23 research outputs found

    Development and validation of kinase activity reporters for the dynamic study of cell response modalities by microscopy

    Get PDF
    Necroptosis is defined as a caspase-independent programmed cell death and relies on a signaling pathway involving two serine-threonine kinases: Receptor-Interacting Protein Kinase 1 and 3 (RIPK1 and RIPK3) and the pseudo-kinase Mixed-Lineage Kinase Like (MLKL). Activation of Extracellular signal-Regulated Kinases 1 and 2 (ERK1/2) was reported to be involved in different modes of programmed cell death. It is now accepted that the regulation of the duration, magnitude and subcellular compartmentalization of ERK1/2 activity by specific spatio-temporal regulators is interpreted by the cell towards cell fate determination. ERK1/2 inhibition delays TNFα-induced necroptosis in L929 cells in a dose dependent manner but did not block it, providing arguments for a pro-necrotic function of ERK1/2. In this context, a compartmentalized biphasic phosphorylation of ERK1/2 was observed. Our results indicate a RIPK1-dependent phosphorylation of ERK1/2. Owing to the importance of ERK1/2 spatio-temporal dynamics in determining cellular responses, we developed a new reporter of ERK2 localization named ERK2-LOC. We observed a transient translocation of ERK2 when necroptosis was triggered in L929 upon TNFα stimulation, followed by progressive ERK2 accumulation in the nucleus. ERK1/2 activities were monitored during necroptosis using a FRET-based kinase biosensor for ERK1/2 (ERK1/2-ACT). Using ERK1/2-ACT, a dedicated spatio-temporal signature of ERK1/2 activity was recorded during necroptosis. Finally, to correlate ERK1/2 activity code with necroptosis occurrence, we also engineered a first generation of FRET biosensors to report on both RIPK1 and RIPK3 activities during necroptosis

    Novel reporter for faithful monitoring of ERK2 dynamics in living cells and model organisms

    Get PDF
    Uncoupling of ERK1/2 phosphorylation from subcellular localization is essential towards the understanding of molecular mechanisms that control ERK1/2-mediated cell-fate decision. ERK1/2 non-catalytic functions and discoveries of new specific anchors responsible of the subcellular compartmentalization of ERK1/2 signaling pathway have been proposed as regulation mechanisms for which dynamic monitoring of ERK1/2 localization is necessary. However, studying the spatiotemporal features of ERK2, for instance, in different cellular processes in living cells and tissues requires a tool that can faithfully report on its subcellular distribution. We developed a novel molecular tool, ERK2-LOC, based on the T2A-mediated coexpression of strictly equimolar levels of eGFP-ERK2 and MEK1, to faithfully visualize ERK2 localization patterns. MEK1 and eGFP-ERK2 were expressed reliably and functionally both in vitro and in single living cells. We then assessed the subcellular distribution and mobility of ERK2-LOC using fluorescence microscopy in non-stimulated conditions and after activation/inhibition of the MAPK/ERK1/2 signaling pathway. Finally, we used our coexpression system in Xenopus laevis embryos during the early stages of development. This is the first report on MEK1/ERK2 T2A-mediated coexpression in living embryos, and we show that there is a strong correlation between the spatiotemporal subcellular distribution of ERK2-LOC and the phosphorylation patterns of ERK1/2. Our approach can be used to study the spatiotemporal localization of ERK2 and its dynamics in a variety of processes in living cells and embryonic tissues

    Nesprins are mechanotransducers that discriminate epithelial-mesenchymal transition programs

    Get PDF
    © 2020 DĂ©jardin et al. This article is distributed under the terms of an Attribution–Noncommercial–Share Alike–No Mirror Sites license for the first six months after the publication date (see http://www.rupress.org/terms/). After six months it is available under a Creative Commons License (Attribution–Noncommercial–Share Alike 4.0 International license, as described at https://creativecommons.org/licenses/by-nc-sa/4.0/).LINC complexes are transmembrane protein assemblies that physically connect the nucleoskeleton and cytoskeleton through the nuclear envelope. Dysfunctions of LINC complexes are associated with pathologies such as cancer and muscular disorders. The mechanical roles of LINC complexes are poorly understood. To address this, we used genetically encoded FRET biosensors of molecular tension in a nesprin protein of the LINC complex of fibroblastic and epithelial cells in culture. We exposed cells to mechanical, genetic, and pharmacological perturbations, mimicking a range of physiological and pathological situations. We show that nesprin experiences tension generated by the cytoskeleton and acts as a mechanical sensor of cell packing. Moreover, nesprin discriminates between inductions of partial and complete epithelial-mesenchymal transitions. We identify the implicated mechanisms, which involve α-catenin capture at the nuclear envelope by nesprin upon its relaxation, thereby regulating ÎČ-catenin transcription. Our data thus implicate LINC complex proteins as mechanotransducers that fine-tune ÎČ-catenin signaling in a manner dependent on the epithelial-mesenchymal transition program.This material is based on work supported by the Centre national de la recherche scientifique (CNRS), Agence nationale de la recherche (ANR; grants ANR-13-JSV5-0007 and ANR-14-CE09-0006), France BioImaging (ANR-10-INBS-04), la Ligue contre le Cancer (REMX17751 to P.M. Davidson), and the Fondation ARC pour la Recherche sur le Cancer (PDF20161205227 to P.M. Davidson). P.S. Carollo has received funding from the European Union’s Horizon 2020 Framework Programme for Research and Innovation (Marie SkƂodowska-Curie grant agreement 665850-INSPIRE) and acknowledges the Ecole Doctorale FrontiĂšres de l'Innovation en Recherche et Éducation (FIRE) Programme Bettencourt. E.R. Gomes was supported by a European Research Council consolidator grant (617676).info:eu-repo/semantics/publishedVersio

    Datalog+, RuleML and OWL 2: Formats and Translations for Existential Rules

    Get PDF
    International audienceThis paper is devoted to formats and translations for Datalog+. We first introduce the dlgp format, which extends classical Datalog format to Datalog+. It allows to encode facts, existential rules (including equality), negative constraints and conjunctive queries. Moreover, for compatibility with Semantic Web languages, this format includes Web notions (IRIs and literals, according to Turtle syntax). Second, we define a translation from dlgp to the Datalog+ fragment of RuleML. Third, we define a translation from OWL 2 to dlgp. We point out that the composition of both translations allows to import OWL 2 to RuleML. The associated parsers and translators are available

    A mechanical G2 checkpoint controls epithelial cell division through E-cadherin-mediated regulation of Wee1-Cdk1

    Get PDF
    Epithelial cell divisions are coordinated with cell loss to preserve epithelial integrity. However, how epithelia adapt their rate of cell division to changes in cell number, for instance during homeostatic turnover or wounding, is not well understood. Here, we show that epithelial cells sense local cell density through mechanosensitive E-cadherin adhesions to control G2/M cell-cycle progression. As local cell density increases, tensile forces on E-cadherin adhesions are reduced, which prompts the accumulation of the G2 checkpoint kinase Wee1 and downstream inhibitory phosphorylation of Cdk1. Consequently, dense epithelia contain a pool of cells that are temporarily halted in G2 phase. These cells are readily triggered to divide following epithelial wounding due to the consequent increase in intercellular forces and resulting degradation of Wee1. Our data collectively show that epithelial cell division is controlled by a mechanical G2 checkpoint, which is regulated by cell-density-dependent intercellular forces sensed and transduced by E-cadherin adhesions.Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved

    Développement et validation de rapporteurs d'activité kinase pour l'étude dynamique des processus cellulaires par microscopie : rÎle de la Mitogen-Activated Protein Kinase / Extracellular signal-Regulated Kinase dans la nécroptose

    No full text
    La nĂ©croptose est une mort programmĂ©e caspase indĂ©pendante, induite par le Tumor Necrosis Factor alpha (TNFα) et fait intervenir une voie de signalisation impliquant deux kinases, les Receptor-Interacting Protein Kinases 1 et 3 (RIPK1 et RIPK3) et la pseudo-kinase Mixed Lineage Kinase domain-Like protein (MLKL). L’activation des kinases Extracellular signal-Regulated Kinase 1 et 2 (ERK1/2) a Ă©tĂ© rapportĂ©e dans plusieurs morts cellulaires programmĂ©es. Par ailleurs, la rĂ©gulation de l’activitĂ© de ERK1/2 en termes d’amplitude, de durĂ©e et de localisation via des rĂ©gulateurs spatio-temporels spĂ©cifiques est nĂ©cessaire pour l’orientation du destin cellulaire. L’inhibition de ERK1/2 retarde la nĂ©croptose induite par le TNFα dans les L929 de maniĂšre dose-dĂ©pendante, sans pour autant la bloquer, suggĂ©rant un rĂŽle pro-nĂ©crotique de ERK1/2. Dans ces conditions expĂ©rimentales, une activitĂ© biphasique et compartimentĂ©e de ERK1/2 a Ă©tĂ© observĂ©e. De plus, nos rĂ©sultats indiquent que la phosphorylation ERK1/2 est RIPK1 dĂ©pendante. Nous avons dĂ©veloppĂ© un nouveau rapporteur de localisation de ERK2, appelĂ© ERK2-LOC. Une translocation transitoire suivi d’une accumulation nuclĂ©aire de ERK2 suite Ă  la stimulation des L929 par le TNFα a Ă©tĂ© observĂ©e. Les mesures d’activitĂ© de ERK1/2 au cours de la nĂ©croptose ont Ă©tĂ© rĂ©alisĂ©es avec un biosenseur FRET d’activitĂ© kinase (ERK1/2-ACT). Son utilisation a rĂ©vĂ©lĂ© une signature spatio-temporelle spĂ©cifique d’activitĂ© au cours de la nĂ©croptose. Afin de corrĂ©ler les signatures d’activitĂ© de ERK1/2 avec celles des kinases RIPK1 et RIPK3, nous avons dĂ©veloppĂ© une premiĂšre gĂ©nĂ©ration de biosenseurs FRET pour ces kinases initiatrices de la nĂ©croptose.Necroptosis is defined as a caspase-independent programmed cell death and relies on a signaling pathway involving two serine-threonine kinases: Receptor-Interacting Protein Kinase 1 and 3 (RIPK1 and RIPK3) and the pseudo-kinase Mixed-Lineage Kinase Like (MLKL). Activation of Extracellular signal-Regulated Kinases 1 and 2 (ERK1/2) was reported to be involved in different modes of programmed cell death. It is now accepted that the regulation of the duration, magnitude and subcellular compartmentalization of ERK1/2 activity by specific spatio-temporal regulators is interpreted by the cell towards cell fate determination. ERK1/2 inhibition delays TNFα-induced necroptosis in L929 cells in a dose dependent manner but did not block it, providing arguments for a pro-necrotic function of ERK1/2. In this context, a compartmentalized biphasic phosphorylation of ERK1/2 was observed. Our results indicate a RIPK1-dependent phosphorylation of ERK1/2. Owing to the importance of ERK1/2 spatio-temporal dynamics in determining cellular responses, we developed a new reporter of ERK2 localization named ERK2-LOC. We observed a transient translocation of ERK2 when necroptosis was triggered in L929 upon TNFα stimulation, followed by progressive ERK2 accumulation in the nucleus. ERK1/2 activities were monitored during necroptosis using a FRET-based kinase biosensor for ERK1/2 (ERK1/2-ACT). Using ERK1/2-ACT, a dedicated spatio-temporal signature of ERK1/2 activity was recorded during necroptosis. Finally, to correlate ERK1/2 activity code with necroptosis occurrence, we also engineered a first generation of FRET biosensors to report on both RIPK1 and RIPK3 activities during necroptosis

    Shining light on cell death processes: a novel biosensor for necroptosis, a newly described cell death program

    No full text
    Cell death contributes to the maintenance of homeostasis, but mounting evidence has confirmed the involvement of programmed cell death in some diseases. The concept of programmed cell death, which was coined several decades ago to refer to apoptosis, now also encompasses necroptosis, a newly characterized cell death program. Research on programmed cell death has become essential for the development of some new therapies. To study cell death signaling and its molecular mechanisms, new biochemical and fluorogenic approaches have been devised. Here, we first provide an overview of programmed cell death modes and the importance of dynamic cell death studies. Next, we focus on both apoptotic and necroptotic signaling and their mechanisms by providing a systematic review of all the methods and approaches that have been used. We emphasize the contribution of advanced approaches based on fluorescent probes, reporters, and Forster resonance energy transfer (FRET)-based biosensors for studying programmed cell death. Because apoptosis and necroptosis signaling pathways share some effectors molecules, we discuss how these new tools could be used to discriminate between apoptosis and necroptosis. We also describe how we developed specific FRET-based biosensors for detecting necroptosis. Finally, we touch on how dynamic measurement of biomolecules in living models will play a role in personalized prognosis and therapy

    Graal: A Toolkit for Query Answering with Existential Rules

    No full text
    International audienceThis paper presents Graal, a java toolkit dedicated to ontological query answering in the framework of existential rules. We consider knowledge bases composed of data and an ontology expressed by existential rules. The main features of Graal are the following: a basic layer that provides generic interfaces to store and query various kinds of data, forward chaining and query rewriting algorithms, structural analysis of decidability properties of a rule set, a textual format and its parser, and import of OWL 2 files. We describe in more detail the query rewriting algorithms, which rely on original techniques, and report some experiments
    corecore