21 research outputs found

    Distinct Fates of Chemokine and Surrogate Molecule Gradients: Consequences for CCR7-Guided Dendritic Cell Migration.

    Get PDF
    Chemokine-guided leukocyte migration is a hallmark of the immune system to cope with invading pathogens. Intruder confronted dendritic cells (DCs) induce the expression of the chemokine receptor CCR7, which enables them to sense and migrate along chemokine gradients to home to draining lymph nodes, where they launch an adaptive immune response. Chemokine-mediated DC migration is recapitulated and intensively studied in 3D matrix migration chambers. A major caveat in the field is that chemokine gradient formation and maintenance in such 3D environments is generally not assessed. Instead, fluorescent probes, mostly labelled dextran, are used as surrogate molecules, thereby neglecting important electrochemical properties of the chemokines. Here, we used site-specifically, fluorescently labelled CCL19 and CCL21 to study the establishment and shape of the chemokine gradients over time in the 3D collagen matrix. We demonstrate that CCL19 and particularly CCL21 establish stable, but short-distance spanning gradients with an exponential decay-like shape. By contrast, dextran with its neutral surface charge forms a nearly linear gradient across the entire matrix. We show that the charged C-terminal tail of CCL21, known to interact with extracellular matrix proteins, is determinant for shaping the chemokine gradient. Importantly, DCs sense differences in the shape of CCL19 and CCL21 gradients, resulting in distinct spatial migratory responses

    Epithelial chemokine CXCL14 synergizes with CXCL12 via allosteric modulation of CXCR4

    Get PDF
    The chemokine receptor, CXC chemokine receptor 4 (CXCR4), is selective for CXC chemokine ligand 12 (CXCL12), is broadly expressed in blood and tissue cells, and is essential during embryogenesis and hematopoiesis. CXCL14 is a homeostatic chemokine with unknown receptor selectivity and preferential expression in peripheral tissues. Here, we demonstrate that CXCL14 synergized with CXCL12 in the induction of chemokine responses in primary human lymphoid cells and cell lines that express CXCR4. Combining subactive concentrations of CXCL12 with 100–300 nM CXCL14 resulted in chemotaxis responses that exceeded maximal responses that were obtained with CXCL12 alone. CXCL14 did not activate CXCR4-expressing cells (i.e., failed to trigger chemotaxis and Ca2+ mobilization, as well as signaling via ERK1/2 and the small GTPase Rac1); however, CXCL14 bound to CXCR4 with high affinity, induced redistribution of cell-surface CXCR4, and enhanced HIV-1 infection by >3-fold. We postulate that CXCL14 is a positive allosteric modulator of CXCR4 that enhances the potency of CXCR4 ligands. Our findings provide new insights that will inform the development of novel therapeutics that target CXCR4 in a range of diseases, including cancer, autoimmunity, and HIV.—Collins, P. J., McCully, M. L., MartŽınez-Muñoz, L., Santiago, C.,Wheeldon, J., Caucheteux, S., Thelen, S., Cecchinato, V., Laufer, J.M., Purvanov, V.,Monneau, Y. R., Lortat-Jacob, H., Legler, D. F., Uguccioni, M., Thelen, M., Piguet, V., Mellado, M., Moser, B. Epithelial chemokine CXCL14 synergizes with CXCL12 via allosteric modulation of CXCR4. FASEB J. 31, 000–000 (2017). www.fasebj.or

    Wnt secretion and gradient formation.

    Get PDF
    Concentration gradients formed by the lipid-modified morphogens of the Wnt family are known for their pivotal roles during embryogenesis and adult tissue homeostasis. Wnt morphogens are also implicated in a variety of human diseases, especially cancer. Therefore, the signaling cascades triggered by Wnts have received considerable attention during recent decades. However, how Wnts are secreted and how concentration gradients are formed remains poorly understood. The use of model organisms such as Drosophila melanogaster has provided important advances in this area. For instance, we have previously shown that the lipid raft-associated reggie/flotillin proteins influence Wnt secretion and spreading in Drosophila. Our work supports the notion that producing cells secrete Wnt molecules in at least two pools: a poorly diffusible one and a reggie/flotillin-dependent highly diffusible pool which allows morphogen spreading over long distances away from its source of production. Here we revise the current views of Wnt secretion and spreading, and propose two models for the role of the reggie/flotillin proteins in these processes: (i) reggies/flotillins regulate the basolateral endocytosis of the poorly diffusible, membrane-bound Wnt pool, which is then sorted and secreted to apical compartments for long-range diffusion, and (ii) lipid rafts organized by reggies/flotillins serve as "dating points" where extracellular Wnt transiently interacts with lipoprotein receptors to allow its capture and further spreading via lipoprotein particles. We further discuss these processes in the context of human breast cancer. A better understanding of these phenomena may be relevant for identification of novel drug targets and therapeutic strategies

    Direkte und funktionale Interaktion zwischen Trimeric G Protein Go und Rab5 in G proteingekoppelte Rezeptorsignalisierung

    No full text
    Die Internalisierung und der intrazellulĂ€re Transport von GPCRs ist ein hoch regulierter und dynamischer Prozess, der fĂŒr Desensibilisierung, Phosphorylierung und Resensibilisierung vieler GPCR entscheidend ist. Viele GPCRs erreichen durch die durch Rab5 gesteuerte Internalisierung ein starkes Signal, da sie den Rezeptor und seine Effektoren in die NĂ€he der nĂ€chsten Komponenten der Signalkaskade bringen. Normalerweise wird das Signal dadurch beendet, dass der Rezeptor zu multivesikulĂ€ren Körperchen gebracht wird. Das schnelle Recyclen durch Rab4 erlaubt mehrere Zyklen von kĂŒrzerer GPCR Aktivierung und starke Signale an der Membran. Im Gegensatz dazu fĂŒhrt das langsamere Rezeptorrecycling durch Rab11 zu einem Signal, das tiefer in das Zytoplasma reicht. Lange Zeit wurde die Funktion von Rab Proteinen, Vesikel von einer zur anderen Membran zu transferieren, als eher passiv angesehen. Unsere Ergebnisse zeigen nun, dass Rezeptoren der Frizzled Familie in vitro direkt mit Rab5 interagieren können und diese GTPase in Drosophila Zellen aktivieren können. Außerdem konnten wir zeigen, dass der Prozess der Frizzled Internalisierung von Gαo abhĂ€ngig ist. ZusĂ€tzlich konnten wir zeigen, dass Gαo in vitro direkt an Rab5 und Rab4 bindet und dass es Rab5 durch Rekrutierung des Proteins an die Membran aktiviert. Dies ist der erste Beweis einer direkten und funktionellen Interaktion eines Gαo-Proteins mit einer Rab-GTPase. Wir zeigen das Mitwirken von Rab4, Rab5 und Rab11 in der planaren ZellpolaritĂ€t und im Wingless-Frizzled Signaltransduktionsweg in Drosophila und schlagen ein Model fĂŒr die Regulierung beider Signalwege vor. Wir schlagen vor, dass die durch Frizzled hervorgerufenen Aktivierung von Gαo zur Rekrutierung von Rab5 in die NĂ€he der Frizzeld-Rezeptoren fĂŒhrt, wodurch die Endozytose des Rezeptors katalysiert und daher die SignalintensitĂ€t amplifiziert wird. Danach definieren unterschiedliche Transportwege der Frizzled Komplexe die SpezifitĂ€t der Aktivierung von Wingless oder planaren ZellpolaritĂ€t Frizzled-Signalwege. Unsere Beobachtungen bringen Erkenntnisse fĂŒr den Frizzled- Signaltransduktionsweg und seine Regulierung, ebenso wie fĂŒr die gesamte GPCR-Biologie.ZusĂ€tzlich haben wir einen in vitro Assay etabliert, um G-Proteine untersuchen zu können und wir konnten zeigen, dass Europium-GTP in den gleichen Experimenten, in denen traditionell radioaktive Nukleotide eingesetzt werden, verwendet werden kann. Daher vermeidet dieser Assay nicht nur die Nachteile von radioaktiven GTPÎłS, die mit der Verwendung von radioaktiven Komponenten und radioaktivem Abfall einhergehen und daher in der modernen Laborpraxis unerwĂŒnscht sind, sondern ermöglicht auch Experimente im "high-throughput" Format durchzufĂŒhren. Unsere Methode verwendet die zeitauflösende Fluorometrie, eine etablierte Technologie, die die fluoreszierenden Eigenschafen von Lanthanoidchelaten verwendet, als leistungsfĂ€hige Alternative fĂŒr Assays mit radioaktiven Isotope

    A Direct and Functional Interaction Between Go and Rab5 During G Protein-Coupled Receptor Signaling

    No full text
    Rab5 is a small guanosine triphosphatase (GTPase) that regulates the early stages of endocytosis and is conserved in eukaryotes. Rab5 regulates the internalization of receptors and other membrane-associated signaling proteins. The function of Rab5 in these processes is considered relatively passive, so that the endocytic capacity of Rab5 is used during, for example, ÎČ-arrestin–dependent internalization of G protein (heterotrimeric guanine nucleotide–binding protein)–coupled receptors (GPCRs). Direct recruitment or activation of Rab5 by the components of these signaling pathways has not been reported. Here, we demonstrate an interaction of Drosophila Rab5 and an immediate transducer of GPCR signaling, the G protein Go, in vitro and in vivo. Rab5 and Go bound to each other as purified proteins, as well as in fly extracts. In cellular assays, Go led to Rab5 activation and endosome fusion. We further showed that the Go-Rab5 interaction functioned in Drosophila planar cell polarity and Wingless signal transduction, pathways initiated by GPCRs of the Frizzled (Fz) family. Additionally, the recycling Rab GTPases Rab4 and Rab11 functioned in Fz- and Go-mediated signaling to favor planar cell polarity over canonical Wingless signaling. The interplay between heterotrimeric G proteins and Rab GTPases controlled receptor internalization, revealing a previously uncharacterized regulatory mechanism in GPCR signaling

    Chemokine Receptor CCR7 Triggers an Endomembrane Signaling Complex for Spatial Rac Activation

    No full text
    Chemokine-guided cell migration is pivotal for many immunological and developmental processes. How chemokine receptor signaling persists to guarantee sustained directional migration despite receptor desensitization and internalization remains poorly understood. Here, we uncover a function for an intracellular pool of the chemokine receptor CCR7 present in human dendritic cells and cellular model systems. We find that CCR7 signaling, initiated at the plasma membrane, is translocated by joint trafficking of ÎČ-arrestin and Src kinase to endomembrane-residing CCR7. There, Src tyrosine phosphorylates CCR7, required for the recruitment of Vav1 to form an endomembrane-residing multi-protein signaling complex comprising CCR7, the RhoGEF Vav1, and its effector, Rac1. Interfering with vesicular trafficking affects CCR7-driven cell migration, whereas CCR7:Vav1 interaction at endomembranes is essential for local Rac1 recruitment to CCR7. Photoactivation of Rac1 at endomembranes leads to lamellipodia formation at the cell's leading edge, supporting the role of sustained endomembrane signaling in guiding cell migration.publishe

    Active site geometry and substrate recognition of the molybdenum hydroxylase quinoline 2-oxidoreductase

    Get PDF
    The soil bacterium Pseudomonas putida 86 uses quinoline as a sole source of carbon and energy. Quinoline 2-oxidoreductase (Qor) catalyzes the first metabolic step converting quinoline to 2-oxo-1,2-dihydroquinoline. Qor is a member of the molybdenum hydroxylases. The molybdenum ion is coordinated by two ene-dithiolate sulfur atoms, two oxo-ligands, and a catalytically crucial sulfido-ligand, whose position in the active site was controversial. The 1.8 Å resolution crystal structure of Qor indicates that the sulfido-ligand occupies the equatorial position at the molybdenum ion. The structural comparison of Qor with the allopurinol-inhibited xanthine dehydrogenase from Rhodobacter capsulatus allows direct insight into the mechanism of substrate recognition and the identification of putative catalytic residues. The active site protein variants QorE743V and QorE743D were analyzed to assess the catalytic role of E743

    Fluorescently Tagged CCL19 and CCL21 to Monitor CCR7 and ACKR4 Functions

    No full text
    Chemokines are essential guidance cues orchestrating cell migration in health and disease. Cognate chemokine receptors sense chemokine gradients over short distances to coordinate directional cell locomotion. The chemokines CCL19 and CCL21 are essential for recruiting CCR7-expressing dendritic cells bearing pathogen-derived antigens and lymphocytes to lymph nodes, where the two cell types meet to launch an adaptive immune response against the invading pathogen. CCR7-expressing cancer cells are also recruited by CCL19 and CCL21 to metastasize in lymphoid organs. In contrast, atypical chemokine receptors (ACKRs) do not transmit signals required for cell locomotion but scavenge chemokines. ACKR4 is crucial for internalizing and degrading CCL19 and CCL21 to establish local gradients, which are sensed by CCR7-expressing cells. Here, we describe the production of fluorescently tagged chemokines by fusing CCL19 and CCL21 to monomeric red fluorescent protein (mRFP). We show that purified CCL19-mRFP and CCL21-mRFP are versatile and powerful tools to study CCR7 and ACKR4 functions, such as receptor trafficking and chemokine scavenging, in a spatiotemporal fashion. We demonstrate that fluorescently tagged CCL19 and CCL21 permit the visualization and quantification of chemokine gradients in real time, while CCR7-expressing leukocytes and cancer cells sense the guidance cues and migrate along the chemokine gradients.publishe

    CAL-1 as Cellular Model System to Study CCR7-Guided Human Dendritic Cell Migration

    Get PDF
    Dendritic cells (DCs) are potent and versatile professional antigen-presenting cells and central for the induction of adaptive immunity. The ability to migrate and transport peripherally acquired antigens to draining lymph nodes for subsequent cognate T cell priming is a key feature of DCs. Consequently, DC-based immunotherapies are used to elicit tumor-antigen specific T cell responses in cancer patients. Understanding chemokine-guided DC migration is critical to explore DCs as cellular vaccines for immunotherapeutic approaches. Currently, research is hampered by the lack of appropriate human cellular model systems to effectively study spatio-temporal signaling and CCR7-driven migration of human DCs. Here, we report that the previously established human neoplastic cell line CAL-1 expresses the human DC surface antigens CD11c and HLA-DR together with co-stimulatory molecules. Importantly, if exposed for three days to GM-CSF, CAL-1 cells induce the endogenous expression of the chemokine receptor CCR7 upon encountering the clinically approved TLR7/8 agonist Resiquimod R848 and readily migrate along chemokine gradients. Further, we demonstrate that CAL-1 cells can be genetically modified to express fluorescent (GFP)-tagged reporter proteins to study and visualize signaling or can be gene-edited using CRISPR/Cas9. Hence, we herein present the human CAL-1 cell line as versatile and valuable cellular model system to effectively study human DC migration and signaling.publishe

    Engineering of Nanobodies Recognizing the Human Chemokine Receptor CCR7

    No full text
    The chemokine receptor CCR7 plays a pivotal role in health and disease. In particular, CCR7 controls homing of antigen-bearing dendritic cells and T cells to lymph nodes, where adaptive immune responses are initiated. However, CCR7 also guides T cells to inflamed synovium and thereby contributes to rheumatoid arthritis and promotes cancer cell migration and metastasis formation. Nanobodies have recently emerged as versatile tools to study G-protein-coupled receptor functions and are being tested in diagnostics and therapeutics. In this study, we designed a strategy to engineer novel nanobodies recognizing human CCR7. We generated a nanobody library based on a solved crystal structure of the nanobody Nb80 recognizing the β2-adrenergic receptor (β2AR) and by specifically randomizing two segments within complementarity determining region 1 (CDR1) and CDR3 of Nb80 known to interact with β2AR. We fused the nanobody library to one half of split-YFP in order to identify individual nanobody clones interacting with CCR7 fused to the other half of split-YFP using bimolecular fluorescence complementation. We present three novel nanobodies, termed Nb1, Nb5, and Nb38, that recognize human CCR7 without interfering with G-protein-coupling and downstream signaling. Moreover, we were able to follow CCR7 trafficking upon CCL19 triggering using Nb1, Nb5, and Nb38
    corecore