32 results on '"Mark G.H. Scott"'
Search Results
2. Control of the Mdm2-p53 signal loop by β-arrestin 2: the ins and outs
- Author
-
Mark G.H. Scott, Hervé Enslen, Elodie Blondel-Tepaz, Enslen, Hervé, Institut Cochin (IC UM3 (UMR 8104 / U1016)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), and SCOTT, Mark
- Subjects
p53 ,Physics ,β-arrestin ,[SDV]Life Sciences [q-bio] ,Signal ,Cell biology ,[SDV] Life Sciences [q-bio] ,Loop (topology) ,Editorial ,Mdm2 ,Oncology ,SUMO ,β arrestin 2 ,RanGAP1 ,Mdm2 p53 - Abstract
International audience; No abstract available
- Published
- 2021
3. The RanBP2/RanGAP1-SUMO complex gates β-arrestin2 nuclear entry to regulate the Mdm2-p53 signaling axis
- Author
-
Stephane Angers, Kusumika Saha, Jane E. Findlay, Evelyne Lima-Fernandes, Michel Bouvier, Justine S. Paradis, Stefano Marullo, Cédric Auffray, George S. Baillie, Hervé Enslen, Mark G.H. Scott, Alessia Zamborlini, Anne Poupon, Elodie Blondel-Tepaz, Badr Sokrat, Milena Kosic, Louis Gaboury, Marie Leverve, Institut Cochin (IC UM3 (UMR 8104 / U1016)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP), Université de Montréal (UdeM), University of Toronto, Institut de Biologie Intégrative de la Cellule (I2BC), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS), CEA- Saclay (CEA), Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Physiologie de la reproduction et des comportements [Nouzilly] (PRC), Institut Français du Cheval et de l'Equitation [Saumur]-Université de Tours-Centre National de la Recherche Scientifique (CNRS)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), University of Glasgow, ANR-11-LABX-0071,WHO AM I,Determinants de l'Identité : de la molécule à l'individu(2011), Institut Français du Cheval et de l'Equitation [Saumur]-Université de Tours (UT)-Centre National de la Recherche Scientifique (CNRS)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), SCOTT, Mark, Determinants de l'Identité : de la molécule à l'individu - - WHO AM I2011 - ANR-11-LABX-0071 - LABX - VALID, Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), Institut Français du Cheval et de l'Equitation [Saumur] (IFCE)-Université de Tours (UT)-Centre National de la Recherche Scientifique (CNRS)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Dynamiques de populations multi-échelles pour des systèmes physiologiques (MUSCA), Inria Saclay - Ile de France, Institut National de Recherche en Informatique et en Automatique (Inria)-Institut National de Recherche en Informatique et en Automatique (Inria)-Physiologie de la reproduction et des comportements [Nouzilly] (PRC), Institut Français du Cheval et de l'Equitation [Saumur] (IFCE)-Université de Tours (UT)-Centre National de la Recherche Scientifique (CNRS)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)-Institut Français du Cheval et de l'Equitation [Saumur] (IFCE)-Université de Tours (UT)-Centre National de la Recherche Scientifique (CNRS)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)-Mathématiques et Informatique Appliquées du Génome à l'Environnement [Jouy-En-Josas] (MaIAGE), and Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)
- Subjects
0301 basic medicine ,Scaffold protein ,Cancer Research ,[SDV]Life Sciences [q-bio] ,SUMO-1 Protein ,SUMO protein ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Neoplasms ,Genetics ,Humans ,Nuclear pore ,Nuclear export signal ,Molecular Biology ,Cytoskeleton ,Cell Nucleus ,Nuclear Export Signals ,GTPase-Activating Proteins ,Sumoylation ,Proto-Oncogene Proteins c-mdm2 ,beta-Arrestin 2 ,Cell biology ,[SDV] Life Sciences [q-bio] ,enzymes and coenzymes (carbohydrates) ,030104 developmental biology ,Nucleocytoplasmic Transport ,Cytoplasm ,030220 oncology & carcinogenesis ,Mutation ,RANBP2 ,Tumor Suppressor Protein p53 ,Nuclear transport ,Signal Transduction - Abstract
International audience; Mdm2 antagonizes the tumor suppressor p53. Targeting the Mdm2-p53 interaction represents an attractive approach for the treatment of cancers with functional p53. Investigating mechanisms underlying Mdm2-p53 regulation is therefore important. The scaffold protein β-arrestin2 (β-arr2) regulates tumor suppressor p53 by counteracting Mdm2. β-arr2 nucleocytoplasmic shuttling displaces Mdm2 from the nucleus to the cytoplasm resulting in enhanced p53 signaling. β-arr2 is constitutively exported from the nucleus, via a nuclear export signal, but mechanisms regulating its nuclear entry are not completely elucidated. β-arr2 can be SUMOylated, but no information is available on how SUMO may regulate β-arr2 nucleocytoplasmic shuttling. While we found β-arr2 SUMOylation to be dispensable for nuclear import, we identified a non-covalent interaction between SUMO and β-arr2, via a SUMO interaction motif (SIM), that is required for β-arr2 cytonuclear trafficking. This SIM promotes association of β-arr2 with the multimolecular RanBP2/RanGAP1-SUMO nucleocytoplasmic transport hub that resides on the cytoplasmic filaments of the nuclear pore complex. Depletion of RanBP2/RanGAP1-SUMO levels result in defective β-arr2 nuclear entry. Mutation of the SIM inhibits β-arr2 nuclear import, its ability to delocalize Mdm2 from the nucleus to the cytoplasm and enhanced p53 signaling in lung and breast tumor cell lines. Thus, a β-arr2 SIM nuclear entry checkpoint, coupled with active β-arr2 nuclear export, regulates its cytonuclear trafficking function to control the Mdm2-p53 signaling axis.
- Published
- 2021
4. TRPV1 Activation Promotes β-arrestin2 Interaction with the Ribosomal Biogenesis Machinery in the Nucleolus: Implications for p53 Regulation and Neurite Outgrowth
- Author
-
Robyn Flynn, Daniel Young, Manon Defaye, Mircea Iftinca, Christophe Altier, Mark G.H. Scott, Nasser Abdullah, Antoine Dufour, Ahmed Hassan, Francina Agosti, University of Calgary, [Institut Cochin] Département Endocrinologie, métabolisme, diabète (EMD) (EMD), Institut Cochin (IC UM3 (UMR 8104 / U1016)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), and SCOTT, Mark
- Subjects
Proteomics ,Nucleolus ,[SDV.NEU.NB]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,lcsh:Chemistry ,0302 clinical medicine ,Treacle protein ,RNA Polymerase I ,Ganglia, Spinal ,lcsh:QH301-705.5 ,Spectroscopy ,β-arrestins ,Neurons ,0303 health sciences ,Chemistry ,General Medicine ,beta-Arrestin 2 ,Computer Science Applications ,Cell biology ,Protein Transport ,Signal transduction ,chronic pain ,Nucleophosmin ,Cell Nucleolus ,Protein Binding ,Neurite ,Neuronal Outgrowth ,neuroplasticity ,TRPV Cation Channels ,Article ,Catalysis ,ribosomal biogenesis ,Inorganic Chemistry ,03 medical and health sciences ,RNA polymerase I ,Animals ,Humans ,Physical and Theoretical Chemistry ,Molecular Biology ,030304 developmental biology ,Organic Chemistry ,[SDV.NEU.NB] Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,Mice, Inbred C57BL ,TRPV1 ,HEK293 Cells ,lcsh:Biology (General) ,lcsh:QD1-999 ,Cytoplasm ,Tumor Suppressor Protein p53 ,Ribosomes ,030217 neurology & neurosurgery ,Biogenesis - Abstract
International audience; Transient receptor potential vanilloids (TRPV1) are non-selective cation channels that sense and transduce inflammatory pain signals. We previously reported that activation of TRPV1 induced the translocation of β-arrestin2 (ARRB2) from the cytoplasm to the nucleus, raising questions about the functional role of ARRB2 in the nucleus. Here, we determined the ARRB2 nuclear signalosome by conducting a quantitative proteomic analysis of the nucleus-sequestered L395Q ARRB2 mutant, compared to the cytosolic wild-type ARRB2 (WT ARRB2), in a heterologous expression system. We identified clusters of proteins that localize to the nucleolus and are involved in ribosomal biogenesis. Accordingly, L395Q ARRB2 or WT ARRB2 after capsaicin treatment were found to co-localize and interact with the nucleolar marker nucleophosmin (NPM1), treacle protein (TCOF1) and RNA polymerase I (POL I). We further investigated the role of nuclear ARRB2 signaling in regulating neuroplasticity. Using neuroblastoma (neuro2a) cells and dorsal root ganglia (DRG) neurons, we found that L395Q ARRB2 mutant increased POL I activity, inhibited the tumor suppressorp53 (p53) level and caused a decrease in the outgrowth of neurites. Together, our results suggest that the activation of TRPV1 promotes the ARRB2-mediated regulation of ribosomal biogenesis in the nucleolus. The ARRB2-TCOF1-p53 checkpoint signaling pathway might be involved in regulating neurite outgrowth associated with pathological pain conditions.
- Published
- 2021
5. Beta-arrestins operate an on/off control switch for focal adhesion kinase activity
- Author
-
Stefano Marullo, Aurélie Borrull, Mireille Lambert, Isaure Lot, Alexandre Beautrait, Michel Bouvier, Revu Ann Alexander, Kusumika Saha, Eleonore Decosta, Mark G.H. Scott, Atef Asnacios, Hervé Enslen, Guillaume Abadie, Hiroyuki Kobayashi, [Institut Cochin] Département Endocrinologie, métabolisme, diabète (EMD) (EMD), Institut Cochin (IC UM3 (UMR 8104 / U1016)), Centre National de la Recherche Scientifique (CNRS)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Department of Biochemistry [Montreal, QC, Canada] (Institute for Research in Immunology and Cancer), Université de Montréal (UdeM), Matière et Systèmes Complexes (MSC (UMR_7057)), Centre National de la Recherche Scientifique (CNRS)-Université Paris Diderot - Paris 7 (UPD7), Enslen, Hervé, Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), and Université Paris Diderot - Paris 7 (UPD7)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Scaffold protein ,genetic structures ,[SDV]Life Sciences [q-bio] ,G-protein-coupled receptors ,[SDV.BC.IC] Life Sciences [q-bio]/Cellular Biology/Cell Behavior [q-bio.CB] ,Receptors, G-Protein-Coupled ,Mice ,0302 clinical medicine ,Neoplasms ,[SDV.BC.IC]Life Sciences [q-bio]/Cellular Biology/Cell Behavior [q-bio.CB] ,Phosphorylation ,beta-Arrestins ,ComputingMilieux_MISCELLANEOUS ,0303 health sciences ,FERM domain ,Chemistry ,Autophosphorylation ,Cell biology ,[SDV] Life Sciences [q-bio] ,030220 oncology & carcinogenesis ,Molecular Medicine ,Arrestin beta 2 ,biological phenomena, cell phenomena, and immunity ,G proteins ,Protein Binding ,G protein ,Vasopressins ,Adaptor Protein Complex 2 ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Receptor, Angiotensin, Type 1 ,Focal adhesion ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,Protein Domains ,GTP-Binding Proteins ,Animals ,Humans ,[SDV.BC] Life Sciences [q-bio]/Cellular Biology ,Molecular Biology ,030304 developmental biology ,G protein-coupled receptor ,Pharmacology ,Beta-arrestin ,FAK ,Beta-Arrestins ,Cell Membrane ,β-Arrestin ,Cell Biology ,AP-2 ,HEK293 Cells ,Focal Adhesion Protein-Tyrosine Kinases ,Multiprotein Complexes ,sense organs - Abstract
International audience; Focal adhesion kinase (FAK) regulates key biological processes downstream of G protein-coupled receptors (GPCRs) in normal and cancer cells, but the modes of kinase activation by these receptors remain unclear. We report that after GPCR stimulation, FAK activation is controlled by a sequence of events depending on the scaffolding proteins β-arrestins and G proteins. Depletion of β-arrestins results in a marked increase in FAK autophosphorylation and focal adhesion number. We demonstrate that β-arrestins interact directly with FAK and inhibit its autophosphorylation in resting cells. Both FAK-β-arrestin interaction and FAK inhibition require the FERM domain of FAK. Following the stimulation of the angiotensin receptor AT1AR and subsequent translocation of the FAK-β-arrestin complex to the plasma membrane, β-arrestin interaction with the adaptor AP-2 releases inactive FAK from the inhibitory complex, allowing its activation by receptor-stimulated G proteins and activation of downstream FAK effectors. Release and activation of FAK in response to angiotensin are prevented by an AP-2-binding deficient β-arrestin and by a specific inhibitor of β-arrestin/AP-2 interaction; this inhibitor also prevents FAK activation in response to vasopressin. This previously unrecognized mechanism of FAK regulation involving a dual role of β-arrestins, which inhibit FAK in resting cells while driving its activation at the plasma membrane by GPCR-stimulated G proteins, opens new potential therapeutic perspectives in cancers with up-regulated FAK.
- Published
- 2020
6. Posttranslational Regulation and Conformational Plasticity of PTEN
- Author
-
Mark G.H. Scott, Eric Chastre, Larissa Kotelevets, and Barbara Trifault
- Subjects
0301 basic medicine ,Phosphatase ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Neoplasms ,medicine ,PTEN ,Tensin ,Humans ,Post-translational regulation ,Genes, Tumor Suppressor ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,biology ,Kinase ,PTEN Phosphohydrolase ,Cell biology ,030104 developmental biology ,Cell Transformation, Neoplastic ,030220 oncology & carcinogenesis ,Lipid phosphatase activity ,biology.protein ,Carcinogenesis ,Perspectives ,Signal Transduction - Abstract
Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a tumor suppressor that is frequently down-modulated in human cancer. PTEN inhibits the phosphatidylinositol 3-phosphate kinase (PI3K)/AKT pathway through its lipid phosphatase activity. Multiple PI3K/AKT-independent actions of PTEN, protein-phosphatase activities and functions within the nucleus have also been described. PTEN, therefore, regulates many cellular processes including cell proliferation, survival, genomic integrity, polarity, migration, and invasion. Even a modest decrease in the functional dose of PTEN may promote cancer development. Understanding the molecular and cellular mechanisms that regulate PTEN protein levels and function, and how these may go awry in cancer contexts, is, therefore, key to fully understanding the role of PTEN in tumorigenesis. Here, we discuss current knowledge on posttranslational control and conformational plasticity of PTEN, as well as therapeutic possibilities toward reestablishment of PTEN tumor-suppressor activity in cancer.
- Published
- 2020
7. Methods to Investigate the Nucleocytoplasmic Shuttling Properties of β-Arrestins
- Author
-
Mark G.H. Scott, Elodie Blondel-Tepaz, Thomas Guilbert, Institut Cochin (IC UM3 (UMR 8104 / U1016)), and Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,Gene isoform ,media_common.quotation_subject ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Leptomycin ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,chemistry ,Cytoplasm ,medicine ,Receptor ,Internalization ,Nuclear export signal ,Nucleus ,030217 neurology & neurosurgery ,ComputingMilieux_MISCELLANEOUS ,G protein-coupled receptor ,media_common - Abstract
β-Arrestins (β-arrs) were originally appreciated for the roles they play in the desensitization and internalization of G protein-coupled receptors (GPCRs). They are also now known to act as molecular scaffolds, providing control in multiple signalling pathways. Through their scaffolding properties, β-arrs dynamically regulate the activity and/or subcellular distribution of protein partners giving rise to an appropriate cellular response. There are two β-arr isoforms, namely, β-arr1 and β-arr2, which share high sequence homology and structural conservation. While the β-arrs often display conserved overlapping roles, decisive differences between the isoforms also exist. A striking example of this is the subcellular distribution of the β-arr isoforms. While β-arr1 is distributed both in cytoplasmic and nuclear compartments, β-arr2 displays an apparent cytoplasmic distribution. Both β-arrs are actively imported into the nucleus, but β-arr2 is constitutively exported by a leptomycin B-sensitive pathway due to a nuclear export signal in its C-terminus that is absent in β-arr1. β-arr2 therefore undergoes constitutive nucleocytoplasmic shuttling enabling the displacement of nuclear binding cargoes, such as Mdm2. Here, we describe methods to explore the differential nucleocytoplasmic shuttling capacities of the β-arrs.
- Published
- 2019
8. Author response: PTEN controls glandular morphogenesis through a juxtamembrane β-Arrestin1/ARHGAP21 scaffolding complex
- Author
-
Ravi K. Deevi, Emma Evergren, Frederick Charles Campbell, Mark G.H. Scott, Arman Javadi, George S. Baillie, and Elodie Blondel-Tepaz
- Subjects
Scaffold ,Morphogenesis ,biology.protein ,PTEN ,Biology ,β arrestin1 ,Cell biology - Published
- 2017
9. PTEN controls glandular morphogenesis through a juxtamembrane β-Arrestin1/ARHGAP21 scaffolding complex
- Author
-
Frederick Charles Campbell, Arman Javadi, Mark G.H. Scott, Ravi K. Deevi, Emma Evergren, George S. Baillie, and Elodie Blondel-Tepaz
- Subjects
0301 basic medicine ,Scaffold protein ,PTEN ,Cell division ,Cell ,CDC42 ,Tissue Culture Techniques ,Cell membrane ,Mice ,Morphogenesis ,Cdc42 protein ,RNA, Small Interfering ,Biology (General) ,cdc42 GTP-Binding Protein ,Cancer Biology ,C2 domain ,Arrestin ,biology ,General Neuroscience ,GTPase-Activating Proteins ,ARHGAP21 ,General Medicine ,3. Good health ,Cell biology ,Organoids ,beta-Arrestin 1 ,medicine.anatomical_structure ,Medicine ,Protein Binding ,Signal Transduction ,Research Article ,Human ,QH301-705.5 ,Science ,Spindle Apparatus ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,medicine ,Animals ,Humans ,Binding Sites ,General Immunology and Microbiology ,Cell Membrane ,PTEN Phosphohydrolase ,Cell Biology ,HCT116 Cells ,Mice, Inbred C57BL ,HEK293 Cells ,030104 developmental biology ,Gene Expression Regulation ,biology.protein ,Caco-2 Cells - Abstract
PTEN controls three-dimensional (3D) glandular morphogenesis by coupling juxtamembrane signaling to mitotic spindle machinery. While molecular mechanisms remain unclear, PTEN interacts through its C2 membrane-binding domain with the scaffold protein β-Arrestin1. Because β-Arrestin1 binds and suppresses the Cdc42 GTPase-activating protein ARHGAP21, we hypothesize that PTEN controls Cdc42 -dependent morphogenic processes through a β-Arrestin1-ARHGAP21 complex. Here, we show that PTEN knockdown (KD) impairs β-Arrestin1 membrane localization, β-Arrestin1-ARHGAP21 interactions, Cdc42 activation, mitotic spindle orientation and 3D glandular morphogenesis. Effects of PTEN deficiency were phenocopied by β-Arrestin1 KD or inhibition of β-Arrestin1-ARHGAP21 interactions. Conversely, silencing of ARHGAP21 enhanced Cdc42 activation and rescued aberrant morphogenic processes of PTEN-deficient cultures. Expression of the PTEN C2 domain mimicked effects of full-length PTEN but a membrane-binding defective mutant of the C2 domain abrogated these properties. Our results show that PTEN controls multicellular assembly through a membrane-associated regulatory protein complex composed of β-Arrestin1, ARHGAP21 and Cdc42., eLife digest The protein PTEN helps to organize cells in the body to form complex structures. In particular, it collects signals from a cells’ surroundings and changes where cells divide so new cells are produced in the right places. The control of cell division by PTEN is also thought to help limit the progression and spread of cancer. PTEN can interact with another protein called β-Arrestin1, which behaves as a so-called scaffolding protein – in other words, one that helps groups of proteins to interact with each other. β-Arrestin1 has been found to control cell division via a series of other proteins, including ARHGAP21 and Cdc42. The relationship between PTEN and these other proteins in dividing cells is still not fully understood. Javadi, Deevi et al. studied PTEN in human cells grown in the laboratory to show that a part of PTEN known as the C2 domain allows it to help organize cells by moving β-Arrestin1 to the outer edge of the cell – the cell membrane. This relocation allows β-Arrestin1 to interact with ARHGAP21 and Cdc42, and control cell division. Active Cdc42 changes the orientation of cell division, allowing cells to organize into single layers of regular cells and similar tightly controlled structures. Further experiments revealed that these proteins are important to form tubes inside the glands of the gut. The C2 region of PTEN also helps to detect signals carried by fat molecules in the cell membrane, so these results provide a direct link between signaling and cell organization via PTEN. The work of Javadi, Deevi et al. provides new understanding of how PTEN links nutrient availability to cell organization during development and may also lead to new insights into the role of PTEN in limiting the growth of tumors.
- Published
- 2017
10. CXCR7 participates in CXCL12-induced CD34+ cell cycling through β-arrestin–dependent Akt activation
- Author
-
Mark G.H. Scott, Stefano Marullo, Marie-Caroline Le Bousse-Kerdilès, Laetitia Boutin, Bernadette Guerton, Denis Clay, Aurélie Chabanon, Adrienne Anginot, Frédéric Torossian, Christophe Desterke, and Jean-Jacques Lataillade
- Subjects
Arrestins ,Cell Survival ,Immunology ,Cell ,Intracellular Space ,Antigens, CD34 ,Biology ,Biochemistry ,Colony-Forming Units Assay ,medicine ,Humans ,Phosphorylation ,Progenitor cell ,Receptor ,Protein kinase B ,beta-Arrestins ,Receptors, CXCR ,Beta-Arrestins ,Cell Cycle ,Cell Biology ,Hematology ,Cell cycle ,Hematopoietic Stem Cells ,Chemokine CXCL12 ,biological factors ,Cell biology ,Enzyme Activation ,Protein Transport ,Haematopoiesis ,medicine.anatomical_structure ,embryonic structures ,Cancer research ,biological phenomena, cell phenomena, and immunity ,Proto-Oncogene Proteins c-akt ,Protein Binding ,Homing (hematopoietic) - Abstract
In addition to its well-known effect on migration and homing of hematopoietic stem/progenitor cells (HSPCs), CXCL12 chemokine also exhibits a cell cycle and survival-promoting factor for human CD34(+) HSPCs. CXCR4 was suggested to be responsible for CXCL12-induced biological effects until the recent discovery of its second receptor, CXCR7. Until now, the participation of CXCR7 in CXCL12-induced HSPC cycling and survival is unknown. We show here that CXCL12 was capable of binding CXCR7 despite its scarce expression at CD34(+) cell surface. Blocking CXCR7 inhibited CXCL12-induced Akt activation as well as the percentage of CD34(+) cells in cycle, colony formation, and survival, demonstrating its participation in CXCL12-induced functional effects in HSPCs. At steady state, CXCR7 and β-arrestin2 co-localized near the plasma membrane of CD34(+) cells. After CXCL12 treatment, β-arrestin2 translocated to the nucleus, and this required both CXCR7 and CXCR4. Silencing β-arrestin expression decreased CXCL12-induced Akt activation in CD34(+) cells. Our results demonstrate for the first time the role of CXCR7, complementary to that played by CXCR4, in the control of HSPC cycling, survival, and colony formation induced by CXCL12. We also provide evidence for the involvement of β-arrestins as signaling hubs downstream of both CXCL12 receptors in primary human HSPCs.
- Published
- 2014
11. Distinct functional outputs of PTEN signalling are controlled by dynamic association with β-arrestins
- Author
-
Emeline Camand, Alexandre Benmerah, Mark G.H. Scott, Cédric Boularan, Stefano Marullo, Larissa Kotelevets, Lucien C.D. Gibson, Sandrine Etienne-Manneville, Evelyne Lima-Fernandes, Lamia Achour, George S. Baillie, Hervé Enslen, Eric Chastre, and Julie A. Pitcher
- Subjects
Scaffold protein ,RHOA ,General Immunology and Microbiology ,Beta-Arrestins ,General Neuroscience ,Cell migration ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,Lipid phosphatase activity ,Cancer research ,biology.protein ,PTEN ,Tensin ,Molecular Biology ,Protein kinase B - Abstract
The tumour suppressor PTEN (phosphatase and tensin deleted on chromosome 10) regulates major cellular functions via lipid phosphatase-dependent and -independent mechanisms. Despite its fundamental pathophysiological importance, how PTEN's cellular activity is regulated has only been partially elucidated. We report that the scaffolding proteins β-arrestins (β-arrs) are important regulators of PTEN. Downstream of receptor-activated RhoA/ROCK signalling, β-arrs activate the lipid phosphatase activity of PTEN to negatively regulate Akt and cell proliferation. In contrast, following wound-induced RhoA activation, β-arrs inhibit the lipid phosphatase-independent anti-migratory effects of PTEN. β-arrs can thus differentially control distinct functional outputs of PTEN important for cell proliferation and migration.
- Published
- 2011
12. Meningococcus Hijacks a β2-Adrenoceptor/β-Arrestin Pathway to Cross Brain Microvasculature Endothelium
- Author
-
Xavier Nassif, Mathieu Coureuil, Hervé Enslen, Mark G.H. Scott, Sandrine Bourdoulous, Cédric Boularan, Hervé Lécuyer, Stefano Marullo, Magali Soyer, and Guillain Mikaty
- Subjects
Endothelium ,Arrestins ,Neisseria meningitidis ,Biology ,Blood–brain barrier ,Cell junction ,Bacterial Adhesion ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Receptors, Adrenergic, beta ,Arrestin ,medicine ,Humans ,beta-Arrestins ,030304 developmental biology ,0303 health sciences ,Biochemistry, Genetics and Molecular Biology(all) ,Beta-Arrestins ,Brain ,Endothelial Cells ,3. Good health ,Cell biology ,Meningococcal Infections ,medicine.anatomical_structure ,Blood-Brain Barrier ,Signal transduction ,Tyrosine kinase ,030217 neurology & neurosurgery ,Signal Transduction ,Proto-oncogene tyrosine-protein kinase Src - Abstract
SummaryFollowing pilus-mediated adhesion to human brain endothelial cells, meningococcus (N. meningitidis), the bacterium causing cerebrospinal meningitis, initiates signaling cascades, which eventually result in the opening of intercellular junctions, allowing meningeal colonization. The signaling receptor activated by the pathogen remained unknown. We report that N. meningitidis specifically stimulates a biased β2-adrenoceptor/β-arrestin signaling pathway in endothelial cells, which ultimately traps β-arrestin-interacting partners, such as the Src tyrosine kinase and junctional proteins, under bacterial colonies. Cytoskeletal reorganization mediated by β-arrestin-activated Src stabilizes bacterial adhesion to endothelial cells, whereas β-arrestin-dependent delocalization of junctional proteins results in anatomical gaps used by bacteria to penetrate into tissues. Activation of β-adrenoceptor endocytosis with specific agonists prevents signaling events downstream of N. meningitidis adhesion and inhibits bacterial crossing of the endothelial barrier. The identification of the mechanism used for hijacking host cell signaling machineries opens perspectives for treatment and prevention of meningococcal infection.PaperFlick
- Published
- 2010
13. CD4-CCR5 interaction in intracellular compartments contributes to receptor expression at the cell surface
- Author
-
Georges Bismuth, Mark G.H. Scott, Catherine Labbé-Jullié, Lamia Achour, Hamasseh Shirvani, Stefano Marullo, Alain Thuret, Marullo, Stefano, Institut Cochin (UMR_S567 / UMR 8104), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), and Université Paris Descartes - Paris 5 (UPD5) - Institut National de la Santé et de la Recherche Médicale (INSERM) - Centre National de la Recherche Scientifique (CNRS)
- Subjects
MESH : Hela Cells ,Chemokine receptor CCR5 ,viruses ,Receptor expression ,MESH: Antigens, CD4 ,Intracellular Space ,MESH : Intracellular Space ,MESH: Cricetinae ,Plasma protein binding ,MESH: Receptors, CCR5 ,Endoplasmic Reticulum ,Biochemistry ,MESH: Cell Compartmentation ,MESH : Protein Transport ,MESH: Cricetulus ,Cell–cell interaction ,Cricetinae ,MESH : CHO Cells ,MESH: Animals ,Receptor ,Cells, Cultured ,MESH : Endoplasmic Reticulum ,biology ,MESH : Cricetinae ,virus diseases ,MESH : Protein Binding ,MESH : Receptors, CCR5 ,Hematology ,Cell biology ,Protein Transport ,Antigens, Surface ,CD4 Antigens ,MESH: Intracellular Space ,MESH : Antigens, Surface ,Intracellular ,MESH: Cells, Cultured ,Protein Binding ,MESH: Protein Transport ,Receptors, CCR5 ,MESH: Antigens, Surface ,MESH : Cricetulus ,Immunology ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,CHO Cells ,Article ,Cricetulus ,MESH: CHO Cells ,MESH: Endoplasmic Reticulum ,MESH : Cells, Cultured ,MESH: Protein Binding ,Animals ,Humans ,MESH : Cell Compartmentation ,[SDV.BC] Life Sciences [q-bio]/Cellular Biology ,MESH: Humans ,Endoplasmic reticulum ,MESH : Humans ,Cell Biology ,Cell Compartmentation ,MESH: Hela Cells ,biology.protein ,MESH : Antigens, CD4 ,MESH : Animals ,CC chemokine receptors ,HeLa Cells - Abstract
International audience; The association of CD4, a glycoprotein involved in T-cell development and antigen recognition, and CC chemokine receptor 5 (CCR5), a chemotactic G protein-coupled receptor, which regulates trafficking and effector functions of immune cells, forms the main receptor for HIV. We observed that the majority of CCR5 is maintained within the intracellular compartments of primary T lymphocytes and in a monocytic cell line, contrasting with its relatively low density at the cell surface. The CCR5-CD4 association, which occurs in the endoplasmic reticulum, enhanced CCR5 export to the plasma membrane in a concentration-dependent manner, whereas inhibition of endogenous CD4 with small interfering RNAs decreased cell-surface expression of endogenous CCR5. This effect was specific for CCR5, as CD4 did not affect cellular distribution of CXCR4, the other HIV coreceptor. These results reveal a previously unappreciated role of CD4, which contributes to regulating CCR5 export to the plasma membrane.
- Published
- 2009
14. β-arrestin 2 oligomerization controls the Mdm2-dependent inhibition of p53
- Author
-
Alexandre Benmerah, Alain Thuret, Stefano Marullo, Mark G.H. Scott, Emmanuel Esteve, Karima Bourougaa, Marc Tramier, Catherine Labbé-Jullié, Maïté Coppey-Moisan, Myriam Bellal, Robin Fåhraeus, Cédric Boularan, Institut Cochin (UMR_S567 / UMR 8104), Université Paris Descartes - Paris 5 (UPD5) - Institut National de la Santé et de la Recherche Médicale (INSERM) - Centre National de la Recherche Scientifique (CNRS), Cibles Moleculaires Dans les Cancers, Université Paris Diderot - Paris 7 (UPD7) - Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Jacques Monod (IJM), Université Paris Diderot - Paris 7 (UPD7) - Centre National de la Recherche Scientifique (CNRS), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Laboratoire de pharmacologie expérimentale et clinique : cibles moléculaires en cancérologie ((U 716)), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Université Paris Diderot - Paris 7 (UPD7)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut Jacques Monod (IJM (UMR_7592)), Université Paris Diderot - Paris 7 (UPD7)-Centre National de la Recherche Scientifique (CNRS), and Marullo, Stefano
- Subjects
beta-arrestin 1 ,Arrestins ,Beta-Arrestin-2 ,chemistry.chemical_compound ,Biopolymers ,Chlorocebus aethiops ,MESH: Animals ,Inositol ,MESH: Tumor Suppressor Protein p53 ,Receptor ,beta-Arrestins ,0303 health sciences ,Multidisciplinary ,COS cells ,030302 biochemistry & molecular biology ,nuclear export signal ,Proto-Oncogene Proteins c-mdm2 ,Biological Sciences ,beta-Arrestin 2 ,ß-arrs ,Cell biology ,MESH: COS Cells ,MESH: Biopolymers ,Biochemistry ,COS Cells ,MESH: Arrestins ,Intracellular ,Agonist ,6-hexakisphosphate ,Phytic Acid ,medicine.drug_class ,Bioluminescence Resonance Energy Transfer ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Biology ,Cell Line ,03 medical and health sciences ,MESH: Proto-Oncogene Proteins c-mdm2 ,medicine ,Animals ,Humans ,MESH: Phytic Acid ,Binding site ,[SDV.BC] Life Sciences [q-bio]/Cellular Biology ,030304 developmental biology ,ß-arr1 ,MESH: Humans ,IP6 ,Binding Sites ,Beta-Arrestins ,MESH: Cercopithecus aethiops ,MESH: Cell Line ,ß-arr2 ,MESH: Binding Sites ,chemistry ,NES ,BRET ,Tumor Suppressor Protein p53 ,inositol 1 - Abstract
International audience; beta-arrestins (beta-arrs), two ubiquitous proteins involved in serpentine heptahelical receptor regulation and signaling, form constitutive homo- and heterooligomers stabilized by inositol 1,2,3,4,5,6-hexakisphosphate (IP6). Monomeric beta-arrs are believed to interact with receptors after agonist activation, and therefore, beta-arr oligomers have been proposed to represent a resting biologically inactive state. In contrast to this, we report here that the interaction with and subsequent titration out of the nucleus of the protooncogene Mdm2 specifically require beta-arr2 oligomers together with the previously characterized nucleocytoplasmic shuttling of beta-arr2. Mutation of the IP6-binding sites impair oligomerization, reduce interaction with Mdm2, and inhibit p53-dependent antiproliferative effects of beta-arr2, whereas the competence for receptor regulation and signaling is maintained. These observations suggest that the intracellular concentration of beta-arr2 oligomers might control cell survival and proliferation.
- Published
- 2007
15. Cooperative Regulation of Extracellular Signal-Regulated Kinase Activation and Cell Shape Change by Filamin A and β-Arrestins
- Author
-
Alain Thuret, Olivier Muntaner, Mark G.H. Scott, Vincenzo Pierotti, Julie A. Pitcher, Stephano Marullo, Hélène Storez, Catherine Labbé-Jullié, Erika Lindberg, Laboratory for Molecular Cell Biology and Department of Pharmacology, University College of London [London] (UCL) - MRC, Institut Cochin (UMR_S567 / UMR 8104), Université Paris Descartes - Paris 5 (UPD5) - Institut National de la Santé et de la Recherche Médicale (INSERM) - Centre National de la Recherche Scientifique (CNRS), and Wellcome Trust, the Agence Nationale pour la Recherche sur le SIDA, Ligue Contre le Cancer (Comité de l'Oise), CNRS, and INSERM. During the performance of this work, M.G.H.S. was supported by postdoctoral fellowships from the Wellcome Trust, Fondation pou
- Subjects
MAPK/ERK pathway ,Arrestins ,ruffle ,Filamin ,two hybrid ,Contractile Proteins ,0302 clinical medicine ,Protein Interaction Mapping ,beta arrestin ,FLNA ,Cells, Cultured ,beta-Arrestins ,Mitogen-Activated Protein Kinase 1 ,0303 health sciences ,Microfilament Proteins ,cytoskeleton ,Articles ,filamin ,Cell biology ,Repetitive Sequences, Amino Acid ,Filamins ,Molecular Sequence Data ,G protein coupled receptor ,Biology ,Receptor, Angiotensin, Type 1 ,03 medical and health sciences ,Two-Hybrid System Techniques ,Animals ,Humans ,Immunoprecipitation ,Amino Acid Sequence ,[SDV.BC] Life Sciences [q-bio]/Cellular Biology ,Cell Shape ,Molecular Biology ,030304 developmental biology ,G protein-coupled receptor ,Binding Sites ,Beta-Arrestins ,Cell Membrane ,Receptor, Muscarinic M1 ,Actin cytoskeleton reorganization ,Cell Biology ,Actin cytoskeleton ,MAPK ,Angiotensin II ,Actins ,Protein Structure, Tertiary ,Enzyme Activation ,Cytoskeletal Proteins ,030217 neurology & neurosurgery - Abstract
14 pages; beta-Arrestins (betaarr) are multifunctional adaptor proteins that can act as scaffolds for G protein-coupled receptor activation of mitogen-activated protein kinases (MAPK). Here, we identify the actin-binding and scaffolding protein filamin A (FLNA) as a betaarr-binding partner using Son of sevenless recruitment system screening, a classical yeast two-hybrid system, coimmunoprecipitation analyses, and direct binding in vitro. In FLNA, the betaarr-binding site involves tandem repeat 22 in the carboxyl terminus. betaarr binds FLNA through both its N- and C-terminal domains, indicating the presence of multiple binding sites. We demonstrate that betaarr and FLNA act cooperatively to activate the MAPK extracellular signal-regulated kinase (ERK) downstream of activated muscarinic M1 (M1MR) and angiotensin II type 1a (AT1AR) receptors and provide experimental evidence indicating that this phenomenon is due to the facilitation of betaarr-ERK2 complex formation by FLNA. In Hep2 cells, stimulation of M1MR or AT1AR results in the colocalization of receptor, betaarr, FLNA, and active ERK in membrane ruffles. Reduction of endogenous levels of betaarr or FLNA and a catalytically inactive dominant negative MEK1, which prevents ERK activation, inhibit membrane ruffle formation, indicating the functional requirement for betaarr, FLNA, and active ERK in this process. Our results indicate that betaarr and FLNA cooperate to regulate ERK activation and actin cytoskeleton reorganization.
- Published
- 2006
16. Phosphorylation and Chronic Agonist Treatment Atypically Modulate GABAB Receptor Cell Surface Stability
- Author
-
Andrew R. Calver, Benjamin P. Fairfax, Andrés Couve, Stephen J. Moss, Mark G.H. Scott, Julie A. Pitcher, and Menelas N. Pangalos
- Subjects
Baclofen ,Time Factors ,Hippocampus ,Biochemistry ,Cyclic AMP ,Phosphorylation ,Receptor ,Cells, Cultured ,Cerebral Cortex ,Neurons ,Arrestin ,Temperature ,Endocytosis ,Cell biology ,COS Cells ,Signal transduction ,Dimerization ,Plasmids ,Protein Binding ,Agonist ,medicine.medical_specialty ,DNA, Complementary ,medicine.drug_class ,GABAB receptor ,Biology ,Cell Line ,Internal medicine ,Receptors, Adrenergic, beta ,medicine ,Animals ,Humans ,Biotinylation ,GABA Agonists ,Molecular Biology ,GABA-B Receptor Agonists ,Cell Membrane ,Cell Biology ,Cyclic AMP-Dependent Protein Kinases ,Precipitin Tests ,Rats ,Enzyme Activation ,Metabotropic receptor ,Endocrinology ,Microscopy, Fluorescence ,Receptors, GABA-B ,nervous system ,Calcium - Abstract
GABA(B) receptors are heterodimeric G protein-coupled receptors that mediate slow synaptic inhibition in the central nervous system. The dynamic control of the cell surface stability of GABA(B) receptors is likely to be of fundamental importance in the modulation of receptor signaling. Presently, however, this process is poorly understood. Here we demonstrate that GABA(B) receptors are remarkably stable at the plasma membrane showing little basal endocytosis in cultured cortical and hippocampal neurons. In addition, we show that exposure to baclofen, a well characterized GABA(B) receptor agonist, fails to enhance GABA(B) receptor endocytosis. Lack of receptor internalization in neurons correlates with an absence of agonist-induced phosphorylation and lack of arrestin recruitment in heterologous systems. We also demonstrate that chronic exposure to baclofen selectively promotes endocytosis-independent GABA(B) receptor degradation. The effect of baclofen can be attenuated by activation of cAMP-dependent protein kinase or co-stimulation of beta-adrenergic receptors. Furthermore, we show that increased degradation rates are correlated with reduced receptor phosphorylation at serine 892 in GABA(B)R2. Our results support a model in which GABA(B)R2 phosphorylation specifically stabilizes surface GABA(B) receptors in neurons. We propose that signaling pathways that regulate cAMP levels in neurons may have profound effects on the tonic synaptic inhibition by modulating the availability of GABA(B) receptors.
- Published
- 2004
17. Nuclear Functions for Plasma Membrane-Associated Proteins?
- Author
-
Viviane Poupon, Stefano Marullo, Alexandre Benmerah, and Mark G.H. Scott
- Subjects
Endocytic cycle ,Cell Biology ,Biology ,Endocytosis ,Biochemistry ,Cell biology ,Cell membrane ,Cell nucleus ,medicine.anatomical_structure ,Membrane protein ,Structural Biology ,Genetics ,medicine ,Nuclear transport ,Nuclear pore ,Molecular Biology ,Lamin - Abstract
There are a growing number of observations that proteins, which were initially thought to perform a specific function in a given subcellular compartment, may also play additional roles in different locations within the cell. Proteins found in adhesion and endocytic structures of the plasma membrane and which also traffic to the nucleus perhaps represent the more spectacular examples of this phenomenon. The mechanisms involved in the transport of these molecules through the nuclear pores and their potential nuclear functions are discussed.
- Published
- 2003
18. Differential Nucleocytoplasmic Shuttling of β-Arrestins
- Author
-
Stefano Marullo, Erwann Le Rouzic, Mark G.H. Scott, Axel Périanin, Vincenzo Pierotti, Alexandre Benmerah, Serge Benichou, and Hervé Enslen
- Subjects
Scaffold protein ,G protein ,Cell Biology ,Leptomycin ,Biology ,Biochemistry ,Clathrin ,Cell biology ,Cytosol ,chemistry.chemical_compound ,chemistry ,Cytoplasm ,biology.protein ,Nuclear transport ,Nuclear export signal ,Molecular Biology - Abstract
β-arrestins (βarrs) are two highly homologous proteins that uncouple G protein-coupled receptors from their cognate G proteins, serve as adaptor molecules linking G protein-coupled receptors to clathrin-coat components (AP-2 complex and clathrin), and act as scaffolding proteins for ERK1/2 and JNK3 cascades. A striking difference between the two βarrs (βarr1 and βarr2) is that βarr1 is evenly distributed throughout the cell, whereas βarr2 shows an apparent cytoplasmic localization at steady state. Here, we investigate the molecular determinants underlying this differential distribution. βarr2 is constitutively excluded from the nucleus by a leptomycin B-sensitive pathway because of the presence of a classical leucine-rich nuclear export signal in its C terminus (L395/L397) that is absent in βarr1. In addition, using a nuclear import assay in yeast we showed that βarr2 is actively imported into the nucleus, suggesting that βarr2 undergoes constitutive nucleocytoplasmic shuttling. In cells expressing βarr2, JNK3 is mostly cytosolic. A point mutation of the nuclear export signal (L395A) in βarr2, which was sufficient to redistribute βarr2 from the cytosol to the nucleus, also caused the nuclear relocalization of JNK3. These data indicate that the nucleocytoplasmic shuttling of βarr2 controls the subcellular distribution of JNK3.
- Published
- 2002
19. Identification of novel polymorphisms within the promoter region of the humanβ2adrenergic receptor gene
- Author
-
Ian P. Hall, Mark G.H. Scott, Caroline Swan, and Amanda Wheatley
- Subjects
Pharmacology ,Genetics ,Start codon ,5' flanking region ,Gene expression ,Adrenergic ,Promoter ,Luciferase ,Transfection ,Biology ,Gene ,Molecular biology - Abstract
By screening the 1470 bp 5' to the start codon of the human beta2 adrenergic receptor gene, we have identified a total of eight polymorphisms (-20 T-->C, -47 T-->C, -367 T-->C, -468 C-->G, -654 G-->A, -1023 G-->A, -1343 A-->G and -1429 T-->A c.f. beta2 adrenergic receptor start codon). Transient transfection of 5' flanking deletion luciferase reporter constructs demonstrated the majority of activity of the human beta2 adrenergic gene 5' flanking region to be present within a 549 bp fragment immediately upstream from the start codon. Because of linkage disequilibrium, some combinations of polymorphisms were particularly frequent. We transiently transfected COS-7 cells with luciferase constructs under the control of the 549 bp of 5' flanking DNA containing the two most frequent extended haplotypes in this region. Luciferase activity was significantly reduced in cells transfected with the 'mutant' construct (-20C, -47C, -367C, -468G) c.f. the 'wild-type' construct (-20T, -47T, -367T, -468C). These data suggest that polymorphisms have the potential to alter human beta2 adrenergic receptor gene expression.
- Published
- 1999
20. Arrestins as Regulatory Hubs in Cancer Signalling Pathways
- Author
-
Mark G.H. Scott, Evelyne Lima-Fernandes, and Hervé Enslen
- Subjects
genetic structures ,biology ,Cancer ,Endocytosis ,medicine.disease ,biology.protein ,medicine ,Mdm2 ,sense organs ,Receptor ,Protein kinase B ,Neuroscience ,Function (biology) ,PI3K/AKT/mTOR pathway ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Non-visual arrestins were initially appreciated for the roles they play in the negative regulation of G protein-coupled receptors through the processes of desensitisation and endocytosis. The arrestins are also now known as protein scaffolding platforms that act downstream of multiple types of receptors, ensuring relevant transmission of information for an appropriate cellular response. They function as regulatory hubs in several important signalling pathways that are often dysregulated in human cancers. Interestingly, several recent studies have documented changes in expression and localisation of arrestins that occur during cancer progression and that correlate with clinical outcome. Here, we discuss these advances and how changes in expression/localisation may affect functional outputs of arrestins in cancer biology.
- Published
- 2013
21. Secreted factors from brain endothelial cells maintain glioblastoma stem-like cell expansion through the mTOR pathway
- Author
-
Hervé Chneiweiss, Cécile Thirant, Armelle Le Guelte, Eva María Galán-Moya, Julie Dwyer, Julie Gavard, Mark G.H. Scott, Nicolas Bidère, Pierre-Olivier Couraud, Marie-Pierre Junier, and Evelyne Lima-Fernandes
- Subjects
endocrine system ,animal structures ,Pyridines ,Blotting, Western ,Biology ,Transfection ,Biochemistry ,Flow cytometry ,RNA interference ,Glioma ,Genetics ,medicine ,Humans ,RNA, Small Interfering ,Furans ,Molecular Biology ,PI3K/AKT/mTOR pathway ,Sirolimus ,medicine.diagnostic_test ,Stem Cells ,TOR Serine-Threonine Kinases ,fungi ,Scientific Reports ,RNA ,Brain ,Endothelial Cells ,medicine.disease ,Flow Cytometry ,Cell biology ,Blot ,Pyrimidines ,Microscopy, Fluorescence ,embryonic structures ,RNA Interference ,Stem cell ,Glioblastoma ,Signal Transduction - Abstract
Glioma stem-cells are associated with the brain vasculature. However, the way in which this vascular niche regulates stem-cell renewal and fate remains unclear. Here, we show that factors emanating from brain endothelial cells positively control the expansion of long-term glioblastoma stem-like cells. We find that both pharmacological inhibition of and RNA interference with the mammalian target of rapamycin (mTOR) pathway reduce their spheroid growth. Conversely, the endothelial secretome is sufficient to promote this mTOR-dependent survival. Thus, interfering with endothelial signals might present opportunities to identify treatments that selectively target malignant stem-cell niches.
- Published
- 2010
22. An escort for GPCRs: implications for regulation of receptor density at the cell surface
- Author
-
Lamia Achour, Stefano Marullo, Catherine Labbé-Jullié, Mark G.H. Scott, Institut Cochin (UMR_S567 / UMR 8104), Université Paris Descartes - Paris 5 (UPD5) - Institut National de la Santé et de la Recherche Médicale (INSERM) - Centre National de la Recherche Scientifique (CNRS), Marullo, Stefano, and Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
MESH: Protein Transport ,Context (language use) ,MESH: Receptors, G-Protein-Coupled ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Biology ,Toxicology ,Endoplasmic Reticulum ,Models, Biological ,Receptors, G-Protein-Coupled ,GPCR ,MESH: Endoplasmic Reticulum ,trafficking ,Humans ,chaperone ,Receptor ,[SDV.BC] Life Sciences [q-bio]/Cellular Biology ,Secretory pathway ,G protein-coupled receptor ,Pharmacology ,MESH: Humans ,Endoplasmic reticulum ,Cell Membrane ,MESH: Models, Biological ,regulation ,Cell biology ,Transport protein ,Protein Transport ,Chaperone (protein) ,biology.protein ,biosynthesis ,Intracellular ,MESH: Cell Membrane - Abstract
International audience; G-protein-coupled receptors (GPCRs) are dynamically regulated by various mechanisms that tune their response to external stimuli. Modulation of their plasma membrane density, via trafficking between subcellular compartments, constitutes an important process in this context. Substantial information has been accumulated on cellular pathways that remove GPCRs from the cell surface for subsequent degradation or recycling. In comparison, much less is known about the mechanisms controlling trafficking of neo-synthesized GPCRs from intracellular compartments to the cell surface. Although GPCR export to the plasma membrane is commonly considered to mostly implicate the default, unregulated secretory pathway, an increasing number of observations indicate that trafficking to the plasma membrane from the endoplasmic reticulum might be tightly regulated and involve specific protein partners. Moreover, a new paradigm is emerging in some cellular contexts, in which stocks of functional receptors retained within intracellular compartments can be rapidly mobilized to the plasma membrane to maintain sustained physiological responsiveness.
- Published
- 2008
23. Targeting of β-Arrestin2 to the Centrosome and Primary Cilium: Role in Cell Proliferation Control
- Author
-
Rania Ghossoub, Mark G.H. Scott, Alexandre Benmerah, Anne Burtey, Jean-Paul Concordet, Sophie Saunier, Cédric Boularan, Anahi Molla-Herman, Marion Zarka, Stefano Marullo, Institut Cochin (IC UM3 (UMR 8104 / U1016)), Centre National de la Recherche Scientifique (CNRS)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Imagine - Institut des maladies génétiques (IMAGINE - U1163), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Structure et Instabilité des Génomes (STRING), Muséum national d'Histoire naturelle (MNHN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut de Chimie du CNRS (INC)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), ANR-BLAN07-3-187842 ANR-BLAN07-3-187842, Association pour la recherche contre le cancer, Ligue Nationale contre le Cancer, Burtey, Anne, Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut Cochin (UMR_S567 / UMR 8104), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), University of Bergen (UiB), Université d'Oslo, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP), Muséum national d'Histoire naturelle (MNHN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), and ANR-BLAN07-3-187842,ANR-BLAN07-3-187842,ANR-BLAN07-3-187842
- Subjects
Axoneme ,[SDV.BIO]Life Sciences [q-bio]/Biotechnology ,Centrosomes ,Centriole ,Arrestins ,[SDV.IB.IMA]Life Sciences [q-bio]/Bioengineering/Imaging ,Cell Biology/Cell Growth and Division ,[SDV.SA.AGRO]Life Sciences [q-bio]/Agricultural sciences/Agronomy ,Kinesins ,lcsh:Medicine ,Microtubules ,Mice ,0302 clinical medicine ,[SDV.EE.ECO] Life Sciences [q-bio]/Ecology, environment/Ecosystems ,lcsh:Science ,beta-Arrestins ,ComputingMilieux_MISCELLANEOUS ,Centrioles ,0303 health sciences ,Multidisciplinary ,Cilium ,Cell Cycle ,Cell staining ,Cell biology ,Protein Transport ,Kinesin ,Research Article ,Protein Binding ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Biology ,Endocytosis ,Cell Line ,03 medical and health sciences ,[SDV.EE.ECO]Life Sciences [q-bio]/Ecology, environment/Ecosystems ,Cell Biology/Membranes and Sorting ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,Microtubule ,Cell Biology/Cytoskeleton ,[SDV.BBM] Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Animals ,Humans ,[SDV.EE.SANT] Life Sciences [q-bio]/Ecology, environment/Health ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Cilia ,[SDV.IB.BIO]Life Sciences [q-bio]/Bioengineering/Biomaterials ,[SDV.BC] Life Sciences [q-bio]/Cellular Biology ,Cell fusion ,Cell Proliferation ,030304 developmental biology ,G protein-coupled receptor ,Centrosome ,[SDV.SA.AGRO] Life Sciences [q-bio]/Agricultural sciences/Agronomy ,[SDV.EE.SANT]Life Sciences [q-bio]/Ecology, environment/Health ,[SDV.BA.MVSA]Life Sciences [q-bio]/Animal biology/Veterinary medicine and animal Health ,Beta-Arrestins ,lcsh:R ,[SDV.BA.MVSA] Life Sciences [q-bio]/Animal biology/Veterinary medicine and animal Health ,DAPI staining ,Cell Biology ,[SDV.EE.IEO] Life Sciences [q-bio]/Ecology, environment/Symbiosis ,Rats ,[SDV.BIO] Life Sciences [q-bio]/Biotechnology ,[SDV.IB.BIO] Life Sciences [q-bio]/Bioengineering/Biomaterials ,Cell cycle and cell division ,[SDV.AEN] Life Sciences [q-bio]/Food and Nutrition ,[SDV.IB.IMA] Life Sciences [q-bio]/Bioengineering/Imaging ,14-3-3 Proteins ,Nuclear staining ,lcsh:Q ,[SDV.AEN]Life Sciences [q-bio]/Food and Nutrition ,030217 neurology & neurosurgery ,[SDV.EE.IEO]Life Sciences [q-bio]/Ecology, environment/Symbiosis - Abstract
International audience; Background: The primary cilium is a sensory organelle generated from the centrosome in quiescent cells and found at the surface of most cell types, from where it controls important physiological processes. Specific sets of membrane proteins involved in sensing the extracellular milieu are concentrated within cilia, including G protein coupled receptors (GPCRs). Most GPCRs are regulated by b-arrestins, barr1 and barr2, which control both their signalling and endocytosis, suggesting that barrs may also function at primary cilium.Methodology/Principal Findings: In cycling cells, βarr2 was observed at the centrosome, at the proximal region of the centrioles, in a microtubule independent manner. However, βarr2 did not appear to be involved in classical centrosome-associated functions. In quiescent cells, both in vitro and in vivo, βarr2 was found at the basal body and axoneme of primary cilia. Interestingly, βarr2 was found to interact and colocalize with 14-3-3 proteins and Kif3A, two proteins known to be involved in ciliogenesis and intraciliary transport. In addition, as suggested for other centrosome or cilia-associated proteins, βarrs appear to control cell cycle progression. Indeed, cells lacking βarr2 were unable to properly respond to serum starvation and formed less primary cilia in these conditions.Conclusions/Significance: Our results show that βarr2 is localized to the centrosome in cycling cells and to the primary cilium in quiescent cells, a feature shared with other proteins known to be involved in ciliogenesis or primary cilium function. Within cilia, βarr2 may participate in the signaling of cilia-associated GPCRs and, therefore, in the sensory functions of this cell “antenna”.
- Published
- 2008
24. The Conserved Isoleucine-Valine-Phenylalanine Motif Couples Activation State and Endocytic Functions of β-Arrestins
- Author
-
Joshua Z. Rappoport, Alexandre Benmerah, Sanford M. Simon, Stefano Marullo, Marijn G. J. Ford, Anne Burtey, Mark G.H. Scott, Eva M. Schmid, Harvey T. McMahon, Institut Cochin (IC UM3 (UMR 8104 / U1016)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP), University of Bergen (UiB), Université d'Oslo, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Potsdam Institute for Climate Impact Research (PIK), Laboratory of Cellular Biophysics, Rockefeller University [New York], Institut Cochin (UMR_S567 / UMR 8104), and Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
[SDV.BIO]Life Sciences [q-bio]/Biotechnology ,Arrestins ,[SDV.IB.IMA]Life Sciences [q-bio]/Bioengineering/Imaging ,Phenylalanine ,Clathrin adaptor complex ,Amino Acid Motifs ,Molecular Sequence Data ,Endocytic cycle ,[SDV.SA.AGRO]Life Sciences [q-bio]/Agricultural sciences/Agronomy ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Biochemistry ,Clathrin ,Receptors, G-Protein-Coupled ,03 medical and health sciences ,[SDV.EE.ECO]Life Sciences [q-bio]/Ecology, environment/Ecosystems ,Structural Biology ,Chlorocebus aethiops ,Genetics ,Arrestin ,Animals ,Humans ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Amino Acid Sequence ,Isoleucine ,Binding site ,[SDV.IB.BIO]Life Sciences [q-bio]/Bioengineering/Biomaterials ,Molecular Biology ,Conserved Sequence ,beta-Arrestins ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology ,G protein-coupled receptor ,[SDV.EE.SANT]Life Sciences [q-bio]/Ecology, environment/Health ,0303 health sciences ,[SDV.BA.MVSA]Life Sciences [q-bio]/Animal biology/Veterinary medicine and animal Health ,Sequence Homology, Amino Acid ,biology ,030302 biochemistry & molecular biology ,Signal transducing adaptor protein ,Valine ,Cell Biology ,Cell biology ,COS Cells ,biology.protein ,Clathrin adaptor proteins ,[SDV.AEN]Life Sciences [q-bio]/Food and Nutrition ,HeLa Cells ,[SDV.EE.IEO]Life Sciences [q-bio]/Ecology, environment/Symbiosis - Abstract
Beta-arrestins (betaarrs) play a central role in the regulation of G-protein-coupled receptors (GPCRs). Their binding to phosphorylated activated GPCRs induces a conformational transition to an active state resulting in the release of their flexible C-terminal tail. Binding sites for clathrin and the adaptor protein (AP)-2 clathrin adaptor complex are then unmasked, which drive the recruitment of betaarrs-GPCR complexes into clathrin-coated pits (CCPs). A conserved isoleucine-valine-phenylalanine (IVF) motif of the C-terminal tail controls betaarr activation through intramolecular interactions. Here, we provide structural, biochemical and functional evidence in living cells that the IVF motif also controls binding to AP-2. While the F residue is directly involved in AP-2 binding, substitutions of I and V residues, markedly enhanced affinity for AP-2 resulting in active betaarr mutants, which are constitutively targeted to CCPs in the absence of any GPCR activation. Conformational change and endocytic functions of betaarrs thus appear to be coordinated via the complex molecular interactions established by the IVF motif.
- Published
- 2007
25. Homo- and Hetero-oligomerization of β-Arrestins in Living Cells
- Author
-
Michel Bouvier, Stefano Marullo, Hassan Issafras, Alexandre Benmerah, Anne Burtey, Mark G.H. Scott, Olivier Muntaner, Maïté Coppey-Moisan, Tristan Piolot, Marc Tramier, Catherine Labbé-Jullié, Hélène Storez, Institut Cochin (UMR_S567 / UMR 8104), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut Jacques Monod (IJM (UMR_7592)), Université Paris Diderot - Paris 7 (UPD7)-Centre National de la Recherche Scientifique (CNRS), Université de Montréal (UdeM), Burtey, Anne, Université Paris Descartes - Paris 5 (UPD5) - Institut National de la Santé et de la Recherche Médicale (INSERM) - Centre National de la Recherche Scientifique (CNRS), Institut Jacques Monod (IJM), Université Paris Diderot - Paris 7 (UPD7) - Centre National de la Recherche Scientifique (CNRS), and Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Descartes - Paris 5 (UPD5)
- Subjects
Gene isoform ,genetic structures ,Arrestins ,Genetic Vectors ,Green Fluorescent Proteins ,[SDV.BBM.BP] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biophysics ,Biology ,[SDV.BC.IC] Life Sciences [q-bio]/Cellular Biology/Cell Behavior [q-bio.CB] ,Biochemistry ,Models, Biological ,Retina ,03 medical and health sciences ,0302 clinical medicine ,Chlorocebus aethiops ,[SDV.BC.IC]Life Sciences [q-bio]/Cellular Biology/Cell Behavior [q-bio.CB] ,medicine ,Arrestin ,Fluorescence Resonance Energy Transfer ,Animals ,Immunoprecipitation ,Protein Isoforms ,[SDV.BBM.BC]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biochemistry [q-bio.BM] ,Receptor ,Molecular Biology ,[SDV.BBM.BC] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biochemistry [q-bio.BM] ,beta-Arrestins ,030304 developmental biology ,G protein-coupled receptor ,Cell Nucleus ,0303 health sciences ,Dose-Response Relationship, Drug ,Cell Biology ,eye diseases ,Cell biology ,[SDV.BBM.BC]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biomolecules [q-bio.BM] ,[SDV.BBM.BP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biophysics ,medicine.anatomical_structure ,Förster resonance energy transfer ,Microscopy, Fluorescence ,COS Cells ,sense organs ,Signal transduction ,Nucleus ,Dimerization ,030217 neurology & neurosurgery ,Function (biology) ,Protein Binding ,Signal Transduction - Abstract
International audience; Arrestins are important proteins, which regulate the function of serpentine heptahelical receptors and contribute to multiple signaling pathways downstream of receptors. The ubiquitous β-arrestins are believed to function exclusively as monomers, although self-association is assumed to control the activity of visual arrestin in the retina, where this isoform is particularly abundant. Here the oligomerization status of β-arrestins was investigated using different approaches, including co-immunoprecipitation of epitope-tagged β-arrestins and resonance energy transfer (BRET and FRET) in living cells. At steady state and at physiological concentrations, β-arrestins constitutively form both homo- and hetero-oligomers. Co-expression of β-arrestin2 and β-arrestin1 prevented β-arrestin1 accumulation into the nucleus, suggesting that hetero-oligomerization may have functional consequences. Our data clearly indicate that β-arrestins can exist as homo- and hetero-oligomers in living cells and raise the hypothesis that the oligomeric state may regulate their subcellular distribution and functions.
- Published
- 2005
26. G Protein-Coupled Receptor Kinase 5 Contains a DNA-Binding Nuclear Localization Sequence
- Author
-
Mark G.H. Scott, Laura Johnson, and Julie A. Pitcher
- Subjects
G-Protein-Coupled Receptor Kinase 5 ,DNA, Complementary ,Calmodulin ,Molecular Sequence Data ,Active Transport, Cell Nucleus ,CHO Cells ,Protein Serine-Threonine Kinases ,Transfection ,Cell Line ,Cytosol ,Catalytic Domain ,Cricetinae ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Phosphorylation ,Nuclear export signal ,Molecular Biology ,Cell Growth and Development ,G protein-coupled receptor ,Cell Nucleus ,Receptor, Muscarinic M3 ,G protein-coupled receptor kinase ,Microscopy, Confocal ,biology ,Ionophores ,Sequence Homology, Amino Acid ,Cell Biology ,DNA ,Cell biology ,Cell nucleus ,medicine.anatomical_structure ,Biochemistry ,Microscopy, Fluorescence ,COS Cells ,Mutation ,biology.protein ,RNA ,Calcium ,Signal transduction ,Nuclear localization sequence ,Protein Binding ,Signal Transduction - Abstract
G protein-coupled receptor kinases (GRKs) mediate desensitization of agonist-occupied G protein-coupled receptors (GPCRs). Here we report that GRK5 contains a DNA-binding nuclear localization sequence (NLS) and that its nuclear localization is regulated by GPCR activation, results that suggest potential nuclear functions for GRK5. As assessed by fluorescence confocal microscopy, transfected and endogenous GRK5 is present in the nuclei of HEp2 cells. Mutation of basic residues in the catalytic domain of GRK5 (between amino acids 388 and 395) results in the nuclear exclusion of the mutant enzyme (GRK5(Delta)(NLS)), demonstrating that GRK5 contains a functional NLS. The nuclear localization of GRK5 is subject to dynamic regulation. Calcium ionophore treatment or activation of Gq-coupled muscarinic-M3 receptors promotes the nuclear export of the kinase in a Ca(2+)/calmodulin (Ca(2+)/CaM)-dependent fashion. Ca(2+)/CaM binding to the N-terminal CaM binding site of GRK5 mediates this effect. Furthermore, GRK5, but not GRK5(Delta)(NLS) or GRK2, binds specifically and directly to DNA in vitro. Consistent with their presence in the nuclei of transfected cells, all the GRK4, but not GRK2, subfamily members contain putative NLSs. These results suggest that the GRK4 subfamily of GRKs may play a signaling role in the nucleus and that GRK4 and GRK2 subfamily members perform divergent cellular functions.
- Published
- 2004
27. Recruitment of Activated G Protein-coupled Receptors to Pre-existing Clathrin-coated Pits in Living Cells
- Author
-
Stefano Marullo, Olivier Muntaner, Alexandre Benmerah, Mark G.H. Scott, Institut Cochin de Génétique Moléculaire [Paris] (CNRS UPR 0415 - ICGM), Hôpital Cochin [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Centre National de la Recherche Scientifique (CNRS), Interactions de l'épithélium intestinal avec les sytème immunitaire et les antigènes intraluminaux, Institut National de la Santé et de la Recherche Médicale (INSERM), and Benmerah, Alexandre
- Subjects
Arrestins ,Receptors, Cell Surface ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,[SDV.GEN] Life Sciences [q-bio]/Genetics ,Transfection ,Endocytosis ,Biochemistry ,Clathrin ,[SDV.MHEP.UN]Life Sciences [q-bio]/Human health and pathology/Urology and Nephrology ,Rhodopsin-like receptors ,HeLa ,Bacterial Proteins ,stomatognathic system ,GTP-Binding Proteins ,Genes, Reporter ,Cell surface receptor ,Receptors, Transferrin ,[SDV.BDD] Life Sciences [q-bio]/Development Biology ,Humans ,Receptor ,Thyrotropin-Releasing Hormone ,Molecular Biology ,[SDV.BDD]Life Sciences [q-bio]/Development Biology ,[SDV.BC] Life Sciences [q-bio]/Cellular Biology ,beta-Arrestins ,G protein-coupled receptor ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,biology ,Receptors, Thyrotropin-Releasing Hormone ,Clathrin-Coated Vesicles ,SUPERFAMILY ,Cell Biology ,biology.organism_classification ,[SDV.MHEP.UN] Life Sciences [q-bio]/Human health and pathology/Urology and Nephrology ,Recombinant Proteins ,Cell biology ,Kinetics ,Luminescent Proteins ,stomatognathic diseases ,biology.protein ,HeLa Cells ,Plasmids ,Subcellular Fractions - Abstract
International audience; The process of clathrin-mediated endocytosis tightly regulates signaling of the superfamily of seven-transmembrane G protein-coupled receptors (GPCRs). A fundamental question in the cell biology of membrane receptor endocytosis is whether activated receptors can initiate the formation of clathrin-coated pits (CPs) or whether they are simply mobilized to pre-existing CPs. Here, using various approaches, including a dynamic assay to monitor the distribution of CPs and GPCR-beta-arrestin complexes in live HeLa cells, we demonstrate for the first time that activated GPCRs do not initiate the de novo formation of CPs but instead are targeted to pre-existing CPs.
- Published
- 2002
28. Modulation of human airway smooth muscle proliferation by type 3 phosphodiesterase inhibition
- Author
-
Mark G.H. Scott, Charlotte K. Billington, Ian P. Hall, Sunil K. Joseph, Caroline Swan, and Timothy M. Jobson
- Subjects
Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Platelet-derived growth factor ,Physiology ,Phosphodiesterase Inhibitors ,Becaplermin ,Biology ,Guanidines ,chemistry.chemical_compound ,Physiology (medical) ,Internal medicine ,1-Methyl-3-isobutylxanthine ,medicine ,Cyclic AMP ,Humans ,Phosphodiesterase inhibitor ,Respiratory system ,Luciferases ,Cells, Cultured ,Platelet-Derived Growth Factor ,Cell growth ,Phosphoric Diester Hydrolases ,Muscle, Smooth ,Cell Biology ,Transfection ,Proto-Oncogene Proteins c-sis ,In vitro ,Cell biology ,Isoenzymes ,Pyridazines ,Trachea ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Enzyme inhibitor ,biology.protein ,Cell Division ,Respiratory tract ,Thymidine - Abstract
Elevation in cell cAMP content can inhibit mitogenic signaling in cultured human airway smooth muscle (HASM) cells. We studied the effects of the type 3-selective phosphodiesterase inhibitor siguazodan, the type 4-selective phosphodiesterase inhibitor rolipram, and the nonselective inhibitor 3-isobutyl-1-methylxanthine (IBMX) on proliferation of cultured HASM cells. At concentrations selective for the type 3 phosphodiesterase isoform, siguazodan inhibited both [3H]thymidine incorporation (IC502 μM) and the increase in cell number (10 μM; 64% reduction) induced by platelet-derived growth factor-BB (20 ng/ml). These effects were mimicked by IBMX. At concentrations selective for type 4 phosphodiesterase inhibition, rolipram was without effect. A 20-min exposure to siguazodan and rolipram did not increase whole cell cAMP levels. However, in HASM cells transfected with a cAMP-responsive luciferase reporter (p6CRE/Luc), increases in cAMP-driven luciferase expression were seen with siguazodan (3.9-fold) and IBMX (16.5-fold). These data suggest that inhibition of the type 3 phosphodiesterase isoform present in airway smooth muscle results in inhibition of mitogenic signaling, possibly through an increase in cAMP-driven gene expression.
- Published
- 1999
29. L’expansion effrénée des fonctions des bêta-arrestines investit le domaine du développement
- Author
-
Mark G.H. Scott and Stefano Marullo
- Subjects
G protein ,Chemistry ,Embryogenesis ,Arrestin ,General Medicine ,Membrane transport ,General Biochemistry, Genetics and Molecular Biology ,Transmembrane protein ,Cell biology - Published
- 2005
30. Effects of cholinoceptor and 5-hydroxytryptamine3 receptor antagonism on erythromycin-induced canine intestinal motility disruption and emesis
- Author
-
Xin Yu Qin, H. H. Thompson, M.A. Pilot, and Mark G.H. Scott
- Subjects
Agonist ,Atropine ,Male ,medicine.medical_specialty ,Metoclopramide ,medicine.drug_class ,Vomiting ,Hexamethonium Compounds ,Biology ,Hexamethonium ,Cholinergic Antagonists ,chemistry.chemical_compound ,Hexamethonium compound ,Bridged Bicyclo Compounds ,Dogs ,Internal medicine ,Intestine, Small ,medicine ,Animals ,Pharmacology ,Electromyography ,Muscle, Smooth ,Receptor antagonist ,Bridged Bicyclo Compounds, Heterocyclic ,Renzapride ,Erythromycin ,Endocrinology ,chemistry ,Benzamides ,Injections, Intravenous ,Female ,Serotonin Antagonists ,medicine.symptom ,Gastrointestinal Motility ,medicine.drug ,Muscle Contraction ,Tropanes ,Research Article - Abstract
1. Erythromycin administration is associated with gastrointestinal problems, disturbed gastrointestinal motility and emesis. This study in the dog investigates the underlying mechanisms. 2. Intestinal myoelectrical activity and the occurrence and latency of emesis were recorded in eight conscious dogs. All drugs were administered intravenously. 3. Erythromycin (7 mg kg-1) increased contractions of the proximal small intestine, and caused emesis in all fasted dogs and in 5 dogs after food. Atropine (50 mg kg-1 min-1) and hexamethonium (10 mg kg-1 h-1) partially inhibited the GI motility effects but did not significantly reduce emesis. 4. Metoclopramide at a high dose (2 mg kg-1 h-1) reduced the incidence of emesis in the presence of increased intestinal motility, but a low dose (150 micrograms kg-1 h-1) was ineffective. 5. A 5-hydroxytryptamine3 (5-HT3) receptor antagonist, MDL 72222 (1 mg kg-1), reduced emesis when given alone and combined with metoclopramide (low dose). The 5-HT4 receptor agonist BRL24924 (Renzapride, 1 mg kg-1) had no effect on emesis either alone in combination with metoclopramide. 6. In conclusion, erythromycin-induced GI motility disturbances and emesis are not causally related. Whereas the increase in intestinal smooth muscle activity is possibly cholinergically mediated, emesis occurs at least in part via a 5-hydroxytryptaminergic mechanism, but does not involve the dopamine system.
- Published
- 1993
31. Endocytose des récepteurs couplés aux protéines G
- Author
-
Stefano Marullo, Alexandre Benmerah, Mark G.H. Scott, Institut Cochin (IC UM3 (UMR 8104 / U1016)), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), and Benmerah, Alexandre
- Subjects
[SDV.GEN]Life Sciences [q-bio]/Genetics ,[SDV.BDD] Life Sciences [q-bio]/Development Biology ,General Medicine ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,[SDV.GEN] Life Sciences [q-bio]/Genetics ,[SDV.BDD]Life Sciences [q-bio]/Development Biology ,[SDV.MHEP.UN]Life Sciences [q-bio]/Human health and pathology/Urology and Nephrology ,[SDV.BC] Life Sciences [q-bio]/Cellular Biology ,[SDV.MHEP.UN] Life Sciences [q-bio]/Human health and pathology/Urology and Nephrology ,General Biochemistry, Genetics and Molecular Biology - Abstract
Les récepteurs couplés aux protéines G sont, pour la plupart, rapidement internalisés après stimulation. L’étude de ce phénomène s’est particulièrement développée depuis une dizaine d’années et a engendré une masse impressionnante d’informations, apparemment discordantes, qui pourraient faire croire que chaque récepteur interprète sa propre partition biologique. Le but de cet article est de souligner, à partir de travaux récents, les convergences qui existent au niveau de la fonction et des mécanismes de lde ces récepteurs, tout en soulignant certaines variations phénotypiques réelles qui reflètent des nécessités biologiques., Most G protein-coupled receptors (GPCR) are rapidly internalized upon agonist stimulation. From the large number of studies available to date on receptor endocytosis, substantial differences seem to exist among GPCRs in terms of both the molecular pathways of receptor internalization and the biological significance of this process. The aim of this review is to outline common themes in GPCR endocytosis and to delineate true phenotypic variations, which reflect specific necessities for receptor’s function.
32. The C-terminus region of β-arrestin1 modulates VE-cadherin expression and endothelial cell permeability
- Author
-
Jagoda K. Hebda, Julie Gavard, Mark G.H. Scott, Sandy Azzi, Nicolas Bidère, Héloïse M. Leclair, Célestin Roussel, Institut Cochin (IC UM3 (UMR 8104 / U1016)), Centre National de la Recherche Scientifique (CNRS)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Mitologics, Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Robert Debré, Régulation de la survie cellulaire et des allogreffes, Université Paris-Sud - Paris 11 (UP11)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Paul Brousse-Institut National de la Santé et de la Recherche Médicale (INSERM), This research was funded by Ligue nationale contre le cancer comite de Paris, Fondation ARC, Fondation pour la Recherche Medicale, ANR JCJC and by a Marie Curie International Reintegration Grant within The Seventh Framework Programme. JKH is supported by doctoral fellowship from Universite Paris Descartes and SA by a post-doctoral fellowship from Fondation ARC., BMC, Ed., Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut Cochin (UM3 (UMR 8104 / U1016)), Université Paris Descartes - Paris 5 (UPD5) - Institut National de la Santé et de la Recherche Médicale (INSERM) - Centre National de la Recherche Scientifique (CNRS), Assistance publique - Hôpitaux de Paris (AP-HP) - Hôpital Robert Debré, and Université Paris-Sud - Paris 11 (UP11) - Assistance publique - Hôpitaux de Paris (AP-HP) - Hôpital Paul Brousse - Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
Short Report ,Biology ,Vascular endothelial growth inhibitor ,Biochemistry ,Permeability ,03 medical and health sciences ,chemistry.chemical_compound ,[SDV.BBM.BC] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biomolecules [q-bio.BM] ,0302 clinical medicine ,Vasculogenesis ,[SDV.BBM] Life Sciences [q-bio]/Biochemistry, Molecular Biology ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,[SDV.BBM.BC]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biochemistry [q-bio.BM] ,[SDV.BBM.BC] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biochemistry [q-bio.BM] ,Molecular Biology ,S1PR1 ,030304 developmental biology ,0303 health sciences ,β-arrestin ,VE-cadherin promoter ,Cell Biology ,VEGF ,Endocytosis ,Cell biology ,Vascular endothelial growth factor B ,Vascular endothelial growth factor ,Endothelial stem cell ,Vascular endothelial growth factor A ,chemistry ,Vascular endothelial growth factor C ,030220 oncology & carcinogenesis - Abstract
Background The endothelial specific cell-cell adhesion molecule, VE-cadherin, modulates barrier function and vascular homeostasis. In this context, we have previously characterized that VEGF (vascular endothelial growth factor) leads to VE-cadherin phosphorylation, β-arrestin2 recruitment and VE-cadherin internalization in mouse endothelial cells. However, exactly how this VE-cadherin/β-arrestin complex contributes to VEGF-mediated permeability in human endothelial cells remains unclear. In this study, we investigated in-depth the VE-cadherin/β-arrestin interactions in human endothelial cells exposed to VEGF. Findings First, we demonstrated that VEGF induces VE-cadherin internalization in a clathrin-dependent manner in human umbilical vein endothelial cells (HUVEC). In addition to the classical components of endocytic vesicles, β-arrestin1 was recruited and bound to phosphorylated VE-cadherin. Molecular mapping of this interaction uncovered that the C-terminus tail of β-arrestin1, that comprises amino acids 375 to 418, was sufficient to directly interact with the phosphorylated form of VE-cadherin. Interestingly, the expression of the C-terminus tail of β-arrestin1 induced loss of surface exposed-VE-cadherin, promoted monolayer disorganization and enhanced permeability. Finally, this effect relied on decreased VE-cadherin expression at the transcriptional level, through inhibition of its promoter activity. Conclusions Altogether, our results demonstrate that β-arrestin1 might play multiple functions collectively contributing to endothelial barrier properties. Indeed, in addition to a direct implication in VE-cadherin endocytosis, β-arrestin1 could also control VE-cadherin transcription and expression. Ultimately, understanding the molecular mechanisms involved in VE-cadherin function might provide therapeutic tools for many human diseases where the vascular barrier is compromised.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.