22 research outputs found

    Systematic protein-protein interaction mapping for clinically relevant human GPCRs

    Get PDF
    G‐protein‐coupled receptors (GPCRs) are the largest family of integral membrane receptors with key roles in regulating signaling pathways targeted by therapeutics, but are difficult to study using existing proteomics technologies due to their complex biochemical features. To obtain a global view of GPCR‐mediated signaling and to identify novel components of their pathways, we used a modified membrane yeast two‐hybrid (MYTH) approach and identified interacting partners for 48 selected full‐length human ligand‐unoccupied GPCRs in their native membrane environment. The resulting GPCR interactome connects 686 proteins by 987 unique interactions, including 299 membrane proteins involved in a diverse range of cellular functions. To demonstrate the biological relevance of the GPCR interactome, we validated novel interactions of the GPR37, serotonin 5‐HT4d, and adenosine ADORA2A receptors. Our data represent the first large‐scale interactome mapping for human GPCRs and provide a valuable resource for the analysis of signaling pathways involving this druggable family of integral membrane proteins

    Human Herpesvirus 7 Open Reading Frames U12 and U51 Encode Functional β-Chemokine Receptors

    No full text
    Human herpesvirus 7 (HHV-7), which belongs to the betaherpesvirus subfamily and infects mainly CD4(+) T cells in vitro, infects children during infancy. HHV-7 contains two genes, U12 and U51, that encode putative homologs of cellular G-protein-coupled receptors. To analyze the biological function of the U12 and U51 genes, we cloned these genes and expressed the proteins in cells. U12 and U51 encoded functional calcium-mobilizing receptors for β-chemokines, which include thymus and activation-regulated chemokine (TARC), macrophage-derived chemokine (MDC), EBI1-ligand chemokine (ELC), and secondary lymphoid-tissue chemokine (SLC), but not for other chemokines, suggesting that the chemokine selectivities of the U12 and U51 products were distinct from those of the known mammalian chemokine receptors. ELC and SLC induced migration in Jurkat cells stably expressing U12, but TARC and MDC did not. In contrast, none of these chemokines induced migration in Jurkat cells stably expressing U51. Together, these data indicate that the products of U12 and U51 may play important and different roles in the pathogenesis of HHV-7 through transmembrane signaling

    Venetoclax efficacy on acute myeloid leukemia is enhanced by the combination with butyrate

    No full text
    Abstract Venetoclax has been approved recently for treatment of Acute myeloid leukemia (AML). Venetoclax is a BH3-mimetic and induces apoptosis via Bcl-2 inhibition. However, venetoclax’s effect is still restrictive and a novel strategy is needed. In the present study, we demonstrate that sodium butyrate (NaB) facilitates the venetoclax’s efficacy of cell death in AML cells. As a single agent, NaB or venetoclax exerted just a weak effect on cell death induction for AML cell line KG-1. The combination with NaB and venetoclax drastically induced cell death. NaB upregulated pro-apoptotic factors, Bax and Bak, indicating the synergistic effect by the collaboration with Bcl-2 inhibition by venetoclax. The combined treatment with NaB and venetoclax strongly cleaved a caspase substrate poly (ADP-ribose) polymerase (PARP) and a potent pan-caspase inhibitor Q-VD-OPh almost completely blocked the cell death induced by the combination, meaning that the combination mainly induced apoptosis. The combination with NaB and venetoclax also strongly induced cell death in another AML cell line SKNO-1 but did not affect chronic myeloid leukemia (CML) cell line K562, indicating that the effect was specific for AML cells. Our results provide a novel strategy to strengthen the effect of venetoclax for AML treatment

    Oligomérisation des protéines humaines et virales à sept domaines transmembranaires

    No full text
    Les récepteurs couplés aux protéines G (RCPG), aussi appelés protéines à sept domaines transmembranaires (7TM), représentent la plus grande famille de protéines. Elle comprend, chez l’homme, environ 900 membres. Ces protéines se lient à une grande variété de ligands ce qui entraîne l’activation de voies de signalisation impliquées dans divers processus cellulaires. Certaines protéines à 7TM, communément appelés orphelines, n’ont pas de ligand identifié, mais semblent jouer un rôle important dans la modulation de la fonction cellulaire via leurs activités constitutives ou leurs interactions avec d’autres protéines à 7TM. Certains virus synthétisent des protéines orphelines à 7TM homologues aux récepteurs de chimiokines humains pour détourner les fonctions de la cellule hôte et promouvoir leur réplication et leur dissémination. En effet, les protéines virales à 7TM sont capables de former des homomères ou des hétéromères avec d’autres protéines virales à 7TM, voire avec des protéines à 7TM de la cellule hôte. L’hétéromérisation des protéines virales à 7TM constitue une stratégie pertinente pour contrôler les fonctions de la cellule hôte

    Human Herpesvirus-6 U14 Induces Cell-Cycle Arrest in G2/M Phase by Associating with a Cellular Protein, EDD.

    Get PDF
    The human herpesvirus-6 (HHV-6) infection induces cell-cycle arrest. In this study, we found that the HHV-6-encoded U14 protein induced cell-cycle arrest at G2/M phase via an association with the cellular protein EDD, a mediator of DNA-damage signal transduction. In the early phase of HHV-6 infection, U14 colocalized with EDD dots in the nucleus, and similar colocalization was also observed in cells transfected with a U14 expression vector. When the carboxyl-terminal region of U14 was deleted, no association of U14 and EDD was observed, and the percentage of cells in G2/M decreased relative to that in cells expressing wild-type U14, indicating that the C-terminal region of U14 and the U14-EDD association are critical for the cell-cycle arrest induced by U14. These results indicate that U14 is a G2/M checkpoint regulator encoded by HHV-6

    Subcellular localization of U14 and EDD in U14-expressing 293T cells.

    No full text
    <p>293T cells were transfected with HHV-6A U14, its deletion mutants, or empty vector (as a control). Cells were harvested at 12 h post-transfection, fixed, and subjected to IFA using antibodies against HA (for U14) and EDD; nuclear DNA was counterstained with Hoechst 33342. Scale bars: 5 μm. Single sections are shown.</p
    corecore