17 research outputs found

    The Human Papillomavirus Type 16 E6 Oncoprotein Activates mTORC1 Signaling and Increases Protein Synthesis ā–æ ā€ 

    No full text
    The mammalian target of rapamycin (mTOR) kinase acts as a cellular rheostat that integrates signals from a variety of cellular signal transduction pathways that sense growth factor and nutrient availability as well as intracellular energy status. It was previously reported that the human papillomavirus type 16 (HPV16) E6 oncoprotein may activate the S6 protein kinase (S6K) through binding and E6AP-mediated degradation of the mTOR inhibitor tuberous sclerosis complex 2 (TSC2) (Z. Lu, X. Hu, Y. Li, L. Zheng, Y. Zhou, H. Jiang, T. Ning, Z. Basang, C. Zhang, and Y. Ke, J. Biol. Chem. 279:35664-35670, 2004; L. Zheng, H. Ding, Z. Lu, Y. Li, Y. Pan, T. Ning, and Y. Ke, Genes Cells 13:285-294, 2008). Our results confirmed that HPV16 E6 expression causes an increase in mTORC1 activity through enhanced phosphorylation of mTOR and activation of downstream signaling pathways S6K and eukaryotic initiation factor binding protein 1 (4E-BP1). However, we did not detect a decrease in TSC2 levels in HPV16 E6-expressing cells. We discovered, however, that HPV16 E6 expression causes AKT activation through the upstream kinases PDK1 and mTORC2 under conditions of nutrient deprivation. We show that HPV16 E6 expression causes an increase in protein synthesis by enhancing translation initiation complex assembly at the 5ā€² mRNA cap and an increase in cap-dependent translation. The increase in cap-dependent translation likely results from HPV16 E6-induced AKT/mTORC1 activation, as the assembly of the translation initiation complex and cap-dependent translation are rapamycin sensitive. Lastly, coexpression of the HPV16 E6 and E7 oncoproteins does not affect HPV16 E6-induced activation of mTORC1 and cap-dependent translation. HPV16 E6-mediated activation of mTORC1 signaling and cap-dependent translation may be a mechanism to promote viral replication under conditions of limited nutrient supply in differentiated, HPV oncoprotein-expressing proliferating cells

    Internalization and degradation of activated receptor species are increased in HPV16 E6 expressing HFKs.

    No full text
    <p>(<b>A</b>) Western blot analysis of EGFR and INSRĪ²/IGFRĪ² following receptor internalization. After washing the cells with PBS they were incubated with biotin disulfide N-hydroxysulfosuccinimide ester for 40 minutes at 4Ā°C followed by inactivation of the biotin reagent with NH<sub>4</sub>Cl for 10 min at 4Ā°C. Cells were then stimulated with KSFM lacking growth factors and supplements for 3 hours at 37Ā°C. Following ligand independent stimulation, cells were either lysed without reduction to measure surface bound receptors (S) or reduced and lysed to measure ligand independent internalization (I), Internalization assays were performed as described in the <a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1003237#s4" target="_blank">Materials and Methods</a>. Levels of EGFR and INSRĪ²/IGFRĪ² in a 5 Āµg sample, representing 2% of the internalization assay, together with actin, are shown in the left panel (Input), and the resulting streptavidin affinity purification (AP) is shown in the right panel. Quantification representing the ratio of phosphorylated receptors to total receptors normalized to control vector (C) internalized phosphorylated receptors is shown below applicable panels. This experiment represents one of two independent experiments. (<b>B</b>) Immunofluorescence analysis of EGF co-staining with the endosomal marker EEA1 in HFKs with stable expression of HPV16 E6 (E6) or pLentiN6.3 control vector (C) following overnight starvation and stimulation with Alexa 647 labeled EGF (EGF) for 15 minutes. Cells were stained with Hoechst to visualize the nuclei. The scaling bar represents 40 Āµm. (<b>C</b>) Quantification of the number of EGF positive early endosomes from panel B. Bar graphs represent average numbers of EGF positive early endosomes per cell from four independent experiments. A total of 115 cells were evaluated. Error bars indicate standard deviations; the asterisk indicates statistical significance (<i>P</i>ā€Š=ā€Š0.0017).</p

    RPTK inhibition reduces mTORC1 activation in HPV16 E6 expressing HFKs.

    No full text
    <p>(<b>A</b>) Western blot analysis of EGFR and mTORC1 activation (S6K) in HFKs with stable expression of HPV16 E6 (E6) or pLentiN6.3 control vector (C) following EGFR inhibition with Gefitinib. Cells were treated with DMSO or 1 ĀµM Gefitinib over a 24 hour time course. Actin blots are shown as loading controls. The experiment shown here represents one of three independent experiments. (<b>B</b>) Western blot analysis of INSRĪ²/IGFRĪ² and mTORC1 activation (S6K) in HFKs with stable expression of HPV16 E6 (E6) or pLentiN6.3 control vector (C) following INSRĪ²/IGFRĪ² chemical inhibition with OSI-906. These data represent one of two independent experiments. Cells were treated with DMSO or 150 nM OSI-906 over a 6 hour time course. Actin blots are shown as a loading control; ā€œactin 1ā€ corresponds to the blots with the phosphospecific antibodies, ā€œactin 2ā€ corresponds the other blots.</p

    The signaling adaptor protein GRB2 is critical for HPV16 E6 mediated EGFR internalization and mTORC1 activation.

    No full text
    <p>(<b>A</b>) Immunofluorescence analysis of EGF co-staining with the endosomal marker EEA1 in GRB2 depleted (siGRB2) or control siRNA transfected (siCtrl) HFKs with stable expression of HPV16 E6 (E6) or pLentiN6.3 control vector (C) following overnight starvation and subsequent stimulation with Alexa 647 labeled EGF (EGF) for 15 minutes. Cells were stained with Hoechst to visualize the nuclei. The scaling bar represents 20 Āµm. (<b>B</b>) Quantification of the number of EGF positive early endosomes from panel A. Bar graphs represent average numbers of EGF positive early endosomes/cell with counts done in triplicate for a total of at least 95 analyzed cells per condition. Error bars indicate standard deviations; the asterisk indicates statistical significance (<i>P</i>ā€Š=ā€Š0.03). (<b>C</b>) Western blot analysis of S6K T389 in GRB2 depleted (siGRB2) or control siRNA transfected (siCtrl) HFK populations with stable expression of HPV16 E6 (E6) or control vector (C). Cells were transfected with the corresponding siRNAs at 48 hours prior to lysis. An actin blot is shown as a loading control and quantifications relative to actin are shown below each panel. A representative experiment taken from four independent experiments is shown in panel C.</p

    ERBB2 phosphorylation is increased in HPV16 E6 expressing HFKs.

    No full text
    <p>(<b>A</b>) Western blot analysis of proteins with phosphorylated tyrosine residues following EGF withdrawal (left) or PBS starvation (right) from HFK lysates stably expressing HPV16 E6 (E6) or the pLentiN6.3 control vector (C). (<b>B</b>) and (<b>C</b>) Western blot analyses probing the phosphorylation status of EGFR (<b>B</b>) or INSRĪ²/IGFRĪ² (<b>C</b>) using phosphospecific antibodies in HPV16 E6 (E6) or LXSN control vector (C) expressing stable HFKs. Representative experiments from three independent experiments are shown in panels A, B and C. Quantifications relative to actin are shown below each panel.</p

    Hypothetical model.

    No full text
    <p>HPV16 E6 expressing cells (right) show enhanced activation of RPTKs at least in part by enhancing internalization of activated receptor species as compared to parental cell (left). As a consequence, downstream signaling circuits, including mTORC1 activity remain active even under conditions of limited growth factor availability. See text for details.</p

    HPV16 E6 increases cellular migration in EGF depleted cells.

    No full text
    <p>(<b>A</b>) Wound healing assays with primary HFKs stably expressing HPV16 E6 (E6) or infected with pLentiN6.3 control vector (C) following wounding of the cellular monolayer in the absence of EGF following RPTK and effector pathway inhibition. Cells were treated with DMSO or 100 nM Rapamycin, 1 ĀµM Gefitinib, 150 nM OSI-906, or 10 ĀµM U0126 and closure of the monolayer was measured over a 25 hour time course. Only images of the DMSO treated samples are shown (<b>B</b>). Quantification of wound closure as shown in (<b>A</b>). Surface areas of wounds were calculated relative to the surface area of the wounds at tā€Š=ā€Š0 hour. Bar represent averages and standard deviations of three experiments; asterisks indicate statistical significance (<i>P</i><0.0001). (<b>C</b>) Quantification of wound closure in the presence of 8 Āµg/ml Mitomycin C to inhibit cellular proliferation. (<b>D</b>) Quantification of wound closure in immortalized HFKs stably expressing HPV16 E6 (E6) or pLentiN6.3 control vector (C) (<b>E</b>). Transwell migration assay with HFKs stably expressing HPV16 E6 (E6) or pLentiN6.3 control vector (C). Cells were seeded in the absence of EGF on the top chamber of a transwell insert and migration to the bottom chamber in the absence of EGF was determined at 30 minutes and 60 minutes after seeding. The bars represent averages and standard deviations of three experiments; the asterisk indicates statistical significance (<i>P</i><0.05).</p

    Signaling pathways downstream of RPTKs are activated in HPV16 E6 expressing HFKs.

    No full text
    <p>(<b>A</b>) Western blot analysis of MAPK signaling by analyzing ERK1/2 and RSK phosphorylation in HFKs with stable expression of HPV16 E6 (E6) or LSXN control vector (C) and experiencing nutrient withdrawal by PBS starvation for 30 minutes. Quantifications relative to actin are shown below each panel. A representative experiment from a total of four independent experiments is shown. (<b>B</b>) HPV16 E6 mediated increase in cap dependent translation is dependent on MEK and mTORC1 signaling. U2OS cells were transfected with pFR_CrPV_xb and the indicated plasmids and processed for firefly and Renilla luciferase activity at 48 hours post transfection. 20 hours prior to lysis, cells were treated with DMSO, 10 ĀµM U0126 (a MEK inhibitor), or a combination of 10 ĀµM U0126 and 100 nM Rapamycin (an mTORC1 inhibitor). Firefly and Renilla luciferase were normalized to levels in DMSO treated cells and are presented as fold changes of normalized firefly relative to normalized Renilla activity. The bar graph represents averages and standard deviations of three independent experiments, each performed in triplicate; the asterisk indicates statistical significance (<i>P</i><0.05).</p
    corecore