47 research outputs found

    Receptors involved in the crosstalk between CXCL12 and HER1.

    No full text
    <p>5637 or HeLa cells were cultured alone, or in the presence of 200 ng/mL CXCL12 or 25 ng/mL HB-EGF. After 20 minutes, the cells were collected and evaluated by flow cytometry for the expression of CXCR4 and HER1. (A) Stimulation with CXCL12 led to downregulation of CXCR4 due to its internalization. (B) Stimulation with HB-EGF induced HER1 internalization. In contrast, no changes in the surface expression of HER1 were detected after stimulation with CXCL12. Thus, CXCL12 binding to CXCR4 did not transactivate HER1 in these cells <i>via</i> shedding of HER1 ligands. Representative flow cytometry patterns and the means ±SD of 10 experiments are depicted.</p

    Regulation of HER1 Y1068 or Y1173 phosphorylation downstream of G-proteins requires calmodulin/calcineurin activity.

    No full text
    <p>(A) and (B) Time-course. HeLa cells were stimulated as indicated in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0034432#pone-0034432-g003" target="_blank">Figure 3</a>. Treatment with W-13 (a calmodulin inhibitor) or CsA (a calcineurin inhibitor) restored HB-EGF-dependent HER1 phosphorylation at Y1068, whereas KN-62 (a CaMKII inhibitor) had no effect. (C) Following prestimulation with [N33A]CXCL12 the HB-EGF-dependent HER1 phosphorylation at Y1068 and Y1173 was abolished at the plateau (15 minutes) and restored by W-13 or CsA; KN-62 had no effect. The means ±SD of 10 experiments are depicted.</p

    Knockdown of β-arrestin 2 protein levels allows for transinhibition of HER1.

    No full text
    <p>5637 or HeLa cells were transfected with control siRNA or β-arrestin 2 siRNA, prestimulated for 1 minute with 200 ng/mL CXCL12 and subsequently stimulated with 25 ng/mL HB-EGF for 2, 4, 5, 7, 10 or 15 minutes. In knockdown cells either Y1068 or Y1173 phosphorylation was inhibited as opposed to non-silenced cells at each time observed, as determined by ELISA (p<0.05). This shows that CXCL12 signaling transinhibits HER1 phosphorylation via G-protein-pathways in the absence of β-arrestin 2 activation and further supports that [N33A]CXCL12, which strongly transhinibits HER1, is a G-protein-biased ligand. The means ±SD out of 4 experiments are shown.</p

    Phosphorylation of HER1 and ERK1/2, and cell proliferation following treatment with HB-EGF and CXCL12.

    No full text
    <p>(A) 5637 and HeLa cells were stimulated with 25 ng/mL HB-EGF for 20 minutes, and cell lysates were analyzed by mass spectrometry following trypsin digestion. Y1068 and Y1173, sites of autophosphorylation coupled to the activation of Ras, MEK and ERK1/2, were evaluated in the following experiments with specific phosphotyrosine mAbs. (B) HB-EGF induced phosphorylation of both HER1 Y1068 and ERK1/2 TY185/187 in 5637 or HeLa cells. In contrast, stimulation with 200 ng/mL CXCL12 for 20 minutes, which induced phosphoERK1/2, led to no phosphorylation of HER1. (C) In dose-response experiments, exposures of CXCL12 ranging from 6 to 400 ng/mL for 20 minutes did not induce HER1 phosphorylation in either cells. (D) No proliferation was induced with 200 ng/mL CXCL12. The means ±SD of 10 experiments are depicted.</p

    CXCL12 regulates HER1 Y1068 and Y1173 phosphorylation via G-proteins.

    No full text
    <p>5637 or HeLa cells were stimulated with 25 ng/mL HB-EGF or with 200 ng/mL CXCL12 or N33A]CXCL12 alone or followed after 1 minute by 25 ng/mL HB-EGF. HER1 Y1068 or ERK1/2 TY185/187 phosphorylation was evaluated at the indicated time-points by using specific mAbs and was expressed as percentages of phosphorylation at the specified times after normalization as phosphorylated molecule/total molecule ratios. (A) HER1 phosphorylation at Y1068 in HeLa cells induced by HB-EGF alone (black) was abolished by prestimulation with [N33A]CXCL12 (blue) and modified by prestimulation with CXCL12 (red): maximum phosphorylation was reached at 4 minutes, and the plateau after the initial spike was around 50% of the maximum at 10 minutes. No phosphorylation was induced by CXCL12 alone. (B) Phosphorylation at Y1173 in 5637 cells displayed the same kind of pattern. (C) Prestimulation with CXCL12 did not modify the mitogenic effect of HB-EGF. (D) Stimulation with CXCL12 induced ERK1/2 phosphorylation (red) resulting from two spikes: a G-protein-dependent (blue) and a β-arrestin-dependent (red) phosphorylation spike. By using the PKC inhibitor Ro-31 the G-protein-dependent spike was abolished, whereas the β-arrestin-dependent spike persisted. [N33A]CXCL12 induced only the G-protein-dependent spike (blue), which was abolished by Ro-31. (E) Ro-31 abolished the effects of prestimulation with CXCL12 or [N33A]CXCL12 at Y1068 in 5637 cells. (F) The same pattern at Y1173 in HeLa cells. The means ±SD of 10 experiments are depicted.</p

    Shotgun Protein Profile of Human Adipose Tissue and Its Changes in Relation to Systemic Amyloidoses

    No full text
    In systemic amyloidosis, accumulation of misfolded proteins as extracellular amyloid fibrils in tissues causes severe organ dysfunction, but the molecular events of tissue damage related to amyloid deposition are still largely unknown. Through the use of the MudPIT proteomic approach, comprehensive protein profiles of human amyloid-affected adipose tissue from patients and its control (non-amyloid-affected) counterpart were acquired. Label-free comparison between patients and controls made it possible to highlight differences related to the presence of amyloid, by describing up- and down-represented proteins, connected into interacting networks. In particular, extracellular matrix (ECM), protein folding, lipid metabolism, and mitochondrial functions were among the most affected structural/functional pathways. The reported results, obtained with no a priori hypotheses, represent a significant step forward in the clarification of the molecular mechanisms involved in amyloidoses at tissue level and are the premise for understanding protein misfolding diseases

    CXCL12 and [N33A]CXCL12 transinhibit HER1 in 5637 or HeLa cells.

    No full text
    <p>Both chemokines signal <i>via</i> G-proteins to calmodulin/calcineurin and modulate the ligand-dependent phosphorylation of HER1. CXCL12 induces a delay in the phosphorylation. [N33A]CXCL12, which is a G-protein-biased ligand, inhibits the HER1 phosphorylation.</p

    Dissecting <i>Escherichia coli</i> Outer Membrane Biogenesis Using Differential Proteomics

    No full text
    <div><p>The cell envelope of Gram-negative bacteria is a complex multi-layered structure comprising an inner cytoplasmic membrane and an additional asymmetric lipid bilayer, the outer membrane, which functions as a selective permeability barrier and is essential for viability. Lipopolysaccharide, an essential glycolipid located in the outer leaflet of the outer membrane, greatly contributes to the peculiar properties exhibited by the outer membrane. This complex molecule is transported to the cell surface by a molecular machine composed of seven essential proteins LptABCDEFG that form a transenvelope complex and function as a single device. While advances in understanding the mechanisms that govern the biogenesis of the cell envelope have been recently made, only few studies are available on how bacterial cells respond to severe envelope biogenesis defects on a global scale. Here we report the use of differential proteomics based on Multidimensional Protein Identification Technology (MudPIT) to investigate how <i>Escherichia coli</i> cells respond to a block of lipopolysaccharide transport to the outer membrane. We analysed the envelope proteome of a <i>lptC</i> conditional mutant grown under permissive and non permissive conditions and identified 123 proteins whose level is modulated upon LptC depletion. Most such proteins belong to pathways implicated in cell envelope biogenesis, peptidoglycan remodelling, cell division and protein folding. Overall these data contribute to our understanding on how <i>E. coli</i> cells respond to LPS transport defects to restore outer membrane functionality.</p></div

    Proteome Profiling of Neuroblastoma-Derived Exosomes Reveal the Expression of Proteins Potentially Involved in Tumor Progression

    No full text
    <div><p>Neuroblastoma (NB) is the most common extracranial solid tumor in childhood, with grim prognosis in a half of patients. Exosomes are nanometer-sized membrane vesicles derived from the multivesicular bodies (MVBs) of the endocytic pathway and released by normal and neoplastic cells. Tumor-derived exosomes have been shown in different model systems to carry molecules that promote cancer growth and dissemination. In this respect, we have here performed the first characterization and proteomic analysis of exosomes isolated from human NB cell lines by filtration and ultracentrifugation. Electron microscopy demonstrated that NB-derived exosomes exhibited the characteristic cup-shaped morphology. Dynamic light scattering studies showed a bell-shaped curve and a polydispersity factor consistent with those of exosomes. Zeta potential values suggested a good nanoparticle stability. We performed proteomic analysis of NB-derived exosomes by two dimension liquid chromatography separation and mass spectrometry analyses using the multidimensional protein identification technology strategy. We found that the large majority of the proteins identified in NB derived exosomes are present in Exocarta database including tetraspanins, fibronectin, heat shock proteins, MVB proteins, cytoskeleton-related proteins, prominin-1 (CD133), basigin (CD147) and B7-H3 (CD276). Expression of the CD9, CD63 and CD81 tetraspanins, fibronectin, CD133, CD147 and CD276 was validated by flow cytometry. Noteworthy, flow cytometric analysis showed that NB-derived exosomes expressed the GD2 disialoganglioside, the most specific marker of NB. In conclusion, this study shows that NB-derived exosomes express a discrete set of molecules involved in defense response, cell differentiation, cell proliferation and regulation of other important biological process. Thus, NB-derived exosomes may play an important role in the modulation of tumor microenvironment and represent potential tumor biomarkers.</p></div
    corecore