25 research outputs found

    Fibroblast activation protein is expressed by rheumatoid myofibroblast-like synoviocytes

    Get PDF
    Fibroblast activation protein (FAP), as described so far, is a type II cell surface serine protease expressed by fibroblastic cells in areas of active tissue remodelling such as tumour stroma or healing wounds. We investigated the expression of FAP by fibroblast-like synoviocytes (FLSs) and compared the synovial expression pattern in rheumatoid arthritis (RA) and osteoarthritis (OA) patients. Synovial tissue from diseased joints of 20 patients, 10 patients with refractory RA and 10 patients with end-stage OA, was collected during routine surgery. As a result, FLSs from intensively inflamed synovial tissues of refractory RA expressed FAP at high density. Moreover, FAP expression was co-localised with matrix metalloproteinases (MMP-1 and MMP-13) and CD44 splice variants v3 and v7/8 known to play a major role in the concert of extracellular matrix degradation. The pattern of signals appeared to constitute a characteristic feature of FLSs involved in rheumatoid arthritic joint-destructive processes. These FAP-expressing FLSs with a phenotype of smooth muscle actin-positive myofibroblasts were located in the lining layer of the synovium and differ distinctly from Thy-1-expressing and non-proliferating fibroblasts of the articular matrix. The intensity of FAP-specific staining in synovial tissue from patients with RA was found to be different when compared with end-stage OA. Because expression of FAP by RA FLSs has not been described before, the findings of this study highlight a novel element in cartilage and bone destruction of arthritic joints. Moreover, the specific expression pattern qualifies FAP as a therapeutic target for inhibiting the destructive potential of fibroblast-like synovial cells

    RP1 is a phosphorylation target of CK2 and is involved in cell adhesion

    Get PDF
    RP1 (synonym: MAPRE2, EB2) is a member of the microtubule binding EB1 protein family, which interacts with APC, a key regulatory molecule in the Wnt signalling pathway. While the other EB1 proteins are well characterized the cellular function and regulation of RP1 remain speculative to date. However, recently RP1 has been implicated in pancreatic cancerogenesis. CK2 is a pleiotropic kinase involved in adhesion, proliferation and anti-apoptosis. Overexpression of protein kinase CK2 is a hallmark of many cancers and supports the malignant phenotype of tumor cells. In this study we investigate the interaction of protein kinase CK2 with RP1 and demonstrate that CK2 phosphorylates RP1 at Ser(236) in vitro. Stable RP1 expression in cell lines leads to a significant cleavage and down-regulation of N-cadherin and impaired adhesion. Cells expressing a Phospho-mimicking point mutant RP1-ASP(236) show a marked decrease of adhesion to endothelial cells under shear stress. Inversely, we found that the cells under shear stress downregulate endogenous RP1, most likely to improve cellular adhesion. Accordingly, when RP1 expression is suppressed by shRNA, cells lacking RP1 display significantly increased cell adherence to surfaces. In summary, RP1 phosphorylation at Ser(236) by CK2 seems to play a significant role in cell adhesion and might initiate new insights in the CK2 and EB1 family protein association

    Serological immune response to cancer testis antigens in patients with pancreatic cancer

    Get PDF
    Serological screening approaches have allowed for the identification of a large number of potentially relevant tumor antigens in cancer patients. Within this group, cancer testis antigens represent promising targets for cancer immunotherapy, since they are widely expressed in a variety of human cancer entities. In pancreatic cancer, however, there are only few data available about the expression pattern and serological response to cancer testis antigens and other serological-defined tumor antigens. Therefore, we investigated the IgG antibody response against 11 cancer testis antigens (SCP-1, GAGE, LAGE-1a,-1b, CT-7, NY-ESO-1, SSX-1-5) recombinantly expressed on yeast surface (RAYS) in patients with pancreatic cancer (n = 96), chronic pancreatitis (n = 18) and healthy donors (n = 48). We found in 14% of all patients antibody responses to SCP-1, but not to other cancer testis antigens (GAGE, LAGE-1a,-1b, CT-7, NY-ESO-1, SSX-1-5). Antibody response correlated with the expression of SCP-1 in the primary tumor of the respective patient as shown by RT-PCR, immunohistochemistry and Western blot. In contrast, no serological response to cancer testis antigens was observed in healthy donors. The humoral immune response against SCP-1 was associated with the size of tumor, but not with other clinico-pathological parameters such as histology, stage, presence of lymph node metastases, grading, age, gender or gemcitabine treatment. In conclusion, antibody response to cancer testis antigen SCP-1 is found in a proportion of pancreatic carcinoma patients. These results indicate that identification of additional tumor antigens by serological screening of tumor cDNA expression libraries by RAYS is a promising goal in pancreatic cancer

    Treatment with 5-Aza-2'-Deoxycytidine Induces Expression of NY-ESO-1 and Facilitates Cytotoxic T Lymphocyte-Mediated Tumor Cell Killing

    No full text
    <div><p>Background</p><p>NY-ESO-1 belongs to the cancer/testis antigen (CTA) family and represents an attractive target for cancer immunotherapy. Its expression is induced in a variety of solid tumors via DNA demethylation of the promoter of CpG islands. However, NY-ESO-1 expression is usually very low or absent in some tumors such as breast cancer or multiple myeloma. Therefore, we established an optimized <i>in vitro</i> treatment protocol for up-regulation of NY-ESO-1 expression by tumor cells using the hypomethylating agent 5-aza-2'-deoxycytidine (DAC).</p><p>Methodology/Principal Findings</p><p>We demonstrated <i>de novo</i> induction of NY-ESO-1 in MCF7 breast cancer cells and significantly increased expression in U266 multiple myeloma cells. This effect was time- and dose-dependent with the highest expression of NY-ESO-1 mRNA achieved by the incubation of 10 μM DAC for 72 hours. NY-ESO-1 activation was also confirmed at the protein level as shown by Western blot, flow cytometry, and immunofluorescence staining. The detection and quantification of single NY-ESO-1 peptides presented at the tumor cell surface in the context of HLA-A*0201 molecules revealed an increase of 100% and 50% for MCF7 and U266 cells, respectively. Moreover, the enhanced expression of NY-ESO-1 derived peptides at the cell surface was accompanied by an increased specific lysis of MCF7 and U266 cells by HLA-A*0201/NY-ESO-1(<sub>157–165</sub>) peptide specific chimeric antigen receptor (CAR) CD8<sup>+</sup> T cells. In addition, the killing activity of CAR T cells correlated with the secretion of higher IFN-gamma levels.</p><p>Conclusions/Significance</p><p>These results indicate that NY-ESO-1 directed immunotherapy with specific CAR T cells might benefit from concomitant DAC treatment.</p></div

    Effects of DAC treatment on NY-ESO-1 mRNA and protein expression in MCF7, U266, and ARK cell lines.

    No full text
    <p>A. NY-ESO-1 specific mRNA, quantified as copy numbers/μg RNA using qRT-PCR. All data are representative of three independent experiments performed in triplicate. B. NY-ESO-1 protein expression analyzed by Western blotting (n = 3). The first and second line show total cell lysate of the respective cell line +/- DAC treatment and detection via a NY-ESO-1 specific or α-Tubulin specific (loading control) antibody. The third line shows a Western blot of recombinant NY-ESO-1 protein as control.</p

    Optimization of DAC treatment for MCF7, U266, and ARK tumor cell lines.

    No full text
    <p>A. Variation of DAC concentration in cell culture medium as indicated (0–15 μM); white bars: isotype control, grey bars: detection with HLA-A2/NY-ESO-1p<sub>157-165</sub> specific Fab-tetramers. B. Variation of DAC-treatment intensity (0–4 times per day). C. Variation of DAC-treatment duration (1–3 days). All data are representative of at least three independent experiments performed in triplicate.</p

    Increased specific lysis of MCF7 and U266 tumor cells by CAR redirected CD8<sup>+</sup> T cells after DAC treatment.

    No full text
    <p>Retrovirally transduced NY-ESO-1-specific and CEA-specific CAR redirected CD8<sup>+</sup> T cells were cocultivated with U266 or MCF7 cells. NY-ESO-1 expression after DAC treatment statistically significantly enhanced the antigen specific killing of anti-NY-ESO-1 CAR redirected T cells in U266 (A), whereas the increased lysis of MCF7 was only detectable after DAC treatment (B). Antigen specific activation of anti-NY-ESO-1 CAR redirected CD8<sup>+</sup> T cells was determined by IFN-gamma (C and D). All data are representative of three independent experiments performed in triplicate.</p

    Quantification of HLA-A2/NY-ESO1p<sub>157-165</sub> complexes at the cell surface of MCF7, U266, and ARK tumor cell lines.

    No full text
    <p>A. Flow cytometry histogram after staining of the indicated tumor cell lines with HLA-A2/NY-ESO-1p<sub>157-165</sub> peptide specific Fab-T1 tetramer (blue/red) and isotype control (black/green). Cells were either untreated (black/blue) or DAC- treated (green/red). B. Quantification of the NY-ESO-1 peptides at the surface of the untreated (white bars) or DAC-treated (grey bars) tumor cell lines. Peptide numbers were calculated as described in materials and methods. C. Relative change in HLA-A2/NY-ESO-1p<sub>157-165</sub> peptide presentation on the tumor cell lines. Increase of NY-ESO-1p<sub>157-165</sub> peptide presentation is shown in relation to the total number of HLA-A2 molecules on the tumor cell lines following DAC treatment. ARK cells were used as a negative control. All data are representative of at least five independent experiments performed in triplicate.</p
    corecore