15 research outputs found

    Reciprocal complementation of the tumoricidal effects of radiation and natural killer cells.

    Get PDF
    The tumor microenvironment is a key determinant for radio-responsiveness. Immune cells play an important role in shaping tumor microenvironments; however, there is limited understanding of how natural killer (NK) cells can enhance radiation effects. This study aimed to assess the mechanism of reciprocal complementation of radiation and NK cells on tumor killing. Various tumor cell lines were co-cultured with human primary NK cells or NK cell line (NK-92) for short periods and then exposed to irradiation. Cell proliferation, apoptosis and transwell assays were performed to assess apoptotic efficacy and cell viability. Western blot analysis and immunoprecipitation methods were used to determine XIAP (X-linked inhibitor of apoptosis protein) and Smac (second mitochondria-derived activator of caspase) expression and interaction in tumor cells. Co-culture did not induce apoptosis in tumor cells, but a time- and dose-dependent enhancing effect was found when co-cultured cells were irradiated. A key role for caspase activation via perforin/granzyme B (Grz B) after cell-cell contact was determined, as the primary radiation enhancing effect. The efficacy of NK cell killing was attenuated by upregulation of XIAP to bind caspase-3 in tumor cells to escape apoptosis. Knockdown of XIAP effectively potentiated NK cell-mediated apoptosis. Radiation induced Smac released from mitochondria and neutralized XIAP and therefore increased the NK killing. Our findings suggest NK cells in tumor microenvironment have direct radiosensitization effect through Grz B injection while radiation enhances NK cytotoxicity through triggering Smac release

    pNK and NK-92 cells sensitized tumor cells.

    No full text
    <p>1×10<sup>5</sup> of various tumor cells were seeded in 96-well tissue-culture plates, co-cultured with 2.5×10<sup>5</sup> pNK cells for 4 h, washed and then exposed to 800 cGy of irradiation and evaluated 48 h late for cell proliferation by the MTS (A). C, cancer cell alone; C/NK, cancer cell and NK coculture; C+RT, cancer cell treated with 800 cGy radiation; C/NK+RT, cancer cell and pNK coculture followed by radiation. The apoptosis of CNE-1 cells after irradiation 48 h under various co-culture conditions that described previously was analyzed by (B) Annexin-V assay and (C) cell cycle analysis. (D) CNE-1 cells were incubated with NK-92 cells in 1∶2.5 ratios for 2, 4, and 8 h and irradiated at indicated doses. (E) 1×10<sup>5</sup> CNE-1 cells were cultured in the lower chambers of transwells, and 2.5×10<sup>5</sup> NK-92 cells were cultured in the upper chambers for 4 and 8 h. Both of (D) and (E) were assayed using Annexin-V to detect apoptotic cells (AnnexinV<sup>+</sup>). (*, <i>p</i><0.05).</p

    The caspase signaling pathway was induced after co-culture.

    No full text
    <p>CNE-1 cells were co-cultured with 2.5 fold of NK-92 cells for 4 h, then NK-92 cells were washed away, and CNE-1 cells were exposed to 800 cGy of radiation. Control cells of CNE-1 alone or CNE-1 cells that had been co-cultured were not irradiated. After 24 h of incubation, cells were harvested for western blot analysis of procaspase/caspase-3, procaspase-8, and procaspase-9 protein in lysates of CNE-1 alone (lane C), CNE-1 cells cultures with NK-92 cells (lane C/N). The arrows indicate cleaved (activated) caspase 3 at about 17 kDa and its precursor, pro-caspase 3, at about 43 kDa; precaspase 8, at about 55kDa; procaspase 9, at about 45kDa. β-actin was used as the internal control.</p

    Primary NK cells sensitized tumor cells with same pathway.

    No full text
    <p>CNE-1 cells were transfected with 80 nM of XIAP siRNA for 16 h and co-cultured with pNK cells for 4 h before pNK cells were washed away. The cells were assayed using Annexin-V to determine the percentage of apoptotic cells.</p

    Caspase-3 was inhibited by XIAP and XIAP was downregulated by binding of Smac after radiation.

    No full text
    <p>CNE-1 cells (lane C) were treated with NK-92 cells for 4 h (lane C/N) before combined treatment with 800 cGy of radiation. (A) Cell lysates were immunoprecipitated with anti-XIAP antibody and immunoblotted with anti-Smac, anti-caspase-3 or anti-XIAP antibody. (B) CNE-1 cells were transfected with 80 nM of XIAP siRNA for 16 h and co-cultured with NK-92 cells for 4 h before NK-92 cells were washed away. The cells were assayed using Annexin-V to determine the percentage of apoptotic cells (AnnexinV<sup>+</sup>). (C) Cell lysates were immunoprecipitated by anti-Smac antibody and detected with anti-XIAP antibody by Western blot. (D) CNE-1 cells were treated with NK-92 cells for 4 h (C/N) before combined treatment with 800 cGy of radiation (C/N+RT) or CNE-1 treated with 800 cGy of radiation alone (C+RT). After treatment, cells were further incubated for 0 min, 15 min, 2 h, or 24 h, then harvested and fractionated into cytosolic (Cyto) and mitochondrial (Mito) fractions for assay by western blot. β-actin was used as the loading control for each fraction. Density of the XIAP normalized with β-actin was assayed by Image J. The bar chart was average of three independent experiments.</p

    Pharmacologically Upregulated Carcinoembryonic Antigen-expression Enhances the Cytolytic Activity of Genetically-modified Chimeric Antigen Receptor NK-92MI Against Colorectal Cancer Cells

    No full text
    [[abstract]]Background: The natural killer cell line, NK-92MI, is cytotoxic against various types of cancer. The aim of this study was to develop chimeric antigen receptor-modified (CAR) NK-92MI cells targeting carcinoembryonic antigenexpressing (CEA) tumours and increase killing efficacy by pharmacologically modifying CEA-expression. Result: We generated anti-CEA-CAR NK-92MI cells by retroviral vector transduction. This genetically-modified cell line recognised and lysed high CEA-expressing tumour cell lines (LS174T) at 47.54 ± 12.60% and moderate CEA-expressing tumour cell lines (WiDr) at 31.14 ± 16.92% at a 5:1 effector: target (E/T) ratio. The cell line did not lyse low CEA-expressing tumour cells (HCT116) as they did their parental cells (NK-92MI cells). The histone deacetylase-inhibitor (HDAC) sodium butyrate (NaB) and the methylation-inhibitor 5-azacytidine (5-AZA), as epigenetic modifiers, induced CEA-expression in HCT116 and WiDr cells. Although the IC50 of 5 fluorouracil (5-FU) increased, both cell lines showed collateral sensitivity to anti-CEA-CAR NK-92MI cells. The cytolytic function of anti-CEA-CAR NK-92MI cells was increased from 22.99 ± 2.04% of lysis background to 69.20 ± 11.92% after NaB treatment, and 69.70 ± 9.93% after 5-AZA treatment, at a 10:1 E/T ratio in HCT116 cells. The WiDr cells showed similar trend, from 22.99 ± 4.01% of lysis background to 70.69 ± 10.19% after NaB treatment, and 59.44 ± 10.92% after 5-AZA treatment, at a 10:1 E/T ratio. Conclusions: This data indicates that the effector-ability of anti-CEA-CAR NK-92MI increased in a CEA-dependent manner. The combination of epigenetic-modifiers like HDAC-inhibitors, methylation-inhibitors, and adoptive-transfer of ex vivo-expanded allogeneic-NK cells may be clinically applicable to patients with in 5-FU resistant condition.[[notice]]補正完

    Effect of NK-92-treated CNE-1 cells on Fas blockage.

    No full text
    <p>CNE-1 cells were co-cultured with 2.5 fold NK-92 cells for 4 h in presence of anti-FasL blocking antibody (10 µg/ml). The percentage of apoptotic cells after irradiation 48 h under various co-culture conditions was analyzed by Annexin-V assay (AnnexinV<sup>+</sup>, D). Results from 3 independent experiments are shown; bars indicate mean ± SD.</p

    Mechanism of reciprocal interaction between NK cells and radiation in target cells.

    No full text
    <p>NK cells damage target cell through perforin/granzyme B and death receptor/caspase mediated pathway. The radiosensitisation effect through NK cell depends more on the perforin/granzyme B pathway. Without radiation, the suboptimal activation of NK cells cause up-regulation of XIAP. With radiation, the mitochondria releases Smac to neutralize XIAP and enhances NK cell-mediated cytotoxicity.</p
    corecore