9 research outputs found

    Mechanism of Calcium Lactate Facilitating Phytic Acid Degradation in Soybean during Germination

    No full text
    Calcium lactate facilitates the growth and phytic acid degradation of soybean sprouts, but the mechanism is unclear. In this study, calcium lactate (Ca) and calcium lactate with lanthanum chloride (Ca+La) were used to treat soybean sprouts to reveal the relevant mechanism. Results showed that the phytic acid content decreased and the availability of phosphorus increased under Ca treatment. This must be due to the enhancement of enzyme activity related to phytic acid degradation. In addition, the energy metabolism was accelerated by Ca treatment. The energy status and energy metabolism-associated enzyme activity also increased. However, the transmembrane transport of calcium was inhibited by La<sup>3+</sup> and concentrated in intercellular space or between the cell wall and cell membrane; thus, Ca+La treatment showed reverse results compared with those of Ca treatment. Interestingly, gene expression did not vary in accordance with their enzyme activity. These results demonstrated that calcium lactate increased the rate of phytic acid degradation by enhancing growth, phosphorus metabolism, and energy metabolism

    Dendritic Cells Pulsed with Leukemia Cell-Derived Exosomes More Efficiently Induce Antileukemic Immunities

    No full text
    <div><p>Dendritic cells (DCs) and tumor cell-derived exosomes have been used to develop antitumor vaccines. However, the biological properties and antileukemic effects of leukemia cell-derived exosomes (LEXs) are not well described. In this study, the biological properties and induction of antileukemic immunity of LEXs were investigated using transmission electron microscopy, western blot analysis, cytotoxicity assays, and animal studies. Similar to other tumor cells, leukemia cells release exosomes. Exosomes derived from K562 leukemia cells (LEX<sub>K562</sub>) are membrane-bound vesicles with diameters of approximately 50–100 μm and harbor adhesion molecules (<i>e.g.</i>, intercellular adhesion molecule-1) and immunologically associated molecules (<i>e.g.</i>, heat shock protein 70). In cytotoxicity assays and animal studies, LEXs-pulsed DCs induced an antileukemic cytotoxic T-lymphocyte immune response and antileukemic immunity more effectively than did LEXs and non-pulsed DCs (<i>P</i><0.05). Therefore, LEXs may harbor antigens and immunological molecules associated with leukemia cells. As such, LEX-based vaccines may be a promising strategy for prolonging disease-free survival in patients with leukemia after chemotherapy or hematopoietic stem cell transplantation.</p></div

    Therapeutic effect of LEX<sub>L1210</sub>-pulsed DCs on established tumors.

    No full text
    <p>To examine the therapeutic effect on established tumors, DBA/2 mice (n = 8 per group) were subcutaneously (s.c.) inoculated with L1210 cells (0.5×10<sup>6</sup> cells/mouse). After 5 d, when tumors became palpable (∼5 mm in diameter), mice were s.c. immunized with L1210-derived exosomes (LEX<sub>L1210</sub>) and different doses of dendritic cells (DCs) pulsed with LEX<sub>L1210</sub> (DC/LEX<sub>L1210</sub>) (1.0–4.0×10<sup>6</sup> cells/mouse). Animal mortality and tumor growth or regression were monitored daily for up to 10 weeks. For ethical treatment of the animals, mice were euthanized when the tumor diameter reached 1.5 cm. Experiments were performed in triplicate. One representative experiment is shown.</p

    LEX<sub>L1210</sub> and LEX<sub>L1210</sub>-pulsed DCs induce anti-leukemia protective immunity against L1210 leukemia cells.

    No full text
    <p>Survival of mice prophylactically immunized with different vaccines. DBA/2 mice (n = 8 per group) were immunized with phosphate-buffered saline (PBS), L1210-derived exosomes (LEX<sub>L1210</sub>), non-pulsed dendritic cells (DCs), and different doses of LEX<sub>L1210</sub>-pulsed DCs (DC/LEX<sub>L1210</sub>). On days 7–10 after immunization, all mice were challenged with L1210 leukemia cells. Vaccination with non-pulsed DCs (median survival time [MST], 20 d) did not significantly prolong the survival of mice as compared to the non-vaccinated PBS group (MST, 15 d), whereas LEX<sub>L1210</sub> vaccination (MST, 30 d) improved survival as compared to the control group of non-vaccinated mice (<i>P</i><0.05). In contrast, mice receiving different doses of DC/LEX<sub>L1210</sub> (particularly 2×10<sup>6</sup> and 4×10<sup>6</sup>) had significantly improved survival as compared to non-vaccinated mice (<i>P</i><0.0001). Results were combined from 2 separate experiments.</p

    Vaccination with LEXO and LEXO-targeted DC protects against tumor growth.

    No full text
    <p>To examine the antitumor immunity conferred by LEX<sub>L1210</sub> and DC/LEX<sub>L1210</sub>, DBA/2 mice were randomly divided into 4 groups (n = 8) and immunized s.c. with the following vaccines on the inner side of their thighs: PBS (control), LEX<sub>L1210</sub> (30 μg), unpulsed DCs (1×10<sup>6</sup> cells), and LEX<sub>L1210</sub>-pulsed DCs (DC/LEX<sub>L1210</sub>) (1×10<sup>6</sup> cells). On day 7 after immunization, all mice were challenged with L1210 leukemia cells on the outer side of the same thighs (0.5×10<sup>6</sup> cells/mice). To determine immune specificity, a group of tumor-free mice after immunization, were challenged s.c. with p388 cells (5×10<sup>5</sup> cells/mouse). Tumor growth was monitored daily for up to 4 weeks using a caliper. For ethical treatment of the animals, all mice were euthanized when the tumor diameter reached 1.5 cm. Three total experiments were performed. One representative experiment is shown.</p

    Morophology and expression of heat shock protein 70 and ABL in LEX<sub>K562</sub>.

    No full text
    <p>(a) Transmission electron micrograph of K562 cell-secreted exosomes (×100K). (b and c) Electron micrograph of heat shock protein 70 (HSP70)- and ABL-labeled exosomes. (d) Western blot analysis demonstrating the presence of HSP70 and ABL molecules in K562 cells and K562-derived exosomes (LEX<sub>K562</sub>).</p

    Cytotoxicity assay.

    No full text
    <p>Cytotoxic responses were evaluated by the lactate dehydrogenase (LDH)-releasing method. Splenic T-cells from mice immunized intravenously with L1210-derived exosomes (LEX<sub>L1210</sub>) and dendritic cells (DCs) pulsed with LEX<sub>L1210</sub> or phosphate-buffered saline (PBS) as a control were harvested and co-cultured with irradiated L1210 cells. At the end of the culture period, viable T-cells were separated using Ficoll-Paque centrifugation and were thereafter referred to as effector cells. The LDH assay is an enzymatic method used to colorimetrically quantify LDH released from lysed target cells, including L1210 or P388 cells, which served as the control and were mixed at different ratios with effector cells after incubation for 4 h at 37°C. The spontaneous/maximal release ratio was <20% in all experiments. Specific lysis (%) was calculated as follows: (experimental LDH release – effector cell spontaneous LDH release – target spontaneous LDH release)/(target maximum LDH release)×100. * <i>P</i><0.05 compared with the PBS, DC, and P388 groups; ** <i>P</i><0.01 compared with the PBS control group; △ <i>P</i><0.05 compared with the LEX<sub>L1210</sub> group; △△ <i>P</i><0.01 compared with the LEX<sub>L1210</sub> group. Experiments were performed in triplicate. One representative experiment is shown.</p

    Exosome-uptaking by dendritic cells (a) Carboxyfluorescein succinimidyl ester (CFSE)-labeled exosomes were co-cultured <i>in vitro</i> with dendritic cells (DCs), and CFSE-positive DCs were detected using flow cytometry at different times during the culture.

    No full text
    <p>Confocal microscopy was used concurrently with flow cytometry to visualize the cultured DCs. (b) Phase changes of CFSE expression in DCs at different time points during the culture. (c) To investigate the rate of decay of exosomes (EXOs) in DCs, DCs were incubated with CFSE-labeled EXOs for 4 h, washed twice with phosphate-buffered saline, cultured in culture medium, and examined at different time points for up to 72 h.</p

    Table2_Anti-hyperuricemia effect of hesperetin is mediated by inhibiting the activity of xanthine oxidase and promoting excretion of uric acid.DOCX

    No full text
    Hesperetin is a natural flavonoid with many biological activities. In view of hyperuricemia treatment, the effects of hesperetin in vivo and in vitro, and the underlying mechanisms, were explored. Hyperuricemia models induced by yeast extract (YE) or potassium oxonate (PO) in mice were created, as were models based on hypoxanthine and xanthine oxidase (XOD) in L-O2 cells and sodium urate in HEK293T cells. Serum level of uric acid (UA), creatinine (CRE), and urea nitrogen (BUN) were reduced significantly after hesperetin treatment in vivo. Hesperetin provided hepatoprotective effects and inhibited xanthine oxidase activity markedly, altered the level of malondialdehyde (MDA), glutathione peroxidase (GSH-PX) and catalase (CAT), downregulated the XOD protein expression, toll-like receptor (TLR)4, nucleotide binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, interleukin-18 (IL-18), upregulated forkhead box O3a (FOXO3a), manganese superoxide dismutase (MnSOD) in a uric acid-synthesis model in mice. Protein expression of organic anion transporter 1 (OAT1), OAT3, organic cationic transporter 1 (OCT1), and OCT2 was upregulated by hesperetin intervention in a uric acid excretion model in mice. Our results proposal that hesperetin exerts a uric acid-lowering effect through inhibiting xanthine oxidase activity and protein expression, intervening in the TLR4-NLRP3 inflammasome signaling pathway, and up-regulating expression of FOXO3a, MnSOD, OAT1, OAT3, OCT1, and OCT2 proteins. Thus, hesperetin could be a promising therapeutic agent against hyperuricemia.</p
    corecore