30 research outputs found

    Roles of bulk γ(L)-Bi₂MoO₆ and surface β-Bi₂Mo₂O₉ in the selective catalytic oxidation of C₃H₆

    Get PDF
    γ(L)-Bi₂MoO₆ (L: low temperature phase) catalysts, whose surface compositions have a Mo/Bi ratio above = 0.5, exhibited high selectivity in the partial oxidation of C₃H₆, while catalysts with Mo/Bi surface ratios near or below = 0.5 exhibited low selectivity. γ(L)-phase catalysts which have Mo/Bi surface ratios greater than = 0.5, were demonstrated to form β-Bi₂Mo₂O₉ on their surface. An interaction between the β- and γ(L)-phases was observed in these catalysts’ UV–vis spectra at 430 nm. The new β-phase material seems to grow along b-axis of γ(L)-phase, i.e., perpendicular to MoO₂–Bi₂O₂ layers. Structure visualizations revealed that the α-Bi₂Mo₃O₁₂, β-, and γ(H)-phases, which are selective catalysts, contain twin Mo tetrahedral structures, and that their Mo and Bi ions lie on the same plane. The pure γ(L)-phase does not contain this structure. A model for the very rapid transfer of oxygen between the γ(L)- and β-phases is discussed in relation to the kinetics of C₃H₆ oxidation.ArticleJournal of molecular catalysis. A, Chemical. 318(1-2):94-100 (2010)journal articl

    Robust and highly efficient hiPSC generation from patient non-mobilized peripheral blood-derived CD34+ cells using the auto-erasable Sendai virus vector

    Get PDF
    Background: Disease modeling with patient-derived induced pluripotent stem cells (iPSCs) is a powerful tool forelucidating the mechanisms underlying disease pathogenesis and developing safe and effective treatments. Patientperipheral blood (PB) cells are used for iPSC generation in many cases since they can be collected with minimuminvasiveness. To derive iPSCs that lack immunoreceptor gene rearrangements, hematopoietic stem and progenitorcells (HSPCs) are often targeted as the reprogramming source. However, the current protocols generally requireHSPC mobilization and/or ex vivo expansion owing to their sparsity at the steady state and low reprogrammingefficiencies, making the overall procedure costly, laborious, and time-consuming.Methods: We have established a highly efficient method for generating iPSCs from non-mobilized PB-derivedCD34+ HSPCs. The source PB mononuclear cells were obtained from 1 healthy donor and 15 patients and werekept frozen until the scheduled iPSC generation. CD34+ HSPC enrichment was done using immunomagnetic beads,with no ex vivo expansion culture. To reprogram the CD34+-rich cells to pluripotency, the Sendai virus vectorSeVdp-302L was used to transfer four transcription factors: KLF4, OCT4, SOX2, and c-MYC. In this iPSC generationseries, the reprogramming efficiencies, success rates of iPSC line establishment, and progression time wererecorded. After generating the iPSC frozen stocks, the cell recovery and their residual transgenes, karyotypes, T cellreceptor gene rearrangement, pluripotency markers, and differentiation capability were examined.Results:We succeeded in establishing 223 iPSC lines with high reprogramming efficiencies from 15 patients with 8 different disease types. Our method allowed the rapid appearance of primary colonies (~ 8 days), all of which were expandable under feeder-free conditions, enabling robust establishment steps with less workload. After thawing, the established iPSC lines were verified to be pluripotency marker-positive and of non-T cell origin. A majority of the iPSC lines were confirmed to be transgene-free, with normal karyotypes. Their trilineage differentiation capability was also verified in a defined in vitro assay.Conclusion:This robust and highly efficient method enables the rapid and cost-effective establishment of transgene-free iPSC lines from a small volume of PB, thus facilitating the biobanking of patient-derived iPSCs and their use for the modeling of various diseases

    Quandles and Symmetric Spaces 2022

    No full text
    corecore