Stem Cell Research & Therapy (Jun 2019)

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

  • Takashi Okumura,
  • Yumi Horie,
  • Chen-Yi Lai,
  • Huan-Ting Lin,
  • Hirofumi Shoda,
  • Bunki Natsumoto,
  • Keishi Fujio,
  • Eri Kumaki,
  • Tsubasa Okano,
  • Shintaro Ono,
  • Kay Tanita,
  • Tomohiro Morio,
  • Hirokazu Kanegane,
  • Hisanori Hasegawa,
  • Fumitaka Mizoguchi,
  • Kimito Kawahata,
  • Hitoshi Kohsaka,
  • Hiroshi Moritake,
  • Hiroyuki Nunoi,
  • Hironori Waki,
  • Shin-ichi Tamaru,
  • Takayoshi Sasako,
  • Toshimasa Yamauchi,
  • Takashi Kadowaki,
  • Hiroyuki Tanaka,
  • Sachiko Kitanaka,
  • Ken Nishimura,
  • Manami Ohtaka,
  • Mahito Nakanishi,
  • Makoto Otsu

DOI
https://doi.org/10.1186/s13287-019-1273-2
Journal volume & issue
Vol. 10, no. 1
pp. 1 – 16

Abstract

Read online

Abstract Background Disease modeling with patient-derived induced pluripotent stem cells (iPSCs) is a powerful tool for elucidating the mechanisms underlying disease pathogenesis and developing safe and effective treatments. Patient peripheral blood (PB) cells are used for iPSC generation in many cases since they can be collected with minimum invasiveness. To derive iPSCs that lack immunoreceptor gene rearrangements, hematopoietic stem and progenitor cells (HSPCs) are often targeted as the reprogramming source. However, the current protocols generally require HSPC mobilization and/or ex vivo expansion owing to their sparsity at the steady state and low reprogramming efficiencies, making the overall procedure costly, laborious, and time-consuming. Methods We have established a highly efficient method for generating iPSCs from non-mobilized PB-derived CD34+ HSPCs. The source PB mononuclear cells were obtained from 1 healthy donor and 15 patients and were kept 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 vector SeVdp-302L was used to transfer four transcription factors: KLF4, OCT4, SOX2, and c-MYC. In this iPSC generation series, the reprogramming efficiencies, success rates of iPSC line establishment, and progression time were recorded. After generating the iPSC frozen stocks, the cell recovery and their residual transgenes, karyotypes, T cell receptor 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.

Keywords