Successful transfer of ADA gene in vitro into human peripheral blood CD34+cells by transfecting EBV-based episomal vectors

FEBS Letters ◽  
1998 ◽  
Vol 441 (1) ◽  
pp. 39-42 ◽  
Author(s):  
Etsuko Satoh ◽  
Hideyo Hirai ◽  
Tohru Inaba ◽  
Chihiro Shimazaki ◽  
Masao Nakagawa ◽  
...  
Blood ◽  
1993 ◽  
Vol 82 (12) ◽  
pp. 3600-3609 ◽  
Author(s):  
N Sato ◽  
K Sawada ◽  
K Koizumi ◽  
T Tarumi ◽  
M Ieko ◽  
...  

Abstract To elucidate the role of recombinant human colony-stimulating factors (CSFs) for expanding peripheral blood (PB) CD34+ cells, these cells were purified up to 94.5% +/- 1.3% and the effects of individual and combined CSFs on the proliferation and differentiation of these cells were studied in a 7-day suspension culture. The majority of CD34+ cells coexpressed CD38 (81.8% +/- 5.1%), but was negative for CD33 (88.5% +/- 3.4%). Among the individual CSFs examined, recombinant interleukin-3 (rIL-3) was identified as the most potent factor for expanding PB progenitor cells and increased nonerythroid progenitor cells 13- +/- 4- fold (P < .01). Recombinant granulocyte-macrophage colony-stimulating factor (rGM-CSF), recombinant granulocyte-CSF (rG-CSF), recombinant macrophage-CSF (rM-CSF), rIL-6, rIL-11, and recombinant stem cell factor (rSCF) did not alone expand nonerythroid progenitor cells. A combination of 5 CSFs, ie, rIL-3, rIL-6, rGM-CSF, rG-CSF, and rSCF, was identified as the most potent combination of those tested and increased nonerythroid progenitor cells 57- +/- 11-fold. After a 7-day suspension culture of CD34+ cells with these 5 CSFs, CD34+ cells expanded 14.5- fold, and CD34+/CD33- cells and CD34+/CD33+ cells were also expanded 2.9-fold and 307-fold, respectively. Most secondary colonies derived from expanded cells were small; however, the absolute number of large- sized colonies expanded 5.9- +/- 3.3-fold. Thus, the combination of CSFs can achieve a degree of amplification of PB CD34+ cells. The capability of in vitro expansion of PB CD34+ cells as an adjunct to PB stem cell transplantation is worthy of consideration.


Blood ◽  
1993 ◽  
Vol 82 (12) ◽  
pp. 3600-3609 ◽  
Author(s):  
N Sato ◽  
K Sawada ◽  
K Koizumi ◽  
T Tarumi ◽  
M Ieko ◽  
...  

To elucidate the role of recombinant human colony-stimulating factors (CSFs) for expanding peripheral blood (PB) CD34+ cells, these cells were purified up to 94.5% +/- 1.3% and the effects of individual and combined CSFs on the proliferation and differentiation of these cells were studied in a 7-day suspension culture. The majority of CD34+ cells coexpressed CD38 (81.8% +/- 5.1%), but was negative for CD33 (88.5% +/- 3.4%). Among the individual CSFs examined, recombinant interleukin-3 (rIL-3) was identified as the most potent factor for expanding PB progenitor cells and increased nonerythroid progenitor cells 13- +/- 4- fold (P < .01). Recombinant granulocyte-macrophage colony-stimulating factor (rGM-CSF), recombinant granulocyte-CSF (rG-CSF), recombinant macrophage-CSF (rM-CSF), rIL-6, rIL-11, and recombinant stem cell factor (rSCF) did not alone expand nonerythroid progenitor cells. A combination of 5 CSFs, ie, rIL-3, rIL-6, rGM-CSF, rG-CSF, and rSCF, was identified as the most potent combination of those tested and increased nonerythroid progenitor cells 57- +/- 11-fold. After a 7-day suspension culture of CD34+ cells with these 5 CSFs, CD34+ cells expanded 14.5- fold, and CD34+/CD33- cells and CD34+/CD33+ cells were also expanded 2.9-fold and 307-fold, respectively. Most secondary colonies derived from expanded cells were small; however, the absolute number of large- sized colonies expanded 5.9- +/- 3.3-fold. Thus, the combination of CSFs can achieve a degree of amplification of PB CD34+ cells. The capability of in vitro expansion of PB CD34+ cells as an adjunct to PB stem cell transplantation is worthy of consideration.


1996 ◽  
Vol 98 (2) ◽  
pp. 344-354 ◽  
Author(s):  
Meir Shalit ◽  
Sudhir Sekhsaria ◽  
Fei Li ◽  
Stephen Mauhorter ◽  
Siddhartha Mahanti ◽  
...  

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4302-4302
Author(s):  
Meng Qin ◽  
Xin Guan ◽  
Yu Zhang ◽  
Qing-yu Zhang ◽  
Wei Dai ◽  
...  

Abstract It is possible to treat ischemia and hemophilia A diseases by producing sufficient functional human endothelial progenitor cells (EPCs)/endothelial cells (ECs) in vitro, for use with cell therapy in the clinic. We have previously reported the ability to produce FVIII-secreting EPCs/ ECs derived from human cord blood CD34+ cells. About 1412±102 fold expansion over initial EPCs was achieved after culturing for 21 days. An acute liver sinusoidal endothelial cells (LSEC) injury model in NOD/SCID mice was also developed to verify the functional migrating ability of the generated EPCs/ ECs in vivo. Here, we further applied this culturing technique to expand and subsequently differentiate CD34+ cells into the EPCs/ ECs derived from mobilized peripheral bloods of both human and cynomolgus monkeys. In brief, the CD34+ cells were isolated from human peripheral bloods or from monkeys (n=10) mobilized with human G-CSF/SCF. In the first 6 days, the isolated CD34+ cells were expanded in modified IMDM medium supplemented with human cytokine combinations of SCF, Flt-3L, TPO, IL-3, GM-CSF, and VEGF. From days 7 to 36, the adhering EPCs/ ECs were subsequently differentiated in EBM-2 basal medium with 20% FBS and endothelial growth factors of VEGF、IGF、EGF、FGF, and fibronectin. The purities and phenotypes of the induced EPCs/ECs were assessed in vitro by antibodies against human CD31, vWF, and FVIII for the human or Dil- acetylated- low density lipoprotein (ac-LDL) and FITC-lectin double staining for the monkey cells.In addition, the safety and efficacy of the induced monkey EPCs/ECs was determined in vivo by autologous transplantation in monkey LSEC injury model, which was induced by a toxic agent, monocrotaline (MCT), to disrupt the sinusoidal endothelial barrier and stimulate the incorporation of transplanted cells into liver parenchyma. In the transplantation group (n=7), each monkey was injected with double labeled autologous EPCs/ECs preparations (2×108 cells/500μl in saline), whereas in the control group (n=3) was injected with the same volume saline via hepatic portal vein injections. The cross-sections (20µm in depth) of fixed hepatic tissues were analyzed for grafting and functional migration of transplanted EPCs/ECs. The transplanted cells were identified by lenti-viral gene expressed with green fluorescent protein (red) or direct observation using anti-monkey IgG -microbead- FITC conjugates (green). For in vitro induced EPCs/ECs derived from human peripheral blood cell, the expansion of 834.58±119.03 fold was achieved from the CD34+/VEGFR2+ EPCs on day 21. Total more than 2x 108 FVIII-producing EPCs / ECs were produced from one collection of human peripheral blood (250 mL). On the other hand, the CD34+/VEGFR2+ EPCs (3.6×104 ±2.1×103) from one collection of monkey peripheral blood (20ml) were expanded up to 1274±166 fold and 7211±372 fold on days 24 and 36, respectively (n=4). The EPCs were reached at a logarithmic growth from days 12 to 45. The induced cells can be frozen and resuscitated during any stage of the culturing process. The formation of EC tubes was observed from day 24. Over 80% of expanded cells were EPC/EC-specific and identified by Dil-ac-LDL and FITC-lectin double staining on day 36. All monkeys recoveredfrom the surgeries of portal vein injection and resumed normal diet and behavior after autologous transplantation with cultured EPCs/ECs. Similarly, the routine blood analysis and liver functional enzymes were at the normal level, and no other apparent side effects were observed. About 3.2±1.4% and 2.1±1.1% of liver cells were observed as Dil-ac-LDL and FITC-lectin double positive in the liver cryosections (25 sections per monkey) on days 7 and 14, respectively, indicating that autologous transplanted EPCs/ECs were capable of repopulating into functional ECs in vivo. Furthermore, the injected EPCs/ECs were scattered in the intercellular spaces of hepatocytes at the hepatic tissues on day 14, suggesting that the transplanted cells could migrate towards injured LSEC sites and reconstitute structurally the sinusoidal endothelial compartment in monkey livers. In summary, the large-scale EPCs/ECs were produced from CD34+ cells of both human and monkey peripheral bloods in vitro. The safety and functions of the EPCs/ECs were confirmed in mice and cynomolgus monkeys, strongly suggesting the potential application of these FVIII-producing EPCs/ECs to future clinical study. Disclosures Qin: Biopharmagen. corp: Employment.


1997 ◽  
Vol 185 (6) ◽  
pp. 1131-1136 ◽  
Author(s):  
Dirk Strunk ◽  
Claudia Egger ◽  
Gerda Leitner ◽  
Daniel Hanau ◽  
Georg Stingl

We have recently described a system for the generation of dendritic cells (DC) and Langerhans cells (LC) from defined CD34+ precursors purified from peripheral blood of healthy adult volunteers (1). This study has now been extended by the characterization of two distinct subpopulations of CD34+ cells in normal human peripheral blood as defined by the expression of the skin homing receptor cutaneous lymphocyte-associated antigen (CLA). CD34+/CLA+ cells from normal peripheral blood were found to be CD71LOW/CD11a+/CD11b+/CD49d+/ CD45RA+ whereas CD34+/CLA− cells displayed the CD71+/CD11aLOW/CD11bLOW/CD49d(+)/ CD45RALOW phenotype. To determine the differentiation pathways of these two cell populations, CD34+ cells were sorted into CLA+ and CLA− fractions, stimulated with GM-CSF and TNF-α in vitro, and then were cultured for 10 to 18 d. Similar to unfractionated CD34+ cells, the progeny of both cell populations contained sizable numbers (12–22%) of dendritically shaped, CD1a+/HLA-DR+++ cells. In addition to differences in their motility, the two dendritic cell populations generated differed from each other by the expression of LC-specific structures. Only the precursors expressing the skin homing receptor were found to differentiate into LC as evidenced by the presence of Birbeck granules. In contrast, CLA− precursor cells generated a CD1a+ DC population devoid of Birbeck granule–containing LC. Provided that comparable mechanisms as found in this study are also operative in vivo, we postulate that the topographic organization of the DC system is already determined, at least in part, at the progenitor level.


Sign in / Sign up

Export Citation Format

Share Document