Efficient isolation of human CD34+CD38- hematopoietic stem cells using immunomagnetic sorting

2017 ◽  
Vol 53 ◽  
pp. S124
Author(s):  
Dariusz Krenz ◽  
Faidra Aivazidou ◽  
Andreas Bosio ◽  
Sebastian Knöbel ◽  
Ute Bissels
PLoS ONE ◽  
2010 ◽  
Vol 5 (10) ◽  
pp. e13109 ◽  
Author(s):  
Yoshinori Sato ◽  
Hiroshi Takata ◽  
Naoki Kobayashi ◽  
Sayaka Nagata ◽  
Naomi Nakagata ◽  
...  

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3178-3178
Author(s):  
Toshinao Kawai ◽  
Uimook Choi ◽  
Po-Ching Liu ◽  
Harry L. Malech

Abstract CD26/DPP-IV (CD26) is a membrane-anchored ectoenzyme with N terminus exopeptidase activity that cleaves X-Pro-dipeptides. Stromal cell-derived factor 1 (SDF-1) and its receptor CXCR4 play a central role in trafficking of hematopoietic stem cells in the bone marrow. SDF-1 has a proline second from the N-terminus and is cleaved by CD26. Mouse hematopoietic progenitors express CD26 and a brief treatment of these cells with Diprotin A (Ile-Pro-Ile), a specific inhibitor of CD26, enhances engraftment. We examined the effect of Diprotin A treatment of peripheral blood human CD34+ stem cells (PBSC) with respect to subsequent responses to SDF-1 and with respect to engraftment in the NOD/SCID mouse xenograft model. We found that human CD34+ PBSC with colony forming potential are unlike mouse hematopoietic stem cells in that they lack the equivalent of CD26; and also are unlike mouse cells in that their response to SDF-1 and their engraftment in the NOD/SCID xenograft model are not affected by pre-treatment with Diprotin A. However, administration of Diprotin A intravenously to the NOD/SCID mouse at the time of transplant of human PBSC greatly enhances engraftment of the human PBSC, suggesting an effect primarily on the mouse stroma. Previous reports suggested that 70% of mouse lineage negative/sca-1 positive hematopoietic stem cells express CD26. However, freshly selected mobilized human CD34+ PBSC do not express detectable CD26, though after 4 days and 7 days of ex vivo culture in growth factors (SCF, flt3-ligand, TPO, IL3) 8.9% and 26.6% of cells express CD26, indicating that CD26 may only appear in later progenitors. At day 4 the cultured human PBSC were sorted by flow cytometry into CD26 positive and negative fractions. Only the CD26 negative fraction contained colony forming cells. 4 day-cultured human PBSC were exposed to Diprotin A 5mM for 15 minutes, washed and used in a filter transwell migration assay in response to SDF-1 at concentrations from 0.5 to 10 nM. There was no statistical difference between migration of Diprotin A treated and control PBSC, even in experiments with longer treatment with Diprotin A. When these human PBSC were transplanted into NOD/SCID mice there was no difference of engraftment between the Diprotin A treated group and control group. However, when 1x106 of 4 day-cultured PBSC were injected into NOD/SCID mice without or together with 2μmol of Diprotin A, there was a profound enhancement on subsequent engraftment in the group of mice injected with Diprotin A at time of transplant. At 6 weeks after transplantation the CD45+ human cell engraftment of the Diprotin A group was 6-fold increased compared to control group (49.6±8.2% vs. 8.1 ± 3.4%, p<.0001). Taken together with the colony assay, the in vitro migration studies, and lack of effect on engraftment when only the human PBSC are treated with Diprotin A, this result suggests that the enhanced engraftment of human PBSC in NOD/SCID mice is due to an action of Diprotin A on endogenous mouse CD26/DPP-IV (where the target is unknown, but possibly stromal cells). Although, further work is required to determine levels of expression of CD26/DPP-IV in human marrow stromal cells, it is possible to speculate that inhibitors of CD26/DPP-IV activity may provide a novel approach to improve stem cell engraftment in humans.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4622-4622
Author(s):  
Nadja Grund ◽  
Patrick Maier ◽  
Uwe Appelt ◽  
Heike Allgayer ◽  
Frederik Wenz ◽  
...  

Abstract Hematologic side effects of cancer chemotherapy like myelosuppression are frequently dose-limiting. Lentiviral gene therapy with cytostatic drug resistance gene transfer to human hematopoietic stem cells (CD34+) is a promising approach to overcome this problem. In this context it is of interest if chemotherapy mediated selection has an impact on lentiviral integration site patterns of transduced hematopoietic stem cells (CD34+). Concerning this issue, human CD34+ cells transduced with a lentiviral self-inactivating (SIN) vector encoding MGMTP140K (the O6-BG resistant mutant of O6-methylguanine- DNA methyltransferase) were in vitro treated with the alkylating agent BCNU. For integration site analysis LM-PCR was performed and integration patterns of the treated and untreated CD34+ cells were analyzed and compared with an in silico set of 106 random integrations. We found different integration preferences of the lentiviral vector between either the treated (82 integrations) or the untreated (30 integrations) CD34+ cells and the in silico set: both groups showed chromosomal preferences, a significant bias for integrations in genes (74,4% in the treated, respectively 70% in the untreated to 40% in the in silico group), especially by favouring introns, a random integration distribution regarding transcription start sites (TSS), and most importantly no significant differences concerning the number of integrations in or near cancer genes. Concerning all integration characteristics we could not find significant differences when comparing the untreated with the treated group. In conclusion, the general distribution of lentiviral integrations in either untreated or treated human CD34+ cells showed no distinct differences between both groups but significant differences compared to the in silico integration set. These results suggest that chemoselection of cells lentivirally overexpressing a specific chemoresistence gene might not influence the integration pattern. Therefore chemotherapy pressure seems not to hamper the safety of lentiviral vectors in gene transfer studies.


Blood ◽  
2003 ◽  
Vol 101 (1) ◽  
pp. 112-118 ◽  
Author(s):  
Mo A. Dao ◽  
Jesusa Arevalo ◽  
Jan A. Nolta

Abstract The cell surface protein CD34 is frequently used as a marker for positive selection of human hematopoietic stem/progenitor cells in research and in transplantation. However, populations of reconstituting human and murine stem cells that lack cell surface CD34 protein have been identified. In the current studies, we demonstrate that CD34 expression is reversible on human hematopoietic stem/progenitor cells. We identified and functionally characterized a population of human CD45+/CD34− cells that was recovered from the bone marrow of immunodeficient beige/nude/xid (bnx) mice 8 to 12 months after transplantation of highly purified human bone marrow–derived CD34+/CD38− stem/progenitor cells. The human CD45+ cells were devoid of CD34 protein and mRNA when isolated from the mice. However, significantly higher numbers of human colony-forming units and long-term culture-initiating cells per engrafted human CD45+ cell were recovered from the marrow of bnx mice than from the marrow of human stem cell–engrafted nonobese diabetic/severe combined immunodeficient mice, where 24% of the human graft maintained CD34 expression. In addition to their capacity for extensive in vitro generative capacity, the human CD45+/CD34− cells recovered from thebnx bone marrow were determined to have secondary reconstitution capacity and to produce CD34+ progeny following retransplantation. These studies demonstrate that the human CD34+ population can act as a reservoir for generation of CD34− cells. In the current studies we demonstrate that human CD34+/CD38− cells can generate CD45+/CD34− progeny in a long-term xenograft model and that those CD45+/CD34− cells can regenerate CD34+ progeny following secondary transplantation. Therefore, expression of CD34 can be reversible on reconstituting human hematopoietic stem cells.


2019 ◽  
Vol 47 (5) ◽  
pp. 656-660 ◽  
Author(s):  
Sundeep Chandra ◽  
Patrizia Cristofori ◽  
Carlos Fonck ◽  
Charles A. O’Neill

A therapeutic option for monogenic disorders is gene therapy with ex vivo-transduced autologous hematopoietic stem cells (HSCs). Safety or efficacy studies of ex vivo-modified HSCs are conducted in humanized mouse models after ablation of the murine bone marrow and transfer of human CD34+ HSCs. Engrafted human CD34+ cells migrate to bone marrow and differentiate into various human hematopoietic lineages. A 12-week study was conducted in NSG™ mice to evaluate engraftment, differentiation, and safety of human CD34+ cells that were transduced ( ex vivo) with a proprietary lentiviral vector encoding a human gene (BMRN-1) or a mock (green fluorescent protein) vector. Several mice intravenously injected with naive CD34+ cells or transduced CD34+ cells had variable lymphohistiocytic inflammatory cell infiltrates and microgranulomas in the liver and lungs consistent with graft-versus-host disease (GVHD). Spleen, bone marrow, stomach, reproductive tract, but not the skin had similar inflammatory changes. Ex vivo viral transduction of CD34+ cells did not impact engraftment or predispose to xenogeneic GVHD.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4744-4744
Author(s):  
Xingbing Wang ◽  
Xin Chen ◽  
Weihua Ren ◽  
Xiucai Xu ◽  
Kaidi Song ◽  
...  

Abstract High-mobility group box 1 protein (HMGB1) is a chromatin protein and acts as a cytokine involved in inflammation, cell proliferation, differentiation, migration and stem cell recruitment. So far, little is known about its effect on hematopoietic stem cells (HSCs). In this study, we investigated whether receptors for HMGB1 are expressed on human CD34+ HSCs, and whether HMGB1 could affect HSCs proliferation, differentiation and migration in vitro. As examined by FACS analysis and RT-PCR, cord blood CD34+ HSCs express the HMGB1 receptors RAGE (receptor for advanced glycation end products), TLR2 (Toll-like receptor 2) and TLR4. To study the effects of HMGB1 on CD34+ HSCs proliferation and differentiation, freshly isolated cord blood CD34+ HSCs were cultured for 7 days in medium alone or in the presence of HMGB1. Flow cytometric analysis showed that HMGB1 (50ng/ml) can induce the differentiation of CD34+ HSCs along the granulo-monocytic (CD14+, CD13+) lineage and erythropoiesis (CD71+). In contrast, HMGB1 did not induce the expression of CD41, a marker for megakaryocyte lineage. The numbers of cells cultured in the presence of HMGB1 were always increased in comparison with controls. Furthermore, higher numbers of granulomonocytic progenitors (CFU-GM), erythroid progenitors (BFU-E), and CFU-MIX were confirmed by CFC assays in a HMGB1 dose dependent manner after 14-day culture. The results suggest that HMGB1 enhances CD34+ HSCs proliferation and differentiation. We next assessed the effects of HMGB1 on HSCs migration by chemotaxis assay using Boyden chambers. HMGB1 dose-dependently increased the chemotactic migration of CD34+ HSCs. A neutralizing anti-RAGE antibody significantly blocked the HMGB1-induced migration of HSCs, whereas neutralizing TLR2 and TLR4 antibodies did not significantly influence HMGB1-stimulated HSCs migration, suggesting that the migratory effect of HMGB1 on human HSCs is predominantly mediated by RAGE. In summary, our results provide the first report of HMGB1 receptors expression profile of human cord blood CD34+ HSCs and demonstrate that HMGB1 can increase the proliferation and migration of HSCs and directly induce of HSCs along myeloid differentiation and erythropoiesis in vitro. Further studies will be needed to clarify the mechanism of HMGB1 activation and the physiological function of HMGB1 in HSCs in vivo.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4099-4099
Author(s):  
Aaron C. Logan ◽  
Agnieszka Czechowicz ◽  
Benjamin V. Kelley ◽  
Theingi M. Thway ◽  
Ivan Magana ◽  
...  

Abstract Abstract 4099 Engraftment of allogeneic hematopoietic stem cells (HSC) requires conditioning to overcome immunologic and anatomic barriers preventing access to the marrow niche. Most patients who undergo allogeneic hematopoietic cell transplantation (allo-HCT) are prepared with cytotoxic chemotherapy and/or radiation to eliminate these barriers, and to facilitate eradication of malignant cells, if present. Many non-malignant conditions, such as primary immunodeficiencies, hemoglobinopathies, and autoimmune diseases may be successfully treated by transplantation of allogeneic HSC, but the toxicity of conventional conditioning regimens is, in many cases, prohibitive. Targeted elimination of barriers to the HSC niche would be a preferable approach. Signaling via the c-Kit receptor (CD117) is critical for the maintenance of pluripotent HSC. Anti-CD117 monoclonal antibodies (mAbs) deplete HSC and facilitate engraftment of donor HSC in a mouse model of severe combined immunodeficiency (SCID) (Czechowicz et al., Science, 2007). Patients with SCID are highly susceptible to infections, but also have limited immunologic barriers to alloengraftment, making this patient population ideal for studying targeted stem cell depletion to facilitate allo-HSC engraftment. We identified a clinical grade humanized anti-human CD117 mAb (anti-hCD117) as a potential candidate for this purpose. Anti-hCD117 significantly inhibited mitosis in human cord blood and bone marrow derived HSC (Lin−CD34+CD38−CD90+CD45RA−) in liquid and methylcellulose culture containing Flt3 ligand, stem cell factor (SCF), thrombopoietin (TPO), IL-3, and IL-6. To assess in vivo activity of anti-hCD117, we employed it alone, or in combination with alemtuzumab (anti-CD52), to deplete human stem and differentiated cells from hematopoietically humanized NOD/scid/IL2Rg−/− (HuNSG) mice. Pups were conditioned with 100cGy and then humanized by injection of 2000–4000 human HSC into the facial vein on day p2 or intrahepatically on day p4–5. After permitting hematopoietic stabilization for 4–6 months, we confirmed multi-lineage xenochimerism in the peripheral blood (PB) and bone marrow (BM) prior to mAb treatment. After a single treatment with anti-hCD117, mice were depleted of total human leukocytes a median 60% (35–100%; n=11) in the PB and 100% (84–100%; n=10) in the BM at 6 weeks after treatment, with >80% depletion of human myeloid cells in both compartments. Partial recovery of human chimerism was observed at 16 weeks, consistent with recovery of some LT-HSC after anti-hCD117 therapy. The addition of anti-CD52 facilitated clearance of human lymphoid cells not eradicated by anti-hCD117. Human HSC and progenitor cells (Lin−CD34+CD117+; HS/PC) in the bone marrow decreased from 0.4% (0–1.7%) to 0% (0–0.1%; n=10) 6 weeks after treatment with anti-hCD117. We then modeled a human transplant by treating HuNSG mice with anti-hCD117, anti-CD52, or both, to deplete their primary human graft. After monitoring mAb catabolism by ELISA, mice received a second (non-HLA matched) human CD34+ HS/PC graft modified to express the green fluorescent protein using a lentivector. After overnight prestimulation in XVIVO-15 supplemented with SCF, Flt3 ligand, TPO, and IL-3, human CD34+ HS/PC were exposed for 18 hours to lentivector at 1×108 TU/mL. Cells were washed and 80,000 transduced CD34+ HS/PC were injected IV into untreated and mAb-conditioned HuNSG mice. After 6 weeks, PB was evaluated and demonstrated GFP+hCD45+cells in 3/5 (60%) mice treated with anti-hCD117 + anti-CD52, 0/5 mice treated with either anti-hCD117 or anti-CD52 alone, and 1/5 untreated mice. Anti-hCD117 is a promising reagent for depletion of human HSC and facilitation of allo-HSC engraftment. Although anti-hCD117 alone capably depletes human CD34+CD117+ HS/PC and myeloid chimerism in HuNSG mice, the addition of anti-CD52 facilitates engraftment, possibly by reducing alloreactive rejection by T cells from the primary graft. Additional HuNSG mice are receiving second human transplants following mAb conditioning to further explore the utility of combining anti-hCD117 and anti-CD52 for this purpose. These studies will lead the way to minimally toxic allogeneic HSC transplant regimen, and in a broader view, to the application of targeted biological therapies that deplete endogenous stem cells and facilitate their replacement with allogeneic or gene-corrected stem cells. Disclosures: Thway: Amgen, Inc.: Employment. Magana:Amgen, Inc.: Employment. Weissman:Amgen, Inc.: Equity Ownership.


Sign in / Sign up

Export Citation Format

Share Document