Detection of Hematopoietic Stem Cells by Flow Cytometry

Author(s):  
Kuanyin K. Lin ◽  
Magaret A. Goodell
2012 ◽  
Vol 83A (1) ◽  
pp. 27-37 ◽  
Author(s):  
Allison Mayle ◽  
Min Luo ◽  
Mira Jeong ◽  
Margaret A. Goodell

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2429-2429
Author(s):  
Tomohiko Ishibashi ◽  
Takafumi Yokota ◽  
Michiko Ichii ◽  
Yusuke Satoh ◽  
Takao Sudo ◽  
...  

Abstract Identification of novel markers associated with hematopoietic stem cells (HSCs) is important to progress basic and clinical research regarding the HSC biology. We previously reported that endothelial cell-selective adhesion molecule (ESAM) marks HSCs throughout life in mice (Yokota et al. Blood, 2009). We also demonstrated that ESAM can be a useful indicator of activated HSCs after bone marrow (BM) injury and that ESAM is functionally important for recovering hematopoiesis by using ESAM knockout mice (Sudo et al. J Immunol, 2012). However, the discrepancy between species has been a long-standing obstacle to apply findings in mice to human. For example, established murine HSC markers such as Sca-1 or CD150 are not expressed on human HSCs. Thus, it is important to know if ESAM marks HSCs beyond species and serves as a functional molecule for the HSC property, but information regarding ESAM expression in human HSCs has been quite limited. In this study, we have examined the ESAM expression pattern on human HSCs derived from diverse sources. In addition, we have performed functional assessment of the ESAM-expressing cells. Cord blood (CB), aspirated BM, and granulocyte-colony stimulating factor-mobilized peripheral blood (GMPB) were obtained from healthy donors. BM was also obtained from head of femora of patients who received the hip replacement surgery. All of the protocols were approved by the Institutional Review Board of Osaka University School of Medicine, and we obtained the written agreement form with informed consent from all participants. Mononuclear cells were separated using Ficoll centrifugation from CB, aspirated BM and GMPB. For preparation of BM cells adjacent to bone tissues, trabecular tissues of femora were treated with 2 mg/ml collagenase IV and DNase and gently agitated for 1 hour at 37 °C. Collected cells were analyzed using flow cytometry for cell surface expression of ESAM and other markers. Further, the CD34+ CD38−cells were fractionated according to the intensity of ESAM expression and evaluated in vivo and in vitro functional assays. Flow cytometry analyses revealed that the majority of CB CD34+ CD38− cells expressed ESAM. According to the expression level, CB CD34+ CD38− cells could be subdivided into three populations, namely ESAM−/Low, ESAMHigh, and ESAMBright. While all CB contained a robust ESAMHigh population in CD34+ CD38− cells, the percentage of ESAMBright cells varied widely among CB samples. The ESAMHigh CD34+ CD38− cells also expressed CD90 and CD133, which are known as HSC markers. Methylcellulose colony-forming assays and limiting dilution assays revealed that ESAMHigh fraction enriches primitive hematopoietic progenitors. Further, ESAMHigh cells also reconstituted the long-term human hematopoiesis in NOD/Shi-scid, IL-2Rγnull (NOG) mice. Therefore, as in mice, ESAMHighmarks authentic HSCs in human. On the other hand, ESAMBright CD34+ CD38− cells showed low colony-forming activities and no reconstitution of human hematopoiesis in NOG mice. These ESAMBright CD34+ CD38− cells expressed CD118/leukemia inhibitor factor receptor and endothelial markers such as VE-Cadherin, Flk-1, and CD146, but not CD45. These results suggested that ESAMBright cells in the CB CD34+ CD38− fraction are non-hematopoietic cells. With respect to the other HSC sources such as aspirated BM and GMPB, almost all CD34+ CD38− cells were ESAMHigh and ESAMBright cells were not found in this fraction. Interestingly, however, ESAMBright cells were found in the CD34+ CD38− fraction isolated from collagenase-treated femora. These BM-derived ESAMBright CD34+ CD38− cells expressed endothelial markers as did the CB-derived cells. They could generate CD31+endothelial cells, but not hematopoietic cells in coculture with MS5 stromal cells with vascular endothelial growth factor, stromal-cell-derived factor, and interleukin 16. In conclusion, ESAM expression serves as a marker to enrich HSCs in human regardless of the HSC sources. In addition, the very high intensity of this marker might be useful to isolate non-hematopoietic progenitors from CD34+ CD38− cells, which has been conventionally used as human HSCs. The common feature of ESAM expression of murine and human HSCs suggests a possibility that functional significance of ESAM expression obtained from mouse studies could be applicable to human. Disclosures: No relevant conflicts of interest to declare.


2004 ◽  
Vol 57A (2) ◽  
pp. 100-107 ◽  
Author(s):  
Joachim Oswald ◽  
Birgitte Jørgensen ◽  
Tilo Pompe ◽  
Fritz Kobe ◽  
Katrin Salchert ◽  
...  

Cytometry ◽  
1987 ◽  
Vol 8 (3) ◽  
pp. 296-305 ◽  
Author(s):  
Kenneth F. McCarthy ◽  
Martha L. Hale ◽  
Paula L. Fehnel

Author(s):  
Mukul Girotra ◽  
Anne-Christine Thierry ◽  
Alexandre Harari ◽  
George Coukos ◽  
Olaia Naveiras ◽  
...  

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1897-1897
Author(s):  
Ramon V. Tiu ◽  
Jennifer J. Powers ◽  
Abdo Haddad ◽  
Ying Jiang ◽  
Jaroslaw P. Maciejewski

Abstract Members of the signaling lymphocytic activation molecule (SLAM) family, including CD150, CD48 and CD244, were shown to precisely distinguish more committed lineage restricted progenitor cells from pluripotent and multipotent murine hematopoietic stem cells (HSC; Kiel et al; 2005 Cell). Similar SLAM profiles may also be present on HSC subsets in humans. We hypothesized that these SLAM markers may be indicators not only of stem cell potential in normal hematopoiesis but also distinguish a subset of the most immature malignant precursors of leukemia. In agreement with the concept of a “cancer stem cell,” the presence of leukemic stem cell population may be an indicator of important clinical and biological properties. We first tested the distribution of CD150, CD48 and CD244 antigens on human CD34+ cells derived from 7 control individuals using 4-color flow cytometry. CD34+ cells were measured in the blast gate based on side scatter and CD45 expression. Within CD34+ blasts, expression of CD48, CD150, and CD244 was detected on 16.71%±9.69, 6.53%±2.93, and 26.92%±6.95 of cells respectively. Subsequently, we investigated SLAM expression in 9 immature leukemic cell lines, including KG-1, K562, U937, HEL, HL60, MKN-95, NB-4, Kasumi and UT7, and found increased expression of SLAM markers in KG-1 (CD48+, CD150+, CD244+) and Kasumi (CD48−, CD150−, CD244+). Consequently, none of the leukemic cells showed pluripotent/multipotent SLAM profiles. We then compared the SLAM marker expression on blasts from patients with AML and MDS with that of CD34+ cells from normal controls. We studied a total of 28 patients: 11 MDS (2 low grade, 5 advanced MDS, 3 MDS/MPD overlap) and 10 AML (FAB: 3 M1, 2 M2, 1 M3, 2 M4/M4E0 and 2 M6). In our cohort, 8/10 AML patients expressed one of the three SLAM markers; 6/10 were CD150−CD48−CD244+ (63.57%±6.96) and 2/10 were CD150+CD48−CD244−(46%±10.96) suggestive of the presence of either pluripotent or multipotent leukemic stem cell phenotype. In the MDS cohort, 8/11 patients expressed one of three SLAM markers, 7/11 were CD150−CD48−CD244+ (41.21% ± 8.9) and 1/11 were CD150+CD48−CD244− (1.26%±0.59) again consistent with a profile derived from either pluripotent or multipotent stem cells. None of the MDS and AML patients had either co-expression of CD244 and CD48 or increased expression of CD48 alone. Two of the M1 type AML patients with CD150−CD48−CD244+ phenotype received prior chemotherapy and achieved complete remission on bone marrow biopsy and flow cytometry using traditional blast markers. In some, we conclude that the SLAM receptor markers may be associated with certain types of leukemic blasts and may be useful in the identification of leukemic stem cell population in both MDS and AML.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3682-3682
Author(s):  
Chiharu Sugimori ◽  
Kanako Mochizuki ◽  
Hirohito Yamazaki ◽  
Shinji Nakao

Abstract Acquired aplastic anemia (AA) is thought to be caused by the immune system attack against hematopoietic stem cells. However, there is no direct evidence that an immune system attack against normal hematopoietic stem cells leads to development of AA. The immune system attack may be directed toward abnormal stem cells given the fact that some patients with myelodysplastic syndrome respond to immunosuppressive therapy. Although the presence of a small population of CD55−CD59− blood cells represents a reliable marker for the immune pathophysiology of AA, little is known regarding when and how such paroxysmal nocturnal hemoglobinuria (PNH)-type cells appear in patients with AA. The development of AA with a small population of PNH-type cells was recently observed in an allogeneic stem cell transplant (SCT) recipient. This patient, a 59-year-old male, who had been treated with allogeneic peripheral blood stem cell transplantation (PBSCT) from an HLA-compatible sibling for treatment of very severe AA in March 2002, developed severe pancytopenia in December 2005. Late graft failure (LGF) without residual recipient cells was diagnosed based on the results of a chimerism analysis. Sensitive flow cytometry failed to reveal any increase in the proportion of CD55−CD59− PNH-type blood cells. The patient underwent a second PBSCT from the original donor without preconditioning in February 2006. Although his pancytopenia was completely resolved by day 20, his blood counts gradually decreased from day 60 without any apparent complications. Flow cytometry revealed small populations of PNH-type granulocytes in his peripheral blood (Figure 1). Both the PNH-type and normal phenotype granulocytes were of donor origin. PIG-A gene analyses showed the PNH-type granulocytes in the patient to be a clonal stem cell with an insertion of thymine at position 593 (codon 198). Similar results were obtained from the sorted PNH-type granulocytes obtained 6 months later. The patient was treated with horse antithymocyte globulin and cyclosporine. The patient required no further transfusions after 88 days of the therapy and remains well as of August, 2007. The small population of PNH-type cells was not detectable in any of 50 SCT recipients showing stable engraftment or in an AA patient suffering graft rejection after a SCT. These findings suggest that some factors expressed by the patient induced an immune system attack against autologous hematopoietic cells, leading to de novo development of donor-cell derived AA. This is the first evidence that an immune system attack against normal hematopoietic stem cells results in AA associated with a clonal expansion of a PIG-A mutant which may originally be present in the donor bone marrow. Figure Figure


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5888-5888
Author(s):  
Laurel Christine Truscott ◽  
Tzu Ting Chiou ◽  
Roy L. Kao ◽  
Satiro N. De Oliveira

Abstract Background:Gene-modified human hematopoietic stem cells (HSC) have been used as tools to introduce genes into organisms to correct metabolic or cellular disorders caused by defective or absent genes, or to prevent or treat diseases. We propose the innovative concept of using gene-modification of HSC to enable persistent generation of multilineage immune effectors able to directly target cancer cells. Stable transgene integration has been done by modification of HSC using retroviral or lentiviral vectors. In early clinical trials, there was a high incidence of retroviral-mediated insertional mutagenesis, prompting the need for alternative approaches. Despite current research data suggesting lentiviral vectors are relatively safer, concerns regarding malignant transformation, abnormal hematopoiesis and autoimmunity still exist, making the co-delivery of a suicide gene a necessary safety measure. We have compared two potential suicide gene systems for this use: the herpes simplex virus thymidine kinase (HSV-sr39TK) and a truncated epidermal growth factor receptor (EGFRt). To enable future clinical applications, those transgenes were co-delivered with anti-CD19 chimeric antigen receptor (CAR). We hypothesize gene-modified HSC can be successfully targeted and ablated using a suicide gene system, and HSV-sr39TK and EGFRt will both be effective suicide gene systems. Methods: Third generation self-inactivating lentiviral vector constructs were used to co-deliver an anti-CD19 CAR and HSV-sr39TK or EGFRt. Each suicide gene's efficacy was tested using cytotoxicity assays. Jurkat and primary cells expressing HSV-sr39TK were incubated with varying concentrations for ganciclovir. For the EGFRt suicide gene system, an antibody-dependent cell-mediated cytotoxicity (ADCC) assay was used with EGFRt-transduced Jurkat and primary cells as targets, with the target cells incubated with leukocytes and varying ratios of the EGFR-specific monoclonal antibody cetuximab. For both assays, the cells were stained and analyzed by flow cytometry to determine the percentage of surviving cells. For in vitro assays, gene-modified HSC were differentiated into myeloid cells over ten days to allow transgene expression before cytotoxicity challenges. Gene-modified HSC were also engrafted into immunodeficient NSG mice for in vivo experiments using treatments with intraperitoneal ganciclovir 50mg/kg/day over 5 days or intraperitoneal cetuximab 1mg/mouse/day over 12 days. Persistence of gene-modified cells was assessed by flow cytometry and ddPCR of animal tissues. Results:For the HSV-sr39TK transgene, primary human T-cells and myeloid cells differentiated from gene-modified human HSC had consistently decreased rates of survival for the HSV-sr39TK transduced cells when incubated with varying concentrations of ganciclovir, as compared to both the non-transduced and control-transduced cells, with remaining survival of gene-modified cells of 20% in the assays employed. For the EGFRt transgene, cytotoxicity was significantly increased (p<0.0001) in target cells expressing EGFRt after 4-hour incubation of leukocytes with target cells and cetuximab 1µg/mL, compared with either EGFRt+ cells without cetuximab, and non-transduced cells with or without cetuximab. This was seen at all effector to target ratios and average remaining gene-modified cells also approached 20% in the assays employed. Mice humanized with gene-modified HSC presented significant ablation of gene-modified cells after treatment with either ganciclovir for the HSV-sr39TK transgene, or cetuximab for the EGFRt transgene, with successful ablation of gene-modified cells. Remaining detected gene-modified cells in both models were close to background on flow cytometry and one to two logs of decrease of vector copy numbers by ddPCR in mouse tissues. Conclusions: Ganciclovir-mediated killing of HSV-sr39TK transduced cells was shown to be effective in cells differentiated from gene-modified human HSC. Cetuximab ADCC of EGFRt-modified cells also determined effective killing. These results give proof of principle for CAR-modified HSC regulated by suicide gene, and further studies are needed to enable full clinical translation of this approach. Different ablation approaches, such as inducible caspase 9 or co-delivery of inert cell surface markers (truncated CD20, truncated EGFR) should be evaluated. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document