Hematopoietic Origin of Fibroblasts and Their Precursors.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 672-672 ◽  
Author(s):  
Yasuhiro Ebihara ◽  
Masahiro Masuya ◽  
Russell Owens ◽  
Su Yang ◽  
Richard P. Visconti ◽  
...  

Abstract Recent studies indicate that hematopoietic stem cells (HSCs) are capable of reconstituting a number of non-hematopoietic organs and tissues. To define the potential of the HSC precisely, we initiated transplantation studies of a clonal population of cells derived from a single HSC. We used the bone marrow (BM) of transgenic enhanced green fluorescent protein (EGFP) mice as the source of donor HSCs. We demonstrated the HSC origin of glomerular mesangial cells (Masuya et al, Blood 101: 2215, 2003) and brain microglial cells and pericyte-like perivascular cells (Hess et al, Exp Neurol 186: 134, 2004). These observations and the fact that glomerular mesangial cells and pericytes are considered myofibroblasts suggested that fibroblasts are also derived from HSCs. In this abstract, we present evidence for HSC origin of fibroblasts and their precursors, BM fibroblast colony-forming units (CFU-F) and peripheral blood (PB) fibrocytes. Lin−Sca-1+c-Kit+CD34− cells from the BM of adult EGFP mice were individually sorted into 96 well Corning plates and cultured for 7 days in the presence of Steel factor and IL-11 or Steel factor and G-CSF. Viable clones consisting of fewer than 20 cells were individually transplanted into lethally irradiated mice. EGFP+ mononuclear cells were sorted from the BM cells of recipients showing high-level, multilineage hematopoietic reconstitution and assayed for CFU-F in Retronectin-treated tissue culture plates. Colonies consisting of more than 50 adherent cells were all EGFP+. The majority of the cells comprising the colonies were fibroblast-like, exhibited spindle-shaped or polygonal cytoplasm and had clear, ovoid nuclei. Flow cytometric analyses revealed that these cells expressed collagen-1 and discoidin domain receptor 2 (DDR2) and exhibited a decreased intensity of CD45. RT-PCR analysis of these cells revealed the presence of mRNA for procollagen 1 alpha-1, vimentin, fibronectin and DDR2. Next we analyzed the PB for donor origin fibrocytes, a fibroblast-like cell type that expresses both fibroblastic and hematopoietic phenotypes in culture. When nucleated PB cells from clonally engrafted mice were cultured on fibronectin-coated dishes, proliferation of EGFP+ fibroblast-like cells was detected. Only one-third of the EGFP+ cells expressed CD45 and most of the EGFP+ cells expressed both collagen-I and DDR2. Similar results were obtained with EGFP+ cells from mice transplanted with 100 uncultured Lin−Sca-1+c-Kit+CD34− cells or 1x106 BM nucleated cells. These studies excluded the effects of short-term culture on HSC differentiation and established the HSC origin of CFU-F and fibrocytes. Classic studies of CFU-F by Friedenstein and others have led to the general belief that mesenchymal stem cells (MSCs), rather than HSCs, generate a number of tissues including adipocytes, osteoblasts, chondrocytes, myocytes and vascular endothelial cells. Here we unequivocally demonstrated that CFU-F are of HSC origin, warranting a re-evaluation of the relationship between HSCs and MSCs.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1188-1188
Author(s):  
Fumihito Tajima ◽  
Yuki Nakamura ◽  
Yoshikazu Murawaki ◽  
Goshi Shiota

Abstract How hematopoietic stem cells (HSC) in bone marrow (BM) contribute to liver regeneration remains to be resolved. The mechanisms which mobilize HSC from BM to peripheral blood (PB) and govern their homing to the liver are unknown. Hepatocyte growth factor (HGF) is locally and generally increased following liver injury, suggesting that it promotes proliferation, adhesion, and survival of hepatocytes. PB stem cells mobilized by cytokines are widely used for clinical transplantation. However, it is not known if HGF can mobilize HSC to PB or if it has the capability to differentiate HSC into hepatocytes. In this study we examined whether HGF can mobilize HSC from BM to PB using the murine stem cell transplantation model. First, we found that HGF transgenic mice, which have high serum levels of HGF concentration, had many colony-forming cells in PB, suggesting HGF increases circulation of HSC and progenitor cells. After determining through RT-PCR analysis that lineage (Lin) − BM cells express the tyrosine kinase receptor c-MET, we speculated that the c-MET-HGF axis modulates the recruitment of HSC from BM to PB. We also examined the effects of exogenous HGF in mobilizing stem cells from BM to PB. To determine whether HGF can mobilize HSC to PB, we investigated the expression of CD34 using flow cytometric analysis. The CD34+ cells in PB mobilized by HGF increased in a dose-dependent pattern and reached a plateau at 0.1mg/kg of recombinant HGF administration. Significant increases in CD34+ cells in PB were noted at 3h after HGF infusion. The continual administration of HGF every 24h increased the CD34+ cells in PB to maximum levels at 4 days. Finally, the absolute number of CD34+ cells in PB after HGF administration was as much as the number of those cells after administration at 12-hour intervals subcutaneously with 0.125mg/kg of recombinant human granulocyte-colony stimulating factor (G-CSF) for 4 consecutive days. To investigate engraftment of the mobilized cells to BM, 0.1mg/kg HGF was injected into Ly-5.1 mice every 24h for 4 days. Lin− cells in PB were collected 3h after the last injection of HGF and then injected into lethally-irradiated Ly-5.2 C57BL/6 mice. Two months after transplantation, the level of engraftment was assessed by analysis of donor (Ly-5.1) cells in the nucleated cells of the PB of recipient mice. The mean percentage of donor cells of mice transplanted with Lin− cells from HGF-treated mice was 1.8%, whereas that of the mice transplanted with untreated PB cells was 0%. Multilineage engraftment was confirmed by the presence of the Thy-1+ cells, B220+ cells, and Mac-1/Gr-1+ cells. When we tested the CD34 expression of the stem cells transplanted, the majority of the cells expressed CD34. Then we tracked single Lin−, Sca-1+, c-kit+, CD34+ PB cells from G-CSF-treated transgenic-enhanced green fluorescent protein (EGFP) mice that were injected into spleen of the CCl4-induced liver-injured B6 mice along with 500 Lin−, Sca-1+, c-kit+, CD34+ PB cells from G-CSF-treated normal B6 mice. Two months later, donor-derived GFP+ cells were identified among recipient hepatocytes in liver-injured mice using immunohistochemistry for GFP. These findings demonstrate that stem cells with long-term engraftment capabilities can be mobilized by HGF, and that HSC in PB mobilized by HGF are capable of differentiating into hepatocytes, suggesting HGF contributes to liver regeneration partly by mobilizing HSC to PB.


Blood ◽  
2000 ◽  
Vol 96 (12) ◽  
pp. 3725-3733 ◽  
Author(s):  
Niels-Bjarne Woods ◽  
Cecilia Fahlman ◽  
Hanna Mikkola ◽  
Isao Hamaguchi ◽  
Karin Olsson ◽  
...  

The ability of lentiviral vectors to transfer genes into human hematopoietic stem cells was studied, using a human immunodeficiency virus 1 (HIV-1)–derived vector expressing the green fluorescence protein (GFP) downstream of the phosphoglycerate kinase (PGK) promoter and pseudotyped with the G protein of vesicular stomatitis virus (VSV). High-efficiency transduction of human cord blood CD34+cells was achieved after overnight incubation with vector particles. Sixteen to 28 percent of individual colony-forming units granulocyte-macrophage (CFU-GM) colonies derived from cord blood CD34+ cells were positive by polymerase chain reaction (PCR) for the GFP gene. The transduction efficiency of SCID-repopulating cells (SRC) within the cord blood CD34+population was assessed by serial transplantation into nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice. When 400 000 cord blood CD34+ cells were transplanted into primary recipients, all primary and secondary recipients contained and expressed the transgene. Over 50% of CFU-GM colonies derived from the bone marrow of these primary and secondary recipients contained the vector on average as determined by PCR. Transplantation of transduced cells in limiting dilution generated GFP+ lymphoid and myeloid progeny cells that may have arisen from a single SRC. Inverse PCR analysis was used to amplify vector-chromosomal junctional fragments in colonies derived from SRC and confirmed that the vector was integrated. These results show that lentiviral vectors can efficiently transduce very primitive human hematopoietic progenitor and stem cells.


2007 ◽  
Vol 25 (18_suppl) ◽  
pp. 17527-17527
Author(s):  
H. Lannert ◽  
T. Able ◽  
S. Leicht ◽  
R. Saffrich ◽  
V. Eckstein ◽  
...  

17527 Background: Stathmin/Op18 is a cytosolic phosphoprotein which regulates the dynamics of microtubules. This regulation is important in mitosis during cell division and in the migration of cells in modification of the cytoskeleton. The process of tumor proliferation and metastasis is characterized by high rates of mitosis and migration into distant tissues. Stathmin itself is regulated by kinases through phosphorylation of mainly 4 different serin sides. In this study, we investigated stathmin- and its kinases expression in native hematopoietic CD34+ stem cells (HSCs) from bone marrow (BM) in comparison to mobilized peripheral blood stem cells (mPBSCs) from G-CSF stimulated donors and leukemic CD34+ cells from patients with AML. Methods: Mononuclear cells were isolated by a standard Ficoll-Hypaque gradient separation method from the different blood sources. An Auto-MACS (Miltenyi) and FACS Vantage SE cell sorter (Becton Dickinson) was used to highly enrich (>99%) CD34+ cells fractions. In comparative proteome analysis, we detected the protein expression of stathmin in mPBSCs, AML CD34+ cells, and in native HSCs from BM. We performed microarray-based gene expression profiles of these cells and focused on kinases regulating stathmin’s activity. Furthermore, we monitored stathmin and its relevant kinases by FACS analyses of the enriched cell fractions and by fluorescence microscopy of bone marrow smears and cytospins. Results: In this study, we have shown in comparative proteome analysis (Q-TOF-MS/MS) that stathmin is expressed in G-CSF mobilized hematopoietic stem cells for the first time and in AML cells. In microarray analysis we indentified up- and down-regulated kinases: MAPK, PAK1, PKC beta/zeta, MEKK3 and CDKs. Accordingly, we demonstrated in FACS analyses and in immunofluorescence microscopy the high intracellular expression of PKCzeta in AML cells and MEKK3 as well PAK1 in mPBSCs. Conclusions: Our findings show that G-CSF stimulates Stathmin expression in mPBSCs and plays a key role in migration into peripheral blood. Furthermore, we show the different expression of kinases acting on stathmin in mPBSCs and AML cells. Consequently, stathmin and its relevant kinases promise to become a future target in therapies of malignant processes. No significant financial relationships to disclose.


2021 ◽  
Author(s):  
Rashmi Bhardwaj ◽  
Lalit Kumar ◽  
Deepika Chhabra ◽  
N K Mehra ◽  
Atul Sharma ◽  
...  

Abstract Fetal liver hematopoietic stem cells because of their proliferative potential have been considered appropriate for management of aplastic anemia. Bone marrow recovery was possible in some cases; engraftment potential of these cells however, was unsatisfactory, possibly due to the availability of smaller number of these cells from a single fetus. Present study was undertaken to see if fetal liver hematopoietic stem cells could be expanded in vitro. Mononuclear cells were isolated and hematopoietic stem cells were identified and analyzed by cell surface marker CD34. CD34+ cells were separated by magnetic cell sorting positive selection method using flow cytometry. Hematopoietic stem cells (CD34+) were cultured by using 5 cytokines, stem cell factor, granulocyte macrophages-colony stimulating factor, interlukin-6, Fms-related tyrosine kinase 3 and erythropoietin, in 4 different combinations along with supplements, in serum free culture media for 21 days. Cell viability continued to be greater than 90% throughout 21 days of culture. The cells expanded best in combination of media, supplements and 4 cytokines, namely SCF, Flt-3, IL6 and Epo to yield large number of total (CD34+ & CD34-) cells. Even though the total number of nucleated cells increased in culture significantly, levels of CD34 antigen expression declined steadily over this period.


Blood ◽  
2003 ◽  
Vol 101 (6) ◽  
pp. 2215-2218 ◽  
Author(s):  
Masahiro Masuya ◽  
Christopher J. Drake ◽  
Paul A. Fleming ◽  
Christopher M. Reilly ◽  
Haiqun Zeng ◽  
...  

It was recently reported that crude bone marrow cells have the ability to differentiate into glomerular mesangial cells. However, the exact nature of the engrafting cells in the bone marrow was not known. We tested the hypothesis that hematopoietic stem cells are capable of reconstituting the mesangial cells by transplanting a clonal population of cells derived from a single stem cell. We cultured Lin−, Sca-1+, c-kit+, CD34− bone marrow cells from transgenic enhanced green fluorescent protein (EGFP) mice (C57BL/6-Ly-5.2 background) individually for 1 week in the presence of interleukin-11 and steel factor. We then transplanted viable clones individually into lethally irradiated C57BL/6-Ly-5.1 mice. Kidneys from 5 recipient mice showing high levels (60%-90%) of multilineage hematopoietic reconstitution were examined 2 to 6 months later, using differential interference contrast and epifluorescence microscopy. EGFP+ cells with a morphology characteristic of mesangial cells were evident within the glomeruli. Transplantation of 100 noncultured Lin−, Sca-1+, c-kit+, CD34− bone marrow cells also generated mesangial cells. Cultured EGFP+ glomerular cells from recipient mice contracted in response to angiotensin II. EGFP+ mesangial cells seen in male-to-male transplants revealed only one Y-chromosome. These data demonstrate that a single hematopoietic stem cell is capable of differentiating into glomerular mesangial cells and that the process does not involve cell fusion.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 396-396
Author(s):  
Mehrdad Abedi ◽  
Deborah A. Greer ◽  
Bethany M. Foster ◽  
Gerald A. Colvin ◽  
Delia A. Demers ◽  
...  

Abstract We have previously shown robust conversion of marrow cells to skeletal muscle (up to 12 percent) in a regenerative environment. We have also recently shown the presence of marrow derived muscle colonies. Evaluation of GFP fluorescent intensity of fibers in individual colonies and comparing them with the single fibers outside the colonies strongly suggests a clonal origin for these colonies. Cotransplantation of marrow cells from Yellow and Cyan Fluorescent Protein transgenic mice also supported this theory. We studied the type of cells responsible for marrow to muscle conversion in an immunocompetent and an immunocomprosied model of marrow transplant. The choice of two models was to clarify any bias related to immunologic reactivity of GFP positive cells that has been well documented in the literature. In the first model, different population of GFP positive marrow cells were transplanted intravenously, after sublethal doses of radiation into C57BL/6 mice. One week after transplant their tibialis anterior muscle were injured with cardiotoxin and their muscles were evaluated for GFP positive, dystrophin positive and CD45 negative muscle fibers. In the second model, different population of GFP positive marrow cells were injected directly into tibilias anterior of Bege/Scid mice one day after cardiotoxin injury. Similar to the first model, GFP positive muscle fibers were evaluated 4 weeks post injury. For each population of marrow cells 3 to 6 animals were transplant with cells either positive or negative for the specific surface markers. Our data demonstrates that conversion potential of marrow cells to skeletal muscle fibers is highly enriched in a CD45 positive, c-Kit positive, Flk-2 positive and Sca-1 positive population. Furthermore, CD34 negative, CXCR4 negative and lineage negative cells have higher potential to participate in marrow to muscle conversion. This indicates that under the conditions of our experiments, mesenchymal stem cells are not the origin of marrow derived GFP muscle fiber. To follow the fate of marrow cells homed into muscle we transplanted GFP marrow cells into radiated C57BL/6 mice and at different timepoints before and after injury, harvested tibialis anterior muscle of these animals. Isolation of mononuclear cells from these samples revealed a new GFP+ CD45 negative population which was very rare at 1 week but more abundant at 4 weeks after transplant. Real time PCR analysis of these cells showed multiple stem cells markers including Sca-1, endoglin, c-Kit and CD34. They did not express any RNA markers for satellite cells or myoblasts (pax 7, MyoD, Myf 5) or hematopoietic cells (CD45, CD11b). We next injured GFP transplanted animals with cardiotoxin and isolated the same CD45-GFP+ population 3 days after injury. Real time PCR analysis at this point showed RNA activity for myoblast including Myf5, MyoD and desmin. Immunofluorescent staining of Lin-, Sca+ cKit+ marrow cells from GFP transgenic mice injected into cardiotoxin injured tibialis anterior muscle of Bege Scid mice confirmed our RT-PCR data and showed the presence of MyoD, Myf5, Myogenin and desmin in donor derived cells in recipient muscle. These data suggests that marrow derived mononuclear cells with hematopoietic stem cell phenotype in the recipient muscle lose their hematopoietic characteristics and obtain myoblast phenomenon after being exposed to injury.


Blood ◽  
2002 ◽  
Vol 100 (13) ◽  
pp. 4391-4400 ◽  
Author(s):  
Wanda Piacibello ◽  
Stefania Bruno ◽  
Fiorella Sanavio ◽  
Sara Droetto ◽  
Monica Gunetti ◽  
...  

The ability of advanced-generation lentiviral vectors to transfer the green fluorescent protein (GFP) gene into human hematopoietic stem cells (HSCs) was studied in culture conditions that allowed expansion of transplantable human HSCs. Following 96 hours' exposure to flt3/flk2 ligand (FL), thrombopoietin (TPO), stem cell factor (SCF), and interleukin-6 (IL-6) and overnight incubation with vector particles, cord blood (CB) CD34+ cells were further cultured for up to 4 weeks. CD34+ cell expansion was similar for both transduced and control cells. Transduction efficiency of nonobese diabetic/severe combined immunodeficient (NOD/SCID) repopulating cells (SRCs) was assessed by transplants into NOD/SCID mice. Mice that received transplants of transduced week 1 and week 4 expanded cells showed higher levels of human engraftment than mice receiving transplants of transduced nonexpanded cells (with transplants of 1 × 105 CD34+ cells, the percentages of CD45+ cells were 20.5 ± 4.5 [week 1, expanded] and 27.2 ± 8.2 [week 4, expanded] vs 11.7 ± 2.5 [nonexpanded]; n = 5). The GFP+/CD45+ cell fraction was similar in all cases (12.5% ± 2.9% and 12.2% ± 2.7% vs 12.7% ± 2.1%). Engraftment was multilineage, with GFP+/lineage+ cells. Clonality analysis performed on the bone marrow of mice receiving transduced and week 4 expanded cells suggested that more than one integrant likely contributed to the engraftment of GFP-expressing cells. Serial transplantations were performed with transduced week 4 expanded CB cells. Secondary engraftment levels were 10.7% ± 4.3% (n = 12); 19.7% ± 6.2% of human cells were GFP+. In tertiary transplants the percentage of CD45+ cells was lower (4.3% ± 1.7%; n = 10); 14.8% ± 5.9% of human cells were GFP+, and human engraftment was multilineage. These results show that lentiviral vectors efficiently transduce HSCs, which can undergo expansion and maintain proliferation and self-renewal ability.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 14-15
Author(s):  
Muhammad Elnaggar ◽  
Igor Pavlovski ◽  
Anjud Al-Mohannadi ◽  
Chiara Cugno ◽  
Jean-Charles Grivel ◽  
...  

Introduction UM171, a novel compound with hematopoietic stem cells (HPSCs) self-renewal properties has shown great potential for expanding CD34+ HPSCs while preserving their stemness and increasing their engrafting abilities. The compound has been increasingly used for the purpose of CD34+ HPSCs expansion, both in pre-clinical research and in clinical applications. Its use is particularly important in the gene therapy (GT) and bone marrow transplantation (BMT) fields, especially when CD34+ expansion is needed to ensure the success of the therapy. Recent reports have demonstrated UM171efficacy to expand enriched CD34+ HPSCs from peripheral blood (PB) in osteopetrosis patients, and after mobilization in lymphoma patients. Here, we demonstrate the feasibility of exploiting the UM171 molecule to directly expand CD34+ HPSCs in bulk PB mononuclear cells (MNCs) without the need of either HPSCs mobilization or CD34+ enrichment. Material and Methods Our protocol entails culturing of bulk MNCs directly after Ficoll gradient separation in IMDM media supplemented with 10% FBS, and HPSCs stemness sparing cytokines, used in clinical suitable concentrations: SCF 300 ng/ml, TPO 100 ng/ml, Flt3-L 300 ng/ml and IL3 60 ng/ml +/- 38 nM UM171 for 20 days. Briefly, quantities of 7.5± 0.5 ml of blood from 2 healthy donors were gradient-separated by Ficoll and yielded a total of 6.6±0.2X10^6 MNCs. Cells were divided in two conditions/donor (Cytokines +/- UM171) and 3.3±0.14X10^6 cell were seeded in 6 well plates at concentration of 1.1X10^6/ml in a total volume of 3 ml. Half of medium was changed every 5 days. Cell count and phenotype of HPSCs were periodically evaluated to determine the optimal expansion window. Cells were manually counted, and phenotype was measured through Symphony A5 flow-cytometry with this antibody panel: Zombie viability dye, CD45, CD34, and in the last timepoint (day 20) CD11b as an exclusion marker for myeloid differentiation. Results UM171 treatment efficiently achieved direct expansion of CD34+ HPSCs in bulk PB MNCs, reaching a peak expansion at day 12 with CD34+ Cells representing 3.9±0.3% of cells (mean±SD of live cells) compared to 0.08±0.04% at baseline (48.75x increase). This increase in CD34+ cell concentration was also partially due to the decline of total PB MNCs cell count during culture time (declined up to 90%). Despite the overall cell count decrease, the CD34+ HPSCs population had an absolute number increase of 6.5±0.916 times with respect to the initial count, peaking at day 12 with an absolute number of 19600±1344 cells compared to 2985±1466 cells at baseline. If projected to a larger scale, this yield proves the feasibility of obtaining of ∼20X10^6 CD34+ HPSCs from a buffy coat of 350 ml blood donation. This amount would be adequate for a pediatric HSCT, and very handy for multiple experiments with CD34+ HPSCs. Conclusion In conclusion, we have provided a proof of concept on the feasibility to directly expand CD34+ HPSCs in bulk PB MNCs. Further characterization of the expanded CD34+ will be needed to ensure stemness, plasticity, and engrafting ability. The advantage of expanding HPSCs in bulk is to preserve the rare naïve CD34+ population that can be lost during the immunoselection process and to take advantage of the myeloid niche, to further enhance the expansion along with UM171. This expansion approach can be performed at larger scale on the buffy coats often dismissed in blood banks to provide considerable number of CD34+ cells for either banking, research or future clinical application. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2000 ◽  
Vol 96 (12) ◽  
pp. 3725-3733 ◽  
Author(s):  
Niels-Bjarne Woods ◽  
Cecilia Fahlman ◽  
Hanna Mikkola ◽  
Isao Hamaguchi ◽  
Karin Olsson ◽  
...  

Abstract The ability of lentiviral vectors to transfer genes into human hematopoietic stem cells was studied, using a human immunodeficiency virus 1 (HIV-1)–derived vector expressing the green fluorescence protein (GFP) downstream of the phosphoglycerate kinase (PGK) promoter and pseudotyped with the G protein of vesicular stomatitis virus (VSV). High-efficiency transduction of human cord blood CD34+cells was achieved after overnight incubation with vector particles. Sixteen to 28 percent of individual colony-forming units granulocyte-macrophage (CFU-GM) colonies derived from cord blood CD34+ cells were positive by polymerase chain reaction (PCR) for the GFP gene. The transduction efficiency of SCID-repopulating cells (SRC) within the cord blood CD34+population was assessed by serial transplantation into nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice. When 400 000 cord blood CD34+ cells were transplanted into primary recipients, all primary and secondary recipients contained and expressed the transgene. Over 50% of CFU-GM colonies derived from the bone marrow of these primary and secondary recipients contained the vector on average as determined by PCR. Transplantation of transduced cells in limiting dilution generated GFP+ lymphoid and myeloid progeny cells that may have arisen from a single SRC. Inverse PCR analysis was used to amplify vector-chromosomal junctional fragments in colonies derived from SRC and confirmed that the vector was integrated. These results show that lentiviral vectors can efficiently transduce very primitive human hematopoietic progenitor and stem cells.


Blood ◽  
2000 ◽  
Vol 96 (6) ◽  
pp. 2125-2133 ◽  
Author(s):  
Robert W. Storms ◽  
Margaret A. Goodell ◽  
Alan Fisher ◽  
Richard C. Mulligan ◽  
Clay Smith

Abstract A novel Hoechst 33342 dye efflux assay was recently developed that identifies a population of hematopoietic cells termed side population (SP) cells. In the bone marrow of multiple species, including mice and primates, the SP is composed primarily of CD34−cells, yet has many of the functional properties of hematopoietic stem cells (HSCs). This report characterizes SP cells from human umbilical cord blood (UCB). The SP in unfractionated UCB was enriched for CD34+ cells but also contained a large population of CD34− cells, many of which were mature lymphocytes. SP cells isolated from UCB that had been depleted of lineage-committed cells (Lin− UCB) contained CD34+ and CD34− cells in approximately equivalent proportions. Similar to previous descriptions of human HSCs, the CD34+Lin− SP cells were CD38dimHLA-DRdimThy-1dimCD45RA−CD71−and were enriched for myelo-erythroid precursors. In contrast, the CD34−Lin− SP cells were CD38−HLA-DR−Thy-1−CD71−and failed to generate myelo-erythroid progeny in vitro. The majority of these cells were CD7+CD11b+CD45RA+, as might be expected of early lymphoid cells, but did not express other lymphoid markers. The CD7+CD34−Lin− UCB SP cells did not proliferate in simple suspension cultures but did differentiate into natural killer cells when cultured on stroma with various cytokines. In conclusion, the human Lin− UCB SP contains both CD34+ multipotential stem cells and a novel CD7+CD34−Lin− lymphoid progenitor. This observation adds to the growing body of evidence that CD34− progenitors exist in humans.


Sign in / Sign up

Export Citation Format

Share Document