Engraftment potential of hematopoietic stem cells (HSCs) is likely to be dependent on several factors including expression of certain adhesion molecules (AMs) and degree of mitotic quiescence. The authors investigated the functional properties and engraftment potential of Sca-1+lin cells subfractionated on the basis of expression, or lack thereof, of CD11a, CD43, CD49d, CD49e, or CD62L and correlated that expression with cell cycle status and proliferative potential of engrafting fractions. Donor-derived chimerism in mice receiving CD49e+ or CD43+ Sca-1+lin cells was greater than that in mice receiving cells lacking these 2 markers, while Sca-1+lin cells positive for CD11a and CD62L and bright for CD49d expression mediated minimal engraftment. AM phenotypes enriched for engraftment potential contained the majority of high proliferative potential–colony forming cells, low proliferative potential–colony forming cells, and cells providing rapid in vitro expansion. Cell cycle analysis of AM subpopulations revealed that, regardless of their bone marrow repopulating potential, Sca-1+lin AM cells contained a higher percentage of cells in G0/G1 than their AM+ counterparts. Interestingly, engrafting phenotypes, regardless of the status of their AM expression, were quicker to exit G0/G1 following in vitro cytokine stimulation than their opposing phenotypes. When engrafting phenotypes of Sca-1+lin AM+ or AMcells were further fractionated by Hoechst 33342 into G0/G1 or S/G2+M, cells providing long-term engraftment were predominantly contained within the quiescent fraction. These results define a theoretical phenotype of a Sca-1+lin engrafting cell as one that is mitotically quiescent, CD43+, CD49e+, CD11a, CD49ddim, and CD62L. Furthermore, these data suggest that kinetics of in vitro proliferation may be a good predictor of engraftment potential of candidate populations of HSCs.

The ability of hematopoietic stem cells (HSCs) to reconstitute normal bone marrow (BM) hematopoiesis following transplantation into suitable recipients relies on the potential of these cells to home to and anchor within the BM microenvironment. Homing is an intricate process by which HSCs, through interactions between adhesion molecules (AMs) and their counter-receptors expressed on BM endothelium, migrate through endothelial cells and into the stromal cell microenvironment. Within the BM microenvironment, the homing process continues as HSCs, again through interactions between AMs and cognant ligands, specifically anchor within appropriate BM niches and begin the process of hematopoiesis. Mobilization of HSCs from their BM niches into the periphery following administration of growth factors or chemotherapeutic agents is likely to involve sequential loss or alterations in adhesive interactions between HSCs and stromal cells, and then endothelium, with final release into the periphery. Although data implicating certain AMs in various stages of stem cell homing and egress from the BM are beginning to accumulate,1-7 much remains to be learned regarding the relationship between AMs and stem cell trafficking, and how this process affects stem cell engraftment and long-term hematopoiesis in transplanted recipients.

While AMs are likely to direct the trafficking of HSCs within the BM microenvironment and periphery, the position of these cells in specific phases of cell cycle is believed to dictate the hematopoietic potential of HSCs.8 A large body of evidence in both in vitro and in vivo systems supports the notion that mitotic quiescence is a fundamental characteristic of HSCs and is essential for the preservation of primitive hematopoietic function. Recent reports in the murine,9 feline,10 and human11systems suggest that the cell cycle of HSCs may be relatively long and that these cells may display reduced engraftment potential at the time of active cell division.12-14 On the other hand, few reports seem to indicate that cells capable of long-term engraftment cycle quickly following transplantation.15These data pose the question of whether changes in AM status, concurrent with entry of these cells into active cell division, influence the engraftment potential of cycling HSCs. We and others have begun to investigate relationships between cell cycle progression and expression or function of AMs on primitive hematopoietic progenitor cells (HPCs). Some reports recently demonstrated an increased expression of certain AMs16-19 and, in some cases, increased adhesion17 of primitive HPCs in active phases of cell cycle. These data, which clearly establish a link between cell cycle status and AM repertoire, suggest the possible existence of regulatory control mechanisms between expression or function of AMs and cell cycle position of HSCs, which may in turn have an impact on the engraftment potential of these cells.

In the present study, we examined the contribution of 6 AMs to the homing and long-term engraftment potential of primitive HPCs using an in vivo murine BM transplantation model. Results of these studies imply a theoretical phenotype of a Sca-1+linengrafting cell as expressing high levels of CD49e and CD43, and low levels of CD11a, CD49d, and CD62L. Cells within this AM-defined phenotype were determined to reside in G0/G1; however, their proliferative response to in vitro cytokine stimulation was rapid and proved to be a good predictor of long-term engraftment potential, as did the content of high proliferative potential–colony forming cells (HPP-CFCs) and low proliferative potential-colony forming cells (LPP-CFCs). Whether this AM phenotype imparts to Sca-1+lin cells more efficient homing and/or anchorage within the BM microenvironment, or enriches for HSCs within Sca-1+lin cells, remains to be determined.

Mice

C57BL/6 female mice (Jackson Laboratories, Bar Harbor, ME) were purchased at 8 to 10 weeks of age and allowed to acclimate for 1 to 2 weeks prior to being used in these studies. C57BL/6 mice are Hbbs/Hbbs (hemoglobin single), are glucose phosphatase isoenzyme type Gpi-1b/Gpi-1b, and express the CD45.2 allotype. Congenic BM donors were of 2 different strains: (1) B6 mice, which are Hbbd/Hbbd(hemoglobin diffuse) and Gpi-1a/Gpi-1a(B6.Gpi-1a) and (2) B6.SJL-PtrcaPep3b/BoyJ mice (B6.BoyJ), which express the CD45.1 allotype. Congenic donor mice were maintained in our breeding colony and used between 8 to 12 weeks of age. These studies were approved by the Indiana University School of Medicine Institutional Animal Care and Use Committee.

Flow cytometric cell sorting and analysis of Sca-1+lin cells expressing one or more AMs

Sca-1+lin cells were isolated from C57BL/6 mice as previously described.20 Briefly, low-density (1.077 g/mL or less) BM mononuclear cells were stained with phycoerythrin (PE)–conjugated Sca-1 and fluorescein isothiocyanate (FITC)–conjugated CD3 and CD45R/B220, and sorted by means of a FACStarplus flow cytometer (Becton Dickinson Immunocytometry Systems, San Jose, CA) to yield Sca-1+lin cells. To isolate AM subfractions of Sca-1+lin cells, mononuclear cells were stained with Sca-1 and lineage monoclonal antibodies as above, along with the biotinylated antibody recognizing one of the following: CD11a (leukocyte-function–associated antigen 1 [LFA-1], clone 2D7), CD43 (Leukosialin, clone S7), CD44 (homing-associated cell AM[HCAM], clone IM7), CD49d (very late antigen 4 [VLA-4], clone 9C10), CD49e (VLA-5, clone 5H10-27; MFR5), or CD62L (L-selectin, clone MEL-14). All antibodies used in this study were obtained from Pharmingen (San Diego, CA). AM antibodies were visualized by streptavidin-APC (allophycocyanin) (Molecular Probes, Eugene, OR). Cells were sorted on a FACStarplus flow cytometer as follows (Figure1): A primary light scatter gate was constructed and used to visualize FITC and PE fluorescence of stained cells. A second gate was constructed to contain PE+ and FITC cells (Sca-1+lincells, Figure 1A), and the APC fluorescence of these gated cells was examined in a single-parameter histogram (Figure 1B-C). The fluorescence of a sample stained with Sca-1–PE, CD3-FITC, B220-FITC, and nonspecific biotinylated antibodies followed by streptavidin-APC was used to determine background APC fluorescence of Sca-1+lin cells. On the basis of this determination, sort regions were created within the APC histogram to isolate Sca-1+lin cells that were positive or negative for CD11a, CD43, CD49e, or CD62L (referred to as Sca-1+lin AM+ or Sca-1+lin AM cells hereafter) (Figure 1B). To ensure adequate separation, cells were gated to include the upper and lower 30% to 40% of the APC histogram, with the middle 30% to 40% of the cells discarded. In the cases of CD44 and CD49d, where nearly 100% of Sca-1+lin cells expressed these 2 markers (Table 1), the brightest and dimmest 30% to 40% of Sca-1+lin cells were sorted and referred to as “bright” or “dim,” respectively (Figure 1C). In some cases, Sca-1+lin cells were sorted while ignoring the signal from the AM antibody, to obtain a control group of Sca-1+lin cells. These cells were used to test for in vivo blocking activity of the AM antibody used for cell sorting. Cells were maintained at 4°C throughout cell sorting and were collected into Iscove's Modified Dulbecco's Medium (IMDM) containing 20% fetal bovine serum (FBS) (Hyclone, Logan, UT).

Fig. 1.

Flow cytometric cell sorting of Sca-1+lin AM+ or AMcells.

Low-density murine BM cells were stained with Sca-1–PE, FITC-conjugated CD3 and B220 (lin), and biotinylated antibodies recognizing either CD11a, CD43, CD44, CD49d, CD49e, or CD62L, followed by the addition of streptavidin-APC. A primary light scatter gate was constructed and used to visualize PE and FITC fluorescence (panel A). A second gate containing PE+ and FITC cells (Sca-1+lin cells, panel A) was drawn, and the APC fluorescence of these gated Sca-1+lin cells were visualized in a single-parameter histogram (filled histograms, panels B and C). Background APC fluorescence of Sca-1+lincells (open histogram, panels B and C) was determined by staining a sample with Sca-1–PE, CD3-FITC, B220-FITC, and nonspecific biotinylated antibodies followed by streptavidin-APC. Sort regions, depicted by the vertical lines and arrows in panels B and C, were created to isolate the upper and lower 30% to 40% of Sca-1+lin cells expressing the particular AM. For CD11a, CD43, CD49e, and CD62L, a distinct “positive” and “negative” AM fraction could be isolated (panel B), but in the cases of CD44 and CD49d, where nearly 100% of Sca-1+lin cells expressed these markers, the brightest and dimmest 30% to 40% of Sca-1+lin cells were sorted and referred to as “bright” and “dim,” respectively. To test for in vivo blocking activity of AM antibodies, a group of Sca-1+lin cells were sorted from each AM group while the APC signal from the AM antibody was ignored. Sort regions are shown for CD49e and CD49d as representative examples. To isolate Sca-1+lin cells on the basis of their expression of both CD49d and CD49e, Sca-1+lin cells were sorted to purity, panel A. Since these cells were FITC, they were stained with FITC-labeled CD49d and biotinylated CD49e, followed by streptavidin-APC (panel D). Based on nonspecific background fluorescence, denoted by dotted box in lower left of panel D, 4 sort regions were created as follows: (1) APC FITC+ cells were gated and sorted as CD49e CD49dbright; (2) APC+ FITC+ cells were sorted as CD49e+ CD49dbright; (3) APCFITC cells were sorted as CD49eCD49ddim; and (4) APC+ FITC cells were sorted as CD49e+ CD49ddim (panel D).

Fig. 1.

Flow cytometric cell sorting of Sca-1+lin AM+ or AMcells.

Low-density murine BM cells were stained with Sca-1–PE, FITC-conjugated CD3 and B220 (lin), and biotinylated antibodies recognizing either CD11a, CD43, CD44, CD49d, CD49e, or CD62L, followed by the addition of streptavidin-APC. A primary light scatter gate was constructed and used to visualize PE and FITC fluorescence (panel A). A second gate containing PE+ and FITC cells (Sca-1+lin cells, panel A) was drawn, and the APC fluorescence of these gated Sca-1+lin cells were visualized in a single-parameter histogram (filled histograms, panels B and C). Background APC fluorescence of Sca-1+lincells (open histogram, panels B and C) was determined by staining a sample with Sca-1–PE, CD3-FITC, B220-FITC, and nonspecific biotinylated antibodies followed by streptavidin-APC. Sort regions, depicted by the vertical lines and arrows in panels B and C, were created to isolate the upper and lower 30% to 40% of Sca-1+lin cells expressing the particular AM. For CD11a, CD43, CD49e, and CD62L, a distinct “positive” and “negative” AM fraction could be isolated (panel B), but in the cases of CD44 and CD49d, where nearly 100% of Sca-1+lin cells expressed these markers, the brightest and dimmest 30% to 40% of Sca-1+lin cells were sorted and referred to as “bright” and “dim,” respectively. To test for in vivo blocking activity of AM antibodies, a group of Sca-1+lin cells were sorted from each AM group while the APC signal from the AM antibody was ignored. Sort regions are shown for CD49e and CD49d as representative examples. To isolate Sca-1+lin cells on the basis of their expression of both CD49d and CD49e, Sca-1+lin cells were sorted to purity, panel A. Since these cells were FITC, they were stained with FITC-labeled CD49d and biotinylated CD49e, followed by streptavidin-APC (panel D). Based on nonspecific background fluorescence, denoted by dotted box in lower left of panel D, 4 sort regions were created as follows: (1) APC FITC+ cells were gated and sorted as CD49e CD49dbright; (2) APC+ FITC+ cells were sorted as CD49e+ CD49dbright; (3) APCFITC cells were sorted as CD49eCD49ddim; and (4) APC+ FITC cells were sorted as CD49e+ CD49ddim (panel D).

Close modal
Table 1.

Expression of AMs on Sca-1+lincells

Adhesion molecule CD designationCD11aCD43CD44CD49dCD49eCD62L
Common name LFA-1 Leukosialin HCAM VLA-4 VLA-5 L-selectin 
Percentage of positive Sca-1+lincells* 77 ± 7.4 83 ± 6.6 99 ± 0.6 94 ± 2.8 63 ± 9.0 46 ± 5.2 
Adhesion molecule CD designationCD11aCD43CD44CD49dCD49eCD62L
Common name LFA-1 Leukosialin HCAM VLA-4 VLA-5 L-selectin 
Percentage of positive Sca-1+lincells* 77 ± 7.4 83 ± 6.6 99 ± 0.6 94 ± 2.8 63 ± 9.0 46 ± 5.2 

AM indicates adhesion molecule; LFA-1, leukocyte-function–associated antigen 1, HCAM, homing-associated cell AM, and VLA, very late antigen

*

Individual expression of 6 AMs on fresh Sca-1+lin cells was determined by flow cytometry as described in “Materials and methods.” Data represent the mean percentage of positive Sca-1+lincells ± SEM; n = 5 to 8 for each AM.

To isolate Sca-1±lin cells on the basis of their expression of both CD49d and CD49e, Sca-1±lin cells were isolated with the use of Sca-1–PE, CD3-FITC, and B220-FITC as described above. These cells, which are PE+ and FITC, were then stained with biotinylated CD49e and FITC-labeled CD49d (Figure 1D). CD49e was developed with streptavidin-APC, and the cells were washed and resuspended for flow cytometric cell sorting on a FACStarplus flow cytometer. Stained Sca-1±lin cells contained within a primary light scatter gate were examined for their FITC and APC fluorescence in a dual parameter dot plot, and 4 sort regions (Figure 1D), each containing 10% to 20% of the Sca-1±lincells, were created as follows: (1) APCFITC± cells were gated and sorted as CD49eCD49dbright; (2) APC± FITC± cells were sorted as CD49e± CD49dbright; (3) APC FITC cells were sorted as CD49e CD49ddim; and (4) APC±FITC cells were sorted as CD49e±CD49ddim. Cells were maintained at 4°C throughout cell sorting and were collected into IMDM containing 20% FBS.

To analyze the dual expression of AM on Sca-1+lin cells, groups of Sca-1+lin AM+ or AMcells were isolated as described above with the use of Sca-1–FITC, CD3-PE, B220-PE, and biotinylated-AM antibodies developed with streptavidin-APC. The resulting groups of Sca-1+lin cells, which are negative for PE fluorescence, were then stained with the PE-conjugate of the remaining 5 AM antibodies not used in the primary sort (ie, Sca-1+lin CD49e+ and Sca-1+ lin CD49e cells would be subsequently stained with CD11a, CD43, CD44, CD49d, and CD62L). The PE fluorescence of these stained cells was examined on a FACScan (Becton Dickinson), and the percentage of each primary adhesion phenotype expressing each of the other AMs was calculated on the basis of background fluorescence of cells stained with PE-conjugated nonspecific myeloma proteins. In every case, background PE fluorescence of Sca-1+lin AM+ or AM cells was less than 5% of that observed after staining these cells with 1 of the 5 PE-conjugated AM antibodies.

Separation of cell cycle subfractions of Sca-1+lin AM+ or AM cells

Sca-1+linAM+ or AM cells enriched for engraftment potential were isolated as described above, resuspended in Hoechst buffer (Hanks' balanced salt solution [Biowhittaker, Walkersville, MD], 20 mmol/L HEPES [Biowhittaker], 1 g/L glucose, and 10% fetal calf serum [FCS]), and stained with Hoechst 33342 (Molecular Probes) at 10 μmol/L for 45 minutes at 37°C as previously described.21 To limit dye efflux via the MDR-1 pump, 100μmol/L verapamil (Sigma Chemical Co, St Louis, MO) was added to the staining buffer.22 Cells were washed and resuspended in Hoechst buffer plus verapamil for cell sorting. Cells falling within a primary light scatter gate and containing 2n DNA were gated and sorted as G0/G1. Cells containing between 2n and 4n DNA were gated and sorted as S/G2+M. Hypodiploid events, when present, were excluded from sort windows. When quantities were sufficient, a small portion of sorted cells were subjected to postsort analyses, either by examining Hoechst fluorescence or by propidium iodide staining, as previously described.23 

HPP-CFC and LPP-CFC assay

Between 0.8 × 103 and 1.5 × 103Sca-1+lin cells or AM subfractions were suspended in triplicate in 1 mL double-layer agar cultures and assayed for HPP-CFCs and LPP-CFCs as previously described.24Cultures were incubated in a 100%-humidified 5% O2, 10% CO2, and 85% N2 environment. Recombinant hematopoietic growth factors were used as follows: 200 U/mL murine interleukin-3 (mIL3), 1000 U/mL mIL1-α, 50 ng/mL murine stem cell factor (mSCF), 25 ng/mL murine granulocyte macrophage–colony stimulating factor (mGM-CSF) (all from PeproTech, Rocky Hill, NJ), and 1600 U/mL human macrophage–CSF-1 (hM-CSF, Genetics Institute, Camden, MA). On day 14, colonies larger than 0.5 mm were scored as HPP-CFC and those smaller than 0.5mm as LPP-CFC.

Transplantation protocol

C57BL/6 female recipients between 10 and 12 weeks of age were lethally irradiated (split dose of 700 centrigrays [cGy] followed by 350 cGy 3 to 4 hours later) from a 137Cs gamma irradiator (GammaCell 40; Nordion International, Kanata, Ontario, Canada). Mice were transplanted via tail vein injections 3 to 6 hours later with 0.25 × 103 to 1.0 × 103 donor B6.Gpi-1a or B6.BoyJ cells, along with 0.3 × 105 to 1.0 × 105 competitor cells (low-density BM cells of C57BL/6 origin), as described in Figures. Recipient mice were bled from the tail vein monthly until 6 or 8 months posttransplantation for analysis of donor-derived hematopoiesis.

Determination of donor chimerism in transplanted recipients

Chimerism in C57BL/6 mice, when B6.Gpi-1amice were used as donors, was determined by analyzing the percentage of donor-derived hemoglobin or Gpi-1 in peripheral blood obtained from tail veins as previously described.25 To examine multilineage engraftment in these mice, whole-blood samples obtained from the retro-orbital sinus were stained separately with FITC-conjugated CD45R/B220, or FITC-conjugated CD3, and PE-conjugated Gr-1, lysed, and then isolated by flow cytometric cell sorting to yield B lymphocytes, T lymphocytes, and granulocytes, respectively. These purified lineages were then analyzed for Gpi-1 content.25 

Chimerism in C57BL/6 mice receiving donor cells of B6 BoyJ origin was determined by means of flow cytometry to calculate the percentage of CD45.2 peripheral blood cells. Whole-blood samples obtained from tail veins were lysed and stained with PE-conjugated CD45 and FITC-conjugated CD45.2. CD45+ cells were gated and examined for the percentage of CD45.2cells (cells of B6.BoyJ origin). Multilineage engraftment in these mice was determined by staining with each of the 3 following combinations of antibodies, CD45.2-FITC and Gr-1–PE, CD45.1-PE and CD3-FITC, and CD45.1-PE and B220-FITC, and analyzing the percentages of donor-derived granulocytes, T lymphocytes, and B lymphocytes, respectively.

Short-term culture

Cultures of 0.2 × 103 to 4 × 103Sca-1+lin AM+ or AMcells, or cell cycle subfractions of these cells, were initiated in IMDM supplemented with 20% FBS and 2 × 10−5 mol/L 2-mercaptoethanol in an atmosphere of 5% CO2 in 100%-humidified air. Cytokines were delivered on day 0 as follows: 100 ng/mL mSCF (PeproTech), 500 U/mL mIL1α (Genzyme, Cambridge, MA), 100 U/mL mIL3 (Genzyme), 100 ng/mL hIL6, and 50 ng/mL human Flt3 ligand (hFlt3-L). The hIL6 and hFlt3-L were kind gifts from Amgen (Thousand Oaks, CA) and Immunex (Seattle, WA), respectively. Care was taken to keep the cell concentration below 1 × 106cells/mL. Fresh cells or aliquots of cultured cells were removed on days 1 and 2 and stained with propidium iodide for cell cycle analysis23 or incubated for an additional 11 to 14 days, at which time cells were harvested, enumerated, and analyzed by flow cytometry for the percentage of cells expressing Sca-1 with the use of anti–Sca-1–PE.

Statistical analysis

Data are expressed as the mean ± SEM where applicable. Differences between groups were analyzed by means of an unpaired 2-sided t test. A probability value of less than .05 was considered significant. Regression analysis was used to analyze the rate of exit from G0/G1 phases of the cell cycle.

Expression of AMs on Sca-1+lin cells

As a first step in defining AMs important for engraftment of primitive HPCs, we examined the pattern of expression of CD11a, CD43, CD44, CD49d, CD49e, and CD62L on fresh Sca-1+lin cells. As seen in Table 1, expression of these 6 AMs was varied, with nearly 100% of Sca-1+lin cells expressing CD44 and CD49d, and approximately 50% expressing CD49e and CD62L. Expression of CD11a and CD43 was observed on approximately 80% of Sca-1+lin cells.

Primitive and mature progenitor cell content of Sca-1+linAM+ or AM cells

Since HPP-CFCs likely contain some of the most primitive HPCs,26 the HPP-CFC frequency of our 12 groups of Sca-1+lin AM+ or AMcells was determined. Data in Table 2show that CD43 and CD49e were expressed on the majority of HPP-CFCs residing in the Sca-1+lin cell fraction, while Sca-1+lin cells lacking expression of either CD11a or CD62L contained a higher fraction of HPP-CFCs than Sca-1+lin CD11a+ or Sca-1+lin CD62L+ cells (Table 2). LPP-CFCs, progenitors more committed to lineage differentiation than HPP-CFCs, were mostly enriched among the same AM profile as HPP-CFCs (Table 2). To ensure that the AM antibody used for cell sorting did not induce any negative influences on primitive HPC clonogenic activity, Sca-1+lin cells treated with each AM antibody but not sorted on adhesion phenotype were also assayed for HPP- and LPP-CFC content. HPP- and LPP-CFC activity of these antibody-treated cells was not different from that of untreated Sca-1+lin cells (data not shown), indicating that the AM antibodies used for cell sorting did not inhibit in vitro activity of progenitor cells. Fractions enriched for HPP- and LPP-CFC activity also exhibited greater cellular expansion during 11 days of in vitro cytokine-stimulated cell culture (Table 2).

Table 2.

HPP-CFC and LPP-CFC content and proliferative potential of Sca-1+linAM+ or AMcells

Sca-1+LinCD11aCD43CD44CD49dCD49eCD62L
PosNegPosNegBrightDimBrightDimPosNegPosNeg
HPP-CFC* 100 126 58 45 86 71 88 186 59 106 
LPP-CFC* 100 10 115 47 35 55 57 58 148 37 69 
Proliferative potential Nd 440 650 480 360 290 190 200 650 230 15 110 570 
Sca-1+LinCD11aCD43CD44CD49dCD49eCD62L
PosNegPosNegBrightDimBrightDimPosNegPosNeg
HPP-CFC* 100 126 58 45 86 71 88 186 59 106 
LPP-CFC* 100 10 115 47 35 55 57 58 148 37 69 
Proliferative potential Nd 440 650 480 360 290 190 200 650 230 15 110 570 

HPP-CFC indicates high proliferative potential–colony forming cells; and LPP-CFC, low proliferative potential-colony forming cells.

*

HPP-CFC and LPP-CFC data are normalized by assigning a value of 100% to the HPP-CFC and LPP-CFC content of Sca-1+lin cells in each experiment, and adjusting remaining values accordingly. Data are means from 3 separate experiments. For ease of presentation, SEM values are excluded from the table, but range from 0 to 35 for HPP-CFC data, and from 0 to 27 for LPP-CFC data.

Proliferative potential is expressed as fold-increase in total cells from day 0 to day 11. AM+ and AMSca-1+lin cells were cultured in vitro with mSCF, mIL1α, mIL3, hIL6, and hFlt3-L for 11 days. Fold-increase was calculated by dividing the cell number on day 11 by the input number on day 0. Data shown are from 1 of 2 experiments with similar results.

Chimerism in mice transplanted with Sca-1+lin AM+ or AM cells

To determine the engraftment potential of the 12 groups of Sca-1+lin AM+ or AMcells, a competitive repopulation assay in lethally irradiated recipient mice was performed. Chimerism in recipient mice transplanted with either total Sca-1+lin cells or Sca-1+lin AM+ or AMcells was evident 4 weeks posttransplantation and continued to increase until 2 months posttransplantation, at which time chimerism stabilized (data not shown). Figure 2A shows that Sca-1+lin cells expressing CD43 and CD49e appeared to be highly enriched for long-term engraftment potential, as Sca-1+lin cells lacking expression of either of these 2 molecules failed to provide measurable chimerism in recipients at 6 months posttransplantation. Sca-1+lin cells expressing low levels of CD11a, CD49d, and CD62L were superior competitors compared with their counterparts expressing higher levels (Figure 2A). Expression of CD44 did not appear to significantly correlate with enhanced or diminished engraftment potential of Sca-1+lin cells. As indicated by the horizontal bars in Figure 2A, anti-AM antibodies used for cell sorting did not interfere with engraftment of Sca-1+lin cells, as mice transplanted with Sca-1+lin cells treated with each anti-AM antibody exhibited chimerism similar to that of mice receiving untreated Sca-1+lin cells. All groups of Sca-1+linAM+ or AM cells contributed equally to lineage-specific hematopoiesis, as indicated by similar levels of donor-derived chimerism in myeloid (Gr-1+) and lymphoid (CD3+ or B220+) cells (data not shown).

Fig. 2.

Percentage of donor-derived chimerism in transplanted mice.

Percentage of donor-derived chimerism in mice transplanted with Sca-1+lin cells, Sca-1+lin AM+ or AMcells (panel A), or Sca-1+lin cells fractionated with the use of CD49d and CD49e simultaneously (panel B). Lethally irradiated C57BL/6 mice were transplanted with 1 × 103 total Sca-1+lin cells (hatched bar), Sca-1+linAM+ (black bars), or Sca-1+linAM (gray bars), all of B6.Gpi-1a origin, along with 3 × 104 C57BL/6 competitor cells. Chimerism was monitored monthly by hemoglobin analysis of peripheral blood cells. Horizontal black lines represent mean engraftment in mice transplanted with “antibody control” cells (Sca-1+lincells treated with the AM antibody but not sorted for the AM phenotype). Data are expressed in panel A as the mean ± SEM of 3 to 12 mice per group from 1 to 5 separate experiments (analyzed at 6 months posttransplantation), and in panel B as the mean ± SEM of 3 to 5 mice per group in 1 experiment. Trends in engraftment similar to that seen in panel A were obtained in experiments in which B6.BoyJ donor cells were used along with 1 × 105 C57BL/6 competitor cells. *The presence of statistical significance between the AM+ and AM fraction of Sca-1+lin cells; P < .05. **The presence of statistical significance between Sca-1+lin CD49e+CD49ddim cells and each of the other 3 phenotypes at 8 months posttransplantation.

Fig. 2.

Percentage of donor-derived chimerism in transplanted mice.

Percentage of donor-derived chimerism in mice transplanted with Sca-1+lin cells, Sca-1+lin AM+ or AMcells (panel A), or Sca-1+lin cells fractionated with the use of CD49d and CD49e simultaneously (panel B). Lethally irradiated C57BL/6 mice were transplanted with 1 × 103 total Sca-1+lin cells (hatched bar), Sca-1+linAM+ (black bars), or Sca-1+linAM (gray bars), all of B6.Gpi-1a origin, along with 3 × 104 C57BL/6 competitor cells. Chimerism was monitored monthly by hemoglobin analysis of peripheral blood cells. Horizontal black lines represent mean engraftment in mice transplanted with “antibody control” cells (Sca-1+lincells treated with the AM antibody but not sorted for the AM phenotype). Data are expressed in panel A as the mean ± SEM of 3 to 12 mice per group from 1 to 5 separate experiments (analyzed at 6 months posttransplantation), and in panel B as the mean ± SEM of 3 to 5 mice per group in 1 experiment. Trends in engraftment similar to that seen in panel A were obtained in experiments in which B6.BoyJ donor cells were used along with 1 × 105 C57BL/6 competitor cells. *The presence of statistical significance between the AM+ and AM fraction of Sca-1+lin cells; P < .05. **The presence of statistical significance between Sca-1+lin CD49e+CD49ddim cells and each of the other 3 phenotypes at 8 months posttransplantation.

Close modal

The importance of CD49e expression in fractionating Sca-1+lin cells into engrafting phenotypes is illustrated in Figure 2B. With the use of both CD49e and CD49d, Sca-1+lin cells were further fractionated to yield 4 groups of cells (each being either positive or negative for either marker). All 4 groups of cells were analyzed for their engraftment potential in the same competitive repopulation assay described above. Figure 2B shows that chimerism in mice transplanted with Sca-1+lin cells expressing a CD49e+ CD49ddim phenotype was higher than that in mice transplanted with similar cells exhibiting a CD49e+CD49dbright phenotype, while mice receiving either group of Sca-1+lin cells devoid of CD49e expression had undetectable engraftment. These results are consistent with data in Figure 2A, in that Sca-1+lin cells that were CD49e+ or CD49ddim were better competitors than their CD49e or CD49dbright counterparts, respectively. Mice receiving Sca-1+linCD49e+ CD49dbright cells exhibited transient chimerism at 2 months posttransplantation that diminished after several months, suggesting that this phenotype consisted of cells capable of short-term engraftment only. Sca-1+lin cells exposed to CD49e and CD49d antibodies provided 50% ± 8% chimerism in recipients, a figure not statistically different from that provided by untreated Sca-1+lin cells (67.8% ± 11% chimerism, P > 0.1) (Figure 1A). Taken together, these results suggest a theoretical phenotype of a Sca-1+lin engrafting cell as being positive for expression of CD43 and CD49e, but negative or low for expression of CD11a, CD49d, and CD62L.

Analysis of AM expression on engrafting vs nonengrafting phenotypes of Sca-1+lin cells

To better identify AMs potentially important in engraftment, subfractions of Sca-1+lin cells demonstrated as having enriched engraftment potential were surveyed for their repertoire of AM expression in comparison with their nonengrafting counterparts. Figure 3shows that expression of CD11a (panel A), CD49dbright(panel D), and CD62L (panel F) was slightly lower on engrafting than on nonengrafting phenotypes, while expression of CD43 and CD49e was greater on engrafting cells. Interestingly, expression of CD43 (panel B) was more than 4 times greater on engrafting phenotypes defined by CD11a and CD49e, and expression of CD49e (Panel E) was more than 2- and 5-fold greater on engrafting phenotypes defined by CD11a and CD43, respectively; this illustrates once again the importance of CD43 and CD49e in engraftment. Examination of mean channel fluorescence and peak channel fluorescence also revealed high levels of expression of CD43 and CD49e on engrafting phenotypes of Sca-1±lin cells (data not shown). Expression of CD44bright (Figure 3C) was slightly increased on engrafting phenotypes.

Fig. 3.

Expression of AMs on engrafting and nonengrafting phenotypes of Sca-1+lin cells.

Sca-1+lin cells were subfractionated by cell sorting into AM+/AMbright or AM/AMdim groups with the use of CD11a (■), CD43 (♦), CD49d (●), CD49e (○), and CD62L (▪) to give engrafting or nonengrafting phenotypes, designated E and NE, respectively. CD44 was omitted in the primary sort since this marker was not useful in defining engrafting phenotypes (Figure 2). These resulting 10 groups of cells were phenotyped for their expression of other AMs not used in the primary sort (listed at the top of each scatter plot), and the percentage of each engrafting (E) and nonengrafting (NE) phenotype positive for expression of other AMs was calculated after subtracting nonspecific background fluorescence. Each individual data point represents the percentage of positive expression; horizontal bars are mean expression. For example, the open circles in Panel A represent the expression of CD11a on CD49e+ cells (E) and CD49e cells (NE). In panel B, open squares represent expression of CD43 on CD11a cells (E) and CD11a+ cells (NE), also shown in panel G. Since 100% of Sca-1+lin cells express CD44 and CD49d (Table 1), only the proportion of engrafting and nonengrafting phenotypes positive for CD44bright and CD49dbright expression was calculated. The histogram in panel G illustrates the high level of expression of CD43 (x-axis) on Sca-1[+lin CD11a cells (open histogram) relative to CD11a+ cells (filled histogram), while panel H shows the high level of expression of CD43 on Sca-1+lin CD49e+ cells (open histogram) relative to CD49e cells (filled histogram). Data are from 1 of 2 experiments with similar results.

Fig. 3.

Expression of AMs on engrafting and nonengrafting phenotypes of Sca-1+lin cells.

Sca-1+lin cells were subfractionated by cell sorting into AM+/AMbright or AM/AMdim groups with the use of CD11a (■), CD43 (♦), CD49d (●), CD49e (○), and CD62L (▪) to give engrafting or nonengrafting phenotypes, designated E and NE, respectively. CD44 was omitted in the primary sort since this marker was not useful in defining engrafting phenotypes (Figure 2). These resulting 10 groups of cells were phenotyped for their expression of other AMs not used in the primary sort (listed at the top of each scatter plot), and the percentage of each engrafting (E) and nonengrafting (NE) phenotype positive for expression of other AMs was calculated after subtracting nonspecific background fluorescence. Each individual data point represents the percentage of positive expression; horizontal bars are mean expression. For example, the open circles in Panel A represent the expression of CD11a on CD49e+ cells (E) and CD49e cells (NE). In panel B, open squares represent expression of CD43 on CD11a cells (E) and CD11a+ cells (NE), also shown in panel G. Since 100% of Sca-1+lin cells express CD44 and CD49d (Table 1), only the proportion of engrafting and nonengrafting phenotypes positive for CD44bright and CD49dbright expression was calculated. The histogram in panel G illustrates the high level of expression of CD43 (x-axis) on Sca-1[+lin CD11a cells (open histogram) relative to CD11a+ cells (filled histogram), while panel H shows the high level of expression of CD43 on Sca-1+lin CD49e+ cells (open histogram) relative to CD49e cells (filled histogram). Data are from 1 of 2 experiments with similar results.

Close modal

Cell cycle status

Since HSCs are believed to represent relatively quiescent cells, the cell cycle status of the 12 phenotypes of Sca-1+lin AM+ or AMcells was evaluated to determine whether each of the AM subfractions of Sca-1+lin cells enriched for engraftment potential would also be more quiescent than the corresponding nonengrafting phenotype. However, 5 out of 6 Sca-1+lin fractions that were negative or dim for expression of AMs were significantly more quiescent than the AM+ fraction (Figure 4). In the cases of CD49d and CD62L, the engrafting phenotype was more quiescent than the nonengrafting, but in the cases of CD43 and CD49e, the engrafting phenotype contained the higher percentage of cycling cells (Figure 4).

Fig. 4.

Cell cycle status of fresh Sca-1+lin AM+ or AMcells or total Sca-1+lin cells.

Fractions of Sca-1+lin AM+ (black bars), Sca-1+lin AM (gray bars), or total Sca-1+lin cells (horizontal line) were stained with propidium iodide and analyzed on a FACScan flow cytometer for cell cycle status as previously described.23Data are reported as the percentage of cells in G0/G1 phase of the cell cycle (mean ± SEM; n = 6 to 7 for every group). *The presence of statistical significance when the AM fraction is compared with its AM+ counterpart; P < .05.

Fig. 4.

Cell cycle status of fresh Sca-1+lin AM+ or AMcells or total Sca-1+lin cells.

Fractions of Sca-1+lin AM+ (black bars), Sca-1+lin AM (gray bars), or total Sca-1+lin cells (horizontal line) were stained with propidium iodide and analyzed on a FACScan flow cytometer for cell cycle status as previously described.23Data are reported as the percentage of cells in G0/G1 phase of the cell cycle (mean ± SEM; n = 6 to 7 for every group). *The presence of statistical significance when the AM fraction is compared with its AM+ counterpart; P < .05.

Close modal

The cell cycle status of Sca-1+lin cells fractionated with the use of both CD49e and CD49d expression also correlated with the level of expression of these 2 AMs. Sca-1+lin cells lacking expression of both markers were more quiescent (98.6% ± 0.4% in G0/G1) than Sca-1+lincells positive for the expression of both CD49e and CD49d (78.3% ± 2.8% in G0/G1). The degree of quiescence of Sca-1+lin cells lacking expression of one but not both markers fell between that of double-positive and double-negative cells (CD49e+/CD49ddim = 93.2% ± 1.9% G0/G1; CD49e/CD49dbright = 96.0% ± 2.0% G0/G1; n = 2 for all cell cycle analyses).

Cell cycle progression of AM subfractions of Sca-1+lin cells

Rapid in vivo proliferation of HSCs following transplantation has been reported to correlate with long-term hematopoietic function of transplanted cells.27 We have already shown that cellular expansion in vitro (Table 2) correlates with long-term engraftment capability of Sca-1+lin AM+ or AM cells (Figure 2). Differences in proliferative potential between engrafting and nonengrafting phenotypes may be related to the rate at which these cells exit G0/G1 in vitro, as shown in Figure5. AM fractions enriched for engraftment potential, regardless of their relative degree of quiescence on day 0, exited G0/G1 more rapidly than nonengrafting phenotypes (Figure 5). Cell cycle progression of Sca-1+lin cells subfractionated with the use of both CD49e and CD49d also correlated with the engraftment potential of these cells. Sca-1+lin cells most enriched for engraftment potential among these 4 phenotypes (CD49e+/CD49ddim) exited G0/G1 more rapidly (slope = −25) than cells lacking expression of CD49e (slopes = −2 to −11), or Sca-1+lin cells expressing both CD49e and CD49d cells (slope = −16).

Fig. 5.

Exit from G0/G1 phases of cell cycle.

Exit from G0/G1 phases of cell cycle of total Sca-1+lin cells (▴), or engrafting phenotypes (dotted lines) and nonengrafting phenotypes (solid lines) of Sca-1+lin AM+ (▪) and AM (○) cells. Up to 4 × 103 cells/mL were supplemented with 20% FBS, 2 × 10−5 mol/L 2-mercaptoethanol, 100 ng/mL mSCF and hIL6, 500 U/mL mIL1α, 100 U/mL mIL3, and 50 ng/mL hFlt3-L. Fresh cells and aliquots of cultured cells removed on days 1 and 2 were stained with propidium iodide and analyzed for cell cycle status as previously described.23 Data are expressed as the mean ± SEM; n = 2 to 3 for each time point. Slopes of linear regression lines generated from these data are indicated next to the appropriate lines to illustrate relative rates of exit from G0/G1 of the different populations of cells. *The presence of statistical significance in slope of regression line of engrafting phenotype when compared with the slope generated from its nonengrafting counterpart; P < .05.

Fig. 5.

Exit from G0/G1 phases of cell cycle.

Exit from G0/G1 phases of cell cycle of total Sca-1+lin cells (▴), or engrafting phenotypes (dotted lines) and nonengrafting phenotypes (solid lines) of Sca-1+lin AM+ (▪) and AM (○) cells. Up to 4 × 103 cells/mL were supplemented with 20% FBS, 2 × 10−5 mol/L 2-mercaptoethanol, 100 ng/mL mSCF and hIL6, 500 U/mL mIL1α, 100 U/mL mIL3, and 50 ng/mL hFlt3-L. Fresh cells and aliquots of cultured cells removed on days 1 and 2 were stained with propidium iodide and analyzed for cell cycle status as previously described.23 Data are expressed as the mean ± SEM; n = 2 to 3 for each time point. Slopes of linear regression lines generated from these data are indicated next to the appropriate lines to illustrate relative rates of exit from G0/G1 of the different populations of cells. *The presence of statistical significance in slope of regression line of engrafting phenotype when compared with the slope generated from its nonengrafting counterpart; P < .05.

Close modal

Engraftment potential of Sca-1+linAM+ or AM cells subfractionated on the basis of position in cell cycle

The relatively high percentage of cycling cells in engrafting phenotypes defined as CD43+ and CD49e+ of Sca-1+lin cells (Figure 4) led us to question whether quiescent or cycling cells within the AM-defined engrafting phenotypes were contributing to engraftment potential and in vitro hematopoiesis of these cells. To this end, engrafting phenotypes of Sca-1+lin AM+ or AMcells were subfractionated into G0/G1 and S/G2+M fractions with the use of Hoechst 33342 and examined for their engraftment potential. Figure 6shows chimerism in mice transplanted with G0/G1or S/G2 + M fractions of total Sca-1+lin cells, Sca-1+lin CD11a, Sca-1+lin CD49ddim, and Sca-1+lin CD49e+ cells. G0/G1 cells, regardless of which AM was used in fractionation, provided greater levels of chimerism than equal numbers of S/G2+M cells (P < .005), illustrating the enhanced engraftment potential of quiescent cells. G0/G1 cells also provided higher levels of chimerism than equal numbers of total Sca-1+lin AM subfraction (data not shown), negating any suggestion that the Hoechst dye may negatively influence the function of primitive HPCs in active phases of cell cycle. Of interest in these analyses is that, although a relatively large percentage of Sca-1+lin CD49e+cells were in active phases of cell cycle (Figure 4), only those in G0/G1 accounted for the majority of the engraftment potential of this phenotype. All groups of cells contributed equally to lineage-specific hematopoiesis (myeloid and lymphoid; data not shown). Table 3 shows that the quiescent fractions of engrafting Sca-1+lin cells possessed greater proliferative potential and gave rise to greater numbers of Sca-1+ cells during 14 days of in vitro cytokine-stimulated cell culture than engrafting cells in active phases of cell cycle.

Fig. 6.

Percentage of donor-derived chimerism of cell cycle subfractions.

Percentage of donor-derived chimerism at 6 months posttransplantation in mice receiving G0/G1 or S/G2+M subfractions of total Sca-1+lin cells (▪), Sca-1+lin CD11a cells (■), Sca-1+lin CD49ddim cells (●), or Sca-1+lin CD49e+ cells (○). Total Sca-1+lin cells or CD11a, CD49ddim, or CD49e+ subfractions of Sca-1+lin cells were isolated by cell sorting and then subfractionated by means of Hoechst 33342 into G0/G1 or S/G2+M fractions. Between 0.4 to 1.0 × 103 sorted donor cells from B6.BoyJ mice were transplanted into lethally irradiated C57BL/6 mice along with 1.0 × 105 C57BL/6 competitor cells. Chimerism was monitored monthly by CD45.2 analysis of peripheral white blood cells. Purity of sorted donor G0/G1 cells was greater than 97%; purity of donor S/G2+M cells was greater than 82%. Data points are values for the percentage of chimerism for individual mice; horizontal bars are mean chimerism levels. n = 10 for G0/G1 mice, n = 12 for S/G2+M mice. *The presence of statistical significance between mice transplanted with G0/G1 and S/G2+M fractions of engrafting phenotypes; P < .005.

Fig. 6.

Percentage of donor-derived chimerism of cell cycle subfractions.

Percentage of donor-derived chimerism at 6 months posttransplantation in mice receiving G0/G1 or S/G2+M subfractions of total Sca-1+lin cells (▪), Sca-1+lin CD11a cells (■), Sca-1+lin CD49ddim cells (●), or Sca-1+lin CD49e+ cells (○). Total Sca-1+lin cells or CD11a, CD49ddim, or CD49e+ subfractions of Sca-1+lin cells were isolated by cell sorting and then subfractionated by means of Hoechst 33342 into G0/G1 or S/G2+M fractions. Between 0.4 to 1.0 × 103 sorted donor cells from B6.BoyJ mice were transplanted into lethally irradiated C57BL/6 mice along with 1.0 × 105 C57BL/6 competitor cells. Chimerism was monitored monthly by CD45.2 analysis of peripheral white blood cells. Purity of sorted donor G0/G1 cells was greater than 97%; purity of donor S/G2+M cells was greater than 82%. Data points are values for the percentage of chimerism for individual mice; horizontal bars are mean chimerism levels. n = 10 for G0/G1 mice, n = 12 for S/G2+M mice. *The presence of statistical significance between mice transplanted with G0/G1 and S/G2+M fractions of engrafting phenotypes; P < .005.

Close modal
Table 3.

Fold-increase in total cells and Sca-1+ cells in cultures initiated with engrafting phenotypes of Sca-1+linAM+ or AMcells fractionated on the basis of position in cell cycle

PhenotypeSca-1+linCD11aCD43+CD49ddimCD49e+CD62L
G0/G1S/G2+MG0/G1S/G2+MG0/G1S/G2+MG0/G1S/G2+MG0/G1S/G2+MG0/G1S/G2+M
Total cells3-150 1200 70 1900 1400 4100 100 840 10 3900 240 4900 50 
Sca1+cells3-151 1030 35 1800 1200 3970 10 330 2200 33 2340 
PhenotypeSca-1+linCD11aCD43+CD49ddimCD49e+CD62L
G0/G1S/G2+MG0/G1S/G2+MG0/G1S/G2+MG0/G1S/G2+MG0/G1S/G2+MG0/G1S/G2+M
Total cells3-150 1200 70 1900 1400 4100 100 840 10 3900 240 4900 50 
Sca1+cells3-151 1030 35 1800 1200 3970 10 330 2200 33 2340 

AM indicates adhesion molecule.

F3-150

Fold-increase in total cells was calculated by dividing the cell number on day 14 of cytokine supplemented in vitro culture by the input number on day 0.

F3-151

Fold-increase in Sca-1+ cells was calculated by multiplying the percentage of Sca-1+ cells on day 14, determined by flow cytometry, by the total number of cells, and then dividing this number by the input number on day 0.

In this report, we define a theoretical phenotype of a Sca-1+lin long-term engrafting cell as a quiescent cell that expresses high levels of CD43 and CD49e, expresses low or negligible levels of CD11a, CD49d, and CD62L, and proliferates rapidly in vitro following cytokine stimulation. These data are a first step in defining AMs on primitive quiescent HPCs potentially important in the homing and/or anchorage of long-term engrafting cells within the BM microenvironment.

To the best of our knowledge, antibodies used in these studies were nonblocking. In mice where long-term engraftment potential was predominantly among AM/AMdim phenotypes (CD11a, CD49d, CD62L), testing of blocking activity was crucial since low chimerism in AM+ mice may have resulted from blockade of homing or engraftment. While other investigators showed variable inhibition using these same clones but with different cell types in different assays,28-30 we found similar chimerism and HPP-CFC frequency in untreated and antibody-treated Sca-1+lin cells, indicating that these clones were not inhibitory to the in vivo or in vitro activity of Sca-1+lin cells.

Although much work has been performed illustrating the importance of several AMs in various aspects of stem cell function, AMs involved in the sequential movement of transplanted or mobilized HSCs through BM endothelium to or from the periphery are not yet defined. It is likely that different AMs are involved at different stages of stem cell homing, from initial tethering of HSCs on BM endothelium, to rolling, attachment, passage through endothelial barriers, movement within BM tissues, and final anchorage within specialized BM niches, similar to that described for lymphocyte trafficking and leukocyte emigration.31 Mobilization could conceivably involve similar AMs but in reverse order. We found that a sizable fraction of Sca-1+lin CD49e+ cells co-expressed CD43, suggesting that either or both AMs could be involved at some step of the homing process. A role for CD49e in murine long-term and nonobese diabetic/severe combined immunodeficiency (SCID) repopulating cell function has been previously suggested by van der Loo et al,3 while other investigators32 showed no involvement of CD49e in homing or mobilization of murine clonogenic HPCs. Expression of CD43 on primitive human33 and murine HPCs,34 and on cells capable of secondary SCID repopulation,35 has been previously documented. These observations do not exclude the involvement, either synergistically or in a redundant fashion, of other, yet unidentified, AMs in homing of HSCs to the BM. Of the 6 AMs examined in our studies, only CD44 was inefficient in fractionating Sca-1+lin cells into engrafting versus nonengrafting phenotypes. Given the cooperation between CD44 and other AMs,36 as well as the high expression of CD44 on Sca-1+lin cells, it is possible that CD44 may not be directly involved in homing, but may regulate the function of other AMs involved in this process.

While a role of CD49d in homing to and egress from the BM of clonogenic progenitors28,37,38 and malignant cells39,40 has been well defined, to date no one has directly examined CD49d in homing of long-term engrafting cells. The laboratory of Dr Papayannopoulou has shown the ability of function-blocking anti-CD49d antibody to mobilize murine long-term engrafting cells7 and, more recently, to possess a role in homing and mobilization of human CD34+ cells in fetal sheep.1 Given the complexity of homing, it is possible that CD49d may not be involved in the initial steps of HSC homing, but may be critical for the lodgment of HSCs within BM microenvironment following homing via different AMs. This notion is supported by our findings demonstrating that Sca-1+lin cells expressing low levels of CD49d were enriched for long-term engrafting cells (Figure 2). It is also possible that cell cycle status16-19 or rapid changes in functional status5,41-43 may be involved in the control of CD49d function.

CD62L expression on human mobilized peripheral blood (MPB) CD34+ cells has been shown to correlate with rapid platelet engraftment,4,44 but was found in our studies to be detrimental to engraftment potential, presenting the possibility that short-term engrafting cells may express CD62L but long-term engrafting cells may not. Such a concept, however, could be in direct conflict with that recently presented by Ziljmans et al,45 who found primitive HPCs to be responsible for both short- and long-term engraftment. Additionally, since the AM profile is reportedly different in human BM and MPB CD34+cells,4,5,44,46-49 it should not be surprising to find differences in the AM profiles of long-term reconstituting cells from either BM or MPB, within or outside species barriers.

A surprising finding in our studies was the rapid in vitro proliferation and cycling of long-term engrafting cells, which are generally believed to be quiescent.8 While it is possible that cytokine-responsive cells in vitro were not the same cells contributing to long-term engraftment, several lines of evidence support the notion that murine long-term engrafting cells cycle rapidly, at least following transplantation.15,27,50 The rate of in vivo proliferation of colony-forming unit–spleen has been demonstrated to predict long-term engraftment potential of transplanted HPC.27,50 In addition, Nilsson et al15 demonstrated cycling of engrafting murine HSCs within the first 12 hours posttransplantation. Collectively, these data suggest that rapid proliferative responses to cytokine stimulation, either in vivo or in vitro, may be predictive of engraftment potential of selected groups of murine primitive HPCs.

The relationship between cell cycle position and engraftment capability of HSCs has been the focus of much investigation.12,13,15,51Sca-1+lin CD43+ and CD49e+ cells were found to contain a relatively large percentage of cycling cells, posing the question of whether engrafting cells were contained within quiescent or cycling compartments. Despite possessing an apparently crucial repertoire of AMs, cycling cells were deficient in engraftment potential, similar to what has been previously reported in human12 and murine51 systems. Since HPCs express some AMs16-19 and primitive markers52,53 in a cell cycle–related fashion, it is possible that changes in AMs on cycling cells lead to an engraftment defect. Alternatively, as HSCs exit G0/G1 and begin to cycle, hematopoietic potential of these cells may be compromised, as has been previously suggested.12-14Characteristics of putative murine HSCs defined in these studies may be applicable for the selection and in vitro manipulation of analogous human HSCs as targets for gene therapy and ex vivo expansion protocols.

Supported by National Institutes of Health grants RO1 HL55716 and RO1 HL62200 (to E.F.S.); Herman B Wells Center for Pediatric Research is a Center of Excellence in Molecular Hematology (NIDDK P30 DK49218).

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Zanjani
E
Flake
A
Almeida-Porada
G
Tran
N
Papayannopoulou
T
Homing of human cells in the fetal sheep model: modulation by antibodies activating or inhibiting very late activation antigen-4-dependent function.
Blood.
94
1999
2515
2
Liesveld
JL
Dipersio
JF
Abboud
CN
Integrins and adhesive receptors in normal and leukemic CD34+ progenitor cells: potential regulatory checkpoints for cellular traffic.
Leuk Lymphoma.
14
1994
19
3
van der Loo
JCM
Xiao
X
McMillin
D
Hashino
K
Kato
I
Williams
DA
VLA-5 is expressed by mouse and human long-term repopulating hematopoietic cells and mediates adhesion to extracellular matrix protein fibronectin.
J Clin Invest.
102
1998
1051
4
Dercksen
MW
Gerritsen
WR
Rodenhuis
S
et al
Expression of adhesion molecules on CD34+ cells: CD34+ L-selectin+ cells predict a rapid platelet recovery after peripheral blood stem cell transplantation.
Blood.
85
1995
3313
5
Leavesley
DI
Oliver
JM
Swart
BW
Berndt
MC
Haylock
DN
Simmons
PJ
Signals from platelet/endothelial cell adhesion molecule enhance the adhesive activity of the very late antigen-4 integrin of human CD34+ hemopoietic progenitor cells.
J Immunol.
153
1994
4673
6
Lewinsohn
D
Nagler
A
Ginzton
N
Greenberg
P
Butcher
E
Hematopoietic progenitor cell expression of the H-CAM (CD44) homing-associated adhesion molecule.
Blood.
75
1990
589
7
Craddock
C
Nakamoto
B
Andrews
R
Priestley
G
Papayannopoulou
T
Antibodies to VLA4 integrin mobilize long-term repopulating cells and augment cytokine-induced mobilization in primates and mice.
Blood.
90
1997
4779
8
Ogawa
M
Differentiation and proliferation of hematopoietic stem cells.
Blood.
81
1993
2844
9
Bradford
G
Williams
B
Rossi
R
Bertoncello
I
Quiescence, cycling, and turnover in the primitive hematopoietic stem cell compartment.
Exp Hematol.
25
1997
445
10
Abkowitz
JL
Catlin
SN
Guttorp
P
Evidence that hematopoiesis may be a stochastic process in vivo.
Nat Med.
2
1996
190
11
Shah
AJ
Smogorzewska
EM
Hannum
C
Crooks
GM
Flt3 ligand induces proliferation of quiescent human bone marrow CD34+CD38− cells and maintains progenitor cells in vitro.
Blood.
87
1996
3563
12
Gothot
A
van der Loo
J
Clapp
W
Srour
E
Cell cycle-related changes in repopulating capacity of human mobilized peripheral blood CD34+ cells in non-obese diabetic/severe combined immune-deficient mice.
Blood.
92
1998
2641
13
Traycoff
CM
Cornetta
K
Yoder
MC
Davidson
A
Srour
EF
Ex vivo expansion of murine hematopoietic progenitor cells generates classes of expanded cells possessing varying levels of bone marrow repopulating potentials.
Exp Hematol.
24
1996
299
14
Peters
SO
Kittler
ELW
Ramshaw
HS
Quesenberry
PJ
Ex vivo expansion of murine marrow cells with interleukin-3 (IL-3), IL-6, IL-11, and stem cell factor leads to impaired engraftment in irradiated hosts.
Blood.
87
1996
30
15
Nilsson
SK
Dooner
MS
Quesenberry
PJ
Synchronized cell-cycle induction of engrafting long-term repopulating stem cells.
Blood.
90
1997
4646
16
Becker
PS
Nilsson
SK
Li
Z
et al
Adhesion receptor expression by hematopoietic cell lines and murine progenitors: modulation by cytokines and cell cycle status.
Exp Hematol.
27
1999
533
17
Yamaguchi
M
Ikebuchi
K
Hirayama
F
et al
Different adhesive characteristics and VLA-4 expression of CD34+ progenitors in G0/G1 versus S+G2/M phases of the cell cycle.
Blood.
92
1998
842
18
Traycoff
C
Yoder
M
Hiatt
K
Srour
E
Cell cycle stage-specific expression of adhesion molecules may augment engraftment potential of quiescent but not mitotically active hematopoietic progenitor cells [abstract].
Blood.
88
1996
475
19
Srour
E
Traycoff
C
Mobilization of primitive hematopoietic progenitor cells into the periphery may involve their exit from the bone marrow microenvironmnet without entry into active phases of the cell cycle [abstract].
Blood.
88
1996
532
20
Traycoff
CM
Hoffman
R
Zanjani
ED
et al
Measurement of marrow repopulating potential of human hematopoietic progenitor and stem cells using a fetal sheep model.
Prog Clin Biol Res.
389
1994
281
21
Leemhuis
T
Yoder
MC
Grigsby
S
Aguero
B
Eder
P
Srour
E
Isolation of primitive human bone marrow hematopoietic progenitor cells using Hoechst 33342 and Rhodamine 123.
Exp Hematol.
24
1996
1215
22
Uchida
N
Friera
A
He
D
Reitsma
M
Tsukamoto
A
Weissman
I
Hydroxyurea can be used to increase mouse c-kit+, Thy-1.1lo, Lin−/lo, Sca-1+ hematopoietic cell number and frequency in cell cycle in vivo.
Blood.
90
1997
4354
23
Srour
EF
Brandt
JE
Leemhuis
T
Ballas
CB
Hoffman
R
Relationship between cytokine-dependent cell cycle progression and MHC class II antigen expression by human CD34+ HLA-DR− bone marrow cells.
J Immunol.
148
1992
815
24
Yoder
MC
King
B
Hiatt
K
Williams
DA
Murine embryonic yolk sac cells promote in vitro proliferation of bone marrow high proliferative potential colony-forming cells.
Blood.
86
1995
1322
25
Harrison
D
Jordan
C
Zhong
R
Astle
C
Primitive hemopoietic stem cells: direct assay of most productive populations by competitve repopulation with simple binomial correlation and covariance calculations.
Exp Hematol.
21
1993
206
26
Wolf
NS
Kone
A
Priestley
GV
Bartelmez
SH
In vivo and in vitro characterization of long-term repopulating primitive hematopoietic cells isolated by sequential Hoechst 33342-rhodamine 123 FACS selection.
Exp Hematol.
21
1993
614
27
Neben
S
Marcus
K
Mauch
P
Mobilization of hematopoietic stem and progenitor cell subpopulations from the marrow to the blood of mice following cyclophosphamide and/or granulocyte colony-stimulating factor.
Blood.
81
1993
1960
28
Vermeulen
M
Le Pesteur
F
Gagnerault
M
Mary
J
Sainteny
F
Lepault
F
Role of adhesion molecules in the homing and mobilization of murine hematopoietic stem and progenitor cells.
Blood.
92
1998
894
29
Gallatin
W
Weissman
I
Butcher
E
A cell surface molecule involved in organ-specific homing of lymphocytes.
Nature.
304
1983
30
30
Carlos
T
Harlan
J
Leukocyte-endothelial adhesion molecules.
Blood.
84
1994
2068
31
Springer
T
Traffic signals for lymphocyte recirculation and leukocyte emigration: the multistep paradigm.
Cell.
76
1994
301
32
Craddock
C
Nakamoto
B
Elices
M
Papayannapoulou
T
The role of CS1 moiety of fibronectin in VLA4-mediated haemopoietic progenitor trafficking.
Br J Haematol.
97
1997
15
33
Bazil
V
Brandt
J
Chen
S
et al
A monoclonal antibody recognizing CD43 (leukosialin) initiates apoptosis of human hematopoietic progenitor cells but not stem cells.
Blood.
87
1996
1272
34
Hirayama
F
Ogawa
M
CD43 expression by murine lymphohemopoietic progenitors.
Int J Hematol.
60
1994
191
35
Moore
T
Huang
S
Terstappen
LW
Bennett
M
Kumar
V
Expression of CD43 on murine and human pluripotent hematopoietic stem cells.
J Immunol.
153
1994
4978
36
Verfaillie
CM
Benis
A
Iida
J
McGlave
PB
McCarthy
JB
Adhesion of committed human hematopoietic progenitors to synthetic peptides from the C-terminal heparin-binding domain of fibronectin: cooperation between the integrin alpha 4 beta 1 and the CD44 adhesion receptor.
Blood.
84
1994
1802
37
Papayannopoulou
T
Nakamoto
B
Peripheralization of hemopoietic progenitors in primates treated with anti-VLA4 integrin.
Proc Natl Acad Sci U S A.
90
1993
9374
38
Papayannopoulou
T
Craddock
C
Nakamoto
B
Priestley
G
Wolf
N
The VLA4/VCAM adhesion pathway defines contrasting mechanisms of lodgement of transplanted murine hemopoietic progenitors between bone marrow and spleen.
Proc Nat Acad Sci U S A.
92
1995
9647
39
Filshie
R
Gottlieb
D
Bradstock
K
VLA-4 is involved in the engraftment of the human pre-B acute lymphoblastic leukemia cell line NALM-6 in SCID mice.
Br J Haematol.
102
1998
1292
40
Matsuura
N
Purzon-McLaughlin
W
Irie
A
Morikawa
Y
Kakudo
K
Takada
Y
Induction of experimental bone metastasis in mice by transfection of integrin alpha 4 beta 1 into tumor cells.
Am J Pathol.
148
1996
55
41
Prosper
F
Stroncek
D
McCarthy
JB
Verfaillie
CM
Mobilization and homing of peripheral blood progenitors is related to reversible downregulation of alpha4 beta1 integrin expression and function.
J Clin Invest.
101
1998
2456
42
Kovach
NL
Lin
N
Yednock
T
Harlan
JM
Broudy
VC
Stem cell factor modulates avidity of alpha 4 beta 1 and alpha 5 beta 1 integrins expressed on hematopoietic cell lines.
Blood.
85
1995
159
43
Levesque
JP
Leavesley
DI
Niutta
S
Vadas
M
Simmons
PJ
Cytokines increase human hemopoietic cell adhesiveness by activation of very late antigen (VLA)-4 and VLA-5 integrins.
J Exp Med.
181
1995
1805
44
Watanabe
T
Dave
B
Heimann
D
Jackson
J
Kessinger
A
Talmadge
J
Cell adhesion molecule expression on CD34+ cells in grafts and time to myeloid and platelet recovery after autologous stem cell transplantation.
Exp Hematol.
26
1998
10
45
Ziljmans
J
Visser
J
Laterveer
L
et al
The early phase of engraftment after murine blood cell transplantation is mediated by hematopoietic stem cells.
Proc Nat Acad Sci U S A.
95
1998
725
46
Turner
ML
McIlwaine
K
Anthony
RS
Parker
AC
Differential expression of cell adhesion molecules by human hematopoietic progenitor cells from bone marrow and mobilized adult peripheral blood.
Stem Cells.
13
1995
311
47
To
LB
Haylock
DN
Dowse
T
et al
A comparative study of the phenotype and proliferative capacity of peripheral blood (PB) CD34+ cells mobilized by four different protocols and those of steady-phase PB and bone marrow CD34+ cells.
Blood.
84
1994
2930
48
Fultz
C
Shivers
S
Smilee
R
Janssen
W
Hematopoietic cell adhesion molecule and very late antigen-4 but not L-selectin are differentially expressed between marrow and blood [abstract].
Exp Hematol.
24
1996
1078
49
Zini
J
Marolleau
J
Miclea
J
Mouton
V
Benbunan
M
Expression of adhesion molecules on CD34+ cells: comparative studies between peripheral blood and bone marrow stem cells [abstract].
Exp Hematol.
24
1996
1034
50
Mauch
P
Hellman
S
Loss of hematopoietic stem cell self-renewal after bone marrow transplantation.
Blood.
74
1989
872
51
Fleming
WH
Alpern
EJ
Uchida
N
Ikuta
K
Spangrude
GJ
Weissman
IL
Functional heterogeneity is associated with the cell cycle status of murine hematopoietic stem cells.
J Cell Biol.
122
1993
897
52
Sato
T
Laver
J
Ogawa
M
Reversible expression of CD34 by murine hematopoietic stem cells.
Blood.
94
1999
2548
53
Orlic
D
Fischer
R
Nishikawa
S
Nienhuis
AW
Bodine
DM
Purification and characterization of heterogeneous pluripotent hematopoietic stem cell populations expressing high levels of c-kit receptor.
Blood.
82
1993
762

Author notes

Christie M. Orschell-Traycoff, Indiana University School of Medicine, 1044 West Walnut St, R4-202, Indianapolis, IN 46202-5254; e-mail: ctraycof@iupui.edu.

Sign in via your Institution