Expression of the CD4 antigen was observed on human fetal liver, fetal bone marrow (BM), and umbilical cord blood progenitors expressing high levels of CD34. Using clonal and liquid-culture assays, CD4+ CD34++ Lin (lineage = CD3, CD8, CD10, CD14, CD15, CD16, CD19, CD20, and glycophorin A) fetal liver progenitors were found to have a greater proliferative potential than CD4 CD34++ Lin progenitors, whereas the CD4 fraction was more enriched for erythroid progenitors. Both the CD4+ and the CD4 progenitor subpopulations also gave rise to multilineage engraftment upon transplantation into human fetal bone fragments, supportive of B-lymphoid and myeloid growth, or into human fetal thymic fragments, supportive of T-cell growth, implanted in scid/scid (SCID) mice. However, in SCID-hu mice transplanted with graded doses of donor cells ranging from 2.0 × 102 to 2.0 × 104 cells, BM reconstitution by the CD4+ fraction of CD34++ Lin cells was more frequent than by the CD4 fraction when low numbers of cells were injected. These functional data strongly suggest that stem cells reside among CD4+ CD34++ Lin fetal liver cells. This hypothesis was further supported by the observations that CD4+ CD34++ Lin fetal liver cells were enriched for CDw90+ (Thy-1), CD117+ (kit), CD123+, HLA-DR+, CD7, CD38, CD45RA, CD71, CD115 (fms), and rhodamine 123dull cells, a phenotypic profile believed to represent fetal stem cells. Furthermore, all CD4+ CD34++ Lin fetal liver cells also expressed CD13 and CD33.

CD4 IS A 56-kD glycoprotein, the expression of which is best known to define a subset of mature T cells.1 The CD4+ mature T cells usually interact with cells expressing class II major histocompatibility complex (MHC).2 The function of CD4 in these interactions is that of a coreceptor for class II molecules that augments the stability of the complex formed by the T-cell receptor (TCR) and the antigen presented by MHC. The requirement of CD4 expression on mature T cells for their function was, in part, shown in CD4-deficient mice that displayed deficient T-cell helper function.3 CD4 has also been shown to function as a receptor for the cytokine, lymphocyte chemoattractant factor, therefore suggesting a functional role for CD4 on cells that do not express TCR.4 

CD4 expression has been observed on a number of human cell types that do not express TCR, including megakaryocytes,5 eosinophils,6 monocytes, and Langerhans cells.7 During intrathymic development in both mice and humans, CD4 is expressed by immature (TCR) and mature (TCR+) thymocytes.8,9 In the human thymus, the existence of CD34+ CD4+ CD3 CD8 cells that are an intermediate stage between CD34+ CD3 CD4 CD8 (triple negative) thymocytes and CD34 CD3± CD4+ CD8+ (double positive [DP]) thymocytes has been recently shown.10,11 Furthermore, several studies in the mouse have documented the expression of CD4 on early multipotent progenitors in both the thymus and bone marrow (BM). Wu et al12 have shown the existence of a murine thymic CD4lo progenitor population that has lost myeloid and erythroid lineage potential but remains capable of T- and B-lymphoid development. Although the expression of CD4 on murine stem cells remains controversial,13,14 low levels of CD4 expression on murine BM progenitors15 and stem cells capable of long-term BM reconstitution have been observed.16-18 

Low levels of CD4 expression on human adult BM progenitors, defined by their expression of CD34, have recently been reported.19,20 Louache et al20 have shown that CD4 is expressed on adult BM CD34+ cells with the phenotypic profile of primitive hematopoietic progenitors. Furthermore, these investigators observed that the CD34+ CD4+ population was also enriched for long-term culture-initiating cells (LTC-IC).20 These studies showed that CD4 expression is not only limited to mature T cells, but that CD4 is also expressed on hematopoietic progenitors at various stages of lineage commitment. In this study, we report the expression of low levels of CD4 by human fetal hematopoietic progenitors expressing high levels of CD34. Moreover, these CD4+ CD34++ cells were found to coexpress a pattern of cell surface antigens consistent with the phenotypic profile believed to represent fetal hematopoietic stem cells. Functionally, this population of CD4+ cells was shown to contain erythroid, myeloid, B-cell, and T-cell progenitors. The CD4+ cells also displayed an extensive in vitro and in vivo proliferative capacity that was greater than that observed of CD4 cells. These data show that cells isolated from the human fetal liver expressing low levels of CD4 appear to contain hematopoietic stem cells.

Cytokines.Recombinant human c-kit ligand (KL) was purchased from R&D Systems, Inc (Minneapolis, MN) and was used at 20 ng/mL. Recombinant human granulocyte-macrophage colony-stimulating factor (GM-CSF ), used at 20 ng/mL, was provided by the Schering-Plough Corp (Kenilworth, NJ). Recombinant human erythropoietin (EPO; Amgen, Thousand Oaks, CA) was used at 1 U/mL.

Antibodies.The following unconjugated, biotinylated, phycoerythrin (PE)-labeled, fluorescein isothiocyanate (FITC)-labeled, or tricolor (TC)-labeled polyclonal or monoclonal antibodies (MoAbs) were purchased from Becton Dickinson (San Jose, CA) or an otherwise stated vendor: mouse IgG1-PE, mouse IgG2a-PE, mouse IgG1-FITC, mouse IgG2a-FITC, mouse IgG1-TC (Caltag Laboratories, South San Francisco, CA), mouse IgG2a-TC (Caltag Laboratories), CD3 (SK7), CD3-PE (SK7), CD3-FITC (SK7), CD4 (SK3), CD4-FITC (SK3 + SK4), CD4-PE (SK3), CD7-FITC (4H9), CD8 (SK1), CD8-FITC (SK1), CD10-PE (5-1B4; Caltag Laboratories), CD13-PE (WM-47; DAKO, Carpinteria, CA), CD14 (MØP9), CD14-PE (MØP9), CD16-PE (B73.1), CD19 (4G7), CD19-PE (4G7), CD20 (L27), CD20-PE (L27), CD33-PE (P67.6), CD33-TC (251; Caltag Laboratories), CD34-biotin (8G12), CD34-PE (8G12), CD34-FITC (8G12), CD38-PE (HB-7), CD38-FITC (HIT2; Caltag Laboratories), CD45RO-PE (UCHL-1), CD45RA-FITC (L48), CD56 (MY31), CD56-PE (MY31), CD71-FITC (L01.1), CDw90-PE (5E10; PharMingen; San Diego, CA), CD115 (3-4A4-E4; Oncogene Science, Uniondale, NY), CD117-PE (95C3; Immunotech, Westbrook, ME), CD123-PE (9F5; PharMingen), HLA-DR-PE (L243), HLA-DR-TC (HL38; Caltag Laboratories), glycophorin A (GPA; 10F7MN; American Type Tissue Culture Collection [ATCC], Rockville, MD), HLA-A2-FITC (CR11351; kindly provided by Dr P. Parham, Stanford, CA), HLA-A3-FITC (GAP A3; ATCC), HLA-A11/A24-FITC (A11.1M; ATCC), and HLA-B7/B40-FITC (MB40.2; ATCC). Allophycocyanin (APC)-labeled streptavidin was also purchased from Becton Dickinson, whereas TC-labeled streptavidin and goat antirat IgG-PE were purchased from Caltag Laboratories.

Human hematopoietic tissues.Human fetal liver and fetal BM were obtained from Advanced Bioscience Resources (Alameda, CA) and were used with the approval of the Committee for the Protection of Human Subjects at our institute. The gestational age of the abortuses, ranging from 16 to 24 weeks, was approximated based on the foot length of the fetus.

Umbilical cord blood was obtained from normal full-term deliveries and was kindly provided by Dr U. Chitkara (Department of Labor and Delivery, Stanford University Medical School, Palo Alto, CA). After delivery of the infant and ligation of the umbilical cord, the umbilical cord blood was harvested, before the expulsion of the placenta, using a 60-mL syringe and transferred to a heparinized 50-mL tube.

Phenotypic analysis of hematopoietic tissues.The expression of cell surface antigens was determined by flow cytometry using a FACScan (Becton Dickinson). In all experiments in which fetal liver, fetal BM, or umbilical cord blood cells were labeled with MoAb, the cells were first preincubated for 10 minutes in a solution of phosphate-buffered saline (PBS) free of added Ca+2 and Mg+2 supplemented with 1 mmol/L ethylenediaminetetraacetic acid, 50 μg/mL gentamicin (GIBCO, Grand Island, NY), 0.5% human γ-globulins (Sigma Chemical Co, St Louis, MO), and 2% normal mouse serum followed by staining with MoAb for at least 30 minutes in the same solution. After staining, the cells were washed twice and resuspended in PBS containing 0.5% bovine serum albumin (BSA; Sigma Chemical Co) and 0.05% NaN3 followed by FACScan analysis. For some analyses, propidium iodide (PI; Sigma Chemical Co) was added at a concentration of 1 μg/mL to the final cell suspension to stain dead cells. A range of 5 × 103 to 4 × 104 events were collected per sample using a live-cell gate based on negative PI staining.

Rhodamine 123 (Rh123; Molecular Probes, Eugene, OR) staining was performed by incubating CD34++ Lin cells, isolated by fluorescence-activated cell sorting (FACS), with 0.1 μg/mL Rh123 for 30 minutes at 37°C. Thereafter, the cells were washed and resuspended in medium without Rh123 for 30 to 120 minutes at 37°C to allow for Rh123 efflux. These cells were then washed at 4°C with PBS containing 0.5% BSA and 0.05% NaN3 and stained for CD4 according to the protocol described above.

Hematopoietic progenitor isolation.Light-density fetal liver (LDFL) cells were prepared by density centrifugation and depleted of GPA+ cells by immunomagnetic bead depletion as previously described.21 GPA light-density fetal BM (LDFBM) cells and GPA light-density umbilical cord blood (LDCB) cells were prepared in a similar manner. In preparation for cell sorting or for some phenotypic analysis, GPA LDFL, LDFBM, or LDCB cells were depleted of lineage (Lin)-committed progenitors and mature hematopoietic cells by immunomagnetic bead depletion with a cocktail of MoAbs detailed in the Results. The MoAb-labeled cells were washed and resuspended in PBS containing 2% fetal calf serum (FCS) and 50 μg/mL gentamicin and FACS was performed under the conditions previously described.21 

In vitro hematopoietic progenitor assays.Progenitors with myeloid potential were assayed in double-layered clonal cultures in 60 × 15 mm tissue culture dishes (Becton Dickinson).21 The culture medium consisted of Iscove's modified Dulbecco's medium (Sigma Chemical Co) supplemented with 20% FCS (JRH Biosciences, Lenexa, KS), 50 μg/mL gentamicin, and 7.5 × 10−5 mol/L α-thioglycerol (Sigma Chemical Co). The bottom layer of the cultures consisted of 2 mL of culture medium solidified with the addition of 0.5% SeaPlaque agarose, a low gelling temperature form of agarose (FMC BioProducts, Rockland, ME). The cytokine combination KL + GM-CSF, which has been shown to stimulate a high frequency as well as a broad spectrum of fetal hematopoietic progenitors,22 was added to the bottom layer of the cultures. Cells were seeded in a volume of 1 mL of culture medium supplemented with 0.36% agarose on top of the solidified bottom layer. The cultures were maintained at 37°C in a fully humidified atmosphere containing 5% CO2 in air. After 3 weeks, the cultures were analyzed for the growth of two types of progenitors based on colony size: low proliferative potential colony-forming cells (LPP-CFC) were defined as progenitors giving rise to colonies of ≥50 cells but smaller than colonies derived from high proliferative potential colony-forming cells (HPP-CFC). HPP-CFC were defined as progenitors giving rise to colonies ≥0.5 mm in diameter that were approximated to consist of 1 × 104 to 1 × 106 cells.

Erythroid progenitors were assayed in 1-mL clonal cultures stimulated by KL + EPO grown in 35 × 10 mm culture dishes (Becton Dickinson). The same culture medium as described above was used except for the substitution of 1.2% methyl cellulose (4,000 cp; Sigma Chemical Co) for agarose as the semisolid support matrix. Burst-forming units-erythroid (BFU-E) were defined as hemoglobinized colonies containing at least 50 cells and were enumerated after 3 weeks of growth in a fully humidified atmosphere containing 5% CO2 in air.

The proliferative potential of the purified progenitor subpopulations was further measured using the delta (Δ) assay as previously described.21 Briefly, sorted fetal liver cells were grown in liquid cultures stimulated by KL + GM-CSF for 7 days and then assayed for their potential to give rise to secondary HPP-CFC and LPP-CFC. The numbers of primary HPP-CFC and LPP-CFC were determined from cultures initiated in parallel with the liquid cultures. The data are presented as the fold increase (Δ) in progenitors during liquid culture. Furthermore, cells generated in liquid cultures were also analyzed for their expression of cell surface markers. In some experiments, the length of time in liquid culture was extended beyond 7 days before the cultures were harvested and the cell numbers were determined and phenotypic analysis was performed. Further details of the cytokines used for these long-term liquid cultures are described in the Results.

In vivo assays of human hematopoietic progenitors.T-cell progenitor activity was assessed by using SCID-hu mice established as described23 by combined fetal liver and fetal thymus implantation under the kidney capsule of CB.17 scid/scid (SCID) mice at least 8 weeks before progenitor transplantation. Fifty to 5.0 × 103 sorted CD4+ CD34++ Lin LDFL cells or CD4 CD34++ Lin LDFL cells were directly injected into human thymic grafts in a volume of 2 μL using a 10-μL Hamilton syringe 1 to 3 hours after whole body γ-irradiation with 2.5 Gy. Additionally, in some experiments, the sorted fetal liver progenitors were injected without prior irradiation of SCID-hu mice that were constructed with only fetal thymic tissue. The degree of donor cell reconstitution was analyzed by three-color FACScan analysis. Engraftment by donor cells was determined by detecting a human leukocyte antigen (HLA)-mismatch between donor and host thymocytes. BM-reconstituting ability, as well as B-cell lineage potential, of the sorted CD4+ CD34++ Lin LDFL or CD4 CD34++ Lin LDFL subpopulations was determined by injecting sorted cells directly into the human BM of SCID-hu mice implanted subcutaneously with human fetal bones at least 8 weeks before.24-26 The SCID-hu mice received 2.5 Gy γ-irradiation 1 to 3 hours before transplantation of the sorted fetal liver cells. At the indicated time points after transplantation, the level of donor cell engraftment, again determined by an HLA-mismatch between donor and host cells, was determined by FACScan analysis.25,26 

Data presentation and statistical analysis.The data from multiple experiments were pooled and are presented as the mean ± 1 standard error of the mean (SEM). Statistical significance was determined on individual experiments (single tissues) using the two-tailed paired Student's t-test. Statistical significance of data pooled from multiple experiments was calculated using the nonparametric paired sign test. Differences in the data were considered significant when P ≤ .05.

Phenotypic characterization of CD4+ human fetal hematopoietic progenitors.The expression of CD4 on CD34+ progenitor cells was investigated using light-density erythrocyte-depleted (GPA) fetal liver, fetal BM, and umbilical cord blood cells (Fig 1). Low levels of CD4 expression were observed on CD34+ cells obtained from each of the three tissue types. In the representative experiment shown in Fig 1, CD34 was found to be expressed on 65% of GPA LDFL cells, on 29% of GPA LDFBM cells, and only on 2% to 3% of GPA LDCB cells. The CD4 antigen expression was found on 53% of CD34+ GPA LDFL cells, 28% of CD34+ GPA LDFBM cells, and on approximately 57% of CD34+ GPA LDCB cells. In control stainings, no clear expression of CD8, as detected by an MoAb of the same isotype as that of the CD4 MoAb, was observed on CD34+ cells. The levels of CD4 expression on CD34+ cells were lower than the levels of CD4 expressed by mature T cells, which were present in abundance among the CD34 GPA fractions of LDFL and LDCB cells. The identification of the CD4++ CD34 LDFL cells as mature T cells was confirmed by the coexpression of CD3 observed in three-color stainings (data not shown). Cells of the monocytic lineage were most likely responsible for the low levels of CD4 expression on CD34 cells, because many of the CD4+ CD34 LDFL cells coexpressed CD11b, CD11c, CD13, CD14, CD15, CD18, and CD33 (data not shown). In contrast, very low levels of CD4 expression were observed on less than 5% of B-cell progenitors defined by their expression of CD34 and either CD10, CD19, or CD20 (data not shown). Further depletion of GPA LDFL, LDFBM, or LDCB cells of Lin+ cells expressing CD3, CD8, CD14, CD19, CD20, and CD56 resulted in an enrichment of CD34+ cells expressing CD4 (Fig 1, bottom panels). After Lin depletion, the expression of low levels of CD4 (CD4+) on a subpopulation of cells expressing the highest levels of CD34 (CD34++) was clearly evident in each of the three hematopoietic tissues.

Fig. 1.

Expression of CD4 by CD34+ human fetal liver, fetal BM, and umbilical cord blood cells. LDFL, LDFBM, and LDCB cells were depleted of GPA+ cells and analyzed for the expression of CD34 in combination with CD8 or CD4. The analysis of antigen expression was limited to cells with a low side scatter, containing the bulk of the CD34+ cells, as defined by the gates shown in the top row. The expression of CD4 and CD34 on LDFL, LDFBM, and LDCB cells, depleted of Lin+ cells by magnetic bead depletion of CD3+, CD8+, CD14+, CD19+, CD20+, and CD56+ cells, is shown in the bottom row. CD4+ CD34++ Lin and CD4 CD34++ Lin LDFL cells are indicated in the bottom-left panel by regions representative of those used for the isolation of these cells by FACS in subsequent experiments. The percentage of events found in each quadrant is indicated. Contour plots were made using a linear density scale at the degree of resolution indicated in the top-right corner of each plot (% Lin). Dot plots are used to show data on cord blood cells containing a low percentage of CD34+ cells. These data are representative of at least three analyses per tissue type.

Fig. 1.

Expression of CD4 by CD34+ human fetal liver, fetal BM, and umbilical cord blood cells. LDFL, LDFBM, and LDCB cells were depleted of GPA+ cells and analyzed for the expression of CD34 in combination with CD8 or CD4. The analysis of antigen expression was limited to cells with a low side scatter, containing the bulk of the CD34+ cells, as defined by the gates shown in the top row. The expression of CD4 and CD34 on LDFL, LDFBM, and LDCB cells, depleted of Lin+ cells by magnetic bead depletion of CD3+, CD8+, CD14+, CD19+, CD20+, and CD56+ cells, is shown in the bottom row. CD4+ CD34++ Lin and CD4 CD34++ Lin LDFL cells are indicated in the bottom-left panel by regions representative of those used for the isolation of these cells by FACS in subsequent experiments. The percentage of events found in each quadrant is indicated. Contour plots were made using a linear density scale at the degree of resolution indicated in the top-right corner of each plot (% Lin). Dot plots are used to show data on cord blood cells containing a low percentage of CD34+ cells. These data are representative of at least three analyses per tissue type.

Close modal

To characterize the phenotype of CD4+ CD34++ cells, fetal liver CD34++ Lin cells were isolated by FACS and analyzed for the expression of CD4 in conjunction with various cell surface markers (Fig 2). Previous studies suggest that hematopoietic stem cells derived from adult and fetal tissues are among CD34++ cells expressing low to negative levels of CD7,27,28 CD38,21,29-31 CD45RA,21,31-33 and CD71.21,31,34 CD4 expression by CD34++ Lin LDFL cells was found to be inversely correlated with the expression of CD7, CD38, CD45RA, and CD71. Moreover, the expression of CD4 by CD34++ Lin LDFL cells correlated with the expression of CDw90 (Thy-1). The detection of CDw90 necessitated the use of a PE-labeled anti-CDw90 MoAb due to the weak expression of this antigen. Because higher levels of CD4 expression were also detected using PE-labeled as compared with FITC-labeled MoAbs, the population of CD34++ Lin LDFL cells coexpressing CD4 and CDw90 is underestimated by the data shown in Fig 2. Most CD4+ cells were also found to express HLA-DR, although CD4 expression was also observed on the minority of CD34++ Lin LDFL cells lacking the expression of HLA-DR. Additionally, all CD4+ CD34++ Lin LDFL cells expressed low levels of CD13 and CD33. We have previously reported that CD13 and CD33, markers associated with myeloid cell differentiation, may be expressed on fetal hematopoietic stem cells.21 The growth factor receptors CD117 (c-kit) and CD123 (the α-chain of the interleukin-3 [IL-3] receptor) were both uniformly expressed on CD4+ CD34++ Lin LDFL cells, whereas the expression of these receptors appeared to diverge on CD4 cells (Fig 2). Almost no expression of CD115, the receptor for macrophage colony-stimulating factor, was seen on CD4+ cells, but high levels of CD115 expression were seen on a subpopulation of CD4 cells. The heterogeneity of growth factor receptor expression on CD4 cells suggests that this population of cells is composed of functionally diverse progenitors.

Fig. 2.

Phenotypic analysis of CD4+ CD34++ Lin LDFL cells. CD34++ Lin LDFL cells were isolated by immunomagnetic bead depletion of GPA LDFL cells after staining with FITC-labeled MoAb against CD3, CD8, CD10, CD14, CD15, CD16, CD19, and CD20. These Lin cells were then stained for CD34 expression in a two-step protocol using anti-CD34-biotin followed by streptavidin-APC or streptavidin-TC. CD34++ Lin PI LDFL cells were isolated by FACS. The sorted cells were stained for CD4 and the other cell-surface markers indicated. Additionally, staining with CD4 and Rh123 was also performed, for which the incubation periods for Rh123 efflux are indicated. These data are shown using linear-density contour plots with one degree of smoothing. A 10% resolution was used for all plots except for the analyses of cytokine receptor expression and HLA-DR, which are shown at 5% resolution. However, the analysis of CD38 versus CD4 expression is shown on a 50% logarithmic-density contour plot to show the expression of CD4 on the rare subpopulation of CD34++ that does not express CD38. These data are compiled from the analyses of multiple tissues and are representative of at least two analyses per phenotype.

Fig. 2.

Phenotypic analysis of CD4+ CD34++ Lin LDFL cells. CD34++ Lin LDFL cells were isolated by immunomagnetic bead depletion of GPA LDFL cells after staining with FITC-labeled MoAb against CD3, CD8, CD10, CD14, CD15, CD16, CD19, and CD20. These Lin cells were then stained for CD34 expression in a two-step protocol using anti-CD34-biotin followed by streptavidin-APC or streptavidin-TC. CD34++ Lin PI LDFL cells were isolated by FACS. The sorted cells were stained for CD4 and the other cell-surface markers indicated. Additionally, staining with CD4 and Rh123 was also performed, for which the incubation periods for Rh123 efflux are indicated. These data are shown using linear-density contour plots with one degree of smoothing. A 10% resolution was used for all plots except for the analyses of cytokine receptor expression and HLA-DR, which are shown at 5% resolution. However, the analysis of CD38 versus CD4 expression is shown on a 50% logarithmic-density contour plot to show the expression of CD4 on the rare subpopulation of CD34++ that does not express CD38. These data are compiled from the analyses of multiple tissues and are representative of at least two analyses per phenotype.

Close modal

Human stem cells have also been enriched on the basis of their low level of staining with the dye Rh123, which is in part due to the expression of the efflux pump P-glycoprotein on these cells.31,35,36 Sorted CD34++ Lin LDFL cells were stained with Rh123, followed by 30 to 120 minutes of efflux. Both CD4+ and CD4 cells were found among those cells expressing the lowest levels of Rh123; however, the CD4+ fraction was enriched for Rh123dull cells (Fig 2).

Colony-forming activities of the CD4+ and CD4 subsets of CD34++ Lin fetal liver cells.Functional studies were performed on the CD4+ and CD4 subsets of CD34++ Lin LDFL cells, which were isolated by FACS using the criteria shown in Fig 1. Consistent with the results from phenotypic analyses (Fig 2) suggesting that CD4+ cells are enriched among the most primitive fetal liver progenitors, the frequency of early myeloid progenitors with a high proliferative capacity (HPP-CFC; Table 1) was found to be significantly greater among CD4+ cells than CD4 cells (paired sign test, P = .031; n = 6 experiments). The frequencies of LPP-CFC detected among CD4+ and CD4 progenitors were similar. We have previously reported that the most primitive HPP-CFC, those progenitors with the greatest proliferative capacity, are enriched among CD34++ CD38 Lin LDFL cells,21 although in the presence of KL + GM-CSF large colony formation can also be detected in cultures of the more mature CD34++ CD38+ Lin LDFL cells.22 Thus, to investigate the expression of CD4 among fetal liver progenitors expressing negative to low levels of CD38, CD34++ CD38 Lin LDFL cells were isolated based on their expression of CD4. The frequency of HPP-CFC among CD4 CD34++ CD38 Lin LDFL cells was found to be significantly less than among CD4+ CD34++ CD38 Lin LDFL cells. In three analyses of cells stained for FACS, the percentage of CD4+ cells among CD34++ CD38 Lin LDFL cells was found to range from 60% to 87%, which is consistent with the phenotypic analysis shown in Fig 2. Thus, the majority of the most primitive HPP-CFC, those residing among CD34++ CD38 Lin LDFL cells, express CD4.

Table 1.

Myeloid and Erythroid Progenitor Activity Detected Among Populations of CD4+ and CD4 Fetal Liver Progenitors

ExperimentCell Population*HPP-CFCLPP-CFCBFU-E
CD4+CD4PCD4+CD4PCD4+CD4P
CD34++ Lin 32.0 ± 2.0 14.2 ± 2.5 .0080 52.9 ± 0.9 40.9 ± 3.2 .0460 2.7 ± 0.8 6.7 ± 0.8 .0351 
CD34++ Lin 96.9 ± 1.6 58.7 ± 10.1 .0712 67.1 ± 8.6 75.6 ± 7.6 .0762 6.7 ± 1.5 25.8 ± 0.4 .0101 
CD34++ Lin 82.2 ± 3.5 45.3 ± 5.4 .0237 79.6 ± 8.0 98.7 ± 7.1 .1439 ND ND ND 
CD34++ Lin 78.2 ± 6.5 28.4 ± 3.8 .0100 54.2 ± 6.2 87.6 ± 12.0 .0325 ND ND ND 
CD34++ Lin 124.4 ± 4.5 52.9 ± 2.7 .0042 149.3 ± 8.0 166.2 ± 7.5 .3844 20.0 ± 6.7 44.9 ± 2.4 .0441 
CD34++ Lin 96.0 ± 4.2 74.7 ± 7.4 .0343 170.7 ± 2.9  130 ± 3.1 .0035 23.1 ± 4.2 48.4 ± 11.6 .1013 
CD34++ CD38 Lin 117.8 ± 13.5 57.8 ± 9.5 .0574 158.9 ± 15.4 95.6 ± 7.3 .0747 ND ND ND 
CD34++ CD38 Lin 284.4 ± 33.2 90.4 ± 15.5 .0230 87.8 ± 15.4 171.9 ± 10.4 .0080 ND ND ND 
CD34++ CD38 Lin 173.3 ± 5.8 125.6 ± 9.5 .0148 255.6 ± 4.0 217.8 ± 13.5 .0984 ND ND ND 
ExperimentCell Population*HPP-CFCLPP-CFCBFU-E
CD4+CD4PCD4+CD4PCD4+CD4P
CD34++ Lin 32.0 ± 2.0 14.2 ± 2.5 .0080 52.9 ± 0.9 40.9 ± 3.2 .0460 2.7 ± 0.8 6.7 ± 0.8 .0351 
CD34++ Lin 96.9 ± 1.6 58.7 ± 10.1 .0712 67.1 ± 8.6 75.6 ± 7.6 .0762 6.7 ± 1.5 25.8 ± 0.4 .0101 
CD34++ Lin 82.2 ± 3.5 45.3 ± 5.4 .0237 79.6 ± 8.0 98.7 ± 7.1 .1439 ND ND ND 
CD34++ Lin 78.2 ± 6.5 28.4 ± 3.8 .0100 54.2 ± 6.2 87.6 ± 12.0 .0325 ND ND ND 
CD34++ Lin 124.4 ± 4.5 52.9 ± 2.7 .0042 149.3 ± 8.0 166.2 ± 7.5 .3844 20.0 ± 6.7 44.9 ± 2.4 .0441 
CD34++ Lin 96.0 ± 4.2 74.7 ± 7.4 .0343 170.7 ± 2.9  130 ± 3.1 .0035 23.1 ± 4.2 48.4 ± 11.6 .1013 
CD34++ CD38 Lin 117.8 ± 13.5 57.8 ± 9.5 .0574 158.9 ± 15.4 95.6 ± 7.3 .0747 ND ND ND 
CD34++ CD38 Lin 284.4 ± 33.2 90.4 ± 15.5 .0230 87.8 ± 15.4 171.9 ± 10.4 .0080 ND ND ND 
CD34++ CD38 Lin 173.3 ± 5.8 125.6 ± 9.5 .0148 255.6 ± 4.0 217.8 ± 13.5 .0984 ND ND ND 

Abbreviation: ND, not determined.

*

CD4+ CD34++ Lin and CD4 CD34++ Lin LDFL cells were isolated by immunomagnetic bead depletion of GPA LDFL cells using a cocktail of PE-labeled MoAbs against CD3, CD8, CD10, CD14, CD16, CD19, CD20, and CD56. These Lin cells were then stained for CD34 and CD4 expression using anti-CD34-biotin followed by streptavidin-APC and anti-CD4-FITC. Electronic gates for FACS were defined as shown in Fig 1. CD4+ CD34++ CD38 Lin and CD4 CD34++ CD38 Lin LDFL cells were isolated using a similar protocol except that the cocktail of MoAbs against Lin markers was FITC-labeled and did not contain anti-CD56 but did contain anti-CD38. Consequently, CD4 expression was detected using anti-CD4-PE.

Results are presented as the mean CFU-C ± SE/1 × 103 cells. Triplicate cultures of CD4+ CD34++ Lin or CD4 CD34++ Lin LDFL cells were initiated with 7.5 × 102 cells/culture dish. Cultures of CD4+ CD34++ CD38 Lin or CD4 CD34++ CD38 Lin LDFL cells were initiated with 2.0 to 3.0 × 102 cells/culture dish.

P values were determined using the Student's t-test.

Both sorted cell populations contained BFU-E detected in cultures stimulated by EPO + KL (Table 1). However, significantly greater numbers of BFU-E were observed among CD4 CD34++ Lin LDFL cells than among CD4+ CD34++ Lin LDFL cells. Because the culture conditions used support the growth of very few erythroid progenitors found among primitive CD34++ CD38 Lin LDFL progenitors, these data reflect the growth of more mature erythroid progenitors that express CD38 and respond to EPO + KL (M.O.M., M.-G.R., and R.N., manuscript in preparation). Thus, among the more mature CD34++ CD38+ Lin LDFL cells, erythroid progenitors are concentrated in the CD4 fraction. In contrast, the predominance of HPP-CFC in the CD4+ fraction of CD34++ Lin LDFL cells suggests that this population is enriched for primitive progenitors.

Ex vivo expansion potential of the CD4+ and CD4 subsets of CD34++ fetal liver progenitors.The growth in liquid cultures of CD4+ CD34++ CD38 Lin and CD4 CD34++ CD38 Lin LDFL cells showed marked differences in these two progenitor subpopulations. After 4 days of culture in the presence of KL + GM-CSF + EPO, both the cultured CD4+ and CD4 progenitors were observed to express CD4 and CD34 (Fig 3A). The mean intensity of CD34 fluorescence was twofold higher on the cultured CD4+ progenitors (1,835) than on the cultured CD4 progenitors (912). After 7 days of growth, higher percentages of cells expressing higher levels of CD34 continued to be observed in cultures of CD4+ progenitors, relative to cultures of CD4 progenitors (Fig 3B). Therefore, the kinetics of the loss of CD34 expression clearly distinguish the CD4+ progenitors from the CD4 progenitors. It is also of interest to note that the CD4 CD34++ CD38 Lin LDFL cells could become CD4+ CD34+ cells in vitro.

Fig. 3.

Analysis of CD4 and CD34 expression on cultured CD4+ CD34++ CD38 Lin and CD4 CD34++ CD38 Lin LDFL cells. (A) Fetal liver cells were prepared for FACS by the method described in the legend to Table 1, with the exception that streptavidin-TC was substituted for streptavidin-APC. After 4 days of culture in KL + GM-CSF + EPO, the cultured cells were restained using the same MoAbs as used for cell sorting and analyzed using a FACScan. (B) CD4+ CD34++ CD38 Lin and CD4 CD34++ CD38 Lin LDFL cells, isolated by the identical procedure used in Table 1, were reanalyzed after 7 days of culture in KL + GM-CSF. CD34 expression was clearly observed to be greater on the cultured CD4+ cells than on the cultured CD4 cells.

Fig. 3.

Analysis of CD4 and CD34 expression on cultured CD4+ CD34++ CD38 Lin and CD4 CD34++ CD38 Lin LDFL cells. (A) Fetal liver cells were prepared for FACS by the method described in the legend to Table 1, with the exception that streptavidin-TC was substituted for streptavidin-APC. After 4 days of culture in KL + GM-CSF + EPO, the cultured cells were restained using the same MoAbs as used for cell sorting and analyzed using a FACScan. (B) CD4+ CD34++ CD38 Lin and CD4 CD34++ CD38 Lin LDFL cells, isolated by the identical procedure used in Table 1, were reanalyzed after 7 days of culture in KL + GM-CSF. CD34 expression was clearly observed to be greater on the cultured CD4+ cells than on the cultured CD4 cells.

Close modal

In Δ-assays, the cellularity of cultures of CD4+ CD34++ CD38 Lin LDFL cells increased 1,000-fold over 7 days, whereas the cellularity of cultures of CD4 CD34++ CD38 Lin LDFL cells increased only 730-fold. The cultures of CD4+ CD34++ CD38 Lin LDFL cells gave rise to a 60-fold increase in HPP-CFC and a 430-fold increase in LPP-CFC, in contrast to the 9.6-fold increase in HPP-CFC and 130-fold increase in LPP-CFC measured from cultures of CD4 CD34++ CD38 Lin LDFL cells (P = .042 and .001, respectively). The ability of CD4+ progenitors to proliferate to a greater degree than CD4 progenitors was confirmed in 13 experiments in cultures maintained for up to 21 days in the presence of various growth stimuli.

Both CD4+ and CD4 fetal liver progenitors can reconstitute lymphoid and myeloid lineages in SCID-hu mice.Multilineage potential as well as the ability of progenitors to maintain long-term hematopoiesis were investigated using SCID-hu mice, which provide an in vivo model system for human hematopoiesis.23-26 The T-cell lineage potential of both the CD4+ and CD4 subsets of fetal liver progenitors was shown by the ability of 5.0 × 101 to 2.0 × 103 of these cells to give rise to T cells upon injection into human fetal thymic tissue implanted into SCID mice (Table 2). Immature CD4+ and CD8+ DP thymocytes of donor origin were observed in all tissues engrafted with either CD4 CD34++ Lin or CD4+ CD34++ Lin LDFL cells (Table 2 and Fig 4). Thus, precluding the possibility that engraftment was a consequence of contaminating single-positive (SP) T cells of fetal liver origin. Furthermore, in all cases of engraftment, CD4+ SP and CD8+ SP thymocytes with the donor HLA phenotype were observed. Interestingly, in most cases, the frequency of CD4+ SP and CD8+ SP cells was similar, in contrast to fresh fetal thymic tissue in which the number of CD4+ SP thymocytes exceeds the number of CD8+ SP thymocytes by twofold.27 In some transplants, as few as 50 cells of each fraction reconstituted thymic T lymphopoiesis for greater than 2 months. These results show that T-cell progenitors are present at high frequencies among both the CD4+ and the CD4 fractions of CD34++ Lin LDFL cells.

Table 2.

Thymic Engraftment in SCID-hu Mice by CD4 CD34++ Lin LDFL and CD4+ CD34++ Lin LDFL Progenitors

DonorProgenitor PopulationNo. InjectedDay of AnalysisFraction Engrafted*% DP% CD4+ SP% CD8+ SP
CD4 2.0 × 103 54 3/4 68.6-79.3 9.6-14.1 10.6-17.4 
CD4+ 2.0 × 103 54 1/1 59.9 19.3 20.9 
CD4 2.0 × 102 68 1/2 8.0 18.9 46.3 
CD4+ 2.0 × 102 68 3/3 58.7-90.1 4.5-16.0 3.6-20.0 
CD4 50 82 2/2 8.6-52.8 2.2-5.3 3.9-11.2 
CD4+ 50 82 1/1 56.4 4.1 2.6 
CD4+ 5.0 × 103 83 2/6 8.2-19.9 40.2-45.0 39.7-44.9 
CD4+ 5.0 × 103 95 2/3 62.9-83.4 7.3-15.5 8.4-18.7 
CD4+ 5.0 × 102 95 0/2 0  0  0  
CD4+ 50 95 1/2 80.3 9.1 10.1 
DonorProgenitor PopulationNo. InjectedDay of AnalysisFraction Engrafted*% DP% CD4+ SP% CD8+ SP
CD4 2.0 × 103 54 3/4 68.6-79.3 9.6-14.1 10.6-17.4 
CD4+ 2.0 × 103 54 1/1 59.9 19.3 20.9 
CD4 2.0 × 102 68 1/2 8.0 18.9 46.3 
CD4+ 2.0 × 102 68 3/3 58.7-90.1 4.5-16.0 3.6-20.0 
CD4 50 82 2/2 8.6-52.8 2.2-5.3 3.9-11.2 
CD4+ 50 82 1/1 56.4 4.1 2.6 
CD4+ 5.0 × 103 83 2/6 8.2-19.9 40.2-45.0 39.7-44.9 
CD4+ 5.0 × 103 95 2/3 62.9-83.4 7.3-15.5 8.4-18.7 
CD4+ 5.0 × 102 95 0/2 0  0  0  
CD4+ 50 95 1/2 80.3 9.1 10.1 
*

Engraftment of human fetal thymic tissue implanted in SCID mice by HLA-mismatched fetal liver progenitors was defined as the presence of donor-derived CD3+, CD4 SP, CD8 SP, and CD4 CD8 DP T cells as determined by FACScan analysis.

The data are presented as the range in the percentage of each subpopulation observed among total thymocytes expressing the donor HLA phenotype.

Fig. 4.

Engraftment of human fetal thymic and BM tissue implanted in SCID mice by CD4 CD34++ Lin LDFL and CD4+ CD34++ Lin LDFL progenitors. Generation of CD3+, CD4 SP, and CD8 SP and DP thymocytes by 50 HLA-A3 CD4+ CD34++ Lin LDFL cells injected into an HLA-A3+ thymic implant 95 days before analysis. Note the presence of a small (<1.9%) fraction of CD3+ HLA-A3+ host cells (A). Generation of CD34+, CD19+, and CD33+ cells by 200 HLA-A3+ CD4+ CD34++ Lin LDFL cells injected into human fetal bone fragments (HLA-A3) 125 days before analysis (B). The data are shown using 3% (A) and 2% (B) linear-density contour plots with 1 degree of smoothing. Analysis of CD4 and CD34 expression on Lin PI LDFL cells before (C, left) and after FACS (C, center and right). Ten thousand PI events were collected for the sort reanalysis shown. These sorted cells are donor B in Table 2 and donor D in Table 3.

Fig. 4.

Engraftment of human fetal thymic and BM tissue implanted in SCID mice by CD4 CD34++ Lin LDFL and CD4+ CD34++ Lin LDFL progenitors. Generation of CD3+, CD4 SP, and CD8 SP and DP thymocytes by 50 HLA-A3 CD4+ CD34++ Lin LDFL cells injected into an HLA-A3+ thymic implant 95 days before analysis. Note the presence of a small (<1.9%) fraction of CD3+ HLA-A3+ host cells (A). Generation of CD34+, CD19+, and CD33+ cells by 200 HLA-A3+ CD4+ CD34++ Lin LDFL cells injected into human fetal bone fragments (HLA-A3) 125 days before analysis (B). The data are shown using 3% (A) and 2% (B) linear-density contour plots with 1 degree of smoothing. Analysis of CD4 and CD34 expression on Lin PI LDFL cells before (C, left) and after FACS (C, center and right). Ten thousand PI events were collected for the sort reanalysis shown. These sorted cells are donor B in Table 2 and donor D in Table 3.

Close modal

The potential of CD4+ CD34++ Lin and CD4 CD34++ Lin LDFL cells to reconstitute BM hematopoiesis when injected into bone grafts implanted in SCID mice was investigated (Table 3). Full engraftment was defined by the presence of CD34+, CD19+, and CD33+ cells of donor HLA origin, as shown in Fig 4. Both CD4+ and CD4 fetal liver cells were observed to contain progenitors with myeloid and/or B-lymphoid potential. Furthermore, both the CD4+ and the CD4 subpopulations had considerable proliferative potential when transplanted into human bone fragments. A similar frequency of donor cell engraftment was observed in the human BM of SCID-hu mice injected with either 2.0 × 104 CD4+ or CD4 cells and analyzed 143 days after transplantation (Table 3, donor A). The discrepancy between these in vivo results and the stark differences in the proliferative capacities between CD4+ and CD4 cells in vitro lead us to hypothesize that the high number of cells injected into the SCID-hu mice may have interfered with the ability to discern any differences in proliferative potential between CD4+ and CD4 cells. Indeed, with the injection of a 10-fold fewer number of cells, a difference between the CD4+ and the CD4 fetal liver subsets was observed. Progenitor (CD34), B-lymphoid (CD19), and myeloid (CD33) engraftment, measured at about 3 months after the injection of 2.0 × 103 CD4+ cells, was observed in 3 of 4 SCID-hu mice receiving injections of donor B cells and in all 4 mice receiving injections of donor C cells, whereas injection of 2.0 × 103 CD4 cells from donor B gave rise to full engraftment in only 3 of 7 mice and no reconstitution was observed with CD4 cells derived from donor C. CD4+ cells were again found to be superior in reconstituting human hematopoiesis when only 2.0 × 102 cells were injected. Reanalysis of 10,000 of these sorted CD4+ and CD4 cells (Fig 4C) indicates that no contamination by the counterpart population occured with the injection of only 2.0 × 102 cells. Approximately 4 months after transplantation, full reconstitution was observed for 3 of 6 mice receiving injections of CD4+ cells, whereas full engraftment was not observed in the 7 mice receiving transplants of CD4 cells (Table 3, donor D). Taken together, these data show that CD4+ progenitors have, on average, a greater proliferative capacity than do CD4 progenitors, although both populations of cells can reconstitute multilineage hematopoiesis in vivo.

Table 3.

BM Engraftment in SCID-hu Mice by CD4 CD34++ Lin LDFL and CD4+ CD34++ Lin LDFL Cells

DonorProgenitor PopulationNo. InjectedDay of AnalysisFraction Engrafted3-150
CD34+CD19+CD33+
CD4 2.0 × 104 143 3/5 2/33-151 2/33-151 
CD4+ 2.0 × 104 143 5/7 4/43-151 4/43-151 
CD4 2.0 × 103 87-88 4/7 3/7 4/7 
CD4+ 2.0 × 103 87-88 3/4 3/4 3/4 
CD4 2.0 × 103 91 0/3 0/3 0/3 
CD4+ 2.0 × 103 91 4/4 4/4 4/4 
CD4 2.0 × 102 125 0/7 0/7 1/7 
CD4+ 2.0 × 102 125 3/6 3/6 4/6 
DonorProgenitor PopulationNo. InjectedDay of AnalysisFraction Engrafted3-150
CD34+CD19+CD33+
CD4 2.0 × 104 143 3/5 2/33-151 2/33-151 
CD4+ 2.0 × 104 143 5/7 4/43-151 4/43-151 
CD4 2.0 × 103 87-88 4/7 3/7 4/7 
CD4+ 2.0 × 103 87-88 3/4 3/4 3/4 
CD4 2.0 × 103 91 0/3 0/3 0/3 
CD4+ 2.0 × 103 91 4/4 4/4 4/4 
CD4 2.0 × 102 125 0/7 0/7 1/7 
CD4+ 2.0 × 102 125 3/6 3/6 4/6 
F3-150

Engraftment of human fetal bone tissue implanted in SCID mice was determined using HLA-mismatch between donor and host cells.

F3-151

Some samples were not analyzed due to an insufficient number of BM cells recovered.

The CD4 molecule has been shown to be expressed by mature cells and progenitors of various hematopoietic lineages.1,5,7,19 We have found CD4 to be expressed by a subset of human hematopoietic progenitors obtained from umbilical cord blood, fetal BM, and fetal liver. These observations are in agreement with recent reports showing low levels of CD4 expression on human CD34+ progenitors isolated from adult BM.19,20 

Although CD4 expression was detected on progenitors expressing both low and high levels of CD34, we focused our study on progenitors expressing the highest levels of CD34. These CD34++ cells have been shown by various means to contain primitive hematopoietic progenitors, including stem cells.21,25,29,31 In the human fetal liver, fetal BM, and umbilical cord blood, the highest levels of CD4 expression on progenitor cells was on those cells also expressing the highest levels of CD34. However, the levels of CD4 expression on CD34++ cells were lower than those found on CD4+ mature T cells. The phenotypic profile of CD4+ CD34++ Lin LDFL cells was similar to the profile suggested to represent fetal and adult pluripotent stem cells: CDw90+ (Thy-1),21,31,37 CD117+ (c-kit),21,31,38-41 CD7,27,28 CD38,21,29-31 CD45RA,21,32-34 CD71 (transferrin receptor),21,31,34 and Rh123dull.31,35,36 Indeed, the expression of CD4 on CD34++ Lin LDFL cells was found to be highest on cells with the lowest expression of CD7, CD38, CD45RA, and CD71. These observations are similar to those of Louache et al,20 who observed that CD4+ adult BM cells are CD38−/low, CD71low, and CDw90+. In summary, these data show the existence of a subpopulation of CD34++ fetal liver cells that is CD4+, CD13+, CD33+, CDw90+, CD117+, CD123+, CD7, CD38, CD45RA, CD71, CD115, and Rh123dull. Based on the phenotypic similarity of these cells to those previously described to be candidate stem cells and on the functional characteristics of CD4+ CD34++ Lin LDFL cells, it is highly likely that this cell population contains fetal hematopoietic stem cells.

However, a number of differences between the expression of cell surface markers on adult and fetal stem cells do appear to exist. Our previous studies on the expression of CD13 and CD33 by fetal liver progenitors suggested that these myeloid-associated antigens are expressed by fetal stem cells,21 in contrast to adult stem cells.42-44 The expression of CD33 by CD5+ CD2+ fetal thymocytes with T-cell and natural killer cell progenitor activity also indicated that CD33 expression is not limited to myeloid cells.45,46 The results reported here confirm this supposition, because all CD4+ CD34++ Lin LDFL cells expressed CD13 and CD33 and these cells were found to give rise to T, B, and myeloid cells in vivo. Although the expression of CD117 by early fetal progenitors is consistent with the distribution of this cytokine receptor on adult progenitors, the expression of the α-chain of the IL-3 receptor (CD123) on CD4+ CD34++ Lin LDFL cells is in contrast with the observation that candidate adult stem cells are refractory to IL-3.47 However, these antithetical findings are likely to represent functional differences between adult and fetal stem cells, because CD34++ CD38 Lin LDFL cells were shown to be responsive to IL-3.22 The expression of HLA-DR by hematopoietic stem cells may also change during human ontogeny. The CD4+ and CD4 subpopulations of CD34++ Lin LDFL cells differed only slightly in their expression of HLA-DR, with both subpopulations containing a minor fraction of HLA-DR cells and a major fraction of HLA-DR+ cells. We have previously failed to observe any difference between the in vitro proliferative potential of HLA-DR and HLA-DR+ fetal liver progenitors,21 whereas others have suggested that the fetal progenitors with the greatest proliferative potential reside among the HLA-DR+ fraction of fetal BM48 or umbilical cord blood cells.49 Conversely, numerous studies of adult hematopoietic progenitors favor the hypothesis that HLA-DR is not expressed by stem cells.50-52 Interestingly, in agreement with our findings on fetal liver cells, CD4+ adult BM progenitors were shown to be predominantly HLA-DR+.20 The observation that CD4 expression is conserved during human ontogeny suggests that this molecule plays an important role in hematopoiesis.

Hematopoietic stem cells are defined by their pluripotentiality and their extensive proliferative capacity. The results of in vitro and in vivo assays of these two functions indicated that most of the stem cell activity found in the human fetal liver was contained in the CD4+ subset. In vitro clonal assays showed that CD4+ CD34++ Lin LDFL cells are enriched for HPP-CFC activity relative to CD4 CD34++ Lin LDFL cells. Likewise, with the further enrichment of primitive progenitors by the isolation of CD34++ CD38 Lin cells,21 greater HPP-CFC activity was in the subset of these cells expressing CD4. More importantly, the Δ-assay showed that the CD4+ fraction consistently generated significantly greater numbers of secondary HPP-CFC than were generated by the CD4 fraction, a finding similar to the observed enrichment of LTC-IC among CD4+ adult BM progenitors.20 Erythroid potential was also observed in both the CD4+ and the CD4 subpopulations of CD34++ Lin LDFL cells. However, similar to the results using adult BM progenitors,19,20 greater numbers of BFU-E were measured in the CD4 fraction of fetal liver progenitors, suggesting a loss of CD4 during erythropoiesis.

Further evidence for the enrichment of stem cells among CD4+ cells was obtained using the SCID-hu mouse model. The incidence of BM engraftment in SCID-hu mice at least 3 months after the transplantation of CD4+ CD34++ Lin LDFL cells was more frequent than observed with CD4 CD34++ Lin LDFL cells. This difference became more evident when low numbers of cells were transplanted. Transplantation of 2 × 102 CD4+ progenitors led to 50% engraftment of progenitors, myeloid cells, and B cells, whereas no engraftment was observed with CD4 progenitors. The injection of 2 × 103 CD4+ progenitors resulted in an 88% rate of engraftment, and a 30% rate of engraftment was obtained with the transplantation of the same number of CD4 progenitors. However, when 2 × 104 cells were injected, a functional difference between the CD4+ and the CD4 progenitors was not apparent (71% v 60% engraftment, respectively). It is possible that a contamination of the CD4 cells by CD4+ cells may contribute to this reconstitution, because only 2.0 × 102 CD4+ cells were sufficient for reconstitution. Alternatively, the transplantation of a large number of CD4 cells may reduce the degree of proliferation these cells undergo in reconstituting the human bone fragments, thereby extending the lifetime of these grafts. These data show that human fetal liver cells are capable of reconstituting human fetal BM hematopoiesis in the in vivo environment of the SCID-hu mouse and that the BM-reconstituting cells are enriched among the fraction of CD4+ CD34++ Lin LDFL cells.

Both populations of CD4+ CD34++ Lin LDFL and CD4 CD34++ Lin LDFL progenitors gave rise to multilineage differentiation in vivo. Multilineage engraftment in bone-implanted SCID mice was indicated by the expression of CD33 (myeloid) and CD19 (B lymphoid). T-cell progenitor activity, indicated by the generation of DP and SP thymocytes, was observed in both the CD4+ and the CD4 subpopulations of CD34++ Lin LDFL cells using SCID-hu mice implanted with human thymic tissue. These results are consistent with our previously reported observation that 1 × 104 fetal liver progenitors, isolated by FACS based on the expression of CD34 and the lack of expression of a panel of lineage markers including CD4, could give rise to multilineage hematopoiesis in human fetal thymic organ cultures. These CD4 CD34+/++ Lin fetal liver cells were observed to give rise to myeloid, dendritic, B, natural killer, and T cells during culture in depleted fetal thymic fragments.10,27,45 A caveat with experiments measuring thymic or BM reconstitution is the inability to distinguish the multilineage engraftment due to pluripotent stem cells from that due to a mixture of lineage-restricted progenitors. Therefore, further studies are required to determine if cells in both the CD4+ and the CD4 fractions of CD34++ Lin fetal liver progenitors are capable of pluripotential differentiation.

In agreement with our findings, the expression of low levels of CD4 on murine stem cells has been shown,16,17 although two more recent reports have suggested that the most primitive murine stem cells reside among CD4 BM or fetal liver cells.13,14 Surprisingly, despite the expression of CD4 on murine hematopoietic progenitors13,14,16-18 and T-cell progenitors,53,54 mice lacking functional CD4 genes were not found to be grossly deficient in their lympho-hematopoietic capabilities.3 The functional significance of CD4 expression on cells other than mature CD4 SP T cells therefore remains to be determined. The expression of CD4 by human fetal hematopoietic progenitors does have implications towards our understanding of the biology of human immunodeficiency virus (HIV). Because CD4 is one of the receptors for HIV,55,56 our data imply that fetal hematopoietic stem cells may be infected by HIV in utero. This possibility is supported by the observation that adult CD4+ hematopoietic progenitors bound HIV gp120 via the CD4 antigen.19,20 Some studies have also found adult hematopoietic progenitors to be infected by HIV, although there is disagreement as to the incidence of these infections and the role of HIV-infected progenitors in the hematologic abnormalities observed in HIV-infected individuals.57-60 The discovery of human fetal hematopoietic tissues as a source of primitive CD4+ progenitors makes the role of these cells in normal and abnormal hematopoiesis amenable to further study.

We are indebted to E. Callas, J. Cupp, V. Hong, and D. Polakoff for help with cell sorting and to S. Antonenko and N. Carballido-Perrig for help in the construction and care of SCID-hu mice. We also thank A. Bárcena for her critical review of this manuscript.

DNAX Research Institute is supported by the Schering-Plough Corp.

Address reprint requests to Reiko Namikawa, MD, PhD, DNAX Research Institute of Molecular and Cellular Biology, 901 California Ave, Palo Alto, CA 94304-1104.

1
Ledbetter
JA
Evans
RL
Lipinski
M
Cunningham-Rundles
C
Good
RA
Herzenberg
LA
Evolutionary conservation of surface molecules that distinguish T lymphocyte helper/inducer and cytotoxic/suppressor subpopulations in mouse and man.
J Exp Med
153
1981
310
2
Doyle
C
Strominger
JL
Interaction between CD4 and class II MHC molecules mediates cell adhesion.
Nature
330
1987
256
3
Rahemtulla
A
Fung-Leung
WP
Schilham
MW
Kündig
TM
Sambhara
SR
Narendran
A
Arabian
A
Wakeham
A
Paige
CJ
Zinkernagel
RM
Miller
RG
Mak
TW
Normal development and function of CD8+ cells but markedly decreased helper cell activity in mice lacking CD4.
Nature
353
1991
180
4
Rand
TH
Cruikshank
WW
Center
DM
Weller
PF
CD4-mediated stimulation of human eosinophils: Lymphocyte chemoattractant factor and other CD4-binding ligands elicit eosinophil migration.
J Exp Med
173
1991
1521
5
Basch
RS
Kouri
YH
Karpatkin
S
Expression of CD4 by human megakaryocytes.
Proc Natl Acad Sci USA
87
1990
8085
6
Lucey
DR
Dorsky
DI
Nicholson-Weller
A
Weller
PF
Human eosinophils express CD4 protein and bind human immunodeficiciency virus gp120.
J Exp Med
169
1989
327
7
Wood
GS
Warner
NL
Warnke
RA
Anti-Leu-3/T4 antibodies react with cells of monocyte/macrophage and langerhans lineage.
J Immunol
131
1983
212
8
Hugo
P
Waanders
GA
Scollay
R
Shortman
K
Boyd
RL
Ontogeny of a novel CD4+ CD8− CD3− thymocyte subpopulation: A comparison with CD4− CD8+ CD3− thymocytes.
Int Immunol
2
1990
209
9
Hori
T
Cupp
J
Wrighton
N
Lee
F
Spits
H
Identification of a novel human thymocyte subset with a phenotype of CD3− CD4+ CD8 alpha + beta−. Possible progeny of the CD3− CD4− CD8− subset.
J Immunol
146
1991
4078
10
Galy
A
Verma
S
Bárcena
A
Spits
H
Precursors of CD3+ CD4+ CD8+ cells in the human thymus are defined by expression of CD34. Delineation of early events in human thymic development.
J Exp Med
178
1993
391
11
Kraft
DL
Weissman
IL
Waller
EK
Differentiation of CD3− 4− 8− human fetal thymocytes in vivo: Characterization of a CD3− 4+ 8− intermediate.
J Exp Med
178
1993
265
12
Wu
L
Antica
M
Johnson
GR
Scollay
R
Shortman
K
Developmental potential of the earliest precursor cells from the adult mouse thymus.
J Exp Med
174
1991
1617
13
Morrison
S
Weissman
I
The long-term repopulating subset of hematopoietic stem cells is deterministic and isolatable by phenotype.
Immunity
1
1994
661
14
Tocci
A
Rezzoug
F
Wahbi
K
Touraine
J-L
Fetal liver generates low CD4 hematopoietic cells in murine stromal cultures.
Blood
85
1995
1463
15
Fredrickson
GG
Basch
RS
L3T4 antigen expression by hemopoietic precursor cells.
J Exp Med
169
1989
1473
16
Onishi
M
Nagayoshi
K
Kitamura
K
Hirai
H
Takaku
F
Nakauchi
H
CD4dull+ hematopoietic progenitor cells in murine bone marrow.
Blood
81
1993
3217
17
Wineman
JP
Gilmore
GL
Gritzmacher
C
Torbett
BE
Muller
SC
CD4 is expressed on murine pluripotent hematopoietic stem cells.
Blood
80
1992
1717
18
Szilvassy
SJ
Cory
S
Phenotypic and functional characterization of competitive long-term repopulating hematopoietic stem cells enriched from 5-fluorouracil–treated murine marrow.
Blood
81
1993
2310
19
Zauli
G
Furlini
G
Vitale
M
Re
M-C
Gibellini
D
Zamai
L
Visani
G
Borgatti
P
Capitani
S
La Placa
M
A subset of human CD34+ hematopoietic progenitors express low levels of CD4, the high-affinity receptor for human immunodeficiency virus-type 1.
Blood
84
1994
1896
20
Louache
F
Debili
N
Marandin
A
Coulombel
L
Vainchenker
W
Expression of CD4 by human hematopoietic progenitors.
Blood
84
1994
3344
21
Muench
MO
Cupp
J
Polakoff
J
Roncarolo
MG
Expression of CD33, CD38 and HLA-DR on CD34+ human fetal liver progenitors with a high proliferative potential.
Blood
83
1994
3170
22
Muench
MO
Roncarolo
MG
Menon
S
Xu
Y
Kastelein
R
Zurawski
S
Hannum
CH
Culpepper
J
Lee
F
Namikawa
R
FLK-2/FLT-3 ligand (FL) regulates the growth of early myeloid progenitors isolated from human fetal liver.
Blood
85
1995
963
23
McCune
JM
Namikawa
R
Kaneshima
H
Shultz
LD
Lieberman
M
Weissman
IL
The SCID-hu mouse: Murine model for the analysis of human hematolymphoid differentiation and function.
Science
241
1988
1632
24
Kyoizumi
S
Baum
CM
Kaneshima
H
McCune
JM
Yee
EJ
Namikawa
R
Implantation and maintenance of functional human bone marrow in SCID-hu mice.
Blood
79
1992
1704
25
Di Giusto
D
Chen
S
Combs
J
Webb
S
Namikawa
R
Tsukamoto
A
Chen
BP
Galy
AHM
Human fetal bone marrow early progenitors for T, B, and myeloid cells are found exclusively in the population expressing high levels of CD34.
Blood
84
1994
421
26
Chen
BP
Galy
A
Kyoizumi
S
Namikawa
R
Scarborough
J
Webb
S
Ford
B
Cen
D-Z
Chen
SC
Engraftment of human hematopoietic precursor cells with secondary transfer potential in SCID-hu mice.
Blood
84
1994
2497
27
Bárcena
A
Muench
MO
Galy
AHM
Cupp
J
Roncarolo
M-G
Phillips
JH
Spits
H
Phenotypic and functional analysis of T-cell precursors in the human fetal liver and thymus: CD7 expression in the early stages of T- and myeloid-cell development.
Blood
82
1993
3401
28
Bárcena
A
Muench
MO
Roncarolo
MG
Spits
H
Tracing the expression of CD7 and other antigens during T- and myeloid-cell differentiation in the human fetal liver and thymus.
Leuk Lymphoma
17
1995
1
29
Terstappen
LWMM
Huang
S
Safford
M
Lansdorp
PM
Loken
MR
Sequential generation of hematopoietic colonies derived from single nonlineage-committed CD34+CD38− progenitor cells.
Blood
77
1991
1218
30
Issaad
C
Croisille
L
Katz
A
Vainchenker
W
Coulombel
L
A murine stromal cell line allows the proliferation of very primitive human CD34++/CD38− progenitor cells in long-term cultures and semisolid assays.
Blood
81
1993
2916
31
Craig
W
Kay
R
Cutler
RL
Lansdorp
PM
Expression of Thy-1 on human hematopoietic progenitor cells.
J Exp Med
177
1993
1331
32
Lansdorp
PM
Sutherland
HJ
Eaves
CJ
Selective expression of CD45 isoforms on functional subpopulations of CD34+ hemopoietic cells from human bone marrow.
J Exp Med
172
1990
363
33
Fritsch
G
Buchinger
P
Printz
D
Fink
FM
Mann
G
Peters
C
Wagner
T
Adler
A
Gadner
H
Rapid discrimination of early CD34+ myeloid progenitors using CD45-RA analysis.
Blood
81
1993
2301
34
Lansdorp
PM
Dragowska
W
Long-term erythropoiesis from constant numbers of CD34+ cells in serum-free cultures initiated with highly purified progenitor cells from human bone marrow.
J Exp Med
175
1992
1501
35
Chaudhary
PM
Roninson
IB
Expression and activity of P-glycoprotein, a multidrug effluc pump, in human hematopoietic stem cells.
Cell
66
1991
85
36
Udomsakdi
C
Eaves
CJ
Sutherland
HJ
Landsdorp
PM
Separation of functionally distinct subpopulations of primitive human hematopoietic cells using rhodamine-123.
Exp Hematol
19
1991
338
37
Baum
CM
Weissman
IL
Tsukamoto
AS
Buckle
AM
Peault
B
Isolation of a candidate human hematopoietic stem-cell population.
Proc Natl Acad Sci USA
89
1992
2804
38
Okada
S
Nakauchi
H
Nagayoshi
K
Nishikawa
S
Nishikawa
S
Miura
Y
Suda
T
Enrichment and characterization of murine hematopoietic stem cells that express c-kit molecule.
Blood
78
1991
1706
39
Okada
S
Nakauchi
H
Nagayoshi
K
Nishikawa
S
Miura
Y
Suda
T
In vivo and in vitro stem cell function of c-kit– and Sca-1–positive murine hematopoietic cells.
Blood
80
1992
3044
40
de Vries
P
Brasel
KA
McKenna
HJ
Williams
DE
Watson
JD
Thymus reconstitution by c-kit-expressing hematopoietic stem cells purified from adult mouse bone marrow.
J Exp Med
176
1992
1503
41
Simmons
PJ
Aylett
GW
Niutta
S
To
LB
Juttner
CA
Ashman
LK
c-kit is expressed by primitive human hematopoietic cells that give rise to colony-forming cells in stroma-dependent or cytokine-supplemented culture.
Exp Hematol
22
1994
157
42
Andrews
RG
Singer
JW
Bernstein
ID
Precursors of colony-forming cells in humans can be distinguished from colony-forming cells by expression of the CD33 and CD34 antigens and light scatter properties.
J Exp Med
169
1989
1721
43
La Russa
VF
Giffin
JD
Kessler
SW
Cutting
MA
Knight
RD
Blattler
WA
Lambert
JM
Wright
DG
Effects of anti-CD33 blocked ricin immunotoxin on the capacity of CD34+ human marrow cells to establish in vitro hematopoiesis in long-term marrow cultures.
Exp Hematol
20
1992
442
44
Shipp
MA
Look
AT
Hematopoietic differentiation antigens that are membrane-associated enzymes: Cutting is the key!
Blood
82
1993
1052
45
Bárcena
A
Galy
AHM
Punnonen
J
Muench
MO
Schols
D
Roncarolo
MG
de Vries
JE
Spits
H
Lymphoid and myeloid differentiation of fetal liver CD34+ lineage− cells in human thymic organ culture.
J Exp Med
180
1994
123
46
Sánchez
MJ
Muench
MO
Roncarolo
MG
Lanier
LL
Phillips
JH
Identification of a common T/NK cell progenitor in the human fetal thymus.
J Exp Med
180
1994
569
47
Berardi
AC
Wang
A
Levine
JD
Lopez
P
Scadden
DT
Functional isolation and characterization of human hematopoietic stem cells.
Science
267
1995
104
48
Huang
S
Terstappen
LWMM
Lymphoid and myeloid differentiation of single human CD34+, HLA-DR+, CD38− hematopoietic cells.
Blood
83
1994
1515
49
Traycoff
CM
Abboud
MR
Laver
J
Brandt
JE
Hoffman
R
Law
P
Ishizawa
L
Srour
EF
Evaluation of the in vitro behavior of phenotypically defined populations of umbilical cord blood hematopoietic progenitor cells.
Exp Hematol
22
1994
215
50
Moore
MAS
Broxmeyer
HE
Sheridan
APC
Meyers
PA
Jacobsen
N
Winchester
RJ
Continuous human bone marrow culture: Ia antigen characterization of probable pluripotential stem cells.
Blood
55
1980
682
51
Keating
A
Powell
J
Takahashi
M
Singer
JW
The generation of human long-term marrow cultures from bone marrow depleted of Ia (HLA-DR) positive cells.
Blood
64
1984
1159
52
Verfaillie
C
Blakomer
K
McGlave
P
Purified primitive human hematopoietic progenitor cells with long-term in vitro repopulating capacity adhere selectively to irradiated bone marrow stroma.
J Exp Med
172
1990
509
53
Wu
L
Scollay
R
Egerton
M
Pearse
M
Spangrude
GJ
Shortman
K
CD4 expressed on earliest T-lineage precursor cells in the adult murine thymus.
Nature
349
1991
71
54
Chervenak
R
Dempsey
D
Soloff
R
Wolcott
RM
Jennings
SR
The expression of CD4 by T cell precursors resident in both the thymus and the bone marrow.
J Immunol
151
1993
4486
55
Dalgleish
AG
Beverley
PCL
Clapham
PR
Crawford
DH
Greaves
MF
Weiss
RA
The CD4 (T4) antigen is an essential component of the receptor for the AIDS retrovirus.
Nature
312
1984
763
56
Klatzmann
D
Champagne
E
Chamaret
S
Gruest
J
Guetard
D
Hercend
T
Gluckman
J-C
Montagnier
L
T-lymphocyte T4 molecule behaves as the receptor for human retrovirus LAV.
Nature
312
1984
767
57
Chelucci
C
Hassan
HJ
Locardi
C
Bulgarini
D
Pelosi
E
Mariani
G
Testa
U
Federico
M
Valtieri
M
Peschle
C
In vitro human immunodeficiency virus-1 infection of purified hematopoietic progenitors in single-cell culture.
Blood
85
1995
1181
58
Stanley
SK
Kessler
SW
Justement
JS
Schnittman
SM
Greenhouse
JJ
Brown
CC
Musongela
L
Musey
K
Kapita
B
Fauci
AS
CD34+ bone marrow cells are infected with HIV in a subset of seropositive individuals.
J Immunol
149
1992
689
59
von Laer
D
Hufert
FT
Fenner
TE
Schwander
S
Dietrich
M
Schmitz
H
Kern
P
CD34+ hematopoietic progenitor cells are not a major reservoir of the human immunodeficiency virus.
Blood
76
1990
1281
60
Molina
J-M
Scadden
DT
Sakaguchi
M
Fuller
B
Woon
A
Groopman
JE
Lack of evidence for infection of or effect on growth of hematopoietic progenitor cells after in vivo or in vitro exposure to human immunodeficiency virus.
Blood
76
1990
2476
Sign in via your Institution