Recently, a collection of surface markers was exploited to isolate viable Lin TdT+ cells from murine bone marrow. These early pro-B cells were enriched for B-lineage lymphocyte precursor activity measured by short-term culture and had little responsiveness to myeloid growth factors. Early precursors can be propagated with remarkably high cloning frequencies in stromal cell–free, serum-free cultures, permitting this analysis of direct regulatory factors. Expression of the interleukin-7 receptor (IL-7Rα) chain marks functional precursors and IL-7 is necessary for progression beyond the CD45RA+ CD19 stage. Efficient survival and differentiation were only observed when stem cell factor and Flt-3 ligand were also present. IL-7–responsive CD19+precursors are estrogen resistant. However, B-lineage differentiation was selectively abrogated when highly purified Lin precursors were treated with hormone in the absence of stromal cells. In addition, early stages of B lymphopoiesis were arrested by limitin, a new interferon (IFN)–like cytokine as well as IFN-α, IFN-γ, or transforming growth factor β (TGF-β), but not by epidermal growth factor (EGF). Lin TdT+early pro-B cells are shown here to be CD27+AA4.1+/−Ki-67+ Ly-6CLy-6A/Sca-1Lo/−Thy-1CD43+CD4+/−CD16/32Lo/−CD44Hi and similar in some respects to the “common lymphoid progenitors” (CLP) identified by others. Although early pro-B cells have lost myeloid differentiation potential, transplantation experiments described here reveal that at least some can generate T lymphocytes. Of particular importance is the demonstration that a pivotal early stage of lymphopoiesis is directly sensitive to negative regulation by hormones and cytokines.

The humoral immune system is replenished throughout life with B cells produced by the bone marrow. This process is being increasingly understood in terms of selective gene expression with transcription factors playing a major role in determining how hematopoietic stem cells adopt one of 8 major fates. However, much remains to be learned about the timing of events and how they are influenced by positive and negative extracellular cues provided by the microenvironment. Multiparameter flow cytometry and cell sorting allows isolation of rare populations of bone marrow cells and it is important to order them in a differentiation sequence that culminates with the production of B lymphocytes. Expression of the enzyme terminal deoxynucleotidyl transferase (TdT) is thought to be an early, lymphocyte-specific event with functional consequences.1TdT contributes to the diversity of antibody combining sites by adding non–germline-encoded nucleotides at sites of V-DJ segment joining during immunoglobulin gene recombination.2,3 We recently developed a means to identify and sort TdT+ bone marrow cells lacking surface markers associated with any of the mature blood cell lineages (Lin).4,5 According to tradition, they were termed “early pro-B” cells and they were highly enriched for functional B-lymphocyte lineage precursor activity. That is, early pro-B cells gave rise to CD45R+CD19 and CD19+ lymphocytes but generated few nonlymphoid cells on contact with stromal cells. These precursors comprise approximately 0.06% of the nucleated cells in marrow and may represent an important developmental compartment that must be transited by differentiating stem cells. We now report that early pro-B cells expand and differentiate under defined, stromal cell–free culture conditions. This model made it possible to examine the importance of functional interleukin-7 (IL-7) receptors (IL-7R) and other factors for their survival, proliferation, and lineage progression. Early pro-B cells were directly sensitive to factors previously described to be negative regulators of B lymphopoiesis in intact animals.

Some of the early pro-B cell characteristics are similar to ones previously used by another group to isolate “common lymphoid progenitors” (CLP).6 At least 20% of Lin Sca-1+ IL-7Rα+ CLP could expand in culture and single cells had the potential for T, B, and natural killer (NK) cell differentiation. Stem cells with long-term repopulating potential can be distinguished from hematopoietic progenitors by the absence of CD27 together with expression of c-kit and Sca-1.7 Still other markers have been exploited to isolate categories of marrow cells with restricted potential for differentiation in nonlymphoid lineages.8 One objective of the present study was to determine features of LinTdT+ cells that would allow more direct comparison to CLP and other progenitor subsets. In addition to CLP, populations designated common myeloid precursors and fraction A0 pre pro-B cells were of interest.8,9 We also assessed the potential of early pro-B cells for T-lymphocyte lineage differentiation in transplantation experiments. The molecular and cellular complexity of the bone marrow was experimentally reduced with the objective of learning when differentiation fate decisions are made and how they are regulated by extracellular signals.

Mice

BALB/c mice were obtained at 5 to 15 weeks of age from the Oklahoma Medical Research Foundation Laboratory Animal Resource Center or Charles Rivers Breeding Laboratories (Wilmington, ME) and used for culture experiments. C57BL/6 mice were obtained from Charles Rivers or the Jackson Laboratory (Bar Harbor, ME) and used for surface marker characterization of Lin TdT+ cells. RAG-1−/− mice (5-10 weeks old) and 5- to 10-week-old C57BL/6 SJL mice (Ly5.1 background) were purchased from the Jackson Laboratory and used in transfer experiments.

Antibodies

Anti-CD19 monoclonal antibody (mAb) (1D3) was purified from the culture supernatant of hybridoma cells grown in our laboratory. Anti-CD45RA (14.8) mAb developed in our laboratory and the anti–Mac-1/CD11b (M1/70) mAb were used as culture supernatants of the respective hybridomas. The rat mAb to mouse IL-7Rα subunit (SB199) was established in our laboratory, purified from hybridoma supernatant and biotinylated.10Phycoerythrin (PE)-conjugated and allophycocyanin (APC)-conjugated antibodies to AA4.1 antigen were kindly provided by Dr R. R. Hardy (Fox Chase Cancer Center, Philadelphia, PA). Purified antierythroid (Ter-119) and anti–Ly6-G (Gr-1; RB6-8C5) mAbs, fluorescein isothiocyanate (FITC)–conjugated anti-CD3, (145-2C11) anti-CD8, (53-6.7) anti-CD45R/B220 (RA3/6B2), anti–Mac-1 (M1/70), and anti–Gr-1 (Ly-6G; RB6-8C5) antibodies, PE-conjugated Ter-119, anti-CD45R/B220 (RA3/6B2), anti-Flk2/Flt3 (A2F10.1), anti-CD19, (1D3) anti–Sca-1 (Ly6A/E; E13-161.7), anti-CD16/CD32 (2.4G2) and anti-Ki67 (B56) antibodies, biotinylated anti-CD27 (LG3A10) antibody and APC-conjugated anti–c-kit, anti-CD8, anti-CD45R, anti–Mac-1 antibodies were all purchased from BD Pharmingen (San Diego, CA). Purified rabbit anti-TdT polyclonal antibody and a FITC-conjugated F(ab′)2 fraction of goat antirabbit IgG antibody were purchased from Supertechs (Rockville, MD). FITC-conjugated goat antimouse IgM antibody was purchased from Zymed (San Francisco, CA).

Reagents

Recombinant mouse IL-7 was purchased from Endogen (Woburn, MA). Recombinant mouse stem cell factor (SCF), Flt-2/Flk-3 ligand (FL), and transforming growth factor-β (TGF-β) were purchased from R&D Systems (Minneapolis, MN). 1, 3, 5[10]-Estratriene-3, 17β-diol (β-estradiol) was purchased from Sigma Chemical (St Louis, MO). Epidermal growth factor (EGF) was purchased from Upstate Biotechnology (Lake Placid, NY). Mouse interferon (IFN)-α and rat IFN-γ were purchased from Life Technologies (Rockville, MD). Limitin-human immunoglobulin (IgG1) heavy-chain constant region fusion protein was prepared as previously described.11 

Isolation of lineage-negative early pro-B cells

Bone marrow cells were collected from 4 to 10 mice and suspended with phosphate-buffered saline (PBS) without Ca++ or Mg++ (PBS) and supplemented with 3% heat-inactivated fetal calf serum (staining wash). Cells were incubated with antibodies to lineage markers (Gr-1 and Mac-1 for myeloid cells, anti-CD19 and anti-CD45R for B lineage cells, and Ter-119 for erythroid cells) for 30 minutes. Then cells were washed and incubated with BioMag goat-antirat IgG-coated magnetic beads (Polysciences, Warrington, PA) for 15 minutes. Cells attached to beads were removed with a magnetic separator. Recovered cells were further incubated with new magnetic beads and subjected to a second round of magnetic separation. These enriched lineage marker–negative bone marrow cells were then stained with a cocktail of labeled antibodies to the lineage markers (FITC-conjugated anti-CD3, anti-CD8, anti–Gr-1, PE-conjugated Ter-119 and anti-CD45R) and APC-conjugated anti–c-kit antibody. In some experiments, PE-labeled anti-FL antibody and biotinylated anti–IL-7Rα antibody were also used. In this case, PE-labeled Ter-119 was eliminated and FITC-conjugated anti-CD45R was used instead of PE-conjugated antibody. Streptavidin-RED613 (Life Technologies) was used as the secondary reagent for biotinylated anti–IL-7Rα. Stained cells were subjected to sorting on a MoFlo (Cytomation, Fort Collins, CO). When reanalyzed after sorting, lineage-negative cells were generally more than 95% pure; that is, less than 5% had even slightly above background levels of staining. Subsets of these Lincells were usually more than 90% pure when sorted according to absence, low density, or high density of c-kit. The IL-7Rα+ cells were relatively homogenous, but the IL-7Rα preparations included some cells with low, but above threshold staining.

Cellculture

Sorted cells were put into 24-well culture plates (Costar, Cambridge, MA) containing 1 mL X-VIVO15 medium (Biowhittaker, Walkersville, MD) containing 1% detoxified bovine serum albumin (Stem Cell Technologies, Vancouver, BC, Canada), 5 × 10−5 M 2-mercaptoethanol (2-ME), 2 mM l-glutamine, 100 U/mL penicillin, 100 mg/mL streptomycin, and cytokines as indicated and cultured at 37°C and 5% CO2 in a humidified atmosphere. At the end of culture, cells were harvested, cell viability was determined with a trypan blue dye exclusion method, and then cells were subjected to flow cytometric analysis. For single-cell sorting experiments, cells were directly sorted into wells of 96-well U-bottom tissue culture plates (Costar) containing 100 μL of the same media. The concentrations of cytokines were IL-7, 1 ng/mL; FL,100 ng/mL; and SCF, 20 ng/mL.

Colony assay for granulocyte-macrophage progenitors

Sorted cells were put into methylcellulose medium containing murine IL-3 (mIL-3), human IL-6 (hIL-6), and murine SCF (mSCF) (MethoCult GF M3534, Stem Cell Technologies) with 200 to 1000 cells plated per dish. After 10 days of culture in a humidified CO2 incubator at 37°C, numbers of colonies were counted with an inverted microscope.

Immunofluorescence staining

For the analysis of surface antigens, cells were incubated with combinations of labeled antibodies in PBS. They were then washed and incubated with streptavidin-RED613 to detect biotinylated primary antibodies. To characterize TdT+ cells, sorted lineage-negative cells were first stained using PE-conjugated, APC-conjugated, or biotinylated antibodies in combination with streptavidin-RED613. Then cells were fixed with 1% formaldehyde in 1.25 × PBS followed by permeabilization with 70% ethanol. Then, intranuclear TdT was stained using anti-TdT polyclonal rabbit antibody and the FITC-conjugated F(ab′)2 fraction of a goat antirabbit IgG antibody. PE-conjugated anti–Ki-67 antibody was included at this step in some experiments. Stained cells were run on a FACScalibur flow cytometer (Becton Dickinson, San Diego, CA) and the data were analyzed with Flojo software (Treestar, San Carlos, CA).

In vivo transfer

Lineage marker–negative cells from C57BL/6 SJL mice were sorted to isolate the c-kitLo Flk-2/Flt-3+IL-7Rα+ fraction. One thousand to 3000 of these early pro-B cells were injected into RAG1−/− mice that had been given 400 rad irradiation. Spleens and thymuses were harvested from these mice 2 or 3 weeks after injection and stained for flow cytometric analysis.

Three cytokines permit survival and differentiation of early lymphocyte precursors

Jacobsen and colleagues reported the production of CD45R/B220+ and then CD19+ lymphocytes in serum-free cultures supported only by recombinant cytokines.12 They used magnetic beads and sorting to enrich for c-kit+ Sca-1+ cells with low expression of lineage markers and their focus was on investigating roles played by SCF and Flt-3 ligand. We now report that their stromal cell–free conditions are also ideal for B lymphopoiesis in cultures initiated with the TdT+ cell-enriched Linc-kitLo fraction of marrow, that is, early pro-B cells. For initial experiments, bone marrow cell suspensions were rigorously depleted of lineage marker–positive (GR-1, CD11b/Mac-1, CD45RA, CD19, TER119, CD3, and CD8) cells and then separated into 3 fractions on the basis of c-kit density.4 At least one third of the cells in the c-kitHi population were myeloid precursors that proliferated in a colony-forming unit in culture (CFU-c) assay (as described in “Materials and methods”). However, small numbers of CD19+ cells were produced during a 7-day culture with SCF, FL, and IL-7 (Figure 1). Although essentially no myeloid progenitors were present in the c-kitLo subset, it produced at least 10 times more (up to 800/input cell) CD19+ B-lineage progeny in culture. Consistent with our previous study,4 small numbers of rapidly differentiating lymphocyte precursors were present in a fraction that completely lacks c-kit. As with stromal cell cocultures, the time required under defined conditions for generation of CD19+ cells from Lin c-kitHicells was longer than when the cultures were initiated with Lin c-kitLo/− cells (data not shown). Indeed, down-regulation of this SCF receptor coincides with loss of myeloid differentiation potential and progression through the early steps of lymphopoiesis.4 Defined culture models of this kind allow detailed investigation of the importance and signals transmitted via such receptors.

Fig. 1.

B-lineage lymphocytes arise from early precursors under defined culture conditions.

Sorted Lin c-kitHi (7500 cells/well), Lin c-kitLo (3000 cells/well), or Lin c-kit cells (10 000 cells/well) were placed in serum-free, stromal cell-free cultures. After 7 days cells were collected, counted, and analyzed by flow cytometry. Each dot represents absolute numbers of CD19+ cells per input in a single experiment. This was determined by simply dividing numbers of recovered lymphocytes by the numbers of precursors used to initiate the cultures.

Fig. 1.

B-lineage lymphocytes arise from early precursors under defined culture conditions.

Sorted Lin c-kitHi (7500 cells/well), Lin c-kitLo (3000 cells/well), or Lin c-kit cells (10 000 cells/well) were placed in serum-free, stromal cell-free cultures. After 7 days cells were collected, counted, and analyzed by flow cytometry. Each dot represents absolute numbers of CD19+ cells per input in a single experiment. This was determined by simply dividing numbers of recovered lymphocytes by the numbers of precursors used to initiate the cultures.

Close modal

A requirement for IL-7 and its receptor

Gene-targeting experiments have established the importance of IL-7 for B lymphopoiesis in mice and expression of the IL-7R must represent an important differentiation milestone.13,14 Therefore, this aspect was investigated under defined culture conditions. At least some CD45R+ cells were produced from Linc-kitLo precursors in 1-week cultures established with any of several cytokine combinations, but CD19+ cells emerged only when IL-7 was present (Figure 2A). IL-7 was also needed for production of CD19+ cells from cultures started with Lin c-kit precursors (Figure 2B). Additional experiments confirmed that the combination of SCF, FL, and IL-7 was optimal and almost no cells were recovered from cultures maintained with IL-7 alone. The LinFlk-2/Flt-3+ c-kitLo population was then sorted into subsets on the basis of IL-7Rα expression and cultured (Figure3). The starting IL-7RαLo/− category included 9% to 37% TdT+ cells, whereas 25% to 72% of the IL-7Rα+ fraction expressed TdT. CD19+ cells were produced when cultures were initiated with IL-7RαLo/− precursors, but less efficiently than in cultures of IL-7Rα+ cells. Single LinFlk-2/Flt-3+ c-kitLo IL-7Rα+cells were then placed in 96 individual culture wells along with SCF, FL, and IL-7 in 2 experiments. After 11 days, growth was observed in 25 or 29 wells and 20 or 25 of them contained CD19+lymphocytes, respectively. The cloning efficiency was approximately 6-fold lower in a comparable experiment done with single Lin Flk-2/Flt-3+ c-kitLoIL-7RαLo/− cells. When considered with the results shown above, these findings indicate that 3 functional cytokine receptors cooperate to deliver signals for survival, proliferation, or differentiation of early lymphocyte precursors, but IL-7 is particularly important for transition to the CD19+stage.

Fig. 2.

IL-7 is required for progression from the CD45R+CD19 stage to the CD45R+CD19+ stage.

Linc-kitLo (A) or Linc-kit cells (B) were sorted to high purity and placed in serum-free, stromal cell–free cultures with the indicated combinations of cytokines. Yields of CD45R+CD19 (▪) and CD45R+CD19+ (▨) cells per input precursor were calculated as described in the legend to Figure 1.

Fig. 2.

IL-7 is required for progression from the CD45R+CD19 stage to the CD45R+CD19+ stage.

Linc-kitLo (A) or Linc-kit cells (B) were sorted to high purity and placed in serum-free, stromal cell–free cultures with the indicated combinations of cytokines. Yields of CD45R+CD19 (▪) and CD45R+CD19+ (▨) cells per input precursor were calculated as described in the legend to Figure 1.

Close modal
Fig. 3.

Acquisition of IL-7R corresponds with rapid B-lineage differentiation in culture.

Cultures were initiated with Lin c-kitLoFlk-2/Flt-3+ IL-7Rα+ (♦) or Lin c-kitLo Flk-2/Flt-3+IL-7RαLo/− (■) cells and evaluated at the indicated intervals. Numbers of CD19+ lymphocytes recovered per input precursor are shown from one of 3 independent experiments that gave very similar results.

Fig. 3.

Acquisition of IL-7R corresponds with rapid B-lineage differentiation in culture.

Cultures were initiated with Lin c-kitLoFlk-2/Flt-3+ IL-7Rα+ (♦) or Lin c-kitLo Flk-2/Flt-3+IL-7RαLo/− (■) cells and evaluated at the indicated intervals. Numbers of CD19+ lymphocytes recovered per input precursor are shown from one of 3 independent experiments that gave very similar results.

Close modal

Early lymphocyte precursors are directly influenced by negative regulators

A variety of evidence suggests that lymphopoiesis in the thymus and bone marrow is limited by sex steroids, but the nature of the target cells remains elusive.15 Linc-kitHi cells with the potential for lymphoid or myeloid differentiation were placed in serum-free, stromal cell–free cultures containing only SCF, FL, and IL-7 (Figure4). Although total numbers of Gr-1+ myeloid progeny were slightly increased by inclusion of β-estradiol in the medium (1.1- to 2.5-fold), production of CD19+ lymphocytes was almost completely prevented. Titration experiments indicated that 10−8 M concentrations of the hormone were sufficient for maximum suppression (not shown). The next putative stage of differentiation also appeared to be directly sensitive to estrogen. That is, the ability of Linc-kitLo or Lin c-kit marrow cells to generate CD19+ lymphocytes in 1-week cultures was suppressed 78% ± 11% (Table 1) or 92% ± 3%, respectively, by β-estradiol. Interestingly, absolute numbers of CD45R+ CD19 cells were actually increased in estrogen- containing cultures as compared to control cultures (Table 1). The same accumulation of CD45R+CD19 cells (168.2% ± 34.8% of controls) occurred in estrogen-treated cultures initiated with Linc-kitHi precursors. We previously found that estrogen had no influence on the IL-7–driven clonal proliferation of lymphoid progenitors in semisolid agar cultures.16 Indeed, the same hormone preparations found here to be suppressive for early stages of lymphopoiesis were ineffective in CFU–IL-7 assays (not shown). Although late events in B lymphopoiesis can be influenced by estrogen only via effects on stromal cells, early precursors may be direct hormone targets.16 

Fig. 4.

Early lymphocyte precursors are directly and selectively inhibited by estrogen in culture.

Lin c-kitHi BM cells were placed in serum-free, stromal cell-free cultures with or without β-estradiol (10−8M). Gr-1+ myeloid and CD19+lymphoid cells determined by flow cytometry are shown after 7 days of culture.

Fig. 4.

Early lymphocyte precursors are directly and selectively inhibited by estrogen in culture.

Lin c-kitHi BM cells were placed in serum-free, stromal cell-free cultures with or without β-estradiol (10−8M). Gr-1+ myeloid and CD19+lymphoid cells determined by flow cytometry are shown after 7 days of culture.

Close modal
Table 1.

Early lymphocyte precursors are direct targets for negative regulation by hormones and cytokines

Factor (concentration)CD45R+,
CD19cells
CD45R+,
CD19+ cells
β-Estradiol (10−8 M)* 239.8 ± 208.7 22.0 ± 11.0  
Limitin (200 ng/mL) 5.6, 4.7 1.2, 0.1 
Interferon-α (10 U/mL) 14.9, 3.3 5.9, 4.2 
Interferon-γ (10 U/mL) 19.5, 17.8 <0.1, 0.3  
TGF-β (1 ng/mL) 8.3, 9.0 11.0, 9.1  
EGF (50 ng/mL) 94.1, 62.9 82.2, 87.2 
Factor (concentration)CD45R+,
CD19cells
CD45R+,
CD19+ cells
β-Estradiol (10−8 M)* 239.8 ± 208.7 22.0 ± 11.0  
Limitin (200 ng/mL) 5.6, 4.7 1.2, 0.1 
Interferon-α (10 U/mL) 14.9, 3.3 5.9, 4.2 
Interferon-γ (10 U/mL) 19.5, 17.8 <0.1, 0.3  
TGF-β (1 ng/mL) 8.3, 9.0 11.0, 9.1  
EGF (50 ng/mL) 94.1, 62.9 82.2, 87.2 

TGF-β indicates transforming growth factor β; EGF, epidermal growth factor.

*

Highly purified Lin c-kitLo cells were held for 1 week in serum-free, stromal cell-free cultures. Yields of CD45R+CD19+ and CD45R+CD19 cells per input precursor were calculated and then normalized as percentages of control values. These data represent means ± SD from 5 independent experiments.

Results from each of two experiments are separated by commas.

B lymphopoiesis was preferentially suppressed when limitin, a new IFN-like cytokine, was injected into mice.11 This factor dramatically reduced the yield of CD19+ cells in 1-week stromal cell-free cultures and, in contrast to estrogen, also reduced numbers of CD45R+ CD19 cells (Table 1). This was true when the cultures were initiated with either Linc-kitHi marrow cells or the more B-lineage precursor-rich Lin c-kit fraction (not shown). Gene-targeted mice and binding assays were used to determine that limitin uses the type I IFN receptor11 and IFN-α is also known to diminish B lymphopoiesis in vivo.17 Therefore, we performed a comparison between limitin and conventional IFNs in defined cultures (Table 1). IFN-α, IFN-γ, and limitin were equivalent in suppressing the production of CD45R+ CD19+cells from early precursors. In contrast to estrogen, the IFN-like cytokines also prevented the generation of CD45R+CD19 cells. Therefore, the B-lymphocyte lineage is also a direct target for negative regulation by these cytokines.

Transforming growth factor-β represents another previously studied inhibitor, but one with the potential for limiting myeloid as well as lymphoid differentiation.18 We now show that both the expansion and lymphoid lineage differentiation of Linprecursors are sensitive to TGF-β, using a neutralizing antibody to confirm specificity (Table 1 and data not shown). Although some evidence indicates that EGF negatively influences hematopoietic cells,19 we saw no evidence for this in our stromal cell–free cultures (Table 1).

Comparison of early pro-B cells with other committed precursors on the basis of surface marker expression

Lin TdT+ cells were uniformly positive for CD27, an antigen recently found to be absent from long-term reconstituting stem cells7 and when a PE-conjugated antibody was used, more than half of the LinTdT+ cells also displayed the AA4.1 antigen20(Figure 5). Large and medium-sized Lin TdT+ cells could be distinguished on the basis of forward light scatter, but we found no differences between them in terms of these surface markers. Both medium and large Lin TdT+ cells stained for the Ki-67 proliferation-associated antigen (Figure 5 and data not shown).21 Ly-6C is a protein of unknown function on some bone marrow cells and we previously found that CD45R+Ly-6C+ cells had little potential for differentiation to CD19+ lymphocytes in culture.5 We now report that less than 8% of Lin TdT+ cells were positive for the Ly-6C antigen.

Fig. 5.

Additional characteristics of LinTdT+ bone marrow cells.

Sorted Lin bone marrow cells were stained for surface antigens followed by intracellular TdT staining and analysis by 4 parameter flow cytometry (described in “Materials and methods”). The Lin TdT+ cells in each panel are shown as black dots, whereas all remaining cells are shown as yellow dots. Percentages of the total population of LinTdT+ cells in each quadrant are also given. Cells were also stained for the Ki-67 proliferation antigen (bottom panel). Expression of Ki-67 in Lin TdT+ cells (bold) was compared with that in Gr-1Lo bone marrow cells (dotted) and in IgM+ B cells (plain) along with the isotype control (tinted).

Fig. 5.

Additional characteristics of LinTdT+ bone marrow cells.

Sorted Lin bone marrow cells were stained for surface antigens followed by intracellular TdT staining and analysis by 4 parameter flow cytometry (described in “Materials and methods”). The Lin TdT+ cells in each panel are shown as black dots, whereas all remaining cells are shown as yellow dots. Percentages of the total population of LinTdT+ cells in each quadrant are also given. Cells were also stained for the Ki-67 proliferation antigen (bottom panel). Expression of Ki-67 in Lin TdT+ cells (bold) was compared with that in Gr-1Lo bone marrow cells (dotted) and in IgM+ B cells (plain) along with the isotype control (tinted).

Close modal

In C57BL/6 mice, 42% to 73% of Lin TdT+cells were positive for Ly-6A/Sca-1, a marker extensively used to isolate stem cells and early lymphocyte precursors.22-24However, we found that early pro-B cells were not homogeneous with respect to Sca-1 density. There was a tendency for the c-kitHi cells to have higher amounts of Sca-1 than did the c-kitLo/− TdT+ cells (Figure 5). We found no expression of Thy1.2 on Lin TdT+ cells, but they were uniformly positive for CD43 (not shown).

Heterogeneity was also found with respect to CD4 or the CD16/CD32 Fcγ receptors identified with the 2.4G2 mAb (Figure 5). Therefore, neither of these 2 markers would allow clean discrimination of Lin TdT+ cells. CD44 is another marker that has been used to characterize early precursors in the thymus and we found that all Lin TdT+ cells in bone marrow had a uniformly high density (not shown). The Linc-kitLo TdT+ fraction of bone marrow cells is highly enriched for B lymphocyte precursors and the flow cytometry data permit a more direct comparison with lymphocyte precursor populations described by others. We conclude that most of these early pro-B cells are CD27+ AA4.1+/−Ki-67+Ly-6CLy-6A/Sca-1Lo/−Thy-1CD43+CD4+/−CD16/32Lo/− and CD44Hi. This extends our previous characterization of them as a Lin c-kitLo Flk-2/Flt-3+IL-7Rα+/ −TdT+ fraction of murine bone marrow.

Marrow fractions enriched for early B-lineage precursors include T-lymphocyte precursors

The emphasis of our studies has been on identifying functional precursors for B-lymphocyte lineage cells within bone marrow and the term “early pro-B cells” seemed appropriate for Linc-kitLo Flk-2/Flt-3+ IL-7Rα+cells.5 However, some of these surface characteristics were also used by others to isolate “common lymphoid progenitors”6 and it was important to learn more about the differentiation potential of early pro-B cells. Immunodeficient Rag-1−/− mice were injected with Linc-kitLo Flk-2/Flt-3+ IL-7Rα+cells (Table 2). This subset contained cells with the potential to reconstitute T- as well as B-cell lineages. Thus, a population of Lin c-kitLoFlk-2/Flt-3+ IL-7Rα+ cells that efficiently gives rise to CD19+ lymphocytes also includes T-lineage precursors.

Table 2.

Early pro-B cells can reconstitute T as well as B lymphoid lineages in vivo

CD45.1+, IgM+ B cells (×103)* recoveredCD45.1+CD4+ or CD8+ T cells (×103)recovered
Day 14Day 21Day 14Day 21
Experiment 1 850, 340 850, 450 17 000, 1500 <100, 15 000 
Experiment 2 120, <100 140, 160 6100, <100 13 000, 40 000 
CD45.1+, IgM+ B cells (×103)* recoveredCD45.1+CD4+ or CD8+ T cells (×103)recovered
Day 14Day 21Day 14Day 21
Experiment 1 850, 340 850, 450 17 000, 1500 <100, 15 000 
Experiment 2 120, <100 140, 160 6100, <100 13 000, 40 000 

There were fewer than 105 CD45.1+ cells per organ in uninjected control mice (data not shown).

*

B-cell reconstitution was determined by the presence of CD45.1+ IgM+ cells in the spleen.

T-cell reconstitution was determined by the presence of CD45.1+ CD4+ cells or CD45+CD8+ cells in the thymus.

The focus of this study was on a population of bone marrow cells that lacks lineage-associated surface markers but expresses intracellular TdT. Our previous analysis indicated that they represent important intermediates between multipotential stem cells and functional B-cell precursors. We show here that a fraction enriched for Lin TdT+ cells efficiently survive in well-defined culture conditions and differentiate under the positive influence of 3 cytokines. They are also sensitive to previously described negative regulators, suggesting they may represent an important control point for B lymphopoiesis. We have learned that the population includes cells with the potential for T-lymphocyte lineage differentiation and shares other properties with common lymphoid progenitors described by another laboratory.

The serum-free, stromal cell–free culture conditions developed by Jacobsen and colleagues12 proved ideal for our analysis of early pro-B cells. Remarkably, the same required cytokines were those whose receptors were used as sort criteria. That is, the cells we placed in culture bore mAbs to c-kit, IL-7Rα, and FLk-2/Flt-3. Despite this receptor ligation, at least one in 4 had the potential to respond to the corresponding factors in serum-free culture. Addition of these cytokines in various combinations and with cells isolated with different sorting strategies was informative. For example, SCF was an effective stimulus even when cells were sorted as negative for the corresponding receptor (Figure 2). We previously found that a subset of c-kit cells transiently reacquired this receptor in culture.4 Also, the minimum receptor density required for cytokine responsiveness might be below that detectable by flow cytometry. Gene-targeting experiments indicate that SCF, IL-7, and FL contribute to efficient lymphocyte production in mice.25Ligation of c-kit by SCF is not essential for B lymphopoiesis,25,26 and we observed small numbers of CD19+ cells without addition of SCF to our cultures. Similarly, CD19+ lymphocytes were produced in reduced numbers when FL was omitted from the medium. However, these 2 factors synergize to maintain a normal-sized population of pro-B cells in vivo25 and were needed for optimal production of B-lineage lymphocytes from early pro-B cells. It is now important to discriminate roles played by each and determine how they interact with other positive signals delivered by stromal cells. An additional goal must be to determine why human lymphocyte precursors have more complex and not completely overlapping requirements in culture.27,28 

Our findings complement and extend previous studies regarding roles played by IL-7. Jacobsen and colleagues concluded that the cytokine is important for commitment of multipotential progenitors to lymphocyte development.12 We showed that cells bearing the IL-7R differentiated slightly better than those lacking this molecule, and particularly when assessed at short culture intervals (Figure 3). This suggests that IL-7RαLo/− progenitors require time to become responsive to the cytokine and the 2 fractions were indistinguishable with respect to IL-7Rα density when examined by flow cytometry after 1 week of culture (not shown). However, the presence of more TdT+ cells in the IL-7Rα+subset than in the IL-7RαLo/− fraction suggest the frequency of B-cell progenitors may not have been equivalent in the 2 starting populations. These findings would be consistent with a sequence of differentiation events where expression of TdT precedes display of the IL-7Rα by cells with the potential for acquisition of CD45R and CD19. Other studies indicate that IL-7 may not only support survival of precursors, but stimulate the immunoglobulin gene rearrangement process.29 Cells did not progress beyond the CD45R+ stage to yield CD19+ lymphocytes in our cultures unless IL-7 was included. Targeting of genes for IL-7, the IL-7Rα chain, or the γc chain component of the IL-7R, as well as the intracellular mediator JAK3 kinase all have severe consequences for B lymphocyte lineage differentiation in the mouse.13,14,30,31 However, IL-7 is unlikely to be the only factor involved in this process for several reasons. The lymphocyte deficiency is more severe in IL-7Rα−/− mice than in IL-7Rγc−/− or IL-7−/− mice, and it has been frequently suggested that thymic stromal lymphopoietin (TSLP) plays a partially overlapping role.13,14,32,33 It is noteworthy that there is little progression to the sIgM+B-cell stage in our cultures supported by SCF, FL, and IL-7. There is some indication from culture studies that TSLP augments the final stages in B-lymphocyte formation.33 

It has become increasingly apparent that negative, as well as positive stimuli, control rates of blood cell production and 3 of the inhibitory agents investigated here are effective in vivo. Castration and androgen receptor abnormalities increase B-cell production, whereas it is selectively suppressed during pregnancy and after treatment with sex steroids.34-37 These observations suggest that steady-state lymphocyte production is under hormonal control, but much remains to be learned about relevant target cells and molecular mechanisms. We previously found that IL-7–responsive pro-B cells are insensitive to estrogen, but that stromal cell cocultures initiated with earlier precursors were suppressed.16 The findings indicated that stromal cells might be induced to produce a soluble negative regulator of B lymphopoiesis. This possibility is still open but we show with stromal cell–free cultures and highly purified precursors that early pro-B cells are also potential hormone targets. Efforts to identify estrogen-regulated genes and consequences of hormone therapy should include an analysis of this rare cell population.

We recently discovered limitin, an IFN-like cytokine that preferentially suppresses B lymphopoiesis.11 This activity has also been described for type I IFN17 and limitin can use the same receptor.11 We demonstrate here that early pro-B cells are directly and similarly suppressed by limitin, type I IFN, and type II IFN. Stromal cells, macrophages, and T lymphocytes are all potential sources of these factors within bone marrow.38-40 However, B-cell precursor populations appear to be normal in type I IFN receptor-mutated mice41 (and data not shown). In contrast to sex steroids, these agents may only influence lymphopoiesis under conditions of trauma, inflammation, or disease.

Transforming growth factor-β is another known inhibitor of B lymphopoiesis and early pro-B cells were directly responsive to it. This extends earlier observations that IL-7–responsive pro-B cells and a pre-B lymphoma are sensitive to TGF-β.42,43 However, it is not a cytokine that is preferentially active on the B-lymphocyte lineage.18,44 In addition to the potentially direct effect on hematopoietic precursors, TGF-β may negatively influence stromal cell IL-7 production.45 

Epidermal growth factor was of interest for several reasons. This is an estrogen-inducible cytokine and one that can elicit estrogen receptor–mediated signaling.46,47 Therefore, involvement of EGF in or mimicking of sex steroid–elicited responses seemed possible. We show here that early pro-B cells are unaffected by EGF. However, EGF may influence components of the bone marrow microenvironment because it promotes growth of bone marrow–derived stromal cells48 as well as fetal thymic architecture49 in culture.

This panel of negative regulators included 2 that selectively inhibit lymphopoiesis, but not myelopoiesis and this direct comparison now makes it possible to appreciate differences in their actions.11,16,50 Although limitin blocked formation of CD45R+ CD19 cells in culture, these presumptive intermediates actually accumulated in cultures containing estrogen (Table 1). We will report elsewhere that very early hematopoietic precursors in intact mice are estrogen sensitive, but it is possible this response is mediated indirectly via stromal cells (K.L.M., manuscript in preparation). Conventional IFNs and TGF-β are known inhibitors of nonlymphoid hematopoietic cells as well as lymphocyte precursors.18,44,51As discussed above, estrogen is the only one of these agents likely to be important as a lineage-specific negative regulator under normal steady-state conditions.

One important objective of this study was to learn more about the surface marker characteristics of Lin TdT+early pro-B cells. The Ly-6A (Sca-1) antigen is useful for identifying stem cells from some strains of mice22-24 and common lymphoid progenitors were isolated with this as a sort criterion.6 We found nearly complete overlap between the “common lymphoid progenitor” and early pro-B cells on the basis of Sca-1 expression (Figure 5). However, there was a range of densities and a tendency for c-kitHi cells to express more than those that were c-kitLo/−. This is consistent with previous findings that stem cells express higher levels of Sca-1 than CLP.6 Sca-1 and c-kit may be progressively down-regulated in parallel as lymphocyte precursors differentiate.4 As expected, early pro-B cells differ from long-term repopulating stem cells by uniform expression of CD27.7 The presence of the Ki-67 antigen indicates that the Lin TdT+cells are cycling, as previously concluded for CLP on the basis of Hoechst dye staining.6,52 CLPs were defined by Kondo and colleagues on the basis of IL-7Rα expression.6 Although most of the Lin TdT+ cells in murine bone marrow express this receptor component,5 questions remain about the timing and requirement for its acquisition. We now show that display of the IL-7R signals readiness to differentiate and that the cytokine is necessary for transition to the CD19+ stage under defined culture conditions.

Several subsets of nonlymphoid progenitors have recently been found among the Lin Sca-1 fraction of bone marrow and resolved on the basis of FcγR expression.8Lin TdT+ cells were not homogeneous for this marker and it does not seem promising for their enrichment (Figure 5). However, most did not have the high FcγR levels characteristic of many granulocyte-macrophage restricted progenitors. We previously found that functional B-cell precursors lack CD34, another distinction from most myeloid progenitors.5,8 

Differences in batches of labeled mAbs and mouse strains as well as other technical reasons complicate comparisons between laboratories concerning extremely rare subsets of bone marrow cells. The functional B-cell precursors we refer to as early pro-B cells only partially resemble cells designated “fraction A0” by Hardy and colleagues.9 Some, but not all, early pro-B cells display the C1qRp receptor recognized by the AA4.1 antibody and many, but not all, expressed the CD4 antigen used by Hardy to define fraction A0.9 CD4 has long been used to identify the earliest thymic immigrants and cells with T-lineage potential within bone marrow.53 That is consistent with our finding that early pro-B cell populations include T-lineage precursors. On the other hand, others found little or no B-lymphocyte lineage potential associated with CD4+ bone marrow cells.54,55Furthermore, we found variable, low level expression of CD4 on CD45R+ CD19Ly-6C+ marrow cells that also failed to differentiate in culture.5 We also found that LinTdT+ cells were negative for Thy-1.2. This is consistent with a recent report that a Lin Sca-1+ c-kit+Thy-1 population is enriched with respect to B-lymphocyte lineage potential, whereas a Lin Sca-1+c-kit+ Thy-1Lo population includes myeloid progenitors.56 In summary, we previously determined that functional B-cell precursors were enriched among the Linc-kitLo Flk-2/Flt-3+IL-7Rα+/−TdT+ fraction of murine bone marrow. The present findings show that the LinTdT+ cells are CD27+AA4.1+/−Ly-6CLy-6A/Sca-1Lo/−Thy-1CD43+CD4+/−CD16/32Lo/− and CD44Hi.

We demonstrated that functional T-lymphocyte lineage progenitors were present in the Lin c-kitLoIL-7Rα+ Flk-2/Flt-3+ fraction of bone marrow (Table 5). This result is entirely consistent with the similarities we found in surface markers expressed by early pro-B cells and the common lymphoid progenitors described by others.6 However, caution must be exercised with respect to terminology and interpretation of differentiation pathways. Kondo and colleagues showed that at least some individual cells with a given phenotype have B and T lymphocytes, but not myeloid differentiation potential.6 It remains to be seen if cells with that unique capability represent a major and obligatory intermediate step between stem cells and lymphocyte precursors. Our previous studies and data presented here are informative about the characteristics of early B-lineage precursors. The efficiency and time required for differentiation of early pro-B cells suggests this is a major pathway to the pre-B cell stage and although myeloid differentiation options appear to have been lost, at least some cells in the population retain T-lineage potential. Some type of single cell analysis, perhaps along the lines used by Katsura and colleagues,57 would be required to learn what fraction of early pro-B cells should be designated common lymphoid progenitors. The dependence of these cells on 3 cytokines and their direct sensitivity to inhibitors suggests they may represent an important control point for lymphocyte formation.

Note added in proof: Thurmond and colleagues recently described transplantation experiments with estrogen receptor α knockout mice and conclude that hematopoietic cells can be direct hormone targets (Endocrinology. 2000;141:2309).

The authors thank Dr Lisa Borghesi for critical reading of the manuscript, as well as Ms Viji Dandapani and Mr Jim Henthorn for cell sorting. The assistance of Ms Shelli Wasson in manuscript preparation is also appreciated.

Supported by grant AI 20069 from the National Institutes of Health.

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
Park
Y-H
Osmond
DG
Dynamics of early B lymphocyte precursor cells in mouse bone marrow: proliferation of cells containing terminal deoxynucleotidyl transferase.
Eur J Immunol.
19
1989
2139
2144
2
Desiderio
SV
Yancopoulos
GD
Paskind
M
et al
Insertion of N regions into heavy-chain genes is correlated with expression of terminal deoxytransferase in B cells.
Nature.
311
1984
752
755
3
Komori
T
Okada
A
Stewart
V
Alt
FW
Lack of N regions in antigen receptor variable region genes of TdT-deficient lymphocytes.
Science.
261
1993
1171
1174
4
Payne
KJ
Medina
KL
Kincade
PW
Loss of c-kit accompanies B lineage commitment and acquisition of CD45R in most murine B lymphocyte precursors.
Blood.
94
1999
713
723
5
Tudor
K-S RS
Payne
KJ
Yamashita
Y
Kincade
PW
Functional assessment of precursors from murine bone marrow suggests a sequence of early B-lineage differentiation events.
Immunity.
12
2000
335
345
6
Kondo
M
Weissman
IL
Akashi
K
Identification of clonogenic common lymphoid progenitors in mouse bone marrow.
Cell.
91
1997
661
672
7
Wiesmann
A
Phillips
RL
Mojica
M
et al
Expression of CD27 on murine hematopoietic stem and progenitor cells.
Immunity.
12
2000
193
199
8
Akashi
K
Traver
D
Miyamoto
T
Weissman
IL
A clonogenic common myeloid progenitor that gives rise to all myeloid lineages.
Nature.
404
2000
193
197
9
Li
YS
Wasserman
R
Hayakawa
K
Hardy
RR
Identification of the earliest B lineage stage in mouse bone marrow.
Immunity.
5
1996
527
535
10
Borghesi
LA
Yamashita
Y
Kincade
PW
Heparan sulfate proteoglycans mediate interleukin-7- dependent B lymphopoiesis.
Blood.
93
1999
140
148
11
Oritani
K
Medina
KL
Tomiyama
Y
et al
Limitin: an interferon-like cytokine that preferentially influences B-lymphocyte precursors.
Nat Med.
6
2000
659
666
12
Borge
OJ
Adolfsson
J
Jacobsen
AM
Lymphoid-restricted development from multipotent candidate murine stem cells: distinct and complimentary functions of the c-kit and flt3- ligands.
Blood.
94
2000
3781
3790
13
Peschon
JJ
Morrissey
PJ
Grabstein
KH
et al
Early lymphocyte expansion is severely impaired in interleukin 7 receptor-deficient mice.
J Exp Med.
180
1994
1955
1960
14
Von Freeden-Jeffry
U
Vieira
P
Lucian
LA
McNeil
T
Burdach
SEG
Murray
R
Lymphopenia in interleukin (IL)-7 gene-deleted mice identifies IL-7 as a nonredundant cytokine.
J Exp Med.
181
1995
1519
1526
15
Medina
KL
Strasser
A
Kincade
PW
Estrogen influences the differentiation, proliferation and survival of early B lineage precursors.
Blood.
95
2000
2059
2067
16
Smithson
G
Medina
K
Ponting
I
Kincade
PW
Estrogen suppresses stromal cell-dependent lymphopoiesis in culture.
J Immunol.
155
1995
3409
3417
17
Lin
Q
Dong
C
Cooper
MD
Impairment of T and B cell development by treatment with a type I interferon.
J Exp Med.
187
1998
79
87
18
Keller
JR
McNiece
IK
Sill
KT
et al
Transforming growth factor β directly regulates primitive murine hematopoietic cell proliferation.
Blood.
75
1990
596
602
19
Dooley
DC
Oppenlander
BK
Spurgin
P
et al
Basic fibroblast growth factor and epidermal growth factor downmodulate the growth of hematopoietic cells in long-term stromal cultures.
J Cell Physiol.
165
1995
386
397
20
Jordan
CT
McKearn
JP
Lemischka
IR
Cellular and developmental properties of fetal hematopoietic stem cells.
Cell.
61
1990
953
963
21
Bruno
S
Darzynkiewicz
Z
Cell cycle dependent expression and stability of the nuclear protein detected by Ki-67 antibody in HL-60 cells.
Cell Prolif.
25
1992
31
40
22
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
2320
23
Aihara
Y
Buhring
H
Aihara
M
Klein
J
An attempt to produce “pre-T” cell hybridomas and to identify their antigens.
Eur J Immunol.
16
1986
1391
1399
24
Spangrude
GJ
Heimfeld
S
Weissman
IL
Purification and characterization of mouse hematopoietic stem cells.
Science.
241
1988
58
62
25
Mackarehtschian
K
Hardin
JD
Moore
K
Boast
S
Goff
SP
Lemischka
I
Targeted disruption of the flk2/flt3 gene leads to deficiencies in primitive hematopoietic progenitors.
Immunity.
3
1995
147
161
26
Takeda
S
Shimizu
T
Rodewald
HR
Interactions between c-kit and stem cell factor are not required for B-cell development in vivo.
Blood.
89
1997
518
525
27
Pribyl
JAR
Lebien
TW
Interleukin 7 independent development of human B cells.
Proc Natl Acad Sci U S A.
93
1996
10348
10353
28
Leonard
WJ
Shores
EW
Love
PE
Role of the common cytokine receptor gamma chain in cytokine signaling and lymphoid development.
Immunol Rev.
148
1995
97
114
29
Corcoran
AE
Riddell
A
Krooshoop
D
Venkitaraman
AR
Impaired immunoglobulin gene rearrangement in mice lacking the IL-7 receptor.
Nature.
391
1998
904
907
30
Cao
X
Shores
EW
Hu-Li
J
et al
Defective lymphoid development in mice lacking expression of the common cytokine receptor γ chain.
Immunity.
2
1995
223
238
31
Wei
C
Zeff
R
Goldschneider
I
Murine pro-B cells require IL-7 and its receptor complex to up-regulate IL-7Rα, terminal deoxynucleotidyltransferase, and c μ expression.
J Immunol.
164
2000
1961
1970
32
Ray
RJ
Furlonger
C
Williams
DE
Paige
CJ
Characterization of thymic stromal-derived lymphopoietin (TSLP) in murine B cell development in vitro.
Eur J Immunol.
26
1996
10
16
33
Levin
SD
Koelling
RM
Friend
SL
et al
Thymic stromal lymphopoietin: a cytokine that promotes the development of IgM+ B cells in vitro and signals via a novel mechanism.
J Immunol.
162
1999
677
683
34
Wilson
CA
Mrose
SA
Thomas
DW
Enhanced production of B lymphocytes after castration.
Blood.
85
1995
1535
1539
35
Medina
KL
Smithson
GM
Kincade
PW
Suppression of B lymphopoiesis during normal pregnancy.
J Exp Med.
178
1993
1507
1515
36
Smithson
G
Beamer
WG
Shultz
KL
Christianson
SW
Shultz
LD
Kincade
PW
Increased B lymphopoiesis in genetically sex steroid-deficient hypogonadal (hpg) mice.
J Exp Med.
180
1994
717
720
37
Smithson
G
Couse
JF
Lubahn
DB
Korach
KS
Kincade
PW
The role of estrogen receptors and androgen receptors in sex steroid regulation of B lymphopoiesis.
J Immunol.
161
1998
27
34
38
Wang
J
Lin
Q
Langston
H
Cooper
MD
Resident bone marrow macrophages produce type 1 interferons that can selectively inhibit interleukin-7-driven growth of B lineage cells.
Immunity.
3
1995
475
484
39
Zoumbos
NC
Djeu
JY
Young
NS
Interferon is the suppressor of hematopoiesis generated by stimulated lymphocytes in vitro.
J Immunol.
133
1984
769
774
40
Shah
G
Dexter
TM
Lanotte
M
Interferon production by human marrow stromal cells.
Br J Haematol.
54
1983
365
372
41
Muller
U
Steinhoff
U
Reis
LF
Functional role of type I and type II interferons in antiviral defense.
Science.
264
1994
1918
1921
42
Lee
G
Ellingsworth
LR
Gillis
S
Wall
R
Kincade
PW
β transforming growth factors are potential regulators of B lymphopoiesis.
J Exp Med.
166
1987
1290
1299
43
Lee
G
Namen
AE
Gillis
S
Ellingsworth
LR
Kincade
PW
Normal B cell precursors responsive to recombinant murine IL-7 and inhibition of IL-7 activity by transforming growth factor-β.
J Immunol.
142
1989
3875
3883
44
Keller
JR
Mantel
C
Sing
GK
Ellingsworth
LR
Ruscetti
SK
Ruscetti
FW
Transforming growth factor β 1 selectively regulates early murine hematopoietic progenitors and inhibits the growth of IL-3-dependent myeloid leukemia cell lines.
J Exp Med.
168
1988
737
750
45
Tang
JH
Nuccie
BL
Ritterman
I
Liesveld
JL
Abboud
CN
Ryan
DH
TGF-β down-regulates stromal IL-7 secretion and inhibits proliferation of human B cell precursors.
J Immunol.
159
1997
117
125
46
Ignar-Trowbridge
DM
Nelson
KG
Bidwell
MC
et al
Coupling of dual signaling pathways: epidermal growth factor action involves the estrogen receptor.
Proc Natl Acad Sci U S A.
89
1992
4658
4662
47
Ignar-Trowbridge
DM
Teng
CT
Ross
KA
Parker
MG
Korach
KS
McLachlan
JA
Peptide growth factors elicit estrogen receptor-dependent transcriptional activation of an estrogen-responsive element.
Mol Endocrinol.
7
1993
992
998
48
Gimble
JM
Pietrangeli
CE
Henley
A
et al
Characterization of murine bone marrow and spleen derived stromal cells: analysis of leukocyte marker and growth factor mRNA transcript levels.
Blood.
74
1989
303
311
49
Shinohara
T
Honjo
T
Epidermal growth factor can replace thymic mesenchyme in induction of embryonic thymus morphogenesis in vitro.
Eur J Immunol.
26
1996
747
752
50
Medina
KL
Kincade
PW
Pregnancy-related steroids are potential negative regulators of B lymphopoiesis.
Proc Natl Acad Sci U S A.
91
1994
5382
5386
51
Klimpel
GR
Fleischmann
WR
Jr
Klimpel
KD
Gamma interferon (IFNα) and IFN α/β suppress murine myeloid colony formation (CFU-C): magnitude of suppression is dependent upon level of colony-stimulating factor (CSF).
J Immunol.
129
1982
76
80
52
du Manoir
S
Guillaud
P
Camus
E
Seigneurin
D
Brugal
G
Ki-67 labeling in postmitotic cells defines different Ki-67 pathways within the 2c compartment.
Cytometry.
12
1991
455
463
53
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
4493
54
Wineman
JP
Gilmore
GL
Gritzmacher
C
Torbett
BE
Müller-Sieburg
CE
CD4 is expressed on murine pluripotent hematopoietic stem cells.
Blood.
80
1992
1717
1724
55
Rolink
A
Ten Boekel
E
Melchers
F
Fearon
DT
Krop
I
Andersson
J
A subpopulation of B220+ cells in murine bone marrow does not express CD19 and contains natural killer cell progenitors.
J Exp Med.
183
1996
187
194
56
Searles
AE
Pohlmann
SJ
Pierce
LJ
et al
Rapid, B lymphoid-restricted engraftment mediated by a primitive bone marrow subpopulation.
J Immunol.
165
2000
67
74
57
Kawamoto
H
Ikawa
T
Ohmura
K
Fujimoto
S
Katsura
Y
T cell progenitors emerge earlier than B cell progenitors in the murine fetal liver.
Immunity.
12
2000
441
450

Author notes

Paul W. Kincade, Immunobiology and Cancer Program, Oklahoma Medical Research Foundation, 825 NE 13th St, Oklahoma City, OK 73104.

Sign in via your Institution