Recently, several lines of evidence have indicated an expanded role for thrombopoietin (TPO) and its receptor, c-mpl, in hematopoiesis. In addition to being the primary physiological regulator of platelet production, it is now apparent that TPO also acts during early hematopoiesis. To futher define the role of TPO in early hematopoiesis we have identified discrete murine and human stem cell populations with respect to c-mpl expression and evaluated their potential for hematopoietic engraftment. Fluorescence-activated cell sorter analysis of enriched stem cell populations showed the presence of c-mpl expressing subpopulations. Approximately 50% of the murine fetal liver stem cell–enriched population, AA4+Sca+c-kit+, expressed c-mpl. Analysis of the murine marrow stem cell population LinloSca+c-kit+ showed that 70% of this population expressed c-mpl. Expression of c-mpl was also detected within the human bone marrow CD34+CD38 stem cell progenitor pool and approximately 70% of that population expressed c-mpl. To rigorously evaluate the role of TPO/c-mpl in early hematopoiesis we compared the repopulation capacity of murine stem cell populations with respect to c-mpl expression in a competitive repopulation assay. When comparing the fetal liver progenitor populations, AA4+Sca+c-kit+c-mpl+and AA4+Sca+c-kit+c-mpl, we found that stem cell activity segregates with c-mpl expression. This result is complemented by the observation that the LinloSca+ population of c-mplgene-deficient mice was sevenfold less potent than LinloSca+ cells from wild-type mice in repopulating activity. The engraftment potential of the human CD34+CD38c-mpl+ population was evaluated in a severe combined immunodeficient-human bone model. In comparison to the CD34+CD38c-mpl population, the CD34+CD38c-mpl+ cells showed significantly better engraftment. These results demonstrate a physiological role for TPO and its receptor, c-mpl, in regulating early hematopoiesis.

THROMBOPOIETIN (TPO) is the primary physiological regulator of platelet production and was initially thought to be a lineage dominant factor primarily affecting megakaryocytopoiesis.1,2 Like other hematopoietic growth factors, however, several lines of evidence indicate that TPO has a more pleiotropic range of activities. This was first realized when TPO was found to accelerate red blood cell recovery in myelosuppressed mice,3-5 as well as to synergize with erythropoietin (EPO) to stimulate erythroid proliferation in vitro.5,6 In addition, in vivo administration of TPO caused an expansion of colony-forming unit granulocyte-macrophage (CFU-GM) and burst-forming unit-erythroid (BFU-E) in normal mice3 and expanded CFU-mixed in rhesus monkeys.7 It was also shown that megakaryocytopoietic activities of TPO are initiated at the early progenitor cell level.8 Recently, several in vitro studies have shown that TPO alone or in combination with the early acting growth factors, c-kit ligand (KL), interleukin-3 (IL-3), or Flt-3 ligand (FL), stimulates the proliferation of early hematopoietic progenitors.9-19Significantly, progenitors expanded by TPO in combination with KL or FL retain a primitive phenotype and maintain the capacity for multilineage differentiation, even in the presence of TPO.9,12-14,16-18TPO also enhances the expansion and viability of CD34+CD38 progenitors in culture.19 

The phenotypic alteration of c-mpl- and TPO-deficient mice also indicate a role for TPO in regulating hematopoietic progenitors. As expected, the primary phenotype of these mice is severe thrombocytopenia. However, detailed analysis of their marrow shows deficiencies in their progenitor pool as well.20-23 In addition to reduced megakaryocyte progenitors, both TPO- andc-mpl–deficient mice show significant reductions in neutrophil, GM, erythroid, and multilineage progenitors in the bone marrow (BM), spleen, and peripheral blood (PB).20,21 A reduction in blast cell–colony-forming cell (CFC) was also observed inc-mpl–deficient mice.21 TPO administration toTPO-deficient mice restores their progenitors to normal or above-normal levels.22 These latter observations further indicate that TPO may act to maintain and/or expand early progenitors.

To rigorously characterize the role of c-mpl and TPO in hematopoiesis we have identified discrete murine hemopoietic progenitor populations with respect to c-mpl, and evaluated their capacity to repopulate lethally irradiated mice. Similarly, the repopulation capacity of hematopoietic progenitors from c-mpl gene-deficient mice was also determined. To further expand our analysis we have characterized the expression of c-mpl on human stem cell populations and their functional capabilities using the severe combined immunodeficient-human (SCID-hu) bone model. Collectively, these data show that the expression of c-mpl is important in early hematopoiesis.

Flow cytometric analysis of murine fetal liver and BM.

Murine fetal liver stem cell populations were isolated from AA4.1+ (called AA4+) cells derived by immune panning from mid-gestation fetal liver essentially as previously described.8 The AA4+ cells were subsequently stained using directly conjugated antibodies against murine c-kit (fluorescein isothiocyanate [FITC]), Sca-1 (R-PE) (Pharmingen, San Diego, CA), and biotinylated hamster anti-murine c-mpl.24 For the biotinylated antibodies, streptavidin Red-670 (GIBCO-BRL, Grand Island, NY) was used for detection.

BM populations were derived using femurs from C57/BL/6 mice. Briefly, the femurs were obtained and flushed with PBS/2% fetal calf serum (FCS) and a single cell suspension was made. Any remaining red blood cells were removed by lysis with 10 mmol/L NH4Cl. The mononuclear fraction was then incubated with monoclonal antibodies (MoAbs) specific for lineage markers consisting of rat anti-mouse CD4/L3T4, CD8/Ly 2,3, B-220/CD45R (Caltag, South San Francisco, CA), Gr-1, Mac-1, and Ter-119 (Pharmigen, San Diego, CA). Lineage marker positive cells were then removed by magnetic bead depletion with a sheep anti-rat antibody conjugated to magnetic beads (Dynal, Great Neck, NY). The lineage depleted cells were then incubated with MoAbs specific for: c-kit (FITC), Sca-1 (R-PE) (purchased from Pharmigen), goat anti-rat Lin (Cascade Blue; Molecular Probes, Eugene, OR), and biotinylated hamster anti-murine c-mpl.24 A second step using Streptavidin Red-670 (GIBCO-BRL) was included for the biotinylated samples. All fetal liver or BM samples were analyzed or sorted on a Coulter EPICS Elite (Hialeah, FL) dual-laser flow cytometer configured with a Coherent model 302 Krypton laser (407 nm) and argon laser (488 nm). The appropriate isotype controls were included for each experiment. All analysis excluded propidium iodide positive cells and all cells with high forward or right angle light scatter.

Competitive repopulation of fetal liver progenitors and c-mpl–deficient BM.

Timed pregnant C57/BL/6 mice were killed and the murine day 14 fetal liver cell populations were isolated as described above. The following enriched populations were then sorted: AA4+Sca+c-mpl+ and AA4+Sca+c-mpl. For the competitive repopulation analysis of c-mpl–deficient mice, the BM population LinloSca+ was isolated from either homozygote mutant mice (c-mpl−/−) or their wild-type (c-mpl+/+) littermate controls. Young adult male C57/BL/6 Ly 5.2 mice were obtained from NCI and used as recipients. A minimum of five animals was used per experimental group. Whole-body irradiation (1,100 rads, 190 cGy/min) was administered as a single dose from a 137Cs source. One million nucleated BM cells from the Ly 5.2 mice were used as a source competitor, and mixed with 1,000 or 2,000 fetal liver donor cells or 10,000 BM-derived donor cells. Cells were administered via tail-vein injection and PB samples (50 to 100 μL) were obtained via the retro-orbital sinus 4, 12, and 24 weeks postreconstitution. The percentage of Ly 5.1 (A20.1) donor cells was determined indirectly by staining with biotin-conjugated Ly 5.2 MoAb (A20.1.7). Briefly, PB was collected in 10 U/mL heparin, 1 mmol/L EDTA in phosphate-buffered saline (PBS) and immediately placed on ice. Erythrocytes were removed by the addition of 5 vol of 4% Dextran T500 (Pharmacia) in PBS followed by incubation at room temperature for 20 minutes. The red blood cell–depleted fraction was then centrifuged for 5 minutes at approximately 200g. The remaining red blood cells were then lysed using 10 mmol/L NH4Cl. The mononuclear fraction was then resuspended in PBS/2% FCS. This procedure removed nearly 100% of the erythrocytes (the remaining red blood cells were excluded by size gating) while leaving the leukocytes 95% viable by propidium iodide exclusion. Fluorescence-activated cell sorter (FACS) analysis of Ly 5.2 PB cells was nearly identical to cells prepared by lysing. Controls for the specificity of Ly 5.2 staining included cells from Ly 5.1 or Ly 5.2 animals and cells from animals repopulated with Ly 5.2 competitor cells only. To confirm repopulation by the donor Ly 5.1 cells in all lineages, PB cells and the BM mononuclear fraction were stained in conjunction with Ly 5.1 for B220, CD4, CD8 (lymphoid lineages), and Gr-1/Mac-1 (myeloid lineages).

The relative repopulating ability of each putative stem cell population was compared by calculating repopulation units (RU) as described by Harrison et al.25 Each RU represents the same repopulating ability as 1 × 105 fresh marrow cells. It has been determined that the long-term repopulating stem cell content of fresh marrow is one stem cell per 105 total marrow cells.25 Therefore, the number of RU from each donor population is defined by the following equation:

%(Donor Repopulation)=100(RU/RU+C),
whereC=number of fresh competitor marrow cells/105RU
=%(C)/(100%of Donor Repopulation)
Isolation of CD34+ cells from human BM.

Fresh human BM aspirates were obtained from healthy donors (Poietic Technologies, Gaithersburg, MD). The mononuclear cells were then enriched for CD34 by immunomagnetic positive selection (Miltenyi Biotech, Inc, Auburn, CA) according to the manufacturer's instruction. CD34 content was assessed by FACS and purity was routinely greater than 90%. The CD34+ fraction was then stained with murine MoAbs against human CD34 (FITC), CD38 (R-PE), (Becton Dickinson, San Jose, CA), and c-mpl (biotin).26 The biotinylated samples were then subsequently stained with Streptavidin Red-670 (GIBCO-BRL).

SCID-hu bone mice.

CB-17 scid/scid mice were implanted with fetal bone as previously described.27 Briefly, human fetal bone fragments from femurs, tibias, and humeri of 17- to 22-week gestational fetuses were implanted subcutaneously in 8-week-old CB-17 scid/scidmice. The BM fragments were allowed to engraft for 8 weeks. Mice were then bled via the retro-orbital sinus and engraftment of the bone fragments was determined by flow cytometric analysis of the blood samples with MoAbs specific for human HLA class 1 epitopes (One Lambda, Canoga Park, CA).

SCID-hu bone mice were administered whole-body irradiation of 200 rads from a 137Cs source and sorted human stem cells were then injected directly into a bone graft using a Hamilton syringe. Mice were then killed 8 weeks postinjection, HLA disparate grafts were removed, and marrow mononuclear cells were analyzed by FACS for donor HLA contribution as well as CD19, CD33, and CD34 (Becton Dickinson).

Expression of c-mpl on murine stem cell populations.

To determine if a murine primitive hematopoietic cell population enriched for stem cell activity expressed c-mpl, we isolated AA4.1+ (refered to as AA4+ here) cells from midgestational murine fetal liver by immunopanning and quantified the expression of c-mpl on the AA4+ fraction that is double-positive for the Sca-1 marker (called Sca here) and c-kit. The AA4+Sca+c-kit+ fraction has been shown to be highly enriched for stem cell activity.28 29Approximately 50% of the AA4+ cells that were positive for both Sca and c-kit (AA4+Sca+c-kit+) were c-mpl+. This c-mpl positive fraction (AA4+Sca+c-kit+ c-mpl+) constituted less than 0.1% of the total fetal liver (Fig 1A).

Fig. 1.

(A) Expression of c-mpl of murine fetal liver stem cell populations. Fetal liver stem cells were enriched for AA4 and stained with MoAbs to murine Sca, c-kit, and c-mpl as described in Materials and Methods. Fetal liver cells positive for both Sca and c-kit were gated and the resultant c-mpl positive cells are shown. Analysis was done a minimum of four times. (B) Expression of c-kit, Sca, and c-mpl on murine BM hematopoietic progenitors. The mononuclear fraction from BM was isolated by a density gradient and stained with a Lin cocktail of antibodies (see Materials and Methods). Lin stained cells were then removed by magnetic bead depletion. The lineage-depleted cells were then stained using directly conjugated MoAbs against Sca (R-PE), c-kit (FITC), c-mpl (biotin), and the Lin cocktail (Cascade Blue). The biotinylated c-mpl was detected using Streptavidin Red-670. Analysis was repeated three times. (C) Expression of CD34, CD38, and c-mpl on enriched human BM progenitor cells. CD34 cells were isolated as described. The enriched cells were then stained with MoAbs to CD34 (FITC), CD38 (R-PE), and c-mpl (Red -670). Cells were then analyzed on a Coulter Epics Elite flow cytometer as described. BM cells were enriched for CD34 using an immunomagnetic column. Analysis was repeated six times.

Fig. 1.

(A) Expression of c-mpl of murine fetal liver stem cell populations. Fetal liver stem cells were enriched for AA4 and stained with MoAbs to murine Sca, c-kit, and c-mpl as described in Materials and Methods. Fetal liver cells positive for both Sca and c-kit were gated and the resultant c-mpl positive cells are shown. Analysis was done a minimum of four times. (B) Expression of c-kit, Sca, and c-mpl on murine BM hematopoietic progenitors. The mononuclear fraction from BM was isolated by a density gradient and stained with a Lin cocktail of antibodies (see Materials and Methods). Lin stained cells were then removed by magnetic bead depletion. The lineage-depleted cells were then stained using directly conjugated MoAbs against Sca (R-PE), c-kit (FITC), c-mpl (biotin), and the Lin cocktail (Cascade Blue). The biotinylated c-mpl was detected using Streptavidin Red-670. Analysis was repeated three times. (C) Expression of CD34, CD38, and c-mpl on enriched human BM progenitor cells. CD34 cells were isolated as described. The enriched cells were then stained with MoAbs to CD34 (FITC), CD38 (R-PE), and c-mpl (Red -670). Cells were then analyzed on a Coulter Epics Elite flow cytometer as described. BM cells were enriched for CD34 using an immunomagnetic column. Analysis was repeated six times.

Close modal

In adult murine marrow, we analyzed the expression of c-mpl on the LinloSca+c-kit+ population which is enriched for stem cell activity.30 In these experiments the mononuclear fraction of BM was depleted of lineage-committed progenitors with immunomagnetic beads (see Materials and Methods) and the Linlo mononuclear cells were then further fractionated into the LinloSca+c-kit+population. FACS analysis of the LinloSca+c-kit+ population showed that 70% of this population was c-mpl+ (Fig 1B). This population comprised less than 0.1% of total adult BM. As expected, we detected c-mpl on the Linhi cells (data not shown) that are presumably the lineage committed megakaryocyte progenitors.

Expression of c-mpl on human stem cell progenitor populations.

To evaluate the presence of c-mpl on human progenitors we analyzed the expression of c-mpl on the CD34+CD38stem cell progenitor population derived from human marrow. The CD34+CD38 population comprises 10% of the total CD34+ population and is highly enriched for multiprogenitor and stem cell activity.31-33CD34+ cells were isolated from adult BM by immunomagnetic positive selection (>90% purity), then stained with MoAbs against human CD34, CD38, and c-mpl for FACS analysis. Similar to the expression pattern on murine progenitors, approximately 70% of the CD34+CD38 population expressed c-mpl (Fig 1C).

Long-term self-renewing stem cell activity segregates with c-mpl expression.

In this study, we used a competitive repopulation assay to compare the long-term functional abilities of murine hematopoietic stem cell populations with respect to c-mpl expression. Briefly, progenitor-enriched donor populations derived from either mid-gestational fetal liver or adult BM were mixed with 106competitor cells from BM of C57/BL6 Ly 5.2 animals and then injected into lethally irradiated C57/BL6 Ly 5.2 recipients. At the 4-, 12-, and 24-week time points, the relative contribution of donor cells to hematopoietic lineages was determined by FACS analysis of the Ly 5.1 and Ly 5.2 allotypes.

Using this model we directly compared the repopulating activity of AA4+Sca+c-mpl+ and AA4+Sca+c-mpl fetal liver cells derived from wild-type mice (Table1). This direct comparison shows that all of the long-term repopulating activity of the AA4+Sca+ population segregated with c-mpl expression. At the 24-week time point more than 50% of the myeloid and lymphoid cells were derived from the AA4+Sca+c-mpl+ donor cells with equal contribution to each of these hematopoietic compartments. At 24 weeks the AA4+Sca+c-mpl+ population exhibited a reconstituting activity of 220 RU. In contrast, the AA4+Sca+c-mpl cells exhibited no repopulating activity at the 24-week time point. The AA4+Sca+c-mpl population did exhibit short-term reconstituting activity, indicating that this population consists primarily of committed progenitors. These data indicate that the enriched fetal liver AA4+Sca+population can be fractionated into functionally and phenotypically different populations based on differential expression of c-mpl, and that c-mpl expression in this subset is associated with pluripotent repopulating activity.

To further expand this analysis, we isolated LinloSca+ cells from the BM ofc-mpl–deficient mice and their wild-type littermate controls. The LinloSca+ cells from wild-type animals contained significantly more RU when compared with LinloSca+ cells from thec-mpl–deficient mice (37 v 5). Preferential engraftment of the lymphoid or myeloid compartments was not observed with the LinloSca+ donor populations isolated from either wild-type or c-mpl–deficient mice. In addition, for the mice injected with wild-type LinloSca+ cells, the majority of lymphoid and myeloid cells were derived from this donor population (Table 1).

To evaluate the in vivo functional characteristics of the human CD34+CD38c-mpl+ population in comparison to the CD34+CD38c-mplfraction, the multilineage engraftment potential of these populations was compared in the SCID-hu bone model.27 Thirty thousand cells of each phenotype were injected directly into the bone fragments of the SCID-hu animals and at 8 weeks postinjection, the mice were killed, and mononuclear cells from the grafts were analyzed by FACS. Cell suspensions were made of each graft and analyzed for overall donor contribution by FACS using HLA haplotype-specific antibodies. In addition, the contribution of donor cells to myeloid (CD33), lymphoid (CD19), and hematopoietic progenitors (CD34) was determined. As shown in Table 2, the CD34+CD38c-mpl+ population engrafted better (donor HLA) than the other populations. The combined results of two separate experiments show that the CD34+CD38c-mpl+ population engrafted in 90%(9 of 10) of the mice injected with this cell population. In contrast, of the mice receiving the CD34+CD38c-mpl cell population only 22% (2 of 9) showed donor derived reconstitution. In addition, the contribution of donor derived cells to the progenitor (CD34+), myeloid (CD33+), or lymphoid (CD19+) compartments was higher with the CD34+CD38c-mpl+ than the CD34+CD38c-mplpopulation 8 weeks postreconstitution. This was especially true for the CD34+ progenitor population. In both experiments, with mice injected with the CD34+CD38c-mpl+ cells, greater than 22% of CD34+ cells were donor derived. This compares to 2% to 4% donor-derived CD34+ cells for the two mice that engrafted with the CD34+CD38c-mpl cells.

Recent in vitro data clearly indicate that TPO may function not only to expand the megakaryocyte compartment, but also to play a role in the expansion and maintenance of early progenitors. TPO alone is capable of activating quiescent progenitors into cycle13,18 and acts in synergy with either IL-3, KL, or FL to expand the early progenitor pool.9-19 In support of these observations, we have recently shown that hematopoietic progenitors expanded in a stroma-free liquid culture using a cytokine cocktail consisting of FL, KL, and TPO maintain their long-term lymphohematopoietic repopulating activity in vivo.34 The hematopoietic deficiencies that occur inc-mpl and TPO-deficient mice, which exhibit a significant reduction of multipotent progenitors, further indicate a role for TPO/c-mpl in early hematopoiesis.20 21 

At the molecular level, expression of c-mpl mRNA has been detected in human CD34+cells35 and noncycling progenitors.36 FACS analysis shows that c-mpl is expressed on the stem cell enriched murine fetal liver population AA4+Sca+.8 To further characterize stem cell populations that express c-mpl, we have identified discrete progenitor populations derived from murine and human sources that express c-mpl. FACS analysis of murine BM (LinloSca+c-kit+) or fetal liver (AA4+Sca+c-kit+) stem cell populations shows that c-mpl is expressed on at least 50% of each of these stem cell–enriched populations.

The differential expression of c-mpl within the AA4+Sca+ fetal liver progenitor population allowed evaluation of its functional characteristics with respect to c-mpl expression. Using a murine competitive repopulation model,25 we directly compared the in vivo hematopoietic reconstituting activity of AA4+Sca+c-mpl+ and AA4+Sca+c-mpl murine fetal liver cell populations. These results show that all of the long-term repopulating activity of the fetal liver progenitor population (AA4+Sca+) segregates with c-mpl expression. Repopulating activity showed equal contribution to the myeloid and lymphoid compartments by the donor cell population (AA4+Sca+c-mpl+). The AA4+Sca+c-mpl population exhibited a low level of short-term reconstituting activity, indicating that this population is devoid of true stem cell activity. Furthermore, marrow progenitors (LinloSca+) derived fromc-mpl gene-deficient mice showed a significant (sevenfold) reduction in repopulating activity when compared with the LinloSca+ progenitor population derived from wild-type littermates. The segregation of stem cell activity with c-mpl expression on the fetal liver AA4+Sca+population indicates that c-mpl is expressed on a pluripotent stem cell and that this subpopulation may further define a long-term self-renewing stem cell.

It is interesting to note that although the level of hematopoietic progenitors is diminished by 50% in adultc-mpl−/−mice, day 14 fetal liver progenitors are not reduced inc-mpl−/− mice,21suggesting that TPO does not play a critical role in early fetal hematopoiesis. However, the inability of the AA4+Sca+c-mpl fetal liver cells to repopulate in the adult mouse indicates that c-mpl expression may be essential for fetal liver progenitors to exhibit repopulating activity in the adult marrow microenviroment. Because the LinloSca+ progenitor population from thec-mpl−/−mice had a low level of stem cell activity and that only platelets, and not other hematopoietic blood cells, are affected in these mice, suggests that for adult hematopoiesis, c-mpl may play a crucial role in the function of primitive stem/progenitors cells only when the homeostasis of the hematopoietic compartment is perturbed. Thus, expression of c-mpl on a subset of stem/progenitor cells may allow them to respond to feedback mechanisms that maintain homeostasis in the hematolymphoid compartment.

As was observed for the murine progenitor populations, c-mpl is also expressed on the stem cell–enriched progenitor population, CD34+CD38, derived from human marrow. The expression pattern of c-mpl expression within this population is similar to that observed for the murine progenitors with greater than 50% of CD34+CD38 expressing c-mpl. These results are in contrast to those of Debili et al,37who suggest that the c-mpl+ cells present in the CD34+ fraction are primarily caused by late megakaryocyte progenitors and transitional cells. Expression of c-mpl on CD34+CD38 cells and the ability of TPO to activate noncycling progenitors13 confirm that c-mpl is indeed expressed on an early progenitor.

Until recently the study of long-term human hematopoiesis in vivo was difficult because of the lack of an appropriate in vivo model. However, using immunodeficient, nonobese diabetic (NOD)/SCID and scid/scid mice, models have been developed in recent years to assay for human hematopoietic stem cells and allow evaluation of their multilineage repopulating capacity.27, 32,38In this study we used the SCID-hu bone model27 to determine the effect of c-mpl expression on the in vivo reconstituting activity of the human progenitor CD34+CD38 cell population. A direct comparison of the CD34+CD38c-mpl+ and CD34+CD38c-mpl cell populations shows that c-mpl expression correlates with significantly better donor-derived engraftment. This was apparent in both overall engraftment and donor contribution to various hematopoietic lineages. With equal numbers of cells injected, 90% of the mice receiving the CD34+CD38c-mpl+ cells engrafted compared with 22% engraftment for the mice receiving the c-mpl fraction. In addition, a higher level of donor contribution to the progenitor, myeloid, and lymphoid compartments was observed with the CD34+CD38c-mpl+ donor cells. This was especially true for the CD34+ fraction in which greater than 20% were donor derived using CD34+CD38c-mpl+ cells compared with only 4% for the CD34+CD38c-mpl donor cells or 8% for the c-mpl unenriched CD34+CD38 fraction. Taken together, these results suggest that the multilineage long-term reconstituting activity of the human CD34+CD38 cell population correlates with c-mpl expression.

The expanded role of TPO in hematopoiesis suggest that TPO may be useful clinically for more than just alleviating thrombocytopenia. Recent in vitro studies have shown that TPO in combination with other early growth factors can cause multilineage expansion of CD34+ cells in a long-term culture.17,33 Ex vivo expansion would be a great benefit for the banking of umbilical cord progenitors for future engraftment procedures. Additionally, recent clinical trials have reported that TPO treatment results in an increase in marrow hematopoietic progenitors and peripheral CD34+ cells.39 These preliminary clinical results again suggest that TPO may be useful not only as a thrombopoietic agent but also in peripheral stem cell harvest procedures.

We thank Jim Chin for excellent technical assistance and Daniel Tumas for critically reviewing the manusript.

Address reprint request to Dan L. Eaton, PhD, Genentech, 1 DNA Way, South San Francisco, CA 94080.

The publication costs of this article were defrayed in part by page charge payment. This article must therefore be hereby marked "advertisement" is accordance with 18 U.S.C. section 1734 solely to indicate this fact.

1
Eaton
DL
de Sauvage
FJ
Thrombopoietin: The primary regulator of magakaryocytopoiesis and thrombopoiesis.
Exp Hematol
25
1997
1
2
Kaushansky
K
Thrombopoietin: The primary regulator of megakaryocytopoiesis.
Trends Exp Med
8
1997
45
3
Kaushaunsky
K
Lin
N
Grossman
A
Humes
J
Sprugel
K
Broudy
V
Thrombopoietin expands erthyroid, granulocyte-macrophage, and megakaryocyte progenitor cell in normal and myelosupressed mice.
Exp Hematol
24
1996
265
4
Farese
AM
Hunt
P
Grab
LB
MacVittie
TJ
Combined administration of recombinant megakaryocyte growth and development factor and granulocyte colony stimulating factor enhances multilineage hematopoietic reconstitution in nonhuman primates after radiation induced marrow aplasia.
J Clin Invest
97
1996
2145
5
Kaushansky
K
Broudy
VC
Grosman
A
Humes
J
Lin
N
Ren
HP
Baily
MC
Papayannopoulou
T
Forstrom
JW
Sprugel
KH
Thrombopoietin expands erthroid progenitors, increases red cell production and enhances erythroid recovery after myelosuppresive therapy.
J Clin Invest
96
1995
1683
6
Kobayashi
M
Laver JH, Kato T, Miyazaki H, Ogawa M: Recombinant human thrombopoietin (Mpl ligand) enhances proliferation of erythroid progenitors.
Blood
8
1995
2494
7
Farese
AM
Hunt
P
Boone
T
MacVittie
TJ
Recombinant human megakaryocyte growth and development factor stimulates thrombopoiesis in normal nonhuman primates.
Blood
86
1995
54
8
Zeigler
FC
de Sauvage
F
Widmer
HR
Keller
GA
Donahue
C
Schreiber
RD
Malloy
B
Hass
P
Eaton
D
Matthews
W
In vitro megakaryocytopoietic and thrombopoietic activity of c-mpl ligand (TPO) on purified murine hematopoietic stem cells.
Blood
84
1994
4045
9
Ku
H
Yonemura
Y
Kaushansky
K
Ogawa
M
Thrombopoietin, the ligand for the mpl receptor, synergizes with steel factor and other early acting cytokines in supporting proliferation of primitive hematopoietic progenitors of mice.
Blood
87
1996
4544
10
Sitnicka
E
Lin
N
Priestley
GV
Fox
N
Broudy
VC
Wolf
NS
Kaushansky
K
The effect of thrombopoietin on the proliferation and differentiation of murine hematopoietic stem cells.
Blood
87
1996
4998
11
Petzer
AL
Zanstra
PW
Piret
JM
Eaves
CJ
Differential cytokine effects on primitive (CD34+CD38−) human hematopoietic cells: Novel responses to flt-3 ligand and thrombopoietin.
J Exp Med
183
1996
2551
12
Kobayashi
M
Laver
JH
Kato
T
Miyazaki
H
Ogawa
M
Thrombopoietin supports proliferation of human primitive hematopoietic cells in synergy with steel factor and/or interleukin-3.
Blood
88
1996
429
13
Young
JC
Bruno
E
Luens
KM
Wu
S
Backer
M
Murray
LJ
Thrombopoietin stimulates megakaryocytopoiesis, myelopoiesis, and expansion of CD34+ progenitor cells from single CD34+Thy-1+Lin− primitive progenitor cells.
Blood
88
1996
1619
14
Rasko
JEJ
O'Flaherty
E
Begley
CG
mpl ligand (MGDF) alone and in combination with stem cell factor (SCF) promotes proliferation and survival of human megakaryocyte, erythroid and granulocyte/macrophage progenitors.
Stem Cells
15
1997
33
15
Ohmizono
Y
Sakabe
H
Kimura
T
Tanimukai
S
Matsumura
T
Miuazaki
H
Lyman
SD
Sonoda
Y
Thrombopoietin augments ex vivo expansion of human cord blood derived hematopoietic progenitors in combination with stem cell factor and flt3 ligand.
Leukemia
11
1997
524
16
Ramsfjell
V
Borge
OJ
Veiby
OP
Cardier
J
Murphy
MJ
Lyman
SD
Lok
S
Jacobsen
SEW
Thrombopoietin, but not erythropoietin, directly stimulates multilineage growth of primitive murine bone marrow progenitor cells in synergy with early cytokines: Distinct interactions with the ligands for c-kit and flt3.
Blood
88
1996
4481
17
Piacibello
W
Sanavio
F
Garetto
L
Severino
A
Bergandi
D
Ferrario
J
Fagioli
F
Berger
M
Aglietta
M
Extensive amplification and self renewal of human primitive hematopoietic stem cells from cord blood.
Blood
89
1997
2644
18
Ramsfjell
V
Borge
OJ
Cui
L
Jacobsen
SEW
Thrombopoietin directly and potently stimulates multilineage growth and progenitor cell expansion from primitive (CD34+CD38−) human bone marrow progenitor cells.
J Immunol
158
1997
5169
19
Borge
OJ
Ramsfjell
V
Cui
L
Jacobsen
SEW
Ability of early acting cytokines to directly promote survival and suppress apoptosis of human primitive CD34+CD38− bone marrow cells with mutilineage potential at the single cell level: Key role of thrombopoietin.
Blood
90
1997
2282
20
Carver-Moore
K
Broxmeyer
HE
Luoh
SM
Cooper
S
Peng
J
Burstein
SA
Moore
MW
de Sauvage
FJ
Low levels of erythroid and myeloid progenitors in thrombopoietin and c-mpl deficient mice.
Blood
88
1996
803
21
Alexander
WS
Roberts
AW
Nicola
NA
Li
R
Metcalf
D
Deficiencies in progenitor cells of multiple hematopoietic lineages and defective megakaryocytopoiesis in mice lacking the thrombopoietin receptor c-mpl.
Blood
87
1996
2162
22
de Sauvage
FJ
Carver-Moore
K
Luoh
S-M
Ryan
A
Dowd
M
Eaton
DL
Moore
MW
Physiological regulation of early and late stages of megakaryocytopoiesis by thrombopoietin.
J Exp Med
183
1995
651
23
Gurney
AL
Carver-Moore
K
de Sauvage
FJ
Moore
MW
Thrombocytopenia in c-mpl deficient mice.
Science
265
1994
1445
24
Shivdasani
RA
Fielder
P
Keller
G
Orkin
S
de Sauvage
FJ
Regulation of the serum concentration of thrombopoietin in thrombocytopenic NF-E2 knockout mice.
Blood
90
1997
1821
25
Harrison
D
Jordan
C
Zhong
R
Astle
C
Primitive hematopoietic stem cells: direct assay of most productive populations by competitive repopulation with simple, binomial, correlation and covariance calculations.
Exp Hematol
21
1993
206
26
Pearce
KH
Potts
BJ
Presta
LG
Bald
LN
Fendly
BM
Wells
JA
Mutational analysis of thrombopoietin for identification of receptor and neutralizing antibody sites.
J Biol Chem
272
1997
20595
27
Kyoizumi
S
Baum
CM
Kaneshima
H
McCune
JM
Yee
EJ
Nauikawa
R
Implantation and maintenance of functional human bone marrow in SCID-hu mice.
Blood
79
1992
1704
28
Zeigler
FC
Bennet
BD
Jordon
CT
Spencer
SD
Baumhueter
S
Carroll
KJ
Hooley
J
Bauer
K
Matthews
BS
Cellular and molecular characterization of the role of the FLK-2/FLT-3 receptor tyrosine kinase in hematopoietic stem cells.
Blood
84
1994
2422
29
Jordan
CT
Astle
CM
Zawadzki
J
Mackarehtschian
K
Lemischka
IR
Harrison
DE
Long-term repopulating abilities of enriched fetal liver stem cells measured by competitive repopulation.
Exp Hematol
23
1995
1011
30
Rebel
VI
Miller
CL
Thornbury
GR
Dragowska
WH
Eaves
CJ
Lansdorp
PM
A comparison of long term repopulating hematopoietic stem cells in fetal liver and adult bone marrow from the mouse.
Exp Hematol
24
1996
638
31
Civin
CI
Almeida-Porada
G
Lee
M-J
Olweus
J
Terstappen
L
Zanjani
ED
Sustained, retransplantable, multilineage engraftment of highly purified adult human bone marrow stem cells in vivo.
Blood
88
1996
4102
32
Larochelle
A
Vormoor
J
Hanenberg
H
Wang
J
Bhatia
M
Lapidot
T
Moritz
T
Murdoch
B
Xiao
XL
Kato
I
Williams
DA
Dick
JE
Identification of primitive human hematopoietic cells capable of repopulating NOD/SCID mouse bone marrow: Implications for gene therapy.
Nature Med
2
1996
1329
33
Terstappen
LWMM
Huang
S
Safford
M
Lansdorp
P
Loken
M
Sequential generations of hematopoietic colonies derived from single non-lineage-committed CD34+CD38− progenitor cells.
Blood
77
1991
1218
34
(abstr, suppl 1)
Solar
G
Chen
Q
de Sauvage
F
Eaton
D
In vitro expansion of early hematopoietic progenitors by flt 3 ligand, kit ligand and TPO.
Blood
90
1997
475a
35
Methia
N
Louache
F
Vainchenker
W
Wendling
F
Oligodeoxynucleotides antisense to the proto-oncogene c-mpl specifically inhibit in vitro megakaryocytopoiesis.
Blood
82
1993
1395
36
AC
Wang
A
Levine
JD
Lopez
P
Scadden
DT
Functional isolation and characterization of human hematopoietic stem cells.
Science
267
1995
104
37
Debili
N
Wendling
F
Cosman
D
Titeux
M
Florindo
C
Dusanter-Fourt
I
Schooley
K
Methia
N
Charon
M
Nador
R
Bettaieb
A
Vainchenker
W
The mpl receptor is expressed in the megakaryocytic lineage from late progenitors to platelets.
Blood
85
1995
391
38
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
39
Murray
LJ
Luens
KM
Estrada
MF
Vadhan-Raj
S
Bruno
E
Hoffman
R
Cohen
RL
Thrombopoietin mobilizes CD34+ cell subsets into peripheral blood and expands multilineage progenitors in bone marrow of patients with sarcoma.
Exp Hematol
26
1998
207
Sign in via your Institution