In acute myeloid leukemia (AML), granulocyte colony-stimulating factor receptor (G-CSFR) proliferative and maturational signaling pathways are uncoupled. Seven human G-CSFR mRNA isoforms exist, named class I through class VII. The 183-amino acid cytosolic domain of the class I isoform provides all signaling activities. The class IV isoform is “differentiation defective” because the carboxy-terminal 87 amino acids are replaced with 34 amino acids of novel sequence. In more than 50% of AML samples, the class IV/class I G-CSFR mRNA ratio is aberrantly elevated compared to normal CD34+ bone marrow cells. We hypothesized that the increased relative expression of class IV G-CSFR in AML uncouples proliferative and maturational G-CSFR signaling pathways. To test this, we transfected the G-CSF–responsive murine cell line 32Dcl3 with class IV G-CSFR cDNA. After 10 days of G-CSF stimulation, clones expressing class IV G-CSFR had greater percentages of myeloblasts and promyelocytes than controls (53% ± 13% versus 3% ± 2%). Differential counts over time demonstrated delayed G-CSF–driven maturation in 5 class IV-expressing clones, with 2 clones demonstrating a subpopulation that completely failed to differentiate. Heterologous class IV expression did not affect G-CSF–dependent proliferation. Class IV/murine G-CSFR mRNA ratios after 24 hours of G-CSF stimulation for 3 of the 5 clones (range, 0.090 to 0.245; mean, 0.152 ± 0.055) are within the range of class IV/class I mRNA ratios seen in patients with AML. This indicates that aberrantly increased relative class IV G-CSFR expression seen in AML can uncouple G-CSFR proliferative and maturational signaling pathways.

Granulocyte colony-stimulating factor (G-CSF), the critical growth factor for the normal production of mature circulating neutrophils, acts on myeloid cells during all stages of development.1-6 Because of the ability of G-CSF to promote the maturation of myeloid cells, it was tested as a potential means of differentiation therapy for acute myeloid leukemia (AML).7-10 However, the typical effect was to stimulate proliferation in blasts of patients with AML, with maturation either aberrant or absent.11-13 This indicated that the proliferative and maturational responses to G-CSF are somehow uncoupled in AML. Mutation of the G-CSF receptor (G-CSFR) or alterations in the levels or functions of the downstream signaling molecules have been proposed as hypotheses to explain this aberrant response in AML.14-19 

The human G-CSFR is known to occur in 7 isoforms (class I through class VII), all produced by alternative splicing of the single gene transcript.20-24 Only the class I and class IV G-CSFR mRNA isoforms are detected at significant levels in myeloid cells.25 Functional mapping studies have shown that the carboxy-terminal 87 amino acids of the class I G-CSFR are crucial to the maturational signaling function of the G-CSFR, whereas the membrane proximal 96 amino acids are fully competent for driving proliferation.14,22,26-31 The class IV G-CSFR isoform replaces the carboxy-terminal 87-amino acid maturation domain of the full-length class I G-CSFR with 34 amino acids of novel peptide sequence, thus leaving the proliferative capacity of this isoform intact. However, when class IV G-CSFR is expressed in isolation, it fails to mediate G-CSF–driven myeloid differentiation.21,22,28 We have previously demonstrated that in blasts of patients with AML, the class IV G-CSFR isoform is coexpressed at relatively increased levels compared with normal immature myeloid cells.25 Given the “differentiation-defective” nature of the class IV G-CSFR isoform, we have examined whether this alteration in the relative levels of G-CSFR isoform coexpression is sufficient to uncouple the proliferative and maturational responses to G-CSF that typify AML.

Cells

The murine myeloblastic cell line 32Dcl3 expresses endogenous murine G-CSFR and differentiates into phenotypic neutrophils in response to G-CSF stimulation.32 The parental cell line is maintained in Iscove's modified Dulbecco's medium supplemented with 10% fetal bovine serum, 100 U/mL penicillin G, 100 μg/mL streptomycin, 1 mmol/LL-glutamine, and 10% WEHI 3B D+cell-conditioned medium as a source of murine interleukin-3 (mIL-3).

Expression vector constructs and transfection

The cDNA clone D-7 (generous gift from S. Zeigler) in pBluescript (Stratagene, La Jolla, CA) was isolated after digestion withEcoRI and NotI. This fragment was then cloned into theEcoRI/NotI digested expression vector pcDNA3.1(−)/NEO (Clontech, Palo Alto, CA). 32Dcl3 cells were then electroporated as described elsewhere33 with either the pcDNA3.1(−)/NEO/D-7 (D-7) or the pcDNA3.1(−)/NEO (NEO) construct, and stable transfectants were selected with 600 μg/mL Geneticin (Life Technologies, Grand Island, NY). Limiting dilution was used to isolate individual clones from each bulk population, and their expression of murine G-CSFR and D-7 cDNA was determined using the polymerase chain reaction (PCR)-based method described below.

Reverse transcription–polymerase chain reaction and quantitative polymerase chain reaction

Total RNA was isolated from each clone using the TRIzol reagent (Life Technologies, Grand Island, NY). Two micrograms RNA was then used in a reverse transcription–polymerase chain reaction (RT-PCR) described elsewhere.25 The PCR primers used for amplification of D-7 cDNA sequence (HGR5, HGR3) have been published.25 The PCR primers used for amplification of the murine G-CSFR were MGR5 5′-CCACTACACCATCTTCTG-3′ and MGR3 5′-CCAAGAGGGGCTGAGTGG-3′. The primer combination for quantitative (Q)-PCR measurement of the D-7/murine G-CSFR mRNA ratio is MGR5 (200 nmol) plus HGR3 (100 nmol) and MGR3 (100 nmol). Data were discarded unless equivalent PCR amplification efficiency was confirmed for the murine G-CSFR and D-7 PCR fragments over a minimum of 3 cycles during the linear phase of amplification.

Maturation assay

32Dcl3 clones were stimulated with rhG-CSF (G-CSF; Amgen, Thousand Oaks, CA) 100 ng/mL, and differential counts were made from preparations of Wright–Giemsa-stained cells at days 5, 7, and 10 of culture. To delay apoptosis and allow up-regulation of endogenous G-CSFR expression, recombinant murine IL-3 (Peprotech, Rocky Hill, NJ) was added at a concentration of 0.1 ng/mL during the initial 72 hours of G-CSF stimulation. This dose of IL-3 was determined to be adequate to suppress apoptosis but not to induce more than 1 cycle of cell division over 72 hours, as measured by the proliferation assay discussed below (data not shown).

Proliferation assay

The proliferative response of D-7 and NEO clones was measured using the Cell Census System (Sigma, St Louis, MO). One million cells were labeled with PHK26 and divided into 3 equal aliquots. Immediately, 1 aliquot was fixed in 2% paraformaldehyde (Sigma) in phosphate-buffered saline (PBS). The remaining 2 aliquots were stimulated with either 100 ng/mL G-CSF or 30 ng/mL recombinant mIL-3. After 72 hours of cytokine stimulation, the cells were fixed in 2% paraformaldehyde in PBS for subsequent flow cytometric evaluation according to the manufacturer's protocol in comparison to unstimulated cells fixed immediately after staining. Flow cytometric data were then analyzed using Cell Census System (Sigma) software, and a “proliferation index” was computed that represented the average number of generations the initial population of cells produced, with the initial population having a proliferation index of 1.00. The proliferation index after G-CSF stimulation for an individual clone was divided by the proliferation index after IL-3 stimulation for that clone as a means of normalizing for the clone's baseline proliferative capacity.

Statistics

The Student t test was used to analyze results from the maturation and proliferation studies (P values). The Pearson product moment correlation coefficient was used to test the correlation between the maturation data and class IV/murine G-CSFR mRNA ratios (r value).

Differentiation-defective G-CSFR expression in 32Dcl3 cells at levels observed in AML cells blocks G-CSF–mediated maturation

To assess the ability of the class IV G-CSFR isoform to uncouple the proliferative and maturational signaling functions of the full-length G-CSFR, we expressed the class IV G-CSFR cDNA (D-7) in the IL-3–dependent murine myeloblastic cell line 32Dcl3. This cell line expresses the murine G-CSFR, a structural and functional homologue of the human class I G-CSFR, and differentiates into phenotypic neutrophils with G-CSF stimulation. Individual clones expressing either the class IV G-CSFR (D-7 number) or empty expression vector (NEO number) were stimulated with G-CSF 100 ng/mL, and their phenotypic maturation was assessed over 10 days. Initial screening demonstrated that all the D-7 clones examined demonstrated a significant subpopulation that failed to enter the postmitotic phase of maturation (46.9% ± 13.4%) in comparison to the NEO clones (97.0% ± 2.2%; P ≤ .001) (Figure1 and Table1). All clones were cytokine dependent based on the observation that more than 99% of cells for each clone were nonviable by trypan blue staining after 72 hours of withdrawing cytokine (data not shown).

Fig. 1.

Photomicrographs of 32Dcl3 cell clones following 10 days of G-CSF stimulation (100 ng/mL).

(A) Clone NEO.1, (B) Clone D-7.1, (C) Clone D-7.3, and (D) Clone D-7.4. (Wright-Giemsa stain, original magnification × 1000 [with oil immersion lens].)

Fig. 1.

Photomicrographs of 32Dcl3 cell clones following 10 days of G-CSF stimulation (100 ng/mL).

(A) Clone NEO.1, (B) Clone D-7.1, (C) Clone D-7.3, and (D) Clone D-7.4. (Wright-Giemsa stain, original magnification × 1000 [with oil immersion lens].)

Close modal
Table 1.

Effect of human class IV murine G-CSFR coexpression on G-CSF–mediated myeloid maturation

32D Clone Postmitotic*after 10 days of G-CSF 100 ng/mL (%)
D-7.1  74 
D-7.2  52  
D-7.3  46  
D-7.4  26  
D-7.5  49 
D-7.6  50  
D-7.7  32  
D-7.8  46  
NEO .1  94 
NEO .2  98  
NEO .3  99 
32D Clone Postmitotic*after 10 days of G-CSF 100 ng/mL (%)
D-7.1  74 
D-7.2  52  
D-7.3  46  
D-7.4  26  
D-7.5  49 
D-7.6  50  
D-7.7  32  
D-7.8  46  
NEO .1  94 
NEO .2  98  
NEO .3  99 
*

Postmitotic = myelocytes + bands + neutrophils.

To examine more closely the dynamics of the effect of class IV G-CSFR coexpression on G-CSF–driven maturation, differential counts were performed at days 5, 7, and 10 of G-CSF stimulation on select D-7 and NEO clones; results are shown in Figure 2. When all clones were compared at day 5 of G-CSF stimulation, it was clear that in the NEO clones, 87.3% ± 0.85% of cells were in the postmitotic phase of differentiation, whereas in the D-7 clones, only 15.8% ± 6.5% (P ≤ .001) of cells were postmitotic. By day 7, this increased to between 94.0% ± 4.4% for the NEO clones but only to 45.0% ± 16.7% (P = .001) for D-7 clones. By 10 days of G-CSF stimulation, 100% ± 0% of the cells in each of the NEO clones were postmitotic. However, only 54.0% ± 21.9% of cells in the D-7 clones were in the postmitotic phase of maturation. When individual clones were examined over time, it was observed that for NEO clones there was a consistent progression of all cells forward along the myelopoietic pathway. In the D-7 clones, this progression was dramatically delayed. Even more intriguing was that in 2 of the 5 D-7 clones studied (clones D-7.2 and D-7.4), a sizable myeloblastic subpopulation expanded between days 7 and 10 of G-CSF stimulation. This demonstrated that coexpression of the class IV G-CSFR isoform delays, and in some circumstances blocks, G-CSF–driven differentiation.

Fig. 2.

Differential counts of 32Dcl3 cell clones over time during G-CSF stimulation.

Clones NEO.1-NEO.3 and D-7.1-D-7.5 were stimulated with G-CSF 100 ng/mL and differential counts performed on Wright-Geimsa stained samples at day 5 (A), day 7 (B), and day 10 (C) of culture.

Fig. 2.

Differential counts of 32Dcl3 cell clones over time during G-CSF stimulation.

Clones NEO.1-NEO.3 and D-7.1-D-7.5 were stimulated with G-CSF 100 ng/mL and differential counts performed on Wright-Geimsa stained samples at day 5 (A), day 7 (B), and day 10 (C) of culture.

Close modal

We have previously observed ratios of class IV/class I G-CSFR mRNA in patients with AML, ranging from 0.057 to 0.158 (mean, 0.106 ± 0.035)25 (and unpublished data). These values are significantly greater than the ratios observed in the CD34+ fraction of normal bone marrow cells (range, 0.050 to 0.080; mean, 0.065 ± 0.009).25 Coexpression of the differentiation-defective class IV G-CSFR isoform in 32Dcl3, which expressed a full-length G-CSFR, acted to disrupt the G-CSFR maturational signaling activities. To determine whether relative overexpression of the class IV G-CSFR isoform could have been responsible for the disruption of G-CSFR maturational signaling that occurred in patients with AML, we proceeded to measure the ratio of class IV/murine G-CSFR mRNA to compare to the class IV/class I mRNA ratios previously observed in patients with AML. This ratio, determined using Q-PCR, was measured on mRNA harvested from 2 different populations—cells maintained in IL-3 and cells stimulated with G-CSF 100 ng/mL for 24 hours. The latter group was included to account for the G-CSF–induced up-regulation of murine G-CSFR mRNA. The results of this analysis are shown in Table 2. At baseline, the class IV/murine G-CSFR mRNA ratios ranged from 0.225 to 0.560 (mean, 0.377 ± 0.123). After 24 hours of G-CSF stimulation and the expected up-regulation of the murine G-CSFR mRNA, these ratios changed to 0.090 to 0.245 (mean, 0.152 ± 0.055). The ratios observed here overlapped significantly with those previously observed in patients with AML (P = .14) and not with those observed for normal immature myeloid cells (P = .03).

Table 2.

Q-PCR analysis of human class IV G-CSFR versus murine G-CSFR mRNA levels

Clone Human class IV/murine G-CSFR mRNA ratio
Baseline (mean ± SD) 24 hours G-CSF 100 ng/mL (mean ± SD)
D-7.1  0.225 ± 0.025 0.178 ± 0.021  
D-7.2  0.455 ± 0.035 0.128 ± 0.013  
D-7.3  0.560 ± 0.040 0.090 ± 0.007  
D-7.4  0.264 ± 0.011 0.245 ± 0.025  
D-7.5 0.382 ± 0.042  0.118 ± 0.009 
Mean ± SEM  0.377 ± 0.123 0.152 ± 0.055 
Clone Human class IV/murine G-CSFR mRNA ratio
Baseline (mean ± SD) 24 hours G-CSF 100 ng/mL (mean ± SD)
D-7.1  0.225 ± 0.025 0.178 ± 0.021  
D-7.2  0.455 ± 0.035 0.128 ± 0.013  
D-7.3  0.560 ± 0.040 0.090 ± 0.007  
D-7.4  0.264 ± 0.011 0.245 ± 0.025  
D-7.5 0.382 ± 0.042  0.118 ± 0.009 
Mean ± SEM  0.377 ± 0.123 0.152 ± 0.055 

The degree of maturation delay or block varied among the D-7 clones examined, as did the class IV/murine G-CSFR mRNA ratio. It was considered that an increasing relative level of class IV G-CSFR mRNA might result in an increase in the degree of maturation delay. Statistical analysis looking for such a correlation between the class IV/murine G-CSFR mRNA ratio and the percentage of cells failing to differentiate terminally after 10 days of G-CSF stimulation found no correlation between these values (r = −0.329) (Figure3).

Fig. 3.

Plot of percent mitotic cells versus human class IV/murine G-CSFR mRNA ratio.

A plot of percent mitotic cells (myeloblasts + promyelocytes) versus the class IV/murine G-CSFR mRNA ratio is shown. The Pearson product moment correlation coefficient [r] for this plot is -0.329 indicating a poor correlation between the class IV/murine G-CSFR mRNA ratio and the fraction of cells that remain in the mitotic phase of myelopoiesis for each clone.

Fig. 3.

Plot of percent mitotic cells versus human class IV/murine G-CSFR mRNA ratio.

A plot of percent mitotic cells (myeloblasts + promyelocytes) versus the class IV/murine G-CSFR mRNA ratio is shown. The Pearson product moment correlation coefficient [r] for this plot is -0.329 indicating a poor correlation between the class IV/murine G-CSFR mRNA ratio and the fraction of cells that remain in the mitotic phase of myelopoiesis for each clone.

Close modal

Differentiation-defective G-CSFR expression in 32Dcl3 cells does not affect G-CSF–mediated proliferation

Coexpression of the class IV G-CSFR with a full-length G-CSFR at levels seen in patients with AML disrupted maturational signaling in a manner similar to that seen in AML. If our model system correctly represented the physiology of class IV G-CSFR activity in AML, then our D-7 clones should have demonstrated a proliferative response to G-CSF that was at least as vigorous as the NEO control clones.

Proliferation was measured using the Cell Census System (Sigma) to generate a proliferation index (PI) as outlined in “Materials and methods,” with the results shown in Figure4. Each clone was stimulated with 30 ng/mL rmIL-3 to determine the baseline proliferative capacity (PI [IL-3]), and then the response to 100 ng/mL G-CSF stimulation for each clone was measured (PI [G-CSF]). For comparison between clones, the PI [G-CSF] was divided by the PI [IL-3] for each clone to normalize for the baseline proliferative capacity of that clone, with the result expressed as the percentage of IL-3 response. The normalized proliferative response to G-CSF was unchanged in the D-7 clones (16.5% to 22.5%; mean, 19.1% ± 0.02%) compared with the NEO clones (15.1% to 3.9%; mean, 18.63% ± 0.04%; P = .88). This clearly demonstrated that coexpression of the class IV G-CSFR isoform with the full-length G-CSFR selectively disrupted the maturational signaling pathway; a pattern identical to that seen in most patients with AML.

Fig. 4.

Proliferative response of 32Dcl3 cell clones to G-CSF or IL-3.

Using the Cell Census Plus System (Sigma) the proliferative response of clones NEO.1-NEO.3 and D-7.1-D-7.5 was measured following stimulation with either G-CSF (100 ng/mL) or IL-3 (30 ng/mL) for 72 hours. The raw data indicating the percent of cells in each generation (Gen 0-Gen 9) and the “proliferation index” (see “Materials and methods”) is shown at the right for clones stimulated with G-CSF (A) or IL-3 (B). The proliferation index for G-CSF stimulation was normalized to the proliferation index for IL-3 stimulation for each clone and this value (% IL-3 response) is presented in C.

Fig. 4.

Proliferative response of 32Dcl3 cell clones to G-CSF or IL-3.

Using the Cell Census Plus System (Sigma) the proliferative response of clones NEO.1-NEO.3 and D-7.1-D-7.5 was measured following stimulation with either G-CSF (100 ng/mL) or IL-3 (30 ng/mL) for 72 hours. The raw data indicating the percent of cells in each generation (Gen 0-Gen 9) and the “proliferation index” (see “Materials and methods”) is shown at the right for clones stimulated with G-CSF (A) or IL-3 (B). The proliferation index for G-CSF stimulation was normalized to the proliferation index for IL-3 stimulation for each clone and this value (% IL-3 response) is presented in C.

Close modal

In this study, we have demonstrated that coexpression of the class IV G-CSFR isoform with the full-length murine G-CSFR is able to block completely G-CSF–driven myeloid differentiation in a cell line capable of terminal phenotypic myeloid maturation. However, this coexpression leaves G-CSF–driven proliferation unaffected. The maturation block is observed at ratios of class IV/murine G-CSFR mRNA in the range of 0.090 to 0.245, values similar to the class IV/class I G-CSFR mRNA ratios we previously demonstrated in patients with AML.25 This implicates overexpression of the class IV G-CSFR isoform in the uncoupling of the proliferative and maturational responses to G-CSF typical of patients with AML.

How coexpression of the class IV G-CSFR uncouples the proliferative and maturational signaling pathways of the full-length G-CSFR is unclear. It is presumed that the class I and class IV G-CSFR isoforms can freely lead to homodimerization and heterodimerization after ligand binding. Based on this assumption, 2 general mechanisms can be hypothesized. One would be that the class IV G-CSFR acts as a dominant-negative to the class I G-CSFR and would then interfere with the activation of all the necessary signaling molecules required to halt cell cycling, initiate granule formation, and promote nuclear segmentation. The other mechanism would involve the activation of novel signaling molecules by the class IV G-CSFR. Both mechanisms would depend on the total number of class IV receptors at the cell surface; however, the dominant-negative mechanism would be more sensitive to the ratio of receptor isoforms coexpressed. We found no correlation between the G-CSFR isoform ratio and the degree of maturation block in our study, but the ratio varied over a narrow range for the isolated clones.

Studies of the comparatively rare severe congenital neutropenia (SCN)-related G-CSFR mutations provide insight into how aberrant relative class IV G-CSFR expression could uncouple the proliferative and maturational G-CSFR signaling pathways in most patients with AML by a dominant-negative mechanism. The mutant G-CSFR isoforms found in a subset of patients with SCN are the result of nonsense mutations that truncate the C-terminal 83 to 98 amino acids of the receptor.14,19,34-37 This results in a receptor structure similar to that of class IV G-CSFR in that they all lack the C-terminal amino acid sequence critical to driving myeloid maturation. However, the SCN mutants cannot activate additional signaling molecules, whereas the class IV G-CSFR isoform, with its novel carboxy-terminal peptide sequence, may indeed activate yet unidentified signaling molecules. Functionally, the SCN-related G-CSFR mutants, when expressed alone or in combination with class I G-CSFR, act to delay markedly the ligand-induced G-CSFR internalization, caused in part by the absence of a di-leucine motif, and they produce a hyperproliferative response to G-CSF stimulation.15,18,19 38 The class IV G-CSFR also lacks the di-leucine internalization motif and could be expected to behave in a manner similar to the SCN-related G-CSFR mutant isoforms. However, in our clones, coexpression of the class IV isoform did not impart a hyperproliferative response to G-CSF stimulation. This might have been caused by a difference in the receptor isoform ratio in our clones versus those in the SCN-related G-CSFR mutant studies, in which relative mRNA levels were not determined. It also might have been caused by signaling activity derived from the unique carboxy-terminus of the class IV G-CSFR. In either setting, it is still possible that the same mechanisms that supported the hyperproliferative response to G-CSF in the studies of SCN-related G-CSFR mutants also participated in the maturation block observed in our class IV G-CSFR–expressing clones.

The delay in G-CSFR internalization seen with the SCN-related G-CSFR mutants is associated with prolonged-activation Stat5 and, to a lesser extent, Stat3.15,16 The relevance of this observation has been discussed by other authors.15 16 Preliminary data from our laboratory (not shown), however, suggest that prolonged G-CSF–mediated Stat5 or Stat3 activation does not occur in the setting of class IV G-CSFR/murine G-CSFR coexpression. If this observation holds true, it may be that it is not the duration of activation but rather the balance between the activation of Stat5 and Stat3 that is most critical to disrupting the myeloid maturation process. Therefore, the prolonged activation of Stat5 may be more relevant to the hyperproliferative response to G-CSF imparted by the SCN-related mutant G-CSFR isoforms. This would also explain why we observed a maturation block without a hyperproliferative response to G-CSF stimulation in our class IV/murine G-CSFR coexpression model system.

Delayed mutant G-CSFR internalization may also contribute to the prolonged activity of the mitogenic inositol triphosphate products of phosphatidyl inositol 3′-kinase (PI-3 kinase) caused by the failure to activate SH-2–containing inositol phosphatase (SHIP).39 Inositol triphosphates are known to support cell survival, prevent apoptosis, and augment G-CSF–mediated proliferation.39 This is another possible mechanism that allows the SCN-related mutant G-CSFR isoforms, and potentially the class IV G-CSFR, to override the maturational signals emanating from the full-length G-CSFR.

An alternative to the dominant-negative mechanism of the SCN-related G-CSFR mutants has the novel carboxy-terminal 38 amino acid sequence of the class IV G-CSFR recruiting a distinct signaling molecule (or molecules) that act to suppress maturation. This mechanism would be independent of the duration of surface expression and potentially would be less sensitive to the relative levels of full-length G-CSFR coexpressed as long as sufficient numbers of class IV G-CSFR molecules were present to signal. Such a mechanism would explain the lack of correlation between the ratio of class IV/murine G-CSFR and the degree of maturation delay or block observed in the D-7 clones studied here. We are currently investigating this possibility.

Supported by National Institutes of Health/National Cancer Institute grants K11 CA68480 (S.M.W.) and R01 CA72261 (D.J.T.).

Reprints:Scott M. White, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Suite 501 Lillian S. Kaufmann Bldg, 3471 Fifth Ave, Pittsburgh, PA 15213.

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
Broxmeyer
HE
Williams
DE
Cooper
S
et al
Synergistic interaction of hematopoietic colony stimulating and growth factors in the regulation of myelopoiesis.
Behring Inst Mitt.
83
1988
80
84
2
Hassan
HT
Zyada
LE
Ragab
MH
Rees
JK
Synergistic interactions between recombinant human interleukin-3, GM-CSF and G-CSF in normal human marrow granulocyte-macrophage colony formation.
Cell Biol Int Rep.
15
1991
211
219
3
Kennedy
MJ
Davis
J
Passos-Coelho
J
et al
Administration of human recombinant granulocyte colony-stimulating factor (filgrastim) accelerates granulocyte recovery following high-dose chemotherapy and autologous marrow transplantation with 4-hydroperoxycyclophosphamide-purged marrow in women with metastatic breast cancer.
Cancer Res.
53
1993
5424
5428
4
Welte
K
Bonilla
MA
Gabrilove
JL
et al
Recombinant human granulocyte-colony stimulating factor: in vitro and in vivo effects on myelopoiesis.
Blood Cells.
13
1987
17
30
5
Lemoli
RM
Fortuna
A
Fogli
M
et al
Proliferative response of human marrow myeloid progenitor cells to in vivo treatment with granulocyte colony-stimulating factor alone and in combination with interleukin-3 after autologous bone marrow transplantation.
Exp Hematol.
23
1995
1520
1526
6
Bodine
DM
Crosier
PS
Clark
SC
Effects of hematopoietic growth factors on the survival of primitive stem cells in liquid suspension culture.
Blood.
78
1991
914
920
7
Moore
MA
G-CSF: its relationship to leukemia differentiation-inducing activity and other hemopoietic regulators.
J Cell Physiol.
1(suppl)
1982
53
64
8
Begley
CG
Metcalf
D
Nicola
NA
Purified colony-stimulating factors (G-CSF and GM-CSF) induce differentiation in human HL60 leukemic cells with suppression of clonogenicity.
Int J Cancer.
39
1987
99
105
9
Tomida
M
Yamamoto-Yamaguchi
Y
Hozumi
M
Okabe
T
Takaku
F
Induction by recombinant human granulocyte colony-stimulating factor of differentiation of mouse myeloid leukemic M1 cells.
FEBS Lett.
207
1986
271
275
10
Nicola
NA
Granulocyte colony-stimulating factor and differentiation-induction in myeloid leukemic cells.
Int J Cell Cloning.
5
1987
1
15
11
Suzuki
T
Morio
T
Tohda
S
et al
Effects of interleukin-6 and granulocyte colony-stimulating factor on the proliferation of leukemic blast progenitors from acute myeloblastic leukemia patients.
Jap J Cancer Res.
81
1990
979
986
12
Visani
G
Manfroi
S
G-CSF in the biology and treatment of acute myeloid leukemias.
Leuk Lymphoma.
18
1995
423
428
13
Itoh
K
Bessho
M
Hirashima
K
Effects of recombinant human G-CSF and GM-CSF on primary human leukemic cells.
Nippon Ketsueki Gakkai Zasshi.
52
1989
988
995
14
Touw
IP
Dong
F
Severe congenital neutropenia terminating in acute myeloid leukemia: disease progression associated with mutations in the granulocyte-colony stimulating factor receptor gene.
Leuk Res.
20
1996
629
631
15
Ward
AC
van Aesch
YM
Schelen
AM
Touw
IP
Defective internalization and sustained activation of truncated granulocyte colony-stimulating factor receptor found in severe congenital neutropenia/acute myeloid leukemia.
Blood.
93
1999
447
458
16
Hermans
MH
Antonissen
C
Ward
AC
Mayen
AE
Ploemacher
RE
Touw
IP
Sustained receptor activation and hyperproliferation in response to granulocyte colony-stimulating factor (G-CSF) in mice with a severe congenital neutropenia/acute myeloid leukemia-derived mutation in the G-CSF receptor gene.
J Exp Med.
189
1999
683
692
17
Avalos
BR
The granulocyte colony-stimulating factor receptor and its role in disorders of granulopoiesis.
Leuk Lymphoma.
28
1998
265
273
18
McLemore
ML
Poursine-Laurent
J
Link
DC
Increased granulocyte colony-stimulating factor responsiveness but normal resting granulopoiesis in mice carrying a targeted granulocyte colony-stimulating factor receptor mutation derived from a patient with severe congenital neutropenia.
J Clin Invest.
102
1998
483
492
19
Hunter
MG
Avalos
BR
Deletion of a critical internalization domain in the G-CSFR in acute myelogenous leukemia preceded by severe congenital neutropenia [in process citation].
Blood.
93
1999
440
446
20
Bernard
T
Gale
RE
Linch
DC
Analysis of granulocyte colony stimulating factor receptor isoforms, polymorphisms and mutations in normal haemopoietic cells and acute myeloid leukaemia blasts.
Br J Haematol.
93
1996
527
533
21
Dong
F
van Paassen
M
van Buitenen
C
Hoefsloot
LH
Lowenberg
B
Touw
IP
A point mutation in the granulocyte colony-stimulating factor receptor (G-CSF-R) gene in a case of acute myeloid leukemia results in the overexpression of a novel G-CSF-R isoform.
Blood.
85
1995
902
911
22
Larsen
A
Davis
T
Curtis
BM
et al
Expression cloning of a human granulocyte colony-stimulating factor receptor: a structural mosaic of hematopoietin receptor, immunoglobulin, and fibronectin domains.
J Exp Med.
172
1990
1559
1570
23
Fukunaga
R
Seto
Y
Mizushima
S
Nagata
S
Three different mRNAs encoding human granulocyte colony-stimulating factor receptor.
Proc Natl Acad Sci U S A.
87
1990
8702
8706
24
Seto
Y
Fukunaga
R
Nagata
S
Chromosomal gene organization of the human granulocyte colony-stimulating factor receptor.
J Immunol.
148
1992
259
266
25
White
SM
Ball
ED
Ehmann
WC
Rao
AS
Tweardy
DJ
Increased expression of the differentiation-defective granulocyte colony-stimulating factor receptor mRNA isoform in acute myelogenous leukemia.
Leukemia.
12
1998
899
906
26
Dong
F
van Buitenen
C
Pouwels
K
Hoefsloot
LH
Lowenberg
B
Touw
IP
Distinct cytoplasmic regions of the human granulocyte colony-stimulating factor receptor involved in induction of proliferation and maturation.
Mol Cell Biol.
13
1993
7774
7781
27
Ziegler
SF
Bird
TA
Morella
KK
Mosley
B
Gearing
DP
Baumann
H
Distinct regions of the human granulocyte-colony-stimulating factor receptor cytoplasmic domain are required for proliferation and gene induction.
Mol Cell Biol.
13
1993
2384
2390
28
Ziegler
SF
Davis
T
Schneringer
JA
et al
Alternative forms of the human G-CSF receptor function in growth signal transduction.
New Biologist.
3
1991
1242
1248
29
Fukunaga
R
Ishizaka-Ikeda
E
Pan
CX
Seto
Y
Nagata
S
Functional domains of the granulocyte colony-stimulating factor receptor.
EMBO J.
10
1991
2855
2865
30
Fukunaga
R
Ishizaka-Ikeda
E
Nagata
S
Growth and differentiation signals mediated by different regions in the cytoplasmic domain of granulocyte colony-stimulating factor receptor.
Cell.
74
1993
1079
1087
31
de Koning
JP
Dong
F
Smith
L
et al
The membrane-distal cytoplasmic region of human granulocyte colony-stimulating factor receptor is required for STAT3 but not STAT1 homodimer formation.
Blood.
87
1996
1335
1342
32
Valtieri
M
Tweardy
DJ
Caracciolo
D
et al
Cytokine-dependent granulocytic differentiation: regulation of proliferative and differentiative responses in a murine progenitor cell line.
J Immunol.
138
1987
3829
3835
33
Steinman
RA
Tweardy
DJ
Granulocyte colony-stimulating factor receptor mRNA upregulation is an immediate early marker of myeloid differentiation and exhibits dysfunctional regulation in leukemic cells.
Blood.
83
1994
119
127
34
Naparstek
E
Granulocyte colony-stimulating factor, congenital neutropenia, and acute myeloid leukemia [editorial; comment].
N Engl J Med.
333
1995
516
518
35
Dong
F
Brynes
RK
Tidow
N
et al
Mutations in the gene for the granulocyte colony-stimulating-factor receptor in patients with acute myeloid leukemia preceded by severe congenital neutropenia [see comments].
N Engl J Med.
333
1995
487
493
36
Tidow
N
Pilz
C
Kasper
B
Welte
K
Frequency of point mutations in the gene for the G-CSF receptor in patients with chronic neutropenia undergoing G-CSF therapy.
Stem Cells.
15
1997
113
119
discussion 120.
37
Imashuku
S
Hibi
S
Kataoka-Morimoto
Y
et al
Myelodysplasia and acute myeloid leukaemia in cases of aplastic anaemia and congenital neutropenia following G-CSF administration.
Br J Haematol.
89
1995
188
190
38
Hermans
MH
Ward
AC
Antonissen
C
Karis
A
Lowenberg
B
Touw
IP
Perturbed granulopoiesis in mice with a targeted mutation in the granulocyte colony-stimulating factor receptor gene associated with severe chronic neutropenia.
Blood.
92
1998
32
39
39
Hunter
MG
Avalos
BR
Phosphatidylinositol 3′-kinase and SH2-containing inositol phosphatase (SHIP) are recruited by distinct positive and negative growth-regulatory domains in the granulocyte colony-stimulating factor receptor.
J Immunol.
160
1998
4979
4987
Sign in via your Institution