Stem cell factor (SCF) is a potent costimulatory molecule for many cytokines. Its synergy with granulocyte colony-stimulating factor (G-CSF) results in important biologic and clinical effects, although the mechanism by which this occurs remains poorly understood. To investigate this interaction, this study used a retroviral vector to transduce the G-CSF receptor into MO7e cells, which are known to express the SCF receptor. The transduced G-CSF receptor is functionally active, and the resultant MO7e-G cells recapitulate the proliferative synergy between SCF and G-CSF. When treated with both cytokines, a marked shortening of the G0/G1 phase of the cell cycle occurs, associated with a suppression of the cyclin-dependent kinase inhibitor p27kip-1. In addition, SCF and G-CSF induce the synergistic activation of c-fos, a proto-oncogene involved in propagation of mitogenic signals in hematopoietic cells. G-CSF, but not SCF, induces the tyrosine phosphorylation of STAT1 and STAT3, transcription factors that can mediate the induction of c-fos. However, SCF induces phosphorylation of STAT3 on serine727 (ser727), which is necessary for maximal STAT transcriptional activity, and the combination of SCF and G-CSF leads to complete STAT3 phosphorylation on ser727. The pathways by which SCF and G-CSF lead to serine phosphorylation of STAT3 are distinct and are partially dependent on phosphatidylinositol-3 kinase and ERKs, pathways that are also necessary for the synergistic effects of SCF and G-CSF on proliferation and c-fos induction. Thus, MO7e-G cells provide a powerful system in which the molecular basis of the synergy between SCF and G-CSF can be dissected.

Stem cell factor (SCF) is a potent costimulatory growth factor in hematopoiesis.1-3 In combination with cytokines, SCF results in a synergistic enhancement of the proliferation, differentiation, and survival of hematopoietic cells, which is critical for an adequate expansion and development of the hematologic lineages.4-13 In particular, its synergy with granulocyte colony-stimulating factor (G-CSF) has important biologic and clinical implications. G-CSF and SCF are crucial factors in ex vivo long-term culture of human primitive hematopoietic cells.14 Furthermore, the combination of SCF and G-CSF in vivo increases the mobilization of peripheral blood progenitor cells (PBPC) over that seen with G-CSF alone15-17 and enhances the antileukemic effect mediated by these PBPC.18 In addition, several clinical trials have reported that the combination of SCF and G-CSF improves yield of CD34+ cells in patients at risk of poor mobilization.19-21 However, the mechanism by which this biologically and clinically important interaction between SCF and G-CSF occurs remains poorly understood.

The G-CSF receptor is a single-chain member of the cytokine receptor superfamily, which lacks tyrosine kinase activity.22Binding of G-CSF to its receptor induces the tyrosine phosphorylation of a number of cellular proteins and activates signaling cascades including the signal transducer and activator of transcription (STAT) and mitogen-activated protein kinase (MAPK) pathways.23 In contrast to the G-CSF receptor, the receptor for SCF, encoded by the proto-oncogene c-kit,24-26 possesses intrinsic tyrosine kinase activity. Binding of SCF to c-kit induces kinase activation and transphosphorylation of the receptor chains. Phosphotyrosine residues in the receptor then function as docking sites for proteins that link c-kit activation to various signaling pathways.27 28 Despite the biologic and clinical importance of the combination of G-CSF and SCF, no system has existed in which to study the interaction of these receptor pathways. To overcome this impediment, we generated a cell line that recapitulates the biologic synergy between SCF and G-CSF and has allowed analysis of the molecular events that underlie it.

Cytokines, antibodies, and other reagents

Recombinant human (rh) granulocyte-macrophage colony-stimulating factor (GM-CSF) (sargramostin) was obtained from Immunex Corporation (Seattle, WA), rhSCF from Biosource International (Camarillo, CA), and rhG-CSF from Amgen (Thousand Oaks, CA). H7, puromycin, tetracycline, polybrene, and fetal calf serum (FCS) were purchased from Sigma Chemical Company (St Louis, MO). Antibodies to STAT1 (E-23) and STAT3 (C-20) were purchased from Santa Cruz Biochemicals (Santa Cruz, CA). PD98059 and antibodies to ERK1 were purchased from New England Biolabs (Beverly, MA). Phycoerythrin (PE)-conjugated antibody to c-kit and antibodies to the retinoblastoma protein (Rb) and G-CSF receptor were purchased from Becton Dickinson (San Jose, CA), to p27kip-1 from Transduction Laboratories (Lexington, KY), and to p21cip-1 from Upstate Biotechnology (Lake Placid, NY). Antibodies to the tyrosine phosphorylated forms of STAT129 and STAT330 and to the ser727-phosphorylated forms of STAT1 and STAT331 were generated as described. RPMI 1640, Dulbecco modified Eagle medium (DMEM), l-glutamine, HEPES, penicillin/streptomycin, and G418 were purchased from Gibco/BRL-Life Technologies (Rockville, MD). Horseradish peroxidase-conjugated antirabbit and antimouse antibodies were purchased from Calbiochem (La Jolla, CA).

Cells

The human factor–dependent myeloid cell line MO7e was obtained from Dr J. Griffin (Dana-Farber Cancer Institute, Boston, MA).8,32 Cells were maintained in RPMI 1640 supplemented with 20% (vol/vol) heat-inactivated fetal calf serum (FCS) and 10 ng/mL GM-CSF. The human 293-derived retroviral packaging cell line 293GPG was obtained from Dr G. Dranoff (Dana-Farber Cancer Institute). This cell line is capable of producing high titers of recombinant Moloney murine leukemia virus particles that incorporate, in a tetracycline-regulated fashion, the vesicular stomatitis virus G (VSV-G) protein.33 The 293GPG cells were grown in DMEM supplemented with 10% (vol/vol) FCS, 2 mmol/L l-glutamine, 10 mmol/L HEPES, 250 U/mL penicillin and streptomycin, 2 μg/mL puromycin, 0.3 mg/mL G418, and 1 μg/mL tetracycline. All cells were cultured at 37°C in a humidified atmosphere containing 5% CO2, and cell counts were obtained with a Z2 cell counter (Beckman-Coulter, Miami, FL). Flow cytometric analysis was performed using an Epics MCL-XL (Coulter).

Whole cell and nuclear extracts

To prepare whole cell extracts, cells were placed on ice, washed once with ice-cold phosphate-buffered saline (PBS; 137 mmol/L NaCl, 2.7 mmol/L KCl, 10 mmol/L Na2HPO4, pH 7.4), and extracted at 4°C for 30 minutes in lysis buffer (10 mmol/L Tris, pH 8.0, 0.5% NP-40, 250 mmol/L NaCl, 10 mmol/L sodium orthovanadate, 100 μmol/L phenylmethylsulfonyl fluoride [PMSF], 1 μg/mL leupeptin, 1 μg/mL pepstatin, and 1 μg/mL aprotinin). Insoluble material was removed by centrifugation at 12 000g for 5 minutes. To generate nuclear extracts, cells were washed once with ice-cold PBS, then resuspended in 5 mL hypotonic buffer (10 mmol/L Tris, pH 7.4, 10 mmol/L NaCl, and 6 mmol/L MgCl2) and incubated on ice for 5 minutes. The cells were then centrifuged and resuspended in 0.8 mL hypotonic buffer containing 1 mmol/L β-mercaptoethanol, 10 μg/mL PMSF, and 1 mmol/L sodium orthovanadate. Cells were disrupted by shearing in a Dounce homogenizer (type b pestle, 25 strokes), then nuclei were collected by a 10-second centrifugation at 12 000g. The supernatant was removed and the nuclear pellet was washed once with hypotonic buffer, then resuspended in 3 volumes of high-salt buffer (20 mmol/L HEPES, pH 7.9, 420 mmol/L NaCl, 25% glycerol, 1.5 mmol/L MgCl2, 0.2 mmol/L EDTA, 1 mmol/L β-mercaptoethanol, 1 mmol/L sodium orthovanadate, 1 μg/mL leupeptin, 1 μg/mL pepstatin, 1 μg/mL aprotinin, and 100 μmol/L PMSF), and rocked at 4°C for 30 minutes. Insoluble material and intact nuclei were removed by centrifugation at 12 000g for 3 minutes at 4°C, and the supernatant was recovered and designated the nuclear extract.

Western blotting

Whole cell extracts (100 μg/lane) were resolved by sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE), transferred to nitrocellulose, and blocked with 5% nonfat dry milk (for anti-STAT, antiphosphotyrosine-STAT, and antiphosphoser727-STAT blots) or 5% bovine serum albumin (BSA) (for other blots) in TBST (100 mmol/L Tris, pH 8.0,150 mmol/L NaCl, and 0.05% Tween-20). The primary antibodies were diluted in TBST containing 3% BSA at a dilution of 1:20 000 (anti-STAT, antiphosphotyrosine-STAT, and antiphosphoser727-STAT) or 1:500 (others) and incubated with the blots for 1 hour at room temperature. After being washed with TBST extensively, membranes were incubated for 1 hour at room temperature with horseradish peroxidase–conjugated secondary antibody diluted 1:20 000 in TBST containing 1% BSA. Membranes were then washed extensively, and bound antibody was detected by chemiluminescence (Renaissance Kit; NEN; Boston, MA). Band intensity was quantitated by densitometry. Membranes were stripped by incubation in 62.5 mmol/L Tris, pH 6.9, containing 2% SDS and 100 mmol/L β-mercaptoethanol for 1 hour at 65°C and then washed in TBST at 4°C overnight.

DNA binding assay

Nuclear extract (2 μL) was mixed with 1 ng32P-labeled oligonucleotide (5′-AGCCTGATTTCCCCAAATGACGGC-3′ and its complement34 in 10 μL binding buffer (25 mmol/L HEPES, pH 7.9, 100 μmol/L EGTA, 200 μmol/L MgCl2, 500 μmol/L dithiothreitol, 1 μg/μL BSA, 0.2 μg/μL poly dI:dC, 1% Ficoll, and 0.1 μg/μL herring testis DNA). The incubation was performed at room temperature for 15 minutes. For antibody competition, antiserum (1:200 dilution) was added at the end of the binding reaction and incubated at 4°C for an additional 15 minutes. The products of the binding reaction were separated on a 4% acrylamide gel in 0.2 × Tris-borate/EDTA, after which the gel was dried and exposed to film.

Semiquantitative reverse transcription–polymerase chain reaction (RT-PCR)

RNA was collected (RNeasy Kit; Qiagen; Valencia, CA) and reverse-transcribed using Superscript IIRT (Life Technologies). PCR to amplify c-fos used primers for a 482-bp fragment (sense: 5′-CTACGAGGCGTCATCCT-3′; antisense: 5′-TCTGTCTCCGCTTGGAGTGTA-3′), and PCR to amplify GAPDH used primers for a 226-bp fragment (Perkin Elmer Applied Biosystems; Branchburg, NJ). Minimal cycle numbers were used to allow quantitative comparisons.

Construction of granulocyte colony-stimulating factor receptor retroviral plasmid

The retroviral vector pMFG was originally constructed in the laboratory of Dr R. C. Mulligan (Harvard Medical School, Boston, MA) and was kindly provided by Dr G. Dranoff. The wild-type hG-CSF receptor complementary DNA (cDNA), a gift from Dr Seth Corey35 (University of Pittsburgh School of Medicine, Pittsburgh, PA), was amplified by PCR using PfuTurbo DNA polymerase (Stratagene; La Jolla, CA) and a set of primers with flanking Xba I and Bgl II restriction site sequences (forward, 5′-ATCCTCTAGACTGCCATGGCAAGGCTGGGAAACTGC-3′; reverse, 5′-TAATCCAGATCTGCCCTAGAAGCTCCCCAGCGCCTC-3′), and then subcloned in frame into pMFG after an intermediate cloning step of the blunt-ended PCR product into the pCR-BluntII-TOPO plasmid (Invitrogen; Carlsbad, CA). Sequence analysis was performed to confirm the fidelity of the pMFG-G-CSF receptor construct.

Retroviral generation and infection of MO7e cells

The pMFG-G-CSF receptor (10 μg) was transfected into 293GPG packaging cells that were at 80% confluence in 10-cm dishes using LipofectAMINE (Gibco/BRL) according to the manufacturer's recommendations. The day after transfection, the media was replaced with fresh media without antibiotics (Tet-Off media). This allowed the expression of the tetracycline-dependent VSV-G gene, and the subsequent release into the supernatant of infectious replication-incompetent retroviruses containing the G-CSF receptor gene. The retrovirus-containing supernatant was collected, filtered, stored at −70°C, and replaced with fresh Tet-Off media for 7 days, after which the supernatants were thawed, pooled, and centrifuged at 50 000g for 90 minutes at 4°C to concentrate the retroviral particles. After centrifugation the supernatant was poured off, and the retroviral pellet was resuspended in 2.5 mL of 10% Hank's balanced salt solution in PBS, aliquoted, and either used fresh for infection of MO7e cells or stored at −70°C. MO7e cells growing in log phase were plated in 4 mL media at 3 × 105cells/mL, incubated overnight with 250 μL pMFG-G-CSF receptor retroviral concentrate in the presence of 8 μg/mL polybrene, and then centrifuged and resuspended in fresh media.

Cell cycle analysis

Cell cycle analysis was performed by flow cytometry after staining DNA with propidium iodide (50 μg/mL) in 0.1% (vol/vol) NP-40 and 0.1% (wt/vol) sodium citrate as described previously.36 Data analysis was performed using Multi-Cycle software (Phoenix Flow, San Diego, CA).

High-efficiency transduction of MO7e cells with granulocyte colony-stimulating factor receptor expressing retrovirus

Given the clinical importance of the synergistic interaction between G-CSF and SCF, we generated a model system in which the intracellular signaling events induced by these cytokines could be studied. The hG-CSF receptor was cloned into the retroviral vector pMFG and transduced into MO7e cells, which constitutively express c-kit. After 72 hours, more than 90% of the infected cells were positive for G-CSF receptor surface expression (MO7e-G cells) in 4 independent infection experiments (Figure1A). The level of expression of the G-CSF receptor in MO7e-G cells was comparable to that in neutrophils isolated from the peripheral blood of healthy donors, indicating that expression of the G-CSF receptor was at physiologic levels (data not shown). Given the level of expression achieved by direct transduction, no further selection or purification steps were necessary. The expression of the G-CSF receptor in MO7e-G cells has remained stable for more than 3 months in culture and had no effect on c-kit expression, because 100% of both MO7e-G and MO7e cells expressed c-kitas measured by direct immunostaining with a PE-conjugated antic-kit monoclonal antibody (data not shown).

Fig. 1.

Transduced MO7e-G cells express the G-CSF receptor and show synergistic proliferation in response to SCF and G-CSF.

(A) Three days after infection, MO7e cells transduced with pMFG-G-CSF receptor retroviral concentrate (MO7e-G cells) were stained with either antihuman G-CSF receptor antibody (filled) or an idiotype control (unfilled), and fluorescein isothiocyanate–conjugated secondary antimouse antibody, and analyzed by flow cytometry. One experiment representative of 4 is presented, in which 94% of the cells express the receptor. Untransfected wild-type MO7e cells did not express the G-CSF receptor (dotted line). (B) Parental MO7e cells and MO7e-G cells were factor starved for 18 hours and then incubated in medium containing no growth factor, 100 ng/mL SCF, 10 ng/mL G-CSF, or both. The data presented represent the means and SD of 4 independent experiments.

Fig. 1.

Transduced MO7e-G cells express the G-CSF receptor and show synergistic proliferation in response to SCF and G-CSF.

(A) Three days after infection, MO7e cells transduced with pMFG-G-CSF receptor retroviral concentrate (MO7e-G cells) were stained with either antihuman G-CSF receptor antibody (filled) or an idiotype control (unfilled), and fluorescein isothiocyanate–conjugated secondary antimouse antibody, and analyzed by flow cytometry. One experiment representative of 4 is presented, in which 94% of the cells express the receptor. Untransfected wild-type MO7e cells did not express the G-CSF receptor (dotted line). (B) Parental MO7e cells and MO7e-G cells were factor starved for 18 hours and then incubated in medium containing no growth factor, 100 ng/mL SCF, 10 ng/mL G-CSF, or both. The data presented represent the means and SD of 4 independent experiments.

Close modal

MO7e-G cells recapitulate the biologic synergy between stem cell factor and granulocyte colony-stimulating factor

To validate MO7e-G cells as a model system for the synergy between SCF and G-CSF, we first assessed the biologic characteristics of these cells. Experimental data suggest that the combination of these cytokines enhances myeloid cell proliferation.37 38 To assess the cytokine-dependent growth of these cells, wild-type MO7e and transduced MO7e-G cells were starved for 18 hours and then cultured in the presence or absence of growth factors (Figure 1B). Parental MO7e cells are factor dependent and neither proliferate nor survive without GM-CSF. The expression of the transduced G-CSF receptor enables MO7e-G cells to proliferate in response to G-CSF in the absence of GM-CSF. This was not due to an interaction between G-CSF and the GM-CSF receptor, because wild-type MO7e cells neither proliferated nor survived in response to G-CSF. Moreover, although SCF had a minimal effect on the proliferation of either MO7e or MO7e-G, the combination of SCF and G-CSF led to a greater-than-additive enhancement of proliferation over that seen with G-CSF alone in MO7e-G cells.

Cotreatment of MO7e-G cells with stem cell factor and granulocyte colony-stimulating factor shortens the G1/G0 phase of the cell cycle

Given the likelihood that the enhanced proliferative state induced by cotreatment with SCF and G-CSF of MO7e-G cells resulted from modifications in cell cycle control, we analyzed the cell cycle distribution induced by these cytokines. Cells were cultured as above, and stained with propidium iodide, after which cell cycle distribution was determined by measuring DNA content with flow cytometry (Figure2A). SCF induced no significant changes in cell cycle distribution compared to untreated samples. G-CSF induced a significant reduction in the percentage of cells in G0/G1 and a parallel increase in the percentages of cells in S and G2/M. However, the combination of G-CSF and SCF led to a significant reduction in the percentage of cells in G1/G0 compared to cells treated with G-CSF alone at all time points (24 hours,P < .01; 48 and 72 hours, P < .05). The duration of each phase was calculated using the doubling times of the cultures and the percentage of cells in each phase of the cell cycle 48 hours after initiation of the culture.39 No significant difference was found in the duration of S and G2/M in cells cultured in G-CSF alone or in combination with SCF. However, a marked shortening of the duration of the G0/G1 was seen in cells cultured with G-CSF and SCF compared to those cultured in G-CSF alone (3.5 versus 8.0 hours; Figure 2B).

Fig. 2.

Cytokine treatment of MO7e-G cells with G-CSF and SCF leads to a shortening of the G1/G0 phase of the cell cycle and a decrease in p27kip-1.

MO7e-G cells were starved overnight and then cultured with cytokines as indicated. At each time point, cells were stained with propidium iodide, and DNA content was determined by flow cytometry. (A) The percentage of cells in each phase of the cell cycle was determined (1 representative experiment of 4 is presented). (B) The duration of each phase was derived from the cell cycle distribution at 48 hours (steady-state exponential growth) and the doubling time of the population. (C,D) MO7e-G cells were cultured as described for the indicated times, after which whole cell extracts were prepared and immunoblots were performed to Rb, p27kip-1, and tubulin.

Fig. 2.

Cytokine treatment of MO7e-G cells with G-CSF and SCF leads to a shortening of the G1/G0 phase of the cell cycle and a decrease in p27kip-1.

MO7e-G cells were starved overnight and then cultured with cytokines as indicated. At each time point, cells were stained with propidium iodide, and DNA content was determined by flow cytometry. (A) The percentage of cells in each phase of the cell cycle was determined (1 representative experiment of 4 is presented). (B) The duration of each phase was derived from the cell cycle distribution at 48 hours (steady-state exponential growth) and the doubling time of the population. (C,D) MO7e-G cells were cultured as described for the indicated times, after which whole cell extracts were prepared and immunoblots were performed to Rb, p27kip-1, and tubulin.

Close modal

Stem cell factor/granulocyte colony-stimulating factor–induced shortening of G0/G1 is associated with a loss of expression of the cyclin-dependent kinase inhibitor p27kip-1

Because the proliferative synergy between SCF and G-CSF is mediated through a shortening of G0/G1, we investigated the effect of G-CSF and SCF cotreatment on several key regulatory components of the G1-S transition. Rb, a critical mediator of the progression from G1 to S phase,40,41 is modulated through phosphorylation by cyclin-dependent kinases (cdks).42 The regulation of the activity of these cdks is critical for progression through the cell cycle.43 In particular, cdk inhibitors (cdkis) such as p27kip-1 or p21cip-1 block cdk activity and prevent progression from G1 to S phase.44-46To analyze the effect of SCF and G-CSF on Rb phosphorylation, MO7e-G cells were starved and then cultured in the presence or absence of cytokines. Both SCF and G-CSF induced hyperphosphorylation of Rb, as measured by decreased gel mobility of Rb as seen in a Western blot (Figure 2C). However, the hyperphosphorylation induced by SCF alone was not complete at any of the time points, because the fastest-migrating hypophosphorylated band was always present. By contrast, the combination of SCF and G-CSF, or G-CSF alone, induced complete hyperphosphorylation of Rb. To investigate the role that cdkis play in this cytokine-induced Rb-phosphorylation, p27kip-1 levels were analyzed by Western blot (Figure 2D). SCF alone did not induce a decrease in p27kip-1 protein level (lanes 1, 2, 5, and 6). Although G-CSF induced a 22% decrease of p27kip-1 protein level at 24 hours compared to untreated cells (lane 7), the combination of SCF and G-CSF induced a 90% reduction in p27kip-1protein level (lane 8). No differences were found in the expression of p21cip-1 among the different treatments at any time point (data not shown). Thus, the combination of G-CSF and SCF leads to specific suppression of p27kip-1 in MO7e-G cells.

Costimulation of MO7e-G cells with stem cell factor and granuloctye colony-stimulating factor leads to an increased expression of c-fos

Because cellular function is dictated by the pattern of gene expression, we analyzed the activation of c-fos, a proto-oncogene involved in cell cycle progression47 48 in response to SCF and G-CSF. MO7e-G cells were starved overnight and then treated with SCF or G-CSF alone or in combination for 30 minutes. Total RNA was harvested and analyzed by semiquantitative RT-PCR (Figure 3). Although SCF was unable to induce c-fos expression, G-CSF at high concentrations (1 ng/mL) but not at low concentrations induced modest expression of this proto-oncogene. Costimulation of MO7e-G cells with SCF and G-CSF led to a marked increase in the expression of c-fos compared with cells treated with G-CSF alone. Thus, SCF and G-CSF, which show synergistic effects in inducing cellular proliferation, also lead to marked enhancement in inducing the expression of a proto-oncogene involved in cell cycle progression.

Fig. 3.

Treatment of MO7e-G cells with the combination of G-CSF and SCF leads to synergistic induction of c-

fos. MO7e-G cells were factor starved overnight, then treated for 30 minutes with SCF (100 ng/mL), G-CSF (1 or 100 ng/mL) or both. RNA was harvested and analyzed by semiquantitative RT-PCR with primers specific for c-fos (upper) andGAPDH (lower).

Fig. 3.

Treatment of MO7e-G cells with the combination of G-CSF and SCF leads to synergistic induction of c-

fos. MO7e-G cells were factor starved overnight, then treated for 30 minutes with SCF (100 ng/mL), G-CSF (1 or 100 ng/mL) or both. RNA was harvested and analyzed by semiquantitative RT-PCR with primers specific for c-fos (upper) andGAPDH (lower).

Close modal

Granulocyte colony-stimulating factor–induced activation of STAT1 and STAT3 is unaffected by stem cell factor

The enhanced activation of c-fos by cotreatment with SCF and G-CSF suggested that one or more signaling pathways activated by these factors was amplified when the cytokines were administered together.23,27,28 In particular, STATs are known to mediate the effects of many cytokines in hematopoietic cells, including G-CSF,49-52 and have been found to control cellular proliferation by regulating p27kip-1expression.53 Therefore, we explored the possibility that enhanced STAT activation mediated the synergistic effects of SCF and G-CSF on proliferation, p27kip-1 regulation, and c-fos induction. To analyze this, MO7e-G cells were factor starved overnight and treated with SCF, G-CSF, or their combination. Whole cell extracts were obtained and Western blots were performed to detect the activated tyrosine-phosphorylated forms of STAT1 and STAT3. G-CSF alone led to phosphorylation of STAT1 and STAT3 (Figure4A; lanes 3 and 4), whereas SCF did not induce any phosphorylation of these STATs (lane 2). The addition of SCF to G-CSF failed to induce any detectable change in the level of tyrosine phosphorylation of either STAT1 or STAT3 compared to that induced by G-CSF alone (lanes 5 and 6). This suggests that the synergy between SCF and G-CSF is not mediated at the level of STAT tyrosine phosphorylation. This is further supported by the finding that SCF did not enhance the phosphorylation of JAK1 and JAK2, the tyrosine kinases activated by G-CSF that mediate STAT tyrosine phosphorylation (data not shown).

Fig. 4.

Tyrosine phosphorylation and DNA binding of STAT1 and STAT3 induced by G-CSF is not affected by SCF.

MO7e-G cells were factor starved overnight, then treated for 15 minutes with SCF (100 ng/mL), G-CSF (1 or 100 ng/mL), or both. (A) Total cell extracts were prepared and Western blots were performed to detect total and tyrosine-phosphorylated forms of STAT1 and STAT3. The same membrane was stripped and reprobed for each blot. (B) Nuclear extracts were prepared and binding was assessed to a 32P-labeled double-stranded oligonucleotide containing a STAT-binding high-affinity sequence. G-CSF was used at a concentration of 1 ng/mL. Ia, Ib, and Ic indicate specific STAT-DNA complexes; II, “super-shifted” complexes; III, nonspecific complexes.

Fig. 4.

Tyrosine phosphorylation and DNA binding of STAT1 and STAT3 induced by G-CSF is not affected by SCF.

MO7e-G cells were factor starved overnight, then treated for 15 minutes with SCF (100 ng/mL), G-CSF (1 or 100 ng/mL), or both. (A) Total cell extracts were prepared and Western blots were performed to detect total and tyrosine-phosphorylated forms of STAT1 and STAT3. The same membrane was stripped and reprobed for each blot. (B) Nuclear extracts were prepared and binding was assessed to a 32P-labeled double-stranded oligonucleotide containing a STAT-binding high-affinity sequence. G-CSF was used at a concentration of 1 ng/mL. Ia, Ib, and Ic indicate specific STAT-DNA complexes; II, “super-shifted” complexes; III, nonspecific complexes.

Close modal

Although SCF did not enhance the tyrosine phosphorylation of STAT1 and STAT3 induced by G-CSF, we considered the possibility that SCF might amplify the actions of G-CSF by increasing STAT translocation or binding to DNA. To address this possibility, nuclear extracts were prepared from cells treated with SCF and G-CSF alone and in combination. The nuclear extracts were incubated with32P-double-stranded oligonucleotide containing a high-affinity STAT-binding sequence,34 and protein-DNA complexes were analyzed in an electrophoretic mobility shift assay (Figure 4B). G-CSF treatment led to the induction of 3 protein-DNA complexes (lane 2; complexes Ia, Ib, and Ic) whose specificity was demonstrated by their abolition by inclusion of a 100-fold excess of unlabeled probe in the binding reaction (lane 11). By contrast, a higher mobility nonspecific complex (complex III) was unaffected by excess unlabeled probe. That these complexes contain STAT1 and STAT3 was demonstrated by including antibodies to each of these proteins in the binding reaction. Antibody to STAT1 abolished the high-mobility complex Ic, and antibody to STAT3 diminished complex Ib while inducing formation of a low-mobility band (complex II, lane 8). It is likely that other proteins are participating in the formation of at least some of these complexes because antibodies to STAT1 and STAT3 do not completely abolish all of the protein-DNA complexes. However, antibodies to other STATs did not affect these complexes (data not shown). In contrast to G-CSF, SCF failed to induce formation of any specific protein-DNA complexes (lane 1), consistent with its inability to induce tyrosine phosphorylation of STAT1 or STAT3. Cotreatment with SCF and G-CSF did not lead to any qualitative or quantitative differences in the protein-DNA complexes induced compared to those seen with G-CSF alone (lane 3). Thus, the synergy between SCF and G-CSF is not mediated by increased STAT tyrosine phosphorylation or DNA binding.

Stem cell factor enhances ser727 phosphorylation of STAT3

The STAT transcription factors can be phosphorylated not only on tyrosine residues, but also on specific serine residues within their transactivation domain (ser727 in STAT1 and STAT3). Although tyrosine phosphorylation directly activates a STAT, ser727 phosphorylation increases the magnitude of the gene transcription mediated by the tyrosine-phosphorylated STATs and is necessary for maximal transcriptional activity.54 Thus, we tested the possibility that SCF enhanced STAT-dependent gene activation by enhancing phosphorylation of the ser727 residue. MO7e-G cells were factor starved and treated with SCF and G-CSF alone and in combination. Whole cell extracts were prepared and immunoblots were performed with antibodies specific for the tyrosine-phosphorylated and the ser727-phosphorylated form of STAT3 (Figure5A). Treatment of MO7e-G cells with SCF led to the prominent phosphorylation of STAT3 on ser727, but not on the activating tyrosine705 (lane 2). A Western blot using an antibody that recognizes all forms of STAT3 revealed that ser727-phosphorylated STAT3 migrated slightly slower than the unphosphorylated form (total-STAT3 blot; lane 2). G-CSF treatment led to both tyrosine and ser727 phosphorylation of STAT3, which added a third band in the pattern of migration of STAT3 (lane 3). Combined treatment of MO7e-G cells with SCF and G-CSF induced an increase in ser727 phosphorylation of STAT3 (lane 4), which was greater than that induced by SCF or G-CSF alone. In 4 independent experiments, the phosphorylation of STAT3 on ser727 induced by the combination was 54% greater than that induced by SCF alone and 67% greater than that induced by G-CSF alone. Cotreatment with SCF and G-CSF also abolished the fastest-migrating unphosphorylated STAT3 band, which neither G-CSF nor SCF did alone (total-STAT3 blot; lanes 1-4). SCF had no effect on the phosphorylation of STAT1 on ser727 (data not shown).

Fig. 5.

SCF and G-CSF induce ser727 phosphorylation of STAT3 through activation of distinct serine/threonine kinases.

MO7e-G cells were factor starved for 18 hours and then either left untreated or treated with SCF (100 ng/mL), G-CSF (10 ng/mL), or both. Where indicated, cells were incubated for 1 hour prior to cytokine treatment with PD98059 (100 μmol/L), wortmannin (500 nmol/L), or H7 (50 μmol/L). Western blots were performed with the indicated antibodies. The same membrane was stripped and reprobed for each blot.

Fig. 5.

SCF and G-CSF induce ser727 phosphorylation of STAT3 through activation of distinct serine/threonine kinases.

MO7e-G cells were factor starved for 18 hours and then either left untreated or treated with SCF (100 ng/mL), G-CSF (10 ng/mL), or both. Where indicated, cells were incubated for 1 hour prior to cytokine treatment with PD98059 (100 μmol/L), wortmannin (500 nmol/L), or H7 (50 μmol/L). Western blots were performed with the indicated antibodies. The same membrane was stripped and reprobed for each blot.

Close modal

Stem cell factor–induced ser727 phosphorylation of STAT3 is partially mediated by PI3 kinase and mitogen-activated protein kinase

Ser727 of STAT3 is contained in the sequence leu-pro-met-ser-pro, a motif found in substrates of proline-directed serine/threonine kinases.55 56 However, the identity of the serine/threonine kinase(s) that phosphorylate this conserved carboxy-terminus serine in STAT family members has remained elusive. To determine the upstream signaling events necessary for the serine phosphorylation of STAT3 induced by these cytokines, MO7e-G cells were factor starved and then incubated for 1 hour with inhibitors of PI3K (wortmannin), MEK1/ERK (PD98059), or p38-MAPK (SB203580). The cells were then treated with SCF and G-CSF alone and in combination. Both wortmannin and PD98059 induced a modest reduction in the level of ser727 phosphorylation of STAT3 induced by SCF, G-CSF, or SCF and G-CSF (Figure 5A). However, the combination of wortmannin and PD98059 led to a much greater inhibition of this phosphorylation for all stimuli. These data suggest that both PI3K and ERK-MAPK pathways are involved in ser727 phosphorylation of STAT3 in response to SCF and G-CSF.

Although it has been suggested that ERKs can phosphorylate STAT serine residues, the fact that PD98059 completely suppresses ERK activity (data not shown) without abolishing ser727 phosphorylation of STAT3 indicates that other kinases must be involved. Several lines of evidence indicate that p38-MAPK can mediate ser727 phosphorylation of STATs.57 58 Inhibition of p38-MAPK with 20 μmol/L SB203580 for 1 hour did not affect the induction of ser727 phosphorylation of STAT3 by SCF and G-CSF, either alone or in combination (data not shown), indicating that this kinase is not involved in ser727 phosphorylation of STAT3 in response to these cytokines.

To further examine the kinase(s) mediating the phosphorylation of STAT3 on ser727, MO7e-G cells were incubated with the serine/threonine kinase inhibitor H7,31 and then left untreated or treated with SCF and G-CSF alone or in combination. Incubation of cells with H7 for 1 hour prior to cytokine treatment led to a marked reduction in the phosphorylation of ser727-STAT3 by G-CSF (Figure 5B, lanes 3 and 7). However, H7 had a minimal effect on the induction of phosphorylation of ser727-STAT3 by SCF (lanes 2 and 6) or by the combination of both cytokines (lanes 4 and 8). These data suggest that SCF and G-CSF induce ser727 phosphorylation of STAT3 through different pathways.

Gene activation and proliferation of MO7e-G cells in response to cotreatment with stem cell factor and granulocyte colony-stimulating factor are dependent on PI3K and ERK

Given that PI3K and ERK-dependent pathways are necessary for the synergistic phosphorylation of STAT3 on ser727 in response to G-CSF and SCF, we directly tested whether inhibition of these pathways would alter gene activation or proliferation induced by this combination of cytokines. To examine the induction of the c-fosproto-oncogene, semiquantitative RT-PCR was performed on total RNA from MO7e-G cells cotreated with SCF and G-CSF for 30 minutes with or without previous incubation with wortmannin, PD98059, or the combination thereof. Treatment with either wortmannin or PD98059 partially reduced the expression of c-fos elicited by the combination of SCF and G-CSF. However, simultaneous inhibition of PI3K and ERK led to a complete abolition of this synergistic effect (Figure6A). Thus, both of these signaling pathways contribute not only to ser727 phosphorylation of STAT3, but also to the activation of a key target gene.

Fig. 6.

Proliferation and c-

fos activation induced by G-CSF and SCF require PI3K and ERK activity. (A) MO7e-G cells were factor starved overnight, incubated with the indicated compounds, then treated for 30 minutes with SCF (100 ng/mL) and G-CSF (10 ng/mL). RNA was extracted and analyzed by semiquantitative RT-PCR with primers specific for c-fos (upper) and GAPDH (lower). (B) MO7e-G cells were factor starved for 18 hours, incubated with or without the combination of PD98059 (P) and wortmannin (W), then cultured in the presence of no growth factor, SCF (100 ng/mL), G-CSF (10 ng/mL), or both. Cell counts were obtained daily and are presented as the means and SD of 4 independent experiments.

Fig. 6.

Proliferation and c-

fos activation induced by G-CSF and SCF require PI3K and ERK activity. (A) MO7e-G cells were factor starved overnight, incubated with the indicated compounds, then treated for 30 minutes with SCF (100 ng/mL) and G-CSF (10 ng/mL). RNA was extracted and analyzed by semiquantitative RT-PCR with primers specific for c-fos (upper) and GAPDH (lower). (B) MO7e-G cells were factor starved for 18 hours, incubated with or without the combination of PD98059 (P) and wortmannin (W), then cultured in the presence of no growth factor, SCF (100 ng/mL), G-CSF (10 ng/mL), or both. Cell counts were obtained daily and are presented as the means and SD of 4 independent experiments.

Close modal

To examine whether these pathways contribute to the synergistic effects on proliferation induced by SCF and G-CSF, MO7e-G cells were incubated in the presence or absence of wortmannin and PD98059, and population growth in response to SCF and G-CSF, alone and in combination, was assessed (Figure 6B). The enhanced growth seen with the combination of SCF and G-CSF was lost in the presence of these inhibitors, and population growth was reduced to that seen with G-CSF alone. Furthermore, the growth of cells cultured in G-CSF alone was not reduced further by these inhibitors. These findings indicate that the added component of growth provided by SCF requires these signaling pathways, whereas growth in the presence of G-CSF alone, or the minimal growth seen with SCF alone, is independent of PI3K and ERKs.

Although the synergy between SCF and G-CSF is of great biologic and clinical importance, the absence of a model system to explore this interaction at the cellular level has impaired the ability to understand its mechanism. In this study, we have developed a system to examine the intracellular events induced by these molecules alone and in combination by using retroviral transduction to introduce the hG-CSF receptor into the SCF-responsive human hematologic cell line, MO7e. This has demonstrated that the use of VSV-G glycoprotein-pseudotyped retroviruses is a feasible approach toward high efficiency and stable transduction of this human acute megakaryocytic leukemia-derived cell line. This may be an important strategy for the introduction of genes into other human hematologic cells and cell lines, which are difficult to transfect by other means.

The transduced G-CSF receptor is functionally active, in that it not only supports G-CSF–dependent proliferation of MO7e-G cells, but also recapitulates the proliferative synergy between SCF and G-CSF (Figure1B). Cell cycle analysis revealed that the enhanced proliferative state induced by SCF and G-CSF cotreatment was associated with a direct effect of these cytokines on cell cycle distribution, specifically a marked shortening of the duration of G0/G1. This is mediated, at least in part, by a marked decrease of expression of the cdki p27kip-1, which is known to set a stoichiometric inhibitory threshold of cdk activity that prevents cdk-induced phosphorylation of Rb and cell cycle progression.45 59 Therefore, the loss of p27kip-1 following treatment with SCF and G-CSF may facilitate cell proliferation.

Mitogenic cytokines induce the activation of immediate-early genes, which are necessary for cellular proliferation. These genes are regulated by transcription factors that are activated by cytokine-induced signaling pathways. SCF and G-CSF, which lead to a marked enhancement of proliferation in MO7e-G cells, also lead to synergistic induction of c-fos, a proto-oncogene necessary for cell cycle progression in many systems. The promoter for c-fos contains elements responsive to a number of transcription factors. Among these, we focused on STATs, which are known to be key mediators of cytokine signaling in hematopoietic cells,49-52 and have been shown to control cellular proliferation by regulating p27kip-1expression.53 As has been reported in other systems, we found that G-CSF induces tyrosine phosphorylation of STAT1 and STAT3 and their subsequent binding to specific DNA elements.60-65 By contrast, SCF failed to induce activation of these STATs in MO7e-G cells. The ability of SCF to activate STATs remains controversial.66-68 In MO7e cells, SCF has been shown to induce activation of STAT1 in some reports,69,70 but not in others.71,72 SCF has not been found to activate STAT3 in any cell type, including MO7e.71 No evidence was found that the synergy between SCF and G-CSF occurs at the level of STAT1 or STAT3 tyrosine phosphorylation, nuclear translocation, or DNA binding (Figure 4).

Although tyrosine phosphorylation is required for STAT dimerization and nuclear translocation, the magnitude of STAT1- or STAT3-mediated (or both) gene induction is modulated by phosphorylation on ser727 within the transactivation domain of both STATs.73,74 We found that SCF induces ser727 phosphorylation of STAT3 in MO7e-G cells, as has been reported by others.71 Furthermore, the combination of SCF and G-CSF maximized the induction of ser727 phosphorylation of STAT3. In fact, all of the STAT3 detectable by Western blot is completely phosphorylated on this residue after treatment with SCF and G-CSF, whereas neither G-CSF nor SCF alone induce this complete shift to the slow-migrating form of STAT3 (Figure 5A). Ser727 is located within a proline-directed serine-threonine kinase consensus site,55,56 which can be phosphorylated by MAPK family members including ERKs75-79 and p38MAPK.57,58,80 81 We found that both PI3K and ERK are upstream of ser727 phosphorylation of STAT3 in MO7e-G cells, but that SCF and G-CSF induce phosphorylation of this residue through different pathways. SCF-induced phosphorylation of STAT3 on ser727 is insensitive to H7, whereas G-CSF is highly sensitive to this kinase inhibitor. Moreover, the fact that complete ERK inhibition does not fully block STAT3 ser727 phosphorylation is additional evidence that even if ERK can phosphorylate ser727, other kinases must be involved in this phosphorylation.

Because PI3K and ERK-dependent pathways are necessary for the synergistic phosphorylation of STAT3 on ser727, we explored whether inhibition of these pathways would affect gene activation or proliferation elicited by cotreatment with SCF and G-CSF. Simultaneous inhibition of these 2 pathways led to complete abolition of the synergistic induction of c-fos. Furthermore, the proliferative synergy between SCF and G-CSF was also completely dependent on PI3K and ERK, because the growth of MO7e-G cells in response to SCF and G-CSF was reduced to that seen with G-CSF alone in the presence of inhibitors of these pathways (Figure 6B).

We have described a novel system to explore the signaling events underlying the important biologic interaction between SCF and G-CSF. MO7e-G cells recapitulate the proliferative synergy between SCF and G-CSF and have allowed us to identify a number of intracellular pathways that may mediate their biologic actions. It remains to be determined whether other pathways play a role in the biologic and clinical effects of SCF and G-CSF.

We thank Drs J. Griffin, G. Dranoff, V. Boussiotis, and S. Corey for generously sharing reagents, and J. Daley and S. Lazo for assistance with flow cytometry.

Department of Adult Oncology, Dana-Farber Cancer Institute; and Departments of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA.

Submitted May 24, 2000; accepted July 26, 2000.

Supported by a grant from the NIH (CA79547) and the Brent Leahey Fund.

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
Maze
R
Miyazawa
K
et al
The Kit receptor and its ligand, Steel factor, as regulators of hemopoiesis.
Cancer Cells.
3
1991
480
487
2
Morstyn
G
Brown
S
Gordon
M
et al
Stem cell factor is a potent synergistic factor in hematopoiesis.
Oncology.
51
1994
205
214
3
Hoffman
R
Tong
J
Brandt
J
et al
The in vitro and in vivo effects of stem cell factor on human hematopoiesis.
Stem Cells.
11
1993
76
82
4
Broxmeyer
HE
Lu
L
Cooper
S
Ruggieri
L
Li
Z
Lyman
SD
Flt-3-ligand stimulates/co-stimulates the growth of myeloid stem/progenitor cells.
Exp Hematol.
23
1995
1121
1129
5
McNiece
IK
Langley
KE
Zsebo
KM
Recombinant human stem cell factor synergises with GM-CSF, G-CSF, IL-3 and Epo to stimulate human progenitor cells of the myeloid and erythroid lineages.
Exp Hematol.
19
1991
226
231
6
Sui
X
Krantz
SB
You
M
Zhao
Z
Synergistic activation of MAP kinase (ERK1/2) by erythropoietin and stem cell factor is essential for expanded erythropoiesis.
Blood.
92
1998
1142
1149
7
Horie
M
Broxmeyer
HE
The combination of Steel factor and GM-CSF blocks apoptosis induced by retinoic acid and upregulates AP-1 in a human growth factor-dependent cell line.
Exp Hematol.
23
1995
168
173
8
Hendrie
PC
Miyazawa
K
Yang
YC
Langefeld
CD
Broxmeyer
HE
Mast cell growth factor (c-kit ligand) enhances cytokine stimulation of proliferation of the human factor-dependent cell line, MO7e.
Exp Hematol.
19
1991
1031
1037
9
Keller
JR
Ortiz
M
Ruscetti
FW
Steel factor (c-kit ligand) promotes the survival of hematopoietic stem/progenitor cells in the absence of cell division.
Blood.
86
1995
1757
1764
10
Gehling
UM
Ryder
JW
Hogan
CJ
et al
Ex vivo expansion of megakaryocyte progenitors: effect of various growth factor combinations on CD34+ progenitor cells from bone marrow and G-CSF-mobilized peripheral blood.
Exp Hematol.
25
1997
1125
1139
11
Fehniger
TA
Carson
WE
Mrozek
E
Caligiuri
MA
Stem cell factor enhances interleukin-2mediated expansion of murine natural killer cells in vivo.
Blood.
90
1997
3647
3653
12
Uittenbogaart
CH
Schmidc
I
Kiertscher
S
et al
Human thymocyte responsiveness to stem cell factor: synergy with interleukin-2 for the generation of NK/LAK cytotoxicity.
Immunol Lett.
52
1996
45
52
13
Avraham
H
Vannier
E
Cowley
S
et al
Effects of the stem cell factor, c-kit ligand, on human megakaryocytic cells.
Blood.
79
1992
365
371
14
Nishi
N
Ishikawa
R
Inoue
H
et al
Granulocyte-colony stimulating factor and stem cell factor are the crucial factors in long-term culture of human primitive hematopoietic cells supported by a murine stromal cell line.
Exp Hematol.
24
1996
1312
1321
15
Briddell
RA
Hartley
CA
Smith
KA
McNiece
IK
Recombinant rat stem cell factor synergises with recombinant human granulocyte-colony stimulating factor in vivo in mice to mobilize peripheral blood progenitor cells that have enhanced repopulating potential.
Blood.
82
1993
1720
1723
16
Andrews
RG
Briddell
RA
Knitter
GH
Rowley
SD
Appelbaum
FR
McNiece
IK
Rapid engraftment by peripheral blood progenitor cells mobilized by recombinant human stem cell factor and recombinant human granulocyte colony-stimulating factor in nonhuman primates.
Blood.
85
1995
15
20
17
de Revel
T
Appelbaum
FR
Storb
R
et al
Effects of granulocyte colony-stimulating factor and stem cell factor, alone and in combination, on the mobilization of peripheral blood cells that engraft lethally irradiated dogs.
Blood.
83
1994
3795
3799
18
Hartung
G
Uharek
L
Zeis
M
et al
Superior antileukemic activity of murine peripheral blood progenitor cell (PBPC) grafts mobilized by G-CSF and stem cell factor (SCF) as compared to G-CSF alone.
Bone Marrow Transplant.
21
1998
S16
S20
19
Facon
T
Harousseau
JL
Maloisel
F
et al
Stem cell factor in combination with filgrastim after chemotherapy improves peripheral blood progenitor cell yield and reduces apheresis requirements in multiple myeloma patients: a randomized, controlled trial.
Blood.
94
1999
1218
1225
20
Shpall
EJ
Wheeler
CA
Turner
SA
et al
A randomized phase 3 study of peripheral blood progenitor cell mobilization with stem cell factor and filgrastim in high-risk breast cancer patients.
Blood.
93
1999
2491
2501
21
Moskowitz
CH
Stiff
P
Gordon
MS
et al
Recombinant methionyl human stem cell factor and filgrastim for peripheral blood progenitor cell mobilization and transplantation in non-Hodgkin's lymphoma patients—results of a phase I/II trial.
Blood.
89
1997
3136
3147
22
Ihle
JN
Cytokine receptor signaling.
Nature.
377
1995
591
594
23
Tidow
N
Welte
K
Advances in understanding postreceptor signaling in response to granulocyte-colony stimulating factor.
Curr Opin Hematol.
4
1997
171
175
24
Witte
ON
Steel locus defines new multipotent growth factor.
Cell.
63
1990
5
8
25
Zsebo
KM
Williams
DA
Geissler
EN
et al
Stem cell factor is encoded at the Sl locus of the mouse and is the ligand for the c-kit tyrosine kinase receptor.
Cell.
63
1990
213
224
26
Huang
E
Nocka
K
Beier
DR
et al
The hematopoietic growth factor KL is encoded by the Sl locus and is the ligand of the c-kit receptor, the gene product of the W locus.
Cell.
63
1990
225
233
27
Aronica
SM
Broxmeyer
HE
Advances in understanding the postreceptor mechanisms of action of GM-CSF, G-CSF and Steel factor.
Curr Opin Hematol.
3
1996
185
190
28
Linnekin
D
Mou
S
Deberry
CS
et al
Stem cell factor, the JAK-STAT pathway and signal transduction.
Leuk Lymphoma.
27
1997
439
444
29
Frank
DA
Robertson
MJ
Bonni
A
Ritz
J
Greenberg
ME
Interleukin 2 signaling involves the phosphorylation of novel Stat proteins.
Proc Natl Acad Sci U S A.
92
1995
7779
7783
30
Bonni
A
Sun
Y
Nadal-Vicens
M
et al
Regulation of gliogenesis in the central nervous system by the JAK-STAT signaling pathway.
Science.
278
1997
477
483
31
Frank
DA
Mahajan
S
Ritz
J
B lymphocytes from patients with chronic lymphocytic leukemia contain signal transducer and activator of transcription (STAT) 1 and STAT3 constitutively phosphorylated on serine residues.
J Clin Invest.
100
1997
3140
3148
32
Avanzi
GC
Lista
P
Giovinazzo
B
et al
Selective growth response to IL-3 of a human leukaemic cell line with megakaryoblastic features.
Br J Hematol.
69
1988
359
366
33
Ory
DS
Neugeboren
BA
Mulligan
RC
A stable human-derived packaging cell line for production of high titer retrovirus/vesicular stomatitis virus G pseudotypes.
Proc Natl Acad Sci U S A.
93
1996
11400
11406
34
Bonni
A
Frank
DA
Schindler
C
Greenberg
ME
Characterization of a pathway for ciliary neurotrophic factor signaling to the nucleus.
Science.
262
1993
1575
1579
35
Corey
SJ
Dombrosky-Ferlan
PM
Zuo
S
et al
Requirement of Src kinase Lyn for induction of DNA synthesis by granulocyte colony-stimulating factor.
J Biol Chem.
273
1998
3230
3235
36
Sattler
M
Winkler
T
Verma
S
et al
Hematopoietic growth factors signal through the formation of reactive oxygen species.
Blood.
93
1999
2928
2935
37
Molineux
G
Migdalska
A
Szmitkowski
M
Zsebo
K
Dexter
TM
The effects on hematopoiesis of recombinant stem cell factor (ligand for c-kit) administered in vivo to mice either alone or in combination with granulocyte colony-stimulating factor.
Blood.
78
1991
961
966
38
Ulich
TR
del Castillo
J
McNiece
IK
et al
Stem cell factor in combination with granulocyte colony-stimulating factor (CSF) or granulocyte-macrophage CSF synergistically increases granulopoiesis in vivo.
Blood.
78
1991
1954
1962
39
Yen
A
Williams
M
Platko
JD
Der
C
Hisaka
M
Expression of activated RAF accelerates cell differentiation and RB protein down-regulation but not hypophosphorylation.
Eur J Cell Biol.
65
1994
103
113
40
Buchkovich
K
Dyson
N
Whyte
P
Harlow
E
The retinoblastoma protein is phosphorylated during specific phases of the cell cycle.
Cell.
58
1989
1097
1105
41
Chen
PL
Scully
P
Shew
JY
Wang
JY
Lee
WH
Phosphorylation of the retinoblastoma gene product is modulated during the cell cycle and cellular differentiation.
Cell.
58
1989
1193
1198
42
Hinds
PW
Mittnacht
S
Dulic
V
Arnold
A
Reed
SI
Weinberg
RA
Regulation of retinoblastoma protein functions by ectopic expression of human cyclins.
Cell.
70
1992
993
1006
43
Ohtsubo
M
Roberts
JM
Cyclin-dependent regulation of G1 in mammalian fibroblasts.
Science.
259
1993
1908
1912
44
Peter
M
Herskowitz
I
Joining the complex: cyclin-dependent kinase inhibitory proteins and the cell cycle.
Cell.
79
1994
181
184
45
Kato
JY
Matsuoka
M
Polyak
K
Massague
J
Sherr
CJ
Cyclic AMP-induced G1 phase arrest mediated by an inhibitor (p27Kip1) of cyclin-dependent kinase 4 activation.
Cell.
79
1994
487
496
46
el-Deiry
WS
Tokino
T
Velculescu
VE
et al
WAF1, a potential mediator of p53 tumor suppression.
Cell.
75
1993
817
825
47
Johansen
FE
Prywes
R
Serum response factor: transcriptional regulation of genes induced by growth factors and differentiation.
Biochim Biophys Acta.
1241
1995
1
10
48
Rivera
VM
Greenberg
ME
Growth factor-induced gene expression: the ups and downs of c-fos regulation.
New Biol.
2
1990
751
758
49
Darnell
JE
Jr
Kerr
IM
Stark
GR
Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins.
Science.
264
1994
1415
1421
50
Leonard
WJ
O'Shea
JJ
Jaks and STATs: biological implications.
Annu Rev Immunol.
16
1998
293
322
51
Frank
DA
STAT signaling in the pathogenesis and treatment of cancer.
Mol Med.
5
1999
432
456
52
Ward
AC
Touw
I
Yoshimura
A
The Jak-Stat pathway in normal and perturbed hematopoiesis.
Blood.
95
2000
19
29
53
Kaplan
MH
Daniel
C
Schindler
U
Grusby
MJ
Stat proteins control lymphocyte proliferation by regulating p27Kip1 expression.
Mol Cell Biol.
18
1998
1996
2003
54
Wen
Z
Zhong
Z
Darnell
JE
Jr
Maximal activation of transcription by Stat1 and Stat3 requires both tyrosine and serine phosphorylation.
Cell.
82
1995
241
250
55
Clark-Lewis
I
Sanghera
JS
Pelech
SL
Definition of a consensus sequence for peptide substrate recognition by p44mpk, the meiosis-activated myelin basic protein kinase.
J Biol Chem.
266
1991
15180
15184
56
Gonzalez
FA
Raden
DL
Davis
RJ
Identification of substrate recognition determinants for human ERK1 and ERK2 protein kinases.
J Biol Chem.
266
1991
22159
22163
57
Gollob
JA
Schnipper
CP
Murphy
EA
Ritz
J
Frank
DA
The functional synergy between IL-12 and IL-2 involves p38 MAP kinase and is associated with the augmentation of STAT serine phosphorylation.
J Immunol.
162
1999
4472
4481
58
Turkson
J
Bowman
T
Adnane
J
et al
Requirement for Ras/Rac1-mediated p38 and c-Jun N-terminal kinase signaling in Stat3 transcriptional activity induced by the Src oncoprotein.
Mol Cell Biol.
19
1999
7519
7528
59
Polyak
K
Lee
MH
Erdjument-Bromage
H
et al
Cloning of p27kip-1 a cyclin-cdk inhibitor and a potential mediator of extracellular antimitogenic signals.
Cell.
78
1994
59
66
60
Shimozaki
K
Nakajima
K
Hirano
T
Nagata
S
Involvement of STAT3 in the granulocyte colony-stimulating factor-induced differentiation of myeloid cells.
J Biol Chem.
272
1997
25184
25189
61
Shin-Shay
T
Lamb
P
Seidel
HM
Stein
RB
Rosen
J
Rapid activation of the STAT3 transcription factor by granulocyte colony-stimulating factor.
Blood.
84
1994
1760
1764
62
Ward
AC
Hermans
MHA
Smith
L
et al
Tyrosine-dependent and -independent mechanisms of STAT3 activation by the human granulocyte colony-stimulating factor (G-CSF) receptor are differentially utilized depending on G-CSF concentration.
Blood.
93
1999
113
124
63
Chakraborty
A
Dyer
KF
Cascio
M
Mietzner
TA
Tweardy
DJ
Identification of a novel Stat3 recruitment and activation motif within the granulocyte colony-stimulating factor receptor.
Blood.
93
1999
15
24
64
Nicholson
SE
Novak
U
Ziegler
SF
Layton
JE
Distinct regions of the granulocyte colony-stimulating factor receptor are required for tyrosine phosphorylation of the signaling molecules JAK2, Stat3, and p42, p44MAPK.
Blood.
86
1995
3698
3704
65
Ward
AC
Smith
L
de Koning
JP
van Aesch
Y
Touw
IP
Multiple signals mediate proliferation, differentiation, and survival from the granulocyte colony-stimulating factor receptor in myeloid 32D cells.
J Biol Chem.
274
1999
14956
14962
66
Jacobs-Helber
SM
Penta
K
Sun
Z
Lawson
A
Sawyer
ST
Distinct signaling from stem cell factor and erythropoietin in HCD57 cells.
J Biol Chem.
272
1997
6850
6853
67
O'Farrell
AM
Ichihara
M
Mui
AL
Miyajima
A
Signaling pathways activated in a unique mast cell line where interleukin-3 supports survival and stem cell factor is required for a proliferative response.
Blood.
87
1996
3655
3668
68
Ryan
JJ
Huang
H
McReynolds
LJ
et al
Stem cell factor activates STAT-5 DNA binding in IL-3-derived bone marrow mast cells.
Exp Hematol.
25
1997
357
362
69
Deberry
C
Mou
S
Linnekin
D
Stat1 associates with c-kit and is activated in response to stem cell factor.
Biochem J.
327
1997
73
80
70
Brizzi
MF
Dentelli
P
Rosso
A
Yarden
Y
Pegoraro
L
STAT protein recruitment and activation in c-Kit deletion mutants.
J Biol Chem.
274
1999
16965
16972
71
Gotoh
A
Takahira
H
Mantel
C
Litz-Jackson
S
Boswell
HS
Broxmeyer
HE
Steel factor induces serine phosphorylation of Stat3 in human growth factor-dependent myeloid cell lines.
Blood.
88
1996
138
145
72
Yip-Schneider
MT
Horie
M
Broxmeyer
HE
Transcriptional induction of pim-1 protein kinase gene expression by interferon gamma and posttranscriptional effects on costimulation with steel factor.
Blood.
85
1995
3494
3502
73
Eilers
A
Georgellis
D
Klose
B
et al
Differentiation-regulated serine phosphorylation of STAT1 promotes GAF activation in macrophages.
Mol Cell Biol.
15
1995
3579
3586
74
Zhang
X
Blenis
J
Li
HC
Schindler
C
ChenKiang
S
Requirement of serine phosphorylation for formation of STAT-promoter complexes.
Science.
267
1995
1990
1994
75
Chung
J
Uchida
E
Grammer
TC
Blenis
J
STAT3 serine phosphorylation by ERKdependent and -independent pathways negatively modulates its tyrosine phosphorylation.
Mol Cell Biol.
17
1997
6508
6516
76
David
M
Petricoin
Er
Benjamin
C
Pine
R
Weber
MJ
Larner
AC
Requirement for MAP kinase (ERK2) activity in interferon alpha- and interferon beta-stimulated gene expression through STAT proteins.
Science.
269
1995
1721
1723
77
Sengupta
TK
Talbot
ES
Scherle
PA
Ivashkiv
LB
Rapid inhibition of interleukin-6 signaling and Stat3 activation mediated by mitogen-activated protein kinases.
Proc Natl Acad Sci U S A.
95
1998
11107
11112
78
Jain
N
Zhang
T
Fong
SL
Lim
CP
Cao
X
Repression of Stat3 activity by activation of mitogen-activated protein kinase (MAPK).
Oncogene.
17
1998
3157
3167
79
Ng
J
Cantrell
D
STAT3 is a serine kinase target in T lymphocytes: interleukin 2 and T cell antigen receptor signals converge upon serine 727.
J Biol Chem.
272
1997
24542
24549
80
van Puijenbroek
AA
van der Saag
PT
Coffer
PJ
Cytokine signal transduction in P19 embryonal carcinoma cells: regulation of Stat3-mediated transactivation occurs independently of p21ras-Erk signaling.
Exp Cell Res.
251
1999
465
476
81
Goh
KC
Haque
SJ
Williams
BR
p38 MAP kinase is required for STAT1 serine phosphorylation and transcriptional activation induced by interferons.
EMBO J.
18
1999
5601
5608

Author notes

David A. Frank, Department of Adult Oncology, Dana-Farber Cancer Institute, 44 Binney St, Boston, MA 02115; e-mail: david_frank@dfci.harvard.edu

Sign in via your Institution