KIT is a tyrosine kinase receptor that is aberrantly activated in several neoplasms. In human pathologies, the most frequent mutation of KIT occurs at codon 816. The resulting KIT mutant protein is activated in the absence of ligand and is resistant to the clinically available inhibitors of KIT. In this report, we provide evidence for an essential function of the cytoplasmic tyrosine kinase FES downstream of KITD816V. FES is phosphorylated on tyrosine residues in cells that carry KITD816V mutation, and this phosphorylation is KIT dependent. Reduction of FES expression using RNA interference results in decreased cell proliferation in human or murine cells harboring KITD816V or the homologous mouse mutation KITD814Y. The reduced cell growth can be rescued using another cytokine (granulocyte-macrophage colony-stimulating factor [GM-CSF]) and is not observed when the closely related fer gene is targeted. Finally, signaling downstream of KITD816V is altered in cells lacking FES expression. This study shows a major function of FES downstream of activated KIT receptor and thereby points to FES as a novel target in KIT-related pathologies.

The proto-oncogene c-kit encodes a receptor with tyrosine kinase activity which is essential for hematopoiesis and the development of melanoblasts, germ cells, and interstitial cells of Cajal. Gain-of-function mutations of KIT are implicated in human pathologies. These mutations have been classified as regulatory-type mutations when they affect domains of the receptor necessary for maintaining KIT autoinhibition and structural-type mutations when they directly affect the catalytic domain. Juxtamembrane mutations of KIT are commonly found in gastrointestinal stromal tumors (GISTs),1  whereas kinase domain mutations are mainly found in mastocytosis,2  hematologic neoplasms,3,5  and germ-cell tumors.6,8  Very recently, both juxtamembrane and kinase domain mutations of KIT have also been reported in melanomas.9  The most frequent mutation affects codon 816 in human c-kit cDNA, which corresponds to codon 814 in the homologous mouse sequence.10  Whereas tyrosine kinase inhibitors such as imatinib are very efficient on wild-type KIT (KITWT) and KIT juxtamembrane mutations, KITD816V is resistant to this treatment.11,13  Therefore, drugs that could target this mutant or an essential downstream effector would be useful tools for the study and the treatment of the associated diseases.

Signaling proteins activated downstream of gain-of-function mutants of KIT include many proteins shown previously to participate in wild-type KIT receptor signal transduction.14,15  They include the classical phosphatidylinositol 3-kinase (PI3K) and extracellular signal-regulated kinase (ERK) pathways, as well as a number of signaling proteins activated by many cell-surface receptors such as p38, JNK, VAV, STAT-1, STAT-3, and STAT-5. Activation of PI3K is critical for both WT and mutant KIT in all models studied, while the activation of the mitogen-activated protein (MAP) kinases ERK-1 and ERK-2 appears to be dependent on the cellular context. The importance of each protein or signaling pathway activated downstream of KITD816V has yet to be fully evaluated.

Fps/fes was originally identified as an oncogene from avian (fps) and feline (fes) retroviruses. FES (feline sarcoma) and FER (FES-related) proteins are the only 2 members of a subfamily of cytoplasmic protein tyrosine kinase.16  FES has been shown to associate with growth factor and cytokine receptors,17,,20  which leads to activation of its catalytic activity through tyrosine phosphorylation. The downstream effectors of FES are not clearly characterized, although several studies point to a link between FES and several signaling proteins such as STATs,21,23  the MAP kinases ERK-1 and ERK-2,24,25  and PI3K.26  Several mouse models of FES have been generated with either a null allele,23,27  a knock-in of an inactive kinase, 22,28  a truncated FES protein,29  or transgenic mice expressing an active form of FES.25,30,31  They all showed a role for FES, albeit nonessential, in the homeostasis of the myeloid cell compartment, and a role in innate immunity has been highlighted in the knock-out mice.23,27,32  Mutations of FES have been recently reported in colorectal cancers.33  The mutant forms of FES appeared to have reduced kinase activity rather than being gain-of-function mutations.34,35  To date, the function of FES in normal physiology and in pathology remains enigmatic.34 

This study was initiated to identify novel proteins required for KITD816V signaling. FES was found to interact with activated KIT receptor in yeast and was identified as a potentially phosphorylated protein in cells that express the KITD816V mutant. In human and mouse cell lines, FES is indeed phosphorylated downstream of KITD816V and downstream of the murine homolog KITD814Y, respectively. Using RNA interference to reduce FES protein expression, we show that FES is required for KITD816V-dependent cell proliferation both in cellular models with transfected KITD816V and in a mastocytoma cell line with endogenous expression of the mutant. In the absence of FES, the rate of proliferation is reduced due to slow progression of cells from G1 to S phase. Furthermore, cells treated with fes siRNA showed aberrant activation of STAT proteins and of mTOR pathway through p70 S6 kinase, suggesting a function for FES upstream of these signaling proteins.

Yeast 2-hybrid screen

A yeast 2-hybrid screen was carried out as described previously.36,37  In brief, human c-kit cDNAs were expressed as the LexA fusion protein using the pBTM116 vector in the L40 yeast strain. A cDNA library cloned in pVP16 derived from the multipotential hematopoietic cell line EML-c1 (kindly provided by Dr S. Tsai, Seattle, WA) was screened using KITD816V as bait. The bait was functionally tested against the SH2 domains of p85 and growth factor receptor-bound protein 2 (GRB-2) before performing the screen.

Cells

TF-1 is a human erythroleukemia cell line, and TF-1 KITD816V cells are TF-1 cells stably transfected with the KITD816V receptor.38  P815 is a mouse mastocytoma cell line with an endogenous KITD814Y mutation. D816VKIT bone marrow-derived mast cells (BMMCs) are from transgenic mice expressing KITD816V.38  HMC-1 is a human mast cell line heterozygous for 2 mutations located on the same allele of c-kit: V560G and D816V (kindly provided by Dr J. H. Butterfield, Rochester, MN). All cells were maintained in RPMI 1640 medium, supplemented with 10% fetal bovine serum, 2 mM l-glutamine, and 100 U/mL penicillin-streptomycin (all from Invitrogen, Carlsbad, CA) and grown at 37°C in 5% CO2 at constant humidity. TF-1 cells were grown in the presence of granulocyte-macrophage colony-stimulating factor (GM-CSF; Berlex, Seattle, WA) or stem cell factor (SCF; Abcys, Paris, France).

siRNA transfection

Small interfering RNA (siRNA) and their sources were as follows. fes A siRNA was an equimolar mix of 2 RNA duplexes which target the human sequences 5′-GGCCAAGUUUCUACAGGAA-3′ and 5′-GCCUGAGGCUGAGUACCAA-3′ (Qiagen, Valencia, CA). The human fes B siRNA targeted the human c-fes sequence 5′-CCAACAUCGUGCGUCUCAUUGGUGU-3′ (Stealth siRNA; Invitrogen, Paisley, United Kingdom). The 2 human fer siRNAs were either a mix of the following 3 duplexes 5′-GGUGAAGUAUAUAAGGGCACAUUAA-3′, 5′-GCCCUAAGUUCAGUGAACUUCAGAA-3′, and 5′-ACGUAUCCAAGUCUUGGCUACUUAU-3′ (Stealth siRNA) or a mix of 4 siRNAs, SMARTpool M-003129-01-0005 (Dharmacon Research, Lafayette, CO). The human c-kit siRNA was as published39  (Qiagen). The mouse fes I siRNA targeted the sequence 5′-UGCGGCAGCAUGCAGAAGAUCUGAA-3′ (Stealth siRNA). The mouse fes II siRNA targeted sequence was 5′-GCAGAGAUAACAAGCCAGACCGAGA-3′ (Stealth siRNA). The control siRNA was 5′-UUCUCCGAACGUGUCACGU-3′ (Qiagen).

A total of 0.2 to 0.8 nmol siRNA were mixed with 5 to 15 × 106 cells in 0.5 mL RPMI 1640 medium in 4-mm electroporation cuvettes. Electroporation were done at room temperature using a Gene Pulser Electroporator II (Bio-Rad Laboratories, Munich, Germany) at 250 V and 400 μF for TF-1 cells40  and 300 V and 950 μF for P815 cells.

Antibodies

Anti-AKT, anti–phospho-AKT (Ser473), anti-KIT, anti-p38 MAP kinase, anti-mTOR, anti–phospho-mTOR (Ser2448), anti-p70 S6 kinase, anti–phospho-p70 S6 kinase (Thr389), anti–phospho-STAT1 (Tyr701), anti–phospho-STAT3 (Tyr705), and anti–phospho-STAT5 (Tyr694) rabbit polyclonal antibodies were from Cell Signaling Technology (Beverly, MA). Anti-ERK2, anti-KIT C-19, anti-STAT1, anti-STAT3, and anti-STAT5 rabbit polyclonal antibodies and anti-FES N-19 goat polyclonal antibody were from Santa Cruz Biotechnology (Heidelberg, Germany). Anti–active pan-ERK (Thr202/Tyr204) and anti–active p38 (Thr180/Tyr182) rabbit polyclonal antibodies were from Promega (Madison, WI). The other antibodies used were antiphosphotyrosine 4G10 (Upstate Biotechnology, Lake Placid, NY), anti-FES F113 rat monoclonal antibody (Calbiochem, Darmstadt, Germany), anti-FER rabbit polyclonal antibody (Millipore, Bedford, MA), anti–rat RARa/IgM rabbit polyclonal antiserum (Nordic Immunology, Tilburg, the Netherlands), mouse IgG2b (Transduction Laboratories, Lexington, KY), and rat IgM (Zymed, South San Francisco, CA).

Immunoprecipitation and Western blotting

Cells washed in phosphate-buffered saline (PBS) were lysed in HNTG buffer (50 mM HEPES [pH 7], 150 mM NaCl, 1% Triton X-100, 10% glycerol, 1.5 mM MgCl2, and 1 mM EGTA) containing protease inhibitor mixture (Roche Applied Science, Mannheim, Germany), 50 mM NaF, and 100 μM Na3VO4. Protein concentration was measured using the Bio-Rad Protein Assay Kit (Bio-Rad Laboratories). For immunoprecipitation, the samples were incubated with antibody and a bed volume of 10 μL protein A- or protein G-sepharose (Amersham Biosciences, Uppsala, Sweden), washed 3 times with HNTG buffer, and eluted in Laemmli buffer at 100°C for 5 minutes. Proteins were separated on SDS-polyacrylamide gels and transferred to polyvinylidene fluoride membrane (Immobilon-P; Millipore). Membranes were incubated with antibodies and treated using Supersignal West pico chemiluminescent substrate (Pierce, Rockford, IL).

Immunoprecipitations of human FES were done with a mix of F113 antibody and anti-RARa/IgM. Phosphotyrosine immunoprecipitations were done with the 4G10 antibody covalently linked to agarose beads (Upstate Biotechnology).

Cell proliferation assay

A total of 104 cells/well were seeded into 96-well plates in 100 μL of RPMI 1640 medium with 10% fetal bovine serum with or without 5 ng/mL GM-CSF or 250 ng/mL human SCF (Abcys). Cells were incubated for 24 hours at 37°C and pulsed for 6 hours with 0.0185 MBq (0.5 μCi) of [methyl3H]-thymidine (Amersham Biosciences). Cells were then transferred onto glass fiber filters (Packard Instruments, Groningen, the Netherlands), and incorporation was measured using a Rackbeta Compact 1212-411 β-counter (LKB, Uppsala, Sweden).

For carboxyfluorescein succinimidyl ester (CSFE) labeling, 5 × 106 cells were washed with PBS and labeled with 5 μM CFSE (Molecular Probes, Leiden, the Netherlands) for 10 minutes at 37°C. Cells were then washed 2 times with culture medium. Fluorescence of cells was analyzed by flow cytometry. A total of 2 × 104 events was collected on a FACScan (Becton Dickinson, San Jose, CA) and analyzed using FlowJo software (Ashland, OR).

BrdU incorporation assay

106 cells were incubated for 15 minutes at 37°C with 1 mM BrdU solution. BrdU and 7-amino-actinomycin D (7-AAD) staining was performed according to the BrdU Flow kit manual (Becton Dickinson, San Diego, CA). A total of 4 × 104 events was collected on a FACScan and the cellular DNA content was analyzed by FlowJo software.

Identification of novel effectors of KIT signaling

To identify new intracellular proteins in the oncogenic pathway of the KIT receptor, 2 strategies were undertaken. A yeast 2-hybrid screen was performed using KITD816V intracellular domain fused to LexA DNA-binding domain as bait and a cDNA library from EML-c1 cells. One of the isolated clones interacted with KITD816V but not with the control baits LexA-Lamin and KITW42, a loss-of-function mutant of KIT. This clone corresponded to FES tyrosine kinase cDNA. In parallel, mass spectrometry was used to identify tyrosine-phosphorylated proteins in cells that express KITD816V, and FES was again identified by this method.

FES is phosphorylated downstream of human KITD816V and murine KITD814Y

Previous published work has correlated the phosphorylation status of FES with its activation status.41,43  If FES is a downstream effector of KIT signaling, it is expected to be activated and therefore phosphorylated on tyrosine residues. To analyze the phosphorylation status of FES in cells that express the D816V mutant, we first used the cell-line TF-1 KITD816V and the parental TF-1 cells that express endogenous KIT protein. Endogenous FES protein expression has been reported in this human erythroleukemic cell line.17  The parental cells can be propagated either with GM-CSF or with KIT-ligand SCF, while TF-1 KITD816V cells proliferate in the absence of exogenous cytokines. Phosphorylation of FES downstream of KITD816V was analyzed by immunoprecipitation of FES followed by Western blotting with an antiphosphotyrosine antibody. Whereas no phosphorylation of FES on tyrosine residues was detected in TF-1 cells grown in SCF, FES was phosphorylated in TF-1 KITD816V (Figure 1A). Accordingly, FES was detected following immunoprecipitation of phosphorylated proteins with the antiphosphotyrosine 4G10 antibody in TF-1 KITD816V, but not in the parental cells (Figure 1B). To demonstrate the direct implication of KITD816V in FES activation, KIT expression was reduced using siRNA, and FES phosphorylation was analyzed. As shown Figure 1C, reduced expression of KIT resulted in dramatic decrease of FES phosphorylation.

Figure 1

FES is phosphorylated in cells that harbor KITD816V. Immunoprecipitations (IPs) were carried out on soluble cell lysates (SCLs) of serum-starved cells. SCLs were also directly probed with anti-FES to show equivalent quantity of proteins in all lysates. (A) IPs were done on SCLs of TF-1 cells with anti-FES antibody or with an isotype-matched irrelevant antibody (IP control), followed by Western blotting (WB) with an antiphosphotyrosine antibody (anti-PY) or anti-FES antibody. (B) IPs were done on SCLs from TF-1 cells with anti-PY antibody, followed by WB with anti-FES antibody. (C) Lysates from TF-1 KITD816V cells treated with siRNA were immunoprecipitated using anti-PY antibody, followed by WB with anti-FES antibody. KIT and FES protein expression were also controlled in the cell lysates. (D) IPs on lysates from mouse (P815 and D816VKIT BMMCs) or human (HMC-1) mastocytomas with anti-PY antibody or with an isotype-matched irrelevant antibody, followed by WB with anti-FES antibody. P-FES indicates phosphorylated FES protein. The data presented in each panel are representative of 1 out of at least 3 independent experiments.

Figure 1

FES is phosphorylated in cells that harbor KITD816V. Immunoprecipitations (IPs) were carried out on soluble cell lysates (SCLs) of serum-starved cells. SCLs were also directly probed with anti-FES to show equivalent quantity of proteins in all lysates. (A) IPs were done on SCLs of TF-1 cells with anti-FES antibody or with an isotype-matched irrelevant antibody (IP control), followed by Western blotting (WB) with an antiphosphotyrosine antibody (anti-PY) or anti-FES antibody. (B) IPs were done on SCLs from TF-1 cells with anti-PY antibody, followed by WB with anti-FES antibody. (C) Lysates from TF-1 KITD816V cells treated with siRNA were immunoprecipitated using anti-PY antibody, followed by WB with anti-FES antibody. KIT and FES protein expression were also controlled in the cell lysates. (D) IPs on lysates from mouse (P815 and D816VKIT BMMCs) or human (HMC-1) mastocytomas with anti-PY antibody or with an isotype-matched irrelevant antibody, followed by WB with anti-FES antibody. P-FES indicates phosphorylated FES protein. The data presented in each panel are representative of 1 out of at least 3 independent experiments.

Close modal

We then investigated FES phosphorylation in mastocyte cell lines. HMC-1 is a human mast-cell leukemia cell line with 2 point mutations at codons 560 and 816 in the c-kit gene. P815 and D816VKIT BMMCs are 2 models of murine mastocytes that carry the homologous murine D814Y mutation endogenously and human KITD816V, respectively. All 3 were analyzed for phosphorylation status of FES. Again, FES was present in the phosphotyrosine immunoprecipitates in all 3 mastocyte cell lines (Figure 1D). These results show constitutive phosphorylation of FES downstream of KITD816V/D814Y mutants.

FES but not FER mediates proliferation of TF-1 KITD816V and P815 cells

Our data suggested that FES tyrosine kinase is activated downstream of KITD816V and not downstream of the wild-type KIT receptor. To determine if FES is implicated in KITD816V-mediated cell growth, we used RNA interference to abolish FES protein expression. As controls in our experiments, we also used siRNAs directed against c-kit and against the fes-related gene fer. Figure 2 illustrates the reduction of expression of FES, FER, and KIT achieved in representative experiments following transfection of the cell lines with siRNAs. All siRNA experiments described were systematically controlled by Western blotting.

Figure 2

Specific reduction of FES, FER, and KIT proteins in cultured cells. FES, FER, and KIT protein expression in TF-1 KITD816V cells (A), TF-1 cells (B), and P815 cells (C) treated with the corresponding specific siRNAs were analyzed by Western blot. SCLs were prepared 48 hours following transfection, and 30 μg of proteins were loaded per lane. As loading control, ERK2 expression was assessed. fes A and fes B are 2 independent human fes siRNAs. fes I and fes II are 2 independent mouse fes siRNAs (“siRNA transfection”). (B) — indicates lysate of cells not treated with siRNA. (C) Arrow shows FES protein. *Nonspecific band in P815 cells.

Figure 2

Specific reduction of FES, FER, and KIT proteins in cultured cells. FES, FER, and KIT protein expression in TF-1 KITD816V cells (A), TF-1 cells (B), and P815 cells (C) treated with the corresponding specific siRNAs were analyzed by Western blot. SCLs were prepared 48 hours following transfection, and 30 μg of proteins were loaded per lane. As loading control, ERK2 expression was assessed. fes A and fes B are 2 independent human fes siRNAs. fes I and fes II are 2 independent mouse fes siRNAs (“siRNA transfection”). (B) — indicates lysate of cells not treated with siRNA. (C) Arrow shows FES protein. *Nonspecific band in P815 cells.

Close modal

To study the biological function of FES downstream of KIT, cell proliferation of TF-1 KITD816V cells was first assayed by [methyl3H]-thymidine incorporation. These cells strictly depend on KIT for cell proliferation, as shown by the cell sample treated with c-kit siRNA (Figure 3A). Silencing of the fes gene in TF-1 KITD816V resulted in a decreased cell proliferation of 35% when compared with control siRNA. A similar reduction was also observed using another independent fes siRNA (data not shown). Furthermore, addition of GM-CSF to the culture medium, which complements the requirement for KIT signaling (Figure 3A; c-kit siRNA + GM-CSF), rescued the cell-proliferation defect associated to FES inactivation (Figure 3A; fes siRNA), indicating specificity of FES in KITD816V-mediated cell proliferation. In these experiments, inactivation of fer did not affect TF-1 KITD816V cell proliferation (Figure 3B).

Figure 3

FES is required for KITD816V-induced cell proliferation. Following siRNA treatment, cells were seeded in 96-well plates and maintained with or without cytokine as indicated. Thymidine incorporation assays were done 24 hours later. Cells were TF-1 KITD816V (A,B), TF-1 (C), and P815 (D). Cell proliferation is represented as a percentage of control siRNA. The data are from 3 independent experiments done in triplicate. Error bars indicate the standard error of the mean.

Figure 3

FES is required for KITD816V-induced cell proliferation. Following siRNA treatment, cells were seeded in 96-well plates and maintained with or without cytokine as indicated. Thymidine incorporation assays were done 24 hours later. Cells were TF-1 KITD816V (A,B), TF-1 (C), and P815 (D). Cell proliferation is represented as a percentage of control siRNA. The data are from 3 independent experiments done in triplicate. Error bars indicate the standard error of the mean.

Close modal

We also investigated the proliferation of the parental TF-1 cells in response to SCF (Figure 3C). These cells do not proliferate in the absence of exogenous cytokines (Figure 3C; black histograms). In the presence of SCF, c-kit siRNA, but not fes siRNA, reduced the proliferation of TF-1 cells, indicating that FES is not required for KITWT-mediated cell proliferation.

Finally, we addressed whether FES is required for cell proliferation in mastocytomas using the P815 cell line, which expresses KITD814Y mutant endogenously (Figure 3D). Proliferation and survival of P815 cells is strictly dependent on KIT, as the use of either the KIT inhibitor dasatinib (BMS-354825) or the reduction of KIT expression using siRNA resulted in cell death within 24 hours (data not shown). We abolished FES expression using 2 independent murine siRNAs. As observed in TF-1 KITD816V, fes silencing resulted in decreased proliferation of P815 cells by 50% (Figure 3D). These results indicate that FES is required for cellular proliferation downstream of the kinase domain mutant KITD816V/KITD814Y.

FES facilitates G1 to S phase transition

We next investigated whether FES is required for cell survival or for cell-cycle progression. Annexin V and 7-AAD staining did not show any evidence for cell death in the absence of FES (data not shown). To delineate the role of FES in cellular proliferation, we stained the cells with CFSE and evaluated the number of cell divisions at various time points following staining. CFSE irreversibly couples to cellular proteins; when cells divide, the fluorescence is then equally distributed in the daughter cells. Cells transfected either with fes siRNAs or control siRNA presented identical profiles following staining (Figure 4A). The staining intensity was then used to evaluate the number of cell divisions in the 2 transfected populations. On day 2, the profile of the control siRNA-treated population was homogenous (Figure 4B,C), with most cells having done 5 divisions. In comparison, the FES-depleted population presented a wider peak with stronger labeling intensity, reflecting the fact that many cells had gone through 2, 3, or 4 cell divisions only (Figure 4B,C). Therefore, the decreased [methyl3H]-thymidine incorporation in the absence of FES is due to reduced rate of cell division rather than to cell death. We concluded that fes gene silencing resulted in slower proliferation of the cells.

Figure 4

Depletion of FES disturbs cell cycle. P815 cells transfected with either control or fes siRNAs were labeled with CFSE. Histogram plots represent the profiles for the 2 populations, fes I siRNA (gray line) and control siRNA (black line) on day 0 (A) and 2 days following labeling (B,C). (C) The estimated number of cell divisions is indicated above the histogram. The analyses were done with FlowJo software. These profiles represent 1 of 3 independent experiments that gave similar results.

Figure 4

Depletion of FES disturbs cell cycle. P815 cells transfected with either control or fes siRNAs were labeled with CFSE. Histogram plots represent the profiles for the 2 populations, fes I siRNA (gray line) and control siRNA (black line) on day 0 (A) and 2 days following labeling (B,C). (C) The estimated number of cell divisions is indicated above the histogram. The analyses were done with FlowJo software. These profiles represent 1 of 3 independent experiments that gave similar results.

Close modal

In parallel to these experiments, we analyzed the cell cycle using BrdU incorporation and DNA staining with 7-AAD. BrdU staining of cells was done 48 hours following electroporation of siRNAs. Whereas in our experimental conditions, most cells transfected with control siRNA entered in S phase (52% in S phase and 38% in G0/G1 phase), fes siRNA-treated cells were mainly in G0/G1 phase (59% versus 33% in S phase; Figure 5). Thus, reduced expression of FES resulted in accumulation of cells in G0/G1, suggesting that FES facilitates the transition between G1 and S phase. Altogether, these results demonstrate a function of FES on cellular proliferation downstream of the KIT kinase mutant.

Figure 5

Absence of FES slows down transition from G1 to S phase. P815 cells transfected with either control or fes siRNAs were stained with anti-BrdU-FITC antibody and 7-AAD. The percentage of cells in G0/G1, S, or G2/M phases were quantified using FlowJo software. The values are from 1 of 3 independent experiments that gave similar results.

Figure 5

Absence of FES slows down transition from G1 to S phase. P815 cells transfected with either control or fes siRNAs were stained with anti-BrdU-FITC antibody and 7-AAD. The percentage of cells in G0/G1, S, or G2/M phases were quantified using FlowJo software. The values are from 1 of 3 independent experiments that gave similar results.

Close modal

FES acts negatively on STATs and positively on p70 S6 kinase activation pathway

To investigate FES contribution to KITD816V downstream signaling, the activation status of several mediators of KIT signaling was analyzed. Cell lysates from control and fes siRNA TF-1 KITD816V cells were probed with phosphospecific antibodies. First, we analyzed the activation of the MAP kinases p38 (Figure 6) and ERK1/2 (data not shown). Their activation was not affected by down-regulation of FES expression. Subsequently, we looked at tyrosine phosphorylation of STATs. We found constitutive tyrosine phosphorylation of STAT-1, STAT-3, and STAT-5 in the control cells as previously described for KITD816V. The depletion of FES led to increased phosphorylation of all 3 STAT proteins (Figure 6). We also analyzed downstream effectors of the PI3K cascade. AKT (data not shown) and mTOR (Figure 6) phosphorylation were not modified following FES inactivation. By contrast, phosphorylation of Thr389, which is required for activation of p70 S6 kinase,44,46  was decreased in cells lacking FES protein (Figure 6). These results suggest that FES negatively regulates the activation of STATs, but acts positively on p70 S6 kinase activation downstream of the activated KIT mutant.

Figure 6

Effect of fes siRNA on phosphorylation of STAT proteins and p70 S6 kinase. TF-1 KITD816V cells treated with either control or fes siRNAs were serum-starved for 16 hours. SCLs were immunoblotted with indicated anti-phospho antibodies and after stripping, reprobed with specific antisera. Similar results were obtained in 3 independent experiments.

Figure 6

Effect of fes siRNA on phosphorylation of STAT proteins and p70 S6 kinase. TF-1 KITD816V cells treated with either control or fes siRNAs were serum-starved for 16 hours. SCLs were immunoblotted with indicated anti-phospho antibodies and after stripping, reprobed with specific antisera. Similar results were obtained in 3 independent experiments.

Close modal

FES, FER, and cell-surface receptors

As an intracellular tyrosine kinase, FES is a downstream effector of signals transduced from cell-surface receptors, such as IL-3, GM-CSF, IL-4 receptors, and gp130-containing receptors.17,,20,47  In primary erythroid cells, the activation of FES and/or FER downstream of wild-type KIT receptor has been suggested using a dual-specific antibody.25  Another study showed increased FER phosphorylation in response to SCF in BMMCs.48  The function of FER in KIT-mediated adhesion and chemotaxis was suggested through the use of a dominant-negative mutant protein.48  The function of these proteins in the context of gain-of-function KIT mutants has not been explored previously. In our models, RNA interference was used to significantly reduce gene expression of FES and FER. We show that FES was dispensable for cell proliferation mediated by KITWT, but is required for the KITD816V proliferation signal. This observation highlights differential signaling mechanisms between wild-type and mutant KIT receptors. In mouse models that harbor kinase-inactivating mutations in both genes, the redundant function of FES and FER has been suggested.16,28  Interestingly, our results indicate that FER does not compensate or participate in KITD816V-mediated proliferation.

FES regulates STAT proteins and p70 S6 kinase

In TF-1 KITD816V cells, FES regulates p70 S6 kinase and STAT-1, STAT-3, and STAT-5 activation, but not ERK1/2, p38, AKT, or mTOR. Interestingly, a similar function of FES upstream of p70 S6 kinase without affecting AKT phosphorylation has already been established in IL-4 signaling in the B lymphoma cell line M12.4.1.20  Several lines of evidence have documented a role for FES in regulating STAT tyrosine phosphorylation. Ectopic expression of FES increased tyrosine phosphorylation of STAT-3 and STAT-5,21  while monocytes from transgenic mice with a kinase-inactive FES show reduced STAT-3 and STAT-5 phosphorylation upon GM-CSF stimulation.22  However, consistent with our observations using RNA interference, in FES knock-out mice, STAT-3 and STAT-5 tyrosine phosphorylation is increased downstream of IL-6 and GM-CSF.23  A working model to reconcile all these observations is that STAT proteins are sequestered and phosphorylated by FES.23  Therefore, our observations and conclusions regarding FES function in KITD816V signaling are consistent with hypotheses raised in other systems. Whether the aberrant STAT tyrosine phosphorylation and/or the impaired activation of p70 S6 kinase are responsible for the proliferative defects remains to be elucidated. A function for p70 S6 kinase in cell growth is consistent with a number of previous studies on this kinase.49,51 

Signaling downstream of KITD816V

KIT gain-of-function mutations activate a large spectrum of proteins. The determination of which proteins are actually required for the oncogenic properties of KIT mutants is an important challenge. Some studies have addressed the requirement for some of these proteins for KIT function. Thus, PI3K is essential for survival and subsequently for KITD816V- or KITD814Y-mediated proliferation.52  In Mo7e cells, STAT-3 but not STAT-1 seems to be required for cell proliferation driven by KITD816V, as suggested by the use of dominant-negative mutants of these proteins.53  Using chemical inhibitors, it has been suggested that NF-κB, mTOR, and PKCδ are important effectors of KITD816V.54,56  These hypotheses have not yet been challenged using gene inactivation techniques. Using antisense oligonucleotides and RNA interference, another study very recently implicated the BCL-2-related protein MCL-1 in KITD816V-dependent cell proliferation.57  We have shown here that FES is a component of KITD816V downstream signaling. Furthermore, using several independent siRNAs, we demonstrated that FES is required for cell proliferation downstream of KITD816V. However, fes siRNA reduced (35% and 50% reduction of TF-1 KITD816V and P815 cells respectively) but did not abolish cell proliferation (Figure 3A,D). Therefore, FES is 1 of several components required for cell proliferation. Mutations at codon 816 are the most frequent mutations of KIT. Yet, other KIT gain-of-function mutations occur in about 70% of GISTs, mainly affecting the regulatory juxtamembrane region. It remains to be determined whether FES is also necessary for the function of juxtamembrane mutants of KIT. Moreover, because of structural and signaling similarities among receptors, our study raises the question of the implication of FES downstream of other constitutively activated tyrosine kinase receptors.

Inhibitors of KITD816V

Several recent studies have identified inhibitors that reduce the growth or the survival mediated by KITD816V in cell cultures. These include drugs that target KIT itself such as dasatinib,58  AMN107,59,60  PP1 and PP2,61  semaxinib (SU5416),62  and PKC412,60,63,64  and drugs that target NF-κB,54  mTOR,56  protein kinase C δ (PKCδ),55  PI3K,52  and heat shock protein 90 (HSP90).65  The present study points to FES as a putative target of KITD816V-mediated cell proliferation. In contrast to NF-κB, mTOR, and PI3K, FES has limited nonredundant functions in normal physiology. Since FES is a kinase, selective ATP competitive inhibitors can be developed. The use of inhibitors for signaling components downstream of an oncoprotein are being considered to target the PI3K pathway.66,67  Considering that KIT kinase mutants show resistance to clinically available KIT inhibitors, and that secondary resistance occur in some patients, this strategy could be used either as an alternative to, or in combination with, the KIT inhibitors.

In conclusion, this study highlights the function of FES in cell proliferation downstream of the kinase-mutant KIT receptor. Since FES is dispensable for wild-type KIT signaling, it is an attractive putative therapeutic target in KIT-related pathologies.

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 USC section 1734.

We thank Jean-Rémy Galindo and Sandrine Sarrazin for help with FACS analysis, J. H. Butterfield for the HMC-1 cell line, and Sheela Ramanathan for critical reading of the manuscript.

This work was supported by INSERM, La Ligue Contre le Cancer “équipe labellisée” and a grant from le Ministère de la Recherche (ACI [Action Concertée Incitative] Molécules et Cibles Thérapeutiques). E.V. was supported by a PhD fellowship from INSERM-Région PACA [Provence-Alpes-Cote d'Azur], and S.L. by a postdoctoral fellowship from la Fondation de France. P.D. and P.D.S. are INSERM scientists.

Contribution: E.V. performed the experiments, analyzed and interpreted the data, and drafted the manuscript. S.L. contributed vital reagents. P.D. interpreted the data, approved the data, and provided logistic and financial support. P.D.S. contributed by designing and supervising the study, writing the manuscript, and approving the data.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Paulo De Sepulveda, INSERM UMR599, 27 Blvd Leï Roure, 13009 Marseille, France; e-mail:sepulveda@marseille.inserm.fr.

1
Hirota
S
Isozaki
K
Moriyama
Y
et al
Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors.
Science
1998
279
577
580
2
Longley
BJ
Jr
Metcalfe
DD
Tharp
M
et al
Activating and dominant inactivating c-KIT catalytic domain mutations in distinct clinical forms of human mastocytosis.
Proc Natl Acad Sci U S A
1999
96
1609
1614
3
Beghini
A
Ripamonti
CB
Cairoli
R
et al
KIT activating mutations: incidence in adult and pediatric acute myeloid leukemia, and identification of an internal tandem duplication.
Haematologica
2004
89
920
925
4
Hongyo
T
Hoshida
Y
Nakatsuka
S
et al
p53, K-ras, c-kit and beta-catenin gene mutations in sinonasal NK/T-cell lymphoma in Korea and Japan.
Oncol Rep
2005
13
265
271
5
Cairoli
R
Beghini
A
Grillo
G
et al
Prognostic impact of c-KIT mutations in core binding factor leukemias: an Italian retrospective study.
Blood
2006
107
3463
3468
6
Tian
Q
Frierson
HF
Jr
Krystal
GW
Moskaluk
CA
Activating c-kit gene mutations in human germ cell tumors.
Am J Pathol
1999
154
1643
1647
7
Looijenga
LH
de Leeuw
H
van Oorschot
M
et al
Stem cell factor receptor (c-KIT) codon 816 mutations predict development of bilateral testicular germ-cell tumors.
Cancer Res
2003
63
7674
7678
8
Kemmer
K
Corless
CL
Fletcher
JA
et al
KIT mutations are common in testicular seminomas.
Am J Pathol
2004
164
305
313
9
Curtin
JA
Busam
K
Pinkel
D
Bastian
BC
Somatic activation of KIT in distinct subtypes of melanoma.
J Clin Oncol
2006
24
4340
4346
10
Valent
P
Akin
C
Sperr
WR
et al
Mastocytosis: pathology, genetics, and current options for therapy.
Leuk Lymphoma
2005
46
35
48
11
Ma
Y
Zeng
S
Metcalfe
DD
et al
The c-KIT mutation causing human mastocytosis is resistant to STI571 and other KIT kinase inhibitors; kinases with enzymatic site mutations show different inhibitor sensitivity profiles than wild-type kinases and those with regulatory-type mutations.
Blood
2002
99
1741
1744
12
Akin
C
Brockow
K
D'Ambrosio
C
et al
Effects of tyrosine kinase inhibitor STI571 on human mast cells bearing wild-type or mutated c-kit.
Exp Hematol
2003
31
686
692
13
Zermati
Y
De Sepulveda
P
Feger
F
et al
Effect of tyrosine kinase inhibitor STI571 on the kinase activity of wild-type and various mutated c-kit receptors found in mast cell neoplasms.
Oncogene
2003
22
660
664
14
Ronnstrand
L
Signal transduction via the stem cell factor receptor/c-Kit.
Cell Mol Life Sci
2004
61
2535
2548
15
Casteran
N
De Sepulveda
P
Beslu
N
et al
Signal transduction by several KIT juxtamembrane domain mutations.
Oncogene
2003
22
4710
4722
16
Greer
P
Closing in on the biological functions of Fps/Fes and Fer.
Nat Rev Mol Cell Biol
2002
3
278
289
17
Hanazono
Y
Chiba
S
Sasaki
K
et al
c-fps/fes protein-tyrosine kinase is implicated in a signaling pathway triggered by granulocyte-macrophage colony-stimulating factor and interleukin-3.
EMBO J
1993
12
1641
1646
18
Izuhara
K
Feldman
RA
Greer
P
Harada
N
Interaction of the c-fes proto-oncogene product with the interleukin-4 receptor.
J Biol Chem
1994
269
18623
18629
19
Matsuda
T
Fukada
T
Takahashi-Tezuka
M
et al
Activation of Fes tyrosine kinase by gp130, an interleukin-6 family cytokine signal transducer, and their association.
J Biol Chem
1995
270
11037
11039
20
Jiang
H
Foltenyi
K
Kashiwada
M
et al
Fes mediates the IL-4 activation of insulin receptor substrate-2 and cellular proliferation.
J Immunol
2001
166
2627
2634
21
Nelson
KL
Rogers
JA
Bowman
TL
Jove
R
Smithgall
TE
Activation of STAT3 by the c-Fes protein-tyrosine kinase.
J Biol Chem
1998
273
7072
7077
22
Senis
Y
Zirngibl
R
McVeigh
J
Haman
A
Hoang
T
Greer
PA
Targeted disruption of the murine fps/fes proto-oncogene reveals that Fps/Fes kinase activity is dispensable for hematopoiesis.
Mol Cell Biol
1999
19
7436
7446
23
Hackenmiller
R
Kim
J
Feldman
RA
Simon
MC
Abnormal Stat activation, hematopoietic homeostasis, and innate immunity in c-fes-/- mice.
Immunity
2000
13
397
407
24
Rovida
E
Marra
F
Baccarini
M
Dello Sbarba
P
Constitutive activation of the MAPK pathway mediates v-fes-induced mitogenesis in murine macrophages.
Blood
2000
95
3959
3963
25
Sangrar
W
Gao
Y
Bates
B
Zirngibl
R
Greer
PA
Activated Fps/Fes tyrosine kinase regulates erythroid differentiation and survival.
Exp Hematol
2004
32
935
945
26
Izuhara
K
Feldman
RA
Greer
P
Harada
N
Interleukin-4 induces association of the c-fes proto-oncogene product with phosphatidylinositol-3 kinase.
Blood
1996
88
3910
3918
27
Zirngibl
RA
Senis
Y
Greer
PA
Enhanced endotoxin sensitivity in fps/fes-null mice with minimal defects in hematopoietic homeostasis.
Mol Cell Biol
2002
22
2472
2486
28
Senis
YA
Craig
AW
Greer
PA
Fps/Fes and Fer protein-tyrosinekinases play redundant roles in regulating hematopoiesis.
Exp Hematol
2003
31
673
681
29
Hackenmiller
R
Simon
MC
Truncation of c-fes via gene targeting results in embryonic lethality and hyperproliferation of hematopoietic cells.
Dev Biol
2002
245
255
269
30
Yee
SP
Mock
D
Greer
P
et al
Lymphoid and mesenchymal tumors in transgenic mice expressing the v-fps protein-tyrosine kinase.
Mol Cell Biol
1989
9
5491
5499
31
Sangrar
W
Gao
Y
Zirngibl
RA
Scott
ML
Greer
PA
The fps/fes proto-oncogene regulates hematopoietic lineage output.
Exp Hematol
2003
31
1259
1267
32
Parsons
SA
Greer
PA
The Fps/Fes kinase regulates the inflammatory response to endotoxin through down-regulation of TLR4, NF-kappaB activation, and TNF-alpha secretion in macrophages.
J Leukoc Biol
2006
80
1522
1528
33
Bardelli
A
Parsons
DW
Silliman
N
et al
Mutational analysis of the tyrosine kinome in colorectal cancers.
Science
2003
300
949
34
Sangrar
W
Zirgnibl
RA
Gao
Y
Muller
WJ
Jia
Z
Greer
PA
An identity crisis for fps/fes: oncogene or tumor suppressor?
Cancer Res
2005
65
3518
3522
35
Delfino
FJ
Stevenson
H
Smithgall
TE
A growth-suppressive function for the c-fes protein-tyrosine kinase in colorectal cancer.
J Biol Chem
2006
281
8829
8835
36
De Sepulveda
P
Okkenhaug
K
Rose
JL
Hawley
RG
Dubreuil
P
Rottapel
R
Socs1 binds to multiple signalling proteins and suppresses steel factor-dependent proliferation.
EMBO J
1999
18
904
915
37
Bayle
J
Letard
S
Frank
R
Dubreuil
P
De Sepulveda
P
Suppressor of cytokine signaling 6 associates with KIT and regulates KIT receptor signaling.
J Biol Chem
2004
279
12249
12259
38
Zappulla
JP
Dubreuil
P
Desbois
S
et al
Mastocytosis in mice expressing human Kit receptor with the activating Asp816Val mutation.
J Exp Med
2005
202
1635
1641
39
Catalano
A
Rodilossi
S
Rippo
MR
Caprari
P
Procopio
A
Induction of stem cell factor/c-Kit/slug signal transduction in multidrug-resistant malignant mesothelioma cells.
J Biol Chem
2004
279
46706
46714
40
Wen
L
Zhuang
L
Luo
X
Wei
P
TL1A-induced NF-kappaB activation and c-IAP2 production prevent DR3-mediated apoptosis in TF-1 cells.
J Biol Chem
2003
278
39251
39258
41
Hjermstad
SJ
Peters
KL
Briggs
SD
Glazer
RI
Smithgall
TE
Regulation of the human c-fes protein tyrosine kinase (p93c-fes) by its src homology 2 domain and major autophosphorylation site (Tyr-713).
Oncogene
1993
8
2283
2292
42
Rogers
JA
Read
RD
Li
J
Peters
KL
Smithgall
TE
Autophosphorylation of the Fes tyrosine kinase. Evidence for an intermolecular mechanism involving two kinase domain tyrosine residues.
J Biol Chem
1996
271
17519
17525
43
Takashima
Y
Delfino
FJ
Engen
JR
Superti-Furga
G
Smithgall
TE
Regulation of c-Fes tyrosine kinase activity by coiled-coil and SH2 domains: analysis with Saccharomyces cerevisiae.
Biochemistry
2003
42
3567
3574
44
Pearson
RB
Dennis
PB
Han
JW
et al
The principal target of rapamycin-induced p70s6k inactivation is a novel phosphorylation site within a conserved hydrophobic domain.
EMBO J
1995
14
5279
5287
45
Alessi
DR
Kozlowski
MT
Weng
QP
Morrice
N
Avruch
J
3-Phosphoinositide-dependent protein kinase 1 (PDK1) phosphorylates and activates the p70 S6 kinase in vivo and in vitro.
Curr Biol
1998
8
69
81
46
Weng
QP
Kozlowski
M
Belham
C
Zhang
A
Comb
MJ
Avruch
J
Regulation of the p70 S6 kinase by phosphorylation in vivo. Analysis using site-specific anti-phosphopeptide antibodies.
J Biol Chem
1998
273
16621
16629
47
Brizzi
MF
Aronica
MG
Rosso
A
Bagnara
GP
Yarden
Y
Pegoraro
L
Granulocyte-macrophage colony-stimulating factor stimulates JAK2 signaling pathway and rapidly activates p93fes, STAT1 p91, and STAT3 p92 in polymorphonuclear leukocytes.
J Biol Chem
1996
271
3562
3567
48
Craig
AW
Greer
PA
Fer kinase is required for sustained p38 kinase activation and maximal chemotaxis of activated mast cells.
Mol Cell Biol
2002
22
6363
6374
49
Lane
HA
Fernandez
A
Lamb
NJ
Thomas
G
p70s6k function is essential for G1 progression.
Nature
1993
363
170
172
50
Fingar
DC
Richardson
CJ
Tee
AR
Cheatham
L
Tsou
C
Blenis
J
mTOR controls cell cycle progression through its cell growth effectors S6K1 and 4E-BP1/eukaryotic translation initiation factor 4E.
Mol Cell Biol
2004
24
200
216
51
Bessard
A
Coutant
A
Rescan
C
et al
An MLCK-dependent window in late G1 controls S phase entry of proliferating rodent hepatocytes via ERK-p70S6K pathway.
Hepatology
2006
44
152
163
52
Chian
R
Young
S
Danilkovitch-Miagkova
A
et al
Phosphatidylinositol 3 kinase contributes to the transformation of hematopoietic cells by the D816V c-Kit mutant.
Blood
2001
98
1365
1373
53
Ning
ZQ
Li
J
Arceci
RJ
Signal transducer and activator of transcription 3 activation is required for Asp(816) mutant c-Kit-mediated cytokine-independent survival and proliferation in human leukemia cells.
Blood
2001
97
3559
3567
54
Tanaka
A
Konno
M
Muto
S
et al
A novel NF-kappaB inhibitor, IMD-0354, suppresses neoplastic proliferation of human mast cells with constitutively activated c-kit receptors.
Blood
2005
105
2324
2331
55
Jelacic
T
Linnekin
D
PKCdelta plays opposite roles in growth mediated by wild-type Kit and an oncogenic Kit mutant.
Blood
2005
105
1923
1929
56
Gabillot-Carre
M
Lepelletier
Y
Humbert
M
et al
Rapamycin inhibits growth and survival of D816V-mutated c-kit mast cells.
Blood
2006
108
1065
1072
57
Aichberger
KJ
Mayerhofer
M
Gleixner
KV
et al
Identification of MCL1 as a novel target in neoplastic mast cells in systemic mastocytosis: inhibition of mast cell survival by MCL1 antisense oligonucleotides and synergism with PKC412.
Blood
2007
109
3031
3041
58
Schittenhelm
MM
Shiraga
S
Schroeder
A
et al
Dasatinib (BMS-354825), a dual SRC/ABL kinase inhibitor, inhibits the kinase activity of wild-type, juxtamembrane, and activation loop mutant KIT isoforms associated with human malignancies.
Cancer Res
2006
66
473
481
59
von Bubnoff
N
Gorantla
SH
Kancha
RK
Lordick
F
Peschel
C
Duyster
J
The systemic mastocytosis-specific activating cKit mutation D816V can be inhibited by the tyrosine kinase inhibitor AMN107.
Leukemia
2005
19
1670
1671
60
Gleixner
KV
Mayerhofer
M
Aichberger
KJ
et al
PKC412 inhibits in vitro growth of neoplastic human mast cells expressing the D816V-mutated variant of KIT: comparison with AMN107, imatinib, and cladribine (2CdA) and evaluation of cooperative drug effects.
Blood
2006
107
752
759
61
Tatton
L
Morley
GM
Chopra
R
Khwaja
A
The Src-selective kinase inhibitor PP1 also inhibits Kit and Bcr-Abl tyrosine kinases.
J Biol Chem
2003
278
4847
4853
62
Kosmider
O
Denis
N
Dubreuil
P
Moreau-Gachelin
F
Semaxinib (SU5416) as a therapeutic agent targeting oncogenic Kit mutants resistant to imatinib mesylate.
Oncogene
2006
26
3904
3908
63
Gotlib
J
Berube
C
Growney
JD
et al
Activity of the tyrosine kinase inhibitor PKC412 in a patient with mast cell leukemia with the D816V KIT mutation.
Blood
2005
106
2865
2870
64
Growney
JD
Clark
JJ
Adelsperger
J
et al
Activation mutations of human c-KIT resistant to imatinib mesylate are sensitive to the tyrosine kinase inhibitor PKC412.
Blood
2005
106
721
724
65
Fumo
G
Akin
C
Metcalfe
DD
Neckers
L
17-Allylamino-17-demethoxygeldanamycin (17-AAG) is effective in down-regulating mutated, constitutively activated KIT protein in human mast cells.
Blood
2004
103
1078
1084
66
Recher
C
Dos Santos
C
Demur
C
Payrastre
B
mTOR, a new therapeutic target in acute myeloid leukemia.
Cell Cycle
2005
4
1540
1549
67
Granville
CA
Memmott
RM
Gills
JJ
Dennis
PA
Handicapping the race to develop inhibitors of the phosphoinositide 3-kinase/Akt/mammalian target of rapamycin pathway.
Clin Cancer Res
2006
12
679
689
Sign in via your Institution