Vascular endothelial growth factor (VEGF) is one of the most important mediators of tumor angiogenesis. In addition to hypoxia, peptide growth factors are known to regulate VEGF expression but the effect of stem cell factor (SCF), the ligand for c-Kit, on VEGF expression has not been characterized. We therefore studied the effect of SCF-mediated c-Kit activation on VEGF expression by the H526 small cell lung cancer (SCLC) cell line. SCF treatment doubled VEGF mRNA expression and VEGF secretion in the absence of other exogenous growth factors, an effect efficiently blocked by imatinib. The increase in VEGF mRNA occurred within the first 2 hours of treatment and was not caused by alterations in mRNA stability. The phosphatidylinositol 3-kinase inhibitor LY294002 blocked the increase in VEGF mRNA, implicating c-Kit-mediated activation of phosphatidylinositol 3-kinase in the phenomenon. VEGF promoter-reporter transfections indicated that a SCF-mediated increase in VEGF promoter activity paralleled the increase in VEGF mRNA, documenting that SCF mediated its effects through enhanced VEGF transcription. Mutation of the core hypoxia-inducible factor (HIF)-1 binding element in the VEGF promoter significantly blunted SCF-responsiveness. SCF increased nuclear levels of the HIF-1α transcription factor, which correlated well with increased HIF-1α binding to a consensus hypoxia-responsive element. SCF-mediated effects on HIF-1α expression were additive with those produced by CoCl2, a hypoxia-mimetic agent. These data indicate that activation of c-Kit by SCF leads to a predominantly HIF-1α-mediated enhancement of VEGF expression and that inhibition of c-Kit signaling with imatinib could result in inhibition of tumor angiogenesis. [Mol Cancer Ther 2006;5(6):1415–22]

Angiogenesis is a critical process in tumor progression that not only provides the growing tumor with required oxygen, nutrients, and growth factors but also provides the circulatory access that allows tumor cells to metastasize (1, 2). Tumor angiogenesis is regulated by a variety of both positive and negative regulatory molecules (1, 3). When the net effects of the positive regulatory factors overwhelm the effects of the negative factors, the recruitment of preexisting blood vessels and growth of new vessels into tumors occurs, in a process known as the “angiogenic switch” (3). In experimental models this angiogenic switch is associated with a rapid increase in tumor size, local invasion, and the development of metastatic disease. Among the most important proangiogenic determinants of the angiogenic switch is the level of vascular endothelial growth factor (VEGF) expression by tumor cells. VEGF expression is regulated by two general types of stimuli: oxygen tension and polypeptide growth factors or cytokines, as well as oncogenically activated components of their downstream signaling pathways (1, 4).

Low oxygen tension regulates VEGF expression by enhancing transcription of the VEGF gene and by stabilization of its mRNA. It is now clear that the transcription factor hypoxia-inducible factor (HIF)-1α is the major regulator of VEGF transcription in response to hypoxia (5, 6). In some cells, hypoxia also results in a prolongation of VEGF mRNA half-life (7), which may also be ultimately attributable to the activity of HIF-1α (8). HIF-1α forms a heterodimer with constitutively expressed HIF-1β that binds to a hypoxia-responsive element (9, 10) present in the promoter or enhancer of many hypoxia-inducible genes (5, 6). HIF-1α levels are tightly regulated by oxygen tension through hydroxylation of prolyl residues, which occurs under normoxic conditions. This prolyl hydroxylation is required for interaction with the von Hippel-Lindau tumor suppressor protein, which is the recognition component of an E3 ubiquitin ligase that mediates the destruction of HIF-1α through the proteasomal degradation pathway. Hypoxic conditions retard the prolyl hydroxylation of HIF-1α and the protein is stabilized, enabling it to transactivate hypoxia-responsive genes, including VEGF (6).

Growth factors and cytokines mediate an increase in VEGF mRNA expression by regulatory mechanisms distinct from those involved in hypoxic regulation (1, 5). Whereas stabilization of HIF-1α protein is the primary mechanism by which hypoxia influences VEGF expression, growth factors enhance HIF-1α protein synthesis and transactivation domain function. Growth factors and cytokines such as epidermal growth factor, heregulin, insulin, insulin-like growth factor I, and interleukin-1β induce VEGF expression by enhancing expression of HIF-1α protein and its DNA-binding capability (5). This occurs under normoxic conditions and can result in an additive effect when growth factor stimulation occurs under hypoxic conditions (11, 12). Growth factor receptor tyrosine kinase stimulation seems to enhance HIF-1α mRNA translation via activation of the phosphatidylinositol 3-kinase-Akt-mammalian target of rapamycin signaling pathway and subsequent phosphorylation of translational regulatory proteins p70 S6 kinase and eukaryotic initiation factor 4E binding protein (11, 12). Activation of mitogen-activated protein kinase (MAPK) pathways correlates with phosphorylation of HIF-1α and enhancement of its transactivation function, as well as phosphorylation of eukaryotic initiation factor 4E (11, 13).

Stem cell factor (SCF), the ligand for the c-Kit receptor tyrosine kinase, has been implicated in the regulation of angiogenesis in hematopoietic and nonhematopoietic malignancies but the mechanisms that mediate this effect have not yet been elucidated. It has been shown that rat mammary tumor cells transfected with a SCF expression vector develop increased microvascular density (14). SCF has also been implicated in the mobilization of endothelial progenitor cells required for angiogenesis (15). Treatment of neuroblastoma cell lines and xenografts with imatinib (Gleevec), a specific inhibitor of c-Kit and platelet-derived growth factor receptor, resulted in decreased VEGF expression (16). Finally, we have recently observed that treatment of small cell lung cancer (SCLC) cell xenografts with SU5416, a multitargeted kinase inhibitor that inhibits both c-Kit and the VEGF receptor-2, resulted in decreased tumor microvascular density. Whereas, in vivo, it could not be determined the degree to which the effect on angiogenesis was due to inhibition of tumor cell c-Kit versus VEGF receptor 2 of endothelial precursors, in vitro studies showed that SU5416 directly inhibited VEGF expression by the tumor cells (17). Because at least 70% of SCLC cells coexpress SCF and c-Kit (18) and tumor microvessel density and VEGF expression have been shown to correlate with stage and prognosis in SCLC cell (19, 20), we have explored the regulation of VEGF expression by SCF in SCLC cells. In addition, we have also determined the effect of imatinib, a highly efficient inhibitor of c-Kit signal transduction (21), on VEGF expression.

SCF treatment of multiple c-Kit-expressing SCLC cell lines increased VEGF secretion by ∼2-fold, an effect efficiently inhibited by imatinib. An increase in VEGF mRNA in the H526 cell line occurred within the first 2 hours of SCF treatment and was not caused by alterations in mRNA stability. The phosphatidylinositol 3-kinase inhibitor LY294002 blocked the increase in VEGF mRNA, implicating c-Kit-mediated activation of phosphatidylinositol 3-kinase in the phenomenon. VEGF promoter-reporter transfections indicated that the SCF-mediated increase in VEGF mRNA occurred through a transcriptional mechanism partially dependent on an intact HIF-1 binding site. SCF enhanced nuclear HIF-1α levels, which correlated well with increased HIF-1α binding to a consensus hypoxia-responsive element. SCF-mediated effects on HIF-1α expression were additive with those produced by CoCl2, a hypoxia-mimetic agent. Thus, activation of c-Kit by SCF has a significant effect on VEGF expression, and inhibition of c-Kit signaling with imatinib could have clinically relevant antiangiogenic effects by inhibiting VEGF expression.

Cell Growth

SCLC cell lines (22) were cultured in complete medium consisting of 10% (v/v) fetal bovine serum (Life Technologies, Invitrogen Corporation, Carlsbad, CA), 2 mmol/L l-glutamine (BioWhittaker, Walkersville, MD), and 50 units/mL penicillin-streptomycin (BioWhittaker) in RPMI 1640 (Life Technologies, Invitrogen). Where indicated, when grown in the absence of serum, 0.1% bovine serum albumin (Sigma, St. Louis, MO) and recombinant SCF (100 ng/mL; Peprotech, Rocky Hill, NJ) were added to the medium. To mimic hypoxia, cells were grown in the presence of 100 μmol/L CoCl2 (Sigma). To block c-Kit signaling, cells were grown in media containing imatinib mesylate (Novartis Pharma, Basel, Switzerland), LY294002, or PD98059 (Calbiochem, San Diego, CA) solubilized in DMSO, as indicated. The concentration of DMSO in all cultures, including controls, was 0.1%.

VEGF Assays

Secretion of VEGF by 5 × 105 cells in 1 mL of culture medium was measured using an ELISA assay kit as directed by the supplier (R&D Systems, Minneapolis, MN). For Northern blotting, a 444-bp cDNA probe was generated by reverse transcription-PCR using commercial oligonucleotide primers (R&D Systems), following the directions supplied by the manufacturer. Radiolabeling was carried out by random priming using the Ready-To-Go kit (Amersham, Piscataway, NJ). RNA was isolated from the H526 cell line using the Trizol reagent (Invitrogen) and Northern blotting was done as previously described (23). For determination of VEGF mRNA half-life, quiescent H526 cells were incubated in serum-free medium containing 10 μg/mL actinomycin D (Sigma) in the presence or absence of 100 ng/mL SCF. RNA was isolated at serial times after addition of actinomycin D and Northern blotting was done as described above. VEGF mRNA was quantitated using a Raytest (Newcastle, DE) phosphoimaging station and the AIDA 2.0 software package. The VEGF signal intensity in each lane was normalized to that of β-actin and half-life was calculated as previously described (23).

VEGF Promoter Reporter Assay

H526 cells were transfected with 25 μg of pGL2-VEGF (pVEGF-KpnI, ATCC MB-4, American Type Culture Collection, Manassas, VA; ref. 24), pGL2-Basic (Promega, Madison, WI), or pGL2-Promoter (Promega) along with 5 μg pSV2-β-Gal using the Lipofectamine 2000 transfection reagent as directed by the manufacturer (Invitrogen). A 4-bp mutation (TACGTG→GAATTC) was introduced into the core HIF-1 binding site (−975) in pGL2-VEGF using the QuickChange Mutagenesis kit (Stratagene, La Jolla, CA) to derive the pGL2-VEGFmutant plasmid. Twenty-four hours after transfection, the cells were made quiescent by incubation overnight in serum-free medium and then stimulated with 100 ng/mL SCF or left unstimulated for an additional 24 hours. Cells were lysed and luciferase and β-galactosidase activities were determined with the Luciferase and Beta-Glo Assay systems (Promega) using a single-photon luminometer. Relative luciferase activity in the presence and absence of SCF was calculated by normalizing luciferase activity to β-galactosidase activity.

Electrophoretic Mobility Shift Assay

Nuclear proteins were isolated using the NE-PER kit (Pierce Biotechnology, Inc., Rockford, IL) as directed by the manufacturer. Double-stranded oligonucleotides encompassing the consensus hypoxia-responsive element and an oligonucleotide with a CGT to AAA mutation within the core binding site were purchased from Santa Cruz Biotechnology (Santa Cruz, CA). Oligonucleotides were end-labeled using polynucleotide kinase and electrophoretic mobility shift assays were done using 10 μg of nuclear protein as previously described (25).

Western Blotting

Whole-cell lysates were prepared by resuspending the cells in cold SDS sample buffer [1% SDS, 0.04 mol/L Tris-HCl (pH 6.8), 5% glycerol]. The lysates were boiled and sheared through a 25-gauge needle and protein concentrations were determined using a commercial assay kit (BCA, Pierce Biotechnology). Nuclear protein extracts were added in a 1:1 ratio to 2× SDS sample buffer and boiled. Fifty micrograms of protein were resolved on a 10% polyacrylamide gel and transferred to polyvinylidene difluoride membranes. The membranes were probed with either anti-HIF-1α rabbit polyclonal antiserum (NB 100-134) or affinity-purified antibody (NB 100-449, Novus Biologicals, Inc., Littleton, CO), β-actin monoclonal antibody (Sigma), or N-myc monoclonal antibody (Santa Cruz Biotechnology). Signals were detected and quantitated using the West Pico chemiluminescent system (Pierce Biotechnology) with the aid of a Raytest cooled CCD camera imaging system and the AIDA 2.0 software package. Total cellular HIF-1α was normalized relative to β-actin and nuclear HIF-1α was normalized relative to N-myc, which is autonomously expressed from a highly amplified locus in the H526 cell line.

SCF Enhances VEGF Secretion and mRNA Expression

To determine the effects of SCF on VEGF expression, quiescent SCLC cell lines that express c-Kit (H526, H69, and WBA) and one that does not (H146) were stimulated with SCF for 24 hours in serum-free medium. The conditioned medium was collected and assayed for VEGF using an ELISA (Fig. 1A). The H146 cell line did not express basal levels of VEGF and could not be stimulated to express VEGF by SCF. VEGF secretion doubled in c-Kit-expressing cells treated with SCF relative to controls. Imatinib (STI571) completely blocked the increase in VEGF, confirming that this phenomenon is mediated by c-Kit activation because the other targets of imatinib (platelet-derived growth factor receptor and Abl) are not expressed or activated by SCF in SCLC cell (21). To further determine the manner in which SCF regulated VEGF expression, RNA was isolated from H526 cells at successive time intervals following SCF stimulation. Figure 1B illustrates that SCF induced a 2-fold increase in VEGF mRNA expression by 2 hours after SCF addition, which persisted at 24 hours after exposure. As illustrated in Fig. 1C, VEGF mRNA levels did drop slightly in the 6- to 10-hour period after the SCF exposure in two of four replicates. The reasons for this inconsistent transient drop in VEGF mRNA are unclear at present. On the whole, however, the increase in VEGF mRNA observed correlated very well with the increase in VEGF secretion and was also inhibited by imatinib.

Figure 1.

SCF enhances VEGF expression ∼2-fold. Quiescent SCLC cell lines were incubated in serum-free medium in the absence (NS) or presence of SCF or SCF plus 5 μmol/L imatinib (SCF + STI) for up to 24 h. A, after 24 h, conditioned medium was collected and assayed for secreted VEGF by ELISA. Columns, mean of duplicate assays; bars, SE. B, at the indicated times after addition of SCF to H526 cells, RNA was isolated and analyzed by Northern blot using VEGF and β-actin probes. Representative of four replicates. C, densitometric quantitation of the Northern blot illustrated in B.

Figure 1.

SCF enhances VEGF expression ∼2-fold. Quiescent SCLC cell lines were incubated in serum-free medium in the absence (NS) or presence of SCF or SCF plus 5 μmol/L imatinib (SCF + STI) for up to 24 h. A, after 24 h, conditioned medium was collected and assayed for secreted VEGF by ELISA. Columns, mean of duplicate assays; bars, SE. B, at the indicated times after addition of SCF to H526 cells, RNA was isolated and analyzed by Northern blot using VEGF and β-actin probes. Representative of four replicates. C, densitometric quantitation of the Northern blot illustrated in B.

Close modal

SCF Enhances VEGF Transcription

Because mRNA stabilization has been shown to be one way in which VEGF expression may be regulated, we determined whether SCF treatment altered VEGF mRNA half-life. H526 cells in serum-free medium were treated with actinomycin D (at a concentration that blocked RNA synthesis by >99%) in the presence or absence of SCF and the decay of VEGF mRNA was followed over time by Northern blotting (Fig. 2A). The decay of VEGF mRNA was not significantly altered by SCF treatment, with a densitometrically calculated mRNA half-life of ∼130 minutes (Fig. 2B). This value is generally consistent with previous reports that have determined the VEGF mRNA half-life to be in the range of 40 minutes to 4 hours under normoxic conditions, with a high degree of cell type dependence (7, 8, 26).

Figure 2.

SCF does not affect VEGF mRNA stability. A, quiescent H526 cells were incubated in serum-free medium containing actinomycin D in the absence or presence of SCF. RNA was isolated at the indicated times after addition of actinomycin D and analyzed by Northern blot using VEGF and β-actin probes. Representative of three replicates. B, densitometric analysis of VEGF mRNA levels normalized to actin, indicating a mRNA half-life of ∼130 min in the presence or absence of SCF.

Figure 2.

SCF does not affect VEGF mRNA stability. A, quiescent H526 cells were incubated in serum-free medium containing actinomycin D in the absence or presence of SCF. RNA was isolated at the indicated times after addition of actinomycin D and analyzed by Northern blot using VEGF and β-actin probes. Representative of three replicates. B, densitometric analysis of VEGF mRNA levels normalized to actin, indicating a mRNA half-life of ∼130 min in the presence or absence of SCF.

Close modal

These results suggested that the increase in VEGF mRNA on SCF treatment was transcriptionally mediated. To determine if this was the case, the VEGF promoter cloned upstream of a luciferase reporter gene was transiently transfected into H526 cells, which were then stimulated with SCF for 24 hours in serum-free medium. SCF treatment led to a 70% increase in relative luciferase activity compared with unstimulated control cells (Fig. 3). The activity produced by a control plasmid containing the SV40 promoter was not affected by SCF treatment. The SCF-mediated increase in promoter activity was completely inhibited by 5 μmol/L imatinib. Furthermore, mutation of the HIF-1 binding site at the −975 position in the VEGF promoter (24) also largely eliminated the SCF-mediated increase in promoter activity. These results indicate that SCF enhances VEGF expression via its effects on the VEGF promoter, predominantly through the HIF-1 binding site. However, because significant stimulation of the VEGF promoter did occur even in the absence of a functional hypoxia-responsive element, c-Kit activation may also enhance VEGF promoter activity through non-HIF-1-dependent mechanisms (27, 28).

Figure 3.

SCF-enhanced VEGF promoter activity is dependent on HIF-1 and completely inhibited by imatinib. H526 cells were transfected with the empty pGL2 reporter plasmid (pGL2 Basic), with pGL2 containing the SV40 promoter (pGL2 Promoter), with pGL2 containing the human VEGF 2.3 kb promoter (pGL2 VEGF), and with pGL2 VEGF promoter containing a 4-bp mutation in the core HIF-1 binding site (pGL2 VEGF mutant). Cells were cotransfected with pSV2βGal. The cells were then incubated in serum-free medium in the absence or presence of SCF and 5 μmol/L imatinib (STI571). Luciferase activity, normalized for β-galactosidase, was determined 24 h after addition of SCF. The SV40 promoter was arbitrarily assigned a value of 1.

Figure 3.

SCF-enhanced VEGF promoter activity is dependent on HIF-1 and completely inhibited by imatinib. H526 cells were transfected with the empty pGL2 reporter plasmid (pGL2 Basic), with pGL2 containing the SV40 promoter (pGL2 Promoter), with pGL2 containing the human VEGF 2.3 kb promoter (pGL2 VEGF), and with pGL2 VEGF promoter containing a 4-bp mutation in the core HIF-1 binding site (pGL2 VEGF mutant). Cells were cotransfected with pSV2βGal. The cells were then incubated in serum-free medium in the absence or presence of SCF and 5 μmol/L imatinib (STI571). Luciferase activity, normalized for β-galactosidase, was determined 24 h after addition of SCF. The SV40 promoter was arbitrarily assigned a value of 1.

Close modal

SCF Stimulates Nuclear HIF-1α Levels

Expression of HIF-1α, the major transcriptional regulator of VEGF, can be enhanced by numerous polypeptide growth factors. To initially determine if SCF partially mediates its effect on VEGF expression by enhancing HIF-1α expression, H526 cells were incubated for 24 hours in serum-free medium containing or lacking SCF and the expression of HIF-1α was assessed by Western blotting of whole-cell lysates (Fig. 4A). HIF-1α was difficult to detect in whole-cell lysates made from unstimulated cells and, surprisingly, we observed no detectable increase in HIF-1α in lysates from cells treated with SCF. However, in the presence of CoCl2, a hypoxia-mimetic agent known to stimulate HIF-1α levels by inhibiting its proteasomal degradation (5), a clear increase in HIF-1α levels was seen in SCF-treated cells relative to cells treated with CoCl2 alone. This SCF-mediated increase in cellular HIF-1α levels was inhibited by imatinib, which had no effect on the CoCl2-mediated increase in HIF-1α. To enhance sensitivity in detection of SCF-mediated changes in HIF-1α expression, we isolated the nuclear protein fraction, which should be enriched in HIF-1α. As illustrated in Fig. 4B and C, SCF induced a nearly 2-fold increase in the amount of nuclear HIF-1α, as opposed to CoCl2, which induced an ∼10-fold increase. Imatinib completely blocked the SCF-mediated increase in HIF-1α. Pretreatment with the phosphatidylinositol 3-kinase inhibitor LY294002 also resulted in nearly complete inhibition of nuclear HIF-1α accumulation whereas treatment with the MAPK/extracellular signal–regulated kinase kinase inhibitor PD98059, which inhibits MAPK activation, reduced the increase in nuclear HIF-1α by only about half. It should be noted that after extended electrophoresis, HIF-1α appeared as multiple bands on staining with several different antibodies (not shown), likely the result of multiple posttranslational modifications that the protein is known to undergo (29). In addition, SCF did not affect nuclear HIF-1β levels whereas CoCl2 induced the same modest increase in HIF-1β in the absence or presence of SCF (data not shown).

Figure 4.

Imatinib completely inhibits SCF-enhanced HIF-1α expression. Quiescent H526 cells were incubated for 24 h in the absence or presence of SCF, CoCl2 (hours 20–24), 5 μmol/L imatinib (STI), 10 μmol/L LY294002 (LY), or 40 μmol/L PD98059 (PD) as indicated. A, whole-cell extracts were made and analyzed by Western blotting using a polyclonal HIF-1α antiserum. The band seen in the extracts from cells not treated with CoCl2 (two leftmost lanes) does not comigrate with HIF-1α when electrophoresis is carried out for longer times and is not present when an affinity-purified antibody is used. Representative of four independent experiments. B, nuclear protein extracts were made and analyzed by Western blot using long electrophoresis times and an affinity-purified polyclonal HIF-1α antibody. HIF-1α migrates as several bands of varying mobility, which can be altered by phosphatase treatment (not shown). However, phosphatase treatment does not greatly simplify the pattern, indicating additional posttranslational modifications must be present. Representative of two independent experiments. C, densitometric quantitation of nuclear HIF-1α levels normalizing for N-myc, which is autonomously expressed in this cell line from a highly amplified locus.

Figure 4.

Imatinib completely inhibits SCF-enhanced HIF-1α expression. Quiescent H526 cells were incubated for 24 h in the absence or presence of SCF, CoCl2 (hours 20–24), 5 μmol/L imatinib (STI), 10 μmol/L LY294002 (LY), or 40 μmol/L PD98059 (PD) as indicated. A, whole-cell extracts were made and analyzed by Western blotting using a polyclonal HIF-1α antiserum. The band seen in the extracts from cells not treated with CoCl2 (two leftmost lanes) does not comigrate with HIF-1α when electrophoresis is carried out for longer times and is not present when an affinity-purified antibody is used. Representative of four independent experiments. B, nuclear protein extracts were made and analyzed by Western blot using long electrophoresis times and an affinity-purified polyclonal HIF-1α antibody. HIF-1α migrates as several bands of varying mobility, which can be altered by phosphatase treatment (not shown). However, phosphatase treatment does not greatly simplify the pattern, indicating additional posttranslational modifications must be present. Representative of two independent experiments. C, densitometric quantitation of nuclear HIF-1α levels normalizing for N-myc, which is autonomously expressed in this cell line from a highly amplified locus.

Close modal

SCF Induces an Increase in Nuclear HIF-1α Binding to Its DNA Consensus Element

To determine if the SCF-mediated increase in HIF-1α resulted in enhanced binding to its consensus recognition sequence, we isolated nuclear proteins from H526 cells treated with SCF and/or CoCl2. The nuclear extracts were analyzed by electrophoretic mobility shift assay for their ability to form specific HIF-1α/DNA complexes with an oligonucleotide containing a consensus hypoxia-responsive element. Figure 5 illustrates that SCF enhanced the ability of nuclear extracts to generate specific HIF-1α/DNA complexes. This effect seemed to be additive with CoCl2 in inducing DNA complex formation. Specificity of the complex formation was documented by showing that wild-type, but not mutant, unlabeled oligonucleotide could compete for HIF-1α binding whereas both mutant and wild-type oligonucleotides eliminated the intense nonspecific bands. In addition, a polyclonal HIF-1α antiserum disrupted the HIF-1α complex but not the nonspecific band, whereas the control rabbit immunoglobulin G failed to disrupt either band.

Figure 5.

Formation of HIF-1α/hypoxia-responsive element complexes is enhanced in nuclear extracts from SCF-treated cells. Quiescent H526 cells were incubated in the absence or presence of SCF and CoCl2 (hours 20–24) for 24 h and nuclear proteins were extracted. The nuclear proteins were then incubated with a radiolabeled double-stranded oligonucleotide containing a consensus hypoxia-responsive element and an electrophoretic mobility shift assay was done. Specificity controls included preincubation with an excess of either unlabeled oligonucleotide (Wt), oligonucleotide with a 3-bp mutation in the hypoxia-responsive element (Mut), polyclonal HIF-1α antiserum, or nonimmune rabbit immunoglobulin G (IgG).

Figure 5.

Formation of HIF-1α/hypoxia-responsive element complexes is enhanced in nuclear extracts from SCF-treated cells. Quiescent H526 cells were incubated in the absence or presence of SCF and CoCl2 (hours 20–24) for 24 h and nuclear proteins were extracted. The nuclear proteins were then incubated with a radiolabeled double-stranded oligonucleotide containing a consensus hypoxia-responsive element and an electrophoretic mobility shift assay was done. Specificity controls included preincubation with an excess of either unlabeled oligonucleotide (Wt), oligonucleotide with a 3-bp mutation in the hypoxia-responsive element (Mut), polyclonal HIF-1α antiserum, or nonimmune rabbit immunoglobulin G (IgG).

Close modal

SCF Mediates Its Effect on VEGF mRNA Expression Predominantly through Phosphatidylinositol 3-Kinase–Mediated Modulation of HIF-1α Activity

Activation of c-Kit by SCF is known to initiate signal transduction down the MAPK and phosphatidylinositol 3-kinase pathways, among others (30). To determine the importance of these signaling pathways in the regulation of VEGF expression, we treated H526 cells with SCF over a 24-hour period in the presence of the MAPK/extracellular signal–regulated kinase kinase inhibitor PD98059, the phosphatidylinositol 3-kinase inhibitor LY294002, or DMSO vehicle. Figure 6A illustrates that whereas inhibition of MAPK signaling had little effect on VEGF mRNA expression, phosphatidylinositol 3-kinase inhibition resulted in complete inhibition of SCF-induced expression. These results are in general agreement with those illustrated in Fig. 4, which documented that LY294002 effectively inhibited the SCF-mediated increase in nuclear HIF-1α. To determine whether changes in VEGF expression correlated with hypoxia-responsive element binding, nuclear proteins were extracted from H526 cells treated with SCF in the absence or presence of LY294002 and PD98059. As previously noted, nuclear extracts from CoCl2-treated cells contained a greater amount of HIF-1α capable of binding its cognate DNA sequence than extracts from SCF-treated cells, which had greater binding activity than extracts from unstimulated cells (Fig. 6B). Treatment with LY294002 resulted in a dramatic and nearly complete loss of HIF-1α DNA binding whereas treatment with PD98059 resulted in a more modest reduction in binding. Thus, inhibition of SCF-mediated VEGF expression by phosphatidylinositol 3-kinase inhibition correlated with a decrease in nuclear HIF-1α and a loss in the ability to form complexes with the hypoxia-responsive transcriptional element.

Figure 6.

SCF enhances VEGF mRNA expression via a phosphatidylinositol 3-kinase–mediated increase in HIF-1α activity. Quiescent H526 cells were incubated for 24 h in the absence or presence of SCF, CoCl2 (hours 20–24), 10 μmol/L LY294002, or 40 μmol/L PD98059 as indicated. A, RNA was isolated and analyzed by Northern blot using VEGF and β-actin probes. B, nuclear proteins were isolated and analyzed by electrophoretic mobility shift assay using a radiolabeled hypoxia-responsive element probe.

Figure 6.

SCF enhances VEGF mRNA expression via a phosphatidylinositol 3-kinase–mediated increase in HIF-1α activity. Quiescent H526 cells were incubated for 24 h in the absence or presence of SCF, CoCl2 (hours 20–24), 10 μmol/L LY294002, or 40 μmol/L PD98059 as indicated. A, RNA was isolated and analyzed by Northern blot using VEGF and β-actin probes. B, nuclear proteins were isolated and analyzed by electrophoretic mobility shift assay using a radiolabeled hypoxia-responsive element probe.

Close modal

Angiogenesis has been shown to be critical for growth and progression of numerous tumor types and VEGF has been shown to be a critical mediator of tumor angiogenesis. In SCLC cell, microvessel density and VEGF expression correlate well with both stage and prognosis (19, 20). To better define the regulation of SCLC cell angiogenesis, we explored the role of SCF and c-Kit, components of a common autocrine loop in SCLC cell, in modulating VEGF expression. Activation of c-Kit by SCF treatment of multiple SCLC cell lines resulted in a doubling of VEGF secretion, with a similar increase in VEGF mRNA in the H526 cell line (Fig. 1). SCF treatment resulted in a 70% increase in luciferase reporter expression mediated by a VEGF promoter-reporter construct without a change in VEGF mRNA stability (Figs. 2 and 3). These results are all very consistent with transcriptional enhancement of VEGF expression through activation of c-Kit by SCF, similar to modulation of VEGF expression by other ligands for receptor tyrosine kinases such as insulin-like growth factor I, heregulin, and epidermal growth factor (11, 12, 31). The fact that this process could be interrupted by imatinib (Figs. 1 and 3) confirms its c-Kit dependence and suggests that these findings could have clinical applicability, especially in light of previous observations on imatinib sensitivity of VEGF expression in neuroblastoma (16). In addition to neuroblastoma, angiogenesis has been shown to be an important factor in the biology of gastrointestinal stromal tumors (32). The success of imatinib in treating this tumor may in part be a consequence of inhibition of VEGF expression, a possibility that could also be relevant to the treatment of other malignancies in which c-Kit plays a pathogenic role (33). In addition to inhibiting VEGF expression, imatinib may also inhibit mobilization of angiogenic precursors (15) and pericyte-induced angiogenic maturation mediated by platelet-derived growth factor receptor (3, 16).

In addition to being sensitive to imatinib, the SCF-mediated increase in VEGF promoter activity was also largely dependent on an intact HIF-1 binding site (Fig. 3). Surprisingly, however, we could not show an increase in total cellular HIF-1α by Western blotting after SCF treatment alone. However, in the presence of the hypoxia-mimetic agent CoCl2, SCF treatment did clearly enhance levels of HIF-1α relative to cells treated with CoCl2 alone (Fig. 4A). This additive response has previously been seen when the mechanism of insulin-like growth factor I–mediated VEGF expression was studied (11). One possible reason for the failure to detect an increase in HIF-1α in cells treated with SCF alone is that HIF-1α levels may have been below the limit of detectability by Western blotting of whole-cell lysates in the absence of CoCl2. To improve sensitivity and to allow for direct correlation with hypoxia-responsive element binding, we extracted nuclear proteins and documented that SCF induced a nearly 2-fold increase in nuclear HIF-1α (Fig. 4B and C), which correlates well with the observed increase in VEGF expression (Fig. 1). Because of our difficulty in detecting HIF-1α in whole-cell lysates, we cannot completely exclude the possibility that SCF enhances transport of HIF-1α to the nucleus. However, in the presence of a hypoxia-mimetic agent, it is clear that SCF enhances total cellular as well as nuclear HIF-1α levels (Fig. 4), suggesting that the predominant effect of SCF is mediated by enhanced HIF-1α expression. HIF-1-independent mechanisms may also be contributory, based on the attenuated stimulation of a hypoxia-responsive element mutant promoter-reporter construct by SCF (Fig. 3). In general, however, the ability to form HIF-1α-containing complexes with the hypoxia-responsive element correlated very well with SCF effects on VEGF expression. However, the marked decrease in hypoxia-responsive element binding induced by LY294002 treatment to levels well below that seen using extracts from quiescent cells (Fig. 6) does seem to be slightly out of proportion to the more modest decline in nuclear HIF-1α content after LY294002 treatment (Fig. 4). This observation suggests that SCF-induced phosphatidylinositol 3-kinase signaling, in addition to enhancing the quantity of HIF-1α, may also enhance its ability to bind its cognate DNA element. This could be caused by phosphatidylinositol 3-kinase–mediated posttranslational modification of HIF-1α itself or another component necessary for the formation of a stable complex with the hypoxia-responsive element. Multiple posttranslational modifications have been described for HIF-1α, including prolyl hydroxylation, ubiquitination, SUMOylation, acetylation, S-nitrosation, and phosphorylation (29). Of these, however, S-nitrosation and phosphorylation have been the only modifications where enhancement of transcriptional activation, the effect mediated by SCF, has been documented.

In summary, we have documented for the first time that SCF induces a doubling in VEGF expression in c-Kit-expressing SCLC cell lines and that this increase is due to enhanced VEGF transcription, which can be completely inhibited by imatinib. This enhanced VEGF transcription correlated well with a SCF-mediated increase in nuclear HIF-1α levels and binding to its cognate DNA element and was additive with the effects of the hypoxia-mimetic agent CoCl2. The SCF-mediated increase in VEGF expression seemed to be primarily dependent on activation of phosphatidylinositol 3-kinase because the phosphatidylinositol 3-kinase inhibitor LY294002 completely blocked hypoxia-responsive element binding and SCF-mediated VEGF expression. These data are consistent with a model in which c-Kit-mediated phosphatidylinositol 3-kinase activation leads to enhanced HIF-1α expression and transactivation potential and suggest that direct inhibition of c-Kit or its activation of the phosphatidylinositol 3-kinase-Akt pathway could result in inhibition of angiogenesis, which would be clinically exploitable in SCLC cell and other malignancies in which c-Kit activation occurs.

Grant support: Merit Review Award from the Department of Veterans Affairs (G.W. Krystal).

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

1
Kerbel R, Folkman J. Clinical translation of angiogenesis inhibitors.
Nat Rev Cancer
2002
;
2
:
727
–39.
2
Fidler IJ. The pathogenesis of cancer metastasis: the “seed and soil” hypothesis revisited.
Nat Rev Cancer
2003
;
3
:
453
–8.
3
Bergers G, Benjamin LE. Tumorigenesis and the angiogenic switch.
Nat Rev Cancer
2003
;
3
:
401
–10.
4
Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors.
Nat Med
2003
;
9
:
669
–76.
5
Semenza GL. Targeting HIF-1 for cancer therapy.
Nat Rev Cancer
2003
;
3
:
721
–32.
6
Pugh CW, Ratcliffe PJ. Regulation of angiogenesis by hypoxia: role of the HIF system.
Nat Med
2003
;
9
:
677
–84.
7
Levy AP, Levy NS, Goldberg MA. Post-transcriptional regulation of vascular endothelial growth factor by hypoxia.
J Biol Chem
1996
;
271
:
2746
–53.
8
Liu LX, Lu H, Luo Y, et al. Stabilization of vascular endothelial growth factor mRNA by hypoxia-inducible factor 1.
Biochem Biophys Res Commun
2002
;
291
:
908
–14.
9
Wang GL, Semenza GL. Characterization of hypoxia-inducible factor 1 and regulation of DNA binding activity by hypoxia.
J Biol Chem
1993
;
268
:
21513
–8.
10
Mukhopadhyay CK, Mazumder B, Fox PL. Role of hypoxia-inducible factor-1 in transcriptional activation of ceruloplasmin by iron deficiency.
J Biol Chem
2000
;
275
:
21048
–54.
11
Fukuda R, Hirota K, Fan F, Jung YD, Ellis LM, Semenza GL. Insulin-like growth factor 1 induces hypoxia-inducible factor 1-mediated vascular endothelial growth factor expression, which is dependent on MAP kinase and phosphatidylinositol 3-kinase signaling in colon cancer cells.
J Biol Chem
2002
;
277
:
38205
–11.
12
Laughner E, Taghavi P, Chiles K, Mahon PC, Semenza GL. HER2 (neu) signaling increases the rate of hypoxia-inducible factor 1α (HIF-1α) synthesis: novel mechanism for HIF-1-mediated vascular endothelial growth factor expression.
Mol Cell Biol
2001
;
21
:
3995
–4004.
13
Richard DE, Berra E, Gothie E, Roux D, Pouyssegur J. p42/p44 mitogen-activated protein kinases phosphorylate hypoxia-inducible factor 1α (HIF-1α) and enhance the transcriptional activity of HIF-1.
J Biol Chem
1999
;
274
:
32631
–7.
14
Zhang W, Stoica G, Tasca SI, Kelly KA, Meininger CJ. Modulation of tumor angiogenesis by stem cell factor.
Cancer Res
2000
;
60
:
6757
–62.
15
Heissig B, Werb Z, Rafii S, Hattori K. Role of c-kit/Kit ligand signaling in regulating vasculogenesis.
Thromb Haemost
2003
;
90
:
570
–6.
16
Beppu K, Jaboine J, Merchant MS, Mackall CL, Thiele CJ. Effect of imatinib mesylate on neuroblastoma tumorigenesis and vascular endothelial growth factor expression.
J Natl Cancer Inst
2004
;
96
:
46
–55.
17
Litz J, Warshamana-Greene GS, Sulanke G, Lipson KE, Krystal GW. The multi-targeted kinase inhibitor SU5416 inhibits SCLC growth and angiogenesis, in part by blocking Kit-mediated VEGF expression.
Lung Cancer
2004
;
46
:
283
–91.
18
Lloyd KP, Krystal GW. Role of small-molecule Kit receptor tyrosine kinase inhibitors in the treatment of SCLC.
Clin Lung Cancer
2002
;
3
:
213
–8.
19
Fontanini G, Faviana P, Lucchi M, et al. A high vascular count and overexpression of vascular endothelial growth factor are associated with unfavourable prognosis in operated small cell lung carcinoma.
Br J Cancer
2002
;
86
:
558
–63.
20
Mall JW, Schwenk W, Philipp AW, et al. Serum vascular endothelial growth factor levels correlate better with tumour stage in SCLC than albumin, neuron-specific enolase or lactate dehydrogenase.
Respirology
2002
;
7
:
99
–102.
21
Krystal GW, Honsawek S, Litz J, Buchdunger E. The selective tyrosine kinase inhibitor STI571 inhibits SCLC growth.
Clin Cancer Res
2000
;
6
:
3319
–26.
22
Carney DN, Gazdar AF, Bepler G, et al. Establishment and identification of SCLC cell lines having classic and variant features.
Cancer Res
1985
;
45
:
2913
–23.
23
Krystal G, Birrer M, Way J, et al. Multiple mechanisms for transcriptional regulation of the myc gene family in SCLC.
Mol Cell Biol
1988
;
8
:
3373
–81.
24
Forsythe JA, Jiang BH, Iyer NV, et al. Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1.
Mol Cell Biol
1996
;
16
:
4604
–13.
25
Park GH, Plummer HK III, Krystal GW. Selective Sp1 binding is critical for maximal activity of the human c-kit promoter.
Blood
1998
;
92
:
4138
–49.
26
Levy NS, Chung S, Furneaux H, Levy AP. Hypoxic stabilization of vascular endothelial growth factor mRNA by the RNA-binding protein HuR.
J Biol Chem
1998
;
273
:
6417
–23.
27
Arsham AM, Plas DR, Thompson CB, Simon MC. Akt and hypoxia-inducible factor-1 independently enhance tumor growth and angiogenesis.
Cancer Res
2004
;
64
:
3500
–7.
28
Pore N, Liu S, Shu HK, et al. Sp1 is involved in Akt-mediated induction of VEGF expression through an HIF-1-independent mechanism.
Mol Biol Cell
2004
;
15
:
4841
–53.
29
Brahimi-Horn C, Mazure N, Pouysségur J. Signalling via the hypoxia-inducible factor-1α requires multiple posttranslational modifications.
Cell Signal
2005
;
17
:
1
–9.
30
Blume-Jensen P, Hunter T. Oncogenic kinase signalling.
Nature
2001
;
411
:
355
–65.
31
Zhong H, Chiles K, Feldser D, et al. Modulation of hypoxia-inducible factor 1α expression by the epidermal growth factor/phosphatidylinositol 3-kinase/PTEN/AKT/FRAP pathway in human prostate cancer cells: implications for tumor angiogenesis and therapeutics.
Cancer Res
2000
;
60
:
1541
–5.
32
Takahashi R, Tanaka S, Kitadai Y, et al. Expression of vascular endothelial growth factor and angiogenesis in gastrointestinal stromal tumor of the stomach.
Oncology
2003
;
64
:
266
–74.
33
Heinrich MC, Blanke CD, Druker BJ, Corless CL. Inhibition of KIT tyrosine kinase activity: a novel molecular approach to the treatment of KIT-positive malignancies.
J Clin Oncol
2002
;
20
:
1692
–703.