The Runx genes (Runx1, 2, and 3) regulate cell fate in development and can operate as either oncogenes or tumor suppressors in cancer. The oncogenic potential of ectopic Runx expression has been shown in transgenic mice that develop lymphoma in potent synergy with overexpressed Myc, and in established fibroblasts that display altered morphology and increased tumorigenicity. Candidate oncogenic functions of overexpressed Runx genes include resistance to apoptosis in response to intrinsic and extrinsic stresses. In a search for gene targets responsible for this aspect of Runx phenotype, we have identified three key enzymes in sphingolipid metabolism (Sgpp1, Ugcg, and St3gal5/Siat9) as direct targets for Runx transcriptional regulation in a manner consistent with survival and apoptosis resistance. Consistent with these changes in gene expression, mass spectrometric analysis showed that ectopic Runx reduces intracellular long-chain ceramides in NIH3T3 fibroblasts and elevated extracellular sphingosine 1 phosphate. Runx expression also opposed the activation of c-Jun-NH2-kinase and p38MAPK, key mediators of ceramide-induced death, and suppressed the onset of apoptosis in response to exogenous tumor necrosis factor α. The survival advantage conferred by ectopic Runx could be partially recapitulated by exogenous sphingosine 1 phosphate and was accompanied by reduced phosphorylation of p38MAPK. These results reveal a novel link between transcription factor oncogenes and lipid signaling pathways involved in cancer cell survival and chemoresistance. Cancer Res; 70(14); 5860–9. ©2010 AACR.

The Runx genes comprise a three-member family of mammalian transcription factors that regulate developmental cell fate decisions in which cells are selected to proliferate, differentiate, survive, or die. Unusually, these genes have been shown to act either as oncogenes or as tumor suppressors according to lineage and host cell genetic context (1). The transcriptional targets for Runx regulation are therefore of considerable interest and have already been shown to include factors involved in lineage-specific development as well as regulators of cell cycle checkpoints (24).

Notably, all three Runx genes have been implicated as oncogenes in lymphoid neoplasia in retroviral mutagenesis screens (58), whereas both Runx1 and Runx2 are potently synergistic with Myc (9, 10). The role of Runx in this context seems to be, at least in part, due to inhibition of Myc-induced apoptosis (11). Further evidence of an oncogenic role for Runx2 is provided by its ectopic expression in breast and prostate cancers in which it is associated with metastasis and survival in the bone environment (12, 13), whereas Runx1 promotes tumorigenicity in fibroblasts (14). Runx-mediated survival has also been reported in vitro. In lymphoid cells, Runx1 confers resistance to apoptosis induced by CD3 ligation (15) or dexamethasone (4), whereas in fibroblasts, all three Runx genes were shown to enhance survival under the stress of medium exhaustion (4, 14).

Despite this evidence for potent prosurvival activity, the relevant pathways regulated by Runx are not well known. Our previous gene array analysis revealed a paucity of well-established regulators of apoptotic pathways among the significantly regulated targets. However, a set of 50 genes selected on the basis of significant regulation by all three Runx-regulated genes included three genes encoding enzymes involved in sphingolipid metabolism (4). Each of these genes [Sgpp1, Ugcg, St3gal5(Siat9)] has been reported to play a role in regulating survival and apoptosis (1618), suggesting that they may be important targets and determinants of Runx oncogenic potential. We now show that all three genes act as direct targets for Runx activation or repression. Moreover, biochemical analyzes show that Runx expression causes significant changes in intracellular sphingolipids and resets the ceramide-sphingosine-1-phosphate “rheostat” (19) in favor of cell survival.

Cell culture and transfections

NIH3T3 fibroblast-derived cells expressing Runx1, Runx2, and Runx3 genes from the pBabe retroviral vector system were maintained as previously described (4). Transfections were carried out using the Superfect Transfection Reagent (Qiagen) according to the manufacturer's instructions. Conditions for transfection have been previously described (4). Live/dead cell counts were carried out using a hemocytometer and trypan blue as a vital indicator. Graphs were generated with Sigma-Plot, and error bars relate to SDs. For tumor necrosis factor-α (TNF-α) experiments, cells were treated with 10 ng/mL TNF-α (Peprotech, Inc.) in the presence of 10 μg/mL cycloheximide (Sigma) for the indicated times.

Quantitative real-time PCR

Runx-expressing and control fibroblasts were grown to confluence in duplicate wells (7 d). Cells expressing the RUNX1-ER construct were treated for a further 1 to 24 hours with 4-OHT. cDNA preparation and the microarray assay were performed as described (4). For quantitative real-time PCR (qt-RT-PCR), aliquots (5 μL) of cDNA were amplified in triplicate using primers for murine endogenous control Hprt or primers for murine Sgpp1, St3gal5 (Qiagen QuantiTect Primer Assays), or Ugcg (779F 5′-tttgctcagtacattgctgaagatta-3′ and 861R 5′-acttgagtagacattgaaaacctccaa-3′). Relative quantification was carried out and calibrated to vector control samples (20).

Gel retardation analysis

DNA-binding reactions were performed as previously described (21) using 10 μg of nuclear extracts and 4 ng of 32P- labeled double-stranded oligonucleotide probe. Reactions were resolved by electrophoresis on 4% (w/v) 0.5× Tris-borate EDTA acrylamide gels at 4°C, after which the gels were fixed and dried and complexes visualized by autoradiography. The following double stranded oligonucleotides were used as probes: Sgpp1 site A, 5′-cctttgcagaccacagctgt-3′; Sgpp1 Site Amut, 5′-cctttgcatagcacagctgt-3′. Where indicated, a 50× excess of cold competitor DNA or 2 μL monoclonal Runx2 antiserum (MWG #D130-3) or Runx3 antiserum (a kind gift from Y. Ito, Institute of Molecular and Cell Biology, Singapore) were added to the reaction 15 minutes before the addition of the probe.

Luciferase reporter assays

Transcription assays were performed as described (22) using 2 × 105 cells per 60-mm-diameter tissue culture dish. The assays shown are representative of at least three independent experiments carried out in duplicate. The luciferase activity was normalized to the protein concentration (Bio-Rad protein assay) present in each extract. pGL3Basic luciferase reporter (Promega) was used in all transcription assays. Expression of luciferase activity was driven by a 362-bp fragment amplified from the murine Sgpp1 promoter spanning two consensus Runx binding sites (−133 to −495). Site-directed mutagenesis (QuikChange II Site-Directed Mutagenesis kit, Stratagene) was used to generate bp mutations in the distal site (gaccacag mutated to tagcacag) or in both sites (gaccacag mutated to tagcacag, and caccgctg mutated to cacgcctg) to determine the efficacy of Runx-mediated Sgpp1 repression. pGL3control (Promega) that contains the SV40 promoter and enhancer driving the expression of the luciferase gene was included in all individual experiments to confirm similar levels of transfection efficiency between experiments.

Mass spectrometry

Fibroblast cell pellets (∼2 × 105 cells) containing 15 ng C17-ceramide as an internal standard were extracted using a modified Folch method [extraction with 4 mL chloroform/2 mL methanol/2 mL 0.88% NaCl for each sample, followed by the extraction of upper phase with 3 mL of synthetic lower phase of chloroform/methanol/0.88% NaCl 2:1:1. The combined lower phases were dried under vacuum and redissolved in 50 μL chloroform/methanol 1:1. Two microliters were injected for liquid chromatography/mass spectrometry (LC-MS) analysis]. Cell culture medium (4 mL from ∼2 × 105 cells) containing 30 ng C17-sphingosine-1-phosphate as internal standard, concentrated to 1 mL at room temperature under vacuum, was extracted with 3 × 1 mL n-butanol, combined, concentrated, and redissolved in 40 μL methanol with 2 μL injected for LC-MS analysis using a Shimadzu IT-TOF liquid chromatography tandem mass spectrometry system. In detail, lipid classes of the Folch extract were separated on a normal phase silica gel column (100Å, 4 μm, 150 × 1 mm, MicoSolv Technology) using a hexane/dichloromethane/chloroform/methanol/acetanitrile/water/ethylamine gradient based on the polarity of head group. Accurate mass (with mass accuracy ∼5 ppm) and tandem MS were used for lipid molecular species identification and quantification. Lipid identity was further confirmed by reference to appropriate lipid standards. Mass spectrometer operation conditions were as follows: ESI interface voltage +4.0 kv for positive ESI and −3.5 kv for negative ESI, heat block temperature of 210oÑ, nebulizing gas flow of 1.0 L/min, and CDL temperature of 200oC, with drying gas on. All the solvents used for lipid extraction and LC-MS analysis were LC-MS grade from Fisher Scientific.

Western blotting and antibodies

Preparation of whole-cell protein extracts was performed as previously described (4). Samples equivalent to 50 μg of protein (Bio-Rad protein assay) were resolved on 10% SDS-polyacrylamide gels and transferred to enhanced chemiluminescence (Amersham) nitrocellulose membranes. The antibodies used were α Phospho-p38 mitogen-activated protein kinase (MAPK) 9211 (Cell Signaling Technology) and α p38 MAPK (C20) sc535 (Santa Cruz Biotechnology).

In vitro c-Jun-NH2-kinase kinase assay

Functional c-Jun-NH2-kinase (JNK) activity was measured using a non–radioactive stress–activated protein kinase (SAPK)/JNK Assay kit #9810 (Cell Signaling Technology) according to the manufacturer's instructions. Parallel lysates all contained an equal concentration of protein (Bio-Rad protein assay). Immunoblotting was performed with an anti–phospho-specific cJun (Ser63) antibody as supplied by the kit. The blot was reprobed against α GST(Z5) sc459 (Santa Cruz Biotechnology) to ensure equal loading of c-Jun (1-89) glutathione S-transferase (GST) fusion protein beads.

Caspase-3 detection of apoptosis

Activated caspase-3 was detected using an activated caspase-3 assay kit (#550914, BD Biosciences) according to the manufacturer's instructions; 10,000 events per sample were analyzed on an Epics XL flow cytometer (Beckman Coulter) using the Expo32 software.

Key enzymes in sphingolipid metabolism are direct targets for Runx regulation

In a previous study, we compared the global transcriptional changes in immortalized fibroblasts expressing ectopic Runx1, 2, or 3. In contrast to primary fibroblasts, which enter a state of senescence-like growth arrest in response to Runx (14, 20, 23, 24), these cells undergo a characteristic phenotypic change resembling mesenchymal to epithelial transition and display greatly enhanced survival in response to stresses including medium exhaustion (4). Gene expression microarray analysis revealed a largely overlapping set of target genes with no evidence of genes that were oppositely regulated by each of the three family members. In a common Runx signature set defined by the 50 most significantly regulated genes, we found a preponderance of genes encoding cell surface molecules or secreted factors, but few candidates with an annotated role in control of apoptosis. However, gene ontology analysis indicated a bias toward a small set of genes encoding enzymes involved in sphingolipid metabolism that were of potential significance in this regard (4).

In this study, we have examined Runx regulation of the three sphingolipid enzymes and assessed its potential significance for Runx-mediated survival and oncogenesis. Figure 1A shows the biochemical steps catalyzed by the respective enzymes. The right-hand side of the figure shows the fold change in mRNA levels induced by constitutive expression of Runx1, Runx2, or Runx3 in 3T3 fibroblasts as measured by the Affymetrix gene expression microarray (http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE11732) and validated by qt-RT-PCR. UDP-glucose ceramide glycosyltransferase (Ugcg) and ST3 β-galactoside α-2,3-sialyltransferase 5 (St3gal5/Siat9/GM3 synthase) are both markedly upregulated by Runx gene expression, whereas sphingosine-1-phosphate phosphatase 1 (Sgpp1) is downregulated. Fig. 1B (bottom) shows a kinetic analysis of the mRNA levels after activation of an inducible RUNX1-ER construct. The fact that the observed regulation happens rapidly supports the hypothesis that all three genes act as direct transcriptional targets for the Runx genes, with Ugcg and St3gal5 as targets for activation, whereas Sgpp1 is repressed. As a further test for direct regulation, we examined the requirement for protein synthesis by inducing RUNX1-ER in the presence of cycloheximide. Although interpretation of these experiments was complicated by the nonspecific effects of cycloheximide on the stability of some transcripts, it was clear that Ugcg induction and Sgpp1 repression were insensitive to cycloheximide inhibition. St3gal5 induction was preserved at very early time points but was lost after 6 hours due to the nonspecific stabilization of transcripts by cycloheximide (Supplementary Fig. S1).

Figure 1.

Functions of Runx-regulated genes in lipid metabolism and validation of microarray data analysis. A, biochemical steps catalyzed by Runx gene targets involved in sphingolipid metabolism and their respective enzyme commission numbers. Validation of array data for selected target genes by qt-RT-PCR (SYBR green). Columns, fold regulation as measured by Affymetrix array and qt-RT-PCR. False discovery rate (FDR) estimates for the individual gene probe sets on the Affymetrix chips are indicated. B, kinetic analysis of mRNA levels of each enzyme after activation of an inducible RUNX1-ER construct.

Figure 1.

Functions of Runx-regulated genes in lipid metabolism and validation of microarray data analysis. A, biochemical steps catalyzed by Runx gene targets involved in sphingolipid metabolism and their respective enzyme commission numbers. Validation of array data for selected target genes by qt-RT-PCR (SYBR green). Columns, fold regulation as measured by Affymetrix array and qt-RT-PCR. False discovery rate (FDR) estimates for the individual gene probe sets on the Affymetrix chips are indicated. B, kinetic analysis of mRNA levels of each enzyme after activation of an inducible RUNX1-ER construct.

Close modal

Screening of the promoter regions of the three genes revealed consensus Runx binding sites close to the transcriptional start site in each case. St3gal5 has three discrete sites (−602, −1,151, and −1834), whereas Ugcg has one site (−345) and Sgpp1 has two closely linked sites (−416 and −348). The Sgpp1 promoter region is the most highly conserved sequence across mammalian species and also contains conserved E box motifs next to the proximal site (B). As shown in Fig. 2A, chromatin immunoprecipitation confirmed Runx binding at each of these sites. Runx2-expressing cells were used in this instance to take advantage of antibodies suitable for use under chromatin immunoprecipitation conditions.

Figure 2.

A, Runx2 occupancy of promoter regions of Ugcg, St3gal5, and Sgpp1 in vivo as assessed by chromatin immunoprecipitation analysis. Specificity was shown by the lack of site recognition by a negative control polyclonal antiserum, α GST, or when primary antibody was not included in the immunoprecipitation reaction. B, DNA mobility shift assay using Runx consensus Sgpp1 oligonucleotide A from the Sgpp1 promoter and nuclear lysates prepared from Runx2- or Runx3-expressing NIH3T3 fibroblasts. Specificity of binding was indicated using excess cold competitor oligonucleotides and monoclonal antibodies specific for Runx2 or Runx3. C, the ability of Runx to trans-repress the Sgpp1 regulatory element is dependent on two Runx consensus motifs in the Sgpp1 promoter. The data are representative of three independent transient transfections into HEK-293T cells and are normalized to the protein concentration present in the extract.

Figure 2.

A, Runx2 occupancy of promoter regions of Ugcg, St3gal5, and Sgpp1 in vivo as assessed by chromatin immunoprecipitation analysis. Specificity was shown by the lack of site recognition by a negative control polyclonal antiserum, α GST, or when primary antibody was not included in the immunoprecipitation reaction. B, DNA mobility shift assay using Runx consensus Sgpp1 oligonucleotide A from the Sgpp1 promoter and nuclear lysates prepared from Runx2- or Runx3-expressing NIH3T3 fibroblasts. Specificity of binding was indicated using excess cold competitor oligonucleotides and monoclonal antibodies specific for Runx2 or Runx3. C, the ability of Runx to trans-repress the Sgpp1 regulatory element is dependent on two Runx consensus motifs in the Sgpp1 promoter. The data are representative of three independent transient transfections into HEK-293T cells and are normalized to the protein concentration present in the extract.

Close modal

In view of the evidence that Sgpp1 plays a central role in the regulation of the ceramide-sphingosine 1 phosphate (S1P) rheostat and cell survival, we focused further efforts on validating the regulatory consequences of Runx binding. We confirmed that Runx proteins from nuclear extracts bound specifically to both site A and site B in electrophoretic mobility shift assay (data not shown; Fig. 2B). A luciferase expression construct with a basal promoter under control of the Sgpp1 regulatory element (−133 to −495) showed negative regulation in the presence of ectopic Runx, which was reduced by mutation of site A and abolished in the double mutant (Fig. 2C). Basal levels of the double mutant were similar to those of the wild-type promoter in the absence of ectopic Runx expression, suggesting that steady-state levels of Runx in fibroblasts are too low to cause significant repression.

Ceramide and S1P levels are modulated by Runx

To explore the biochemical consequences of altered transcription of genes encoding sphingolipid enzymes, we examined the ceramide/S1P profiles by MS in the presence and absence of ectopic Runx. Levels of extracellular S1P were found to be essentially undetectable in serum-free cultures of control cells. However, although the data were somewhat variable, we were able to detect S1P in the culture medium of cells expressing Runx with values ranging from 0 to 0.88 ng S1P/5 mL culture medium of 2 × 105 cells cultured serum free. In contrast, we consistently observed a marked reduction in the level of several long-chain ceramides in the presence of ectopic Runx (Fig. 3A). Ceramides 16:0, 24:1, and 24:0 are typical of the salvage pathway of ceramide synthesis, suggesting that Runx expression opposes sphingosine recycling through this route (17). The most dramatic effect observed was on the levels of 16.0 ceramide (Fig. 3A), which has previously been reported to respond to Sgpp1 expression and is implicated as a critical component of ceramide-induced apoptosis (17, 25, 26).

Figure 3.

A, the ceramides in cultures of Puro vector control and Runx1 expressing NIH3T3 fibroblasts were extracted, separated, identified, and semi-quantitated by high-performance liquid chromatography mass spectrometry. Columns, means in which n = 3 from one experiment typical of three; bars, SD. B, Puro vector control and Runx1-expressing NIH3T3 fibroblasts were grown under conditions of medium exhaustion for 10 d. Dead cell counts were determined each day by trypan blue exclusion. C, on the indicated days, adherent cells from the same cultures were lysed and probed against antibodies to phospho-p38MAPK or p38MAPK as a loading control. D, parallel cell pellets were lysed for immunoprecipitation and incubated with c-Jun (1–89) GST fusion protein beads. Functional JNK activity was quantified in the presence of ATP by immunoblotting with a phospho-specific antibody to c-Jun (Ser 63).

Figure 3.

A, the ceramides in cultures of Puro vector control and Runx1 expressing NIH3T3 fibroblasts were extracted, separated, identified, and semi-quantitated by high-performance liquid chromatography mass spectrometry. Columns, means in which n = 3 from one experiment typical of three; bars, SD. B, Puro vector control and Runx1-expressing NIH3T3 fibroblasts were grown under conditions of medium exhaustion for 10 d. Dead cell counts were determined each day by trypan blue exclusion. C, on the indicated days, adherent cells from the same cultures were lysed and probed against antibodies to phospho-p38MAPK or p38MAPK as a loading control. D, parallel cell pellets were lysed for immunoprecipitation and incubated with c-Jun (1–89) GST fusion protein beads. Functional JNK activity was quantified in the presence of ATP by immunoblotting with a phospho-specific antibody to c-Jun (Ser 63).

Close modal

Runx expression reduces stress-associated kinase signaling

To investigate the consequences of long-chain ceramide depletion for Runx-dependent cell survival, we examined the functional activity of p38MAPK and JNK, which play central roles in stress-induced cell death and apoptotic signaling by ceramide (2729). For this purpose, NIH3T3 fibroblasts expressing ectopic Runx1 or control vector were grown under conditions of medium exhaustion, and adherent cells were harvested every 1 to 3 days for analysis of phospho-p38MAPK expression and JNK activity. Consistent with our previous data (4), expression of ectopic Runx conferred protection against stress-induced cell death under these growth conditions (Fig. 3B). Furthermore, Runx-dependent cell survival correlated with reduced activation of the p38MAPK and JNK signaling pathways as measured by the accumulation of phosphorylated p38MAPK and c-Jun, respectively (Fig. 3C and D).

Runx delays TNF-α–induced cell death

TNF-α has been reported to induce cell death through an accumulation of intracellular ceramide and downstream activation of the JNK signaling pathway (28). To determine whether this response is modified by ectopic Runx1, we examined the effects of TNF-α on the survival of vector control and Runx1-expressing NIH3T3 fibroblasts. Cycloheximide was included in short-term assays to suppress the synthesis of short-lived antiapoptotic proteins that can reduce the effects of TNF-α (30). As shown in Fig. 4A, morphologic differences were observed as early as 2 to 4 hours after TNF-α and cycloheximide treatment, with vector control cells rounding up and detaching from the substratum far more readily than their Runx1-expressing counterparts. Quantitation of adherent and nonadherent cells by trypan blue exclusion confirmed greater survival in the presence of ectopic Runx1 (Fig. 4B). The effect was exaggerated after 5 hours of exposure to TNF-α and cycloheximide (Fig. 4B) but lost by 24 hours when widespread cell death was observed throughout (data not shown). To determine whether Runx1 was protecting against apoptosis, fluorescence-activated cell sorting (FACS) analysis was performed on caspase-3–labeled cells grown in the presence and absence of TNF-α for longer time periods. A highly significant difference between Runx and control cultures was observed from the earliest time point up to 48 hours (Fig. 4C), with Runx-expressing cells showing a marked resistance to induction of caspase-3 over the time course.

Figure 4.

A, Puro vector control and Runx1-expressing fibroblasts were grown in the presence and absence of 10 ng/mL TNF-α and 10 μg/mL cycloheximide (CHX) for up to 4 h. Cultures were photographed every 2 h. B, adherent and nonadherent cells from parallel cultures were harvested after 3 and 5 h of TNF-α/cycloheximide treatment, and viability counts were performed by trypan blue exclusion. Ctrl, control. Ci, cells treated with 10 ng/mL TNF-α alone for up to 48 h were stained for intracellular activated caspase-3 and analyzed by FACS at 6, 12, 17, 24, and 48 h; Cii, representative histogram plots showing caspase-3 activation in control and TNF-α-treated cells at 24 h.

Figure 4.

A, Puro vector control and Runx1-expressing fibroblasts were grown in the presence and absence of 10 ng/mL TNF-α and 10 μg/mL cycloheximide (CHX) for up to 4 h. Cultures were photographed every 2 h. B, adherent and nonadherent cells from parallel cultures were harvested after 3 and 5 h of TNF-α/cycloheximide treatment, and viability counts were performed by trypan blue exclusion. Ctrl, control. Ci, cells treated with 10 ng/mL TNF-α alone for up to 48 h were stained for intracellular activated caspase-3 and analyzed by FACS at 6, 12, 17, 24, and 48 h; Cii, representative histogram plots showing caspase-3 activation in control and TNF-α-treated cells at 24 h.

Close modal

These data suggest that modulation of sphingolipid enzymes by ectopic Runx resets the sphingolipid rheostat against intracellular ceramide-induced apoptosis and in favor of cell survival.

Runx survival is partially reproduced by exogenous S1P

The reduction of intracellular ceramide in Runx-expressing cells may be due to the cumulative action of the three target enzymes leading to the synthesis of glycosylated forms or gangliosides (Ugcg, St3gal5) or to the accumulation of S1P (Fig. 5). S1P functions as a lipid signal that can promote proliferation and suppress ceramide-mediated apoptosis. S1P is actively transported outside the cell where it signals through a family of G protein–coupled receptors. In addition, S1P seems to be able to act through as yet uncharacterized intracellular receptors (31). Although the detection of extracellular S1P was variable in Runx-expressing fibroblasts, it is probable that this pathway is constitutively activated because other pathways of S1P removal are found in cells and S1P was essentially undetectable in control cultures. To explore this hypothesis, we tested whether Runx-expressing cells retained sensitivity to growth regulation by exogenous S1P. NIH3T3 fibroblasts expressing ectopic Runx1 or control vector were grown to confluence and cultured under conditions of medium exhaustion for 8 days in the presence or absence of increasing concentrations of S1P. Viability counts were then performed on detached cells in the supernatant and the adherent cultures harvested to determine the phosphorylation status of p38MAPK.

Figure 5.

A model depicting the sphingolipid rheostat and the enzyme activities of three Runx gene targets relative to the central exchange of intracellular ceramide and S1P. The dynamic balance is normally tightly regulated and has direct consequences for cell survival and apoptosis.

Figure 5.

A model depicting the sphingolipid rheostat and the enzyme activities of three Runx gene targets relative to the central exchange of intracellular ceramide and S1P. The dynamic balance is normally tightly regulated and has direct consequences for cell survival and apoptosis.

Close modal

As shown in Fig. 6A, exogenous S1P reduced cell death in a dose-dependent manner in both cell backgrounds, but the effect was more pronounced in the absence of ectopic Runx1, supporting the proposal that S1P signaling was already activated by elevated S1P generation in the Runx-expressing fibroblasts. Cell viability counts confirmed this trend, with the difference between Runx-expressing and control cultures reduced to only 3% in the presence of 1 × 10−6 mol/L S1P (Fig. 6B). These data support previous studies in NIH3T3 fibroblasts showing a reduction in stress-induced apoptosis in the presence of exogenous S1P (32). Furthermore, the improved survival of control cultures correlated with reduced expression levels of phosphorylated p38MAPK (Fig. 6C). At higher doses of S1P, expression levels of phospho p38MAPK were comparable with those in the Runx-expressing cultures, which were relatively refractory to S1P (Fig. 6C). These data suggest that although p38MAPK inhibition correlates with improved cell survival of control cultures, ectopic expression of Runx has already saturated the relevant survival pathway.

Figure 6.

S1P signaling enhances cell survival and p38MAPK dephosphorylation in control fibroblasts but is effectively silenced in the presence of ectopic Runx1. Confluent cultures were grown under conditions of nutrient starvation for 8 d in the presence of increasing concentrations of S1P. The data are representative of two independent experiments. A, day 8 dead cell suspension counts quantified by trypan blue exclusion. B, corresponding viability counts showing the reduced survival advantage of Runx-expressing fibroblasts compared with control cultures in the presence of extracellular S1P. C, adherent cells from the same cell cultures were harvested and analyzed for phosphorylated p38MAPK expression by Western blotting using a phospho-specific p38MAPK antibody. Levels are expressed relative to the total p38MAPK loading control.

Figure 6.

S1P signaling enhances cell survival and p38MAPK dephosphorylation in control fibroblasts but is effectively silenced in the presence of ectopic Runx1. Confluent cultures were grown under conditions of nutrient starvation for 8 d in the presence of increasing concentrations of S1P. The data are representative of two independent experiments. A, day 8 dead cell suspension counts quantified by trypan blue exclusion. B, corresponding viability counts showing the reduced survival advantage of Runx-expressing fibroblasts compared with control cultures in the presence of extracellular S1P. C, adherent cells from the same cell cultures were harvested and analyzed for phosphorylated p38MAPK expression by Western blotting using a phospho-specific p38MAPK antibody. Levels are expressed relative to the total p38MAPK loading control.

Close modal

Overexpression of Runx genes has context-specific effects, inducing growth arrest or senescence in most primary cell types, but promoting proliferation and/or survival in cells expressing collaborating oncogenes or mutated tumor suppressor pathways (1, 14, 24, 33). In NIH3T3 fibroblasts, which lack Cdkn2a (Ink4a/Arf) function, overexpression of Runx potently induces survival under stress conditions. Our previous exploration of the likely effector genes in this phenotype revealed a paucity of candidates with established roles in apoptosis or survival, with the notable exception of a small set of genes involved in sphingolipid metabolism that were overrepresented as a gene ontology subset (4). In this study, we have shown that the Runx genes can directly regulate three genes encoding enzymes with central roles in sphingolipid pathways (Fig. 5). Of particular note is the ability of all three Runx genes to repress expression of Sgpp1, a key player in the sphingosine rheostat that governs the interchange between proapoptotic ceramides (34) and S1P, a well-established ligand in survival signaling (19). Moreover, ectopic Runx expression was shown to drive a major reduction in the level of specific intracellular ceramide species, a finding that was associated with reduced induction of stress-associated p38MAPK and JNK activity. The selective reduction in a subset of ceramide species points to these lipids as being specific substrates for Ugcg and also suggests that these particular ceramides signal through specific target proteins to mediate proapoptotic effects. The selective action of particular species within a lipid class has been previously recognized for diacylglycerol and phosphatidic acid (35). Resistance to death induced by TNF-α, which operates through ceramide, was also confirmed in Runx-expressing cells, whereas the Runx-associated survival phenotype could be at least partially recapitulated by the addition of exogenous S1P. These results suggest that modulation of sphingolipid metabolism underlies the oncogenic and prosurvival effects of ectopic Runx expression in multiple contexts (4, 11, 15) and indicates the possibility of a wider role for Runx in cancer chemoresistance.

Published studies provide strong support for the hypothesis that the three novel Runx target genes we have described play a significant role in the observed survival phenotype. For example, RNAi knockdown of Sgpp1 expression was shown to promote cell survival with concomitant accumulation of intracellular and extracellular S1P, whereas ectopic expression of Sgpp1 was reported to drive the conversion of S1P to long-chain ceramides and a subsequent apoptotic response (32). Ectopic expression of Ugcg has been reported to rescue cells from ceramide-induced apoptosis (16). Furthermore, multidrug resistance of breast cancer cell lines has been associated with the shunting of ceramide into a glucosyl ceramide sink and can be reversed by small interfering RNA or oligonucleotide blockade of Ugcg (36). The effects of upregulation of St3gal5 are less readily predicted, as high levels of GM3 are associated with the induction of apoptosis in some cancer cells (18), whereas reintroduction of St3gal5 into lines that fail to synthesize GM3 was shown to enhance their ability to form colonies in soft agar and increase their resistance to stress-induced apoptosis (37). We suggest that the outcome may reflect the steady-state levels of GM3 in different cellular backgrounds. In this regard, it is notable that the upregulation of Ugcg by Runx is much more marked than St3gal5, and that glycosylation of ceramide is also a key step in the synthesis of cerebrosides and gangliosides other than GM3.

Improved survival of Runx-expressing fibroblasts also correlated with reduced levels of intracellular long-chain ceramides. The induction of apoptosis through ceramide production is complex, involving both direct and indirect modes of action and multiple signaling pathways. Mitochondrial damage and caspase release is driven by the formation of ceramide channels in the mitochondrial membrane (38) but also through the activation of stress-activated kinases such as p38MAPK, which further activate proapoptotic Bax and Bak, and inhibit antiapoptotic Bcl2 and Bcl-XL (39). It has been suggested that the solubility of these target molecules enables ceramide at nonmitochondrial locations such as the endoplasmic reticulum membranes to participate in the apoptotic process (40). Notably, we observed reduced steady-state p38MAPK phosphorylation and JNK activity in stressed cells expressing ectopic Runx, and both of these kinase pathways have been reported to be induced by intracellular ceramide (29, 39). Further evidence of a role for sphingolipid-mediated changes in the Runx survival phenotype was provided by the greater resistance of Runx-expressing cells to TNF-α–induced apoptosis, which has also been reported to operate through ceramide and JNK signaling pathways (29).

It is notable that we were able to partially recapitulate the survival phenotype of Runx-expressing cells through exogenous administration of S1P to control cells. S1P can mediate survival by binding to a series of cell surface receptors (reviewed in ref. 41) but can also act by a less well-characterized intracellular route (17). The variable detection of free S1P in the medium of Runx-expressing cells could suggest that the latter route is operative here. Nevertheless, it seems that S1P pathways are fully engaged in these Runx-expressing cells, as exogenous S1P caused no additional survival benefit in this context. Sgpp1 is not the only plasma membrane–located enzyme able to degrade S1P, there being a family of lipid phosphate phosphatases able to hydrolyse a range of signaling lipids including S1P and lysophosphatidate (42). The lipid phosphate phosphatases seem to be regulated by substrate availability; thus, although a reduction in Sgpp1 could reduce the degradation of S1P in the proximity of the S1P receptor following its release from the cell, it will not necessarily induce sustained elevation of medium S1P concentration. In support of this, it was reported that reduced expression levels of lipid phosphate phosphatase 1 in ovarian cancer cells was not accompanied by a corresponding increase in extracellular lysophosphatidate (43).

The involvement of sphingolipid enzymes in cancer and chemoresistance (27, 44) suggests a wider role for Runx in these phenomena. Our results establish a link between previous reports of the Runx genes as targets for retroviral activation in an in vivo model of acquired imatinib resistance in CML (45) and studies showing that imatinib resistance can be conferred through changes in the ceramide-S1P rheostat (46). Moreover, the ability of Runx to confer metastatic potential may be mediated by the ability to promote close adhesion (4, 14) and sphingolipid-mediated cell survival under adverse conditions in vivo. Finally, the Runx genes have been widely studied as essential players in lineage-specific differentiation in which they can act as both positive and negative regulators of proliferation according to context (4749). The possibility that these regulatory steps are mediated through sphingolipid metabolism merits further investigation.

No potential conflicts of interest were disclosed.

We thank Dr. S. Wotton for helpful comments and discussions.

Grant Support: Cancer Research UK and Leukaemia and Lymphoma Research to the Molecular Oncology Laboratory. Lipidomic analyses were performed at the Babraham Institute and supported by grants from Cancer Research UK and the Biotechnology and Biological Sciences Research Council (M.J.O. Wakelam).

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
Blyth
K
,
Cameron
ER
,
Neil
JC
. 
The Runx gene family: gain or loss of function in cancer
.
Nat Rev Cancer
2005
;
5
:
376
87
.
2
Lutterbach
B
,
Hiebert
SW
. 
Role of the transcription factor AML-1 in acute leukemia and hematopoietic differentiation
.
Gene
2000
;
245
:
223
35
.
3
Otto
F
,
Lubbert
M
,
Stock
M
. 
Upstream and downstream targets of RUNX proteins
.
J Cell Biochem
2003
;
89
:
9
18
.
4
Wotton
S
,
Terry
A
,
Kilbey
A
, et al
. 
Gene array analysis reveals a common Runx transcriptional programme controlling cell adhesion and survival
.
Oncogene
2008
;
27
:
5856
66
.
5
Stewart
M
,
Terry
A
,
Hu
M
, et al
. 
Proviral insertions induce the expression of bone-specific isoforms of PEBP2αA (CBFA1): evidence for a new myc collaborating oncogene
.
Proc Natl Acad Sci U S A
1997
;
94
:
8646
51
.
6
Wotton
S
,
Stewart
M
,
Blyth
K
, et al
. 
Proviral insertion indicates a dominant oncogenic role for Runx1/AML1 in T-cell lymphoma
.
Cancer Res
2002
;
62
:
7181
5
.
7
Stewart
M
,
Mackay
N
,
Cameron
ER
,
Neil
JC
. 
The common retroviral insertion locus Dsi1 maps 30 kb upstream of the P1 promoter of the murine Runx3/Cbfa3/Aml2 gene
.
J Virol
2002
;
76
:
4364
9
.
8
Lund
AH
,
van Lohuizen
M
. 
RUNX: a trilogy of cancer genes
.
Cancer Cell
2002
;
1
:
213
5
.
9
Vaillant
F
,
Blyth
K
,
Terry
A
, et al
. 
A full length Cbfa1 gene product perturbs T-cell development and promotes lymphomagenesis in synergy with MYC
.
Oncogene
1999
;
18
:
7124
34
.
10
Blyth
K
,
Terry
A
,
Mackay
N
, et al
. 
Runx2: a novel oncogenic effector revealed by in vivo complementation and retroviral tagging
.
Oncogene
2001
;
20
:
295
302
.
11
Blyth
K
,
Vaillant
F
,
Mackay
N
, et al
. 
Runx2 and MYC collaborate in lymphoma development by suppressing apoptotic and growth arrest pathways in vivo
.
Cancer Res
2006
;
66
:
2195
201
.
12
Pratap
J
,
Javed
A
,
Barnes
GL
, et al
. 
Runx2 regulates tumor cell invasive properties and contributes to the osteolytic bone disease associated with metastatic cancer cells
.
J Bone Miner Res
2005
;
20
:
16
.
13
Barnes
GL
,
Javed
A
,
Waller
SM
, et al
. 
Osteoblast-related transcription factors Runx2 (Cbfa1/AML3) and MSX2 mediate the expression of bone sialoprotein in human metastatic breast cancer cells
.
Cancer Res
2003
;
63
:
2631
7
.
14
Wotton
S
,
Blyth
K
,
Kilbey
A
, et al
. 
RUNX1 transformation of primary embryonic fibroblasts is revealed in the absence of p53
.
Oncogene
2004
;
23
:
5476
86
.
15
Fujii
M
,
Hayashi
K
,
Niki
M
, et al
. 
Overexpression of AML1 renders a T hybridoma resistant to T cell receptor-mediated apoptosis
.
Oncogene
1998
;
17
:
1813
20
.
16
Bleicher
RJ
,
Cabot
MC
. 
Glucosylceramide synthase and apoptosis
.
Biochimica et biophysica acta-molecular and cell biology of lipids
2002
;
1585
:
172
8
.
17
Le Stunff
H
,
Galve-Roperh
I
,
Peterson
C
,
Milstien
S
,
Spiegel
S
. 
Sphingosine-1-phosphate phosphohydrolase in regulation of sphingolipid metabolism and apoptosis
.
J Cell Biol
2002
;
158
:
1039
49
.
18
Watanabe
R
,
Ohyama
C
,
Aoki
H
, et al
. 
Ganglioside G(M3) overexpression induces apoptosis and reduces malignant potential in murine bladder cancer
.
Cancer Res
2002
;
62
:
3850
4
.
19
Maceyka
M
,
Payne
SG
,
Milstien
S
,
Spiegel
S
. 
Sphingosine kinase, sphingosine-1-phosphate, and apoptosis
.
Biochim Biophys Acta
2002
;
1585
:
193
201
.
20
Kilbey
A
,
Blyth
K
,
Wotton
S
, et al
. 
Runx2 disruption promotes immortalization and confers resistance to oncogene-induced senescence in primary murine fibroblasts
.
Cancer Res
2007
;
67
:
11263
71
.
21
Kilbey
A
,
Black
EJ
,
Unlu
M
,
Gillespie
DAF
. 
The v-Jun oncoprotein replaces p39 c-Jun as the predominant AP-1 constituent in ASV17-transformed fibroblasts: implications for SAPK/JNK-mediated signal transduction
.
Oncogene
1996
;
12
:
2409
18
.
22
Taylor
ER
,
Boner
W
,
Dornan
ES
,
Corr
EM
,
Morgan
IM
. 
UVB irradiation reduces the half-life and transactivation potential of the human papillomavirus 16 E2 protein
.
Oncogene
2003
;
22
:
4469
77
.
23
Kilbey
A
,
Terry
A
,
Cameron
ER
,
Neil
JC
. 
Oncogene induced senescence: an essential role for Runx
.
Cell Cycle
2008
;
7
:
2333
40
.
24
Wolyniec
K
,
Wotton
S
,
Kilbey
A
, et al
. 
RUNX1 and its fusion oncoprotein derivative RUNX1-ETO induce senescence-like growth arrest independently of replicative stress
.
Oncogene
2009
;
28
:
2502
12
.
25
Kolesnick
R
,
Hannun
YA
. 
Ceramide and apoptosis
.
Trends Biochem Sci
1999
;
24
:
224
5
.
26
Le Stunff
H
,
Giussani
P
,
Maceyka
M
,
Lepine
S
,
Milstien
S
,
Spiegel
S
. 
Recycling of sphingosine is regulated by the concerted actions of sphingosine-1-phosphate phosphohydrolase 1 and sphingosine kinase 2
.
J Biol Chem
2007
;
282
:
34372
80
.
27
Oskouian
B
,
Sooriyakumaran
P
,
Borowsky
AD
, et al
. 
Sphingosine-1-phosphate lyase potentiates apoptosis via p53-and p38-dependent pathways and is down-regulated in colon cancer
.
Proc Natl Acad Sci U S A
2006
;
103
:
17384
9
.
28
Cuvillier
O
. 
Sphingosine in apoptosis signaling
.
Biochim Biophys Acta
2002
;
1585
:
153
62
.
29
Verheij
M
,
Bose
R
,
Lin
XH
, et al
. 
Requirement for ceramide-initiated SAPK/JNK signalling in stress-induced apoptosis
.
Nature
1996
;
380
:
75
9
.
30
Jin
S
,
Ray
RM
,
Johnson
LR
. 
TNF-α/cycloheximide-induced apoptosis in intestinal epithelial cells requires Rac1-regulated reactive oxygen species
.
Am J Physiol
2008
;
294
:
G928
37
.
31
Spiegel
S
,
Milstien
S
. 
Sphingosine-1-phosphate: an enigmatic signalling lipid
.
Nat Rev Mol Cell Biol
2003
;
4
:
397
407
.
32
Mandala
SM
,
Thornton
R
,
Galve-Roperh
I
, et al
. 
Molecular cloning and characterization of a lipid phosphohydrolase that degrades sphingosine-1-phosphate and induces cell death
.
Proc Natl Acad Sci U S A
2000
;
97
:
7859
64
.
33
Linggi
B
,
Muller-Tidow
C
,
van de Locht
L
, et al
. 
The t(8;21) fusion protein, AML1 ETO, specifically represses the transcription of the p14(ARF) tumor suppressor in acute myeloid leukemia
.
Nat Med
2002
;
8
:
743
50
.
34
Obeid
LM
,
Linardic
CM
,
Karolak
LA
,
Hannun
YA
. 
Programmed cell-death induced by ceramide
.
Science
1993
;
259
:
1769
71
.
35
Hodgkin
MN
,
Pettitt
TR
,
Martin
A
,
Michell
RH
,
Pemberton
AJ
,
Wakelam
MJO
. 
Diacylglycerols and phosphatidates: Which molecular species are intracellular messengers?
Trends Biochem Sci
1998
;
23
:
200
4
.
36
Zhang
XF
,
Li
J
,
Qiu
ZG
,
Gao
P
,
Wu
XJ
,
Zhou
GY
. 
Co-suppression of MDR1 (multidrug resistence 1) and GCS (glucosylceramide synthase) restores sensitivity to mutidrug resistance breast cancer cells by RNA interference (RNAi)
.
Cancer Biol Ther
2009
;
8
:
1117
21
.
37
Uemura
S
,
Kabayama
K
,
Noguchi
M
,
Igarashi
Y
,
Inokuchi
J
. 
Sialylation and sulfation of lactosylceramide distinctly regulate anchorage-independent growth, apoptosis, and gene expression in 3LL Lewis lung carcinoma cells
.
Glycobiology
2003
;
13
:
207
16
.
38
Siskind
LJ
. 
Mitochondrial ceramide and the induction of apoptosis
.
J Bioenerg Biomembr
2005
;
37
:
143
53
.
39
Chen
CL
,
Lin
CF
,
Chang
WT
,
Huang
WC
,
Teng
CF
,
Lin
YS
. 
Ceramide induces p38 MAPK and JNK activation through a mechanism involving a thioredoxin-interacting protein-mediated pathway
.
Blood
2008
;
111
:
4365
74
.
40
Simon
CG
,
Holloway
PW
,
Gear
ARL
. 
Exchange of C-16-ceramide between phospholipid vesicles
.
Biochemistry
1999
;
38
:
14676
82
.
41
Pyne
S
,
Pyne
N
. 
Sphingosine 1-phosphate signalling via the endothelial differentiation gene family of G-protein-coupled receptors
.
Pharmacol Ther
2000
;
88
:
115
31
.
42
Brindley
DN
,
Pilquil
C
. 
Lipid phosphate phosphatases and signaling
.
J Lipid Res
2009
;
50
:
S225
30
.
43
Tanyi
JL
,
Hasegawa
Y
,
Lapushin
R
, et al
. 
Role of decreased levels of lipid phosphate phosphatase-1 in accumulation of lysophosphatidic acid in ovarian cancer
.
Clin Cancer Res
2003
;
9
:
3534
45
.
44
Liu
YY
,
Yu
JY
,
Yin
DM
, et al
. 
A role for ceramide in driving cancer cell resistance to doxorubicin
.
FASEB J
2008
;
22
:
2541
51
.
45
Miething
C
,
Grundler
R
,
Mugler
C
, et al
. 
Retroviral insertional mutagenesis identifies RUNX genes involved in chronic myeloid leukemia disease persistence under imatinib treatment
.
Proc Natl Acad Sci U S A
2007
;
104
:
4594
9
.
46
Baran
Y
,
Salas
A
,
Senkal
CE
, et al
. 
Alterations of ceramide/sphingosine 1-phosphate rheostat involved in the regulation of resistance to imatinib-induced apoptosis in K562 human chronic myeloid leukemia cells
.
J Biol Chem
2007
;
282
:
10922
34
.
47
Theriault
FM
,
Nuthall
HN
,
Dong
ZF
, et al
. 
Role for Runx1 in the proliferation and neuronal differentiation of selected progenitor cells in the mammalian nervous system
.
J Neurosci
2005
;
25
:
2050
61
.
48
Pratap
J
,
Galindo
M
,
Zaidi
SK
, et al
. 
Cell growth regulatory role of Runx2 during proliferative expansion of preosteoblasts
.
Cancer Res
2003
;
63
:
5357
62
.
49
Levanon
D
,
Bettoun
D
,
Harris-Cerruti
C
, et al
. 
The Runx3 transcription factor regulates development and survival of TrkC dorsal root ganglia neurons
.
EMBO J
2002
;
21
:
3454
63
.

Supplementary data