Ceramide is a lipid second messenger derived from the hydrolysis of sphingomyelin by sphingomyelinases (SMases) and implicated in diverse cellular responses, including growth arrest, differentiation, and apoptosis. Defects in the neutral SMase (nSMase) gene Smpd3, the primary regulator of ceramide biosynthesis, are responsible for developmental defects of bone; regulation of ceramide levels have been implicated in macrophage differentiation, but this pathway has not been directly implicated in human cancer. In a genomic screen for gene copy losses contributing to tumorigenesis in a mouse osteosarcoma model, we identified a somatic homozygous deletion specifically targeting Smpd3. Reconstitution of SMPD3 expression in mouse tumor cells lacking the endogenous gene enhanced tumor necrosis factor (TNF)–induced reduction of cell viability. Nucleotide sequencing of the highly conserved SMPD3 gene in a large panel of human cancers revealed mutations in 5 (5%) of 92 acute myeloid leukemias (AMLs) and 8 (6%) of 131 acute lymphoid leukemias (ALLs), but not in other tumor types. In a subset of these mutations, functional analysis indicated defects in protein stability and localization. Taken together, these observations suggest that disruption of the ceramide pathway may contribute to a subset of human leukemias.

Somatic genetic alterations that arise during cancer progression may constitute incidental so-called passenger mutations, or they may be drivers of malignant proliferation, some of which may be appropriate therapeutic targets.1  While large-scale nucleotide sequencing efforts hold the promise of comprehensive analysis of the cancer genome, initial studies have focused on known candidate cancer genes within relatively small numbers of tumors.2-4  To complement such studies, genomic screens for gene copy number alterations have pointed to loci that may harbor recurrent abnormalities.5,6  In fact, the initial identification of many critical tumor suppressor genes has relied on the study of rare tumors with homozygous genomic deletions targeting the relevant locus, identified by classical strategies, as well as genomic screens. These led to the discoveries of the retinoblastoma gene RB, the Wilms tumor suppressors WT1 and WTX, the melanoma susceptibility gene p16/INK4a, the familial breast cancer gene BRCA2, and the tumor suppressor PTEN.7-11 

Homozygous genomic deletions may arise by a number of mechanisms, including the overlap of 2 different allelic losses. The relatively small size of homozygous deletions, compared with much larger regions of single allelic deletions detected by loss of heterozygosity (LOH), reflects such coincidental deletion events, as well as potential selection against the homozygous loss of essential genes that may flank the relevant tumor suppressor gene. Strategies to detect gene copy losses include comparative genomic hybridization (CGH), which have resulted in the identification of novel tumor suppressors,12,13  although the signal-noise ratio in CGH favors detection of gene amplification rather than deletion. A powerful but lower-throughput method designed specifically for identifying homozygous deletions is genomic representational difference analysis (RDA),14  a polymerase chain reaction (PCR)–based subtractive hybridization of genomic tumor DNA from matching normal tissue. Both PTEN11  and BRCA215  deletion loci were first identified using RDA. However, the extreme sensitivity of RDA, which can detect as small as 15 kb (W.J.K. and D.A.H., unpublished data, April, 2003), complicates its application to primary human cancers, since it routinely detects polymorphic deletions, which are now known to be common in the human genome.16  To maximize the success of RDA screening for homozygous deletions associated with tumorigenesis, we adapted this strategy to analyze cell lines derived from syngeneic mouse tumor models.17  Mouse tumors initiated by loss of TP53 constitute a particularly interesting model, since their genomic instability may favor chromosomal events over point mutations during progression of tumorigenesis. As such, any initial discovery of candidate tumor progression genes in this model may provide clues to novel modifiers in a variety of human cancers.17 

Ceramide is a lipid second messenger that triggers signal transduction pathways in response to cytokines or extrinsic cellular stresses, leading to a variety of cellular responses, including growth suppression and apoptosis.18  Ceramide may be synthesized de novo or generated from the hydrolysis of sphingomyelin by sphingomyelinases (SMases). The pleiotrophic effect of ceramide-dependent signaling has been attributed to distinct pools of ceramide generated by different catalytic enzymes at different cellular loci.19  To date, 4 SMases have been identified. SMPD1 encodes lysosomal acidic SMase (aSMase), whose deficiency is associated with the autosomal recessive disorder Niemann-Pick disease.20  A total of 3 neutral SMases (nSMases)—SMPD-2, SMPD-3, and SMPD-4—are localized to different cellular compartments and expressed in different tissues.21-23  SMPD2 is localized to the endoplasmic reticulum (ER) and expressed in all cell types, but the mouse knockout has no phenotype.24  SMPD4 is localized to the ER as well as to the Golgi, and has been shown to be activated in response to tumor necrosis factor α (TNFα), although its physiologic role has yet to be defined.23  In contrast to these 2 nSMase genes, Smpd3-null mice have significant developmental defects, including dwarfism and delayed puberty, attributed to hypothalamic pituitary deficiency,25  and a naturally occurring mouse model of noncollagenous osteogenesis imperfecta (fro/fro), associated with skeletal deformities and fractures, results from inactivation of Smpd3.26 SMPD3 is expressed at highest levels in the brain, is activated by TNFα, and contributes to TNFα-induced apoptosis in cultured cells.27-29 

Here, we identified a gene-specific homozygous deletion of Smpd3 in a mouse TP53-driven osteosarcoma by RDA, and extended this analysis to a panel of human tumors, demonstrating missense mutations of SMPD3 in human leukemias. These observations raise the possibility that the ceramide pathway may be directly implicated in a subset of human cancers.

Cell lines and culture conditions

Mouse osteosarcoma cell lines derived from TP53 heterozygous mice30,31  and MDCK cells were maintained in Dulbecco modified Eagle medium supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin at 37°C in a humidified 5% CO2 incubator. Transient transfection of SMPD3 expression constructs was performed using Lipofectamine 2000 reagent (Invitrogen, Carlsbad, CA) according to the manufacturer's instruction. Retroviral transduction of SMPD3 expression constructs into adherent F4328 cells was modified from a previously described method for suspension culture.32 

RDA and genomic deletion mapping

RDA of F4328 mouse osteosarcoma cell line was carried out as described previously.17  For genomic Southern blot, genomic DNA was digested by BamHI (New England Biolabs, Ipswich, MA), transferred onto nitrocellulose membrane (GE Healthcare, Little Chalfont, United Kingdom) and probed by radiolabeled RDA fragments cloned and excised from the pCR2.1 plasmid (Invitrogen). Primers for genomic PCR were designed from the sequences in the mouse chromosome 8 genomic contig NT 039467 (National Center for Biotechnology Information [NCBI]) and listed in Table S1A (available on the Blood website; see the Supplemental Materials link at the top of the online article).

Cell proliferation and viability assays

For determination of cellular proliferation, cells were plated in 96-well tissue-culture plate, grown, and fixed at appropriate time points by 4% paraformaldehyde in phosphate-buffered saline (PBS). All plates were stained by SYTO60 dye (Molecular Probes, Eugene, OR) simultaneously and quantitated using the Odyssey Imaging System (LI-COR, Lincoln, NE). TNFα-mediated cell viability was measured by MTS assay. In brief, cells in 96-well tissue-culture plates were treated with appropriate concentration of recombinant mouse TNFα (Pierce Endogen, Rockford, IL). After 4 days of incubation, 10 μL MTS solution (Promega, Madison, WI) was added to each well and incubated for 2 hours to develop color. The absorbance was measured spectrophotometrically at a wavelength of 490 nm.

Mutation analysis of SMPD3 in human cancer cell lines and primary tumor samples

All coding exons of SMPD3 were amplified by genomic PCR from a panel of 67 sporadic human cancer cell lines, including lung (NCI-H460, NCI-522, HOP92, NCI-15C2, NCI-1734, NCI-446, NCI-H128, NCI-H596, and NCI-H2009), ovarian (ES-2, IGROV-1, MDAH2774, OV1063, OVCAR3, OVCAR4, OVCAR5, OVCAR8, SKOV3, and SW626), brain (SF-295, SNB-19, U-251, CCF-STTG1, SW-1088, SW1783, T98G, MO59K, SK-N-DZ, and SK-N-MC), leukemia (CCRF-CEM, MOLT-4, SR, K562, and RPMI8226), prostate (DU-145 and PC-3), colon (COLO-205, HCT-116, HCT-15, HT29, and SW-620), renal (786-O, ACHN, CAKI-1, SN-12C, and UO-31), skin (LOX-IMV1, M14, and SKMEL2), bone (SAOS-2), and breast (BT549, BT483, HS157, UACC893, HS467T, HS496T, HS578T, MCF7, MDAMB175, MDAMB231, MDAMB415, MDAMB436, MDAMB 453, MDAMB468, T47D, BR-412, and MCF7ADR) cancers. For mutation screening of SMPD3 from leukemia samples, genomic DNA from 92 primary acute myeloid leukemia (AML; 43 from Massachusetts General Hospital [MGH] and 49 from Inserm), 95 primary acute lymphoblastic leukemia (ALL), and 33 T-cell ALL (T-ALL) cell lines (ALLSIL, BE13, CCRFHSB2, CMLT1, DND41, EOL1, HBPALL, JMJURKAT, K3P, KARPAS45, KE37, KGI, KOPTK1, LOUCY, Mk-B1, MOLT-13, MOLT-15, MOLT-16, MV411, P12ICHIKAWA, PEER, PF382, REH, REX, SKM, SL-W3, SUPT1, SUPT11, SUPT13, SUPT7, TA527, TALL1, and THP; Dana-Farber Cancer Institute, Boston, MA) were collected and analyzed by genomic sequencing. Epstein-Barr virus (EBV)–immortalized lymphoblastoid cell lines established from 170 healthy blood donors provided for normal population controls. PCR primers for exon amplification and nucleotide sequencing are listed in Table S1B.

Expression constructs and site-directed mutagenesis

The coding sequence of human SMPD3 was amplified by PCR from cDNA and cloned into pcDNA6/V5-HIS or pcDNA4/Flag (Invitrogen) or Flag-tag version of MSV-SV-GFP32  plasmids to generate cytomegalovirus (CMV) promoter- or retrovirus-driven expression constructs, respectively. Both V5 and Flag tags were located at the C-terminus of SMPD3. To generate SMPD3 mutant alleles, PCR mutagenesis was used as described.33 

nSMase activity assay

F4328 cells were disrupted by repeated freezing and thawing in lysis buffer (25 mM Tris-Cl [pH 7.4], 5 mM EDTA, 10 mM DTT, and protease inhibitor cocktail [Roche, Indianapolis, IN]). Crude membrane fractions were sedimented by centrifugation for 2 minutes at 2500g and extracted with lysis buffer containing 0.2% Triton X-100. After centrifugation, 10 μg of supernatants were incubated with 20 nM [choline-methyl-14C] sphingomyelin (PerkinElmer, Waltham, MA) for 30 minutes at 37°C in 100 μL of 100 mM Tris-Cl (pH 7.4), 10 mM MgCl2, 0.2% Triton X-100, and 10 mM dithiothreitol. Reactions were terminated by the addition of 1 mL chloroform/methanol (2:1), and the aqueous phase was separated by the addition of 200 μL water. The radioactivity of the upper phase containing phosphorylcholine was measured using a liquid scintillation counter.

Cycloheximde treatment

F4328 cells in a 6-well plate format were transfected with 1 μg V5-tagged wild-type or D358G SMPD3 in pcDNA6. At 48 hours after transfection, cells were treated with 100 μg/mL cycloheximide for various times, and total cell lysates were prepared for Western blot analysis.

Indirect immunofluorescence microscopy

MDCK cells grown on coverslips were transiently transfected with either Flag- or V5-tagged SMPD3 or mutant SMPD3 in pcDNA vector using Lipofectamine 2000 (Invitrogen). After 48 hours, cells were fixed with 4% paraformaldehyde in PBS for 10 minutes, permeabilized with 0.5% Triton X-100 in PBS for 10 minutes, and blocked with blocking solution (5% goat serum, 5% horse serum, 0.2% fish gelatin, and 0.2% Tween 20 in PBS) for 30 minutes at 37°C. Cells were then incubated with anti-V5 (1:250) or anti-Flag (1:250) monoclonal antibodies in blocking solution for 1 hour at 37°C, washed, probed with anti-mouse Alexa 488 (1:500; Invitrogen), washed and mounted with DAPI. A Nikon Eclipse 90i fluorescence microscope (Nikon, Melville, NY) was used for visualization of staining. NIS-Element AR 2.30 software (Nikon) was used for the acquisition of images.

Antibodies

Anti-V5, anti-Flag, and antitubulin antibodies were purchased from Invitrogen, Sigma-Aldrich (St Louis, MO), and Santa Cruz Biotechnology (Santa Cruz, CA), respectively.

Homozygous deletion of Smpd3 in a mouse osteosarcoma initiated by loss of TP53

We undertook an RDA screen of 3 osteosarcoma cell lines derived from mouse tumors with a heterozygous germ-line TP53 deletion.30  No homozygous deletions were identified in 2 cell lines, while the third (F4328) harbored homozygous deletions at a number of loci, including the known tumor suppressor gene PTEN. Of 4 deletion loci that did not harbor a known tumor suppressor, we selected a small homozygous deletion on chromosome 8q for detailed analysis. The chromosome 8q deletion produced 4 nonredundant RDA fragments (Figure 1A), which were used to confirm the homozygous deletion by genomic Southern blot (Figure 1B). Based on the sequence of mouse chromosome 8 genomic contig NT 039467 (NCBI), we mapped the boundary of the deletion by genomic PCR, defining a deletion of 100 kb. This gene-poor region contains the first 2 5′ exons of the Smpd3 gene, which is localized to the centromeric border of the deletion. The coding exons of the neighboring gene Zfp90, which are localized at the telomeric border of the deletion, are not affected. However, since the deletion affects sequences upstream of this gene, we confirmed expression of Zfp90 in F4328 cells by reverse transcription (RT)–PCR. In contrast, Smpd3 expression is absent (Figure 1C).

Figure 1

RDA isolation of homozygous genomic deletion at the Smpd3 locus in F4328 mouse osteosarcoma cells. (A) Schematic representation of the homozygous deletion of the mouse chromosome 8 locus in F4328. A total of 4 nonredundant RDA clones (-1, -5, -14, and -22 [arrow])—were found in the intergenic region between Zfp90 and Smpd3. Genomic PCR analysis of the first exon of Zfp90 (1), intergenic region (2), the first (3) and the second (3) exons, and the second intron (4 and 5) of Smpd3 in the F4328 and unrelated control F3733 and F4711 cell lines are shown. (B) Mouse genomic DNAs from F4328 and control F3081, F3678, and F3587 cell lines were digested by BamHI and subjected to Southern blot analysis using RDA clone-1 or -14 as probes. A DNA fragment that was isolated by RDA due to a BglII polymorphism served as a probe for the loading control. (C) Expression of Smpd3 and Zfp90 genes in the F4328 cell line was analyzed by RT-PCR analysis. Primers were chosen in exons 6 and 10 for Smpd3, which are outside the genomic deletion. Gapdh was used as an internal loading control. (D,E) Reconstitution of F4328 cell lines with SMPD3. SMPD3 (□) or empty vector control (■) constructs were retrovirally transduced into F4328 cells. (D) The resulting cells were plated in 96-well format for measurement of growth rate. Cell numbers were measured by the absorbance at 600 nm after staining with DNA staining dye CYTO60 and plotted as fold increase compared with day 1. Error bar indicates standard error of mean. Protein expression of SMPD3, detected by anti-Flag antibody, is shown in the inset. (E) Cells in 96-well plate format were treated with the indicated concentration of TNFα for 4 days, and cell viability was measured by MTS assay. Percentage of viability is plotted compared with the mock-treated cells. Statistical significance of each matching data point was calculated (*P < .001; **P > .05).

Figure 1

RDA isolation of homozygous genomic deletion at the Smpd3 locus in F4328 mouse osteosarcoma cells. (A) Schematic representation of the homozygous deletion of the mouse chromosome 8 locus in F4328. A total of 4 nonredundant RDA clones (-1, -5, -14, and -22 [arrow])—were found in the intergenic region between Zfp90 and Smpd3. Genomic PCR analysis of the first exon of Zfp90 (1), intergenic region (2), the first (3) and the second (3) exons, and the second intron (4 and 5) of Smpd3 in the F4328 and unrelated control F3733 and F4711 cell lines are shown. (B) Mouse genomic DNAs from F4328 and control F3081, F3678, and F3587 cell lines were digested by BamHI and subjected to Southern blot analysis using RDA clone-1 or -14 as probes. A DNA fragment that was isolated by RDA due to a BglII polymorphism served as a probe for the loading control. (C) Expression of Smpd3 and Zfp90 genes in the F4328 cell line was analyzed by RT-PCR analysis. Primers were chosen in exons 6 and 10 for Smpd3, which are outside the genomic deletion. Gapdh was used as an internal loading control. (D,E) Reconstitution of F4328 cell lines with SMPD3. SMPD3 (□) or empty vector control (■) constructs were retrovirally transduced into F4328 cells. (D) The resulting cells were plated in 96-well format for measurement of growth rate. Cell numbers were measured by the absorbance at 600 nm after staining with DNA staining dye CYTO60 and plotted as fold increase compared with day 1. Error bar indicates standard error of mean. Protein expression of SMPD3, detected by anti-Flag antibody, is shown in the inset. (E) Cells in 96-well plate format were treated with the indicated concentration of TNFα for 4 days, and cell viability was measured by MTS assay. Percentage of viability is plotted compared with the mock-treated cells. Statistical significance of each matching data point was calculated (*P < .001; **P > .05).

Close modal

To test for functional consequences of the Smpd3 deletion in F4328 cells, we reconstituted human SMPD3 expression in these cells using retroviral infection with a bicistronic green fluorescent protein (GFP)–linked vector, allowing monitoring of infected cells by flow cytometry. The more than 80% infection rate allowed analysis of pooled infected F4328 cells, avoiding clonal selection bias. SMPD3-reconstituted cells displayed a modest suppression of proliferation (Figure 1D) and showed no obvious difference in contact inhibition-induced growth arrest (data not shown). However, cell viability following treatment with TNFα was significantly decreased in SMPD3-reconstituted cells compared with parental F4328 cells lacking the endogenous gene (Figure 1E). While the functional consequences of restoring SMPD3 expression and ceramide biogenesis in cancer cells with a deletion of the endogenous gene may be complex, TNFα-induced reduction of cell viability provides an initial measure of ceramide signaling.

SMPD3 mutations in human leukemias

To determine whether SMPD3 is also targeted by mutations in human cancers, we first screened a cohort of 67 sporadic human cancer cell lines from various tissue origins, including lung, ovary, brain, leukemia, prostate, colon, renal, skin, bone, and breast by bidrectional nucleotide sequencing. A total of 2 missense mutations in SMPD3 were identified in the leukemia cell lines CCRF-CEM (ALL) and MOLT-4 (ALL). We therefore expanded our analysis to a panel of 92 cases of primary AML, 95 cases of primary ALL samples, and 33 ALL cell lines, identifying SMPD3 mutations in an additional 11 cases. Thus, in total, SMPD3 mutations were present in 8 (6%) of 131 ALLs and 5 (5%) of 92 AMLs (Table 1). All mutations led to nonsynonymous missense changes. Of 7 cell lines with SMPD3 mutations, 4 are heterozygous, and 2 are compound heterozygotes. One cell line has 3 missense mutations including one homozygous mutation (E499G). All primary leukemia specimens displayed both mutant and wild-type sequences (Figure S1), presumably due to normal cell contamination. A total of 2 SMPD3 mutations were recurrent: D194N was present in 5 independent cases and D358G was found in 3 cases. Although remission samples from leukemia cases were not available for analysis, all mutations identified in these leukemias were absent from 170 healthy population controls (340 alleles), and from the NCBI single nucleotide polymorphism database.34  Amino acid alignment of SMPD3 from various vertebrate animals showed that of the 11 mutations, 9 mutations occurred in amino acids that are conserved in all vertebrate animals for which SMPD3 sequence is known, while 5 of them are conserved in mammals (Figure 2). Thus, most of the SMPD3 mutations in human leukemias affect highly conserved amino acids. The overall amino acid identity in SMPD3 from these species compared with human is 91% (murine), 89% (bovis and canine), 72% (chicken), 69% (xenopus), and 56% (zebrafish). In addition, computational prediction of potential impact of an amino acid substitution on the structure and function of a human protein using the Polyphen program (http://genetics.bwh.harvard.edu/pph) resulted in 4 benign, 3 possibly damaging, and 4 probably damaging mutations that are relatively well correlated with their conservation among species (Table S2).

Table 1

SMPD3 mutations within human leukemia

HistologyNucleotide changeAmino acid changeFrequency in controls
AML primary    
    AML1 c.580G>A Asp194Asn 0/166 
    AMLB5 c.580G>A Asp194Asn 0/166 
    AMLB6 c.580G>A Asp194Asn 0/166 
    LEUK139 c.580G>A Asp194Asn 0/166 
    LEUK7 c.1519G>A Val507Ile 0/172 
ALL primary, ALL. 88-89 c.580G>A Asp194Asn 0/166 
ALL cell line    
    KE37 c.127T>C Tyr43His 0/171 
     c.1073A>G Asp358Gly 0/176 
    Mk-B1 c.284A>T Tyr95Phe 0/171 
     c.1073A>G Asp358Gly 0/176 
     c.1495G>C Glu499Gly 0/174 
    JMJURKAT c.151G>A Asp51Asn 0/171 
     c.742G>A Gly248Ser 0/166 
    SUPT1 c.584G>A Gly195Glu 0/166 
    MOLT-4 c.736C>T Arg246Cys 0/166 
    SUPT11 c.1006C>T Arg336Cys 0/166 
    CCRF-CEM c.1073A>G Asp358Gly 0/176 
Polymorphisms    
    Ovarian cancer    
        MDAH2774 c.1850G>T Cys617Tyr 3/43 
    Renal cancer    
        786-O c.1850G>T Cys617Tyr 3/43 
    Myeloma    
        RPMI8226 c.391C>G Val131Leu 0/171 
HistologyNucleotide changeAmino acid changeFrequency in controls
AML primary    
    AML1 c.580G>A Asp194Asn 0/166 
    AMLB5 c.580G>A Asp194Asn 0/166 
    AMLB6 c.580G>A Asp194Asn 0/166 
    LEUK139 c.580G>A Asp194Asn 0/166 
    LEUK7 c.1519G>A Val507Ile 0/172 
ALL primary, ALL. 88-89 c.580G>A Asp194Asn 0/166 
ALL cell line    
    KE37 c.127T>C Tyr43His 0/171 
     c.1073A>G Asp358Gly 0/176 
    Mk-B1 c.284A>T Tyr95Phe 0/171 
     c.1073A>G Asp358Gly 0/176 
     c.1495G>C Glu499Gly 0/174 
    JMJURKAT c.151G>A Asp51Asn 0/171 
     c.742G>A Gly248Ser 0/166 
    SUPT1 c.584G>A Gly195Glu 0/166 
    MOLT-4 c.736C>T Arg246Cys 0/166 
    SUPT11 c.1006C>T Arg336Cys 0/166 
    CCRF-CEM c.1073A>G Asp358Gly 0/176 
Polymorphisms    
    Ovarian cancer    
        MDAH2774 c.1850G>T Cys617Tyr 3/43 
    Renal cancer    
        786-O c.1850G>T Cys617Tyr 3/43 
    Myeloma    
        RPMI8226 c.391C>G Val131Leu 0/171 
Figure 2

Amino acid alignment of SMPD3 from various species surrounding SMPD3 mutations. The SMPD3 amino acid sequence from mammals (Homo sapiens, Mus musculus, Bos taurus, and Canis familiaris) and other vertebrate animals (Gallus gallus, Xenopus laevis, and Danio rerio) are aligned using MultAlin software (http://bioinfo.genopole-toulouse.prd.fr/multalin/multalin.html). Blue and red letters indicate amino acids that are conserved within mammals and vertebrates, respectively.

Figure 2

Amino acid alignment of SMPD3 from various species surrounding SMPD3 mutations. The SMPD3 amino acid sequence from mammals (Homo sapiens, Mus musculus, Bos taurus, and Canis familiaris) and other vertebrate animals (Gallus gallus, Xenopus laevis, and Danio rerio) are aligned using MultAlin software (http://bioinfo.genopole-toulouse.prd.fr/multalin/multalin.html). Blue and red letters indicate amino acids that are conserved within mammals and vertebrates, respectively.

Close modal

To study the effects of leukemia-associated mutations, we generated retroviral expression constructs of wild-type SMPD3 and the mutant alleles. Following infection of F4328 cells, nSMase activity of cellular lysates was quantified by measuring hydrolysis of sphingomyelin into phosphorylcholine and ceramide. nSMase activity was minimal in parental F4328 cells, indicating that SMPD3 is the primary contributor to sphingomyelin hydrolysis activity in these cells. As expected, cells infected with wild-type construct had increased nSMase activity that was appropriately restricted to the crude membrane fraction, with minimal enzymatic activity in cytosolic fractions. Cells infected with the various SMPD3 mutants showed no gross defect in nSMase activity with the exception of the recurrent mutation D358G, which showed no increase in enzymatic activity compared with the empty vector control. (Figure 3A). While similar infection rates were observed for each construct by flow cytometry, Western blot analysis showed strikingly decreased protein expression of D358G SMPD3. The consistently low levels of D358G mutant expression raised the possibility of an inherently unstable protein. Indeed, transient transfection of a CMV-driven construct into F4328 cells showed comparable levels of RNA expression for wild-type and the D358G mutant, but a marked reduction in D358G protein expression (Figure 3B,C). To determine whether the D358G mutation leads to SMPD3 protein instability, we compared the half-life of wild-type and mutant proteins by measuring protein levels following treatment with cycloheximide to block new protein synthesis. Whereas protein turnover, as measured by cycloheximide/Western blot analysis, was undetectable for wild-type SMPD3 (t1/2 over 12 hours), the half-life of the D358G mutant was as short as 8 hours (Figure 3D,E). Thus, the recurrent D358G mutation appears to encode a grossly unstable SMPD3 protein.

Figure 3

The D358G mutation in SMPD3 causes protein instability. (A) nSMase assay for wild-type and D358G SMPD3 lysate. Crude membrane fractions from F4328 cells expressing retrovirally transduced wild-type or D358G SMPD3 were incubated with 14C-labeled sphingomyelin in the presence of the Mg2+ for 30 minutes at 37°C. The release of 14C-phosphorylcholine in the aqueous phase was measured by scintillation counter. Error bars indicate standard error of the mean. (B,C) F4328 cells were transiently transfected with empty vector, V5-tagged wild-type, or D358G SMPD3. Cells were harvested 48 hours after transfection and were split for Northern (B) and Western blot analysis (C). (D) F4328 cells transiently transfected with either V5-tagged wild-type or D358G SMPD3 were treated with 100 μg/mL cycloheximide for the indicated times, and the total cell lysates were prepared for Western blot by anti-V5 antibody. The middle panel shows a longer exposure of the same blot. (E) Western blot results were quantitated by QuantityOne software (Bio-Rad, Hercules, CA) and protein stability was plotted by percentage of the intensity of band compared with that of the untreated sample. □ indicates wild-type SMPD3; ■, D358G SMPD3.

Figure 3

The D358G mutation in SMPD3 causes protein instability. (A) nSMase assay for wild-type and D358G SMPD3 lysate. Crude membrane fractions from F4328 cells expressing retrovirally transduced wild-type or D358G SMPD3 were incubated with 14C-labeled sphingomyelin in the presence of the Mg2+ for 30 minutes at 37°C. The release of 14C-phosphorylcholine in the aqueous phase was measured by scintillation counter. Error bars indicate standard error of the mean. (B,C) F4328 cells were transiently transfected with empty vector, V5-tagged wild-type, or D358G SMPD3. Cells were harvested 48 hours after transfection and were split for Northern (B) and Western blot analysis (C). (D) F4328 cells transiently transfected with either V5-tagged wild-type or D358G SMPD3 were treated with 100 μg/mL cycloheximide for the indicated times, and the total cell lysates were prepared for Western blot by anti-V5 antibody. The middle panel shows a longer exposure of the same blot. (E) Western blot results were quantitated by QuantityOne software (Bio-Rad, Hercules, CA) and protein stability was plotted by percentage of the intensity of band compared with that of the untreated sample. □ indicates wild-type SMPD3; ■, D358G SMPD3.

Close modal

Biochemical activity for nSMase has been observed primarily in the plasma membrane, and relocalization of SMPD3 from Golgi to the plasma membrane has been observed upon its activation by contact inhibition in MCF7 cells,27  by oxidative stress in lung epithelial cells,35  and at baseline in Hep-G2 cells.36  To further analyze functional defects of SMPD3 mutants, we determined the subcellular localization of SMPD3 and each mutant form in the well-characterized MDCK kidney epithelial cell line. SMPD3 was localized to the plasma membrane in confluent MDCK cells (Figure 4D). Of the 11 mutants tested, 2 SMPD3 mutants, D358G and G248S, failed to localize to the plasma membrane under these conditions, showing diffuse staining throughout the cytoplasm (Figure 4G,J).

Figure 4

Subcellular localization of wild-type and mutant SMPD3 in MDCK cells. Tagged version of wild-type and mutant SMPD3 (G247S and D358G) were transfected, and their subcellular localization was determined by indirect fluorescence microscopy. More than 70% of stained cells within each sample showed similar staining as depicted in the panels. Magnification: 20×/0.75 NA Plan Apo objective of samples in Vectashield mounting medium for dluorescence with DAPI, H-1200 (Vector Laboratories, Burlingame, CA) stained with Alexa Fluor 488 goat antimouse IgG H+L (Invitrogen). Images were acquired using a Retiga 200R digital camera (QImaging, Surrey, BC) and HIS-Elements Ar 2.30 software (Nikon) and processed using Adobe Photoshop 7.0 (Adobe Systems, San Jose, CA).

Figure 4

Subcellular localization of wild-type and mutant SMPD3 in MDCK cells. Tagged version of wild-type and mutant SMPD3 (G247S and D358G) were transfected, and their subcellular localization was determined by indirect fluorescence microscopy. More than 70% of stained cells within each sample showed similar staining as depicted in the panels. Magnification: 20×/0.75 NA Plan Apo objective of samples in Vectashield mounting medium for dluorescence with DAPI, H-1200 (Vector Laboratories, Burlingame, CA) stained with Alexa Fluor 488 goat antimouse IgG H+L (Invitrogen). Images were acquired using a Retiga 200R digital camera (QImaging, Surrey, BC) and HIS-Elements Ar 2.30 software (Nikon) and processed using Adobe Photoshop 7.0 (Adobe Systems, San Jose, CA).

Close modal

Concluding remarks

We have demonstrated that a subset of human leukemias have mutations in the nSMase gene SMPD3, which encode the major catalytic enzyme responsible for the generation of ceramide in response to various cellular stresses. Of the 11 mutations identified, 2 mutations showed gross defects in either protein stability and/or subcellular localization. The functional significance of the other 9 mutations is uncertain, and may require more physiologic analyses of hematopoietic differentiation. Matched normal tissues or remission specimens are not available for analysis, a common problem in mutational studies in leukemia, and we therefore cannot exclude the possibility that at least some of the other SMPD3 variants represent rare germ-line mutations. However, their identification as likely somatic mutations is supported by their absence in large number of population controls, the absence of SMPD3 mutations in other tumor types analyzed, and the fact that most mutations affect highly conserved residues across a different evolutionary species. Distinguishing true “driver” versus “passenger” mutations in human cancers may require detailed functional studies, as recently demonstrated in an analysis of FLT3 mutations in leukemia.37  Consistent with that study, the presence of clearly defective SMPD3 alleles in a subset of leukemias supports that other SMPD3 mutations may have more moderate effects that are less readily defined in vitro. Together, these observations point to the ceramide pathway as being targeted by mutations in human leukemias.

Ceramide has been proposed as an antioncogenic lipid second messenger in multiple cancer types, where decreased levels of ceramide or increased levels of the counterbalancing sphingolipid sphingosine-1-phosphate or its metabolic enzyme sphingosine kinase 1 have been observed in cancers (reviewed in Ogretmen and Hannun38 ). However, genetic evidence supporting a specific role of the ceramide pathway in human cancer has been lacking. It is of interest that the sphingomyelin cycle was first discovered in HL-60 human myelocytic leukemia cells, where induction of differentiation along the monocytic lineage using vitamin D3, TNFα, and γ-interferon, but not along other lineages, activates cellular nSMase, triggering sphingomyelin hydrolysis and ceramide accumulation.19,39-41  Thus, ceramide metabolism may play a particularly important role in hematopoetic differentiation, and its dysregulation may contribute to a subset of human leukemias. In addition to the point mutations observed here, we note the chromosomal localization of SMPD3 on chromosome band 16q22.1, a locus rich in leukemia-associated chromosomal abnormalities.42-44  Taken together, the finding that a significant subset of human leukemias harbors mutations in SMPD3 provides the first genetic evidence for alterations in ceramide metabolism in human cancers.

The online version of this article contains a data supplement.

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 are grateful to Drs D. Gary Gilliland, Vijay Yajnik, Charles Paulding, and Ryn Miake-Lye for helpful discussions, reagents, and critical reading.

This work was supported by grants from the National Foundation for Cancer Research (D.A.H.), the Doris Duke Distinguished Clinical Scientist Award (D.A.H.), and the National Insitutes of Health (CA68484 and CA109901 to A.T.L.)

National Institutes of Health

Contribution: W.J.K., R.A.O., S.M.H., L.E.P., M.G., and F.D. performed experiments; A.I.M. provided mouse osteosarcoma cell lines; O.A.B., and A.T.L. provided leukemia DNA samples; W.J.K., D.A.S., D.W.B., D.T.S., and D.A.H analyzed data; W.J.K and D.A.H designed the research and wrote the paper.

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

Correspondence: Daniel A. Haber, Cancer Center, Massachusetts General Hospital, 149 13th St, Charlestown, MA 02129; e-mail: haber@helix.mgh.harvard.edu.

1
Haber
 
DA
Settleman
 
J
Cancer: drivers and passengers.
Nature
2007
, vol. 
446
 (pg. 
145
-
146
)
2
Bardelli
 
A
Parsons
 
DW
Silliman
 
N
, et al. 
Mutational analysis of the tyrosine kinome in colorectal cancers.
Science
2003
, vol. 
300
 pg. 
949
 
3
Greenman
 
C
Stephens
 
P
Smith
 
R
, et al. 
Patterns of somatic mutation in human cancer genomes.
Nature
2007
, vol. 
446
 (pg. 
153
-
158
)
4
Sjoblom
 
T
Jones
 
S
Wood
 
LD
, et al. 
The consensus coding sequences of human breast and colorectal cancers.
Science
2006
, vol. 
314
 (pg. 
268
-
274
)
5
Maser
 
RS
Choudhury
 
B
Campbell
 
PJ
, et al. 
Chromosomally unstable mouse tumours have genomic alterations similar to diverse human cancers.
Nature
2007
, vol. 
447
 (pg. 
966
-
971
)
6
Zender
 
L
Spector
 
MS
Xue
 
W
, et al. 
Identification and validation of oncogenes in liver cancer using an integrative oncogenomic approach.
Cell
2006
, vol. 
125
 (pg. 
1253
-
1267
)
7
Lee
 
WH
Bookstein
 
R
Hong
 
F
Young
 
LJ
Shew
 
JY
Lee
 
EY
Human retinoblastoma susceptibility gene: cloning, identification, and sequence.
Science
1987
, vol. 
235
 (pg. 
1394
-
1399
)
8
Call
 
KM
Glaser
 
T
Ito
 
CY
, et al. 
Isolation and characterization of a zinc finger polypeptide gene at the human chromosome 11 Wilms' tumor locus.
Cell
1990
, vol. 
60
 (pg. 
509
-
520
)
9
Nobori
 
T
Miura
 
K
Wu
 
DJ
Lois
 
A
Takabayashi
 
K
Carson
 
DA
Deletions of the cyclin-dependent kinase-4 inhibitor gene in multiple human cancers.
Nature
1994
, vol. 
368
 (pg. 
753
-
756
)
10
Wooster
 
R
Bignell
 
G
Lancaster
 
J
, et al. 
Identification of the breast cancer susceptibility gene BRCA2.
Nature
1995
, vol. 
378
 (pg. 
789
-
792
)
11
Li
 
J
Yen
 
C
Liaw
 
D
, et al. 
PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer.
Science
1997
, vol. 
275
 (pg. 
1943
-
1947
)
12
Rivera
 
MN
Kim
 
WJ
Wells
 
J
, et al. 
An X chromosome gene, WTX, is commonly inactivated in Wilms tumor.
Science
2007
, vol. 
315
 (pg. 
642
-
645
)
13
Mullighan
 
CG
Goorha
 
S
Radtke
 
I
Downing
 
JR
, et al. 
Genome-wide analysis of genetic alterations in acute lymphoblastic leukaemia.
Nature
2007
, vol. 
446
 (pg. 
758
-
764
)
14
Lisitsyn
 
N
Wigler
 
M
Cloning the differences between two complex genomes.
Science
1993
, vol. 
259
 (pg. 
946
-
951
)
15
Schutte
 
M
da Costa
 
LT
Hahn
 
SA
, et al. 
Identification by representational difference analysis of a homozygous deletion in pancreatic carcinoma that lies within the BRCA2 region.
Proc Natl Acad Sci U S A
1995
, vol. 
92
 (pg. 
5950
-
5954
)
16
Iafrate
 
AJ
Feuk
 
L
Rivera
 
MN
, et al. 
Detection of large-scale variation in the human genome.
Nat Genet
2004
, vol. 
36
 (pg. 
949
-
951
)
17
Yajnik
 
V
Paulding
 
C
Sordella
 
R
, et al. 
DOCK4, a GTPase activator, is disrupted during tumorigenesis.
Cell
2003
, vol. 
112
 (pg. 
673
-
684
)
18
Hannun
 
YA
Functions of ceramide in coordinating cellular responses to stress.
Science
1996
, vol. 
274
 (pg. 
1855
-
1859
)
19
Levade
 
T
Jaffrezou
 
JP
Signalling sphingomyelinases: which, where, how and why?
Biochim Biophys Acta
1999
, vol. 
1438
 (pg. 
1
-
17
)
20
Schuchman
 
EH
Suchi
 
M
Takahashi
 
T
Sandhoff
 
K
Desnick
 
RJ
Human acid sphingomyelinase: isolation, nucleotide sequence and expression of the full-length and alternatively spliced cDNAs.
J Biol Chem
1991
, vol. 
266
 (pg. 
8531
-
8539
)
21
Tomiuk
 
S
Hofmann
 
K
Nix
 
M
Zumbansen
 
M
Stoffel
 
W
Cloned mammalian neutral sphingomyelinase: functions in sphingolipid signaling?
Proc Natl Acad Sci U S A
1998
, vol. 
95
 (pg. 
3638
-
3643
)
22
Hofmann
 
K
Tomiuk
 
S
Wolff
 
G
Stoffel
 
W
Cloning and characterization of the mammalian brain-specific, Mg2+-dependent neutral sphingomyelinase.
Proc Natl Acad Sci U S A
2000
, vol. 
97
 (pg. 
5895
-
5900
)
23
Krut
 
O
Wiegmann
 
K
Kashkar
 
H
Yazdanpanah
 
B
Kronke
 
M
Novel tumor necrosis factor-responsive mammalian neutral sphingomyelinase-3 is a C-tail-anchored protein.
J Biol Chem
2006
, vol. 
281
 (pg. 
13784
-
13793
)
24
Zumbansen
 
M
Stoffel
 
W
Neutral sphingomyelinase 1 deficiency in the mouse causes no lipid storage disease.
Mol Cell Biol
2002
, vol. 
22
 (pg. 
3633
-
3638
)
25
Stoffel
 
W
Jenke
 
B
Block
 
B
Zumbansen
 
M
Koebke
 
J
Neutral sphingomyelinase 2 (smpd3) in the control of postnatal growth and development.
Proc Natl Acad Sci U S A
2005
, vol. 
102
 (pg. 
4554
-
4559
)
26
Aubin
 
I
Adams
 
CP
Opsahl
 
S
, et al. 
A deletion in the gene encoding sphingomyelin phosphodiesterase 3 (Smpd3) results in osteogenesis and dentinogenesis imperfecta in the mouse.
Nat Genet
2005
, vol. 
37
 (pg. 
803
-
805
)
27
Marchesini
 
N
Osta
 
W
Bielawski
 
J
Luberto
 
C
Obeid
 
LM
Hannun
 
YA
Role for mammalian neutral sphingomyelinase 2 in confluence-induced growth arrest of MCF7 cells.
J Biol Chem
2004
, vol. 
279
 (pg. 
25101
-
25111
)
28
Luberto
 
C
Hassler
 
DF
Signorelli
 
P
, et al. 
Inhibition of tumor necrosis factor-induced cell death in MCF7 by a novel inhibitor of neutral sphingomyelinase.
J Biol Chem
2002
, vol. 
277
 (pg. 
41128
-
41139
)
29
Clarke
 
CJ
Truong
 
TG
Hannun
 
YA
Role for neutral sphingomyelinase-2 in tumor necrosis factor alpha-stimulated expression of vascular cell adhesion molecule-1 (VCAM) and intercellular adhesion molecule-1 (ICAM) in lung epithelial cells: p38 MAPK is an upstream regulator of nSMase2.
J Biol Chem
2007
, vol. 
282
 (pg. 
1384
-
1396
)
30
Jacks
 
T
Remington
 
L
Williams
 
BO
, et al. 
Tumor spectrum analysis in p53-mutant mice.
Curr Biol
1994
, vol. 
4
 (pg. 
1
-
7
)
31
McClatchey
 
AI
Saotome
 
I
Mercer
 
K
, et al. 
Mice heterozygous for a mutation at the Nf2 tumor suppressor locus develop a range of highly metastatic tumors.
Genes Dev
1998
, vol. 
12
 (pg. 
1121
-
1133
)
32
Ellisen
 
LW
Carlesso
 
N
Cheng
 
T
Scadden
 
DT
Haber
 
DA
The Wilms tumor suppressor WT1 directs stage-specific quiescence and differentiation of human hematopoietic progenitor cells.
EMBO J
2001
, vol. 
20
 (pg. 
1897
-
1909
)
33
Higuchi
 
R
Krummel
 
B
Saiki
 
RK
A general method of in vitro preparation and specific mutagenesis of DNA fragments: study of protein and DNA interactions.
Nucleic Acids Res
1988
, vol. 
16
 (pg. 
7351
-
7367
)
34
National Center for Biotechnology Information
Single Polymorphism Nucleotide database
Accessed January 22, 2008 
35
Levy
 
M
Castillo
 
SS
Goldkorn
 
T
nSMase2 activation and trafficking are modulated by oxidative stress to induce apoptosis.
Biochem Biophys Res Commun
2006
, vol. 
344
 (pg. 
900
-
905
)
36
Karakashian
 
AA
Giltiay
 
NV
Smith
 
GM
Nikolova-Karakashian
 
MN
Expression of neutral sphingomyelinase-2 (NSMase-2) in primary rat hepatocytes modulates IL-beta-induced JNK activation.
FASEB J
2004
, vol. 
18
 (pg. 
968
-
970
)
37
Frohling
 
S
Scholl
 
C
Levine
 
RL
, et al. 
Identification of driver and passenger mutations of FLT3 by high-throughput DNA sequence analysis and functional assessment of candidate alleles.
Cancer Cell
2007
, vol. 
12
 (pg. 
501
-
513
)
38
Ogretmen
 
B
Hannun
 
YA
Biologically active sphingolipids in cancer pathogenesis and treatment.
Nat Rev Cancer
2004
, vol. 
4
 (pg. 
604
-
616
)
39
Kim
 
MY
Linardic
 
C
Obeid
 
L
Hannun
 
Y
Identification of sphingomyelin turnover as an effector mechanism for the action of tumor necrosis factor alpha and gamma-interferon: specific role in cell differentiation.
J Biol Chem
1991
, vol. 
266
 (pg. 
484
-
489
)
40
Okazaki
 
T
Bell
 
RM
Hannun
 
YA
Sphingomyelin turnover induced by vitamin D3 in HL-60 cells. Role in cell differentiation.
J Biol Chem
1989
, vol. 
264
 (pg. 
19076
-
19080
)
41
Liu
 
B
Obeid
 
LM
Hannun
 
YA
Sphingomyelinases in cell regulation.
Semin Cell Dev Biol
1997
, vol. 
8
 (pg. 
311
-
322
)
42
Betts
 
DR
Rohatiner
 
AZ
Evans
 
ML
Rassam
 
SM
Lister
 
TA
Gibbons
 
B
Abnormalities of chromosome 16q in myeloid malignancy: 14 new cases and a review of the literature.
Leukemia
1992
, vol. 
6
 (pg. 
1250
-
1256
)
43
Marlton
 
P
Keating
 
M
Kantarjian
 
H
, et al. 
Cytogenetic and clinical correlates in AML patients with abnormalities of chromosome 16.
Leukemia
1995
, vol. 
9
 (pg. 
965
-
971
)
44
Hogge
 
DE
Misawa
 
S
Parsa
 
NZ
Pollak
 
A
Testa
 
JR
Abnormalities of chromosome 16 in association with acute myelomonocytic leukemia and dysplastic bone marrow eosinophils.
J Clin Oncol
1984
, vol. 
2
 (pg. 
550
-
557
)
Sign in via your Institution