Journal list menu

Volume 3, Issue 5-6 p. 425-438
Paper
Open Access

Characterizing DNA methylation patterns in pancreatic cancer genome

Aik Choon Tan

Aik Choon Tan

The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, MD, USA

Search for more papers by this author
Antonio Jimeno

Antonio Jimeno

The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, MD, USA

Search for more papers by this author
Steven H. Lin

Steven H. Lin

Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA

Search for more papers by this author
Jenna Wheelhouse

Jenna Wheelhouse

The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, MD, USA

Search for more papers by this author
Fonda Chan

Fonda Chan

The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, MD, USA

Search for more papers by this author
Anna Solomon

Anna Solomon

The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, MD, USA

Search for more papers by this author
N.V. Rajeshkumar

N.V. Rajeshkumar

The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, MD, USA

Search for more papers by this author
Belen Rubio-Viqueira

Belen Rubio-Viqueira

Centro Integral Oncologico “Clara Campal”, Calle Oña, 10, 28050 Madrid, Spain

Search for more papers by this author
Manuel Hidalgo

Corresponding Author

Manuel Hidalgo

The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, MD, USA

Centro Integral Oncologico “Clara Campal”, Calle Oña, 10, 28050 Madrid, Spain

Correspondence to: Manuel Hidalgo, MD, PhD, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, 1650 Orleans St., Room 489, Baltimore, MD 21231, USA. Tel.: +1 410 5029746; fax: +1 410 6149006.Search for more papers by this author
First published: 22 April 2009
Citations: 118

Abstract

We performed a global methylation profiling assay on 1505 CpG sites across 807 genes to characterize DNA methylation patterns in pancreatic cancer genome. We found 289 CpG sites that were differentially methylated in normal pancreas, pancreatic tumors and cancer cell lines. We identified 23 and 35 candidate genes that are regulated by hypermethylation and hypomethylation in pancreatic cancer, respectively. We also identified candidate methylation markers that alter the expression of genes critical to gemcitabine susceptibility in pancreatic cancer. These results indicate that aberrant DNA methylation is a frequent epigenetic event in pancreatic cancer; and by using global methylation profiling assay, it is possible to identify these markers for diagnostic and therapeutic purposes in this disease.

1 Introduction

Cancer is a complex disease characterized by multiple genetic and epigenetic genomic alterations (Esteller, 2007; Feinberg et al., 2006; Jones and Baylin, 2002). DNA methylation is one of the most important epigenetic alterations and plays a critical functional role in development, differentiation and diseases (Jones and Baylin, 2002). Through the activity of DNA methyltransferases (DNMTs), DNA methylation occurs at the cytosine residue in the context of 5′-CG-3′ (CpG dinucleotide) across human genome (Laird, 2003). During the developmental process, DNA methylation plays an essential role in X chromosome inactivation in female somatic cells and in the mono-allelic silencing of parentally imprinted genes (Herman and Baylin, 2003). Once these DNA methylation patterns are acquired in the early embryo stage, these patterns are inherited and maintained in successive cell generations. Promoter regions are usually enriched with CpG dinucleotides, known as CpG islands; and hypermethylation of these islands correlates with transcriptional silencing of tumor suppressor genes (Herman and Baylin, 2003). Conversely, increased expressions of oncogenes were associated with hypomethylation (Cui et al., 2002). This hypomethylation is known to contribute to cancer cell phenotypes through loss of imprinting (LOI) and genomic instability that characterizes tumors (Cui et al., 2002). Furthermore, tumorigenesis of several cancers was also marked by specific methylation changes in their genomes (Yegnasubramanian et al., 2004). Therefore, it is useful to construct a global methylation profile to discover candidate genes and to predict therapeutic outcomes (Shen et al., 2007) and patient survival in cancer (Rosenbaum et al., 2005).

Pancreatic cancer is the fourth most common cause of cancer-related death in the United States, and the death rates for this disease closely mirror incidence rates (Jemal et al., 2007). Lack of early diagnostic biomarkers and ineffectiveness of current therapies for this cancer are among the major factors that contribute to its low survival rate (Jimeno and Hidalgo, 2006). In an effort to identify such biomarkers, many groups have used high-throughput molecular profiling technologies, including oligonucleotide and cDNA arrays and Serial Analysis of Gene Expression (SAGE), to analyzed gene expression data (Han et al., 2002; Iacobuzio-Donahue et al., 2002, 2003; Logsdon et al., 2003; Lowe et al., 2007; Ryu et al., 2002). Many candidate genes have been identified through these studies; however, the mechanism for the regulation of these genes is not fully understood. Using the candidate gene approach, Sato et al. (2003) identified seven overexpression genes (CLDN4, LCN2, MSLN, PSCA, S100A4, SFN and TFF2) in pancreatic cancer when compared to normal pancreatic duct were due to hypomethylation. Although the results are encouraging, the limitations for previous studies are the small number of genes examined (Esteller et al., 2001; Sato et al., 2003), which may fail to uncover other candidate genes regulated by DNA methylation in pancreatic cancer. Throughput this paper, we used the word methylation to refer DNA methylation. To address this issue, we have employed a global methylation profiling platform in this work to comprehensively survey a large panel of CpG sites across 800 genes in pancreatic cancer genome. We compared the DNA methylation profiles of the pancreatic tumors and normal tissues in order to unravel methylation markers for diagnostic purposes. We correlated the methylation markers with global gene expression profiles to identify candidate genes that were transcriptional regulated by methylation. By correlating methylation profiles with drug responses, we have identified candidate methylation markers that alter the expression of genes critical to gemcitabine susceptibility in pancreatic cancer.

2 Results

2.1 Methylation profiling in pancreatic cancer cell lines, tumors and normal tissues

To investigate the aberrant patterns of DNA methylation in pancreatic cancer, we employed global analysis to assess the methylation status of 1505 CpG sites across 807 genes in fourteen pancreatic cell lines, thirty pancreatic tumors and seven normal tissues (Supplementary Table 1). The methylation level of each CpG site is represented by the β-value (0≤β≤1), where 1 represents fully methylated and 0 represents unmethylated. The Illumina GoldenGate methylation assay (Bibikova et al., 2006) generates the profiles for these biological phenotypes, enabling us to perform global dissection of the aberrant DNA methylation patterns in pancreatic cancer. To our knowledge, this comprehensive database represents the largest survey of methylation status, many target sites per sample and many samples per study, in pancreatic cancer.

Table 1. Methylation markers in pancreatic cancer genome.
Chr Gene Description Classa
Hypermethylation
1 GSTM2 glutathione-S-transferase M2 d
NGFB nerve growth factor, beta polypeptide precursor d
PTCH2 patched 2 d
TAL1 T-cell acute lymphocytic leukemia 1 d
2 ALK anaplastic lymphoma kinase Ki-1 b
DES desmin d
FRZB frizzled-related protein d
IGFBP5 insulin-like growth factor binding protein 5 b
POMC proopiomelanocortin b
TMEFF2 transmembrane protein with EGF-like and two follistatin-like domains 2 d
4 CASP3 caspase 3 preproprotein b
CASP6 caspase 6 isoform alpha preproprotein d
EPHA5 ephrin receptor EphA5 isoform b d
FGF2 fibroblast growth factor 2 b
FGF5 fibroblast growth factor 5 isoform 1 precursor b
HHIP hedgehog-interacting protein d
IGFBP7 insulin-like growth factor binding protein 7 b
KDR kinase insert domain receptor (a type III receptor tyrosine kinase) d
KIT v-kit Hardy–Zuckerman 4 feline sarcoma viral oncogene homolog precursor d
PITX2 paired-like homeodomain transcription factor 2 isoform c b
SLIT2 slit homolog 2 d
5 FLT4 fms-related tyrosine kinase 4 isoform 2 d
SCGB3A1 secretoglobin, family 3A, member 1 d
6 ESR1 estrogen receptor 1 d
EYA4 eyes absent 4 isoform a d
7 COL1A2 alpha 2 type I collagen b
FZD9 frizzled 9 d
HOXA11 homeobox protein A11 d
HOXA9 homeobox protein A9 isoform b b
NPY neuropeptide Y d
SMO smoothened d
TFPI2 tissue factor pathway inhibitor 2 d
TWIST1 twist b
8 FGFR1 fibroblast growth factor receptor 1 isoform 1 precursor d
MOS v-mos Moloney murine sarcoma viral oncogene homolog d
NEFL neurofilament, light polypeptide 68kDa d
NRG1 neuregulin 1 isoform HRG-beta3 d
PENK proenkephalin d
SFRP1 secreted frizzled-related protein 1 d
SOX17 SRY-box 17 d
TUSC3 tumor suppressor candidate 3 isoform b b
9 APBA1 amyloid beta A4 precursor protein-binding, family A, member 1 d
DBC1 deleted in bladder cancer 1 d
FANCG Fanconi anemia, complementation group G b
GAS1 growth arrest-specific 1 d
TMEFF1 transmembrane protein with EGF-like and two follistatin-like domains 1 b
10 FGF8 fibroblast growth factor 8 isoform B precursor d
RET ret proto-oncogene isoform c d
11 ASCL2 achaete–scute complex homolog-like 2 d
BDNF brain-derived neurotrophic factor isoform a preproprotein b
FGF3 fibroblast growth factor 3 precursor d
FLI1 Friend leukemia virus integration 1 d
HSD17B12 steroid dehydrogenase homolog b
IGF2AS insulin-like growth factor 2 antisense d
LMO1 LIM domain only 1 d
MYOD1 myogenic differentiation 1 d
THY1 Thy-1 cell surface antigen d
WT1 Wilms tumor 1 isoform B d
12 CCND2 cyclin D2 d
ITPR2 inositol 1,4,5-triphosphate receptor, type 2 d
13 CCNA1 cyclin A1 d
FLT1 fms-related tyrosine kinase 1 d
FLT3 fms-related tyrosine kinase 3 d
SOX1 SRY (sex determining region Y)-box 1 d
14 CHGA chromogranin A precursor b
DLK1 delta-like 1 homolog isoform 1 b
15 NTRK3 neurotrophic tyrosine kinase, receptor, type 3 isoform b precursor d
RASGRF1 Ras protein-specific guanine nucleotide-releasing factor 1 isoform 1 b
16 CDH13 cadherin 13 preproprotein d
HS3ST2 heparan sulfate D-glucosaminyl 3-O-sulfotransferase 2 d
MMP2 matrix metalloproteinase 2 preproprotein d
MYH11 smooth muscle myosin heavy chain 11 isoform SM2 d
TUBB3 tubulin, beta, 4 b
17 ALOX12 arachidonate 12-lipoxygenase d
COL1A1 alpha 1 type I collagen preproprotein b
GAS7 growth arrest-specific 7 isoform a d
HIC1 hypermethylated in cancer 1 d
18 ADCYAP1 adenylate cyclase activating polypeptide precursor b
GALR1 galanin receptor 1 b
20 NTSR1 neurotensin receptor 1 b
21 ERG v-ets erythroblastosis virus E26 oncogene like isoform 2 d
22 HIC2 hypermethylated in cancer 2 d
SEZ6L seizure related 6 homolog (mouse)-like precursor d
TBX1 T-box 1 isoform A d
TIMP3 tissue inhibitor of metalloproteinase 3 precursor d
Hypomethylation
1 DHCR24 24-dehydrocholesterol reductase precursor a
EPHX1 epoxide hydrolase 1, microsomal (xenobiotic) a
HDAC1 histone deacetylase 1 a
MUC1 MUC1 mucin isoform 1 precursor c
NBL1 neuroblastoma, suppression of tumorigenicity 1 precursor a
NES nestin c
NID1 nidogen (enactin) c
S100A2 S100 calcium binding protein A2 c
SFN stratifin a
TGFB2 transforming growth factor, beta 2 a
2 CASP8 caspase 8 isoform A a
CLK1 CDC-like kinase 1 isoform 1 a
GLI2 GLI-Kruppel family member GLI2 isoform alpha c
IHH Indian hedgehog homolog c
IL1RN interleukin 1 receptor antagonist isoform 4 c
UGT1A1 UDP glycosyltransferase 1 family, polypeptide A1 precursor c
VAMP8 vesicle-associated membrane protein 8 a
3 ACVR2B activin A type IIB receptor precursor a
CCR5 chemokine (C–C motif) receptor 5 c
MST1R macrophage stimulating 1 receptor a
PPARG peroxisome proliferative activated receptor gamma isoform 1 c
TNFSF10 tumor necrosis factor (ligand) superfamily, member 10 c
4 CCKAR cholecystokinin A receptor c
CXCL9 small inducible cytokine B9 precursor c
IL2 interleukin 2 precursor c
IL8 interleukin 8 precursor a
SPP1 secreted phosphoprotein 1 a
5 CSF1R colony stimulating factor 1 receptor precursor c
CSF2 colony stimulating factor 2 precursor a
FGF1 fibroblast growth factor 1 (acidic) isoform 2 precursor c
FGFR4 fibroblast growth factor receptor 4 isoform 1 precursor c
IL12B interleukin 12B precursor c
ITK IL2-inducible T-cell kinase a
6 CCND3 cyclin D3 a
FRK fyn-related kinase c
NOTCH4 notch4 preproprotein c
PPARD peroxisome proliferative activated receptor, delta isoform 1 c
SLC22A3 solute carrier family 22 member 3 c
SPDEF SAM pointed domain containing ets transcription factor a
7 ASB4 ankyrin repeat and SOCS box-containing protein 4 isoform a c
CLDN4 claudin 4 a
CPA4 carboxypeptidase A4 preproprotein a
EPHB4 ephrin receptor EphB4 precursor a
FLJ20712 hypothetical protein LOC55025 c
LMTK2 lemur tyrosine kinase 2 c
MEST mesoderm specific transcript isoform a a
NOS3 nitric oxide synthase 3 (endothelial cell) c
PRSS1 protease, serine, 1 preproprotein c
SEMA3C semaphorin 3C c
TRIP6 thyroid hormone receptor interactor 6 a
8 CDH17 cadherin 17 precursor c
DLC1 deleted in liver cancer 1 isoform 1 c
E2F5 E2F transcription factor 5 a
PLAT plasminogen activator, tissue type isoform 2 precursor a
PSCA prostate stem cell antigen c
SFTPC surfactant, pulmonary-associated protein C c
TNFRSF10A tumor necrosis factor receptor superfamily, member 10a a
9 DAPK1 death-associated protein kinase 1 c
LCN2 lipocalin 2 (oncogene 24p3) c
SYK spleen tyrosine kinase a
VAV2 vav 2 oncogene a
10 ABCC2 ATP-binding cassette, subfamily C (CFTR/MRP), member 2 a
CYP2E1 cytochrome P450, family 2, subfamily E, polypeptide 1 c
FGFR2 fibroblast growth factor receptor 2 isoform 12 precursor c
MAP3K8 mitogen-activated protein kinase kinase kinase 8 c
SFTPA1 surfactant, pulmonary-associated protein A1 c
SNCG synuclein, gamma (breast cancer-specific protein 1) c
TCF7L2 transcription factor 7-like 2 (T-cell specific, HMG-box) c
11 APOA1 apolipoprotein A-I preproprotein c
CCND1 cyclin D1 a
DDB2 damage-specific DNA binding protein 2 (48kD) c
INS proinsulin precursor c
MMP1 matrix metalloproteinase 1 preproprotein a
MMP7 matrix metalloproteinase 7 preproprotein a
PHLDA2 pleckstrin homology-like domain family A member 2 a
SPI1 spleen focus forming virus (SFFV) proviral integration oncogene spi1 c
TMPRSS4 transmembrane protease, serine 4 isoform 1 a
TRIM29 tripartite motif protein TRIM29 isoform alpha c
TSG101 tumor susceptibility gene 101 a
12 ARHGDIB Rho GDP dissociation inhibitor (GDI) beta a
IAPP islet amyloid polypeptide precursor a
IFNG interferon, gamma a
KRT1 keratin 1 a
PTHLH parathyroid hormone-like hormone isoform 2 preproprotein c
PTPN6 protein tyrosine phosphatase, non-receptor type 6 isoform 2 a
UNG uracil-DNA glycosylase isoform UNG1 precursor a
14 BMP4 bone morphogenetic protein 4 preproprotein c
KIAA0125 hypothetical protein LOC9834 a
RIPK3 receptor-interacting serine-threonine kinase 3 a
15 APBA2 amyloid beta A4 precursor protein-binding, family A, member 2 c
MAGEL2 MAGE-like protein 2 c
NDN necdin a
TJP1 tight junction protein 1 isoform b a
16 CARD15 NOD2 protein c
CREBBP CREB binding protein c
NQO1 NAD(P)H menadione oxidoreductase 1, dioxin-inducible isoform c a
PRSS8 prostasin preproprotein a
17 BRCA1 breast cancer 1, early onset isoform BRCA1-delta9-11 c
CRK v-crk sarcoma virus CT10 oncogene homolog isoform b a
CSF3 colony stimulating factor 3 isoform c c
ITGB4 integrin beta 4 isoform 1 precursor a
LIG3 ligase III, DNA, ATP-dependent isoform alpha precursor a
NOS2A nitric oxide synthase 2A isoform 1 a
SEPT9 septin 9 a
18 SERPINB5 serine (or cysteine) proteinase inhibitor, clade B (ovalbumin), member 5 a
19 BAX BCL2-associated X protein isoform sigma c
BSG basigin isoform 1 a
CEACAM1 carcinoembryonic antigen-related cell adhesion molecule 1 isoform 1 precursor c
EMR3 egf-like module-containing mucin-like receptor 3 isoform b c
JAK3 Janus kinase 3 c
NOTCH3 Notch homolog 3 c
PLAUR plasminogen activator, urokinase receptor isoform 3 precursor a
PTPRH protein tyrosine phosphatase, receptor type, H precursor a
20 DNMT3B DNA cytosine-5 methyltransferase 3 beta isoform 2 a
ID1 inhibitor of DNA binding 1 isoform a a
MMP9 matrix metalloproteinase 9 preproprotein c
PI3 elafin preproprotein c
PTK6 PTK6 protein tyrosine kinase 6 c
SRC proto-oncogene tyrosine-protein kinase SRC c
21 B3GALT5 UDP-Gal:betaGlcNAc beta 1,3-galactosyltransferase 5 c
RIPK4 ankyrin repeat domain 3 a
TFF1 trefoil factor 1 precursor a
TFF2 trefoil factor 2 precursor a
22 BCR breakpoint cluster region isoform 2 a
LIF leukemia inhibitory factor (cholinergic differentiation factor) a
X BGN biglycan preproprotein c
GRPR gastrin-releasing peptide receptor c
GUCY2F guanylate cyclase 2F c
MAGEA1 melanoma antigen family A, 1 a
  • a Classes: (a) genes with CpG sites' hypomethylation and increased mRNA expression; (b) genes with CpG sites' hypermethylation and mRNA increased expression; (c) genes with CpG sites' hypomethylation and decreased mRNA expression; and (d) genes with CpG sites' hypermethylation and mRNA decreased expression.

2.2 Methylation assay reproducibility

To demonstrate the reproducibility of the high-throughput DNA methylation profiling technique used in this study, biological replicates were profiled on the xenograft samples. We obtained highly reproducible DNA methylation profiles between these biological replicates with an average R2 of 0.94±0.16 when the β-values were compared. As illustrated in Supplementary Figure 1, when β-values for matching normal (JH033-Normal) and cancer (JH033-Cancer) samples were compared, differential methylation was readily detected.

Details are in the caption following the image
Distribution of methylation status in pancreatic cancer and normal genomes. Globally, the methylation levels of the pancreatic cancer cell lines, cancerous and normal tissues were distributed bimodally, with higher percentage of CpG sites populated either at the high (hypermethylation) or low (hypomethylation) end of the β-value scale.

2.3 Distribution of methylation status in pancreatic cancer and normal genomes

To characterize the global methylation status of pancreatic cancer genome, we first analyzed the distribution of β-values across 1505 CpG sites in pancreatic cancer cell lines, tumors and normal tissues. Globally, the methylation levels of for these samples were distributed bimodally, with higher percentage of CpG sites populated either at the high (hypermethylation) or low (hypomethylation) end of the β-value scale (Figure 1). On average, hypomethylation (β≤0.1) was observed in 38%, 49% and 44% of the studied CpG sites in pancreatic cancer cell lines, pancreatic cancer and normal tissues, respectively. Pancreatic cancer cell lines showed higher (25%) hypermethylation (β>0.9) of the CpG sites in this study when compared to 17% and 15% in pancreatic cancer and normal tissues, respectively. This may indicates that the pancreatic cancer cell lines are less stable in their epigenomes. There is not statistically significance between the distribution of methylation sites in the cancer cell lines, cancerous and normal tissues in this study on the global scale; however, each individual tissue and cell line poses unique methylation patterns. Genes located at the two ends are the most interesting in this study, as they are likely regulated by epigenetic events (hypermethylation or hypomethylation) in pancreatic cancer genome.

2.4 Identifying methylation markers in pancreatic cancer

To investigate whether global DNA methylation in CpG sites can distinguish between normal and pancreatic cancer, we performed unsupervised clustering on the seven matched cancer–tumor pairs on 1505 CpG sites based on their methylation status (β-values). Figure 2a shows the unsupervised clustering dendogram. It clearly shows that the normal and cancerous tissues can be separated by the global DNA methylation status of these CpG sites. This demonstrates that methylation profiles can be used to distinguish normal samples from cancers.

Details are in the caption following the image
Unsupervised clustering. (a) Unsupervised clustering of the seven pancreatic cancer–normal pairs based on global methylation profiles. It clearly shows that the normal and cancerous tissues can be separated by the global DNA methylation status of these CpG sites. This demonstrates that methylation profiles can be used to distinguish normal samples from cancers. (b) Unsupervised clustering of the thirty pancreatic cancers, fourteen pancreatic cancer cell lines and seven normal samples based on the methylation profiles of the 289 CpG sites. It shows three distinct clusters where the normal samples are tightly clustered together. The pancreatic cancer cell lines and tumors were clustered together, yet distinct methylation patterns exist between them.

Next, we used significance analysis of microarrays (SAM) (Tusher et al., 2001) to identify CpG sites (methylation markers) that are differentially methylated in the seven matched normal–tumor pairs. At the false discovery rate equals to 0, a total of 289 CpG sites in 214 genes with an average methylation level at least 2-fold higher or lower in pancreas adenocarcinomas compared to non-adenocarcinoma samples were identified. Of these, 120 and 169 CpG sites were hypermethylated and hypomethylated in the pancreatic cancer samples, respectively (Supplementary Table 2). These CpG sites were deemed as statistical significance methylation markers as their differential methylation status can be used to distinguish pancreatic cancer from normal tissue samples.

2.5 Clustering of independent sample sets based on the identified methylation markers

We next attempted to cluster the pancreatic cancer cell lines and independent pancreatic cancers based on these methylation markers. We employed the hierarchical clustering algorithm to organize these samples based on the CpG methylation status of these markers. Figure 2b shows the dendrogram of these relationships where the length of the branches reflects the relatedness of the samples. Overall, pancreatic tumor samples, cancer cell lines and normal tissues formed three distinct clusters based on the methylation status of these CpG sites. Two tumors (265 and JH034) and one cell line (Panc1) formed a cluster more closely to the normal tissues. One cell line (ASPC1) was clustered together with the tumors. This suggests that the methylation patterns for these markers are different in pancreatic cancer cell lines, tumors and normal samples.

2.6 Hypermethylation sites are enriched in CpG islands

Many studies have showed that methylation patterns are severely disrupted in human tumors. Aberrant hypermethylation of CpG islands in promoter regions represents one of the most frequent epigenetic events associated with gene silencing in cancer. For the identified methylation markers, we observed 119/120 (99.2%) hypermethylation sites are located within CpG islands (hypergeometric test, p<9×10−21) (Supplementary Table 2). However, no statistically enrichment of the 57/169 (33.7%) hypomethylation sites are located within CpG islands. We next asked whether there is an enrichment of these methylation markers in the promoter regions. We found 67/120 (55.8%) of the hypermethylation sites are located in the promoter regions compared to the 122/169 (72.2%) of the hypomethylation sites, but neither of them is statistically significance (Supplementary Table 2).

2.7 Methylation markers in distinguishing pancreatic cancers from normal tissues

The 289 methylation markers identified were corresponding to 85 and 129 genes that were hypermethylated and hypomethylated in the pancreatic cancer genome, respectively (Table 1). DNA methylation status for many of these genes was known to be altered in human cancers, including pancreatic cancer (Esteller, 2007). Most notably is the cyclin D2 (CCND2) gene, which is known to be hypermethylated in pancreatic cancer, and is currently being used as a clinical methylation marker to diagnose this disease (Matsubayashi et al., 2003).

Many hypermethylated genes identified in Table 1 were known to be correlated with various cancers (Esteller, 2007; Esteller et al., 2001; Feinberg et al., 2006), including cadherins (CDH13 and CDH17), transcription factors (HIC1 and HIC2), homeobox proteins (HOXA9 and HOXA11), growth factor binding proteins (IGFBP5 and IGFBP7), SFRP1, DBC1, MOS (Scholz et al., 2005), TMEF2, TIMP3 and WT1(Satoh et al., 2003a,b). Transcriptional silencing of these genes has been shown to be associated with CpG island promoter hypermethylation in human cancers (Esteller, 2007). This indicates that methylation patterns are unique to specific cancer, and can be used to differentiate between malignancies (Bibikova et al., 2006).

We identified five imprinting genes (CPA4, MEST, MAGEL2, NDN, and SLC22A3) and four X-linked genes (BGN, GPRP, GUCY2F and MAGEA1) that were hypomethylated in the pancreatic cancers as compared to the normal tissues (Table 1). This suggests that LOI and activation of these genes may contribute to pancreas tumorigenesis. Interestingly, hypomethylation of MAGEA1, a male germline specific gene, was also found in many types of tumors (De Smet et al., 2004). Somatic mutation for one of the X-linked genes, GUCY2F, has been observed in breast, lung, colon and pancreatic cancer (Wood et al., 2006). This indicates that, according to the two-hit hypothesis (Herman and Baylin, 2003), some of the tumors may have loss transcriptional repression in GUCY2F and could cause the increased expression of the mutant copy in pancreatic cancer.

We also observed several transcription factors (E2F5, TFF1, TFF2 and ID1) that were hypomethylated in pancreatic cancers as compared to normal samples (Table 1). This suggests that cancer cell cycle may be disrupted through the increased activities of these hypomethylated transcription factors.

2.8 Correlating methylation markers and gene expression profiles

To investigate the relationship between DNA methylation and gene expression in pancreatic cancer genome, we correlated the DNA methylation status of the methylation markers with the global gene expression profiles. We performed linear regression analysis to correlate changes in CpG methylation patterns and basal gene expression levels on the thirty tumor cases. On average, an inverse correlation (−0.45±0.1) was observed between methylation levels and mRNA expression levels (T-test, p<0.05). This demonstrates that increased mRNA expression levels were correlated with hypomethylation or vice versa (decreased mRNA expression levels were correlated with hypermethylation).

From this correlation analysis, aberrant DNA methylation in the 214 genes can be classified into four categories: (a) 63 genes with CpG sites' hypomethylation and increased mRNA expression; (b) 24 genes with CpG sites' hypermethylation and mRNA increased expression; (c) 66 genes with CpG sites' hypomethylation and decreased mRNA expression; and (d) 61 genes with CpG sites' hypermethylation and mRNA decreased expression (Table 1). Genes that showed concordance between methylation patterns and mRNA expression levels (categories (b) and (c)) may suggest that the expressions of these genes were not regulated by DNA methylation. Another possible explanation is that the CpG sites used in this study were limited in covering all the important CpG dinucleotide sites of these genes, therefore, this may missed out some of the inverse correlations observed between methylation patterns and transcript expression levels for these genes. In contrast, genes within the two discordant categories (a) and (d) are the most interesting in this study, as they are likely candidate genes regulated by hypermethylation or hypomethylation. They represent 124 of the 807 (15.4%) genes we investigated in this study.

2.9 Identifying candidate genes regulated by aberrant DNA methylation

To pinpoint the candidate genes regulated by aberrant DNA methylation in pancreatic cancer, we focused on the 124 genes of discordant categories (a) and (d) in Table 1. Using the gene expression profiles of the two normal pancreas cell lines as baseline, we compare these genes in the thirty pancreatic tumors and fourteen cancer cell lines to the normal cell line profiles. We call the expression of a gene is transcriptional silenced by DNA methylation (hypermethylation) if its expression is under-expressed in all the tumors and cancer cell lines when compared to the expression of the normal cell lines. Conversely, if a gene has increased its expression in all the tumors and cancer cell lines when compared to the baseline expressions, then that gene has loss its fidelity in maintaining methylation status (hypomethylation). Using these criteria, we identified 23 and 35 candidate genes that are regulated by hypermethylation and hypomethylation, respectively (Figure 3 and Table 2).

Details are in the caption following the image
Average expression of the candidate genes in pancreatic cancer genome regulated by DNA methylation. Xeno, PCL and NCL columns represent xenografts, pancreatic cancer cell lines and normal cell lines, respectively. Rows represent candidate genes. Red and green colors represent relative gene overexpression and underexpression.
Table 2. Candidate gene list where their expression is regulated by DNA methylation.
Gene Description
Hypermethylation
ALOX12 arachidonate 12-lipoxygenase
DBC1 deleted in bladder cancer 1
DES desmin
EPHA5 EPH receptor A5
ERG v-ets erythroblastosis virus E26 oncogene homolog (avian)
ESR1 estrogen receptor 1
EYA4 eyes absent homolog 4 (Drosophila)
FLT1 fms-related tyrosine kinase 1 (vascular endothelial growth factor/vascular permeability factor receptor)
FLT3 fms-related tyrosine kinase 3
GAS7 growth arrest-specific 7
HS3ST2 heparan sulfate (glucosamine) 3-O-sulfotransferase 2
KIT v-kit Hardy–Zuckerman 4 feline sarcoma viral oncogene homolog
MOS v-mos Moloney murine sarcoma viral oncogene homolog
MYH11 myosin, heavy chain 11, smooth muscle
NEFL neurofilament, light polypeptide 68kDa
NGFB nerve growth factor, beta polypeptide
NTRK3 neurotrophic tyrosine kinase, receptor, type 3
PENK proenkephalin
PTCH2 patched homolog 2 (Drosophila)
SEZ6L seizure related 6 homolog (mouse)-like
SLIT2 slit homolog 2 (Drosophila)
SMO smoothened homolog (Drosophila)
THY1 Thy-1 cell surface antigen
Hypomethylation
ACVR2B activin A receptor, type IIB
BCR breakpoint cluster region
BSG basigin (Ok blood group)
CASP8 caspase 8, apoptosis-related cysteine peptidase
CCND1 cyclin D1
CCND3 cyclin D3
CLK1 CDC-like kinase 1
CPA4 carboxypeptidase A4
CRK v-crk sarcoma virus CT10 oncogene homolog (avian)
DNMT3B DNA (cytosine-5)-methyltransferase 3 beta
EPHB4 EPH receptor B4
EPHX1 epoxide hydrolase 1, microsomal (xenobiotic)
HDAC1 histone deacetylase 1
IAPP islet amyloid polypeptide
ID1 inhibitor of DNA binding 1, dominant negative helix-loop-helix protein
KIAA0125 KIAA0125
LIF leukemia inhibitory factor (cholinergic differentiation factor)
LIG3 ligase III, DNA, ATP-dependent
MEST mesoderm specific transcript homolog (mouse)
MMP7 matrix metallopeptidase 7 (matrilysin, uterine)
MST1R macrophage stimulating 1 receptor (c-met-related tyrosine kinase)
NBL1 neuroblastoma, suppression of tumorigenicity 1
NOS2A nitric oxide synthase 2A (inducible, hepatocytes)
PHLDA2 pleckstrin homology-like domain, family A, member 2
PLAT plasminogen activator, tissue
PLAUR plasminogen activator, urokinase receptor
PTPN6 protein tyrosine phosphatase, non-receptor type 6
PTPRH protein tyrosine phosphatase, receptor type, H
SEPT9 septin 9
SFN stratifin
SPDEF SAM pointed domain containing ets transcription factor
TGFB2 transforming growth factor, beta 2
TJP1 tight junction protein 1 (zona occludens 1)
TSG101 tumor susceptibility gene 101
VAV2 vav 2 oncogene

Among the candidate genes listed in Table 2, the overexpression of SFN (also known as 14-3-3-σ) in pancreatic cancer is due to hypomethylation was confirmed by Sato et al. (2003). This supports our approach is valid in identifying candidate genes regulated by aberrant DNA methylation in pancreatic cancer. Cyclin D1 and D3 (CCND1 and CCND3) have been showed to be overexpressed in human pancreatic cancer (Ebert et al., 2001; Gansauge et al., 1997). Here, CCND1 and CCND3 were identified as hypomethylated candidate genes, suggesting that overexpression of these cyclins (Ebert et al., 2001; Gansauge et al., 1997) in pancreatic cancer is associated with the loss of methylation. We also identified MMP7 as a hypomethylated candidate gene, where the overexpression of this gene has been shown to be a useful biomarker in pancreatic cancer (Kuhlmann et al., 2007).

To further characterize the candidate genes regulated by DNA methylation, we used DAVID annotation program to group genes according to their biological functional classes. Eight of the hypomethylated candidate genes (LIG3, CCND3, CCND1, SFN, CLK1, NBL1, TGFB2, and ID1) were involved in cell cycle and cell proliferation according to Gene Ontology classification. Six of the hypomethylated candidate genes (CASP8, PHLDA2, PTPN2, TGFB2, HDAC1 and IAPP) were involved in apoptosis. This suggests that increased expression of these genes due to hypomethylation may contribute to the uncontrollable cell proliferation and genomic instability in pancreatic cancer.

For the hypermethylated candidate genes, five of them (KIT, FLT1, FLT3, NTRK3 and EPHA5) are members of the tyrosine kinase family. This suggests that underexpression of these genes may contribute to the lack of sensitivity in certain tyrosine kinase target drugs, such as imatinib (Chen et al., 2006), in pancreatic cancer.

2.10 Validation by methylation specific PCR (MSP)

We performed methylation specific PCR (MSP) validation on three hypermethylated genes (DBC1, FLT1 and EYA4) from Table 1 (candidate genes) in four tumors used in this study (410, 420, JH011 and JH015). Figure 4a shows that the CpG sites identified as hypermethylation by the Illumina Bead arrays (high β-values) were indeed methylated, as validated by MSP (Figure 4b). Methylation of DBC1, FLT1 and EYA4 was observed by MSP in 75% (3/4), 100% (4/4) and 100% (4/4) tumors respectively (Figure 4b). We also performed MSP on two normal pancreatic cell lines (HPDE and HPNE) to validate these methylation changes are specific in tumors. Indeed, we found that the two normal cell lines were not methylated in these genes, except HPNE showed methylation pattern in DBC1 (Figure 4c). The primers used for the MSP analysis are listed in the Supplementary Table 3.

Details are in the caption following the image
Validation by the methylation specific PCR. (a) β-values of CpG sites in three candidate genes (DBC1, FLT1 and EYA4) as assessed as hypermethylation by the Illumina Bead Array platform. Green color indicates that the methylation calls were validated by MSP method. (b) MSP validation of these genes. U and M columns indicate unmethylated and methylated bands. DKO and IVO were used as the unmethylated (U) and methylated (M) controls. (c) MSP results for the two normal pancreas cell lines on these genes.

2.11 Correlating DNA methylation patterns with gemcitabine sensitivity

We next asked whether DNA methylation could be used as molecular markers to predict sensitivity or resistance to chemotherapy. Gemcitabine has been approved as the first-line treatment for pancreatic cancer patients; however, about 75% of patients have minimal benefit from this therapy. Moreover, the genetic factors that predict gemcitabine sensitivity are not fully understood. We hypothesized that aberrant DNA methylation patterns may be used as biomarkers to predict gemcitabine susceptibility in pancreatic cancer. To test this hypothesis, we treated the thirty direct-patient xenograft models with gemcitabine. Relative tumor growth inhibition (TGI) values for these tumors were measured after 28 days of gemcitabine treatment. The thirty xenografts showed varying degrees of gemcitabine sensitivity, ranging from −88% to 59% TGI. For simplicity, in this study, we stratify the tumors into sensitive and resistant groups. We considered tumors with TGI≤−20% as sensitive and tumors with TGI>−20% as resistant to gemcitabine, respectively. Using this cut-off, ten tumors passed the sensitive threshold, and the other twenty tumors were regarded as resistant. Next, we ran SAM on the DNA methylation profiles of the thirty tumors to identify differentially methylated DNA markers in sensitive versus resistant groups. From the SAM analysis, GSTM1_P266_F and ONECUT2_E96_F, two DNA markers were identified as statistically significant in distinguishing sensitive from resistant tumors.

We next sought to ask whether these DNA markers can be used as predictor for gemcitabine sensitivity in pancreatic cancer. To test the applicability of these DNA markers, we build a weighted voting predictor from the thirty tumors' methylation profiles (training set) and predict the gemcitabine sensitivity in the twelve pancreatic cancer cell lines (independent test set). The predictor predicts ten and two cancer cell lines as sensitive and resistant to gemcitabine, respectively (Table 3). To validate the prediction of the predictor based on DNA methylation markers, we performed MTT assay on the twelve pancreatic cancer cell lines for gemcitabine exposure at 10μM for 72h. From the MTT analysis, eleven of the twelve cancer cell lines are sensitive to gemcitabine where viable cell mass after gemcitabine treatment is ≤50% compared to controls; and one cell line (MiaPaca2) is resistant to gemcitabine with viable cell mass>50% compared to controls (Table 3). Hence, the predictor correctly predicts the gemcitabine sensitivity in eleven cell lines, and only missed out one prediction (Panc1) (Table 3).

Table 3. Gemcitabine sensitivity predicted by methylation markers and validated by MTT assay.
Cancer cell lines Prediction MTT (% viable cell mass compared to control) Correct prediction?
Panc430 SEN 10 Yes
XPA3 SEN 12 Yes
Panc203 SEN 14 Yes
Panc1 RES 19 No
Panc1005 SEN 21 Yes
E3LZ10 SEN 23 Yes
HS776T SEN 23 Yes
ASPC1 SEN 30 Yes
XPA4 SEN 38 Yes
L36PL SEN 46 Yes
E3JD13 SEN 49 Yes
MiaPaca2 RES 79 Yes

In the gemcitabine sensitive tumors, GSTM1_P266_F has higher methylation level (βSEN=0.95±0.06) when compared with the resistant tumors (βRES=0.72±0.19). This higher methylation level may reduce the gene expression of GSTM1 in the sensitive cases. Glutathione-S-transferases μ 1 (GSTM1) is a key enzyme involved in the glutathione detoxification pathway in mammalian cells. This class of enzyme catalyzed the formation of glutathione conjugates with xenobiotics, toxic superoxides, or antineoplastic agents. Once the conjugates were formed, they can be exported from cells by ATP-dependent glutathione-S-conjugate complex export pump. Some cancer cell line studies have suggested that the activation or elevated of GST expression decreases drug effects; however, down-regulation and inhibiting GST enhances drug sensitivity (Yang et al., 2006). Indeed, we observed this inversed correlation between GSTM1 gene expression and methylation levels in these thirty cases (Figure 5), and the effect is more profound in the sensitive cases (TGI≤−20%).

Details are in the caption following the image
Correlation of DNA methylation and gene expression levels with gemcitabine sensitivity (a) xenografts models and (b) pancreatic cell lines. GSTM1_Affy and ONECUT2_Affy represent relative gene expression profiles obtained from Affymetrix GeneChip data. GSTM1_Methyl and ONECUT2_Methyl represent relative methylation status obtained from the methylation profiling data. Red and green colors indicate high and low expression/methylation, respectively.

The other DNA methylation marker for predicting gemcitabine sensitivity is ONECUT2_E96_F, a transcription factor characterized by the presence of a single ‘cut’ domain and an atypical homeodomain (one cut domain, family member 2). This gene has been suggested to participate in the network of transcription factors required for liver differentiation and metabolism (Jacquemin et al., 1999). However, the role of ONECUT2 in pancreatic cancer is still unknown. Similar inversed correlation between ONECUT2 gene expression and methylation patterns was observed in the tumor cases (Figure 5). Our current data suggest that hypermethylation of GSTM1 and ONECUT2 may synergized to gemcitabine susceptibility in a subset of pancreatic cancer. (Chen et al., 2006)

3 Discussion

In this study, we investigated the methylation status of a relatively large panel of genes in a series of pancreatic cancer cell lines, primary pancreatic carcinoma and normal pancreas tissues using global methylation profiling assay. By comparing the methylation profiles and gene expression data, we identified a set of candidate genes in pancreatic cancer cell lines and tumors that show an inverse correlation between DNA methylation and mRNA expression. These results suggest that expression changes in these candidate genes may due to abnormal DNA methylation at the promoter regions. Hypermethylation of silencers may also increase the expression of these candidate genes. We found that it is more common to detect hypomethylation of genes that are overexpressed in pancreatic cancer when they are normally methylated and not expressed in non-neoplastic pancreas. This is also supported by the observation of Sato et al. (2003) where DNA hypomethylation is a frequent epigenetic alteration in pancreatic cancer and is associated with the overexpression of affected genes. Several hypermethylated genes identified in this study were also preserved the same methylation pattern in various cancers (Esteller, 2007). Our results strengthen that aberrant DNA methylation is a ubiquitous feature of cancer.

Recent studies have been suggested that aberrant DNA methylation can affect the sensitivity of cancers to chemotherapeutics by altering expression of genes critical to drug response. These studies have showed that methylation of MGMT and CHFR indicates sensitivity to carmustine and microtubule inhibitors in gliomas and in gastric cancers, respectively (Esteller et al., 2000; Satoh et al., 2003a,b). Shen et al. (2007) found association between p73 hypermethylation and increased sensitivity to alkylating agents by correlating drug activity with methylation status of 32 CpG island-associated genes in the NCI-60 cancer cell lines. In this study, we identified DNA methylation of GSTM1 and ONECUT2 was associated with gemcitabine sensitivity in pancreatic cancer.

In summary, we demonstrate the use of methylation profiling to identify a list of candidate genes where their expressions are regulated by DNA methylation and demethylation in pancreatic cancer. We also show that integrating high-throughput methylation and gene expression analyses are useful in identifying biomarkers for cancer drug sensitivity.

4 Experimental procedures

4.1 Direct-patient xenografts, normal and pancreatic cancer cell lines

Pancreatic cancer xenografts were established from surgically resected primary pancreatic carcinomas, and thirty xenografts were selected for this study. We also collected seven adjacent normal pancreas tissues from the subset of these xenografts. Fourteen human pancreatic cancer cell lines, Panc1, Panc203, Panc430, Panc504, Panc813, Panc1005, HS766T, MiaPaca2, E3.JD13, E3.LZ10, ASPC1, L3.6PL, XPA3 and XPA4, were used in this study. We used HPNE and HPDE, two non-transformed pancreatic ductal epithelial cell lines, as the normal pancreas cell lines in this study.

4.2 DNA methylation profiling assay

The GoldenGate Methylation Cancer Panel I contains 1505 CpG sites distributed across 807 genes; 1044 CpG sites are located within CpG islands, and 461 are located outside of CpG islands (Supplementary Table 1). This technology adapts the Illumina high-throughput single nucleotide polymorphism genotyping system to assay the bead-based array platform. Each methylation data site is captured by fluorescent signals from the M (methylated) and U (unmethylated) alleles. To determine the intensity for each CpG site (β), background intensity was subtracted from each data point and the fluorescent signals' ratio from the two alleles is computed as: β=(max(M,0))/(|U|+|M|+100). The β-value (0≤β≤1) reflects the methylation level of each CpG site where 1 represents fully methylated and 0 represents unmethylated (Bibikova et al., 2006).

4.3 Microarray gene expression profiling data

Gene expressions of the thirty pancreatic xenograft tumors, normal and pancreatic cancer cell lines were profiled using Affymetrix U133 Plus 2.0 gene arrays. Sample preparation and processing procedure were performed as described in the Affymetrix GeneChip® Expression Analysis Manual (Affymetrix Inc., Santa Clara, CA).

4.4 Significance analysis of microarray (SAM)

We used SAM (Tusher et al., 2001) to select genes that show statistically significant changes in their methylation patterns. We obtained the SAM algorithm implemented as a Microsoft Excel add-in from http://www-stat.stanford.edu/∼tibs/SAM/.

4.5 Hierarchical clustering

We used the Euclidean distance metric on the expression matrix and performed complete linkage hierarchical clustering using Cluster 3.0 (de Hoon et al., 2004) and visualized it in Java TreeView (v1.0.13) (Saldanha, 2004).

4.6 In vivo growth inhibition studies

Six-week-old female athymic nude mice (Harlan, IN, US) were used to evaluate the in vivo growth inhibition studies. The xenografts were generated according to previous published methodology (Rubio-Viqueira et al., 2006), and the research protocol was approved by the Johns Hopkins University Animal Care and Use Committee and animals were maintained in accordance to guidelines of the American Association of Laboratory Animal Care. In brief, surgical non-diagnostic specimens of patients operated at the Johns Hopkins Hospital were reimplanted subcutaneously to 1–2 mice for each patient, with 2 small pieces per mouse (F1 generation). Tumors were let to grow to a size of 1.5cm3 at which point were harvested, divided, and transplanted to another 5 mice (F2 generation). After a second growth passage tumors were excised and propagated to cohorts of 20 mice or more, that constituted the treatment cohort (F3 generation). Tumors in the PancXenoBank are kept as a live bank that is expanded as required for drug testing and biologic studies. Tumors from this treatment cohort were allowed to grow until reaching ∼200mm3, at which time mice were randomized in the following treatment groups: 1) control and 2) gemcitabine 100mg/kg 2 times a week ip; with five to six mice (at least ten tumors) in each group. Treatment was given for 28 days. Mice were monitored daily for signs of toxicity and were weighed three times per week. Tumor size was evaluated two times per week by caliper measurements using the following formula: tumor volume=[length×width2]/2. Relative tumor growth inhibition (TGI) was calculated by relative tumor growth of treated mice divided by relative tumor growth of control mice since the initiation of therapy.

4.7 Methylation specific PCR (MSP)

Bisulfite modification of genomic DNA was carried out using the EZ DNA methylation Kit (Zymo Research, Orange, CA). We performed methylation analysis of the promoter regions of various genes using MSP primer pairs covering the putative transcriptional start site in the 5′ CpG island with 1μL of bisulfite-treated DNA as template and JumpStart Red Taq DNA Polymerase (Sigma, St. Louis, MO) for amplification as previously described (1). MSP primers were designed using the online software program MSPPrimer (http://www.mspprimer.org). DKO and IVO were used as the unmethylated (U) and methylated (M) controls.

4.8 In vitro drug sensitivity assay

To evaluate the in vitro drug sensitivity, pancreatic cancer cell lines treated with gemcitabine were assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT; Sigma, St Louis, MO). 5000 cells were seeded in 96-well plates, transfected during 48h, and then exposed to either vehicle or gemcitabine at a concentration of 10μM for 72h. Each experiment was performed in sextuplicate independently at least three times.

4.9 Weighted voting predictor

Weighted voting predictor (Golub et al., 1999) was used to distinguish gemcitabine sensitive from resistant cases based on methylation patterns. Let μ12) and δ12) represent mean and standard deviation for the sensitive (resistant) cases. Signal-to-noise ratio (S2N) for a marker g is computed as (μ1−μ2)/(δ12) and the average mean Mg between two classes is calculated as (μ12)/2. To make the prediction for a new case X, for each methylation marker g, the weight for its vote is computed as vg=S2Ng×(Xg−Mg), with a positive value indicating a vote for sensitive and a negative value indicating a vote for resistant. If the total vote for class sensitive (obtained by summing the absolute values of the positive votes over the methylation markers) is greater than the total vote for class resistant (obtained by summing the absolute values of the negative votes), the weighted voting predictor predicts the new case X as sensitive and vise versa.

Acknowledgements

Supported in part by the Sol Golman Center for Pancreatic Cancer Research, the Viragh Family Foundation and the Lee family.

    Conflict of interest

    None.

    Supplementary data A

    Supplementary data associated with this article can be found, in the online version, at doi:10.1016/j.molonc.2009.03.004.