ANTICANCER RESEARCH 26: 55-58 (2006)

Lymph Node Metastasis is Infrequent in Patients with Highly-methylated Colorectal Cancer

KENJI HIBI1 and AKIMASA NAKAO2

1Department of Surgery, Seirei Hospital, 56 Kawanayama-machi, Showa-ku, Nagoya 466-8633; 2Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8560, Japan

Abstract. In this study, the combined methylation status of p16, was found to harbor promoter hypermethylation p16, p14, HLTF, CDH13 and RUNX3 was examined in 59 associated with the loss of protein expression in cancer cells resected primary colorectal cancers using methylation-specific (7). Though homozygous deletions of the p16 were not PCR and the methylation status was correlated with the present (8), p16 promoter methylation was detected in clinicopathological features of the affected patients. A colorectal cancer (9). Subsequently, it has been found that significant difference was found in lymph node metastasis human p14 was also silenced by promoter hypermethylation (p=0.0359) on comparing the methylation status in in colorectal cancer (10). p14 interacts with the MDM2 colorectal cancers with lymph node metastasis to those protein and neutralizes the MDM2-mediated degradation without. There was also a significant gender difference of p53. Thus, p14 acts as a tumor suppressor via (p=0.0248) when the methylation status was compared. inhibition of p53 degradation. These studies indicated that Colorectal cancer without lymph node metastasis frequently p16 and p14 inactivation due to promoter methylation was exhibited aberrant methylation of the p16, p14, HLTF, important for colorectal tumorigenesis. We previously CDH13 and RUNX3 . examined the methylation status of these genes in 86 primary colorectal cancers using methylation-specific PCR There is now strong evidence that a series of genetic (MSP) (11). Aberrant promoter methylation of the p16 and alterations in both dominant oncogenes and tumor suppressor p14 genes was detected in 43 out of 86 (50 %) and 25 out genes are involved in the pathogenesis of human colorectal of 86 (29%) colorectal cancers, respectively. cancer. The activation of oncogenes such as the ras gene and The loss of expression of a -like transcription inactivation of tumor suppressor genes such as the APC and factor (HLTF), a SWI/SNF family member gene, p53 genes have been identified (1-3). In addition, we found accompanied by HLTF promoter methylation was reported that several other genes are related to the pathogenesis of in colon cancer (12). In this report, all colon cancer cell colorectal cancer (4, 5), and investigation of genetic changes is lines that lacked HLTF demonstrated important to clarify its tumorigenic pathway (6). methylation of CpG sites within the putative HLTF Several tumor suppressor genes contain CpG islands in promoter, while methylation was not detected in the HLTF- their promoters, prompting many investigations into the expressing cell lines. Moreover, HLTF methylation was role of methylation in silencing these genes. Many tumor detected in 25 out of 76 primary colon cancers, suggesting suppressor genes show evidence of methylation silencing, that HLTF is a common target for methylation and providing a potential new pathway for the deactivation of epigenetic gene silencing in colon cancer and qualifies as a tumor suppressor genes. At first, a tumor suppressor gene, potential colon cancer suppressor gene (13). Subsequently, it has also recently become clear that CDH13 (H-cadherin, T-cadherin) expression is frequently silenced by aberrant methylation in colorectal cancer and Correspondence to: Kenji Hibi, Department of Surgery, Seirei adenoma (14, 15). CDH13 encodes a protein belonging to Hospital, 56 Kawanayama-machi, Showa-ku, Nagoya 466-8633, the cadherin family of cell surface glycoproteins responsible Japan. Tel: +81-52-744-2245, Fax: +81-52-744-2255, e-mail: [email protected] for selective cell recognition and adhesion (16). Ubiquitous methylation of CDH13 in colorectal cancer and adenoma Key Words: Methylation, colorectal cancer, p16, p14, HLTF, indicated that such methylation occurs at an early stage in CDH13, RUNX3. the multistage process of oncogenesis.

0250-7005/2006 $2.00+.40 55 ANTICANCER RESEARCH 26: 55-58 (2006)

Recently, the loss of expression of RUNX3, a as described previously (11,13,15,18). The methylation transcription factor containing a highly conserved domain, status of the p16, p14, HLTF, CDH13 and RUNX3 genes designated as a ‘runt domain’, accompanied by RUNX3 gene were originally examined in 86, 86, 76, 84 and 92 methylation was reported in gastric cancer (17). In this colorectal cancer specimens, respectively. Fifty-nine report, between 45% and 60% of human gastric cancer cells specimens were tested for the methylation status of all 5 did not significantly express RUNX3 due to hemizygous genes. Aberrant methylation of the p16, p14, HLTF, deletion and hypermethylation of the RUNX3 gene. CDH13 and RUNX3 genes was detected in 34 (58%), 20 Tumorigenicity of human gastric cancer cell lines in nude (34%), 18 (31%), 22 (37%) and 22 (37%) of 59 colorectal mice was inversely related to their level of RUNX3 cancer specimens, respectively (Figure 1). All tumors also expression, suggesting that RUNX3 is a common target for exhibited non-methylation of each gene, which might be methylation and epigenetic gene silencing in gastric cancer the result of the contamination of the tumor specimens and qualifies as a potential gastric cancer suppressor gene. with non-tumor cells. Otherwise, it might be possible that Aberrant promoter methylation of the RUNX3 gene was these gene expressions had not been completely inhibited detected in 31 out of 92 (34%) colorectal cancers (18). in these cancer specimens. In this study, the combined methylation status of p16, The correlation between the methylation status and p14, HLTF, CDH13 and RUNX3 was examined in 59 clinicopathological data was analyzed (Table I). Colorectal resected primary colorectal cancers using methylation- cancers with more than 3 methylated genes were included specific PCR (MSP). To determine whether the methylation in the highly-methylated group (HMG), and colorectal status could be a marker for the malignancy of colorectal cancers with less than 2 methylated genes in the low- cancer, the methylation status was correlated with the methylated group (LMG). There was a significant difference clinicopathological features of the affected patients. in lymph node metastasis (p=0.0359) when the methylation status in colorectal cancers with and without lymph node Materials and Methods metastases was compared, indicating that the colorectal cancers without lymph node metastasis were highly- Sample collection and DNA preparation. Fifty-nine primary tumors methylated. This result suggested that the aberrant and corresponding normal tissues were collected consecutively at methylation of these genes occurred early in the Nagoya University Hospital, Japan, from colorectal cancer patients. carcinogenesis of colorectal cancer, although it was not All tissues were confirmed histologically. The collected samples often shown in advanced colorectal cancer with lymph node were stored immediately at –80ÆC until analysis. DNA was prepared, as described previously (19). metastasis. There was also a significant gender difference (p=0.0248) on comparing the methylation status, indicating Sodium bisulfite modification. One Ìg of genomic DNA extracted that the colorectal cancer were highly-methylated in males. from tumors and corresponding normal colorectal tissues was No other factors such as age, tumor size, extent of tumor, subjected to bisulfite treatment, as described previously (20). histology, or tumor site correlated with the number of Briefly, alkali-denatured DNA was modified by 2.1 M sodium methylated genes. bisulfite / 0.5 mM hydroquinone at pH 5.0. The bisulfite-reacted DNA was then treated by NaOH, purified using the Wizard DNA Clean-Up System® (Promega, Madison, WI, USA), precipitated Discussion with ethanol and resuspended in distilled water. Our results indicated that colorectal cancer without lymph Methylation-specific PCR. The bisulfite-treated DNA was amplified node metastasis exhibited a higher number of methylated with MSP. The primers and PCR conditions were described genes including p16, p14, HLTF, CDH13 and RUNX3. previously (7,10,12,14,17). Ten ml of each PCR product was loaded Therefore, the methylation of these genes is a low directly onto non-denaturing 8% polyacrylamide gels, stained with malignancy factor, as for lymph node metastasis. Shannon ethidium bromide and visualized under UV illumination. et al. also investigated the methylation status of the hMLH1, Statistical analysis. The possible association between the number of p16 and MDR1 genes in 58 colorectal cancers (21). They methylated genes and clinicopathological features was examined found methylation of the hMLH1, p16 and MDR1 genes in using the Chi-squared test or Student’s t-test. The computed p-values 23, 29 and 28% of the colorectal cancers, respectively. As a were two-tailed. Statistical significance was considered to be p<0.05. result, these 3 genes were significantly methylated in MSI+ colorectal cancers compared to MSI– cancers. Additionally, Results they were significantly associated with poor histological differentiation. Although the results of our studies differed The methylation status of the p16, p14, HLTF, CDH13 from theirs, Shannon et al.’s results coincide with ours in and RUNX3 genes were examined in colorectal cancer that the aberrant methylation was already manifest in the and corresponding normal tissue specimens using MSP, early stages of colorectal carcinogenesis (21).

56 Hibi and Nakao: Lymph Node Metastasis is Infrequent in Patients with Highly-methylated Colorectal Cancer

Table I. Clinicopathological features and methylation status in colorectal cancer patients.

Clinicopatho- Variables No. Methylation status p-value logical features of cases HMG LMG

Sex Male 33 12 21 0.0248a Female 26 3 23

Age 22-83 59 59±10c 63±10 0.325b

Maximum 15-100 mm 59 46±18 45±17 0.855b tumor size

Extent of tumor ≤pmd 17 2 15 0.106a pm< 45 13 29

Histology Welle 54 12 42 0.0853a Poorf 53 2

Tumor site C, A, Tg 18 4 14 0.706a D, S, Rh 41 11 30

Lymph node – 34 12 22 0.0359a metastasis + 25 3 22

Total 59 15 44

HMG, highly-methylated group; LMG, low-methylated group aChi-squared test, bStudent’s t-test, cmean±S.D. dproper muscle ewell- or moderately-differentiated adenocarcinoma fpoorly-differentiated or mucinous adenocarcinoma gC, cecum; A, ascending colon; T, transverse colon hD, descending colon; S, sigmoid colon; R, rectum Figure 1. Methylation status of p16, p14, HLTF, CDH13 and RUNX3 genes in 59 colorectal cancers. Methylated genes were shown as closed boxes. Colorectal cancers with more than 3 methylated genes were included in the highly-methylated group (HMG) and colorectal cancers with less than 2 methylated genes in the low-methylated group (LMG). process of carcinogenesis and not be related to the later spread of cancer cells to the lymph nodes, that would be mainly caused by p53 mutations. Taken together, it is possible that colorectal cancer with higher methylation and The CpG island methylator phenotype (CIMP) is no p53 mutations is less aggressive compared to cancer with characterized by the simultaneous methylation of multiple lower methylation and p53 mutations. CpG islands (9). Toyota et al. reported that more frequent Colorectal cancer, one of the most aggressive cancers, has a K-ras mutations were found in CIMP+ compared to CIMP– high incidence rate in most countries (23). To eliminate this colorectal cancers (22). This result supports the notion that fatal cancer, surgical operations and subsequent chemotherapy aberrant methylation occurs early in the carcinogenesis of and radiotherapy are performed. For this purpose, it is colorectal cancer because ras mutations have already important to seek new genetic and epigenetic alteration occurred in colorectal adenoma (6). On the other hand, p53 parameters to estimate the malignancy of the cancer. mutations were more frequently found in CIMP– compared to CIMP+ phenotypes (22). This might be the reason that References the aberrant methylation was not often shown in advanced colorectal cancers with lymph node metastasis because 1 Bos JL, Fearon ER, Hamilton SR, Verlaan-de Vries M, van colorectal cancers with p53 mutations were more likely to Boom JH, van der Eb AJ and Vogelstein B: Prevalence of ras metastasize (6). Therefore, the methylation of the p16, p14, gene mutations in human colorectal cancers. Nature 327: 293- HLTF, CDH13 and RUNX3 genes might simply be an early 297, 1987.

57 ANTICANCER RESEARCH 26: 55-58 (2006)

2 Nishisho I, Nakamura Y, Miyoshi Y, Miki Y, Ando H, Horii A, 14 Toyooka S, Toyooka KO, Harada K, Miyajima K, Makarla P, Koyama K, Utsunomiya J, Baba S and Hedge P: Mutations of Sathyanarayama UG, Yin J, Sato F, Shivapurkar N, Meltzer SJ 5q21 genes in FAP and colorectal cancer patients. and Gazdar AF: Aberrant methylation of the CDH13 (H- Science 253: 665-669, 1991. cadherin) promoter region in colorectal cancers and adenomas. 3 Baker SJ, Markowitz S, Fearon ER, Willson JK and Vogelstein Cancer Res 62: 3382-3386, 2002. B: Suppression of human colorectal carcinoma cell growth by 15 Hibi K, Nakayama H, Kodera Y, Ito K, Akiyama S and Nakao wild-type p53. Science 249: 912-915, 1990. A: CDH13 promoter region is specifically methylated in poorly 4 Hibi K, Nakamura H, Hirai A, Fujikake Y, Kasai Y, Akiyama S, differentiated colorectal cancer. Br J Cancer 90: 1030-1033, Ito K and Takagi H: Loss of H19 imprinting in esophageal 2004. cancer. Cancer Res 56: 480-482, 1996. 16 Takeichi M: Cadherin cell adhesion receptors as a morphogenetic 5 Hibi K, Taguchi M, Nakamura H, Hirai A, Fujikake Y, Matsui regulator. Science 251: 1451-1455, 1991. T, Kasai Y, Akiyama S, Ito K and Takagi H: Alternative 17 Li QL, Ito K, Sakakura C, Fukamachi H, Inoue K, Chi XZ, Lee splicing of the FHIT gene in colorectal cancer. Jpn J Cancer KY, Nomura S, Lee CW, Han SB, Kim HM, Kim WJ, Res 88: 385-388, 1997. Yamamoto H, Yamashita N, Yano T, Ikeda T, Itohara S, 6 Vogelstein B, Fearon ER, Hamilton SR, Kern SE, Preisinger Inazawa J, Abe T, Hagiwara A, Yamagishi H, Ooe A, Kaneda AC, Leppert M, Nakamura Y, White R, Smits AM and Bos JL: A, Sugimura T, Ushijima T, Bae SC and Ito Y: Causal Genetic alterations during colorectal-tumor development. N relationship between the loss of RUNX3 expression and gastric Engl J Med 319: 525-532, 1988. cancer. Cell 109: 113-124, 2002. 7 Herman JG, Graff JR, Myohanen S, Nelkin BD and Baylin SB: 18 Imamura Y, Hibi K, Koike M, Fujiwara M, Kodera Y, Ito K Methylation-specific PCR: a novel PCR assay for methylation and Nakao A: RUNX3 promoter region is specifically status of CpG islands. Proc Natl Acad Sci USA 93: 9821-9826, methylated in poorly-differentiated colorectal cancer. 1996. Anticancer Res 25: 2627-2630, 2005 8 Cairns P, Polascik TJ, Eby Y, Tokino K, Califano J, Merlo A, 19 Hibi K, Takahashi T, Yamakawa K, Ueda R, Sekido Y, Mao L, Herath J, Jenkins R, Westra W, Rutter JL, Buckler A, Ariyoshi Y, Suyama M, Takagi H and Nakamura Y: Three Gavielson E, Tockman M, Cho KR, Hedrick L, Bova GS, Issacs distinct regions involved in 3p deletion in human lung cancer. W, Schwab D and Sidransky D: Frequency of homozygous Oncogene 7: 445-449, 1992. deletion at p16/CDKN2 in primary human tumours. Nat Genet 20 Hibi K, Taguchi M, Nakayama H, Takase T, Kasai Y, Ito K, 11: 210-212, 1995. Akiyama S and Nakao A: Molecular detection of p16 promoter 9 Toyota M, Ahuja N, Ohe-Toyota M, Herman JG, Baylin SB methylation in the serum of patients with esophageal squamous and Issa J-P: CpG island methylator phenotype in colorectal cell carcinoma. Clin Cancer Res 7: 3135-3138, 2001. cancer. Proc Natl Acad Sci USA 96: 8681-8686, 1999. 21 Shannon BA and Iacopetta BJ: Methylation of the hMLH1, 10 Esteller M, Tortola S, Toyota M, Capella G, Peinado MA, p16, and MDR1 genes in colorectal carcinoma: associations with Baylin SB and Herman JG: Hypermethylation-associated clinicopathological features. Cancer Lett 167: 91-97, 2001. inactivation of p14ARF is independent of p16INK4a methylation 22 Toyota M, Ohe-Toyota M, Ahuja N and Issa J-P: Distinct and p53 mutational status. Cancer Res 60: 129-133, 2000. genetic profiles in colorectal tumors with or without the CpG 11 Hibi K, Nakayama H, Koike M, Kasai Y, Ito K, Akiyama S and island methylator phenotype. Proc Natl Acad Sci USA 97: 710- Nakao A: Colorectal cancers with both p16 and p14 methylation 715, 2000. show invasive characteristics. Jpn J Cancer Res 93: 883-887, 2002. 23 Greenlee RT, Murray T, Bolden S and Wingo PA: Cancer 12 Moinova HR, Chen W-D, Shen L, Smiraglia D, Olechnowicz J, statistics, 2000. CA Cancer J Clin 50: 7-33, 2000. Ravi L, Kasturi L, Myeroff L, Plass C, Parsons R, Minna J, Willson JK and Green SB: HLTF gene silencing in human colon cancer. Proc Natl Acad Sci USA 99: 4562-4567, 2002. 13 Hibi K, Nakayama H, Kanyama Y, Kodera Y, Ito K, Akiyama S and Nakao A: Methylation pattern of HLTF gene in digestive Received October 10, 2005 tract cancers. Int J Cancer 104: 433-436, 2003. Accepted October 20, 2005

58