期刊论文详细信息
BMC Cancer
The application of methylation specific electrophoresis (MSE) to DNA methylation analysis of the 5' CpG island of mucin in cancer cells
Seiya Yokoyama3  Sho Kitamoto3  Norishige Yamada3  Izumi Houjou3  Tamotsu Sugai4  Shin-ichi Nakamura1  Yoshifumi Arisaka5  Kyoichi Takaori2  Michiyo Higashi3  Suguru Yonezawa3 
[1] DPR Co., LTD. 4-10-53, Mitake, Morioka 020-0122, Japan
[2] Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Kyoto University Graduate School of Medicine, 54 Kawara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
[3] Department of Human Pathology, Field of Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
[4] Division of Pathology, Central Clinical Laboratory, School of Medicine, Iwate Medical University, Morioka, Japan
[5] Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
关键词: Cancer;    Pancreatic juice;    Colonic crypt;    Mucin;    Epigenetics;    DNA methylation pattern;   
Others  :  1080540
DOI  :  10.1186/1471-2407-12-67
 received in 2011-06-29, accepted in 2012-02-14,  发布年份 2012
PDF
【 摘 要 】

Background

Methylation of CpG sites in genomic DNA plays an important role in gene regulation and especially in gene silencing. We have reported mechanisms of epigenetic regulation for expression of mucins, which are markers of malignancy potential and early detection of human neoplasms. Epigenetic changes in promoter regions appear to be the first step in expression of mucins. Thus, detection of promoter methylation status is important for early diagnosis of cancer, monitoring of tumor behavior, and evaluating the response of tumors to targeted therapy. However, conventional analytical methods for DNA methylation require a large amount of DNA and have low sensitivity.

Methods

Here, we report a modified version of the bisulfite-DGGE (denaturing gradient gel electrophoresis) using a nested PCR approach. We designated this method as methylation specific electrophoresis (MSE). The MSE method is comprised of the following steps: (a) bisulfite treatment of genomic DNA, (b) amplification of the target DNA by a nested PCR approach and (c) applying to DGGE. To examine whether the MSE method is able to analyze DNA methylation of mucin genes in various samples, we apply it to DNA obtained from state cell lines, ethanol-fixed colonic crypts and human pancreatic juices.

Result

The MSE method greatly decreases the amount of input DNA. The lower detection limit for distinguishing different methylation status is < 0.1% and the detectable minimum amount of DNA is 20 pg, which can be obtained from only a few cells. We also show that MSE can be used for analysis of challenging samples such as human isolated colonic crypts or human pancreatic juices, from which only a small amount of DNA can be extracted.

Conclusions

The MSE method can provide a qualitative information of methylated sequence profile. The MSE method allows sensitive and specific analysis of the DNA methylation pattern of almost any block of multiple CpG sites. The MSE method can be applied to analysis of DNA methylation status in many different clinical samples, and this may facilitate identification of new risk markers.

【 授权许可】

   
2012 Yokoyama et al; licensee BioMed Central Ltd.

【 预 览 】
附件列表
Files Size Format View
20141203015625965.pdf 1244KB PDF download
Figure 5. 87KB Image download
Figure 4. 100KB Image download
Figure 3. 52KB Image download
Figure 2. 27KB Image download
Figure 1. 123KB Image download
【 图 表 】

Figure 1.

Figure 2.

Figure 3.

Figure 4.

Figure 5.

【 参考文献 】
  • [1]Kononen J, Bubendorf L, Kallioniemi A, Barlund M, Schraml P, Leighton S, Torhorst J, Mihatsch MJ, Sauter G, Kallioniemi OP: Tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med 1998, 4(7):844-847.
  • [2]Kitamoto S, Yamada N, Yokoyama S, Houjou I, Higashi M, Goto M, Batra SK, Yonezawa S: DNA methylation and histone H3-K9 modifications contribute to MUC17 expression. Glycobiology 2011, 21(2):247-256.
  • [3]Vincent A, Perrais M, Desseyn JL, Aubert JP, Pigny P, Van Seuningen I: Epigenetic regulation (DNA methylation, histone modifications) of the 11p15 mucin genes (MUC2, MUC5AC, MUC5B, MUC6) in epithelial cancer cells. Oncogene 2007, 26(45):6566-6576.
  • [4]Yamada N, Nishida Y, Yokoyama S, Tsutsumida H, Houjou I, Kitamoto S, Goto M, Higashi M, Yonezawa S: Expression of MUC5AC, an early marker of pancreatobiliary cancer, is regulated by DNA methylation in the distal promoter region in cancer cells. J Hepatobiliary Pancreat Sci 2010, 17(6):844-854.
  • [5]Yonezawa S, Goto M, Yamada N, Higashi M, Nomoto M: Expression profiles of MUC1, MUC2, and MUC4 mucins in human neoplasms and their relationship with biological behavior. Proteomics 2008, 8(16):3329-3341.
  • [6]Hamada T, Goto M, Tsutsumida H, Nomoto M, Higashi M, Sugai T, Nakamura S, Yonezawa S: Mapping of the methylation pattern of the MUC2 promoter in pancreatic cancer cell lines, using bisulfite genomic sequencing. Cancer Lett 2005, 227(2):175-184.
  • [7]Yamada N, Nishida Y, Tsutsumida H, Hamada T, Goto M, Higashi M, Nomoto M, Yonezawa S: MUC1 expression is regulated by DNA methylation and histone H3 lysine 9 modification in cancer cells. Cancer Res 2008, 68(8):2708-2716.
  • [8]Yamada N, Hamada T, Goto M, Tsutsumida H, Higashi M, Nomoto M, Yonezawa S: MUC2 expression is regulated by histone H3 modification and DNA methylation in pancreatic cancer. Int J Cancer 2006, 119(8):1850-1857.
  • [9]Yamada N, Nishida Y, Tsutsumida H, Goto M, Higashi M, Nomoto M, Yonezawa S: Promoter CpG methylation in cancer cells contributes to the regulation of MUC4. Br J Cancer 2009, 100(2):344-351.
  • [10]Kitamoto S, Yamada N, Yokoyama S, Houjou I, Higashi M, Yonezawa S: Promoter hypomethylation contributes to the expression of MUC3A in cancer cells. Biochem Biophys Res Commun 2010, 397(2):333-339.
  • [11]Ushijima T, Okochi-Takada E: Aberrant methylations in cancer cells: where do they come from? Cancer Sci 2005, 96(4):206-211.
  • [12]Ushijima T: Detection and interpretation of altered methylation patterns in cancer cells. Nat Rev Cancer 2005, 5(3):223-231.
  • [13]Bastian PJ, Palapattu GS, Lin X, Yegnasubramanian S, Mangold LA, Trock B, Eisenberger MA, Partin AW, Nelson WG: Preoperative serum DNA GSTP1 CpG island hypermethylation and the risk of early prostate-specific antigen recurrence following radical prostatectomy. Clin Cancer Res 2005, 11(11):4037-4043.
  • [14]Fujiwara K, Fujimoto N, Tabata M, Nishii K, Matsuo K, Hotta K, Kozuki T, Aoe M, Kiura K, Ueoka H, et al.: Identification of epigenetic aberrant promoter methylation in serum DNA is useful for early detection of lung cancer. Clin Cancer Res 2005, 11(3):1219-1225.
  • [15]Hiraki M, Kitajima Y, Sato S, Nakamura J, Hashiguchi K, Noshiro H, Miyazaki K: Aberrant gene methylation in the peritoneal fluid is a risk factor predicting peritoneal recurrence in gastric cancer. World J Gastroenterol 2010, 16(3):330-338.
  • [16]Klump B, Hsieh CJ, Dette S, Holzmann K, Kiebetalich R, Jung M, Sinn U, Ortner M, Porschen R, Gregor M: Promoter methylation of INK4a/ARF as detected in bile-significance for the differential diagnosis in biliary disease. Clin Cancer Res 2003, 9(5):1773-1778.
  • [17]Lenhard K, Bommer GT, Asutay S, Schauer R, Brabletz T, Goke B, Lamerz R, Kolligs FT: Analysis of promoter methylation in stool: a novel method for the detection of colorectal cancer. Clin Gastroenterol Hepatol 2005, 3(2):142-149.
  • [18]Matsubayashi H, Canto M, Sato N, Klein A, Abe T, Yamashita K, Yeo CJ, Kalloo A, Hruban R, Goggins M: DNA methylation alterations in the pancreatic juice of patients with suspected pancreatic disease. Cancer Res 2006, 66(2):1208-1217.
  • [19]Aggerholm A, Guldberg P, Hokland M, Hokland P: Extensive intra--and interindividual heterogeneity of p15INK4B methylation in acute myeloid leukemia. Cancer Res 1999, 59(2):436-441.
  • [20]Sugai T, Takahashi H, Habano W, Nakamura S, Sato K, Orii S, Suzuki K: Analysis of genetic alterations, classified according to their DNA ploidy pattern, in the progression of colorectal adenomas and early colorectal carcinomas. J Pathol 2003, 200(2):168-176.
  • [21]Nakamura S, Goto J, Kitayama M, Kino I: Application of the crypt-isolation technique to flow-cytometric analysis of DNA content in colorectal neoplasms. Gastroenterology 1994, 106(1):100-107.
  • [22]Habano W, Sugai T, Nakamura S, Yoshida T: A novel method for gene analysis of colorectal carcinomas using a crypt isolation technique. Lab Invest 1996, 74(5):933-940.
  • [23]Shinozuka N, Okada K, Torii T, Hirooka E, Tabuchi S, Aikawa K, Tawara H, Ozawa S, Ogawa N, Miyazawa M, et al.: Endoscopic pancreatic duct drainage and stenting for acute pancreatitis and pancreatic cyst and abscess. J Hepatobiliary Pancreat Surg 2007, 14(6):569-574.
  • [24]Schafer H, Servais P, Muyzer G: Successional changes in the genetic diversity of a marine bacterial assemblage during confinement. Arch Microbiol 2000, 173(2):138-145.
  • [25]Yonezawa S, Kitajima S, Higashi M, Osako M, Horinouchi M, Yokoyama S, Kitamoto S, Yamada N, Tamura Y, Shimizu T, et al.: A novel anti-MUC1 antibody against the MUC1 cytoplasmic tail domain: use in sensitive identification of poorly differentiated cells in adenocarcinoma of the stomach. Gastric Cancer 2012. published online DOI 10.1007/s10120-011-0125-2
  • [26]Li M, Chen WD, Papadopoulos N, Goodman SN, Bjerregaard NC, Laurberg S, Levin B, Juhl H, Arber N, Moinova H, et al.: Sensitive digital quantification of DNA methylation in clinical samples. Nat Biotechnol 2009, 27(9):858-863.
  文献评价指标  
  下载次数:49次 浏览次数:17次