Somatically acquired hypomethylation of IGF2 in breast and colorectal cancer

Yoko Ito, Thibaud Koessler, Ashraf E K Ibrahim, Sushma Rai, Sarah L Vowler, Sayeda Abu-Amero, Ana-Luisa Silva, Ana-Teresa Maia, Joanna E Huddleston, Santiago Uribe-Lewis, Kathryn Woodfine, Maja Jagodic, Raffaella Nativio, Alison Dunning, Gudrun Moore, Elena Klenova, Sheila Bingham, Paul D P Pharoah, James D Brenton, Stephan Beck, Manjinder S Sandhu, Adele Murrell, Yoko Ito, Thibaud Koessler, Ashraf E K Ibrahim, Sushma Rai, Sarah L Vowler, Sayeda Abu-Amero, Ana-Luisa Silva, Ana-Teresa Maia, Joanna E Huddleston, Santiago Uribe-Lewis, Kathryn Woodfine, Maja Jagodic, Raffaella Nativio, Alison Dunning, Gudrun Moore, Elena Klenova, Sheila Bingham, Paul D P Pharoah, James D Brenton, Stephan Beck, Manjinder S Sandhu, Adele Murrell

Abstract

The imprinted insulin-like growth factor 2 (IGF2) gene is expressed predominantly from the paternal allele. Loss of imprinting (LOI) associated with hypomethylation at the promoter proximal sequence (DMR0) of the IGF2 gene was proposed as a predisposing constitutive risk biomarker for colorectal cancer. We used pyrosequencing to assess whether IGF2 DMR0 methylation is either present constitutively prior to cancer or whether it is acquired tissue-specifically after the onset of cancer. DNA samples from tumour tissues and matched non-tumour tissues from 22 breast and 42 colorectal cancer patients as well as peripheral blood samples obtained from colorectal cancer patients [SEARCH (n=case 192, controls 96)], breast cancer patients [ABC (n=case 364, controls 96)] and the European Prospective Investigation of Cancer [EPIC-Norfolk (n=breast 228, colorectal 225, controls 895)] were analysed. The EPIC samples were collected 2-5 years prior to diagnosis of breast or colorectal cancer. IGF2 DMR0 methylation levels in tumours were lower than matched non-tumour tissue. Hypomethylation of DMR0 was detected in breast (33%) and colorectal (80%) tumour tissues with a higher frequency than LOI indicating that methylation levels are a better indicator of cancer than LOI. In the EPIC population, the prevalence of IGF2 DMR0 hypomethylation was 9.5% and this correlated with increased age not cancer risk. Thus, IGF2 DMR0 hypomethylation occurs as an acquired tissue-specific somatic event rather than a constitutive innate epimutation. These results indicate that IGF2 DMR0 hypomethylation has diagnostic potential for colon cancer rather than value as a surrogate biomarker for constitutive LOI.

Figures

Figure 1.
Figure 1.
Pyrosequencing assay to detect methylation levels at the IGF2 DMR0. (A) The human IGF2 gene showing positions of DMRs relative to exons and active promoters on the paternal allele. IGF2 promoters access enhancers downstream of H19 (ovals) when the CTCF binding site within the H19 DMR is methylated. IGF2 and H19 genes are 100 kb apart. The DMR0 region within the IGF2 has been reported to be hypomethylated in colorectal cancer. The expanded region shows the 26 CpGs within this region and the CpGs that we assayed for methylation by pyrosequencing. Our assay encompassed a 255 bp region, which includes six CpGs (CpG12–17 NCBI36 2125904–2126160). CpG15–17 were previously reported to be hypomethylated in colorectal patients with LOI at IGF2 (15). Arrows indicate position and direction of biotinylated, pyrosequencing (PSQ) and sequencing primers. Circles indicate individual CpGs on expanded DMR0. (B) Linearity of pyrosequencing assay: unmethylated template was obtained by PCR amplification of target DMR0 sequence prior to bisulphite conversion. After bisulphite conversion, the unmethylated template was serially diluted with PCR product obtained from bisulphite converted genomic DNA. At 100% genomic DNA, we have close to 50% methylation which is as expected for normal imprinting. (C) Pyrosequencing profiles for allele-specific methylation compared to total methylation. Total methylation levels reliably reflected the average of the two alleles. (D) Methylation diagrams for 12 cloned PCR products from bisulphite-treated DNA showing that alleles separate into methylated and unmethylated alleles and that methylation at CpG13 is not allele-specific. Filled and open circles denote methylated and unmethylated CpGs.
Figure 2.
Figure 2.
Distribution of average CpG methylation across IGF2 DMR0 and population frequency of methylation in a cohort of peripheral blood and placentae. (A) Graph showing average methylation levels of each CpG across the IGF2 DMR0 in peripheral blood, normal colon, normal breast and term placentae samples. (B) Cumulative frequency distribution of average methylation levels for CpG12, 14–17 in the IGF2 DMR0 in placentae and peripheral blood. At the 90% percentile average methylation levels for the DMR0 was 48%; at the 10% percentile, the average methylation level was less than 35%.
Figure 3.
Figure 3.
IGF2 DMR0 methylation levels in colorectal and breast cancers compared to normal tissue from same patients and non-cancer patients: prevalence of hypomethylation in breast and colorectal cancer tumours. (A) DMR0 methylation levels in DNA from colon tissue from patients with no cancer (Colon NC), and colorectal cancer patients' tumour tissue (CRC-T) and matched control biopsies of non-tumour tissues (CRC-NT). Tumours have significantly lower methylation than non-tumour tissue from same patients and this is lower than in colons from patients without cancer. (B) Line plots showing individual data comparisons between colorectal tumour (CRC-T) and matched control biopsies of non-tumour tissues (CRC NT). (C) LOI relative to IGF2 DMR0 methylation in colorectal cancer tissue: colorectal cancer patients with LOI have lower methylation in colon tissue than patients without LOI. Colorectal tumours (CRC-T) have lower methylation than non-tumour tissues (CRC NT) regardless of LOI. (D) DMR0 methylation levels in DNA from breast tissue from individuals with no cancer (Breast NC) and breast cancer patients' tumours (BC-T) and matched control biopsies of non-tumour tissue (BC-NT). Breast cancer tumour tissues have significantly lower methylation levels than non-tumour tissue. Non-tumour tissue from breast cancer patients has a wider variation of methylation levels than tissue from non-cancer patients, but median levels are not significantly different. (E) Line plots showing individual data comparisons between breast tumour (BC-T) and matched control biopsies of non-tumour tissues (BC NT). (Box and whisker plots represent Median and IQR with max and min values. P-values calculated by Wilcoxon Rank sum test.)

References

    1. Reik W., Walter J. Genomic imprinting: parental influence on the genome. Nat. Rev. Genet. 2001;2:21–32.
    1. van Roozendaal C.E., Gillis A.J., Klijn J.G., van Ooijen B., Claassen C.J., Eggermont A.M., Henzen-Logmans S.C., Oosterhuis J.W., Foekens J.A., Looijenga L.H. Loss of imprinting of IGF2 and not H19 in breast cancer, adjacent normal tissue and derived fibroblast cultures. FEBS Lett. 1998;437:107–111.
    1. Kim K.S., Lee Y.I. Biallelic expression of the H19 and IGF2 genes in hepatocellular carcinoma. Cancer Lett. 1997;119:143–148.
    1. Kim H.T., Choi B.H., Niikawa N., Lee T.S., Chang S.I. Frequent loss of imprinting of the H19 and IGF-II genes in ovarian tumors. Am. J. Med. Genet. 1998;80:391–395.
    1. Murphy S.K., Huang Z., Wen Y., Spillman M.A., Whitaker R.S., Simel L.R., Nichols T.D., Marks J.R., Berchuck A. Frequent IGF2/H19 domain epigenetic alterations and elevated IGF2 expression in epithelial ovarian cancer. Mol. Cancer Res. 2006;4:283–292.
    1. Byun H.M., Wong H.L., Birnstein E.A., Wolff E.M., Liang G., Yang A.S. Examination of IGF2 and H19 loss of imprinting in bladder cancer. Cancer Res. 2007;67:10753–10758.
    1. Liou J.-M., Wu M.-S., Lin J.-T., Wang H.-P., Huang S.-P., Chiu H.-M., Lee Y.-C., Lin Y.-B., Shun C.-T., Liang T. Loss of imprinting of insulin-like growth factor II is associated with increased risk of proximal colon cancer. Eur. J. Cancer. 2007;43:1276–1282.
    1. Sasaki J., Konishi F., Kawamura Y.J., Kai T., Takata O., Tsukamoto T. Clinicopathological characteristics of colorectal cancers with loss of imprinting of insulin-like growth factor 2. Int. J. Cancer. 2006;119:80–83.
    1. Maenaka S., Hikichi T., Imai M.A., Minamoto T., Kawahara E. Loss of imprinting in IGF2 in colorectal carcinoma assessed by microdissection. Oncol. Rep. 2006;15:791–795.
    1. Kaneda A., Feinberg A.P. Loss of imprinting of IGF2: a common epigenetic modifier of intestinal tumor risk. Cancer Res. 2005;65:11236–11240.
    1. Woodson K., Flood A., Green L., Tangrea J.A., Hanson J., Cash B., Schatzkin A., Schoenfeld P. Loss of insulin-like growth factor-II imprinting and the presence of screen-detected colorectal adenomas in women. J. Natl Cancer Inst. 2004;96:407–410.
    1. Sasamoto H., Nagasaka T., Notohara K., Ozaki K., Isozaki H., Tanaka N., Matsubara N. Allele-specific methylation analysis on upstream promoter region of H19 by methylation-specific PCR with confronting two-pair primers. Int. J. Oncol. 2004;25:1273–1278.
    1. Cruz-Correa M., Cui H., Giardiello F.M., Powe N.R., Hylind L., Robinson A., Hutcheon D.F., Kafonek D.R., Brandenburg S., Wu Y., et al. Loss of imprinting of insulin growth factor II gene: a potential heritable biomarker for colon neoplasia predisposition. Gastroenterology. 2004;126:964–970.
    1. Cui H., Cruz-Correa M., Giardiello F.M., Hutcheon D.F., Kafonek D.R., Brandenburg S., Wu Y., He X., Powe N.R., Feinberg A.P. Loss of IGF2 imprinting: a potential marker of colorectal cancer risk. Science. 2003;299:1753–1755.
    1. Cui H., Onyango P., Brandenburg S., Wu Y., Hsieh C.L., Feinberg A.P. Loss of imprinting in colorectal cancer linked to hypomethylation of H19 and IGF2. Cancer Res. 2002;62:6442–6446.
    1. Arslan A.A., Gold L.I., Mittal K., Suen T.-C., Belitskaya-Levy I., Tang M.-S., Toniolo P. Gene expression studies provide clues to the pathogenesis of uterine leiomyoma: new evidence and a systematic review. Hum. Reprod. 2005;20:852–863.
    1. Bertucci F., Salas S., Eysteries S., Nasser V., Finetti P., Ginestier C., Charafe-Jauffret E., Loriod B., Bachelart L., Montfort J., et al. Gene expression profiling of colon cancer by DNA microarrays and correlation with histoclinical parameters. Oncogene. 2004;23:1377–1391.
    1. Giordano T.J., Thomas D.G., Kuick R., Lizyness M., Misek D.E., Smith A.L., Sanders D., Aljundi R.T., Gauger P.G., Thompson N.W., et al. Distinct transcriptional profiles of adrenocortical tumors uncovered by DNA microarray analysis. Am. J. Pathol. 2003;162:521–531.
    1. Schwienbacher C., Angioni A., Scelfo R., Veronese A., Calin G.A., Massazza G., Hatada I., Barbanti-Brodano G., Negrini M. Abnormal RNA expression of 11p15 imprinted genes and kidney developmental genes in Wilms’ tumor. Cancer Res. 2000;60:1521–1525.
    1. Cooper W.N., Luharia A., Evans G.A., Raza H., Haire A.C., Grundy R., Bowdin S.C., Riccio A., Sebastio G., Bliek J., et al. Molecular subtypes and phenotypic expression of Beckwith-Wiedemann syndrome. Eur. J. Hum. Genet. 2005;13:1025–1032.
    1. Reik W., Constancia M., Dean W., Davies K., Bowden L., Murrell A., Feil R., Walter J., Kelsey G. Igf2 imprinting in development and disease. Int. J. Dev. Biol. 2000;44:145–150.
    1. Weksberg R., Nishikawa J., Caluseriu O., Fei Y.L., Shuman C., Wei C., Steele L., Cameron J., Smith A., Ambus I., et al. Tumor development in the Beckwith-Wiedemann syndrome is associated with a variety of constitutional molecular 11p15 alterations including imprinting defects of KCNQ1OT1. Hum. Mol. Genet. 2001;10:2989–3000.
    1. Thorvaldsen J.L., Duran K.L., Bartolomei M.S. Deletion of the H19 differentially methylated domain results in loss of imprinted expression of H19 and Igf2. Genes Dev. 1998;12:3693–3702.
    1. Constancia M., Dean W., Lopes S., Moore T., Kelsey G., Reik W. Deletion of a silencer element in Igf2 results in loss of imprinting independent of H19. Nat. Genet. 2000;26:203–206.
    1. Lopes S., Lewis A., Hajkova P., Dean W., Oswald J., Forne T., Murrell A., Constancia M., Bartolomei M., Walter J., et al. Epigenetic modifications in an imprinting cluster are controlled by a hierarchy of DMRs suggesting long-range chromatin interactions. Hum. Mol. Genet. 2003;12:295–305.
    1. Monk D., Sanches R., Arnaud P., Apostolidou S., Hills F.A., Abu-Amero S., Murrell A., Friess H., Reik W., Stanier P., et al. Imprinting of IGF2 P0 transcript and novel alternatively spliced INS-IGF2 isoforms show differences between mouse and human. Hum. Mol. Genet. 2006;15:1259–1269.
    1. Murrell A., Heeson S., Bowden L., Constancia M., Dean W., Kelsey G., Reik W. An intragenic methylated region in the imprinted Igf2 gene augments transcription. EMBO Rep. 2001;2:1101–1106.
    1. Bell A.C., Felsenfeld G. Methylation of a CTCF-dependent boundary controls imprinted expression of the Igf2 gene. Nature. 2000;405:482–485.
    1. Hark A.T., Schoenherr C.J., Katz D.J., Ingram R.S., Levorse J.M., Tilghman S.M. CTCF mediates methylation-sensitive enhancer-blocking activity at the H19/Igf2 locus. Nature. 2000;405:486–489.
    1. Kanduri C., Pant V., Loukinov D., Pugacheva E., Qi C.F., Wolffe A., Ohlsson R., Lobanenkov V.V. Functional association of CTCF with the insulator upstream of the H19 gene is parent of origin-specific and methylation-sensitive. Curr. Biol. 2000;10:853–856.
    1. Szabo P., Tang S.H., Rentsendorj A., Pfeifer G.P., Mann J.R. Maternal-specific footprints at putative CTCF sites in the H19 imprinting control region give evidence for insulator function. Curr. Biol. 2000;10:607–610.
    1. Lewis A., Murrell A. Genomic imprinting: CTCF protects the boundaries. Curr. Biol. 2004;14:R284–R286.
    1. Kurukuti S., Tiwari V.K., Tavoosidana G., Pugacheva E., Murrell A., Zhao Z., Lobanenkov V., Reik W., Ohlsson R. CTCF binding at the H19 imprinting control region mediates maternally inherited higher-order chromatin conformation to restrict enhancer access to Igf2. Proc. Natl Acad. Sci. USA. 2006;103:10684–10689.
    1. Murrell A., Heeson S., Reik W. Interaction between differentially methylated regions partitions the imprinted genes Igf2 and H19 into parent-specific chromatin loops. Nat. Genet. 2004;36:889–893.
    1. Moore T., Constancia M., Zubair M., Bailleul B., Feil R., Sasaki H., Reik W. Multiple imprinted sense and antisense transcripts, differential methylation and tandem repeats in a putative imprinting control region upstream of mouse Igf2. Proc. Natl Acad. Sci. USA. 1997;94:12509–12514.
    1. Murrell A., Ito Y., Verde G., Huddleston J., Woodfine K., Silengo M., Spreafico F., Perotti D., De Crescenzo A., Sparago A., et al. Distinct methylation changes at the IGF2-H19 locus in congenital growth disorders and cancer. PlosONE. 2008;3:e1849.
    1. Sullivan M.J., Taniguchi T., Jhee A., Kerr N., Reeve A.E. Relaxation of IGF2 imprinting in Wilms tumours associated with specific changes in IGF2 methylation. Oncogene. 1999;18:7527–7534.
    1. Xu W., Fan H., He X., Zhang J., Xie W. LOI of IGF2 is associated with esophageal cancer and linked to methylation status of IGF2 DMR. J. Exp. Clin. Cancer Res. 2006;25:543–547.
    1. Ulaner G.A., Vu T.H., Li T., Hu J.F., Yao X.M., Yang Y., Gorlick R., Meyers P., Healey J., Ladanyi M., et al. Loss of imprinting of IGF2 and H19 in osteosarcoma is accompanied by reciprocal methylation changes of a CTCF-binding site. Hum. Mol. Genet. 2003;12:535–549.
    1. Day N., Oakes S., Luben R., Khaw K.T., Bingham S., Welch A., Wareham N. EPIC-Norfolk: study design and characteristics of the cohort. European Prospective Investigation of Cancer. Br. J. Cancer. 1999;80(Suppl. 1):95–103.
    1. Colella S., Shen L., Baggerly K.A., Issa J.P., Krahe R. Sensitive and quantitative universal Pyrosequencing methylation analysis of CpG sites. Biotechniques. 2003;35:146–150.
    1. Tost J., Gut I.G. Analysis of gene-specific DNA methylation patterns by pyrosequencing technology. Methods Mol. Biol. 2007;373:89–102.
    1. Cebrian A., Lesueur F., Martin S., Leyland J., Ahmed S., Luccarini C., Smith P.L., Luben R., Whittaker J., Pharoah P.D., et al. Polymorphisms in the initiators of RET (rearranged during transfection) signaling pathway and susceptibility to sporadic medullary thyroid carcinoma. J. Clin. Endocrinol. Metab. 2005;90:6268–6274.
    1. Easton D.F., Bishop D.T., Ford D., Crockford G.P. Genetic linkage analysis in familial breast and ovarian cancer: results from 214 families. The Breast Cancer Linkage Consortium. Am. J. Hum. Genet. 1993;52:678–701.
    1. Linhart H.G., Lin H., Yamada Y., Moran E., Steine E.J., Gokhale S., Lo G., Cantu E., Ehrich M., He T., et al. Dnmt3b promotes tumorigenesis in vivo by gene-specific de novo methylation and transcriptional silencing. Genes Dev. 2007;21:3110–3122.
    1. Barreto G., Schafer A., Marhold J., Stach D., Swaminathan S.K., Handa V., Doderlein G., Maltry N., Wu W., Lyko F., et al. Gadd45a promotes epigenetic gene activation by repair-mediated DNA demethylation. Nature. 2007;445:671–675.
    1. Morgan H.D., Dean W., Coker H.A., Reik W., Petersen-Mahrt S.K. Activation-induced cytidine deaminase deaminates 5-methylcytosine in DNA and is expressed in pluripotent tissues: implications for epigenetic reprogramming. J. Biol. Chem. 2004;279:52353–52360.
    1. Ahuja N., Li Q., Mohan A.L., Baylin S.B., Issa J.P. Aging and DNA methylation in colorectal mucosa and cancer. Cancer Res. 1998;58:5489–5494.
    1. Heijmans B.T., Kremer D., Tobi E.W., Boomsma D.I., Slagboom P.E. Heritable rather than age-related environmental and stochastic factors dominate variation in DNA methylation of the human IGF2/H19 locus. Hum. Mol. Genet. 2007;16:547–554.
    1. Chen W.-D., Han Z.J., Skoletsky J., Olson J., Sah J., Myeroff L., Platzer P., Lu S., Dawson D., Willis J., et al. Detection in fecal DNA of colon cancer-specific methylation of the nonexpressed vimentin gene. J. Natl Cancer Inst. 2005;97:1124–1132.
    1. Kann L., Han J., Ahlquist D., Levin T., Rex D., Whitney D., Markowitz S., Shuber A. Improved marker combination for detection of de novo genetic variation and aberrant DNA in colorectal neoplasia. Clin. Chem. 2006;52:2299–2302.
    1. Koinuma K., Kaneda R., Toyota M., Yamashita Y., Takada S., Choi Y.L., Wada T., Okada M., Konishi F., Nagai H., et al. Screening for genomic fragments that are methylated specifically in colorectal carcinoma with a methylated MLH1 promoter. Carcinogenesis. 2005;26:2078–2085.
    1. Zou H., Harrington J.J., Shire A.M., Rego R.L., Wang L., Campbell M.E., Oberg A.L., Ahlquist D.A. Highly methylated genes in colorectal neoplasia: implications for screening. Cancer Epidemiol. Biomarkers Prev. 2007;16:2686–2696.
    1. Kronborg O., Fenger C., Olsen J., Jorgensen O.D., Sondergaard O. Randomised study of screening for colorectal cancer with faecal-occult-blood test. Lancet. 1996;348:1467–1471.
    1. Hardcastle J.D., Chamberlain J.O., Robinson M.H., Moss S.M., Amar S.S., Balfour T.W., James P.D., Mangham C.M. Randomised controlled trial of faecal-occult-blood screening for colorectal cancer. Lancet. 1996;348:1472–1477.
    1. Alexander F., Weller D. Evaluation of the UK Colorectal Cancer Screening Pilot. Final Report. 2003 .
    1. Sakatani T., Wei M., Katoh M., Okita C., Wada D., Mitsuya K., Meguro M., Ikeguchi M., Ito H., Tycko B., et al. Epigenetic heterogeneity at imprinted loci in normal populations. Biochem. Biophys. Res. Commun. 2001;283:1124–1130.
    1. Sandovici I., Leppert M., Hawk P.R., Suarez A., Linares Y., Sapienza C. Familial aggregation of abnormal methylation of parental alleles at the IGF2/H19 and IGF2R differentially methylated regions. Hum. Mol. Genet. 2003;12:1569–1578.
    1. Clopper C.J., Pearson E. The use of confidence or fiducial limits illustrated in the case of the binomial. Biometrika. 1934;26:404–413.

Source: PubMed

3
订阅