Open Access

Epimutation and cancer: a new carcinogenic mechanism of Lynch syndrome (Review)

  • Authors:
    • Kouji Banno
    • Iori Kisu
    • Megumi Yanokura
    • Kosuke Tsuji
    • Kenta Masuda
    • Arisa Ueki
    • Yusuke Kobayashi
    • Wataru Yamagami
    • Hiroyuki Nomura
    • Eiichiro Tominaga
    • Nobuyuki Susumu
    • Daisuke Aoki
  • View Affiliations

  • Published online on: June 25, 2012     https://doi.org/10.3892/ijo.2012.1528
  • Pages: 793-797
  • Copyright: © Banno et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 3.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Epimutation is defined as abnormal transcriptional repression of active genes and/or abnormal activation of usually repressed genes caused by errors in epigenetic gene repression. Epimutation arises in somatic cells and the germline, and constitutional epimutation may also occur. Epimutation is the first step of tumorigenesis and can be a direct cause of carcinogenesis. Cancers associated with epimutation include Lynch syndrome (hereditary non-polyposis colorectal cancer, HNPCC), chronic lymphocytic leukemia, breast cancer and ovarian cancer. Epimutation has been shown for many tumor suppressor genes, including RB, VHL, hMLH1, APC and BRCA1, in sporadic cancers. Methylation has recently been shown in DNA from normal tissues and peripheral blood in cases of sporadic colorectal cancer and many studies show constitutive epimutation in cancers. Epimutation of DNA mismatch repair (MMR) genes (BRCA1, hMLH1 and hMSH2) involved in development familial cancers has also been found. These results have led to a focus on epimutation as a novel oncogenic mechanism.

Contents

Introduction

What is epimutation?

Germline epimutation and disease

Epimutation and cancer

Epimutation and Lynch syndrome

Conclusion

Introduction

Epimutation includes epigenetic repression of active genes for which expression is not suppressed, or epigenetic activation of genes for which expression is normally repressed. Disease susceptibility is generally determined not only by DNA sequence mutation, but also by changes in the activities of genes and chromosomal regions. Epigenetic repression has attracted attention as a mechanism underlying these changes in activities. Epigenetic modification causes repression of gene activity and is required for cell division and histogenesis. Phenotypic variation in genetically identical cells is caused by differences in epigenetic profiles. In addition, epimutation may be the first step of tumorigenesis in specific cancers and this provides a direct link to predisposition for cancer. In this article, we review the relationship between epimutation and cancer.

What is epimutation?

Epimutation was first used as a term for epigenetic changes, including methylation, that have no effect on DNA sequence (1). It is currently defined as abnormal transcriptional repression in active genes and/or abnormal activation of usually repressed genes caused by errors in epigenetic gene repression (13). Epimutation affects one or both alleles of a gene. By preventing transcription of the affected allele, epimutation effectively reduces the level of the gene product. Tumor cells are a typical consequence of epimutation in mammals. Epimutation in cancer generally occurs in somatic cells and manifests as tumor progression. Different kinds of epimutation occur in many cancers, but epimutation is particularly common in tumor suppressor genes. Epimutation may also arise in the germline, and constitutional epimutation has been shown, but it is thought that epigenetic mutation generally occurs stochastically in genes. Epimutation is also found in non-cancerous tissues in the stage prior to tumorigenesis. Methylation in normal tissues is referred to as a field effect and has been shown to have a relationship with clonally-related malignant cells in tissues.

Germline epimutation is defined as an event that occurs in the genes of germ cells and is maintained in fertilization and embryonic development, resulting in persistence in all adult somatic cells. In this manner, epigenetic characteristics are transmitted through generations, and therefore, individuals with the same germline epimutation have a similar oncogenic risk. However, epimutation is not always inherited and epimutation has been shown to have a hereditary form that does not follow Mendelian inheritance (47). Epimutation has also been shown to disappear in spermatogenesis (8). Previous studies have indicated epimutation is inherited from the mother alone, which suggests that epimutation is unlikely to disappear during oogenesis (7,8). Some somatic aberrations of genomic imprinting are also considered to be germline epimutations (3). Constitutional epimutation is defined as that occurring in the stage of early embryonic development and in all human tissues in the stage prior to triploblastic differentiation. Epimutation is mosaic in somatic cells and is not found in all cells, with no study confirming transmission from the previous generation. Epimutation is also the first step of tumorigenesis and can be a direct cause of carcinogenesis.

Germline epimutation and disease

Epimutation is involved in genomic imprinting (Table I), in which a gene inherited from one parent is selectively expressed and genetic disease develops if this gene has a deficiency, even if the other allele is normal. Phenotypes specific to genomic imprinting are regulated by imprinting control centers (ICs). ICs are short sequences in an imprinted gene. Only one allele of an IC is methylated and transcribed, permitting regulation of imprinting. Various genetic changes including microdeletion have been found in ICs and these are considered to be caused by epimutation in Angelman syndrome (AS), Prader-Willi syndrome (PWS) and Beckwith-Wiedemann syndrome (BWS). BWS is a congenital disorder with a high risk of embryonal tumors such as Wilms’ tumor, hepatoblastoma and rhabdomyosarcoma. Chromosome 11p15.5 has been identified as the disease locus. The 11p15.5 locus contains two imprinted domains, the cyclin-dependent kinase inhibitor 1C/KCNQ1 opposite antisense transcript 1 (CDKN1C/KCNQ1OT1) and insulin-like growth factor 2 (IGF2)/H19 domains, through which expression of imprinted genes surrounding the imprinting regulation regions is controlled. In 30–50% of cases of BWS, expression of CDKN1C decreases due to DNA hypomethylation, while expression of IGF2 increases due to DNA hypermethylation of the IGF2/H19 domain (9). Silver-Russell syndrome (SRS) is a disorder characterized by intrauterine growth retardation and severe postnatal growth retardation, and is caused by epimutation of the H19 gene in the 11p15.5 region (10). Thus, these diseases develop due to abnormality of the respective ICs.

Table I

Epimutation and disease.

Table I

Epimutation and disease.

Gene nameEpimutation typeDiseaseReference
hMLH1Germline, constitutionalLynch syndrome(4,24,43)
hMSH2GermlineLynch syndrome(28)
DAPK1UnknownB-cell CLL(29)
HBA2Unknownα-thalassemia(12)
BRCA2ConstitutionalSporadic breast cancer(40)
KIP2/LIT1UnknownBeckwith-Wiedemann syndrome(9)
IGF2UnknownBeckwith-Wiedemann syndrome(9)
H19UnknownSilver-Russell syndrome(10)

Epimutation is also involved in onset of α-thalassemia. Epimutations occur due to variations such as genomic insertion and deletion, and changes in the length of tandem repeats, which are referred to as copy number variation (CNV) (11). In α-thalassemia, the deletion locus of the LUC7-like (LUC7L) gene is co-located with Hemoglobin α 2 (HBA2), an α globin gene, resulting in methylation of the HBA2 promoter (12).

Epimutation and cancer

Studies of familial cancer have shown that specific gene groups inactivated by mutation cause a predisposition to cancer. The tumor suppressor gene retinoblastoma (RB) is a disease gene for hereditary cancer that was initially identified through mutations found in cases of retinoblastoma (13). Subsequently, Nishisho et al found mutations in adenomatous polyposis coli (APC) in familial adenomatous polyposis (14), and Hussussian et al identified mutations in cyclin-dependent kinase inhibitor 2A (CDKN2A) in familial melanoma (15). The mutations in different tumor suppressor genes indicate various underlying mechanisms as causes of cancer. The DNA mismatch repair genes breast cancer susceptibility gene 1 (BRCA1), human MutL homologue 1 (hMLH1) and human MutS homologue 2 (hMSH2) have also been linked to a predisposition for familial cancer. Inactivation of genes induced by mutation in hereditary cancer is recessive inheritance and most carriers have no abnormal phenotype. However, mutation, inactivation and loss of heterozygosity are likely to occur in other normal alleles, and consequently, cancer morbidity is frequently dominantly inherited.

RB methylation was identified as the first example of epimutation in a cancer-related gene (16,17). Subsequently, many other oncogenes, including Von Hippel-Lindau (VHL), hMLH1, APC and BRCA1, have been shown to be methylated in sporadic cancers (1820). Among DNA mismatch genes, hMLH1 and hMSH2 methylation causes a predisposition for endometrial, small intestine and ovarian cancers, in addition to colon cancer. hMLH1 and hMSH2 encode mismatch repair proteins and inactivation of these genes causes microsatellite instability (MSI) in tumor cells (21). hMLH1 is also methylated in cases of sporadic colorectal cancer (19) with the same phenotype of mismatch repair defect and clinicopathologic characteristics similar to those of hereditary tumors. Such sporadic colorectal cancer also has a close relationship with cancer with a CpG island methylator phenotype (CIMP). CIMP-positive cancers frequently have methylation in CpG islands in a specific promoter region (22). These cancers usually occur in the ascending colon and are particularly common in elderly women.

Gazzoli et al first found that hMLH1 may be methylated in peripheral blood, as well as in tumor cells, in patients with colorectal cancer (23). In a study of 14 patients with Lynch syndrome with MSI, hypermethylated hMLH1 was found in normal blood DNA in one 25-year-old female patient (23). Allele methylation in tissues derived from an embryologically discrete germ layer suggests the presence of a constitutional or germline methylation pattern. Since no mutation was found in specimens of her parents, hereditary evidence for epimutation was not obtained; however, it is of interest that methylation occurred in a young patient. It has also been shown that patients with colorectal cancer with methylation in one allele of hMLH1 have constitutional methylation (24). Suter et al showed hMLH1 methylation in a phenotype derived from a triploblastic origin in 2 patients with colorectal cancer (24). Tissues of their parents were not examined, but no methylation was found in tissues in 4 of their 5 children.

Much controversy exists regarding constitutional epimutation; i.e., whether this is transmitted from a mother or father, or occurs de novo in early embryonic development. Miyakura et al showed that complete methylation in the hMLH1 promoter region played an important role in hMLH1 inactivation in patients with sporadic colorectal cancer with high MSI (25). This methylation occurred in both alleles and methylation in the upper hMLH1 promoter region was also found in normal colonic mucosa adjacent to cancer tissue in one-third of patients with colorectal cancer associated with complete methylation (26). Miyakura et al subsequently examined methylation in the hMLH1 promoter region of peripheral blood lymphocytes (PBLs) in 30 patients with early-onset sporadic colorectal cancer or multiple primary cancer. Four of these patients (2 with early-onset sporadic colon cancer, 1 with colon cancer and 1 with multiple cancer including endometrial cancer) had complete methylation in the hMLH1 promoter region in PBLs (27). Methylation was found only in one allele. No methylation was detected in PBLs of the sister of a patient with early-onset sporadic colorectal cancer. MSI was found in all patients and methylation was also detected in normal tissues of the large intestine, digestive mucosa, endometrium and bone marrow of 3 patients. It is of interest that loss of heterozygosity (LOH) in both alleles of hMLH1, loss of G alleles in somatic cells of the hMLH1 locus and methylation of both alleles of hMLH1 were detected in cases of colon cancer. This finding is consistent with the mechanism of carcinogenesis based on germline epimutation proposed by Suter et al based on Knudson’s ‘two hit’ hypothesis (Fig. 1) (24).

Epimutation is not always inherited and has also been shown to have a hereditary form that does not follow Mendelian inheritance (47). Epimutation disappears in spermatogenesis, and may by inherited from the mother alone, suggesting that disappearance during oogenesis is unlikely (7,8). In a cohort study of 160 patients with Lynch syndrome without germline mismatch repair gene mutations, constitutional methylation of hMLH1 was found in only one patient. No methylation of hMLH1 was detected in this patient’s parents or siblings. This finding suggests that clinicopathologic characteristics are a better indicator than family history if constitutional epimutation of a tumor suppressor gene is identified in a cancer patient (4).

Epimutation is known to have a relationship with chronic lymphocytic leukemia (CLL). In CLL, apoptosis in leukemia cells is inhibited by enhanced production of B-cell lymphoma 2 (BCL2) and methylation of the promoter region of death-associated protein kinase 1 (DAPK1) (29). DAPK1 was identified as a familial tumor suppressor gene and methylation is found in the DAPK1 promoter region in CLL (29). Expression of homeobox B7 (HOXB7) proteins connected with the upper promoter region increases due to methylation, and 75% of DAPK1 genes are downregulated. Inactivation of DAPK1 due to methylation is a cause of both familial and sporadic CLL, and hypomethylation of DAPK1 has been found in peripheral blood mononuclear cells (PBMCs) of healthy individuals (30); however, the relationship with hypomethylation in CLL is unclear.

Many studies have examined the relationship between breast cancer and BRCA1 mutation. Armes and Venter (31) and Lakhani et al(32) showed that breast cancer in patients with germline BRCA1 mutations had histological characteristics including a high mitotic index and lymphatic infiltration. This form is currently referred to as the basal-like type. Foulkes et al showed that 80–90% of cancers that developed in carriers of germline mutations in BRCA1 were of the basal-like type (32). Subsequently, methylation in the promoter region of BRCA1 has been found in sporadic breast cancer (34). The relationship between BRCA1 mutation and methylation has been examined based on the hypothesis that a sporadic tumor with BRCA1 methylation should be similar to a tumor with BRCA1 mutation, if BRCA1 methylation causes tumorigenesis. Catteau and Morris showed that sporadic tumors with BRCA1 methylation had pathological characteristics similar to those of hereditary breast cancer (35) and Hedenfalk et al found that the phenotypes of the cancers were similar to each other (34). Many studies have shown that tumors with BRCA1 methylation have high-grade, estrogen receptor-negative and progesterone receptor-negative characteristics and a high incidence in young women (36). These characteristics are referred to as BRCA1-like. Esteller et al detected BRCA1 methylation in 67% of medullary carcinomas and 55% of mucinous adenocarcinomas, and showed that this phenotype was frequently found in families with BRCA1 mutation (34). Snell et al found methylation in the promoter region of BRCA1 in normal tissues of breast cancer patients with a specific BRCA1-like tissue type (37). No germline mutation of BRCA1 and BRCA2 was detected in these patients. This finding suggests constitutional epimutation of BRCA1 in breast cancer patients. Methylation of BRCA1 is considered to be the first hit and the histologically characteristic type occurs due to deletion of both BRCA1 genes.

Epimutation and Lynch syndrome

Lynch syndrome (HNPCC) is a typical familial tumor with autosomal dominant inheritance. A study of morbidity in colorectal cancer showed that Lynch syndrome develops in approximately 3% of patients with colorectal cancer (38). Aberrant mismatch repair (MMR) genes are involved in carcinogenesis of Lynch syndrome. A total of 6 MMR genes have been cloned to date: hMSH2, hMLH1, human MutS homologue 3 (hMSH3) and 6 (hMSH6), and postmeiotic segregation increased 1 (PMS1) and 2 (PMS2). Lynch syndrome kindreds have been confirmed to have mutations in 3 of these genes: hMSH2, hMLH1 and hMSH6(37). hMLH1 and hMSH2 mutations are particularly considered to be a cause of Lynch syndrome. Such mutations also cause a predisposition for endometrial, small intestine and ovarian cancer, in addition to rectal cancer. hMLH1 and hMSH2 encode MMR proteins and inactivation of these genes may cause MSI in tumors (21). Microsatellites are short-tandem repeats (STRs) that are typically in the non-coding region, and therefore, have no relationship with production of aberrant proteins with mutations. However, some STRs exist in regions encoding important genes such as BCL2-associated X protein (BAX), which is related to apoptosis induction, and insulin-like growth factor 2 receptor (IGF2R), which is involved in inhibition of cell proliferation. Mutation of these genes is associated with carcinogenesis.

Recently, a new type of Lynch syndrome has been found with no pathologic mutation in MMR genes, but epimutation in the promoter region of hMLH1 or hMSH2(40). This finding suggested that epimutation in germline hMLH1 can be a cause of Lynch syndrome. In families with hMSH2 methylation, germline mutation of epithelial cell adhesion molecule (EPCAM) is a cause of epimutation in the upper hMSH2 promoter (41). EPCAM is highly expressed in epithelial tissues and cancer (42). A 3′ end deletion in EPCAM causes readthrough with hMSH2, which leads to hypermethylation of CpG islands in the hMSH2 promoter (43). Furthermore, methylation of hMSH2 in Lynch syndrome kindreds has been shown to be transmitted genetically (28). It is of interest that no methylation of hMSH2 has currently been shown. In contrast to many patients with constitutional methylation of hMLH1, for which allele methylation was already confirmed, methylation of an hMSH2 allele was detected in approximately 50%. The level of methylation also depends on the tissues tested. Methylation of hMSH2 follows Mendelian inheritance, in contrast to epimutation of hMLH1, and patients with Lynch syndrome due to epimutation had a different level of methylation from that of epimutation carriers in their family and/or different levels among different tissues.

Conclusion

Epimutation has characteristics of inheritance, deletion in embryonic development, and a hereditary form that does not follow Mendelian inheritance. Cancers involved in epimutation include Lynch syndrome (HNPCC), chronic lymphocytic leukemia, breast cancer, and ovarian cancer. Tumors with epimutation may have specific histological characteristics, and methylation patterns in normal tissues may indicate the cancer tissue type. Use of this information may reduce the need for current invasive tests, including biopsy of tumor tissues. The cancer risk of healthy individuals may also be accessible through a minimally invasive procedure based on differences in methylation patterns between healthy individuals and cancer patients. Development of such techniques requires elucidation of the many factors causing methylation. It is currently unknown how the many variations of methylation occur in normal somatic tissues at an individual level; however, methylation patterns are maintained in individuals. Identical twins have different patterns of DNA methylation and these differences are increased in twins living in different environments (44). CpG islands are methylated age-dependently, and methylation may influence methyl group metabolism such as that involving folic acid, methionine, choline, vitamin B12 and betaine due to metabolite ingestion, which may change methylation patterns. Environmental factors, particularly in early development, may be associated with predisposition to diseases, including cancer, and other epigenomic changes (45). The possible association of methylation with environmental factors and aging, in addition to genetic factors, indicates the need for additional studies of the relationship between epimutation and these factors.

Research efforts are also currently directed at improvement of epigenetic abnormalities based on epimutation studies. Epigenetic therapy to enhance re-expression of tumor suppressor genes using DNA methyltransferase (DNMT) inhibitors (azacitidine and decitabine) has some effect on hematologic malignancies (44). However, epigenetic therapy with a combination of a DNMT inhibitor and a histone deacetylase (HDAC) inhibitor cannot completely remodel chromosomes, and consequently has no effect on re-expression of stable genes. Furthermore, many studies show that re-expressed genes are inhibited again after withdrawal of epigenetic therapy. Thus, epigenetic therapy still has many limitations and further studies are needed to improve this approach.

Acknowledgements

The authors gratefully acknowledge the grant support from the Japan Society for the Promotion of Science (JSPS) through a Grant-in-Aid for Scientific Research (KAKENHI); a Grant-in-Aid for Scientific Research (C) (22591866), and a Grant-in-Aid for Young Scientists (B) (21791573); the Ichiro Kanehara Foundation; Kobayashi Foundation for Cancer Research; and the Keio University Medical Science Fund through a Research Grant for Life Sciences and Medicine.

References

1 

Holliday R: The inheritance of epigenetic defects. Science. 238:163–170. 1987. View Article : Google Scholar : PubMed/NCBI

2 

Das OP and Messing J: Variegated phenotype and developmental methylation changes of a maize allele originating from epimutation. Genetics. 136:1121–1141. 1994.PubMed/NCBI

3 

Schofield PN, Joyce JA, Lam WK, Grandjean V, Ferguson-Smith A, Reik W and Maher ER: Genomic imprinting and cancer; new paradigms in the genetics of neoplasia. Toxicol Lett. 120:151–160. 2001. View Article : Google Scholar : PubMed/NCBI

4 

Hitchins M, Williams R, Cheong K, et al: MLH1 germline epimutations as a factor in hereditary nonpolyposis colorectal cancer. Gastroenterology. 129:1392–1399. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Hitchins MP, Wong JJ, Suthers G, Suter CM, Martin DI, Hawkins NJ and Ward RL: Inheritance of a cancer-associated MLH1 germ-line epimutation. N Engl J Med. 356:697–705. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Valle L, Carbonell P, Fernandez V, Dotor AM, Sanz M, Benitez J and Urioste M: MLH1 germline epimutations in selected patients with early-onset non-polyposis colorectal cancer. Clin Genet. 71:232–237. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Morak M, Schackert HK, Rahner N, et al: Further evidence for heritability of an epimutation in one of 12 cases with MLH1 promoter methylation in blood cells clinically displaying HNPCC. Eur J Hum Genet. 16:804–811. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Hitchins MP and Ward RL: Erasure of MLH1 methylation in spermatozoa-implications for epigenetic inheritance. Nat Genet. 39:12892007. View Article : Google Scholar : PubMed/NCBI

9 

Cooper WN, Luharia A, Evans GA, et al: Molecular subtypes and phenotypic expression of Beckwith-Wiedemann syndrome. Eur J Hum Genet. 13:1025–1032. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Schönherr N, Meyer E, Roos A, Schmidt A, Wollmann HA and Eggermann T: The centromeric 11p15 imprinting centre is also involved in Silver-Russell syndrome. J Med Genet. 44:59–63. 2007.PubMed/NCBI

11 

Conrad DF, Pinto D, Redon R, et al: Origins and functional impact of copy number variation in the human genome. Nature. 464:704–712. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Tufarelli C, Stanley JA, Garrick D, Sharpe JA, Ayyub H, Wood WG and Higgs DR: Transcription of antisense RNA leading to gene silencing and methylation as a novel cause of human genetic disease. Nat Genet. 34:157–165. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Friend SH, Bernards R, Rogelj S, Weinberg RA, Rapaport JM, Albert DM and Dryja TP: A human DNA segment with properties of the gene that predisposes to retinoblastoma and osteosarcoma. Nature. 323:643–646. 1986. View Article : Google Scholar : PubMed/NCBI

14 

Nishisho I, Nakamura Y, Miyoshi Y, et al: Mutations of chromosome 5q21 genes in FAP and colorectal cancer patients. Science. 253:665–669. 1991. View Article : Google Scholar : PubMed/NCBI

15 

Hussussian CJ, Struewing JP, Goldstein AM, et al: Germline p16 mutations in familial melanoma. Nat Genet. 8:15–21. 1994. View Article : Google Scholar : PubMed/NCBI

16 

Greger V, Passarge E, Höpping W, Messmer E and Horsthemke B: Epigenetic changes may contribute to the formation and spontaneous regression of retinoblastoma. Hum Genet. 83:155–158. 1989. View Article : Google Scholar : PubMed/NCBI

17 

Sakai T, Toguchida J, Ohtani N, Yandell DW, Rapaport JM and Dryja TP: Allele-specific hypermethylation of the retinoblastoma tumor-suppressor gene. Am J Hum Genet. 48:880–888. 1991.PubMed/NCBI

18 

Herman JG, Latif F, Weng Y, et al: Silencing of the VHL tumor-suppressor gene by DNA methylation in renal carcinoma. Proc Natl Acad Sci USA. 91:9700–9704. 1994. View Article : Google Scholar : PubMed/NCBI

19 

Kane MF, Loda M, Gaida GM, Lipman J, Mishra R, Goldman H, Jessup JM and Kolodner R: Methylation of the hMLH1 promoter correlates with lack of expression of hMLH1 in sporadic colon tumors and mismatch repair-defective human tumor cell lines. Cancer Res. 57:808–811. 1997.PubMed/NCBI

20 

Dobrovic A and Simpfendorfer D: Methylation of the BRCA1 gene in sporadic breast cancer. Cancer Res. 57:3347–3350. 1997.PubMed/NCBI

21 

de la Chapelle A: Genetic predisposition to human disease: allele-specific expression and low-penetrance regulatory loci. Oncogene. 28:3345–3348. 2009.PubMed/NCBI

22 

Weisenberger DJ, Siegmund KD, Campan M, et al: CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat Genet. 38:787–793. 2006. View Article : Google Scholar

23 

Gazzoli I, Loda M, Garber J, Syngal S and Kolodner RD: A hereditary nonpolyposis colorectal carcinoma case associated with hypermethylation of the MLH1 gene in normal tissue and loss of heterozygosity of the unmethylated allele in the resulting microsatellite instability-high tumor. Cancer Res. 62:3925–3928. 2002.

24 

Suter CM, Martin DI and Ward RL: Germline epimutation of MLH1 in individuals with multiple cancers. Nat Genet. 36:497–501. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Miyakura Y, Sugano K, Konishi F, et al: Extensive methylation of hMLH1 promoter region predominates in proximal colon cancer with microsatellite instability. Gastroenterology. 121:1300–1309. 2001. View Article : Google Scholar : PubMed/NCBI

26 

Miyakura Y, Sugano K, Konishi F, et al: Methylation profile of the MLH1 promoter region and their relationship to colorectal carcinogenesis. Genes Chromosomes Cancer. 36:17–25. 2003. View Article : Google Scholar : PubMed/NCBI

27 

Miyakura Y, Sugano K, Akasu T, et al: Extensive but hemiallelic methylation of the hMLH1 promoter region in early-onset sporadic colon cancers with microsatellite instability. Clin Gastroenterol Hepatol. 2:147–156. 2004. View Article : Google Scholar : PubMed/NCBI

28 

Chan TL, Yuen ST, Kong CK, et al: Heritable germline epimutation of MSH2 in a family with hereditary nonpolyposis colorectal cancer. Nat Genet. 38:1178–1183. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Raval A, Tanner SM, Byrd JC, et al: Downregulation of death-associated protein kinase 1 (DAPK1) in chronic lymphocytic leukemia. Cell. 129:879–890. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Reddy AN, Jiang WW, Kim M, Benoit N, Taylor R, Clinger J, Sidransky D and Califano JA: Death-associated protein kinase promoter hypermethylation in normal human lymphocytes. Cancer Res. 63:7694–7698. 2003.PubMed/NCBI

31 

Armes JE and Venter DJ: The pathology of inherited breast cancer. Pathology. 34:309–314. 2002. View Article : Google Scholar : PubMed/NCBI

32 

Lakhani SR, Van De Vijver MJ, Jacquemier J, Anderson TJ, Osin PP, MaGuffog L and Easton DF: The pathology of familial breast cancer: predictive value of immunohistochemical markers estrogen receptor, progesterone receptor, HER-2, and p53 in patients with mutations in BRCA1 and BRCA2. J Clin Oncol. 20:2310–2318. 2002. View Article : Google Scholar

33 

Foulkes WD, Stefansson IM, Chappuis PO, Bégin LR, Goffin JR, Wong N, Trudel M and Akslen LA: Germline BRCA1 mutations and a basal epithelial phenotype in breast cancer. J Natl Cancer Inst. 95:1482–1485. 2003. View Article : Google Scholar : PubMed/NCBI

34 

Esteller M, Silva JM, Dominguez G, et al: Promoter hypermethylation and BRCA1 inactivation in sporadic breast and ovarian tumors. J Natl Cancer Inst. 92:564–569. 2000. View Article : Google Scholar : PubMed/NCBI

35 

Catteau A and Morris JR: BRCA1 methylation: a significant role in tumour development? Semin Cancer Biol. 12:359–371. 2002. View Article : Google Scholar : PubMed/NCBI

36 

Hedenfalk I, Duggan D, Chen Y, et al: Gene-expression profiles in hereditary breast cancer. N Engl J Med. 344:539–548. 2001. View Article : Google Scholar : PubMed/NCBI

37 

Snell C, Krypuy M, Wong EM; kConFab investigators; Loughrey MB and Dobrovic A: BRCA1 promoter methylation in peripheral blood DNA of mutation negative familial breast cancer patients with a BRCA1 tumour phenotype. Breast Cancer Res. 10:R122008. View Article : Google Scholar : PubMed/NCBI

38 

Hampel H, Frankel WL, Martin E, et al: Feasibility of screening for Lynch syndrome among patients with colorectal cancer. J Clin Oncol. 26:5783–5788. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Peltomäki P and Vasen H: Mutations associated with HNPCC predisposition - Update of ICG-HNPCC/INSiGHT mutation database. Dis Markers. 20:269–276. 2004.PubMed/NCBI

40 

Hirata K, Kanemitsu S, Nakayama Y, Nagata N, Itoh H, Ohnishi H, Ishikawa H and Furukawa Y; HNPCC registry and genetic testing project of the Japanese Society for Cancer of the Colon and Rectum (JSCCR): A novel germline mutation of MSH2 in a hereditary nonpolyposis colorectal cancer patient with liposarcoma. Am J Gastroenterol. 101:193–196. 2006. View Article : Google Scholar : PubMed/NCBI

41 

Naruse H, Ikawa N, Yamaguchi K, et al: Determination of splice-site mutations in Lynch syndrome (hereditary nonpolyposis colorectal cancer) patients using functional splicing assay. Fam Cancer. 8:509–517. 2009. View Article : Google Scholar

42 

Winter MJ, Nagtegaal ID, van Krieken JH and Litvinov SV: The epithelial cell adhesion molecule (Ep-CAM) as a morpho-regulatory molecule is a tool in surgical pathology. Am J Pathol. 163:2139–2148. 2003. View Article : Google Scholar : PubMed/NCBI

43 

Ligtenberg MJ, Kuiper RP, Chan TL, et al: Heritable somatic methylation and inactivation of MSH2 in families with Lynch syndrome due to deletion of the 3′ exons of TACSTD1. Nat Genet. 41:112–117. 2009.PubMed/NCBI

44 

Fraga MF, Ballestar E, Paz MF, et al: Epigenetic differences arise during the lifetime of monozygotic twins. Proc Natl Acad Sci USA. 102:10604–10609. 2005. View Article : Google Scholar : PubMed/NCBI

45 

Issa JP, Ottaviano YL, Celano P, Hamilton SR, Davidson NE and Baylin SB: Methylation of the oestrogen receptor CpG island links ageing and neoplasia in human colon. Nat Genet. 7:536–540. 1994. View Article : Google Scholar : PubMed/NCBI

46 

Rose MG: Hematology: azacitidine improves survival in myelodysplastic syndromes. Nat Rev Clin Oncol. 6:502–503. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September 2012
Volume 41 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Banno K, Kisu I, Yanokura M, Tsuji K, Masuda K, Ueki A, Kobayashi Y, Yamagami W, Nomura H, Tominaga E, Tominaga E, et al: Epimutation and cancer: a new carcinogenic mechanism of Lynch syndrome (Review). Int J Oncol 41: 793-797, 2012
APA
Banno, K., Kisu, I., Yanokura, M., Tsuji, K., Masuda, K., Ueki, A. ... Aoki, D. (2012). Epimutation and cancer: a new carcinogenic mechanism of Lynch syndrome (Review). International Journal of Oncology, 41, 793-797. https://doi.org/10.3892/ijo.2012.1528
MLA
Banno, K., Kisu, I., Yanokura, M., Tsuji, K., Masuda, K., Ueki, A., Kobayashi, Y., Yamagami, W., Nomura, H., Tominaga, E., Susumu, N., Aoki, D."Epimutation and cancer: a new carcinogenic mechanism of Lynch syndrome (Review)". International Journal of Oncology 41.3 (2012): 793-797.
Chicago
Banno, K., Kisu, I., Yanokura, M., Tsuji, K., Masuda, K., Ueki, A., Kobayashi, Y., Yamagami, W., Nomura, H., Tominaga, E., Susumu, N., Aoki, D."Epimutation and cancer: a new carcinogenic mechanism of Lynch syndrome (Review)". International Journal of Oncology 41, no. 3 (2012): 793-797. https://doi.org/10.3892/ijo.2012.1528