eISSN: 1897-4309
ISSN: 1428-2526
Contemporary Oncology/Współczesna Onkologia
Current issue Archive Manuscripts accepted About the journal Supplements Addendum Special Issues Editorial board Reviewers Abstracting and indexing Subscription Contact Instructions for authors Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
4/2011
vol. 15
 
Share:
Share:
Original paper

Methylation profile of selected TSGs in non-cancerous thyroid tissue adjacent to primary PTC

Monika Migdalska-Sęk
,
Dorota Pastuszak-Lewandoska
,
Karolina Czarnecka
,
Ewa Nawrot
,
Daria Domańska
,
Jan Brzeziński
,
Marek Dedecjus
,
Ewa Brzeziańska

Wspolczesna Onkol 2011; 15 (4): 191–197
Online publish date: 2011/09/09
Article file
- Methylation profile.pdf  [0.13 MB]
Get citation
 
PlumX metrics:
 

Introduction

Tumour suppressor genes (TSGs) encode proteins involved in various crucial processes, such as cell proliferation, cell-cycle control, apoptosis, DNA damage detection and repair, cell adhesion, migration and invasion, senescence, and carcinogen detoxification [1]. The current definition of bona fide tumour suppressor specifies that a proven TSG loses its function in the development of a cancer and its in vivo inactivation enhances initiation, growth or progression of a tumour [2].

Thus far, many TSGs have been identified and confirmed to be associated with development and progression of many types of cancers. It is widely accepted that both functional copies of TSG are inactivated during carcinogenesis, via the “two-hit” model of tumour suppressor inactivation [3-5]. Initially, a mutational mechanism for the loss of TSG function was considered. Now it is complemented with a nonmutational TSG inactivation pathway, i.e. silencing through epigenetic mechanisms [5-9].

Epigenetic modifications are defined as heritable changes in gene expression without alteration in the DNA sequence [10]. A predominant mode of epigenetic alteration in cancer is gene silencing via promoter hypermethylation associated with addition of a methyl group (CH3) at the carbon 5 position of the cytosine ring in CpG dinucleotides [4, 10]. It is known that methylation of CpG islands interferes with active transcription by recruiting methyl-cytosine-binding proteins and histone deacetylases, which in a coordinated fashion modify nucleosomes to form transcriptionally repressive chromatin [4, 5, 8, 11].

A huge amount of data has been accumulated indicating epigenetic silencing of a number of TSGs in many human malignancies, e.g., in breast, prostate, colon, stomach, oesophagus, blood, central nervous system, and lung cancer [1, 5, 8, 9, 11]. Numerous studies have defined cancer-type specific methylation profiles [11-14]. Generally, aberrant DNA methylation is significantly associated with poorer tumour differentiation, tumour aggressiveness and poor prognosis [15].

On the other hand, accumulating evidence suggests that alterations of methylation are involved in the early and precancerous stages. It is hypothesized that precancerous conditions showing alterations of DNA methylation may progress rapidly and lead to more malignant cancers [6, 15-17].

In thyroid tumorigenesis hypermethylation of certain genes is associated with the inactivation of various signalling pathways [18-22]. But there is a small number of reports indicating that changes in DNA methylation profile can be observed in thyroid noncancerous tissue.

The aim of our study was to assess the methylation profile of a panel of eight TSGs (ARHI, CDH1, KCNQ1, MEST, p16INK4A, RASSF1A, SLC5A8, VHL) in noncancerous thyroid tissue adjacent to the primary tumour, i.e., papillary thyroid carcinoma (PTC), and to determine whether it parallels the methylation level of the studied TSGs in PTCs.

Material and methods

The procedures used in the study had been approved by the Ethical Committee of the Medical University of Lodz, Poland.

Thyroid tissue samples (100-150 mg) were obtained from patients who had undergone total

thyroidectomy at the Department of General, Oncological and Endocrine Surgery, Medical University of Lodz, and at the Department of General and Oncological Surgery, Medical University of Lodz, Poland, during the years 2006-2009. For the analysis, two samples of thyroid tissue were obtained from each patient: from the centre of the primary lesion and the matching noncancerous thyroid tissue (macroscopically unchanged, procured at the most distant site from the resected specimen). Tissue samples, immediately after resection, were collected in lysis buffer (RNAlater buffer, Qiagen Sciences, USA), homogenized and frozen at –70°C until use. The collected biological material consisted of 11 pairs of tissue samples (PTC and adjacent noncancerous tissue, NPTC), obtained from 8 women and 3 men, mean age 41.8 (range 25-66).

Regarding PTC tissue, some samples have already been analysed, but in a different context, and the results have been published in our previous study [23].

Isolation of genomic DNA from thyroid tissue was performed using QIAamp DNA Mini Kit (Qiagen, Germany), according to the manufacturer’s protocol. Quality and quantity of each DNA sample were spectrophotometrically assessed (NanoDrop Spectrophotometer ND-1000, ThermoScientific, USA).

Methylation status of the studied genes (ARHI, CDH1, KCNQ1, MEST, p16INK4A, RASSF1A, SLC5A8, VHL) was assessed by methylation-specific polymerase chain reaction (MSP) using bisulfite converted DNA. Genomic DNA (1.0 g) was modified with sodium bisulfite, using CpGenome™ DNA Modification Kit (CHEMICON International, Millipore, USA), according to the manufacturer’s protocol. Concentration and purity of the modified DNA were determined spectrophotometrically (NanoDrop Spectrophotometer ND-1000, ThermoScientific, USA).

Primers for MSP were designed using Methyl Primer Express® Software according to the guidelines on the website www.appliedbiosystems.com/methylprimerexpress, and obtained from METAbion (Germany). Sequences and length of MSP primers have been described in our previous paper [23].

The conventional MSP method was performed according to Herman et al. [24] with some modifications. Briefly, MSP was run in triplicate for each sodium bisulfite modified DNA sample, using AmpliTaq Gold® DNA Polymerase Kit (Applied Biosystems, USA). Amplification reactions were conducted in a total volume of 25 l and the MSP master mix contained 1000 ng of DNA, 0.7 l (100 pmol) of each oligonucleotide primer (forward and reverse), 2.5 l (2.5 mM) of dNTPs mix, 2 l of 25 mM MgCl2, Hot Start AmpliTaq Gold® DNA Polymerase (5 U/l), 2.5 µl of 10 × AmpliTaq Gold buffer and nuclease-free water. MSP reactions were run for 35 cycles. Gene-specific annealing temperatures were determined experimentally and are presented in Table 1.

In each PCR reaction, positive and negative MSP controls were included. CpGenome Universal Methylated DNA (enzymatically methylated human male genomic DNA) served as a positive methylation control and CpGenome Universal Unmethylated DNA (human fetal cell line) was used as a negative control (CHEMICON International, Millipore, USA). Additionally, blank samples with nuclease-free water were used instead of DNA as a control for PCR contamination.

The obtained MSP products were separated on polyacrylamide gel (8% PAA) to visualize (in UV light, after ethidium bromide staining) unmethylated and methylated DNA alleles (quality assessment).

Additionally, concentrations (ng) of MSP products were spectrophotometrically estimated, using DNA1000 LabChip Kit, in an Agilent 2100 Bioanalyzer (Agilent Technologies, USA). Based on the quantitative results, methylation indexes (MIs) were calculated for each sample, according to the formula MI = (M)/(M + U), where (M) stands for the methylated and (U) for the unmethylated allele concentration.

The differences in methylation levels (MI values) of the studied genes between cancerous and noncancerous thyroid tissue were statistically analysed using non-parametric Mann-Whitney test and Student’s t-test, and presented as mean or median ± SEM and ± SD values. Statistical significance was determined at the level of p-value less than 0.05. For calculations, Statistica for Windows 7.0 program was applied.

Results

Qualitative assessment of methylation frequency

Analysis of methylation status of the studied TSGs (ARHI, CDH1, KCNQ1, MEST, p16INK4A, RASSF1A, SLC5A8, VHL) revealed the presence of both methylated and nonmethylated alleles in the majority of tissue specimens (PTC and adjacent noncancerous tissue, NPTC). The frequencies of methylated alleles in the studied tissue groups, for all TSGs, are given in Table 2.

The highest frequency of methylated alleles in both tumour and adjacent tissue samples was

observed for ARHI, CDH1, p16INK4A and RASSF1A genes. Only one gene, SLC5A8, was not methylated in PTC and very infrequently in NPTC. Relatively low methylation frequency was found for KCNQ1 (in both tissue groups) and VHL (especially in noncancerous thyroid tissue). The remaining gene, MEST, showed a similarly high level of methylation in both PTC and NPTC. In each tumour and adjacent tissue at least two genes were methylated, as listed in Table 3. Methylation of multiple genes was common, both in cancerous and noncancerous tissue. There were no samples with no methylated gene at all.

We also analysed the concordance of methylation of the studied TSGs between tumour and matched noncancerous thyroid tissue. The results are summarized in Table 4. In most cases, clear overlapping of TSG hypermethylation could be seen between paired samples. The number of cases with hypermethylated genes in tumour and no methylation in paired noncancerous tissue was small. Most genes were never or rarely methylated in noncancerous tissue in the absence of methylation in the paired cancerous tissue. Two of the studied genes – SLC5A8 and KCNQ1 – were unmethylated in most of the matched samples (Table 4).

Quantitative assessment of methylation level

Based on spectrophotometric estimation performed in the Agilent 2100 Bioanalyzer, fluorescence units (FU) of MSP products were quantified (ng/l), according to DNA size marker (DNA ladder, Agilent Technologies, USA). The assessed concentrations of methylated and unmethylated alleles served for MI value calculations, for each gene and in each tissue sample (Table 5).

Based on the median values, we estimated the percentage of samples with high promoter methylation (MI  median value) and with low promoter methylation (MI < median value) for each gene. Among the 8 studied genes, 3 of them – KCNQ1, SLC5A8 and VHL – revealed a very low methylation level in most paired samples. Especially, in the case of SLC5A8 100% and 90% of specimens were completely unmethylated in PTC and NPTC respectively. Similarly, VHL showed methylation only in two cases (2/7) of PTC (although with a high level of MIs) and in one case of NPTC. Regarding KCNQ1, the number of samples with low promoter methylation was two-fold higher than the number of samples with high promoter methylation (66.7% vs. 33.3%). For the remaining genes, both groups of samples (highly and low methylated) were similar, regarding the number of cases, ranging from 42.8% to 57.14%, with a small predominance of highly methylated specimens. The highest diversity was observed for p16INK4A in NPTC tissues: a high level of promoter methylation was found in 72.7% of samples and low promoter methylation in 27.3% of specimens.

Statistical analysis

Statistical analysis comparing MI values between tumours (PTC) and adjacent noncancerous tissue (NPTC) for each TSG was performed. Non-parametric Mann-Whitney test revealed no significant differences regarding each TSG between the two studied tissue groups (p > 0.05). These results were confirmed by Student’s t-test.

Methylation profile, i.e., total methylation level of all studied TSGs, in the PTC group in comparison with the NPTC group was not significantly different (p > 0.05, Student’s t-test). The relevant “box and whisper” plot is shown in Fig. 1.

Regarding the groups classified according to high and low methylation levels (for each gene), there were no statistically significant differences between both groups, PTC vs. NPTC (p > 0.05, two tailed Fisher exact test). The results were confirmed by 2 correlation.

The results for PTCs, regarding patients’ age and sex, as well as tumour staging according to TNM and AJCC classifications, have been published in our previous study [23].

Discussion

Now it is widely accepted that loss of tumour suppressor function leads to the initiation and progression of human cancer. Inactivation of TSG can result from both genetic mechanisms such as mutations or epigenetic modifications such as promoter hypermethylation. The results of many studies indicate that, particularly in the case of TSGs with a low incidence of mutations, epigenetic inactivation is a more frequent event than genetic alteration, especially that only a few genetic changes are known to be responsible for cancer development, particularly in the earlier, precancerous stages [5, 6, 25].

The aim of our study was to assess the methylation status of 8 tumour suppressor genes – ARHI, CDH1, KCNQ1, MEST, p16INK4A, RASSF1A, SLC5A8, VHL – in noncancerous thyroid tissue adjacent to the primary tumour (papillary thyroid carcinoma) and compare it with the methylation level of the studied TSGs in PTC tissues, in pairs.

The results obtained in our study indicated that promoter hypermethylation of TSGs in thyroid tissue undergoing malignant transformation was a frequent event. Among the 8 studied TSGs the highest methylation rate, i.e. 100% of methylated specimens in the PTC group, was found for 4 genes: ARHI, CDH1, p16INK4A and RASSF1A; in the case of MEST 80% of PTC samples were methylated. The same genes displayed similarly high frequency of promoter methylation in paired noncancerous samples (see Tables 2 and 4). In the case of each of them, 50% and even more DNA specimens revealed high promoter methylation levels (according to median MI values) both in PTC and in NPTC.

Regarding the remaining 3 studied TSGs, i.e., KCNQ1, SLC5A8 and VHL, the frequency of promoter methylation was low in both cancerous and noncancerous tissue groups, affecting only 2-3 cases. We did not observe any methylated alleles of SLC5A8 in PTC specimens.

There are several published reports describing epigenetic modifications in thyroid carcinomas. In a panel of analysed TSGs, promoter hypermethylation of CDH1, p16INK4A, RASSF1A and SLC5A8 in malignant thyroid tumours was confirmed [21]. Methylation frequency of those genes observed in PTCs ranged from 22% to 83% for CDH1 [18, 26-28], 25-41% for p16INK4A [29-32], 15-60% for RASSF1A [18, 33-35] and 33-90% for SLC5A8 [20, 36], depending on the study and the technique used for analysis. In the majority of experiments the most frequently used method for methylation analysis was methylation-specific PCR (MSP). It is a sensitive technique, although qualitative, with a high risk of false-positive results. As proved in some studies, the quantitative analysis of methylation showed better results for the discrimination of malignant thyroid tumours [18, 32]. The quantitative method used in our study enabled us to calculate MI values based on concentrations of methylated and unmethylated alleles. Expressing MI × 100%, we found that the methylation levels of 3 genes in our study – ARHI, CDH1 and RASSF1A – generally fell within the range of frequencies reported in the literature.

However, our findings with regard to methylation of SLC5A8 differed from those reported [20, 36], as we did not observe its promoter hypermethylation in PTC. We did not find it in the adjacent thyroid tissue either, which has not been studied so far. Moreover, in our previous study, we did not observe SLC5A8 methylation in nodular goitre (NG) [23]. Our results could support the findings of other authors indicating SLC5A8 methylation at a later stage during thyroid carcinogenesis and its association with poor pathological characteristics of PTC, predicting increased progression and aggressiveness [20, 36]. Additionally, the results of the study performed by Schagdarsurengin et al. [37] indicated preferential SLC5A8 methylation in undifferentiated thyroid carcinomas (UTC) as compared to other thyroid lesions (PTC; follicular thyroid carcinoma, FTC; medullary thyroid carcinoma, MTC; follicular adenoma, FA; NG).

Regarding ARHI and MEST, which were found to be frequently methylated in our study, their roles and epigenetic as well as genetic mechanisms of inactivation in thyroid neoplastic transformation require further analyses, due to the paucity of studies and their controversial results [23, 38-41]. However, our observations of similar frequencies and levels of methylation in paired samples for each of those genes indicated that the same molecular mechanism of regulation associated with carcinogenesis might occur in the earliest stage of thyroid tumourigenesis, although different in the case of each gene, as described below. According to the results of numerous studies, ARHI silencing in cancers can be caused by loss of heterozygosity (LOH), DNA methylation, histone deacetylation and transcriptional regulation [42, 43]. The functional effect of TSG inactivation also depends on gene localization, i.e., within the IR (imprinted) or NIR (non-imprinted) genome region. During tumourigenesis maternally imprinted ARHI can lose its function with a “single hit”, via inactivation of one functional allele. The results obtained in our study, i.e., 100% frequency of methylation observed in the studied samples and very high MI values (ME = 0.99), indicated that the functional paternal allele of the gene could be hypermethylated, especially that the study performed by Weber et al. [39] showed infrequent LOH at the ARHI locus in PTC samples. In the case of the other imprinted gene involved in our study, MEST, high frequency of methylation observed in PTC and NPTC samples revealed the presence of methylated alleles in all, except two, specimens. However, MI values did not exceed 0.50 (ME < 0.50) and in most cases were even lower. This means that the only functional allele of MEST possibly remained unmethylated and, moreover, the imprinted allele could be subjected to LOH or loss of imprinting (LOI). LOI at the MEST locus has been described in some carcinomas, including breast and lung cancers [44-46], and the same molecular event could possibly be involved in thyroid tumourigenesis. On the other hand, although data on LOH involving MEST in thyroid tumours are controversial [38, 40, 41], the results of our previous study indicated the possible role of LOH in MEST silencing in PTC [23].

Summing up, we found frequent promoter methylation of 5 tumour suppressor genes, i.e., ARHI, CDH1, p16INK4A, MEST and RASSF1A, and 3 among them (ARHI, CDH1 and RASSF1A) showed high methylation levels (MI values), both in PTC and NPTC. So far, TSG methylation in thyroid lesions has not been analysed widely in tissue adjacent to the primary tumour. However, promoter methylation of some genes, including CDH1, p16INK4A and RASSF1A, was also found in benign thyroid tumours (follicular adenomas), as well as in nodular goitres [18, 23, 26, 29, 32-35], indicating epigenetic modifications as an early event in thyroid tumourigenesis. Moreover, several studies have revealed that the methylation pattern of some TSGs (including CDH1, p16INK4A and RASSF1A) was nearly identical when comparing cancerous and noncancerous tissues [18, 32]. Our results confirmed promoter methylation of CDH1, p16INK4A and RASSF1A as an early event in malignant thyroid transformation.

The presence of epigenetic modifications of TSGs in noncancerous tissue adjacent to the primary tumour may be explained by the concept of field cancerization. Originally, it referred to genetic changes occurring in normal tissue, explaining the local relapses [47-49]. The presence of a field (areas) composed of genetically altered cells is a risk factor for future carcinogenesis. Now, it has become obvious that epigenetic alterations also play a role in this phenomenon. An epigenetic field defect has been observed in several carcinomas, including head and neck cancers, oesophageal, lung, urothelial and stomach cancer [50-52]. So far, however, such changes have not been well documented in thyroid lesions. According to our knowledge, only one report regarding TSG methylation analysis in malignant thyroid tumours in comparison with adjacent thyroid tissue has been published so far [28]. The results did not show any differences in the methylation status of 4 genes (CDH1, DAPK, ATM and TSHR) between cancerous and noncancerous thyroid tissue. The authors hypothesized that it might represent field cancerization, supported by the prevalence of multifocal thyroid carcinomas. The results of our study are similar: they indicate a nearly identical methylation profile of individual TSGs in PTC and adjacent tissue. Only a few specimens of NPTC have revealed no methylation for a given TSGs methylated in PTC.

In conclusion, in our study promoter methylation of multiple genes, especially of ARHI, CDH1, p16INK4A and RASSF1A, was common, both in cancerous and adjacent thyroid tissue. This finding makes it impossible to establish a panel of methylated TSGs specific for thyroid papillary carcinoma, although it improves our understanding of PTC biology. Epigenetic modifications of those genes may be of functional importance for thyroid carcinogenesis, especially in its very early stage. Loss of function of the studied genes, via promoter hypermethylation, may lead to gain of uncontrolled growth advantage or apoptosis inhibition.

The findings of our study expand the knowledge on the molecular alterations occurring before the development of thyroid cancer. The obtained results might be the basis for a future gene-targeted therapy of preneoplastic lesions in the thyroid gland. Additionally, they could have great value in risk assessment, early cancer detection and monitoring of disease progression.

Acknowledgement

This work was supported by Project of the State Committee for Scientific Research (KBN), No. N402 124 32/4129.

The authors gratefully acknowledge Professor Andrzej Lewiński for the possibility of conducting the research work at the Department of Endocrinology and Metabolic Diseases, Medical University of Lodz, Poland.

References

 1. Palii SS, Robertson KD. Epigenetic control of tumor suppression. Crit Rev Eukaryot Gene Expr 2007; 17: 295-316.

 2. Paige AJ. Redefining tumour suppressor genes: exceptions to the two-hit hypothesis. Cell Mol Life Sci 2003; 60: 2147-63.

 3. Knudson AG. Two genetic hits (more or less) to cancer. Nat Rev Cancer 2001; 1: 157-62.

 4. Herman JG, Baylin SB. Gene silencing in cancer in association with promoter hypermethylation. N Engl J Med 2003; 349: 2042-54.

 5. Sharma S, Kelly TK, Jones PA. Epigenetics in cancer. Carcinogenesis 2010; 31: 27-36.

 6. Jones PA, Baylin SB. The epigenomics of cancer. Cell 2007; 128: 683-92.  

7. Zhu J, Yao X. Use of DNA methylation for cancer detection and molecular classification. J Biochem Mol Biol 2007; 40: 135-44.  

8. Esteller M. Epigenetics in cancer. N Engl J Med 2008; 358: 1148-59.

 9. Sadikovic B, Al-Romaih K, Squire JA, Zielenska M. Cause and consequences of genetic and epigenetic alterations in human cancer. Curr Genomics 2008; 9: 394-408.

10. Bird A. DNA methylation patterns and epigenetic memory. Genes Dev 2002; 16: 6-21.

11. Esteller M, Corn PG, Baylin SB, Herman JG. A gene hypermethylation profile of human cancer. Cancer Res 2001; 61: 3225-9.

12. Costello JF, Frühwald MC, Smiraglia DJ, et al. Aberrant CpG-island methylation has non-random and tumour-type-specific patterns. Nat Genet 2000; 24: 132-8.

13. Park SY, Kim BH, Kim JH, Cho NY, Choi M, Yu EJ, Lee S, Kang GH. Methylation profiles of CpG island loci in major types of human cancers. J Korean Med Sci 2007; 22: 311-7.

14. Hoque MO, Kim MS, Ostrow KL, et al. Genome-wide promoter analysis uncovers portions of the cancer methylome. Cancer Res 2008; 68: 2661-70.

15. Kanai Y. Genome-wide DNA methylation profiles in precancerous conditions and cancers. Cancer Sci 2010; 101: 36-45.

16. Kanai Y, Hirohashi S. Alterations of DNA methylation associated with abnormalities of DNA methyltransferases in human cancers during transition from a precancerous to a malignant state. Carcinogenesis 2007; 28: 2434-42.

17. Feinberg AP, Ohlsson R, Henikoff S. The epigenetic progenitor origin of human cancer. Nat Rev Genet 2006; 7: 21-33.

18. Hoque MO, Rosenbaum E, Westra WH, Xing M, Ladenson P, Zeiger MA, Sidransky D, Umbricht CB. Quantitative assessment of promoter methylation profiles in thyroid neoplasms. J Clin Endocrinol Metab 2005; 90: 4011-8.

19. Hu S, Ewertz M, Tufano RP, et al. Detection of serum deoxyribonucleic acid methylation markers: a novel diagnostic tool for thyroid cancer. J Clin Endocrinol Metab 2006; 91: 98-104.

20. Hu S, Liu D, Tufano RP, et al. Association of aberrant methylation of tumor suppressor genes with tumor aggressiveness and BRAF mutation in papillary thyroid cancer. Int J Cancer 2006; 119: 2322-9.

21. Xing M. Gene methylation in thyroid tumorigenesis. Endocrinology 2007; 148: 948-53.

22. Hou P, Ji M, Xing M. Association of PTEN gene methylation with genetic alterations in the phosphatidylinositol 3-kinase/AKT signaling pathway in thyroid tumors. Cancer 2008; 113: 2440-7.

23. Czarnecka K, Pastuszak-Lewandoska D, Migdalska-Sek M, Nawrot E, Brzezinski J, Dedecjus M, Pomorski L, Brzezianska E. Aberrant methylation as a main mechanism of TSGs silencing in PTC. Front Biosci 2011 (Elite Ed.); 3: 137-57.

24. Herman JG, Graff JR, Myohanen S, Nelkin BD, Baylin SB. Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci USA 1996; 93: 9821-6.

25. Chan TA, Glockner S, Yi JM, et al. Convergence of mutation and epigenetic alterations identifies common genes in cancer that predict for poor prognosis. PLoS Med 2008; 5: e114.

26. Graff JR, Greenberg VE, Herman JG, et al. Distinct patterns of E-cadherin CpG island methylation in papillary, follicular, Hurthle’s cell, and poorly differentiated human thyroid carcinoma. Cancer Res 1998; 58: 2063-6.

27. Rocha AS, Soares P, Seruca R, Máximo V, Matias-Guiu X, Cameselle-Teijeiro J, Sobrinho-Simo~es M. Abnormalities of the E-cadherin/catenin adhesion complex in classical papillary thyroid carcinoma and in its diffuse sclerosing variant. J Pathol 2001; 194: 358-66.

28. Smith JA, Fan CY, Zou C, Bodenner D, Kokoska MS. Methylation status of genes in papillary thyroid carcinoma, Arch Otolaryngol Head Neck Surg 2007; 133: 1006-11.

29. Boltze C, Zack S, Quednow C, Bettge S, Roessner A, Schneider-Stock R. Hypermethylation of the CDKN2/p16INK4A promotor in thyroid carcinogenesis. Pathol Res Pract 2003; 199: 399-404.

30. Ishida E, Nakamura M, Shimada K, Higuchi T, Takatsu K, Yane K, Konishi N. DNA hypermethylation status of multiple genes in papillary thyroid carcinomas. Pathobiology 2007; 74: 344-52.

31. Lam AK, Lo CY, Leung P, Lang BH, Chan WF, Luk JM. Clinicopathological roles of alterations of tumor suppressor gene p16 in papillary thyroid carcinoma. Ann Surg Oncol 2007; 14: 1772-9.

32. Mohammadi-Asl J, Larijani B, Khorgami Z, Tavangar SM, Haghpanah V, Kheirollahi M, Mehdipour P. Qualitative and quantitative promoter hypermethylation patterns of the P16, TSHR, RASSF1A and RARbeta2 genes in papillary thyroid carcinoma. Med Oncol 2010 [Epub ahead of print].

33. Schagdarsurengin U, Gimm O, Hoang-Vu C, Dralle H, Pfeifer GP, Dammann R. Frequent epigenetic silencing of the CpG island promoter of RASSF1A in thyroid carcinoma. Cancer Res 2002; 62: 3698-701.

34. Xing M, Cohen Y, Mambo E, Tallini G, Udelsman R, Ladenson PW, Sidransky D. Early occurrence of RASSF1A hypermethylation and its mutual exclusion with BRAF mutation in thyroid tumorigenesis. Cancer Res 2004; 64: 1664-8.

35. Nakamura N, Carney JA, Jin L, et al. RASSF1A and NORE1A methylation and BRAFV600E mutations in thyroid tumors. Lab Invest 2005; 85: 1065-75.

36. Porra V, Ferraro-Peyret C, Durand C, et al. Silencing of the tumor suppressor gene SLC5A8 is associated with BRAF mutations in classical papillary thyroid carcinomas. J Clin Endocrinol Metab 2005; 90: 3028-35.

37. Schagdarsurengin U, Gimm O, Dralle H, Hoang-Vu C, Dammann R. CpG island methylation of tumor-related promoters occurs preferentially in undifferentiated carcinoma. Thyroid 2006; 16: 633-42.

38. Trovato M, Fraggetta F, Villari D, Batolo D, Mackey K, Trimarchi F, Benvenga S. Loss of heterozygosity of the long arm of chromosome 7 in follicular and anaplastic thyroid cancer, but not in papillary thyroid cancer. J Clin Endocrinol Metab 1999; 84: 3235-40.

39. Weber F, Aldred MA, Morrison CD, Plass C, Frilling A, Broelsch CE, Waite KA, Eng C. Silencing of the maternally imprinted tumor suppressor ARHI contributes to follicular thyroid carcinogenesis. J Clin Endocrinol Metab 2005; 90: 1149-55.

40. Sarquis MS, Weber F, Shen L, Broelsch CE, Jhiang SM, Zedenius J, Frilling A, Eng C. High frequency of loss of heterozygosity in imprinted, compared with nonimprinted, genomic regions in follicular thyroid carcinomas and atypical adenomas. J Clin Endocrinol Metab 2006; 91: 262-9.

41. Lin X, Finkelstein SD, Zhu B, Silverman JF. Molecular analysis of multifocal papillary thyroid carcinoma. J Mol Endocrinol 2008; 41: 195-203.

42. Yu Y, Fujii S, Yuan J, et al. Epigenetic regulation of ARHI in breast and ovarian cancer cells. Ann N Y Acad Sci 2003; 983: 268-77.

43. Feng W, Marquez RT, Lu Z, et al. Imprinted tumor suppressor genes ARHI and PEG3 are the most frequently down-regulated in human ovarian cancers by loss of heterozygosity and promoter methylation. Cancer 2008; 112: 1489-502.

44. Pedersen IS, Dervan PA, Broderick D, et al. Frequent loss of imprinting of PEG1/MEST in invasive breast cancer. Cancer Res 1999; 59: 5449-51.

45. Kohda M, Hoshiya H, Katoh M, Tanaka I, Masuda R, Takemura T, Fujiwara M, Oshimura M. Frequent loss of imprinting of IGF2 and MEST in lung adenocarcinoma. Mol Carcinog 2001; 31: 184-91.

46. Nakanishi H, Suda T, Katoh M, et al. Loss of imprinting of PEG1/MEST in lung cancer cell lines. Oncol Rep 2004; 12: 1273-8.

47. Braakhuis BJ, Tabor MP, Kummer JA, Leemans CR, Brakenhoff RH. A genetic explanation of Slaughter’s concept of field cancerization: evidence and clinical implications. Cancer Res 2003; 63: 1727-30.

48. Nakajima T, Enomoto S, Ushijima T. DNA methylation: a marker for carcinogen exposure and cancer risk. Environ Health Prev Med 2008; 13: 8-15.

49. Tan HK, Saulnier P, Auperin A, Lacroix L, Casiraghi O, Janot F, Fouret P, Temam S. Quantitative methylation analyses of resection margins predict local recurrences and disease-specific deaths in patients with head and neck squamous cell carcinomas. Br J Cancer 2008; 99: 357-63.

50. Ushijima T. Epigenetic field for cancerization. J Biochem Mol Biol 2007; 40: 142-50.

51. Chai H, Brown RE. Field effect in cancer – an update. Ann Clin Lab Sci 2009; 39: 331-7.

52. Wolff EM, Chihara Y, Pan F, et al. Unique DNA methylation patterns distinguish noninvasive and invasive urothelial cancers and establish an epigenetic field defect in premalignant tissue. Cancer Res 2010; 70: 8169-78.

Address for correspondence

Monika Migdalska-Sęk , MSc

Department of Molecular Bases of Medicine

Medical University of Lodz

Pomorska 251

92-213 Lodz

phone/fax: +48 42 675 77 12

e-mail: migdalska5@wp.pl
Copyright: © 2011 Termedia Sp. z o. o. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.