Skip to main content
Erschienen in: BMC Cancer 1/2017

Open Access 01.12.2017 | Research article

Investigation of DNA repair-related SNPs underlying susceptibility to papillary thyroid carcinoma reveals MGMT as a novel candidate gene in Belarusian children exposed to radiation

verfasst von: Christine Lonjou, Francesca Damiola, Monika Moissonnier, Geoffroy Durand, Irina Malakhova, Vladimir Masyakin, Florence Le Calvez-Kelm, Elisabeth Cardis, Graham Byrnes, Ausrele Kesminiene, Fabienne Lesueur

Erschienen in: BMC Cancer | Ausgabe 1/2017

Abstract

Background

Genetic factors may influence an individual’s sensitivity to ionising radiation and therefore modify his/her risk of developing papillary thyroid carcinoma (PTC). Previously, we reported that common single nucleotide polymorphisms (SNPs) within the DNA damage recognition gene ATM contribute to PTC risk in Belarusian children exposed to fallout from the Chernobyl power plant accident. Here we explored in the same population the contribution of a panel of DNA repair-related SNPs in genes acting downstream of ATM.

Methods

The association of 141 SNPs located in 43 DNA repair genes was examined in 75 PTC cases and 254 controls from the Gomel region in Belarus. All subjects were younger than 15 years at the time of the Chernobyl accident. Conditional logistic regressions accounting for radiation dose were performed with PLINK using the additive allelic inheritance model, and a linkage disequilibrium (LD)-based Bonferroni correction was used for correction for multiple testing.

Results

The intronic SNP rs2296675 in MGMT was associated with an increased PTC risk [per minor allele odds ratio (OR) 2.54 95% CI 1.50, 4.30, P per allele = 0.0006, P corr.= 0.05], and gene-wide association testing highlighted a possible role for ERCC5 (P Gene = 0.01) and PCNA (P Gene = 0.05) in addition to MGMT (P Gene = 0.008).

Conclusions

These findings indicate that several genes acting in distinct DNA repair mechanisms contribute to PTC risk. Further investigation is needed to decipher the functional properties of the methyltransferase encoded by MGMT and to understand how alteration of such functions may lead to the development of the most common type of thyroid cancer.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12885-017-3314-5) contains supplementary material, which is available to authorized users.
Abkürzungen
BER
Base excision repair
DDR
DNA damage response
DR
Direct repair
DSBR
Double strand break repair
DTC
Differentiated thyroid carcinoma
FTC
Follicular thyroid carcinoma
GWAS
Genome-Wide Association Study
HWE
Hardy-Weinberg equilibrium
MAF
Minor allele frequency
MMR
Mismatch repair
NER
Nucleotide excision repair
PTC
Papillary thyroid carcinoma
QCs
Quality controls
SNP
Single nucleotide polymorphisms
TC
Thyroid cancer

Background

Thyroid cancer (TC), with an age-standardised incident rate of 4.0 per 100,000 men and women in developed countries (http://​ci5.​iarc.​fr) is the most common type of endocrine malignancy and it is now the fifth most common cancer diagnosed in women [1]. Papillary thyroid carcinoma (PTC) along with follicular thyroid carcinoma (FTC) and Hürthle cell carcinoma (a subtype of FTC) is termed differentiated thyroid carcinoma (DTC). All together these TC types account for more than 90% of all TC and PTC is the most common subtype, representing approximately 80% of cases. During the last three decades incidence rates of TC, and in particular of PTC, have increased in nearly all countries [2]. This trend is partly attributable in high-resource countries to increased surveillance of the thyroid gland and improved detection methods [3], but it is also possible that changes in exposure to environmental factors and ionizing radiation, a well-established risk factor for this cancer especially when it occurs during childhood or as a young adult, could influence PTC incidence [46]. Other risk factors for PTC include iodine deficiency and excess [7], previous history of benign thyroid disease, such as nodules and autoimmune thyroid disease, as well as a family history of DTC. Remarkably, DTC has a strong familial component and first-degree relatives of DTC patients have up to eight times higher risk of developing DTC than the general population, indicating that genetic factors play an important role in DTC risk [8, 9]. The observed familial risk could be partly explained by high-penetrance mutations in yet unidentified genes or by the additive effect of numerous low-penetrance variants [10, 11]. This latter hypothesis would explain the paucity of families with more than two affected members. Recent case-control studies, in particular Genome-Wide Association Study (GWAS) have highlighted a number of low-penetrance alleles contributing to sporadic DTC risk. The association of DTC with the 9q22 (rs965513) locus close to the thyroid specific factor FOXE1 has been detected in all association studies conducted so far in different populations [12]. Weaker associations have been reported, among others, with rs944289 at 14q13.3 (near NKX2–1), rs966423 at 2q35 (in DIRC3), rs334725 at 1p31.3 (in NFIA) and rs2439302 at 8p12 (in NRG1) [1214]. Since the associations described so far only explain about 10% of the familial risk of DTC [15], additional studies on specific populations are of particular relevance, especially studies of high-risk populations having been exposed to known environmental risk factors. Notably a sharp increase in incidence of thyroid malignancies, virtually all PTC, has been observed in children and adolescents that were exposed to radioactive fallout from the Chernobyl nuclear power plant accident in April 1986. Variation in clinical course, ranging from highly aggressive tumours developing after the shorter latency to more indolent carcinomas with longer latent period has been reported in this population [16], which suggests that some predisposing genetic factors could influence an individual’s sensitivity to radiation and therefore modify the risk of developing TC in exposed population [17, 18].
DNA repair is an important defence mechanism against DNA damage caused by normal metabolic activities and environmental factors [19]. DNA damage is recognized and processed by a series of distinct pathways called the “DNA damage response (DDR)” [20]. It includes direct repair (DR), base and nucleotide excision repair (BER and NER), mismatch repair (MMR), double strand break repair (DSBR) and interstrand cross-links repair system [21, 22]. Because ionizing radiation produces DNA lesions and DNA repair genes play a critical role in maintaining genome integrity, it has been proposed that inherited variations in such genes may reduce DNA repair capacity and influence cancer development in exposed subjects. Few case-control studies on radiation-related PTC have been conducted to date and published data suggest that alterations in expression levels and polymorphisms in some candidate DNA repair genes belonging to the different pathways are implicated in risk of developing thyroid cancer [2326]. In particular, others and we have reported association with the coding SNP rs1801516 (p.Asp1853Asn) in the ATM gene, a key regulator of signalling following DNA double-strand breaks (DSBs) [2527]. Other possibly involved DNA repair-related SNPs include rs25487 (p.Arg399Gln) in XRCC1 involved in BER [25, 2729] and rs13181 (p.Lys751Gln) in ERCC2 involved in NER [30]. To follow up with these findings, we hypothesized that alterations in other genes related to the different DNA repair mechanisms may be correlated with tumour susceptibility in subjects originated from the Gomel region of Belarus and having been exposed to radioactive fallout from the Chernobyl nuclear power plant accident during childhood. Here we evaluated the risk association between SNPs in DNA repair genes present on the Cancer SNP panel array (Illumina) and PTC in this very unique population. Findings of this study may increase understanding of PTC aetiology and help identify subjects who may be particularly susceptible to the carcinogenic effects of radiation on the thyroid.

Methods

Study population

A total of 83 PTC cases and 324 matched and unrelated controls living in the Gomel region, one of the most contaminated areas of Belarus, were included in the present study. Participants correspond to a sub-group of subjects from the population-based case-control study carried out at the International Agency for Research on Cancer to evaluate the risk of thyroid cancer after exposure to radioactive iodine in childhood [17]. As we reported previously [26], all subjects “were younger that 15 years at the time of the Chernobyl accident” and “the control subjects were matched to the PTC cases by age (within 1 year for those who were 18 months or older at the time of the accident; within 6 months for those who aged 12-18 months; and within 1 month for who were younger than 12 months) and sex. The cases were diagnosed within 6 to 12 years following the accident with histologically verified PTC, mostly solid/follicular subtype, confirmed by the international panel of pathologists. Two thirds of the cases developed PTC before the age of 15 and the remaining cases before the age of 25. For more than 60% of cases, the latency (time between radiation exposure and diagnosis) was less than 10 years” [26] (Table 1). Individual radiation dose to the thyroid was reconstructed based on the study participant’s whereabouts and dietary habits, and information on environmental contamination for each settlement [17, 26, 31]. The radiation dose-response was similar for subjects included in the current analysis (β = 1.51, 95% CI 0.46–2.55) compared to those who did not consent to blood drawing (β = 1.55, 95% CI 1.04–2.07) [26].
Table 1
Characteristics and distribution of study participants
 
Cases
Controls
Total
Gender
 Male
35 (42%)
127 (39%)
162
 Female
48 (58%)
197 (61%)
245
Age at exposure
 < 1 y
9 (11%)
73 (22.5%)
82
 1–1.9 y
14 (17%)
46 (14%)
60
 2–2.9 y
9 (11%)
40 (12%)
49
 3–3.9 y
17 (20%)
40 (12%)
57
 4–4.9 y
9 (11%)
43 (13%)
52
 5–5.9 y
4 (5%)
18 (5.5%)
22
 6–9 y
13 (15%)
35 (11%)
48
 10–14 y
8 (10%)
29 (10%)
37
Age at diagnosis
 < 11.5 y
24 (29%)
119 (37%)
143
 11.5–14.9 y
33 (40%)
120 (37%)
153
 15+ y
26 (31%)
85 (26%)
111
The study population was rural and highly dependent on the local food produced in known iodine deficient areas [32]. As previously described in this population, “the level of stable iodine intake was correlated with the iodine concentration in the agricultural lands around their places of residence” [33], and “stable iodine intake status was estimated for each individual as a crude index based on the average level of stable iodine soil content in the settlement of residence of the study subject at the time of the Chernobyl accident” [33].

Genotyping

Genomic DNA was extracted from peripheral blood samples using a standard inorganic method [34] as previously described [26].
Study participants were genotyped for a total of 1421 SNPs located in 407 genes involved in cancer-related pathways using the Illumina GoldenGate Assay (Illumina Inc., USA) according to the manufacturer’s recommendations. The Cancer SNP Panel array contains SNPs within genes involved in the aetiology of various types of cancer selected from the National Cancer Institute’s Cancer Genome Anatomy Project SNP500Cancer Database [35]. It contains more than 3 SNPs, on average, for each gene represented on the panel. The complete list of the annotated SNPs present on the array is provided in Additional file 1: Table S1.

Selection of SNPs in DNA repair related genes

For the purpose of this study, we chose to focus the analysis of the genotyping data on candidate SNPs located within genes involved in DNA repair pathways as annotated in the Atlas of Cancer Signalling Network (ACSN) [36]. According to the ACSN, 178 out of the 1421 SNPs present on the Cancer SNP Panel array are located in a DNA repair gene. Among the 178 SNPs, 141 passed the genotyping quality controls (QCs) and had a minor allele frequency (MAF) in the control group greater than 0.05; those SNPs were included in the analyses. The list of the 141 analysed SNPs is accessible in Additional file 2: Table S2.

Statistical analyses

The raw genotyping data were imported into GenomeStudio V2011.1 (Illumina) for SNP clustering and the generation of genotype calls. The standard summary statistics used for quality control of the genotyping were performed using PLINK [37].
We excluded 22 samples with an overall call rate < 90% and 34 SNPs with a call rate < 90%. The deviation of the genotype proportions from Hardy-Weinberg equilibrium (HWE) was assessed in the controls using Chi-squared test with one degree of freedom. Doing so, 3 SNPs with p-values < 0.001 showing significant deviations from HWE and were removed. This resulted in the inclusion of a total of 141 SNPs and 329 subjects (75 cases and 254 controls) in the analyses.
Conditional logistic regressions accounting for radiation dose to the thyroid were performed with PLINK [37] to assess the contribution of genetic factors to PTC risk. SNPs were included as a log additive model (i.e. multiplicative model of inheritance), which assumes the same increment in risk for each allele at a given locus. Dominant and recessive models were also examined for SNPs showing significant association in the log-additive model. Multiple testing was adjusted for using a Bonferroni correction after linkage disequilibrium (LD)-pruning to omit highly correlated SNPs, i.e. SNP pairs with r2 ≥ 0.8. After LD pruning, the number of tested markers was reduced to 94 SNPs.
As previously described, radiation doses were transformed as log(1 + dose), with the raw dose measured in Gy, in order to approximate the linear excess risk model for small doses in the conditional logistic regression [26]. In that way, “for the rare disease approximation, the relative risk of disease is modelled as approximately (1+dose)β, which for doses less than 1 Gy is approximately 1 + β dose” [26].
In addition to the single-marker-based association tests with PLINK, we employed the Versatile Gene-based Association Study (VEGAS) [38] and PLINK set [37] methods to examine whether test statistics for a group of related SNPs or genes have consistent yet moderate deviation from chance. Both VEGAS and PLINK set-based test combine p-values from single-SNP analyses but differ in how an appropriate null distribution is obtained. The gene-based association test performed by VEGAS relies on simulation of LD structure from a reference data set (here we used our control set) whereas PLINK set-based tests resort to permutation testing. For VEGAS, we used as input data the SNP association p-values obtained from the PLINK SNP-based logistic test, and the studied controls (option “poptem”) to estimate LD structure within each gene. The set test in PLINK is related to that used for pathway analysis; however here we used it only for genes and DNA repair module-based analysis. It calculates the average of all test statistics as a module enrichment scores, using independent and significant (by preselecting p-value cut-off) SNPs in the module [39]. PLINK set test generates empirical p-values using the max (T) permutation approach for pointwise estimates. The significance level of each gene was obtained through 10,000 permutations.

Results

SNP-based analysis

Out of the 407 study participants (83 cases and 324 controls) with blood DNA available (Table 1), 75 PTC patients and 254 matched controls were successfully genotyped using the Illumina SNP Cancer Panel array. Among those, 12 cases and 16 controls had received ionizing radiation doses above 2 Gy, and were excluded from the analyses because data were too sparse to allow proper fitting of the model described previously [26]. Doing so, analysis was restricted to 63 (84%) cases and 238 (93.7%) controls.
A total of 141 SNPs located in 43 DNA repair genes were present on the SNP Cancer Panel Array, passed the genotyping quality controls, were in agreement with HWE in controls (P > 0.001) and had a MAF in the control group greater than 0.05. The 43 genes containing these SNPs are involved in distinct DNA mechanisms that can be organized into 10 functional modules, as described in the ACSN [36]. The distribution of these 43 genes, as well as the number of tested SNPs, per functional module is shown in Table 2. As indicated in the legend of Table 2 some of the tested genes are involved in several DNA repair modules.
Table 2
Distribution of the 43 DNA repair genes per DNA repair module as described in the ACSN. The number of tested SNPs per group of genes is indicated in italic and in brackets
 
NER
BER
MMR
SSA
NHEJ
MMEJ
HR
FANCONI
DR
TLS
NER
14 [30]
         
BER
8 [19]
17 [47]
        
MMR
3 [9]
6 [21]
10 [47]
       
SSA
1 [2]
1 [9]
2 [13]
4 [16]
      
NHEJ
-
1 [5]
-
-
4 [13]
     
MMEJ
5 [8]
3 [4]
2 [2]
1 [2]
-
7 [13]
    
HR
-
2 [10]
1 [1]
1 [1]
1 [5]
2 [5]
13 [47]
   
FANCONI
2 [4]
-
-
1 [2]
-
2 [4]
4 [21]
7 [34]
  
DR
-
-
-
-
-
-
-
-
1 [4]
 
TLS
1 [3]
1 [3]
1 [3]
-
-
-
2 [9]
3 [18]
-
4 [21]
NER nucleotide excision repair, BER base excision repair, MMR mismatch repair, SSA Single strand annealing, NHEJ non-homologous end joining, MMEJ microhomology-mediated end joining, HR homologous recombination, FANCONI Fanconi pathway, DR Direct repair, TLS translesion synthesis
The 43 tested genes per DNA repair modules, with number of tested SNPs per gene indicated in brackets, are the following (some genes act in several modules): NER: ERCC1 [2], ERCC2 [2], ERCC3 [2], ERCC4 [2], ERCC5 [2], ERCC6 [2], LIG1 [5], LIG3 [1], PCNA [3], POLD1 [1], RAD23B [3], XPA [1], XPC [2], XRCC1 [2]; BER: APEX1 [1], ERCC5 [2], LIG1 [5], LIG3 [1], MBD4 [1], MLH1 [2], MSH2 [9], MSH6 [1], OGG1 [2], PARP1 [5], PCNA [3], POLB [2], POLD1 [1], RAD23B [3], WRN [5], XPC [2], XRCC1 [2]; MMR: EXO1 [1], LIG1 [5], MLH1 [2], MSH2 [9], MSH3 [4], MSH6 [1], PCNA [3], PMS1 [18], PMS2 [3], POLD1 [1]; SSA: ERCC1 [2], MSH2 [9], MSH3 [4], RAD52 [1]; NHEJ: LIG4 [1], PARP1 [5], XRCC4 [3], XRCC5 [4]; MMEJ: ERCC1 [2], ERCC4 [2], EXO1 [1], LIG3 [1], NBN [4], POLD1 [1], XRCC1 [2]; HR: BARD1 [1], BLM [4], BRCA1 [7], BRCA2 [5], BRIP1 [5], EXO1 [1], NBN [4], PARP1 [5], RAD51 [7], RAD52 [1], RAD54L [1], WRN [5], XRCC3 [1]; FANCONI: BLM [4], BRCA1 [7], BRCA2 [5], BRIP1 [5], ERCC1 [2], ERCC4 [2], FANCA [9]; DR: MGMT [4]; TLS: BLM [4], BRIP1 [5], FANCA [9], PCNA [3]
Results of the single-marker association test for the 7 SNPs showing the strongest association in the log-additive model (P per allele < 0.05) are presented in Table 3. These SNPs are located in MGMT, XRCC5, ERCC5, PARP1, PCNA, PMS2 and OGG1 acting in 8 distinct DNA repair mechanisms. However, after adjustment for multiple testing, only the intronic SNP rs2296675 in MGMT acting in the DR module was significantly associated with PTC risk (Table 3). Other genetic models were also further examined for these 7 SNPs but associations did not reach statistical significance in these subsequent analyses. We also conducted a sensitivity analysis including the 28 subjects who received radiation doses above 2 Gy and results were similar (data not shown). Results of the association tests using different genetic models for the 141 DNA repair SNP and without including radiation dose in the models, are presented in Additional file 2: Table S2.
Table 3
Single marker associations with PTC (only SNPs with P per allele < 0.05 are shown)
Gene
DNA repair module
SNP
Nucleotide change
Location relative to gene
MAF in cases
MAF in controls
Genetic model
OR (95% CI) a
P a
P corr b
MGMT
DR
rs2296675
A > G
Intronic
0.21
0.10
    
      
Risk per G allele c
2.54 (1.50, 4.30)
0.0006
0.05
      
A/G + GG versus A/A d
2.72 (1.48, 5.03)
0.001
0.13
      
G/G versus A/G + AA e
5.30 (1.25, 22.48)
0.02
1
XRCC5
NHEJ
rs1051685
A > G
3’UTR
0.09
0.16
    
      
Risk per G allele c
0.39 (0.20, 0.78)
0.008
0.73
      
A/G + GG versus A/A d
0.39 (0.18, 0.83)
0.01
1
      
G/G versus A/G + AA e
N/A
N/A
N/A
ERCC5
BER, NER
rs1047768
C > T
Coding (p.His46His)
0.49
0.39
    
     
Risk per T allele c
1.58 (1.06, 2.35)
0.02
1
     
C/T + T/T versus C/C d
1.89 (1.05, 3.42)
0.03
1
     
T/T versus C/T + CC e
1.80 (0.88, 3.66)
0.11
1
PARP1
BER, HR, NHEJ
rs747659
C > T
Intergenic
0.12
0.18
    
     
Risk per T allele c
0.49 (0.26, 0.94)
0.03
1
     
C/T + T/T versus C/C d
0.49 (0.25, 0.97)
0.04
1
     
T/T versus C/T + CC e
N/A
N/A
N/A
PCNA
BER, MMR, NER, TLS
rs17349
C > T
5’UTR
0.14
0.08
    
     
Risk per T allele c
1.98 (1.07, 3.68)
0.03
1
     
C/T + T/T versus C/C d
1.93 (0.96, 3.85)
0.06
1
     
T/T versus C/T + CC e
8.29 (0.88, 78.3)
0.06
1
PMS2
MMR
rs3735295
G > A
Intronic
0.12
0.19
    
      
Risk per A allele c
0.52 (0.28, 0.97)
0.04
1
      
G/A + A/A versus G/G d
0.49 (0.24, 1.00)
0.05
1
      
A/A versus G/A + A/A e
0.33 (0.05, 2.07)
0.24
1
OGG1
BER
rs125701
G > A
Intergenic
0.27
0.21
    
      
Risk per A allele c
1.65 (1.01, 2.68)
0.04
1
      
G/A + A/A versus G/G d
1.87 (1.03, 3.40)
0.04
1
      
A/A versus G/A + A/A e
1.73 (0.47, 6.37)
0.41
1
When 2 or more SNPs in the same gene showed significant association, only the best SNP is reported
a Dose was accounted for by including the continuous variable log(1 + dose). Subjects who received more than 2.0 Gy were excluded
b P corr : Bonferroni corrected p-value
c A log-additive model (i.e. a multiplicative model of inheritance), which assumes the same increment in risk for each allele at a given locus was used
d Dominant model of inheritance (combined heterozygotes and rare homozygotes versus common homozygotes)
e Recessive model of inheritance (rare homozygotes versus combined heterozygotes and common homozygotes)
SNP single nucleotide polymorphism, MAF minor allele frequency, OR odds ratio, 95% CI 95% confidence interval, UTR untranslated region, N/A not applicable

Gene-based analysis

We then conducted a gene-based analysis using VEGAS [38] which assigns SNPs to genes and calculates gene-based empirical association p-values while accounting for the LD structure within a gene. Using this approach, two genes, namely ERCC5 and MGMT were significantly associated (P Gene < 0.05) with PTC susceptibility (Table 4), suggesting that DR, BER, and NER DNA repair mechanisms may all play a role in the development of thyroid cancer. We also used PLINK set-based test to test each of the 10 DNA repair modules and observed significant association after Bonferroni correction for module DR (data not shown).
Table 4
Results of the gene-based association test from VEGAS (only genes with P Gene < 0.05 or best SNP P per allele < 0.05 are shown)
Chr
Gene
Start
Stop
Number of tested SNPs
Number of simulations
Test
P Gene
Best SNP
P per allele
10
MGMT
131,215,453
131,615,783
4
1e + 05
13.93
0.008
rs2296675
0.0006
13
ERCC5
103,448,190
103,578,351
2
1e + 06
9.39
0.01
rs1047768
0.02
20
PCNA
5,045,598
5,157,268
3
1e + 05
9.30
0.05
rs17349
0.03
7
PMS2
5,962,869
6,098,737
4
1000
8.70
0.09
rs3735295
0.04
1
PARP1
226,498,391
226,645,801
5
1000
11.05
0.11
rs747659
0.03
3
OGG1
9,741,627
9,858,353
2
1000
4.30
0.13
rs125701
0.05
2
XRCC5
216,924,019
217,121,016
4
1000
7.79
0.13
rs1051685
0.008
Chr chromosome, Start coordinate of the 5′ end of the gene on the chromosome, Stop coordinate of the 3′ end of the gene on the chromosome.

Discussion

Understanding the aetiology of PTC and increased susceptibility to exposure to ionising radiation is an important aim for radiation protection policy. Radiation exposure during childhood is a strong risk factor for PTC, and polymorphisms in DNA repair genes are likely to affect this risk, but few studies have been designed to determine the role of such genes as modulators of PTC risk [40]. For this reason we chose to evaluate the association between a panel of common candidate SNPs located within 43 DNA repair genes and PTC. Our results showed significant association for rs2296675 located in MGMT encoding the O6-methylguanine DNA methyltransferase, and suggestive association for variants in ERCC5 encoding a single-strand specific DNA endonuclease and variants in PCNA encoding the proliferating cell nuclear antigen. Hence our findings support the involvement of several mechanisms that could be mobilised by the follicular cells of the thyroid gland to repair the different types of DNA damages that could occur after exposure to radiation.
There are several limitations to this study that should be noted. First, the power to detect genes with small effect sizes may be low due to the relatively small number of subjects included in this study because of the uniqueness of the studied population. Indeed for SNPs with low MAF in controls (5%), the power of our study for evidencing an association between a candidate SNP and PTC could reach 80% only for an OR of 3.5 or higher for P < 0.05. For SNPs with MAF in controls of about 20%, our study had a power of 80% for evidencing an association if OR is about 1.90 P < 0.05.
Second, the genotyped markers in the Illumina SNP Cancer Panel array are scarce (on average 3.6 SNPs per cancer gene on the array, and 4.1 SNPs per DNA repair) and do not cover all of variations in the candidate genes. In addition, due to small sample size, we only included markers with a MAF ≥ 0.05, and common SNPs usually have small effects. Since the most significant SNPs that we identified are non-conding variants further sequencing and functional studies are required to confirm whether the disease associations of reported markers are causal.
Nevertheless, the association found between MGMT and PTC susceptibility is a novel interesting finding. MGMT is known to be one of the most important DNA repair proteins, and it catalyzes the transfer of the methyl group from O6-methylguanine adducts of double-stranded DNA induced by the alkylating agents to the cysteine residue in its own molecule and thus prevents the transition from G:C to A:T point mutations by removing alkyl adducts from the O6 position of guanine. Loss of MGMT expression has been associated with aggressive tumour behaviour and progression in several types of neoplasia, including esophageal, hepatocellular, lung, gastric and breast carcinomas [4144]. The gene that is located at chromosome 10q26 spans nearly 300 kb of genomic DNA, where heterozygous deletion can often be observed in glioblastoma multiform patients [45].
Interestingly, the minor allele G of rs2296675 in MGMT had been previously shown to increase overall cancer risk of cancer across multiple tissues [per minor allele OR = 1.30, 95% CI 1.19,1.43, P = 4.1 × 10−8] [46], but risk of thyroid cancer was not specifically examined in the published CLUE II cohort study. Nevertheless, a link between MGMT and thyroid cancer had already been established in few studies. First, it was shown that expression level of MGMT protein was significantly downregulated in malignant compared to benign thyroid lesions [47]. Secondly, the methylation status of MGMT has also been investigated in thyroid neoplasia [48, 49]. Correlation of MGMT expression and promoter methylation with genomic instability in PTC patients had been then reported [50]. Functional studies are needed to decipher the biological properties of the methyltransferase and to understand how sequence variants may alter its functions and lead to the development of PTC. To our knowledge, the suggestive association between PCNA and PTC risk has not been observed in other populations, while a protective effect for carriers of the minor allele of rs2227869 in ERCC5, a SNP that is not present on the SNP Cancer Panel array, was reported in the Portuguese population [51]. Hence replication of the association study with a focus on MGMT, ERCC5 and PCNA SNPs in other populations, including both irradiated and non-irradiated PTC patients and matched healthy subjects is also warranted. More widely, identification of genetic modifiers of radiation-associated carcinogenesis may thus be a step forward to allow future personalized cancer risk prediction and may serve in prevention endeavours or public health response to widespread radiation exposure.

Conclusions

To conclude with, this study confirms that genetic variants in several genes operating in distinct DNA repair mechanisms are implicated in the development of PTC. In particular we report a new association between the minor allele G of SNP rs2296675 in MGMT and PTC risk in a unique population sample of Belarusian subjects who have been exposed to ionizing radiation during childhood. Further investigation is needed to decipher the functional properties of the methyltransferase encoded by this gene in order to understand how alteration of such functions may lead to the development of the most common type of thyroid cancer.

Acknowledgments

We are most grateful to all the subjects who participated in this study. We would like to thank Vanessa Tenet, Christophe Lallemand and Jocelyne Michelon for their technical expertise.

Funding

This work was supported by the European Union (Nuclear Fission Safety and INCO-Copernicus Programmes contracts FI4C-CT96–0014 and ERBIC15-CT96–0308), the Sasakawa Memorial Health Foundation (Chernobyl Sasakawa Health and Medical Cooperation Project) and Electricité de France (EDF) (grant 5500-AAP-5910065238 - DPN RB 2010–17 and grant 5100-AAP-5910078316 – DPN RB 2011–20).
These funds were used to collect biological material and epidemiological data (European Union and Sasakawa Memorial Health Foundation) and to perform SNP genotyping (EDF).

Availability of data and material

Genotyping data described in the manuscript are available from the authors upon request.

Authors’ contributions

FLC-K, AK and FL designed the research protocol; FD and GD performed genotyping; CL, FD, MM, GB, AK and FL analysed data; IM, VM, EC were involved in performing investigations, collecting data and helped conceive and design the study; FL wrote the paper. All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.
Not applicable.
Written informed consent was obtained from all participants. The study was carried out with the approval of the International Agency for Research on Cancer (IARC) ethics committee and of the Belarus Coordinating Council for Studies of the Medical Consequences of the Chernobyl Accident.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Jemal A, Siegel R, Xu J, Ward E. Cancer statistics, 2010. CA Cancer J Clin. 2010;60:277–300.CrossRefPubMed Jemal A, Siegel R, Xu J, Ward E. Cancer statistics, 2010. CA Cancer J Clin. 2010;60:277–300.CrossRefPubMed
2.
Zurück zum Zitat La Vecchia C, Malvezzi M, Bosetti C, Garavello W, Bertuccio P, Levi F, Negri E. Thyroid cancer mortality and incidence: a global overview. Int J Cancer. 2015;136:2187–95.CrossRefPubMed La Vecchia C, Malvezzi M, Bosetti C, Garavello W, Bertuccio P, Levi F, Negri E. Thyroid cancer mortality and incidence: a global overview. Int J Cancer. 2015;136:2187–95.CrossRefPubMed
3.
Zurück zum Zitat Vaccarella S, Dal Maso L, Laversanne M, Bray F, Plummer M, Franceschi S. The impact of diagnostic changes on the rise in thyroid cancer incidence: a population-based study in selected high-resource countries. Thyroid. 2015;25:1127–36.CrossRefPubMed Vaccarella S, Dal Maso L, Laversanne M, Bray F, Plummer M, Franceschi S. The impact of diagnostic changes on the rise in thyroid cancer incidence: a population-based study in selected high-resource countries. Thyroid. 2015;25:1127–36.CrossRefPubMed
4.
Zurück zum Zitat Sinnott B, Ron E, Schneider AB. Exposing the thyroid to radiation: a review of its current extent, risks, and implications. Endocr Rev. 2010;31:756–73.CrossRefPubMedPubMedCentral Sinnott B, Ron E, Schneider AB. Exposing the thyroid to radiation: a review of its current extent, risks, and implications. Endocr Rev. 2010;31:756–73.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Pellegriti G, Frasca F, Regalbuto C, Squatrito S, Vigneri R. Worldwide increasing incidence of thyroid cancer: update on epidemiology and risk factors. J Cancer Epidemiol. 2013;2013:965212.CrossRefPubMedPubMedCentral Pellegriti G, Frasca F, Regalbuto C, Squatrito S, Vigneri R. Worldwide increasing incidence of thyroid cancer: update on epidemiology and risk factors. J Cancer Epidemiol. 2013;2013:965212.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Zhu C, Zheng T, Kilfoy BA, Han X, Ma S, Ba Y, Bai Y, Wang R, Zhu Y, Zhang Y. A birth cohort analysis of the incidence of papillary thyroid cancer in the United States, 1973-2004. Thyroid. 2009;19:1061–6.CrossRefPubMedPubMedCentral Zhu C, Zheng T, Kilfoy BA, Han X, Ma S, Ba Y, Bai Y, Wang R, Zhu Y, Zhang Y. A birth cohort analysis of the incidence of papillary thyroid cancer in the United States, 1973-2004. Thyroid. 2009;19:1061–6.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Franceschi S, Boyle P, Maisonneuve P, La Vecchia C, Burt AD, Kerr DJ, MacFarlane GJ. The epidemiology of thyroid carcinoma. Crit Rev Oncog. 1993;4:25–52.PubMed Franceschi S, Boyle P, Maisonneuve P, La Vecchia C, Burt AD, Kerr DJ, MacFarlane GJ. The epidemiology of thyroid carcinoma. Crit Rev Oncog. 1993;4:25–52.PubMed
8.
Zurück zum Zitat Goldgar DE, Easton DF, Cannon-Albright LA, Skolnick MH. Systematic population-based assessment of cancer risk in first-degree relatives of cancer probands. J Natl Cancer Inst. 1994;86:1600–8.CrossRefPubMed Goldgar DE, Easton DF, Cannon-Albright LA, Skolnick MH. Systematic population-based assessment of cancer risk in first-degree relatives of cancer probands. J Natl Cancer Inst. 1994;86:1600–8.CrossRefPubMed
9.
Zurück zum Zitat Hemminki K, Li X. Familial risk of cancer by site and histopathology. Int J Cancer. 2003;103:105–9.CrossRefPubMed Hemminki K, Li X. Familial risk of cancer by site and histopathology. Int J Cancer. 2003;103:105–9.CrossRefPubMed
10.
Zurück zum Zitat Landa I, Robledo M. Association studies in thyroid cancer susceptibility: are we on the right track? J Mol Endocrinol. 2011;47:R43–58.CrossRef Landa I, Robledo M. Association studies in thyroid cancer susceptibility: are we on the right track? J Mol Endocrinol. 2011;47:R43–58.CrossRef
11.
Zurück zum Zitat Bonora E, Rizzato C, Diquigiovanni C, Oudot-Mellakh T, Campa D, Vargiolu M, Guedj M, Consortium N, JD MK, Romeo G, Canzian F, Lesueur F. The FOXE1 locus is a major genetic determinant for familial nonmedullary thyroid carcinoma. Int J Cancer. 2014;134:2098–107.CrossRefPubMed Bonora E, Rizzato C, Diquigiovanni C, Oudot-Mellakh T, Campa D, Vargiolu M, Guedj M, Consortium N, JD MK, Romeo G, Canzian F, Lesueur F. The FOXE1 locus is a major genetic determinant for familial nonmedullary thyroid carcinoma. Int J Cancer. 2014;134:2098–107.CrossRefPubMed
12.
Zurück zum Zitat Gudmundsson J, Sulem P, Gudbjartsson DF, Jonasson JG, Sigurdsson A, Bergthorsson JT, He H, Blondal T, Geller F, Jakobsdottir M, Magnusdottir DN, Matthiasdottir S, Stacey SN, Skarphedinsson OB, Helgadottir H, Li W, Nagy R, Aguillo E, Faure E, Prats E, Saez B, Martinez M, Eyjolfsson GI, Bjornsdottir US, Holm H, Kristjansson K, Frigge ML, Kristvinsson H, Gulcher JR, Jonsson T, Rafnar T, Hjartarsson H, Mayordomo JI, de la Chapelle A, Hrafnkelsson J, Thorsteinsdottir U, Kong A, Stefansson K. Common variants on 9q22.33 and 14q13.3 predispose to thyroid cancer in European populations. Nat Genet. 2009;41:1122–6. Gudmundsson J, Sulem P, Gudbjartsson DF, Jonasson JG, Sigurdsson A, Bergthorsson JT, He H, Blondal T, Geller F, Jakobsdottir M, Magnusdottir DN, Matthiasdottir S, Stacey SN, Skarphedinsson OB, Helgadottir H, Li W, Nagy R, Aguillo E, Faure E, Prats E, Saez B, Martinez M, Eyjolfsson GI, Bjornsdottir US, Holm H, Kristjansson K, Frigge ML, Kristvinsson H, Gulcher JR, Jonsson T, Rafnar T, Hjartarsson H, Mayordomo JI, de la Chapelle A, Hrafnkelsson J, Thorsteinsdottir U, Kong A, Stefansson K. Common variants on 9q22.33 and 14q13.3 predispose to thyroid cancer in European populations. Nat Genet. 2009;41:1122–6.
13.
Zurück zum Zitat Figlioli G, Elisei R, Romei C, Melaiu O, Cipollini M, Bambi F, Chen B, Kohler A, Cristaudo A, Hemminki K, Gemignani F, Forsti A, Landi S. A comprehensive meta-analysis of case-control association studies to evaluate polymorphisms associated with the risk of differentiated thyroid carcinoma. Cancer Epidemiol Biomarkers Prev. 2016;25:700–13. Figlioli G, Elisei R, Romei C, Melaiu O, Cipollini M, Bambi F, Chen B, Kohler A, Cristaudo A, Hemminki K, Gemignani F, Forsti A, Landi S. A comprehensive meta-analysis of case-control association studies to evaluate polymorphisms associated with the risk of differentiated thyroid carcinoma. Cancer Epidemiol Biomarkers Prev. 2016;25:700–13.
14.
Zurück zum Zitat Mancikova V, Cruz R, Inglada-Perez L, Fernandez-Rozadilla C, Landa I, Cameselle-Teijeiro J, Celeiro C, Pastor S, Velazquez A, Marcos R, Andia V, Alvarez-Escola C, Meoro A, Schiavi F, Opocher G, Quintela I, Ansede-Bermejo J, Ruiz-Ponte C, Santisteban P, Robledo M, Carracedo A. Thyroid cancer GWAS identifies 10q26.12 and 6q14.1 as novel susceptibility loci and reveals genetic heterogeneity among populations. Int J Cancer. 2015;137:1870–8.CrossRefPubMed Mancikova V, Cruz R, Inglada-Perez L, Fernandez-Rozadilla C, Landa I, Cameselle-Teijeiro J, Celeiro C, Pastor S, Velazquez A, Marcos R, Andia V, Alvarez-Escola C, Meoro A, Schiavi F, Opocher G, Quintela I, Ansede-Bermejo J, Ruiz-Ponte C, Santisteban P, Robledo M, Carracedo A. Thyroid cancer GWAS identifies 10q26.12 and 6q14.1 as novel susceptibility loci and reveals genetic heterogeneity among populations. Int J Cancer. 2015;137:1870–8.CrossRefPubMed
15.
Zurück zum Zitat Liyanarachchi S, Wojcicka A, Li W, Czetwertynska M, Stachlewska E, Nagy R, Hoag K, Wen B, Ploski R, Ringel MD, Kozlowicz-Gudzinska I, Gierlikowski W, Jazdzewski K, He H, de la Chapelle A. Cumulative risk impact of five genetic variants associated with papillary thyroid carcinoma. Thyroid. 2013;23:1532–40.CrossRefPubMedPubMedCentral Liyanarachchi S, Wojcicka A, Li W, Czetwertynska M, Stachlewska E, Nagy R, Hoag K, Wen B, Ploski R, Ringel MD, Kozlowicz-Gudzinska I, Gierlikowski W, Jazdzewski K, He H, de la Chapelle A. Cumulative risk impact of five genetic variants associated with papillary thyroid carcinoma. Thyroid. 2013;23:1532–40.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Williams D. Twenty years' experience with post-Chernobyl thyroid cancer. Best Pract Res Clin Endocrinol Metab. 2008;22:1061–73.CrossRefPubMed Williams D. Twenty years' experience with post-Chernobyl thyroid cancer. Best Pract Res Clin Endocrinol Metab. 2008;22:1061–73.CrossRefPubMed
17.
Zurück zum Zitat Cardis E, Kesminiene A, Ivanov V, Malakhova I, Shibata Y, Khrouch V, Drozdovitch V, Maceika E, Zvonova I, Vlassov O, Bouville A, Goulko G, Hoshi M, Abrosimov A, Anoshko J, Astakhova L, Chekin S, Demidchik E, Galanti R, Ito M, Korobova E, Lushnikov E, Maksioutov M, Masyakin V, Nerovnia A, Parshin V, Parshkov E, Piliptsevich N, Pinchera A, Polyakov S, Shabeka N, Suonio E, Tenet V, Tsyb A, Yamashita S, Williams D. Risk of thyroid cancer after exposure to 131I in childhood. J Natl Cancer Inst. 2005;97:724–32.CrossRefPubMed Cardis E, Kesminiene A, Ivanov V, Malakhova I, Shibata Y, Khrouch V, Drozdovitch V, Maceika E, Zvonova I, Vlassov O, Bouville A, Goulko G, Hoshi M, Abrosimov A, Anoshko J, Astakhova L, Chekin S, Demidchik E, Galanti R, Ito M, Korobova E, Lushnikov E, Maksioutov M, Masyakin V, Nerovnia A, Parshin V, Parshkov E, Piliptsevich N, Pinchera A, Polyakov S, Shabeka N, Suonio E, Tenet V, Tsyb A, Yamashita S, Williams D. Risk of thyroid cancer after exposure to 131I in childhood. J Natl Cancer Inst. 2005;97:724–32.CrossRefPubMed
18.
19.
Zurück zum Zitat Friedberg EC, McDaniel LD, Schultz RA. The role of endogenous and exogenous DNA damage and mutagenesis. Curr Opin Genet Dev. 2004;14:5–10.CrossRefPubMed Friedberg EC, McDaniel LD, Schultz RA. The role of endogenous and exogenous DNA damage and mutagenesis. Curr Opin Genet Dev. 2004;14:5–10.CrossRefPubMed
22.
Zurück zum Zitat Muniandy PA, Liu J, Majumdar A, Liu ST, Seidman MM. DNA interstrand crosslink repair in mammalian cells: step by step. Crit Rev Biochem Mol Biol. 2010;45:23–49.CrossRefPubMedPubMedCentral Muniandy PA, Liu J, Majumdar A, Liu ST, Seidman MM. DNA interstrand crosslink repair in mammalian cells: step by step. Crit Rev Biochem Mol Biol. 2010;45:23–49.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Gatzidou E, Michailidi C, Tseleni-Balafouta S, Theocharis S. An epitome of DNA repair related genes and mechanisms in thyroid carcinoma. Cancer Lett. 2010;290:139–47.CrossRefPubMed Gatzidou E, Michailidi C, Tseleni-Balafouta S, Theocharis S. An epitome of DNA repair related genes and mechanisms in thyroid carcinoma. Cancer Lett. 2010;290:139–47.CrossRefPubMed
24.
Zurück zum Zitat Takahashi M, Saenko VA, Rogounovitch TI, Kawaguchi T, Drozd VM, Takigawa-Imamura H, Akulevich NM, Ratanajaraya C, Mitsutake N, Takamura N, Danilova LI, Lushchik ML, Demidchik YE, Heath S, Yamada R, Lathrop M, Matsuda F, Yamashita S. The FOXE1 locus is a major genetic determinant for radiation-related thyroid carcinoma in Chernobyl. Hum Mol Genet. 2010;19:2516–23.CrossRefPubMed Takahashi M, Saenko VA, Rogounovitch TI, Kawaguchi T, Drozd VM, Takigawa-Imamura H, Akulevich NM, Ratanajaraya C, Mitsutake N, Takamura N, Danilova LI, Lushchik ML, Demidchik YE, Heath S, Yamada R, Lathrop M, Matsuda F, Yamashita S. The FOXE1 locus is a major genetic determinant for radiation-related thyroid carcinoma in Chernobyl. Hum Mol Genet. 2010;19:2516–23.CrossRefPubMed
25.
Zurück zum Zitat Akulevich NM, Saenko VA, Rogounovitch TI, Drozd VM, Lushnikov EF, Ivanov VK, Mitsutake N, Kominami R, Yamashita S. Polymorphisms of DNA damage response genes in radiation-related and sporadic papillary thyroid carcinoma. Endocr Relat Cancer. 2009;16:491–503.CrossRefPubMed Akulevich NM, Saenko VA, Rogounovitch TI, Drozd VM, Lushnikov EF, Ivanov VK, Mitsutake N, Kominami R, Yamashita S. Polymorphisms of DNA damage response genes in radiation-related and sporadic papillary thyroid carcinoma. Endocr Relat Cancer. 2009;16:491–503.CrossRefPubMed
26.
Zurück zum Zitat Damiola F, Byrnes G, Moissonnier M, Pertesi M, Deltour I, Fillon A, Le Calvez-Kelm F, Tenet V, McKay-Chopin S, McKay JD, Malakhova I, Masyakin V, Cardis E, Lesueur F, Kesminiene A. Contribution of ATM and FOXE1 (TTF2) to risk of papillary thyroid carcinoma in Belarusian children exposed to radiation. Int J Cancer. 2014;134:1659–68.CrossRefPubMed Damiola F, Byrnes G, Moissonnier M, Pertesi M, Deltour I, Fillon A, Le Calvez-Kelm F, Tenet V, McKay-Chopin S, McKay JD, Malakhova I, Masyakin V, Cardis E, Lesueur F, Kesminiene A. Contribution of ATM and FOXE1 (TTF2) to risk of papillary thyroid carcinoma in Belarusian children exposed to radiation. Int J Cancer. 2014;134:1659–68.CrossRefPubMed
27.
Zurück zum Zitat Halkova T, Dvorakova S, Sykorova V, Vaclavikova E, Vcelak J, Vlcek P, Sykorova P, Kodetova D, Betka J, Lastuvka P, Bavor P, Hoch J, Katra R, Bendlova B. Polymorphisms in selected DNA repair genes and cell cycle regulating genes involved in the risk of papillary thyroid carcinoma. Cancer Biomark. 2016;17:97–106.CrossRefPubMed Halkova T, Dvorakova S, Sykorova V, Vaclavikova E, Vcelak J, Vlcek P, Sykorova P, Kodetova D, Betka J, Lastuvka P, Bavor P, Hoch J, Katra R, Bendlova B. Polymorphisms in selected DNA repair genes and cell cycle regulating genes involved in the risk of papillary thyroid carcinoma. Cancer Biomark. 2016;17:97–106.CrossRefPubMed
28.
Zurück zum Zitat Ho T, Li G, Lu J, Zhao C, Wei Q, Sturgis EM. Association of XRCC1 polymorphisms and risk of differentiated thyroid carcinoma: a case-control analysis. Thyroid. 2009;19:129–35.CrossRefPubMed Ho T, Li G, Lu J, Zhao C, Wei Q, Sturgis EM. Association of XRCC1 polymorphisms and risk of differentiated thyroid carcinoma: a case-control analysis. Thyroid. 2009;19:129–35.CrossRefPubMed
29.
Zurück zum Zitat Santos LS, Branco SC, Silva SN, Azevedo AP, Gil OM, Manita I, Ferreira TC, Limbert E, Rueff J, Gaspar JF. Polymorphisms in base excision repair genes and thyroid cancer risk. Oncol Rep. 2012;28:1859–68.PubMed Santos LS, Branco SC, Silva SN, Azevedo AP, Gil OM, Manita I, Ferreira TC, Limbert E, Rueff J, Gaspar JF. Polymorphisms in base excision repair genes and thyroid cancer risk. Oncol Rep. 2012;28:1859–68.PubMed
30.
Zurück zum Zitat Silva SN, Gil OM, Oliveira VC, Cabral MN, Azevedo AP, Faber A, Manita I, Ferreira TC, Limbert E, Pina JE, Rueff J, Gaspar J. Association of polymorphisms in ERCC2 gene with non-familial thyroid cancer risk. Cancer Epidemiol Biomark Prev. 2005;14:2407–12.CrossRef Silva SN, Gil OM, Oliveira VC, Cabral MN, Azevedo AP, Faber A, Manita I, Ferreira TC, Limbert E, Pina JE, Rueff J, Gaspar J. Association of polymorphisms in ERCC2 gene with non-familial thyroid cancer risk. Cancer Epidemiol Biomark Prev. 2005;14:2407–12.CrossRef
31.
Zurück zum Zitat Drozdovitch V, Khrouch V, Maceika E, Zvonova I, Vlasov O, Bratilova A, Gavrilin Y, Goulko G, Hoshi M, Kesminiene A, Shinkarev S, Tenet V, Cardis E, Bouville A. Reconstruction of radiation doses in a case-control study of thyroid cancer following the Chernobyl accident. Health Phys. 2010;99:1–16.CrossRefPubMedPubMedCentral Drozdovitch V, Khrouch V, Maceika E, Zvonova I, Vlasov O, Bratilova A, Gavrilin Y, Goulko G, Hoshi M, Kesminiene A, Shinkarev S, Tenet V, Cardis E, Bouville A. Reconstruction of radiation doses in a case-control study of thyroid cancer following the Chernobyl accident. Health Phys. 2010;99:1–16.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Korobova E, Anoshko Y, Kesminiene A, Kouvyline A, Romanov S, Tenet V, Suonio E, Cardis E. Evaluation of stable iodine status of the areas affected by the Chernobyl accident in an epidemiological study in Belarus and the Russian Federation. J G Explo. 2010;107:124–35.CrossRef Korobova E, Anoshko Y, Kesminiene A, Kouvyline A, Romanov S, Tenet V, Suonio E, Cardis E. Evaluation of stable iodine status of the areas affected by the Chernobyl accident in an epidemiological study in Belarus and the Russian Federation. J G Explo. 2010;107:124–35.CrossRef
34.
Zurück zum Zitat Grimberg J, Nawoschik S, Belluscio L, McKee R, Turck A, Eisenberg A. A simple and efficient non-organic procedure for the isolation of genomic DNA from blood. Nucleic Acids Res. 1989;17:8390.CrossRefPubMedPubMedCentral Grimberg J, Nawoschik S, Belluscio L, McKee R, Turck A, Eisenberg A. A simple and efficient non-organic procedure for the isolation of genomic DNA from blood. Nucleic Acids Res. 1989;17:8390.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Packer BR, Yeager M, Burdett L, Welch R, Beerman M, Qi L, Sicotte H, Staats B, Acharya M, Crenshaw A, Eckert A, Puri V, Gerhard DS, Chanock SJ. SNP500Cancer: a public resource for sequence validation, assay development, and frequency analysis for genetic variation in candidate genes. Nucleic Acids Res. 2006;34:D617–21.CrossRefPubMed Packer BR, Yeager M, Burdett L, Welch R, Beerman M, Qi L, Sicotte H, Staats B, Acharya M, Crenshaw A, Eckert A, Puri V, Gerhard DS, Chanock SJ. SNP500Cancer: a public resource for sequence validation, assay development, and frequency analysis for genetic variation in candidate genes. Nucleic Acids Res. 2006;34:D617–21.CrossRefPubMed
36.
Zurück zum Zitat Kuperstein I, Bonnet E, Nguyen HA, Cohen D, Viara E, Grieco L, Fourquet S, Calzone L, Russo C, Kondratova M, Dutreix M, Barillot E, Zinovyev A. Atlas of cancer Signalling network: a systems biology resource for integrative analysis of cancer data with Google maps. Oncogene. 2015;4:e160.CrossRef Kuperstein I, Bonnet E, Nguyen HA, Cohen D, Viara E, Grieco L, Fourquet S, Calzone L, Russo C, Kondratova M, Dutreix M, Barillot E, Zinovyev A. Atlas of cancer Signalling network: a systems biology resource for integrative analysis of cancer data with Google maps. Oncogene. 2015;4:e160.CrossRef
37.
Zurück zum Zitat Purcell S, Neale B, Todd-Brown K, Thomas L, Ferreira MA, Bender D, Maller J, Sklar P, de Bakker PI, Daly MJ, Sham PC. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet. 2007;81:559–75.CrossRefPubMedPubMedCentral Purcell S, Neale B, Todd-Brown K, Thomas L, Ferreira MA, Bender D, Maller J, Sklar P, de Bakker PI, Daly MJ, Sham PC. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet. 2007;81:559–75.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Liu JZ, McRae AF, Nyholt DR, Medland SE, Wray NR, Brown KM, Investigators A, Hayward NK, Montgomery GW, Visscher PM, Martin NG, Macgregor S. A versatile gene-based test for genome-wide association studies. Am J Hum Genet. 2010;87:139–45.CrossRefPubMedPubMedCentral Liu JZ, McRae AF, Nyholt DR, Medland SE, Wray NR, Brown KM, Investigators A, Hayward NK, Montgomery GW, Visscher PM, Martin NG, Macgregor S. A versatile gene-based test for genome-wide association studies. Am J Hum Genet. 2010;87:139–45.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Wang K, Li M, Hakonarson H. Analysing biological pathways in genome-wide association studies. Nat Rev Genet. 2010;11:843–54.CrossRefPubMed Wang K, Li M, Hakonarson H. Analysing biological pathways in genome-wide association studies. Nat Rev Genet. 2010;11:843–54.CrossRefPubMed
40.
Zurück zum Zitat Garcia-Quispes WA, Perez-Machado G, Akdi A, Pastor S, Galofre P, Biarnes F, Castell J, Velazquez A, Marcos R. Association studies of OGG1, XRCC1, XRCC2 and XRCC3 polymorphisms with differentiated thyroid cancer. Mutat Res. 2011;709-710:67–72.CrossRefPubMed Garcia-Quispes WA, Perez-Machado G, Akdi A, Pastor S, Galofre P, Biarnes F, Castell J, Velazquez A, Marcos R. Association studies of OGG1, XRCC1, XRCC2 and XRCC3 polymorphisms with differentiated thyroid cancer. Mutat Res. 2011;709-710:67–72.CrossRefPubMed
41.
Zurück zum Zitat Matsukura S, Miyazaki K, Yakushiji H, Ogawa A, Harimaya K, Nakabeppu Y, Sekiguchi M. Expression and prognostic significance of O6-methylguanine-DNA methyltransferase in hepatocellular, gastric, and breast cancers. Ann Surg Oncol. 2001;8:807–16.CrossRefPubMed Matsukura S, Miyazaki K, Yakushiji H, Ogawa A, Harimaya K, Nakabeppu Y, Sekiguchi M. Expression and prognostic significance of O6-methylguanine-DNA methyltransferase in hepatocellular, gastric, and breast cancers. Ann Surg Oncol. 2001;8:807–16.CrossRefPubMed
42.
Zurück zum Zitat Kohya N, Miyazaki K, Matsukura S, Yakushiji H, Kitajima Y, Kitahara K, Fukuhara M, Nakabeppu Y, Sekiguchi M. Deficient expression of O(6)-methylguanine-DNA methyltransferase combined with mismatch-repair proteins hMLH1 and hMSH2 is related to poor prognosis in human biliary tract carcinoma. Ann Surg Oncol. 2002;9:371–9.PubMed Kohya N, Miyazaki K, Matsukura S, Yakushiji H, Kitajima Y, Kitahara K, Fukuhara M, Nakabeppu Y, Sekiguchi M. Deficient expression of O(6)-methylguanine-DNA methyltransferase combined with mismatch-repair proteins hMLH1 and hMSH2 is related to poor prognosis in human biliary tract carcinoma. Ann Surg Oncol. 2002;9:371–9.PubMed
43.
Zurück zum Zitat Matsukura S, Miyazaki K, Yakushiji H, Ogawa A, Chen Y, Sekiguchi M. Combined loss of expression of O6-methylguanine-DNA methyltransferase and hMLH1 accelerates progression of hepatocellular carcinoma. J Surg Oncol. 2003;82:194–200.CrossRefPubMed Matsukura S, Miyazaki K, Yakushiji H, Ogawa A, Chen Y, Sekiguchi M. Combined loss of expression of O6-methylguanine-DNA methyltransferase and hMLH1 accelerates progression of hepatocellular carcinoma. J Surg Oncol. 2003;82:194–200.CrossRefPubMed
44.
Zurück zum Zitat Sawhney M, Rohatgi N, Kaur J, Gupta SD, Deo SV, Shukla NK, Ralhan R. MGMT expression in oral precancerous and cancerous lesions: correlation with progression, nodal metastasis and poor prognosis. Oral Oncol. 2007;43:515–22.CrossRefPubMed Sawhney M, Rohatgi N, Kaur J, Gupta SD, Deo SV, Shukla NK, Ralhan R. MGMT expression in oral precancerous and cancerous lesions: correlation with progression, nodal metastasis and poor prognosis. Oral Oncol. 2007;43:515–22.CrossRefPubMed
45.
Zurück zum Zitat Zhong Y, Huang Y, Huang Y, Zhang T, Ma C, Zhang S, Fan W, Chen H, Qian J, Lu D. Effects of O6-methylguanine-DNA methyltransferase (MGMT) polymorphisms on cancer: a meta-analysis. Mutagenesis. 2010;25:83–95.CrossRefPubMed Zhong Y, Huang Y, Huang Y, Zhang T, Ma C, Zhang S, Fan W, Chen H, Qian J, Lu D. Effects of O6-methylguanine-DNA methyltransferase (MGMT) polymorphisms on cancer: a meta-analysis. Mutagenesis. 2010;25:83–95.CrossRefPubMed
46.
Zurück zum Zitat Alberg AJ, Jorgensen TJ, Ruczinski I, Wheless L, Shugart YY, Berthier-Schaad Y, Kessing B, Hoffman-Bolton J, Helzlsouer KJ, Kao WH, Francis L, Alani RM, Smith MW, Strickland PT. DNA repair gene variants in relation to overall cancer risk: a population-based study. Carcinogenesis. 2013;34:86–92.CrossRefPubMed Alberg AJ, Jorgensen TJ, Ruczinski I, Wheless L, Shugart YY, Berthier-Schaad Y, Kessing B, Hoffman-Bolton J, Helzlsouer KJ, Kao WH, Francis L, Alani RM, Smith MW, Strickland PT. DNA repair gene variants in relation to overall cancer risk: a population-based study. Carcinogenesis. 2013;34:86–92.CrossRefPubMed
47.
Zurück zum Zitat Giaginis C, Michailidi C, Stolakis V, Alexandrou P, Tsourouflis G, Klijanienko J, Delladetsima I, Theocharis S. Expression of DNA repair proteins MSH2, MLH1 and MGMT in human benign and malignant thyroid lesions: an immunohistochemical study. Med Sci Monit. 2011;17:BR81–90.PubMedPubMedCentral Giaginis C, Michailidi C, Stolakis V, Alexandrou P, Tsourouflis G, Klijanienko J, Delladetsima I, Theocharis S. Expression of DNA repair proteins MSH2, MLH1 and MGMT in human benign and malignant thyroid lesions: an immunohistochemical study. Med Sci Monit. 2011;17:BR81–90.PubMedPubMedCentral
48.
Zurück zum Zitat 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.CrossRefPubMed 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.CrossRefPubMed
49.
Zurück zum Zitat 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.CrossRefPubMed 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.CrossRefPubMed
50.
Zurück zum Zitat Santos JC, Bastos AU, Cerutti JM, Ribeiro ML. Correlation of MLH1 and MGMT expression and promoter methylation with genomic instability in patients with thyroid carcinoma. BMC Cancer. 2013;13:79.CrossRefPubMedPubMedCentral Santos JC, Bastos AU, Cerutti JM, Ribeiro ML. Correlation of MLH1 and MGMT expression and promoter methylation with genomic instability in patients with thyroid carcinoma. BMC Cancer. 2013;13:79.CrossRefPubMedPubMedCentral
51.
Zurück zum Zitat Santos LS, Gomes BC, Gouveia R, Silva SN, Azevedo AP, Camacho V, Manita I, Gil OM, Ferreira TC, Limbert E, Rueff J, Gaspar JF. The role of CCNH Val270Ala (rs2230641) and other nucleotide excision repair polymorphisms in individual susceptibility to well-differentiated thyroid cancer. Oncol Rep. 2013;30:2458–66.PubMed Santos LS, Gomes BC, Gouveia R, Silva SN, Azevedo AP, Camacho V, Manita I, Gil OM, Ferreira TC, Limbert E, Rueff J, Gaspar JF. The role of CCNH Val270Ala (rs2230641) and other nucleotide excision repair polymorphisms in individual susceptibility to well-differentiated thyroid cancer. Oncol Rep. 2013;30:2458–66.PubMed
Metadaten
Titel
Investigation of DNA repair-related SNPs underlying susceptibility to papillary thyroid carcinoma reveals MGMT as a novel candidate gene in Belarusian children exposed to radiation
verfasst von
Christine Lonjou
Francesca Damiola
Monika Moissonnier
Geoffroy Durand
Irina Malakhova
Vladimir Masyakin
Florence Le Calvez-Kelm
Elisabeth Cardis
Graham Byrnes
Ausrele Kesminiene
Fabienne Lesueur
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2017
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-017-3314-5

Weitere Artikel der Ausgabe 1/2017

BMC Cancer 1/2017 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.