Research Paper Volume 10, Issue 10 pp 2944—2953

XRCC1 gene polymorphisms and risk of neuroblastoma in Chinese children

Jiao Zhang1, , Zhenjian Zhuo2, , Wenya Li1, , Jinhong Zhu3, , Jing He4, , Jinsong Su5, ,

  • 1 Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
  • 2 School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China
  • 3 Department of Clinical Laboratory, Molecular Epidemiology Laboratory, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
  • 4 Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
  • 5 Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
* Equal contribution

Received: August 27, 2018       Accepted: October 15, 2018       Published: October 25, 2018      

https://doi.org/10.18632/aging.101601
How to Cite

Copyright: © 2018 Zhang et al. This is an open access article distributed under the terms of the Creative Commons Attribution License (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Abstract

Neuroblastoma is a common pediatric extra-cranial tumor of the sympathetic nervous system. XRCC1 is a scaffold protein that participates in DNA single-strand break repair by complexing with other proteins. XRCC1 gene polymorphisms are being increasingly explored in cancer epidemiology studies. However, the contribution of XRCC1 gene polymorphisms to neuroblastoma risk remains unclarified. Herein, we conducted a case-control study with 393 neuroblastoma patients and 812 controls to explore the association of XRCC1 gene polymorphisms (rs1799782 G>A, rs25487 C>T, rs25489 C>T and rs915927 T>C) with neuroblastoma risk. Results showed that none of the studied polymorphisms was associated with neuroblastoma risk. However, individuals with 2 risk genotypes seemed to be at significantly higher risk for neuroblastoma compared with those without risk genotype (adjusted odds ratio=1.69; 95% confidence interval=1.06-2.69). Stratified analysis revealed that the XRCC1 rs25489 CT/TT was strongly associated with reduced risk of neuroblastoma in the children ≤ 18 months of age and subgroup with clinical stage I+II+4s diseases, compared with CC genotypes. We also identified an increased neuroblastoma risk for carrier of 2-3 risk genotypes among children ≤ 18 months of age and subgroup with clinical stage I+II+4s. More evidence of the association between XRCC1 gene polymorphisms and neuroblastoma risk is needed.

Introduction

Neuroblastoma is an extracranial solid tumor of the developing sympathetic nervous system [1,2]. This disease is the most common cancer in children under age of 1 year old, and accounts for about 7% of all childhood malignancies [2]. The prognosis of neuroblastoma varies greatly from spontaneous regression without chemotherapy to aggressive, incurable disease [3,4]. Approximately, 50% of affected children are diagnosed with a localized low- or intermediate- risk disease while rest of patients are of metastatic high-risk neuroblastomas [46]. The long-term survival rates of high-risk neuroblastoma cases remain disappointingly low despite of great advances in treatments [68].

The etiology of neuroblastoma is not fully elucidated [4]. Previous evidence has suggested that environmental risk factors for neuroblastoma, but no direct linkage is established in between [9,10]. Neuroblastoma is characterized by genomic instability and chromosomal abnormalities. Growing evidence indicated that the carcinogenesis of neuroblastoma is also partially attributed to the genetic factors and the gene-environment interactions. For instance, amplification of MYCN is observed in approximately 20% of neuroblastoma cases and associated with poor prognosis [11], and ALK mutation is identified in nearly 1% of neuroblastoma cases [12,13]. Recently, genome-wide association studies (GWASs) identified several neuroblastoma susceptibility loci [1419]. Candidate gene approaches and fine mapping analysis also revealed that genetic variants in the NEFL [20], CNKN1B [21], and BARD1 gene [22] are associated with neuroblastoma susceptibility.

X-ray repair cross-complementing group 1 (XRCC1) was the first identified mammalian gene that is involved in the single strand DNA break repair [23]. XRCC1 serves as the scaffold protein without catalytic activity; alternatively, it facilitates single strand DNA break repair by cooperating with other proteins such as DNA polymerase β and DNA ligase III [2426]. XRCC1 is mainly responsible for repairing DNA damage caused by ionizing radiation, alkylation, and UV exposure [2729]. The abnormal function of XRCC1 protein could cause human cells more vulnerable to ionizing radiation and DNA-damaging agents. The associations between XRCC1 polymorphisms and the risk of cancer have been widely investigated, including lung cancer [30], gastric cancer [31], and colorectal cancer [32]. However, no such association study has been performed in neuroblastoma. Give the significant role of XRCC1 in DNA repair, we conducted a case-control study to explore the association between XRCC1 polymorphisms and risk of neuroblastoma.

Results

XRCC1 gene polymorphisms and neuroblastoma susceptibility

The characteristics of participants were described in our previous studies (Supplementary Table 1) [3336]. The genotype frequencies of the four XRCC1 polymorphisms (rs1799782 G>A, rs25487 C>T, rs25489 C>T and rs915927 T>C) in the neuroblastoma cases and controls are presented in Table 1. In the single locus analysis, none of the analyzed polymorphism was significantly associated with neuroblastoma risk. In analyzing the combined effect of risk genotypes, we found that subjects carrying 2 risk genotypes had a significantly increased neuroblastoma risk at an adjusted odds ratio (OR) of 1.69 [95% confidence interval (CI)=1.06-2.69] when compared with those carrying 0 risk genotype.

Table 1. Association between XRCC1 gene polymorphisms and neuroblastoma susceptibility.

GenotypeCases
(N=393)
Controls
(N=812)
PaCrude OR
(95% CI)
PAdjusted OR
(95% CI) b
Pb
rs1799782 G>A
GG182 (46.31)406 (50.00)1.001.00
GA177 (45.04)333 (41.01)1.19 (0.92-1.53)0.1861.19 (0.92-1.53)0.187
AA34 (8.65)73 (8.99)1.04 (0.67-1.62)0.8661.04 (0.67-1.62)0.872
Additive0.4091.08 (0.90-1.30)0.3991.08 (0.90-1.30)0.402
Dominant211 (53.69)406 (50.00)0.2301.16 (0.91-1.48)0.2301.16 (0.91-1.48)0.231
Recessive359 (91.35)739 (91.01)0.8460.96 (0.63-1.47)0.8470.96 (0.63-1.47)0.841
rs25487 C>T
CC221 (56.23)471 (58.00)1.001.00
CT151 (38.42)278 (34.24)1.16 (0.90-1.49)0.2601.16 (0.90-1.50)0.257
TT21 (5.34)63 (7.76)0.71 (0.42-1.19)0.1970.71 (0.42-1.20)0.199
Additive0.1600.98 (0.81-1.19)0.8670.98 (0.81-1.20)0.874
Dominant172 (43.77)341 (42.00)0.5601.08 (0.84-1.37)0.5601.08 (0.84-1.37)0.554
Recessive372 (94.66)749 (92.24)0.1230.67 (0.40-1.12)0.1250.67 (0.40-1.12)0.126
rs25489 C>T
CC326 (82.95)636 (78.33)1.001.00
CT64 (16.28)170 (20.94)0.73 (0.54-1.01)0.0560.73 (0.53-1.01)0.055
TT3 (0.76)6 (0.74)0.98 (0.24-3.93)0.9720.99 (0.25-4.01)0.994
Additive0.1600.77 (0.57-1.03)0.0790.77 (0.57-1.03)0.078
Dominant67 (17.05)176 (21.67)0.0610.74 (0.54-1.01)0.0610.74 (0.54-1.01)0.060
Recessive390 (99.24)806 (99.26)0.9631.03 (0.26-4.15)0.9631.05 (0.26-4.24)0.942
rs915927 T>C
TT294 (74.81)627 (77.22)1.001.00
TC97 (24.68)169 (20.81)1.22 (0.92-1.63)0.1651.23 (0.92-1.64)0.158
CC2 (0.51)16 (1.97)0.27 (0.06-1.17)0.0790.27 (0.06-1.16)0.078
Additive0.0561.04 (0.81-1.35)0.7401.05 (0.81-1.35)0.729
Dominant99 (25.19)185 (22.78)0.3561.14 (0.86-1.51)0.3561.15 (0.86-1.52)0.345
Recessive391 (99.49)796 (98.03)0.0500.26 (0.06-1.11)0.0690.25 (0.06-1.11)0.068
Combined effect of risk genotypes c
031 (7.89)90 (11.08)1.001.00
1240 (61.07)509 (62.68)1.37 (0.89-2.12)0.1581.38 (0.89-2.13)0.150
2121 (30.79)210 (25.86)1.67 (1.05-2.66)0.0301.69 (1.06-2.69)0.028
31 (0.25)3 (0.37)0.97 (0.10-9.65)0.9780.96 (0.10-9.54)0.969
Trend0.1541.25 (1.02-1.53)0.0321.25 (1.02-1.54)0.030
0-1271 (68.96)599 (73.77)1.001.00
2-3122 (31.04)213 (26.23)0.0811.27 (0.97-1.65)0.0811.27 (0.97-1.65)0.078
OR: odds ratio; CI: confidence interval. aχ2 test for genotype distributions between neuroblastoma patients and controls. b Adjusted for age and gender. c The risk genotypes were rs1799782 GA/AA, rs25487 CT/TT, rs25489 TT and rs915927 TC/CC.

Stratification analysis

We then further conducted stratification analysis for rs25489 C>T by age, gender, tumor sites of origin and clinical stages (Table 2). Compared to the rs25489 CC genotype, the protective effect of CT/TT genotypes was more predominant for children ≤ 18 months of age (adjusted OR=0.54; 95% CI=0.31-0.95) and those with clinical stage I+II+4s disease (adjusted OR=0.53, 95% CI=0.33-0.87). With the 0-1 risk genotypes as the reference group, the presence of 2-3 risk genotypes was associated with an elevated neuroblastoma risk among children ≤ 18 months of age (adjusted OR=1.87; 95% CI=1.19-2.93) and those with clinical stage I+II+4s tumor (adjusted OR=1.56, 95% CI=1.09-2.23).

Table 2. Stratification analysis of XRCC1 rs25489 C>T polymorphism and combined risk genotypes with neuroblastoma susceptibility.

Variablesrs25489
(cases/controls)
OR (95% CI)PAOR(95% CI) aPaCombined
(cases/controls)
OR (95% CI)PAOR (95% CI)aPa
CCCT/TT0-12-3
Age, month
≤18108/233
(27.48/28.69)
18/72
(4.58/8.87)
0.54
(0.31-0.95)
0.0320.54
(0.31-0.95)
0.03281/23545/701.87
(1.19-2.93)
0.0071.87
(1.19-2.93)
0.007
>18218/403
(55.47/49.63)
49/104
(12.47/12.81)
0.87
(0.60-1.27)
0.4730.87
(0.60-1.27)
0.469190/36477/1431.03
(0.74-1.43)
0.8521.03
(0.74-1.43)
0.851
Gender
Females141/262
(35.88/32.27)
27/80
(6.87/9.85)
0.63
(0.39-1.02)
0.0580.63
(0.39-1.02)
0.058115/25253/901.29
(0.86-1.93)
0.2171.28
(0.86-1.93)
0.227
Males185/374
(47.07/46.06)
40/96
(10.18/11.82)
0.84
(0.56-1.27)
0.4110.84
(0.56-1.26)
0.402156/34769/1231.25
(0.88-1.77)
0.2151.25
(0.88-1.78)
0.206
Sites of origin
Adrenal gland127/636
(32.31/78.32)
26/176
(6.62/21.67)
0.74
(0.47-1.17)
0.1930.73
(0.47-1.16)
0.183107/59946/2131.21
(0.83-1.77)
0.3271.22
(0.83-1.78)
0.313
Retroperitoneal74/636
(18.83/78.32)
13/176
(3.31/21.67)
0.64
(0.34-1.17)
0.1460.64
(0.35-1.18)
0.15160/59927/2131.27
(0.78-2.05)
0.3371.27
(0.78-2.05)
0.332
Mediastinum90/636
(22.90/78.32)
19/176
(4.83/21.67)
0.76
(0.45-1.29)
0.3100.75
(0.45-1.27)
0.28275/59934/2131.28
(0.83-1.97)
0.2731.28
(0.83-1.98)
0.265
Others27/636
(6.87/78.32)
9/176
(2.29/21.67)
1.21
(0.56-2.61)
0.6371.21
(0.56-2.62)
0.63623/59913/2131.59
(0.79-3.19)
0.1931.60
(0.80-3.21)
0.189
Clinical stages
I+II+4s141/636
(35.88/78.32)
21/176
(5.34/21.67)
0.54
(0.33-0.88)
0.0130.53
(0.33-0.87)
0.011104/59958/2131.57
(1.10-2.24)
0.0141.56
(1.09-2.23)
0.015
III+IV168/636
(42.75/78.32)
43/176
(10.94/21.67)
0.93
(0.64-1.35)
0.6830.93
(0.64-1.35)
0.698154/59957/2131.04
(0.74-1.47)
0.8171.04
(0.74-1.47)
0.823
AOR: adjusted odds ratio; CI: confidence interval. a Adjusted for age and gender in logistic regress models, without the corresponding stratify factor.

Discussion

In this two-center Chinese population study, we examined the impact of polymorphisms in the XRCC1 gene on neuroblastoma susceptibility. We found that XRCC1 polymorphisms might play a low-penetrant role on the neuroblastoma risk.

XRCC1 gene is located on chromosome 19q13.2 and encodes a 633 amino acid protein [37]. It consists of 17 exons, spanning a genomic distance of 32 kb. By now, more than 8,453 polymorphisms in the XRCC1 have been identified. Among them, three common polymorphisms, rs25487 C>T (Arg399Gln), rs25489 C>T (Arg280His), and rs1799782 G>A (Arg194Trp), are mostly investigated. These three polymorphisms could cause amino acid changes in evolutionarily conserved regions [23,38], and may further lead to the alteration in the XRCC1 protein activity. Previous study showed that XRCC1 polymorphism 399Gln allele might affect the function of XRCC1 protein, consequently impairing DNA repair capacity [39].

Increasing epidemiological studies have been performed regarding the association between XRCC1 polymorphisms and cancer risk. Hong et al. observed that rs25487 T allele and the 3 common allele combinations (rs25487 C>T, rs25489 C>T, and rs1799782 G>A) were associated with an increased risk of colorectal cancer in a study of 209 colorectal cancer cases and 209 controls recruited from Korean [40]. Divine et al. reported that XRCC1 rs25487 TT genotype conferred susceptibility to lung adenocarcinoma at an OR of 2.8 (95% CI=1.2-7.9) in a case-control study [41]. In a population-based case-control study conducted among Korean population, Lee et al. found that none of the polymorphisms (rs25487 C>T, rs25489 C>T, and rs1799782 G>A) contributed to the risk of gastric cancer [42]. Duellet al. also reported a null association between XRCC1 rs1799782 G>A and breast cancer risk in a population-based case-control study in North Carolina [43]. However, they observed a positive association for rs25487 CT or TT genotypes compared with CC among African Americans, but not Caucasians. This conflicting role of XRCC1 polymorphisms indicates their risk effects may depend on cancer types and ethnic groups.

In the present study, we found that none of the four polymorphisms of XRCC1 gene were associated with the risk of neuroblastoma. Failure to detect the association between XRCC1 polymorphisms and neuroblastoma risk might be due to the small sample size and the low-penetrant effect of common polymorphisms. In the combined analysis, subjects carrying 2 risk genotypes tend to have increased neuroblastoma risk, when those without risk genotypes serve as a reference. Stratification analysis revealed that the rs25489 CT/TT is associated with favorable clinical markers such as ≤ 18 months of age and clinical stage I+II+4s. While exploring cumulative effects of risk genotypes, we found that the presence of 2-3 risk genotypes was associated with neuroblastoma risk among children ≤ 18 months of age and those with clinical stage I+II+4s tumors. It is believed that cancer susceptibility results from interactions of genetic and environmental factors and is subjected to modification of clinical markers such as age and clinical stage; in other words, the effect of one SNP on cancer susceptibility maybe modified by other genetic variants, environment exposures, and clinical makers. Single-locus analysis in the current study only focused on one genetic factor. the rs25489. Stratified analysis by age and clinical stage involve the effects of the rs25489 and clinical makers. While SNPs were combined, stratified analysis by age and clinical stage involve interaction of multiple SNPs and clinical makers. It is reasonable that analyses involved in different variables may lead to different results. Therefore, results considering interplays of gene-gene, gene-environmental factors and clinical makers maybe more meaningful in the clinical setting. Alternatively, these results could be chance findings, which calls for larger validation studies.

The study was limited by several points. Firstly, the relative small number of participants in the study impaired the statistical power. Therefore, the null association obtained in this study should be cautiously explained. Secondly, the insufficient information about environmental factors limited our ability to analyze gene-environment interactions. Thirdly, only four polymorphisms were investigated in the current study. More polymorphisms should be tested in the future work. Fourthly, due to the restriction to Chinese descent, these findings cannot be directly extrapolated to other ethnicities before validation studies. Last, our work lacks of in vitro data to support a role for these XRCC1 polymorphisms in neuroblastoma, which should be completed in the future.

In conclusion, we report that polymorphisms in XRCC1 gene may exert a low-penetrant effect on neuroblastoma risk in Chinese population. Our study helps to provide novel insight into the role of XRCC1 gene polymorphisms in neuroblastoma risk. The findings should be further confirmed in other independent case-control studies.

Materials and Methods

Study populations

Details of the selection criteria and characteristics of the study participants were provided elsewhere [3336]. In brief, 393 neuroblastoma cases and 812 healthy controls were included in this study. The cases were from two medical centers, Guangzhou Women and Children’s Medical Center and the First Affiliated Hospital of Zhengzhou University. All the controls were selected from the same region as cases during the same period. Informed consent was obtained at the time of enrollment. This study was conducted under the approval of the Institutional Review Board of the participating hospitals.

Polymorphism selection

Potentially functional polymorphisms in the XRCC1 gene were selected using dbSNP database (http://www.ncbi.nlm.nih.gov/) and SNPinfo (http://snpinfo.niehs.nih.gov/) [44]. In final, four polymorphisms: rs1799782 G>A, rs25487 C>T, rs25489 C>T and rs915927 T>C, were adopted for analysis. As shown in our Supplementary Figure 1, there was no significant LD (R2<0.8) among these SNPs of XRCC1 (R2=0.032 between rs25487 and rs25489, R2=0.039 between rs25487 and rs915927, R2=0.122 between rs25487 and rs1799782; R2=0.014 between rs25489 and rs915927, R2=0.043 between rs25489 and rs1799782, R2=0.046 between rs915927 and rs1799782).

Genotyping

We used a TIANamp Blood DNA Kit (TianGen Biotech Co. Ltd., Beijing, China) for DNA extraction. Polymorphisms were genotyped applying TaqMan platform (Applied Biosystem, Foster City, CA, USA) without knowing the status of the participants [4547]. Quality control was conducted by including eight negative controls with water in each plate. We also randomly selected 10% of the samples to perform a second genotyping, and the concordance rate was 100%.

Statistical analysis

The Hardy-Weinberg equilibrium (HWE) was performed for each polymorphism among controls using the goodness-of-fit χ2 test. Two-sided chi-square test was applied to compare the demographic variables between the case group and controls. Tests for association between XRCC1 polymorphisms and neuroblastoma risk were conducted by comparing the genotype frequencies between cases and controls using the χ2 test. We also applied stratified analyses by age, gender, tumor sites, and clinical stages. All analyses were conducted with SAS release 9.4 (SAS Institute, Cary, NC). P< 0.05 was defined as statistically significant.

Conflicts of Interest

The authors declare no conflict of interest.

Funding

This work was supported by grants from National Natural Science Foundation of China (No: 81502187), and Pearl River S&T Nova Program of Guangzhou (No: 201710010086).

References

  • 1. Maris JM, Hogarty MD, Bagatell R, Cohn SL. Neuroblastoma. Lancet. 2007; 369:2106–20. https://doi.org/10.1016/S0140-6736(07)60983-0 [PubMed]
  • 2. Maris JM. Recent advances in neuroblastoma. N Engl J Med. 2010; 362:2202–11. https://doi.org/10.1056/NEJMra0804577 [PubMed]
  • 3. Capasso M, Diskin SJ. Genetics and genomics of neuroblastoma. Cancer Treat Res. 2010; 155:65–84. https://doi.org/10.1007/978-1-4419-6033-7_4 [PubMed]
  • 4. Cheung NK, Dyer MA. Neuroblastoma: developmental biology, cancer genomics and immunotherapy. Nat Rev Cancer. 2013; 13:397–411. https://doi.org/10.1038/nrc3526 [PubMed]
  • 5. Matthay KK, Maris JM, Schleiermacher G, Nakagawara A, Mackall CL, Diller L, Weiss WA. Neuroblastoma. Nat Rev Dis Primers. 2016; 2:16078. https://doi.org/10.1038/nrdp.2016.78 [PubMed]
  • 6. Irwin MS, Park JR. Neuroblastoma: paradigm for precision medicine. Pediatr Clin North Am. 2015; 62:225–56. https://doi.org/10.1016/j.pcl.2014.09.015 [PubMed]
  • 7. Park JR, Bagatell R, Cohn SL, Pearson AD, Villablanca JG, Berthold F, Burchill S, Boubaker A, McHugh K, Nuchtern JG, London WB, Seibel NL, Lindwasser OW, et al. Revisions to the International Neuroblastoma Response Criteria: A Consensus Statement From the National Cancer Institute Clinical Trials Planning Meeting. J Clin Oncol. 2017; 35:2580–87. https://doi.org/10.1200/JCO.2016.72.0177 [PubMed]
  • 8. Esposito MR, Aveic S, Seydel A, Tonini GP. Neuroblastoma treatment in the post-genomic era. J Biomed Sci. 2017; 24:14. https://doi.org/10.1186/s12929-017-0319-y [PubMed]
  • 9. Menegaux F, Olshan AF, Neglia JP, Pollock BH, Bondy ML. Day care, childhood infections, and risk of neuroblastoma. Am J Epidemiol. 2004; 159:843–51. https://doi.org/10.1093/aje/kwh111 [PubMed]
  • 10. Cook MN, Olshan AF, Guess HA, Savitz DA, Poole C, Blatt J, Bondy ML, Pollock BH. Maternal medication use and neuroblastoma in offspring. Am J Epidemiol. 2004; 159:721–31. https://doi.org/10.1093/aje/kwh108 [PubMed]
  • 11. Huang M, Weiss WA. Neuroblastoma and MYCN. Cold Spring Harb Perspect Med. 2013; 3:a014415. https://doi.org/10.1101/cshperspect.a014415 [PubMed]
  • 12. Chen Y, Takita J, Choi YL, Kato M, Ohira M, Sanada M, Wang L, Soda M, Kikuchi A, Igarashi T, Nakagawara A, Hayashi Y, Mano H, Ogawa S. Oncogenic mutations of ALK kinase in neuroblastoma. Nature. 2008; 455:971–74. https://doi.org/10.1038/nature07399 [PubMed]
  • 13. Mossé YP, Laudenslager M, Longo L, Cole KA, Wood A, Attiyeh EF, Laquaglia MJ, Sennett R, Lynch JE, Perri P, Laureys G, Speleman F, Kim C, et al. Identification of ALK as a major familial neuroblastoma predisposition gene. Nature. 2008; 455:930–35. https://doi.org/10.1038/nature07261 [PubMed]
  • 14. Maris JM, Mosse YP, Bradfield JP, Hou C, Monni S, Scott RH, Asgharzadeh S, Attiyeh EF, Diskin SJ, Laudenslager M, Winter C, Cole KA, Glessner JT, et al. Chromosome 6p22 locus associated with clinically aggressive neuroblastoma. N Engl J Med. 2008; 358:2585–93. https://doi.org/10.1056/NEJMoa0708698 [PubMed]
  • 15. Capasso M, Devoto M, Hou C, Asgharzadeh S, Glessner JT, Attiyeh EF, Mosse YP, Kim C, Diskin SJ, Cole KA, Bosse K, Diamond M, Laudenslager M, et al. Common variations in BARD1 influence susceptibility to high-risk neuroblastoma. Nat Genet. 2009; 41:718–23. https://doi.org/10.1038/ng.374 [PubMed]
  • 16. Nguyen B, Diskin SJ, Capasso M, Wang K, Diamond MA, Glessner J, Kim C, Attiyeh EF, Mosse YP, Cole K, Iolascon A, Devoto M, Hakonarson H, et al. Phenotype restricted genome-wide association study using a gene-centric approach identifies three low-risk neuroblastoma susceptibility Loci. PLoS Genet. 2011; 7:e1002026. https://doi.org/10.1371/journal.pgen.1002026 [PubMed]
  • 17. Wang K, Diskin SJ, Zhang H, Attiyeh EF, Winter C, Hou C, Schnepp RW, Diamond M, Bosse K, Mayes PA, Glessner J, Kim C, Frackelton E, et al. Integrative genomics identifies LMO1 as a neuroblastoma oncogene. Nature. 2011; 469:216–20. https://doi.org/10.1038/nature09609 [PubMed]
  • 18. Diskin SJ, Capasso M, Schnepp RW, Cole KA, Attiyeh EF, Hou C, Diamond M, Carpenter EL, Winter C, Lee H, Jagannathan J, Latorre V, Iolascon A, et al. Common variation at 6q16 within HACE1 and LIN28B influences susceptibility to neuroblastoma. Nat Genet. 2012; 44:1126–30. https://doi.org/10.1038/ng.2387 [PubMed]
  • 19. Chang X, Zhao Y, Hou C, Glessner J, McDaniel L, Diamond MA, Thomas K, Li J, Wei Z, Liu Y, Guo Y, Mentch FD, Qiu H, et al. Common variants in MMP20 at 11q22.2 predispose to 11q deletion and neuroblastoma risk. Nat Commun. 2017; 8:569. https://doi.org/10.1038/s41467-017-00408-8 [PubMed]
  • 20. Capasso M, Diskin S, Cimmino F, Acierno G, Totaro F, Petrosino G, Pezone L, Diamond M, McDaniel L, Hakonarson H, Iolascon A, Devoto M, Maris JM. Common genetic variants in NEFL influence gene expression and neuroblastoma risk. Cancer Res. 2014; 74:6913–24. https://doi.org/10.1158/0008-5472.CAN-14-0431 [PubMed]
  • 21. Capasso M, McDaniel LD, Cimmino F, Cirino A, Formicola D, Russell MR, Raman P, Cole KA, Diskin SJ. The functional variant rs34330 of CDKN1B is associated with risk of neuroblastoma. J Cell Mol Med. 2017; 21:3224–30. https://doi.org/10.1111/jcmm.13226 [PubMed]
  • 22. Cimmino F, Avitabile M, Diskin SJ, Vaksman Z, Pignataro P, Formicola D, Cardinale A, Testori A, Koster J, de Torres C, Devoto M, Maris JM, Iolascon A, Capasso M. Fine mapping of 2q35 high-risk neuroblastoma locus reveals independent functional risk variants and suggests full-length BARD1 as tumor-suppressor. Int J Cancer. 2018 August 21. https://doi.org/10.1002/ijc.31822 [PubMed]
  • 23. Lamerdin JE, Montgomery MA, Stilwagen SA, Scheidecker LK, Tebbs RS, Brookman KW, Thompson LH, Carrano AV. Genomic sequence comparison of the human and mouse XRCC1 DNA repair gene regions. Genomics. 1995; 25:547–54. https://doi.org/10.1016/0888-7543(95)80056-R [PubMed]
  • 24. Masson M, Niedergang C, Schreiber V, Muller S, Menissier-de Murcia J, de Murcia G. XRCC1 is specifically associated with poly(ADP-ribose) polymerase and negatively regulates its activity following DNA damage. Mol Cell Biol. 1998; 18:3563–71. https://doi.org/10.1128/MCB.18.6.3563 [PubMed]
  • 25. Kubota Y, Nash RA, Klungland A, Schär P, Barnes DE, Lindahl T. Reconstitution of DNA base excision-repair with purified human proteins: interaction between DNA polymerase beta and the XRCC1 protein. EMBO J. 1996; 15:6662–70. https://doi.org/10.1002/j.1460-2075.1996.tb01056.x [PubMed]
  • 26. Nash RA, Caldecott KW, Barnes DE, Lindahl T. XRCC1 protein interacts with one of two distinct forms of DNA ligase III. Biochemistry. 1997; 36:5207–11. https://doi.org/10.1021/bi962281m [PubMed]
  • 27. Thompson LH, West MG. XRCC1 keeps DNA from getting stranded. Mutat Res. 2000; 459:1–18. https://doi.org/10.1016/S0921-8777(99)00058-0 [PubMed]
  • 28. Caldecott KW. XRCC1 and DNA strand break repair. DNA Repair (Amst). 2003; 2:955–69. https://doi.org/10.1016/S1568-7864(03)00118-6 [PubMed]
  • 29. Shen MR, Zdzienicka MZ, Mohrenweiser H, Thompson LH, Thelen MP. Mutations in hamster single-strand break repair gene XRCC1 causing defective DNA repair. Nucleic Acids Res. 1998; 26:1032–37. https://doi.org/10.1093/nar/26.4.1032 [PubMed]
  • 30. Tang J, Zhao J, Zhao J. The relationship between genetic variants of XRCC1 gene and lung cancer susceptibility in Chinese Han population. Med Oncol. 2014; 31:157. https://doi.org/10.1007/s12032-014-0157-7 [PubMed]
  • 31. Yuan T, Deng S, Chen M, Chen W, Lu W, Huang H, Xia J. Association of DNA repair gene XRCC1 and XPD polymorphisms with genetic susceptibility to gastric cancer in a Chinese population. Cancer Epidemiol. 2011; 35:170–74. https://doi.org/10.1016/j.canep.2010.08.008 [PubMed]
  • 32. Gsur A, Bernhart K, Baierl A, Feik E, Führlinger G, Hofer P, Leeb G, Mach K, Micksche M. No association of XRCC1 polymorphisms Arg194Trp and Arg399Gln with colorectal cancer risk. Cancer Epidemiol. 2011; 35:e38–41. https://doi.org/10.1016/j.canep.2011.03.005 [PubMed]
  • 33. He J, Zou Y, Liu X, Zhu J, Zhang J, Zhang R, Yang T, Xia H. Association of common genetic variants in pre-microRNAs and neuroblastoma susceptibility: a two-center study in Chinese children. Mol Ther Nucleic Acids. 2018; 11:1–8. https://doi.org/10.1016/j.omtn.2018.01.003 [PubMed]
  • 34. Zhuo ZJ, Zhang R, Zhang J, Zhu J, Yang T, Zou Y, He J, Xia H. Associations between lncRNA MEG3 polymorphisms and neuroblastoma risk in Chinese children. Aging (Albany NY). 2018; 10:481–91. https://doi.org/10.18632/aging.101406 [PubMed]
  • 35. Wu Q, Zhuo ZJ, Zeng J, Zhang J, Zhu J, Zou Y, Zhang R, Yang T, Zhu D, He J, Xia H. Association between NEFL gene polymorphisms and neuroblastoma risk in Chinese children: a two-center case-control study. J Cancer. 2018; 9:535–39. https://doi.org/10.7150/jca.22681 [PubMed]
  • 36. Yang X, He J, Chang Y, Luo A, Luo A, Zhang J, Zhang R, Xia H, Xu L. HOTAIR gene polymorphisms contribute to increased neuroblastoma susceptibility in Chinese children. Cancer. 2018; 124:2599–606. https://doi.org/10.1002/cncr.31353 [PubMed]
  • 37. Siciliano MJ, Carrano AV, Thompson LH. Assignment of a human DNA-repair gene associated with sister-chromatid exchange to chromosome 19. Mutat Res. 1986; 174:303–08. https://doi.org/10.1016/0165-7992(86)90051-5 [PubMed]
  • 38. Shen MR, Jones IM, Mohrenweiser H. Nonconservative amino acid substitution variants exist at polymorphic frequency in DNA repair genes in healthy humans. Cancer Res. 1998; 58:604–08. [PubMed]
  • 39. Lunn RM, Langlois RG, Hsieh LL, Thompson CL, Bell DA. XRCC1 polymorphisms: effects on aflatoxin B1-DNA adducts and glycophorin A variant frequency. Cancer Res. 1999; 59:2557–61. [PubMed]
  • 40. Hong YC, Lee KH, Kim WC, Choi SK, Woo ZH, Shin SK, Kim H. Polymorphisms of XRCC1 gene, alcohol consumption and colorectal cancer. Int J Cancer. 2005; 116:428–32. https://doi.org/10.1002/ijc.21019 [PubMed]
  • 41. Divine KK, Gilliland FD, Crowell RE, Stidley CA, Bocklage TJ, Cook DL, Belinsky SA. The XRCC1 399 glutamine allele is a risk factor for adenocarcinoma of the lung. Mutat Res. 2001; 461:273–78. https://doi.org/10.1016/S0921-8777(00)00059-8 [PubMed]
  • 42. Lee SG, Kim B, Choi J, Kim C, Lee I, Song K. Genetic polymorphisms of XRCC1 and risk of gastric cancer. Cancer Lett. 2002; 187:53–60. https://doi.org/10.1016/S0304-3835(02)00381-6 [PubMed]
  • 43. Duell EJ, Millikan RC, Pittman GS, Winkel S, Lunn RM, Tse CK, Eaton A, Mohrenweiser HW, Newman B, Bell DA. Polymorphisms in the DNA repair gene XRCC1 and breast cancer. Cancer Epidemiol Biomarkers Prev. 2001; 10:217–22. [PubMed]
  • 44. Zhuo ZJ, Liu W, Zhang J, Zhu J, Zhang R, Tang J, Yang T, Zou Y, He J, Xia H. Functional polymorphisms at ERCC1/XPF genes confer neuroblastoma risk in Chinese children. EBioMedicine. 2018; 30:113–19. https://doi.org/10.1016/j.ebiom.2018.03.003 [PubMed]
  • 45. Chang J, Tian J, Yang Y, Zhong R, Li J, Zhai K, Ke J, Lou J, Chen W, Zhu B, Shen N, Zhang Y, Gong Y, et al. A rare missense variant in TCF7L2 associates with colorectal cancer risk by interacting with a GWAS-identified regulatory variant in the MYC enhancer. Cancer Res. 2018; 78:5164–72. https://doi.org/10.1158/0008-5472.CAN-18-0910 [PubMed]
  • 46. Li J, Chang J, Tian J, Ke J, Zhu Y, Yang Y, Gong Y, Zou D, Peng X, Yang N, Mei S, Wang X, Cheng L, et al. A rare variant P507L in TPP1 interrupts TPP1-TIN2 interaction, influences telomere length, and confers colorectal cancer risk in Chinese population. Cancer Epidemiol Biomarkers Prev. 2018; 27:1029–35. https://doi.org/10.1158/1055-9965.EPI-18-0099 [PubMed]
  • 47. Zou D, Lou J, Ke J, Mei S, Li J, Gong Y, Yang Y, Zhu Y, Tian J, Chang J, Zhong R, Gong J, Miao X. Integrative expression quantitative trait locus-based analysis of colorectal cancer identified a functional polymorphism regulating SLC22A5 expression. Eur J Cancer. 2018; 93:1–9. https://doi.org/10.1016/j.ejca.2018.01.065 [PubMed]