Open access peer-reviewed chapter

Genetic Single Nucleotide Polymorphisms (GSNPs) in the DNA Repair Genes and Hepatocellular Carcinoma Related to Aflatoxin B1 among Guangxiese Population

Written By

Xue-Ming Wu, Zhi-Feng Xi, Jun Lu, Xing-Zhizi Wang, Tian-Qi Zhang, Xiao-Ying Huang, Jin-Guang Yao, Chao Wang, Zhong-Heng Wei, Chun-Ying Luo, Bing-Chen Huang, Qun-Qing Xu, Wen-Pei Yang, Qiang Xia and Xi-Dai Long

Reviewed: 02 May 2017 Published: 06 September 2017

DOI: 10.5772/intechopen.69530

From the Edited Volume

Genetic Polymorphisms

Edited by Narasimha Reddy Parine

Chapter metrics overview

1,175 Chapter Downloads

View Full Metrics

Abstract

Aflatoxin B1 (AFB1) is an important environmental carcinogen for the development of hepatocellular carcinoma (HCC). HCC is a complex disease likely resulting from genetic single nucleotide polymorphisms (GSNPs) of multiple interacting genes and gene-environment interactions. Recent efforts have been made to analyze the associations between risk of this malignancy and GSNPs in genes involved in the repair of DNA damage induced by AFB1. Here, we reviewed the results of published case-control studies that have examined the effects of common alleles of all susceptible DNA repair genes, including XRCC1, XRCC3, XRCC4, XRCC7, XPC, and XPD, on risk of AFB1-related HCC among Guangxi population. Statistically significant differences in genotype frequencies found in case-control comparisons were rs25487, rs80309960, rs861539, rs7003908, rs28383151, rs3734091, rs13181, and rs2228001 polymorphism. The overall effects of these GNSPs were moderate in terms of relative risk, with ORs ranging from 2 to 10. Furthermore, some evidence of the interaction of GSNPs in DNA repair genes and AFB1 exposure modulate risk of this cancer was also found, although the results require confirmation with larger sample size studies.

Keywords

  • genetic single nucleotide polymorphism
  • hepatocellular carcinoma
  • DNA repair
  • DNA damage
  • aflatoxin B1
  • Guangxiese population

1. Introduction

In China, hepatocellular carcinoma (HCC) is the fourth most common malignant tumors and accounts for about 50% of the world’s HCC cases [1, 2]. This malignancy occurs more often in some hot and humid areas such as Guangxi area, mainly because of high aflatoxin B1(AFB1) exposure [3, 4]. However, accumulating epidemiological evidence has shown that although many people are exposed to this risk factor, only a relatively small proportion of people with chronic AFB1 exposure develop HCC [3, 4]. This suggests that an individual susceptibility related to genetic factors including genetic single nucleotide polymorphisms (GSNPs) in DNA repair genes might be correlated with HCC tumorigenesis [35]. In past several decade years, evidence has been accumulated to support the hypothesis that GSNPs in DNA repair genes may be of importance in determining individual susceptibility to AFB1-related HCC [68]. Here, we review recent efforts in identifying genetic single nucleotide polymorphisms (SNPs) in DNA repair genes, which may have impact on AFB1-related HCC risk among Guangxiese population.

Advertisement

2. Epidemiology of AFB1-related HCC among Guangxi areas

In Guangxi, a known high AFB1 exposure area, HCC is the most common occurring cancer [5]. Epidemiologically, this type tumor is characterized as follows: (a) the incidence rate displaying a noticeable mountain shape, gradually increasing with age increasing before shape top and gradually decreasing with age increasing after shape top and (b) more than three times risk of HCC among males than among females, especially in the high AFB1-exposure areas (including Baise and Nanning) [5]. During 1990–1992, the incidence rates of this tumor in Guangxi area increased markedly compared between the late 1970s and the early 1980s (25.22 per 100,000 vs. 15.85 per 100,000), especially male Guangxiese population (Figure 1A) [9]. This might result from the increasing infective rate of hepatitis viruses and advance of diagnostic ability [10]. The following epidemiological investigations showed that the incidence rate of HCC among Guangxiese population significantly markedly decrease during 2004 and 2005 (mainly because of the control of hepatitis virus infection) (Figure 1) [9]. However, the incident rates of this tumor in high AFB1-exposure areas have little change. According to epidemiological investigation from AFB1-exposures areas, during May 2007–April 2008, incidence rates were 103.1/100,000 and 117.8/100,000 for Fusui in Nanning and Xiangzhou in Baise (two main high AFB1 exposure areas), respectively [11]. This is similar to the results before 10 years [11].

Figure 1.

The incidence and mortality rates of hepatocellular carcinoma (HCC) in Guangxi during 1971 and 2005. Total incidence rate (A) and mortality rates (B), regardless of male or female population, were significantly increasing from during 1971 and 1973 to during 1990 and 1992. However, they would decrease during 2004 and 2005.

Because of the very poor prognosis, HCC is the first most common cause of death from cancer in Guangxi [9]. In the past 40 years, the total mortality rate of HCC gradually increased and followed by gradually decreased (Figure 1B), regardless of male or female population. Furthermore, this trend was not only associated with age but also more noticeable in male population than female population (Figure 2), possibly because male individuals featured more AFB1 exposure [4]. Supporting above-mentioned hypothesis, molecular epidemiological studies from high AFB1-exposure areas of Guangxi have exhibited that these individuals featuring more AFB1 exposure would face decreasing 5-year survival rate and increasing death risk [1214].

Figure 2.

The gender and age distributions of hepatocellular carcinoma (HCC) mortality among Guangxiese population. (A) The gender and age distributions of HCC mortality between 1990 and 1992. (B) The gender and age distributions of HCC mortality between 2004 and 2005.

Advertisement

3. DNA damage induced byAFB1 and DNA repair

Several previous reviews have significantly summarized the toxicology of AFB1 [35, 15]. Generally, AFB1, an important mycotoxin and a category I known human carcinogen produced by Aspergillus parasiticus and Aspergillus flavus [4, 5], has been found as toxic contaminants of human food such as ground nuts and core in tropical areas as a result of fungal contamination during growth and after harvest which under hot and humid conditions [15]. Once this toxigenic agent is taken into body, it is metabolized by phase I detoxification enzymes to its reactive form, also called AFB1-8,9-epoxide (AFBO). This reactive product can also covalently bind to DNA and induce DNA damage, which might ultimately result to the development of HCC [16, 17].

AFB1-induced DNA damage types consist of DNA adducts, oxidative damage, and gene mutations. For AFB1-DNA adducts, AFB1-N7-Gua adduct, the most common adduct type, is primarily identified and confirmed in vivo research studies [1722], whereas the formamidopyridine AFB1 adduct (AFB1-FAPy) is a ring-opened DNA adduct [23, 24]. Because of the accumulations of AFB1-FAPy, it is characterized by a time-dependent and non-enzyme way and exhibits apparent persistence in DNA; it may be of biological basis of genes mutation. Furthermore, AFB1-N7-Gua and AFB1-FAPy can induce error-prone DNA repair and result in the damage of DNA strands, including double-strand breaks (DSBs), single-strand breaks (SSBs), base pair substitution, frame shift mutations, and oxidation DNA damage such as 8-oxodeoxyguanosine (8-oxyG) [2528]. For genes mutations induced by AFB1 exposure, G:C > T:A mutation in codon 249 of TP53 gene has been frequently reported [2936]. This mutation is tested in more than 40% of HCC from high-AFB1-exposure areas, but in either very rare or absent for those from low or null AFB1 exposure areas [3639]. Therefore, G:C > T:A mutation in codon 249 of TP53 gene is regarded as the hotspot mutation caused by AFB1 and the molecular symbol of AFB1-related HCC [4042].

DNA damage by AFB1 exposure, if not repaired, may cause genomic instability and ultimately result in cellular malignant transformation and tumor formation [20]. In human cells, several evolved surveillance mechanisms, including base excision repair (BER), nucleotide excision repair (NER), single-strand break repair (SSBR), and double-strand break repair (DSBR), can monitor the integrity of genome and repair and minimize the consequences of detrimental genome damage [3, 40]. DNA repair genes play an important role during aforementioned repair pathways of DNA damage; therefore, GSNPs in the DNA repair genes may be associated with risk of developing AFB1-related HCC [4].

Advertisement

4. Xeroderma pigmentosum C (XPC) GSNPs and AFB1-HCC among Guangxiese population

Xeroderma pigmentosum C (XPC) gene locates on chromosome 3p25 and spans 33kb. Its encoding protein consists of 940 amino acids and acts as a DNA damage recognition molecule during the global genome NER pathway [43, 44]. XPC can bind tightly with RAD23 homolog B (RAD23B, also called HR23B) to form a stable XPC/RAD23B complex. This complex can recognize DNA adducts formed by exogenous carcinogens such as AFB1 and binds to the DNA damage sites [44]. Thus, XPC may play a role in the pathogenesis of HCC-related AFB1. The pathological and cellular researches have shown that the abnormal expression of this gene is correlated with hepatocarcinogenesis [45]. Some studies have shown that GSNPs of XPC (rs2228001 (also called codon Lys939Gln polymorphism) can decrease DNA repair capacity related to AFB1-induced DNA damage [4649]. In the past several decade years, a total of four studies from Guangxi area reported XPC rs2228001 polymorphism was involved in AFB1 detoxification (Table 1) [12, 4951]. The first study conducted by Wu et al. [51] is from Baise area (a famous high AFB1 exposure area with high incidence rate of HCC). In this study, researchers investigated the association between XPC rs2228001 polymorphism and HCC risk via an hospital-based case-control study (including 98 HCC patients and 157 age-, sex-, race-, and hepatitis viruses-matching controls) method and found XPC rs2228001 polymorphism increased HCC risk of female individuals (odds ratios [ORs] were 5.44 with 95% CI 1.38–21.50), but not tumor risk of male population. Noticeably, they found a significant interaction of XPC variables and AFB1 exposure levels [51].

No.Ref.YearAFB1 exposureaMethodsMatching factorCasesControlsRisk valueb
(n)(n)(OR)
1Wu et al. [51]2010HighCase-controlAge, sex, HBV, HCV, race981571.46–2.08 (P < 0.05)
2Long et al. [12]2010HighCase-controlAge, sex, HBV, HCV, race115614021.25–1.81 (P < 0.001)
3Li et al. [50]2010HighCase-controlNo5005071.20–1.81 (P > 0.05)
4Yao et al. [49]2014HighCase-controlAge, sex, HBV, HCV, race148619961.57–2.19 (P < 0.001)

Table 1.

Characteristics of studies about GSNP at codon 939 of XPC and AFB1-related HCC risk.

a Defined by means of Henry et al. (Science, 1999).


b AFB1-related HCC risk will increase if OR > 1 and corresponding P-value < 0.05; will decrease if OR < 1 and corresponding P-value < 0.05; and will not change if OR is about 1 and/or corresponding P-value > 0.05.


Results from the matching-design studies with large-size samples show that the mutant genotypes of XPC rs2228001 polymorphism increased about two times risk of HCC [12, 49]. The following quantitative analysis exhibited that AFB1 exposure interacted with risk genotypes of XPC rs2228001 polymorphism on HCC risk (22.33 > 1.88 × 8.69 for the interaction of AFB1-exposure years and XPC risk genotypes and 18.38 > 1.11 × 4.62 for the interaction of AFB1-exposure levels and XPC risk genotypes) [12, 49]. Furthermore, mutant alleles were correlated with the decrease of XPC expression levels in cancerous tissues and with the overall survival of HCC patients [12]. However, another study also from Nanning area (another high AFB1 exposure area in Guangxi) exhibited that this polymorphism does not change HCC risk [50], possibly because it bases on nonmatching design and some confounders might affect their results. Interestingly, a significant risk value for HCC among female population was observed in the stratified analyses (OR = 2.17, 95% CI = 1.01–4.64), similar with findings from Wu et al. [51]. Given these different effects, we accomplished meta-analysis and found XPC rs2228001 polymorphism increased AFB1-related risk (combined OR = 1.89, P < 0.01). Therefore, these results demonstrate that GSNP at codon 939 of XPC gene is not only a genetic determinant in the development of HCC caused by AFB1 in Guangxiese population but also an independent prognostic factor influencing the survival of HCC, like AFB1 exposure.

Advertisement

5. Xeroderma pigmentosum D (XPD) GSNPs and AFB1-HCC among Guangxiese population

Xeroderma pigmentosum gene, contains 22 introns and 23 exons, locates on 19q13.3 and spans about 20 kb [3, 52]. Its encoded product is one of the TFIIH complexes (a DNA-dependent ATPase/helicase complex), it has classically been linked to the damage verification and the opening of the DNA helix step of NER pathway [53]. To date, molecular epidemiological studies on two GSNPs in this gene, namely, rs369191500 polymorphism (codon 312, Asp to Asn) and rs13181 polymorphism (codon 751, Lys to Gln), have been conducted by different laboratories [54]. Genotype-phenotype analyses have shown that these two GSNPs are associated with low DNA repair ability, which might modify DNA-adduct levels, p53 gene mutation, and risk of cancer [55, 56]. For genetic polymorphisms among Guangxiese population, a total of four epidemiological studies have been conducted via case-control design (Table 2). In 2009, Zeng et al. [57] investigated the association between XPD GSNPs and HCC and observed about 2.60-time risk of HCC for these with mutant genotypes of XPD codon 751. After that, results from Long et al. [58] and Yao et al. [49] have exhibited that the variants of XPD rs13181 polymorphism (including rs13181-LG and -GG) was significantly different between controls and HCC cases (26.3 vs. 35.9% for rs13181-LG and 8.6 vs. 20.1% for rs13181-GG, respectively, P < 0.001). Mutant alleles increased the risk of developing the cancer with a risk value of 1.75 (1.30–2.37) for Lys/Gln and 2.47 (1.62–3.76) for Gln/Gln, respectively [58]. Furthermore, the interactive effects of mutant genotypes of rs13181 polymorphism in XPD gene and AFB1 exposure on HCC risk were also found (Pinteraction < 0.05) [49]. Recently, Hu et al. [59] have reported higher frequency of XPD codon 751 Gln alleles among HCC families than among non-HCC families. This suggests that genetic susceptibility resulting from XPD GSNPs might have a potential effect on AFB1-related HCC risk among Guangxiese population. Meta-analysis further improved the above-mentioned hypothesis [56]. However, the studies from molecular epidemiological studies exhibit that another GSNP of XPD is not significantly correlate with the risk of HCC related to AFB1 [49, 58].

No.Ref.YearAFB1 exposureaMethodsMatching factorCasesControlsRisk valueb
(n)(n)(OR)
1Zeng et al. [57]2009HighCase-controlNo300312About 2.60 (P < 0.001)
2Long et al. [58]2009HighCase-controlAge, sex, HBV, HCV, race6187121.75–2.47 (P < 0.001)
3Yao et al. [49]2014HighCase-controlAge, sex, HBV, HCV, race148619962.19–4.27 (P < 0.001)
4Hu et al. [59]2016HighCase-controlNo1781782.33–3.38 (P < 0.05)

Table 2.

Characteristics of studies about genetic polymorphism at codon 751 of XPD and AFB1-related HCC risk.

a Defined by means of Henry et al. (Science, 1999).


b AFB1-related HCC risk will increase if OR > 1 and corresponding P-value < 0.05; will decrease if OR < 1 and corresponding P-value < 0.05; and will not change if OR is about 1 and/or corresponding P-value > 0.05.


Advertisement

6. The human 8-oxoguanine DNA glycosylase (hOGG1) GSNPs and AFB1-HCC among Guangxiese population

As described previously [60], DNA glycosylases play a central role in the BER pathway because they can recognize and catalyze the removal of damaged bases. Among having been reported GSNPs in DNA glycosylase-encoded genes, only hOGG1 correlates with DNA repair capacity [5, 61]. This gene consists of 7 exons and 6 introns and spans 17 kb on chromosome 3p26.2. It encodes a 546-amino acid protein, a specific DNA glycosylase that catalyzes the release of 8-oxyG and the cleavage of DNA at the AP site (Figure 3) [61, 62]. The presence of several polymorphisms within hOGG1 locus is identified and only rs1052133 polymorphism (at codon 326, Ser → Cys) is suggested to modify DNA repair capacity [6365]. Therefore, low capacity of 8-oxyG repair resulting from hOGG1 326Cys polymorphism might contribute to the persistence of 8-oxyG in genomic DNA in vivo, which, in turn, could be associated with increased cancer risk [66]. Several reports imply this polymorphism may be associated with HCC related to AFB1 exposure [6770]. Through a hospital-based case-control study (including 500 cases with HCC and 507 healthy controls) conducted in Nanning area from Guangxi, Ji et al. [67] investigated the relationship between hOGG1 rs1052133 polymorphism and HCC risk and found significantly different distribution of three genotypes (Ser/Ser, Ser/Cys, and Cys/Cys) between cases and controls (71.40, 8.00, and 20.60% for patients with HCC, 84.22, 10.06, and 5.72% for controls). Regression logistic analysis shows that mutant alleles of this polymorphism increase AFB1-related HCC risk (OR = 2.14, 95% CI = 1.57–2.91). Molecular epidemiological studies from other researchers have also proved higher frequencies of Cys alleles in HCC cases than controls and this change increased HCC risk among Guangxiese population featuring AFB1-exposure history [69, 70]. Given that hOGG1 expression is significantly related to HCC carcinogenesis [71, 72], Peng et al. [68] explored the effects of hOGG1 rs1052133 polymorphism on 8-oxyG levels and hOGG1 expression in Guangxiese population with AFB1 exposure. They observed that Cys alleles downregulated hOGG1 expression and increased 8-oxyG levels. These findings suggested the pathogenic role of hOGG1 Cys326Ser polymorphism in the carcinogenesis of AFB1-related HCC.

Figure 3.

The base excision repair for oxidative DNA damage 8-oxyG in mammalian cells. In this repair pathway, human 8-oxoguanine DNA glycosylase (hOGG1) can catalyze the release of 8-oxyG (Red) and the cleavage of DNA at the AP site.

Advertisement

7. The XRCC1 GSNPs and AFB1-HCC among Guangxiese population

X-ray repair cross-complementing group 1 (XRCC1), also called RCC, spans about 32 kb on chromosome 19q13.2 and contains 17 exons and 16 introns. The encoding protein of XRCC1 acts as a scaffold protein in SSBR and BER pathways via the interaction with Pol β, DNA ligase III, and PARP [73]. According to data from SNP database, more than 50 GSNPs in the coding region of XRCC1 gene, which lead to amino acid substitution, have been identified. Of GSNPs, rs25487 polymorphism (at codon 399, Arg → Gln) is of special concern, because this GSNP resides in functionally significant regions and may decrease DNA repair activity and increase the risk of gene mutation cancers [3].

In Guangxi area, a total of five molecular epidemiological studies were found in PubMed Database, Web of Science Database, Wangfang Database, Google Database, and Weipu database (Table 3). In 2005, Long et al. [74] investigated the effects of XRCC1 rs25487 polymorphism on AFB1-related HCC risk. In their study, they included 140 patients with pathologically diagnosed HCC and 536 age-, sex-, race-, and hepatitis virus-matching controls and found significantly different distribution of three genotypes of XRCC1 rs25487 polymorphism (Arg/Arg, Arg/Gln, and Gln/Gln) between cases and controls (51.43, 45.00, and 3.57% for patients with HCC, 6.54, 29.66, and 2.88% for controls). Regression logistic analysis shows that mutant alleles of this polymorphism increase AFB1-related HCC risk (OR = 2.18, 95% CI = 1.27–3.74). They also observed more noticeable risk role for HCC under the conditions of individuals with both high AFB1 exposure and mutant genotypes of XRCC1 rs25487 polymorphism (risk value 1.84–10.87). The several following molecular epidemiological studies furthermore prove these results [49, 75, 77]. However, associations between XRCC1 rs25487 polymorphism and individual susceptibility to HCC have been reported in a case-control study with the insignificant results [76]. This may be because of nonmatching design and the effects of confounders such as age, gender, race, and hepatitis viruses. Supporting aforementioned conclusions, meta-analysis from data in high AFB1 exposure areas also exhibited that these subjects with Gln alleles of XRCC1 rs25487 polymorphism had increasing HCC risk (OR > 1, P < 0.01) [78]. This suggests that XRCC1 rs25487 polymorphism should be a risk biomarker of Guangxiese HCC related to AFB1 exposure.

No.Ref.YearAFB1 exposureaMethodsMatching factorCases (n)Controls (n)Risk valueb (OR)
1Long et al. [74]2005HighCase-controlAge, sex, HBV, HCV, race1405362.18 (P = 0.0001)
2Long et al. [75]2006HighCase-controlAge, sex, HBV, HCV, race2576492.47 (P = 0.0001)
3Zeng et al. [76]2010HighCase-controlNo500507about 1 (P > 0.05)
4He et al. [77]2012HighCase-controlAge, sex1191192.50 (P < 0.05)
5Yao et al. [49]2014HighCase-controlAge, sex, HBV, HCV, race148619962.19–4.27 (P < 0.001)

Table 3.

Characteristics of studies about genetic polymorphism at codon 399 of XRCC1 and AFB1-related HCC risk.

Notes: a Defined by means of Henry et al. (Science, 1999, 286: 2453-2454).


b AFB1-related HCC risk will increase if OR > 1 and corresponding P-value < 0.05; will decrease if OR < 1 and corresponding P-value < 0.05; and will not change if OR is about 1 and/or corresponding P-value > 0.05.


Advertisement

8. The XRCC3 GSNPs and AFB1-HCC among Guangxiese population

X-ray repair cross-complementing group 3 (XRCC3) is an important paralogs of the strand-exchange protein RAD51 and is primarily found and cloned by functional complementation in the Chinese hamster cell line irs1SF. This gene spans about 17.8 kb on chromosome 14q32.33 and contains 10 exons. Functionally, XRCC3 can bind directly with DNA breaks and facilitate the formation of the RAD51 nucleoprotein filament and protein-protein interaction of RAD51 family members in the SSBR pathway [79]. In XRCC3-defective irs1SF hamster cells, spontaneous chromosomal aberrations, high sensitivity to cross-linking agents, mild sensitivity to gamma rays, and significantly attenuated Rad51 focus formation after gamma ray exposure were observed, which suggests that XRCC3 is crucial for the SSBR pathway [80].

Recent molecular epidemiological studies have shown that the function of XRCC3 is modified by its GSNPs. Of the known GSNPs, only rs2228001 polymorphism (at codon 241, Thr → Met) is of special concern, because this GSNP results the change Thr to Met at codon 241 [8183]. Up to now, three reports from high AFB1-exposure areas of Guangxi supported the above-mentioned conclusions [49, 84, 85]. In the first frequent case-control study in Guangxi population [85], higher mutant frequencies at codon 241 of XRCC3 are observed among cases with HCC than controls (33.01 vs. 61.48%, P < 0.001). The results from logistic regression analysis further prove that this mutate increases about 2- to 10-time risk of HCC. The following two relatively larger sample size molecular epidemiological studies (study 1 conducted by Long et al. [84] consisting of 491 patients with HCC and 862 healthy controls; study 2 from Yao et al. [49] including 1486 cases and 1996 controls) were accomplished in high AFB1 exposure. Similar to data from the first study, individuals having the mutant genotypes of XRCC3 rs2228001 polymorphism would feature higher risk of developing HCC. Interactive analysis of risk genotypes and AFB1 exposure further demonstrated that this allele multiplicatively interacted with AFB1 exposure in the process of hepatotumorigenesis [49]. Taken together, these results suggest that the GSNPs in XRCC3 gene, such as rs2228001 polymorphism, might a genetic determinant in the HCC carcinogenesis caused by AFB1 exposure among Guangxiese population.

Advertisement

9. The XRCC7 GSNPs and AFB1-HCC among Guangxiese population

X-ray repair cross-complementing 7 (XRCC7), also called protein kinase DNA-activated catalytic polypeptide (PRKDC), spans about 197 kb on 8q11.21 and contains 85 exons. Protein encoded by his gene is a member of the PI3/PI4-kinase family and acts as the catalytic subunit of the DNA-dependent protein kinase (DNA-PK). Functionally, it interacts with the Ku70/Ku80 heterodimer protein in the nonhomologous end joining (NHEJ) repair and recombination [8688]. More than 200 of GSNPs have been reported in the XRCC7 gene, some of which are reported to affect tumorigenesis of malignant tumors [8993]. In Guangxi area, several reports imply XRCC7 rs7003908 polymorphism (T to G) may be associated with HCC related to AFB1 exposure [49, 94]. Through a hospital-based case-control study (including 348 cases with HCC and 597 age-, gender-, race-, and hepatitis viruses-matched healthy controls) conducted in high AFB1 exposure area from Guangxi, Long et al. [94] investigated the association between XRCC7 rs7003908 polymorphism and HCC risk and found significantly different distribution of three genotypes (rs7003908-TT, -TG, and -GG) between cases and controls (25.9, 44.5, and 29.6% for patients with HCC, 58.1, 30.0, and 11.9% for controls). Regression logistic analysis shows that mutant alleles of this polymorphism increase AFB1-related HCC risk, with risk value of 3.45 (2.40–4.94) for rs7003908-TG and 5.04 (3.28–7.76) for rs7003908-GG, respectively. Furthermore, this GSNP was also related to higher the amounts of DNA adducts induced by AFB1 (r = 0.142) [94]. Interestingly, it also multiplicatively interacts with AFB1-exposure variable (interactive value: OR = 1.74; 95% CI = 1.66–1.82; P = 2.21 × 10−133) [49]. Taken together, these data exhibited that the XRCC7 rs7003908 polymorphism might modify AFB1-related HCC risk via decreasing NHEJ capacity. However, more studies are inquired to support this conclusion.

Advertisement

10. The XRCC4 GSNPs and AFB1-HCC among Guangxiese population

X-ray repair cross-complementing 4 (XRCC4, also known as SSMED) is an important NHEJ pathway gene locating on 5q14.2 and spanning about 293 kb. This gene is a generally expressed protein of 334 amino acids residues and functions together with DNA ligase IV and DNA-PK in the repair of DNA double-strand breaks. XRCC4 plays an important role in both nonhomologous end joining and the completion of V(D)J recombination [95, 96]. Increasing evidence has shown that mutations in XRCC4 can cause short stature, microcephaly, and endocrine dysfunction (SSMED) resulting from more-deficient NHEJ capacity [97, 98]. This suggests that the low repair capacity of this gene from genetic changes might be associated with cancer risk [99, 100]. Up to now, only two GSPs in the XRCC4 gene, namely, rs28383151 (at codon 56, Ala to Thr) and rs3734091 (at codon 247, Ala to Ser), have reported to be correlated with risk of AFB1-related HCC among Guangxiese population [13, 14, 49]. In these studies, Long et al. [13, 14] have examined the association between genetic polymorphisms (Ala56Thr and Ala247Ser) in XRCC4 and risk of HCC in a large case-control study in at-risk Guangxiese population. Genotypes were determined in 1499 patients with HCC and 2045 individually matched controls. We observed that individuals with Thr/Thr genotype at Arg194Trp site and Ser/Ser genotype at Ala247Ser site of XRCC4 had about twofold and threefold increased risk for the cancer compared with wild types, respectively. Furthermore, when risk genotypes of XRCC4 and AFB1-exposure were combined, this risk role was more noticeable, with the adjusted OR being11.26 (95% CI, 8.36–15.18) for rs28383151 polymorphism and 14.43 (95% CI, 7.98–26.09) for rs3734091 polymorphism, respectively. Interactive analysis further exhibited this multiplicative interaction (interactive OR = 1.64, 95% CI = 1.55–1.73, 6.41 × 10−73) [49]. Functional exploration showed that these two polymorphisms increased AFB1-DNA adducts levels and TP53M risk, and AFB1-related HCC prognosis, suggesting they should be important modified factors of AFB1 toxicological effects [5, 13, 14]. Together, these results imply that XRCC4 rs28383151 and rs3734091 polymorphisms might play a role in the carcinogenesis of the liver under the conditions of AFB1 exposure.

11. Future directions

In the past decades, great progress has been made in understanding the molecular mechanisms of the GSNPs affecting risk of AFB1-related HCC. However, we are still far from a comprehensive view of this effect. The molecular mechanism about GSNPs in the DNA repair genes modifying the risk of HCC caused by AFB1 remains largely unknown. Although several reports have displayed that GSNPs may progress tumorigenesis of AFB1-related HCC via downregulating expression of DNA repair genes, breaking the structure of DNA repair proteins, or decreasing the function of DNA repair genes, more direct evidence is still missing. Disclosing the roles of different phenotypes of DNA repair genes will greatly benefit our understanding of pathological mechanisms of GSNPs affecting AFB1-related HCC risk, and will shed important light on the prevention values for individuals with risk types.

12. Conclusion

Like most other human cancers, HCC is a complex disease attributed to environment variation and genetic susceptive factors. In high incidence areas of HCC in Guangxi, AFB1 is an important environment variation. In the process of AFB1 hepatocarcinogenesis, DNA damage induced by AFB1 exposure plays a central role because of their genotoxicity and interactions with genetic susceptive factors. Numerous studies reviewed in this chapter have demonstrated that the hereditary variations in DNA repair genes (including XRCC1, XRCC3, XRCC4, XRCC7, hOGG1, XPC, and XPD) are related to the susceptibility to AFB1-related HCC among Chinese population. These molecular epidemiological studies have significantly contributed to our knowledge of the importance of genetic polymorphisms in DNA repair genes in the etiology of HCC related to AFB1 exposure. It would be expected that genetic susceptibility factors involved in DNA repair genes for HCC could serve as useful biomarkers for identifying at-risk individuals and, therefore, targeting prevention of this malignant tumor.

However, there are several issues to be noted. Because of conflicting data existing in the same ethnic population in view of between some genotypes of DNA repair genes and the risk of HCC related to AFB1, the conclusions should first be drawn carefully. For example, whether the XRCC1 Arg399Gln polymorphism is a risk factor or not remains controversial and further studies are needed. Second, because of the difference in population frequencies corresponding to genetic polymorphisms that depend on ethnicity, caution should be taken, particularly in extrapolating these data to other ethnic populations, especially from other high AFB1-exposure areas. Third, when risk of a specific polymorphism is considered, AFB1 exposure should be stressed because AFB1 exposure may differ from areas to areas and from individuals to individuals. Last, in view of that fact the development of AFB1-related HCC is “polygenic,” a panel of susceptive biomarkers are warranted to define individuals at high risk for this cancer.

Acknowledgments

This study was supported in part by the National Natural Science Foundation of China (Nos. 81572353, 81372639, 81472243, 81660495, and 81460423), the Innovation Program of Guangxi Municipal Education Department (Nos. 201204LX674 and 201204LX324), the Natural Science Foundation of Guangxi (Nos. 2016GXNSFDA380003, 2015GXNSFAA139223, 2013GXNSFAA019251, 2014GXNSFDA118021, and 2014GXNSFAA118144), Innovation Program of Guangxi Health Department (No. Z2013781), Research Program of Guangxi Clinic Research Center of Hepatobiliary Diseases (No. 2016-11), and Open Research Program from Molecular Immunity Study Room Involving in Acute & Severe Diseases in Guangxi Colleges and Universities (Nos. kfkt20160062 and kfkt20160063).

Abbreviations

8-oxyG8-oxodeoxyguanosine
AFB1aflatoxin B1
AFBOAFB1-8,9-epoxide
AFB1-FAPy8,9-dihydro-8-(2,6-diamino-4-oxo-3,4-dihydropyrimid-5-yl formamide)-9-hydroxy-AFB1
AFB1-N7-Gua(8,9-dihydro-8-N 7-guanyl-9-hydroxy-AFB1)
BERbase excision repair
DSBRdouble-strand break repair
DSBsdouble-strand breaks
GSNPgenetic single nucleotide polymorphism
HCChepatocellular carcinoma
HRhazard ratio
hOGG1human 8-oxoguanine DNA glycosylase
NERnucleotide excision repair
NHEJnonhomologous end joining
ORodds ratio
SSBssingle-strand breaks
SSBRsingle-strand break repair
SSMEDshort stature, microcephaly, and endocrine dysfunction
TP53Mthe hotspot mutation at codon 249 of TP53 gene;
XPCxeroderma pigmentosum C
XPDxeroderma pigmentosum D
XRCC1X-ray repair cross-complementing group 1
XRCC3X-ray repair cross-complementing group 3
XRCC4X-ray repair cross-complementing group 4
XRCC7X-ray repair cross-complementing group 7

References

  1. 1. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA A Cancer Journal for Clinicians. 2015;65:87-108. DOI: 10.3322/caac.21262
  2. 2. Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ, He J. Cancer statistics in China, 2015. CA A Cancer Journal for Clinicians. 2016;66:115-132. DOI: 10.3322/caac.21338
  3. 3. Xia Q, Huang XY, Xue F, Zhang JJ, Zhai B, Kong DC, Wang C, Huang ZQ, Long XD. Genetic polymorphisms of DNA repair genes and DNA repair capacity related to aflatoxin b1 (AFB1)-induced DNA damages. In: Chen C, editor. New Research Directions in DNA Repair. 1st ed. Rijeka: InTech; 2013. pp. 377-412. DOI: 10.5772/53967
  4. 4. Long XD, Yao JG, Zeng Z, Huang CH, Huang ZS, Huang YZ, Ban FZ, Huang XY, Yao LM, Fan LD, Fu GH. DNA repair capacity-related to genetic polymorphisms of DNA repair genes and aflatoxin B1-related hepatocellular carcinoma among Chinese population. In: Kruman I, editor. DNA Repair. Rijeka: InTech; 2011. pp. 505-524. DOI: 10.5772/20792
  5. 5. Long XD, Yao JD, Yang Q, Huang CH, Liao P, Nong LG, Tang YJ, Huang XY, Wang C, Wu XM, Huang BC, Ban FZ, Zeng LX, Ma Y, Zhai B, Zhang JQ, Xue F, Lu CX, Xia Q. Polymorphisms of DNA repair genes and toxicological effects of aflatoxin B1 exposure In: Faulkner AG, editor. Aflatoxins: Food Sources, Occurrence and Toxicological Effects. 1st ed. New York: Nova Science Publishers; 2014. pp. 107-124. DOI: 978-1-63117-298-4
  6. 6. Clifford RJ, Zhang J, Meerzaman DM, Lyu MS, Hu Y, Cultraro CM, Finney RP, Kelley JM, Efroni S, Greenblum SI, Nguyen CV, Rowe WL, Sharma S, Wu G, Yan C, Zhang H, Chung YH, Kim JA, Park NH, Song IH, Buetow KH. Genetic variations at loci involved in the immune response are risk factors for hepatocellular carcinoma. Hepatology. 2010;52:2034-2043. DOI: 10.1002/hep.23943
  7. 7. Dominguez-Malagon H, Gaytan-Graham S. Hepatocellular carcinoma: An update. Ultrastructural Pathology. 2001;25:497-516. DOI: 10.1080/25.497516
  8. 8. Long XD, Tang YH, Qu DY, Ma Y. The toxicity and role of aflatoxin B1 and DNA repair (corresponding DNA repair enzymes). Youjiang Medical College for Nationalities Xue Bao. 2006;28:278-280. DOI: 10.3969/j.issn.1001-5817.2006.02.092
  9. 9. Zhang CY, Huang TR, Yu JH, Zhang ZQ, Li JL, Deng W, Ye SY, Zhou DN, He ZF. Epidemiological analysis of primary liver cancer in the early 21st century in Guangxi province of China. Chinese Journal of Cancer. 2010;29:545-550. DOI: 10.3969/j.issn.0253-4304.2000.04.003
  10. 10. Huang ZB, Wu ZB, Xu GC. A study on serum epidemiology of several hepatitis viruses in Guangxi areas with high incident areas of liver cancer. Guangxi Medical Unveristy Xue Bao. 2000;17:79. DOI: 10.3969/j.issn.1005-930X.2000.01.037
  11. 11. Ma AM, Luo YX, Wan JL, Long XD, Huang YZ. An investigative analysis of incidence, prevention, and treatment of hepatocellular carcinoma in Xiangzhou, Baise, Guangxi Autonomous Region, China. Youjiang Minzu Yixueyuan Bao. 2010;32:165-166. DOI: 10.3969/j.issn.1001-5817.2010.02.024
  12. 12. Long XD, Ma Y, Zhou YF, Ma AM, Fu GH. Polymorphism in xeroderma pigmentosum complementation group C codon 939 and aflatoxin B1-related hepatocellular carcinoma in the Guangxi population. Hepatology. 2010;52:1301-1309. DOI: 10.1002/hep.23807
  13. 13. Long XD, Zhao D, Wang C, Huang XY, Yao JG, Ma Y, Wei ZH, Liu M, Zeng LX, Mo XQ, Zhang JJ, Xue F, Zhai B, Xia Q. Genetic polymorphisms in DNA repair genes XRCC4 and XRCC5 and aflatoxin B1-related hepatocellular carcinoma. Epidemiology. 2013;24:671-681. DOI: 10.1097/EDE.0b013e31829d2744
  14. 14. Long XD, Yao JG, Zeng Z, Ma Y, Huang XY, Wei ZH, Liu M, Zhang JJ, Xue F, Zhai B, Xia Q. Polymorphisms in the coding region of X-ray repair complementing group 4 and aflatoxin B1-related hepatocellular carcinoma. Hepatology. 2013;58:171-181. DOI: 10.1002/hep.26311
  15. 15. Kensler TW, Roebuck BD, Wogan GN, Groopman JD. Aflatoxin: A 50-year odyssey of mechanistic and translational toxicology. Toxicological Sciences: An Official Journal of the Society of Toxicology. 2011;120(Suppl 1):S28–S48. DOI: 10.1093/toxsci/kfq283
  16. 16. Guengerich FP, Johnson WW, Shimada T, Ueng YF, Yamazaki H, Langouet S. Activation and detoxication of aflatoxin B1. Mutation Research. 1998;402:121-128. DOI: S0027-5107(97)00289-3
  17. 17. Ben Rejeb I, Arduini F, Arvinte A, Amine A, Gargouri M, Micheli L, Bala C, Moscone D, Palleschi G. Development of a bio-electrochemical assay for AFB1 detection in olive oil. Biosensors and Bioelectronics. 2009;24:1962-1968. DOI: 10.1016/j.bios.2008.10.002
  18. 18. Giri I, Stone MP. Wobble DC. dA pairing 5' to the cationic guanine N7 8,9-dihydro-8-(N7-guanyl)-9-hydroxyaflatoxin B1 adduct: Implications for nontargeted AFB1 mutagenesis. Biochemistry. 2003;42:7023-7034. DOI: 10.1021/bi020688n
  19. 19. Habib SL, Said B, Awad AT, Mostafa MH, Shank RC. Novel adenine adducts, N7-guanine-AFB1 adducts, and p53 mutations in patients with schistosomiasis and aflatoxin exposure. Cancer Detection and Prevention. 2006;30:491-498. DOI: 10.1016/j.cdp.2006.10.006
  20. 20. Wang JS, Groopman JD. DNA damage by mycotoxins. Mutation Research. 1999;424:167-181. DOI: 01.2013/0027-5107.10064859
  21. 21. Essigmann JM, Croy RG, Nadzan AM, Busby WF, Jr, Reinhold VN, Buchi G, Wogan GN. Structural identification of the major DNA adduct formed by aflatoxin B1 in vitro. Proceedings of the National Academy of Sciences of the United States of America. 1977;74:1870-1874. DOI: 10.2013/0027-8424.266709
  22. 22. Croy RG, Essigmann JM, Reinhold VN, Wogan GN. Identification of the principal aflatoxin B1-DNA adduct formed in vivo in rat liver. Proceedings of the National Academy of Sciences of the United States of America. 1978;75:1745-1749. DOI: 10.2013/0027-8427. 273905
  23. 23. Croy RG, Wogan GN. Temporal patterns of covalent DNA adducts in rat liver after single and multiple doses of aflatoxin B1. Cancer Research. 1981;41:197-203. DOI: 10.2013/0008-5472.7448760
  24. 24. Groopman JD, Croy RG, Wogan GN. In vitro reactions of aflatoxin B1-adducted DNA. Proceedings of the National Academy of Sciences of the United States of America. 1981;78:5445-5449. DOI: 10.2013/0027-8424.6795633
  25. 25. Chen JD, Liu CJ, Lee PH, Chen PJ, Lai MY, Kao JH, Chen DS. Hepatitis B genotypes correlate with tumor recurrence after curative resection of hepatocellular carcinoma. Clinical Gastroenterology and Hepatology. 2004;2:64-71. DOI: S1542356503002933
  26. 26. Gradelet S, Le Bon AM, Berges R, Suschetet M, Astorg P. Dietary carotenoids inhibit aflatoxin B1-induced liver preneoplastic foci and DNA damage in the rat: Role of the modulation of aflatoxin B1 metabolism. Carcinogenesis. 1998;19:403-411. DOI: 10.2013/0143-3334. 9525273
  27. 27. Farombi EO, Adepoju BF, Ola-Davies OE, Emerole GO. Chemoprevention of aflatoxin B1-induced genotoxicity and hepatic oxidative damage in rats by kolaviron, a natural bioflavonoid of Garcinia kola seeds. European Journal of Cancer Prevention: The Official Journal of the European Cancer Prevention Organisation. 2005;14:207-214. DOI: 10.2013/0959-8278. 15901988
  28. 28. Liu J, Yang CF, Lee BL, Shen HM, Ang SG, Ong CN. Effect of Salvia miltiorrhiza on aflatoxin B1-induced oxidative stress in cultured rat hepatocytes. Free Radical Research. 1999;31:559-568. DOI: 10.2013/1071-5762.10630680
  29. 29. Wild CP, Montesano R. A model of interaction: Aflatoxins and hepatitis viruses in liver cancer aetiology and prevention. Cancer Letters. 2009;286:22-28. DOI: 10.1016/j.canlet.2009.02.053
  30. 30. Shen HM, Ong CN. Mutations of the p53 tumor suppressor gene and ras oncogenes in aflatoxin hepatocarcinogenesis. Mutation Research. 1996;366:23-44. DOI: 10.2013/0027-5107.8921985
  31. 31. Golli-Bennour EE, Kouidhi B, Bouslimi A, Abid-Essefi S, Hassen W, Bacha H. Cytotoxicity and genotoxicity induced by aflatoxin B1, ochratoxin A, and their combination in cultured Vero cells. Journal of Biochemical and Molecular Toxicology. 2010;24:42-50. DOI: 10.1002/jbt.20310
  32. 32. Paget V, Sichel F, Garon D, Lechevrel M. Aflatoxin B1-induced TP53 mutational pattern in normal human cells using the FASAY (functional analysis of separated alleles in yeast). Mutation Research. 2008;656:55-61. DOI: 10.1016/j.mrgentox.2008.07.009
  33. 33. Van Vleet TR, Watterson TL, Klein PJ, Coulombe Jr. RA. Aflatoxin B1 alters the expression of p53 in cytochrome P450-expressing human lung cells. Toxicological Sciences. 2006;89:399-407. DOI: 10.1093/toxsci/kfj039
  34. 34. Vahakangas K. TP53 mutations in workers exposed to occupational carcinogens. Human Mutation. 2003;21:240-251. DOI: 10.1002/humu.10182
  35. 35. Staib F, Hussain SP, Hofseth LJ, Wang XW, Harris CC. TP53 and liver carcinogenesis. Human Mutation. 2003;21:201-216. DOI: 10.1002/humu.10176
  36. 36. Chan KT, Hsieh DP, Lung ML. In vitro aflatoxin B1-induced p53 mutations. Cancer Letters. 2003;199:1-7. DOI: S0304383503003379
  37. 37. Long XD, Ma Y, Huang HD, Yao JG, Qu de Y, Lu YL. Polymorphism of XRCC1 and the frequency of mutation in codon 249 of the p53 gene in hepatocellular carcinoma among Guangxi population, China. Molecular Carcinogenesis. 2008;47:295-300. DOI: 10.1002/mc.20384
  38. 38. Li D, Cao Y, He L, Wang NJ, Gu JR. Aberrations of p53 gene in human hepatocellular carcinoma from China. Carcinogenesis. 1993;14:169-173. DOI: 10.2013/0143-3334. 8382111
  39. 39. Kirk GD, Camus-Randon AM, Mendy M, Goedert JJ, Merle P, Trepo C, Brechot C, Hainaut P, Montesano R. Ser-249 p53 mutations in plasma DNA of patients with hepatocellular carcinoma from the Gambia. Journal of the National Cancer Institute. 2000;92:148-153. DOI: 10.2013/0027-8874.10639517
  40. 40. Aguilar F, Hussain SP, Cerutti P. Aflatoxin B1 induces the transversion of G-->T in codon 249 of the p53 tumor suppressor gene in human hepatocytes. Proceedings of the National Academy of Sciences of the United States of America. 1993;90:8586-8590. DOI: 10.2013/0027-8424.8397412
  41. 41. Denissenko MF, Koudriakova TB, Smith L, O'Connor TR, Riggs AD, Pfeifer GP. The p53 codon 249 mutational hotspot in hepatocellular carcinoma is not related to selective formation or persistence of aflatoxin B1 adducts. Oncogene. 1998;17:3007-3014. DOI: 10.1038/sj.onc.1202214
  42. 42. Duflot A, Hollstein M, Mehrotra R, Trepo C, Montesano R, Cova L. Absence of p53 mutation at codon 249 in duck hepatocellular carcinomas from the high incidence area of Qidong (China). Carcinogenesis. 1994;15:1353-1357. DOI: 10.2017/0143-3334.8033311
  43. 43. Sugasawa K. XPC: Its product and biological roles. Advances in Experimental Medicine and Biology. 2008;637:47-56. DOI: 10.2016/0065-2598.19181110
  44. 44. Nemzow L, Lubin A, Zhang L, Gong F. XPC: Going where no DNA damage sensor has gone before. DNA Repair (Amst). 2015;36:19-27. DOI: 10.1016/j.dnarep.2015.09.004
  45. 45. Fautrel A, Andrieux L, Musso O, Boudjema K, Guillouzo A, Langouet S. Overexpression of the two nucleotide excision repair genes ERCC1 and XPC in human hepatocellular carcinoma. Journal of Hepatology. 2005;43:288-293. DOI: 10.1016/j.jhep.2005.02.020
  46. 46. Strom SS, Estey E, Outschoorn UM, Garcia-Manero G. Acute myeloid leukemia outcome: Role of nucleotide excision repair polymorphisms in intermediate risk patients. Leuk Lymphoma. 2010;51:598-605. DOI: 10.3109/10428190903582804
  47. 47. Khan SG, Yamanegi K, Zheng ZM, Boyle J, Imoto K, Oh KS, Baker CC, Gozukara E, Metin A, Kraemer KH. XPC branch-point sequence mutations disrupt U2 snRNP binding, resulting in abnormal pre-mRNA splicing in xeroderma pigmentosum patients. Human Mutation. 2010;31:167-175. DOI: 10.1002/humu.21166
  48. 48. Gangwar R, Mandhani A, Mittal RD. XPC gene variants: A risk factor for recurrence of urothelial bladder carcinoma in patients on BCG immunotherapy. Journal of Cancer Research and Clinical Oncology. 2010;136:779-786. DOI: 10.1007/s00432-009-0717-y
  49. 49. Yao JG, Huang XY, Long XD. Interaction of DNA repair gene polymorphisms and aflatoxin B1 in the risk of hepatocellular carcinoma. International Journal of Clinical and Experimental Pathology. 2014;7:6231-6244. DOI: 10.2016/1936-2625.25337275
  50. 50. Li LM, Zeng XY, Ji L, Fan XJ, Li YQ, Hu XH, Qiu XQ, Yu HP. Association of XPC and XPG polymorphisms with the risk of hepatocellular carcinoma. Zhonghua gan zang bing za zhi = Zhonghua ganzangbing zazhi = Chinese Journal of Hepatology. 2010;18:271-275. DOI: 10.3760/cma.j.issn.1007-3418.2010.04.009
  51. 51. Wu XM, Ma Y, Deng ZL, Long XD. The polymorphism at codon 939 of xeroderma pigmentosum C gene and hepatocellular carcinoma among Guangxi population. Zhonghua Xiaohua Zazhi. 2010;30:846-848. DOI: 10.3760/cma.j.issn.0254-1432.2010.11.018
  52. 52. Van Houten B, Kuper J, Kisker C. Role of XPD in cellular functions: To TFIIH and beyond. DNA Repair (Amst). 2016;44:136-142. DOI: 10.1016/j.dnarep.2016.05.019
  53. 53. Manuguerra M, Saletta F, Karagas MR, Berwick M, Veglia F, Vineis P, Matullo G. XRCC3 and XPD/ERCC2 single nucleotide polymorphisms and the risk of cancer: A HuGE review. American Journal of Epidemiology. 2006;164:297-302. DOI: 10.1093/aje/kwj189
  54. 54. Sandoval-Carrillo A, Mendez-Hernandez EM, Vazquez-Alaniz F, Aguilar-Duran M, Tellez-Valencia A, Barraza-Salas M, Castellanos-Juarez FX, Llave-Leon OL, Salas-Pacheco JM. Polymorphisms in DNA repair genes (APEX1, XPD, XRCC1 and XRCC3) and risk of preeclampsia in a Mexican mestizo population. International Journal of Molecular Sciences. 2014;15:4273-4283. DOI: 10.3390/ijms15034273
  55. 55. Guo XF, Wang J, Lei XF, Zeng YP, Dong WG. XPD Lys751Gln polymorphisms and the risk of esophageal cancer: An updated meta-analysis. Internal Medicine Journal. 2015;54:251-259. DOI: 10.2169/internalmedicine.54.3256
  56. 56. Peng Q, Li S, Lao X, Chen Z, Li R, Qin X. Association between XPD Lys751Gln and Asp312Asn polymorphisms and hepatocellular carcinoma risk: A systematic review and meta-analysis. Medicine (Baltimore). 2014;93:e330. DOI: 10.1097/MD.0000000000000330
  57. 57. Zeng XY, Qiu XQ, Ji L, Yu HP. Study on the relationship between hepatoceilular carcinoma and the interaction between polymorphisms in DNA repair gene XPD and environmental factors. Zhong Hua Liu Xing Bing Xue Za Zhi. 2009;30:702-705. DOI: 10.3760/cma.j.issn.0254-6450.2009.07.014
  58. 58. Long XD, Ma Y, Zhou YF, Yao JG, Ban FZ, Huang YZ, Huang BC. XPD Codon 312 and 751 polymorphisms, and AFB1 exposure, and hepatocellular carcinoma risk. BMC Cancer. 2009;9:400. DOI: 10.1186/1471-2407-9-400
  59. 59. Hu HB, Tang CL, Li YB, Qin WB, Lu WX. Genic polymorphisms of XPD and risk of liver cancer among Guangxi Zhuang population. Shan Dong Yi Yao. 2016;56:33-35. DOI: 10.3969/j.issn.1002-266X.2016.13.012
  60. 60. Wallace SS. Base excision repair: A critical player in many games. DNA Repair (Amst). 2014;19:14-26. DOI: 10.1016/j.dnarep.2014.03.030
  61. 61. Kuznetsov NA, Koval VV, Fedorova OS. Mechanism of recognition and repair of damaged DNA by human 8-oxoguanine DNA glycosylase hOGG1. Biochemistry (Mosc). 2011;76:118-130. DOI: 10.2016/1608-3040.21568844
  62. 62. Boiteux S, Radicella JP. The human OGG1 gene: Structure, functions, and its implication in the process of carcinogenesis. Archives of Biochemistry and Biophysics. 2000;377:1-8. DOI: 10.1006/abbi.2000.1773
  63. 63. Weiss JM, Goode EL, Ladiges WC, Ulrich CM. Polymorphic variation in hOGG1 and risk of cancer: A review of the functional and epidemiologic literature. Molecular Carcinogenesis. 2005;42:127-141. DOI: 10.1002/mc.20067
  64. 64. Kohno T, Shinmura K, Tosaka M, Tani M, Kim SR, Sugimura H, Nohmi T, Kasai H, Yokota J. Genetic polymorphisms and alternative splicing of the hOGG1 gene, that is involved in the repair of 8-hydroxyguanine in damaged DNA. Oncogene. 1998;16:3219-3225. DOI: 10.1038/sj.onc.1201872
  65. 65. Kamali M, Kargar S, Heiranizadeh N, Zare M, Kargar S, Zare Shehneh M, Neamatzadeh H. Lack of any Association between the Hogg1 Ser326Cys polymorphism and breast cancer risk: A systematic review and meta-analysis of 18 studies. Asian Pacific Journal of Cancer Prevention. 2017;18:245-251. DOI: 10.22034/APJCP.2017.18.1.245
  66. 66. Wang W, Dang S, Li Y, Sun M, Jia X, Wang R, Liu J. hOGG1 Ser326Cys polymorphism and risk of hepatocellular carcinoma among East Asians: A meta-analysis. PLoS One. 2013;8:e60178. DOI: 10.1371/journal.pone.0060178
  67. 67. Ji L, Zeng XY, Li LM, Qiu XQ, Chen SX, Yu HP. Interaction between the single-nucleotide polymorphism of DNA repair gene hOGG1 and HBV infection and its susceptibility to hepatocellular carcinoma. Journal of Hygiene Research. 2011;40:705-708. DOI: 10.2017/1000-8020.2011.06.017
  68. 68. Peng T, Shen HM, Liu ZM, Yan LN, Peng MH, Li LQ, Liang RX, Wei ZL, Halliwell B, Ong CN. Oxidative DNA damage in peripheral leukocytes and its association with expression and polymorphisms of hOGG1: A study of adolescents in a high risk region for hepatocellular carcinoma in China. World Journal of Gastroenterology. 2003;9:2186-2193. DOI: 10.2016/1007-9327.14562375
  69. 69. Yuan T, Wei J, Luo J, Liu M, Deng S, Chen P. Polymorphisms of base-excision repair genes hOGG1 326cys and XRCC1 280His increase hepatocellular carcinoma risk. Digestive Diseases and Sciences. 2012;57:2451-2457. DOI: 10.1007/s10620-012-2192-6
  70. 70. Zhang H, He BC, He FC. Impact of DNA repair gene hOGG1 Ser326Cys polymorphism on the risk of hepatocellular carcinoma. Shijie Huaren xiaohua zazhi. 2006;14:2311-2314. DOI: 10.3969/j.issn.1009-3079.2006.23.010
  71. 71. Cheng B, Jungst C, Lin J, Caselmann WH. Potential role of human DNA-repair enzymes hMTH1, hOGG1 and hMYHalpha in the hepatocarcinogenesis. Journal of Huazhong University of Science and Technology. Medical sciences. 2002;22:206-211, 215. DOI: 10.2016/1672-0733.12658805
  72. 72. Su ZX, Li LQ, Peng T, Ye XP, Xiao KY, Shang LM. The implications of hOGG1 expression in hepatocellular carcinoma and cirrhosis tissues. China Medical Abstracts: Oncology. 2009;1:3-6. DOI: 10.3969/j.issn.1674-5671.2009.01.003
  73. 73. London RE. The structural basis of XRCC1-mediated DNA repair. DNA Repair (Amst). 2015;30:90-103. DOI: 10.1016/j.dnarep.2015.02.005
  74. 74. Long XD, Ma Y, Wei YP, Deng ZL. X-ray repair cross-complementing group 1 (XRCC1) Arg 399 Gln polymorphism and aflatoxin B1 (AFB1)-related hepatocellular carcinoma (HCC) in Guangxi population. Chinese Journal of Cancer Research. 2005;17:17-21. DOI: 10.1007/s11670-005-0004-7
  75. 75. Long XD, Ma Y, Wei YP, Deng ZL. The polymorphisms of GSTM1, GSTT1, HYL1*2, and XRCC1, and aflatoxin B1-related hepatocellular carcinoma in Guangxi population, China. Hepatology Research. 2006;36:48-55. DOI: 10.1016/j.hepres.2006.06.004
  76. 76. Zeng XY, Yu HP, Qiu XQ, Ji L, Li LM. A case-control study of polymorphism of XRCC1 gene and the risk of hepatocellular carcinoma. Zhonghua Ji Bing Kong Zhi Za Zhi. 2010;14:760-763. DOI: 10.3760/cma.j.issn.0254-6448.2010.11.018
  77. 77. He GZ, Huang TR, Deng JH, Zhang CY, Li JL. Research on association between XRCC1Arg399Gln polymorphism and liver cancer. Guangxi Zhongyi Xueyuan Xuebao. 2012;15:1-3. DOI: 10.3969/j.issn.1008-7486.2012.03.001
  78. 78. Chen BP, Long XD, Fu GH. Meta-analysis of XRCC1 Codon 399 polymorphism and susceptibility of hepatocellular carcinoma. Journal of Shanghai Jiao Tong University (Medical Science). 2011;31:1588-1602. DOI: 10.3969/j.issn.1674-8115.2011.11.018
  79. 79. Bishop DK, Ear U, Bhattacharyya A, Calderone C, Beckett M, Weichselbaum RR, Shinohara A. Xrcc3 is required for assembly of Rad51 complexes in vivo. Journal of Biological Chemistry. 1998;273:21482-21488. DOI: 10.2016/0021-9258. 9705276
  80. 80. Kawabata M, Kawabata T, Nishibori M. Role of recA/RAD51 family proteins in mammals. Acta Medica Okayama. 2005;59:1-9. DOI: 10.2016/0386-300X.PMID.15902993
  81. 81. Cheng S, Wang L, Wang L, Wang Z. Association of XRCC3 gene rs861539 polymorphism with gastric cancer risk: Evidence from a case-control study and a meta-analysis. International Journal of Clinical and Experimental Pathology. 2015;8:1911-1919. DOI: 10.2016/1936-2625.10086
  82. 82. Bei L, Xiao-Dong T, Yu-Fang G, Jian-Ping S, Zhao-Yu Y. DNA repair gene XRCC3 Thr241Met polymorphisms and lung cancer risk: A meta-analysis. Bull Cancer. 2015;102:332-339. DOI: 10.1016/j.bulcan.2015.02.003
  83. 83. Zhang X, Zhang L, Tian C, Yang L, Wang Z. Genetic variants and risk of cervical cancer: Epidemiological evidence, meta-analysis and research review. British Journal of Obstetrics and Gynaecology. 2014;121:664-674. DOI: 10.1111/1471-0528.12638
  84. 84. Long XD, Ma Y, Qu de Y, Liu YG, Huang ZQ, Huang YZ, Lin ZH, Wei NB, Zhou SC. The polymorphism of XRCC3 codon 241 and AFB1-related hepatocellular carcinoma in Guangxi population, China. Annals of Epidemiology. 2008;18:572-578. DOI: 10.1016/j.annepidem.2008.03.003
  85. 85. Long XD, Ma Y, Deng ZL, Huang YZ, Wei NB. Association of the Thr241Met polymorphism of DNA repair gene XRCC3 with genetic susceptibility to AFB1-related hepatocellular carcinoma in Guangxi population. Zhonghua Yi Xue Yi Chuan Xue Za Zhi. 2008;25:268-271. DOI: 940625058
  86. 86. Chen BP, Li M, Asaithamby A. New insights into the roles of ATM and DNA-PKcs in the cellular response to oxidative stress. Cancer Letters. 2012;327:103-110. DOI: 10.1016/j.canlet.2011.12.004
  87. 87. Shrivastav M, De Haro LP, Nickoloff JA. Regulation of DNA double-strand break repair pathway choice. Cell Research. 2008;18:134-147. DOI: 10.1038/cr.2007.111
  88. 88. Jolly CJ, Cook AJ, Manis JP. Fixing DNA breaks during class switch recombination. The Journal of Experimental Medicine. 2008;205:509-513. DOI: 10.1084/jem.20080356
  89. 89. Zhou C, Tang H, Yu J, Zhuang D, Zhang H. Blood-based DNA methylation of DNA repair genes in the non-homologous end-joining (NEHJ) pathway in patient with glioma. International journal of Clinical and Experimental Pathology. 2015;8:9463-9467. DOI: 10.2016/1936-2625.PMID.26464705
  90. 90. Xiao M, Shen Y, Chen L, Liao Z, Wen F. The rs7003908 (T>G) polymorphism in the XRCC7 gene and the risk of cancers. Molecular Biology Reports. 2014;41:3577-3582. DOI: 10.1007/s11033-014-3220-8
  91. 91. Jahantigh D, Salimi S, Mousavi M, Moossavi M, Mohammadoo-Khorasani M, Narooei-nejad M, Sandoughi M. Association between functional polymorphisms of DNA Double-Strand breaks in repair genes XRCC5, XRCC6 and XRCC7 with the risk of systemic lupus erythematosus in South East Iran. DNA Cell Biology. 2015;34:360-366. DOI: 10.1089/dna.2014.2465
  92. 92. Hsieh YH, Chang WS, Tsai CW, Tsai JP, Hsu CM, Jeng LB, Bau DT. DNA double-strand break repair gene XRCC7 genotypes were associated with hepatocellular carcinoma risk in Taiwanese males and alcohol drinkers. Tumour Biology. 2015;36:4101-4106. DOI: 10.1007/s13277-014-2934-5
  93. 93. Hsia TC, Chang WS, Chen WC, Liang SJ, Tu CY, Chen HJ, Liang JA, Tsai CW, Hsu CM, Tsai CH, Bau DT. Genotype of DNA double-strand break repair gene XRCC7 is associated with lung cancer risk in Taiwan males and smokers. Anticancer Research. 2014;34:7001-7005. DOI: 10.2016/0250-7005.PMID.25503126
  94. 94. Long XD, Yao JG, Huang YZ, Huang XY, Ban FZ, Yao LM, Fan LD. DNA repair gene XRCC7 polymorphisms (rs#7003908 and rs#10109984) and hepatocellular carcinoma related to AFB1 exposure among Guangxi population, China. Hepatology Research. 2011;41:1085-1093. DOI: 10.1111/j.1872-034X.2011.00866.x
  95. 95. Koike M, Yutoku Y, Koike A. Cloning, localization and focus formation at DNA damage sites of canine XRCC4. The Journal of Veterinary Medical Science. 2017;78:1865-1871. DOI: 10.1292/jvms.16-0381
  96. 96. Hammel M, Yu Y, Radhakrishnan SK, Chokshi C, Tsai MS, Matsumoto Y, Kuzdovich M, Remesh SG, Fang S, Tomkinson AE, Lees-Miller SP, Tainer JA. An intrinsically disordered APLF links Ku, DNA-PKcs, and XRCC4-DNA Ligase IV in an extended flexible Non-homologous end joining complex. Journal of Biological Chemistry. 2016;291:26987-27006. DOI: 10.1074/jbc.M116.751867
  97. 97. Mizuta R, Cheng HL, Gao Y, Alt FW. Molecular genetic characterization of XRCC4 function. International Immunology. 1997;9:1607-1613. DOI: 10.2016/0953-8178.PMID.9352367
  98. 98. Li Z, Otevrel T, Gao Y, Cheng HL, Seed B, Stamato TD, Taccioli GE, Alt FW. The XRCC4 gene encodes a novel protein involved in DNA double-strand break repair and V(D)J recombination. Cell. 1995;83:1079-1089. DOI: 0092-8674(95)90135-3
  99. 99. Yang CH, Lin YD, Yen CY, Chuang LY, Chang HW. A systematic Gene-Gene and Gene-Environment interaction analysis of DNA repair genes XRCC1, XRCC2, XRCC3, XRCC4, and oral cancer risk. OMICS. 2015;19:238-247. DOI: 10.1089/omi.2014.0121
  100. 100. Jung SW, Park NH, Shin JW, Park BR, Kim CJ, Lee JE, Shin ES, Kim JA, Chung YH. Polymorphisms of DNA repair genes in Korean hepatocellular carcinoma patients with chronic hepatitis B: Possible implications on survival. Journal of Hepatology. 2012;57:621-627. DOI: 10.1016/j.jhep.2012.04.039

Written By

Xue-Ming Wu, Zhi-Feng Xi, Jun Lu, Xing-Zhizi Wang, Tian-Qi Zhang, Xiao-Ying Huang, Jin-Guang Yao, Chao Wang, Zhong-Heng Wei, Chun-Ying Luo, Bing-Chen Huang, Qun-Qing Xu, Wen-Pei Yang, Qiang Xia and Xi-Dai Long

Reviewed: 02 May 2017 Published: 06 September 2017