Open access

The Molecular Epidemiology of DNA Repair Polymorphisms in Carcinogenesis

Written By

Paul W. Brandt-Rauf, Yongliang Li, Changmin Long and Regina Monaco

Submitted: 13 September 2012 Published: 22 May 2013

DOI: 10.5772/53936

From the Edited Volume

New Research Directions in DNA Repair

Edited by Clark Chen

Chapter metrics overview

2,039 Chapter Downloads

View Full Metrics

1. Introduction

There are well-established examples of highly penetrant mutations in genes that are directly involved in carcinogenesis and result in a high risk of cancer in the individuals who carry these mutations. Some of the best examples include syndromes of defective DNA repair, such as xeroderma pigmentosum [1]. However, these examples tend to be very rare and thus contribute minimally to the overall burden of cancer risk. Nevertheless, it has long been suspected that less penetrant susceptibility may be produced by much more common variants in the same cancer-related genes, for example, in the form of single nucleotide polymorphisms (SNPs), that presumably would be less disruptive and therefore produce more subtle effects on the function of the encoded proteins but which could contribute greatly to overall cancer attributable risk in populations due to their widespread occurrence [1]. Because several of these common polymorphisms occur in DNA repair proteins, many epidemiologic studies have examined their relationship to cancer risk [2-4].

These studies have looked at all different types of cancer, many different at-risk populations, several different DNA repair pathways, and a variety of polymorphisms at different sites [5-18]. The results to date at best have been inconsistent, conflicting and confusing with many examples of positive, negative or null associations between particular polymorphisms and particular cancers, even in multiple large meta-analyses of the data. For example, a very recent large, rigorous and systematic review of the literature on the involvement of DNA repair polymorphisms in human cancer reached the conclusion that because of the inconsistencies in the literature “none of the cancer genome-wide association studies (GWAs) published so far showed highly statistically significant associations for any of the common DNA repair gene variants” and “clarification of the discrepancies in the literature is needed.” [4] It was suggested that one way to proceed would be that “gene/environment and gene/lifestyle interactions for carcinogenic mechanisms involving DNA repair should be investigated more systematically and with less classification error.” [4] However, even in studies of populations with exposures to known environmental carcinogens and the cancers most closely associated with those exposures, the results of DNA repair polymorphism studies have not always been clear-cut; these inconsistencies may also be the result of poor exposure classification, multiple confounders and/or poor understanding of the exact mechanisms of DNA damage and/or repair [19-23]. In other words, what is needed is to study model systems where there are clear linkages between the exposure to the carcinogenic risk factor and the specific DNA damage that it produces with the DNA repair mechanisms that would correct those particular defects.

In environmental carcinogenesis studies of DNA repair polymorphisms, the majority of the work has focused on base excision repair (BER) or nucleotide excision repair (NER) pathways, since these are thought to play dominant roles in the repair of damage from exogenous carcinogens, including chemical carcinogens. In both of these pathways, numerous polymorphisms in numerous proteins that make up the DNA repair machinery have been examined. However, much of the focus has been on the particular proteins in the respective pathways that contain the most common polymorphic variants, in particular the x-ray cross complementing-1 (XRCC1) protein in BER and the xeroderma pigmentosum-D (XPD) protein in NER [24-38].

This is also understandable because of the critical roles that each of these proteins play in their respective pathways. For example, in BER the particular type of damage produced by exposure to a chemical carcinogen is usually recognized and removed by a specific DNA glycoslase. The BER apparatus includes numerous other proteins that complete the repair at the resultant abasic site once the damage is removed: apurinic/apyrimidinic endonuclease (APE1), poly(ADP-ribose) polymerase-1 (PARP-1), poly(ADP-ribose) polymerase-2 (PARP-2), DNA polymerase β (Pol β) and DNA ligase IIIα (Lig III). AP endonuclease is responsible for cleaving the phosphodiester bond at the abasic site created by the glycosylase. PARP-1 and to a lesser extent PARP-2 participate in the repair process by catalyzing ribosylation of a number of DNA-bound proteins, thereby decreasing the affinity of these proteins for DNA, and allowing the repair machinery to access the damaged site. Pol β, the polymerase involved in short patch repair, provides two essential activities, deoxyribophosphodiesterase activity which releases the 5' sugar phosphate group, and gap filling synthesis, where one nucleotide is added to the 3' OH. Finally, Lig III seals the nick in an ATP-dependent manner [39, 40]. The XRCC1 protein is critical to this process since it acts as a scaffold protein in this pathway and appears to enhance the activity of the other BER proteins. Although XRCC1 has not been demonstrated to contain enzymatic activity of its own, it is thus necessary for coordinating and regulating the early and late stages of BER through its protein interaction modules [41, 42].

XRCC1 is known to contain three common polymorphic sites that might be expected to have an effect on XRCC1 structure and function because they occur in or near important protein domains [11]. For example, the polymorphism at amino acid residue 194, which results in the substitution of a tryptophan for the normal arginine, occurs in the XRCC1 N-terminal domain from amino acid residues 1-195 that has been observed to mediate its interaction with the palm-thumb domain of Pol β [43]. A second polymorphism at amino acid residue 280, which results in the substitution of a histidine for the normal arginine, occurs in the region between the N-terminal domain and the BRCA1 carboxy terminal (BRCT1) domain of the protein and close to the nuclear localization signal site and thus could affect the relationship between these two critical domains and/or the protein’s localization ability [44]. The third and most common polymorphism in XRCC1 occurs at amino acid residue 399, resulting in the substitution of a glutamine for the normal arginine, within the highly conserved BRCT1 domain from amino acid residues 315-403, which has been associated with the functioning of PARP1, PARP2 and APE1 [45].

Like BER, NER occurs in a series of steps: damage recognition, unwinding and demarcation of the DNA, excision of the single-stranded fragment containing the damaged site, and DNA re-synthesis. NER is accomplished primarily through the action of proteins of the xeroderma pigmentosum family of genes which are categorized into 7 different groups (A-G). XPC and XPE proteins are involved in recognition of different types of DNA damage. XPB and XPD are DNA helicases that function as subunits of the transcription factor IIH complex (TFIIH) to promote DNA bubble formation at the damaged site by unwinding the DNA as XPA complexes with replication protein A (RPA) for demarcation. XPF and XPG are structure-specific endonucleases for excision of the damaged site. Finally, replicative DNA polymerase and DNA ligase I complete the repair [46, 47]. XPD is one of the major players in NER and is essential for life [48, 49].

XPD is also known to contain at least two common polymorphic sites, namely at amino acid residues 312 (aspartic acid->asparagine) and 751 (lysine->glutamine) [50]. The 751 site is assumed to be particularly important for XPD function since it occurs in the C-terminal domain of the protein which has been suggested to interact with the p44 helicase activator protein of the TFIIH complex [51]; also, it is been shown that an XPD mutation that results in the loss of the final 17 C-terminal amino acids, including residue 751, results in the clinical disease phenotype of trichothiodystrophy [52].

In summary, an ideal system for investigating the role of DNA repair polymorphisms in carcinogenesis might be an exposure to a known chemical carcinogen that produces specific types of DNA damage that are repaired by the BER and/or NER pathways where the effects of common polymorphisms in XRCC1 and XPD on the damage and repair could be studied.

Advertisement

2. A model for the study of the epidemiology of dna repair polymorphisms in carcinogenesis

Such a potential model system for the study of the role of DNA repair polymorphisms in chemical carcinogenesis is provided by the known carcinogen vinyl chloride (VC) because considerable detail is available concerning the molecular biology of its pathogenic pathway which allows for careful study of the role of DNA damage and repair in the carcinogenic process in exposed human populations through the application of molecular epidemiologic approaches (Figure 1).

As noted, VC is a well-established animal and human carcinogen. It is most strongly associated with liver cancer, in particular the rare, sentinel neoplasm of angiosarcoma of the liver (ASL), a malignant tumor of the endothelial cells of the liver [53]. However, VC has also been identified as a cause of hepatocellular carcinoma (HCC), the corresponding malignant tumor of the parenchymal cells of the liver [54]. In addition, it has been associated with other malignancies, e.g., lung and brain, although these associations remain much more controversial. The most significant exposures to VC occur in the petrochemical and plastics industries because VC is used in the manufacture of polyvinyl chloride, one of most high-volume plastics in the world. For example, it is estimated that worldwide more than 2,200,000 workers are probably occupationally exposed to VC. General population exposures also occur primarily through the air and water. For example, elevated levels of VC have been found not only in the air near VC manufacturing and processing facilities but also in the vicinity of many hazardous waste sites and municipal landfills, either due to the direct disposal of VC or from the microbial degradation of other chlorinated solvents to form VC. In some cases, dangerously high levels have been detected in the air at some of these landfills [53]. General population exposures may also occur from tobacco smoke, drinking water from PVC pipe, and consumption of food and beverages from PVC packaging and bottles, although probably at much lower levels.

VC is a gas so the most significant exposures are respiratory. Following inhalation, absorption is rapid in humans and most subsequent metabolism occurs in the liver [53]. Phase I metabolism is primarily via the cytochrome P-450 isoenzyme 2E1 (CYP2E1) to generate the reactive intermediates chloroethylene oxide (CEO) and chloracetaldehyde (CAA) which are further metabolized in phase II reactions by glutathione-S-transferases (GSTs) and aldehyde dehydrogenase 2 (ALDH2) to end products for ultimate excretion. However, CEO and CAA can readily interact with cellular macromolecules, including DNA, to produce promutagenic effects. VC biotransformation to CEO probably occurs principally in hepatocytes, but the epoxide can also reach and react with adjacent sinusoidal lining cells, so that mutagenic effects can occur in parenchymal liver cells and non-parenchymal endothelial cells, providing a logical rationale for the association between VC exposure and ASL as well as HCC [55]. The major VC-associated liver DNA adduct is 7-(2-oxoethyl)guanine, comprising up to 98% of all adducts formed. However, this adduct is eliminated from the DNA with a very short half-life, principally by chemical depurination, and is not considered to be promutagenic. On the other hand, three etheno DNA adducts are also formed in much less abundance, but they are known to be promutagenic. These are: N2,3-ethenoguanine (εG) ; 1,N6-ethenoadenine (εA) ; and 3,N4-ethenocytosine (εC) [56].

The promutagenic properties of etheno-DNA adducts that are not fully repaired by one or another of the DNA repair pathways have been well documented in experimental systems in vitro, as well as in vivo in bacterial and mammalian cells. The εA adduct generates A->T, A->G and A->C base changes; the εG adduct generates G->A base changes; and the εC adduct generates C->A and C->T base changes [55]. These experimental results are consistent with the tumor mutational spectra identified in exposed animals and humans in oncogenes and tumor suppressor genes. Of particular interest have been the A->T transversions at codons 179, 249 and 255 of the TP53 tumor suppressor gene generated by εA adducts and the G->A transitions at codon 13 of the K-ras oncogene generated by εG adducts, because of their frequent occurrence in human ASLs from VC-exposed individuals but not in sporadic ASLs in individuals without VC exposure. In addition, other results suggest that these VC-associated mutations, particularly the codon 13 K-ras mutation, may be a relatively early event in VC carcinogenesis, and thus the occurrence of these mutations may be useful biomarkers of cancer risk in exposed individuals, as discussed below.

Figure 1.

Proposed mechanism of VC-induced DNA damage and repair as a model system for the study of the effects of polymorphisms in BER and NER pathways.

The G->A transition at codon 13 of K-ras results in the substitution of an aspartic acid for the normal glycine at amino acid residue 13 in the encoded p21 protein product. This substitution is believed to be oncogenic, having been identified in other human tumors as well. The oncogenic mechanism of action of this substitution is thought to be through the production of a conformational change in p21 which may be responsible for altering its intrinsic GTPase activity, thus affecting signal transduction within the cell leading to uncontrolled growth and division [57]. Similarly, the A->T transversions at various codons of p53 produce their corresponding amino acid substitutions in the encoded p53 protein product, all changes that have been shown to cause the protein to adopt its so-called “malignant” conformation with a concomitant loss of its normal tumor suppressor activity [57]. These protein changes provide a useful indicator of the pathogenic consequences of the occurrence of the corresponding mutations, as well as convenient intermediate biomarkers of VC effect to study the molecular epidemiology of VC carcinogenesis in exposed human populations, including the effects of polymorphisms in the relevant DNA repair pathways.

It has been shown that the mutant ras-p21 protein containing aspartic acid for glycine at amino acid residue 13 can be distinguished from the wild-type protein and other mutant ras-p21 proteins immunologically with a mouse monoclonal antibody specific for this protein. For cells in culture that contain the mutant ras gene, it is possible to use this monoclonal antibody to detect mutant ras-p21 expression in the cells by immunocytochemistry and in the extracellular supernatant by immunoblotting. In analogous situations in vivo, mutant Asp 13 ras-p21 can be detected in tumor tissue by immunohistochemistry and in the serum by immunoblotting of VC-exposed workers with ASLs known to contain the mutant ras gene but not in the serum of VC-exposed workers with ASLs that do not contain the mutation or in unexposed controls [57-59].

An analogous, although slightly more complicated situation occurs with p53. As noted, all of the VC-induced mutations in the p53 gene have been shown to cause a similar conformational change in the encoded p53 protein that results in the exposure of a common epitope, which is normally not immunologically detectable in the wild-type protein. Thus, these mutant p53 proteins can be distinguished from wild-type p53 immunologically with a mouse monoclonal antibody that binds to this mutant-specific epitope. For cells in culture that contain the mutant p53 genes, it is possible to use this monoclonal antibody to detect mutant p53 protein expression in the cells by immunocytochemistry and in the extracellular supernatant by immunoblotting or by enzyme-linked immunosorbent assay (ELISA). In the analogous situation in vivo, mutant p53 can be detected in the tumor tissue by immunohistochemistry and in the serum by immunoblotting or ELISA of VC-exposed workers with ASLs known to contain the mutant p53 genes but not of VC-exposed workers with ASLs that do not contain the mutations or in unexposed controls. In some cases of mutant p53-positive tumors, it is known that individuals can also develop an antibody response to the mutant p53 which can obscure the detection of the mutant p53 protein itself. However, it is also possible to detect these auto-antibodies to mutant p53 using an ELISA. Thus, the detection in serum of mutant p53 protein and/or an antibody response to mutant p53 protein can be used together to best identify individuals who have a p53 mutation in their tumors [57, 60, 61].

Based on the above evidence, it seems that these serum biomarkers for mutant ras-p21 and mutant p53 accurately reflect the occurrence of the corresponding DNA damage in the target tissue of VC-exposed workers. In addition, these biomarkers have been identified not only in VC-exposed workers with ASLs but also in VC-exposed workers with non-malignant (but potentially pre-malignant) angiomatous lesions and in VC-exposed workers without any apparent neoplastic disease [57, 62-64]. In a large cohort of French VC workers, the presence of these biomarkers was found to occur with a highly statistically significant dose-response relationship with regard to estimated, cumulative VC exposure, supporting the claim that the generation of the biomarkers was indeed the result of the exposure [65]. Similar results with these biomarkers have been noted in several other VC workers cohorts around the world [66-71]. To date in these various studies, at least five VC-exposed biomarker-positive workers without ASL have developed subsequent liver lesions presumed to be ASLs, also suggesting that these biomarkers may have predictive value for the subsequent occurrence of cancer.

However, at any given level of VC exposure, some workers will have none, one or both mutant biomarkers. One possible explanation for this inter-individual variability is genetic differences in the proteins that metabolize VC or repair the DNA damage it produces. Although polymorphisms in the proteins involved in metabolizing VC have been shown to have an effect, polymorphisms in DNA repair proteins have been found to be even more significant.

There are several potential mechanisms by which VC-induced adducts could be repaired before they have a chance to cause mutations. As noted above, the oxoethyl adduct is removed rapidly by chemical depurination. The potential repair of the etheno adducts, however, is more complicated and involves the BER and NER pathways.

For example, the 1,N6-εA adducts are recognized and removed by 3-methyl adenine DNA glycosylase which is part of the BER pathway [55]. Likewise, the 3,N4-ethenocytosine adducts are also repaired with high efficiency by BER via the thymine DNA glycosylase. Therefore, polymorphisms in the BER pathway that could decrease DNA repair efficiency, particularly the polymorphisms in XRCC1, might be expected to result in an increase in εA and εC adduct levels at any given level of exposure in VC-exposed individuals with a resultant increase in the VC-associated mutant biomarkers, particularly the mutant p53 biomarker. In contrast, the N2,3-ethenoguanine adducts have been shown to be not very efficiently repaired by BER [56, 72]. Thus, if they are repaired, it is likely to be by a different DNA repair pathway such as NER. Therefore, polymorphisms in the NER pathway that could decrease DNA repair efficiency, particularly the polymorphisms in XPD, might be expected to result in an increase in εG adduct levels at any given level of exposure in VC-exposed individuals with a resultant increase in the VC-associated mutant biomarkers, particularly the mutant ras-p21 biomarker.

In fact in the aforementioned French VC worker cohort, we have been able to identify the effect of the XRCC1 polymorphisms on the occurrence of the mutant p53 biomarker, but not the mutant ras-p21 biomarker [73-75]. The difference in effect on the two biomarkers is expected, since, as noted the εA adducts that result in the mutant p53 biomarker are repaired efficiently by BER but the εG adducts that result in the mutant ras–p21 biomarker are not, so changes in XRCC1 might affect the former but should not affect the latter. Among the three XRCC1 polymorphisms, the most significant effect on the mutant p53 biomarker was attributable to the residue 399 polymorphism. In this case, individuals who were homozygous variant Gln-Gln at 399 had a statistically significant 1.9-fold risk of occurrence of the mutant p53 biomarker compared to homozygous Arg-Arg wild-type individuals, even after controlling for potential confounders including cumulative VC exposure, and the gene-environment interaction between the polymorphism and VC exposure appeared to be potentially supra-multiplicative [75]. Studies in other VC worker populations have found similar effects of the XRCC1 polymorphisms, particularly the 399 polymorphism, on the mutant p53 biomarker, as well as other biomarkers of DNA damage [76-79].

This is also consistent with various experimental results examining this model system. For example, molecular modeling of the BRCT1 domains of the normal and polymorphic forms of XRCC1 demonstrates that the 399 substitution produces significant conformational changes in this domain, including the loss of secondary structural features such as α-helices that can be critical for mediating protein-protein interactions that would allow XRCC1 to coordinate BER [80]. Also, studies of lymphoblasts from individuals of different genotypes exposed in vitro to the reactive metabolites of VC showed that cells with the XRCC1 399 homozygous variant Gln-Gln genotype had an approximate 4-fold decrease in efficiency of repair of εA DNA adducts compared to cells with the homozygous wild-type Arg-Arg genotype [74, 81], resulting in an approximate 1.8-fold increase in mutation frequency in the polymorphic cells compared to the wild-type cells as determined by the hypoxanthine-guanine phosphoribosyltransferase (HPRT) assay [82]. Based on mutational spectrum studies in CAA-exposed human cell lines [83], the resultant increase in  A DNA adducts would especially result in an increase in A->T transversions consistent with those found in the tumors of VC-exposed workers, as noted above.

Furthermore, in the French VC worker cohort, we have been able to identify the effect of the XPD polymorphisms on the occurrence of both mutant biomarkers, although the most marked and statistically significant effect was on the mutant ras-p21 biomarker, as expected [75]. In this case, individuals who were homozygous variant at either residue 312 or 751 had a statistically significant 2.6-3.0-fold increased risk of occurrence of the mutant ras–p21 biomarker compared to homozygous wild-type individuals, even after controlling for potential confounders including cumulative VC exposure. Furthermore, in the case of the residue 751 polymorphism, the gene-environment interaction between the polymorphism and VC exposure, as well as the gene-gene interaction between the XPD and CYP2E1 polymorphisms (which could increase VC metabolism to its promutagenic reactive metabolites and thus also increase etheno-DNA adducts at any given level of VC exposure with a resultant increase in the mutant biomarkers) appeared to be potentially multiplicative [75]. Once again, studies in other VC worker populations have found similar effects of the XPD polymorphisms on other biomarkers of DNA damage [77].

This is also consistent with various experimental results in this model system. For example, molecular modeling of the normal and polymorphic forms of XPD demonstrates that these substitutions produce discrete local conformational changes in the protein which affect its overall structure and could affect its function [82, 84], and, in particular, are projected to interfere with its protein-protein interactions and binding to other components of the TFIIH complex (Figure 2; adapted from Gibbons et al. [85]). Also, studies of lymphoblasts from individuals of different genotypes exposed in vitro to the reactive metabolites of VC showed that cells with the XPD 751 homozygous variant Gln-Gln genotype had an approximate 5-fold decrease in efficiency of repair of εG DNA adducts compared to cells with the homozygous wild-type Lys-Lys genotype [82], resulting in an approximate 4.8-fold increase in mutation frequency in the polymorphic cells compared to the wild-type cells as determined by the HPRT assay, even though there is no difference in the level of expression of the XPD protein among cells that are homozygous wild-type, heterozygous or homozygous polymorphic at this codon (Figure 3). Once again, based on mutational spectrum studies in CAA-exposed human cell lines [83], the resultant increase in  G DNA adducts would especially result in an increase in G->A transitions consistent with those found in the tumors of VC-exposed workers.

A thorough understanding of the molecular biology and molecular epidemiology of VC carcinogenesis can provide the basis for new molecular approaches to the prevention of VC-induced cancers and potentially other cancers related to DNA-damaging agents. For example, one approach to secondary prevention could be based on “personalized prevention” derived from knowledge of the status of individual’s DNA repair capability. Although little is currently known about methods for altering DNA repair activity, there is some evidence to suggest augmenting DNA repair may be possible. Several in vitro studies have shown that DNA repair processes can be increased by selenium-based compounds in response to radiation or chemically induced DNA damage [86]. More recently, a study in mice has suggested that selenocystine administration, although it did not protect against immediate DNA damage following ionizing radiation exposure, was nevertheless protective because it enhanced the rate of repair of the induced DNA damage [87]. In cohorts exposed to DNA damaging agents, determination of the dose of selenium compounds to provide an optimum effect on DNA repair could be based on the genetic status of the exposed individuals in terms of the presence of polymorphisms in key components of the repair apparatus, and the success of such interventions could be effectively monitored by following mutant biomarkers of DNA damage.

Figure 2.

Protein backbone structures showing the proposed interaction effect of XPD (blue), cyclin H (pink) and cdk7 (green) in the TFIIH complex.

Figure 3.

Levels of expression of the XPD protein in lymphocyte cell lines that are homozygous wild-type, heterozygous or homozygous polymorphic at codon 751.

Advertisement

3. Conclusion

VC provides an instructive model for the study of the role of DNA repair polymorphisms in chemical carcinogenesis. A detailed understanding of the molecular biology of VC carcinogenesis has provided new ways of studying the molecular epidemiology of VC carcinogenesis in exposed humans, which in turn may provide the basis for new approaches to the prevention and treatment of VC-related cancer. This model could also have much broader implications, since other potential carcinogenic exposures share some of the same molecular biologic pathways of damage and repair as VC similar molecular epidemiologic biomarkers could be useful for monitoring their carcinogenic process and the effect of altered susceptibility due to changes in DNA repair capability. Such studies in additional model systems would further help to define the exact significance of DNA repair polymorphisms in the development of human cancers.

Advertisement

Acknowledgments

This work was supported in part by grants to PWB-R from NIOSH, R01-OH04192 and R01-OH07590.

References

  1. 1. Au WW (2006) Heritable Susceptibility Factors for the Development of Cancer. J. radiat. res. 47:B13-B17.
  2. 2. Berwick M, Vineis P (2000) Markers of DNA Repair and Susceptibility to Cancer in Humans: An Epidemiologic Review. J. natl. cancer inst. 92:874-897.
  3. 3. Goode EL, Ulrich CM, Potter JD (2002) Polymorphisms in DNA Repair Genes and Associations with Cancer Risk. Cancer epidemiol biomarkers prev. 11:1513-1530.
  4. 4. Ricceri F, Matullo G, Vineis P (2012) Is There Evidence of Involvement of DNA Repair Polymorphisms in Human Cancer? Mutat. res. 736:117-121.
  5. 5. Jiricny J, Nystrom-Lahti M (2000) Mismatch Repair Defects in Cancer. Curr. opin. genet. dev. 10:157-161.
  6. 6. Hung RJ, Hall J, Brennan P, Boffetta P (2005) Genetic Polymorphisms in the Base Excision Repair Pathway and Cancer Risk: A HuGE Review. Am. j. epidemiol. 162:925-942.
  7. 7. Weiss JM, Goode EL, Ladiges WC, Ulrich CM (2005) Polymorphic Variation in hOGG1 and Risk of Cancer: A review of the Functional and Epidemiologic Literature. Mol. carcinog. 42:127-141.
  8. 8. Clarkson SG, Wood RD (2005) Polymorphisms in the Human XPD (ERCC2) Gene, DNA Repair Capacity and Cancer Susceptibility: An Appraisal. DNA repair 4:1068-1074.
  9. 9. Hu Z, Ma H, Chen F, Wei Q, Shen H (2005) XRCC1 Polymorphisms and Cancer Risk: A Meta-Analysis of 38 Case-Control Studies. Cancer epidemiol. biomarkers prev. 14:1810-1818.
  10. 10. Manuguerra M, Saletta F, Karagas MR, Berwick M, Vegila F, Vineis P, Matullo G (2006) XRCC3 and XPD/ERCC2 Single Nucleotide Polymorphisms and the Risk of Cancer: A HuGE Review. Am. j. epidemiol. 164:297-302.
  11. 11. Tudek B (2007) Base Excision Repair Modulation as a Risk Factor for Human Cancers. Mol. aspects med. 28:258-275.
  12. 12. Bugni JM, Han J, Tsai M, Hunter DJ, Samson LD (2007) Genetic Association and Functional Studies of Major Polymorphic Variants of MGMT. DNA repair 6:1116-1126.
  13. 13. Qiu L, Wang Z, Shi X, Wang Z (2008) Associations between XPC Polymorphisms and Risk of Cancers: A Meta-Analysis. Eur. j. cancer 44:2241-2253.
  14. 14. Wang F, Chang D, Hu F, Sui H, Han B, Li D, Zhao Y (2008) DNA Repair Gene XPD Polymorphisms and Cancer Risk: A Meta-Analysis Based on 56 Case-Control Studies. Cancer epidemiol. biomarkers prev. 17:507-517.
  15. 15. Jiang J, Zhang X, Yang H, Wang W (2009) Polymorphisms of DNA Repair Genes: ADPRT, XRCC1, and XPD and Cancer Risk in Genetic Epidemiology. In: Verma M, editor. Methods in Molecular Biology, Cancer Epidemiology, vol. 471. Totowa: Humana Press. pp. 305-333.
  16. 16. Wilson DM, Kim D, Berquist BR, Sigurdson AJ (2011) Variation in Base Excision Repair Capacity. Mutat. res. 711:100-112.
  17. 17. Ding D, Zhang Y, Yu H, Guo Y, Jiang L, He X, Ma W, Zheng W (2012) Genetic Variation of XPA Gene and Risk of Cancer: A Systematic Review and Pooled Analysis. Int. j. cancer 131:488-496.
  18. 18. Gossage L, Perry C, Abbotts R, Madhusudan S (2012) Base Excision Repair Factors are Promising Prognostic and Predictive Markers in Cancer. Curr. mol. pharmacol. 5:115-124.
  19. 19. Hulla JE, Miller MS, Taylor JA, Hein DW, Furlong CE, Omiecinski CJ, Kunkel TA (1999) Symposium Overview: The Role of Genetic Polymorphism and Repair Deficiencies in Environmental Disease. Toxicol. sci. 47:135-143.
  20. 20. de Boer JG (2002) Polymorphisms in DNA Repair and Environmental Interactions. Mutat. res. 509:201-210.
  21. 21. Belitsky GA, Yakubovskaya MG (2008) Genetic Polymorphism and Variability of Chemical Carcinogenesis. Biochemistry 73:543-554.
  22. 22. Klaunig JE, Wang Z, Pu X, Zhou S (2011) Oxidative Stress and Oxidative Damage in Chemical Carcinogenesis. Toxicol. appl. pharmacol. 254:86-99.
  23. 23. Simonelli V, Mazzei F, D’Errico M, Dogliotti E (2012) Gene Susceptibility to Oxidative Damage: From Single Nucleotide Polymorphisms to Function. Mutat. res. 736:104-116.
  24. 24. Stern MC, Johnson LR, Bell DA, Taylor JA (2002) XPD Codon 751 Polymorphism, Metabolism Genes, Smoking, and Bladder Cancer Risk. Cancer epidemiol. biomarkers prev. 11:1004-1011.
  25. 25. Shen M, Hung RJ, Brennan P, Malaveille C, Donato F, Placidi D, Carta A, Hautefeuille A, Boffetta P, Porru S (2003) Polymorphisms of the DNA Repair Genes XRCC1, XRCC3, XPD, Interaction with Environmental Exposures, and Bladder Cancer Risk in a Case-Control Study in Northern Italy. Cancer epidemiol. biomarkers prev. 12:1234-1240.
  26. 26. Gao WM, Romkes M, Day RD, Siegfried JM, Luketich JD, Mady HH, Melhem MF, Keohavong P (2003) Association of the DNA Repair Gene XPD Asp312Asn Polymorphism with p53 Gene Mutations in Tobacco-Related Non-Small Cell Lung Cancer. Carcinogenesis 24:1671-1676.
  27. 27. Terry MB, Gammon MD, Zhang FF, Eng SM, Sagiv SK, Paykin AB, Wang Q, Hayes S, Teitelbaum SL, Neugut AI, Santella RM (2004) Polymorphism in the DNA Repair Gene XPD, Polycyclic Aromatic Hydrocarbon-DNA Adducts, Cigarette Smoking, and Breast Cancer Risk. Cancer epidemiol. biomarkers prev. 13:2053-2058.
  28. 28. Han J, Hankinson SE, Colditz GA, Hunter DJ (2004) Genetic Variation in XRCC1, Sun Exposure, and Risk of Skin Cancer. Br. j. cancer 91:1604-1609.
  29. 29. Neumann AS, Sturgis EM, Wei Q (2005) Nucleotide Excision Repair as a Marker for Susceptibility to Tobacco-Related Cancers: A Review of Molecular Epidemiological Studies. Mol. carcinog. 42:65-92.
  30. 30. Han J, Colditz GA, Liu JS, Hunter DJ (2005) Genetic Variation in XPD, Sun Exposure, and Risk of Skin Cancer. Cancer epidemiol. biomarkers prev. 14:1539-1544.
  31. 31. Leng S, Cheng J, Zhang L, Niu Y, Dai Y, Pan Z, Li B, He F, Zheng Y (2005) The Association of XRCC1 Haplotypes and Chromosomal Damage Levels in Peripheral Blood Lymphocytes Among Coke-Oven Workers. Cancer epidemiol. biomarkers prev. 14: 1295-1301.
  32. 32. Zhao XH, Jia G, Liu YQ, Liu SW, Yan L, Jin Y, Liu N (2006) Association Between Polymorphisms of DNA Repair Gene XRCC1 and DNA Damage in Asbestos-Exposed Workers. Biomed. environ. sci. 19:232-238.
  33. 33. Long XD, Ma Y, Wei YP, Deng ZL (2006) The Polymorphisms of GSTM1, GSTT1, HYL1*2, and XRCC1, and Aflatoxin B1-Related Hepatocellular Carcinoma in Guangxi Population, China. Hepatol. res. 36:48-55.
  34. 34. Neri M, Ugolini D, Dianzani I, Gemignani F, Landi S, Cesario A, Magnani C, Mutti L, Puntoni R, Bonassi S (2008) Genetic Susceptibility to Malignant Pleural Mesothelioma and Other Asbestos-Associated Diseases. Mutat. res. 659:126-136.
  35. 35. Long XD, Ma Y, Zhou YF, Yao JG, Ban FZ, Huang YZ, Huang BC (2009) XPD Codon 312 and 751 Polymorphisms, and AFB1 Exposure, and Hepatocellular Carcinoma Risk. BMC cancer 9:400.
  36. 36. Sterpone S, Cozzi R (2010) Influence of XRCC1 Genetic Polymorphisms on Ionizing Radiation-Induced DNA Damage and Repair. J. nucleic acids
  37. 37. Toumpanakis D, Theocharis SE (2011) DNA Repair Systems in Malignant Mesothelioma. Cancer lett. 312:143-149.
  38. 38. Zhang XH, Zhang X, Zhang L, Chen Q, Yang Z, Yu J, Fu H, Zhu YM (2012) XRCC1 Arg399Gln Was Associated with Repair Capacity for DNA Damage Induced by Occupational Chromium Exposure. BMC res. notes 5:263.
  39. 39. Fortini P, Pascucci B, Parlanti E, D’Errico M, Simonelli V, Dogliotti E (2003) The Base Excision Repair: Mechanisms and Its Relevance for Cancer Susceptibility. Biochemie 85:1053-1071.
  40. 40. Baute J, Depicker A (2008) Base Excision Repair and Its Role in Maintaining Genome Stability. Crit. rev. biochem mol. biol. 43:239-276.
  41. 41. Thompson LH, West MG (2000) XRCC1 Keeps DNA from Getting Stranded. Mutat. res. 459:1-18.
  42. 42. Caldecott KW (2003) XRCC1 and DNA Strand Break Repair. DNA repair 2:955-969.
  43. 43. Marintchev A, Mullen MA, Maciejewski MW, Pan B, Gryk MR, Mullen GP (1999) Solution Structure of the Single-Strand Break Repair Protein XRCC1 N-Terminal Domain. Nat. struct. biol. 6: 884-893.
  44. 44. Marintchev A, Robertson A, Dimitriadis EK, Prasad R, Wilson SH, Mullen GP (2000) Domain Specific Interaction in the XRCC1 Polymerase Beta Complex. Nucleic acids res. 28:2049-2059.
  45. 45. Vidal AE, Boiteux S, Hickson ID, Radicella JP (2001) XRCC1 Coordinates the Initial and Late Stages of DNA Abasic Site Repair through Protein-Protein Interactions. EMBO j. 20:6530-6539.
  46. 46. Nouspikel T (2009) DNA Repair in Mammalian Cells: Nucleotide Excision Repair: Variations in Versatility. Cell. mol. life sci. 66:994-1009.
  47. 47. Bergoglio V, Magnaldo T (2006) Nucleotide Excision Repair and Related Human Diseases. Genome dyn. 1:35-52.
  48. 48. Oksenych V, Coin F (2010) The Long Unwinding Road: XPB and XPD Helicases in Damaged DNA Opening. Cell cycle 9:90-96.
  49. 49. Lehmann AR (2008) XPD Structure Reveals Its Secrets. DNA repair 7:1912-1915.
  50. 50. Benhamou S, Sarasin A (2002) ERCC2/XPD Gene Polymorphisms and Cancer Risk. Mutagenesis 17:463-469.
  51. 51. Bienstock RJ, Skovaga M. Mandavilli BS, Van Houten B (2003) Structural and Functional Characterization of the Human DNA Repair Helicase XPD by Comparative Molecular Modeling and Site-Directed Mutagenesis of the Bacterial Repair Protein UvrB. J. biol. chem.. 278:5309-5316.
  52. 52. Botta E, Nardo T, Broughton BC, Marinoni S, Lehmann AR, Stefanini M (1998) Analysis of Mutations in the XPD Gene in Italian Patients with Trichothiodystrophy: Site of Mutation Correlates with Repair Deficiency, but Gene Dosage Appears to Determine Clinical Severity. J. hum. genet. 63:1036-1048.
  53. 53. Agency for Toxic Substances and Disease Registry (2006) Toxicological Profile for Vinyl Chloride. Atlanta: US Department of Health and Human Services.
  54. 54. Grosse Y, Baan R, Straif K, Secretan B, El Ghissassi F, Bouvard V, Altieri A, Cogliano V (2007) Carcinogenicity of 1,3-Butadiene, Ethylene Oxide, Vinyl Chloride, Vinyl Fluoride, and Vinyl Bromide. Lancet oncol. 8:679-680.
  55. 55. Dogliotti E (2006) Molecular Mechanisms of Carcinogenesis by Vinyl Chloride. Ann. 1st super sanita 42:163-169.
  56. 56. Cohen SM, Storer RD, Criswell KA, Doerrer NG, Dellarco VL, Pegg DG, Wojcinski ZW, Malarkey DE, Jacobs AC, Klaunig JE, Swenberg JA, Cook JC (2009) Hemangiosarcoma in Rodents: Mode-of-Action Evaluation and Human Relevance. Toxicol. sci. 111:4-18.
  57. 57. Li Y, Asherova M, Marion MJ, Brandt-Rauf PW (1998) Mutant Oncoprotein Biomarkers in Chemical Carcinogenesis. In: Mendelsohn ML, Mohr LC, Peeters JP, editors. Biomarkers – Medical and Workplace Applications. Washington: Joseph Henry Press. pp. 345-353.
  58. 58. DeVivo I, Marion MJ, Smith SJ, Carney WP, Brandt-Rauf PW (1994) Mutant c-Ki-ras p21 Protein in Chemical Carcinogenesis in Humans Exposed to Vinyl Chloride. Cancer causes control 5:273-278.
  59. 59. Li Y, Marion MJ, Asherova M, Coulibaly D, Smith SJ, Do T, Carney WP, Brandt-Rauf PW (1998) Mutant p21ras in Vinyl Chloride Exposed Workers. Biomarkers 3:433-439.
  60. 60. Brandt-Rauf PW, Chen JM, Marion MJ, Smith SJ, Luo JC, Carney W, Pincus MR (1996) Conformational Effects in the p53 Protein of Mutations Induced during Chemical Carcinogenesis: Molecular Dynamic and Immunologic Analyses. J. protein chem. 15:367-375.
  61. 61. Smith SJ, Li Y, Whitney R, Marion MJ, Partilo S, Carney WP, Brandt-Rauf PW (1998) Molecular Epidemiology of p53 Protein Mutations in Workers Exposed to Vinyl Chloride. Am. j. epidemiol. 147:302-308.
  62. 62. Brandt-Rauf PW, Luo JC, Cheng TJ, Du CL, Wang JD, Marion MJ (2000) Mutant Oncoprotein Biomarkers of Vinyl Chloride Exposure: Application to Risk Assessment. In: Anderson D, Karakaya AE, Sram RJ, editors. Human Monitoring after Environmental and Occupational Exposure to Chemical and Physical Agents. Amsterdam: IOS Press. pp. 243-248.
  63. 63. Brandt-Rauf PW, Luo JC, Cheng TJ, Du CL, Wang JD, Rosal R, Do T, Marion MJ (2002) Molecular Biomarkers and Epidemiologic Risk Assessment. Hum. ecol. risk assess. 8:1295-1301.
  64. 64. Marion MJ, DeVivo I, Smith S, Luo JC, Brandt-Rauf PW (1996) The Molecular Epidemiology of Occupational Carcinogenesis in Vinyl Chloride Exposed Workers. Int. arch. occup. environ. health 68:394-398.
  65. 65. Schindler J, Li Y, Marion MJ, Paroly A, Brandt-Rauf PW (2007) The Effect of Genetic Polymorphisms in the Vinyl Chloride Metabolic Pathway on Mutagenic Risk. J. hum. genet. 52:448-455.
  66. 66. Trivers GE, Cawley HL, DeBenedetti VM, Hollstein M, Marion MJ, Bennett ML, Hoover ML, Prives CC, Tamburro CC, Harris CC (1995) Anti-p53 Antibodies in Sera of Workers Occupationally Exposed to Vinyl Chloride. J. natl. cancer inst. 87:1400-1407.
  67. 67. Luo JC, Liu HT, Cheng TJ, Du CL, Wang JD (1998) Plasma Asp13-Ki-ras Oncoprotein Expression in Vinyl Chloride Monomer Workers in Taiwan. J. occup. environ. med. 40:1053-1058.
  68. 68. Luo JC, Liu HT, Cheng TJ, Du CL, Wang JD (1999) Plasma p53 Protein and Anti-p53 Antibody Expression in Vinyl Chloride Monomer Workers in Taiwan. J. occup. environ. med. 41:521-526.
  69. 69. Luo JC, Cheng TJ, Du CL, Wang JD (2003) Molecular Epidemiology of Plasma Oncoproteins in Vinyl Chloride Monomer Workers in Taiwan. Cancer detect. prev. 27:94-101.
  70. 70. Mocci F, De Biasio AL, Nettuno M (2003) Anti-p53 Antibodies as Markers of Carcinogenesis in Exposures to Vinyl Chloride. G. ital. med. lav. ergon. 25:S21-S23.
  71. 71. Mocci F, Nettuno M (2006) Plasma Mutant-p53 Protein and Anti-p53 Antibody as a Marker: An Experience in Vinyl Chloride Workers in Italy. J. occup. environ. med. 48:158-164.
  72. 72. Dosanjh MK, Chenna A, Kim E, Fraenkel-Conrat H, Samson L, Singer B (1994) All Four Known Cyclic Adducts Formed in DNA by the Vinyl Chloride Metabolite Chloroacetaldehyde are Released by a Human DNA Glycosylase. Proc. natl. acad. sci. usa 91:1024-1028.
  73. 73. Li Y, Marion MJ, Rundle A, Brandt-Rauf PW (2003) A common Polymorphism in XRCC1 as a Biomarker of Susceptibility for Chemically Induced Genetic Damage. Biomarkers 8:408-414.
  74. 74. Li Y, Marion MJ, Zipprich J, Freyer G, Santella RM, Kanki C, Brandt-Rauf PW (2006) The Role of XRCC1 Polymorphisms in Base Excision Repair of Etheno-DNA Adducts in French Vinyl Chloride Workers. Int. j. occup. med. environ. health 19:45-52.
  75. 75. Li Y, Marion MJ, Zipprich J, Santella RM, Freyer G, Brandt-Rauf PW (2009) Gene-Environment Interactions between DNA Repair Polymorphisms and Exposure to the Carcinogen Vinyl Chloride. Biomarkers 14:148-155.
  76. 76. Wong RH, Du CL, Wang JD, Chan CC, Luo JC, Cheng TJ (2002) XRCC1 and CYP2E1 Polymorphisms as Susceptibility Factors of Plasma Mutant p53 Protein and Anti-p53 Antibody Expression in Vinyl Chloride Monomer-Exposed Polyvinyl Chloride Workers. Cancer epidemiol. biomarkers prev. 11:475-482.
  77. 77. Zhu SM, Xia ZL, Wang AH, Ren XF, Jiao J, Zhao NQ, Qian J, Jin L, Christiani DC (2008) Polymorphisms and Haplotypes of DNA Repair and Xenobiotic Metabolism Genes and Risk of DNA Damage in Chinese Vinyl Chloride Monomer (VCM)-Exposed Workers. Toxicol. let. 178:88-94.
  78. 78. Ji F, Wang W, Xia ZL, Zheng YJ, Qiu YL, Wu F, Miao WB, Jin RF, Qian J, Jin L, Zhu YL, Christiani DC (2010) Prevalence and Persistence of Chromosomal Damage and Susceptible Genotypes of Metabolic and DNA Repair Genes in Chinese Vinyl Chloride-Exposed Workers. Carcinogenesis 31:648-653.
  79. 79. Wang Q, Ji F, Sun Y, Qiu YL, Wang W, Wu F, Miao WB, Li Y, Brandt-Rauf PW, Xia ZL (2010) Genetic Polymorphisms of XRCC1, HOGG1 and MGMT and Micronucleus Occurrence in Chinese Vinyl Chloride-Exposed Workers. Carcinogenesis 31:1068-1073.
  80. 80. Monaco R, Rosal R, Dolan MA, Pincus MR, Brandt-Rauf PW (2007) Conformational Effects of a Common Codon 399 Polymorphism on the BRCT1 Domain of the X-Ray Cross-Complementing-1 Protein. Protein j. 26:541-546.
  81. 81. Li Y, Long C, Lin G, Marion MJ, Freyer G. Santella RM, Brandt-Rauf PW (2009) Effects of the XRCC1 Codon 399 Polymorphism on the Repair of Vinyl Chloride Metabolite-Induced DNA Damage. J. carcinogen. 8:108-112.
  82. 82. Brandt-Rauf PW, Li Y, Monaco R, Kovvali G, Marion MJ (2012) Plastics and Carcinogenesis: The Example of Vinyl Chloride. J. carcinogen. 11:50-60.
  83. 83. Matsuda T, Yagi T, Kawanishi M, Matsui S, Takebe H (1995) Molecular Analysis of Mutations Induced by 2-Chloroacetaldehyde, the Ultimate Carcinogenic Form of Vinyl Chloride, in Human Cells Using Shuttle Vectors. Carcinogenesis 16:2389-2394.
  84. 84. Monaco R, Rosal R, Dolan MA, Pincus MR, Freyer G, Brandt-Rauf PW (2009) Conformational Effects of a Common Codon 751 Polymorphism on the C-Terminal Domain of the Xeroderma Pigmentosum D Protein. J carcinogen. 8:93-97.
  85. 85. Gibbons BJ, Brignole EJ, Azubel M, Murakami K, Voss NR, Bushnell DA, Asturias FJ, Kornberg RD (2012) Subunit Architecture of General Transcription Factor TFIIH. Proc. natl. acad. sci. usa 109:1949-1954.
  86. 86. Valdiglesias V, Pasaro E, Mendez J, Laffon B (2010) In Vitro Evaluation of Selenium Genotoxic, Cytotoxic, and Protective Effects: A Review. Arch. toxicol. 84:337-351.
  87. 87. Kunwar A, Jayakumar S, Bhilwade HN, Bag PP, Bhatt H, Chaubey RC, Priyadarsini KI (2011) Protective Effects of Selenocystine Against γ-Radiation-Induced Genotoxicity in Swiss Albino Mice. Radiat. environ. biophys. 50:271-280.

Written By

Paul W. Brandt-Rauf, Yongliang Li, Changmin Long and Regina Monaco

Submitted: 13 September 2012 Published: 22 May 2013