Formamidopyrimidine adducts are detected using the comet assay in human cells treated with reactive metabolites of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK)

https://doi.org/10.1016/j.mrfmmm.2006.04.005Get rights and content

Abstract

4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) is the most lung-specific of the carcinogens present in tobacco smoke. Its bioactivation in cells leads to a small amount of methylation or pyridyloxobutylation DNA damage. Considering its great sensitivity, the comet assay seems a technique of choice to investigate NNK-related damage. Several strategies were used to impart some specificity to the assay: (1) using analogs that produce a limited variety of DNA lesions, as they mimic either the methylation or the pyridyloxobutylation pathway; (2) using cells with different bioactivation abilities; (3) using alkali conversion and/or enzymes specific for cleaving particular classes of damage; (4) using different lysis conditions to convert a specific class of DNA lesions into enzyme-sensitive lesions. We determined that several NNK-associated lesions can be detected with some specificity with the comet assay. For the methylation pathway, they are AP sites and the more frequent formamidopyrimidine (fapy) adducts. These fapy adducts correspond to N7-methylguanines generated in the cells that were ring-opened during the assay by the lysis solution at pH 10. For the pyridyloxobutylation pathway, alkylphosphotriesters and a roughly equal frequency of fapy sites were detected. By analogy to the methylation damage, these fapy adducts are thought to be the ring-opened form of N7-pyridyloxobutylguanines (N7-pobG). N7-pobG are unstable and this constitutes the first indirect demonstration of their formation in cells. But contrary to N7-m-fapy, the lysis time or pH did not influence the frequency of N7-pob-fapy adducts detected, suggesting that they already exist in the cells and are not related to the experimental conditions. These N7-pob-fapy have a strong mutagenic potential and we think that the comet assay, in spite of its limitations, is a good way to study them considering their low frequency and the inherent instability of the adduct from which they originate.

Introduction

N-Nitrosamines are thought to play a major role in carcinogenesis related to tobacco consumption. Among the tobacco specific nitrosamines, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) is the most abundant and the strongest carcinogen [1], [2]. It exhibits lung-selective toxicity and induces primarily adenocarcinoma of the lung on laboratory animals [3]. This led to the assumption that NNK accounts for recent increases in the relative incidence of human lung adenocarcinoma in smokers [4], [5].

Bioactivation to reactive intermediates is necessary for NNK to be genotoxic and its activation has been extensively studied [6] in freshly isolated human lung cells [7] and in peripheral human lung microsomes [8]. The two α carbons of the nitrosamino group of NNK can be hydroxylated, yielding unstable intermediates that can lead to DNA alkylation (Fig. 1). The α-methylene hydroxylation of NNK leads to the formation of methyldiazohydroxides, which can methylate DNA and the α-methyl hydroxylation of NNK results in the formation of oxobutyldiazohydroxides, which can alkylate DNA through a reaction of pyridyloxobutylation. Both types of α-carbon hydroxylation clearly exist in human lung cells [3], [7] and both pathways seem to participate in NNK-related lung carcinogenesis in humans [3], [9]. But the extent to which they do, remains to be determined. These pathways can be investigated separately through the use of analogs that generate either one type of reactive species or the other. For instance, N-nitroso(acetoxymethyl)methylamine (NDMAOAc) and 4-(acetoxymethylnitrosamino)-1-(3-pyridyl)-1-butanone (NNKOAc) mimic respectively the methylation or the pyridyloxobutylation pathway (Fig. 1). NNK bioactivation, that is the biotransformation in alkylating reactive species, is not as frequent as other possible biotransformations corresponding for example to detoxification pathways, but it is thought to be responsible for the carcinogenetic effect of NNK. In fact, the relative occurrence of bioactivation in reactive species seems significantly higher in alveolar type II cells and macrophages than in other pulmonary cell types, making them potential target for NNK toxicity [7].

DNA adducts generated by NNK through the methylation or the pyridyloxobutylation pathway (Table 1) have been extensively studied to determinate which of them could be relevant to carcinogenesis [3]. Laboratory animals treated with NNK show the following adducts in their lung: N7-methylguanine (N7-mG), O6-methylguanine (O6-mG) and to a lesser extend of O4-methylthymine (O4-mT), all these are due to the methylation pathway. Some pyridyloxobutyl adducts have also been detected such as O6-pyridyloxobutylguanine (O6-pobG) or phosphate adducts (pyridyloxobutylphosphotriesters) [6], [10], [11]. NNK generates single-strand breaks [6] but exactly how they are produced remains undetermined. It has been suggested that they derived from the methylation pathway by spontaneous or enzymatic depurination of N7-mG or N3-mA [3] but they more likely result from the pyridyloxobutylation pathway by the generation of pyridyloxobutylphosphotriesters [10], [11]. The O6-alkyl adducts (O6-mG and O6-pobG) are considered as major players in NNK-related carcinogenicity because of their mutagenetic potential and the fact that they are repaired both by the same O6-methylguanine methyltransferase which is easily overwhelmed by low adduct levels [3]. Still, carcinogenicity of the pyridyloxobutylation pathway has been also associated with the presence of another type of damage. This adduct type, that accounts for at least 50% of the DNA binding obtained through this pathway, is the most prevalent adduct from the pyridyloxobutylation pathway and remained uncharacterized until recently. It was therefore only qualified according to its ability to release 4-hydroxy-1-(3-pyridyloxobutyl)-1-butanone (HPB) (Fig. 1) upon acid or neutral thermal hydrolysis. Recently, several HPB-releasing adducts have been obtained and identified in DNA and/or deoxyguanosine treated with NNKOAc in vitro: N7-pyridyloxobutylguanine (N7-pobG) [12] and O2-pyridyloxobutylcytosine (O2-pobC) [13], the former being by far the most abundant [14]. Other adducts have also been characterized that do not release HPB. They derive from the pyridyloxobutylation of the N2 position of guanine [12] and more frequently of the O2 position of thymine [13]. None of these newly characterized adduct (HPB-releasing or not) have yet been demonstrated to occur in animals or cells, probably because of their low frequency of occurrence and/or their instability, which makes them difficult to observe [12], [14].

The comet assay is a commonly used, sensitive method to investigate DNA damage in individual cells [15]. It has already been used to detect NNK-associated damage in the metabolically competent lymphoblastoid cell line MCL-5 [16] and in white blood cells where NNK seems to act synergistically with UVA to generate DNA damage [17]. In both studies, the type of damage detected could not be identified. In fact, this technique completely lacks specificity in that it can determine only one parameter: the average break frequency. On the contrary, a technique like ligation-mediated PCR (LMPCR), which is commonly used in DNA damage studies, gives several information concerning the breaks: the exact genomic base position along with some chemical information such as the existence of a 5′ phosphate termination. Nevertheless, the comet assay is extremely sensitive, detecting breaks as infrequent as 1 break/108 bases [18] versus 1 break/5 × 104 bases for LMPCR [19] and can therefore be useful to detect DNA damage that is infrequent. In fact, specificity must be sacrificed for sensitivity in cases like NNK-associated damage where damage frequency can be limited by factors such as the ability of the cell to biotransform the exogenous pre-mutagen into intracellular DNA-damaging species. Moreover, some specificity can be imparted to the assay by the following treatments: (1) using analogs that produce a limited variety of DNA lesions such as NDMAOAc or NNKOAc; (2) using a variety of cells with different activation abilities: U937, a histiocytic lymphoma cell line that bioactivates NNK [20], NCI-H23, a lung adenocarcinoma cell line originally obtained from a smoker and normal lymphocytes; (3) generating the single-strand breaks from DNA using chemical treatment (comet assay at different pH) and/or enzymes specific for cleaving particular classes of DNA lesions; (4) using chemical treatments (different lysis conditions) that convert a specific class of DNA lesions into enzyme-sensitive lesions. We used all four specificity enhancement methods to distinguish various types of NNK-associated damage that can be specifically studied using the comet assay.

Section snippets

Caution

All work involving NNK and its derivatives should be performed with protective clothing and in a well-ventilated fume hood.

Cell culture

U937 (human histiocytic lymphoma CRL-1593.2) and NCI-H23 (human lung adenocarcinoma CRL-5800) cell lines were obtained from the ATCC. Both were grown in RPMI 1640 with 2 mM l-glutamine modified to contain 10 mM HEPES, 1 mM sodium pyruvate, 4.5 g/L glucose, 1.5 g/L bicarbonate and supplemented with 10% FBS (v/v) at 37 °C in humidified atmosphere of 5% CO2. U937 cells were

Results

NNK-associated damage has been characterized by others (Table 1). We report here that the comet assay, when used with different strategies to increase its specificity, can be used to detect several types of damage quite specifically with the great sensitivity permitted by this technique.

Discussion

The results of our present study show that the comet assay is sufficiently sensitive and specific to measure NNK-related DNA damage. In peculiar, it permits an investigation of potentially important damage of the pyridyloxobutylation pathway which is far less documented than that of the methylation pathway. A major damage of this pathway correspond to N7-pobG, an HPB-releasing adduct, that had never been shown in cells to this day. Our result indicate that this adduct could exist in its fapy

Acknowledgements

The authors are grateful to Dr. Jean-François Cloutier for the synthesis of NNKOAc, Dr. Serge Boiteux for kindly supplying the endonuclease III and the formamidopyrimidine glycosylase, and Dr. R. Stephen Lloyd for the T4 endonuclease V. The authors sincerely thank Dr. Gerald P. Holmquist for important discussion and carefully editing the manuscript. The research carried out in the laboratory of R.D. was funded by the Institute of Genetics of the CIHR and the Canada Research Chairs Program. R.D.

References (40)

  • M.H. Green et al.

    Protective effect of deoxyribonucleosides on UV-irradiated human peripheral blood T-lymphocytes: possibilities for the selective killing of either cycling or non-cycling cells

    Mutat. Res.

    (1996)
  • J. Laval et al.

    Physiological properties and repair of apurinic/apyrimidinic sites and imidazole ring-opened guanines in DNA

    Mutat. Res.

    (1990)
  • S.S. Hecht

    Tobacco smoke carcinogens and lung cancer

    J. Natl. Cancer Inst.

    (1999)
  • D. Hoffmann et al.

    The biological significance of tobacco-specific N-nitrosamines: smoking and adenocarcinoma of the lung

    Crit. Rev. Toxicol.

    (1996)
  • S.S. Devesa et al.

    Changing patterns of lung cancer incidence by histological type

    Cancer Epidemiol. Biomarkers Prev.

    (1991)
  • S.S. Hecht

    Biochemistry, biology, and carcinogenicity of tobacco-specific N-nitrosamines

    Chem. Res. Toxicol.

    (1998)
  • G.B. Smith et al.

    Biotransformation of the tobacco-specific carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) in freshly isolated human lung cells

    Carcinogenesis

    (1999)
  • G.B. Smith et al.

    Biotransformation of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) in peripheral human lung microsomes

    Drug Metab. Dispos.

    (2003)
  • D.H. Phillips

    Smoking-related DNA and protein adducts in human tissues

    Carcinogenesis

    (2002)
  • J. Haglund et al.

    Evidence for phosphate adducts in DNA from mice treated with 4-(N-Methyl-N-nitrosamino)-1-(3-pyridyl)-1-butanone (NNK)

    Chem. Res. Toxicol.

    (2002)
  • Cited by (25)

    • Comet assay and hormesis

      2024, Environmental Pollution
    • Assessment of RTG-W1, RTL-W1, and PLHC-1 fish cell lines for genotoxicity testing of environmental pollutants by means of a Fpg-modified comet assay

      2012, Toxicology in Vitro
      Citation Excerpt :

      The present results confirm the sensitivity of Fpg towards N7-guanine alkylation as previously shown (Smith et al., 2006; Speit et al., 2004). As described by Lacoste et al. (2006), N7-methyl guanine adducts unstable in alkaline condition, can lead to ring opened N7 methyl fapy guanine during the lysis step (pH 10) and becomes subsequently a Fpg-sensitive lesion. The sensitivity of the Fpg toward N7-purine adducts is extremely relevant in a context of accurate detection of environmental genotoxicity since alkylating compounds are thought to be among the most potent and abundant genotoxic contaminants in aquatic ecosystems (Kuehl et al., 1994).

    • Genotoxic evaluation of the non-halogenated disinfection by-products nitrosodimethylamine and nitrosodiethylamine

      2011, Journal of Hazardous Materials
      Citation Excerpt :

      Besides detecting single and double DNA strand breaks, this technique can also detect incomplete excision repair and alkali-labile sites. It must be emphasized that due to its high sensitivity it is able to detect DNA damage in frequencies of less than 1 damaged base in 107 bases [18]. In addition, the comet assay also allows the analysis of oxidative DNA damage, when the bacterial repair enzymes formamidopyrimidine DNA glycosilase (FPG) and endonuclease III (endoIII) are used.

    • DNA damage induction by two halogenated acetaldehydes, byproducts of water disinfection

      2010, Water Research
      Citation Excerpt :

      One is the single-cell gel electrophoresis (SCGE) assay or comet assay, which is a rapid, relatively simple and sensitive technique for measuring primary DNA damage (Fairbairn et al., 1995). The comet assay is able to detect DNA damage frequencies less than 1 in 107 bases (Lacoste et al., 2006). In addition to DNA single and double-strand breaks, the comet assay is also able to detect incomplete excision repair and alkali-labile sites (Collins, 2004).

    • Chapter 5 Molecular Mechanisms of 4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanone-Induced Lung Carcinogenesis

      2009, Advances in Molecular Toxicology
      Citation Excerpt :

      Additional studies are required to explain these discrepant results. Indirect evidence for the formation of formamidopyrimidine (fapy) adducts in DNA from NNKOAc‐treated human cells was obtained using the comet assay [98]. The contribution of these adducts to the overall damage observed in pyridyloxobutyl DNA is unknown since the method of detection assumed that pyridyloxobutyl fapy adducts equally good substrates for formamidopyrimidine glycosylase as methyl fapy adducts.

    View all citing articles on Scopus
    View full text