Next Article in Journal
The ABCC6 Transporter: A New Player in Biomineralization
Next Article in Special Issue
Induced Pluripotent Stem Cells: Advances in the Quest for Genetic Stability during Reprogramming Process
Previous Article in Journal
Time Course of the Phenotype of Blood and Bone Marrow Monocytes and Macrophages in the Lung after Cigarette Smoke Exposure In Vivo
Previous Article in Special Issue
The Role of PALB2 in the DNA Damage Response and Cancer Predisposition
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Mechanisms of Acetaldehyde-Mediated Carcinogenesis in Squamous Epithelium

1
Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
2
Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
3
Research Center for Environmental Quality Management, Kyoto University, Otsu 520-0811, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2017, 18(9), 1943; https://doi.org/10.3390/ijms18091943
Submission received: 28 July 2017 / Revised: 29 August 2017 / Accepted: 7 September 2017 / Published: 10 September 2017
(This article belongs to the Special Issue DNA Injury and Repair Systems)

Abstract

:
Acetaldehyde is a highly reactive compound that causes various forms of damage to DNA, including DNA adducts, single- and/or double-strand breaks (DSBs), point mutations, sister chromatid exchanges (SCEs), and DNA–DNA cross-links. Among these, DNA adducts such as N2-ethylidene-2′-deoxyguanosine, N2-ethyl-2′-deoxyguanosine, N2-propano-2′-deoxyguanosine, and N2-etheno-2′-deoxyguanosine are central to acetaldehyde-mediated DNA damage because they are associated with the induction of DNA mutations, DNA–DNA cross-links, DSBs, and SCEs. Acetaldehyde is produced endogenously by alcohol metabolism and is catalyzed by aldehyde dehydrogenase 2 (ALDH2). Alcohol consumption increases blood and salivary acetaldehyde levels, especially in individuals with ALDH2 polymorphisms, which are highly associated with the risk of squamous cell carcinomas in the upper aerodigestive tract. Based on extensive epidemiological evidence, the International Agency for Research on Cancer defined acetaldehyde associated with the consumption of alcoholic beverages as a “group 1 carcinogen” (definite carcinogen) for the esophagus and/or head and neck. In this article, we review recent advances from studies of acetaldehyde-mediated carcinogenesis in the squamous epithelium, focusing especially on acetaldehyde-mediated DNA adducts. We also give attention to research on acetaldehyde-mediated DNA repair pathways such as the Fanconi anemia pathway and refer to our studies on the prevention of acetaldehyde-mediated DNA damage.

Graphical Abstract

1. Acetaldehyde, Acetaldehyde Metabolism, and Risk of Cancers

Acetaldehyde, a low molecular weight organic aldehyde with the formula CH3CHO, is a highly reactive compound that causes DNA damage [1,2]. It is found in food and drinks such as yogurt, ripe fruits, cheese, coffee, and alcoholic beverages [3,4], and in tobacco smoke [5]. In addition, acetaldehyde can be produced by microorganisms such as yeasts and bacteria in the human oral cavity [6,7,8]. Thus, acetaldehyde can be ingested orally in a variety of ways. In particular, alcoholic beverages such as Calvados and other spirits contain high quantities of “free” acetaldehyde (e.g., Calvados: 1781 ± 861 μM), and frequent consumption of these beverages is associated with an increased risk of esophageal squamous cell carcinoma (ESCC) [4,9], although “free” acetaldehyde present in alcoholic beverages appears to cause only a short time (1–2 min) direct exposure to the organs [10].
More importantly, acetaldehyde is also generated endogenously by alcohol metabolism. Ingested alcohol is absorbed from the upper gastrointestinal tract and transported to the liver, where it is mainly metabolized into acetaldehyde by alcohol dehydrogenase 1B (ADH1B), and then detoxified to acetic acid by aldehyde dehydrogenase 2 (ALDH2) (Figure 1) [11,12]. Genetic polymorphisms in ADH1B and/or ALDH2 can result in different enzymatic activities that have a major impact on the risk of ESCC as well as head and neck squamous cell carcinoma (HNSCC) [13,14,15,16].
ADH1B has two alleles, ADH1B*1 (less active ADH1B) and ADH1B*2 (active ADH1B, Arg47His). Therefore, ADH1B is divided into three genotypes; ADH1B*1/*1, less active slow metabolizing ADH1B, and ADH1B*1/*2 and ADH1B*2/*2, active ADH1B [17]. Since alcohol metabolism is slow in individuals homozygous for ADH1B*1/*1, acetaldehyde remains in the body for a long time. Meta-analysis has shown that individuals with ADH1B*1/*1 have a 2.77- and 2.35-fold increased risk of ESCC [18] and HNSCC [19], respectively, compared with carriers of the ADH1B*2 allele (ADH1B*1/*2 and ADH1B*2/*2).
ALDH2 has two alleles, ALDH2*1 (active ALDH2) and ALDH2*2 (inactive ALDH2, Glu504Lys). As ALDH2 is a tetrameric enzyme and ALDH2*2 acts in a dominant negative manner, the phenotypic loss of ALDH2 activity is found in both heterozygous (ALDH2*1/*2) and homozygous (ALDH2*2/*2) genotypes [20,21]. Subsequently, ALDH2 genotypes are classified as follows: ALDH2*1/*1, active (100% activity) ALDH2; ALDH2*1/*2, inactive (< 10% activity) ALDH2; and ALDH2*2/*2, inactive (0% activity) ALDH2 [22]. Carriers of the ALDH2*2 allele (ALDH2*1/*2 and ALDH2*2/*2) account for approximately 40% of East Asian populations [23,24,25], whereas these genotypes are quite rare in Caucasoid or Negroid populations [26]. Meta-analysis has shown that individuals with ALDH2*1/*2 have a 7.12- and 1.83-fold increased risk of ESCC [14] and HNSCC [27], respectively, compared with carriers of ALDH2*1/*1. Moreover, alcoholics with the ALDH2*1/*2 genotype have a 13.5- and 18.52-fold increased risk of ESCC and HNSCC, respectively, compared with ALDH2*1/*1 genotypes [15].
Thus, extensive epidemiological evidence suggests that acetaldehyde is deeply involved in the carcinogenesis of the squamous epithelium of the esophagus, and head and neck. In addition, the International Agency for Research on Cancer has defined acetaldehyde associated with the consumption of alcoholic beverages as a “group 1 carcinogen” (definite carcinogen) for the esophagus and/or head and neck [28].

2. Field Cancerization in the Esophagus, and Head and Neck

In some patients, ESCC occurs synchronously and/or metachronously in conjunction with HNSCC (Figure 2A) [12,29]. In such patients, widespread epithelial oncogenic alterations are frequently observed in the esophagus and can be visible as multiple Lugol-voiding lesions (LVLs) by Lugol chromoendoscopy (Figure 2B) [30,31]. Thus, multiple occurrences of neoplastic changes in the upper aerodigestive tract have been explained by the phenomenon of “field cancerization” [32]. We reported previously that the ALDH2*2 allele is the strongest contributing factor (OR: 17.6) for the development of multiple LVLs [29]. Our recent prospective cohort study also revealed that the severity of LVLs is associated with the amount of average alcohol consumption, and individuals with multiple LVLs in their esophagus are especially at high risk for metachronous multiple ESCC and HNSCC [33]. Thus, alcohol consumption in individuals with the ALDH2*2 allele is proven to be associated with the development of field cancerization in the esophagus, and head and neck.

3. Blood and Salivary Acetaldehyde Level after Alcohol Intake

Alcohol consumption increases acetaldehyde concentrations in the blood, saliva, and breath [29,34,35]. In particular, acetaldehyde concentration reaches a very high level in saliva compared with blood [6]. When ALDH2*1/*1 or ALDH2*1/*2 carriers drink 0.6 g ethanol/kg body weight, salivary acetaldehyde concentrations immediately reach 24 to 53 μM in ALDH2*1/*1 carriers and 37 to 76 μM in ALDH2*1/*2 carriers, respectively [36]. The reason for the high acetaldehyde concentrations in saliva is considered to be associated with the formation of acetaldehyde from ethanol via microbial [6] and/or mucosal ADH [37]. Moreover, secretion from salivary glands also influences acetaldehyde concentration in saliva. Indeed, alcohol drinking (0.5 g ethanol/kg body weight) increases acetaldehyde concentrations in parotid duct saliva on ALDH2*1/*2 carriers, while it does not affect those on ALDH2*1/*1 carriers [38]. Furthermore, breath acetaldehyde is also thought to dissolve into saliva. The acetaldehyde concentrations in the oral cavity thus produced are equivalent to the concentration that can induce DNA damage in vitro [6,38]. Therefore, alcohol consumption in ALDH2*1/*2 carriers could promote the direct contact of high acetaldehyde-containing saliva to the surface of the oropharynx, hypopharynx, and esophagus and has the potential to induce DNA damage in the squamous epithelium. Taken together, sustained high acetaldehyde-containing saliva is considered to play an important role in the carcinogenesis of upper digestive tract cancers and it could be involved in “field cancerization.”

4. Acetaldehyde Reacts with DNA to Form DNA Adducts

Acetaldehyde reacts directly with the exocyclic amino group of deoxyguanosine (dG) to form DNA adducts such as N2-ethylidene-2′-deoxyguanosine (N2-ethylidene-dG) [39], N2-ethyl-2′-deoxyguanosine (N2-Et-dG) [40,41], and α-S- and α-R-methyl-γ-hydroxy-1, N2-propano-2′-deoxyguanosine (CrPdG) (Figure 3) [39,42].
N2-ethylidene-dG is generated by a single molecule of acetaldehyde and is the most abundant DNA adduct derived from acetaldehyde [43]. N2-ethylidene-dG is unstable at the nucleoside level and is therefore difficult to measure [39]. N2-ethylidene-dG can be stabilized by the chemical reduction of the Schiff base to the stable product, N2-Et-dG. As endogenous N2-Et-dG is extremely low, the level of N2-Et-dG that is converted from N2-ethylidene-dG by chemical reduction (e.g., NaBH3CN) indicates the endogenous N2-ethylidene-dG level [44]. Thus, N2-ethylidene-dG is used for analysis of acetaldehyde-mediated DNA damage [43,45,46] as a biomarker for acetaldehyde-specific DNA damage [47]. Indeed, alcohol consumption increases oral N2-ethylidene-dG levels [48,49]. Furthermore, blood N2-ethylidene-dG levels are definitely increased by alcohol consumption [50] and/or tobacco smoking [51]. Additionally, blood N2-ethylidene-dG levels in alcoholics with the ALDH2*2 allele are higher than those with the ALDH2*1/*1 allele [46]. Importantly, alcohol consumption increases the esophageal N2-ethylidene-dG levels in Aldh2-knockout mice to a higher level than that of wild-type mice [47,52]. This evidence indicates that drinking alcohol definitely increases acetaldehyde exposure to the esophageal tissues in individuals with the ALDH2*2 allele.
CrPdG is generated by the reaction of two molecules of acetaldehyde with DNA [53] and exists in a ring-opened or ring-closed form [54,55]. Here, two molecules of acetaldehyde are converted into crotonaldehyde and then react with DNA to form CrPdG [56]. The levels of CrPdG are also related to the amount of acetaldehyde produced [57].
An ethenobase adduct, 1,N2-etheno-2′-deoxyguanosine (NεG), is generated in human cells treated with acetaldehyde [53]. NεG is a product from 2′-deoxyguanosine and α,β-unsaturated aldehydes that can be formed during lipid peroxidation mediated by acetaldehyde (Figure 3) [53,58]. As acetaldehyde induces reactive oxygen species (ROS) that leads to lipid peroxidation [59], generation of NεG can be triggered by acetaldehyde, ROS, or both.

5. DNA Adducts Induce Severe DNA Damage

N2-Et-dG blocks DNA synthesis and induces DNA mutations [60,61,62,63]. Moreover, N2-Et-dG inhibits translesion DNA synthesis (TLS), which leads to a majority of frameshift deletions and a minority of G:C > T:A transversions in human cells [62]. N2-Et-dG can rotate around the exocyclic nitrogen and the alpha carbon of acetaldehyde because it has a single bond, whereas N2-ethylidene-dG has a double bond, which makes it more hydrophobic than N2-Et-dG. These differences may result in significantly different mutagenic potential between N2-Et-dG and N2-ethylidene-dG [2].
CrPdG induces DNA interstrand [64] and intrastrand cross-links [65]. The ring-opened form of CrPdG can react with dG on the opposite strand of the DNA to form DNA interstrand cross-links [66]. A similar mechanism has been suggested for the formation of DNA intrastrand cross-links [2]. Whereas the ring-closed form of CrPdG would prevent Watson–Crick base pairing with cytosine in the anti conformation, Hoogsteen base pairing with cytosine would be possible in the syn conformation [55]. CrPdG-mediated disruption of the DNA replication process is thought to cause DNA damage [55,67,68,69].
NεG inhibits a replicative polymerase δ in complex with proliferating cell nuclear antigen (PCNA) while translesion polymerases η, ι, and κ can bypass the lesion with varying mutagenic consequences [70,71,72]. In cells, replication of a plasmid containing a site-specific NεG induces base-pair mutations at the NεG site as well as deletions, rearrangements, double mutants, and base-pair substitutions near the NεG site [73]. These mutations near the NεG site could be triggered by error-prone processing of DNA double-strand breaks (DSBs) resulting from a replication fork collapse caused by NεG [2]. Certainly, acetaldehyde blocks DNA replication and increases the level of phosphorylated histone H2AX (γ-H2AX), a DSB marker, in cells [74].
Acetaldehyde exposure of human cells increases rates of sister chromatid exchange (SCE) [75]. SCE is thought to result from replication-blocking DNA lesions [76]. Although CrPdGs, NεG, and interstrand cross-links are shown to inhibit replication, the adducts or cross-links that relate to the formation of SCEs have not been elucidated.

6. Carcinogenic Effects of Acetaldehyde

To elaborate on details mentioned previously in part, acetaldehyde causes DNA adducts [39,40,41,42], DNA single-strand breaks, DSBs [77], point mutations [69], SCEs [78,79,80], DNA–DNA cross-links [81], micronuclei [82], and gross chromosomal aberrations [65,80]. Accumulations of these genetic abnormalities are considered to proceed cancer development. Exposure of acetaldehyde directly induces mutations, most frequently G:C > A:T transitions in the TP53 gene [83]. This transition pattern is consistent with that found in a study of the HPRT reporter gene [69]. In addition, G:C > T:A transversions are the most frequent miscoding events induced by CrPdG, followed by G:C > C:G and G:C > A:T mutations [67,68,69]. This spectrum of mutations corresponds with the gene variation pattern observed in ESCC [84,85] and HNSCC [86]. Furthermore, inhalation of acetaldehyde causes nasal and respiratory squamous cell carcinoma in rats and hamsters [87,88]. These results indicate that acetaldehyde has direct carcinogenic effects in animals.

7. Repair Pathways of Acetaldehyde-Mediated DNA Damage

Recent research has revealed that cells coordinate multiple processes, such as the Fanconi anemia (FA) pathway, nucleotide excision repair (NER), homologous recombination (HR), TLS, base excision repair (BER), fork protection complex, and ATR-dependent cell cycle checkpoint activation, to prevent and repair acetaldehyde-mediated DNA damage [89].
The specific repair processes for N2-ethylidene-dG and N2-Et-dG remain unknown. The efforts to identify the repair mechanism for N2-Et-dG are reported to be unsuccessful [2,90].
The most plausible repair pathway of CrPdG is NER [91]. CrPdG generates interstrand cross-links [64], which can be repaired by the FA pathway [2]. This pathway is composed of at least 19 genes (FANCA, B, C, D1, D2, E–G, I, J, L–T) and the deficiency of these genes can cause FA [92]. FANCA, B, C, E–G, L, and M form a core complex at the site of interstrand cross-links and then promote ubiquitination of the FANCD2–FANCI complex. This ubiquitination leads to the activation of downstream effector proteins, FANCD1, O, P, and Q. They promote the nucleolytic processing of interstrand cross-links, followed by DNA repair via HR [93,94,95,96,97]. Indeed, the FA–BRCA network is activated when cells are treated with ethanol or aldehyde [98,99]. Cells derived from an FA patient are hypersensitive to acetaldehyde exposure [99,100]. Cells deficient for FANCG, FANCQ, or HR protein Rad51D also show many chromosomal aberrations in response to acetaldehyde, while cells deficient for BER and nonhomologous end-joining show subtle increases in chromosome aberration [101,102]. In vivo, when mice with disrupted Aldh2 locus (Aldh2+/− or Aldh2−/−) and Fancd2 heterozygosity (Fancd2+/−) are crossed and then challenged with ethanol exposure, the numbers of double-knockout offspring (Aldh2−/−, Fancd2−/−) are significantly reduced [103]. Treatment with ethanol in adult double-knockout mice (Aldh2−/−, Fancd2−/−) results in dramatic reductions of bone marrow cells. Moreover, these mice develop leukemia, even without ethanol administration [103]. These results indicate that Fancd2 plays an important role in the protection from acetaldehyde-induced genotoxicity.
Acetaldehyde-mediated DSB is repaired by HR [74]. Acetaldehyde accumulates γ-H2AX, which colocalizes with foci of the HR protein Rad51 in cells [74]. Moreover, recombination-defective cells are hypersensitive to acetaldehyde [74].

8. Prevention of Acetaldehyde-Mediated DNA Damage

Acetaldehyde-mediated DNA damage is influenced by ALDH2 expression level [52]. ALDH2 is known to express in various tissues including the liver, kidney, muscle, and heart [104]. Recently, we found that alcohol consumption in mice promoted ALDH2 protein production in esophageal epithelium [52]. In vitro experiments revealed that ALDH2 is induced by acetaldehyde exposure in esophageal keratinocytes. ALDH2 knockdown resulted in an increase of susceptibility to acetaldehyde. Conversely, ALDH2 overexpression prevented acetaldehyde-mediated DNA damage in esophageal keratinocytes, although overexpression of mutant ALDH2 (ALDH2*2) offered no protection. Thus, enhancement of ALDH2 expression level may prevent acetaldehyde-mediated DNA damage.

9. Conclusions

Previous studies have provided substantial evidence that acetaldehyde induces various forms of DNA damage leading to cancer development (Figure 4). DNA adduct formation might be the key to acetaldehyde-mediated DNA damage; however, the role of DNA adducts in carcinogenesis has not been completely elucidated. Further studies are necessary to reveal the complete mechanisms of acetaldehyde-mediated cancer development.

Acknowledgments

This work was supported by a Grant-in-Aid for Scientific Research 16K09281 (Shinya Ohashi), the Takeda Science Foundation (Shinya Ohashi), and for practical research for innovative cancer control from the Japan Agency for Medical Research and Development, AMED (Manabu Muto). None of the funding sources contributed to the writing of the manuscript.

Conflicts of Interest

The authors declare that they have no competing interests.

Abbreviations

ADH1BAlcohol dehydrogenase 1B
ALDH2Aldehyde dehydrogenase 2
BERBase excision repair
CrPdGα-S- and α-R-methyl-γ-hydroxy-1, N2-propano-2′-deoxyguanosine
dGDeoxyguanosine
DSBDouble-strand break
ESCCEsophageal squamous cell carcinoma
FAFanconi anemia
HNSCCHead and neck squamous cell carcinoma
HRHomologous recombination
LVLLugol-voiding lesion
NERNucleotide excision repair
N2-Et-dGN2-ethyl-2′-deoxyguanosine
N2-ethylidene-dGN2-ethylidene-2′-deoxyguanosine
NεG1,N2-etheno-2′-deoxyguanosine
PCNAProliferating cell nuclear antigen
ROSReactive oxygen species
SCESister chromatid exchange
TLSTranslesion DNA synthesis
γ-H2AXPhosphorylated histone H2AX

References

  1. Seitz, H.K.; Stickel, F. Molecular mechanisms of alcohol-mediated carcinogenesis. Nat. Rev. Cancer 2007, 7, 599–612. [Google Scholar] [CrossRef] [PubMed]
  2. Brooks, P.J.; Zakhari, S. Acetaldehyde and the genome: Beyond nuclear DNA adducts and carcinogenesis. Environ. Mol. Mutagen. 2014, 55, 77–91. [Google Scholar] [CrossRef] [PubMed]
  3. Uebelacker, M.; Lachenmeier, D.W. Quantitative determination of acetaldehyde in foods using automated digestion with simulated gastric fluid followed by headspace gas chromatography. J. Autom. Methods Manag. Chem. 2011, 2011, 907317. [Google Scholar] [CrossRef] [PubMed]
  4. Launoy, G.; Milan, C.; Day, N.E.; Pienkowski, M.P.; Gignoux, M.; Faivre, J. Diet and squamous-cell cancer of the oesophagus: A french multicentre case-control study. Int. J. Cancer 1998, 76, 7–12. [Google Scholar] [CrossRef]
  5. Salaspuro, V.J.; Hietala, J.M.; Marvola, M.L.; Salaspuro, M.P. Eliminating carcinogenic acetaldehyde by cysteine from saliva during smoking. Cancer Epidemiol. Biomark. Prev. 2006, 15, 146–149. [Google Scholar] [CrossRef] [PubMed]
  6. Homann, N.; Jousimies-Somer, H.; Jokelainen, K.; Heine, R.; Salaspuro, M. High acetaldehyde levels in saliva after ethanol consumption: Methodological aspects and pathogenetic implications. Carcinogenesis 1997, 18, 1739–1743. [Google Scholar] [CrossRef] [PubMed]
  7. Salaspuro, M.P. Acetaldehyde, microbes, and cancer of the digestive tract. Crit. Rev. Clin. Lab. Sci. 2003, 40, 183–208. [Google Scholar] [CrossRef] [PubMed]
  8. Muto, M.; Hitomi, Y.; Ohtsu, A.; Shimada, H.; Kashiwase, Y.; Sasaki, H.; Yoshida, S.; Esumi, H. Acetaldehyde production by non-pathogenic neisseria in human oral microflora: Implications for carcinogenesis in upper aerodigestive tract. Int. J. Cancer 2000, 88, 342–350. [Google Scholar] [CrossRef]
  9. Linderborg, K.; Joly, J.P.; Visapaa, J.P.; Salaspuro, M. Potential mechanism for calvados-related oesophageal cancer. Food Chem. Toxicol. 2008, 46, 476–479. [Google Scholar] [CrossRef] [PubMed]
  10. Linderborg, K.; Salaspuro, M.; Vakevainen, S. A single sip of a strong alcoholic beverage causes exposure to carcinogenic concentrations of acetaldehyde in the oral cavity. Food Chem. Toxicol. 2011, 49, 2103–2106. [Google Scholar] [CrossRef] [PubMed]
  11. Brooks, P.J.; Enoch, M.A.; Goldman, D.; Li, T.K.; Yokoyama, A. The alcohol flushing response: An unrecognized risk factor for esophageal cancer from alcohol consumption. PLoS Med. 2009, 6, e50. [Google Scholar] [CrossRef] [PubMed]
  12. Ohashi, S.; Miyamoto, S.; Kikuchi, O.; Goto, T.; Amanuma, Y.; Muto, M. Recent advances from basic and clinical studies of esophageal squamous cell carcinoma. Gastroenterology 2015, 149, 1700–1715. [Google Scholar] [CrossRef] [PubMed]
  13. Matsuo, K.; Hamajima, N.; Shinoda, M.; Hatooka, S.; Inoue, M.; Takezaki, T.; Tajima, K. Gene-environment interaction between an aldehyde dehydrogenase-2 (ALDH2) polymorphism and alcohol consumption for the risk of esophageal cancer. Carcinogenesis 2001, 22, 913–916. [Google Scholar] [CrossRef] [PubMed]
  14. Yang, S.J. Relationship between genetic polymorphisms of ALDH2 and ADH1B and esophageal cancer risk: A meta-analysis. World J. Gastroenterol. 2010, 16, 4210. [Google Scholar] [CrossRef] [PubMed]
  15. Yokoyama, A.; Muramatsu, T.; Omori, T.; Yokoyama, T.; Matsushita, S.; Higuchi, S.; Maruyama, K.; Ishii, H. Alcohol and aldehyde dehydrogenase gene polymorphisms and oropharyngolaryngeal, esophageal and stomach cancers in japanese alcoholics. Carcinogenesis 2001, 22, 433–439. [Google Scholar] [CrossRef] [PubMed]
  16. Lachenmeier, D.W.; Salaspuro, M. ALDH2-deficiency as genetic epidemiologic and biochemical model for the carcinogenicity of acetaldehyde. Regul. Toxicol. Pharmacol. 2017, 86, 128–136. [Google Scholar] [CrossRef] [PubMed]
  17. Neumark, Y.D.; Friedlander, Y.; Durst, R.; Leitersdorf, E.; Jaffe, D.; Ramchandani, V.A.; O’Connor, S.; Carr, L.G.; Li, T.K. Alcohol dehydrogenase polymorphisms influence alcohol-elimination rates in a male jewish population. Alcohol Clin. Exp. Res. 2004, 28, 10–14. [Google Scholar] [CrossRef] [PubMed]
  18. Zhang, L.; Jiang, Y.; Wu, Q.; Li, Q.; Chen, D.; Xu, L.; Zhang, C.; Zhang, M.; Ye, L. Gene—Environment interactions on the risk of esophageal cancer among Asian populations with the G48A polymorphism in the alcohol dehydrogenase-2 gene: A meta-analysis. Tumour Biol. 2014, 35, 4705–4717. [Google Scholar] [CrossRef] [PubMed]
  19. Zhang, Y.; Gu, N.; Miao, L.; Yuan, H.; Wang, R.; Jiang, H. Alcohol dehydrogenase-1B Arg47His polymorphism is associated with head and neck cancer risk in Asian: A meta-analysis. Tumour Biol. 2015, 36, 1023–1027. [Google Scholar] [CrossRef] [PubMed]
  20. Enomoto, N.; Takase, S.; Yasuhara, M.; Takada, A. Acetaldehyde metabolism in different aldehyde dehydrogenase-2 genotypes. Alcohol Clin. Exp. Res. 1991, 15, 141–144. [Google Scholar] [CrossRef] [PubMed]
  21. Hoshi, H.; Hao, W.; Fujita, Y.; Funayama, A.; Miyauchi, Y.; Hashimoto, K.; Miyamoto, K.; Iwasaki, R.; Sato, Y.; Kobayashi, T.; et al. Aldehyde-stress resulting from ALDH2 mutation promotes osteoporosis due to impaired osteoblastogenesis. J. Bone Miner. Res. 2012, 27, 2015–2023. [Google Scholar] [CrossRef] [PubMed]
  22. Yokoyama, A.; Mizukami, T.; Yokoyama, T. Genetic polymorphisms of alcohol dehydrogense-1B and aldehyde dehydrogenase-2, alcohol flushing, mean corpuscular volume, and aerodigestive tract neoplasia in japanese drinkers. Adv. Exp. Med. Biol. 2015, 815, 265–279. [Google Scholar] [PubMed]
  23. Harada, S.; Agarwal, D.P.; Goedde, H.W. Aldehyde dehydrogenase deficiency as cause of facial flushing reaction to alcohol in Japanese. Lancet (Lond. Engl.) 1981, 2, 982. [Google Scholar] [CrossRef]
  24. Yoshida, A.; Huang, I.Y.; Ikawa, M. Molecular abnormality of an inactive aldehyde dehydrogenase variant commonly found in orientals. Proc. Natl. Acad. Sci. USA 1984, 81, 258–261. [Google Scholar] [CrossRef] [PubMed]
  25. Higuchi, S.; Matsushita, S.; Murayama, M.; Takagi, S.; Hayashida, M. Alcohol and aldehyde dehydrogenase polymorphisms and the risk for alcoholism. Am. J. Psychiatry 1995, 152, 1219–1221. [Google Scholar] [PubMed]
  26. Goedde, H.W.; Agarwal, D.P.; Fritze, G.; Meier-Tackmann, D.; Singh, S.; Beckmann, G.; Bhatia, K.; Chen, L.Z.; Fang, B.; Lisker, R.; et al. Distribution of ADH2 and ALDH2 genotypes in different populations. Hum. Genet. 1992, 88, 344–346. [Google Scholar] [CrossRef] [PubMed]
  27. Boccia, S.; Hashibe, M.; Galli, P.; De Feo, E.; Asakage, T.; Hashimoto, T.; Hiraki, A.; Katoh, T.; Nomura, T.; Yokoyama, A.; et al. Aldehyde dehydrogenase 2 and head and neck cancer: A meta-analysis implementing a mendelian randomization approach. Cancer Epidemiol. Biomark. Prev. 2009, 18, 248–254. [Google Scholar] [CrossRef] [PubMed]
  28. Secretan, B.; Straif, K.; Baan, R.; Grosse, Y.; El Ghissassi, F.; Bouvard, V.; Benbrahim-Tallaa, L.; Guha, N.; Freeman, C.; Galichet, L.; et al. A review of human carcinogens—Part E: Tobacco, areca nut, alcohol, coal smoke, and salted fish. Lancet Oncol. 2009, 10, 1033–1034. [Google Scholar] [CrossRef]
  29. Muto, M.; Nakane, M.; Hitomi, Y.; Yoshida, S.; Sasaki, S.; Ohtsu, A.; Yoshida, S.; Ebihara, S.; Esumi, H. Association between aldehyde dehydrogenase gene polymorphisms and the phenomenon of field cancerization in patients with head and neck cancer. Carcinogenesis 2002, 23, 1759–1765. [Google Scholar] [CrossRef] [PubMed]
  30. Mori, M.; Adachi, Y.; Matsushima, T.; Matsuda, H.; Kuwano, H.; Sugimachi, K. Lugol staining pattern and histology of esophageal lesions. Am. J. Gastroenterol. 1993, 88, 701–705. [Google Scholar] [PubMed]
  31. Muto, M.; Hironaka, S.; Nakane, M.; Boku, N.; Ohtsu, A.; Yoshida, S. Association of multiple lugol-voiding lesions with synchronous and metachronous esophageal squamous cell carcinoma in patients with head and neck cancer. Gastrointest. Endosc. 2002, 56, 517–521. [Google Scholar] [CrossRef]
  32. Slaughter, D.P.; Southwick, H.W.; Smejkal, W. Field cancerization in oral stratified squamous epithelium; clinical implications of multicentric origin. Cancer 1953, 6, 963–968. [Google Scholar] [CrossRef]
  33. Katada, C.; Yokoyama, T.; Yano, T.; Kaneko, K.; Oda, I.; Shimizu, Y.; Doyama, H.; Koike, T.; Takizawa, K.; Hirao, M.; et al. Alcohol consumption and multiple dysplastic lesions increase risk of squamous cell carcinoma in the esophagus, head, and neck. Gastroenterology 2016, 151, 860–869. [Google Scholar] [CrossRef] [PubMed]
  34. Yokoyama, A.; Tsutsumi, E.; Imazeki, H.; Suwa, Y.; Nakamura, C.; Yokoyama, T. Polymorphisms of alcohol dehydrogenase-1B and aldehyde dehydrogenase-2 and the blood and salivary ethanol and acetaldehyde concentrations of japanese alcoholic men. Alcohol Clin. Exp. Res. 2010, 34, 1246–1256. [Google Scholar] [CrossRef] [PubMed]
  35. Aoyama, I.; Ohashi, S.; Amanuma, Y.; Hirohashi, K.; Mizumoto, A.; Funakoshi, M.; Tsurumaki, M.; Nakai, Y.; Tanaka, K.; Hanada, M.; et al. Establishment of a quick and highly accurate breath test for ALDH2 genotyping. Clin. Transl. Gastroenterol. 2017, 8, e96. [Google Scholar] [CrossRef] [PubMed]
  36. Yokoyama, A.; Tsutsumi, E.; Imazeki, H.; Suwa, Y.; Nakamura, C.; Mizukami, T.; Yokoyama, T. Salivary acetaldehyde concentration according to alcoholic beverage consumed and aldehyde dehydrogenase-2 genotype. Alcohol Clin. Exp. Res. 2008, 32, 1607–1614. [Google Scholar] [CrossRef] [PubMed]
  37. Dong, Y.J.; Peng, T.K.; Yin, S.J. Expression and activities of class Ⅳ alcohol dehydrogenase and class Ⅲ aldehyde dehydrogenase in human mouth. Alcohol 1996, 13, 257–262. [Google Scholar] [CrossRef]
  38. Vakevainen, S.; Tillonen, J.; Agarwal, D.P.; Srivastava, N.; Salaspuro, M. High salivary acetaldehyde after a moderate dose of alcohol in ALDH2-deficient subjects: Strong evidence for the local carcinogenic action of acetaldehyde. Alcohol Clin. Exp. Res. 2000, 24, 873–877. [Google Scholar] [CrossRef] [PubMed]
  39. Wang, M.; McIntee, E.J.; Cheng, G.; Shi, Y.; Villalta, P.W.; Hecht, S.S. Identification of DNA adducts of acetaldehyde. Chem. Res. Toxicol. 2000, 13, 1149–1157. [Google Scholar] [CrossRef] [PubMed]
  40. Fang, J.L.; Vaca, C.E. Development of a 32P-postlabelling method for the analysis of adducts arising through the reaction of acetaldehyde with 2’-deoxyguanosine-3’-monophosphate and DNA. Carcinogenesis 1995, 16, 2177–2185. [Google Scholar] [CrossRef] [PubMed]
  41. Fang, J.L.; Vaca, C.E. Detection of DNA adducts of acetaldehyde in peripheral white blood cells of alcohol abusers. Carcinogenesis 1997, 18, 627–632. [Google Scholar] [CrossRef] [PubMed]
  42. Hecht, S.S.; McIntee, E.J.; Wang, M. New DNA adducts of crotonaldehyde and acetaldehyde. Toxicology 2001, 166, 31–36. [Google Scholar] [CrossRef]
  43. Matsuda, T.; Matsumoto, A.; Uchida, M.; Kanaly, R.A.; Misaki, K.; Shibutani, S.; Kawamoto, T.; Kitagawa, K.; Nakayama, K.I.; Tomokuni, K.; et al. Increased formation of hepatic N2-ethylidene-2’-deoxyguanosine DNA adducts in aldehyde dehydrogenase 2-knockout mice treated with ethanol. Carcinogenesis 2007, 28, 2363–2366. [Google Scholar] [CrossRef] [PubMed]
  44. Wang, M.; Yu, N.; Chen, L.; Villalta, P.W.; Hochalter, J.B.; Hecht, S.S. Identification of an acetaldehyde adduct in human liver DNA and quantitation as N2-ethyldeoxyguanosine. Chem. Res. Toxicol. 2006, 19, 319–324. [Google Scholar] [CrossRef] [PubMed]
  45. Nagayoshi, H.; Matsumoto, A.; Nishi, R.; Kawamoto, T.; Ichiba, M.; Matsuda, T. Increased formation of gastric N2-ethylidene-2’-deoxyguanosine DNA adducts in aldehyde dehydrogenase-2 knockout mice treated with ethanol. Mutat. Res. 2009, 673, 74–77. [Google Scholar] [CrossRef] [PubMed]
  46. Yukawa, Y.; Muto, M.; Hori, K.; Nagayoshi, H.; Yokoyama, A.; Chiba, T.; Matsuda, T. Combination of ADH1B*2/ALDH2*2 polymorphisms alters acetaldehyde-derived DNA damage in the blood of japanese alcoholics. Cancer Sci. 2012, 103, 1651–1655. [Google Scholar] [CrossRef] [PubMed]
  47. Yukawa, Y.; Ohashi, S.; Amanuma, Y.; Nakai, Y.; Tsurumaki, M.; Kikuchi, O.; Miyamoto, S.; Oyama, T.; Kawamoto, T.; Chiba, T.; et al. Impairment of aldehyde dehydrogenase 2 increases accumulation of acetaldehyde-derived DNA damage in the esophagus after ethanol ingestion. Am. J. Cancer Res. 2014, 4, 279–284. [Google Scholar] [PubMed]
  48. Balbo, S.; Meng, L.; Bliss, R.L.; Jensen, J.A.; Hatsukami, D.K.; Hecht, S.S. Kinetics of DNA adduct formation in the oral cavity after drinking alcohol. Cancer Epidemiol. Biomarkers Prev. 2012, 21, 601–608. [Google Scholar] [CrossRef] [PubMed]
  49. Balbo, S.; Juanes, R.C.; Khariwala, S.; Baker, E.J.; Daunais, J.B.; Grant, K.A. Increased levels of the acetaldehyde-derived DNA adduct N2-ethyldeoxyguanosine in oral mucosa DNA from rhesus monkeys exposed to alcohol. Mutagenesis 2016, 31, 553–558. [Google Scholar] [CrossRef] [PubMed]
  50. Balbo, S.; Hashibe, M.; Gundy, S.; Brennan, P.; Canova, C.; Simonato, L.; Merletti, F.; Richiardi, L.; Agudo, A.; Castellsague, X.; et al. N2-ethyldeoxyguanosine as a potential biomarker for assessing effects of alcohol consumption on DNA. Cancer Epidemiol. Biomarkers Prev. 2008, 17, 3026–3032. [Google Scholar] [CrossRef] [PubMed]
  51. Chen, L.; Wang, M.; Villalta, P.W.; Luo, X.; Feuer, R.; Jensen, J.; Hatsukami, D.K.; Hecht, S.S. Quantitation of an acetaldehyde adduct in human leukocyte DNA and the effect of smoking cessation. Chem. Res Toxicol. 2007, 20, 108–113. [Google Scholar] [CrossRef] [PubMed]
  52. Amanuma, Y.; Ohashi, S.; Itatani, Y.; Tsurumaki, M.; Matsuda, S.; Kikuchi, O.; Nakai, Y.; Miyamoto, S.; Oyama, T.; Kawamoto, T.; et al. Protective role of ALDH2 against acetaldehyde-derived DNA damage in oesophageal squamous epithelium. Sci. Rep. 2015, 5, 14142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Garcia, C.C.; Angeli, J.P.; Freitas, F.P.; Gomes, O.F.; de Oliveira, T.F.; Loureiro, A.P.; di Mascio, P.; Medeiros, M.H. [13C2]-acetaldehyde promotes unequivocal formation of 1,N2-propano-2’-deoxyguanosine in human cells. J. Am. Chem. Soc. 2011, 133, 9140–9143. [Google Scholar] [CrossRef] [PubMed]
  54. Mao, H.; Schnetz-Boutaud, N.C.; Weisenseel, J.P.; Marnett, L.J.; Stone, M.P. Duplex DNA catalyzes the chemical rearrangement of a malondialdehyde deoxyguanosine adduct. Proc. Natl. Acad. Sci. USA 1999, 96, 6615–6620. [Google Scholar] [CrossRef] [PubMed]
  55. Minko, I.G.; Kozekov, I.D.; Harris, T.M.; Rizzo, C.J.; Lloyd, R.S.; Stone, M.P. Chemistry and biology of DNA containing 1,N2-deoxyguanosine adducts of the α,β-unsaturated aldehydes acrolein, crotonaldehyde, and 4-hydroxynonenal. Chem. Res. Toxicol. 2009, 22, 759–778. [Google Scholar] [CrossRef] [PubMed]
  56. Theruvathu, J.A.; Jaruga, P.; Nath, R.G.; Dizdaroglu, M.; Brooks, P.J. Polyamines stimulate the formation of mutagenic 1,N2-propanodeoxyguanosine adducts from acetaldehyde. Nucleic Acids Res. 2005, 33, 3513–3520. [Google Scholar] [CrossRef] [PubMed]
  57. Matsuda, T.; Yabushita, H.; Kanaly, R.A.; Shibutani, S.; Yokoyama, A. Increased DNA damage in ALDH2-deficient alcoholics. Chem. Res. Toxicol. 2006, 19, 1374–1378. [Google Scholar] [CrossRef] [PubMed]
  58. Loureiro, A.P.; di Mascio, P.; Gomes, O.F.; Medeiros, M.H. Trans,trans-2,4-decadienal-induced 1, N2-etheno-2′-deoxyguanosine adduct formation. Chem. Res. Toxicol. 2000, 13, 601–609. [Google Scholar] [CrossRef] [PubMed]
  59. Tanaka, K.; Whelan, K.A.; Chandramouleeswaran, P.M.; Kagawa, S.; Rustgi, S.L.; Noguchi, C.; Guha, M.; Srinivasan, S.; Amanuma, Y.; Ohashi, S.; et al. ALDH2 modulates autophagy flux to regulate acetaldehyde-mediated toxicity thresholds. Am. J. Cancer Res. 2016, 6, 781–796. [Google Scholar] [PubMed]
  60. Matsuda, T.; Terashima, I.; Matsumoto, Y.; Yabushita, H.; Matsui, S.; Shibutani, S. Effective utilization of N2-ethyl-2′-deoxyguanosine triphosphate during DNA synthesis catalyzed by mammalian replicative DNA polymerases. Biochemistry 1999, 38, 929–935. [Google Scholar] [CrossRef] [PubMed]
  61. Terashima, I.; Matsuda, T.; Fang, T.W.; Suzuki, N.; Kobayashi, J.; Kohda, K.; Shibutani, S. Miscoding potential of the N2-ethyl-2′-deoxyguanosine DNA adduct by the exonuclease-free klenow fragment of escherichia coli DNA polymerase i. Biochemistry 2001, 40, 4106–4114. [Google Scholar] [CrossRef] [PubMed]
  62. Upton, D.C.; Wang, X.; Blans, P.; Perrino, F.W.; Fishbein, J.C.; Akman, S.A. Replication of N2-ethyldeoxyguanosine DNA adducts in the human embryonic kidney cell line 293. Chem. Res. Toxicol. 2006, 19, 960–967. [Google Scholar] [CrossRef] [PubMed]
  63. Perrino, F.W.; Blans, P.; Harvey, S.; Gelhaus, S.L.; McGrath, C.; Akman, S.A.; Jenkins, G.S.; LaCourse, W.R.; Fishbein, J.C. The N2-ethylguanine and the O6-ethyl- and O6-methylguanine lesions in DNA: Contrasting responses from the "bypass" DNA polymerase eta and the replicative DNA polymerase α. Chem. Res. Toxicol. 2003, 16, 1616–1623. [Google Scholar] [CrossRef] [PubMed]
  64. Brooks, P.J.; Theruvathu, J.A. DNA adducts from acetaldehyde: Implications for alcohol-related carcinogenesis. Alcohol 2005, 35, 187–193. [Google Scholar] [CrossRef] [PubMed]
  65. Matsuda, T.; Kawanishi, M.; Yagi, T.; Matsui, S.; Takebe, H. Specific tandem GG to TT base substitutions induced by acetaldehyde are due to intra-strand crosslinks between adjacent guanine bases. Nucleic Acids Res. 1998, 26, 1769–1774. [Google Scholar] [CrossRef] [PubMed]
  66. Cho, Y.J.; Wang, H.; Kozekov, I.D.; Kurtz, A.J.; Jacob, J.; Voehler, M.; Smith, J.; Harris, T.M.; Lloyd, R.S.; Rizzo, C.J.; et al. Stereospecific formation of interstrand carbinolamine DNA cross-links by crotonaldehyde- and acetaldehyde-derived α-CH3-γ-OH-1,N2-propano-2’-deoxyguanosine adducts in the 5′-CpG-3′ sequence. Chem. Res. Toxicol. 2006, 19, 195–208. [Google Scholar] [CrossRef] [PubMed]
  67. Fernandes, P.H.; Kanuri, M.; Nechev, L.V.; Harris, T.M.; Lloyd, R.S. Mammalian cell mutagenesis of the DNA adducts of vinyl chloride and crotonaldehyde. Environ. Mol. Mutagen. 2005, 45, 455–459. [Google Scholar] [CrossRef] [PubMed]
  68. Stein, S.; Lao, Y.; Yang, I.Y.; Hecht, S.S.; Moriya, M. Genotoxicity of acetaldehyde- and crotonaldehyde-induced 1, N2-propanodeoxyguanosine DNA adducts in human cells. Mutat. Res. 2006, 608, 1–7. [Google Scholar] [CrossRef] [PubMed]
  69. Noori, P.; Hou, S.M. Mutational spectrum induced by acetaldehyde in the HPRT gene of human t lymphocytes resembles that in the p53 gene of esophageal cancers. Carcinogenesis 2001, 22, 1825–1830. [Google Scholar] [CrossRef] [PubMed]
  70. Choi, J.Y.; Guengerich, F.P. Adduct size limits efficient and error-free bypass across bulky N2-guanine DNA lesions by human DNA polymerase eta. J. Mol. Biol. 2005, 352, 72–90. [Google Scholar] [CrossRef] [PubMed]
  71. Choi, J.Y.; Guengerich, F.P. Kinetic evidence for inefficient and error-prone bypass across bulky N2-guanine DNA adducts by human DNA polymerase iota. J. Biol. Chem. 2006, 281, 12315–12324. [Google Scholar] [CrossRef] [PubMed]
  72. Choi, J.Y.; Angel, K.C.; Guengerich, F.P. Translesion synthesis across bulky N2-alkyl guanine DNA adducts by human DNA polymerase κ. J. Biol Chem. 2006, 281, 21062–21072. [Google Scholar] [CrossRef] [PubMed]
  73. Akasaka, S.; Guengerich, F.P. Mutagenicity of site-specifically located 1,N2-ethenoguanine in chinese hamster ovary cell chromosomal DNA. Chem. Res. Toxicol. 1999, 12, 501–507. [Google Scholar] [CrossRef] [PubMed]
  74. Kotova, N.; Vare, D.; Schultz, N.; Gradecka Meesters, D.; Stepnik, M.; Grawe, J.; Helleday, T.; Jenssen, D. Genotoxicity of alcohol is linked to DNA replication-associated damage and homologous recombination repair. Carcinogenesis 2013, 34, 325–330. [Google Scholar] [CrossRef] [PubMed]
  75. Jansson, T. The frequency of sister chromatid exchanges in human lymphocytes treated with ethanol and acetaldehyde. Hereditas 1982, 97, 301–303. [Google Scholar] [CrossRef] [PubMed]
  76. Wilson, D.M., 3rd; Thompson, L.H. Molecular mechanisms of sister-chromatid exchange. Mutat. Res. 2007, 616, 11–23. [Google Scholar] [CrossRef] [PubMed]
  77. Singh, N.P.; Khan, A. Acetaldehyde: Genotoxicity and cytotoxicity in human lymphocytes. Mutat. Res. 1995, 337, 9–17. [Google Scholar] [CrossRef]
  78. Obe, G.; Jonas, R.; Schmidt, S. Metabolism of ethanol in vitro produces a compound which induces sister-chromatid exchanges in human peripheral lymphocytes in vitro: Acetaldehyde not ethanol is mutagenic. Mutat. Res. 1986, 174, 47–51. [Google Scholar] [CrossRef]
  79. Dellarco, V.L. A mutagenicity assessment of acetaldehyde. Mutat. Res. 1988, 195, 1–20. [Google Scholar] [CrossRef]
  80. Helander, A.; Lindahl-Kiessling, K. Increased frequency of acetaldehyde-induced sister-chromatid exchanges in human lymphocytes treated with an aldehyde dehydrogenase inhibitor. Mutat. Res. 1991, 264, 103–107. [Google Scholar] [CrossRef]
  81. Lambert, B.; Chen, Y.; He, S.M.; Sten, M. DNA cross-links in human leucocytes treated with vinyl acetate and acetaldehyde in vitro. Mutat. Res. 1985, 146, 301–303. [Google Scholar] [CrossRef]
  82. Kayani, M.A.; Parry, J.M. The in vitro genotoxicity of ethanol and acetaldehyde. Toxicol. In Vitro 2010, 24, 56–60. [Google Scholar] [CrossRef] [PubMed]
  83. Paget, V.; Lechevrel, M.; Sichel, F. Acetaldehyde-induced mutational pattern in the tumour suppressor gene tp53 analysed by use of a functional assay, the fasay (functional analysis of separated alleles in yeast). Mutat. Res. 2008, 652, 12–19. [Google Scholar] [CrossRef] [PubMed]
  84. Lin, D.C.; Hao, J.J.; Nagata, Y.; Xu, L.; Shang, L.; Meng, X.; Sato, Y.; Okuno, Y.; Varela, A.M.; Ding, L.W.; et al. Genomic and molecular characterization of esophageal squamous cell carcinoma. Nat. Genet. 2014, 46, 467–473. [Google Scholar] [CrossRef] [PubMed]
  85. Sawada, G.; Niida, A.; Uchi, R.; Hirata, H.; Shimamura, T.; Suzuki, Y.; Shiraishi, Y.; Chiba, K.; Imoto, S.; Takahashi, Y.; et al. Genomic landscape of esophageal squamous cell carcinoma in a Japanese population. Gastroenterology 2016, 150, 1171–1182. [Google Scholar] [CrossRef] [PubMed]
  86. Cancer Genome Atlas, N. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature 2015, 517, 576–582. [Google Scholar]
  87. Woutersen, R.A.; Appelman, L.M.; van Garderen-Hoetmer, A.; Feron, V.J. Inhalation toxicity of acetaldehyde in rats. Ⅲ. Carcinogenicity study. Toxicology 1986, 41, 213–231. [Google Scholar] [CrossRef]
  88. Feron, V.J.; Kruysse, A.; Woutersen, R.A. Respiratory tract tumours in hamsters exposed to acetaldehyde vapour alone or simultaneously to benzo(a)pyrene or diethylnitrosamine. Eur. J. Cancer Clin. Oncol. 1982, 18, 13–31. [Google Scholar] [CrossRef]
  89. Noguchi, C.; Grothusen, G.; Anandarajan, V.; Martinez-Lage Garcia, M.; Terlecky, D.; Corzo, K.; Tanaka, K.; Nakagawa, H.; Noguchi, E. Genetic controls of DNA damage avoidance in response to acetaldehyde in fission yeast. Cell Cycle 2017, 16, 45–58. [Google Scholar] [CrossRef] [PubMed]
  90. Koivisto, P.; Robins, P.; Lindahl, T.; Sedgwick, B. Demethylation of 3-methylthymine in DNA by bacterial and human DNA dioxygenases. J. Biol. Chem. 2004, 279, 40470–40474. [Google Scholar] [CrossRef] [PubMed]
  91. Choudhury, S.; Pan, J.; Amin, S.; Chung, F.L.; Roy, R. Repair kinetics of trans-4-hydroxynonenal-induced cyclic 1,N2-propanodeoxyguanine DNA adducts by human cell nuclear extracts. Biochemistry 2004, 43, 7514–7521. [Google Scholar] [CrossRef] [PubMed]
  92. Dong, H.; Nebert, D.W.; Bruford, E.A.; Thompson, D.C.; Joenje, H.; Vasiliou, V. Update of the human and mouse fanconi anemia genes. Hum. Genomics 2015, 9, 32. [Google Scholar] [CrossRef] [PubMed]
  93. Clauson, C.; Scharer, O.D.; Niedernhofer, L. Advances in understanding the complex mechanisms of DNA interstrand cross-link repair. Cold Spring Harb. Perspect. Biol. 2013, 5, a012732. [Google Scholar] [CrossRef] [PubMed]
  94. Kottemann, M.C.; Smogorzewska, A. Fanconi anaemia and the repair of watson and crick DNA crosslinks. Nature 2013, 493, 356–363. [Google Scholar] [CrossRef] [PubMed]
  95. Moldovan, G.L.; D’Andrea, A.D. How the fanconi anemia pathway guards the genome. Annu. Rev. Genet. 2009, 43, 223–249. [Google Scholar] [CrossRef] [PubMed]
  96. Walden, H.; Deans, A.J. The fanconi anemia DNA repair pathway: Structural and functional insights into a complex disorder. Annu. Rev. Biophys. 2014, 43, 257–278. [Google Scholar] [CrossRef] [PubMed]
  97. Thompson, L.H.; Hinz, J.M. Cellular and molecular consequences of defective fanconi anemia proteins in replication-coupled DNA repair: Mechanistic insights. Mutat. Res. 2009, 668, 54–72. [Google Scholar] [CrossRef] [PubMed]
  98. Abraham, J.; Balbo, S.; Crabb, D.; Brooks, P.J. Alcohol metabolism in human cells causes DNA damage and activates the fanconi anemia-breast cancer susceptibility (FA-BRCA) DNA damage response network. Alcohol Clin. Exp. Res. 2011, 35, 2113–2120. [Google Scholar] [CrossRef] [PubMed]
  99. Marietta, C.; Thompson, L.H.; Lamerdin, J.E.; Brooks, P.J. Acetaldehyde stimulates FANCD2 monoubiquitination, H2AX phosphorylation, and BRCA1 phosphorylation in human cells in vitro: Implications for alcohol-related carcinogenesis. Mutat. Res. 2009, 664, 77–83. [Google Scholar] [CrossRef] [PubMed]
  100. Obe, G.; Natarajan, A.T.; Meyers, M.; Hertog, A.D. Induction of chromosomal aberrations in peripheral lymphocytes of human blood in vitro, and of sces in bone-marrow cells of mice in vivo by ethanol and its metabolite acetaldehyde. Mutat. Res. 1979, 68, 291–294. [Google Scholar] [CrossRef]
  101. Mechilli, M.; Schinoppi, A.; Kobos, K.; Natarajan, A.T.; Palitti, F. DNA repair deficiency and acetaldehyde-induced chromosomal alterations in CHO cells. Mutagenesis 2008, 23, 51–56. [Google Scholar] [CrossRef] [PubMed]
  102. Lorenti Garcia, C.; Mechilli, M.; Proietti De Santis, L.; Schinoppi, A.; Kobos, K.; Palitti, F. Relationship between DNA lesions, DNA repair and chromosomal damage induced by acetaldehyde. Mutat. Res. 2009, 662, 3–9. [Google Scholar] [CrossRef] [PubMed]
  103. Langevin, F.; Crossan, G.P.; Rosado, I.V.; Arends, M.J.; Patel, K.J. FANCD2 counteracts the toxic effects of naturally produced aldehydes in mice. Nature 2011, 475, 53–58. [Google Scholar] [CrossRef] [PubMed]
  104. Stewart, M.J.; Malek, K.; Crabb, D.W. Distribution of messenger RNAs for aldehyde dehydrogenase 1, aldehyde dehydrogenase 2, and aldehyde dehydrogenase 5 in human tissues. J. Investig. Med. 1996, 44, 42–46. [Google Scholar] [PubMed]
Figure 1. Ethanol and acetaldehyde metabolism after alcohol ingestion. Ethanol is metabolized to acetaldehyde by alcohol dehydrogenase 1B (ADH1B), and then acetaldehyde is degraded to acetic acid by aldehyde dehydrogenase 2 (ALDH2).
Figure 1. Ethanol and acetaldehyde metabolism after alcohol ingestion. Ethanol is metabolized to acetaldehyde by alcohol dehydrogenase 1B (ADH1B), and then acetaldehyde is degraded to acetic acid by aldehyde dehydrogenase 2 (ALDH2).
Ijms 18 01943 g001
Figure 2. Lugol chromoendoscopic images. (A): “Field cancerization” in a patient with esophageal squamous cell carcinoma (ESCC) and head and neck squamous cell carcinoma (HNSCC) synchronously. Location of (a) oropharynx, (b) uvula, (c) upper thoracic esophagus, and (d) lower thoracic esophagus. Lesions are indicated by arrowheads; (B): (a) normal esophageal mucosa, (b) esophageal mucosa with multiple dysplastic lesions known as multiple Lugol-voiding lesions. Scale bar = 0.5 cm.
Figure 2. Lugol chromoendoscopic images. (A): “Field cancerization” in a patient with esophageal squamous cell carcinoma (ESCC) and head and neck squamous cell carcinoma (HNSCC) synchronously. Location of (a) oropharynx, (b) uvula, (c) upper thoracic esophagus, and (d) lower thoracic esophagus. Lesions are indicated by arrowheads; (B): (a) normal esophageal mucosa, (b) esophageal mucosa with multiple dysplastic lesions known as multiple Lugol-voiding lesions. Scale bar = 0.5 cm.
Ijms 18 01943 g002
Figure 3. Formation of acetaldehyde-mediated DNA adducts. A single molecule of acetaldehyde reacts with deoxyguanosine (dG) to generate N2-ethylidene-2′-deoxyguanosine (N2-ethylidene-dG), which can be reduced to the stable adducts, N2-ethyl-2′-deoxyguanosine (N2-Et-dG). α-S- and α-R-methyl-γ-hydroxy-1, N2-propano-2′-deoxyguanosine (CrPdG) is derived from dG and two molecules of acetaldehyde. N2-etheno-2′-deoxyguanosine (NεG) is formed from dG and α,β-unsaturated aldehydes during lipid peroxidation, which is mediated by acetaldehyde or reactive oxygen species (ROS).
Figure 3. Formation of acetaldehyde-mediated DNA adducts. A single molecule of acetaldehyde reacts with deoxyguanosine (dG) to generate N2-ethylidene-2′-deoxyguanosine (N2-ethylidene-dG), which can be reduced to the stable adducts, N2-ethyl-2′-deoxyguanosine (N2-Et-dG). α-S- and α-R-methyl-γ-hydroxy-1, N2-propano-2′-deoxyguanosine (CrPdG) is derived from dG and two molecules of acetaldehyde. N2-etheno-2′-deoxyguanosine (NεG) is formed from dG and α,β-unsaturated aldehydes during lipid peroxidation, which is mediated by acetaldehyde or reactive oxygen species (ROS).
Ijms 18 01943 g003
Figure 4. Summary of acetaldehyde-mediated DNA damage. Acetaldehyde causes DNA adducts, DNA single-strand breaks, DNA double-strand breaks (DSBs), point mutations, micronuclei, frameshift mutations, base-pair mutations, deletions, DNA–DNA interstrand or intrastrand cross-links, rearrangements, and sister chromatid exchanges (SCEs). DNA adducts are considered to be partly (but deeply) involved in their formation.
Figure 4. Summary of acetaldehyde-mediated DNA damage. Acetaldehyde causes DNA adducts, DNA single-strand breaks, DNA double-strand breaks (DSBs), point mutations, micronuclei, frameshift mutations, base-pair mutations, deletions, DNA–DNA interstrand or intrastrand cross-links, rearrangements, and sister chromatid exchanges (SCEs). DNA adducts are considered to be partly (but deeply) involved in their formation.
Ijms 18 01943 g004

Share and Cite

MDPI and ACS Style

Mizumoto, A.; Ohashi, S.; Hirohashi, K.; Amanuma, Y.; Matsuda, T.; Muto, M. Molecular Mechanisms of Acetaldehyde-Mediated Carcinogenesis in Squamous Epithelium. Int. J. Mol. Sci. 2017, 18, 1943. https://doi.org/10.3390/ijms18091943

AMA Style

Mizumoto A, Ohashi S, Hirohashi K, Amanuma Y, Matsuda T, Muto M. Molecular Mechanisms of Acetaldehyde-Mediated Carcinogenesis in Squamous Epithelium. International Journal of Molecular Sciences. 2017; 18(9):1943. https://doi.org/10.3390/ijms18091943

Chicago/Turabian Style

Mizumoto, Ayaka, Shinya Ohashi, Kenshiro Hirohashi, Yusuke Amanuma, Tomonari Matsuda, and Manabu Muto. 2017. "Molecular Mechanisms of Acetaldehyde-Mediated Carcinogenesis in Squamous Epithelium" International Journal of Molecular Sciences 18, no. 9: 1943. https://doi.org/10.3390/ijms18091943

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop