Next Article in Journal
Urine CA125 and HE4 for the Detection of Ovarian Cancer in Symptomatic Women
Next Article in Special Issue
Sun-Protective Clothing Worn Regularly during Early Childhood Reduces the Number of New Melanocytic Nevi: The North Queensland Sun-Safe Clothing Cluster Randomized Controlled Trial
Previous Article in Journal
Improving Automatic Melanoma Diagnosis Using Deep Learning-Based Segmentation of Irregular Networks
Previous Article in Special Issue
Genetics and RNA Regulation of Uveal Melanoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Ethanol Metabolism and Melanoma

1
Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
2
Department of Social Medicine, School of Medicine, Saga University, Saga 849-8501, Japan
3
Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
4
Department of Veterans Affairs Medical Center, VA Eastern Colorado Health Care System, Aurora, CO 80045, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2023, 15(4), 1258; https://doi.org/10.3390/cancers15041258
Submission received: 31 December 2022 / Revised: 11 February 2023 / Accepted: 13 February 2023 / Published: 16 February 2023
(This article belongs to the Special Issue Epidemiology and Biological Features of Melanoma)

Abstract

:

Simple Summary

Acetaldehyde (AcAH) is a carcinogenic byproduct of ethanol metabolism. Ethanol-associated malignancies commonly occur in the upper gastrointestinal tract exposed to AcAH after ethanol ingestion. Unexpectedly but true, emerging epidemiological evidence supports a link between alcohol consumption and cutaneous melanoma, suggesting skin exposure to ethanol and AcAH as potential causes of skin cancer. Humans are unavoidably exposed to ethanol and AcAH daily in multiple ways, such as through alcohol consumption, food ingestion, inhalation, skin contact, and bodily microbiota. This review examines the sources of ethanol and AcAH in the skin, their metabolic pathways, and the consequences of dysfunctional ethanol and AcAH metabolizing enzymes, focusing on the role of these factors in melanoma development and progression.

Abstract

Malignant melanoma is the deadliest form of skin cancer. Despite significant efforts in sun protection education, melanoma incidence is still rising globally, drawing attention to other socioenvironmental risk factors for melanoma. Ethanol and acetaldehyde (AcAH) are ubiquitous in our diets, medicines, alcoholic beverages, and the environment. In the liver, ethanol is primarily oxidized to AcAH, a toxic intermediate capable of inducing tumors by forming adducts with proteins and DNA. Once in the blood, ethanol and AcAH can reach the skin. Although, like the liver, the skin has metabolic mechanisms to detoxify ethanol and AcAH, the risk of ethanol/AcAH-associated skin diseases increases when the metabolic enzymes become dysfunctional in the skin. This review highlights the evidence linking cutaneous ethanol metabolism and melanoma. We summarize various sources of skin ethanol and AcAH and describe how the reduced activity of each alcohol metabolizing enzyme affects the sensitivity threshold to ethanol/AcAH toxicity. Data from the Gene Expression Omnibus database also show that three ethanol metabolizing enzymes (alcohol dehydrogenase 1B, P450 2E1, and catalase) and an AcAH metabolizing enzyme (aldehyde dehydrogenase 2) are significantly reduced in melanoma tissues.

1. Introduction

Melanoma is the most serious type of skin cancer, with an increasing incidence worldwide [1,2]. Cumulative evidence indicates that the risk of melanoma correlates with genetic factors [3,4], personal lifestyles [5,6], and phenotypic risk factors that reflect gene/personal lifestyle interactions [7,8]. Individual lifestyle factors associated with melanoma risk include UV exposure, cigarette smoking, alcohol use, overweight and obesity, poor diet, environmental pollution, and stress [5,6]. Recently, we summarized epidemiological data on the association between alcohol consumption and cutaneous melanoma [9]. Nearly half (14 out of 29) of the studies on the relationship between alcohol consumption and melanoma, including 10 cohort and 19 case-control studies, have shown a positive correlation, while only 2 showed a negative correlation. Further, of 20 studies assessing alcohol dose effects, 13 (65%) demonstrated an association between alcohol dose and melanoma risk [9]. These associations became stronger in multiple meta-analyses with larger sample sizes [10,11,12,13]. A systematic meta-analysis by Gandini et al. found that individuals in the highest category of recent alcohol intake had a 30% increased risk of melanoma compared to those in the lowest category, and a nearly two-fold increased risk of melanoma was found with cumulative alcohol consumption [14].
Approximately 4% of cancer cases worldwide are caused by alcohol consumption [15]. The main culprit for this is acetaldehyde (AcAH), an immediate metabolite of ethanol. AcAH is a mutagen and carcinogen implicated in a wide range of cancers by forming adducts with proteins and DNA and disrupting cellular functions [16,17]. However, ethanol and AcAh are derived not just from alcohol drinking but from various sources, some of which exist naturally [18,19]. In fact, our skin is exposed to ethanol and AcAH every day, regularly at extremely low and safe levels [18,19,20]. While our skin, like the liver, is equipped with ethanol metabolism mechanisms to reduce the concentration of ethanol and AcAH [18,20], dysfunction of these ethanol and AcAH metabolizing enzymes in the skin may greatly influence the skin biology and increase the risk of ethanol/AcAH-associated skin diseases.
This review summarizes various sources of ethanol and AcAH in the skin and explains how ethanol metabolism can affect an individual’s sensitivity threshold to AcAH carcinogenesis.

2. Exposure to Ethanol and AcAH

Humans are in a chemical and toxicological environment [21] and are exposed to ethanol and AcAH in many ways. Once in the bloodstream, ethanol and AcAH can reach many organs and tissues, including the skin [22]. The skin is also directly exposed to alcohols and aldehydes from natural chemicals or industrialized products [19,20].

2.1. Sources of Ethanol

Ethanol is not only the active ingredient of alcoholic beverages (beers, wines, and spirits) but also is a ubiquitous substance from various sources (Figure 1). It is one of the main indoor and outdoor pollutants [19]. In addition to alcoholic beverages and air pollutants, non-alcoholic beverages on the market can contain as much as 0.5% ethanol [23]. Certain herbal medicines, including those used to treat coughs, colds, and gastrointestinal (GI) diseases, are also sources of ethanol [24]. Furthermore, many foods contain ethanol, which is produced from sugar through fermentation. Examples include fermented foods (i.e., bread, yogurt, vinegar, and kimchi), preservatives, bakery products, fruit, and fruit juices [25]. Brewers and bakers use yeast to make a variety of alcoholic beverages and expand the dough.
Even without the exogenous ethanol intake mentioned above, our bodies contain low levels of ethanol. Baseline ethanol levels in the blood and breath can reach 0.02–0.15 mg/dL and 0.07–0.39 mg/L, respectively, without consuming alcohol [26]. These low levels of ethanol are generated by microbial fermentation. Fermenting yeasts such as Saccharomyces and Candida (C.) and fermenting bacteria such as Zymomonas mobilis and Sarcina ventriculi are present in our oral cavity and digestive tract. These microbiotas use anaerobic respiration to convert non-ethanol, carbohydrate-rich foods such as glucose and lactose to ethanol by fermentation in the oral cavity, GI system, or urinary system [27]. This microbial ethanol production is particularly interesting in certain medical conditions.
In auto-brewery syndrome (ABS), also known as gut fermentation syndrome, microbiota-derived ethanol concentrations in the body are comparable to those produced by directly consuming alcoholic beverages [28,29]. In a study conducted by Malik et al., the blood alcohol concentration (BAC) in an ABS patient reached 400 mg/dL [30], which is comparable to a BAC that can cause respiratory depression, coma, and death [31]. The species causing the ABS include Klebsiella pneumoniae, C. albicans, C. glabrata, Saccharomyces cerevisiae, C. intermedia, C. parapsilosis, and C. kefyr [32]. ABS is a rare condition. This syndrome could be underdiagnosed, as the symptoms may be mood changes, delirium, and brain fog or mimic a food allergy [30,32,33]. Triggers of this ABS include meals high in carbohydrates, psychological stress, and reduced dietary intake. ABS may also be related to pre-existing conditions, including a history of antibiotic use and comorbidities, such as type 2 diabetes, obesity, liver cirrhosis, and Crohn’s disease [27].
Ethanol can also be produced from AcAH by microbial reduction [34].

2.2. Sources of AcAH

Similar to ethanol exposure, human exposure to AcAH is not only from alcohol consumption but also from other diverse sources (Figure 1). AcAH can be produced endogenously in any tissue with high ethanol metabolizing enzyme activity [35]. Bodily AcAH may come from oral and gut microbes that metabolize ethanol to AcAH. Salivary AcAH levels reached up to 140 µM after ingesting a moderate amount of ethanol (0.5 g/kg body weight) [35]. Long-term exposure to locally produced AcAH in saliva may explain the higher risk of upper GI cancers in heavy drinkers [35].
The atmospheric AcAH are from photochemical production, net ocean emissions, biogenic emissions, biomass burning emissions, and anthropogenic emissions, accounting for 60%, 27%, 11%, 1.6%, and < 1% of global AcAH production, respectively [36]. The largest source is the photo-oxidation of volatile organic compounds such as alkanes and alkenes [36]. The second-largest source is water bodies that degrade dissolved organic compounds through the photochemical mechanism and emit AcAH into the atmosphere [37]. Most plant cells and some microorganisms use anaerobic respiration to break down glucose to AcAH and release carbon dioxide (biogenic emissions). Biomass burning emissions are from wildfire smoke and biofuel burning. Another source of AcAH includes urban and industrial pollution, such as residential fireplaces, woodstoves, ethanol fuel, vehicle exhaust fumes, coal refining, and waste processing. Therefore, like ethanol, AcAH is a common noxious environmental pollutant [19]. Nevertheless, sinks of atmospheric AcAH include reaction with hydroxyl radical, photolysis, and wet and dry deposition, leading to an overall atmospheric lifetime for AcAH of approximately 20 h [36,38].
AcAH sources also include occupational exposure. Individuals may be at risk of higher AcAH exposure when working in gas stations [39], transportation vehicles [40], waterpipe café [41], bakeries [42], and beauty salons [43], as well as in industries using AcAH as a solvent for perfumes, polyester resins, acetic acid, mirror silvering, tanning leather, denaturing alcohol, fuel compositions, gelatin fiber hardeners, glue and casein products, paper, cosmetics, aniline dyes, plastics, and synthetic rubber [44].
AcAH is also contained in various foods (e.g., fermented food, roasted coffee, bread, and ripe fruit) and beverages and is used as a flavoring agent and a preservative for fruits and fish [45,46].
Furthermore, AcAH is a byproduct of tobacco smoking [47]. When coupled with nicotine, AcAH has been shown to increase the addictive potential of smoking [48,49]. Nieminen et al. reported that the concentration of AcAH in saliva remains as high as 261 μM with one cigarette, which is higher than the AcAH concentration after drinking high-concentration (40%) ethanol for a short period [47]. Smoking can increase AcAH production from ethanol in saliva by 60–75%; for heavy drinking, the increase in AcAH is up to 100% [47].
Another source of AcAH is pyruvate, an important energy source produced during glycolysis. In anaerobic conditions, yeast use pyruvate decarboxylase (PDC) to convert pyruvate to AcAH, and C. albicans, an opportunistic pathogenic yeast, has been shown to contribute to oral squamous cell carcinoma progression by producing high levels of AcAH from glucose under low oxygen conditions [50,51]. Some bacteria also use PDC to convert pyruvate to AcAH. However, as a common pathway in bacteria, pyruvate is first decarboxylated to acetyl-CoA by pyruvate ferredoxin oxidoreductase and/or pyruvate formate-lyase. Acetyl-CoA is then converted into AcAH by acetylating acetyl-CoA reductase in bacteria [52]. Since the enzymes to produce AcAH from pyruvate have not been reported in humans, pyruvate-derived AcAH is likely produced from bodily microbiota rather than human cells.
Collectively, ethanol and AcAH are part of our life. The skin is exposed to these toxic metabolites through the air, water, land, smoke, food, and bodily microbiota, among other means. Since ethanol and AcAH are readily degraded in the environment or metabolized in the body, the anticipated skin exposure levels are very low or safe. However, when the body’s metabolic process becomes dysfunctional, high levels of ethanol and AcAH can cause health consequences, including skin diseases.

3. Ethanol Metabolism and Its Contribution to Human Diseases

Due to their ubiquitous nature, ethanol and AcAH can affect our health daily, particularly in industrialized countries [19]. In this context, ethanol metabolism is important since it directly determines the fate of ethanol and AcAH in the body, with varying degrees of biological consequences.
The first-pass metabolism of ethanol after its ingestion occurs in the GI mucosa and flora before it reaches the bloodstream and then in the liver, where most ethanol metabolizing enzymes are present [53,54]. Overall, over 90% of ingested ethanol is metabolized, and the remainder is excreted through breath (1–3%), urine (1–3%), and sweat (traces) without modification [55,56]. As our bodies, including the skin, are exposed to ethanol [18,20], humans have evolved to harbor a set of metabolic detoxifying enzymes to reduce the toxic effects of alcohols, aldehydes, and other xenobiotics from various sources. This chapter will explain the roles of each enzyme, its expression, and the potential consequences when the enzyme becomes dysfunctional, particularly its carcinogenic effects.

3.1. Ethanol Metabolizing Enzymes and Their Impacts on Humans

The main pathway of ethanol metabolism is oxidation into the highly reactive AcAH [57] (Figure 2a). The predominant enzyme that oxidizes ethanol to AcAH is cytosolic alcohol dehydrogenase (ADH) [58]. Other enzymes that convert ethanol to AcAH are microsomal cytochrome P450 2E1 (CYP2E1) and peroxisomal catalase (CAT) [54,59].
There are seven isoforms of ADH [60]. So far, all of them are known to participate in ethanol oxidation except ADH6 [61]. Although ADH families have a limited distribution in human tissues, primarily in hepatocytes, they are also found in the GI tract and certain tissues, including the skin epidermis and dermis [18,58].
ADH1A, ADH1B, and ADH1C (referred to as ADH1-3) play a dominant role in the oxidative metabolism of ethanol in the liver after low to moderate alcohol consumption [60,61]. Genetic polymorphisms with physiological significance occur in ADH1B and ADH1C [61]. The common ADH1B*2 allele in East Asia (75%) and the relatively common ADH1B*3 allele in Eastern Africa (10-30%) [61] display quick ethanol turnover, leading to rapid accumulation of AcAH following ethanol intake [62]. A meta-analysis of 18 studies demonstrates that ADH1B polymorphisms, particularly rs1229984 Arg47His with a faster metabolic character, increase the risk of bladder cancer, colorectal cancer, and upper aerodigestive tract cancers (tumors of the oral cavity, pharynx, larynx, and esophagus) for alcoholics [63]. On the contrary, the ADH1C*2 allele metabolizes ethanol < 2.5 times slower than the ADH1C*1 allele. Therefore, Caucasians with ADH1C*1/2 heterogeneity have an increased risk for alcohol-related esophageal, hepatocellular, and head and neck cancers due to slow ethanol turnover [64]. Interestingly, a paper reports that these enzymes are also expressed in the epidermis of human skin (foreskin, breast, and abdomen) [18].
ADH4, a class II ADH, is active only when large quantities of ethanol are consumed and is expressed almost exclusively in the liver [61]. ADH5, a class III ADH, is ubiquitously expressed and implicated in the first-pass metabolism of ethanol [65,66]. ADH5 has two other main functions: it inactivates formaldehyde and nitric oxide by converting them into formic acid and S-nitrosoglutathione, respectively, in a glutathione-dependent manner [67,68]. Formaldehyde, an endogenous byproduct of oxidative demethylation reactions, is genotoxic and carcinogenic, as it cross-links proteins and nucleic acids [69]. Therefore, ADH5 dysfunction in scavenging formaldehyde is linked to defective hematopoiesis and increased leukemia [70]. ADH7, a class III ADH, is expressed in the GI tract endothelial cells and implicated in ethanol’s first-pass metabolism [65].
CYP2E1 constitutes the microsomal ethanol oxidizing system that converts ethanol to AcAH, accounting for around 10% of the total ethanol oxidizing capacity of the liver [54]. CYP2E1 is highly expressed in the liver but is also detectable in extrahepatic tissues, including the lung, kidney, skin, brain, placenta, and testis [71,72]. Although CYP2E1 is critical for oxidizing higher levels of ethanol due to its lower affinity for ethanol than most ADH isoforms, the fact is that CYP2E1 is inducible up to 10-fold by ethanol [60,73]. An alcohol-induced increase in the microsomal pathway contributes to liver pathology due to the generation of reactive oxygen species (ROS) during this reaction [54,57,60], leading to the development of hepatocellular carcinoma [74]. Diet, lifestyle, and physiological factors substantially influence CYP2E1 phenotype.
Hepatic CAT plays a minimal role in ethanol metabolism. However, blood CAT activity significantly correlates with alcohol consumption [75]. Since its oxidation of ethanol is hydrogen peroxide (H2O2)-dependent, CAT works more efficiently under elevated ROS levels and oxidative stress following heavy drinking [54]. Since CAT-catalyzed ethanol oxidation occurs in the brain, this gene may impact susceptibility to alcohol dependence [76]. While CAT has a limited role in ethanol clearance, its polymorphism rs1001179 (Cys262Thr) has been shown to increase prostate cancer risk because of increased ROS [77,78]. Several reports have demonstrated that CAT is highly expressed in the skin. CAT activity in the epidermis is more than eight times higher than in the underlying dermis [79] and plays a key role in protecting skin against UV radiation and skin aging [80,81]. In epidermal melanocytes, the expression and activity of CAT are directly related to melanin content, acting synergistically to defend against solar UV damage [82]. Consistent with these reports, reduced CAT levels and increased H2O2 levels in the skin have been related to vitiligo and xeroderma pigmentosum [83,84,85,86].
While a majority of ingested ethanol undergoes oxidative metabolism, a small fraction (0.1–0.2%) can undergo non-oxidative metabolism [56] (Figure 2b), resulting in the enzymatic conjugation of ethanol to endogenous metabolites to form ethyl glucuronide (EtG), ethyl sulfate (EtS), phosphatidylethanol (PEth), and fatty acid ethyl ester (FAEE) [87,88]. EtG and EtS can be retained in the blood (1–3 h), urine (2–4 weeks), and keratinized matrices such as hair (months), and these metabolic products are used as metabolic biomarkers of recent alcohol consumption [89,90]. For example, transdermal sensors have been developed to measure skin concentrations of ethanol or its metabolite EtG after alcohol drinking [91]. The metabolites produced by non-oxidative metabolism are not considered non-toxic. EtG and EtS affect toll-like receptor signaling and reduce energy metabolism [92]. Phospholipase D, an enzyme synthesizing PEth, is involved in keratinocyte differentiation [93]. FAEE interferes with cell signaling pathways and disrupts organelle function, contributing to ethanol toxicity in tissues with a limited oxidative capacity [88].
Additionally, ethanol itself affects the human body. Ethanol can enhance the activity of adenylyl cyclase, one of the main targets of ethanol in the cAMP/protein kinase A (PKA) signaling pathway [94,95]. PKA phosphorylates and dephosphorylates many proteins, mediating various cellular processes, including glucose and lipid metabolism, cell growth, differentiation, and death [96].
In conclusion, ethanol metabolizing enzymes and pathways are related to the development of cancers. While most exogenous ethanol is metabolized in the GI tract and liver, a fraction goes to the peripheral tissues. Due to the lack of plasma protein binding, ethanol is readily distributed in all body tissues except fat and bone [60]. Therefore, the skin may have evolved to express ethanol metabolizing enzymes.

3.2. AcAH Metabolizing Enzymes and Their Effects on Humans

Since AcAH is incriminated in pathogenesis by its potent and lasting damage to cellular macromolecules and its mutagenic and carcinogenic effects on DNA, removing AcAH from the body is essential [97]. In the second step of oxidative ethanol metabolism, AcAH is further oxidized to acetate by aldehyde dehydrogenase (ALDH), mainly cytosolic ALDH1A1, mitochondrial ALDH1B1, and mitochondrial ALDH2 (Figure 2a). ALDH1A1 has an affinity for AcAH but metabolizes AcAH less efficiently. ALDH1B1 and ALDH2 have a broad tissue distribution and are catalytically active toward a wide range of aldehyde substrates [20,98,99]. ALDH1B1 shares a 75% peptide sequence homology with ALDH2 [100].
ALDH1A1 and ALDH1B1 have limited involvement in AcAH metabolism under physiological conditions, as they have a higher Km for AcAH compared to ALDH2 (with Km values of 180, 55, and 0.2 μM, respectively) [100]. Polymorphisms of ALDH1A1 and ALDH1B1 have been identified in Europeans and are associated with alcohol-related phenotypes [101,102].
ALDH2 is involved in oxidizing toxic aldehydes of both exogenous and endogenous sources, including those generated from endoplasmic reticulum stress, oxidative stress, and other stress-inducing conditions [99,103]. Thus, ALDH2 has special physiopathological significance, and its downregulation and inactivation have been associated with many health conditions, including cardiovascular diseases, neurodegenerative diseases, alcohol-related liver disease, and cancer [99].
ALDH2 downregulation and inactivation are the results of the complex interplay between genetic susceptibility (e.g., allelic variation, polymorphism, and epigenetics) and multiple environmental factors (e.g., alcohol, smoking, drugs, and high-fat diet) [104]. One of the well-studied ALDH2 polymorphisms is the ALDH2*2 allele (rs671), which results from a single point mutation changing the glutamic acid at the 487th position to a lysine (Glu487Lys of mature protein or Glu504Lys of the precursor protein) [61]. Due to the tetrameric structure of ALDH2 and the dominant inactive phenotype of the variant, individuals can be categorized into three ALDH2 genotypes. The wild-type ALDH2*1/*1 genotype possesses a normal enzymatic activity, while the heterozygous ALDH2*1/*2 genotype retains approximately 15.6% of normal activity, and the homozygous ALDH2*2/*2 genotype loses total enzyme activity [105]. The ALDH2*2 (either ALDH2*1/*2 or ALDH2*2/*2) mutation is one of the most common genotypes carried by more than 8% of the world population and is frequent in East Asians, with a prevalence of 30–50% [106]. When similar amounts of ethanol are consumed, ALDH2-deficient individuals have significantly higher AcAH-derived DNA adducts in their blood than individuals with the wild-type genotype [107].
The mechanistic basis behind ALDH2 downregulation without ALDH2 polymorphisms is complex and multi-factorial: (1) ALDH2 expression can be epigenetically regulated through gene methylation and hyperacetylation [108,109,110,111]; (2) ALDH2 expression is regulated by several transcriptional factors [112,113,114] and can be negatively regulated by cAMP, potentially through phosphorylation of its transcription factor hepatocyte nuclear factor 4 by PKA [115]; (3) ALDH2 is negatively regulated by small non-coding RNAs, like miR-193 and -27a-3p [116,117]; (4) ALDH2 activity can be inactivated by chemical modifications at several functional groups, such as oxidation at Cys residue(s), phosphorylation at Ser residue(s), nitration at Tyr residue(s), and acetylation at Lys residue(s) [104,118]; and (5) ALDH2 activity is regulated by several signaling molecules, including c-Jun N-terminal kinase, AMP-activated protein kinase, ε protein kinase C, and sirtuin 3, through phosphorylating ALDH2 protein or decreasing its acetylation level [119].
These mechanisms will have biological effects on ALDH2 function, ultimately contributing to carcinogenesis. Indeed, ALDH2 dysfunction has been widely reported to correlate with tumor initiation and progression in various cancers [120]. Specifically, the ALDH2*2 allele with a slower AcAH-metabolizing capacity represents an increased risk of alcohol-related cancers such as esophageal and head and neck cancers [106,121]. When this slow ALDH2*2 allele is combined with the rapid ADH1B*2/*2 allele (rs1229984), a clear positive dose-response relationship can be seen between alcohol consumption and worse survival in patients with bladder and head and neck cancers [122,123], which may be related to the accumulation of large amounts of AcAH. However, ADH1B polymorphisms do no appear to result in significant differences in circulating AcAH levels between ALDH2 wild-type and heterozygous genotypes [124]. Moreover, it is worth mentioning that the effects of the ALDH2*2 allele on alcohol-related cancers may be complex due to its direct carcinogenesis and indirect protective effect of alcohol avoidance [125].
In addition to the enzymes mentioned above, CYP2E1 also has the ability to oxidize AcAH (Figure 2a) and plays an alternative role in clearing AcAH from the liver after alcohol consumption [126]. The joint effects of ALDH2*2 and CYP2E1 (e.g., rs2031920) polymorphisms on tumor susceptibility in alcohol drinkers deserve to be evaluated [127].

4. Ethanol Metabolism and Its Contribution to Melanoma

As most research on ethanol or AcAH concentrates on the liver and GI tract with little focus on the skin [128,129], we previously summarized their potential roles in skin biology and melanoma [9]. Similarly, despite the well-known roles of ethanol and AcAH metabolizing enzymes, there is a lack of direct evidence for their effects on melanoma. This chapter will examine the expression of these enzymes in melanoma and consider the potential association with melanoma biology.

4.1. Potential Roles of Dysfunctional Ethanol and AcAH Metabolism in Melanoma

Ethanol exposure may disrupt a range of melanocyte development processes leading to defective skin pigmentation [130], in which the role of ethanol metabolism is unknown. On the other hand, as previously mentioned in Section 3.1, ethanol increases cAMP/PKA pathway activity, which has been implicated in melanocyte pigmentation, melanomagenesis [131,132], and therapy resistance [133]. Therefore, ethanol may directly affect melanoma initiation and progression.
The enzymes metabolizing ethanol and AcAH have various polymorphic forms, each with distinct catalytic kinetics, leading to various AcAH production. ALDH2 dysfunction is a significant cause of toxic AcAH accumulation [124]. Additionally, AcAH is produced by normal human microbiota, including those on the skin, and damages DNA, impairs DNA repair, and increases ROS production. High ethanol intake and redox imbalance in ALDH2-related mitochondrial respiration can elevate CYP2E1 levels, causing further increases in ROS levels [73,134]. Excessive ROS production has widespread impacts on cell biology, including inducing inflammation and altering signaling pathways that control cancer stem cell renewal, cell proliferation, differentiation, and angiogenesis, creating a favorable microenvironment for cancer initiation [9]. Evidence suggests that a high pro-oxidant state with impaired antioxidant defenses is related to melanocyte malignant transformation and melanoma progression [135,136].
Ethanol consumption directly affects the skin, disrupting skin physiology and homeostasis by influencing cutaneous metabolism, immune response, vasculature, and antioxidant system [137]. This skin damage from ethanol is exacerbated by UV exposure, and ethanol and UV radiation can act synergistically to induce mutations and weaken protection mechanisms, including reducing melanin production and glutathione levels [138]. Ethanol and UV radiation also activate cAMP/PKA, mitogen-activated protein kinase, phosphatidylinositol 3-kinase/protein kinase B, and Wnt/β-catenin signaling pathways, leading to altered expression of the melanocyte transcription factor microphthalmia-associated transcriptional factor (MITF) [9]. MITF regulates melanocyte biology, and its dysregulation is directly linked to melanoma development [139].
Animal studies have also shown that ethanol and AcAH promote melanoma progression and metastasis [140]. The exact mechanism is unclear, but ethanol and AcAH may remodel the tumor microenvironment to facilitate tumor migration and invasion. For example, ethanol intake reduces circulating CD8+ T cells and NK cells [141,142], activates the inflammasome [143], induces hypoxia-inducible factor 1 expression [144,145], and enhances extracellular matrix degradation by matrix metalloproteinases [146], all of which contribute to the progression of melanoma.
Overall, dysfunctional ethanol and AcAH metabolism would amplify the impact of ethanol and AcAH in tumor formation. For more information on how ethanol and AcAH contribute to melanoma initiation and progression, please refer to reference [9].

4.2. Ethanol Metabolizing Enzymes in Human Melanoma

Several recent studies have suggested the potential roles of ethanol metabolizing enzymes in human melanoma by identifying differentially expressed genes using publicly available microarray data. Liu et al. analyzed three microarray datasets from the Gene Expression Omnibus (GEO) database, including GSE15605, GSE46517, and GSE114445, and found that ADH1B was significantly downregulated in primary melanoma samples vs. normal skin samples [147]. Their Cox regression analysis revealed that ADH1B is an independent prognostic factor in human melanoma [147]. Another study confirmed the downregulation (~9-fold) of ADH1B in human primary melanoma samples compared to normal skin tissues by analyzing two of the three datasets (GSE15605 and GSE46517) and another dataset GDS1375 [148].
Since the above studies did not include metastatic melanoma samples, we further analyzed the gene expressions of ethanol metabolizing enzymes in the datasets GSE15605, GSE7553, and GSE46517 [149,150,151] (Figure 3a–i, left). Among the ethanol metabolizing enzymes (ADH1-7, CYP2E1, and CAT), ADH1B, ADH5, and CAT were relatively highly expressed in all three datasets. When compared to normal skin tissues, primary melanoma tissues showed reduced ADH1B, CYP2E1, and CAT gene expression in at least two of three datasets (Figure 3b,h,i, left), consistent with previous observations for ADH1B [147,148]. Moreover, CYP2E1 was shown to be further downregulated in metastatic melanoma compared to primary melanoma in two of three datasets (Figure 3h, left). Unexpectedly, ADH5 was significantly upregulated in primary melanoma samples of one dataset and showed an increasing trend in melanoma in the other two datasets (Figure 3e, left).
Since melanocytes, the skin pigment-producing cells that give rise to melanoma, make up only a small portion of normal skin tissue [153,154], we further compared the gene expression levels of ethanol metabolizing enzymes in normal human skin, nevus, and melanoma tissues (Figure 3a–i, right). We analyzed another dataset GSE114445, which contained normal skin, common nevi, dysplastic nevi, and primary melanoma [152]. Results showed no differences in the gene expression levels of ethanol metabolizing enzymes between normal skin and melanocytic nevi except for downregulated ADH1A and ADH1B in common nevi (Figure 3a,b, right). Although we did not find decreased ADH1B and CAT in primary melanoma compared to normal skin, a decrease in CYP2E1 and an increase in ADH5 were observed when compared to normal skin and dysplastic nevi (Figure 3b,e,h,i, right).
These data suggest a potential role for altered gene expression levels of ethanol metabolizing enzymes in human melanoma. The mechanism behind these data is unknown, but their expression levels may ultimately relate to the production and metabolism of AcAH.

4.3. AcAH Metabolizing Enzymes in Human Melanoma

Among four AcAH metabolizing enzymes, upregulation of ALDH1A1 and ALDH1B1 have been reported in various cancers. ALDH1A1 is involved in cancer stem cell maintenance, metabolism, and drug resistance in multiple cancer types, including melanoma [155]. ALDH1B1 is upregulated in several cancers, such as colorectal and pancreatic cancers, and acts as an oncogene [156,157]. The role of ALDH1B1 in melanoma remains unclear. On the other hand, ALDH2 expression has been shown to be downregulated in breast, lung, esophageal, and head and neck cancers, and this reduction in ALDH2 expression has been linked to a poor prognosis of cancer patients [158,159].
We compared the gene expression levels of ALDH1A1, ALDH1B1, and ALDH2 in normal human skin vs. primary and metastatic melanoma by analyzing the gene profiling datasets GSE15605, GSE7553, and GSE46517 as explained in Section 4.2 [149,150,151]. We found that ALDH2 expression levels were significantly downregulated in three GEO melanoma datasets (Figure 4a–c, left). Wu et al. also observed downregulated ALDH2 gene expression in primary human melanoma [160]. Moreover, analysis of the GSE11445 dataset [152] revealed that some patients with primary melanoma exhibited decreased expression of the ALDH2 gene compared to normal skin and melanocytic nevi (Figure 4c, right). These findings suggest that the downregulation of ALDH2 expression could impact clinical outcomes in melanoma patients.
Considering the consequences of low ALDH2 expression, leading to AcAH accumulation and compensation by other enzymes, it is plausible to explain how ALDH2 downregulation in tumor tissues contributes to the poor prognosis of cancer patients. ALDH2 dysfunction has been widely linked to an increased risk of alcohol-related cancers [120], and individuals carrying the ALDH2*2 allele may face a 10-fold increased risk of developing upper esophageal and pharyngeal cancers with chronic alcohol intake [97].
However, the clinical significance of ALDH2 polymorphism in melanoma remains unclear. The ALDH2*2 allele is prevalent in populations with low melanoma incidence, such as Mongolians [161], but rare in high-risk populations like Caucasians with high alcohol consumption [6,161]. Therefore, we analyzed global melanoma data, alcohol consumption, and published ALDH2 information and found that the wild-type ALDH2 allele was strongly positively correlated with melanoma incidence (R = 0.70; p < 0.001), while the allelic variants had a modest to strong negative correlation (R = −0.70; p < 0.001 and R = −0.51; p = 0.01 for ALDH2*1/*2 and ALDH2*2/*2, respectively) [6]. Interestingly, alcohol consumption showed similar trends: people with the wild-type ALDH2 allele tended to drink more alcohol (R = 0.39; p = 0.07), while the allelic variants consumed less alcohol (R = −0.38; p = 0.07 and R = −0.25; p = 0.26 for ALDH2*1/*2 and ALDH2*2/*2, respectively) [6]. These data suggest that carriers of the ALDH2 mutation may develop less melanoma because they drink less alcohol [105]. This notion is supported by observations by Koyanagi et al. on GI tract cancers [125], who split the ALDH2 allele effects into the carcinogenic effect (direct effect from increased AcAH exposure) and the protective effect (indirect effect from decreased alcohol intake). They found that while the ALDH2 allele increased the risk of GI tract cancers, especially those of the upper GI tract, the risk protection was also prominent for all GI tract cancers observed except small intestine cancer [125]. These demonstrate the complexity of the relationship between ALDH2 expression, ALDH2 polymorphism, and melanoma outcomes. Understanding the mechanisms underlying ALDH2 downregulation or inactivation in cancers not involving ALDH2 polymorphisms is crucial.

5. Conclusions and Future Directions

Alcohol consumption has long be recognized as a risk factor for many GI tract cancers due to their exposure to mutagenic AcAH. However, growing evidence suggests that alcohol consumption is a potential culprit in cutaneous melanoma. The underlying mechanisms by which ethanol and AcAH induce carcinogenesis in melanoma remain unclear. It is important to investigate whether there are shared mechanisms between melanoma and other cancers. On the other hand, the skin is exposed to toxic environmental metabolites in addition to the ethanol and AcAH that circulate from the digestive system following alcohol drinking and food intake. All these different sources of ethanol and AcAH, combined with other risk factors such as UV radiation and smoking, make the pathogenesis of melanoma complex. Understanding the contributions of each source of ethanol and AcAH in melanoma development is essential.
A simple but important question is how ethanol and AcAH contribute to melanomagenesis beyond the formation of carcinogenic macromolecular adducts. One possible explanation is that the levels of ethanol and AcAH in the skin exceed the detoxification capacity of metabolic enzymes, thereby activating the cAMP/PKA signaling and promoting the formation of macromolecular adducts. Like the liver, the skin harbors a set of ethanol and AcAH metabolizing enzymes that play a crucial role in preventing the excessive accumulation of ethanol and toxic metabolites. Additionally, the cutaneous antioxidant system can help reduce oxidative stress caused by ethanol metabolism. However, it is still unclear whether there are differences in the biological properties of ethanol and AcAH detoxification and ROS scavenging between skin tissue and the digestive system.
Nevertheless, evidence supports that downregulation and inactivation of ethanol or AcAH metabolizing enzymes due to genetic polymorphisms and somatic mutations are associated with various human diseases. Therefore, restoring the activity of these metabolic enzymes, particularly the ALDH2 enzyme, may be important to prevent melanoma initiation and progression.

Author Contributions

Conceptualization, M.F., Z.Z. and T.Y.; original draft preparation, Z.Z., T.Y., S.S., V.H. and A.M.; review and editing, M.F., Z.Z. and A.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the National Institutes of Health (NIH)/National Cancer Institute (NCI) R01CA197919 (to M.F.), NIH/National Institute of Allergy and Infectious Diseases (NIAID) R01AI156534-01A1 (to M.F.), Veterans Affairs Merit Review Award 5I01BX001228 (to M.F.), NIH/National Institute on Alcohol Abuse and Alcoholism (NIAAA) R21AA028904 (to M.F.), Cancer League of Colorado (to M.F. and T.Y.), Dermatology Foundation (to T.Y.), University of Colorado Cancer Center-Nutrition Obesity Research Center (UCCC-NORC) Metabolism and Cancer Pilot Grant (to M.F.), and Melanoma Research Alliance (to T.Y.).

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the writing of the manuscript.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2022. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef] [PubMed]
  2. Whiteman, D.C.; Green, A.C.; Olsen, C.M. The growing burden of invasive melanoma: Projections of incidence rates and numbers of new cases in six susceptible populations through 2031. J. Investig. Dermatol. 2016, 136, 1161–1171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Read, J.; Wadt, K.A.; Hayward, N.K. Melanoma genetics. J. Med. Genet. 2016, 53, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Hawkes, J.E.; Truong, A.; Meyer, L.J. Genetic predisposition to melanoma. Semin. Oncol. 2016, 43, 591–597. [Google Scholar] [CrossRef]
  5. Sawada, Y.; Nakamura, M. Daily lifestyle and cutaneous malignancies. Int. J. Mol. Sci. 2021, 22, 5527. [Google Scholar] [CrossRef]
  6. Batta, N.; Shangraw, S.; Nicklawsky, A.; Yamauchi, T.; Zhai, Z.; Menon, D.R.; Gao, D.; Dellavalle, R.P.; Fujita, M. Global melanoma correlations with obesity, smoking, and alcohol consumption. JMIR Dermatol. 2021, 4, e31275. [Google Scholar] [CrossRef]
  7. Ribero, S.; Glass, D.; Bataille, V. Genetic epidemiology of melanoma. Eur. J. Dermatol. 2016, 26, 335–339. [Google Scholar] [CrossRef]
  8. Tagliabue, E.; Gandini, S.; Bellocco, R.; Maisonneuve, P.; Newton-Bishop, J.; Polsky, D.; Lazovich, D.; Kanetsky, P.A.; Ghiorzo, P.; Gruis, N.A.; et al. MC1R variants as melanoma risk factors independent of at-risk phenotypic characteristics: A pooled analysis from the M-SKIP project. Cancer Manag. Res. 2018, 10, 1143–1154. [Google Scholar] [CrossRef] [Green Version]
  9. Yamauchi, T.; Shangraw, S.; Zhai, Z.; Ravindran Menon, D.; Batta, N.; Dellavalle, R.P.; Fujita, M. Alcohol as a non-UV social-environmental risk factor for melanoma. Cancers 2022, 14, 5010. [Google Scholar] [CrossRef]
  10. Rota, M.; Pasquali, E.; Bellocco, R.; Bagnardi, V.; Scotti, L.; Islami, F.; Negri, E.; Boffetta, P.; Pelucchi, C.; Corrao, G.; et al. Alcohol drinking and cutaneous melanoma risk: A systematic review and dose-risk meta-analysis. Br. J. Dermatol. 2014, 170, 1021–1028. [Google Scholar] [CrossRef]
  11. Miura, K.; Zens, M.S.; Peart, T.; Holly, E.A.; Berwick, M.; Gallagher, R.P.; Mack, T.M.; Elwood, J.M.; Karagas, M.R.; Green, A.C. Alcohol consumption and risk of melanoma among women: Pooled analysis of eight case-control studies. Arch. Dermatol. Res. 2015, 307, 819–828. [Google Scholar] [CrossRef] [PubMed]
  12. Bagnardi, V.; Rota, M.; Botteri, E.; Tramacere, I.; Islami, F.; Fedirko, V.; Scotti, L.; Jenab, M.; Turati, F.; Pasquali, E.; et al. Alcohol consumption and site-specific cancer risk: A comprehensive dose-response meta-analysis. Br. J. Cancer 2015, 112, 580–593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Rivera, A.; Nan, H.; Li, T.; Qureshi, A.; Cho, E. Alcohol intake and risk of incident melanoma: A pooled analysis of three prospective studies in the United States. Cancer Epidemiol. Biomark. Prev. 2016, 25, 1550–1558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Gandini, S.; Masala, G.; Palli, D.; Cavicchi, B.; Saieva, C.; Ermini, I.; Baldini, F.; Gnagnarella, P.; Caini, S. Alcohol, alcoholic beverages, and melanoma risk: A systematic literature review and dose-response meta-analysis. Eur. J. Nutr. 2018, 57, 2323–2332. [Google Scholar] [CrossRef]
  15. Rumgay, H.; Shield, K.; Charvat, H.; Ferrari, P.; Sornpaisarn, B.; Obot, I.; Islami, F.; Lemmens, V.; Rehm, J.; Soerjomataram, I. Global burden of cancer in 2020 attributable to alcohol consumption: A population-based study. Lancet Oncol. 2021, 22, 1071–1080. [Google Scholar] [CrossRef]
  16. Rumgay, H.; Murphy, N.; Ferrari, P.; Soerjomataram, I. Alcohol and Cancer: Epidemiology and biological mechanisms. Nutrients 2021, 13, 3173. [Google Scholar] [CrossRef]
  17. Heymann, H.M.; Gardner, A.M.; Gross, E.R. Aldehyde-induced DNA and protein adducts as biomarker tools for alcohol use disorder. Trends Mol. Med. 2018, 24, 144–155. [Google Scholar] [CrossRef]
  18. Cheung, C.; Smith, C.K.; Hoog, J.O.; Hotchkiss, S.A. Expression and localization of human alcohol and aldehyde dehydrogenase enzymes in skin. Biochem. Biophys. Res. Commun. 1999, 261, 100–107. [Google Scholar] [CrossRef]
  19. Pargoletti, E.; Rimoldi, L.; Meroni, D.; Cappelletti, G. Photocatalytic removal of gaseous ethanol, acetaldehyde and acetic acid: From a fundamental approach to real cases. Int. Mater. Rev. 2022, 67, 864–897. [Google Scholar] [CrossRef]
  20. Cheung, C.; Davies, N.G.; Hoog, J.O.; Hotchkiss, S.A.; Smith Pease, C.K. Species variations in cutaneous alcohol dehydrogenases and aldehyde dehydrogenases may impact on toxicological assessments of alcohols and aldehydes. Toxicology 2003, 184, 97–112. [Google Scholar] [CrossRef]
  21. Ahn, J.; Hayes, R.B. Environmental influences on the human microbiome and implications for noncommunicable disease. Annu. Rev. Public Health 2021, 42, 277–292. [Google Scholar] [CrossRef] [PubMed]
  22. Seitz, H.K.; Stickel, F. Molecular mechanisms of alcohol-mediated carcinogenesis. Nat. Rev. Cancer 2007, 7, 599–612. [Google Scholar] [CrossRef] [PubMed]
  23. Goh, Y.I.; Verjee, Z.; Koren, G. Alcohol content in declared non-to low alcoholic beverages: Implications to pregnancy. Can. J. Clin. Pharmacol. 2010, 17, e47–e50. [Google Scholar] [PubMed]
  24. Kelber, O.; Steinhoff, B.; Nauert, C.; Biller, A.; Adler, M.; Abdel-Aziz, H.; Okpanyi, S.N.; Kraft, K.; Nieber, K. Ethanol in herbal medicinal products for children: Data from pediatric studies and pharmacovigilance programs. Wien. Med. Wochenschr. 2017, 167, 183–188. [Google Scholar] [CrossRef] [Green Version]
  25. Gorgus, E.; Hittinger, M.; Schrenk, D. Estimates of ethanol exposure in children from food not labeled as alcohol-containing. J. Anal. Toxicol. 2016, 40, 537–542. [Google Scholar] [CrossRef] [Green Version]
  26. Jones, A.W. Excretion of low-molecular weight volatile substances in human breath: Focus on endogenous ethanol. J. Anal. Toxicol. 1985, 9, 246–250. [Google Scholar] [CrossRef]
  27. Painter, K.; Cordell, B.J.; Sticco, K.L. Auto-brewery syndrome. In StatPearls; StatPearls Publishing LLC.: Treasure Island, FL, USA, 2022. [Google Scholar]
  28. Hafez, E.M.; Hamad, M.A.; Fouad, M.; Abdel-Lateff, A. Auto-brewery syndrome: Ethanol pseudo-toxicity in diabetic and hepatic patients. Hum. Exp. Toxicol. 2017, 36, 445–450. [Google Scholar] [CrossRef]
  29. Welch, B.T.; Coelho Prabhu, N.; Walkoff, L.; Trenkner, S.W. Auto-brewery syndrome in the setting of long-standing Crohn’s disease: A case report and review of the literature. J. Crohn’s Colitis 2016, 10, 1448–1450. [Google Scholar] [CrossRef] [Green Version]
  30. Malik, F.; Wickremesinghe, P.; Saverimuttu, J. Case report and literature review of auto-brewery syndrome: Probably an underdiagnosed medical condition. BMJ Open Gastroenterol. 2019, 6, e000325. [Google Scholar] [CrossRef]
  31. Vonghia, L.; Leggio, L.; Ferrulli, A.; Bertini, M.; Gasbarrini, G.; Addolorato, G.; Alcoholism Treatment Study, G. Acute alcohol intoxication. Eur. J. Intern. Med. 2008, 19, 561–567. [Google Scholar] [CrossRef]
  32. Bayoumy, A.B.; Mulder, C.J.J.; Mol, J.J.; Tushuizen, M.E. Gut fermentation syndrome: A systematic review of case reports. United Eur. Gastroenterol. J. 2021, 9, 332–342. [Google Scholar] [CrossRef] [PubMed]
  33. Dinis-Oliveira, R.J. The auto-brewery syndrome: A perfect metabolic “storm” with clinical and forensic implications. J. Clin. Med. 2021, 10, 4637. [Google Scholar] [CrossRef] [PubMed]
  34. Pronk, J.T.; Yde Steensma, H.; Van Dijken, J.P. Pyruvate metabolism in Saccharomyces cerevisiae. Yeast 1996, 12, 1607–1633. [Google Scholar] [CrossRef]
  35. 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]
  36. Millet, D.B.; Guenther, A.; Siegel, D.A.; Nelson, N.B.; Singh, H.B.; de Gouw, J.A.; Warneke, C.; Williams, J.; Eerdekens, G.; Sinha, V.; et al. Global atmospheric budget of acetaldehyde: 3-D model analysis and constraints from in-situ and satellite observations. Atmos. Chem. Phys. 2010, 10, 3405–3425. [Google Scholar] [CrossRef] [Green Version]
  37. Singh, H.B.; Salas, L.J.; Chatfield, R.B.; Czech, E.; Fried, A.; Walega, J.; Evans, M.J.; Field, B.D.; Jacob, D.J.; Blake, D.; et al. Analysis of the atmospheric distribution, sources, and sinks of oxygenated volatile organic chemicals based on measurements over the Pacific during TRACE-P. J. Geophys. Res.-Atmos. 2004, 109, D15S07. [Google Scholar] [CrossRef] [Green Version]
  38. Custer, T.; Schade, G. Methanol and acetaldehyde fluxes over ryegrass. Tellus B 2007, 59, 673–684. [Google Scholar] [CrossRef] [Green Version]
  39. Cruz, L.P.S.; Luz, S.R.; Campos, V.P.; Santana, F.O.; Alves, R.S. Determination and risk assessment of formaldehyde and acetaldehyde in the ambient air of gas stations in Salvador, Bahia, Brazil. J. Brazil. Chem. Soc. 2020, 31, 1137–1148. [Google Scholar] [CrossRef]
  40. Hadei, M.; Shahsavani, A.; Hopke, P.K.; Kermani, M.; Yarahmadi, M.; Mahmoudi, B. Comparative health risk assessment of in-vehicle exposure to formaldehyde and acetaldehyde for taxi drivers and passengers: Effects of zone, fuel, refueling, vehicle’s age and model. Environ. Pollut. 2019, 254, 112943. [Google Scholar] [CrossRef]
  41. Naddafi, K.; Nabizadeh, R.; Rostami, R.; Ghaffari, H.R.; Fazlzadeh, M. Formaldehyde and acetaldehyde in the indoor air of waterpipe cafes: Measuring exposures and assessing health effects. Build. Environ. 2019, 165, 106392. [Google Scholar] [CrossRef]
  42. Chang, P.T.; Hung, P.C.; Tsai, S.W. Occupational exposures of flour dust and airborne chemicals at bakeries in Taiwan. J. Occup. Environ. Hyg. 2018, 15, 580–587. [Google Scholar] [CrossRef] [PubMed]
  43. Hadei, M.; Hopke, P.K.; Shahsavani, A.; Moradi, M.; Yarahmadi, M.; Emam, B.; Rastkari, N. Indoor concentrations of VOCs in beauty salons; association with cosmetic practices and health risk assessment. J. Occup. Med. Toxicol. 2018, 13, 30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Karaffa, L.S. The Merck Index: An Encyclopedia of Chemicals, Drugs, and Biologicals; RSC Publishing: London, UK, 2013. [Google Scholar]
  45. Miyake, T.; Shibamoto, T. Quantitative analysis of acetaldehyde in foods and beverages. J. Agric. Food Chem. 1993, 41, 1968–1970. [Google Scholar] [CrossRef]
  46. 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] [Green Version]
  47. Nieminen, M.T.; Salaspuro, M. Local acetaldehyde-An essential role in alcohol-related upper gastrointestinal tract carcinogenesis. Cancers 2018, 10, 11. [Google Scholar] [CrossRef] [Green Version]
  48. Talhout, R.; Opperhuizen, A.; van Amsterdam, J.G. Role of acetaldehyde in tobacco smoke addiction. Eur. Neuropsychopharmacol. 2007, 17, 627–636. [Google Scholar] [CrossRef]
  49. Rabinoff, M.; Caskey, N.; Rissling, A.; Park, C. Pharmacological and chemical effects of cigarette additives. Am. J. Public Health 2007, 97, 1981–1991. [Google Scholar] [CrossRef]
  50. Marttila, E.; Bowyer, P.; Sanglard, D.; Uittamo, J.; Kaihovaara, P.; Salaspuro, M.; Richardson, M.; Rautemaa, R. Fermentative 2-carbon metabolism produces carcinogenic levels of acetaldehyde in Candida albicans. Mol. Oral Microbiol. 2013, 28, 281–291. [Google Scholar] [CrossRef]
  51. Kumamoto, C.A. Inflammation and gastrointestinal Candida colonization. Curr. Opin. Microbiol. 2011, 14, 386–391. [Google Scholar] [CrossRef] [Green Version]
  52. Eram, M.S.; Ma, K. Decarboxylation of pyruvate to acetaldehyde for ethanol production by hyperthermophiles. Biomolecules 2013, 3, 578–596. [Google Scholar] [CrossRef] [Green Version]
  53. Barron, K.A.; Jeffries, K.A.; Krupenko, N.I. Sphingolipids and the link between alcohol and cancer. Chem. Biol. Interact. 2020, 322, 109058. [Google Scholar] [CrossRef] [PubMed]
  54. Jiang, Y.; Zhang, T.; Kusumanchi, P.; Han, S.; Yang, Z.; Liangpunsakul, S. Alcohol metabolizing enzymes, microsomal ethanol oxidizing system, cytochrome P450 2E1, catalase, and aldehyde dehydrogenase in alcohol-associated liver disease. Biomedicines 2020, 8, 50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Norberg, A.; Jones, A.W.; Hahn, R.G.; Gabrielsson, J.L. Role of variability in explaining ethanol pharmacokinetics: Research and forensic applications. Clin. Pharmacokinet. 2003, 42, 1–31. [Google Scholar] [CrossRef] [PubMed]
  56. Jones, A.W. Alcohol, its absorption, distribution, metabolism, and excretion in the body and pharmacokinetic calculations. WIREs Forensic Sci. 2019, 1, 26. [Google Scholar] [CrossRef]
  57. Le Dare, B.; Lagente, V.; Gicquel, T. Ethanol and its metabolites: Update on toxicity, benefits, and focus on immunomodulatory effects. Drug. Metab. Rev. 2019, 51, 545–561. [Google Scholar] [CrossRef]
  58. Estonius, M.; Svensson, S.; Hoog, J.O. Alcohol dehydrogenase in human tissues: Localisation of transcripts coding for five classes of the enzyme. FEBS Lett. 1996, 397, 338–342. [Google Scholar] [CrossRef] [Green Version]
  59. Hartman, J.H.; Miller, G.P.; Meyer, J.N. Toxicological implications of mitochondrial localization of CYP2E1. Toxicol. Res. 2017, 6, 273–289. [Google Scholar] [CrossRef] [Green Version]
  60. Cederbaum, A.I. Alcohol metabolism. Clin. Liver Dis. 2012, 16, 667–685. [Google Scholar] [CrossRef] [Green Version]
  61. Hurley, T.D.; Edenberg, H.J. Genes encoding enzymes involved in ethanol metabolism. Alcohol Res. 2012, 34, 339–344. [Google Scholar]
  62. Edenberg, H.J. The genetics of alcohol metabolism: Role of alcohol dehydrogenase and aldehyde dehydrogenase variants. Alcohol Res. Health 2007, 30, 5–13. [Google Scholar]
  63. Guo, H.; Zhang, G.; Mai, R. Alcohol dehydrogenase-1B Arg47His polymorphism and upper aerodigestive tract cancer risk: A meta-analysis including 24,252 subjects. Alcohol. Clin. Exp. Res. 2012, 36, 272–278. [Google Scholar] [CrossRef] [PubMed]
  64. Homann, N.; Stickel, F.; Konig, I.R.; Jacobs, A.; Junghanns, K.; Benesova, M.; Schuppan, D.; Himsel, S.; Zuber-Jerger, I.; Hellerbrand, C.; et al. Alcohol dehydrogenase 1C*1 allele is a genetic marker for alcohol-associated cancer in heavy drinkers. Int. J. Cancer 2006, 118, 1998–2002. [Google Scholar] [CrossRef] [PubMed]
  65. Chi, Y.C.; Lee, S.L.; Lee, Y.P.; Lai, C.L.; Yin, S.J. Modeling of human hepatic and gastrointestinal ethanol metabolism with kinetic-mechanism-based full-rate equations of the component alcohol dehydrogenase isozymes and allozymes. Chem. Res. Toxicol. 2018, 31, 556–569. [Google Scholar] [CrossRef] [PubMed]
  66. Lee, S.L.; Wang, M.F.; Lee, A.I.; Yin, S.J. The metabolic role of human ADH3 functioning as ethanol dehydrogenase. FEBS Lett. 2003, 544, 143–147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Barnett, S.D.; Buxton, I.L.O. The role of S-nitrosoglutathione reductase (GSNOR) in human disease and therapy. Crit. Rev. Biochem. Mol. 2017, 52, 340–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Oka, Y.; Hamada, M.; Nakazawa, Y.; Muramatsu, H.; Okuno, Y.; Higasa, K.; Shimada, M.; Takeshima, H.; Hanada, K.; Hirano, T.; et al. Digenic mutations in ALDH2 and ADH5 impair formaldehyde clearance and cause a multisystem disorder, AMeD syndrome. Sci. Adv. 2020, 6, eabd7197. [Google Scholar] [CrossRef] [PubMed]
  69. Nadalutti, C.A.; Prasad, R.; Wilson, S.H. Perspectives on formaldehyde dysregulation: Mitochondrial DNA damage and repair in mammalian cells. DNA Repair 2021, 105, 103134. [Google Scholar] [CrossRef]
  70. Dingler, F.A.; Wang, M.; Mu, A.; Millington, C.L.; Oberbeck, N.; Watcham, S.; Pontel, L.B.; Kamimae-Lanning, A.N.; Langevin, F.; Nadler, C.; et al. Two aldehyde clearance systems are essential to prevent lethal formaldehyde accumulation in mice and humans. Mol. Cell 2020, 80, 996–1012.e9. [Google Scholar] [CrossRef]
  71. Hrycay, E.G.; Bandiera, S.M. Cytochrome P450 Enzymes. Preclinical Development Handbook: ADME and Biopharmaceutical Properties; Wiley: Hoboken, NY, USA, 2008; pp. 627–696. [Google Scholar]
  72. Katen, A.L.; Sipila, P.; Mitchell, L.A.; Stanger, S.J.; Nixon, B.; Roman, S.D. Epididymal CYP2E1 plays a critical role in acrylamide-induced DNA damage in spermatozoa and paternally mediated embryonic resorptionsdagger. Biol. Reprod. 2017, 96, 921–935. [Google Scholar] [CrossRef]
  73. Jin, M.; Ande, A.; Kumar, A.; Kumar, S. Regulation of cytochrome P450 2e1 expression by ethanol: Role of oxidative stress-mediated pkc/jnk/sp1 pathway. Cell Death. Dis. 2013, 4, e554. [Google Scholar] [CrossRef] [Green Version]
  74. Harjumaki, R.; Pridgeon, C.S.; Ingelman-Sundberg, M. CYP2E1 in alcoholic and non-alcoholic liver injury. Roles of ROS, reactive intermediates and lipid overload. Int. J. Mol. Sci. 2021, 22, 8221. [Google Scholar] [CrossRef] [PubMed]
  75. Koechling, U.M.; Amit, Z. Relationship between blood catalase activity and drinking history in a human population, a possible biological marker of the affinity to consume alcohol. Alcohol Alcohol. 1992, 27, 181–188. [Google Scholar] [PubMed]
  76. Plemenitas, A.; Kastelic, M.; Porcelli, S.; Serretti, A.; Rus Makovec, M.; Kores Plesnicar, B.; Dolzan, V. Genetic variability in CYP2E1 and catalase gene among currently and formerly alcohol-dependent male subjects. Alcohol Alcohol. 2015, 50, 140–145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Wang, C.D.; Sun, Y.; Chen, N.; Huang, L.; Huang, J.W.; Zhu, M.; Wang, T.; Ji, Y.L. The role of catalase C262T gene polymorphism in the susceptibility and survival of cancers. Sci. Rep. 2016, 6, 26973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Liu, K.; Liu, X.; Wang, M.; Wang, X.; Kang, H.; Lin, S.; Yang, P.; Dai, C.; Xu, P.; Li, S.; et al. Two common functional catalase gene polymorphisms (rs1001179 and rs794316) and cancer susceptibility: Evidence from 14,942 cancer cases and 43,285 controls. Oncotarget 2016, 7, 62954–62965. [Google Scholar] [CrossRef] [Green Version]
  79. Shindo, Y.; Witt, E.; Han, D.; Epstein, W.; Packer, L. Enzymic and non-enzymic antioxidants in epidermis and dermis of human skin. J. Investig. Dermatol. 1994, 102, 122–124. [Google Scholar] [CrossRef] [Green Version]
  80. Wagener, F.A.; Carels, C.E.; Lundvig, D.M. Targeting the redox balance in inflammatory skin conditions. Int. J. Mol. Sci. 2013, 14, 9126–9167. [Google Scholar] [CrossRef]
  81. Rhie, G.E.; Seo, J.Y.; Chung, J.H. Modulation of catalase in human skin in vivo by acute and chronic UV radiation. Mol. Cells 2001, 11, 399–404. [Google Scholar]
  82. Maresca, V.; Flori, E.; Briganti, S.; Mastrofrancesco, A.; Fabbri, C.; Mileo, A.M.; Paggi, M.G.; Picardo, M. Correlation between melanogenic and catalase activity in in vitro human melanocytes: A synergic strategy against oxidative stress. Pigment Cell Melanoma Res. 2008, 21, 200–205. [Google Scholar] [CrossRef]
  83. Schallreuter, K.U.; Wood, J.M.; Berger, J. Low catalase levels in the epidermis of patients with vitiligo. J. Investig. Dermatol. 1991, 97, 1081–1085. [Google Scholar] [CrossRef] [Green Version]
  84. Schallreuter, K.U.; Moore, J.; Wood, J.M.; Beazley, W.D.; Peters, E.M.; Marles, L.K.; Behrens-Williams, S.C.; Dummer, R.; Blau, N.; Thony, B. Epidermal H(2)O(2) accumulation alters tetrahydrobiopterin (6BH4) recycling in vitiligo: Identification of a general mechanism in regulation of all 6BH4-dependent processes? J. Investig. Dermatol. 2001, 116, 167–174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Schallreuter, K.U.; Moore, J.; Wood, J.M.; Beazley, W.D.; Gaze, D.C.; Tobin, D.J.; Marshall, H.S.; Panske, A.; Panzig, E.; Hibberts, N.A. In vivo and in vitro evidence for hydrogen peroxide (H2O2) accumulation in the epidermis of patients with vitiligo and its successful removal by a UVB-activated pseudocatalase. J. Investig. Dermatol. Symp. Proc. 1999, 4, 91–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Rezvani, H.R.; Ged, C.; Bouadjar, B.; de Verneuil, H.; Taieb, A. Catalase overexpression reduces UVB-induced apoptosis in a human xeroderma pigmentosum reconstructed epidermis. Cancer Gene Ther. 2008, 15, 241–251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Dinis-Oliveira, R.J. Oxidative and non-oxidative metabolomics of ethanol. Curr. Drug Metab. 2016, 17, 327–335. [Google Scholar] [CrossRef] [Green Version]
  88. Heier, C.; Xie, H.; Zimmermann, R. Nonoxidative ethanol metabolism in humans-from biomarkers to bioactive lipids. IUBMB Life 2016, 68, 916–923. [Google Scholar] [CrossRef] [Green Version]
  89. Appenzeller, B.M.; Agirman, R.; Neuberg, P.; Yegles, M.; Wennig, R. Segmental determination of ethyl glucuronide in hair: A pilot study. Forensic Sci. Int. 2007, 173, 87–92. [Google Scholar] [CrossRef]
  90. D’Alessandro, A.; Fu, X.; Reisz, J.A.; Stone, M.; Kleinman, S.; Zimring, J.C.; Busch, M.; Recipient Epidemiology and Donor Evaluation Study-III (REDS III). Ethyl glucuronide, a marker of alcohol consumption, correlates with metabolic markers of oxidant stress but not with hemolysis in stored red blood cells from healthy blood donors. Transfusion 2020, 60, 1183–1196. [Google Scholar] [CrossRef]
  91. Paprocki, S.; Qassem, M.; Kyriacou, P.A. Review of ethanol intoxication sensing technologies and techniques. Sensors 2022, 22, 6819. [Google Scholar] [CrossRef]
  92. Lewis, S.S.; Hutchinson, M.R.; Zhang, Y.; Hund, D.K.; Maier, S.F.; Rice, K.C.; Watkins, L.R. Glucuronic acid and the ethanol metabolite ethyl-glucuronide cause toll-like receptor 4 activation and enhanced pain. Brain Behav. Immun. 2013, 30, 24–32. [Google Scholar] [CrossRef] [Green Version]
  93. Seishima, M.; Aoyama, Y.; Mori, S.; Nozawa, Y. Involvement of phospholipase D in ganglioside GQ1b-induced biphasic diacylglycerol production in human keratinocytes. J. Investig. Dermatol. 1995, 104, 835–838. [Google Scholar] [CrossRef] [Green Version]
  94. Hill, R.A.; Xu, W.; Yoshimura, M. Role of an adenylyl cyclase isoform in ethanol’s effect on cAMP regulated gene expression in NIH 3T3 cells. Biochem. Biophys. Rep. 2016, 8, 162–167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Yoshimura, M.; Tabakoff, B. Ethanol’s actions on cAMP-mediated signaling in cells transfected with type VII adenylyl cyclase. Alcohol. Clin. Exp. Res. 1999, 23, 1457–1461. [Google Scholar] [CrossRef] [PubMed]
  96. Torres-Quesada, O.; Mayrhofer, J.E.; Stefan, E. The many faces of compartmentalized PKA signalosomes. Cell Signal. 2017, 37, 1–11. [Google Scholar] [CrossRef] [PubMed]
  97. Seitz, H.K.; Stickel, F. Acetaldehyde as an underestimated risk factor for cancer development: Role of genetics in ethanol metabolism. Genes Nutr. 2010, 5, 121–128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. 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]
  99. Chen, C.H.; Ferreira, J.C.; Gross, E.R.; Mochly-Rosen, D. Targeting aldehyde dehydrogenase 2: New therapeutic opportunities. Physiol. Rev. 2014, 94, 1–34. [Google Scholar] [CrossRef] [Green Version]
  100. Singh, S.; Arcaroli, J.; Thompson, D.C.; Messersmith, W.; Vasiliou, V. Acetaldehyde and retinaldehyde-metabolizing enzymes in colon and pancreatic cancers. Adv. Exp. Med. Biol. 2015, 815, 281–294. [Google Scholar]
  101. Lind, P.A.; Eriksson, C.J.; Wilhelmsen, K.C. The role of aldehyde dehydrogenase-1 (ALDH1A1) polymorphisms in harmful alcohol consumption in a Finnish population. Hum. Genomics 2008, 3, 24–35. [Google Scholar] [CrossRef] [Green Version]
  102. Linneberg, A.; Gonzalez-Quintela, A.; Vidal, C.; Jorgensen, T.; Fenger, M.; Hansen, T.; Pedersen, O.; Husemoen, L.L. Genetic determinants of both ethanol and acetaldehyde metabolism influence alcohol hypersensitivity and drinking behaviour among Scandinavians. Clin. Exp. Allergy 2010, 40, 123–130. [Google Scholar] [CrossRef]
  103. Zhang, Y.; Ren, J. ALDH2 in alcoholic heart diseases: Molecular mechanism and clinical implications. Pharmacol. Ther. 2011, 132, 86–95. [Google Scholar] [CrossRef] [Green Version]
  104. Song, B.J.; Abdelmegeed, M.A.; Yoo, S.H.; Kim, B.J.; Jo, S.A.; Jo, I.; Moon, K.H. Post-translational modifications of mitochondrial aldehyde dehydrogenase and biomedical implications. J. Proteom. 2011, 74, 2691–2702. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Matsumoto, A. The bidirectional effect of defective ALDH2 polymorphism and disease prevention. Adv. Exp. Med. Biol. 2019, 1193, 69–87. [Google Scholar] [PubMed]
  106. Chang, J.S.; Hsiao, J.R.; Chen, C.H. ALDH2 polymorphism and alcohol-related cancers in Asians: A public health perspective. J. Biomed. Sci. 2017, 24, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. 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]
  108. Haschemi Nassab, M.; Rhein, M.; Hagemeier, L.; Kaeser, M.; Muschler, M.; Glahn, A.; Pich, A.; Heberlein, A.; Kornhuber, J.; Bleich, S.; et al. Impaired regulation of ALDH2 protein expression revealing a yet unknown epigenetic impact of rs886205 on specific methylation of a negative regulatory promoter region in alcohol-dependent patients. Eur. Addict. Res. 2016, 22, 59–69. [Google Scholar] [CrossRef]
  109. Pathak, H.; Frieling, H.; Bleich, S.; Glahn, A.; Heberlein, A.; Haschemi Nassab, M.; Hillemacher, T.; Burkert, A.; Rhein, M. Promoter polymorphism rs886205 genotype interacts with DNA methylation of the ALDH2 regulatory region in alcohol dependence. Alcohol Alcohol. 2017, 52, 269–276. [Google Scholar] [CrossRef] [Green Version]
  110. Xue, L.; Xu, F.; Meng, L.; Wei, S.; Wang, J.; Hao, P.; Bian, Y.; Zhang, Y.; Chen, Y. Acetylation-dependent regulation of mitochondrial ALDH2 activation by SIRT3 mediates acute ethanol-induced eNOS activation. FEBS Lett. 2012, 586, 137–142. [Google Scholar] [CrossRef] [Green Version]
  111. Shankarappa, B.; Mahadevan, J.; Murthy, P.; Purushottam, M.; Viswanath, B.; Jain, S.; Devarbhavi, H.; Mysore, A.V. Genetics and epigenetics of aldehyde dehydrogenase (ALDH2) in alcohol related liver disease. medRxiv 2021. [Google Scholar] [CrossRef]
  112. Stewart, M.J.; Dipple, K.M.; Stewart, T.R.; Crabb, D.W. The role of nuclear factor NF-Y/CP1 in the transcriptional regulation of the human aldehyde dehydrogenase 2-encoding gene. Gene 1996, 173, 155–161. [Google Scholar] [CrossRef]
  113. Stewart, M.J.; Dipple, K.M.; Estonius, M.; Nakshatri, H.; Everett, L.M.; Crabb, D.W. Binding and activation of the human aldehyde dehydrogenase 2 promoter by hepatocyte nuclear factor 4. Biochim. Biophys. Acta 1998, 1399, 181–186. [Google Scholar] [CrossRef]
  114. Pinaire, J.; Hasanadka, R.; Fang, M.; Chou, W.Y.; Stewart, M.J.; Kruijer, W.; Crabb, D. The retinoid X receptor response element in the human aldehyde dehydrogenase 2 promoter is antagonized by the chicken ovalbumin upstream promoter family of orphan receptors. Arch. Biochem. Biophys. 2000, 380, 192–200. [Google Scholar] [CrossRef] [PubMed]
  115. You, M.; Fischer, M.; Cho, W.K.; Crabb, D. Transcriptional control of the human aldehyde dehydrogenase 2 promoter by hepatocyte nuclear factor 4: Inhibition by cyclic AMP and COUP transcription factors. Arch. Biochem. Biophys. 2002, 398, 79–86. [Google Scholar] [CrossRef] [PubMed]
  116. Mao, L.; Zuo, M.L.; Hu, G.H.; Duan, X.M.; Yang, Z.B. mir-193 targets ALDH2 and contributes to toxic aldehyde accumulation and tyrosine hydroxylase dysfunction in cerebral ischemia/reperfusion injury. Oncotarget 2017, 8, 99681–99692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Sun, Y.; Wang, Q.; Zhang, Y.; Geng, M.; Wei, Y.; Liu, Y.; Liu, S.; Petersen, R.B.; Yue, J.; Huang, K.; et al. Multigenerational maternal obesity increases the incidence of HCC in offspring via miR-27a-3p. J. Hepatol. 2020, 73, 603–615. [Google Scholar] [CrossRef] [PubMed]
  118. Song, B.J.; Akbar, M.; Abdelmegeed, M.A.; Byun, K.; Lee, B.; Yoon, S.K.; Hardwick, J.P. Mitochondrial dysfunction and tissue injury by alcohol, high fat, nonalcoholic substances and pathological conditions through post-translational protein modifications. Redox Biol. 2014, 3, 109–123. [Google Scholar] [CrossRef] [Green Version]
  119. Zhong, S.; Li, L.; Zhang, Y.L.; Zhang, L.; Lu, J.; Guo, S.; Liang, N.; Ge, J.; Zhu, M.; Tao, Y.; et al. Acetaldehyde dehydrogenase 2 interactions with LDLR and AMPK regulate foam cell formation. J. Clin. Investig. 2019, 129, 252–267. [Google Scholar] [CrossRef] [Green Version]
  120. Zhang, H.; Fu, L. The role of ALDH2 in tumorigenesis and tumor progression: Targeting ALDH2 as a potential cancer treatment. Acta Pharm. Sin. B 2021, 11, 1400–1411. [Google Scholar] [CrossRef]
  121. Zuo, W.; Zhan, Z.; Ma, L.; Bai, W.; Zeng, S. Effect of ALDH2 polymorphism on cancer risk in Asians: A meta-analysis. Medicine 2019, 98, e14855. [Google Scholar] [CrossRef] [PubMed]
  122. Masaoka, H.; Ito, H.; Soga, N.; Hosono, S.; Oze, I.; Watanabe, M.; Tanaka, H.; Yokomizo, A.; Hayashi, N.; Eto, M.; et al. Aldehyde dehydrogenase 2 (ALDH2) and alcohol dehydrogenase 1B (ADH1B) polymorphisms exacerbate bladder cancer risk associated with alcohol drinking: Gene-environment interaction. Carcinogenesis 2016, 37, 583–588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Tsai, S.T.; Wong, T.Y.; Ou, C.Y.; Fang, S.Y.; Chen, K.C.; Hsiao, J.R.; Huang, C.C.; Lee, W.T.; Lo, H.I.; Huang, J.S.; et al. The interplay between alcohol consumption, oral hygiene, ALDH2 and ADH1B in the risk of head and neck cancer. Int. J. Cancer 2014, 135, 2424–2436. [Google Scholar] [CrossRef] [PubMed]
  124. Peng, G.S.; Yin, S.J. Effect of the allelic variants of aldehyde dehydrogenase ALDH2*2 and alcohol dehydrogenase ADH1B*2 on blood acetaldehyde concentrations. Hum. Genomics 2009, 3, 121–127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Koyanagi, Y.N.; Suzuki, E.; Imoto, I.; Kasugai, Y.; Oze, I.; Ugai, T.; Iwase, M.; Usui, Y.; Kawakatsu, Y.; Sawabe, M.; et al. Across-site differences in the mechanism of alcohol-induced digestive tract carcinogenesis: An evaluation by mediation analysis. Cancer. Res. 2020, 80, 1601–1610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Kunitoh, S.; Imaoka, S.; Hiroi, T.; Yabusaki, Y.; Monna, T.; Funae, Y. Acetaldehyde as well as ethanol is metabolized by human CYP2E1. J. Pharmacol. Exp. Ther. 1997, 280, 527–532. [Google Scholar] [PubMed]
  127. Ye, X.; Wang, X.; Shang, L.; Zhu, G.; Su, H.; Han, C.; Qin, W.; Li, G.; Peng, T. Genetic variants of ALDH2-rs671 and CYP2E1-rs2031920 contributed to risk of hepatocellular carcinoma susceptibility in a Chinese population. Cancer Manag. Res. 2018, 10, 1037–1050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Subramaniyan, V.; Chakravarthi, S.; Jegasothy, R.; Seng, W.Y.; Fuloria, N.K.; Fuloria, S.; Hazarika, I.; Das, A. Alcohol-associated liver disease: A review on its pathophysiology, diagnosis and drug therapy. Toxicol. Rep. 2021, 8, 376–385. [Google Scholar] [CrossRef]
  129. Bode, C.; Bode, J.C. Alcohol’s role in gastrointestinal tract disorders. Alcohol Health Res. World 1997, 21, 76–83. [Google Scholar]
  130. Azimian Zavareh, P.; Silva, P.; Gimhani, N.; Atukorallaya, D. Effect of embryonic alcohol exposure on craniofacial and skin melanocyte development: Insights from zebrafish (Danio rerio). Toxics 2022, 10, 544. [Google Scholar] [CrossRef]
  131. Bang, J.; Zippin, J.H. Cyclic adenosine monophosphate (cAMP) signaling in melanocyte pigmentation and melanomagenesis. Pigment Cell Melanoma Res. 2021, 34, 28–43. [Google Scholar] [CrossRef]
  132. Larribere, L.; Utikal, J. NF1-dependent transcriptome regulation in the melanocyte lineage and in melanoma. J. Clin. Med. 2021, 10, 3350. [Google Scholar] [CrossRef]
  133. Johannessen, C.M.; Johnson, L.A.; Piccioni, F.; Townes, A.; Frederick, D.T.; Donahue, M.K.; Narayan, R.; Flaherty, K.T.; Wargo, J.A.; Root, D.E.; et al. A melanocyte lineage program confers resistance to MAP kinase pathway inhibition. Nature 2013, 504, 138–142. [Google Scholar] [CrossRef] [Green Version]
  134. Rodriguez-Zavala, J.S.; Calleja, L.F.; Moreno-Sanchez, R.; Yoval-Sanchez, B. Role of aldehyde dehydrogenases in physiopathological processes. Chem. Res. Toxicol. 2019, 32, 405–420. [Google Scholar] [CrossRef] [PubMed]
  135. Denat, L.; Kadekaro, A.L.; Marrot, L.; Leachman, S.A.; Abdel-Malek, Z.A. Melanocytes as instigators and victims of oxidative stress. J. Investig. Dermatol. 2014, 134, 1512–1518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Kaminski, K.; Kazimierczak, U.; Kolenda, T. Oxidative stress in melanogenesis and melanoma development. Contemp. Oncol. 2022, 26, 1–7. [Google Scholar] [CrossRef] [PubMed]
  137. Cohen, A.D.; Halevy, S. Alcohol intake, immune response, and the skin. Clin. Dermatol. 1999, 17, 411–412. [Google Scholar] [CrossRef] [PubMed]
  138. Brand, R.M.; Stottlemyer, J.M.; Paglia, M.C.; Carey, C.D.; Falo, L.D., Jr. Ethanol consumption synergistically increases ultraviolet radiation induced skin damage and immune dysfunction. J. Dermatol. Sci. 2021, 101, 40–48. [Google Scholar] [CrossRef]
  139. Bertolotto, C.; Lesueur, F.; Giuliano, S.; Strub, T.; de Lichy, M.; Bille, K.; Dessen, P.; d’Hayer, B.; Mohamdi, H.; Remenieras, A.; et al. A SUMOylation-defective MITF germline mutation predisposes to melanoma and renal carcinoma. Nature 2011, 480, 94–98. [Google Scholar] [CrossRef]
  140. Meadows, G.G.; Zhang, H. Effects of alcohol on tumor growth, metastasis, immune response, and host survival. Alcohol Res. 2015, 37, 311–322. [Google Scholar]
  141. Zhang, H.; Zhu, Z.; Meadows, G.G. Chronic alcohol consumption impairs distribution and compromises circulation of B cells in B16BL6 melanoma-bearing mice. J. Immunol. 2012, 189, 1340–1348. [Google Scholar] [CrossRef] [Green Version]
  142. Zhang, H.; Zhu, Z.; Meadows, G.G. Chronic alcohol consumption decreases the percentage and number of NK cells in the peripheral lymph nodes and exacerbates B16BL6 melanoma metastasis into the draining lymph nodes. Cell. Immunol. 2011, 266, 172–179. [Google Scholar] [CrossRef] [Green Version]
  143. Hoyt, L.R.; Randall, M.J.; Ather, J.L.; DePuccio, D.P.; Landry, C.C.; Qian, X.; Janssen-Heininger, Y.M.; van der Vliet, A.; Dixon, A.E.; Amiel, E.; et al. Mitochondrial ROS induced by chronic ethanol exposure promote hyper-activation of the NLRP3 inflammasome. Redox Biol. 2017, 12, 883–896. [Google Scholar] [CrossRef]
  144. Nishiyama, Y.; Goda, N.; Kanai, M.; Niwa, D.; Osanai, K.; Yamamoto, Y.; Senoo-Matsuda, N.; Johnson, R.S.; Miura, S.; Kabe, Y.; et al. HIF-1alpha induction suppresses excessive lipid accumulation in alcoholic fatty liver in mice. J. Hepatol. 2012, 56, 441–447. [Google Scholar] [CrossRef]
  145. D’Aguanno, S.; Mallone, F.; Marenco, M.; Del Bufalo, D.; Moramarco, A. Hypoxia-dependent drivers of melanoma progression. J. Exp. Clin. Cancer Res. 2021, 40, 159. [Google Scholar] [CrossRef] [PubMed]
  146. Wang, Y.; Xu, M.; Ke, Z.J.; Luo, J. Cellular and molecular mechanisms underlying alcohol-induced aggressiveness of breast cancer. Pharmacol. Res. 2017, 115, 299–308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Liu, Y.; Sun, J.; Han, D.; Cui, S.; Yan, X. Identification of potential biomarkers and small molecule drugs for cutaneous melanoma using integrated bioinformatic analysis. Front. Cell Dev. Biol. 2022, 10, 858633. [Google Scholar] [CrossRef]
  148. Sumantran, V.N.; Mishra, P.; Bera, R.; Sudhakar, N. Microarray analysis of differentially-expressed genes encoding CYP450 and phase II drug metabolizing enzymes in psoriasis and melanoma. Pharmaceutics 2016, 8, 4. [Google Scholar] [CrossRef] [Green Version]
  149. Raskin, L.; Fullen, D.R.; Giordano, T.J.; Thomas, D.G.; Frohm, M.L.; Cha, K.B.; Ahn, J.; Mukherjee, B.; Johnson, T.M.; Gruber, S.B. Transcriptome profiling identifies HMGA2 as a biomarker of melanoma progression and prognosis. J. Investig. Dermatol. 2013, 133, 2585–2592. [Google Scholar] [CrossRef] [Green Version]
  150. Riker, A.I.; Enkemann, S.A.; Fodstad, O.; Liu, S.; Ren, S.; Morris, C.; Xi, Y.; Howell, P.; Metge, B.; Samant, R.S.; et al. The gene expression profiles of primary and metastatic melanoma yields a transition point of tumor progression and metastasis. BMC Med. Genomics. 2008, 1, 13. [Google Scholar] [CrossRef] [Green Version]
  151. Kabbarah, O.; Nogueira, C.; Feng, B.; Nazarian, R.M.; Bosenberg, M.; Wu, M.; Scott, K.L.; Kwong, L.N.; Xiao, Y.; Cordon-Cardo, C.; et al. Integrative genome comparison of primary and metastatic melanomas. PLoS ONE 2010, 5, e10770. [Google Scholar] [CrossRef]
  152. Yan, B.Y.; Garcet, S.; Gulati, N.; Kiecker, F.; Fuentes-Duculan, J.; Gilleaudeau, P.; Sullivan-Whalen, M.; Shemer, A.; Mitsui, H.; Krueger, J.G. Novel immune signatures associated with dysplastic naevi and primary cutaneous melanoma in human skin. Exp. Dermatol. 2019, 28, 35–44. [Google Scholar] [CrossRef]
  153. Cichorek, M.; Wachulska, M.; Stasiewicz, A.; Tyminska, A. Skin melanocytes: Biology and development. Postep. Dermatol. Alergol. 2013, 30, 30–41. [Google Scholar] [CrossRef]
  154. Reemann, P.; Reimann, E.; Ilmjarv, S.; Porosaar, O.; Silm, H.; Jaks, V.; Vasar, E.; Kingo, K.; Koks, S. Melanocytes in the skin--comparative whole transcriptome analysis of main skin cell types. PLoS ONE 2014, 9, e115717. [Google Scholar] [CrossRef] [Green Version]
  155. Luo, Y.; Dallaglio, K.; Chen, Y.; Robinson, W.A.; Robinson, S.E.; McCarter, M.D.; Wang, J.; Gonzalez, R.; Thompson, D.C.; Norris, D.A.; et al. ALDH1A isozymes are markers of human melanoma stem cells and potential therapeutic targets. Stem Cells 2012, 30, 2100–2113. [Google Scholar] [CrossRef] [Green Version]
  156. Mameishvili, E.; Serafimidis, I.; Iwaszkiewicz, S.; Lesche, M.; Reinhardt, S.; Bolicke, N.; Buttner, M.; Stellas, D.; Papadimitropoulou, A.; Szabolcs, M.; et al. Aldh1b1 expression defines progenitor cells in the adult pancreas and is required for Kras-induced pancreatic cancer. Proc. Natl. Acad. Sci. USA 2019, 116, 20679–20688. [Google Scholar] [CrossRef] [Green Version]
  157. Feng, Z.; Hom, M.E.; Bearrood, T.E.; Rosenthal, Z.C.; Fernandez, D.; Ondrus, A.E.; Gu, Y.; McCormick, A.K.; Tomaske, M.G.; Marshall, C.R.; et al. Targeting colorectal cancer with small-molecule inhibitors of ALDH1B1. Nat. Chem. Biol. 2022, 18, 1065–1075. [Google Scholar] [CrossRef]
  158. Chang, P.M.; Chen, C.H.; Yeh, C.C.; Lu, H.J.; Liu, T.T.; Chen, M.H.; Liu, C.Y.; Wu, A.T.H.; Yang, M.H.; Tai, S.K.; et al. Transcriptome analysis and prognosis of ALDH isoforms in human cancer. Sci. Rep. 2018, 8, 2713. [Google Scholar] [CrossRef] [Green Version]
  159. Ma, B.; Liu, Z.; Xu, H.; Liu, L.; Huang, T.; Meng, L.; Wang, L.; Zhang, Y.; Li, L.; Han, X. Molecular characterization and clinical relevance of ALDH2 in human cancers. Front. Med. 2021, 8, 832605. [Google Scholar] [CrossRef]
  160. Wu, L.; Dong, B.; Zhang, F.; Li, Y.; Liu, L. Prediction of the engendering mechanism and specific genes of primary melanoma by bioinformatics analysis. Dermatol. Sin. 2016, 34, 6. [Google Scholar] [CrossRef] [Green Version]
  161. Dimitriou, F.; Krattinger, R.; Ramelyte, E.; Barysch, M.J.; Micaletto, S.; Dummer, R.; Goldinger, S.M. The world of melanoma: Epidemiologic, genetic, and anatomic differences of melanoma across the globe. Curr. Oncol. Rep. 2018, 20, 87. [Google Scholar] [CrossRef]
Figure 1. Schematic overview of the major factors leading to human skin exposure to ethanol and/or acetaldehyde. ACR, acetyl-CoA reductase; PDC, pyruvate decarboxylase; PFL, pyruvate formate-lyase; POR, pyruvate ferredoxin oxidoreductase.
Figure 1. Schematic overview of the major factors leading to human skin exposure to ethanol and/or acetaldehyde. ACR, acetyl-CoA reductase; PDC, pyruvate decarboxylase; PFL, pyruvate formate-lyase; POR, pyruvate ferredoxin oxidoreductase.
Cancers 15 01258 g001
Figure 2. Schematic overview of ethanol metabolism. Ethanol can be metabolized by oxidative (a) or non-oxidative (b) pathways. ADH, alcohol dehydrogenase; ALDH, aldehyde dehydrogenase; CAT, catalase; CYP2E1, cytochrome P450 2E1; EtG, ethyl glucuronide; EtS, ethyl sulfate; FAEE, fatty acid ethyl ester; FAEES, fatty acid ethyl ester synthase; PEth, phosphatidyl ethanol; PLD, phospholipase D; SULT, sulfotransferase; UGT, UDP-glucuronosyltransferase. Modified from [9].
Figure 2. Schematic overview of ethanol metabolism. Ethanol can be metabolized by oxidative (a) or non-oxidative (b) pathways. ADH, alcohol dehydrogenase; ALDH, aldehyde dehydrogenase; CAT, catalase; CYP2E1, cytochrome P450 2E1; EtG, ethyl glucuronide; EtS, ethyl sulfate; FAEE, fatty acid ethyl ester; FAEES, fatty acid ethyl ester synthase; PEth, phosphatidyl ethanol; PLD, phospholipase D; SULT, sulfotransferase; UGT, UDP-glucuronosyltransferase. Modified from [9].
Cancers 15 01258 g002
Figure 3. Gene expression of ethanol metabolizing enzymes in normal human skin, melanocytic nevi, and melanoma. Ethanol metabolizing enzyme genes include ADH1A (a), ADH1B (b), ADH1C (c), ADH4 (d), ADH5 (e), ADH6 (f), ADH7 (g), CYP2E1 (h), and CAT (i). Data from four independent gene profiling studies were analyzed: GSE15605 (16 normal skin (NS) samples, 46 primary melanoma (PM) samples, and 12 metastatic melanoma (MM) samples [149]), GSE7553 (4 NS, 14 PM, and 40 MM [150]), GSE46517 (7 NS, 31 PM, and 73 MM [151]), and GSE114445 (6 NS, 5 common nevus (CN) samples, 7 dysplastic nevus (DN) samples, and 16 PM [152]). In these datasets, if two or more probes were used for a certain gene, the gene expression values of these probes per patient were averaged since they used the same controls. Data are shown as scatter dot plot with median and interquartile range (IQR). * p < 0.05, ** p < 0.01, *** p < 0.001, and **** p < 0.0001 (Kruskal–Wallis’s non-parametric test (GSE15605, GSE7553, and GSE46517) and nonparametric estimation of Spearman’s rank correlation (GSE114445)). n/a, not available.
Figure 3. Gene expression of ethanol metabolizing enzymes in normal human skin, melanocytic nevi, and melanoma. Ethanol metabolizing enzyme genes include ADH1A (a), ADH1B (b), ADH1C (c), ADH4 (d), ADH5 (e), ADH6 (f), ADH7 (g), CYP2E1 (h), and CAT (i). Data from four independent gene profiling studies were analyzed: GSE15605 (16 normal skin (NS) samples, 46 primary melanoma (PM) samples, and 12 metastatic melanoma (MM) samples [149]), GSE7553 (4 NS, 14 PM, and 40 MM [150]), GSE46517 (7 NS, 31 PM, and 73 MM [151]), and GSE114445 (6 NS, 5 common nevus (CN) samples, 7 dysplastic nevus (DN) samples, and 16 PM [152]). In these datasets, if two or more probes were used for a certain gene, the gene expression values of these probes per patient were averaged since they used the same controls. Data are shown as scatter dot plot with median and interquartile range (IQR). * p < 0.05, ** p < 0.01, *** p < 0.001, and **** p < 0.0001 (Kruskal–Wallis’s non-parametric test (GSE15605, GSE7553, and GSE46517) and nonparametric estimation of Spearman’s rank correlation (GSE114445)). n/a, not available.
Cancers 15 01258 g003aCancers 15 01258 g003b
Figure 4. Gene expression of acetaldehyde (AcAH) metabolizing enzymes in normal human skin, melanocytic nevi, and melanoma. AcAH metabolizing enzyme genes include ALDH1A1 (a), ALDH1B1 (b), and ALDH2 (c). See Figure 3 for CYP2E1. Data from four independent gene profiling studies were analyzed as described in Figure 3: GSE15605 (16 normal skin (NS) samples, 46 primary melanoma (PM) samples, and 12 metastatic melanoma (MM) samples [149]), GSE7553 (4 NS, 14 PM, and 40 MM [150]), GSE46517 (7 NS, 31 PM, and 73 MM [151]), and GSE114445 (6 NS, 5 common nevus (CN) samples, 7 dysplastic nevus (DN) samples, and 16 PM [152]). Data are shown as scatter dot plot with median and interquartile range (IQR). * p < 0.05, ** p < 0.01, and **** p < 0.0001 (Kruskal–Wallis’s non-parametric test (GSE15605, GSE7553, and GSE46517) and nonparametric estimation of Spearman’s rank correlation (GSE114445)).
Figure 4. Gene expression of acetaldehyde (AcAH) metabolizing enzymes in normal human skin, melanocytic nevi, and melanoma. AcAH metabolizing enzyme genes include ALDH1A1 (a), ALDH1B1 (b), and ALDH2 (c). See Figure 3 for CYP2E1. Data from four independent gene profiling studies were analyzed as described in Figure 3: GSE15605 (16 normal skin (NS) samples, 46 primary melanoma (PM) samples, and 12 metastatic melanoma (MM) samples [149]), GSE7553 (4 NS, 14 PM, and 40 MM [150]), GSE46517 (7 NS, 31 PM, and 73 MM [151]), and GSE114445 (6 NS, 5 common nevus (CN) samples, 7 dysplastic nevus (DN) samples, and 16 PM [152]). Data are shown as scatter dot plot with median and interquartile range (IQR). * p < 0.05, ** p < 0.01, and **** p < 0.0001 (Kruskal–Wallis’s non-parametric test (GSE15605, GSE7553, and GSE46517) and nonparametric estimation of Spearman’s rank correlation (GSE114445)).
Cancers 15 01258 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhai, Z.; Yamauchi, T.; Shangraw, S.; Hou, V.; Matsumoto, A.; Fujita, M. Ethanol Metabolism and Melanoma. Cancers 2023, 15, 1258. https://doi.org/10.3390/cancers15041258

AMA Style

Zhai Z, Yamauchi T, Shangraw S, Hou V, Matsumoto A, Fujita M. Ethanol Metabolism and Melanoma. Cancers. 2023; 15(4):1258. https://doi.org/10.3390/cancers15041258

Chicago/Turabian Style

Zhai, Zili, Takeshi Yamauchi, Sarah Shangraw, Vincent Hou, Akiko Matsumoto, and Mayumi Fujita. 2023. "Ethanol Metabolism and Melanoma" Cancers 15, no. 4: 1258. https://doi.org/10.3390/cancers15041258

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