Next Article in Journal
Reversal of Hyperglycemia and Suppression of Type 1 Diabetes in the NOD Mouse with Apoptotic DNA Immunotherapy™ (ADi™), ADi-100
Next Article in Special Issue
Alcoholic-Hepatitis, Links to Brain and Microbiome: Mechanisms, Clinical and Experimental Research
Previous Article in Journal
Effects of an 8-Week Protein Supplementation Regimen with Hyperimmunized Cow Milk on Exercise-Induced Organ Damage and Inflammation in Male Runners: A Randomized, Placebo Controlled, Cross-Over Study
Previous Article in Special Issue
Global Burden of Alcohol Use Disorders and Alcohol Liver Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Alcohol Metabolizing Enzymes, Microsomal Ethanol Oxidizing System, Cytochrome P450 2E1, Catalase, and Aldehyde Dehydrogenase in Alcohol-Associated Liver Disease

1
Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
2
Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, USA
*
Authors to whom correspondence should be addressed.
Biomedicines 2020, 8(3), 50; https://doi.org/10.3390/biomedicines8030050
Submission received: 24 January 2020 / Revised: 20 February 2020 / Accepted: 29 February 2020 / Published: 4 March 2020
(This article belongs to the Special Issue Alcoholic Liver Disease: Diagnostics and Therapeutics)

Abstract

:
Once ingested, most of the alcohol is metabolized in the liver by alcohol dehydrogenase to acetaldehyde. Two additional pathways of acetaldehyde generation are by microsomal ethanol oxidizing system (cytochrome P450 2E1) and catalase. Acetaldehyde can form adducts which can interfere with cellular function, leading to alcohol-induced liver injury. The variants of alcohol metabolizing genes encode enzymes with varied kinetic properties and result in the different rate of alcohol elimination and acetaldehyde generation. Allelic variants of these genes with higher enzymatic activity are believed to be able to modify susceptibility to alcohol-induced liver injury; however, the human studies on the association of these variants and alcohol-associated liver disease are inconclusive. In addition to acetaldehyde, the shift in the redox state during alcohol elimination may also link to other pathways resulting in activation of downstream signaling leading to liver injury.

1. Introduction

Alcohol associated liver disease (ALD) is one of the major causes of chronic liver disease worldwide [1,2]. The risk of ALD development is known to be associated with the quantity of alcohol (beverages containing ethanol) consumed [3,4]. Early studies suggested that a long-term consumption of more than 80 g per day was correlated with increased risk of cirrhosis, but subsequent estimates of the harmful drinking threshold have been below this level, especially for women [5,6]. In another study, 13,285 subjects were prospectively followed for 12 years to determine the association between the risk for development of future liver disease and self-reported alcohol consumption [7]. The patients were asked in multiple-choice form on the type of alcoholic beverages they consumed, wine (glasses), beer (bottles), or spirits (units) and frequency (i.e., daily, weekly, monthly, and hardly/never). If daily alcohol intake was reported, the average number of beverages of each type per week was recorded. In this study, one beverage equals to 12 g of alcohol [7]. There was no apparent risk of alcohol-induced liver injury if the range of alcohol consumption was between 1 to 6 beverages per week (12–72 g per week); however, drinking beyond this level (7 to 13 beverages (84–156 g) per week for women and 14 to 27 beverages (168–324 g) per week for men) resulted in a significant rise in the relative risk of alcohol associated liver disease [7]. In addition to the quantity of alcohol consumption, studies also suggested that the risk of ALD may also depend on the patterns and types of alcohol intake independently of the level of alcohol consumption [8]. Red wine drinkers may have a lower risk of ALD compared to other beverage drinkers [9]. It is currently unknown whether this is due to an effect of the red wine per se or to confounding protective lifestyle factors. Of importance, it is difficult to systematically study the relationship between the quantity of alcohol consumption and the development of ALD because data collection always involves rough estimate, differences in drinking patterns among studies, and inconsistencies in data collection tools. ALD represents a range of pathological changes ranging from steatosis, alcoholic steatohepatitis, advanced fibrosis, and cirrhosis [1,10]. Alcoholic steatosis can occur in patients who consumed 120–150 g of alcohol per day for 2–3 weeks [11]. Most cases develop macrovesicular steatosis primarily in zone 3 or throughout the liver in severe cases [12]. This condition is reversible upon abstinence [13]. Hepatic steatosis was thought to be a benign condition; however, a subset of patients can progress to fibrosis or cirrhosis in 10 years, especially if the drinking continues [14].
Only a group of patients with excessive alcohol use develop alcoholic hepatitis, a severe manifestation of ALD with high morbidity and mortality [15,16,17]. Approximately, 10% to 20% of patients with alcoholic hepatitis are likely to progress to cirrhosis annually, and up to 70% of patients with alcoholic hepatitis ultimately will become cirrhotic [12]. Abstinence is essential and has the impact on the survival [15,18]. Overall, 15% to 20% of excessive drinkers will progress to advanced liver disease and cirrhosis [19]. Once developed, patients with alcoholic cirrhosis have clinical presentations and complications similar to those with other types of cirrhosis. Of importance, mortality due to alcoholic cirrhosis has been shifted towards a younger population between 25 and 34 years old [20]. There are also important gender differences in ALD pathogenesis. At any given level of alcohol consumption, women have a higher likelihood of developing ALD than men [5]. The mechanism is likely due to lower volume of distribution in women leading to higher blood alcohol concentrations per unit of alcohol consumed [21]. Additionally, recent studies also showed the role of hormone like estrogen which can increase gut permeability and endotoxemia leading to the increase in the production of inflammatory cytokines by Kupffer cells and risk of ALD [22]. Some of the enzymes involved in alcohol metabolism are genetically polymorphic leading to the alteration in enzymatic properties and varying amounts of acetaldehyde generation after alcohol consumption [23]. The roles of these enzymes in association with ALD are the subject of this overview.

2. Alcohol Metabolism

When alcohol is ingested, a small amount is immediately metabolized in the stomach. Most of the remaining alcohol is subsequently absorbed from the gastrointestinal tract, primarily the stomach and upper small intestine [24]. After the absorption, alcohol is transported to the liver through the portal vein, where the alcohol concentration is significantly higher than that measured from the peripheral blood [25]. Absorption rate of ethanol is a first-order kinetic process in proportion to the concentration of alcohol in the stomach. Once the gastric alcohol concentration decreases, the rate of absorption slows down [26]. After the initial passage through the liver; the remainder of the ingested alcohol enters systemic circulation and is distributed throughout the body’s tissues. Once the rate of absorption is equal to the rate of alcohol metabolism (approximately 10–30 mg% per hour), the peripheral blood alcohol concentration reaches a maximum point and starts to decline as alcohol is eliminated from the body [27].
Liver is the major organ which metabolizes more than 90% of ingested alcohol into acetaldehyde through several enzymatic and non-enzymatic mechanisms [28,29]. Many of the effects of ethanol are mediated by its byproduct acetaldehyde, which is mainly generated by alcohol dehydrogenase (ADH), cytochrome P450 2E1 (CYP2E1), and catalase (Figure 1). Acetaldehyde is subsequently converted to acetate by aldehyde dehydrogenases (ALDH), which is released from the liver and metabolized by muscle and heart [29]. The rate of ethanol metabolism by alcohol metabolizing enzymes may be critical in determining its toxicity because of the toxic intermediates.

3. Alcohol Metabolizing Enzymes

3.1. Alcohol Dehydrogenase (ADH)

ADH is a zinc-dependent enzyme located in the cytosol. It uses NAD+ as a cofactor and is responsible for the majority of alcohol oxidation in the liver [30,31]. Human ADHs are categorized into five classes based on their structural and kinetic characteristics [23,29]. These enzymes are dimeric and identified by Greek letters with a molecular weight of 40kDa per subunit [23,29]. Under physiological conditions, the main isoenzymes involved in human ethanol metabolism are ADHs from classes I, II, and IV [23,29].
Class I ADH contains α, β, and γ isoenzymes. These enzymes have a low Michaelis constant (Km) < 5 mM for ethanol and therefore play a significant role in ethanol metabolism [23]. Class I ADH is highly abundant in the liver and is sensitive to inhibition by pyrazole derivatives [32]. Class I ADH consists of three genes, ADH1A, ADH1B, and ADH1C, making up gene clusters located on chromosome 4q21-23, spanning about 80 kb [33]. Among these genes, polymorphisms with physiological significance exist in the ADH1B and ADH1C loci [23]. These variants result in the difference in their efficiency for ethanol oxidation. People of different racial backgrounds inherit different sets of ADH isoenzymes [29,34,35]. African Americans who carry a copy of the ADH1B*3 gene, encoding an enzyme with high maximum velocity, have a somewhat faster alcohol metabolism rate [36,37]. Class II ADH (π ADH) has a higher Km for ethanol and is less sensitive to pyrazole inhibition than class I enzymes [38]. Because of its Km, its role in ethanol metabolism is less than that of Class I ADH [39]. Class III ADH is ubiquitous and it can metabolize longer chain alcohols as well as ω-oxidation of fatty acid [40]. The class IV ADH (µ-ADH, for the mucosal isozyme or σ-ADH, for the stomach isoenzyme) has been found in the esophagus and stomach and is responsible for alcohol first pass metabolism [41]. Among ADHs, σ-ADH has the highest Vmax and is very active towards retinol. This may be pertinent to its expression at the epithelia, which are dependent on retinol for their integrity [42]. The kinetic properties of each ADH class, population and tissue distribution are shown in Table 1.
Tissue distribution of ADH: ADH mRNAs are expressed in a variety of tissues with high levels of class I ADH in the liver, stomach, kidney, and colon, and with lower levels in lung, small intestine, brain, and muscle [43]. Of importance, class I ADH is also found in blood vessels, which may be linked to alcohol-induced flushing. Class II ADH was found in the liver and duodenum [43]. σ-ADH is primarily found in the gastric mucosa, though its expression is absent in approximately one-third of the Asians population [44]. Microorganisms can produce alcohol [45,46] and express numerous forms of alcohol dehydrogenase, which can contribute to the formation of acetaldehyde in the gastrointestinal tract, or wherever microbial overgrowth occurs [47,48].
Regulation of ADH expression: The promotors of hepatic ADH1 can interact with ubiquitous transcription factors (i.e., upstream stimulatory factor [USF], TATAA binding factors and CAAT box-binding transcription factor/nuclear factor-1 [CTF/NF-I]), and tissue-specific factors (i.e., CCAAT-enhancer binding proteins [C/EBPα and β] hepatocyte nuclear factor 1 [HNF-1], and D-box binding protein [DBP]) [49,50,51,52]. An HNF-1 site was found to serve as a key regulator for all three class I genes [53]. Upstream regions of ADH1 genes are also found to be the binding sites for retinoic acid, glucocorticoid and thyroid hormones [23,54,55,56]. Hepatic ADH activity also depends on the route of alcohol administration. While the quantity of ethanol consumed from liquid diets did not modify hepatic ADH activity, higher doses of ethanol administered by intragastric infusion induced hepatic ADH activity due to the induction of the transcription factor C/EBPβ and suppression of C/EBPγ and a truncated, inhibitory form of C/EBPβ [57]. Additionally, intragastric ethanol infusion can induce hepatic Class I ADH mRNA, protein, and activity levels by 3.9-, 3.3-, and 1.7-fold, respectively through transcription factor USF due to the increase in endotoxin in the portal system [57,58].
Level of alcohol concentration and ADH: Because of the low Km of Class I ADH, this enzyme is saturated with any types of drinking patterns once blood alcohol concentration >15–20 mg% [27]. While class I ADH with low Km for ethanol is active even at a low alcohol concentration, those with high Km (i.e., σ-ADH) are more active when blood ethanol concentrations are high or in tissues of the upper gastrointestinal tract that are directly exposed to beverage ethanol. Modelling of ethanol oxidation indicated that ethanol metabolism by ADH is regulated by the quantity of ADH enzymes, the byproduct acetaldehyde, and the NADH to NAD+ ratio in the cytosol [59,60].

3.2. Microsomal Ethanol Oxidizing System

In addition to ADH, ethanol can be oxidized by a microsomal ethanol oxidizing system (MEOS) involving primarily the cytochrome P450 2E1 (CYP2E1) [61]. Two other cytochromes contribute to the MEOS, albeit to a lesser extent, CYP1A2 and CYP3A4 [62,63,64]. CYP2E1 is associated with NADPH-cytochrome P450 reductase in the endoplasmic reticulum and reduces oxygen molecule to water when ethanol is oxidized to acetaldehyde [61]. It has a low ethanol catalytic efficiency with the Km for ethanol ~10 mM when compared to ADH and therefore is responsible for a minor role and accounting for approximately 10% of ethanol metabolism [65]. However, CYP2E1 may assume a greater role at high blood alcohol concentrations. The levels of CYP2E1 are induced by chronic alcohol consumption (more detail in section below) notably in the perivenular zone leading to the increase in alcohol elimination rate among excessive drinkers [23]. In addition, it is induced in diabetes, high fat diet, and fasting, which may relate to its ability to oxidize the ketone body acetone [66,67,68,69]. CYP2E1 is a major contributor of oxidative stress in the hepatocytes by generating several reactive oxygen species (ROS) such as hydrogen peroxide (H2O2), hydroxyethyl radical (HER·), hydroxyl radical (OH) and superoxide (O2) [64,67].
Regulation of CYP2E1 expression: The human CYP2E1 gene spans 11 kb, contains 9 exons, and contains a typical TATAA box [70]. Its expression is regulated at the transcriptional (mRNA) level by ethanol concentration [71] and at the post translational level by protein stabilization and reducing the proteasomal degradation [64]. The expression of CYP2E1 can also be induced by cytokines such as interleukin-4 [72]. CYP2E1 can also be regulated by hsa-miR-214–3p [73]. In a HepG2-derived CYP2E1 over-expression cell model, hsa-miR-214-3p showed strong suppression of CYP2E1 expression by targeting the coding region of its mRNA transcript [73]. Treatment with hsa-miR-214-3p mimics partially blocked ethanol-dependent increases in CYP2E1 mRNA and protein levels in HepG2 cells [73].
Level of alcohol concentration and CYP2E1: Since CYP2E1 has a high Km for ethanol, it will generate acetaldehyde at a high level of ethanol concentrations [23]. CYP2E1 plays a more important role in alcohol metabolism when high blood concentration is reached. Another important aspect of CYP2E1 is that binge drinking can induce its activity, making it more effective in alcohol metabolism at its higher blood concentration [74]. Additionally, the increase in enzyme activity is the underlying mechanism to explain faster alcohol clearance among excessive drinkers [63]. Unlike ADH, it does not appear that CYP2E1 is inhibited by acetaldehyde; in fact, CYP2E1 can oxidize acetaldehyde to acetate, but not in the presence of ethanol [23]. Because CYP2E1 also metabolizes several medications, excessive drinkers with the induction in CYP2E1 activity will demonstrate increased metabolic rates for those medications which may lead to adverse consequences [75].

3.3. Catalase

The peroxisomal catalase, a tetrameric, heme-containing enzyme, converts hydrogen peroxide (H2O2) to oxygen and water [76]. Catalase can also oxidize ethanol to acetaldehyde as the end product in an H2O2-dependent fashion, though this is not a key pathway for ethanol elimination [23,77].
Tissue distribution of catalase: Catalase is ubiquitously expressed in almost all tissues. Catalase is also expressed by colonic floras which may lead to acetaldehyde production in the lower gastrointestinal tract [78,79].
Regulation of catalase expression: In rodents, catalase gene is a single-copy gene with 33 kb. The promoter region of this gene lacks a TATAA box and an initiator consensus sequence [23]. It contains multiple transcription initiation sites as well as multiple CCAAT boxes and GC boxes [23]. The increase in catalase activity was reported after chronic ethanol feeding [80]. The rat catalase promoter contains a peroxisome proliferator responsive element (PPRE), therefore it can be induced by peroxisome proliferators [81].
Level of alcohol concentration and catalase: Catalase activity relies on the cellular level of H2O2. The ability of catalase to metabolize ethanol may be increased under oxidative stress with the increase in cellular H2O2 production [82]. This was observed with perfused rat liver, when fatty acids were added and the process of peroxisomal β oxidation lead to the generation of H2O2 and oxidation of ethanol. This raises the possibility that under conditions of oxidant stress (and H2O2 production) catalase-mediated ethanol oxidation may be increased [23].

3.4. Aldehyde Dehydrogenase (ALDH)

The second step of ethanol metabolism is the process to convert acetaldehyde to acetate by alcohol dehydrogenase (ALDH). ALDH enzymes use NAD+ as a cofactor during the oxidization of acetaldehyde but are expressed in a wider range of tissues than the ADH isoenzymes [83]. In addition to its role in elimination of acetaldehyde, ALDH also plays a critical role in the detoxification of lipid peroxidation byproduct such as 4-hydroxynonenal (4-HNE) which is generated during oxidative stress [84]. There are two major ALDH isoforms, cytosolic and mitochondrial, encoded by the aldehyde ALDH1 and ALDH2 genes, respectively. Of the two isoforms, mitochondrial ALDH2 plays the central role in human acetaldehyde metabolism because of its submicromolar Km for acetaldehyde [29]. Tissue distribution and Km for acetaldehyde of human ALDHs are shown in Table 2.
Regulation of ALDH2 expression: ALDH2 expression can be regulated at the transcriptional level [85]. ALDH2 gene promoter contains a nuclear receptor response element that is bound by retinoic acid receptors, peroxisome proliferator activated receptors (PPARs), hepatocyte nuclear factor 4 (HNF4), and members of the chicken ovalbumin upstream promoter transcription factor (COUP-TF) family [85,86,87]. ALDH2 activity can also be modified through post-translational regulation by serine/threonine phosphorylation through epsilon protein kinase C (εPKC) [84]. Additionally, the activation of ALDH2 by ethanol has been shown to be mediated by epigenetic regulation through hyperacetylation of ALDH2 gene by sirtuin 3 (SIRT3) inactivation [88].

3.5. Other Minor Pathways for the Generation of Acetaldehyde

Cytosolic fractions of xanthine oxidoreductase such as hypoxanthine, xanthine, and caffeine can oxidize ethanol to acetaldehyde [89]. This process can be inhibited by allopurinol and not by pyrazole [89]. Ethanol can also be metabolized to acetaldehyde and 1-hydroxyethyl radical by cytochrome P450 reductase [90]. There are other reports that other oxidant species (hydroxyl radical) formed non-enzymatically might be able to oxidize ethanol to acetaldehyde. In addition, acetaldehyde can be formed during the degradation of threonine, putatively by threonine aldolase [23,29].

4. Genetic Polymorphisms of Alcohol-Metabolizing Enzymes and ALD

ADH: Because of the differences in the enzymatic activity to metabolize alcohol to acetaldehyde, it has been postulated that those with alleles encoding for higher enzymatic activity such as ADH1B*2 and ADH1C*1 alleles, are at increased risk of developing ALD and modifying the risk for alcohol dependence due to higher acetaldehyde exposure at any given alcohol concentration [91,92]. ADH1B*2 allele has been shown to reduce the occurrence of alcohol abuse especially in population with high prevalence such as Jewish and Asian populations [29]. Several other studies also found similar results on the higher frequency of ADH1B*2 among moderate or non-drinkers compared to heavy drinkers [93,94,95]. The reason behind these findings is likely due to the development of unpleasant side effects associated with the faster accumulation of acetaldehyde after alcohol consumption [96], though the levels of venous acetaldehyde concentration in those with and without this allele have not previously shown to be different [97,98]. A recent study from Japan found that those with slow metabolizing ADH1B enzyme increase susceptibility to hepatic steatosis in men with alcohol dependence [99]. However, to date, the results on the association between genetic variants of ADH and ALD have been inconsistent, owing to the difference in the ethnicities of the study cohorts and limitations in sample size, as well as the low allele frequency of ADH1B*2 among Caucasians [92].
CYP2E1: The CYP2E1 c1 allele is located in the 5’-flanking region of the CYP2E1 gene while in vitro transcriptional assays showed the c2 allele to be more active enzyme [100,101]. The allele frequency of CYP2E1 variants is varied depending on ethnicity, the c2 variant is found at a higher frequency in East Asians (~25–35%) than that in Caucasians (~2–8%) [23,102]. In contrast to ADH, CYP2E1 is inducible and its activity can increase significantly after alcohol consumption [23], causing a higher level of reactive oxygen species and acetaldehyde [103]. The allele frequency of c2 variant is higher than that of c1 in patients with ALD, compared with random population controls [92]. However, the association between CYP2E1 variants and ALD is inconclusive [92].
Catalase: Transcription of the catalase gene is influenced by the -262 C/T and -844 A/G polymorphisms. A common polymorphism -262 C/T in the promoter region has been found to be associated with altered catalase activities and is associated with the susceptibility to alcohol dependence [104,105]. To date, there is no convincing evidence on the role of catalase polymorphism and ALD.
ALDH: Similar to ADH and CYP2E1 variants, polymorphisms in the ALDH2 demonstrate a vital role in regulating ALDH2 activity and are hypothesized to alter genetic susceptibility to alcohol dependence and alcohol-induced liver diseases. A variant at exon 12 predicts lysine at residue 504 instead of glutamic acid [106]. The common form of the SNP (rs671) (504glu) encodes the glu (G) allele (previously referred to as the ALDH2 *1 allele); the 504lys (A, formerly ALDH2 *2 and 487lys) allele produces an inactive isozyme and thus limits its activity to convert acetaldehyde into acetate [106,107]. Individuals with the lys allele tend to have the accumulation of acetaldehyde after alcohol ingestion with the peak acetaldehyde concentration approximately 6- to 19-fold higher in those with heterozygotes or homozygotes for 504lys allele than those with common allele [97]. These patients commonly experience unpleasant side effects such as nausea, vomiting, or flushing after alcohol ingestion [108,109]. ALDH2 is inhibited by disulfiram (Antabuse) and consumption of alcohol while taking disulfiram or a number of other drugs such as metronidazole results in the accumulation of acetaldehyde, producing vasodilation, facial flushing, tachycardia, nausea, and vomiting [37]. Variants in genes encoding ALDH2 are important determinants of risk for alcohol abuse or dependence and to a lesser extent for cirrhosis, especially in Asian populations [92]. Previous studies have shown that the inactive ALDH2 504lys allele occurred mainly in Asians [29,37]. Most patients with alcohol abuse or dependence with inactive ALDH2 alleles were heterozygous while homozygous lys/lys was rarely found, although it was often observed in controls [106]. These findings are consistent with what we observed in our study [110]. ALDH2 504lys allele is reported to be less prevalent in patients with alcoholic cirrhosis than that in controls [110]. Depending on the geographic region, the allele frequency for 504lys has been reported to be up to 40% among East Asian population [111], however, its frequency among patients with ALD is around 1.0%–11.6%, depending on the population being studied [106]. Taken together, there are several evidence linking the genetic variants of alcohol metabolizing enzyme genes with the risk of alcoholism/alcohol dependence; however, the association of these variants and ALD is still inconclusive.

5. Ethanol Metabolism and Its Association in ALD Pathogenesis

5.1. Redox State Alterations and ALD

Ethanol metabolism by alcohol metabolizing enzymes leads to the changes in intracellular redox state which may underlie the pathogenesis of ALD. One of the earliest responses to excessive alcohol use is the development of hepatic steatosis secondary to the accumulation of triglyceride droplets in the hepatocytes [10]. One of the pathophysiological mechanisms leading to hepatic steatosis is the imbalance between triglyceride synthesis and the ability to oxidize or export it in the form of very low-density lipoprotein (VLDL) particles [10]. Increasing intracellular NADH levels secondary to ethanol metabolism from ADH and ALDH inhibit fatty acid oxidation [112]. Additionally, the induction of a few enzymes involved in fatty acid synthesis as well as increase in the intracellular level of fatty acids and L-glycerol-3-phosphate result in the increase in the synthesis of triglycerides [10,113]. The change in the intracellular redox state leading to triglyceride accumulation has been confirmed in cell lines with stably expressed ADH. These cell lines can oxidize ethanol resulting in the alterations in intracellular redox potential as indicated by an increase in the lactate/pyruvate ratio. More importantly, they can accumulate large amounts of triglyceride upon alcohol treatment [114].
The alterations in the redox state, among several mechanisms, may interfere with sirtuin-1 (SIRT-1), a nicotinamide adenine dinucleotide (NAD+, NADH)-dependent class III histone deacetylase [115,116]. Ethanol-mediated SIRT1 inhibition may lead to hyper-acetylation of a set of molecules including histone H3, transcription factors such as sterol regulatory element-binding protein (SREBP) and peroxisome proliferator-activated receptor alpha (PPARα) [116]. This subsequently leads to the alterations in several downstream pathways which are involved in inflammatory responses and lipid metabolism leading to the development of ALD (see more detail in [116]).

5.2. Role of Oxidative Stress

As previously mentioned, CYP2E1 is induced by chronic alcohol use and is “leaky,” in that electrons transferred to it from CYP450 reductase can eventually be transferred to oxygen molecule forming superoxide [67,117]. The consequence of intracellular oxidative stress is lipid peroxidation [118], a reaction of lipid radical formation that follows the removal of an electron from polyunsaturated fatty acid (PUFA) [118,119]. The lipid free radical then form a lipid hydroperoxide with the addition of oxygen molecule, and ultimately decomposition to form reactive aldehydes such as 4-hydroxynonenal (4-HNE) and malondialdehyde (MDA) [120,121,122,123]. ALD has been shown to be associated with increase in lipid peroxidation, formation of 1-hydroxyethyl radical and lipid radicals, and decrease in hepatic antioxidants, especially glutathione [67,82,124]. In addition to the role of CYP2E1-induced oxidative stress, alcohol can also acetylate mitochondrial superoxide dismutase and impact its antioxidant ability [125]. Changes in diets that either promote or reduce oxidative stress have an impact on the degree of liver injury. When PUFA were substituted with either medium-chain triglycerides or saturated fats, the extent of alcohol-induced liver injury was ameliorated because of the reduction in lipid peroxidation [126,127]. CYP2E1 may be an important regulator of liver injury in obese patients with excessive alcohol use. Obesity is an independent risk factor for hepatic steatosis, alcoholic hepatitis, and cirrhosis among excessive drinkers [8,128]. Obesity and alcohol synergistically enhance the degree of liver injury [129,130]. Several studies indicate combined pathological effects of alcohol and obesity or fatty acid levels, respectively, on hepatocellular lipid accumulation and injury as well as hepatic inflammation, fibrosis, and cancerogenesis [130]. The induction of CYP2E1 is commonly observed in patients with excessive alcohol use and interestingly it can also be activated in the presence of obesity and metabolic syndrome by increasing levels of fatty acids [63,131,132]. The co-activation of CYP2E1 by both ethanol and endogenous substrates such as fatty acids, can augment the severity and accelerate the progression of liver injury [132,133]. In fact, a recent study in a cohort of 233 patients with alcoholic hepatitis from the United Kingdom and United States demonstrated that obesity is common in AH and associated with a greater than two-fold increase in short-term mortality [134].

5.3. Role of Protein Adduct Formation

Protein adducts formation during alcohol-induced liver injury are covalent linkages developed between amino acid side chains of liver proteins and compounds such as acetaldehyde, and byproducts of lipid peroxidation 4-HNE and MDA [135]. Once generated, acetaldehyde can form the covalent linkages with amino acid side chains of hepatic proteins to form acetaldehyde adducts. These adducts are located primarily in the perivenous region and develop early in the process of alcohol-induced liver injury [136]. Once the disease progresses, they are prevalent in areas of inflammation and fibrosis [137,138]. It has been well documented that these adducts elicit an immune response and can interfere with the functions of proteins especially at the lysine residues [139], such as lysine-dependent enzymes as well as cytoskeletal proteins [140]. There are reports that show acetaldehyde adducts can impair microtubule function leading to defects in protein transport which in turn attributes to endoplasmic reticulum stress in alcohol-induced liver injury [140].

6. Conclusions

ADH, CYP2E1, and catalase can generate acetaldehyde after alcohol ingestion. The ability of acetaldehyde to alter cellular function will depend on the enzymatic activity and acetaldehyde formation. Because of the differences in the capacity to metabolize alcohol to acetaldehyde, it has been suggested that individuals with high activity of alcohol metabolizing enzymes are susceptible to alcohol-induced liver injury due to a higher level of acetaldehyde exposure. However, the results on the allelic variants of the genes encoding for ADH, CYP2E1 genes, and ALD are inconclusive. Alcohol metabolizing enzyme, notably CYP2E1, may be an important regulator leading to liver injury especially in obese patients with excessive alcohol use. In addition to acetaldehyde, the shift in the redox state during alcohol elimination may also inter link to other pathways and trigger downstream effects leading to ALD. The knowledge gain from the studies of alcohol metabolism by these enzymes may perhaps open the new avenue for the treatment of ALD [141].

Author Contributions

Conceptualization and designing the contents for the manuscript, Y.J., T.Z., P.K., S.H., Z.Y. and S.L.; literature review Y.J., T.Z., P.K., S.H., Z.Y.; drafting the manuscript, Y.J., Z.Y. and S.L.; critical review of the draft Y.J., T.Z., P.K., S.H., Z.Y.; finalizing manuscript Z.Y. and S.L. All authors have read and agreed to the published version of the manuscript.

Funding

This work is partly supported by NIH K01AA26385 (to Z.Y.); R01 DK107682, R01 AA025208, U01 AA026917, UH2 AA026903 and VA Merit Award 1I01CX000361 (to S.L.).

Conflicts of Interest

All authors declare no conflict of interest with this manuscript.

Abbreviations

ALDAlcohol associated liver disease
ADHAlcohol dehydrogenase
ALDHAldehyde dehydrogenase
C/EBPCCAAT-enhancer-binding proteins
COUP-TFChicken ovalbumin upstream promoter transcription factor
CTF/NF-ITATAA binding factors and CAAT box-binding transcription factor/nuclear factor-1
CYPCytochrome
CYP2E1Cytochrome P450 2E1
DBPD-box binding protein
HNEHydroxynonenal
HNFhepatocyte nuclear factor
MDAMalondialdehyde
MEOSMicrosomal ethanol oxidizing system
PKCProtein kinase C
NADNicotinamide adenine dinucleotide
NADPNicotinamide adenine dinucleotide phosphate
PKCprotein kinase C
PPARsPeroxisome proliferator activated receptors
PPREPeroxisome proliferator responsive element
PUFAPolyunsaturated fatty acid
ROSReactive oxygen species
SIRTSirtuin
SREBPSterol regulatory element-binding protein
USFUpstream stimulatory factor
VLDLVery low density lipoprotein

References

  1. Liangpunsakul, S.; Haber, P.; McCaughan, G.W. Alcoholic Liver Disease in Asia, Europe, and North America. Gastroenterology 2016, 150, 1786–1797. [Google Scholar] [CrossRef] [Green Version]
  2. Rehm, J.; Samokhvalov, A.V.; Shield, K.D. Global burden of alcoholic liver diseases. J. Hepatol. 2013, 59, 160–168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Cutright, P.; Fernquist, R.M. Predictors of per capita alcohol consumption and gender-specific liver cirrhosis mortality rates: Thirteen European countries, circa 1970–1984 and 1995–2007. Omega (Westport) 2010, 62, 269–283. [Google Scholar] [CrossRef] [PubMed]
  4. Ramstedt, M. Alcohol consumption and liver cirrhosis mortality with and without mention of alcohol—The case of Canada. Addiction 2003, 98, 1267–1276. [Google Scholar] [CrossRef] [PubMed]
  5. Tuyns, A.J.; Pequignot, G. Greater risk of ascitic cirrhosis in females in relation to alcohol consumption. Int. J. Epidemiol. 1984, 13, 53–57. [Google Scholar] [CrossRef] [PubMed]
  6. Corrao, G.; Bagnardi, V.; Zambon, A.; Arico, S. Exploring the dose-response relationship between alcohol consumption and the risk of several alcohol-related conditions: A meta-analysis. Addiction 1999, 94, 1551–1573. [Google Scholar] [CrossRef]
  7. Becker, U.; Deis, A.; Sorensen, T.I.; Gronbaek, M.; Borch-Johnsen, K.; Muller, C.F.; Schnohr, P.; Jensen, G. Prediction of risk of liver disease by alcohol intake, sex, and age: A prospective population study. Hepatology 1996, 23, 1025–1029. [Google Scholar] [CrossRef]
  8. Liangpunsakul, S.; Puri, P.; Shah, V.; Kamath, P.; Sanyal, A.; Urban, T.; Ren, X.; Katz, B.; Radaeva, S.; Chalasani, N.; et al. Effects of Age, Sex, Body Weight, and Quantity of Alcohol Consumption on Occurrence and Severity of Alcoholic Hepatitis. Clin. Gastroenterol. Hepatol. 2016. [Google Scholar] [CrossRef] [Green Version]
  9. Becker, U.; Gronbaek, M.; Johansen, D.; Sorensen, T.I. Lower risk for alcohol-induced cirrhosis in wine drinkers. Hepatology 2002, 35, 868–875. [Google Scholar] [CrossRef]
  10. Sozio, M.S.; Liangpunsakul, S.; Crabb, D. The role of lipid metabolism in the pathogenesis of alcoholic and nonalcoholic hepatic steatosis. Semin. Liver Dis. 2010, 30, 378–390. [Google Scholar] [CrossRef]
  11. Lieber, C.S.; JONES, D.P.; DECARLI, L.M. Effects of prolonged ethanol intake: production of fatty liver despite adequate diets. J. Clin. Investig. 1965, 44, 1009–1021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Schwartz, J.M.; Reinus, J.F. Prevalence and natural history of alcoholic liver disease. Clin. Liver Dis. 2012, 16, 659–666. [Google Scholar] [CrossRef] [PubMed]
  13. Lane, B.P.; Lieber, C.S. Ultrastructural alterations in human hepatocytes following ingestion of ethanol with adequate diets. Am. J. Pathol. 1966, 49, 593–603. [Google Scholar] [PubMed]
  14. Teli, M.R.; Day, C.P.; Burt, A.D.; Bennett, M.K.; James, O.F. Determinants of progression to cirrhosis or fibrosis in pure alcoholic fatty liver. Lancet 1995, 346, 987–990. [Google Scholar] [CrossRef]
  15. Lourens, S.; Sunjaya, D.B.; Singal, A.; Liangpunsakul, S.; Puri, P.; Sanyal, A.; Ren, X.; Gores, G.J.; Radaeva, S.; Chalasani, N.; et al. Acute Alcoholic Hepatitis: Natural History and Predictors of Mortality Using a Multicenter Prospective Study. Mayo Clin. Proc. Innov. Qual. Outcomes 2017, 1, 37–48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Jinjuvadia, R.; Liangpunsakul, S. Trends in Alcoholic Hepatitis-related Hospitalizations, Financial Burden, and Mortality in the United States. J. Clin. Gastroenterol. 2015, 49, 506–511. [Google Scholar] [CrossRef] [Green Version]
  17. Chayanupatkul, M.; Liangpunsakul, S. Alcoholic hepatitis: A comprehensive review of pathogenesis and treatment. World J. Gastroenterol. 2014, 20, 6279–6286. [Google Scholar] [CrossRef]
  18. Peeraphatdit, T.B.; Kamath, P.S.; Karpyak, V.M.; Davis, B.; Desai, V.; Liangpunsakul, S.; Sanyal, A.; Chalasani, N.; Shah, V.H.; Simonetto, D.A. Alcohol Rehabilitation Within 30 Days of Hospital Discharge is Associated With Reduced Readmission, Relapse, and Death in Patients with Alcoholic Hepatitis. Clin. Gastroenterol. Hepatol. 2019. [Google Scholar] [CrossRef]
  19. Mills, S.J.; Harrison, S.A. Comparison of the natural history of alcoholic and nonalcoholic fatty liver disease. Curr. Gastroenterol. Rep. 2005, 7, 32–36. [Google Scholar] [CrossRef]
  20. Tapper, E.B.; Parikh, N.D. Mortality due to cirrhosis and liver cancer in the United States, 1999–2016: Observational study. BMJ 2018, 362, k2817. [Google Scholar] [CrossRef] [Green Version]
  21. Frezza, M.; di, P.C.; Pozzato, G.; Terpin, M.; Baraona, E.; Lieber, C.S. High blood alcohol levels in women. The role of decreased gastric alcohol dehydrogenase activity and first-pass metabolism. N. Engl. J. Med. 1990, 322, 95–99. [Google Scholar] [CrossRef] [PubMed]
  22. Enomoto, N.; Yamashina, S.; Schemmer, P.; Rivera, C.A.; Bradford, B.U.; Enomoto, A.; Brenner, D.A.; Thurman, R.G. Estriol sensitizes rat Kupffer cells via gut-derived endotoxin. Am. J. Physiol. 1999, 277, G671–G677. [Google Scholar] [CrossRef] [PubMed]
  23. Crabb, D.W.; Liangpunsakul, S. Acetaldehyde generating enzyme systems: Roles of alcohol dehydrogenase, CYP2E1 and catalase, and speculations on the role of other enzymes and processes. Novartis. Found. Symp. 2007, 285, 4–16. [Google Scholar] [PubMed]
  24. Paton, A. Alcohol in the body. BMJ 2005, 330, 85–87. [Google Scholar] [CrossRef] [PubMed]
  25. Beck, I.T.; Paloschi, G.B.; Dinda, P.K.; Beck, M. Effect of intragastric administration of alcohol on the ethanol concentrations and osmolality of pancreatic juice, bile, and portal and peripheral blood. Gastroenterology 1974, 67, 484–489. [Google Scholar] [CrossRef]
  26. Lieber, C.S. Metabolism of alcohol. Clin. Liver Dis. 2005, 9, 1–35. [Google Scholar] [CrossRef]
  27. Jones, A.W. Alcohol, its absorption, distribution, metabolism, and excretion in the body and pharmacokinetic calculations. WIREs Forensic Sci. 2019, 1, e1340. [Google Scholar] [CrossRef]
  28. Ramchandani, V.A.; Bosron, W.F.; Li, T.K. Research advances in ethanol metabolism. Pathol. Biol. (Paris) 2001, 49, 676–682. [Google Scholar] [CrossRef]
  29. Crabb, D.W.; Matsumoto, M.; Chang, D.; You, M. Overview of the role of alcohol dehydrogenase and aldehyde dehydrogenase and their variants in the genesis of alcohol-related pathology. Proc. Nutr. Soc. 2004, 63, 49–63. [Google Scholar] [CrossRef] [Green Version]
  30. Sun, H.W.; Plapp, B.V. Progressive sequence alignment and molecular evolution of the Zn-containing alcohol dehydrogenase family. J. Mol. Evol. 1992, 34, 522–535. [Google Scholar] [CrossRef]
  31. Jornvall, H.; Persson, B.; Jeffery, J. Characteristics of alcohol/polyol dehydrogenases. The zinc-containing long-chain alcohol dehydrogenases. Eur. J. Biochem. 1987, 167, 195–201. [Google Scholar] [CrossRef] [PubMed]
  32. Donohue, T.M.; Osna, N.A.; Clemens, D.L. Recombinant Hep G2 cells that express alcohol dehydrogenase and cytochrome P450 2E1 as a model of ethanol-elicited cytotoxicity. Int. J. Biochem. Cell Biol. 2006, 38, 92–101. [Google Scholar] [CrossRef] [PubMed]
  33. Yoshida, A.; Hsu, L.C.; Yasunami, M. Genetics of human alcohol-metabolizing enzymes. Prog. Nucleic Acid Res. Mol. Biol. 1991, 40, 255–287. [Google Scholar] [PubMed]
  34. Borras, E.; Coutelle, C.; Rosell, A.; Fernandez-Muixi, F.; Broch, M.; Crosas, B.; Hjelmqvist, L.; Lorenzo, A.; Gutierrez, C.; Santos, M.; et al. Genetic polymorphism of alcohol dehydrogenase in europeans: The ADH2*2 allele decreases the risk for alcoholism and is associated with ADH3*1. Hepatology 2000, 31, 984–989. [Google Scholar] [CrossRef]
  35. Eng, M.Y.; Luczak, S.E.; Wall, T.L. ALDH2, ADH1B, and ADH1C genotypes in Asians: A literature review. Alcohol Res. Health 2007, 30, 22–27. [Google Scholar]
  36. Thomasson, H.R.; Beard, J.D.; Li, T.K. ADH2 gene polymorphisms are determinants of alcohol pharmacokinetics. Alcohol. Clin. Exp. Res. 1995, 19, 1494–1499. [Google Scholar] [CrossRef]
  37. Thomasson, H.R.; Crabb, D.W.; Edenberg, H.J.; Li, T.K.; Hwu, H.G.; Chen, C.C.; Yeh, E.K.; Yin, S.J. Low frequency of the ADH2*2 allele among Atayal natives of Taiwan with alcohol use disorders. Alcohol. Clin. Exp. Res. 1994, 18, 640–643. [Google Scholar] [CrossRef]
  38. Ehrig, T.; Bosron, W.F.; Li, T.K. Alcohol and aldehyde dehydrogenase. Alcohol Alcohol. 1990, 25, 105–116. [Google Scholar] [CrossRef]
  39. Svensson, S.; Some, M.; Lundsjo, A.; Helander, A.; Cronholm, T.; Hoog, J.O. Activities of human alcohol dehydrogenases in the metabolic pathways of ethanol and serotonin. Eur. J. Biochem. 1999, 262, 324–329. [Google Scholar] [CrossRef] [Green Version]
  40. Holmes, R.S. Alcohol dehydrogenases: A family of isozymes with differential functions. Alcohol Alcohol. Suppl. 1994, 2, 127–130. [Google Scholar]
  41. Han, C.L.; Liao, C.S.; Wu, C.W.; Hwong, C.L.; Lee, A.R.; Yin, S.J. Contribution to first-pass metabolism of ethanol and inhibition by ethanol for retinol oxidation in human alcohol dehydrogenase family--implications for etiology of fetal alcohol syndrome and alcohol-related diseases. Eur. J. Biochem. 1998, 254, 25–31. [Google Scholar] [CrossRef] [Green Version]
  42. Oliveira, L.M.; Teixeira, F.M.E.; Sato, M.N. Impact of Retinoic Acid on Immune Cells and Inflammatory Diseases. Mediat. Inflamm. 2018, 2018, 3067126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. 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]
  44. Dohmen, K.; Baraona, E.; Ishibashi, H.; Pozzato, G.; Moretti, M.; Matsunaga, C.; Fujimoto, K.; Lieber, C.S. Ethnic differences in gastric sigma-alcohol dehydrogenase activity and ethanol first-pass metabolism. Alcohol. Clin. Exp. Res. 1996, 20, 1569–1576. [Google Scholar] [CrossRef] [PubMed]
  45. Yuan, J.; Chen, C.; Cui, J.; Lu, J.; Yan, C.; Wei, X.; Zhao, X.; Li, N.; Li, S.; Xue, G.; et al. Fatty Liver Disease Caused by High-Alcohol-Producing Klebsiella pneumoniae. Cell Metab. 2019, 30, 675–688. [Google Scholar] [CrossRef]
  46. 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]
  47. Salaspuro, M. Microbial metabolism of ethanol and acetaldehyde and clinical consequences. Addict. Biol. 1997, 2, 35–46. [Google Scholar] [CrossRef]
  48. Tagaino, R.; Washio, J.; Abiko, Y.; Tanda, N.; Sasaki, K.; Takahashi, N. Metabolic property of acetaldehyde production from ethanol and glucose by oral Streptococcus and Neisseria. Sci. Rep. 2019, 9, 10446. [Google Scholar] [CrossRef] [Green Version]
  49. Potter, J.J.; Cheneval, D.; Dang, C.V.; Resar, L.M.; Mezey, E.; Yang, V.W. The upstream stimulatory factor binds to and activates the promoter of the rat class I alcohol dehydrogenase gene. J. Biol. Chem. 1991, 266, 15457–15463. [Google Scholar]
  50. Stewart, M.J.; McBride, M.S.; Winter, L.A.; Duester, G. Promoters for the human alcohol dehydrogenase genes ADH1, ADH2, and ADH3: Interaction of CCAAT/enhancer-binding protein with elements flanking the ADH2 TATA box. Gene 1990, 90, 271–279. [Google Scholar] [CrossRef]
  51. Edenberg, H.J. Regulation of the mammalian alcohol dehydrogenase genes. Prog. Nucleic Acid Res. Mol. Biol. 2000, 64, 295–341. [Google Scholar] [PubMed]
  52. van Ooij, C.; Snyder, R.C.; Paeper, B.W.; Duester, G. Temporal expression of the human alcohol dehydrogenase gene family during liver development correlates with differential promoter activation by hepatocyte nuclear factor 1, CCAAT/enhancer-binding protein alpha, liver activator protein, and D-element-binding protein. Mol. Cell. Biol. 1992, 12, 3023–3031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Su, J.S.; Tsai, T.F.; Chang, H.M.; Chao, K.M.; Su, T.S.; Tsai, S.F. Distant HNF1 site as a master control for the human class I alcohol dehydrogenase gene expression. J. Biol. Chem. 2006, 281, 19809–19821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Duester, G. Retinoids and the alcohol dehydrogenase gene family. EXS 1994, 71, 279–290. [Google Scholar] [PubMed]
  55. Dong, Y.; Poellinger, L.; Okret, S.; Hoog, J.O.; von Bahr-Lindstrom, H.; Jornvall, H.; Gustafsson, J.A. Regulation of gene expression of class I alcohol dehydrogenase by glucocorticoids. Proc. Natl. Acad. Sci. USA 1988, 85, 767–771. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Kim, D.S.; Lee, C.B.; Park, Y.S.; Ahn, Y.H.; Kim, T.W.; Kee, C.S.; Kang, J.S.; Om, A.S. Effect of thyroid hormone on the alcohol dehydrogenase activities in rat tissues. J. Korean Med. Sci. 2001, 16, 313–316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. He, L.; Ronis, M.J.; Badger, T.M. Ethanol induction of class I alcohol dehydrogenase expression in the rat occurs through alterations in CCAAT/enhancer binding proteins beta and gamma. J. Biol. Chem. 2002, 277, 43572–43577. [Google Scholar] [CrossRef] [Green Version]
  58. Potter, J.J.; Rennie-Tankersley, L.; Mezey, E. Endotoxin enhances liver alcohol dehydrogenase by action through upstream stimulatory factor but not by nuclear factor-kappa B. J. Biol. Chem. 2003, 278, 4353–4357. [Google Scholar] [CrossRef] [Green Version]
  59. Bosron, W.F.; Crabb, D.W.; Li, T.K. Relationship between kinetics of liver alcohol dehydrogenase and alcohol metabolism. Pharmacol. Biochem. Behav. 1983, 18 (Suppl. S1), 223–227. [Google Scholar] [CrossRef]
  60. Crabb, D.W.; Bosron, W.F.; Li, T.K. Steady-state kinetic properties of purified rat liver alcohol dehydrogenase: Application to predicting alcohol elimination rates in vivo. Arch. Biochem. Biophys. 1983, 224, 299–309. [Google Scholar] [CrossRef]
  61. Lieber, C.S.; DeCarli, L.M. Ethanol oxidation by hepatic microsomes: Adaptive increase after ethanol feeding. Science 1968, 162, 917–918. [Google Scholar] [CrossRef] [PubMed]
  62. Niemela, O.; Parkkila, S.; Juvonen, R.O.; Viitala, K.; Gelboin, H.V.; Pasanen, M. Cytochromes P450 2A6, 2E1, and 3A and production of protein-aldehyde adducts in the liver of patients with alcoholic and non-alcoholic liver diseases. J. Hepatol. 2000, 33, 893–901. [Google Scholar] [CrossRef]
  63. Liangpunsakul, S.; Kolwankar, D.; Pinto, A.; Gorski, J.C.; Hall, S.D.; Chalasani, N. Activity of CYP2E1 and CYP3A enzymes in adults with moderate alcohol consumption: A comparison with nonalcoholics. Hepatology 2005, 41, 1144–1150. [Google Scholar] [CrossRef] [PubMed]
  64. Lieber, C.S. The discovery of the microsomal ethanol oxidizing system and its physiologic and pathologic role. Drug Metab. Rev. 2004, 36, 511–529. [Google Scholar] [CrossRef] [PubMed]
  65. Quertemont, E. Genetic polymorphism in ethanol metabolism: Acetaldehyde contribution to alcohol abuse and alcoholism. Mol. Psychiatry 2004, 9, 570–581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Wang, Z.; Hall, S.D.; Maya, J.F.; Li, L.; Asghar, A.; Gorski, J.C. Diabetes mellitus increases the in vivo activity of cytochrome P450 2E1 in humans. Br. J. Clin. Pharmacol. 2003, 55, 77–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Lu, Y.; Cederbaum, A.I. CYP2E1 and oxidative liver injury by alcohol. Free Radic. Biol. Med. 2008, 44, 723–738. [Google Scholar] [CrossRef] [Green Version]
  68. Hong, J.Y.; Pan, J.M.; Gonzalez, F.J.; Gelboin, H.V.; Yang, C.S. The induction of a specific form of cytochrome P-450 (P-450j) by fasting. Biochem. Biophys. Res. Commun. 1987, 142, 1077–1083. [Google Scholar] [CrossRef]
  69. Johansson, I.; Lindros, K.O.; Eriksson, H.; Ingelman-Sundberg, M. Transcriptional control of CYP2E1 in the perivenous liver region and during starvation. Biochem. Biophys. Res. Commun. 1990, 173, 331–338. [Google Scholar] [CrossRef]
  70. Zanger, U.M.; Schwab, M. Cytochrome P450 enzymes in drug metabolism: Regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacol. Ther. 2013, 138, 103–141. [Google Scholar] [CrossRef]
  71. Takahashi, T.; Lasker, J.M.; Rosman, A.S.; Lieber, C.S. Induction of cytochrome P-4502E1 in the human liver by ethanol is caused by a corresponding increase in encoding messenger RNA. Hepatology 1993, 17, 236–245. [Google Scholar] [PubMed]
  72. Lagadic-Gossmann, D.; Lerche, C.; Rissel, M.; Joannard, F.; Galisteo, M.; Guillouzo, A.; Corcos, L. The induction of the human hepatic CYP2E1 gene by interleukin 4 is transcriptional and regulated by protein kinase C. Cell Biol. Toxicol. 2000, 16, 221–233. [Google Scholar] [CrossRef] [PubMed]
  73. Wang, Y.; Yu, D.; Tolleson, W.H.; Yu, L.R.; Green, B.; Zeng, L.; Chen, Y.; Chen, S.; Ren, Z.; Guo, L.; et al. A systematic evaluation of microRNAs in regulating human hepatic CYP2E1. Biochem. Pharmacol. 2017, 138, 174–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Tsutsumi, M.; Lasker, J.M.; Takahashi, T.; Lieber, C.S. In vivo induction of hepatic P4502E1 by ethanol: Role of increased enzyme synthesis. Arch. Biochem. Biophys. 1993, 304, 209–218. [Google Scholar] [CrossRef] [PubMed]
  75. Weathermon, R.; Crabb, D.W. Alcohol and medication interactions. Alcohol Res. Health 1999, 23, 40–54. [Google Scholar]
  76. Glorieux, C.; Calderon, P.B. Catalase, a remarkable enzyme: Targeting the oldest antioxidant enzyme to find a new cancer treatment approach. Biol. Chem. 2017, 398, 1095–1108. [Google Scholar] [CrossRef] [Green Version]
  77. Oshino, N.; Oshino, R.; Chance, B. The characteristics of the “peroxidatic” reaction of catalase in ethanol oxidation. Biochem. J. 1973, 131, 555–563. [Google Scholar] [CrossRef] [Green Version]
  78. Rodriguez, E.; Peiroten, A.; Landete, J.M.; Medina, M.; Arques, J.L. Gut Catalase-Positive Bacteria Cross-Protect Adjacent Bifidobacteria from Oxidative Stress. Microbes Environ. 2015, 30, 270–272. [Google Scholar] [CrossRef] [Green Version]
  79. Tillonen, J.; Kaihovaara, P.; Jousimies-Somer, H.; Heine, R.; Salaspuro, M. Role of catalase in in vitro acetaldehyde formation by human colonic contents. Alcohol. Clin. Exp. Res. 1998, 22, 1113–1119. [Google Scholar] [CrossRef]
  80. Orellana, M.; Rodrigo, R.; Valdes, E. Peroxisomal and microsomal fatty acid oxidation in liver of rats after chronic ethanol consumption. Gen. Pharmacol. 1998, 31, 817–820. [Google Scholar] [CrossRef]
  81. Girnun, G.D.; Domann, F.E.; Moore, S.A.; Robbins, M.E. Identification of a functional peroxisome proliferator-activated receptor response element in the rat catalase promoter. Mol. Endocrinol. 2002, 16, 2793–2801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Wu, D.; Cederbaum, A.I. Alcohol, oxidative stress, and free radical damage. Alcohol Res. Health 2003, 27, 277–284. [Google Scholar] [PubMed]
  83. Thomasson, H.R.; Crabb, D.W.; Edenberg, H.J.; Li, T.K. Alcohol and aldehyde dehydrogenase polymorphisms and alcoholism. Behav. Genet. 1993, 23, 131–136. [Google Scholar] [CrossRef] [PubMed]
  84. Nene, A.; Chen, C.H.; Disatnik, M.H.; Cruz, L.; Mochly-Rosen, D. Aldehyde dehydrogenase 2 activation and coevolution of its epsilonPKC-mediated phosphorylation sites. J. Biomed. Sci. 2017, 24, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. 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]
  86. 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]
  87. 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]
  88. 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]
  89. Castro, G.D.; Delgado de Layno, A.M.; Costantini, M.H.; Castro, J.A. Cytosolic xanthine oxidoreductase mediated bioactivation of ethanol to acetaldehyde and free radicals in rat breast tissue. Its potential role in alcohol-promoted mammary cancer. Toxicology 2001, 160, 11–18. [Google Scholar] [CrossRef]
  90. Diaz Gomez, M.I.; Castro, G.D.; de Layno, A.M.; Costantini, M.H.; Castro, J.A. Cytochrome P450 reductase-mediated anaerobic biotransformation of ethanol to 1-hydroxyethyl-free radicals and acetaldehyde. Toxicology 2000, 154, 113–122. [Google Scholar] [CrossRef]
  91. Couzigou, P.; Coutelle, C.; Fleury, B.; Iron, A. Alcohol and aldehyde dehydrogenase genotypes, alcoholism and alcohol related disease. Alcohol Alcohol. Suppl. 1994, 2, 21–27. [Google Scholar] [PubMed]
  92. Anstee, Q.M.; Seth, D.; Day, C.P. Genetic Factors That Affect Risk of Alcoholic and Nonalcoholic Fatty Liver Disease. Gastroenterology 2016, 150, 1728–1744. [Google Scholar] [CrossRef] [PubMed]
  93. Chambers, G.K.; Marshall, S.J.; Robinson, G.M.; Maguire, S.; Newton-Howes, J.; Chong, N.L. The genetics of alcoholism in Polynesians: Alcohol and aldehyde dehydrogenase genotypes in young men. Alcohol. Clin. Exp. Res. 2002, 26, 949–955. [Google Scholar] [CrossRef] [PubMed]
  94. Chen, W.J.; Loh, E.W.; Hsu, Y.P.; Chen, C.C.; Yu, J.M.; Cheng, A.T. Alcohol-metabolising genes and alcoholism among Taiwanese Han men: Independent effect of ADH2, ADH3 and ALDH2. Br. J. Psychiatry 1996, 168, 762–767. [Google Scholar] [CrossRef]
  95. Higuchi, S. Polymorphisms of ethanol metabolizing enzyme genes and alcoholism. Alcohol Alcohol. Suppl. 1994, 2, 29–34. [Google Scholar]
  96. Yin, S.J. Alcohol dehydrogenase: Enzymology and metabolism. Alcohol Alcohol. Suppl. 1994, 2, 113–119. [Google Scholar]
  97. Mizoi, Y.; Yamamoto, K.; Ueno, Y.; Fukunaga, T.; Harada, S. Involvement of genetic polymorphism of alcohol and aldehyde dehydrogenases in individual variation of alcohol metabolism. Alcohol Alcohol. 1994, 29, 707–710. [Google Scholar]
  98. 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]
  99. Yokoyama, A.; Taniki, N.; Hara, S.; Haysashi, E.; Nakamoto, N.; Mizukami, T.; Maruyama, K.; Yokoyama, T. Slow-metabolizing ADH1B and inactive heterozygous ALDH2 increase vulnerability to fatty liver in Japanese men with alcohol dependence. J. Gastroenterol. 2018, 53, 660–669. [Google Scholar] [CrossRef]
  100. Grove, J.; Brown, A.S.; Daly, A.K.; Bassendine, M.F.; James, O.F.; Day, C.P. The RsaI polymorphism of CYP2E1 and susceptibility to alcoholic liver disease in Caucasians: Effect on age of presentation and dependence on alcohol dehydrogenase genotype. Pharmacogenetics 1998, 8, 335–342. [Google Scholar] [CrossRef]
  101. Hayashi, S.; Watanabe, J.; Kawajiri, K. Genetic polymorphisms in the 5’-flanking region change transcriptional regulation of the human cytochrome P450IIE1 gene. J. Biochem. 1991, 110, 559–565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Zhu, L.; He, Y.; Niu, F.; Yan, M.; Li, J.; Yuan, D.; Jin, T. Polymorphisms of drug-metabolizing enzyme CYP2E1 in Chinese Uygur population. Medicine 2018, 97, e9970. [Google Scholar] [CrossRef] [PubMed]
  103. Watanabe, J.; Hayashi, S.; Kawajiri, K. Different regulation and expression of the human CYP2E1 gene due to the RsaI polymorphism in the 5’-flanking region. J. Biochem. 1994, 116, 321–326. [Google Scholar] [CrossRef] [PubMed]
  104. Kodydkova, J.; Vavrova, L.; Kocik, M.; Zak, A. Human catalase, its polymorphisms, regulation and changes of its activity in different diseases. Folia Biol. (Praha) 2014, 60, 153–167. [Google Scholar]
  105. 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] [Green Version]
  106. Li, D.; Zhao, H.; Gelernter, J. Strong protective effect of the aldehyde dehydrogenase gene (ALDH2) 504lys (*2) allele against alcoholism and alcohol-induced medical diseases in Asians. Hum. Genet. 2012, 131, 725–737. [Google Scholar] [CrossRef] [Green Version]
  107. 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] [Green Version]
  108. Whitfield, J.B.; Martin, N.G. Aversive reactions and alcohol use in Europeans. Alcohol. Clin. Exp. Res. 1993, 17, 131–134. [Google Scholar] [CrossRef]
  109. Petersen, E.N. The pharmacology and toxicology of disulfiram and its metabolites. Acta Psychiatr. Scand. Suppl. 1992, 369, 7–13. [Google Scholar] [CrossRef]
  110. Chang, B.; Hao, S.; Zhang, L.; Gao, M.; Sun, Y.; Huang, A.; Teng, G.; Li, B.; Crabb, D.W.; Kusumanchi, P.; et al. Association Between Aldehyde Dehydrogenase 2 Glu504Lys Polymorphism and Alcoholic Liver Disease. Am. J. Med. Sci. 2018, 356, 10–14. [Google Scholar] [CrossRef]
  111. Li, H.; Borinskaya, S.; Yoshimura, K.; Kal’ina, N.; Marusin, A.; Stepanov, V.A.; Qin, Z.; Khaliq, S.; Lee, M.Y.; Yang, Y.; et al. Refined geographic distribution of the oriental ALDH2*504Lys (nee 487Lys) variant. Ann. Hum. Genet. 2009, 73, 335–345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Grunnet, N.; Kondrup, J. The effect of ethanol on the beta-oxidation of fatty acids. Alcohol. Clin. Exp. Res. 1986, 10, 64S–68S. [Google Scholar] [CrossRef] [PubMed]
  113. Liangpunsakul, S.; Bennett, R.; Westerhold, C.; Ross, R.A.; Crabb, D.W.; Lai, X.; Witzmann, F.A. Increasing serum pre-adipocyte factor-1 (Pref-1) correlates with decreased body fat, increased free fatty acids, and level of recent alcohol consumption in excessive alcohol drinkers. Alcohol 2014, 48, 795–800. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Galli, A.; Price, D.; Crabb, D. High-level expression of rat class I alcohol dehydrogenase is sufficient for ethanol-induced fat accumulation in transduced HeLa cells. Hepatology 1999, 29, 1164–1170. [Google Scholar] [CrossRef]
  115. Chang, H.C.; Guarente, L. SIRT1 and other sirtuins in metabolism. Trends Endocrinol. Metab. 2014, 25, 138–145. [Google Scholar] [CrossRef]
  116. You, M.; Jogasuria, A.; Taylor, C.; Wu, J. Sirtuin 1 signaling and alcoholic fatty liver disease. Hepatobiliary Surg. Nutr. 2015, 4, 88–100. [Google Scholar] [CrossRef]
  117. Cederbaum, A.I. Alcohol metabolism. Clin. Liver Dis. 2012, 16, 667–685. [Google Scholar] [CrossRef] [Green Version]
  118. Cichoz-Lach, H.; Michalak, A. Oxidative stress as a crucial factor in liver diseases. World J. Gastroenterol. 2014, 20, 8082–8091. [Google Scholar] [CrossRef]
  119. Shahidi, F.; Zhong, Y. Lipid oxidation and improving the oxidative stability. Chem. Soc. Rev. 2010, 39, 4067–4079. [Google Scholar] [CrossRef] [PubMed]
  120. Kamimura, S.; Gaal, K.; Britton, R.S.; Bacon, B.R.; Triadafilopoulos, G.; Tsukamoto, H. Increased 4-hydroxynonenal levels in experimental alcoholic liver disease: Association of lipid peroxidation with liver fibrogenesis. Hepatology 1992, 16, 448–453. [Google Scholar] [CrossRef]
  121. Shearn, C.T.; Backos, D.S.; Orlicky, D.J.; Smathers-McCullough, R.L.; Petersen, D.R. Identification of 5’ AMP-activated kinase as a target of reactive aldehydes during chronic ingestion of high concentrations of ethanol. J. Biol. Chem. 2014, 289, 15449–15462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Ayala, A.; Munoz, M.F.; Arguelles, S. Lipid peroxidation: Production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. Oxid. Med. Cell. Longev. 2014, 2014, 360438. [Google Scholar] [CrossRef] [PubMed]
  123. Smathers, R.L.; Galligan, J.J.; Stewart, B.J.; Petersen, D.R. Overview of lipid peroxidation products and hepatic protein modification in alcoholic liver disease. Chem. Biol. Interact. 2011, 192, 107–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Nanji, A.A.; Zhao, S.; Sadrzadeh, S.M.; Dannenberg, A.J.; Tahan, S.R.; Waxman, D.J. Markedly enhanced cytochrome P450 2E1 induction and lipid peroxidation is associated with severe liver injury in fish oil-ethanol-fed rats. Alcohol. Clin. Exp. Res. 1994, 18, 1280–1285. [Google Scholar] [CrossRef]
  125. Assiri, M.A.; Roy, S.R.; Harris, P.S.; Ali, H.; Liang, Y.; Shearn, C.T.; Orlicky, D.J.; Roede, J.R.; Hirschey, M.D.; Backos, D.S.; et al. Chronic Ethanol Metabolism Inhibits Hepatic Mitochondrial Superoxide Dismutase via Lysine Acetylation. Alcohol. Clin. Exp. Res. 2017, 41, 1705–1714. [Google Scholar] [CrossRef]
  126. Kirpich, I.A.; Miller, M.E.; Cave, M.C.; Joshi-Barve, S.; McClain, C.J. Alcoholic Liver Disease: Update on the Role of Dietary Fat. Biomolecules 2016, 6, 1. [Google Scholar] [CrossRef] [Green Version]
  127. Nanji, A.A.; Jokelainen, K.; Tipoe, G.L.; Rahemtulla, A.; Dannenberg, A.J. Dietary saturated fatty acids reverse inflammatory and fibrotic changes in rat liver despite continued ethanol administration. J. Pharmacol. Exp. Ther. 2001, 299, 638–644. [Google Scholar]
  128. Chiang, D.J.; McCullough, A.J. The impact of obesity and metabolic syndrome on alcoholic liver disease. Clin. Liver Dis. 2014, 18, 157–163. [Google Scholar] [CrossRef]
  129. Ruhl, C.E.; Everhart, J.E. Joint effects of body weight and alcohol on elevated serum alanine aminotransferase in the United States population. Clin. Gastroenterol. Hepatol. 2005, 3, 1260–1268. [Google Scholar] [CrossRef]
  130. Mahli, A.; Hellerbrand, C. Alcohol and Obesity: A Dangerous Association for Fatty Liver Disease. Dig Dis 2016, 34 (Suppl. S1), 32–39. [Google Scholar] [CrossRef] [Green Version]
  131. Emery, M.G.; Fisher, J.M.; Chien, J.Y.; Kharasch, E.D.; Dellinger, E.P.; Kowdley, K.V.; Thummel, K.E. CYP2E1 activity before and after weight loss in morbidly obese subjects with nonalcoholic fatty liver disease. Hepatology 2003, 38, 428–435. [Google Scholar] [CrossRef] [PubMed]
  132. Cederbaum, A.I. CYP2E1 potentiates toxicity in obesity and after chronic ethanol treatment. Drug Metab. Drug Interact. 2012, 27, 125–144. [Google Scholar] [CrossRef] [PubMed]
  133. Song, B.J.; Akbar, M.; Jo, I.; Hardwick, J.P.; Abdelmegeed, M.A. Translational Implications of the Alcohol-Metabolizing Enzymes, Including Cytochrome P450-2E1, in Alcoholic and Nonalcoholic Liver Disease. Adv. Pharmacol. 2015, 74, 303–372. [Google Scholar] [CrossRef] [PubMed]
  134. Parker, R.; Kim, S.J.; Im, G.Y.; Nahas, J.; Dhesi, B.; Vergis, N.; Sinha, A.; Ghezzi, A.; Rink, M.R.; McCune, A.; et al. Obesity in acute alcoholic hepatitis increases morbidity and mortality. EBioMedicine 2019, 45, 511–518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Israel, Y.; Hurwitz, E.; Niemela, O.; Arnon, R. Monoclonal and polyclonal antibodies against acetaldehyde-containing epitopes in acetaldehyde-protein adducts. Proc. Natl. Acad. Sci. USA 1986, 83, 7923–7927. [Google Scholar] [CrossRef] [Green Version]
  136. Tuma, D.J.; Casey, C.A. Dangerous byproducts of alcohol breakdown--focus on adducts. Alcohol Res. Health 2003, 27, 285–290. [Google Scholar]
  137. Niemela, O. Distribution of ethanol-induced protein adducts in vivo: Relationship to tissue injury. Free Radic. Biol. Med. 2001, 31, 1533–1538. [Google Scholar] [CrossRef]
  138. Niemela, O. Aldehyde-protein adducts in the liver as a result of ethanol-induced oxidative stress. Front. Biosci. 1999, 4, D506–D513. [Google Scholar] [CrossRef] [Green Version]
  139. Tuma, D.J.; Hoffman, T.; Sorrell, M.F. The chemistry of acetaldehyde-protein adducts. Alcohol Alcohol. Suppl. 1991, 1, 271–276. [Google Scholar]
  140. Tuma, D.J.; Smith, S.L.; Sorrell, M.F. Acetaldehyde and microtubules. Ann. N. Y. Acad. Sci. 1991, 625, 786–792. [Google Scholar] [CrossRef]
  141. Guillot, A.; Ren, T.; Jourdan, T.; Pawlosky, R.J.; Han, E.; Kim, S.J.; Zhang, L.; Koob, G.F.; Gao, B. Targeting liver aldehyde dehydrogenase-2 prevents heavy but not moderate alcohol drinking. Proc. Natl. Acad. Sci. USA 2019, 116, 25974–25981. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The major pathways of acetaldehyde formation after alcohol ingestion, alcohol dehydrogenase (ADH), cytochrome P450 2E1 (CYP2E1), catalase, and aldehyde dehydrogenase (ALDH) are demonstrated with their cofactors, and substrates.
Figure 1. The major pathways of acetaldehyde formation after alcohol ingestion, alcohol dehydrogenase (ADH), cytochrome P450 2E1 (CYP2E1), catalase, and aldehyde dehydrogenase (ALDH) are demonstrated with their cofactors, and substrates.
Biomedicines 08 00050 g001
Table 1. The kinetic properties of each ADH class, population, and tissue distribution [23,29].
Table 1. The kinetic properties of each ADH class, population, and tissue distribution [23,29].
Gene LocusSubunitKm for Ethanol (mM)Vmax (min−1)Major EthnicityTissue Distribution
Class I
ADH1B*1β10.0554Caucasians, African-AmericansLiver, lung
ADH1B*2β20.9-Asians
ADH1B*3β234-African-Americans
ADH1C*1γ11-All groupsLiver, stomach
ADH1C*2γ20.63-Caucasians
Class II: ADH2π3440 Liver
Class III: ADH3χ1000- Ubiquitous
Class IV: ADH4σ, µ201510 Stomach and esophagus
“-“: no data available.
Table 2. Km for acetaldehyde and tissue distribution of human aldehyde dehydrogenase (ALDH) [23,29].
Table 2. Km for acetaldehyde and tissue distribution of human aldehyde dehydrogenase (ALDH) [23,29].
Gene LocusKm for AcetaldehydeTissue Distribution
Class I ALDH130 µMLiver and many other tissues
Class II ALDH2<1 µMLow levels in most tissues with the expression highest in liver compared to kidney and muscle
Class III ALDH311 mMStomach, liver, cornea

Share and Cite

MDPI and ACS Style

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. https://doi.org/10.3390/biomedicines8030050

AMA Style

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(3):50. https://doi.org/10.3390/biomedicines8030050

Chicago/Turabian Style

Jiang, Yanchao, Ting Zhang, Praveen Kusumanchi, Sen Han, Zhihong Yang, and Suthat Liangpunsakul. 2020. "Alcohol Metabolizing Enzymes, Microsomal Ethanol Oxidizing System, Cytochrome P450 2E1, Catalase, and Aldehyde Dehydrogenase in Alcohol-Associated Liver Disease" Biomedicines 8, no. 3: 50. https://doi.org/10.3390/biomedicines8030050

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