Next Article in Journal
Flavonoid–Phenolic Acid Hybrids Are Potent Inhibitors of Ferroptosis via Attenuation of Mitochondrial Impairment
Next Article in Special Issue
Natural Products as Dietary Agents for the Prevention and Mitigation of Oxidative Damage and Inflammation in the Intestinal Barrier
Previous Article in Journal
The Binding of HSPA8 and Mitochondrial ALDH2 Mediates Oxygen-Glucose Deprivation-Induced Fibroblast Senescence
Previous Article in Special Issue
The Antioxidant Properties of Salvia verbenaca Extract Contribute to Its Intestinal Antiinflammatory Effects in Experimental Colitis in Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Melatonin Prevents Alcohol- and Metabolic Dysfunction- Associated Steatotic Liver Disease by Mitigating Gut Dysbiosis, Intestinal Barrier Dysfunction, and Endotoxemia

by
Karli R. LeFort
*,
Wiramon Rungratanawanich
and
Byoung-Joon Song
*
Section of Molecular Pharmacology and Toxicology, National Institute on Alcohol Abuse and Alcoholism, 9000 Rockville Pike, Bethesda, MD 20892, USA
*
Authors to whom correspondence should be addressed.
Antioxidants 2024, 13(1), 43; https://doi.org/10.3390/antiox13010043
Submission received: 21 November 2023 / Revised: 19 December 2023 / Accepted: 22 December 2023 / Published: 25 December 2023
(This article belongs to the Special Issue Significance of Antioxidant Mechanisms in Intestinal Inflammation)

Abstract

:
Melatonin (MT) has often been used to support good sleep quality, especially during the COVID-19 pandemic, as many have suffered from stress-related disrupted sleep patterns. It is less known that MT is an antioxidant, anti-inflammatory compound, and modulator of gut barrier dysfunction, which plays a significant role in many disease states. Furthermore, MT is produced at 400–500 times greater concentrations in intestinal enterochromaffin cells, supporting the role of MT in maintaining the functions of the intestines and gut–organ axes. Given this information, the focus of this article is to review the functions of MT and the molecular mechanisms by which it prevents alcohol-associated liver disease (ALD) and metabolic dysfunction-associated steatotic liver disease (MASLD), including its metabolism and interactions with mitochondria to exert its antioxidant and anti-inflammatory activities in the gut–liver axis. We detail various mechanisms by which MT acts as an antioxidant, anti-inflammatory compound, and modulator of intestinal barrier function to prevent the progression of ALD and MASLD via the gut–liver axis, with a focus on how these conditions are modeled in animal studies. Using the mechanisms of MT prevention and animal studies described, we suggest behavioral modifications and several exogenous sources of MT, including food and supplements. Further clinical research should be performed to develop the field of MT in preventing the progression of liver diseases via the gut–liver axis, so we mention a few considerations regarding MT supplementation in the context of clinical trials in order to advance this field of research.

Graphical Abstract

1. Introduction: MT

Melatonin (MT), known as N-acetyl-5-methoxytryptamine, is a sleep-regulating hormone known to be secreted from the hypothalamus–pineal gland axis [1]. However, recent reports demonstrate that the gastrointestinal tract (GIT) is responsible for producing 400 [2] to 500 [3] times more MT than the pineal gland, suggesting that MT may be more prominent in regulating the gut and other peripheral organs [3,4,5].
This review details how MT made from the intestines interacts in the gut–liver axis to prevent liver diseases. We have explained that MT produced in intestinal mitochondria targets hepatic mitochondria by regulating oxidative stress mainly generated by one-electron leakage from the electron transport chain [6] as well as from the activities of cytochrome P450-2E1 (CYP2E1), inducible nitric oxide synthetase (iNOS), and NADPH-dependent oxidases (NOXs). Consequently, MT attenuates mitochondrial dysfunction, and mitigates metabolic alterations and inflammatory processes that are involved in the progression of liver diseases. For instance, MT prevents the worsened progression of alcohol-associated liver disease (ALD) and metabolic dysfunction-associated steatotic liver disease (MASLD) by protecting the intestines against gut dysbiosis, intestinal barrier dysfunction, and endotoxemia.

1.1. Synthesis of MT

MT synthesis mainly occurs in the brain and intestines. In the brain, MT synthesis occurs in pinealocytes, which are photoreceptor cells [7,8,9]. Intestinal MT synthesis occurs in enterochromaffin cells in the mucosal layer of the villi [10,11]. MT production in the brain and gut is highly influenced by environmental stimuli such as the light–dark cycle or dietary exposures to the precursor of MT, L-tryptophan [7,8,9]. It is theorized that intestinal MT synthesis is stimulated mainly by feeding cues, and photoreceptor synthesis of MT (stimulated by light–dark cycles) occurs within an opposite phase, suggesting that MT is constantly being synthesized [11]. MT production in the gut may also be inversely related to cortisol levels, as these two hormones are involved in circadian rhythm-coordinated metabolism in the brain and many peripheral tissues [1,5,12].
In all cells that synthesize MT, L-tryptophan is hydroxylated to 5-hydroxytryptophan in mitochondria and then converted to 5-hydroxytryptamine (serotonin) in the cytoplasm [13]. Serotonin is then acetylated to N-acetylserotonin and converted to MT [13]. In total, four enzymes including tryptophan-5-hydroxylase (TPH), L-aromatic amino acid decarboxylase (AADC), arylalkylamine N-acetyltransferase (AA-NAT), and N-acetylserotonin-O-methyltransferase (ASMT), as well as three membrane receptors including MT1, MT2, and MT3 are involved in MT production and signaling [14].
While MT supplementation is widely known, sufficient amounts of MT can be consumed in foods high in its precursor, L-tryptophan, and in organisms associated with food cultivation. MT can be produced from many types of bacteria, protists, fungi, macroalgae, plants, and animals and is abundant in fruits, vegetables, nuts, seeds, grains, and olive oil (see Section 5 for more sources) [15,16,17,18]. In general, the Mediterranean Diet also contains MT, but it should be noted that foods have certain health benefits via the synergistic effects of the combination of compounds in their composition [12]. MT may even be produced by human commensal bacteria, as reviewed [3,4,5,19,20].
MT stores are distributed throughout the body in the intestines, brain, eyes, skin, bones, blood, immune system, thymus, spleen, GIT, inner ear, pancreas, liver, kidneys, reproductive tract, upper respiratory tract, heart, and thyroid [5]. MT is also found in many bodily fluids, including blood, amniotic fluid, synovial fluid, bile, saliva, breast milk, and cerebrospinal fluid (CSF) [12]. Importantly, it should be noted that MT accumulates in these tissues and contributes to the circadian rhythm of all these tissues/fluids. Likewise, the metabolic functions of these tissues are interrelated and are affected by depleted MT stores. MT has been shown to regulate the functions of the immune system, heart, gut, liver, kidney, reproductive organs, and endocrine glands and to act in organ axes to prevent chronic and acute conditions [21,22].
Given the number of cells in which MT is stored and synthesized, it is essential to support sufficient biological stores of MT in our cells for optimal homeostatic communication and prevention of liver diseases. In sum, this review details how supplementation and restored levels of MT affect several components in the gut–liver axis to prevent the progression of ALD and MASLD.

1.2. Contributing Factors of ALD within the Gut–Liver Axis

The liver is the main site of oxidative and non-oxidative ethanol metabolism, including enzymes such as alcohol dehydrogenase, aldehyde dehydrogenase 2 (ALDH2), Cytochrome P450-2E1 (CYP2E1), and iNOS [23,24]. Redox imbalance [25], poor nutritional status [26], mitochondrial dysfunction [27], cellular injury (apoptosis/necrosis) [28], inflammation [29], altered cell signaling pathways [30], and endotoxemia [31] all contribute to the pathology of ALD [25,26,27,28,29,30,31].
Fat accumulation (steatosis) can happen from increased de novo lipid synthesis, lipid transport from adipose tissues, and mitochondrial dysfunction that causes decreased fat degradation [32]. However, alcohol-induced accumulation of reactive oxidative species (ROS) [33], lipid peroxides (LPOs) (e.g., acrolein, malondialdehyde (MDA), and 4-hydroxynonenal (4-HNE)) [34], and hepatocyte death pathways may all contribute to the activation of hepatic stellate cells (HSCs), which then stimulate fibrosis and cirrhosis, leading to hepatocellular carcinoma (HCC) or liver failure [32]. One study demonstrated that ROS-, LPO- and hepatocyte apoptosis-mediated HSC activation may take place via the increased production and accumulation of α-smooth muscle actin (α-SMA) and collagen in the extracellular matrices, upregulated pro-fibrogenic metalloproteinase-2 (MMP2), and downregulated anti-fibrogenic metalloproteinase-1 (MMP1) via the MDA/4-HNE pathway [25,35,36].
Chronic and binge alcohol exposure can cause intestinal barrier dysfunction, worsening the progression of liver disease [37,38,39,40]. Elevated intestinal permeability makes endotoxemia a likely outcome, measured by increased serum LPS [25,41]. Acetaldehyde, a highly reactive aldehyde produced during oxidative ethanol metabolism, can increase permeability in the intestinal barrier by disrupting the distribution and organization of tight junction and adherens junction (TJ/AJ) proteins in the intestines [25]. Alcohol (ethanol) also changes the composition of the microbiome in ALD [31,42,43,44,45], even at the early stages of ALD, causing dysbiosis and bacterial translocation [45]. Even AUD patients exhibit microbial characteristics such as upregulation of GABA metabolism pathways that could serve as intestinal fingerprint biomarkers with a 93% accuracy [29,46].
Alterations of ALDH2, CYP2E1, and iNOS can also contribute to intestinal permeability and endotoxemia in the progression of ALD. Inhibition of ALDH2 can directly increase intestinal permeability via elevated acetaldehyde, as demonstrated in Aldh2-KO mice exposed to the alcohol liquid diet for four weeks [47] or binge alcohol [48]. LPS released from the gut interacts with TLR4 in specialized macrophages in the liver (Kupffer cells) and other immune cells to stimulate an inflammatory response and ROS production [49]. These changes promote hepatocyte damage, activate hepatic stellate cells, and contribute to fibrosis [49].
Another cellular mechanism of hepatic inflammation via the gut–liver axis may happen when LPS-mediated induction of iNOS results in toxic peroxynitrite production in the presence of ROS [23,24]. Elevated peroxynitrite can nitrate or S-nitrosylate proteins and promote the accumulation of 4-HNE protein adducts in mitochondria [50,51]; these post-translational modifications (PTMs) of various proteins in many subcellular compartments, including mitochondria, stimulate mitochondrial dysfunction and caspase-mediated apoptosis of hepatocytes [50,51].
Lastly, CYP2E1 is a significant contributor to alcohol-induced oxidative stress and ALD in periods of excessive alcohol use [36,52,53,54,55,56]. CYP2E1 is expressed in the intestine, induced by alcohol exposure, and involved in oxidative intestinal injury [57,58]. Specifically, it is induced by protein stabilization (i.e., protection from its rapid degradation via the ubiquitin-proteosome-dependent proteolytic pathway) after alcohol exposure [59,60]. CYP2E1-mediated ROS can also stimulate endotoxemia and elevate serum levels of LPS, which can travel to the liver, upregulating pro-inflammatory mediators, including tumor necrosis factor-alpha (TNF-α) [61] and hypoxia-inducible factor 1-alpha (HIF1-α) [49]. Elevated TNF-α and HIF1-α can activate mitogen-activated protein kinases (MAPKs) and decrease autophagy [49,61]. Upregulated CYP2E1 activity can also promote various PTMs, such as oxidation, S-nitrosylation, nitration, MAPK-mediated phosphorylation, acetylation, and adduct formations in mitochondria and endoplasmic reticuli, leading to mitochondrial dysfunction, endoplasmic reticulum (ER) stress, and apoptosis of hepatocytes and other cells like gut enterocytes [24,62]. CYP2E1-mediated oxidative stress can also sensitize the liver to toxicity via endotoxemia and intestinal TNF-α via a CYP2E1-thioredoxin-ASK1-JNK1 pathway [36,52,53,54,55,56,57].

1.3. Contributing Factors of MASLD via the Gut–Liver Axis

The incidence of obesity and metabolic syndrome has steadily increased over the last two decades. Obesity is affiliated with insulin resistance and metabolic syndrome, which may negatively affect the function of the liver, in addition to the heart, blood vessels, kidneys, and muscle mass [63]. MASLD and metabolic dysfunction-associated steatohepatitis (MASH) are considered co-morbidities of obesity and Type II Diabetes Mellitus that may be caused by substances other than alcohol, such as fructose/sucrose and Western-style high-fat diets (HFDs) containing high levels of n-6 fatty acids [64,65]. MASLD may also have hepatic manifestations similar to ALD. Like the progression of steatosis in ALD, increased de novo fat synthesis, fat transport from adipose tissues, and decreased fat degradation are typically seen in MASLD/MASH [66]. Oxidative and nitrative stress can significantly contribute to the progression of MASLD/MASH, partly via the involvement of CYP2E1 [30,67,68,69], additional oxidative stress [70], and various PTMs [30,62,67,68,69,70]. Oxidative stress and PTM accumulation can ultimately lead to mitochondrial dysfunction with decreased lipid degradation and insufficient energy (ATP) supplies [71]. CYP2E1 has been found to contribute to liver injuries caused by many substances including high fructose [64], HFDs [72], carcinogens [73], narcotics [74], pharmacotherapies [75], acetaminophen (APAP) [76,77], hepatotoxic substances to model MASLD (i.e., carbon tetrachloride (CCl4) [78,79] and thioacetamide (TAA) [78]), and nicotine [80]. CYP2E1 can also play a causal role in creating oxidative stress in MASLD [30,68,72,81] and insulin resistance [52,81,82,83].
It is well established that multiple hits are needed to initiate and develop MASLD/MASH [84]. MASLD has a wide variety of appearances, from as little as a simple accumulation of lipid droplets to active inflammation seen in MASH, with a small part progressing to fibrosis and HCC, and during this progression, multiple events occur. The original report of the “Two-hit Hypothesis” [84] described the development of steatosis as the first hit, and increased oxidative stress represents part of the second hit in the progression of MASLD [84]. According to others, the second hits also include intestinal barrier dysfunction [46], endotoxemia [46], inflammation [46], changed hepatocyte apoptosis pathways, and stimulated HSCs, all of which allow the evolution of milder cases of MASLD to progress to more severe cases, including MASH and fibrosis [46,66,85,86]. Others build on this hypothesis by suggesting that the “Multiple-hit Hypothesis” involves dysregulated lipid metabolism, mitochondrial dysfunction with suppressed fat degradation, and oxidative stress [66,85,86]. However, it is still unclear whether MASLD stimulates mitochondrial dysfunction and oxidative stress or if mitochondrial dysfunction and oxidative stress cause MASLD [66,85,86].
An explanation for this phenomenon is that chronic inflammation triggers oxidative stress, which stimulates more inflammation in a vicious cycle during the progression of MASLD [68,81,87,88]. During the phase of steatosis, mitochondrial respiration is increased to meet the elevated need for energy [89]. Mitochondrial respiration will increase the production of ROS and activate antioxidant responses [89]. Lipid accumulation may manifest as an excess of free fatty acids (FFAs), which can activate stress-activated c-Jun protein kinase JNK, leading to apoptosis of hepatocytes accompanied by damaged mitophagy of mitochondria [89]. Increased inflammation and oxidative stress trigger apoptosis, mitochondrial respiration, mitophagy, and antioxidant pathways that may compensate depending on the conditions. During cirrhosis, few compensatory mechanisms exist, suggesting that there are low levels of respiration, biogenesis, antioxidant pathways, or mitophagy. Further stimulation of nonparenchymal cells, such as HSCs, as well as inflammation, oxidative stress, and increased apoptosis of hepatocytes, occur at later stages of liver diseases [90,91].
Gut dysbiosis and endotoxemia are certainly causal risk factors in MASLD/MASH [28,92] and other liver disorders [93,94], and the intestinal “fingerprint” caused by the susceptibility and progression of MASLD differs from that of ALD [94]. For example, one study found that while patients with ALD tend to have a decreased abundance of Ruminococcus and Faecalibacterium prausnitzii, patients with MASLD had an increased abundance of Proteobacteria, Enterobacteriaceae, and Escherichia [94]. Furthermore, microbial products or metabolites such as LPS, short-chain fatty acids (SCFAs), bile acids, choline, trimethylamine-N-oxide (TMAO), and endogenously produced ethanol in the gastrointestinal tract [95,96,97] have been found to accumulate in the progression of MASLD [64,95,96,97,98]. More importantly, the intestines contribute to the progression of MASLD via excessive oxidative stress and systemic inflammation from leaked LPS [99]. The intestines may even alter lipid metabolism pathways via microbial SCFA metabolites [100] and disrupt mitochondrial function, leading to hepatocyte death pathways, inflammation, and fibrosis [101].

1.4. Functions of MT in the Gut–Liver Axis

The liver and gut contain cytochrome P450 isoforms involved in the deacetylation of N-acetylserotonin to form MT and the subsequent formation of 6-hydroxy-melatonin [102]. The pineal gland is responsible for the hydroxylation and decarboxylation of tryptophan to MT. However, the liver and intestine are among the few organs that express AA-NAT and ASMT enzymes to synthesize MT from serotonin [103]. AA-NAT and ASMT handle acetylation and then methylation of serotonin molecules to MT, respectively [3,5,104]. MT is also metabolized by cytochrome P450 isoforms in the liver when ingested orally or administered intraperitoneally [5]. Given its metabolism and localization in the liver and intestines, MT is likely to exert its beneficial effects on the gut–liver axis.
The metabolism of MT contributes to its unique function to combat oxidative stress in the gut-mitochondria axis. To begin, MT is a unique antioxidant because it produces metabolites that scavenge free radicals as well. MT can donate electrons to two free radicals or be hydroxylated to produce N-acetyl-N-formyl-5-methoxykynuramine (AFMK), which is then oxidized to another metabolite called N-acetyl-5-methoxykynuramine (AMK) [105,106]. AMK has been demonstrated to have many effects on oxidative stress in mitochondrial metabolism, including inhibiting and downregulating cyclooxygenases and suppressing iNOS and mitochondrial nitric oxide synthase (mtNOS) enzymes [106]. MT also directly upregulates the electron transport chain, oxidative phosphorylation, and ATP synthesis in mitochondria by interacting with complexes I and IV in healthy cells. In addition, MT has been shown to recover glutathione levels and decrease the activity and expression of iNOS and mtNOS in models of septic damage to mitochondria [107,108]. Thus, MT has an exponential antioxidant effect on mitochondrial metabolism by directly scavenging free radicals and producing metabolites that also scavenge free radicals.

1.4.1. Intestinal Barrier Dysfunction

The function of MT in intestinal health may be revealed via studies on the effects of intestinal MT deficits in sleep-deprived clinical and animal models. MT deficits have been reported in night shift workers showing signs of intestinal barrier dysfunction and increased sensitivity to oxidative stress, inflammation, and several diseases, including ALD, MASLD, ischemic liver injury, and other metabolic disorders that may relate to intestinal barrier dysfunction [109,110]. It has been well documented that having a deficit of MT can result in multi-organ damage, most notably those tied to the gut [41]. These models of disrupted circadian rhythms may be correlated with elevated gut dysbiosis and intestinal barrier dysfunction [111,112]. A clinical study involving people with alcohol use disorder (AUD) reported decreased hours of sleep and MT levels, as well as increased intestinal hyperpermeability and endotoxemia [113], as recently reviewed [114]. Based on these studies, MT deficits are associated with alcohol-induced susceptibility to intestinal barrier dysfunction.

1.4.2. Oxidative Stress and PTMs in Mitochondria

MT is primarily localized in the mitochondria of many cells [115,116,117,118], maintaining energy homeostasis [116,119,120] and reducing oxidative stress. Reactive oxygen species (ROS) are mainly produced from the one-electron leakage from the electron transport chain [6]. ROS may also be produced by the ethanol-inducible CYP2E1 enzyme [62,121], which is typically localized in microsomes but may translocate to mitochondria [122,123,124], causing mitochondrial dysfunction and fat accumulation [62,125]. MT also counteracts oxidative stress because it improves the functions of mitochondria in hepatocytes [126]. MT receptors, including MT1, MT2, and MT3, are localized in the gut, and MT3, also known as quinone reductase 2, has been known to have antioxidant properties [127]. MT also up-regulates NAD+-dependent protein-deacetylase Sirtuins (SIRTs) such as Sirtuin-1 (SIRT1) [128,129,130,131,132], Sirtuin-3 (SIRT3) [117,118,131,132,133,134,135,136,137], and Sirtuin-6 (SIRT6) [138,139], as shown in Figure 1.
Sirtuins prevent the accumulation of protein acetylation, one type of PTM, in mitochondria, preventing mitochondrial dysfunction in liver diseases and aging [140]. MT also promotes mitophagy and autophagy in cells [134,135,137,141,142,143,144,145] and reduces apoptosis [146,147], protecting various organs from acute tissue injury and aging-related chronic diseases. In later sections, we will discuss more specific mechanisms by which MT exerts its effects as an antioxidant to prevent chronic and acute liver injuries via the gut–liver axis [128,148,149].

1.4.3. Gut–Mitochondria Axis

Changes in the intestinal microbiota (i.e., gut dysbiosis) can lead to increased intestinal permeability and local inflammation, which can cause endotoxemia and decrease the production of SCFAs [150,151]. A high abundance of Gram-negative bacteria typically contributes to increased LPS and lowered SCFA production, which is commonly seen in intestinal barrier dysfunction and endotoxemia [152]. Consequently, these two events promote an inflammatory-related pathway, which causes oxidative stress, negatively affecting mitochondrial functions and the production of MT [42]. However, MT can positively regulate its own production by increasing acetyl-CoA production from the pyruvate dehydrogenase complex, which activates the mitochondrial melatonergic pathway as a co-substrate [42], as shown in Figure 1.

2. Melatonin in the Gut–Liver Axis of ALD

Circadian disruptions make the intestines more susceptible to alcohol-induced tissue injuries [153]. Because several microbial changes and intestinal MT alterations occur in the progression of ALD, MT supplementation has the potential to mitigate alcohol-induced gut dysbiosis and liver injury. More specifically, MT has a profound impact in the context of alcohol-induced intestinal permeability in the gut–liver axis. In one study, alcohol that was metabolized with CYP2E1 with subsequent ROS byproducts enhanced the expression of circadian (i.e., CLOCK and PER2) proteins (loosely affiliated with MT), which were later shown to be involved in ethanol-induced intestinal hyperpermeability [154]. Circadian disruptions (perhaps due to abnormal MT levels) from irregular patterns of eating, sleeping, working, and drinking should be minimized for people who struggle with AUD or ALD [154]. Related to circadian genes, MT depletion and circadian disruption in human studies increased night shift workers’ susceptibility to intestinal permeability during social drinking via the upregulated transcription of CLOCK and BMAL1 genes [155]. Depleted MT was also a sign of increased intestinal permeability in people with AUD who may suffer from sleep loss as a side effect of alcohol consumption [113]. Withdrawal from chronic alcohol consumption in people struggling with AUD was shown to increase MT levels because secretion of MT is regulated by the noradrenergic system, which is activated when withdrawing from alcohol [156].
MT supplementation has a unique set of roles in preventing alcohol-related disorders. First of all, MT exerts an anti-stressor effect which includes (a) antioxidant effects (i.e., neutralizing hydroxyl radicals, lipid peroxides, and peroxynitrite, and regulating the activities of glutathione peroxidase, superoxide dismutase, glucose 6-phosphate dehydrogenase (G6PDH), and nitric oxide synthase) and (b) anti-inflammatory effects to prevent aging [114]. MT also has been implicated in various signaling pathways such as the epidermal growth factor receptor–Brahma-related gene-1–telomerase reverse transcriptase (EGFR-BRG1-TERT) axis [157], Sirtuin1-Wnt/β-catenin-NOD-, LRR- and pyrin domain-containing protein 3 (SIRT1-Wnt/β-catenin-NLRP3) axis [158,159], SIRT1-Cereblon-Yin Yang 1-CYP2E1 (SIRT1-CRBN-YY1-CYP2E1) axis [160], and matrix metalloproteinase-9–nuclear factor–kappa B (MMP9-NF-κB) axis [161]. MT can also exhibit hepatoprotective effects when taken with other drugs, such as celecoxib (a nonsteroidal anti-inflammatory drug), by suppressing alcohol-induced apoptosis and inflammation via JNK and TNF-α signaling cascades and by mitigating oxidative stress [162]. MT also acts comparably to metformin [163] by upregulating the activity of AMPK and attenuating alcohol-related lipid accumulation to mitigate steatosis [164]. Specific to ALD and AUD, MT targets circadian rhythm disorders, alcohol withdrawal, and sleep apoplexy while acting as an antidepressant and sedative [22].
The preventive/therapeutic effects of MT on interconnected organ systems have been shown in many studies on ALD affecting the gut–liver axis [109,165]. MT has been shown to attenuate sub-clinical LPS and oxidative stress caused by alcohol [166] and to ameliorate alcohol-induced bile acid synthesis by enhancing miR-497 expression [167].

Translational Research on the Effects of MT on the Progression of ALD

Many laboratories have reported the effects of MT on ALD in various types of animal studies (Table 1).

3. MT in the Gut–Liver Axis of MASLD

The kynurenine, indole, serotonin, and MT pathways are interconnected, and tryptophan metabolism is an integral driver in the progression of MASLD via the gut–liver axis and the immune system [168]. Related to the gut–liver axis, intestinal MT production levels, in sync with feeding cycle cues, may compensate for many factors of overnutrition accompanied with the progression of MASLD. For example, MT supplementation could potentially treat the obesity-related vicious cycle of insulin resistance, lipogenesis, and oxidative stress in the progression of MASLD/MASH by contributing to more brown adipose tissue (rich in mitochondria) in the body [169,170]. One study detailing the beneficial role of MT in managing obesity demonstrated that MT supplementation stimulated thermogenesis in brown fat cells and attenuated lipogenesis in white fat cells and hepatocytes to support that MT can prevent the development or progression of MASLD caused by a Western-style HFD [171]. MT may also regulate feeding hormones such as ghrelin [20] and the inflammatory processes [172,173,174] that commonly contribute to obesity-related MASLD [20].
The preventive and therapeutic effects of this antioxidant-like hormone have been shown to positively affect the gut–liver axis in multiple animal studies modeling MASLD [168,175,176,177]. MT has been shown to inhibit the metabolic effects of fructose [178], which have been found to contribute to MASLD [64,179]. MT has also been shown to alleviate gut-related liver inflammation from Ochratoxin A by restoring TJ/AJ proteins in the physical barrier of the gut, restoring liver function, and returning inflammatory markers connecting the gut and liver (i.e., LPS, interleukins, and TNF-α) [180]. Catalase plays a role in MASLD by preserving mitochondrial function, and MT prevents MASLD by supporting the effects of catalase [181]. Catalase-KO mice (CKO) exposed to HFD showed cellular lipid accumulation and decreased mitochondrial biogenesis, which were both treated with MT administration [181]. Overall, MT prevented fatty liver development and maintained the mitochondrial integrity of hepatocytes [181]. In another model of a short-term HFD, MT was able to reduce the effects of several pathological factors on the gut–liver axis and fat accumulation in the liver [176]. MT has also acted in the orphan nuclear receptor subfamily 4 group A member 1/DNA-dependent protein kinase catalytic subunit/tumor protein 53 (NR4A1/DNA-PKcs/p53) axis to reduce mitochondrial fission and promote mitophagy to prevent MASLD [142]. Some theorize that MT can even prevent HCC, which has been known to be irreversible [165].

Translational Research on the Effects of MT in MASLD

Many laboratories have reported the effects of MT on the progression of MASLD in various animal models (Table 2).

4. Acute Toxicity in the Gut–Liver Axis: MT and Septic Hepatotoxicity

MT has been shown to affect septic hepatotoxicity, an acute liver condition, in similar ways to the way by which it prevents the beginning stages of ALD and MASLD. In one model of LPS-induced acute hepatotoxicity by cecal ligation and puncture, MT upregulated SIRT3 and SIRT1, which mitigated oxidative stress (by increased SOD2 activity via protein deacetylation), preserved mitochondrial function, and mitigated autophagy of intestinal epithelial cells. In this model, MT also attenuated inflammation by upregulating deacetylated nuclear factor-kappa B (NF-κB) and interleukin-10 (IL-10), and decreasing serum levels of TNF-α and interleukin-6 (IL-6) in the gut–liver axis [185]. In another septic hepatotoxicity study, MT alleviated LPS-induced hepatic SREBP-1c activation. It also mitigated lipid accumulation in mice which was shown via the significant decrease of liver weight, liver-to-body weight ratio, serum levels of very low density lipoproteins (VLDL), serum triglyceride (TG) levels, SREBP-1 expression, fatty acid synthase (FAS) mRNA expression, and acetyl-CoA carboxylase (ACC) mRNA expression, and increased liver X receptor α (LXR-α) expression [183]. These results suggest the beneficial role of MT as an alleviating compound in the beginning stages of endotoxin-evoked MASLD [183]. MT also mitigates cellular stress in the liver of septic mice by reducing ROS and increasing the unfolded protein response via the upregulation of PKR-like ER kinase (PERK) and C/EBP homologous protein (CHOP), and preventing the downregulation of cyclic AMP-response element-binding protein H (CREBH) [186].

5. Concluding Remarks

5.1. Suggestion of Behavioral Changes That May Increase MT to Prevent or Treat Liver Diseases

To efficiently manage ALD and MASLD, we should understand the underlying mechanisms of liver disease stages, including the bidirectional interactions of the gut–liver axis [88,150,187]. One example is that compensatory mechanisms occur to mitigate oxidative stress at the beginning stages of MASLD, but these compensatory mechanisms tend to fail at later, irreversible stages (i.e., cirrhosis) [88,187].
Exercise effectively prevents MASLD by mitigating oxidative stress and inflammatory factors, reducing intrahepatic fat content, increasing β-oxidation of fatty acids, overexpressing PPAR-γ, and attenuating apoptosis [188]. However, it is crucial to exercise at wakeful moments of the day because too much exercise during restful hours disrupts MT secretion [189], which does not allow for the prevention of liver diseases if MT is already low in biological stores.
Other lifestyle changes can be used to target oxidative stress to ameliorate liver and intestinal diseases, such as losing weight, eating beneficial diets with probiotics, and eliminating liver disease risk factors (e.g., overconsumption of alcohol, candy, and high-fructose corn syrup, and fast food-HFD) [70]. However, compliance with lifestyle changes could be improved. Increasing one’s antioxidant supply to maintain a homeostatic balance of ROS and antioxidants could be a promising approach and make patients more likely to stay compliant. Using antioxidants (like MT) that target mitochondria has a tremendous therapeutic potential to prevent and treat oxidative stress-related disorders because they cross the mitochondrial membranes [90,190].
Many dietary factors have been shown to prevent gut dysbiosis and LPS-induced ALD and MASLD that may be related to low MT production. Prebiotics (e.g., oligofructose, galactose, and inulin) and probiotics (e.g., Bifidobacterium longum, Bifidobacterium infantis, Lactobacillus plantarum, and Lactobacillus acidenophilus) [191] may have a role in upregulating MT production, possibly via bacterial L-tryptophan production (shown in a mouse model of ALD) [192]. Foods containing polyphenols (i.e., grape seed proanthocyanidins, resveratrol, quercetin, genistein, and isoflavones), other dietary compounds (i.e., rhein, phlorizin, capsaicin, rutin, lycopene, broccoli sprout extract, cranberry extract, and green tea extract), seeds (i.e., Alfalfa, almonds, anise, black mustard, celery, coriander, fennel, fenugreek, flax, green cardamom, milk thistle, poppy, sunflower, white mustard, and wolf berries), nuts (i.e., walnuts), fruits (i.e., apples, bananas, grapes, kiwis, pineapples, strawberries, tart cherries, and tomatoes), vegetables (i.e., asparagus, cabbage, carrots, celery, cucumbers, Indian spinach, Japanese radishes, beets, and corn), starches (i.e., oats, rice, tall fescue, and barley), and oil (i.e., olive oil) are all sources of MT, with seeds being the richest sources of MT [191,193,194]. In general, the Mediterranean Diet is a rich source of MT and has been demonstrated to prevent gut dysbiosis as well as intestinal and systemic inflammation [194,195].

5.2. Discussion of Future Studies and Considerations

Eating foods that contain MT may prevent the progression of liver diseases via the gut–liver axis, but if food consumption is not enough to raise serum MT levels then supplementation may be necessary [194]. The United States Food and Drug Administration does not regulate MT or supplement use [196]. In other countries, MT is prescribed and used for its intended purposes. In the United States, continuing the regulation of MT as a supplement may be part of the reason why 1 in 8 people taking MT take more than 5 milligrams (resulting in peak concentrations that last more than 4–8 h) and why it is being utilized for other benefits than its intended purpose of treating circadian rhythm disorders [197]. Supplementation should not be used for medical use until it is FDA-regulated, but if MT is administered then serum 6-hydroxymelatonin levels should be measured. A clinical trial in healthy men noted that in groups given 20 mg, 30 mg, 50 mg, or 100 mg MT, serum levels of the MT metabolite, 6-hydoxymelatonin sulfate, fluctuated less between participants in the same dosage groups than serum MT levels [198], reflecting that perhaps 6-hydroxymelatonin sulfate should be a biomarker of MT metabolism in the body.
Few clinical studies on MT supplementation in patients with ALD or MASLD have been detailed, especially from the perspective of the gut–liver axis, and this deserves more research [194,199]. However, MT has been found to be safe for people undergoing major liver surgery in doses of 50 mg/kg of body weight [200]. Since cirrhotic patients produce less MT [174] and may have high Model for End-Stage Liver Disease (MELD) scores that make them eligible for liver transplantation surgeries, MT supplementation may serve multiple purposes for cirrhotic patients [174,200]. Perhaps some of the animal studies detailed in Table 1 and Table 2 may be translated into clinical research study designs in order to promote this field of research.
One reason and consideration for designing MT supplementation clinical trials is that MT has been shown to be safe for supplementation in clinical studies. It can be taken in amounts up to 300 mg/day [157,198,201,202,203,204,205,206,207], with only reported mild side effects of dizziness, sleepiness, nausea, and headaches [204,208,209]. One case report detailed a female successfully using MT as her primary treatment for Multiple Sclerosis in a range of 50–300 mg daily for four years [210].
This brings us to the next consideration for further research, which is using MT as a complementary therapy for liver diseases. Co-administration with MT has been shown to increase the efficacy of drugs in treating and preventing the onset of liver diseases [211,212]. MT may also be taken in tandem with drugs that cause oxidative stress because it may allow higher doses of drugs to be used in cases where their usages are limited by toxicity [162,211]. MT has not been shown to interfere with drugs, but for future studies, for drug-drug interactions with compounds that are also metabolized by CYP1A2 [213], TPH, AADC, AA-NAT, or ASMT should be considered. Expanding our understanding of how MT co-administration may prevent liver diseases via the gut–liver axis in clinical trials is a promising direction for the future of liver disease research.

5.3. Conclusions

In this review, we summarized how MT may exert its benefits within the gut–liver axis to prevent the development and progression of ALD and MASLD. MT has many functions in mitigating oxidative stress and LPS-induced inflammation, preventing damage to the intestines and liver, and replenishing mitochondrial antioxidant levels. We also detailed many animal models in which MT supplementation prevents the progression of liver diseases and have discussed multiple exogenous sources of MT and lifestyle factors that affect human MT levels. Lastly, we considered aspects of MT supplementation in clinical trials, including its safety and information on the few known MT and liver disease clinical trials, with the hope of expanding the field of research in MT supplementation for the prevention and treatment of liver diseases via the gut–liver axis.

Author Contributions

Manuscript conceptualization: K.R.L. and B.-J.S.; Literature search and article acquisition: K.R.L. and B.-J.S.; Writing drafts and revision: K.R.L. and B.-J.S.; Figure drawing and table conceptualization: K.R.L.; Proofreading of the drafts and critical comments: K.R.L., W.R. and B.-J.S.; Final manuscript review and editing: K.R.L., W.R. and B.-J.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Intramural Research Fund (to B.J.S.) from the National Institute on Alcohol Abuse and Alcoholism.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Figure 1 was created with software from Biorender.com.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ALD, alcohol-associated liver disease; ALDH2, mitochondrial aldehyde dehydrogenase 2; AUD, alcohol use disorder; CKO, catalase-knockout; DNA-PKcs, DNA-dependent protein kinase catalytic subunit; GIT, gastrointestinal tract; HFD, Western-style high fat diet; 4-HNE, 4-hydroxynonenal; HSCs, hepatic stellate cells; IL-6, interleukin-6; IL-10, interleukin-10; iNOS, inducible nitric oxide synthetase; LPOs, lipid peroxides; LPS, lipopolysaccharide; MAPKs, mitogen-activated protein kinases; MASH, metabolic dysfunction-associated steatohepatitis; MASLD, metabolic dysfunction-associated steatotic liver disease; MDA, malondialdehyde; MT, melatonin; NOX, NADPH-dependent oxidase; NRF2, Nuclear factor erythroid 2-related factor 2; NR4A1, orphan nuclear receptor subfamily 4 group A member 1; PTMs, post-translational modifications; P53, tumor protein 53; ROS, reactive oxygen species; SCFAs, short chain fatty acids; Serotonin, 5-hydroxytryptamine; SIRT1, Sirtuin1; SIRT3, Sirtuin3; SIRT6, Sirtuin6; SOD, superoxide dismutase; TGs, triacylglycerols, triglycerides; TNF-α, tumor necrosis factor-alpha; TJ/AJ, tight junction and adherens junction proteins.

References

  1. Baekelandt, S.; Mandiki, S.N.M.; Kestemont, P. Are cortisol and melatonin involved in the immune modulation by the light environment in pike perch Sander lucioperca? J. Pineal Res. 2019, 67, e12573. [Google Scholar] [CrossRef] [PubMed]
  2. Bubenik, G.A.; Brown, G.M. Pinealectomy reduces melatonin levels in the serum but not in the gastrointestinal tract of rats. Neurosignals 1997, 6, 40–44. [Google Scholar] [CrossRef] [PubMed]
  3. Konturek, S.J.; Konturek, P.C.; Brzozowski, T.; Bubenik, G.A. Role of melatonin in upper gastrointestinal tract. J. Physiol. Pharmacol. 2007, 58 (Suppl. S6), 23–52. [Google Scholar] [PubMed]
  4. Bubenik, G.A. Gastrointestinal melatonin: Localization, function, and clinical relevance. Dig. Dis. Sci. 2002, 47, 2336–2348. [Google Scholar] [CrossRef]
  5. Tordjman, S.; Chokron, S.; Delorme, R.; Charrier, A.; Bellissant, E.; Jaafari, N.; Fougerou, C. Melatonin: Pharmacology, Functions and Therapeutic Benefits. Curr. Neuropharmacol. 2017, 15, 434–443. [Google Scholar] [CrossRef]
  6. Cadenas, E.; Davies, K.J. Mitochondrial free radical generation, oxidative stress, and aging. Free. Radic. Biol. Med. 2000, 29, 222–230. [Google Scholar] [CrossRef]
  7. Maffei, M.E. 5-Hydroxytryptophan (5-HTP): Natural Occurrence, Analysis, Biosynthesis, Biotechnology, Physiology and Toxicology. Int. J. Mol. Sci. 2020, 22, 181. [Google Scholar] [CrossRef]
  8. Sangsopha, J.; Johns, N.P.; Johns, J.; Moongngarm, A. Dietary sources of melatonin and benefits from production of high melatonin pasteurized milk. J. Food Sci. Technol. 2020, 57, 2026–2037. [Google Scholar] [CrossRef]
  9. Zuraikat, F.M.; Wood, R.A.; Barragán, R.; St-Onge, M.P. Sleep and Diet: Mounting Evidence of a Cyclical Relationship. Annu. Rev. Nutr. 2021, 41, 309–332. [Google Scholar] [CrossRef]
  10. Mukherjee, S.; Maitra, S.K. Gut Melatonin in Vertebrates: Chronobiology and Physiology. Front. Endocrinol. 2015, 6, 112. [Google Scholar] [CrossRef]
  11. Yasmin, F.; Sutradhar, S.; Das, P.; Mukherjee, S. Gut melatonin: A potent candidate in the diversified journey of melatonin research. Gen. Comp. Endocrinol. 2021, 303, 113693. [Google Scholar] [CrossRef] [PubMed]
  12. Acuña-Castroviejo, D.; Escames, G.; Venegas, C.; Díaz-Casado, M.E.; Lima-Cabello, E.; López, L.C.; Rosales-Corral, S.; Tan, D.X.; Reiter, R.J. Extrapineal melatonin: Sources, regulation, and potential functions. Cell. Mol. Life Sci. 2014, 71, 2997–3025. [Google Scholar] [CrossRef] [PubMed]
  13. Markus, R.P.; Sousa, K.S.; da Silveira Cruz-Machado, S.; Fernandes, P.A.; Ferreira, Z.S. Possible Role of Pineal and Extra-Pineal Melatonin in Surveillance, Immunity, and First-Line Defense. Int. J. Mol. Sci. 2021, 22, 12143. [Google Scholar] [CrossRef] [PubMed]
  14. Ma, N.; Zhang, J.; Reiter, R.J.; Ma, X. Melatonin mediates mucosal immune cells, microbial metabolism, and rhythm crosstalk: A therapeutic target to reduce intestinal inflammation. Med. Res. Rev. 2020, 40, 606–632. [Google Scholar] [CrossRef] [PubMed]
  15. Terziev, D.; Terzieva, D. Experimental Data on the Role of Melatonin in the Pathogenesis of Nonalcoholic Fatty Liver Disease. Biomedicines 2023, 11, 1722. [Google Scholar] [CrossRef] [PubMed]
  16. Ma, Y.; Jiao, J.; Fan, X.; Sun, H.; Zhang, Y.; Jiang, J.; Liu, C. Endophytic Bacterium Pseudomonas fluorescens RG11 May Transform Tryptophan to Melatonin and Promote Endogenous Melatonin Levels in the Roots of Four Grape Cultivars. Front. Plant Sci. 2016, 7, 2068. [Google Scholar] [CrossRef]
  17. Jiao, J.; Ma, Y.; Chen, S.; Liu, C.; Song, Y.; Qin, Y.; Yuan, C.; Liu, Y. Melatonin-Producing Endophytic Bacteria from Grapevine Roots Promote the Abiotic Stress-Induced Production of Endogenous Melatonin in Their Hosts. Front. Plant Sci. 2016, 7, 1387. [Google Scholar] [CrossRef]
  18. Danilovich, M.E.; Alberto, M.R.; Juárez Tomás, M.S. Microbial production of beneficial indoleamines (serotonin and melatonin) with potential application to biotechnological products for human health. J. Appl. Microbiol. 2021, 131, 1668–1682. [Google Scholar] [CrossRef]
  19. Arnao, M.B.; Giraldo-Acosta, M.; Castejón-Castillejo, A.; Losada-Lorán, M.; Sánchez-Herrerías, P.; El Mihyaoui, A.; Cano, A.; Hernández-Ruiz, J. Melatonin from Microorganisms, Algae, and Plants as Possible Alternatives to Synthetic Melatonin. Metabolites 2023, 13, 72. [Google Scholar] [CrossRef]
  20. Bubenik, G.A.; Pang, S.F. The role of serotonin and melatonin in gastrointestinal physiology: Ontogeny, regulation of food intake, and mutual serotonin-melatonin feedback. J. Pineal Res. 1994, 16, 91–99. [Google Scholar] [CrossRef]
  21. Slominski, R.M.; Reiter, R.J.; Schlabritz-Loutsevitch, N.; Ostrom, R.S.; Slominski, A.T. Melatonin membrane receptors in peripheral tissues: Distribution and functions. Mol. Cell. Endocrinol. 2012, 351, 152–166. [Google Scholar] [CrossRef] [PubMed]
  22. Kurhaluk, N. Alcohol and melatonin. Chronobiol. Int. 2021, 38, 785–800. [Google Scholar] [CrossRef] [PubMed]
  23. Osna, N.A.; Donohue, T.M., Jr.; Kharbanda, K.K. Alcoholic Liver Disease: Pathogenesis and Current Management. Alcohol. Res. 2017, 38, 147–161. [Google Scholar]
  24. Rungratanawanich, W.; Qu, Y.; Wang, X.; Essa, M.M.; Song, B.J. Advanced glycation end products (AGEs) and other adducts in aging-related diseases and alcohol-mediated tissue injury. Exp. Mol. Med. 2021, 53, 168–188. [Google Scholar] [CrossRef] [PubMed]
  25. Ceni, E.; Mello, T.; Galli, A. Pathogenesis of alcoholic liver disease: Role of oxidative metabolism. World J. Gastroenterol. 2014, 20, 17756–17772. [Google Scholar] [CrossRef] [PubMed]
  26. Vatsalya, V.; Kong, M.; Cave, M.C.; Liu, N.; Schwandt, M.L.; George, D.T.; Ramchandani, V.A.; McClain, C.J. Association of serum zinc with markers of liver injury in very heavy drinking alcohol-dependent patients. J. Nutr. Biochem. 2018, 59, 49–55. [Google Scholar] [CrossRef] [PubMed]
  27. Ali, H.R.; Assiri, M.A.; Harris, P.S.; Michel, C.R.; Yun, Y.; Marentette, J.O.; Huynh, F.K.; Orlicky, D.J.; Shearn, C.T.; Saba, L.M.; et al. Quantifying Competition among Mitochondrial Protein Acylation Events Induced by Ethanol Metabolism. J. Proteome Res. 2019, 18, 1513–1531. [Google Scholar] [CrossRef]
  28. Cho, Y.E.; Yu, L.R.; Abdelmegeed, M.A.; Yoo, S.H.; Song, B.J. Apoptosis of enterocytes and nitration of junctional complex proteins promote alcohol-induced gut leakiness and liver injury. J. Hepatol. 2018, 69, 142–153. [Google Scholar] [CrossRef]
  29. Addolorato, G.; Ponziani, F.R.; Dionisi, T.; Mosoni, C.; Vassallo, G.A.; Sestito, L.; Petito, V.; Picca, A.; Marzetti, E.; Tarli, C.; et al. Gut microbiota compositional and functional fingerprint in patients with alcohol use disorder and alcohol-associated liver disease. Liver Int. 2020, 40, 878–888. [Google Scholar] [CrossRef]
  30. Harjumäki, 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]
  31. Chen, L.; Zhu, Y.; Hou, X.; Yang, L.; Chu, H. The Role of Gut Bacteria and Fungi in Alcohol-Associated Liver Disease. Front. Med. 2022, 9, 840752. [Google Scholar] [CrossRef] [PubMed]
  32. Namachivayam, A.; Valsala Gopalakrishnan, A. A review on molecular mechanism of alcoholic liver disease. Life Sci. 2021, 274, 119328. [Google Scholar] [CrossRef]
  33. Butura, A.; Nilsson, K.; Morgan, K.; Morgan, T.R.; French, S.W.; Johansson, I.; Schuppe-Koistinen, I.; Ingelman-Sundberg, M. The impact of CYP2E1 on the development of alcoholic liver disease as studied in a transgenic mouse model. J. Hepatol. 2009, 50, 572–583. [Google Scholar] [CrossRef] [PubMed]
  34. Zhao, X.; Wang, C.; Dai, S.; Liu, Y.; Zhang, F.; Peng, C.; Li, Y. Quercetin Protects Ethanol-Induced Hepatocyte Pyroptosis via Scavenging Mitochondrial ROS and Promoting PGC-1α-Regulated Mitochondrial Homeostasis in L02 Cells. Oxid. Med. Cell. Longev. 2022, 2022, 4591134. [Google Scholar] [CrossRef] [PubMed]
  35. Chen, J.; Jiang, S.; Wang, J.; Renukuntla, J.; Sirimulla, S.; Chen, J. A comprehensive review of cytochrome P450 2E1 for xenobiotic metabolism. Drug Metab. Rev. 2019, 51, 178–195. [Google Scholar] [CrossRef] [PubMed]
  36. Seitz, H.K. The role of cytochrome P4502E1 in the pathogenesis of alcoholic liver disease and carcinogenesis. Chem. Biol. Interact. 2020, 316, 108918. [Google Scholar] [CrossRef] [PubMed]
  37. Keshavarzian, A.; Farhadi, A.; Forsyth, C.B.; Rangan, J.; Jakate, S.; Shaikh, M.; Banan, A.; Fields, J.Z. Evidence that chronic alcohol exposure promotes intestinal oxidative stress, intestinal hyperpermeability and endotoxemia prior to development of alcoholic steatohepatitis in rats. J. Hepatol. 2009, 50, 538–547. [Google Scholar] [CrossRef] [PubMed]
  38. Dubinkina, V.B.; Tyakht, A.V.; Odintsova, V.Y.; Yarygin, K.S.; Kovarsky, B.A.; Pavlenko, A.V.; Ischenko, D.S.; Popenko, A.S.; Alexeev, D.G.; Taraskina, A.Y.; et al. Links of gut microbiota composition with alcohol dependence syndrome and alcoholic liver disease. Microbiome 2017, 5, 141. [Google Scholar] [CrossRef]
  39. Baraona, E.; Julkunen, R.; Tannenbaum, L.; Lieber, C.S. Role of intestinal bacterial overgrowth in ethanol production and metabolism in rats. Gastroenterology 1986, 90, 103–110. [Google Scholar] [CrossRef]
  40. Malaguarnera, G.; Giordano, M.; Nunnari, G.; Bertino, G.; Malaguarnera, M. Gut microbiota in alcoholic liver disease: Pathogenetic role and therapeutic perspectives. World J. Gastroenterol. 2014, 20, 16639–16648. [Google Scholar] [CrossRef]
  41. Anderson, G. Linking the biological underpinnings of depression: Role of mitochondria interactions with melatonin, inflammation, sirtuins, tryptophan catabolites, DNA repair and oxidative and nitrosative stress, with consequences for classification and cognition. Prog. Neuropsychopharmacol. Biol. Psychiatry 2018, 80, 255–266. [Google Scholar] [CrossRef] [PubMed]
  42. Anderson, G.; Maes, M. Gut Dysbiosis Dysregulates Central and Systemic Homeostasis via Suboptimal Mitochondrial Function: Assessment, Treatment and Classification Implications. Curr. Top Med. Chem. 2020, 20, 524–539. [Google Scholar] [CrossRef] [PubMed]
  43. Chu, H.; Duan, Y.; Lang, S.; Jiang, L.; Wang, Y.; Llorente, C.; Liu, J.; Mogavero, S.; Bosques-Padilla, F.; Abraldes, J.G.; et al. The Candida albicans exotoxin candidalysin promotes alcohol-associated liver disease. J. Hepatol. 2020, 72, 391–400. [Google Scholar] [CrossRef] [PubMed]
  44. Jew, M.H.; Hsu, C.L. Alcohol, the gut microbiome, and liver disease. J. Gastroenterol. Hepatol. 2023, 38, 1205–1210. [Google Scholar] [CrossRef]
  45. Maccioni, L.; Gao, B.; Leclercq, S.; Pirlot, B.; Horsmans, Y.; De Timary, P.; Leclercq, I.; Fouts, D.; Schnabl, B.; Stärkel, P. Intestinal permeability, microbial translocation, changes in duodenal and fecal microbiota, and their associations with alcoholic liver disease progression in humans. Gut Microbes 2020, 12, 1782157. [Google Scholar] [CrossRef]
  46. Betrapally, N.S.; Gillevet, P.M.; Bajaj, J.S. Changes in the Intestinal Microbiome and Alcoholic and Nonalcoholic Liver Diseases: Causes or Effects? Gastroenterology 2016, 150, 1745–1755.e3. [Google Scholar] [CrossRef]
  47. Chaudhry, K.K.; Samak, G.; Shukla, P.K.; Mir, H.; Gangwar, R.; Manda, B.; Isse, T.; Kawamoto, T.; Salaspuro, M.; Kaihovaara, P.; et al. ALDH2 Deficiency Promotes Ethanol-Induced Gut Barrier Dysfunction and Fatty Liver in Mice. Alcohol. Clin. Exp. Res. 2015, 39, 1465–1475. [Google Scholar] [CrossRef]
  48. Rungratanawanich, W.; Lin, Y.; Wang, X.; Kawamoto, T.; Chidambaram, S.B.; Song, B.J. ALDH2 deficiency increases susceptibility to binge alcohol-induced gut leakiness, endotoxemia, and acute liver injury in mice through the gut-liver axis. Redox Biol.. 2023, 59, 102577. [Google Scholar] [CrossRef]
  49. Lu, Y.; Cederbaum, A.I. Cytochrome P450s and Alcoholic Liver Disease. Curr. Pharm. Des. 2018, 24, 1502–1517. [Google Scholar] [CrossRef]
  50. Moon, K.H.; Hood, B.L.; Kim, B.J.; Hardwick, J.P.; Conrads, T.P.; Veenstra, T.D.; Song, B.J. Inactivation of oxidized and S-nitrosylated mitochondrial proteins in alcoholic fatty liver of rats. Hepatology 2006, 44, 1218–1230. [Google Scholar] [CrossRef]
  51. Moon, K.H.; Hood, B.L.; Mukhopadhyay, P.; Rajesh, M.; Abdelmegeed, M.A.; Kwon, Y.I.; Conrads, T.P.; Veenstra, T.D.; Song, B.J.; Pacher, P. Oxidative inactivation of key mitochondrial proteins leads to dysfunction and injury in hepatic ischemia reperfusion. Gastroenterology 2008, 135, 1344–1357. [Google Scholar] [CrossRef] [PubMed]
  52. Cederbaum, A.I. Role of CYP2E1 in ethanol-induced oxidant stress, fatty liver and hepatotoxicity. Dig. Dis. 2010, 28, 802–811. [Google Scholar] [CrossRef] [PubMed]
  53. Cederbaum, A.I. Alcohol metabolism. Clin. Liver Dis. 2012, 16, 667–685. [Google Scholar] [CrossRef] [PubMed]
  54. Song, B.J. Ethanol-inducible cytochrome P450 (CYP2E1): Biochemistry, molecular biology and clinical relevance: 1996 update. Alcohol. Clin. Exp. Res. 1996, 20, 138a–146a. [Google Scholar] [CrossRef] [PubMed]
  55. Morgan, K.; French, S.W.; Morgan, T.R. Production of a cytochrome P450 2E1 transgenic mouse and initial evaluation of alcoholic liver damage. Hepatology 2002, 36, 122–134. [Google Scholar] [CrossRef] [PubMed]
  56. Lu, Y.; Wu, D.; Wang, X.; Ward, S.C.; Cederbaum, A.I. Chronic alcohol-induced liver injury and oxidant stress are decreased in cytochrome P4502E1 knockout mice and restored in humanized cytochrome P4502E1 knock-in mice. Free. Radic. Biol. Med. 2010, 49, 1406–1416. [Google Scholar] [CrossRef] [PubMed]
  57. Cederbaum, A.I.; Yang, L.; Wang, X.; Wu, D. CYP2E1 Sensitizes the Liver to LPS- and TNF α-Induced Toxicity via Elevated Oxidative and Nitrosative Stress and Activation of ASK-1 and JNK Mitogen-Activated Kinases. Int. J. Hepatol. 2012, 2012, 582790. [Google Scholar] [CrossRef] [PubMed]
  58. Forsyth, C.B.; Voigt, R.M.; Keshavarzian, A. Intestinal CYP2E1: A mediator of alcohol-induced gut leakiness. Redox Biol. 2014, 3, 40–46. [Google Scholar] [CrossRef]
  59. Roberts, B.J.; Song, B.J.; Soh, Y.; Park, S.S.; Shoaf, S.E. Ethanol induces CYP2E1 by protein stabilization. Role of ubiquitin conjugation in the rapid degradation of CYP2E1. J. Biol. Chem. 1995, 270, 29632–29635. [Google Scholar] [CrossRef]
  60. Roberts, B.J.; Shoaf, S.E.; Jeong, K.S.; Song, B.J. Induction of CYP2E1 in liver, kidney, brain and intestine during chronic ethanol administration and withdrawal: Evidence that CYP2E1 possesses a rapid phase half-life of 6 hours or less. Biochem. Biophys. Res. Commun. 1994, 205, 1064–1071. [Google Scholar] [CrossRef]
  61. Hansen, J.; Cherwitz, D.L.; Allen, J.I. The role of tumor necrosis factor-alpha in acute endotoxin-induced hepatotoxicity in ethanol-fed rats. Hepatology 1994, 20, 461–474. [Google Scholar] [CrossRef] [PubMed]
  62. 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] [PubMed]
  63. Yazıcı, D.; Sezer, H. Insulin Resistance, Obesity and Lipotoxicity. Adv. Exp. Med. Biol. 2017, 960, 277–304. [Google Scholar] [CrossRef] [PubMed]
  64. Cho, Y.E.; Kim, D.K.; Seo, W.; Gao, B.; Yoo, S.H.; Song, B.J. Fructose Promotes Leaky Gut, Endotoxemia, and Liver Fibrosis Through Ethanol-Inducible Cytochrome P450-2E1-Mediated Oxidative and Nitrative Stress. Hepatology 2021, 73, 2180–2195. [Google Scholar] [CrossRef] [PubMed]
  65. Drożdż, K.; Nabrdalik, K.; Hajzler, W.; Kwiendacz, H.; Gumprecht, J.; Lip, G.Y.H. Metabolic-Associated Fatty Liver Disease (MAFLD), Diabetes, and Cardiovascular Disease: Associations with Fructose Metabolism and Gut Microbiota. Nutrients 2021, 14, 103. [Google Scholar] [CrossRef] [PubMed]
  66. Karkucinska-Wieckowska, A.; Simoes, I.C.M.; Kalinowski, P.; Lebiedzinska-Arciszewska, M.; Zieniewicz, K.; Milkiewicz, P.; Górska-Ponikowska, M.; Pinton, P.; Malik, A.N.; Krawczyk, M.; et al. Mitochondria, oxidative stress and nonalcoholic fatty liver disease: A complex relationship. Eur. J. Clin. Investig. 2022, 52, e13622. [Google Scholar] [CrossRef]
  67. Abdelmegeed, M.A.; Ha, S.K.; Choi, Y.; Akbar, M.; Song, B.J. Role of CYP2E1 in Mitochondrial Dysfunction and Hepatic Injury by Alcohol and Non-Alcoholic Substances. Curr. Mol. Pharmacol. 2017, 10, 207–225. [Google Scholar] [CrossRef]
  68. Abdelmegeed, M.A.; Banerjee, A.; Yoo, S.H.; Jang, S.; Gonzalez, F.J.; Song, B.J. Critical role of cytochrome P450 2E1 (CYP2E1) in the development of high fat-induced non-alcoholic steatohepatitis. J. Hepatol. 2012, 57, 860–866. [Google Scholar] [CrossRef]
  69. Kathirvel, E.; Chen, P.; Morgan, K.; French, S.W.; Morgan, T.R. Oxidative stress and regulation of anti-oxidant enzymes in cytochrome P4502E1 transgenic mouse model of non-alcoholic fatty liver. J. Gastroenterol. Hepatol. 2010, 25, 1136–1143. [Google Scholar] [CrossRef]
  70. Arroyave-Ospina, J.C.; Wu, Z.; Geng, Y.; Moshage, H. Role of Oxidative Stress in the Pathogenesis of Non-Alcoholic Fatty Liver Disease: Implications for Prevention and Therapy. Antioxidants 2021, 10, 174. [Google Scholar] [CrossRef]
  71. Song, B.J.; Abdelmegeed, M.A.; Henderson, L.E.; Yoo, S.H.; Wan, J.; Purohit, V.; Hardwick, J.P.; Moon, K.H. Increased nitroxidative stress promotes mitochondrial dysfunction in alcoholic and nonalcoholic fatty liver disease. Oxid. Med. Cell. Longev. 2013, 2013, 781050. [Google Scholar] [CrossRef] [PubMed]
  72. Zheng, W.; Song, Z.; Li, S.; Hu, M.; Shaukat, H.; Qin, H. Protective Effects of Sesamol against Liver Oxidative Stress and Inflammation in High-Fat Diet-Induced Hepatic Steatosis. Nutrients 2021, 13, 4484. [Google Scholar] [CrossRef] [PubMed]
  73. Bae, C.S.; Yun, C.H.; Ahn, T. Extracts from Erythronium japonicum and Corylopsis coreana Uyeki reduce 1,3-dichloro-2-propanol-mediated oxidative stress in human hepatic cells. Food Sci. Biotechnol. 2019, 28, 175–180. [Google Scholar] [CrossRef] [PubMed]
  74. Roque Bravo, R.; Carmo, H.; Valente, M.J.; Silva, J.P.; Carvalho, F.; Bastos, M.L.; Dias da Silva, D. 4-Fluoromethamphetamine (4-FMA) induces in vitro hepatotoxicity mediated by CYP2E1, CYP2D6, and CYP3A4 metabolism. Toxicology 2021, 463, 152988. [Google Scholar] [CrossRef] [PubMed]
  75. Sharma, V.; Kaur, R.; Sharma, V.L. Ameliorative potential of Adhatoda vasica against anti-tubercular drugs induced hepatic impairments in female Wistar rats in relation to oxidative stress and xeno-metabolism. J. Ethnopharmacol. 2021, 270, 113771. [Google Scholar] [CrossRef] [PubMed]
  76. Xiao, Q.; Zhao, Y.; Ma, L.; Piao, R. Orientin reverses acetaminophen-induced acute liver failure by inhibiting oxidative stress and mitochondrial dysfunction. J. Pharmacol. Sci. 2022, 149, 11–19. [Google Scholar] [CrossRef] [PubMed]
  77. Luo, D.D.; Chen, J.F.; Liu, J.J.; Xie, J.H.; Zhang, Z.B.; Gu, J.Y.; Zhuo, J.Y.; Huang, S.; Su, Z.R.; Sun, Z.H. Tetrahydrocurcumin and octahydrocurcumin, the primary and final hydrogenated metabolites of curcumin, possess superior hepatic-protective effect against acetaminophen-induced liver injury: Role of CYP2E1 and Keap1-Nrf2 pathway. Food Chem. Toxicol. 2019, 123, 349–362. [Google Scholar] [CrossRef] [PubMed]
  78. Hong, F.; Si, C.; Gao, P.; Cederbaum, A.I.; Xiong, H.; Lu, Y. The role of CYP2A5 in liver injury and fibrosis: Chemical-specific difference. Naunyn Schmiedebergs Arch. Pharmacol. 2016, 389, 33–43. [Google Scholar] [CrossRef]
  79. Jang, S.; Yu, L.R.; Abdelmegeed, M.A.; Gao, Y.; Banerjee, A.; Song, B.J. Critical role of c-jun N-terminal protein kinase in promoting mitochondrial dysfunction and acute liver injury. Redox Biol. 2015, 6, 552–564. [Google Scholar] [CrossRef]
  80. Stading, R.; Couroucli, X.; Lingappan, K.; Moorthy, B. The role of cytochrome P450 (CYP) enzymes in hyperoxic lung injury. Expert Opin. Drug Metab. Toxicol. 2021, 17, 171–178. [Google Scholar] [CrossRef]
  81. Zong, H.; Armoni, M.; Harel, C.; Karnieli, E.; Pessin, J.E. Cytochrome P-450 CYP2E1 knockout mice are protected against high-fat diet-induced obesity and insulin resistance. Am. J. Physiol. Endocrinol. Metab. 2012, 302, E532–E539. [Google Scholar] [CrossRef] [PubMed]
  82. Aubert, J.; Begriche, K.; Knockaert, L.; Robin, M.A.; Fromenty, B. Increased expression of cytochrome P450 2E1 in nonalcoholic fatty liver disease: Mechanisms and pathophysiological role. Clin. Res. Hepatol. Gastroenterol. 2011, 35, 630–637. [Google Scholar] [CrossRef] [PubMed]
  83. Abdelmegeed, M.A.; Choi, Y.; Godlewski, G.; Ha, S.K.; Banerjee, A.; Jang, S.; Song, B.J. Cytochrome P450-2E1 promotes fast food-mediated hepatic fibrosis. Sci. Rep. 2017, 7, 39764. [Google Scholar] [CrossRef] [PubMed]
  84. Day, C.P.; James, O.F. Steatohepatitis: A tale of two “hits”? Gastroenterology 1998, 114, 842–845. [Google Scholar] [CrossRef] [PubMed]
  85. Buzzetti, E.; Pinzani, M.; Tsochatzis, E.A. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism 2016, 65, 1038–1048. [Google Scholar] [CrossRef] [PubMed]
  86. Clare, K.; Dillon, J.F.; Brennan, P.N. Reactive Oxygen Species and Oxidative Stress in the Pathogenesis of MAFLD. J. Clin. Transl. Hepatol. 2022, 10, 939–946. [Google Scholar] [CrossRef]
  87. Dallio, M.; Sangineto, M.; Romeo, M.; Villani, R.; Romano, A.D.; Loguercio, C.; Serviddio, G.; Federico, A. Immunity as Cornerstone of Non-Alcoholic Fatty Liver Disease: The Contribution of Oxidative Stress in the Disease Progression. Int. J. Mol. Sci. 2021, 22, 436. [Google Scholar] [CrossRef]
  88. Chen, Z.; Tian, R.; She, Z.; Cai, J.; Li, H. Role of oxidative stress in the pathogenesis of nonalcoholic fatty liver disease. Free. Radic. Biol. Med. 2020, 152, 116–141. [Google Scholar] [CrossRef]
  89. Malhi, H.; Bronk, S.F.; Werneburg, N.W.; Gores, G.J. Free fatty acids induce JNK-dependent hepatocyte lipoapoptosis. J. Biol. Chem. 2006, 281, 12093–12101. [Google Scholar] [CrossRef]
  90. Shum, M.; Ngo, J.; Shirihai, O.S.; Liesa, M. Mitochondrial oxidative function in NAFLD: Friend or foe? Mol. Metab. 2021, 50, 101134. [Google Scholar] [CrossRef]
  91. Ginès, P.; Krag, A.; Abraldes, J.G.; Solà, E.; Fabrellas, N.; Kamath, P.S. Liver cirrhosis. Lancet 2021, 398, 1359–1376. [Google Scholar] [CrossRef] [PubMed]
  92. Abdelmegeed, M.A.; Banerjee, A.; Jang, S.; Yoo, S.H.; Yun, J.W.; Gonzalez, F.J.; Keshavarzian, A.; Song, B.J. CYP2E1 potentiates binge alcohol-induced gut leakiness, steatohepatitis, and apoptosis. Free. Radic. Biol. Med. 2013, 65, 1238–1245. [Google Scholar] [CrossRef] [PubMed]
  93. Ma, H.D.; Wang, Y.H.; Chang, C.; Gershwin, M.E.; Lian, Z.X. The intestinal microbiota and microenvironment in liver. Autoimmun. Rev. 2015, 14, 183–191. [Google Scholar] [CrossRef] [PubMed]
  94. Fukui, H. Role of Gut Dysbiosis in Liver Diseases: What Have We Learned So Far? Diseases 2019, 7, 58. [Google Scholar] [CrossRef] [PubMed]
  95. Cope, K.; Risby, T.; Diehl, A.M. Increased gastrointestinal ethanol production in obese mice: Implications for fatty liver disease pathogenesis. Gastroenterology 2000, 119, 1340–1347. [Google Scholar] [CrossRef] [PubMed]
  96. Zhu, L.; Baker, S.S.; Gill, C.; Liu, W.; Alkhouri, R.; Baker, R.D.; Gill, S.R. Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: A connection between endogenous alcohol and NASH. Hepatology 2013, 57, 601–609. [Google Scholar] [CrossRef] [PubMed]
  97. Engstler, A.J.; Aumiller, T.; Degen, C.; Dürr, M.; Weiss, E.; Maier, I.B.; Schattenberg, J.M.; Jin, C.J.; Sellmann, C.; Bergheim, I. Insulin resistance alters hepatic ethanol metabolism: Studies in mice and children with non-alcoholic fatty liver disease. Gut 2016, 65, 1564–1571. [Google Scholar] [CrossRef] [PubMed]
  98. Vallianou, N.; Christodoulatos, G.S.; Karampela, I.; Tsilingiris, D.; Magkos, F.; Stratigou, T.; Kounatidis, D.; Dalamaga, M. Understanding the Role of the Gut Microbiome and Microbial Metabolites in Non-Alcoholic Fatty Liver Disease: Current Evidence and Perspectives. Biomolecules 2021, 12, 56. [Google Scholar] [CrossRef]
  99. Ferro, D.; Baratta, F.; Pastori, D.; Cocomello, N.; Colantoni, A.; Angelico, F.; Del Ben, M. New Insights into the Pathogenesis of Non-Alcoholic Fatty Liver Disease: Gut-Derived Lipopolysaccharides and Oxidative Stress. Nutrients 2020, 12, 2762. [Google Scholar] [CrossRef]
  100. Schoeler, M.; Caesar, R. Dietary lipids, gut microbiota and lipid metabolism. Rev. Endocr. Metab. Disord. 2019, 20, 461–472. [Google Scholar] [CrossRef]
  101. Pierantonelli, I.; Svegliati-Baroni, G. Nonalcoholic Fatty Liver Disease: Basic Pathogenetic Mechanisms in the Progression From NAFLD to NASH. Transplantation 2019, 103, e1–e13. [Google Scholar] [CrossRef] [PubMed]
  102. Hardeland, R. Taxon- and Site-Specific Melatonin Catabolism. Molecules 2017, 22, 2015. [Google Scholar] [CrossRef] [PubMed]
  103. Popović, B.; Velimirović, M.; Stojković, T.; Brajović, G.; De Luka, S.R.; Milovanović, I.; Stefanović, S.; Nikolić, D.; Ristić-Djurović, J.L.; Petronijević, N.D.; et al. The influence of ageing on the extrapineal melatonin synthetic pathway. Exp. Gerontol. 2018, 110, 151–157. [Google Scholar] [CrossRef] [PubMed]
  104. Esteban-Zubero, E.; López-Pingarrón, L.; Alatorre-Jiménez, M.A.; Ochoa-Moneo, P.; Buisac-Ramón, C.; Rivas-Jiménez, M.; Castán-Ruiz, S.; Antoñanzas-Lombarte, Á.; Tan, D.X.; García, J.J.; et al. Melatonin’s role as a co-adjuvant treatment in colonic diseases: A review. Life Sci. 2017, 170, 72–81. [Google Scholar] [CrossRef] [PubMed]
  105. Rosales-Corral, S.A.; Reiter, R.J.; Tan, D.-X.; Manchester, L.C.; Liu, X. Chapter 18: Antioxidant and Anti-Inflammatory Role of Melatonin in Alzheimer’s Neurodegeneration; Academic Press: Cambridge, MA, USA, 2014. [Google Scholar]
  106. Hardeland, R. Melatonin metabolism in the central nervous system. Curr. Neuropharmacol. 2010, 8, 168–181. [Google Scholar] [CrossRef] [PubMed]
  107. Acuña-Castroviejo, D.; Escames, G.; León, J.; Carazo, A.; Khaldy, H. Mitochondrial regulation by melatonin and its metabolites. Adv. Exp. Med. Biol. 2003, 527, 549–557. [Google Scholar] [CrossRef] [PubMed]
  108. Mokhtari, B.; Yavari, R.; Badalzadeh, R.; Mahmoodpoor, A. An Overview on Mitochondrial-Based Therapies in Sepsis-Related Myocardial Dysfunction: Mitochondrial Transplantation as a Promising Approach. Can. J. Infect. Dis. Med. Microbiol. 2022, 2022, 3277274. [Google Scholar] [CrossRef] [PubMed]
  109. Swanson, G.R.; Siskin, J.; Gorenz, A.; Shaikh, M.; Raeisi, S.; Fogg, L.; Forsyth, C.; Keshavarzian, A. Disrupted diurnal oscillation of gut-derived Short chain fatty acids in shift workers drinking alcohol: Possible mechanism for loss of resiliency of intestinal barrier in disrupted circadian host. Transl. Res. 2020, 221, 97–109. [Google Scholar] [CrossRef]
  110. Engen, P.A.; Green, S.J.; Voigt, R.M.; Forsyth, C.B.; Keshavarzian, A. The Gastrointestinal Microbiome: Alcohol Effects on the Composition of Intestinal Microbiota. Alcohol. Res. 2015, 37, 223–236. [Google Scholar]
  111. Tran, L.; Jochum, S.B.; Shaikh, M.; Wilber, S.; Zhang, L.; Hayden, D.M.; Forsyth, C.B.; Voigt, R.M.; Bishehsari, F.; Keshavarzian, A.; et al. Circadian misalignment by environmental light/dark shifting causes circadian disruption in colon. PLoS ONE 2021, 16, e0251604. [Google Scholar] [CrossRef]
  112. Voigt, R.M.; Forsyth, C.B.; Keshavarzian, A. Circadian rhythms: A regulator of gastrointestinal health and dysfunction. Expert Rev. Gastroenterol. Hepatol. 2019, 13, 411–424. [Google Scholar] [CrossRef] [PubMed]
  113. Swanson, G.R.; Gorenz, A.; Shaikh, M.; Desai, V.; Forsyth, C.; Fogg, L.; Burgess, H.J.; Keshavarzian, A. Decreased melatonin secretion is associated with increased intestinal permeability and marker of endotoxemia in alcoholics. Am. J. Physiol. Gastrointest Liver Physiol. 2015, 308, G1004–G1011. [Google Scholar] [CrossRef] [PubMed]
  114. Kurhaluk, N.; Tkachenko, H. Melatonin and alcohol-related disorders. Chronobiol. Int. 2020, 37, 781–803. [Google Scholar] [CrossRef] [PubMed]
  115. Reiter, R.J.; Tan, D.X.; Rosales-Corral, S.; Galano, A.; Zhou, X.J.; Xu, B. Mitochondria: Central Organelles for Melatonin’s Antioxidant and Anti-Aging Actions. Molecules 2018, 23, 509. [Google Scholar] [CrossRef] [PubMed]
  116. Paradies, G.; Paradies, V.; Ruggiero, F.M.; Petrosillo, G. Mitochondrial bioenergetics decay in aging: Beneficial effect of melatonin. Cell. Mol. Life Sci. 2017, 74, 3897–3911. [Google Scholar] [CrossRef] [PubMed]
  117. Reiter, R.J.; Rosales-Corral, S.; Tan, D.X.; Jou, M.J.; Galano, A.; Xu, B. Melatonin as a mitochondria-targeted antioxidant: One of evolution’s best ideas. Cell. Mol. Life Sci. 2017, 74, 3863–3881. [Google Scholar] [CrossRef] [PubMed]
  118. Reiter, R.J.; Sharma, R.; Rosales-Corral, S.; de Campos Zuccari, D.A.P.; de Almeida Chuffa, L.G. Melatonin: A mitochondrial resident with a diverse skill set. Life Sci. 2022, 301, 120612. [Google Scholar] [CrossRef] [PubMed]
  119. Li, J.; Li, N.; Yan, S.; Lu, Y.; Miao, X.; Gu, Z.; Shao, Y. Melatonin attenuates renal fibrosis in diabetic mice by activating the AMPK/PGC1α signaling pathway and rescuing mitochondrial function. Mol. Med. Rep. 2019, 19, 1318–1330. [Google Scholar] [CrossRef]
  120. Chen, W.R.; Zhou, Y.J.; Sha, Y.; Wu, X.P.; Yang, J.Q.; Liu, F. Melatonin attenuates vascular calcification by inhibiting mitochondria fission via an AMPK/Drp1 signalling pathway. J. Cell Mol. Med. 2020, 24, 6043–6054. [Google Scholar] [CrossRef]
  121. Robin, M.A.; Sauvage, I.; Grandperret, T.; Descatoire, V.; Pessayre, D.; Fromenty, B. Ethanol increases mitochondrial cytochrome P450 2E1 in mouse liver and rat hepatocytes. FEBS Lett. 2005, 579, 6895–6902. [Google Scholar] [CrossRef]
  122. Neve, E.P.; Ingelman-Sundberg, M. Molecular basis for the transport of cytochrome P450 2E1 to the plasma membrane. J. Biol. Chem. 2000, 275, 17130–17135. [Google Scholar] [CrossRef] [PubMed]
  123. Neve, E.P.; Ingelman-Sundberg, M. A soluble NH(2)-terminally truncated catalytically active form of rat cytochrome P450 2E1 targeted to liver mitochondria(1). FEBS Lett. 1999, 460, 309–314. [Google Scholar] [CrossRef] [PubMed]
  124. Lieber, C.S. Cytochrome P-4502E1: Its physiological and pathological role. Physiol. Rev. 1997, 77, 517–544. [Google Scholar] [CrossRef] [PubMed]
  125. Bai, J.; Cederbaum, A.I. Overexpression of CYP2E1 in mitochondria sensitizes HepG2 cells to the toxicity caused by depletion of glutathione. J. Biol. Chem. 2006, 281, 5128–5136. [Google Scholar] [CrossRef] [PubMed]
  126. Solís-Muñoz, P.; Solís-Herruzo, J.A.; Fernández-Moreira, D.; Gómez-Izquierdo, E.; García-Consuegra, I.; Muñoz-Yagüe, T.; García Ruiz, I. Melatonin improves mitochondrial respiratory chain activity and liver morphology in ob/ob mice. J. Pineal Res. 2011, 51, 113–123. [Google Scholar] [CrossRef] [PubMed]
  127. Chen, C.Q.; Fichna, J.; Bashashati, M.; Li, Y.Y.; Storr, M. Distribution, function and physiological role of melatonin in the lower gut. World J. Gastroenterol. 2011, 17, 3888–3898. [Google Scholar] [CrossRef]
  128. Hardeland, R. Aging, Melatonin, and the Pro- and Anti-Inflammatory Networks. Int. J. Mol. Sci. 2019, 20, 1223. [Google Scholar] [CrossRef]
  129. Zhao, L.; An, R.; Yang, Y.; Yang, X.; Liu, H.; Yue, L.; Li, X.; Lin, Y.; Reiter, R.J.; Qu, Y. Melatonin alleviates brain injury in mice subjected to cecal ligation and puncture via attenuating inflammation, apoptosis, and oxidative stress: The role of SIRT1 signaling. J. Pineal Res. 2015, 59, 230–239. [Google Scholar] [CrossRef]
  130. Guo, Y.; Sun, J.; Bu, S.; Li, B.; Zhang, Q.; Wang, Q.; Lai, D. Melatonin protects against chronic stress-induced oxidative meiotic defects in mice MII oocytes by regulating SIRT1. Cell Cycle 2020, 19, 1677–1695. [Google Scholar] [CrossRef]
  131. Chandramowlishwaran, P.; Vijay, A.; Abraham, D.; Li, G.; Mwangi, S.M.; Srinivasan, S. Role of Sirtuins in Modulating Neurodegeneration of the Enteric Nervous System and Central Nervous System. Front. Neurosci. 2020, 14, 614331. [Google Scholar] [CrossRef]
  132. Zhang, G.Z.; Deng, Y.J.; Xie, Q.Q.; Ren, E.H.; Ma, Z.J.; He, X.G.; Gao, Y.C.; Kang, X.W. Sirtuins and intervertebral disc degeneration: Roles in inflammation, oxidative stress, and mitochondrial function. Clin. Chim. Acta 2020, 508, 33–42. [Google Scholar] [CrossRef]
  133. Jiang, J.; Liang, S.; Zhang, J.; Du, Z.; Xu, Q.; Duan, J.; Sun, Z. Melatonin ameliorates PM(2.5) -induced cardiac perivascular fibrosis through regulating mitochondrial redox homeostasis. J. Pineal Res. 2021, 70, e12686. [Google Scholar] [CrossRef]
  134. Ma, S.; Chen, J.; Feng, J.; Zhang, R.; Fan, M.; Han, D.; Li, X.; Li, C.; Ren, J.; Wang, Y.; et al. Melatonin Ameliorates the Progression of Atherosclerosis via Mitophagy Activation and NLRP3 Inflammasome Inhibition. Oxid Med. Cell. Longev. 2018, 2018, 9286458. [Google Scholar] [CrossRef]
  135. Wu, J.; Yang, Y.; Gao, Y.; Wang, Z.; Ma, J. Melatonin Attenuates Anoxia/Reoxygenation Injury by Inhibiting Excessive Mitophagy Through the MT2/SIRT3/FoxO3a Signaling Pathway in H9c2 Cells. Drug Des. Devel. Ther. 2020, 14, 2047–2060. [Google Scholar] [CrossRef]
  136. Reiter, R.J.; Tan, D.X.; Rosales-Corral, S.; Galano, A.; Jou, M.J.; Acuna-Castroviejo, D. Melatonin Mitigates Mitochondrial Meltdown: Interactions with SIRT3. Int. J. Mol. Sci. 2018, 19, 2439. [Google Scholar] [CrossRef]
  137. Bai, Y.; Yang, Y.; Gao, Y.; Lin, D.; Wang, Z.; Ma, J. Melatonin postconditioning ameliorates anoxia/reoxygenation injury by regulating mitophagy and mitochondrial dynamics in a SIRT3-dependent manner. Eur. J. Pharmacol. 2021, 904, 174157. [Google Scholar] [CrossRef]
  138. Yu, L.M.; Dong, X.; Xue, X.D.; Xu, S.; Zhang, X.; Xu, Y.L.; Wang, Z.S.; Wang, Y.; Gao, H.; Liang, Y.X.; et al. Melatonin attenuates diabetic cardiomyopathy and reduces myocardial vulnerability to ischemia-reperfusion injury by improving mitochondrial quality control: Role of SIRT6. J. Pineal Res. 2021, 70, e12698. [Google Scholar] [CrossRef]
  139. Kim, H.G.; Huang, M.; Xin, Y.; Zhang, Y.; Zhang, X.; Wang, G.; Liu, S.; Wan, J.; Ahmadi, A.R.; Sun, Z.; et al. The epigenetic regulator SIRT6 protects the liver from alcohol-induced tissue injury by reducing oxidative stress in mice. J. Hepatol. 2019, 71, 960–969. [Google Scholar] [CrossRef]
  140. Zeng, C.; Chen, M. Progress in Nonalcoholic Fatty Liver Disease: SIRT Family Regulates Mitochondrial Biogenesis. Biomolecules 2022, 12, 1079. [Google Scholar] [CrossRef]
  141. Wang, S.; Wang, L.; Qin, X.; Turdi, S.; Sun, D.; Culver, B.; Reiter, R.J.; Wang, X.; Zhou, H.; Ren, J. ALDH2 contributes to melatonin-induced protection against APP/PS1 mutation-prompted cardiac anomalies through cGAS-STING-TBK1-mediated regulation of mitophagy. Signal Transduct. Target. Ther. 2020, 5, 119. [Google Scholar] [CrossRef]
  142. Zhou, H.; Du, W.; Li, Y.; Shi, C.; Hu, N.; Ma, S.; Wang, W.; Ren, J. Effects of melatonin on fatty liver disease: The role of NR4A1/DNA-PKcs/p53 pathway, mitochondrial fission, and mitophagy. J. Pineal Res. 2018, 64, e12450. [Google Scholar] [CrossRef]
  143. Stacchiotti, A.; Favero, G.; Rodella, L.F. Impact of Melatonin on Skeletal Muscle and Exercise. Cells 2020, 9, 288. [Google Scholar] [CrossRef]
  144. Chen, C.; Yang, C.; Wang, J.; Huang, X.; Yu, H.; Li, S.; Li, S.; Zhang, Z.; Liu, J.; Yang, X.; et al. Melatonin ameliorates cognitive deficits through improving mitophagy in a mouse model of Alzheimer’s disease. J. Pineal Res. 2021, 71, e12774. [Google Scholar] [CrossRef]
  145. Doğanlar, Z.B.; Doğanlar, O.; Kurtdere, K.; Güçlü, H.; Chasan, T.; Turgut, E. Melatonin prevents blood-retinal barrier breakdown and mitochondrial dysfunction in high glucose and hypoxia-induced in vitro diabetic macular edema model. Toxicol. Vitr. 2021, 75, 105191. [Google Scholar] [CrossRef]
  146. Wang, Z.; Zhou, F.; Dou, Y.; Tian, X.; Liu, C.; Li, H.; Shen, H.; Chen, G. Melatonin Alleviates Intracerebral Hemorrhage-Induced Secondary Brain Injury in Rats via Suppressing Apoptosis, Inflammation, Oxidative Stress, DNA Damage, and Mitochondria Injury. Transl. Stroke Res. 2018, 9, 74–91. [Google Scholar] [CrossRef]
  147. Yapislar, H.; Haciosmanoglu, E.; Sarioglu, T.; Ekmekcioglu, C. The melatonin MT(2) receptor is involved in the anti-apoptotic effects of melatonin in rats with type 2 diabetes mellitus. Tissue Cell 2022, 76, 101763. [Google Scholar] [CrossRef]
  148. Galano, A.; Tan, D.X.; Reiter, R.J. On the free radical scavenging activities of melatonin’s metabolites, AFMK and AMK. J. Pineal Res. 2013, 54, 245–257. [Google Scholar] [CrossRef]
  149. Tan, D.X.; Manchester, L.C.; Terron, M.P.; Flores, L.J.; Reiter, R.J. One molecule, many derivatives: A never-ending interaction of melatonin with reactive oxygen and nitrogen species? J. Pineal Res. 2007, 42, 28–42. [Google Scholar] [CrossRef]
  150. Ballway, J.W.; Song, B.J. Translational Approaches with Antioxidant Phytochemicals against Alcohol-Mediated Oxidative Stress, Gut Dysbiosis, Intestinal Barrier Dysfunction, and Fatty Liver Disease. Antioxidants 2021, 10, 384. [Google Scholar] [CrossRef]
  151. Kim, D.H.; Sim, Y.; Hwang, J.H.; Kwun, I.S.; Lim, J.H.; Kim, J.; Kim, J.I.; Baek, M.C.; Akbar, M.; Seo, W.; et al. Ellagic Acid Prevents Binge Alcohol-Induced Leaky Gut and Liver Injury through Inhibiting Gut Dysbiosis and Oxidative Stress. Antioxidants 2021, 10, 1386. [Google Scholar] [CrossRef]
  152. Whitfield, C.; Trent, M.S. Biosynthesis and export of bacterial lipopolysaccharides. Annu. Rev. Biochem. 2014, 83, 99–128. [Google Scholar] [CrossRef]
  153. Forsyth, C.B.; Voigt, R.M.; Shaikh, M.; Tang, Y.; Cederbaum, A.I.; Turek, F.W.; Keshavarzian, A. Role for intestinal CYP2E1 in alcohol-induced circadian gene-mediated intestinal hyperpermeability. Am. J. Physiol. Gastrointest Liver Physiol. 2013, 305, G185–G195. [Google Scholar] [CrossRef] [PubMed]
  154. Forsyth, C.B.; Voigt, R.M.; Burgess, H.J.; Swanson, G.R.; Keshavarzian, A. Circadian rhythms, alcohol and gut interactions. Alcohol 2015, 49, 389–398. [Google Scholar] [CrossRef] [PubMed]
  155. Swanson, G.R.; Gorenz, A.; Shaikh, M.; Desai, V.; Kaminsky, T.; Van Den Berg, J.; Murphy, T.; Raeisi, S.; Fogg, L.; Vitaterna, M.H.; et al. Night workers with circadian misalignment are susceptible to alcohol-induced intestinal hyperpermeability with social drinking. Am. J. Physiol. Gastrointest Liver Physiol. 2016, 311, G192–G201. [Google Scholar] [CrossRef] [PubMed]
  156. Fonzi, S.; Solinas, G.P.; Costelli, P.; Parodi, C.; Murialdo, G.; Bo, P.; Albergati, A.; Montalbetti, L.; Savoldi, F.; Polleri, A. Melatonin and cortisol circadian secretion during ethanol withdrawal in chronic alcoholics. Chronobiologia 1994, 21, 109–112. [Google Scholar]
  157. Che, Z.; Song, Y.; Xu, C.; Li, W.; Dong, Z.; Wang, C.; Ren, Y.; So, K.F.; Tipoe, G.L.; Wang, F.; et al. Melatonin alleviates alcoholic liver disease via EGFR-BRG1-TERT axis regulation. Acta Pharm. Sin. B 2023, 13, 100–112. [Google Scholar] [CrossRef]
  158. Arioz, B.I.; Tarakcioglu, E.; Olcum, M.; Genc, S. The Role of Melatonin on NLRP3 Inflammasome Activation in Diseases. Antioxidants 2021, 10, 1020. [Google Scholar] [CrossRef]
  159. Ashrafizadeh, M.; Najafi, M.; Kavyiani, N.; Mohammadinejad, R.; Farkhondeh, T.; Samarghandian, S. Anti-Inflammatory Activity of Melatonin: A Focus on the Role of NLRP3 Inflammasome. Inflammation 2021, 44, 1207–1222. [Google Scholar] [CrossRef]
  160. Lee, S.E.; Koh, H.; Joo, D.J.; Nedumaran, B.; Jeon, H.J.; Park, C.S.; Harris, R.A.; Kim, Y.D. Induction of SIRT1 by melatonin improves alcohol-mediated oxidative liver injury by disrupting the CRBN-YY1-CYP2E1 signaling pathway. J. Pineal Res. 2020, 68, e12638. [Google Scholar] [CrossRef]
  161. Mishra, A.; Paul, S.; Swarnakar, S. Downregulation of matrix metalloproteinase-9 by melatonin during prevention of alcohol-induced liver injury in mice. Biochimie 2011, 93, 854–866. [Google Scholar] [CrossRef]
  162. Ullah, U.; Badshah, H.; Malik, Z.; Uddin, Z.; Alam, M.; Sarwar, S.; Aman, A.; Khan, A.U.; Shah, F.A. Hepatoprotective effects of melatonin and celecoxib against ethanol-induced hepatotoxicity in rats. Immunopharmacol. Immunotoxicol. 2020, 42, 255–263. [Google Scholar] [CrossRef] [PubMed]
  163. Song, Y.M.; Lee, Y.H.; Kim, J.W.; Ham, D.S.; Kang, E.S.; Cha, B.S.; Lee, H.C.; Lee, B.W. Metformin alleviates hepatosteatosis by restoring SIRT1-mediated autophagy induction via an AMP-activated protein kinase-independent pathway. Autophagy 2015, 11, 46–59. [Google Scholar] [CrossRef] [PubMed]
  164. Rui, B.B.; Chen, H.; Jang, L.; Li, Z.; Yang, J.M.; Xu, W.P.; Wei, W. Melatonin Upregulates the Activity of AMPK and Attenuates Lipid Accumulation in Alcohol-induced Rats. Alcohol Alcohol. 2016, 51, 11–19. [Google Scholar] [CrossRef] [PubMed]
  165. Zhang, J.J.; Meng, X.; Li, Y.; Zhou, Y.; Xu, D.P.; Li, S.; Li, H.B. Effects of Melatonin on Liver Injuries and Diseases. Int. J. Mol. Sci. 2017, 18, 673. [Google Scholar] [CrossRef] [PubMed]
  166. Kurhaluk, N.; Tkachenko, H.; Lukash, O. Melatonin modulates oxidative phosphorylation, hepatic and kidney autophagy-caused subclinical endotoxemia and acute ethanol-induced oxidative stress. Chronobiol. Int. 2020, 37, 1709–1724. [Google Scholar] [CrossRef] [PubMed]
  167. Kim, Y.D.; Hwang, S.L.; Lee, E.J.; Kim, H.M.; Chung, M.J.; Elfadl, A.K.; Lee, S.E.; Nedumaran, B.; Harris, R.A.; Jeong, K.S. Melatonin ameliorates alcohol-induced bile acid synthesis by enhancing miR-497 expression. J. Pineal Res. 2017, 62, e12386. [Google Scholar] [CrossRef] [PubMed]
  168. Teunis, C.; Nieuwdorp, M.; Hanssen, N. Interactions between Tryptophan Metabolism, the Gut Microbiome and the Immune System as Potential Drivers of Non-Alcoholic Fatty Liver Disease (NAFLD) and Metabolic Diseases. Metabolites 2022, 12, 514. [Google Scholar] [CrossRef]
  169. Genario, R.; Cipolla-Neto, J.; Bueno, A.A.; Santos, H.O. Melatonin supplementation in the management of obesity and obesity-associated disorders: A review of physiological mechanisms and clinical applications. Pharmacol. Res. 2021, 163, 105254. [Google Scholar] [CrossRef]
  170. Halpern, B.; Mancini, M.C.; Bueno, C.; Barcelos, I.P.; de Melo, M.E.; Lima, M.S.; Carneiro, C.G.; Sapienza, M.T.; Buchpiguel, C.A.; do Amaral, F.G.; et al. Melatonin Increases Brown Adipose Tissue Volume and Activity in Patients With Melatonin Deficiency: A Proof-of-Concept Study. Diabetes 2019, 68, 947–952. [Google Scholar] [CrossRef]
  171. Kim, M.; Lee, S.M.; Jung, J.; Kim, Y.J.; Moon, K.C.; Seo, J.H.; Ha, T.K.; Ha, E. Pinealectomy increases thermogenesis and decreases lipogenesis. Mol. Med. Rep. 2020, 22, 4289–4297. [Google Scholar] [CrossRef]
  172. Sohrabi, M.; Gholami, A.; Amirkalali, B.; Taherizadeh, M.; Kolahdoz, M.; SafarnezhadTameshkel, F.; Aghili, S.; Hajibaba, M.; Zamani, F.; Nasiri Toosi, M.; et al. Is melatonin associated with pro-inflammatory cytokine activity and liver fibrosis in non-alcoholic fatty liver disease (NAFLD) patients? Gastroenterol. Hepatol. Bed Bench 2021, 14, 229–236. [Google Scholar] [PubMed]
  173. Sohrabi, M.; Ajdarkosh, H.; Gholami, A.; Amirkalali, B.; Mansorian, M.R.; Aten, S.; Sohrabi, M.; Nasiri-Toosi, M.; Zamani, F.; Keyvani, H. Association between Melatonin Value and Interleukins1B, -18, and -33 Levels in Patients with Different Stages of Non-Alcoholic Fatty Liver Disease. Middle East J. Dig. Dis. 2022, 14, 110–117. [Google Scholar] [CrossRef] [PubMed]
  174. Lane, E.A.; Moss, H.B. Pharmacokinetics of melatonin in man: First pass hepatic metabolism. J. Clin. Endocrinol. Metab. 1985, 61, 1214–1216. [Google Scholar] [CrossRef] [PubMed]
  175. Zhang, H.; Yan, A.; Liu, X.; Ma, Y.; Zhao, F.; Wang, M.; Loor, J.J.; Wang, H. Melatonin ameliorates ochratoxin A induced liver inflammation, oxidative stress and mitophagy in mice involving in intestinal microbiota and restoring the intestinal barrier function. J. Hazard. Mater. 2021, 407, 124489. [Google Scholar] [CrossRef] [PubMed]
  176. Yildirim, A.; Arabacı Tamer, S.; Sahin, D.; Bagriacik, F.; Kahraman, M.M.; Onur, N.D.; Cayirli, Y.B.; Cilingir Kaya Ö, T.; Aksu, B.; Akdeniz, E.; et al. The effects of antibiotics and melatonin on hepato-intestinal inflammation and gut microbial dysbiosis induced by a short-term high-fat diet consumption in rats. Br. J. Nutr. 2019, 122, 841–855. [Google Scholar] [CrossRef] [PubMed]
  177. Liu, S.; Kang, W.; Mao, X.; Ge, L.; Du, H.; Li, J.; Hou, L.; Liu, D.; Yin, Y.; Liu, Y.; et al. Melatonin mitigates aflatoxin B1-induced liver injury via modulation of gut microbiota/intestinal FXR/liver TLR4 signaling axis in mice. J. Pineal Res. 2022, 73, e12812. [Google Scholar] [CrossRef] [PubMed]
  178. Valenzuela-Melgarejo, F.J.; Caro-Díaz, C.; Cabello-Guzmán, G. Potential Crosstalk between Fructose and Melatonin: A New Role of Melatonin-Inhibiting the Metabolic Effects of Fructose. Int. J. Endocrinol. 2018, 2018, 7515767. [Google Scholar] [CrossRef]
  179. Pafili, K.; Roden, M. Nonalcoholic fatty liver disease (NAFLD) from pathogenesis to treatment concepts in humans. Mol. Metab. 2021, 50, 101122. [Google Scholar] [CrossRef]
  180. Caballero, M.E.; Berlanga, J.; Ramirez, D.; Lopez-Saura, P.; Gozalez, R.; Floyd, D.N.; Marchbank, T.; Playford, R.J. Epidermal growth factor reduces multiorgan failure induced by thioacetamide. Gut 2001, 48, 34–40. [Google Scholar] [CrossRef]
  181. Shin, S.K.; Cho, H.W.; Song, S.E.; Bae, J.H.; Im, S.S.; Hwang, I.; Ha, H.; Song, D.K. Ablation of catalase promotes non-alcoholic fatty liver via oxidative stress and mitochondrial dysfunction in diet-induced obese mice. Pflugers Arch. 2019, 471, 829–843. [Google Scholar] [CrossRef]
  182. Rishi, P.; Bharrhan, S.; Bhalla, M.P.; Koul, A.; Chopra, K. Inhibition of endotoxin-induced hepatotoxicity by melatonin in rats. Int. J. Biomed. Sci. 2008, 4, 103–112. [Google Scholar] [CrossRef] [PubMed]
  183. Chen, X.; Zhang, C.; Zhao, M.; Shi, C.E.; Zhu, R.M.; Wang, H.; Zhao, H.; Wei, W.; Li, J.B.; Xu, D.X. Melatonin alleviates lipopolysaccharide-induced hepatic SREBP-1c activation and lipid accumulation in mice. J. Pineal Res. 2011, 51, 416–425. [Google Scholar] [CrossRef] [PubMed]
  184. Xia, D.; Yang, L.; Li, Y.; Chen, J.; Zhang, X.; Wang, H.; Zhai, S.; Jiang, X.; Meca, G.; Wang, S.; et al. Melatonin alleviates Ochratoxin A-induced liver inflammation involved intestinal microbiota homeostasis and microbiota-independent manner. J. Hazard. Mater. 2021, 413, 125239. [Google Scholar] [CrossRef] [PubMed]
  185. Xu, S.; Li, L.; Wu, J.; An, S.; Fang, H.; Han, Y.; Huang, Q.; Chen, Z.; Zeng, Z. Melatonin Attenuates Sepsis-Induced Small-Intestine Injury by Upregulating SIRT3-Mediated Oxidative-Stress Inhibition, Mitochondrial Protection, and Autophagy Induction. Front. Immunol. 2021, 12, 625627. [Google Scholar] [CrossRef] [PubMed]
  186. Kleber, A.; Kubulus, D.; Rössler, D.; Wolf, B.; Volk, T.; Speer, T.; Fink, T. Melatonin modifies cellular stress in the liver of septic mice by reducing reactive oxygen species and increasing the unfolded protein response. Exp. Mol. Pathol. 2014, 97, 565–571. [Google Scholar] [CrossRef]
  187. Borrelli, A.; Bonelli, P.; Tuccillo, F.M.; Goldfine, I.D.; Evans, J.L.; Buonaguro, F.M.; Mancini, A. Role of gut microbiota and oxidative stress in the progression of non-alcoholic fatty liver disease to hepatocarcinoma: Current and innovative therapeutic approaches. Redox Biol. 2018, 15, 467–479. [Google Scholar] [CrossRef] [PubMed]
  188. Farzanegi, P.; Dana, A.; Ebrahimpoor, Z.; Asadi, M.; Azarbayjani, M.A. Mechanisms of beneficial effects of exercise training on non-alcoholic fatty liver disease (NAFLD): Roles of oxidative stress and inflammation. Eur. J. Sport Sci. 2019, 19, 994–1003. [Google Scholar] [CrossRef] [PubMed]
  189. Buxton, O.M.; L’Hermite-Balériaux, M.; Hirschfeld, U.; Cauter, E. Acute and delayed effects of exercise on human melatonin secretion. J. Biol. Rhythms. 1997, 12, 568–574. [Google Scholar] [CrossRef]
  190. Oyewole, A.O.; Birch-Machin, M.A. Mitochondria-targeted antioxidants. FASEB J. 2015, 29, 4766–4771. [Google Scholar] [CrossRef]
  191. Croci, S.; D’Apolito, L.I.; Gasperi, V.; Catani, M.V.; Savini, I. Dietary Strategies for Management of Metabolic Syndrome: Role of Gut Microbiota Metabolites. Nutrients 2021, 13, 1389. [Google Scholar] [CrossRef]
  192. Wrzosek, L.; Ciocan, D.; Hugot, C.; Spatz, M.; Dupeux, M.; Houron, C.; Lievin-Le Moal, V.; Puchois, V.; Ferrere, G.; Trainel, N.; et al. Microbiota tryptophan metabolism induces aryl hydrocarbon receptor activation and improves alcohol-induced liver injury. Gut 2021, 70, 1299–1308. [Google Scholar] [CrossRef] [PubMed]
  193. Fuke, N.; Nagata, N.; Suganuma, H.; Ota, T. Regulation of Gut Microbiota and Metabolic Endotoxemia with Dietary Factors. Nutrients 2019, 11, 2277. [Google Scholar] [CrossRef] [PubMed]
  194. Bonnefont-Rousselot, D.; Collin, F. Melatonin: Action as antioxidant and potential applications in human disease and aging. Toxicology 2010, 278, 55–67. [Google Scholar] [CrossRef] [PubMed]
  195. Tomasello, G.; Mazzola, M.; Leone, A.; Sinagra, E.; Zummo, G.; Farina, F.; Damiani, P.; Cappello, F.; Gerges Geagea, A.; Jurjus, A.; et al. Nutrition, oxidative stress and intestinal dysbiosis: Influence of diet on gut microbiota in inflammatory bowel diseases. Biomed. Pap. Med. Fac. Univ. Palacky Olomouc. Czech Repub. 2016, 160, 461–466. [Google Scholar] [CrossRef] [PubMed]
  196. Suhail Kasim, B.Z.; Wafa Harrouk, K.A. Melatonin: Pharmacy Compounding Advisory Committee Meeting; Food and Drug Administration: Silver Spring, MD, USA, 2021. [Google Scholar]
  197. Kuehn, B.M. Climbing Melatonin Use for Insomnia Raises Safety Concerns. JAMA 2022, 328, 605–607. [Google Scholar] [CrossRef] [PubMed]
  198. Galley, H.F.; Lowes, D.A.; Allen, L.; Cameron, G.; Aucott, L.S.; Webster, N.R. Melatonin as a potential therapy for sepsis: A phase I dose escalation study and an ex vivo whole blood model under conditions of sepsis. J. Pineal Res. 2014, 56, 427–438. [Google Scholar] [CrossRef] [PubMed]
  199. Malhotra, S.; Sawhney, G.; Pandhi, P. The therapeutic potential of melatonin: A review of the science. MedGenMed 2004, 6, 46. [Google Scholar]
  200. Nickkholgh, A.; Schneider, H.; Sobirey, M.; Venetz, W.P.; Hinz, U.; Pelzl le, H.; Gotthardt, D.N.; Cekauskas, A.; Manikas, M.; Mikalauskas, S.; et al. The use of high-dose melatonin in liver resection is safe: First clinical experience. J. Pineal Res. 2011, 50, 381–388. [Google Scholar] [CrossRef]
  201. Seabra, M.L.; Bignotto, M.; Pinto, L.R., Jr.; Tufik, S. Randomized, double-blind clinical trial, controlled with placebo, of the toxicology of chronic melatonin treatment. J. Pineal Res. 2000, 29, 193–200. [Google Scholar] [CrossRef]
  202. Lissoni, P.; Barni, S.; Rovelli, F.; Brivio, F.; Ardizzoia, A.; Tancini, G.; Conti, A.; Maestroni, G.J. Neuroimmunotherapy of advanced solid neoplasms with single evening subcutaneous injection of low-dose interleukin-2 and melatonin: Preliminary results. Eur. J. Cancer 1993, 29, 185–189. [Google Scholar] [CrossRef]
  203. Arendt, J.; Aldhous, M.; Marks, V. Alleviation of jet lag by melatonin: Preliminary results of controlled double blind trial. Br. Med. J. (Clin. Res. Ed.) 1986, 292, 1170. [Google Scholar] [CrossRef] [PubMed]
  204. Lieberman, H.R.; Waldhauser, F.; Garfield, G.; Lynch, H.J.; Wurtman, R.J. Effects of melatonin on human mood and performance. Brain Res. 1984, 323, 201–207. [Google Scholar] [CrossRef] [PubMed]
  205. James, S.P.; Sack, D.A.; Rosenthal, N.E.; Mendelson, W.B. Melatonin administration in insomnia. Neuropsychopharmacology 1990, 3, 19–23. [Google Scholar] [PubMed]
  206. Samel, A.; Wegmann, H.M.; Vejvoda, M.; Maass, H.; Gundel, A.; Schütz, M. Influence of melatonin treatment on human circadian rhythmicity before and after a simulated 9-hr time shift. J. Biol. Rhythms. 1991, 6, 235–248. [Google Scholar] [CrossRef]
  207. Gramajo, A.L.; Marquez, G.E.; Torres, V.E.; Juárez, C.P.; Rosenstein, R.E.; Luna, J.D. Therapeutic benefit of melatonin in refractory central serous chorioretinopathy. Eye 2015, 29, 1036–1045. [Google Scholar] [CrossRef] [PubMed]
  208. Menczel Schrire, Z.; Phillips, C.L.; Chapman, J.L.; Duffy, S.L.; Wong, G.; D’Rozario, A.L.; Comas, M.; Raisin, I.; Saini, B.; Gordon, C.J.; et al. Safety of higher doses of melatonin in adults: A systematic review and meta-analysis. J. Pineal Res. 2022, 72, e12782. [Google Scholar] [CrossRef] [PubMed]
  209. Andersen, L.P.; Gögenur, I.; Rosenberg, J.; Reiter, R.J. The Safety of Melatonin in Humans. Clin. Drug Investig. 2016, 36, 169–175. [Google Scholar] [CrossRef]
  210. López-González, A.; Álvarez-Sánchez, N.; Lardone, P.J.; Cruz-Chamorro, I.; Martínez-López, A.; Guerrero, J.M.; Reiter, R.J.; Carrillo-Vico, A. Melatonin treatment improves primary progressive multiple sclerosis: A case report. J. Pineal Res. 2015, 58, 173–177. [Google Scholar] [CrossRef]
  211. Reiter, R.J.; Tan, D.X.; Sainz, R.M.; Mayo, J.C.; Lopez-Burillo, S. Melatonin: Reducing the toxicity and increasing the efficacy of drugs. J. Pharm. Pharmacol. 2002, 54, 1299–1321. [Google Scholar] [CrossRef]
  212. Reiter, R.J.; Mayo, J.C.; Tan, D.X.; Sainz, R.M.; Alatorre-Jimenez, M.; Qin, L. Melatonin as an antioxidant: Under promises but over delivers. J. Pineal Res. 2016, 61, 253–278. [Google Scholar] [CrossRef]
  213. Gunes, A.; Dahl, M.L. Variation in CYP1A2 activity and its clinical implications: Influence of environmental factors and genetic polymorphisms. Pharmacogenomics 2008, 9, 625–637. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The beneficial effects of melatonin (MT) on the gut–liver axis and its underlying mechanisms. MT produced in the gut acts on hepatic mitochondria. Within hepatic mitochondria, MT can elevate the transcription factor Nuclear factor erythroid 2-related factor 2 (Nrf2) to increase the expression of its downstream antioxidant enzymes and Sirtuin isoforms, as illustrated. As an antioxidant, MT also prevents oxidative stress and promotes autophagy/mitophagy to activate proper mitochondrial metabolic functions and energy production. MT also attenuates gut dysbiosis and intestinal barrier dysfunction, inflammation, and endotoxemia to prevent liver disease via the gut–liver axis. The molecular effects of MT on the different systems are also listed. The up and down arrows indicate the increased and decreased cellular functions and systems, respectively.
Figure 1. The beneficial effects of melatonin (MT) on the gut–liver axis and its underlying mechanisms. MT produced in the gut acts on hepatic mitochondria. Within hepatic mitochondria, MT can elevate the transcription factor Nuclear factor erythroid 2-related factor 2 (Nrf2) to increase the expression of its downstream antioxidant enzymes and Sirtuin isoforms, as illustrated. As an antioxidant, MT also prevents oxidative stress and promotes autophagy/mitophagy to activate proper mitochondrial metabolic functions and energy production. MT also attenuates gut dysbiosis and intestinal barrier dysfunction, inflammation, and endotoxemia to prevent liver disease via the gut–liver axis. The molecular effects of MT on the different systems are also listed. The up and down arrows indicate the increased and decreased cellular functions and systems, respectively.
Antioxidants 13 00043 g001
Table 1. Effects of MT shown in animal models of ALD.
Table 1. Effects of MT shown in animal models of ALD.
Stage of ALD and ModelSpecies UsedMT Dose and
Treatment Duration
Signaling Pathway
Affected by MT
Consequences of MT EffectsReference
Acute Ethanol-Induced Stress Male 2–3-month-old white mice10 mg/kg for 10 days↑ Mitochondrial
function
↑ Mitochondrial
respiration
↑ RCR, ADP/O, and
Vph
Modulates oxidative
phosphorylation;
mitigates subclinical endotoxemia
and oxidative stress
[166]
Steatosis/hepatitisAML-12 cells and 7-week-old C57BL wild-type mice 10 μmol/L in cell model and 5 mg/kg for 10 days in
Gao Binge Model
EGFR-BRG1-TERT
pathway
Downstream effects
of MT
[157]
HepatotoxicityAdult Male Rats50 mg/kg for 11 consecutive days ↓ Serum transaminases
↓ ALP
↑ GSH, GST
↓ NO
↓ TNF-α, p-NF-κB,
COX2
↓ Hepatic cellular
apoptosis
Decreased EtOH-
induced apoptosis
and inflammation
via JNK and TNF-α
signaling cascades
[162]
SteatosisMice and human samples10 mg/kg orally for last 2 weeks of 4-week alcohol
exposure
Once daily for
7 days via tail-vein
CRBN-YY1-CYP2E1

↓ CYP2E1
↓ ROS
↓ Serum AST and ↓ALT
↓ IL-6 and ↓TNF-α
↓ Hepatic TGs
↓ Hepatic cholesterol
Induction of SIRT1
acts via the CRBN-
YY1-CYP2E1 pathway to mitigate oxidative stress, improve liver function, prevent hepatic fat accumulation and
inflammation
[160]
Fat accumulationAdult male Sprague
Dawley rats
20 and 40 mg/kg
administration
↓ ALT, AST, and serum
and hepatic TG
↑ SOD
↓ MDA
↑ p-AMPK
↑ MT1R expression
↑ AMPK
↓ Lipid
accumulation
[164]
SteatosisFemale
Balb/C Mice
15 mg/kg via i.p. prior to ethanol for
3 days
↓ MMP-9 activity,
which then prevented
NF-κB translocation
to the nucleus after
EtOH exposure
↓ Total pathology score
but no significant effect
on transaminases
Prevented
inflammation
[161]
Chronic ALDMouse
model
10 mg/kg daily oral gavage for last 2 weeks of 4 weeks of ethanol↑ miR-497 expressionAmeliorates alcohol-
induced bile acid
synthesis by up-
regulating miR-497
expression and
attenuating the
BTG2-YY1 pathway
[167]
Note: Upward arrows (↑) indicate increase or elevation, while downward arrows (↓) represent decrease or suppression of the specific parameter(s) after MT exposure(s).
Table 2. Effects of MT shown in animal models of MASLD.
Table 2. Effects of MT shown in animal models of MASLD.
Stage of MASLD or
Liver Injury
Species Used in Study MT Dose/
Treatment Duration
Signaling Pathway Affected
by MT
MT EffectsReference
Endotoxin-
induced
Hepatotoxicity
Female Wistar rats10 mg/kg MT 30 min before LPS and 2 h after LPS↓ LPS
↑ GSH levels
↑ SOD activity
↑ Catalase activity
↓ Serum nitrite NO2
↓ TNF-α
↓ Hepatic necrosis
Mitigates endotoxin-
induced hepatotoxicity
↑ Antioxidant stores
↓ Oxidative stress
↓ Hepatic inflammation
and cellular death
pathways
[182]
MASLD
(Steatosis)
±HFD
Catalase-KO mice (CKO)
and HepG2
cells
500 μg/kg/day MT for 6 weeks↓ Liver weight
↓ Fat accumulation
Restored Aspect
Ratio (AR) and
Form Factor (FF)
values as measures
of mitochondrial
function
↑ mRNA expression
of FOXO1, PGC1β,
and PPAR-γ
Improved mitochon-
drial morphology
↑ mRNA expression
of CPT1, CPT2,
COX1, FGF21, Lcad,
Mcad, Aconitase,
IDH, SDH, MDH
↑ Hepatic [ATP]
Ablation of catalase
plays a role in MASLD,
and MT supports the
function of
mitochondria;
HFD-exposed CKO
mice exhibited cellular
lipid accumulation and
decreased mitochon-
drial biogenesis, which
was recovered with MT;
MT prevented fatty liver
development and main-
tained mitochondrial in-
tegrity in hepatocytes;
Mitigated oxidative
stress-induced mito-
chondrial dysfunction
and progression of
MASLD
[181]
HFD-mediated MASLD
lipogenesis and fibrosis
  • Liver-specific DNA PKcs-Knockout Mice
  • NR4A1-Knockout mice
20 mg/kg/day MT via i.p. for 12 weeks after 12 weeks HFD or LFDIn HFD-fed mice, MT
↓ NR4A1 level
↓ Hepatocyte vacu-
olization, steatosis,
and fibrosis
↓ MMP9
↓ VCAM1
↓ IL-6
↓ TNF-α
↓ TGF-β
↓ Mitochondrial
ROS production
↓ Mitochondrial
PTP opening
↑ ΔΨm mitochon-
drial inner mem-
brane potential
NR4A1/DNA-PKcs/p53
pathway, mitochondrial
fission, and mitophagy;
Prevented fat accumula-
tion and fibrosis by
inhibiting NR4A1. NR4A1 then activates
Drp-1-mediated mito-
chondrial fission, and
repressing BNIP3-medi-
ated mitophagy, which
protects mitochondria;
MT mitigated oxidative
stress and calcium over-
load by suppressing
fission
[142]
MASHMale 6–8-week-old CD-1 mice on a regular
diet
5 mg/kg MT
30 min before
2 mg/kg LPS
then 5 mg/kg
150 min after
LPS
↓ LPS-induced acti-
vation of SREBP-1c
↓ expression of
SREBP-1c genes
↓ serum and hepatic
TG levels
Prevents LPS-induced
fat accumulation
[183]
HepatitisMale 1-day-old Cherry Valley ducklings0.2 mg/mL MT
supplemented in drinking water for 2 weeks
Bacteriodetes
induced by
Ochratoxin A (OTA)
Firmicutes
Bacteriodetes/Firmi-
cutes Ratio
Bacteroides
Bacteroides uniformis
Turicibacter sangui-
nis↓ Serum LPS levels
↑ Protein expression
of Occludin and
tight junction pro-
tein-1 (TJP-1)
↑ Villi height and
crypt depth
↑ Villi height/crypt
depth ratio
Restored gut
histology
↓ TLR4, MyD88, p-
-IKBα, p-IKBα/IKBα
ratio, p-p65, liver
IL-1β level, liver IL-
6, liver TNF-α
↑ Liver IL-10
↓ Percentage of
inflammatory liver cells in histology quantified
Anti-inflammatory
Restored the physical
barrier of gut
Restored liver function
and inflammatory
markers
[184]
HFD-induced hepato-intestinal dysfunction and
inflammation
Male Sprague Dawley rats4 mg/kg/day for 2 weeks ↓ Perirenal fat
↓ Blood glucose
↑ TG levels by 3–4x
↓ Intestinal motility
↓ Liver weight
↑ GSH levels
↓ Myeloperoxidase
(MPO) activity
↓ Serum ALT levels
↑ Hypertrophic
goblet cell levels,
epithelium, and
brush border
↓ Vacuolization of
hepatocytes
Kept mitochondria
intact
Restored Liver function
and prevented fat
accumulation
Improved antioxidant
levels and prevented
oxidative stress
Improved intestinal
function and motility
[176]
Note: Upward arrows (↑) indicate increase or elevation, while downward arrows (↓) represent decrease or suppression of the specific parameter(s) after MT exposure(s).
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

LeFort, K.R.; Rungratanawanich, W.; Song, B.-J. Melatonin Prevents Alcohol- and Metabolic Dysfunction- Associated Steatotic Liver Disease by Mitigating Gut Dysbiosis, Intestinal Barrier Dysfunction, and Endotoxemia. Antioxidants 2024, 13, 43. https://doi.org/10.3390/antiox13010043

AMA Style

LeFort KR, Rungratanawanich W, Song B-J. Melatonin Prevents Alcohol- and Metabolic Dysfunction- Associated Steatotic Liver Disease by Mitigating Gut Dysbiosis, Intestinal Barrier Dysfunction, and Endotoxemia. Antioxidants. 2024; 13(1):43. https://doi.org/10.3390/antiox13010043

Chicago/Turabian Style

LeFort, Karli R., Wiramon Rungratanawanich, and Byoung-Joon Song. 2024. "Melatonin Prevents Alcohol- and Metabolic Dysfunction- Associated Steatotic Liver Disease by Mitigating Gut Dysbiosis, Intestinal Barrier Dysfunction, and Endotoxemia" Antioxidants 13, no. 1: 43. https://doi.org/10.3390/antiox13010043

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