Next Article in Journal
Valorization of Avocado Seed Wastes for Antioxidant Phenolics and Carbohydrates Recovery Using Deep Eutectic Solvents (DES)
Previous Article in Journal
Loss of PRDX6 Aborts Proliferative and Migratory Signaling in Hepatocarcinoma Cell Lines
Previous Article in Special Issue
COVID-19: Focusing on the Link between Inflammation, Vitamin D, MAPK Pathway and Oxidative Stress Genetics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Melatonin: A Potential Regulator of DNA Methylation

1
Department of Human Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Lwowska 1, 87-100 Toruń, Poland
2
Dr. Phillip Frost Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33125, USA
3
Department of Dermatology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
4
Pathology and Laboratory Medicine Service, VA Medical Center, Birmingham, AL 35294, USA
5
Department of Cell Systems and Anatomy, UT Health, Long School of Medicine, San Antonio, TX 78229, USA
6
Department of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany
*
Authors to whom correspondence should be addressed.
Antioxidants 2023, 12(6), 1155; https://doi.org/10.3390/antiox12061155
Submission received: 15 March 2023 / Revised: 17 May 2023 / Accepted: 24 May 2023 / Published: 25 May 2023
(This article belongs to the Special Issue Melatonin and Vitamin D in Diseases and Health)

Abstract

:
The pineal gland-derived indoleamine hormone, melatonin, regulates multiple cellular processes, ranging from chronobiology, proliferation, apoptosis, and oxidative damage to pigmentation, immune regulation, and mitochondrial metabolism. While melatonin is best known as a master regulator of the circadian rhythm, previous studies also have revealed connections between circadian cycle disruption and genomic instability, including epigenetic changes in the pattern of DNA methylation. For example, melatonin secretion is associated with differential circadian gene methylation in night shift workers and the regulation of genomic methylation during embryonic development, and there is accumulating evidence that melatonin can modify DNA methylation. Since the latter one impacts cancer initiation, and also, non-malignant diseases development, and that targeting DNA methylation has become a novel intervention target in clinical therapy, this review discusses the potential role of melatonin as an under-investigated candidate epigenetic regulator, namely by modulating DNA methylation via changes in mRNA and the protein expression of DNA methyltransferases (DNMTs) and ten-eleven translocation (TET) proteins. Furthermore, since melatonin may impact changes in the DNA methylation pattern, the authors of the review suggest its possible use in combination therapy with epigenetic drugs as a new anticancer strategy.

1. Melatonin Biosynthesis and Function

It has been widely discussed that melatonin has pleiotropic biological effects. This hormone is synthesized in a multistep process from tryptophan via tryptophan hydroxylase (TPH) to 5-hydroxytryptofan (tryptamine). Next, tryptamine is transformed using amino acid decarboxylase (AAD) to 5-hydroxytryptamine (serotonin), with its subsequent conversion to N-acetylserotonin (NAS) via aralkylamine N-acetyltransferase (AANAT). NAS is the immediate precursor of melatonin (N-acetyl-5-hydroxytryptmine), and this step is conducted using hydroxyindole-O-methyltransferase (HIOMT) (Figure 1) [1,2].
Due to its chemical structure, melatonin is a lipophilic compound that can easily penetrate biological membranes, thus defending enzymes, proteins, lipids and DNA from oxidative damage [3,4]. It has been shown to play a key role in the protection of cellular organelles, such as nuclei or mitochondria. This indoleamine hormone is synthesized and secreted in a circadian manner by the pineal gland. Its rhythmic secretion affects the sleep/wake cycle [1,5]. In addition, melatonin is produced in the nervous system, as well as in numerous peripheral organs, including the bone marrow, lymphocytes, eyes, gastrointestinal tract [6,7,8,9,10,11,12], and human [13,14,15,16] and rodent skin [17,18]. It exerts anti-oxidative actions by restoring UVB-induced oxidative stress and mitochondrial alterations [4,19], anti-inflammatory effects by decreasing the expression of heat shock protein 70 and pro-inflammatory cytokines [20], or anti-apoptotic properties; thus, melatonin and its metabolites significantly protect against DNA damage via the inhibition of 8-OH-deoxyguanosine formation and the enhancement of p53, as well as in promoting the expression of repair and anti-apoptotic proteins [13,21,22]. These pleiotropic effects are mediated by ubiquitously distributed cell membrane receptors (MT1 and MT2), and they have an impact on members of the RZR/ROR nuclear receptor family, e.g., retinoic acid-related orphan receptor α (RORα), and on membrane receptor-independent mechanisms [11,23,24,25,26].
The anti-oxidative attributes of melatonin are associated with its ability to decrease the number of reactive oxygen species (ROS) or reactive nitrogen species (RNS) involved in the activation of antioxidant enzymes [27]. The latter way requires transcription factor—nuclear erythroid factor 2 (Nrf2)—to play a specific role. Under physiological conditions, Nrf2 is ubiquitinated via ligase E3 complex Keap1-Cul3-Rbx1, which is followed by its proteasomal degradation. However, under oxidative stress, Nrf2 degradation is suppressed, allowing for its translocation to the nucleus and binding to antioxidant response element 2 (ARE) [28,29], which is located in the regulatory regions of genes encoding numerous antioxidant enzymes, including superoxide dismutase (Mn-SOD, Cu/Zn-SOD), catalase (CAT), γ-glutamylcysteine synthetase (γ-GCS), heme oxygenase-1 (HO-1), NADPH quinone dehydrogenase-1 (NQO1), or glutathione peroxidase (GPx) [4,19,30,31,32,33]. Apart from this, melatonin affects Nrf2 via the inhibition of degradation and the promotion of its nuclear translocation [29,31,34]. Moreover, a recent in vitro study revealed that 1 nM melatonin can effectively stimulate Nrf2 via an increase in the expression levels of antioxidant and proteasome genes linked with the Keap1-Nrf2-ARE pathway [28].
Earlier in vitro and in vivo studies revealed that melatonin can inhibit cancer cell growth. Melatonin regulates the immunosurveillance of cancer cells, via stimulating natural killer (NK) cells, monocytes, leukocytes, interleukins, and interferon-γ, as well as activating the cytokine system and cytotoxic activity (Figure 2) [35]. Melatonin reduces the rates of cancer initiation, progression, metastasis and migration [36,37,38,39], angiogenesis, and inflammation [40,41,42]. On the other hand, melatonin can regulate the immune system [43,44] and oxidative stress [19,30,38,45,46,47,48], thereby repressing many malignancies [49].

2. Alterations in DNA Methylation

2.1. DNA Methylation

DNA methylation results from the attachment of a methyl group to the fifth carbon atom of cytosine in DNA [50]. DNA methylation takes part in fundamental processes in an organism. Under physiological conditions, DNA methylation patterns are not random, and they are controlled and tissue-specific [51]. The process of DNA methylation is driven by DNA methyltransferases (DNMTs), which have the ability to transfer a methyl group from S-adenosyl-L-methionine to a DNA base—cytosine [52]. There are two primary means of DNA methylation: maintenance methylation driven by DNMT1 [53], and de novo methylation caused by DNMT3A and DNMT3B [54] (Figure 3).
The presence of modified cytosine in promoter regions can affect gene expression. As a general rule, the promoter regions of active genes are hypomethylated, while silenced genes are typically hypermethylated [55]. There are two plausible means of gene expression regulation driven by changes in DNA methylation machinery. Regarding the first of these, 5-mC in DNA is an obstacle for transcription factors by the inhibition of RNA polymerase II binding and the following transcription complexes formation [56]. Recent research revealed, however, that some of transcription factors are dependent of methylated CpG islands; hence, they can interact with hypermethylated DNA [57]. The second one involves 5-mC interaction with specific proteins that recognize the methylated bases of CpG islands—methyl-CpG binding proteins (MeCPs). MeCPs are a heterogeneous group that contain a methyl-CpG binding domain (MBD) [58]. They regulate transcription process through the inhibition of transcription factor binding to DNA or through interaction with other proteins, leading to silencing via heterochromatin formation, which is an inactive form of chromatin [59].

2.2. Active DNA Demethylation

A milestone in understanding the dynamic changes in the epigenome was the discovery of ten-eleven translocation (TET) enzymes (TET1, TET2, and TET3). In 2009, Tahiliani et al. [60] demonstrated that TET enzymes (initially regarded as chromosomal translocations in genes MLL and LCX (t(10;11)(q22;q23)) in leukemia patients [61,62] have the ability to change the DNA methylation pattern [60]. TET enzymes cause the oxidation of 5-mC to 5-hydroxymethylcytosine (5-hmC), leading to DNA demethylation [60]. Subsequent studies revealed that 5-hmC can be further hydroxylated to 5-formylcytosine (5-fC) and 5-carboxylcytosine (5-caC) [63]. Modified cytosines (5-fC or 5-caC) are recognized and excised by thymine DNA glycosylase (TDG), a key enzyme belonging to base excision repair (BER) machinery. As a result, 5-fC or 5-caC are replaced by unmodified cytosine; thus, TDG supports the active DNA demethylation process [64,65] (Figure 4). The loss of methylation marks (5-mC or 5-hmC) can passively occur as a result of mitosis. However, active DNA demethylation is a dynamic process that is independent of the cell cycle.
Apart from the direct regulation of the 5-mC level, TETs can also interact with other proteins, thus affecting DNA methylation. A study on mice embryonic cells indicated that TET1 can attach to Polycomb repressive complex 2 (PRC2) and impact the reduction of 5-hmC in promoter regions, followed by gene repression [66]. It was suggested that it is one of the mechanisms that occurs during cell differentiation, as it was detected in the promoter regions of developmental genes [67]. The association between TET1 and Sin3a—a multifunctional transcription regulator—was also indicated as one of the processes involved in the development and reprogramming associated with transcription inhibition [68,69]. TET proteins can have an interplay with O-GlcNAc transferase (OGT). This interaction can positively influence stem cell production in tet mutant zebrafish embryos. Moreover, it can affect the TET2 and 5-mC levels [70].
Modifications in the DNA methylation profile may be a trigger for transcriptional changes, including the activation or silencing of oncogenes and suppressor genes contributing to cell cycle changes, apoptosis inhibition, and the uncontrolled growth of cells, followed by carcinogenesis [71,72,73]. Global DNA hypomethylation is a characteristic hallmark of cancer [74,75], which manifests more frequently in the advanced stages of carcinogenesis [76,77,78,79,80]. In addition, another commonly observed phenomenon in cancer cells is the hypermethylation of CpG islands located in the promoter regions of genes [75]. Many reports have documented that the balance between DNA methylation and demethylation is disturbed in a variety of tumors, as well as TET and DNMT expression [74,81,82,83]. Most changes in TET and DNMT activity are not associated with genetic mutations, indicating that other factors may be responsible for these changes. Based on published data, it is suggested that melatonin may be one of the chemicals that plays a significant role in epigenetics and maintaining the DNA methylation pattern.

3. Melatonin and Its Role in DNA Methylation

3.1. Melatonin Regulates the Expression of DNMTs

The wide range of the biological effects of melatonin suggests that this indoleamine has a key function in modulating gene expression, including genes related to epigenetic processes. It was demonstrated that melatonin can impact the DNA methylation level, thus stimulating cell differentiation [84]. Additionally, melatonin significantly decreases the methylation level (5-mC) via downregulating the expression of DNMT1 and MeCP2. Of note, the expression levels of DNMT3A and DNMT3B were not affected by the addition of melatonin itself [84]. On this basis, it may be assumed that melatonin exerts a greater impact on the maintenance of DNA methylation occurring during DNA replication. Herein, a question should be stated: how does melatonin influence DNA methyltransferases? One of the answers was considered by Korkmaz and Reiter [85], and the authors hypothesized that based on their structural resemblance, melatonin and/or its derivatives might potentially serve as DNMT inhibitors via impacting DNMTs transcription or via binding to their catalytic center [85] (Figure 5).

3.2. Melatonin and Its Possible Impact on TET Proteins

Recent studies imply that the melatoninergic regulation of DNA methylation, which is followed by changes in gene expression, is not only associated with its impact on DNMTs, but also with other pathways possibly controlled by this indoleamine. A lot of the DNA methylation status in the genome stems from DNA demethylation driven by TET proteins [86]. Since melatonin influences DNMTs expression [84], it seems plausible that this compound may be a potential candidate that influences active DNA demethylation as well. Indeed, mouse embryos with knocked-out aralkylamine N-acetyltransferase (AANAT), which is the rate-limiting enzyme in melatonin biosynthesis, resulted in a decrease in TET2 expression and in changes in DNA methylation. Moreover, this process was reversible after melatonin supplementation [87]. Hence, the authors proposed that physiological embryo development requires the secretion of melatonin, as well as TET2 expression, to preserve the DNA methylation pattern and to achieve correct cellular differentiation [87]. Melatonin’s effect on TET2 upregulation was also shown in an in vitro study, where melatonin supplementation led to an increase in TET2, followed by the methylation inhibition of ubiquinol-cytochrome c reductase core protein 1 (UQCRC1), which eventually resulted in improved mitochondrial function [88]. Melatonin was also successfully used to inhibit TET1 expression and the DNA demethylation of metabotropic glutamine receptor subtype 5 (mGluR5), which is implicated in neuropathic pain [89]. The effect of the downregulation of TET1, followed by the decrease in the 5-hmC level at the position of the mGluR5 promoter, was demonstrated via the interaction of melatonin with MT2 receptor [89]. Moreover, melatonin can also significantly increase the TET1 and TET3 expression levels after exposure of mycotoxin Enniatin B1 (EB1) to porcine embryos [90]. This was consistent in another study on bovine oocytes, where melatonin supplementation significantly increased the TET1, TET3, and TET2 mRNA and protein levels [91]. It seems that melatonin can differently impact the regulation of TET1, TET2, and TET3 expression. However, it needs to be emphasized that TETs differ significantly in terms of expression during cell development and differentiation [92]. Moreover, DNA demethylation may be not driven the same way by all TET proteins. It was implied that hydroxylation of 5-mC to 5-hmC can be led by all TET proteins; however, the subsequent oxidation of 5-hmC into 5-fC and/or into 5-caC may only be driven by TET2 or TET3 [93].
These data suggest the possibility that melatonin may influence TET gene expression; although, the mechanism remains unknown. One explanation could be the association between melatonin and mitochondrial metabolism, especially in terms of tricarboxylic acid cycle metabolites. As it was demonstrated in mice study, melatonin plays a significant role in increasing the α-ketoglutarate (α-KG, 2-oxoglutarate) level, which is followed by TET-mediated DNA demethylation [94]. TET proteins, as members of 2-oxoglutarate and Fe2+-dependent dioxygenases (2-OGDD), require 2-oxoglutarate as a cofactor to maintain their catalytic activity [95,96] (Figure 5). Thus, this action of melatonin may be of importance in the regulation of DNA methylation due to its potential impact on TET expression.

3.3. Melatonin and DNA Methylation Changes under Artificial Light at Night

Melatonin synthesis is disrupted under artificial light at night (ALAN) [97]. Pineal melatonin synthesis is regulated by intrinsically photosensitive retinal ganglion cells containing photopigment melanopsin, which are particularly sensitive to exposure to short-wavelength light [98]. Even brief exposition [99] to ALAN may result in the suppression of melatonin secretion. Previous studies demonstrated that reduced melatonin levels provoked via ALAN may be involved in increased cancer incidence, especially hormone-dependent cancers (extensively reviewed in [100]). Further studies revealed that ALAN may be responsible not only for the increase in tumor growth, but also for developing global DNA hypomethylation in the tumors of BALB/c short-day-acclimated mice inoculated with 4T1 breast cancer cells [101]. The same mouse model was also used to detect lower levels of DNMT, along with global DNA methylation, after repeated exposure to ALAN [102]. Interestingly, a previous study documented that the ALAN wavelength is critical for evoking changes in DNA methylation status. A shorter wavelength, up to 500 nm, which is equivalent to that of normal blue LED light, is correlated with tumor development, melatonin suppression, and global DNA methylation. Exposition to longer wavelengths clearly has a weaker impact on melatonin suppression and the DNA methylation status [103]. In each of these experimental studies, melatonin administration significantly diminished ALAN effects on tumor growth, as well as on the DNA methylation level [101,102,103]. Changes in the DNA methylation status after ALAN exposure were also observed in rats lacking malignancies [104]. Moreover, it was previously reported that melatonin can impact cancer metabolism, possibly via epigenetic mechanisms [105,106]. In line with the aforementioned studies, it was demonstrated that ALAN exposure is associated with DNA hypomethylation in pancreatic tissue, as well as with lower levels of glucose and insulin in rats, suggesting its significant effect on metabolic responses [104]. In addition, a recent study indicated that ALAN exposure, particularly among elderly patients, is associated with diabetes, cardiovascular diseases, and obesity incidence [107]. Furthermore, the hypermethylation of MTNR1B was recently suggested as a novel epigenetic marker for atherosclerosis [108], which is considered to be a metabolic disease. Taken together, these studies demonstrate the close correlation between melatonin function and the status of DNA methylation. There is a potential role for melatonin in promoting alterations of DNA methylation of metabolic- and carcinogenic-related genes. It is suggested that the association between these factors should be investigated in future studies, as exposition to artificial light at night is acknowledged to be a cancer risk factor [109].

3.4. Melatonin and Circadian Clock Epigenetics

The latest research has highlighted that changes in melatonin synthesis and the circadian rhythm are related to cancer development [110,111,112]. There is a relationship between disturbances in the circadian rhythm and genomic instability [113,114]. Moreover, previous research revealed that the maintenance of circadian rhythmicity also involves epigenetic mechanisms [115,116]. Furthermore, the hypermethylation of promotor regions of circadian-cycle-dependent genes was observed in breast cancer cells [114], and melanoma [117], pan-renal cell carcinoma [118], cervical and esophageal cancer [119], and hepatocellular carcinoma cells [120].
The studies involving night shift workers indicated that melatonin secretion is associated with distinctive methylation pattern of genes implicated in the circadian cycle [112,121]. A cross-sectional study, including nurses and midwives who worked day shifts and rotating night shifts, showed differential blood-based DNA methylation statuses in the circadian genes in the latter group [121]. The hypomethylation of promoters was demonstrated by the CLOCK genes, PER1 and PER2 [121], which play an important role in regulation during sleep and sleep disorders [122]. Interestingly, hypomethylation status of rotating-shift workers was associated with the reduced methylation of selected circadian genes promoters [121]. A recent study involving female night shift workers in Canada indicated that promoters of circadian genes, PER3 (a member of PERIOD genes, along with PER1 and PER2) and MTNR1A (encoding MT1 melatonin receptor), are hypomethylated [112]. Changes in the DNA methylation of circadian genes have also been associated with the differential secretion of melatonin: night shift workers had increased levels of melatonin and peak melatonin secretion was reached later [112]. Furthermore, a large cohort study in Finland revealed the presence of a single-nucleotide polymorphism (SNP), rs12506228, downstream of the MT1 gene in the vast majority of shift workers. As shown in the study, individuals carrying rs12506228 had significantly lower levels of MT1, which is linked to hypermethylation of the regulatory region of MT1 [123].

3.5. Melatonin and Methylation during Cell Development and Differentiation

It has emerged in the literature that melatonin can also regulate methylation patterns during cell development and differentiation. The study involving breeding hamsters indicated that winter-like melatonin significantly decreased the expression of DNA methyltransferases in hypothalamus, followed by the hypomethylation of the dio3 promoter responsible for gonadal regression [124]. In a mouse study, females treated with decabromodiphenyl ethane (DBDPE)—a flame retardant, proven to have an impact on oocyte toxicity—showed the beneficial effect of melatonin treatment on preimplantation embryo development via the reduction of the number of ROS and the decrease in DNA methylation [68]. Similarly, a favorable effect of melatonin was observed on mice oocytes treated with mycotoxin deoxynivalenol (DON). Exposure of melatonin to mice oocytes during maturation decreased the level of DNA methylation evoked via the previous treatment with DON [125]. Furthermore, porcine embryos exposed to another mycotoxin, EB1, demonstrated alterations in the expression of genes involved in DNA methylation and demethylation, followed by changes in the DNA methylation pattern and the inhibition of embryos development. After melatonin supplementation, methylation changes were mitigated [90]. Melatonin can also promote the development of cloned embryos via an increase in blastocysts formation, the inhibition of embryos apoptosis, and the enhancement of nuclear remodeling [126]. In addition, it was demonstrated that melatonin’s impact on embryos development is associated with its influence on DNA methylation reprogramming. Melatonin can promote changes in the DNA methylation of pluripotency and tissue-specific genes, leading to the improvement of the embryo developmental progress [126]. A recent study reveal that mice embryos have the ability to gradually synthesize melatonin during development. However, the knockdown of rate-limiting enzyme in melatonin synthesis resulted in DNA hypomethylation in blastocysts, followed by the inhibition of further development [87]. Melatonin was also found to have an impact on stem cell differentiation. A recent in vitro study on human dental pulp cells (hDPCs) indicated that melatonin can promote the odontogenic differentiation of hDPCs, with the simultaneous downregulation of global DNA methylation and MeCP [84]. It needs to be emphasized that the loss of methylation stimulates stem cell differentiation, whereas hypermethylation promotes the stemness of cells [127,128]. Moreover, the expression level of DNMT1 was decreased; however, this change was melatonin-independent [84]. Thus, the mechanism underlying hDPCs differentiation after melatonin supplementation is related to a specific interaction between the proteins involved in global DNA methylation changes.
The aforementioned research suggest that melatonin secretion or supplementation can alter the DNA methylation pattern. Amongst the epigenetics mechanisms, changes in the DNA methylation pattern are the most often described ones, and 5-mC is regarded as a central epigenetic marker [129]. We hypothesize that, apart from many other functions, melatonin may also drive DNA methylation changes. Therefore, the exploration of the potential role of melatonin in DNA methylation changes may contribute to a better understanding of the underlying mechanisms of cancer pathogenesis. Furthermore, given the fact that epigenetic modifications are reversible [130,131,132,133], the modulation of these changes is emerging as a therapeutic intervention. Novel developments in this field have increased the need for inventing new drugs or combined therapies for use in cancer treatment on the epigenome.

4. Melatonin as a Part of Combined Epigenetic Therapy

4.1. Epigenetic Drugs Limitations

There are several drugs that modulate DNA methylation. The first agents that were proven to exert demethylation activity were the nucleoside derivatives: 5-azacytidine (5-AZA) and its analog—5-aza-2-deoxycytidine (decitabine, DAC) [134]. These compounds are hypomethylating agents, as they have the ability to be incorporated into DNA, instead of cytosine. This results in the arresting of DNMTs due to covalent bond formation, followed by the inhibition of DNMT function [135]. Moreover, these bonds disturb DNA functions, leading to the initiation of DNA damage signaling and DNMTs degradation. The subsequent round of DNA replication is, therefore, associated with the loss of DNA methylation markers [135,136,137]. Despite their clinical success in comparison to that of conventional anti-cancer therapy [138,139,140,141,142], the use of DNMT inhibitors has a number of limitations. Several studies have shown that the treatment responses among patients receiving these hypomethylating agents vary widely. The largest discrepancies are associated with the transcriptional responses to demethylation and the activation of previously silenced promotors after treatment [143,144,145,146,147]. The second limitation of these agents is the instability of 5-AZA and DAC in alkaline and acidic solutions [148,149]. These drugs are only stable in neutral solutions at temperatures relevant for routine clinical use and maintenance, with the preservation of their ability to prevent DNA methylation [135]. Moreover, the activity of azanucleosides is based not only on methylation inhibition, but also on dose-related toxicity, which limits their usefulness [150]. Both, 5-AZA and DAC exhibit significant cytotoxicity at the highest tolerated concentrations in chronic myeloid leukemia cell lines, as well in patients with hematopoietic malignancies [151,152]. Furthermore, prolonged treatment with 5-AZA and DAC (48 h) has a more significant impact on DNMT1 depletion, the induction of DNA damage, and the upregulation of gene expression [153]. Both drugs, however, have relatively short substance elimination half-lives [154,155]. At present, 5-azacytidyne and 5-aza-2-deoxycytidine are the only DNMT inhibitors that have been approved for the treatment of hematological malignancies, including acute myeloid leukemia (AML), chronic myelomonocytic leukemia (CMML), and myelodysplastic disorders (MDS) [156,157]. Currently, there are ongoing clinical trials investigating the plausible use of DNMT inhibitors for the treatment of solid tumors [158]. In fact, the limitations of studies on DNMT inhibitors have raised many questions in need for the further investigation of novel DNA methylation inhibitors or combined therapies relative to their potential minimal toxicity and improved pharmacological properties.

4.2. Melatonin’s Potential in Combined Therapy

In line with exploring new combined anticancer therapies, some experimental and clinical research has indicated the beneficial effect of melatonin when it is combined with conventional chemo- or radiotherapy. A recent meta-analysis of sixteen trials, in which a combined treatment with melatonin and chemotherapy was used, revealed that patients who received melatonin with other therapies had better outcomes and survival rates, as well as less frequent side effects, than the patients who did not receive supplemental melatonin did [159]. Interestingly, the most striking result from this meta-analysis is that the same beneficial effects of combined chemotherapy with melatonin were noticed in patients with multiple types of cancer. Furthermore, in vitro studies demonstrated that melatonin can enhance the effect of 5-fluorouracil in colon cancer cells via the activation of caspase-9 and caspase-3 in the mitochondrial apoptotic pathway [160], increasing oxidative stress, downregulating genes and proteins related to apoptosis resistance [161], and inhibiting NF-κB/iNOS and PI3K/AKT signaling pathways [162]. Further evidence for the augmenting effect of melatonin on the anticancer activity of conventional chemotherapy comes from studies of doxorubicin [163,164] and paclitaxel [165].
In light of the recent findings that melatonin combined with conventional anti-cancer therapy could limit the adverse effect of chemotherapy and increase its therapeutic efficacy, including reducing drug resistance [159,160,161,162,166,167], its simultaneous use with DNMT inhibitors or other epigenetic drugs may be a more efficient approach to avoid the limitations of epigenetic drugs. A growing number of clinical trials has investigated the plausible favorable effects of therapies when anticancer drugs are combined with DNMT inhibitors. Currently, 255 clinical trials for 5-AZA with chemotherapy and 175 clinical trials for DAC with chemotherapy have already been registered (clinicaltrials.gov). An in vitro study by Hartung et al. [168] demonstrated that treatment of rat C6 glioma cells with 5-AZA significantly upregulated the expression of the MT1 melatonin receptor. Epigenetic silencing via the hypermethylation of CpG islands of MT1 gene in oral squamous cell carcinoma (OSCC) cell lines was mitigated after a treatment with 5-AZA [169]. Notably, it was previously reported that MT1 loss is associated with a worse prognosis in oral squamous cell cancer [169], triple-negative breast cancer [170,171], and in leiomyosarcoma patients [172]. Moreover, increased MT1 expression has a favorable effect on the resensitization of breast cancer cells to paclitaxel with the concomitant upregulation of tumor suppressor genes [173] (Table 1). This is in accordance with earlier studies, where the association between MT1 and cancer incidence was reviewed by Hill et al. [174]. The decrease in melatonin and MT1, which is associated with age, was suggested to be one of the risk factors of breast tumor growth [174]. Moreover, not only silencing itself, but also specific methylation pattern of MT1 is significant. A recent study by Lesicka et al. [175] revealed that the profiles of MT1 methylation were significantly different among patients with depression in comparison to those of healthy subjects. In addition, specific single nucleotide polymorphism (SNP) variants in MTNR1B combined with CDKN2A and MGMT genes hypermethylation were found to cause worse 5-year overall survival in colorectal cancer patients, suggesting significant role of SNP variants of MTNR1B in cancer pathogenesis [176]. A particular SNP variant of the MT1 gene was also detected downstream of MT1 [123]. In silico analyses demonstrated that this variant may be responsible for the silencing of MT1 in the brain via DNA methylation. Moreover, authors suggested that the mentioned SNP variant is related to the exhaustion of shift workers, potentially leading to a decrease in melatonin signaling and an increase in cancer incidence in this group [123].

4.3. Melatonin Plays Significant Role in Changing of DNA Methylation Pattern of Cancer-Related Genes

Lee et al. [177] who studied the characteristics of DNA methylation profiles in breast cancer cells (MCF-7), identified several genes candidates as those affected by melatonin exposure. The authors analyzed genes which mRNA expression level was negatively correlated with the DNA methylation status. After a treatment with melatonin, MCF-7 cells displayed the significant upregulation of tumor suppressor gene (GPC3), as well as the downregulation of oncogenic genes (EGR3 and POU4F2-Brn-3b) [177]. Another study on brain tumor stem cells and glioma cells (A172) proved that melatonin in combination with chemotherapeutic agents influences the hypermethylation of the promoter of the ABCG2/BCRP gene responsible for multidrug resistance and tumor recurrence [178] (Table 1). These findings suggest the novel anti-cancer activity of melatonin. It is, therefore, likely that melatonin may be involved in changing the DNA methylation status of cancer-related genes.
Table 1. Recent findings promoting melatonin as a methylation modulator.
Table 1. Recent findings promoting melatonin as a methylation modulator.
Studied Cell Line/Mice ModelMain FindingReferences
Oral cell carcinoma cell linesHypermethylation of CpG islands in MT1 gene restored after treatment with 5-AZA[169]
Rat C6 glioma cellsTreatment with 5-AZA increases expression of MT1 gene[168]
Porcine oocytesTreatment with melatonin increased level of 5-mC and DNMT in prolonged culture[179]
4T1 mouse breast cancer modelMelatonin can mitigate the changes in DNMT activity and global DNA methylation level in mice exposed to artificial light at night[102,103]
Brain tumor stem cell, A172 malignant glioma tumor cellsMelatonin treatment increases methylation of genes ABCG2/BCRP associated with multidrug resistance[178]
MCF-7 mice breast cancer modelMelatonin treatment increases the favorable effect of paclitaxel by impact on IL-6, STAT3 and DNMT1 gene expression[173]
MCF-7 breast cancer cellsSupplementation of melatonin increases expression of tumor suppressor gene and decreases expression of oncogenic genes possibly by impacting on methylation level[177]
It should be emphasized that melatonin itself may impact DNMTs via regulating mRNA or protein expression [84] (discussed above). Furthermore, alterations in the methylation of promoters of circadian rhythm genes may also impact the sensitivity of anticancer drugs [118]. Hence, it is hypothesized that DNMT inhibitors improve the therapeutic potential of melatonin and that the improvement may be substantial. Future studies on the therapeutic efficacy of these agents in combination are, therefore, needed.

5. Conclusions and Future Perspectives

A large and growing body of literature has investigated melatonin as a promising candidate for cancer prevention or treatments. Herein, we discussed melatonin’s potential to modulate DNA methylation. Since night shift work is associated with the hypermethylation of genes involved in the circadian biorhythm [180], which is also involved in changes in melatonin secretion [112], it is plausible that melatonin may also directly change DNA methylation. Given the fact that DNA methylation modifications are the most prevalent epigenetic processes [181], melatonin may affect the regulation of gene expression. Additionally, well-described melatonin roles, such as anti-apoptotic or anti-oxidative molecule ones, can be also related to alterations in the DNA methylation pattern. Previous studies have noted the importance of changes in DNA methylation pattern in genes related to apoptosis in cancer cells undergoing this process [182,183,184]. Moreover, a recent in vivo study revealed that mice exposed to formaldehyde, known for provoking oxidative stress and inflammation, showed a more obvious trend toward global DNA hypermethylation compared to that of the formaldehyde-treated group due to simultaneous melatonin supplementation [185]. Furthermore, the treatment of cumulus cells of prepubertal lambs with melatonin was shown to decrease the methylation level of promoter regions of SOD, GPx, and CAT genes [186]. Therefore, it seems plausible that the anti-oxidative effect of melatonin is also associated with epigenetic changes in the genes involved in oxidative homeostasis.
Lastly, due to the fact that epigenetic alterations are eminently reversible [130,131,132], they have emerged as attractive targets for cancer therapy. However, the currently used DNMT inhibitors have limitations (an inconsistent demethylation response and high toxicity level during prolonged use), which can be potentially overcome by using combined anti-cancer therapies. Accordingly, melatonin has beneficial effects when it is combined with chemotherapy [159,160,161,162,166,167], and it can regulate the expression of DNMT [84] or TET [87,89]. Therefore, it may also be more efficient when it is added to epigenetic therapy. Hence, the administration of melatonin, along with DNMT inhibitors in anticancer therapy, especially in tumors where expression of MT receptors is associated with a better prognosis [169,170,171,172], should be, therefore, under consideration at both experimental and clinical levels.

Author Contributions

Conceptualization, writing original draft preparation and supervision, K.L. and K.K.; extensive editing, revision of the manuscript, K.L., A.T.S., R.J.R., R.P., M.B., K.S. and K.K. All authors have read and agreed to the published version of the manuscript.

Funding

Writing of this review was supported by the Polish National Science Center (2018/29/N/NZ3/02514) (K.L.), Mobility Grant of Nicolaus Copernicus University postdoctoral fellows under the “Excellence Initiative—Research University” program Emerging Fields (EF)/Sustainable development (SD) funds (4101.00000024) (K.L.), and in part by an Endowed Frost Scholarship (R.P.), by National Institutes of Health (NIH): R01AR073004-01A1 (A.T.S.), R01AR071189-01A1 (A.T.S.), R21AI149267-01A1 (A.T.S.), by VA merit award (1I01BX004293-01A1) (A.T.S.), and by the German Research Foundation (Deutsche Forschungsgemeinschaft (DFG)): KL2900/2-1 (K.K.), TR156/C05-246807620 (K.S.), SFB1009/B11-194468054 (K.S.), SFB1066/B06-213555243 (K.S.), SFB1450/C06-431460824 (K.S.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Reiter, R.J. Pineal Melatonin: Cell Biology of Its Synthesis and of Its Physiological Interactions. Endocr. Rev. 1991, 12, 151–180. [Google Scholar] [CrossRef]
  2. Roseboom, P.H.; Namboodiri, M.A.; Zimonjic, D.B.; Popescu, N.C.; Rodriguez, I.R.; Gastel, J.A.; Klein, D.C. Natural Melatonin “knockdown” in C57BL/6J Mice: Rare Mechanism Truncates Serotonin N-Acetyltransferase. Mol. Brain Res. Brain Res. 1998, 63, 189–197. [Google Scholar] [CrossRef]
  3. Naranjo, M.C.; Guerrero, J.M.; Rubio, A.; Lardone, P.J.; Carrillo-Vico, A.; Carrascosa-Salmoral, M.P.; Jiménez-Jorge, S.; Arellano, M.V.; Leal-Noval, S.R.; Leal, M.; et al. Melatonin Biosynthesis in the Thymus of Humans and Rats. Cell. Mol. Life Sci. 2007, 64, 781–790. [Google Scholar] [CrossRef]
  4. Janjetovic, Z.; Jarrett, S.G.; Lee, E.F.; Duprey, C.; Reiter, R.J.; Slominski, A.T. Melatonin and Its Metabolites Protect Human Melanocytes against UVB-Induced Damage: Involvement of NRF2-Mediated Pathways. Sci. Rep. 2017, 7, 1274. [Google Scholar] [CrossRef]
  5. Grant, S.G.; Melan, M.A.; Latimer, J.J.; Witt-Enderby, P.A. Melatonin and Breast Cancer: Cellular Mechanisms, Clinical Studies and Future Perspectives. Expert Rev. Mol. Med. 2009, 11, e5. [Google Scholar] [CrossRef]
  6. Tosini, G.; Fukuhara, C. Photic and Circadian Regulation of Retinal Melatonin in Mammals. J. Neuroendocrinol. 2003, 15, 364–369. [Google Scholar] [CrossRef]
  7. Konturek, S.J.; Konturek, P.C.; Brzozowska, I.; Pawlik, M.; Sliwowski, Z.; Cześnikiewicz-Guzik, M.; Kwiecień, S.; Brzozowski, T.; Bubenik, G.A.; Pawlik, W.W. Localization and Biological Activities of Melatonin in Intact and Diseased Gastrointestinal Tract (GIT). J. Physiol. Pharmacol. 2007, 58, 381–405. [Google Scholar]
  8. 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]
  9. Wiechmann, A.F.; Summers, J.A. Circadian Rhythms in the Eye: The Physiological Significance of Melatonin Receptors in Ocular Tissues. Prog. Retin. Eye Res. 2008, 27, 137–160. [Google Scholar] [CrossRef]
  10. Reiter, R.J.; Tan, D.-X.; Fuentes-Broto, L. Melatonin: A Multitasking Molecule. Prog. Brain Res. 2010, 181, 127–151. [Google Scholar] [CrossRef]
  11. Hardeland, R.; Cardinali, D.P.; Srinivasan, V.; Spence, D.W.; Brown, G.M.; Pandi-Perumal, S.R. Melatonin—A Pleiotropic, Orchestrating Regulator Molecule. Prog. Neurobiol. 2011, 93, 350–384. [Google Scholar] [CrossRef]
  12. Lanoix, D.; Lacasse, A.-A.; Reiter, R.J.; Vaillancourt, C. Melatonin: The Smart Killer: The Human Trophoblast as a Model. Mol. Cell. Endocrinol. 2012, 348, 1–11. [Google Scholar] [CrossRef]
  13. Slominski, A.T.; Semak, I.; Fischer, T.W.; Kim, T.-K.; Kleszczyński, K.; Hardeland, R.; Reiter, R.J. Metabolism of Melatonin in the Skin: Why Is It Important? Exp. Dermatol. 2017, 26, 563–568. [Google Scholar] [CrossRef]
  14. Slominski, A.T.; Hardeland, R.; Zmijewski, M.A.; Slominski, R.M.; Reiter, R.J.; Paus, R. Melatonin: A Cutaneous Perspective on Its Production, Metabolism, and Functions. J. Investig. Dermatol. 2018, 138, 490–499. [Google Scholar] [CrossRef]
  15. Slominski, A.T.; Kim, T.-K.; Kleszczyński, K.; Semak, I.; Janjetovic, Z.; Sweatman, T.; Skobowiat, C.; Steketee, J.D.; Lin, Z.; Postlethwaite, A.; et al. Characterization of Serotonin and N-Acetylserotonin Systems in the Human Epidermis and Skin Cells. J. Pineal Res. 2020, 68, e12626. [Google Scholar] [CrossRef]
  16. Bocheva, G.; Slominski, R.M.; Janjetovic, Z.; Kim, T.-K.; Böhm, M.; Steinbrink, K.; Reiter, R.J.; Kleszczyński, K.; Slominski, A.T. Protective Role of Melatonin and Its Metabolites in Skin Aging. Int. J. Mol. Sci. 2022, 23, 1238. [Google Scholar] [CrossRef]
  17. Slominski, A.; Baker, J.; Rosano, T.G.; Guisti, L.W.; Ermak, G.; Grande, M.; Gaudet, S.J. Metabolism of Serotonin to N-Acetylserotonin, Melatonin, and 5-Methoxytryptamine in Hamster Skin Culture. J. Biol. Chem. 1996, 271, 12281–12286. [Google Scholar] [CrossRef]
  18. Slominski, A.; Pisarchik, A.; Semak, I.; Sweatman, T.; Szczesniewski, A.; Wortsman, J. Serotoninergic System in Hamster Skin. J. Investig. Dermatol. 2002, 119, 934–942. [Google Scholar] [CrossRef]
  19. Kleszczyński, K.; Bilska, B.; Stegemann, A.; Flis, D.J.; Ziolkowski, W.; Pyza, E.; Luger, T.A.; Reiter, R.J.; Böhm, M.; Slominski, A.T. Melatonin and Its Metabolites Ameliorate UVR-Induced Mitochondrial Oxidative Stress in Human MNT-1 Melanoma Cells. Int. J. Mol. Sci. 2018, 19, 3786. [Google Scholar] [CrossRef]
  20. Kleszczyński, K.; Zwicker, S.; Tukaj, S.; Kasperkiewicz, M.; Zillikens, D.; Wolf, R.; Fischer, T.W. Melatonin Compensates Silencing of Heat Shock Protein 70 and Suppresses Ultraviolet Radiation-Induced Inflammation in Human Skin Ex Vivo and Cultured Keratinocytes. J. Pineal Res. 2015, 58, 117–126. [Google Scholar] [CrossRef]
  21. Janjetovic, Z.; Nahmias, Z.P.; Hanna, S.; Jarrett, S.G.; Kim, T.-K.; Reiter, R.J.; Slominski, A.T. Melatonin and Its Metabolites Ameliorate Ultraviolet B-Induced Damage in Human Epidermal Keratinocytes. J. Pineal Res. 2014, 57, 90–102. [Google Scholar] [CrossRef] [PubMed]
  22. Skobowiat, C.; Brożyna, A.A.; Janjetovic, Z.; Jeayeng, S.; Oak, A.S.W.; Kim, T.-K.; Panich, U.; Reiter, R.J.; Slominski, A.T. Melatonin and Its Derivatives Counteract the Ultraviolet B Radiation-Induced Damage in Human and Porcine Skin Ex Vivo. J. Pineal Res. 2018, 65, e12501. [Google Scholar] [CrossRef] [PubMed]
  23. Pandi-Perumal, S.R.; Srinivasan, V.; Maestroni, G.J.M.; Cardinali, D.P.; Poeggeler, B.; Hardeland, R. Melatonin. Nature’s Most Versatile Biological Signal? FEBS J. 2006, 273, 2813–2838. [Google Scholar] [CrossRef] [PubMed]
  24. Reiter, R.J.; Tan, D.X.; Manchester, L.C.; Pilar Terron, M.; Flores, L.J.; Koppisepi, S. Medical Implications of Melatonin: Receptor-Mediated and Receptor-Independent Actions. Adv. Med. Sci. 2007, 52, 11–28. [Google Scholar]
  25. Slominski, A.T.; Zmijewski, M.A.; Jetten, A.M. RORα Is Not a Receptor for Melatonin. Bioessays 2016, 38, 1193–1194. [Google Scholar] [CrossRef]
  26. Hardeland, R. Melatonin and Retinoid Orphan Receptors: Demand for New Interpretations after Their Exclusion as Nuclear Melatonin Receptors. Melatonin Res. 2018, 1, 78–93. [Google Scholar] [CrossRef]
  27. 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]
  28. Torres, F.F.; Bernardo, V.S.; de Paula, C.P.; da Silva, J.P.M.d.O.; de Almeida, E.A.; da Cunha, A.F.; da Silva, D.G.H. Influence of Melatonin Treatment on Cellular Mechanisms of Redox Adaptation in K562 Erythroleukemic Cells. Genes 2022, 13, 2337. [Google Scholar] [CrossRef]
  29. Vriend, J.; Reiter, R.J. The Keap1-Nrf2-Antioxidant Response Element Pathway: A Review of Its Regulation by Melatonin and the Proteasome. Mol. Cell. Endocrinol. 2015, 401, 213–220. [Google Scholar] [CrossRef]
  30. Kleszczyński, K.; Zillikens, D.; Fischer, T.W. Melatonin Enhances Mitochondrial ATP Synthesis, Reduces Reactive Oxygen Species Formation, and Mediates Translocation of the Nuclear Erythroid 2-Related Factor 2 Resulting in Activation of Phase-2 Antioxidant Enzymes (γ-GCS, HO-1, NQO1) in Ultraviolet Radiation-Treated Normal Human Epidermal Keratinocytes (NHEK). J. Pineal Res. 2016, 61, 187–197. [Google Scholar] [CrossRef]
  31. Deng, Y.; Zhu, J.; Mi, C.; Xu, B.; Jiao, C.; Li, Y.; Xu, D.; Liu, W.; Xu, Z. Melatonin Antagonizes Mn-Induced Oxidative Injury through the Activation of Keap1-Nrf2-ARE Signaling Pathway in the Striatum of Mice. Neurotox. Res. 2015, 27, 156–171. [Google Scholar] [CrossRef] [PubMed]
  32. Ryšavá, A.; Vostálová, J.; Rajnochová Svobodová, A. Effect of Ultraviolet Radiation on the Nrf2 Signaling Pathway in Skin Cells. Int. J. Radiat. Biol. 2021, 97, 1383–1403. [Google Scholar] [CrossRef] [PubMed]
  33. Suwannakot, K.; Sritawan, N.; Prajit, R.; Aranarochana, A.; Sirichoat, A.; Pannangrong, W.; Wigmore, P.; Welbat, J.U. Melatonin Protects against the Side-Effects of 5-Fluorouracil on Hippocampal Neurogenesis and Ameliorates Antioxidant Activity in an Adult Rat Hippocampus and Prefrontal Cortex. Antioxidants 2021, 10, 615. [Google Scholar] [CrossRef] [PubMed]
  34. Sobhani, S.; Tehrani, A.-A.; Sobhani, G.; Fatima, S.; Ulloa, L.; Motaghinejad, M.; Atif, A. Melatonin Protects Against Titanium Oxide-Induced Neurotoxicity: Neurochemical, Neurobehavioral, and Histopathological Evidences. Biol. Trace Elem. Res. 2022. [Google Scholar] [CrossRef] [PubMed]
  35. Srinivasan, V.; Spence, D.W.; Trakht, I.; Pandi-Perumal, S.R.; Cardinali, D.P.; Maestroni, G.J. Immunomodulation by Melatonin: Its Significance for Seasonally Occurring Diseases. Neuroimmunomodulation 2008, 15, 93–101. [Google Scholar] [CrossRef]
  36. Cos, S.; Sánchez-Barceló, E.J. Melatonin and Mammary Pathological Growth. Front. Neuroendocrinol. 2000, 21, 133–170. [Google Scholar] [CrossRef]
  37. Reiter, R.J.; Rosales-Corral, S.A.; Tan, D.-X.; Acuna-Castroviejo, D.; Qin, L.; Yang, S.-F.; Xu, K. Melatonin, a Full Service Anti-Cancer Agent: Inhibition of Initiation, Progression and Metastasis. Int. J. Mol. Sci. 2017, 18, 843. [Google Scholar] [CrossRef]
  38. Bilska, B.; Schedel, F.; Piotrowska, A.; Stefan, J.; Zmijewski, M.; Pyza, E.; Reiter, R.J.; Steinbrink, K.; Slominski, A.T.; Tulic, M.K.; et al. Mitochondrial Function Is Controlled by Melatonin and Its Metabolites in Vitro in Human Melanoma Cells. J. Pineal Res. 2021, 70, e12728. [Google Scholar] [CrossRef]
  39. Nyamsambuu, A.; Khan, M.A.; Zhou, X.; Chen, H.-C. Molecular Mechanism of Inhibitory Effects of Melatonin on Prostate Cancer Cell Proliferation, Migration and Invasion. PLoS ONE 2022, 17, e0261341. [Google Scholar] [CrossRef]
  40. Park, S.-Y.; Jang, W.-J.; Yi, E.-Y.; Jang, J.-Y.; Jung, Y.; Jeong, J.-W.; Kim, Y.-J. Melatonin Suppresses Tumor Angiogenesis by Inhibiting HIF-1alpha Stabilization under Hypoxia. J. Pineal Res. 2010, 48, 178–184. [Google Scholar] [CrossRef]
  41. Alvarez-García, V.; González, A.; Alonso-González, C.; Martínez-Campa, C.; Cos, S. Regulation of Vascular Endothelial Growth Factor by Melatonin in Human Breast Cancer Cells. J. Pineal Res. 2013, 54, 373–380. [Google Scholar] [CrossRef]
  42. Maschio-Signorini, L.B.; Gelaleti, G.B.; Moschetta, M.G.; Borin, T.F.; Jardim-Perassi, B.V.; Lopes, J.R.; Lacerda, J.Z.; Roela, R.A.; Bordin, N.A.; Corrêa, L.A.; et al. Melatonin Regulates Angiogenic and Inflammatory Proteins in MDA-MB-231 Cell Line and in Co-Culture with Cancer-Associated Fibroblasts. Anticancer Agents Med. Chem. 2016, 16, 1474–1484. [Google Scholar] [CrossRef]
  43. Gonzalez-Haba, M.G.; Garcia-Mauriño, S.; Calvo, J.R.; Goberna, R.; Guerrero, J.M. High-Affinity Binding of Melatonin by Human Circulating T Lymphocytes (CD4+). FASEB J. 1995, 9, 1331–1335. [Google Scholar] [CrossRef]
  44. Plaimee, P.; Khamphio, M.; Weerapreeyakul, N.; Barusrux, S.; Johns, N.P. Immunomodulatory Effect of Melatonin in SK-LU-1 Human Lung Adenocarcinoma Cells Co-Cultured with Peripheral Blood Mononuclear Cells. Cell Prolif. 2014, 47, 406–415. [Google Scholar] [CrossRef]
  45. Kleszczyński, K.; Kim, T.-K.; Bilska, B.; Sarna, M.; Mokrzynski, K.; Stegemann, A.; Pyza, E.; Reiter, R.J.; Steinbrink, K.; Böhm, M.; et al. Melatonin Exerts Oncostatic Capacity and Decreases Melanogenesis in Human MNT-1 Melanoma Cells. J. Pineal Res. 2019, 67, e12610. [Google Scholar] [CrossRef]
  46. Reiter, R.J.; Sharma, R.; Ma, Q.; Rorsales-Corral, S.; de Almeida Chuffa, L.G. Melatonin Inhibits Warburg-Dependent Cancer by Redirecting Glucose Oxidation to the Mitochondria: A Mechanistic Hypothesis. Cell. Mol. Life Sci. 2020, 77, 2527–2542. [Google Scholar] [CrossRef]
  47. Reiter, R.J.; Sharma, R.; Ma, Q. Switching Diseased Cells from Cytosolic Aerobic Glycolysis to Mitochondrial Oxidative Phosphorylation: A Metabolic Rhythm Regulated by Melatonin? J. Pineal Res. 2021, 70, e12677. [Google Scholar] [CrossRef]
  48. Chok, K.C.; Koh, R.Y.; Ng, M.G.; Ng, P.Y.; Chye, S.M. Melatonin Induces Autophagy via Reactive Oxygen Species-Mediated Endoplasmic Reticulum Stress Pathway in Colorectal Cancer Cells. Molecules 2021, 26, 5038. [Google Scholar] [CrossRef]
  49. Kleszczyński, K.; Böhm, M. Can Melatonin and Its Metabolites Boost the Efficacy of Targeted Therapy in Patients with Advanced Melanoma? Exp. Dermatol. 2020, 29, 860–863. [Google Scholar] [CrossRef]
  50. Cheng, X. Structure and Function of DNA Methyltransferases. Annu. Rev. Biophys. Biomol. Struct. 1995, 24, 293–318. [Google Scholar] [CrossRef]
  51. Illingworth, R.; Kerr, A.; Desousa, D.; Jørgensen, H.; Ellis, P.; Stalker, J.; Jackson, D.; Clee, C.; Plumb, R.; Rogers, J.; et al. A Novel CpG Island Set Identifies Tissue-Specific Methylation at Developmental Gene Loci. PLoS Biol. 2008, 6, e22. [Google Scholar] [CrossRef]
  52. Hermann, A.; Gowher, H.; Jeltsch, A. Biochemistry and Biology of Mammalian DNA Methyltransferases. Cell. Mol. Life Sci. 2004, 61, 2571–2587. [Google Scholar] [CrossRef]
  53. Yoder, J.A.; Soman, N.S.; Verdine, G.L.; Bestor, T.H. DNA (Cytosine-5)-Methyltransferases in Mouse Cells and Tissues. Studies with a Mechanism-Based Probe. J. Mol. Biol. 1997, 270, 385–395. [Google Scholar] [CrossRef]
  54. Okano, M.; Bell, D.W.; Haber, D.A.; Li, E. DNA Methyltransferases Dnmt3a and Dnmt3b Are Essential for de Novo Methylation and Mammalian Development. Cell 1999, 99, 247–257. [Google Scholar] [CrossRef]
  55. Vinson, C.; Chatterjee, R. CG Methylation. Epigenomics 2012, 4, 655–663. [Google Scholar] [CrossRef]
  56. Wu, H.; Zhang, Y. Mechanisms and Functions of Tet Protein-Mediated 5-Methylcytosine Oxidation. Genes Dev. 2011, 25, 2436–2452. [Google Scholar] [CrossRef]
  57. Zhu, H.; Wang, G.; Qian, J. Transcription Factors as Readers and Effectors of DNA Methylation. Nat. Rev. Genet. 2016, 17, 551–565. [Google Scholar] [CrossRef]
  58. Hendrich, B.; Bird, A. Identification and Characterization of a Family of Mammalian Methyl-CpG Binding Proteins. Mol. Cell. Biol. 1998, 18, 6538–6547. [Google Scholar] [CrossRef]
  59. Ballestar, E.; Paz, M.F.; Valle, L.; Wei, S.; Fraga, M.F.; Espada, J.; Cruz Cigudosa, J.; Huang, T.H.-M.; Esteller, M. Methyl-CpG Binding Proteins Identify Novel Sites of Epigenetic Inactivation in Human Cancer. EMBO J. 2003, 22, 6335–6345. [Google Scholar] [CrossRef]
  60. Tahiliani, M.; Koh, K.P.; Shen, Y.; Pastor, W.A.; Bandukwala, H.; Brudno, Y.; Agarwal, S.; Iyer, L.M.; Liu, D.R.; Aravind, L.; et al. Conversion of 5-Methylcytosine to 5-Hydroxymethylcytosine in Mammalian DNA by MLL Partner TET1. Science 2009, 324, 930–935. [Google Scholar] [CrossRef]
  61. Ono, R.; Taki, T.; Taketani, T.; Taniwaki, M.; Kobayashi, H.; Hayashi, Y. LCX, Leukemia-Associated Protein with a CXXC Domain, Is Fused to MLL in Acute Myeloid Leukemia with Trilineage Dysplasia Having t(10;11)(Q22;Q23). Cancer Res. 2002, 62, 4075–4080. [Google Scholar]
  62. Lorsbach, R.B.; Moore, J.; Mathew, S.; Raimondi, S.C.; Mukatira, S.T.; Downing, J.R. TET1, a Member of a Novel Protein Family, Is Fused to MLL in Acute Myeloid Leukemia Containing the t(10;11)(Q22;Q23). Leukemia 2003, 17, 637–641. [Google Scholar] [CrossRef]
  63. Ito, S.; Shen, L.; Dai, Q.; Wu, S.C.; Collins, L.B.; Swenberg, J.A.; He, C.; Zhang, Y. Tet Proteins Can Convert 5-Methylcytosine to 5-Formylcytosine and 5-Carboxylcytosine. Science 2011, 333, 1300–1303. [Google Scholar] [CrossRef]
  64. He, Y.-F.; Li, B.-Z.; Li, Z.; Liu, P.; Wang, Y.; Tang, Q.; Ding, J.; Jia, Y.; Chen, Z.; Li, L.; et al. Tet-Mediated Formation of 5-Carboxylcytosine and Its Excision by TDG in Mammalian DNA. Science 2011, 333, 1303–1307. [Google Scholar] [CrossRef]
  65. Maiti, A.; Drohat, A.C. Thymine DNA Glycosylase Can Rapidly Excise 5-Formylcytosine and 5-Carboxylcytosine: Potential Implications for Active Demethylation of CpG Sites. J. Biol. Chem. 2011, 286, 35334–35338. [Google Scholar] [CrossRef]
  66. Neri, F.; Incarnato, D.; Krepelova, A.; Rapelli, S.; Pagnani, A.; Zecchina, R.; Parlato, C.; Oliviero, S. Genome-Wide Analysis Identifies a Functional Association of Tet1 and Polycomb Repressive Complex 2 in Mouse Embryonic Stem Cells. Genome Biol. 2013, 14, R91. [Google Scholar] [CrossRef]
  67. Li, Y.; Zheng, H.; Wang, Q.; Zhou, C.; Wei, L.; Liu, X.; Zhang, W.; Zhang, Y.; Du, Z.; Wang, X.; et al. Genome-Wide Analyses Reveal a Role of Polycomb in Promoting Hypomethylation of DNA Methylation Valleys. Genome Biol. 2018, 19, 18. [Google Scholar] [CrossRef]
  68. Shi, F.; Qiu, J.; Zhang, S.; Zhao, X.; Feng, D.; Feng, X. Exogenous Melatonin Protects Preimplantation Embryo Development from Decabromodiphenyl Ethane-Induced Circadian Rhythm Disorder and Endogenous Melatonin Reduction. Environ. Pollut. 2022, 292, 118445. [Google Scholar] [CrossRef]
  69. Chandru, A.; Bate, N.; Vuister, G.W.; Cowley, S.M. Sin3A Recruits Tet1 to the PAH1 Domain via a Highly Conserved Sin3-Interaction Domain. Sci. Rep. 2018, 8, 14689. [Google Scholar] [CrossRef]
  70. Hrit, J.; Goodrich, L.; Li, C.; Wang, B.-A.; Nie, J.; Cui, X.; Martin, E.A.; Simental, E.; Fernandez, J.; Liu, M.Y.; et al. OGT Binds a Conserved C-Terminal Domain of TET1 to Regulate TET1 Activity and Function in Development. eLife 2018, 7, e34870. [Google Scholar] [CrossRef]
  71. Yuan, J.; Luo, R.Z.; Fujii, S.; Wang, L.; Hu, W.; Andreeff, M.; Pan, Y.; Kadota, M.; Oshimura, M.; Sahin, A.A.; et al. Aberrant Methylation and Silencing of ARHI, an Imprinted Tumor Suppressor Gene in Which the Function Is Lost in Breast Cancers. Cancer Res. 2003, 63, 4174–4180. [Google Scholar] [PubMed]
  72. Han, L.-L.; Hou, L.; Zhou, M.-J.; Ma, Z.; Lin, D.-L.; Wu, L.; Ge, Y. Aberrant NDRG1 Methylation Associated with Its Decreased Expression and Clinicopathological Significance in Breast Cancer. J. Biomed. Sci. 2013, 20, 52. [Google Scholar] [CrossRef]
  73. Xiang, T.-X.; Yuan, Y.; Li, L.-L.; Wang, Z.-H.; Dan, L.-Y.; Chen, Y.; Ren, G.-S.; Tao, Q. Aberrant Promoter CpG Methylation and Its Translational Applications in Breast Cancer. Chin. J. Cancer 2013, 32, 12–20. [Google Scholar] [CrossRef] [PubMed]
  74. Li, J.; Huang, Q.; Zeng, F.; Li, W.; He, Z.; Chen, W.; Zhu, W.; Zhang, B. The Prognostic Value of Global DNA Hypomethylation in Cancer: A Meta-Analysis. PLoS ONE 2014, 9, e106290. [Google Scholar] [CrossRef] [PubMed]
  75. Hinshelwood, R.A.; Clark, S.J. Breast Cancer Epigenetics: Normal Human Mammary Epithelial Cells as a Model System. J. Mol. Med. 2008, 86, 1315–1328. [Google Scholar] [CrossRef]
  76. Yi, J.; Gao, R.; Chen, Y.; Yang, Z.; Han, P.; Zhang, H.; Dou, Y.; Liu, W.; Wang, W.; Du, G.; et al. Overexpression of NSUN2 by DNA Hypomethylation Is Associated with Metastatic Progression in Human Breast Cancer. Oncotarget 2017, 8, 20751–20765. [Google Scholar] [CrossRef]
  77. Soares, J.; Pinto, A.E.; Cunha, C.V.; André, S.; Barão, I.; Sousa, J.M.; Cravo, M. Global DNA Hypomethylation in Breast Carcinoma: Correlation with Prognostic Factors and Tumor Progression. Cancer 1999, 85, 112–118. [Google Scholar] [CrossRef]
  78. Widschwendter, M.; Jiang, G.; Woods, C.; Müller, H.M.; Fiegl, H.; Goebel, G.; Marth, C.; Müller-Holzner, E.; Zeimet, A.G.; Laird, P.W.; et al. DNA Hypomethylation and Ovarian Cancer Biology. Cancer Res. 2004, 64, 4472–4480. [Google Scholar] [CrossRef]
  79. Yegnasubramanian, S.; Haffner, M.C.; Zhang, Y.; Gurel, B.; Cornish, T.C.; Wu, Z.; Irizarry, R.A.; Morgan, J.; Hicks, J.; DeWeese, T.L.; et al. DNA Hypomethylation Arises Later in Prostate Cancer Progression than CpG Island Hypermethylation and Contributes to Metastatic Tumor Heterogeneity. Cancer Res. 2008, 68, 8954–8967. [Google Scholar] [CrossRef]
  80. Barciszewska, A.-M. Global DNA Demethylation as an Epigenetic Marker of Human Brain Metastases. Biosci. Rep. 2018, 38, BSR20180731. [Google Scholar] [CrossRef]
  81. Feinberg, A.P.; Vogelstein, B. Hypomethylation Distinguishes Genes of Some Human Cancers from Their Normal Counterparts. Nature 1983, 301, 89–92. [Google Scholar] [CrossRef] [PubMed]
  82. Gama-Sosa, M.A.; Slagel, V.A.; Trewyn, R.W.; Oxenhandler, R.; Kuo, K.C.; Gehrke, C.W.; Ehrlich, M. The 5-Methylcytosine Content of DNA from Human Tumors. Nucleic Acids Res. 1983, 11, 6883–6894. [Google Scholar] [CrossRef] [PubMed]
  83. Haffner, M.C.; Chaux, A.; Meeker, A.K.; Esopi, D.M.; Gerber, J.; Pellakuru, L.G.; Toubaji, A.; Argani, P.; Iacobuzio-Donahue, C.; Nelson, W.G.; et al. Global 5-Hydroxymethylcytosine Content Is Significantly Reduced in Tissue Stem/Progenitor Cell Compartments and in Human Cancers. Oncotarget 2011, 2, 627–637. [Google Scholar] [CrossRef] [PubMed]
  84. Li, J.; Deng, Q.; Fan, W.; Zeng, Q.; He, H.; Huang, F. Melatonin-Induced Suppression of DNA Methylation Promotes Odontogenic Differentiation in Human Dental Pulp Cells. Bioengineered 2020, 11, 829–840. [Google Scholar] [CrossRef]
  85. Korkmaz, A.; Reiter, R.J. Epigenetic Regulation: A New Research Area for Melatonin? J. Pineal Res. 2008, 44, 41–44. [Google Scholar] [CrossRef] [PubMed]
  86. López-Moyado, I.F.; Tsagaratou, A.; Yuita, H.; Seo, H.; Delatte, B.; Heinz, S.; Benner, C.; Rao, A. Paradoxical Association of TET Loss of Function with Genome-Wide DNA Hypomethylation. Proc. Natl. Acad. Sci. USA 2019, 116, 16933–16942. [Google Scholar] [CrossRef]
  87. Yang, M.; Tao, J.; Wu, H.; Guan, S.; Liu, L.; Zhang, L.; Deng, S.; He, C.; Ji, P.; Liu, J.; et al. Aanat Knockdown and Melatonin Supplementation in Embryo Development: Involvement of Mitochondrial Function and DNA Methylation. Antioxid. Redox Signal. 2019, 30, 2050–2065. [Google Scholar] [CrossRef]
  88. Zeng, J.; Tao, J.; Xia, L.; Zeng, Z.; Chen, J.; Wang, Z.; Meng, J.; Liu, L. Melatonin Inhibits Vascular Endothelial Cell Pyroptosis by Improving Mitochondrial Function via Up-Regulation and Demethylation of UQCRC1. Biochem. Cell Biol. 2021, 99, 339–347. [Google Scholar] [CrossRef]
  89. Hsieh, M.-C.; Ho, Y.-C.; Lai, C.-Y.; Chou, D.; Wang, H.-H.; Chen, G.-D.; Lin, T.-B.; Peng, H.-Y. Melatonin Impedes Tet1-Dependent MGluR5 Promoter Demethylation to Relieve Pain. J. Pineal Res. 2017, 63, e12436. [Google Scholar] [CrossRef]
  90. Wang, X.; Sun, M.; Li, J.; Song, X.; He, H.; Huan, Y. Melatonin Protects against Defects Induced by Enniatin B1 during Porcine Early Embryo Development. Aging 2021, 13, 5553–5570. [Google Scholar] [CrossRef]
  91. Zhao, X.-M.; Wang, N.; Hao, H.-S.; Li, C.-Y.; Zhao, Y.-H.; Yan, C.-L.; Wang, H.-Y.; Du, W.-H.; Wang, D.; Liu, Y.; et al. Melatonin Improves the Fertilization Capacity and Developmental Ability of Bovine Oocytes by Regulating Cytoplasmic Maturation Events. J. Pineal Res. 2018, 64, e12445. [Google Scholar] [CrossRef] [PubMed]
  92. Yang, J.; Bashkenova, N.; Zang, R.; Huang, X.; Wang, J. The Roles of TET Family Proteins in Development and Stem Cells. Development 2020, 147, dev183129. [Google Scholar] [CrossRef] [PubMed]
  93. Putiri, E.L.; Tiedemann, R.L.; Thompson, J.J.; Liu, C.; Ho, T.; Choi, J.-H.; Robertson, K.D. Distinct and Overlapping Control of 5-Methylcytosine and 5-Hydroxymethylcytosine by the TET Proteins in Human Cancer Cells. Genome Biol. 2014, 15, R81. [Google Scholar] [CrossRef] [PubMed]
  94. Liu, Z.; Gan, L.; Zhang, T.; Ren, Q.; Sun, C. Melatonin Alleviates Adipose Inflammation through Elevating α-Ketoglutarate and Diverting Adipose-Derived Exosomes to Macrophages in Mice. J. Pineal Res. 2018, 64, e12455. [Google Scholar] [CrossRef] [PubMed]
  95. Clifton, I.J.; McDonough, M.A.; Ehrismann, D.; Kershaw, N.J.; Granatino, N.; Schofield, C.J. Structural Studies on 2-Oxoglutarate Oxygenases and Related Double-Stranded Beta-Helix Fold Proteins. J. Inorg. Biochem. 2006, 100, 644–669. [Google Scholar] [CrossRef]
  96. McDonough, M.A.; Loenarz, C.; Chowdhury, R.; Clifton, I.J.; Schofield, C.J. Structural Studies on Human 2-Oxoglutarate Dependent Oxygenases. Curr. Opin. Struct. Biol. 2010, 20, 659–672. [Google Scholar] [CrossRef]
  97. Reiter, R.J.; Rosales-Corral, S.; Coto-Montes, A.; Boga, J.A.; Tan, D.-X.; Davis, J.M.; Konturek, P.C.; Konturek, S.J.; Brzozowski, T. The Photoperiod, Circadian Regulation and Chronodisruption: The Requisite Interplay between the Suprachiasmatic Nuclei and the Pineal and Gut Melatonin. J. Physiol. Pharmacol. 2011, 62, 269–274. [Google Scholar]
  98. Cajochen, C.; Münch, M.; Kobialka, S.; Kräuchi, K.; Steiner, R.; Oelhafen, P.; Orgül, S.; Wirz-Justice, A. High Sensitivity of Human Melatonin, Alertness, Thermoregulation, and Heart Rate to Short Wavelength Light. J. Clin. Endocrinol. Metab. 2005, 90, 1311–1316. [Google Scholar] [CrossRef]
  99. Thapan, K.; Arendt, J.; Skene, D.J. An Action Spectrum for Melatonin Suppression: Evidence for a Novel Non-Rod, Non-Cone Photoreceptor System in Humans. J. Physiol. 2001, 535, 261–267. [Google Scholar] [CrossRef]
  100. Walker, W.H.; Bumgarner, J.R.; Walton, J.C.; Liu, J.A.; Meléndez-Fernández, O.H.; Nelson, R.J.; DeVries, A.C. Light Pollution and Cancer. Int. J. Mol. Sci. 2020, 21, 9360. [Google Scholar] [CrossRef]
  101. Schwimmer, H.; Metzer, A.; Pilosof, Y.; Szyf, M.; Machnes, Z.M.; Fares, F.; Harel, O.; Haim, A. Light at Night and Melatonin Have Opposite Effects on Breast Cancer Tumors in Mice Assessed by Growth Rates and Global DNA Methylation. Chronobiol. Int. 2014, 31, 144–150. [Google Scholar] [CrossRef] [PubMed]
  102. Agbaria, S.; Haim, A.; Fares, F.; Zubidat, A.E. Epigenetic Modification in 4T1 Mouse Breast Cancer Model by Artificial Light at Night and Melatonin—The Role of DNA-Methyltransferase. Chronobiol. Int. 2019, 36, 629–643. [Google Scholar] [CrossRef] [PubMed]
  103. Zubidat, A.E.; Fares, B.; Fares, F.; Haim, A. Artificial Light at Night of Different Spectral Compositions Differentially Affects Tumor Growth in Mice: Interaction with Melatonin and Epigenetic Pathways. Cancer Control 2018, 25, 1073274818812908. [Google Scholar] [CrossRef]
  104. Yonis, M.; Haim, A.; Zubidat, A.E. Altered Metabolic and Hormonal Responses in Male Rats Exposed to Acute Bright Light-at-Night Associated with Global DNA Hypo-Methylation. J. Photochem. Photobiol. B 2019, 194, 107–118. [Google Scholar] [CrossRef] [PubMed]
  105. Korkmaz, A.; Sanchez-Barcelo, E.J.; Tan, D.-X.; Reiter, R.J. Role of Melatonin in the Epigenetic Regulation of Breast Cancer. Breast Cancer Res. Treat. 2009, 115, 13–27. [Google Scholar] [CrossRef] [PubMed]
  106. Korkmaz, A.; Topal, T.; Tan, D.-X.; Reiter, R.J. Role of Melatonin in Metabolic Regulation. Rev. Endocr. Metab. Disord. 2009, 10, 261–270. [Google Scholar] [CrossRef]
  107. Kim, M.; Vu, T.-H.; Maas, M.B.; Braun, R.I.; Wolf, M.S.; Roenneberg, T.; Daviglus, M.L.; Reid, K.J.; Zee, P.C. Light at Night in Older Age Is Associated with Obesity, Diabetes, and Hypertension. Sleep 2023, 46, zsac130. [Google Scholar] [CrossRef]
  108. Kim, J.Y.; Jelinek, J.; Lee, Y.H.; Kim, D.H.; Kang, K.; Ryu, S.H.; Moon, H.R.; Cho, K.; Rha, S.H.; Cha, J.K.; et al. Hypomethylation in MTNR1B: A Novel Epigenetic Marker for Atherosclerosis Profiling Using Stenosis Radiophenotype and Blood Inflammatory Cells. Clin. Epigenet. 2023, 15, 11. [Google Scholar] [CrossRef]
  109. Al-Naggar, R.A.; Anil, S. Artificial Light at Night and Cancer: Global Study. Asian Pac. J. Cancer Prev. 2016, 17, 4661–4664. [Google Scholar] [CrossRef]
  110. Gehlert, S.; Clanton, M. Shift Work and Breast Cancer. Int. J. Environ. Res. Public Health 2020, 17, 9544. [Google Scholar] [CrossRef]
  111. Szkiela, M.; Kusideł, E.; Makowiec-Dąbrowska, T.; Kaleta, D. Night Shift Work—A Risk Factor for Breast Cancer. Int. J. Environ. Res. Public Health 2020, 17, 659. [Google Scholar] [CrossRef] [PubMed]
  112. Ritonja, J.A.; Aronson, K.J.; Leung, M.; Flaten, L.; Topouza, D.G.; Duan, Q.L.; Durocher, F.; Tranmer, J.E.; Bhatti, P. Investigating the Relationship between Melatonin Patterns and Methylation in Circadian Genes among Day Shift and Night Shift Workers. Occup. Environ. Med. 2022, 79, 673–680. [Google Scholar] [CrossRef] [PubMed]
  113. Shadan, F.F. Circadian Tempo: A Paradigm for Genome Stability? Med. Hypotheses 2006, 68, 883–891. [Google Scholar] [CrossRef] [PubMed]
  114. Sultan, A.; Parganiha, A.; Sultan, T.; Choudhary, V.; Pati, A.K. Circadian Clock, Cell Cycle, and Breast Cancer: An Updated Review. Biol. Rhythm. Res. 2017, 48, 353–369. [Google Scholar] [CrossRef]
  115. Doi, M.; Hirayama, J.; Sassone-Corsi, P. Circadian Regulator CLOCK Is a Histone Acetyltransferase. Cell 2006, 125, 497–508. [Google Scholar] [CrossRef]
  116. Hirayama, J.; Sahar, S.; Grimaldi, B.; Tamaru, T.; Takamatsu, K.; Nakahata, Y.; Sassone-Corsi, P. CLOCK-Mediated Acetylation of BMAL1 Controls Circadian Function. Nature 2007, 450, 1086–1090. [Google Scholar] [CrossRef]
  117. Huang, C.-H.; Han, W.; Wu, Y.-Z.; Shen, G.-L. Identification of Aberrantly Methylated Differentially Expressed Genes and Pro-Tumorigenic Role of KIF2C in Melanoma. Front. Genet. 2022, 13, 817656. [Google Scholar] [CrossRef]
  118. Liu, S.; Cheng, Y.; Wang, S.; Liu, H. Circadian Clock Genes Modulate Immune, Cell Cycle and Apoptosis in the Diagnosis and Prognosis of Pan-Renal Cell Carcinoma. Front. Mol. Biosci. 2021, 8, 747629. [Google Scholar] [CrossRef]
  119. Van der Watt, P.J.; Roden, L.C.; Davis, K.T.; Parker, M.I.; Leaner, V.D. Circadian Oscillations Persist in Cervical and Esophageal Cancer Cells Displaying Decreased Expression of Tumor-Suppressing Circadian Clock Genes. Mol. Cancer Res. 2020, 18, 1340–1353. [Google Scholar] [CrossRef]
  120. Zhang, Z.; Liang, Z.; Gao, W.; Yu, S.; Hou, Z.; Li, K.; Zeng, P. Identification of Circadian Clock Genes as Regulators of Immune Infiltration in Hepatocellular Carcinoma. J. Cancer 2022, 13, 3199–3208. [Google Scholar] [CrossRef]
  121. Reszka, E.; Wieczorek, E.; Przybek, M.; Jabłońska, E.; Kałużny, P.; Bukowska-Damska, A.; Zienolddiny, S.; Pepłońska, B. Circadian Gene Methylation in Rotating-Shift Nurses: A Cross-Sectional Study. Chronobiol. Int. 2018, 35, 111–121. [Google Scholar] [CrossRef] [PubMed]
  122. Dijk, D.-J.; Archer, S.N. PERIOD3, Circadian Phenotypes, and Sleep Homeostasis. Sleep Med. Rev. 2010, 14, 151–160. [Google Scholar] [CrossRef] [PubMed]
  123. Sulkava, S.; Ollila, H.M.; Alasaari, J.; Puttonen, S.; Härmä, M.; Viitasalo, K.; Lahtinen, A.; Lindström, J.; Toivola, A.; Sulkava, R.; et al. Common Genetic Variation Near Melatonin Receptor 1A Gene Linked to Job-Related Exhaustion in Shift Workers. Sleep 2017, 40, zsw011. [Google Scholar] [CrossRef] [PubMed]
  124. Stevenson, T.J.; Prendergast, B.J. Reversible DNA Methylation Regulates Seasonal Photoperiodic Time Measurement. Proc. Natl. Acad. Sci. USA 2013, 110, 16651–16656. [Google Scholar] [CrossRef] [PubMed]
  125. Lan, M.; Han, J.; Pan, M.-H.; Wan, X.; Pan, Z.-N.; Sun, S.-C. Melatonin Protects against Defects Induced by Deoxynivalenol during Mouse Oocyte Maturation. J. Pineal Res. 2018, 65, e12477. [Google Scholar] [CrossRef]
  126. Qu, J.; Sun, M.; Wang, X.; Song, X.; He, H.; Huan, Y. Melatonin Enhances the Development of Porcine Cloned Embryos by Improving DNA Methylation Reprogramming. Cell Reprogram. 2020, 22, 156–166. [Google Scholar] [CrossRef]
  127. Sørensen, A.L.; Timoskainen, S.; West, F.D.; Vekterud, K.; Boquest, A.C.; Ahrlund-Richter, L.; Stice, S.L.; Collas, P. Lineage-Specific Promoter DNA Methylation Patterns Segregate Adult Progenitor Cell Types. Stem. Cells Dev. 2010, 19, 1257–1266. [Google Scholar] [CrossRef]
  128. Berdasco, M.; Melguizo, C.; Prados, J.; Gómez, A.; Alaminos, M.; Pujana, M.A.; Lopez, M.; Setien, F.; Ortiz, R.; Zafra, I.; et al. DNA Methylation Plasticity of Human Adipose-Derived Stem Cells in Lineage Commitment. Am. J. Pathol. 2012, 181, 2079–2093. [Google Scholar] [CrossRef]
  129. Gibney, E.R.; Nolan, C.M. Epigenetics and Gene Expression. Heredity 2010, 105, 4–13. [Google Scholar] [CrossRef]
  130. Rahman, M.M.; Brane, A.C.; Tollefsbol, T.O. MicroRNAs and Epigenetics Strategies to Reverse Breast Cancer. Cells 2019, 8, 1214. [Google Scholar] [CrossRef]
  131. Tompkins, J.D.; Hall, C.; Chen, V.C.; Li, A.X.; Wu, X.; Hsu, D.; Couture, L.A.; Riggs, A.D. Epigenetic Stability, Adaptability, and Reversibility in Human Embryonic Stem Cells. Proc. Natl. Acad. Sci. USA 2012, 109, 12544–12549. [Google Scholar] [CrossRef] [PubMed]
  132. Kaur, J.; Daoud, A.; Eblen, S.T. Targeting Chromatin Remodeling for Cancer Therapy. Curr. Mol. Pharmacol. 2019, 12, 215–229. [Google Scholar] [CrossRef] [PubMed]
  133. Kim, A.; Mo, K.; Kwon, H.; Choe, S.; Park, M.; Kwak, W.; Yoon, H. Epigenetic Regulation in Breast Cancer: Insights on Epidrugs. Epigenomes 2023, 7, 6. [Google Scholar] [CrossRef] [PubMed]
  134. Jones, P.A.; Taylor, S.M. Cellular Differentiation, Cytidine Analogs and DNA Methylation. Cell 1980, 20, 85–93. [Google Scholar] [CrossRef] [PubMed]
  135. Stresemann, C.; Lyko, F. Modes of Action of the DNA Methyltransferase Inhibitors Azacytidine and Decitabine. Int. J. Cancer 2008, 123, 8–13. [Google Scholar] [CrossRef] [PubMed]
  136. Schermelleh, L.; Spada, F.; Easwaran, H.P.; Zolghadr, K.; Margot, J.B.; Cardoso, M.C.; Leonhardt, H. Trapped in Action: Direct Visualization of DNA Methyltransferase Activity in Living Cells. Nat. Methods 2005, 2, 751–756. [Google Scholar] [CrossRef]
  137. Gnyszka, A.; Jastrzebski, Z.; Flis, S. DNA Methyltransferase Inhibitors and Their Emerging Role in Epigenetic Therapy of Cancer. Anticancer Res. 2013, 33, 2989–2996. [Google Scholar]
  138. Fenaux, P.; Mufti, G.J.; Hellstrom-Lindberg, E.; Santini, V.; Finelli, C.; Giagounidis, A.; Schoch, R.; Gattermann, N.; Sanz, G.; List, A.; et al. Efficacy of Azacitidine Compared with That of Conventional Care Regimens in the Treatment of Higher-Risk Myelodysplastic Syndromes: A Randomised, Open-Label, Phase III Study. Lancet Oncol. 2009, 10, 223–232. [Google Scholar] [CrossRef]
  139. Seymour, J.F.; Döhner, H.; Butrym, A.; Wierzbowska, A.; Selleslag, D.; Jang, J.H.; Kumar, R.; Cavenagh, J.; Schuh, A.C.; Candoni, A.; et al. Azacitidine Improves Clinical Outcomes in Older Patients with Acute Myeloid Leukaemia with Myelodysplasia-Related Changes Compared with Conventional Care Regimens. BMC Cancer 2017, 17, 852. [Google Scholar] [CrossRef]
  140. Platzbecker, U.; Middeke, J.M.; Sockel, K.; Herbst, R.; Wolf, D.; Baldus, C.D.; Oelschlägel, U.; Mütherig, A.; Fransecky, L.; Noppeney, R.; et al. Measurable Residual Disease-Guided Treatment with Azacitidine to Prevent Haematological Relapse in Patients with Myelodysplastic Syndrome and Acute Myeloid Leukaemia (RELAZA2): An Open-Label, Multicentre, Phase 2 Trial. Lancet Oncol. 2018, 19, 1668–1679. [Google Scholar] [CrossRef]
  141. Seymour, J.F.; Fenaux, P.; Silverman, L.R.; Mufti, G.J.; Hellström-Lindberg, E.; Santini, V.; List, A.F.; Gore, S.D.; Backstrom, J.; McKenzie, D.; et al. Effects of Azacitidine Compared with Conventional Care Regimens in Elderly (≥75 Years) Patients with Higher-Risk Myelodysplastic Syndromes. Crit. Rev. Oncol. Hematol. 2010, 76, 218–227. [Google Scholar] [CrossRef] [PubMed]
  142. Fenaux, P.; Mufti, G.J.; Hellström-Lindberg, E.; Santini, V.; Gattermann, N.; Germing, U.; Sanz, G.; List, A.F.; Gore, S.; Seymour, J.F.; et al. Azacitidine Prolongs Overall Survival Compared with Conventional Care Regimens in Elderly Patients with Low Bone Marrow Blast Count Acute Myeloid Leukemia. J. Clin. Oncol. 2010, 28, 562–569. [Google Scholar] [CrossRef] [PubMed]
  143. Kantarjian, H.M.; O’Brien, S.; Shan, J.; Aribi, A.; Garcia-Manero, G.; Jabbour, E.; Ravandi, F.; Cortes, J.; Davisson, J.; Issa, J.-P. Update of the Decitabine Experience in Higher Risk Myelodysplastic Syndrome and Analysis of Prognostic Factors Associated with Outcome. Cancer 2007, 109, 265–273. [Google Scholar] [CrossRef] [PubMed]
  144. Jabbour, E.; Garcia-Manero, G.; Ravandi, F.; Faderl, S.; O’Brien, S.; Fullmer, A.; Cortes, J.E.; Wierda, W.; Kantarjian, H. Prognostic Factors Associated with Disease Progression and Overall Survival in Patients with Myelodysplastic Syndromes Treated with Decitabine. Clin. Lymphoma Myeloma Leuk. 2013, 13, 131–138. [Google Scholar] [CrossRef]
  145. Ramos, M.-P.; Wijetunga, N.A.; McLellan, A.S.; Suzuki, M.; Greally, J.M. DNA Demethylation by 5-Aza-2′-Deoxycytidine Is Imprinted, Targeted to Euchromatin, and Has Limited Transcriptional Consequences. Epigenetics Chromatin 2015, 8, 11. [Google Scholar] [CrossRef]
  146. Giri, A.K.; Aittokallio, T. DNMT Inhibitors Increase Methylation in the Cancer Genome. Front. Pharmacol. 2019, 10, 385. [Google Scholar] [CrossRef]
  147. Cabezón, M.; Malinverni, R.; Bargay, J.; Xicoy, B.; Marcé, S.; Garrido, A.; Tormo, M.; Arenillas, L.; Coll, R.; Borras, J.; et al. Different Methylation Signatures at Diagnosis in Patients with High-Risk Myelodysplastic Syndromes and Secondary Acute Myeloid Leukemia Predict Azacitidine Response and Longer Survival. Clin. Epigenet. 2021, 13, 9. [Google Scholar] [CrossRef]
  148. Notari, R.E.; DeYoung, J.L. Kinetics and Mechanisms of Degradation of the Antileukemic Agent 5-Azacytidine in Aqueous Solutions. J. Pharm Sci. 1975, 64, 1148–1157. [Google Scholar] [CrossRef]
  149. Rogstad, D.K.; Herring, J.L.; Theruvathu, J.A.; Burdzy, A.; Perry, C.C.; Neidigh, J.W.; Sowers, L.C. The Chemical Decomposition of 5-Aza-2′-Deoxycytidine (Decitabine): Kinetic Analyses and Identification of Products by NMR, HPLC, and Mass Spectrometry. Chem. Res. Toxicol. 2009, 22, 1194–1204. [Google Scholar] [CrossRef]
  150. Agrawal, K.; Das, V.; Vyas, P.; Hajdúch, M. Nucleosidic DNA Demethylating Epigenetic Drugs—A Comprehensive Review from Discovery to Clinic. Pharmacol. Ther. 2018, 188, 45–79. [Google Scholar] [CrossRef]
  151. Issa, J.-P.J.; Garcia-Manero, G.; Giles, F.J.; Mannari, R.; Thomas, D.; Faderl, S.; Bayar, E.; Lyons, J.; Rosenfeld, C.S.; Cortes, J.; et al. Phase 1 Study of Low-Dose Prolonged Exposure Schedules of the Hypomethylating Agent 5-Aza-2′-Deoxycytidine (Decitabine) in Hematopoietic Malignancies. Blood 2004, 103, 1635–1640. [Google Scholar] [CrossRef] [PubMed]
  152. Schnekenburger, M.; Grandjenette, C.; Ghelfi, J.; Karius, T.; Foliguet, B.; Dicato, M.; Diederich, M. Sustained Exposure to the DNA Demethylating Agent, 2′-Deoxy-5-Azacytidine, Leads to Apoptotic Cell Death in Chronic Myeloid Leukemia by Promoting Differentiation, Senescence, and Autophagy. Biochem. Pharmacol. 2011, 81, 364–378. [Google Scholar] [CrossRef] [PubMed]
  153. Hollenbach, P.W.; Nguyen, A.N.; Brady, H.; Williams, M.; Ning, Y.; Richard, N.; Krushel, L.; Aukerman, S.L.; Heise, C.; MacBeth, K.J. A Comparison of Azacitidine and Decitabine Activities in Acute Myeloid Leukemia Cell Lines. PLoS ONE 2010, 5, e9001. [Google Scholar] [CrossRef] [PubMed]
  154. Zhao, M.; Rudek, M.A.; He, P.; Hartke, C.; Gore, S.; Carducci, M.A.; Baker, S.D. Quantification of 5-Azacytidine in Plasma by Electrospray Tandem Mass Spectrometry Coupled with High-Performance Liquid Chromatography. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2004, 813, 81–88. [Google Scholar] [CrossRef]
  155. Liu, Z.; Marcucci, G.; Byrd, J.C.; Grever, M.; Xiao, J.; Chan, K.K. Characterization of Decomposition Products and Preclinical and Low Dose Clinical Pharmacokinetics of Decitabine (5-Aza-2′-Deoxycytidine) by a New Liquid Chromatography/Tandem Mass Spectrometry Quantification Method. Rapid Commun. Mass Spectrom. 2006, 20, 1117–1126. [Google Scholar] [CrossRef]
  156. Gros, C.; Fahy, J.; Halby, L.; Dufau, I.; Erdmann, A.; Gregoire, J.-M.; Ausseil, F.; Vispé, S.; Arimondo, P.B. DNA Methylation Inhibitors in Cancer: Recent and Future Approaches. Biochimie 2012, 94, 2280–2296. [Google Scholar] [CrossRef]
  157. Bohl, S.R.; Bullinger, L.; Rücker, F.G. Epigenetic Therapy: Azacytidine and Decitabine in Acute Myeloid Leukemia. Expert Rev. Hematol. 2018, 11, 361–371. [Google Scholar] [CrossRef]
  158. Hu, C.; Liu, X.; Zeng, Y.; Liu, J.; Wu, F. DNA Methyltransferase Inhibitors Combination Therapy for the Treatment of Solid Tumor: Mechanism and Clinical Application. Clin. Epigenet. 2021, 13, 166. [Google Scholar] [CrossRef]
  159. Seely, D.; Wu, P.; Fritz, H.; Kennedy, D.A.; Tsui, T.; Seely, A.J.E.; Mills, E. Melatonin as Adjuvant Cancer Care with and without Chemotherapy: A Systematic Review and Meta-Analysis of Randomized Trials. Integr. Cancer Ther. 2012, 11, 293–303. [Google Scholar] [CrossRef]
  160. Pariente, R.; Bejarano, I.; Rodríguez, A.B.; Pariente, J.A.; Espino, J. Melatonin Increases the Effect of 5-Fluorouracil-Based Chemotherapy in Human Colorectal Adenocarcinoma Cells in Vitro. Mol. Cell Biochem. 2018, 440, 43–51. [Google Scholar] [CrossRef]
  161. Mihanfar, A.; Yousefi, B.; Ghazizadeh Darband, S.; Sadighparvar, S.; Kaviani, M.; Majidinia, M. Melatonin Increases 5-Flurouracil-Mediated Apoptosis of Colorectal Cancer Cells through Enhancing Oxidative Stress and Downregulating Survivin and XIAP. Bioimpacts 2021, 11, 253–261. [Google Scholar] [CrossRef] [PubMed]
  162. Gao, Y.; Xiao, X.; Zhang, C.; Yu, W.; Guo, W.; Zhang, Z.; Li, Z.; Feng, X.; Hao, J.; Zhang, K.; et al. Melatonin Synergizes the Chemotherapeutic Effect of 5-Fluorouracil in Colon Cancer by Suppressing PI3K/AKT and NF-ΚB/INOS Signaling Pathways. J. Pineal Res. 2017, 62, e12380. [Google Scholar] [CrossRef] [PubMed]
  163. Liu, D.; Ma, Z.; Di, S.; Yang, Y.; Yang, J.; Xu, L.; Reiter, R.J.; Qiao, S.; Yuan, J. AMPK/PGC1α Activation by Melatonin Attenuates Acute Doxorubicin Cardiotoxicity via Alleviating Mitochondrial Oxidative Damage and Apoptosis. Free Radic. Biol. Med. 2018, 129, 59–72. [Google Scholar] [CrossRef]
  164. Niu, G.; Yousefi, B.; Qujeq, D.; Marjani, A.; Asadi, J.; Wang, Z.; Mir, S.M. Melatonin and Doxorubicin Co-Delivered via a Functionalized Graphene-Dendrimeric System Enhances Apoptosis of Osteosarcoma Cells. Mater. Sci. Eng. C Mater. Biol. Appl. 2021, 119, 111554. [Google Scholar] [CrossRef] [PubMed]
  165. Erdogan, C.S.; Al Hassadi, Y.; Aru, B.; Yılmaz, B.; Gemici, B. Combinatorial Effects of Melatonin and Paclitaxel Differ Depending on the Treatment Scheme in Colorectal Cancer in Vitro. Life Sci. 2022, 308, 120927. [Google Scholar] [CrossRef] [PubMed]
  166. Rohilla, S.; Singh, M.; Priya, S.; Almalki, W.H.; Haniffa, S.M.; Subramaniyan, V.; Fuloria, S.; Fuloria, N.K.; Sekar, M.; Singh, S.K.; et al. Exploring the Mechanical Perspective of a New Anti-Tumor Agent: Melatonin. J. Environ. Pathol. Toxicol. Oncol. 2023, 42, 1–16. [Google Scholar] [CrossRef] [PubMed]
  167. Pathipaka, R.; Thyagarajan, A.; Sahu, R.P. Melatonin as a Repurposed Drug for Melanoma Treatment. Med. Sci. 2023, 11, 9. [Google Scholar] [CrossRef] [PubMed]
  168. Hartung, E.E.; Mukhtar, S.Z.; Shah, S.M.; Niles, L.P. 5-Azacytidine Upregulates Melatonin MT1 Receptor Expression in Rat C6 Glioma Cells: Oncostatic Implications. Mol. Biol. Rep. 2020, 47, 4867–4873. [Google Scholar] [CrossRef]
  169. Nakamura, E.; Kozaki, K.; Tsuda, H.; Suzuki, E.; Pimkhaokham, A.; Yamamoto, G.; Irie, T.; Tachikawa, T.; Amagasa, T.; Inazawa, J.; et al. Frequent Silencing of a Putative Tumor Suppressor Gene Melatonin Receptor 1 A (MTNR1A) in Oral Squamous-Cell Carcinoma. Cancer Sci. 2008, 99, 1390–1400. [Google Scholar] [CrossRef]
  170. Oprea-Ilies, G.; Haus, E.; Sackett-Lundeen, L.; Liu, Y.; McLendon, L.; Busch, R.; Adams, A.; Cohen, C. Expression of Melatonin Receptors in Triple Negative Breast Cancer (TNBC) in African American and Caucasian Women: Relation to Survival. Breast Cancer Res. Treat. 2013, 137, 677–687. [Google Scholar] [CrossRef]
  171. Goyal, R.; Gupta, T.; Bal, A.; Sahni, D.; Singh, G. Role of Melatonin in Breast Carcinoma: Correlation of Expression Patterns of Melatonin-1 Receptor with Estrogen, Progesterone, and HER2 Receptors. Appl. Immunohistochem. Mol. Morphol. 2020, 28, 518–523. [Google Scholar] [CrossRef] [PubMed]
  172. Dauchy, R.T.; Blask, D.E.; Dauchy, E.M.; Davidson, L.K.; Tirrell, P.C.; Greene, M.W.; Tirrell, R.P.; Hill, C.R.; Sauer, L.A. Antineoplastic Effects of Melatonin on a Rare Malignancy of Mesenchymal Origin: Melatonin Receptor-Mediated Inhibition of Signal Transduction, Linoleic Acid Metabolism and Growth in Tissue-Isolated Human Leiomyosarcoma Xenografts. J. Pineal Res. 2009, 47, 32–42. [Google Scholar] [CrossRef] [PubMed]
  173. Xiang, S.; Dauchy, R.T.; Hoffman, A.E.; Pointer, D.; Frasch, T.; Blask, D.E.; Hill, S.M. Epigenetic Inhibition of the Tumor Suppressor ARHI by Light at Night-Induced Circadian Melatonin Disruption Mediates STAT3-Driven Paclitaxel Resistance in Breast Cancer. J. Pineal Res. 2019, 67, e12586. [Google Scholar] [CrossRef] [PubMed]
  174. Hill, S.M.; Cheng, C.; Yuan, L.; Mao, L.; Jockers, R.; Dauchy, B.; Blask, D.E. Age-Related Decline in Melatonin and Its MT1 Receptor Are Associated with Decreased Sensitivity to Melatonin and Enhanced Mammary Tumor Growth. Curr. Aging Sci. 2013, 6, 125–133. [Google Scholar] [CrossRef]
  175. Lesicka, M.; Dmitrzak-Weglarz, M.; Jablonska, E.; Wieczorek, E.; Kapelski, P.; Szczepankiewicz, A.; Pawlak, J.; Reszka, E. Methylation of Melatonin Receptors in Patients with Unipolar and Bipolar Depression. Mech. Ageing Dev. 2023, 211, 111776. [Google Scholar] [CrossRef]
  176. Lee, C.-C.; Kuo, Y.-C.; Hu, J.-M.; Chang, P.-K.; Sun, C.-A.; Yang, T.; Li, C.-W.; Chen, C.-Y.; Lin, F.-H.; Hsu, C.-H.; et al. MTNR1B Polymorphisms with CDKN2A and MGMT Methylation Status Are Associated with Poor Prognosis of Colorectal Cancer in Taiwan. World J. Gastroenterol. 2021, 27, 5737–5752. [Google Scholar] [CrossRef]
  177. Lee, S.E.; Kim, S.J.; Yoon, H.-J.; Yu, S.Y.; Yang, H.; Jeong, S.I.; Hwang, S.Y.; Park, C.-S.; Park, Y.S. Genome-Wide Profiling in Melatonin-Exposed Human Breast Cancer Cell Lines Identifies Differentially Methylated Genes Involved in the Anticancer Effect of Melatonin. J. Pineal Res. 2013, 54, 80–88. [Google Scholar] [CrossRef]
  178. Martín, V.; Sanchez-Sanchez, A.M.; Herrera, F.; Gomez-Manzano, C.; Fueyo, J.; Alvarez-Vega, M.A.; Antolín, I.; Rodriguez, C. Melatonin-Induced Methylation of the ABCG2/BCRP Promoter as a Novel Mechanism to Overcome Multidrug Resistance in Brain Tumour Stem Cells. Br. J. Cancer 2013, 108, 2005–2012. [Google Scholar] [CrossRef]
  179. Nie, J.; Xiao, P.; Wang, X.; Yang, X.; Xu, H.; Lu, K.; Lu, S.; Liang, X. Melatonin Prevents Deterioration in Quality by Preserving Epigenetic Modifications of Porcine Oocytes after Prolonged Culture. Aging 2018, 10, 3897–3909. [Google Scholar] [CrossRef]
  180. Ritonja, J.A.; Aronson, K.J.; Flaten, L.; Topouza, D.G.; Duan, Q.L.; Durocher, F.; Tranmer, J.E.; Bhatti, P. Exploring the Impact of Night Shift Work on Methylation of Circadian Genes. Epigenetics 2022, 17, 1259–1268. [Google Scholar] [CrossRef]
  181. Villicaña, S.; Bell, J.T. Genetic Impacts on DNA Methylation: Research Findings and Future Perspectives. Genome Biol. 2021, 22, 127. [Google Scholar] [CrossRef] [PubMed]
  182. Miyakuni, K.; Nishida, J.; Koinuma, D.; Nagae, G.; Aburatani, H.; Miyazono, K.; Ehata, S. Genome-Wide Analysis of DNA Methylation Identifies the Apoptosis-Related Gene UQCRH as a Tumor Suppressor in Renal Cancer. Mol. Oncol. 2022, 16, 732–749. [Google Scholar] [CrossRef]
  183. Ding, K.; Jiang, J.; Chen, L.; Xu, X. Methylenetetrahydrofolate Dehydrogenase 1 Silencing Expedites the Apoptosis of Non-Small Cell Lung Cancer Cells via Modulating DNA Methylation. Med. Sci. Monit. 2018, 24, 7499–7507. [Google Scholar] [CrossRef] [PubMed]
  184. Dos Santos, P.W.d.S.; Machado, A.R.T.; De Grandis, R.A.; Ribeiro, D.L.; Tuttis, K.; Morselli, M.; Aissa, A.F.; Pellegrini, M.; Antunes, L.M.G. Transcriptome and DNA Methylation Changes Modulated by Sulforaphane Induce Cell Cycle Arrest, Apoptosis, DNA Damage, and Suppression of Proliferation in Human Liver Cancer Cells. Food Chem. Toxicol. 2020, 136, 111047. [Google Scholar] [CrossRef]
  185. Bernardini, L.; Barbosa, E.; Charão, M.F.; Goethel, G.; Muller, D.; Bau, C.; Steffens, N.A.; Santos Stein, C.; Moresco, R.N.; Garcia, S.C.; et al. Oxidative Damage, Inflammation, Genotoxic Effect, and Global DNA Methylation Caused by Inhalation of Formaldehyde and the Purpose of Melatonin. Toxicol. Res. 2020, 9, 778–789. [Google Scholar] [CrossRef]
  186. Fang, Y.; Zhang, J.; Li, Y.; Guo, X.; Li, J.; Zhong, R.; Zhang, X. Melatonin-Induced Demethylation of Antioxidant Genes Increases Antioxidant Capacity through RORα in Cumulus Cells of Prepubertal Lambs. Free Radic Biol. Med. 2019, 131, 173–183. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Melatonin synthesis pathway. Abbreviations: TPH: tryptophan hydroxylase; AAD: amino acid decarboxylase; AANAT: aralkylamine N-acetyltransferase; HIOMT: hydroxyindole-O-methyltransferase; NAS: N-acetyloserotonin.
Figure 1. Melatonin synthesis pathway. Abbreviations: TPH: tryptophan hydroxylase; AAD: amino acid decarboxylase; AANAT: aralkylamine N-acetyltransferase; HIOMT: hydroxyindole-O-methyltransferase; NAS: N-acetyloserotonin.
Antioxidants 12 01155 g001
Figure 2. Melatonin stimulates cells of the immune system, NK cells, monocytes, leukocytes, interleukins, and interferon gamma to invoke immune responses triggering organism defense from cancerous cells.
Figure 2. Melatonin stimulates cells of the immune system, NK cells, monocytes, leukocytes, interleukins, and interferon gamma to invoke immune responses triggering organism defense from cancerous cells.
Antioxidants 12 01155 g002
Figure 3. DNA methylation processes. Unmethylated DNA can undergo de novo methylation process via DNMT3A and DNMT3B. During the replication process, methylation marks are restored by DNMT1 through maintenance DNA methylation process. Abbreviations: DNMT1, DNA methyltransferase 1; DNMT3A, DNA methyltransferase 3A; DNMT3B, DNA methyltransferase 3B.
Figure 3. DNA methylation processes. Unmethylated DNA can undergo de novo methylation process via DNMT3A and DNMT3B. During the replication process, methylation marks are restored by DNMT1 through maintenance DNA methylation process. Abbreviations: DNMT1, DNA methyltransferase 1; DNMT3A, DNA methyltransferase 3A; DNMT3B, DNA methyltransferase 3B.
Antioxidants 12 01155 g003
Figure 4. Active DNA demethylation process. 5-methylcytosine is transformed into 5-hydroxymethylcytosine, which is further hydroxylated into 5-formylcytosine and 5-carboxylcytosine. Modified cytosines (5-formylcytosine and 5-carboxylcytosine) are recognized and excised by thymine DNA glycosylase (TDG), a key enzyme belonging to base excision repair (BER) machinery. Abbreviations: BER machinery, base excision repair machinery; DNMT, DNA methyltransferase; TET, ten-eleven translocation proteins; TDG, thymine DNA glycosylase.
Figure 4. Active DNA demethylation process. 5-methylcytosine is transformed into 5-hydroxymethylcytosine, which is further hydroxylated into 5-formylcytosine and 5-carboxylcytosine. Modified cytosines (5-formylcytosine and 5-carboxylcytosine) are recognized and excised by thymine DNA glycosylase (TDG), a key enzyme belonging to base excision repair (BER) machinery. Abbreviations: BER machinery, base excision repair machinery; DNMT, DNA methyltransferase; TET, ten-eleven translocation proteins; TDG, thymine DNA glycosylase.
Antioxidants 12 01155 g004
Figure 5. Possible impact of melatonin on DNA methylation and demethylation processes. Melatonin can impact TET proteins via the regulation of α-ketoglutarate level, thus promoting active DNA demethylation. Moreover, melatonin may also regulate DNMT, resulting in DNA methylation inhibition. Abbreviations: DNMT, DNA methyltransferase; TET, ten-eleven translocation proteins; 5-mC, 5-methylcytosine; 5-hmC, 5-hydroxymethylcytosine; α-KG, α-ketoglutarate.
Figure 5. Possible impact of melatonin on DNA methylation and demethylation processes. Melatonin can impact TET proteins via the regulation of α-ketoglutarate level, thus promoting active DNA demethylation. Moreover, melatonin may also regulate DNMT, resulting in DNA methylation inhibition. Abbreviations: DNMT, DNA methyltransferase; TET, ten-eleven translocation proteins; 5-mC, 5-methylcytosine; 5-hmC, 5-hydroxymethylcytosine; α-KG, α-ketoglutarate.
Antioxidants 12 01155 g005
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

Linowiecka, K.; Slominski, A.T.; Reiter, R.J.; Böhm, M.; Steinbrink, K.; Paus, R.; Kleszczyński, K. Melatonin: A Potential Regulator of DNA Methylation. Antioxidants 2023, 12, 1155. https://doi.org/10.3390/antiox12061155

AMA Style

Linowiecka K, Slominski AT, Reiter RJ, Böhm M, Steinbrink K, Paus R, Kleszczyński K. Melatonin: A Potential Regulator of DNA Methylation. Antioxidants. 2023; 12(6):1155. https://doi.org/10.3390/antiox12061155

Chicago/Turabian Style

Linowiecka, Kinga, Andrzej T. Slominski, Russel J. Reiter, Markus Böhm, Kerstin Steinbrink, Ralf Paus, and Konrad Kleszczyński. 2023. "Melatonin: A Potential Regulator of DNA Methylation" Antioxidants 12, no. 6: 1155. https://doi.org/10.3390/antiox12061155

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