Volume 61, Issue 3 p. 253-278
Review Article
Free Access

Melatonin as an antioxidant: under promises but over delivers

Russel J. Reiter

Corresponding Author

Russel J. Reiter

Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA

Correspondence

Russel J. Reiter, Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA.

Email: [email protected]

Search for more papers by this author
Juan C. Mayo

Juan C. Mayo

Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA

Search for more papers by this author
Dun-Xian Tan

Dun-Xian Tan

Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA

Search for more papers by this author
Rosa M. Sainz

Rosa M. Sainz

Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA

Search for more papers by this author
Moises Alatorre-Jimenez

Moises Alatorre-Jimenez

Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA

Search for more papers by this author
Lilan Qin

Lilan Qin

Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA

Search for more papers by this author
First published: 08 August 2016
Citations: 1,064

Abstract

Melatonin is uncommonly effective in reducing oxidative stress under a remarkably large number of circumstances. It achieves this action via a variety of means: direct detoxification of reactive oxygen and reactive nitrogen species and indirectly by stimulating antioxidant enzymes while suppressing the activity of pro-oxidant enzymes. In addition to these well-described actions, melatonin also reportedly chelates transition metals, which are involved in the Fenton/Haber–Weiss reactions; in doing so, melatonin reduces the formation of the devastatingly toxic hydroxyl radical resulting in the reduction of oxidative stress. Melatonin's ubiquitous but unequal intracellular distribution, including its high concentrations in mitochondria, likely aid in its capacity to resist oxidative stress and cellular apoptosis. There is credible evidence to suggest that melatonin should be classified as a mitochondria-targeted antioxidant. Melatonin's capacity to prevent oxidative damage and the associated physiological debilitation is well documented in numerous experimental ischemia/reperfusion (hypoxia/reoxygenation) studies especially in the brain (stroke) and in the heart (heart attack). Melatonin, via its antiradical mechanisms, also reduces the toxicity of noxious prescription drugs and of methamphetamine, a drug of abuse. Experimental findings also indicate that melatonin renders treatment-resistant cancers sensitive to various therapeutic agents and may be useful, due to its multiple antioxidant actions, in especially delaying and perhaps treating a variety of age-related diseases and dehumanizing conditions. Melatonin has been effectively used to combat oxidative stress, inflammation and cellular apoptosis and to restore tissue function in a number of human trials; its efficacy supports its more extensive use in a wider variety of human studies. The uncommonly high-safety profile of melatonin also bolsters this conclusion. It is the current feeling of the authors that, in view of the widely diverse beneficial functions that have been reported for melatonin, these may be merely epiphenomena of the more fundamental, yet-to-be identified basic action(s) of this ancient molecule.

1 Introduction: Melatonin and the Evolution of Ideas

The ubiquitous distribution and functional diversity of melatonin as currently envisioned far exceeds that of original expectations.1 In particular, due to definitive studies within the last 15 years, melatonin has been linked to a wide range of functions including anti-inflammation, antioxidant, oncostatic, circadian rhythm regulation, etc. Likely prompted by early observations made on pinealectomized animals2-4 prior to the discovery of melatonin,5, 6 this indoleamine was initially tested as to its effects on reproduction.7-9 This led to the discovery that the changing photoperiod, with the pineal gland and the melatonin rhythm as intermediates, unequivocally regulates seasonal reproductive capability in photosensitive species.10-12

Seasonal breeding is of utmost importance because it ensures delivery of the young during the most propitious season, thereby improving the chances of survival of the young and the continuation of the species. That the introduction of artificial light at night, which is commonplace in the outdoor environment in economically developed regions, negatively impacts this essential annual cycle was recently reported in a publication from Australia. In a 5-year study on the seasonal reproductive behavior of tammar wallabies living in different photoperiodic environments, it became apparent that ambient light pollution disturbs this cycle.13 The comparisons were made on wallabies living in a forested area without artificial light at night vs wallabies inhabiting a nearby urban area where anthropogenic light was rampant. In the wallabies witnessing light pollution at night, their circulating melatonin levels were suppressed and the birth of the young was delayed, that is, occurred at an unusual time of the year. These results were entirely predictable given that light at night is well known to interfere with the amplitude and the duration of the nocturnal melatonin signal,14, 15 which dictates the annual reproductive cycle of seasonal breeding mammals.10, 16 The finding also illustrates the dangers of exposing seasonally breeding wild animals to light pollution. This probably is becoming more common, since a number of wild species cohabitate very successfully with residents of urban communities and light pollution is becoming more widespread. The reproductive cycle may be only one of several metabolic disturbances the wallabies and other species experience when exposed to artificial light at night. For example, it may be increasing the frequency of some types of cancer and reducing the total antioxidative capacity of vertebrates.

Even before melatonin was identified, light microscopic observations on the pineal gland showed that its morphology depends on the light:dark environment to which the animals are exposed.17, 18 Both of these authors noted that the cytological changes in the pinealocytes during exposure of the animals to short days were consistent with presumed elevated synthetic activity. This portended that the physiology of the pineal gland may be impacted by the photoperiod. Whether these microscopic observations were, however, used as a justification for studies related to the dark-dependent synthesis of melatonin was not explicitly stated in the introduction to the reports where pineal melatonin production was documented to be confined to the dark phase of the light:dark cycle (Fig. 1).19-21

Details are in the caption following the image
The nocturnal rise in the synthesis and release by pineal melatonin drives the circadian rhythms of melatonin in blood and cerebrospinal fluid (CSF). During the day, the central circadian oscillator (the biological clock in the suprachiasmatic nuclei, i.e., the SCN) receives neural messages from highly specialized intrinsically photoreceptive retinal ganglion cells (ipRGC) via the retinohypothalamic pathway in the optic nerves, which prevent the SCN from signaling the pineal gland to gear up melatonin production. When the inhibitory signal from the ipRGC (which are especially sensitive to blue wavelengths of visible light) is lifted at night, the SCN contacts the pineal gland via a multisynaptic pathway in the central and peripheral sympathetic nervous system, which is relayed through the superior cervical ganglia (SCN), to upregulate the melatonin synthetic machinery. The pineal releases melatonin directly into the third ventricular CSF where it generates a larger amplitude melatonin rhythm (not shown in this figure) than exists in the blood; it is the CSF rhythm, in our opinion, which modulates the activity of the SCN rather than the melatonin cycle in the blood. Melatonin released into the blood accesses every cell in the organism and presumably influences the circadian genes in these cells. Additionally, pineal-derived melatonin has numerous functions (some of which are tabulated in the box at the lower right) in multiple organs, where it protects critical molecules from pathophysiology. In addition to melatonin of pineal origin, melatonin produced in many (perhaps all) cells has similar beneficial actions in the cells where it is synthesized (autocrine actions) and in neighboring cells (paracrine actions)

Initially, the rate limiting enzyme in melatonin synthesis was deduced to be hydroxyindole-O-methyltransferase (HIOMT), the enzyme currently known as N-acetylserotonin methyltransferase (ASMT).22-24 However, after the discovery of the marked circadian rhythm in the activity of N-acetyltransferase (NAT), the enzyme that acetylates serotonin to form N-acetylserotonin,25 interest in this step as being the determinant of maximal melatonin production peaked. While this assumption generally persists until today, there are some who have argued otherwise.26 More recently, we have raised the issue as to whether the sequence of enzymatic events that convert serotonin to melatonin is always correct.27 In some plant species at least, we feel that serotonin may be first methylated to form 5-methoxytryptamine followed by its acetylation to melatonin. Moreover, in plants, melatonin may not be the terminal product but rather an intermediate that is subsequently hydroxylated to 2-hydroxymelatonin.28

The key studies to confirm the essential nature of the sympathetic nerve terminals in the pineal as being important for its biochemistry and physiology23, 29 were driven by the meticulous morphological studies of Kappers;30 he showed that the pineal received a rich postganglionic sympathetic neural input from perikarya located in the superior cervical ganglia. Subsequently, surgical removal of the ganglia was shown to biochemically and functionally incapacitate the pineal gland.23, 29 The loss of melatonin due to surgical removal of either the pineal gland or superior cervical ganglia eliminates all known functions of the pineal gland. Yet, in a 1965 report, it was claimed that melatonin synthesis (as judged by the elevated HIOMT activity) was actually increased after superior cervical ganglionectomy.31 This error may have been made since, when the superior cervical ganglia are surgically removed, the degenerating nerve terminals in the pineal gland release stored norepinephrine that causes a transient rise in pineal synthetic activity.32

While melatonin, subsequent to its discovery, was never denied as being a pineal secretory product, it was not always promoted as the major pineal secretion. Rather than melatonin, a number of peptides extracted from pineal tissue, most of which were never structurally identified, were advanced as being the responsible agents for mediating the gland's action on the pituitary–gonadal axis.33-36 So far as the authors know, there is only a single group that still believes there are regulatory peptides of any type of pineal origin.37, 38 The release of these peptides from the pineal gland has never been verified, an obvious requirement if they are to have actions throughout the organism. If these mythological peptides do exist, they have never been tested for the diversity of functions exhibited by melatonin, for example, as antioxidants, although in one report the authors claimed that the pineal peptide, epithalamin, provides better protection against free radical damage than does melatonin.38 This seems doubtful, and this observation requires confirmation in an independent laboratory.

Because melatonin was initially discovered in the pineal gland, it was often surmised that this molecule would be unique to the pineal tissue of vertebrates, given that they are the only species that has this organ. This, however, was not found to be the case. Within less than a decade after the characterization of melatonin, the melatonin-forming enzyme (HIOMT) was also uncovered in the retina by Quay21, 39 and slightly later by Cardinali and Rosner.40 The presence of melatonin in the vertebrate retina perhaps should not have been a major surprise, as the epithalamus of some nonmammalian vertebrates has a structure reminiscent of the retina of the lateral eyes.41 This structure has been referred to as the third eye (or parietal eye). In some extinct quadrupeds, the third eye in the epithalamus was believed to be organized like a retina and capable of light perception followed by the transmission of action potentials to the central nervous system; however, this extra eye was probably not capable of forming images. Retinal melatonin of the lateral eyes exhibits a circadian rhythm like that in the pineal gland, but the melatonin-forming cells in the eyes of mammals do not discharge this product into the blood. The retinal melatonin rhythm influences dopamine metabolism and the function of retinal clocks,42 presumably much like the melatonin cycle in the cerebrospinal fluid (CSF) modulates the superchiasmatic nucleus (SCN).43-45 The pineal gland of mammals and the third eye from which it evolved (or related structures) have other common features.46, 47

In addition to the pineal gland and retinas, numerous vertebrate organs produce melatonin. This is perhaps best exemplified by its production throughout much of the gastrointestinal system,48, 49 where its synthesis is not known to exhibit a daily cycle.50 The one non-neural organ that may display a 24-hour rhythm of melatonin is the Harderian gland,51 an intraorbital exocrine gland that is found in only some mammals (but not in the human); the function of these large organs remains a mystery.52, 53 There is a disagreement whether the melatonin cycle in the Harderian glands is disrupted by pinealectomy or changes in response to the light:dark cycle.54 As with other peripheral organs, the Harderian glands do not contribute melatonin to the systemic circulation.

The concentrations of melatonin in the serum of vertebrates have always been described as being uncommonly low (in the pg/mL range), even during darkness when the values rarely exceed 200 pg/mL. A recent revelation by Dauchy et al.55 suggests that these values, measured in animals and humans maintained under what is considered low intensity artificial light (which is not similar either in intensity or wavelength to sunlight) during the day and relative darkness at night, may not represent the true melatonin cycle in reference to its nocturnal amplitude. In the study in question, these workers found that when male and female pigmented nude rats were maintained in blue-tinted polycarbonate cages, the nocturnal melatonin peak was up to seven times higher at night compared to that in rats housed in clear polycarbonate cages (Fig. 2); rather than serum melatonin values rising to 150 pg/mL at night, peak concentrations rose to 1000 pg/mL serum in rats kept in blue-tinted cages. Thus, the spectral content of daytime light [in this case light enriched with blue wavelengths (450–495 nmol/L)] profoundly impacted the amplitude of the peak serum melatonin on subsequent nights. Although not examined, this exaggerated rise was likely related to increased synthesis and release of melatonin by the pineal gland and was presumably associated with a proportionally elevated rise of melatonin in other bodily fluids, for example, CSF, and possibly also in somatic cells.

Details are in the caption following the image
Twenty-four hour plasma melatonin rhythms in pigmented nude male rats maintained under a 12:12 light:dark cycle (darkness from 18:00 to 06:00 h) for 6 wk. Animals were kept in either clear polycarbonate cages (red dots and lines) or polycarbonate blue-tinted cages (blue squares and lines). The lighting conditions in the room were identical for both groups of rats (light intensity 300 lux, 125 μW/cm2) with absolute darkness at night. The average nighttime increase in plasma melatonin in rats kept in clear cages was 9.6-fold over daytime levels while, for animals kept in blue-tinted cages, the average nighttime rise was 55.3-fold. Clearly, rats that experienced light during the day that was enriched with blue wavelengths (450–495 nmol/L) had a much greater nighttime rise in plasma melatonin levels. The supposition is that pineal melatonin synthesis and release is enhanced during darkness if the animals witness blue-enriched light during the day. Less likely would be that the blue light during the day slowed nocturnal hepatic melatonin metabolism. Red and blue * signify significant differences. Data are double-plotted for clarity. Data points are means ±1 SD. From Dauchy et al.55 with permission

These findings are of particular interest since exposure to blue-enriched light during the day leads to an obvious enhancement of maximal nighttime melatonin concentrations, yet it is also blue light exposure at night that is maximally inhibitory to circulating melatonin concentrations.56 The latter response is primarily mediated by the intrinsically photosensitive retinal ganglion cells (ipRGC), which contain a unique photopigment, melanopsin, that is especially sensitive to blue light wavelengths.57, 58 Whether blue wavelengths of light detected by ipRGC during the day have a function in relation to the exaggerated melatonin peak on the subsequent nights should be examined.

There is another implication of the findings reported by Dauchy et al.55 This group has a history of publishing eloquent studies on the inhibitory actions of physiological concentrations (1 nmol/L) of melatonin on tumor growth.59-61 They have also shown that the tumor suppressive actions of melatonin are mediated by the well-characterized MT1 membrane melatonin receptor.61 The Kd of this receptor is consistent with 1 nmol/L melatonin concentration to which it is routinely exposed.62 If the nighttime rise in melatonin is substantially greater than originally believed, the newly described melatonin concentration could exceed the Kd of the receptor, possibly leading to its downregulation and rendering it incapable of mediating an inhibitory response in terms of cancer inhibition. That was not the case, however, since the higher melatonin values actually had a greater inhibitory effect on prostate tumor growth.55 Whether the highly elevated circulating melatonin levels described by Dauchy et al. will be found to influence the function of the MT1 and MT2 membrane receptors awaits further experimentation. Considering the very wide differences in melatonin concentrations in different body fluids (blood levels vs those in the CSF63 and bile64), receptors in different locations are normally exposed to markedly different levels of the indoleamine.

The data related to the markedly elevated nocturnal circulating melatonin concentrations in rats kept under blue-enriched daytime light55 have implications beyond inhibition of tumor growth. The ability of melatonin to reduce oxidative stress via its free radical scavenging actions is directly related to its concentration. At higher concentrations, there are more molecules of the antioxidant available to quench free radicals thereby lowering oxidative damage and related diseases. Induction of a lifelong melatonin deficit due to pinealectomy leads to an elevation in the amount of oxidative tissue damage in late life relative to the amount of molecular damage seen in pineal-intact rats with a preserved melatonin rhythm.65 A corollary of this is that the higher concentrations of melatonin, as observed by Dauchy et al.55 would also be expected to reduce oxidative damage to a greater degree than the commonly accepted lower amplitude rhythm.

Melatonin production is not limited to vertebrates but is also present in all organisms examined including bacteria,66 unicells,67 invertebrates,68, 69 and vascular plants.70-72 None of these species has a pineal gland and some consist of only a single cell. Considering this, melatonin's association with the pineal gland of vertebrates may be only coincidental and perhaps was necessitated by the fact that, in order for the gland to produce melatonin in a circadian manner dependent on the light:dark cycle, it had to be regulated by neural information from organs responsible for light perception, that is, the lateral eyes. Although not examined in most nonvertebrate species, a 24-hour rhythm of melatonin has been described in the dinoflagellate Gonyaulax polyedra,73 and in some plants,74 at least one of the melatonin's functions is preserved in all species where it has been found, for example, its ability to detoxify free radicals.75-78

2 Melatonin as an Antioxidant: Waging War on Free Radicals

2.1 Preparing for battle: melatonin's physiological weapons

The direct free radical scavenging activity of melatonin has been known for almost 25 years.79 A subsequent report of this process also identified a novel melatonin metabolite, cyclic-3-hydroxymelatonin (c3OHM), which is formed when melatonin scavenges two free radicals; in this report, we also deduced the pathway by which c3OHM is formed.80 This discovery was followed shortly by a series of studies, conducted both in vitro81, 82 and in vivo,83-87 which documented the ability of melatonin to quell oxidative damage to molecules, cells and tissues, including human cells.88, 89 Since then, there have been numerous reports confirming the ability of melatonin to directly scavenge oxygen-centered radicals and toxic reactive oxygen species (ROS)90-95 and to diminish oxidative mutilation to key cellular macromolecules.96-102 These direct free radical scavenging actions of melatonin and its metabolites have been summarized in a number of reviews,103-105 so they will not be discussed in detail here.

Other developments in the mid-1990s further advanced melatonin as an effective countermeasure to oxidative insults. Within 2 years after its discovery as a direct free radical scavenger, melatonin was found to stimulate antioxidative enzymes including glutathione peroxidase and glutathione reductase.106-111 Furthermore, melatonin upregulates the synthesis of glutathione,112-114 a highly effective intrinsic antioxidant, and synergizes with classic free radical scavengers to improve the reductive potential of tissues and fluids.115, 116 These indirect antioxidant functions of melatonin further leveraged this molecule as being a key endogenous factor in limiting free radical damage. Finally, melatonin was found to neutralize nitrogen-based toxicants, that is, nitric oxide and the peroxynitrite anion, both of which promote nitrosative damage,117, 118 and to suppress the pro-oxidative enzyme, nitric oxide synthase.119, 120 When the total antioxidant capacity of human blood was compared to both day and night endogenous melatonin concentrations, these parameters were found to be positively correlated (Fig. 3).121 This correlation documents that not only pharmacological levels of melatonin, but likewise physiological concentrations, likely provide protection against damaging free radicals.122

Details are in the caption following the image
The day and nighttime levels of melatonin in the blood correlated with the total antioxidant status (TAS) of this fluid throughout the life of humans. As individuals age, the nighttime melatonin levels wane; likewise, the TSA concentrations drop accordingly. The findings indicate that physiological levels melatonin in the blood enhance the free radical scavenging potential of that medium. From Benot et al.121 with permission

As pointed out above, when melatonin functions as a scavenger, one resulting product is the metabolite c3OHM. When tested for its antioxidant capacity, c3OHM proved also to function in radical detoxification123, 124 as do its downstream metabolites, N-acetyl-N-formyl-5-methoxykynuramine (AFMK) and N-acetyl-5-methoxykynuramine (AMK),125-130 in what has been defined as melatonin's antioxidant cascade.126 Hence, the first-, second-, and third-generation metabolites of melatonin all have proven to be excellent radical scavengers.131, 132 This cascade predictably allows melatonin to neutralize up to 10 radical products, which contrasts with classic free radical scavengers, which detoxify a single oxidizing molecule. As melatonin is present in plants,51, 52 its function is also that of a radical scavenger in these organisms.133-137

Another potentially important action that is often overlooked as being relevant to melatonin's capacity to quench oxidative damage is its ability to bind heavy metals. In 1998, using absorptive voltammetry as a means of assessment, Limson et al.138 reported that melatonin binds aluminum, cadmium, copper, iron, lead, and zinc not unlike metallothionein. The interaction of melatonin with these metals was found to be concentration dependent. Melatonin chelates both iron (III) and iron (II), which is the form that participates in the Fenton reaction to generate the hydroxyl radical. If the iron is bound to a protein, for example, hemoglobin, melatonin restores the highly covalent iron such as oxyferryl (FeIV-O) hemoglobin back to the iron (III) thereby re-establishing the biological activity of the protein. This would be similar to the reducing action of melatonin when it encounters the highly toxic hydroxyl radical. Particularly in the brain, metallothionein plays a less important role regarding its binding of transition metals. Because it is a protein, any bond metallothionein forms with a transition metal would be damaged by the free radical the metal would generate. By comparison, melatonin would neutralize the generated free radical and reduce the damage. This may be especially important in the brain where, as noted, metallothionein has a reduced role in binding metals. We have recently discussed the possibility that the high levels of melatonin in the CSF, relative to the concentration in the blood, may afford the brain extra protection from oxidative stress.43, 63, 139 In the brain, melatonin, in addition to its direct scavenging activity and indirect antioxidant actions, may have replaced or supplemented metallothionein as a major binder of transition metals.

Parmar et al.140 perused these original observations by investigating melatonin's ability to reduce copper-mediated lipid peroxidation in hepatic homogenates. In this study, melatonin may well have reduced lipid damage by directly scavenging radicals sufficiently toxic to initiate lipid peroxidation; additionally, however, electrochemical studies found that melatonin bound both Cu(II) and Cu(I). These actions likely conspired to reduce the oxidation of hepatic lipids. Soon after the report by Parmar et al.,140 Mayo et al.141 showed that protein damage resulting from exposure to Cu(II)/H2O2 was alleviated by melatonin while Gulcin et al.,142 in a comparative investigation, found that melatonin had a higher Fe(II) chelating activity of this ion than either α-tocopherol or the synthetic antioxidants, butylated hydroxybutylanisole or butylated hydroxytoluene. Melatonin also markedly reduced the interaction of Al(III), Zn(II), Cu(II), Mn(II), and Fe(II) with amyloid β-peptide.143

In the most recent study related to the metal-chelating activity of melatonin, Galano et al.144 examined the copper sequestering ability of melatonin as well as that of its metabolites c3OHM, AFMK, and AMK. This group pointed out that while copper is essential for optimal cell physiology, when it is in high concentrations, it participates in the Fenton/Haber–Weiss reactions, which generate the hydroxyl radical. Also, a deficiency of copper compromises antioxidant defense processes due to a reduction in the synthesis of the cytosolic antioxidant enzyme, copper superoxide dismutase (CuSOD). Moreover, several neurological diseases including Alzheimer's disease,145, 146 Parkinson's disease,147, 148 Huntington's disease,149 and hepatolenticular degeneration (Wilson disease)150, 151 are characterized by an overload of copper and/or other metals. Molecular damage associated with some of these conditions is likely a result of the pro-oxidative actions of an excess of copper ions. Considering this, it is important to regulate the levels of copper consistent with cellular needs. When the cooper-chelating ability of melatonin, c3OHM, AFMK, and AMK were compared in the framework of the density function theory, we reported that melatonin as well as its metabolites yielded stable complexes when they bond copper ions (Fig. 4).144 Two mechanisms were modeled; these were the direct-chelation mechanism (DCM) and the coupled-deprotonation-chelation mechanism (CDCM). Under physiological conditions, it was predicted that the CDCM was the major route of Cu(II) chelation. Melatonin, as well as its metabolites chelated Cu(II) and completely inhibited oxidative stress induced in a Cu(II)/ascorbate mixture. Similarly, melatonin, c3OHM, and AFMK prevented the initial step in the Haber–Weiss reaction consequently reducing the formation of the highly oxidizing hydroxyl radical. On the basis of these findings, Galano et al.144 proposed that melatonin, besides being the initial molecule in the free radical scavenging casade,152 is also involved in a metal-chelating cascade as summarized in Fig. 4. A review related to the metal-catalyzed molecular damage that occurs in organisms where the ability of melatonin to chelate these damaging ions may be consequential was recently published.153

Details are in the caption following the image
The free radical scavenging cascade (vertical) and the metal-chelating cascade (horizontal) of melatonin and its metabolites. The chelation structures shown on the right side of the figure are those that are predicted to be the most abundant. The most likely mechanism for the formation of the predicted complexes is a coupled-deprotonation-chelation mechanism (CDCM). From Galano et al.144 with permission

In addition to the means already discussed, a variety of other factors probably aid melatonin in reducing the total oxidative burden of an organism. As summarized in Fig. 5, melatonin not only neutralizes a host of toxic reactive molecules, but also modulates the activities of a wide variety of enzymes that determine the quantity of ROS/RNS produced. Moreover, there are physiological and metabolic factors that probably contribute to the high efficacy of melatonin, as well as its metabolites, in reducing oxidative damage. For example, melatonin reportedly limits electron leakage from the mitochondrial respiratory chain leading to fewer oxygen molecules being reduced to the superoxide anion radical; this process is referred to as radical avoidance.154 Melatonin's anti-inflammatory actions indirectly reduce free radical damage given that the inflammatory response typically is accompanied by free radical generation155 while the ability of melatonin to strengthen circadian rhythms also aids in fewer oxidative processes since chronodisruption enhances the production of oxidizing molecules.156

Details are in the caption following the image
This figure summarizes the multiple actions of melatonin in reducing oxidative stress. The red area indicates the reactive oxygen (ROS) and reactive nitrogen species (RNS) that have been shown to be neutralized by melatonin and metabolites that are formed during its antioxidant cascade. The blue area identifies enzymes that impact the redox state of the cell because they either cause the generation of radicals or metabolized them to inactive products. The former are upregulated while the latter are downregulated by melatonin and/or its metabolites. Glutamyl cysteine ligase induces the formation of glutathione, an important intracellular antioxidant. The black areas list features that aid melatonin in terms of its ability to quench free radicals and reduce oxidative damage

2.2 Melatonin, a mitochondria-targeted antioxidant

Mitochondria are specifically designed for certain critical functions including the generation of ATP; in normal aerobic cells, oxidative phosphorylation accounts for the efficient production of 95% of the total ATP generated. While performing this essential task, mitochondria also are a major site for the production of oxygen-based toxic species, that is, ROS,157, 158 the majority of which must be detoxified before they irreparably damage these organelles and severely compromise ATP production. Indeed, a major theory of aging, that is, the mitochondrial theory, incriminates damage to these organelles as being responsible for the aging of cells, of organs and of organisms.159-161 As oxidative damage of mitochondria is central to a number of serious pathologies and to aging, conventional antioxidants should be useful in forestalling these diseases or delaying degenerative processes associated with advanced age. Yet, the evidence is remarkably sparse regarding the successful application of regularly used antioxidants to influence the progression of the diseases or aging.162-166

One reason for the failure of conventional antioxidants to ameliorate the severity of ROS-related diseases may be a result of their inability to concentrate in mitochondria where free radical production is maximal. Thus, it seemed like a worthwhile strategy would be to develop mitochondria-targeted antioxidants; this has been done and they were shown effective in reducing mitochondrial damage and the resulting apoptosis.167-169 As an example, to achieve a high concentration in mitochondria, the ubiquinone moiety of endogenous co-enzyme Q10 was conjugated with the lipophilic triphenyl phosphonium cation (TPP).170 Combining TPP with Q10 allowed the resulting molecule, called MitoQ, to rapidly cross the cell and mitochondrial membranes and to accumulate in concentrations up to several hundred-fold greater in the mitochondrial matrix than that of the unconjugated antioxidant (Fig. 6). Tocopherol (vitamin E) also has been conjugated with TPP with a similar degree of success in terms of targeting it to the mitochondrial matrix; this complex is identified as MitoE.170 Both MitoQ and MitoE achieve improved protection of mitochondria against free radical damage over that provided by the unconjugated forms of the antioxidants.171, 172, 168, 173, 174 Both MitoQ and MitoE are recycled in the mitochondrial matrix thereby increasing their efficacy in minimizing local molecular damage.175

Details are in the caption following the image
The structures of MitoE and MitoQ, mitochondria-targeted antioxidants. Melatonin is an endogenously produced molecule that, based on its relative ability to protect against inflammation and oxidative stress when compared to MitoQ and MitoE, may be capable of accumulating in the mitochondria. MitoQ and MitoE are synthetic mitochondria-targeted antioxidants. They are produced when the ubiquinone moiety of Q10 and tocopherol, respectively, is conjugated with triphenyl phosphonium cation. MitoE and MitoQ accumulate in high concentrations in the mitochondria. Melatonin is as effective and in some cases more effective than MitoE and MitoQ in reducing oxidative damage and inflammation (See Fig. 7)

Lowes et al.176 compared the relative efficacies of the two mitochondria-targeted antioxidants, MitoQ and MitoE, with melatonin in reducing inflammation and oxidative damage. A worst case scenario was used to create the molecular carnage. Adult male rats were given both bacteria lipopolysaccharide (LPS) and peptidoglycan (PepG) via a tail vein infusion to induce massive oxidative damage. Shortly, thereafter, the animals received, via the same route, either MitoQ, MitoE or melatonin. Five hours later, plasma and tissue samples were collected. The authors described broadly equivalent protective actions of the three antioxidants relative to their improvement in maintaining mitochondrial respiration, reducing oxidative damage and depressing pro-inflammatory cytokine levels. Additionally, each of the antioxidants had roughly similar protective effects in preserving biochemical parameters of organ physiology as plasma levels of alanine transaminase and creatinine did not differ statistically among the three antioxidant-treated groups. The data relative to the hepatic protein carbonyls and oxidized lipids are summarized in Fig. 7. From the data in this figure, it seems apparent that the most effective antioxidant related to these parameters was melatonin given the lower mean values of damage molecules and their more uniform inhibition in the animals of this group.

Details are in the caption following the image
Concentration of hepatic protein carbonyls (A) and plasma lipid hydroperoxides (B) in placebo-treated control rats and in animals given toxic bacterial lipopolysaccharide (LPS) and peptidoglycan (PepG) to induce oxidative damage. Some LPS + PepG-treated rats were also infused with the synthetic mitochondria targeted antioxidants, MitoE or MitoQ, or with the endogenously produced antioxidant melatonin. Each of these antioxidants significantly reduced oxidatively damaged hepatic proteins and plasma lipids, with melatonin seemingly being the most effective. #P-values are relative to the LPS + PepG control group. Redrawn and with approval from Lowes et al.176

The combination of LPS + PepG is a very aggressive challenge to the defensive makeup of mammals and in this study melatonin handled the attack as well as or better than the synthetic mitochondria-targeted antioxidants.176 A major implication of these findings is that melatonin should be classified as an endogenous mitochondria-targeted antioxidant (Fig. 6). This would be consistent with the much higher melatonin levels in hepatic mitochondria than in the plasma as reported177, 178 and with the proposal that mitochondria might be the site of intracellular melatonin synthesis.179 Positioning itself in mitochondria may be a critical aspect of melatonin's potent antioxidant activity. Maintaining the reductive potential of these organelles is important as mitochondria are often the major site of massive free radical generation. In view of the Surviving Sepsis Campaign, a program to identify agents that can counteract the rampant damage that occurs during sepsis and septic shock,180 melatonin may prove to be a critical component of a treatment paradigm. Of the three antioxidants used, at the conclusion of their report, Lowes et al.176 state that melatonin may be the most accessible agent to resist the molecular damage and mortality that occurs in septic humans.

2.3 Melatonin as an antioxidant: evidence from ischemia-reperfusion studies

There are a large number of published reports confirming that melatonin overcomes oxidative destruction of key molecules and death of cells in tissues that are temporarily deprived of oxygenated blood and then reperfused with blood rich in oxygen. During both hypoxia (ischemia) and reoxygenation (reperfusion), cataclysmic levels of ROS/RNS are generated that molest essential molecules leading to the accumulation of molecular debris that compromises both the function and the survival of cells.181, 182 Melatonin's relentless quest to curb such damage stems in part from its antioxidant potential has been documented during ischemia/reperfusion (IR) of many organs (Fig. 8).

Details are in the caption following the image
A diagrammatic representation of some of the consequences of hypoxia/reoxygenation (ischemia/reperfusion) as they occur in the brain during a stroke or in the heart during a heart attack. Similar changes occur in any organ that experiences hypoxia/reoxygenation. Massive quantities of reactive oxygen (ROS) and reactive nitrogen species are generated during both hypoxia and reperfusion. These toxic agents initiate the release of previously sequestered calcium (Ca2+) into the cytosol and damage mitochondria, which allows the escape of cytochrome c (Cyt c). Released Cyt c activates the apoptotic cascade. Hypoxia/reoxygenation is also associated with an inflammatory response that involves the release of NF-kB and its translocation into the nucleus. This activates the synthesis of chemokines and cytokines which results in the augmentation of ROS production. Melatonin has multiple actions by which it abates the damage inflicted by ROS; these actions include direct free radical scavenging, stimulation of antioxidant enzymes and chelation of transition metals. As a result of these actions, melatonin attenuates cellular apoptosis and tissue loss, thereby partially preserving the function of the damaged organs

While any IR event is always serious, when it involves the brain (stroke) or the heart (heart attack), it is especially critical and often life threatening. In those individuals who do survive a stroke or a heart attack, the neurobehavioral or physiological consequences are often debilitating and persistent and compromise life quality. Identifying molecules that can prevent or significantly reduce the damage caused by episodes of IR are a major interest of the scientific community.183, 184

Table 1 summarizes a few of the numerous studies in which melatonin has been effectively used to combat IR damage in the brain and in the heart. As seen in the table, the most common rodent model used to temporarily interrupt the blood supply to a focal area of the brain is middle cerebral artery occlusion (MCAO) with the usual doses of melatonin used to counter the associated neural damage being 4–10 mg/kgbody weight (BW). This model is of interest since it is representative of the localized stroke that humans often experience.

Table 1. A summary of the results of some of the published reports (there are many more), which illustrate the beneficial effects of melatonin in experimental and clinical ischemia/reperfusion injury (stroke) in the brain and in the heart (heart attack). The majority of studies were performed using rodents as the experimental models
Reference Species Type/duration ischemia Melatonin dose
Brain, animal
Guerrero et al.185 Gerbil 10 min bilateral common carotid clamp 10 mg/kg BW
Kilic et al.186 Rat 120 min MCAO 4 mg/kg BW pinealectomy
Kilic et al.187 Mouse 90 min MCAO 4 mg/kg BW
Kilic et al.188 Mouse 90 min MCAO 4 mg/kg BW
Carloni et al.189 Newborn rat Permanent right common carotid ligation 15 mg/kg BW
Li et al.190 Rat 120 min MCAO 5 mg/kg BW
Zheng et al.191 Rat 90 min MCAO 5 or 10 mg/kg BW
Paredes et al.192 2, 6, 14 mon old rats Permanent MCAO 10 mg/kg BW
Brain, human
Fulia et al.193 Newborn During difficult vaginal delivery 80 mg total (first 6 h after birth)
Aly et al.194 Newborn Hypoxic ischemic encephalopathy 50 mg total (5 × 10 mg) + hypothermia
Heart, animal
Tan et al.195 Rat heart ex vivo 10 min ligation of left anterior descending artery Perfused with 1, 10 or 50 μmol/L
Petrosillo et al.196 Rat heart ex vivo 30 min global ischemia Perfused with 50 μmol/L
Liu et al.197 Rat 10 min ligation of left coronary artery 2.5, 5 or 10 mg/kg
Yu et al.198 Rat 30 min ligation of left anterior descending coronary artery 10 mg/kg/7d15 mg/kg
He et al.199 Mouse 30 min ligation of left coronary artery 150 mg/kg
Nduhirabandi et al.200 Rat heart ex vivo 30 min global ischemia Perfused with 75 μg/L
Heart, human
Gogener et al.201 Adult Ischemia during abdominal aortic aneurism repair Perfused with 50 mg for 2 h; 10 mg/3 d after surgery
Dwaich et al.202 Adult Coronary artery bypass surgery Oral 10 or 20 mg daily for 5 d
  • MCAO, middle cerebral artery occlusion.

The most recent and certainly the most captivating study, although not registered with ClinicalTrials.gov, related to the use of melatonin to overcome the perturbed heart function associated with transitory ischemia and reperfusion is that of the Dwaich et al.202 When they gave either 10 or 20 mg of melatonin orally for 5 days before coronary bypass surgery to male and female patients (15 individuals per treatment group), the physiological dividend reaped from this treatment was substantial. Twenty-four hours following surgery, there was a significant increase in the cardiac ejection fraction (measured using echocardiography) accompanied with a reduction in heart rate (relative to 15 surgical patients not treated with melatonin). Moreover, there were significant reductions in plasma cardiac troponin 1, interleukin-1β, inducible nitric oxide synthase (iNOS), and caspase 3 due to melatonin treatment. The improvements were greater in the patients who were given 20 mg of melatonin compared with those given 10 mg of the indole; thus, the effects were dose dependent. The results of Dwiach et al.202 showed that melatonin treatment attenuated myocardial injury (as measured by the ejection fraction and troponin 1), limited the inflammatory response (IL-1β), decreased oxidative stress (iNOS), and arrested the degree of apoptosis (caspase 3). As the responses measured varied with the dose of melatonin given, higher doses of the indole or its administration via another route (e.g., infusion during surgery) may further improve cardiac parameters. Hopefully, such studies are being pursued.

A wide variety of endpoints ranging from infarct volume to molecular markers of cellular damage have been measured in the IR studies to prove the value of melatonin in suppressing brain damage that results from oxygen deprivation followed by oxygen restoration. The majority of the studies concluded that a significant portion of the protective effects of melatonin related either to its direct scavenging actions or to its indirect functions in promoting other free radical neutralizing activities. One report noted that blocking the MT1 and MT2 membrane receptors, which are widely distributed in the brain,203 did not interfere with melatonin's ability to douse cellular damage.186 That does not exclude the possibility, however, that the MT3 (quinone reductase, a cytosolic detoxifying enzyme204) or nuclear binding sites (ROR, RZR205) did not mediate some of the neuroprotective actions of melatonin.

The number of human studies related to hypoxia and melatonin use is obviously limited. Fulia et al.193 were the first to show that giving 80 mg of melatonin (eight doses of 10 mg each) during the first 6 hours after birth to asphyxiated newborns (due to difficult birth) reduced circulating levels of oxidized lipids and nitrite/nitrate concentrations and decreased mortality (three of 10 asphyxiated newborns not given melatonin died while zero of 10 melatonin-treated asphyxiated infants succumbed). While this is not direct evidence that melatonin protected the brain from the period of hypoxia, this organ is especially sensitive to oxygen deprivation206 and melatonin readily crosses the blood–brain barrier;207 so it can be safely assumed that the exogenously administered melatonin relieved the brain of some of the redox imbalance it suffered due to the hypoxia (Fig. 8).

The report by Aly et al.194 speaks more directly to the neuroprotective actions of melatonin in human neonates. In this prospective study, 5-day hypothermia combined with enteral melatonin treatment reduced numerous oxidative parameters in newborns suffering with hypoxic ischemic encephalopathy (HIE). The neurological endpoints included fewer seizures in the hypothermic melatonin-treated infants and less white matter damage as visualized using magnetic resonance imaging. Finally, the combined treatment was efficacious in terms of improving survival and causing favorable neurodevelopmental outcomes at a month after birth.194

The use of melatonin to protect the heart from ST-segment elevation myocardial infarction (STEMI) has been a major interest to the group of Dominguez-Rodriguez et al.208-210 The safety and efficacy of melatonin as an antioxidant and the participation of free radicals in mediating cardiac damage in STEMI patients were the basis for the design and rationale of the MARIA trial.211 This group212-214 and others215, 216 have published summaries of literature reports that have used melatonin to overcome heart damage from ROS/RNS, whatever the cause.

The neurological damage resulting from a stroke or heart stoppage leaves in its wake a variety of physiological, neurobehavioral, and cognitive residuals that lead to physical and mental debilitation. To limit these devastating conditions, several damaging processes must be targeted including oxidative/nitrosative stress, inflammation, and glutamate excitotoxicity.217 Each of these processes are modulated by melatonin. As discussed herein, melatonin is among the best molecules available in terms of fighting against the molecular carnage inflicted by oxygen- and nitrogen-based toxic reactants. Furthermore, its anti-inflammatory actions are well described and mechanistically defined,218, 219 and glutamate toxicity, which involves destructive free radicals, is negated by melatonin.220, 221 When measured in experimental studies, the severity of the long-term neurobehavioral deficits associated with stroke have been also shown to be reduced when melatonin was given coincident with the IR episode.222, 223 Many of the molecular details that are involved in melatonin's protective actions during IR have been elucidated in recent reports.224, 190, 225-227

Herein, emphasis was placed on the neuroprotective and cardioprotective actions of melatonin that result from IR as hypoxia/reoxygenation in these organs often has dire consequences. These are, however, not the only organs where melatonin has preserved the morphological and functional integrity when they are subjected to IR. Published reports have shown that the lung,228-230 liver,231-233 kidney,234 pancreas,235 intestine,236 urinary bladder,237, 238 corpus cavernosum,239 skeletal muscle240, 241 spinal cord,242, 243 and stem cells244 are also protected by melatonin. It would be expected that if a molecule limits IR damage in one organ, it would do so in all, as was shown to be the case for melatonin. Stem cells in culture sometimes suffer from periods of hypoxia and this also happens when they are implanted. That an endogenously produced, nontoxic molecule protects them from damage and death may prove to be of great importance given that stem cell transplantation is increasing in frequency.

2.4 Melatonin as an antioxidant: evidence from organ transplantation studies

Organ transplantation is a valuable procedure for individuals suffering with end-stage organ failure. Among a number of factors that compromise success of transplanted organs are immune intolerance and apoptotic/necrotic cell death due to hypoxia/reoxygenation.245, 246 In reference to this latter point, the information related to the efficacy of melatonin in reducing IR-mediated cellular injury is relevant to the transplantation procedure. This molecule could be useful in protecting transplantable organs from hypoxia associated with organ storage and reoxygenation when the tissue is reperfused after transplantation. The utility of melatonin for this purpose was initially recognized by Viarette et al.247 In this study, the liver was isolated from rats and immersed in either University of Wisconsin (UW) or Celsior storage solutions for 20 hours at 4°C. Thereafter, the hepatic tissues were perfused with Krebs Henseleit bicarbonate (KHB) buffer with or without melatonin (25, 50, 100 or 200 μmol/L). Perfusing the livers with melatonin caused a dose–response rise in bile production (Fig. 9) and in the amount of bilirubin in the bile. All doses of melatonin induced a comparable increase in hepatic ATP levels. Both hepatic and biliary concentrations rose proportional to the melatonin dose. The authors concluded that the addition of melatonin to the perfusion fluid led to more healthy hepatocytes increasing the likelihood that, if transplanted, the liver would have an improved its chance of survival.

Details are in the caption following the image
Melatonin improves bile production from livers prepared for transplantation. Surgically removed livers were flushed with Wisconsin (A) or Celsior (B) preservation solutions. They were then stored for 20 h at 4°C. Thereafter, they were perfused with Krebs–Henseleit buffer containing melatonin (100 μmol/L) or no melatonin. Melatonin (solid points) significantly (*P<.05) improved bile flow compared to that from livers not perfused with melatonin (hollow points). From Viaretti et al.247 with permission

In a follow-up study,248 this group histochemically examined the levels of ROS in situ in cold-preserved livers that were subsequently perfused with warm KHB solution with or without melatonin (100 μmol/L). Cold storage was achieved in either UW or Celsior preservation solution. The presence of melatonin in the reperfusion medium reduced histochemical evidence of ROS production in hepatocytes and also maintained a more normal morphology of the cells.

Many livers destined for transplantation are steatotic; therefore, they more likely to functionally fail when transplanted. Zaouali et al.249 performed studies similar to those described above to determine whether melatonin would also improve the function of fatty livers. Steatotic and nonsteatotic livers were obtained from obese and lean Zucker rats, respectively, and were stored for 24 hours at 4°C in either UW or Institute Georges Lopez (IGL-1) solution with or without melatonin (100 μmol/L); thereafter, they were subjected to ex vivo normothermic reperfusion (2 hours at 37°C). In both liver types, melatonin lowered the release of transaminases (indicative of fewer damaged hepatocytes), improved bile output, enhanced bromosulfophthalein clearance, and caused a diminution in vascular resistance. These benefits were consistent with the observed reduction in oxidative stress and lowered cytokine release. The implication is that the use of melatonin in organ storage solutions may improve the function of these organs once they are transplanted. Also, the fact that melatonin recouped the function of the steatotic livers suggests moderately damaged livers could potentially be used for successful transplantation if they were treated with melatonin; this is particularly important given the acute shortage of healthy transplantable organs.

The most thorough investigation as the utility of melatonin in organ transplantation was provided by Garcia-Gil et al.250 who performed pancreas allotransplants in pigs. In this study, the efficacy of two antioxidants was compared, that is, melatonin and ascorbic acid (AA). The antioxidants were intravenously administered to the donor and recipient pigs during surgery and for 7 days postsurgery; these antioxidants were also added to the UW storage solution before the organs were transplanted. Melatonin proved highly effective in prolonging allograft survival (25 days) relative to the survival of grafts from control (8 days) or AA-treated pigs (9 days) (Fig. 10). Melatonin also had a greater inhibitory effect on indices of lipid peroxidation (malondialdehyde and 4-hydroxyalkenal) in pancreatic tissues. Moreover, melatonin reduced serum pig's major acute-phase protein/inter-α-trypsin inhibitor heavy chain 4 (PMAP/ITIH4) in the early post-transplantation period. By all indices, the benefits of melatonin exceed those of AA and suggest tests of this important molecule in additional transplantation studies, including in clinical trials. The findings related to the likely utility of melatonin in organ transplantation have recently been reviewed,251, 252 and, in a separate report, melatonin was also suggested for use in ovary transplantation.253

Details are in the caption following the image
Duration of survival of pancreaticoduodenal allografts in pigs. The untreated control pigs rejected the grafts by 12 d postoperatively (solid line). Ascorbic acid (heavy dashed line) did not prolong the autografts beyond those of the control pigs. Melatonin treatment (dotted line) significantly prolonged the survival time of the pancreaticoduodenal grafts. Each group contained eight pigs. From Garcia-Gil250 with permission

2.5 Melatonin as an antioxidant: evidence from toxic drug studies

Drugs for the treatment of diseases are approved on the basis of their cost/benefit ratio. Often drugs have a significant physiological downside, but when the benefits are presumed to outweigh the damage they inflict, they are sanctioned. Some of the side effects of these drugs progress to the point where the damage becomes life threatening. In many cases, the damage that drugs cause are a consequence of molecular processes within cells that culminate in free radical generation leading to oxidative stress and cellular malfunction. Because of this, over a decade ago, we introduced the idea that toxic drugs should be taken in combination with melatonin, so the associated free radical damage could be mitigated.254, 255 Melatonin has not been found to interfere with the efficacy of prescription drugs and, in those cases, where a drug's use is limited by its toxicity, for example, doxorubicin, if given it in combination with melatonin may allow the use of a larger dose with greater efficacy.255

Cholesterol-lowering statins are some of the most widely prescribed drugs in the world, and their side effects are well documented. Well-known adverse effects of statin use include myalgia and myopathy; occasionally these progress to rhabdomyolysis,256 a serious consequence that can lead to incapacitation and death. Moreover, rhabdomyolysis can cause acute renal failure, electrolyte disturbances, disseminated intravascular coagulation and other negative effects.257 Other potential negative consequences of regular statin use include elevation in the levels of serum aminotransferase,258 cognitive impairment,259 and what has been referred to as new-onset diabetes mellitus, while rare, older patients may be at greater risk for the latter complication.260

Each of the side effects of statin use likely involves free radical production, and, mechanistically, this is especially the case with the most serious complication, rhabdomyolysis. While the causes of this degenerative muscle condition are complex, a final common pathway involves large increases in free ionized Ca2+ in the sarcoplasm and mitochondria of muscle cells.261 The rise in free Ca2+ leads to downstream events that culminate in mitochondrial damage, reduced ATP production, and generation of free radicals, which cause further damage and malfunction.262 At the level of the kidney, myoglobin released from the damaged sarcomeres induces oxidative damage and dysfunction of renal mitochondria leading to acute renal failure.263

As repeatedly stated herein, melatonin is a potent protector against oxidative stress, a major contributory factor to the side effects of statins. Moreover, melatonin regulates free Ca2+ movement intracellularly.264-266 To potentially improve the utility and safety of statins, these authors urged the performance of both experimental studies and clinical trials to determine whether melatonin has the ability to forestall the toxicity of these very widely used, cholesterol-lowering drugs.

There are only a few studies where melatonin and statin drugs have been examined in the same report. Atorvastatin, in addition to lowering cholesterol, has protective actions on endothelial cells, which retard the development of arthrosclerosis. This statin promotes the expression of endothelial nitric oxide synthase (eNOS) resulting in vasodilation. As melatonin also has beneficial actions at the level of the endothelium, Dayaub et al.267 tested the synergistic effects of melatonin and atorvastatin on human umbilical vein endothelial cells (HUVEC) incubated with bacterial LPS. The combination of drugs induced higher eNOS protein expression than they did individually. Melatonin, but not the statin, exhibited the predictable antioxidant actions; however, when the drugs were combined, the protection against LPS was further improved. These findings are consistent with melatonin's ability to provide beneficial effects to atorvastatin while reducing oxidative stress associated with the inflammation advanced by LPS.267

Statins reportedly have additional benefits including oncostatic actions and antifibrillating and defibrillating potential. Melatonin was found to exaggerate the cancer-inhibiting actions of pitavastatin268 and pravastatin269 against breast cancer in experimental studies. Melatonin also has antiarrhythmic potential equivalent to that of atorvastatin in an isolated heart model.270 These findings support a closer examination of melatonin as an adjunct treatment with statins.

Methamphetamine is a common drug of abuse. This toxin, in addition to destroying the gingiva and periodontium in the oral cavity,271 has even more serious effects in the central nervous system.272, 273 There is general agreement that the toxicity of methamphetamine involves oxidative stress.274 This likely prompted the group led by Govitrapong to test whether melatonin could ameliorate the effects of this drug on the brain.275 In in vitro studies, they have shown that melatonin reduces methamphetamine-elicited autophagy,276 inflammation,277 and hippocampal progenitor cell death278 and conserves blood–brain barrier integrity of brain microvascular endothelial cells.279 They and others also have conducted in vivo studies and report that melatonin prevented the changes in neuronal nestin, doublecortin, and beta-III tubulin in mice treated with methamphetamine (Fig. 11).280 The toxic drug also suppressed neuronal nitrogen-activate protein kinase and altered the expression of the N-methyl-D-aspartate receptor subunits NR2A and NR2B; each of these effects was attenuated when the mice were given melatonin. Using mice and a liposomal melatonin preparation, Nguyen et al.281 found that one of the major targets by which melatonin reduces methamphetamine-related neuronal damage is due to the inhibition of the PKC-δ gene. This could account for the ability of melatonin to protect against mitochondrial dysfunction, apoptosis, and dopaminergic degeneration which occurs when mice are treated with methamphetamine.

Details are in the caption following the image
Effects of methamphetamine (METH) (500 μmol/L) without/with melatonin (100 μmol/L) on (A) nestin, (B) doublecortin (DCX), (C) BIII tubulin, and (D) glial fibrillary acid protein in neuronal phenotypes in culture. * and ** indicate P<.05 or P<.01, respectively, compared to controls; # indicates P<.05 compared to the METH group. From Ekthuwapranee et al.278 with permission

2.6 Melatonin as an antioxidant: food for thought

In the United States and many other countries, alcohol consumption and cigarette smoking are permitted. These habits reduce the quality of life of hundreds of thousands of humans annually. They contribute greatly to healthcare costs, which are already strained and their use causes the premature death of numerous humans. Yet, their use is endorsed. In contrast, getting support for an endogenously produced molecule such as melatonin, which experimentally at least, reduces the toxicity of alcohol consumption (Fig. 12)282, 283 and cigarette smoke,284, 285 has not been easy.

Details are in the caption following the image
(A) Histological evidence showing that melatonin (10−6 mol/L) reduced lipid accumulation (evaluated by oil red O staining) in alcohol plus oleic acid-treated HepG2 hepatocytes only (b); (a) are untreated control cells. (B) Dose–response inhibition of lipid accumulation by melatonin. (C) Dose–response inhibition of triglycerides in hepatocytes. O, oleic acid; A, alcohol. ##P<.01 verses untreated controls; **P<.01 verses untreated control cells; *P<.05 and P<.01 alcohol-treated cells (second histogram from the left in both B and C). From Rui et al.283 with permission

One criticism that is often levied against melatonin is that the potential negative consequences of its chronic use are not known. As noted above, melatonin is a component of the metabolic machinery perhaps of every organism, extinct and living, including organisms from bacteria to humans and plants; it predictably evolved a couple billion years ago.286 Humans and all other species have managed to survive even though melatonin is continually endogenously produced throughout the life time of these species; thus, at least at physiological concentrations, melatonin has been “tested” in the long term. Pharmacological levels could, of course, have negative effects that have not yet revealed themselves. The vast majority of the published data, however, document that melatonin has a high-safety profile and many publications have verified its beneficial actions. Regarding tests to define the consequences of its long-term use, it should be noted there are numerous highly toxic drugs approved for use in humans. Moreover, in at least some cases, melatonin reduces the toxicity of these pharmacological agents in normal cells254, 287, 288 while enhancing the cancer-killing actions (also, see below) of conventional chemotherapeutic agents.254, 289-291 Yet, melatonin has not been sanctioned for use with these drugs or chemotherapies when they are given.

Glioblastoma, a common and deadly brain cancer that rapidly invades the surrounding tissue, is often refractory to conventional therapies that are used to kill them. The resistant glioblastoma cells do not respond well to TRAIL, the death receptor ligand, which promotes apoptosis signaling cascades.292 When TRAIL was combined with melatonin for the treatment of A172 and U87 human glioblastoma cells, however, apoptotic cell death was greatly exaggerated over that caused by TRAIL alone (Fig. 13).13 Based in their results, the authors proposed that the observed effect was related to a modulation of protein kinase c which reduced Akt activation resulting in a rise in death receptor 5 (DR5) levels; concurrently, the combination treatment reduced concentrations of the anti-apoptotic proteins, Bcl-2 and survivin. These observations are consistent with the repeated confirmation that melatonin enhances apoptotic cell death in many cancer types while reducing apoptosis in normal cells.293 Because of these differential responses, the effects of melatonin on apoptosis are defined as being context specific.

Details are in the caption following the image
Melatonin sensitizes two human glioma cell lines, that is, A172 and U87, to TRAIL-mediated apoptosis. (A) Melatonin (1 mmol/L for 24 h) greatly increased apoptosis in both glioma cell types treated with TRAIL (100 ng/mL) added after melatonin. Apoptosis was assessed using the annexin V-binding assay. *P<.05 verses untreated controls; #P<.05 verses TRAIL alone. (B) the pan-caspase inhibitor, ZVAD-fmk, reduced the apoptotic effects of combined melatonin/TRAIL treatment in both glioma cell types. ZVAD was added 4 h before melatonin. Cell viability was determined using the MTT assay. *P<.05 verses untreated controls; #P<.05 verses TRAIL alone. (C) Western blots of caspase cleavage after combined melatonin/TRAIL treatment. From Martin et al.292 with permission

The finding of Martin et al.292 using glioblastoma cells are not an isolated observation. The same group reported that Ewing sarcoma, the second most common bone cancer, was also more profoundly killed when melatonin was in the mix.294 Thus, Ewing cancer cells exhibit a greatly exaggerated apoptotic response when vincristine or ifosfamide treatment is combined with melatonin. Again, the major action seems to involve the extrinsic apoptotic pathway with marked increases in caspases 3, 8, and 9 and Bid when the treatments are combined. Also, in these cells, there was a substantial rise in free radical production that likely aided in apoptosis induction. The pro-oxidant action of melatonin is common in cancer cells, while in normal cells, the indoleamine is a powerful antioxidant.104 This, again, points out the context specificity of melatonin's actions.

Cultured human breast cancer cells otherwise moderately sensitive to ionizing radiation were increasing susceptible to radiotherapy when they were treated for a week with physiological concentrations of melatonin.295, 296 Molecular studies of these cells indicated that the elevated sensitivity of the cancer cells involved a host of intracellular processes concerned with the regulation of proteins related to double-strand DNA breaks and to estrogen biosynthesis. Similar studies in human lung adenocarcinoma cells (SK-LV-1) showed that melatonin also increased their sensitivity to the chemotherapy, cisplatin.297 In this case, the reduced cell proliferation was mediated by cell cycle arrest in the S phase.

In vivo, as well, melatonin changes the sensitivity of cancer cells to chemotherapies. Some breast cancers are resistant to the chemotherapeutic agent, doxorubicin. Xiang et al.60 showed that MCF-7 human breast cancer cells growing in athymic nude rats grew faster when the daily dark period (animals on a 12:12 LD cycle) was contaminated with a light intensity that reduced the nocturnal endogenous melatonin peak (Fig. 14). Conversely, in rats experiencing darkness at night, which allowed the nighttime rise in melatonin, the tumor latency to onset, tumor regression, and reduced tumor metabolism were observed. Moreover, tumors growing in the rats exposed to darkness at night greatly increased their sensitivity to doxorubicin. The authors reported, in a related publication, that metabolically the tumors grown in rats exposed to light pollution at night are markedly different from the metabolism of those in rats exposed to darkness at night. The conclusion is that chronodisruption and melatonin suppression due to light at night accounted for the decreased sensitivity of the tumors to doxorubicin.

Details are in the caption following the image
Effect of doxorubicin (DOX) on the growth and regression of MCF-7 (ER α+) breast tumor xenografts growing in a athymic nude female rats exposed to a light:dark cycle of 12:12 with the dark period contaminated with dim light exposure at night (dLEN) or dLEN supplemented with melatonin during the dim light period. (A) Estimated tumor weight (based on tumor measurements) in rats exposed to a dLEN lighting schedule and left untreated (red triangles) or DOX (blue diamonds) or exposed to dLEN and supplement with melatonin (black triangles) or dLEN and melatonin plus DOX (inverted green triangles). Photographs of tumors in rats maintained in either LD 12:12 + dLEN (B) or LD12:12 + dLEN but supplemented with melatonin (C). Panels (D) and (E) show tumors in the animals after 45 d after tumor implantation in animals kept in dLEN + melatonin (D) or dLEN + diluent (D). * identifies the location of the tumors. From Xiang et al.60 with permission

The collective data on the association of melatonin with cancer indicate that while melatonin itself has intrinsic cytotoxic actions in cancer cells,59, 61, 298-300 it also sensitizes some cancers to conventional therapies and it reduces the toxicity of chemotherapies in normal cells, that is, it reduces the side effects of these drugs. This latter action would allow the chemotherapy to be given at higher doses, which would likely increase its cancer-killing activity. Overall, this information should be of interest to clinical oncologists; it is the hope of the authors that this information does not merely languish in the published literature. In view of the published data related to melatonin's ability to change the sensitivity of cancer cells to therapeutic agents, it is interesting to imagine that bacteria that become insensitive to drugs would perhaps exhibit renewed sensitivity if they were exposed to melatonin.

As already noted, a major consideration for the approval of any drug is its cost-to-benefit ratio. If the benefits derived from the use of even a highly damaging drug are determined to outweigh the physiological impairment it causes, its use may be approved. Using the same formula to evaluate melatonin, the data are overwhelmingly in favor of its benefits far exceeding the potential negative side effects, which under the worst case scenarios, seem minimal.

Melatonin has been available to the public for about 20 years, and, based on published sales figures, it may be taken regularly by tens of thousands of individuals. There are few reports of serious side effects due to its regular use and, if highly damaging, individuals would be “dropping like flies.” If industry had a patentable molecule as efficacious as melatonin, it likely would have been tested and approved for large-scale, long-term use years ago. There are a number of patented melatonin analogs that are already sanctioned as drugs which, as they do not exist in nature, are always given in pharmacological doses. It seems reasonable to assume that the likelihood of them having toxicity in the long term would be greater than that for melatonin. There should be long duration trials of melatonin against serious diseases (where few treatments are available) where it has been shown beneficial in limited clinical studies or where the experimental evidence is compelling. Some examples include melatonin's ability to forestall Alzheimer's disease,301-303 Parkinson's disease,304-306 multiple sclerosis,307, 308 osteoporosis,309-311 diabetes and metabolic syndrome,312-315 sepsis,316-318 cancer,319-321 tropical diseases,322-325 snake and nematocyst venom toxicity,326-329 etc. In some cases, rather than a treatment for these conditions, melatonin should be more strongly considered in terms of its preventative actions as prevention always trumps treatment and is usually less expensive and certainly less debilitating.

Finally, during the Ebola epidemic in West Africa in 2014, two groups independently proposed the use of melatonin to slow the progression of this disease so as to improve survival of the affected individuals.330, 331 In our publication, we highlighted the scientific evidence, which prompted our suggestion to use melatonin against this dreaded condition. Ebola virus disease is characterized by severe inflammation, coagulopathy, and endothelial disruption,332 changes not unlike those caused by LPS-mediated sepsis, which has been successfully treated with melatonin.333, 334 Numerous reports also have documented the anti-inflammatory actions of melatonin.335-337 Another feature of melatonin is its ability to reduce endothelial damage.338, 339 Whereas the evidence may be somewhat less compelling, melatonin's favorable effects on coagulopathy also have been described.340 The rationale for the use of melatonin as a potential treatment voiced by Anderson et al.331 was similar to that proposed by Tan et al.330 Anderson et al.331 also noted that melatonin upregulates heme oxygenase, which inhibits the replication of the Ebola virus.

The most recent viral scourge is that of the Zika virus.341 Based on the antagonistic effects of melatonin on viral infections generally,342-345 and as, like Ebola, there are few treatment options for Zika, perhaps melatonin should be given consideration to combat this viral infection as well.

3 Epilogue and Perspective

Melatonin has a very large physiological footprint and some of the mechanisms by which this is achieved are illustrated in Fig. 15. There is likely no organ or cell that is not impacted by this molecule. As summarized in this report, melatonin has a plethora of actions that make it extraordinarily efficacious in reducing the subcellular turmoil induced by oxidative destruction of key cellular elements which, when damaged, compromise the optimal function of cells often resulting in their disintegration via apoptosis or necrosis. Melatonin, in its capacity as an antioxidant, is proposed to have been the original function of this ancient and ubiquitously distributed molecule. Melatonin seems to be a linchpin of the highly complex antioxidative defense system.

Details are in the caption following the image
Some of the multiple molecular actions of melatonin, which account for its efficacy in reducing oxidative damage. Melatonin directly scavenges (illustrated on the left) ROS/RNS via receptor-independent actions thereby reducing mitochondrial damage and the apoptotic cascade. Melatonin may also act on cytosolic quinone reductase (MT3) to eliminate free radicals and reduce oxidative damage. The receptor-mediated actions are summarized on the right. Melatonin acts via membrane receptors (MT1/MT2) to stimulate a cascade of events which increase transcriptional activity; this leads to an upregulation of antioxidant enzymes and a downregulation of pro-oxidant enzymes as well as a reduction in toxic cytokine synthesis. Melatonin also binds to calmodulin to modulate nitric oxide production. Finally, some of these actions may also involve nuclear binding sites (RoR-α and RZR). Figure provided by Dr. Nicola Robertson

In addition to its steadfastness in resisting oxidative stress, melatonin has a very wide number of essential molecular mechanisms (Fig. 15). What is usually measured as a result of melatonin actions, however, may merely be epiphenomena of its yet-to-be identified most fundamental ethos. Because of its highly divergent manifested actions, since its discovery almost six decades ago, melatonin has been designated as a regulator of regulators,346 a refiner of physiology,347 a tranquilizing agent,348 a multitasking molecule349 nature's most versatile signal,156 etc. Recently, it was even classified as a biological Higgs boson,350 a phrase that may actually best characterize this ingenious agent. It is the authors’ current opinion that melatonin's basic function has yet to be uncovered or to put it in less formal terms, we are “seeing the smoke but not the fire.”

In our estimation, it is unfortunate that melatonin is not more in the forefront of biomedical research. While it has gained some traction at the clinical level, its low toxicity profile and high efficacy in many pathophysiological states should make it a molecule more commonly tested/used in the medical and veterinary arenas. Certainly, one goal of this review was to strongly urge more attention be directed to melatonin in terms of is likely usefulness as a preventative and treatment for human and animal diseases.

    The full text of this article hosted at iucr.org is unavailable due to technical difficulties.