Next Article in Journal
Low Doses of Cuscuta reflexa Extract Act as Natural Biostimulants to Improve the Germination Vigor, Growth, and Grain Yield of Wheat Grown under Water Stress: Photosynthetic Pigments, Antioxidative Defense Mechanisms, and Nutrient Acquisition
Next Article in Special Issue
Role of Melatonin in the Synchronization of Asexual Forms in the Parasite Plasmodium falciparum
Previous Article in Journal
α-Lactalbumin, Amazing Calcium-Binding Protein
Previous Article in Special Issue
Cellular Mechanisms of Melatonin: Insight from Neurodegenerative Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Melatonin’s Impact on Antioxidative and Anti-Inflammatory Reprogramming in Homeostasis and Disease

by
Diana Maria Chitimus
1,†,
Mihaela Roxana Popescu
2,†,
Suzana Elena Voiculescu
1,
Anca Maria Panaitescu
3,
Bogdan Pavel
1,
Leon Zagrean
1 and
Ana-Maria Zagrean
1,*
1
Division of Physiology and Neuroscience, Department of Functional Sciences, “Carol Davila” University of Medicine and Pharmacy, 010164 Bucharest, Romania
2
Department of Cardiology, “Carol Davila” University of Medicine and Pharmacy, Elias University Hospital, 010164 Bucharest, Romania
3
Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, Filantropia Clinical Hospital, 010164 Bucharest, Romania
*
Author to whom correspondence should be addressed.
These authors contributed equally to the work.
Biomolecules 2020, 10(9), 1211; https://doi.org/10.3390/biom10091211
Submission received: 1 July 2020 / Revised: 30 July 2020 / Accepted: 18 August 2020 / Published: 20 August 2020
(This article belongs to the Special Issue Recent Advance of Melatonin)

Abstract

:
There is a growing consensus that the antioxidant and anti-inflammatory properties of melatonin are of great importance in preserving the body functions and homeostasis, with great impact in the peripartum period and adult life. Melatonin promotes adaptation through allostasis and stands out as an endogenous, dietary, and therapeutic molecule with important health benefits. The anti-inflammatory and antioxidant effects of melatonin are intertwined and are exerted throughout pregnancy and later during development and aging. Melatonin supplementation during pregnancy can reduce ischemia-induced oxidative damage in the fetal brain, increase offspring survival in inflammatory states, and reduce blood pressure in the adult offspring. In adulthood, disturbances in melatonin production negatively impact the progression of cardiovascular risk factors and promote cardiovascular and neurodegenerative diseases. The most studied cardiovascular effects of melatonin are linked to hypertension and myocardial ischemia/reperfusion injury, while the most promising ones are linked to regaining control of metabolic syndrome components. In addition, there might be an emerging role for melatonin as an adjuvant in treating coronavirus disease 2019 (COVID 19). The present review summarizes and comments on important data regarding the roles exerted by melatonin in homeostasis and oxidative stress and inflammation related pathologies.

1. Introduction

Melatonin is a pineal hormone produced and released in relation to the circadian rhythm, while also synthesized in extrapineal tissues, like heart, liver, placenta, skin, kidney, gut, etc. [1,2,3,4]. Melatonin is an important regulator of physiologic processes and a guardian of body homeostatic balance. Its level varies during the day from 5 to 200 pg/mL [5].
Melatonin has antioxidant effects exerted through direct and indirect mechanisms that make this unrivaled multitasking molecule an endogenous protector against highly toxic oxygen- and nitrogen-derived free radicals. The main mechanisms of action attributed to melatonin are free radical scavenging, endogenous antioxidative enzymes stimulation, and improving the efficiency of other antioxidants. Melatonin’s particularity is that together with its metabolites, which act as antioxidants themselves, creates an antioxidant cascade that yields radical scavenger products [6], limiting the oxidative damage through a variety of mechanisms [7,8]. Thus, the radical quencher property of melatonin against the hydroxyl radical (OH) is superior to that of glutathione, while its action against the peroxyl radical (ROO) involves single electron transfer, hydrogen atom transfer, or radical adduct formation [8,9]. Besides lowering the amount of free radicals, melatonin can also interact with non-radical oxidants such as hydrogen peroxide (H2O2), singlet oxygen (1O2), and peroxynitrite (HNOO) [6,10]. Melatonin is effective in inhibiting metal-induced oxidation, as it was reported for copper, a redox generating metal, but it also chelates iron, lead, zinc, and aluminum [8].
Along with its free radical scavenging properties, melatonin protects the mitochondria against oxidative stress by influencing the mitochondrial membrane potential, thus facilitating electron transfer antioxidant processes within the cell [11]. The roles of melatonin within mitochondria are exemplified in Figure 1.
Melatonin works through both receptor-independent and receptor-dependent antioxidant processes. Through its receptor-mediated actions, melatonin either inhibits pro-oxidative enzymes such as xanthine oxidase or enhances the activity of superoxide dismutase (SOD), glutathione peroxidase, and catalase [12,13].
Growing evidence supports the anti-inflammatory role of melatonin, both in acute and chronic inflammation processes. However, most of the data are obtained from in vitro and in vivo experimental studies, while clinical studies have proven inconsistent results [14]. Administration of exogenous melatonin in animal studies, prior to acute conditions, have shown a decrease in the inflammatory response, a reduction of pro-inflammatory cytokines, interleukine-1β (IL-1β), and tumor necrosis factor-α (TNF-α), and an increase in anti-inflammatory cytokine IL-4 levels in serum [15,16]. In addition, melatonin inhibits the expression of cyclooxygenase (COX) and inducible nitric oxide synthase (iNOS), while reducing the production of high concentrations of prostanoids and leukotrienes, as well as other mediators of the inflammatory process, such as chemokines and adhesion molecules [17,18].
Melatonin also exerts an anti-apoptotic effect that depends on its capacity to optimize the mitochondrial function, through antioxidative mechanisms. Some of the anti-apoptotic effects of melatonin are associated with an increase in the anti-apoptotic factor Bcl2, and a decrease in the pro-apoptotic factors Bax and caspase 3 [19,20,21]. However, an anti-angiogenic and pro-apoptotic effect through vascular endothelial growth factor (VEGF), hypoxia-induced factor 1α (HIF1α), Janus kinase 2/signal transducers and activators of transcription 3 (JAK/STAT3) inhibition, was observed in hepatic carcinoma [22]. It seems that melatonin tips the scales of the pro/anti-apoptotic balance according to local needs, in order to maintain homeostasis.
Melatonin has been shown to have pleiotropic effects in numerous neurology, endocrinology, cardiology, fetal medicine, and oncology studies [20,23]. Moreover, its protective and allostatic effect spreads over many organs and systems (Figure 2).
The allostatic load implies the incapacity of the body systems to effectively adapt and cooperate in order to discard the effect of external stressors. Chronodisruption and sleep-deprivation elevate the allostatic load by altering the biological rhythms of hormones.
Melatonin plays an important role in establishing continuous chrono-biological homeostasis, as revealed by the increased load of allostasis in circadian disruptive patterns. Hunter et al. described a 33.8% reduction in melatonin production in night-shift workers over 24 h. Although there is no clear connection between increased cancer incidence, overall mortality, chronodisruption, and melatonin suppression, multiple studies reported a significant increment in breast or ovarian cancer in night-workers. Moreover, shift workers display a slow adaptation over the following week and have a lower night-time melatonin blood concentration [24]. This might explain why insufficient melatonin production favors an overload of stress and/or inefficient management of the body resources.
As melatonin has a rather short half-life (0.57–0.67 h), the need for longer acting substances has prompted the synthesis of analogues such as ramelteon, piromelatine, agomelatine, and tasimelteon. Ramelteon has a half-life of 1.5–2 h, with a metabolite with an even longer half-life (2–5 h) [25]. Ramelteon has a higher affinity for melatonin receptors (3–16 fold than melatonin) and is recommended in 8 mg dose for onset insomnia, decreasing sleep latency, and increasing sleep duration. Agomelatine and tasimelteon reach peak levels after 1–2 h and 0.5–3 h, respectively [25]. Agomelatine is indicated in treating sleep disorders associated to major depressive disorder. Its dose is 25 mg and has good oral absorption (80%). Tasimelteon is indicated in non-24-h sleep-wake disorder, in a dose of 20 mg [26,27].
With regard to exogenous melatonin administration and its side effects, up to date, only few studies focused on investigating possible adverse reactions as their primary objectives. Andersen et al. summarized in their analysis the safety of melatonin administration in humans, in both infants and adults. The most common side effects reported throughout literature were dizziness, headache, nausea, and sleepiness, while the long-term administration registered few to none adverse reactions [28].
In addition to the previously mentioned attributes, melatonin is intimately linked to reproduction and its most prominent role is to mediate reproductive effects in mammals that breed seasonally. In humans, melatonin influences reproduction as suggested by the widespread distribution of its receptors in reproductive organs and systems. Melatonin modulates the production and function of the human gonadotropins and steroid hormones and is considered to influence the onset of puberty, sexual maturation, follicular genesis and ovulation, pregnancy, and menopause [29]. In late pregnancy, melatonin concentration increases steadily towards the last trimester, returning to normal values after delivery [30,31]. The embryo and the fetus express receptors for melatonin since early stages of pregnancy suggesting that melatonin plays a crucial role in normal development during intrauterine life.
In the early stages of embryo development, the DNA suffers dynamic changes through active and passive demethylation and de novo methylation [32,33], crucial for fertilization, development, and tissue differentiation [34]. The rate-limiting enzyme for melatonin production, aralkylamine N-acetyltransferase (Aanat), mostly identified at the mitochondrial level, was linked to DNA demethylation. Thus, Aanat knockdown inhibited ten-eleven-translocation (TET) methylcytosine dioxygenase 2 (TET2) expression and DNA demethylation in the blastocyst, severely altering embryonic development and cellular differentiation [35]. These effects were counteracted by melatonin supplementation [36].
Melatonin was also explored as a neuroprotective molecule in ischemic conditions, like stroke, for its antioxidant and anti-inflammatory actions. Several studies on animal models showed that melatonin therapy diminishes the toxicity induced by the glutamate-triggered increase in intracellular calcium in focal cerebral ischemia [37], increases the survival rate, and reduces neurodegeneration and post-ischemic neuronal loss [38]. The effectiveness of melatonin to improve neural viability not only following acute injury, but also as a long-term therapeutic agent, grants its position as a promising neuroprotector [39].
Given its high production in the neuronal mitochondria, melatonin is capable of interfering with neuronal dysfunction in the early stages of neurodegeneration [40]. Cellular senescence is accelerated by free radicals such as reactive oxygen (ROS) and nitrogen (RNS) species, which endorse caspase activation, cytochrome c release from mitochondria, and p53 protein cycle control [41,42] (Figure 1). Melatonin displays neuroprotection by diminishing cytochrome c release and caspase activation, and by reducing the activation of pro-inflammatory cytokines and thus, neurodegeneration [43]. Furthermore, melatonin lowers cell mortality rate and the mitochondrial dysfunction via the silent information regulator 1 (SIRT1) signaling pathway in cerebral ischemic lesions, in a similar way as it does in the myocardial injury [44].
Melatonin’s ability to prevent the neuronal death induced by kainate, a glutamate receptor agonist, supports its anti-excitotoxic activity [45]. Excitotoxicity is one of the most important pathophysiological mechanisms in ischemia/reperfusion (I/R) brain injury. The reactions following brain I/R injury are interlinked and often consist of calcium homeostasis alteration, an accumulation of reactive free radicals and activation of pro-inflammatory agents [46]. With regard to the in vitro experimental designs, melatonin was proven to counteract the destructive effects of hypoxia followed by reperfusion in primary cultures of rat cortical neurons [47].
An overview of melatonin’s mechanisms of action and protective effects is presented in Table 1.
As an endogenous and therapeutic molecule, melatonin ubiquitously exerts important benefits for human health. The present review summarizes significant and new data regarding the roles played by melatonin in homeostasis, development, and in oxidative-stress related pathologies, which favor allostatic overload. We chose to include some of the highest impact subjects on the matter, namely, the effects of melatonin on maternal-fetal health, cardiovascular diseases, neuroinflammation-related pathologies like degenerative neurological disorders, and respiratory distress caused by SARS-CoV-2 infection.

2. Melatonin Involvement in Maternal and Fetal Health

Melatonin’s role in pregnancy has been studied extensively lately and growing understanding became available of its physiological functions and its potential therapeutic use to improve maternal and neonatal outcomes. Melatonin is now considered a key signaling molecule between the mother and the fetus and a new potential candidate in the prevention of such complications as maternal preeclampsia or neonatal encephalopathy [73].
Melatonin exogenous supplementation during pregnancy could be beneficial both for the mother and the fetus, mainly but not only due to the antioxidant proprieties of melatonin [74].

2.1. Melatonin in Maternal-Fetal Signaling

During pregnancy, all maternal organs and systems sustain important changes to support the proper development of the fetus, in a continuous interplay between the maternal and fetal systems that promotes normal development.
The pineal gland also undergoes structural and functional changes. Serum levels of melatonin increase in the second and third trimester, pick at term, and decline to normal immediately postpartum [75]. The specific cyclic night-time pattern of production for melatonin is maintained during normal pregnancy and lactation [30]. The placenta contributes to melatonin production but in a non-rhythmic manner [76]. Placental melatonin predominantly acts in an autocrine and paracrine fashion [50]. Because of its lipophilic structure, melatonin is transferred easily across the placenta into the fetal circulation [77]. The fetus relies on melatonin transferred from the mother since its pineal gland, though present in late gestation, begins functioning only after 3–5 months postnatally [78]. Fetal tissues present melatonin receptors, hence it is expected that maternal melatonin is involved in fetal growth and development [79]. In particular, the fetal brain displays melatonin receptors in multiple areas, thus maternal melatonin may play a role in the early stages of fetal neurodevelopment [80].
Recent evidence suggests that the circadian rhythm of the fetus and the newborn relay on the programming imprinted by the maternal circadian rhythms. Maternal melatonin is considered a key molecule for maternal-to-fetal signaling of circadian rhythm as it readily crosses the placenta and the levels in the maternal blood are mirrored in the fetal circulation, the mother seems to be the sole source and the fetus presents receptors in relevant areas. The circadian-generating systems in the fetus, particularly the suprachiasmatic nucleus, are considered to be under the influence of maternally derived melatonin [76]. Disturbances in the fetal circadian system may trigger long-term consequences. Interference to normal light/dark cycle in late pregnancy or during the perinatal period may have detrimental effects on the behavior and metabolic functions of the offspring, possibly contributing to conditions such as metabolic syndrome, obesity, attention deficit-hyperactivity disorder, or autism spectrum disorders [81,82,83,84].
Melatonin is excreted in the human milk early post-partum, especially at night, to compensate for the neonate lack of melatonin, as melatonin sustained rhythmic production starts only after several months or even later in preterm babies [85,86].

2.2. Melatonin for the Prevention of Maternal Pregnancy Complications

Alterations in normal melatonin production have been studied as a possible contributor to adverse pregnancy outcomes. A recent systematic review and meta-analysis by Cai at al. showed that pregnant women working rotating and night shifts, when compared to those working day-time only, have an increased risk of preterm birth (PTB), delivery of a small-for-gestational age baby, developing preeclampsia (PE) or gestational hypertension [87]. A possible explanation for these findings could be that reduced secretion of melatonin as a consequence of repeated disruption of the circadian rhythm and exposure to light in night workers could interfere with maternal and fetal hormone homeostasis, placental implantation, and fetal growth [55,88]. A recent study on mice where a model of induced maternal inflammation was used to mimic the clinical setting of preterm birth, showed that in mothers pre-treated with melatonin, there were fewer preterm deliveries and fewer cases of perinatal brain injuries in the pups [89]. The authors proposed that, because melatonin is considered generally safe in pregnancy, a study testing its effect for the prevention of PTB in humans should be initiated. Preterm birth (delivery of the baby before 37 weeks of gestation) is the big unresolved problem of modern obstetrics. It complicates around 7% of all pregnancies in industrialized countries and its incidence has remained the same in the last 30 years, even though obstetrical care has improved considerably [56]. While the outcomes of babies born preterm are improving due to the scientific and technical advancements in the field of neonatology, preventive, and therapeutic strategies for the mothers should be continually sought. The anti-inflammatory and antioxidant proprieties of melatonin are well recognized, and evidence from fundamental research studies is encouraging enough to consider testing melatonin in a randomized controlled trial (RCT) in the prevention of PTB in humans. This kind/type of trial would have to include around 1400 singleton pregnancies treated with either melatonin or placebo for a hypothetical reduction with 50% in the incidence of PTB (from 7% to 3.5%), with a power of 0.8 and alpha of 0.05. Other issues would regard the timing and duration of melatonin treatment (prenatal administration, during the first trimester or later in pregnancy), dosage, targeted population (low versus high-risk groups), and the route of administration. Melatonin supplementation is considered generally safe in humans. When given at a high dose by intravenous route, it does not induce somnolence or sedation or any other adverse events. Even with long-term administration, there are no associated adverse effects reported [54,57]. From studies with pregnant animals exposed to various doses of melatonin, there are no reported treatment-related side effects [90]. Even though data from human studies are limited, melatonin seems to have a good safety profile in human pregnancy, with no reported teratogenic effects [91].
Melatonin levels are altered in women with abnormally functioning placentas in preeclampsia and fetal growth restriction (FGR). Preeclampsia is characterized by new-onset hypertension after 20 weeks of gestation in association with maternal end-organ dysfunction and is a condition specific to human pregnancy with the only cure being the delivery of the fetus with the placenta. It is one of the leading causes of maternal mortality and morbidity, fetal death, and an important contributor to neonatal morbidity because of its need for iatrogenic preterm delivery. The pathogenesis of preeclampsia is likely to involve the mother, fetus, and placenta. In PE, there is defective placentation from early pregnancy, reduced blood perfusion in the placenta, hypoxia, and high levels of oxidative stress, with the release of trophoblast-derived factors (e.g., SFlt-1, soluble endoglin), which enter the maternal circulation and cause generalized endothelial dysfunction and an exaggerated inflammatory response [92]. For the fetus, this leads to growth restriction, preventing development to its full genetic potential. Serum levels of melatonin and placental distribution of its receptors are significantly reduced in pregnant women with preeclampsia or growth-restricted fetuses [30,93]. As a potent antioxidant and free-radical scavenger safe to be used during pregnancy, melatonin is a candidate drug for the prevention and treatment of PE and FGR. In a xanthine/xanthine oxidase (X/XO) placental explant model, melatonin was shown to reduce oxidative stress and enhance antioxidant markers, but it did not affect explant production of antiangiogenic factors (e.g., sFlt, soluble endoglin) [94]. In a model using primary trophoblast cell cultures, melatonin increased the expression of antioxidant enzymes and reduced the production of sFlt-1 [95]. In a clinical trial, 20 pregnant women diagnosed with early-PE (at the beginning of the third trimester) received 10 mg of melatonin three times daily from a median gestational age of 28 weeks until when the delivery was required for fetal or maternal indications. Compared to matched historical controls, those treated with melatonin, had a prolongation of pregnancy of ~6 days and a reduction of doses of antihypertensive medication [96]. While these effects are encouraging, more extensive, well-designed, randomized, double-blind, placebo-controlled trials are needed to test melatonin for the treatment of PE. Another approach to reducing the incidence of PE, would be to select a group of high-risk women who could possibly benefit from melatonin supplementation from the first trimester. This approach has been recently tested in a large RCT for which aspirin supplementation from 12 to 36 weeks of pregnancy in those high-risk, significantly reduced the incidence of PE [97].
FGR is frequently associated with PE. Chronic placental dysfunction, as seen in the placentas of growth-restricted fetuses, leads to intrauterine hypoxia, increases oxidative stress and ROS generation [98]. In a pilot phase I clinical trial in human pregnancy by Miller et al., melatonin (4 mg twice daily in a prolonged-release form) was administered to six pregnant women with severe early-onset FGR from a median gestational age of 27 weeks. Compared to historical control matched for diagnosis and gestational age placentas, oxidative stress was significantly reduced for those treated with melatonin [99].

2.3. Antioxidative Effects of Melatonin in Embryo-Fetal Development: Function and Therapeutic Approaches

Melatonin synthesis was identified at different stages of embryonic development and the main synthesis site has been reported to be the mitochondria [100] (Figure 1). Studies report that the melatonin level in the mitochondria was ~100-fold higher than the plasmatic concentration [48].
Melatonin plays a key role in embryonic development [101]. It has been shown that melatonin promotes embryo development in different species such as sheep, mouse, bovine, and pig [102,103,104]. All these positive effects exerted by the hormone on development have been attributed to its abilities to improve mitochondrial function and to reduce mitochondrial oxidative stress [33] combined with its function in the regulation of genomic methylation levels [105], which are key processes involved in embryonic development [106].
Melatonin is considered to act directly at the mitochondrial level, where it reduces free radical formation and also protects ATP synthesis, by stimulating key enzymatic complexes I and IV [50]. Direct scavenging of free oxygen and nitrogen species is also present at the mitochondrial level, by this protecting the membrane against disruption and supporting the continuity of the electron chain. Melatonin seems to protect mitochondrial DNA [49,107,108], and also prevents the opening of mitochondrial permeability transition pore (mPTP) in pathological conditions like traumatic brain injury and hypoxic ischemia. The opening of this pore allows macromolecules smaller than 1.5 kDa bypass the membrane, thus leading to mitochondrial swelling and death [109].
Melatonin’s role in the embryo and fetus is highly concentrated in nervous system development. The presence of a high number of melatonin receptors has been found in both nervous system organs and endocrine glands, thus supporting its function as a regulatory molecule. Melatonin receptors have been identified in human embryos and fetuses in the leptomeninges, cerebellum, thalamus, hypothalamus, brain stem, arcuate, ventromedial and mamillary nuclei. At brain stem level, there are melatonin receptor-positive areas in the nuclei of cranial nerves, such as oculomotor, trochlear, trigeminal motor and sensory, facial, and cochlear [110]. As melatonin’s main functions are represented by its antioxidative and DNA protective effects, one might assume that its highly expressed receptors in the nervous system organs during neurodevelopment point toward the same key-functions in the brain, during a period of time dominated by differentiation and subsequently high ROS production.
Hypoxic ischemia is a cause of severe brain injury in newborns and it represents one of the main causes of death in this age group and also a disability inducer [111]. During a hypoxic-ischemic injury, ROS production is increased massively, as that free species cannot be eliminated by antioxidant enzymes, leading to ROS accumulation. ROS accumulation is associated with structural changes to proteins, lipids, and DNA, thus leading to mitochondrial dysfunction, one of the main causes involved in the pathophysiology of this condition [112]. Mitochondria regulate the oxidative reactions standing at the basis of membrane potential maintenance and cellular ionic balance. Therefore, mitochondria play a crucial role in neonatal neurodegeneration. Up to date, the only efficient treatment is controlled hypothermia, but due to difficult clinical implementation and complications, it is not always applicable. New therapeutic strategies need to be searched, conceptualized, and methodically explored.
Mitochondria-based therapeutic strategies have been reported in animal models, but have not been extensively tested in humans. Part of these treatment tactics originate from the roles that melatonin plays in mitochondrial physiology and pathophysiology. Along with its antioxidant properties, it has been shown to assert anti-inflammatory functions, by inhibiting the production of various anti-inflammatory molecules, like 8-isoprostanes, an important mediator involved in hypoxic-ischemic inflammation [52]. Melatonin also exhibits anti-apoptosis properties, by inhibiting Cytochrome C (Cyt C) release, Bax and Bad pro-apoptosis molecules production and preventing DNA disruption [53,113,114].
In animal studies, melatonin was shown to enhance the antioxidant enzyme glutathione peroxidase, and to reduce oxidative stress-induced DNA and lipids damage in fetal brain homogenates, when administered intraperitoneally in pregnant rats prior to 20 min bilateral utero-ovarian artery occlusion [40]. In another study, the prophylactic administration of melatonin to pregnant rats immediately prior to an acute ischemic episode and regularly throughout pregnancy, decreased ischemia-reperfusion-induced oxidative impairment in the premature fetal rat brain [41]. In addition, melatonin supplementation was shown to reduce maternal hyperthermia-induced embryo death, prevent preterm labor, and increase offspring survival in a mouse model of lipopolysaccharide (LPS)-induced inflammation [42,43].
Melatonin depletion through continuous light exposure during pregnancy was linked to intrauterine growth retardation in a rat model, which is avertible by maternal melatonin administration [44].
Melatonin toxicity has been assessed in human studies and is extremely low when including available data from children with severe neurological conditions or neonates with sepsis. One pilot RCT tested the clinical outcomes of melatonin in neonates with hypoxic-ischemic encephalopathy (HIE) [115]. The study included 45 newborns: 30 with HIE and 15 healthy controls. HIE infants were either treated with controlled hypothermia, or hypothermia plus melatonin. Serum nitric oxide (NO), plasma superoxide dismutase (SOD), and melatonin levels were measured for the 45 neonates on admission and at 5 days since the beginning of the study. The study showed that the HIE groups had increased melatonin, SOD, and NO. After 5 days, the melatonin/hypothermia group had a significantly higher level of melatonin and a decrease in NO and SOD. This group also had a lower rate of seizures on follow-up electroencephalogram and less extensive white matter abnormalities on MRI. Importantly, at 6 months, the melatonin/hypothermia group had a significantly better survival rate. Melatonin appears to be safe and beneficial in HIE treatment. However, larger randomized controlled trials are required before melatonin can be approved for usage as a neuroprotective molecule.

3. Melatonin’s Role in Programming Adult Cardiovascular Homeostasis and Allostasis

Melatonin accompanies the development and homeostasis of the cardiovascular system all throughout life with distinctive effects in both the perinatal and adult periods. As previously discussed, the most prominent protective mechanism lies in its antioxidant potential [19,116]. In the field of cardiovascular effects, melatonin protects against ischemia/reperfusion injury (IRI) [62,117,118,119,120,121] and hypertension [122,123,124], as an endogenous effect, nutritional supplement, or acute parenteral administration [125,126]. In addition, it appears that other promising effects such as anti-apoptotic, anti-inflammatory [127], preconditioning [118,128], myocardial infarction [129,130,131], and even an implication in heart failure [132,133,134,135] have been documented. However, it seems than in some conditions, melatonin can have deleterious effects, as exaggerated collagen and glycosaminoglycans deposition and increased size in post-myocardial infarction scar have also been reported [136,137]. A reduction in cardiovascular risk factors such as diabetes and hyperlipidemia could be of use in preventive cardiovascular care. Even before disease manifests, melatonin contributes to allostasis, as it is involved in regulating prediabetes, inflammation, and lipid metabolism [124]. These risk factors precede conditions like atherosclerosis, coronary artery disease, cerebrovascular disease, etc. Slowing-down of these processes through the positive effects of melatonin could be an effective prophylactic therapy. The allostatic effects are not efficient in some individuals, as melatonin levels decrease with age and with cumulating risk factors [124]. If there is a threshold below which these positive effects are no longer observed, or if there is a specific moment in the day when the melatonin level should surpass that threshold, it is still not clear.

3.1. Prenatal and Neonatal Cardiovascular Effects

Melatonin has been implicated in prenatal programming in more than one aspect. The antioxidant role is significant for babies at risk of birth asphyxia, as the new-born myocardium has reduced antioxidant defense [138]. In addition, in an experimental setting of intrauterine growth restriction, melatonin counteracts the vascular and myocardial impairment seen in the untreated group [139].
Melatonin was shown to prevent the development of hypertension by increasing the level of nitric oxide (NO) [140], although the exact mechanism has not yet been agreed upon. Supposedly, the antioxidant properties of melatonin might aid in restoring the NO/ROS balance against programmed hypertension. The offspring subjected to high methyl-donor diet-induced programmed hypertension also displayed a lower SDMA (symmetric dimethylarginine) plasma level, which is an endogenous NO inhibitor, following maternal melatonin administration [141,142]. In a rat model, chronic photoperiod shifts (CPS) during 85% of the pregnancy were shown to generate increased systolic BP at night and higher heart rate variability at rest in the young adult offspring (raised and tested under normal light/dark conditions) [143].

3.2. Antioxidant and Anti-Inflammatory Roles in Cardiovascular Pathologies

The roles of antioxidant and anti-inflammatory agent are intertwined in the case of melatonin. The anti-inflammatory role of melatonin in clinical practice is still mechanistically challenged. Melatonin reduced free radical production, lipid peroxidation, and IL-6 levels caused by bacterial lipopolysaccharide in vitro [116]. In a human experimental sepsis model, melatonin reduces IL-1β, but does not affect IL-6 and TNF-α [144]. However, obese women who received melatonin supplementation showed lower TNF-α and IL-6 levels, a fact that was not demonstrated in the previously mentioned study [144]. The differences between the two studies were that in the sepsis model study, melatonin was given in a single, higher dose of 100 mg, and in the obesity trial, the subjects received daily doses of 6 mg over 40 days (the overall dose was 2.4× higher). Another clinical study mentions higher IL-6 levels, and lower melatonin concentration in myocardial infarction patients compared to the control group [145]. The authors suggest that there is a causal relationship between melatonin and IL-6 levels. Different research stresses that the C-reactive protein (CRP) is increased in ST-elevation myocardial infarction (STEMI) patients compared to healthy subjects. This correlates with a diminished surge in the night-time melatonin levels and also with an increased risk of adverse events in the following six months [146]. The link between CRP and melatonin is represented by IL-6. This interleukin induces CRP production and also shows an increase in relation to melatonin [145]. Inflammation plays an important role in the pathogenesis of the atherosclerotic disease [147,148]. Thus, the anti-inflammatory properties of melatonin should be exploited in a preventative capacity.

3.3. Effect on Adult Cardiovascular Risk Factors

The metabolic syndrome is a pool of cardiovascular risk factors, like obesity, hypertension, dyslipidemia, and pre-diabetes. It is also accompanied by low-grade adipose tissue inflammation, which acts as a precursor to insulin resistance and diabetes. Patients with metabolic syndrome have a 1.5–2.5 higher risk of cardiovascular death. Melatonin proved to be useful in managing the metabolic syndrome components mainly through its antioxidant, anti-inflammatory, and immunomodulatory actions (Figure 3). Along with their main use in sleep disorders, melatonin’s analogs have been shown to be beneficial in managing metabolic syndrome components in rodents. Thus, ramelteon lowered blood pressure values and body weight [149], while piromelatine, a newer melatonin analog, improved insulin resistance and stabilized the metabolic profile [150].
Age is also a cardiovascular risk factor and unfortunately, melatonin production progressively decreases with age, in the pineal gland, as well as in the other extra-pineal tissues (heart, liver, spleen). Consequently, cardiovascular protective mechanisms diminish. Inflammation is a key process in aging and in age-related diseases, thus, the term inflammaging was introduced to highlight this strong connection [124]. Melatonin levels were inversely correlated with inflammation in a study on more than 1000 elderly subjects [151], while experimental data suggest that melatonin is also involved in slowing of myocardial aging [152,153].
In elderly individuals, a connection between night-time decreased melatonin secretion, and hypertension has been established [154]. This relationship is not maintained in patients already treated for hypertension. In addition, melatonin reduces nocturnal blood pressure values [155]. Studies in sleep patterns showed susceptibility to hypertension in patients with longstanding insomnia [156].
Nitric oxide (NO) production causes vasodilation and reduction of blood pressure values. Melatonin associated NO increase has been documented in certain circumstances [122,123,157]. However, other studies report the inhibition of NO production, as part of the antioxidant role of melatonin.
Oxidative stress is a major factor in the development of diabetic complications. A potent antioxidant as melatonin has proven beneficial effects on ROS decrease and beta-cell protection in diabetic patients [158]. It also contributes to a decrease in insulin resistance [159] and melatonin supplementation has shown positive results in some clinical studies [158].
Melatonin improves dyslipidemia and body weight in several animal models of hyperadiposity [124]. The reduction of oxidized low-density lipoproteins and triglycerides was also linked to melatonin administration [160]. As previously mentioned, melatonin demonstrates both an anti-inflammatory and antioxidative effect in obesity [161,162]. Part of these properties are attributed to activation of the silent information regulator 1 (SIRT1) [162]. Furthermore, obese women with low melatonin levels have a higher risk of myocardial infarction [163]. Moreover, melatonin is abundant in the gastrointestinal system, approximately 400 times the amount found in the pineal gland [164]. Melatonin reprograms the gut microbiota and improves lipid metabolism [165].

3.4. Vascular and Myocardial Effects

Atherosclerosis (ATS) is the consequence of cholesterol build up and chronic inflammation, in the context of a dysfunctional endothelium. Endothelial dysfunction is partially caused by insufficient NO production via eNOS synthesis, and partially by free radicals such as ROS, significantly involved in vascular disease pathogenesis. Thus, the association between a hypocholesterolemic agent, such as statins and melatonin seem logical. Indeed, a study using both substances proved that melatonin reduces the oxidative stress and improves the statins’ capacity to stimulate eNOS and increase NO production with consequent vasodilation [116].
Melatonin decreases the sympathetic activity and shifts the neurovegetative balance in favor of the parasympathetic system [122]. This is reinforced by the fact that plasma melatonin levels rise as a counter-regulatory mechanism in hypertension triggered by sympathetic overstimulation [123].
Melatonin is also vasoprotective by dampening the inflammation subsequent to the atherosclerotic process (Figure 3) [166]. Through interference with ATS progression, melatonin also possibly contributes to slower development of ischemic heart disease [127]. Moreover, it inhibits calcium accumulation, leucocyte infiltration, and endothelial damage [167,168]. In addition, it has antithrombotic effects through a reduction in platelet reactivity [129].
The cardiac insult produced by experimental myocardial infarction induces an increase in pineal melatonin secretion [169]. This suggests that endogenous melatonin displays a cardioprotective effect (Figure 3). On the other hand, it seems that in patients with coronary artery disease, this mechanism is impaired.
The day/night pattern of melatonin secretion and the inverse correlation to cortisol levels have a connection to the timing of certain adverse events. Myocardial infarction, sudden cardiac death, and even malignant arrhythmias occur early in the morning when melatonin levels are low [170,171]. This relationship could be confounded with the diurnal pattern of sympathetic system activation, which is also influenced by melatonin. It was demonstrated that corticosterone has a dual effect on melatonin production, influenced by the sympathetic system. This effect is based on the stimulation pattern of adrenoceptors, only beta1, or beta1 and alpha1. Thus, sympathetic system activation is necessary for the control of melatonin synthesis in certain conditions and vice-versa [123,172]. A recent study shows norepinephrine (NE) levels accompanying a myocardial infarction are indeed influenced by melatonin administration [173].
Melatonin has demonstrated its potential role in protecting the myocardium against ischemia/reperfusion injury in multiple settings [62,117,120,121,128]. At low concentrations (4 mg/kg/day), it activates several signaling pathways that offer protection against IRI [20,174]. These pathways include JAK-STAT3, and nuclear factor erythroid 2-related factor 2 (Nrf2) (Table 1).
For the coronary artery bypass graft (CABG) patients, a five-day melatonin administration (10 or 20 mg/day) before surgery showed beneficial effects in a dose-dependent manner, as compared to placebo [65,136], recommending melatonin as a preventive method.
Some studies showed that melatonin produces a decrease in the myocardial scar, reduced fibrosis [134], and remodeling after myocardial infarction [130]. In the MARIA (Melatonin Adjunct in the acute myocaRdial Infarction treated with Angioplasty) study on reperfused STEMI patients, melatonin reduces infarct size 40% if administered in the first 2.5 h after symptom onset [125,129]. In addition, melatonin improves reverse remodeling after cardiac resynchronization therapy [132]. Melatonin has lower levels not only in patients with myocardial infarction, but also in dilated cardiomyopathy, and is correlated with cardiac output [133].
Cardiotoxicity is a known side effect of cytostatic therapy. Melatonin protects against doxorubicin-induced cardiotoxicity by decreasing apoptosis and oxidative stress [175].
Insomnia is a common side effect of first and second-generation beta-blockers, a common cardiovascular medication. Newer molecules have been tailored as not to modify the melatonin levels, i.e., nebivolol and carvedilol vs. the other beta-blockers [176,177].

3.5. Neutral and Deleterious Effects

After several promising small animal studies on the effect of melatonin on MI size, a large animal model failed to prove melatonin’s role in MI cardioprotection, when administered prior to myocardial reperfusion either intravenously or intracoronary, thus raising questions on the relevance of melatonin in this context [126]. Whether the lack of efficacy is due to an inappropriate dose, timing or administration route is yet to be investigated. This initial translational fiasco should point to scrupulously testing and tweaking any novel therapy before going further.
Patients with longer-lasting ischemia symptoms (>3.5 h) exhibited an increase in MI size after melatonin administration compared to the placebo group [131]. This effect, as well as lower ejection fraction and higher left ventricle end-diastolic and systolic volumes [125], might be explained by reports on collagen and glycosaminoglycan deposition following melatonin treatment [178,179,180].
Despite numerous clinical and preclinical trials, authors with vast experience in the field of ischemia/reperfusion injury, preconditioning, and cardioprotection do not believe melatonin to be relevant to MI scar reduction in STEMI reperfused patients [137].

4. Melatonin Effects in Neuronal Inflammation and Degenerative Neurological Diseases

Neurological diseases represent the second most common cause of death with a mortality rate of 16.8% worldwide, according to the Global Burden of Disease Study 2015. In the last 25 years, the burden of neurological disorders has increased worldwide. Alzheimer’s disease (AD) and other dementias are one of the four major contributors and account for up to 10% of the disability-adjusted life-years (DALYs) [181].
It was shown that generation or aggravation of various neuropathological conditions are accompanied by a low-grade inflammation and blood–brain barrier impairment, and these conditions could be associated with alteration of sleep and melatonin secretion [182].
The low-grade inflammatory state in the aged brain, known as inflammaging, mainly refers to an excessive production of pro-inflammatory cytokines and proteins such as integrin alpha-M (CD11b), IL-1β, IL-16, and TNF-α and downregulation of agents such as brain-derived neurotrophic factor (BDNF), which were significantly altered in aged-animal models [69]. In lipopolysaccharide (LPS)-treated animals, neuroinflammation is augmented by enhanced oxidative stress, pro-inflammatory cytokines, such as TNF-α, IL-6, IL-1β, and NF-κB phosphorylation.
Various studies showed that exogenous melatonin administration reduces the damaging effects of LPS on nerve cells [183,184]. In a recent review, the close relationship between neuroinflammation and neurodegeneration has been linked by the dysregulation of glial-neuronal communications. Thus, altered signaling promotes glial activation, NF-κB activation and release of proinflammatory cytokines that further determine overactivation of protein kinases and favor amyloid beta and tau proteins accumulation [185]. Together with the aging process, the risk for neurodegenerative diseases such as mild mild cognitive impairment (MCI) and Alzheimer’s disease increases.
Multiple pathological patterns that lead to neurodegeneration have been described, the most popular ones being oxidative stress, mitochondrial dysfunction, low degree of inflammation, excitotoxicity, and impaired pruning mechanisms that lead to accumulation of neurotoxic aggregates such as beta amyloid oligomers [186]. Given the melatonin’s main characteristics, as a circadian rhythm regulator and as a potent antioxidant, multiple studies have investigated its potential role in preventing neurodegeneration. Melatonin secretion is regulated by the suprachiasmatic nucleus, according to environmental light/dark cycles. Its main role is to synchronize the phase and the amplitude of the peripheral biological rhythms. On the other hand, melatonin’s cytoprotective properties allow it to partially reverse the inflammatory damage seen in neurodegenerative disorders and aging [187].
However, notwithstanding the antioxidant and anti-inflammatory characteristics of melatonin, some studies have brought significant data regarding its efficacy against brain injury in humans. Zhao et al. recently demonstrated that melatonin might have a protective effect on cerebral I/R injury in both rats and humans on cerebral I/R-related injury after carotid endarterectomy surgery [39].
Melatonin has been shown to be beneficial in cerebral ischemia. In addition, its antioxidant properties might help slow down neuronal aging, especially when considering its low blood levels in the elderly. In experimental models of Alzheimer’s and Parkinson’s diseases, the neurodegeneration observed is partially prevented by melatonin [188]. Generally, the progression of these neurological conditions and other forms of dementia is associated with low melatonin secretion [189].

4.1. Alzheimer’s Disease

Alzheimer’s disease is the most common neurodegenerative disease of the brain. The pathogenesis of AD is characterized by the presence within the nerve cell of fiber-like deposits of the microtubular tau protein and by the accumulation of amyloid beta. The antioxidant role of melatonin and inhibiting action on oxidative stress was acknowledged in various neurodegenerative disorders such as Alzheimer’s or Parkinson’s disease. In AD, the intracellular accumulation of abnormal proteins become senile plaques consisting of tau filaments and amyloid beta proteins which disrupt the neuronal homeostasis and damage axonal conduction [190]. Oxidative stress causes structural changes in cellular DNA, proteins, and lipids that eventually form damaging protein deposits. Physiologically, melatonin is released directly into the cerebrospinal fluid, thus having a higher concentration in comparison with its blood level. In patients with AD, melatonin concentration in the cerebrospinal fluid is decreased, suggesting that a deficiency of this molecule is correlated with the pathophysiology of this condition [191]. During sleep, the elimination of amyloid beta molecules increases considerably. The disruption of the circadian rhythm in AD, hence, might favor the progression of the disease by slowing the amyloid beta clearance. Moreover, melatonin effectively inhibits amyloid beta synthesis and tau filament formation [192].
Mild cognitive impairment and AD are frequently associated with low melatonin circulating concentrations in animal models [193]. With regard to clinical trials involving melatonin administration to humans, recent studies support the efficacy of melatonin in improving cognitive deterioration and reducing the so-called “sundown” effect [194]. The sundown phenomenon implies the aggravation of specific symptoms such as disorganized thinking, agitation, emotional lability, and attention deficit in the evening.

4.2. Parkinson’s Disease

Parkinson’s disease (PD) comprises the progressive neurodegeneration of dopamine containing neurons located in the pars compacta of the substantia nigra. The pathophysiology of PD is multidimensional, with no existing treatment meant to stop the development of the disease. Current medication is symptomatic and designed to alleviate the motor component of the disease. However, PD affects multiple neurotransmitter systems and involves a non-motor component regarding the autonomic nervous system. Most common symptoms include genitourinary impairment, decreased bowel movements, respiratory and cardiovascular disorders, neuropsychiatric and sleep-related disorders [97]. The pathological patterns which cause the neurodegeneration process in PD are represented by neuro-inflammation, microglial activation, and lymphocytic infiltration. By adjusting the activity of antioxidative enzymes like SOD, mitochondrial complex-I activity, and glutathione (GSH), melatonin decreased oxidative stress in a rat model of PD disease [195]. A study reported that administration of melatonin improved non-motor disorders in patients with PD [196].
The mechanisms through which melatonin helps averting neurodegeneration in Parkinson’s are closely related to its anti-excitotoxic activity and scavenger properties. Melatonin was efficient in inhibiting α-synuclein assembly and in softening kainic acid-induced neurotoxicity [197] and arsenite-induced apoptosis [198]. Moreover, it was proved to block α-synuclein fibril formation and to disrupt pre-synthesized fibrils by hindering protofibril creation and protein secondary structure transitions, while reducing α-synuclein cytotoxicity [198,199].

5. Melatonin as an Adjuvant in Respiratory Distress Caused by SARS-CoV-2

In light of the recent global health events, the search for anti-inflammatory molecules has brought attention to melatonin for its combined anti-inflammatory and antioxidative effects for the treatment of SARS-CoV-2 respiratory infection. As a protector against acute respiratory distress syndrome (ARDS), melatonin proved to be a good adjuvant in respiratory diseases, while also improving the cardiac function in pulmonary arterial hypertension [14]. In a recent report on ramelteon, a melatonin receptor agonist, it was demonstrated that melatonin averts volume induced lung injury in a rat model, by decreasing lung edema, nuclear factor kappa light chain enhancer of activated B cells (NF-κB) activation, iNOS levels, apoptosis, and IL-10 production [71]. Moreover, melatonin prevents lung fibrosis through multiple mechanisms. It interferes with the fibrogenic response, activation of effector cells, elaboration of the extracellular matrix, and its active deposition. Melatonin also diminishes bleomycin-induced lung fibrosis in mice by inhibiting transforming growth factor β 1 (TGF-β1) [200,201] and it is protective in radiation-induced lung injury by suppressing the NOD-like receptor protein 3 (NLRP3) inflammasome [70,202]. Through its antioxidant and anti-inflammatory effects, melatonin is beneficial in pulmonary hypertension [203,204,205], enhances the NO-independent acetylcholine’s vasodilatory effect, decreases the systolic pressure of the right ventricle, and prevents right ventricular hypertrophy [206]. By diminishing TGF-β1 activity, apoptosis, and endoplasmic reticulum stress via Sirtuin-1 upregulation in animal studies, melatonin could be protective in chronic obstructive pulmonary disease (COPD) [72,207]. It also decreases dyspnea and 8-isoprostane in the exhaled air, in humans [208].
COVID 19 is a disease induced by the human body infection with the SARS-CoV-2 virus, which affects especially the lungs, but also the gastrointestinal and nervous system. Respiratory failure in this type of infection is associated with severe hypoxemia. Clinically, two phenotypes were described: L-type and H-type. The L-type is characterized by low elastance (high compliance), low ventilation-to-perfusion ratio (V/Q), low lung weight, low recruitability, whereas the H-type is accompanied by high elastance, high right-to-left shunt, high lung weight, and high lung recruitability [209]. Another important feature of this disease is the presence of venous thrombosis and lung micro-thrombosis [210].
The ARDS present in COVID-19 is induced by the “cytokine storm”. Dendritic and epithelial cells infected with SARS-CoV-2 produce several inflammatory cytokines (IL-1β, Il-2, Il-6, Il-8, TNF-α) and chemoattractants (monocyte chemoattractant protein-1 (MCP-1)/CCL-2 (C-C Motif Chemokine Ligand 2), CCL-3, CCL-5, interferon γ (IFNγ)–induce protein 10 (IP-10)) that attract other neutrophils and monocytes/macrophages resulting in an excessive inflammatory response. These processes lead to alveolar-capillary membrane damage in the lungs [211]. Other mechanisms reported in this type of respiratory failure are vascular leakage, apoptosis of epithelial and endothelial cells.
An intuitive reason for melatonin’s therapeutic effect in COVID-19 is based on its pleiotropic traits (anti-inflammatory, antioxidant, and immunomodulatory) and the observation that children and young patients, known to have higher melatonin levels, show milder forms of disease than the elderly [212]. In a review on the effects of melatonin in COVID 19, Zhang et al. describe its multiple theoretical benefits. Its anti-inflammatory effects are mediated by the proper regulation of Sirtuin-1, suppression of NF-kB, and a reduction of pro-inflammatory cytokines [213]. The antioxidant effect is performed by down-regulating NOS and direct scavenger. The immunomodulatory effect is mediated by the regulation of lymphocytes, macrophages–antigen-presenting cell (APC) activity, and inhibition of NLPR-3 inflammasome.
Melatonin has a vasodilatory effect on the pulmonary circulation and decreases pulmonary blood pressure. However, this effect does not seem to improve the V/Q mismatch with systemic administration in L-type infection, because theoretically, it could increase the hypoxemia [204,205]. Intratracheal administration might be the best choice, to induce a NO-like effect and consequently improve the perfusion in ventilated alveoli. This type of melatonin administration is safe and has proven diminished inflammation via NLPR-3 inflammasome inhibition [214]. Nevertheless, there is a debate if NO administration is really beneficial due to the supposed pulmonary vasoplegia present in the L-type ARDS [209]. Moreover, a lack of pulmonary vessels’ response to NO makes the administration of melatonin probably ineffective.
Melatonin decreases D-dimers levels in stress-induced coagulopathy [215], and has a suppressive effect on burn-induced coagulopathy. However, melatonin was not able to prevent the disseminated intravascular coagulation induced by sepsis, and presumably its administration might not contribute to the prevention of COVID-19 related thrombosis [216].
Melatonin through its anti-inflammatory, antioxidant, and immunomodulatory effects is a perfect candidate as a treatment add-in in COVID-19 but its efficacy is yet to be demonstrated in clinical trials. One certain effect is that it reduces the ICU delirium and this could be a strong recommendation for long-term sedated patients with COVID-19 requiring mechanical ventilation [217].

6. Promising Clinical Targets And Pending Questions

Melatonin has been administered to humans by intranasal, transdermal, oral, transmucosal, and intravenous routes, however, it is not known whether these routes are suitable for pregnancy and if melatonin’s pharmacokinetics is different in pregnant women [57]. These are questions that would need to be addressed if we were to think of melatonin as a potential intervention to prevent pregnancy complications. Importantly, there is need for solid data not only on the safety and pharmacokinetic profile of exogenously administered melatonin during pregnancy but ideally also on the long-term follow-up for the babies exposed in utero.
The Mediterranean diet has been confirmed as protective for individuals at high risk of cardiovascular events [218]. Part of this beneficial effect might be attributed to the melatonin content of some of the components of this type of diet, such as red wine, pistachio nuts, olives, fish, and seasonal fruits [219,220]. Concerning exogenous melatonin administration, a relevant question is whether dietary melatonin is enough or a higher dose would be necessary for an efficient cardioprotection. Furthermore, the issues regarding the ideal administration route and timing are still unresolved.
Is there a specific endogenous level below which melatonin becomes inefficient as a protective mechanism and an exogenous supplementation might be beneficial? If so, what is that level, what are the efficient doses and when should they be implemented? All these questions are still open-ended.
Additionally, melatonin analogues (ramelteon, agomelatine, tasimelteon, piromelatine), with a long-lasting effect are currently being explored in clinical trials. Interestingly, the doses used for these analogues are significantly higher than for melatonin. Thus, maybe it is justified to also test higher doses of melatonin (50–100 mg/day) in order to assess its role in cardiovascular prevention [124].
Although very promising in preclinical trials, melatonin seems to fail to deliver clinically. Melatonin failed the translational trial in established cardiovascular disease [137], but nevertheless acts on several cardiovascular disease risk factors (diabetes, obesity, hypertension, dyslipidemia). Should this be a clue it ought to be included in the prevention guidelines for patients at risk of developing cardiovascular diseases?
As both the neurodegenerative disorders and cardiovascular diseases represent a potential target for melatonin’s beneficial prophylactic effects, maybe a joint guideline for patients at risk would be of clinical use.
Fortunately, clinical trials involving melatonin administration do not cease to appear. Even in higher doses, melatonin proved to have a satisfying safe profile and elicited a nontoxic effect. Hence, its applications as a neuronal protector were exploited both in acute disease, especially brain ischemia and in degenerative conditions, such as Alzheimer’s, Parkinson’s, or Huntington’s diseases. The pathological accumulation of intracellular proteins that later form senile plaques in AD is reduced by melatonin. Furthermore, its dual dimension, as a chrono-biological regulator and antioxidant was critical in improving cognitive deterioration and reducing the “sundown” effect in AD. Its ability to adjust antioxidative enzymes rendered a lowering of neuronal oxidative stress in PD in animal models, whilst yielding favorable results in human trials with respect to non-motor symptoms in PD.
Here and now, an important question is whether melatonin will prove itself useful in managing the cytokine storm associated to COVID 19. A better mechanistic understanding of the disease will either support the initiation of research on the matter or discourage it altogether. At the moment, the only clear recommendation for the use of melatonin is the association of 6–12 mg at night, according to the recently released MATH+ protocol (Methylprednisolone, Ascorbic acid, Thiamine, Heparine + melatonine, zinc, vitamin D, famotidine, magnesium) [221].
As more and more data accumulate around the subject of melatonin and its clinical use, it becomes abundantly clear that even if some useful properties are “lost in translation”, there are still a lot of questions that, when answered, will perhaps be a breakthrough.

Author Contributions

Conceptualization, A.-M.Z., D.M.C., M.R.P.; methodology, all authors; writing—original draft preparation, all authors; writing—review and editing, all authors; visualization, A.-M.Z., L.Z., M.R.P.; supervision, A.-M.Z., L.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Acuña-Castroviejo, D.; Escames, G.; Venegas, C.; Díaz-Casado, M.E.; Lima-Cabello, E.; López, L.C.; Rosales-Corral, S.; Tan, D.X.; Reiter, R.J. Extrapineal Melatonin: Sources, Regulation, and Potential Functions. Cell. Mol. Life Sci. 2014, 71, 2997–3025. [Google Scholar] [CrossRef] [PubMed]
  2. Jiki, Z.; Lecour, S.; Nduhirabandi, F. Cardiovascular Benefits of Dietary Melatonin: A Myth or a Reality? Front. Physiol. 2018, 528, 1–17. [Google Scholar] [CrossRef] [PubMed]
  3. Venegas, C.; García, J.A.; Escames, G.; Ortiz, F.; López, A.; Doerrier, C.; García-Corzo, L.; López, L.C.; Reiter, R.J.; Acuña-Castroviejo, D. Extrapineal Melatonin: Analysis of Its Subcellular Distribution and Daily Fluctuations. J. Pineal Res. 2012, 52, 217–227. [Google Scholar] [CrossRef] [PubMed]
  4. Scholtens, R.M.; Van Munster, B.C.; Van Kempen, M.F.; De Rooij, S.E.J.A. Physiological Melatonin Levels in Healthy Older People: A Systematic Review. J. Psychosom. Res. 2016, 86, 20–27. [Google Scholar] [CrossRef] [PubMed]
  5. Hickie, I.B.; Rogers, N.L. Novel Melatonin-Based Therapies: Potential Advances in the Treatment of Major Depression. Lancet 2011, 378, 621–631. [Google Scholar] [CrossRef]
  6. 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]
  7. Poeggeler, B.; Reiter, R.J.; Tan, D.-X.; Chen, L.-D.; Manchester, L.C. Melatonin, Hydroxyl Radical-mediated Oxidative Damage, and Aging: A Hypothesis. J. Pineal Res. 1993, 14, 151–168. [Google Scholar] [CrossRef]
  8. Galano, A.; Tan, D.X.; Reiter, R.J. Melatonin: A Versatile Protector against Oxidative DNA Damage. Molecules 2018, 23, 530. [Google Scholar] [CrossRef] [Green Version]
  9. Álvarez-Diduk, R.; Galano, A.; Tan, D.X.; Reiter, R.J. N-Acetylserotonin and 6-Hydroxymelatonin against Oxidative Stress: Implications for the Overall Protection Exerted by Melatonin. J. Phys. Chem. B 2015, 119, 8535–8543. [Google Scholar] [CrossRef] [Green Version]
  10. Tan, D.X.; Hardeland, R.; Manchester, L.C.; Paredes, S.D.; Korkmaz, A.; Sainz, R.M.; Mayo, J.C.; Fuentes-Broto, L.; Reiter, R.J. The Changing Biological Roles of Melatonin during Evolution: From an Antioxidant to Signals of Darkness, Sexual Selection and Fitness. Biol. Rev. 2010, 85, 607–623. [Google Scholar] [CrossRef]
  11. Tan, D.X.; Manchester, L.C.; Qin, L.; Reiter, R.J. Melatonin: A Mitochondrial Targeting Molecule Involving Mitochondrial Protection and Dynamics. Int. J. Mol. Sci. 2016, 17, 2124. [Google Scholar] [CrossRef] [PubMed]
  12. Huo, X.; Wang, C.; Yu, Z.; Peng, Y.; Wang, S.; Feng, S.; Zhang, S.; Tian, X.; Sun, C.; Liu, K.; et al. Human Transporters, PEPT1/2, Facilitate Melatonin Transportation into Mitochondria of Cancer Cells: An Implication of the Therapeutic Potential. J. Pineal Res. 2017, 62, e12390. [Google Scholar] [CrossRef] [PubMed]
  13. Mayo, J.C.; Sainz, R.M.; González-Menéndez, P.; Hevia, D.; Cernuda-Cernuda, R. Melatonin Transport into Mitochondria. Cell. Mol. Life Sci. 2017, 74, 3927–3940. [Google Scholar] [CrossRef] [PubMed]
  14. Nabavi, S.M.; Nabavi, S.F.; Sureda, A.; Xiao, J.; Dehpour, A.R.; Shirooie, S.; Silva, A.S.; Baldi, A.; Khan, H.; Daglia, M. Anti-Inflammatory Effects of Melatonin: A Mechanistic Review. Crit. Rev. Food Sci. Nutr. 2018, 59, S4–S16. [Google Scholar] [CrossRef] [PubMed]
  15. El-Shenawy, S.M.; Abdel-Salam, O.M.E.; Baiuomy, A.R.; El-Batran, S.; Arbid, M.S. Studies on the Anti-Inflammatory and Anti-Nociceptive Effects of Melatonin in the Rat. Pharm. Res. 2002, 46, 235–243. [Google Scholar] [CrossRef]
  16. Carrasco, C.; Marchena, A.M.; Holguín-Arévalo, M.S.; Martín-Partido, G.; Rodríguez, A.B.; Paredes, S.D.; Pariente, J.A. Anti-Inflammatory Effects of Melatonin in a Rat Model of Caerulein-Induced Acute Pancreatitis. Cell Biochem. Funct. 2013, 31, 585–590. [Google Scholar] [CrossRef]
  17. Liu, Z.; Gan, L.; Xu, Y.; Luo, D.; Ren, Q.; Wu, S.; Sun, C. Melatonin Alleviates Inflammasome-Induced Pyroptosis through Inhibiting NF-ΚB/GSDMD Signal in Mice Adipose Tissue. J. Pineal Res. 2017, 63, e12414. [Google Scholar] [CrossRef]
  18. Deng, W.G.; Tang, S.T.; Tseng, H.P.; Wu, K.K. Melatonin Suppresses Macrophage Cyclooxygenase-2 and Inducible Nitric Oxide Synthase Expression by Inhibiting P52 Acetylation and Binding. Blood 2006, 108, 518–524. [Google Scholar] [CrossRef]
  19. Yang, Y.; Sun, Y.; Yi, W.; Li, Y.; Fan, C.; Xin, Z.; Jiang, S.; Di, S.; Qu, Y.; Reiter, R.J.; et al. A Review of Melatonin as a Suitable Antioxidant against Myocardial Ischemia-Reperfusion Injury and Clinical Heart Diseases. J. Pineal Res. 2014, 57, 357–366. [Google Scholar] [CrossRef]
  20. Opie, L.H.; Lecour, S. Melatonin Has Multiorgan Effects. Eur. Heart J. Cardiovasc. Pharm. 2016, 2, 258–265. [Google Scholar] [CrossRef] [Green Version]
  21. Yang, C.H.; Xu, J.H.; Ren, Q.C.; Duan, T.; Mo, F.; Zhang, W. Melatonin Promotes Secondary Hair Follicle Development of Early Postnatal Cashmere Goat and Improves Cashmere Quantity and Quality by Enhancing Antioxidant Capacity and Suppressing Apoptosis. J. Pineal Res. 2019, 67, e12569. [Google Scholar] [CrossRef]
  22. Carbajo-Pescador, S.; Ordoñez, R.; Benet, M.; Jover, R.; García-Palomo, A.; Mauriz, J.L.; González-Gallego, J. Inhibition of VEGF Expression through Blockade of Hif1α and STAT3 Signalling Mediates the Anti-Angiogenic Effect of Melatonin in HepG2 Liver Cancer Cells. Br. J. Cancer 2013, 109, 83–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Wang, Y.; Wang, P.; Zheng, X.; Du, X. Therapeutic Strategies of Melatonin in Cancer Patients: A Systematic Review and Meta-Analysis. OncoTargets Ther. 2018, 11, 7895–7908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Hunter, C.M.; Figueiro, M.G. Measuring Light at Night and Melatonin Levels in Shift Workers: A Review of the Literature. Biol. Res. Nurs. 2017, 19, 365–374. [Google Scholar] [CrossRef] [PubMed]
  25. Laudon, M.; Frydman-Marom, A. Therapeutic Effects of Melatonin Receptor Agonists on Sleep and Comorbid Disorders. Int. J. Mol. Sci. 2014, 15, 15924–15950. [Google Scholar] [CrossRef] [Green Version]
  26. Liu, J.; Clough, S.J.; Hutchinson, A.J.; Adamah-Biassi, E.B.; Popovska-Gorevski, M.; Dubocovich, M.L. MT 1 and MT 2 Melatonin Receptors: A Therapeutic Perspective. Annu. Rev. Pharmacol. Toxicol. 2016, 56, 361–383. [Google Scholar] [CrossRef] [Green Version]
  27. Atkin, T.; Comai, S.; Gobbi, G. Drugs for Insomnia beyond Benzodiazepines: Pharmacology, Clinical Applications, and Discovery. Pharmacol. Rev. 2018, 70, 197–245. [Google Scholar] [CrossRef]
  28. Andersen, L.P.H.; Gögenur, I.; Rosenberg, J.; Reiter, R.J. The Safety of Melatonin in Humans. Clin. Drug Investig. 2015, 36, 169–175. [Google Scholar] [CrossRef]
  29. Zagrean, A.-M.; Chitimus, D.M.; Badiu, C.; Panaitescu, A.M.; Peltecu, G.; Zagrean, L. The Pineal Gland and Its Function in Pregnancy and Lactation. In Maternal-Fetal and Neonatal Endocrinology; Elsevier: Amsterdam, The Netherlands, 2020; pp. 15–37. [Google Scholar] [CrossRef]
  30. Nakamura, Y.; Tamura, H.; Kashida, S.; Takayama, H.; Yamagata, Y.; Karube, A.; Sugino, N.; Kato, H. Changes of Serum Melatonin Level and Its Relationship to Feto-Placental Unit during Pregnancy. J. Pineal Res. 2001, 30, 29–33. [Google Scholar] [CrossRef]
  31. Tamura, H.; Takayama, H.; Nakamura, Y.; Reiter, R.J.; Sugino, N. Fetal/Placental Regulation of Maternal Melatonin in Rats. J. Pineal Res. 2008, 44, 335–340. [Google Scholar] [CrossRef]
  32. Reik, W. Stability and Flexibility of Epigenetic Gene Regulation in Mammalian Development. Nature 2007, 447, 425–432. [Google Scholar] [CrossRef] [PubMed]
  33. Santos, F.; Hendrich, B.; Reik, W.; Dean, W. Dynamic Reprogramming of DNA Methylation in the Early Mouse Embryo. Dev. Biol. 2002, 241, 172–182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Guo, H.; Zhu, P.; Yan, L.; Li, R.; Hu, B.; Lian, Y.; Yan, J.; Ren, X.; Lin, S.; Li, J.; et al. The DNA Methylation Landscape of Human Early Embryos. Nature 2014, 511, 606–610. [Google Scholar] [CrossRef]
  35. Verma, N.; Pan, H.; Doré, L.C.; Shukla, A.; Li, Q.V.; Pelham-Webb, B.; Teijeiro, V.; González, F.; Krivtsov, A.; Chang, C.J.; et al. TET Proteins Safeguard Bivalent Promoters from de Novo Methylation in Human Embryonic Stem Cells. Nat. Genet. 2018, 50, 83–95. [Google Scholar] [CrossRef]
  36. 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]
  37. Koh, P.O. Melatonin Regulates the Calcium-Buffering Proteins, Parvalbumin and Hippocalcin, in Ischemic Brain Injury. J. Pineal Res. 2012, 53, 358–365. [Google Scholar] [CrossRef] [PubMed]
  38. Cuzzocrea, S.; Costantino, G.; Gitto, E.; Mazzon, E.; Fulia, F.; Serraino, I.; Cordaro, S.; Barberi, I.; De Sarro, A.; Caputi, A.P. Protective Effects of Melatonin in Ischemic Brain Injury. J. Pineal Res. 2000, 29, 217–227. [Google Scholar] [CrossRef] [PubMed]
  39. Zhao, Z.; Lu, C.; Li, T.; Wang, W.; Ye, W.; Zeng, R.; Ni, L.; Lai, Z.; Wang, X.; Liu, C. The Protective Effect of Melatonin on Brain Ischemia and Reperfusion in Rats and Humans: In Vivo Assessment and a Randomized Controlled Trial. J. Pineal Res. 2018, 65, e12521. [Google Scholar] [CrossRef] [PubMed]
  40. Phillipson, O.T. Alpha-Synuclein, Epigenetics, Mitochondria, Metabolism, Calcium Traffic, & Circadian Dysfunction in Parkinson’s Disease. An Integrated Strategy for Management. Ageing Res. Rev. 2017, 40, 149–167. [Google Scholar] [CrossRef]
  41. Sinha, K.; Das, J.; Pal, P.B.; Sil, P.C. Oxidative Stress: The Mitochondria-Dependent and Mitochondria-Independent Pathways of Apoptosis. Arch. Toxicol. 2013, 87, 1157–1180. [Google Scholar] [CrossRef]
  42. Chandra, J.; Samali, A.; Orrenius, S. Triggering and Modulation of Apoptosis by Oxidative Stress. Free Radic. Biol. Med. 2000, 29, 323–333. [Google Scholar] [CrossRef]
  43. Zhang, H.M.; Zhang, Y. Melatonin: A Well-Documented Antioxidant with Conditional pro-Oxidant Actions. J. Pineal Res. 2014, 57, 131–146. [Google Scholar] [CrossRef] [PubMed]
  44. Yang, Y.; Jiang, S.; Dong, Y.; Fan, C.; Zhao, L.; Yang, X.; Li, J.; Di, S.; Yue, L.; Liang, G.; et al. Melatonin Prevents Cell Death and Mitochondrial Dysfunction via a SIRT1-Dependent Mechanism during Ischemic-Stroke in Mice. J. Pineal Res. 2015, 58, 61–70. [Google Scholar] [CrossRef] [PubMed]
  45. Giusti, P.; Upartiti, M.; Franceschini, D.; Schiavo, N.; Floreani, M.; Manev, H. Neuroprotection by Melatonin from Kainate-induced Excitotoxicity in Rats. FASEB J. 1996, 10, 891–896. [Google Scholar] [CrossRef] [Green Version]
  46. Allen, C.L.; Bayraktutan, U. Oxidative Stress and Its Role in the Pathogenesis of Ischaemic Stroke. Int. J. Stroke 2009, 4, 461–470. [Google Scholar] [CrossRef]
  47. Cazevieille, C.; Safa, R.; Osborne, N.N. Melatonin Protects Primary Cultures of Rat Cortical Neurones from NMDA Excitotoxicity and Hypoxia/Reoxygenation. Brain Res. 1997, 768, 120–124. [Google Scholar] [CrossRef]
  48. Martín, M.; Macías, M.; Escames, G.; León, J.; Acuña-Castroviejo, D. Melatonin but Not Vitamins C and E Maintains Glutathione Homeostasis in T-butyl Hydroperoxide-induced Mitochondrial Oxidative Stress. FASEB J. 2000, 14, 1677–1679. [Google Scholar] [CrossRef]
  49. Loren, P.; Sánchez, R.; Arias, M.E.; Felmer, R.; Risopatrón, J.; Cheuquemán, C. Melatonin Scavenger Properties against Oxidative and Nitrosative Stress: Impact on Gamete Handling and In Vitro Embryo Production in Humans and Other Mammals. Int. J. Mol. Sci. 2017, 18, 1119. [Google Scholar] [CrossRef] [Green Version]
  50. García, J.J.; Lõpez-Pingarrõn, L.; Almeida-Souza, P.; Tres, A.; Escudero, P.; García-Gil, F.A.; Tan, D.X.; Reiter, R.J.; Ramírez, J.M.; Bernal-Pérez, M. Protective Effects of Melatonin in Reducing Oxidative Stress and in Preserving the Fluidity of Biological Membranes: A Review. J. Pineal Res. 2014, 56, 225–237. [Google Scholar] [CrossRef]
  51. Reiter, R.J.; Tan, D.X.; Korkmaz, A.; Rosales-Corral, S.A. Melatonin and Stable Circadian Rhythms Optimize Maternal, Placental and Fetal Physiology. Hum. Reprod. Update 2014, 20, 293–307. [Google Scholar] [CrossRef] [Green Version]
  52. Welin, A.K.; Svedin, P.; Lapatto, R.; Sultan, B.; Hagberg, H.; Gressens, P.; Kjellmer, I.; Mallard, C. Melatonin Reduces Inflammation and Cell Death in White Matter in the Mid-Gestation Fetal Sheep Following Umbilical Cord Occlusion. Pediatr. Res. 2007, 61, 153–158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Wang, X.; Zhu, S.; Pei, Z.; Drozda, M.; Stavrovskaya, I.G.; Del Signore, S.J.; Cormier, K.; Shimony, E.M.; Wang, H.; Ferrante, R.J.; et al. Inhibitors of Cytochrome c Release with Therapeutic Potential for Huntington’s Disease. J. Neurosci. 2008, 28, 9473–9485. [Google Scholar] [CrossRef]
  54. Jan, J.E.; Wasdell, M.B.; Freeman, R.D.; Bax, M. Evidence Supporting the Use of Melatonin in Short Gestation Infants. J. Pineal Res. 2007, 42, 22–27. [Google Scholar] [CrossRef] [PubMed]
  55. Schernhammer, E.S.; Kroenke, C.H.; Dowsett, M.; Folkerd, E.; Hankinson, S.E. Urinary 6-Sulfatoxymelatonin Levels and Their Correlations with Lifestyle Factors and Steroid Hormone Levels. J. Pineal Res. 2006, 40, 116–124. [Google Scholar] [CrossRef] [PubMed]
  56. Tucker, J.; McGuire, W. ABC of Preterm Birth Epidemiology of Preterm Birth. BMJ 2004, 329, 675–678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Zetner, D.; Andersen, L.P.H.; Rosenberg, J. Pharmacokinetics of Alternative Administration Routes of Melatonin: A Systematic Review. Drug Res. 2015, 66, 169–173. [Google Scholar] [CrossRef] [Green Version]
  58. Robertson, N.J.; Faulkner, S.; Fleiss, B.; Bainbridge, A.; Andorka, C.; Price, D.; Powell, E.; Lecky-Thompson, L.; Thei, L.; Chandrasekaran, M.; et al. Melatonin Augments Hypothermic Neuroprotection in a Perinatal Asphyxia Model. Brain 2013, 136, 90–105. [Google Scholar] [CrossRef]
  59. Petrosillo, G.; Colantuono, G.; Moro, N.; Ruggiero, F.M.; Tiravanti, E.; Di Venosa, N.; Fiore, T.; Paradies, G. Melatonin Protects against Heart Ischemia-Reperfusion Injury by Inhibiting Mitochondrial Permeability Transition Pore Opening. Am. J. Physiol. Circ. Physiol. 2009, 297, H1487–H1493. [Google Scholar] [CrossRef] [Green Version]
  60. Paradies, G.; Paradies, V.; Ruggiero, F.M.; Petrosillo, G. Protective Role of Melatonin in Mitochondrial Dysfunction and Related Disorders. Arch. Toxicol. 2015, 89, 923–939. [Google Scholar] [CrossRef]
  61. Petrosillo, G.; Moro, N.; Ruggiero, F.M.; Paradies, G. Melatonin Inhibits Cardiolipin Peroxidation in Mitochondria and Prevents the Mitochondrial Permeability Transition and Cytochrome c Release. Free Radic. Biol. Med. 2009, 47, 969–974. [Google Scholar] [CrossRef]
  62. Yang, Y.; Duan, W.; Jin, Z.; Yi, W.; Yan, J.; Zhang, S.; Wang, N.; Liang, Z.; Li, Y.; Chen, W.; et al. JAK2/STAT3 Activation by Melatonin Attenuates the Mitochondrial Oxidative Damage Induced by Myocardial Ischemia/Reperfusion Injury. J. Pineal Res. 2013, 55, 275–286. [Google Scholar] [CrossRef] [PubMed]
  63. Yeung, H.M.; Hung, M.W.; Fung, M.L. Melatonin Ameliorates Calcium Homeostasis in Myocardial and Ischemia-Reperfusion Injury in Chronically Hypoxic Rats. J. Pineal Res. 2008, 45, 373–382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Liu, Y.; Li, L.N.; Guo, S.; Zhao, X.Y.; Liu, Y.Z.; Liang, C.; Tu, S.; Wang, D.; Li, L.; Dong, J.Z.; et al. Melatonin Improves Cardiac Function in a Mouse Model of Heart Failure with Preserved Ejection Fraction. Redox Biol. 2018, 18, 211–221. [Google Scholar] [CrossRef]
  65. Javanmard, S.H.; Ziaei, A.; Ziaei, S.; Ziaei, E.; Mirmohammad-Sadeghi, M. The Effect of Preoperative Melatonin on Nuclear Erythroid 2-Related Factor 2 Activation in Patients Undergoing Coronary Artery Bypass Grafting Surgery. Oxidative Med. Cell. Longev. 2013, 2013, 1–6. [Google Scholar] [CrossRef] [PubMed]
  66. Ortiz, F.; García, J.A.; Acuña-Castroviejo, D.; Doerrier, C.; Lõpez, A.; Venegas, C.; Volt, H.; Luna-Sánchez, M.; Lõpez, L.C.; Escames, G. The Beneficial Effects of Melatonin against Heart Mitochondrial Impairment during Sepsis: Inhibition of i NOS and Preservation of n NOS. J. Pineal Res. 2014, 56, 71–81. [Google Scholar] [CrossRef]
  67. Veneroso, C.; Tuñón, M.J.; González-Gallego, J.; Collado, P.S. Melatonin Reduces Cardiac Inflammatory Injury Induced by Acute Exercise. J. Pineal Res. 2009, 47, 184–191. [Google Scholar] [CrossRef]
  68. Oliveira, L.G.R.; Kuehn, C.C.; Dos Santos, C.D.; Miranda, M.A.; Da Costa, C.M.B.; Mendonça, V.J.; Do Prado, J.C. Protective Actions of Melatonin against Heart Damage during Chronic Chagas Disease. Acta Trop. 2013, 128, 652–658. [Google Scholar] [CrossRef]
  69. Permpoonputtana, K.; Tangweerasing, P.; Mukda, S.; Boontem, P.; Nopparat, C.; Govitrapong, P. Long-Term Administration of Melatonin Attenuates Neuroinflammation in the Aged Mouse Brain. EXCLI J. 2018, 17, 634–646. [Google Scholar] [CrossRef]
  70. Wu, X.; Ji, H.; Wang, Y.; Gu, C.; Gu, W.; Hu, L.; Zhu, L. Melatonin Alleviates Radiation-Induced Lung Injury via Regulation of MiR-30e/NLRP3 Axis. Oxidative Med. Cell. Longev. 2019, 2019, 1–14. [Google Scholar] [CrossRef] [Green Version]
  71. Wu, G.C.; Peng, C.K.; Liao, W.I.; Pao, H.P.; Huang, K.L.; Chu, S.J. Melatonin Receptor Agonist Protects against Acute Lung Injury Induced by Ventilator through Up-Regulation of IL-10 Production. Respir. Res. 2020, 21, 1–17. [Google Scholar] [CrossRef] [Green Version]
  72. Shin, N.R.; Ko, J.W.; Kim, J.C.; Park, G.; Kim, S.H.; Kim, M.S.; Kim, J.S.; Shin, I.S. Role of Melatonin as an SIRT1 Enhancer in Chronic Obstructive Pulmonary Disease Induced by Cigarette Smoke. J. Cell. Mol. Med. 2020, 24, 1151–1156. [Google Scholar] [CrossRef] [Green Version]
  73. Zagrean, A.-M.; Chitimus, D.M.; Paslaru, F.G.; Voiculescu, S.E.; Badiu, C.; Peltecu, G.; Panaitescu, A.M. Pineal Gland Disorders and Circadian Rhythm Alterations in Pregnancy and Lactation. In Maternal-Fetal and Neonatal Endocrinology; Elsevier: Amsterdam, The Netherlands, 2020; pp. 241–257. [Google Scholar] [CrossRef]
  74. Zhu, J.L.; Hjollund, N.H.; Olsen, J. Shift Work, Duration of Pregnancy, and Birth Weight: The National Birth Cohort in Denmark. Am. J. Obstet. Gynecol. 2004, 191, 285–291. [Google Scholar] [CrossRef] [PubMed]
  75. Wierrani, F.; Grin, W.; Hlawka, B.; Kroiss, A.; Grünberger, W. Elevated Serum Melatonin Levels during Human Late Pregnancy and Labour. J. Obstet. Gynaecol. 1997, 17, 449–451. [Google Scholar] [CrossRef]
  76. Reiter, R.J.; Tamura, H.; Tan, D.X.; Xu, X.Y. Melatonin and the Circadian System: Contributions to Successful Female Reproduction. Fertil. Steril. 2014, 102, 321–328. [Google Scholar] [CrossRef]
  77. Okatani, Y.; Hayashi, K.; Sagara, Y. Effect of Estrogen on Melatonin Synthesis in Female Peripubertal Rats as Related to Adenylate Cyclase Activity. J. Pineal Res. 1998, 25, 245–250. [Google Scholar] [CrossRef] [PubMed]
  78. Kennaway, D.J. Programming of the Fetal Suprachiasmatic Nucleus and Subsequent Adult Rhythmicity. Trends Endocrinol. Metab. 2002, 13, 398–402. [Google Scholar] [CrossRef]
  79. Conway, S.; Drew, J.E.; Canning, S.J.; Barrett, P.; Jockers, R.; Strosberg, A.D.; Guardiola-Lemaitre, B.; Delagrange, P.; Morgan, P.J. Identification of Mel1a Melatonin Receptors in the Human Embryonic Kidney Cell Line HEK293: Evidence of G Protein-Coupled Melatonin Receptors Which Do Not Mediate the Inhibition of Stimulated Cyclic AMP Levels. FEBS Lett. 1997, 407, 121–126. [Google Scholar] [CrossRef]
  80. Yu, X.; Li, Z.; Zheng, H.; Ho, J.; Chan, M.T.V.; Wu, W.K.K. Protective Roles of Melatonin in Central Nervous System Diseases by Regulation of Neural Stem Cells. Cell Prolif. 2017, 50, e12323. [Google Scholar] [CrossRef] [Green Version]
  81. Hsu, C.N.; Tain, Y.L. Light and Circadian Signaling Pathway in Pregnancy: Programming of Adult Health and Disease. Int. J. Mol. Sci. 2020, 21, 2232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Mahoney, M.M. Shift Work, Jet Lag, and Female Reproduction. Int. J. Endocrinol. 2010, 2010, 813764. [Google Scholar] [CrossRef]
  83. Hardeland, R.; Madrid, J.A.; Tan, D.-X.; Reiter, R.J. Melatonin, the Circadian Multioscillator System and Health: The Need for Detailed Analyses of Peripheral Melatonin Signaling. J. Pineal Res. 2012, 52, 139–166. [Google Scholar] [CrossRef] [PubMed]
  84. Kaur, S.; Teoh, A.N.; Shukri, N.H.M.; Shafie, S.R.; Bustami, N.A.; Takahashi, M.; Lim, P.J.; Shibata, S. Circadian Rhythm and Its Association with Birth and Infant Outcomes: Research Protocol of a Prospective Cohort Study. BMC Pregnancy Childbirth 2020, 20, 96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Katzer, D.; Pauli, L.; Mueller, A.; Reutter, H.; Reinsberg, J.; Fimmers, R.; Bartmann, P.; Bagci, S. Melatonin Concentrations and Antioxidative Capacity of Human Breast Milk According to Gestational Age and the Time of Day. J. Hum. Lact. 2016, 32, NP105–NP110. [Google Scholar] [CrossRef] [PubMed]
  86. Qin, Y.; Shi, W.; Zhuang, J.; Liu, Y.; Tang, L.; Bu, J.; Sun, J.; Bei, F. Variations in Melatonin Levels in Preterm and Term Human Breast Milk during the First Month after Delivery. Sci. Rep. 2019, 9, 1–5. [Google Scholar] [CrossRef]
  87. Cai, C.; Vandermeer, B.; Khurana, R.; Nerenberg, K.; Featherstone, R.; Sebastianski, M.; Davenport, M.H. The Impact of Occupational Shift Work and Working Hours during Pregnancy on Health Outcomes: A Systematic Review and Meta-Analysis. Am. J. Obstet. Gynecol. 2019, 221, 563–576. [Google Scholar] [CrossRef]
  88. Stevens, R.G.; Schernhammer, E. Epidemiology of Urinary Melatonin in Women and Its Relation to Other Hormones and Night Work. Cancer Epidemiol. Biomark. Prev. 2005, 14, 551. [Google Scholar] [CrossRef] [Green Version]
  89. Lee, J.Y.; Song, H.; Dash, O.; Park, M.; Shin, N.E.; McLane, M.W.; Lei, J.; Hwang, J.Y.; Burd, I. Administration of Melatonin for Prevention of Preterm Birth and Fetal Brain Injury Associated with Premature Birth in a Mouse Model. Am. J. Reprod. Immunol. 2019, 82, e13151. [Google Scholar] [CrossRef]
  90. Jahnke, G.; Marr, M.; Myers, C.; Wilson, R.; Travlos, G.; Price, C. Maternal and Developmental Toxicity Evaluation of Melatonin Administered Orally to Pregnant Sprague-Dawley Rats. Toxicol. Sci. 1999, 50, 271–279. [Google Scholar] [CrossRef]
  91. Freeman, M.P.; Sosinsky, A.Z.; Moustafa, D.; Viguera, A.C.; Cohen, L.S. Supplement Use by Women during Pregnancy: Data from the Massachusetts General Hospital National Pregnancy Registry for Atypical Antipsychotics. Arch. Womens Ment. Health 2016, 19, 437–441. [Google Scholar] [CrossRef]
  92. Karumanchi, S.A.; Granger, J.P. Preeclampsia and Pregnancy-Related Hypertensive Disorders. Hypertension 2016, 67, 238–242. [Google Scholar] [CrossRef]
  93. Zeng, K.; Gao, Y.; Wan, J.; Tong, M.; Lee, A.C.; Zhao, M.; Chen, Q. The Reduction in Circulating Levels of Melatonin May Be Associated with the Development of Preeclampsia. J. Hum. Hypertens. 2016, 30, 666–671. [Google Scholar] [CrossRef] [PubMed]
  94. Hobson, S.R.; Gurusinghe, S.; Lim, R.; Alers, N.O.; Miller, S.L.; Kingdom, J.C.; Wallace, E.M. Melatonin Improves Endothelial Function in Vitro and Prolongs Pregnancy in Women with Early-Onset Preeclampsia. J. Pineal Res. 2018, 65, e12508. [Google Scholar] [CrossRef] [PubMed]
  95. Hannan, N.J.; Binder, N.K.; Beard, S.; Nguyen, T.V.; Kaitu’u-Lino, T.J.; Tong, S. Melatonin Enhances Antioxidant Molecules in the Placenta, Reduces Secretion of Soluble Fms-like Tyrosine Kinase 1 (SFLT) from Primary Trophoblast but Does Not Rescue Endothelial Dysfunction: An Evaluation of Its Potential to Treat Preeclampsia. PLoS ONE 2018, 13, e0187082. [Google Scholar] [CrossRef] [PubMed]
  96. Hobson, S.R.; Lim, R.; Gardiner, E.E.; Alers, N.O.; Wallace, E.M. Phase I Pilot Clinical Trial of Antenatal Maternally Administered Melatonin to Decrease the Level of Oxidative Stress in Human Pregnancies Affected by Pre-Eclampsia (PAMPR): Study Protocol. BMJ Open 2013, 3, e003788. [Google Scholar] [CrossRef] [Green Version]
  97. Nutt, J.G.; Bohnen, N.I. Non-Dopaminergic Therapies. J. Park. Dis. 2018, 8, S73–S78. [Google Scholar] [CrossRef] [Green Version]
  98. Cetin, I.; Alvino, G. Intrauterine Growth Restriction: Implications for Placental Metabolism and Transport. A Review. Placenta 2009, 30, 77–82. [Google Scholar] [CrossRef]
  99. Miller, S.L.; Yan, E.B.; Castillo-Meléndez, M.; Jenkin, G.; Walker, D.W. Melatonin Provides Neuroprotection in the Late-Gestation Fetal Sheep Brain in Response to Umbilical Cord Occlusion. Dev. Neurosci. 2005, 27, 200–210. [Google Scholar] [CrossRef]
  100. Wang, L.; Feng, C.; Zheng, X.; Guo, Y.; Zhou, F.; Shan, D.; Liu, X.; Kong, J. Plant Mitochondria Synthesize Melatonin and Enhance the Tolerance of Plants to Drought Stress. J. Pineal Res. 2017, 63, e12429. [Google Scholar] [CrossRef]
  101. Voiculescu, S.E.; Zygouropoulos, N.; Zahiu, C.D.; Zagrean, A.M. Role of Melatonin in Embryo Fetal Development. J. Med. Life. 2014, 7, 488–492. [Google Scholar]
  102. Rodriguez-Osorio, N.; Kim, I.J.; Wang, H.; Kaya, A.; Memili, E. Melatonin Increases Cleavage Rate of Porcine Preimplantation Embryos in Vitro. J. Pineal Res. 2007, 43, 283–288. [Google Scholar] [CrossRef]
  103. Sampaio, R.V.; Conceição, D.S.B.; Miranda, M.S.; Sampaio, L.d.F.S.; Ohashi, O.M. MT3 Melatonin Binding Site, MT1 and MT2 Melatonin Receptors Are Present in Oocyte, but Only MT1 Is Present in Bovine Blastocyst Produced In Vitro. Reprod. Biol. Endocrinol. 2012, 10, 103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Wang, F.; Tian, X.; Zhang, L.; Tan, D.; Reiter, R.J.; Liu, G. Melatonin Promotes the in Vitro Development of Pronuclear Embryos and Increases the Efficiency of Blastocyst Implantation in Murine. J. Pineal Res. 2013, 55, 267–274. [Google Scholar] [CrossRef]
  105. 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]
  106. Smith, Z.D.; Meissner, A. DNA Methylation: Roles in Mammalian Development. Nat. Rev. Genet. 2013, 14, 204–220. [Google Scholar] [CrossRef] [PubMed]
  107. Vázquez, J.; González, B.; Sempere, V.; Mas, A.; Torija, M.J.; Beltran, G. Melatonin Reduces Oxidative Stress Damage Induced by Hydrogen Peroxide in Saccharomyces Cerevisiae. Front. Microbiol. 2017, 8, 1066. [Google Scholar] [CrossRef] [PubMed]
  108. Gitto, E.; Aversa, S.; Reiter, R.J.; Barberi, I.; Pellegrino, S. Update on the Use of Melatonin in Pediatrics. J. Pineal Res. 2011, 50, 21–28. [Google Scholar] [CrossRef] [PubMed]
  109. Gitto, E.; Pellegrino, S.; Gitto, P.; Barberi, I.; Reiter, R.J. Oxidative Stress of the Newborn in the Pre- and Postnatal Period and the Clinical Utility of Melatonin. J. Pineal Res. 2009, 46, 128–139. [Google Scholar] [CrossRef] [PubMed]
  110. Thomas, L.; Purvis, C.C.; Drew, J.E.; Abramovich, D.R.; Williams, L.M. Melatonin Receptors in Human Fetal Brain: 2-[125I]Iodomelatonin Binding and MT1 Gene Expression. J. Pineal Res. 2002, 33, 218–224. [Google Scholar] [CrossRef]
  111. McDonald, C.A.; Penny, T.R.; Paton, M.C.B.; Sutherland, A.E.; Nekkanti, L.; Yawno, T.; Castillo-Melendez, M.; Fahey, M.C.; Jones, N.M.; Jenkin, G.; et al. Effects of Umbilical Cord Blood Cells, and Subtypes, to Reduce Neuroinflammation Following Perinatal Hypoxic-Ischemic Brain Injury. J. Neuroinflamm. 2018, 15, 47. [Google Scholar] [CrossRef]
  112. Cao, W.; Carney, J.M.; Duchon, A.; Floyd, R.A.; Chevion, M. Oxygen Free Radical Involvement in Ischemia and Reperfusion Injury to Brain. Neurosci. Lett. 1988, 88, 233–238. [Google Scholar] [CrossRef]
  113. Jang, M.H.; Jung, S.B.; Lee, M.H.; Kim, C.J.; Oh, Y.T.; Kang, I.; Kim, J.; Kim, E.H. Melatonin Attenuates Amyloid Beta25-35-Induced Apoptosis in Mouse Microglial BV2 Cells. Neurosci. Lett. 2005, 380, 26–31. [Google Scholar] [CrossRef] [PubMed]
  114. Ebadi, M.; Sharma, S.K.; Ghafourifar, P.; Brown-Borg, H.; El ReFaey, H. Peroxynitrite in the Pathogenesis of Parkinson’s Disease and the Neuroprotective Role of Metallothioneins. Methods Enzymol. 2005, 396, 276–298. [Google Scholar] [CrossRef] [PubMed]
  115. Aly, H.; Elmahdy, H.; El-Dib, M.; Rowisha, M.; Awny, M.; El-Gohary, T.; Elbatch, M.; Hamisa, M.; El-Mashad, A.R. Melatonin Use for Neuroprotection in Perinatal Asphyxia: A Randomized Controlled Pilot Study. J. Perinatol. 2015, 35, 186–191. [Google Scholar] [CrossRef] [PubMed]
  116. Dayoub, J.C.; Ortiz, F.; Lõpez, L.C.; Venegas, C.; Del Pino-Zumaquero, A.; Roda, O.; Sánchez-Montesinos, I.; Acuña-Castroviejo, D.; Escames, G. Synergism between Melatonin and Atorvastatin against Endothelial Cell Damage Induced by Lipopolysaccharide. J. Pineal Res. 2011, 51, 324–330. [Google Scholar] [CrossRef]
  117. Hu, S.; Zhu, P.; Zhou, H.; Zhang, Y.; Chen, Y. Melatonin-Induced Protective Effects on Cardiomyocytes against Reperfusion Injury Partly through Modulation of IP3R and SERCA2a via Activation of ERK1. Arq. Bras. Cardiol. 2018, 110, 44–51. [Google Scholar] [CrossRef]
  118. Giacomo, C.G.; Antonio, M. Melatonin in Cardiac Ischemia/Reperfusion-Induced Mitochondrial Adaptive Changes. Cardiovasc. Hematol. Disord. Drug Targets 2007, 7, 163–169. [Google Scholar] [CrossRef]
  119. Qi, X.; Wang, J. Melatonin Improves Mitochondrial Biogenesis through the AMPK / PGC1α Pathway to Attenuate Ischemia/Reperfusion -Induced Myocardial Damage. Aging 2020, 12, 1–14. [Google Scholar] [CrossRef]
  120. Stroethoff, M.; Goetze, L.; Torregroza, C.; Bunte, S.; Raupach, A.; Heinen, A.; Mathes, A.; Hollmann, M.W.; Huhn, R. The Melatonin Receptor Agonist Ramelteon Induces Cardioprotection That Requires MT2 Receptor Activation and Release of Reactive Oxygen Species. Cardiovasc. Drugs Ther. 2020, 34, 303–310. [Google Scholar] [CrossRef] [Green Version]
  121. Zhou, H.; Ma, Q.; Zhu, P.; Ren, J.; Reiter, R.J.; Chen, Y. Protective Role of Melatonin in Cardiac Ischemia-Reperfusion Injury: From Pathogenesis to Targeted Therapy. J. Pineal Res. 2018, 64, 1–21. [Google Scholar] [CrossRef] [Green Version]
  122. Baker, J.; Kimpinski, K. Role of melatonin in blood pressure regulation: An adjunct anti-hypertensive agent. Clin. Exp. Pharmacol. Physiol. 2018, 45, 755–766. [Google Scholar] [CrossRef]
  123. Pechanova, O.; Paulis, L.; Simko, F. Peripheral and Central Effects of Melatonin on Blood Pressure Regulation. Int. J. Mol. Sci. 2014, 15, 17920–17937. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Cardinali, D.P.; Hardeland, R. Inflammaging, Metabolic Syndrome and Melatonin: A Call for Treatment Studies. Neuroendocrinology 2017, 104, 382–397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Dominguez-Rodriguez, A.; Abreu-Gonzalez, P.; De La Torre-Hernandez, J.M.; Gonzalez-Gonzalez, J.; Garcia-Camarero, T.; Consuegra-Sanchez, L.; Garcia-Saiz, M.d.M.; Aldea-Perona, A.; Virgos-Aller, T.; Azpeitia, A.; et al. Effect of Intravenous and Intracoronary Melatonin as an Adjunct to Primary Percutaneous Coronary Intervention for Acute ST-Elevation Myocardial Infarction: Results of the Melatonin Adjunct in the Acute MyocaRdial Infarction Treated with Angioplasty Trial. J. Pineal Res. 2017, 62, e12374. [Google Scholar] [CrossRef] [PubMed]
  126. Ekeløf, S.V.; Halladin, N.L.; Jensen, S.E.; Zaremba, T.; Aarøe, J.; Kjærgaard, B.; Simonsen, C.W.; Rosenberg, J.; Gögenur, I. Effects of Intracoronary Melatonin on Ischemia–Reperfusion Injury in ST-Elevation Myocardial Infarction. Heart Vessel. 2016, 31, 88–95. [Google Scholar] [CrossRef]
  127. Virag, J.; Lust, R. Circadian Influences on Myocardial Infarction. Front. Physiol. 2014, 5, 1–10. [Google Scholar] [CrossRef]
  128. Singhanat, K.; Apaijai, N.; Chattipakorn, S.C.; Chattipakorn, N. Roles of Melatonin and Its Receptors in Cardiac Ischemia—Reperfusion Injury. Cell. Mol. Life Sci. 2018, 75, 4125–4149. [Google Scholar] [CrossRef]
  129. Dominguez-Rodriguez, A.; Abreu-Gonzalez, P.; Reiter, R.J. Melatonin for Cardioprotection in ST Elevation Myocardial Infarction: Are We Ready for the Challenge? Heart 2016, 103, 647–648. [Google Scholar] [CrossRef]
  130. Dominguez-Rodriguez, A.; Abreu-Gonzalez, P.; Arroyo-Ucar, E.; Reiter, R.J. Decreased Level of Melatonin in Serum Predicts Left Ventricular Remodelling after Acute Myocardial Infarction. J. Pineal Res. 2012, 53, 319–323. [Google Scholar] [CrossRef]
  131. Dominguez-Rodriguez, A.; Abreu-Gonzalez, P.; De La Torre-Hernandez, J.M.; Consuegra-Sanchez, L.; Piccolo, R.; Gonzalez-Gonzalez, J.; Garcia-Camarero, T.; Del Mar Garcia-Saiz, M.; Aldea-Perona, A.; Reiter, R.J.; et al. Usefulness of Early Treatment With Melatonin to Reduce Infarct Size in Patients With ST-Segment Elevation Myocardial Infarction Receiving Percutaneous Coronary Intervention (From the Melatonin Adjunct in the Acute Myocardial Infarction Treated With Angioplasty Trial). Am. J. Cardiol. 2017, 120, 522–526. [Google Scholar] [CrossRef]
  132. Dominguez-Rodriguez, A.; Abreu-Gonzalez, P.; Piccolo, R.; Galasso, G.; Reiter, R.J. Melatonin Is Associated with Reverse Remodeling after Cardiac Resynchronization Therapy in Patients with Heart Failure and Ventricular Dyssynchrony. Int. J. Cardiol. 2016, 221, 359–363. [Google Scholar] [CrossRef]
  133. Misaka, T.; Yoshihisa, A.; Yokokawa, T.; Sato, T.; Oikawa, M.; Kobayashi, A.; Yamaki, T.; Sugimoto, K.; Kunii, H.; Nakazato, K.; et al. Plasma Levels of Melatonin in Dilated Cardiomyopathy. J. Pineal Res. 2019, 66, e12564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Che, H.; Wang, Y.; Li, H.; Li, Y.; Sahil, A.; Lv, J.; Liu, Y.; Yang, Z.; Dong, R.; Xue, H.; et al. Melatonin Alleviates Cardiac Fibrosis via Inhibiting LncRNA MALAT1/MiR-141-Mediated NLRP3 Inflammasome and TGF-Β1/Smads Signaling in Diabetic Cardiomyopathy. FASEB J. 2020, 34, 5282–5298. [Google Scholar] [CrossRef] [PubMed]
  135. Crnko, S.; Printezi, M.I.; Jansen, T.P.J.; Leiteris, L.; Van Der Meer, M.G.; Schutte, H.; Van Faassen, M.; Du Pré, B.C.; De Jonge, N.; Asselbergs, F.W.; et al. Prognostic Biomarker Soluble ST2 Exhibits Diurnal Variation in Chronic Heart Failure Patients. ESC Heart Fail. 2020, 7, 1224–1233. [Google Scholar] [CrossRef] [PubMed]
  136. Dwaich, K.H.; Al-Amran, F.G.Y.; AL-Sheibani, B.I.M.; Al-Aubaidy, H.A. Melatonin Effects on Myocardial Ischemia–Reperfusion Injury: Impact on the Outcome in Patients Undergoing Coronary Artery Bypass Grafting Surgery. Int. J. Cardiol. 2016, 221, 977–986. [Google Scholar] [CrossRef]
  137. Dominguez-Rodriguez, A.; Abreu-Gonzalez, P. Future Strategies for Acute Cardioprotection: “Melatonin as Promising Therapy.”. Cardiovasc. Res. 2017, 113, 1418. [Google Scholar] [CrossRef]
  138. Popescu, M.R.; Panaitescu, A.M.; Pavel, B.; Zagrean, L.; Peltecu, G.; Zagrean, A.M. Getting an Early Start in Understanding Perinatal Asphyxia Impact on the Cardiovascular System. Front. Pediatr. 2020, 8, 68. [Google Scholar] [CrossRef]
  139. Tare, M.; Parkington, H.C.; Wallace, E.M.; Sutherland, A.E.; Lim, R.; Yawno, T.; Coleman, H.A.; Jenkin, G.; Miller, S.L. Maternal Melatonin Administration Mitigates Coronary Stiffness and Endothelial Dysfunction, and Improves Heart Resilience to Insult in Growth Restricted Lambs. J. Physiol. 2014, 592, 2695–2709. [Google Scholar] [CrossRef] [Green Version]
  140. Tain, Y.L.; Huang, L.T.; Hsu, C.N. Developmental Programming of Adult Disease: Reprogramming by Melatonin? Int. J. Mol. Sci. 2017, 18, 426. [Google Scholar] [CrossRef] [Green Version]
  141. Tain, Y.L.; Huang, L.T.; Hsu, C.N.; Lee, C.-T. Melatonin Therapy Prevents Programmed Hypertension and Nitric Oxide Deficiency in Offspring Exposed to Maternal Caloric Restriction. Oxidative Med. Cell. Longev. 2014, 2014, 1–21. [Google Scholar] [CrossRef]
  142. Tain, Y.L.; Chan, J.Y.H.; Lee, C.-T.; Hsu, C.N. Maternal Melatonin Therapy Attenuates Methyl-Donor Diet-Induced Programmed Hypertension in Male Adult Rat Offspring. Nutrients 2018, 10, 1407. [Google Scholar] [CrossRef] [Green Version]
  143. Mendez, N.; Halabi, D.; Spichiger, C.; Salazar, E.R.; Vergara, K.; Alonso-Vasquez, P.; Carmona, P.; Sarmiento, J.M.; Richter, H.G.; Seron-Ferre, M.; et al. Gestational Chronodisruption Impairs Circadian Physiology in Rat Male Offspring, Increasing the Risk of Chronic Disease. Endocrinology 2016, 157, 4654–4668. [Google Scholar] [CrossRef] [PubMed]
  144. Alamili, M.; Bendtzen, K.; Lykkesfeldt, J.; Rosenberg, J.; Gögenur, I. Melatonin Suppresses Markers of Inflammation and Oxidative Damage in a Human Daytime Endotoxemia Model. J. Crit. Care 2014, 29, 184.e9–184.e13. [Google Scholar] [CrossRef] [PubMed]
  145. Dominguez-Rodriguez, A.; Abreu-Gonzalez, P.; Garcia, M.; Ferrer, J.; De La Rosa, A.; Vargas, M.; Reiter, R.J. Light/Dark Patterns of Interleukin-6 in Relation to the Pineal Hormone Melatonin in Patients with Acute Myocardial Infarction. Cytokine 2004, 26, 89–93. [Google Scholar] [CrossRef] [PubMed]
  146. Dominguez-Rodriguez, A.; Garcia-Gonzalez, M.; Abreu-Gonzalez, P.; Ferrer, J.; Kaski, J.C. Relation of Nocturnal Melatonin Levels to C-Reactive Protein Concentration in Patients with ST-Segment Elevation Myocardial Infarction. Am. J. Cardiol. 2006, 97, 10–12. [Google Scholar] [CrossRef]
  147. Libby, P. Inflammation in Atherosclerosis. Arter. Thromb. Vasc. Biol. 2012, 32, 2045–2051. [Google Scholar] [CrossRef] [Green Version]
  148. Bäck, M.; Yurdagul, A.; Tabas, I.; Öörni, K.; Kovanen, P.T. Inflammation and Its Resolution in Atherosclerosis: Mediators and Therapeutic Opportunities. Nat. Rev. Cardiol. 2019, 1, 389–406. [Google Scholar] [CrossRef]
  149. Oxenkrug, G.F.; Summergrad, P. Ramelteon Attenuates Age-Associated Hypertension and Weight Gain in Spontaneously Hypertensive Rats. In Annals of the New York Academy of Sciences; Blackwell Publishing Inc.: New York, NY, USA, 2010; Volume 1199, pp. 114–120. [Google Scholar] [CrossRef]
  150. She, M.; Hu, X.; Su, Z.; Zhang, C.; Yang, S.; Ding, L.; Laudon, M.; Yin, W. Piromelatine, a Novel Melatonin Receptor Agonist, Stabilizes Metabolic Profiles and Ameliorates Insulin Resistance in Chronic Sleep Restricted Rats. Eur. J. Pharmacol. 2014, 727, 60–65. [Google Scholar] [CrossRef]
  151. Obayashi, K.; Saeki, K.; Kurumatani, N. Higher Melatonin Secretion Is Associated with Lower Leukocyte and Platelet Counts in the General Elderly Population: The Kyo-Heijo Cohort. J. Pineal Res. 2015, 58, 227–233. [Google Scholar] [CrossRef]
  152. Paredes, S.D.; Forman, K.A.; García, C.; Vara, E.; Escames, G.; Tresguerres, J.A.F. Protective Actions of Melatonin and Growth Hormone on the Aged Cardiovascular System. Horm. Mol. Biol. Clin. Investig. 2014, 18, 79–88. [Google Scholar] [CrossRef]
  153. Favero, G.; Franceschetti, L.; Buffoli, B.; Moghadasian, M.H.; Reiter, R.J.; Rodella, L.F.; Rezzani, R. Melatonin: Protection against Age-Related Cardiac Pathology. Ageing Res. Rev. 2017, 35, 336–349. [Google Scholar] [CrossRef]
  154. Obayashi, K.; Saeki, K.; Tone, N.; Kurumatani, N. Relationship between Melatonin Secretion and Nighttime Blood Pressure in Elderly Individuals with and without Antihypertensive Treatment: A Cross-Sectional Study of the HEIJO-KYO Cohort. Hypertens. Res. 2014, 37, 908–913. [Google Scholar] [CrossRef] [PubMed]
  155. Reiter, R.J.; Tan, D.X.; Korkmaz, A. The Circadian Melatonin Rhythm and Its Modulation: Possible Impact on Hypertension. J. Hypertens. 2009, 27, S17–S20. [Google Scholar] [CrossRef] [PubMed]
  156. Li, Y.; Vgontzas, A.N.; Fernandez-Mendoza, J.; Bixler, E.O.; Sun, Y.; Zhou, J.; Ren, R.; Li, T.; Tang, X. Insomnia with Physiological Hyperarousal Is Associated with Hypertension. Hypertension 2015, 65, 644–650. [Google Scholar] [CrossRef] [PubMed]
  157. Akbari, M.; Ostadmohammadi, V.; Tabrizi, R.; Lankarani, K.B.; Heydari, S.T.; Amirani, E.; Reiter, R.J.; Asemi, Z. The Effects of Melatonin Supplementation on Inflammatory Markers among Patients with Metabolic Syndrome or Related Disorders: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Inflammopharmacology 2018, 26, 899–907. [Google Scholar] [CrossRef] [PubMed]
  158. Zephy, D.; Ahmad, J. Type 2 Diabetes Mellitus: Role of Melatonin and Oxidative Stress. Diabetes Metab. Syndr. Clin. Res. Rev. 2015, 9, 127–131. [Google Scholar] [CrossRef] [PubMed]
  159. Korkmaz, A.; Ma, S.; Topal, T.; Rosales-Corral, S.; Tan, D.X.; Reiter, R.J. Glucose: A Vital Toxin and Potential Utility of Melatonin in Protecting against the Diabetic State. Mol. Cell. Endocrinol. 2012, 349, 128–137. [Google Scholar] [CrossRef]
  160. Rezzani, R.; Favero, G.; Stacchiotti, A.; Rodella, L.F. Endothelial and Vascular Smooth Muscle Cell Dysfunction Mediated by Cyclophylin A and the Atheroprotective Effects of Melatonin. Life Sci. 2013, 92, 875–882. [Google Scholar] [CrossRef]
  161. Mesri Alamdari, N.; Mahdavi, R.; Roshanravan, N.; Lotfi Yaghin, N.; Ostadrahimi, A.R.; Faramarzi, E. A Double-Blind, Placebo-Controlled Trial Related to the Effects of Melatonin on Oxidative Stress and Inflammatory Parameters of Obese Women. Horm. Metab. Res. 2014, 47, 504–508. [Google Scholar] [CrossRef] [Green Version]
  162. Favero, G.; Franco, C.; Stacchiotti, A.; Rodella, L.F.; Rezzani, R. Sirtuin1 Role in the Melatonin Protective Effects Against Obesity-Related Heart Injury. Front. Physiol. 2020, 11, 11. [Google Scholar] [CrossRef] [Green Version]
  163. McMullan, C.J.; Rimm, E.B.; Schernhammer, E.S.; Forman, J.P. A Nested Case-Control Study of the Association between Melatonin Secretion and Incident Myocardial Infarction. Heart 2016, 103, 694–701. [Google Scholar] [CrossRef]
  164. Talafi Noghani, M.; Namdar, H. Migraine Associated with Gastrointestinal Disorders: A Pathophysiological Explanation. Med. Hypotheses 2019, 125, 90–93. [Google Scholar] [CrossRef] [PubMed]
  165. Yin, J.; Li, Y.; Han, H.; Chen, S.; Gao, J.; Liu, G.; Wu, X.; Deng, J.; Yu, Q.; Huang, X.; et al. Melatonin Reprogramming of Gut Microbiota Improves Lipid Dysmetabolism in High-Fat Diet-Fed Mice. J. Pineal Res. 2018, 65, e12524. [Google Scholar] [CrossRef] [PubMed]
  166. Hu, Z.P.; Fang, X.L.; Fang, N.; Wang, X.B.; Qian, H.Y.; Cao, Z.; Cheng, Y.; Wang, B.N.; Wang, Y. Melatonin Ameliorates Vascular Endothelial Dysfunction, Inflammation, and Atherosclerosis by Suppressing the TLR4/NF-ΚB System in High-Fat-Fed Rabbits. J. Pineal Res. 2013, 55, 388–398. [Google Scholar] [CrossRef] [PubMed]
  167. Chen, W.R.; Zhou, Y.J.; Yang, J.Q.; Liu, F.; Wu, X.P.; Sha, Y. Melatonin Attenuates Calcium Deposition from Vascular Smooth Muscle Cells by Activating Mitochondrial Fusion and Mitophagy via an AMPK/OPA1 Signaling Pathway. Oxidative Med. Cell. Longev. 2020, 2020, 1–23. [Google Scholar] [CrossRef]
  168. Xu, F.; Zhong, J.Y.; Lin, X.; Shan, S.K.; Guo, B.; Zheng, M.H.; Wang, Y.; Li, F.; Cui, R.R.; Wu, F.; et al. Melatonin Alleviates Vascular Calcification and Ageing through Exosomal MiR-204/MiR-211 Cluster in a Paracrine Manner. J. Pineal Res. 2020, 68, 1–20. [Google Scholar] [CrossRef] [Green Version]
  169. Sallinen, P.; Mänttäri, S.; Leskinen, H.; Ilves, M.; Vakkuri, O.; Ruskoaho, H.; Saarela, S. The Effect of Myocardial Infarction on the Synthesis, Concentration and Receptor Expression of Endogenous Melatonin. J. Pineal Res. 2007, 42, 254–260. [Google Scholar] [CrossRef]
  170. Altun, A.; Yaprak, M.; Aktoz, M.; Vardar, A.; Betul, U.A.; Ozbay, G. Impaired Nocturnal Synthesis of Melatonin in Patients with Cardiac Syndrome X. Neurosci. Lett. 2002, 327, 143–145. [Google Scholar] [CrossRef]
  171. Domínguez-Rodríguez, A.; Abreu-González, P.; García, M.J.; Sanchez, J.; Marrero, F.; De Armas-Trujillo, D. Decreased Nocturnal Melatonin Levels during Acute Myocardial Infarction. J. Pineal Res. 2002, 33, 248–252. [Google Scholar] [CrossRef]
  172. Fernandes, P.A.; Tamura, E.K.; D’Argenio-Garcia, L.; Muxel, S.M.; Da Silveira Cruz-Machado, S.; Marçola, M.; Carvalho-Sousa, C.E.; Cecon, E.; Ferreira, Z.S.; Markus, R.P. Dual Effect of Catecholamines and Corticosterone Crosstalk on Pineal Gland Melatonin Synthesis. Neuroendocrinology 2016, 104, 126–134. [Google Scholar] [CrossRef]
  173. Yang, J.B.; Kang, Y.M.; Zhang, C.; Yu, X.J.; Chen, W.S. Infusion of Melatonin into the Paraventricular Nucleus Ameliorates Myocardial Ischemia-Reperfusion Injury by Regulating Oxidative Stress and Inflammatory Cytokines. J. Cardiovasc. Pharmacol. 2019, 74, 336–347. [Google Scholar] [CrossRef]
  174. Nduhirabandi, F.; Du Toit, E.F.; Blackhurst, D.; Marais, D.; Lochner, A. Chronic Melatonin Consumption Prevents Obesity-Related Metabolic Abnormalities and Protects the Heart against Myocardial Ischemia and Reperfusion Injury in a Prediabetic Model of Diet-Induced Obesity. J. Pineal Res. 2011, 50, 171–182. [Google Scholar] [CrossRef] [PubMed]
  175. Li, H.R.; Wang, C.; Sun, P.; Liu, D.; Du, G.; Tian, J.W. Melatonin Attenuates Doxorubicin-Induced Cardiotoxicity through Preservation of YAP Expression. J. Cell. Mol. Med. 2020, 24, 3634–3646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Stoschitzky, K.; Stoschitzky, G.; Brussee, H.; Bonell, C.; Dobnig, H. Comparing Beta-Blocking Effects of Bisoprolol, Carvedilol and Nebivolol. Cardiology 2006, 106, 199–206. [Google Scholar] [CrossRef] [PubMed]
  177. Stoschitzky, K.; Sakotnik, A.; Lercher, P.; Zweiker, R.; Maier, R.; Liebmann, P.; Lindner, W. Influence of Beta-Blockers on Melatonin Release. Eur. J. Clin. Pharmacol. 1999, 55, 111–115. [Google Scholar] [CrossRef] [PubMed]
  178. Drobnik, J.; Owczarek, K.; Piera, L.; Tosik, D.; Olczak, S.; Ciosek, J.; Hrabec, E. Melatonin-Induced Augmentation of Collagen Deposition in Cultures of Fibroblasts and Myofibroblasts Is Blocked by Luzindole—A Melatonin Membrane Receptors Inhibitor. Pharmacol. Rep. 2013, 65, 642–649. [Google Scholar] [CrossRef]
  179. Drobnik, J.; Karbownik-Lewińska, M.; Szczepanowska, A.; Słotwińska, D.; Olczak, S.; Jakubowski, L.; Da̧browski, R. Regulatory Influence of Melatonin on Collagen Accumulation in the Infarcted Heart Scar. J. Pineal Res. 2008, 45, 285–290. [Google Scholar] [CrossRef]
  180. Drobnik, J.; Tosik, D.; Piera, L.; Szczepanowska, A.; Olczak, S.; Zielinska, A.; Liberski, P.P.; Ciosek, J. Melatonin-Induced Glycosaminoglycans Augmentation in Myocardium Remote to Infarction. J. Physiol. Pharm. 2013, 64, 737–744. [Google Scholar]
  181. Feigin, V.L.; Nichols, E.; Alam, T.; Bannick, M.S.; Beghi, E.; Blake, N.; Culpepper, W.J.; Dorsey, E.R.; Elbaz, A.; Ellenbogen, R.G.; et al. Global, Regional, and National Burden of Neurological Disorders, 1990–2016: A Systematic Analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019, 18, 459–480. [Google Scholar] [CrossRef] [Green Version]
  182. Hurtado-Alvarado, G.; Domínguez-Salazar, E.; Pavon, L.; Velázquez-Moctezuma, J.; Gómez-González, B. Blood-Brain Barrier Disruption Induced by Chronic Sleep Loss: Low-Grade Inflammation May Be the Link. J. Immunol. Res. 2016, 2016, 1–15. [Google Scholar] [CrossRef] [Green Version]
  183. Ali, T.; Rehman, S.U.; Shah, F.A.; Kim, M.O. Acute Dose of Melatonin via Nrf2 Dependently Prevents Acute Ethanol-Induced Neurotoxicity in the Developing Rodent Brain. J. Neuroinflamm. 2018, 15, 119. [Google Scholar] [CrossRef]
  184. Tyagi, E.; Agrawal, R.; Nath, C.; Shukla, R. Effect of Melatonin on Neuroinflammation and Acetylcholinesterase Activity Induced by LPS in Rat Brain. Eur. J. Pharmacol. 2010, 640, 206–210. [Google Scholar] [CrossRef] [PubMed]
  185. Guzman-Martinez, L.; Maccioni, R.B.; Andrade, V.; Navarrete, L.P.; Pastor, M.G.; Ramos-Escobar, N. Neuroinflammation as a Common Feature of Neurodegenerative Disorders. Front. Pharmacol. 2019, 10, 1008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Tan, S.H.; Karri, V.; Tay, N.W.R.; Chang, K.H.; Ah, H.Y.; Ng, P.Q.; Ho, H.S.; Keh, H.W.; Candasamy, M. Emerging Pathways to Neurodegeneration: Dissecting the Critical Molecular Mechanisms in Alzheimer’s Disease, Parkinson’s Disease. Biomed. Pharmacother. 2019, 111, 765–777. [Google Scholar] [CrossRef] [PubMed]
  187. Rudnitskaya, E.A.; Maksimova, K.Y.; Muraleva, N.A.; Logvinov, S.V.; Yanshole, L.V.; Kolosova, N.G.; Stefanova, N.A. Beneficial Effects of Melatonin in a Rat Model of Sporadic Alzheimer’s Disease. Biogerontology 2015, 16, 303–316. [Google Scholar] [CrossRef]
  188. Cardinali, D.P. Melatonin: Clinical Perspectives in Neurodegeneration. Front. Endocrinol. 2019, 10, 480. [Google Scholar] [CrossRef]
  189. Alghamdi, B.S. The Neuroprotective Role of Melatonin in Neurological Disorders. J. Neurosci. Res. 2018, 96, 1136–1149. [Google Scholar] [CrossRef]
  190. Sayre, L.; Smith, M.; Perry, G. Chemistry and Biochemistry of Oxidative Stress in Neurodegenerative Disease. Curr. Med. Chem. 2012, 8, 721–738. [Google Scholar] [CrossRef]
  191. Maurizi, C.P. Alzheimer’s Disease: Roles for Mitochondrial Damage, the Hydroxyl Radical, and Cerebrospinal Fluid Deficiency of Melatonin. Med. Hypotheses 2001, 57, 156–160. [Google Scholar] [CrossRef]
  192. Wongprayoon, P.; Govitrapong, P. Melatonin Attenuates Methamphetamine-Induced Neurotoxicity. Curr. Pharm. Des. 2016, 22, 1022–1032. [Google Scholar] [CrossRef]
  193. Şirin, F.B.; Kumbul Doğuc, D.; Vural, H.; Eren, İ.; İnanli, İ.; Sütçü, R.; Delibaş, N. Plasma 8-IsoPGF2α and Serum Melatonin Levels in Patients with Minimal Cognitive Impairment and Alzheimer Disease. Turk. J. Med. Sci. 2015, 45, 1073–1077. [Google Scholar] [CrossRef]
  194. Menegardo, C.S.; Friggi, F.A.; Scardini, J.B.; Rossi, T.S.; Vieira, T.D.S.; Tieppo, A.; Morelato, R.L. Sundown Syndrome in Patients with Alzheimer’s Disease Dementia. Dement. Neuropsychol. 2019, 13, 469–474. [Google Scholar] [CrossRef] [PubMed]
  195. Paul, R.; Phukan, B.C.; Justin Thenmozhi, A.; Manivasagam, T.; Bhattacharya, P.; Borah, A. Melatonin Protects against Behavioral Deficits, Dopamine Loss and Oxidative Stress in Homocysteine Model of Parkinson’s Disease. Life Sci. 2018, 192, 238–245. [Google Scholar] [CrossRef] [PubMed]
  196. Dowling, G.A.; Mastick, J.; Colling, E.; Carter, J.H.; Singer, C.M.; Aminoff, M.J. Melatonin for Sleep Disturbances in Parkinson’s Disease. Sleep Med. 2005, 6, 459–466. [Google Scholar] [CrossRef] [PubMed]
  197. Chang, C.F.; Huang, H.J.; Lee, H.C.; Hung, K.C.; Wu, R.T.; Lin, A.M.Y. Melatonin Attenuates Kainic Acid-Induced Neurotoxicity in Mouse Hippocampus via Inhibition of Autophagy and α-Synuclein Aggregation. J. Pineal Res. 2012, 52, 312–321. [Google Scholar] [CrossRef] [PubMed]
  198. Lin, A.M.Y.; Fang, S.F.; Chao, P.L.; Yang, C.H. Melatonin Attenuates Arsenite-Induced Apoptosis in Rat Brain: Involvement of Mitochondrial and Endoplasmic Reticulum Pathways and Aggregation of α-Synuclein. J. Pineal Res. 2007, 43, 163–171. [Google Scholar] [CrossRef]
  199. Brito-Armas, J.M.; Baekelandt, V.; Castro-Hernández, J.R.; González-Hernández, T.; Rodríguez, M.; Fuentes, R.C. Melatonin Prevents Dopaminergic Cell Loss Induced by Lentiviral Vectors Expressing A30P Mutant Alpha-Synuclein. Histol. Histopathol. 2013, 28, 999–1006. [Google Scholar] [CrossRef]
  200. Hu, W.; Ma, Z.; Jiang, S.; Fan, C.; Deng, C.; Yan, X.; Di, S.; Lv, J.; Reiter, R.J.; Yang, Y. Melatonin: The Dawning of a Treatment for Fibrosis? J. Pineal Res. 2016, 60, 121–131. [Google Scholar] [CrossRef]
  201. Zhao, X.; Sun, J.; Su, W.; Shan, H.; Zhang, B.; Wang, Y.; Shabanova, A.; Shan, H.; Liang, H. Melatonin Protects against Lung Fibrosis by Regulating the Hippo/YAP Pathway. Int. J. Mol. Sci. 2018, 19, 1118. [Google Scholar] [CrossRef] [Green Version]
  202. Yildirim, Z.; Kotuk, M.; Erdogan, H.; Iraz, M.; Yagmurca, M.; Kuku, I.; Fadillioglu, E. Preventive Effect of Melatonin on Bleomycin-Induced Lung Fibrosis in Rats. J. Pineal Res. 2006, 40, 27–33. [Google Scholar] [CrossRef]
  203. Wang, R.; Zhou, S.; Wu, P.; Li, M.; Ding, X.; Sun, L.; Xu, X.; Zhou, X.; Zhou, L.; Cao, C.; et al. Identifying Involvement of H19-MiR-675-3p-IGF1R and H19-MiR-200a-PDCD4 in Treating Pulmonary Hypertension with Melatonin. Mol. Ther. Nucleic Acids 2018, 13, 44–54. [Google Scholar] [CrossRef] [Green Version]
  204. Gonzalez-Candia, A.; Veliz, M.; Carrasco-Pozo, C.; Castillo, R.L.; Cárdenas, J.C.; Ebensperger, G.; Reyes, R.V.; Llanos, A.J.; Herrera, E.A. Antenatal Melatonin Modulates an Enhanced Antioxidant/pro-Oxidant Ratio in Pulmonary Hypertensive Newborn Sheep. Redox Biol. 2019, 22, 101128. [Google Scholar] [CrossRef] [PubMed]
  205. Hung, M.W.; Yeung, H.M.; Lau, C.F.; Poon, A.M.S.; Tipoe, G.L.; Fung, M.L. Melatonin Attenuates Pulmonary Hypertension in Chronically Hypoxic Rats. Int. J. Mol. Sci. 2017, 18, 1125. [Google Scholar] [CrossRef] [PubMed]
  206. Maarman, G.; Blackhurst, D.; Thienemann, F.; Blauwet, L.; Butrous, G.; Davies, N.; Sliwa, K.; Lecour, S. Melatonin as a Preventive and Curative Therapy against Pulmonary Hypertension. J. Pineal Res. 2015, 59, 343–353. [Google Scholar] [CrossRef] [PubMed]
  207. He, B.; Zhang, W.; Qiao, J.; Peng, Z.; Chai, X. Melatonin Protects against COPD by Attenuating Apoptosis and Endoplasmic Reticulum Stress via Upregulating SIRT1 Expression in Rats. Can. J. Physiol. Pharmacol. 2019, 97, 386–392. [Google Scholar] [CrossRef]
  208. De Matos Cavalcante, A.G.; De Bruin, P.F.C.; De Bruin, V.M.S.; Nunes, D.M.; Pereira, E.D.B.; Cavalcante, M.M.; Andrade, G.M. Melatonin Reduces Lung Oxidative Stress in Patients with Chronic Obstructive Pulmonary Disease: A Randomized, Double-Blind, Placebo-Controlled Study. J. Pineal Res. 2012, 53, 238–244. [Google Scholar] [CrossRef]
  209. Gattinoni, L.; Chiumello, D.; Caironi, P.; Busana, M.; Romitti, F.; Brazzi, L.; Camporota, L. COVID-19 Pneumonia: Different Respiratory Treatments for Different Phenotypes? Intensive Care Med. 2020, 46, 1099–1102. [Google Scholar] [CrossRef]
  210. Wichmann, D.; Sperhake, J.-P.; Lütgehetmann, M.; Steurer, S.; Edler, C.; Heinemann, A.; Heinrich, F.; Mushumba, H.; Kniep, I.; Schröder, A.S.; et al. Autopsy Findings and Venous Thromboembolism in Patients With COVID-19. Ann. Intern. Med. 2020, 173, 268–277. [Google Scholar] [CrossRef]
  211. Ye, Q.; Wang, B.; Mao, J. The Pathogenesis and Treatment of the ‘Cytokine Storm’ in COVID-19. J. Infect. 2020, 80, 607–613. [Google Scholar] [CrossRef]
  212. Shneider, A.; Kudriavtsev, A.; Vakhrusheva, A. Can Melatonin Reduce the Severity of COVID-19 Pandemic? Int. Rev. Immunol. 2020, 39, 153–162. [Google Scholar] [CrossRef]
  213. Zhang, R.; Wang, X.; Ni, L.; Di, X.; Ma, B.; Niu, S.; Liu, C.; Reiter, R.J. COVID-19: Melatonin as a Potential Adjuvant Treatment. Life Sci. 2020, 250, 117583. [Google Scholar] [CrossRef]
  214. Zhang, Y.; Li, X.; Grailer, J.J.; Wang, N.; Wang, M.; Yao, J.; Zhong, R.; Gao, G.F.; Ward, P.A.; Tan, D.-X.; et al. Melatonin Alleviates Acute Lung Injury through Inhibiting the NLRP3 Inflammasome. J. Pineal Res. 2016, 60, 405–414. [Google Scholar] [CrossRef] [PubMed]
  215. Wirtz, P.H.; Bärtschi, C.; Spillmann, M.; Ehlert, U.; Von Känel, R. Effect of Oral Melatonin on the Procoagulant Response to Acute Psychosocial Stress in Healthy Men: A Randomized Placebo-Controlled Study. J. Pineal Res. 2008, 44, 358–365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Bekyarova, G.; Tancheva, S.; Hristova, M. The Effects of Melatonin on Burn-Induced Inflammatory Responses and Coagulation Disorders in Rats. Methods Find. Exp. Clin. Pharmacol. 2010, 32, 299–303. [Google Scholar] [CrossRef]
  217. Baumgartner, L.; Lam, K.; Lai, J.; Barnett, M.; Thompson, A.; Gross, K.; Morris, A. Effectiveness of Melatonin for the Prevention of Intensive Care Unit Delirium. Pharmacotherapy 2019, 39, 280–287. [Google Scholar] [CrossRef]
  218. Martínez-González, M.A.; Gea, A.; Ruiz-Canela, M. The Mediterranean Diet and Cardiovascular Health: A Critical Review. Circ. Res. 2019, 124, 779–798. [Google Scholar] [CrossRef]
  219. Guerrini, S.; Mangani, S.; Romboli, Y.; Luti, S.; Pazzagli, L.; Granchi, L. Impact of Saccharomyces Cerevisiae Strains on Health-Promoting Compounds in Wine. Fermentation 2018, 4, 26. [Google Scholar] [CrossRef] [Green Version]
  220. Papada, E.; Forbes, A.; Amerikanou, C.; Torović, L.; Kalogeropoulos, N.; Tzavara, C.; Triantafillidis, J.; Kaliora, A. Antioxidative Efficacy of a Pistacia Lentiscus Supplement and Its Effect on the Plasma Amino Acid Profile in Inflammatory Bowel Disease: A Randomised, Double-Blind, Placebo-Controlled Trial. Nutrients 2018, 10, 1779. [Google Scholar] [CrossRef] [Green Version]
  221. Treatment Protocol—Frontline COVID-19 Critical Care Alliance. Available online: https://covid19criticalcare.com/treatment-protocol/ (accessed on 24 July 2020).
Figure 1. The roles of melatonin within the mitochondria. Melatonin is transported into mitochondria through PEPT1/2 oligopeptide and Glut/SLC2A transporters, but it is also synthesized within mitochondria [12,13]. Melatonin lowers the formation of free radicals and protects ATP synthesis at the mitochondrial level. It scavenges free oxygen (ROS) and nitrogen (RNS) reactive species, by preventing mitochondrial apoptosis and disruption of the electron transport chain. Melatonin interacts with MT1 and MT2 melatonin receptors, inhibits pro-apoptosis protein synthesis, and the subsequently cytochrome C leakage at the level of the membrane. It also protects mitochondrial DNA and prevents the opening of the mitochondrial permeability transition pore (mPTP) [13].
Figure 1. The roles of melatonin within the mitochondria. Melatonin is transported into mitochondria through PEPT1/2 oligopeptide and Glut/SLC2A transporters, but it is also synthesized within mitochondria [12,13]. Melatonin lowers the formation of free radicals and protects ATP synthesis at the mitochondrial level. It scavenges free oxygen (ROS) and nitrogen (RNS) reactive species, by preventing mitochondrial apoptosis and disruption of the electron transport chain. Melatonin interacts with MT1 and MT2 melatonin receptors, inhibits pro-apoptosis protein synthesis, and the subsequently cytochrome C leakage at the level of the membrane. It also protects mitochondrial DNA and prevents the opening of the mitochondrial permeability transition pore (mPTP) [13].
Biomolecules 10 01211 g001
Figure 2. The widespread effects of melatonin at organ and system level.
Figure 2. The widespread effects of melatonin at organ and system level.
Biomolecules 10 01211 g002
Figure 3. Cardiovascular effects of melatonin. ATS: atherosclerotic disease, BP: blood pressure, CAD: coronary artery disease, IRI: ischemia/reperfusion injury, HTN: hypertension, MI: myocardial infarction, MS: metabolic syndrome, NO: nitric oxide.
Figure 3. Cardiovascular effects of melatonin. ATS: atherosclerotic disease, BP: blood pressure, CAD: coronary artery disease, IRI: ischemia/reperfusion injury, HTN: hypertension, MI: myocardial infarction, MS: metabolic syndrome, NO: nitric oxide.
Biomolecules 10 01211 g003
Table 1. Overview of the mechanisms of action and the protective effects of melatonin.
Table 1. Overview of the mechanisms of action and the protective effects of melatonin.
MechanismEffectStudy Type
Maternal & Fetal HealthImproves mitochondrial function, reduces mitochondrial oxidative stress [48]Promotes embryo developmentIn vivo
TET genes function [36]Regulation of genomic DNA methylation levelsIn vitro
Counteracts the effects of Aanat knockdown [33]Protects fertilization and tissue differentiationIn vitro
Direct scavenger of hydroxyl groups and nitrogen reactive species [49]Detoxification of superoxide anionsIn vitro
Mitochondrial complexes I and IV [50]Protects ATP synthesisIn vitro
Prevents pathological opening of mPTP [51]Protects mitochondrial DNAIn vitro
Inhibits anti-inflammatory molecules, like 8-isoprostanes [52]Reduces hypoxic-ischemic inflammationIn vivo
Inhibits Cytochrome C release [53]Anti-apoptoticIn vitro
Increases Bax and Bad pro-apoptosis molecules production [54]Anti-apoptotic
Prevention of DNA disruption
In vitro
Enhances glutathione peroxidase levels [55]AntioxidantIn vitro
Not known [56]Increases offspring survival in a model of lipopolysaccharide-induced inflammationIn vivo
Not known [57]Prevents intrauterine growth retardation associated with continuous light exposure during pregnancyIn vivo
Increases plasma NO and SOD levels [58]Beneficial in hypoxic-ischemic encephalopathy of the newbornIn vivo
Cardiovascular ImmediateIntermediateFinal
Inhibits MPTP opening [59,60]Decrease apoptosisDecrease cell deathPrevent/reduce myocardial IRIIn vitro
Inhibit Cyt C release [61]Decrease apoptosisDecrease cell deathIn vitro
Activates JAK/STAT3 [62]Decrease Bax, Increase BclDecrease apoptosisIn vitro
Improves TAC [60,61]Scavenger activity Increase endogenous antioxidant capacity Stop cardiolipin peroxidationDecrease oxidative damageIn vitro
Improves calcium handling [63]Ca2+-calmodulin modulationDecrease cell death, reduce apoptosisIn vivo
Nrf 2 [64,65]Transactivate HO-1Decrease inflammation and oxidationIn vivo
iNOS inhibition [66,67]Lower NO levelsDecrease oxidative stressIn vivo
In vitro
Inhibits inflammatory cytokine release [67]Decrease of TNF-α, IL-1β, IL-6Decrease inflammation In vitro
Stimulates anti-inflammatory cytokines [68]Increase of IL-10Decrease inflammation In vivo
NeuroinflammationMaintains the levels of parvalbumin and hippocalcin [37]Prevents neuronal death in cerebral ischemiaIn vivo
In vitro
Decreases NO, peroxynitrite formation [38]Reduces hyperactivity linked to neurodegeneration induced by cerebral ischemia and reperfusion;
Reduces PARS and brain edema
In vivo
In vitro
Activation of SIRT1 signaling [44]Reduced infarct volume, lowered brain edema, increased neurological scores in IRIIn vivo
In vitro
Prevents accumulation of free radicals [47]Counteracts the destructive effects of NMDA or hypoxia/reperfusionIn vitro
Decreases pro-inflammatory
cytokines IL-1β, IL-6, and TNF-α in PFC [69]
Attenuates neuroinflammation in the aged mouse brainIn vivo
In vitro
RespiratorySuppresses the NLRP3 inflammasome [70]Protectects against radiation-induced lung injuryIn vivo
In vitro
Decreasies lung edema and reduces NF-κB activation, enhances the secretion of IL-10 [71]Averts volume induced lung injuryIn vivo
In vitro
Sirtuin-1 upregulation [72]Diminishes TGF-β1 activity, apoptosis, and endoplasmic reticulum stressIn vivo
In vitro
AANAT: aralkylamine N-acetyltransferase; Cyt C: cytochrome C; DNA: deoxyribonucleic acid; IRI: ischemia reperfusion injury; JAK/STAT3: Janus kinase 2/signal transducers and activators of transcription 3; iNOS: inducible nitric oxide synthase; HO-1: heme oxygenase-1; IRI: ischemia-reperfusion injury; mPTP: mitochondrial permeability transition pore; NO: nitric oxide; NF-κB: nuclear factor kappa light chain enhancer of activated B cells NLRP3: NOD-like receptor protein 3 Nrf 2: nuclear factor erythroid 2-related factor 2; PARS: poly (ADP-Ribose) synthetase (PARS); SIRT1: Silent information regulator 1; SOD: superoxide dismutase; TAC: total antioxidant capacity; TET = ten-eleven-translocation; PFC: pre-frontal cortex.

Share and Cite

MDPI and ACS Style

Chitimus, D.M.; Popescu, M.R.; Voiculescu, S.E.; Panaitescu, A.M.; Pavel, B.; Zagrean, L.; Zagrean, A.-M. Melatonin’s Impact on Antioxidative and Anti-Inflammatory Reprogramming in Homeostasis and Disease. Biomolecules 2020, 10, 1211. https://doi.org/10.3390/biom10091211

AMA Style

Chitimus DM, Popescu MR, Voiculescu SE, Panaitescu AM, Pavel B, Zagrean L, Zagrean A-M. Melatonin’s Impact on Antioxidative and Anti-Inflammatory Reprogramming in Homeostasis and Disease. Biomolecules. 2020; 10(9):1211. https://doi.org/10.3390/biom10091211

Chicago/Turabian Style

Chitimus, Diana Maria, Mihaela Roxana Popescu, Suzana Elena Voiculescu, Anca Maria Panaitescu, Bogdan Pavel, Leon Zagrean, and Ana-Maria Zagrean. 2020. "Melatonin’s Impact on Antioxidative and Anti-Inflammatory Reprogramming in Homeostasis and Disease" Biomolecules 10, no. 9: 1211. https://doi.org/10.3390/biom10091211

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