Open access peer-reviewed chapter - ONLINE FIRST

Sirtuins and Melatonin: Linking Chronobiology to Inflammation and Aging

Written By

Anca Ungurianu, Cristina Manuela Drăgoi, Alina Crenguța Nicolae, Ion-Bogdan Dumitrescu, Daniela Grădinaru and Denisa Margină

Submitted: 27 September 2023 Reviewed: 22 November 2023 Published: 16 March 2024

DOI: 10.5772/intechopen.1003914

Advances in Geriatrics and Gerontology - Challenges of the New Millennium IntechOpen
Advances in Geriatrics and Gerontology - Challenges of the New Mi... Edited by Sara Palermo

From the Edited Volume

Advances in Geriatrics and Gerontology - Challenges of the New Millennium [Working Title]

Ph.D. Sara Palermo

Chapter metrics overview

34 Chapter Downloads

View Full Metrics

Abstract

In recent years, the intricate interplay between sirtuins and melatonin has emerged as a fascinating area of research, with profound implications on various aspects of human health. This comprehensive chapter delves into the complex relationship between sirtuins and melatonin, as well as their essential roles in the regulation of circadian rhythms, inflammation, and aging. The attention is primarily directed to their impact on a range of critical health focal points, including cardiovascular diseases, central nervous system disorders, metabolic imbalances, musculoskeletal disorders, neoplasms, and the overarching process of aging, detailing all the complex biochemical mechanisms and physiological pathways that validate the intimately tailored functional relationship between the indoleamine hormone synthesized in the pinealocytes and the NAD+-dependent histone deacetylases. These two components interact in complex ways, influencing processes such as cellular homeostasis, oxidative stress, and inflammatory cascade regulation. Age-related reductions in SIRT1 expression, influenced by melatonin levels, can deeply impact cellular functions. By elucidating the complex connections between sirtuins, melatonin, and chronobiological processes, we contribute to a deeper understanding of the fundamental mechanisms that trigger inflammation and aging-related diseases, and in the meantime underscore the promising avenues for future research and clinical interventions aimed at enhancing human health and extending the quality of life.

Keywords

  • sirtuin
  • melatonin
  • inflammation
  • aging
  • metabolic diseases

1. Introduction

Melatonin is mainly known for its involvement in sleep and circadian rhythm regulation, among other neuroendocrine processes [1], with reported anti-inflammatory, antioxidant, and antitumor effects [2, 3]. Melatonin is a hormone synthesized by the pineal gland that subsequently enters the bloodstream, enabling its distribution throughout various bodily systems. Moreover, it has the capability to penetrate the third ventricle of the brain via the pineal recess [4]. Melatonin receptors are mainly found throughout the central nervous system (CNS) and in immune cells, with various effects. Aside from its well-known role in circadian rhythm and sleep regulation, melatonin also acts as an anti-excitatory molecule in the CNS and is involved in the regulation of metabolic pathways, modulation of hormone secretion and of pro- and anti-inflammatory cytokines release, and it can even directly activate monocytes [45]. Moreover, it maintains redox homeostasis by upregulating antioxidant enzymes, downregulating reactive oxygen- (ROS) and reactive nitrogen species (RNS)-generating enzymes, and also via its mitochondria-protective effects [4, 6, 7, 8].

Sirtuins are NAD+-dependent enzymes with numerous physiological functions, regulating energy metabolism, inflammation, stress response, DNA repair, cell survival, and also being involved in circadian rhythms [9, 10]. Moreover, recent literature data links the sirtuin family to neurodegenerative, inflammation, and aging-associated diseases [11]. In humans, this enzyme family comprises seven isoforms, with different subcellular distribution and functions. Three enzymes are nuclear—SIRT1, SIRT6 and SIRT7, three mitochondrial—SIRT3, SIRT4, and SIRT5, and one cytosolic—SIRT2 [9]. However, SIRT1 often shuttles to the cytoplasm, while SIRT2 and SIRT3 can migrate to the nucleus, under certain conditions [9, 11]. The nuclear sirtuins are transcriptional and epigenetic regulators, stabilizing chromatin and deacetylating histones and non-histone proteins, such as transcriptional factors or DNA repair proteins [12, 13, 14]. They also modulate stress and oxidative stress response, maintain telomere integrity, and regulate apoptosis [9]. SIRT2 intervenes in several cellular processes, including cell cycle, apoptosis, DNA repair, metabolism, and senescence [11]. The mitochondrial sirtuins are mainly involved in metabolic regulation, energy metabolism, and mitochondrial function, maintaining redox and energy homeostasis [11].

Thus far, two sirtuin isoforms, SIRT1 and SIRT3, seem to be essential for the normal functioning of the circadian system, via multiple cellular pathways [4]. SIRT1 was established as a secondary mediator of melatonin’s cellular actions, as numerous in vivo and in vitro studies confirmed its upregulation by melatonin [15]. Also, melatonin signaling in a SIRT1-mediated way is supported by the lack of melatonin effects in the case of SIRT1 inhibition or knockdown [16]. SIRT1 was reported to interact with the core circadian oscillator complex BMAL1:CLOCK (basic helix-loop-helix ARNT-like 1: circadian locomotor output cycles kaput), as to intervene in the positive feedback loop involving nicotinamide phosphoribosyltransferase (NAMPT) and nicotinamide adenine dinucleotide (NAD+), influencing the expression of the period circadian regulator 2 (Per2) gene, a central player in circadian rhythm regulation [17, 18, 19]. SIRT3 can also intervene in the NAD+ cycle, linking circadian rhythms to mitochondrial oxidative metabolism [20]. SIRT3 plays a pivotal role in mitochondrial antioxidant defense, increasing the expression of superoxide dismutase 2 (SOD 2) and catalase, two enzymes of paramount importance in counteracting the deleterious effects of oxidative stress [21, 22]. Moreover, intracellularly, melatonin is primarily concentrated in the mitochondria, its concentration in this organelle being higher than in any other [23]. Mitochondrial melatonin is not released in the systemic circulation and its synthesis is independent of light exposure [24]. Both melatonin and SIRT3 were reported to fight against oxidative stress by enhancing the expression of antioxidant enzymes [24, 25, 26, 27, 28], and melatonin’s antioxidant effects seem to be SIRT3-mediated [29]. Consequently, its mitochondrial accumulation goes hand in hand with its antioxidant and antitumor actions, contributing to the maintenance of redox homeostasis and combatting malignant cell transformation [23].

Melatonin can act both as a pro-inflammatory and an anti-inflammatory molecule. This duality might come as a surprise, however, just as with other hormones, its function may vary under different conditions and when concerning various cell types [16, 30]. Its pro-inflammatory effect can be deemed beneficial when considering its action as an immune stimulatory agent concerning leukocytes and their ability to fight off pathogens [30, 31, 32] while proving detrimental in autoimmune maladies. The anti-inflammatory effects usually take center stage as they can be the basis of melatonin-based therapies in diseases with a low-grade inflammatory component, such as neurodegenerative or metabolic diseases, or characterized by high-grade inflammation, such as ischemia-reperfusion or brain injury and sepsis [16, 33, 34].

In this chapter, we aimed to construct a summary of the current state of understanding on a wide topic concerning the link between melatonin’s effects and sirtuin signaling, concerning regulation of circadian rhythms, inflammation, and aging, in the most prevalent noncommunicable diseases currently associated with increased mortality and morbidity, selecting the most relevant, novel, and comprehensive research previously published by other scientists. The attention was primarily focused on their impact on cardiovascular diseases, central nervous system disorders, metabolic imbalances, neoplasms, and the process of aging, detailing the complex biochemical mechanisms involved.

Advertisement

2. Cardiovascular diseases

The imbalance of melatonin, which is one of the master regulators of the internal clocks in humans, is clearly associated with an increased risk of diseases, correlated with impaired sleep and aging-associated pathology, mainly cardiovascular, metabolic, and neurodegenerative disease [35, 36, 37, 38, 39, 40, 41, 42].

One of the main pathways responsible for the correlation between melatonin and age-related chronic disease is represented by sirtuins [36, 37]. The interplay between the circadian machinery and sirtuins promotes cardiac health in a complex biochemistry of regulatory systems, mainly by modulating metabolic homeostasis and cell death or survival genes and influencing energy metabolism [43]. Recent research suggested that sirtuins in general, but SIRT1 in particular, have a crucial role in connecting the cellular metabolism to the circadian/internal clock [43, 44].

SIRT1 is directly implicated in the mechanistic development of cardiomyocytes, being responsible for regulating the voltage-gated cardiac sodium ion channels, reducing the risk of atherosclerotic plaque build-up, protection against oxidative damage, and lowering thrombotic risk [43, 45].

Melatonin is an amphiphilic molecule, so it can be found in all subcellular components, with a high concentration in cellular and subcellular membranes [8, 24, 46, 47]. As a result, it has the ability to act as a stabilizer of membrane processes acting against lipid peroxidation and oxidative impairment of mitochondrial DNA [28, 48, 49]. Melatonin is concentrated in the mitochondria and, as a consequence, it improves the electron transport chain efficiency and stimulates ATP production [50]. Its subcellular localization is somewhat overlapping with SIRT isoforms, supporting the intertwining of their signaling pathways; for example, recent data argues that melatonin and SIRT3 may act synergistically in regulating free radical generation and shielding mitochondria from oxidative damage [24].

Melatonin acts through different signaling pathways, either membrane- or organelle-focused, influencing the dynamics of physiological processes and protecting from pathological shifts. One of the key pathways modulated by melatonin concerning its protective actions is represented by modulating SIRT1 expression [9, 11]. Melatonin induces the transcriptional activation of nuclear factor erythroid 2-related factor 2 (Nrf2) and, consequently, antioxidant response element (ARE) through a SIRT1-dependent mechanism [51, 52]. Nrf2 is transcription factor that is able to bind to DNA and regulate the gene expression concerning antioxidant defense, as part of a master antioxidant and cytoprotective pathway, also inhibiting inflammation-enhancing signaling, such as the NLR family pyrin domain containing 3 (NLRP3) inflammasome [53, 54, 55].

Moreover, melatonin as well as its metabolites acts as ROS scavengers, stimulating the synthesis of antioxidant enzymes [7, 25, 28, 56]. Owing to its antioxidant action, melatonin was able to protect against ischemia-reperfusion injury in all organs, the activation sirtuins being most likely involved [10]. In a model of ischemia-reperfusion injury, the protective effects exerted by melatonin were dependent on the mitochondrial SIRT3. Melatonin’s action was correlated with the stimulation of the adenosine monophosphate-activated kinase (AMPK)—peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α)—SIRT3 signaling, the activation of mitochondrial SOD and the enhancement of Nrf2 and mitochondrial transcription factor A (TFAM) expression [24, 57]. AMPK has a pivotal role in energy metabolism and homeostasis, adapting cell response to stress and nutrient availability. Recently, it was reported that AMPK functions as a redox sensor, also influencing autophagy, cell proliferation, and apoptosis, seemingly being involved in cardiovascular health and disease [58, 59]. PGC-1α is a key regulator of mitochondrial metabolism, being central to quite a few cellular pathways combating oxidative stress and inflammation [60]. This molecule is a crucial factor in the cellular stress response in the ischemic myocardium [60, 61]. TFAM is a mitochondrial DNA-binding protein vital for the maintenance of the mitochondrial genome, involved in the inflammatory stress response. An altered TFAM function was linked to pathological changes, especially in neurodegenerative diseases and aging [62, 63]. SIRT3 inhibition hinders mitochondrial SOD2 upregulation, leading to oxidative stress, which prevents melatonin’s ability to protect the myocardium from free radical destruction [64].

Melatonin attenuates sepsis-induced myocardial injury by inhibiting caspase-3-induced apoptosis via SIRT1 activation [65]. Caspase-3 is a protease involved in tissue differentiation and regeneration, neural development, and, most famously, cell apoptosis, being possible target in the therapy of cardiovascular diseases, neurodegenerative disorders, and malignancies [66, 67]. Melatonin also exerts anti-inflammatory SIRT1-dependent effects, as the downregulation/inhibition of SIRT1 was reversed under the effect of melatonin in a H2O2-induced pro-inflammatory cell model [68]. On the other hand, experimental research shows that melatonin upregulates sirtuins, with a consequent downregulation of transcription for pro-inflammatory proteins and kappa-light-chain-enhancer of activated B cells (NF-κB) by suppressing the activation of toll-like receptor 4 (TLR4) and NLRP3 inflammasome [4, 69]. TLR4 activation leads to pro-inflammatory signaling (i.e., NF-κB) and synthesis of pro-inflammatory cytokines [70, 71]. NLRP3 inflammasome is a protein complex that assembles in response to cellular stress, promoting inflammation; its chronic aberrant activation is part of the etiopathogenesis of numerous diseases characterized by low-grade inflammation [72].

Furthermore, melatonin downregulates inflammation-associated enzymes such as inducible nitric oxide synthase (NOS) and cyclooxygenase 2 (COX-2), leading to lower levels of pro-inflammatory molecules, also contributing to an increase anti-inflammatory cytokines (e.g., interleukin 10, IL-10), thus exerting a protective effect against cardiovascular, metabolic, and autoimmune disease, which are all associated with oxidative stress and inflammation [73, 74, 75, 76]. In a model of apolipoprotein E-deficient mice, melatonin decreased endothelial impairment, as well as the loss of SIRT1 and endothelial NOS activities, lowered tumor protein p53 and endothelin-1 expression. Administering melatonin formulated as a long-release dose and was more effective in counteracting endothelial dysfunction through multiple mechanisms, including SIRT modulation [46, 77].

Human studies confirm the correlation between melatonin, sirtuins, and the risk of cardiovascular disease. A clinical trial published in 2017 showed that the time of day (morning vs. afternoon) when patients underwent isolated aortic valve replacement interventions clearly influenced the overall survival, with a direct advantage of patients involved in afternoon intervention, who were characterized by fewer post-interventional events; also, that hypoxia-reoxygenation tolerance of the human myocardium is higher in the afternoon [35, 78]. These results confirm the observations regarding the higher incidence rate of cardiovascular events (myocardial infarction, stroke, arrhythmias, and sudden cardiac deaths) in the morning than in the evening. Also, from a chronotherapeutic perspective, the efficacy of antihypertensive treatments is higher when administered in the afternoon, according to both animal and human studies [41, 79, 80, 81].

There are several studies supporting the synergistic effects of melatonin and sirtuins, with cardiovascular beneficial outcome through antioxidant and anti-inflammatory mechanisms; in experimental/preclinical studies mimicking severe pathology, such as cardiac ischemia-reperfusion of normal and diabetic rats, endoplasmic reticulum stress in cardiomyocytes, lipopolysaccharide (LPS)-treated microglial cell lines, in brain injury by cecal ligation/puncture in mice, these results are confirmed [10, 51, 69, 82, 83]. This synergy is also supported by results showing that melatonin effects are antagonized by sirtuin inhibitors or by silencing the protein.

Advertisement

3. CNS disorders

CNS disorders encompass a wide variety of diseases from neurodegenerative diseases, such as Alzheimer’s, Parkinson’s, or multiple sclerosis, to neuropsychiatric disorders, such as depression, anxiety, or substance abuse, all being associated with a decreased quality of life [84, 85, 86]. Their etiopathogenesis is very complex, with multiple processes and alterations being involved, including neuroinflammation, disruption of autophagy, protein and lipid metabolism, redox, and energy and circadian homeostasis [86, 87, 88].

SIRT1 is widely expressed in the CNS, with anti-inflammatory and neuroprotective actions in numerous neurodegenerative diseases experimental models [1], and its recent link to melatonin signaling opens a new research path in the therapy of neurodegenerative diseases [89].

One of the mechanisms of melatonin’s neuroprotective effect involves the increase of SIRT1 expression and the activation of SIRT1/Nrf2 pathway and the inhibition of the NLRP3 inflammasome [90, 91]. These might prove pivotal in the melatonin-based therapeutic approaches of some CNS disorders, seeing as the NLRP3 inflammasome is involved in the development or progression of neurodegenerative diseases [92], ischemia-reperfusion injury [93], traumatic brain injury [94], and cerebral tumors [95].

Neuroinflammation can be triggered by numerous factors, such as cellular damage or pathogens, and results into extracellular matrix damage and immunological reactions that can ultimately lead to neuronal oxidative damage and neurotransmitter dysfunction [1]. Further, the decrease in melatonin results in circadian dysregulation, decreased antioxidant defense, and alteration of normal mitochondrial functioning [26].

Inflammation is central to the pathogenesis of major depressive disorder (MMD) [91]. SIRT1 plays an important role in numerous cellular processes, including inflammation, in the hippocampus and central cortex [11], being recently linked to depression [96]. Mediation of inflammation by SIRT1, mainly via NF-κB and NLRP3 inflammasome downregulation, was shown to alleviate depression and anxiety-related behavioral deficits [91, 97, 98]. The NF-κB family of transcription factors is a key regulator of inflammation, immune responses, and cell proliferation, which, along with the NLRP3 inflammasome, contributes to amplifying inflammation [99, 100, 101].

Mitochondrial changes seem to play a crucial role in the development of MDD [26], and sirtuins are key regulators of mitochondrial processes, seeing as three of the seven family members are mitochondria-based [9]. The enhancement of the SIRT1–PGC-1α pathway is another signaling route via which melatonin exerts its protective effects, this time mitochondria being the main target, as PGC-1α is known as the master mitochondrial regulator, increasing their biogenesis and function [26]. SIRT1 enhancement by melatonin as a secondary signaling pathway [89] could correct some of the oxidative, mitochondrial, and neurotransmitter imbalances characteristic for depression and other neuropsychiatric disorders [102, 103], and contribute to uncovering more of the cellular pathways involved in melatonin’s antidepressant-like effect [104, 105].

Neurodegenerative disease diagnosis had a sharp escalation in the last decades, increasing elderly morbidity and mortality [35]. Inflammation is part of normal aging; however, it is also part of the pathogenesis of several maladies, including neurodegenerative diseases [4]. This type of neuroinflammation is not of an infectious cause but entails moderate, slowly progressing microglia activation, supported by oxidative stress and mitochondria dysfunction, encompassing immune cells, astrocytes, and neurons [4].

Brain inflammation is a hallmark of neurodegenerative diseases, most notably Alzheimer’s disease (AD) [4]. Both AD and Parkinson’s disease (PD) are associated with an altered circadian rhythm, alongside impaired homeostasis of redox and inflammatory processes [4, 106]. AD is the most prevalent form of dementia in the elderly, being characterized by modified sleep patterns, abnormal melatonin secretion, and circadian dysregulation [26, 107, 108], shifting sleeping habits being reported early in its progression [35]. SIRT1 is an important link between circadian rhythm and redox homeostasis [35]. Melatonin is intimately linked to SIRT1 function in aging cells [109, 110]. Age-associated NAD+ and SIRT1 deficiency, changes which are observed in neurodegenerative diseases also, are associated with mitochondrial dysfunction, autophagy, and circadian rhythm alterations, which can be reversed by melatonin [4, 111]. In preclinical and in vitro neurodegenerative diseases models, melatonin proved beneficial [112], while in clinical settings results varied [112], but the majority of studies reported improved sleep quality and reduced daytime sleepiness, stabilizing the circadian rhythm, and slowing down the progression of cognitive impairment [113, 114, 115, 116, 117].

Traumatic brain injury is a worldwide leading cause of mortality and morbidity, with debilitating long-term sequels. Sleep alterations are among the most common long-term post-injury implications. Animal studies showed that melatonin improved cognition as well as behavior; it also reduced post-injury cognitive decline and the risk of developing dementia, while human studies are scarce [118, 119]. The development of secondary injury following traumatic brain injury is dependent on the inflammatory response in the cerebral cortex, the NLRP3 inflammasome playing a central part [120, 121, 122]. SIRT1 was reported to have a protective role against traumatic brain injury, seeing as it mitigates oxidative stress and ROS production, which can, in turn, activate the NLRP3 inflammasome [1]. Further, resveratrol, a well-known SIRT1 activator [11], attenuated inflammation and oxidative stress by suppressing the NLRP3 inflammasome in a SIRT1-dependent manner [120]. Taking into consideration the melatonin-SIRT1 relationship, this neurohormone is a possible candidate as an additional therapeutic option in traumatic brain injury [118, 119].

Advertisement

4. Metabolic imbalances

Metabolic diseases, such as diabetes mellitus, metabolic syndrome, and obesity, have exponentially increased in the last decades, posing a serious threat to human health. They are characterized by inflammation and oxidative stress, along with impairments of cell metabolism, energy homeostasis, insulin secretion and function, and microbiota alterations [123, 124, 125, 126].

Melatonin is involved in energy metabolism pathways and regulates epigenetic processes in neuronal cells, being biochemically interconnected with signaling pathways responsible for adjusting energy metabolism, such as insulin/insulin-like growth factor 1 (IGF-1), Forkhead box O (FoxO), and sirtuin pathways [110, 127, 128, 129, 130, 131]. Alterations of the expression and activity of circadian rhythm components are commonly found in patients with neurodegenerative, metabolic disorders, and cancer [37, 127, 132]. Also, melatonin levels and CLOCK expressions are reduced in patients with neurodegenerative and metabolic disorders [133, 134, 135, 136, 137, 138, 139].

All these pathological impairments have an underlying component of oxidative stress and mitochondrial function failure. Sirtuins, and especially SIRT1, as well as the peroxiredoxin protein family, are directly involved in the relationship between redox homeostasis and circadian rhythm, regulated by melatonin [37127132, 133, 134, 135, 136, 137, 138, 139].

Metabolic syndrome and diabetes are associated with oxidative stress and inflammation, reunited under the umbrella of inflammaging, and would clearly benefit from the melatonin/SIRT synergy [69], seeing as melatonin is a key player in energy sensing/energy expenditure and body weight regulation. Animal studies showed that removing the pineal gland from rats led to a body weight increase that could be reversed by exogenous melatonin administration, along with a decrease of visceral fat; the results were found in animals fed either high fat or high fructose diets [140, 141]. Also, rat pinealectomy was associated with decreased insulin sensitivity and reduced glucose transporter type 4 (GLUT4) gene expression [142, 143]. In animal models, melatonin, as well as selective melatonin receptor agonists, induced a reduction of body weight and blood pressure, increased insulin sensitivity, and restored lipid homeostasis [34, 144]. These preclinical reports, among others [145, 146], highlight the potential of melatonin therapy in improving glucose metabolism and contribute to diabetes mellitus prevention [145, 146].

Impairments of melatonergic signaling due to genetic polymorphism support the development of a prediabetic status, type 2 diabetes, elevated cholesterol, triglycerides, and coronary heart disease; mice knocked out for the melatonin receptor MT1 or with pinealectomy exhibit insulin resistance [137, 142, 143, 147]. These metabolic alterations were reversed by melatonin, which decreased pro-inflammatory signaling (TNF-α, IL-1β) and inducible NOS by suppressing NF-κB expression in a SIRT-dependent manner [44, 148, 149].

Human studies confirm the metabolic protective action of melatonin, reporting antihyperlipidemic effects and a reduced insulin release (via pancreatic β-cells receptors), also contributing to alleviating metabolic syndrome via SIRT regulation, enhancing antioxidant and anti-inflammatory pathways [139, 150]. In type 2 diabetic patients low-circulating levels of melatonin were found, as well as increased mRNA for the melatonin membrane receptor [150, 151], while genetic variations of melatonin receptors are associated with impaired levels of fasting blood glucose and increased risk of type 2 diabetes, and also with polycystic ovary syndrome [45, 152, 153, 154]. Also, coronary artery disease patients show decreased melatonin levels; exogenous melatonin was effective in reducing blood pressure and cardiovascular rhythm alterations, preserving the availability of nitric oxide and yielding anti-remodeling cardiac effects, thus providing cardiovascular protection in metabolic syndrome patients [45, 137, 155]. Controlled clinical studies confirmed the antihypertensive properties of melatonin, and also underlined its ability to improve lipid profiles, with an increase of HDL, in metabolic syndrome patients [156].

Advertisement

5. Musculoskeletal disorders

Skeletal muscle is essential for posture and movement, but it is also directly involved in glucose uptake, thermal regulation, and nutritional balance, among other important physiological roles [157, 158, 159]. Therefore, deterioration of skeletal muscle mass is associated with impaired glucose homeostasis, and not only with posture/movement-associated difficulties (falls, fractures, disability) [160]. Moreover, skeletal muscle ailments are considerably increasing in aging, thus bringing up the costs of healthcare and having a negative impact on the quality of life [159].

Melatonin was reported to support muscle activity through its ability to maintain mitochondrial function, alongside oxidative stress reduction and inhibition of cardiolipin peroxidation [15, 24, 69]. Cardiolipin is a dimeric phospholipid found in the inner mitochondrial membrane that undergoes oxidation and translocation to the cytosolic side of the outer mitochondrial membrane under oxidative stress conditions, signaling a dysfunctional mitochondria [161, 162]. In Refs., [163, 164] dystrophic muscle diseases are biochemically characterized by inflammation, redox imbalance, and mitochondrial dysfunction, and could benefit from melatonin treatment [160, 165]. This is attributable to its lipophilic nature, making it possible to pass through cells and mitochondrial membranes and the blood-brain barrier, as well as its effect as a calcium homeostasis regulator during muscle contraction [166, 167]. When administered as a nutraceutical in preclinical, but also in clinical studies, it improved muscle metabolism and strength [163, 164]. These positive effects are also pointed out in age-related sarcopenia and muscle weakness [160].

Chronic melatonin administration in rat and mouse models of muscle injury reduced apoptosis, increased twitch force, and accelerated the regeneration of satellite cells. Women with fibromyalgia benefit from melatonin administration which induces reduction of symptoms such as chronic muscular pain, cognitive dysfunctions, and sleep disorders [159, 168, 169].

Calpain is a receptor of calcium, found in the cytoplasm of skeletal muscle cells in an inactive form, being controlled by intracellular calcium ion concentration and calpain inhibitory protein. An increase in the skeletal muscle cells’ cytoplasmic Ca2+ concentration activates calpain, resulting in the hydrolysis of skeletal muscle fibers, leading to reduced contractility. Melatonin was reported to inhibit calpain, but more in-depth studies are required to establish its clinical potential [170, 171, 172].

In Refs., [159, 168, 169] literature data reveal melatonin to be a promising agent for muscle regeneration and maintenance, with a possible use in chronic diseases, especially those associated with aging, sirtuins being just one of the signaling pathways involved. Nevertheless, further studies, both preclinical and clinical, are needed to establish its muscle-protective mechanisms and clinical use aspects.

Advertisement

6. Neoplasms

Malignancies have an ever-increasing prevalence and a cancer diagnosis has a severe impact on the quality of life and mental well-being of patients [173]. The antitumor effect of melatonin was reported in different types of cancer, interfering with various cancer hallmarks, mitigating cancer initiation, progression, and metastasis [174, 175].

The disruption of circadian rhythm due to exposure to excessive light or frequent long-distance travel entails an alteration of melatonin synthesis and secretion, with an associated increased risk of cancer development [176, 177]. In vitro studies in breast cancer cells showed that exposure to white fluorescent light led to decreased melatonin levels and increased tumor growth [178], while the blood of volunteers exposed to white fluorescent light during nighttime had lower melatonin levels and proved a better tumor growth medium [178].

Melatonin administration decreased proliferation parameters and induced a reduction in tumor growth, concomitantly downregulating SIRT1 [179]. The downregulation or inhibition of SIRT1 led to increased pro-oxidant and antitumor activity [180, 181], while its activation decreased melatonin’s anticancer action [182].

The relationship between SIRT1 and melatonin in cancer cells is opposite to that in nontumor cells, melatonin acting as an inhibitor of SIRT1 activity [44]. This might come as a surprise, but seeing as SIRT1 is overexpressed in some types of cancer [183, 184], a context-specific role for melatonin in regulating the activity of this sirtuin is plausible [185]. The dual role of melatonin concerning SIRT1 regulation in normal and malignant cells seems to entail its ability to either stimulate or inhibit the activity of SIRT1. Moreover, this regulation might not only target cell proliferation but also the control of circadian regulation genes, such as BMAL1 or Per2, which are key players in maintaining tissue homeostasis [90, 185].

Advertisement

7. Aging

Aging is a ubiquitous phenomenon that encompasses numerous biological changes that, in time, lead to the decline of an organism [186]. In humans, aging entails a gradual accumulation of physical and cognitive alterations, with an increased risk of developing various maladies, such as cardiovascular, metabolic, or neurodegenerative diseases and malignancies [186, 187, 188]. These often cause a marked decline in the quality of life, being associated with higher morbidity and mortality.

Aging is associated with an alteration of circadian rhythm synchrony and reduced secretion of melatonin [1, 35]. Also, a reduction of SIRT1 activity was observed in senescence, while its inhibition abolished a number of melatonin’s cellular effects [69]. Lower SIRT1 levels were observed in the suprachiasmatic nucleus (SCN) of aging mice, affecting the functioning of the core circadian oscillator BMAL1:CLOCK, while its overexpression prevented aging-depending circadian rhythm alterations and its silencing in young animals decreased BMAL1 and Per2 gene expression [189].

Low-grade inflammation is a major component of physiological aging, especially considering its association with the alteration of brain function, neurodegeneration, and mood disorders [137, 190]. The contribution of inflammation to the aging process is known as inflammaging [4, 16]. Apart from playing a central role in longevity, regulating cellular processes as cell cycle, apoptosis, or DNA repair, SIRT1 is involved in modulating antioxidant and anti-inflammatory processes [11]. SIRT1 seems to be an important factor in trying to assess the extent of melatonin’s effects on aging and aging-associated low-grade inflammation [1]. Moreover, melatonin enhances the antioxidant defense of senescent cells, regulating redox homeostasis. A central player in this effect is SIRT1, whose upregulation results in the increased expression of antioxidants via Nrf2 and FOXO pathways, modulating mitochondrial ROS production and autophagy, while inhibiting NF-κB signaling [35].

Despite all these, some conflicting results regarding the effect of melatonin treatment on SIRT2 activity were reported in preclinical models of aging. One research group found no effect on SIRT2 in neurons from the dentate gyrus [191], while another group observed that melatonin treatment led to a decrease in SIRT2 activity in the hippocampus of adult rats [192], and in the colon and hippocampus of aged rats [193, 194], reducing oxidative stress parameters and pro-apoptotic proteins.

Both the pro-inflammatory effect of melatonin, as well as the anti-inflammatory, must be considered when addressing its potential use in mitigating some aging-associated signs and symptoms [69]. Most data are supportive of its beneficial, anti-inflammatory actions. However, some reports concerning autoimmune diseases, such as rheumatoid arthritis or multiple sclerosis, bring to the fore its possible detrimental effects [195, 196, 197]. Its protective actions fall mainly under the umbrella of the above-mentioned and well-documented antioxidant and anti-inflammatory effects, along with its stimulation of the immune system, promoting healing and maintaining homeostasis [187, 188, 198, 199]. A special melatonin-mediated pathway, central to the aging process, is the enhancement of SIRT1 activity.

Advertisement

8. Discussion

Melatonin, a hormone primarily synthesized in the pineal gland, has emerged as a critical regulator of circadian rhythms and a multifunctional molecule with antioxidant, anti-inflammatory, and neuroprotective properties, involving numerous cellular signaling pathways (Figure 1).

Figure 1.

An overview of the SIRT1-mediated melatonin effects in noncancerous and malignant cells. ROS-reactive oxygen species, AO-antioxidant, BMAL1:CLOCK-basic helix-loop-helix ARNT-like 1: circadian locomotor output cycles kaput, Per2-period circadian regulator 2 gene, PGC-1α-peroxisome proliferator-activated receptor gamma coactivator 1-alpha, Nrf2-nuclear factor erythroid 2-related factor 2, FOXO-Forkhead box O, NF-κB-kappa-light-chain-enhancer of activated B cells, NLRP3-NLR family pyrin domain containing 3 inflammasome.

The anti-inflammatory activity of melatonin involves both immunological and non-immunological processes [16]. The latter mainly includes protection against oxidative stress by promoting antioxidant defense and decreasing the formation of reactive oxygen and nitrogen species, and the preventing mitochondrial dysfunction [16]. It contributes to an anti-inflammatory pathway involving sirtuin activation, namely SIRT1, Nrf2 upregulation, and nuclear factor NF-κB downregulation [1, 69]. Also, it was reported to downregulate COX-2 and neuronal NOS, to prevent TLR4 and NLRP3 inflammasome activation [16]. These resulted in an increased secretion of anti-inflammatory cytokines and decreased production of ROS and pro-inflammatory cytokines [1, 16].

Exploring the melatonin-sirtuins interaction holds significant promise in advancing our understanding of their joint impact on human health. On the other hand, sirtuins, a family of deacetylase enzymes, play fundamental roles in cellular processes such as gene expression, DNA repair, and stress response. The interplay between melatonin and sirtuins has been implicated in a spectrum of biological phenomena, ranging from circadian rhythm regulation to cellular homeostasis and physiological aging. Investigating the intricate crosstalk between melatonin and sirtuins has the potential to unlock novel insights into the mechanisms governing these processes and different pathological milieu modulation.

Moreover, understanding how melatonin influences sirtuins activity and vice versa could pave the way for the development of innovative therapeutic strategies targeting a wide array of health conditions, including sleep disorders, cardiovascular and metabolic diseases, neurodegenerative disorders, and cancer. These melatonin-sirtuins studies not only shed light on the fundamental principles of circadian biology and cellular physiology but also offer promising avenues for enhancing human health and well-being.

Advertisement

9. Outlook

Melatonin is considered one of the master regulators of the circadian rhythm, being intensively studied for its pleiotropic effects concerning redox imbalance, inflammation, immune response, aging, cell proliferation, and even fertility. The present chapter aims to critically analyze the latest scientific information regarding the interplay between sirtuins and melatonin in order to better understand the role of this complex system and its potential modulation in preventing/treating various afflictions. As a result, we pointed out that sirtuin signaling is directly involved in the cardio- and neuroprotective effects attributed to melatonin, as well as its ability to support musculoskeletal function and regeneration and to restore metabolic and energy homeostasis. Regarding malignancies, the relationship between SIRT1 and melatonin in cancer cells is opposite to that in non-tumor cells, with an overall antitumor action. All these reported effects are integrated as important pathways, justifying the protective effect of melatonin in aging-associated pathology through SIRT-mediated pathways. In this complex picture, there is an acute need for further studies to substantiate all these scientific claims, since there is a great imbalance between in vitro, preclinical and clinical studies, for each of the above-mentioned effects. Also, a systematic review of the latest literature data, encompassing the cellular pathways through which melatonin modulates physio-pathological processes, focusing on the interconnection with sirtuins, is highly needed considering the current heterogeneous research output.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Chen H et al. Involvement of the SIRT1-NLRP3 pathway in the inflammatory response. Cell Communication and Signaling: CCS. 2023;21(1):185
  2. 2. Tordjman S et al. Melatonin: Pharmacology, functions and therapeutic benefits. Current Neuropharmacology. 2017;15(3):434-443
  3. 3. Zhou H et al. Melatonin suppresses platelet activation and function against cardiac ischemia/reperfusion injury via PPARgamma/FUNDC1/mitophagy pathways. Journal of Pineal Research. 2017;63(4)
  4. 4. Hardeland R et al. Melatonin and brain inflammaging. Progress in Neurobiology. 2015;127-128:46-63
  5. 5. Hardeland R. Melatonin and the theories of aging: A critical appraisal of melatonin’s role in antiaging mechanisms. Journal of Pineal Research. 2013;55(4):325-356
  6. 6. Drăgoi CM, Nicolae AC, editors. Melatonin - Molecular Biology, Clinical and Pharmaceutical Approaches. IntechOpen. 2018. DOI: 10.5772/intechopen.74993
  7. 7. Galano A, Tan DX, Reiter RJ. On the free radical scavenging activities of melatonin’s metabolites, AFMK and AMK. Journal of Pineal Research. 2013;54(3):245-257
  8. 8. Menendez-Pelaez A, Reiter RJ. Distribution of melatonin in mammalian tissues: The relative importance of nuclear versus cytosolic localization. Journal of Pineal Research. 1993;15(2):59-69
  9. 9. Ungurianu A, Zanfirescu A, Margina D. Regulation of gene expression through food-curcumin as a Sirtuin activity modulator. Plants (Basel). 2022;11(13)
  10. 10. Yu L et al. Melatonin receptor-mediated protection against myocardial ischemia/reperfusion injury: Role of SIRT1. Journal of Pineal Research. 2014;57(2):228-238
  11. 11. Ungurianu A, Zanfirescu A, Margina D. Sirtuins, resveratrol and the intertwining cellular pathways connecting them. Ageing Research Reviews. 2023;88:101936
  12. 12. Toiber D, Sebastian C, Mostoslavsky R. Characterization of nuclear sirtuins: Molecular mechanisms and physiological relevance. Handbook of Experimental Pharmacology. 2011;206:189-224
  13. 13. Soetikno V et al. Curcumin prevents diabetic cardiomyopathy in streptozotocin-induced diabetic rats: Possible involvement of PKC-MAPK signaling pathway. European Journal of Pharmaceutical Sciences. 2012;47(3):604-614
  14. 14. Houtkooper RH, Pirinen E, Auwerx J. Sirtuins as regulators of metabolism and healthspan. Nature Reviews. Molecular Cell Biology. 2012;13(4):225-238
  15. 15. Hardeland R. Melatonin and the pathologies of weakened or dysregulated circadian oscillators. Journal of Pineal Research. 2017;62(1)
  16. 16. Hardeland R. Melatonin and inflammation-story of a double-edged blade. Journal of Pineal Research. 2018;65(4):e12525
  17. 17. Ramsey KM et al. Circadian clock feedback cycle through NAMPT-mediated NAD+ biosynthesis. Science. 2009;324(5927):651-654
  18. 18. Nakahata Y et al. Circadian control of the NAD+ salvage pathway by CLOCK-SIRT1. Science. 2009;324(5927):654-657
  19. 19. Imai S. “Clocks” in the NAD world: NAD as a metabolic oscillator for the regulation of metabolism and aging. Biochimica et Biophysica Acta. 2010;1804(8):1584-1590
  20. 20. Peek CB et al. Circadian clock NAD+ cycle drives mitochondrial oxidative metabolism in mice. Science. 2013;342(6158):1243417
  21. 21. Tao R et al. Sirt3-mediated deacetylation of evolutionarily conserved lysine 122 regulates MnSOD activity in response to stress. Molecular Cell. 2010;40(6):893-904
  22. 22. Rangarajan P et al. Sirtuin 3 regulates Foxo3a-mediated antioxidant pathway in microglia. Neuroscience. 2015;311:398-414
  23. 23. Reiter RJ et al. Melatonin: A mitochondrial resident with a diverse skill set. Life Sciences. 2022;301:120612
  24. 24. Reiter RJ et al. Melatonin mitigates mitochondrial meltdown: Interactions with SIRT3. International Journal of Molecular Sciences. 2018;19(8)
  25. 25. Allegra M et al. The chemistry of melatonin’s interaction with reactive species. Journal of Pineal Research. 2003;34(1):1-10
  26. 26. Anderson G. Linking the biological underpinnings of depression: Role of mitochondria interactions with melatonin, inflammation, sirtuins, tryptophan catabolites, DNA repair and oxidative and nitrosative stress, with consequences for classification and cognition. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2018;80(Pt C):255-266
  27. 27. Drăgoi CM, Nicolae AC. Introductory Chapter: Melatonin, the Integrative Molecule within the Human Architecture. Melatonin - Molecular Biology, Clinical and Pharmaceutical Approaches. IntechOpen. 2018. DOI: 10.5772/intechopen.81071
  28. 28. Garcia JJ et al. Protective effects of melatonin in reducing oxidative stress and in preserving the fluidity of biological membranes: A review. Journal of Pineal Research. 2014;56(3):225-237
  29. 29. Chen Y et al. Melatonin protects hepatocytes against bile acid-induced mitochondrial oxidative stress via the AMPK-SIRT3-SOD2 pathway. Free Radical Research. 2015;49(10):1275-1284
  30. 30. Drăgoi CM et al. In vitro effects of some bio-indoles on the transmembrane potential of Jurkat E6. 1 limphoblasts. Farmácia. 2012;60(2):240-248
  31. 31. Carrillo-Vico A et al. Melatonin: Buffering the immune system. International Journal of Molecular Sciences. 2013;14(4):8638-8683
  32. 32. Carrillo-Vico A et al. A review of the multiple actions of melatonin on the immune system. Endocrine. 2005;27(2):189-200
  33. 33. Nicolae AC et al. In vitro P-GP expression after administration of CNS active drugs. Farmácia. 2016;64(6):844-850
  34. 34. Dragoi CM et al. Characteristics of glucose homeostasis and lipidic profile in a hamster metabolic syndrome model, after the co-administration of melatonin and Irbesartan in a multiparticulate pharmaceutical formation. In: 2nd International Conference on Interdisciplinary Management of Diabetes Mellitus and its Complications, Interdiab 2016. Romania: Bucharest; 2016
  35. 35. Yanar K, Simsek B, Cakatay U. Integration of melatonin related redox homeostasis, aging, and circadian rhythm. Rejuvenation Research. 2019;22(5):409-419
  36. 36. Reiter RJ. Pineal melatonin: Cell biology of its synthesis and of its physiological interactions. Endocrine Reviews. 1991;12(2):151-180
  37. 37. Salminen A, Kaarniranta K, Kauppinen A. Crosstalk between oxidative stress and SIRT1: Impact on the aging process. International Journal of Molecular Sciences. 2013;14(2):3834-3859
  38. 38. Dragoi CM, Andreea LA, Cristina ED-P, Ion BD, Daniela EP, George TA, et al. Melatonin: A Silent Regulator of the Glucose Homeostasis. Carbohydrate. InTech. 2017. DOI: 10.5772/66625
  39. 39. Drăgoi CM et al. 1 cell line studies regarding the effects of some bio-indoles on the membrane fluidity. Farmácia. 2012;60(1):13-20
  40. 40. Nicolae AC et al. Clinical implications of the indolergic system and oxidative stress in physiological gestational homeostasis. Farmácia. 2015;63(1):46-51
  41. 41. Nicolae AC et al. Chronotherapy Advances in the Management of Chronic Neurological and Cardiovascular Diseases: Complex Interactions of Circadian Rhythm Environmental Inputs, Nutrition and Drug Administration and their Impact on Human Health, in Circadian Rhythm-New Insights into Physiological and Pathological Implications. London: IntechOpen; 2022
  42. 42. Stanciu AE et al. Clinical significance of serum melatonin in predicting the severity of oral squamous cell carcinoma. Oncology Letters. 2020;19(2):1537-1543
  43. 43. Soni SK et al. Sirtuins and the circadian clock interplay in cardioprotection: Focus on sirtuin 1. Cellular and Molecular Life Sciences. 2021;78(6):2503-2515
  44. 44. Jung-Hynes B, Reiter RJ, Ahmad N. Sirtuins, melatonin and circadian rhythms: Building a bridge between aging and cancer. Journal of Pineal Research. 2010;48(1):9-19
  45. 45. Song YJ, Zhong CB, Wu W. Cardioprotective effects of melatonin: Focusing on its roles against diabetic cardiomyopathy. Biomedicine & Pharmacotherapy. 2020;128:110260
  46. 46. Ramis MR et al. Caloric restriction, resveratrol and melatonin: Role of SIRT1 and implications for aging and related-diseases. Mechanisms of Ageing and Development. 2015;146-148:28-41
  47. 47. Costa EJ, Lopes RH, Lamy- Freund MT. Permeability of pure lipid bilayers to melatonin. Journal of Pineal Research. 1995;19(3):123-126
  48. 48. Karbownik M et al. Renal toxicity of the carcinogen delta-aminolevulinic acid: Antioxidant effects of melatonin. Cancer Letters. 2000;161(1):1-7
  49. 49. Drăgoi C et al. DNA targeting AS a molecular mechanism underlying endogenous indoles biological effects. Farmácia. 2019;67(2)
  50. 50. Acuna-Castroviejo D et al. Characterization of high-affinity melatonin binding sites in purified cell nuclei of rat liver. Journal of Pineal Research. 1994;16(2):100-112
  51. 51. Shah SA et al. Melatonin stimulates the SIRT1/Nrf2 Signaling pathway counteracting lipopolysaccharide (LPS)-induced oxidative stress to rescue postnatal rat brain. CNS Neuroscience & Therapeutics. 2017;23(1):33-44
  52. 52. Ding YW et al. SIRT1 exerts protective effects against paraquat-induced injury in mouse type II alveolar epithelial cells by deacetylating NRF2 in vitro. International Journal of Molecular Medicine. 2016;37(4):1049-1058
  53. 53. Gureev AP, Popov VN, Starkov AA. Crosstalk between the mTOR and Nrf2/ARE signaling pathways as a target in the improvement of long-term potentiation. Experimental Neurology. 2020;328:113285
  54. 54. Ghareghomi S et al. Nrf2 modulation in breast cancer. Biomedicine. 2022;10(10)
  55. 55. Ahmed SM et al. Nrf2 signaling pathway: Pivotal roles in inflammation. Biochimica et Biophysica Acta - Molecular Basis of Disease. 2017;1863(2):585-597
  56. 56. Fischer TW et al. Melatonin as a major skin protectant: From free radical scavenging to DNA damage repair. Experimental Dermatology. 2008;17(9):713-730
  57. 57. Yu L et al. Melatonin ameliorates myocardial ischemia/reperfusion injury in type 1 diabetic rats by preserving mitochondrial function: Role of AMPK-PGC-1alpha-SIRT3 signaling. Scientific Reports. 2017;7:41337
  58. 58. Wu S, Zou MH. AMPK, mitochondrial function, and cardiovascular disease. International Journal of Molecular Sciences. 2020;21(14)
  59. 59. Wang S, Song P, Zou MH. AMP-activated protein kinase, stress responses and cardiovascular diseases. Clinical Science (London, England). 2012;122(12):555-573
  60. 60. Aggarwal R et al. Novel therapeutic approaches enhance PGC1-alpha to reduce oxidant stress-inflammatory Signaling and improve functional recovery in hibernating myocardium. Antioxidants (Basel). 2022;11(11)
  61. 61. Butterick TA et al. Pioglitazone increases PGC1-alpha signaling within chronically ischemic myocardium. Basic Research in Cardiology. 2016;111(3):37
  62. 62. Mposhi A et al. Regulation of mitochondrial gene expression, the epigenetic enigma. Frontiers in Bioscience (Landmark edition). 2017;22(7):1099-1113
  63. 63. Kang I, Chu CT, Kaufman BA. The mitochondrial transcription factor TFAM in neurodegeneration: Emerging evidence and mechanisms. FEBS Letters. 2018;592(5):793-811
  64. 64. Zhai M et al. Melatonin ameliorates myocardial ischemia reperfusion injury through SIRT3-dependent regulation of oxidative stress and apoptosis. Journal of Pineal Research. 2017;63(2)
  65. 65. Liu Y et al. Melatonin: A potential adjuvant therapy for septic myopathy. Biomedicine & Pharmacotherapy. 2023;158:114209
  66. 66. Shalini S et al. Old, new and emerging functions of caspases. Cell Death and Differentiation. 2015;22(4):526-539
  67. 67. Asadi M et al. Caspase-3: Structure, function, and biotechnological aspects. Biotechnology and Applied Biochemistry. 2022;69(4):1633-1645
  68. 68. Lim HD et al. Cytoprotective and anti-inflammatory effects of melatonin in hydrogen peroxide-stimulated CHON-001 human chondrocyte cell line and rabbit model of osteoarthritis via the SIRT1 pathway. Journal of Pineal Research. 2012;53(3):225-237
  69. 69. Hardeland R. Aging, melatonin, and the pro- and anti-inflammatory networks. International Journal of Molecular Sciences. 2019;20(5)
  70. 70. Vaure C, Liu Y. A comparative review of toll-like receptor 4 expression and functionality in different animal species. Frontiers in Immunology. 2014;5:316
  71. 71. Li Y, Jiang Q , Wang L. Appetite regulation of TLR4-induced inflammatory signaling. Frontiers in Endocrinology. 2021;12:777997
  72. 72. Sharma BR, Kanneganti TD. NLRP3 inflammasome in cancer and metabolic diseases. Nature Immunology. 2021;22(5):550-559
  73. 73. Chattree V et al. A comprehensive review on modulation of SIRT1 signaling pathways in the immune system of COVID-19 patients by phytotherapeutic melatonin and epigallocatechin-3-gallate. Journal of Food Biochemistry. 2022;46(12):e14259
  74. 74. Martin Gimenez VM et al. New proposal involving nanoformulated melatonin targeted to the mitochondria as a potential COVID-19 treatment. Nanomedicine (London, England). 2020;15(29):2819-2821
  75. 75. Niu Z, Li R. Clinical study of novel coronavirus pneumonia prevention by melatonin. Reproductive Biomedicine Online. 2020;41(6):1156
  76. 76. Ozturk G, Akbulut KG, Guney S. Melatonin, aging, and COVID-19: Could melatonin be beneficial for COVID-19 treatment in the elderly? Turkish Journal of Medical Sciences. 2020;50(6):1504-1512
  77. 77. Rodella LF et al. Aging and vascular dysfunction: Beneficial melatonin effects. Age (Dordrecht, Netherlands). 2013;35(1):103-115
  78. 78. Montaigne D et al. Daytime variation of perioperative myocardial injury in cardiac surgery and its prevention by Rev-Erbalpha antagonism: A single-Centre propensity-matched cohort study and a randomised study. Lancet. 2018;391(10115):59-69
  79. 79. Kuehn BM. The Heart’s circadian rhythms point to potential treatment strategies. Circulation. 2016;134(23):1907-1908
  80. 80. Martino TA et al. The primary benefits of angiotensin-converting enzyme inhibition on cardiac remodeling occur during sleep time in murine pressure overload hypertrophy. Journal of the American College of Cardiology. 2011;57(20):2020-2028
  81. 81. Zairi I et al. Effect of intermittent fasting and chronotherapy on blood pressure control in hypertensive patients during Ramadan. Arterial Hypertension. 2022;26(2):67-72
  82. 82. Yu L et al. Reduced silent information regulator 1 signaling exacerbates myocardial ischemia-reperfusion injury in type 2 diabetic rats and the protective effect of melatonin. Journal of Pineal Research. 2015;59(3):376-390
  83. 83. Zhao L et al. Melatonin alleviates brain injury in mice subjected to cecal ligation and puncture via attenuating inflammation, apoptosis, and oxidative stress: The role of SIRT1 signaling. Journal of Pineal Research. 2015;59(2):230-239
  84. 84. Zschucke E, Gaudlitz K, Strohle A. Exercise and physical activity in mental disorders: Clinical and experimental evidence. Journal of Preventive Medicine and Public Health. 2013;46(Suppl. 1):S12-S21
  85. 85. Mihai DP et al. Effects of venlafaxine, risperidone and Febuxostat on Cuprizone-induced demyelination, Behavioral deficits and oxidative stress. International Journal of Molecular Sciences. 2021;22(13)
  86. 86. Bogie JFJ et al. Fatty acid metabolism in the progression and resolution of CNS disorders. Advanced Drug Delivery Reviews. 2020;159:198-213
  87. 87. Nassan M, Videnovic A. Circadian rhythms in neurodegenerative disorders. Nature Reviews. Neurology. 2022;18(1):7-24
  88. 88. Haidar M et al. Lipophagy: A new player in CNS disorders. Trends in Endocrinology and Metabolism. 2021;32(11):941-951
  89. 89. Hardeland R. Melatonin and microglia. International Journal of Molecular Sciences. 2021;22(15)
  90. 90. Mayo JC et al. Melatonin and sirtuins: A “not-so unexpected” relationship. Journal of Pineal Research. 2017;62(2)
  91. 91. Arioz BI et al. Melatonin attenuates LPS-induced acute depressive-like Behaviors and microglial NLRP3 Inflammasome activation through the SIRT1/Nrf2 pathway. Frontiers in Immunology. 2019;10:1511
  92. 92. Hanslik KL, Ulland TK. The role of microglia and the Nlrp3 Inflammasome in Alzheimer’s disease. Frontiers in Neurology. 2020;11:570711
  93. 93. Minutoli L et al. ROS-mediated NLRP3 Inflammasome activation in brain, heart, kidney, and testis ischemia/reperfusion injury. Oxidative Medicine and Cellular Longevity. 2016;2016:2183026
  94. 94. Liu HD et al. Expression of the NLRP3 inflammasome in cerebral cortex after traumatic brain injury in a rat model. Neurochemical Research. 2013;38(10):2072-2083
  95. 95. Li L, Liu Y. Aging-related gene signature regulated by Nlrp3 predicts glioma progression. American Journal of Cancer Research. 2015;5(1):442-449
  96. 96. Lei Y et al. SIRT1 in forebrain excitatory neurons produces sexually dimorphic effects on depression-related behaviors and modulates neuronal excitability and synaptic transmission in the medial prefrontal cortex. Molecular Psychiatry. 2020;25(5):1094-1111
  97. 97. Abe-Higuchi N et al. Hippocampal sirtuin 1 signaling mediates depression-like behavior. Biological Psychiatry. 2016;80(11):815-826
  98. 98. Fan J et al. SIRT1 mediates Apelin-13 in ameliorating chronic normobaric hypoxia-induced anxiety-like behavior by suppressing NF-kappaB pathway in mice hippocampus. Neuroscience. 2018;381:22-34
  99. 99. Wu JT, Kral JG. The NF-kappaB/IkappaB signaling system: A molecular target in breast cancer therapy. The Journal of Surgical Research. 2005;123(1):158-169
  100. 100. Mitchell S, Vargas J, Hoffmann A. Signaling via the NFkappaB system. Wiley Interdisciplinary Reviews. Systems Biology and Medicine. 2016;8(3):227-241
  101. 101. Dolcet X et al. NF-kB in development and progression of human cancer. Virchows Archiv. 2005;446(5):475-482
  102. 102. Rajkhowa B et al. Activation of SIRT-1 signalling in the prevention of bipolar disorder and related Neurocomplications: Target activators and influences on neurological dysfunctions. Neurotoxicity Research. 2022;40(2):670-686
  103. 103. Lu G et al. Role and possible mechanisms of Sirt1 in depression. Oxidative Medicine and Cellular Longevity. 2018;2018:8596903
  104. 104. Won E, Na KS, Kim YK. Associations between melatonin, neuroinflammation, and brain alterations in depression. International Journal of Molecular Sciences. 2021;23(1)
  105. 105. Tonon AC et al. Melatonin and depression: A translational perspective from animal models to clinical studies. Frontiers in Psychiatry. 2021;12:638981
  106. 106. Erdogan ME et al. The effects of lipoic acid on redox status in brain regions and systemic circulation in streptozotocin-induced sporadic Alzheimer’s disease model. Metabolic Brain Disease. 2017;32(4):1017-1031
  107. 107. Hung CW et al. Ageing and neurodegenerative diseases. Ageing Research Reviews. 2010;9(Suppl. 1):S36-S46
  108. 108. Zhou L et al. Degeneration and energy shortage in the suprachiasmatic nucleus underlies the circadian rhythm disturbance in ApoE(−/−) mice: Implications for Alzheimer’s disease. Scientific Reports. 2016;6:36335
  109. 109. Cuesta S et al. Melatonin can improve insulin resistance and aging-induced pancreas alterations in senescence-accelerated prone male mice (SAMP8). Age (Dordrecht, Netherlands). 2013;35(3):659-671
  110. 110. Cristofol R et al. Neurons from senescence-accelerated SAMP8 mice are protected against frailty by the sirtuin 1 promoting agents melatonin and resveratrol. Journal of Pineal Research. 2012;52(3):271-281
  111. 111. Luo F et al. Melatonin and autophagy in aging-related neurodegenerative diseases. International Journal of Molecular Sciences. 2020;21(19)
  112. 112. Chen D, Zhang T, Lee TH. Cellular mechanisms of melatonin: Insight from neurodegenerative diseases. Biomolecules. 2020;10(8)
  113. 113. Wade AG et al. Add-on prolonged-release melatonin for cognitive function and sleep in mild to moderate Alzheimer’s disease: A 6-month, randomized, placebo-controlled, multicenter trial. Clinical Interventions in Aging. 2014;9:947-961
  114. 114. Singer C et al. A multicenter, placebo-controlled trial of melatonin for sleep disturbance in Alzheimer’s disease. Sleep. 2003;26(7):893-901
  115. 115. Riemersma-van der Lek RF et al. Effect of bright light and melatonin on cognitive and noncognitive function in elderly residents of group care facilities: A randomized controlled trial. JAMA. 2008;299(22):2642-2655
  116. 116. Dowling GA et al. Melatonin and bright-light treatment for rest-activity disruption in institutionalized patients with Alzheimer’s disease. Journal of the American Geriatrics Society. 2008;56(2):239-246
  117. 117. Asayama K et al. Double blind study of melatonin effects on the sleep-wake rhythm, cognitive and non-cognitive functions in Alzheimer type dementia. Journal of Nippon Medical School. 2003;70(4):334-341
  118. 118. Blum B et al. Melatonin in traumatic brain injury and cognition. Cureus. 2021;13(9):e17776
  119. 119. Bell A et al. Traumatic brain injury, sleep, and melatonin-intrinsic changes with therapeutic potential. Clocks Sleep. 2023;5(2):177-203
  120. 120. Zou P et al. Resveratrol pretreatment attenuates traumatic brain injury in rats by suppressing NLRP3 inflammasome activation via SIRT1. Molecular Medicine Reports. 2018;17(2):3212-3217
  121. 121. Zhang YM et al. XingNaoJing injection ameliorates cerebral ischaemia/reperfusion injury via SIRT1-mediated inflammatory response inhibition. Pharmaceutical Biology. 2020;58(1):16-24
  122. 122. Qu XY et al. XingNaoJing injections protect against cerebral ischemia/reperfusion injury and alleviate blood-brain barrier disruption in rats, through an underlying mechanism of NLRP3 inflammasomes suppression. Chinese Journal of Natural Medicines. 2019;17(7):498-505
  123. 123. Xu X et al. Therapeutic effect of berberine on metabolic diseases: Both pharmacological data and clinical evidence. Biomedicine & Pharmacotherapy. 2021;133:110984
  124. 124. Ungurianu A et al. Interleukins and redox impairment in type 2 diabetes mellitus: Mini-review and pilot study. Current Medical Research and Opinion. 2022;38(4):511-522
  125. 125. Lu C et al. Novel role of the SIRT1 in endocrine and metabolic diseases. International Journal of Biological Sciences. 2023;19(2):484-501
  126. 126. Gradinaru D et al. Insulin-leptin axis, cardiometabolic risk and oxidative stress in elderly with metabolic syndrome. Experimental and Clinical Endocrinology & Diabetes. 2018
  127. 127. Jenwitheesuk A et al. Melatonin regulates aging and neurodegeneration through energy metabolism, epigenetics, autophagy and circadian rhythm pathways. International Journal of Molecular Sciences. 2014;15(9):16848-16884
  128. 128. Gutierrez-Cuesta J et al. Evaluation of potential pro-survival pathways regulated by melatonin in a murine senescence model. Journal of Pineal Research. 2008;45(4):497-505
  129. 129. Ostrowska Z et al. Influence of pinealectomy and long-term melatonin administration on GH-IGF-I axis function in male rats. Neuro Endocrinology Letters. 2001;22(4):255-262
  130. 130. Tajes M et al. Anti-aging properties of melatonin in an in vitro murine senescence model: Involvement of the sirtuin 1 pathway. Journal of Pineal Research. 2009;47(3):228-237
  131. 131. Vriend J, Sheppard MS, Borer KT. Melatonin increases serum growth hormone and insulin-like growth factor I (IGF-I) levels in male Syrian hamsters via hypothalamic neurotransmitters. Growth, Development, and Aging. 1990;54(4):165-171
  132. 132. Wilking M et al. Circadian rhythm connections to oxidative stress: Implications for human health. Antioxidants & Redox Signaling. 2013;19(2):192-208
  133. 133. Cai Y et al. Expression of clock genes Per1 and Bmal1 in total leukocytes in health and Parkinson’s disease. European Journal of Neurology. 2010;17(4):550-554
  134. 134. Ding H et al. Decreased expression of Bmal2 in patients with Parkinson’s disease. Neuroscience Letters. 2011;499(3):186-188
  135. 135. Slats D et al. Reciprocal interactions between sleep, circadian rhythms and Alzheimer’s disease: Focus on the role of hypocretin and melatonin. Ageing Research Reviews. 2013;12(1):188-200
  136. 136. Almoosawi S et al. Chronotype: Implications for epidemiologic studies on chrono-nutrition and cardiometabolic health. Advances in Nutrition. 2019;10(1):30-42
  137. 137. Cardinali DP, Hardeland R. Inflammaging, metabolic syndrome and melatonin: A call for treatment studies. Neuroendocrinology. 2017;104(4):382-397
  138. 138. Purdel C, Ungurianu A, Margina D. Metabolic and metabolomic insights regarding the Omega-3 PUFAs intake in type 1 diabetes mellitus. Frontiers in Molecular Biosciences. 2021;8:783065
  139. 139. Srinivasan V et al. Metabolic syndrome, its pathophysiology and the role of melatonin. Recent Patents on Endocrine Metabolic & Immune Drug Discovery. 2013;7(1):11-25
  140. 140. Puchalski SS, Green JN, Rasmussen DD. Melatonin effect on rat body weight regulation in response to high-fat diet at middle age. Endocrine. 2003;21(2):163-167
  141. 141. Wolden-Hanson T et al. Daily melatonin administration to middle-aged male rats suppresses body weight, intraabdominal adiposity, and plasma leptin and insulin independent of food intake and total body fat. Endocrinology. 2000;141(2):487-497
  142. 142. Zanquetta MM et al. Calorie restriction reduces pinealectomy-induced insulin resistance by improving GLUT4 gene expression and its translocation to the plasma membrane. Journal of Pineal Research. 2003;35(3):141-148
  143. 143. Nogueira TC et al. Absence of melatonin induces night-time hepatic insulin resistance and increased gluconeogenesis due to stimulation of nocturnal unfolded protein response. Endocrinology. 2011;152(4):1253-1263
  144. 144. She M et al. NEU-P11, a novel melatonin agonist, inhibits weight gain and improves insulin sensitivity in high-fat/high-sucrose-fed rats. Pharmacological Research. 2009;59(4):248-253
  145. 145. Karamitri A, Jockers R. Melatonin in type 2 diabetes mellitus and obesity. Nature Reviews. Endocrinology. 2019;15(2):105-125
  146. 146. Sartori C et al. Melatonin improves glucose homeostasis and endothelial vascular function in high-fat diet-fed insulin-resistant mice. Endocrinology. 2009;150(12):5311-5317
  147. 147. Tchio C et al. Removal of melatonin receptor type 1 signalling induces dyslipidaemia and hormonal changes in mice subjected to environmental circadian disruption. Endocrinology, Diabetes & Metabolism. 2021;4(1):e00171
  148. 148. Jung KH et al. Melatonin downregulates nuclear erythroid 2-related factor 2 and nuclear factor-kappaB during prevention of oxidative liver injury in a dimethylnitrosamine model. Journal of Pineal Research. 2009;47(2):173-183
  149. 149. Jung KH et al. Melatonin ameliorates cerulein-induced pancreatitis by the modulation of nuclear erythroid 2-related factor 2 and nuclear factor-kappaB in rats. Journal of Pineal Research. 2010;48(3):239-250
  150. 150. Peschke E. Melatonin, endocrine pancreas and diabetes. Journal of Pineal Research. 2008;44(1):26-40
  151. 151. Peschke E et al. Melatonin and type 2 diabetes - a possible link? Journal of Pineal Research. 2007;42(4):350-358
  152. 152. Huber M et al. Genetics of melatonin receptor type 2 is associated with left ventricular function in hypertensive patients treated according to guidelines. European Journal of Internal Medicine. 2013;24(7):650-655
  153. 153. Prokopenko I et al. Variants in MTNR1B influence fasting glucose levels. Nature Genetics. 2009;41(1):77-81
  154. 154. Zheng C et al. A common variant in the MTNR1b gene is associated with increased risk of impaired fasting glucose (IFG) in youth with obesity. Obesity (Silver Spring). 2015;23(5):1022-1029
  155. 155. Gubin DG et al. Daily melatonin administration attenuates age-dependent disturbances of cardiovascular rhythms. Current Aging Science. 2016;9(1):5-13
  156. 156. Tamura H et al. Melatonin treatment in peri- and postmenopausal women elevates serum high-density lipoprotein cholesterol levels without influencing total cholesterol levels. Journal of Pineal Research. 2008;45(1):101-105
  157. 157. Gouspillou G et al. Protective role of parkin in skeletal muscle contractile and mitochondrial function. The Journal of Physiology. 2018;596(13):2565-2579
  158. 158. Meynial-Denis D et al. New strategies to fight against sarcopenia at old age. Journal of Aging Research. 2012;2012:676042
  159. 159. Stacchiotti A, Favero G, Rodella LF. Impact of melatonin on skeletal muscle and exercise. Cell. 2020;9(2)
  160. 160. Salucci S et al. Melatonin role in skeletal muscle disorders. European Review for Medical and Pharmacological Sciences. 2021;25(2):1024-1033
  161. 161. Pizzuto M, Pelegrin P. Cardiolipin in immune signaling and cell death. Trends in Cell Biology. 2020;30(11):892-903
  162. 162. Li XX et al. Cardiolipin and its different properties in mitophagy and apoptosis. The Journal of Histochemistry and Cytochemistry. 2015;63(5):301-311
  163. 163. Hibaoui Y et al. Melatonin improves muscle function of the dystrophic mdx5Cv mouse, a model for Duchenne muscular dystrophy. Journal of Pineal Research. 2011;51(2):163-171
  164. 164. McCormick R, Vasilaki A. Age-related changes in skeletal muscle: Changes to life-style as a therapy. Biogerontology. 2018;19(6):519-536
  165. 165. Heydemann A. Skeletal muscle metabolism in Duchenne and Becker muscular dystrophy-implications for therapies. Nutrients. 2018;10(6)
  166. 166. Gomez-Pinilla PJ, Camello PJ, Pozo MJ. Protective effect of melatonin on Ca2+ homeostasis and contractility in acute cholecystitis. Journal of Pineal Research. 2008;44(3):250-260
  167. 167. Yeung HM, Hung MW, Fung ML. Melatonin ameliorates calcium homeostasis in myocardial and ischemia-reperfusion injury in chronically hypoxic rats. Journal of Pineal Research. 2008;45(4):373-382
  168. 168. Caumo W et al. Melatonin is a biomarker of circadian dysregulation and is correlated with major depression and fibromyalgia symptom severity. Journal of Pain Research. 2019;12:545-556
  169. 169. Stratos I et al. Melatonin restores muscle regeneration and enhances muscle function after crush injury in rats. Journal of Pineal Research. 2012;52(1):62-70
  170. 170. Cohen S. Role of calpains in promoting desmin filaments depolymerization and muscle atrophy. Biochimica et Biophysica Acta. 2020;1867(10):118788
  171. 171. Smith IJ, Lecker SH, Hasselgren PO. Calpain activity and muscle wasting in sepsis. American Journal of Physiology. Endocrinology and Metabolism. 2008;295(4):E762-E771
  172. 172. Tamtaji OR et al. Melatonin, a calpain inhibitor in the central nervous system: Current status and future perspectives. Journal of Cellular Physiology. 2019;234(2):1001-1007
  173. 173. van den Beuken-van Everdingen MH et al. Update on prevalence of pain in patients with cancer: Systematic review and meta-analysis. Journal of Pain and Symptom Management. 2016;51(6):1070-1090 e9
  174. 174. Talib WH. Melatonin and cancer hallmarks. Molecules. 2018;23(3)
  175. 175. Reiter RJ et al. Melatonin, a full service anti-cancer agent: Inhibition of initiation, progression and metastasis. International Journal of Molecular Sciences. 2017;18(4)
  176. 176. Reiter RJ et al. Light at night, chronodisruption, melatonin suppression, and cancer risk: A review. Critical Reviews in Oncogenesis. 2007;13(4):303-328
  177. 177. Voiculescu SE et al. Behavioral and molecular effects of prenatal continuous light exposure in the adult rat. Brain Research. 2016;1650:51-59
  178. 178. Blask DE et al. Melatonin-depleted blood from premenopausal women exposed to light at night stimulates growth of human breast cancer xenografts in nude rats. Cancer Research. 2005;65(23):11174-11184
  179. 179. Jung-Hynes B et al. Melatonin, a novel Sirt1 inhibitor, imparts antiproliferative effects against prostate cancer in vitro in culture and in vivo in TRAMP model. Journal of Pineal Research. 2011;50(2):140-149
  180. 180. Cheng Y et al. SIRT1 inhibition by melatonin exerts antitumor activity in human osteosarcoma cells. European Journal of Pharmacology. 2013;715(1-3):219-229
  181. 181. Hill SM et al. Molecular mechanisms of melatonin anticancer effects. Integrative Cancer Therapies. 2009;8(4):337-346
  182. 182. Proietti S et al. Melatonin down-regulates MDM2 gene expression and enhances p53 acetylation in MCF-7 cells. Journal of Pineal Research. 2014;57(1):120-129
  183. 183. Frazzi R. SIRT1 in secretory organ cancer. Frontiers in Endocrinology (Lausanne). 2018;9:569
  184. 184. Sharma A et al. Shedding light on structure, function and regulation of human sirtuins: A comprehensive review. 3 Biotech. 2023;13(1):29
  185. 185. Rodriguez-Santana C et al. Role of melatonin in cancer: Effect on clock genes. International Journal of Molecular Sciences. 2023;24(3)
  186. 186. Kritsilis M et al. Ageing, cellular senescence and neurodegenerative disease. International Journal of Molecular Sciences. 2018;19(10)
  187. 187. Favero G et al. Melatonin: Protection against age-related cardiac pathology. Ageing Research Reviews. 2017;35:336-349
  188. 188. Majidinia M et al. The role of melatonin, a multitasking molecule, in retarding the processes of ageing. Ageing Research Reviews. 2018;47:198-213
  189. 189. Chang HC, Guarente L. SIRT1 mediates central circadian control in the SCN by a mechanism that decays with aging. Cell. 2013;153(7):1448-1460
  190. 190. Franceschi C, Campisi J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences. 2014;69(Suppl. 1):S4-S9
  191. 191. Kireev RA, Vara E, Tresguerres JA. Growth hormone and melatonin prevent age-related alteration in apoptosis processes in the dentate gyrus of male rats. Biogerontology. 2013;14(4):431-442
  192. 192. Keskin-Aktan A et al. The effects of melatonin and curcumin on the expression of SIRT2, Bcl-2 and Bax in the hippocampus of adult rats. Brain Research Bulletin. 2018;137:306-310
  193. 193. Keskin-Aktan A et al. SIRT2 and FOXO3a expressions in the cerebral cortex and hippocampus of young and aged male rats: Antioxidant and anti-apoptotic effects of melatonin. Biologia Futura. 2022;73(1):71-85
  194. 194. Akbulut KG, Aktas SH, Akbulut H. The role of melatonin, sirtuin2 and FoXO1 transcription factor in the aging process of colon in male rats. Biogerontology. 2015;16(1):99-108
  195. 195. Maestroni GJ et al. Does melatonin play a disease-promoting role in rheumatoid arthritis? Journal of Neuroimmunology. 2005;158(1-2):106-111
  196. 196. Ghareghani M et al. Melatonin exacerbates acute experimental autoimmune encephalomyelitis by enhancing the serum levels of lactate: A potential biomarker of multiple sclerosis progression. Clinical and Experimental Pharmacology & Physiology. 2017;44(1):52-61
  197. 197. Cutolo M, Maestroni GJ. The melatonin-cytokine connection in rheumatoid arthritis. Annals of the Rheumatic Diseases. 2005;64(8):1109-1111
  198. 198. Spinedi E, Cardinali DP. Neuroendocrine-metabolic dysfunction and sleep disturbances in neurodegenerative disorders: Focus on Alzheimer’s disease and melatonin. Neuroendocrinology. 2019;108(4):354-364
  199. 199. Rosales-Corral SA et al. Alzheimer’s disease: Pathological mechanisms and the beneficial role of melatonin. Journal of Pineal Research. 2012;52(2):167-202

Written By

Anca Ungurianu, Cristina Manuela Drăgoi, Alina Crenguța Nicolae, Ion-Bogdan Dumitrescu, Daniela Grădinaru and Denisa Margină

Submitted: 27 September 2023 Reviewed: 22 November 2023 Published: 16 March 2024