Next Article in Journal
Attachment Trauma Is Associated with White Matter Fiber Microstructural Alterations in Adolescents with Anorexia Nervosa before and after Exposure to Psychotherapeutic and Nutritional Treatment
Next Article in Special Issue
Parkinson’s Disease: A Narrative Review on Potential Molecular Mechanisms of Sleep Disturbances, REM Behavior Disorder, and Melatonin
Previous Article in Journal
Amalia Revisited—A Reanalysis of Amalia’s Dreams with the Method Structural Dream Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Melatonin as a Chronobiotic/Cytoprotective Agent in REM Sleep Behavior Disorder

by
Daniel P. Cardinali
1,* and
Arturo Garay
2
1
CENECON, Faculty of Medical Sciences, University of Buenos Aires, Buenos Aires C1431FWO, Argentina
2
Unidad de Medicina del Sueño-Sección Neurología, Centro de Educación Médica e Investigaciones Clínicas “Norberto Quirno” (CEMIC), Buenos Aires C1431FWO, Argentina
*
Author to whom correspondence should be addressed.
Brain Sci. 2023, 13(5), 797; https://doi.org/10.3390/brainsci13050797
Submission received: 23 April 2023 / Revised: 8 May 2023 / Accepted: 10 May 2023 / Published: 13 May 2023
(This article belongs to the Special Issue Sleep Disorders in Parkinson’s Disease)

Abstract

:
Dream-enactment behavior that emerges during episodes of rapid eye movement (REM) sleep without muscle atonia is a parasomnia known as REM sleep behavior disorder (RBD). RBD constitutes a prodromal marker of α-synucleinopathies and serves as one of the best biomarkers available to predict diseases such as Parkinson disease, multiple system atrophy and dementia with Lewy bodies. Most patients showing RBD will convert to an α-synucleinopathy about 10 years after diagnosis. The diagnostic advantage of RBD relies on the prolonged prodromal time, its predictive power and the absence of disease-related treatments that could act as confounders. Therefore, patients with RBD are candidates for neuroprotection trials that delay or prevent conversion to a pathology with abnormal α-synuclein metabolism. The administration of melatonin in doses exhibiting a chronobiotic/hypnotic effect (less than 10 mg daily) is commonly used as a first line treatment (together with clonazepam) of RBD. At a higher dose, melatonin may also be an effective cytoprotector to halt α-synucleinopathy progression. However, allometric conversion doses derived from animal studies (in the 100 mg/day range) are rarely employed clinically regardless of the demonstrated absence of toxicity of melatonin in phase 1 pharmacological studies with doses up to 100 mg in normal volunteers. This review discusses the application of melatonin in RBD: (a) as a symptomatic treatment in RBD; (b) as a possible disease-modifying treatment in α-synucleinopathies. To what degree melatonin has therapeutic efficacy in the prevention of α-synucleinopathies awaits further investigation, in particular multicenter double-blind trials.

1. Introduction

Rapid-eye-movement (REM) sleep behavior disorder (RBD) is a REM sleep parasomnia that shows abnormal motor behaviors emerging during REM sleep in a context of diminished normal hypotonia [1]. Various visual disturbances are associated with synucleinopathies [2]. It has been established that isolated RBD (iRBD) is a reliable prodromal manifestation of any α-synucleinopathy within 20 years of onset of iRBD [3]. Melatonin, a chronobiotic methoxyindole with a well-known cytoprotective and antioxidant and free radical scavenging properties, has shown effectiveness for symptomatic treatment of RBD [4]. This is a necessary intervention to avoid the risk of injury to oneself or one’s bed partner and also to avoid the sleep disruption that these episodes entail. In addition, in the absence of specific treatments, patients with iRBD are candidates for neuroprotection trials or even better, disease-modifying treatments, that delay or prevent conversion to a pathology with abnormal α-synuclein metabolism [5]. This review will discuss two different aspects related to the action of melatonin in RBD: 1. As a symptomatic treatment in RBD and 2. As a possible disease-modifying treatment in α-synucleinopathies.

2. Circadian Modulation of REM Sleep

Sleep is regulated by two interacting processes: circadian and homeostatic processes [6]. The circadian process is driven by the body’s internal biological clock. The circadian process promotes wakefulness during the day and sleepiness at night, with a peak in sleepiness occurring during the late night and early morning hours. The second process is the homeostatic process, which is driven by the body’s need for sleep and is based on the concept of sleep debt. The homeostatic process monitors the quality and amount of an individual’s sleep and adjusts the drive for sleep accordingly. The longer an individual is awake, the greater the homeostatic sleep pressure, leading to increased sleepiness and a stronger drive to sleep [6]. Timing and duration of sleep interact. The circadian process regulates the timing of sleep and wakefulness, while the homeostatic process regulates the duration and depth of sleep. Together, these processes help to maintain a balance between sleep and wakefulness, promoting healthy and restorative sleep [6]. Neurodegenerative processes include early disruption of sleep and circadian rhythms, and both types of disturbances are recognized as key factors in neurodegeneration [7,8]. RBD gives a window for insight into the early development of α-synucleinopathies, i.e., Parkinson’s disease (PD), multiple system atrophy and Lewy body dementia.
Circadian oscillations are based on interconnected transcriptional and post-translational feedback loops that are regulated by core clock genes [9]. The transcriptional/translational feedback loops comprising the core clockwork were studied by transgenic gene deletion technology in rats and mice. Delays in the feedback loops, partly depending on phosphorylation of the clock proteins that affects their stability and transcription complex formation, are the basis of the 24 h oscillation in clock gene expression [10]. As the Clock and Bmal1 genes in mammals are transcribed, the transcription factors BMAL1 and CLOCK are produced, which then dimerize via basic helix-loop-helix (bHLH) domains. The dimers subsequently stimulate the transcription of Cry and Per, the proteins CRY and PER inhibiting the expression of BMAL1 and CLOCK. The cycle is restarted as PER and CRY decline over time [10]. Per and Cry mRNA levels in the suprachiasmatic nucleus (SCN) have a maximum in middle/late afternoon [11]. Bmal1 mRNA levels rise around midnight, but Clock mRNA is present in the SCN at a constant concentration all day [12]. Production of PER and CRY is limited by binding to the E-box element of the promoter regions of Bmal1, Clock, Rev-Erb and other clock-controlled genes via the CLOCK/BMAL1 complex [10]. For translocation to the nucleus, PER and CRY are phosphorylated by casein kinase 1 δ/ε [12]. Four secondary regulatory loops modulate master oscillation further. A first one involves ROR (retinoid-related orphan receptor) and the nuclear receptors REV-ERB. REV-ERB suppresses Bmal1, whereas ROR promotes it by attaching to the RORE (response element-binding site) sequence in the Bmal1’s promoter region [13,14]. A second regulatory loop is given by the protein ROR that also binds to the RORE element in the promotor of CLOCK, and to NFIL3 (nuclear factor, interleukin 3 regulated), thus inducing their transcription. NR1D1 (nuclear receptor subfamily 1 group D member 1) and possibly other proteins from this family inhibit ROR binding to the RORE element. A third regulatory loop comprises DBP (D-box binding PAR b ZIP transcription factor), a protein whose expression is controlled by BMAL1:CLOCK from the first loop, which binds to the D-box in the promotor region of PER. NFIL3 from the second loop regulates this binding negatively [10]. A fourth regulatory loop is given by DEC (AKA Basic helix-loop-helix family member e40 loop) as an ancillary circadian loop characterized by the expression of DEC and other circadian-controlled genes under regulation by BMAL1:CLOCK. DEC, in turn, inhibits BMAL1:CLOCK binding to the E-box element, regulating its expression [15].
In normal REM sleep, two neuronal networks, one suppressing motor-skeletal activity and the other generating muscle atonia, are involved [16]. Active inhibition by neurons in the medulla explains the muscle atonia. The thalamus influences spinal motor neurons, and locomotion implicates input from the forebrain. The laterodorsal tegmental nucleus, the pedunculo-pontine nucleus and the peri-locus coeruleus region are brainstem regions involved in RBD pathophysiology [17]. REM atonia is handled by supra-spinal mechanisms. Muscle atonia in REM sleep depends on the stimulation by nuclei in the pons and medulla, which then transmit descending inhibitory projections to hyperpolarize spinal α- motoneurons. Obliteration of this mechanism leads to muscle activity during the REM sleep [18]. At the early stages of α-synucleinopathies, other sleep characteristics besides REM associated atonia remain relatively unaffected.
Under usual entrained conditions, the probability of REM sleep to occur (i.e., REM sleep propensity) is highest during the second half of night sleep [19]. A circadian profile characterizes REM sleep, which is more dependent on the circadian phase than on previous wakefulness duration [20]. Almost every aspect of REM sleep is modulated by the circadian system. However, these parameters are not altered in iRBD. Animal research supports such a circadian modulation in SCN lesioned rats, in which this circadian modulation of REM sleep vanished [21]. Experiments with crepuscular mammals indicate that both diurnal and nocturnal REM sleep deprivation lead to comparable REM sleep debt, but only after nocturnal deprivation does a consistent REM sleep rebound occur, indicating active promotion of REM sleep by the circadian system [22].
The therapeutic effect of melatonin is another indication on the involvement of the circadian system in α-synucleinopathies. Kunz et al. have proposed that a reason for controversial results lies on the specific mode of action of melatonin as a chronobiotic [23,24]. Melatonin is commonly used as a conventional sleeping aid, administered 1–2 h prior to bedtime. However, in several studies, administration of melatonin in a chronobiotic protocol at the moment of expect maximum effect (around 0100 h) adjusted for chronotype, exerts a gradual therapeutic effect on RBD seen within 1 to 2 weeks [23,24]. α-Synucleinopathies are seen as a complex combination of motor and non-motor disturbance preceded by a prodromal, non-motor phase including alterations of sleep, autonomic disorders, sensory deficits and cognitive impairment, among others [8,25]. Since diurnal fluctuations are detected in non-motor symptoms during the course of the disease, it is logical to associate the symptom fluctuations with chronodisruption [26].
An example is given by PD [27]. Both during the course of the disease, as well as after dopaminergic treatments used to mitigate Parkinsonian symptoms, clock genes and melatonin significantly change [28,29]. In addition to be the best prognostic biomarker for the development of α-synucleinopathies, iRBD represents the ongoing neurodegenerative process itself [3,30]. The rhythmicity of circadian clock genes and circulating melatonin levels were measured in iRBD patients [31]. Abolition of circadian rhythmicity of Bmal1, Per2 and Nr1d1, intactness of rhythmicity of Per1, and a reduced amplitude of Per3 rhythm were documented in iRBD patients. No change in the diurnal melatonin rhythm was observed. RBD patients had a more dispersed range of acrophases of melatonin rhythm (>11 h) as compared to the control group exhibiting stable acrophases with approximately 5 h dispersion. A phase delay of about 1 h was seen in iRBD patients as compared to controls [31].
Disturbances of biological rhythms emerge in fully developed α-synucleinopathy. For example, PD patients exhibited lower expression of BMAL1 [32,33,34]. In a sample of 1253 Chinese PD patients vs. 1342 controls, polymorphism in PER1 and ARNTL genes were uncovered reinforcing the link of circadian alterations with α-synucleinopathies. Symptomatic severity of RBD correlated with an altered body temperature rhythm [35]. Disturbances in blood pressure or cortisol circadian rhythms were also observed [36,37]. In the sleep study of the longitudinal cohort Osteoporotic Fractures in Men Study (comprising more than 2900 men), a higher risk of incident α-synucleinopathy was associated with disrupted circadian rhythmicity [38]. Non-REM sleep symptoms remain poorly understood in α-synucleinopathies [39].

3. RBD as a Prodrome of α-Synucleinopathies

As mentioned, α-synucleinopathies are associated with RBD or daytime sonmnolence years in advance to a diagnosis based on motor, behavioral or autonomic disturbances. Sleep deprivation leads to impairment of clearance of waste proteins by the glymphatic system with concomitant neurodegeneration. In a recent study, glymphatic system activity, evaluated by diffusion tensor image analysis along the perivascular space (ALPS), indicated a lower ALPS index (impairment of glymphatic system activity) in PD and iRBD patients as compared to healthy controls. The ALPS index and elevated disease severity were negatively correlated in the iRBD and PD subgroups [40]. The recently described subarachnoid lymphatic-like membrane in murine and human brains provides fundamental insights into brain fluid transport and immune barriers and opens questions about its function in the presentation of neurodegenerative diseases and related sleep disorders [41].
iRBD has proven to be a robust predictor of disease since more than 80% of iRBD patients will develop some α-synucleinopathy with a conversion rate of 6.3% of diagnosed patients by year [41,42]. On clinical grounds, the first observation corresponded to James Parkinson that pointed out the presence of excessive motor activity during sleep resembling subsequent descriptions of RBD in 1817 [43]. He wrote: “In this stage the sleep becomes much disturbed. The tremulous motion of the limbs occurs during sleep, and augments until they awaken the patient, and frequently with much agitation and alarm”. In modern science, the first observation of loss of atonia during REM sleep was that of Jouvet and co-workers [44,45] followed later on by the contributions of Hendricks et al. [17]. The complete description of the phenomenon in humans was made by Schenk and colleagues [46].
Dream-enactment motor behaviors that emerge during episodes of REM sleep define RBD as a parasomnia. Two types of RBD occur, i.e., isolated or symptomatic. In the latter case it can be a comorbidity related to neurological disorders like neurodegenerative diseases, or narcolepsy, to autoimmune or paraneoplastic diseases, or to substance abuse or withdrawal. It is important to highlight that not all of the patients with α-synucleinopathies have RBD. It is found in 50% of PD, 90% of multiple system atrophy and 80% of dementia with Lewy bodies. It is recommended that once patients are identified with isolated RBD it is mandatory to stratified them in conjunction with other markers of disease since the diagnosis of iRBD is not sufficient to distinguish subtypes of α-synucleinopathy [47]. Despite these limitations, iRBD constitutes a robust prodromal marker of α-synucleinopathies and to date, the best biomarker available to predicts α-synucleinopathy diseases [48], with the following “pros”: (1) A prolonged prodromal time, (2) An excellent predictive power and when diagnosed as iRBD, and (3) The absence of disease-related treatments that could act as confounders [42,49].

4. Basic Physiology and Biochemistry of Melatonin Relevant to RBD

Two major functions are ascribed to melatonin and are relevant for the neurodegeneration. Melatonin is a chronobiotic and at a higher dose, an effective cytoprotector. Thus to have a full view of melatonin utility in α-synucleinopathies both aspects should be considered. Melatonin is the prototype of the endogenous signals regulating the circadian system (the so-called “chronobiotics”) [50,51]. The sleep/wake cycle in both normal and blind patients is strongly related to the circadian rhythm of melatonin in blood [52]. The association of plasma melatonin levels with the circadian processes that govern sleep proclivity is clearly established [53,54]. A fundamental information on the time of year regulating neuroendocrine seasonality depends on the secretion of pineal melatonin which is proportional to night duration [55].
Circulating melatonin is produced almost exclusively by the pineal gland. As soon as it is synthesized, melatonin diffuses out into the capillary blood [56] and cerebrospinal fluid (CSF) [57]. Melatonin is found earlier in the third ventricle than in the lateral ventricle CSF. Levels of melatonin in ventricle CSF were up to 30 times higher than circulating levels [57], whereas spinal CSF values were in the range of those found in blood [58]. Hypothalamic melatonin concentrations up to 50 times greater than in plasma were described by high pressure liquid chromatography [59] or radioimmunoassay [60]. This indicates the existence of two compartments of melatonin affecting physiological function, i.e., in CSF affecting neurally mediated functions and in plasma acting on peripheral organs. Circulating melatonin derived from the pineal gland is about 5% of the total melatonin produced in the body. Peak concentration of circulating melatonin occurs at night with maximum at a younger age [61]. Increased levels of oxidative stress and associated degenerative changes seen in α-synucleinopathies are probably related to the age-associated decline in melatonin production [62]. Melatonin is synthesized locally in most cells [63] and the hypothesis that it is produced in all animal cells that have mitochondria [64] and that this mitochondrial function is critical for cytoprotection [65] is widely accepted.
The chronobiotic function of melatonin relies mainly on melatonergic receptors (MT1 and MT2), which belong to the superfamily of membrane receptors associated with G proteins [66]. GPR50 is another member of the melatonin receptor family which rather than binding melatonin it forms homo and heteromers with MT1 and MT2 [67]. The SCN and several other CNS areas, like the cerebral and cerebellar cortex and the midbrain, display MT1 and MT2 receptor activity [68]. Substantia nigra, caudate-putamen, ventral tegmental areas and nucleus accumbens contain melatonin receptors [69]. In the case of substantia nigra MT1 and MT2 receptors, a depressed signal was found in PD patients [70]. Cell membranes are crossed easily by melatonin due to its amphiphilic properties. In the cytoplasm melatonin interacts with proteins like calmodulin and tubulin [71]. In the nucleus, indirect interaction of melatonin with orphan RZR/ROR superfamily of receptors [65] via the activation of sirtuin-1 has been detected [72].
However, interaction with MT1 and MT2 receptors cannot explain fully the cytoprotective effect of the methoxyindole. The melatonin doses needed to modify the intracellular melatonin are significantly higher than those used clinically as a chronobiotic [73]. Although in cell cultures, nanomolar concentrations of melatonin are effective [74], pharmacological doses largely exceeding receptor saturation are needed for cytoprotection [75]. Excellent summaries on the activity of melatonin to reverse altered signaling mechanisms in neurodegeneration have been published [76,77,78]. The antioxidant effects of melatonin on free radical production are largely independent of receptors. In addition to be a free radical scavenger itself, a cascade of compounds with higher antioxidant activity is produced after melatonin oxidation. As an indirect antioxidant, melatonin also promotes the synthesis of antioxidant enzymes while inhibiting that of pro-oxidant enzymes [79]. Under ischemic conditions (not related to free radicals) the stabilizing activity of melatonin on mitochondrial membranes explain its antiapoptotic and cytoprotective effects [64].
The mitochondrial aspects of α-synucleinopathies that arise from the activity of α-synuclein as an excitotoxin must be considered when discussing melatonin activity [80,81]. They include free radical generation and calcium overload-mediated changes in mitochondrial membrane potential and in permeability transition pore (mtPTP). Mitochondria are not only a major site of radical oxygen species (ROS) generation, but also the primary target of attack for ROS and radical nitrogen species (RNS) [82]. A vicious cycle leading to further generation of free radicals arises after damage of the mitochondrial respiratory chain via breakdown of the proton potential and opening of the mtPTP; apoptosis is thus elicited. Indeed, mitochondrial effects explain the neuroprotective role of melatonin in α-synucleinopathies. Besides ROS and RNS scavenging, additional actions of melatonin are stimulation of glutathione (GSH) synthesis, support of the GSH disulfide (GSSG) reduction and protection of mitochondrial membranes and DNA from oxidative insults [83,84]. Some others are indirect antioxidant effects of melatonin, like the maintenance of mitochondrial electron flux [85,86]. In contrast to other antioxidants, the balanced amphiphilicity of melatonin allows crossing of the cell membranes to be concentrated within mitochondrial compartments [86,87,88]. Melatonin administration increased the activities of mitochondrial respiratory complexes I and IV in the brain [89]. Melatonin antagonizes mitochondrial protein misfolding damage given by free radicals and changes in membrane potential due to overexcitation caused by excess calcium.
The brain has a high oxygen consumption rate and is relatively rich in polyunsaturated fatty acids which can be peroxidized under oxidative stress [90]. Lipid peroxidation leading to oxidative protein modifications can be effectively suppressed by melatonin, particularly in the central nervous system [91]. Melatonin is very effective to scavenge free-radicals like the extremely reactive hydroxyl radical, carbonate radicals and RNS [92]. Metabolites of melatonin, like cyclic 3-hydroxymelatonin, N1-acetyl-N2-formyl-5-methoxykynuramine (AFMK) and N1-acetyl-5-methoxykynuramine (AMK) also share the activity of the mother molecule [84,93]. GSH peroxidase was consistently upregulated by melatonin in the brain [84,93]. Down-regulation of prooxidant enzymes such as lipoxygenases and nitric oxide (NO) synthases is also seen after melatonin. Thus, by avoiding peroxynitrite-derived radicals and NO-dependent neuronal excitation, and by inhibiting inflammatory reactions, the oxidative and nitrosative damage is attenuated by melatonin [94]. Moreover, melatonin decreases Pp53, Bax, and caspase 9 expression [95] and increases Bcl-2 and p53 levels [96] leading to the inhibition of the apoptosis pathway. Microglial activation in neuroinflammation plays a causative role in α-synucleinopathies exacerbating the pathological consequences of the disease. Increased inflammatory factors including nuclear factor κB (NF-κB), IL-1, IL-6, cyclooxygenase (Cox)-2, TNF-α, iNOS, and INF-γ in glial cells, and elevated oxidative stress due to excessive free radical generation following mitochondrial damage, play essential roles in the progression of α-synucleinopathies [97,98].
Dopaminergic neuron loss and α-synuclein accumulation is found in 1-tmethyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced murine model of PD. Death of dopaminergic neurons depends on toll-like receptor 4 (TLR4) stimulation via cytokine release evoked by NF-κB in activated glial cells. In the MPTP mouse model melatonin reduces dopaminergic neuron loss, decreases cytokine release due to the inflammatory response and suppresses TLR4-mediated neuroinflammation [99]. Melatonin displays both pro-inflammatory and anti-inflammatory effects [100,101]. The inhibition of COX [102] (mainly Cox-2) [103] and of NF κB binding to DNA, and the down-regulation of inducible NO synthase receptors, underlie melatonin anti-inflammatory activity [65]. Other anti-inflammatory pathways include prevention of NLRP3 inflammasome activation and up-regulation of nuclear factor erythroid 2-related factor 2 [65]. Ultimately, these effects lead to an augmented production of anti-inflammatory cytokines and decreased levels of pro-inflammatory cytokines [100,101].
Brain magnetic resonance spectroscopy in PD patients documented a γ-aminobutyric acid (GABA)-ergic dysregulation in basal ganglia [104]. Dopaminergic neurodegeneration probably led to a GABA hypofunction in basal ganglia because striatal dopaminergic axons release GABA [105]. Administration of the GABA agonist baclofen in a murine model of PD ameliorated motor symptoms and protected dopamine cell bodies [106]. Besides its anti-excitatory and sedative effects [107,108] melatonin may act on the GABAergic system to mediate neuroprotection. Through the activation of GABAergic receptors melatonin protects neurons from ß-amyloid peptide toxicity [109]. The efficacy of the benzodiazepine antagonist flumazenil to modify upregulation of GABA activity by melatonin, and the lack of activity of the melatonin receptor antagonist luzindole, support the occurrence of an allosteric modulation of GABAA receptors by melatonin [110]. Melatonin has a potent anti-excitotoxic activity. Melatonin administration prevents CA1 neuron death in the hippocampus after transient ischemia [111] or due to high doses of glucocorticoids [112]. The neuronal death induced by the agonist of the ionotropic glutamate receptor kainite is also prevented by melatonin [113]. Neither MT1 nor MT2 melatonin receptor blockers are able to modify this anti-excitotoxic activity of melatonin [114].
Of importance in relation to α-synucleinopathies is the activity of melatonin to counteract an increased insulin resistance (IR) [115]. Both brain and systemic IR are documented in PD [116,117] and type-2 diabetes (T2D) is a risk factor aggravating PD development [118]. The pathological signs of PD are Lewy bodies and neurites, composed of amyloid aggregates of misfolded α-synuclein [119,120]. Aggregation of islet amyloid polypeptide (amylin) is found in the pancreas in T2D, the deposit of amylin accelerating the formation of α-synuclein amyloid [119]. direct interaction between amylin and α-synuclein was indicated by studies in pancreatic ß-cells of patients with a neuropathological diagnosis of α-synucleinopathy [121]. In experimental models of PD, melatonin increases the concentration of nigral and striatal dopamine [122]; it also reportedly prevents the depletion of dopamine and disruption of dopaminergic neurons [95] and neurotoxins-induced dopaminergic neuron death [123]. An elevation in the activity of superoxide dismutase, catalase, and GSH peroxidase and reduction in the malondialdehyde level and death of dopaminergic neurons were documented after melatonin treatment in the substantia nigra of rats with PD model induced by 6-hydroxydopamine [124]. Administration of melatonin also reduces oxidative stress in the MPTP murine model of PD [125]. In view of melatonin’s capacity to suppress free radicals, transfer electrons, and repair damaged biomolecules, it may effectively protect neurons and glial cells from the oxidative stress pathway in α-synucleinopathies. In MPTP-induced PD mice, melatonin prevents the rise of iNOS, as a pathologic hallmark of neuroinflammation [126]. The protective effect of melatonin on α-synuclein-induced damage to dopaminergic neurons in the substantia nigra has been observed in animal models [123]. Melatonin prevents α-synuclein assembly and fibril formation by suppressing protofibril development and instability in precursor fibrils.
The expression of aquaporine-4 (AQ4) is significantly reduced in PD patient brains compared to the healthy individuals [127] The AQ4 water channels have an essential role in lowering CSF α-synuclein levels [128]. Melatonin preserves the function of glymphatic system and increases AQ4 expression [129,130]: hence, it has a favorable effect on PD patients. However, further clinical trials are required to definitively prove the beneficial effects of melatonin on PD patients [131] (Figure 1).

5. Melatonin and Melatonin Analogs in RBD

Table 1 summarizes data on the use of melatonin and melatonin analogs to treat RBD. In 1997 the first observation on melatonin as a treatment was published [133]. A 64-year-old man with clinically and polysomnographically confirmed RBD was examined. He was also an insomniac with excessive daily somnolence and short time memory problem and did not receive clonazepam because of comorbidities. After melatonin administration (3 mg p.o./daily at bedtime) the RBD symptoms, cognitive/mnestic deficits and sleep abnormalities showed a complete clinical recovery [133].
The findings were confirmed in the same laboratory in an open-labeled trial with 6 patients (melatonin 3 mg daily) [24] and also in a randomized, double-blind, placebo-controlled trial in a crossover design with 8 patients (placebo or melatonin 3 mg daily, each for a period of 4 weeks [23]. Video-polysomnographic examination (vPSG) was used in all cases to confirm RBD. Twelve out of the 14 patients improved clinically and showed significantly decreased REM sleep without atonia (RWA) in vPSG (Table 1).
In a prospective observational study, the effect of melatonin (3–9 mg daily) was examined in 15 patients with vPSG-confirmed RBD [134]. A mild to strong improvement of symptoms was reported by 13 patients. Melatonin administration significantly reduced percentage of tonic REM activity in vPSG. Baseline melatonin levels were low in the presence of a higher response in RBD patients.
The efficacy of melatonin as monotherapy and in combination with clonazepam was examined in a retrospective observational study [135]. Fourteen RBD patients were treated with melatonin (3–12 mg daily). Coexisting comorbidities were PD, narcolepsy, dementia with Lewy bodies and multiple system atrophy. RBD was controlled by melatonin in 6 patients, significantly improved in 4, and transiently improved in 2. Continued benefit with melatonin beyond 12 months of therapy was seen in 8 subjects [135].
Anderson and Shneerson [136] reviewed 39 patients with confirmed RBD. In 58% of the patients using clonazepam adverse effects were reported. Twenty-one patients continued to take clonazepam, 8 used another medication, and 4 required a combination of medications to control symptoms adequately. Two patients successfully used 10 mg melatonin p.o./daily. A combination therapy (lonazepam/gabapentin/melatonin) was used and found effective [136].
A survey of 45 consecutive RBD patients seen at Mayo Clinic between 2008–2010 was published [137]. The primary outcome variables were RBD visual analog scale (VAS) score and injury frequency. Twenty-five patients received melatonin (6 mg/daily), 18 received clonazepam (0.5 mg daily) and 2 received both as initial treatment. RBD VAS ratings were significantly improved after treatments. Significantly reduced injuries and fewer adverse effects were observed in melatonin-treated patients [137].
Another retrospective analysis evaluated the data from 28 patients with PSG-confirmed RBD [138]. Comorbidities observed were PD (10 patients) and cognitive decline (4 patients). All patients received melatonin (3–6 mg daily). After 4 months treatment with 6 mg melatonin nightly, 26 patients showed a clear clinical improvement. Following the first period of melatonin monotherapy, all patients received a combination therapy with clonazepam 0.5–1 mg per night. A significant reduction in the percent of wakefulness after sleep onset was found in the patients with 6 mg melatonin first and subsequently combined with clonazepam [138,139].
Obstructive sleep apnea (OSA) is known to provoke RBD-like symptoms and both sleep-related pathologies usually coincide because of overlapping prevalence in similar age groups. In a case series study on four patients with a severe clinical RBD syndrome and concomitant OSA, daily administration of 2 mg prolonged release melatonin resulted in improvement of RBD symptoms but persistence of the sleep related breathing disorder [140]. REM sleep without atonia values in PSG with melatonin were abnormally high, probably because of the untreated OSA. indicating the necessity of treatment of both disorders for an optimal therapeutic response.
In a case report study, a 72-year-old man was clinically diagnosed as RBD in 2011 [141]. A reduced DA transporter density typically indicating an impending PD was diagnosed by DA transporter scintigraphy (DaTSCAN). After 6 months of daily melatonin treatment (2 mg prolonged release) the clinical signs of RBD disappeared. A control PSG in 2014 confirmed the presence of a normal REM sleep. Additional DaTSCANs were performed in 2013 and 2015. Compared to the clear PD signs in the 2011 scan, the 2013 scan was borderline and the 2015 scan was without any sign of PD. The authors interpreted the results as a possible neuroprotective role for melatonin in α-synucleinopathy [141].
A single-center, observational cohort study of 209 consecutive iRBD patients treated with melatonin was undertaken to assess treatment effects, time course and confounding factors [4]. A total of 171 patients received 2 mg prolonged release melatonin (2 mg, ≥6 months), 13 were under melatonin for about 1–3 months, and 25 received mixed treatments. RBD symptomatology gradually improved over the first 4 weeks of treatment with melatonin and remained stably improved for up to 21.7 years [4].
It must be noted that negative results with melatonin (prolonged release) have also been reported. In a randomized, double–blind, placebo–controlled pilot study on 30 iRBD to assess the effects of prolonged-release melatonin (2 or 6 mg/day), primary outcomes (scores from the Clinical Global Impression-Improvement and the Korean version of the RBD questionnaire-Hong Kong) and secondary outcomes (Pittsburgh Sleep Quality Index score, the Epworth Sleepiness Scale score, and the frequency of dream–enacting behaviors) remained unchanged after melatonin administration [142].
A dose of 4 mg of prolonged-release melatonin or placebo p.o. once-daily before bedtime was given to 30 PD patients with RBD in a double-blind, placebo-controlled trial [143]. No differences between melatonin-treated and placebo were found. The authors concluded that prolonged-release melatonin 4 mg did not reduce RBD disorder in PD [143]. Presumably, melatonin at low dose is relatively ineffective when RBD is co-morbid with PD or OSA [140].
In a prospective, open-label, randomized trial undertaken to compare efficacy and safety of melatonin and clonazepam, RBD patients received either clonazepam 0.5 mg or prolonged-release melatonin 2 mg at bedtime for 4 weeks [144]. In 34 patients with probable RBD, scoring parameters of RWA improved after clonazepam treatment but not after melatonin treatment. Daytime sleepiness and insomnia symptoms were reduced by melatonin but not by clonazepam. The proportion of N2 sleep was increased, and N3 and REM sleep were decreased only in the clonazepam group. Depressive symptoms increased after clonazepam [144].
Concerning melatonergic agonists only a limited number of studies examined their efficacy in RBD. Three publications examined the effect of ramelteon. In an open-labeled trial, 12 consecutive patients with idiopathic RBD were treated for at least 4 weeks with 8 mg ramelteon 30 min before bedtime [145]. Ramelteon treatment did not have any effect on RBD severity scale or REM sleep without atonia measured by vPSG.
Two patients with secondary RBD complications along with neurodegenerative diseases including multiple system atrophy and PD received ramelteon (8 mg per night) in monotherapy due to contraindications to clonazepam [147]. Ramelteon treatment improved RBD severity scale or REM sleep without atonia measured by vPSG.
In another study 35 patients from multiple centers with idiopathic PD comorbid with sleep disorders were evaluated for ramelteon response [146]. The patients received 8 mg of ramelteon before sleep once daily for 12 weeks. Twenty-four out of the 35 patients examined were diagnosed with probable RBD (PRBD) using the Japanese version of the RBD screening questionnaire. Sleep disruption in PD patients was curtailed by ramelteon [146].
Concerning agomelatine, a case review described its positive effects in 3 patients with clinical and PSG confirmed iRBD [148]. Aggressive behavior fully remitted in 1 patient after treatment with 25 mg per night. The other 2 patients received 50 mg per day agomelatine with clearly reduced numbers of RBD episodes.
A systematic review of randomized controlled trials and observational studies that addressed interventions for the management of RBD was published on behalf of The American Academy of Sleep Medicine [149]. The study concluded that the overall certainty of evidence for melatonin to treat iRBD was low. Clinical improvement in decreasing frequency and/or intensity of dream enactment episodes were noted among patients taking immediate-release melatonin and to a lesser degree prolonged-release melatonin. In a critical review Gilat et al. concluded that the effectiveness of the two first-line therapies for RBD (melatonin and clonazepam) are probably overestimated [150].
The differences in efficacy of fast release and prolonged release melatonin may depend on the concentrations attained at the phase delay and phase advance portions of the night. Although it is widely accepted that natural melatonin’s chronobiotic influence is mediated by MT receptors, a chronobiotic effect can also be observed when pharmaceutical amounts of fast-release melatonin (that saturate receptors) are utilized. Even at such high doses, melatonin ingested as a fast-release preparation at a single time of day (bedtime) maintains chronobiotic effects contrasting with the more or less similar pharmacological concentrations during the whole night attained by prolonged release melatonin [151].

6. Concluding Remarks

Utilizing preclinical models has significant potential to advance the understanding of the interplay of α-synucleinopathies with the circadian system and sleep. In melatonin research, animal models has been widely studied but many of their findings are overlooked. In particular the translation doses calculated by allometry [152] is generally disregarded to discuss melatonin use in humans, regardless than these studies are very useful to calculate the initial doses of compounds use in clinical Phase 1 studies. The effect of melatonin in animal models of α-synucleinopathies indicates that, from the melatonin doses used in each case, a human equivalent dose (HED) of melatonin is in the 100 mg/day range [132].
Dosage escalation trials showed melatonin’s absence of toxicity in humans in doses up to 100 mg [153,154]. Melatonin has a high safety profile and is generally well tolerated (see ref. [75]). In the USA, the number of individuals aged >65 years who have used melatonin in the last month increases 3-fold over the last two decades [155]. Typically few, mild to moderate in intensity, and either self-limiting or resolved promptly after treatment discontinuation, adverse effects of melatonin have been documented [156,157,158].
In the United States an estimated 3.1 million individuals (1.3 percent of adults) consume melatonin daily [159]. The manufacturing quality and bioavailability of melatonin and the potential contaminants are questionable in these unlicensed melatonin preparations [160,161]. In that aspect, commercial melatonin labeled with the U.S. Pharmacopeia (USP) Verification Mark may provide the most consistent dosing among treatment options.
To what degree melatonin has therapeutic efficacy in the prevention or treatment of α-synucleinopathies awaits further investigation, in particular multicenter double-blind trials. Because of the HED of melatonin determined from preclinical studies, melatonin dosages need to be reviewed. Indeed, given the number of scientific/medical papers that have recommended its use, melatonin’s failure to garner attention as a potential treatment for α-synucleinopathies is disappointing. The fact that no significant group has advocated for its therapeutic usage in treating this condition is one of several potential causes for this.
The pharmaceutical business is not motivated to promote the use of melatonin because it is not patented and affordable. Nonetheless, it would be wise for the pharmaceutical business to research the possibility of a profitable and medically effective combination of melatonin with other medications. Due to its low cost, minimal toxicity, and ability to be taken orally, melatonin would be particularly advantageous. This is particularly true in underdeveloped nations where individuals have less money to spend on treating age-related α-synucleinopathies.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

D.P.C. is an Emeritus Superior Investigator from the Argentine National Research Council and Emeritus Professor at the University of Buenos Aires.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. American Academy of Sleep Medicine Darien IL The International Classification of Sleep Disorders; American Academy of Sleep Medicine: Darien, IL, USA, 2015.
  2. Maurage, C.A.; Ruchoux, M.M.; De Vos, R.; Surguchov, A.; Destee, A. Retinal Involvement in Dementia with Lewy Bodies: A Clue to Hallucinations? Ann. Neurol. 2003, 54, 542–547. [Google Scholar] [CrossRef] [PubMed]
  3. Postuma, R.B.; Iranzo, A.; Hu, M.; Högl, B.; Boeve, B.F.; Manni, R.; Oertel, W.H.; Arnulf, I.; Ferini-Strambi, L.; Puligheddu, M.; et al. Risk and Predictors of Dementia and Parkinsonism in Idiopathic REM Sleep Behaviour Disorder: A Multicentre Study. Brain 2019, 142, 744–759. [Google Scholar] [CrossRef] [PubMed]
  4. Kunz, D.; Stotz, S.; Bes, F. Treatment of Isolated REM Sleep Behavior Disorder Using Melatonin as a Chronobiotic. J. Pineal Res. 2021, 71, e12759. [Google Scholar] [CrossRef] [PubMed]
  5. Nassan, M.; Videnovic, A. Circadian Rhythms in Neurodegenerative Disorders. Nat. Rev. Neurol. 2022, 18, 7–24. [Google Scholar] [CrossRef]
  6. Borbély, A.A.; Daan, S.; Wirz-Justice, A.; Deboer, T. The Two-Process Model of Sleep Regulation: A Reappraisal. J. Sleep. Res. 2016, 25, 131–143. [Google Scholar] [CrossRef]
  7. Colwell, C.S. Defining Circadian Disruption in Neurodegenerative Disorders. J. Clin. Investig. 2021, 131, e148288. [Google Scholar] [CrossRef]
  8. Shen, Y.; Lv, Q.-K.; Xie, W.-Y.; Gong, S.-Y.; Zhuang, S.; Liu, J.-Y.; Mao, C.-J.; Liu, C.-F. Circadian Disruption and Sleep Disorders in Neurodegeneration. Transl. Neurodegener. 2023, 12, 8. [Google Scholar] [CrossRef]
  9. Welz, P.S.; Benitah, S.A. Molecular Connections Between Circadian Clocks and Aging. J. Mol. Biol. 2020, 432, 3661–3679. [Google Scholar] [CrossRef]
  10. Takahashi, J.S. Transcriptional Architecture of the Mammalian Circadian Clock. Nat. Rev. Genet. 2017, 18, 164–179. [Google Scholar] [CrossRef]
  11. Hastings, M.H.; Smyllie, N.J.; Patton, A.P. Molecular-Genetic Manipulation of the Suprachiasmatic Nucleus Circadian Clock. J. Mol. Biol. 2020, 432, 3639–3660. [Google Scholar] [CrossRef]
  12. Lee, C.; Etchegaray, J.P.; Cagampang, F.R.A.; Loudon, A.S.I.; Reppert, S.M. Posttranslational Mechanisms Regulate the Mammalian Circadian Clock. Cell 2001, 107, 855–867. [Google Scholar] [CrossRef]
  13. Fontaine, C.; Staels, B. The Orphan Nuclear Receptor Rev-Erbα: A Transcriptional Link between Circadian Rhythmicity and Cardiometabolic Disease. Curr. Opin. Lipidol. 2007, 18, 141–146. [Google Scholar] [CrossRef]
  14. Preitner, N.; Brown, S.; Ripperger, J.; Le-Minh, N.; Damiola, F.; Schibler, U. Sehgal Orphan Nuclear Receptors, Molecular Clockwork, and the Entrainment of Peripheral Oscillators. Novartis Found. Symp. 2003, 253, 89–101. [Google Scholar] [CrossRef]
  15. Ono, D.; Honma, K.I.; Schmal, C.; Takumi, T.; Kawamoto, T.; Fujimoto, K.; Kato, Y.; Honma, S. CHRONO and DEC1/DEC2 Compensate for Lack of CRY1/CRY2 in Expression of Coherent Circadian Rhythm but Not in Generation of Circadian Oscillation in the Neonatal Mouse SCN. Sci. Rep. 2021, 11, 19240. [Google Scholar] [CrossRef]
  16. Garay, A.; Cardinali, D.P. New Concepts in the Neurophysiology of Sleep and Wakefulness. Physiol. Minirev. 2016, 9, 26–36. [Google Scholar]
  17. Hendricks, J.C.; Morrison, A.R.; Mann, G.L. Different Behaviors during Paradoxical Sleep without Atonia Depend on Pontine Lesion Site. Brain. Res. 1982, 239, 81–105. [Google Scholar] [CrossRef]
  18. Boeve, B.F.; Silber, M.H.; Saper, C.B.; Ferman, T.J.; Dickson, D.W.; Parisi, J.E.; Benarroch, E.E.; Ahlskog, J.E.; Smith, G.E.; Caselli, R.C.; et al. Pathophysiology of REM Sleep Behaviour Disorder and Relevance to Neurodegenerative Disease. Brain 2007, 130, 2770–2788. [Google Scholar] [CrossRef]
  19. Czeisler, C.A.; Weitzman, E.D.; Moore-Ede, M.C.; Zimmerman, J.C.; Knauer, R.S. Human Sleep: Its Duration and Organization Depend on Its Circadian Phase. Science 1980, 210, 1264–1267. [Google Scholar] [CrossRef]
  20. Endo, S.; Kobayashi, T.; Yamamoto, T.; Fukuda, H.; Sasaki, M.; Ohta, T. Persistence of the Circadian Rhythm of REM Sleep: A Variety of Experimental Manipulations of the Sleep-Wake Cycle. Sleep 1981, 4, 319–328. [Google Scholar] [CrossRef]
  21. Wurts, S.W.; Edgar, D.M. Circadian and Homeostatic Control of Rapid Eye Movement (REM) Sleep: Promotion of REM Tendency by the Suprachiasmatic Nucleus. J. Neurosci. 2000, 20, 4300–4310. [Google Scholar] [CrossRef]
  22. Ocampo-Garcés, A.; Hernández, F.; Palacios, A.G. REM Sleep Phase Preference in the Crepuscular Octodon Degus Assessed by Selective REM Sleep Deprivation. Sleep 2013, 36, 1247–1256. [Google Scholar] [CrossRef] [PubMed]
  23. Kunz, D.; Mahlberg, R. A Two-Part, Double-Blind, Placebo-Controlled Trial of Exogenous Melatonin in REM Sleep Behaviour Disorder. J. Sleep Res. 2010, 19, 591–596. [Google Scholar] [CrossRef] [PubMed]
  24. Kunz, D.; Bes, F. Melatonin as a Therapy in REM Sleep Behavior Disorder Patients: An Open-Labeled Pilot Study on the Possible Influence of Melatonin on REM-Sleep Regulation. Mov. Disord. 1999, 14, 507–511. [Google Scholar] [CrossRef] [PubMed]
  25. Titova, N.; Chaudhuri, K.R. Non-Motor Parkinson Disease: New Concepts and Personalised Management. Med. J. Aust. 2018, 208, 404–409. [Google Scholar] [CrossRef] [PubMed]
  26. Kunz, D.; Oster, H.; Rawashdeh, O.; Neumann, W.; Münte, T.; Berg, D. Sleep and Circadian Rhythms in α-Synucleinopathies-Perspectives for Disease Modification. Acta Physiol. 2023, 238, e13966. [Google Scholar] [CrossRef]
  27. Asadpoordezaki, Z.; Coogan, A.N.; Henley, B.M. Chronobiology of Parkinson’s Disease: Past, Present and Future. Eur. J. Neurosci. 2023, 57, 178–200. [Google Scholar] [CrossRef]
  28. Liu, Y.; Niu, L.; Liu, X.; Cheng, C.; Le, W. Recent Progress in Non-Motor Features of Parkinson’s Disease with a Focus on Circadian Rhythm Dysregulation. Neurosci. Bull. 2021, 37, 1010–1024. [Google Scholar] [CrossRef]
  29. Schenck, C.H.; Boeve, B.F.; Mahowald, M.W. Delayed Emergence of a Parkinsonian Disorder or Dementia in 81% of Older Men Initially Diagnosed with Idiopathic Rapid Eye Movement Sleep Behavior Disorder: A 16-Year Update on a Previously Reported Series. Sleep. Med. 2013, 14, 744–748. [Google Scholar] [CrossRef]
  30. Weissová, K.; Škrabalová, J.; Skálová, K.; Červená, K.; Bendová, Z.; Miletínová, E.; Kopřivová, J.; Šonka, K.; Dudysová, D.; Bartoš, A.; et al. Circadian Rhythms of Melatonin and Peripheral Clock Gene Expression in Idiopathic REM Sleep Behavior Disorder. Sleep Med. 2018, 52, 1–6. [Google Scholar] [CrossRef]
  31. Cai, Y.; Liu, S.; Sothern, R.B.; Xu, S.; Chan, P. Expression of Clock Genes Per1 and Bmal1 in Total Leukocytes in Health and Parkinson’s Disease. Eur. J. Neurol. 2010, 17, 550–554. [Google Scholar] [CrossRef]
  32. Ding, H.; Liu, S.; Yuan, Y.; Lin, Q.; Chan, P.; Cai, Y. Decreased Expression of Bmal2 in Patients with Parkinson’s Disease. Neurosci. Lett. 2011, 499, 186–188. [Google Scholar] [CrossRef]
  33. Breen, D.P.; Vuono, R.; Nawarathna, U.; Fisher, K.; Shneerson, J.M.; Reddy, A.B.; Barker, R.A. Sleep and Circadian Rhythm Regulation in Early Parkinson Disease. JAMA Neurol. 2014, 71, 589–595. [Google Scholar] [CrossRef]
  34. Zhong, G.; Bolitho, S.; Grunstein, R.; Naismith, S.L.; Lewis, S.J.G. The Relationship between Thermoregulation and REM Sleep Behaviour Disorder in Parkinson’s Disease. PLoS ONE 2013, 8, e72661. [Google Scholar] [CrossRef]
  35. Berganzo, K.; Díez-Arrola, B.; Tijero, B.; Somme, J.; Lezcano, E.; Llorens, V.; Ugarriza, I.; Ciordia, R.; Gómez-Esteban, J.C.; Zarranz, J.J. Nocturnal Hypertension and Dysautonomia in Patients with Parkinson’s Disease: Are They Related? J. Neurol. 2013, 260, 1752–1756. [Google Scholar] [CrossRef]
  36. Ahsan Ejaz, A.; Sekhon, I.S.; Munjal, S. Characteristic Findings on 24-h Ambulatory Blood Pressure Monitoring in a Series of Patients with Parkinson’s Disease. Eur. J. Intern. Med. 2006, 17, 417–420. [Google Scholar] [CrossRef]
  37. Leng, Y.; Blackwell, T.; Cawthon, P.M.; Ancoli-Israel, S.; Stone, K.L.; Yaffe, K. Association of Circadian Abnormalities in Older Adults with an Increased Risk of Developing Parkinson Disease. JAMA Neurol. 2020, 77, 1270–1278. [Google Scholar] [CrossRef]
  38. Bugalho, P.; Magriço, M. Sleep Stability in Isolated Rapid Eye Movement Sleep Behavior Disorder, Parkinson’s Disease, and Dementia with Lewy Bodies. Acta. Neurol. Scand. 2022, 146, 545–552. [Google Scholar] [CrossRef]
  39. Si, X.; Guo, T.; Wang, Z.; Fang, Y.; Gu, L.; Cao, L.; Yang, W.; Gao, T.; Song, Z.; Tian, J.; et al. Neuroimaging Evidence of Glymphatic System Dysfunction in Possible REM Sleep Behavior Disorder and Parkinson’s Disease. NPJ. Park. Dis. 2022, 8, 54. [Google Scholar] [CrossRef]
  40. Møllgård, K.; Beinlich, F.R.M.; Kusk, P.; Miyakoshi, L.M.; Delle, C.; Plá, V.; Hauglund, N.L.; Esmail, T.; Rasmussen, M.K.; Gomolka, R.S.; et al. A Mesothelium Divides the Subarachnoid Space into Functional Compartments. Science 2023, 379, 84–88. [Google Scholar] [CrossRef]
  41. Postuma, R.B.; Gagnon, J.F.; Vendette, M.; Montplaisir, J.Y. Markers of Neurodegeneration in Idiopathic Rapid Eye Movement Sleep Behaviour Disorder and Parkinson’s Disease. Brain 2009, 132, 3298–3307. [Google Scholar] [CrossRef]
  42. Postuma, R.B. Neuroprotective Trials in REM Sleep Behavior Disorder: The Way Forward Becomes Clearer. Neurology 2022, 99, 19–25. [Google Scholar] [CrossRef] [PubMed]
  43. Parkinson, J. An Essay on the Shaking Palsy. 1817. J. Neuropsychiatry Clin. Neurosci. 2002, 14, 223–236. [Google Scholar] [CrossRef] [PubMed]
  44. Jouvet, M. Paradoxical Sleep—A Study of Its Nature and Mechanisms. Prog. Brain. Res. 1965, 18, 20–62. [Google Scholar] [CrossRef] [PubMed]
  45. Sastre, P.J.P.; Jouvet, M. Oneiric Behavior in Cats. Physiol. Behav. 1979, 22, 979–989. [Google Scholar] [CrossRef] [PubMed]
  46. Schenck, C.H.; Bundlie, S.R.; Ettinger, M.G.; Mahowald, M.W. Chronic Behavioral Disorders of Human REM Sleep: A New Category of Parasomnia. Sleep 1986, 9, 293–308. [Google Scholar] [CrossRef]
  47. Dauvilliers, Y. Identifying the Best Biomarkers for α-Synucleinopathies. Lancet Neurol. 2021, 20, 593–594. [Google Scholar] [CrossRef]
  48. Dauvilliers, Y.; Schenck, C.H.; Postuma, R.B.; Iranzo, A.; Luppi, P.H.; Plazzi, G.; Montplaisir, J.; Boeve, B. REM Sleep Behaviour Disorder. Nat. Rev. Dis. Primers. 2018, 4, 19. [Google Scholar] [CrossRef]
  49. Videnovic, A.; Ju, Y.E.S.; Arnulf, I.; Cochen-De Cock, V.; Cochen-De Cock, V.; Högl, B.; Kunz, D.; Provini, F.; Provini, F.; Ratti, P.L.; et al. Clinical Trials in REM Sleep Behavioural Disorder: Challenges and Opportunities. J. Neurol. Neurosurg. Psychiatry 2020, 91, 740–749. [Google Scholar] [CrossRef]
  50. Dawson, D.; Armstrong, S.M. Chronobiotics-Drugs That Shift Rhythms. Pharmacol. Ther. 1996, 69, 15–36. [Google Scholar] [CrossRef]
  51. Skene, D.J.; Arendt, J. Circadian Rhythm Sleep Disorders in the Blind and Their Treatment with Melatonin. Sleep. Med. 2007, 8, 651–655. [Google Scholar] [CrossRef]
  52. Emens, J.S.; Eastman, C.I. Diagnosis and Treatment of Non-24-h Sleep-Wake Disorder in the Blind. Drugs 2017, 77, 637–650. [Google Scholar] [CrossRef]
  53. Gobbi, G.; Comai, S. Sleep Well. Untangling the Role of Melatonin MT1 and MT2 Receptors in Sleep. J. Pineal Res. 2018, 66, e12544. [Google Scholar] [CrossRef]
  54. Auld, F.; Maschauer, E.L.; Morrison, I.; Skene, D.J.; Riha, R.L. Evidence for the Efficacy of Melatonin in the Treatment of Primary Adult Sleep Disorders. Sleep. Med. Rev. 2017, 34, 10–22. [Google Scholar] [CrossRef]
  55. Pandi-Perumal, S.R.; Trakht, I.; Srinivasan, V.; Spence, D.W.; Maestroni, G.J.M.; Zisapel, N.; Cardinali, D.P. Physiological Effects of Melatonin: Role of Melatonin Receptors and Signal Transduction Pathways. Prog. Neurobiol. 2008, 85, 335–353. [Google Scholar] [CrossRef]
  56. Arendt, J. Melatonin: Countering Chaotic Time Cues. Front. Endocrinol. 2019, 10, 391. [Google Scholar] [CrossRef]
  57. Tricoire, H.; Møller, M.; Chemineau, P.; Malpaux, B. Origin of Cerebrospinal Fluid Melatonin and Possible Function in the Integration of Photoperiod. Reprod. Suppl. 2003, 61, 311–321. [Google Scholar] [CrossRef]
  58. Reiter, R.J.; Tan, D.X. Role of CSF in the Transport of Melatonin. J. Pineal Res. 2002, 33, 61. [Google Scholar] [CrossRef]
  59. Cardinali, D.P.; Rosenstein, R.; Golombek, D.; Chuluyan, E.; Kanterewicz, B.; Del Zar, M.; Vacas, M. Melatonin Binding Sites in Brain: Single or Multiple? Adv. Pineal Res. 1991, 5, 159–165. [Google Scholar]
  60. Pang, S.F.; Brown, G.M. Regional Concentrations of Melatonin in the Rat Brain in the Light and Dark Period. Life. Sci. 1983, 33, 1199–1204. [Google Scholar] [CrossRef]
  61. Godfrey, S.; Iversen, H.K.; West, A.S. Melatonin Profile in Healthy, Elderly Subjects-A Systematic Literature Review. Chronobiol. Int. 2022, 39, 476–492. [Google Scholar] [CrossRef]
  62. Tchekalarova, J.; Tzoneva, R. Oxidative Stress and Aging as Risk Factors for Alzheimer’s Disease and Parkinson’s Disease: The Role of the Antioxidant Melatonin. Int. J. Mol. Sci. 2023, 24, 3022. [Google Scholar] [CrossRef] [PubMed]
  63. 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]
  64. Tan, D.-X.; Reiter, R.J. Mitochondria: The Birth Place, Battle Ground and the Site of Melatonin Metabolism in Cells. Melatonin Res. 2019, 2, 44–66. [Google Scholar] [CrossRef]
  65. Hardeland, R.; Cardinali, D.P.; Srinivasan, V.; Spence, D.W.; Brown, G.M.; Pandi-Perumal, S.R. Melatonin-A Pleiotropic, Orchestrating Regulator Molecule. Prog. Neurobiol. 2011, 93, 350–384. [Google Scholar] [CrossRef] [PubMed]
  66. Dubocovich, M.L.; Delagrange, P.; Krause, D.N.; Sugden, D.; Cardinali, D.P.; Olcese, J. International Union of Basic and Clinical Pharmacology. LXXV. Nomenclature, Classification, and Pharmacology of G Protein-Coupled Melatonin Receptors. Pharmacol. Rev. 2010, 62, 343–380. [Google Scholar] [CrossRef]
  67. Cecon, E.; Oishi, A.; Jockers, R. Melatonin Receptors: Molecular Pharmacology and Signalling in the Context of System Bias. Br. J. Pharmacol. 2018, 175, 3263–3280. [Google Scholar] [CrossRef]
  68. Ng, K.Y.; Leong, M.K.; Liang, H.; Paxinos, G. Melatonin Receptors: Distribution in Mammalian Brain and Their Respective Putative Functions. Brain Struct. Funct. 2017, 222, 2921–2939. [Google Scholar] [CrossRef]
  69. Uz, T.; Arslan, A.D.; Kurtuncu, M.; Imbesi, M.; Akhisaroglu, M.; Dwivedi, Y.; Pandey, G.N.; Manev, H. The Regional and Cellular Expression Profile of the Melatonin Receptor MT1 in the Central Dopaminergic System. Mol. Brain Res. 2005, 136, 45–53. [Google Scholar] [CrossRef]
  70. Adi, N.; Mash, D.C.; Ali, Y.; Singer, C.; Shehadeh, L.; Papapetropoulos, S. Melatonin MT1 and MT2 Receptor Expression in Parkinson’s Disease. Med. Sci. Monat. 2010, 16, BR61-7. [Google Scholar]
  71. Jiménez-Rubio, G.; Ortíz-López, L.; Benítez-King, G. Melatonin Modulates Cytoskeletal Organization in the Rat Brain Hippocampus. Neurosci. Lett. 2012, 511, 47–51. [Google Scholar] [CrossRef]
  72. Hardeland, R. Recent Findings in Melatonin Research and Their Relevance to the CNS. Cent. Nerv. Syst. Agents Med. Chem. 2018, 18, 102–114. [Google Scholar] [CrossRef]
  73. Venegas, C.; García, J.A.; Doerrier, C.; Volt, H.; Escames, G.; Lõpez, L.C.; Reiter, R.J.; Acuña-Castroviejo, D. Analysis of the Daily Changes of Melatonin Receptors in the Rat Liver. J. Pineal Res. 2013, 54, 313–321. [Google Scholar] [CrossRef]
  74. Zemková, H.; Vaněček, J. Inhibitory Effect of Melatonin on Gonadotropin-Releasing Hormone-Induced Ca2+ Oscillations in Pituitary Cells of Newborn Rats. Neuroendocrinology 1997, 65, 276–283. [Google Scholar] [CrossRef]
  75. Cardinali, D.P. Are Melatonin Doses Employed Clinically Adequate for Melatonin-Induced Cytoprotection? Melatonin Res. 2019, 2, 106–132. [Google Scholar] [CrossRef]
  76. Shukla, M.; Chinchalongporn, V.; Govitrapong, P.; Reiter, R.J. The Role of Melatonin in Targeting Cell Signaling Pathways in Neurodegeneration. Ann. N. Y. Acad. Sci. 2019, 1443, 75–96. [Google Scholar] [CrossRef]
  77. Tan, H.Y.; Ng, K.Y.; Koh, R.Y.; Chye, S.M. Pharmacological Effects of Melatonin as Neuroprotectant in Rodent Model: A Review on the Current Biological Evidence. Cell. Mol. Neurobiol. 2020, 40, 25–51. [Google Scholar] [CrossRef]
  78. Tamtaji, O.R.; Reiter, R.J.; Alipoor, R.; Dadgostar, E.; Kouchaki, E.; Asemi, Z. Melatonin and Parkinson Disease: Current Status and Future Perspectives for Molecular Mechanisms. Cell. Mol. Neurobiol. 2020, 40, 15–23. [Google Scholar] [CrossRef]
  79. Reiter, R.J.; Rosales-Corral, S.; Tan, D.X.; Jou, M.J.; Galano, A.; Xu, B. Melatonin as a Mitochondria-Targeted Antioxidant: One of Evolution’s Best Ideas. Cell. Mol. Life. Sci. 2017, 74, 3863–3881. [Google Scholar] [CrossRef]
  80. Zhang, J.; Shi, Y. In Search of the Holy Grail: Toward a Unified Hypothesis on Mitochondrial Dysfunction in Age-Related Diseases. Cells 2022, 11, 1906. [Google Scholar] [CrossRef]
  81. Bazzani, V.; Equisoain Redin, M.; McHale, J.; Perrone, L.; Vascotto, C. Mitochondrial DNA Repair in Neurodegenerative Diseases and Ageing. Int. J. Mol. Sci. 2022, 23, 11391. [Google Scholar] [CrossRef]
  82. Raha, S.; Robinson, B.H. Mitochondria, Oxygen Free Radicals, Disease and Ageing. Trends. Biochem. Sci. 2000, 25, 502–508. [Google Scholar] [CrossRef] [PubMed]
  83. Hardeland, R.; Pandi-Perumal, S.R. Melatonin, a Potent Agent in Antioxidative Defense: Actions as a Natural Food Constituent, Gastrointestinal Factor, Drug and Prodrug. Nutr. Metab. 2005, 2, 22. [Google Scholar] [CrossRef] [PubMed]
  84. Hardeland, R. Antioxidative Protection by Melatonin: Multiplicity of Mechanisms from Radical Detoxification to Radical Avoidance. Endocrine 2005, 27, 119–130. [Google Scholar] [CrossRef] [PubMed]
  85. Castroviejo, D.; Escames, G.; Carazo, A.; Leon, J.; Khaldy, H.; Reiter, R. Melatonin, Mitochondrial Homeostasis and Mitochondrial-Related Diseases. Curr. Top. Med. Chem. 2002, 2, 133–151. [Google Scholar] [CrossRef] [PubMed]
  86. Verma, A.K.; Singh, S.; Rizvi, S.I. Therapeutic Potential of Melatonin and Its Derivatives in Aging and Neurodegenerative Diseases. Biogerontology 2023, 24, 183–206. [Google Scholar] [CrossRef]
  87. Morén, C.; de Souza, R.M.; Giraldo, D.M.; Uff, C. Antioxidant Therapeutic Strategies in Neurodegenerative Diseases. Int. J. Mol. Sci. 2022, 23, 9328. [Google Scholar] [CrossRef]
  88. Chen, D.; Zhang, T.; Lee, T.H. Cellular Mechanisms of Melatonin: Insight from Neurodegenerative Diseases. Biomolecules 2020, 10, 1158. [Google Scholar] [CrossRef]
  89. Reiter, R.J.; Tan, D.X.; Rosales-Corral, S.; Galano, A.; Jou, M.J.; Acuna-Castroviejo, D. Melatonin Mitigates Mitochondrial Meltdown: Interactions with SIRT3. Int. J. Mol. Sci. 2018, 19, 2439. [Google Scholar] [CrossRef]
  90. Angelova, P.R.; Abramov, A.Y. Alpha-Synuclein and Beta-Amyloid-Different Targets, Same Players: Calcium, Free Radicals and Mitochondria in the Mechanism of Neurodegeneration. Biochem. Biophys. Res. Commun. 2017, 483, 1110–1115. [Google Scholar] [CrossRef]
  91. Ikram, M.; Park, H.Y.; Ali, T.; Kim, M.O. Melatonin as a Potential Regulator of Oxidative Stress, and Neuroinflammation: Mechanisms and Implications for the Management of Brain Injury-Induced Neurodegeneration. J. Inflamm. Res. 2021, 14, 6251–6264. [Google Scholar] [CrossRef]
  92. Manchester, L.C.; Coto-Montes, A.; Boga, J.A.; Andersen, L.P.H.; Zhou, Z.; Galano, A.; Vriend, J.; Tan, D.X.; Reiter, R.J. Melatonin: An Ancient Molecule That Makes Oxygen Metabolically Tolerable. J. Pineal. Res. 2015, 59, 403–419. [Google Scholar] [CrossRef]
  93. Hardeland, R. Melatonin: Signaling Mechanisms of a Pleiotropic Agent. BioFactors 2009, 35, 183–192. [Google Scholar] [CrossRef]
  94. Reiter, R.J.; Ma, Q.; Sharma, R. Melatonin in Mitochondria: Mitigating Clear and Present Dangers. Physiology 2020, 35, 86–95. [Google Scholar] [CrossRef]
  95. Singhal, N.K.; Srivastava, G.; Patel, D.K.; Jain, S.K.; Singh, M.P. Melatonin or Silymarin Reduces Maneb- and Paraquat-Induced Parkinsons Disease Phenotype in the Mouse. J. Pineal Res. 2011, 50, 97–109. [Google Scholar] [CrossRef]
  96. Yildirim, F.B.; Ozsoy, O.; Tanriover, G.; Kaya, Y.; Ogut, E.; Gemici, B.; Dilmac, S.; Ozkan, A.; Agar, A.; Aslan, M. Mechanism of the Beneficial Effect of Melatonin in Experimental Parkinson’s Disease. Neurochem. Int. 2014, 79, 1–11. [Google Scholar] [CrossRef]
  97. 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]
  98. Michel, P.P.; Hirsch, E.C.; Hunot, S. Understanding Dopaminergic Cell Death Pathways in Parkinson Disease. Neuron 2016, 90, 675–691. [Google Scholar] [CrossRef]
  99. Yildirim, S.; Ozkan, A.; Aytac, G.; Agar, A.; Tanriover, G. Role of Melatonin in TLR4-Mediated Inflammatory Pathway in the MTPT-Induced Mouse Model. Neurotoxicology 2022, 88, 168–177. [Google Scholar] [CrossRef]
  100. Hardeland, R. Melatonin and Inflammation—Story of a Double-Edged Blade. J. Pineal Res. 2018, 65, e12525. [Google Scholar] [CrossRef]
  101. Carrillo-Vico, A.; Lardone, P.J.; Álvarez-Śnchez, N.; Rodrĩguez-Rodrĩguez, A.; Guerrero, J.M. Melatonin: Buffering the Immune System. Int. J. Mol. Sci. 2013, 14, 8638–8683. [Google Scholar] [CrossRef]
  102. Cardinali, D.P.; Ritta, M.N.; Fuentes, A.M.; Gimeno, M.F.; Gimeno, A.L. Prostaglandin E Release by Rat Medial Basal Hypothalamus in Vitro. Inhibition by Melatonin at Submicromolar Concentrations. Eur. J. Pharmacol. 1980, 67, 151–153. [Google Scholar] [CrossRef] [PubMed]
  103. 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] [PubMed]
  104. Huang, L.; Ren, Y.; Zeng, Z.; Ren, H.; Li, S.; He, S.; He, F.; Li, X. Comparative Study of Striatum GABA Concentrations and Magnetic Resonance Spectroscopic Imaging in Parkinson’s Disease Monkeys. BMC. Neurosci. 2019, 20, 42. [Google Scholar] [CrossRef] [PubMed]
  105. O’Gorman Tuura, R.L.; Baumann, C.R.; Baumann-Vogel, H. Beyond Dopamine: GABA, Glutamate, and the Axial Symptoms of Parkinson Disease. Front. Neurol. 2018, 9, 806. [Google Scholar] [CrossRef] [PubMed]
  106. Lozovaya, N.; Eftekhari, S.; Cloarec, R.; Gouty-Colomer, L.A.; Dufour, A.; Riffault, B.; Billon-Grand, M.; Pons-Bennaceur, A.; Oumar, N.; Burnashev, N.; et al. GABAergic Inhibition in Dual-Transmission Cholinergic and GABAergic Striatal Interneurons Is Abolished in Parkinson Disease. Nat. Commun. 2018, 9, 1422. [Google Scholar] [CrossRef]
  107. Golombek, D.A.; PéVet, P.; Cardinali, D.P. Melatonin Effects on Behavior: Possible Mediation by the Central GABAergic System. Neurosci. Biobehav. Rev. 1996, 20, 403–412. [Google Scholar] [CrossRef]
  108. Caumo, W.; Levandovski, R.; Hidalgo, M.P.L. Preoperative Anxiolytic Effect of Melatonin and Clonidine on Postoperative Pain and Morphine Consumption in Patients Undergoing Abdominal Hysterectomy: A Double-Blind, Randomized, Placebo-Controlled Study. J. Pain 2009, 10, 100–108. [Google Scholar] [CrossRef]
  109. Louzada, P.R.; Lima, A.C.P.; Mendonca-Silva, D.L.; Noël, F.; De Mello, F.G.; Ferreira, S.T. Taurine Prevents the Neurotoxicity of Β-amyloid and Glutamate Receptor Agonists: Activation of GABA Receptors and Possible Implications for Alzheimer’s Disease and Other Neurological Disorders. FASEB J. 2004, 18, 511–518. [Google Scholar] [CrossRef]
  110. Cheng, X.P.; Sun, H.; Ye, Z.Y.; Zhou, J.N. Melatonin Modulates the GABAergic Response in Cultured Rat Hippocampal Neurons. J. Pharmacol. Sci. 2012, 119, 177–185. [Google Scholar] [CrossRef]
  111. Cho, S.; Joh, T.H.; Baik, H.H.; Dibinis, C.; Volpe, B.T. Melatonin Administration Protects CA1 Hippocampal Neurons after Transient Forebrain Ischemia in Rats. Brain. Res. 1997, 755, 335–338. [Google Scholar] [CrossRef]
  112. Furio, A.M.; Fontao, R.; Falco, N.; Ruiz, J.I.; Caccuri, R.; Cardinali, D.P. Neuroprotective Effect of Melatonin on Glucocorticoid Toxicity in the Rat Hippocampus. Open. Physiol. J. 2014, 1, 23–27. [Google Scholar] [CrossRef]
  113. 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]
  114. Escames, G.; León, J.; López, L.C.; Acuña-Castroviejo, D. Mechanisms of N-Methyl-D-Aspartate Receptor Inhibition by Melatonin in the Rat Striatum. J. Neuroendocrinol. 2004, 16, 929–935. [Google Scholar] [CrossRef]
  115. Do Amaral, F.G.; Andrade-Silva, J.; Kuwabara, W.M.T.; Cipolla-Neto, J. New Insights into the Function of Melatonin and Its Role in Metabolic Disturbances. Expert. Rev. Endocrinol. Metab. 2019, 14, 293–300. [Google Scholar] [CrossRef]
  116. Aviles-Olmos, I.; Limousin, P.; Lees, A.; Foltynie, T. Parkinson’s Disease, Insulin Resistance and Novel Agents of Neuroprotection. Brain 2013, 136, 374–384. [Google Scholar] [CrossRef]
  117. Athauda, D.; Foltynie, T. Insulin Resistance and Parkinson’s Disease: A New Target for Disease Modification? Prog. Neurobiol. 2016, 145–146, 98–120. [Google Scholar] [CrossRef]
  118. De Pablo-Fernandez, E.; Goldacre, R.; Pakpoor, J.; Noyce, A.J.; Warner, T.T. Association between Diabetes and Subsequent Parkinson Disease: A Record-Linkage Cohort Study. Neurology 2018, 91, e139–e142. [Google Scholar] [CrossRef]
  119. Horvath, I.; Wittung-Stafshede, P. Cross-Talk between Amyloidogenic Proteins in Type-2 Diabetes and Parkinson’s Disease. Proc. Natl. Acad. Sci. USA 2016, 113, 12473–12477. [Google Scholar] [CrossRef]
  120. Surguchov, A.; Surguchev, A. Synucleins: New Data on Misfolding, Aggregation and Role in Diseases. Biomedicines 2022, 10, 3241. [Google Scholar] [CrossRef]
  121. Martinez-Valbuena, I.; Amat-Villegas, I.; Valenti-Azcarate, R.; del Mar Carmona-Abellan, M.; Marcilla, I.; Tuñon, M.-T.; Luquin, M.-R. Interaction of Amyloidogenic Proteins in Pancreatic β Cells from Subjects with Synucleinopathies. Acta. Neuropathol. 2018, 135, 877–886. [Google Scholar] [CrossRef]
  122. Patki, G.; Lau, Y.S. Melatonin Protects against Neurobehavioral and Mitochondrial Deficits in a Chronic Mouse Model of Parkinson’s Disease. Pharmacol. Biochem. Behav. 2011, 99, 704–711. [Google Scholar] [CrossRef] [PubMed]
  123. 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] [PubMed]
  124. Ozsoy, O.; Yildirim, F.B.; Ogut, E.; Kaya, Y.; Tanriover, G.; Parlak, H.; Agar, A.; Aslan, M. Melatonin Is Protective against 6-Hydroxydopamine-Induced Oxidative Stress in a Hemiparkinsonian Rat Model. Free. Radic. Res. 2015, 49, 1004–1014. [Google Scholar] [CrossRef] [PubMed]
  125. Ortiz, G.G.; Pacheco-Moisés, F.P.; Gómez-Rodríguez, V.M.; González-Renovato, E.D.; Torres-Sánchez, E.D.; Ramírez-Anguiano, A.C. Fish Oil, Melatonin and Vitamin E Attenuates Midbrain Cyclooxygenase-2 Activity and Oxidative Stress after the Administration of 1-Methyl-4-Phenyl-1,2,3,6- Tetrahydropyridine. Metab. Brain Dis. 2013, 28, 705–709. [Google Scholar] [CrossRef]
  126. López, A.; Ortiz, F.; Doerrier, C.; Venegas, C.; Fernández-Ortiz, M.; Aranda, P.; Díaz-Casado, M.E.; Fernández-Gil, B.; Barriocanal-Casado, E.; Escames, G.; et al. Mitochondrial Impairment and Melatonin Protection in Parkinsonian Mice Do Not Depend of Inducible or Neuronal Nitric Oxide Synthases. PLoS ONE 2017, 12, e0183090. [Google Scholar] [CrossRef]
  127. Hoshi, A.; Tsunoda, A.; Tada, M.; Nishizawa, M.; Ugawa, Y.; Kakita, A. Expression of Aquaporin 1 and Aquaporin 4 in the Temporal Neocortex of Patients with Parkinson’s Disease. Brain Pathol. 2017, 27, 160–168. [Google Scholar] [CrossRef]
  128. Schirinzi, T.; Sancesario, G.M.; Di Lazzaro, G.; Biticchi, B.; Colona, V.L.; Mercuri, N.B.; Bernardini, S.; Pisani, A. CSF α-Synuclein Inversely Correlates with Non-Motor Symptoms in a Cohort of PD Patients. Parkinsonism Park. Relat. Disord. 2019, 61, 203–206. [Google Scholar] [CrossRef]
  129. Pappolla, M.A.; Matsubara, E.; Vidal, R.; Pacheco-Quinto, J.; Poeggeler, B.; Zagorski, M.; Sambamurti, K. Melatonin Treatment Enhances Aβ Lymphatic Clearance in a Transgenic Mouse Model of Amyloidosis. Curr. Alzheimer. Res. 2018, 15, 637–642. [Google Scholar] [CrossRef]
  130. Li, Y.; Zhang, J.; Wan, J.; Liu, A.; Sun, J. Melatonin Regulates Aβ Production/Clearance Balance and Aβ Neurotoxicity: A Potential Therapeutic Molecule for Alzheimer’s Disease. Biomed. Pharmacother. 2020, 132, 110887. [Google Scholar] [CrossRef]
  131. Hu, X.; Li, J.; Wang, X.; Liu, H.; Wang, T.; Lin, Z.; Xiong, N. Neuroprotective Effect of Melatonin on Sleep Disorders Associated with Parkinson’s Disease. Antioxidants 2023, 12, 396. [Google Scholar] [CrossRef]
  132. Pérez-Lloret, S.; Cardinali, D.P. Melatonin as a Chronobiotic and Cytoprotective Agent in Parkinson’s Disease. Front. Pharmacol. 2021, 12, 650597. [Google Scholar] [CrossRef]
  133. Kunz, D.; Bes, F. Melatonin Effects in a Patient with Severe REM Sleep Behavior Disorder: Case Report and Theoretical Considerations. Neuropsychobiology 1997, 36, 211–214. [Google Scholar] [CrossRef]
  134. Takeuchi, N.; Uchimura, N.; Hashizume, Y.; Mukai, M.; Etoh, Y.; Yamamoto, K.; Kotorii, T.; Ohshima, H.; Ohshima, M.; Maeda, H. Melatonin Therapy for REM Sleep Behavior Disorder. Psychiatry Clin. Neurosci. 2001, 55, 267–269. [Google Scholar] [CrossRef]
  135. Boeve, B.F.; Silber, M.H.; Ferman, T.J. Melatonin for Treatment of REM Sleep Behavior Disorder in Neurologic Disorders: Results in 14 Patients. Sleep Med. 2003, 4, 281–284. [Google Scholar] [CrossRef]
  136. Anderson, K.N.; Shneerson, J.M. Drug Treatment of REM Sleep Behavior Disorder: The Use of Drug Therapies Other than Clonazepam. J. Clin. Sleep. Med. 2009, 5, 235–239. [Google Scholar] [CrossRef]
  137. McCarter, S.J.; Boswell, C.L.; St. Louis, E.K.; Dueffert, L.G.; Slocumb, N.; Boeve, B.F.; Silber, M.H.; Olson, E.J.; Tippmann-Peikert, M. Treatment Outcomes in REM Sleep Behavior Disorder. Sleep. Med. 2013, 14, 237–242. [Google Scholar] [CrossRef]
  138. Lin, C.-M.; Chiu, H.-Y.; Guilleminault, C. Melatonin and REM Behavior Disorder. J. Sleep Disord. Ther. 2013, 2, 1000118. [Google Scholar] [CrossRef]
  139. Fernández-Arcos, A.; Iranzo, A.; Serradell, M.; Gaig, C.; Santamaria, J. The Clinical Phenotype of Idiopathic Rapid Eye Movement Sleep Behavior Disorder at Presentation: A Study in 203 Consecutive Patients. Sleep 2016, 39, 121–132. [Google Scholar] [CrossRef]
  140. Schaefer, C.; Kunz, D.; Bes, F. Melatonin Effects in REM Sleep Behavior Disorder Associated with Obstructive Sleep Apnea Syndrome: A Case Series. Curr. Alzheimer Res. 2017, 14, 1. [Google Scholar] [CrossRef]
  141. Kunz, D.; Bes, F. Twenty Years after: Another Case Report of Melatonin Effects on REM Sleep Behavior Disorder, Using Serial Dopamine Transporter Imaging. Neuropsychobiology 2018, 76, 100–104. [Google Scholar] [CrossRef]
  142. Jun, J.S.; Kim, R.; Byun, J.I.; Kim, T.J.; Lim, J.A.; Sunwoo, J.S.; Lee, S.T.; Jung, K.H.; Park, K.I.; Chu, K.; et al. Prolonged-Release Melatonin in Patients with Idiopathic REM Sleep Behavior Disorder. Ann. Clin. Transl. Neurol. 2019, 6, 716–722. [Google Scholar] [CrossRef] [PubMed]
  143. Gilat, M.; Coeytaux Jackson, A.; Marshall, N.S.; Hammond, D.; Mullins, A.E.; Hall, J.M.; Fang, B.A.M.; Yee, B.J.; Wong, K.K.H.; Grunstein, R.R.; et al. Melatonin for Rapid Eye Movement Sleep Behavior Disorder in Parkinson’s Disease: A Randomised Controlled Trial. Mov. Disord. 2020, 35, 344–349. [Google Scholar] [CrossRef] [PubMed]
  144. Byun, J.I.; Shin, Y.Y.; Seong, Y.A.; Yoon, S.M.; Hwang, K.J.; Jung, Y.J.; Cha, K.S.; Jung, K.Y.; Shin, W.C. Comparative Efficacy of Prolonged-Release Melatonin versus Clonazepam for Isolated Rapid Eye Movement Sleep Behavior Disorder. Sleep Breath 2023, 27, 309–318. [Google Scholar] [CrossRef] [PubMed]
  145. Esaki, Y.; Kitajima, T.; Koike, S.; Fujishiro, H.; Iwata, Y.; Tsuchiya, A.; Hirose, M.; Iwata, N. An Open-Labeled Trial of Ramelteon in Idiopathic Rapid Eye Movement Sleep Behavior Disorder. J. Clin. Sleep Med. 2016, 12, 689–693. [Google Scholar] [CrossRef] [PubMed]
  146. Kashihara, K.; Nomura, T.; Maeda, T.; Tsuboi, Y.; Mishima, T.; Takigawa, H.; Nakashima, K. Beneficial Effects of Ramelteon on Rapid Eye Movement Sleep Behavior Disorder Associated with Parkinson’s Disease-Results of a Multicenter Open Trial. Intern. Med. 2016, 55, 231–236. [Google Scholar] [CrossRef]
  147. Nomura, T.; Kawase, S.; Watanabe, Y.; Nakashima, K. Use of Ramelteon for the Treatment of Secondary REM Sleep Behavior Disorder. Intern. Med. 2013, 52, 2123–2126. [Google Scholar] [CrossRef]
  148. Bonakis, A.; Economou, N.T.; Papageorgiou, S.G.; Vagiakis, E.; Nanas, S.; Paparrigopoulos, T. Agomelatine May Improve REM Sleep Behavior Disorder Symptoms. J. Clin. Psychopharmacol. 2012, 32, 732–734. [Google Scholar] [CrossRef]
  149. Howell, M.; Avidan, A.Y.; Foldvary-Schaefer, N.; Malkani, R.G.; During, E.H.; Roland, J.P.; McCarter, S.J.; Zak, R.S.; Carandang, G.; Kazmi, U.; et al. Management of REM Sleep Behavior Disorder: An American Academy of Sleep Medicine Clinical Practice Guideline. J. Clin. Sleep. Med. 2022, 19, 769–810. [Google Scholar] [CrossRef]
  150. Gilat, M.; Marshall, N.S.; Testelmans, D.; Buyse, B.; Lewis, S.J.G. A Critical Review of the Pharmacological Treatment of REM Sleep Behavior Disorder in Adults: Time for More and Larger Randomized Placebo-Controlled Trials. J. Neurol. 2022, 269, 125–148. [Google Scholar] [CrossRef]
  151. Cardinali, D.P.; Brown, G.M.; Reiter, R.J.; Pandi-Perumal, S.R. Elderly as a High-Risk Group during COVID-19 Pandemic: Effect of Circadian Misalignment, Sleep Dysregulation and Melatonin Administration. Sleep Vigil. 2020, 4, 81–87. [Google Scholar] [CrossRef]
  152. Huang, Q.; Riviere, J.E. The Application of Allometric Scaling Principles to Predict Pharmacokinetic Parameters across Species. Expert. Opin. Drug. Metab. Toxicol. 2014, 10, 1241–1253. [Google Scholar] [CrossRef]
  153. Galley, H.F.; Lowes, D.A.; Allen, L.; Cameron, G.; Aucott, L.S.; Webster, N.R. Melatonin as a Potential Therapy for Sepsis: A Phase i Dose Escalation Study and an Ex Vivo Whole Blood Model under Conditions of Sepsis. J. Pineal Res. 2014, 56, 427–438. [Google Scholar] [CrossRef]
  154. Andersen, L.P.H.; Gögenur, I.; Rosenberg, J.; Reiter, R.J. The Safety of Melatonin in Humans. Clin. Drug. Investig. 2016, 36, 169–175. [Google Scholar] [CrossRef]
  155. Li, J.; Somers, V.K.; Xu, H.; Lopez-Jimenez, F.; Covassin, N. Trends in Use of Melatonin Supplements Among US Adults, 1999–2018. JAMA 2022, 327, 483–485. [Google Scholar] [CrossRef]
  156. Besag, F.M.C.; Vasey, M.J.; Lao, K.S.J.; Wong, I.C.K. Adverse Events Associated with Melatonin for the Treatment of Primary or Secondary Sleep Disorders: A Systematic Review. CNS Drugs 2019, 33, 1167–1186. [Google Scholar] [CrossRef]
  157. Menczel Schrire, Z.; Phillips, C.L.; Chapman, J.L.; Duffy, S.L.; Wong, G.; D’Rozario, A.L.; Comas, M.; Raisin, I.; Saini, B.; Gordon, C.J.; et al. Safety of Higher Doses of Melatonin in Adults: A Systematic Review and Meta-Analysis. J. Pineal. Res. 2022, 72, e12782. [Google Scholar] [CrossRef]
  158. Li, C.; Ma, D.; Li, M.; Wei, T.; Zhao, X.; Heng, Y.; Ma, D.; Anto, E.O.; Zhang, Y.; Niu, M.; et al. The Therapeutic Effect of Exogenous Melatonin on Depressive Symptoms: A Systematic Review and Meta-Analysis. Front. Psychiatry. 2022, 13, 737972. [Google Scholar] [CrossRef]
  159. Clarke, T.C.; Black, L.I.; Stussman, B.J.; Barnes, P.M.; Nahin, R.L. Trends in the Use of Complementary Health Approaches Among Adults: United States, 2002–2012. Natl. Health Stat. Rep. 2015, 79, 1–16. [Google Scholar]
  160. Erland, L.A.E.; Saxena, P.K. Melatonin Natural Health Products and Supplements: Presence of Serotonin and Significant Variability of Melatonin Content. J. Clin. Sleep Med. 2017, 13, 275–281. [Google Scholar] [CrossRef]
  161. Goldstein, C.A.; Burgess, H.J. Hit or Miss: The Use of Melatonin Supplements. J. Clin. Sleep Med. 2020, 16, 29S–30S. [Google Scholar] [CrossRef]
Figure 1. Melatonin (M) and synucleinopathies. The Figure depicts the multiple effects of melatonin and the different degree of overlap (interrelations and mutual influences) they have (modified from [132]).
Figure 1. Melatonin (M) and synucleinopathies. The Figure depicts the multiple effects of melatonin and the different degree of overlap (interrelations and mutual influences) they have (modified from [132]).
Brainsci 13 00797 g001
Table 1. Studies including RBD patients treated with melatonin or melatonin receptor agonists.
Table 1. Studies including RBD patients treated with melatonin or melatonin receptor agonists.
Subjects and Concomitant DiseaseDesign and Diagnostic CriteriaStudy’s DurationTreatmentMeasuredNumber of Patients RespondingPSGRef.
64 year-old male iRBD patientCase report5 months3 mg melatonin p.o./daily at bedtimeActigraphy, PSGActigraphy indicated a decrease in motor activity during sleep.↑ % REM-Sleep, ↓ RWA, ↓ phasic electromyographic activity in REM[133]
6 RBD patients, 50% males,
mean age 54 years, 1 PD, 3 with memory and concentration deficits
Open-label prospective case series6 weeks3 mg melatonin p.o./daily at bedtimePSGIn 5 patients, improvement of symptoms was observed during treatment and extended for weeks or months after treatment.↑ REM atonia,
↓ stage shifts in REM epochs
[24]
8 male RBD patients, mean age 54 years, 2 narcolepsia, 1 PDDouble blind, placebo-controlled trial4 weeks3 mg melatonin p.o./daily at bedtime.PSGImproved clinical global impression in 7 patients.↓ frequency of RBD episodes
↓ 30-s epochs of RWA
[23]
14 male RBD patients, mean age 63.5 years,
RBD with no further disclosures
Open-label prospective case seriesvariable3–9 mg melatonin p.o./daily at bedtimePSGNormalized problem sleep behavior in 13 patients after melatonin treatment. Patients with low melatonin levels tended to respond to melatonin therapy better.↓ % tonic REM activity after melatonin administration[134]
13 male RBD, onset age 56 years, 7 DLB, 2 mild cognitive impairment, 2 narcolepsia, 1 PDRetrospective case series14 months3–12 mg melatonin p.o./daily at bedtime, +clonazepam 0.5–1 mg in 7 patientsClinical Global ImpressionAfter 12 months of treatment, 8 patients had improved symptoms of RBD.No[135]
38 Male RBD patients,
mean age 66 years
1 mild cognitive impairment + mild OSA
Retrospective case series All initially treated with clonazepam. When melatonin was used, it was given at a 10 mg p.o./daily at bedtime.Clinical Global ImpressionTo control symptoms, 21 patients received clonazepam, 8 used another medication, and 4 needed a combination of adequately. Melatonin was successfully used in 2 patients. Combination therapy (lonazepam/gabapentin/melatonin) was used in 1 patient.No[136]
45 RBD patients.
77.8% Male,
mean age 66 years
Retrospective case series27–53 monthsIn 25 patients, 6 mg melatonin p.o./daily at bedtime.
In 18 patients 0.5 mg clonazepam monotherapy
Clinical Global Impression89% improvement on clonazepam, 68% improvement on melatonin.
Melatonin ↓ injuries significantly.
Patients receiving melatonin reported fewer adverse effects.
No[137]
28 patients, 72% male, 66.5 years mean age,
10 PD, 16 with OSA, 6 with cognitive decline
Retrospective review4 monthsmelatonin 3–6 mg per night for 4 months, +0.5–3 mg clonazepamPSG26 with 6 mg melatonin in monotherapy.↓ % tonic REM activity and wake after sleep onset[138]
203 consecutive patients with iRBDRetrospective case series4 years6 mg melatonin as a median dose (range 1.9–9 mg), 1 mg clonazepam as a median dose (range, 0.25–4 mg)PSGMelatonin treatment effective in 32 patients (15.7%). Clonazepam was replaced by melatonin
In 24 subjects who experienced side effects or in whom clonazepam was not effective, melatonin was given instead.
↓ % tonic REM activity after melatonin or clonazepam administration.[139]
4 RBD patients with concomitant obstructive sleep apneaOpen label4 weeks2 mg prolonged release melatonin p.o./daily at bedtimePSGTreatment led to a relevant clinical improvement of RBD symptoms in all patients.REM without atonia incidence was high probably because of the untreated comorbid condition[140]
72 year-old male iRBD patientCase report5 years2 mg prolonged release melatonin p.o./daily at bedtimePSG and DA transporter scintigraphy (DaTSCAN)In 2011, the patient was clinically suspected of PD. DaTSCAN revealed reduced DA transporter density and PSG confirmed the diagnosis of RBD. After 6 months of melatonin treatment, clinical signs of RBD were absent.Control PSG in 2014 indicated normalized REM sleep with atonia. Additional DaTSCANs were performed in 2013 and 2015 indicated normalization of DA transporter density[141]
30 iRBD patients, 66% male, mean age of 66.4 yearsDouble blind, placebo-controlled trial4 weeks2 mg or 6 mg prolonged release melatonin o placebo p.o./daily at bedtimeKorean version of the RBD questionnaire. Impression Improvement (Clinical Global Impression)Neither primary nor secondary outcomes examined were affected by treatment.No[142]
209 RBD patientsSingle-center, observational cohort study171 patients ≥6 months, 13 patients 1–3 months, 25 patients received a mixed treatment171 patients took melatonin (2 mg prolonged release), ≥6 months, always-at-the-same-clock time, 10–11pm, corrected for chronotype), 13 patients had applied melatonin for about 1–3 months, and 25 patients underwent mixed treatments1529 clinical evaluations performed, including Clinical Global Impression and RBDsymptom severity scale (Ikelos-RS), analyzed using linear mixed modelsWith melatonin, RBD symptomatology regressed gradually up to 4 weeks of treatment and remained improved thereafter. When melatonin was discontinued after 6 months, symptoms remained stably improved. When administered for only 1–3 months, RBD symptoms gradually returned.Video-PSG-confirmed RBD[4]
30 RBD patientsRandomized, double-blind, placebo controlled, parallel-group trial8 weeks4 mg prolonged release melatonin p.o./daily at bedtimeAggregate of RBD incidents as captured in sleep diariesMild adverse effects (headaches, fatigue, morning sleepiness) in 4 subjects with melatonin and in 5 with placebo.No[143]
34 RBD patientsProspective, open-label, randomized trial4 weeksClonazepam 0.5 mg or prolonged-release melatonin 2 mg 30 minbefore bedtimePSG, clinical global improvement-impression scale and sleepquestionnaire scoreRBD symptom improvement tended to be better after clonazepam than after melatonin. Daytime sleepiness and insomnia symptoms were decreased by melatonin.Clonazepam, but not PR melatonin, ↓ % RWA[144]
12 iRBD patientsopen-labeled trial ramelteon 8 mg daily for 4 weeksPSGAfter ramelteon, a trend toward significant improvement occurred.No statistically significant effect on RWA, RBD severity scale or other sleep parameters[145]
35 patients, PD + sleep disorders;
24 with PRBD
open-labeled trial12 weeksramelteon 8 mg daily,RBD questionnaire↓scores in the Japanese RBD questionnaireNo[146]
59 year-old male RBD, 76 year-old female RBD,
1 PD, 1 multiple system atrophy + OSA
case series2–3 yearsramelteon 8 mg monotherapy in 1 patient, +clonazepam 1 mg in 1 patientPSGImprovement in 2 patients, 1 with rebound after discontinuation.↓ % RWA[147]
3 iRBD patientscase review6 monthsagomelatine, 25–50 mg per nightPSG3 (1 at 25 mg, 2 at 50 mg).% of REM epochs with muscle atonia (atonic density) only slightly increased. In 2 cases the percentage of REM epochs with high tonic density decreased[148]
IRBD: idiopathic rapid eye movement sleep behavior disorder; OSA: obstructive sleep apnea; PD: Parkinson disease; REM: rapid eye movement; RWA: REM sleep without atonia.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cardinali, D.P.; Garay, A. Melatonin as a Chronobiotic/Cytoprotective Agent in REM Sleep Behavior Disorder. Brain Sci. 2023, 13, 797. https://doi.org/10.3390/brainsci13050797

AMA Style

Cardinali DP, Garay A. Melatonin as a Chronobiotic/Cytoprotective Agent in REM Sleep Behavior Disorder. Brain Sciences. 2023; 13(5):797. https://doi.org/10.3390/brainsci13050797

Chicago/Turabian Style

Cardinali, Daniel P., and Arturo Garay. 2023. "Melatonin as a Chronobiotic/Cytoprotective Agent in REM Sleep Behavior Disorder" Brain Sciences 13, no. 5: 797. https://doi.org/10.3390/brainsci13050797

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