02.05.2015 Views

Melatonin: circadian rhythm regulator, chronobiotic, antioxidant and ...

Melatonin: circadian rhythm regulator, chronobiotic, antioxidant and ...

Melatonin: circadian rhythm regulator, chronobiotic, antioxidant and ...

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

Clinics in Dermatology (2009) 27, 202–209<br />

<strong>Melatonin</strong>: <strong>circadian</strong> <strong>rhythm</strong> <strong>regulator</strong>, <strong>chronobiotic</strong>,<br />

<strong>antioxidant</strong> <strong>and</strong> beyond<br />

Bruno Berra, PhD ⁎ , Angela Maria Rizzo, PhD<br />

Institute of General Physiology <strong>and</strong> Biochemistry, “Giovanni Esposito”, University of Milan, Faculty of Pharmacy,<br />

Via Trentacoste 2, Milan 20134, Italy<br />

Abstract For many years, melatonin has been known to interact with <strong>circadian</strong> <strong>rhythm</strong>s. New evidence<br />

indicates that melatonin acts as a free radical scavenger <strong>and</strong> <strong>antioxidant</strong>. Moreover, melatonin prevents<br />

apoptosis in different types of cells, because it induces mRNA levels of several <strong>antioxidant</strong> enzymes. It<br />

is evident that melatonin is involved in the cellular bioenergetic system as a mechanism that counteracts<br />

the progression of Alzheimer's disease.<br />

© 2009 Elsevier Inc. All rights reserved.<br />

Introduction<br />

<strong>Melatonin</strong>, the major secretory product of the pineal gl<strong>and</strong>,<br />

has been known to interact with the neuroendocrine axis <strong>and</strong><br />

<strong>circadian</strong> <strong>rhythm</strong>s. Recently, it has been reported that<br />

melatonin acts as a free radical scavenger <strong>and</strong> <strong>antioxidant</strong>, 1,2<br />

<strong>and</strong> thus as an antiapopotic agent. 3 Mayo <strong>and</strong> colleagues 4<br />

reported that melatonin prevents apoptosis in undifferentiated<br />

<strong>and</strong> differentiated PC12 cells <strong>and</strong> suggested that the<br />

protective effect of melatonin may be associated with the<br />

increase in the mRNA levels of several antioxidative<br />

enzymes.<br />

Pineal gl<strong>and</strong> <strong>and</strong> melatonin<br />

In humans, the pineal gl<strong>and</strong> is 5 mm long, 1-4 mm thick,<br />

<strong>and</strong> weighs about 100 mg, in men <strong>and</strong> women. 5 The pineal<br />

gl<strong>and</strong> contains two major cell types: neuroglial cells <strong>and</strong> the<br />

predominant pinealocytes that produce melatonin. The<br />

pineal gl<strong>and</strong> is a central structure in the cardian system that<br />

⁎ Corresponding author. Tel.: +39 02 50315777; fax: +39 02 50315775.<br />

E-mail address: bruno.berra@unimi.it (B. Berra).<br />

is innervated by a neural multi-synaptic pathway originating<br />

in the suprachiasmatic nucleus (SCN) located in the anterior<br />

hypothalamus. The SCN is the major <strong>circadian</strong> pacemaker of<br />

the mammalian brain <strong>and</strong> plays a central role in the<br />

generation <strong>and</strong> regulation of biological <strong>rhythm</strong>s. 6 The pineal<br />

gl<strong>and</strong> produces melatonin in a marked <strong>circadian</strong> fashion, 7<br />

reflecting signals originating in the SCN. The human SCN<br />

innervates only a small number of hypothalamic nuclei<br />

directly. 8 However, it may impose <strong>circadian</strong> fluctuations<br />

indirectly on the organism by means of melatonin released<br />

from the pineal gl<strong>and</strong>. 9<br />

<strong>Melatonin</strong> biosynthesis <strong>and</strong> its regulation<br />

The biosynthetic pathway of melatonin has been studied<br />

thoroughly. L-trytophan is taken from circulation <strong>and</strong><br />

converted to serotonin (5-HT) by tryptophan hydroxylase. 5-<br />

HT is metabolized by the rate-limiting enzyme arylalkylamine<br />

N-acetyltransferase (AA-NAT) to N-acetyl-5-hydroxytryptamine,<br />

<strong>and</strong> in turn by hydroxyindole-O-methyltransferase to<br />

melatonin. In all vertebrates, the activity of the <strong>rhythm</strong>generating<br />

enzyme AA-NAT increases at night by a factor of<br />

7-150, depending on the species. The molecular mechanisms<br />

regulating AA-NAT also are remarkably different among<br />

0738-081X/$ – see front matter © 2009 Elsevier Inc. All rights reserved.<br />

doi:10.1016/j.clindermatol.2008.04.003


<strong>Melatonin</strong>: <strong>circadian</strong> <strong>rhythm</strong> <strong>regulator</strong>, <strong>chronobiotic</strong>, <strong>antioxidant</strong> <strong>and</strong> beyond<br />

203<br />

species. For instance, in rats, pineal AA-NAT is regulated<br />

at both the mRNA level <strong>and</strong> protein level; however, in sheep<br />

<strong>and</strong> rhesus macaque, the mRNA pineal AA-RAT levels<br />

show relatively little change over a 24-hr period, <strong>and</strong> changes<br />

in AA-NAT activity are regulated primarily at the protein<br />

level. 10 In the human pineal gl<strong>and</strong>, significant daily fluctuations<br />

in AA-NAT activity may be regulated mainly at the post<br />

transcriptional level. 11<br />

Light intensity is the main environmental control of the<br />

pineal melatonin synthesis. Light perceived by the retina<br />

reaches the SCN through the retinohypothalamic tract, which<br />

has been revealed in the human hypothalamus. 12 The SCN<br />

innervates the pineal gl<strong>and</strong>, resulting in the <strong>rhythm</strong>ic<br />

secrection of melatonin. 13 The importance of ocular light<br />

as a temporal clue has been demonstrated in <strong>circadian</strong> studies<br />

of blind people, bilaterally enucleated, showing desychronized<br />

melatonin <strong>and</strong> cortisol <strong>rhythm</strong>. 14 Abundant evidence<br />

indicates that, in humans, the sympathetic stimulus is crucial<br />

for melatonin secrection. The primary neurotransmitter<br />

released from the postganglionic sympathetic terminals in<br />

the pineal gl<strong>and</strong> is norepinephrine (NE); during darkness at<br />

night, NE is discharged onto the pinealocyte, where it<br />

couples (especially with beta 1-adrenoceptors). This process<br />

is potientated further by stimulations of alfa 1- adrenoceptors.<br />

This leads to a marked rise in intracellular cAMP levels<br />

to denovo protein synthesis <strong>and</strong> eventually to the stimulation<br />

of AA-NAT.<br />

Unlike other endocrine organs, the pineal does not store<br />

melatonin for later release after its synthesis; melatonin<br />

quickly diffuses out of the pinealocytes into the rich<br />

capillary bed within the gl<strong>and</strong> <strong>and</strong> directly into the<br />

cerebrospinal fluid. <strong>Melatonin</strong> displays high lipid <strong>and</strong><br />

water solubility, which facilitates passage across cell<br />

membranes. After release into the circulation, it gains access<br />

to various fluids, tissues, <strong>and</strong> cellular compartments (saliva,<br />

urine, cerebrospinal fluid, preovulatory follicles, semen,<br />

amniotic fluid, <strong>and</strong> milk).<br />

Three mammalian melatonin receptors have been<br />

described: MT1, MT2, MT3. The first two are G-proteincoupled<br />

receptors, <strong>and</strong> their activation modulates a wide<br />

range of intracellular messangers (eg, cAMP, cGMP, or<br />

calcium concentrations). The MT1 receptor, with high<br />

affinity, is mainly expressed in the SCN <strong>and</strong> hypophyseal<br />

pars-tuberalis. The MT1 receptor with low affinity is<br />

expressed mainly in the retina. The MT3-binding site has<br />

been identified as a quinone reductase protein, but its<br />

physiological significance needs to be clarified.<br />

A very large goiter may compress the superior cervical<br />

ganglia (SCG), thus altering melatonin synthesis in<br />

patients. 15 After bilateral T1-T2 ganglionectomy in a patient<br />

with hyperhidrosis, melatonin levels in the cerebrospinal<br />

fluid (CSF) <strong>and</strong> plasma were reduced, <strong>and</strong> the diurnal <strong>rhythm</strong><br />

was abolished. 16 A <strong>circadian</strong> <strong>rhythm</strong> of β1-adrenergic<br />

receptors has been found in human pinealocytes. 17 Propanolol,<br />

a β-adrenergic receptor antagonist, causes a dosedependent<br />

decrease in melatonin levels or completely<br />

abolishes nighttime surge. 18 In turn, melatonin elicits two<br />

distinct, separable effects on the SCN: acute neuronal<br />

inhibition <strong>and</strong> phase shifting. 19 The ability of melatonin to<br />

phase-shift in the <strong>circadian</strong> system has been investigated<br />

extensively in humans. 20<br />

Jet lag<br />

Jet lag is a considerable problem in the modern world<br />

with widespread air travel. When the internal body clock<br />

(or <strong>circadian</strong> <strong>rhythm</strong>) is not synchronized with the external<br />

“local” time (light-dark cycle), jet lag is experienced. The<br />

symptoms, which vary between individuals, include tiredness,<br />

inability to sleep at the new bedtime, inability to<br />

concentrate, disturbed sleep for several days after a long<br />

flight, headache, <strong>and</strong> gastrointestinal problems. All of these<br />

symptoms can interfere with normal activities. Jet lag also<br />

may cause considerable problems for training <strong>and</strong> performance<br />

in sports competitions <strong>and</strong> should be taken into<br />

account when planning journey times for sportsmen <strong>and</strong><br />

women. Symptoms are more marked in older travellers,<br />

when more time zones are crossed, <strong>and</strong> when travelling in<br />

an easterly direction. 21<br />

A recent Cochrane review 22 assessed 10 trials comparing<br />

melatonin with a placebo <strong>and</strong> compared it with the hypnotic<br />

zolpidem. In 9 out of 10 trials, melatonin decreased jet lag<br />

resulting from crossing five or more time zones when taken<br />

close to the desired bedtime at the destination (10 P.M. to<br />

midnight). The time of dosing was very important, because if<br />

it is taken at the wrong time, melatonin could cause a delay in<br />

adaptation to the local time. The safety profile was very high<br />

in these trials, <strong>and</strong> the authors concluded that melatonin can<br />

be recommended safely to adults travelling across five or<br />

more time zones, particularly those who have experienced jet<br />

lag previously.<br />

Delayed sleep phase disorder<br />

According to the International Classifications of Sleep<br />

Disorders, individuals with delayed sleep phase disorder<br />

(DSPD) have difficulty falling asleep at their desired bedtime<br />

<strong>and</strong> an inability to wake spontaneously at the planned<br />

time in the morning. 23 Weitzman <strong>and</strong> colleagues 24 first<br />

defined this disorder <strong>and</strong> described its characteristics: long<br />

sleep onset latencies <strong>and</strong> late sleep onset times. This is<br />

caused by a delay of the major sleep period.<br />

There is considerable evidence that DSPD arises from a<br />

delayed endogenous <strong>circadian</strong> <strong>rhythm</strong>. 25 Sleep parameters,<br />

melatonin, <strong>and</strong> core body temperature <strong>rhythm</strong>s have been<br />

delayed in individuals with sleep onset insomnia <strong>and</strong> DSPD<br />

when compared to control groups. 26 If the core body<br />

temperature <strong>and</strong> melatonin <strong>circadian</strong> <strong>rhythm</strong>s were phase<br />

delayed, then the “wake maintenance zone” would be<br />

delayed also. 27 Although <strong>circadian</strong> <strong>rhythm</strong> phase delay is<br />

seen as the major contributor to DSPD, there are some


204 B. Berra, A.M. Rizzo<br />

important behavioral <strong>and</strong> cognitive factors that should be<br />

addressed to improve treatment effectiveness.<br />

Treatments that change the <strong>circadian</strong> <strong>rhythm</strong> phase or<br />

timing (such as morning bright light, exogenous melatonin,<br />

<strong>and</strong> chronotherapy) have been effective in treating of delayed<br />

<strong>circadian</strong> <strong>rhythm</strong> sleep disorders. Bright light is an effective<br />

intervention for phase advancing <strong>circadian</strong> <strong>rhythm</strong>.<br />

The timing <strong>and</strong> duration of the light stimulus <strong>and</strong> the<br />

brightness <strong>and</strong> wavelength of light affects the magnitude of<br />

phase shift. The human phase response curve to light<br />

suggests that a phase advance of the <strong>circadian</strong> <strong>rhythm</strong> is<br />

achieved when the light stimulus is presented immediately<br />

after the normal <strong>circadian</strong> time (CT). 28<br />

Exogenous melatonin administration also is capable of<br />

shifting the <strong>circadian</strong> <strong>rhythm</strong> to a more desired time. 29 In<br />

the evening phase, melatonin advances <strong>circadian</strong> <strong>rhythm</strong><br />

when combined with optimal time of administration <strong>and</strong><br />

greater doses: (3-5 mg) 4 to 8 hours prior to the onset<br />

of endogenous melatonin <strong>and</strong> smaller doses (0.3-0.5 mg)<br />

3 hours before the beginning of melatonin production. 30 A<br />

recent study 31 showed that the addition of evening<br />

melatonin administration to morning bright light therapy<br />

produced a significantly greater phase advance than the<br />

morning bright light alone, suggesting that the two<br />

therapies are additive.<br />

Although exogenous melatonin appears to be safe with<br />

short-term use (less than 3 months), there is little<br />

information available on its long-term administration. 32<br />

Because typical doses (3-5 mg) in many studies can elevate<br />

melatonin concentrations well above normal physiological<br />

plasma levels, it would be prudent to use much lower<br />

doses. Fortunately, the <strong>chronobiotic</strong> effects of low doses<br />

(0.3-0.5 mg) appear to be sufficient without requiring<br />

excessive supraphysiological levels. 31 Adverse side effects<br />

reported following melatonin administration include headache,<br />

dizziness, nausea, <strong>and</strong> drowsiness. 32 The effects of<br />

exogenous melatonin on sleep was the object of a recent<br />

meta-analysis report, 33 which states: “ this meta analysis<br />

supports the hypothesis that melatonin decreases sleep<br />

onset latensy, increases sleep efficiency <strong>and</strong> total sleep<br />

duration. In spite of the heterogeneity of the data the<br />

present meta analysis does l<strong>and</strong> statistical support to the<br />

notion that melatonin preparations can improve sleep<br />

quality with regard to sleep onset latensy, sleep efficiency<br />

<strong>and</strong> sleep duration.”<br />

<strong>Melatonin</strong> as an <strong>antioxidant</strong>, radical scavenger,<br />

<strong>and</strong> anti aging product<br />

<strong>Melatonin</strong> is present in bacteria, plants, eukaryotes, fungi,<br />

<strong>and</strong> all phyla of multicellular animals; its original evolutionary<br />

role probably was to act as an <strong>antioxidant</strong>. 34 Its<br />

<strong>antioxidant</strong> properties have been seen in tissue cultures <strong>and</strong><br />

intact animals. One problem still discussed is whether<br />

melatonin acts in this manner directly or activates critical<br />

pathways involved in the disposition of free radicals. 35 The<br />

evidence for a direct effect is seen when melatonin acts as a<br />

power-free radical scavenger in isolated cell-free-systems 36 ;<br />

there are, however, reports that melatonin can act as a prooxidant<br />

in such systems. 37<br />

<strong>Melatonin</strong> is present within brain at a concentration of<br />

only 5% of that found in serum. 38 Therefore, unless it is<br />

highly concentrated in a localized area, it can contribute<br />

little to scavenging when compared to predominant<br />

<strong>antioxidant</strong> species like glutathione <strong>and</strong> alfa-tocopherol. 39<br />

<strong>Melatonin</strong> receptors <strong>and</strong> enzyme induction<br />

There are three major plasma membrane receptors for<br />

melatonin in the brain. However, the presence of additional<br />

melatonin binding sides in the nuclei of many cell types<br />

suggests the existence of a mechanism different from the<br />

mechanism mediated by the interaction with plasma<br />

membrane receptors. 40 The specificity of melatonin may<br />

reside in its properties as a neurohormone, which affects<br />

transcriptional events in the CNS. 41<br />

A wide range of <strong>antioxidant</strong> enzymes is induced by<br />

melatonin (eg, glutathione peroxidase, catalase, <strong>and</strong> superoxide<br />

dismutases). 42 These protein changes are paralleled by<br />

altered levels of gene expression of oxidative emzymes. 43 In<br />

addition, levels of some pro-oxidant enzymes (such as<br />

lipoxygenase <strong>and</strong> nitric oxide synthetase) are depressed after<br />

melatonin treatment. 44<br />

Presently, melatonin functions seem to fall into three<br />

categories: receptor-mediated, protein-mediated, <strong>and</strong> nonreceptor-mediated<br />

effects. Receptor-mediated melatonin<br />

events involve membrane <strong>and</strong> nuclear receptors. Although<br />

membrane melatonin receptors are well-characterized in<br />

humans, 45 some of the receptor-related <strong>antioxidant</strong> effects of<br />

melatonin seem to be related to its nuclear receptors. 46 With<br />

this information, the interaction between membrane <strong>and</strong><br />

nuclear melatonin signalling has been proposed. 47<br />

<strong>Melatonin</strong> <strong>and</strong> aging<br />

One of the most recent theoretical advances in basic<br />

gerontology is the free radical theory of aging. This thoery<br />

proposes that reactive ogygen species (ROS) (including<br />

superoxide [O ● 2], hydroxyl (OH ● ) free radicals, hydrogen<br />

peroxide [H 2 O 2 ] <strong>and</strong> possibly singlet oxygen) are generated<br />

as by-products of cellular respiration <strong>and</strong> other<br />

metabolic processes <strong>and</strong> damage cellular macromolecules.<br />

This results in mutations <strong>and</strong> genome instability, <strong>and</strong> all of<br />

these abnormalities can lead to the development of agerelated<br />

pathological phenomena, including cancer, circulatory<br />

diseases, immuno depression, brain disfunction, <strong>and</strong><br />

cataracts. 48 Much evidence indicates that aging is characterized<br />

by a progressive deterioration of <strong>circadian</strong> time<br />

keeping. 49


205<br />

<strong>Melatonin</strong>: <strong>circadian</strong> <strong>rhythm</strong> <strong>regulator</strong>, <strong>chronobiotic</strong>, <strong>antioxidant</strong> <strong>and</strong> beyond<br />

tissue <strong>and</strong> were compared with those of other known expression of apoptosis-related factors in vivo. 63<br />

Besides the age-related decline of melatonin production, <strong>antioxidant</strong>s. 54 After oxidative stress, virtually all GSH<br />

age-related changes in the timing of the melatonin <strong>rhythm</strong><br />

have been reported. 50 The possible mechanism of age-related<br />

melatonin changes were associated with changes in the<br />

pineal gl<strong>and</strong> morphology <strong>and</strong> its calcification; these data are<br />

in agreement with the assertion of decreased melatonin<br />

production with age. 51,52 These findings suggest that the<br />

changes observed in the melatonin <strong>rhythm</strong> may be part of a<br />

general effect of aging in particularly on the central clock<br />

of SCN <strong>and</strong> its regulation.<br />

in mitochondria is oxidized to GSSG, <strong>and</strong> the activity of<br />

both GPx <strong>and</strong> GRd are reduced almost to zero. In this<br />

situation, melatonin counteracted these effects, restored basal<br />

levels of GHS <strong>and</strong> the normal activities of both GPx <strong>and</strong><br />

GRd. To characterize the effects of melatonin on mitochondrial<br />

ETC activity, submitochondrial particles were used.<br />

<strong>Melatonin</strong> increased the activity of the C-I <strong>and</strong> C-IV<br />

complexes in a dose-dependent manner, as previously<br />

shown on intact mitochondria.<br />

These results suggest a direct effect of melatonin on<br />

mitochondrial energy metabolism <strong>and</strong> provide a new<br />

<strong>Melatonin</strong>, mitochondria, <strong>and</strong><br />

homeostatic mechanism for regulation of mitochondrial<br />

function. The findings also identify a new mechanism of<br />

cellular bioenergetics<br />

action of melatonin at the mitochondrial level. Thus, melatonin<br />

improves the bioenergetics of the cell, by providing<br />

Aerobic cells use oxygen for the production of 90%-95% more efficient nuclear <strong>and</strong> mitochondrial genomic repair<br />

of the total amount of ATP they use. The synthesis of ATP mechanisms, increasing GSH levels, elevating ATP production,<br />

<strong>and</strong> improving ATP-dependent functions, including<br />

is the result of electron transport along the mitochondrial<br />

electron chain, resulting in the ultimate oxygen reduction neurotransmission.<br />

<strong>and</strong> coupled to oxidative phosphorylation. Under normal<br />

conditions, a small percentage of oxygen may be reduced<br />

by one, two, or three electrons only, yielding superoxide<br />

anion, hydrogen peroxide, <strong>and</strong> the hydroxyl radical, <strong>Melatonin</strong> <strong>and</strong> Alzheimer's disease<br />

respectively. The main radical produced by mitochondria<br />

is superoxide anion, <strong>and</strong> the intramitochondria <strong>antioxidant</strong> Alzheimer's disease (AD) is characterized by the presence<br />

systems must scavenge this radical to avoid oxidative of β-amyloid deposits <strong>and</strong> neurofibrillary tangles (NFT) in<br />

damage to the mitochondrial membrane, which leads to the brains of affected individuals. The development of early<br />

impaired ATP production.<br />

diagnostic tools <strong>and</strong> quantitative markers are crucial for<br />

During aging <strong>and</strong> some neurodegenerative diseases, exploring promising therapeutic strategies. 55 Anti-inflammatory<br />

agents, <strong>antioxidant</strong>s, vaccinations, cholesterol-low-<br />

oxidatively damaged mitochondria are unable to maintain<br />

the energy dem<strong>and</strong>s of the cells, leading to a further ering agents, <strong>and</strong> hormone therapy are examples of new<br />

increased production of free radicals. Both defective ATP approaches being developed for treating or delaying the<br />

production <strong>and</strong> increased oxygen radical may induce progression of AD.<br />

mitochondrial-dependent apoptic cell death.<br />

Recent evidence indicates that melatonin reduces the<br />

<strong>Melatonin</strong> has been reported to exert neuroprotective neuronal damage mediated by oxygen-based reactive<br />

effects in several experimental <strong>and</strong> clinical situations involving<br />

neurotoxicity <strong>and</strong> exocitotoxicity. Additionally, in a radical scavenger <strong>and</strong> <strong>antioxidant</strong>. 56,57 Several clinical<br />

species in experimental models of AD by acting as a free<br />

series of pathologies in which high production of free radicals studies also have indicated that melatonin levels are<br />

is the primary cause of the diseases, melatonin is protective. decreased in AD patients. 58 Thus, melatonin's receptorindependent<br />

scavenging effects <strong>and</strong> receptor-mediated<br />

The common features of these diseases is the existence of<br />

mitochondrial damage caused by oxidative stress.<br />

influences on enzyme activities (which counteract the effect<br />

of oxidative stress) may account for its possible beneficial<br />

<strong>Melatonin</strong> <strong>and</strong> mitochondria<br />

effects in AD.<br />

Increased awareness of the role oxidative stress plays in<br />

Two main considerations support melatonin's role in<br />

mitochondrial homeostasis: 1) mitochondria produce high<br />

the pathogenesis of AD has highlighted the issue of whether<br />

oxidative damage is a fundamental step in pathogenesis or a<br />

amounts of ROS <strong>and</strong> RNS, <strong>and</strong> 2) mitochondria depend on result of the disease-associated pathology. 59,60 Several<br />

the GSH uptake from the cytoplasm, although they have GPx<br />

<strong>and</strong> GRd to maintain GSH redox cycling. Thus, the<br />

<strong>antioxidant</strong> effect of melatonin <strong>and</strong> its ability to increase<br />

GSH levels may be of great importance for mitochondrial<br />

physiology. 53<br />

The protective effects of melatonin were analyzed in<br />

isolated mitochondria prepared from rat brain <strong>and</strong> liver<br />

studies have demonstrated the presence of lipid, protein,<br />

<strong>and</strong> DNA oxidation products in postmortem examinations of<br />

the brains of AD patients. 61,62<br />

By observing the specific markers of in vivo oxidative<br />

stress <strong>and</strong> the expression of apoptotic-related factors,<br />

researchers demonstrated that melatonin suppresses brain<br />

lipid peroxidation in transgenic mice, <strong>and</strong> reduces the


206 B. Berra, A.M. Rizzo<br />

Inhibitory role of melatonin in tau protein<br />

hyperphosphorylation<br />

The cytoskeleton plays a crucial role in maintaining the<br />

highly asymmetrical shape <strong>and</strong> structural polarity of neurons<br />

essential for normal physiology. In AD, the cytoskeleton is<br />

assembled abnormally into NFT, <strong>and</strong> impairment of<br />

neurotransmission occurs. Microtubule-associated protein<br />

tau is capable of binding to tubulin to form the micro-tubules,<br />

essential structures for neuronal viability NFT.<br />

The micro-tubule-stabilizing function of tau is diminished<br />

greatly by its hyperphosphorylation, which results in<br />

poor binging to tubulin. 64 Glycogen synthase kinase-3, a<br />

downstream element of phosphoinositol-3 kinase, is one of<br />

the most active enzymes in phosphorylating tau in vivo.<br />

Protein kinase A (PKA) is another crucial kinase in ADlike<br />

tau hyperphosphorylation. Isoproterenol, the specific<br />

PKA activator, can induce tau hyperphosphorylation.<br />

Furthermore, melatonin enhances SOD activity <strong>and</strong><br />

decreases the level of MDA. It has been suggested that<br />

isoproterenol may induce abnormal hyperphosphorylation<br />

of tau through not only the activation of PKA, but also by<br />

increasing oxidative stress.<br />

Circadian <strong>rhythm</strong> disruptions in Alzheimer's disease<br />

The fragmented sleep-wake pattern that occurs in elderly<br />

people is even more pronounced in AD patients. 65 Many<br />

patients also suffer often from <strong>circadian</strong> system related<br />

bahavioral disturbances, such as daytime agitation <strong>and</strong><br />

nightly restlessness. 66<br />

<strong>Melatonin</strong> changes in preclinical <strong>and</strong> clinical<br />

Alzheimer's disease<br />

Many studies demonstrate that nocturnal melatonin levels<br />

are selectively decreased in AD, 11 <strong>and</strong> daytime melatonin<br />

levels are increased in AD patients. 67 A strong reduction was<br />

observed in postmortem CSF melatonin levels of AD<br />

patients; CSF melatonin levels in AD patients were only<br />

one-fifth of those in control subjects. 68<br />

The melatonin levels in CSF decrease with the progression<br />

of AD neuropathology. 69 More strikingly, CSF<br />

melatonin levels are already reduced in preclinical AD<br />

patients, who are cognitively intact <strong>and</strong> are showing only the<br />

earliest signs of AD neuropathology.<br />

A significant high correlation exists between pineal melatonin<br />

content <strong>and</strong> CSF melatonin levels 70 <strong>and</strong> between CSF<br />

<strong>and</strong> plasma melatonin amount, 71 suggesting that reduced<br />

melatonin levels may be an early marker for the first stages of<br />

AD that could not be monitored another way. <strong>Melatonin</strong><br />

deficiency is not only a consequence of the AD process; it<br />

may contribute to the pathogenesis of AD, because it acted as<br />

an <strong>antioxidant</strong> <strong>and</strong> neuroprotector in in vitro <strong>and</strong> in vivo<br />

experiments. 70,72<br />

Mechanism underlying the melatonin changes in<br />

the progression of Alzheimer's disease<br />

The pineal gl<strong>and</strong> shows molecular changes in preclinical<br />

<strong>and</strong> clinical AD. However, cells or afferent fibers are clear of<br />

the neuropathological hallmarks of AD (ie, neurofibrillary<br />

tangles, accumulation of neurofilaments, <strong>and</strong> hyperphosphorylated<br />

tau or β/A4 amyloid deposition). 73<br />

The <strong>circadian</strong> melatonin <strong>rhythm</strong> disappears because of<br />

decreased nocturnal melatonin levels in AD preclinical <strong>and</strong><br />

advanced patients. Moreover, the <strong>circadian</strong> <strong>rhythm</strong> of β1-<br />

adrenergic receptor mRNA disappears in both patient<br />

groups, which suggests a dysfunction of the SCN innervation<br />

to the pineal. The biological SCN clock is affected<br />

severely. It shows prominent degenerative changes 74 <strong>and</strong><br />

the typical cytoskeletal alterations caused by pretangles 75<br />

<strong>and</strong> tangles. 76 These degenerative changes in the SCN<br />

most likely result in a disrupted melatonin synthesis <strong>and</strong><br />

may underline the clinically common <strong>circadian</strong> <strong>rhythm</strong><br />

disorders in AD.<br />

The input of environmental light to the <strong>circadian</strong> timing<br />

system is also disrupted in AD. Besides the degenerative<br />

changes that are present in the SCN, several factors attenuate<br />

the input of environmental light to the <strong>circadian</strong> timing system;<br />

AD patients are exposed to less environmental light than their<br />

age-matched controls. 77 Furthermore, the retina <strong>and</strong> optic<br />

nerve show degenerative changes, but without neurofibrillary<br />

tangles, neuritic plaques, or amyloid angiopathy. 78,79<br />

<strong>Melatonin</strong> supplementation<br />

In AD patients, melatonin has been suggested to<br />

improve <strong>circadian</strong> <strong>rhythm</strong>icity, decreasing agitated behavior,<br />

confusion, <strong>and</strong> sundowning in uncontrolled studies.<br />

80,81 <strong>Melatonin</strong> also may have beneficial effects on<br />

memory, 82 possibly through protection against oxidative<br />

stress <strong>and</strong> neuroprotective capabilities. However, these<br />

suggestions need to be confirmed in well-controlled studies,<br />

<strong>and</strong> a few r<strong>and</strong>omized placebo-controlled trials of melatonin<br />

administration to AD patients did not find improved in<br />

the sleep-wake pattern. 83,84<br />

Conclusions<br />

This contribution summarizes the actions of melatonin in<br />

reducing the effects of jet lag, delayed sleep phase disorder,<br />

<strong>and</strong> molecular damage caused by free radicals. In<br />

particular, melatonin has effects the reduction of oxidative<br />

damage in the CNS as it cross the blood-brain barrier.<br />

However, it is unlikely that all the actions by which melatonin<br />

reduces free radical damage have been uncovered. The<br />

simplest way to account for the multiple effects of melatonin<br />

is to hypothesize that it modifies early alterations of<br />

gene expression consisting in the depression of mRNAs for


<strong>Melatonin</strong>: <strong>circadian</strong> <strong>rhythm</strong> <strong>regulator</strong>, <strong>chronobiotic</strong>, <strong>antioxidant</strong> <strong>and</strong> beyond<br />

207<br />

immuno-related cytokines <strong>and</strong> in the elevation of mRNA<br />

for <strong>antioxidant</strong> enzymes. Also, transcription factors are<br />

activated after binding to cytoplasmic melatonin receptors<br />

or by changes caused by melatonin acting directly on<br />

molecular receptors.<br />

The reported beneficial effects of melatonin on oxidative<br />

stress-related damage were supported by the improvement<br />

of mitochondrial function, which was achieved by<br />

counteracting the oxidation of mitochondrial membrane<br />

<strong>and</strong> resulting in the amelioration of cell bioenergetic. This<br />

leads to a more efficient nuclear genomic repair mechanism,<br />

to increased GSH levels, the elevation of ATP<br />

production, <strong>and</strong> the improvement of ATP-dependent functions,<br />

including neurotransmission. All these mechanisms<br />

may represent the basis for the potential anti-aging<br />

properties of melatonin <strong>and</strong> its effective use in neurodegenerative<br />

diseases.<br />

<strong>Melatonin</strong> could be considered an evolutionarily ancient<br />

neurohormone that has a very low toxicity <strong>and</strong> no carcinogenic<br />

properties which makes it a very safe compound that<br />

is available at a low cost. However more research is needed<br />

on the effects of therapeutic modulation of the melatoninergic<br />

system on <strong>circadian</strong> haemodynamies <strong>and</strong> the possible<br />

impact on morbility <strong>and</strong> mortality in humans.<br />

Aknowledgment<br />

The authors thank Francesca Tunesi for her collaboration<br />

in the accurate preparation of this contribution.<br />

References<br />

1. Lezoual'h F, Skutella T, Widmann M, et al. <strong>Melatonin</strong> prevents<br />

oxidative stress-induced cell death in hippocampal cells. NeuroReport<br />

1996;7:2071-7.<br />

2. Borlongan CV, Yamamoto M, Takei N, et al. Glial cell survival is<br />

enhanced during melatonin-induced neuroprotection against cerebral<br />

ischemia. FASEB J 1998;12:725-31.<br />

3. Chen ST, Chuang JI. The <strong>antioxidant</strong> melatonin reduces cortical<br />

neuronal death after intrastriatal injection of kainate in the rat. Exp<br />

Brain Res 1999;124:241-7.<br />

4. Mayo JC, sainz RM, Uria H, et al. <strong>Melatonin</strong> prevents apoptosis<br />

induced by 6-hydroxydopamine in neuronal cells: implications for<br />

Parkinson's disease. J Pineal Res 1998;24:179-92.<br />

5. Hasegawa A, Ohtsubo K, Mori W. Pineal gl<strong>and</strong> in old age; quantitative<br />

<strong>and</strong> qualitative morphological study of 168 human autopsy cases. Brain<br />

Res 1987;409:343-9.<br />

6. Swaab DF. In: Aminoff MJ, Francois B, Swaab DF, editors. The human<br />

hypothalamus basic <strong>and</strong> clinical aspects. H<strong>and</strong>book of Clinical<br />

Neurology, vol 79. Amsterdam: Elsevier; 2003. p. 63-123.<br />

7. Arendt J. <strong>Melatonin</strong> <strong>and</strong> the Mammalian Pineal Gl<strong>and</strong>. London:<br />

Chapman & Hall; 1995. p. 27-59.<br />

8. Dai J, Swaab DF, van der Vliet J, et al. Postmortem tracing reveals the<br />

organization of hypothalamic projections of the suprachiasmatic<br />

nucleus in the human brain. J Comp Neurol 1998;400:87-102.<br />

9. Reiter RJ. The melatonin <strong>rhythm</strong>: both a clock <strong>and</strong> a calendar.<br />

Experientia 1993;49:654-64.<br />

10. Coon SL, Del Olmo E, Young III WS, et al. <strong>Melatonin</strong> synthesis<br />

enzymes in Mecaca mulatta: focus on arylalkylamine N-acetyltransferase<br />

(EC 2. 3. 1. 87). J Clin Endocrinol Metab 2002;87:4699-706.<br />

11. Wu YH, Feenstra MG, Zhou JN, et al. Molecular changes underlying<br />

reduced pineal melatonin levels in Alzheimer disease: alterations in<br />

preclinical <strong>and</strong> clinical stages. J Clin Endocrinol Metab 2003;88:<br />

5898-906.<br />

12. Dai J, Van Der Vliet J, Swaab DF, et al. Humans retinohypothalamic<br />

tract as revealed by in vitro post-mortem tracing. J Comp Neurol 1998;<br />

397:357-70.<br />

13. Teclemariam-Mesbah R, Ter Horst GJ, Postema F, et al. Anatomical<br />

demonstration of the suprachiasmatic nucleus-pineal pathway. J Comp<br />

Neurol 1999;406:171-82.<br />

14. Skene DJ, Lockely SW, James K, et al. Correlation between urinary<br />

cortisol <strong>and</strong> 6-sulphatoxymeltonin <strong>rhythm</strong>s in field studies of blind<br />

subjects. Clin Endocrinol 1999;50:715-9.<br />

15. Karasek M, Stankiewicz A, B<strong>and</strong>urska-Stankiewicz E, et al. <strong>Melatonin</strong><br />

concentrations in patients with large goiter before <strong>and</strong> after surgery.<br />

Neuroendocrinol Lett 2000;21:437-9.<br />

16. Bruce J, Tamarkin L, Riedel C, et al. Sequential cerebrospinal fluid <strong>and</strong><br />

plasm sampling in humans: 24-hour melatonin measurements in normal<br />

subjects <strong>and</strong> after peripheral sympathectomy. J Clin Endocrinol Metab<br />

1991;72:819-23.<br />

17. Oxenkrug GF, Anderson GF, Gragovic L, et al. Circadian <strong>rhythm</strong>s of<br />

human pineal melatonin, related indoles, <strong>and</strong> beta adrenoreceptors:<br />

post-mortem evaluation. J Pineal Res 1990;9:1-11.<br />

18. Stoschitzky K, Sakotnik A, Lercher P, et al. Influence of beta-blockers<br />

on melatonin release. Eur J Clin Pharmacol 1999;55:111-5.<br />

19. Liu C, Weaver DR, Jin X, et al. Molecular dissection of two dinstinct<br />

actions of melatonin on the suprachiasmatic <strong>circadian</strong> clock. Neuron<br />

1997;19:91-102.<br />

20. Fisher S, Smolnik R, Herms M, et al. <strong>Melatonin</strong> acutely improves the<br />

neuroendocrine architecture of sleep in blind individuals. J Clin<br />

Endocrinol Metab 2003;88:5315-20.<br />

21. Spitzer RL, Terman M, Williamns JBW, et al. Jet lag: clinical features,<br />

validation of a new syndrome-specific scale, <strong>and</strong> lack of response to<br />

melatonin in a r<strong>and</strong>omised, double-blind trial. Am J Psychiatry 1999;<br />

156:1392-6.<br />

22. Herxhaimer A, Petrie KJ. <strong>Melatonin</strong> for the prevention <strong>and</strong> treatment of<br />

jet lag (Cochrane Review). Cochrane Library, Issue 3. Oxford: Update<br />

Software; 2001. p. 8.<br />

23. Westchester IL. The international classification of sleep disorders:<br />

diagnostic <strong>and</strong> coding manual. Am Acad Sleep Med 2005.<br />

24. Weitzman ED, Czeister CA, Coleman RM, Spielman AJ, Zimmerman<br />

JC, Dement W. Delayed sleep phase syndrome: a chronobiological<br />

disorder with sleep-onset insomnia. Arch Gen Psychiatry 1981;38:<br />

137-46.<br />

25. Kerkhof G, Vian Vianen B. Circadian phase estimation of chronic<br />

insomniacs relates to their sleep characteristics. Arch Physiol Biochem<br />

1999;107:383-92.<br />

26. Watanabe T, Kajimura N, Kato M, et al. Sleep <strong>and</strong> <strong>circadian</strong> <strong>rhythm</strong><br />

disturbances in patients with Delayed Sleep Phase Syndrome. Sleep<br />

2003;26:657-61.<br />

27. Strogatz Sh, Kronauer RE. Circadian wake maintenance zones <strong>and</strong><br />

insomnia in man. Sleep Res 1985;14:219.<br />

28. Dawson D, Lack L, Morris M. Phase resetting of the human <strong>circadian</strong><br />

pacemaker with use of a single pulse of bright light. Chronobiol Int<br />

1999;10:94-102.<br />

29. Sack R, Lewy A, Hughes R. Use of melatonin for sleep <strong>and</strong> <strong>circadian</strong><br />

<strong>rhythm</strong> disorders. Ann Med 1998;30:115-21.<br />

30. Lewy A, Ahmed S, Jackson JM, Sack RL. <strong>Melatonin</strong> shifts human<br />

<strong>circadian</strong> <strong>rhythm</strong>s according to phase-response curve. Chronobiol Int<br />

1992;9:380-92.<br />

31. Revell VL, Burgess HJ, Gazda CJ, Smith MR, Fogg LF, Eastman CI.<br />

Advancing human <strong>circadian</strong> <strong>rhythm</strong>s with afternoon melatonin <strong>and</strong><br />

morning intermittent bright light. J Clin Endocrinol Metab 2006;91:<br />

54-9.


208 B. Berra, A.M. Rizzo<br />

32. Buscemi N, V<strong>and</strong>ermeer B, Hooton N, et al. The efficacy <strong>and</strong> safety of<br />

exogenous melatonin for primary sleep disorders: A meta-analysis.<br />

J Gen Intern Med 2005;20:1151-8.<br />

33. Brzezinski A, Vangel MG, Wurtman RJ, et al. Effects of exogenous<br />

melatonin on sleep: a meta-analysis. Sleep Med Rev 2005;9:41-50.<br />

34. Hardel<strong>and</strong> R, Poeggeler B. Non-vertebrate melatonin. J Pineal Res<br />

2003;34:233-4.<br />

35. Srinivasan V, P<strong>and</strong>i-Perumal SR, Cardinali DP, Poeggler B, Harderl<strong>and</strong><br />

R. <strong>Melatonin</strong> in Alzheimer's disease <strong>and</strong> other neurodegenerative<br />

disorders. Behav Brain Funct 2006:2-15.<br />

36. Tan DX, Reiter RJ, Manchester LC, et al. Chemical <strong>and</strong> physical properties<br />

<strong>and</strong> potential mechanism: melatonin as a broad spectrum <strong>antioxidant</strong><br />

<strong>and</strong> free rdical scavenger. Curr Top Med Chem 2002;2:181-97.<br />

37. Buyukavci M, Ozdemir O, Buck S, Stout M, Ravindranath Y, Savasan<br />

S. <strong>Melatonin</strong> cytotoxicity in human leukaemia cells: relation with its<br />

prooxidant effect. Fundam Clin Pharmacol 2006;20:73-9.<br />

38. Lahiri DK, Ge YW, Sharman EH, Bondy SC. Age-related changes in<br />

serum melatonin in mice: higher levels of combined melatonin <strong>and</strong> 6-<br />

hydroxymelatonin sulphate in the cerebral cortex than serum, heart,<br />

liver <strong>and</strong> kidney tissues. J Pineal Res 2004;36:217-23.<br />

39. Sanchez-Moreno C, Dorfman SE, Lichtenstein AH, Martin A. Dietary<br />

fat type affects vitamins C <strong>and</strong> E <strong>and</strong> biomarkers of oxidative status in<br />

peripheral <strong>and</strong> brain tissue of golden Syrian hamsters. J Nutr 2004;134:<br />

655-60.<br />

40. Filadelfi AM, Castrucci AM. Comparative aspects of the pineal<br />

melatonin system of poikilothermic vertebrates. J Pineal Res 1996;20:<br />

175-86.<br />

41. Kotler M, Rodriguez C, Sainz RM, Antolin I, Menendez-Pelaez A.<br />

<strong>Melatonin</strong> increases gene expression for <strong>antioxidant</strong> enzymes in rat<br />

brain cortex. J Pineal Res 1998;24:83-9.<br />

42. Rodriguez C, Mayo JC, Sainz RM, Antolin I, Herrera F, Martin V,<br />

Reiter RJ. Regulation of <strong>antioxidant</strong> enzymes: a significant role for<br />

melatonin. J Pineal Res 2004;36:1-9.<br />

43. Anisimov SV, Popovic N. Genetic aspects of melatonin biology. Rev<br />

Neurosci 2004;15:209-30.<br />

44. Reiter RJ, Acuna-Castroviejo D, Tan DX, Burkhardt S. Free radicalmediated<br />

molecular damage. Mechanisms for the protective actions of<br />

melatonin in the central nervous system. Ann N Y Acad Sci 2001;939:<br />

200-15.<br />

45. Conway S, Drew JE, Mowat P, Barret P, Delagrange P, Morgan PJ.<br />

Chimeric melatonin mtl <strong>and</strong> melatonin-related receptors. Identification<br />

of domains <strong>and</strong> residues participating in lig<strong>and</strong> binding <strong>and</strong> receptor<br />

activation of the melatonin mtl receptor. J Biol Chem 2000;275:<br />

20602-9.<br />

46. Garcia-Maurino S, Pozo D, Calvo JR, Guerrero JM. Correlation<br />

between nuclear melatonin receptor expression <strong>and</strong> enhanced cytokine<br />

production in human lymphocytic <strong>and</strong> monocytic cell lines. J Pineal Res<br />

2000;29:129-37.<br />

47. Carlberg C, Wiesenberg I. The orphan receptor family RZR/ROR,<br />

melatonin <strong>and</strong> 5-lipoxygenase. An unexpected relationship. J Pineal<br />

Res 1995;18:171-8.<br />

48. Yu BP, Chung HV. Adaptive mechanism to oxidative stress during<br />

aging. Mech Ageing Dev 2006;127:436-43.<br />

49. Skene DJ, Swaab DF. <strong>Melatonin</strong> <strong>rhythm</strong>icity: effect of age <strong>and</strong><br />

Alzheimer's disease. Exp Gerontol 2003;38:199-206.<br />

50. Duffy JF, Zeitzer JM, Rimmer DW, et al. Peak of <strong>circadian</strong> melatonin<br />

<strong>rhythm</strong> occurs later within the sleep of older subjects. Am J Physiol<br />

Endocrinol Metab 2002;282:E297-303.<br />

51. Kunz D, Schmitz S, Mahlberg R, et al. A new concept for melatonin<br />

deficit: on pineal calcification <strong>and</strong> melatonin excretion. Neuropsychopharmacology<br />

1999;21:765-72.<br />

52. Kunz D, Bes F, Schlattmann P, et al. On pineal calcification <strong>and</strong> its<br />

relation to subjective sleep perception: a hypothesis-driven pilot study.<br />

Psychiatry Res 1998;82:187-91.<br />

53. Urata Y, Honma S, Goto S, et al. <strong>Melatonin</strong> induces glutamylcysteine<br />

synthase mediated by activator protein-I in human vascular endothelial<br />

cells. Free Radic Biol Med 1999;27:838-47.<br />

54. Martin M, Macias G, Escames G, et al. <strong>Melatonin</strong>-induced increased<br />

activity of the respiratory chain complexes I <strong>and</strong> IV can prevent<br />

mitochondrial damage induced by ruthenium red in vivo. J Pineal Res<br />

2000;28:242-8.<br />

55. Harrison T, Churcher I, Beher D. Gamma-secretase as a target for drug<br />

intervention in Alzheimer's disease. Curr Opin Drug Discov Devel<br />

2004;7:709-19.<br />

56. Zatta P, Tognon G, Carampin P. <strong>Melatonin</strong> prevents free radical<br />

formation due to the interaction between beta-amyloid peptides <strong>and</strong><br />

metal ions [Al(III), Zn(II), Cu(II), Mn(II), Fe(II)]. J Pineal Res 2003;35:<br />

98-103.<br />

57. Acuna-Castroviejo D, Martin M, Macias M, et al. <strong>Melatonin</strong>,<br />

mitochondria, <strong>and</strong> cellular bioenergetics. J Pineal Res 2001;30:<br />

65-74.<br />

58. Liu RY, Zhou JN, Van Heerikhuize J, Hofman MA, Swaab DF.<br />

Decreased melatonin levels in postmortem cerebrospinal fluid in<br />

relation to aging, Alzheimer's disease, <strong>and</strong> apolipoprotein E-epsilon4/4<br />

genotype. J Clin Endocrinol Metab 1999;84:323-7.<br />

59. Beal MF. Mitochondria, free radicals, <strong>and</strong> neurodegeneration. Curr<br />

Opin Neurobiol 1996;6:661-6.<br />

60. Floyd RA, Hensley K. Oxidative stress in brain aging, Implications fot<br />

therapeutics of neurodegenerative diseases. Neurobiol Aging 2002;23:<br />

795-807.<br />

61. Huang X, Moir RD, Tanzi RE, Bush AI, Rogers JT. Redox-active<br />

metals, oxidative stress, <strong>and</strong> Alzheimer's disease pathology. Ann N Y<br />

Acad Sci 2004;1012:153-63.<br />

62. Zhu X, Raina AK, Perry G, Smith MA. Alzheimer's disease:the two-hit<br />

hypothesis. Lancet Neurol 2004;3:219-26.<br />

63. Cheng Y, Feng Z, Zhang QZ, Zhang JT. Beneficial effects of melatonin<br />

in experimental models of Alzheimer disease. Acta Pharmacol Sin<br />

2006;2:129-39.<br />

64. Blennow K, Cowburn RF. The neurochemistry of Alzheimer's disease.<br />

Acta Neurol Sc<strong>and</strong> 1996;168(Suppl):77-86.<br />

65. Ancoli -Israel S, Klauber MR, Jones DW, et al. Variations in <strong>circadian</strong><br />

<strong>rhythm</strong>s of activity, sleep, <strong>and</strong> light exposure related to dementia in<br />

nursing-home patients. Sleep 1997;20:18-23.<br />

66. Martin J, Marler M, Shochat T, et al. Circadian <strong>rhythm</strong>s of agitation in<br />

institutionalized patients with Alzheimer's disease. Chronobiol Int<br />

2000;17:405-18.<br />

67. Ohashi Y, Okamoto N, Uchida K, et al. Daily <strong>rhythm</strong> of serum<br />

melatonin levels <strong>and</strong> effect of light exposure in patients with dementia<br />

of the Alzheimer's type. Biol Psychiatry 1999;45:1646-52.<br />

68. Wu YH, Swaab DF. The human pineal gl<strong>and</strong> <strong>and</strong> melatonin in aging<br />

<strong>and</strong> Alzheimer's disease. J Pineal Res 2005;38:145-52.<br />

69. Zhou JN, Liu RY, Kamphorst W, et al. Early neuropathological<br />

Alzheimer's changes in aged individuals are accompanied by<br />

decreased cerebrospinal fluid melatonin levels. J Pineal Res 2003;35:<br />

125-30.<br />

70. Pappola MA, Chyan YJ, Poeggeler B, et al. An assessment of the<br />

<strong>antioxidant</strong> <strong>and</strong> antiamyloidogenic properties of melatonin: Implications<br />

for Alzheimer's disease. J Neural Transm 2000;107:203-31.<br />

71. Rousseau A, Petren S, Plannthin J, et al. Serum <strong>and</strong> cerebrospinal fluid<br />

concentrations of melatonin: a pilot study in healthy male volunteers.<br />

J Neural Transm 1999;106:883-8.<br />

72. Reiter RJ, Tan DX, Machester LC, et al. <strong>Melatonin</strong> reduces oxidant<br />

damage <strong>and</strong> promotes mitochondrial respiration: implications for aging.<br />

Ann N Y Acad Sci 2002;959:238-50.<br />

73. Pardo CA, Martin LJ, Troncoso JC, et al. The human pineal gl<strong>and</strong> in<br />

aging <strong>and</strong> Alzheimer's disease: patterns of cytoskeletal antigen<br />

immunoreactivity. Acta Neuropathol 1990;80:535-40.<br />

74. Liu RY, Zhou JN, Hoogenduk WJ, et al. Decreased vasopressing<br />

gene expression in the biological clock of Alzheimer disease patients<br />

with <strong>and</strong> without depression. J Neuropathol Exp Neurol 2000;59:<br />

314-22.<br />

75. Van De Nes JA, Kamphorst W, Ravid R, et al. Comparison of betaprotein/A4<br />

deposits <strong>and</strong> Alz-50-stained cytoskeletal changes in the<br />

hypothalamus <strong>and</strong> adjoining areas of Alzheimer's disease patients:


<strong>Melatonin</strong>: <strong>circadian</strong> <strong>rhythm</strong> <strong>regulator</strong>, <strong>chronobiotic</strong>, <strong>antioxidant</strong> <strong>and</strong> beyond<br />

209<br />

amorphic plaques <strong>and</strong> cytoskeletal changes occur independently. Acta<br />

Neuropathol 1998;96:129-38.<br />

76. Stopa EG, Volicer L, Kuo-Leblanc V, et al. Pathologic evaluation of the<br />

human suprachiasmatic nucleus in severe dementia. J Neuropathol Exp<br />

Neurol 1999;58:29-39.<br />

77. Campbell SS, Kripke DF, Gillin JC, et al. Exposure to light in healthy<br />

elderly subjects <strong>and</strong> Alzheimer's patients. Physiol Behav 1998;42:<br />

141-4.<br />

78. Blanks JC, Torigoe Y, Hinton DR, et al. Retinal pathology in<br />

Alzheimer's disease. I. Ganglion cell loss in foveal/parafoveal retina.<br />

Neurobiol Aging 1996;17:377-84.<br />

79. Blanks JC, Schmidt SY, Torigoe T, et al. Retinal pathology in<br />

Alzheimer's disease. II. Regional neuron loss <strong>and</strong> glial changes in<br />

GCL. Neurobiol Aging 1996;17:385-95.<br />

80. Cohen-Mansfield J, Garfinkel D, Lipson S. <strong>Melatonin</strong> for treatment of<br />

sundowning in elderly persons with dementia – a preliminary study.<br />

Arch Gerontol Geriatr 2000;31:65-76.<br />

81. Brusco LI, Marquez M, Cardinali DP. <strong>Melatonin</strong> treatment stabilizes<br />

chronobiologic <strong>and</strong> cognitive symptoms in Alzheimer's disease.<br />

Neuroendocrinol. Letter 2000;21:39-42.<br />

82. Cardinali DP, Brusco LI, Liberczuk C, et al. REVIEW. The use of<br />

melatonin in Alzheimer's disease. Neuroendocrinol Lett 2002;23:20-3.<br />

83. Singer C, Tractenberg RE, Kaye J, et al. A multicenter, placebocontrolled<br />

trial of melatonin for sleep disturbance in Alzheimer's<br />

disease. Sleep 2003;26:893-901.<br />

84. Sefarty M, Kennel-Webb S, Warner J, et al. Double blind rendomised<br />

placebo controlled trial of low dose melatonin for sleep disorders in<br />

dementia. Int J Geriatr Psychiatry 2002;17:1120-7.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!