You are on page 1of 36

J. Pineal Res.

2012; 52:167–202  2011 John Wiley & Sons A/S


Doi:10.1111/j.1600-079X.2011.00937.x Journal of Pineal Research

REVIEW ARTICLE
AlzheimerÕs disease: pathological mechanisms and the beneficial
Molecular, Biological, Physiological and Clinical Aspects of Melatonin

role of melatonin
Abstract: AlzheimerÕs disease (AD) is a highly complex neurodegenerative Sergio A. Rosales-Corral1,2, Dario
disorder of the aged that has multiple factors which contribute to its etiology Acuña-Castroviejo3, Ana Coto-
in terms of initiation and progression. This review summarizes these diverse Montes2, Jose A. Boga2, Lucien
aspects of this form of dementia. Several hypotheses, often with overlapping C. Manchester2, Lorena Fuentes-
features, have been formulated to explain this debilitating condition. Perhaps Broto2, Ahmet Korkmaz2, Shuran
Ma2, Dun- Xian Tan2 and Russel
the best-known hypothesis to explain AD is that which involves the role of
J. Reiter2
the accumulation of amyloid-b peptide in the brain. Other theories that have
1
been invoked to explain AD and summarized in this review include the Centro de Investigación Biomédica de
Occidente del Instituto Mexicano del Seguro
cholinergic hypothesis, the role of neuroinflammation, the calcium Social, Guadalajara, Jalisco, México;
hypothesis, the insulin resistance hypothesis, and the association of AD with 2
Department of Cellular and Structural Biology,
peroxidation of brain lipids. In addition to summarizing each of the theories University of Texas Health Science Center at
San Antonio, San Antonio, TX, USA;
that have been used to explain the structural neural changes and the 3
Departamento de Fisiologı́a, Instituto de
pathophysiology of AD, the potential role of melatonin in influencing each Biotecnologı́a, Universidad de Granada,
of the theoretical processes involved is discussed. Melatonin is an Granada, Spain

endogenously produced and multifunctioning molecule that could


theoretically intervene at any of a number of sites to abate the changes Key words: AlzheimerÕs disease, amyloid-b
associated with the development of AD. Production of this indoleamine peptide, calcium, cholinergic
diminishes with increasing age, coincident with the onset of AD. In addition neurotransmission, free radicals,
inflammation, insulin resistance, melatonin,
to its potent antioxidant and anti-inflammatory activities, melatonin has a oxidative stress
multitude of other functions that could assist in explaining each of the
hypotheses summarized above. The intent of this review is to stimulate
Address reprint requests to Sergio A. Rosales-
interest in melatonin as a potentially useful agent in attenuating and/or Corral, Centro de Investigación Biomédica de
delaying AD. Occidente del Instituto Mexicano del Seguro
Social, Sierra Mojada 800 colonia Indepen-
dencia, Guadalajara, Jalisco, CP 45150,
México.
E-mail: espiral17@gmail.com
Received September 30, 2011;
Accepted October 4, 2011.

Introduction There currently is no cure for AD. A recent meta-analysis


of functional outcomes for commercially available acetyl-
AlzheimerÕs disease (AD) is a devastating disorder affecting cholinesterase inhibitors and memantine in the treatment of
around 35 million people worldwide [1]. Ten years ago, patients with AD, the only FDA-approved drugs for AD,
there were 4.5 million persons with AD in the US popu- revealed only a modest trend favoring active treatment over
lation alone; the number has increased to 5.3 million people placebo [4, 5]. Anti-inflammatory agents may reduce the
in 2010, according to the AlzheimerÕs Association [2]. It is risk of developing AD [6], but, on the contrary, according
estimated there will be 13.2 million people with this to results obtained from an elderly community–based
neurodegenerative disorder by 2050 [3]. cohort study, anti-inflammatory agents could even be
AlzheimerÕs disease is a primary, progressive neurological dangerous for cognitive abilities [7]. It is also possible that
disease, which is of unknown etiology in more than 90% of anti-inflammatory drugs have no influence at all with the
the cases. Some characteristic neuropathological and neu- exemption of their well-known collateral effects [8, 9].
rochemical features lead to irreversible loss of neurons. Vitamin E, estrogens, omega-3 fatty acids, and Ginkgo
Owing to the nature of the primarily affected neuronal biloba have been tested in different studies, and they yielded
circuits, the clinical hallmarks of AD are progressive contradictory results. And there is a long list of experi-
impairment in memory, judgment, decision making, orien- mental therapies targeting different protagonists in the
tation to physical surroundings, and distorted language. pathology of AD, such as tau protein, amyloid-b (Ab
This illness is the leading cause of dementia in older people targets: formation, aggregation, or toxicity), Ab receptors
[2]. or N-methyl-D-aspartate (NMDA) antagonists, serotonin

167
Rosales-Corral et al.

Fig. 1. Major targets for AlzheimerÕs


disease therapy.

receptors, loss of acetylcholine neurons, cholesterol, and


Melatonin levels in AD
antiaging drugs (reviewed in [10]; Fig. 1).
Likewise, there is insufficient clinical evidence to support Cerebral spinal fluid (CSF) melatonin levels are reportedly
the effectiveness of melatonin by itself in managing the significantly decreased in aged individuals with early
cognitive and noncognitive sequelae of people with demen- neuropathological AD-related changes in the temporal
tia [11]. However, there are molecular and physiological cortex [22]. In aged patients, melatonin levels in CSF have
bases that are worth analyzing, because melatonin may been found to be one-half those in young control subjects,
have an effective influence on several of the above- but in patients with AD, the CSF melatonin levels are only
mentioned AD protagonists and the most prominent one-fifth those in young subjects [23]. In fact, it is possible
hypotheses to explain the cause of this disease, as reviewed to replicate hippocampal CA1 and CA3 pyramidal neuron
below. There is also a growing body of evidence indicating loss in rats by merely removing the pineal gland (which
the potential role of melatonin as an effective adjuvant in lowers melatonin levels) with this effect being reversed by
AD management [12–17]. On the contrary, there is one melatonin replacement in the drinking water [24]. Also,
report indicating essentially negative results after using constant light exposure, which decreases serum melatonin,
melatonin in patients with AD [18]. Even worse, there is a is enough to cause Alzheimer-like damage, such as memory
single recent publication which claims that melatonin may deficits, tau hyperphosphorylation at multiple sites, activa-
aggravate this neurodegenerative disorder [19]. tion of glycogen synthase kinase-3 and protein kinase A, as
The authors of the current review declare no conflict of well as suppression of protein phosphatase-1 and promi-
interest related to this paper or financial relationships with nent oxidative stress [25].
commercial entities. The aim is to put together evidence on
melatoninÕs role in the best-known hypotheses that cur-
Melatonin, mechanisms of action
rently attempt to explain AD pathogenic mechanisms,
starting with the fact that AD-related changes begin at the Melatonin is derived from the aminoacid tryptophan in a
age when melatonin levels fall significantly. However, it is multistep process involving the synthesis of serotonin,
also clear from the beginning that more than 100 yr after which is subsequently N-acetylated and O-methylated [26].
the first clinical report of a case of AD, there is not yet a Melatonin is 5-methoxy-N-acetyltryptamine produced by
satisfactory hypothesis or a model capable of explaining or the functional elements of the pineal gland; once released, it
reproducing the pathogenic mechanisms of this devastating acts both as an endocrine product and as an antioxidant.
disease. Thus, all the proposed treatments for AD are Several precursors of melatonin including tryptophan and
groping for optimal experimental outcomes in regard to serotonin are reduced by aging, and their reduction may be
obviously incomplete hypotheses. This incompleteness may linked to AD appearance [27, 28] (Fig. 2). Tryptophan
explain, at least in part, how different models yield widely deficiency is related to an accelerated degradation attrib-
different results. For example, long-term oral administra- uted, as reviewed below, to homocysteine, a risk factor for
tion of melatonin in an amyloid precursor protein dementia and AD [29]. Serotonin deficiency, on the other
(APP) + PS1 double transgenic mice model of AD pro- hand, is linked to severe psychiatric symptoms in AD [28],
tects against cognitive deficits and markers of neurode- although serotonin dysfunction may appear long before
generation [20], while it fails to protect animals expressing psychiatric symptoms; these symptoms are associated with
the Swedish AD mutant gene (Tg2576 mice) exposed to altered brain serotonin transporter and glucose metabolism
aluminum [21]. as identified using in vivo molecular imaging [30].

168
AlzheimerÕs disease and melatonin

Fig. 2. Melatonin and its precursors,


tryptophan and serotonin (*), appear
reduced in aging, which is particularly
significant in AlzheimerÕs disease brain.

Besides the pineal gland, melatonin is also presumably Currently, melatonin is recognized both as a free radical
produced in gastrointestinal tract, airway epithelium, pan- scavenger and as an antioxidant [59]. Thanks to its electron-
creas, adrenal glands, thyroid gland, thymus, urogenital rich aromatic indole ring, melatonin directly donates an
tract, placenta, and other organs [31]. Even nonendocrine electron to free radicals at a potential of 715 mV and avoids
cells, such as mast cells, natural killer cells, eosinophilic redox recycling (reviewed in [60]), while it scavenges, with
leukocytes, platelets, and endothelial cells, may produce varying degrees of efficiency, the hydroxyl radical [56, 60–
melatonin [32]; this wide synthesis underlines its diverse 64], hydrogen peroxide [65], hypochlorous acid [66], singlet
physiological activities: from the control of biological oxygen [67], superoxide anion radical [68], nitric oxide [69],
rhythms [33, 34], metabolism of free radicals [35–39], and the peroxynitrite anion [70] (Fig. 3). Radicals may also
immune responsiveness [40–42], monitoring of mood and be added in the C3 amide side chain of melatonin, which
sleep [43–45], cell proliferation and differentiation [46, 47], possesses an N–C=O functional group [71]. The rate
and control of seasonal reproduction [48, 49]. Importantly, constant for the scavenging of the ·OH by melatonin is
melatonin production declines with age [50–53] because of calculated to be on the order of 2.7 · 1010/M/s [72].
dysfunction of the sympathetic regulation of pineal mela- The free radical-scavenging capacity of melatonin also
tonin synthesis by the suprachiasmatic nucleus (SCN), a extends to its secondary, tertiary, and quaternary meta-
condition probably linked to early AD stages, once that the bolites in a free radical-scavenging cascade that prolongs
reactivation of the circadian system using light therapy and its useful life [73–75]. Thus, the interaction of melatonin
melatonin has shown promising positive results [54]. with free radicals produces the oxidative pyrrole-ring
Firmly established as the key mediator controlling cleavage, giving N1-acetyl-N2-formyl-5-methoxykynur-
circadian rhythms [33, 34, 55], it has been discovered that amine (AFMK), and this substituted kynuramine may
melatonin, a small, lipophilic molecule, also had the donate two electrons at different potentials (456 and
capacity to directly scavenge the hydroxyl radical (·OH) 668 mV, respectively) to function as a reductive molecule
[56, 57]. Almost immediately, a link between melatoninÕs capable to destroy reactive species and to protect macro-
hydroxyl radical-scavenging activity and aging was envi- molecules against oxidative damage [76]. Thus, via the
sioned [58] as well as realizing that aging and Ab-induced AFMK pathway, a single melatonin molecule may scav-
oxidative stress play a key role in AD as well. enge up to 10 reactive oxygen and reactive nitrogen species

Fig. 3. Major oxidant pathways and the


role of melatonin as an antioxidant
(dashed blue arrows), promoting the
activity of antioxidant enzymes. Derived
from metabolic activity, particularly
from mitochondria in aging, melatonin
plays an important role as a free radical
scavenger (blue balloons).

169
Rosales-Corral et al.

(ROS/RNS) [73]. Further AFMK deformylation by the diabetic skin fibroblasts demonstrated that melatonin
action of arylamineformamidase or hemoperoxidase en- increases the activity of superoxide dismutase (SOD),
zymes produces N1-acetyl-5-methoxykynuramine (AMK) catalase (CAT), and glutathione peroxidase (GPx) and
[77], which, in addition to its ability to react with various the level of glutathione (GSH) [87] (Fig. 3). Similar results
oxidizing and nitrosating free radical species, particularly have been obtained in fetal rat brain [88], in experimental
singlet oxygen and nitrogen species, also may destroy brain trauma [89], as well as in cultured dopaminergic cells
carbonate and peroxyl radicals (reviewed in [78]) and [90] and, of course, in AD transgenic mouse brain [91, 92].
function as an antioxidant [79]. AFMK and AMK metab- These observations allow for the conclusion that melatonin
olism may lead to other oxidation products, such as 3- exerts an antioxidant action by increasing the mRNA levels
indolinone, cinnolinone, and quinazoline compounds, for of the antioxidant enzymes SOD, CAT, and the GSH
which no specific functions have been identified to date [78]. system, but it may depend somewhat on the model system
Interestingly, the parallel orientation of b-sheets, such as investigated.
tau and Ab filaments, generates channels extending along Finally, melatonin exhibits a lower-affinity binding site to
the length of the filament to which aromatic small mole- a cytosolic quinine oxidoreductase 2 or QR2, also known as
cules such as indolinones can bind via p–p interactions, MT3 (reviewed in [93]). This enzyme, as any other quinone,
stacked arrangement of aromatic molecules [80]. Using has the ability to transform its substrates into more highly
fluorescence spectroscopy, atomic force microscopy, and reactive compounds that are able to cause cellular damage.
electron microscopy to screen 29 indole derivatives, Cohen Once melatonin binds to its large active site, the enzyme
et al. [81] identified three potent inhibitors of amyloid fibril produces fewer hydrogen bonds and hydrophobic contacts,
formation and cytotoxicity, and the indole-3-carbinol was which diminishes their reactivity [94] (Fig. 4).
among them. The interaction of melatonin with Ab will be MelatoninÕs best-known receptors, MT1 and MT2, are
reviewed below. Additionally, the 3-substituted indolinones transmembrane G-protein-coupled heterodimers whose
have been identified as kinase inhibitors [82], which could signaling pathways lead to downstream effects on Ca2+
be related to the anti-inflammatory actions of melatonin channels, Ca2+ signaling, and changes in MAP and ERK
and its metabolites [83]. kinases and/or PI3K/Akt pathways [95] (Fig. 4). This
Melatonin may also prevent abnormal elevation of means a broad spectrum of possibilities for melatonin is
reactive nitrogen species, stimulate other antioxidant sys- one factor that gives the indoleamine a pleiotropic nature
tems, and/or inhibit some pro-oxidant enzymes; these [96, 97].
indirect actions of melatonin contribute to its potent Once again, it is worth noting that the pathogenic
antioxidant activity [84–86]. An evaluation in human mechanisms in AD are not well understood, and there are

Fig. 4. Receptor-mediated or acting directly on its substrates, melatonin exhibits a broad diversity of effects to reduce neurodegenerative
changes in the central nervous system. It is a pleiotropic indoleamine, actually. All the protagonists in neurodegenerative diseases express
melatonin receptors; when and how cells or their molecular effectors become activated or inhibited according to the expression of their
melatonin receptors remains unclear. The scheme shows some of the published observations to date, all related to central nervous system
and/or neurodegeneration. Thanks to its ability to transfer electrons, melatonin may repair damaged biomolecules derived from DNA
oxidation, such as guanosine. LTP, long-term potentiation; MT, melatonin receptor (G-protein-coupled receptors); CaM, calmodulin; CRT,
calreticulin; APP, amyloid b precursor protein; 5-LOX, 5-lipoxygenase; COX-2, cyclooxygenase; iNOS, inducible nitric oxide synthase;
PLA2, phospholipase A2.

170
AlzheimerÕs disease and melatonin

many hypotheses regarding this major cause of dementia. amino acids may suffer consecutive cleavage events. The
The three major hypotheses as well as their derivatives will large extramembranous N-terminal region may undergo
be the common thread throughout this review. What has proteolysis by the a-secretase that cleaves the molecule
been published regarding melatonin and its potential role in between Lys687 and Leu688, releasing a large (105–
each proposed mechanism will be added herein, where 125 kDa), soluble ectodomain known as sAPPa [108]. This
appropriate. sAPPa carries a portion of the Ab sequence [109] that
normally includes 28 amino acids of the extracellular and
12–15 residues of the membrane-spanning region of APP;
Pathogeny of AD
thus, the subsequent formation of amyloidogenic peptides
There is a failure of the intercommunication between may be precluded. This a-secretase may be modulated by
neuronal circuits in AlzheimerÕs disease resulting from metal ions and metalloprotease inhibitors and three related
synaptic loss and the destruction of neurons. As a conse- disintegrin and metalloprotease enzymes, ADAM9 [110],
quence, working memory is not transferred through the ADAM10 [108], and ADAM17 [111], which seem to exert
hippocampus to long-term memory circuits. The discon- an a-secretase activity. It has been speculated that the
nection progresses over time and affects some other activation of these proteases, representing a nonamyloido-
functions in addition to memory, so that behavior, exec- genic pathway, may offer a therapeutic method in AD [112,
utive functioning, judgment, movement coordination, and 113]. As a matter of fact, it is remarkable that levels of a-
pattern recognition may become eventually affected. secretase ADAM-10 and sAPPa are reduced in the CSF of
Three major hypotheses have been primarily explored in patients with AD compared to that of controls [114].
an attempt to explain AD: (i) cholinergic hypothesis, (ii) The amyloidogenic pathway is established by the con-
amyloid cascade hypothesis, and (iii) mitochondrial cascade certed action of two secretases, the b-secretase, which
hypothesis. Even though the amyloid cascade is the most cleaves the APP-N terminus, and the c-secretase, which
extended hypothesis [98], the pathogenic role for Ab is cleaves the APP-C terminus in the secondary transmem-
under debate because of reports showing a poor relation- brane region. b-secretase is an aspartyl protease of 501
ship between Ab accumulation and cell death in the brain, amino acids with two aspartic protease active site motifs,
in addition to other results demonstrating a weak correla- which is known as the b-site APP cleaving enzyme I or
tion between Ab and cognitive decline [99]. Furthermore, BACE-1 [115] and considered a prime drug target for
people with Ab deposits do not necessarily suffer AD [100]. lowering cerebral Ab levels in the treatment for and/or
Even more importantly, there are some published data prevention of AD. It is the initiating and rate-limiting
demonstrating that Ab may be protective in brain disease enzyme in Ab generation. BACE-1 activity on APP is
[101, 102]. Thus, the pathogenic role of Ab in AD deserves related to the accessibility of APP within a lipid raft zone of
further scrutiny because all of the hypotheses mentioned the membrane [116, 117]. Once APP escapes from process-
include a pathological role of Ab in AD. ing at the a-site, it is cleaved at the luminal domain,
resulting in a 12 COOH-terminal fragment (C99), which
remains membrane bound, and the soluble APPb NH2-
Amyloid-b processing terminal fragment (sAPPb) [118, 119]. Then, C99 is cleaved
Although far from conclusive, Ab is the most studied factor from the membrane by the c-secretase, a multisubunit
related to pathogenic mechanisms of AD. Ab is derived protease complex composed of a presenilin catalytic sub-
from the catalytic cleavage of an integral membrane unit in addition to nicastrin, the anterior pharynx-defective
protein, the APP, a ubiquitously expressed type I trans- 1 (APH-1), and the presenilin enhancer 2 (PEN-2) [120,
membrane protein whose primary function is not well 121]. Binding of cholesterol to C99 appears to favor the
known. This is the first obstacle in AlzheimerÕs research, amyloidogenic pathway in cells by promoting localization
actually. It is known that the lack of APP in hippocampal of C99 in lipid rafts [122]. The resultant peptide of 39–43
neurons enhances neuritic growth, which influences only amino acid residues, Ab, is delivered to the extracellular
the synapse number at an early age but not in adult animals milieu where it forms insoluble aggregates and becomes the
[103]. The conserved APP intracellular domain, genetically major component of senile plaques. Ab1–40 and Ab1–42 are
uncoupled from APP processing and Ab pathogenesis, has the most common Ab isoforms. Aberrant Ab1–42 accumu-
a key role in survival, proper high-affinity choline trans- lation within distal neurites and synapses is directly
porter (ChT) targeting, and neuromuscular synapse devel- associated with subcellular pathology and neurotransmit-
opment [104]. ChT is responsible for choline uptake from ters [123–125], while Ab1–40 is the predominant form of the
the synaptic cleft, a rate-limiting step in acetylcholine Ab peptides but less prone to form fibrils [126]. As a
synthesis [105]. An extensive review [106] tackles in detail consequence of re-internalization from the extracellular
the possible role of APP in synaptic transmission and space [127, 128] or directly by the cleavage of APP in
neural plasticity, with its respective implications for learn- endosomes generated from the endoplasmic reticulum (ER)
ing and memory. or the Golgi apparatus, Ab peptide accumulation also
Closely associated with lipid rafts in membranes, com- occurs inside neurons leading to trafficking problems, early
posed mostly of sphingolipids and cholesterol, the enzymes axonopathy, synaptic loss, and neuron death [129–131].
in charge of APP cleavage are proteases generating soluble What determines which enzyme will gain access to APP,
isoforms of membrane proteins, a process first related to thus determining the course of events? There are clues
the secretion of angiotensin-converting enzyme in the indicating that cholesterol in lipid rafts directly binds the
kidney [107]. Thus, the APP chain of 695, 751, or 770 C-terminal transmembrane domain of APP, and this

171
Rosales-Corral et al.

Fig. 5. Several routes to prevent the formation of Ab neurotoxic aggregates are used by melatonin. It directly intervenes, affecting the
stability of amyloid b-sheets by disrupting Asp) -His+ salt bridges or affecting the synthesis and maturation of APP, where its ability to
suppress cAMP activity may have a role, because of the cAMP-responsive regions on the APP promoter gene. However, its indirect actions
are significant as well because melatonin may reduce the activity of GSK3 required for the amyloidogenic APP processing, by activating
and/or enhancing the activity of PKC, or by inducing Akt. Both PKC and Akt may turn off GSK-3 through phosphorylation. COX-2,
related to APP synthesis in astrocytes, is controlled by melatonin and its metabolites. Finally, melatonin has a key role in cholesterol and
fatty acid distribution in membranes, as reviewed later. This is important because, as illustrated, amyloidogenic APP processing seems to be
favored by cholesterol/sphingomyelin-enriched lipid rafts. APP, amyloid precursor protein; bs, b secretase; cs, c-secretase; Ab, amyloid b;
MT, melatonin receptors; PLC, phospholipase C; DAG, diacyl-glycerol; PKC, protein kinase C; PI3K, phosphatidylinositol-3-kinase; Akt,
a serine/threonine protein kinase; PKA, cAMP-dependent protein kinase.

interaction may be a determinant in favor of the amyloi- nonamyloidogenic secreted form of APP [138]. Impor-
dogenic pathway [122] (Fig. 5). Cholesterol decreases the tantly, this insulin–PI3K pathway locates and halts glyco-
secretion of soluble amyloid precursor protein (sAPP) by gen synthase kinase-3 (GSK-3) to promote glucose storage
interfering with APP maturation and inhibiting glycosyla- as glycogen, as part of intermediary metabolism. However,
tion in the protein secretory pathway, in such a manner that GSK-3 plays another role. Its a-isoform may interact
APP cannot be cleaved by a-secretase [132]. Processing APP directly with presenilins within the c-secretase complex, and
at the b-site also requires proper orientation to be accessed it is required for the amyloidogenic APP processing. This is
by BACE-1 [133], which in turn localizes largely within the reason why GSK-3 has become a target for the
cholesterol-rich lipid rafts [117, 134]. It is proposed that treatment for AD [139]. Insulin deficiency in brain leads
APP is actually a cholesterol sensor [117]. The AD brain to enhancement of GSK3a/b activation, increases cerebral
shows significant cholesterol retention and high b- and c- amyloidosis, and exacerbates behavioral deficits, as dem-
secretase activities as compared to age-matched nonde- onstrated in APP/PS1 transgenic mouse model of AD by
mented controls, while cholesterol depletion may be asso- impairing insulin downstream GSK3 and JNK pathways
ciated with reduced cellular cholesterol, b-secretase activity, [140].
and Ab secretion [116]. Not only fibrillar Ab but a variety of Ab oligomers may
Another factor influencing the access to APP in cell cause cellular damage. Soluble oligomers, referred to as
membranes seems to be insulin, a significant association amorphous aggregates, micelles, protofibrils, prefibrillar
that links AD to diabetes mellitus. Insulin accelerates APP aggregates, amyloid b-derived diffusible legends (ADDLs),
trafficking from the trans-Golgi network to the plasma Ab*56, globulomers, amylospheroids, toxic soluble Ab,
membrane [135], while the insulin-degrading enzyme (IDE) ÔparanucleiÕ, and annular protofibrils [141], appear within
degrades not only insulin but also Ab and the intracellular neuronal processes and synapses rather than within the
domain of APP [135, 136]. Thus, insulin reduces intracel- extracellular space. They are neurotoxic rather than amy-
lular levels of amyloid and increases amyloid secretion in a loid fibrils found in amyloid plaques [142] and may inhibit
process that probably involves the activation of the MAPK critical neuronal functions including long-term potentiation
cascade [137], although it might also use the phosphatidyl [143], a classic experimental paradigm for memory and
inositol 3 kinase (PI3K)-pathway to release sAPP, the synaptic plasticity [143, 144]. Even more, as a consequence

172
AlzheimerÕs disease and melatonin

of inhibition of the proteasome function, soluble oligomers cAMP production, protecting white matter against a
may cause cell death [124]. Extracellular soluble Ab species, neonatal excitotoxic challenge. This neuroprotective effect
on the other hand, are deposited around neuronal cell may be prevented by luzindole, a well-known membrane
bodies and may interact with the lipid bilayer within melatonin receptor antagonist, or by forskolin, an adenyl-
dendritic arbors at discrete points, appearing co-localized ate cyclase activator [158]. Thus, in a receptor-mediated
with the postsynaptic density protein 95 (PSD-95) [145], manner and by inhibiting adenylyl cyclase, melatonin may
which is related to synapse stabilization and plasticity [146]. impair cAMP signaling, which is probably involved in the
activation of the APP gene promoter. Therefore, melatonin
could interfere with APP synthesis (Fig. 5).
MelatoninÕs role on amyloid-b processing Acting through its MT2 receptor, melatonin stimulates
There are some clues indicating an interaction between phospholipase C (PLC) and, via diacylglycerol (DAG),
melatonin and Ab. By using a thioflavin T (Th T) activates protein kinase C (PKC) [159], which in turn
fluorescence assay, which measures the binding abilities of phosphorylates and inactivates GSK-3, whose participation
different compounds with Ab, it is possible to demonstrate in APP synthesis is key, as mentioned before. However,
that melatonin directly interacts with Ab and prevents its PKC is also capable to directly promote a-secretase-
aggregation [147]. This fact has been well known since 1997, mediated cleavage of APP favoring the nonamyloidogenic
when it was documented using circular dichroism, electron pathway [160]. Thus, by activating PKC, melatonin might
microscopy, and nuclear magnetic resonance spectroscopy impair Ab overproduction (Fig. 5). Moreover, acting
[148]. This phenomenon is not related to the antioxidant through its membrane receptors, melatonin uses a PI3K-
properties of melatonin [149] and involves the disruption of dependent pathway to activate Akt, a serine/threonine
the His+-Asp) salt bridges in Ab peptide, which are protein kinase, which, besides participating in multiple
determinants for the formation and stabilization of b-sheet survival pathways, phosphorylates and inactivates GSK-3
structures [150]. Thus, 24 hr after the incubation with [161]. PI3K/Akt is the same pathway employed by insulin
melatonin, Pappolla et al. [148] showed that original and by the insulin growth factor-1 receptor (IGF-1R) to
b-sheet content of Ab was significantly diminished in interrupt GSK-3b activity under oxidative conditions [162]
opposition to the increase in b-sheet content when Ab (Fig. 5).
was incubated alone (Fig. 5). Because the JNK pathway also could be involved in
A direct interaction between the 5-methoxy group on GSK-3 activation [140], we speculate that melatonin, which
melatonin and His-13 of Ab may occur as well. This prevents JNK activation under oxidative stress conditions
eventuality may be attributed to the higher binding energies [163], may also employ this mechanism to prevent the
in the 5-methoxyindole group, according to single-point activation of GSK-3. It is also feasible that melatonin, in its
energy calculations [151]. Further investigations by electro- role as antioxidant, might enhance PKC anti-GSK3 activity
spray ionization mass spectrometry (ESI-MS), the hydro- by avoiding PKC inactivation. This may occur because
phobic nature of Ab, and melatonin interaction has been PKC is redox sensitive and may be S-glutathiolated and
unveiled, and the proteolytic assessment suggests that the inactivated during oxidative stress in the brain. Oxidative
interaction takes place on the 29–40 Ab-peptide segment stress is a well-documented phenomenon in AlzheimerÕs
[152]. As compared with some other antiamyloidogenic [164].
agents, such as daunomycin or the melatonin analogue 3- Stopping GSK-3 activity could be important not only in
indolepropionic acid, melatonin exhibits a moderate degree interrupting APP synthesis but also to reduce tau hyper-
of inhibition of aggregation, as evaluated by ESI-MS [153]. phosphorylation, because GSK-3 phosphorylates tau [165,
It is also possible that melatonin could regulate the 166]. It is worth remembering that the microtubule-associ-
synthesis and full maturation of APP, as it was demon- ated protein tau is the other key protagonist in AD
strated in melatonin-treated PC12 cells, which responded pathology, being responsible, once hyperphosphorylated,
by decreasing its mRNA encoding b-APP. According to for paired helical filament (PHF) formation.
Lahiri and Song [154, 155], melatonin accomplishes this An indirect interaction between melatonin and Ab
while potentiating the nerve growth factor–mediated processing has been also proposed involving the hypoxia-
differentiation. inducible factor-1 (HIF-1), which upregulates BACE-1,
Because APP gene promoter contains c-AMP-responsive facilitating the generation of cytotoxic Ab peptide [167]. In
regions, it is possible that c-AMP signaling pathways may fact, at a pharmacological dose, melatonin may prevent the
induce APP synthesis, and this eventuality could be a link generation of Ab peptides by reducing both the BACE-1
between neuroinflammation and neurodegeneration, as protein and its mRNA, as demonstrated in a rat model
explored by Lee et al. [156]. They found that prostaglandins employed to evaluate the effect of chronic intermittent
produced by brain injury or inflammation increases cAMP hypoxia (CIH) on the Ab generation in the hippocampus.
formation and stimulates overexpression of APP mRNA Being a redox-sensitive transcription factor, HIF-1 is
and holoprotein in primary cultures of cortical astrocytes. susceptible to melatonin redox modulation [164, 168] and,
On the other hand, the relationship between melatonin or via this indirect manner, melatonin might prevent the
its metabolites and neuroinflammation relies importantly formation of Ab. HIF-1 has been observed to be abundant
on its ability to inhibit prostaglandins by interfering with in AD microvessels where it regulates proinflammatory
the COX-2/PGE pathway [157], which implies the partic- gene expression [169]. On the contrary, it is also known that
ipation of cAMP as a second messenger. In fact, acting the accumulation of Ab by using a nonhypoxic mechanism
through its membrane receptors, melatonin may block may induce the accumulation and nuclear translocation of

173
Rosales-Corral et al.

HIF-1, which in turn mediates a neuroprotective response, [140]. Years later by damaging cholinergic input to the
presumably by regulating glucose metabolism [170]. HIF-1 neocortex or hippocampus from the basal forebrain, it was
has a half-life of approximately 5 min in normoxic condi- possible to reproduce a memory impairment as observed in
tions and less than a minute under hypoxic conditions. AD [141]. Based on this old hypothesis, increasing the
Thus, the role of melatonin in these HIF-1 dependent synaptic levels of acetylcholine (Ach) with the use of
mechanisms is currently only a matter of speculation. acetylcholinesterase (AchE) inhibitors has been employed
Conformational changes in Ab occur in minutes after as a treatment and is considered the standard of care for the
addition of melatonin. In fact, the ability of melatonin to treatment for mild-to-moderate AD; meanwhile, the search
induce conformational changes in Ab has been used to for new AchE inhibitors continues [185]. However,
investigate the conformation and topology of Ab peptides although it remains as a rational approach, nowadays the
interacting with peptide-tethered planar lipid bilayers [171, real efficacy of this treatment is under debate [142].
172]. Similarly, it has been also demonstrated that lipid Both Ab and oxidative stress may reduce Ach synthesis
composition of membrane bilayers plays a dominant role in by reducing choline acetyltransferase activity [186, 187].
mediating conformational changes and in AD pathogeny, However, acetylcholine depletion in the AD brain is also
as reviewed below. related to free cytosolic ionic calcium and oxidative stress.
Levels of Ab aggregates in the brain were reduced by It is well known that Ab induces elevations of intracellular
melatonin in aging mice [173], and in 8-month-old APP 695 free Ca2+ by increasing calcium entry through L-type
transgenic [174] mice or in APP + PS1 double-transgenic voltage-dependent calcium channels [188], and AchE
mice. The latter were supplemented with melatonin from 2 release is a Ca2+-dependent phenomenon [189]. In this
to 2.5 months to age 7.5 months [20]. However, in old, manner, Ab may elevate AChE activity, as demonstrated in
amyloid plaque–bearing Tg2576 mice, which started mel- P19 cells [190]. Furthermore, oxidative stress, which is a key
atonin treatment as late as 14 months of age (5 months protagonist in AD, also plays a role in the enhancement of
later from the onset of the pathology [175]), melatonin acetylcholinesterase activity induced by Ab peptide [191].
failed to reproduce its antiamyloid effects (it seems to even
fail to prevent oxidative stress [176]).
The melatonin/Ab interaction could be an inconvenience
MelatoninÕs role on the cholinergic
hypothesis
according to the Ôbioflocculant hypothesisÕ. Even though
the investigation related to inhibitors of Ab aggregation as The Ab-induced AchE activity may be significantly reduced
a real promise for many investigators [177], blocking or by melatonin, protecting mice from Ab-induced acetylcho-
inhibiting Ab could be a mistake owing to the soluble forms line degradation [147]. Even in LPS injected mice, which
of this peptide, according to this hypothesis. This is because exhibit AchE overactivity, melatonin inhibits this effect as
Ab could have a primordial function: binding to unwanted demonstrated in the neocortical and hippocampal regions
solutes in the extracellular fluid, which then precipitates to in vivo [192].
build deposits or aggregates. Thus, Ab plaques would be an Ab selectively interacts with the potentially neurotoxic
efficient means of presenting neurotoxins to phagocytes NMDA receptor via a postsynaptic site [193], leading to
[178]. dysregulation of Ca2+, which is explained because an
Extracellular Ab may suppress synaptic plasticity or intense stimulation of NMDA-type glutamate receptors
inhibit long-term potentiation (LTP) [179]. There is, in fact, results in a sustained elevation of cytosolic free calcium
an odds ratio homocysteine/AD of 4.5 for histopatholog- ([Ca2+]c) and its consequential dysregulation [194]. The
ically confirmed AD in a case–control study [180], which mechanism of AchE control may be related to the stabil-
also demonstrated that patients with AD and high homo- ization of Ca2+ levels, because it seems that the increase in
cysteine levels showed a more rapid progression over the AChE expression around amyloid plaques is a consequence
following 3 yr. Even more so, high homocysteine levels, of a disturbance in calcium homeostasis; in fact, intracel-
considered a risk factor for dementia and AD [29], are lular calcium mobilization upregulates AChE expression by
related to apoptosis [181]. This methionine derivative has modulating promoter activity and mRNA stability and, on
the ability to induce proapoptotic caspases (caspase 3 and the contrary, chelation of intracellular Ca2+ may inhibit
caspase 9), DNA fragmentation, and the Bcl-2–associated acetylcholinesterase expression [195]. Thus, by controlling
X protein (Bax) while reducing the antiapoptotic Bcl-2 [Ca2+]c, it is possible to control AchE activity. Melatonin
protein; these effects may be inhibited in the presence of controls Ca2+ influx through different pathways, and by
melatonin [182]. these means, it could control acetylcholinesterase expres-
sion as reviewed below. Hence, melatonin is important as
an acetylcholinesterase and butyrylcholinesterase inhibitor,
Cholinergic hypothesis especially the cyclic 3-hydroxymelatonin analogues, which
The cholinergic hypothesis asserts that degeneration of exhibit structural similarity to cholinesterase inhibitor
cholinergic neurons in the basal forebrain and the associ- drugs [196, 197] (Fig. 6). Melatonin, like insulin and the
ated loss of cholinergic neurotransmission in the cerebral anticholinesterase drug, donepezil, exhibits an antiamnesic
cortex cause deterioration in cognitive function as seen in effect in amnesic mice mediated by enhancement of
patients with AD [183, 184]. This theory was introduced in cholinergic activity at the expense of decreasing AChE
1971 when it was demonstrated that cholinergic synapses activity [198].
were modified as a result of learning and that loss of Considering that oxidative stress and particularly perox-
sensitivity to acetylcholine was related to forgetfulness ynitrite (ONOO)) overproduction is significant in AD

174
AlzheimerÕs disease and melatonin

levels as observed in the transgenic animals [174]. It is


possible to find ChAT oxidatively modified by the lipid
peroxidation product, 4-hydroxy-2-nonenal (HNE) [203], a
diffusible electrophile that covalently binds to proteins via
Michael addition to Cys, His, and Lys residues [164, 204,
205]. Thus, owing to its free radical-scavenging activity as
well as its indirect antioxidant actions, melatonin may
reduce ChAT nitrosylation and/or oxidation [186] (Fig. 6).
However, in rats infused intracerebroventricularly with
amyloid-beta for 14 days, where ChAT activity was signif-
icantly reduced, melatonin was unable to restore the
activity of this enzyme [205].

Oxidative stress and neuroinflammation in


the pathology of AD
It is well known that the accumulation of Ab in plaques as
well as Ab oligomers may produce sequential inflamma-
tory/oxidative events and excitotoxicity, causing neurode-
generation and cognitive impairment [206]. In one way or
another, all the proposed mechanisms for explaining AD
pathogenic mechanisms are connected to oxidative stress
and neuroinflammation, widely known hallmarks not only
for AD but in general for all neurodegenerative diseases
and obviously linked to the amyloid cascade [207–209].
Metabolic signs of oxidative stress in AD are always
evident in neocortex and hippocampus, related to altera-
Fig. 6. Melatonin may have a role as an acetylcholine enhancer by
tions in synaptic density. In response to elevated brain
blocking the Ca2+-dependent release of anticholinesterase enzyme peroxide metabolism, AD brains show increased cerebral
(red cross #1) or allowing the proper reentry of choline (red cross glucose-6-phosphate dehydrogenase activity [210], which is
#2) by avoiding ChT oxidation. It is possible that melatonin re- the first and rate-limiting enzyme of the pentose phosphate
stores ChAT activity under oxidative stress conditions (red cross pathway, central to maintenance of the cytosolic pool of
#3) as observed in APP transgenic mice. However, the role of NADPH and thus the cellular redox balance. Even the
melatonin in cholinergic hypothesis remains to be clarified because
brains of preclinical AD individuals, with normal antemor-
oxidative stress along with calcium dysregulation, as observed in
chronic cellular stress, is relevant in acetylcholine expression and its tem neuropsychological test scores but abundant AD
metabolism as well as Ach receptor activity (red cross #4). Mit, pathology at autopsy, may exhibit increased levels of the
mitochondria; ER, endoplasmic reticulum; ChAT, choline acetyl major product of lipid peroxidation, 4-hydroxynonenal,
transferase; Ach, acetylcholine; ChT, choline transporter; AchE, and acrolein, a powerful marker of oxidative damage to
acetylcholinesterase; AchR, acetylcholine receptor. protein [211]. Inferior parietal lobule samples from early
AD patients compared to age-matched controls have been
brain, where the latter mediates neurotoxicity of Ab [199], it examined for proteomic identification of nitrated brain
is possible that S-nitrosylation of metabolic Ach interme- proteins that revealed significant alterations in antioxidant
diaries has a significant role. Among the multiple targets of defense proteins and energy metabolism enzymes, with all
OONO) in AD brain, choline acetyltransferase (ChAT) of them being directly or indirectly linked to AD pathology
may be a good candidate. ChAT, whose activity has been [212].
shown to decrease in the AD brain [200], may undergo Ab neurotoxic properties depend heavily on free radicals.
S-nitrosylation followed by lysis and oligomerization, as The overproduction of free radicals in the pathogeny of AD
demonstrated in cholinergic nerve endings and synaptic may come from the microglial respiratory burst in response
vesicles from Torpedo marmorata electroneurons [186]. to Ab-induced neuroinflammatory events [213–216]. The
Even though the mechanism is not fully understood, the microglial respiratory burst in AD may result from (i) the
high-affinity ChT, which provides choline for acetylcholine interaction of Ab with specialized receptors, (ii) the
synthesis in neurons, seems to be regulated by OONO) as astrocyte/microglia intercommunication, or (iii) detection
well [201]. Thus, we speculate that melatonin may promote of damage-associated molecular patterns (DAMPs)
choline transport [200, 202] (Fig. 6). through their corresponding receptors, leading to the
There are some other factors also involving melatonin in activation of the phagocytic nicotinamide adenine dinucle-
the cholinergic hypothesis even though the mechanism is otide phosphate (NADPH)-oxidase (PHOX). The activa-
not fully understood. For example, ChAT, which binds tion of the NADPH oxidase probably both in neurons and
acetyl coenzyme A to choline in Ach synthesis, has been in glia [217–219] links redox control and neuroinflamma-
found decreased in the frontal cortex and hippocampus of tory signaling pathways [220].
APP 695 transgenic mouse model of AD, and melatonin, Ab causes microglial proliferation mediated by PHOX,
chronically administered for 4 months, restored ChAT which is demonstrated by a marked translocation of the

175
Rosales-Corral et al.

cytosolic factors p47phox and p67phox to the microglial PHOX complex, a crucial step where SOD plays a key role
membrane in brains of patients with AD, and this is acting as a stabilizer of Rac [225]. Once integrated, PHOX
correlated with proinflammatory events, such as TNF-a transfers electrons from NADPH to molecular oxygen
and IL-1b overproduction [221, 222]. The synergy between generating O2 .
oxidative and nitrosative stress plus neuroinflammation Because Ab induces oxidative stress that is related to
may increase the overproduction of OONO) by a mitochondrial damage, a mechanism closely linked to
1,000,000-fold [223]. PHOX is a multicomponent enzyme apoptosis is established [226–229]. Reciprocally, oxidative
system composed of two integral membrane proteins, stress may induce intracellular accumulation of Ab,
p22phox and gp91phox, integrated as cytochrome b558, enhancing the amyloidogenic pathway [226, 230, 231]
three essential cytosolic components, p47phox, p67phox, (Fig. 7).
p40phox, and the above-mentioned GTPase Rac1, of the Additionally, Ab1–42 may initiate free radical chain
Rho family of small G proteins. In general terms, the reactions by itself. It has a critical methionine residue at
complex begins its integration when the cytosolic p47phox position 35, which is highly hydrophobic and possesses a
subunit becomes phosphorylated and transports the total sulfur atom sensitive to oxidation (:S: fi O=S: fi
cytosolic components to the docking site where they O=S=O) [231], or if the lone pair of electrons on the S
assemble to the flavocytochrome b558 (reviewed in [224]). atom undergoes one-electron oxidation, it produces a
GTP-bound Rac coordinates the translocation of the positively charged sulfuranyl radical (MetS+) [232]
p47phox/p67phox/p40phox complex and its dissociation (Fig. 7). In this manner, S–O bonded MetS+ may initiate
from GTP permits the subsequent inactivation of the free radical chain reactions with allylic H atoms on

Fig. 7. Ab plaques and oligomers are in the middle of a complex set of interactions among astrocytes, microglia, and neurons originating a
neuroinflammatory response. This is linked to reactive oxygen species (ROS) and NOS overproduction (gray clouds) culminating in
oxidative stress, which in turn feeds back on neuroinflammation. Organelle dysfunction, particularly mitochondria, adds more free radicals
and aggravates the situation. Even worse, oxidative stress and Ab are interdependent phenomena; thus, the more the oxidation, the
more the amyloid accumulation. Newly formed Ab contributes to more neuroinflammation and oxidative stress, closing the vicious cycle. In
fact, Ab can be an oxidant by itself, as shown. During inflammatory and oxidative stress, communication between cellular protagonists is
importantly mediated by calcium waves (blue waves) apart from cytokines. Melatonin (green) and its major metabolites AFMK and
AMK play key roles by scavenging free radicals directly, while they enhance endogenous antioxidant systems, as shown in Fig. 3. AMK is
relevant particularly in mitochondria, where it takes ETC components as electron donors or acceptors. Going further, melatonin and its
metabolites have a role in neuroinflammation by regulating both proinflammatory signals and oxidative stress mediators, such as COX2
and iNOS by avoiding NF-jB full integration. ctk, cytokines; Ab, amyloid-beta; AFMK, N1-acetyl-N2-formyl-5-methoxykynuramine;
AMK, N1-acetyl-5-methoxykynuramine; 3-OHM, cyclic 3-hydroxymelatonin; ETC, electron transport chain; NF-jB, nuclear factor kappa
B; COX-2, cyclooxygenase 2; iNOS, inducible nitric oxide synthase; PGE2, prostaglandin E2.

176
AlzheimerÕs disease and melatonin

unsaturated acyl chains of lipids making the lipid hydro- 248]. ROS, in turn, may come from the innate immune
peroxide and propagating the chain reaction [233]. response promoted by danger signals [249, 250] or from the
Ab can also directly trap molecular oxygen, reducing it to damaged mitochondria [251, 252].
H2O2 in the presence of iron (Fenton reaction), as it has Ab peptides may activate microglia through (i) Toll-like
been demonstrated by spectrochemistry in AD brain [234]. receptors 2 (TLR2), (ii) scavenger receptor (SR), (iii)
Fe2+ ions are generated via a redox cycling of iron (Fe2+ receptor for advanced glycation end products (RAGE),
M Fe3+), and in the presence of a metal chelator, such as (iv) a cell surface receptor complex, and (v) TNFR1, whose
clioquinol, this Ab neurotoxicity is reduced [235]. The deletion, as observed in APP23 transgenic mice (APP23/
matter is relevant because significant alterations in Cu, Zn, TNFR1()/))), may inhibit Ab generation and diminishes Ab
and Fe have been found in AD brain in those areas showing plaque formation in the brain [253]. Ab aggregates as
severe histopathologic alterations [236, 237]. In general, foreign protein particles are recognized by TLRs, and these
drugs that prevent oxidative stress include antioxidants, become important Ab innate immune receptors, as dem-
modifiers of the enzymes involved in ROS generation and onstrated in antisense knockdown of TLR2 or using
metabolism, metal-chelating agents and agents, such as functional blocking antibodies against TLR2, which may
anti-inflammatory drugs, that can remove the stimulus for suppress Ab-induced expression of proinflammatory mol-
ROS generation. ecules and integrin markers in microglia [254]. Even TLR4
H2O2 is a well-known uncoupler of the mitochondrial could also play a role, as demonstrated in mouse models
respiratory activity, producing a concentration-dependent homozygous for a destructive mutation of TLR4; these
inhibition of state 3 (ADP-stimulated) respiration and show significant increases in diffuse and fibrillar Ab
reducing substantially the ADP:O ratio [238]. An evalua- deposits [255]. However, it is not clear whether TLR
tion of electron transport chain complexes and Krebs cycle signaling pathways involve the clearance of Ab deposits in
enzymes revealed that a-ketoglutarate dehydrogenase, suc- the brain or they initiate a neuroinflammatory response,
cinate dehydrogenase, and aconitase are susceptible to responsible for the synaptic impairment observed in AD
H2O2 inactivation, which is a reversible process [239]. pathology [256]. Important receptors, such as the class B
Under normal conditions, excessive ROS are neutralized scavenger receptor CD36 and the LPS-binding molecule
by the action of endogenous and exogenous antioxidant CD14, signal through TLR2. CD36 recognizes a variety of
defense systems. In addition to the above-mentioned ischemic by-products acting as ligands, including oxidized
oxidant-generating properties, Ab may bind the peroxidase low-density lipoprotein (LDL), long-chain fatty acids,
enzyme, CAT with high affinity, inhibiting H2O2 break- thrombospondin-1, and, again, Ab. In microglia and in
down [240] and thus worsening redox conditions. However, other tissue macrophages, Ab initiates a CD36-dependent
all the antioxidant mechanisms play roles in the AD brain. signaling cascade involving the Src kinase family members,
Thus, the overexpression of superoxide dismutase-2 (SOD- Lyn and Fyn, as well as the mitogen-activated protein
2), which is localized to mitochondria, scavenges hippo- kinase, p44/42. Ab also causes the blockade of Src kinases
campal superoxide and prevents memory deficits in Tg2576 downstream of CD36 and inhibits macrophage inflamma-
AD mice [241], which carry both mutant APP and tory responses to b-amyloid [257].
presenilin 1 transgenes [242]. In another AD mouse model, Another scavenger receptor, the macrophage receptor
3xTg-AD, there are significant rises in the activities of SOD with collagenous structure (MARCO) along with the che-
and GPx, compared with the controls, whereas levels of motactic G-protein-coupled receptor formyl-peptide-recep-
reduced GSH are significantly decreased with a concomi- tor-like-1 (FPRL1) has been documented to be essential in
tant rise in oxidized glutathione (GSSG). This set of events the amyloid b-induced signal transduction in glial cells [258].
implicates a high oxidative state and depletion of proton Neurons, microglia, and endothelial cells, which surround
donors [243]. The 3xTg-AD mouse harbors PS1M146 V, the senile plaques in the AD brain, express higher levels of
APPSwe, and tauP301L mutations and progressively develops RAGE, which may trigger oxidative stress and NF-jB
extracellular senile plaques and intracellular neurofibrillary activation [259]. The interaction of Ab with RAGE may be a
tangles (NFTs) as well as cognitive impairments [244]. direct interaction [216], or it may involve damaged molecular
Interestingly, even the exogenous antioxidant systems seem patterns, such as the S100B protein. In primary cortical
to fail in AD, which are apparently not related to under- neurons, the transcription factor Sp1 mediates IL-1b induc-
nourishment because, as demonstrated in 79 patients where tion by S100B without evidence of a role for NF-jB, whereas
the plasma chain-breaking antioxidants a-carotene, in microglia, S100B stimulates NF-jB or AP-1 transcrip-
b-carotene, lycopene, vitamin A, vitamin C, and vitamin tional activity and upregulates Cox-2, IL-1b, IL-6, and TNF-
E were measured by HPLC in addition to a total antiox- a expression through RAGE engagement [260, 261]. Finally,
idant capacity assay, a tool for measuring the inhibitory a cell surface receptor complex for fibrillar Ab, linked to the
effect of antioxidants [245]; all of the measured parameters small GTPase Rac1 and critical in signaling to PHOX, has
were below the normal range. been described. This molecular complex mediates microglial
activation through the stimulation of intracellular tyrosine
kinase–based signaling cascades, and it is integrated by the
Microglia activation and neuroinflammatory
B-class scavenger receptor CD36, the integrin-associated
response
protein/CD47, and the a6b1-integrin [262].
A common factor in AD pathogeny is the overactivation of NF-jB may be activated from a variety of pathways,
microglia with the consequent overexpression of proinflam- from the canonical pathway where the proinflammatory
matory cytokines and a significant increase in ROS [246– TNF-a, IL-1, and LPS exert their action in addition to

177
Rosales-Corral et al.

DAMPS, to the noncanonical pathway where CD40 and lating factor, IL-10, IL-b, and ApoE modulate microglia
lymphotoxin receptors activate a p52/relB complex. More- activity [274–276]. Glial Ca2+ waves can trigger responses
over, there are other atypical pathways, where genotoxic in microglial cells, and the calcium waves arise, in vitro, in
stress, hypoxia, UV light, H2O2, or the epidermal growth response to Ab administration [277]. Extracellular ATP, in
factor receptor 2, among others, may intervene (reviewed in its role as a DAMP and as part of the innate immune
[263]). The link between NF-jB and neurodegenerative receptor surveillance behind the Ab-induced inflammasome
disorders, particularly AD, is an old one [264, 265]. activation [278], may also elicit Ca2+ waves and activate a
In rat primary cultures of microglial cells and human microglial inflammatory response [279]. As will be ex-
neutrophils and monocytes, Ab activates PHOX, and this plained below, melatonin also has a role in this process.
effect may be potentiated by the proinflammatory stimulus, Even though the underlying mechanisms and their scopes
such as interferon-gamma or TNF-a, but blocked by in neuroinflammation remain to be unveiled, it has been
tyrosine kinase inhibitors [266]. Mediated by PHOX, suggested that melatonin could have modulatory effects on
oligomeric Ab may induce ROS production, possibly ATP-dependent gliotransmission or glial calcium waves
through N-methyl-D-aspartate receptors (NMDAR), and derived from different brain regions and species, regulating
these PHOX-related ROS, in turn, release the prostanoid astroglial function [280]. Studied in the context of rhythmic
precursor arachidonic acid through the activation of ERKs, circadian outputs to pervasive neurobehavioral states, a
which phosphorylate cytosolic phospholipase A2a [219]. functional shift in the mode of intercellular communication
The rate-limiting enzyme, COX-2, can be induced by between junctional coupling and calcium waves in glial cells
multiple cellular factors such as growth factors or the was found to be induced by melatonin [281]. However, it is
proinflammatory cytokines IL-1b and TNF-a in neurons, well known that melatonin modulates intracellular free
astrocytes, and microglia. COX-2 in turn regulates PGE2 Ca2+, and by this means, melatonin may protect cells from
signaling in neurons [267] and can activate APP transcrip- calcium-dependent pathways of death, such as calpain and
tion in astrocytes [156], as well as glutamate release from caspase-3 in cells undergoing excitotoxicity and oxidative
astrocytes, which is responsible for excitotoxic damage in stress, as demonstrated in vitro in rat C6 astroglial cells
AD [193, 268, 269]. PGE2 and COX-2 feedback each other [282]. By controlling Ca2+ influx, melatonin attenuates
and modulate neuroinflammation, regulating the produc- glutamate-mediated excitotoxicity, which is responsible for
tion of multiple inflammatory molecules. NMDAR-mediated damage of neurons. This in turn is one
of the postulated Ab-mechanisms of damage, as mentioned
above [219]. In in vitro experiments with a hippocampal cell
MelatoninÕs role in neuroinflammation and line challenged with H2O2, Ab, or glutamate, cell death was
oxidative stress
prevented by the melatonin derivative, AFMK, which is
Importantly, melatonin has a key role in Ab-induced formed by the interaction of melatonin with H2O2 or O_2
assembly of PHOX and the subsequent production of [76] (Fig. 7). Additionally, melatonin may inhibit not only
ROS, as demonstrated in cultures of microglia incubated in glutamate-induced ion currents but also ion currents from
the presence of fibrillar Ab. According to Zhou et al. [270], the other ionotropic glutamate receptors, kainate, and
melatonin may impair the assembly of PHOX by inhibiting AMPA [283]. Also, it is possible that melatonin antagonizes
the translocation of p47phox and p67phox subunits of glutamate release, as observed in cortical synaptosomes in
PHOX from the cytosol to the plasma membrane. This old mice and in neurotoxicity induced by KCl [284].
becomes feasible owing to blockade of the phosphorylation We reported in vivo that melatonin significantly reduced
of p47phox, a PI3K-dependent phenomenon, and conse- the proinflammatory response, decreasing by nearly 50%
quently impairing the binding of p47phox to gp91phox. the Ab-induced levels of proinflammatory cytokines IL1-b,
This mechanism is related to melatoninÕs capacity to inhibit IL6, and TNF-a [285]. We speculated that melatonin
Akt (protein kinase B) activity in microglia, which is the affected NF-jB DNA binding activity based on a previous
Ser/Thr kinase downstream of PI3K in these cells [270]. It is report by Natarajan et al. [286]and Chuang et al. [287],
worth mentioning that the activation of the PI3K/Akt who found that NF-jB DNA binding activity was inhibited
pathway may be mediated by H2O2, acting as an intracel- by melatonin and was lower at night when endogenous
lular messenger [271]; melatonin is known to directly melatonin levels are high. Furthermore, 60 min after an
scavenge H2O2 [234, 239] (Fig. 7). intraperitoneal injection of melatonin, a reduction in NF-
Melatonin directly detoxifies H2O2 and produces the jB DNA binding activity was replicated. More recently, it
biogenic amine AFMK and a potent free radical scavenger, has been demonstrated in Ab-treated brain slices that
which in turn may suffer deformylation, giving rise melatonin reduces NF-jB-induced IL-6 in a concentration-
AMK.This latter antioxidant and free radical scavenger is dependent manner [288]. By administering melatonin, it is
particularly relevant in mitochondria [78, 157, 239] (Fig. 7). also possible to reduce Ab-induced impairment in learning
Melatonin and/or its metabolites function as antioxidants and memory in rats along with a significant decrease in
[91, 272], free radical scavengers, and antiapoptosis agents positive glial cells expressing NF-jB-induced IL-1b in
and prevent abnormal nitric oxide (NO) elevation [273] in addition to C1q in hippocampus [289], both of which are
the cerebral cortex. involved in glial activation (Fig. 7). The critical comple-
Between microglia and astrocytes, a fluid communication ment component C1q, in turn, may induce the translocation
exists. Several astrocyte factors released including trans- of NF-jB p50p50 homodimers, at least as observed in
forming growth factor b (TGF-b), macrophage colony- human monocytes [290], and it is always related to AD
stimulating factor, granulocyte/macrophage colony-stimu- pathology usually linked to fibrillar b-amyloid [291].

178
AlzheimerÕs disease and melatonin

The pleiotropic transcription factor NF-jB, composed of It seems that melatonin may thus regulate neuroinflam-
homo- and heterodimers of five members of the Rel family mation through free radical control and modulation of
including NF-jB1 (p50), NF-jB2 (p52), RelA (p65), RelB, important proinflammatory transcription factors and their
and c-Rel (Rel) plays a key role in inflammatory processes signaling pathways while reducing glutamate excitotoxicity,
but also it is a protagonist in plasticity and neuronal whether it be by inhibiting glutamate-induced ion currents
development (reviewed in [292]). Thus, it is complicated to or by controlling the glutamate delivery. On the other hand,
point out that NF-jB inhibition may be a therapeutic target even though functional cytoplasmic membrane melatonin
in AD. Nonetheless, by using an immunological assay, it is receptors have been described in astrocytes derived from
possible to demonstrate how melatonin prevents the Ab- chick brain [301], which could suggest a role for melatonin
induced expression of NF-jB [147]; more specifically, as a metabolism regulator in astrocytes, these receptors
according to Deng et al. [293], melatonin inhibits p52 have not been corroborated in human glia.
NF-jB binding as demonstrated by examining the expres- The nuclear hormone retinoid z receptor/retinoid orphan
sion of LPS-induced iNOS and COX-2. The latter action receptor (RZR/ROR), from the retinoid-related orphan
involves a promiscuous histone acetyltransferase (HAT), receptors family, are likely associated with melatonin
within the nuclear cofactor p300, which is essential for signaling and have been identified in the promoter region
COX-2 transcriptional activation by proinflammatory of 5-lipoxygenase (5-LOX), a key protagonist in neuroin-
mediators. By inhibiting p300 HAT activity, melatonin flammation. By repressing the expression of 5-LOX mRNA
may suppress p52 acetylation, binding, and transactivation in human B lymphocytes, melatonin may reduce the
[293]. In this manner, it is possible to block the rate-limiting proinflammatory response via nuclear receptor RZR/
enzyme COX-2 (Fig. 7). This enzyme is induced by multiple RORa [302]. Furthermore, the transcriptional activation
cellular factors, such as growth factors or the proinflam- of RZR/RORa by melatonin is possible even in the
matory cytokines IL-1b and TNF-a in neurons, astrocytes, nanomolar range [303, 304]. There are no reports confirm-
and microglia. COX-2 in turn regulates PGE2 signaling in ing this effect of melatonin in the CNS, but 5-LOX is widely
neurons [267] and can activate APP transcription in expressed in the brain [305] where it has neuromodulatory
astrocytes [156], as well as glutamate release from astro- and neuroendocrine functions and plays an important role
cytes, which is responsible for excitotoxic damage in AD in aging and AD, as we will review later. It is worth
[268, 269, 293]. PGE2 and COX-2 feedback on each other mentioning that, in addition to AA-derived leukotrienes,
and modulate neuroinflammation, regulating the produc- 5-LOX also modulates the c-secretase activity in mem-
tion of multiple inflammatory molecules. branes, favoring Ab formation [306].
Experiments using transformed lymphatic-derived endo- Although there is an isolated report indicating that
thelial cell line demonstrated the ability of melatonin to melatonin is not an important modulator of macrophage
prevent TNF-a induced phosphorylation of NF-jB p65, and microglia function [307], melatoninÕs role controlling
although the mechanism is unclear [294]. The administra- the primarily microglia-guided neuroinflammatory
tion of melatonin 1 hr after closed head injury also may response is demonstrated in multiple reports. This is a
inhibit the activation of NF-jB during the late phase consequence of its regulation of the NF-jB overexpression
(8 days), an effect attributed to its prolonged antioxidant [308], the amount of LPS-induced proinflammatory cyto-
effect at the site of injury. However, melatonin did not alter kines [192], or prevention of GSK-3b activation and
early phase (24 hr after closed head injury), which implies a neuroinflammation in response to Ab, as observed in
selective mechanism of neuroprotection [295]. One could astrocytes and microglial cells [166]. Meanwhile, a cumu-
expect that such an interference with NF-jB would also lative dose of 10 mg/kg melatonin may attenuate kainic
affect the role of this transcription factor in plasticity and acid-induced neuronal death, lipid peroxidation, and mi-
neurogenesis. Thus, even though not linked to these NF- croglial activation, reducing the number of DNA breaks in
jB-dependent mechanisms, there are reports indicating a vivo, as demonstrated in adult male Sprague–Dawley rats
significant depression as well as instability of synaptic [309].
transmission in the central nervous system (CNS), The antioxidant and immunomodulatory effects have
although melatonin-dependent fluctuations in synaptic inserted melatonin into the two-hit hypothesis [310], which
potentials were apparent only when the involved circuit states that although either oxidative stress or abnormalities
was tetanized [296]. Such depressive effects of melatonin in in mitotic signaling can, independently, serve as initiators in
synaptic transmission would be expected to influence AD, both processes are necessary to propagate the path-
epileptic seizure activity [297]. Nonetheless, other results ological features of the disease.
indicate that, instead of depressing synaptic transmission,
melatonin modulates neuronal excitability in the hippo-
campus, and this modulatory activity depends on its
The mitochondrial cascade hypothesis
receptors [298, 299]. In fact, melatonin may modulate According to Swerdlow and Khan [311], the mitochondrial
specific forms of plasticity in hippocampal pyramidal cascade hypothesis asserts that inheritance determines
neurons, as demonstrated by electrophysiological methods mitochondrial baseline function and durability, and mito-
[300] where neurons exposed to melatonin were found to chondrial durability influences how mitochondria change
change their excitability in response to repetitive stimula- with age. Thus, according to this hypothesis, once a
tion, which reveals melatonin as an activity-dependent threshold of mitochondrial changes is reached, AD histo-
modulator of subsequent synaptic plasticity (metaplastic- pathology and symptoms ensue. Even though it was
ity; Fig. 4). formulated in 2004 as a formal hypothesis, some strong

179
Rosales-Corral et al.

evidence linking AD to mitochondrial damage in brain of patients with AD where mitochondrial distribution
cells had been reported many years earlier. For example, tends to be predominantly perinuclear and fission or
in 1980, by investigating the mechanism for the production fragmentation prevails over fusion, a phenomenon related
of acetyl-CoA used in acetylcholine synthesis, a small, but to a low metabolic capability [327, 328]. These events
significant, reduction in the activity of the pyruvate involve large dynamin-related GTPases, such as the
dehydrogenase (besides ATP-citrate lyase and acetoace- dynamin-related protein (Drp1). Localized to mitochon-
tyl-CoA thiolase) was found in postmortem brain tissue dria, Drp1 is a key factor in mitochondrial division and
from cases of AD [312]. In 1985, the activity of the particularly sensitive to redox regulation [328]. It has been
pyruvate dehydrogenase complex was reported to be reported that NO overproduced in response to Ab protein
reduced to about 30% of control values in histologically could be responsible for the impairment of Drp1 via
unaffected occipital cortex as well as in histologically S-nitrosylation [329], and this eventuality may lead to an
affected frontal cortex of the brains of patients with AD imbalance of fission/fusion in mitochondria, which in turn
[313]. Likewise, AD as Ôa primary defect in cytochrome is correlated with neuronal damage and synaptic loss
oxidaseÕ was proposed years later [314]. However, it [330].
currently is debatable whether Ab is a downstream Another important feature related to AD pathogeny is
product of the mitochondrial functional decline or whether mtDNA damage. Perhaps because it is not protected by
Ab-induced mitochondrial damage is an extension of the histones, mtDNA with its 37 genes is more susceptible to
amyloid cascade hypothesis. The amyloid cascade, pro- oxidative stress-induced deletions and point mutations
posed 20 yr ago, suggested that faulty metabolism of APP than nuclear DNA. Even though the consequences of
was the initiating event in AD pathogenesis, leading these alterations remain to be clarified [331], mtDNA
subsequently to the aggregation of Ab, specifically Ab1– damage is usually linked to dysfunction (decrement of
42 [315, 316]. However, long before the appearance of mitochondrial electron transport chain efficiency) and
extracellular Ab deposits, they are detectable within apoptosis [332, 333].
mitochondria [317]. Also related to Ab-induced oxidative stress, mitochon-
Beyond hypotheses, some other important features have drial proteins and lipids become disturbed leading to
been found since those early years. It has been demon- dysfunction. We have found significant alterations in
strated that Ab1–42 uncouple the mitochondrial respiratory cholesterol and fatty acids content in mitochondrial mem-
chain, and this event plays a key role in pathology of AD branes following the injection of Ab (Rosales-Corral et al.,
[311]. Structurally, Ab induces swelling of isolated mito- unpublished data), associated with functional impairment;
chondria [318, 319] and, functionally, decreases ATP as a consequence of increased membrane permeability and
synthesis and the activity of various mitochondrial en- changes in lipid polarity owing to oxidative injury,
zymes, as demonstrated in vivo [320] and in vitro in cytochrome c is released from the intermembrane space of
cultured neuronal cells or in Ab-exposed astrocytes [319– mitochondria [334], behaving as an important intermediate
321]. Later, different neurotoxic mechanisms for Ab were in apoptosis and associated with impaired mitochondrial
proposed, including disruption of mitochondrial function respiration, as observed in brain, platelets, and fibroblasts
via binding of the Ab-binding alcohol dehydrogenase of patients with AD [335].
(ABAD) protein [322]or the formation of ion channels An important feature related to the direct interaction
allowing calcium uptake, which induces neuritic abnormal- between Ab and cyclophilin D (CypD) has been found
ity in a dose- and time-dependent manner [323], or the [336]. Ca2+-associated CypD is part of the mitochondrial
opening of the mitochondrial permeability transition pore permeability transition pore (MtPTP) and translocates
coupled to inhibition of respiratory complexes [324, 325]. from the matrix to the inner membrane where it appears
We have found (Rosales-Corral et al., unpublished data) linked to oxidative stress, and by facilitating the opening of
that following the intracerebral injection of fibrillar Ab, the mPTP, CypD causes mitochondrial swelling with cellular
peptide is revealed both intracellularly and intramitoc- and synaptic perturbations [337, 338]. The importance of
hondrially, deep in the cristae, coinciding with other reports the association of Ab/CypD is underlined by the fact that a
which demonstrate that Ab progressively accumulates in deficiency in CypD may attenuate Ab-induced mitochon-
mitochondria where it is associated with diminished enzy- drial oxidative stress, an effect accompanied by improved
matic activity of the respiratory chain complexes III and IV synaptic function and an improved cognitive performance,
[317]. Presence of Ab in mitochondria is related to a as observed in APP transgenic/CypD double-mutant mice
reduction in the rate of oxygen consumption by the electron [336] (Fig. 8).
transport chain [317]. The enzymes in charge of importing As a result of Ab entrance and mitochondrial damage,
Ab to mitochondria have been identified as a complex of energy demands of cells become impaired. We have found
translocases, i.e., translocases of the outer membrane functional disorders of F0F1-ATPase in submitochondrial
(TOM) and the translocase of the inner membrane (TIM) particles obtained from platelets of patients with Alzhei-
[326] (Fig. 8). merÕs-type dementia [339], but the impairment of ion-
In addition to direct effects caused by Ab in mitochon- motive ATPases in response to Ab is reproducible in
dria, there are severe changes attributed to the Ab-induced hippocampal neurons in culture [340]. Nonetheless, another
oxidative stress. A disturbance of mitochondrial dynamics, report on F0F1-ATPase, searching in isolated mitochon-
a term that includes fission, fusion, movement, and dria from platelets and postmortem motor cortex and
mitochondrial architecture, seems to be implicated in hippocampus from patients with AD, did not find abnor-
AD pathogeny. It has been demonstrated in human brains malities in F0F1-ATPase functioning [341].

180
AlzheimerÕs disease and melatonin

Fig. 8. Both Ab and abnormal phosphorylated tau play key roles in mitochondrial dysfunction long before amyloid plaques appear.
Hyperphosphorylated tau may cause ETC dysfunction by impairing complex I activity, although its major capacity for damaging may come
from its ability to interact with ANT from MtPTP, which leads to swelling, mitochondrial dysfunction, and cell death, ultimately. Amyloid-
b, whose mitochondrial receptors (ABAD and Hsp) have been identified, causes ETC dysfunction by interrupting the activity of complex III
and complex IV, and possibly it may disrupt ion-motive ATPase. Moreover, Ab impairs energy metabolism by inhibiting directly the activity
of the a-ketoglutarate enzyme during the tricarboxylic acid cycle and the pyruvate dehydrogenase before the cycle. Ab also disrupts Ca2+
homeostasis, which overloads mitochondrial matrix and may lead to complex II deficiency, membrane potential loss, ATP reduction, and
ROS overproduction in addition to MtPTP disturbance. Ab-induced oxidative stress has been related to membrane dysfunction, oxidation
of ETC components, free radical leak, and MtDNA damage because MtDNA is particularly vulnerable to these events. The way CytC
escapes from mitochondria is not completely clear, but once released, it initiates a chain of events leading to apoptosis. Melatonin
(represented as red crosses) tends to accumulate inside mitochondria, where (1) it may reduce oxidative stress and its deleterious conse-
quences on MtDNA, proteins, and membrane lipids, such as cardiolipin; (2) it strongly inhibits MtPTP currents and prevents cytochrome c
release in a dose-dependent manner; (3) it may recycle electron carriers, such as NADH; (4) it may prevent apoptosis by impairing Cytc
release from mitochondria; (5) Ca2+ regulation by melatonin may protect mitochondrial functioning. ROS/RNS, reactive oxygen/nitrogen
species; Ub, ubiquitin; ABAD, amyloid-b binding alcohol dehydrogenase; Hsp, heat-shock protein; Cytc, cytochrome c; ETC, electron
transport chain; MtPTP, mitochondrial permeability transition pore; MtDNA, mitochondrial DNA; APAF-1, apoptosis-activating factor 1.

levels could be even 100 times higher than melatonin levels


MelatoninÕs role in mitochondrial
in plasma [346]; this claim, however, requires confirmation.
hypothesis
Indole propionamide, similar to melatonin but with a
MelatoninÕs role in this context is mostly related to its longer half-life has been proven to protect against mito-
ability to scavenge free radicals in addition to its indirect chondrial toxins capable of collapsing the mitochondrial
antioxidant properties because it enhances endogenous proton potential, causing severe mitochondrial dysfunction
antioxidant systems in mitochondria [342–348]. It does this and ATP deprivation. Under those circumstances, this
basically by maintaining and regenerating the GSH con- recently discovered endogenous indole may act as a
tent, which is an important antioxidant mechanism in recyclable electron and proton carrier, restoring the proton
mitochondria [346]. Similarly, melatonin reduces peroxida- gradient and mitochondrial ATP synthesis [349].
tion of lipids in mitochondrial membranes and free radical In reference to MtPTP, the antiapoptotic effects of
leakage from this organelle. Thus, it is possible to repro- melatonin have been explained by its ability to inhibit the
duce in vitro mtDNA damage by adding Ab1–42 to neurons opening of the protein channels responsible for calcium and
in culture, but the addition of melatonin prevents signifi- cytochrome c (cyt c) release from mitochondria. Also, it
cantly mtDNA damage [14, 347]. Added to drinking water may function by reducing the loss of the mitochondrial
and chronically administered, melatonin prevents mito- membrane potential in the presence of glucose deprivation-
chondrial impairment, maintaining or even increasing ATP related events [350] (Fig. 8).
production in senescent-prone mice suffering age-dependent Cardiolipin is an important component (20%) of the
mitochondrial dysfunction accompanied by an important inner mitochondria membrane. Being particularly suscep-
oxidative/nitrosative stress [348]. It is worth noting that, tible to oxidative stress, cardiolipin becomes implicated in
additionally, melatonin seems to accumulate in the mito- cyt c release during apoptotic events [351], in part because it
chondria, in such a manner that mitochondrial melatonin sensitizes mitochondria to Ca2+ mPTP [352]. One of the

181
Rosales-Corral et al.

probable mechanisms of mitochondrial protection by neurotransmitter glutamate, may lead to an excessive


melatonin relates to the prevention of cardiolipin oxidation, glutamate activity and consequently excessive influx of
avoiding MtPTP opening and restoring Ca2+ balance (as cations, Ca2+ in particular [268, 269]. Glutamate receptors,
reviewed in [353]). However, it is also possible that especially NMDA, are deeply involved in AD pathology
melatonin directly inhibits MtPTP at single-channel and [370]by controlling Ca2+ influx. Ionic calcium, in turn,
cellular levels, as demonstrated in patch-clamp recordings activates a number of enzymes, including phospholipases,
on the inner mitochondrial membrane [350]. endonucleases, xanthine oxidase, neuronal nitric oxide
Three other important proapoptotic factors related to synthase, as well as proteases, such as the calcium-depen-
mitochondrial functioning and signaling have been dem- dent cysteine protease, calpains, among others [190, 269,
onstrated to be modulated by melatonin in brain. The 338, 367, 369]. Thus, the glutamate-induced overestimation
executory caspase-3, which is known to be directly linked to of NMDA receptors becomes neurotoxic; this process is
cyt c release and widely linked to cell death in AD [99, 320, common to several neurodegenerative diseases, and it is
354], can be downregulated by melatonin [92]. On the well known as glutamate excitotoxicity (reviewed in [269]).
contrary, melatonin may enhance bcl-2 expression as
demonstrated in AD transgenic mice [92] and in ischemic
brain [355]. Bcl-2 is recognized as an antiapoptotic protec-
MelatoninÕs role in calcium hypothesis
tive factor, and its relation to Ab is also widely known, Melatonin may reduce NMDA-induced high [Ca2+]c levels
because Ab may deplete bcl-2 as demonstrated in human in addition to its ability to directly inhibit the mitochondrial
primary neuron cultures [356], in microglia [357], or in permeability transition pores, a mechanism linked partic-
human neurons from patients with AD [358]. Furthermore, ularly to oxidative stress, as mentioned before [350]. Several
the proapoptotic bcl-2–associated X protein (Bax), which other mechanisms have been postulated to explain the
moves from the cytosol to the mitochondria, binds to bcl-2, regulation of intracellular Ca2+ by melatonin. As an
and promotes cyt c release, is increased in the presence of example, it is possible that melatonin acting on its MT2
Ab in human neurons [356]. However, under a variety of receptor may inhibit adenylyl cyclase, and with this, it
experimental conditions, melatonin has demonstrated its decreases cAMP formation, blocking the cAMP-dependent
utility in diminishing bax [174, 359–361]. Thus, melatonin protein kinase (PKA), which would activate calcium release
modulates mitochondrial pathways to apoptosis. channels [370–372]. The reader is reminded that pineal and
Mitochondrial damage is linked not only to energy cortical melatonin receptors, MT1 and MT2, are signifi-
dysmetabolism and leakage of free radicals, which in turn cantly decreased in AD brain [373]. Conversely, it has been
feeds back to induce oxidative stress, but also to increased known from many years that calcium influx regulates
leakage of Ca2+ currents, besides the above-mentioned melatonin production in the pineal gland [374] (Fig. 9).
apoptosis-inducing factors. Melatonin also may inhibit the mobilization of Ca2+
from ER as well as Ca2+ influx through voltage-sensitive
channels [375]. Importantly, melatonin controls the
Calcium hypothesis
NMDA receptor whose activation comprises multiple
Other hypotheses have been proposed, complementing the regulatory sites controlling Ca2+ influx into the cell. On
amyloid cascade theory. For example, it has been proposed the contrary, in the presence of the Ca2+ ionophore A-
that a calcium-signaling deficit causes accumulation of APP 23187, the inhibitory effect on Ca2+ by melatonin is
because APP a-processing is a Ca2+-dependent process, suppressed [376], returning to a glutamate-derived excit-
and this phenomenon provides excessive substrate for atory state. The mechanism involved in NMDA-R control
b- and c-secretases, the enzymes responsible for APP may also imply redox modulation [350, 377]. However, it is
processing and Ab overproduction (Fig. 1) [362]. That Ab also possible that melatonin increases the concentration of
increases calcium uptake has been demonstrated in PC12 the NMDA receptor subunits 2A and 2B, as demonstrated
cells [363], in human cortical neurons linked to glutamate in rat hippocampus, in a dose-dependent manner [378].
excitotoxicity [364], in AD brain frontal cortex, and in Another melatonin mechanism of protection related to
plasma membrane vesicles from both rat and human brain calcium influx control is the multifunctional calcium-
[365]. This occurs via stimulation of L voltage-sensitive modulated protein (calmodulin, or CaM) [379], which
calcium channels, as demonstrated in cultured neurons mediates the calcium requirement for retrograde axonal
[366], but Ab also may increase calcium uptake via transport of AchE [380]. Melatonin interaction with CaM is
potassium channels, the NMDA receptor, the nicotinic so avid that CaM had been considered a receptor for
receptor, or even by its own calcium-conducting pores melatonin [381, 382] (Fig. 4). Even though more recent
(reviewed in [367]). Conversely, calcium accelerates Ab NMR and molecular dynamic studies suggest a lower
aggregation, even at physiological concentrations [368], and affinity [383], it has been demonstrated that melatonin
it is exacerbated by synthetic calcium ionophores [369]. At decreases, in a specific manner, the activity and autophos-
the same time, Ab-induced calcium waves feed the neuro- phorylation of CaM kinase II, a key protein kinase
inflammatory response, and this increases Ab aggregation involved in neurite maturation [384], and causes neurite
and calcium waves, as mentioned previously [277–279]. enlargement through an increase in tubulin polymerization
During the events leading to oxidative stress and neur- derived from its CaM antagonism [385, 386]. In this
oinflammation in AD pathogeny, the glutamate-override of manner, apart from its antioxidant and antiapoptotic
the glutamate/cystine-antiporter system, which controls the effects as well as its anti-AchE actions related to Ca2+
levels of glutamate by exchanging cystine in cells for the and CaM modulatory effects, melatonin may preclude

182
AlzheimerÕs disease and melatonin

Fig. 9. Some of the most relevant issues in the calcium hypothesis are related to the following: (1) its functional association with c-and b-
secretase activity; thus, APP amyloidogenic processing seems to be a Ca2+-dependent process; (2) Ab, in turn, facilitates Ca2+ uptake using
a variety of calcium channels; (3) Ca2+ accelerates Ab aggregation, which in turn causes neuroinflammation, oxidative stress, and glutamate
release; (4) calcium massive entrance causes excessive accumulation of Ca2+ within ER and mitochondria, one dysfunctional and apoptosis-
related phenomenon; (5) dysregulated intracellular Ca2+ activates a number of enzymes including CaM, which in turn activates CaM-
dependent kinases responsible for tau phosphorylation; (6) cytosolic PLA2 is also a Ca-dependent enzyme and, when activated, causes
arachidonic acid release and the subsequent activation of the neuroinflammatory pathway via COX-2. Reports on melatonin (red crosses)
reveal a role for it: (1) as a free radical scavenger and antioxidant, (2) as an antiamyloidogenic, (3) as an inhibitor of Ca2+ influx, (4) as a
CaM antagonist, and melatonin also may impair Ca2+ leaking from ER, and (5) by regulating calreticulin, it is tempting to speculate that
melatonin may delay ER stress. APP, amyloid precursor protein; ROS/RNS, reactive oxygen and reactive nitrogen species; bs, beta
secretase; cs, gamma secretase; NMDA-R, n-methyl-daspartate receptor; L-Type R, voltage dependent L-type calcium receptor; AC,
adenylyl cyclase; cPLA2, cytoplasmic phospholipase A2; AA, arachidonic acid; ER, endoplasmic reticulum; UPR, unfolded protein re-
sponse; MIT, mitochondria; CaM, calmodulin.

microfilament and microtubule collapse, as demonstrated in ER stress, so it is tempting to speculate that ER stress-
N1E-115 cells [372]. Even more, by activating the Ca2+- induced CRT augments the folding capacity of the ER. By
dependent a isoform of PKC [387], melatonin may restore regulating calcium influx and release within the cytoplasm,
neurite formation, microtubule enlargement, and microfil- melatonin regulates not only cellular homeostasis but
ament organization in microspikes and growth cones in apoptosis as well (Fig. 9).
cells damaged with H2O2. On the contrary, the PKC It is obvious that in addition to the inhibition of ER and
inhibitor, bisindolylmaleimide, blocks neurite formation mitochondrial-related mechanisms of apoptosis, melatonin
and microfilament reorganization elicited by melatonin. may also prevent spontaneous neuronal apoptosis by acting
Thus, by regulating Ca2+ and CaM, melatonin possesses on downstream signal targets of the protein kinase B or
modulatory actions on cytoskeletal protein phosphoryla- Akt: the forkhead box protein O1 (FOXO-1) and the GSK-
tion, suggesting that it may re-establish neurite formation 3b [392]. Akt is linked to the PI3-K/Akt survival pathway
and the basal levels of phosphorylated tau, at least in N1E- and is required for the expression of long-term potentiation
115 cells treated with okadaic acid [388, 389]. in learning and memory. It is also linked to the insulin
There is another Ca2+-related melatonin receptor, which receptor substrate (IRS) for the purpose of regulating
may be relevant in AD, i.e., calreticulin (CRT) [390]. This glucose uptake through a series of phosphorylation events.
ER resident protein is known to have a role in the folding Moreover, PI3K is phosphorylated upon NMDA receptor-
and assembly of oligomeric membrane proteins. In fact, dependent CamKII activity [393], which is also a melatonin
CRT may function as a molecular chaperone for the Ab target, as mentioned above [386–388]. According to Tajes
precursor protein, as it has been demonstrated in HEK 293 et al. [392], melatonin increases the activation of prosur-
cells transfected with APP [391]. Calreticulin may initiate vival PI3K/Akt, whereas it inhibits GSK-3b and causes an

183
Rosales-Corral et al.

increase in FOXO-1 phosphorylation. This is a proapop-


Insulin resistance hypothesis
totic transcription factor involved in insulin activity and in
the cellular response to oxidative stress. Finally, GSK-3b, As can be seen, a number of glucose metabolism related
primarily linked to the inactivation of the glycogen factors begin to appear. GSK-3b is closely controlled by
synthase, has been also associated with AD and fronto- Akt and insulin signal transduction [162], while GSK-3a
temporal dementia [394] where it is related to microtubule increases Ab production [139]. Insulin-activated PI3K-Akt
stability because it phosphorylates the microtubule-associ- signaling pathway may be interfered with by Ab [396], and
ated protein tau [395] whose hyperphosphorylation is an it is well known that an impaired insulin signal transduction
early event preceding the appearance of neurofibrillary is involved in AD pathogeny [397]. On the other hand, the
tangles (NFT) in AD (Fig. 10). redox-sensitive insulin-degrading enzyme, IDE [398], the
most important degradative system for insulin and insulin-
like growth factor 1 (IGF-1), is also a major Ab-degrading
enzyme [399]. All of this leads us to another hypothesis,
which is provocative because it links AD to diabetes,
mitochondrial dysfunction, obesity, and even the choliner-
gic hypothesis (reviewed in [400]) (Fig. 10).
Also linked to Ab aggregates and oxidative stress, this
insulin resistance hypothesis involves IGF-1 and maintains
that a reduced input of this hormone, by causing insulin
resistance in the brain, may be the primary pathogenic
event in late-onset AD [401, 402]. In fact, many years before
the protective effect of IGF-1 in hippocampal neurons in
vitro subjected to Ab toxicity was described [403], epide-
miological studies revealed a significant association be-
tween diabetes mellitus and AD [165, 404, 405]. Later, it
was demonstrated that insulin deficiency and diabetes
mellitus exacerbate cerebral amyloidosis and behavioral
deficits in transgenic mice [406].
Insulin-like growth factor 1 is a natural molecule similar
to insulin, which controls insulin action and is responsible
for promoting growth effects, such as increased nitrogen
retention and cell proliferation. Beyond glucose homeosta-
sis, insulin is involved in cellular transport of Ab, and a lack
of an adequate insulin input has been related to intracel-
lular Ab accumulation [135]. In fact, by feeding Tg2576
mice with an insulin resistance-inducing diet, it is possible
to promote Ab aggregates, to increase c-secretase activities,
and to decrease IDE activity [406]. Ab peptides, particularly
Fig. 10. Ab and other associated events, such as high cholesterol
ADDLs, compete for insulin binding to the insulin recep-
levels, have insulin resistance in common. Insulin resistance inter-
rupts the neuroprotector PKB/Akt pathway, leading to activation tor, making the cell insulin resistant [407], very similar to
of GSK3 and diminishing IDE levels. Once GSK3 becomes active, IGF-1 receptor [408], and causing IGF-1/insulin dysfunc-
it phosphorylates tau while increasing Ab production, because tion; this in turn will affect amyloid trafficking via the
GSK3 is required for the amyloidogenic processing of APP. IDE, inappropriate MAPk and PI3K/Akt activation [401]. The
normally stimulated by Akt, reduces its function under insulin PI3K/Akt pathway also allows for IGF-1 to inhibit GSK-
resistance conditions. In this manner, insulin degradation is avoi-
3b [409], a kinase involved in the phosphorylation of tau, as
ded. However, because amyloid is also a target for IDE, Ab deg-
radation decreases in parallel with the former event, and the mentioned before, which is another hallmark of AD
neurotoxic peptide tends to accumulate as a consequence and pathology.
competes for insulin binding to the insulin receptor. Ab may even
inhibit IDEÕs activation by interfering with the PI3K/PKB/Akt
pathway. There are reports indicating that melatonin (red crosses) MelatoninÕs role in the insulin resistance
(1) may increase IGF-1 levels, (2) may activate the insulin receptor hypothesis
b-subunit tyrosine kinase, which then phosphorylates and activates
IRS, (3) may activate directly the PI3K/PKB/Akt pathway through
Male Syrian hamsters receiving melatonin for 10 wk show a
its MT receptors, and (4) may act on CaMKII melatonin inhibits significant increase in serum levels of IGF-1 [410], while the
tau phosphorylation, but if CaMKII were responsible for PI3K protective effect of melatonin on hepatocytes of CCl4-
activation (above left), then it would inhibit the PI3K/PKB/Akt exposed rats [411]has been attributed to increased IGF-1
pathway. MT, melatonin receptors; IGF-1, insulin-like growth levels [412]. Evaluated in the context of hypoxia-induced
factor; IR, insulin receptor; PI3K, phosphoinositide 3 kinase; IRS, periventricular white matter injury, melatonin reduces brain
insulin receptor substrate; PIP, phosphatidylinositol bis- and tri-
damage by enhancing IGF-1 activity, while it diminishes
phosphate; PDK-1, Phosphoinositide-dependent kinase; CREB,
cAMP response element binding; FOXO-1, Forkhead box protein glutamate release and the proinflammatory response
O1; IDE, insulin-degrading enzyme; GSK3, Glycogen synthase induced by hypoxia [413]. On the other hand, 6 months
kinase; ADDL, Ab-Derived Diffusible Ligands. after the administration on daily basis of 2 mg of melatonin

184
AlzheimerÕs disease and melatonin

in elderly women, IGF-I was found to be slightly but cytes express MT1 and MT2 receptors, and both seem to be
significantly increased [414]. necessary to activate the Akt/PI3k signaling pathway [161].
An explanation as to how melatonin raises IGF-1 levels In microglial cells, what or how many melatonin receptors
is related to melatonin-induced sleep improvement [415]. would be necessary to activate this same pathway, or even if
Sleep disturbances, as observed in obesity or in sleep apnea they are necessary, remains to be clarified. By resolving that
syndrome, cause alterations in the growth hormone/IGF-1 question, we might explain some differences between
system [416]. Sleep restriction, in fact, is related to increased neurons, microglia, and astrocytes in response to melato-
insulin resistance, as demonstrated both in obese subjects nin. This is relevant in relation to insulin because melatonin
and in those with AD. may influence insulin secretion [428, 429].
By acting on its receptors, which are present also in the Experimental evidence demonstrates that melatonin
hippocampus [298, 417], melatonin may induce a rapid improves glucose tolerance and increases insulin receptors
tyrosine phosphorylation and activation of the insulin in muscle and the expression of GLUT-4, a glucose
receptor b-subunit tyrosine kinase (IR) and downstream transporter, in addition to glucose clearance from the
Akt serine phosphorylation [418]. Thus, by using the PI3K- blood [430, 431]. A final explanation describes these
Akt signaling pathway, melatonin seems to activate Akt phenomena as protective actions by melatonin to prevent
(although other receptors acting on the same pathway hypoglycemia during winter dormancy in animals [432]. In
could also be involved [161]). The activation of Akt may the AD brain, where glucose uptake and metabolism are
have at least four consequences: (i) antiapoptotic activity, impaired even before the appearance of neuronal degener-
especially by reducing p53 transcriptional activity, which is ation [433], melatonin effects might contribute to delay the
particularly true in the hippocampus [419], in addition to progression of the disease, as has been experimentally
the phosphorylative inactivation of proapoptotic factors demonstrated in Ab-injected mice [147] as well as in high-
such as FOXO-1 [392] and the Bcl-2-associated death fat-diet-fed insulin-resistant mice [434] or in vitro by using
promoter (BAD) [420]; (ii) inhibition of tau hyperphosph- in C2C12 murine skeletal muscle cells where it was also
orylation and the consequent paired helical filament shown that the glucose transport amelioration by melato-
formation by inhibiting the tau kinase GSK-3b [392, 409]; nin may occur via insulin receptor substrate-1/PI3-kinase
(iii) activation in astrocytes of neurotrophic factors such as pathway [435].
cAMP response element binding (CREB) and the glial cell- Feeding rabbits a cholesterol-enriched diet also increases
derived neurotrophic factor (GDNF), while the neuroin- Ab levels in the hippocampus. This is an intriguing
flammatory control and neuronal plasticity are approached phenomenon, because cholesterol metabolism in the brain
through the astrocytic and neuronal overexpression of has been widely known to be independent of cholesterol
NF-jB; and (iv) the reactivation of IGF-1 could reduce metabolism in the body. Even more so, cholesterol in the
insulin resistance [397, 421], ameliorate glucose transport blood does not pass the blood brain barrier [436]. How
[422], and additionally reduce Ab accumulation from the hypercholesterolemia increases Ab levels in the brain may
cerebral parenchyma [401, 408, 409, 423] (Fig. 10). be explained by the oxidized cholesterol metabolite 27-
Here, it is necessary to look at melatoninÕs role in Akt hydroxycholesterol, which, acting on IGF-1 signaling,
signaling. In isolated microglial cells exposed to Ab, activates Akt. This survival promoter may decrease IDE,
melatonin has shown inhibitory, dose-dependent effects while it increases GSK-3 (a and b), and both phenomena
on the activity of Akt. This impairs the NADPH oxidase appear associated with elevated Ab levels, as observed in
assembly [270] as we have seen before at the purpose of AD organotypic hippocampal slices [423] (Fig. 10).
neuroinflammatory response. No apparent phospho-GSK-
3 was detected in those experiments. On the contrary, in
The lipid connection
isolated astrocytes, melatonin may induce Akt, and by this
means, it may inhibit GSK-3b [161]. These apparently The first question is how cholesterol in the blood increases
contradictory effects may be related to MT1/MT2 dimer- cholesterol in brain and how cholesterol in membranes
ization as a mechanism determining the receptor-mediated influences Ab overproduction. Answers to these questions
biological effects of melatonin [424]. As an example, the would help to explain why cholesterol may be considered
ERK activity is increased by melatonin in mouse neuro- an early risk factor for AD. There is a link between APP
blastoma cells that express only MT1 receptors [425], processing, Ab production, and the lipid environment [437,
whereas in human umbilical vein endothelial cells express- 438]. APP is a type I membrane protein that undergoes
ing both MT1 and MT2 receptors, ERK is inhibited by proteolytic cleavage within its ectodomain, followed by
melatonin [426]. However, differences related to types of intramembrane cleavage on its C-terminal fragment. These
melatonin receptors among astrocytes, microglia, and proteolytic pathways lead to the generation of Ab, and both
neurons have not been established yet, except in injured processes involve proteolytic enzymes known as secretases,
white matter of neonatal rats where MT1 and MT2 seem to as previously explained. The intramembrane cleavage site
be strongly expressed in both astrocytes and microglial cells of the APP depends on the length of its transmembrane
and to a lesser extent in neurons and immature oligoden- domain [439], while membrane composition and its physical
drocyte [427]. In the AD brain, an increased MT1 immu- state modulate the ratio between the APP derivatives Ab1–
noreactivity in pyramidal hippocampal neurons has been 42 and Ab1–40 [440]. Ab1–42 is more pathogenic than Ab1–40
documented [417], but in this case, differences between as described earlier in this review. The cleavage of APP may
astrocytes, microglia, and neurons in reference to their occur at the exact center of the lipid bilayer [441] where
melatonin receptors were not established (Fig. 4). Astro- cholesterol may enhance c-secretase-mediated Ab produc-

185
Rosales-Corral et al.

tion [442, 443] and, reciprocally, Ab aggregates preferen- Other important relationships between lipids and AD
tially bind cholesterol within the lipidic rafts [444]. Thus, a involve the polyunsaturated fatty acids (PUFAs), particu-
drastic reduction in membrane cholesterol may result in larly docosahexaenoic acid (DHA, 22:6x-3) and arachi-
decreased amyloid production [442, 443]. On the contrary, donic acid (AA, 20:4x-6), a precursor in the production of
it has also been shown that neuronal membrane cholesterol eicosanoids. Arachidonic acid is a well-known neuroin-
loss may enhance amyloid peptide generation [445], which flammation by-product, delivered from cellular membranes
calls into question the population-based studies which have by phospholipases A2, cyclooxygenases, and lipoxygenases
demonstrated that cholesterol is an early risk factor for the under the effect of cytokines and chemokines once the
development of amyloid pathology [446]. proinflammatory response to injury is established [164].
Sphingomyelin (SM), another particularly significant This is particularly true in the AD brain [449, 450].
lipid in signal transduction, when it accumulates, provides Importantly, AD is associated with depletion of DHA in
a favorable milieu for GM1 ganglioside-induced assembly the brain [451, 452]. DHA comprises about 40% of the
of Ab-protein [447]. According to Grimm et al. [438], while PUFAs in the brain [453], it is found predominantly in
the control of cholesterol and SM metabolism involves APP neuronal membranes in the gray matter, and it has an
processing via regulation of the activity of the c-secretase important role in cholinergic stimulated signal transduction
enzyme, Ab directly activates neutral sphingomyelinase, the at the synapse and phospholipid-mediated signal transduc-
enzyme responsible for metabolizing SM to phosphocholine tion involving activation of phospholipase A2 and/or PLC
and ceramide and downregulates SM levels or reduces de [454]. In fact, regional gray matter volumes in the anterior
novo cholesterol synthesis by inhibiting the hydroxymethyl- cingulate cortex, amygdala, and hippocampus, calculated
glutaryl-CoA reductase (HMGR) activity. Thus, Ab using optimized voxel-based morphometry on high-resolu-
becomes a cholesterol and sphingomyelin regulator, while tion structural magnetic resonance images in adult humans,
cholesterol and glycosphingolipids may elevate Ab produc- revealed a positive association between long-chain omega-3
tion and SM may reduce Ab production by inhibition of fatty acid intake and corticolimbic gray matter increase in
c-secretase [438]. This is particularly true in individuals volume [455]. There is experimental evidence supporting the
carrying the ApoE4 genotype, because this isoform binds role of DHA in the formation of new memories; for
more avidly to Ab [448] (Fig. 11). example, in fish oil-deficient rats tested for learning ability

Fig. 11. Cholesterol may interfere with a-secretase activity or may enhance c-secretase (cs) activity, favoring the amyloidogenic pathway;
also, it is possible that cholesterol contributes by relocalizing APP near to the b-secretase (bs) influence. Ab, correspondingly, affects
cholesterol influx and efflux because of its ability to bind cholesterol transporters, while it inhibits cholesterol esterification. Sphingomyelin,
which makes up about 10% of the lipids of brain, has been related to c-secretase (cs) activity and the amyloidogenic pathway. Ab for its part
may induce apoptosis by activating the cPLA2-dependent sphingomyelinase–ceramide pathway, which is a phenomenon related to caspase 3
activation, PI3K/Akt inhibition, and arachidonic acid release. MelatoninÕs role (red crosses) in this hypothesis is related to the following: (1)
functional stability of the membrane by avoiding the lipid peroxidation cascade, (2) interfering with lipid-derived proinflammatory signals,
(3) it is also possible that melatonin may interfere with PLA2 activity, (4) melatonin reduces cholesterol absorption or augments endogenous
cholesterol clearance mechanisms, and (5) melatonin may interfere with cholesterol transport as well or directly interact with cholesterol;
even though the effects derived from this possible interaction remain to be clarified, it has been demonstrated that melatonin may favor the
displacement of cholesterol from the phospholipid bilayer. Reciprocally, cholesterol might influence melatoninÕs antioxidant capacity.

186
AlzheimerÕs disease and melatonin

related to reference memory and working memory, chronic triacylglyceride, very low-density lipoprotein cholesterol,
administration of DHA led to an improvement in reference and low-density lipoprotein cholesterol in plasma and the
memory-related learning both in young [456] and in old rats concentrations of cholesterol and triacylglyceride in the
[457]. The diet-induced increase in brain DHA levels has liver; this was demonstrated in rats fed on high-cholesterol
been related to enhanced reference and working memory diet [472]. In a very particular study (hypercholesterolemic
performance as well [458]. Moreover, DHA may promote mice fed with an atherogenic diet), it has reported that
the differentiation of neural stem cells into neurons by melatonin induces atherosclerotic lesions in the proximal
promoting cell cycle exit and suppressing cell death as aorta as a consequence of exacerbated LDL oxidation
evaluated in neural stem cells obtained from 15.5-day-old [473]. On the contrary, in normolipidemic postmenopausal
rat embryos [459] probably by regulating basic helix–loop– women, melatonin has shown to inhibit oxidative modifi-
helix transcription factors [460]. Ab oligomer-induced cation of low-density lipoprotein particles [474].
effects, such as phosphorylation of tau and inactivation of The cholesterol–melatonin relationship in the biological
insulin receptor substrate via c-Jun N-terminal kinase membranes is, from a functional point of view, particularly
signaling, may be suppressed by the dietary intake of fish relevant. Evaluated in dry cholesterol/lecithin mixed
oil/DHA, and this may be accompanied by improvement in reversed micelles, melatonin is mainly located in and
Y-maze performance, as demonstrated in 3xTg-AD mice oriented in the proximity of the polar heads where it
[461]. appears to compete with cholesterol for the hydrophilic
binding sites of lecithin; this is particularly true at high
cholesterol concentrations. Thus, free radical displacement
MelatoninÕs role in the lipid connection coming from the aqueous compartment to a high concen-
MelatoninÕs utility is attributable in great part to its tration of cholesterol could influence melatoninÕs antioxi-
amphipathic nature, which allows it to pass easily through dant activity. At the same time, the more concentrated
all morphophysiological barriers, and this property in- cholesterol is in membranes, the greater the membrane
volves an active interaction with membrane lipids. The rigidity. It is speculated that competition with melatonin
most widely known effect of melatonin on lipids is to may favor the displacement of cholesterol from the
prevent their peroxidation [14, 462, 463]. In fact, there is an phospholipid bilayer [475]. This feature gains relevance
inverse correlation between melatonin levels and peroxida- because decreasing membrane cholesterol in mature neu-
tion of lipids following the intracerebral injection of Ab rons may reduce their susceptibility to Ab, tau production,
[464]. By doing this, melatonin avoids some of the major and cell death, whereas increasing membrane cholesterol in
problems derived from cell membrane dysfunction and young neurons enhances the Ab-mediated cellular pro-
subsequent neurotoxicity [462, 465]. However, the role of cesses, as demonstrated in hippocampal neurons [476]
melatonin seems to be more complex (Fig. 11). (Fig. 11).
Melatonin may interfere with proinflammatory signals by
blocking two principal enzymatic pathways: COX and LOX.
Concluding remarks
Both of these insert molecular oxygen into molecules of free
and esterified polyunsaturated fatty acids, such as arachi- Is it just a coincidence that while melatonin declines with
donic acid, and thereby synthesize several different biolog- age, the probability of experiencing AD grows? New
ically active eicosanoids. 12/15-LOX protein levels and findings on CSF flow, possibly moving from the choroid
enzyme activity, linked to mechanisms of oxidative stress fissure into the ventricular system, could help to explain
and neurodegeneration, have been demonstrated to be why melatonin is found in higher concentration in the CSF
increased in AD brain [466], the same as 5-LOX. This latter than in simultaneously sampled blood. Thus, neural tissue
is also known as an active Ab inducer [467], and 5-LOX gene in contact with the ventricular system via hypothetical
expression may be suppressed by melatonin through its high- choroid plexus portals would have high levels of cellular
affinity nuclear receptors, as demonstrated in hippocampus melatonin [477]. A CSF deficiency of melatonin has been
[468]. It is noted that a drop in melatonin levels, as observed demonstrated to precede clinical symptoms of AD [22, 265]
in elderly subjects with an aging-associated melatonin and, the loss of this lipophilic antioxidant normally
deficiency, has been related to 5-LOX overexpression [469]. concentrated in the ventricular CSF exposes highly active
On the contrary, a proradical effect has been described for and vulnerable brain tissue to self-generated oxygen radi-
melatonin in which it uses these pathways [470], which is a cals.
transitory, early melatonin effect observed in leukocytes, MelatoninÕs more common indication in patients with
accompanied by strong liberation of arachidonic acid and AD is sleep regulation because sleep disruptions, nightly
explained as a consequence of calmodulin binding. restlessness, and sundowning are frequently observed in
Lipid mobilization to obtain polyunsaturated fatty acids elderly and particularly in patients with AD [44]. This is
from the membranes for LOX and COX enzymes requires related to decreased levels of both melatonin [415] and
the intervention of an enzyme, the calcium-dependent melatonin receptors in the SCN [478]. It has also a potential
phospholipase A2 (PLA2); melatonin may be a negative to treat mood disorders [30, 43], commonly associated with
endogenous regulator of cytosolic PLA2 presumably AD [28, 30].
through a melatonin receptor-mediated process as demon- By taking into account the most compelling hypotheses
strated in the rat pineal gland in vitro [471]. trying to explain the cellular and biomolecular alterations
Melatonin may also significantly reduce cholesterol in AlzheimerÕs disease, however, a growing body of
absorption causing significant decreases in total cholesterol, evidence supports the protective role of melatonin, exceed-

187
Rosales-Corral et al.

ing the above-mentioned conventional uses. Thus, melato- caused the memory impairment actually, because the
nin has a role in each of the different reviewed hypotheses: electroconvulsive stimulation has been often related to
(i) it prevents amyloid overproduction, (ii) it reduces memory impairment by itself; and not only in rodents but
hyperphosphorylation of tau [15, 166], (iii) it is an antiox- in humans (reviewed in [483]). On the other hand, not in
idant and free radical scavenger, (iv) it modulates proin- rats but in epileptic children, a randomized, double-blind,
flammatory processes, (v) it works well as an placebo-controlled trial demonstrated that melatonin im-
anticholinesterase agent, (vi) it prevents mitochondrial proved cognitive and social function as well as emotional
damage and the apoptotic phenomena related with AD, well-being and behavior [484]. It has been also reported that
(vii) it may impair calcium-dependent toxicity, (viii) it ramelteon, a synthetic melatonin derivative, administrated
reduces insulin resistance as well as glucose transport, and prior to a short (2 hr) evening nap, impairs significantly
finally (ix) it is able to maintain the integrity and function- neurobehavioral performance for up to 12 hr after awak-
ality of cellular membranes, thanks to its ability to interact ening [485].However, a 2-hr nap is not a short nap; naps
with lipids or against their neuroinflammatory or proapop- longer than 30 min have been largely associated with a loss
totic signals when the lipid balance becomes affected. of productivity and sleep inertia [486]. Thus, probably
The functional translation of these biomolecular effects ramelteon was not responsible for a low neurobehavioral
has been also well documented. MT2 receptor-deficient performance, but the long nap by itself.
mice undergo impairment of synaptic plasticity and learn- Melatonin doses ranging from 3 to 6.6 g/day for more
ing-dependent behavior, suggesting that MT2 receptors than 30 days were administrated in patients with Parkin-
participate in hippocampal synaptic plasticity and in sonÕs disease, and the number of collateral effects, such as
memory processes [299]. Also relevant are the protective headache, somnolence, or abdominal cramps, were isolated,
effects of melatonin on cognition in a variety of tasks of with melatonin being Ôremarkably wellÕ tolerated [487].
working memory, spatial reference learning/memory, and More severe collateral effects have never been observed. In
basic mnemonic function, as observed in a transgenic model fact, doses up to 800 mg/kg failed to produce death in mice
of AD [20]. It is worth remembering the increased mela- [488]; indeed, no lethal dose of melatonin has been
tonin 1a-receptor immunoreactivity in the hippocampus of established overall; melatonin has been repeatedly shown,
patients with AD, which may be a compensatory response at any dose to be free of significant side effects.
to impaired melatonin levels [417]. Other concerns go in the opposite direction: melatoninÕs
It is also true that in transgenic animals, additionally extremely short half-life in the circulation, a question that
exposed to aluminum for months (aluminum has been have led to the development of synthetic melatonergic drugs
circumstantially linked to AD [16, 479, 480]), melatonin did with substantially longer half-life than melatonin [489]. It
not ameliorate the behavioral effects [21] even though they should be noted however that the short half-time of
did respond well to the antioxidant actions of [92]. Limited melatonin in the blood does not necessarily translate into
or even null results in memory performance or other high a short half-life within the cells.
mental functions using melatonin have been observed in This review underlines the potential of melatonin to slow
some clinical trials [11]. However, these changes are the progressive deterioration of AD brain, in light of the
associated with the loss of brain cells. The greater the loss known hypotheses that attempt to explain the neurodegen-
of brain cells, the more severe the deterioration in high eration. As observed in AD models and based on a
mental functions, and no drug exists that is capable of multitude of experimental results, melatonin benefits may
regenerating lost neurons. Once the brain tissue has well stem from actions that exceed its well-known antiox-
degenerated, there is just a little chance of recovering [176]. idant properties. Unfortunately, while there are a number
There are several concerns about melatonin. From a of well-founded hypotheses, the real cause of neurodegen-
cellular, basic perspective, we find a single report where eration in AD is still unknown. Very likely, there are many
melatonin reportedly worsens the neurodegenerative contributing causes to this highly complex disease.
pathology. It is a rotenone-induced ParkinsonÕs disease-
specific model, where melatonin not only failed to impair
neuronal degeneration but potentiated neurodegeneration
Acknowledgements
[19]. However, synergistic effects of melatonin against A. Coto-Montes is an associate professor at Universidad de
MPTP-induced mitochondrial damage and dopamine Oviedo (Spain). Jose A. Boga is a researcher of HUCA/
depletion have been also reported [481]. Otherwise, the FICYT (Oviedo, Spain). His stay at UTHSCSA has been
evidence indicating the protective role of melatonin on subsidized by Instituto de Salud Carlos III (Madrid, Spain).
mitochondria within CNS is vast [239, 342–346, 348–350,
352, 353, 355, 359].
On the other hand, there are several clinical concerns.
References
One refers to dose and side effects. In a few isolated studies, 1. Querfurth HW, Laferla FM. AlzheimerÕs disease. N Engl
melatonin has been related to sleepiness, dizziness, head- J Med 2010; 362:329–344.
aches, nightmares, confusion, sleepwalking, daytime sleep- 2. AlzheimerÕs Association, Thies W, Bleiler L. 2011
iness, and abdominal discomfort, even though some results AlzheimerÕs disease facts and figures. Alzheimers Dement
deserve a re-analysis. For example, using a high dose of 2011; 7:208–244.
melatonin (20 mg/kg) in mice undergoing electroconvulsive 3. Hebert LE, Scherr PA, Bienias JL et al. Alzheimer disease
stimulation, a strong long-term memory deficit was attrib- in the US population: prevalence estimates using the 2000
uted to melatonin [482]. However, it was not clear what census. Arch Neurol 2003; 60:1119–1122.

188
AlzheimerÕs disease and melatonin

4. Martorana A, Esposito Z, Koch G. Beyond the cholinergic panied by decreased cerebrospinal fluid melatonin levels.
cypothesis: do current drugs work in AlzheimerÕs disease? J Pineal Res 2003; 35:125–130.
CNS Neurosci Ther 2010; 16:235–245. 23. Liu RY, Zhou JN, Van HJ et al. Decreased melatonin levels
5. Hansen RA, Gartlehner G, Lohr KN et al. Functional in postmortem cerebrospinal fluid in relation to aging, Alz-
outcomes of drug treatment in AlzheimerÕs disease: a systematic heimerÕs disease, and apolipoprotein E-epsilon4/4 genotype.
review and meta-analysis. Drugs Aging 2007; 24:155–167. J Clin Endocrinol Metab 1999; 84:323–327.
6. Hayden KM, Zandi PP, Khachaturian AS et al. Does 24. De BM, Pappas BA. Pinealectomy causes hippocampal CA1
NSAID use modify cognitive trajectories in the elderly? The and CA3 cell loss: reversal by melatonin supplementation.
Cache County study. Neurology 2007; 69:275–282. Neurobiol Aging 2007; 28:306–313.
7. Breitner JC, Haneuse SJ, Walker R et al. Risk of dementia 25. Ling ZQ, Tian Q, Wang L et al. Constant illumination
and AD with prior exposure to NSAIDs in an elderly com- induces Alzheimer-like damages with endoplasmic reticulum
munity-based cohort. Neurology 2009; 72:1899–1905. involvement and the protection of melatonin. J Alzheimers
8. Aisen PS, Davis KL, Berg JD et al. A randomized Dis 2009; 16:287–300.
controlled trial of prednisone in AlzheimerÕs disease. Alzhei- 26. Reiter RJ. Pineal melatonin: cell biology of its synthesis and
merÕs Disease Cooperative Study. Neurology 2000; 54:588– of its physiological interactions. Endocr Rev 1991; 12:151–180.
593. 27. Greilberger J, Fuchs D, Leblhuber F et al. Carbonyl
9. Reines SA, Block GA, Morris JC et al. Rofecoxib: no effect proteins as a clinical marker in AlzheimerÕs disease and its
on AlzheimerÕs disease in a 1-year, randomized, blinded, relation to tryptophan degradation and immune activation.
controlled study. Neurology 2004; 62:66–71. Clin Lab 2010; 56:441–448.
10. Neugroschl J, Sano M. Current treatment and recent clin- 28. Zubenko GS, Moossy J, Martinez AJ et al. Neuropatho-
ical research in AlzheimerÕs disease. Mt Sinai J Med 2010; logic and neurochemical correlates of psychosis in primary
77:3–16. dementia. Arch Neurol 1991; 48:619–624.
11. Jansen SL, Forbes DA, Duncan V et al. Melatonin for 29. Seshadri S, Beiser A, Selhub J et al. Plasma homocysteine
cognitive impairment. Cochrane Database Syst Rev 2006; as a risk factor for dementia and AlzheimerÕs disease. N Engl
25:CD003802. J Med 2002; 346:476–483.
12. Spuch C, Antequera D, Isabel Fernandez-Bachiller M 30. Ouchi Y, Yoshikawa E, Futatsubashi M et al. Altered
et al. A new tacrine-melatonin hybrid reduces amyloid burden brain serotonin transporter and associated glucose metabo-
and behavioral deficits in a mouse model of AlzheimerÕs dis- lism in Alzheimer disease. J Nucl Med 2009; 50:1260–1266.
ease. Neurotox Res 2010; 17:421–431. 31. Kvetnoy IM. Extrapineal melatonin: location and role
13. Cardinali DP, Brusco LI, Liberczuk C et al. The use of within diffuse neuroendocrine system. Histochem J 1999;
melatonin in AlzheimerÕs disease. Neuro Endocrinol Lett 31:1–12.
2002; 23(Suppl 1):20–23. 32. Kvetnoy IM, Reiter RJ, Khavinson VK. Letter to the
14. Pappolla MA, Sos M, Omar RA et al. Melatonin prevents Editor. Claude Bernard was right: hormones may be pro-
death of neuroblastoma cells exposed to the Alzheimer amy- duced by Ônon-endocrineÕ cells. Neuro Endocrinol Lett 2000;
loid peptide. J Neurosci 1997; 17:1683–1690. 21:173–174.
15. Wang XC, Zhang J, Yu X et al. Prevention of isoproterenol- 33. Pevet P. Melatonin and biological rhythms. Biol Signals
induced tau hyperphosphorylation by melatonin in the rat. Recept 2000; 9:203–212.
Sheng Li Xue Bao 2005; 57:7–12. 34. Dominguez-Rodriguez A, Abreu-Gonzalez P, Sanchez-
16. Van Rensburg SJ, Daniels WM, Potocnik FC et al. A new Sanchez JJ et al. Melatonin and circadian biology in human
model for the pathophysiology of AlzheimerÕs disease. Alu- cardiovascular disease. J Pineal Res 2010; 49:14–22.
minium toxicity is exacerbated by hydrogen peroxide and 35. Reiter RJ, Cuna-Castroviejo D, Tan DX et al. Free rad-
attenuated by an amyloid protein fragment and melatonin. S ical-mediated molecular damage. Mechanisms for the pro-
Afr Med J 1997; 87:1111–1115. tective actions of melatonin in the central nervous system.
17. Yang X, Yang Y, Fu Z et al. Melatonin ameliorates Alz- Ann N Y Acad Sci 2001; 939:200–215.
heimer-like pathological changes and spatial memory reten- 36. Jou MJ, Peng TI, Hsu LF et al. Visualization of melatoninÕs
tion impairment induced by calyculin A. J Psychopharmacol multiple mitochondrial levels of protection against mito-
2010; 25:1118–1125. chondrial Ca(2+)-mediated permeability transition and
18. Singer C, Tractenberg RE, Kaye J et al. A multicenter, beyond in rat brain astrocytes. J Pineal Res 2010; 48:20–38.
placebo-controlled trial of melatonin for sleep disturbance in 37. Romero A, Egea J, Garcia AG et al. Synergistic neuro-
AlzheimerÕs disease. Sleep 2003; 26:893–901. protective effect of combined low concentrations of galanta-
19. Tapias V, Cannon JR, Greenamyre JT. Melatonin treat- mine and melatonin against oxidative stress in SH-SY5Y
ment potentiates neurodegeneration in a rat rotenone Par- neuroblastoma cells. J Pineal Res 2010; 49:141–148.
kinsonÕs disease model. J Neurosci Res 2010; 88:420–427. 38. Milczarek R, Hallmann A, Sokolowska E et al. Mela-
20. Olcese JM, Cao C, Mori T et al. Protection against cogni- tonin enhances antioxidant action of alpha-tocopherol and
tive deficits and markers of neurodegeneration by long-term ascorbate against N. J Pineal Res 2010; 49:149–155.
oral administration of melatonin in a transgenic model of 39. Reiter RJ, Manchester LC, Tan DX. Neurotoxins: free
Alzheimer disease. J Pineal Res 2009; 47:82–96. radical mechanisms and melatonin protection. Curr Neuro-
21. Garcia T, Ribes D, Colomina MT et al. Evaluation of the pharmacol 2010; 8:194–210.
protective role of melatonin on the behavioral effects of alu- 40. Carrillo-Vico A, Guerrero JM, Lardone PJ et al. A
minum in a mouse model of AlzheimerÕs disease. Toxicology review of the multiple actions of melatonin on the immune
2009; 265:49–55. system. Endocrine 2005; 27:189–200.
22. Zhou JN, Liu RY, Kamphorst W et al. Early neuropatho- 41. Jung KH, Hong SW, Zheng HM et al. Melatonin amelio-
logical AlzheimerÕs changes in aged individuals are accom- rates cerulein-induced pancreatitis by the modulation of

189
Rosales-Corral et al.

nuclear erythroid 2-related factor 2 and nuclear factor-kap- 64. Fukutomi J, Fukuda A, Fukuda S et al. Scavenging activity
paB in rats. J Pineal Res 2010; 48:239–250. of indole compounds against cisplatin-induced reactive oxy-
42. Chahbouni M, Escames G, Venegas C et al. Melatonin gen species. Life Sci 2006; 80:254–257.
treatment normalizes plasma pro-inflammatory cytokines and 65. Tan DX, Manchester LC, Reiter RJ et al. Melatonin di-
nitrosative/oxidative stress in patients suffering from Duch- rectly scavenges hydrogen peroxide: a potentially new meta-
enne muscular dystrophy. J Pineal Res 2010; 48:282–289. bolic pathway of melatonin biotransformation. Free Radic
43. Srinivasan V, Smits M, Spence W et al. Melatonin in mood Biol Med 2000; 29:1177–1185.
disorders. World J Biol Psychiatry 2006; 7:138–151. 66. Zavodnik IB, Lapshina EA, Zavodnik LB et al. Hypo-
44. Ferguson SA, Rajaratnam SM, Dawson D. Melatonin ag- chlorous acid-induced oxidative stress in Chinese hamster B14
onists and insomnia. Expert Rev Neurother 2010; 10:305–318. cells: viability, DNA and protein damage and the protective
45. Galecki P, Szemraj J, Bartosz G et al. Single-nucleotide action of melatonin. Mutat Res 2004; 559:39–48.
polymorphisms and mRNA expression for melatonin syn- 67. Matuszak Z, Bilska MA, Reszka KJ et al. Interaction of
thesis rate-limiting enzyme in recurrent depressive disorder. singlet molecular oxygen with melatonin and related indoles.
J Pineal Res 2010; 48:311–317. Photochem Photobiol 2003; 78:449–455.
46. Hill SM, Frasch T, Xiang S et al. Molecular mechanisms 68. Ximenes VF, Silva SO, Rodrigues MR et al. Superoxide-
of melatonin anticancer effects. Integr Cancer Ther 2009; dependent oxidation of melatonin by myeloperoxidase. J Biol
8:337–346. Chem 2005; 280:38160–38169.
47. Sotthibundhu A, Phansuwan-Pujito P, Govitrapong P. 69. Aydogan S, Yerer MB, Goktas A. Melatonin and nitric
Melatonin increases proliferation of cultured neural stem cells oxide. J Endocrinol Invest 2006; 29:281–287.
obtained from adult mouse subventricular zone. J Pineal Res 70. Reiter RJ, Tan DX, Manchester LC et al. Biochemical
2010; 49:291–300. reactivity of melatonin with reactive oxygen and nitrogen
48. Reiter RJ. The melatonin rhythm: both a clock and a cal- species: a review of the evidence. Cell Biochem Biophys 2001;
endar. Experientia 1993; 49:654–664. 34:237–256.
49. Reiter RJ, Tan DX, Manchester LC et al. Melatonin and 71. Tan DX, Reiter RJ, Manchester LC et al. Chemical and
reproduction revisited. Biol Reprod 2009; 81:445–456. physical properties and potential mechanisms: melatonin as a
50. Reiter RJ, Richardson BA, Johnson LY et al. Pineal broad spectrum antioxidant and free radical scavenger. Curr
melatonin rhythm: reduction in aging Syrian hamsters. Sci- Top Med Chem 2002; 2:181–197.
ence 1980; 210:1372–1373. 72. Matuszak Z, Reszka K, Chignell CF. Reaction of mela-
51. Reiter RJ, Craft CM, Johnson JE Jr et al. Age-associated tonin and related indoles with hydroxyl radicals: EPR and
reduction in nocturnal pineal melatonin levels in female rats. spin trapping investigations. Free Radic Biol Med 1997;
Endocrinology 1981; 109:1295–1297. 23:367–372.
52. Sack RL, Lewy AJ, Erb DL et al. Human melatonin pro- 73. Tan DX, Manchester LC, Terron MP et al. One molecule,
duction decreases with age. J Pineal Res 1986; 3:379–388. many derivatives: a never-ending interaction of melatonin
53. Reiter RJ. The ageing pineal gland and its physiological with reactive oxygen and nitrogen species? J Pineal Res 2007;
consequences. Bioessays 1992; 14:169–175. 42:28–42.
54. Wu YH, Swaab DF. The human pineal gland and melatonin 74. Niu S, Li F, Tan DX et al. Analysis of N1-acetyl-N2-formyl-5-
in aging and AlzheimerÕs disease. J Pineal Res 2005; 38:145– methoxykynuramine/N1-acetyl-5-methoxy-kynuramine for-
152. mation from melatonin in mice. J Pineal Res 2010; 49:106–114.
55. Lynch HJ, Wurtman RJ, Moskowitz MA et al. Daily 75. Kuesel JT, Hardeland R, Pfoertner H et al. Reactions of
rhythm in human urinary melatonin. Science 1975; 187:169– the melatonin metabolite N(1)-acetyl-5-methoxykynuramine
171. with carbamoyl phosphate and related compounds. J Pineal
56. Tan DX, Chen LD, Poeggeler B et al. Melatonin: a potent, Res 2010; 48:47–54.
endogenous hydroxyl radical scavenger. 1993; 1:57–60. 76. Tan DX, Manchester LC, Burkhardt S et al. N1-acetyl-
57. Galano A. On the direct scavenging activity of melatonin N2-formyl-5-methoxykynuramine, a biogenic amine and
towards hydroxyl and a series of peroxyl radicals. Phys Chem melatonin metabolite, functions as a potent antioxidant.
Chem Phys 2011; 13:7178–7188. FASEB J 2001; 15:2294–2296.
58. Poeggeler B, Reiter RJ, Tan DX et al. Melatonin, hy- 77. Hirata F, Hayaishi O, Tokuyama T et al. In vitro and in
droxyl radical-mediated oxidative damage, and aging: a vivo formation of two new metabolites of melatonin. J Biol
hypothesis. J Pineal Res 1993; 14:151–168. Chem 1974; 249:1311–1313.
59. Reiter RJ, Paredes SD, Manchester LC et al. Reducing 78. Hardeland R, Tan DX, Reiter RJ. Kynuramines, metab-
oxidative/nitrosative stress: a newly-discovered genre for olites of melatonin and other indoles: the resurrection of an
melatonin. Crit Rev Biochem Mol Biol 2009; 44:175–200. almost forgotten class of biogenic amines. J Pineal Res 2009;
60. Reiter RJ, Tan DX, Terron MP et al. Melatonin and its 47:109–126.
metabolites: new findings regarding their production and their 79. Ressmeyer AR, Mayo JC, Zelosko V et al. Antioxidant
radical scavenging actions. Acta Biochim Pol 2007; 54:1–9. properties of the melatonin metabolite N1-acetyl-5-meth-
61. Stasica P, Ulanski P, Rosiak JM. Melatonin as a hydroxyl oxykynuramine (AMK): scavenging of free radicals and pre-
radical scavenger. J Pineal Res 1998; 25:65–66. vention of protein destruction. Redox Rep 2003; 8:205–213.
62. Ebelt H, Peschke D, Bromme HJ et al. Influence of mela- 80. Honson NS, Johnson RL, Huang W et al. Differentiating
tonin on free radical-induced changes in rat pancreatic beta- Alzheimer disease-associated aggregates with small molecules.
cells in vitro. J Pineal Res 2000; 28:65–72. Neurobiol Dis 2007; 28:251–260.
63. Velkov ZA, Velkov YZ, Galunska BT et al. Melatonin: 81. Cohen T, Frydman-Marom A, Rechter M et al. Inhibition
quantum-chemical and biochemical investigation of antioxi- of amyloid fibril formation and cytotoxicity by hydroxyindole
dant activity. Eur J Med Chem 2009; 44:2834–2839. derivatives. Biochemistry (Mosc) 2006; 45:4727–4735.

190
AlzheimerÕs disease and melatonin

82. Sun L, Tran N, Tang F et al. Synthesis and biological 102. Gentleman SM, Greenberg BD, Savage MJ et al. A beta
evaluations of 3-substituted indolin-2-ones: a novel class of 42 is the predominant form of amyloid beta-protein in the
tyrosine kinase inhibitors that exhibit selectivity toward par- brains of short-term survivors of head injury. Neuroreport
ticular receptor tyrosine kinases. J Med Chem 1998; 41:2588– 1997; 8:1519–1522.
2603. 103. Priller C, Bauer T, Mitteregger G et al. Synapse for-
83. Guerrero JM, Reiter RJ. Melatonin-immune system rela- mation and function is modulated by the amyloid precursor
tionships. Curr Top Med Chem 2002; 2:167–179. protein. J Neurosci 2006; 26:7212–7221.
84. Barlow-Walden LR, Reiter RJ, Abe M et al. Melatonin 104. Li H, Wang Z, Wang B et al. Genetic dissection of the
stimulates brain glutathione peroxidase activity. Neurochem amyloid precursor protein in developmental function and
Int 1995; 26:497–502. amyloid pathogenesis. J Biol Chem 2010; 285:30598–30605.
85. Pablos MI, Agapito MT, Gutierrez R et al. Melatonin 105. Okuda T, Haga T. High-affinity choline transporter. Neu-
stimulates the activity of the detoxifying enzyme glutathione rochem Res 2003; 28:483–488.
peroxidase in several tissues of chicks. J Pineal Res 1995; 106. Turner PR, OÕconnor K, Tate WP et al. Roles of amyloid
19:111–115. precursor protein and its fragments in regulating neural activ-
86. Rodriguez C, Mayo JC, Sainz RM et al. Regulation of ity, plasticity and memory. Prog Neurobiol 2003; 70:1–32.
antioxidant enzymes: a significant role for melatonin. J Pineal 107. Hooper NM, Keen J, Pappin DJ et al. Pig kidney angio-
Res 2004; 36:1–9. tensin converting enzyme. Purification and characterization of
87. Kilanczyk E, Bryszewska M. The effect of melatonin on amphipathic and hydrophilic forms of the enzyme establishes
antioxidant enzymes in human diabetic skin fibroblasts. Cell C-terminal anchorage to the plasma membrane. Biochem J
Mol Biol Lett 2003; 8:333–336. 1987; 247:85–93.
88. Wakatsuki A, Okatani Y, Shinohara K et al. Melatonin 108. Lammich S, Kojro E, Postina R et al. Constitutive and
protects fetal rat brain against oxidative mitochondrial regulated alpha-secretase cleavage of AlzheimerÕs amyloid
damage. J Pineal Res 2001; 30:22–28. precursor protein by a disintegrin metalloprotease. Proc Natl
89. Kerman M, Cirak B, Ozguner MF et al. Does melatonin Acad Sci USA 1999; 96:3922–3927.
protect or treat brain damage from traumatic oxidative stress? 109. Haass C, Hung AY, Schlossmacher MG et al. beta-Amy-
Exp Brain Res 2005; 163:406–410. loid peptide and a 3-kDa fragment are derived by distinct
90. Mayo JC, Sainz RM, Uria H et al. Inhibition of cell pro- cellular mechanisms. J Biol Chem 1993; 268:3021–3024.
liferation: a mechanism likely to mediate the prevention of 110. Hotoda N, Koike H, Sasagawa N et al. A secreted form of
neuronal cell death by melatonin. J Pineal Res 1998; 25:12–18. human ADAM9 has an alpha-secretase activity for APP.
91. Feng Z, Qin C, Chang Y et al. Early melatonin supple- Biochem Biophys Res Commun 2002; 293:800–805.
mentation alleviates oxidative stress in a transgenic mouse 111. Buxbaum JD, Liu KN, Luo Y et al. Evidence that tumor
model of AlzheimerÕs disease. Free Radic Biol Med 2006; necrosis factor alpha converting enzyme is involved in regu-
40:101–109. lated alpha-secretase cleavage of the Alzheimer amyloid
92. Garcia T, Esparza JL, Nogues MR et al. Oxidative stress protein precursor. J Biol Chem 1998; 273:27765–27767.
status and RNA expression in hippocampus of an animal 112. Bandyopadhyay S, Goldstein LE, Lahiri DK et al. Role
model of AlzheimerÕs disease after chronic exposure to alu- of the APP non-amyloidogenic signaling pathway and tar-
minum. Hippocampus 2010; 20:218–225. geting alpha-secretase as an alternative drug target for treat-
93. Dubocovich ML. Pharmacology and function of melatonin ment of AlzheimerÕs disease. Curr Med Chem 2007; 14:2848–
receptors. FASEB J 1988; 2:2765–2773. 2864.
94. Calamini B, Santarsiero BD, Boutin JA et al. Kinetic, 113. Postina R. A closer look at alpha-secretase. Curr Alzheimer
thermodynamic and X-ray structural insights into the inter- Res 2008; 5:179–186.
action of melatonin and analogues with quinone reductase 2. 114. Colciaghi F, Borroni B, Pastorino L et al. [alpha]-Secre-
Biochem J 2008; 413:81–91. tase ADAM10 as well as [alpha]APPs is reduced in platelets
95. Dubocovich ML, Yun K, Al-Ghoul WM et al. Selective and CSF of Alzheimer disease patients. Mol Med 2002; 8:67–
MT2 melatonin receptor antagonists block melatonin-medi- 74.
ated phase advances of circadian rhythms. FASEB J 1998; 115. Cole SL, Vassar R. BACE1 structure and function in health
12:1211–1220. and AlzheimerÕs disease. Curr Alzheimer Res 2008; 5:100–120.
96. Hardeland R, Cardinali DP, Srinivasan V et al. Mela- 116. Xiong H, Callaghan D, Jones A et al. Cholesterol reten-
tonin-A pleiotropic, orchestrating regulator molecule. Prog tion in AlzheimerÕs brain is responsible for high beta- and
Neurobiol 2010; 93:350–358. gamma-secretase activities and Abeta production. Neurobiol
97. Reiter RJ, Tan DX, Fuentes-Broto L. Melatonin: a mul- Dis 2008; 29:422–437.
titasking molecule. Prog Brain Res 2010; 181:127–151. 117. Ehehalt R, Keller P, Haass C et al. Amyloidogenic pro-
98. Cummings JL. AlzheimerÕs disease. N Engl J Med 2004; cessing of the Alzheimer beta-amyloid precursor protein
351:56–67. depends on lipid rafts. J Cell Biol 2003; 160:113–123.
99. Aliev G, Palacios HH, Walrafen B et al. Brain mito- 118. Kitazume S, Tachida Y, Oka R et al. AlzheimerÕs beta-
chondria as a primary target in the development of treatment secretase, beta-site amyloid precursor protein-cleaving en-
strategies for Alzheimer disease. Int J Biochem Cell Biol 2009; zyme, is responsible for cleavage secretion of a Golgi-resident
41:1989–2004. sialyltransferase. Proc Natl Acad Sci USA 2001; 98:13554–
100. Bishop GM, Robinson SR. The amyloid hypothesis: let 13559.
sleeping dogmas lie? Neurobiol Aging 2002; 23:1101–1105. 119. Wolfe MS, Xia W, Ostaszewski BL et al. Two transmem-
101. Plant LD, Boyle JP, Smith IF et al. The production of brane aspartates in presenilin-1 required for presenilin endo-
amyloid beta peptide is a critical requirement for the viability proteolysis and gamma-secretase activity. Nature 1999;
of central neurons. J Neurosci 2003; 23:5531–5535. 398:513–517.

191
Rosales-Corral et al.

120. De SB, Saftig P, Craessaerts K et al. Deficiency of prese- 139. Phiel CJ, Wilson CA, Lee VM et al. GSK-3alpha regulates
nilin-1 inhibits the normal cleavage of amyloid precursor production of AlzheimerÕs disease amyloid-beta peptides.
protein. Nature 1998; 391:387–390. Nature 2003; 423:435–439.
121. Kaether C, Haass C, Steiner H. Assembly, trafficking and 140. Wang X, Zheng W, Xie JW et al. Insulin deficiency exac-
function of gamma-secretase. Neurodegener Dis 2006; 3:275– erbates cerebral amyloidosis and behavioral deficits in an
283. Alzheimer transgenic mouse model. Mol Neurodegener 2010;
122. Beel AJ, Sakakura M, Barrett PJ et al. Direct binding of 5:46–59.
cholesterol to the amyloid precursor protein: an important 141. Glabe CG. Structural classification of toxic amyloid oligo-
interaction in lipid-AlzheimerÕs disease relationships? Biochim mers. J Biol Chem 2008; 283:29639–29643.
Biophys Acta 2010; 1801:975–982. 142. Mandal PK, Pettegrew JW. AlzheimerÕs disease: soluble
123. Selkoe DJ. AlzheimerÕs disease is a synaptic failure. Science oligomeric Abeta(1–40) peptide in membrane mimic envi-
2002; 298:789–791. ronment from solution NMR and circular dichroism studies.
124. Almeida CG, Tampellini D, Takahashi RH et al. Beta- Neurochem Res 2004; 29:2267–2272.
amyloid accumulation in APP mutant neurons reduces PSD- 143. Walsh DM, Klyubin I, Fadeeva JV et al. Naturally se-
95 and GluR1 in synapses. Neurobiol Dis 2005; 20:187–198. creted oligomers of amyloid beta protein potently inhibit
125. Snyder EM, Nong Y, Almeida CG et al. Regulation of hippocampal long-term potentiation in vivo. Nature 2002;
NMDA receptor trafficking by amyloid-beta. Nat Neurosci 416:535–539.
2005; 8:1051–1058. 144. Lambert MP, Barlow AK, Chromy BA et al. Diffusible,
126. Kamenetz F, Tomita T, Hsieh H et al. APP processing and nonfibrillar ligands derived from Abeta1–42 are potent cen-
synaptic function. Neuron 2003; 37:925–937. tral nervous system neurotoxins. Proc Natl Acad Sci USA
127. Bi X, Gall CM, Zhou J et al. Uptake and pathogenic effects 1998; 95:6448–6453.
of amyloid beta peptide 1–42 are enhanced by integrin 145. Lacor PN, Buniel MC, Chang L et al. Synaptic targeting
antagonists and blocked by NMDA receptor antagonists. by AlzheimerÕs-related amyloid beta oligomers. J Neurosci
Neuroscience 2002; 112:827–840. 2004; 24:10191–10200.
128. Knauer MF, Soreghan B, Burdick D et al. Intracellular 146. El-Husseini AE, Schnell E, Chetkovich DM et al. PSD-
accumulation and resistance to degradation of the Alzheimer 95 involvement in maturation of excitatory synapses. Science
amyloid A4/beta protein. Proc Natl Acad Sci USA 1992; 2000; 290:1364–1368.
89:7437–7441. 147. Masilamoni JG, Jesudason EP, Dhandayuthapani S
129. Bayer TA, Wirths O. Intracellular accumulation of amy- et al. The neuroprotective role of melatonin against amyloid
loid-Beta – a predictor for synaptic dysfunction and neuron beta peptide injected mice. Free Radic Res 2008; 42:661–
loss in AlzheimerÕs disease. Front Aging Neurosci 2010; 2:1– 673.
10. 148. Pappolla M, Bozner P, Soto C et al. Inhibition of Alzhei-
130. Gouras GK, Tsai J, Naslund J et al. Intraneuronal mer beta-fibrillogenesis by melatonin. J Biol Chem 1998;
Abeta42 accumulation in human brain. Am J Pathol 2000; 273:7185–7188.
156:15–20. 149. Poeggeler B, Miravalle L, Zagorski MG et al. Melatonin
131. Sakono M, Zako T. Amyloid oligomers: formation and reverses the profibrillogenic activity of apolipoprotein E4 on
toxicity of Abeta oligomers. FEBS J 2010; 277:1348–1358. the Alzheimer amyloid Abeta peptide. Biochemistry (Mosc)
132. Galbete JL, Martin TR, Peressini E et al. Cholesterol 2001; 40:14995–15001.
decreases secretion of the secreted form of amyloid precursor 150. Fraser PE, Nguyen JT, Surewicz WK et al. pH-dependent
protein by interfering with glycosylation in the protein structural transitions of Alzheimer amyloid peptides. Biophys
secretory pathway. Biochem J 2000; 348(Pt 2):307–313. J 1991; 60:1190–1201.
133. Qahwash I, He W, Tomasselli A et al. Processing amyloid 151. Carter MD, Weaver DF. Ab initio molecular modeling of
precursor protein at the beta-site requires proper orientation imadazolium interaction with 5-hydroxy- and 5-methoxyin-
to be accessed by BACE1. J Biol Chem 2004; 279:39010– dole: implications for melatonin-based inhibition of Alzhei-
39016. mer [beta]-amyloid fibril formation. J Mol Struct (Theochem)
134. Riddell DR, Christie G, Hussain I et al. Compartmen- 2003; 626:279–285.
talization of beta-secretase (Asp2) into low-buoyant density, 152. Skribanek Z, Balaspiri L, Mak M. Interaction between
noncaveolar lipid rafts. Curr Biol 2001; 11:1288–1293. synthetic amyloid-beta-peptide (1–40) and its aggregation
135. Farris W, Mansourian S, Chang Y et al. Insulin-degrading inhibitors studied by electrospray ionization mass spectrom-
enzyme regulates the levels of insulin, amyloid beta-protein, etry. J Mass Spectrom 2001; 36:1226–1229.
and the beta-amyloid precursor protein intracellular domain 153. Matsubara E, Bryant-Thomas T, Pacheco QJ et al. Mel-
in vivo. Proc Natl Acad Sci USA 2003; 100:4162–4167. atonin increases survival and inhibits oxidative and amyloid
136. Qiu WQ, Walsh DM, Ye Z et al. Insulin-degrading enzyme pathology in a transgenic model of AlzheimerÕs disease.
regulates extracellular levels of amyloid beta-protein by deg- J Neurochem 2003; 85:1101–1108.
radation. J Biol Chem 1998; 273:32730–32738. 154. Song W, Lahiri DK. Melatonin alters the metabolism of the
137. Gasparini L, Gouras GK, Wang R et al. Stimulation of beta-amyloid precursor protein in the neuroendocrine cell line
beta-amyloid precursor protein trafficking by insulin reduces PC12. J Mol Neurosci 1997; 9:75–92.
intraneuronal beta-amyloid and requires mitogen-activated 155. Lahiri DK. Melatonin affects the metabolism of the beta-
protein kinase signaling. J Neurosci 2001; 21:2561–2570. amyloid precursor protein in different cell types. J Pineal Res
138. Solano DC, Sironi M, Bonfini C et al. Insulin regulates 1999; 26:137–146.
soluble amyloid precursor protein release via phosphatidyl 156. Lee RK, Knapp S, Wurtman RJ. Prostaglandin E2 stimu-
inositol 3 kinase-dependent pathway. FASEB J 2000; lates amyloid precursor protein gene expression: inhibition by
14:1015–1022. immunosuppressants. J Neurosci 1999; 19:940–947.

192
AlzheimerÕs disease and melatonin

157. Mayo JC, Sainz RM, Tan DX et al. Anti-inflammatory ac- 174. Feng Z, Chang Y, Cheng Y et al. Melatonin alleviates
tions of melatonin and its metabolites, N1-acetyl-N2-formyl- behavioral deficits associated with apoptosis and choliner-
5-methoxykynuramine (AFMK) and N1-acetyl-5-meth- gic system dysfunction in the APP 695 transgenic mouse
oxykynuramine (AMK), in macrophages. J Neuroimmunol model of AlzheimerÕs disease. J Pineal Res 2004; 37:129–
2005; 165:139–149. 136.
158. Husson I, Mesples B, Bac P et al. Melatoninergic neuro- 175. Hsiao K, Chapman P, Nilsen S et al. Correlative memory
protection of the murine periventricular white matter against deficits, Abeta elevation, and amyloid plaques in transgenic
neonatal excitotoxic challenge. Ann Neurol 2002; 51:82–92. mice. Science 1996; 274:99–102.
159. Mcarthur AJ, Hunt AE, Gillette MU. Melatonin action 176. Quinn J, Kulhanek D, Nowlin J et al. Chronic melatonin
and signal transduction in the rat suprachiasmatic circadian therapy fails to alter amyloid burden or oxidative damage in
clock: activation of protein kinase C at dusk and dawn. old Tg2576 mice: implications for clinical trials. Brain Res
Endocrinology 1997; 138:627–634. 2005; 1037:209–213.
160. Zhu G, Wang D, Lin YH et al. Protein kinase C epsilon 177. Amijee H, Madine J, Middleton DA et al. Inhibitors of
suppresses Abeta production and promotes activation of alpha- protein aggregation and toxicity. Biochem Soc Trans 2009;
secretase. Biochem Biophys Res Commun 2001; 285:997–1006. 37:692–696.
161. Kong PJ, Byun JS, Lim SY et al. Melatonin Induces Akt 178. Robinson SR, Bishop GM. Abeta as a bioflocculant: impli-
Phosphorylation through Melatonin Receptor- and PI3K- cations for the amyloid hypothesis of AlzheimerÕs disease.
Dependent Pathways in Primary Astrocytes. Korean J Physiol Neurobiol Aging 2002; 23:1051–1072.
Pharmacol 2008; 12:37–41. 179. Hasegawa T. Prolonged stress will induce AlzheimerÕs dis-
162. Duarte AI, Santos P, Oliveira CR et al. Insulin neuro- ease in elderly people by increased release of homocysteic
protection against oxidative stress is mediated by Akt and acid. Med Hypotheses 2007; 69:1135–1139.
GSK-3beta signaling pathways and changes in protein 180. Clarke R, Smith AD, Jobst KA et al. Folate, vitamin B12,
expression. Biochim Biophys Acta 2008; 1783:994–1002. and serum total homocysteine levels in confirmed Alzheimer
163. Chetsawang B, Govitrapong P, Ebadi M. The neuropro- disease. Arch Neurol 1998; 55:1449–1455.
tective effect of melatonin against the induction of c-Jun 181. Ganapathy PS, Moister B, Roon P et al. Endogenous ele-
phosphorylation by 6-hydroxydopamine on SK-N-SH cells. vation of homocysteine induces retinal neuron death in the
Neurosci Lett 2004; 371:205–208. cystathionine-beta-synthase mutant mouse. Invest Ophthal-
164. Rosales-Corral S, Reiter RJ, Tan DX et al. Functional mol Vis Sci 2009; 50:4460–4470.
aspects of redox control during neuroinflammation. Antioxid 182. Baydas G, Reiter RJ, Akbulut M et al. Melatonin inhibits
Redox Signal 2010; 13:193–247. neural apoptosis induced by homocysteine in hippocampus of
165. Biessels GJ, Kappelle LJ. Increased risk of AlzheimerÕs rats via inhibition of cytochrome c translocation and caspase-
disease in Type II diabetes: insulin resistance of the brain or 3 activation and by regulating pro- and anti-apoptotic protein
insulin-induced amyloid pathology? Biochem Soc Trans 2005; levels. Neuroscience 2005; 135:879–886.
33:1041–1044. 183. Bartus RT, Dean RL III, Beer B et al. The cholinergic
166. Hoppe JB, Frozza RL, Horn AP et al. Amyloid-beta neu- hypothesis of geriatric memory dysfunction. Science 1982;
rotoxicity in organotypic culture is attenuated by melatonin: 217:408–414.
involvement of GSK-3beta, tau and neuroinflammation. 184. Francis PT, Palmer AM, Snape M et al. The cholinergic
J Pineal Res 2010; 48:230–238. hypothesis of AlzheimerÕs disease: a review of progress.
167. Zhang X, Zhou K, Wang R et al. Hypoxia-inducible factor J Neurol Neurosurg Psychiatry 1999; 66:137–147.
1alpha (HIF-1alpha)-mediated hypoxia increases BACE1 185. Spencer JP, Middleton LJ, Davies CH. Investigation into
expression and beta-amyloid generation. J Biol Chem 2007; the efficacy of the acetylcholinesterase inhibitor, donepezil,
282:10873–10880. and novel procognitive agents to induce gamma oscillations in
168. Yasinska IM, Sumbayev VV. S-nitrosation of Cys-800 of rat hippocampal slices. Neuropharmacology 2010; 59:437–
HIF-1alpha protein activates its interaction with p300 and 443.
stimulates its transcriptional activity. FEBS Lett 2003; 186. Guermonprez L, Ducrocq C, Gaudry-Talarmain YM.
549:105–109. Inhibition of acetylcholine synthesis and tyrosine nitration
169. Grammas P, Samany PG, Thirumangalakudi L. Throm- induced by peroxynitrite are differentially prevented by an-
bin and inflammatory proteins are elevated in AlzheimerÕs tioxidants. Mol Pharmacol 2001; 60:838–846.
disease microvessels: implications for disease pathogenesis. 187. Pedersen WA, Kloczewiak MA, Blusztajn JK. Amyloid
J Alzheimers Dis 2006; 9:51–58. beta-protein reduces acetylcholine synthesis in a cell line de-
170. Soucek T, Cumming R, Dargusch R et al. The regulation rived from cholinergic neurons of the basal forebrain. Proc
of glucose metabolism by HIF-1 mediates a neuroprotective Natl Acad Sci USA 1996; 93:8068–8071.
response to amyloid beta peptide. Neuron 2003; 39:43–56. 188. Mattson MP. Metal-catalyzed disruption of membrane
171. Song H, Sinner EK, Knoll W. Peptid-tethered bilayer lipid protein and lipid signaling in the pathogenesis of neurode-
membranes and their interaction with Amyloid beta-peptide. generative disorders. Ann NY Acad Sci 2004; 1012:37–50.
Biointerphases 2007; 2:151–158. 189. Schweitzer ES. Regulated and constitutive secretion of dis-
172. Song H, Ritz S, Knoll W et al. Conformation and topology tinct molecular forms of acetylcholinesterase from PC12 cells.
of amyloid beta-protein adsorbed on a tethered artificial J Cell Sci 1993; 106(Pt 3):731–740.
membrane probed by surface plasmon field-enhanced fluo- 190. Sberna G, Saez-Valero J, Beyreuther K et al. The amy-
rescence spectroscopy. J Struct Biol 2009; 168:117–124. loid beta-protein of AlzheimerÕs disease increases acetylcho-
173. Lahiri DK, Chen D, Ge YW et al. Dietary supplementation linesterase expression by increasing intracellular calcium in
with melatonin reduces levels of amyloid beta-peptides in the embryonal carcinoma P19 cells. J Neurochem 1997; 69:1177–
murine cerebral cortex. J Pineal Res 2004; 36:224–231. 1184.

193
Rosales-Corral et al.

191. Melo JB, Agostinho P, Oliveira CR. Involvement of oxi- 210. Martins RN, Harper CG, Stokes GB et al. Increased
dative stress in the enhancement of acetylcholinesterase cerebral glucose-6-phosphate dehydrogenase activity in Alz-
activity induced by amyloid beta-peptide. Neurosci Res 2003; heimerÕs disease may reflect oxidative stress. J Neurochem
45:117–127. 1986; 46:1042–1045.
192. Tyagi E, Agrawal R, Nath C et al. Effect of melatonin 211. Bradley MA, Markesbery WR, Lovell MA. Increased
on neuroinflammation and acetylcholinesterase activity in- levels of 4-hydroxynonenal and acrolein in the brain in pre-
duced by LPS in rat brain. Eur J Pharmacol 2010; 640:206– clinical Alzheimer disease. Free Radic Biol Med 2010;
210. 48:1570–1576.
193. Wu J, Anwyl R, Rowan MJ. beta-Amyloid selectively aug- 212. Reed TT, Pierce WM Jr, Turner DM et al. Proteomic
ments NMDA receptor-mediated synaptic transmission in rat identification of nitrated brain proteins in early AlzheimerÕs
hippocampus. Neuroreport 1995; 6:2409–2413. disease inferior parietal lobule. J Cell Mol Med 2009;
194. Peng TI, Jou MJ, Sheu SS et al. Visualization of NMDA 13:2019–2029.
receptor-induced mitochondrial calcium accumulation in 213. Mattson MP, Rydel RE. AlzheimerÕs disease. Amyloid
striatal neurons. Exp Neurol 1998; 149:1–12. ox–tox transducers. Nature 1996; 382:674–675.
195. Zhu H, Gao W, Jiang H et al. Regulation of acetylcholin- 214. Rosales-Corral S, Tan DX, Reiter RJ et al. Kinetics of
esterase expression by calcium signaling during calcium ion- the neuroinflammation-oxidative stress correlation in rat
ophore A2. Int J Biochem Cell Biol 2007; 39:93–108. brain following the injection of fibrillar amyloid-beta onto the
196. Zhang QZ, Zhang JT. Inhibitory effects of melatonin on free hippocampus in vivo. J Neuroimmunol 2004; 150:20–28.
intracellular calcium in mouse brain cells. Zhongguo Yao Li 215. Weldon DT, Rogers SD, Ghilardi JR et al. Fibrillar beta-
Xue Bao 1999; 20:206–210. amyloid induces microglial phagocytosis, expression of
197. Siwicka A, Moleda Z, Wojtasiewicz K et al. The oxidation inducible nitric oxide synthase, and loss of a select population
products of melatonin derivatives exhibit acetylcholinesterase of neurons in the rat CNS in vivo. J Neurosci 1998; 18:2161–
and butyrylcholinesterase inhibitory activity. J Pineal Res 2173.
2008; 45:40–49. 216. Yan SD, Chen X, Fu J et al. RAGE and amyloid-beta
198. Agrawal R, Tyagi E, Shukla R et al. Effect of insulin and peptide neurotoxicity in AlzheimerÕs disease. Nature 1996;
melatonin on acetylcholinesterase activity in the brain of 382:685–691.
amnesic mice. Behav Brain Res 2008; 189:381–386. 217. Serrano F, Chang A, Hernandez C et al. NADPH oxidase
199. Smith MA, Richey Harris PL, Sayre LM et al. Widespread mediates beta-amyloid peptide-induced activation of ERK in
peroxynitrite-mediated damage in AlzheimerÕs disease. hippocampal organotypic cultures. Mol Brain 2009; 2:31–41.
J Neurosci 1997; 17:2653–2657. 218. Lecanu L, Greeson J, Papadopoulos V. Beta-amyloid and
200. Davies P. Challenging the cholinergic hypothesis in Alzhei- oxidative stress jointly induce neuronal death, amyloid
mer disease. JAMA 1999; 281:1433–1434. deposits, gliosis, and memory impairment in the rat brain.
201. Pinthong M, Black SA, Ribeiro FM et al. Activity and Pharmacology 2006; 76:19–33.
subcellular trafficking of the sodium-coupled choline trans- 219. Shelat PB, Chalimoniuk M, Wang JH et al. Amyloid beta
porter CHT is regulated acutely by peroxynitrite. Mol Phar- peptide and NMDA induce ROS from NADPH oxidase and
macol 2008; 73:801–812. AA release from cytosolic phospholipase A2 in cortical neu-
202. Wang JZ, Wang ZF. Role of melatonin in Alzheimer-like rons. J Neurochem 2008; 106:45–55.
neurodegeneration. Acta Pharmacol Sin 2006; 27:41–49. 220. Roher AE, Weiss N, Kokjohn TA et al. Increased A beta
203. Butterfield DA, Lauderback CM. Lipid peroxidation and peptides and reduced cholesterol and myelin proteins char-
protein oxidation in AlzheimerÕs disease brain: potential acterize white matter degeneration in AlzheimerÕs disease.
causes and consequences involving amyloid beta-peptide- Biochemistry (Mosc) 2002; 41:11080–11090.
associated free radical oxidative stress. Free Radic Biol Med 221. Jekabsone A, Mander PK, Tickler A et al. Fibrillar beta-
2002; 32:1050–1060. amyloid peptide Abeta1–40 activates microglial proliferation
204. Uchida K, Shiraishi M, Naito Y et al. Activation of stress via stimulating TNF-alpha release and H2O2 derived from
signaling pathways by the end product of lipid peroxidation. NADPH oxidase: a cell culture study. J Neuroinflammation
4-hydroxy-2-nonenal is a potential inducer of intracellular 2006; 3:24–37.
peroxide production. J Biol Chem 1999; 274:2234–2242. 222. Shimohama S, Tanino H, Kawakami N et al. Activation of
205. Tang F, Nag S, Shiu SY et al. The effects of melatonin and NADPH oxidase in AlzheimerÕs disease brains. Biochem
Ginkgo biloba extract on memory loss and choline acetyl- Biophys Res Commun 2000; 273:5–9.
transferase activities in the brain of rats infused intracere- 223. Pacher P, Beckman JS, Liaudet L. Nitric oxide and perox-
broventricularly with beta-amyloid 1-40. Life Sci 2002; ynitrite in health and disease. Physiol Rev 2007; 87:315–424.
71:2625–2631. 224. Sorce S, Krause KH. NOX enzymes in the central nervous
206. Hardy JA, Higgins GA. AlzheimerÕs disease: the amyloid system: from signaling to disease. Antioxid Redox Signal
cascade hypothesis. Science 1992; 256:184–185. 2009; 11:2481–2504.
207. Wang JY, Wen LL, Huang YN et al. Dual effects of an- 225. Harraz MM, Marden JJ, Zhou W et al. SOD1 mutations
tioxidants in neurodegeneration: direct neuroprotection disrupt redox-sensitive Rac regulation of NADPH oxidase in
against oxidative stress and indirect protection via suppres- a familial ALS model. J Clin Invest 2008; 118:659–670.
sion of glia-mediated inflammation. Curr Pharm Des 2006; 226. Melov S, Adlard PA, Morten K et al. Mitochondrial
12:3521–3533. oxidative stress causes hyperphosphorylation of tau. PLoS
208. Delegge MH, Smoke A. Neurodegeneration and inflam- ONE 2007; 2:e536–e548.
mation. Nutr Clin Pract 2008; 23:35–41. 227. Takuma K, Yao J, Huang J et al. ABAD enhances Abeta-
209. Golden TR, Patel M. Catalytic antioxidants and neurode- induced cell stress via mitochondrial dysfunction. FASEB J
generation. Antioxid Redox Signal 2009; 11:555–570. 2005; 19:597–598.

194
AlzheimerÕs disease and melatonin

228. Yao J, Irwin RW, Zhao L et al. Mitochondrial bioenergetic 245. Foy CJ, Passmore AP, Vahidassr MD et al. Plasma chain-
deficit precedes AlzheimerÕs pathology in female mouse model breaking antioxidants in AlzheimerÕs disease, vascular
of AlzheimerÕs disease. Proc Natl Acad Sci USA 2009; dementia and ParkinsonÕs disease. Q J Med 1999; 92:39–45.
106:14670–14675. 246. Streit WJ, Mrak RE, Griffin WS. Microglia and neuro-
229. Hsu MJ, Sheu JR, Lin CH et al. Mitochondrial mechanisms inflammation: a pathological perspective. J Neuroinflamma-
in amyloid beta peptide-induced cerebrovascular degenera- tion 2004; 1:14–18.
tion. Biochim Biophys Acta 2010; 1800:290–296. 247. Akiyama H, Barger S, Barnum S et al. Inflammation and
230. Misonou H, Morishima-Kawashima M, Ihara Y. Oxida- AlzheimerÕs disease. Neurobiol Aging 2000; 21:383–421.
tive stress induces intracellular accumulation of amyloid beta- 248. Shaftel SS, Kyrkanides S, Olschowka JA et al. Sustained
protein (Abeta) in human neuroblastoma cells. Biochemistry hippocampal IL-1 beta overexpression mediates chronic
(Mosc) 2000; 39:6951–6959. neuroinflammation and ameliorates Alzheimer plaque
231. Butterfield DA, Boyd-Kimball D. The critical role of pathology. J Clin Invest 2007; 117:1595–1604.
methionine 35 in AlzheimerÕs amyloid beta-peptide (1–42)- 249. Bianchi ME. DAMPs, PAMPs and alarmins: all we need to
induced oxidative stress and neurotoxicity. Biochim Biophys know about danger. J Leukoc Biol 2007; 81:1–5.
Acta 2005; 1703:149–156. 250. Pais TF, Figueiredo C, Peixoto R et al. Necrotic neurons
232. Pogocki D, Schoneich C. Redox properties of Met(35) in enhance microglial neurotoxicity through induction of gluta-
neurotoxic beta-amyloid peptide. A molecular modeling minase by a MyD88-dependent pathway. J Neuroinflamma-
study. Chem Res Toxicol 2002; 15:408–418. tion 2008; 5:43.
233. Butterfield DA, Reed T, Newman SF et al. Roles of 251. Atlante A, Calissano P, Bobba A et al. Glutamate neu-
amyloid beta-peptide-associated oxidative stress and brain rotoxicity, oxidative stress and mitochondria. FEBS Lett
protein modifications in the pathogenesis of AlzheimerÕs dis- 2001; 497:1–5.
ease and mild cognitive impairment. Free Radic Biol Med 252. Brookes PS. Mitochondrial H(+) leak and ROS generation:
2007; 43:658–677. an odd couple. Free Radic Biol Med 2005; 38:12–23.
234. Huang X, Atwood CS, Hartshorn MA et al. The A beta 253. He P, Zhong Z, Lindholm K et al. Deletion of tumor
peptide of AlzheimerÕs disease directly produces hydrogen necrosis factor death receptor inhibits amyloid beta genera-
peroxide through metal ion reduction. Biochemistry (Mosc) tion and prevents learning and memory deficits in AlzheimerÕs
1999; 38:7609–7616. mice. J Cell Biol 2007; 178:829–841.
235. Rival T, Page RM, Chandraratna DS et al. Fenton 254. Jana M, Palencia CA, Pahan K. Fibrillar amyloid-beta
chemistry and oxidative stress mediate the toxicity of the beta- peptides activate microglia via TLR2: implications for Alz-
amyloid peptide in a Drosophila model of AlzheimerÕs dis- heimerÕs disease. J Immunol 2008; 181:7254–7262.
ease. Eur J Neurosci 2009; 29:1335–1347. 255. Tahara K, Kim HD, Jin JJ et al. Role of toll-like receptor
236. Deibel MA, Ehmann WD, Markesbery WR. Copper, iron, signalling in Abeta uptake and clearance. Brain 2006;
and zinc imbalances in severely degenerated brain regions in 129:3006–3019.
AlzheimerÕs disease: possible relation to oxidative stress. 256. Salminen A, Suuronen T, Kaarniranta K. ROCK, PAK,
J Neurol Sci 1996; 143:137–142. and Toll of synapses in AlzheimerÕs disease. Biochem Biophys
237. Lovell MA, Robertson JD, Teesdale WJ et al. Copper, Res Commun 2008; 371:587–590.
iron and zinc in AlzheimerÕs disease senile plaques. J Neurol 257. Moore KJ, El KJ, Medeiros LA et al. A CD36-initiated
Sci 1998; 158:47–52. signaling cascade mediates inflammatory effects of beta-
238. Sims NR, Anderson MF, Hobbs LM et al. Impairment of amyloid. J Biol Chem 2002; 277:47373–47379.
brain mitochondrial function by hydrogen peroxide. Brain 258. Brandenburg LO, Konrad M, Wruck CJ et al. Functional
Res Mol Brain Res 2000; 77:176–184. and physical interactions between formyl-peptide-receptors
239. Reiter RJ, Tan DX, Manchester LC et al. Melatonin and scavenger receptor MARCO and their involvement in
reduces oxidant damage and promotes mitochondrial respi- amyloid beta 1–42-induced signal transduction in glial cells.
ration: implications for aging. Ann NY Acad Sci 2002; J Neurochem 2010; 113:749–760.
959:238–250. 259. Lue LF, Walker DG, Jacobson S et al. Receptor for ad-
240. Milton NG. Amyloid-beta binds catalase with high affinity vanced glycation end products: its role in AlzheimerÕs disease
inhibits hydrogen peroxide breakdown. Biochem J 1999; and other neurological diseases. Future Neurol 2009; 4:167–
344(Pt 2):293–296. 177.
241. Massaad CA, Washington TM, Pautler RG et al. Over- 260. Bianchi R, Giambanco I, Donato R. S100B/RAGE-
expression of SOD-2 reduces hippocampal superoxide and dependent activation of microglia via NF-kappaB and AP-1
prevents memory deficits in a mouse model of AlzheimerÕs Co-regulation of COX-2 expression by S100B, IL-1beta and
disease. Proc Natl Acad Sci USA 2009; 106:13576–13581. TNF-alpha. Neurobiol Aging 2010; 31:665–677.
242. Holcomb L, Gordon MN, Mcgowan E et al. Accelerated 261. Li Y, Barger SW, Liu L et al. S100beta induction of the
Alzheimer-type phenotype in transgenic mice carrying both proinflammatory cytokine interleukin-6 in neurons. J Neu-
mutant amyloid precursor protein and presenilin 1 transgenes. rochem 2000; 74:143–150.
Nat Med 1998; 4:97–100. 262. Bamberger ME, Harris ME, Mcdonald DR et al. A cell
243. Resende R, Moreira PI, Proenca T et al. Brain oxidative surface receptor complex for fibrillar beta-amyloid mediates
stress in a triple-transgenic mouse model of Alzheimer dis- microglial activation. J Neurosci 2003; 23:2665–2674.
ease. Free Radic Biol Med 2008; 44:2051–2057. 263. Gupta SC, Sundaram C, Reuter S et al. Inhibiting
244. Oddo S, Caccamo A, Shepherd JD et al. Triple-transgenic NF-kappaB activation by small molecules as a therapeutic
model of AlzheimerÕs disease with plaques and tangles: strategy. Biochim Biophys Acta 2010; 1799:775–787.
intracellular Abeta and synaptic dysfunction. Neuron 2003; 264. Barger SW, Horster D, Furukawa K et al. Tumor
39:409–421. necrosis factors alpha and beta protect neurons against

195
Rosales-Corral et al.

amyloid beta-peptide toxicity: evidence for involvement of a citotoxicity and oxidative stress. J Pineal Res 2008; 45:117–
kappa B-binding factor and attenuation of peroxide and 124.
Ca2+ accumulation. Proc Natl Acad Sci USA 1995; 92:9328– 283. Kim YS, Kim H, Sung YH et al. The effect of melatonin on
9332. glutamate- and its subtype agonists- induced ion currents in
265. Kaltschmidt B, Baeuerle PA, Kaltschmidt C. Potential rat hippocampal CA1 neurons. FASEB J 2007; 21:A1277–
involvement of the transcription factor NF-kappa B in neu- A127c.
rological disorders. Mol Aspects Med 1993; 14:171–190. 284. Zhang QZ, Gong YS, Zhang JT. Antagonistic effects
266. Bianca VD, Dusi S, Bianchini E et al. beta-amyloid acti- of melatonin on glutamate release and neurotoxicity in
vates the O-2 forming NADPH oxidase in microglia, mono- cerebral cortex. Zhongguo Yao Li Xue Bao 1999; 20:829–
cytes, and neutrophils. A possible inflammatory mechanism 834.
of neuronal damage in AlzheimerÕs disease. J Biol Chem 1999; 285. Rosales-Corral S, Tan DX, Reiter RJ et al. Orally
274:15493–15499. administered melatonin reduces oxidative stress and proin-
267. Chen C, Bazan NG. Endogenous PGE2 regulates membrane flammatory cytokines induced by amyloid-beta peptide in rat
excitability and synaptic transmission in hippocampal CA1 brain: a comparative, in vivo study versus vitamin C and E.
pyramidal neurons. J Neurophysiol 2005; 93:929–941. J Pineal Res 2003; 35:80–84.
268. Del RP, Montiel T, Massieu L. Contribution of NMDA 286. Natarajan M, Sadeghi K, Reiter RJ et al. The neuro-
and non-NMDA receptors to in vivo glutamate-induced cal- hormone melatonin inhibits cytokine, mitogen and ionizing
pain activation in the rat striatum. Relation to neuronal radiation induced NF-kappa B. Biochem Mol Biol Int 1995;
damage. Neurochem Res 2008; 33:1475–1483. 37:1063–1070.
269. Lau A, Tymianski M. Glutamate receptors, neurotoxicity 287. Chuang JI, Natarajan M, Meltz ML et al. Effect of mel-
and neurodegeneration. Pflugers Arch 2010; 460:525–542. atonin on NF-kappa-B DNA-binding activity in the rat
270. Zhou J, Zhang S, Zhao X et al. Melatonin impairs NADPH spleen. Cell Biol Int 1996; 20:687–692.
oxidase assembly and decreases superoxide anion production 288. Clapp-Lilly KL, Smith MA, Perry G et al. Melatonin
in microglia exposed to amyloid-beta1–42. J Pineal Res 2008; reduces interleukin secretion in amyloid-beta stressed mouse
45:157–165. brain slices. Chem Biol Interact 2001; 134:101–107.
271. Lahair MM, Howe CJ, Rodriguez-Mora O et al. Molec- 289. Shen Y, Zhang G, Liu L et al. Suppressive effects of mela-
ular pathways leading to oxidative stress-induced phosphor- tonin on amyloid-beta-induced glial activation in rat hippo-
ylation of Akt. Antioxid Redox Signal 2006; 8:1749–1756. campus. Arch Med Res 2007; 38:284–290.
272. Feng Z, Zhang JT. Long-term melatonin or 17beta-estradiol 290. Fraser DA, Arora M, Bohlson SS et al. Generation of
supplementation alleviates oxidative stress in ovariectomized inhibitory NFkappaB complexes and phosphorylated cAMP
adult rats. Free Radic Biol Med 2005; 39:195–204. response element-binding protein correlates with the anti-
273. Leon J, Escames G, Rodriguez MI et al. Inhibition of inflammatory activity of complement protein C1q in human
neuronal nitric oxide synthase activity by N1-acetyl-5-meth- monocytes. J Biol Chem 2007; 282:7360–7367.
oxykynuramine, a brain metabolite of melatonin. J Neuro- 291. Afagh A, Cummings BJ, Cribbs DH et al. Localization and
chem 2006; 98:2023–2033. cell association of C1q in AlzheimerÕs disease brain. Exp
274. Dewitt DA, Perry G, Cohen M et al. Astrocytes regulate Neurol 1996; 138:22–32.
microglial phagocytosis of senile plaque cores of AlzheimerÕs 292. Mattson MP, Camandola S. NF-kappaB in neuronal
disease. Exp Neurol 1998; 149:329–340. plasticity and neurodegenerative disorders. J Clin Invest 2001;
275. Boche D, Cunningham C, Docagne F et al. TGFbeta1 107:247–254.
regulates the inflammatory response during chronic neu- 293. Deng WG, Tang ST, Tseng HP et al. Melatonin suppresses
rodegeneration. Neurobiol Dis 2006; 22:638–650. macrophage cyclooxygenase-2 and inducible nitric oxide
276. Gee JR, Keller JN. Astrocytes: regulation of brain synthase expression by inhibiting p52 acetylation and binding.
homeostasis via apolipoprotein E. Int J Biochem Cell Biol Blood 2006; 108:518–524.
2005; 37:1145–1150. 294. Sasaki M, Jordan P, Joh T et al. Melatonin reduces TNF-a
277. Chow SK, Yu D, Macdonald CL et al. Amyloid beta- induced expression of MAdCAM-1 via inhibition of NF-
peptide directly induces spontaneous calcium transients, kappaB. BMC Gastroenterol 2002; 2:9–14.
delayed intercellular calcium waves and gliosis in rat cortical 295. Beni SM, Kohen R, Reiter RJ et al. Melatonin-induced
astrocytes. ASN Neuro 2010; 2:e00026. neuroprotection after closed head injury is associated with
278. Halle A, Hornung V, Petzold GC et al. The NALP3 increased brain antioxidants and attenuated late-phase
inflammasome is involved in the innate immune response to activation of NF-kappaB and AP-1. FASEB J 2004; 18:149–
amyloid-beta. Nat Immunol 2008; 9:857–865. 151.
279. Schipke CG, Boucsein C, Ohlemeyer C et al. Astrocyte 296. Talaei SA, Sheibani V, Salami M. Light deprivation
Ca2+ waves trigger responses in microglial cells in brain improves melatonin related suppression of hippocampal
slices. FASEB J 2002; 16:255–257. plasticity. Hippocampus 2010; 20:447–455.
280. Peters JL, Earnest BJ, Tjalkens RB et al. Modulation of 297. Zeise ML, Semm P. Melatonin lowers excitability of guinea
intercellular calcium signaling by melatonin in avian and pig hippocampal neurons in vitro. J Comp Physiol A 1985;
mammalian astrocytes is brain region-specific. J Comp Neu- 157:23–29.
rol 2005; 493:370–380. 298. Musshoff U, Riewenherm D, Berger E et al. Melatonin
281. Peters JL, Cassone VM, Zoran MJ. Melatonin modulates receptors in rat hippocampus: molecular and functional
intercellular communication among cultured chick astrocytes. investigations. Hippocampus 2002; 12:165–173.
Brain Res 2005; 1031:10–19. 299. Larson J, Jessen RE, Uz T et al. Impaired hippocampal
282. Das A, Belagodu A, Reiter RJ et al. Cytoprotective effects long-term potentiation in melatonin MT2 receptor-deficient
of melatonin on C6 astroglial cells exposed to glutamate ex- mice. Neurosci Lett 2006; 393:23–26.

196
AlzheimerÕs disease and melatonin

300. El-Sherif Y, Tesoriero J, Hogan MV et al. Melatonin 319. Clementi ME, Marini S, Coletta M et al. Abeta(31–35)
regulates neuronal plasticity in the hippocampus. J Neurosci and Abeta(25–35) fragments of amyloid beta-protein induce
Res 2003; 72:454–460. cellular death through apoptotic signals: Role of the redox
301. Adachi A, Natesan AK, Whitfield-Rucker MG et al. state of methionine-35. FEBS Lett 2005; 579:2913–2918.
Functional melatonin receptors and metabolic coupling in 320. Aliev G, Seyidova D, Lamb BT et al. Mitochondria and
cultured chick astrocytes. Glia 2002; 39:268–278. vascular lesions as a central target for the development of
302. Steinhilber D, Brungs M, Werz O et al. The nuclear AlzheimerÕs disease and Alzheimer disease-like pathology in
receptor for melatonin represses 5-lipoxygenase gene expres- transgenic mice. Neurol Res 2003; 25:665–674.
sion in human B lymphocytes. J Biol Chem 1995; 270:7037– 321. Alvarez G, Ramos M, Ruiz F et al. Pyruvate protection
7040. against beta-amyloid-induced neuronal death: role of mito-
303. Becker-Andre M, Wiesenberg I, Schaeren-Wiemers N chondrial redox state. J Neurosci Res 2003; 73:260–269.
et al. Pineal gland hormone melatonin binds and activates an 322. Lustbader JW, Cirilli M, Lin C et al. ABAD directly links
orphan of the nuclear receptor superfamily. J Biol Chem Abeta to mitochondrial toxicity in AlzheimerÕs disease. Sci-
1994; 269:28531–28534. ence 2004; 304:448–452.
304. Wiesenberg I, Missbach M, Kahlen JP et al. Transcrip- 323. Lin H, Bhatia R, Lal R. Amyloid beta protein forms ion
tional activation of the nuclear receptor RZR alpha by the channels: implications for AlzheimerÕs disease pathophysiol-
pineal gland hormone melatonin and identification of CGP ogy. FASEB J 2001; 15:2433–2444.
52608 as a synthetic ligand. Nucleic Acids Res 1995; 23:327– 324. Parks JK, Smith TS, Trimmer PA et al. Neurotoxic Abeta
333. peptides increase oxidative stress in vivo through NMDA-
305. Chinnici CM, Yao Y, Pratico D. The 5-lipoxygenase receptor and nitric-oxide-synthase mechanisms, and inhibit
enzymatic pathway in the mouse brain: young versus old. complex IV activity and induce a mitochondrial permeability
Neurobiol Aging 2007; 28:1457–1462. transition in vitro. J Neurochem 2001; 76:1050–1056.
306. Firuzi O, Zhuo J, Chinnici CM et al. 5-Lipoxygenase gene 325. Canevari L, Clark JB, Bates TE. beta-Amyloid fragment
disruption reduces amyloid-beta pathology in a mouse model 25–35 selectively decreases complex IV activity in isolated
of AlzheimerÕs disease. FASEB J 2008; 22:1169–1178. mitochondria. FEBS Lett 1999; 457:131–134.
307. Shafer LL, Mcnulty JA, Young MR. Assessment of mel- 326. Hansson Petersen CA, Alikhani N, Behbahani H et al.
atoninÕs ability to regulate cytokine production by macro- The amyloid beta-peptide is imported into mitochondria via
phage and microglia cell types. J Neuroimmunol 2001; the TOM import machinery and localized to mitochondrial
120:84–93. cristae. Proc Natl Acad Sci USA 2008; 105:13145–13150.
308. Negi G, Kumar A, Sharma SS. Melatonin modulates 327. Bonda DJ, Wang X, Perry G et al. Mitochondrial dynamics
neuroinflammation and oxidative stress in experimental dia- in AlzheimerÕs disease: opportunities for future treatment
betic neuropathy: effects on NF-kappaB and Nrf2 cascades. strategies. Drugs Aging 2010; 27:181–192.
J Pineal Res 2010; 50:124–131. 328. Wang X, Su B, Lee HG et al. Impaired balance of mito-
309. Chung SY, Han SH. Melatonin attenuates kainic acid-in- chondrial fission and fusion in AlzheimerÕs disease. J Neurosci
duced hippocampal neurodegeneration and oxidative stress 2009; 29:9090–9103.
through microglial inhibition. J Pineal Res 2003; 34:95–102. 329. Cho DH, Nakamura T, Fang J et al. S-nitrosylation of
310. Zhu X, Raina AK, Perry G et al. AlzheimerÕs disease: the Drp1 mediates beta-amyloid-related mitochondrial fission
two-hit hypothesis. Lancet Neurol 2004; 3:219–226. and neuronal injury. Science 2009; 324:102–105.
311. Swerdlow RH, Khan SM. The AlzheimerÕs disease mito- 330. Mattson MP, Liu D. Energetics and oxidative stress in
chondrial cascade hypothesis: an update. Exp Neurol 2009; synaptic plasticity and neurodegenerative disorders. Neuro-
218:308–315. molecular Med 2002; 2:215–231.
312. Perry EK, Perry RH, Tomlinson BE et al. Coenzyme A- 331. Mancuso M, Calsolaro V, Orsucci D et al. Is there a
acetylating enzymes in AlzheimerÕs disease: possible cholin- primary role of the mitochondrial genome in AlzheimerÕs
ergic ÔcompartmentÕ of pyruvate dehydrogenase. Neurosci disease? J Bioenerg Biomembr 2009; 41:411–416.
Lett 1980; 18:105–110. 332. Kujoth GC, Hiona A, Pugh TD et al. Mitochondrial DNA
313. Sheu KF, Kim YT, Blass JP et al. An immunochemical mutations, oxidative stress, and apoptosis in mammalian
study of the pyruvate dehydrogenase deficit in AlzheimerÕs aging. Science 2005; 309:481–484.
disease brain. Ann Neurol 1985; 17:444–449. 333. Swerdlow RH, Khan SM. A Ômitochondrial cascade
314. Parker WD Jr, Filley CM, Parks JK. Cytochrome oxidase hypothesisÕ for sporadic AlzheimerÕs disease. Med Hypotheses
deficiency in AlzheimerÕs disease. Neurology 1990; 40:1302– 2004; 63:8–20.
1303. 334. Rodrigues CM, Sola S, Brito MA et al. Amyloid beta-
315. Hardy J, Selkoe DJ. The amyloid hypothesis of AlzheimerÕs peptide disrupts mitochondrial membrane lipid and protein
disease: progress and problems on the road to therapeutics. structure: protective role of tauroursodeoxycholate. Biochem
Science 2002; 297:353–356. Biophys Res Commun 2001; 281:468–474.
316. Hardy J, Allsop D. Amyloid deposition as the central event 335. Mancuso M, Orsucci D, Siciliano G et al. Mitochondria,
in the aetiology of AlzheimerÕs disease. Trends Pharmacol Sci mitochondrial DNA and AlzheimerÕs disease. What comes
1991; 12:383–388. first? Curr Alzheimer Res 2008; 5:457–468.
317. Caspersen C, Wang N, Yao J et al. Mitochondrial Abeta: a 336. Du H, Guo L, Fang F et al. Cyclophilin D deficiency
potential focal point for neuronal metabolic dysfunction in attenuates mitochondrial and neuronal perturbation and
AlzheimerÕs disease. FASEB J 2005; 19:2040–2041. ameliorates learning and memory in AlzheimerÕs disease. Nat
318. Gao X, Zheng CY, Yang L et al. Huperzine A protects Med 2008; 14:1097–1105.
isolated rat brain mitochondria against beta-amyloid peptide. 337. Connern CP, Halestrap AP. Recruitment of mitochondrial
Free Radic Biol Med 2009; 46:1454–1462. cyclophilin to the mitochondrial inner membrane under

197
Rosales-Corral et al.

conditions of oxidative stress that enhance the opening of a 355. Ling X, Zhang LM, Lu SD et al. Protective effect of mela-
calcium-sensitive non-specific channel. Biochem J 1994; tonin on injured cerebral neurons is associated with bcl-2
302(Suppl 2):321–324. protein over-expression. Zhongguo Yao Li Xue Bao 1999;
338. Ichas F, Mazat JP. From calcium signaling to cell death: 20:409–414.
two conformations for the mitochondrial permeability tran- 356. Paradis E, Douillard H, Koutroumanis M et al. Amyloid
sition pore. Switching from low- to high-conductance state. beta peptide of AlzheimerÕs disease downregulates Bcl-2 and
Biochim Biophys Acta 1998; 1366:33–50. upregulates bax expression in human neurons. J Neurosci
339. Martinez-Cano E, Ortiz-Genaro G, Pacheco-Moises F 1996; 16:7533–7539.
et al. [Functional disorders of FOF1-ATPase in submito- 357. Jang MH, Jung SB, Lee MH et al. Melatonin attenuates
chondrial particles obtained from platelets of patients with a amyloid beta25–35-induced apoptosis in mouse microglial
diagnosis of probable AlzheimerÕs disease]. Rev Neurol 2005; BV2 cells. Neurosci Lett 2005; 380:26–31.
40:81–85. 358. OÕbarr S, Schultz J, Rogers J. Expression of the pro-
340. Mark RJ, Hensley K, Butterfield DA et al. Amyloid tooncogene bcl-2 in AlzheimerÕs disease brain. Neurobiol
beta-peptide impairs ion-motive ATPase activities: evidence Aging 1996; 17:131–136.
for a role in loss of neuronal Ca2+ homeostasis and cell 359. Juknat AA, Mendez MV, Quaglino A et al. Melatonin
death. J Neurosci 1995; 15:6239–6249. prevents hydrogen peroxide-induced Bax expression in cul-
341. Bosetti F, Brizzi F, Barogi S et al. Cytochrome c oxidase tured rat astrocytes. J Pineal Res 2005; 38:84–92.
and mitochondrial F1F0-ATPase (ATP synthase) activities in 360. Wisessmith W, Phansuwan-Pujito P, Govitrapong P et al.
platelets and brain from patients with AlzheimerÕs disease. Melatonin reduces induction of Bax, caspase and cell death in
Neurobiol Aging 2002; 23:371–376. methamphetamine-treated human neuroblastoma SH-SY5Y
342. Jou MJ, Peng TI, Reiter RJ et al. Visualization of the an- cultured cells. J Pineal Res 2009; 46:433–440.
tioxidative effects of melatonin at the mitochondrial level 361. Feng Z, Zhang JT. Melatonin reduces amyloid beta-induced
during oxidative stress-induced apoptosis of rat brain astro- apoptosis in pheochromocytoma (PC12) cells. J Pineal Res
cytes. J Pineal Res 2004; 37:55–70. 2004; 37:257–266.
343. Leon J, Acuna-Castroviejo D, Sainz RM et al. Melatonin 362. Chen M. The AlzheimerÕs plaques, tangles and memory def-
and mitochondrial function. Life Sci 2004; 75:765–790. icits may have a common origin. Part III: animal model.
344. Jou MJ, Peng TI, Yu PZ et al. Melatonin protects against Front Biosci 1998; 3:A47–A51.
common deletion of mitochondrial DNA-augmented mito- 363. Fukuyama R, Wadhwani KC, Galdzicki Z et al. beta-
chondrial oxidative stress and apoptosis. J Pineal Res 2007; Amyloid polypeptide increases calcium-uptake in PC12 cells:
43:389–403. a possible mechanism for its cellular toxicity in AlzheimerÕs
345. Acuna CD, Lopez LC, Escames G et al. Melatonin-mito- disease. Brain Res 1994; 667:269–272.
chondria interplay in health and disease. Curr Top Med 364. Mattson MP, Cheng B, Davis D et al. beta-Amyloid pep-
Chem 2011; 11:221–240. tides destabilize calcium homeostasis and render human cor-
346. Martin M, Macias M, Escames G et al. Melatonin but not tical neurons vulnerable to excitotoxicity. J Neurosci 1992;
vitamins C and E maintains glutathione homeostasis in t- 12:376–389.
butyl hydroperoxide-induced mitochondrial oxidative stress. 365. Wu A, Derrico CA, Hatem L et al. AlzheimerÕs amyloid-
FASEB J 2000; 14:1677–1679. beta peptide inhibits sodium/calcium exchange measured in
347. Bozner P, Grishko V, Ledoux SP et al. The amyloid beta rat and human brain plasma membrane vesicles. Neurosci-
protein induces oxidative damage of mitochondrial DNA. ence 1997; 80:675–684.
J Neuropathol Exp Neurol 1997; 56:1356–1362. 366. Ho R, Ortiz D, Shea TB. Amyloid-beta promotes calcium
348. Carretero M, Escames G, Lopez LC et al. Long-term influx and neurodegeneration via stimulation of L voltage-
melatonin administration protects brain mitochondria from sensitive calcium channels rather than NMDA channels in
aging. J Pineal Res 2009; 47:192–200. cultured neurons. J Alzheimers Dis 2001; 3:479–483.
349. Poeggeler B, Sambamurti K, Siedlak SL et al. A novel 367. Green KN, Smith IF, Laferla FM. Role of calcium in the
endogenous indole protects rodent mitochondria and extends pathogenesis of AlzheimerÕs disease and transgenic models.
rotifer lifespan. PLoS ONE 2010; 5:e10206–e10214. Subcell Biochem 2007; 45:507–521.
350. Andrabi SA, Sayeed I, Siemen D et al. Direct inhibition of 368. Isaacs AM, Senn DB, Yuan M et al. Acceleration of amy-
the mitochondrial permeability transition pore: a possible loid beta-peptide aggregation by physiological concentrations
mechanism responsible for anti-apoptotic effects of melato- of calcium. J Biol Chem 2006; 281:27916–27923.
nin. FASEB J 2004; 18:869–871. 369. Querfurth HW, Selkoe DJ. Calcium ionophore increases
351. Orrenius S, Zhivotovsky B. Cardiolipin oxidation sets amyloid beta peptide production by cultured cells. Biochem-
cytochrome c free. Nat Chem Biol 2005; 1:188–189. istry (Mosc) 1994; 33:4550–4561.
352. Petrosillo G, Moro N, Ruggiero FM et al. Melatonin 370. Danysz W, Parsons CG. The NMDA receptor antagonist
inhibits cardiolipin peroxidation in mitochondria and memantine as a symptomatological and neuroprotective
prevents the mitochondrial permeability transition and treatment for AlzheimerÕs disease: preclinical evidence. Int J
cytochrome c release. Free Radic Biol Med 2009; 47: Geriatr Psychiatry 2003; 18:S23–S32.
969–974. 371. Vanecek J. Melatonin inhibits increase of intracellular cal-
353. Paradies G, Petrosillo G, Paradies V et al. Melatonin, cium and cyclic AMP in neonatal rat pituitary via indepen-
cardiolipin and mitochondrial bioenergetics in health and dent pathways. Mol Cell Endocrinol 1995; 107:149–153.
disease. J Pineal Res 2010; 48:297–310. 372. Slanar O, Pelisek V, Vanecek J. Melatonin inhibits pitui-
354. Louneva N, Cohen JW, Han LY et al. Caspase-3 is enriched tary adenylyl cyclase-activating polypeptide-induced increase
in postsynaptic densities and increased in AlzheimerÕs disease. of cyclic AMP accumulation and [Ca2+]i in cultured cells of
Am J Pathol 2008; 173:1488–1495. neonatal rat pituitary. Neurochem Int 2000; 36:213–219.

198
AlzheimerÕs disease and melatonin

373. Brunner P, Sozer-Topcular N, Jockers R et al. Pineal and 391. Johnson RJ, Xiao G, Shanmugaratnam J et al. Calreticulin
cortical melatonin receptors MT1 and MT2 are decreased in functions as a molecular chaperone for the beta-amyloid
AlzheimerÕs disease. Eur J Histochem 2006; 50:311–316. precursor protein. Neurobiol Aging 2001; 22:387–395.
374. Zatz M, Mullen DA. Does calcium influx regulate mela- 392. Tajes OM, Pelegri GC, Vilaplana HJ et al. An evaluation
tonin production through the circadian pacemaker in chick of the neuroprotective effects of melatonin in an in vitro
pineal cells? Effects of nitrendipine, Bay K 8644, Co2+, experimental model of age-induced neuronal apoptosis.
Mn2+, and low external Ca2+. Brain Res 1988; 463:305– J Pineal Res 2009; 46:262–267.
316. 393. Sanna PP, Cammalleri M, Berton F et al. Phosphati-
375. Ayar A, Martin DJ, Ozcan M et al. Melatonin inhibits dylinositol 3-kinase is required for the expression but not
high voltage activated calcium currents in cultured rat dorsal for the induction or the maintenance of long-term poten-
root ganglion neurones. Neurosci Lett 2001; 313:73–77. tiation in the hippocampal CA1 region. J Neurosci 2002;
376. Escames G, Macias M, Leon J et al. Calcium-dependent 22:3359–3365.
effects of melatonin inhibition of glutamatergic response in rat 394. Schaffer BA, Bertram L, Miller BL et al. Association of
striatum. J Neuroendocrinol 2001; 13:459–466. GSK3B with Alzheimer disease and frontotemporal demen-
377. Escames G, Leon J, Lopez LC et al. Mechanisms of N-me- tia. Arch Neurol 2008; 65:1368–1374.
thyl-D-aspartate receptor inhibition by melatonin in the rat 395. Bhat RV, Budd SL. GSK3beta signalling: casting a wide net
striatum. J Neuroendocrinol 2004; 16:929–935. in AlzheimerÕs disease. Neurosignals 2002; 11:251–261.
378. Sutcu R, Yonden Z, Yilmaz A et al. Melatonin increases 396. Lee HK, Kumar P, Fu Q et al. The insulin/Akt signaling
NMDA receptor subunits 2A and 2B concentrations in rat pathway is targeted by intracellular beta-amyloid. Mol Biol
hippocampus. Mol Cell Biochem 2006; 283:101–105. Cell 2009; 20:1533–1544.
379. Pozo D, Reiter RJ, Calvo JR et al. Inhibition of cerebellar 397. Frolich L, Blum-Degen D, Bernstein HG et al. Brain
nitric oxide synthase and cyclic GMP production by melato- insulin and insulin receptors in aging and sporadic Alzhei-
nin via complex formation with calmodulin. J Cell Biochem merÕs disease. J Neural Transm 1998; 105:423–438.
1997; 65:430–442. 398. Cordes CM, Bennett RG, Siford GL et al. Nitric oxide
380. Tiberi M, Lavoie PA. Inhibition of the retrograde axonal inhibits insulin-degrading enzyme activity and function
transport of acetylcholinesterase by the anti-calmodulin through S-nitrosylation. Biochem Pharmacol 2009; 77:1064–
agents amitriptyline and desipramine. J Neurobiol 1985; 1073.
16:245–248. 399. Kurochkin IV, Goto S. AlzheimerÕs beta-amyloid peptide
381. Benitez-King G, Anton-Tay F. Calmodulin mediates specifically interacts with and is degraded by insulin degrad-
melatonin cytoskeletal effects. Experientia 1993; 49:635–641. ing enzyme. FEBS Lett 1994; 345:33–37.
382. Bettahi I, Pozo D, Osuna C et al. Melatonin reduces nitric 400. Sridhar GR, Thota H, Allam AR et al. AlzheimerÕs disease
oxide synthase activity in rat hypothalamus. J Pineal Res and type 2 diabetes mellitus: the cholinesterase connection?
1996; 20:205–210. Lipids Health Dis 2006; 5:28.
383. Turjanski AG, Estrin DA, Rosenstein RE et al. The 401. Carro E, Torres-Aleman I. Insulin-like growth factor I and
interaction between melatonin to calmodulin: a NMR and AlzheimerÕs disease: therapeutic prospects? Expert Rev Neu-
molecular dynamics study. In: Melatonin – From Molecules rother 2004; 4:79–86.
to Therapy, Pandi-Perumal SR, Cardinali DP, eds., Nova 402. Niikura T, Hashimoto Y, Okamoto T et al. Insulin-like
Science, New York, 2007; pp. 47–68. growth factor I (IGF-I) protects cells from apoptosis by
384. Benitez-King G, Rios A, Martinez A et al. In vitro inhi- AlzheimerÕs V642I mutant amyloid precursor protein through
bition of Ca2+/calmodulin-dependent kinase II activity by IGF-I receptor in an IGF-binding protein-sensitive manner.
melatonin. Biochim Biophys Acta 1996; 1290:191–196. J Neurosci 2001; 21:1902–1910.
385. Huerto-Delgadillo L, Anton-Tay F, Benitez-King G. 403. Dore S, Kar S, Quirion R. Insulin-like growth factor I
Effects of melatonin on microtubule assembly depend on protects and rescues hippocampal neurons against beta-
hormone concentration: role of melatonin as a calmodulin amyloid- and human amylin-induced toxicity. Proc Natl Acad
antagonist. J Pineal Res 1994; 17:55–62. Sci USA 1997; 94:4772–4777.
386. Benitez-King G, Tunez I, Bellon A et al. Melatonin 404. Janson J, Laedtke T, Parisi JE et al. Increased risk of type 2
prevents cytoskeletal alterations and oxidative stress induced diabetes in Alzheimer disease. Diabetes 2004; 53:474–481.
by okadaic acid in N1E-115 cells. Exp Neurol 2003; 182:151– 405. Luchsinger JA, Tang MX, Stern Y et al. Diabetes mellitus
159. and risk of AlzheimerÕs disease and dementia with stroke in a
387. Benitez-King G, Hernandez ME, Tovar R et al. Melato- multiethnic cohort. Am J Epidemiol 2001; 154:635–641.
nin activates PKC-alpha but not PKC-epsilon in N1E-115 406. Ho L, Qin W, Pompl PN et al. Diet-induced insulin resis-
cells. Neurochem Int 2001; 39:95–102. tance promotes amyloidosis in a transgenic mouse model of
388. Ortiz GG, Itez-King GA, Rosales-Corral SA et al. Cel- AlzheimerÕs disease. FASEB J 2004; 18:902–904.
lular and biochemical actions of melatonin which protect 407. Xie L, Helmerhorst E, Taddei K et al. AlzheimerÕs beta-
against free radicals: role in neurodegenerative disorders. amyloid peptides compete for insulin binding to the insulin
Curr Neuropharmacol 2008; 6:203–214. receptor. J Neurosci 2002; 22:RC221.
389. Benitez-King G, Ortiz-Lopez L, Jimenez-Rubio G. 408. Ullrich A, Gray A, Tam AW et al. Insulin-like growth
Melatonin precludes cytoskeletal collapse caused by factor I receptor primary structure: comparison with insulin
hydrogen peroxide: participation of protein kinase C. 2005; receptor suggests structural determinants that define func-
2:767–778. tional specificity. EMBO J 1986; 5:2503–2512.
390. Macias M, Escames G, Leon J et al. Calreticulin-melatonin. 409. Quevedo C, Alcazar A, Salinas M. Two different signal
An unexpected relationship. Eur J Biochem 2003; 270:832– transduction pathways are implicated in the regulation
840. of initiation factor 2B activity in insulin-like growth

199
Rosales-Corral et al.

factor-1-stimulated neuronal cells. J Biol Chem 2000; 427. Olivier P, Fontaine RH, Loron G et al. Melatonin pro-
275:19192–19197. motes oligodendroglial maturation of injured white matter in
410. Vriend J, Sheppard MS, Bala RM. Melatonin increases neonatal rats. PLoS ONE 2009; 4:e7128–e7141.
serum insulin-like growth factor-I in male Syrian hamsters. 428. Peschke E. Melatonin, endocrine pancreas and diabetes. J
Endocrinology 1988; 122:2558–2561. Pineal Res 2008; 44:26–40.
411. Daniels WM, Reiter RJ, Melchiorri D et al. Melatonin 429. Peschke E, Schucht H, Muhlbauer E. Long-term enteral
counteracts lipid peroxidation induced by carbon tetrachlo- administration of melatonin reduces plasma insulin and in-
ride but does not restore glucose-6 phosphatase activity. creases expression of pineal insulin receptors in both Wistar
J Pineal Res 1995; 19:1–6. and type 2-diabetic Goto-Kakizaki rats. J Pineal Res 2010;
412. Jale O, Liter K, Hakan O. Melatonin Increases the 49:373–381.
Expression of Insulin-like Growth Factor I in Rats with 430. Ghosh G, De K, Maity S et al. Melatonin protects against
Carbontetrachlorid-Induced Hepatic Damage. Anim Vet Adv oxidative damage and restores expression of GLUT4 gene in
2009; 8:2251–2256. the hyperthyroid rat heart. J Pineal Res 2007; 42:71–82.
413. Sivakumar V, Ling EA, Lu J et al. Role of glutamate and its 431. Nduhirabandi F, Du Toit EF, Blackhurst D et al.
receptors and insulin-like growth factors in hypoxia induced Chronic melatonin consumption prevents obesity-related
periventricular white matter injury. Glia 2010; 58:507–523. metabolic abnormalities and protects the heart against myo-
414. Pawlikowski M, Kolomecka M, Wojtczak A et al. Effects cardial ischemia and reperfusion injury in a prediabetic model
of six months melatonin treatment on sleep quality and serum of diet-induced obesity. J Pineal Res 2011; 50:171–182.
concentrations of estradiol, cortisol, dehydroepiandrosterone 432. Srivastava RK, Krishna A. Melatonin modulates glucose
sulfate, and somatomedin C in elderly women. Neuro Endo- homeostasis during winter dormancy in a vespertilionid bat,
crinol Lett 2002; 23(Suppl 1):17–19. Scotophilus heathi. Comp Biochem Physiol A Mol Integr
415. Garfinkel D, Laudon M, Nof D et al. Improvement of Physiol 2010; 155:392–400.
sleep quality in elderly people by controlled-release melatonin. 433. Pettegrew JW, Panchalingam K, Klunk WE et al.
Lancet 1995; 346:541–544. Alterations of cerebral metabolism in probable AlzheimerÕs
416. Gianotti L, Pivetti S, Lanfranco F et al. Concomitant disease: a preliminary study. Neurobiol Aging 1994; 15:117–
impairment of growth hormone secretion and peripheral 132.
sensitivity in obese patients with obstructive sleep apnea 434. Sartori C, Dessen P, Mathieu C et al. Melatonin improves
syndrome. J Clin Endocrinol Metab 2002; 87:5052–5057. glucose homeostasis and endothelial vascular function in
417. Savaskan E, Olivieri G, Meier F et al. Increased melatonin high-fat diet-fed insulin-resistant mice. Endocrinology 2009;
1a-receptor immunoreactivity in the hippocampus of Alzhei- 150:5311–5317.
merÕs disease patients. J Pineal Res 2002; 32:59–62. 435. Ha E, Yim SV, Chung JH et al. Melatonin stimulates glucose
418. Anhe GF, Caperuto LC, Pereira-Da-Silva M et al. In vivo transport via insulin receptor substrate-1/phosphatidylinosi-
activation of insulin receptor tyrosine kinase by melatonin in tol 3-kinase pathway in C2C12 murine skeletal muscle cells.
the rat hypothalamus. J Neurochem 2004; 90:559–566. J Pineal Res 2006; 41:67–72.
419. Yamaguchi A, Tamatani M, Matsuzaki H et al. Akt 436. Lange Y, Ye J, Rigney M et al. Regulation of endoplasmic
activation protects hippocampal neurons from apoptosis by reticulum cholesterol by plasma membrane cholesterol.
inhibiting transcriptional activity of p53. J Biol Chem 2001; J Lipid Res 1999; 40:2264–2270.
276:5256–5264. 437. Hartmann T, Kuchenbecker J, Grimm MO. AlzheimerÕs
420. Datta SR, Dudek H, Tao X et al. Akt phosphorylation of disease: the lipid connection. J Neurochem 2007; 103(Suppl
BAD couples survival signals to the cell-intrinsic death 1):159–170.
machinery. Cell 1997; 91:231–241. 438. Grimm MO, Grimm HS, Patzold AJ et al. Regulation of
421. Zenobi PD, Graf S, Ursprung H et al. Effects of insulin- cholesterol and sphingomyelin metabolism by amyloid-beta
like growth factor-I on glucose tolerance, insulin levels, and and presenilin. Nat Cell Biol 2005; 7:1118–1123.
insulin secretion. J Clin Invest 1992; 89:1908–1913. 439. Lichtenthaler SF, Beher D, Grimm HS et al. The intra-
422. Cheng CM, Reinhardt RR, Lee WH et al. Insulin-like membrane cleavage site of the amyloid precursor protein
growth factor 1 regulates developing brain glucose metabo- depends on the length of its transmembrane domain. Proc
lism. Proc Natl Acad Sci USA 2000; 97:10236–10241. Natl Acad Sci USA 2002; 99:1365–1370.
423. Sharma S, Prasanthi RPJ, Schommer E et al. Hypercho- 440. Grimm MO, Tschape JA, Grimm HS et al. Altered mem-
lesterolemia-induced Abeta accumulation in rabbit brain is brane fluidity and lipid raft composition in presenilin-deficient
associated with alteration in IGF-1 signaling. Neurobiol Dis cells. Acta Neurol Scand Suppl 2006; 185:27–32.
2008; 32:426–432. 441. Grziwa B, Grimm MO, Masters CL et al. The transmem-
424. Hirsch-Rodriguez E, Imbesi M, Manev R et al. The pat- brane domain of the amyloid precursor protein in microsomal
tern of melatonin receptor expression in the brain may membranes is on both sides shorter than predicted. J Biol
influence antidepressant treatment. Med Hypotheses 2007; Chem 2003; 278:6803–6808.
69:120–124. 442. Simons M, Keller P, De SB et al. Cholesterol depletion
425. Bordt SL, Mckeon RM, Li PK et al. N1E-115 mouse neu- inhibits the generation of beta-amyloid in hippocampal neu-
roblastoma cells express MT1 melatonin receptors and pro- rons. Proc Natl Acad Sci USA 1998; 95:6460–6464.
duce neurites in response to melatonin. Biochim Biophys Acta 443. Fassbender K, Simons M, Bergmann C et al. Simvastatin
2001; 1499:257–264. strongly reduces levels of AlzheimerÕs disease beta -amyloid
426. Cui P, Yu M, Luo Z et al. Intracellular signaling pathways peptides Abeta 42 and Abeta 40 in vitro and in vivo. Proc
involved in cell growth inhibition of human umbilical Natl Acad Sci USA 2001; 98:5856–5861.
vein endothelial cells by melatonin. J Pineal Res 2008; 44:107– 444. Avdulov NA, Chochina SV, Igbavboa U et al. Lipid
114. binding to amyloid beta-peptide aggregates: preferential

200
AlzheimerÕs disease and melatonin

binding of cholesterol as compared with phosphatidylcholine 462. Reiter R, Tang L, Garcia JJ, Muñoz-Hoyos A. Pharma-
and fatty acids. J Neurochem 1997; 69:1746–1752. cological actions of melatonin in oxygen radical pathophysi-
445. Abad-Rodriguez J, Ledesma MD, Craessaerts K et al. ology. Life Sci 1997; 60:2255–2271.
Neuronal membrane cholesterol loss enhances amyloid pep- 463. Reiter RJ, Tan DX, Korkmaz A et al. Obesity and metabolic
tide generation. J Cell Biol 2004; 167:953–960. syndrome: association with chronodisruption, sleep depriva-
446. Kivipelto M, Helkala EL, Laakso MP et al. Midlife vas- tion, and melatonin suppression. Ann Med 2011; in press.
cular risk factors and AlzheimerÕs disease in later life: longi- 464. Rosales-Corral S, Ortiz G, Valdivia-Velazquez M.
tudinal, population based study. Br Med J 2001; 322:1447– Oxidative stress in rat brain induced by amyloid-beta under
1451. continuous light conditions. 2004; 154:170–170.
447. Yuyama K, Yanagisawa K. Sphingomyelin accumulation 465. Sultana R, Butterfield DA. Alterations of some mem-
provides a favorable milieu for GM1 ganglioside-induced brane transport proteins in AlzheimerÕs disease: role of amy-
assembly of amyloid beta-protein. Neurosci Lett 2010; loid beta-peptide. Mol Biosyst 2008; 4:36–41.
481:168–172. 466. Pratico D, Zhukareva V, Yao Y et al. 12/15-lipoxygenase
448. Jaeger S, Pietrzik CU. Functional role of lipoprotein is increased in AlzheimerÕs disease: possible involvement in
receptors in AlzheimerÕs disease. Curr Alzheimer Res 2008; brain oxidative stress. Am J Pathol 2004; 164:1655–1662.
5:15–25. 467. Chu J, Pratico D. 5-lipoxygenase as an endogenous modu-
449. Frank-Cannon TC, Alto LT, Mcalpine FE et al. Does lator of amyloid beta formation in vivo. Ann Neurol 2010;
neuroinflammation fan the flame in neurodegenerative dis- 69:34–46.
eases? Mol Neurodegener 2009; 4:47–60. 468. Uz T, Longone P, Manev H. Increased hippocampal 5-lip-
450. Moses GS, Jensen MD, Lue LF et al. Secretory PLA2-IIA: a oxygenase mRNA content in melatonin-deficient, pinealec-
new inflammatory factor for AlzheimerÕs disease. J Neuroin- tomized rats. J Neurochem 1997; 69:2220–2223.
flammation 2006; 3:28–39. 469. Manev H, Uz T, Sugaya K et al. Putative role of neuronal 5-
451. Soderberg M, Edlund C, Kristensson K et al. Fatty acid lipoxygenase in an aging brain. FASEB J 2000; 14:1464–1469.
composition of brain phospholipids in aging and in Alzhei- 470. Radogna F, Sestili P, Martinelli C et al. Lipoxygenase-
merÕs disease. Lipids 1991; 26:421–425. mediated pro-radical effect of melatonin via stimulation of
452. Tully AM, Roche HM, Doyle R et al. Low serum cho- arachidonic acid metabolism. Toxicol Appl Pharmacol 2009;
lesteryl ester-docosahexaenoic acid levels in AlzheimerÕs dis- 238:170–177.
ease: a case–control study. Br J Nutr 2003; 89:483–489. 471. Li B, Zhang H, Akbar M et al. Negative regulation of
453. Haag M. Essential fatty acids and the brain. Can J Psychiatry cytosolic phospholipase A(2) by melatonin in the rat pineal
2003; 48:195–203. gland. Biochem J 2000; 351(Pt 3):709–716.
454. Jones CR, Arai T, Rapoport SI. Evidence for the involve- 472. Hussain SA. Effect of melatonin on cholesterol absorption in
ment of docosahexaenoic acid in cholinergic stimulated signal rats. J Pineal Res 2007; 42:267–271.
transduction at the synapse. Neurochem Res 1997; 22:663– 473. Tailleux A, Torpier G, Bonnefont-Rousselot D et al.
670. Daily melatonin supplementation in mice increases athero-
455. Conklin SM, Gianaros PJ, Brown SM et al. Long-chain sclerosis in proximal aorta. Biochem Biophys Res Commun
omega-3 fatty acid intake is associated positively with corti- 2002; 293:1114–1123.
colimbic gray matter volume in healthy adults. Neurosci Lett 474. Wakatsuki A, Okatani Y, Ikenoue N et al. Melatonin
2007; 421:209–212. inhibits oxidative modification of low-density lipoprotein
456. Gamoh S, Hashimoto M, Sugioka K et al. Chronic particles in normolipidemic post-menopausal women. J Pineal
administration of docosahexaenoic acid improves reference Res 2000; 28:136–142.
memory-related learning ability in young rats. Neuroscience 475. Bongiorno D, Ceraulo L, Ferrugia M et al. Localization
1999; 93:237–241. and interactions of melatonin in dry cholesterol/lecithin
457. Gamoh S, Hashimoto M, Hossain S et al. Chronic admin- mixed reversed micelles used as cell membrane models.
istration of docosahexaenoic acid improves the performance J Pineal Res 2005; 38:292–298.
of radial arm maze task in aged rats. Clin Exp Pharmacol 476. Nicholson AM, Ferreira A. Increased membrane choles-
Physiol 2001; 28:266–270. terol might render mature hippocampal neurons more sus-
458. Chung WL, Chen JJ, Su HM. Fish oil supplementation of ceptible to beta-amyloid-induced calpain activation and tau
control and (n-3) fatty acid-deficient male rats enhances ref- toxicity. J Neurosci 2009; 29:4640–4651.
erence and working memory performance and increases brain 477. Maurizi CP. Choroid plexus portals and a deficiency of
regional docosahexaenoic acid levels. J Nutr 2008; 138:1165– melatonin can explain the neuropathology of AlzheimerÕs
1171. disease. Med Hypotheses 2010; 74:1059–1066.
459. Kawakita E, Hashimoto M, Shido O. Docosahexaenoic 478. Wu YH, Zhou JN, Van HJ et al. Decreased MT1 melatonin
acid promotes neurogenesis in vitro and in vivo. Neuroscience receptor expression in the suprachiasmatic nucleus in aging and
2006; 139:991–997. AlzheimerÕs disease. Neurobiol Aging 2007; 28:1239–1247.
460. Katakura M, Hashimoto M, Shahdat HM et al. Doco- 479. Eichhorn GL. Is there any relationship between aluminum
sahexaenoic acid promotes neuronal differentiation by regu- and AlzheimerÕs disease? Exp Gerontol 1993; 28:493–498.
lating basic helix-loop-helix transcription factors and cell 480. Trapp GA, Miner GD, Zimmerman RL et al. Aluminum
cycle in neural stem cells. Neuroscience 2009; 160:651–660. levels in brain in AlzheimerÕs disease. Biol Psychiatry 1978;
461. Ma QL, Yang F, Rosario ER et al. Beta-amyloid oligomers 13:709–718.
induce phosphorylation of tau and inactivation of insulin 481. Khaldy H, Escames G, Leon J et al. Synergistic effects of
receptor substrate via c-Jun N-terminal kinase signaling: melatonin and deprenyl against MPTP-induced mitochon-
suppression by omega-3 fatty acids and curcumin. J Neurosci drial damage and DA depletion. Neurobiol Aging 2003;
2009; 29:9078–9089. 24:491–500.

201
Rosales-Corral et al.

482. Borowicz KK, Kaminski R, Gasior M et al. Influence of for up to 12 hours after awakening. J Clin Sleep Med 2010;
melatonin upon the protective action of conventional anti- 6:565–571.
epileptic drugs against maximal electroshock in mice. Eur 486. Dhand R, Sohal H. Good sleep, bad sleep! The role of
Neuropsychopharmacol 1999; 9:185–190. daytime naps in healthy adults. Curr Opin Pulm Med 2006;
483. Squire LR. Memory functions as affected by electroconvul- 12:379–382.
sive therapy. Ann N Y Acad Sci 1986; 462:307–314. 487. Papavasiliou PS, Cotzias GC, Duby SE et al. Melatonin
484. Gupta M, Aneja S, Kohli K. Add-on melatonin improves and parkinsonism. JAMA 1972; 221:88–89.
quality of life in epileptic children on valproate monotherapy: 488. Barchas J, Dacosta F, Spector S. Acute pharmacology of
a randomized, double-blind, placebo-controlled trial. Epi- melatonin. Nature 1967; 214:919–920.
lepsy Behav 2004; 5:316–321. 489. Hardeland R, Poeggeler B, Srinivasan V et al. Mela-
485. Cohen DA, Wang W, Klerman EB et al. Ramelteon prior tonergic drugs in clinical practice. Arzneimittelforschung
to a short evening nap impairs neurobehavioral performance 2008; 58:1–10.

202

You might also like