ArticlePDF AvailableLiterature Review

A brief review about melatonin, a pineal hormone

Authors:

Abstract and Figures

Melatonin is a ubiquitous molecule in nature, being locally synthesized in several cells and tissues, besides being a hormone that is centrally produced in the pineal gland of vertebrates, particularly in mammals. Its pineal synthesis is timed by the suprachiasmatic nucleus, that is synchronized to the light-dark cycle via the retinohypothalamic tract, placing melatonin synthesis at night, provided its dark. This unique trait turns melatonin into an internal synchronizer that adequately times the organism’s physiology to the daily and seasonal demands. Besides being amphiphilic, melatonin presents specific mechanisms and ways of action devoted to its role as a time-giving agent, being widely spread in the organism. The present review aims to focus on melatonin as a pineal hormone with specific mechanisms and ways of action, besides presenting the clinical syndromes related to its synthesis and/or function disruptions. © 2018, Sociedade Brasileira de Endocrinologia e Metabologia. All rights reserved.
Content may be subject to copyright.
Copyright© AE&M all rights reserved.
472
review
Arch Endocrinol Metab. 2018;62/4
1 Departamento de Fisiologia,
Universidade Federal de São Paulo
(Unifesp), São Paulo, SP, Brasil
2 Departamento de Fisiologia e
Biofísica, Instituto de Ciências
Biomédicas, Universidade de São
Paulo (USP), São Paulo, SP, Brasil
Correspondence to:
José Cipolla-Neto
Department of Physiology and
Biophysics, Neurobiology Lab,
Institute of Biomedical Sciences,
Bldg 1, University of São Paulo
Av. Lineu Prestes, 1524
05508-000 – São Paulo, SP, Brazil
cipolla@icb.usp.br
Received on Ago/31/2018
Accepted on Ago/31/2018
DOI: 10.20945/2359-3997000000066
A brief review about melatonin,
a pineal hormone
Fernanda Gaspar do Amaral1, José Cipolla-Neto2
ABSTRACT
Melatonin is a ubiquitous molecule in nature, being locally synthesized in several cells and tissues,
besides being a hormone that is centrally produced in the pineal gland of vertebrates, particularly
in mammals. Its pineal synthesis is timed by the suprachiasmatic nucleus, that is synchronized to
the light-dark cycle via the retinohypothalamic tract, placing melatonin synthesis at night, provided
its dark. This unique trait turns melatonin into an internal synchronizer that adequately times the
organism’s physiology to the daily and seasonal demands. Besides being amphiphilic, melatonin
presents specic mechanisms and ways of action devoted to its role as a time-giving agent, being
widely spread in the organism. The present review aims to focus on melatonin as a pineal hormone
with specic mechanisms and ways of action, besides presenting the clinical syndromes related to its
synthesis and/or function disruptions.
Arch Endocrinol Metab. 2018;62(4):472-9
Keywords
Melatonin; pineal; hypermelatoninemia; hypomelatoninemia
MELATONIN
Melatonin, N-acetyl-5-methoxytryptamine, is a
ubiquitous molecule in nature, being found in almost
all living organisms. It is an indolamine present in
any compartment of the organism for its amphiphilic
characteristics of diffusion (Figure 1). In vertebrates,
mammals in particular, in addition of local production
in several tissues, melatonin is centrally produced by the
pineal gland and directly released in the blood, acting as
a hormone. The pineal gland is an unpaired epithalamic
neuroendocrine gland originating from the roof of the
third ventricle containing, in mammals, melatonin-
producing cells called pinealocytes, in addition to
astrocytes and other cell types (1).
Melatonin synthesis by the pinealocytes in the
pineal gland is under the control of a neural system
originating in the hypothalamic paraventricular nuclei,
projecting directly and indirectly to the preganglionic
sympathetic neurons of the rst thoracic segments of
the spinal cord. Following, through a projection of the
postganglionary sympathetic neuron of the superior
cervical ganglia, nerve bers forming the conary nerves
reach the pineal gland (Figure 2).
Norepinephrine released by the sympathetic
terminals interacts with the classical beta and alpha
noradrenergic receptors in the membrane of pinealocytes
and activates cAMP-PKA-CREB and PLC-Ca++-PKC
pathways to trigger melatonin synthesis (2).
Melatonin synthesis initiates with tryptophan
that, under the action of tryptophan hydroxylase, is
transformed in 5-hydroxytryptophan that, in turn,
is converted to serotonin, which is acetylated, by
arylalkylamine N-acetyltransferase (AANAT) to
N-acetylserotonin (NAS) that is converted to melatonin
by acetylserotonin O-methyltransferase (ASMT) former
called hydroxy-indole-O-methyltransferase (HIOMT).
The three enzymes above are under the control of neural
and endocrine systems that regulate time, duration and
amount of produced melatonin (3) (Figure 3).
The major control is exerted by the circadian timing
system, mainly the hypothalamic suprachiasmatic
nuclei, that times melatonin synthesis so that it is daily
produced in synchrony to the light/dark cycle, being
tightly restricted to the night, provided it is dark. Light
stimulus (mainly in the blue range) activates melanopsin
breakdown in retinal photoreceptive ganglion cells that
Copyright© AE&M all rights reserved.
473
Melatonin hormonal ways of action
Arch Endocrinol Metab. 2018;62/4
project, via the retinohypothalamic pathway, to the
hypothalamus, inhibiting melatonin synthesis (4).
Due to its amphiphilic nature, melatonin is not
stored inside the pinealocytes, being released as it is
synthetized. The pineal gland is profusely vascularized
and its attachment, dorsal and posterior, to the third
ventricle wall allows melatonin to be released into
the cerebrospinal uid of the central nervous system
during the night, as well as into the blood stream. In
the blood, melatonin is usually bound to albumin,
metabolized to 6-hydroxymelatonin by cytochrome
P450 isoforms (mainly CYP1A2) and conjugated to
6-sulfatoxymelatonin in the liver, for the subsequent
urinary excretion. 6-sulfatoxymelatonin production
perfectly reects the plasma levels of melatonin, so
its urinary measurement is a less intrusive method
to evaluate the pineal function and melatonin
production (Figure 3). In the central nervous system
melatonin is degraded to N-acetyl- N2-formyl-5-
methoxykynuramine (AFMK) that is deformylated to
N-acetyl-5- methoxykynuramine (AMK) (5).
Melatonin, as an ancient chemical messenger,
developed several pleiotropic mechanisms of action
(6). First, there are mechanisms that are not mediated
by cellular receptors and involve the direct interaction
of melatonin and other molecules, as its antioxidant
action. Melatonin is one of the most powerful natural
antioxidants, not only by directly chelating oxygen and
nitrogen reactive species, but also by mobilizing the
intracellular antioxidant enzymatic system. Second, as
any other hormone, melatonin acts through specic
cellular receptors. Membrane melatonin receptors,
in mammals, are of two types, MT1 (MTNR1A, in
humans) and MT2 (MTNR1B in humans). These
membrane melatonin receptors are heterotrimeric Gi/
Go and Gq/11 protein-coupled receptors that interact
with downstream messengers such as adenylyl cyclase,
phospholipase A2 and phospholipase C, generally
decreasing cAMP and cGMP production and/or
increasing diacylglycerol and IP3 formation. MT1 and
MT2 receptors are found in almost all peripheral tissues,
as well as in the central nervous system. In addition to
acting through membrane receptors, melatonin might
interact with ROR/RZR (retinoid orphan receptors/
retinoid Z receptors) nuclear receptors (7) (Figure 4).
As mentioned before, melatonin hormonal pineal
production is restricted to the night and a light
signal, mainly with the characteristics of day-light
(predominance of the blue range), blocks melatonin
Figure 1. Melatonin molecule (232,2 molecular weight).
Figure 2. Neural control of pineal melatonin synthesis. RHT:
retinohypothalamic tract. SCN: suprachiasmatic nucleus. PVH:
paraventricular nucleus. SCG: superior cervical ganglion.
secretion. This complex and very well-organized neural
system control is the product of natural selection that
turned the melatonin nocturnal prole into the internal
representative of the environmental night. In addition,
and as a consequence, the duration of melatonin
nocturnal secretion episode follows the duration of the
night as it varies along the year. Long winter nights
determine long plasma melatonin duration episodes
and the reverse occurs following the short nights of
summer time.
Copyright© AE&M all rights reserved.
474
Melatonin hormonal ways of action
Arch Endocrinol Metab. 2018;62/4
Figure 3. Melatonin synthesis pathway and hepatic metabolization. The
enzymes are written in italic and the derived molecules are underlined.
Melatonin synthesis circadian rhythm synchronized
to day/night cycle, restricted to the night and to
its duration, converts melatonin to the internal
representative of the daily and seasonal photoperiod.
As a consequence of being a representative of the
environmental photoperiod, melatonin has, in some
way, to control the organism physiology during the 24
hours of the day and all through the seasons of the year.
To do that, melatonin, using the classical hormonal
mechanisms of action, developed several especial ways
of action (8).
As any other hormone, melatonin acts through the
classical way in the sense that its effects are seen as a
direct and immediate consequence of its interaction with
molecular effectors. These are called immediate effects
(Figure 5). Depending on the molecular effectors and
the involved mechanisms of action, such are the possible
effects: antioxidant action; reduction of cAMP-PKA-
CREB and cGMP; increased DAG, IP3, PKC activity;
regulation of potassium and calcium channels; etc. As
it would be expected for any hormone, the effects are
dependent on the target tissue and the correspondent
intracellular melatonin signaling pathway.
However, in addition to this classical and expected
hormonal way of action, melatonin developed another
one that is not seen during the night when melatonin is
being produced and released but, instead, it is only seen
during the day, being triggered in the absence of plasma
melatonin, provided it was present in the immediate
previous night. These are called prospective effects and
they are of two types (Figure 5). The rst one, called
proximal or consecutive, is seen right at the beginning
of the morning, immediately after the cessation of
melatonin production, when it is maximal and may
extend to several hours. One example is the cAMP/
PKA/CREB pathway super or hypersensitization that is
seen in several peripheral and central systems (9). That
is to say that in consequence of the nocturnal sustained
and prolonged adenylyl cyclase inhibition induced by
melatonin through its Gi-protein coupled receptors,
this signaling pathway shows, following the cessation
of the inhibitory signal, an increased and potentiated
response to any agonists that activates adenylyl cyclase
through any Gs-protein coupled receptors. Depending
on the system (suprachiasmatic nucleus, pancreatic
beta cells, Leydig cells, pars tuberalis, etc.) such is
the magnitude and the duration of the potentiating
prospective consecutive melatonin effect (10-13). That
is to say that melatonin, in spite of being produced
only during the night, determines effects that are best
seen during the day, when melatonin is not present
anymore, and in this case, especially in the morning.
The second type of melatonin prospective effects are
called distal or prolonged effects. These are dependent
on the action of melatonin controlling the transcription
and/or translation of the well-known clock genes (CG)
and the clock-controlled genes (CCG) (14-20). The
clock genes are part of a complex molecular machinery
that includes a cycle of transcription and translation of
several genes and the resulting proteins might either
reinforce the process or inhibit it, so that the cycle
has a duration of approximately 24 hours (21). These
proteins can also control, throughout the 24-hour
cycle, the transcription and translation of other genes,
called clock-controlled genes, that are responsible for
Figure 4. Melatonin mechanisms of action. Classical receptor mediated
and non-mediated pathways and the involvement of nuclear, cytosolic and
membrane receptors.
Copyright© AE&M all rights reserved.
475
Melatonin hormonal ways of action
Arch Endocrinol Metab. 2018;62/4
almost all cell functions. That is to say that melatonin,
through its prospective prolonged effects, controlling
the cycling of the CG and CCG, is able to control cell
and tissue function all over the 24-hours of the day,
even being only produced and released during the
night.
Mediated by these immediate and prospective
effects, melatonin developed others ways of action.
One of them depends on the circadian characteristic
of the melatonin signal and on the contrast between
nocturnal and diurnal values of circulating melatonin.
The precise daily repetition of melatonin signal and
its perfect relation to the dark phase of the day turns
melatonin into the internal synchronizer of circadian
rhythms. This shapes the so-called chronobiotic effect
of melatonin (Figure 5). Melatonin is one of the most
powerful synchronizers of human circadian rhythms and
is used in clinics to adjust circadian rhythmicity in cases
like phase-delayed sleep onset disorder, jet lag, etc. (22-
25) The chronobiotic effect of melatonin depends on
its action at several levels of the circadian timing system,
including the suprachiasmatic nucleus and the clock
genes located in peripheral tissues like, adipose tissue,
muscle, pancreatic beta cells, reproductive organs, etc.
Another important synchronizing ef fect of melatonin
is related to the seasonal rhythms. It is its seasonal effect
(26-28) (Figure 5). The duration of melatonin diurnal
prole and, most importantly, the direction of the daily
change of its duration (towards increasing or decreasing
nights resulting in increasing or decreasing duration of
melatonin production) that is dependent on the typical
relative duration of the day and night along the year,
turns melatonin into the most powerful synchronizer of
seasonal rhythms, being fundamental for the organism
to anticipate and to adapt to the evolving environmental
change along the year. The seasonal synchronizing effect
of melatonin is mediated by its action on the pituitary
pars tuberalis. From there, the signal is transduced to
the hypothalamus, mediated by special glial cells called
tanycytes, and to the distal hypophysis through several
other mediators. In consequence of this functional
system, melatonin is able to control seasonal events like
reproduction, energy metabolism, immune response
and thermogenesis, growth, body weight control, etc.
(29-33).
Finally, another melatonin way of action called
transgenerational effect should be mentioned (Figure 5).
In mammals, in humans in particular, pineal melatonin
production increases as the gestation progress (34).
Maternal melatonin freely crosses the placenta and
reaches the fetus circulation, being its only source of
melatonin (35-37). Considering this, several of the effects
of melatonin that can be seen in the maternal organism
are seen in the fetus, particularly, the chronobiotic
Figure 5. Melatonin ways of action. The upper boxes represent the different ways of action. The second-line boxes explain how melatonin causes the
correspondent effects. The third-line boxes show examples of the correspondent effects. Note that the Prospective effects are further classied in
Proximal ou consecutive and Distal or prolonged effects.
Copyright© AE&M all rights reserved.
476
Melatonin hormonal ways of action
Arch Endocrinol Metab. 2018;62/4
and seasonal effects (38-40). Maternal melatonin is
responsible for the circadian timing and priming of
the fetus organism. Similarly, the seasonal timing is
transferred from the mother to the fetus, preparing its
neuroendocrine system to the future environment to
be dealt with (41,42). In addition, maternal melatonin
is necessary for the adequate neurodevelopment of the
fetus (43).
MELATONIN PHYSIOLOGY, CLINICAL SYNDROMES
AND THERAPEUTICS
Melatonin, due to its phylogenetic history, its pleiotropic
mechanisms of action and its unique ways of action,
as described above, in addition of being delivered to
the blood stream and directly in the CNS, is able to
regulate several, if not all, the physiological and neural
functions. Among them, circadian and seasonal timing
of organism; sleep and wakefulness cycle; endocrine
functions, as energy metabolism, glycemic control,
blood lipid prole and reproduction, gestation and
fetal development and programming; cardiovascular
system; immune system; neural development, neural
protection and neuroplasticity, etc. A detailed discussion
of this subject can be seen in Cipolla-Neto and Amaral,
2018 (8).
As any other hormone, from the clinical point
of view, melatonin hormonal dysfunction can be
classied as hypo (hypomelatoninemia) or hyper
(hypermelatoninemia) production by the pineal gland.
Hypomelatoninemia is dened by decreased
melatonin nocturnal peak value or total production
when compared to what is expected for the age- and
sex-paired population. This syndrome can be classied
as primary, dependent on factors that directly affect
the pineal gland and/or its innervation, and secondary,
developed as a consequence of a primary event, such as a
systemic disease (e.g. hyperglycemia) or environmental
factor (e.g light at night) (44-50).
Hypermelatoninemia is dened as hyperproduction
of pineal melatonin, usually associated to other diseases
like hypogonadotrophic hypogonadism, anorexia
nervosa, polycystic ovarian syndrome, Rabson-
Mendenhall syndrome and spontaneous hypothermia
hyperhidrosis (51-54).
A third syndrome associated to pineal melatonin
dysfunction is due to what can be called inappropriate
melatonin receptor-mediated response. This melatonin-
receptor dysfunction is usually a consequence of
melatonin receptors genetic variations (e.g. single
nucleotide polymorphisms) and affect either MT1 or
MT2 receptors (55,56).
Finally, in addition to these classical hormonal
dysfunctions and due to melatonin specic
characteristics of production and ways of action, it is
possible to dene another syndrome associated to
the time displacement of the nocturnal melatonin
production causing a phase-displacement of its plasma
prole that is called melatonin circadian displacement
syndrome. The result is a misalignment of the organism
to the circadian timing domain, causing sleep/wake,
metabolic and cardiovascular disturbances, among
other symptoms. This syndrome is usually associated
to the Smith-Magenis disease, the phase-delayed
sleep-wake disorder, and as a consequence of indoors
illumination during the evening/night, among others
(57,58).
Melatonin pharmacokinetics will depend on the
way of administration (oral, fast and/or slow-release,
intravenous, nasal spray, anal suppository, skin patches
or cream, etc.) and on the individual absorption and
hepatic metabolization rates (dependent on the activity
of cytochrome P450 complex, mainly CYP1A2).
Each of these aspects might vary depending on age
and sex. Usually, in a young/middle-aged human
patient, pharmacokinetic studies show that plasma
concentration reaches the peak at approximately 45
minutes after orally administered melatonin, resulting
in a low bioavailability due to the rst pass liver
metabolism (59).
Melatonin administration should always be done
during the evening/night, mimicking the physiological
production. The moment of administration during the
evening/night will depend on the desired effect that
will be determined by the well-known melatonin phase-
response curve. That is to say that depending on the
moment of administration, melatonin is able to act on
the circadian clock resulting in phase-advance, phase-
delay or even no phase-displacement of the circadian
rhythms. If administered in the late afternoon/
beginning of the evening, melatonin phase-advances
the circadian rhythms (as is the case for the treatment of
the sleep-wake phase-delay disorder); if administered in
the end of the night/early morning, melatonin would
phase-delay the circadian clock; if administered during
the evening, beginning around 1 hour before the usual
bedtime and extending to 2 to 4 hours afterwards,
melatonin does not phase displace the circadian
Copyright© AE&M all rights reserved.
477
Melatonin hormonal ways of action
Arch Endocrinol Metab. 2018;62/4
rhythms, regulating the circadian clock pace (this is the
time of choice if melatonin is being replaced, as in the
pinealectomized or elder patients).
It should be said that, considering the above
discussed melatonin ways of action, one should always
avoid its chronic administration during the day.
The dosage is always a concern and will depend
on the desired effect. If replacement therapy is the
goal, oral melatonin in the range of 0.1 to 0.5 mg
usually generates a plasma concentration varying from
100 to 500 pg/mL, that is 1 to 5 times the expected
physiological concentration at the nocturnal peak in
young people. In addition, 0.5 to 1.0 mg is usually used
for proper circadian timing, as in jetlag for example.
Melatonin-responsive sleep disorders are usually treated
with 1.0 to 5.0 mg oral melatonin. However, one
should always keep in mind that the dosage must be
individually adjusted by the evaluation of the evolution
of the symptoms and the occurrence of adverse effects
as diurnal somnolence or nocturnal nightmares.
CONCLUSIONS
Nowadays, any quick search in PubMed/NCBI shows
that there are about 33,000 papers about melatonin
or pineal and about 1,200 new articles per year, in
addition to a dedicated journal (Journal of Pineal
Research) showing the 2017 impact factor of 11.613
(6th out of 143 journals in the area of Endocrinology
& Metabolism). In spite of this, melatonin hormonal
action is scarcely targeted in the classical Physiology
or Endocrinology books. In Williams’ textbook of
Endocrinology, for example, the pineal gland was
included for a long time in the circumventricular
organs section and is, nowadays, located in a separate
scanty part just after it. Jameson & De Groot devoted,
for a long time, a full chapter to the pineal gland that
is mostly dedicated to pineal tumors. Melatonin in all
its complexity is barely reported. In addition, in several
medical schools all over the world, pineal gland and/or
melatonin is never systematically taught. So, it is about
time to change this picture, considering melatonin as a
hormone decisively important to the mammalian and
human physiology and pathophysiology.
Concerning the clinical aspects of melatonin
dysfunction some observations should be additionally
done. The most common mistake that we nd in
clinical endocrinology when dealing with melatonin
is to expect, as far as hypo or hyper production is
concerned, exactly the same that we would be expected
to occur with the classical glands syndromes: immediate
effects and immediate health repercussions. It is not
the case with pineal and melatonin as it is a time-
domain acting hormone that organizes physiology and
behavior in the circadian and seasonal time. Its absence
induces an unhealthy state whose clinical pictures
is not so tinted as is expressed in other glands but is
still there and will be reected in the long-term health
status rather then immediately. For example, a subtle
alteration in the sleep organization as little as shorter
30 minutes sleep episode every day will not be even
perceived by the patient and the physician. However,
it will determine, in the medium/long term, several
critical alterations in metabolism (insulin resistance,
overweight, etc.), cardiovascular system (hypertension),
loss of performance, GIT events, reproduction/sexual
repercussions, etc. (60). It should be considered, still,
that the sleep/wake cycle is only one among several
others affected systems in melatonin-decient adults
and children. Extend this to several of the circadian
aspects of physiology and the clinical picture becomes
much more serious, resulting in systemic repercussion
reaching every other aspect of human physiology and
behavior, jeopardizing its health and quality of life and
even longevity.
Funding statement: the present work was funded by São Paulo
Research Foundation – Fapesp (2014/50457-0).
Disclosure: no potential conict of interest relevant to this article
was reported.
REFERENCES
1. Vollrath L. The Pineal Organ. Mollendorff WaB, W., editor. Heild-
berg, Germany: Springer-Verlag; 1981. p. 659
2. Kappers JA. The development, topographical relations and in-
nervation of the epiphysis cerebri in the albino rat. Z Zellforsch
Mikrosk Anat. 1960;52:163-215.
3. Afeche SC, do Amaral FG, Villela DCM, Abrahão MV, Peres R,
Cipolla-Neto J. Melatonin and the Pineal Gland. In: Romano E, De
Luca S, editors. New Research on Neurosecretory Systems. New
York: Nova Biomedical Books; 2008. p. 151-77.
4. Reiter RJ. Pineal melatonin: cell biology of its synthesis and of its
physiological interactions. Endocr Rev. 1991;12(2):151-80.
5. Hardeland R. Taxon- and Site-Specic Melatonin Catabolism.
Molecules. 2017; 22(11):pii: E2015.
6. Hardeland R, Balzer I, Poeggeler B, Fuhrberg B, Uria H, Behrmann
G, et al. On the primary functions of melatonin in evolution: me-
diation of photoperiodic signals in a unicell, photooxidation, and
scavenging of free radicals. J Pineal Res. 1995;18(2):104-11.
Copyright© AE&M all rights reserved.
478
Melatonin hormonal ways of action
Arch Endocrinol Metab. 2018;62/4
7. Jockers R, Delagrange P, Dubocovich ML, Markus RP, Renault N,
Tosini G, et al. Update on melatonin receptors: IUPHAR Review
20. Br J Pharmacol. 2016;173(18):2702-25.
8. Cipolla-Neto J, Amaral FG. Melatonin as an hormone: New physi-
ological and clinical insights. Endocrine Reviews. 2018, in press.
DOI 10.1210/er.2018-00084.
9. Hazlerigg DG, Gonzalez-Brito A, Lawson W, Hastings MH, Morgan
PJ. Prolonged exposure to melatonin leads to time-dependent
sensitization of adenylate cyclase and down-regulates melatonin
receptors in pars tuberalis cells from ovine pituitary. Endocrinol-
ogy. 1993;132(1):285-92.
10. von Gall C, Garabette ML, Kell CA, Frenzel S, Dehghani F, Schumm-
Draeger PM, et al. Rhythmic gene expression in pituitary depends
on heterologous sensitization by the neurohormone melatonin.
Nat Neurosci. 2002;5(3):234-8.
11. Valenti S, Guido R, Giusti M, Giordano G. In vitro acute and pro-
longed effects of melatonin on puried rat Leydig cell steroido-
genesis and adenosine 3’,5’-monophosphate production. Endo-
crinology. 1995;136(12):5357-62.
12. Witt-Enderby PA, Masana MI, Dubocovich ML. Physiological
exposure to melatonin supersensitizes the cyclic adenosine
3’,5’-monophosphate-dependent signal transduction cascade in
Chinese hamster ovary cells expressing the human mt1 melato-
nin receptor. Endocrinology. 1998;139(7):3064-71.
13. Kemp DM, Ubeda M, Habener JF. Identication and functional
characterization of melatonin Mel 1a receptors in pancreatic beta
cells: potential role in incretin-mediated cell function by sensitiza-
tion of cAMP signaling. Mol Cell Endocrinol. 2002;191(2):157-66.
14. Nagy AD, Iwamoto A, Kawai M, Goda R, Matsuo H, Otsuka T, et
al. Melatonin adjusts the expression pattern of clock genes in the
suprachiasmatic nucleus and induces antidepressant-like effect
in a mouse model of seasonal affective disorder. Chronobiol Int.
2015;32(4):447-57.
15. Kandalepas PC, Mitchell JW, Gillette MU. Melatonin Signal
Transduction Pathways Require E-Box-Mediated Transcription
of Per1 and Per2 to Reset the SCN Clock at Dusk. PLoS One.
2016;11(6):e0157824.
16. de Farias Tda S, de Oliveira AC, Andreotti S, do Amaral FG, Ch-
imin P, de Proenca AR, et al. Pinealectomy interferes with the cir-
cadian clock genes expression in white adipose tissue. J Pineal
Res. 2015;58(3):251-61.
17. Coelho LA, Peres R, Amaral FG, Reiter RJ, Cipolla-Neto J. Daily
differential expression of melatonin-related genes and clock
genes in rat cumulus-oocyte complex: changes after pinealec-
tomy. J Pineal Res. 2015;58(4):490-9.
18. Valenzuela FJ, Torres-Farfan C, Richter HG, Mendez N, Campino C,
Torrealba F, et al. Clock gene expression in adult primate supra-
chiasmatic nuclei and adrenal: is the adrenal a peripheral clock
responsive to melatonin? Endocrinology. 2008;149(4):1454-61.
19. Hiragaki S, Baba K, Coulson E, Kunst S, Spessert R, Tosini G. Mel-
atonin signaling modulates clock genes expression in the mouse
retina. PLoS One. 2014;9(9):e106819.
20. Zeman M, Herichova I. Melatonin and clock genes expression in
the cardiovascular system. Front Biosci (Schol Ed). 2013;5:743-53.
21. Takahashi JS. Finding new clock components: past and future. J
Biol Rhythms. 2004;19(5):339-47.
22. Arendt J, Broadway J. Light and melatonin as zeitgebers in man.
Chronobiol Int. 1987;4(2):273-82.
23. Arendt J, Skene DJ. Melatonin as a chronobiotic. Sleep Med Rev.
2005;9(1):25-39.
24. Lewy AJ, Ahmed S, Jackson JM, Sack RL. Melatonin shifts human
circadian rhythms according to a phase-response curve. Chrono-
biol Int. 1992;9(5):380-92.
25. Lewy AJ. Clinical applications of melatonin in circadian disor-
ders. Dialogues Clin Neurosci. 2003;5(4):399-413.
26. Robinson JE, Karsch FJ. Photoperiodic history and a changing
melatonin pattern can determine the neuroendocrine response
of the ewe to daylength. J Reprod Fertil. 1987;80(1):159-65.
27. Lewis JE, Ebling FJ. Tanycytes As Regulators of Seasonal Cycles
in Neuroendocrine Function. Front Neurol. 2017;8:79.
28. Ebling FJP, Lewis JE. Tanycytes and hypothalamic control of en-
ergy metabolism. Glia. 2018;66(6):1176-84.
29. Dopico XC, Evangelou M, Ferreira RC, Guo H, Pekalski ML, Smyth
DJ, et al. Widespread seasonal gene expression reveals annual
differences in human immunity and physiology. Nat Commun.
2015;6:7000.
30. Roenneberg T, Aschoff J. Annual rhythm of human reproduction:
II. Environmental correlations. J Biol Rhythms. 1990;5(3):217-39.
31. Sivan Y, Laudon M, Tauman R, Zisapel N. Melatonin production
in healthy infants: evidence for seasonal variations. Pediatr Res.
2001;49(1):63-8.
32. Wehr TA. Photoperiodism in humans and other primates: evi-
dence and implications. J Biol Rhythms. 2001;16(4):348-64.
33. Arendt J, Middleton B. Human seasonal and circadian stud-
ies in Antarctica (Halley, 75 degrees S). Gen Comp Endocrinol.
2018;258:250-8.
34. Tamura H, Nakamura Y, Terron MP, Flores LJ, Manchester LC, Tan
DX, et al. Melatonin and pregnancy in the human. Reprod Toxicol.
2008;25(3):291-303.
35. Klein DC. Evidence for the placental transfer of 3 H-acetyl-melato-
nin. Nat New Biol. 1972;237(73):117-8.
36. Reppert SM, Chez RA, Anderson A, Klein DC. Maternal-fetal
transfer of melatonin in the non-human primate. Pediatr Res.
1979;13(6):788-91.
37. Okatani Y, Okamoto K, Hayashi K, Wakatsuki A, Tamura S, Sagara
Y. Maternal-fetal transfer of melatonin in pregnant women near
term. J Pineal Res. 1998;25(3):129-34.
38. Mendez N, Abarzua-Catalan L, Vilches N, Galdames HA, Spich-
iger C, Richter HG, et al. Timed maternal melatonin treatment re-
verses circadian disruption of the fetal adrenal clock imposed by
exposure to constant light. PLoS One. 2012;7(8):e42713.
39. Seron-Ferre M, Mendez N, Abarzua-Catalan L, Vilches N, Valenzu-
ela FJ, Reynolds HE, et al. Circadian rhythms in the fetus. Mol Cell
Endocrinol. 2012;349(1):68-75.
40. Seron-Ferre M, Valenzuela GJ, Torres-Farfan C. Circadian clocks
during embryonic and fetal development. Birth Defects Res C Em-
bryo Today. 2007;81(3):204-14.
41. Weaver DR, Reppert SM. Maternal melatonin communicates
daylength to the fetus in Djungarian hamsters. Endocrinology.
1986;119(6):2861-3.
42. Weaver DR, Keohan JT, Reppert SM. Denition of a prenatal sen-
sitive period for maternal-fetal communication of day length. Am
J Physiol. 1987;253(6 Pt 1):E701-4.
43. Motta-Teixeira LC, Machado-Nils AV, Battagello DS, Diniz GB,
Andrade-Silva J, Silva-Junior S, et al. The absence of maternal
pineal melatonin rhythm during pregnancy and lactation impairs
offspring physical growth, neurodevelopment, and behavior.
Horm Behav. 2018 Aug 13. DOI: S0018-506X(17)30530-5.
44. Amaral FG, Castrucci AM, Cipolla-Neto J, Poletini MO, Mendez
N, Richter HG, et al. Environmental control of biological rhythms:
effects on development, fertility and metabolism. J Neuroendo-
crinol. 2014;26(9):603-12.
45. Amaral FG, Turati AO, Barone M, Scialfa JH, do Carmo Buon-
glio D, Peres R, et al. Melatonin synthesis impairment as a new
deleterious outcome of diabetes-derived hyperglycemia. J Pineal
Res. 2014;57(1):67-79.
46. Wetterberg L, Bratlid T, von Knorring L, Eberhard G, Yuwiler A. A
multinational study of the relationships between nighttime uri-
nary melatonin production, age, gender, body size, and latitude.
Eur Arch Psychiatry Clin Neurosci. 1999;249(5):256-62.
Copyright© AE&M all rights reserved.
479
Melatonin hormonal ways of action
Arch Endocrinol Metab. 2018;62/4
47. Rommel T, Demisch L. Inuence of chronic beta-adrenoreceptor block-
er treatment on melatonin secretion and sleep quality in patients with
essential hypertension. J Neural Transm Gen Sect. 1994;95(1):39-48.
48. Cox MA, Davis M, Voin V, Shoja M, Oskouian RJ, Loukas M, et
al. Pineal Gland Agenesis: Review and Case Illustration. Cureus.
2017;9(6):e1314.
49. Hanish AE, Butman JA, Thomas F, Yao J, Han JC. Pineal hypopla-
sia, reduced melatonin and sleep disturbance in patients with
PAX6 haploinsufciency. J Sleep Res. 2016;25(1):16-22.
50. Veatch OJ, Pendergast JS, Allen MJ, Leu RM, Johnson CH, Elsea
SH, et al. Genetic variation in melatonin pathway enzymes in chil-
dren with autism spectrum disorder and comorbid sleep onset
delay. J Autism Dev Disord. 2015;45(1):100-10.
51. Arendt J, Bhanji S, Franey C, Mattingly D. Plasma melatonin lev-
els in anorexia nervosa. Br J Psychiatry. 1992;161:361-4.
52. Tarquini R, Bruni V, Perfetto F, Bigozzi L, Tapparini L, Tarquini B.
Hypermelatoninemia in women with polycystic ovarian syn-
drome. Eur J Contracept Reprod Health Care. 1996;1(4):349-50.
53. Luboshitzk y R, Shen-Orr Z, Ishai A, Lavie P. Melatonin hyper-
secretion in male patients with adult-onset idiopathic hypo-
gonadotropic hypogonadism. Exp Clin Endocrinol Diabetes.
2000;108(2):142-5.
54. Duman O, Durmaz E, Akcurin S, Serteser M, Haspolat S. Sponta-
neous endogenous hypermelatoninemia: a new disease? Horm
Res Paediatr. 2010;74(6):444-8.
55. Bonnefond A, Froguel P. The case for too little melatonin signal-
ling in increased diabetes risk. Diabetologia. 2017;60(5):823-5.
56. Tarnowski M, Malinowski D, Safranow K, Dziedziejko V, Pawlik A.
MTNR1A and MTNR1B gene polymorphisms in women with ges-
tational diabetes. Gynecol Endocrinol. 2017;33(5):395-8.
57. Barboni MTS, Bueno C, Nagy BV, Maia PL, Vidal KSM, Alves RC,
et al. Melanopsin System Dysfunction in Smith-Magenis Syn-
drome Patients. Invest Ophthalmol Vis Sci. 2018;59(1):362-9.
58. Eckel RH, Depner CM, Perreault L, Markwald RR, Smith MR,
McHill AW, et al. Morning Circadian Misalignment during Short
Sleep Duration Impacts Insulin Sensitivity. Curr Biol. 2015;25(22):
3004-10.
59. Harpsoe NG, Andersen LP, Gogenur I, Rosenberg J. Clinical phar-
macokinetics of melatonin: a systematic review. Eur J Clin Phar-
macol. 2015;71(8):901-9.
60. Arora T, Chen MZ, Cooper AR, Andrews RC, Taheri S. The Impact
of Sleep Debt on Excess Adiposity and Insulin Sensitivity in
Patients with Early Type 2 Diabetes Mellitus. J Clin Sleep Med.
2016;12(5):673-80.
... Ello podría deberse a su acción antioxidante frente a las especies reactivas de oxígeno (ROS), inhibiendo el poro de transición de la permeabilidad mitocondrial y activando las proteínas desacopladoras 11 . Aproximadamente 30 µg/día de melatonina se secretan constantemente en humanos adultos 1 . En individuos sanos, la síntesis de melatonina se inicia al oscurecer, siguiendo el ritmo circadiano, entre las 8 horas p.m. y las 10 horas p.m., alcanzando unos niveles máximos plasmáticos durante la noche, entre las 2-4 horas de la madrugada. ...
... Después, la síntesis de melatonina decrece progresiva-mente hasta alcanzar valores en adultos mayores de 29,2 ± 6,1 pg/ml. Estas concentraciones bajas de melatonina podrían contribuir al empeoramiento progresivo de la función orgánica asociada al envejecimiento 1,7,14 (figura 3). ...
... La localización anatómica estratégica de la glándula pineal y su alta vascularización adyacente permiten a la melanocortina secretarse y acceder rápidamente al torrente sanguíneo y al líquido cefalorraquídeo, lo que le facilita penetrar a los tejidos profundos del SNC y al resto de los tejidos periféricos 1,6 . Debido a su alta solubilidad, por su propiedad anfipática y su bajo peso molecular, tiene una elevada capacidad de difusión intracelular y extracelular, siendo capaz de cruzar las membranas celulares y la barrera hematoencefálica 6,15 . ...
Article
Full-text available
MMelatonin (N-acetyl-5 methoxy-tryptamine) is an indolic compound present in almost all fungi, plants, and animals. This neurohormone is synthesized and secreted into the internal environment mainly by the pineal gland, although there are other non-endocrine extrapineal locations. Its biosynthesis varies substantially depending on chronological age; it is between 4-7 years when its maximum concentration (329.5 ± 42.0 pg/ml) then progressively decreases until reaching minimum values (29.2 ± 6.1 pg/ml). ml) in older adults that would contribute to the progressive worsening of organic function. Eggs, fish, nuts, cereals, seeds, fruits and legumes are excellent dietary sources of melatonin increase its plasma concentration, but also as a dietary supplement with a health claim, according to the US Food and Drug Administration, and even the European Agency. of the Medicine authorized its marketing as a drug. This molecule of pleiotropic bioactions by interaction with its membrane receptors and cell nucleus located in multiple tissues exerts, in addition to regulating the circadian rhythm, antioxidant, anti-inflammatory, immunostimulant, cardioprotective, antidiabetic, antiobesity, neuroprotective, anti-aging actions and against several types of cancer. Its anticancer activity includes antioxidant mechanisms, modulation of MT1/MT2 receptors, apoptotic stimulation, pro-survival signaling, angiogenic and metastatic inhibition, and induction of epigenetic alterations. Knowing the pathways of action that support anticancer efficacy, which would make melatonin a potential adjuvant to oncological therapies, could be useful for clinical nutrition professionals by expanding their therapeutic action because it is feasible to increase its plasma concentration by incorporating it into the nutritional strategy. rich foods or nutritional supplements
... The light cycle plays an important role in regulating the circadian rhythm. Melatonin is programmed to be synthesized at night by the suprachiasmatic nucleus in sync with the light-dark cycle via the retino-hypothalamic pathway [8][9][10]. The circadian rhythm of melatonin and the influence of human sleep have been extensively studied by many authors [11,12]. ...
... Melatonin (N-acetyl-5-methoxytryptamine) is a neurohormone produced by the pineal gland in the human brain. Is a ubiquitous biomolecule in nature, also produced by other species of animals, plants, and microorganisms [114]. Additionally, this hormone is synthesized in human ocular tissues, including the retina and ciliary body, which have specific melatonin receptors [115]. ...
Article
Full-text available
The majority of neurodegenerative eye disorders occur with aging and significantly impair quality of life. Age-related macular degeneration (AMD) is the third most common cause of visual impairment and blindness worldwide. One of the most important elements in the pathophys-iology of neurodegenerative eye disease is certainly oxidative stress, with neuroinflammation and ocular ischemia which may also be significant factors. Antioxidants, either by food or oral supple-mentation, may be able to mitigate the deleterious effects of reactive oxygen species that build as a result of oxidative stress, ischemia, and inflammation. Over the past few decades, a number of research works examining the potential adjuvant impact of antioxidants in AMD have been published. In fact, there is not only more and more interest in already known molecules but also in new molecules that can help clinicians in the management of this complex multifactorial disease, such as astaxanthin and melatonin. However, while some studies showed encouraging outcomes, others were conflicting. In addition, more and more attention is also being paid to nutrition, considered a pivotal key point, especially to prevent AMD. For this reason, the purpose of this review is to analyze the main antioxidant molecules currently used as oral supplements for AMD treatment, as well as the role of diet and food intake in this ocular disease, to better understand how all these factors can improve the clinical management of AMD patients.
... A regulação da síntese de melatonina está relacionada com a ausência de luz ambiental (Santos, 2019), sendo sua maior produção durante o período noturno (Neto, 2008;Araujo et al., 2019). Assim, a presença de luz é capaz de bloquear a secreção de melatonina, pois inibe a função da glândula pineal (Amaral & Cipolla-Neto, 2018). ...
Chapter
Melatonin (MT) from plant species has attracted attention in the past decade. It is still the only conserved molecule that is broad spectrum as a regulator of biological functions since it has been identified in the unicellular alga, higher plants, and animals. In plants, MT is produced from tryptophan converted to tryptamine in the chloroplast. Subsequent pathways involve the conversion of tryptamine into serotonin, which can form methoxytryptamine and N-acetyl-serotonin resulting in MT. MT has been documented to play a crucial role in governing various aspects of plant growth and development, including the regulation of circadian rhythms and photoperiodic responses. Moreover, it has been found to contribute significantly to antioxidation and stress resistance in plants. Its use as an anti-inflammatory and immunomodulated drug against human diseases also gained significant attention in humans. There is an increasingly shared agreement that MT is valuable in maintaining body functions and homeostasis, particularly during adulthood, owing to its antioxidant, anti-inflammatory, and immunomodulatory properties. MT can mitigate symptoms such as cough, dyspnea, and fatigue and shorten hospitalization time in COVID patients. Moreover, it could reduce thrombotic and septic processes. In Alzheimer’s patients, it improves sleep quality and cognitive function. In Parkinson’s disease, it can enhance sleep quality and non-motor symptoms and reduce oxidant marker levels. In multiple sclerosis it improves sleep time and lowers fatigue scores and inflammatory and oxidative scenarios. In polycystic ovary syndrome, MT reduced insulin, HOMA, LDL-c, body weight, waist circumference, and TNF-α levels. In dermatitis, it led to fewer signs of dermatitis compared to placebo. It decreased the degree of liver fat compared to the placebo group in non-alcoholic fat liver disease.
Article
The study provides literature data on the impact of melatonin on blood pressure (BP) in elderly individuals with arterial hypertension (AH). AH is a significant risk factor for cardiovascular diseases (CVDs) and is a key focus of medical research. Melatonin, which plays a crucial role in the body's regulatory mechanisms, particularly when its production is disrupted, can significantly influence the development of AH. Melatonin is not a specific treatment for a particular disease but a multifunctional element that helps maintain homeostasis. Combining melatonin with antihypertensive therapy in elderly individuals with AH has been found to lead to more effective treatment outcomes. Exploring the effects of melatonin on BP levels in elderly individuals with AH is of great scientific interest, especially in understanding the relationship between individual melatonin secretion characteristics and predisposition to cardiovascular complications. This topic requires further research for a deeper understanding. Over 400 sources were analyzed on various aspects of the impact of melatonin on BP levels using materials from the PubMed and Google Scholar electronic databases. Out of these, 44 sources were selected and included in the study, considering relevant keywords. Keywords: Melatonin, blood pressure, arterial hypertension, cardiovascular disease, old age.
Article
Full-text available
The circadian clock regulates biological cycles across species and is crucial for physiological activities and biochemical reactions, including cancer onset and development. The interplay between the circadian rhythm and cancer involves regulating cell division, DNA repair, immune function, hormonal balance, and the potential for chronotherapy. This highlights the importance of maintaining a healthy circadian rhythm for cancer prevention and treatment. This article investigates the complex relationship between the circadian rhythm and cancer, exploring how disruptions to the internal clock may contribute to tumorigenesis and influence cancer progression. Numerous databases are utilized to conduct searches for articles, such as NCBI, MEDLINE, and Scopus. The keywords used throughout the academic archives are “circadian rhythm”, ”cancer”, and ”circadian clock”. Maintaining a healthy circadian cycle involves prioritizing healthy sleep habits and minimizing disruptions, such as consistent sleep schedules, reduced artificial light exposure, and meal timing adjustments. Dysregulation of the circadian clock gene and cell cycle can cause tumor growth, leading to the need to regulate the circadian cycle for better treatment outcomes. The circadian clock components significantly impact cellular responses to DNA damage, influencing cancer development. Understanding the circadian rhythm’s role in tumor diseases and their therapeutic targets is essential for treating and preventing cancer. Disruptions to the circadian rhythm can promote abnormal cell development and tumor metastasis, potentially due to immune system imbalances and hormonal fluctuations.
Article
Full-text available
Melatonin (N-acetyl-5 methoxytryptamine) is an indolic compound present in almost all fungi, plants, and animals. This neurohormone is synthesized and secreted into the internal environment mainly by the pineal gland, present in most vertebrates. Non-endocrine extrapineal locations have not been documented. This molecule with pleiotropic bioactions regulates the circadian rhythm, antioxidant, anti-inflammatory, immunostimulant, cardioprotective, antidiabetic, antiobesity, neuroprotective, and antiaging actions. Furthermore, in recent years, many studies have described the key role of melatonin in the prevention and development of cancer. The objective of this narrative review is to describe the different mechanisms through which melatonin exerts its action as an adjuvant in the modulation of carcinogenesis. The general anticarcinogenic mechanisms include epigenetic control, modulation of cell proliferation, regulation of cell cycle, induction of apoptosis, and telomerase inhibition. Melatonin also exerts antiestrogenic activity, which is particularly significant in hormone-dependent tumors, regulating the expression and transactivation of the estrogen receptor, and modulating the enzymes involved in the local synthesis of 22 estrogens. Modulation of metastasis by melatonin includes increased expression of cell adhesion molecules such as E-cadherin and β1-integrin, inhibition of angiogenesis, and control of fat metabolism by inhibiting the uptake of fatty acids by membrane transporters. Finally, immunomodulatory properties include enhanced production of anti-inflammatory interleukins and other cytokines in lymphocytes and monocytes and modulation of antioxidant activity by neutralizing free radicals. Despite all the mentioned properties, the use of melatonin in daily clinical practice is very limited, and additional studies are needed to better establish the role of this hormone in oncological clinical applications against different types of cancer.
Article
Full-text available
Melatonin is a ubiquitous molecule present in almost every live being from bacteria to humans. In vertebrates, besides being produced in peripheral tissues and acting as an autocrine and paracrine signal, melatonin is centrally synthetized by a neuroendocrine organ, the pineal gland. Independently of the considered specie, pineal hormone melatonin is always produced during the night and its production and secretory episode duration is directly dependent on the length of the night. As its production is tightly linked to the light/dark cycle, melatonin main hormonal systemic integrative action is to coordinate behavioral and physiological adaptations to the environmental geophysical day and season. The circadian signal is dependent on its daily production regularity, on the contrast between day and night concentrations and on specially developed ways of action. During its daily secretory episode, melatonin coordinates the night adaptive physiology through immediate effects and primes the day adaptive responses through prospective effects that will only appear at daytime, when melatonin is absent. Similarly, the annual history of the daily melatonin secretory episode duration primes the central nervous/endocrine system to the seasons to come. Remarkably, maternal melatonin programs the fetuses’ behavior and physiology to cope with the environmental light/dark cycle and season after birth. These unique ways of action turn melatonin into a biological time-domain acting molecule. The present review focus on the above considerations, proposes a putative classification of clinical melatonin dysfunctions and discuss general guidelines to the therapeutic use of melatonin.
Article
Full-text available
Maternal melatonin provides photoperiodic information to the fetus and thus influences the regulation and timing of the offspring's internal rhythms and preparation for extra-uterine development. There is clinical evidence that melatonin deprivation of both mother and fetus during pregnancy, and of the neonate during lactation, results in negative long-term health outcomes. As a consequence, we hypothesized that the absence of maternal pineal melatonin might determine abnormal brain programming in the offspring, which would lead to long-lasting implications for behavior and brain function. To test our hypothesis, we investigated in rats the effects of maternal melatonin deprivation during gestation and lactation (MMD) to the offspring and the effects of its therapeutic replacement. The parameters evaluated were: (1) somatic, physical growth and neurobehavioral development of pups of both sexes; (2) hippocampal-dependent spatial learning and memory of the male offspring; (3) adult hippocampal neurogenesis of the male offspring. Our findings show that MMD significantly delayed male offspring's onset of fur development, pinna detachment, eyes opening, eruption of superior incisor teeth, testis descent and the time of maturation of palmar grasp, righting reflex, free-fall righting and walking. Conversely, female offspring neurodevelopment was not affected. Later on, male offspring show that MMD was able to disrupt both spatial reference and working memory in the Morris Water Maze paradigm and these deficits correlate with changes in the number of proliferative cells in the hippocampus. Importantly, all the observed impairments were reversed by maternal melatonin replacement therapy. In summary, we demonstrate that MMD delays the appearance of physical features, neurodevelopment and cognition in the male offspring, and points to putative public health implications for night shift working mothers.
Preprint
Full-text available
Purpose: Smith-Magenis syndrome (SMS) causes sleep disturbance that is related to an abnormal melatonin profile. It is not clear how the genomic disorder leads to a disturbed synchronization of the sleep/wake rhythm in SMS patients. To evaluate the integrity of the intrinsically photosensitive retinal ganglion cell (ipRGC)/melanopsin system, the transducers of the light-inhibitory effect on pineal melatonin synthesis, we recorded pupillary light responses (PLR) in SMS patients. Methods: Subjects were SMS patients (n = 5), with molecular diagnosis and melatonin levels measured for 24 hours and healthy controls (n = 4). Visual stimuli were 1-second red light flashes (640 nm; insignificant direct ipRGC activation), followed by a 470-nm blue light, near the melanopsin peak absorption region (direct ipRGC activation). Blue flashes produce a sustained pupillary constriction (ipRGC driven) followed by baseline return, while red flashes produce faster recovery. Results: Pupillary light responses to 640-nm red flash were normal in SMS patients. In response to 470-nm blue flash, SMS patients had altered sustained responses shown by faster recovery to baseline. SMS patients showed impairment in the expected melatonin production suppression during the day, confirming previous reports. Conclusions: SMS patients show dysfunction in the sustained component of the PLR to blue light. It could explain their well-known abnormal melatonin profile and elevated circulating melatonin levels during the day. Synchronization of daily melatonin profile and its photoinhibition are dependent on the activation of melanopsin. This retinal dysfunction might be related to a deficit in melanopsin-based photoreception, but a deficit in rod function is also possible.
Article
Full-text available
Melatonin is catabolized both enzymatically and nonenzymatically. Nonenzymatic processes mediated by free radicals, singlet oxygen, other reactive intermediates such as HOCl and peroxynitrite, or pseudoenzymatic mechanisms are not species- or tissue-specific, but vary considerably in their extent. Higher rates of nonenzymatic melatonin metabolism can be expected upon UV exposure, e.g., in plants and in the human skin. Additionally, melatonin is more strongly nonenzymatically degraded at sites of inflammation. Typical products are several hydroxylated derivatives of melatonin and N1-acetyl-N2-formyl-5-methoxykynuramine (AFMK). Most of these products are also formed by enzymatic catalysis. Considerable taxon- and site-specific differences are observed in the main enzymatic routes of catabolism. Formation of 6-hydroxymelatonin by cytochrome P450 subforms are prevailing in vertebrates, predominantly in the liver, but also in the brain. In pineal gland and non-mammalian retina, deacetylation to 5-methoxytryptamine (5-MT) plays a certain role. This pathway is quantitatively prevalent in dinoflagellates, in which 5-MT induces cyst formation and is further converted to 5-methoxyindole-3-acetic acid, an end product released to the water. In plants, the major route is catalyzed by melatonin 2-hydroxylase, whose product is tautomerized to 3-acetamidoethyl-3-hydroxy-5-methoxyindolin-2-one (AMIO), which exceeds the levels of melatonin. Formation and properties of various secondary products are discussed.
Research
Full-text available
Purpose : Smith-Magenis Syndrome (SMS) is a rare disease caused by a deletion (or a point mutation < 10% of the patients) in the gene RAI1 of chromosome 17. This gene plays an important role during the development of the brain. SMS patients show a disrupted sleep pattern caused by an alteration in daily profile of melatonin production that unusually peaks during the day. Here we used pupillometry in order to investigate the sustained component of the pupillary light response (PLR). This component reveals melanopsin activity since it is mediated by melanopsin expressing intrinsically photosensitive retinal ganglion cells (ipRGCs). Among several functions, the ipRGCs contribute to photopic regulation of melatonin and therefore to the regulation of circadian rhythms of sleep. Methods : PLRs were recorded monocularly (both eyes) in five SMS patients and three controls. A dark adaptation period of 10 minutes preceded PLR recordings in response to 1 second light flashes of different wavelengths: a 640 nm red light, which falls outside the absorption spectrum of melanopsin (no direct activation of the ipRGCs), followed by a 470 nm blue light, which is close to the peak absorption of melanopsin (direct activation of the ipRGCs) at 100 2
Article
Full-text available
Agenesis of the pineal gland has rarely been reported in the medical literature. Herein, we report a cadaveric specimen found to have agenesis of the pineal gland. The remaining gross examination of the brain was normal. A review of the literature was performed on this unusual finding.
Article
Studies from a number of areas of neuroendocrinology indicate that hypothalamic tanycytes play a key role in control of energy metabolism. First, profound annual changes in gene expression have been identified in these unusual glial cells in seasonal mammals, for example in genes relating to the transport and metabolism of thyroid hormone into the hypothalamus. The consequent changes in local thyroid hormone availability in the hypothalamus have been shown experimentally to regulate annual cycles in energy intake, storage and expenditure in seasonal species. This is reflected in overt seasonal changes in appetite, body fat composition and torpor. Second, studies in laboratory rodents demonstrate that hypothalamic tanycytes possess transport mechanisms and receptors that indicate they have a cellular function as nutrient sensors. Ex vivo studies with organotypic tanycyte cultures confirm that acute changes in nutrient availability alter calcium and purinergic signalling within and between tanycytes. Finally, tanycytes are components of a stem cell niche in the hypothalamus whose activity can be regulated by the nutritional environment. Experimental depletion of cell division in the hypothalamus alters the homeostatic response to nutrient excess in mice raised in high fat diets. These convergent lines of evidence suggest that tanycytes are nutrient and metabolite sensors that impact upon plasticity and neuronal function in the surrounding hypothalamus, and consequently have an important role in energy intake and expenditure.
Article
It is well established that hypothalamic neurons producing the peptide corticotropin-releasing factor (CRF) play a key role in stress adaptation, including reduction of food intake when a threat or stressor is present. We have previously reported on the presence of an intrinsic CRF signaling system within the optic tectum (OT), a brain area that plays a key role in visually guided prey capture/predator avoidance decisions. To better understand the potential role of tectal CRF neurons in regulating adaptive behavior and energy balance during stress we examined evidence for modulation of tectal CRF neuronal activity after stressor exposure and food deprivation in the African clawed frog Xenopus laevis. We tested two predictions, 1) that exposure to categorically distinct stressors (ether vapors and shaking) will reduce food intake and modulate the activity of tectal CRF cells, and 2) that food deprivation will modulate the activity of tectal CRF cells. Exposure to ether increased tectal content of CRF and CRF transcript, but lowed CRFR1 transcript abundance. Two weeks of food deprivation reduced total fat stores in frogs and decreased tectal content of CRF content while having no effect on CRF and CRFR1 transcript abundance. Our data are consistent with a role for tectal CRF neurons in modulating food intake in response to certain stressors.
Article
Living for extended periods in Antarctica exposes base personnel to extremes of daylength (photoperiod) and temperature. At the British Antarctic Survey base of Halley, 75(0)S, the sun does not rise for 110 d in the winter and does not set for 100 d in summer. Photoperiod is the major time cue governing the timing of seasonal events such as reproduction in many species. The neuroendocrine signal providing photoperiodic information to body physiology is the duration of melatonin secretion which reflects the length of the night: longer in the short days of winter and shorter in summer. Light of sufficient intensity and spectral composition serves to suppress production of melatonin and to set the circadian timing and the duration of the rhythm. In humans early observations suggested that bright (>2000 lux) white light was needed to suppress melatonin completely. Shortly thereafter winter depression (Seasonal Affective Disorder or SAD) was described, and its successful treatment by an artificial summer photoperiod of bright white light, sufficient to shorten melatonin production. At Halley dim artificial light intensity during winter was measured, until 2003, at a maximum of approximately 500 lux in winter. Thus a strong seasonal and circadian time cue was absent. It seemed likely that winter depression would be common in the extended period of winter darkness and could be treated with an artificial summer photoperiod. These observations, and predictions, inspired a long series of studies regarding human seasonal and circadian status, and the effects of light treatment, in a small overwintering, isolated community, living in the same conditions for many months at Halley. We found little evidence of SAD, or change in duration of melatonin production with season. However the timing of the melatonin rhythm itself, and/or that of its metabolite 6-sulphatoxymelatonin (aMT6s), was used as a primary marker of seasonal, circadian and treatment changes. A substantial phase delay of melatonin in winter was advanced to summer phase by a two pulse 'skeleton' bright white light treatment. Subsequently a single morning pulse of bright white light was effective with regard to circadian phase and improved daytime performance. The circadian delay evidenced by melatonin was accompanied by delayed sleep (logs and actigraphy): poor sleep is a common complaint in Polar regions. Appropriate extra artificial light, both standard white, and blue enriched, present throughout the day, effectively countered delay in sleep timing and the aMT6s rhythm. The most important factor appeared to be the maximum light experienced. Another manifestation of the winter was a decline in self-rated libido (men only on base at this time). Women on the base showed lower aspects of physical and mental health compared to men. Free-running rhythms were seen in some subjects following night shift, but were rarely found at other times, probably because this base has strongly scheduled activity and leisure time. Complete circadian adaptation during a week of night shift, also seen in a similar situation on North Sea oil rigs, led to problems readapting back to day shift in winter, compared to summer. Here again timed light treatment was used to address the problem. Sleep, alertness and waking performance are critically dependent on optimum circadian phase. Circadian desynchrony is associated with increased risk of major disease in shift workers. These studies provide some groundwork for countering/avoiding circadian desynchrony in rather extreme conditions.