Abstract

BACKGROUND

Research within the last decade has shown melatonin to have previously-unsuspected beneficial actions on the peripheral reproductive organs. Likewise, numerous investigations have documented that stable circadian rhythms are also helpful in maintaining reproductive health. The relationship of melatonin and circadian rhythmicity to maternal and fetal health is summarized in this review.

METHODS

Databases were searched for the related published English literature up to 15 May 2013. The search terms used in various combinations included melatonin, circadian rhythms, biological clock, suprachiasmatic nucleus, ovary, pregnancy, uterus, placenta, fetus, pre-eclampsia, intrauterine growth restriction, ischemia-reperfusion, chronodisruption, antioxidants, oxidative stress and free radicals. The results of the studies uncovered are summarized herein.

RESULTS

Both melatonin and circadian rhythms impact reproduction, especially during pregnancy. Melatonin is a multifaceted molecule with direct free radical scavenging and indirect antioxidant activities. Melatonin is produced in both the ovary and in the placenta where it protects against molecular mutilation and cellular dysfunction arising from oxidative/nitrosative stress. The placenta, in particular, is often a site of excessive free radical generation due to less than optimal adhesion to the uterine wall, which leads to either persistent hypoxia or intermittent hypoxia and reoxygenation, processes that cause massive free radical generation and organ dysfunction. This may contribute to pre-eclampsia and other disorders which often complicate pregnancy. Melatonin has ameliorated free radical damage to the placenta and to the fetus in experiments using non-human mammals. Likewise, the maintenance of a regular maternal light/dark and sleep/wake cycle is important to stabilize circadian rhythms generated by the maternal central circadian pacemaker, the suprachiasmatic nuclei. Optimal circadian rhythmicity in the mother is important since her circadian clock, either directly or indirectly via the melatonin rhythm, programs the developing master oscillator of the fetus. Experimental studies have shown that disturbed maternal circadian rhythms, referred to as chronodisruption, and perturbed melatonin cycles have negative consequences for the maturing fetal oscillators, which may lead to psychological and behavioral problems in the newborn. To optimize regular circadian rhythms and prevent disturbances of the melatonin cycle during pregnancy, shift work and bright light exposure at night should be avoided, especially during the last trimester of pregnancy. Finally, melatonin synergizes with oxytocin to promote delivery of the fetus. Since blood melatonin levels are normally highest during the dark period, the propensity of childbirth to occur at night may relate to the high levels of melatonin at this time which work in concert with oxytocin to enhance the strength of uterine contractions.

CONCLUSIONS

A number of conclusions naturally evolve from the data summarized in this review: (i) melatonin, of both pineal and placental origin, has essential functions in fetal maturation and placenta/uterine homeostasis; (ii) circadian clock genes, which are components of all cells including those in the peripheral reproductive organs, have important roles in reproductive and organismal (fetal and maternal) physiology; (iii) due to the potent antioxidant actions of melatonin, coupled with its virtual absence of toxicity, this indoleamine may have utility in the treatment of pre-eclampsia, intrauterine growth restriction, placental and fetal ischemia/reperfusion, etc. (iv) the propensity for parturition to occur at night may relate to the synergism between the nocturnal increase in melatonin and oxytocin.

Introduction

Even before the discovery of melatonin in the pineal gland (Lerner et al., 1958), the organ was experimentally linked to reproduction, but the findings were unpersuasive (Quay, 1956; Mogler, 1958). The decade of the 1960s, however, was accompanied by a series of seminal studies which showed that the gland functioned as a photoneuroendocrine transducer (Quay, 1963; Axelrod et al., 1964, 1965) and it was definitively proved to have a physiological association with the hypothalamo-pituitary-gonadal axis (Hoffman and Reiter, 1965, 1966; Reiter and Hester, 1966). In particular, the daily changes in the duration of darkness under natural photoperiod conditions were found, via the pineal gland, to drive seasonal variations in reproductive capability in photosensitive species (Reiter, 1973, 1974). That melatonin was the mediator of pineal origin which determined the waxing and waning of reproductive competence under these conditions was uncovered shortly thereafter (Reiter et al., 1976; Tamarkin et al., 1976; Carter and Goldman, 1983).

In addition to its importance in intervening between the changing light:dark cycle and seasonal reproduction in photoperiodic species (Revel et al., 2009; Lincoln and Hazlerigg, 2010), melatonin has numerous other actions on the gonads and adnexae of non-seasonally breeding mammals, including the human, that are beneficial to optimal peripheral reproductive organ health (Reiter et al., 2009c, 2013a). The new data relating melatonin to pregnancy and delivery are summarized in this survey.

Research on circadian rhythms progressed concurrent with that of melatonin. Given that a normal light:dark cycle is of 24 h duration and has been for eons, it was advantageous for animals to modulate their metabolism, locomotor activity, etc. over a 24-h period to preserve energy, reduce predation pressure and avoid the likelihood of antagonistic cellular processes, e.g. the maximal activities of lipolytic and lipogenic enzymes in hepatocytes, from occurring simultaneously (Li et al., 2012; Thut et al., 2012). Thus, vertebrate species developed a light-sensitive biological clock whose function is governed by the prevailing light:dark cycle, the most regular recurring variable in the environment (Brainard et al., 1983, 1984; Meng et al., 2011; Ruger et al., 2013). This master clock is located in the suprachiasmatic nucleus (SCN) of the hypothalamus (Swanson and Cowan, 1975; Lydic et al., 1980). The SCN synchronizes many circadian rhythms in humans as in other vertebrates (Gillette and Tischkau, 1999; Hofman, 2000). Since the circadian production of melatonin in the pineal gland is also under the influence of the SCN, it is often difficult to separate the biological effects of general circadian disruption from those following from disturbances in the circadian melatonin cycle (Reiter et al., 2009b, 2012b).

In the following survey, the importance of the circadian clock to optimal reproductive physiology is often discussed separately from that due to alterations in melatonin (and vice versa). In reality, however, there is often no way of knowing whether a particular abnormal process relates to a derangement of the biological clock or to a perturbation or total suppression of the cyclic production of melatonin (Dumont et al., 2012).

Methods

The published literature utilized to compile this review was retrieved from a number of sources. Because of the relative diversity of subjects reviewed (i.e. mechanisms of melatonin synthesis in the pineal and in the placenta, peripheral melatonin receptors, free radical biology and the role of melatonin and its derivatives as free radical scavengers and as antioxidants, functional aspects of the central and peripheral circadian clocks, maturation and development of fetal physiology, diseases of pregnancy and the control of parturition), search terms were broad and numerous. These included, in various combinations, melatonin, circadian rhythms, biological clock, SCN, ovary, pregnancy, uterus, placenta, fetus, pre-eclampsia, intrauterine growth restriction, ischemia/reperfusion, chronodisruption, antioxidants oxidative stress and free radicals. All germane literature uncovered is summarized in this report.

Melatonin as a free radical scavenger and as an antioxidant

Whereas an extensive discussion of the actions of melatonin as a direct free radical scavenger (Tan et al., 1993; Galano et al., 2011) and as an indirect antioxidant (Barlow-Walden et al., 1995; Rodriguez et al., 2004) is beyond the scope of this review, the data overwhelmingly show that this indoleamine is highly effective in reducing oxidative stress throughout the body (Maldonado et al., 2007; de Matos et al., 2012; Tamura et al., 2013) including in the ovary, placenta, fetus and mother (see below). In studies where melatonin was compared with other better-known antioxidants (e.g. vitamins C, E, etc.) in terms of their protective efficiency against the damaging actions of toxic oxygen and nitrogen-based reactants, melatonin has proved more effective (Martin et al., 2000a; Reiter et al., 2009a; Milczarek et al., 2010). Recently, investigators also found that when melatonin was compared with chemically designed mitochondria-targeted antioxidants, again melatonin was superior in reducing molecular damage resulting from free radicals (Lowes et al., 2013).

There are several features that make melatonin highly efficient in protecting beleaguered macromolecules from oxidative/nitrosative stress (Fig. 1). First, there are no morphophysiological barriers to melatonin, e.g. it readily crosses the blood–brain barrier and the placenta (Schenker et al., 1998; Okatani et al., 1999), and so in addition to all maternal organs it also protects the placenta and the fetus. Melatonin readily gains access to the major sites of free radical generation, e.g. mitochondria. Besides being located in several subcellular compartments (Menendez-Pelaez and Reiter, 1993; Venegas et al., 2012), melatonin actually may be produced in the mitochondria (Tan et al., 2013) where free radical generation is especially high. Not only melatonin but also several of its metabolites that are formed when it functions as a direct free radical scavenger, i.e. cyclic 3-hydroxymelatonin, N1-acetyl-N2-formyl-5-methoxykynuramine (AFMK), N1-acetyl-5-methoxykynuramine (AMK), etc. are also radical scavengers (Tan et al., 2007; Hardeland et al., 2009; Hardeland, 2012) and some may be better at doing so than melatonin itself (Galano et al., 2013). Thus, melatonin is the progenitor of a variety of free radical scavengers which function in an antioxidant cascade to prevent radical-mediated damage (Fig. 2) (Tan et al., 2002). Finally, in terms of its indirect functions in limiting oxidative stress, melatonin stimulates several antioxidative enzymes (Barlow-Walden et al., 1995; Pablos et al., 1998; Rodriguez et al., 2004), likely via receptor-mediated processes (Tomas-Zapico and Coto-Montes, 2005). Its ubiquitous distribution, ease of transfer between subcellular compartments and broad spectrum capabilities make melatonin highly effective in combatting free radical damage in the reproductive system and elsewhere.

Figure 1

Pathways showing the conversion of molecular oxygen to reactive oxygen and nitrogen species. An estimated 1–4% of the O2 inhaled is eventually converted to reactive products. When produced in excess, free radicals and other derivatives of oxygen are highly destructive to peripheral reproductive tissues. A single electron reduction of melatonin generates the superoxide anion radical (O2•–) which is either quickly metabolized by superoxide dismutase (SOD) to hydrogen peroxide (H2O2) or it couples with nitric oxide (NO) to produce the peroxynitrite anion (ONOO). H2O2 is transformed to the hydroxyl radical (OH) in the presence of transition metals, represented here by iron. O2 can also be converted to singlet oxygen (1O2), a less common but toxic species. O2•–, 1O2, H2O2 and OH are often classified as reactive oxygen species (ROS), whereas NO and ONOO are classified as reactive nitrogen species (RNS). Of these products, the OH and ONOO are considered the most reactive and damaging. The items marked with an asterisk are scavenged by melatonin and/or its metabolites. The up and down arrows associated with the enzymes indicate whether the action of melatonin is either stimulatory or inhibitory to its activity, respectively. H2O2 is metabolized via several routes which eventually leads to the production of non-toxic species. CAT, catalase; GCL, glutamyl cysteine ligase; GPx, glutathione peroxidase; GRd, glutathione reductase; GSSG, glutathione disulfide; HOCl, hypochlorous acid; MPO, myeloperoxidase; NOS, nitric oxide synthase.

Figure 2

In what is referred to as the antioxidant cascade, melatonin and its metabolites function in the reduction of oxidative stress. Melatonin is a powerful antioxidant due to its ability to neutralize toxic-free radicals in both the mother and the fetus. The metabolism of melatonin is a continuum when it functions as a radical scavenger such that each metabolite, like melatonin, also detoxifies radical species. This pathway is referred to as melatonin's antioxidant cascade. In this process, not only melatonin—but also cyclic 3-hydroxymelatonin (cyclic-3OH-melatonin), N1-acetyl-N2-formyl-5-methoxykynuramine (known as AFMK) and N1-acetyl-5-methoxykynuramine (known as AMK)—all function as radical scavengers. Finally, preliminary findings indicate that the molecule(s) produced when AMK neutralizes radicals may also function as a scavenger(s). Among the metabolic products listed, cyclic-3OH-melatonin and AMK may actually be more effective radical scavengers than melatonin itself. Thus, melatonin could be referred to as a pro-antioxidant or pro-drug.

In addition to removing toxic oxygen and nitrogen species from cells by either directly quenching them or by metabolizing them to non-reactive products, melatonin also exhibits significant anti-inflammatory activity (Carrillo-Vico et al., 2005; Mauriz et al., 2013). Melatonin's efficacy as an anti-inflammatory agent stems from its ability to reduce gene expression and activities of inducible nitric oxide synthase and cyclo-oxygenase, and by limiting the production of a variety of pro-inflammatory molecules including prostanoids, leukotrienes, cytokines and adhesion molecules. Melatonin modulates the production of these factors by regulating several transcription factors including nuclear factor -κB, nuclear factor erythroid-2 related factor 2 and hypoxia-inducible factor (Mauriz et al., 2013). As a component of this process, melatonin reduces the recruitment of leukocytes to areas of injury; this also limits oxidative damage since leukocytes can be a major source of free radicals and, therefore, of the associated molecular damage.

While melatonin's multi-faceted antioxidant actions are of fundamental importance and were the presumed original functions of melatonin during evolution (Tan et al., 2013), it is not the exclusive role of melatonin to quench and/or neutralize free radicals (Reiter et al., 2010a; 2012a). Rather, this ubiquitously-acting molecule has a variety of other functions that have been well documented in mammals including man; some of these may involve not only changes in the absolute levels of melatonin but also perturbations of its circadian rhythm. Thus, changes in melatonin have been linked to sleep disturbances (Santhi et al., 2012), psychological depression (Cardinali et al., 2012), metabolism leading to body weight regulation (Fenn et al., 2011), attention deficit/hyperactivity disorder (Chaste et al., 2011) and many others. Research on melatonin and circadian rhythms is progressing at a rapid pace and it seems likely that the discoveries to date are only a fraction of the real functions of melatonin and circadian rhythmicity. How all these facets of melatonin and circadian rhythms impact reproductive physiology are only beginning to be uncovered.

Melatonin production and function in the placenta

Melatonin, N-acetyl-5-methoxytryptamine, is synthesized from tryptophan (Fig. 3) and was initially thought to be solely of pineal origin (Lerner et al., 1958). This idea was dispelled, however, when melatonin showed up in other organs along with the enzymes that produce it (Reiter et al., 2013b). While the pineal gland is the only organ in which melatonin has been found to be generated in a light:dark-dependent circadian manner (Axelrod et al., 1965; Panke et al., 1979), as reflected in the blood levels of this indole (Vaughan et al., 1976; Wehr, 1991; Kennaway et al., 1992), it is now clear that many, perhaps all, organs have acquired the ability to generate this important molecule. If melatonin is synthesized in mitochondria, as recently proposed (Tan et al., 2013), then its ubiquitous production in organisms would be assured since all eukaryotic cells are endowed with these energy-generating organelles. The advantage of mitochondria being a source of this antioxidant would certainly be fortuitous since these organelles are a major site of origin of damaging oxygen derivatives. Thus in this location, melatonin, due to its potent radical scavenging activity (Tan et al., 1993; Allegra et al., 2003; Reiter et al., 2009a; Hardeland, 2012), would be ideally situated to prevent these destructive agents from mutilating neighboring molecules. In essence, melatonin would provide on-site protection. Certainly, this is consistent with the high levels of melatonin in mitochondria (Venegas et al., 2012) and the ability of melatonin to protect this organelle from damage and preserve its physiology (Martin et al., 2000b; Acuna-Castroviejo et al., 2002, 2011; Lowes et al., 2013). The vastly different levels of melatonin in fluids and subcellular organelles have led to a debate about what constitutes a physiological concentration of melatonin (Reiter and Tan, 2003).

Figure 3

Conversion of the amino acid, tryptophan, to the indoleamine, melatonin, as documented in the pineal gland. This pathway is also utilized by a large number of other cells, including some in the peripheral reproductive system, to generate melatonin that is used locally to protect the cells from toxic-free radicals, among other functions. The villous cytotrophoblasts and the syncytiotrophoblasts from the human placenta also contain the two enzymes that metabolize serotonin to melatonin, i.e. serotonin N-acetyltransferase and N-acetylserotonin methyltransferase (also referred to as hydroxylindole-O-methyltransferase).

Specifically in the reproductive system, melatonin is reportedly produced in the ovary (Itoh et al., 1999), in the oocyte per se (Sakaguchi et al., 2013), in the cumulus cells surrounding the oocyte (El-Raey et al., 2011) and in the placenta (Lanoix et al., 2008). Moreover, melatonin levels are higher in the ovarian follicular fluid than they are in the general circulation (Brzezinski et al., 1987; Röunberg et al., 1990; Nakamura et al., 2003). In the follicular fluid, melatonin has access to the surrounding tissues where it may have the important function of protecting the oocyte from oxidative damage during both maturation and ovulation (Tamura et al., 2013), a time when free radical formation is elevated (Brannstrom and Norman, 1993; Behrman et al., 2001; Sugino, 2005). This idea is consistent with the observation that melatonin was effective in reducing molecular damage to human ova (Tamura et al., 2008a, b) that were used for IVF-embryo transfer. Just as melatonin prevents free radical damage to ova, it also does so for sperm (Ortiz et al., 2011; Reiter et al., 2013a).

The placental villous trophoblasts are not only a source of melatonin but also they contain the classic transmembrane receptors for the indole, i.e. MT1 and MT2 (Lanoix et al., 2008). To document this, villous cytotrophoblasts were isolated from human term placentas (37–41 weeks of gestation) after vaginal delivery. The two enzymes that convert the precursor, serotonin, to melatonin, i.e. arylalkylamine N-acetyltransferase (AANAT) and acetylserotonin methyltransferase (ASMT) (formerly known as hydroxyindole-O-methyltransferase) (Fig. 3) are expressed and are active in both cytotrophoblast and syncytiotrophoblast cells as well as in JEG-3 and BeWo choriocarcinoma cells, commonly used as in vitro models for human trophoblasts. Given that villous trophoblasts apparently have the capability of generating melatonin and they also possess membrane receptors, Lanoix et al. (2008) speculated that locally produced melatonin likely has paracrine, autocrine and/or intracrine actions in the placenta. In addition to working via MT1 and MT2 receptors, melatonin also could directly scavenge radicals and reduce oxidative damage to placental tissues, which would be important given the large number of radicals that are often produced in this tissue when its function becomes compromised (Pringle et al., 2010).

In the placenta, the primary villous trophoblasts include two distinct categories of cells, i.e. the mononuclear villous cytotrophoblasts (vCTB) and the multinucleated syncytiotrophoblast (STB). The fusion of the vCTB to form the STB is a highly regulated process. To ensure the transition of vCTB to the STB, the vCTB cells are generally not lost due to apoptosis. Based on the recent in vitro findings of Lanoix et al. (2012b), melatonin reduces the loss of human vCTB cells by seemingly increasing their resistance to apoptosis via the intrinsic apoptotic pathway. The suppression of the apoptotic response by melatonin in vCTB cells was shown to be a membrane receptor-mediated process, given that the addition of luzindole, an MT1/MT2 receptor antagonist, to the culture medium blocked the anti-apoptotic actions of melatonin in vCTB cells.

The STB is a non-proliferative tissue with a pro-apoptotic phenotype (Vaillancourt et al., 2009). Thus, throughout pregnancy, lost STB must be continually regenerated by fusion of vCTB cells. In the absence of the persistent input of vCTB cells into the STB, the latter would quickly be reduced in size since its life span is short (Huppertz and Kingdom, 2004; Vaillancourt et al., 2009). Because melatonin preserves the vCTB and thereby makes them available for formation of the STB, melatonin may have an important role in maintaining homeostasis in the placenta (Fig. 4). STB degeneration is a consequence of syncytial loss due to both the intrinsic and extrinsic pathways (Heazell et al., 2008). The specific actions of melatonin on the apoptotic processes of STB has yet to be defined; this action could be the same or different than melatonin's function at the level of the vCTB.

Figure 4

The functions of locally produced melotonin in the villous cytotrophoblasts of the placenta. The enzymatic machinery required for the synthesis of melatonin has been identified in the human villous cytotrophoblasts and in the syncytiotrophoblasts. Although clock genes have been identified in the placenta, whether there is a 24-h rhythm in melatonin production in this tissue is yet to be examined. The classic membrane receptors, MT1 and MT2, for this indoleamine are also present in the placenta. Locally generated melatonin likely functions in the protection of the placenta from oxidative stress employing both receptor-dependent and receptor-independent processes. Additionally, melatonin probably reduces the loss of villous cytotrophoblasts by preventing apoptosis of these cells; melatonin's anti-apoptotic actions are well known in normal cells. Via its influence on cell survival, melatonin presumably maintains the stable turnover of the syncytiotrophoblast. Whether melatonin has any actions at the level of the extravillous trophoblasts, especially during invasion of the uterine wall should be examined.

Interestingly, trophoblast-derived cancer cells respond differently to melatonin than do the vCTB cells. Several choriocarcinoma cell lines have been established. One of the best studied is the BeWo line (Lewis et al., 1994). BeWo cells also have the capability of fusing to form a syncytium and they synthesize melatonin and possess its receptors (Lanoix et al., 2006, 2008). As with numerous other cancer cells (Sainz et al., 2003; Uguz et al., 2012; Wang et al., 2012), BeWo cells respond to melatonin by undergoing apoptosis, a response that is opposite that of normal vCTB. Thus, the actions of melatonin in the placenta are clearly context specific. As already mentioned, it is common that melatonin preserves normal cells while promoting processes that cause degeneration and death of cancer cells (Choi et al., 2008; Proietti et al., 2013; Rodriguez et al., 2013).

Germane to a discussion of melatonin's action in the placenta is a report which claimed that night-time circulating melatonin levels are depressed in women suffering with severe pre-eclampsia when compared with those in women with a normal pregnancy or with mild pre-eclampsia (Nakamura et al., 2001). Since especially severe pre-eclampsia is believed to involve the excessive production of free radicals, the lower melatonin levels could be a consequence of a more rapid utilization of this free radical scavenger or a consequence of a reduced production. Also, given that the night-time level of melatonin was measured at a single time point in these three groups of women, the apparently lower values in those with severe pre-eclampsia could have been a result of a shifted nocturnal melatonin peak rather than an actual depression of its absolute concentration.

Consistent with their continuing interest in the relevance of melatonin to placental physiology, Lanoix et al. (2012b) compared the levels of melatonin, its precursor, its synthesizing enzymes and its membrane receptors in pre-eclamptic placentas with those from gestation-matched normotensive control placentas. They observed that relative to control tissues, pre-eclamptic placentas had reduced AANAT gene expression and enzyme activity along with lower ASMT activity. Correlating with these observations were dramatically depressed concentrations of melatonin and elevated levels of its precursor, serotonin. Finally, pre-eclampsia was associated with a significant reduction in both the MT1 and MT2 receptor in the placenta. In view of these outcomes, it seems clear that the markedly depressed level of melatonin in pre-eclamptic placentas is a consequence of impaired production of the indoleamine due to a major deficiency in the rate-limiting enzyme AANAT. The drop in melatonin in the pre-eclamptic placenta may help to explain the lower levels of blood melatonin in women with pre-eclampsia (Nakamura et al., 2001); however, this correlation implies that placenta-derived melatonin is normally released into the blood, perhaps especially in near-term pregnancy, when blood levels of the indoleamine reach their maximal values (Tamura et al., 2008a). Lanoix et al. (2012b) also suggested that depressed circulating melatonin levels during pregnancy may be a biomarker for the diagnosis of pre-eclampsia. They also surmise that melatonin may be useful as a treatment for pre-eclampsia.

Programming fetal circadian rhythmicity

The maternal circadian system is organized by the master clock, a small group of neurons located bilaterally at the base of the diencephalon/telencephalon junction in the anterior hypothalamus (Mason and Lincoln, 1976). These neurons, referred to as the SCN, regulate circadian rhythms of most if not all organs in the body. At the molecular level, the rhythms in the SCN are driven by a transcriptional/translational feedback loop that involves genes Per 1, Cry 1, Cry 2, Clock and Bmal1 (Bass and Takahashi, 2010); these rhythms in turn regulate circadian oscillations in peripheral organs (Menaker et al., 2013).

Clock genes have been described in two functionally distinct zones (junctional and labyrinth) of the rat placenta near the time of delivery (Wharfe et al., 2011). Clock gene expression was compared in placentas collected at four time points throughout a light:dark cycle (collected at 0800, 1400, 2000 and 0200 h). The findings revealed that although the canonical circadian genes were expressed in both placenta zones, the circadian changes were not robust nor were they well co-ordinated. Despite these data, Waddell et al. (2012) caution against discounting a potential role of circadian rhythms as a key component of the normal placental phenotype.

A primary entrainer of the cyclic function of the SCN is the light:dark cycle, as detected by the retinas. The eyes are connected to the SCN via a specialized group of axons from a select population of retinal ganglion cells (Sand et al., 2011; Lucas, 2013); the axons of these cells are referred to as the retinohypothalamic tract (Mason and Lincoln, 1976) and they course with the optic nerve to the SCN.

Axons of SCN cells project to adjacent hypothalamic neurons where they synchronize overt circadian rhythms, such as body temperature, sleeping/waking, feeding and adrenocorticotrophic hormone/corticosteroid. Additionally, via a more complex neural route involving central and peripheral sympathetic neurons, the SCN regulates the production and release of melatonin from the pineal gland (Stehle et al., 2011). Blood melatonin levels are typically 10–15 times higher at night than during the day as a result of the nocturnal production and release of the indole from the pineal gland. The maternal melatonin rhythm may have a major role in influencing the development of the fetus since unaltered melatonin readily crosses the placenta (Okatani et al., 1999; Schenker et al., 1998). Melatonin produced in organs other than the pineal gland is minimally released into the blood, but rather is used in the organ where it is synthesized as an autocoid or paracoid (Tan et al., 2003).

The circadian clocks in many peripheral organs, for example the kidney, adrenal gland, lung, are entrained by SCN-derived information supplied to them by their autonomic innervation (Bass and Takahashi, 2010). Also, daily fluctuations in body temperature and cortisol may aid in synchronizing the clocks of peripheral organs. Finally, the day/night changes in circulating melatonin cue peripheral organs of the prevailing light:dark environment (Pevet and Challet, 2011). Thus, the metabolism of cells throughout the body is exposed to a variety of circadian messages that serve to regulate gene expression. The expression of an estimated 5–15% of the genes in peripheral organs is circadian in nature (Richards and Gumz, 2012).

The maturation and synchronization of the fetal circadian system has eluded detailed examination to date. Evidence from studies conducted in humans and non-human primates has revealed entrained 24-h rhythms in fetal heart rate and respiratory movements during the latter half of pregnancy (Seron-Ferre et al., 2007). Whether the circadian system of the fetus, particularly in late pregnancy, has anything reminiscent of the pervasive influence of the maternal SCN is unknown. It has been demonstrated that the post-natal development of the peripheral circadian system in offspring of mothers lacking a functional master clock (due to lesions of the SCN or to gene knockouts) is unexpectedly relatively normal (Jud and Albrecht, 2006).

In those mammals (including the human) where studies have been done, the SCN is already morphologically distinguishable in the basal hypothalamus of the fetus by mid-gestation. Before birth it exhibits rhythms in mRNA for vasopressin (an important transmitter in the SCN) and cfos mRNA and protein (Reppert and Schwartz, 1983; Novakova et al., 2010). Moreover, the innervating fibers from the retinohypothalamic tract to the SCN are apparent before term delivery (Weinert, 2005). The oscillations that occur in the fetal SCN are typically of lower amplitude than those in the adult master clock (Watanabe et al., 2006). While the retinohypothalamic input to the SCN is apparent in the late-gestation fetus, the output of the fetal clock to the pineal gland seems to be incomplete until after birth (Reiter, 1991). Thus, there is no evidence that a fetal melatonin rhythm influences any aspect of fetal development since the pineal gland requires an innervation from the SCN via the peripheral sympathetic nervous system to produce melatonin.

Melatonin of maternal origin, however, does influence the fetus by virtue of its rhythm and the ease with which it crosses the placenta. Studies in the human have repeatedly confirmed that the cycle of melatonin in the maternal blood also occurs in the fetal circulation (Kennaway et al., 1996; Okatani et al., 1999). This rhythm, when abolished by exposure of the mother to constant light, changes the rhythmic expression in fetal clock genes; these changes are reversed when daily melatonin injections are given to the mother (Torres-Farfan et al., 2006). This documents that the fetal clock is imprinted by melatonin, which under normal circumstances is of maternal origin. The human fetus displays 24-h rhythms in temperature and oxygen consumption by 32–33 weeks of gestation (Bauer et al., 2009).

In addition to the maternal melatonin rhythm, judging from the cyclic fluctuation of cortisol levels in the umbilical artery and vein, the term fetus is also exposed to this rhythm (Seron-Ferre et al., 2001). The so-called fetal zone of the fetal adrenal gland also secretes large amounts of dehydroepiandrosterone sulfate (DHEAS), which causes a 24-h rhythm in the fetal circulation; DHEAS is a necessary precursor for placental estrogen production during pregnancy (Seron-Ferre et al., 2007). Whether there is any functional interaction of the fetal melatonin and DHEAS rhythms at the level of the fetal SCN or elsewhere is unknown.

Evidence accumulated to date suggests that the circadian architecture in the fetus, particularly in the SCN, is entrained by the maternal melatonin cycle (Fig. 5) and possibly to a lesser extent by manipulation of the feeding times (Novakova et al., 2010). As mentioned above, exposure of pregnant mothers to constant light disrupts the maternal activity cycle and suppresses the circadian melatonin rhythm; these perturbations impact gene expression in the fetal SCN. Studies in rodents have shown that maternal-generated rhythms, when disrupted during pregnancy, also alter the post-natal behavioral circadian rhythms in the offspring. For example, maternal pinealectomy, which abolishes the circadian melatonin cycle, significantly interferes with the drinking rhythm in the offspring while this cycle is restored when the pinealectomized mothers are given regular melatonin injections in late pregnancy (Bellavia et al., 2006). These observations emphasize the potential danger of constant light exposure, unusual light/dark cycles or night shift work for pregnant humans especially during the last trimester. This is certainly not trivial given that throughout the world an estimated one-fourth to one-third of female employees work at night and, moreover, considering the high degree of light pollution in urban areas, avoiding light at night sufficient to alter the function of the biological clock and circulating melatonin levels is becoming progressively more difficult.

Figure 5

Potential routes by which the photoperiod, melatonin and the maternal master circadian clock, the suprachiasmatic nuclei (SCN), influence the development and/or maturation of the SCN in the fetal brain. The rhythmic activity of the maternal SCN is governed by the light/dark cycle. In the maternal compartment (top), the SCN controls the circadian production and secretion of melatonin from the pineal gland (PG) and imposes its activity on clocks in peripheral organs directly via neural signals and perhaps indirectly via the 24-h melatonin rhythm. The maternal SCN also influences daily changes in body temperature and feeding rhythms. Melatonin is also produced in the placenta but whether it is generated in a circadian manner in this tissue has not been determined. Also, whether placenta-derived melatonin has any bearing on fetal maturation or development is unknown. In the fetal compartment (bottom), the development and maturation of the SCN may be influenced by maternal temperature and feeding cycles and by the maternal circadian rhythm of melatonin, which easily passes through the placenta to the fetal circulation. In the fetus, these processes may also influence the adrenal corticosteroid rhythm and those of the pituitary. These organs, in turn, influence rhythms of clocks in the peripheral organs. Given that the maternal master circadian clock (SCN) is under the strong influence of the light/dark environment, it may be important that pregnant females maintain a regular light/dark cycle, especially during the last trimester. RHT, retinohypothalamic tract.

There is now general agreement that maternal circadian rhythms are influential in the entrainment and programming of fetal and newborn circadian rhythms (Fig. 5), but what rhythms are affected may be species specific. While information in this field is still rudimentary, evidence has shown that disturbances of the fetal circadian system, regardless of the cause of those perturbations, have long-term consequences in the offspring. As an example, women who engage in shift work during pregnancy have an increased incidence of spontaneous abortions, premature deliveries and low birthweight infants (Zhu et al., 2004). Shift work greatly alters the melatonin cycle, the sleep/wake rhythm and feeding times which also could be instrumental in contributing to these complications. Exposure of rats to simulated shift work caused increased hyperleptinemia and adiposity of the offspring at 3 months after birth and altered glucose tolerance and insulin resistance, reminiscent of that seen in metabolic syndrome, when the offspring were 1 year of age (Varcoe et al., 2011). Clearly, the impact of a disturbed light:dark cycle in late pregnancy and during the perinatal period may have major effects on subsequent behavioral and metabolic functions (Ferreira et al., 2012). Given the remarkable rise in the diagnosis of metabolic syndrome, obesity, attention deficit-hyperactivity disorder, autism spectrum disorders, etc., it may be worthwhile to be more attentive to the light:dark environment during pregnancy (Hardeland et al., 2012). This is especially true since the frequency of these conditions has run in parallel with excessive use of light at night, a change that is difficult to avoid in current societies.

Offspring that are delivered prematurely are not exposed to a normal maternal melatonin rhythm that they would be if they were still in utero and they do not generate a melatonin rhythm on their own (Kennaway et al., 1992). Thus, preterm infants are deprived of exposure to the melatonin cycle during a critical interval of their development. How or whether the lack of exposure to this rhythm in these premature infants has consequences on any aspect of development remains unexamined. This problem could be potentially partially rectified if premature infants would be breast fed since a melatonin rhythm normally exists in human breast milk, with higher levels at night than during the day (Illnerova et al., 1993). This still would require, however, that the mother be in darkness for several hours before and at the time of night-time breast feeding and also the much lower levels of melatonin in the breast milk (relative to those in the blood) may render these concentrations insignificant in terms of programming the SCN of the newborn.

Pre-eclampsia and placental damage: protection by melatonin

Pre-eclampsia is a dangerous complication of pregnancy which causes significant maternal and fetal morbidity and death (Hung and Burton, 2006; Nelissen et al., 2011; Redman, 2011). It is commonly diagnosed in the latter half of pregnancy but it may involve poor placentation during the earliest stage of pregnancy (Fig. 6). It can progress to an even more serious condition, eclampsia, which is accompanied by life-threatening seizures.

Figure 6

The potential sequence of events that contribute to pre-eclampsia. The oxidative/nitrosative stress that occurs may be a result of permanent hypoxia or intermittent hypoxia/reoxygenation resulting from placental maldevelopment because of inadequate attachment of the placenta to the uterine wall. Melatonin, due to its potent antioxidative actions, reduces ischemia/reperfusion injury to the placenta under experimental conditions and could potentially alleviate some of the signs of pre-eclampsia for the same reason. Melatonin also has antihypertensive actions which may aid in lowering the blood pressure in pre-eclamptic women. Not illustrated in this figure are the negative effects that chronodisruption has during pregnancy (see text).

Pre-eclampsia has multiple facets with a commonly observed sign being hypertension. As the condition progresses, the damaged placenta releases molecules into the circulation which cause endothelial dysfunction leading to an elevation in blood pressure (Roberts et al., 1989; Roberts and Hubel, 2009) and eventually more systemic damage, for example at the level of the kidney: these latter changes initiate proteinuria as a result of glomerular endotheliosis (Gaber et al., 1994). Several features which characterize pre-eclampsia contribute to the pathophysiology of the disease including hypertension, inflammation and oxidative/nitrosative stress. The only effective treatment for pre-eclampsia is the delivery of the fetus and the placenta, although the condition can occur during the first 2 weeks after child birth.

The role of oxidative and/or nitrosative stress in pre-eclampsia is well accepted (Myatt et al., 1996; Hung and Burton, 2006), although it is debated as to what causes the generation of the toxic oxygen derivatives that contribute to molecular damage. One theory states that free radicals and other reactive species are a consequence of a compromised uteroplacental arterial flow because of the improper remodeling of the spiral arteries during placentation (Kaufmann et al., 2003). This leads to a low oxygen tension and a relative hypoxic state in the placenta. Another view is that it is not a matter of a chronically diminished blood flow to the placenta, but rather the problem resides with the intermittency of blood flow to this tissue. This results in intervals of ample arterial blood flow followed by intervals when blood flow is inadequate, which leads to hypoxia/reoxygenation injury (Hung and Burton, 2006). These presumptive explanations differ in that in the first case the placenta is in a chronic hypoxic state while in the second situation there are repeated intervals of hypoxia followed by reoxygenation. Under both conditions, massive numbers of free radicals would be generated thereby ensuring elevated molecular damage to the uterine trophoblasts and the release of a wide range of factors that are known to be altered in pre-eclampsia that then promote inflammation. A review by Redman and Sargent (2009) describes these inflammatory agents in detail and an extensive discussion of these data is considered beyond the scope of this report.

Considering the proposed involvement of free radicals and oxidative stress as contributors to the placental damage that occurs early in pre-eclampsia, it was anticipated that antioxidants may be beneficial in ameliorating the severity or progression of this disease. When tested, however, the data do not generally support the use of either vitamins C or E as having benefit in the treatment of pre-eclampsia (Conde-Agudelo et al., 2011; Rossi and Mullin, 2011). This is also the case with some other antioxidants, for example co-enzyme Q10, that have been examined (Briceno-Perez et al., 2009).

As noted above, melatonin as an antioxidant has a much greater capacity to scavenge radicals and reduce oxidative damage than do either vitamins C or E, including in the placenta (Martin et al., 2000a; Milczarek et al., 2010). Melatonin and its secondary, tertiary and quaternary metabolites function as radical scavengers and so melatonin neutralizes many more toxic reactants than do the vitamin antioxidants (Tan et al., 1993, 2002; Galano et al., 2011, 2013). Moreover, melatonin acts as an indirect antioxidant by stimulating antioxidative enzymes (Barlow-Walden et al., 1995; Pablos et al., 1998; Rodriguez et al., 2004) and glutathione production (Urata et al, 1999), a tripeptide with significant antioxidative activity.

Since the initial assault that leads to pre-eclampsia may be oxidative damage to the placenta arising from chronic hypoxia (Kaufmann et al., 2003) or repeated hypoxia and reoxygenation (Hung and Burton, 2006) of the placental tissue, Okatani et al. (2001) examined whether melatonin would protect against placental ischemia and reperfusion. To achieve this, the authors occluded the utero-ovarian arteries bilaterally for 20 min on Day 19 of pregnancy in rats, half of which were treated with melatonin (10 mg/kg) 60 min in advance of arterial occlusion. The hypoxic/reoxygenation episode was followed by a reduction in the placental mitochondrial respiratory control index and drop in the ratio of adenosine-5-diphosphate (ADP) concentration to oxygen consumption (ADP/O) during state 3 respiration, while the level of oxidized lipids in the placenta rose. These changes were prevented when melatonin was given documenting that the indole, likely due to its antioxidant activity, is capable of protecting the placenta from ischemia/reperfusion injury. Moreover, the authors surmised that melatonin may be a beneficial treatment in other situations where free radicals contribute to fetal problems, e.g. fetal growth restriction and fetal hypoxia associated with a difficult delivery. Interestingly, subsequent reports have proved this speculation valid; melatonin reduces fetal growth restriction in rats due to either placental ischemia/reperfusion (Nagai et al., 2008) or undernourishment during pregnancy (Richter et al., 2009) and in sheep resulting from a period of ischemia followed by reperfusion (Lemley et al., 2012). In humans as well, melatonin reduced oxidative damage in newborns that experienced a period of asphyxia due to difficult delivery (Fulia et al., 2001).

Lanoix et al. (2012a, 2013) recently completed a series of in vitro studies in which they used melatonin to protect primary human STB cells from apoptosis due to 4-h hypoxia followed by 18 h of reoxygenation. The hypoxia/reoxygenation procedure clearly caused oxidative stress in the STB cells, which led to an activation of the Bax/Bcl-2 mitochondrial apoptosis pathway and DNA fragmentation. In this in vitro model, melatonin markedly reduced both apoptosis and the associated DNA damage. These findings support the possibility that melatonin may be useful in vivo to limit complications of pregnancy that involve damage to STB cells and their premature loss. For example, STB apoptosis is elevated in pre-eclampsia and intrauterine growth restriction (Heazell et al., 2008; Tomas et al., 2011).

In addition to oxidative damage to the placenta, etc., circadian misalignment may be one of many factors that contribute to pre-eclampsia (Ditisheim et al., 2013). Chronodisruption has been tentatively experimentally linked to other reproductive malfunctions including interruption of pregnancy, spontaneous abortion and low birthweight infants (Mahoney, 2010; Summa et al., 2012). The epidemiological data, however, have not uniformly supported an association between pre-eclampsia and shift work, a known circadian disrupter (Wergeland and Strand, 1997; Haelterman et al., 2007; Chang et al., 2010).

Gestational hypertension is one aspect of pre-eclampsia that may be linked to circadian disruption given that these rhythmic perturbations are also often associated with melatonin suppression. Normally, blood pressure in humans varies over a 24-h period with lowest pressure occurring at night (de la Sierra et al., 2009; Reiter et al., 2010b). This nocturnal reduction is mediated by the concurrent rise in circulating melatonin (Obayashi et al., 2013). Moreover, exogenously administered melatonin reduces hypertension in humans (Arangino et al., 1999). Thus, with the elimination of the night-time elevation of melatonin due to shift work, night-time light exposure, etc., some degree of hypertension would be expected, such as that which occurs in pre-eclampsia. Finally, placental diseases including pre-eclampsia may have an impact on the cardiovascular health of the mother later in life (Mosca et al., 2011). The cause of this association presumably relates to the endothelial damage sustained during a difficult pregnancy (Siddiqui and Hladunewich, 2011). Thus, if melatonin would have utility in reducing the severity of pre-eclampsia one downstream effect may also be an improvement of the cardiovascular health of the mother in the long term.

Pre-eclampsia is often accompanied by what is referred to as the HELLP syndrome; this condition includes hemolysis, elevated liver enzymes and low platelet count (Weinstein, 1982). Since it occurs coincident with pre-eclampsia, it is speculated to have a similar causative basis in terms of abnormal placental development or physiology (Abildgaard and Heimdal, 2013). Whether the signs of HELLP have any relation to melatonin has never been investigated. Platelets do, however, normally contain melatonin and receptors for this indole (Vacas et al., 1991; Morera and Abreu, 2005). Also, hemolysis and elevated liver enzymes are frequently a result of oxidative damage to the respective cells (Pacini et al., 2011). This being the case, melatonin, due to its antioxidant properties, may be beneficial in this condition. Finally, melatonin was observed to have anti-epileptic actions in humans (Molino-Carballo et al., 1997; Sanchez-Barcelo et al., 2011). This suggests that melatonin may reduce the likelihood of pre-eclampsia progressing to eclampsia.

Lipopolysaccharide (LPS) from the cell membranes of Gram-negative bacteria is a commonly used toxin in experimental situations to promote free radical-mediated oxidative damage. In a comprehensive molecular study designed to test whether exogenously administered melatonin would combat oxidative destruction at the level of the placenta, Wang et al. (2011) treated mice at 15 days of pregnancy with 300 µg/kg LPS without or with concurrent melatonin (5 mg/kg) treatment. Virtually every placental change induced by LPS, including depressed glutathione, elevated inducible nitric oxide synthase (iNOS), enhanced levels of 3-nitrotyrosine residues, reduced GRP78 expression, elevated elF2α and JNK phosphorylation and increased CHOP expression, was negated in mice treated with melatonin. The results of this thorough study strongly emphasize the potentially important role that endogenously produced placental melatonin could play in preserving homeostasis in this pregnancy-critical organ.

Melatonin not only protects the placenta from LPS-mediated oxidative toxicity but also, likewise, reduces the damage inflicted by LPS on the fetus. When mice were treated (on gravid Days 15–17) with LPS, the toxin increased intrauterine fetal death, caused intrauterine growth retardation (IUGR) and induced biochemical changes consistent with elevated oxidative stress in both the mothers and the fetuses (Chen et al., 2006). As in other experiments of this type, melatonin was effective in eliminating almost all the changes resulting from LPS toxicity, although it did not totally restore the reduced fetal weights of the recovered fetuses.

Other actions of melatonin at the placental level also document that the indole enhances the total antioxidative capacity of this organ. When rats that were undernourished (35% food reduction) from gestation days 15–20 were given melatonin in their drinking water, the relative expression of the antioxidative enzymes manganese superoxide dismutase and catalase were highly significantly elevated over those in the underfed animals not treated with melatonin (Richter et al., 2009). While a similar stimulation of glutathione peroxidase expression was not observed, the findings do show the likely value of melatonin in protecting the placenta from molecular mutilation by free radicals. The enzyme increases were accompanied by an improvement in the growth of the placenta (reflected in the fetal weight to placental weight ratio), which benefitted the fetuses since mean fetal bodyweights were also elevated. Thus in this case, melatonin overcame IUGR which was a result of undernourishment, a procedure believed to involve excessive free radical generation (Franco et al., 2006).

Melatonin in relation to parturition

The processes of labor and delivery of a child involve increasingly frequent and robust contractions of the myometrium accompanied by cervical effacement and expulsion of the fetus. While these events obviously occur at any time during the day or night, there are reports noting that the onset of human term labor more commonly takes place in the late night and early morning hours (Glattre and Bjerkedal, 1983; Cagnacci and Soldani, 1998; Lindow et al., 2000). Similar claims have been made relative to preterm labor (Iams et al., 2002), at least after 28 weeks of gestation (Vatish et al., 2010). During evolution there were likely selective survival advantages for both the mother and the newborn to enter parturition at night since predation pressure was lowest at that time. As in the human, similar 24-h circadian variations of birth are seen in other species (Lincoln and Porter, 1976; Zahn and Hattensperger, 1993; Farber et al., 1997).

The circadian signals that drive the nocturnal changes that culminate in the delivery of offspring in late term human pregnancy are only beginning to be unraveled (Olcese, 2012; Olcese et al., 2013). Moreover, the circadian mechanisms related to night-time parturition are probably breaking down because of the widespread misuse of light at night. Throughout evolution where the regularly recurring light:dark cycles were undisturbed, internal rhythms were more strongly coupled to the photoperiodic environment (Golombek et al., 2013). With the advent of the developments in artificial lighting, the natural environmental light/dark cycles are being markedly subverted (witness, for example, the light environments in a hospital setting) and normally entrained circadian rhythms are likewise under increasingly greater pressure. The dysynchronization of circadian rhythms, also referred to as chronodisruption (Erren and Reiter, 2009; 2013), due to perturbed or unusual light/dark cycles generally negatively influence physiological and metabolic processes and sometimes lead to disease (Blask et al., 2011; Bass, 2012; Münch and Bromundt, 2012; Golombek et al., 2013).

Within the last decade, melatonin, which exhibits a pronounced circadian rhythm in both the pineal gland and the circulation of all mammals (Vaughan et al., 1976; Panke et al., 1979; Reiter, 1986; Stehle et al., 2011), has been shown to modulate uterine physiology. With the aid of receptor autoradiography and radioreceptor assays, Schlabritz-Loutsevitch et al. (2003) identified specific high-affinity, G-protein coupled, melatonin-binding sites on the membranes of uterine myometrial cells obtained from both non-pregnant and pregnant humans. These findings are consistent with the presence of the well-defined 7-transmembrane melatonin receptors typical of many cells (Stankov and Reiter, 1990; Dubocovich and Markowska, 2005; Slominski et al., 2012). Moreover, this same group (Schlabritz-Loutsevitch et al., 2003) reported that the accumulation of cyclic adenosine monophosphate (cAMP) in cultured primary myocytes obtained from the human uterus was not suppressed by melatonin after treating the cells with forskolin. This showed that the common Gi protein coupling of the uterine melatonin receptors to adenylyl cyclase was not essential in these cells (Dubocovich and Markowska, 2005). Interestingly, however, it was found that melatonin activates the same intracellular signaling pathway as does oxytocin in uterine myometrial cells. These intracellular events include stimulation of phospholipase C, protein kinase C and myosin light chain kinase (Sharkey et al., 2009, 2010). This being the case, oxytocin and melatonin may act synergistically to promote and sustain strong uterine contractility which aids in the delivery of the fetus. This interaction between oxytocin and melatonin, which is normally elevated during darkness, could explain the higher night-time delivery of offspring (Lincoln and Porter, 1976; Zahn and Hattensperger, 1993; Farber et al., 1997). That melatonin actually sensitizes the human uterus to oxytocin is supported by the finding that cultured myometrial cells treated with a concentration of oxytocin that by itself did not induce contractions undergo powerful contractions when melatonin is added with the low oxytocin concentration (Sharkey et al., 2009, 2010).

An examination of the potential mechanisms involved in the oxytocin/melatonin synergy showed that melatonin also enhances gap junction activity and mRNA as well as protein expression for connexin 43, a gap junction protein common to many cells, including uterine myocytes. Thus, melatonin seems to assist in the co-ordination of uterine muscle cells during labor to intensify contractile strength (Olcese et al., 2013). Further examination of myometrial cells collected during labor revealed elevated 2-I125-melatonin binding compared with smooth muscle cells harvested from the non-laboring uterus. Since 2-I125-melatonin binds to both the MT1 and MT2 membrane receptors, the obvious implication is that these receptors are more abundant in the uterus during labor than during a more quiescent phase.

Preliminary studies recently performed in humans support a significant role for melatonin in aiding in the delivery of the fetus by enhancing the strength of uterine contractions (Olcese et al., 2013). Pregnant volunteers (>38 weeks gestation) were monitored continuously from 19:00–07:00 h under dim white light as to the frequency and strength of their uterine contractions. When these females were exposed to a 10 000 lux full spectrum light at 23:00 h for 1 h, the strength of their uterine contractions was diminished during the remainder of the night. This light intensity is known to suppress endogenous circulating melatonin levels (Lewy et al., 1980; Revell and Skene, 2007), although in this study melatonin was not measured. The regularity and the strength of the uterine contractions were monitored by an experienced obstetrical nurse but the study was not blinded. The results, however, generally support the idea that the nocturnal rise in melatonin contributes to the increased night-time labor onset and delivery of the offspring due to its synergistic interaction with oxytocin.

The results summarized above are intriguing in view of what is known about the melatonin rhythm during pregnancy. The amplitude of the nocturnal melatonin peak becomes steadily higher in late pregnancy such that, near term, the night-time melatonin levels are higher than those measured at any other time (Fig. 7) (Nakamura et al., 2001; Tamura et al., 2008a) thereby enhancing the likelihood of its synergy with oxytocin. This rise may be an evolutionary adaptation to enhance uterine contractions near the time when delivery should occur. Immediately after delivery, nocturnal melatonin levels in the mother return to values typical of those in non-pregnant women (Tamura et al., 2008a).

Figure 7

Melatonin synergizes with oxytocin from the posterior pituitary (PP) to strengthen the force of uterine myometrial contractions during labor. Since maximal melatonin levels occur at night in darkness, its synergistic actions with oxytocin may explain the elevated frequency of labor onset at night. In developed countries, the widespread use of manufactured light at night alters circadian rhythms and suppresses the nocturnal production of melatonin thereby likely reducing nighttime labor onset and spreading this process throughout the 24-h period. AP, anterior pituitary; PG, pineal gland; PVN, paraventricular nucleus; SON, supraoptic nucleus.

Several conclusions and/or recommendations follow from the data summarized: (i) considering the increasingly widespread use of manufactured light, to the point where darkness in urban areas is disappearing and humans are becoming progressively more melatonin deficient, it seems likely that chronodisruption and the loss of melatonin will remain a continuing problem; (ii) since melatonin seems to synergize with oxytocin to strengthen uterine contractions, when spontaneous labor begins melatonin could be given as a supplement to augment uterine contractions, to reduce the duration of labor and make delivery easier. Parenthetically, melatonin also may be useful during delivery since it has been shown to alleviate pain (Gitto et al., 2012; Srinivasan et al., 2012). (iii) When labor, especially during the day, is induced using oxytocin, it could be given with melatonin to intensify the strength of uterine contractions; (iv) women who have a history of early delivery (e.g. before 32 weeks of gestation) should probably avoid melatonin use during pregnancy and (v) throughout pregnancy, but especially during the last trimester, women should maintain a regular light/dark cycle, without interrupting the period of darkness with even brief periods of light, for the purpose of stabilizing their circadian rhythms and maximizing the nocturnal melatonin rise, both of which influence the maturation of the fetal circadian system.

Conclusion

Both melatonin and circadian rhythms are influential in determining optimal reproductive physiology during pregnancy. The discoveries related to melatonin production in several peripheral reproductive organs suggest that this indole has several important functions in these tissues, one of which very likely relates to its ability to obviate oxidative/nitrosative stress and placental dysfunction. The production of this highly effective antioxidant in the placenta may be of special significance since it would likely protect against conditions that involve less than optimal placentation, a situation that leads to either prolonged or intermittent hypoxia/reoxygenation and elevated free radical generation. Moreover, pre-eclampsia, which many researchers agree involves exaggerated free radical generation that leads to the manifestation of the signs of this condition, also may be ameliorated by this indoleamine. Given that melatonin readily crosses the placenta it also protects the fetus from oxidative/nitrosative stress. Considering the uncommonly low, or lack of, toxicity of melatonin, including during pregnancy (Jahnke et al., 1999), melatonin may prove to be highly valuable in optimizing the physiology of the reproductive system not only during pregnancy but also at other times (Reiter et al., 2013a). Certainly, it is the hope that research in this area will flourish during the next decade.

Circadian rhythms of the mother are seemingly highly important in programming the fetal clock, either via the melatonin signal and/or by other means. This being the case, it is imperative that during pregnancy, and perhaps especially during the last trimester, pending mothers maintain a regular sleep/wake and light:dark cycle. The light:dark cycle is especially important because the photic environment is an important impeller of the maternal master biological clock, the SCN, which in turn directly or indirectly provides circadian information to the fetus. Thus, shift work and unusual exposure to bright light at night should be avoided since they cause chronodisruption and melatonin suppression. There is a large amount of experimental and clinical data showing that disturbances caused by these processes have untoward consequences for the fetus.

Authors' roles

R.J.R. performed literature searches, had critical discussion of the data with each of the co-workers and wrote the preliminary and final drafts of the paper. D.X.T. performed literature searches, analyzed and discussed data and read drafts of the paper and made suggestions for changes/additions. A.K. and S.A.R.-C. prepared figures, discussed data and read all drafts of the paper.

Funding

No funding was obtained from any source to support this work.

Conflict of interest

The authors declare no conflict of interest.

References

Abildgaard
U
Heimdal
K
Pathogenesis of the syndrome of hemolysis, elevated liver enzymes, and low platelet county (HELLP): a review
Eur J Obstet Gynecol Reprod Biol
2013
, vol. 
166
 (pg. 
117
-
123
)
Acuna-Castroviejo
D
Escames
G
Carazo
A
Leon
J
Khaldy
H
Reiter
RJ
Melatonin, mitochondrial homeostasis and mitochondrial-related diseases
Curr Top Med Chem
2002
, vol. 
2
 (pg. 
133
-
151
)
Acuna-Castroviejo
D
Lopez
LC
Escames
G
Lopez
A
Garcia
JA
Reiter
RJ
Melatonin-mitochondria interplay in health and disease
Curr Top Med Chem
2011
, vol. 
11
 (pg. 
221
-
240
)
Allegra
M
Reiter
RJ
Tan
DX
Gentile
C
Tesoriere
L
Livrea
MA
The chemistry of melatonin's interaction with reactive species
J Pineal Res
2003
, vol. 
34
 (pg. 
1
-
10
)
Arangino
S
Cagnacci
A
Angiolucci
M
Vacca
AM
Longu
G
Volpe
A
Melis
GB
Effects of melatonin on vascular reactivity, catecholamine levels, and blood pressure in healthy men
Am J Cardiol
1999
, vol. 
83
 (pg. 
1417
-
1419
)
Axelrod
J
Wurtman
RJ
Winget
CM
Melatonin synthesis in the hen pineal gland and its control by light
Nature
1964
, vol. 
201
 pg. 
1134
 
Axelrod
J
Wurtman
RJ
Snyder
SH
Control of hydroxyindole-O-methyltransferase activity in the rat pineal gland by environmental lighting
J Biol Chem
1965
, vol. 
240
 (pg. 
949
-
954
)
Barlow-Walden
LR
Reiter
RJ
Abe
M
Cablos
M
Menendez-Pelaez
A
Chen
LD
Poeggeler
B
Melatonin stimulates brain glutathione peroxidase activity
Neurochem Int
1995
, vol. 
26
 (pg. 
497
-
502
)
Bass
J
Circadian topology of metabolism
Nature
2012
, vol. 
491
 (pg. 
348
-
356
)
Bass
J
Takahashi
JS
Circadian integration of metabolism and energetics
Science
2010
, vol. 
37
 (pg. 
1349
-
1354
)
Bauer
J
Janecke
A
Gerss
J
Masjosthusmann
K
Werner
C
Hoffmann
G
Circadian variation in oxygen consumption in preterm infants
J Perinat Med
2009
, vol. 
37
 (pg. 
413
-
417
)
Behrman
HR
Kodaman
PH
Preston
SL
Gao
S
Oxidative stress and the ovary
J Soc Gynecol Investig
2001
, vol. 
8
 (pg. 
S40
-
S42
)
Bellavia
SL
Carpentieri
AR
Vaque
AM
Macchione
AF
Vermouth
NT
Pup circadian rhythm entrainment-effect of maternal ganglionectomy or pinealectomy
Physiol Behav
2006
, vol. 
89
 (pg. 
342
-
349
)
Blask
DE
Hill
SM
Dauchy
RT
Xiang
S
Yuan
L
Duplessis
T
Mao
L
Dauchy
E
Saver
LA
Circadian regulation of molecular, dietary and metabolic signaling mechanisms of human breast cancer growth by the nocturnal melatonin signal and the consequences of its disruption by light at night
J Pineal Res
2011
, vol. 
51
 (pg. 
259
-
269
)
Brainard
GC
Richardson
BA
King
TS
Matthews
SA
Reiter
RJ
The suppression of pineal melatonin content and N-acetyltransferase activity by different light irradiances in the syrian hamster: a dose–response relationship
Endocrinology
1983
, vol. 
113
 (pg. 
293
-
298
)
Brainard
GC
Richardson
BA
King
TS
Reiter
RJ
Influence of different light spectra on the suppression of pineal melatonin content in the Syrian hamster
Brain Res
1984
, vol. 
294
 (pg. 
333
-
339
)
Brannstrom
M
Norman
RJ
Involvement of leukocytes and cytokines in the ovulatory process and corpus luteum function
Hum Reprod
1993
, vol. 
8
 (pg. 
1762
-
1775
)
Briceno-Perez
C
Briceno-Sanabria
L
Vigil-De Garcia
P
Prediction and preeclampsia
Hyperten Pregnancy
2009
, vol. 
28
 (pg. 
138
-
155
)
Brzezinski
A
Seibel
MM
Lynch
HJ
Deng
MH
Wurtman
RJ
Melatonin in human preovulatory follicular fluid
J Clin Endocrinol Metab
1987
, vol. 
64
 (pg. 
864
-
867
)
Cagnacci
A
Soldani
R
Melis
GB
Volpe
A
Diurnal rhythms of labor and delivery in women: modulation by parity and reasons
Am J Obst Gynecol
1998
, vol. 
178
 
1 Pt 1
(pg. 
140
-
145
)
Cardinali
DP
Srinivasan
V
Brzezinski
A
Brown
GM
Melatonin and its analogs in insomnia and depression
J Pineal Res
2012
, vol. 
52
 (pg. 
365
-
375
)
Carrillo-Vico
A
Guerrero
JM
Lardone
PJ
Reiter
RJ
A review of the multiple actions of melatonin on the immune system
Endocrine
2005
, vol. 
27
 (pg. 
189
-
200
)
Carter
DS
Goldman
BD
Antigonadal effect of timed melatonin infusion in pinealectomized male Djungarian hamsters (Phodopus sungorus sungorus): duration is the critical parameter
Endocrinology
1983
, vol. 
113
 (pg. 
1261
-
1267
)
Chang
PJ
Chu
LC
Hsieh
WS
Chuang
YL
Lin
SJ
Chen
PC
Working hours and risk of gestational hypertension and pre-eclampsia
Occup Med
2010
, vol. 
60
 (pg. 
66
-
71
)
Chaste
P
Clement
N
Botros
HG
Guillaume
JL
Konyukh
M
Pagan
C
Scheid
I
Nygren
G
Anckarsater
H
Rastam
M
, et al. 
Genetic variations of the melatonin pathway in patients with attention-deficit and hyperactivity disorders
J Pineal Res
2011
, vol. 
51
 (pg. 
394
-
399
)
Chen
YH
Xu
DX
Wang
JP
Wang
H
Wei
LZ
Sun
MF
Wei
W
Melatonin protects against lipopolysaccharide-induced intrauterine fetal death and growth retardation in mice
J Pineal Res
2006
, vol. 
40
 (pg. 
40
-
47
)
Choi
J
Park
SM
Lee
E
Kim
JH
Jeong
YI
Lee
JY
Park
SW
Kim
HS
Hossein
MS
Jeong
YW
, et al. 
Anti-apoptotic effect of melatonin on preimplantation development of porcine parthenogenetic embryos
Mol Reprod Dev
2008
, vol. 
75
 (pg. 
1127
-
1135
)
Conde-Agudelo
A
Romero
R
Kusanovic
JP
Hassan
SS
Supplementation with vitamins C and E during pregnancy for the prevention of preeclampsia and other adverse maternal and perinatal outcomes: a systematic review and meta analysis
Am J Obstet Gynecol
2011
, vol. 
204
 (pg. 
503.e1
-
503.e12
)
de la Sierra
A
Redon
J
Banegas
JR
Segura
J
Parati
G
Gorostidi
M
de la Cruz
JJ
Sobrino
J
Llisterri
JL
Alonso
J
, et al. 
Prevalence and factors associated with circadian blood pressure patterns in hypertensive patients
Hypertension
2009
, vol. 
53
 (pg. 
466
-
472
)
de Matos Cavalcante
AG
Carvalhedo de Bruin
PF
Sales de Bruin
VM
Nunes
DM
Pereira
ED
Cavalcante
MM
Andrade
GM
Melatonin reduces lung oxidative stress in patients with chronic obstructive pulmonary disease: a randomized, double-blind, placebo-controlled study
J Pineal Res
2012
, vol. 
53
 (pg. 
238
-
244
)
Ditisheim
AJ
Dibner
C
Phillippe
J
Pechere-Bertschi
A
Biological rhythms and preeclampsia
Front Endocrinol
2013
Dubocovich
ML
Markowska
M
Functional MT1 and MT2 melatonin receptors in mammals
Endocrine
2005
, vol. 
27
 (pg. 
101
-
110
)
Dumont
M
Lanctot
V
Cadieux-Viau
R
Paquet
J
Melatonin production and light exposure of rotating night workers
Chronobiol Int
2012
, vol. 
29
 (pg. 
203
-
210
)
El-Raey
M
Geshi
M
Somfai
T
Kaneda
M
Hirako
M
Abdel-Ghaffar
AE
Sosa
GA
El-Roos
ME
Nagai
T
Evidence of melatonin synthesis in the cumulus oocyte complexes and it role in embracing oocyte maturation in vitro in cattle
Mol Reprod Dev
2011
, vol. 
78
 (pg. 
250
-
262
)
Erren
TC
Reiter
RJ
Defining chronodisruption
J Pineal Res
2009
, vol. 
46
 (pg. 
245
-
247
)
Erren
TC
Reiter
RJ
Revisiting chronodisruption: when the physiological nexus between internal and external time splits in humans
Naturwissenschaften
2013
, vol. 
100
 (pg. 
291
-
298
)
Farber
DM
Giussani
DA
Jenkins
SC
Mecenas
CA
Winter
SA
Wentworth
RA
Nathanielsz
PW
Timing of the switch from myometrial contractures to contractions in late-gestation pregnant Rhesus monkeys as recorded by myometrial electromyogram during spontaneous term and androstenedione-induced labor
Biol Reprod
1997
, vol. 
56
 (pg. 
557
-
562
)
Fenn
AM
Fonken
LK
Nelson
RJ
Sustained melatonin treatment blocks body mass, pelage, reproductive and fever responses to short day lengths in female Siberian hamsters
J Pineal Res
2011
, vol. 
51
 (pg. 
180
-
186
)
Ferreira
DS
Amaral
FG
Mesquita
CC
Barbosa
AP
Lellis-Santos
C
Turati
AO
Santos
LR
Sollon
CS
Gomes
PR
Faria
JA
Maternal melatonin programs the daily pattern of energy metabolism in adult offspring
PLoS One
2012
, vol. 
7
 pg. 
e38795
 
Franco
MC
Akamine
EH
Reboucas
N
Carvalho
MH
Tostes
RC
Nigro
D
Fortes
ZB
Long-term effects of intrauterine malnutrition on vascular function in female offspring: implications of oxidative stress
Life Sci
2006
, vol. 
80
 (pg. 
709
-
715
)
Fulia
F
Gitto
E
Cuzzocrea
S
Reiter
RJ
Dugo
L
Gittoi
P
Barberi
S
Cordaro
S
Barberi
I
Increased levels of malondialdehyde and nitrite/nitrate in the blood of asphyxiated newborns: reduction by melatonin
J Pineal Res
2001
, vol. 
31
 (pg. 
343
-
349
)
Gaber
LW
Spargo
BH
Lindheimer
MD
Renal pathology in pre-eclampsia
Baillieres Clin Obstet Gynoecol
1994
, vol. 
8
 (pg. 
443
-
468
)
Galano
A
Tan
DX
Reiter
RJ
Melatonin as a natural ally against oxidative stress: a physiochemical examination
J Pineal Res
2011
, vol. 
51
 (pg. 
1
-
16
)
Galano
A
Tan
DX
Reiter
RJ
On the free radical scavenging activities of melatonin's metabolites, AFMK and AMK
J Pineal Res
2013
, vol. 
54
 (pg. 
245
-
257
)
Gillette
MU
Tischkau
SA
Suprachiasmatic nucleus: the brain's circadian clock
Recent Prog Horm Res
1999
, vol. 
54
 (pg. 
33
-
58
)
Gitto
E
Aversa
S
Salpietro
CD
Barberi
I
Arrigo
T
Trimarchi
G
Reiter
RJ
Pelligrino
S
Pain in neonatal intensive care: role of melatonin as an analgesic antioxidant
J Pineal Res
2012
, vol. 
52
 (pg. 
291
-
295
)
Glattre
E
Bjerkedal
T
The 24-hour rhythmicity of birth: a population study
Acta Obstet Gynecol Scand
1983
, vol. 
62
 (pg. 
31
-
36
)
Golombek
DA
Casiraghi
LP
Agostino
PV
Paladino
N
Duhart
JM
Plano
SA
Chiesa
JJ
The times they're a-changing: effects of circadian desynchronization on physiology and disease
J Physiol Paris
2013
, vol. 
107
 (pg. 
310
-
322
)
Haelterman
E
Marcoux
S
Croteau
A
Dramaix
M
Population-based study on occupational risk factors for preeclampsia and gestational hypertension
J Work Environ Health
2007
, vol. 
33
 (pg. 
304
-
317
)
Hardeland
R
Melatonin in aging and disease—multiple consequences of reduced secretion, options and limits of treatment
Aging Dis
2012
, vol. 
3
 (pg. 
194
-
225
)
Hardeland
R
Tan
DX
Reiter
RJ
Kynuramines, metabolites of melatonin and other indoles: the resurrection of an almost forgotten class of biogenic amines
J Pineal Res
2009
, vol. 
47
 (pg. 
109
-
126
)
Hardeland
R
Madrid
JA
Tan
DX
Reiter
RJ
Melatonin, the circadian multioscillator system and health: the need for detailed analyses of peripheral melatonin signaling
J Pineal Res
2012
, vol. 
52
 (pg. 
139
-
166
)
Heazell
AE
Buttle
HR
Baker
PN
Crocker
IP
Altered expression of regulators of caspase activity within trophoblast of normal pregnancies and pregnancies complicated by preeclampsia
Reprod Sci
2008
, vol. 
15
 (pg. 
1034
-
1043
)
Hofman
MA
The human circadian clock and aging
Chronobiol Int
2000
, vol. 
17
 (pg. 
245
-
259
)
Hoffman
RA
Reiter
RJ
Pineal gland: influence on gonads of male hamsters
Science
1965
, vol. 
148
 (pg. 
1609
-
1611
)
Hoffman
RA
Reiter
RJ
Influence of compensatory mechanisms and the pineal gland on dark-induced gonadal atrophy in male hamsters
Nature
1966
, vol. 
207
 (pg. 
656
-
657
)
Hung
TH
Burton
G
Hypoxia and reoxygenation: a possible mechanism for placental oxidative stress in pre-eclampsia
Taiwan J Obstet Gynec
2006
, vol. 
45
 (pg. 
189
-
200
)
Huppertz
B
Kingdom
JCP
Apoptosis in the trophoblast—role of apoptosis in placental morphogenesis
J Soc Gynecol Investig
2004
, vol. 
11
 (pg. 
353
-
362
)
Iams
JD
Newman
RB
Thom
EA
Goldenberg
RL
Mueller-Heubach
E
Maowad
A
Sibai
BM
Caritis
SN
Miodovnik
M
Paul
RH
, et al. 
Frequency of uterine contractions and the risk of spontaneous preterm delivery
N Engl J Med
2002
, vol. 
346
 (pg. 
250
-
255
)
Illnerova
H
Buresova
M
Presl
J
Melatonin rhythm in human milk
J Clin Endocrinol Metab
1993
, vol. 
77
 (pg. 
838
-
841
)
Itoh
MT
Ishizuka
B
Kuribayashi
Y
Amemiya
A
Sumi
Y
Melatonin, its precursors, and synthesizing enzyme activities in the human ovary
Mol Human Reprod
1999
, vol. 
5
 (pg. 
402
-
408
)
Jahnke
G
Marr
M
Myers
C
Wilson
R
Travlos
G
Price
C
Maternal and developmental toxicity evaluation of melatonin administered orally to pregnant Sprague–Dawley rats
Toxicol Sci
1999
, vol. 
50
 (pg. 
271
-
279
)
Jud
C
Albrecht
U
Circadian rhythms in murine pups develop in the absence of a functional maternal circadian clock
J Biol Rhythms
2006
, vol. 
21
 (pg. 
149
-
154
)
Kaufmann
P
Black
S
Huppertz
B
Endovascular trophoblast invasion: implications for the pathogenesis of intrauterine growth retardation and preeclampsia
Biol Reprod
2003
, vol. 
69
 (pg. 
1
-
7
)
Kennaway
DJ
Stamp
GE
Goble
FC
Development of melatonin production in infants and impact of prematurity
J Clin Endocrinol Metab
1992
, vol. 
75
 (pg. 
397
-
399
)
Kennaway
DJ
Goble
FC
Stamp
GE
Factors influencing the development of melatonin rhythmicity in humans
J Clin Endocrinol Metab
1996
, vol. 
81
 (pg. 
1525
-
1532
)
Lanoix
D
Ouellette
R
Vaillancourt
C
Expression of melatoninergic receptors in human placenta choriocarcinoma cell lines
Hum Reprod
2006
, vol. 
21
 (pg. 
1981
-
1988
)
Lanoix
D
Beghdadi
H
Lafond
J
Vaillancourt
C
Human placental trophoblasts synthesize melatonin and express its receptors
J Pineal Res
2008
, vol. 
45
 (pg. 
50
-
60
)
Lanoix
D
Lacasse
AA
Reiter
RJ
Vaillancourt
C
Melatonin: the smart killer: the human trophoblast as a model
Mol Cell Endrocrinol
2012a
, vol. 
348
 (pg. 
1
-
11
)
Lanoix
D
Guerin
P
Vaillancourt
C
Placental melatonin production and melatonin receptor expression are altered in preeclampsia: new insights into the role of this hormone in pregnancy
J Pineal Res
2012b
, vol. 
54
 (pg. 
417
-
425
)
Lanoix
D
Lacasse
AA
Reiter
RJ
Vaillancourt
C
Melatonin: the watchdog of villous trophoblast homeostasis against hypoxia/reoxygenation-induced oxidative stress and apoptosis
Mol Cell Endocrinol
2013
, vol. 
38
 (pg. 
35
-
45
)
Lemley
CO
Meyer
AM
Camacho
LE
Neville
TL
Newman
DJ
Caton
JS
Vonnahme
KA
Melatonin supplementation alters uteroplacental hemodynamic and fetal development in an ovine model of intrauterine growth restriction
Am J Physiol Regul Integr Comp Physiol
2012
, vol. 
302
 (pg. 
R454
-
R467
)
Lerner
AB
Case
JD
Takahashi
Y
Mori
W
Isolation of melatonin, the pineal gland factor that lightens melanocytes
J Am Chem Soc
1958
, vol. 
80
 pg. 
2587
 
Lewis
MP
Sullivan
MH
Elder
MG
Regulation of interlookin-1 beta of growth and collagenase production by choriocarcinoma cells
Placenta
1994
, vol. 
15
 (pg. 
13
-
20
)
Lewy
AJ
Wehr
TA
Goodwin
FK
Newsome
DA
Markey
SP
Light suppresses melatonin secretion in humans
Science
1980
, vol. 
210
 (pg. 
1267
-
1269
)
Li
MD
Li
CM
Wang
Z
The role of circadian clocks in metabolic disease
Yale J Biol Med
2012
, vol. 
85
 (pg. 
387
-
401
)
Lincoln
GA
Hazlerigg
DG
Mammalian circannual pacemakers
Soc Reprod Fertil
2010
, vol. 
67
 
Suppl.
(pg. 
171
-
186
)
Lincoln
DW
Porter
DG
Timing of the photoperiod and the hour of delivery in rats
Nature
1976
, vol. 
260
 (pg. 
780
-
781
)
Lindow
SW
Jha
RR
Thompson
JW
24-hour rhythm to the onset of preterm labor
Br J Obstet Gynecol
2000
, vol. 
107
 (pg. 
1145
-
1148
)
Lowes
DA
Webster
NR
Murphy
MP
Galley
HF
Antioxidants that protect mitochondria reduce interleukin-6 and oxidative stress, improve mitochondrial function, and reduce biochemical markers of organ dysfunction in a rat model of acute sepsis
Br J Anaesth
2013
, vol. 
110
 (pg. 
472
-
480
)
Lucas
RJ
Mammalian inner retinal photoreception
Curr Biol
2013
, vol. 
23
 (pg. 
R125
-
R133
)
Lydic
R
Schoene
WC
Czeisler
CA
Moore-Ede
MC
Suprachiasmatic region of the human hypothalamus: homolog to the primate circadian pacemaker?
Sleep
1980
, vol. 
2
 (pg. 
355
-
361
)
Mahoney
MM
Shift work, jet lag, and female reproduction
Int J Endocrinol
2010
 
813764
Maldonado
MD
Murrillo-Cabezas
F
Terron
MP
Flores
LJ
Tan
DX
Manchester
LC
Reiter
RJ
The potential of melatonin in reducing morbidity-mortality after craniocerebral trauma
J Pineal Res
2007
, vol. 
42
 (pg. 
1
-
11
)
Martin
M
Macias
M
Escames
G
Leon
J
Acuna-Castroviejo
D
Melatonin but not vitamins C and E maintains glutathione homeostasis in t-butyl hydroperoxide-induced mitochondrial oxidative stress
FASEB J
2000a
, vol. 
14
 (pg. 
1677
-
1679
)
Martin
M
Macias
M
Escames
G
Reiter
RJ
Agapito
MT
Ortiz
GG
Acuna-Castroviejo
D
Melatonin-induced increased activity of the respiratory chain complexes I and IV can prevent mitochondrial damage induced by ruthenium red in vivo
J Pineal Res
2000b
, vol. 
28
 (pg. 
242
-
248
)
Mason
CA
Lincoln
DW
Visualization of the retino-hypothalamic projection in the rat by cobalt precipitation
Cell Tiss Res
1976
, vol. 
168
 (pg. 
117
-
131
)
Mauriz
JL
Collado
PS
Veneroso
C
Reiter
RJ
Gonzalez-Gallego
J
A review of the molecular aspects of melatonin's anti-inflammatory actions: recent insights and new perspectives
J Pineal Res
2013
, vol. 
54
 (pg. 
1
-
14
)
Menaker
M
Murphy
ZC
Sellix
MT
Central control of peripheral oscillators
Curr Opin Neurobiol
2013
, vol. 
23
 (pg. 
741
-
746
)
Menendez-Pelaez
A
Reiter
RJ
Distribution of melatonin in mammalian tissue: the relative importance of nuclear verses cytosolic localization
J Pineal Res
1993
, vol. 
15
 (pg. 
59
-
69
)
Meng
Y
He
Z
Yin
J
Zhang
Y
Zhang
T
Quantitative calculation of human melatonin suppression induced by inappropriate light at night
Med Biol Eng Comput
2011
, vol. 
49
 (pg. 
1083
-
1088
)
Milczarek
R
Hallmann
A
Sokolowska
E
Kaletha
K
Klimek
J
Melatonin enhances antioxidant action of alpha-tocopherol and ascorbate against NADPH- and iron-dependent lipid peroxidation in human placental mitochondria
J Pineal Res
2010
, vol. 
49
 (pg. 
149
-
155
)
Mogler
RKH
Das Endokrine System des Syrischen Goldhamsters unter natürlichen Winterschlaf
Z Morphol Oekal Tiere
1958
, vol. 
47
 (pg. 
267
-
308
)
Molino-Carballo
A
Munoz-Hoyos
A
Reiter
RJ
Sanchez-Forte
M
Moreno-Madrid
F
Rufo-Campos
M
Molino-Font
JA
Acuna-Castroviejo
D
Utility of high doses of melatonin as adjunctive anticonvulsant therapy in a child with severe myoclonic epilepsy: two years’ experience
J Pineal Res
1997
, vol. 
23
 (pg. 
97
-
105
)
Morera
AL
Abreu
P
Existence of melatonin in human platelets
J Pineal Res
2005
, vol. 
39
 (pg. 
432
-
433
)
Mosca
L
Benjamin
EJ
Berra
K
Bezanson
JL
Dolor
RJ
Lloyd-Jones
DM
Newby
LK
Pina
IL
Roger
VL
Shawl
J
, et al. 
Effectiveness-based guidelines for the prevention of cardiovascular disease in women—2011 update: a guideline from the American Heart Association
J Am Coll Cardiol
2011
, vol. 
57
 (pg. 
1404
-
1423
)
Münch
M
Bromundt
V
Light and chronobiology: implications for health and disease
Dialogues Clin Neurosci
2012
, vol. 
14
 (pg. 
448
-
453
)
Myatt
L
Rosenfield
RB
Eis
AL
Brockman
DE
Greer
I
Lyall
F
Nitrotyrosine residues in placenta. Evidence of peroxynitrite formation and action
Hypertension
1996
, vol. 
28
 (pg. 
488
-
493
)
Nagai
R
Watanabe
K
Wakatsuki
A
Hamada
F
Shinohara
K
Hayashi
Y
Imamura
R
Fukaya
T
Melatonin preserves fetal growth in rats by protecting against ischemia/reperfusion-induced oxidative/nitrosative mitochondrial damage in the placenta
J Pineal Res
2008
, vol. 
45
 (pg. 
271
-
276
)
Nakamura
Y
Tamura
H
Kashida
S
Takayama
H
Yamagata
Y
Karube
A
Sugino
N
Kato
H
Changes of serum melatonin level and its relationship to feto-placental unit
J Pineal Res
2001
, vol. 
30
 (pg. 
29
-
33
)
Nakamura
Y
Tamura
H
Takayama
H
Kato
H
Increased endogenous level of melatonin in preovulatory human follicles does not directly influence progesterone production
Fertil Steril
2003
, vol. 
80
 (pg. 
1012
-
1016
)
Nelissen
EC
van Montfoort
AP
Dumoulin
JC
Evers
JL
Epigenetics and the placenta
Hum Reprod Update
2011
, vol. 
17
 (pg. 
397
-
417
)
Novakova
M
Sladek
M
Sumova
A
Exposure of pregnant rats to restricted feeding schedule synchronizes the SCN clocks of their fetuses under constant light but not under a light:dark regime
J Biol Rhythms
2010
, vol. 
25
 (pg. 
350
-
360
)
Obayashi
K
Saeki
K
Iwamoto
J
Okamoto
N
Tomioka
K
Nezu
S
Ikada
Y
Kurumatani
N
Nocturnal urinary melatonin excretion is associated with non-dipper pattern in elderly hypertensives
Hyperten Res
2013
, vol. 
36
 (pg. 
736
-
740
)
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
1999
, vol. 
25
 (pg. 
129
-
134
)
Okatani
Y
Wakatsuki
A
Schinohara
K
Taniguchi
K
Fukaya
T
Melatonin protects against oxidative mitochondrial damage induced by rat placenta by ischemia and reperfusion
J Pineal Res
2001
, vol. 
31
 (pg. 
173
-
178
)
Olcese
J
Circadian aspects of mammalian parturition: a review
Mol Cell Endocrinol
2012
, vol. 
349
 (pg. 
62
-
67
)
Olcese
J
Lazier
S
Paradise
C
Melatonin and the circadian timing of human parturition
Reprod Sci
2013
, vol. 
20
 (pg. 
168
-
174
)
Ortiz
A
Espino
J
Bejarano
I
Lozano
GM
Monllor
F
Garcia
JF
Pariente
JA
Rodriguez
A
High endogenous melatonin concentrations enhance sperm quality and short-term in vitro exposure to melatonin improves aspects of sperm motility
J Pineal Res
2011
, vol. 
50
 (pg. 
132
-
139
)
Pablos
MI
Reiter
RJ
Ortiz
GG
Guerro
JM
Agapito
MT
Chuang
JI
Sewerynek
E
Rhythms of glutathione peroxidase and glutathione reductase in brain of chick and their inhibition by light
Neurochem Int
1998
, vol. 
32
 (pg. 
69
-
75
)
Pacini
N
Ferrari
A
Menozzi
M
Borziani
F
Melatonin enhances the in vitro action of cytochalasin B on globular resistance and osmotic fragility of erythrocytes
Neuro Endocrinol Lett
2011
, vol. 
32
 (pg. 
292
-
300
)
Panke
E
Rollag
MD
Reiter
RJ
Pineal melatonin concentrations in the Syrian hamster
Endocrinology
1979
, vol. 
104
 (pg. 
194
-
197
)
Pevet
P
Challet
E
Melatonin: both master clock output and internal time-giver in the circadian clocks network
J Physiol (Paris)
2011
, vol. 
105
 (pg. 
170
-
182
)
Pringle
KG
Kind
KL
Sferruzzi-Perri
AN
Thompson
JG
Roberts
CT
Beyond oxygen: complex regulation and activity of hypoxia inducible factors in pregnancy
Human Reprod Update
2010
, vol. 
16
 (pg. 
415
-
431
)
Proietti
S
Cucina
A
Reiter
RJ
Bizzarri
M
Molecular mechanisms of melatonin's inhibitory actions on breast cancer
Cell Mol Life Sci
2013
, vol. 
70
 (pg. 
2139
-
2157
)
Quay
WB
Volumetric and cytologic variation in the pineal body of Peromyscus leucopus (Rodentia) with respect to sex, captivity and daylength
J Morphol
1956
, vol. 
98
 (pg. 
471
-
495
)
Quay
WB
Circadian rhythm in rat pineal serotonin and its modification by estrous cycle and photoperiod
Gen Comp Endocrinol
1963
, vol. 
3
 (pg. 
473
-
479
)
Redman
CWG
Pre-eclampsia: a multi-stress disorder
Rev Med Int
2011
, vol. 
32
 
Suppl. 1
(pg. 
S41
-
S44
)
Redman
CWG
Sargent
IL
Placental stress and pre-eclampsia: a revised review
Placenta
2009
, vol. 
30
 
Suppl. A
(pg. 
S38
-
S42
)
Reiter
RJ
Pineal control of a seasonal reproductive rhythm in male golden hamsters exposed to natural daylight and temperature
Endocrinology
1973
, vol. 
92
 (pg. 
423
-
430
)
Reiter
RJ
Circannual reproductive rhythms in mammals related to photoperiod and pineal function
Chronobiologia
1974
, vol. 
1
 (pg. 
365
-
395
)
Reiter
RJ
Normal patterns of melatonin levels in the pineal gland and body fluids of humans and experimental animals
J Neural Transm
1986
, vol. 
35
 
Suppl. 21
(pg. 
35
-
54
)
Reiter
RJ
Pineal melatonin: cell biology of its synthesis and of its physiological interactions
Endocrine Rev
1991
, vol. 
12
 (pg. 
151
-
180
)
Reiter
RJ
Hester
RJ
Interrelationships of the pineal gland, the superior cervical ganglia and the photoperiod in the regulation of the endocrine system of hamsters
Endocrinology
1966
, vol. 
79
 (pg. 
1168
-
1170
)
Reiter
RJ
Tan
DX
What constitutes a physiological concentration of melatonin?
J Pineal Res
2003
, vol. 
34
 (pg. 
79
-
80
)
Reiter
RJ
Blask
DE
Johnson
LY
Rudeen
PK
Vaughan
MK
Waring
PJ
Melatonin inhibition of reproduction in the male hamsters: its dependency on time of day of administration and on an intact and sympathetically innervated pineal gland
Neuroendocrinology
1976
, vol. 
22
 (pg. 
107
-
116
)
Reiter
RJ
Paredes
SD
Manchester
LC
Tan
DX
Reducing oxidative/nitrosative stress: a newly-discovered genre for melatonin
Crit Rev Biochem Mol Biol
2009a
, vol. 
44
 (pg. 
175
-
200
)
Reiter
RJ
Tan
DX
Erren
TC
Fuentes-Broto
L
Paredes
SD
Light-mediated perturbations of circadian timing and cancer risk: a mechanistic analysis
Integr Cancer Ther
2009b
, vol. 
8
 (pg. 
354
-
360
)
Reiter
RJ
Tan
DX
Manchester
LC
Paredes
SD
Mayo
JC
Sainz
RM
Melatonin and reproduction revisited
Biol Reprod
2009c
, vol. 
81
 (pg. 
445
-
456
)
Reiter
RJ
Tan
DX
Fuentes-Broto
L
Melatonin: a multitasking molecule
Prog Brain Rec
2010a
, vol. 
181
 (pg. 
127
-
151
)
Reiter
RJ
Tan
DX
Paredes
SD
Fuentes-Broto
L
Beneficial effects of melatonin in cardiovascular disease
Ann Med
2010b
, vol. 
42
 (pg. 
276
-
285
)
Reiter
RJ
Tan
DX
Korkmaz
A
Ma
S
Obesity and metabolic syndrome: association with chronodisruption, sleep deprivation and melatonin suppression
Ann Med
2012a
, vol. 
44
 (pg. 
564
-
577
)
Reiter
RJ
Tan
DX
Madrid
JA
Erren
TC
When the circadian clock becomes a ticking time bomb
Chronobiol Int
2012b
, vol. 
29
 (pg. 
1286
-
1287
)
Reiter
RJ
Rosales-Corral
SA
Manchester
LC
Tan
DX
Peripheral reproductive organ health and melatonin: ready for prime time
Int J Mol Sci
2013a
, vol. 
14
 (pg. 
7231
-
7272
)
Reiter
RJ
Tan
DX
Rosales-Corral
SA
Manchester
LC
The universal nature, unequal distribution and antioxidant functions of melatonin and its derivatives
Mini-Rev Med Chem
2013b
, vol. 
13
 (pg. 
373
-
384
)
Reppert
SM
Schwartz
WJ
Maternal coordination of the fetal biological clock in utero
Science
1983
, vol. 
220
 (pg. 
969
-
971
)
Revel
FG
Masson-Pevet
M
Pevet
P
Mikkelsen
JD
Simonneaux
V
Melatonin controls seasonal breeding by a network of hypothalamic targets
Neuroendocrinology
2009
, vol. 
90
 (pg. 
1
-
4
)
Revell
VL
Skene
DJ
Light-induced melatonin suppression in humans with polychromatic and monochromatic light
Chronobiol Int
2007
, vol. 
24
 (pg. 
1125
-
1137
)
Richards
J
Gumz
M
Advances in understanding the peripheral circadian clocks
FASEB J
2012
, vol. 
26
 (pg. 
3602
-
3613
)
Richter
HG
Hansell
JA
Raut
S
Giussani
DA
Melatonin improves placenta efficiency and birth weight and increases the placental expression of antioxidant enzymes in undernourished rats
J Pineal Res
2009
, vol. 
46
 (pg. 
357
-
364
)
Roberts
JM
Hubel
CA
The two stage model of preeclampsia: variations on the theme
Placenta
2009
, vol. 
23
 
Suppl. A
(pg. 
S43
-
S48
)
Roberts
JM
Taylor
RN
Musci
TJ
Rodgers
GM
Hubel
CA
McLaughlin
MK
Preeclampsia: an endothelial cell disorder
Am J Obst Gynecol
1989
, vol. 
161
 (pg. 
1200
-
1204
)
Rodriguez
C
Mayo
JC
Sainz
RM
Antolini
I
Herrera
F
Martin
V
Reiter
RJ
Regulation of antioxidant enzymes: a significant role for melatonin
J Pineal Res
2004
, vol. 
36
 (pg. 
1
-
9
)
Rodriguez
C
Martin
V
Herrera
F
Garcia-Santos
G
Rodriguez-Blanco
J
Casado-Zapico
S
Sanchez-Sanchez
AM
Suarez
S
Puente-Moncada
N
Anitua
MJ
, et al. 
Mechanisms involved in the pro-apoptotic effect of melatonin in cancer cells
Int J Mol Sci
2013
, vol. 
14
 (pg. 
6597
-
6613
)
Rossi
AC
Mullin
PM
Prevention of pre-eclampsia with low dose aspirin or vitamins C and E in women at high and low risk: a systemic review with meta-analysis
Eur J Obstet Gynecol Reprod Biol
2011
, vol. 
158
 (pg. 
9
-
16
)
Röunberg
I
Kauppila
A
Leppaluoto
J
Martikainen
H
Vakkuri
O
Circadian and seasonal variation in human preovulatory follicular fluid melatonin concentration
J Clin Endocrinol Metab
1990
, vol. 
71
 (pg. 
492
-
496
)
Ruger
M
St Hilaire
MA
Brainard
GC
Khalsa
SB
Kronauer
RE
Czeisler
CA
Lockley
SW
Human phase response curve to a single 6.5 h pulse of short-wavelength light
J Physiol
2013
, vol. 
591
 (pg. 
353
-
363
)
Sainz
RM
Mayo
JC
Rodriguez
C
Tan
DX
Lopez-Burillo
S
Reiter
RJ
Melatonin and cell death: differential actions on apoptosis in normal and cancer cells
Cell Mol Life Sci
2003
, vol. 
60
 (pg. 
1407
-
1426
)
Sakaguchi
K
Itoh
MT
Takahashi
N
Tarumi
W
Ishizuka
B
The rat oocyte synthesizes melatonin
Reprod Fertil Rev
2013
, vol. 
25
 (pg. 
674
-
682
)
Sanchez-Barcelo
EJ
Mediavilla
MD
Reiter
RJ
Clinical uses of melatonin in pediatrics
Int J Pediatr
2011
 
892624
Sand
A
Schmidt
TM
Kofuji
P
Diverse types of ganglion cell photoreceptors in the mammalian retina
Prog Retin Eye Res
2011
, vol. 
51
 (pg. 
17
-
43
)
Santhi
N
Thorne
HC
van der Veen
DR
Johnsen
S
Mills
SL
Hommes
V
Schlangen
LJ
Archer
SN
Dijk
DJ
The spectral composition of evening light and individual differences in the suppression of melatonin and delay of sleep in humans
J Pineal Res
2012
, vol. 
53
 (pg. 
47
-
59
)
Schenker
S
Yang
Y
Perez
A
Acuff
RV
Papas
AM
Henderson
G
Lee
MP
Antioxidant transport by the human placenta
Clin Nutr
1998
, vol. 
17
 (pg. 
159
-
167
)
Schlabritz-Loutsevitch
N
Hellner
N
Middendorf
R
Müller
D
Olcese
J
The human myometrium as a target for melatonin
J Clin Endocrinol Metab
2003
, vol. 
88
 (pg. 
908
-
913
)
Seron-Ferre
M
Riffo
R
Valenzuela
GJ
Germain
AM
Twenty-four-hour pattern of cortisol in the human fetus at term
Am J Obst Gynecol
2001
, vol. 
184
 (pg. 
1278
-
1283
)
Seron-Ferre
M
Valenzuela
GJ
Torres-Farfan
C
Circadian clocks during embryonic and fetal development
Birth Defects Res C Embryo Today
2007
, vol. 
81
 (pg. 
204
-
214
)
Sharkey
JT
Puttaramu
R
Word
RA
Olcese
J
Melatonin synergizes with oxytocin to enhance contractility of human myometrial smooth muscle cells
J Clin Endocrinol Metab
2009
, vol. 
94
 (pg. 
421
-
427
)
Sharkey
JT
Cable
C
Olcese
J
Melatonin sensitizes human myometrial cells to oxytocin in a PKCα/ERK-dependent manner
J Clin Endocrinol Metab
2010
, vol. 
95
 (pg. 
2902
-
2908
)
Siddiqui
N
Hladunewich
M
Understanding the link between the placenta and future cardiovascular disease
Trends Cardiovasc Med
2011
, vol. 
21
 (pg. 
188
-
193
)
Slominski
RM
Reiter
RJ
Schlabritz-Loutsevitch
N
Ostrom
RS
Slominski
AT
Melatonin membrane receptors in peripheral tissues: distribution and functions
Mol Cell Endocrinol
2012
, vol. 
351
 (pg. 
152
-
166
)
Srinivasan
V
Lauterbach
EC
Ho
KY
Acuna-Castroviejo
D
Zakaria
R
Brzezinski
A
Melatonin in antinociception: its therapeutic applications
Curr Neuropharmacol
2012
, vol. 
10
 (pg. 
167
-
178
)
Stankov
B
Reiter
RJ
Melatonin receptors: current status, facts and hypotheses
Life Sci
1990
, vol. 
46
 (pg. 
971
-
982
)
Stehle
JH
Saade
A
Rawashdeh
O
Ackermann
K
Jilg
A
Sebesteny
T
Maronde
E
A survey of molecular details in the human pineal gland in the light of phylogeny, structure, function and chronobiological diseases
J Pineal Res
2011
, vol. 
51
 (pg. 
17
-
43
)
Sugino
N
Reactive oxygen species in ovarian physiology
Reprod Med Biol
2005
, vol. 
4
 (pg. 
31
-
44
)
Summa
KC
Vitaterna
MH
Turek
FW
Environment perturbation of the circadian clock disrupts pregnancy in the mouse
PLoS
2012
, vol. 
7
 pg. 
e637668
 
Swanson
LW
Cowan
WM
The efferent connections of the suprachiasmatic nucleus of the hypothalamus
J Comp Neurol
1975
, vol. 
160
 (pg. 
1
-
12
)
Tamarkin
L
Westrom
WK
Hamill
AI
Goldman
BD
Effect of melatonin on the reproductive system of male and female Syrian hamsters: diurnal rhythm in sensitivity to melatonin
Endocrinology
1976
, vol. 
99
 (pg. 
1534
-
1541
)
Tamura
H
Nakamura
Y
Terron
MP
Flores
LJ
Manchester
LC
Tan
DX
Sugino
N
Reiter
RJ
Melatonin and pregnancy in the human
Reprod Toxicol
2008a
, vol. 
25
 (pg. 
291
-
303
)
Tamura
H
Takasaki
A
Miwa
I
Taniguchi
K
Maekawa
R
Asada
H
Taketani
T
Matsuoka
A
Yamagata
Y
Shimamura
K
, et al. 
Oxidative stress impairs oocyte quality and melatonin protects oocytes from free radical damage and improves fertilization rates
J Pineal Res
2008b
, vol. 
44
 (pg. 
280
-
287
)
Tamura
H
Takasaki
A
Taketani
T
Tanabe
M
Kuzuka
F
Lee
L
Tamura
I
Maekawa
R
Asada
H
Yamagata
Y
, et al. 
Melatonin as a free radical scavenger in the ovarian follicle
Endocr J
2013
, vol. 
60
 (pg. 
1
-
13
)
Tan
DX
Chen
LD
Poeggeler
B
Manchester
LC
Reiter
RJ
Melatonin: a potent, endogenous hydroxyl radical scavenger
Endocr J
1993
, vol. 
1
 (pg. 
57
-
60
)
Tan
DX
Reiter
RJ
Manchester
LC
Yan
MT
El-Sawi
M
Sainz
RM
Mayo
JC
Kohen
R
Allegra
M
Hardeland
R
Chemical and physical properties and potential mechanisms: melatonin as a broad spectrum antioxidant and free radical scavenger
Curr Top Med Chem
2002
, vol. 
2
 (pg. 
181
-
197
)
Tan
DX
Manchester
LC
Hardeland
R
Lopez-Burillo
S
Mayo
JC
Sainz
RM
Reiter
RJ
Melatonin: a hormone, a tissue factor, an autocoid, a paracoid, and an antioxidant vitamin
J Pineal Res
2003
, vol. 
34
 (pg. 
75
-
78
)
Tan
DX
Manchester
LC
Terron
MP
Flores
LJ
Reiter
RJ
One molecule, many derivatives: a never-ending interaction of melatonin with reactive oxygen and nitrogen species?
J Pineal Res
2007
, vol. 
42
 (pg. 
28
-
42
)
Tan
DX
Manchester
LC
Liu
X
Rosales-Corral
SA
Acuna-Castroviejo
D
Reiter
RJ
Mitochondria and chloroplasts as the original sites of melatonin synthesis: a hypothesis related to melatonin's primary function and evolution in eukaryotes
J Pineal Res
2013
, vol. 
54
 (pg. 
127
-
138
)
Thut
G
Miniussi
C
Gross
J
The functional importance of rhythmic activity in the brain
Curr Biol
2012
, vol. 
22
 (pg. 
R658
-
R663
)
Tomas
SZ
Prusac
IK
Roje
D
Tadin
I
Trophoblast apoptosis in placentas from pregnancies complicated with pre-eclampsia
Gynecol Obstet Invest
2011
, vol. 
71
 (pg. 
250
-
255
)
Tomas-Zapico
C
Coto-Montes
A
A proposed mechanism to explain the stimulatory effect of melatonin on antioxidative enzymes
J Pineal Res
2005
, vol. 
39
 (pg. 
99
-
104
)
Torres-Farfan
C
Rocco
V
Monso
C
Valenzuela
FJ
Campino
C
Germain
A
Torrealba
F
Valenzuela
GJ
Seron-Ferre
M
Maternal melatonin effects on clock gene expression in a nonhuman primate fetus
Endocrinology
2006
, vol. 
147
 (pg. 
4618
-
4626
)
Uguz
AC
Cig
B
Espino
J
Bejarano
I
Naziroglu
M
Rodriguez
AB
Pariente
JA
Melatonin potentiates chemotherapy-induced cytotoxicity and apoptosis in pancreatic tumor cells
J Pineal Res
2012
, vol. 
53
 (pg. 
91
-
98
)
Urata
Y
Honma
S
Goto
S
Todoroki
S
Iida
T
Cho
S
Honma
K
Kondo
T
Melatonin induces gamma-glutamylcysteine synthetase mediated by activator protein-1 in human vascular endothelial cells
Free Radic Bio Med
1999
, vol. 
27
 (pg. 
838
-
847
)
Vacas
MI
Del Zar
MM
Martinuzzo
M
Cardinali
DP
Binding sites for [3H]-melatonin in humans platelets
J Pineal Res
1991
, vol. 
11
 (pg. 
135
-
139
)
Vaillancourt
C
Lanoix
D
Le Bellego
F
Daoud
G
Lafond
J
Involvement of MAPK signaling in human villous trophoblast differentiation
Mini-Rev Med Chem
2009
, vol. 
9
 (pg. 
962
-
973
)
Varcoe
TJ
Wight
N
Voultsios
A
Salkeld
MD
Kennaway
DJ
Chronic phase shifts of the photoperiod throughout pregnancy programs glucose intolerance and insulin resistance in the rat
PLoS One
2011
, vol. 
6
 pg. 
e18504
 
Vatish
M
Steer
PJ
Blanks
AM
Horn
M
Thorton
S
Diurnal variation is lost in preterm deliveries before 28 weeks of gestation
Br J Obstet Gynecol
2010
, vol. 
107
 (pg. 
1145
-
1148
)
Vaughan
GM
Pelham
RW
Pang
SF
Loughlin
LL
Wilson
KM
Sandock
KL
Vaughan
MK
Koslow
SH
Reiter
RJ
Nocturnal elevation of plasma melatonin and urinary 5-hydroxyindoleacetic acid in young men: attempts at modification by brief changes in environmental lighting and sleep and by autonomic drugs
J Clin Endocrinol Metab
1976
, vol. 
42
 (pg. 
752
-
764
)
Venegas
C
Garcia
JA
Escames
G
Ortiz
F
Lopez
A
Doerrier
C
Garcia-Corzo
L
Lopez
LC
Reiter
RJ
Acuna-Castroviejo
D
Extrapineal melatonin: analysis of its subcellular distribution and daily fluctuations
J Pineal Res
2012
, vol. 
52
 (pg. 
217
-
227
)
Waddell
BJ
Wharfe
MD
Crew
RC
Mark
PJ
A rhythmic placenta? Circadian variation, clock genes and placental function
Placenta
2012
, vol. 
33
 (pg. 
533
-
539
)
Wang
H
Li
L
Zhao
M
Zhang
ZH
Zhang
C
Ji
YL
Meng
XH
Xu
DX
Melatonin alleviates lipopolysaccharide-induced placental cellular stress response in mice
J Pineal Res
2011
, vol. 
50
 (pg. 
418
-
426
)
Wang
J
Xiao
X
Zhang
Y
Shi
D
Chen
W
Fu
L
Liu
L
Xie
F
Kang
T
Huang
W
, et al. 
Simultaneous modulation of COX-2, p300, Akt, and Apaf-1 signaling by melatonin to inhibit proliferation and induce apoptosis in breast cancer cells
J Pineal Res
2012
, vol. 
53
 (pg. 
77
-
90
)
Watanabe
T
Kojima
M
Tomida
S
Nakamura
TJ
Yamamura
T
Nakao
N
Yasuo
S
Yoshimura
T
Ebihara
S
Peripheral clock gene expression in CS mice with bimodal locomotor rhythms
Neurosci Res
2006
, vol. 
54
 (pg. 
295
-
301
)
Wehr
TA
The durations of human melatonin secretion and sleep respond to changes in daylength
J Clin Endocrinol Metab
1991
, vol. 
73
 (pg. 
1276
-
1280
)
Weinert
D
Ontogenetic development of the mammalian circadian system
Chronobiol Int
2005
, vol. 
22
 (pg. 
179
-
205
)
Weinstein
L
Syndrome of hemolysis, elevated liver enzymes, and low platelet count: a severe consequence of hypertension in pregnancy
Am J Obstet Gynecol
1982
, vol. 
142
 (pg. 
159
-
167
)
Wergeland
E
Strand
K
Working conditions and prevalence of pre-eclampsia, Norway 1989
Int J Gynecol Obstet
1997
, vol. 
58
 (pg. 
189
-
196
)
Wharfe
MD
Mark
PJ
Waddell
BJ
Circadian variation in placental and hepatic clock genes in rat pregnancy
Endocrinology
2011
, vol. 
152
 (pg. 
3552
-
3560
)
Zahn
V
Hattensperger
W
Circadian rhythm of pregnancy contractions
Z Geburtshilfe Perinatol
1993
, vol. 
197
 (pg. 
1
-
10
)
Zhu
JL
Hjollund
NH
Andersen
AM
Olsen
J
Shift work, job stress, and late fetal loss: the National Birth Cohort in Denmark
J Occup Environ Med
2004
, vol. 
46
 (pg. 
1144
-
1149
)