当前位置: 首页 > 期刊 > 《内分泌学杂志》 > 2006年第2期 > 正文
编号:11416432
Multiple Effects of Melatonin on Rhythmic Clock Gene Expression in the Mammalian Pars Tuberalis
http://www.100md.com 《内分泌学杂志》
     School of Biological Sciences (J.D.J, D.G.H.), University of Aberdeen, Zoology Building, Aberdeen AB24 2TZ, Scotland, United Kingdom

    Neurobiologie des Rhythmes laboratory, Unite Mixte de Recherche 7518 Centre National de la Recherche Scientifique/Universite Louis Pasteur (B.B.T., M.M.-P.),67084 Strasbourg, France

    Medical Research Council Human Reproductive Sciences Unit (H.A., G.A.L.), Centre for Reproductive Biology, The Queen’s Medical Research Institute, Edinburgh EH16 4TJ, Scotland, United Kingdom

    Abstract

    In mammals, changing day length modulates endocrine rhythms via nocturnal melatonin secretion. Studies of the pituitary pars tuberalis (PT) suggest that melatonin-regulated clock gene expression is critical to this process. Here, we considered whether clock gene rhythms continue in the PT in the absence of melatonin and whether the effects of melatonin on the expression of these genes are temporally gated. Soay sheep acclimated to long photoperiod (LP) were transferred to constant light for 24 h, suppressing endogenous melatonin secretion. Animals were infused with melatonin at 4-h intervals across the final 24 h, and killed 3 h after infusion. The expression of five clock genes (Per1, Per2, Cry1, Rev-erb, and Bmal1) was measured by in situ hybridization. In sham-treated animals, PT expression of Per1, Per2, and Rev-erb showed pronounced temporal variation despite the absence of melatonin, with peak times occurring earlier than predicted under LP. The time of peak Bmal1 expression remained LP-like, whereas Cry1 expression was continually low. Melatonin infusion induced Cry1 expression at all times and suppressed other genes, but only when they showed high expression in sham-treated animals. Hence, 3 h after melatonin treatment, clock gene profiles were driven to a similar state, irrespective of infusion time. In contrast to the PT, melatonin infusions had no clear effect on clock gene expression in the suprachiasmatic nuclei. Our results provide the first example of acute sensitivity of multiple clock genes to one endocrine stimulus and suggest that rising melatonin levels may reset circadian rhythms in the PT, independently of previous phase.

    Introduction

    MELATONIN, THE PRINCIPAL product of the vertebrate pineal gland, undergoes marked temporal variation in secretion and drives a wide range of rhythmic physiology. In mammals, the synthesis and secretion of pineal melatonin occurs at night, under the control of the circadian clock present in the suprachiasmatic nuclei (SCN) of the hypothalamus. The duration of this nocturnal signal varies in proportion to the length of the night, and thence melatonin secretion transduces both daily and seasonal time throughout the body (1).

    The pituitary pars tuberalis (PT) is strongly implicated in the photoperiodic regulation of prolactin secretion and, because of its high density of melatonin receptors, has become a key model tissue for the understanding of melatonin signal readout (2, 3, 4). It is now clear that melatonin is a key regulator of clock gene expression in the mammalian PT, because modulation of the endogenous profile of melatonin secretion, by manipulation of ambient photoperiod or by administration of exogenous melatonin, changes PT clock gene profiles (5, 6, 7, 8, 9).

    To date, the documented effects of melatonin in the PT can be divided into those occurring in the morning (Per1) and those occurring in the evening (Cry1, Rev-rb). The former depend on melatonin withdrawal at the end of the night, probably mediated by the resultant increase in cAMP production (2, 5). In rodents unable to synthesize melatonin, either because of genetic deficiency (10) or pinealectomy (11), rhythmic expression of Per1 expression is absent, suggesting that melatonin is necessary for maintained rhythmic gene expression in this tissue.

    Collectively, these observations suggest that the PT contains a circadian oscillator that is either driven or modulated by melatonin and raise questions about 1) whether a rhythmic melatonin signal is necessary for the 24-h variation of clock gene expression in the PT and 2) whether melatonin actions on gene expression in the PT is restricted to time windows around the morning and evening changes in illumination. To test these ideas, we returned to the sheep as a model because this species has been extensively used for investigations into the molecular basis of melatonin-dependent photoperiodic responses (2, 6, 12). We placed animals in constant light for one 24-h cycle (thereby suppressing endogenous melatonin secretion) and treated them at 4-h intervals with either melatonin-containing sc implants or sham procedure.

    Materials and Methods

    Animals and housing

    All animals were treated in accordance with the U.K. Animals (Scientific Procedures) Act, 1986. Forty-eight pubertal female Soay sheep, obtained from specialist breeders in Scotland (12), were brought indoors in winter (November). They were housed in light-sealed rooms and fed a standardized diet of grass pellets (500 g per animal; Vitagrass, Cumbria, UK) given daily 1 h into the light phase. Hay and water were available ad libitum. White fluorescent strip lights provided approximately 160 lux at the animals’ eye level during the light phase, whereas dim red light (<5 lux) was provided during the dark phase.

    Light and melatonin treatment

    The animals were initially acclimatized under a winter-like short photoperiod (SP; 8 h light, 16 h darkness) for 4 wk and then exposed to long photoperiod (LP; 16 h light, 8 h dark) for 6 wk. Blood samples were collected twice weekly from the jugular vein from representative animals (n = 12), and the blood plasma separated by centrifugation within 30 min and stored at –20 C, to measure prolactin concentration as evidence of photoresponsiveness.

    Over the final 24 h of the experiment, lights were left on to suppress endogenous pineal melatonin secretion. Animals were treated with a single sc implant of melatonin (Regulin; CEVA Animal Health, Chesham, Bucks, UK) placed on the inside of the hind leg using a standard injector, or a sham treatment with no implant as control at 4-h intervals across the final 24 h (n = 4 per time point), and killed 3 h after treatment by pentobarbital injection. Immediately before death, blood samples were collected from the jugular vein and processed as above to permit analysis of the effect of treatments on melatonin concentration. The hypothalamus and upper pituitary gland were removed within 10 min of death and snap frozen in isopentane kept at –30 C by dry ice. The expression of five clock genes (Per1, Per2, Cry1, Rev-erb, and Bmal1) was measured in the PT and SCN by in situ hybridization.

    RIA

    Prolactin concentrations were measured by a validated RIA (13). The lower limit of sensitivity was 0.5 ng/ml prolactin standard (NIH-PRL-S13), and the intraassay coefficient of variation was 8.3%. Melatonin concentrations were also measured by a standard RIA (14), using a commercial antibody (PF-1288; SPI-BIO, Paris, France). The sensitivity of the assay was 5 pg/ml, and the intraassay coefficient of variation was 12.0%.

    In situ hybridization

    Coronal cryosections (20 μm) cut through the SCN and PT were thaw-mounted onto poly-L-lysine-coated glass slides and stored at –80 C. Expression of the clock genes Per1, Per2, Cry1, Bmal1, and Rev-erb was then detected using radioactive riboprobes and procedures described previously (15).

    Statistics

    Weekly changes in plasma prolactin concentration during acclimation to LP were analyzed by one-way ANOVA with repeated measures. Clock gene expression profiles in sham-treated animals were analyzed by one-way ANOVA. The comparison between melatonin and sham treatment was analyzed by two-way ANOVA and Bonferroni’s post hoc testing when appropriate.

    Results

    Photosensitivity of experimental animals

    The pretreatment exposure to LP induced a significant (P < 0.01, one-way ANOVA with repeated measures) increase in plasma prolactin concentrations from 19.2 ± 4.6, at the change from SP to LP, to 52.4 ± 11.1 ng/ml at wk 6, illustrating the physiological and photosensitive state of the experimental animals before the final day on constant light (LL) (Fig. 1).

    Effect of treatment on endocrine profiles

    LL treatment resulted in suppressed plasma melatonin concentration across the 24-h cycle (Fig. 2A; melatonin concentration in sham group was 10.4 ± 0.9 pg/ml, mean ± SEM, n = 24). Melatonin implants significantly increased plasma melatonin concentration within 3 h (Fig. 2A; P < 0.001 for effect of treatment, two-way ANOVA). This effect was independent of infusion time (P > 0.05 for treatment x time interaction, two-way ANOVA). The resulting plasma melatonin concentrations (216.9 ± 28.0 pg/ml, mean ± SEM, n = 24) were within the nocturnal physiological range commonly observed in this species (12).

    Although prolactin concentration appeared to be low in sham-treated animals killed at zeitgeber time 16 (ZT16) (Fig. 2B), there was no statistically significant 24-h variation in either the sham- or melatonin-treated animals (P > 0.05 for effect of time in both experimental groups, one-way ANOVA). Administration of melatonin during LL decreased mean plasma prolactin concentration within 3 h of treatment (Fig. 2B; plasma prolactin concentration was 29.8 ± 4.0 vs. 18.6 ± 2.6 ng/ml, sham vs. melatonin-treated group, mean ± SEM, n = 24; P < 0.01 for effect of treatment, two-way ANOVA). This acute response to melatonin was not time dependent (P > 0.05 for treatment x time interaction, two-way ANOVA).

    Clock gene expression in the PT

    Despite the suppressed plasma melatonin concentrations in sham-treated animals, the expression of four of the five clock genes analyzed in the PT exhibited significant (P < 0.05, one-way ANOVA) variation over the 24-h cycle (Fig. 3). For Rev-erb and Per1, which show peak PT expression in the early light phase in LP acclimated animals (6, 8), a melatonin-independent expression peak was seen after approximately 20 h of constant light treatment. For Per2, which peaks slightly later than Per1 under LP (6), a melatonin-independent increase in expression was seen after 24 h of exposure to LL. Similar to the case in LP acclimated animals (6), Bmal1 expression peaked between ZT12 and ZT16 in LL.

    In contrast to the other clock genes, Cry1 expression in sham-treated animals did not vary significantly over the LL sampling period (P > 0.05, one-way ANOVA) and was low at all sampling points (Fig. 4). Melatonin infusion for a period of 3 h caused a large increase in the expression of Cry1 mRNA in the PT (Fig. 4) (P < 0.001 for effect of treatment, two-way ANOVA). This increase occurred at all sampling points throughout the 24-h cycle (P < 0.05 melatonin vs. sham, two-way ANOVA with Bonferroni’s post hoc test). The expression of Per1, Per2, Rev-erb, and Bmal1 was also sensitive to melatonin treatment, but in these instances the effects of melatonin were inhibitory and time dependent (P < 0.05 melatonin vs. sham, two-way ANOVA with Bonferroni’s post hoc test) (Fig. 5). As a result of these effects, the expression of clock genes in the PT 3 h after melatonin treatment was qualitatively similar, irrespective of the time of infusion; Cry1 mRNA was elevated, whereas all other mRNAs were suppressed (Figs. 4 and 5).

    Clock gene expression in the SCN

    The expression of all five clock genes was rhythmic in the SCN over 24 h of LL exposure (P < 0.001 for effect of time, two-way ANOVA). Melatonin had a significant effect on Rev-erb expression (P = 0.05 for effect of treatment, two-way ANOVA), although this effect was not dependent upon time of melatonin treatment (P > 0.05 for treatment x time interaction, two-way ANOVA). For all other genes, melatonin had no significant effect on mRNA expression (P > 0.05 effect of treatment, two-way ANOVA) (Fig. 6).

    Discussion

    Recent studies have identified rhythmic expression of multiple clock genes in the mammalian PT and proposed models by which these genes may mediate the photoperiodic effects of melatonin (16). Here, we have used photosensitive Soay sheep to demonstrate that clock gene rhythms persist in the absence of endogenous melatonin secretion. Furthermore, in these animals, melatonin infusions acutely alter the expression of multiple clock genes at all phases of the 24-h cycle in the PT, without any corresponding effects in the SCN. These data are consistent with the possibility that the PT functions as a circadian oscillator that can be synchronized by an increase in melatonin concentration in the bloodstream, irrespective of when this occurs relative to lights on. More generally, this study provides the first example of acute responsiveness of multiple clock genes to a single defined endocrine stimulus.

    Since the classical lesioning experiments performed over 30 yr ago (17, 18), the SCN has been viewed as a master circadian pacemaker that drives and synchronizes overt rhythms of physiology and behavior. The recent identification of circadian oscillators in peripheral tissues and in immortalized cell lines suggests that the role of the SCN is primarily to coordinate circadian oscillations throughout the organism (19) and focuses attention on the mechanisms through which such circadian synchronization takes place. The precise manner by which a given SCN-dependent circadian signal influences peripheral oscillator function may be expected to reflect the physiological functions of the tissues upon which it acts. In mammals, melatonin is one such SCN-dependent circadian output, whose primary function is to modulate neuroendocrine physiology, by relaying information about changes in day length (20). The pineal melatonin signal communicates the phase of night onset and of the subsequent dawn; melatonin is secreted continuously between these transitions, giving a signal whose duration is proportional to the length of the night. This ability of the melatonin signal to represent night duration is crucial for photoperiodic responses in seasonal mammals (21).

    The PT is a largely homogeneous population of type 1 melatonin (MT1) receptor-expressing thyrotrophic cells (22, 23), making it an ideal tissue for the study of melatonin action. Long-term loss of melatonin synthesis or loss of functional MT1 receptors abolishes rhythmic clock gene expression in the mammalian PT (10, 11, 24, 25), indicating that the molecular clockwork of this tissue is ultimately melatonin dependent. The data presented here, however, demonstrate clear temporal variation in PT expression of four core clock genes over a 24-h period in the absence of a rhythmic melatonin signal and thus strongly support the existence of an endogenous PT oscillator. We therefore suspect that arrhythmic Per1 gene expression in the PT of hamsters that have been pinealectomized for 7 d (11) derives from progressive damping of the PT circadian oscillator. Based on recent studies of damping in cell culture systems (26, 27, 28), we speculate that this is a result of desynchronization of circadian oscillators in individual PT cells. Accordingly, in genetically melatonin-synthesis- or melatonin-receptor-deficient mice (10, 24, 25), PT gene expression is likely arrhythmic because PT cells never become synchronized during fetal or postnatal development.

    The present study has examined only one aspect of the information inherent in the endogenous melatonin signal: rising melatonin levels associated with night onset. Our data suggest that a dusk-like response (high Cry1, low Per, Rev-erb, and Bmal1) can be elicited by increasing melatonin levels occurring at any phase throughout the 24-h cycle. This suggests that PT cells exhibit a type 0 resetting response (29) to rising melatonin levels, that is to say that the PT molecular clockwork resets to a dusk state in response to rising melatonin, independent of its state immediately beforehand. This dusk state is transient, because our previous data show that a state of low Cry1 and high Bmal1 expression develops during continued melatonin presence through the middle to the end of the night (6). Because the effects of melatonin on cAMP-dependent signal transduction and gene expression persist during prolonged melatonin exposure (2), it is likely that cAMP-independent mechanisms account for disappearance of the dusk state under physiological conditions.

    Interestingly, the PT of the Japanese quail also rhythmically expresses clock genes, of which Cry1 may be acutely induced by light. However, photic induction of Cry1 is subject to circadian gating in this tissue (30), suggesting that the avian PT may contain a more robust oscillator than is the case in mammals.

    For sheep in a natural environment, the times of dawn and dusk move in opposite directions during the annual cycle, raising the possibility of a similar flexibility in the resetting response of the PT oscillator to declining melatonin levels associated with dawn. Although we have yet to perform an experiment explicitly to address this, our published data agree with this prediction; the timing of the dawn state, characterized by high Per1, can be delayed by an acute light manipulation in the preceding evening that delays the dawn-associated decline in melatonin secretion (8). Hence the circadian clockwork of the sheep PT shows a remarkable plasticity in its response to changing patterns of melatonin secretion.

    Early work in the mammalian PT focused on the ability of melatonin to modulate adenylate cyclase activity (2). Melatonin-regulated expression of Per1, which is considered a cAMP-regulated immediate-early response gene in the PT (31), is likely to occur through this pathway (24). However, other clock genes are not directly sensitive to cAMP signaling, suggesting that melatonin-dependent signaling may occur by additional and as yet undefined signaling pathways. Consistent with this possibility, previous studies of many tissues, including the ovine PT, have revealed a wide range of intracellular pathways that are sensitive to melatonin (32). At present, we cannot entirely rule out the possibility that induction of Cry1 drives changes in the expression of other clock genes. However, we feel that this putative mechanism is unlikely, because it would require not only rapid transcription and translation of Cry1 but also a high level of mRNA instability for other clock genes to be affected within the short interval (3 h) between melatonin infusion and tissue collection used in this study.

    Additional to its role in seasonal neuroendocrine control, melatonin may influence the function of the SCN itself. Exogenous melatonin administration has been shown to synchronize circadian behavioral rhythmicity in rodents (33, 34) and in humans (35), and melatonin can reportedly phase-shift circadian rhythms of electrical activity in mouse SCN slice preparations (36). Knockout of the MT1 and MT2 melatonin receptor subtypes abolishes phase-shifting responses to melatonin in mice and acute effects of melatonin on SCN electrical activity (37, 38). Hence it is possible that circadian effects of melatonin are mediated through melatonin receptors located in the SCN. Nevertheless, a recent study reported no effect of melatonin injection on clock gene expression in the rat SCN (39). In sheep, melatonin receptors are at best weakly expressed in the SCN (40), and in the present study a mild effect of melatonin injection was seen for only one gene, Rev-erb, at one time point only. Possibly our data and that of Poirel et al. (39) reflect different coupling between melatonin receptors and circadian molecular clockwork in SCN compared with PT cells. Alternatively the heterogeneous cellular composition of the SCN and its sensitivity to multiple inputs in addition to melatonin (41) may account for these results.

    A final point of interest is the regulation of plasma prolactin concentration in this study. We have previously described a LP-induced increase in prolactin concentration within 24 h (8). In this study, the animals required approximately 4–6 wk of LP exposure before showing such a rise. This difference can be explained by two reasons. In the current study, animals were likely to exhibit early refractoriness to shortening winter photoperiod when they were transferred to controlled lighting regimes; furthermore, animals were not habituated to blood sampling and it is therefore likely that initial prolactin concentrations were affected by stress of experimental manipulation at this stage. During the final 24 h of this experiment, melatonin caused an acute inhibition of prolactin release in the sheep under constant light. This is unlikely to reflect a direct effect on lactotroph cells, which do not appear to express melatonin receptors (22, 42) and are unresponsive to melatonin in primary cell culture (43). It is unclear whether melatonin acts at the level of the PT or hypothalamus to acutely suppress prolactin concentration. Although hypothalamic monoamines are not believed to drive photoperiodic changes of prolactin secretion (44, 45), there is some evidence that melatonin may stimulate secretion of the prolactin-inhibiting factor dopamine from hypothalamic neurons (46, 47). Alternatively, melatonin infusions may have acutely inhibited the secretion of prolactin-releasing factor(s), termed tuberalin, from the PT (43, 48, 49, 50, 51). However, the acute effects of melatonin on tuberalin secretion are as yet unknown.

    In summary, we propose that the PT may become an important model tissue to address the function of peripheral circadian oscillators as well as the mechanisms of photoperiodic time measurement. A strength of the PT as a circadian model is that it has a well defined circadian input, melatonin. As well as testing the generality of the results described here in other species, we are currently developing in vitro techniques to further evaluate the effect of melatonin on the PT circadian clockwork. These studies will extend beyond the scope of the current in vivo work and will hopefully provide novel insights into the chronotherapeutic effects of melatonin.

    Acknowledgments

    We are grateful to Marshall Building staff (Edinburgh) and Nigel Graham and Jean-Michel Fustin (Aberdeen) for technical assistance and to Prof. Ueli Schibler (University of Geneva, Switzerland) for the generous gift of the Rev-erb probe template. Purified preparations of ovine prolactin were provided by the National Hormone and Peptide Program.

    Footnotes

    Present address for J.D.J.: School of Biomedical and Molecular Sciences, University of Surrey, Guildford GU2 7XH, United Kingdom.

    This work was supported by the Medical Research Council (to H.A. and G.A.L.) and a grant from the Biotechnology and Biological Sciences Research Council (to D.G.H.).

    First Published Online November 3, 2005

    1 J.D.J. and B.B.T. are joint first authors.

    Abbreviations: LL, Constant light; LP, long photoperiod; MT1, melatonin type 1; PT, pars tuberalis; SCN, suprachiasmatic nuclei; SP, short photoperiod; ZT, zeitgeber time.

    Accepted for publication October 26, 2005.

    References

    Simonneaux V, Ribelayga C 2003 Generation of the melatonin endocrine message in mammals: a review of the complex regulation of melatonin synthesis by norepinephrine, peptides, and other pineal transmitters. Pharmacol Rev 55:325–395

    Hazlerigg DG, Morgan PJ, Messager S 2001 Decoding photoperiodic time and melatonin in mammals: what can we learn from the pars tuberalis J Biol Rhythms 16:326–335

    Johnston JD 2004 Photoperiodic regulation of prolactin secretion: changes in intra-pituitary signalling and lactotroph heterogeneity. J Endocrinol 180:351–356

    Kell CA, Stehle JH 2005 Just the two of us: melatonin and adenosine in rodent pituitary function. Ann Med 37:105–120

    Messager S, Ross AW, Barrett P, Morgan PJ 1999 Decoding photoperiodic time through Per1 and ICER gene amplitude. Proc Natl Acad Sci USA 96:9938–9943

    Lincoln G, Messager S, Andersson H, Hazlerigg D 2002 Temporal expression of seven clock genes in the suprachiasmatic nucleus and the pars tuberalis of the sheep: evidence for an internal coincidence timer. Proc Natl Acad Sci USA 99:13890–13895

    Dardente H, Menet JS, Poirel VJ, Streicher D, Gauer F, Vivien-Roels B, Klosen P, Pevet P, Masson-Pevet M 2003 Melatonin induces Cry1 expression in the pars tuberalis of the rat. Mol Brain Res 114:101–106

    Hazlerigg DG, Andersson H, Johnston JD, Lincoln GA 2004 Molecular characterization of the long-day response in the Soay sheep, a seasonal mammal. Curr Biol 14:334–339

    Johnston JD, Ebling FJP, Hazlerigg DG 2005 Photoperiod regulates multiple gene expression in the suprachiasmatic nuclei and pars tuberalis of the Siberian hamster (Phodopus sungorus). Eur J Neurosci 21:2967–2974

    Sun ZS, Albrecht U, Zhuchenko O, Bailey J, Eichele G, Lee CC 1997 RIGUI, a putative mammalian ortholog of the Drosophila Period gene. Cell 90:1003–1011

    Messager S, Garabette ML, Hastings MH, Hazlerigg DG 2001 Tissue-specific abolition of Per1 expression in the pars tuberalis by pinealectomy in the Syrian hamster. NeuroReport 12:579–582

    Lincoln GA 2002 Neuroendocrine regulation of seasonal gonadotrophin and prolactin rhythms: lessons from the Soay ram model. Reprod Suppl 19:131–147

    McNeilly AS, Andrews P 1974 Purification and characterization of caprine prolactin. J Endocrinol 60:359–367

    Fraser S, Cowan P, Franclin M, Franey C, Arendt J 1983 Direct radioimmunoassay for melatonin in plasma. Clin Chem 29:396–397

    Lincoln GA, Johnston JD, Andersson H, Wagner G, Hazlerigg DG 2005 Photorefractoriness in mammals: dissociating a seasonal timer from the circadian-based photoperiod response. Endocrinology 146:3782–3790

    Lincoln GA, Andersson H, Loudon A 2003 Clock genes in calendar cells as the basis of annual timekeeping in mammals: a unifying hypothesis. J Endocrinol 179:1–13

    Moore RY, Eichler VB 1972 Loss of a circadian adrenal corticosterone rhythm following suprachiasmatic lesions in the rat. Brain Res 42:201–206

    Stephan FK, Zucker I 1972 Circadian rhythms in drinking behavior and locomotor activity of rats are eliminated by hypothalamic lesions. Proc Natl Acad Sci USA 69:1583–1586

    Reppert SM, Weaver DR 2002 Coordination of circadian timing in mammals. Nature 418:935–941

    Goldman BD 2001 Mammalian photoperiodic system: formal properties and neuroendocrine mechanisms of photoperiodic time measurement. J Biol Rhythms 16:283–301

    Bartness TJ, Powers JB, Hastings MH, Bittman EL, Goldman BD 1993 The timed infusion paradigm for melatonin delivery: what has it taught us about the melatonin signal, its reception, and the photoperiodic control of seasonal responses J Pineal Res 15:161–190

    Klosen P, Bienvenu C, Demarteau O, Dardente H, Guerrero H, Pevet P, Masson-Pevet M 2002 The mt1 melatonin receptor and ROR receptor are co-localized in specific TSH-immunoreactive cells in the pars tuberalis of the rat pituitary. J Histochem Cytochem 50:1647–1657

    Dardente H, Klosen P, Pevet P, Masson-Pevet M 2003 MT1 melatonin receptor mRNA expressing cells in the pars tuberalis of the European hamster: effect of photoperiod. J Neuroendocrinol 15:778–786

    von Gall C, Garabette ML, Kell CA, Frenzel S, Dehghani F, Schumm-Draeger PM, Weaver DR, Korf HW, Hastings MH, Stehle JH 2002 Rhythmic gene expression in pituitary depends on heterologous sensitization by the neurohormone melatonin. Nat Neurosci 5:234–238.

    von Gall C, Weaver DR, Moek J, Jilg A, Stehle JH, Korf HW 2005 Melatonin plays a crucial role in the regulation of rhythmic clock gene expression in the mouse pars tuberalis. Ann NY Acad Sci 1040:508–511

    Nagoshi E, Saini C, Bauer C, Laroche T, Naef F, Schibler U 2004 Circadian gene expression in individual fibroblasts: cell-autonomous and self-sustained oscillators pass time to daughter cells. Cell 119:693–705

    Welsh DK, Yoo SH, Liu AC, Takahashi JS, Kay SA 2004 Bioluminescence imaging of individual fibroblasts reveals persistent, independently phased circadian rhythms of clock gene expression. Curr Biol 14:2289–2295

    Carr AJ, Whitmore D 2005 Imaging of single light-responsive clock cells reveals fluctuating free-running periods. Nat Cell Biol 7:319–321

    Roenneberg T, Daan S, Merrow M 2003 The art of entrainment. J Biol Rhythms 18:183–194

    Yasuo S, Watanabe M, Tsukada A, Takagi T, Iigo M, Shimada K, Ebihara S, Yoshimura T 2004 Photoinducible phase-specific light induction of Cry1 gene in the pars tuberalis of Japanese quail. Endocrinology 145:1612–1616

    Morgan PJ, Ross AW, Graham ES, Adam C, Messager S, Barrett P 1998 oPer1 is an early response gene under photoperiodic regulation in the ovine pars tuberalis. J Neuroendocrinol 10:319–323

    Vanecek J 1998 Cellular mechanisms of melatonin action. Physiol Rev 78:687–721

    Redman J, Armstrong S, Ng KT 1983 Free-running activity rhythms in the rat: entrainment by melatonin. Science 219:1089–1091

    Grosse J, Velickovic A, Davis FC 1996 Entrainment of Syrian hamster circadian activity rhythms by neonatal melatonin injections Am J Physiol 270:R533–R540

    Arendt J, Skene DJ 2005 Melatonin as a chronobiotic. Sleep Med Rev 9:25–39

    Gillette MU, McArthur AJ 1996 Circadian actions of melatonin at the suprachiasmatic nucleus. Behav Brain Res 73:135–139

    Liu C, Weaver DR, Jin X, Shearman LP, Pieschl RL, Gribkoff VK, Reppert SM 1997 Molecular dissection of two distinct actions of melatonin on the suprachiasmatic circadian clock. Neuron 19:91–102

    Jin X, von Gall C, Pieschl RL, Gribkoff VK, Stehle JH, Reppert SM, Weaver DR 2003 Targeted disruption of the mouse Mel(1b) melatonin receptor. Mol Cell Biol 23:1054–1060

    Poirel VJ, Boggio V, Dardente H, Pevet P, Masson-Pevet M, Gauer F 2003 Contrary to other non-photic cues, acute melatonin injection does not induce immediate changes of clock gene mRNA expression in the rat suprachiasmatic nuclei. Neuroscience 120:745–755

    Bittman EL, Weaver DR 1990 The distribution of melatonin binding sites in neuroendocrine tissues of the ewe. Biol Reprod 43:986–993

    Piggins HD, Cutler DJ 2003 The roles of vasoactive intestinal polypeptide in the mammalian circadian clock. J Endocrinol 177:7–15

    Williams LM, Lincoln GA, Mercer JG, Barrett P, Morgan PJ, Clarke IJ 1997 Melatonin receptors in the brain and pituitary gland of hypothalamo-pituitary disconnected Soay rams. J Neuroendocrinol 9:639–643

    Stirland JA, Johnston JD, Cagampang FR, Morgan PJ, Castro MG, White MR, Davis JR, Loudon AS 2001 Photoperiodic regulation of prolactin gene expression in the Syrian hamster by a pars tuberalis-derived factor. J Neuroendocrinol 13:147–157

    Lincoln GA, Clarke IJ 1995 Evidence that melatonin acts in the pituitary gland through a dopamine-independent mechanism to mediate effects of daylength on the secretion of prolactin in the ram. J Neuroendocrinol 7:637–643

    Lincoln GA, Clarke IJ 2002 Noradrenaline and dopamine regulation of prolactin secretion in sheep: role in prolactin homeostasis but not photoperiodism. J Neuroendocrinol 14:36–44

    Shieh KR, Chu YS, Pan JT 1997 Circadian change of dopaminergic neuron activity: effects of constant light and melatonin. Neuroreport 8:2283–2287

    Chu YS, Shieh KR, Yuan ZF, Pan JT 2000 Stimulatory and entraining effect of melatonin on tuberoinfundibular dopaminergic neuron activity and inhibition on prolactin secretion. J Pineal Res 28:219–226

    Hazlerigg DG, Hastings MH, Morgan PJ 1996 Production of a prolactin releasing factor by the ovine pars tuberalis. J Neuroendocrinol 8:489–492

    Morgan PJ, Webster CA, Mercer JG, Ross AW, Hazlerigg DG, MacLean A, Barrett P 1996 The ovine pars tuberalis secretes a factor(s) that regulates gene expression in both lactotropic and nonlactotropic pituitary cells. Endocrinology 137:4018–4026

    Lafarque M, Oliveros L, Aguado L 1998 Effect of adenohypophyseal pars tuberalis secretions on pars distalis prolactin liberation. Medicina 58:36–40

    Johnston JD, Cagampang FR, Stirland JA, Carr AJ, White MR, Davis JR, Loudon AS 2003 Evidence for an endogenous per1- and ICER-independent seasonal timer in the hamster pituitary gland. FASEB J 17:810–815(Jonathan D. Johnston1, Benjamin B. Tourn)