Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Neuroendocrinology ; 94(2): 101-12, 2011.
Article in English | MEDLINE | ID: mdl-21778697

ABSTRACT

In mammals, the nocturnal melatonin signal is well established as a key hormonal indicator of seasonal changes in day-length, providing the brain with an internal representation of the external photoperiod. The pars tuberalis (PT) of the pituitary gland is the major site of expression of the G-coupled receptor MT1 in the brain and is considered as the main site of integration of the photoperiodic melatonin signal. Recent studies have revealed how the photoperiodic melatonin signal is encoded and conveyed by the PT to the brain and the pituitary, but much remains to be resolved. The development of new animal models and techniques such as cDNA arrays or high throughput sequencing has recently shed the light onto the regulatory networks that might be involved. This review considers the current understanding of the mechanisms driving photoperiodism in the mammalian PT with a particular focus on the seasonal prolactin secretion.


Subject(s)
Melatonin/physiology , Photoperiod , Pituitary Gland, Anterior/physiology , Humans , Melatonin/biosynthesis , Receptor, Melatonin, MT1/biosynthesis , Receptor, Melatonin, MT1/genetics , Seasons
2.
Curr Biol ; 20(24): 2193-8, 2010 Dec 21.
Article in English | MEDLINE | ID: mdl-21129971

ABSTRACT

Seasonal synchronization based on day length (photoperiod) allows organisms to anticipate environmental change. Photoperiodic decoding relies on circadian clocks, but the underlying molecular pathways have remained elusive [1]. In mammals and birds, photoperiodic responses depend crucially on expression of thyrotrophin ß subunit RNA (TSHß) in the pars tuberalis (PT) of the pituitary gland [2-4]. Now, using our well-characterized Soay sheep model [2], we describe a molecular switch governing TSHß transcription through the circadian clock. Central to this is a conserved D element in the TSHß promoter, controlled by the circadian transcription factor thyrotroph embryonic factor (Tef). In the PT, long-day exposure rapidly induces expression of the coactivator eyes absent 3 (Eya3), which synergizes with Tef to maximize TSHß transcription. The pineal hormone melatonin, secreted nocturnally, sets the phase of rhythmic Eya3 expression in the PT to peak 12 hr after nightfall. Additionally, nocturnal melatonin levels directly suppress Eya3 expression. Together, these effects form a switch triggering a strong morning peak of Eya3 expression under long days. Species variability in the TSHß D element influences sensitivity to TEF, reflecting species variability in photoperiodic responsiveness. Our findings define a molecular pathway linking the circadian clock to the evolution of seasonal timing in mammals.


Subject(s)
Circadian Clocks/physiology , Mammals/physiology , Photoperiod , Sheep/physiology , Thyrotropin, beta Subunit/genetics , Animals , Base Sequence , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Melatonin/metabolism , Molecular Sequence Data , Pituitary Gland/anatomy & histology , Pituitary Gland/physiology , Promoter Regions, Genetic , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Seasons , Sequence Alignment , Thyrotropin, beta Subunit/metabolism , Transcription, Genetic
3.
Proc Natl Acad Sci U S A ; 107(34): 15240-5, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20696890

ABSTRACT

Circadian pacemaking requires the orderly synthesis, posttranslational modification, and degradation of clock proteins. In mammals, mutations in casein kinase 1 (CK1) epsilon or delta can alter the circadian period, but the particular functions of the WT isoforms within the pacemaker remain unclear. We selectively targeted WT CK1epsilon and CK1delta using pharmacological inhibitors (PF-4800567 and PF-670462, respectively) alongside genetic knockout and knockdown to reveal that CK1 activity is essential to molecular pacemaking. Moreover, CK1delta is the principal regulator of the clock period: pharmacological inhibition of CK1delta, but not CK1epsilon, significantly lengthened circadian rhythms in locomotor activity in vivo and molecular oscillations in the suprachiasmatic nucleus (SCN) and peripheral tissue slices in vitro. Period lengthening mediated by CK1delta inhibition was accompanied by nuclear retention of PER2 protein both in vitro and in vivo. Furthermore, phase mapping of the molecular clockwork in vitro showed that PF-670462 treatment lengthened the period in a phase-specific manner, selectively extending the duration of PER2-mediated transcriptional feedback. These findings suggested that CK1delta inhibition might be effective in increasing the amplitude and synchronization of disrupted circadian oscillators. This was tested using arrhythmic SCN slices derived from Vipr2(-/-) mice, in which PF-670462 treatment transiently restored robust circadian rhythms of PER2::Luc bioluminescence. Moreover, in mice rendered behaviorally arrhythmic by the Vipr2(-/-) mutation or by constant light, daily treatment with PF-670462 elicited robust 24-h activity cycles that persisted throughout treatment. Accordingly, selective pharmacological targeting of the endogenous circadian regulator CK1delta offers an avenue for therapeutic modulation of perturbed circadian behavior.


Subject(s)
Casein Kinase 1 epsilon/antagonists & inhibitors , Casein Kinase Idelta/antagonists & inhibitors , Circadian Rhythm/physiology , Animals , Base Sequence , Casein Kinase 1 epsilon/physiology , Casein Kinase Idelta/deficiency , Casein Kinase Idelta/genetics , Casein Kinase Idelta/physiology , Circadian Rhythm/drug effects , Gene Knockdown Techniques , In Vitro Techniques , Mice , Mice, Knockout , Mice, Transgenic , Period Circadian Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , RNA, Small Interfering/genetics , Receptors, Vasoactive Intestinal Peptide, Type II/deficiency , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Suprachiasmatic Nucleus/drug effects , Suprachiasmatic Nucleus/physiology
4.
Curr Biol ; 20(9): 829-35, 2010 May 11.
Article in English | MEDLINE | ID: mdl-20434341

ABSTRACT

Seasonally breeding mammals such as sheep use photoperiod, encoded by the nocturnal secretion of the pineal hormone melatonin, as a critical cue to drive hormone rhythms and synchronize reproduction to the most optimal time of year. Melatonin acts directly on the pars tuberalis (PT) of the pituitary, regulating expression of thyrotropin, which then relays messages back to the hypothalamus to control reproductive circuits. In addition, a second local intrapituitary circuit controls seasonal prolactin (PRL) release via one or more currently uncharacterized low-molecular-weight peptides, termed "tuberalins," of PT origin. Studies in birds have identified the transcription factor Eya3 as the first molecular response activated by long photoperiod (LP). Using arrays and in situ hybridization studies, we demonstrate here that Eya3 is the strongest LP-activated gene in sheep, revealing a common photoperiodic molecular response in birds and mammals. We also demonstrate TAC1 (encoding the tachykinins substance P and neurokinin A) to be strongly activated by LP within the sheep PT. We show that these PRL secretagogues act on primary pituitary cells and thus are candidates for the elusive PT-expressed tuberalin seasonal hormone regulator.


Subject(s)
Genes, Regulator/physiology , Photoperiod , Pituitary Gland/physiology , Sheep/physiology , Animals , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Genes, Regulator/genetics , Oligonucleotide Array Sequence Analysis , Periodicity , Prolactin/blood , Prolactin/metabolism , RNA, Messenger/genetics , Receptors, Tachykinin/physiology , Seasons , Sheep/genetics , Tachykinins/physiology , Thyrotrophs/physiology
5.
Endocrinology ; 149(11): 5527-39, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18669596

ABSTRACT

The pars tuberalis (PT) of the pituitary gland expresses a high density of melatonin (MEL) receptors and is believed to regulate seasonal physiology by decoding changes in nocturnal melatonin secretion. Circadian clock genes are known to be expressed in the PT in response to the decline (Per1) and onset (Cry1) of MEL secretion, but to date little is known of other molecular changes in this key MEL target site. To identify transcriptional pathways that may be involved in the diurnal and photoperiod-transduction mechanism, we performed a whole genome transcriptome analysis using PT RNA isolated from sheep culled at three time points over the 24-h cycle under either long or short photoperiods. Our results reveal 153 transcripts where expression differs between photoperiods at the light-dark transition and 54 transcripts where expression level was more globally altered by photoperiod (all time points combined). Cry1 induction at night was associated with up-regulation of genes coding for NeuroD1 (neurogenic differentiation factor 1), Pbef / Nampt (nicotinamide phosphoribosyltransferase), Hif1alpha (hypoxia-inducible factor-1alpha), and Kcnq5 (K+ channel) and down-regulation of Rorbeta, a key clock gene regulator. Using in situ hybridization, we confirmed day-night differences in expression for Pbef / Nampt, NeuroD1, and Rorbeta in the PT. Treatment of sheep with MEL increased PT expression for Cry1, Pbef / Nampt, NeuroD1, and Hif1alpha, but not Kcnq5. Our data thus reveal a cluster of Cry1-associated genes that are acutely responsive to MEL and novel transcriptional pathways involved in MEL action in the PT.


Subject(s)
Gene Expression Regulation/drug effects , Melatonin/pharmacology , Pituitary Gland/drug effects , Pituitary Hormones/genetics , Seasons , Sheep/genetics , Animals , Circadian Rhythm/genetics , Female , Gene Expression Profiling , Male , Oligonucleotide Array Sequence Analysis , Photoperiod , Pituitary Gland/metabolism
6.
Am J Physiol Endocrinol Metab ; 294(1): E176-82, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17957037

ABSTRACT

The X-linked orphan receptor GPR50 shares 45% homology with the melatonin receptors, yet its ligand and physiological function remain unknown. Here we report that mice lacking functional GPR50 through insertion of a lacZ gene into the coding sequence of GPR50 exhibit an altered metabolic phenotype. GPR50 knockout mice maintained on normal chow exhibit lower body weight than age-matched wild-type littermates by 10 wk of age. Furthermore, knockout mice were partially resistant to diet-induced obesity. When placed on a high-energy diet (HED) for 5 wk, knockout mice consumed significantly more food per unit body weight yet exhibited an attenuated weight gain and reduced body fat content compared with wild-type mice. Wheel-running activity records revealed that, although GPR50 knockout mice showed no alteration of circadian period, the overall levels of activity were significantly increased over wild types in both nocturnal and diurnal phases. In line with this, basal metabolic rate (O2 consumption, CO2 production, and respiratory quotient) was found to be elevated in knockout mice. Using in situ hybridization (wild-type mice) and beta-galactosidase activity (from LacZ insertion element in knockout mice), brain expression of GPR50 was found to be restricted to the ependymal layer of the third ventricle and dorsomedial nucleus of the hypothalamus. GPR50 expression was highly responsive to energy status, showing a significantly reduced expression following both fasting and 5 wk of HED. These data implicate GPR50 as an important regulator of energy metabolism.


Subject(s)
Dorsomedial Hypothalamic Nucleus/metabolism , Energy Metabolism/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Obesity/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Animals , Circadian Rhythm/physiology , Fasting/physiology , Male , Mice , Mice, Knockout , Motor Activity/physiology , Obesity/physiopathology , Weight Gain/physiology
7.
Curr Biol ; 17(6): R216-7, 2007 Mar 20.
Article in English | MEDLINE | ID: mdl-17371762

ABSTRACT

A recent study has revealed new insight into how the annual clock may drive seasonal hormone rhythms in mammals; the data suggest that melatonin-receptor-containing cells in the pituitary gland may operate as key calendar cells, transmitting seasonal temporal information to the endocrine system.


Subject(s)
Circadian Rhythm , Seasons , Sheep/physiology , Animals , Melatonin/physiology , Photoperiod , Receptors, Melatonin/physiology , Sexual Behavior, Animal , Signal Transduction
8.
J Endocrinol ; 191(3): 687-98, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17170225

ABSTRACT

Tanycytes in the ependymal layer of the third ventricle act both as a barrier and a communication gateway between the cerebrospinal fluid, brain and portal blood supply to the pituitary gland. However, the range, importance and mechanisms involved in the function of tanycytes remain to be explored. In this study, we have utilized a photoperiodic animal to examine the expression of three unrelated gene sequences in relation to photoperiod-induced changes in seasonal physiology and behaviour. We demonstrate that cellular retinol binding protein [corrected] (CRBP1), a retinoic acid transport protein, GPR50, an orphan G-protein-coupled receptor and nestin, an intermediate filament protein, are down-regulated in short-day photoperiods. The distribution of the three sequences is very similar, with expression located in cells with tanycyte morphology in the region of the ependymal layer where tanycytes are located. Furthermore, CRBP1 expression in the ependymal layer is shown to be independent of a circadian clock and altered testosterone levels associated with testicular regression in short photo-period. Pinealectomy of Siberian hamsters demonstrates CRBP1 expression is likely to be dependent on melatonin output from the pineal gland. This provides evidence that tanycytes are seasonally responsive cells and are likely to be an important part of the mechanism to facilitate seasonal physiology and behaviour in the Siberian hamster.


Subject(s)
Ependyma/metabolism , Intermediate Filament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Phodopus/physiology , Photoperiod , Receptors, G-Protein-Coupled/metabolism , Receptors, Retinoic Acid/metabolism , Animals , Biomarkers/analysis , Blood-Brain Barrier , Cricetinae , Ependyma/cytology , Gene Expression Regulation , In Situ Hybridization/methods , Male , Mice , Mice, Knockout , Nestin , Phodopus/anatomy & histology , Pineal Gland/physiology , Pineal Gland/surgery , Receptors, G-Protein-Coupled/analysis , Stem Cells/cytology , Third Ventricle
9.
Med Sci (Paris) ; 21(10): 854-9, 2005 Oct.
Article in French | MEDLINE | ID: mdl-16197903

ABSTRACT

The synthesis and secretion of thyroid hormones (TH: T3, T4) must be strictly regulated. TH act on their own production via a negative feedback system. The synthesis of thyrotropin-releasing hormone (TRH), produced in the hypothalamus, and thyrotropin (TSH) in the pituitary is inhibited at the transcriptional level by TH. TRH and TSH stimulate production of TH. An outstanding, still open, question is the molecular basis of T3-dependent transcription repression of TRH and TSH genes. However, some regulatory components have been identified, with the b-TH receptor (TRb) playing a specific regulatory role (versus TRa) in the negative feedback effects of T3 on production of TRH and TSH. Moreover, the N-terminus of TRb is known to be a key element in this regulation. A hypothesis to explain this isoform specificity could be that TRb and TRa interact differentially with transcriptional comodulators. Thus, it is critical to characterize these comodulators and to analyse their contribution to the transcription regulation of TRH.


Subject(s)
Gene Expression Regulation , Thyrotropin-Releasing Hormone/biosynthesis , Thyrotropin-Releasing Hormone/genetics , Humans , Hypothalamus/physiology , Pituitary Gland/physiology , Protein Isoforms , Transcription, Genetic
10.
Endocrinology ; 145(5): 2337-45, 2004 May.
Article in English | MEDLINE | ID: mdl-14726446

ABSTRACT

Thyroid hormones (TH) are essential regulators of vertebrate development and metabolism. Central mechanisms governing their production have evolved, with the beta-TH receptor (TRbeta) playing a key regulatory role in the negative feedback effects of circulating TH levels on production of hypothalamic TRH and hypophyseal TSH. Both TRbeta-isoforms (TRbeta1 and TRbeta2) are expressed in the hypothalamus and pituitary. However, their respective roles in TH-dependent transcriptional regulation of TRH are undefined. We confirmed the preferential role of TRbeta vs. TRalpha isoforms in TRH regulation in wild-type mice in vivo by using the TRbeta preferential agonist GC-1. We next determined the effects of tissue-specific rescue of TRbeta1 and TRbeta2 isoforms by somatic gene transfer in hypothalami of TRbeta null (TRbeta(-/-)) mice. TH-dependent TRH transcriptional repression was impaired in TRbeta(-/-) mice, but was restored by cotransfection of either TRbeta1 or TRbeta2 into the hypothalamus. TRbeta1, but not TRbeta2, displayed a role in ligand-independent activation. In situ hybridization was used to examine endogenous TRH expression in the paraventricular nucleus of the hypothalamus of TRbeta(-/-) or TRalpha null (TRalpha(o/o)) mice under different thyroid states. In contrast to published data on TRbeta2(-/-) mice, we found that both ligand-independent TRH activation and ligand-dependent TRH repression were severely impaired in TRbeta(-/-) mice. This study thus provides functional in vivo data showing that both TRbeta1 and TRbeta2 isoforms have specific roles in regulating TRH transcription.


Subject(s)
Gene Expression Regulation , Hypothalamus/chemistry , Thyroid Hormone Receptors beta/physiology , Thyrotropin-Releasing Hormone/genetics , Animals , Luciferases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/analysis , Recombinant Fusion Proteins , Thyroid Hormone Receptors beta/deficiency , Thyroid Hormone Receptors beta/genetics , Transcription, Genetic/genetics , Transfection , Triiodothyronine/pharmacology
11.
Mol Endocrinol ; 16(7): 1652-66, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12089358

ABSTRACT

The beta thyroid hormone receptor (TRbeta), but not TRalpha1, plays a specific role in mediating T(3)-dependent repression of hypothalamic TRH transcription. To investigate the structural basis of isoform specificity, we compared the transcriptional regulation and DNA binding obtained with chimeric and N-terminally deleted TRs. Using in vivo transfection assays to follow hypothalamic TRH transcription in the mouse brain, we found that TRbeta1 and chimeras with the TRbeta1 N terminus did not affect either transcriptional activation or repression from the rat TRH promoter, whereas N-terminally deleted TRbeta1 impaired T(3)-dependent repression. TRalpha1 or chimeras with the TRalpha1 N terminus reduced T(3)-independent transcriptional activation and blocked T(3)-dependent repression of transcription. Full deletion of the TRalpha1 N terminus restored ligand-independent activation of transcription. No TR isoform specificity was seen after transcription from a positive thyroid hormone response element. Gel mobility assays showed that all TRs tested bound specifically to the main negative thyroid hormone response element in the TRH promoter (site 4). Addition of neither steroid receptor coactivator 1 nor nuclear extracts from the hypothalamic paraventricular nuclei revealed any TR isoform specificity in binding to site 4. Thus N-terminal sequences specify TR T(3)-dependent repression of TRH transcription but not DNA recognition, emphasizing as yet unknown neuron-specific contributions to protein-promoter interactions in vivo.


Subject(s)
Feedback, Physiological , Hypothalamus/physiology , Receptors, Thyroid Hormone/metabolism , Thyrotropin-Releasing Hormone/genetics , Transcription, Genetic , Animals , Binding Sites , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dimerization , Histone Acetyltransferases , Mice , Mice, Inbred Strains , Nuclear Receptor Coactivator 1 , Promoter Regions, Genetic , Protein Isoforms , Rats , Receptors, Thyroid Hormone/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Response Elements , Substrate Specificity , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Receptors beta , Thyrotropin-Releasing Hormone/metabolism , Transcription Factors/metabolism , Triiodothyronine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...