Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Sandrine M. Dupre is active.

Publication


Featured researches published by Sandrine M. Dupre.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Entrainment of disrupted circadian behavior through inhibition of casein kinase 1 (CK1) enzymes

Qing Jun Meng; Elizabeth S. Maywood; David A. Bechtold; Wei Qun Lu; Jian Li; Julie Gibbs; Sandrine M. Dupre; Johanna E. Chesham; Francis Rajamohan; John D. Knafels; Blossom Sneed; Laura E. Zawadzke; Jeffrey F. Ohren; Kevin Walton; Travis T. Wager; Michael H. Hastings; Andrew Loudon

Circadian pacemaking requires the orderly synthesis, posttranslational modification, and degradation of clock proteins. In mammals, mutations in casein kinase 1 (CK1) ε or δ can alter the circadian period, but the particular functions of the WT isoforms within the pacemaker remain unclear. We selectively targeted WT CK1ε and CK1δ 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, CK1δ is the principal regulator of the clock period: pharmacological inhibition of CK1δ, but not CK1ε, 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 CK1δ 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 CK1δ 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 CK1δ offers an avenue for therapeutic modulation of perturbed circadian behavior.


Endocrinology | 2008

Identification of Melatonin-Regulated Genes in the Ovine Pituitary Pars Tuberalis, a Target Site for Seasonal Hormone Control

Sandrine M. Dupre; Dave Burt; Richard Talbot; Alison Downing; Daphne Mouzaki; D. Waddington; Benoît Malpaux; Julian R. E. Davis; Gerald A. Lincoln; Andrew Loudon

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.


Neuroendocrinology | 2011

Encoding and Decoding Photoperiod in the Mammalian Pars Tuberalis

Sandrine M. Dupre

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.


Journal of Neuroendocrinology | 2011

Evidence for RGS4 modulation of melatonin and thyrotrophin signalling pathways in the pars tuberalis.

Sandrine M. Dupre; Hugues Dardente; Mike J. Birnie; Andrew Loudon; Gerald A. Lincoln; David G. Hazlerigg

In mammals, the pineal hormone melatonin is secreted nocturnally and acts in the pars tuberalis (PT) of the anterior pituitary to control seasonal neuroendocrine function. Melatonin signals through the type 1 Gi‐protein coupled melatonin receptor (MT1), inhibiting adenylate cyclase (AC) activity and thereby reducing intracellular concentrations of the second messenger, cAMP. Because melatonin action ceases by the end of the night, this allows a daily rise in cAMP levels, which plays a key part in the photoperiodic response mechanism in the PT. In addition, melatonin receptor desensitisation and sensitisation of AC by melatonin itself appear to fine‐tune this process. Opposing the actions of melatonin, thyroid‐stimulating hormone (TSH), produced by PT cells, signals through its cognate Gs‐protein coupled receptor (TSH‐R), leading to increased cAMP production. This effect may contribute to increased TSH production by the PT during spring and summer, and is of considerable interest because TSH plays a pivotal role in seasonal neuroendocrine function. Because cAMP stands at the crossroads between melatonin and TSH signalling pathways, any protein modulating cAMP production has the potential to impact on photoperiodic readout. In the present study, we show that the regulator of G‐protein signalling RGS4 is a melatonin‐responsive gene, whose expression in the PT increases some 2.5‐fold after melatonin treatment. Correspondingly, RGS4 expression is acutely sensitive to changing day length. In sheep acclimated to short days (SP, 8 h light/day), RGS4 expression increases sharply following dark onset, peaking in the middle of the night before declining to basal levels by dawn. Extending the day length to 16 h (LP) by an acute 8‐h delay in lights off causes a corresponding delay in the evening rise of RGS4 expression, and the return to basal levels is delayed some 4 h into the next morning. To test the hypothesis that RGS4 expression modulates interactions between melatonin‐ and TSH‐dependent cAMP signalling pathways, we used transient transfections of MT1, TSH‐R and RGS4 in COS7 cells along with a cAMP‐response element luciferase reporter (CRE‐luc). RGS4 attenuated MT1‐mediated inhibition of TSH‐stimulated CRE‐luc activation. We propose that RGS4 contributes to photoperiodic sensitivity in the morning induction of cAMP‐dependent gene expression in the PT.


Current Biology | 2007

Circannual clocks: annual timers unraveled in sheep.

Sandrine M. Dupre; Andrew Loudon

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.


Molecular Endocrinology | 2013

Npas4 Is Activated by Melatonin, and Drives the Clock Gene Cry1 in the Ovine Pars Tuberalis

A. West; Sandrine M. Dupre; Le Yu; I.R. Paton; K. Miedzinska; Alan S. McNeilly; Julian R. E. Davis; David W. Burt; Andrew Loudon

Seasonal mammals integrate changes in the duration of nocturnal melatonin secretion to drive annual physiologic cycles. Melatonin receptors within the proximal pituitary region, the pars tuberalis (PT), are essential in regulating seasonal neuroendocrine responses. In the ovine PT, melatonin is known to influence acute changes in transcriptional dynamics coupled to the onset (dusk) and offset (dawn) of melatonin secretion, leading to a potential interval-timing mechanism capable of decoding changes in day length (photoperiod). Melatonin offset at dawn is linked to cAMP accumulation, which directly induces transcription of the clock gene Per1. The rise of melatonin at dusk induces a separate and distinct cohort, including the clock-regulated genes Cry1 and Nampt, but little is known of the up-stream mechanisms involved. Here, we used next-generation sequencing of the ovine PT transcriptome at melatonin onset and identified Npas4 as a rapidly induced basic helix-loop-helix Per-Arnt-Sim domain transcription factor. In vivo we show nuclear localization of NPAS4 protein in presumptive melatonin target cells of the PT (α-glycoprotein hormone-expressing cells), whereas in situ hybridization studies identified acute and transient expression in the PT of Npas4 in response to melatonin. In vitro, NPAS4 forms functional dimers with basic helix loop helix-PAS domain cofactors aryl hydrocarbon receptor nuclear translocator (ARNT), ARNT2, and ARNTL, transactivating both Cry1 and Nampt ovine promoter reporters. Using a combination of 5′-deletions and site-directed mutagenesis, we show NPAS4-ARNT transactivation to be codependent upon two conserved central midline elements within the Cry1 promoter. Our data thus reveal NPAS4 as a candidate immediate early-response gene in the ovine PT, driving molecular responses to melatonin.


Current Biology | 2010

A molecular switch for photoperiod responsiveness in mammals

Hugues Dardente; Cathy A. Wyse; Mike J. Birnie; Sandrine M. Dupre; Andrew Loudon; Gerald A. Lincoln; David G. Hazlerigg


Journal of Endocrinology | 2006

Photoperiodic regulation of cellular retinoic acid-binding protein 1, GPR50 and nestin in tanycytes of the third ventricle ependymal layer of the Siberian hamster

Perry Barrett; Elena A. Ivanova; E. Scott Graham; Alexander W. Ross; Dana Wilson; Helene Ple; Julian G. Mercer; Francis J. P. Ebling; Sandrine Schuhler; Sandrine M. Dupre; Andrew Loudon; Peter J. Morgan


American Journal of Physiology-endocrinology and Metabolism | 2008

Altered metabolism in the melatonin-related receptor (GPR50) knockout mouse

Elena A. Ivanova; David A. Bechtold; Sandrine M. Dupre; John Brennand; Perry Barrett; Simon M. Luckman; Andrew Loudon


Current Biology | 2010

Identification of Eya3 and TAC1 as Long-Day Signals in the Sheep Pituitary

Sandrine M. Dupre; Katarzyna Miedzinska; Chloe V. Duval; Le Yu; Robert L. Goodman; Gerald A. Lincoln; Julian R. E. Davis; Alan S. McNeilly; David D. Burt; Andrew Loudon

Collaboration


Dive into the Sandrine M. Dupre's collaboration.

Top Co-Authors

Avatar

Andrew Loudon

University of Manchester

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Le Yu

University of Edinburgh

View shared research outputs
Top Co-Authors

Avatar

Dave Burt

University of Edinburgh

View shared research outputs
Top Co-Authors

Avatar

Bob Paton

University of Edinburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge