Mammalian target of rapamycin signaling modulates photic entrainment of the suprachiasmatic circadian clock

R Cao, A Li, H Cho, B Lee, K Obrietan - Journal of Neuroscience, 2010 - Soc Neuroscience
R Cao, A Li, H Cho, B Lee, K Obrietan
Journal of Neuroscience, 2010Soc Neuroscience
Inducible gene expression appears to be an essential event that couples light to entrainment
of the master mammalian circadian clock located in the suprachiasmatic nucleus (SCN) of
the hypothalamus. Recently, we reported that light triggers phase-dependent activation of
the mammalian target of rapamycin (mTOR) signaling pathway, a major regulator of protein
synthesis, in the SCN, thus raising the possibility that mTOR-evoked mRNA translation
contributes to clock entrainment. Here, we used a combination of cellular, molecular, and …
Inducible gene expression appears to be an essential event that couples light to entrainment of the master mammalian circadian clock located in the suprachiasmatic nucleus (SCN) of the hypothalamus. Recently, we reported that light triggers phase-dependent activation of the mammalian target of rapamycin (mTOR) signaling pathway, a major regulator of protein synthesis, in the SCN, thus raising the possibility that mTOR-evoked mRNA translation contributes to clock entrainment. Here, we used a combination of cellular, molecular, and behavioral assays to address this question. To this end, we show that the in vivo infusion of the mTOR inhibitor rapamycin led to a significant attenuation of the phase-delaying effect of early-night light. Conversely, disruption of mTOR during the late night augmented the phase-advancing effect of light. To assess the role of mTOR signaling within the context of molecular entrainment, the effects of rapamycin on light-induced expression of PERIOD1 and PERIOD2 were examined. At both the early- and late-night time points, abrogation of mTOR signaling led to a significant attenuation of light-evoked PERIOD protein expression. Our results also reveal that light-induced mTOR activation leads to the translation of mRNAs with a 5′-terminal oligopyrimidine tract such as eukaryotic elongation factor 1A and the immediate early gene JunB. Together, these data indicate that the mTOR pathway functions as potent and selective regulator of light-evoked protein translation and SCN clock entrainment.
Soc Neuroscience