Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Jul 12.
Published in final edited form as: Adv Genet. 2011;74:175–230. doi: 10.1016/B978-0-12-387690-4.00006-4

Genetics of Circadian Rhythms in Mammalian Model Organisms

Phillip L Lowrey 1, Joseph S Takahashi 2
PMCID: PMC3709251  NIHMSID: NIHMS483358  PMID: 21924978

Abstract

The mammalian circadian system is a complex hierarchical temporal network which is organized around an ensemble of uniquely coupled cells comprising the principal circadian pacemaker in the suprachiasmatic nucleus of the hypothalamus. This central pacemaker is entrained each day by the environmental light/dark cycle and transmits synchronizing cues to cell-autonomous oscillators in tissues throughout the body. Within cells of the central pacemaker and the peripheral tissues, the underlying molecular mechanism by which oscillations in gene expression occur involves interconnected feedback loops of transcription and translation. Over the past 10 years we have learned much regarding the genetics of this system, including how it is particularly resilient when challenged by single-gene mutations, how accessory transcriptional loops enhance the robustness of oscillations, how epigenetic mechanisms contribute to the control of circadian gene expression, and how, from coupled neuronal networks, emergent clock properties arise. Here we will explore the genetics of the mammalian circadian system from cell-autonomous molecular oscillations, to interactions among central and peripheral oscillators and ultimately, to the daily rhythms of behavior observed in the animal.

I. INTRODUCTION

The rising and setting of the sun each day causes predictable environmental changes to which most organisms on earth have adapted by evolving endogenous biological timing systems with a period of approximately 24 hours (Young and Kay, 2001). These circadian (~24 hr) clocks anticipate environmental cycles and control daily rhythms in biochemistry, physiology and behavior. Across phyla, all circadian clocks share several fundamental properties: they are synchronized (entrained) each day to external cues, they are self-sustained and produce oscillations that persist in the absence of any external cues, they are temperature compensated such that temperature changes in the physiological range do not alter their endogenous period, and of particular relevance to this review, they are cell-autonomous and genetically-determined. In all of the major model organisms in which circadian rhythms have been studied there has emerged a central organizing principle of the molecular clockwork: within cells a set of clock genes and their protein products together participate in autoregulatory feedback loops of transcription and translation to produce an oscillation with a period of about 24 hr (Lowrey and Takahashi, 2004; Takahashi et al., 2008).

Recent work, however, has prompted a reappraisal of the transcription/translation model as the sole generative mechanism of the molecular circadian oscillator in mammals. For example, it is now clear that oscillations of some mammalian core clock components are dispensable for circadian function (Fan et al., 2007; Liu et al., 2008), and there is some evidence, albeit preliminary, for circadian rhythms in the absence of transcription in some mammalian cells (O'Neill and Reddy, 2011). Perhaps more importantly, however, limitations of the conventional perturbation analysis methods that helped elucidate the transcription/translation model have become apparent. No longer is it sufficient to knock out a clock gene in a mouse and then assess the consequences on behavior (locomotor activity) or gene expression (changes in RNA and protein levels in cells) alone. We now appreciate that the mammalian circadian clock is a more complex hierarchical system than originally imagined, and thus understanding it requires analysis at many levels.

New technologies and clock models have revealed higher-order genetic properties of the mammalian clock system in which the elimination of one component may be compensated for by other components in ways that are more complex than simple redundancy, and they have demonstrated the important roles of accessory feedback loops and gene networks in conferring stability and robustness on the system (Ueda et al., 2005; Ukai-Tadenuma et al., 2008; Baggs et al., 2009). Further, novel approaches have elucidated the importance of networks of coupled cells from which emergent circadian clock properties arise and even buffer the system against the effects of mutations (Liu et al., 2007b; Abraham et al., 2010; Buhr et al., 2010; Ko et al., 2010). These, and other advances, are making clearer the fundamental properties of each level of organization of the mammalian circadian system from cell-autonomous molecular oscillations, to tissue-specific properties, to the interaction of central and peripheral oscillators, and ultimately to the overt daily rhythms of behavior observed in the animal.

Here we present some of the key findings in the field of mammalian circadian biology over the past 10 years and introduce many of the new technologies that are revolutionizing our understanding of the clock system. Our emphasis will be primarily on work from the principal model organism used to study mammalian biology—the mouse. Indeed, for no other mammalian model is there the extensive repertoire of experimental resources and techniques as for the mouse (Silver, 1995; Nagy et al., 2003; Hedrich and Bullock, 2004; Fox et al., 2007; Adams and van der Weyden, 2008; Blake et al., 2010). We will not, however, explore in depth the intriguing link between the mammalian circadian clock and metabolism, first proposed by McKnight and colleagues a decade ago (Rutter et al., 2002), and now well established, as it is beyond the scope of this review. Instead, we refer the reader to several recent comprehensive treatments of this specific topic (Green et al., 2008; Bass and Takahashi, 2010; Maury et al., 2010; Asher and Schibler, 2011).

II. THE BEGINNING OF MAMMALIAN CLOCK GENETICS

A. Serendipitous discovery of the Syrian hamster tau mutant

Before discussing the current state of mammalian clock genetics and the details of the molecular clockwork in mammals, we would first like to reflect back briefly on the period from approximately 1985 to 2000 when the study of mammalian clock genetics began. Indeed, it was in 1985 that Martin Ralph, at the time a graduate student in the laboratory of Michael Menaker (then at the University of Oregon), identified a single outbred Syrian hamster (Mesocricetus auratus) with an unusually early onset of locomotor activity. Following transfer from a light/dark (LD) cycle to constant darkness (DD), this animal exhibited an endogenous free-running period of 22 hr compared to 23.5 hr, the shortest previously reported circadian period for this species. Recognizing the implications of possibly discovering the first mammalian circadian mutant, Ralph had the foresight to cross this animal to wild-type hamsters and analyze the behavioral rhythms in the offspring. The free-running periods for the resulting F1 progeny (1:1 ratio; 22 hr : 24 hr) confirmed that the aberrant phenotype was heritable. Intercrosses produced an F2 generation with a 1:2:1 Mendelian ratio of 20 hr : 22 hr : 24 hr periods. Thus, this spontaneous mutation designated tau (after the circadian symbol for period length), segregated in a semidominant manner and seemed to involve a single autosomal locus (Table 1) (Ralph and Menaker, 1988).

Table 1.

Behavioral phenotypes of mutations in mouse clock and clock-related genes

Gene(s) Protein product(s) Mutant allele(s) Mutant phenotype(s) References
Clock bHLH-PAS transcription factor ClockΔ19/Δ19
Clock−/−
4 hr longer pd/arrhythmic
0.4 hr shorter pd
(Vitaterna et al., 1994)
(Debruyne et al., 2006)
Npas2 (Mop4) bHLH-PAS transcription factor Npas2−/− 0.2 hr shorter pd (Dudley et al., 2003)
Clock/Npas2 bHLH-PAS transcription factors Clock−/−/Npas2−/− Arrhythmic (DeBruyne et al., 2007a)
Bmal1 (Arntl, Mop3) bHLH-PAS transcription factor Bmal1−/− Arrhythmic (Bunger et al., 2000)
Cry1 flavoprotein Cry1−/− 1 hr shorter pd (van der Horst et al., 1999; Vitaterna et al., 1999)
Cry2 flavoprotein Cry2−/− 1 hr longer pd (Thresher et al., 1998;
van der Horst et al., 1999)
Cry1/Cry2 flavoproteins Cry1−/−/Cry2−/− Arrhythmic (van der Horst et al., 1999; Vitaterna et al., 1999)
Per1 PAS protein Per1−/− 0.7 hr shorter pd (Cermakian et al., 2001)
Per1brdml 1 hr shorter pd (Zheng et al., 2001)
Per1ldc 0.5 hr shorter pd/arrhythmic (Bae et al., 2001)
Per2 PAS protein Per2brdml 1.5 hr shorter pd/arrhythmic (Zheng et al., 1999)
Per2ldc Arrhythmic (Bae et al., 2001)
Per3 PAS protein Per3−/− 0–0.5 hr shorter pd (Shearman et al., 2000a)
Per1/Per2 PAS proteins Per1brdml/Per2brdml Arrhythmic (Zheng et al., 2001)
Per1brdml/Per2brdml Arrhythmic (Bae et al., 2001)
Per1/Cry1 PAS protein/flavoprotein Per1brdml/Cry1−/− Normal behavior (Oster et al., 2003)
Per1/Cry2 PAS protein/flavoprotein Per1brdm1/Cry2−/− <6 months, 1.5 hr longer pd;
>6 months, arrhythmic
(Oster et al., 2003)
Per2/Cry1 PAS protein/flavoprotein Per2brdm1/Cry1−/− Arrhythmic (Oster et al., 2002)
Per2/Cry2 PAS protein/flavoprotein Per2brdm1/Cry2−/− 0–0.4 hr shorter pd (Oster et al., 2002)
Rev-erbα (Nr1d1) nuclear receptor Rev-erbα−/− 0.5 hr shorter pd; disrupted
entrainment
(Preitner et al., 2002)
Rev-erbβ (Nr1d2) nuclear receptor
Rorα (Rora) nuclear receptor Rorα−/− (staggerer) 0.5 hr shorter pd; disrupted
entrainment
(Sato et al., 2004)
Rorβ (Rorb) nuclear receptor Rorβ−/− 0.5 hr longer pd (Masana et al., 2007)
Rorγ (Rorc) nuclear receptor Rorγ−/− Normal behavior (Liu et al., 2008)
Dec1 (Bhlhe40, Stra13, Sharp-2) bHLH transcription factor Stra13−/− 0.15 hr longer pd (Nakashima et al., 2008)
Dec2 (Bhlhe41, Sharp-1) bHLH transcription factor Sharp-1−/− Delayed resetting (Rossner et al., 2008)
CK1δ (Csnk1d) casein kinase 1 CK1δ+/− 0–0.5 hr longer pd (Xu et al., 2005;
Etchegaray et al., 2009)
CK1ε (Csnk1e) casein kinase 1 CK1ε−/− 0.2–0.4 hr longer pd (Meng et al., 2008;
Etchegaray et al., 2009)
*CK1εtau 4 hr shorter pd (Lowrey et al., 2000;
Meng et al., 2008)
CK1α (Csnk1a1) casein kinase 1
Fbxl3 F-box protein Fbxl3Ovtm 2 hr longer pd (Siepka et al., 2007)
Fbxl3Afh 3 hr longer pd (Godinho et al., 2007)
Bmal2 (Arntl2, Mop9, Clif) bHLH-PAS transcription factor
Pgc1a (Ppargc1a) transcriptional coactivator Pgc1a−/− 0.3 hr longer pd (Liu et al., 2007c)
Mtnr1a (Mel1a) G protein-coupled receptor Mtnr1a−/− Normal behavior (Liu et al., 1997a)
Mtnr1b (Mel1b) G protein-coupled receptor Mtnr1b−/− Normal behavior (Jin et al., 2003)
Opn4 melanopsin; opsin 4 Opn4−/− Attenuated photic responses (Panda et al., 2002b;
Ruby et al., 2002)
Dbp PAR bZIP transcription factor Dbp−/− 0.5 hr shorter pd (Lopez-Molina et al., 1997)
Vipr2 G protein-coupled receptor Vipr2−/− Disrupted locomotor rhythm (Harmar et al., 2002;
Cutler et al., 2003)
Vip peptide hormone Vip−/− 1 hr shorter pd/arrhythmic (Colwell et al., 2003)
Prok2 (PK2) secreted protein Prok2−/− Reduced locomotor activity (Li et al., 2006)
Nocturnin (Ccrn4l) deadenylase Noc−/− Normal behavior (Green et al., 2007)

Gene symbols listed here are the predominate forms used in the scientific literature; alternate forms are given in parentheses. When the predominant symbol differs from standard mouse gene nomenclature, the standard form is given in parentheses. For genes with more than two symbols in parentheses, the form adhering to standard mouse gene nomenclature is underlined.

*

First identified as a mutation in Syrian hamster (Mesocricetus auratus).

As the first mammalian circadian mutation, tau figured prominently in many studies addressing behavioral and physiological aspects of mammalian circadian biology. Perhaps the most important result obtained from the tau model was the definitive demonstration through transplantation experiments that the suprachiasmatic nucleus (SCN) of the hypothalamus harbors the central circadian pacemaker in mammals. Adult hamsters rendered behaviorally arrhythmic by SCN lesioning exhibited restored rhythmicity following transplantation of donor SCN tissue into the third ventricle. Not only was host rhythmicity rescued, but the restored rhythms had periods reflecting the genotype of the donor animal (Ralph et al., 1990). Further, when firing rate rhythms of individual SCN neurons on fixed microelectrode plates were recorded, cells from tau animals helped show that the circadian period of the whole tissue/animal is determined by averaging widely dispersed periods of individual SCN clock cells. This was the first demonstration that the tau mutation affects circadian function in a cell-autonomous manner (Liu et al., 1997b). Two additional seminal findings using the tau model include the first report that a diffusible signal can drive circadian rhythms in a mammal (Silver et al., 1996), and that SCN-independent circadian oscillators reside in the mammalian retina (Tosini and Menaker, 1996; Tosini and Menaker, 1998).

Despite the importance to the field of mammalian circadian biology of the tau model, from a genetic standpoint it is unfortunate that the mutation occurred in the Syrian hamster rather than in the mouse, a mammalian model for which even in 1985 more comprehensive genetic resources were available. Efforts to develop dense genetic maps and physical mapping reagents for the mouse were well underway at the inception of the Human Genome Project and its inclusion of the mouse as a model genome sequencing project (Muller and Grossniklaus, 2010). The Syrian hamster, however, became one of many “orphan genomes” not included in the publically-funded mapping effort (Jacob, 1996). And so the quest to clone and characterize the tau mutation would not come to fruition until twelve years after Ralph and Menaker's 1988 report of the mutant. Our laboratory, using a comparative genomics approach called positional syntenic cloning, demonstrated that the tau mutation results from a single nucleotide change in the gene encoding casein kinase I epsilon (CK1ε) (Lowrey et al., 2000).

B. Forward genetics and the Clock mutation

With the realization that the tau mutant hamster, while advantageous for physiological studies, was not immediately a genetically tractable model, our laboratory, in collaboration with William F. Dove, Lawrence H. Pinto, and Fred W. Turek, initiated a dominant circadian behavioral screen of first-generation offspring of male C57BL/6J mice mutagenized with the alkylating agent N-ethyl-N-nitrosourea (ENU). We adopted this forward genetics approach in mice encouraged by the successful mutagenesis screens in Drosophila for circadian defects some 20 years earlier by Seymour Benzer's group (Konopka and Benzer, 1971). Of the 304 animals tested, we recovered a mouse with a single-gene, semidominant mutation, Clock (for circadian locomotor output cycles kaput), that significantly lengthened its free-running period (24.8 hr) compared to wild-type C57BL/6J mice (23.3–23.8 hr). Homozygous Clock mutant animals exhibited an extremely long initial free-running period in DD of 26–29 hr, followed by a complete loss of rhythmicity after two weeks in DD (Table 1). We mapped the mutation to mouse chromosome 5 (Vitaterna et al., 1994) and subsequently identified the gene through a combination of positional cloning and transgenic rescue of the mutant phenotype (Antoch et al., 1997; King et al., 1997b). Sequence analysis revealed that the Clock mutation is caused by an A→T transversion in a splice donor site in the intron between exons 18 and 19 of the gene, resulting in a transcript missing exon 19 (ClockΔ19). This deletion disrupts the transactivation domain of the basic helix-loop-helix (bHLH)-PAS (Period-Arnt-Single-minded) transcription factor encoded by Clock. Additional genetic approaches revealed that Clock is an antimorph—a specific type of dominant negative mutation (King et al., 1997a).

Identification of the Clock mutation was proof of principle that, as in Drosophila, forward genetic screens for behavioral defects in mice are feasible (Takahashi et al., 1994; Bacon et al., 2004; Clark et al., 2004; Siepka and Takahashi, 2005). Identifying mammalian clock genes by recovering mutants was, however, not the only approach during this “birth” of mammalian clock genetics in the 1990s. Several of what proved to be mammalian core clock genes were cloned by homology to known genes in other organisms, or by identification of paralogs in the same organism. These include three mouse orthologs of the Drosophila period gene (Per1, Per2, and Per3), two mouse Cryptochrome orthologs (Cry1 and Cry2), and brain and muscle ARNT-like protein 1 (Bmal1 or Mop3), another bHLH-PAS protein, all reviewed comprehensively elsewhere (Lowrey and Takahashi, 2000; Young and Kay, 2001; Reppert and Weaver, 2002; Lowrey and Takahashi, 2004). Indeed, the 1990s witnessed the description of a molecular model of the mammalian core circadian oscillator based on a transcription/translation feedback loop with striking similarity to models proposed for other, phylogenetically divergent organisms, including Drosophila, Arabidopsis, and Neurospora (Dunlap, 1999). This rapid elucidation of the basic mechanism by which mammalian cells keep time was aptly characterized by one colleague as a “clockwork explosion” (Reppert, 1998).

III. OVERVIEW OF THE MAMMALIAN CLOCK SYSTEM

The mammalian circadian system is organized around three major physiological components: an input pathway by which environmental cues (most importantly light) are transmitted to the central or 'master' pacemaker, the central pacemaker itself, and finally a set of output pathways by which the central pacemaker regulates circadian rhythms throughout the body (Takahashi et al., 2001; Quintero et al., 2003; Lowrey and Takahashi, 2004). Light entrainment of the circadian system relies on the eye (Nelson and Zucker, 1981; Foster et al., 1991) where, within the retina, the rods and cones and a recently discovered subset (~1%) of intrinsically-photosensitive retinal ganglion cells (ipRGCs) reside (Do and Yau, 2010). The ipRGCs respond to light stimulation independently of the rod-cone system (Berson et al., 2002; Hattar et al., 2002), and are directly photosensitive owing to their expression of the photopigment melanopsin (Provencio et al., 1998; Gooley et al., 2001; Dacey et al., 2005; Fu et al., 2005; Melyan et al., 2005; Panda et al., 2005; Qiu et al., 2005). Mice lacking either the rod-cone system (Freedman et al., 1999), or melanopsin (Opn4−/−) (Panda et al., 2002b; Ruby et al., 2002; Lucas et al., 2003), exhibit normal entrainment to light. Loss of both the rod-cone system and melanopsin, however, render mice unable to entrain to photic stimuli (Hattar et al., 2003; Panda et al., 2003). Because the ipRGCs, via which all retinal input to the SCN is transmitted, receive synaptic input containing non-visual information from the rods and cones, selective destruction of the ipRGCs in mice also prevents circadian photoentrainment (Goz et al., 2008; Hatori et al., 2008).

Photic information received by the retina is transmitted via the retinohypothalamic tract (RHT) which is formed from the axons of the ipRGCs, to the bilaterally-paired suprachiasmatic nuclei of the hypothalamus, the location of the central pacemaker in mammals (Moore and Eichler, 1972; Moore and Lenn, 1972; Stephan and Zucker, 1972; Moore et al., 1995; Gooley et al., 2001). Light stimulation of the ipRGCs causes their axon terminals to release glutamate and pituitary adenylate cyclase-activating polypeptide (PACAP) onto postsynaptic SCN neurons (Ebling, 1996; Hannibal, 2002; Hannibal et al., 2004; Michel et al., 2006; Morin and Allen, 2006). Glutamate-induced calcium influx activates several protein kinase pathways in SCN neurons which ultimately lead to phosphorylation of Ca2+/cAMP-response element binding protein (CREB) (Golombek and Rosenstein, 2010). Within the promoters of many core clock genes, reside Ca2+/cAMP-response elements (CREs) to which phospho-CREB homodimers bind to activate transcription (Zhang et al., 2005). Two particularly important CREB clock targets with respect to photic entrainment are the Per1 and Per2 genes, both of which contain CREs in their promoters (Travnickova-Bendova et al., 2002) and are rapidly induced in SCN neurons by nocturnal light exposure (Albrecht et al., 1997; Shearman et al., 1997; Shigeyoshi et al., 1997). Circadian rhythms of locomotor activity in mice are phase advanced or phase delayed depending on the time at night during which a photic stimulus occurs (Golombek and Rosenstein, 2010). The resulting increase in PER protein presumably affects the molecular clock in SCN neurons by opposing the action of the positive effectors of the core clock feedback loop discussed later (Yan and Silver, 2004).

Each SCN of the mouse contains approximately 10,000 neurons (Abrahamson and Moore, 2001). When dissociated from SCN tissue (Welsh et al., 1995; Liu et al., 1997b; Herzog et al., 1998; Honma et al., 1998) or when grown as immortalized cells (Earnest et al., 1999), these neurons can independently generate self-sustained circadian rhythms. Intact SCN neurons, however, couple to form a network that expresses synchronized rhythms (Welsh et al., 2010). Ongoing work seeks to clarify the nature of the coupling mechanisms that give rise to the unique SCN network, yet it is clear that neurotransmitters, neuropeptides, gap junctions, and chemical synaptic mechanisms are involved (Welsh et al., 2010). For example, the presence of vasoactive intestinal polypeptide (VIP) and its G-protein coupled receptor, VPAC2, are important for maintaining circadian rhythmicity of gene expression in SCN cells, and for normal expression of rhythmic behavior in mice. Disrupting the genes for VIP (Vip−/−) (Colwell et al., 2003) or its receptor VPAC2 (Vipr2−/−) (Harmar et al., 2002; Cutler et al., 2003), leads to severely compromised circadian rhythms in behavior, neuronal firing, and gene expression, owing to intercellular desynchronization among SCN neurons (Table 1) (Aton et al., 2005; Maywood et al., 2006; Brown et al., 2007; Hughes et al., 2008). In normal mice, circadian rhythms generated within the SCN network are much more robust than those produced by individual neurons (Yamaguchi et al., 2003), even in the presence of clock gene mutations (Liu et al., 2007b). Indeed, recent work has shown that when the autonomous circadian oscillation of individual SCN neurons is eliminated by core clock gene mutation, molecular noise and intercellular coupling are sufficient to elicit stochastic, quasi-circadian oscillations as an emergent property of the SCN network (Ko et al., 2010).

Photic input to the SCN via the RHT is transduced into neural and humoral output signals that synchronize other rhythms in the body, including those of temperature, hormone secretion, and rest/wake (Aston-Jones et al., 2001; Buijs and Kalsbeek, 2001; Brown et al., 2002). Synchronizing signals reach peripheral tissues by both autonomic neural connections (Buijs and Kalsbeek, 2001; Vujovic et al., 2008), and through the release of hormones such as glucocorticoids (Balsalobre et al., 2000; Le Minh et al., 2001). In the absence of the SCN, circadian rhythms in most peripheral tissues damp out after a few days from desynchronization among the cells in the tissue, yet at the single cell level circadian rhythms persist (Balsalobre et al., 1998; Yamazaki et al., 2000; Nagoshi et al., 2004; Welsh et al., 2004; Yoo et al., 2004). The circadian rhythm of locomotor activity commonly monitored in mice and other rodents to determine endogenous circadian period relies on diffusible signals released from the SCN (Silver et al., 1996), several candidate molecules for which have been identified including transforming growth factor α (TGFα) (Kramer et al., 2001; Kramer et al., 2005), cardiotrophin-like cytokine (Kraves and Weitz, 2006), prokineticin 2 (PK2) (Cheng et al., 2002; Li et al., 2006) and potentially others (Hatcher et al., 2008).

It is important to note that there are oscillators in some mammalian brain regions and tissues that, in the absence of the SCN, can drive local physiological rhythms. Two such well-characterized regions include the retina (Tosini and Menaker, 1996) and the olfactory bulb (Granados-Fuentes et al., 2004a; Granados-Fuentes et al., 2004b; Granados-Fuentes et al., 2006). Further, two extra-SCN pacemakers, the food-entrainable oscillator (FEO) and the methamphetamine-sensitive circadian oscillator (MASCO), can drive circadian behavioral and endocrine rhythms in the absence of the SCN or functional canonical clock genes (Honma et al., 2008; Honma and Honma, 2009; Mohawk et al., 2009; Storch and Weitz, 2009; Pezuk et al., 2010).

IV. THE MAMMALIAN CIRCADIAN MOLECULAR OSCILLATOR

The mammalian circadian molecular oscillator model proposed following the discovery of the core clock genes described earlier encompasses our current understanding of the circadian control of gene expression in cells throughout the body. Core circadian clock genes are genes whose protein products are necessary components for the generation and regulation of circadian rhythms; that is, proteins which form the primary molecular circadian oscillatory mechanism within individual cells. In this model, positive and negative core clock components form a feedback loop with a time constant of about 24 hr per cycle (Figure 1). This loop begins during the day when two bHLH-PAS transcription factors, CLOCK and BMAL1 heterodimerize, translocate to the nucleus, and initiate transcription from genes containing E-box (5′-CACGTG-3′) or E′-box (5′-CACGTT-3′) cis-regulatory elements, including the Per and Cry genes (King et al., 1997b; Gekakis et al., 1998; Kume et al., 1999; Bunger et al., 2000; Zheng et al., 2001; Yoo et al., 2005). PER and CRY proteins heterodimerize and, along with other proteins such as CK1ε, form a complex in the cytoplasm that translocates to the nucleus where they accumulate and subsequently represses transcription of their own (and other) genes by directly inhibiting CLOCK/BMAL1 (Griffin et al., 1999; Kume et al., 1999; Lee et al., 2001; Sato et al., 2006). Thus, the CLOCK/BMAL1 heterodimer forms the positive, or transactivating component in this loop, while the PER/CRY complex acts as the negative, or transinhibiting component (Figure 1). Following several posttranscriptional and posttranslational steps discussed later, the PER/CRY complex is targeted for degradation via the proteasomal pathway, thereby relieving inhibition such that CLOCK/BMAL1 can initiate a new cycle of transcription. This relatively straightforward feedback loop forms what has become known as the mammalian “core” oscillator mechanism.

Figure 1.

Figure 1

Model of the mammalian cell-autonomous oscillator as described in the text. Abbreviations: CCG, clock-controlled gene; P, phosphate; U, ubiquitin.

The general molecular mechanism just described governs circadian output rhythms in all cells throughout the body, although there are tissue-specific differences. For example, in the forebrain, neuronal PAS domain protein 2 (NPAS2) appears to be the more relevant BMAL1 partner (Reick et al., 2001). Thus, the CLOCK(NPAS2)/BMAL1 complex initiates the rhythmic transcription of clock-controlled genes in tissues throughout the body. Microarray studies have shown that approximately 10–15% of all mammalian transcripts exhibit a circadian oscillation from one cell type/tissue to another (Akhtar et al., 2002; Duffield et al., 2002; Panda et al., 2002a; Storch et al., 2002; Oishi et al., 2003; Miller et al., 2007). These studies, however, may underrepresent the true number of genes under circadian control. Powerful statistical tests are required to identify cycling transcripts from noisy microarray data sets (Doherty and Kay, 2010). Development of new nonparametric statistical algorithms promises to provide more accurate measurements of period, phase and amplitude than traditional analysis methods (Hughes et al., 2010). Hence, continued improvements in data analysis methods should allow the identification of rhythmic transcripts in noisy, low-amplitude data and provide a more precise estimate of the number of genes under circadian control in various mammalian tissues.

Following the discovery of several other bona fide clock genes, it soon became evident that accessory regulatory loops interconnect with the core loop just described and add not only robustness and stability to the clock mechanism, but also provide additional layers of control and link to a myriad of other pathways in the cell. The first of these accessory loops involves members of the large nuclear receptor family. The mouse Bmal1 promoter contains two cognate RevErbA/ROR-binding elements (ROREs) via which the nuclear receptors RORα (retinoic acid receptor-related orphan receptor α), RORβ, or RORγ activate (Sato et al., 2004; Akashi and Takumi, 2005; Guillaumond et al., 2005), and REV-ERBα (reverse orientation c-erbA α) or REV-ERBβ repress (Preitner et al., 2002; Triqueneaux et al., 2004; Guillaumond et al., 2005), Bmal1 expression, although the REV-ERBs are likely more important in this process (Figure 1) (Liu et al., 2008). Interestingly, the expression of the aforementioned nuclear receptors is circadian and relies on CLOCK/BMAL1-mediated activation through E-boxes in their promoters, although the Ror and Rev-erb transcripts cycle antiphase to each other (Yang et al., 2006). The RORα-mediated activation of Bmal1 transcription is enhanced by peroxisome proliferator-activated receptor-γ (PPARγ) coactivator 1 α (PGC1α), the expression of which cycles in liver and muscle (Liu et al., 2007c). Members of the PAR bZIP transcription factor family, including the activators DBP (D-box binding protein), TEF (thyrotroph embryonic factor), HLF (hepatic leukemia factor), and the repressor E4BP4 (E4 promoter-binding protein 4), act via D-box elements in target genes to form a second accessory feedback loop (Mitsui et al., 2001; Ueda et al., 2005; Ohno et al., 2007).

V. BEHAVIORAL, MOLECULAR, AND CELL/TISSUE EFFECTS OF CIRCADIAN CLOCK GENE MUTATIONS

Naturally-occurring, chemically-induced, or targeted mutations exist for all of the core clock genes (Table 1). These mutations have helped define the role of each component of the molecular oscillator (Lowrey and Takahashi, 2004; Ko and Takahashi, 2006; Takahashi et al., 2008). At times, however, results from disruption of clock components have been unexpected.

A. Behavioral and molecular effects

One of these surprises occurred with the generation of a mouse Clock knockout model. Interestingly, unlike ClockΔ19 animals, Clock−/− mice continue to express circadian rhythms of locomotor activity in DD, albeit with a slightly shorter period compared to wild-type animals, and experience only modest alterations in circadian gene expression in the SCN (Table 1) (Debruyne et al., 2006). Subsequent work has revealed that in the SCN, NPAS2 can compensate for CLOCK by heterodimerizing with BMAL1 to activate transcription from E-box-containing target genes (DeBruyne et al., 2007a), but that molecular circadian rhythms in peripheral tissues are dependent on the presence of CLOCK (DeBruyne et al., 2007b). Npas2−/− animals, however, experience only subtle changes in circadian locomotor activity and gene expression (Dudley et al., 2003; DeBruyne et al., 2007a), suggesting that CLOCK has a more important role in the molecular oscillator. Hence, although there is partial functional redundancy between CLOCK and NPAS2, it is clearly tissue specific (Reick et al., 2001; DeBruyne et al., 2007b). As expected, CLOCK/NPAS2 double knockout animals are completely arrhythmic in DD (DeBruyne et al., 2007a).

Knockout of Bmal1 in mice results in behavioral arrhythmicity in DD and disrupted molecular rhythms of gene expression even though its paralog, BMAL2 (MOP9), is also expressed in the SCN and can form a transcriptionally-competent complex with CLOCK (Table 1) (Bunger et al., 2000; Hogenesch et al., 2000; Dardente et al., 2007). Indeed, Bmal1 is the only core clock gene for which loss of function causes an immediate loss of circadian locomotor behavior in DD. Constitutive expression of Bmal1 in Bmal1−/− mice or cells restores circadian rhythmicity (McDearmon et al., 2006; Liu et al., 2008), demonstrating that cycling Bmal1 mRNA is not necessary for circadian rhythm generation. That BMAL2 cannot rescue the Bmal1−/− phenotype most likely results from the dependence of Bmal2 expression on CLOCK/BMAL1-mediated activation. Hence, disrupting Bmal1 is likely functionally equivalent to creating a double Bmal1/Bmal2 null animal (Shi et al., 2010).

Per1 null mutations have been independently generated by three groups (Table 1). The mutant progeny from these lines exhibit subtle differences in circadian behavior. Homozygous Per1ldc mice have about a 0.5 hr shorter free-running period in DD than wild-type controls and experience a gradual loss of rhythmicity after two weeks in DD (Bae et al., 2001). Per1Brdm1 mice express a free-running period approximately 1 hr shorter than wild-type animals and maintain rhythmicity (Zheng et al., 2001). This result is consistent with the circadian behavior of the Per1−/− mutant line generated by a third group which exhibits a 0.7 hr shorter free-running period with no loss of rhythmicity (Cermakian et al., 2001). The phenotypic disparities observed among the three Per1 null studies may result from differences in targeting approaches or genetic backgrounds. Two independent null mutations in Per2 have been reported (Table 1). Both mutant lines exhibit a loss of behavioral rhythmicity in DD, yet the Per2Brdm1 line expresses a 1.5 hr shorter period for several days before experiencing arrhythmicity (Zheng et al., 1999). Most animals of the Per2ldc line exhibit immediate arrhythmicity upon exposure to DD (Bae et al., 2001). As expected from the results just presented, double Per1/Per2 knockout animals experience behavioral and molecular arrhythmicity in constant conditions (Bae et al., 2001; Zheng et al., 2001). Because loss of Per3 has no effect on circadian rhythms either in Per1/Per3 or Per2/Per3 double mutant mice, Per3 is not an essential component of the circadian core clock mechanism (Shearman et al., 2000a; Bae et al., 2001).

Targeted disruption of either of the two Cry genes results in opposite effects on circadian behavior—Cry1−/− animals have a 1 hr shorter and Cry2−/− animals have a 1 hr longer free-running period in DD compared to wild-type animals (Thresher et al., 1998; van der Horst et al., 1999; Vitaterna et al., 1999) (Table 1). Similar to Per1/Per2 double knockouts, Cry1/Cry2 double knockout animals experience a complete loss of behavioral and molecular rhythmicity when transferred to DD (Thresher et al., 1998; Okamura et al., 1999; van der Horst et al., 1999; Vitaterna et al., 1999). Per/Cry compound knockouts also exhibit interesting behavioral phenotypes (Table 1). Per1Brdm1/Cry1−/− mice have normal circadian rhythms of behavior and gene expression (Oster et al., 2003). Deletion of Per1 rescues the short period phenotype observed in Cry1−/− mutants, revealing that Per1 is a nonallelic suppressor of Cry1. Per1Brdm1/Cry2−/− mice have more complex phenotypes. Mutants up to six months of age express behavioral rhythms 1.5 hr longer than wild-type controls and have normal rhythms of Per2 expression. After approximately 6 months of age, Per1Brdm1/Cry2−/− animals exhibit disrupted entrainment to LD cycles, and subsequently experience arrhythmicity upon release into DD (Oster et al., 2003). In addition, the older animals have blunted Per2 rhythms in the SCN revealing an age-sensitive effect in this compound mutant. Per2Brdm1/Cry1−/− mutants experience immediate behavioral and molecular arrhythmicity in DD (Oster et al., 2002). Per2Brdm1/Cry2−/− animals, however, maintain behavioral and molecular circadian rhythmicity in DD with a slightly shorter free-running period compared to wild-type controls. Thus, inactivation of Cry2 in Per2 null animals restores circadian rhythmicity. As a result, Cry2 is a nonallelic suppressor of Per2 (Oster et al., 2002).

The double Per1/Per2 and Cry1/Cry2 knockout results make sense given that the PER/CRY complex is necessary to directly inhibit CLOCK/BMAL1-mediated transcription (Griffin et al., 1999; Kume et al., 1999; Shearman et al., 2000b; Lee et al., 2001; Sato et al., 2006). Until recently, transient transfection assays pointed to a more prominent role for CRY in inhibiting CLOCK/BMAL1 (Dardente et al., 2007), yet new evidence suggests that PER may be the more important of the negative effectors as its constitutive expression disrupts the circadian clock in fibroblasts and hepatocytes. Furthermore, constitutive PER2 expression in the SCN of transgenic mice results in the loss of circadian rhythms of locomotor behavior in a conditional and reversible manner (Chen et al., 2009). Finally, biochemical evidence demonstrates that PER2 directly binds to the CLOCK/BMAL1 complex in a rhythmic way, and that it brings CRY into contact with CLOCK/BMAL1. Rhythmic expression of PER in turn drives the rhythmic inhibition of CLOCK/BMAL1, and it is PER that is the rate-limiting component of the inhibitor complex (Chen et al., 2009). This is substantiated by independent work demonstrating that PER2 is also a more potent inhibitor of CLOCK/BMAL2-mediated transactivation than is CRY (Sasaki et al., 2009).

B. Cell/tissue effects

The analysis of behavioral (locomotor activity) and molecular (RNA/protein) rhythms in mice with mutations in core circadian clock genes just described is insufficient to provide a comprehensive view of molecular clock function. For example, most of the above studies do not take into account differences in central versus peripheral oscillators, potential intercellular interactions in producing the observed phenotypes, or reveal unique properties of individual cellular oscillators. New methods allowing continuous monitoring of circadian rhythms in cultured tissues and individual cells in real time for periods of 20 days or more via bioluminescent technology have revealed many clock properties not evident from behavioral and molecular analyses alone.

By crossing clock gene knockout mice to the Per2Luciferase (Per2Luc) knockin reporter mouse line in which a luciferase gene is fused to the 3'-end of the endogenous Per2 gene (Yoo et al., 2004), one group has measured the effects of clock gene perturbations at the level of tissues, populations of cultured cells, and single dissociated cells from both the SCN and peripheral tissues (Liu et al., 2007b). In SCN tissue explants, disruption of Per1, Per3, Cry1 or Cry2 individually has no effect on the maintenance of circadian rhythmicity, and the observed period for each mutant SCN tissue reflects the free-running behavioral period of the corresponding mutant animal model (Table 1 and Table 2). In peripheral tissue explants (e.g., liver, lung, cornea), unlike SCN explants, Cry1−/− and Per1−/− are required for robust, sustained circadian rhythmicity (Table 2), a property of peripheral tissues not evident from the previously described behavioral and molecular studies (Table 1). Cry2−/− and Per3−/− mutant peripheral tissues maintain rhythmicity with slightly longer and shorter periods, respectively, compared to wild-type controls, again consistent with behavioral results (Table 1). In dissociated fibroblast cells in culture, Per1, Per2 and Cry1 are required to maintain circadian oscillations. Thus it seems that, in fibroblast cultures at least, Per1 and Per2 are not functionally redundant (Liu et al., 2007b).

Table 2.

Cell and tissue phenotypes of mutations in mouse clock and clock-related genes

Gene(s) Mutant allele(s) Cellular phenotype(s) Tissue phenotype(s) References
Clock Clock−/− SCN: WT; lung, liver: AR (DeBruyne et al., 2007a;
DeBruyne et al., 2007b)
Npas2 (Mop4) Npas2−/− SCN, lung, liver: WT (DeBruyne et al., 2007b)
Bmal1 (Arntl, Mop3) Bmal1−/− Fibroblasts, SCN neurons: AR SCN: variable/stochastic; pituitary,
liver, lung, cornea: AR
(Liu et al., 2008; Ko etal., 2010)
Cry1 Cry1−/− Fibroblasts, SCN neurons: AR SCN: short; lung, liver, cornea: AR (Liu et al., 2007b)
Cry2 Cry2−/− Fibroblasts, SCN neurons: long SCN, lung, liver, cornea: long (Liu et al., 2007b)
Cry1/Cry2 Cry1−/−/Cry2−/− Fibroblasts: AR SCN, lung, liver, cornea: AR (Yagita et al., 2001; Liu et al., 2007b)
Per1 Per1ldc Fibroblasts, SCN neurons: AR SCN: WT; lung: AR (Liu et al., 2007b)
Per2 Per2ldc Fibroblasts: AR (Liu et al., 2007b)
Per3 Per3−/− Fibroblasts: short SCN, lung: short (Liu et al., 2007b)
Rev-erbα (Nr1d1) Rev-erbα−/− Fibroblasts: WT (Liu et al., 2008)
Rorα (Rora) Rorα−/− (staggerer) Fibroblasts: WT (Liu et al., 2008)
Rorγ (Rorc) Rorγ−/− Fibroblasts: WT Lung, liver: WT (Liu et al., 2008)
CK1δ (Csnk1d) CK1δ−/− Fibroblasts: long *SCN, liver: long (Etchegaray et al., 2009;
Lee et al., 2009;
Etchegaray et al., 2010)
CK1ε (Csnk1e) CK1ε−/− Fibroblasts: WT *SCN, liver: WT (Etchegaray et al., 2009;
Etchegaray et al., 2010)
CK1εtau Fibroblasts: short SCN, pituitary, lung, kidney: short (Meng et al., 2008)

Gene symbols are as in Table 1. Abbreviations: AR, arrhythmic; WT, wild type;

*

neonatal tissue. Table modified from (Baggs et al., 2009).

When single fibroblast cells are imaged for circadian rhythms of bioluminescence, again Per1 and Cry1 prove necessary to sustain circadian oscillations, confirming the results observed in fibroblast cultures (Liu et al., 2007b). Single Cry2−/− fibroblast cells are rhythmic with a slightly longer period compared to individual wild-type cells, consistent with the behavioral phenotype of Cry2 null mice (Table 1 and Table 2). To measure rhythms of bioluminescence from single SCN neurons, they must first be uncoupled by mechanical dissociation into single cells (Welsh et al., 1995; Herzog et al., 1998). Similar to the result in single fibroblasts, single Cry2−/− SCN neurons are rhythmic with a period longer than wild-type SCN neurons (Table 2). Single Cry1−/− and Per1−/− SCN neurons, in contrast to single Cry2−/− SCN neurons or Cry1−/− and Per1−/− SCN tissue explants, exhibit arrhythmicity (Table 2). This is an important result not apparent in earlier behavioral studies of Per1 and Cry1 null mutants and demonstrates that the robustness of circadian oscillations observed in Cry1−/− and Per1−/− SCN explant tissue is not a cell-autonomous property. Instead, the ability of Cry1−/− and Per1−/− SCN tissue to maintain circadian rhythmicity despite mutations in core clock components that, at the single-cell level cause arrhythmicity, depends on intercellular coupling among SCN neurons (Liu et al., 2007b). This property of intercellular coupling, and not unique intracellular molecular mechanisms, is what distinguishes SCN neurons from cells of peripheral tissues.

As mentioned previously, Bmal1 null mutant animals experience an immediate loss of circadian behavior upon transfer to constant conditions (Bunger et al., 2000). Using bioluminescence monitoring methods with Bmal1−/− SCN and peripheral tissues similar to those just described, a recent study has elucidated interesting properties of the SCN network not discovered in previous behavioral and molecular investigations (Ko et al., 2010). SCN explants from Bmal1−/− mice crossed to the Per2Luc reporter line exhibit rhythmic but highly variable (noisy) oscillations of PER2::LUC bioluminescence for more than 35 days in culture (Table 2). This is an unexpected result given that the behavioral and molecular rhythms of gene expression in Bmal1−/− animals are arrhythmic (Table 1). The authors of this study refer to these quasi-circadian rhythms generated by the Bmal1−/− SCN explants as stochastic. As expected, bioluminescence monitoring demonstrates that all peripheral tissues from Bmal1−/− animals are arrhythmic (Table 2). Analysis of single dispersed Bmal1−/− SCN neurons, however, reveals that they are arrhythmic and do not exhibit the stochastic rhythms of bioluminescence observed in Bmal1−/− SCN explants. This result seems not to depend from what subtype of SCN neuron recordings are made or from what region of the SCN the neurons are obtained—all dispersed Bmal1−/− SCN neurons exhibit arrhythmicity. Further, Bmal1−/− SCN slices treated with tetrodotoxin (TTX) experience an immediate cessation of stochastic rhythmicity, a result of loss of rhythmicity at the single-cell level. Upon removal of TTX from SCN slices, stochastic rhythmicity is restored thereby confirming the importance of intercellular coupling in generating the observed PER2::LUC rhythms in the SCN slices. Taken together, these bioluminescence results demonstrate that the quasi-circadian rhythms in Bmal1−/− SCN explants is not a cell-autonomous property, but rather an emergent rhythmic property of the SCN intercellular network (Ko et al., 2010).

VI. POSTTRANSLATIONAL MODIFICATION OF CLOCK PROTEINS

Posttranslational modifications of the core clock components play a crucial role in generating the delays necessary to establish the ~24 hr rhythm of the mammalian circadian clock. Some of these modifications are absolutely essential to clock function while others simply fine-tune the rhythm. Phosphorylation of clock proteins was the first posttranslational process observed in the mammalian molecular clock, and we understand more about this mechanism than any other. The list of identified posttranslational modifications of mammalian clock proteins has grown rapidly and now, in addition to phosphorylation, includes dephosphorylation, ubiquitination, sumoylation, and acetylation.

A. Phosphorylation

1. Casein kinase 1 (CK1)

Posttranslational modification as an important clock-related process in higher eukaryotes became apparent with the identification in Drosophila of doubletime, a gene encoding a fly casein kinase 1 ortholog that phosphorylates PER (Kloss et al., 1998; Price et al., 1998). Subsequently, we identified casein kinase 1 epsilon (CK1ε) as the gene affected by the Syrian hamster tau mutation, and showed that in vitro CK1εtau is hypomorphic toward various substrates, including mammalian PER1 and PER2 proteins (Lowrey et al., 2000). Others demonstrated that, in vitro and in cultured cells, CK1ε and the closely-related family member, CK1δ, can phosphorylate PER (Keesler et al., 2000; Vielhaber et al., 2000; Camacho et al., 2001; Akashi et al., 2002). Further work revealed that CK1δ/ε-mediated phosphorylation regulates PER subcellular localization and its ability to repress CLOCK/BMAL1-mediated transcription, and promotes its ubiquitin-degradation via the 26S proteasome (Vielhaber et al., 2000; Eide et al., 2005b; Shirogane et al., 2005; Vanselow et al., 2006; Ohsaki et al., 2008).

Although our work showed that CK1εtau was a hypomorph in in vitro assays, a study using mathematical modeling and in vivo analyses has reported that the CK1εtau anion binding site mutation causes loss of enzyme function toward canonical acidic CK1ε substrates, but gain of function toward the non-canonical β-transducin repeat-containing protein (βTrCP) binding site on PER2 (Gallego et al., 2006a). Indeed, biochemical evidence substantiates the model's prediction as do behavioral, neurophysiological and cellular studies from a mouse model of the hamster tau mutation (Gallego et al., 2006a; Meng et al., 2008). Another group, however, has published findings that contradict this interpretation. They report that CK1εtau is actually a partial loss of function mutation as the mutant kinase is unable to phosphorylate sites that promote nuclear localization of PER2, but that it can phosphorylate amino acids required for PER2 proteasomal degradation (Vanselow et al., 2006). By mapping all of the CK1ε phosphorylation sites on PER2, they opine that the different sites differentially target PER2 to two cellular locations—nucleus or proteasome. Both interpretations agree, however, that the tau allele is a particularly interesting mutation biochemically as it differentially affects CK1ε activity toward specific substrates.

The tau mutation focused much attention on the role of CK1ε in the mammalian molecular clock, yet as mentioned above, CK1δ also phosphorylates PER proteins and targets them for degradation (Camacho et al., 2001; Xu et al., 2005), and both kinases associate with PER/CRY repressor complexes in vivo (Figure 1) (Lee et al., 2001). Thus, to better define the role of these two CK1 enzymes, null mutants of both have been generated independently by different laboratories. The free-running period of the locomotor activity rhythm of CK1ε−/− mice is slightly, but significantly, longer than wild-type controls (Table 1) (Meng et al., 2008; Etchegaray et al., 2009). Two groups have reported that CK1δ−/− mice die during the perinatal period, thus the free-running behavioral period has been studied in CK1δ+/− heterozygous animals. In one case, one copy of the CK1δ null allele results in no difference in free-running period compared to controls (Xu et al., 2005), while another group's heterozygous null animal exhibits a slight increase in circadian period (Etchegaray et al., 2009). In addition, compared to CK1ε-deficient tissue, mouse embryonic fibroblasts (MEFs) and liver tissue deficient in CK1δ have about a one to two hour longer circadian period in vitro (Etchegaray et al., 2009; Lee et al., 2009). When monitored from neonatal SCN explants, PER2::LUC bioluminescence rhythms from CK1δ−/− mice are longer compared to wild-type controls, yet there is no significant difference in PER2::LUC rhythms from CK1ε−/− SCN compared to controls (Etchegaray et al., 2010).

Pharmacological approaches have also been used to study the roles of CK1ε and CK1δ in the mammalian clock with the general CK1δ/ε inhibitors CKI-7, IC261 and D4476, all of which lengthen circadian period in cultured cells (Eide et al., 2005a; Vanselow et al., 2006; Reischl et al., 2007; Hirota et al., 2008). An inhibitor specific for CK1ε (PF-4800567) has only a slight effect on the period of oscillating rat-1 fibroblasts stably transfected with a Per2::luc reporter compared to the dual CK1δ/ε inhibitor (PF-670462) which causes an increase in fibroblast circadian period (Walton et al., 2009). Single injections into rats of the dual CK1δ/ε inhibitor induce large phase delays in circadian locomotor rhythms under free-running and entrained conditions (Badura et al., 2007; Sprouse et al., 2010). Daily treatment with PF-670462 significantly lengthens locomotor behavioral rhythms in a dose-dependent manner in wild-type, CK1εtau, and CK1ε−/− mice (Meng et al., 2010). Selective inhibition of CK1ε with PF-4800567 has no significant effect on behavioral rhythms in wild-type or CK1ε−/− mice, yet it lengthens the free-running locomotor activity rhythm of CK1εtau animals. How does inhibition of the CK1 enzymes affect molecular clock function? PF-670462 seems to work by stabilizing PER2 nuclear localization in SCN neurons and peripheral tissues. This prolongs PER2-mediated negative feedback, thereby lengthening circadian period (Meng et al., 2010). Together with the CK1 knockout experiments, these results suggest that CK1δ has a more prominent role compared to CK1ε in the mammalian clockwork; that is, the two kinases seem not to be equally redundant. It is clear, however, that loss of both CK1ε and CK1δ causes arrhythmicity in MEFs (Lee et al., 2009), and that knockdown of both kinases in human U2OS (osteosarcoma) cells additively lengthens circadian period to more than 30 hr (Isojima et al., 2009). It remains to be determined if this partial functional redundancy derives from unequal expression levels of the two kinases in cells throughout the body. Some evidence suggests that, at least in MEFs, CK1δ is twice as abundant as CK1ε (Lee et al., 2009).

One group has reported a surprising result in their work with CK1δ/ε in which a novel, non-catalytic clock-related role for these kinases is revealed (Lee et al., 2009). Overexpression of the CK1δ/ε-binding domain of PER2 (CKBD-P2) in MEFs severely disrupts PER2::LUC rhythms of bioluminescence, presumably because the CKBD-P2 fragment interferes (competes) with the interaction between CK1δ/ε and PER. In addition, PER1 and PER2::LUC levels are lower in the CKBD-P2-expressing MEFs, while CK1δ/ε levels are higher than normal. This suggests that the CKBD-P2 enhances the stability of CK1δ/ε via physical interaction. Furthermore, the low levels of PER observed in the CKBD-P2-expressing MEFs may result from the inhibition of CK1δ/ε-specific phosphorylation of PER, and from the reduced physical interaction between CK1δ/ε and PER which, under normal circumstances, may confer stability to PER (Lee et al., 2009). Overexpression of the CKBD region of PER3 in MEFs does not have an effect on the circadian rhythms in these cells, mirroring previous reports showing that PER3 does not interact with CK1δ/ε. That the stabilizing role of CK1δ/ε toward PER is non-catalytic is supported by experiments in which dominant-negative CK1δ/ε (K38R)-expressing MEFs do not experience reduced PER levels (Lee et al., 2009). Interestingly, a non-catalytic circadian role has also been reported recently for the Drosophila CK1 family member, DBT (Yu et al., 2009).

Finally, in a high-throughput screen of approximately 120,000 compounds using U2OS cells expressing luciferase under the control of the mouse Bmal1 promoter, another CK1 family member, CK1α, has been identified as a mammalian clock regulatory kinase (Hirota et al., 2010). A purine derivative identified in this screen, longdaysin, inhibits CK1δ, CK1α, and ERK2 (MAPK1) and prevents them from phosphorylating PER1, causing a dramatic 13 hr lengthening of period in U2OS cells. Knockdown by siRNA of one of the aforementioned kinases alone is insufficient to recapitulate the 13 hr period lengthening effect, yet combinatorial knockdown of all three kinases additively increases period and closely mirrors the effect of longdaysin treatment (Hirota et al., 2010). Results from this interesting study suggest that multiple kinases participate in a network to enhance robustness of the molecular clock mechanism.

2. Glycogen synthase kinase-3β (GSK-3β)

Glycogen synthase kinase-3 (GSK-3) is a serine-threonine, phosphate-directed protein kinase of which there are two isoforms in mammals: GSK-3α and GSK-3β (Ali et al., 2001). GSK-3 was initially characterized as a kinase involved in metabolism and energy storage, yet it has since been shown to play a role in many intracellular pathways (Doble and Woodgett, 2003). Knockout models for both isoforms have been generated, but Gsk-3β−/− mice experience embryonic lethality (Hoeflich et al., 2000; MacAulay et al., 2007). Interestingly, GSK-3 is sensitive to lithium. Presumably, Li+ competes directly for binding to GSK-3 with Mg2+, a required cofactor for GSK-3 function (Klein and Melton, 1996; Stambolic et al., 1996; Ryves and Harwood, 2001). Several studies have documented the effects of lithium treatment on circadian rhythms in mammals, including a consistent effect of lengthening the free-running period of behavioral rhythms, notably those of locomotor activity and drinking (Seggie et al., 1982; LeSauter and Silver, 1993; Iwahana et al., 2004). This period-lengthening effect of lithium is also observed for the firing rate rhythms in isolated mouse SCN neurons (Abe et al., 2000). Other work has shown that GSK-3β is rhythmically expressed in the SCN and liver of mice, and that it undergoes a daily cycle in phosphorylation in vivo as well as in serum-shocked NIH3T3 mouse fibroblasts (Harada et al., 2005; Iitaka et al., 2005). Lithium chloride treatment phase delays, while overexpression of GSK-3β, phase advances clock gene expression in fibroblasts (Iitaka et al., 2005). In addition, Gsk-3α RNAi knockdown in Gsk-3β−/− MEFs, induces a phase delay in the Per2 RNA rhythm, as does treatment of MEFs with kenpaullone, a GSK-3 antagonist (Kaladchibachi et al., 2007). Surprisingly, however, a recent high-throughput screen has identified small molecule inhibitors of GSK-3β that shorten circadian period, a result confirmed by siRNA knockdown of GSK-3β (Hirota et al., 2008). These effects of inhibition of GSK-3β on the molecular clock in cells and on clock-controlled behavior in mammals have prompted further investigation into the potential clock-related targets of this enzyme. In addition, work in Drosophila has shown that shaggy, the fly ortholog of mammalian GSK-3β, is an important component in determining circadian period length in that organism (Martinek et al., 2001).

Mammalian targets of GSK-3β phosphorylation include the positive and negative components of the core circadian transcriptional/translational feedback loop, CLOCK/BMAL1 and PER/CRY, respectively. Phosphorylation by GSK-3β of PER2 promotes its nuclear localization (Iitaka et al., 2005). GSK-3β-mediated phosphorylation at Ser553 of CRY2 leads to its degradation by the proteasome (Harada et al., 2005), yet GSK-3β phosphorylation promotes the stabilization of REV-ERBα (Yin et al., 2006). Lithium treatment of cultured cells results in the rapid proteasomal degradation of REV-ERBα and concomitant derepression of Bmal1 transcription owing to inhibition of GSK-3β (Yin et al., 2006). Others have identified a phosho-degron region on the CLOCK protein that is phosphorylated by GSK-3β, and which promotes degradation of CLOCK (Spengler et al., 2009). Finally, BMAL1, the major CLOCK dimerization partner, is phosphorylated by GSK-3β on Ser17 and Thr21. This modification targets BMAL1 for ubiquitination and subsequent degradation by the proteasome (Sahar et al., 2010).

3. Casein kinase 2 (CK2)

Casein kinase 2, a serine/threonine kinase, is a tetramer composed of two α catalytic and two β regulatory subunits, and, similar to CK1 prefers acidic substrates (Meggio and Pinna, 2003). A role for this kinase in the regulation of circadian rhythms was first reported in Arabidopsis, Drosophila, and Neurospora (Gallego and Virshup, 2007). Recently, participation of CK2 in the mammalian molecular clock mechanism has been reported by three groups. First, BMAL1 is a substrate for CK2α at Ser90, a residue that undergoes rhythmic phosphorylation (Tamaru et al., 2009). Nuclear localization of BMAL1 seems to depend on CK2α phosphorylation, as knockdown of CK2α results in cytoplasmic BMAL1 accumulation and a concomitant disruption of Per2 mRNA rhythms (Tamaru et al., 2009).

PER2 is also a CK2α substrate. One study has shown that CK2α both phosphorylates PER2 at Ser53 and enhances CK1ε-mediated PER2 destabilization (Tsuchiya et al., 2009). These two actions of CK2α on PER2 are independent, as the CK2α-mediated potentiation of CK1ε-dependent degradation of PER2 does not require phosphorylation of Ser53. Although it has not been demonstrated, it may be that CK2α acts to phosphorylate CK1ε thereby upregulating its activity toward PER2, given that a catalytically inactive form of CK2α (CK2α-K68A) fails to enhance CK1ε-dependent PER2 degradation (Tsuchiya et al., 2009). Finally, in a recent RNAi screen for clock-related components, downregulation of either CK2α or CK2β lengthens circadian period, while knockdown of both subunits leads to arrhythmicity (Maier et al., 2009). Furthermore, overexpression of CK2α has a period-shortening effect. This same study showed that CK2α binds PER2 and promotes phosphorylation near the N-terminus, although the proposed sites do not correspond to Ser53 mentioned before. Also, unlike the previous study, CK2α phosphorylation is proposed to stabilize PER2 rather than enhance its degradation. Inhibition of CK2α was shown to delay PER2 nuclear accumulation, suggesting that CK2α phosphorylation of PER2 may provide a signal for nuclear entry (Maier et al., 2009). These, and other questions, need to be addressed to better understand the role of CK2 in the mammalian clock mechanism.

4. Other kinases

BMAL1 has been identified as a substrate for several kinases including CK1ε and GSK-3β as described above. In addition, mitogen-activated protein kinase (MAPK) phosphorylates BMAL1 on several residues, including Thr534 which negatively regulates the transactivation potential of the CLOCK/BMAL1 complex at E-boxes (Sanada et al., 2002). Another study has shown recently that RACK1 (receptor for activated C kinase-1) recruits activated protein kinase Cα (PKCα) to the CLOCK/BMAL1 complex during the negative feedback phase of the circadian cycle in the nucleus where it phosphorylates BMAL1 and suppresses CLOCK/BMAL1 transcriptional activity (Robles et al., 2010). Knockdown of either RACK1 or PKCα in fibroblast cultures shortens circadian period. Prkca−/− mice have a normal circadian period, yet they experience impaired photic resetting of behavioral rhythms (Jakubcakova et al., 2007). This difference between the knockout phenotype and cell culture results highlights the need for further work.

CLOCK has been shown to be phosphorylated throughout the circadian cycle, yet it is hyperphosphorylated during the negative feedback phase (Yoshitane et al., 2009). Specific residues have been identified as phosphorylation sites on CLOCK, including Ser38, Ser42, and Ser427. The kinase or kinases responsible for phosphorylating these sites, however, remain undetermined. CLOCKΔ19, which lacks the binding site for CLOCK-interacting protein circadian (CIPC), a PER/CRY-independent negative regulator of CLOCK/BMAL1-mediated transcription, is less phosphorylated and more stable than wild-type CLOCK (Zhao et al., 2007). Others have shown that CLOCK is a substrate for phosphorylation by protein kinase G II (PKG-II) and by two protein kinase C isoforms (PKCα and PKCγ) (Tischkau et al., 2004; Shim et al., 2007).

Finally, it will be important to assess the contribution of sequential phosphorylation events on clock proteins. For example, some kinases such as CK1δ/ε require priming phosphorylation of their target sites (Knippschild et al., 2005). Phosphorylation of human PER2 at the Ser662 familial advanced sleep phase syndrome (FASPS) site occurs by an unidentified kinase and is necessary for subsequent CK1δ/ε phosphorylation (Xu et al., 2007). Similarly, GSK-3β phosphorylation of CRY2 requires priming phosphorylation by DYRK1A (dual-specificity tyrosine-phosphorylated and regulated kinase 1A) (Kurabayashi et al., 2010). In many cases, the priming kinases for these so-called “phosphate directed” kinases such as CK1δ/ε and GSK-3 remain to be identified for particular circadian substrates.

B. Dephosphorylation

Protein phosphatases, although fewer in number in the mammalian genome relative to kinases, also play an important role in the molecular clock (Gallego and Virshup, 2007). Reversible phosphorylation of clock-relevant substrates presumably confers on clock the flexibility to respond appropriately to various stimuli. Few studies, however, have explored the role of phosphatases on the mammalian clock mechanism. The major clock kinases, CK1ε and CK1δ, both undergo autophosphorylation which downregulates their activity. At least eight autophosphorylation sites must be dephosphorylated by phosphatases to activate CK1δ/ε (Gietzen and Virshup, 1999). Additional evidence suggests that CK1δ/ε participates in a futile autophosphorylation/dephosphorylation cycle in vivo which acts to regulate kinase activity (Rivers et al., 1998). One group has shown that it is protein phosphatase 5 (PP5) that dephosphorylates CK1δ/ε. Furthermore, the same study demonstrated that the CRY proteins interact with and inhibit noncompetitively, PP5. As a result, the CRY proteins seem to indirectly regulate the activity of CK1δ/ε by inhibiting the phosphatase activity of PP5 (Partch et al., 2006). Another study has found a role for PP1 in the clock mechanism (Gallego et al., 2006b). PP1 can bind to and dephosphorylate CK1δ/ε-phosphorylated PER2 thereby negatively regulating its degradation by the proteasome. Overexpression of a dominant negative form of PP1, or use of PP1 inhibitors, results in accelerated degradation of PER2 (Gallego et al., 2006b).

C. Ubiquitination

One of the major pathways for protein degradation in cells is the ubiquitin-dependent proteasomal mechanism. This system requires the attachment of multiple ubiquitin molecules to lysine residues on the target protein (Ciechanover et al., 2000; Nandi et al., 2006). Polyubiquitinated proteins are directed to the 26S proteasome, a large multicatalytic protease, where they are then degraded to small peptides (Nandi et al., 2006). Attachment of ubiquitin to a protein is a three-step process. First, ubiquitin is adenylated by an activating enzyme (E1), then transferred to a conjugating enzyme (E2), and finally linked to the target protein by a ligase (E3) (Wilkinson, 1999; Ciechanover et al., 2000). The specificity of this system is determined by the E3 ligases which can be categorized into at least six subtypes. With respect to the mammalian clock mechanism, the SCF E3 subtype is the most relevant. SCF complexes are multimers and are named for their constituent protein components, Skp1, Cdc53 or Cullin, and any one of a number of proteins containing an F-box motif, each of which recognizes a particular target protein (Nandi et al., 2006). It is the F-box protein that confers specificity to each SCF complex (Cardozo and Pagano, 2004). The SCF complexes are constitutively active enzymes that recognize and ubiquitinate only phosphorylated substrates (Kornitzer and Ciechanover, 2000). Consequently, this system links protein phosphorylation to proteolytic degradation by the 26S proteasome (Cardozo and Pagano, 2004).

Turnover of the PER/CRY repressor complex is an important event in relieving inhibition of CLOCK/BMAL1 such that a new circadian day can begin. Insight into how this repressor complex is cleared has come from several studies. Recently, our laboratory and another independently recovered long period behavioral mutants, Overtime (Siepka et al., 2007) and After-hours (Godinho et al., 2007), respectively, through ENU mutagenesis screens of mice. Both mutations affect the same gene, Fbxl3, which encodes the F-box and leucine-rich repeat protein 3 (FBXL3). FBXL3 is involved in ubiquitination of the CRY proteins, which targets them for degradation (Busino et al., 2007). The proteasomal-mediated degradation of CRY1 and CRY2, however, appears to be differentially regulated. CRY1 is phosphorylated by AMPK (AMP-activated protein kinase) which promotes its FBXL13-dependent degradation (Figure 1) (Lamia et al., 2009). CRY2, however, is phosphorylated by GSK-3β on Ser553 (Harada et al., 2005), which first requires priming phosphorylation by DYRK1A on Ser557, both of which are at the C-terminal tail of CRY2 (Kurabayashi et al., 2010). This Ser557/Ser553 phosphorylation mechanism promotes proteasomal degradation of CRY2 by what is likely an FBXL13-independent mechanism. Indeed, it seems that Ser557/Ser553 phosphorylation and subsequent turnover of CRY2 slows its cytosolic accumulation rate and allows its timely nuclear translocation (Kurabayashi et al., 2010). The FBXL13-dependent degradation mechanism acts during the declining phase of negative feedback when CRY2 nuclear clearance occurs (Godinho et al., 2007). Taken together, these two mechanisms of CRY2 degradation suggest a model by which negative feedback is controlled.

As discussed earlier, phosphorylation of the PER proteins by CK1δ/ε promotes their polyubiquitination (Eide et al., 2005a; Shirogane et al., 2005; Reischl et al., 2007; Ohsaki et al., 2008). Ubiquitination of the PERs is mediated by the F-box proteins βTrCP1 and/or βTrCP2 (Figure 1) (Vielhaber et al., 2000; Eide et al., 2005b; Shirogane et al., 2005; Vanselow et al., 2006; Ohsaki et al., 2008). Following ubiquitination, the PER proteins are degraded by the 26S proteasome (Gallego and Virshup, 2007).

D. Sumoylation

The BMAL1 protein undergoes extensive posttranslational modification in cells, including phosphorylation, acetylation, ubiquitination, and sumoylation. Small ubiquitin-like modifier (SUMO) proteins are covalently attached at lysine residues of target proteins to modify their function (Wilkinson and Henley, 2010). Sumoylation has been shown to affect nuclear/cytosolic localization of proteins, progression through the cell cycle, protein stability, and transcriptional regulation (Gareau and Lima, 2010). In mice and humans, there are three Sumo paralogs (Sumo1–3). SUMO2 and SUMO3 are 95% identical and are often referred to as SUMO2/3. Two groups have independently demonstrated that BMAL1 is rhythmically polysumoylated at a conserved lysine residue (K259) in the PAS domain linker region by all three SUMO proteins, and that this process is dependent on CLOCK, the BMAL1 dimerization partner (Cardone et al., 2005; Lee et al., 2008). Furthermore, sumoylation localizes BMAL1 to the promyelocytic leukemia nuclear body, potentiates CLOCK/BMAL1 transactivation of clock-controlled genes, and promotes BMAL1 ubiquitin-dependent proteasomal degradation (Lee et al., 2008).

E. Acetylation, deacetylation and chromatin remodeling

Rhythmic changes in chromatin architecture participate in the activation and repression of transcription via posttranslational modifications at histone N-terminal tail regions (Imhof and Becker, 2001). Acetylation of lysine residues or phosphorylation of serine residues in histone tails induces a transcription-permissive nucleosome conformation, whereas deacetylation, dephosphorylation, or methylation of histone lysine residues promotes a transcription-inhibitory nucleosome conformation. These, and other covalent modifications that occur at histones to alter the degree of chromatin condensation, have become known as the “histone code” (Strahl and Allis, 2000; Jenuwein and Allis, 2001). Chromatin remodeling as a possible circadian regulatory mechanism was first suggested by an experiment demonstrating that in mice, light pulses during the subjective night promote phosphorylation of serine 10 of histone H3 (Crosio et al., 2000). Subsequent work has shown that at the Per1 and Per2 promoters, lysine 9 of histone H3 is rhythmically acetylated (Etchegaray et al., 2003) and that CLOCK and NPAS2 may act to recruit histone acetyltransferases (HATs) to the Per1 promoter (Etchegaray et al., 2003; Curtis et al., 2004).

Interestingly, CLOCK has been shown to exhibit intrinsic HAT activity toward lysine residues of histones H3 and H4 (Doi et al., 2006). This suggests that CLOCK, while activating transcription with its partner, BMAL1, may rhythmically acetylate histones at clock-controlled genes and thereby participate in chromatin remodeling. CLOCK also acetylates non-histone substrates, including BMAL1 at lysine 537 (Hirayama et al., 2007), as well as the glucocorticoid receptor (Nader et al., 2009). Additional work is needed to clarify the relationship of CLOCK HAT activity to the circadian control of gene expression. Recently, a role for CLOCK HAT activity in facilitating herpes simplex virus gene expression in infected mammalian cells has been reported (Kalamvoki and Roizman, 2010), hence this activity of CLOCK may not be restricted solely to circadian regulation.

Histone deacetylases (HDACs) act to remove acetyl groups from histone tails to promote transcriptional repression (Finkel et al., 2009). The NAD+-dependent HDAC Sirtuin 1 (SIRT1) has been shown recently to be involved in the mammalian molecular clock mechanism. SIRT1 acts to deacetylate lysines 9 and 14 of histone H3 as well as lysine 16 of histone H4 leading to chromatin condensation and transcriptional repression (Vaquero et al., 2004; Nakahata et al., 2008). Furthermore, SIRT1 seems to bind CLOCK-BMAL1 to form a complex that is recruited in a circadian manner to promoters of clock-controlled genes. Through its interaction with CLOCK-BMAL1, SIRT1 participates in the circadian expression of Bmal1, Rorc, Per2 and Cry1 (Asher et al., 2008). Interestingly, SIRT1 also acts directly on two core clock components—it reverses the CLOCK-mediated acetylation of lysine 537 of BMAL1 (Nakahata et al., 2008), and deacetylates and promotes the degradation of PER2 (Asher et al., 2008).

It is interesting to note here that because SIRT1 is controlled by the cellular NAD+:NADH ratio, its activity is intimately tied to a cell’s redox state. This finding fits nicely with earlier work showing that the transcriptional activity of CLOCK and NPAS2 is regulated by cellular redox state (Rutter et al., 2001). Expression of the gene encoding the rate-limiting enzyme in the NAD+ salvage pathway, nicotinamide phosphoribosyltransferase (NAMPT), is circadian owing to E-box-mediated CLOCK/BMAL1 activation. The resulting daily oscillation in NAMPT activity produces rhythmic levels of NAD+ in cells (Nakahata et al., 2009; Ramsey et al., 2009). Hence, a novel feedback loop connecting cellular metabolism and the circadian clock has been uncovered whereby CLOCK/BMAL1 positively regulates NAD+ levels, and thus SIRT1 activity through the circadian control of Nampt expression. SIRT1, in turn, negatively regulates CLOCK/BMAL1 activity by promoting transcriptional repression and participates in the oscillation of its own coenzyme, NAD+. As mentioned earlier, this and several other connections between the circadian system and metabolic pathways represent an emerging area of investigation as reviewed comprehensively elsewhere (Green et al., 2008; Bass and Takahashi, 2010; Bellet and Sassone-Corsi, 2010; Maury et al., 2010; Asher and Schibler, 2011).

VII. POSTTRANSCRIPTIONAL CLOCK MECHANISMS

Despite significant progress in elucidating the role of posttranslational regulation of the molecular clock in mammals, only recently have the contributions of posttranscriptional regulatory processes to clock function been explored (Kojima et al., 2011; Staiger and Koster, 2011). Because many of the core clock genes, as well as clock-controlled genes, exhibit circadian oscillations in their transcript levels it is important to determine what processes mediate daily mRNA turnover in mammalian cells. Furthermore, in mice only between 33–50% of the genes that encode rhythmic proteins also manifest rhythmic transcript levels, indicating that the other mRNAs are regulated at the posttranscriptional level (Reddy et al., 2006; Deery et al., 2009).

A. MicroRNAs

Of particular relevance to the elucidation of posttranscriptional clock mechanisms, is recent work revealing circadian control of the expression of microRNAs (miRNAs) and their role in the cycling of clock gene transcripts in mammals. Transcribed from non-coding genomic regions, miRNAs are short, single-stranded RNA molecules 19–25 nucleotides in length that interact with the 3′ untranslated regions (3′UTRs) of target transcripts to induce the cleavage/destabilization of, or to repress translation of, the target mRNA (Bushati and Cohen, 2007; Rana, 2007; Bartel, 2009; Guo et al., 2010). If one report is correct in positing that most mammalian mRNAs are conserved targets of miRNAs (Friedman et al., 2008), it will be important to understand how this regulatory mechanism affects the molecular clock.

Results from a genome-wide screen to identify CREB-regulated miRNAs (Impey et al., 2004) have encouraged analysis of miRNA expression specifically in the SCN of mice (Cheng et al., 2007). Two miRNAs, miR-219 and miR-132, exhibit circadian expression in the SCN and harbor cAMP-reponse elements (CREs) in their promoters, yet only in miR-219 has an E-box element (noncanonical) been identified. Coexpression of CLOCK and BMAL1 in PC12 cells induces expression of miR-219, but not of miR-132. Under constant conditions, both miRNAs exhibit a circadian oscillation solely in the SCN, peaking during the mid-subjective day. This rhythm is abolished in Cry1/Cry2 double mutant mice. Evidence that miR-132 downregulates translation of Per2 comes from treatment of mice with an antagomir to this miRNA. Antagomirs, modified oligoribonucleotides complementary to a specific miRNA, act to block miRNA function (Krutzfeldt et al., 2005). The miR-132 antagomir results in increased PER2 expression in mice. Finally, circadian period length and light-dependent clock resetting are altered by the antagomir-mediated silencing of miR-219 and miR-132, respectively (Cheng et al., 2007).

To further investigate the role of miR-132 in photic entrainment of the SCN clock, one group has generated a transgenic mouse model that conditionally expresses miR-132 in the SCN and forebrain (Alvarez-Saavedra et al., 2011). Following photic stimulation, the miR-123 transgenic animals experience attenuation in light-induced resetting of behavioral rhythms, and a concomitant decline in PER1 and PER2 levels. Analysis of putative miR-132 targets in the SCN has revealed several that are involved in chromatin remodeling and translational control. Through posttranscriptional regulation of these genes, it is proposed that miR-132 is able to control chromatin remodeling and translation within SCN neurons, thus mediating clock entrainment by regulating Per expression. Indeed, the Per1 and Per2 promoters are bound to, and activated by, methyl CpG island binding protein 2 (MeCP2), whereas PABP-interacting protein 2A (PAIP2A) and B-cell translocation gene 2 (BTG2) have the opposite effect—they suppress PER translation by promoting Per mRNA decay (Alvarez-Saavedra et al., 2011).

Another group has identified a role for the miR-192/194 cluster in the regulation of the Per family in HeLa and NIH3T3 cells (Nagel et al., 2009). The 3′UTRs of all three Per genes contain putative target sites for miR-192, miR-194, or both. Evidence shows that miR-192/194 downregulates the Per genes by acting on mRNA. This downregulation causes a shortening of the circadian period (Nagel et al., 2009).

In liver, the most abundant microRNA, miR-122, is transcribed in a rhythmic manner with pri-mir-122 peaking at zeitgeber time 0 (ZT0) and reaching a nadir at ZT12, although the levels of miR-122 remain constant throughout the day (Gatfield et al., 2009). The expression of miR-122 is most likely regulated by REV-ERBα/β through two conserved ROREs in this gene’s promoter as Rev-erbα−/− mice experience attenuation in pri-mir-122 accumulation. Microarray analyses of gene expression in liver tissue of mice treated with a miR-122 antisense oligonucleotide have demonstrated several mRNAs that are under circadian regulation including peroxisome proliferator-activated receptor β/δ (PPARβ/δ) and the PPARα coactivator SMARCD1/BAF60a (SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily d, member 1), both circadian regulators of metabolism (Yang et al., 2006; Li et al., 2008). Indeed, the 3′UTR of Pparβ/δ contains target sites for miR-122, and this protein is upregulated by two-to-threefold upon inactivation of miR-122 (Gatfield et al., 2009). A similar mechanism of miR-122 regulation at the 3′UTR of Smarcd1/Baf60a is apparent. The identification of the circadian regulation of miR-122 expression in liver and its role in the circadian control of downstream targets involved in metabolism provides further insight into tissue-specific links between the circadian system and metabolic function.

B. RNA binding proteins

In addition to the miRNA mechanisms just described, several RNA binding proteins have been identified that regulate clock-related RNA transcripts by different mechanisms. Most RNAs contain cis-acting elements in their 3′UTRs to which trans-acting protein factors can bind and regulate splicing, transport, stability and translation (Moore, 2005; Gratacós and Brewer, 2010). The first RNA binding protein with a demonstrated effect on the mouse molecular clockwork is LARK, for which there are two forms, LARK1 (RBM4) and LARK2 (RBM4B) (Kojima et al., 2007). Although transcripts for both Lark isoforms are found in the SCN, only the protein levels oscillate. Interestingly, the phase of the LARK protein oscillation is similar to that for PER1. Investigation of the Per1 3′UTR has revealed a specific cis element to which LARK binds and promotes translation. Knockdown of Lark transcript causes a shorter circadian period, while overexpression of LARK protein increases circadian period length. Thus, LARK seems to act on Per1 posttranscriptionally by enhancing translation and conferring robustness on the PER1 protein oscillation (Kojima et al., 2007).

One of the more common cis-acting elements in mammalian 3′UTRs is the AU-rich element (ARE) (Gratacós and Brewer, 2010). A protein which binds these AREs and is abundant in many tissues is polypyrimidine tract-binding protein 1 (PTBP1; also known as hnRNP I). Recently, circadian oscillation of the mouse Per2 mRNA has been shown to be regulated by PTBP1 (Woo et al., 2009). PTBP1 binds to the 3′UTR of the Per2 transcript and promotes its decay. Cytoplasmic PTBP1 levels increase at the time at which there is a concomitant rapid decline in Per2 RNA levels. Upon knockdown of PTBP1 expression with siRNA, the peak amplitude of Per2 expression increased. Because several other RNA binding proteins have been detected at the Per2 3′UTR, it may be that the effect of PTBP1 on Per2 RNA decay relies on additional proteins (Woo et al., 2009).

Another RNA binding protein implicated in the regulation of a circadian gene is heterogeneous nuclear ribonucleoprotein D (HNRNPD; also known as AUF1). HNRNPD binds to specific sequences in target mRNA 3′UTRs and regulates transcript stability or the promotion of translation (Gratacós and Brewer, 2010). One group's examination of the Cry1 transcript has revealed a 610-bp 3′UTR which contains an ARE that, when deleted, results in Cry1 mRNA stability (Woo et al., 2010). HNRNPD has been identified as the protein responsible for binding to the Cry1 ARE and promoting mRNA turnover. Similar to the findings for PTPB1 and Per2, the cytoplasmic levels of HNRNPD levels exhibit a maximum as Cry1 mRNA levels are in decline. Knockdown of HNRNPD results in stabilized Cry1 transcript and enhanced oscillation amplitude (Woo et al., 2010). Thus, cytoplasmic destabilization of both the Per2 and Cry1 transcripts share many similarities. It will be interesting to determine what posttranscriptional processes act on the other core clock gene transcripts.

Decay pathways for mRNAs in mammalian cells are either deadenylation-dependent or deadenylation-independent (Gratacós and Brewer, 2010). Both PTBP1 and HNRNPD are involved in the deadenylation-dependent pathway. Deadenylation involves the removal of a transcript's poly(A) tail followed by breakdown of the RNA. It is interesting to note that a circadian deadenylase gene, Nocturnin (Ccrn4l), has been characterized in mammals where it is rhythmically expressed in multiple tissues, including the SCN (Wang et al., 2001). Noc circadian expression is damped in the liver of Clock mutant mice, and is constitutively elevated in Cry double mutant animals (Oishi et al., 2003). Although Noc−/− mice exhibit normal locomotor activity rhythms and clock gene expression, they do have several aberrant metabolic phenotypes (Green et al., 2007). Thus, Noc is a clock-controlled gene—a clock output. Interestingly, it has recently been shown that circadian expression of Noc in mouse liver is controlled by miR-122 (Kojima et al., 2010).

VIII. EFFECTS OF TEMPERATURE ON THE MAMMALIAN CLOCK

A. Temperature as an entraining agent

Temperature is an important environmental entraining agent for many organisms, yet in homoeothermic vertebrates, including mammals, changes in ambient temperature either do not entrain circadian rhythms of locomotor activity, or do so poorly (Hoffmann, 1969; Aschoff and Tokura, 1986; Francis and Coleman, 1997; Palkova et al., 1999). Mammals do, however, experience circadian rhythms in core body temperature with a fluctuation of 1–4°C that are regulated by the SCN (Refinetti and Menaker, 1992). As mentioned previously, cells of most peripheral tissues throughout the mammalian body harbor cell-autonomous circadian oscillators (Welsh et al., 1995; Balsalobre et al., 1998; Nagoshi et al., 2004; Welsh et al., 2004; Yoo et al., 2004) which are synchronized to external cues by rhythmic signals from the SCN (Silver et al., 1996; Earnest et al., 1999; Buijs and Kalsbeek, 2001). These observations raise the intriguing question as to what effect, if any, the normal circadian variation in core body temperature may have upon the cell-autonomous oscillators in peripheral tissues. Could body temperature entrain peripheral oscillators in mammals and, perhaps just as interesting, what properties of the SCN prevent it from being synchronized by environmental temperature cycles?

Studies have demonstrated that circadian rhythms of gene expression in cultures of rat-1 fibroblasts (Brown et al., 2002) and rat astrocytes (Prolo et al., 2005) can be entrained to temperature fluctuations of 4°C and 1.5°C, respectively. Although rhythmicity in both cell types damps within a few cycles upon cessation of the temperature rhythms, damping can be delayed by exposing fibroblasts to natural body temperature oscillations (Brown et al., 2002) or by co-culturing astrocytes with SCN explants (Prolo et al., 2005). Furthermore, when mice are exposed to inverted environmental temperature cycles of 37°C during the day and 24°C during the night, circadian rhythms of gene expression in the liver are reversed without affecting the central clock in the SCN (Brown et al., 2002). It should be mentioned that this phenomenon of decoupling peripheral circadian rhythms from the SCN has also been observed under paradigms of restricted feeding in mammals, and that restricted feeding can alter body temperature rhythms (Damiola et al., 2000; Stokkan et al., 2001). Indeed, it has been proposed that food availability and environmental temperature are related, but independent, entraining cues (Brown et al., 2002). Thus, it is clear that in cultured nonneuronal mammalian cells, and in peripheral tissues in vivo in mammals, temperature cycles can entrain circadian rhythms of gene expression.

In contrast to peripheral oscillators, SCN rhythms in vivo are unaffected by environmental temperature changes that phase shift circadian rhythms in other brain regions (Brown et al., 2002). Recent work in our lab has extended this finding by demonstrating that in mice, the resistance of the SCN to entrainment by ambient temperature is not a cell-autonomous property of individual SCN neurons. Rather, this phenomenon is an emergent property of the SCN network and is therefore dependent upon intercellular coupling among neurons (Buhr et al., 2010). Compelling evidence for the role of intercellular coupling comes from experiments showing that blocking either voltage-gated Na+ channels with TTX or L-type, but not T-type, Ca2+ channels with nimodipine, renders the SCN susceptible to resetting by temperature pulses. Furthermore, communication between the neurochemically-distinct ventral and dorsal regions of the SCN is necessary as cultures of either of these regions alone are shifted in response to temperature changes (Buhr et al., 2010).

Work implying a role for heat shock factor 1 (HSF1) and other components of the heat shock response pathway in circadian gene expression in mammalian liver (Kornmann et al., 2007; Reinke et al., 2008) has encouraged investigation of the possible involvement of this pathway in temperature resetting in mammals. When SCN cultures are pulsed for 1 hr with the heat shock pathway antagonist KNK437, no phase shifts occur. In contrast, the same treatment induces strong phase shifts in pituitary and lung cultures, indicating that in non-SCN tissues, KNK437 mimics the effect of 1-hr cool (33.5°C) pulses in reducing HSF1-mediated transcription (Buhr et al., 2010). Phase shifts to warm (38.6°C) pulses observed in lung and pituitary are blocked both by KNK437 and quercetin, another HSF1 inhibitor. Continuous inhibition of HSF1-mediated transcription via chronic KNK437 administration to SCN, lung and pituitary cultures causes an increase in circadian period. This effect is consistent with period lengthening of the circadian rhythm of locomotor activity observed in Hsf1−/− mice (Reinke et al., 2008). Taken together, these results suggest a molecular mechanism that involves HSF1 in temperature resetting in mammalian peripheral tissues (Buhr et al., 2010).

It is important to mention here that two reports, in contrast to the work just presented, provide evidence that rat SCN slice cultures do exhibit entrainment to temperature cycles (Ruby et al., 1999; Herzog and Huckfeldt, 2003). Several differences in these studies may account for this discrepancy. First, the work of Ruby et al. (1999) relied on extracellular recordings from single neurons as glass electrodes advanced along tracks through SCN slices maintained in culture for up to 60 hr. Herzog and Huckfeldt (2003) measured PER1::LUC rhythms from neonatal and juvenile rat SCN slices in cultures for up to two weeks and observed that these slice preparations were more sensitive to temperature pulses than adult tissues. In contrast, our studies used organotypic cultures of SCN and peripheral tissues from Per2Luc reporter mice and measured in real time the PER2::LUC bioluminescence rhythm continuously for several days (Yoo et al., 2004; Buhr et al., 2010). The in vivo study of Brown et al. (2002) examined mice exposed to environmental temperature changes over a several day period and measured Dbp expression in SCN sections by in situ hybridization. Thus, the SCN culture techniques and output rhythms measured in each study differed. A final possibility is that species-specific differences between mice and rats account for the differences observed in some of these studies. Further work is necessary to clarify these issues.

B. Temperature compensation

One of the hallmarks of circadian rhythms in all organisms, from cyanobacteria to mammals, is that they are temperature compensated—daily rhythms remain constant as temperature increases or decreases across a physiologically viable range (Pittendrigh, 1993). This clock property can be expressed quantitatively as the Q10, or temperature coefficient—the change in the rate of a biological rhythm or biochemical reaction as a result of increasing the ambient temperature by 10°C. The Q10 for most biochemical reactions is 2 to 3, yet circadian rhythms usually have a Q10 between 0.8–1.2 (Sweeney and Hastings, 1960). Efforts to elucidate a molecular mechanism underlying the temperature compensation property of circadian clocks have been unsuccessful until recently. The striking demonstration that the circadian rhythm of phosphorylation of the cyanobacterial clock protein, KaiC, can be reconstituted in vitro with just three Kai proteins and ATP, has led to experiments revealing that this in vitro oscillator is also temperature compensated (Nakajima et al., 2005; Tomita et al., 2005). Hence, as the cyanobacterial example aptly illustrates, temperature compensation must be an inherent property of at least some of the biochemical reactions comprising the molecular mechanism of the circadian clock of any given species.

In mammals, temperature compensation has been demonstrated in cell culture for rat-1 (Izumo et al., 2003) and mouse (Tsuchiya et al., 2003; Dibner et al., 2009) fibroblasts, and for neonatal rat SCN neurons (Herzog and Huckfeldt, 2003). As for whole neural tissues, hamster retina (Tosini and Menaker, 1998), and rat (Ruby et al., 1999), mouse (Buhr et al., 2010), and ground squirrel (Ruby and Heller, 1996) SCN are temperature compensated. Several mammalian peripheral tissues also exhibit this property (Reyes et al., 2008), as do human red blood cells (O'Neill and Reddy, 2011). Work by our group has also shown that the temperature compensation observed in SCN tissue is a cell-autonomous property and does not rely on intercellular coupling, unlike the resistance to temperature resetting of the SCN which is an emergent property of the SCN network (Buhr et al., 2010).

Genetic mutations affecting the period of the circadian clock in mammals can also affect temperature compensation as shown for isolated tau mutant hamster retinal cultures (Tosini and Menaker, 1998). This is particularly intriguing in light of recent work demonstrating both in cell culture and in cell-free in vitro reactions, that the enzymatic activity of CK1δ/ε toward circadian substrates such as PER2 is temperature-insensitive, but temperature-sensitive toward non-circadian substrates (Isojima et al., 2009). Both wild-type and CK1εtau exhibit temperature-insensitive phosphorylation of a peptide containing the PER2 βTrCP binding region, yet when a peptide substrate derived from the FASPS region of PER2 is tested, an increase in the temperature sensitivity of the CK1εtau mutant enzyme is revealed (Isojima et al., 2009). An effect of autophosphorylation state on CK1δ/ε activity toward circadian-relevant substrates in vitro is also apparent. Thus, similar to the cyanobacterial system mentioned previously, the temperature-insensitive phosphorylation of mammalian circadian substrates by CK1δ/ε can be reconstituted in vitro, and this property of CK1δ/ε is dependent both on the substrate and on the phosphorylation state of the enzyme (Isojima et al., 2009). Additional work will be necessary to replicate these results and to elucidate the biochemical mechanism underlying CK1δ/ε circadian-specific temperature compensation.

IX. UNRESOLVED ISSUES AND FUTURE DIRECTIONS

As mentioned at the beginning of this review, extensive work has shown that across phyla, the primary molecular mechanism underlying cell-autonomous circadian oscillators is composed of autoregulatory feedback loops of transcription and translation. Hence, the existence of transcription-independent oscillations and a potential role for such oscillations in the function of the cellular clock in mammals and other organisms seems surprising. Indeed, it was the cell-free recapitulation in a test tube of the circadian rhythm of KaiC phosphorylation by colleagues working on the cyanobacterial circadian clock (Nakajima et al., 2005; Tomita et al., 2005) that generated recent interest in transcription-independent clock processes. Further work in cyanobacteria has shown that both circadian transcriptional/translational mechanisms and transcription-independent posttranslational mechanisms are necessary for a competent circadian clock in this organism (Kitayama et al., 2008).

Hints that transcription-independent processes in the mammalian circadian clock exist come from several studies. The surprising discovery that rhythmic transcription of the core clock genes Bmal1, Cry1, and Cry2 (Fan et al., 2007; Liu et al., 2008) is not necessary for circadian clock function in mammalian cells, and that the mammalian cellular clock is particularly resilient to attenuation of transcription (Dibner et al., 2009), suggests that mechanisms other than the transcriptional/translational feedback loop are involved in the generation of oscillations. Perhaps the most striking demonstration of a transcription-independent circadian rhythm in mammals is the recent report of a daily oscillation in human red blood cells of the oxidation and subsequent monomer-to-dimer transition of peroxiredoxin (PRX), a protein that inactivates reactive oxygen species (O'Neill and Reddy, 2011). In liver cells, expression of PRX is circadian (Reddy et al., 2006), but this is not possible in mature erythrocytes which have no nucleus. Inhibitors of transcription and translation have no effect on the circadian rhythm of PRX oxidation in human red blood cells further suggesting that the observed PRX oxidation rhythm in erythrocytes is a transcription-independent process. Moreover, detection of another circadian rhythm in erythrocytes—the transition of hemoglobin between dimer and tetramer states—seems also to be transcription-independent. A circadian rhythm of PRX oxidation has also been demonstrated by the same group in the green algae Ostreococcus tauri, a primitive eukaryote, even when gene expression is halted by exposing these cells to DD (O'Neill et al., 2011). Although these findings must be repeated and validated in other model organisms, they suggest an intriguing avenue for further work.

A major goal toward understanding any biological system is to successfully model that system mathematically. Accomplishing this requires that biologists and modelers work together to incorporate experimental results into models such that they may be used to make testable predictions. The ongoing development of models of the mammalian circadian clock has been particularly helpful recently as they take into account the “combinatorial complexity” of clock component interactions as well as stochastic properties (Yamada and Forger, 2010). Model-based predictions regarding the effects of mutations in core clock genes (Liu et al., 2007b), the role of posttranslational processes on clock components (Gallego et al., 2006a), and the electrical properties of the SCN (Belle et al., 2009), have been validated empirically. The necessity for both molecular noise and intercellular coupling to induce rhythms in a population of SCN neurons has also been predicted via modeling and subsequently confirmed by experiment (Ko et al., 2010). A future challenge to clock modelers will be to address the complex connections and interactions between the mammalian circadian clock and other systems, including the many emerging links between the clock and metabolism.

While there are many potentially fruitful paths of pursuit in the study of mammalian circadian clock genetics, we will conclude by mentioning a few here. Large-scale screening of small-molecule libraries may yield promising targets for therapeutic intervention of circadian-related genetic disorders in humans including FASPS and seasonal affective disorder (Liu et al., 2007a). New approaches in synthetic biology in which artificial transcriptional circuits are used to define networks of oscillating genes or to interrogate the function of circadian-related cis-acting elements promise to further our understanding of clock transcriptional pathways (Ueda et al., 2005; Kumaki et al., 2008; Ukai-Tadenuma et al., 2008). Understanding circadian phenotypes that occur in mammals in the absence of underlying changes in DNA sequence–via epigenetic processes–will require ongoing work (Bellet and Sassone-Corsi, 2010; Ripperger and Merrow, 2011). Elucidation of the molecular and biochemical mechanisms of temperature-insensitive phenomena in mammalian clock systems remains an important goal (Brown et al., 2002; Isojima et al., 2009; Buhr et al., 2010). In all of these endeavors, the mouse will remain the mammalian genetic model of choice.

X. CONCLUSION

Over the past 10 years remarkable progress has been made in our understanding of the genetics of the mammalian circadian clock. The transcriptional/translational feedback loop model of the molecular oscillator within cells, for which there is evidence across phyla, has formed the foundation of our understanding of the molecular clockwork. This model, however, must be modified given the new levels of hierarchy and complexity evident from recent work. It is necessary to study the mammalian clock at all levels from single cells, to cell-cell interactions within a tissue, to tissue-level properties, and finally, at the level of behavior. Emergent clock properties arise from the interactions among cells—properties that cannot be studied at the single-cell level. Likewise, focusing on behavior or tissue-specific processes alone will overlook cell-autonomous clock properties. New advances in reporter technology, microarrays, mathematical modeling, perturbation analysis methods, and systems biology will continue to elucidate properties of the molecular clock. Hence, a lesson learned from the work presented herein is that with respect to the mammalian circadian clock, a systems-wide approach must be advocated. In the near future we should look forward to a better understanding of how the mammalian clockwork is integrated with the other physiological systems of the body, and perhaps be better able to develop therapies for human clock-related disorders.

ACKNOWLEDGEMENTS

This work was supported by NIH grant 1R15GM086825-01 to P.L.L. and NIH grants U01 MH61915, P50 MH074924, and R01 MH078024 to J.S.T. J.S.T. is an Investigator in the Howard Hughes Medical Institute.

REFERENCES

  1. Abe M, Herzog ED, Block GD. Lithium lengthens the circadian period of individual suprachiasmatic nucleus neurons. NeuroReport. 2000;11:3261–3264. doi: 10.1097/00001756-200009280-00042. [DOI] [PubMed] [Google Scholar]
  2. Abraham U, Granada AE, Westermark PO, Heine M, Kramer A, Herzel H. Coupling governs entrainment range of circadian clocks. Mol. Syst. Biol. 2010;6:438. doi: 10.1038/msb.2010.92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Abrahamson EE, Moore RY. Suprachiasmatic nucleus in the mouse: retinal innervation, intrinsic organization and efferent projections. Brain Res. 2001;916:172–191. doi: 10.1016/s0006-8993(01)02890-6. [DOI] [PubMed] [Google Scholar]
  4. Adams DJ, van der Weyden L. Contemporary approaches for modifying the mouse genome. Physiol. Genomics. 2008;34:225–238. doi: 10.1152/physiolgenomics.90242.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Akashi M, Tsuchiya Y, Yoshino T, Nishida E. Control of intracellular dynamics of mammalian period proteins by casein kinase I ɛ (CKIɛ) and CKIδ in cultured cells. Mol. Cell. Biol. 2002;22:1693–1703. doi: 10.1128/MCB.22.6.1693-1703.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Akashi M, Takumi T. The orphan nuclear receptor RORα regulates circadian transcription of the mammalian core-clock Bmal1 . Nat. Struct. Mol. Biol. 2005;12:441–448. doi: 10.1038/nsmb925. [DOI] [PubMed] [Google Scholar]
  7. Akhtar RA, Reddy AB, Maywood ES, Clayton JD, King VM, Smith AG, Gant TW, Hastings MH, Kyriacou CP. Circadian cycling of the mouse liver transcriptome, as revealed by cDNA microarray, is driven by the suprachiasmatic nucleus. Curr. Biol. 2002;12:540–550. doi: 10.1016/s0960-9822(02)00759-5. [DOI] [PubMed] [Google Scholar]
  8. Albrecht U, Sun ZS, Eichele G, Lee CC. A differential response of two putative mammalian circadian regulators, mper1 and mper2, to light. Cell. 1997;91:1055–1064. doi: 10.1016/s0092-8674(00)80495-x. [DOI] [PubMed] [Google Scholar]
  9. Ali A, Hoeflich KP, Woodgett JR. Glycogen synthase kinase-3: properties, functions, and regulation. Chem. Rev. 2001;101:2527–2540. doi: 10.1021/cr000110o. [DOI] [PubMed] [Google Scholar]
  10. Alvarez-Saavedra M, Antoun G, Yanagiya A, Oliva-Hernandez R, Cornejo-Palma D, Perez-Iratxeta C, Sonenberg N, Cheng HY. miRNA-132 orchestrates chromatin remodeling and translational control of the circadian clock. Hum. Mol. Genet. 2011;20:731–751. doi: 10.1093/hmg/ddq519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Antoch MP, Song EJ, Chang AM, Vitaterna MH, Zhao Y, Wilsbacher LD, Sangoram AM, King DP, Pinto LH, Takahashi JS. Functional identification of the mouse circadian Clock gene by transgenic BAC rescue. Cell. 1997;89:655–667. doi: 10.1016/s0092-8674(00)80246-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Aschoff J, Tokura H. Circadian activity rhythms in squirrel monkeys: entrainment by temperature cycles. J. Biol. Rhythms. 1986;1:91–99. doi: 10.1177/074873048600100201. [DOI] [PubMed] [Google Scholar]
  13. Asher G, Gatfield D, Stratmann M, Reinke H, Dibner C, Kreppel F, Mostoslavsky R, Alt FW, Schibler U. SIRT1 regulates circadian clock gene expression through PER2 deacetylation. Cell. 2008;134:317–328. doi: 10.1016/j.cell.2008.06.050. [DOI] [PubMed] [Google Scholar]
  14. Asher G, Schibler U. Crosstalk between components of circadian and metabolic cycles in mammals. Cell Metab. 2011;13:125–137. doi: 10.1016/j.cmet.2011.01.006. [DOI] [PubMed] [Google Scholar]
  15. Aston-Jones G, Chen S, Zhu Y, Oshinsky ML. A neural circuit for circadian regulation of arousal. Nat. Neurosci. 2001;4:732–738. doi: 10.1038/89522. [DOI] [PubMed] [Google Scholar]
  16. Aton SJ, Colwell CS, Harmar AJ, Waschek J, Herzog ED. Vasoactive intestinal polypeptide mediates circadian rhythmicity and synchrony in mammalian clock neurons. Nat. Neurosci. 2005;8:476–483. doi: 10.1038/nn1419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Bacon Y, Ooi A, Kerr S, Shaw-Andrews L, Winchester L, Breeds S, Tymoska-Lalanne Z, Clay J, Greenfield AG, Nolan PM. Screening for novel ENU-induced rhythm, entrainment and activity mutants. Genes Brain Behav. 2004;3:196–205. doi: 10.1111/j.1601-183X.2004.00070.x. [DOI] [PubMed] [Google Scholar]
  18. Badura L, Swanson T, Adamowicz W, Adams J, Cianfrogna J, Fisher K, Holland J, Kleiman R, Nelson F, Reynolds L, et al. An inhibitor of casein kinase Iɛ induces phase delays in circadian rhythms under free-running and entrained conditions. J. Pharmacol. Exp. Ther. 2007;322:730–738. doi: 10.1124/jpet.107.122846. [DOI] [PubMed] [Google Scholar]
  19. Bae K, Jin X, Maywood ES, Hastings MH, Reppert SM, Weaver DR. Differential functions of mPer1, mPer2, and mPer3 in the SCN circadian clock. Neuron. 2001;30:525–536. doi: 10.1016/s0896-6273(01)00302-6. [DOI] [PubMed] [Google Scholar]
  20. Baggs JE, Price TS, DiTacchio L, Panda S, Fitzgerald GA, Hogenesch JB. Network features of the mammalian circadian clock. PLoS Biol. 2009;7:e52. doi: 10.1371/journal.pbio.1000052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Balsalobre A, Damiola F, Schibler U. A serum shock induces circadian gene expression in mammalian tissue culture cells. Cell. 1998;93:929–937. doi: 10.1016/s0092-8674(00)81199-x. [DOI] [PubMed] [Google Scholar]
  22. Balsalobre A, Brown SA, Marcacci L, Tronche F, Kellendonk C, Reichardt HM, Schutz G, Schibler U. Resetting of circadian time in peripheral tissues by glucocorticoid signaling. Science. 2000;289:2344–2347. doi: 10.1126/science.289.5488.2344. [DOI] [PubMed] [Google Scholar]
  23. Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136:215–233. doi: 10.1016/j.cell.2009.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Bass J, Takahashi JS. Circadian integration of metabolism and energetics. Science. 2010;330:1349–1354. doi: 10.1126/science.1195027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Belle MD, Diekman CO, Forger DB, Piggins HD. Daily electrical silencing in the mammalian circadian clock. Science. 2009;326:281–284. doi: 10.1126/science.1169657. [DOI] [PubMed] [Google Scholar]
  26. Bellet MM, Sassone-Corsi P. Mammalian circadian clock and metabolism - the epigenetic link. J. Cell Sci. 2010;123:3837–3848. doi: 10.1242/jcs.051649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Berson DM, Dunn FA, Takao M. Phototransduction by retinal ganglion cells that set the circadian clock. Science. 2002;295:1070–1073. doi: 10.1126/science.1067262. [DOI] [PubMed] [Google Scholar]
  28. Blake JA, Bult CJ, Kadin JA, Richardson JE, Eppig JT. The Mouse Genome Database (MGD): premier model organism resource for mammalian genomics and genetics. Nucleic Acids Res. 2010;39:D842–D848. doi: 10.1093/nar/gkq1008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Brown S, Zumbrunn G, Fleury-Olela F, Preitner N, Schibler U. Rhythms of mammalian body temperature can sustain peripheral circadian clocks. Curr. Biol. 2002;12:1574–1583. doi: 10.1016/s0960-9822(02)01145-4. [DOI] [PubMed] [Google Scholar]
  30. Brown TM, Colwell CS, Waschek JA, Piggins HD. Disrupted neuronal activity rhythms in the suprachiasmatic nuclei of vasoactive intestinal polypeptide-deficient mice. J. Neurophysiol. 2007;97:2553–2558. doi: 10.1152/jn.01206.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Buhr ED, Yoo SH, Takahashi JS. Temperature as a universal resetting cue for mammalian circadian oscillators. Science. 2010;330:379–385. doi: 10.1126/science.1195262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Buijs RM, Kalsbeek A. Hypothalamic integration of central and peripheral clocks. Nat. Rev. Neurosci. 2001;2:521–526. doi: 10.1038/35081582. [DOI] [PubMed] [Google Scholar]
  33. Bunger MK, Wilsbacher LD, Moran SM, Clendenin C, Radcliffe LA, Hogenesch JB, Simon C, Takahashi JS, Bradfield CA. Mop3 is an essential component of the master circadian pacemaker in mammals. Cell. 2000;103:1009–1017. doi: 10.1016/s0092-8674(00)00205-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Bushati N, Cohen SM. microRNA functions. Annu. Rev. Cell. Dev. Biol. 2007;23:175–205. doi: 10.1146/annurev.cellbio.23.090506.123406. [DOI] [PubMed] [Google Scholar]
  35. Busino L, Bassermann F, Maiolica A, Lee C, Nolan PM, Godinho SI, Draetta GF, Pagano M. SCFFbxl3 controls the oscillation of the circadian clock by directing the degradation of cryptochrome proteins. Science. 2007;316:900–904. doi: 10.1126/science.1141194. [DOI] [PubMed] [Google Scholar]
  36. Camacho F, Cilio M, Guo Y, Virshup DM, Patel K, Khorkova O, Styren S, Morse B, Yao Z, Keesler GA. Human casein kinase Iδ phosphorylation of human circadian clock proteins period 1 and 2. FEBS Lett. 2001;489:159–165. doi: 10.1016/s0014-5793(00)02434-0. [DOI] [PubMed] [Google Scholar]
  37. Cardone L, Hirayama J, Giordano F, Tamaru T, Palvimo JJ, Sassone-Corsi P. Circadian clock control by SUMOylation of BMAL1. Science. 2005;309:1390–1394. doi: 10.1126/science.1110689. [DOI] [PubMed] [Google Scholar]
  38. Cardozo T, Pagano M. The SCF ubiquitin ligase: insights into a molecular machine. Nat. Rev. Mol. Cell Biol. 2004;5:739–751. doi: 10.1038/nrm1471. [DOI] [PubMed] [Google Scholar]
  39. Cermakian N, Monaco L, Pando MP, Dierich A, Sassone-Corsi P. Altered behavioral rhythms and clock gene expression in mice with a targeted mutation in the Period1 gene. EMBO J. 2001;20:3967–3974. doi: 10.1093/emboj/20.15.3967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Chen R, Schirmer A, Lee Y, Lee H, Kumar V, Yoo SH, Takahashi JS, Lee C. Rhythmic PER abundance defines a critical nodal point for negative feedback within the circadian clock mechanism. Mol. Cell. 2009;36:417–430. doi: 10.1016/j.molcel.2009.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Cheng HY, Papp JW, Varlamova O, Dziema H, Russell B, Curfman JP, Nakazawa T, Shimizu K, Okamura H, Impey S, et al. microRNA modulation of circadian-clock period and entrainment. Neuron. 2007;54:813–829. doi: 10.1016/j.neuron.2007.05.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Cheng MY, Bullock CM, Li C, Lee AG, Bermak JC, Belluzzi J, Weaver DR, Leslie FM, Zhou QY. Prokineticin 2 transmits the behavioural circadian rhythm of the suprachiasmatic nucleus. Nature. 2002;417:405–410. doi: 10.1038/417405a. [DOI] [PubMed] [Google Scholar]
  43. Ciechanover A, Orian A, Schwartz AL. Ubiquitin-mediated proteolysis: biological regulation via destruction. Bioessays. 2000;22:442–451. doi: 10.1002/(SICI)1521-1878(200005)22:5<442::AID-BIES6>3.0.CO;2-Q. [DOI] [PubMed] [Google Scholar]
  44. Clark AT, Goldowitz D, Takahashi JS, Vitaterna MH, Siepka SM, Peters LL, Frankel WN, Carlson GA, Rossant J, Nadeau JH, et al. Implementing large-scale ENU mutagenesis screens in North America. Genetica. 2004;122:51–64. doi: 10.1007/s10709-004-1436-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Colwell CS, Michel S, Itri J, Rodriguez W, Tam J, Lelievre V, Hu Z, Liu X, Waschek JA. Disrupted circadian rhythms in VIP- and PHI-deficient mice. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2003;285:R939–R949. doi: 10.1152/ajpregu.00200.2003. [DOI] [PubMed] [Google Scholar]
  46. Crosio C, Cermakian N, Allis CD, Sassone-Corsi P. Light induces chromatin modification in cells of the mammalian circadian clock. Nat. Neurosci. 2000;3:1241–1247. doi: 10.1038/81767. [DOI] [PubMed] [Google Scholar]
  47. Curtis AM, Seo SB, Westgate EJ, Rudic RD, Smyth EM, Chakravarti D, FitzGerald GA, McNamara P. Histone acetyltransferase-dependent chromatin remodeling and the vascular clock. J. Biol. Chem. 2004;279:7091–7097. doi: 10.1074/jbc.M311973200. [DOI] [PubMed] [Google Scholar]
  48. Cutler DJ, Haraura M, Reed HE, Shen S, Sheward WJ, Morrison CF, Marston HM, Harmar AJ, Piggins HD. The mouse VPAC2 receptor confers suprachiasmatic nuclei cellular rhythmicity and responsiveness to vasoactive intestinal polypeptide in vitro . Eur. J. Neurosci. 2003;17:197–204. doi: 10.1046/j.1460-9568.2003.02425.x. [DOI] [PubMed] [Google Scholar]
  49. Dacey DM, Liao HW, Peterson BB, Robinson FR, Smith VC, Pokorny J, Yau KW, Gamlin PD. Melanopsin-expressing ganglion cells in primate retina signal colour and irradiance and project to the LGN. Nature. 2005;433:749–754. doi: 10.1038/nature03387. [DOI] [PubMed] [Google Scholar]
  50. Damiola F, Le Minh N, Preitner N, Kornmann B, Fleury-Olela F, Schibler U. Restricted feeding uncouples circadian oscillators in peripheral tissues from the central pacemaker in the suprachiasmatic nucleus. Genes Dev. 2000;14:2950–2961. doi: 10.1101/gad.183500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Dardente H, Fortier EE, Martineau V, Cermakian N. Cryptochromes impair phosphorylation of transcriptional activators in the clock: a general mechanism for circadian repression. Biochem. J. 2007;402:525–536. doi: 10.1042/BJ20060827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Debruyne JP, Noton E, Lambert CM, Maywood ES, Weaver DR, Reppert SM. A clock shock: mouse CLOCK is not required for circadian oscillator function. Neuron. 2006;50:465–477. doi: 10.1016/j.neuron.2006.03.041. [DOI] [PubMed] [Google Scholar]
  53. DeBruyne JP, Weaver DR, Reppert SM. CLOCK and NPAS2 have overlapping roles in the suprachiasmatic circadian clock. Nat. Neurosci. 2007a;10:543–545. doi: 10.1038/nn1884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. DeBruyne JP, Weaver DR, Reppert SM. Peripheral circadian oscillators require CLOCK. Curr. Biol. 2007b;17:R538–R539. doi: 10.1016/j.cub.2007.05.067. [DOI] [PubMed] [Google Scholar]
  55. Deery MJ, Maywood ES, Chesham JE, Sladek M, Karp NA, Green EW, Charles PD, Reddy AB, Kyriacou CP, Lilley KS, et al. Proteomic analysis reveals the role of synaptic vesicle cycling in sustaining the suprachiasmatic circadian clock. Curr. Biol. 2009;19:2031–2036. doi: 10.1016/j.cub.2009.10.024. [DOI] [PubMed] [Google Scholar]
  56. Dibner C, Sage D, Unser M, Bauer C, d'Eysmond T, Naef F, Schibler U. Circadian gene expression is resilient to large fluctuations in overall transcription rates. EMBO J. 2009;28:123–134. doi: 10.1038/emboj.2008.262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Do MT, Yau KW. Intrinsically photosensitive retinal ganglion cells. Physiol. Rev. 2010;90:1547–1581. doi: 10.1152/physrev.00013.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Doble BW, Woodgett JR. GSK-3: tricks of the trade for a multi-tasking kinase. J. Cell Sci. 2003;116:1175–1186. doi: 10.1242/jcs.00384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Doherty CJ, Kay SA. Circadian control of global gene expression patterns. Annu. Rev. Genet. 2010;44:419–444. doi: 10.1146/annurev-genet-102209-163432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Doi M, Hirayama J, Sassone-Corsi P. Circadian regulator CLOCK is a histone acetyltransferase. Cell. 2006;125:497–508. doi: 10.1016/j.cell.2006.03.033. [DOI] [PubMed] [Google Scholar]
  61. Dudley CA, Erbel-Sieler C, Estill SJ, Reick M, Franken P, Pitts S, McKnight SL. Altered patterns of sleep and behavioral adaptability in NPAS2-deficient mice. Science. 2003;301:379–383. doi: 10.1126/science.1082795. [DOI] [PubMed] [Google Scholar]
  62. Duffield GE, Best JD, Meurers BH, Bittner A, Loros JJ, Dunlap JC. Circadian programs of transcriptional activation, signaling, and protein turnover revealed by microarray analysis of mammalian cells. Curr. Biol. 2002;12:551–557. doi: 10.1016/s0960-9822(02)00765-0. [DOI] [PubMed] [Google Scholar]
  63. Dunlap JC. Molecular bases for circadian clocks. Cell. 1999;96:271–290. doi: 10.1016/s0092-8674(00)80566-8. [DOI] [PubMed] [Google Scholar]
  64. Earnest DJ, Liang FQ, Ratcliff M, Cassone VM. Immortal time: circadian clock properties of rat suprachiasmatic cell lines. Science. 1999;283:693–695. doi: 10.1126/science.283.5402.693. [DOI] [PubMed] [Google Scholar]
  65. Ebling FJ. The role of glutamate in the photic regulation of the suprachiasmatic nucleus. Prog. Neurobiol. 1996;50:109–132. doi: 10.1016/s0301-0082(96)00032-9. [DOI] [PubMed] [Google Scholar]
  66. Eide EJ, Kang H, Crapo S, Gallego M, Virshup DM. Casein kinase I in the mammalian circadian clock. Methods Enzymol. 2005a;393:408–418. doi: 10.1016/S0076-6879(05)93019-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Eide EJ, Woolf MF, Kang H, Woolf P, Hurst W, Camacho F, Vielhaber EL, Giovanni A, Virshup DM. Control of mammalian circadian rhythm by CKIɛ-regulated proteasome-mediated PER2 degradation. Mol. Cell. Biol. 2005b;25:2795–2807. doi: 10.1128/MCB.25.7.2795-2807.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Etchegaray JP, Lee C, Wade PA, Reppert SM. Rhythmic histone acetylation underlies transcription in the mammalian circadian clock. Nature. 2003;421:177–182. doi: 10.1038/nature01314. [DOI] [PubMed] [Google Scholar]
  69. Etchegaray JP, Machida KK, Noton E, Constance CM, Dallmann R, Di Napoli MN, DeBruyne JP, Lambert CM, Yu EA, Reppert SM, et al. Casein kinase 1 delta regulates the pace of the mammalian circadian clock. Mol. Cell. Biol. 2009;29:3853–3866. doi: 10.1128/MCB.00338-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Etchegaray JP, Yu EA, Indic P, Dallmann R, Weaver DR. Casein kinase 1 delta (CK1δ) regulates period length of the mouse suprachiasmatic circadian clock in vitro . PLoS One. 2010;5:e10303. doi: 10.1371/journal.pone.0010303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Fan Y, Hida A, Anderson DA, Izumo M, Johnson CH. Cycling of CRYPTOCHROME proteins is not necessary for circadian-clock function in mammalian fibroblasts. Curr. Biol. 2007;17:1091–1100. doi: 10.1016/j.cub.2007.05.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Finkel T, Deng CX, Mostoslavsky R. Recent progress in the biology and physiology of sirtuins. Nature. 2009;460:587–591. doi: 10.1038/nature08197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Foster RG, Provencio I, Hudson D, Fiske S, De Grip W, Menaker M. Circadian photoreception in the retinally degenerate mouse (rd/rd) J. Comp. Physiol. A. 1991;169:39–50. doi: 10.1007/BF00198171. [DOI] [PubMed] [Google Scholar]
  74. Fox J, Barthold S, Davvison M, Newcomer C, Quimby F, Smith A, editors. The mouse in biomedical research. 2nd ed. 4 vols. Boston: Elsevier; 2007. [Google Scholar]
  75. Francis AJ, Coleman GJ. Phase response curves to ambient temperature pulses in rats. Physiol. Behav. 1997;62:1211–1217. doi: 10.1016/s0031-9384(97)00202-3. [DOI] [PubMed] [Google Scholar]
  76. Freedman MS, Lucas RJ, Soni B, von Schantz M, Munoz M, David-Gray Z, Foster R. Regulation of mammalian circadian behavior by non-rod, non-cone, ocular photoreceptors. Science. 1999;284:502–504. doi: 10.1126/science.284.5413.502. [DOI] [PubMed] [Google Scholar]
  77. Friedman RC, Farh KKH, Burge CB, Bartel DP. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 2008;19:92–105. doi: 10.1101/gr.082701.108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Fu Y, Zhong H, Wang MH, Luo DG, Liao HW, Maeda H, Hattar S, Frishman LJ, Yau KW. Intrinsically photosensitive retinal ganglion cells detect light with a vitamin A-based photopigment, melanopsin. Proc. Natl. Acad. Sci. USA. 2005;102:10339–10344. doi: 10.1073/pnas.0501866102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Gallego M, Eide EJ, Woolf MF, Virshup DM, Forger DB. An opposite role for tau in circadian rhythms revealed by mathematical modeling. Proc. Natl. Acad. Sci. USA. 2006a;103:10618–10623. doi: 10.1073/pnas.0604511103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Gallego M, Kang H, Virshup DM. Protein phosphatase 1 regulates the stability of the circadian protein PER2. Biochem. J. 2006b;399:169–175. doi: 10.1042/BJ20060678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Gallego M, Virshup DM. Post-translational modifications regulate the ticking of the circadian clock. Nat. Rev. Mol. Cell Biol. 2007;8:139–148. doi: 10.1038/nrm2106. [DOI] [PubMed] [Google Scholar]
  82. Gareau JR, Lima CD. The SUMO pathway: emerging mechanisms that shape specificity, conjugation and recognition. Nat. Rev. Mol. Cell Biol. 2010;11:861–871. doi: 10.1038/nrm3011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Gatfield D, Le Martelot G, Vejnar CE, Gerlach D, Schaad O, Fleury-Olela F, Ruskeepaa AL, Oresic M, Esau CC, Zdobnov EM, et al. Integration of microRNA miR-122 in hepatic circadian gene expression. Genes Dev. 2009;23:1313–1326. doi: 10.1101/gad.1781009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Gekakis N, Staknis D, Nguyen HB, Davis FC, Wilsbacher LD, King DP, Takahashi JS, Weitz CJ. Role of the CLOCK protein in the mammalian circadian mechanism. Science. 1998;280:1564–1569. doi: 10.1126/science.280.5369.1564. [DOI] [PubMed] [Google Scholar]
  85. Gietzen KF, Virshup DM. Identification of inhibitory autophosphorylation sites in casein kinase Iɛ. J. Biol. Chem. 1999;274:32063–32070. doi: 10.1074/jbc.274.45.32063. [DOI] [PubMed] [Google Scholar]
  86. Godinho SI, Maywood ES, Shaw L, Tucci V, Barnard AR, Busino L, Pagano M, Kendall R, Quwailid MM, Romero MR, et al. The after-hours mutant reveals a role for Fbxl3 in determining mammalian circadian period. Science. 2007;316:897–900. doi: 10.1126/science.1141138. [DOI] [PubMed] [Google Scholar]
  87. Golombek DA, Rosenstein RE. Physiology of circadian entrainment. Physiol. Rev. 2010;90:1063–1102. doi: 10.1152/physrev.00009.2009. [DOI] [PubMed] [Google Scholar]
  88. Gooley JJ, Lu J, Chou TC, Scammell TE, Saper CB. Melanopsin in cells of origin of the retinohypothalamic tract. Nat. Neurosci. 2001;4:1165. doi: 10.1038/nn768. [DOI] [PubMed] [Google Scholar]
  89. Goz D, Studholme K, Lappi DA, Rollag MD, Provencio I, Morin LP. Targeted destruction of photosensitive retinal ganglion cells with a saporin conjugate alters the effects of light on mouse circadian rhythms. PLoS One. 2008;3:e3153. doi: 10.1371/journal.pone.0003153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Granados-Fuentes D, Prolo LM, Abraham U, Herzog ED. The suprachiasmatic nucleus entrains, but does not sustain, circadian rhythmicity in the olfactory bulb. J. Neurosci. 2004a;24:615–619. doi: 10.1523/JNEUROSCI.4002-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Granados-Fuentes D, Saxena MT, Prolo LM, Aton SJ, Herzog ED. Olfactory bulb neurons express functional, entrainable circadian rhythms. Eur. J. Neurosci. 2004b;19:898–906. doi: 10.1111/j.0953-816x.2004.03117.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Granados-Fuentes D, Tseng A, Herzog ED. A circadian clock in the olfactory bulb controls olfactory responsivity. J. Neurosci. 2006;26:12219–12225. doi: 10.1523/JNEUROSCI.3445-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Gratacós FM, Brewer G. The role of AUF1 in regulated mRNA decay. WIREs RNA. 2010;1:457–473. doi: 10.1002/wrna.26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Green CB, Douris N, Kojima S, Strayer CA, Fogerty J, Lourim D, Keller SR, Besharse JC. Loss of Nocturnin, a circadian deadenylase, confers resistance to hepatic steatosis and diet-induced obesity. Proc. Natl. Acad. Sci. USA. 2007;104:9888–9893. doi: 10.1073/pnas.0702448104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Green CB, Takahashi JS, Bass J. The meter of metabolism. Cell. 2008;134:728–742. doi: 10.1016/j.cell.2008.08.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Griffin EA, Jr, Staknis D, Weitz CJ. Light-independent role of CRY1 and CRY2 in the mammalian circadian clock. Science. 1999;286:768–771. doi: 10.1126/science.286.5440.768. [DOI] [PubMed] [Google Scholar]
  97. Guillaumond F, Dardente H, Giguere V, Cermakian N. Differential control of Bmal1 circadian transcription by REV-ERB and ROR nuclear receptors. J. Biol. Rhythms. 2005;20:391–403. doi: 10.1177/0748730405277232. [DOI] [PubMed] [Google Scholar]
  98. Guo H, Ingolia NT, Weissman JS, Bartel DP. Mammalian microRNAs predominantly act to decrease target mRNA levels. Nature. 2010;466:835–840. doi: 10.1038/nature09267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Hannibal J. Neurotransmitters of the retino-hypothalamic tract. Cell Tissue Res. 2002;309:73–88. doi: 10.1007/s00441-002-0574-3. [DOI] [PubMed] [Google Scholar]
  100. Hannibal J, Hindersson P, Ostergaard J, Georg B, Heegaard S, Larsen PJ, Fahrenkrug J. Melanopsin is expressed in PACAP-containing retinal ganglion cells of the human retinohypothalamic tract. Invest. Ophthalmol. Vis. Sci. 2004;45:4202–4209. doi: 10.1167/iovs.04-0313. [DOI] [PubMed] [Google Scholar]
  101. Harada Y, Sakai M, Kurabayashi N, Hirota T, Fukada Y. Ser-557-phosphorylated mCRY2 is degraded upon synergistic phosphorylation by glycogen synthase kinase-3β. J. Biol. Chem. 2005;280:31714–31721. doi: 10.1074/jbc.M506225200. [DOI] [PubMed] [Google Scholar]
  102. Harmar AJ, Marston HM, Shen S, Spratt C, West KM, Sheward WJ, Morrison CF, Dorin JR, Piggins HD, Reubi JC, et al. The VPAC2 receptor is essential for circadian function in the mouse suprachiasmatic nuclei. Cell. 2002;109:497–508. doi: 10.1016/s0092-8674(02)00736-5. [DOI] [PubMed] [Google Scholar]
  103. Hatcher NG, Atkins N, Jr, Annangudi SP, Forbes AJ, Kelleher NL, Gillette MU, Sweedler JV. Mass spectrometry-based discovery of circadian peptides. Proc. Natl. Acad. Sci. USA. 2008;105:12527–12532. doi: 10.1073/pnas.0804340105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Hatori M, Le H, Vollmers C, Keding SR, Tanaka N, Buch T, Waisman A, Schmedt C, Jegla T, Panda S. Inducible ablation of melanopsin-expressing retinal ganglion cells reveals their central role in non-image forming visual responses. PLoS One. 2008;3:e2451. doi: 10.1371/journal.pone.0002451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Hattar S, Liao HW, Takao M, Berson DM, Yau KW. Melanopsin-containing retinal ganglion cells: architecture, projections, and intrinsic photosensitivity. Science. 2002;295:1065–1070. doi: 10.1126/science.1069609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Hattar S, Lucas RJ, Mrosovsky N, Thompson S, Douglas RH, Hankins MW, Lem J, Biel M, Hofmann F, Foster RG, et al. Melanopsin and rod-cone photoreceptive systems account for all major accessory visual functions in mice. Nature. 2003;424:75–81. doi: 10.1038/nature01761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Hedrich HJ, Bullock G, editors. The Laboratory Mouse. San Diego, CA: Elsevier Academic Press; 2004. [Google Scholar]
  108. Herzog ED, Takahashi JS, Block GD. Clock controls circadian period in isolated suprachiasmatic nucleus neurons. Nat. Neurosci. 1998;1:708–713. doi: 10.1038/3708. [DOI] [PubMed] [Google Scholar]
  109. Herzog ED, Huckfeldt RM. Circadian entrainment to temperature, but not light, in the isolated suprachiasmatic nucleus. J. Neurophysiol. 2003;90:763–770. doi: 10.1152/jn.00129.2003. [DOI] [PubMed] [Google Scholar]
  110. Hirayama J, Sahar S, Grimaldi B, Tamaru T, Takamatsu K, Nakahata Y, Sassone-Corsi P. CLOCK-mediated acetylation of BMAL1 controls circadian function. Nature. 2007;450:1086–1090. doi: 10.1038/nature06394. [DOI] [PubMed] [Google Scholar]
  111. Hirota T, Lewis WG, Liu AC, Lee JW, Schultz PG, Kay SA. A chemical biology approach reveals period shortening of the mammalian circadian clock by specific inhibition of GSK-3β. Proc. Natl. Acad. Sci. USA. 2008;105:20746–20751. doi: 10.1073/pnas.0811410106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Hirota T, Lee JW, Lewis WG, Zhang EE, Breton G, Liu X, Garcia M, Peters EC, Etchegaray JP, Traver D, et al. High-throughput chemical screen identifies a novel potent modulator of cellular circadian rhythms and reveals CKIα as a clock regulatory kinase. PLoS Biol. 2010;8:e1000559. doi: 10.1371/journal.pbio.1000559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Hoeflich KP, Luo J, Rubie EA, Tsao MS, Jin O, Woodgett JR. Requirement for glycogen synthase kinase-3β in cell survival and NF-κB activation. Nature. 2000;406:86–90. doi: 10.1038/35017574. [DOI] [PubMed] [Google Scholar]
  114. Hoffmann K. Die relative Wirksamkeit von Zeitgebern. Oecologia. 1969;3:184–206. doi: 10.1007/BF00416981. [DOI] [PubMed] [Google Scholar]
  115. Hogenesch JB, Gu YZ, Moran SM, Shimomura K, Radcliffe LA, Takahashi JS, Bradfield CA. The basic helix-loop-helix-PAS protein MOP9 is a brain-specific heterodimeric partner of circadian and hypoxia factors. J. Neurosci. 2000;20 doi: 10.1523/JNEUROSCI.20-13-j0002.2000. RC83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Honma K, Honma S. The SCN-independent clocks, methamphetamine and food restriction. Eur. J. Neurosci. 2009;30:1707–1717. doi: 10.1111/j.1460-9568.2009.06976.x. [DOI] [PubMed] [Google Scholar]
  117. Honma S, Shirakawa T, Katsuno Y, Namihira M, Honma K. Circadian periods of single suprachiasmatic neurons in rats. Neurosci. Lett. 1998;250:157–160. doi: 10.1016/s0304-3940(98)00464-9. [DOI] [PubMed] [Google Scholar]
  118. Honma S, Yasuda T, Yasui A, van der Horst GT, Honma K. Circadian behavioral rhythms in Cry1/Cry2 double-deficient mice induced by methamphetamine. J. Biol. Rhythms. 2008;23:91–94. doi: 10.1177/0748730407311124. [DOI] [PubMed] [Google Scholar]
  119. Hughes AT, Guilding C, Lennox L, Samuels RE, McMahon DG, Piggins HD. Live imaging of altered period1 expression in the suprachiasmatic nuclei of Vipr2−/− mice. J. Neurochem. 2008;106:1646–1657. doi: 10.1111/j.1471-4159.2008.05520.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Hughes ME, Hogenesch JB, Kornacker K. JTK_CYCLE: an efficient nonparametric algorithm for detecting rhythmic components in genome-scale data sets. J. Biol. Rhythms. 2010;25:372–380. doi: 10.1177/0748730410379711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Iitaka C, Miyazaki K, Akaike T, Ishida N. A role for glycogen synthase kinase-3β in the mammalian circadian clock. J. Biol. Chem. 2005;280:29397–29402. doi: 10.1074/jbc.M503526200. [DOI] [PubMed] [Google Scholar]
  122. Imhof A, Becker PB. Modifications of the histone N-terminal domains. Evidence for an “epigenetic code”? Mol. Biotechnol. 2001;17:1–13. doi: 10.1385/MB:17:1:01. [DOI] [PubMed] [Google Scholar]
  123. Impey S, McCorkle SR, Cha-Molstad H, Dwyer JM, Yochum GS, Boss JM, McWeeney S, Dunn JJ, Mandel G, Goodman RH. Defining the CREB regulon: a genome-wide analysis of transcription factor regulatory regions. Cell. 2004;119:1041–1054. doi: 10.1016/j.cell.2004.10.032. [DOI] [PubMed] [Google Scholar]
  124. Isojima Y, Nakajima M, Ukai H, Fujishima H, Yamada RG, Masumoto KH, Kiuchi R, Ishida M, Ukai-Tadenuma M, Minami Y, et al. CKIɛ/δ-dependent phosphorylation is a temperature-insensitive, period-determining process in the mammalian circadian clock. Proc. Natl. Acad. Sci. USA. 2009;106:15744–15749. doi: 10.1073/pnas.0908733106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Iwahana E, Akiyama M, Miyakawa K, Uchida A, Kasahara J, Fukunaga K, Hamada T, Shibata S. Effect of lithium on the circadian rhythms of locomotor activity and glycogen synthase kinase-3 protein expression in the mouse suprachiasmatic nuclei. Eur. J. Neurosci. 2004;19:2281–2287. doi: 10.1111/j.0953-816X.2004.03322.x. [DOI] [PubMed] [Google Scholar]
  126. Izumo M, Johnson CH, Yamazaki S. Circadian gene expression in mammalian fibroblasts revealed by real-time luminescence reporting: temperature compensation and damping. Proc. Natl. Acad. Sci. USA. 2003;100:16089–16094. doi: 10.1073/pnas.2536313100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Jacob HJ. A landmark for orphan genomes? Njat. Genet. 1996;13:14–15. doi: 10.1038/ng0596-14. [DOI] [PubMed] [Google Scholar]
  128. Jakubcakova V, Oster H, Tamanini F, Cadenas C, Leitges M, van der Horst GT, Eichele G. Light entrainment of the mammalian circadian clock by a PRKCA-dependent posttranslational mechanism. Neuron. 2007;54:831–843. doi: 10.1016/j.neuron.2007.04.031. [DOI] [PubMed] [Google Scholar]
  129. Jenuwein T, Allis CD. Translating the histone code. Science. 2001;293:1074–1080. doi: 10.1126/science.1063127. [DOI] [PubMed] [Google Scholar]
  130. Jin X, von Gall C, Pieschl RL, Gribkoff VK, Stehle JH, Reppert SM, Weaver DR. Targeted disruption of the mouse Mel1b melatonin receptor. Mol. Cell. Biol. 2003;23:1054–1060. doi: 10.1128/MCB.23.3.1054-1060.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Kaladchibachi SA, Doble B, Anthopoulos N, Woodgett JR, Manoukian AS. Glycogen synthase kinase 3, circadian rhythms, and bipolar disorder: a molecular link in the therapeutic action of lithium. J. Circadian Rhythms. 2007;5:3. doi: 10.1186/1740-3391-5-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Kalamvoki M, Roizman B. Circadian CLOCK histone acetyl transferase localizes at ND10 nuclear bodies and enables herpes simplex virus gene expression. Proc. Natl. Acad. Sci. USA. 2010;107:17721–17726. doi: 10.1073/pnas.1012991107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Keesler GA, Camacho F, Guo Y, Virshup D, Mondadori C, Yao Z. Phosphorylation and destabilization of human period I clock protein by human casein kinase I epsilon. NeuroReport. 2000;11:951–955. doi: 10.1097/00001756-200004070-00011. [DOI] [PubMed] [Google Scholar]
  134. King DP, Vitaterna MH, Chang AM, Dove WF, Pinto LH, Turek FW, Takahashi JS. The mouse Clock mutation behaves as an antimorph and maps within the W 19H deletion, distal of Kit . Genetics. 1997a;146:1049–1060. doi: 10.1093/genetics/146.3.1049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. King DP, Zhao Y, Sangoram AM, Wilsbacher LD, Tanaka M, Antoch MP, Steeves TD, Vitaterna MH, Kornhauser JM, Lowrey PL, et al. Positional cloning of the mouse circadian Clock gene. Cell. 1997b;89:641–653. doi: 10.1016/s0092-8674(00)80245-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Kitayama Y, Nishiwaki T, Terauchi K, Kondo T. Dual KaiC-based oscillations constitute the circadian system of cyanobacteria. Genes Dev. 2008;22:1513–1521. doi: 10.1101/gad.1661808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Klein PS, Melton DA. A molecular mechanism for the effect of lithium on development. Proc. Natl. Acad. Sci. USA. 1996;93:8455–8459. doi: 10.1073/pnas.93.16.8455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Kloss B, Price JL, Saez L, Blau J, Rothenfluh A, Wesley CS, Young MW. The Drosophila clock gene double-time encodes a protein closely related to human casein kinase Iɛ. Cell. 1998;94:97–107. doi: 10.1016/s0092-8674(00)81225-8. [DOI] [PubMed] [Google Scholar]
  139. Knippschild U, Gocht A, Wolff S, Huber N, Lohler J, Stoter M. The casein kinase 1 family: participation in multiple cellular processes in eukaryotes. Cell. Signal. 2005;17:675–689. doi: 10.1016/j.cellsig.2004.12.011. [DOI] [PubMed] [Google Scholar]
  140. Ko CH, Takahashi JS. Molecular components of the mammalian circadian clock. Hum. Mol. Genet. 2006;15 Spec(No 2):R271–R277. doi: 10.1093/hmg/ddl207. [DOI] [PubMed] [Google Scholar]
  141. Ko CH, Yamada YR, Welsh DK, Buhr ED, Liu AC, Zhang EE, Ralph MR, Kay SA, Forger DB, Takahashi JS. Emergence of noise-induced oscillations in the central circadian pacemaker. PLoS Biol. 2010;8:e1000513. doi: 10.1371/journal.pbio.1000513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Kojima S, Matsumoto K, Hirose M, Shimada M, Nagano M, Shigeyoshi Y, Hoshino S, Ui-Tei K, Saigo K, Green CB, et al. LARK activates posttranscriptional expression of an essential mammalian clock protein, PERIOD1. Proc. Natl. Acad. Sci. USA. 2007;104:1859–1864. doi: 10.1073/pnas.0607567104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Kojima S, Gatfield D, Esau CC, Green CB. MicroRNA-122 modulates the rhythmic expression profile of the circadian deadenylase Nocturnin in mouse liver. PLoS One. 2010;5:e11264. doi: 10.1371/journal.pone.0011264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Kojima S, Shingle DL, Green CB. Post-transcriptional control of circadian rhythms. J. Cell Sci. 2011;124:311–320. doi: 10.1242/jcs.065771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Konopka RJ, Benzer S. Clock mutants of Drosophila melanogaster . Proc. Natl. Acad. Sci. USA. 1971;68:2112–2116. doi: 10.1073/pnas.68.9.2112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Kornitzer D, Ciechanover A. Modes of regulation of ubiquitin-mediated protein degradation. J. Cell. Physiol. 2000;182:1–11. doi: 10.1002/(SICI)1097-4652(200001)182:1<1::AID-JCP1>3.0.CO;2-V. [DOI] [PubMed] [Google Scholar]
  147. Kornmann B, Schaad O, Bujard H, Takahashi JS, Schibler U. System-driven and oscillator-dependent circadian transcription in mice with a conditionally active liver clock. PLoS Biol. 2007;5:e34. doi: 10.1371/journal.pbio.0050034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Kramer A, Yang FC, Snodgrass P, Li X, Scammell TE, Davis FC, Weitz CJ. Regulation of daily locomotor activity and sleep by hypothalamic EGF receptor signaling. Science. 2001;294:2511–2515. doi: 10.1126/science.1067716. [DOI] [PubMed] [Google Scholar]
  149. Kramer A, Yang FC, Kraves S, Weitz CJ. A screen for secreted factors of the suprachiasmatic nucleus. Methods Enzymol. 2005;393:645–663. doi: 10.1016/S0076-6879(05)93034-6. [DOI] [PubMed] [Google Scholar]
  150. Kraves S, Weitz CJ. A role for cardiotrophin-like cytokine in the circadian control of mammalian locomotor activity. Nat. Neurosci. 2006;9:212–219. doi: 10.1038/nn1633. [DOI] [PubMed] [Google Scholar]
  151. Krutzfeldt J, Rajewsky N, Braich R, Rajeev KG, Tuschl T, Manoharan M, Stoffel M. Silencing of microRNAs in vivo with 'antagomirs'. Nature. 2005;438:685–689. doi: 10.1038/nature04303. [DOI] [PubMed] [Google Scholar]
  152. Kumaki Y, Ukai-Tadenuma M, Uno KD, Nishio J, Masumoto KH, Nagano M, Komori T, Shigeyoshi Y, Hogenesch JB, Ueda HR. Analysis and synthesis of high-amplitude Cis-elements in the mammalian circadian clock. Proc. Natl. Acad. Sci. USA. 2008;105:14946–14951. doi: 10.1073/pnas.0802636105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Kume K, Zylka MJ, Sriram S, Shearman LP, Weaver DR, Jin X, Maywood ES, Hastings MH, Reppert SM. mCRY1 and mCRY2 are essential components of the negative limb of the circadian clock feedback loop. Cell. 1999;98:193–205. doi: 10.1016/s0092-8674(00)81014-4. [DOI] [PubMed] [Google Scholar]
  154. Kurabayashi N, Hirota T, Sakai M, Sanada K, Fukada Y. DYRK1A and glycogen synthase kinase 3β, a dual-kinase mechanism directing proteasomal degradation of CRY2 for circadian timekeeping. Mol. Cell. Biol. 2010;30:1757–1768. doi: 10.1128/MCB.01047-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Lamia KA, Sachdeva UM, DiTacchio L, Williams EC, Alvarez JG, Egan DF, Vasquez DS, Juguilon H, Panda S, Shaw RJ, et al. AMPK regulates the circadian clock by cryptochrome phosphorylation and degradation. Science. 2009;326:437–440. doi: 10.1126/science.1172156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Le Minh N, Damiola F, Tronche F, Schutz G, Schibler U. Glucocorticoid hormones inhibit food-induced phase-shifting of peripheral circadian oscillators. EMBO J. 2001;20:7128–7136. doi: 10.1093/emboj/20.24.7128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Lee C, Etchegaray JP, Cagampang FR, Loudon AS, Reppert SM. Posttranslational mechanisms regulate the mammalian circadian clock. Cell. 2001;107:855–867. doi: 10.1016/s0092-8674(01)00610-9. [DOI] [PubMed] [Google Scholar]
  158. Lee H, Chen R, Lee Y, Yoo S, Lee C. Essential roles of CKIδ and CKIɛ in the mammalian circadian clock. Proc. Natl. Acad. Sci. USA. 2009;106:21359–21364. doi: 10.1073/pnas.0906651106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Lee J, Lee Y, Lee MJ, Park E, Kang SH, Chung CH, Lee KH, Kim K. Dual modification of BMAL1 by SUMO2/3 and ubiquitin promotes circadian activation of the CLOCK/BMAL1 complex. Mol. Cell. Biol. 2008;28:6056–6065. doi: 10.1128/MCB.00583-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. LeSauter J, Silver R. Lithium lengthens the period of circadian rhythms in lesioned hamsters bearing SCN grafts. Biol. Psychiatry. 1993;34:75–83. doi: 10.1016/0006-3223(93)90259-g. [DOI] [PubMed] [Google Scholar]
  161. Li JD, Hu WP, Boehmer L, Cheng MY, Lee AG, Jilek A, Siegel JM, Zhou QY. Attenuated circadian rhythms in mice lacking the prokineticin 2 gene. J. Neurosci. 2006;26:11615–11623. doi: 10.1523/JNEUROSCI.3679-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Li S, Liu C, Li N, Hao T, Han T, Hill DE, Vidal M, Lin JD. Genome-wide coactivation analysis of PGC-1alpha identifies BAF60a as a regulator of hepatic lipid metabolism. Cell Metab. 2008;8:105–117. doi: 10.1016/j.cmet.2008.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Liu AC, Lewis WG, Kay SA. Mammalian circadian signaling networks and therapeutic targets. Nat. Chem. Biol. 2007a;3:630–639. doi: 10.1038/nchembio.2007.37. [DOI] [PubMed] [Google Scholar]
  164. Liu AC, Welsh DK, Ko CH, Tran HG, Zhang EE, Priest AA, Buhr ED, Singer O, Meeker K, Verma IM, et al. Intercellular coupling confers robustness against mutations in the SCN circadian clock network. Cell. 2007b;129:605–616. doi: 10.1016/j.cell.2007.02.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Liu AC, Tran HG, Zhang EE, Priest AA, Welsh DK, Kay SA. Redundant function of REV-ERBα and β and non-essential role for Bmal1 cycling in transcriptional regulation of intracellular circadian rhythms. PLoS Genet. 2008;4:e1000023. doi: 10.1371/journal.pgen.1000023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Liu C, Weaver DR, Jin X, Shearman LP, Pieschl RL, Gribkoff VK, Reppert SM. Molecular dissection of two distinct actions of melatonin on the suprachiasmatic circadian clock. Neuron. 1997a;19:91–102. doi: 10.1016/s0896-6273(00)80350-5. [DOI] [PubMed] [Google Scholar]
  167. Liu C, Weaver DR, Strogatz SH, Reppert SM. Cellular construction of a circadian clock: period determination in the suprachiasmatic nuclei. Cell. 1997b;91:855–860. doi: 10.1016/s0092-8674(00)80473-0. [DOI] [PubMed] [Google Scholar]
  168. Liu C, Li S, Liu T, Borjigin J, Lin JD. Transcriptional coactivator PGC-1α integrates the mammalian clock and energy metabolism. Nature. 2007c;447:477–481. doi: 10.1038/nature05767. [DOI] [PubMed] [Google Scholar]
  169. Lopez-Molina L, Conquet F, Dubois-Dauphin M, Schibler U. The DBP gene is expressed according to a circadian rhythm in the suprachiasmatic nucleus and influences circadian behavior. EMBO J. 1997;16:6762–6771. doi: 10.1093/emboj/16.22.6762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Lowrey PL, Shimomura K, Antoch MP, Yamazaki S, Zemenides PD, Ralph MR, Menaker M, Takahashi JS. Positional syntenic cloning and functional characterization of the mammalian circadian mutation tau . Science. 2000;288:483–492. doi: 10.1126/science.288.5465.483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Lowrey PL, Takahashi JS. Genetics of the mammalian circadian system: Photic entrainment, circadian pacemaker mechanisms, and posttranslational regulation. Annu. Rev. Genet. 2000;34:533–562. doi: 10.1146/annurev.genet.34.1.533. [DOI] [PubMed] [Google Scholar]
  172. Lowrey PL, Takahashi JS. Mammalian circadian biology: elucidating genome-wide levels of temporal organization. Annu. Rev. Genom. Hum. Genet. 2004;5:407–441. doi: 10.1146/annurev.genom.5.061903.175925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Lucas RJ, Hattar S, Takao M, Berson DM, Foster RG, Yau KW. Diminished pupillary light reflex at high irradiances in melanopsin-knockout mice. Science. 2003;299:245–247. doi: 10.1126/science.1077293. [DOI] [PubMed] [Google Scholar]
  174. MacAulay K, Doble BW, Patel S, Hansotia T, Sinclair EM, Drucker DJ, Nagy A, Woodgett JR. Glycogen synthase kinase 3α-specific regulation of murine hepatic glycogen metabolism. Cell Metab. 2007;6:329–337. doi: 10.1016/j.cmet.2007.08.013. [DOI] [PubMed] [Google Scholar]
  175. Maier B, Wendt S, Vanselow JT, Wallach T, Reischl S, Oehmke S, Schlosser A, Kramer A. A large-scale functional RNAi screen reveals a role for CK2 in the mammalian circadian clock. Genes Dev. 2009;23:708–718. doi: 10.1101/gad.512209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Martinek S, Inonog S, Manoukian AS, Young MW. A role for the segment polarity gene shaggy/GSK-3 in the Drosophila circadian clock. Cell. 2001;105:769–779. doi: 10.1016/s0092-8674(01)00383-x. [DOI] [PubMed] [Google Scholar]
  177. Masana MI, Sumaya IC, Becker-Andre M, Dubocovich ML. Behavioral characterization and modulation of circadian rhythms by light and melatonin in C3H/HeN mice homozygous for the RORβ knockout. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2007;292:R2357–R2367. doi: 10.1152/ajpregu.00687.2006. [DOI] [PubMed] [Google Scholar]
  178. Maury E, Ramsey KM, Bass J. Circadian rhythms and metabolic syndrome: from experimental genetics to human disease. Circ. Res. 2010;106:447–462. doi: 10.1161/CIRCRESAHA.109.208355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Maywood ES, Reddy AB, Wong GK, O'Neill JS, O'Brien JA, McMahon DG, Harmar AJ, Okamura H, Hastings MH. Synchronization and maintenance of timekeeping in suprachiasmatic circadian clock cells by neuropeptidergic signaling. Curr. Biol. 2006;16:599–605. doi: 10.1016/j.cub.2006.02.023. [DOI] [PubMed] [Google Scholar]
  180. McDearmon EL, Patel KN, Ko CH, Walisser JA, Schook AC, Chong JL, Wilsbacher LD, Song EJ, Hong HK, Bradfield CA, et al. Dissecting the functions of the mammalian clock protein BMAL1 by tissue-specific rescue in mice. Science. 2006;314:1304–1308. doi: 10.1126/science.1132430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Meggio F, Pinna LA. One-thousand-and-one substrates of protein kinase CK2? FASEB J. 2003;17:349–368. doi: 10.1096/fj.02-0473rev. [DOI] [PubMed] [Google Scholar]
  182. Melyan Z, Tarttelin EE, Bellingham J, Lucas RJ, Hankins MW. Addition of human melanopsin renders mammalian cells photoresponsive. Nature. 2005;433:741–745. doi: 10.1038/nature03344. [DOI] [PubMed] [Google Scholar]
  183. Meng QJ, Logunova L, Maywood ES, Gallego M, Lebiecki J, Brown TM, Sladek M, Semikhodskii AS, Glossop NR, Piggins HD, et al. Setting clock speed in mammals: the CK1ɛ tau mutation in mice accelerates circadian pacemakers by selectively destabilizing PERIOD proteins. Neuron. 2008;58:78–88. doi: 10.1016/j.neuron.2008.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Meng QJ, Maywood ES, Bechtold DA, Lu WQ, Li J, Gibbs JE, Dupre SM, Chesham JE, Rajamohan F, Knafels J, et al. Entrainment of disrupted circadian behavior through inhibition of casein kinase 1 (CK1) enzymes. Proc. Natl. Acad. Sci. USA. 2010;107:15240–15245. doi: 10.1073/pnas.1005101107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Michel S, Itri J, Han JH, Gniotczynski K, Colwell CS. Regulation of glutamatergic signalling by PACAP in the mammalian suprachiasmatic nucleus. BMC Neurosci. 2006;7:15. doi: 10.1186/1471-2202-7-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Miller BH, McDearmon EL, Panda S, Hayes KR, Zhang J, Andrews JL, Antoch MP, Walker JR, Esser KA, Hogenesch JB, et al. Circadian and CLOCK-controlled regulation of the mouse transcriptome and cell proliferation. Proc. Natl. Acad. Sci. USA. 2007;104:3342–3347. doi: 10.1073/pnas.0611724104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Mitsui S, Yamaguchi S, Matsuo T, Ishida Y, Okamura H. Antagonistic role of E4BP4 and PAR proteins in the circadian oscillatory mechanism. Genes Dev. 2001;15:995–1006. doi: 10.1101/gad.873501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Mohawk JA, Baer ML, Menaker M. The methamphetamine-sensitive circadian oscillator does not employ canonical clock genes. Proc. Natl. Acad. Sci. USA. 2009;106:3519–3524. doi: 10.1073/pnas.0813366106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Moore MJ. From birth to death: the complex lives of eukaryotic mRNAs. Science. 2005;309:1514–1518. doi: 10.1126/science.1111443. [DOI] [PubMed] [Google Scholar]
  190. Moore RY, Eichler VB. Loss of a circadian adrenal corticosterone rhythm following suprachiasmatic lesions in the rat. Brain Res. 1972;42:201–206. doi: 10.1016/0006-8993(72)90054-6. [DOI] [PubMed] [Google Scholar]
  191. Moore RY, Lenn NJ. A retinohypothalamic projection in the rat. J. Comp. Neurol. 1972;146:1–14. doi: 10.1002/cne.901460102. [DOI] [PubMed] [Google Scholar]
  192. Moore RY, Speh JC, Card JP. The retinohypothalamic tract originates from a distinct subset of retinal ganglion cells. J. Comp. Neurol. 1995;352:351–366. doi: 10.1002/cne.903520304. [DOI] [PubMed] [Google Scholar]
  193. Morin LP, Allen CN. The circadian visual system, 2005. Brain Res. Rev. 2006;51:1–60. doi: 10.1016/j.brainresrev.2005.08.003. [DOI] [PubMed] [Google Scholar]
  194. Muller B, Grossniklaus U. Model organisms--A historical perspective. J. Proteomics. 2010;73:2054–2063. doi: 10.1016/j.jprot.2010.08.002. [DOI] [PubMed] [Google Scholar]
  195. Nader N, Chrousos GP, Kino T. Circadian rhythm transcription factor CLOCK regulates the transcriptional activity of the glucocorticoid receptor by acetylating its hinge region lysine cluster: potential physiological implications. FASEB J. 2009;23:1572–1583. doi: 10.1096/fj.08-117697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Nagel R, Clijsters L, Agami R. The miRNA-192/194 cluster regulates the Period gene family and the circadian clock. FEBS J. 2009;276:5447–5455. doi: 10.1111/j.1742-4658.2009.07229.x. [DOI] [PubMed] [Google Scholar]
  197. Nagoshi E, Saini C, Bauer C, Laroche T, Naef F, Schibler U. Circadian gene expression in individual fibroblasts; cell-autonomous and self-sustained oscillators pass time to daughter cells. Cell. 2004;119:693–705. doi: 10.1016/j.cell.2004.11.015. [DOI] [PubMed] [Google Scholar]
  198. Nagy A, Gertsenstein M, Vintersten K, Behringer R, editors. Manipulating the Mouse Embryo: A Laboratory Manual. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press; 2003. [Google Scholar]
  199. Nakahata Y, Kaluzova M, Grimaldi B, Sahar S, Hirayama J, Chen D, Guarente LP, Sassone-Corsi P. The NAD+-dependent deacetylase SIRT1 modulates CLOCK-mediated chromatin remodeling and circadian control. Cell. 2008;134:329–340. doi: 10.1016/j.cell.2008.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Nakahata Y, Sahar S, Astarita G, Kaluzova M, Sassone-Corsi P. Circadian control of the NAD+ salvage pathway by CLOCK-SIRT1. Science. 2009;324:654–657. doi: 10.1126/science.1170803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  201. Nakajima M, Imai K, Ito H, Nishiwaki T, Murayama Y, Iwasaki H, Oyama T, Kondo T. Reconstitution of circadian oscillation of cyanobacterial KaiC phosphorylation in vitro. Science. 2005;308:414–415. doi: 10.1126/science.1108451. [DOI] [PubMed] [Google Scholar]
  202. Nakashima A, Kawamoto T, Honda KK, Ueshima T, Noshiro M, Iwata T, Fujimoto K, Kubo H, Honma S, Yorioka N, et al. DEC1 modulates the circadian phase of clock gene expression. Mol. Cell. Biol. 2008;28:4080–4092. doi: 10.1128/MCB.02168-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  203. Nandi D, Tahiliani P, Kumar A, Chandu D. The ubiquitin-proteasome system. J. Biosci. 2006;31:137–155. doi: 10.1007/BF02705243. [DOI] [PubMed] [Google Scholar]
  204. Nelson RJ, Zucker I. Absence of extraocular photoreception in diurnal and nocturnal rodents exposed to direct sunlight. Comp. Biochem. Physiol. 1981;69A:145–148. [Google Scholar]
  205. O'Neill JS, Reddy AB. Circadian clocks in human red blood cells. Nature. 2011;469:498–503. doi: 10.1038/nature09702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  206. O'Neill JS, van Ooijen G, Dixon LE, Troein C, Corellou F, Bouget FY, Reddy AB, Millar AJ. Circadian rhythms persist without transcription in a eukaryote. Nature. 2011;469:554–558. doi: 10.1038/nature09654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Ohno T, Onishi Y, Ishida N. A novel E4BP4 element drives circadian expression of mPeriod2 . Nucleic Acids Res. 2007;35:648–655. doi: 10.1093/nar/gkl868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. Ohsaki K, Oishi K, Kozono Y, Nakayama K, Nakayama KI, Ishida N. The role of β-TrCP1 and β-TrCP2 in circadian rhythm generation by mediating degradation of clock protein PER2. J. Biochem. Tokyo. 2008;144:609–618. doi: 10.1093/jb/mvn112. [DOI] [PubMed] [Google Scholar]
  209. Oishi K, Miyazaki K, Kadota K, Kikuno R, Nagase T, Atsumi G, Ohkura N, Azama T, Mesaki M, Yukimasa S, et al. Genome-wide expression analysis of mouse liver reveals CLOCK-regulated circadian output genes. J. Biol. Chem. 2003;278:41519–41527. doi: 10.1074/jbc.M304564200. [DOI] [PubMed] [Google Scholar]
  210. Okamura H, Miyake S, Sumi Y, Yamaguchi S, Yasui A, Muijtjens M, Hoeijmakers JH, van der Horst GT. Photic induction of mPer1 and mPer2 in Cry-deficient mice lacking a biological clock. Science. 1999;286:2531–2534. doi: 10.1126/science.286.5449.2531. [DOI] [PubMed] [Google Scholar]
  211. Oster H, Yasui A, van der Horst GT, Albrecht U. Disruption of mCry2 restores circadian rhythmicity in mPer2 mutant mice. Genes Dev. 2002;16:2633–2638. doi: 10.1101/gad.233702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Oster H, Baeriswyl S, Van Der Horst GT, Albrecht U. Loss of circadian rhythmicity in aging mPer1−/− mCry2−/− mutant mice. Genes Dev. 2003;17:1366–1379. doi: 10.1101/gad.256103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Palkova M, Sigmund L, Erkert HG. Effect of ambient temperature on the circadian activity rhythm in common marmosets, Callithrix j. jacchus (primates) Chronobiol. Int. 1999;16:149–161. doi: 10.3109/07420529909019082. [DOI] [PubMed] [Google Scholar]
  214. Panda S, Antoch MP, Miller BH, Su AI, Schook AB, Straume M, Schultz PG, Kay SA, Takahashi JS, Hogenesch JB. Coordinated transcription of key pathways in the mouse by the circadian clock. Cell. 2002a;109:307–320. doi: 10.1016/s0092-8674(02)00722-5. [DOI] [PubMed] [Google Scholar]
  215. Panda S, Sato TK, Castrucci AM, Rollag MD, DeGrip WJ, Hogenesch JB, Provencio I, Kay SA. Melanopsin (Opn4) requirement for normal light-induced circadian phase shifting. Science. 2002b;298:2213–2216. doi: 10.1126/science.1076848. [DOI] [PubMed] [Google Scholar]
  216. Panda S, Provencio I, Tu DC, Pires SS, Rollag MD, Castrucci AM, Pletcher MT, Sato TK, Wiltshire T, Andahazy M, et al. Melanopsin is required for non-image-forming photic responses in blind mice. Science. 2003;301:525–527. doi: 10.1126/science.1086179. [DOI] [PubMed] [Google Scholar]
  217. Panda S, Nayak SK, Campo B, Walker JR, Hogenesch JB, Jegla T. Illumination of the melanopsin signaling pathway. Science. 2005;307:600–604. doi: 10.1126/science.1105121. [DOI] [PubMed] [Google Scholar]
  218. Partch CL, Shields KF, Thompson CL, Selby CP, Sancar A. Posttranslational regulation of the mammalian circadian clock by cryptochrome and protein phosphatase 5. Proc. Natl. Acad. Sci. USA. 2006;103:10467–10472. doi: 10.1073/pnas.0604138103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  219. Pezuk P, Mohawk JA, Yoshikawa T, Sellix MT, Menaker M. Circadian organization is governed by extra-SCN pacemakers. J. Biol. Rhythms. 2010;25:432–441. doi: 10.1177/0748730410385204. [DOI] [PubMed] [Google Scholar]
  220. Pittendrigh CS. Temporal organization: reflections of a Darwinian clock-watcher. Annu. Rev. Physiol. 1993;55:16–54. doi: 10.1146/annurev.ph.55.030193.000313. [DOI] [PubMed] [Google Scholar]
  221. Preitner N, Damiola F, Lopez-Molina L, Zakany J, Duboule D, Albrecht U, Schibler U. The orphan nuclear receptor REV-ERBα controls circadian transcription within the positive limb of the mammalian circadian oscillator. Cell. 2002;110:251–260. doi: 10.1016/s0092-8674(02)00825-5. [DOI] [PubMed] [Google Scholar]
  222. Price JL, Blau J, Rothenfluh A, Abodeely M, Kloss B, Young MW. double-time is a novel Drosophila clock gene that regulates PERIOD protein accumulation. Cell. 1998;94:83–95. doi: 10.1016/s0092-8674(00)81224-6. [DOI] [PubMed] [Google Scholar]
  223. Prolo LM, Takahashi JS, Herzog ED. Circadian rhythm generation and entrainment in astrocytes. J. Neurosci. 2005;25:404–408. doi: 10.1523/JNEUROSCI.4133-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  224. Provencio I, Jiang G, De Grip WJ, Hayes WP, Rollag MD. Melanopsin: An opsin in melanophores, brain, and eye. Proc. Natl. Acad. Sci. USA. 1998;95:340–345. doi: 10.1073/pnas.95.1.340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  225. Qiu X, Kumbalasiri T, Carlson SM, Wong KY, Krishna V, Provencio I, Berson DM. Induction of photosensitivity by heterologous expression of melanopsin. Nature. 2005;433:745–749. doi: 10.1038/nature03345. [DOI] [PubMed] [Google Scholar]
  226. Quintero JE, Kuhlman SJ, McMahon DG. The biological clock nucleus: a multiphasic oscillator network regulated by light. J. Neurosci. 2003;23:8070–8076. doi: 10.1523/JNEUROSCI.23-22-08070.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  227. Ralph MR, Menaker M. A mutation of the circadian system in golden hamsters. Science. 1988;241:1225–1227. doi: 10.1126/science.3413487. [DOI] [PubMed] [Google Scholar]
  228. Ralph MR, Foster RG, Davis FC, Menaker M. Transplanted suprachiasmatic nucleus determines circadian period. Science. 1990;247:975–978. doi: 10.1126/science.2305266. [DOI] [PubMed] [Google Scholar]
  229. Ramsey KM, Yoshino J, Brace CS, Abrassart D, Kobayashi Y, Marcheva B, Hong HK, Chong JL, Buhr ED, Lee C, et al. Circadian clock feedback cycle through NAMPT-mediated NAD+ biosynthesis. Science. 2009;324:651–654. doi: 10.1126/science.1171641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  230. Rana TM. Illuminating the silence: understanding the structure and function of small RNAs. Nat. Rev. Mol. Cell Biol. 2007;8:23–36. doi: 10.1038/nrm2085. [DOI] [PubMed] [Google Scholar]
  231. Reddy AB, Karp NA, Maywood ES, Sage EA, Deery M, O'Neill JS, Wong GK, Chesham J, Odell M, Lilley KS, et al. Circadian orchestration of the hepatic proteome. Curr. Biol. 2006;16:1107–1115. doi: 10.1016/j.cub.2006.04.026. [DOI] [PubMed] [Google Scholar]
  232. Refinetti R, Menaker M. The circadian rhythm of body temperature. Physiol. Behav. 1992;51:613–637. doi: 10.1016/0031-9384(92)90188-8. [DOI] [PubMed] [Google Scholar]
  233. Reick M, Garcia JA, Dudley C, McKnight SL. NPAS2: an analog of Clock operative in the mammalian forebrain. Science. 2001;293:506–509. doi: 10.1126/science.1060699. [DOI] [PubMed] [Google Scholar]
  234. Reinke H, Saini C, Fleury-Olela F, Dibner C, Benjamin IJ, Schibler U. Differential display of DNA-binding proteins reveals heat-shock factor 1 as a circadian transcription factor. Genes Dev. 2008;22:331–345. doi: 10.1101/gad.453808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  235. Reischl S, Vanselow K, Westermark PO, Thierfelder N, Maier B, Herzel H, Kramer A. Beta-TrCP1-mediated degradation of PERIOD2 is essential for circadian dynamics. J. Biol. Rhythms. 2007;22:375–386. doi: 10.1177/0748730407303926. [DOI] [PubMed] [Google Scholar]
  236. Reppert SM. A clockwork explosion! Neuron. 1998;21:1–4. doi: 10.1016/s0896-6273(00)80234-2. [DOI] [PubMed] [Google Scholar]
  237. Reppert SM, Weaver DR. Coordination of circadian timing in mammals. Nature. 2002;418:935–941. doi: 10.1038/nature00965. [DOI] [PubMed] [Google Scholar]
  238. Reyes BA, Pendergast JS, Yamazaki S. Mammalian peripheral circadian oscillators are temperature compensated. J. Biol. Rhythms. 2008;23:95–98. doi: 10.1177/0748730407311855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Ripperger JA, Merrow M. Perfect timing: Epigenetic regulation of the circadian clock. FEBS Lett. 2011;585:1406–1411. doi: 10.1016/j.febslet.2011.04.047. [DOI] [PubMed] [Google Scholar]
  240. Rivers A, Gietzen KF, Vielhaber E, Virshup DM. Regulation of casein kinase I ɛ and casein kinase I δ by an in vivo futile phosphorylation cycle. J. Biol. Chem. 1998;273:15980–15984. doi: 10.1074/jbc.273.26.15980. [DOI] [PubMed] [Google Scholar]
  241. Robles MS, Boyault C, Knutti D, Padmanabhan K, Weitz CJ. Identification of RACK1 and protein kinase Cα as integral components of the mammalian circadian clock. Science. 2010;327:463–466. doi: 10.1126/science.1180067. [DOI] [PubMed] [Google Scholar]
  242. Rossner MJ, Oster H, Wichert SP, Reinecke L, Wehr MC, Reinecke J, Eichele G, Taneja R, Nave KA. Disturbed clockwork resetting in Sharp-1 and Sharp-2 single and double mutant mice. PLoS One. 2008;3:e2762. doi: 10.1371/journal.pone.0002762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  243. Ruby NF, Heller HC. Temperature sensitivity of the suprachiasmatic nucleus of ground squirrels and rats in vitro. J. Biol. Rhythms. 1996;11:126–136. doi: 10.1177/074873049601100205. [DOI] [PubMed] [Google Scholar]
  244. Ruby NF, Burns DE, Heller HC. Circadian rhythms in the suprachiasmatic nucleus are temperature-compensated and phase-shifted by heat pulses in vitro . J. Neurosci. 1999;19:8630–8636. doi: 10.1523/JNEUROSCI.19-19-08630.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  245. Ruby NF, Brennan TJ, Xie X, Cao V, Franken P, Heller HC, O'Hara BF. Role of melanopsin in circadian responses to light. Science. 2002;298:2211–2213. doi: 10.1126/science.1076701. [DOI] [PubMed] [Google Scholar]
  246. Rutter J, Reick M, Wu LC, McKnight SL. Regulation of Clock and NPAS2 DNA binding by the redox state of NAD cofactors. Science. 2001;293:510–514. doi: 10.1126/science.1060698. [DOI] [PubMed] [Google Scholar]
  247. Rutter J, Reick M, McKnight SL. Metabolism and the control of circadian rhythms. Annu. Rev. Biochem. 2002;71:307–331. doi: 10.1146/annurev.biochem.71.090501.142857. [DOI] [PubMed] [Google Scholar]
  248. Ryves WJ, Harwood AJ. Lithium inhibits glycogen synthase kinase-3 by competition for magnesium. Biochem. Biophys. Res. Commun. 2001;280:720–725. doi: 10.1006/bbrc.2000.4169. [DOI] [PubMed] [Google Scholar]
  249. Sahar S, Zocchi L, Kinoshita C, Borrelli E, Sassone-Corsi P. Regulation of BMAL1 protein stability and circadian function by GSK3β-mediated phosphorylation. PLoS One. 2010;5:e8561. doi: 10.1371/journal.pone.0008561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  250. Sanada K, Okano T, Fukada Y. Mitogen-activated protein kinase phosphorylates and negatively regulates basic helix-loop-helix-PAS transcription factor BMAL1. J. Biol. Chem. 2002;277:267–271. doi: 10.1074/jbc.M107850200. [DOI] [PubMed] [Google Scholar]
  251. Sasaki M, Yoshitane H, Du NH, Okano T, Fukada Y. Preferential inhibition of BMAL2-CLOCK activity by PER2 reemphasizes its negative role and a positive role of BMAL2 in the circadian transcription. J. Biol. Chem. 2009;284:25149–25159. doi: 10.1074/jbc.M109.040758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  252. Sato TK, Panda S, Miraglia LJ, Reyes TM, Rudic RD, McNamara P, Naik KA, FitzGerald GA, Kay SA, Hogenesch JB. A functional genomics strategy reveals Rora as a component of the mammalian circadian clock. Neuron. 2004;43:527–537. doi: 10.1016/j.neuron.2004.07.018. [DOI] [PubMed] [Google Scholar]
  253. Sato TK, Yamada RG, Ukai H, Baggs JE, Miraglia LJ, Kobayashi TJ, Welsh DK, Kay SA, Ueda HR, Hogenesch JB. Feedback repression is required for mammalian circadian clock function. Nat. Genet. 2006;38:312–319. doi: 10.1038/ng1745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  254. Seggie J, Werstiuk ES, Grota L. Effect of chronic lithium treatment on twenty four hour variation in plasma and red blood cell lithium and sodium concentrations, drinking behavior, body weight, kidney weight, and corticosterone levels. Prog. Neuro-Psychopharmacol. Biol. Psychiatry. 1982;6:455–458. doi: 10.1016/s0278-5846(82)80127-9. [DOI] [PubMed] [Google Scholar]
  255. Shearman LP, Zylka MJ, Weaver DR, Kolakowski LF, Jr, Reppert SM. Two period homologs: circadian expression and photic regulation in the suprachiasmatic nuclei. Neuron. 1997;19:1261–1269. doi: 10.1016/s0896-6273(00)80417-1. [DOI] [PubMed] [Google Scholar]
  256. Shearman LP, Jin X, Lee C, Reppert SM, Weaver DR. Targeted disruption of the mPer3 gene: subtle effects on circadian clock function. Mol. Cell. Biol. 2000a;20:6269–6275. doi: 10.1128/mcb.20.17.6269-6275.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  257. Shearman LP, Sriram S, Weaver DR, Maywood ES, Chaves I, Zheng B, Kume K, Lee CC, van der Horst GT, Hastings MH, et al. Interacting molecular loops in the mammalian circadian clock. Science. 2000b;288:1013–1019. doi: 10.1126/science.288.5468.1013. [DOI] [PubMed] [Google Scholar]
  258. Shi S, Hida A, McGuinness OP, Wasserman DH, Yamazaki S, Johnson CH. Circadian clock gene Bmal1 is not essential; functional replacement with its paralog, Bmal2 . Curr. Biol. 2010;20:316–321. doi: 10.1016/j.cub.2009.12.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  259. Shigeyoshi Y, Taguchi K, Yamamoto S, Takekida S, Yan L, Tei H, Moriya T, Shibata S, Loros JJ, Dunlap JC, et al. Light-induced resetting of a mammalian circadian clock is associated with rapid induction of the mPer1 transcript. Cell. 1997;91:1043–1053. doi: 10.1016/s0092-8674(00)80494-8. [DOI] [PubMed] [Google Scholar]
  260. Shim HS, Kim H, Lee J, Son GH, Cho S, Oh TH, Kang SH, Seen DS, Lee KH, Kim K. Rapid activation of CLOCK by Ca2+-dependent protein kinase C mediates resetting of the mammalian circadian clock. EMBO Rep. 2007;8:366–371. doi: 10.1038/sj.embor.7400920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  261. Shirogane T, Jin J, Ang XL, Harper JW. SCFβ-TRCP controls clock-dependent transcription via casein kinase 1-dependent degradation of the mammalian period-1 (Per1) protein. J. Biol. Chem. 2005;280:26863–26872. doi: 10.1074/jbc.M502862200. [DOI] [PubMed] [Google Scholar]
  262. Siepka SM, Takahashi JS. Forward genetic screens to identify circadian rhythm mutants in mice. Methods Enzymol. 2005;393:219–229. doi: 10.1016/S0076-6879(05)93007-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  263. Siepka SM, Yoo SH, Park J, Song W, Kumar V, Hu Y, Lee C, Takahashi JS. Circadian mutant Overtime reveals F-box protein FBXL3 regulation of Cryptochrome and Period gene expression. Cell. 2007;129:1011–1023. doi: 10.1016/j.cell.2007.04.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  264. Silver LM. "Mouse Genetics: Concepts and Applications.". New York, NY: Oxford University Press; 1995. [Google Scholar]
  265. Silver R, LeSauter J, Tresco PA, Lehman MN. A diffusible coupling signal from the transplanted suprachiasmatic nucleus controlling circadian locomotor rhythms. Nature. 1996;382:810–813. doi: 10.1038/382810a0. [DOI] [PubMed] [Google Scholar]
  266. Spengler ML, Kuropatwinski KK, Schumer M, Antoch MP. A serine cluster mediates BMAL1-dependent CLOCK phosphorylation and degradation. Cell Cycle. 2009;8:4138–4146. doi: 10.4161/cc.8.24.10273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  267. Sprouse J, Reynolds L, Kleiman R, Tate B, Swanson TA, Pickard GE. Chronic treatment with a selective inhibitor of casein kinase I δ/ɛ yields cumulative phase delays in circadian rhythms. Psychopharmacology (Berl) 2010;210:569–576. doi: 10.1007/s00213-010-1860-5. [DOI] [PubMed] [Google Scholar]
  268. Staiger D, Koster T. Spotlight on post-transcriptional control in the circadian system. Cell. Mol. Life Sci. 2011;68:71–83. doi: 10.1007/s00018-010-0513-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  269. Stambolic V, Ruel L, Woodgett JR. Lithium inhibits glycogen synthase kinase-3 activity and mimics wingless signalling in intact cells. Curr. Biol. 1996;6:1664–1668. doi: 10.1016/s0960-9822(02)70790-2. [DOI] [PubMed] [Google Scholar]
  270. Stephan FK, Zucker I. Circadian rhythms in drinking behavior and locomotor activity of rats are eliminated by hypothalamic lesions. Proc. Natl. Acad. Sci. USA. 1972;69:1583–1586. doi: 10.1073/pnas.69.6.1583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  271. Stokkan KA, Yamazaki S, Tei H, Sakaki Y, Menaker M. Entrainment of the circadian clock in the liver by feeding. Science. 2001;291:490–493. doi: 10.1126/science.291.5503.490. [DOI] [PubMed] [Google Scholar]
  272. Storch KF, Lipan O, Leykin I, Viswanathan N, Davis FC, Wong WH, Weitz CJ. Extensive and divergent circadian gene expression in liver and heart. Nature. 2002;417:78–83. doi: 10.1038/nature744. [DOI] [PubMed] [Google Scholar]
  273. Storch KF, Weitz CJ. Daily rhythms of food-anticipatory behavioral activity do not require the known circadian clock. Proc. Natl. Acad. Sci. USA. 2009;106:6808–6813. doi: 10.1073/pnas.0902063106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  274. Strahl BD, Allis CD. The language of covalent histone modifications. Nature. 2000;403:41–45. doi: 10.1038/47412. [DOI] [PubMed] [Google Scholar]
  275. Sweeney BM, Hastings JW. Effects of temperature upon diurnal rhythms. Cold Spring Harbor Symp. Quant. Biol. 1960;25:87–104. doi: 10.1101/sqb.1960.025.01.009. [DOI] [PubMed] [Google Scholar]
  276. Takahashi JS, Pinto LH, Vitaterna MH. Forward and reverse genetic approaches to behavior in the mouse. Science. 1994;264:1724–1733. doi: 10.1126/science.8209253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  277. Takahashi JS, Turek FW, Moore RY, editors. Handbook of Behavioral Neurobiology: Circadian Clocks. Vol. 12. New York: Kluwer Acad./Plenum Publ.; 2001. [Google Scholar]
  278. Takahashi JS, Hong HK, Ko CH, McDearmon EL. The genetics of mammalian circadian order and disorder: implications for physiology and disease. Nat. Rev. Genet. 2008;9:764–775. doi: 10.1038/nrg2430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  279. Tamaru T, Hirayama J, Isojima Y, Nagai K, Norioka S, Takamatsu K, Sassone-Corsi P. CK2α phosphorylates BMAL1 to regulate the mammalian clock. Nat. Struct. Mol. Biol. 2009;16:446–448. doi: 10.1038/nsmb.1578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  280. Thresher RJ, Vitaterna MH, Miyamoto Y, Kazantsev A, Hsu DS, Petit C, Selby CP, Dawut L, Smithies O, Takahashi JS, et al. Role of mouse cryptochrome blue-light photoreceptor in circadian photoresponses. Science. 1998;282:1490–1494. doi: 10.1126/science.282.5393.1490. [DOI] [PubMed] [Google Scholar]
  281. Tischkau SA, Mitchell JW, Pace LA, Barnes JW, Barnes JA, Gillette MU. Protein kinase G type II is required for night-to-day progression of the mammalian circadian clock. Neuron. 2004;43:539–549. doi: 10.1016/j.neuron.2004.07.027. [DOI] [PubMed] [Google Scholar]
  282. Tomita J, Nakajima M, Kondo T, Iwasaki H. No transcription-translation feedback in circadian rhythm of KaiC phosphorylation. Science. 2005;307:251–254. doi: 10.1126/science.1102540. [DOI] [PubMed] [Google Scholar]
  283. Tosini G, Menaker M. Circadian rhythms in cultured mammalian retina. Science. 1996;272:419–421. doi: 10.1126/science.272.5260.419. [DOI] [PubMed] [Google Scholar]
  284. Tosini G, Menaker M. The tau mutation affects temperature compensation of hamster retinal circadian oscillators. NeuroReport. 1998;9:1001–1005. doi: 10.1097/00001756-199804200-00009. [DOI] [PubMed] [Google Scholar]
  285. Travnickova-Bendova Z, Cermakian N, Reppert SM, Sassone-Corsi P. Bimodal regulation of mPeriod promoters by CREB-dependent signaling and CLOCK/BMAL1 activity. Proc. Natl. Acad. Sci. USA. 2002;99:7728–7733. doi: 10.1073/pnas.102075599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  286. Triqueneaux G, Thenot S, Kakizawa T, Antoch MP, Safi R, Takahashi JS, Delaunay F, Laudet V. The orphan receptor Rev-erbα gene is a target of the circadian clock pacemaker. J. Mol. Endocrinol. 2004;33:585–608. doi: 10.1677/jme.1.01554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  287. Tsuchiya Y, Akashi M, Nishida E. Temperature compensation and temperature resetting of circadian rhythms in mammalian cultured fibroblasts. Genes Cells. 2003;8:713–720. doi: 10.1046/j.1365-2443.2003.00669.x. [DOI] [PubMed] [Google Scholar]
  288. Tsuchiya Y, Akashi M, Matsuda M, Goto K, Miyata Y, Node K, Nishida E. Involvement of the protein kinase CK2 in the regulation of mammalian circadian rhythms. Sci. Signal. 2009;2 doi: 10.1126/scisignal.2000305. ra26. [DOI] [PubMed] [Google Scholar]
  289. Ueda HR, Hayashi S, Chen W, Sano M, Machida M, Shigeyoshi Y, Iino M, Hashimoto S. System-level identification of transcriptional circuits underlying mammalian circadian clocks. Nat. Genet. 2005;37:187–192. doi: 10.1038/ng1504. [DOI] [PubMed] [Google Scholar]
  290. Ukai-Tadenuma M, Kasukawa T, Ueda HR. Proof-by-synthesis of the transcriptional logic of mammalian circadian clocks. Nat. Cell Biol. 2008;10:1154–1163. doi: 10.1038/ncb1775. [DOI] [PubMed] [Google Scholar]
  291. van der Horst GT, Muijtjens M, Kobayashi K, Takano R, Kanno S, Takao M, de Wit J, Verkerk A, Eker AP, van Leenen D, et al. Mammalian Cry1 and Cry2 are essential for maintenance of circadian rhythms. Nature. 1999;398:627–630. doi: 10.1038/19323. [DOI] [PubMed] [Google Scholar]
  292. Vanselow K, Vanselow JT, Westermark PO, Reischl S, Maier B, Korte T, Herrmann A, Herzel H, Schlosser A, Kramer A. Differential effects of PER2 phosphorylation: molecular basis for the human familial advanced sleep phase syndrome (FASPS) Genes Dev. 2006;20:2660–2672. doi: 10.1101/gad.397006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  293. Vaquero A, Scher M, Lee D, Erdjument-Bromage H, Tempst P, Reinberg D. Human SirT1 interacts with histone H1 and promotes formation of facultative heterochromatin. Mol. Cell. 2004;16:93–105. doi: 10.1016/j.molcel.2004.08.031. [DOI] [PubMed] [Google Scholar]
  294. Vielhaber E, Eide E, Rivers A, Gao ZH, Virshup DM. Nuclear entry of the circadian regulator mPER1 is controlled by mammalian casein kinase I ɛ. Mol. Cell. Biol. 2000;20:4888–4899. doi: 10.1128/mcb.20.13.4888-4899.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  295. Vitaterna MH, King DP, Chang AM, Kornhauser JM, Lowrey PL, McDonald JD, Dove WF, Pinto LH, Turek FW, Takahashi JS. Mutagenesis and mapping of a mouse gene, Clock, essential for circadian behavior. Science. 1994;264:719–725. doi: 10.1126/science.8171325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  296. Vitaterna MH, Selby CP, Todo T, Niwa H, Thompson C, Fruechte EM, Hitomi K, Thresher RJ, Ishikawa T, Miyazaki J, et al. Differential regulation of mammalian period genes and circadian rhythmicity by cryptochromes 1 and 2. Proc. Natl. Acad. Sci. USA. 1999;96:12114–12119. doi: 10.1073/pnas.96.21.12114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  297. Vujovic N, Davidson AJ, Menaker M. Sympathetic input modulates, but does not determine, phase of peripheral circadian oscillators. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2008;295:R355–R360. doi: 10.1152/ajpregu.00498.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  298. Walton KM, Fisher K, Rubitski D, Marconi M, Meng QJ, Sladek M, Adams J, Bass M, Chandrasekaran R, Butler T, et al. Selective inhibition of casein kinase 1ɛ minimally alters circadian clock period. J. Pharmacol. Exp. Ther. 2009;330:430–439. doi: 10.1124/jpet.109.151415. [DOI] [PubMed] [Google Scholar]
  299. Wang Y, Osterbur DL, Megaw PL, Tosini G, Fukuhara C, Green CB, Besharse JC. Rhythmic expression of Nocturnin mRNA in multiple tissues of the mouse. BMC Dev. Biol. 2001;1:9. doi: 10.1186/1471-213X-1-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  300. Welsh DK, Logothetis DE, Meister M, Reppert SM. Individual neurons dissociated from rat suprachiasmatic nucleus express independently phased circadian firing rhythms. Neuron. 1995;14:697–706. doi: 10.1016/0896-6273(95)90214-7. [DOI] [PubMed] [Google Scholar]
  301. Welsh DK, Yoo SH, Liu AC, Takahashi JS, Kay SA. Bioluminescence imaging of individual fibroblasts reveals persistent, independently phased circadian rhythms of clock gene expression. Curr. Biol. 2004;14:2289–2295. doi: 10.1016/j.cub.2004.11.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  302. Welsh DK, Takahashi JS, Kay SA. Suprachiasmatic nucleus: cell autonomy and network properties. Annu. Rev. Physiol. 2010;72:551–577. doi: 10.1146/annurev-physiol-021909-135919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  303. Wilkinson, Kevin A, Henley, Jeremy M. Mechanisms, regulation and consequences of protein SUMOylation. Biochem. J. 2010;428:133–145. doi: 10.1042/BJ20100158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  304. Wilkinson KD. Ubiquitin-dependent signaling: the role of ubiquitination in the response of cells to their environment. J. Nutr. 1999;129:1933–1936. doi: 10.1093/jn/129.11.1933. [DOI] [PubMed] [Google Scholar]
  305. Woo KC, Kim TD, Lee KH, Kim DY, Kim W, Lee KY, Kim KT. Mouse period 2 mRNA circadian oscillation is modulated by PTB-mediated rhythmic mRNA degradation. Nucleic Acids Res. 2009;37:26–37. doi: 10.1093/nar/gkn893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  306. Woo KC, Ha DC, Lee KH, Kim DY, Kim TD, Kim KT. Circadian amplitude of cryptochrome 1 is modulated by mRNA stability regulation via cytoplasmic hnRNP D oscillation. Mol. Cell. Biol. 2010;30:197–205. doi: 10.1128/MCB.01154-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  307. Xu Y, Padiath QS, Shapiro RE, Jones CR, Wu SC, Saigoh N, Saigoh K, Ptacek LJ, Fu YH. Functional consequences of a CKIδ mutation causing familial advanced sleep phase syndrome. Nature. 2005;434:640–644. doi: 10.1038/nature03453. [DOI] [PubMed] [Google Scholar]
  308. Xu Y, Toh KL, Jones CR, Shin JY, Fu YH, Ptacek LJ. Modeling of a human circadian mutation yields insights into clock regulation by PER2. Cell. 2007;128:59–70. doi: 10.1016/j.cell.2006.11.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  309. Yagita K, Tamanini F, van Der Horst GT, Okamura H. Molecular mechanisms of the biological clock in cultured fibroblasts. Science. 2001;292:278–281. doi: 10.1126/science.1059542. [DOI] [PubMed] [Google Scholar]
  310. Yamada YR, Forger DB. Multiscale complexity in the mammalian circadian clock. Curr. Opin. Genet. Dev. 2010;20:626–633. doi: 10.1016/j.gde.2010.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  311. Yamaguchi S, Isejima H, Matsuo T, Okura R, Yagita K, Kobayashi M, Okamura H. Synchronization of cellular clocks in the suprachiasmatic nucleus. Science. 2003;302:1408–1412. doi: 10.1126/science.1089287. [DOI] [PubMed] [Google Scholar]
  312. Yamazaki S, Numano R, Abe M, Hida A, Takahashi R, Ueda M, Block GD, Sakaki Y, Menaker M, Tei H. Resetting central and peripheral circadian oscillators in transgenic rats. Science. 2000;288:682–685. doi: 10.1126/science.288.5466.682. [DOI] [PubMed] [Google Scholar]
  313. Yan L, Silver R. Resetting the brain clock: time course and localization of mPER1 and mPER2 protein expression in suprachiasmatic nuclei during phase shifts. Eur. J. Neurosci. 2004;19:1105–1109. doi: 10.1111/j.1460-9568.2004.03189.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  314. Yang X, Downes M, Yu RT, Bookout AL, He W, Straume M, Mangelsdorf DJ, Evans RM. Nuclear receptor expression links the circadian clock to metabolism. Cell. 2006;126:801–810. doi: 10.1016/j.cell.2006.06.050. [DOI] [PubMed] [Google Scholar]
  315. Yin L, Wang J, Klein PS, Lazar MA. Nuclear receptor Rev-erbα is a critical lithium-sensitive component of the circadian clock. Science. 2006;311:1002–1005. doi: 10.1126/science.1121613. [DOI] [PubMed] [Google Scholar]
  316. Yoo SH, Yamazaki S, Lowrey PL, Shimomura K, Ko CH, Buhr ED, Siepka SM, Hong HK, Oh WJ, Yoo OJ, et al. PERIOD2::LUCIFERASE real-time reporting of circadian dynamics reveals persistent circadian oscillations in mouse peripheral tissues. Proc. Natl. Acad. Sci. USA. 2004;101:5339–5346. doi: 10.1073/pnas.0308709101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  317. Yoo SH, Ko CH, Lowrey PL, Buhr ED, Song EJ, Chang S, Yoo OJ, Yamazaki S, Lee C, Takahashi JS. A noncanonical E-box enhancer drives mouse Period2 circadian oscillations in vivo . Proc. Natl. Acad. Sci. USA. 2005;102:2608–2613. doi: 10.1073/pnas.0409763102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  318. Yoshitane H, Takao T, Satomi Y, Du NH, Okano T, Fukada Y. Roles of CLOCK phosphorylation in suppression of E-box-dependent transcription. Mol. Cell. Biol. 2009;29:3675–3686. doi: 10.1128/MCB.01864-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  319. Young MW, Kay SA. Time zones: a comparative genetics of circadian clocks. Nat. Rev. Genet. 2001;2:702–715. doi: 10.1038/35088576. [DOI] [PubMed] [Google Scholar]
  320. Yu W, Zheng H, Price JL, Hardin PE. DOUBLETIME plays a noncatalytic role to mediate CLOCK phosphorylation and repress CLOCK-dependent transcription within the Drosophila circadian clock. Mol. Cell. Biol. 2009;29:1452–1458. doi: 10.1128/MCB.01777-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  321. Zhang X, Odom DT, Koo SH, Conkright MD, Canettieri G, Best J, Chen H, Jenner R, Herbolsheimer E, Jacobsen E, et al. Genome-wide analysis of cAMP-response element binding protein occupancy, phosphorylation, and target gene activation in human tissues. Proc. Natl. Acad. Sci. USA. 2005;102:4459–4464. doi: 10.1073/pnas.0501076102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  322. Zhao WN, Malinin N, Yang FC, Staknis D, Gekakis N, Maier B, Reischl S, Kramer A, Weitz CJ. CIPC is a mammalian circadian clock protein without invertebrate homologues. Nat. Cell Biol. 2007;9:268–275. doi: 10.1038/ncb1539. [DOI] [PubMed] [Google Scholar]
  323. Zheng B, Larkin DW, Albrecht U, Sun ZS, Sage M, Eichele G, Lee CC, Bradley A. The mPer2 gene encodes a functional component of the mammalian circadian clock. Nature. 1999;400:169–173. doi: 10.1038/22118. [DOI] [PubMed] [Google Scholar]
  324. Zheng B, Albrecht U, Kaasik K, Sage M, Lu W, Vaishnav S, Li Q, Sun ZS, Eichele G, Bradley A, et al. Nonredundant roles of the mPer1 and mPer2 genes in the mammalian circadian clock. Cell. 2001;105:683–694. doi: 10.1016/s0092-8674(01)00380-4. [DOI] [PubMed] [Google Scholar]

RESOURCES