circadian posttranscriptional regulatory mechanisms in mammals

12
Circadian Posttranscriptional Regulatory Mechanisms in Mammals Carla B. Green Department of Neuroscience, Universityof Texas Southwestern Medical Center, Dallas, Texas 75390-9111 Correspondence: [email protected] The circadian clock drives rhythms in the levels of thousands of proteins in the mammalian cell, arising in part from rhythmic transcriptional regulation of the genes that encode them. However, recent evidence has shown that posttranscriptional processes also playa major role in generating the rhythmic protein makeup and ultimately the rhythmic physiologyof the cell. Regulation of steps throughout the life of the messenger RNA (mRNA), ranging from initial mRNA processing and export from the nucleus to extensive control of translation and degradation in the cytosol have been shown to be important for producing the final rhythms in protein levels critical for proper circadian rhythmicity. These findings will be reviewed here. I n mammals, cell-autonomous circadian clocks control rhythmic expression of thousands of messenger RNAs (mRNAs), ultimately generat- ing daily rhythms in biochemistry, physiology, and behavior (Pittendrigh 1981a,b; Akhtar et al. 2002; Panda et al. 2002; Storch et al. 2002; Ueda et al. 2002; Duffield 2003; Welsh et al. 2004; Reddy et al. 2006). This dynamic control of gene expression is a hallmark of the mammalian circadian clock, which is comprised of inter- locking transcriptional–translational feedback loops (Lowrey and Takahashi 2004; Takahashi et al. 2008). Steady-state levels of 5% – 10% of the mRNAs in any given tissue are rhythmic (Duffield 2003; Rey et al. 2011; Koike et al. 2012; Menet et al. 2012). Many of these genes are regulated transcriptionally, either by direct rhythmic transactivation by CLOCK/BMAL1, the central transcriptional activators of the core clock mechanism, or indirectly by other rhythmic transcription factors that are down- stream from the core loop. However, many steps occur between transcriptional initiation and ul- timate protein function and many, if not all, of these steps could conceivably be subject to cir- cadian regulation. Transcripts are capped and spliced while they are still being transcribed and then the 3 0 ends are cleaved and polyadenylated. Each of these steps requires many accessory proteins and/or small RNAs and frequently many alter- nate splice forms and alternate polyadenylation sites can be chosen. RNAs are coated with RNA- binding proteins, forming ribonucleoprotein complexes and are exported to the cytoplasm (and in some cases to specific regions of the cell), where they are translated and eventually degraded. During the time in the cytoplasm, the Editors: Paolo Sassone-Corsi, Michael W. Young, and Akhilesh B.Reddy Additional Perspectives on Circadian Rhythms available at www.cshperspectives.org Copyright # 2018 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a030692 Cite this article as Cold Spring Harb Perspect Biol 2018;10:a030692 1 on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/ Downloaded from

Upload: others

Post on 03-Oct-2021

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Circadian Posttranscriptional Regulatory Mechanisms in Mammals

Circadian Posttranscriptional RegulatoryMechanisms in Mammals

Carla B. Green

Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111

Correspondence: [email protected]

The circadian clock drives rhythms in the levels of thousands of proteins in the mammaliancell, arising in part from rhythmic transcriptional regulation of the genes that encode them.However, recent evidence has shown that posttranscriptional processes also play a majorrole in generating the rhythmic protein makeup and ultimately the rhythmic physiology ofthe cell. Regulation of steps throughout the life of the messenger RNA (mRNA), rangingfrom initial mRNA processing and export from the nucleus to extensive control of translationand degradation in the cytosol have been shown to be important for producing the finalrhythms in protein levels critical for proper circadian rhythmicity. These findings will bereviewed here.

In mammals, cell-autonomous circadian clockscontrol rhythmic expression of thousands of

messenger RNAs (mRNAs), ultimately generat-ing daily rhythms in biochemistry, physiology,and behavior (Pittendrigh 1981a,b; Akhtar et al.2002; Panda et al. 2002; Storch et al. 2002; Uedaet al. 2002; Duffield 2003; Welsh et al. 2004;Reddy et al. 2006). This dynamic control ofgene expression is a hallmark of the mammaliancircadian clock, which is comprised of inter-locking transcriptional–translational feedbackloops (Lowrey and Takahashi 2004; Takahashiet al. 2008). Steady-state levels of �5%–10% ofthe mRNAs in any given tissue are rhythmic(Duffield 2003; Rey et al. 2011; Koike et al.2012; Menet et al. 2012). Many of these genesare regulated transcriptionally, either by directrhythmic transactivation by CLOCK/BMAL1,the central transcriptional activators of the

core clock mechanism, or indirectly by otherrhythmic transcription factors that are down-stream from the core loop. However, many stepsoccur between transcriptional initiation and ul-timate protein function and many, if not all, ofthese steps could conceivably be subject to cir-cadian regulation.

Transcripts are capped and spliced whilethey are still being transcribed and then the 30

ends are cleaved and polyadenylated. Each ofthese steps requires many accessory proteinsand/or small RNAs and frequently many alter-nate splice forms and alternate polyadenylationsites can be chosen. RNAs are coated with RNA-binding proteins, forming ribonucleoproteincomplexes and are exported to the cytoplasm(and in some cases to specific regions of thecell), where they are translated and eventuallydegraded. During the time in the cytoplasm, the

Editors: Paolo Sassone-Corsi, Michael W. Young, and Akhilesh B. Reddy

Additional Perspectives on Circadian Rhythms available at www.cshperspectives.org

Copyright # 2018 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a030692

Cite this article as Cold Spring Harb Perspect Biol 2018;10:a030692

1

on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 2: Circadian Posttranscriptional Regulatory Mechanisms in Mammals

mRNAs may be deadenylated and stored in atranslationally silent form, followed by laterreadenylation to make them translationallycompetent. Regulation of the protein productsthat result from translating these mRNAs de-pends on extensive regulation at many or all ofthese steps.

Much of the work on the mechanisms ofcircadian gene expression has focused on tran-scriptional control and the primary readoutshave been steady-state mRNA levels, as a resultlargely of the ease of making these measure-ments. However, extensive evidence is accumu-lating that posttranscriptional control is an im-portant mechanism for generating appropriatecircadian outputs. For example, the develop-ment of methods to examine nascent transcripts(or pre-mRNAs) around the circadian cyclehave revealed that many rhythmic mRNAs arenot transcribed rhythmically (Koike et al. 2012;Menet et al. 2012) and mathematical modelingof these types of datasets support the idea thatmRNA degradation and other posttranscrip-tional mechanisms must be involved in gener-ating the rhythms in steady-state levels (Lucket al. 2014; Luck and Westermark 2016). Inaddition, a significant fraction of rhythmicproteins is encoded by nonrhythmic mRNAs,suggesting additional regulatory mechanisms(Reddy et al. 2006; Mauvoisin et al. 2014; Robleset al. 2014). It has also been found that the clockis quite insensitive to large fluctuations of tran-scription rate (Dibner et al. 2009), suggestingthat posttranscriptional mechanisms may beable to buffer the system to generate reliablerhythms. Moreover, circadian rhythms can existin red blood cells devoid of nuclei (O’Neill andReddy 2011; O’Neill et al. 2011), showing thatmechanisms beyond transcription can alsodrive rhythmic physiology.

This review will focus on posttranscrip-tional regulatory mechanisms in the circadiansystem in mammals. There are other excellentreviews of circadian posttranscriptional regula-tion in mammals and other organisms (e.g.,Kojima et al. 2011; Staiger and Koster 2011;Zhang et al. 2011; Lim and Allada 2013; Kojimaand Green 2015; Romanowski and Yanovsky2015; Preussner and Heyd 2016).

PROCESSING OF THE PRIMARYTRANSCRIPT

Following transcriptional initiation, DNA-de-pendent RNA polymerase II (Pol II) enters theelongation phase, proceeding down the gene togenerate the nascent RNA transcript. As thenewly synthesized transcript emerges from thepolymerase, processing and modifications be-gin to occur, starting with the addition of the7-methylguanosine cap at the 50 end followed bybinding by the Cap-binding complex (CBC)(reviewed in Carmody and Wente 2009). In-hibiting this process by knockdown of themethylase that generates the 7-methylguano-sine (Rnmt) or the major unit of the CBC(Ncbp1) causes significant lengthening of thecircadian period (.2 h) as measured by lucif-erase reporter under the control of the Bmal1promoter in rhythmic U2OS human osteo-sarcoma cells (Fustin et al. 2013). The mecha-nism by which this lengthens the period isnot known, but suggests that this first step ofmRNA processing must contribute to correctperiodicity.

As the transcript lengthens, any introns thatare present are spliced out shortly after theyemerge from Pol II by the huge ribonucleopro-tein complex called the spliceosome. This stepis extensively regulated and a significant frac-tion of genes can be alternatively spliced, gen-erating more than one (and in some cases,many) different isoforms of the final mRNA.For example, many of the core clock genes havealternative splice forms, as was first reportedfor Clock (King et al. 1997) and Bmal1 whenthey were initially cloned (Ikeda and Nomura1997).

More recently, it was reported that alterna-tive splicing of Per2 in human keratinocytesproduces an extremely short form of PER2called PER2S (Avitabile et al. 2014). This formcontains only the amino-terminal part of thenormal PER2 and is lacking more than half ofthe protein. PER2S is unique from PER2 in thatit is localized to the nucleolus. When nucleolarstructure is transiently and reversibly perturbed,the PER2S is released and resets the clock as seenfrom synchronization of cells following such

C.B. Green

2 Cite this article as Cold Spring Harb Perspect Biol 2018;10:a030692

on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 3: Circadian Posttranscriptional Regulatory Mechanisms in Mammals

perturbation. This finding represents a novelmechanism for synchronization in response tonucleolar function.

The first examination of alternative splicingon a genome-wide level across the circadian cy-cle in mammals was performed using exon ar-rays to estimate splice forms from mouse liver(McGlincy et al. 2012). They found alternativesplicing of many exons over the circadian dayand also found changes that were influencedby fasting and feeding, suggesting that this is amechanism used by the clock to regulate thespecific isoforms of mRNAs needed for eachtime of day and for nutrient availability.

Several additional cases have recently beenreported in which alternative transcripts gener-ated by alternative splicing can regulate variousaspects of clock function. For example, it hasbeen found that the Opn4 gene, which encodesthe nonvisual retinal photoreceptor melanop-sin, is alternatively spliced to generate a longand a short isoform, which differ only in theircarboxy-terminal tails (Pires et al. 2009; Jagan-nath et al. 2015). Both isoforms form functionalphotopigments, but mediate different behavio-ral responses to light, presumably through dif-ferent downstream signaling pathways resultingfrom the different carboxyl termini. Althoughboth isoforms contribute to phase shifting,OPN4S is solely responsible for pupillary con-striction and OPN4L regulates negative mask-ing to light.

A particularly intriguing example of rhyth-mic alternative splicing is that reported for theU2af26 gene, which was shown to control PE-RIOD1 protein stability and the circadian clockin mice (Preussner et al. 2014). The alternativesplicing changes the reading frame so the pro-tein encodes a carboxy-terminal domain withsimilarity to the circadian clock protein timelessfrom Drosophila (dTIM). This variant contain-ing the dTIM-like domain interacts with PER1and destabilizes it. Mice lacking U2af26 havedisruption in peripheral rhythms, arrhythmicPER protein levels, and also have rapid phaseadvances to shifted light. This alternative formis both rhythmic and induced acutely by light,and the authors of this paper suggest that alter-native splicing of this gene is a buffering mech-

anism that limits PER1 induction to preventinappropriate phase shifts.

OTHER ALTERNATE TRANSCRIPTS

Functionally analogous to alternative splicing(although not technically “posttranscription-al”), other alternate transcripts can be generatedthrough alternate transcription start site usageor through termination at alternate polyadeny-lation sites. Several examples of this type of reg-ulation have been reported for transcripts in theclock mechanism. In genome-wide analysis oftranscripts from the mouse hypothalamic su-prachiasmatic nucleus (SCN; the locus of thecentral clock in mammals), it was noticed thata previously undescribed exon was expressed inthe Cry1 gene (Pembroke et al. 2015). The in-clusion of this exon adds sequence to the 50 endof the transcript. Both isoforms are rhythmical-ly expressed but cycle in antiphase with eachother. The function of this alternate transcriptis not known.

The decision where to terminate transcrip-tion is also a highly regulated process and ChIP-seq analysis of mouse liver revealed that RNAPol II accumulates at termination sites of the Perand Cry genes, where it interacts with the PE-RIOD repressive complex of proteins during thefirst part of the repressive phase (Padmanabhanet al. 2012). The interaction of the PER complexinhibits SETX, a helicase that promotes tran-scription termination. As levels of the PER com-plex increase, this inhibition prevents normalRNA Pol II release and termination, causingthe polymerase to accumulate near the termi-nation site. Because transcription initiation andtermination are linked, this loss of terminationcauses subsequent loss of initiation, thereforegenerating another negative feedback mecha-nism by the PER complex, but at a posttran-scriptional level.

Alternative polyadenylation site usage isalso used by the circadian system to posttran-scriptionally regulate gene expression. TheRNA-binding proteins CIRBP and RBM3 areboth cold-inducible and their rhythmic expres-sion patterns are driven by the low amplituderhythms in body temperature. Analysis of their

Circadian Posttranscriptional Regulatory Mechanisms

Cite this article as Cold Spring Harb Perspect Biol 2018;10:a030692 3

on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 4: Circadian Posttranscriptional Regulatory Mechanisms in Mammals

binding showed that the 30 UTR binding sites areenriched near polyadenylation sites. Depletionof either protein shortened the UTR and coldtemperature (which increases the proteins lev-els) lengthened them (Liu et al. 2013). Changingthe length of the 30 UTR can have profoundimpact on mRNA levels, because this canchange the presence of sites for microRNAs(miRNAs) and RNA-binding proteins that reg-ulate mRNA export, stability, and translation.One of the RNAs bound by CIRBP is the ClockmRNA and knockdown of CIRBP causes re-duced rhythmicity with low amplitude rhythms(Morf et al. 2012). In the knockout, CLOCKprotein levels are low and arrhythmic and ClockmRNA was reduced in the cytoplasm but un-changed in the nucleus, suggesting that CIRBPregulates Clock mRNA nuclear export or pro-motes cytosol-specific decay. Interestingly, itwas recently reported that the threefold rhythmof Cirbp mRNA, driven by the daily smallfluctuation in body temperature, is itself post-transcriptionally generated and is the resultof temperature-dependent regulation of splic-ing efficiency (Gotic et al. 2016). This rhythmin splicing efficiency also affects the accumula-tion of other mRNAs, and represents anotherposttranscriptional mechanism in the circadiantoolbox.

EXPORT FROM THE NUCLEUS

Once the mature mRNA is generated, it must beexported from the nucleus so that it can betranslated in the cytoplasm. The export processis also highly regulated and examples of clockcontrol of this process are beginning to emerge.Evidence that rate of mRNA export contributesto the correct period length determinationcame unexpectedly from experiments probingthe role of transmethylation of mRNA (Fustinet al. 2013). Inhibition of RNA methylation, bythe global inhibitor 3-deazaadenosine or bysuppression of the m6A methylase Mettl3, wasfound to cause prolonged nuclear retention ofcircadian RNAs such as Per2 and Arntl (Bmal1mRNA), independent of transcription. Thisincreased nuclear retention caused decreasedcytoplasmic mRNA and protein levels and pro-

duced long circadian periods in MEF and U2OScells, in SCN slices and in locomotor activity inmice (when inhibitors were infused into thethird ventricle near the SCN). It is unclearfrom these studies whether the circadian length-ening effects of methylation are caused by theslowing of RNA processing at one or more steps(capping, splicing, etc.) or whether this is atargeted effect specifically on mRNA export.However, the long periods caused by inhibitionof the m7 capping are additive with the m6Amethylase inhibition, resulting in extremelylong periods of more than 30 h, suggestingthat these are independent mechanisms bothcontributing to period determination.

Other evidence for circadian control ofnuclear export comes from the demonstrationthat the nuclear bodies called paraspeckles arerhythmic. Paraspeckles are thought to functionto prevent some mRNAs containing specific se-quences (inverted Alu repeats) from being ex-ported from the nucleus and therefore preventtranslation (Chen et al. 2008). These large ribo-nucleoprotein complexes are made from a longnoncoding RNA called Neat1 bound by proteinsincluding NONO, SFPQ, RBM14, and PSPC1.In rat pituitary cells, the numbers of para-speckles and the levels of the RNA and proteincomponents change over the circadian day(Guillaumond et al. 2011; Torres et al. 2016).Furthermore, reporter RNAs containing the in-verted Alu sequences were retained in the nu-cleus and released into the cytoplasm with acorresponding circadian rhythm. The disrup-tion of paraspeckles caused loss of rhythmicityof some mRNAs, therefore showing the im-portance of this mechanism for regulatingrhythmic expression, at least in this cell type.Interestingly, NONO, one of the componentsof the paraspeckles, was originally identified asa PER1-interacting protein in Rat1 fibroblastsand knockdown of this protein in cyclingNIH3T3 cells or in Drosophila caused attenua-tion of rhythms (Brown et al. 2005). Whetherthis is the result of changes in paraspeckles or toother functions of NONO is not known, buttogether these studies support a role for nuclearRNA processing and export in generating prop-er rhythmicity.

C.B. Green

4 Cite this article as Cold Spring Harb Perspect Biol 2018;10:a030692

on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 5: Circadian Posttranscriptional Regulatory Mechanisms in Mammals

TRANSLATIONAL REGULATION

Once an mRNA is exported from the nucleusand arrives in the cytosol, numerous other reg-ulatory mechanisms determine its fate. SomemRNAs are delivered to specific cellular loca-tions, such as mRNAs that are transported topostsynaptic sites in neurons or to the leadingedge of migrating cells for local translation. Re-gardless of the location, whether or not themRNA is translated is determined by many fac-tors, including signaling pathways that regulatetranslational initiation, levels, and compositionof ribosomes, length of the mRNA poly(A) tailsthat promote circularization and ribosome ini-tiation, the composition of RNA-binding pro-teins and miRNAs that are bound to the mRNA,which can determine the stability of the mRNA.Recent global analysis from proteomic and tran-scriptomic datasets have revealed that there islittle correlation between mRNA levels and pro-tein levels, suggesting that extensive regulationoccurs after the mRNA is synthesized (Vogeland Marcotte 2012). It is clear from a numberof studies that the circadian clock impacts many,if not all, of these steps in the generation of theappropriate rhythmic protein complement ofthe cell.

Comparison of mRNA levels with proteinlevels by transcriptomic and proteomic analysesof mouse liver over the circadian day has re-vealed poor correlation, suggesting extensivecircadian control over translation and/or pro-tein degradation (Reddy et al. 2006; Mauvoisinet al. 2014; Robles et al. 2014). However, prob-lems with the sensitivity of the proteomic anal-ysis only allow the most abundant proteins to bedetected and, therefore, the extent of such reg-ulation was difficult to determine conclusivelyfrom these studies.

Examination of mRNAs bound to poly-somes (and therefore assumed to be activelytranslated) at different times of day found thattranscripts that encoded ribosomal proteinswere preferentially bound to polysomes at thebeginning of night (Jouffe et al. 2013). This cor-responds, in mice, to the beginning of the activephase and feeding phase and these mRNAs con-tained 50 UTR elements called 50-terminal oli-

gopyrimidine (50-TOP) motifs, which are regu-lated by the mammalian target of rapamycin(mTOR) complex 1 in response to nutrient sta-tus. Translation initiation is thought to be therate-limiting step in protein synthesis for mostproteins and this begins with the recognition ofthe 50 cap by eukaryotic initiation factor 4E(eIF4E; which binds as a complex with eIF4Aand eIF4G). eIF4E’s abundance and activityare highly regulated and one of these mecha-nisms is through mTOR-regulated eIF4E-BPs,which prevent eIF4E binding to eIF4G andtherefore prevent the binding to the cap. Indeed,this study found that many of these transla-tion initiation factors, the signaling pathwaysthat regulate them, and the components of themTOR1 complex were all under circadian con-trol in the mouse liver.

Control of translation initiation may be ageneral feature of circadian clocks, because thesepathways are also rhythmic in the mouse SCNand are regulated by light (Cao et al. 2008, 2010,2011; Cao and Obrietan 2010). eIF4E-BP1 ex-pression and phosphorylation are rhythmic inthe SCN and mice lacking Eif4ebp1 entrain tophase shifts more rapidly as measured by loco-motor activity and PER rhythmicity and arealso resistant to desynchrony induced by cons-tant light (Cao et al. 2013). It was also foundthat there was enhanced vasointestinal peptide(VIP) translation in the Eif4ebp1 knockoutmice, suggesting that VIP, a critical neuropep-tide in the SCN, is under translational control bythis mechanism in the SCN. Further analysis ofthis pathway revealed that signaling pathwaysthat phosphorylate eIF4E are also rhythmic inthe SCN and acutely induced by light (Cao etal. 2015). Knockin mice carrying a mutation(S209A) of eIF4E that cannot be phosphorylat-ed were shown to have deficits in phase shiftingand did not entrain well to non-24-h T cycles.Although the precise mechanism for these def-icits are not known, phosphorylation of this sitein eIF4e promotes PER1 and PER2 translationin MEFs and the S209A mutant mice also havereduced levels of PER proteins over the circadi-an cycle and following a light pulse.

Another intriguing link between the mTORpathway and the circadian clock comes from the

Circadian Posttranscriptional Regulatory Mechanisms

Cite this article as Cold Spring Harb Perspect Biol 2018;10:a030692 5

on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 6: Circadian Posttranscriptional Regulatory Mechanisms in Mammals

demonstration that the core clock transcrip-tional activator BMAL1 has a surprising cyto-solic role as a translation factor (Lipton et al.2015). It was shown that BMAL1 can be phos-phorylated by the mTOR-effector S6K1 and,when phosphorylated, BMAL1 can associatewith the translational machinery and broadlystimulate protein synthesis. These findings di-rectly connect the core circadian timing ma-chinery to the control of protein production.

More extensive analysis of rhythmic trans-lation comes from ribosomal profiling methodsin mouse liver and in rhythmic U2OS cellswith high temporal and nucleotide resolution(Jang et al. 2015; Janich et al. 2015). In themouse liver study (Janich et al. 2015), 150“high-confidence” mRNAs were identified thatwere not rhythmic in their steady-state levelsbut were rhythmically translated and a some-what smaller set with similar profiles were iden-tified in the human U2OS cells. Together, thesestudies suggest that the circadian clock controlstranslation through a number of differentmechanisms and this contributes to the finalrhythmic protein makeup of the cell.

Mathematical modeling of the effect of dif-ferent PER translation kinetics suggested thatregulation of this step could be important forgenerating self-sustained oscillations with char-acteristic period, amplitude, and phase lag (thetime delays between Per mRNA and PER pro-tein) (Nieto et al. 2015). However, both ribo-some profiling studies found that the core clockgenes had matching mRNA and ribosome pro-files, suggesting that the lag between mRNA andprotein peaks over the circadian cycle is not theresult of translational control. However, up-stream open reading frames (uORFs) were iden-tified in several clock genes in both cell types.Because active uORFs usually decrease transla-tion of the downstream main ORF, the presenceof these suggest that these mRNAs may besubject to translational control under some cir-cumstances, for example in response to envi-ronmental signals.

Additionally, a number of studies have im-plicated various RNA-binding proteins andmiRNAs in translational regulation of the coreclock mRNAs and various circadian output

mRNAs. The RNA-binding protein AUF (alsoknown as hnRNP D) is rhythmic and was shownto bind rhythmically to Cry1 30 UTR in anti-phase with peak Cry1 mRNA levels, then asso-ciates with the translation initiation factoreIF3B, recruiting the 40S subunit to 50 end, lead-ing to time-dependent translation of CRY1 (Leeet al. 2014). Another protein, LARK, was shownto bind rhythmically to the Per1 30 UTR andincreases PER1 protein expression, most likelythrough translational regulation (Kojima et al.2003; Kojima et al. 2007). LARK protein levels(but not mRNA levels) cycle in the SCN andknockdown or overexpression of this gene incycling cells produces short or long periods, re-spectively. Per1 undergoes both cap-dependentand IRES-mediated translation and the proteinhnRNP Q has been reported to control time-dependent IRES-mediated translation of Per1through rhythmic interaction with the Per1mRNA (Lee et al. 2012a,b) and to regulate thetranslation of AANAT, the rate-limiting enzymein melatonin production in the pineal gland(Kim et al. 2007).

Another potential role for translational reg-ulation is suggested by the phenotypes of micelacking the genes for the translational repressorproteins, Fmr1 (encodes FMRP) and Fxr2(Zhang et al. 2008). In mice, knockout of eitherFmr1 or Fxr2 have short periods and Fmr1/Fxr2double knockouts (both homo- and heterozy-gous) are arrhythmic. The mechanism behindthis is not known and although they did seechanges in several core clock gene mRNA levelsin liver; this study did not examine the levels ofthese proteins, so no conclusion about transla-tional control can be drawn.

Several RNA-binding proteins have alsobeen identified that contribute to the circadianregulation of mRNA half-life, which can indi-rectly impact translation. These include AUF1,which modulates Cry1 stability (Woo et al.2010), polypyrimidine tract-binding protein(PTB), which binds to Per2 30 UTR and regu-lates its stability (Woo et al. 2009), KSRP, whichregulates the stability of Per2 (Chou et al. 2015),and AUF1 and hnRNP K, which both interactwith Per3 30 UTR but have opposite effects, withhnRNP K stabilizing the mRNA and AUF1 de-

C.B. Green

6 Cite this article as Cold Spring Harb Perspect Biol 2018;10:a030692

on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 7: Circadian Posttranscriptional Regulatory Mechanisms in Mammals

stabilizing it (Kim et al. 2015). In addition,hnRNP Q, hnRNP R, and hnRNP L have allbeen shown to be expressed rhythmically inthe pineal gland and induce the degradationof the mRNA encoding AANAT (Kim et al.2005, 2007).

miRNAs

In addition to the many RNA-binding proteinsthat regulate translation and mRNA decay,miRNAs also play a critical role. And they tooappear to play a role in shaping the circadianproteome. A number of miRNAs have been im-plicated in the regulation of specific mRNAs,including the highly abundant, liver-specificmiRNA-122, which is synthesized rhythmicallyunder the control of the circadian transcrip-tional repressor Rev-erba, although the levelsof the mature form are not rhythmic becauseof its long half-life. Knockdown of this miRNAresults in both increases and decreases of hun-dreds of mRNAs, a large fraction of which arerhythmic. These include mRNAs encodingproteins in many important metabolic path-ways in the liver, including lipid and cholesterolmetabolism (Gatfield et al. 2009). Among thesemRNA targets is the mRNA encoding Noctur-nin, an enzyme that removes poly(A) tails frommRNAs and is itself involved in circadian post-transcriptional regulation (Kojima et al. 2010).

There are a number of other examples ofspecific miRNAs that are involved in circadianrhythms, including the brain-specific miRNAsmiR-219 and miR-132, which appear to con-tribute to both light-induced clock resettingand period length regulation (Cheng et al.2007; Alvarez-Saavedra et al. 2011), miR-155in macrophages, which targets the 30 UTR ofBMAL1 (Curtis et al. 2015), miR-185, whichoscillates in antiphase to CRY1 levels and con-trols Cry1 mRNA translation in NIH3T3 cells(Lee et al. 2013).

Despite these examples of specific miRNAsregulating core clock genes, the effects ofmiRNAs on the clock is less clear when globalmiRNA biogenesis is prevented by Dicer knock-down. In liver, using an inducible Dicer knock-down system, extensive changes in rhythmic

mRNA profiles are seen, but these changes arelargely modulatory, such as changes in phase oramplitude (Du et al. 2014). The core clock genemRNAs were largely not affected by miRNA lossexcept for Per2, which showed a twofold in-crease in amplitude. Liver explants from thesemice had slight period lengthening. In contrast,knockdown of Dicer in NIH3T3 cells increasedthe Cry1 30 UTR reporter activity (Lee et al.2013) and Dicer knockdown in mouse embryofibroblast cells produced short periods, whichwas attributed to faster Per1 and Per2 translation(Chen et al. 2013). Since such different resultshave been noted in these different cell types,it may be that miRNAs are important for tis-sue-specific tuning and shaping of circadiangene-expression patterns, and perhaps couldcontribute to known tissue-specific differencesin phases and free-running period lengths (Yooet al. 2004).

POLY(A) TAIL LENGTH

One of the first observations of circadian post-transcriptional control in mammals was the ob-servation that the arginine vasopressin (AVP)mRNA changed size over the circadian day inthe SCN (Robinson et al. 1988). This change insize was shown to be caused by changes in thelength of the poly(A) tails at the 30 ends of themRNA. Long poly(A) tails are added to mRNAsin the nucleus following transcription termina-tion and shortening and removal of the tails byenzymes called deadenylases is a critical step indegradation of the mRNA and is regulated byRNA-binding proteins and miRNAs (Eckmannet al. 2011). Although it was originally thoughtthat shortening the tails determined decay, it isnow known that some mRNAs can exist inshort-tailed forms and be readenylated in thecytoplasm by cytoplasmic poly(A) polymerases(reviewed in Weill et al. 2012). Circadian profil-ing of poly(A) tail lengths from mouse liversamples revealed that hundreds of mRNAshave rhythmic poly(A) tail lengths and thatthese lengths correlate well with protein levelsencoded by those mRNAs, even when themRNAs were not rhythmic at the steady-statelevel (Kojima et al. 2012). Proteins implicated in

Circadian Posttranscriptional Regulatory Mechanisms

Cite this article as Cold Spring Harb Perspect Biol 2018;10:a030692 7

on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 8: Circadian Posttranscriptional Regulatory Mechanisms in Mammals

cytoplasmic polyadenylation were also found tobe rhythmic in the mouse liver and one of these,CPEB2, was found to regulate rhythmic poly(A)addition for at least some mRNAs. Althoughpoly(A) tail length has not been found tostrongly correlate with translatability in general,it can regulate translation in certain cases weretranscriptional control is not useful such as inthe early embryo or in the postsynaptic regionof neurons (Subtelny et al. 2014). The strongcorrelation between poly(A) tail length and pro-tein rhythms in the mouse liver suggest that thecircadian system is another case in which thismechanism is used.

There are at least eight deadenylases inmammals (Goldstrohm and Wickens 2008)and several of them show low amplituderhythms, peaking in the early day (Kojima etal. 2012). However, one of them, the deadeny-lase called Nocturnin (Baggs and Green 2003;Garbarino-Pico et al. 2007), shows very highamplitude rhythms, peaking at night (Greenand Besharse 1996; Wang et al. 2001; Stubble-field et al. 2012; Godwin et al. 2013). Mice lack-ing Nocturnin have a strong metabolic pheno-type, with protection from obesity on a high-fatdiet and alterations in many circadian metabolicpathways (Green et al. 2007; Douris and Green2008; Douris et al. 2011; Stubblefield et al.2012). Comparison of poly(A) tail lengths, us-ing an approach similar to the one describedabove for the circadian analysis, revealedmRNAs with longer tails in the Nocturninknockout liver and these represent candidatesfor Nocturnin target mRNAs (Kojima et al.2015). Surprisingly, these mRNAs showed littleoverlap with the set of mRNAs with cyclingpoly(A) tail lengths or with the known rhythmicmRNAs, suggesting that the role of Nocturnin isnot to generate rhythmic tails. However, the as-say used to identify the poly(A) tail lengths inboth experiments (differential elution from oli-go[dT] beads) could not reliably detect mRNAsthat are present at low copy number. More workwith more sensitive assays is needed to definethe role for Nocturnin in the circadian clock.

Finally, the role of cytoplasmic granulessuch as processing bodies (P-bodies) in regulat-ing circadian protein expression has not been

carefully examined, but they are likely to be in-volved. These bodies store mRNAs that are notbeing translated (such as those that have beendeadenylated) until they are either degraded orplaced back into the translatable pool (Deckerand Parker 2012). Recent ribosome profiling ofU2OS cells revealed that the LSM1 protein, acritical component of P-bodies is rhythmicallytranslated and subsequent experiments showedthat cytoplasmic P-body formation is rhythmicin these cells (Jang et al. 2015). This findingprovides further evidence that the circadianclock governs the fate of mRNAs in the cyto-plasm in previously unappreciated ways.

CONCLUDING REMARKS

Circadian rhythms regulate critical functionsthroughout the mammalian body, includingbiochemical and metabolic pathways, higher or-der cell and tissue functions, and complex be-havior. These functions are generated by therhythmic patterns of protein function (notmRNA levels) and the emerging data discussedhere indicate that posttranscriptional regulatorymechanisms are not simply “refinements” oftranscriptional regulation, but are critical forgenerating the proper rhythmic protein outputsof the clock. Despite these exciting new insights,it is clear that there is much more to be learnedabout how the clock uses posttranscriptionalmechanisms to drive rhythmic physiology.

ACKNOWLEDGMENTS

I thank the National Institutes of Health (NIH)for funding support (R01 GM112991, R01GM111387, and R01 AG045795).

REFERENCES

Akhtar RA, Reddy AB, Maywood ES, Clayton JD, King VM,Smith AG, Gant TW, Hastings MH, Kyriacou CP. 2002.Circadian cycling of the mouse liver transcriptome, asrevealed by cDNA microarray, is driven by the suprachi-asmatic nucleus. Curr Biol 12: 540–550.

Alvarez-Saavedra M, Antoun G, Yanagiya A, Oliva-Hernan-dez R, Cornejo-Palma D, Perez-Iratxeta C, Sonenberg N,Cheng HY. 2011. miRNA-132 orchestrates chromatin re-modeling and translational control of the circadian clock.Hum Mol Genet 20: 731–751.

C.B. Green

8 Cite this article as Cold Spring Harb Perspect Biol 2018;10:a030692

on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 9: Circadian Posttranscriptional Regulatory Mechanisms in Mammals

Avitabile D, Genovese L, Ponti D, Ranieri D, Raffa S, Calo-gero A, Torrisi MR. 2014. Nucleolar localization and cir-cadian regulation of Per2S, a novel splicing variant of thePeriod 2 gene. Cell Mol Life Sci 71: 2547–2559.

Baggs JE, Green CB. 2003. Nocturnin, a deadenylase in Xen-opus laevis retina: A mechanism for posttranscriptionalcontrol of circadian-related mRNA. Curr Biol 13: 189–198.

Brown SA, Ripperger J, Kadener S, Fleury-Olela F, Vilbois F,Rosbash M, Schibler U. 2005. PERIOD1-associated pro-teins modulate the negative limb of the mammalian cir-cadian oscillator. Science 308: 693–696.

Cao R, Obrietan K. 2010. mTOR signaling and entrainmentof the mammalian circadian clock. Mol Cell Pharmacol 2:125–130.

Cao R, Lee B, Cho HY, Saklayen S, Obrietan K. 2008. Photicregulation of the mTOR signaling pathway in the supra-chiasmatic circadian clock. Mol Cell Neurosci 38: 312–324.

Cao R, Li A, Cho HY, Lee B, Obrietan K. 2010. Mammaliantarget of rapamycin signaling modulates photic entrain-ment of the suprachiasmatic circadian clock. J Neurosci30: 6302–6314.

Cao R, Anderson FE, Jung YJ, Dziema H, Obrietan K. 2011.Circadian regulation of mammalian target of rapamycinsignaling in the mouse suprachiasmatic nucleus. Neuro-science 181: 79–88.

Cao R, Robinson B, Xu H, Gkogkas C, Khoutorsky A, AlainT, Yanagiya A, Nevarko T, Liu AC, Amir S, et al. 2013.Translational control of entrainment and synchrony ofthe suprachiasmatic circadian clock by mTOR/4E-BP1signaling. Neuron 79: 712–724.

Cao R, Gkogkas CG, de Zavalia N, Blum ID, Yanagiya A,Tsukumo Y, Xu H, Lee C, Storch KF, Liu AC, et al. 2015.Light-regulated translational control of circadian behav-ior by eIF4E phosphorylation. Nat Neurosci 18: 855–862.

Carmody SR, Wente SR. 2009. mRNA nuclear export at aglance. J Cell Sci 122: 1933–1937.

Chen LL, DeCerbo JN, Carmichael GG. 2008. Alu element-mediated gene silencing. EMBO J 27: 1694–1705.

Chen R, D’Alessandro M, Lee C. 2013. miRNAs are requiredfor generating a time delay critical for the circadian os-cillator. Curr Biol 23: 1959–1968.

Cheng HY, Papp JW, Varlamova O, Dziema H, Russell B,Curfman JP, Nakazawa T, Shimizu K, Okamura H, ImpeyS, et al. 2007. microRNA modulation of circadian-clockperiod and entrainment. Neuron 54: 813–829.

Chou CF, Zhu X, Lin YY, Gamble KL, Garvey WT, Chen CY.2015. KSRP is critical in governing hepatic lipid metab-olism through controlling Per2 expression. J Lipid Res 56:227–240.

Curtis AM, Fagundes CT, Yang G, Palsson-McDermott EM,Wochal P, McGettrick AF, Foley NH, Early JO, Chen L,Zhang H, et al. 2015. Circadian control of innate immu-nity in macrophages by miR-155 targeting Bmal1. ProcNatl Acad Sci 112: 7231–7236.

Decker CJ, Parker R. 2012. P-bodies and stress granules:Possible roles in the control of translation and mRNAdegradation. Cold Spring Harb Perspect Biol 4: a012286.

Dibner C, Sage D, Unser M, Bauer C, d’Eysmond T, Naef F,Schibler U. 2009. Circadian gene expression is resilient to

large fluctuations in overall transcription rates. EMBO J28: 123–134.

Douris N, Green CB. 2008. NOC out the fat: A short reviewof the circadian deadenylase Nocturnin. Ann Med 40:622–626.

Douris N, Kojima S, Pan X, Lerch-Gaggl AF, Duong SQ,Hussain MM, Green CB. 2011. Nocturnin regulatescircadian trafficking of dietary lipid in intestinal enter-ocytes. Curr Biol 21: 1347–1355.

Du NH, Arpat AB, De Matos M, Gatfield D. 2014. Micro-RNAs shape circadian hepatic gene expression on a tran-scriptome-wide scale. eLife 3: e02510.

Duffield GE. 2003. DNA microarray analyses of circadiantiming: The genomic basis of biological time. J Neuro-endocrinol 15: 991–1002.

Eckmann CR, Rammelt C, Wahle E. 2011. Control ofpoly(A) tail length. Wiley Interdiscip Rev RNA 2: 348–361.

Fustin JM, Doi M, Yamaguchi Y, Hida H, Nishimura S,Yoshida M, Isagawa T, Morioka MS, Kakeya H, ManabeI, et al. 2013. RNA-methylation-dependent RNA process-ing controls the speed of the circadian clock. Cell 155:793–806.

Garbarino-Pico E, Niu S, Rollag MD, Strayer CA, BesharseJC, Green CB. 2007. Immediate early response of thecircadian polyA ribonuclease nocturnin to two extracel-lular stimuli. RNA 13: 745–755.

Gatfield D, Le Martelot G, Vejnar CE, Gerlach D, Schaad O,Fleury-Olela F, Ruskeepaa AL, Oresic M, Esau CC, Zdob-nov EM, et al. 2009. Integration of microRNA miR-122in hepatic circadian gene expression. Genes Dev 23:1313–1326.

Godwin AR, Kojima S, Green CB, Wilusz J. 2013. Kiss yourtail goodbye: The role of PARN, Nocturnin, and Angeldeadenylases in mRNA biology. Biochim Biophys Acta1829: 571–579.

Goldstrohm AC, Wickens M. 2008. Multifunctional dead-enylase complexes diversify mRNA control. Nat Rev MolCell Biol 9: 337–344.

Gotic I, Omidi S, Fleury-Olela F, Molina N, Naef F, SchiblerU. 2016. Temperature regulates splicing efficiency of thecold-inducible RNA-binding protein gene Cirbp. GenesDev 30: 2005–2017.

Green CB, Besharse JB. 1996. Identification of a novel ver-tebrate circadian clock-regulated gene encoding the pro-tein nocturnin. Proc Natl Acad Sci 93: 14884–14888.

Green CB, Douris N, Kojima S, Strayer CA, Fogerty J,Lourim D, Keller SR, Besharse JC. 2007. Loss of Noctur-nin, a circadian deadenylase, confers resistance to hepaticsteatosis and diet-induced obesity. Proc Natl Acad Sci104: 9888–9893.

Guillaumond F, Boyer B, Becquet D, Guillen S, Kuhn L,Garin J, Belghazi M, Bosler O, Franc JL, Francois-BellanAM. 2011. Chromatin remodeling as a mechanism forcircadian prolactin transcription: Rhythmic NONO andSFPQ recruitment to HLTF. FASEB J 25: 2740–2756.

Ikeda M, Nomura M. 1997. cDNA cloning and tissue-spe-cific expression of a novel basic helix–loop–helix/PASprotein (BALM1) and identification of alternativelyspliced variants with alternative translation initiationsite usage. Biochem Biophys Res Commun 233: 258–264.

Circadian Posttranscriptional Regulatory Mechanisms

Cite this article as Cold Spring Harb Perspect Biol 2018;10:a030692 9

on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 10: Circadian Posttranscriptional Regulatory Mechanisms in Mammals

Jagannath A, Hughes S, Abdelgany A, Pothecary CA, DiPretoro S, Pires SS, Vachtsevanos A, Pilorz V, BrownLA, Hossbach M, et al. 2015. Isoforms of melanopsinmediate different behavioral responses to light. CurrBiol 25: 2430–2434.

Jang C, Lahens NF, Hogenesch JB, Sehgal A. 2015. Ribosomeprofiling reveals an important role for translational con-trol in circadian gene expression. Genome Res 25: 1836–1847.

Janich P, Arpat AB, Castelo-Szekely V, Lopes M, Gatfield D.2015. Ribosome profiling reveals the rhythmic liver trans-latome and circadian clock regulation by upstream openreading frames. Genome Res 25: 1848–1859.

Jouffe C, Cretenet G, Symul L, Martin E, Atger F, Naef F,Gachon F. 2013. The circadian clock coordinates ribo-some biogenesis. PLoS Biol 11: e1001455.

Kim TD, Kim JS, Kim JH, Myung J, Chae HD, Woo KC, JangSK, Koh DS, Kim KT. 2005. Rhythmic serotonin N-ace-tyltransferase mRNA degradation is essential for themaintenance of its circadian oscillation. Mol Cell Biol25: 3232–3246.

Kim TD, Woo KC, Cho S, Ha DC, Jang SK, Kim KT. 2007.Rhythmic control of AANAT translation by hnRNP Q incircadian melatonin production. Genes Dev 21: 797–810.

Kim SH, Lee KH, Kim DY, Kwak E, Kim S, Kim KT. 2015.Rhythmic control of mRNA stability modulates circadianamplitude of mouse Period3 mRNA. J Neurochem 132:642–656.

King DP, Zhao Y, Sangoram AM, Wilsbacher LD, Tanaka M,Antoch MP, Steeves TD, Vitaterna MH, Kornhauser JM,Lowrey PL, et al. 1997. Positional cloning of the mousecircadian Clock gene. Cell 89: 641–653.

Koike N, Yoo SH, Huang HC, Kumar V, Lee C, Kim TK,Takahashi JS. 2012. Transcriptional architecture andchromatin landscape of the core circadian clock in mam-mals. Science 338: 349–354.

Kojima S, Green CB. 2015. Circadian genomics reveal a rolefor post-transcriptional regulation in mammals. Bio-chemistry 54: 124–133.

Kojima S, Hirose M, Tokunaga K, Sakaki Y, Tei H. 2003.Structural and functional analysis of 30 untranslated re-gion of mouse Period1 mRNA. Biochem Biophys Res Com-mun 301: 1–7.

Kojima S, Matsumoto K, Hirose M, Shimada M, Nagano M,Shigeyoshi Y, Hoshino S, Ui-Tei K, Saigo K, Green CB, etal. 2007. LARK activates posttranscriptional expressionof an essential mammalian clock protein, PERIOD1. ProcNatl Acad Sci 104: 1859–1864.

Kojima S, Gatfield D, Esau CC, Green CB. 2010. MicroRNA-122 modulates the rhythmic expression profile of thecircadian deadenylase Nocturnin in mouse liver. PLoSONE 5: e11264.

Kojima S, Shingle DL, Green CB. 2011. Post-transcriptionalcontrol of circadian rhythms. J Cell Sci 124: 311–320.

Kojima S, Sher-Chen EL, Green CB. 2012. Circadian controlof mRNA polyadenylation dynamics regulates rhythmicprotein expression. Genes Dev 26: 2724–2736.

Kojima S, Gendreau KL, Sher-Chen EL, Gao P, Green CB.2015. Changes in poly(A) tail length dynamics from theloss of the circadian deadenylase Nocturnin. Sci Rep 5:17059.

Lee KH, Kim SH, Kim DY, Kim S, Kim KT. 2012a. Internalribosomal entry site-mediated translation is importantfor rhythmic PERIOD1 expression. PLoS ONE 7: e37936.

Lee KH, Woo KC, Kim DY, Kim TD, Shin J, Park SM, JangSK, Kim KT. 2012b. Rhythmic interaction between Peri-od1 mRNA and hnRNP Q leads to circadian time-depen-dent translation. Mol Cell Biol 32: 717–728.

Lee KH, Kim SH, Lee HR, Kim W, Kim DY, Shin JC, Yoo SH,Kim KT. 2013. MicroRNA-185 oscillation controls circa-dian amplitude of mouse Cryptochrome 1 via transla-tional regulation. Mol Biol Cell 24: 2248–2255.

Lee KH, Kim SH, Kim HJ, Kim W, Lee HR, Jung Y, Choi JH,Hong KY, Jang SK, Kim KT. 2014. AUF1 contributes toCryptochrome1 mRNA degradation and rhythmic trans-lation. Nucleic Acids Res 42: 3590–3606.

Lim C, Allada R. 2013. Emerging roles for post-transcrip-tional regulation in circadian clocks. Nat Neurosci 16:1544–1550.

Lipton JO, Yuan ED, Boyle LM, Ebrahimi-Fakhari D, Kwiat-kowski E, Nathan A, Guttler T, Davis F, Asara JM, SahinM. 2015. The circadian protein BMAL1 regulates trans-lation in response to S6K1-mediated phosphorylation.Cell 161: 1138–1151.

Liu Y, Hu W, Murakawa Y, Yin J, Wang G, Landthaler M, YanJ. 2013. Cold-induced RNA-binding proteins regulatecircadian gene expression by controlling alternative poly-adenylation. Sci Rep 3: 2054.

Lowrey PL, Takahashi JS. 2004. Mammalian circadian biol-ogy: Elucidating genome-wide levels of temporal organi-zation. Annu Rev Genomics Hum Genet 5: 407–441.

Luck S, Westermark PO. 2016. Circadian mRNA expression:Insights from modeling and transcriptomics. Cell MolLife Sci 73: 497–521.

Luck S, Thurley K, Thaben PF, Westermark PO. 2014.Rhythmic degradation explains and unifies circadiantranscriptome and proteome data. Cell Rep 9: 741–751.

Mauvoisin D, Wang J, Jouffe C, Martin E, Atger F, Waridel P,Quadroni M, Gachon F, Naef F. 2014. Circadian clock-dependent and -independent rhythmic proteomes im-plement distinct diurnal functions in mouse liver. ProcNatl Acad Sci 111: 167–172.

McGlincy NJ, Valomon A, Chesham JE, Maywood ES, Has-tings MH, Ule J. 2012. Regulation of alternative splicingby the circadian clock and food related cues. Genome Biol13: R54.

Menet JS, Rodriguez J, Abruzzi KC, Rosbash M. 2012. Na-scent-Seq reveals novel features of mouse circadian tran-scriptional regulation. eLife 1: e00011.

Morf J, Rey G, Schneider K, Stratmann M, Fujita J, Naef F,Schibler U. 2012. Cold-inducible RNA-binding proteinmodulates circadian gene expression posttranscription-ally. Science 338: 379–383.

Nieto PS, Revelli JA, Garbarino-Pico E, Condat CA, GuidoME, Tamarit FA. 2015. Effects of different per translation-al kinetics on the dynamics of a core circadian clockmodel. PLoS ONE 10: e0115067.

O’Neill JS, Reddy AB. 2011. Circadian clocks in human redblood cells. Nature 469: 498–503.

O’Neill JS, van Ooijen G, Dixon LE, Troein C, Corellou F,Bouget FY, Reddy AB, Millar AJ. 2011. Circadian rhythms

C.B. Green

10 Cite this article as Cold Spring Harb Perspect Biol 2018;10:a030692

on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 11: Circadian Posttranscriptional Regulatory Mechanisms in Mammals

persist without transcription in a eukaryote. Nature 469:554–558.

Padmanabhan K, Robles MS, Westerling T, Weitz CJ. 2012.Feedback regulation of transcriptional termination by themammalian circadian clock PERIOD complex. Science337: 599–602.

Panda S, Antoch MP, Miller BH, Su AI, Schook AB, StraumeM, Schultz PG, Kay SA, Takahashi JS, Hogenesch JB.2002. Coordinated transcription of key pathways in themouse by the circadian clock. Cell 109: 307–320.

Pembroke WG, Babbs A, Davies KE, Ponting CP, Oliver PL.2015. Temporal transcriptomics suggest that twin-peak-ing genes reset the clock. eLife 4: e10518.

Pires SS, Hughes S, Turton M, Melyan Z, Peirson SN, ZhengL, Kosmaoglou M, Bellingham J, Cheetham ME, LucasRJ, et al. 2009. Differential expression of two distinctfunctional isoforms of melanopsin (Opn4) in the mam-malian retina. J Neurosci 29: 12332–12342.

Pittendrigh CS. 1981a. Circadian systems: Entrainment. InHandbook of behavioral neurobiology, Vol 4: Biologicalrhythms (ed. Aschoff J), pp. 95–124. Plenum, New York.

Pittendrigh C. 1981b. Circadian systems: General perspec-tive. In Handbook of behavioral neurobiology, Vol 4: Bio-logical rhythms (ed. Aschoff J), pp. 57–80. Plenum,New York.

Preussner M, Heyd F. 2016. Post-transcriptional control ofthe mammalian circadian clock: Implications for healthand disease. Pflugers Arch 468: 983–991.

Preussner M, Wilhelmi I, Schultz AS, Finkernagel F, MichelM, Moroy T, Heyd F. 2014. Rhythmic U2af26 alternativesplicing controls PERIOD1 stability and the circadianclock in mice. Mol Cell 54: 651–662.

Reddy AB, Karp NA, Maywood ES, Sage EA, Deery M,O’Neill JS, Wong GK, Chesham J, Odell M, Lilley KS, etal. 2006. Circadian orchestration of the hepatic prote-ome. Curr Biol 16: 1107–1115.

Rey G, Cesbron F, Rougemont J, Reinke H, Brunner M, NaefF. 2011. Genome-wide and phase-specific DNA-bindingrhythms of BMAL1 control circadian output functions inmouse liver. PLoS Biol 9: e1000595.

Robinson BG, Frim DM, Schwartz WJ, Majzoub JA. 1988.Vasopressin mRNA in the suprachiasmatic nuclei: Dailyregulation of polyadenylate tail length. Science 241: 342–344.

Robles MS, Cox J, Mann M. 2014. In-vivo quantitative pro-teomics reveals a key contribution of post-transcriptionalmechanisms to the circadian regulation of liver metabo-lism. PLoS Genet 10: e1004047.

Romanowski A, Yanovsky MJ. 2015. Circadian rhythms andpost-transcriptional regulation in higher plants. FrontPlant Sci 6: 437.

Staiger D, Koster T. 2011. Spotlight on post-transcriptionalcontrol in the circadian system. Cell Mol Life Sci 68: 71–83.

Storch K, Lipan O, Leykin I, Viswanathan N, Davis F, WongW, Weitz C. 2002. Extensive and divergent circadian geneexpression in liver and heart. Nature 417: 78–83.

Stubblefield JJ, Terrien J, Green CB. 2012. Nocturnin: At thecrossroads of clocks and metabolism. Trends EndocrinolMetab 23: 326–333.

Subtelny AO, Eichhorn SW, Chen GR, Sive H, Bartel DP.2014. Poly(A)-tail profiling reveals an embryonic switchin translational control. Nature 508: 66–71.

Takahashi JS, Hong HK, Ko CH, McDearmon EL. 2008. Thegenetics of mammalian circadian order and disorder:Implications for physiology and disease. Nat Rev Genet9: 764–775.

Torres M, Becquet D, Blanchard MP, Guillen S, Boyer B,Moreno M, Franc JL, Francois-Bellan AM. 2016. Circa-dian RNA expression elicited by 30-UTR IRAlu-para-speckle associated elements. eLife 5: e14837.

Ueda HR, Chen W, Adachi A, Wakamatsu H, Hayashi S,Takasugi T, Nagano M, Nakahama K, Suzuki Y, SuganoS, et al. 2002. A transcription factor response elementfor gene expression during circadian night. Nature 418:534–539.

Vogel C, Marcotte EM. 2012. Insights into the regulation ofprotein abundance from proteomic and transcriptomicanalyses. Nat Rev Genet 13: 227–232.

Wang Y, Osterbur DL, Megaw PL, Tosini G, Fukuhara C,Green CB, Besharse JC. 2001. Rhythmic expression ofNocturnin mRNA in multiple tissues of the mouse.BMC Dev Biol 1: 9.

Weill L, Belloc E, Bava FA, Mendez R. 2012. Translationalcontrol by changes in poly(A) tail length: RecyclingmRNAs. Nat Struct Mol Biol 19: 577–585.

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

Woo KC, Kim TD, Lee KH, Kim DY, Kim W, Lee KY, Kim KT.2009. Mouse period 2 mRNA circadian oscillation ismodulated by PTB-mediated rhythmic mRNA degrada-tion. Nucleic Acids Res 37: 26–37.

Woo KC, Ha DC, Lee KH, Kim DY, Kim TD, Kim KT. 2010.Circadian amplitude of cryptochrome 1 is modulated bymRNA stability regulation via cytoplasmic hnRNP D os-cillation. Mol Cell Biol 30: 197–205.

Yoo SH, Yamazaki S, Lowrey PL, Shimomura K, Ko CH,Buhr ED, Siepka SM, Hong HK, Oh WJ, Yoo OJ, et al.2004. PERIOD2::LUCIFERASE real-time reporting ofcircadian dynamics reveals persistent circadian oscilla-tions in mouse peripheral tissues. Proc Natl Acad Sci101: 5339–5346.

Zhang J, Fang Z, Jud C, Vansteensel MJ, Kaasik K, Lee CC,Albrecht U, Tamanini F, Meijer JH, Oostra BA, et al. 2008.Fragile X-related proteins regulate mammalian circadianbehavioral rhythms. Am J Hum Genet 83: 43–52.

Zhang L, Weng W, Guo J. 2011. Posttranscriptional mecha-nisms in controlling eukaryotic circadian rhythms. FEBSLett 585: 1400–1405.

Circadian Posttranscriptional Regulatory Mechanisms

Cite this article as Cold Spring Harb Perspect Biol 2018;10:a030692 11

on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 12: Circadian Posttranscriptional Regulatory Mechanisms in Mammals

August 4, 20172018; doi: 10.1101/cshperspect.a030692 originally published onlineCold Spring Harb Perspect Biol 

 Carla B. Green Circadian Posttranscriptional Regulatory Mechanisms in Mammals

Subject Collection Circadian Rhythms

Mechanisms in MammalsCircadian Posttranscriptional Regulatory

Carla B. GreenCircadian Clocks Generates Rhythmic BehaviorCoordination between Differentially Regulated

Deniz Top and Michael W. Young

Timekeeping in Eukaryotic Circadian ClocksDesign Principles of Phosphorylation-Dependent

Koji L. Ode and Hiroki R. Ueda

Introduction to Chronobiology

DuffySandra J. Kuhlman, L. Michon Craig and Jeanne F.

ClockInterplay between Microbes and the Circadian

Sassone-CorsiPaola Tognini, Mari Murakami and Paolo

Cellular Timekeeping: It's Redox o'Clock

ReddyNikolay B. Milev, Sue-Goo Rhee and Akhilesh B.

Expression, and Circadian RhythmsA 50-Year Personal Journey: Location, Gene

Michael RosbashFlies and MammalsMolecular Mechanisms of Sleep Homeostasis in

Ravi Allada, Chiara Cirelli and Amita Sehgal

Cell-Autonomous and Circuit-Level MechanismsCircadian Clockwork: Interplay between Regulating the Suprachiasmatic Nucleus (SCN)

Smyllie, et al.Erik D. Herzog, Tracey Hermanstyne, Nicola J.

Circadian ClocksMembrane Currents, Gene Expression, and

Christopher S. ColwellCharles N. Allen, Michael N. Nitabach and

Regulation in a 24-Hour Periodic WorldSystems Chronobiology: Global Analysis of Gene

Jérôme Mermet, Jake Yeung and Felix NaefManagerTimekeeper to a Complex Developmental

The Plant Circadian Clock: From a Simple

Sabrina E. Sanchez and Steve A. Kay

http://cshperspectives.cshlp.org/cgi/collection/ For additional articles in this collection, see

Copyright © 2018 Cold Spring Harbor Laboratory Press; all rights reserved

on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from