Regulation of circadian behaviour and metabolism by REV ... · Regulation of circadian behaviour...

5
LETTER doi:10.1038/nature11048 Regulation of circadian behaviour and metabolism by REV-ERB-a and REV-ERB-b Han Cho 1 , Xuan Zhao 1 , Megumi Hatori 2 , Ruth T. Yu 1 , Grant D. Barish 1 , Michael T. Lam 3 , Ling-Wa Chong 1 , Luciano DiTacchio 2 , Annette R. Atkins 1 , Christopher K. Glass 3 , Christopher Liddle 4 , Johan Auwerx 5 , Michael Downes 1 , Satchidananda Panda 2 & Ronald M. Evans 1,6 The circadian clock acts at the genomic level to coordinate internal behavioural and physiological rhythms via the CLOCK–BMAL1 transcriptional heterodimer. Although the nuclear receptors REV-ERB-a and REV-ERB-b have been proposed to form an accessory feedback loop that contributes to clock function 1,2 , their precise roles and importance remain unresolved. To establish their regulatory potential, we determined the genome-wide cis-acting targets (cistromes) of both REV-ERB isoforms in murine liver, which revealed shared recognition at over 50% of their total DNA binding sites and extensive overlap with the master circadian regulator BMAL1. Although REV-ERB-a has been shown to regu- late Bmal1 expression directly 1,2 , our cistromic analysis reveals a more profound connection between BMAL1 and the REV-ERB-a and REV-ERB-b genomic regulatory circuits than was previously suspected. Genes within the intersection of the BMAL1, REV-ERB-a and REV-ERB-b cistromes are highly enriched for both clock and metabolic functions. As predicted by the cistromic analysis, dual depletion of Rev-erb-a and Rev-erb-b function by creating double- knockout mice profoundly disrupted circadian expression of core circadian clock and lipid homeostatic gene networks. As a result, double-knockout mice show markedly altered circadian wheel- running behaviour and deregulated lipid metabolism. These data now unite REV-ERB-a and REV-ERB-b with PER, CRY and other components of the principal feedback loop that drives circadian expression and indicate a more integral mechanism for the coordi- nation of circadian rhythm and metabolism. The circadian clock is a transcriptional mechanism that coordinates both behavioural and physiological processes such as the sleep–wake cycle and food intake. The existing model for the mammalian core molecular clock involves a transcriptional negative-feedback loop in which the transactivation of E-box-containing genes by CLOCK and BMAL1 is inhibited by the expression of Per1 and Cry1, themselves under transcriptional control of E boxes 3 . As functional redundancy is common for core clock components, deletion of multiple paralogues in mice is often required to uncover relevant phenotypes such as perturbations in circadian gene expression, metabolism and wheel-running behaviour 4–7 . As REV-ERB-a, REV-ERB-b, ROR-a, ROR-b and ROR-c bind to a common response element (the RORE), their intrinsic repressive and inductive activities, respectively, are believed to establish the rhythmic expression of target genes such as Bmal1. However, the partial penetrance and mild period phenotype of Rev-erb-a 2/2 mice 1 (essentially intact circadian wheel-running behaviour) and the modest rhythmic pheno- type upon partial Rev-erb-b depletion of Rev-erb-a 2/2 cultured cells 2 , suggested that they are not required for principal core clock function. Rather, REV-ERBs are proposed to form an accessory feedback loop that stabilizes the clock and has a role in receiving input signals or transmitting output pathways 2,8 . To clarify the regulatory potential of REV-ERB-a and REV-ERB-b in circadian regulation, we generated isoform-specific antibodies (Supplementary Methods and Supplementary Fig. 1) and determined their genome-wide cis-acting targets (cistromes) in the liver at Zeitgeber time (ZT) 8, the peak of their protein expression (data not shown and ref. 9). De novo motif analysis (Fig. 1a) revealed that in vivo, in addition to the classic REV-ERB direct repeat 2 (DR2) motifs, other nuclear receptor binding sites (particularly DR1) are predominant in the DNA elements bound by both REV-ERB-a and REV-ERB-b 10 . Although not surprising, extensive overlap was observed between the REV-ERB cistromes, with commonly bound peaks accounting for 54.8% and 60.7% of the total REV-ERB-a and REV-ERB-b peaks, respectively (Fig. 1b). The limited overlap of our REV-ERB-a cistrome with one that has been published previously 11 (,50%) can most likely be attributed to differences in the antibody specificities (Supplemen- tary Figs 2 and 3). Pathway analyses of our REV-ERB-a and REV- ERB-b overlapping peaks 12 revealed an enrichment in lipid metabolism genes (Fig. 1c), consistent with the hyperlipidaemic phenotype previously observed in Rev-erb-a null mice 13 . Notably, loci encoding circadian clock genes (Clk, Bmal1, Cry1, Cry2, Per1, Per2; see Fig. 1d) were also enriched in the REV-ERB-a/b cistromic overlap, suggesting that the coordinated actions of both REV-ERB isoforms are directly linked with clock function. A comparison of the REV-ERB-a/b cistrome with published BMAL1 binding sites 14 revealed that 28% of BMAL1 peaks (at ZT6 and ZT10) were shared with the REV-ERB-a/b (FT8) cistrome and 68% of these peaks (781) were occupied by all three transcription factors. Clear binding sites for each of these tran- scription factors were found on ‘core clock’ gene loci as well as on many clock-controlled target genes (Rev-erb-a, Rev-erb-b, Ror, Dbp, Hlf, Tef, Nfil3; see Fig. 1d, e). In addition to circadian annotated loci, the BMAL1, REV-ERB-a, REV-ERB-b triple intersection is highly enriched for genes in the receptor tyrosine kinase signalling pathway as well as those known for energy homeostasis (Supplementary Table 1). Their confluence at hundreds of clock and clock output genes indicates that beyond a simple ‘binary relationship’, REV- ERB-a/b and BMAL1 cooperate to regulate clock and clock output genes coordinately. To test the above proposal, we used Cre/lox recombination to generate three genetically modified mouse lines in C57BL/6 backgrounds harbouring global, tissue-specific, or conditional knockouts of Rev- erb-a and Rev-erb-b loci. Global Rev-erb-a 2/2 and Rev-erb-b 2/2 single knockout mice were generated using a CMV-cre transgenic allele (detailed in Supplementary Methods). Homozygous deletion of Rev- erb-b did not cause overt gross abnormalities, lethality or infertility, whereas Rev-erb-a 2/2 mice, although viable, show frequent postnatal lethality (before 2weeks of age) (Supplementary Table 2), with survivors exhibiting diminished fertility in both sexes. In contrast, 1 Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, California 92037, USA. 2 Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California 92037, USA. 3 Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093-0651, USA. 4 The Storr Liver Unit, Westmead Millennium Institute and University of Sydney, Westmead Hospital, Westmead, New South Wales 2145, Australia. 5 Ecole Polytechnique Fe ´de ´ rale in Lausanne CH-1015, Lausanne, Switzerland. 6 Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, California 92037, USA. 3 MAY 2012 | VOL 485 | NATURE | 123 Macmillan Publishers Limited. All rights reserved ©2012

Transcript of Regulation of circadian behaviour and metabolism by REV ... · Regulation of circadian behaviour...

Page 1: Regulation of circadian behaviour and metabolism by REV ... · Regulation of circadian behaviour and metabolism by REV-ERB-a and REV-ERB-b Han Cho1, Xuan Zhao1, Megumi Hatori2, Ruth

LETTERdoi:10.1038/nature11048

Regulation of circadian behaviour and metabolismby REV-ERB-a and REV-ERB-bHan Cho1, Xuan Zhao1, Megumi Hatori2, Ruth T. Yu1, Grant D. Barish1, Michael T. Lam3, Ling-Wa Chong1, Luciano DiTacchio2,Annette R. Atkins1, Christopher K. Glass3, Christopher Liddle4, Johan Auwerx5, Michael Downes1, Satchidananda Panda2

& Ronald M. Evans1,6

The circadian clock acts at the genomic level to coordinate internalbehavioural and physiological rhythms via the CLOCK–BMAL1transcriptional heterodimer. Although the nuclear receptorsREV-ERB-a and REV-ERB-b have been proposed to form anaccessory feedback loop that contributes to clock function1,2, theirprecise roles and importance remain unresolved. To establish theirregulatory potential, we determined the genome-wide cis-actingtargets (cistromes) of both REV-ERB isoforms in murine liver,which revealed shared recognition at over 50% of their totalDNA binding sites and extensive overlap with the master circadianregulator BMAL1. Although REV-ERB-a has been shown to regu-late Bmal1 expression directly1,2, our cistromic analysis reveals amore profound connection between BMAL1 and the REV-ERB-aand REV-ERB-b genomic regulatory circuits than was previouslysuspected. Genes within the intersection of the BMAL1, REV-ERB-aand REV-ERB-b cistromes are highly enriched for both clock andmetabolic functions. As predicted by the cistromic analysis, dualdepletion of Rev-erb-a and Rev-erb-b function by creating double-knockout mice profoundly disrupted circadian expression of corecircadian clock and lipid homeostatic gene networks. As a result,double-knockout mice show markedly altered circadian wheel-running behaviour and deregulated lipid metabolism. These datanow unite REV-ERB-a and REV-ERB-b with PER, CRY and othercomponents of the principal feedback loop that drives circadianexpression and indicate a more integral mechanism for the coordi-nation of circadian rhythm and metabolism.

The circadian clock is a transcriptional mechanism that coordinatesboth behavioural and physiological processes such as the sleep–wakecycle and food intake. The existing model for the mammalian coremolecular clock involves a transcriptional negative-feedback loop inwhich the transactivation of E-box-containing genes by CLOCK andBMAL1 is inhibited by the expression of Per1 and Cry1, themselves undertranscriptional control of E boxes3. As functional redundancy is commonfor core clock components, deletion of multiple paralogues in mice isoften required to uncover relevant phenotypes such as perturbations incircadian gene expression, metabolism and wheel-running behaviour4–7.

As REV-ERB-a, REV-ERB-b, ROR-a, ROR-b and ROR-c bind to acommon response element (the RORE), their intrinsic repressive andinductive activities, respectively, are believed to establish the rhythmicexpression of target genes such as Bmal1. However, the partial penetranceand mild period phenotype of Rev-erb-a2/2 mice1 (essentially intactcircadian wheel-running behaviour) and the modest rhythmic pheno-type upon partial Rev-erb-b depletion of Rev-erb-a2/2 cultured cells2,suggested that they are not required for principal core clock function.Rather, REV-ERBs are proposed to form an accessory feedback loopthat stabilizes the clock and has a role in receiving input signals ortransmitting output pathways2,8.

To clarify the regulatory potential of REV-ERB-a and REV-ERB-bin circadian regulation, we generated isoform-specific antibodies(Supplementary Methods and Supplementary Fig. 1) and determinedtheir genome-wide cis-acting targets (cistromes) in the liver atZeitgeber time (ZT) 8, the peak of their protein expression (data notshown and ref. 9). De novo motif analysis (Fig. 1a) revealed that in vivo,in addition to the classic REV-ERB direct repeat 2 (DR2) motifs, othernuclear receptor binding sites (particularly DR1) are predominant inthe DNA elements bound by both REV-ERB-a and REV-ERB-b10.Although not surprising, extensive overlap was observed betweenthe REV-ERB cistromes, with commonly bound peaks accountingfor 54.8% and 60.7% of the total REV-ERB-a and REV-ERB-b peaks,respectively (Fig. 1b). The limited overlap of our REV-ERB-a cistromewith one that has been published previously11 (,50%) can most likelybe attributed to differences in the antibody specificities (Supplemen-tary Figs 2 and 3). Pathway analyses of our REV-ERB-a and REV-ERB-b overlapping peaks12 revealed an enrichment in lipid metabolismgenes (Fig. 1c), consistent with the hyperlipidaemic phenotypepreviously observed in Rev-erb-a null mice13. Notably, loci encodingcircadian clock genes (Clk, Bmal1, Cry1, Cry2, Per1, Per2; see Fig. 1d)were also enriched in the REV-ERB-a/b cistromic overlap, suggestingthat the coordinated actions of both REV-ERB isoforms are directlylinked with clock function. A comparison of the REV-ERB-a/bcistrome with published BMAL1 binding sites14 revealed that 28% ofBMAL1 peaks (at ZT6 and ZT10) were shared with the REV-ERB-a/b(FT8) cistrome and 68% of these peaks (781) were occupied by allthree transcription factors. Clear binding sites for each of these tran-scription factors were found on ‘core clock’ gene loci as well as on manyclock-controlled target genes (Rev-erb-a, Rev-erb-b, Ror, Dbp, Hlf, Tef,Nfil3; see Fig. 1d, e). In addition to circadian annotated loci, theBMAL1, REV-ERB-a, REV-ERB-b triple intersection is highlyenriched for genes in the receptor tyrosine kinase signalling pathwayas well as those known for energy homeostasis (SupplementaryTable 1). Their confluence at hundreds of clock and clock outputgenes indicates that beyond a simple ‘binary relationship’, REV-ERB-a/b and BMAL1 cooperate to regulate clock and clock outputgenes coordinately.

To test the above proposal, we used Cre/lox recombination to generatethree genetically modified mouse lines in C57BL/6 backgroundsharbouring global, tissue-specific, or conditional knockouts of Rev-erb-a and Rev-erb-b loci. Global Rev-erb-a2/2 and Rev-erb-b2/2 singleknockout mice were generated using a CMV-cre transgenic allele(detailed in Supplementary Methods). Homozygous deletion of Rev-erb-b did not cause overt gross abnormalities, lethality or infertility,whereas Rev-erb-a2/2 mice, although viable, show frequent postnatallethality (before 2 weeks of age) (Supplementary Table 2), withsurvivors exhibiting diminished fertility in both sexes. In contrast,

1Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, California 92037, USA. 2Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California 92037, USA.3Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093-0651, USA. 4The Storr Liver Unit, WestmeadMillennium Institute and University of Sydney, Westmead Hospital, Westmead, New South Wales 2145, Australia. 5Ecole Polytechnique Federale in Lausanne CH-1015, Lausanne, Switzerland. 6HowardHughes Medical Institute, Salk Institute for Biological Studies, La Jolla, California 92037, USA.

3 M A Y 2 0 1 2 | V O L 4 8 5 | N A T U R E | 1 2 3

Macmillan Publishers Limited. All rights reserved©2012

Page 2: Regulation of circadian behaviour and metabolism by REV ... · Regulation of circadian behaviour and metabolism by REV-ERB-a and REV-ERB-b Han Cho1, Xuan Zhao1, Megumi Hatori2, Ruth

Rev-erb-a2/2 mice on a mixed 129/Sv and C57BL/6 background werereported to have reduced fertility but no postnatal lethality15.

Liver-specific Rev-erb-a2/2 Rev-erb-b2/2 mice were generated toassess combinatorial REV-ERB signalling in hepatic circadian geneexpression via an albumin-Cre transgene16. These liver-specific doubleknockout mice (Rev-erb-alox/lox Rev-erb-blox/lox albumin-Cre (L-DKO))were born at the expected Mendelian ratio, effectively bypassing thefrequent lethality of the Rev-erb-a2/2 global deletion. Depletion of bothRev-erb-a and Rev-erb-b (Fig. 2a, b) disrupted the circadian, and oftentotal, expression of many hepatic core clock genes (Fig. 2c–f) and pre-sumed output genes (Fig. 2g–i).

Microarray analysis of gene expression over a 24-h time course(presented as Zeitgeber time) revealed massive differences betweenthe wild-type and L-DKO liver. Using CircWave v3.3 (ref. 17) soft-ware, we determined that of the ,900 genes showing circadianexpression in wild-type liver, the rhythmicity of more than 90% wasperturbed in the L-DKO liver (Fig. 3a). The severity of the circadiandisruption was comparable to that described for mice deficient in coreclock components18,19, emphasizing the functional impact associatedwith the extensive intersection between REV-ERB-a/b and BMAL1cistromes. Furthermore, the gene ontology of the REV-ERB-a/b-dependent circadian transcriptome (those genes that lose rhythm inL-DKO mice; Fig. 3b) also mirrors that of their cistromes (Fig. 1c andSupplementary Table 1). This is of interest as the disrupted oscillationof these genes in the liver occurred in vivo in the presence of otherwisewild-type entraining signals and fully intact extra-hepatic clockwork(Supplementary Fig. 5). Notably, residual rhythmic expression of somegenes was maintained in the L-DKO mice, indicating that they may becontrolled by systemic cues, as suggested previously20.

To link the cistromic and transcriptomic data functionally, weexamined the gene expression levels of REV-ERB/BMAL1 commoncistromic sites. A comparison of the liver expression levels across ZT8to ZT16 revealed that 45% of the co-occupied genes were significantlyperturbed in the liver-specific DKO compared with wild-type liver(Supplementary Table 3). This strong correlation between REV-ERBoccupancy and perturbed gene expression corroborates a direct rolefor REV-ERBs in maintaining rhythmic expression patterns for manyof the above genes.

The benchmark assay for circadian dysfunction is wheel-runningbehaviour, where the persistence or fragmentation of running rhythmis assessed in constant darkness after prior entrainment to a 12-h light/dark cycle. Actograms recorded for Rev-erb-a2/2 mice in a pureC57BL/6 background displayed a fully penetrant period shorteningof 30 min (Fig. 4a, b), contrasting with the lack of fully penetrantphenotype reported in mixed background studies1,15. No changes inthe activity levels, activity consolidation, or phase angle of entrainment(activity onset relative to dark onset) were observed for Rev-erb-a2/2

mice. Rev-erb-b2/2 mice showed no change in any of the parametersof circadian activity rhythms (Fig. 4a, b), raising the possibility that thecircadian clock function is independent of REV-ERB-b. Given thefunctional redundancy demonstrated for the established componentsof the core clock, we sought to address whether REV-ERB-a andREV-ERB-bmight cooperatively influence circadian rhythms in wheelrunning. Using the tamoxifen-activated creER transgene, inducibledouble knockout (iDKO) mice were generated21, allowing ubiquitousdeletion of both genes in adulthood (see Supplementary Methods).Whereas the wheel-running activity of Rev-erb-alox/lox Rev-erb-blox/lox

mice treated with tamoxifen was normal, the phenotype of mice

HNF4A(NR/DR1)CEBPA

Onecut

HNF4A

NF1:FOXA1

PPARG::RXRA

TAL1::GATA1

ZSCAN4

ASCL2NF1

De novoAnnotated

Clock

50 kb

29.6 -

0 _44 -

0 _18.3 -

0.1 _

ArntlArntl

50 kb

22.1 -

0 _15.1 -

0 _25.3 -

0.1 _

Cry1

20 kb

Cry1

47 -

0 _51.4 -

0 _29.6 -

0.1 _

Cry2D930015M05Rik

Cry2

10 kb

18.3 -

0 _34.8 -

0 _71.8 -

0.1 _

Per1Per1

10 kb

64.8 -

0 _27.2 -

0 _99.1 -

0.1 _

Per2

20 kb

50.3 -

0 _116.7 -

0 _50.2 -

0.1 _

a b

c

e

d

Term name

Genes involved in metabolism of lipids and lipoproteins

PPAR signalling pathway

Genes involved in regulation of lipid metabolism by PPAR-αMap kinase inactivation of SMRT corepressor

Genes involved in lipoprotein metabolism

Binomial

rank

Binomial

raw

P value

1

2

3

4

5

8.7051 × 10–51

3.3434 × 10–39

2.4520 × 10–35

5.7027 × 10–26

2.4802 × 10–21

781

2,959

10,7748,589

12,632

274958,684 11,04813,413

β α

BMAL1ClockBmal1Cry1Cry2Per1Per2Rev-erb-αRev-erb-βRorcNfil3DbpTefHlf

REV-ERB-α

REV-ERB-β

BMAL1

REV-ERB-α

REV-ERB-β

BMAL1

REV-ERB-α

REV-ERB-β

BMAL1

REV-ERB-α

REV-ERB-β

BMAL1

REV-ERB-α

REV-ERB-β

BMAL1

REV-ERB-α

REV-ERB-β

BMAL1

RE

V-E

RB

-αR

EV-E

RB

αβ

Clock Cry2

Per1

Per2Cry1

Bmal1

Figure 1 | Cistromic analyses of REV-ERB-a and REV-ERB-b in liver. a, Denovo HOMER motif analysis of in vivo REV-ERB-a and REV-ERB-b binding.b, Venn diagram depicting the unique and common REV-ERB-a and REV-ERB-b bound peaks. c, Commonly bound REV-ERB-a and REV-ERB-b peaksare enriched for genes involved in lipid metabolism and associated with PPARs.

d, REV-ERB-a, REV-ERB-b and BMAL1 binding at canonical circadian clockgenes. The left axis indicates sequence-tag counts. e, BMAL1 cistromesignificantly overlaps with REV-ERB-a and REV-ERB-b. Examples of clock-related genes in overlap are listed, and selected peaks are shown inSupplementary Fig. 4.

RESEARCH LETTER

1 2 4 | N A T U R E | V O L 4 8 5 | 3 M A Y 2 0 1 2

Macmillan Publishers Limited. All rights reserved©2012

Page 3: Regulation of circadian behaviour and metabolism by REV ... · Regulation of circadian behaviour and metabolism by REV-ERB-a and REV-ERB-b Han Cho1, Xuan Zhao1, Megumi Hatori2, Ruth

bearing the creER transgene was more severe than the additive effectsof the individual knockouts (Fig. 4a–c). The iDKO mice showedreduced and severely fragmented activity, and advanced phase angleof entrainment, features also found in Bmal12/2 mice22. Furthermore,

as shown in the periodogram in Fig. 4c, in mice with detectable activityrhythms, the free-running period length under constant darkness wasshortened by as much as 2.5 h (see also Supplementary Tables 4 and 5).The severe activity defects in iDKO mice is unexpected when oneconsiders the properties of the single knockouts, and supports astrongly selected, novel and cooperative role for REV-ERB-a andREV-ERB-b in rhythmic behaviour4–7.

Mice and humans with disrupted circadian rhythms have beenshown to develop metabolic disorders including hyperlipidaemiaand hyperglycaemia, suggesting a link between proper clock functionand metabolism23. To investigate if REV-ERB activity is similarlyrequired for normal metabolic regulation, we compared metabolicparameters of tamoxifen-treated control and iDKO mice. TreatediDKO mice displayed increased circulating glucose and triglyceridelevels, and a reduction in the level of free fatty acids compared tocontrol littermates (Fig. 4d). The reduced fatty acids may reflect a moreoxidative metabolism of the double knockout mice as exemplified byreduced RERs. We also note a distinct RER phase shift in constant darkconditions (Fig. 4e), indicating dysregulation of overall body metabol-ism. Although previous studies have linked REV-ERB-a with lipidhomeostasis13,24,25, the presence of both REV-ERB-a and REV-ERB-b on multiple key lipid and bile acid regulatory genes including LXRa(also called Nr1h3), Fxr (Nr1h4), Apoc3, Cyp7a1, SHP (Nr0b2), Insig2and SREBP (Srebf1) provides a molecular mechanism for rhythmiccholesterol and bile acid metabolism. The current cistromic studieselevate REV-ERB-b to equal prominence with REV-ERB-a in thetranscriptional regulation of these pathways, and identify additionaldirect gene targets (Supplementary Fig. 6). The severity of the meta-bolic phenotype observed in the iDKO mice compared to the REV-ERB-a knockout mice is consistent with the cistromic analyses of theREV-ERBs.

Defining the genetic mechanisms that comprise the circadian clockis central to understanding how genomic rhythms are transformedinto behavioural and metabolic physiology. In this study, using a com-bination of genome-wide cistromic profiling, mRNA expression ana-lysis and three new targeted gene knockout mouse models, weunequivocally demonstrate a more critical and central role for REV-ERB-a/b circadian clock regulation than previously suspected. Thegenomic analysis confirms Bmal1 as a direct REV-ERB-a/b target,while at the genome-wide level we find a predominance of both coreclock and circadian output genes targeted by REV-ERBs, indicating amore integral role for this family in the core clock than previouslyconsidered. These findings indicate that a dynamic balance betweenBMAL1 and REV-ERB-a/b cistromes is used to regulate both coreclock and clock output genes by co-localizing opposing epigeneticregulators to shared genomic targets. In vivo, using targeted and indu-cible double knockout mice, we demonstrate that REV-ERB-a andREV-ERB-b together function as integral drivers of the circadianclock, rather than simply as stabilizers of an output, thereby redefiningthe established paradigm for these receptors. The similarity of theDKO circadian phenotype to compound core clock mutants(Per12/2Per22/2, Cry12/2Cry22/2)5–7 in severity, penetrance andstrong period shortening is more reflective of a pacemaker rather thana stabilizer of rhythm. Together, this leads to a model of mutual directregulation, with BMAL1 controlling one loop and REV-ERB-a/b asecond loop of the core clock, with both loops using cistromic conver-gence to coordinate key clock and metabolic functions (Fig. 4f). Theadaptive feature of the circadian clock enables its control of sleep–wakecycles, physiological rhythms, energy homeostasis and behaviour. Incontrast, disruption of rhythm spawns a range of problems from jetlag, to more profound sleep disorders, obesity, metabolic disease,immune function and cancer26–29. As partnered regulators, the recentdevelopment of both potent REV-ERB agonists (that enhance repres-sion)25 and REV-ERB antagonists (that relieve repression)30 provides anew therapeutic approach to both reset disrupted rhythms and re-establish metabolic balance.

–0.50 –0.40 –0.30 –0.20 –0.10 0.00 0.10 0.20 0.30 0.40 0.50

No

rmaliz

ed

exp

ressio

n

log

2

Albumin Cre– Albumin Cre+

0 4 8 12 16 20 0 4 8 12 16 20

a

ZT

bCount P value

Insulin signalling pathway 19 1.24 × 10–3

Circadian rhythm 5 5.71 × 10–3

Glycine, serine and threonine metabolism 7 1.02 × 10–2

GnRH signalling pathway 13 1.18 × 10–2

PPAR signalling pathway 11 1.78 × 10–2

Biosynthesis of unsaturated fatty acids 6 1.97 × 10–2

Porphyrin and chlorophyll metabolism 6 3.00 × 10–2

Citrate cycle (TCA cycle) 6 3.41 × 10–2

Glycerophospholipid metabolism 9 4.37 × 10–2

Adipocytokine signalling pathway 9 4.37 × 10–2

ABC transporters 7 4.84 × 10–2

KEGG pathway term

Figure 3 | Broad disruption of circadian transcriptome in the absence ofRev-erb-a and Rev-erb-b. a, Heat map of genes with circadian expression inwild-type (left panel) and L-DKO (right panel) livers. A total of 1,227 uniqueaccession numbers were selected based on false discovery rate ,0.05. b, Genesexpressed in a circadian manner that lose rhythm are highly associated withcircadian and energy homeostasis functions as assessed by KEGG pathwayanalysis.

0

2

4

6

0 4 8 121620012345

0 4 8 121620

0

1

2

0 4 8 1216200

1

2

3

0 4 8 121620

01234

0 4 8 121620

0

1

2

3

0 4 8 1216200

1

2

0 4 8 1216200

1

2

3

0 4 8 121620

a b c

d e f

g h i

ZT (h)

RU

ZT (h)

RU

ZT (h)

RU

ZT (h)

RU

ZT (h)

RU

ZT (h)

RU

ZT (h)

RU

ZT (h)

RU

Sco2

012345

0 4 8 121620

Per2

RU***

***

***

*****

**

****

***

***

*** ***

* ***

*

*****

**

*

*

*

*

ZT (h)

Rev-erb-α Rev-erb-β Cry1

Bmal1Clock

PparaPor

Figure 2 | Circadian gene expression of many canonical core clock genesand output genes are disrupted in livers of Rev-erb-alox/lox Rev-erb-blox/lox

albumin-Cre (L-DKO) mice. a–i, The expression levels of Rev-erb-a (a), Rev-erb-b (b), canonical core clock genes (Cry1, Clock, Bmal1 and Per2) (c–f) andpresumed output genes (Por, Ppara and Sco2) (g–i) in livers from L-DKO(albumin-Cre positive, red labels) and wild-type (albumin-Cre negative, blacklabels) mice are shown. Livers (n 5 3) were collected at each indicated ZT undera 12-h light/dark cycle. QPCR was performed in technical triplicates. Relativeunits (RU) normalized with 36B4. Error bars indicate standard error of themean; statistical significance was determined by Student’s t-test (*P , 0.05,**P , 0.01, ***P , 0.001).

LETTER RESEARCH

3 M A Y 2 0 1 2 | V O L 4 8 5 | N A T U R E | 1 2 5

Macmillan Publishers Limited. All rights reserved©2012

Page 4: Regulation of circadian behaviour and metabolism by REV ... · Regulation of circadian behaviour and metabolism by REV-ERB-a and REV-ERB-b Han Cho1, Xuan Zhao1, Megumi Hatori2, Ruth

METHODS SUMMARYFor chromatin immunoprecipitation (ChIP), 5-month-old male C57BL/6J micewere killed by CO2 asphyxiation at ZT8. Livers were removed and pooled for ChIP,processing and sequencing. Tamoxifen-induction of Cre recombinase activity wasaccomplished by daily intraperitoneal injection of 2 mg tamoxifen (Sigma) in100ml corn oil (Sigma) for 7 days. Three-month-old tamoxifen-treated animalswere subjected to wheel-running assays for 7 days after the end of treatment. Forgene expression analysis, 5–6-month-old males were killed by cervical dislocationat indicated ZT points for rapid dissection and snap freezing of the tissues. Duringthe dark cycle, procedures were performed under red light. Detailed methods areprovided in the Supplementary Information.

Received 14 February; accepted 21 March 2012.

Published online 29 March 2012.

1. Preitner, N. et al. The orphan nuclear receptor REV-ERBa controls circadiantranscription within the positive limb of the mammalian circadian oscillator. Cell110, 251–260 (2002).

2. Liu, A. C. et al. Redundant function of REV-ERBa and b and non-essential role forBmal1 cycling in transcriptional regulation of intracellular circadian rhythms.PLoS Genet. 4, e1000023 (2008).

3. Gekakis, N. et al. Role of the CLOCK protein in the mammalian circadianmechanism. Science 280, 1564–1569 (1998).

4. DeBruyne, J. P.,Weaver,D.R.& Reppert, S.M.CLOCKandNPAS2haveoverlappingroles in the suprachiasmatic circadian clock. Nature Neurosci. 10, 543–545(2007).

5. Zheng, B. et al. Nonredundant roles of the mPer1 and mPer2 genes in themammalian circadian clock. Cell 105, 683–694 (2001).

6. van der Horst, G. T. et al. Mammalian Cry1 and Cry2 are essential for maintenanceof circadian rhythms. Nature 398, 627–630 (1999).

7. Vitaterna, M. H. et al. Differential regulation of mammalian period genes andcircadian rhythmicity by cryptochromes 1 and 2. Proc. Natl Acad. Sci. USA 96,12114–12119 (1999).

8. Levi, F. & Schibler, U. Circadian rhythms: mechanisms and therapeuticimplications. Annu. Rev. Pharmacol. Toxicol. 47, 593–628 (2007).

9. Ukai-Tadenuma, M. et al. Delay in feedback repression by cryptochrome 1 isrequired for circadian clock function. Cell 144, 268–281 (2011).

10. Yang, X. et al. Nuclear receptor expression links the circadian clock to metabolism.Cell 126, 801–810 (2006).

11. Feng, D. et al. A circadian rhythm orchestrated by histone deacetylase 3 controlshepatic lipid metabolism. Science 331, 1315–1319 (2011).

12. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-basedapproach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci.USA 102, 15545–15550 (2005).

13. Le Martelot, G. et al. REV-ERBa participates in circadian SREBP signaling and bileacid homeostasis. PLoS Biol. 7, e1000181 (2009).

14. Rey, G. et al. Genome-wide and phase-specific DNA-binding rhythms of BMAL1control circadian output functions in mouse liver. PLoS Biol. 9, e1000595(2011).

15. Chomez, P. et al. Increased cell death and delayed development in the cerebellumof mice lacking the rev-erbAa orphan receptor. Development 127, 1489–1498(2000).

16. Postic, C. et al. Dual roles for glucokinase inglucose homeostasis as determined byliver and pancreatic beta cell-specific gene knock-outs using Cre recombinase.J. Biol. Chem. 274, 305–315 (1999).

17. Oster, H., Damerow, S., Hut, R. A. & Eichele, G. Transcriptional profiling in theadrenal gland reveals circadian regulation of hormone biosynthesis genes andnucleosome assembly genes. J. Biol. Rhythms 21, 350–361 (2006).

0

10

20

30

Mean c

ounts

min

–1

20 24 28 320

1

2

3

23.50

Period length (h)

0

10

20

30

40

20 24 28 320

1

2

3

Am

plit

ud

e (

×1,0

00)

23.08

Period length (h)

0

10

20

30

40

20 24 28 320

1

2

3

4

23.42

Period length (h)

05

10152025

30

20 24 28 32

1

2

3

21.29

Period length (h)

a

b

c

d

e

f

Rev-erb-α–/– iDKOWild type

Am

plit

ud

e (

×1,0

00)

Am

plit

ud

e (

×1,0

00)

Am

plit

ud

e (

×1,0

00)

0

50iDKOWild type

0

50

100

150

0

0.5

1.0

1.5

0

0.5

1.0

1.5

2.0

P<0.002 P<0.002 P<0.05

Glu

co

se

(mg

dl–

1)

Fre

e f

att

y a

cid

(m

M)

Trig

lycerid

e

(m

M)

CreER

+

CreER

CreER

+

CreER

CreER

+

CreER

Wild type iDKO

24 36 48 72 84 9660

1.00

0.95

0.90

0.85

0.80

0.75

RE

R

Zeitgeber time

(h)

Rev-erb-β–/–

Mean c

ounts

min

–1

Mean c

ounts

min

–1

Mean c

ounts

min

–1Rev-erb-β–/–Rev-erb-α–/–

Rev-erb-α–/– Rev-erb-β–/–

CRYPER1

CLOCK BMAL1

RORE E box

Cry

Per

Clock

Bmal1

Rev-erb-α

Rev-erb-β

REV-ERB-α REV-ERB-β

REV-ERB-α REV-ERB-β

L/D L/D L/D L/D

D/D D/D D/D D/D

Figure 4 | Loss of both Rev-erb-a and Rev-erb-b results in disruptedcircadian wheel-running behaviour and metabolic shift. a–c, Voluntarylocomotor activity of wild-type, Rev-erb-a2/2, Rev-erb-b2/2 and Rev-erb-a2/2

Rev-erb-b2/2 (iDKO) mice. a, Actograms showing wheel-running activity inconstant darkness after prior entrainment in light/dark. b, Activity profilesduring light/dark cycles. c, Chi-squared periodogram of the initial 20 days inconstant darkness (n 5 5–9 for each mutant strain; n 5 5–6 littermate

controls). Representative actograms from individual mice are shown. Theperiod length of wild-type mice is shown as a vertical orange line. d, Fastingglucose (n 5 6), free fatty acid (n 5 6) and triglyceride (n 5 6) levels in iDKOand wild-type mice. e, Respiratory exchange ratio (RER, vCO2

/vO2) for wild-type

(black) and iDKO (red) mice (n 5 4). f, Model depicting the activating(CLOCK–BMAL1) and repressive (REV-ERB-a–REV-ERB-b) transcriptionalcomplexes, the coordinate actions of which generate rhythmic gene expression.

RESEARCH LETTER

1 2 6 | N A T U R E | V O L 4 8 5 | 3 M A Y 2 0 1 2

Macmillan Publishers Limited. All rights reserved©2012

Page 5: Regulation of circadian behaviour and metabolism by REV ... · Regulation of circadian behaviour and metabolism by REV-ERB-a and REV-ERB-b Han Cho1, Xuan Zhao1, Megumi Hatori2, Ruth

18. Miller, B. H. et al. Circadian and CLOCK-controlled regulation of the mousetranscriptome and cell proliferation. Proc. Natl Acad. Sci. USA 104, 3342–3347(2007).

19. Hatanaka, F. et al. Genome-wide profiling of the core clock protein BMAL1 targetsreveals a strict relationship with metabolism. Mol. Cell. Biol. 30, 5636–5648(2010).

20. Kornmann, B. et al. System-driven and oscillator-dependent circadiantranscription in mice with a conditionally active liver clock. PLoS Biol. 5, e34(2007).

21. Hayashi, S. & McMahon, A. P. Efficient recombination in diverse tissues by atamoxifen-inducible form of Cre: a tool for temporally regulated gene activation/inactivation in the mouse. Dev. Biol. 244, 305–318 (2002).

22. Bunger, M. K. et al. Mop3 is an essential component of the master circadianpacemaker in mammals. Cell 103, 1009–1017 (2000).

23. Huang, W. et al. Circadian rhythms, sleep, and metabolism. J. Clin. Invest. 121,2133–2141 (2011).

24. Raspe, E. et al. Identification of Rev-erba as a physiological repressor of apoC-IIIgene transcription. J. Lipid Res. 43, 2172–2179 (2002).

25. Kumar, N. et al. Regulation of adipogenesis by natural and synthetic REV-ERBligands. Endocrinology 151, 3015–3025 (2010).

26. Gibbs, J. E. et al. The nuclear receptor REV-ERBa mediates circadian regulation ofinnate immunity through selective regulationof inflammatorycytokines.Proc.NatlAcad. Sci. USA 109, 582–587 (2012).

27. Huang, W., Ramsey, K. M., Marcheva, B. & Bass, J. Circadian rhythms, sleep, andmetabolism. J. Clin. Invest. 121, 2133–2141 (2011).

28. Lamia, K. A. et al. AMPK regulates the circadian clock by cryptochromephosphorylation and degradation. Science 326, 437–440 (2009).

29. Lamia, K. A. et al. Cryptochromes mediate rhythmic repression of theglucocorticoid receptor. Nature 480, 552–556 (2011).

30. Kojetin, D., Wang, Y., Kamenecka, T. M. & Burris, T. P. Identification of SR8278, asynthetic antagonist of the nuclear heme receptor REV-ERB. ACS Chem. Biol. 6,131–134 (2011).

Supplementary Information is linked to the online version of the paper atwww.nature.com/nature.

Acknowledgements We thank S. Kaufman, J. Alvarez, E. Banayo, H. Juguilon, S. Jacintoand H. Le for technical assistance; and L. Ong and S. Ganley for administrativeassistance. We also thank L. Pei for discussion. R.M.E is an Investigator of the HowardHughes Medical Institute at The Salk Institute for Biological Studies and March ofDimes Chair in Molecular and Developmental Biology. H.C. is a recipient of NationalResearch Service Award (T32-HL007770). This work was supported by NationalInstitutes of Health Grants (DK062434, DK057978, DK090962, DK091618 andHL105278), National Health and Medical Research Council of Australia Project Grants(NHMRC 512354 and 632886), the Helmsley Charitable Trust, the Glenn Foundationand the Howard Hughes Medical Institute.

Author Contributions X.Z., G.D.B., R.T.Y., M.D. and C.L. performed and/or analysed theresults from ChIP-seq. R.T.Y., M.D., M.T.L. and C.K.G. performed and/or analysed theresults from the microarray experiment. M.H., L.D. and S.P. performed and/or analysedthe wheel-running assay and the real-time luciferase assay. J.A. performed genetargeting. H.C. and L.-W.C. performed all experiments. H.C. and R.M.E. designed allexperiments, analysed all results and H.C., R.T.Y., M.D., A.R.A., S.P. and R.M.E. wrote themanuscript.

Author Information Microarray and ChIP-seq data sets have been deposited in theNCBI Gene Expression Omnibus with the accession number GSE34020. Reprints andpermissions information is available at www.nature.com/reprints. The authors declareno competing financial interests. Readers are welcome to comment on the onlineversion of this article at www.nature.com/nature. Correspondence and requests formaterials should be addressed to R.M.E. ([email protected]).

LETTER RESEARCH

3 M A Y 2 0 1 2 | V O L 4 8 5 | N A T U R E | 1 2 7

Macmillan Publishers Limited. All rights reserved©2012