6
experimental error (~7 s), but these were not sta- tistically different from that of eIF4A alone. How- ever, ternary complexes eIF4ABG 682-1105 and eIF4AHG 682-1105 had pause lifetimes three times shorter than that of eIF4A, with durations of 2.9 T 0.1 s and 3.6 T 0.2 s (mean T SEM), respectively. These findings suggest that, in addition to enhanc- ing the directionality of eIF4A, the accessory factors eIF4B, eIF4H, and eIF4G 682-1105 also in- crease the mRNA unwinding efficiency by de- creasing pausing. Pause locations were distributed along the reporter hairpin (Fig. 3C and fig. S3). The two reporter hairpins used in this study were based on randomized nucleotide sequences, and we could identify no obvious relationship between the locations and durations of pauses and the local base composition. However, many addition- al sequences would need to be scored to identify any possible sequence dependence of helicase activity. Finally, analysis of unwinding activity indi- cates that eIF4A, alone or in complexes, has no memory associated with a given series of steps and that the translocation mechanism is governed by a single rate-limiting step (sup- plementary text). Although DEAD-box helicases have some- times been regarded as nonprocessive enzymes (29), we find here that eIF4A displays a factor- mediated processivity requiring at least two cofactors (eIF4B/H and eIF4G 682-1105 ). This ac- quired processivity is characterized by a prefer- ential directionality (presumably 5-to-3), with burst openings of 11 T 2 bp (mean T SEM) and dramatically reduced pausing. In eukaryotic translation initiation, the eIF4ABG 682-1105 com- plex comprises a minimal processive unit that is thought to facilitate sequential, step-by-step ribosomal scanning. In light of our findings, we anticipate that the processivity and dynamics of other DEAD-box family members may be influenced by accessory cofactors, so that en- zymatic activities scored in their absence may not fully reflect the in vivo function of the proteins. REFERENCES AND NOTES 1. A. Parsyan et al., Nat. Rev. Mol. Cell Biol. 12, 235245 (2011). 2. P. Linder, E. Jankowsky, Nat. Rev. Mol. Cell Biol. 12, 505516 (2011). 3. I. Jarmoskaite, R. Russell, Annu. Rev. Biochem. 83, 697725 (2014). 4. S. Rocak, P. Linder, Nat. Rev. Mol. Cell Biol. 5, 232241 (2004). 5. I. Jarmoskaite, R. Russell, Wiley Interdiscip. Rev.: RNA 2, 135152 (2011). 6. S. A. Woodson, RNA Biol. 7, 677686 (2010). 7. M. E. Fairman-Williams, U.-P. Guenther, E. Jankowsky, Curr. Opin. Struct. Biol. 20, 313324 (2010). 8. J. M. Caruthers, E. R. Johnson, D. B. McKay, Proc. Natl. Acad. Sci. U.S.A. 97, 1308013085 (2000). 9. C. E. Aitken, J. R. Lorsch, Nat. Struct. Mol. Biol. 19, 568576 (2012). 10. A. Marintchev et al., Cell 136, 447460 (2009). 11. K. H. Nielsen et al., Nucleic Acids Res. 39, 26782689 (2011). 12. Y. Sun et al., Nucleic Acids Res. 40, 61996207 (2012). 13. R. D. Abramson, T. E. Dever, W. C. Merrick, J. Biol. Chem. 263, 60166019 (1988). 14. Y. Sun et al., Structure 22, 941948 (2014). 15. F. Liu, A. Putnam, E. Jankowsky, Proc. Natl. Acad. Sci. U.S.A. 105, 2020920214 (2008). 16. M. Kozak, J. Cell Biol. 108, 229241 (1989). 17. A. Z. Andreou, D. Klostermeier, RNA Biol. 10, 1932 (2013). 18. E. De Gregorio, T. Preiss, M. W. Hentze, EMBO J. 18, 48654874 (1999). 19. K. Feoktistova, E. Tuvshintogs, A. Do, C. S. Fraser, Proc. Natl. Acad. Sci. U.S.A. 110, 1333913344 (2013). 20. N. L. Korneeva, E. A. First, C. A. Benoit, R. E. Rhoads, J. Biol. Chem. 280, 18721881 (2005). 21. J. R. Lorsch, D. Herschlag, Biochemistry 37, 21942206 (1998). 22. M. L. Peck, D. Herschlag, RNA 5, 12101221 (1999). 23. F. Rozen et al., Mol. Cell. Biol. 10, 11341144 (1990). 24. G. W. Rogers Jr., N. J. Richter, W. F. Lima, W. C. Merrick, J. Biol. Chem. 276, 3091430922 (2001). 25. G. W. Rogers Jr., N. J. Richter, W. C. Merrick, J. Biol. Chem. 274, 1223612244 (1999). 26. A. Z. Andreou, D. Klostermeier, J. Mol. Biol. 426, 5161 (2014). 27. M. J. Schnitzer, S. M. Block, Nature 388, 386390 (1997). 28. J. W. J. Kerssemakers et al., Nature 442, 709712 (2006). 29. A. M. Pyle, Annu. Rev. Biophys. 37, 317336 (2008). 30. S. Myong, M. M. Bruno, A. M. Pyle, T. Ha, Science 317, 513516 (2007). ACKNOWLEDGMENTS We thank R. Landick for providing RNAP and J. Hershey, E. Abrash, and members of the Fraser and Block labs for helpful comments. This work was supported by National Institute of General Medical Sciences grants R37GM057035 (S.M.B.) and R01GM092927 (C.S.F.), an Alejandro and Lisa Zaffaroni Graduate Fellowship (C.G.G.), a Stanford Graduate Fellowship (K.L.F.), and training grant T32 GM-007377 (K.F.). SUPPLEMENTARY MATERIALS www.sciencemag.org/content/348/6242/1486/suppl/DC1 Materials and Methods Supplementary Text Figs. S1 to S15 References (3151) 2 January 2015; accepted 20 May 2015 10.1126/science.aaa5089 GENE REGULATION Discrete functions of nuclear receptor Rev-erbα couple metabolism to the clock Yuxiang Zhang, 1 * Bin Fang, 1 * Matthew J. Emmett, 1 Manashree Damle, 1 Zheng Sun, 1,2 Dan Feng, 1 Sean M. Armour, 1 Jarrett R. Remsberg, 1 Jennifer Jager, 1 Raymond E. Soccio, 1 David J. Steger, 1 Mitchell A. Lazar 1 Circadian and metabolic physiology are intricately intertwined, as illustrated by Rev-erba, a transcription factor (TF) that functions both as a core repressive component of the cell-autonomous clock and as a regulator of metabolic genes. Here, we show that Rev-erba modulates the clock and metabolism by different genomic mechanisms. Clock control requires Rev-erba to bind directly to the genome at its cognate sites, where it competes with activating ROR TFs. By contrast, Rev-erba regulates metabolic genes primarily by recruiting the HDAC3 co-repressor to sites to which it is tethered by cell typespecific transcription factors. Thus, direct competition between Rev-erba and ROR TFs provides a universal mechanism for self-sustained control of the molecular clock across all tissues, whereas Rev-erba uses lineage-determining factors to convey a tissue-specific epigenomic rhythm that regulates metabolism tailored to the specific need of that tissue. C ircadian rhythmicity is a common fea- ture of nearly all physiological processes (14). Each cell of the body contains a mo- lecular clock composed of transcription factors that act on one another in inter- locking feedback loops that generate near24- hour oscillations (3, 5). A core component of the molecular clock, the nuclear receptor Rev-erba, is expressed with a circadian rhythm (6) and represses BMAL1, a positive regulator of clock output genes (7). Rev-erba represses many genes, often to regulate metabolism in a circadian and tissue-dependent manner (811). Thus, Rev-erba is central to complex interactions between the core clock and metabolism. Because Rev-erba is a core clock component but also has tissue-specific functions, we were in- terested in comparing its cistromes in different mouse tissues, including liver, brain, and epidid- ymal adipose tissue. The majority of Rev-erba binding sites were tissue-specific (Fig. 1A), and gene ontology analyses were consistent with specialized functions of Rev-erba (fig. S1). How- ever, a common Rev-erba cistrome included binding at clock genes in all tissues, consistent with its universal function in the core clock (fig. S1) (6, 7, 12). 1488 26 JUNE 2015 VOL 348 ISSUE 6242 sciencemag.org SCIENCE 1 Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Department of Genetics, and the Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA. 2 Department of Molecular and Cellular Biology, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA. *These authors contributed equally to this work. Corresponding author. E-mail: [email protected] RESEARCH | REPORTS on February 8, 2020 http://science.sciencemag.org/ Downloaded from

Discrete functions of nuclear receptor Rev-erbα couple ... · RevDR2 and retinoid-related orphan recep-tor (ROR)–response element (RORE) were the most enriched motifs at Rev-erba

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Discrete functions of nuclear receptor Rev-erbα couple ... · RevDR2 and retinoid-related orphan recep-tor (ROR)–response element (RORE) were the most enriched motifs at Rev-erba

experimental error (~7 s), but these were not sta-tistically different from that of eIF4A alone. How-ever, ternary complexes eIF4A•B•G682-1105 andeIF4A•H•G682-1105 had pause lifetimes three timesshorter than that of eIF4A, with durations of 2.9 T0.1 s and 3.6 T 0.2 s (mean T SEM), respectively.These findings suggest that, in addition to enhanc-ing the directionality of eIF4A, the accessoryfactors eIF4B, eIF4H, and eIF4G682-1105 also in-crease the mRNA unwinding efficiency by de-creasing pausing. Pause locationswere distributedalong the reporter hairpin (Fig. 3C and fig. S3).The two reporter hairpins used in this study werebased on randomized nucleotide sequences, andwe could identify no obvious relationship betweenthe locations and durations of pauses and thelocal base composition.However,many addition-al sequences would need to be scored to identifyany possible sequence dependence of helicaseactivity.Finally, analysis of unwinding activity indi-

cates that eIF4A, alone or in complexes, hasno memory associated with a given series ofsteps and that the translocation mechanism isgoverned by a single rate-limiting step (sup-plementary text).Although DEAD-box helicases have some-

times been regarded as nonprocessive enzymes(29), we find here that eIF4A displays a factor-mediated processivity requiring at least twocofactors (eIF4B/H and eIF4G682-1105). This ac-quired processivity is characterized by a prefer-ential directionality (presumably 5′-to-3′), withburst openings of 11 T 2 bp (mean T SEM) anddramatically reduced pausing. In eukaryotictranslation initiation, the eIF4A•B•G682-1105 com-plex comprises a minimal processive unit thatis thought to facilitate sequential, step-by-stepribosomal scanning. In light of our findings, weanticipate that the processivity and dynamicsof other DEAD-box family members may beinfluenced by accessory cofactors, so that en-zymatic activities scored in their absence maynot fully reflect the in vivo function of theproteins.

REFERENCES AND NOTES

1. A. Parsyan et al., Nat. Rev. Mol. Cell Biol. 12, 235–245(2011).

2. P. Linder, E. Jankowsky, Nat. Rev. Mol. Cell Biol. 12, 505–516(2011).

3. I. Jarmoskaite, R. Russell, Annu. Rev. Biochem. 83, 697–725(2014).

4. S. Rocak, P. Linder, Nat. Rev. Mol. Cell Biol. 5, 232–241(2004).

5. I. Jarmoskaite, R. Russell, Wiley Interdiscip. Rev.: RNA 2,135–152 (2011).

6. S. A. Woodson, RNA Biol. 7, 677–686 (2010).7. M. E. Fairman-Williams, U.-P. Guenther, E. Jankowsky, Curr.

Opin. Struct. Biol. 20, 313–324 (2010).8. J. M. Caruthers, E. R. Johnson, D. B. McKay, Proc. Natl. Acad.

Sci. U.S.A. 97, 13080–13085 (2000).9. C. E. Aitken, J. R. Lorsch, Nat. Struct. Mol. Biol. 19, 568–576

(2012).10. A. Marintchev et al., Cell 136, 447–460 (2009).11. K. H. Nielsen et al., Nucleic Acids Res. 39, 2678–2689

(2011).12. Y. Sun et al., Nucleic Acids Res. 40, 6199–6207 (2012).13. R. D. Abramson, T. E. Dever, W. C. Merrick, J. Biol. Chem. 263,

6016–6019 (1988).14. Y. Sun et al., Structure 22, 941–948 (2014).

15. F. Liu, A. Putnam, E. Jankowsky, Proc. Natl. Acad. Sci. U.S.A.105, 20209–20214 (2008).

16. M. Kozak, J. Cell Biol. 108, 229–241 (1989).17. A. Z. Andreou, D. Klostermeier, RNA Biol. 10, 19–32 (2013).18. E. De Gregorio, T. Preiss, M. W. Hentze, EMBO J. 18,

4865–4874 (1999).19. K. Feoktistova, E. Tuvshintogs, A. Do, C. S. Fraser, Proc. Natl.

Acad. Sci. U.S.A. 110, 13339–13344 (2013).20. N. L. Korneeva, E. A. First, C. A. Benoit, R. E. Rhoads, J. Biol.

Chem. 280, 1872–1881 (2005).21. J. R. Lorsch, D. Herschlag, Biochemistry 37, 2194–2206

(1998).22. M. L. Peck, D. Herschlag, RNA 5, 1210–1221 (1999).23. F. Rozen et al., Mol. Cell. Biol. 10, 1134–1144 (1990).24. G. W. Rogers Jr., N. J. Richter, W. F. Lima, W. C. Merrick, J. Biol.

Chem. 276, 30914–30922 (2001).25. G. W. Rogers Jr., N. J. Richter, W. C. Merrick, J. Biol. Chem.

274, 12236–12244 (1999).26. A. Z. Andreou, D. Klostermeier, J. Mol. Biol. 426, 51–61

(2014).27. M. J. Schnitzer, S. M. Block, Nature 388, 386–390 (1997).28. J. W. J. Kerssemakers et al., Nature 442, 709–712 (2006).

29. A. M. Pyle, Annu. Rev. Biophys. 37, 317–336 (2008).30. S. Myong, M. M. Bruno, A. M. Pyle, T. Ha, Science 317, 513–516

(2007).

ACKNOWLEDGMENTS

We thank R. Landick for providing RNAP and J. Hershey,E. Abrash, and members of the Fraser and Block labs for helpfulcomments. This work was supported by National Institute ofGeneral Medical Sciences grants R37GM057035 (S.M.B.) andR01GM092927 (C.S.F.), an Alejandro and Lisa Zaffaroni GraduateFellowship (C.G.G.), a Stanford Graduate Fellowship (K.L.F.), andtraining grant T32 GM-007377 (K.F.).

SUPPLEMENTARY MATERIALS

www.sciencemag.org/content/348/6242/1486/suppl/DC1Materials and MethodsSupplementary TextFigs. S1 to S15References (31–51)

2 January 2015; accepted 20 May 201510.1126/science.aaa5089

GENE REGULATION

Discrete functions of nuclearreceptor Rev-erbα couplemetabolism to the clockYuxiang Zhang,1* Bin Fang,1* Matthew J. Emmett,1 Manashree Damle,1

Zheng Sun,1,2 Dan Feng,1 Sean M. Armour,1 Jarrett R. Remsberg,1 Jennifer Jager,1

Raymond E. Soccio,1 David J. Steger,1 Mitchell A. Lazar1†

Circadian and metabolic physiology are intricately intertwined, as illustrated byRev-erba, a transcription factor (TF) that functions both as a core repressivecomponent of the cell-autonomous clock and as a regulator of metabolic genes.Here, we show that Rev-erba modulates the clock and metabolism by differentgenomic mechanisms. Clock control requires Rev-erba to bind directly to the genomeat its cognate sites, where it competes with activating ROR TFs. By contrast, Rev-erbaregulates metabolic genes primarily by recruiting the HDAC3 co-repressor to sites towhich it is tethered by cell type–specific transcription factors. Thus, direct competitionbetween Rev-erba and ROR TFs provides a universal mechanism for self-sustained controlof the molecular clock across all tissues, whereas Rev-erba uses lineage-determiningfactors to convey a tissue-specific epigenomic rhythm that regulates metabolism tailoredto the specific need of that tissue.

Circadian rhythmicity is a common fea-ture of nearly all physiological processes(1–4). Each cell of the body contains a mo-lecular clock composed of transcriptionfactors that act on one another in inter-

locking feedback loops that generate near–24-hour oscillations (3, 5). A core component of themolecular clock, the nuclear receptor Rev-erba,is expressed with a circadian rhythm (6) and

represses BMAL1, a positive regulator of clockoutput genes (7). Rev-erba represses many genes,often to regulate metabolism in a circadian andtissue-dependent manner (8–11). Thus, Rev-erbais central to complex interactions between thecore clock and metabolism.Because Rev-erba is a core clock component

but also has tissue-specific functions, we were in-terested in comparing its cistromes in differentmouse tissues, including liver, brain, and epidid-ymal adipose tissue. The majority of Rev-erbabinding sites were tissue-specific (Fig. 1A), andgene ontology analyses were consistent withspecialized functions of Rev-erba (fig. S1). How-ever, a common Rev-erba cistrome includedbinding at clock genes in all tissues, consistentwith its universal function in the core clock (fig.S1) (6, 7, 12).

1488 26 JUNE 2015 • VOL 348 ISSUE 6242 sciencemag.org SCIENCE

1Division of Endocrinology, Diabetes, and Metabolism,Department of Medicine, Department of Genetics, and theInstitute for Diabetes, Obesity, and Metabolism, PerelmanSchool of Medicine, University of Pennsylvania, Philadelphia,PA 19104, USA. 2Department of Molecular and CellularBiology, Division of Diabetes, Endocrinology and Metabolism,Department of Medicine, Baylor College of Medicine,Houston, TX 77030, USA.*These authors contributed equally to this work. †Correspondingauthor. E-mail: [email protected]

RESEARCH | REPORTSon F

ebruary 8, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 2: Discrete functions of nuclear receptor Rev-erbα couple ... · RevDR2 and retinoid-related orphan recep-tor (ROR)–response element (RORE) were the most enriched motifs at Rev-erba

RevDR2 and retinoid-related orphan recep-tor (ROR)–response element (RORE) were themost enriched motifs at Rev-erba binding sitesshared among tissues (Fig. 1A). This is consistentwith earlier reports that the function of Rev-erba as a repressive component of the molecu-lar clock involves binding to two RORE motifsthat function in the transcriptional regulationof the Bmal1 gene (7, 13). Rev-erba recruits thenuclear receptor co-repressor–histone deacety-lase 3 (NCoR-HDAC3) complex to actively re-press Bmal1 transcription (13), and liver-specificdeletion of HDAC3 induced Bmal1 expression(fig. S2B) at Zeitgeber time (ZT) 10, consistentwith previous reports (9, 13). However, the loss ofHDAC3 did not dampen circadian rhythmicityof Bmal1 or other clock components as much asthe loss of Rev-erba itself, suggesting an addi-tional mechanism (fig. S2, A and B).Another, non–mutually exclusive mechanism

posits competition with the activating nuclearreceptor ROR for the DNA binding site, whichcontains RevDR2/RORE motifs bound by bothreceptors (14–17). The a and g isoforms of ROR aremost abundant in liver (18) and are expressed ina circadian manner with a peak at ZT18, antiphaseto Rev-erba (19), although the circadian varia-tion of RORa is modest and of unclear biologicalimportance (fig. S3, A and B) (19). Liver-specificdeletion of RORa and g markedly dampenedthe circadian oscillation of core clock genes inliver (fig. S3, C to E), consistent with previousreports (19). To determine target genes com-mon to RORs and Rev-erba, we compared geneexpression in livers depleted of RORa and g, attheir peak time of expression, with gene ex-pression from Rev-erba knockout (KO) livers (9)(Fig. 1B). Intriguingly, genes regulated both byRev-erba and the RORs included clock genessuch as Bmal1, Npas2, Cry1, and E4bp4 (Fig. 1Band fig. S3F) and were expressed with large cir-cadian amplitudes, consistent with the modelthat Rev-erba and RORs are both critical reg-ulators of the clock (Fig. 1C). By contrast, Rev-erba–specific genes had modest circadian rhythmsand were enriched for liver metabolic processes(Fig. 1C and fig. S3F).Although RORa expression was similar at

ZT10 and ZT22, there was a marked differencebetween RORa binding to ROREs at the clockgenes Bmal1 and Npas2 at these times (fig. S4A).Deletion of Rev-erba enhanced RORa recruit-ment to these sites at ZT10, and this was poten-tiated by loss of Rev-erbb (Fig. 1D), consistentwith lower binding of RORa at ZT10 being dueto competition with Rev-erbs. Conversely, hepaticoverexpression of Rev-erba reduced RORa re-cruitment to Bmal1 and Npas2 sites at ZT22 (Fig.1E). Genome-wide, ~44% of RORa binding sitesoverlapped with Rev-erba, and these were morelikely to be circadian than were RORa-specificsites (Fig. 1F). In addition, sites of increased RORabinding at ZT22 were enriched for the RevDR2/RORE motifs bound by both Rev-erba and RORa(fig. S4, B and C). Moreover, oscillating RORabinding sites were enriched near common targetgenes of RORs and Rev-erba (Fig. 1G), further

suggesting that RORa and Rev-erba compete forbinding at highly circadian genes, including corecomponents of the molecular clock. In contrast,consistent with its expression, RORg had a cir-cadian binding pattern at overlapped and non-overlapped sites (fig. S4D).To understand why Rev-erba and ROR tended

to compete near clock genes but not Rev-erba–

specific genes, we performed chromatin immuno-precipitation (ChIP)–exonuclease followed byhigh-throughput sequencing (ChIP-exo) (20) inmouse liver at ZT10 to better resolve Rev-erbabinding (fig. S5A). At clock genes regulated byRev-erba and RORs, exemplified by Bmal1 andCry1, the RevDR2/RORE motif was detected atChIP-exo peaks (Fig. 2A, left). However, Rev-erba

SCIENCE sciencemag.org 26 JUNE 2015 • VOL 348 ISSUE 6242 1489

Fig. 1. Rev-erba represses clock genes by competing with RORa at its cognate sites. (A) Overlapof Rev-erba cistromes in liver (9), brain, and epididymal adipose tissue (eWAT). Most significantlyenriched known motifs (abundance >10%) in common and tissue-specific cistromes are shown. (B)Heat map showing expression fold changes of genes deactivated by RORa/γ double KO (DKO/WT < –1.3,P < 0.01) and derepressed by Rev-erba KO (aKO/WT > 1.3, P < 0.01). (C) Mean relative global run-onsequencing (GRO-seq) transcription (left) throughout 24-hour light–dark cycle, as well as oscillationamplitudes (right) of RORs/Rev-erba common targets (red) and Rev-erba–specific targets (blue).Time points were duplicated for clearer visualization. (D) RORa binding at clock and control genespromoters at ZT10, in WT, Rev-erba KO (aKO), Rev-erbb knockdown (bKD), and aKO/bKD mice liver,analyzed by ChIP-PCR. Data are expressed as mean T SEM (* Student’s t test, P < 0.05, n = 4 mice).(E) RORa binding at clock and control gene promoters at ZT10 or ZT22 in Rev-erba overexpression(OE) mouse liver, analyzed by ChIP-PCR. Data are expressed as mean T SEM (* Student’s t test, P <0.05, n = 6 or 7 mice). (F) Circadian binding of RORa at sites overlapped or not overlapped with Rev-erba cistrome (N.S., not significant; Student’s t test, P > 0.05). (G) Percentage of common or Rev-erba–specific target genes containing high-confidence oscillating RORa binding sites [ZT22 > 2reads per million (rpm), ZT22/ZT10 > 1.5] within 50 kb of transcription start sites (P value fromhypergeometric test).

RESEARCH | REPORTSon F

ebruary 8, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 3: Discrete functions of nuclear receptor Rev-erbα couple ... · RevDR2 and retinoid-related orphan recep-tor (ROR)–response element (RORE) were the most enriched motifs at Rev-erba

ChIP-exo peaks were most commonly enrichedfor the motif bound by liver-lineage determiningTF hepatocyte nuclear factor 6 (HNF6) (Fig. 2B).As exemplified by Cyp2b13 and Slc45a3, theseRev-erba binding sites colocalized with HNF6 inmouse liver (Fig. 2A, right). Overall, the HNF6motif was found at 1108 Rev-erba ChIP-exo sites(Fig. 2C), the vast majority of which were alsodetected by HNF6 ChIP-exo in liver (21) yet didnot have an RORE motif nearby (fig. S5B). Thegenes located nearest to these Rev-erba/HNF6binding sites (“Rev-erba/HNF6-exo sites”) wereenriched for lipid metabolic processes (fig. S5C),similar to Rev-erba–specific gene regulation. In-deed, enhancer RNAs (eRNAs) at these sites boundby Rev-erba and HNF6 had a robust circadianexpression pattern (Fig. 2D) and were markedlyup-regulated in livers depleted of Rev-erba, in-dicating active repression of enhancer functionat these sites (Fig. 2E) (22).To test whether the binding of Rev-erba to the

genome can be indirect, we used a mouse mod-el with a conditional deletion of the Rev-erbaDNA binding domain (DBD). These mice havebeen previously studied as a model of Rev-erbadeletion (12, 23), but the targeting strategy is pre-dicted to lead to in-frame deletion of the DBD (fig.S6A), and Rev-erba immunoblot of mouse liverafter Cre-recombination revealed an abundantspecies at the approximate molecular weightof the protein lacking the DBD (fig. S6B). Theidentity of this protein as full-length Rev-erbalacking its DBD was confirmed by mass spec-trometric analysis of Rev-erba immunoprecipi-tates from recombined liver extracts (fig. S6C).Thus, this model is actually a knock-in of a DBDmutation, rather than a complete knockout of theRev-erba protein. We studied the function of thisRev-erba DBD mutant in mice whose livers werealso depleted of Rev-erbb to eliminate its com-pensatory effects (6, 12).ChIP-seq analysis of Rev-erba in livers express-

ing only the Rev-erba DBD mutant (“DBDm”) re-vealed a comparable level of binding at a subsetof wild-type (WT) sites (“DBD-independent sites”),whereas binding was markedly reduced at manyother sites (“DBD-dependent sites”) (Fig. 2E andfig. S7, A and B). HNF6 ChIP-seq signals (24) weremore enriched at Rev-erba DBD-independentsites than at the DBD-dependent sites (fig. S7C),suggesting that HNF6 might tether Rev-erba tothe DNA even in the absence of Rev-erba DBDdomain.DBD-dependent sites were enriched in RORE

as well as dimeric RevDR2 motifs, in agreementwith direct DNA binding (fig. S7D). These mo-tifs are also recognized by ROR, and indeed thebinding of RORa at these sites decreased marked-ly at ZT10, when Rev-erba competition is stron-gest (fig. S7E). RevDR2 motifs were depleted inDBD-independent sites, whereas ROREs still existin a minority of sites (fig. S7D), suggesting thatthe RORE may facilitate, but is not required for,DBD-independent binding. The HNF6 motifwas markedly enriched at DBD-independentsites (fig. S7D), and Rev-erba binding at Rev-erba/HNF6-exo sites was comparable between WT

and DBDm mice, following the same pattern asthat seen at DBD-independent sites (fig. S7F).The simultaneous binding of Rev-erba and HNF6at these sites was confirmed by ChIP-re-ChIP ex-periments in WT liver, whereas HNF6 and Rev-erba were not colocalized at DBD-dependent sites(Fig. 2G and fig. S7G). Enhancer RNA transcrip-tion showed circadian oscillation in phase ZT22,at DBD-dependent and -independent sites (fig.S7H), suggesting active repression of Rev-erbain both cases. In agreement with Rev-erba func-tioning by recruiting the co-repressor complex,

HDAC3 binding at Rev-erba sites was reducedin Rev-erb–depleted livers (fig. S7I). In addition,the HDAC3 ChIP-seq signal in the DBDm wasreduced at DBD-dependent sites but not at sitesthat are DBD-independent (Fig. 2H), suggestingactive repression by Rev-erba via recruitmentof HDAC3.To determine whether HNF6 is required for

Rev-erba DBD-independent binding, we per-formed ChIP-seq for Rev-erba in the liver of129S1/SvlmJ mice and compared this result withthat obtained in the C57BL/6J mice. The two

1490 26 JUNE 2015 • VOL 348 ISSUE 6242 sciencemag.org SCIENCE

Fig. 2. Rev-erba binds to the genome using both DBD-dependent and DBD-independent mecha-nisms. (A) Genome browser view of Rev-erba and HNF6 ChIP-exo signals under Rev-erba ChIP-seqpeaks near clock and metabolic genes. Blue bars indicate locations of RevDR2/RORE and HNF6 motif.(B) Highly enriched known motifs found in Rev-erba ChIP-exo peak pairs 22 to 26 base pairs apart. (C)Heat map showing 5′-end tag densities of Rev-erba ChIP-exo centered at HNF6 motifs within 1108 peakpairs. Red and blue indicate tag density on the plus and minus strand, respectively. (D) Mean relativeeRNA transcription (22) at Rev-erba/HNF6-exo sites throughout 24-hour light–dark cycles. Data weredouble plotted for clearer visualization. (E) eRNA tag density (22) centered at Rev-erba/HNF6-exo sitesnear Rev-erba target genes, in Rev-erba KO and WT liver. (F) Heat map showing Rev-erba ChIP-seq tagdensities (at ZT10) in WT, DBD mutant (DBDm), and Rev-erba KO (aKO) mice, at DBD-dependentand -independent sites identified among 5792 high-confidence Rev-erba peaks (peak height > 1 rpm,WT/Rev-erba KO > 3). (G) Sequential ChIP of Rev-erba followed by either HNF6 or immunoglobulinG ChIP in WTand DBDmmouse liver at ZT10. Data are expressed as mean T SEM (* Student’s t test, P < 0.05,n = 3 or 4 mice per group). (H) Binding of HDAC3 at DBD-dependent and -independent sites in WTand DBDm

liver (N.S., Student’s t test, P > 0.05).

RESEARCH | REPORTSon F

ebruary 8, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 4: Discrete functions of nuclear receptor Rev-erbα couple ... · RevDR2 and retinoid-related orphan recep-tor (ROR)–response element (RORE) were the most enriched motifs at Rev-erba

strains differ by ~5.4 million single-nucleotidepolymorphisms (SNPs), and SNPs were pre-dicted to cripple the HNF6 motif at 107 Rev-erb

binding sites in C57BL/6J mice and 71 Rev-erbbinding sites in 129S1/SvlmJ mice. Notably,Rev-erba binding was markedly diminished at

the sites in which the SNPs disrupt either theRevDR2/RORE or the HNF6 motif in C57BL/6Jor 129S1/SvlmJ mice, whereas Rev-erba bindingat random SNPs tended to be unaffected (Fig. 3,A and B, and fig. S8A). Specific examples ofstrain-dependent binding of Rev-erba at HNF6sites are shown in Fig. 3, A and B, and differentialbinding in the two mouse strains was confirmedby ChIP–polymerase chain reaction (PCR) forHNF6 and Rev-erba (fig. S8, B and C). Interest-ingly, although the HNF6 motif was most signif-icantly associated with strain specific Rev-erbabinding by the same analysis, we also foundsignificant association with motifs for severalother TFs that play important roles in liver func-tion, suggesting involvement of other partners inRev-erba binding in the absence of RORE andRevDR2 (Fig. 3C).The preserved binding of HDAC3, mediated by

Rev-erba DBD mutant, at metabolic genes sug-gested that hepatic expression of these genesmight also be intact relative to livers of mice inwhich the Rev-erba protein is deleted. To testthis hypothesis, we compared the gene expres-sion changes in mice lacking Rev-erba in liverwith published results using the DBDm mousemodel used here, in which Rev-erba is con-verted to the DBD mutant and Rev-erbb is alsodeleted (12). Circadian clock genes were dere-pressed in both situations, demonstrating thatthe regulation of these genes required directbinding at RevDR2/RORE sites by Rev-erba.Overall, only ~25% of Rev-erba target genes thatwere derepressed in Rev-erba KO mice were alsoderepressed in the DBDm mice (“DBD-dependentRev-erba targets”) (Fig. 4A). Genes derepressed spe-cifically in Rev-erba KO mice (“DBD-independentRev-erba targets”) showed circadian expres-sion peaking at ZT22 (fig. S9A) and were en-riched for lipid metabolic functions (fig. S9B),suggesting that Rev-erba regulates circadian

SCIENCE sciencemag.org 26 JUNE 2015 • VOL 348 ISSUE 6242 1491

Fig. 4. DBD-independent Rev-erba sites reg-ulate metabolic genes in liver. (A) Top pan-el shows the number of DBD-dependent and-independent Rev-erba target genes identified withmicroarrays in Rev-erba KO (9) and DBDm mice(12). Bar graph shows ratios of DBD-independentand -dependent Rev-erba/HDAC3 binding sites(29) located near two groups of Rev-erba tar-get genes (P value from hypergeometric test). (B)mRNA expression of lipid metabolic genes nor-malized to Arpp, measured by reverse transcription–quantitative polymerase chain reaction (RT-QPCR),in livers of Rev-erba KO mice and WTmice at ZT10.(C) mRNA expression of lipid metabolic genesnormalized to Arpp, measured by RT-QPCR, inlivers of Rev-erba DBDm (Rev-erba/b double floxedmice injected with adeno-associated virus express-ing cre-recombinase under a hepatocyte-specificthyroxine-binding globulin promoter (AAV-Tbg-Cre)) or control mice (floxed mice injected withAAV-Tbg–green fluorescent protein (GFP)) at ZT10.

Data are expressed as mean T SEM (* Student’s t test, P < 0.05, n = 4 mice per group). (D) Hepatic triglyceride (TG)concentrations in the same mice as in (B). (E) HepaticTG concentrations in mice as in (C) (* Student’s t test, P < 0.05, n = 4 miceper group).

Fig. 3. SNP-associatedstrain-specific occu-pancy suggests HNF6-mediated binding ofRev-erba. (A) Heat mapshowing log2 fold changesof Rev-erba binding in 129and B6 mice. The leftcolumn in the heat mapcontains 141 Rev-erbapeaks where RevDR2/RORE motif scores arehigher in 129 mice andlower in B6 mice, owing tothe SNPs (illustrated inthe left panel). Similarly,the right column contains101 SNP-bearing Rev-erbapeaks with better RevDR2/RORE in the B6 genome.P value was calculated withStudent’s t test. (B) Sameanalysis as in (A), focusingon SNPs disrupting HNF6motif under Rev-erbapeaks. (C) Heat mapshowing –log10 P valuesfor other motifs that areenriched in DBD-independent Rev-erbapeaks.

RESEARCH | REPORTSon F

ebruary 8, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 5: Discrete functions of nuclear receptor Rev-erbα couple ... · RevDR2 and retinoid-related orphan recep-tor (ROR)–response element (RORE) were the most enriched motifs at Rev-erba

lipid metabolic genes independent of its DBD.In support of this, DBD-independent Rev-erba/HDAC3 sites were enriched near DBD-independentRev-erba targets, whereas DBD-dependent Rev-erba/HDAC3 sites, where Rev-erba and HDAC3binding was markedly reduced in DBDm mice,were more enriched near DBD-dependent Rev-erba targets (Fig. 4A). Examples of the deletion-specific regulation of metabolic genes are shownin Fig. 4, B and C. Consistent with preservedmetabolic gene expression in the DBDm micelivers, these mice did not display hepatosteatosisas is characteristic of the mice with completedeletion of Rev-erba (Fig. 4, D and E, and fig.S9C) (6, 9).These findings demonstrate that Rev-erba has

a DBD-independent function that contributes toits regulation of liver metabolism. Other nuclearreceptors, including estrogen receptor and glu-cocorticoid receptor, have DBD-independent ac-tivities through protein-protein interactions withother TFs, either directly or indirectly (25, 26). Inliver, Rev-erba is tethered to chromatin by he-patic lineage–determining TFs (fig. S10), and thismechanism of binding explains much of the non-overlapping cistromes of Rev-erba in differenttissues, as well as the large proportion of bind-ing sites without the RevDR2/RORE motif. Inliver, the tethered cistrome is more enriched forgenes with specialized function in hepatic me-tabolism, whereas the DBD-dependent cistromeis enriched for circadian clock genes and com-mon to multiple tissues.Circadian clocks and metabolism are tightly

connected (1, 3, 4), and Rev-erba has emergedas a transcriptional link from circadian rhythmsto metabolism in multiple tissues (8). Our find-

ings delineate a molecular hierarchy that gov-erns how the clock is wired with metabolism.Direct competition between Rev-erb and RORprovides a universal mechanism for self-sustainedcontrol of the molecular clock across all tissues.On top of this basic landscape, circadianly ex-pressed Rev-erb uses lineage-determination fac-tors to convey a tissue-specific epigenomic rhythmthat, through co-repressor and HDAC3, regulatesmetabolism tailored to the specific need of thattissue. These two modes of action may bestowon Rev-erba the ability to stabilize the circadianoscillations of clock gene, while coupling livermetabolism to environmental and metabolicchanges, perhaps through its endogenous ligandheme (27, 28). This raises the possibility thatsynthetic ligands that specifically affect Rev-erbainteraction with NCoR/HDAC3 without disrupt-ing DNA binding could modulate liver metabo-lism with lesser effects on the integrity of thecircadian clock.

REFERENCES AND NOTES

1. D. Feng, M. A. Lazar, Mol. Cell 47, 158 (2012).2. G. Asher, P. Sassone-Corsi, Cell 161, 84 (2015).3. J. S. Takahashi, H. K. Hong, C. H. Ko, E. L. McDearmon,

Nat. Rev. Genet. 9, 764 (2008).4. J. Bass, J. S. Takahashi, Science 330, 1349 (2010).5. P. E. Hardin, S. Panda, Curr. Opin. Neurobiol. 23, 724

(2013).6. A. Bugge et al., Genes Dev. 26, 657 (2012).7. N. Preitner et al., Cell 110, 251 (2002).8. L. J. Everett, M. A. Lazar, Trends Endocrinol. Metab. 25, 586

(2014).9. D. Feng et al., Science 331, 1315 (2011).10. Z. Gerhart-Hines et al., Nature 503, 410 (2013).11. E. Woldt et al., Nat. Med. 19, 1039 (2013).12. H. Cho et al., Nature 485, 123 (2012).13. L. Yin, M. A. Lazar, Mol. Endocrinol. 19, 1452 (2005).14. V. Giguère et al., Genes Dev. 8, 538 (1994).

15. B. M. Forman et al., Mol. Endocrinol. 8, 1253 (1994).16. H. P. Harding, M. A. Lazar, Mol. Cell. Biol. 15, 4791

(1995).17. E. Stashi et al., Cell Rep. 6, 633 (2014).18. L. A. Solt, T. P. Burris, Trends Endocrinol. Metab. 23, 619

(2012).19. Y. Takeda, R. Jothi, V. Birault, A. M. Jetten, Nucleic Acids Res.

40, 8519 (2012).20. H. S. Rhee, B. F. Pugh, Cell 147, 1408 (2011).21. L. Wang et al., Nucleic Acids Res. 42, e156 (2014).22. B. Fang et al., Cell 159, 1140 (2014).23. M. T. Lam et al., Nature 498, 511 (2013).24. A. J. Faure et al., Genome Res. 22, 2163 (2012).25. N. Heldring et al., Mol. Endocrinol. 25, 564 (2011).26. C. K. Glass, K. Saijo, Nat. Rev. Immunol. 10, 365 (2010).27. L. Yin et al., Science 318, 1786 (2007).28. S. Raghuram et al., Nat. Struct. Mol. Biol. 14, 1207 (2007).29. Materials and methods are available as supplementary

materials on Science Online.

ACKNOWLEDGMENTS

ChIP-seq and microarray data have been deposited in theGene Expression Omnibus (GSE67973). We thank S. Sidoli andB. A. Garcia for assistance with mass spectrometry. Weacknowledge the Functional Genomics Core and the Viral VectorCore of the Penn Diabetes Research Center (P30 DK19525)for next-generation sequencing and virus preparation, respectively.We thank the Penn Digestives Disease Center Morphology Core(P30 DK050306) for histology studies and the MolecularProfiling Core for microarray analysis. This work was supportedby NIH R01 DK45586 (M.A.L.), K08 DK094968 (R.E.S.), R00DK099443 (Z.S.), R01 DK098542 (D.J.S.), F32 DK102284 (S.M.A.),F30 DK104513 (M.J.E.), T32 GM0008275 (J.R.R.), and the CoxMedical Research Institute.

SUPPLEMENTARY MATERIALS

www.sciencemag.org/content/348/6242/1488/suppl/DC1Materials and MethodsFigs. S1 to S10Table S1References (30–39)

14 April 2015; accepted 22 May 2015Published online 4 June 2015;10.1126/science.aab3021

1492 26 JUNE 2015 • VOL 348 ISSUE 6242 sciencemag.org SCIENCE

RESEARCH | REPORTSon F

ebruary 8, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 6: Discrete functions of nuclear receptor Rev-erbα couple ... · RevDR2 and retinoid-related orphan recep-tor (ROR)–response element (RORE) were the most enriched motifs at Rev-erba

couple metabolism to the clockαDiscrete functions of nuclear receptor Rev-erb

Remsberg, Jennifer Jager, Raymond E. Soccio, David J. Steger and Mitchell A. LazarYuxiang Zhang, Bin Fang, Matthew J. Emmett, Manashree Damle, Zheng Sun, Dan Feng, Sean M. Armour, Jarrett R.

originally published online June 4, 2015DOI: 10.1126/science.aab3021 (6242), 1488-1492.348Science 

, this issue p. 1488Sciencefactors that regulate metabolic gene expression in a tissue-specific manner.displacing a competing transcription factor. At metabolic genes, it interacts not with DNA but with other transcriptionand metabolic genes through distinct mechanisms. At clock genes, it binds directly to a specific DNA sequence,

regulates expression of both clockα found that a multitasking transcription factor called Rev-erb-et al.disease. Zhang may increase the risk of metabolic−−for example, through shift work−−Chronic disruption of our circadian rhythms

Multitasking around the clock

ARTICLE TOOLS http://science.sciencemag.org/content/348/6242/1488

MATERIALSSUPPLEMENTARY http://science.sciencemag.org/content/suppl/2015/06/03/science.aab3021.DC1

CONTENTRELATED

http://stke.sciencemag.org/content/sigtrans/9/429/ec120.abstracthttp://stke.sciencemag.org/content/sigtrans/7/342/re6.fullhttp://stke.sciencemag.org/content/sigtrans/5/244/pt4.fullhttp://stke.sciencemag.org/content/sigtrans/2003/209/tr7.abstract

REFERENCES

http://science.sciencemag.org/content/348/6242/1488#BIBLThis article cites 38 articles, 9 of which you can access for free

PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

Terms of ServiceUse of this article is subject to the

is a registered trademark of AAAS.ScienceScience, 1200 New York Avenue NW, Washington, DC 20005. The title (print ISSN 0036-8075; online ISSN 1095-9203) is published by the American Association for the Advancement ofScience

Copyright © 2015, American Association for the Advancement of Science

on February 8, 2020

http://science.sciencem

ag.org/D

ownloaded from