We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Published Online:https://doi.org/10.4155/fmc.11.9

Research efforts spanning the past two decades have established a clear link between nuclear receptor function, regulation of the circadian clock and lipid homeostasis. As such, this family of receptors represents an important area of research. Recent advances in the field have identified two nuclear receptor subfamilies, the REV-ERBs and the ‘retinoic acid receptor-related orphan receptors’ (RORs), as critical regulators of the circadian clock with significant roles in lipid homeostasis. In this review, the latest information garnered from cutting-edge research on these two nuclear receptor subfamilies will be discussed. Through direct targeting of the REV-ERBs and RORs with synthetic ligands, generation of novel tools aimed at characterizing their function in vivo have been developed, which may lead to novel therapeutics for the treatment of metabolic disorders.

Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

Bibliography

  • Sladek FM. What are nuclear receptor ligands? Mol. Cell Endocrinol.334(1–2),3–13 (2010).
  • Burris TP. Nuclear hormone receptors for heme: REV-ERBα and REV-ERBβ are ligand-regulated components of the mammalian clock. Mol. Endocrinol.22(7),1509–1520 (2008).
  • Mangelsdorf DJ, Thummel C, Beato M et al. The nuclear receptor superfamily: the second decade. Cell83(6),835–839 (1995).
  • Mangelsdorf DJ, Evans RM. The RXR heterodimers and orphan receptors. Cell83(6),841–850 (1995).
  • Raghuram S, Stayrook KR, Huang P et al. Identification of heme as the ligand for the orphan nuclear receptors REV-ERBα and REV-ERBβ. Nat. Struct. Mol. Biol.14(12),1207–1213 (2007).▪▪ In conjunction with [6], these studies identified heme as an endogenous physiological ligand for the orphan nuclear receptors REV-ERBα and β.
  • Yin L, Wu N, Curtin JC et al. REV-ERBα, a heme sensor that coordinates metabolic and circadian pathways. Science318(5857),1786–1789 (2007).▪▪ In conjunction with [5], these studies identified heme as an endogenous physiological ligand for the orphan nuclear receptors REV-ERBα and β.
  • Becker-Andre M, Andre E, DeLamarter JF. Identification of nuclear receptor mRNAs by RT-PCR amplification of conserved zinc-finger motif sequences. Biochem. Biophys. Res. Commun.194(3),1371–1379 (1993).
  • Giguere V, Tini M, Flock G et al. Isoform-specific amino-terminal domains dictate DNA-binding properties of RORα, a novel family of orphan hormone nuclear receptors. Genes Dev.8(5),538–553 (1994).
  • Carlberg C, Hooft van Huijsduijnen R, Staple JK, DeLamarter JF, Becker-André M. RZRs, a new family of retinoid-related orphan receptors that function as both monomers and homodimers. Mol. Endocrinol.8(6),757–770 (1994).
  • 10  Hirose T, Smith RJ, Jetten AM. RORγ: the third member of ROR/RZR orphan receptor subfamily that is highly expressed in skeletal muscle. Biochem. Biophys. Res. Commun.205(3),1976–1983 (1994).
  • 11  Jetten AM. Retinoid-related orphan receptors (RORs): critical roles in development, immunity, circadian rhythm, and cellular metabolism. Nucl. Recept. Signal.7,e003 (2009).
  • 12  Wang Y, Kumar N, Solt LA et al. Modulation of retinoic acid receptor-related orphan receptor α and γ activity by 7-oxygenated sterol ligands. J. Biol. Chem.285(7),5013–5025 (2010).
  • 13  Burke L, Downes M, Carozzi A, Giguère V, Muscat GE. Transcriptional repression by the orphan steroid receptor RVR/REV-ERBβ is dependent on the signature motif and helix 5 in the E region: functional evidence for a biological role of RVR in myogenesis. Nucleic Acids Res.24(18),3481–3489 (1996).
  • 14  Downes M, Burke LJ, Bailey PJ, Muscat GE. Two receptor interaction domains in the corepressor, N-CoR/RIP13, are required for an efficient interaction with REV-ERBAα and RVR: physical association is dependent on the E region of the orphan receptors. Nucleic Acids Res.24(22),4379–4386 (1996).
  • 15  Zamir I, Harding HP, Atkins GB et al. A nuclear hormone receptor corepressor mediates transcriptional silencing by receptors with distinct repression domains. Mol. Cell Biol.16(10),5458–5465 (1996).
  • 16  Harding HP, Lazar MA. The orphan receptor REV-ERBAα activates transcription via a novel response element. Mol. Cell Biol.13(5),3113–3121 (1993).
  • 17  Harding HP, Lazar MA. The monomer-binding orphan receptor REV-ERB represses transcription as a dimer on a novel direct repeat. Mol. Cell Biol.15(9),4791–4802 (1995).
  • 18  Green CB, Takahashi JS, Bass J. The meter of metabolism. Cell134(5),728–742 (2008).
  • 19  DeBruyne JP, Weaver DR, Reppert SM. CLOCK and NPAS2 have overlapping roles in the suprachiasmatic circadian clock. Nat. Neurosci.10(5),543–545 (2007).
  • 20  Gachon F, Nagoshi E, Brown SA, Ripperger J, Schibler U. The mammalian circadian timing system: from gene expression to physiology. Chromosoma113(3),103–112 (2004).
  • 21  Duez H, Pavlic M, Lewis GF. Mechanism of intestinal lipoprotein overproduction in insulin resistant humans. Atheroscler. Suppl.9(2),33–38 (2008).
  • 22  Guillaumond F, Dardente H, Giguère V, Cermakian N. Differential control of Bmal1 circadian transcription by REV-ERB and ROR nuclear receptors. J. Biol. Rhythms20(5),391–403 (2005).
  • 23  Sato TK, Panda S, Miraglia LJ et al. A functional genomics strategy reveals Rora as a component of the mammalian circadian clock. Neuron43(4),527–537 (2004).▪ Study suggesting that the opposing activities of the nuclear receptors RORa and REV-ERBa are important in the maintenance of the core mammalian circadian clock.
  • 24  Akashi M, Takumi T. The orphan nuclear receptor RORα regulates circadian transcription of the mammalian core-clock Bmal1. Nat. Struct. Mol. Biol.12(5),441–448 (2005).▪▪ Study identified RORα as a functional component of the cell-autonomous core circadian clock.
  • 25  Crumbley C, Wang Y, Kojetin DJ, Burris TP. Characterization of the core mammalian clock component, NPAS2, as a REV-ERB{α}/ROR{α} target gene. J. Biol. Chem.285,35386–35392 (2010).
  • 26  Preitner N, Damiola F, Molina L et al. The orphan nuclear receptor REV-ERBα controls circadian transcription within the positive limb of the mammalian circadian oscillator. Cell110(2),251–260 (2002).▪▪ Study identified REV-ERBα as being an integral part of the regulation of the circadian clock.
  • 27  Masana MI, Sumaya IC, Becker-Andre M, Dubocovich ML. Behavioral characterization and modulation of circadian rhythms by light and melatonin in C3H/HeN mice homozygous for the RORβ knockout. Am. J. Physiol. Regul. Integr. Comp. Physiol.292(6),R2357–R2367 (2007).
  • 28  Schaeren-Wiemers N, André E, Kapfhammer JP, Becker-André M. The ExDression pattern of the orphan nuclear receptor RORβ in the developing and adult rat nervous system suggests a role in the processing of sensory information and in circadian rhythm. Eur. J. Neurosci.9(12),2687–2701 (1997).
  • 29  Liu AC, Tran HG, Zhang EE, Priest AA, Welsh DK, Kay SA. Redundant function of REV-ERBα and β and non-essential role for Bmal1 cycling in transcriptional regulation of intracellular circadian rhythms. PLoS Genet.4(2),e1000023 (2008).
  • 30  Ando H, Yanagihara H, Hayashi Y et al. Rhythmic messenger ribonucleic acid expression of clock genes and adipocytokines in mouse visceral adipose tissue. Endocrinology146(12),5631–5636 (2005).
  • 31  Panda S, Antoch MP, Miller BH et al. Coordinated transcription of key pathways in the mouse by the circadian clock. Cell109(3),307–320 (2002).
  • 32  Hamilton BA, Frankel WN, Kerrebrock AW et al. Disruption of the nuclear hormone receptor RORα in staggerer mice. Nature379(6567),736–739 (1996).
  • 33  Vu-Dac N, Gervois P, Grötzinger T et al. Transcriptional regulation of apolipoprotein A-I gene expression by the nuclear receptor RORα. J. Biol. Chem.272(36),22401–22404 (1997).
  • 34  Raspe E, Duez H, Gervois P et al. Transcriptional regulation of apolipoprotein C-III gene expression by the orphan nuclear receptor RORα. J. Biol. Chem.276(4),2865–2871 (2001).
  • 35  Mamontova A, Séguret-Macé S, Esposito B et al. Severe atherosclerosis and hypoαlipoproteinemia in the staggerer mouse, a mutant of the nuclear receptor RORα. Circulation98(24),2738–2743 (1998).
  • 36  Lau P, Fitzsimmons RL, Raichur S, Wang SC, Lechtken A, Muscat GE. The orphan nuclear receptor, RORα, regulates gene expression that controls lipid metabolism: staggerer (SG/SG) mice are resistant to diet-induced obesity. J. Biol. Chem.283(26),18411–18421 (2008).
  • 37  Kang HS, Angers M, Beak JY et al. Gene expression profiling reveals a regulatory role for RORα and RORγ in Phase I and Phase II metabolism. Physiol. Genomics31(2),281–294 (2007).
  • 38  Wada T, Kang HS, Angers M et al. Identification of oxysterol 7α-hydroxylase (Cyp7b1) as a novel retinoid-related orphan receptor α (RORα) (NR1F1) target gene and a functional cross-talk between RORα and liver X receptor (NR1H3). Mol. Pharmacol.73(3),891–899 (2008).
  • 39  Kharitonenkov A, Shiyanova TL, Koester A et al. FGF-21 as a novel metabolic regulator. J. Clin. Invest.115(6),1627–1635 (2005).
  • 40  Mayer D. The circadian rhythm of synthesis and catabolism of cholesterol. Arch. Toxicol.36(3–4),267–276 (1976).
  • 41  Edwards PA, Muroya H, Gould RG. In vivo demonstration of the circadian thythm of cholesterol biosynthesis in the liver and intestine of the rat. J. Lipid Res.13(3),396–401 (1972).
  • 42  Chopra AR, Louet JF, Saha P et al. Absence of the SRC-2 coactivator results in a glycogenopathy resembling Von Gierke’s disease. Science322(5906),1395–1399 (2008).▪ Identified G6Pase as an ROR target gene, and linked ROR transcriptional activity with glucose metabolism.
  • 43  Badman MK, Pissios P, Kennedy AR, Koukos G, Flier JS, Maratos-Flier E. Hepatic fibroblast growth factor 21 is regulated by PPARα and is a key mediator of hepatic lipid metabolism in ketotic states. Cell Metab.5(6),426–437 (2007).
  • 44  Inagaki T, Dutchak P, Zhao G et al. Endocrine regulation of the fasting response by PPARα-mediated induction of fibroblast growth factor 21. Cell Metab.5(6),415–425 (2007).
  • 45  Duez H, Staels B. REV-ERBα: an integrator of circadian rhythms and metabolism. J. Appl. Physiol.107(6),1972–1980 (2009).
  • 46  Chomez P, Neveu I, Mansén A et al. Increased cell death and delayed development in the cerebellum of mice lacking the REV-ERB(α) orphan receptor. Development127(7),1489–1498 (2000).
  • 47  Raspe E, Duez H, Mansén A et al. Identification of REV-ERBα as a physiological repressor of apoC-III gene transcription. J. Lipid Res.43(12),2172–2179 (2002).
  • 48  Shachter NS. Apolipoproteins C-I and C-III as important modulators of lipoprotein metabolism. Curr. Opin. Lipidol.12(3),297–304 (2001).
  • 49  Wang J, Yin L, Lazar MA. The orphan nuclear receptor REV-ERBα regulates circadian expression of plasminogen activator inhibitor type 1. J. Biol. Chem.281(45),33842–33848 (2006).
  • 50  Anzulovich A, Mir A, Brewer M, Ferreyra G, Vinson C, Baler R. Elovl3: a model gene to dissect homeostatic links between the circadian clock and nutritional status. J. Lipid Res.47(12),2690–2700 (2006).
  • 51  Vu-Dac N, Chopin-Delannoy S, Gervois P et al. The nuclear receptors peroxisome proliferator-activated receptor α and REV-ERBα mediate the species-specific regulation of apolipoprotein A-I expression by fibrates. J. Biol. Chem.273(40),25713–25720 (1998).
  • 52  Estall JL, Ruas JL, Choi CS et al. PGC-1α negatively regulates hepatic FGF21 expression by modulating the heme/REV-ERB(α) axis. Proc. Natl Acad. Sci. USA106(52),22510–22515 (2009).
  • 53  Zheng J, Shan Y, Lambrecht RW, Donohue SE, Bonkovsky HL. Differential regulation of human ALAS1 mRNA and protein levels by heme and cobalt protoporphyrin. Mol. Cell. Biochem.319(1–2),153–161 (2008).
  • 54  Ramakrishnan SN, Lau P, Burke LJ, Muscat GE. REV-ERBβ regulates the expression of genes involved in lipid absorption in skeletal muscle cells: evidence for cross-talk between orphan nuclear receptors and myokines. J. Biol. Chem.280(10),8651–8659 (2005).
  • 55  Gronemeyer H, Gustafsson JA, Laudet V. Principles for modulation of the nuclear receptor superfamily. Nat. Rev. Drug Discov.3(11),950–964 (2004).
  • 56  Rosenfeld MG, Lunyak VV, Glass CK. Sensors and signals: a coactivator/corepressor/epigenetic code for integrating signal-dependent programs of transcriptional response. Genes Dev.20(11),1405–1428 (2006).
  • 57  Wiesenberg I, Missbach M, Kahlen JP, Schräder M, Carlberg C. Transcriptional activation of the nuclear receptor RZRα by the pineal gland hormone melatonin and identification of CGP 52608 as a synthetic ligand. Nucleic Acids Res.23(3),327–333 (1995).
  • 58  Kallen JA, Schlaeppi JM, Bitsch F et al. X-ray structure of the hRORα LBD at 1.63 Å: structural and functional data that cholesterol or a cholesterol derivative is the natural ligand of RORα. Structure10(12),1697–1707 (2002).▪▪ Solved the first crystal structure of RORa’s ligand-binding domain (LBD) revealing cholesterol bound within the ligand-binding pocket.
  • 59  Kallen JA, Schlaeppi JM, Bitsch F, Delhon I, Fournier B. Crystal structure of the human RORα ligand binding domain in complex with cholesterol sulfate at 2.2 Å. J. Biol. Chem.279(14),14033–14038 (2004).▪ Through x-ray crystallography, this group identified cholesterol sulfate within the ligand-binding domain of RORα, and characterized it as another endogenous ligand for ROR.
  • 60  Stehlin C, Wurtz JM, Steinmetz A et al. X-ray structure of the orphan nuclear receptor RORβ ligand-binding domain in the active conformation. EMBO J.20(21),5822–5831 (2001).
  • 61  Stehlin-Gaon C, Willmann D, Zeyer D et al. All-trans retinoic acid is a ligand for the orphan nuclear receptor RORβ. Nat. Struct. Biol.10(10),820–825 (2003).
  • 62  Wang Y, Kumar N, Crumbley C, Griffin PR, Burris TP. A second class of nuclear receptors for oxysterols: regulation of RORα and RORγ activity by 24S-hydroxycholesterol (cerebrosterol). Biochim. Biophys. Acta.1801(8),917–923 (2010).
  • 63  Jin L, Martynowski D, Zheng S, Wada T, Xie W, Li Y. Structural basis for hydroxycholesterols as natural ligands of orphan nuclear receptor RORγ. Mol. Endocrinol.24(5),923–929 (2010).▪▪ Identified several hydroxycholesterols as endogenous ligands for RORγ through crystallographic methods.
  • 64  Adare A, Afanasiev S, Aidala C et al. Enhanced production of direct photons in Au + Au collisions at square root(S(NN)) = 200 GeV and implications for the initial temperature. Phys. Rev. Lett.104(13),132301 (2010).
  • 65  Pardee KI, Xu X, Reinking J et al. The structural basis of gas-responsive transcription by the human nuclear hormone receptor REV-ERBβ. PLoS Biol.7(2),e43 (2009).▪ Provided the crystal structure of REV-ERBβ when bound to heme.
  • 66  Woo EJ, Jeong DG, Lim MY et al. Structural insight into the constitutive repression function of the nuclear receptor REV-ERBβ. J. Mol. Biol.373(3),735–744 (2007).
  • 67  Phelan CA, Gampe RT Jr, Lambert MH et al. Structure of REV-ERBα bound to N-CoR reveals a unique mechanism of nuclear receptor-co-repressor interaction. Nat. Struct. Mol. Biol.17(7),808–814 (2010).▪ Provides the structure of the LBD of REV-ERBα when bound to the corepressor NCoR and suggests that REV-ERBα may be active in the absence of heme.
  • 68  Kumar N, Solt LA, Conkright JJ et al. The benzenesulfoamide T0901317 [N-(2,2,2-trifluoroethyl)-N-[4-[2,2,2-trifluoro-1-hydroxy-1-(trifluorometh yl)ethyl]phenyl]-benzenesulfonamide] is a novel retinoic acid receptor-related orphan receptor-α/γ inverse agonist. Mol. Pharmacol.77(2),228–236 (2010).▪▪ Identifies the first synthetic ligand that modulates both RORα and RORγ activity through direct binding of the receptors, resulting in the modulation of the receptor’s ability to interact with transcriptional cofactor proteins.
  • 69  Houck KA, Borchert KM, Hepler CD et al. T0901317 is a dual LXR/FXR agonist. Mol. Genet. Metab.83(1–2),184–187 (2004).
  • 70  Mitro N, Vargas L, Romeo R, Koder A, Saez E. T0901317 is a potent PXR ligand: implications for the biology ascribed to LXR. FEBS Lett.581(9),1721–6172 (2007).
  • 71  Wang Y, Kumar N, Nuhant P et al. Identification of SR1078, a synthetic agonist for the orphan nuclear receptors RORα and RORγ. ACS Chem. Biol.5(11),1029–1034 (2010).▪▪ Identified and characterized the first selective RORα/γ synthetic agonist.
  • 72  Kumar N, Kojetin DJ, Solt LA et al. Identification of ML176 (SR3335): a synthetic RORα selective inverse agonist. ACS Chem. Biol. DOI: 10.1021/cb1002762 (2010). (Epub ahead of print).
  • 73  Meng QJ, McMaster A, Beesley S et al. Ligand modulation of REV-ERBα function resets the peripheral circadian clock in a phasic manner. J. Cell Sci.121(Pt. 21),3629–3635 (2008).▪▪ Identified and characterized the first selective REV-ERBα synthetic agonist, GSK4112.
  • 74  Fu M, Sun T, Bookout AL et al. A nuclear receptor atlas: 3T3-L1 adipogenesis. Mol. Endocrinol.19(10),2437–2450 (2005).
  • 75  Fontaine C, Rigamonti E, Pourcet B et al. The orphan nuclear receptor REV-ERBα is a peroxisome proliferator-activated receptor (PPAR) γ target gene and promotes PPARγ-induced adipocyte differentiation. J. Biol. Chem.278(39),37672–37680 (2003).
  • 76  Chawla A, Lazar MA. Induction of REV-ERBAα, an orphan receptor encoded on the opposite strand of the α-thyroid hormone receptor gene, during adipocyte differentiation. J. Biol. Chem.268(22),16265–16269 (1993).
  • 77  Duez H, Duhem C, Laitinen S et al. Inhibition of adipocyte differentiation by RORα. FEBS Lett.583(12),2031–2036 (2009).
  • 78  Austin S, Medvedev A, Yan ZH, Adachi H, Hirose T, Jetten AM. Induction of the nuclear orphan receptor RORγ during adipocyte differentiation of D1 and 3T3-L1 cells. Cell Growth Differ.9(3),267–276 (1998).
  • 79  Kumar N, Solt LA, Wang Y et al. Regulation of adipogenesis by natural and synthetic REV-ERB ligands. Endocrinology151(7),3015–3025 (2010).▪ Series of studies suggesting that heme, functioning as a REV-ERB ligand, is an important regulator of adipogenesis and that synthetic ligands can modulate adipogenesis and may be useful therapeutics for the treatment of metabolic diseases.
  • 80  Grant D, Yin L, Collins JL et al. GSK4112, a small-molecule chemical probe for the cell biology of the nuclear heme receptor REV-analogα. ACS Chem. Biol.5(10),925–932 (2010).
  • 81  Kojetin D, Wang Y, Kamenecka TM, Burris TP. Identification of SR8278, a synthetic antagonist of the nuclear heme receptor REV-ERB. ACS Chem. Biol.6(2),131–134 (2011).▪▪ Study that identified and characterized the first synthetic REV-ERB antagonist, SR8278.
  • 82  Lund J, Arendt J, Hampton SM, English J, Morgan LM. Postprandial hormone and metabolic responses amongst shift workers in Antarctica. J. Endocrinol.171(3),557–564 (2001).
  • 83  Kawachi I, Colditz GA, Stampfer MJ et al. Prospective study of shift work and risk of coronary heart disease in women. Circulation92(11),3178–3182 (1995).
  • 84  Knutsson A, Hallquist J, Reuterwall C, Theorell T, Akerstedt T. Shiftwork and myocardial infarction: a case-control study. Occup. Environ. Med.56(1),46–50 (1999).
  • 85  Ellingsen T, Bener A, Gehani AA. Study of shift work and risk of coronary events. J. R. Soc. Promot. Health127(6),265–267 (2007).
  • 86  Hermansson J, Gillander Gådin K, Karlsson B, Lindahl B, Stegmayr B, Knutsson A. Ischemic stroke and shift work. Scand. J. Work Environ. Health33(6),435–943 (2007).
  • 87  Van Cauter E, Polonsky KS, Scheen AJ. Roles of circadian rhythmicity and sleep in human glucose regulation. Endocr. Rev.18(5),716–738 (2007).
  • 88  Holterhus PM, Odendahl R, Oesingmann S et al. Classification of distinct baseline insulin infusion patterns in children and adolescents with Type I diabetes on continuous subcutaneous insulin infusion therapy. Diabetes Care30(3),568–573 (2007).
  • 89  Wang Y, Kumar N, Solt LA et al. Modulation of ROR{α} and ROR{γ} activity by 7-oxygenated sterol ligands. J. Biol. Chem.285(1),5013–5025 (2009).▪▪ Study demonstrating that 7-oxygenated sterols function as high-affinity ligands for both RORα and RORγ leading to suppression of the transcriptional activity of the receptors.
  • 90  Brown AJ, Jessup W. Oxysterols and atherosclerosis. Atherosclerosis142(1),1–28 (1999).
  • 91  Brown AJ, Leong SL, Dean RT, Jessup W. 7-hydroperoxycholesterol and its products in oxidized low-density lipoprotein and human atherosclerotic plaque. J. Lipid Res.38(9),1730–1745 (1997).
  • 92  Gelissen IC, Brown AJ, Mander EL, Kritharides L, Dean RT, Jessup W. Sterol efflux is impaired from macrophage foam cells selectively enriched with 7-ketocholesterol. J. Biol. Chem.271(30),17852–17860 (1996).
  • 93  Zieden B, Kaminskas A, Kristenson M et al. Increased plasma 7β-hydroxycholesterol concentrations in a population with a high risk for cardiovascular disease. Arterioscler. Thromb. Vasc. Biol.19(4),967–971 (1999).
  • 94  Duez H, van der Veen JN, Duhem C et al. Regulation of bile acid synthesis by the nuclear receptor REV-ERBα. Gastroenterology135(2),689–698 (2008).
  • 95  Le Martelot G, Claudel T, Gatfield D et al. REV-ERBα participates in circadian SREBP signaling and bile acid homeostasis. PLoS Biol.7(9),e1000181 (2009).