1F. Retinoic acid-related orphans: Introduction

The IUPHAR Compendium of the Pharmacology and Classification of the Nuclear Receptor Superfamily 2006 provides a useful general overview on nuclear hormone receptors. This volume is available online from Pharmacological Reviews. The following chapters from the compendium may be of interest for this particular set of receptors [4,17].

The retinoic acid-related orphan receptors alpha, beta, and gamma (RORα-γ or NR1F1-3) constitute a subfamily of nuclear receptors [28,30,73]. Each gene generates several isoforms that regulate different genes and biological processes. RORs function as ligand-dependent transcription factors that regulate the transcription of target genes by binding as monomers to ROR response elements (ROREs). Genome-wide mapping of cis-acting binding sites (cistrome) in liver and Th17 cells, identified the RORE consensus in vivo as well as genes directly regulated by RORα and/or RORγ, including several clock genes, glucose and lipid metabolic genes, and Th17 related genes [25,80,82,95].


ROR ligands

Initial crystallographic studies identified cholesterol and several of its analogs as RORα agonists [33-34] and several retinoids as inverse agonists of RORβ [76]. Studies realizing the potential of RORs antagonists in the management of autoimmune disease, metabolic syndrome, insulin resistance, and neuropsychiatric disorders [16,22-23,36,38,41,48,70,72,82,85,95-96] provided an incentive for many to identify novel ROR ligands. This led to the discovery of many ROR (ant)agonists of various chemical classes and include various oxysterols [31,91-92], other cholesterol-related compounds [96], vitamin D metabolites [71], derivatives of LXR agonist T0901317 [38-39,75], digoxin and its analogs [22], and a number of different sulfonamides [13,88]. A comprehensive summary on ROR ligands can be found in several recent reviews [13,38,45,73]. Recent studies demonstrated that intermediates of the cholesterol biosynthetic pathway can bind and regulate RORγ activity and function as endogenous ligands for RORγ and include desmosterol and its sulfate, and 4α-carboxy, 4β-methyl-zymosterol (4ACD8) [21,63].

Crystal structure analyses revealed that RORs contain a large ligand binding pocket (>700Å3) that can accommodate large ligands, such as cholesterol sulfate [31,34,63,73,76,91-92]. Analysis of ROR-agonist crystal structures further revealed that the H12 was in the active conformation thereby allowing co-activator interaction, whereas binding of inverse agonists inhibited this interaction. Certain inverse agonists were found to inhibit the binding of RORγ to ROREs, whereas the ability of RORγ to bind DNA target sites was mostly preserved with others [95]. ROR antagonists have been shown to inhibit the expression of a number of ROR target genes, including several clock genes, various glucose and lipid metabolic genes, and Th17 related genes [5,22,75,80-83,92-93,96-97].


RORγ Immune Functions

Early studies revealed that the RORγt isoform is critical for thymopoiesis and the development of secondary lymphoid organs, Peyer’s patches, and intestinal lymphoid follicles [37,40,78]. This was shown to be due to the absence of a subset of type 3 innate lymphoid cells (ILC3), referred to as lymphoid tissue inducer (LTi) cells [7,10,47]. Retinoic acid (RA) was found to control LTi cell maturation by directly regulating RORγt expression through the recruitment of RA receptors (RARs) to the promoter region of RORγt [87]. RORγt also regulates different subsets of ILC3s in the intestine [20,69]. Moreover, RORγt is essential for the differentiation of naïve T cells into Th17 cells, which have an important role in fighting bacterial and fungal infections and are implicated in several autoimmune diseases [8,24,29,55,95,97]. RORγt was shown to directly regulate the transcription of several Th17 marker genes, including Ilf17a, Il17f, Irf4, and Il23r [23,97-98,100]. Loss of RORγt expression in mice abolished the expression of many Th17 cytokines and protects against the development of several experimental autoimmune diseases as well as allergen-induced lung inflammation [24,84,97]. Inversely, overexpression of RORγt in mice induced a steroid-insensitive neutrophilic inflammation [3]. RORγ has also been reported to directly regulate the transcription of the L-phenylalanine oxidase IL41, which is involved in the control of TCR signaling [62].


RORα Immune Functions

RORα has a small role in Th17 differentiation [29,97], but is essential for the development and function of type 2 ILCs, which are implicated in immune defense and allergy-induced inflammation [18,61,90,94]. A role for RORα in the control of allergy-induced inflammation is supported by studies showing an association of RORA SNPs with increased susceptibility to asthma [49,51,60] and reduced Th2 cytokine production, airway hyper-reactivity, and allergic skin inflammation in RORα-deficient mice [18,27,61]. RORα has also been reported to regulate the diurnal expression of several pathogen recognition receptors, including Nod2 and the Toll-like receptors, TLR1-5 and TLR9, in the intestinal epithelium where it also directly regulates the diurnal expression of interleukin-1 receptor-associated kinase 1 (IRAK1) and Toll-interleukin 1 receptor domain containing adaptor protein (TIRAP), and several clock genes [53].


RORα/β Functions in Brain and Retina

Loss of RORα in Purkinje cells results in impairment of the hedgehog signaling pathway and cerebellar degeneration that leads to the development of a staggerer phenotype [19,77]. RORα has also been implicated in several neuropsychiatric disorders, including autism spectrum disorder [2,6,12,44,50,54]. Several genes directly regulated by RORα were found to be repressed in individuals with ASD [64-66]. It has been suggested that protecting brain cells from the damaging effects of injury and stress might be relevant to several brain disorders [32].

RORβ has a role in the regulation of circadian, motor and visual functions [30,68] and controls the cytoarchitectural patterning of neocortical neurons during mouse development [26]. GWAS studies observed a correlation between RORβ and verbal intelligence [11]. RORβ1 is critical for the differentiation of retinal progenitors into amacrine and horizontal interneurons [43]. This involves direct transcriptional regulation of Ptf1a by RORβ1. Both RORβ1 and -β2 directly regulate neural retina leucine zipper factor (NRL) [15]. In turn, NRL enhances the transcription of RORβ2. NRL and RORβ form two positive feedback loops that synergistically promote the commitment to a rod cell lineage.


ROR Functions in Metabolism

Both RORα and RORγ have been implicated in the regulation of metabolism and energy homeostasis [14,30,35,41-42,58-59,81-83,93]. RORα-deficient mice are less sensitive to developing metabolic syndrome when fed a high fat diet (HFD) as indicated by a reduced adiposity, inflammation, and hepatic steatosis, and improved insulin sensitivity [36,41]. RORα regulates Glut4 expression in skeletal muscle [14,41] and the hepatic expression of several genes involved in triglyceride synthesis, cholesterol metabolism, glucose and lipid homeostasis and inflammation [35-36,56-57,86,89,93]. ChIP and promoter analysis demonstrated that several of these genes are directly regulated by RORα. An RORα SNP was found to be associated with an increased risk for type 2 diabetes [16].

RORγ as well has a role in the regulation of glucose and lipid metabolism and insulin sensitivity [35,48,56,56,59,79,82-83,85]. Genome-wide cistromic profiling showed that in liver RORγ directly regulates the diurnal expression of a number of metabolic genes critical in the control of glycolysis and gluconeogenesis pathways, and lipid metabolism [82]. A role for RORγ in glucose homeostasis was supported by findings showing a positive correlation between the level of RORγ expression and insulin resistance [48,85].


RORs and Circadian Rhythm

Earlier studies provided evidence for a role of RORs in the regulation of circadian rhythm and clock gene expression [1,5,9,28,30,46,67,74,80-81]. RORγ exhibits a robust circadian pattern of expression that is under the direct control of the circadian clock [52,80]. RORγ regulates the diurnal expression of several glucose and lipid metabolic genes and insulin sensitivity downstream of the clock machinery [35,57,80,82-83]. Recently, RORγt was found to play also a role in the diurnal regulation of Th17 differentiation and TLR signaling [53,99].

References

Show »

1. Akashi M, Takumi T. (2005) The orphan nuclear receptor RORalpha regulates circadian transcription of the mammalian core-clock Bmal1. Nat Struct Mol Biol, 12 (5): 441-8. [PMID:15821743]

2. Amstadter AB, Sumner JA, Acierno R, Ruggiero KJ, Koenen KC, Kilpatrick DG, Galea S, Gelernter J. (2013) Support for association of RORA variant and post traumatic stress symptoms in a population-based study of hurricane exposed adults. Mol Psychiatry, 18 (11): 1148-9. [PMID:23319003]

3. Ano S, Morishima Y, Ishii Y, Yoh K, Yageta Y, Ohtsuka S, Matsuyama M, Kawaguchi M, Takahashi S, Hizawa N. (2013) Transcription factors GATA-3 and RORγt are important for determining the phenotype of allergic airway inflammation in a murine model of asthma. J Immunol, 190 (3): 1056-65. [PMID:23293351]

4. Benoit G, Cooney A, Giguere V, Ingraham H, Lazar M, Muscat G, Perlmann T, Renaud JP, Schwabe J, Sladek F, Tsai MJ, Laudet V. (2006) International Union of Pharmacology. LXVI. Orphan nuclear receptors. Pharmacol Rev, 58 (4): 798-836. [PMID:17132856]

5. Crumbley C, Wang Y, Kojetin DJ, Burris TP. (2010) Characterization of the core mammalian clock component, NPAS2, as a REV-ERBalpha/RORalpha target gene. J Biol Chem, 285 (46): 35386-92. [PMID:20817722]

6. Devanna P, Vernes SC. (2014) A direct molecular link between the autism candidate gene RORa and the schizophrenia candidate MIR137. Sci Rep, 4: 3994. [PMID:24500708]

7. Diefenbach A, Colonna M, Koyasu S. (2014) Development, differentiation, and diversity of innate lymphoid cells. Immunity, 41 (3): 354-65. [PMID:25238093]

8. Dong C. (2008) TH17 cells in development: an updated view of their molecular identity and genetic programming. Nat Rev Immunol, 8 (5): 337-48. [PMID:18408735]

9. Duez H, Staels B. (2010) Nuclear receptors linking circadian rhythms and cardiometabolic control. Arterioscler Thromb Vasc Biol, 30 (8): 1529-34. [PMID:20631353]

10. Eberl G, Marmon S, Sunshine MJ, Rennert PD, Choi Y, Littman DR. (2004) An essential function for the nuclear receptor RORgamma(t) in the generation of fetal lymphoid tissue inducer cells. Nat Immunol, 5 (1): 64-73. [PMID:14691482]

11. Ersland KM, Christoforou A, Stansberg C, Espeseth T, Mattheisen M, Mattingsdal M, Hardarson GA, Hansen T, Fernandes CP, Giddaluru S et al.. (2012) Gene-based analysis of regionally enriched cortical genes in GWAS data sets of cognitive traits and psychiatric disorders. PLoS ONE, 7 (2): e31687. [PMID:22384057]

12. Etain B, Jamain S, Milhiet V, Lajnef M, Boudebesse C, Dumaine A, Mathieu F, Gombert A, Ledudal K, Gard S et al.. (2014) Association between circadian genes, bipolar disorders and chronotypes. Chronobiol Int, 31 (7): 807-14. [PMID:24716566]

13. Fauber BP, Magnuson S. (2014) Modulators of the nuclear receptor retinoic acid receptor-related orphan receptor-γ (RORγ or RORc). J Med Chem, 57 (14): 5871-92. [PMID:24502334]

14. Fitzsimmons RL, Lau P, Muscat GE. (2012) Retinoid-related orphan receptor alpha and the regulation of lipid homeostasis. J Steroid Biochem Mol Biol, 130 (3-5): 159-68. [PMID:21723946]

15. Fu Y, Liu H, Ng L, Kim JW, Hao H, Swaroop A, Forrest D. (2014) Feedback induction of a photoreceptor-specific isoform of retinoid-related orphan nuclear receptor β by the rod transcription factor NRL. J Biol Chem, 289 (47): 32469-80. [PMID:25296752]

16. Gamboa-Meléndez MA, Huerta-Chagoya A, Moreno-Macías H, Vázquez-Cárdenas P, Ordóñez-Sánchez ML, Rodríguez-Guillén R, Riba L, Rodríguez-Torres M, Guerra-García MT, Guillén-Pineda LE et al.. (2012) Contribution of common genetic variation to the risk of type 2 diabetes in the Mexican Mestizo population. Diabetes, 61 (12): 3314-21. [PMID:22923468]

17. Germain P, Staels B, Dacquet C, Spedding M, Laudet V. (2006) Overview of nomenclature of nuclear receptors. Pharmacol Rev, 58 (4): 685-704. [PMID:17132848]

18. Halim TY, MacLaren A, Romanish MT, Gold MJ, McNagny KM, Takei F. (2012) Retinoic-acid-receptor-related orphan nuclear receptor alpha is required for natural helper cell development and allergic inflammation. Immunity, 37 (3): 463-74. [PMID:22981535]

19. Hamilton BA, Frankel WN, Kerrebrock AW, Hawkins TL, FitzHugh W, Kusumi K, Russell LB, Mueller KL, van Berkel V, Birren BW, Kruglyak L, Lander ES. (1996) Disruption of the nuclear hormone receptor RORalpha in staggerer mice. Nature, 379 (6567): 736-9. [PMID:8602221]

20. Hoorweg K, Peters CP, Cornelissen F, Aparicio-Domingo P, Papazian N, Kazemier G, Mjösberg JM, Spits H, Cupedo T. (2012) Functional Differences between Human NKp44(-) and NKp44(+) RORC(+) Innate Lymphoid Cells. Front Immunol, 3: 72. [PMID:22566953]

21. Hu X, Wang Y, Hao LY, Liu X, Lesch CA, Sanchez BM, Wendling JM, Morgan RW, Aicher TD, Carter LL et al.. (2015) Sterol metabolism controls T(H)17 differentiation by generating endogenous RORγ agonists. Nat Chem Biol, 11 (2): 141-7. [PMID:25558972]

22. Huh JR, Leung MW, Huang P, Ryan DA, Krout MR, Malapaka RR, Chow J, Manel N, Ciofani M, Kim SV et al.. (2011) Digoxin and its derivatives suppress TH17 cell differentiation by antagonizing RORγt activity. Nature, 472 (7344): 486-90. [PMID:21441909]

23. Huh JR, Littman DR. (2012) Small molecule inhibitors of RORγt: targeting Th17 cells and other applications. Eur J Immunol, 42 (9): 2232-7. [PMID:22949321]

24. Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ, Cua DJ, Littman DR. (2006) The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell, 126 (6): 1121-33. [PMID:16990136]

25. Ivanov II, Zhou L, Littman DR. (2007) Transcriptional regulation of Th17 cell differentiation. Semin Immunol, 19 (6): 409-17. [PMID:18053739]

26. Jabaudon D, Shnider SJ, Tischfield DJ, Galazo MJ, Macklis JD. (2012) RORβ induces barrel-like neuronal clusters in the developing neocortex. Cereb Cortex, 22 (5): 996-1006. [PMID:21799210]

27. Jaradat M, Stapleton C, Tilley SL, Dixon D, Erikson CJ, McCaskill JG, Kang HS, Angers M, Liao G, Collins J et al.. (2006) Modulatory role for retinoid-related orphan receptor alpha in allergen-induced lung inflammation. Am J Respir Crit Care Med, 174 (12): 1299-309. [PMID:16973978]

28. Jetten AM. (2009) Retinoid-related orphan receptors (RORs): critical roles in development, immunity, circadian rhythm, and cellular metabolism. Nucl Recept Signal, 7: e003. [PMID:19381306]

29. Jetten AM. (2011) Immunology: A helping hand against autoimmunity. Nature, 472 (7344): 421-2. [PMID:21525918]

30. Jetten AM, Kang HS, Takeda Y. (2013) Retinoic acid-related orphan receptors α and γ: key regulators of lipid/glucose metabolism, inflammation, and insulin sensitivity. Front Endocrinol (Lausanne), 4: 1. [PMID:23355833]

31. Jin L, Martynowski D, Zheng S, Wada T, Xie W, Li Y. (2010) Structural basis for hydroxycholesterols as natural ligands of orphan nuclear receptor RORgamma. Mol Endocrinol, 24 (5): 923-9. [PMID:20203100]

32. Jolly S, Journiac N, Naudet F, Gautheron V, Mariani J, Vernet-der Garabedian B. (2011) Cell-autonomous and non-cell-autonomous neuroprotective functions of RORα in neurons and astrocytes during hypoxia. J Neurosci, 31 (40): 14314-23. [PMID:21976517]

33. Kallen J, Schlaeppi JM, Bitsch F, Delhon I, Fournier B. (2004) Crystal structure of the human RORalpha Ligand binding domain in complex with cholesterol sulfate at 2.2 A. J Biol Chem, 279 (14): 14033-8. [PMID:14722075]

34. Kallen JA, Schlaeppi JM, Bitsch F, Geisse S, Geiser M, Delhon I, Fournier B. (2002) X-ray structure of the hRORalpha LBD at 1.63 A: structural and functional data that cholesterol or a cholesterol derivative is the natural ligand of RORalpha. Structure, 10 (12): 1697-707. [PMID:12467577]

35. Kang HS, Angers M, Beak JY, Wu X, Gimble JM, Wada T, Xie W, Collins JB, Grissom SF, Jetten AM. (2007) Gene expression profiling reveals a regulatory role for ROR alpha and ROR gamma in phase I and phase II metabolism. Physiol Genomics, 31 (2): 281-94. [PMID:17666523]

36. Kang HS, Okamoto K, Takeda Y, Beak JY, Gerrish K, Bortner CD, DeGraff LM, Wada T, Xie W, Jetten AM. (2011) Transcriptional profiling reveals a role for RORalpha in regulating gene expression in obesity-associated inflammation and hepatic steatosis. Physiol Genomics, 43 (13): 818-28. [PMID:21540300]

37. Kiss EA, Diefenbach A. (2012) Role of the Aryl Hydrocarbon Receptor in Controlling Maintenance and Functional Programs of RORγt(+) Innate Lymphoid Cells and Intraepithelial Lymphocytes. Front Immunol, 3: 124. [PMID:22666222]

38. Kojetin DJ, Burris TP. (2014) REV-ERB and ROR nuclear receptors as drug targets. Nat Rev Drug Discov, 13 (3): 197-216. [PMID:24577401]

39. Kumar N, Solt LA, Conkright JJ, Wang Y, Istrate MA, Busby SA, Garcia-Ordonez RD, Burris TP, Griffin PR. (2010) The benzenesulfoamide T0901317 [N-(2,2,2-trifluoroethyl)-N-[4-[2,2,2-trifluoro-1-hydroxy-1-(trifluoromethyl)ethyl]phenyl]-benzenesulfonamide] is a novel retinoic acid receptor-related orphan receptor-alpha/gamma inverse agonist. Mol Pharmacol, 77 (2): 228-36. [PMID:19887649]

40. Kurebayashi S, Ueda E, Sakaue M, Patel DD, Medvedev A, Zhang F, Jetten AM. (2000) Retinoid-related orphan receptor gamma (RORgamma) is essential for lymphoid organogenesis and controls apoptosis during thymopoiesis. Proc Natl Acad Sci USA, 97 (18): 10132-7. [PMID:10963675]

41. Lau P, Fitzsimmons RL, Pearen MA, Watt MJ, Muscat GE. (2011) Homozygous staggerer (sg/sg) mice display improved insulin sensitivity and enhanced glucose uptake in skeletal muscle. Diabetologia, 54 (5): 1169-80. [PMID:21279323]

42. Lau P, Fitzsimmons RL, Raichur S, Wang SC, Lechtken A, Muscat GE. (2008) The orphan nuclear receptor, RORalpha, regulates gene expression that controls lipid metabolism: staggerer (SG/SG) mice are resistant to diet-induced obesity. J Biol Chem, 283 (26): 18411-21. [PMID:18441015]

43. Liu H, Kim SY, Fu Y, Wu X, Ng L, Swaroop A, Forrest D. (2013) An isoform of retinoid-related orphan receptor β directs differentiation of retinal amacrine and horizontal interneurons. Nat Commun, 4: 1813. [PMID:23652001]

44. Logue MW, Baldwin C, Guffanti G, Melista E, Wolf EJ, Reardon AF, Uddin M, Wildman D, Galea S, Koenen KC et al.. (2013) A genome-wide association study of post-traumatic stress disorder identifies the retinoid-related orphan receptor alpha (RORA) gene as a significant risk locus. Mol Psychiatry, 18 (8): 937-42. [PMID:22869035]

45. Marciano DP, Chang MR, Corzo CA, Goswami D, Lam VQ, Pascal BD, Griffin PR. (2014) The therapeutic potential of nuclear receptor modulators for treatment of metabolic disorders: PPARγ, RORs, and Rev-erbs. Cell Metab, 19 (2): 193-208. [PMID:24440037]

46. Matsumura R, Matsubara C, Node K, Takumi T, Akashi M. (2013) Nuclear receptor-mediated cell-autonomous oscillatory expression of the circadian transcription factor, neuronal PAS domain protein 2 (NPAS2). J Biol Chem, 288 (51): 36548-53. [PMID:24196956]

47. McKenzie AN, Spits H, Eberl G. (2014) Innate lymphoid cells in inflammation and immunity. Immunity, 41 (3): 366-74. [PMID:25238094]

48. Meissburger B, Ukropec J, Roeder E, Beaton N, Geiger M, Teupser D, Civan B, Langhans W, Nawroth PP, Gasperikova D et al.. (2011) Adipogenesis and insulin sensitivity in obesity are regulated by retinoid-related orphan receptor gamma. EMBO Mol Med, 3 (11): 637-51. [PMID:21853531]

49. Melén E, Kho AT, Sharma S, Gaedigk R, Leeder JS, Mariani TJ, Carey VJ, Weiss ST, Tantisira KG. (2011) Expression analysis of asthma candidate genes during human and murine lung development. Respir Res, 12: 86. [PMID:21699702]

50. Miller MW, Wolf EJ, Logue MW, Baldwin CT. (2013) The retinoid-related orphan receptor alpha (RORA) gene and fear-related psychopathology. J Affect Disord, 151 (2): 702-8. [PMID:24007783]

51. Moffatt MF, Gut IG, Demenais F, Strachan DP, Bouzigon E, Heath S, von Mutius E, Farrall M, Lathrop M, Cookson WO et al.. (2010) A large-scale, consortium-based genomewide association study of asthma. N Engl J Med, 363 (13): 1211-21. [PMID:20860503]

52. Mongrain V, Ruan X, Dardente H, Fortier EE, Cermakian N. (2008) Clock-dependent and independent transcriptional control of the two isoforms from the mouse Rorgamma gene. Genes Cells, 13 (12): 1197-210. [PMID:19076641]

53. Mukherji A, Kobiita A, Ye T, Chambon P. (2013) Homeostasis in intestinal epithelium is orchestrated by the circadian clock and microbiota cues transduced by TLRs. Cell, 153 (4): 812-27. [PMID:23663780]

54. Nguyen A, Rauch TA, Pfeifer GP, Hu VW. (2010) Global methylation profiling of lymphoblastoid cell lines reveals epigenetic contributions to autism spectrum disorders and a novel autism candidate gene, RORA, whose protein product is reduced in autistic brain. FASEB J, 24 (8): 3036-51. [PMID:20375269]

55. Nurieva R, Yang XO, Martinez G, Zhang Y, Panopoulos AD, Ma L, Schluns K, Tian Q, Watowich SS, Jetten AM et al.. (2007) Essential autocrine regulation by IL-21 in the generation of inflammatory T cells. Nature, 448 (7152): 480-3. [PMID:17581589]

56. Ou Z, Shi X, Gilroy RK, Kirisci L, Romkes M, Lynch C, Wang H, Xu M, Jiang M, Ren S et al.. (2013) Regulation of the human hydroxysteroid sulfotransferase (SULT2A1) by RORα and RORγ and its potential relevance to human liver diseases. Mol Endocrinol, 27 (1): 106-15. [PMID:23211525]

57. Pathak P, Li T, Chiang JY. (2013) Retinoic acid-related orphan receptor α regulates diurnal rhythm and fasting induction of sterol 12α-hydroxylase in bile acid synthesis. J Biol Chem, 288 (52): 37154-65. [PMID:24226095]

58. Raichur S, Fitzsimmons RL, Myers SA, Pearen MA, Lau P, Eriksson N, Wang SM, Muscat GE. (2010) Identification and validation of the pathways and functions regulated by the orphan nuclear receptor, ROR alpha1, in skeletal muscle. Nucleic Acids Res, 38 (13): 4296-312. [PMID:20338882]

59. Raichur S, Lau P, Staels B, Muscat GE. (2007) Retinoid-related orphan receptor gamma regulates several genes that control metabolism in skeletal muscle cells: links to modulation of reactive oxygen species production. J Mol Endocrinol, 39 (1): 29-44. [PMID:17601883]

60. Ramasamy A, Kuokkanen M, Vedantam S, Gajdos ZK, Couto Alves A, Lyon HN, Ferreira MA, Strachan DP, Zhao JH, Abramson MJ et al.. (2012) Genome-wide association studies of asthma in population-based cohorts confirm known and suggested loci and identify an additional association near HLA. PLoS ONE, 7 (9): e44008. [PMID:23028483]

61. Salimi M, Barlow JL, Saunders SP, Xue L, Gutowska-Owsiak D, Wang X, Huang LC, Johnson D, Scanlon ST, McKenzie AN et al.. (2013) A role for IL-25 and IL-33-driven type-2 innate lymphoid cells in atopic dermatitis. J Exp Med, 210 (13): 2939-50. [PMID:24323357]

62. Santarlasci V, Maggi L, Capone M, Querci V, Beltrame L, Cavalieri D, D'Aiuto E, Cimaz R, Nebbioso A, Liotta F et al.. (2012) Rarity of human T helper 17 cells is due to retinoic acid orphan receptor-dependent mechanisms that limit their expansion. Immunity, 36 (2): 201-14. [PMID:22326581]

63. Santori FR, Huang P, van de Pavert SA, Douglass Jr EF, Leaver DJ, Haubrich BA, Keber R, Lorbek G, Konijn T, Rosales BN et al.. (2015) Identification of natural RORγ ligands that regulate the development of lymphoid cells. Cell Metab, 21 (2): 286-97. [PMID:25651181]

64. Sarachana T, Hu VW. (2013) Differential recruitment of coregulators to the RORA promoter adds another layer of complexity to gene (dys) regulation by sex hormones in autism. Mol Autism, 4 (1): 39. [PMID:24119295]

65. Sarachana T, Hu VW. (2013) Genome-wide identification of transcriptional targets of RORA reveals direct regulation of multiple genes associated with autism spectrum disorder. Mol Autism, 4 (1): 14. [PMID:23697635]

66. Sarachana T, Xu M, Wu RC, Hu VW. (2011) Sex hormones in autism: androgens and estrogens differentially and reciprocally regulate RORA, a novel candidate gene for autism. PLoS ONE, 6 (2): e17116. [PMID:21359227]

67. Sato TK, Panda S, Miraglia LJ, Reyes TM, Rudic RD, McNamara P, Naik KA, FitzGerald GA, Kay SA, Hogenesch JB. (2004) A functional genomics strategy reveals Rora as a component of the mammalian circadian clock. Neuron, 43 (4): 527-37. [PMID:15312651]

68. Schaeren-Wiemers N, André E, Kapfhammer JP, Becker-André M. (1997) The expression pattern of the orphan nuclear receptor RORbeta 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): 2687-701. [PMID:9517474]

69. Serafini N, Klein Wolterink RG, Satoh-Takayama N, Xu W, Vosshenrich CA, Hendriks RW, Di Santo JP. (2014) Gata3 drives development of RORγt+ group 3 innate lymphoid cells. J Exp Med, 211 (2): 199-208. [PMID:24419270]

70. Skepner J, Ramesh R, Trocha M, Schmidt D, Baloglu E, Lobera M, Carlson T, Hill J, Orband-Miller LA, Barnes A et al.. (2014) Pharmacologic inhibition of RORγt regulates Th17 signature gene expression and suppresses cutaneous inflammation in vivo. J Immunol, 192 (6): 2564-75. [PMID:24516202]

71. Slominski AT, Kim TK, Takeda Y, Janjetovic Z, Brozyna AA, Skobowiat C, Wang J, Postlethwaite A, Li W, Tuckey RC et al.. (2014) RORα and ROR γ are expressed in human skin and serve as receptors for endogenously produced noncalcemic 20-hydroxy- and 20,23-dihydroxyvitamin D. FASEB J, 28 (7): 2775-89. [PMID:24668754]

72. Solt LA, Banerjee S, Campbell S, Kamenecka TM, Burris TP. (2015) ROR inverse agonist suppresses insulitis and prevents hyperglycemia in a mouse model of type 1 diabetes. Endocrinology, 156 (3): 869-81. [PMID:25560829]

73. Solt LA, Burris TP. (2012) Action of RORs and their ligands in (patho)physiology. Trends Endocrinol Metab, 23 (12): 619-27. [PMID:22789990]

74. Solt LA, Kojetin DJ, Burris TP. (2011) The REV-ERBs and RORs: molecular links between circadian rhythms and lipid homeostasis. Future Med Chem, 3 (5): 623-38. [PMID:21526899]

75. Solt LA, Kumar N, Nuhant P, Wang Y, Lauer JL, Liu J, Istrate MA, Kamenecka TM, Roush WR, Vidović D et al.. (2011) Suppression of TH17 differentiation and autoimmunity by a synthetic ROR ligand. Nature, 472 (7344): 491-4. [PMID:21499262]

76. Stehlin-Gaon C, Willmann D, Zeyer D, Sanglier S, Van Dorsselaer A, Renaud JP, Moras D, Schüle R. (2003) All-trans retinoic acid is a ligand for the orphan nuclear receptor ROR beta. Nat Struct Biol, 10 (10): 820-5. [PMID:12958591]

77. Steinmayr M, André E, Conquet F, Rondi-Reig L, Delhaye-Bouchaud N, Auclair N, Daniel H, Crépel F, Mariani J, Sotelo C, Becker-André M. (1998) staggerer phenotype in retinoid-related orphan receptor alpha-deficient mice. Proc Natl Acad Sci USA, 95 (7): 3960-5. [PMID:9520475]

78. Sun Z, Unutmaz D, Zou YR, Sunshine MJ, Pierani A, Brenner-Morton S, Mebius RE, Littman DR. (2000) Requirement for RORgamma in thymocyte survival and lymphoid organ development. Science, 288 (5475): 2369-73. [PMID:10875923]

79. Takeda Y, Jetten AM. (2013) Prospero-related homeobox 1 (Prox1) functions as a novel modulator of retinoic acid-related orphan receptors α- and γ-mediated transactivation. Nucleic Acids Res, 41 (14): 6992-7008. [PMID:23723244]

80. Takeda Y, Jothi R, Birault V, Jetten AM. (2012) RORγ directly regulates the circadian expression of clock genes and downstream targets in vivo. Nucleic Acids Res, 40 (17): 8519-35. [PMID:22753030]

81. Takeda Y, Kang HS, Angers M, Jetten AM. (2011) Retinoic acid-related orphan receptor γ directly regulates neuronal PAS domain protein 2 transcription in vivo. Nucleic Acids Res, 39 (11): 4769-82. [PMID:21317191]

82. Takeda Y, Kang HS, Freudenberg J, DeGraff LM, Jothi R, Jetten AM. (2014) Retinoic acid-related orphan receptor γ (RORγ): a novel participant in the diurnal regulation of hepatic gluconeogenesis and insulin sensitivity. PLoS Genet, 10 (5): e1004331. [PMID:24831725]

83. Takeda Y, Kang HS, Lih FB, Jiang H, Blaner WS, Jetten AM. (2014) Retinoid acid-related orphan receptor γ, RORγ, participates in diurnal transcriptional regulation of lipid metabolic genes. Nucleic Acids Res, 42 (16): 10448-59. [PMID:25143535]

84. Tilley SL, Jaradat M, Stapleton C, Dixon D, Hua X, Erikson CJ, McCaskill JG, Chason KD, Liao G, Jania L et al.. (2007) Retinoid-related orphan receptor gamma controls immunoglobulin production and Th1/Th2 cytokine balance in the adaptive immune response to allergen. J Immunol, 178 (5): 3208-18. [PMID:17312169]

85. Tinahones FJ, Moreno-Santos I, Vendrell J, Chacon MR, Garrido-Sanchez L, García-Fuentes E, Macias-González M. (2012) The retinoic acid receptor-related orphan nuclear receptor γ1 (RORγ1): a novel player determinant of insulin sensitivity in morbid obesity. Obesity (Silver Spring), 20 (3): 488-97. [PMID:21904299]

86. Tuong ZK, Lau P, Yeo JC, Pearen MA, Wall AA, Stanley AC, Stow JL, Muscat GE. (2013) Disruption of Rorα1 and cholesterol 25-hydroxylase expression attenuates phagocytosis in male Rorαsg/sg mice. Endocrinology, 154 (1): 140-9. [PMID:23239817]

87. van de Pavert SA, Ferreira M, Domingues RG, Ribeiro H, Molenaar R, Moreira-Santos L, Almeida FF, Ibiza S, Barbosa I, Goverse G et al.. (2014) Maternal retinoids control type 3 innate lymphoid cells and set the offspring immunity. Nature, 508 (7494): 123-7. [PMID:24670648]

88. van Niel MB, Fauber BP, Cartwright M, Gaines S, Killen JC, René O, Ward SI, de Leon Boenig G, Deng Y, Eidenschenk C et al.. (2014) A reversed sulfonamide series of selective RORc inverse agonists. Bioorg Med Chem Lett, 24 (24): 5769-76. [PMID:25453817]

89. Wada T, Kang HS, Angers M, Gong H, Bhatia S, Khadem S, Ren S, Ellis E, Strom SC, Jetten AM et al.. (2008) Identification of oxysterol 7alpha-hydroxylase (Cyp7b1) as a novel retinoid-related orphan receptor alpha (RORalpha) (NR1F1) target gene and a functional cross-talk between RORalpha and liver X receptor (NR1H3). Mol Pharmacol, 73 (3): 891-9. [PMID:18055760]

90. Walker JA, Barlow JL, McKenzie AN. (2013) Innate lymphoid cells--how did we miss them?. Nat Rev Immunol, 13 (2): 75-87. [PMID:23292121]

91. Wang Y, Kumar N, Crumbley C, Griffin PR, Burris TP. (2010) A second class of nuclear receptors for oxysterols: Regulation of RORalpha and RORgamma activity by 24S-hydroxycholesterol (cerebrosterol). Biochim Biophys Acta, 1801 (8): 917-23. [PMID:20211758]

92. Wang Y, Kumar N, Solt LA, Richardson TI, Helvering LM, Crumbley C, Garcia-Ordonez RD, Stayrook KR, Zhang X, Novick S et al.. (2010) Modulation of retinoic acid receptor-related orphan receptor alpha and gamma activity by 7-oxygenated sterol ligands. J Biol Chem, 285 (7): 5013-25. [PMID:19965867]

93. Wang Y, Solt LA, Burris TP. (2010) Regulation of FGF21 expression and secretion by retinoic acid receptor-related orphan receptor alpha. J Biol Chem, 285 (21): 15668-73. [PMID:20332535]

94. Wong SH, Walker JA, Jolin HE, Drynan LF, Hams E, Camelo A, Barlow JL, Neill DR, Panova V, Koch U et al.. (2012) Transcription factor RORα is critical for nuocyte development. Nat Immunol, 13 (3): 229-36. [PMID:22267218]

95. Xiao S, Yosef N, Yang J, Wang Y, Zhou L, Zhu C, Wu C, Baloglu E, Schmidt D, Ramesh R et al.. (2014) Small-molecule RORγt antagonists inhibit T helper 17 cell transcriptional network by divergent mechanisms. Immunity, 40 (4): 477-89. [PMID:24745332]

96. Xu T, Wang X, Zhong B, Nurieva RI, Ding S, Dong C. (2011) Ursolic acid suppresses interleukin-17 (IL-17) production by selectively antagonizing the function of RORgamma t protein. J Biol Chem, 286 (26): 22707-10. [PMID:21566134]

97. Yang XO, Pappu BP, Nurieva R, Akimzhanov A, Kang HS, Chung Y, Ma L, Shah B, Panopoulos AD, Schluns KS et al.. (2008) T helper 17 lineage differentiation is programmed by orphan nuclear receptors ROR alpha and ROR gamma. Immunity, 28 (1): 29-39. [PMID:18164222]

98. Yosef N, Shalek AK, Gaublomme JT, Jin H, Lee Y, Awasthi A, Wu C, Karwacz K, Xiao S, Jorgolli M et al.. (2013) Dynamic regulatory network controlling TH17 cell differentiation. Nature, 496 (7446): 461-8. [PMID:23467089]

99. Yu X, Rollins D, Ruhn KA, Stubblefield JJ, Green CB, Kashiwada M, Rothman PB, Takahashi JS, Hooper LV. (2013) TH17 cell differentiation is regulated by the circadian clock. Science, 342 (6159): 727-30. [PMID:24202171]

100. Zhang F, Meng G, Strober W. (2008) Interactions among the transcription factors Runx1, RORgammat and Foxp3 regulate the differentiation of interleukin 17-producing T cells. Nat Immunol, 9 (11): 1297-306. [PMID:18849990]

How to cite this page

To cite this family introduction, please use the following:

Database page citation (select format):