Abstract

Overview: Nuclear receptors are specialised transcription factors with commonalities of sequence and structure, which bind as homo- or heterodimers to specific consensus sequences of DNA (response elements) in the promoter region of particular target genes. They regulate (either promoting or repressing) transcription of these target genes in response to a variety of endogenous ligands. Endogenous agonists are hydrophobic entities which, when bound to the receptor promote conformational changes in the receptor to allow recruitment (or dissociation) of protein partners, generating a large multiprotein complex. Two major subclasses of nuclear receptors with identified endogenous agonists can be identified: steroid and non-steroid hormone receptors. Steroid hormone receptors function typically as dimeric entities and are thought to be resident outside the nucleus in the unliganded state in a complex with chaperone proteins, which are liberated upon agonist binding. Migration to the nucleus and interaction with other regulators of gene transcription, including RNA polymerase, acetyltransferases and deacetylases, allows gene transcription to be regulated. Non-steroid hormone receptors typically exhibit a greater distribution in the nucleus in the unliganded state and interact with other nuclear receptors to form heterodimers, as well as with other regulators of gene transcription, leading to changes in gene transcription upon agonist binding. Selectivity of gene regulation is brought about through interaction of nuclear receptors with particular consensus sequences of DNA, which are arranged typically as repeats or inverted palindromes to allow accumulation of multiple transcription factors in the promoter regions of genes. Germain P, Staels B, Dacquet C, Spedding M, Laudet V (2006). Overview of nomenclature of nuclear receptors. Pharmacol Rev58: 685–704. In man, 48 nuclear receptors have been identified from sequence analysis of the genome (see Benoit et al., 2006; Germain et al., 2006), only half of which have been ‘assigned’ a ligand by Nomenclature Committees of NC-IUPHAR. 19 families of nuclear receptors have been identified, allowing a systematic nomenclature of the format NRXYZ, where NR represents nuclear receptor, X the subfamily (1, 2, 3, 4, 5, 6 or 0), Y the group (A, B, C, D, F, H or I) and Z the individual member. Listed below are a number of putative nuclear receptors identified by NC-IUPHAR, for which only preliminary evidence for an endogenous ligand has been published. Abbreviations: AC45594, 4-heptoxyphenol; DY131, N-(4-(diethylaminobenzylidenyl)-N′-(4-hydroxybenzoyl)-hydrazine; GSK4112, 1,1-dimethylethyl-N-[(4-chlorophenyl)methyl]-N-[(5-nitro-2-thienyl)methyl])glycinate, also known as SR6452; GSK4716, 4-hydroxy-2-[(1E)-[4-(1-methylethyl)phenyl]methylene]hydrazide; SID7969543, ethyl 2-[2-[2-(2,3-dihydro-1,4-benzodioxin-7-ylamino)-2-oxoethyl]-1-oxoisoquinolin-5-yl]oxypropanoate; XCT790, (E)-3-[4-[[2,4-bis(trifluoromethyl)phenyl]methoxy]-3-methoxyphenyl]-2-cyano-N-[5-(trifluoromethyl)-1,3,4-thiadiazol-2-yl]prop-2-enamide Benoit G, Cooney A, Giguere V, Ingraham H, Lazar M, Muscat G et al. (2006). International Union of Pharmacology. LXVI. Orphan nuclear receptors. Pharmacol Rev58: 798–836. Chanda D, Park JH, Choi HS (2008). Molecular basis of endocrine regulation by orphan nuclear receptor Small Heterodimer Partner. Endocr J55: 253–268. Deblois G, Giguere V (2011). Functional and physiological genomics of estrogen-related receptors (ERRs) in health and disease. Biochim Biophys Acta1812: 1032–1040. Duez H, Staels B (2009). Rev-erb-α: an integrator of circadian rhythms and metabolism. J Appl Physiol107: 1972–1980. Duez H, Staels B (2010). Nuclear receptors linking circadian rhythms and cardiometabolic control. Arterioscler Thromb Vasc Biol30: 1529–1534. El-Khairi R, Martinez-Aguayo A, Ferraz-de-Souza B, Lin L, Achermann JC (2011). Role of DAX-1 (NR0B1) and steroidogenic factor-1 (NR5A1) in human adrenal function. Endocr Dev20: 38–46. Fernandez-Marcos PJ, Auwerx J, Schoonjans K (2011). Emerging actions of the nuclear receptor LRH-1 in the gut. Biochim Biophys Acta (BBA) Mol Basis Dis1812: 947–955. Germain P, Staels B, Dacquet C, Spedding M, Laudet V (2006). Overview of nomenclature of nuclear receptors. Pharmacol Rev58: 685–704. Gui H, Li ML, Tsai CC (2011). A tale of tailless. Dev Neurosci33: 1–13. Huang P, Chandra V, Rastinejad F (2010). Structural overview of the nuclear receptor superfamily: insights into physiology and therapeutics. Annu Rev Physiol72: 247–272. Hwang-Verslues WW, Sladek FM (2010). HNF4α– role in drug metabolism and potential drug target? Curr Opin Pharmacol10: 698–705. Lin FJ, Qin J, Tang K, Tsai SY, Tsai MJ (2011). Coup d'Etat: an Orphan Takes Control. Endocr Rev32: 404–421. Mlynarczuk J, Rekawiecki R (2010). The role of the orphan receptor SF-1 in the development and function of the ovary. Reprod Biol10: 177–193. Pearen MA, Muscat GE (2010). Nuclear hormone receptor 4A signaling: implications for metabolic disease. Mol Endocrinol24: 1891–1903. Phelan CA, Gampe RT, Jr., Lambert MH, Parks DJ, Montana V, Bynum J et al. (2010). Structure of Rev-erbα bound to N-CoR reveals a unique mechanism of nuclear receptor-co-repressor interaction. Nat Struct Mol Biol17: 808–814. Schimmer BP, White PC (2010). Steroidogenic factor 1: its roles in differentiation, development, and disease. Mol Endocrinol24: 1322–1337. Sladek FM (2011). What are nuclear receptor ligands? Mol Cell Endocrinol334: 3–13. Yin L, Wu N, Lazar MA (2010). Nuclear receptor Rev-erbα: a heme receptor that coordinates circadian rhythm and metabolism. Nucl Recept Signal8: e001. Zhang Y, Hagedorn CH, Wang L (2011). Role of nuclear receptor SHP in metabolism and cancer. Biochim Biophys Acta (BBA) Mol Basis Dis1812: 893–908. Zhao Y, Bruemmer D (2010). NR4A orphan nuclear receptors: transcriptional regulators of gene expression in metabolism and vascular biology. Arterioscler Thromb Vasc Biol30: 1535–1541. Overview: Liver X and farnesoid X receptors (LXR and FXR, nomenclature as agreed by NC-IUPHAR Committee on Nuclear Receptors, see Moore et al., 2006) are members of a steroid analogue-activated nuclear receptor subfamily (ENSF00000000720), which form heterodimers with members of the retinoid X receptor family. Endogenous ligands for LXRs include hydroxycholesterols (OHC), while FXRs appear to be activated by bile acids. TO901317 (Repa et al., 2000) and GW3965 (Collins et al., 2002) are synthetic agonists acting at both LXRα and LXRβ with less than 10-fold selectivity. Abbreviations: ECDCA, 6α-ethyl-chenodeoxycholate, also known as INT747; guggulsterone, trans-4,17(20)-pregnadiene-3,16-dione; GW3965, 3-(3-[N-{2-chloro-3-trifluoromethylbenzyl}-{2,2-diphenylethyl}amino]propyloxy)phenylacetic acid hydrochloride; GW4064, 3-[(E)-2-[2-chloro-4-[[3-(2,6-dichlorophenyl)-5-propan-2-yl-1,2-oxazol-4-yl]methoxy]phenyl]ethenyl]benzoic acid; OHC, hydroxycholesterol; TO901317, N-(2,2,2-trifluoroethyl)-N-(4-[2,2,2-trifluoro-1-hydroxy-1-{trifluoromethyl}ethyl]phenyl)-benzenesulfonamide Beltowski J, Semczuk A (2010). Liver X receptor (LXR) and the reproductive system – a potential novel target for therapeutic intervention. Pharmacol Rep62: 15–27. Calkin AC, Tontonoz P (2010). Liver X receptor signaling pathways and atherosclerosis. Arterioscler Thromb Vasc Biol30: 1513–1518. Cariou B, Staels B (2007). FXR: a promising target for the metabolic syndrome? Trends Pharmacol Sci28: 236–243. Chen WD, Wang YD, Meng Z, Zhang L, Huang W (2011). Nuclear bile acid receptor FXR in the hepatic regeneration. Biochim Biophys Acta (BBA) Mol Basis Dis1812: 888–892. Claudel T, Zollner G, Wagner M, Trauner M (2011). Role of nuclear receptors for bile acid metabolism, bile secretion, cholestasis, and gallstone disease. Biochim Biophys Acta (BBA) Mol Basis Dis1812: 867–878. El-Hajjaji FZ, Oumeddour A, Pommier AJC, Ouvrier A, Viennois E, Dufour J et al. (2011). Liver X receptors, lipids and their reproductive secrets in the male. Biochim Biophys Acta (BBA) Mol Basis Dis1812: 974–981. Faulds MH, Zhao C, Dahlman-Wright K (2010). Molecular biology and functional genomics of liver X receptors (LXR) in relationship to metabolic diseases. Curr Opin Pharmacol10: 692–697. Fievet C, Staels B (2009). Liver X receptor modulators: effects on lipid metabolism and potential use in the treatment of atherosclerosis. Biochem Pharmacol77: 1316–1327. Fiorucci S, Mencarelli A, Palladino G, Cipriani S (2009). Bile-acid-activated receptors: targeting TGR5 and farnesoid-X-receptor in lipid and glucose disorders. Trends Pharmacol Sci30: 570–580. Fiorucci S, Cipriani S, Baldelli F, Mencarelli A (2010). Bile acid-activated receptors in the treatment of dyslipidemia and related disorders. Prog Lipid Res49: 171–185. Gabbi C, Warner M, Gustafsson JA (2009). Liver X receptor beta: emerging roles in physiology and diseases. Mol Endocrinol23: 129–136. Gadaleta RM, van Mil SW, Oldenburg B, Siersema PD, Klomp LW, van Erpecum KJ (2010). Bile acids and their nuclear receptor FXR: relevance for hepatobiliary and gastrointestinal disease. Biochim Biophys Acta1801: 683–692. Gardmo C, Tamburro A, Modica S, Moschetta A (2011). Proteomics for the discovery of nuclear bile acid receptor FXR targets. Biochim Biophys Acta (BBA) Mol Basis Dis1812: 836–841. González N, Castrillo A (2011). Liver X receptors as regulators of macrophage inflammatory and metabolic pathways. Biochim Biophys Acta (BBA) Mol Basis Dis1812: 982–994. Hageman J, Herrema H, Groen AK, Kuipers F (2010). A role of the bile salt receptor FXR in atherosclerosis. Arterioscler Thromb Vasc Biol30: 1519–1528. Kemper JK (2011). Regulation of FXR transcriptional activity in health and disease: emerging roles of FXR cofactors and post-translational modifications. Biochim Biophys Acta (BBA) Mol Basis Dis1812: 842–850. Kuipers F, Stroeve JH, Caron S, Staels B (2007). Bile acids, farnesoid X receptor, atherosclerosis and metabolic control. Curr Opin Lipidol18: 289–297. Lefebvre P, Cariou B, Lien F, Kuipers F, Staels B (2009). Role of bile acids and bile acid receptors in metabolic regulation. Physiol Rev89: 147–191. Mencarelli A, Fiorucci S (2010). FXR an emerging therapeutic target for the treatment of atherosclerosis. J Cell Mol Med14: 79–92. Moore DD, Kato S, Xie W, Mangelsdorf DJ, Schmidt DR, Xiao R et al. (2006). International Union of Pharmacology. LXII. The NR1H and NR1I receptors: constitutive androstane receptor, pregnene X receptor, farnesoid X receptor α, farnesoid X receptor β, liver X receptor α, liver X receptor β, and vitamin D receptor. Pharmacol Rev58: 742–759. Schmuth M, Jiang YJ, Dubrac S, Elias PM, Feingold KR (2008). Thematic review series: skin lipids. Peroxisome proliferator-activated receptors and liver X receptors in epidermal biology. J Lipid Res49: 499–509. Wang YD, Chen WD, Moore DD, Huang W (2008). FXR: a metabolic regulator and cell protector. Cell Res18: 1087–1095. Zhang Y, Edwards PA (2008). FXR signaling in metabolic disease. FEBS Lett582: 10–18. Zollner G, Trauner M (2009). Nuclear receptors as therapeutic targets in cholestatic liver diseases. Br J Pharmacol156: 7–27. Overview: Peroxisome proliferator-activated receptors (PPARs, nomenclature as agreed by NC-IUPHAR Committee on Nuclear Receptors, see Michalik et al., 2006) are nuclear hormone receptors of the NR1C family, with diverse roles regulating lipid homeostasis, cellular differentiation, proliferation and the immune response. PPARs have many potential endogenous agonists (see Michalik et al., 2006), including 15-deoxy-Δ12,14-prostaglandin J2, prostacyclin, many fatty acids and their oxidation products, lysophosphatidic acid (McIntyre et al., 2003), 13-HODE, 15-HETE, Paz-PC, azelaoyl-PAF, and leukotriene B4. These receptors also bind hypolipidaemic drugs (PPARα) and anti-diabetic thiazolidinediones (PPARγ), as well as many non-steroidal anti-inflammatory drugs, such as sulindac and indomethacin. Once activated by a ligand, the receptor forms a heterodimer with members of the retinoid X receptor family and can act as a transcription factor. Although radioligand binding assays have been described for all three receptors, the radioligands are not commercially available. Commonly, receptor occupancy studies are conducted using fluorescent ligands and truncated forms of the receptor limited to the ligand binding domain. As with the estrogen receptor antagonists, many agents show tissue-selective efficacy (e.g. Bishop-Bailey, 2000; Rocchi et al., 2001; Nakamuta et al., 2002). Agonists with mixed activity at PPARα and PPARγ have also been described (e.g. Doebber et al., 2004; Guo et al., 2004; Xu et al., 2004). Abbreviations: 13-HODE, 13-hydroxyoctadecadienoic acid; 15-HETE, 15-hydroxyeicosatetraenoic acid; azelaoyl-PAF, 1-O-hexadecyl-2-O-(9-carboxyoctanoyl)-sn-glyceryl-3-phosphocholine; BADGE, bisphenol A diglycidyl ether; CDDO, 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid; CDDO-Me, 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid methyl ester; GSK0660, ; GW1929, (2S)-([2-benzoylphenyl]amino)-3-(4-[2-{methylpyridin-2-ylamino}ethoxy]phenyl)propionic acid; GW501516, 2-methyl-4([{(4-methyl-2-[4-trifluoromethylphenyl]-1,3-thiazol-5-yl)methyl}sulfanyl]phenoxy)acetic acid, GW7647, 2-([4-{2-([{cyclohexylamino}carbonyl][4-cyclohexylbutyl]amino)ethyl}phenyl]thio)-2-methylpropanoic acid; GW9662, 2-chloro-5-nitro-N-phenylbenzamide; L165041, (4-[3-{4-acetyl-3-hydroxy-2-propylphenoxy}propoxyl]phenoxy)acetic acid; Paz-PC, 1-palmitoyl-2-azelaoyl-sn-glycero-3-phosphocholine; T0070907, 2-chloro-5-nitro-N-(4-pyridyl)benzamide; WY14643, N-(3-[2-quinolinylmethoxy]phenyl)-trifluoromethanesulphonamide Belvisi MG, Mitchell JA (2009). Targeting PPAR receptors in the airway for the treatment of inflammatory lung disease. Br J Pharmacol158: 994–1003. Chaturvedi RK, Beal MF (2008). PPAR: a therapeutic target in Parkinson's disease. J Neurochem106: 506–518. Christodoulides C, Vidal-Puig A (2010). PPARs and adipocyte function. Mol Cell Endocrinol318: 61–68. Ehrenborg E, Krook A (2009). Regulation of skeletal muscle physiology and metabolism by peroxisome proliferator-activated receptor delta. Pharmacol Rev61: 373–393. Hiukka A, Maranghi M, Matikainen N, Taskinen MR (2010). PPARα: an emerging therapeutic target in diabetic microvascular damage. Nat Rev Endocrinol6: 454–463. Itoh T, Yamamoto K (2008). Peroxisome proliferator activated receptor gamma and oxidized docosahexaenoic acids as new class of ligand. Naunyn Schmiedebergs Arch Pharmacol377: 541–547. Kawai M, Rosen CJ (2010). PPARγ: a circadian transcription factor in adipogenesis and osteogenesis. Nat Rev Endocrinol6: 629–636. Kawai M, Sousa KM, MacDougald OA, Rosen CJ (2010). The many facets of PPARγ: novel insights for the skeleton. Am J Physiol Endocrinol Metab299: E3–E9. Lockyer P, Schisler JC, Patterson C, Willis MS (2010). Won't get fooled again: the nonmetabolic roles of peroxisome proliferator-activated receptors (PPARs) in the heart. Mol Endocrinol24: 1111–1119. Michalik L, Wahli W (2008). PPARs mediate lipid signaling in inflammation and cancer. PPAR Res, 2008: 134059. Michalik L, Auwerx J, Berger JP, Chatterjee VK, Glass CK, Gonzalez FJ et al. (2006). International Union of Pharmacology. LXI. Peroxisome proliferator-activated receptors. Pharmacol Rev58: 726–741. Reilly SM, Lee CH (2008). PPAR δ as a therapeutic target in metabolic disease. FEBS Lett582: 26–31. Robinson E, Grieve DJ (2009). Significance of peroxisome proliferator-activated receptors in the cardiovascular system in health and disease. Pharmacol Ther, 122: 246–263. Varga T, Czimmerer Z, Nagy L (2011). PPARs are a unique set of fatty acid regulated transcription factors controlling both lipid metabolism and inflammation. Biochim Biophys Acta (BBA) Mol Basis Dis1812: 1007–1022. Villacorta L, Schopfer FJ, Zhang J, Freeman BA, Chen YE (2009). PPARγ and its ligands: therapeutic implications in cardiovascular disease. Clin Sci, 116: 205–218. Wan Y (2010). PPARγ in bone homeostasis. Trends Endocrinol Metab21: 722–728. Wang N, Yin R, Liu Y, Mao G, Xi F (2011). Role of Peroxisome Proliferator-Activated Receptor-gamma in Atherosclerosis. Circ J75: 528–535. Youssef J, Badr M (2011). Peroxisome Proliferator-Activated Receptors and Cancer Challenges and Opportunities. Br J Pharmacol164: 68–82. Yu S, Reddy JK (2007). Transcription coactivators for peroxisome proliferator-activated receptors. Biochim Biophys Acta1771: 936–951. Ziouzenkova O, Plutzky J (2008). Retinoid metabolism and nuclear receptor responses: new insights into coordinated regulation of the PPAR-RXR complex. FEBS Lett582: 32–38. Overview: Cytoplasmic cellular retinoid binding proteins I (ENSG00000114115), II (ENSG00000114113), III (ENSG00000139194) and IV (ENSG00000162444) are thought to control the levels of intracellular retinoids available for interaction with their receptors (Li, 1999). [3H]-ATRA and [3H]-9-cis-retinoic acid have been used to label RARs and RXRs, respectively. Retinoic acid receptors (nomenclature as agreed by NC-IUPHAR Committee on Nuclear Receptors, see Germain et al., 2006a) are nuclear hormone receptors of the NR1B family activated by the vitamin A-derived agonists all-trans retinoic acid (ATRA) and 9-cis-retinoic acid, and the RAR-selective synthetic agonists TTNPB and adapalene. Ro415253 has been suggested to be a PPARγ agonist (Schupp et al., 2007). LE135 is an antagonist with selectivity for RARα and RARβ compared to RARγ (Li et al., 1999). Retinoid X receptors (nomenclature as agreed by NC-IUPHAR Committee on Nuclear Receptors, see Germain et al., 2006b) are NR2B family members activated by 9-cis-retinoic acid and the RXR-selective agonists LGD1069 and LG100268, sometimes referred to as rexinoids. These receptors form RXR–RAR heterodimers and RXR–RXR homodimers (Mangelsdorf and Evans, 1995; Chambon, 1996). UVI3003 has been described as a pan-RXR antagonist (Nahoum et al., 2007). Retinoic acid-related orphan receptors (ROR, nomenclature as agreed by NC-IUPHAR Committee on Nuclear Receptors, see Benoit et al., 2006) have yet to be assigned a definitive endogenous ligand, although RORα may be synthesised with a ‘captured’ agonist such as cholesterol (Kallen et al., 2002; 2004). ATRA shows selectivity for RORβ within the ROR family (Stehlin-Gaon et al., 2003). RORα has been suggested to be a nuclear receptor responding to melatonin (Wiesenberg et al., 1995). Abbreviations: 7-OHC, 7-hydroxycholesterol; AC261066, 4-(4-[2-butoxyethoxy]-5-methyl-2-thiazolyl)-2-fluorobenzoic acid; AC55649, 4′-octyl-[1,1′-biphenyl]-4-carboxylic acid; AHPN, 6-[3-(1-adamantyl)-4-hydroxyphenyl]-2-naphthalene carboxylic acid, also known as CD437; ATRA, all-trans-retinoic acid; CD1530, 4-(6-hydroxy-7-tricyclo[3.3.1.13,7]dec-1-yl-2-naphthalenyl)benzoic acid; CD3254, (E)-3-[4-hydroxy-3-(3,5,5,8,8-pentamethyl-6, 7-dihydronaphthalen-2-yl)phenyl]prop-2-enoic acid; LE135, 4-(7,8,9,10-tetrahydro-5,7,7,10,10-pentamethyl-5H-benzo[e]naphtho[2,3-b][1,4]diazepin-13-yl)benzoic acid; LG100268, 6-(1-[3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydronaphthalen-2-yl]cyclopropyl) nicotinic acid; LGD1069, 4-(1-[3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl]ethenyl) benzoic acid; MM11253, 6-(2-[5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl]-1,3-dithiolan-2-yl)-2-naphthalenecarboxylic acid; Ro406055, 4-([5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl]carboxamido)benzoic acid (also known as AM580); Ro415253, 4-[2-(7-heptoxy-4,4-dimethyl-1,1-dioxo-2,3-dihydrothiochromen-6-yl)prop-1-enyl]benzoic acid, also known as LG629; TTNPB, (E)-4-(2-[5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl]-1-propenyl)benzoic acid; UVI3003, 3-(4-hydroxy-3-[5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-3-{pentyloxy}-2-naphthalenyl]phenyl)-2-propenoic acid Benoit G, Cooney A, Giguere V, Ingraham H, Lazar M, Muscat G et al. (2006). International Union of Pharmacology. LXVI. Orphan nuclear receptors. Pharmacol Rev58: 798–836. Bour G, Lalevee S, Rochette-Egly C (2007). Protein kinases and the proteasome join in the combinatorial control of transcription by nuclear retinoic acid receptors. Trends Cell Biol17: 302–309. Duong V, Rochette-Egly C (2011). The molecular physiology of nuclear retinoic acid receptors. From health to disease. Biochim Biophys Acta (BBA) Mol Basis Dis1812: 1023–1031. Germain P, Chambon P, Eichele G, Evans RM, Lazar MA, Leid M et al. (2006a). International Union of Pharmacology. LX. Retinoic acid receptors. Pharmacol Rev58: 712–725. Germain P, Chambon P, Eichele G, Evans RM, Lazar MA, Leid M et al. (2006b). International Union of Pharmacology. LXIII. Retinoid X receptors. Pharmacol Rev58: 760–772. Lefebvre P, Benomar Y, Staels B (2010). Retinoid X receptors: common heterodimerization partners with distinct functions. Trends Endocrinol Metab21: 676–683. Maden M (2007). Retinoic acid in the development, regeneration and maintenance of the nervous system. Nat Rev Neurosci8: 755–765. Mark M, Ghyselinck NB, Chambon P (2006). Function of retinoid nuclear receptors: lessons from genetic and pharmacological dissections of the retinoic acid signaling pathway during mouse embryogenesis. Annu Rev Pharmacol Toxicol46: 451–480. Perez E, Bourguet W, Gronemeyer H, de Lera AR (2011). Modulation of RXR function through ligand design. Biochim Biophys Acta in press. Overview: Steroid hormone receptors (nomenclature as agreed by NC-IUPHAR Committee on Nuclear Receptors, see Dahlman-Wright et al., 2006; Lu et al., 2006) are nuclear hormone receptors of the NR3 class, with endogenous agonists that may be divided into 3-hydroxysteroids (oestrone and oestradiol) and 3-ketosteroids (5α-dihydrotestosterone [DHT], aldosterone, cortisol, corticosterone, progesterone and testosterone). These receptors exist as dimers coupled with chaperone molecules (such as HSP90 [ENSG00000166598] and immunophilin FKBP52 [ENSG00000004478]), which are shed on binding the steroid hormone. Although rapid signalling phenomena are observed (see Levin, 2008; Prossnitz and Maggiolini, 2009), the principal signalling cascade appears to involve binding of the activated receptors to nuclear hormone response elements of the genome, with a 15-nucleotide consensus sequence AGAACAnnnTGTTCT (i.e. an inverted palindrome) as homo- or heterodimers. They also affect transcription by protein–protein interactions with other transcription factors, such as activator protein 1 (AP-1) and nuclear factor κB (NF-κB). Splice variants of each of these receptors can form functional or nonfunctional monomers that can dimerize to form functional or non-functional receptors. For example, alternative splicing of PR mRNA produces A and B monomers that combine to produce functional AA, AB and BB receptors with distinct characteristics (Vegeto et al., 1993). A 7TM receptor responsive to estrogen (GPE, also known as GPR30, ENSG00000164850, see Prossnitz et al., 2008) has been described. Human orthologues of 7TM ‘membrane progestin receptors’ (ENSG00000182749, ENSG00000170915 and ENSG00000137819), initially discovered in fish (Zhu et al., 2003a; 2003b), appear to localize to intracellular membranes and appear to respond to ‘non-genomic’ progesterone analogues independently of G proteins (Smith et al., 2008). [3H]-Dexamethasone also binds to MR in vitro. PR antagonists have been suggested to subdivide into Type I (e.g. onapristone) and Type II (e.g. ZK112993) groups. These groups appear to promote binding of PR to DNA with different efficacies and evoke distinct conformational changes in the receptor, leading to a transcription-neutral complex (Gass et al., 1998; Leonhardt et al., 1998). Mutations in AR underlie testicular feminization and androgen insensibility syndromes, spinal and bulbar muscular atrophy (Kennedy's disease). r,r-THC exhibits partial agonist activity at ERα (Meyers et al., 1999; Sun et al., 1999). Estrogen receptors may be blocked non-selectively by tamoxifen and raloxifene, and labelled by [3H]-estradiol and [3H]-tamoxifen. Many agents thought initially to be antagonists at estrogen receptors appear to have tissue-specific efficacy (e.g. tamoxifen is an antagonist at estrogen receptors in the breast, but is an agonist at estrogen receptors in the uterus), hence the descriptor SERM (selective estrogen receptor modulator) or SnuRM (selective nuclear receptor modulator). Y134 has been suggested to be an ERα-selective estrogen receptor modulator (Ning et al., 2007). Additional ‘orphan’ estrogen-receptor-related proteins have been described (ERRα ENSG00000173153; ERRβ ENSG00000119715; ERRγ ENSG00000057103); DY131 is an agonist with selectivity for ERRβ and ERRγ compared to ERRα, ERα and ERβ (Yu and Forman, 2005). Abbreviations: DHT, 5α-dihydrotestosterone; DPN, 2,3-bis(4-hydroxyphenyl)propionitrile; DY131, N′-([1E]-[4-(diethylamino)phenyl]methylene)-4-hydroxybenzohydrazide; MPP, 1,3-bis(4-hydroxyphenyl)-4-methyl-5-(4-[2-piperidinylethoxy]phenol)-1H-pyrazole; ORG2058, 16-α-ethyl-21-hydroxy-19-norpregn-4-ene-3,20-dione; PPT, 4,4′,4″-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol; PHTPP, 4-(2-phenyl-5,7-bis[trifluoromethyl]pyrazolo[1,5-a]pyrimidin-3-yl)phenol ; R1881, 17β-hydroxy-17α-methyl-estra-4,9,11-triene-3-one, also known as methyltrienolone; r,r-THC, r,r-tetrahydrochrysene; RU26988, 11β,17β-dihydroxy-21-methyl-17α-pregna-1,4,6-trien-20-yl-3-on; RU28318, 3-oxo-7-propyl-17-hydroxy-androstan-4-en-17-yl; RU28362, 11β,17β-dihydroxy-6-methyl-17-(1-propionyl)androsta-1,4,6-triene-3-one; Y134, (6-hydroxy-2-[4-hydroxyphenyl]-benzo[b]thiophen-3-yl)-(4-[4-isopropylpiperazin-1-yl]-phenyl)methanone; WAY200070, 7-bromo-2-(4-hydroxyphenyl)-1,3-benzoxazol-5-ol; ZK112993, 11β-(4-acetylphenyl)-17β-hydroxyl-17α-(1-propinyl)-4,8-estradiene-3-one Blaustein JD (2008). Progesterone and progestin receptors in the brain: the neglected ones. Endocrinology149: 2737–2738. Briet M, Schiffrin EL (2010). Aldosterone: effects on the kidney and cardiovascular system. Nat Rev Nephrol6: 261–273. Brinton RD (2009). Estrogen-induced plasticity from cells to circuits: predictions for cognitive function. Trends Pharmacol Sci30: 212–222. Callewaert F, Boonen S, Vanderschueren D (2010). Sex steroids and the male skeleton: a tale of two hormones. Trends Endocrinol Metab21: 89–95. Chen Y, Sawyers CL, Scher HI (2008). Targeting the androgen receptor pathway in prostate cancer. Curr Opin Pharmacol8: 440–448. Dahlman-Wright K, Cavailles V, Fuqua SA, Jordan VC, Katzenellenbogen JA, Korach KS et al. (2006). International Union of Pharmacology. LXIV. Estrogen receptors. Pharmacol Rev58: 773–781. Feldman RD, Gros R (2011). Unraveling the mechanisms underlying the rapid vascular effects of steroids: sorting out the receptors and the pathways. Br J Pharmacol163: 1163–1169. Gross KL, Cidlowski JA (2008). Tissue-specific glucocorticoid action: a family affair. Trends Endocrinol Metab19: 331–339. Hadoke PW, Iqbal J, Walker BR (2009). Therapeutic manipulation of glucocorticoid metabolism in cardiovascular disease. Br J Pharmacol156: 689–712. Handelsman DJ (2008). Indirect androgen doping by oestrogen blockade in sports. Br J Pharmacol154: 598–605. Joels M, Karst H, DeRijk R, de Kloet ER (2008). The coming out of the brain mineralocorticoid receptor. Trends Neurosci31: 1–7. Kellner M, Wiedemann K (2008). Mineralocorticoid receptors in brain, in health and disease: possibilities for new pharmacotherapy. Eur J Pharmacol583: 372–378. Kerkhofs S, Denayer S, Haelens A, Claessens F (2009). Androgen receptor knockout and knock-in mouse models. J Mol Endocrinol42: 11–17. Lange CA, Gioeli D, Hammes SR, Marker PC (2007). Integration of rapid signaling events with steroid hormone receptor action in breast and prostate cancer. Annu Rev Physiol69: 171–199. Levin ER (2008). Rapid signaling by steroid receptors. Am J Physiol Regul Integr Comp Physiol295: R1425–R1430. Lu NZ, Wardell SE, Burnstein KL, Defranco D, Fuller PJ, Giguere V et al. (2006). International Union of Pharmacology. LXV. The pharmacology and classification of the nuclear receptor superfamily: glucocorticoid, mineralocorticoid, progesterone, and androgen receptors. Pharmacol Rev58: 782–797. Matsumoto T, Shiina H, Kawano H, Sato T, Kato S (2008). Androgen receptor functions in male and female physiology. J Steroid Biochem Mol Biol109: 236–241. McMaster A, Ray DW (2008). Drug insight: selective agonists and antagonists of the glucocorticoid receptor. Nat Clin Pract Endocrinol Metab4: 91–101. Mohler ML, Bohl CE, Jones A et al. (2009). Nonsteroidal selective androgen receptor modulators (SARMs): dissociating the anabolic and androgenic activities of the androgen receptor for therapeutic benefit. J Med Chem52: 3597–3617. Pippal JB, Fuller PJ (2008). Structure-function relationships in the mineralocorticoid receptor. J Mol Endocrinol41: 405–413. Prossnitz ER, Maggiolini M (2009). Non-genomic signaling by steroids. Mol Cell Endocrinol308: 1–2. Prossnitz ER, Arterburn JB, Smith HO et al. (2008). Estrogen signaling through the transmembrane G protein-coupled receptor GPR30. Annu Rev Physiol70: 165–190. Schumacher M, Sitruk-Ware R, De Nicola AF (2008). Progesterone and progestins: neuroprotection and myelin repair. Curr Opin Pharmacol8: 740–746. Simons SS, Jr. (2010). Glucocorticoid receptor cofactors as therapeutic targets. Curr Opin Pharmacol10: 613–619. Taplin ME (2007). Drug insight: role of the androgen receptor in the development and progression of prostate cancer. Nat Clin Pract Oncol4: 236–244. Weiser MJ, Foradori CD, Handa RJ (2008). Estrogen receptor β in the brain: from form to function. Brain Res Rev57: 309–320. Woolley CS (2007). Acute effects of estrogen on neuronal physiology. Annu Rev Pharmacol Toxicol47: 657–680. Overview: Thyroid hormone receptors (TRs, nomenclature as agreed by NC-IUPHAR Committee on Nuclear Receptors, see Flamant et al., 2006) are nuclear hormone receptors of the NR1A family, with diverse roles regulating macronutrient metabolism, cognition and cardiovascular homeostasis. TRs are activated by thyroxine (T4) and thyroid hormone (T3). Once activated by a ligand, the receptor acts as a transcription factor either as a monomer, homodimer or heterodimer with members of the retinoid X receptor family. An interaction with integrin αVβ3 has been suggested to underlie plasma membrane localization of thyroid hormone receptors and non-genomic signalling (Bergh et al., 2005). One splice variant, TRα2, lacks a functional DNA-binding domain and appears to act as a transcription suppressor. Although radioligand binding assays have been described for these receptors, the radioligands are not commercially available. NH-3 has been described as an antagonist at thyroid hormone receptors with modest selectivity for TRβ (Nguyen et al., 2002). Abbreviations: GC1, (3,5-dimethyl-4-[4-hydroxy-3-isopropylbenzyl]phenyloxy)acetate; NH-3, (4-[4-hydroxy-3-isopropyl-5-{4-nitrophenylethynyl}-benzyl]-3,5-dimethylphenoxy)acetate Baxter JD, Webb P (2009). Thyroid hormone mimetics: potential applications in atherosclerosis, obesity and type 2 diabetes. Nat Rev Drug Discov8: 308–320. Bernal J (2007). Thyroid hormone receptors in brain development and function. Nat Clin Pract Endocrinol Metab3: 249–259. Cheng SY, Leonard JL, Davis PJ (2010). Molecular aspects of thyroid hormone actions. Endocr Rev31: 139–170. Davis PJ, Zhou M, Davis FB, Lansing L, Mousa SA, Lin HY (2010). Mini-review: cell surface receptor for thyroid hormone and nongenomic regulation of ion fluxes in excitable cells. Physiol Behav99: 237–239. Davis PJ, Davis FB, Mousa SA, Luidens MK, Lin HY (2011). Membrane receptor for thyroid hormone: physiologic and pharmacologic implications. Annu Rev Pharmacol Toxicol51: 99–115. Flamant F, Baxter JD, Forrest D, Refetoff S, Samuels H, Scanlan TS et al. (2006). International Union of Pharmacology. LIX. The pharmacology and classification of the nuclear receptor superfamily: thyroid hormone receptors. Pharmacol Rev58: 705–711. Furuya F, Lu C, Guigon CJ, Cheng SY (2009). Nongenomic activation of phosphatidylinositol 3-kinase signaling by thyroid hormone receptors. Steroids74: 628–634. Grover GJ, Mellstrom K, Malm J (2007). Therapeutic potential for thyroid hormone receptor-β selective agonists for treating obesity, hyperlipidemia and diabetes. Curr Vasc Pharmacol5: 141–154. Liu YY, Brent GA (2010). Thyroid hormone crosstalk with nuclear receptor signaling in metabolic regulation. Trends Endocrinol Metab21: 166–173. Pramfalk C, Pedrelli M, Parini P (2011). Role of thyroid receptor β in lipid metabolism. Biochim Biophys Acta (BBA) Mol Basis Dis1812: 929–937. Visser WE, Friesema EC, Jansen J, Visser TJ (2008). Thyroid hormone transport in and out of cells. Trends Endocrinol Metab19: 50–56. Yen PM, Ando S, Feng X, Liu Y, Maruvada P, Xia X (2006). Thyroid hormone action at the cellular, genomic and target gene levels. Mol Cell Endocrinol246: 121–127. Overview: Vitamin D (VDR), Pregnane X (PXR) and Constitutive Androstane (CAR) receptors (nomenclature as agreed by NC-IUPHAR Committee on Nuclear Receptors, Moore et al., 2006) are members of the NR1I family of nuclear receptors, which form heterodimers with members of the retinoid X receptor family. PXR and CAR are activated by a range of exogenous compounds, with no established endogenous physiological agonists, although high concentrations of bile acids and bile pigments activate PXR and CAR (see Moore et al., 2006). Abbreviations: CITCO, 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde-O-(3,4-dichlorobenzyl)oxime; EB1089, (1R,3S,5Z)-5-[(2E)-2-[(1R,3aS,7aR)-1-[(1R,2E,4E)-6-ethyl-6-hydroxy-1-methyl-2,4-octadien-1-yl]-octahydro-7α-methyl-4H-inden-4-ylidene]ethylidene]-4-methylene-1,3-cyclohexanediol, also known as seocalcitol; TEI9647, (23s)-25-dehydro-1α hydroxyvitamin D3-26,23-lactone; TCPOBOP, 1,4-bis-[2-(3,5-dichloropyridyloxy)]benzene; ZK159222, butyl 1-[(E,1R,4R)-4-[(1R,3aS,4E,7aR)-4-[(2Z)-2-[(3S,5R)-3,5-dihydroxy-2-methylidenecyclohexylidene]ethylidene]-7a-methyl-2,3,3a,5,6,7-hexahydro-1H-inden-1-yl]-1-hydroxypent-2-enyl]cyclopropane-1-carboxylate Bikle D (2009). Nonclassic actions of vitamin D. J Clin Endocrinol Metab94: 26–34. Bikle DD (2011). Vitamin D: an ancient hormone. Exp Dermatol20: 7–13. Bouillon R, Carmeliet G, Verlinden L et al. (2008). Vitamin D and human health: lessons from vitamin D receptor null mice. Endocr Rev29: 726–776. Campbell FC, Xu H, El-Tanani M, Crowe P, Bingham V (2010). The yin and yang of vitamin D receptor (VDR) signaling in neoplastic progression: operational networks and tissue-specific growth control. Biochem Pharmacol79: 1–9. Danescu LG, Levy S, Levy J (2009). Vitamin D and diabetes mellitus. Endocrine35: 11–17. Deeb KK, Trump DL, Johnson CS (2007). Vitamin D signalling pathways in cancer: potential for anticancer therapeutics. Nat Rev Cancer7: 684–700. Haussler MR, Haussler CA, Bartik L et al. (2008). Vitamin D receptor: molecular signaling and actions of nutritional ligands in disease prevention. Nutr Rev66: S98–112. Ihunnah CA, Jiang M, Xie W (2011). Nuclear receptor PXR, transcriptional circuits and metabolic relevance. Biochim Biophys Acta (BBA) Mol Basis Dis1812: 956–963. Kohle C, Bock KW (2009). Coordinate regulation of human drug-metabolizing enzymes, and conjugate transporters by the Ah receptor, pregnane X receptor and constitutive androstane receptor. Biochem Pharmacol77: 689–699. Krishnan AV, Feldman D (2011). Mechanisms of the anti-cancer and anti-inflammatory actions of vitamin D. Annu Rev Pharmacol Toxicol51: 311–336. Moore DD, Kato S, Xie W, Mangelsdorf DJ, Schmidt DR, Xiao R et al. (2006). International Union of Pharmacology. LXII. The NR1H and NR1I receptors: constitutive androstane receptor, pregnene X receptor, farnesoid X receptor α, farnesoid X receptor β, liver X receptor α, liver X receptor β, and vitamin D receptor. Pharmacol Rev58: 742–759. Pittas AG, Dawson-Hughes B (2010). Vitamin D and diabetes. J Steroid Biochem Mol Biol121: 425–429. Plum LA, DeLuca HF (2010). Vitamin D, disease and therapeutic opportunities. Nat Rev Drug Discov9: 941–955. Timsit YE, Negishi M (2007). CAR and PXR: the xenobiotic-sensing receptors. Steroids72: 231–246. Urquhart BL, Tirona RG, Kim RB (2007). Nuclear receptors and the regulation of drug-metabolizing enzymes and drug transporters: implications for interindividual variability in response to drugs. J Clin Pharmacol47: 566–578.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call