The orphan nuclear receptors at their 25-year reunion

in Journal of Molecular Endocrinology
Authors:
Shannon E MullicanDivision of Endocrinology, Diabetes, and Metabolism, Department of Medicine, and The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA

Search for other papers by Shannon E Mullican in
Current site
Google Scholar
PubMed
Close
,
Joanna R DiSpiritoDivision of Endocrinology, Diabetes, and Metabolism, Department of Medicine, and The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA

Search for other papers by Joanna R DiSpirito in
Current site
Google Scholar
PubMed
Close
, and
Mitchell A LazarDivision of Endocrinology, Diabetes, and Metabolism, Department of Medicine, and The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA

Search for other papers by Mitchell A Lazar in
Current site
Google Scholar
PubMed
Close
View More View Less

Free access

The nuclear receptor superfamily includes many receptors, identified based on their similarity to steroid hormone receptors but without a known ligand. The study of how these receptors are diversely regulated to interact with genomic regions to control a plethora of biological processes has provided critical insight into development, physiology, and the molecular pathology of disease. Here we provide a compendium of these so-called orphan receptors and focus on what has been learned about their modes of action, physiological functions, and therapeutic promise.

Abstract

The nuclear receptor superfamily includes many receptors, identified based on their similarity to steroid hormone receptors but without a known ligand. The study of how these receptors are diversely regulated to interact with genomic regions to control a plethora of biological processes has provided critical insight into development, physiology, and the molecular pathology of disease. Here we provide a compendium of these so-called orphan receptors and focus on what has been learned about their modes of action, physiological functions, and therapeutic promise.

Introduction

In the late 1980s the observation that the steroid hormone receptors shared highly conserved domains encouraged researchers to search for additional members of the nuclear receptor (NR) family (Mangelsdorf et al. 1995, Giguère 1999, Willson & Moore 2002, Benoit et al. 2006). Unlike that of the classic NRs that had previously been identified with prior knowledge of a naturally occurring ligand, these new members were initially without ligand, and therefore referred to as orphans. Over the span of a decade, using probes designed from conserved NR domains to screen cDNA libraries and degenerate primers for target amplification, as well as automated searches of EST databases, 36 vertebrate orphan NRs were identified (Fig. 1; Willson & Moore 2002).

Figure 1
Figure 1

Orphan NR Timeline. From the late 1980s till the end of the 1990s, there was rapid growth of the NR superfamily with the addition of 36 orphan members. Once the cloning and discovery of novel members plateaued, the focus of the field shifted to intense functional characterization. Here each orphan is listed by its common name and color-coded based on its designated NR subfamily.

Citation: Journal of Molecular Endocrinology 51, 3; 10.1530/JME-13-0212

Orphan NRs consist of the four major domains that characterize classic nuclear hormone receptors (Aranda & Pascual 2001, Huang et al. 2010, Helsen et al. 2012). The amino terminus contains the A/B domain consisting of activation function 1 (AF1) and among orphans, this region is quite variable in size. The DNA-binding domain (DBD) consists of two zinc finger motifs and confers response element specificity; it is typically highly conserved within orphan receptor subgroups. Linking the DBD to the carboxy-terminal ligand-binding domain (LBD) is the hinge region, whose length varies between subfamilies. The pocket formed by the LBD can also vary greatly in size and by the absence or presence of the AF2 region that mediates coactivator interaction.

Classic NRs are transcription factors regulated by the high affinity binding of naturally occurring small molecules, which dictate receptor subcellular localization and conformation. The latter determines coactivator–repressor interactions and thereby transactivation potential (Mangelsdorf et al. 1995, Aranda & Pascual 2001). In contrast, while the regulation of gene transcription by orphan NRs also depends on interactions with coactivator and corepressor complexes, the role of ligand varies (Benoit et al. 2004, Markov & Laudet 2011). Nevertheless, once an endogenous ligand has been identified, the corresponding orphan is then considered ‘adopted’ (Benoit et al. 2006).

Due to their potential ligand regulation, orphan NRs have the prospect of serving as therapeutic targets of small molecules (Mukherjee & Mani 2010). Thus, there has been an intense focus on the physiological roles and molecular mechanisms of orphan NRs over the last 25 years (Benoit et al. 2006). All vertebrate orphan NRs have been globally deleted in mice and some have been overexpressed and/or selectively targeted spatially and/or temporally. To illustrate the enormous impact of this technology on our understanding of orphan NR biology, all relevant mouse models are summarized in Table 1.

Table 1

Phenotypes of orphan receptor mutant mouse strains. Orphan NR mutant strains that have been published and are listed in the Mouse Genome Informatics (MGI) database. We have listed the general physiological systems and functions affected by each mutation. For more details, see the MGI resource (www.informatics.jax.org) from which this information was collated

ReceptorEmbryonic lethalWhole body mutantsTissue-specific mutants
DAX1 (NR0B1)NoKO: fertility (males)NR
SHP (NR0B2)NoKO: fertility; energy expenditure, cholesterol metabolism, hepatic inflammation (diet-induced)Liver: cholesterol and TG metabolism
PPARα (NR1C1)NoKO: fatty acid oxidation (fasting); cholesterol and TG metabolism; glycemic control (HFD); cardiac fibrosis; wound healing Muscle (transgene): glycemic control (HFD)
Heart (transgene): TG and cholesterol metabolism
PPARβ/δ (NR1C2)YesKO: placentation; growth; dyslipidemia; cancer; myelination; energy expenditure; cholesterol and TG metabolismAdipose/brain/macrophage: normal

Pan-hematopoietic: atherosclerosis; glycemic control (HFD); hematopoietic

Myeloid: autoimmunity; glycemic control (HFD)

Skeletal muscle: glycemic control; fiber type; regeneration

Pancreas: insulin secretion

Heart (transgene): glycemic control
PPARγ (NR1C3)YesKO: death by E10.5 (placentation)

KI (hypomorphic): premature death, AT deposition, glycemic control, TG metabolism

KI (Pro12Ala): body weight; glycemic control; TG + cholesterol metabolism

KI (P465L): embryonic lethal; BAT activity (het)KI (L466A): embryonic lethal; AT deposition, TG metabolism, glycemic control (hets)

KI (S112A): glycemic control

KO (Mox2-Cre): AT deposition, glycemic control, blood pressure; circadian rhythm

KO (γ2 isoform only): AT deposition (strain-dependent); glycemic control

KO (γ2 isoform only): AT deposition (strain-dependent); glycemic control
Pancreas: beta cell mass

I-Pancreas: no beta cell defects

Heart: hypertrophy, systolic function

Myeloid: autoimmunity; glycemic control; polarization; glycemic control (HFD), immunity to infection; atherosclerosis

Kidney: body weight, blood volume

Liver: TG metabolism, glycemic control

Muscle: AT deposition; glycemic control

Lung epithelium: lung structure

Mammary epithelium: no mammary defects

Ovary: ovulation (pre-ovulatory follicles)

Endothelium/BM: TG metabolism; blood pressure rhythms

Treg: adipose inflammation (HFD)

B cell: antibody production

CNS: no abnormalities reported

Adipose (aP2): AT deposition; body weight; glycemic control

I-Adipose (aP2): AT maintenance
REV-ERBα (NR1D1)NoKO: postnatal lethality; fertility; circadian (behavior, cholesterol and TG metabolism, immune response); bile acids; cerebellarNR
REV-ERBβ (NR1D2)NoKO: no reported abnormalitiesNR
REV-ERBα/βNoNRDKO liver: circadian; dyslipidemia
RORα (NR1F1)NoENU: postnatal lethality; cerebellar; paralysisNR
KO: viability; atherosclerosis; cerebellar; cholesterol and TG homeostasis; fertility; adrenal; hematopoietic
RORβ (NR1F2)NoKO: postnatal lethality; growth; fertility; learning/memory; motor; ocular NR
RORγ (NR1F3)NoKO: hematopoieticT cell: autoimmunity
LXRβ (NR1H2)NoKO: fertility; skin; CNS (amyloid deposition)NR
LXRα (NR1H3)NoKO: cholesterol and TG homeostasis (high cholesterol diet); infectionLiver: cholesterol and TG homeostasis (high cholesterol diet); atherosclerosis
KI: constitutively active LXRa confers resistance to LPS-induced lung injury
LXRαβ NoDKO: CNS (lipid deposits); cholesterol and TG metabolism; immunity to infection; fertility
FXR (NR1H4)NoKO: bile acids; dyslipidemiaLiver: bile acids; dyslipidemia
Intestine: bile acids
PXR (NR1I2)NoKO: xenobiotic responseNR
KI: species-specific xenobiotic responses (human)
CAR (NR1I3)NoKO: xenobiotic response; lipid homeostasis; body weight (caloric restriction)NR
KI: species-specific xenobiotic responses (human)
HNF4α (NR2A1)YesKO: embryogenesis, die before E10.5Liver: hepatogenesis; glycogen; lipid homeostasis body weight; viability
KI: glycemic control (a1 isoform only)
KI: dyslipidemia (a7 isoform only)Beta cell: insulin secretion
HNF4γ (NR2A2)NoKO: body weight; locomotor activity and energy expenditure (circadian)NR
RXRα (NR2B1)YesKO: cardiac, ocular and placental defects, die before E16.5KO AF-1 only: partial postnatal lethality; cardiac; ocular; growth retardationLiver: lipid and cholesterol metabolism, bile acids (high cholesterol diet); body weight, glycemic control (HFD)

I-Liver: regeneration

I-Adipose (aP2): body weight

Macrophage: autoimmunity; endotoxin response

Epicardium: cardiovascular developmental defects

Retina: photoreceptors

Skin: hair and coat color

Prostate: pre-neoplastic lesions
RXRβ (NR2B2)PartialKO: viability, fertilityNR
KO AF2 only: cholesterol metabolism
RXRγ (NR2B3)NoKO: thyroid; cholinergic neurons; CNS re-myelinationNR
RXRα/β/γMultiple combinatorial KO, see MGI database for details
TR2 (NR2C1)NoKO: fetal hemoglobin NR
TR4 (NR2C2)PartialKO: viability; aging; growth; fertility; cerebellar; body weight, glycemic control, lipid homeostasis, energy expenditure, inflammation (HFD)NR
TR2/4 YesDKO: embryogenesis, die before E12
TLX (NR2E1)NoKO: CNS (limbic development); neurogenesis (adult); growth; fertility; ocular; aggressionCNS: aggression
I-CNS: spatial learning
PNR (NR2E3)NoKO: ocular (retina, rods and cones)NR
COUP-TFI (NR2F1)NoKO: viability (brain development)
COUP-TFII (NR2F2)YesKO: angiogenesis and cardiac development

I-KO: lymphatic (induction of systemic Cre at E8.5); fertility (induction of Cre at P14)
Uterus: placentation

Endothelium: venous and lymphatic development

Beta cells: insulin secretion

Stomach: morphogenesis
COUP-TFI/II NoDKO eye: morphogenesis
EAR-2 (NR2F6)NoKO: circadian rhythm; thermal nocioception; autoimmunityNR
ERRα (NR3B1)NoKO: adipose deposition; stress susceptibility (cardiac, temperature, infection); bone structureNR
ERRβ (NR3B2)YesKO: death by E10.5 (placentation) Embryonic: hearing (inner ear development)
ERRγ (NR3B3)NoKO: viability; cardiac; spinal cord; kidney NR
NURR77 (NR4A1)NoKO: body weight (HFD); atherosclerosisNR
NURR1 (NR4A2)NoKO: complete postnatal lethality by 12 h; selective loss of dopaminergic neurons NR
NOR1 (NR4A3)YesKO: death by E8.5 (gastrulation); seizures; inner ear; atherosclerosisNR
NURR77/NOR1 DKONoKO: viability; leukemia (growth, hepatomegaly, anemia)NR
SF1 (NR5A1)NoKO: viability (adrenals); gonads; hypothalamus

KI: K119 and K194 mutated to R, loss of sumoylation on those residues associated with abnormal adrenal function and infertility
Anterior pituitary: fertility (gonad maturation)

Gonads: fertility (gonad maturation)

CNS: behavior (anxiety); body weight (HFD)
LRH-1 (NR5A2)YesKO: death before E7.5Liver: slight dyslipidemia; bile acid composition
HET: intestinal crypts Intestine: slight dyslipidemia
Gonads: fertility (ovulation)
GCF (NR6A1)YesKO: death by E10.5 (cardiac development)Oocyte: fertility (oocyte morphology, steroidogenesis)

KO, knock-out; DKO, double knock-out; KI, knock-in; I, inducible Cre transgene; NR, not yet reported; AT, adipose tissue; HFD, high-fat diet; TG, triglyceride.

There are many orphan NRs in mammals as well as in lower organisms. Orphan NRs in Drosophila melanogaster and Caenorhabditis elegans have been reviewed elsewhere (Taubert et al. 2011, Fahrbach et al. 2012). Here, we review current knowledge about each of the 36 orphan NRs that has a human ortholog. The entire NR superfamily has been categorized into six structurally distinct groups based on phylogenetic analysis, producing a unified nomenclature system that identifies each NR with less ambiguity (Laudet 1997, Nuclear Receptors Nomenclature Committee 1999, Germain et al. 2006). In this review, each orphan is introduced as part of its official NR group, then addressed by its most commonly used name. The discussion of each is necessarily brief, highlighting its discovery, regulation, and physiological functions, particularly those with therapeutic implications. For more detailed information on individual NRs, readers are directed to the NURSA website (www.nursa.org) and to more comprehensive reviews.

The odd ones: orphans of the NR0B group

Nr0b1/Dax1 and Nr0b2/Shp

DAX1 and SHP are atypical NRs harboring a classifiable NR LBD in their carboxyl-terminus, but lack a classic NR DBD and instead have a region resembling the NR interaction motifs characteristic of coactivators (Zanaria et al. 1994, Seol et al. 1996, Lalli & Sassone-Corsi 2003, Båvner et al. 2005, Ehrlund & Treuter 2012). The name small heterodimer partner (Shp) reflects the ability of SHP and DAX1 to bind to the AF2 of other NRs, preventing coactivator recruitment while recruiting corepressor complexes and acting as transcriptional repressors (Lalli & Sassone-Corsi 2003, Båvner et al. 2005, Ehrlund & Treuter 2012). While most reports indicate that DAX1 and SHP function as transcriptional repressors, they may also enhance transcription (Kim et al. 2001, Nishizawa et al. 2002, Xu et al. 2009, Kelly et al. 2010).

To date, no ligands for the NR0B receptors have been well-established, although retinoid-related molecules have been reported to bind and enhance the repressive function of SHP (Miao et al. 2011, Ehrlund & Treuter 2012). Indeed, a crystal structure study of DAX1 demonstrated a ligand-binding pocket (LBP) filled with amino acid side chains, suggesting typical ligand regulation of this orphan to be unlikely (Sablin et al. 2008). Sequence conservation between Nr0b group members suggests this to be a shared characteristic, and thus, it appears that the Nr0b group is regulated at the level of expression, alternative splicing, posttranslational modifications, and miRNA targeting (Hossain et al. 2004, Miao et al. 2009, Xiao et al. 2012, Seok et al. 2013).

Although DAX1 and SHP are mechanistically very similar, their tissue expression patterns are distinct, suggesting different physiological roles (Ehrlund & Treuter 2012). Nr0b1 was originally identified as a gene frequently deleted or mutated in X-linked adrenal hypoplasia congenita (AHC) and also duplicated in dosage-sensitive sex reversal (DSS), leading to its more common name, DSS-AHC critical region on the X, gene 1, or Dax1 (Muscatelli et al. 1994, Zanaria et al. 1994, Lalli & Sassone-Corsi 2003). Dax1 is highly expressed in the hypothalamic–pituitary–adrenal–gonadal axis and has essential roles in gametogenesis and sex specification mediated in part by repression of NR5A1 (SF1; Lalli & Sassone-Corsi 2003, Iyer & McCabe 2004). In contrast, Shp is highly expressed in the liver and small intestine, where it represses NR5A2 (LRH1) to regulate cholesterol, bile acid (BA), and glucose metabolism (Båvner et al. 2005, Chanda et al. 2008).

Both members of the Nr0b group have also been reported to repress the transcriptional activity of other factors besides NRs (Kim et al. 2004, 2012, Suh et al. 2006, Kinsey et al. 2009, Sun et al. 2009, Yuk et al. 2011). For example, SHP functions as a corepressor of NF-κB during inflammatory Toll-like receptor signaling (Yuk et al. 2011). Moreover, the ability to bind and repress OCT4 activity has identified DAX1 as a critical component in maintaining embryonic stem cell pluriopotency (Wang et al. 2006, Kim et al. 2008, Sun et al. 2009). Finally, the potential role of Nr0b receptors in carcinogenesis is highlighted by the functional interaction of DAX1 with the EWS/FLI fusion protein in Ewing's sarcoma (Kinsey et al. 2009).

There for dinner: orphans of the NR1C group

Nr1c1/Pparα, Nr1c2/Pparβ or δ, and Nr1c3/Pparγ

Peroxisome proliferator-activated receptors (PPARs) regulate carbohydrate and lipid metabolism in major metabolic organs (liver, adipose, and muscle) as well as in peripheral tissues with metabolic roles (intestine, hematopoietic, and hypothalamic). The ‘PPAR’ name followed the discovery that PPARα was activated by clofibrate, a lipid-lowering drug that causes peroxisome proliferation in the rodent liver (Issemann & Green 1990); however, the β/δ and γ subtypes have not been shown to cause peroximsomal proliferation. Despite ongoing searches for dominant endogenous ligands, the PPARs are now commonly thought of as ‘sensors’ for a wide range of fatty acid molecules, a promiscuity that may be explained by their large LBP (Nolte et al. 1998, Uppenberg et al. 1998, Xu et al. 1999, 2001, Michalik 2006). However, high affinity and relatively specific synthetic ligands exist for each PPAR (Bensinger & Tontonoz 2008). As noted earlier, lipid-lowering fibrates activate PPARα, and agonists of PPARγ function as anti-diabetic drugs (Lehrke & Lazar 2005).

Activation of PPARα and PPARβ/δ supports fatty acid oxidation (FAO) in the heart, liver, and muscle, while PPARγ promotes lipid storage in adipose tissue (Michalik 2006). Thus, loss of PPARα in the liver blocks elevated FAO in response to fasting, leading to hepatic and myocardial lipid accumulation, hypoglycemia, and elevated serum–free fatty acids (Kersten et al. 1999, Leone et al. 1999). Similarly, PPARβ/δ activity in the muscle promotes formation of highly oxidative slow-twitch fibers, in part through its positive regulation of PGC1α (Evans et al. 2004, Schuler et al. 2006). Interestingly, these two receptors have opposing roles in cardiac myocytes of the diabetic heart, where PPARβ/δ promotes glucose uptake, utilization, and overall cardiac health, while the PPARα pathway decreases glucose utilization and promotes the myocardial lipid accumulation associated with cardiac disease (Burkart et al. 2007). PPARγ is strongly induced during adipocyte differentiation and its activity is necessary and, in some cases, sufficient for adipogenesis (Chawla & Lazar 1994, Tontonoz et al. 1994, Tontonoz & Spiegelman 2008). Two isoforms, γ1 and γ2, identical except for an additional 30 amino acids at the N-terminus of γ2, are highly expressed in adipocytes, while other cell types express comparatively lower levels of the γ1 isoform (Tontonoz et al. 1994, Nagy et al. 2013). Loss of PPARγ expression causes lipodystrophy in mice as do dominant negative mutants of PPARγ in both mice and humans (Barak et al. 2002, Freedman et al. 2005, Semple 2006, Duan et al. 2007). PPARγ also regulates lipid metabolism in macrophages and appears to have an overlapping but non-redundant role with PPARδ in the regulation of macrophage polarization and homeostatic scavenging of apoptotic cells (Ahmadian et al. 2013).

PPARs bind DNA as obligate heterodimers with Nr2b1 (Rxr) family members to a direct repeat (DR) of the sequence AGGTCA separated by one nucleotide, usually an adenine (Kliewer et al. 1992b, IJpenberg et al. 1997, Varga et al. 2011). This DR1 element was initially defined using in vitro binding and transactivation assays and has been confirmed as the predominant recognition motif at PPAR binding sites identified on a genome-wide scale in vivo (Dreyer et al. 1992, Kliewer et al. 1992b, Tugwood et al. 1992, Lefterova et al. 2008, 2010, Nielsen et al. 2008, Mikkelsen et al. 2010, Boergesen et al. 2012).

It's about time: orphans of the NR1D group

Nr1d1/Rev-erbα and Nr1d2/Rev-erbβ

Rev-erbα was discovered as a coding sequence on the reverse strand of the c-erbα gene, which encodes a thyroid hormone receptor, and the identification of Rev-erbβ followed several years later (Lazar et al. 1989, Dumas et al. 1994, Forman et al. 1994, Retnakaran et al. 1994). Structurally, the Rev-erbs are unique among NRs because they lack a critical alpha-helix (H12) in the AF2 domain that mediates coactivator interaction, resulting in a conformation more suitable for corepressor recruitment (Renaud et al. 2000, Ramakrishnan & Muscat 2006, Yin et al. 2010). Both Rev-erbα and Rev-erbβ act as receptors for heme, which binds within the classic NR LBP (Reinking et al. 2005, Raghuram et al. 2007, Yin et al. 2007, Burris 2008). Several synthetic agonists have been developed and show potential for modulating Rev-erb in vivo (Yin et al. 2010, Solt et al. 2012). Besides ligand binding, regulation of Rev-erb activity also occurs at the level of gene expression, posttranslational modifications, and protein–protein interactions (Yin et al. 2010, Chini et al. 2013). For example, Rev-erb expression in most tissues is circadian and is tightly controlled by the core circadian clock TFs, BMAL1 and CLOCK, as well as repressing its own transcription (Feng & Lazar 2012). Furthermore, Rev-erbα is stabilized by GSK3β-mediated phosphorylation that can be blocked by lithium exposure; in hippocampal neurons, oligophrenin 1 alters cellular localization of Rev-erbα, blocking its repressive function (Valnegri et al. 2011, Feng & Lazar 2012).

Acting as dedicated transcriptional repressors, both Rev-erbs bind to extended half-sites (AGGTCA) with an A/T-rich 5′ end (RORE), a sequence identical to that recognized by NR1F (ROR) orphan receptors but with opposing effects. Importantly, Rev-erb monomer's association with isolated ROREs is not sufficient for active transcriptional repression (although it can still compete with ROR binding to the RORE); however, two neighboring monomeric Rev-erbs can cooperate to recruit corepressors to repress gene expression (Yin et al. 2010). Heme stabilizes the interaction with NCoR, augmenting repression of transcription and defining the Rev-erb group as ligand-stimulated transcriptional repressors (Pardee et al. 2009). Rev-erbα and β also dimerize and function at a DR of their unique half-site spaced by two additional nucleotides (Rev-DR2; Yin et al. 2010).

The Rev-erbs have been implicated in a variety of physiological processes, including cerebellar development, osteoarthritis, adipogenesis, and mitochondria biogenesis, but the most well-known cellular function is to act as integrators of circadian rhythm and metabolic pathways (Chomez et al. 2000, Chaturvedi et al. 2006, Ramakrishnan & Muscat 2006, Wang & Lazar 2008, Yin et al. 2010, Woldt et al. 2013). Genome-wide studies have indicated that the cyclic expression and genomic presence of both Rev-erbs result in the proper rhythmic expression of circadian clock and lipid metabolism genes and underscoring the importance of this convergence, Rev-erb deficiency in the liver results in hepatic steatosis and dysregulation of the cell autonomous hepatic clock (Bugge et al. 2012, Cho et al. 2012). Recent work has also described that Rev-erbs represent a key link between the cellular clock and macrophage function, where they function cyclically to limit the expression of cytokines impacting responses to pathogens (Fontaine et al. 2008, Gibbs et al. 2012, Chandra et al. 2013, Lam et al. 2013).

More time for discussion: orphans of the NR1F group

Nr1f1/Rorα, Nr1f2/Rorβ, and Nr1f3/Rorγ

The RAR-related orphan NRs comprise three structurally related orphan NRs with roles in retinal development, lipid and glucose homeostasis, atherosclerosis, carcinogenesis, circadian rhythm, bone formation, and immunity (Becker-André et al. 1993, Medvedev et al. 1996, Jetten 2009, Solt & Burris 2012). Of particular note, RORα is required for cerebellar development, and RORγ plays a critical role in lymphoid cells (Hamilton et al. 1996, Kurebayashi et al. 2000, Ivanov et al. 2006, Solt & Burris 2012).

RORs bind as monomers to the same RORE sequence bound by Rev-erbs and are described as constitutive activators, recruiting coactivators in absence of ligand (Jetten 2009). However, independent investigations have suggested that oxysterols and cholesterol derivatives act as agonist and inverse agonist ligands of RORα and γ (Kallen et al. 2002, Jin et al. 2010, Wang et al. 2010). In addition, retinoids and retinoic acid have been shown to antagonize the activity of RORβ (Stehlin-Gaon et al. 2003). Despite the uncertainty surrounding endogenous ligand regulation of RORs, the generation of synthetic agonists and inverse agonists has great therapeutic promise, particularly in modulating the function of the RORγ-t isoform in TH17 lymphocytes during autoimmunity (Yang et al. 2008, Huh et al. 2011, Solt et al. 2011).

Like other NRs, activity of RORs can be regulated by posttranslational modifications, including phosphorylation and sumoylation (Hwang et al. 2009, Onishi et al. 2009, Lee et al. 2010). Intriguingly, a ‘methyl degron’ sequence has been recently described in RORα, making it the first non-histone substrate targeted by methyltransferases to regulate protein stability (Lee et al. 2012). Antagonism with other TFs also mediates ROR activity, demonstrated by the recent observation that RORγ function is inhibited by direct interaction with Foxp3 (Ichiyama et al. 2008, Zhou et al. 2008). Furthermore, the ability of RORα and Rev-erbα to bind to overlapping sites and counteract each other's activity at target genes has been demonstrated in the circadian control of Bmal1 and Rev-erbα expression (Guillaumond et al. 2005). In addition to direct transcriptional regulation at specific response elements, the transrepression of Wnt/β-catenin-mediated gene activation has been reported (Lee et al. 2010). Furthermore supporting the DNA-binding independent functions of RORs, RORα has been reported to bind and stabilize p53 protein and thereby indirectly regulate p53 target gene expression (Kim et al. 2011, Wang et al. 2012).

Watching what they eat: orphans of the NR1H group

Nr1h2/Lxrβ, Nr1h3/Lxrα, and Nr1h4/Fxrα

The Nr1h family includes liver X receptors, LXRα and LXRβ, and farnesol X receptors, FXRα and FXRβ, although FXRβ is found in mice but not in humans (Moore et al. 2006). LXRs and FXRs have metabolically intertwined roles that converge on minimizing the buildup of cholesterol by responding to elevated levels of sterols and BAs respectively (Calkin & Tontonoz 2012). Endogenous LXR ligands that have been reported include: 24(S), 25-epoxycholesterol in the liver, 24(S)-hydroxycholesterol in the brain, and 27-hydroxycholesterol in macrophages (Moore et al. 2006). Synthetic, high affinity pan-LXR agonists are in wide experimental use. These compounds promote the cholesterol-lowering activity of LXRs, but this benefit is offset by activation of the SREBP1C pathway of de novo lipogenesis in the liver, which has hindered the advancement of LXR agonists as therapies for human metabolic disorders (Repa et al. 2000, Joseph et al. 2002, Tangirala et al. 2002, Levin et al. 2005). FXR was initially named based on its activation by very high concentrations of farnesol and its metabolites in vitro, but was later found to respond to physiological concentrations of BAs and is now widely viewed as a nuclear BA receptor and central regulator of BA homeostasis (Forman et al. 1995, Makishima et al. 1999, Parks et al. 1999, Wang et al. 1999). BAs vary in their affinity for FXR with the strongest agonist being the primary BA chenodeoxycholic acid (CDCA; Modica et al. 2010). Importantly, synthetic FXR agonists are efficacious in treating mouse models of cholestatic diseases (Moschetta et al. 2004).

Subtype and cell type-specific deletions of LXR have uncovered their roles in the regulation of whole-body cholesterol homeostasis. In atherosclerosis-prone mouse strains, LXRα is required for preventing cholesterol overload in several tissues including the liver, where LXRα activation cannot be compensated for by LXRβ; macrophages, where it contributes equally with LXRβ; and likely other tissues including adipose and intestine (Lehrke et al. 2005, Bradley et al. 2007, Hong et al. 2012, Zhang et al. 2012). Similarly, LXRβ has a non-redundant role in preventing cholesterol accumulation in BA-metabolizing cholangiocytes, the CNS, spinal cord, and male gonadal cells (Gabbi et al. 2009, Xia et al. 2012). For FXR, activation by BAs initiates a negative feedback loop that limits BA synthesis and promotes their transit from hepatocytes to the enterohepatic circulation. Consistent with this, FXR-deficient mice challenged with high levels of dietary BAs suffer severe hepatotoxicity (Sinal et al. 2000).

All Nr1h members heterodimerize with RXR and recent genome-wide binding studies have demonstrated widespread use of sequence motifs identified using older low-throughput methods. In liver, the vast majority of FXR-binding sites identified by ChIP-seq, contained an IR1 motif (inverted repeats of the canonical NR half site AGGTCA separated by one nucleotide) which was initially reported as the preferred motif for FXR-RXR heterodimers (Laffitte et al. 2000, Chong et al. 2010, Thomas et al. 2010). Similarly, genome-wide profiling confirmed co-occupancy of LXR–RXR at DR4 motifs, initially characterized almost 20 years ago (Willy et al. 1995, Boergesen et al. 2012).

The defenders: orphans of the NR1I group

Nr1i2/Pxr and Nr1i3/Car

The pregnane X receptor (PXR) and constitutive androstane receptor (CAR) play a key role in the body's defense against xenobiotics (foreign materials encountered in the environment). Mouse and human PXRs were discovered almost simultaneously, revealing PXR as a conserved, direct regulator of Cyp3a family enzymes important in xenobiotic and endobiotic metabolism (Bertilsson et al. 1998, Blumberg et al. 1998, Kliewer et al. 1998, Lehmann et al. 1998). PXR can bind structurally distinct ligands that range in size from 268 to 823 Da and are highly species-specific, in accordance with the relatively low conservation of the PXR LBP among orthologs (Reschly & Krasowski 2006). This diverse array of xenobiotics, dietary compounds, and endobiotics includes rifampicin, clotrimazole, phenobarbital, the herbal antidepressant St John's wort, HIV protease inhibitors, and BAs (Zhou et al. 2009, Tolson & Wang 2010). Initial cloning of CAR reported its transcriptional activity in the absence of an exogenous ligand, leading to the name constitutive active receptor before androstanes were identified as inverse agonists (Baes et al. 1994, Forman et al. 1998). To date, many xenobiotic agonists have been identified with phenobarbital-like compounds being the most widely used to study CAR's function (Gao & Xie 2010).

Both receptors induce phase I and II drug metabolizing enzymes in the liver and intestine (Tolson & Wang 2010, Ihunnah et al. 2011). Though this activity is clearly beneficial, it can also enhance drug toxicity, such as during the PXR-mediated accumulation of toxic aspirin metabolites (Guo et al. 2004). Similarly, CAR deficiency is protective against hepatotoxicity caused by the breakdown of phenobarbital and related compounds (Wei et al. 2000). Roles of PXR and CAR in endobiotic metabolism have also come to light. PXR can promote BA excretion from the liver into the urine, which may explain the profound susceptibility of PXR-deficient mice to challenge with a high cholesterol-BA diet (Staudinger et al. 2001, Xie et al. 2001, Sonoda et al. 2005). Ligand-activated PXR can also suppress hepatic gluconeogenesis and fasting-induced FAO and has been implicated in the regulation of reverse cholesterol transport, steroid hormone synthesis, and the catabolism of androgen, bilirubins, and retinoic acid (Ihunnah et al. 2011). Ligand-activated CAR protects against diet-induced obesity and its related metabolic complications (Gao et al. 2009).

Ligand activation of both PXR and CAR induces cytoplasmic to nuclear shuttling that can be regulated by phosphorylation (Squires et al. 2004, Pondugula et al. 2009). Once in the nucleus, PXR binds DNA as a heterodimer with RXR, predominantly at DR4 motifs, as revealed by recent genome-wide binding analysis in the liver (Kliewer et al. 1998, Cui et al. 2010). Notably, this study found no evidence for PXR binding to everted repeats (ER6 or ER8), as was previously reported (Kliewer et al. 2002). CAR also binds as an obligate heterodimer with RXR to DR4 motifs, but this preference has yet to be shown on a genome-wide scale (Kawamoto et al. 1999, Sueyoshi & Negishi 2001).

Arrested development: orphans of the NR2A group

Nr2a1/Hnfα and Nr2a2/Hnfγ

The hepatocyte nuclear factors HNF4α and HNF4γ are encoded by two distinct but highly homologous genes (Drewes et al. 1996). Cloning of HNF4α from human liver revealed several variants regulated by alternative promoter usage and splicing (Chartier et al. 1994, Kritis et al. 1996). There are two predominant isoforms: the longer HNF4α1 containing an additional N-terminal AF1 domain expressed highly in liver, and the shorter HNF4α7 expressed highly in the pancreas (Eeckhoute et al. 2003). HNF4γ was cloned from a human kidney cDNA library and an early report found it in the pancreas, liver, brain, and lung (Plengvidhya et al. 1999). HNF4α appears to function as a ligand-independent, constitutively active receptor (Ruse et al. 2002). However, several crystal structures of purified LBDs identified fatty acids in its binding pocket and there is evidence that linoleic acid binds to HNF4α in the livers of fed, but not fasted, mice (Dhe-Paganon et al. 2002, Wisely et al. 2002, Yuan et al. 2009).

Mutations in HNF4α are associated with a rare form of early-onset, autosomal dominant diabetes called maturity onset diabetes of the young, and therefore HNFα is considered a MODY1 gene (Yamagata et al. 1996, Navas et al. 1999). Mouse models have shown that it is required in the pancreas for glucose-stimulated insulin secretion and beta cell expansion (Gupta et al. 2005, 2007). HNF4α has pleiotropic roles in other enterohepatic tissues, most prominently in the liver, where it is required for the maturation, maintenance, and differentiated functions of hepatocytes (Sladek et al. 1990, Chen et al. 1994b). Disruption of this program by hepatocyte-specific ablation of HNF4α results in hepatomegaly, fatty liver, reduced serum cholesterol and TG, increased serum BAs, and premature death (Hayhurst et al. 2001). In intestinal epithelial cells, HNF4α regulates fatty acid re-absorption and barrier function (Cattin et al. 2009, Frochot et al. 2012). Importantly, HNF4α is required for endoderm formation during development and at multiple stages of liver maturation (Duncan 2003, Parviz et al. 2003). Less is known about the physiological roles of HNF4γ, but it appears to be dispensable for embryogenesis. Rather, it promotes normal energy expenditure and locomoter activity (Gerdin et al. 2006).

HNF4α can bind as a homodimer or as a heterodimer with HNF4γ to DR1 recognition motifs (Daigo et al. 2011, Fang et al. 2012). Although many other NRs bind the DR1, one specific half-site motif (CAAAGTCCA) is preferred by HNF4α in vitro and in vivo, suggesting a mechanism underlying its distinct target gene networks and functions (Odom et al. 2004, Gupta et al. 2005, Fang et al. 2012).

Partners for life: orphans of the NR2B group

Nr2b1/Rxrα, Nr2b2/Rxrβ, and Nr2b3/Rxrγ

The Nr2b family includes retinoid X receptors, RXRα, RXRβ, and RXRγ. Cloning of RXRα as a retinoic acid-responsive factor that shared modest homology with retinoic acid receptors (RARs) was followed shortly by identification of RXRβ and RXRγ (Mangelsdorf et al. 1990, Rowe et al. 1991, Yu et al. 1991). RXRs dimerize with and strengthen the DNA-binding and transcriptional activity of other NRs, a list that now includes TR, RARs, VDR, PPARs, LXRs, FXR, PXR, CAR, NGFIB, and NURR1 (Yu et al. 1991, Kliewer et al. 1992a, Leid et al. 1992, Marks et al. 1992, Mark & Chambon 2003). RXRs bind 9-cis-retinoic acid with high affinity, and their heterodimer partners can be defined as ‘permissive’ or ‘non-permissive’ based on whether an RXR ligand activates the complex (Levin et al. 1992, Mangelsdorf et al. 1992, Lefebvre et al. 2010). The production of 9-cis-RA in vivo is controversial, but several specific and high affinity pharmacological ligands have been developed (Wolf 2006, Pérez et al. 2012).

Because of their wide array of binding partners, RXRs may have the most disparate biological roles of all NRs. For instance, RXRα ablation is embryonic lethal due to cardiac defects likely to result from its participation in complexes with RAR (Kastner et al. 1994, Sucov et al. 1994, Gruber et al. 1996). In the liver, where RXRα is the most abundant subtype, hepatocyte-specific deletion results in elevated serum triglycerides, serum cholesterol, and dramatic intolerance to a high cholesterol diet, likely via a loss of LXR, FXR, and possibly PXR and CAR activities (Mangelsdorf et al. 1992, Wan et al. 2000). Ablation of RXRβ is lethal in some genetic backgrounds and it has a non-redundant role in spermatogenesis (Kastner et al. 1996). RXRγ is not essential for development but appears to be important for proper sensitivity to thyroid hormone, as knock-out mice have increased metabolic activity, serum T4 and TSH, and are resistant to diet-induced obesity (Brown et al. 2000, Haugen et al. 2004). It is also required in the CNS for proper functioning of cholinergic and dopaminergic pathways and for oligodendrocyte differentiation (Saga et al. 1999, Krzyzosiak et al. 2010, Huang et al. 2011). Compound mutants in RXR family members have been generated and have been comprehensively reviewed elsewhere (Mark et al. 2006).

Recent ChIP-seq studies have profiled the binding specificities of RXRs on a genome-wide scale, revealing extensive co-occupancy of RXRs with PPARγ in adipocytes and LXR in liver. These studies also reported substantial numbers of RXR-only binding sites, suggesting the likely cooperation of RXR with additional NR partners in these tissues (Nielsen et al. 2008, Boergesen et al. 2012). A similar study found RXRα bound near ∼80% of genes expressed in the liver, though expression of only a small fraction were affected by hepatocyte-specific RXRα knockout, illustrating the challenge of elucidating the role of RXRs in tissue-specific regulatory networks (Zhan et al. 2012).

What's in a name: orphans of the NR2C group

Nr2c1/Tr2 and Nr2c2/Tr4

Testicular orphan NR 2 (Tr2) was originally identified by screening a human testis cDNA library for novel genes containing sequences similar to previously identified NR DBDs and the identification of related receptor Tr4 (Tak1) was reported a few years later (Chang et al. 1989, 1994, Hirose et al. 1994, Law et al. 1994, Lee et al. 2002). Tr2 and Tr4 display a widespread expression pattern in embryonic and adult tissues, suggesting pleiotropic physiological functions (Lee et al. 2002). Indeed, loss of function studies implicated TR4 in many diverse biological systems, including the CNS, reproduction, and metabolism (Kim et al. 2003, Chen et al. 2005b, Kang et al. 2011, Lin et al. 2012). Interestingly, mice lacking TR2 appeared normal suggesting that the Nr2c group may function redundantly and this is supported by the dramatic defect in stem cell self-renewal, commitment, and differentiation observed in mice lacking both orphan NRs (Shyr et al. 2002a, 2009).

TR2 and 4 can bind to response elements consisting of a DR of the canonical NR half site (AGGTCA) with various types of spacing (DR1–5) and, consistent with this relatively promiscuous binding, part of the group's functions seems to be mediated by sharing or competing with other NRs for response elements (Lin et al. 1995, Yan et al. 1998, Xie et al. 2009a). Direct regulation of transcription appears to be dependent on homo- or heterodimerization between the Nr2c group members (Lee et al. 1998, Zhou et al. 2011). In contrast, the analysis of TR4-enriched genomic regions obtained with ChIP-seq revealed no obvious DR element, suggesting that the genomic localization of NR2C members may be indirect (O'Geen et al. 2010). Consistent with this, several studies have reported interaction with and regulation of other NRs independent of NR2C binding to DNA (Hu et al. 2002, Shyr et al. 2002b, Mu & Chang 2003).

To date, ligands for Nr2c group members have not been described, although there is some evidence that they can be bound and activated by polyunsaturated fatty acids (Xie et al. 2009a). A recent crystal structure showed ligand-free TR4 to be in an autorepressed conformation that can be modulated by retinoid treatment introducing another potential ligand of the Nr2c group (Zhou et al. 2011). Receptor phosphorylation, acetylation, and sumoylation status can also cause the exchange of corepressors and coactivators and thereby convert NR2C receptors from transcriptional repressors to activators and vice versa (Khan et al. 2005, Gupta et al. 2009, Xie et al. 2011). This is exemplified by the regulation of the Oct4 promoter in stem cells by TR2, where phosphorylation of the receptor causes repression of Oct4, a critical switch in the balance of pluriopotent cell self-renewal and differentiation (Gupta et al. 2008). In addition to posttranslational modification, the expression level of TR2/4 appears to be an important means of activity modulation and showing therapeutic promise, genetic overexpression of Tr2/Tr4 in the erythroid lineage of a sickle cell disease model conferred enhanced expression of target gene fetal hemoglobin and alleviated disease symptoms (Campbell et al. 2011).

Seeing is believing: orphans of the NR2E group

Nr2e1/Tlx and Nr2e3/Pnr

Tlx is the vertebrate homologue of the Drosophila gene tailess (tll), which was characterized in 1990 (Gui et al. 2011). A screening for similar genes led to the photoreceptor NR (PNR), making it the latest vertebrate NR to be described (Chen et al. 1999, Kobayashi et al. 1999). TLX and PNR negatively regulate target gene transcription as monomers or homodimers, recruiting corepressors to the half sites or DR1 elements although there have been reports of gene activation as well (Chen et al. 2005a, Zhang et al. 2006, Sun et al. 2007, Yokoyama et al. 2008, Qu et al. 2010). Furthermore, a recent study has reported the ability of PNR to interact with and positively regulate p53 in a manner independent of PNR DNA-binding (Wen et al. 2012). While very little is known about the ligand dependency of the Nr2e group, activity of PNR and TLX is regulated by interaction with other NRs, posttranslational modification, alternative splicing, and miRNA targeting (Cheng et al. 2004, Wolkenberg et al. 2006, Onishi et al. 2009, Zhao et al. 2009, 2010, Shibata et al. 2011, Qin et al. 2013b).

In contrast to most orphan NRs, the Nr2e members have very specialized physiological roles limited mainly to the development of the rods and cones of the retina and CNS function (Gui et al. 2011, Forrest & Swaroop 2012). The essential role for PNR in retinal development is reflected in its mutation being highly associated with enhanced S-cone syndrome, retinitis pigmentosa, and other retinopathy in humans (Forrest & Swaroop 2012). Retinal degeneration also occurs in mice with either a spontaneous or targeted deletion (Forrest & Swaroop 2012). TLX also functions in preventing retinal degeneration and in addition, mouse models have demonstrated a critical role in the development of the limbic system, where deletion leads to extreme aggression (Monaghan et al. 1997, Yu et al. 2000, Young et al. 2002). More recently, the essential role of TLX in maintaining adult neural stem cells in an undifferentiated, proliferative state has come to light and this cellular function has been reported to impact both spatial learning and brain tumor expansion, suggesting its potential as a therapeutic target (Shi et al. 2004, Liu et al. 2010, Zou et al. 2012).

Leaders of the chicken dance: orphans of the NR2F group

Nr2f1/Coup-tfI, Nr2f2/Coup-tfII, and Nr2f6/Ear2

Chicken ovalbumin upstream promoting TF 1 (COUP-TFI) was identified as a long-sought after regulator of the chicken ovalubin gene (Wang et al. 1989). It, as well as ERBA-related protein 2 (EAR2), was originally described as the genes encoding proteins with homology to thyroid receptor, while COUP-TFII, a Nr2f member highly related to COUP-TFI, was identified in a later screen (Miyajima et al. 1988, Ritchie et al. 1990, Ladias & Karathanasis 1991).

COUP-TFI and II bind as dimers to repress transcription via DR1 elements (Tsai & Tsai 1997, Alfano et al. 2013). The COUP-TFs can also promiscuously recognize many other direct, inverted and everted NR half-site (AGGTCA) repeats, enabling them to compete with and antagonize the action of other NRs (Tsai & Tsai 1997, Alfano et al. 2013). These receptors also have transpressive effects mediated by heterodimerization with other TFs or their partner RXR. EAR2 also appears to be a transcriptional repressor able to heterodimerize with other NRs including the COUP-TFs (Jonk et al. 1994, Zhu et al. 2000, Warnecke et al. 2005). For example, EAR2 was recently demonstrated to bind and inhibit the function of RORγt in TH17 lympocytes (Hermann-Kleiter et al. 2012). Despite the large body of evidence describing this family as transcriptional repressors, there have been some reports of NR2F-mediated gene activation but in vivo relevance of these findings has not been substantiated (Tsai & Tsai 1997). To date, the Nr2f members remain as true orphan NRs, but in addition to their level of gene expression, activity of these TFs is regulated by posttranslational modifications including phosphorylation (Tsai & Tsai 1997, Hermann-Kleiter et al. 2008).

The Nr2f group is widely expressed and loss of function models has been a key in the dissection of their many physiological roles (Lin et al. 2011). By influencing cellular processes including, survival, migration, fate determination, and differentiation, COUP-TFI and II have unique and essential functions in neural development and organogenesis respectively (Lin et al. 2011). However, they do share nearly identical DBDs and LBDs and recent analyses of mice deficient in both receptors have uncovered a redundant role in retinal development (Satoh et al. 2009, Tang et al. 2010). In addition to loss of function studies, genome-wide profiling of histone modifications and analyses of enhancer sequences predicted a role for both COUP-TFI and II in the phenotype of embryonic neural crest cells (NCC) which was further supported by knockdown experiments in human NCCs (Rada-Iglesias et al. 2012). Besides direct developmental roles, the NR2F receptors have recently been implicated as potential targets in cancer therapeutic intervention (Litchfield & Klinge 2012). For example, overexpression of COUP-TFII results in the inhibition of SMAD4-mediated transactivation, which normally prevents cancer progression in prostate epithelium (Qin et al. 2013a). In contrast to COUP-TFI and II, much less is known about EAR2; however, knock-out mouse studies have suggested the roles in the CNS and immune system (Warnecke et al. 2005, Hermann-Kleiter et al. 2008).

Lots of energy: orphans of the NR3B group

Nr3b1/Errα, Nr3b2/Errβ, and Nr3b3/Errγ

Estrogen-related receptors, α (ERRα) and β (ERRβ) were discovered by screening libraries with a probe consisting of the sequence of the DBD of estrogen receptor (ER) and were the first orphan NRs to be discovered (Giguère et al. 1988, Deblois & Giguère 2011). Nearly a decade later, ERRγ was discovered as a gene deleted in a critical region of Usher syndrome, a genetic disorder resulting in hearing and vision loss, and subsequently described by two additional groups (Eudy et al. 1998, Hong et al. 1999, Heard et al. 2000). The ERRs are constitutive activators and crystal structure studies suggest that classic ligand binding modulation is unlikely due to an inaccessible LBP. However, the presence of certain inverse agonists are able to induce a conformational change favoring corepressor interaction (Kallen et al. 2007, Xie et al. 2009b). Although no endogenous ligand for the ERRs has been reported, many inverse agonists and antagonists have been described, including pesticides, synthetic and phyto-estrogens, and 4-hydroxytamoxifen (4-OHT), underscoring the notion of functional crosstalk between ERR and ER signaling (Coward et al. 2001, Tremblay et al. 2001a,b, Bonnelye & Aubin 2013). Adding to this complexity, bisphenol A, the synthetic estrogen and endocrine disruptor found in many plastics, binds ERRγ and prevents 4-OHT-induced coactivator dissociation (Takayanagi et al. 2006). Posttranslational modifications including phosphorylation, acetylation, and sumoylation also influence ERR activity (Ariazi et al. 2007, Tremblay et al. 2008, Wilson et al. 2010). The constitutive activity of the ERR group is also negatively regulated by DAX1 and SHP (Sanyal et al. 2002, Uranishi et al. 2013). The ERRs all recognize an extended half-site termed the ERRE (TNAAGGTCA) as monomers, homodimers, or heterodimers (Dufour et al. 2007, Tremblay & Giguère 2007). Although ERRs may also have affinity for classic estrogen and thyroid receptor elements in vitro, recent genome wide localization studies have suggested that the ERRE is the main site of ERR occupancy (Johnston et al. 1997, Vanacker et al. 1998, Dufour et al. 2007, Tremblay & Giguère 2007).

ERRα and ERRγ are widely expressed and regulate genes that modulate cellular energy metabolism by directing mitochondrial biogenesis and function impacting a variety of tissues under various physiological stresses including bone, adipose, heart, immune cells, kidney, and liver, implicating them as important therapeutic targets in a variety of metabolic disorders (Tremblay & Giguère 2007, Giguère 2008, Deblois & Giguère 2011). In addition, ERRs have the potential to modulate cancer cell energy production and, recently, ERRγ has been identified as a miRNA target in breast cancer cells, leading to a shift from oxidative to glycolytic metabolism (Eichner et al. 2010, Deblois & Giguère 2013). ERRβ has emerged as a component of cell pluripotency, being a key target of both NANOG and GSK/TCF3 pathways that modulates the expression of genes important in self-renewal (Ivanova et al. 2006, Chen et al. 2008, Festuccia et al. 2012, Martello et al. 2012). ERRβ also functions during development of the placenta, retina, and endolymph of the inner ear and, and mutations have been linked to hearing loss in humans (Chen & Nathans 2007, Collin et al. 2008, Onishi et al. 2010, Ben Saïd et al. 2011, Lee et al. 2011b).

First to the party: orphans of the NR4A group

Nr4a1/Nur77, Nr4a2/Nurr1, and Nr4a3/Nor1

The Nr4a group consists of three related genes with many names reflecting discovery by many different groups: Nur77 (Ngfi-b, Tr3, N10, Nak-1, St-59), Nurr1 (Rnr-1, Not, Tinur) and Nor1 (Tec, Minor, Chn) (Hazel et al. 1988, Milbrandt 1988, Chang et al. 1989, Ryseck et al. 1989, Nakai et al. 1990, Law et al. 1992, Scearce et al. 1993, Mages et al. 1994, Ohkura et al. 1994, Hedvat & Irving 1995, Labelle et al. 1995, Clark et al. 1996, Chen & Nathans 2007, Collin et al. 2008, Onishi et al. 2010, Ben Saïd et al. 2011, Lee et al. 2011b). Depending on cellular and signal context, these receptors can function uniquely or redundantly as developmental modulators or immediate early genes whose expression is rapidly induced by a variety of stimuli to control cellular proliferation, function, or death (Li et al. 2006, Maxwell & Muscat 2006). Key roles for the NR4A receptors have been demonstrated in many cell types including neuronal, immune, hematopoietic, cancer, endocrine, and metabolic (Li et al. 2006, Maxwell & Muscat 2006, Pei et al. 2006, Pearen & Muscat 2010, Zhao & Bruemmer 2010, Luo 2012, Mohan et al. 2012, van Tiel & de Vries 2012).

Nr4a orphans harbor a unique LBD filled with bulky hydrophobic residues that is unlikely to accommodate a typical ligand (Baker et al. 2003, Wang et al. 2003). Furthermore, nonclassical coactivator interfaces have been described for these receptors (Wansa et al. 2002, Codina et al. 2004). Therefore, regulation of the NR4A receptor activity depends on gene expression, alternative splicing, posttranslational modification, interaction with other NRs, microRNA targeting, and cellular localization (Maruyama et al. 1998, Maxwell & Muscat 2006, Malewicz et al. 2011, McMorrow & Murphy 2011, Yang et al. 2012, Li et al. 2013). It should be noted that despite the unique structure of the LBD, pharmacological agonists have been described including 1,1-bis(3-indolyl)-1-(p-anisyl)methane, anti-neoplastic and anti-inflammatory agent 6-mercaptopurine, and cytosporone B (Wansa et al. 2003, Chintharlapalli et al. 2005, Zhan et al. 2008).

NURR1, NUR77, and NOR1 can bind as monomers to half-site recognition motifs called the NBRE (AAAGGTCA), but they can also homodimerize or heterodimerize with each other and with RXR and bind NurRE, a DR element (Maruyama et al. 1998, Maxwell & Muscat 2006). The importance of the transactivation function of the NR4A receptors has been demonstrated in vivo. For example, Nor1 is a gene component within chromosomal translocations that occur frequently in human extraskeletal myxoid sarcoma (Labelle et al. 1995, Clark et al. 1996). The oncogenic fusion protein product of this translocation is frequently a strong activation domain of another TF such as EWS linked to a full length NOR1 leading to misregulation of NOR1 gene targets (Filion & Labelle 2012). Critical gene regulation by NR4A receptors is also exemplified by the direct regulation of tyrosine hydroxylase in neurons by NURR1 to maintain the dopaminergic phenotype which is hampered in Parkinson's disease and by obligatory direct regulation of Foxp3 by all three NR4A members acting redundantly during the development of regulatory T cells thereby preventing autoimmunity (Sakurada et al. 1999, Luo 2012, Sekiya et al. 2013). Like other NRs, transrepression by the NR4A group has been reported. Specifically, NURR1 in concert with the CoREST complex is able to dock on the p65 subunit of NFκB and drive its clearance from inflammatory target genes in astrocytes and microglia, providing an additional mechanism for the protective function of NURR1 in Parkinson's disease (Saijo et al. 2009). Finally, NUR77 and NOR1 play essential roles in apoptosis in many cells and there is mounting evidence that this is accomplished in a non-genomic manner by converting BCL-2 proteins into pro-apoptotic molecules at the mitochondria (Li et al. 2006, Mohan et al. 2012). Underscoring the potential impact of this finding, small peptide mimics of NUR77 have been designed successfully to induce cancer cell death (Kolluri et al. 2008).

On steroids: orphans of the NR5A group

Nr5a1/Sf1 and Nr5a2/Lrh1

Steroidogenic factor 1 (SF1) was discovered by Lala et al. (1992) as a major regulator of steroidogenic enzyme gene expression. Liver receptor homolog 1 (LRH-1) was cloned by several groups as a regulator of heptatitis virus and albumin gene expression (Tsukiyama et al. 1992, Becker-André et al. 1993, Galarneau et al. 1996, Li et al. 1998, Nitta et al. 1999). SF1 and LRH1 have large, hydrophobic LBPs (Schimmer & White 2010). Initial studies reported stable active state conformations of both receptor LBDs without addition of an exogenous ligand (Desclozeaux et al. 2002, Sablin et al. 2003). However, in 2005, three groups discovered phospholipid species in the LBPs of SF1 and LRH1 purified from bacteria (Krylova et al. 2005, Li et al. 2005, Ortlund et al. 2005). Intriguingly, an unusual phosphatidylcholine species was identified as an LRH1 ligand that, when administered to WT, but not Lrh1−/− mice, improved their glucose homeostasis and ameliorated hepatic steatosis during high fat diet feeding (Lee et al. 2011a).

SF1 and LRH1 are transcriptional activators that bind DNA as monomers recognizing consensus of AGGTCA sequences. An apparent preference for YCA (where Y is a pyrimidine) 5′ to the hexamer may be due to a C-terminal extension (CTE) of the DBD shared with other monomeric DNA-binding NRs (Wilson et al. 1993, Solomon et al. 2005). SF1 and LRH1 also contain an additional 20 amino acid extension after the CTE called the Ftz-f1 motif (after Drosophila Ftz-f1) that is unique to NR5A family members (Ingraham & Redinbo 2005). Crystal structures revealed that this motif does not contact DNA but rather affects interactions with co-activating proteins (Solomon et al. 2005). Recent genome-wide profiling of SF1 binding in adrenocortical cells found the NR half site as its preferential motif, but did not find enrichment of the predicted 5′ pyrimidine, suggesting that it may not be required for SF1 binding to DNA in chromatin (Doghman et al. 2013).

SF1 is required for the differentiation of steroidogenic tissues, with homozygous null mice dying shortly after birth due to corticosteroid insufficiency (Luo et al. 1994, Sadovsky et al. 1995). SF1 function is also required for cell autonomously in pituitary gonadotropes, developing gonads and postnatal ovarian granulosa cells (Schimmer & White 2010). These mouse models of sexual maturation defects are important to human disease, as shown by the reproductive dysfunction in patients with naturally occurring SF1 mutations (El-Khairi & Achermann 2012, Lalli et al. 2013). LRH1 is involved in the regulation of steroid, BA and cholesterol homeostasis, processes that are consistent with its restricted expression in the pancreas, liver, intestines, and ovaries (Fernandez-Marcos et al. 2011). Hepatocyte-specific deletion of LRH1 changes the composition of the BA pool, resulting in decreased intestinal lipid absorption and BA recycling (Mataki et al. 2007, Lee et al. 2008). LRH1 has also been implicated in regulating glucose metabolism in the liver and proliferation in the intestine and pancreas (Schoonjans et al. 2005, Oosterveer et al. 2012). Intriguingly, GWAS studies have found a link between LRH1 SNPs and pancreatic cancer susceptibility (Petersen et al. 2010).

The family jewel: orphans of the NR6A group

Nr6a1/Gcnf

Germ cell nuclear factor (Gcnf, Rtr, Ncnf) is unique enough within the NR superfamily to be the only member of the NR6 subfamily (Hummelke & Cooney 2001, Germain et al. 2006). Originally named because of its robust and restricted expression in the germ cells, GCNF lacks a classic AF2 domain and acts as a transcriptional repressor through ligand-independent interactions with corepressor complexes (Chen et al. 1994a, Hirose et al. 1995, Bauer et al. 1997, Hummelke & Cooney 2001, Mullen et al. 2007). To date, a ligand for GCNF has not been identified, and its activity appears to be controlled by regulation of its gene expression (Heinzer et al. 1998, Gurtan et al. 2013, Krill et al. 2013, Wang et al. 2013). While recombinant GCNF forms homodimers upon binding to a DR of the classic NR half-site (AGGTCA) with no additional central nucleotides (DR0), endogenous DR0-bound GCNF exists as an oligomer in a DNA-dependent manner unlike that of typical NR dimerization (Gu et al. 2005b).

One of the direct targets of GCNF is Oct4, a core TF in the maintenance of embryonic stem cell pluriopotency (Fuhrmann et al. 2001). Indeed, loss of function studies in mice has demonstrated that the GCNF-mediated repression of Oct4 is essential for stem cell differentiation during embryogenesis (Chung et al. 2001, Gu et al. 2005a). The regulation of Oct4 and other pluriopotency factors makes GCNF an attractive target for therapeutic manipulation and biomarking in stem cells (Mullen et al. 2007, Akamatsu et al. 2009, Wang et al. 2013).

Summary and future perspectives

Advances in chemistry and molecular biology have led to an enormous accumulation of knowledge about the number, regulation, and function of orphan NRs over the past 25 years since their initial discovery. This review has attempted to provide a high-level overview of this progress, focusing on how each orphan was discovered, its regulation by ligand, and its regulation of gene expression in different tissues in health and disease. Much more is known about each orphan NR, and readers are encouraged to seek out additional information from the printed and virtual scientific literature.

Although the family is much grown up, current rapid progress in high throughput nucleotide sequencing, proteomics, metabolomics, and computational biology promises to lead to a more complete and integrated understanding of the orphan NRs and how they are regulated in different tissues, and what they regulate at many developmental stages, and in disease states. This approach is presently somewhat limited by the lack of adequate antibodies and data analysis pipelines, and by the added complexity of many orphan NRs playing critical roles in cell types that are minority components of heterogenous tissues. Technical advances in these areas will be critical for the discovery of novel therapeutic targets as well as strategies that avoid off target effects while maintaining beneficial actions.

Possibly the largest question still unanswered concerns the identification of the most relevant endogenous ligands for most, if not all, of the orphan NRs. Beginning shortly after the initial discovery of orphan NRs, the issue of whether they would all have endogenous ligands was hotly debated (‘Do Orphan Receptors Have Ligands?’, see www.sarahgreene.net/previous/hmsbeagle/html/content/03/cutedge/overview.htm) and this question still remains. While endogenous and synthetic ligands have been discovered for many of the orphan NRs, others seem likely to be bona fide ligand-independent TFs, yet this is almost impossible to prove (Schupp & Lazar 2010). Further, in most cases, there is not a consensus about the physiological role of putative endogenous ligands. This is in clear contrast with NR superfamily members that are receptors for hormones, where the hormones were discovered before the ligands and, in most cases, are produced by discrete endocrine organs such as the thyroid and adrenals, whose function is largely based on secretion of their cognate hormones.

Nevertheless, the identification of biological functions and ligands that activate the orphan NRs has yielded remarkable insight into dozens of diverse physiological processes, from embryonic stem cell self renewal and differentiation to inflammation, circadian rhythm, and metabolism. This has led to a more complete understanding of the mechanism of action for certain drug classes, the identification of targets mediating adverse effects of environmental pollutants, better strategies for the reprograming of pluripotent cells, and development of novel small molecules for the treatment of human disease. Further dissection of orphan NR networks, including their mechanisms of action and the genes and cellular processes that they regulate, continues to have great potential to elucidate the molecular pathology of diseases as well as the underlying physiology, leading to safer, more specific, and more effective therapeutic strategies that are likely to be evident at the next orphan NR reunion.

Declaration of interest

The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the review.

Funding

Work on orphan NRs in the Lazar lab is supported by NIH DK45586 and DK49780 and the Cox Institute for Medical Research.

Acknowledgements

This review encompasses an enormous area and the authors apologize that they were not able to cite all important papers on orphan NRs.

References

  • Ahmadian M, Suh JM, Hah N, Liddle C, Atkins AR, Downes M & Evans RM 2013 PPARγ signaling and metabolism: the good, the bad and the future. Nature Medicine 19 557566. (doi:10.1038/nm.3159).

    • Search Google Scholar
    • Export Citation
  • Akamatsu W, DeVeale B, Okano H, Cooney AJ & van der Kooy D 2009 Suppression of Oct4 by germ cell nuclear factor restricts pluripotency and promotes neural stem cell development in the early neural lineage. Journal of Neuroscience 29 21132124. (doi:10.1523/JNEUROSCI.4527-08.2009).

    • Search Google Scholar
    • Export Citation
  • Alfano C, Magrinelli E, Harb K & Studer M The nuclear receptors COUP-TF: a long-lasting experience in forebrain assembly Cellular and Molecular Life Sciences 2013 [in press] doi:10.1007/s00018-013-1320-6).

    • Search Google Scholar
    • Export Citation
  • Aranda A & Pascual A 2001 Nuclear hormone receptors and gene expression. Physiological Reviews 81 12691304.

  • Ariazi EA, Kraus RJ, Farrell ML, Jordan VC & Mertz JE 2007 Estrogen-related receptor α1 transcriptional activities are regulated in part via the ErbB2/HER2 signaling pathway. Molecular Cancer Research 5 7185. (doi:10.1158/1541-7786.MCR-06-0227).

    • Search Google Scholar
    • Export Citation
  • Baes M, Gulick T, Choi HS, Martinoli MG, Simha D & Moore DD 1994 A new orphan member of the nuclear hormone receptor superfamily that interacts with a subset of retinoic acid response elements. Molecular and Cellular Biology 14 15441552.

    • Search Google Scholar
    • Export Citation
  • Baker KD, Shewchuk LM, Kozlova T, Makishima M, Hassell A, Wisely B, Caravella JA, Lambert MH, Reinking JL & Krause H et al. 2003 The Drosophila orphan nuclear receptor DHR38 mediates an atypical ecdysteroid signaling pathway. Cell 113 731742. (doi:10.1016/S0092-8674(03)00420-3).

    • Search Google Scholar
    • Export Citation
  • Barak Y, Liao D, He W, Ong ES, Nelson MC, Olefsky JM, Boland R & Evans RM 2002 Effects of peroxisome proliferator-activated receptor δ on placentation, adiposity, and colorectal cancer. PNAS 99 303308. (doi:10.1073/pnas.012610299).

    • Search Google Scholar
    • Export Citation
  • Bauer UM, Schneider-Hirsch S, Reinhardt S, Pauly T, Maus A, Wang F, Heiermann R, Rentrop M & Maelicke A 1997 Neuronal cell nuclear factor – a nuclear receptor possibly involved in the control of neurogenesis and neuronal differentiation. European Journal of Biochemistry/FEBS 249 826837. (doi:10.1111/j.1432-1033.1997.t01-1-00826.x).

    • Search Google Scholar
    • Export Citation
  • Båvner A, Sanyal S, Gustafsson J-Å & Treuter E 2005 Transcriptional corepression by SHP: molecular mechanisms and physiological consequences. Trends in Endocrinology and Metabolism 16 478488. (doi:10.1016/j.tem.2005.10.005).

    • Search Google Scholar
    • Export Citation
  • Becker-André M, André E & DeLamarter JF 1993 Identification of nuclear receptor mRNAs by RT-PCR amplification of conserved zinc-finger motif sequences. Biochemical and Biophysical Research Communications 194 13711379. (doi:10.1006/bbrc.1993.1976).

    • Search Google Scholar
    • Export Citation
  • Benoit G, Malewicz M & Perlmann T 2004 Digging deep into the pockets of orphan nuclear receptors: insights from structural studies. Trends in Cell Biology 14 369376. (doi:10.1016/j.tcb.2004.05.007).

    • Search Google Scholar
    • Export Citation
  • Benoit G, Cooney A, Giguere V, Ingraham H, Lazar M, Muscat G, Perlmann T, Renaud J-P, Schwabe J & Sladek F et al. 2006 International Union of Pharmacology. LXVI. Orphan nuclear receptors. Pharmacological Reviews 58 798836. (doi:10.1124/pr.58.4.10).

    • Search Google Scholar
    • Export Citation
  • Ben Saïd M, Ayedi L, Mnejja M, Hakim B, Khalfallah A, Charfeddine I, Khifagi C, Turki K, Ayadi H & Benzina Z et al. 2011 A novel missense mutation in the ESRRB gene causes DFNB35 hearing loss in a Tunisian family. European Journal of Medical Genetics 54 e535e541. (doi:10.1016/j.ejmg.2011.06.008).

    • Search Google Scholar
    • Export Citation
  • Bensinger SJ & Tontonoz P 2008 Integration of metabolism and inflammation by lipid-activated nuclear receptors. Nature 454 470477. (doi:10.1038/nature07202).

    • Search Google Scholar
    • Export Citation
  • Bertilsson G, Heidrich J, Svensson K, Asman M, Jendeberg L, Sydow-Bäckman M, Ohlsson R, Postlind H, Blomquist P & Berkenstam A 1998 Identification of a human nuclear receptor defines a new signaling pathway for CYP3A induction. PNAS 95 1220812213. (doi:10.1073/pnas.95.21.12208).

    • Search Google Scholar
    • Export Citation
  • Blumberg B, Sabbagh W Jr, Juguilon H, Bolado J Jr, van Meter CM, Ong ES & Evans RM 1998 SXR, a novel steroid and xenobiotic-sensing nuclear receptor. Genes and Development 12 31953205. (doi:10.1101/gad.12.20.3195).

    • Search Google Scholar
    • Export Citation
  • Boergesen M, Pedersen , Gross B, van Heeringen SJ, Hagenbeek D, Bindesbøll C, Caron S, Lalloyer F, Steffensen KR & Nebb HI et al. 2012 Genome-wide profiling of liver X receptor, retinoid X receptor, and peroxisome proliferator-activated receptor α in mouse liver reveals extensive sharing of binding sites. Molecular and Cellular Biology 32 852867. (doi:10.1128/MCB.06175-11).

    • Search Google Scholar
    • Export Citation
  • Bonnelye E & Aubin JE 2013 An energetic orphan in an endocrine tissue: a revised perspective of the function of estrogen receptor-related receptor α in bone and cartilage. Journal of Bone and Mineral Research 28 225233. (doi:10.1002/jbmr.1836).

    • Search Google Scholar
    • Export Citation
  • Bradley MN, Hong C, Chen M, Joseph SB, Wilpitz DC, Wang X, Lusis AJ, Collins A, Hseuh WA & Collins JL et al. 2007 Ligand activation of LXRβ reverses atherosclerosis and cellular cholesterol overload in mice lacking LXRα and apoE. Journal of Clinical Investigation 117 23372346. (doi:10.1172/JCI31909).

    • Search Google Scholar
    • Export Citation
  • Brown NS, Smart A, Sharma V, Brinkmeier ML, Greenlee L, Camper SA, Jensen DR, Eckel RH, Krezel W & Chambon P et al. 2000 Thyroid hormone resistance and increased metabolic rate in the RXR-γ-deficient mouse. Journal of Clinical Investigation 106 7379. (doi:10.1172/JCI9422).

    • Search Google Scholar
    • Export Citation
  • Bugge A, Feng D, Everett LJ, Briggs ER, Mullican SE, Wang F, Jager J & Lazar MA 2012 Rev-erbα and Rev-erbβ coordinately protect the circadian clock and normal metabolic function. Genes and Development 26 657667. (doi:10.1101/gad.186858.112).

    • Search Google Scholar
    • Export Citation
  • Burkart EM, Sambandam N, Han X, Gross RW, Courtois M, Gierasch CM, Shoghi K, Welch MJ & Kelly DP 2007 Nuclear receptors PPARβ/δ and PPARα direct distinct metabolic regulatory programs in the mouse heart. Journal of Clinical Investigation 117 39303939. (doi:10.1172/JCI32578).

    • Search Google Scholar
    • Export Citation
  • Burris TP 2008 Nuclear hormone receptors for heme: REV-ERBα and REV-ERBβ are ligand-regulated components of the mammalian clock. Molecular Endocrinology 22 15091520. (doi:10.1210/me.2007-0519).

    • Search Google Scholar
    • Export Citation
  • Calkin AC & Tontonoz P 2012 Transcriptional integration of metabolism by the nuclear sterol-activated receptors LXR and FXR. Nature Reviews. Molecular Cell Biology 13 213224. (doi:10.1038/nrm3312).

    • Search Google Scholar
    • Export Citation
  • Campbell AD, Cui S, Shi L, Urbonya R, Mathias A, Bradley K, Bonsu KO, Douglas RR, Halford B & Schmidt L et al. 2011 Forced TR2/TR4 expression in sickle cell disease mice confers enhanced fetal hemoglobin synthesis and alleviated disease phenotypes. PNAS 108 1880818813. (doi:10.1073/pnas.1104964108).

    • Search Google Scholar
    • Export Citation
  • Cattin A-L, Le Beyec J, Barreau F, Saint-Just S, Houllier A, Gonzalez FJ, Robine S, Pinçon-Raymond M, Cardot P & Lacasa M et al. 2009 Hepatocyte nuclear factor 4α, a key factor for homeostasis, cell architecture, and barrier function of the adult intestinal epithelium. Molecular and Cellular Biology 29 62946308. (doi:10.1128/MCB.00939-09).

    • Search Google Scholar
    • Export Citation
  • Chanda D, Park J-H & Choi H-S 2008 Molecular basis of endocrine regulation by orphan nuclear receptor small heterodimer partner. Endocrine Journal 55 253268. (doi:10.1507/endocrj.K07E-103).

    • Search Google Scholar
    • Export Citation
  • Chandra V, Mahajan S, Saini A, Dkhar HK, Nanduri R, Raj EB, Kumar A & Gupta P 2013 Human IL10 gene repression by Rev-erbα ameliorates Mycobacterium tuberculosis clearance. Journal of Biological Chemistry 288 1069210702. (doi:10.1074/jbc.M113.455915).

    • Search Google Scholar
    • Export Citation
  • Chang C, Kokontis J, Liao SS & Chang Y 1989 Isolation and characterization of human TR3 receptor: a member of steroid receptor superfamily. Journal of Steroid Biochemistry 34 391395. (doi:10.1016/0022-4731(89)90114-3).

    • Search Google Scholar
    • Export Citation
  • Chang C, Da Silva SL, Ideta R, Lee Y, Yeh S & Burbach JP 1994 Human and rat TR4 orphan receptors specify a subclass of the steroid receptor superfamily. PNAS 91 60406044. (doi:10.1073/pnas.91.13.6040).

    • Search Google Scholar
    • Export Citation
  • Chartier FL, Bossu JP, Laudet V, Fruchart JC & Laine B 1994 Cloning and sequencing of cDNAs encoding the human hepatocyte nuclear factor 4 indicate the presence of two isoforms in human liver. Gene 147 269272. (doi:10.1016/0378-1119(94)90079-5).

    • Search Google Scholar
    • Export Citation
  • Chaturvedi P, Pratta M, Steplewski K, Connor J & Kumar S 2006 Functional characterization of an orphan nuclear receptor, Rev-ErbAα, in chondrocytes and its potential role in osteoarthritis. Arthritis and Rheumatism 54 35133522. (doi:10.1002/art.22170).

    • Search Google Scholar
    • Export Citation
  • Chawla A & Lazar MA 1994 Peroxisome proliferator and retinoid signaling pathways co-regulate preadipocyte phenotype and survival. PNAS 91 17861790. (doi:10.1073/pnas.91.5.1786).

    • Search Google Scholar
    • Export Citation
  • Chen J & Nathans J 2007 Estrogen-related receptor β/NR3B2 controls epithelial cell fate and endolymph production by the stria vascularis. Developmental Cell 13 325337. (doi:10.1016/j.devcel.2007.07.011).

    • Search Google Scholar
    • Export Citation
  • Chen F, Cooney AJ, Wang Y, Law SW & O'Malley BW 1994a Cloning of a novel orphan receptor (GCNF) expressed during germ cell development. Molecular Endocrinology 8 14341444. (doi:10.1210/me.8.10.1434).

    • Search Google Scholar
    • Export Citation
  • Chen WS, Manova K, Weinstein DC, Duncan SA, Plump AS, Prezioso VR, Bachvarova RF & Darnell JE 1994b Disruption of the HNF-4 gene, expressed in visceral endoderm, leads to cell death in embryonic ectoderm and impaired gastrulation of mouse embryos. Genes and Development 8 24662477. (doi:10.1101/gad.8.20.2466).

    • Search Google Scholar
    • Export Citation
  • Chen F, Figueroa DJ, Marmorstein AD, Zhang Q, Petrukhin K, Caskey CT & Austin CP 1999 Retina-specific nuclear receptor: a potential regulator of cellular retinaldehyde-binding protein expressed in retinal pigment epithelium and Müller glial cells. PNAS 96 1514915154. (doi:10.1073/pnas.96.26.15149).

    • Search Google Scholar
    • Export Citation
  • Chen J, Rattner A & Nathans J 2005a The rod photoreceptor-specific nuclear receptor Nr2e3 represses transcription of multiple cone-specific genes. Journal of Neuroscience 25 118129. (doi:10.1523/JNEUROSCI.3571-04.2005).

    • Search Google Scholar
    • Export Citation
  • Chen Y-T, Collins LL, Uno H & Chang C 2005b Deficits in motor coordination with aberrant cerebellar development in mice lacking testicular orphan nuclear receptor 4. Molecular and Cellular Biology 25 27222732. (doi:10.1128/MCB.25.7.2722-2732.2005).

    • Search Google Scholar
    • Export Citation
  • Chen X, Xu H, Yuan P, Fang F, Huss M, Vega VB, Wong E, Orlov YL, Zhang W & Jiang J et al. 2008 Integration of external signaling pathways with the core transcriptional network in embryonic stem cells. Cell 133 11061117. (doi:10.1016/j.cell.2008.04.043).

    • Search Google Scholar
    • Export Citation
  • Cheng H, Khanna H, Oh ECT, Hicks D, Mitton KP & Swaroop A 2004 Photoreceptor-specific nuclear receptor NR2E3 functions as a transcriptional activator in rod photoreceptors. Human Molecular Genetics 13 15631575. (doi:10.1093/hmg/ddh173).

    • Search Google Scholar
    • Export Citation
  • Chini CCS, Escande C, Nin V & Chini EN 2013 DBC1 (deleted in breast cancer 1) modulates the stability and function of the nuclear receptor Rev-erbα. Biochemical Journal 451 453461. (doi:10.1042/BJ20121085).

    • Search Google Scholar
    • Export Citation
  • Chintharlapalli S, Burghardt R, Papineni S, Ramaiah S, Yoon K & Safe S 2005 Activation of Nur77 by selected 1,1-bis(3′-indolyl)-1-(p-substituted phenyl)methanes induces apoptosis through nuclear pathways. Journal of Biological Chemistry 280 2490324914. (doi:10.1074/jbc.M500107200).

    • Search Google Scholar
    • Export Citation
  • Cho H, Zhao X, Hatori M, Yu RT, Barish GD, Lam MT, Chong L-W, DiTacchio L, Atkins AR & Glass CK et al. 2012 Regulation of circadian behaviour and metabolism by REV-ERB-α and REV-ERB-β. Nature 485 123127. (doi:10.1038/nature11048).

    • Search Google Scholar
    • Export Citation
  • Chomez P, Neveu I, Mansén A, Kiesler E, Larsson L, Vennström B & Arenas E 2000 Increased cell death and delayed development in the cerebellum of mice lacking the rev-erbA(α) orphan receptor. Development 127 14891498.

    • Search Google Scholar
    • Export Citation
  • Chong HK, Infante AM, Seo Y-K, Jeon T-I, Zhang Y, Edwards PA, Xie X & Osborne TF 2010 Genome-wide interrogation of hepatic FXR reveals an asymmetric IR-1 motif and synergy with LRH-1. Nucleic Acids Research 38 60076017. (doi:10.1093/nar/gkq397).

    • Search Google Scholar
    • Export Citation
  • Chung AC, Katz D, Pereira FA, Jackson KJ, DeMayo FJ, Cooney AJ & O'Malley BW 2001 Loss of orphan receptor germ cell nuclear factor function results in ectopic development of the tail bud and a novel posterior truncation. Molecular and Cellular Biology 21 663677. (doi:10.1128/MCB.21.2.663-677.2001).

    • Search Google Scholar
    • Export Citation
  • Clark J, Benjamin H, Gill S, Sidhar S, Goodwin G, Crew J, Gusterson BA, Shipley J & Cooper CS 1996 Fusion of the EWS gene to CHN, a member of the steroid/thyroid receptor gene superfamily, in a human myxoid chondrosarcoma. Oncogene 12 229235.

    • Search Google Scholar
    • Export Citation
  • Codina A, Benoit G, Gooch JT, Neuhaus D, Perlmann T & Schwabe JWR 2004 Identification of a novel co-regulator interaction surface on the ligand binding domain of Nurr1 using NMR footprinting. Journal of Biological Chemistry 279 5333853345. (doi:10.1074/jbc.M409096200).

    • Search Google Scholar
    • Export Citation
  • Collin RWJ, Kalay E, Tariq M, Peters T, van der Zwaag B, Venselaar H, Oostrik J, Lee K, Ahmed ZM & Caylan R et al. 2008 Mutations of ESRRB encoding estrogen-related receptor β cause autosomal-recessive nonsyndromic hearing impairment DFNB35. American Journal of Human Genetics 82 125138. (doi:10.1016/j.ajhg.2007.09.008).

    • Search Google Scholar
    • Export Citation
  • Coward P, Lee D, Hull MV & Lehmann JM 2001 4-Hydroxytamoxifen binds to and deactivates the estrogen-related receptor γ. PNAS 98 88808884. (doi:10.1073/pnas.151244398).

    • Search Google Scholar
    • Export Citation
  • Cui JY, Gunewardena SS, Rockwell CE & Klaassen CD 2010 ChIPing the cistrome of PXR in mouse liver. Nucleic Acids Research 38 79437963. (doi:10.1093/nar/gkq654).

    • Search Google Scholar
    • Export Citation
  • Daigo K, Kawamura T, Ohta Y, Ohashi R, Katayose S, Tanaka T, Aburatani H, Naito M, Kodama T & Ihara S et al. 2011 Proteomic analysis of native hepatocyte nuclear factor-4α (HNF4α) isoforms, phosphorylation status, and interactive cofactors. Journal of Biological Chemistry 286 674686. (doi:10.1074/jbc.M110.154732).

    • Search Google Scholar
    • Export Citation
  • Deblois G & Giguère V 2011 Functional and physiological genomics of estrogen-related receptors (ERRs) in health and disease. Biochimica et Biophysica Acta 1812 10321040. (doi:10.1016/j.bbadis.2010.12.009).

    • Search Google Scholar
    • Export Citation
  • Deblois G & Giguère V 2013 Oestrogen-related receptors in breast cancer: control of cellular metabolism and beyond. Nature Reviews. Cancer 13 2736. (doi:10.1038/nrc3396).

    • Search Google Scholar
    • Export Citation
  • Desclozeaux M, Krylova IN, Horn F, Fletterick RJ & Ingraham HA 2002 Phosphorylation and intramolecular stabilization of the ligand binding domain in the nuclear receptor steroidogenic factor 1. Molecular and Cellular Biology 22 71937203. (doi:10.1128/MCB.22.20.7193-7203.2002).

    • Search Google Scholar
    • Export Citation
  • Dhe-Paganon S, Duda K, Iwamoto M, Chi Y-I & Shoelson SE 2002 Crystal structure of the HNF4α ligand binding domain in complex with endogenous fatty acid ligand. Journal of Biological Chemistry 277 3797337976. (doi:10.1074/jbc.C200420200).

    • Search Google Scholar
    • Export Citation
  • Doghman M, Figueiredo BC, Volante M, Papotti M & Lalli E 2013 Integrative analysis of SF-1 transcription factor dosage impact on genome-wide binding and gene expression regulation. Nucleic Acids Research 41 88968907. (doi:10.1093/nar/gkt658).

    • Search Google Scholar
    • Export Citation
  • Drewes T, Senkel S, Holewa B & Ryffel GU 1996 Human hepatocyte nuclear factor 4 isoforms are encoded by distinct and differentially expressed genes. Molecular and Cellular Biology 16 925931.

    • Search Google Scholar
    • Export Citation
  • Dreyer C, Krey G, Keller H, Givel F, Helftenbein G & Wahli W 1992 Control of the peroxisomal β-oxidation pathway by a novel family of nuclear hormone receptors. Cell 68 879887. (doi:10.1016/0092-8674(92)90031-7).

    • Search Google Scholar
    • Export Citation
  • Duan SZ, Ivashchenko CY, Whitesall SE, D'Alecy LG, Duquaine DC, Brosius FC III, Gonzalez FJ, Vinson C, Pierre MA & Milstone DS et al. 2007 Hypotension, lipodystrophy, and insulin resistance in generalized PPARγ-deficient mice rescued from embryonic lethality. Journal of Clinical Investigation 117 812822. (doi:10.1172/JCI28859).

    • Search Google Scholar
    • Export Citation
  • Dufour CR, Wilson BJ, Huss JM, Kelly DP, Alaynick WA, Downes M, Evans RM, Blanchette M & Giguère V 2007 Genome-wide orchestration of cardiac functions by the orphan nuclear receptors ERRα and γ. Cell Metabolism 5 345356. (doi:10.1016/j.cmet.2007.03.007).

    • Search Google Scholar
    • Export Citation
  • Dumas B, Harding HP, Choi HS, Lehmann KA, Chung M, Lazar MA & Moore DD 1994 A new orphan member of the nuclear hormone receptor superfamily closely related to Rev-Erb. Molecular Endocrinology 8 9961005. (doi:10.1210/me.8.8.996).

    • Search Google Scholar
    • Export Citation
  • Duncan SA 2003 Mechanisms controlling early development of the liver. Mechanisms of Development 120 1933. (doi:10.1016/S0925-4773(02)00328-3).

    • Search Google Scholar
    • Export Citation
  • Eeckhoute J, Moerman E, Bouckenooghe T, Lukoviak B, Pattou F, Formstecher P, Kerr-Conte J, Vandewalle B & Laine B 2003 Hepatocyte nuclear factor 4α isoforms originated from the P1 promoter are expressed in human pancreatic β-cells and exhibit stronger transcriptional potentials than P2 promoter-driven isoforms. Endocrinology 144 16861694. (doi:10.1210/en.2002-0024).

    • Search Google Scholar
    • Export Citation
  • Ehrlund A & Treuter E 2012 Ligand-independent actions of the orphan receptors/corepressors DAX-1 and SHP in metabolism, reproduction and disease. Journal of Steroid Biochemistry and Molecular Biology 130 169179. (doi:10.1016/j.jsbmb.2011.04.007).

    • Search Google Scholar
    • Export Citation
  • Eichner LJ, Perry M-C, Dufour CR, Bertos N, Park M, St-Pierre J & Giguère V 2010 miR-378(∗) mediates metabolic shift in breast cancer cells via the PGC-1β/ERRγ transcriptional pathway. Cell Metabolism 12 352361. (doi:10.1016/j.cmet.2010.09.002).

    • Search Google Scholar
    • Export Citation
  • El-Khairi R & Achermann JC 2012 Steroidogenic factor-1 and human disease. Seminars in Reproductive Medicine 30 374381. (doi:10.1055/s-0032-1324720).

    • Search Google Scholar
    • Export Citation
  • Eudy JD, Yao S, Weston MD, Ma-Edmonds M, Talmadge CB, Cheng JJ, Kimberling WJ & Sumegi J 1998 Isolation of a gene encoding a novel member of the nuclear receptor superfamily from the critical region of Usher syndrome type IIa at 1q41. Genomics 50 382384. (doi:10.1006/geno.1998.5345).

    • Search Google Scholar
    • Export Citation
  • Evans RM, Barish GD & Wang Y-X 2004 PPARs and the complex journey to obesity. Nature Medicine 10 355361. (doi:10.1038/nm1025).

  • Fahrbach SE, Smagghe G & Velarde RA 2012 Insect nuclear receptors. Annual Review of Entomology 57 83106. (doi:10.1146/annurev-ento-120710-100607).

    • Search Google Scholar
    • Export Citation
  • Fang B, Mane-Padros D, Bolotin E, Jiang T & Sladek FM 2012 Identification of a binding motif specific to HNF4 by comparative analysis of multiple nuclear receptors. Nucleic Acids Research 40 53435356. (doi:10.1093/nar/gks190).

    • Search Google Scholar
    • Export Citation
  • Feng D & Lazar MA 2012 Clocks, metabolism, and the epigenome. Molecular Cell 47 158167. (doi:10.1016/j.molcel.2012.06.026).

  • Fernandez-Marcos PJ, Auwerx J & Schoonjans K 2011 Emerging actions of the nuclear receptor LRH-1 in the gut. Biochimica et Biophysica Acta 1812 947955. (doi:10.1016/j.bbadis.2010.12.010).

    • Search Google Scholar
    • Export Citation
  • Festuccia N, Osorno R, Halbritter F, Karwacki-Neisius V, Navarro P, Colby D, Wong F, Yates A, Tomlinson SR & Chambers I 2012 Esrrb is a direct Nanog target gene that can substitute for Nanog function in pluripotent cells. Cell Stem Cell 11 477490. (doi:10.1016/j.stem.2012.08.002).

    • Search Google Scholar
    • Export Citation
  • Filion C & Labelle Y 2012 Identification of genes regulated by the EWS/NR4A3 fusion protein in extraskeletal myxoid chondrosarcoma. Tumour Biology 33 15991605. (doi:10.1007/s13277-012-0415-2).

    • Search Google Scholar
    • Export Citation
  • Fontaine C, Rigamonti E, Pourcet B, Duez H, Duhem C, Fruchart J-C, Chinetti-Gbaguidi G & Staels B 2008 The nuclear receptor Rev-erbα is a liver X receptor (LXR) target gene driving a negative feedback loop on select LXR-induced pathways in human macrophages. Molecular Endocrinology 22 17971811. (doi:10.1210/me.2007-0439).

    • Search Google Scholar
    • Export Citation
  • Forman BM, Chen J, Blumberg B, Kliewer SA, Henshaw R, Ong ES & Evans RM 1994 Cross-talk among RORα1 and the Rev-erb family of orphan nuclear receptors. Molecular Endocrinology 8 12531261. (doi:10.1210/me.8.9.1253).

    • Search Google Scholar
    • Export Citation
  • Forman BM, Umesono K, Chen J & Evans RM 1995 Unique response pathways are established by allosteric interactions among nuclear hormone receptors. Cell 81 541550. (doi:10.1016/0092-8674(95)90075-6).

    • Search Google Scholar
    • Export Citation
  • Forman BM, Tzameli I, Choi HS, Chen J, Simha D, Seol W, Evans RM & Moore DD 1998 Androstane metabolites bind to and deactivate the nuclear receptor CAR-β. Nature 395 612615. (doi:10.1038/26996).

    • Search Google Scholar
    • Export Citation
  • Forrest D & Swaroop A 2012 Minireview: the role of nuclear receptors in photoreceptor differentiation and disease. Molecular Endocrinology 26 905915. (doi:10.1210/me.2012-1010).

    • Search Google Scholar
    • Export Citation
  • Freedman BD, Lee E-J, Park Y & Jameson JL 2005 A dominant negative peroxisome proliferator-activated receptor-γ knock-in mouse exhibits features of the metabolic syndrome. Journal of Biological Chemistry 280 1711817125. (doi:10.1074/jbc.M407539200).

    • Search Google Scholar
    • Export Citation
  • Frochot V, Alqub M, Cattin A-L, Carrière V, Houllier A, Baraille F, Barbot L, Saint-Just S, Ribeiro A & Lacasa M et al. 2012 The transcription factor HNF-4α: a key factor of the intestinal uptake of fatty acids in mouse. American Journal of Physiology. Gastrointestinal and Liver Physiology 302 G1253G1263. (doi:10.1152/ajpgi.00329.2011).

    • Search Google Scholar
    • Export Citation
  • Fuhrmann G, Chung AC, Jackson KJ, Hummelke G, Baniahmad A, Sutter J, Sylvester I, Schöler HR & Cooney AJ 2001 Mouse germline restriction of Oct4 expression by germ cell nuclear factor. Developmental Cell 1 377387. (doi:10.1016/S1534-5807(01)00038-7).

    • Search Google Scholar
    • Export Citation
  • Gabbi C, Warner M & Gustafsson J-A 2009 Minireview: liver X receptor β: emerging roles in physiology and diseases. Molecular Endocrinology 23 129136. (doi:10.1210/me.2008-0398).

    • Search Google Scholar
    • Export Citation
  • Galarneau L, Paré JF, Allard D, Hamel D, Levesque L, Tugwood JD, Green S & Bélanger L 1996 The α1-fetoprotein locus is activated by a nuclear receptor of the Drosophila FTZ-F1 family. Molecular and Cellular Biology 16 38533865.

    • Search Google Scholar
    • Export Citation
  • Gao J, He J, Zhai Y, Wada T & Xie W 2009 The constitutive androstane receptor is an anti-obesity nuclear receptor that improves insulin sensitivity. Journal of Biological Chemistry 284 2598425992. (doi:10.1074/jbc.M109.016808).

    • Search Google Scholar
    • Export Citation
  • Gao J & Xie W 2010 Pregnane X receptor and constitutive androstane receptor at the crossroads of drug metabolism and energy metabolism. Drug Metabolism and Disposition 38 20912095. (doi:10.1124/dmd.110.035568).

    • Search Google Scholar
    • Export Citation
  • Gerdin AK, Surve VV, Jönsson M, Bjursell M, Björkman M, Edenro A, Schuelke M, Saad A, Bjurström S & Lundgren EJ et al. 2006 Phenotypic screening of hepatocyte nuclear factor (HNF) 4-γ receptor knockout mice. Biochemical and Biophysical Research Communications 349 825832. (doi:10.1016/j.bbrc.2006.08.103).

    • Search Google Scholar
    • Export Citation
  • Germain P, Staels B, Dacquet C, Spedding M & Laudet V 2006 Overview of nomenclature of nuclear receptors. Pharmacological Reviews 58 685704. (doi:10.1124/pr.58.4.2).

    • Search Google Scholar
    • Export Citation
  • Gibbs JE, Blaikley J, Beesley S, Matthews L, Simpson KD, Boyce SH, Farrow SN, Else KJ, Singh D & Ray DW et al. 2012 The nuclear receptor REV-ERBα mediates circadian regulation of innate immunity through selective regulation of inflammatory cytokines. PNAS 109 582587. (doi:10.1073/pnas.1106750109).

    • Search Google Scholar
    • Export Citation
  • Giguère V 1999 Orphan nuclear receptors: from gene to function. Endocrine Reviews 20 689725. (doi:10.1210/er.20.5.689).

  • Giguère V 2008 Transcriptional control of energy homeostasis by the estrogen-related receptors. Endocrine Reviews 29 677696. (doi:10.1210/er.2008-0017).

    • Search Google Scholar
    • Export Citation
  • Giguère V, Yang N, Segui P & Evans RM 1988 Identification of a new class of steroid hormone receptors. Nature 331 9194. (doi:10.1038/331091a0).

    • Search Google Scholar
    • Export Citation
  • Gruber PJ, Kubalak SW, Pexieder T, Sucov HM, Evans RM & Chien KR 1996 RXRα deficiency confers genetic susceptibility for aortic sac, conotruncal, atrioventricular cushion, and ventricular muscle defects in mice. Journal of Clinical Investigation 98 13321343. (doi:10.1172/JCI118920).

    • Search Google Scholar
    • Export Citation
  • Gu P, LeMenuet D, Chung AC-K, Mancini M, Wheeler DA & Cooney AJ 2005a Orphan nuclear receptor GCNF is required for the repression of pluripotency genes during retinoic acid-induced embryonic stem cell differentiation. Molecular and Cellular Biology 25 85078519. (doi:10.1128/MCB.25.19.8507-8519.2005).

    • Search Google Scholar
    • Export Citation
  • Gu P, Morgan DH, Sattar M, Xu X, Wagner R, Raviscioni M, Lichtarge O & Cooney AJ 2005b Evolutionary trace-based peptides identify a novel asymmetric interaction that mediates oligomerization in nuclear receptors. Journal of Biological Chemistry 280 3181831829. (doi:10.1074/jbc.M501924200).

    • Search Google Scholar
    • Export Citation
  • Gui H, Li M-L & Tsai C-C 2011 A tale of tailless. Developmental Neuroscience 33 113. (doi:10.1159/000321585).

  • Guillaumond F, Dardente H, Giguère V & Cermakian N 2005 Differential control of Bmal1 circadian transcription by REV-ERB and ROR nuclear receptors. Journal of Biological Rhythms 20 391403. (doi:10.1177/0748730405277232).

    • Search Google Scholar
    • Export Citation
  • Guo GL, Moffit JS, Nicol CJ, Ward JM, Aleksunes LA, Slitt AL, Kliewer SA, Manautou JE & Gonzalez FJ 2004 Enhanced acetaminophen toxicity by activation of the pregnane X receptor. Toxicological Sciences 82 374380. (doi:10.1093/toxsci/kfh286).

    • Search Google Scholar
    • Export Citation
  • Gupta RK, Vatamaniuk MZ, Lee CS, Flaschen RC, Fulmer JT, Matschinsky FM, Duncan SA & Kaestner KH 2005 The MODY1 gene HNF-4α regulates selected genes involved in insulin secretion. Journal of Clinical Investigation 115 10061015. (doi:10.1172/JCI22365).

    • Search Google Scholar
    • Export Citation
  • Gupta RK, Gao N, Gorski RK, White P, Hardy OT, Rafiq K, Brestelli JE, Chen G, Stoeckert CJ Jr & Kaestner KH 2007 Expansion of adult β-cell mass in response to increased metabolic demand is dependent on HNF-4α. Genes and Development 21 756769. (doi:10.1101/gad.1535507).

    • Search Google Scholar
    • Export Citation
  • Gupta P, Ho P-C, Huq MM, Ha SG, Park SW, Khan AA, Tsai N-P & Wei L-N 2008 Retinoic acid-stimulated sequential phosphorylation, PML recruitment, and SUMOylation of nuclear receptor TR2 to suppress Oct4 expression. PNAS 105 1142411429. (doi:10.1073/pnas.0710561105).

    • Search Google Scholar
    • Export Citation
  • Gupta P, Ho P-C, Ha SG, Lin Y-W & Wei L-N 2009 HDAC3 as a molecular chaperone for shuttling phosphorylated TR2 to PML: a novel deacetylase activity-independent function of HDAC3. PLoS ONE 4 e4363. (doi:10.1371/journal.pone.0004363).

    • Search Google Scholar
    • Export Citation
  • Gurtan AM, Ravi A, Rahl PB, Bosson AD, JnBaptiste CK, Bhutkar A, Whittaker CA, Young RA & Sharp PA 2013 Let-7 represses Nr6a1 and a mid-gestation developmental program in adult fibroblasts. Genes and Development 27 941954. (doi:10.1101/gad.215376.113).

    • Search Google Scholar
    • Export Citation
  • Hamilton BA, Frankel WN, Kerrebrock AW, Hawkins TL, FitzHugh W, Kusumi K, Russell LB, Mueller KL, van Berkel V & Birren BW et al. 1996 Disruption of the nuclear hormone receptor RORα in staggerer mice. Nature 379 736739. (doi:10.1038/379736a0).

    • Search Google Scholar
    • Export Citation
  • Haugen BR, Jensen DR, Sharma V, Pulawa LK, Hays WR, Krezel W, Chambon P & Eckel RH 2004 Retinoid X receptor γ-deficient mice have increased skeletal muscle lipoprotein lipase activity and less weight gain when fed a high-fat diet. Endocrinology 145 36793685. (doi:10.1210/en.2003-1401).

    • Search Google Scholar
    • Export Citation
  • Hayhurst GP, Lee Y-H, Lambert G, Ward JM & Gonzalez FJ 2001 Hepatocyte nuclear factor 4α (nuclear receptor 2A1) is essential for maintenance of hepatic gene expression and lipid homeostasis. Molecular and Cellular Biology 21 13931403. (doi:10.1128/MCB.21.4.1393-1403.2001).

    • Search Google Scholar
    • Export Citation
  • Hazel TG, Nathans D & Lau LF 1988 A gene inducible by serum growth factors encodes a member of the steroid and thyroid hormone receptor superfamily. PNAS 85 84448448. (doi:10.1073/pnas.85.22.8444).

    • Search Google Scholar
    • Export Citation
  • Heard DJ, Norby PL, Holloway J & Vissing H 2000 Human ERRγ, a third member of the estrogen receptor-related receptor (ERR) subfamily of orphan nuclear receptors: tissue-specific isoforms are expressed during development and in the adult. Molecular Endocrinology 14 382392. (doi:10.1210/me.14.3.382).

    • Search Google Scholar
    • Export Citation
  • Hedvat CV & Irving SG 1995 The isolation and characterization of MINOR, a novel mitogen-inducible nuclear orphan receptor. Molecular Endocrinology 9 16921700. (doi:10.1210/me.9.12.1692).

    • Search Google Scholar
    • Export Citation
  • Heinzer C, Süsens U, Schmitz TP & Borgmeyer U 1998 Retinoids induce differential expression and DNA binding of the mouse germ cell nuclear factor in P19 embryonal carcinoma cells. Biological Chemistry 379 349359. (doi:10.1515/bchm.1998.379.3.349).

    • Search Google Scholar
    • Export Citation
  • Helsen C, Kerkhofs S, Clinckemalie L, Spans L, Laurent M, Boonen S, Vanderschueren D & Claessens F 2012 Structural basis for nuclear hormone receptor DNA binding. Molecular and Cellular Endocrinology 348 411417. (doi:10.1016/j.mce.2011.07.025).

    • Search Google Scholar
    • Export Citation
  • Hermann-Kleiter N, Gruber T, Lutz-Nicoladoni C, Thuille N, Fresser F, Labi V, Schiefermeier N, Warnecke M, Huber L & Villunger A et al. 2008 The nuclear orphan receptor NR2F6 suppresses lymphocyte activation and T helper 17-dependent autoimmunity. Immunity 29 205216. (doi:10.1016/j.immuni.2008.06.008).

    • Search Google Scholar
    • Export Citation
  • Hermann-Kleiter N, Meisel M, Fresser F, Thuille N, Müller M, Roth L, Katopodis A & Baier G 2012 Nuclear orphan receptor NR2F6 directly antagonizes NFAT and RORγt binding to the Il17a promoter. Journal of Autoimmunity 39 428440. (doi:10.1016/j.jaut.2012.07.007).

    • Search Google Scholar
    • Export Citation
  • Hirose T, Fujimoto W, Tamaai T, Kim KH, Matsuura H & Jetten AM 1994 TAK1: molecular cloning and characterization of a new member of the nuclear receptor superfamily. Molecular Endocrinology 8 16671680. (doi:10.1210/me.8.12.1667).

    • Search Google Scholar
    • Export Citation
  • Hirose T, O'Brien DA & Jetten AM 1995 RTR: a new member of the nuclear receptor superfamily that is highly expressed in murine testis. Gene 152 247251. (doi:10.1016/0378-1119(94)00656-D).

    • Search Google Scholar
    • Export Citation
  • Hong H, Yang L & Stallcup MR 1999 Hormone-independent transcriptional activation and coactivator binding by novel orphan nuclear receptor ERR3. Journal of Biological Chemistry 274 2261822626. (doi:10.1074/jbc.274.32.22618).

    • Search Google Scholar
    • Export Citation
  • Hong C, Bradley MN, Rong X, Wang X, Wagner A, Grijalva V, Castellani LW, Salazar J, Realegeno S & Boyadjian R et al. 2012 LXRα is uniquely required for maximal reverse cholesterol transport and atheroprotection in ApoE-deficient mice. Journal of Lipid Research 53 11261133. (doi:10.1194/jlr.M022061).

    • Search Google Scholar
    • Export Citation
  • Hossain A, Li C & Saunders GF 2004 Generation of two distinct functional isoforms of dosage-sensitive sex reversal-adrenal hypoplasia congenita-critical region on the X chromosome gene 1 (DAX-1) by alternative splicing. Molecular Endocrinology 18 14281437. (doi:10.1210/me.2003-0176).

    • Search Google Scholar
    • Export Citation
  • Hu Y-C, Shyr C-R, Che W, Mu X-M, Kim E & Chang C 2002 Suppression of estrogen receptor-mediated transcription and cell growth by interaction with TR2 orphan receptor. Journal of Biological Chemistry 277 3357133579. (doi:10.1074/jbc.M203531200).

    • Search Google Scholar
    • Export Citation
  • Huang P, Chandra V & Rastinejad F 2010 Structural overview of the nuclear receptor superfamily: insights into physiology and therapeutics. Annual Review of Physiology 72 247272. (doi:10.1146/annurev-physiol-021909-135917).

    • Search Google Scholar
    • Export Citation
  • Huang JK, Jarjour AA, Nait Oumesmar B, Kerninon C, Williams A, Krezel W, Kagechika H, Bauer J, Zhao C & Baron-Van Evercooren A et al. 2011 Retinoid X receptor γ signaling accelerates CNS remyelination. Nature Neuroscience 14 4553. (doi:10.1038/nn.2702).

    • Search Google Scholar
    • Export Citation
  • Huh JR, Leung MWL, Huang P, Ryan DA, Krout MR, Malapaka RRV, 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 486490. (doi:10.1038/nature09978).

    • Search Google Scholar
    • Export Citation
  • Hummelke GC & Cooney AJ 2001 Germ cell nuclear factor is a transcriptional repressor essential for embryonic development. Frontiers in Bioscience: A Journal and Virtual Library 6 D1186D1191. (doi:10.2741/Hummelke).

    • Search Google Scholar
    • Export Citation
  • Hwang EJ, Lee JM, Jeong J, Park JH, Yang Y, Lim J-S, Kim JH, Baek SH & Kim KI 2009 SUMOylation of RORα potentiates transcriptional activation function. Biochemical and Biophysical Research Communications 378 513517. (doi:10.1016/j.bbrc.2008.11.072).

    • Search Google Scholar
    • Export Citation
  • Ichiyama K, Yoshida H, Wakabayashi Y, Chinen T, Saeki K, Nakaya M, Takaesu G, Hori S, Yoshimura A & Kobayashi T 2008 Foxp3 inhibits RORγt-mediated IL-17A mRNA transcription through direct interaction with RORγt. Journal of Biological Chemistry 283 1700317008. (doi:10.1074/jbc.M801286200).

    • Search Google Scholar
    • Export Citation
  • Ihunnah CA, Jiang M & Xie W 2011 Nuclear receptor PXR, transcriptional circuits and metabolic relevance. Biochimica et Biophysica Acta 1812 956963. (doi:10.1016/j.bbadis.2011.01.014).

    • Search Google Scholar
    • Export Citation
  • IJpenberg A, Jeannin E, Wahli W & Desvergne B 1997 Polarity and specific sequence requirements of peroxisome proliferator-activated receptor (PPAR)/retinoid X receptor heterodimer binding to DNA. A functional analysis of the malic enzyme gene PPAR response element. Journal of Biological Chemistry 272 2010820117. (doi:10.1074/jbc.272.32.20108).

    • Search Google Scholar
    • Export Citation
  • Ingraham HA & Redinbo MR 2005 Orphan nuclear receptors adopted by crystallography. Current Opinion in Structural Biology 15 708715. (doi:10.1016/j.sbi.2005.10.009).

    • Search Google Scholar
    • Export Citation
  • Issemann I & Green S 1990 Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators. Nature 347 645650. (doi:10.1038/347645a0).

    • Search Google Scholar
    • Export Citation
  • Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ, Cua DJ & Littman DR 2006 The orphan nuclear receptor RORγt directs the differentiation program of proinflammatory IL-17+T helper cells. Cell 126 11211133. (doi:10.1016/j.cell.2006.07.035).

    • Search Google Scholar
    • Export Citation
  • Ivanova N, Dobrin R, Lu R, Kotenko I, Levorse J, DeCoste C, Schafer X, Lun Y & Lemischka IR 2006 Dissecting self-renewal in stem cells with RNA interference. Nature 442 533538. (doi:10.1038/nature04915).

    • Search Google Scholar
    • Export Citation
  • Iyer AK & McCabe ERB 2004 Molecular mechanisms of DAX1 action. Molecular Genetics and Metabolism 83 6073. (doi:10.1016/j.ymgme.2004.07.018).

    • Search Google Scholar
    • Export Citation
  • Jetten AM 2009 Retinoid-related orphan receptors (RORs): critical roles in development, immunity, circadian rhythm, and cellular metabolism. Nuclear Receptor Signaling 7 e003. (doi:10.1621/nrs.07003).

    • Search Google Scholar
    • Export Citation
  • Jin L, Martynowski D, Zheng S, Wada T, Xie W & Li Y 2010 Structural basis for hydroxycholesterols as natural ligands of orphan nuclear receptor RORγ. Molecular Endocrinology 24 923929. (doi:10.1210/me.2009-0507).

    • Search Google Scholar
    • Export Citation
  • Johnston SD, Liu X, Zuo F, Eisenbraun TL, Wiley SR, Kraus RJ & Mertz JE 1997 Estrogen-related receptor α 1 functionally binds as a monomer to extended half-site sequences including ones contained within estrogen-response elements. Molecular Endocrinology 11 342352. (doi:10.1210/me.11.3.342).

    • Search Google Scholar
    • Export Citation
  • Jonk LJ, de Jonge ME, Pals CE, Wissink S, Vervaart JM, Schoorlemmer J & Kruijer W 1994 Cloning and expression during development of three murine members of the COUP family of nuclear orphan receptors. Mechanisms of Development 47 8197. (doi:10.1016/0925-4773(94)90098-1).

    • Search Google Scholar
    • Export Citation
  • Joseph SB, McKilligin E, Pei L, Watson MA, Collins AR, Laffitte BA, Chen M, Noh G, Goodman J & Hagger GN et al. 2002 Synthetic LXR ligand inhibits the development of atherosclerosis in mice. PNAS 99 76047609. (doi:10.1073/pnas.112059299).

    • Search Google Scholar
    • Export Citation
  • Kallen JA, Schlaeppi J-M, Bitsch F, Geisse S, Geiser M, Delhon I & Fournier B 2002 X-ray structure of the hRORα LBD at 1.63 A: structural and functional data that cholesterol or a cholesterol derivative is the natural ligand of RORα. Structure 10 16971707. (doi:10.1016/S0969-2126(02)00912-7).

    • Search Google Scholar
    • Export Citation
  • Kallen J, Lattmann R, Beerli R, Blechschmidt A, Blommers MJJ, Geiser M, Ottl J, Schlaeppi J-M, Strauss A & Fournier B 2007 Crystal structure of human estrogen-related receptor α in complex with a synthetic inverse agonist reveals its novel molecular mechanism. Journal of Biological Chemistry 282 2323123239. (doi:10.1074/jbc.M703337200).

    • Search Google Scholar
    • Export Citation
  • Kang HS, Okamoto K, Kim Y-S, Takeda Y, Bortner CD, Dang H, Wada T, Xie W, Yang X-P & Liao G et al. 2011 Nuclear orphan receptor TAK1/TR4-deficient mice are protected against obesity-linked inflammation, hepatic steatosis, and insulin resistance. Diabetes 60 177188. (doi:10.2337/db10-0628).

    • Search Google Scholar
    • Export Citation
  • Kastner P, Grondona JM, Mark M, Gansmuller A, LeMeur M, Decimo D, Vonesch JL, Dollé P & Chambon P 1994 Genetic analysis of RXRα developmental function: convergence of RXR and RAR signaling pathways in heart and eye morphogenesis. Cell 78 9871003. (doi:10.1016/0092-8674(94)90274-7).

    • Search Google Scholar
    • Export Citation
  • Kastner P, Mark M, Leid M, Gansmuller A, Chin W, Grondona JM, Décimo D, Krezel W, Dierich A & Chambon P 1996 Abnormal spermatogenesis in RXRβ mutant mice. Genes and Development 10 8092. (doi:10.1101/gad.10.1.80).

    • Search Google Scholar
    • Export Citation
  • Kawamoto T, Sueyoshi T, Zelko I, Moore R, Washburn K & Negishi M 1999 Phenobarbital-responsive nuclear translocation of the receptor CAR in induction of the CYP2B gene. Molecular and Cellular Biology 19 63186322.

    • Search Google Scholar
    • Export Citation
  • Kelly VR, Xu B, Kuick R, Koenig RJ & Hammer GD 2010 Dax1 up-regulates Oct4 expression in mouse embryonic stem cells via LRH-1 and SRA. Molecular Endocrinology 24 22812291. (doi:10.1210/me.2010-0133).

    • Search Google Scholar
    • Export Citation
  • Kersten S, Seydoux J, Peters JM, Gonzalez FJ, Desvergne B & Wahli W 1999 Peroxisome proliferator-activated receptor α mediates the adaptive response to fasting. Journal of Clinical Investigation 103 14891498. (doi:10.1172/JCI6223).

    • Search Google Scholar
    • Export Citation
  • Khan SA, Park SW, Huq M & Wei L-N 2005 Protein kinase C-mediated phosphorylation of orphan nuclear receptor TR2: effects on receptor stability and activity. Proteomics 5 38853894. (doi:10.1002/pmic.200402062).

    • Search Google Scholar
    • Export Citation
  • Kim YS, Han CY, Kim SW, Kim JH, Lee SK, Jung DJ, Park SY, Kang H, Choi HS & Lee JW et al. 2001 The orphan nuclear receptor small heterodimer partner as a novel coregulator of nuclear factor-kappa b in oxidized low density lipoprotein-treated macrophage cell line RAW 264.7. Journal of Biological Chemistry 276 3373633740. (doi:10.1074/jbc.M101977200).

    • Search Google Scholar
    • Export Citation
  • Kim E, Xie S, Yeh S-D, Lee Y-F, Collins LL, Hu Y-C, Shyr C-R, Mu X-M, Liu N-C & Chen Y-T et al. 2003 Disruption of TR4 orphan nuclear receptor reduces the expression of liver apolipoprotein E/C-I/C-II gene cluster. Journal of Biological Chemistry 278 4691946926. (doi:10.1074/jbc.M304088200).

    • Search Google Scholar
    • Export Citation
  • Kim J-Y, Chu K, Kim H-J, Seong H-A, Park K-C, Sanyal S, Takeda J, Ha H, Shong M & Tsai M-J et al. 2004 Orphan nuclear receptor small heterodimer partner, a novel corepressor for a basic helix–loop–helix transcription factor BETA2/neuroD. Molecular Endocrinology 18 776790. (doi:10.1210/me.2003-0311).

    • Search Google Scholar
    • Export Citation
  • Kim J, Chu J, Shen X, Wang J & Orkin SH 2008 An extended transcriptional network for pluripotency of embryonic stem cells. Cell 132 10491061. (doi:10.1016/j.cell.2008.02.039).

    • Search Google Scholar
    • Export Citation
  • Kim H, Lee JM, Lee G, Bhin J, Oh SK, Kim K, Pyo KE, Lee JS, Yim HY & Kim KI et al. 2011 DNA damage-induced RORα is crucial for p53 stabilization and increased apoptosis. Molecular Cell 44 797810. (doi:10.1016/j.molcel.2011.09.023).

    • Search Google Scholar
    • Export Citation
  • Kim YD, Li T, Ahn S-W, Kim D-K, Lee J-M, Hwang S-L, Kim Y-H, Lee C-H, Lee I-K & Chiang JYL et al. 2012 Orphan nuclear receptor small heterodimer partner negatively regulates growth hormone-mediated induction of hepatic gluconeogenesis through inhibition of signal transducer and activator of transcription 5 (STAT5) transactivation. Journal of Biological Chemistry 287 3709837108. (doi:10.1074/jbc.M112.339887).

    • Search Google Scholar
    • Export Citation
  • Kinsey M, Smith R, Iyer AK, McCabe ERB & Lessnick SL 2009 EWS/FLI and its downstream target NR0B1 interact directly to modulate transcription and oncogenesis in Ewing's sarcoma. Cancer Research 69 90479055. (doi:10.1158/0008-5472.CAN-09-1540).

    • Search Google Scholar
    • Export Citation
  • Kliewer SA, Umesono K, Mangelsdorf DJ & Evans RM 1992a Retinoid X receptor interacts with nuclear receptors in retinoic acid, thyroid hormone and vitamin D3 signalling. Nature 355 446449. (doi:10.1038/355446a0).

    • Search Google Scholar
    • Export Citation
  • Kliewer SA, Umesono K, Noonan DJ, Heyman RA & Evans RM 1992b Convergence of 9-cis retinoic acid and peroxisome proliferator signalling pathways through heterodimer formation of their receptors. Nature 358 771774. (doi:10.1038/358771a0).

    • Search Google Scholar
    • Export Citation
  • Kliewer SA, Moore JT, Wade L, Staudinger JL, Watson MA, Jones SA, McKee DD, Oliver BB, Willson TM & Zetterström RH et al. 1998 An orphan nuclear receptor activated by pregnanes defines a novel steroid signaling pathway. Cell 92 7382. (doi:10.1016/S0092-8674(00)80900-9).

    • Search Google Scholar
    • Export Citation
  • Kliewer SA, Goodwin B & Willson TM 2002 The nuclear pregnane X receptor: a key regulator of xenobiotic metabolism. Endocrine Reviews 23 687702. (doi:10.1210/er.2001-0038).

    • Search Google Scholar
    • Export Citation
  • Kobayashi M, Takezawa S, Hara K, Yu RT, Umesono Y, Agata K, Taniwaki M, Yasuda K & Umesono K 1999 Identification of a photoreceptor cell-specific nuclear receptor. PNAS 96 48144819. (doi:10.1073/pnas.96.9.4814).

    • Search Google Scholar
    • Export Citation
  • Kolluri SK, Zhu X, Zhou X, Lin B, Chen Y, Sun K, Tian X, Town J, Cao X & Lin F et al. 2008 A short Nur77-derived peptide converts Bcl-2 from a protector to a killer. Cancer Cell 14 285298. (doi:10.1016/j.ccr.2008.09.002).

    • Search Google Scholar
    • Export Citation
  • Krill KT, Gurdziel K, Heaton JH, Simon DP & Hammer GD 2013 Dicer deficiency reveals microRNAs predicted to control gene expression in the developing adrenal cortex. Molecular Endocrinology 27 754768. (doi:10.1210/me.2012-1331).

    • Search Google Scholar
    • Export Citation
  • Kritis AA, Argyrokastritis A, Moschonas NK, Power S, Katrakili N, Zannis VI, Cereghini S & Talianidis I 1996 Isolation and characterization of a third isoform of human hepatocyte nuclear factor 4. Gene 173 275280. (doi:10.1016/0378-1119(96)00183-7).

    • Search Google Scholar
    • Export Citation
  • Krylova IN, Sablin EP, Moore J, Xu RX, Waitt GM, MacKay JA, Juzumiene D, Bynum JM, Madauss K & Montana V et al. 2005 Structural analyses reveal phosphatidyl inositols as ligands for the NR5 orphan receptors SF-1 and LRH-1. Cell 120 343355. (doi:10.1016/j.cell.2005.01.024).

    • Search Google Scholar
    • Export Citation
  • Krzyzosiak A, Szyszka-Niagolov M, Wietrzych M, Gobaille S, Muramatsu S & Krezel W 2010 Retinoid X receptor γ control of affective behaviors involves dopaminergic signaling in mice. Neuron 66 908920. (doi:10.1016/j.neuron.2010.05.004).

    • Search Google Scholar
    • Export Citation
  • Kurebayashi S, Ueda E, Sakaue M, Patel DD, Medvedev A, Zhang F & Jetten AM 2000 Retinoid-related orphan receptor γ (RORγ) is essential for lymphoid organogenesis and controls apoptosis during thymopoiesis. PNAS 97 1013210137. (doi:10.1073/pnas.97.18.10132).

    • Search Google Scholar
    • Export Citation
  • Labelle Y, Zucman J, Stenman G, Kindblom LG, Knight J, Turc-Carel C, Dockhorn-Dworniczak B, Mandahl N, Desmaze C & Peter M 1995 Oncogenic conversion of a novel orphan nuclear receptor by chromosome translocation. Human Molecular Genetics 4 22192226. (doi:10.1093/hmg/4.12.2219).

    • Search Google Scholar
    • Export Citation
  • Ladias JA & Karathanasis SK 1991 Regulation of the apolipoprotein AI gene by ARP-1, a novel member of the steroid receptor superfamily. Science 251 561565. (doi:10.1126/science.1899293).

    • Search Google Scholar
    • Export Citation
  • Laffitte BA, Kast HR, Nguyen CM, Zavacki AM, Moore DD & Edwards PA 2000 Identification of the DNA binding specificity and potential target genes for the farnesoid X-activated receptor. Journal of Biological Chemistry 275 1063810647. (doi:10.1074/jbc.275.14.10638).

    • Search Google Scholar
    • Export Citation
  • Lala DS, Rice DA & Parker KL 1992 Steroidogenic factor I, a key regulator of steroidogenic enzyme expression, is the mouse homolog of fushi tarazu-factor I. Molecular Endocrinology 6 12491258. (doi:10.1210/me.6.8.1249).

    • Search Google Scholar
    • Export Citation
  • Lalli E & Sassone-Corsi P 2003 DAX-1, an unusual orphan receptor at the crossroads of steroidogenic function and sexual differentiation. Molecular Endocrinology 17 14451453. (doi:10.1210/me.2003-0159).

    • Search Google Scholar
    • Export Citation
  • Lalli E, Doghman M, Latre de Late P, Wakil AE & Mus-Veteau I 2013 Beyond steroidogenesis: novel target genes for SF-1 discovered by genomics. Molecular and Cellular Endocrinology 371 154159. (doi:10.1016/j.mce.2012.11.005).

    • Search Google Scholar
    • Export Citation
  • Lam MTY, Cho H, Lesch HP, Gosselin D, Heinz S, Tanaka-Oishi Y, Benner C, Kaikkonen MU, Kim AS & Kosaka M et al. 2013 Rev-Erbs repress macrophage gene expression by inhibiting enhancer-directed transcription. Nature 498 511515. (doi:10.1038/nature12209).

    • Search Google Scholar
    • Export Citation
  • Laudet V 1997 Evolution of the nuclear receptor superfamily: early diversification from an ancestral orphan receptor. Journal of Molecular Endocrinology 19 207226. (doi:10.1677/jme.0.0190207).

    • Search Google Scholar
    • Export Citation
  • Law SW, Conneely OM, DeMayo FJ & O'Malley BW 1992 Identification of a new brain-specific transcription factor, NURR1. Molecular Endocrinology 6 21292135. (doi:10.1210/me.6.12.2129).

    • Search Google Scholar
    • Export Citation
  • Law SW, Conneely OM & O'Malley BW 1994 Molecular cloning of a novel member of the nuclear receptor superfamily related to the orphan receptor, TR2. Gene Expression 4 7784.

    • Search Google Scholar
    • Export Citation
  • Lazar MA, Hodin RA, Darling DS & Chin WW 1989 A novel member of the thyroid/steroid hormone receptor family is encoded by the opposite strand of the rat c-erbAα transcriptional unit. Molecular and Cellular Biology 9 11281136.

    • Search Google Scholar
    • Export Citation
  • Lee CH, Chinpaisal C & Wei LN 1998 A novel nuclear receptor heterodimerization pathway mediated by orphan receptors TR2 and TR4. Journal of Biological Chemistry 273 2520925215. (doi:10.1074/jbc.273.39.25209).

    • Search Google Scholar
    • Export Citation
  • Lee Y-F, Lee H-J & Chang C 2002 Recent advances in the TR2 and TR4 orphan receptors of the nuclear receptor superfamily. Journal of Steroid Biochemistry and Molecular Biology 81 291308. (doi:10.1016/S0960-0760(02)00118-8).

    • Search Google Scholar
    • Export Citation
  • Lee Y-K, Schmidt DR, Cummins CL, Choi M, Peng L, Zhang Y, Goodwin B, Hammer RE, Mangelsdorf DJ & Kliewer SA 2008 Liver receptor homolog-1 regulates bile acid homeostasis but is not essential for feedback regulation of bile acid synthesis. Molecular Endocrinology 22 13451356. (doi:10.1210/me.2007-0565).

    • Search Google Scholar
    • Export Citation
  • Lee JM, Kim IS, Kim H, Lee JS, Kim K, Yim HY, Jeong J, Kim JH, Kim J-Y & Lee H et al. 2010 RORα attenuates Wnt/β-catenin signaling by PKCα-dependent phosphorylation in colon cancer. Molecular Cell 37 183195. (doi:10.1016/j.molcel.2009.12.022).

    • Search Google Scholar
    • Export Citation
  • Lee JM, Lee YK, Mamrosh JL, Busby SA, Griffin PR, Pathak MC, Ortlund EA & Moore DD 2011a A nuclear-receptor-dependent phosphatidylcholine pathway with antidiabetic effects. Nature 474 506510. (doi:10.1038/nature10111).

    • Search Google Scholar
    • Export Citation
  • Lee K, Khan S, Ansar M, Santos-Cortez RLP, Ahmad W & Leal SM 2011b A novel ESRRB deletion is a rare cause of autosomal recessive nonsyndromic hearing impairment among Pakistani families. Genetics Research International 2011 368915. (doi:10.4061/2011/368915).

    • Search Google Scholar
    • Export Citation
  • Lee JM, Lee JS, Kim H, Kim K, Park H, Kim J-Y, Lee SH, Kim IS, Kim J & Lee M et al. 2012 EZH2 generates a methyl degron that is recognized by the DCAF1/DDB1/CUL4 E3 ubiquitin ligase complex. Molecular Cell 48 572586. (doi:10.1016/j.molcel.2012.09.004).

    • Search Google Scholar
    • Export Citation
  • Lefebvre P, Benomar Y & Staels B 2010 Retinoid X receptors: common heterodimerization partners with distinct functions. Trends in Endocrinology and Metabolism 21 676683. (doi:10.1016/j.tem.2010.06.009).

    • Search Google Scholar
    • Export Citation
  • Lefterova MI, Zhang Y, Steger DJ, Schupp M, Schug J, Cristancho A, Feng D, Zhuo D, Stoeckert CJ & Liu XS et al. 2008 PPAR and C/EBP factors orchestrate adipocyte biology via adjacent binding on a genome-wide scale. Genes and Development 22 29412952. (doi:10.1101/gad.1709008).

    • Search Google Scholar
    • Export Citation
  • Lefterova MI, Steger DJ, Zhuo D, Qatanani M, Mullican SE, Tuteja G, Manduchi E, Grant GR & Lazar MA 2010 Cell-specific determinants of peroxisome proliferator-activated receptor γ function in adipocytes and macrophages. Molecular and Cellular Biology 30 20782089. (doi:10.1128/MCB.01651-09).