Abstract
Cholesterol and its metabolites are bioactive lipids that interact with and regulate the activity of various proteins and signaling pathways that are implicated in the control of a variety of physiological and pathological processes. Recent studies revealed that retinoic acid-related orphan receptors, RORα and γ, members of the ligand-dependent nuclear receptor superfamily, exhibit quite a wide binding specificity for a number of sterols. Several cholesterol intermediates and metabolites function as natural ligands of RORα and RORγ and act as agonists or inverse agonists. Changes in cholesterol homeostasis that alter the level or type of sterol metabolites in cells, can either enhance or inhibit ROR transcriptional activity that subsequently result in changes in the physiological processes regulated by RORs, including various immune responses and metabolic pathways. Consequently, this might negatively or positively impact pathologies, in which RORs are implicated, such as autoimmune disease, inflammation, metabolic syndrome, cancer, and several neurological disorders. Best studied are the links between cholesterol metabolism, RORγt activity, and their regulation of Th17 differentiation and autoimmune disease. The discovery that Th17-dependent inflammation is significantly attenuated in RORγ-deficient mice in several experimental autoimmune disease models, initiated a search for ROR modulators that led to the identification of a number of small molecular weight RORγ inverse agonists. The inverse agonists suppress Th17 differentiation and IL-17 production and protect against autoimmunity. Together, these studies suggest that RORγt may provide an attractive therapeutic target in the management of several (inflammatory) diseases.
Keywords: retinoic acid-related orphan receptor, RORγ, oxysterols, metabolism, (inverse) agonists, cholesterol biosynthesis, vitamin D3, transcription, Th17 cells, autoimmune disease, inflammation
1. Introduction
The retinoic acid-related orphan receptors alpha, beta, and gamma (RORα-γ encoded by RORA-C or NR1F1-3) constitute a subfamily of nuclear receptors [1–4]. RORs exhibit a typical nuclear receptor domain structure consisting of an N-terminal domain, a highly-conserved DNA-binding domain (DBD) consisting of two C2-C2 zinc finger motifs, a ligand-binding domain (LBD), and a hinge domain spacing the DBD and LBD [1]. The DBD of RORs recognizes ROR response elements (ROREs) consisting of the RGGTCA consensus preceded by an A/T-rich sequence. RORs regulate transcription by binding as monomers to ROREs in the regulatory regions of target genes and the recruitment of co-activators or co-repressors [5,6].
Over the last ten years, it has become evident that RORs function as ligand-dependent transcription factors. RORα/γ transcriptional activity can be modulated by various sterols and synthetic ligands that bind ROR and function either as agonists or inverse agonists [2,4,7–17]. The discovery that ROR transcriptional activity can be modulated by small (synthetic) molecules, opened the possibility that RORs may provide novel therapeutic targets in the management of various pathologies, in which RORs are implicated, such as autoimmune disease and type 2 diabetes. In addition, RORs may provide a target for various xenobiotics and thereby a mechanism by which environmental agents affect immunity and disease. This chapter provides an overview of the various links between cholesterol/sterol metabolism, RORs, and their regulation of immunity, particularly T helper 17 (Th17) cells, and their relationship to inflammatory disease.
2. ROR isoforms have different functions
The activity and function of RORs are controlled at many levels, including the generation of different isoforms. Via alternative splicing and/or alternative promoter usage the human RORA gene generates 4 isoforms (α1–4) and human RORC two isoforms, RORγ1 and RORγt (RORγ2), while human RORB encodes one protein [1,18–21]. These ROR isoforms, which differ only in their N-terminus, exhibit different patterns of expression and regulate distinct genes and physiological processes. A clear example of this is RORγ. RORγ1 is expressed in many peripheral tissues, including adipose, liver, kidney and muscle, where it regulates circadian rhythm, glucose and lipid metabolism [1,4,22–25]. In contrast, RORγt, to is selectively expressed in a variety of immune cells, including CD4+CD8+ (DP) thymocytes, T helper Th17 cells, type 3 innate lymphoid cells (ILC3), and lymphoid tissue inducer (LTi) cells, which constitute an ILC3 subtype [1,4,19,20,26–30]. RORγt regulates the survival and apoptosis of DP thymocytes and is essential for the development of ILC3 and Th17 cells, and the production of IL-17. RORγt is also required for the generation of LTi cells, which are essential for the development of peripheral lymph nodes and Peyer’s patches. RORγ-deficient mice lack Lti cells and peripheral lymph nodes [19,20]. RORγt is also expressed in several other immune cells, including subtypes of CD8+Tc17, invariant natural killer and γδT-cells, and Foxp3+ T regulatory cells (Treg) [31–36]. Foxp3+RORγt+ Tregs are preferentially expressed in colon and, as Th17 cells, their induction is regulated by the gut microbiota. However, in contrast to Th17 and ILC3 cells, RORγt appears not to be a significant regulator of IL-17 expression in Foxp3+RORγt+ Tregs indicating that the control of target gene transcription by RORγ is cell context-dependent. Th17, ILC3, and Foxp3+RORγt+ Tregs all contribute to intestinal homeostasis and tolerance. Its expression in various immune cell populations with diverse functions indicates that RORγt has multiple roles in regulating immunologic homeostasis and inflammatory disease, including autoimmune disease. The complexity is further complicated by the plasticity of the various immune cell populations.
3. Regulation of ROR activity
The activity and function of RORs can be modulated by various posttranslational modifications (PTMs) [37]. Although the RORs are phosphorylated at multiple sites and can be acetylated, sumoylated, and ubiquitinated, the function of many of these PTMs has not yet been fully elucidated. Phosphorylation by protein kinase A (PKA) and sumoylation of RORα has been reported to positively regulate its transcriptional activity [38,39]. Sumoylation of RORα at Lys240 was enhanced by protein inhibitor of activated STAT (PIAS) proteins and shown to stimulate RORα transcriptional activity. The histone acetylase, p300, acetylates RORγ at several positions, while the deacetylase SIRT1 promotes deacetylation of RORγ and increases its transcriptional activity, which consequently enhances the generation of Th17 cells and promotes autoimmunity [40]. The HECT E3 ubiquitin ligase, ITCH, was found to bind RORγ and mediate its K48 poly-ubiquitination and subsequently its proteasomal degradation [41]. A recent study showed that a two-amino acid substitution in the hinge domain of RORγ (Ser92Ala-Leu93Ala) decreased ubiquitination at Lys69 within the RORγ DBD; however, this ubiquitination did not affect RORγt protein stability [42]. Mice containing this mutation exhibited defective Th17 differentiation, while thymopoiesis and lymph node genesis were not affected, except for the development of Peyer’s patches. ILC3 cells development was not disrupted in these mutant mice, but cells did not produce IL-17. The mechanism of this selectivity is not fully understood and might be due to differences in promoter context of target genes or differential expression and/or recruitment of transcriptional mediators in different RORγ expressing cells.
4. Links between sterols and ROR signaling
Agonists and inverse agonists provide another mechanism to regulate ROR activity. A clear connection has been established between cholesterol intermediates/metabolites and their regulation of ROR activity. The first evidence for a link between RORs and sterols came from crystallography studies showing that cholesterol and cholesterol sulfate bind to the ligand-binding pocket of RORα and function as RORα agonists [7,43]. Subsequent studies demonstrated that several intermediates of the cholesterol biosynthesis pathway bind RORγ and enhance RORγ activity suggesting that they may function as endogenous ligands for RORγ [8,11] (Figure 1). These studies identified zymosterone, zymosterol, 7-dehydrocholesterol, and desmosterol among the most effective sterols activating RORγ, while lanosterol, T-MAS, and cholesterol, were weak agonists. Cholesterol and other sterols can be metabolized by a number of cytochrome P450 enzymes, including CYP27A1, CYP46A1, CYP7A1 and CYP7B1, that lead to formation of a wide range of oxysterols [44–46]. CYP7A1 catalyzes the formation of 7-keto- and 7α-hydroxysterols, while CYP27A1 catalyzes the formation of 25- and 27-hydroxysterols, and CYP46A1 the formation of hydroxysterols, such as 24(S)-hydroxycholesterol. The formation of 25-hydroxycholesterol can also be catalyzed by cholesterol 25-hydroxylase (Ch25h). A number of these oxysterols can act as ligands for RORα and RORγ [8–13,47–49]. These reports demonstrated that 7α- and 7β-hydroxycholesterol, 7-ketocholesterol, and 24S- and 24R-hydroxycholesterol, and 24,25-epoxycholesterol function as inverse agonists for RORα and/or RORγ, while 25-hydroxycholesterol, 27-hydroxycholesterol, 20α-hydroxycholesterol, 22R-hydroxycholesterol, 7α,27- and 7β,27-dihydroxycholesterol act as agonists of RORγ [2,10,45,50]. In addition to being hydroxylated, sterols can be sulfoconjugated by sulfotransferase SULT2B1 and generate sulfated sterols, such as cholesterol- and desmosterol sulfate, which function as RORα/γ agonists [7,8].
Figure 1. Intermediates of the cholesterol biosynthetic pathway function as RORγ agonists.
Shown is a schematic view of the Bloch cholesterol biosynthetic pathway. In peroxisomes, acetyl CoA is converted to mevalonate by HMGCR, a step that is inhibited by statins, and then via multiple steps into farnesyl pyrophosphate, which is then converted into squalene in the endoplasmic reticulum. This is subsequently converted into cholesterol via the steps indicated. Several cholesterol intermediates can act as RORγ agonists, but zymosterol and desmosterol exhibit the highest activity. Deficiency in Fdft1 or Cyp51A1, enzymes acting upstream in the cholesterol biosynthetic pathway, inhibit the synthesis of cholesterol and the availability of RORγ agonists. DHCR24 can convert 7-dehydrodesmosterol to 7-dehydrocholesterol, which acts as an agonist of RORγ. ACAT1, Acetyl-CoA Acetyltransferase 1; HMGCS1, 3-Hydroxy-3-Methylglutaryl-CoA Synthase 1; HMGCR, 3-Hydroxy-3-Methylglutaryl-CoA Reductase; FDT1, Farnesyl-Diphosphate Farnesyltransferase 1; SQLE, Squalene Epoxidase; LSS, Lanosterol Synthase; TM7SF2, Transmembrane 7 Superfamily Member 2; FAXDC2/SC4MOL, Fatty Acid Hydroxylase Domain Containing 2; NSDHL, NAD(P) Dependent Steroid Dehydrogenase-Like; HSD17B7, Hydroxysteroid 17-Beta Dehydrogenase 7; EBP, Emopamil Binding Protein; SC5D, Sterol-C5-Desaturase; DHCR7, 7-Dehydrocholesterol Reductase; DHCR24, 24-Dehydrocholesterol Reductase.
Triterpenes, which are produced by many plants, fruits, herbs, and fungi, are structurally closely related to sterols (Figure 2). Several triterpenes have been reported to bind RORγ and function as natural RORγ ligands. Among them, ursolic acid, which functions as an RORγ inverse agonist [51] and ganoderone A, an antiviral triterpenoid produced by the fungus Ganoderma pfeifferi, which was recently identified as a very potent RORγ agonist [16](Figure 2).
Figure 2.
Chemical structure of two sterol RORγ agonists, cholesterol sulfate, 25-hydroxycholesterol, and two triterpenes, ganoderone A and ursolic acid, which act as an RORγ agonist and RORγ inverse agonist, respectively.
As was demonstrated for the binding of cholesterol sulfate to the RORα(LBD) [7], X-ray crystal structure analysis confirmed the interaction of cholesterol sulfate and hydroxysterols with the ligand binding pocket of RORγ [9,16,52,53]. Studies examining the crystal structures of apo (unbound) RORγ(LBD) showed that, in the absence of ligand, RORγ is in an active conformation that is capable of recruiting co-activators [9,16,52]. Study of X-ray structures of agonist-bound RORγ(LBD) further revealed that binding of agonists stabilizes the active conformation of RORγ and its interaction with coactivators, whereas most inverse agonists do the opposite. The hydrogen bond between Hε2-His479 in helix 11 and OH-Tyr502 in helix 12 (referred to as His-Tyr lock) plays a critical role in stabilizing the active conformation of RORγt. These studies further indicated that this stabilization involves the formation of different bonds with RORγ [9,16,52]. For example, 25-hydroxycholesterol stabilizes H12 by interacting with Tyr502 indirectly via a water-mediated hydrogen bond with its 25-OH group leaving the His-Tyr lock in place, whereas ganoderone A holds helix 12 (H12) in the agonist position through the formation of two strong hydrogen bonds with His479 and Tyr502, thereby eliminating the His-Tyr lock. It is conceivable that such differences in agonist-RORγ interaction might bring about different changes in RORγ conformation that affect the specificity by which RORγ recruits co-activator complexes and activates target gene transcription. Consequently, this might affect the regulation of RORγ functions as well.
5. Vitamin D and lumisterol metabolites and RORs
Recently, several vitamin D3 and lumisterol metabolites were identified that function as weak RORα/γ inverse agonists [15,53–55]. Previtamin D3, and subsequently vitamin D3, are formed in the skin from 7-dehydrocholesterol upon photochemical transformation by UVB. In the classical pathway, CYP2R1 and CYP27B1 convert vitamin D3 to 1,25(OH)D3, the active form that binds the vitamin D receptor (VDR), but also acts as a weak inverse agonist of RORγ [11,15,53](Figure 3). In an alternative pathway in the skin, CYP11A1 catalyzes the formation of additional D3 metabolites, including 20(OH)D3, 22(OH)D3, 20,23(OH)D3, 20,22(OH)D3 and 17,20,23(OH)D3 [54,56]. Some of these metabolites are further hydroxylated by CYP27B1 at C1α, by CYP24A1 and CYP27A1 at C24, C25, and C26 [54,56,57]. Several of the D3 metabolites, including 20(OH)D3, 20,23(OH)2D3, 1,20,25(OH)3D3, 17,20,23(OH)3D3, 1,20(OH)2D3, and 20,26(OH)2D3 were able to inhibit RORE-mediated transactivation as well as the interaction between the RORα/γ LBD with an LXXLL coactivator peptide [11,15,53]. Molecular modeling using established crystal structures of RORα and RORγ LBDs, further showed that these inverse agonists exhibit high docking scores supporting interaction of these vitamin D3 metabolites with the ligand binding pocket of RORα/γ [15,53]. These findings suggest these noncalcemic D3 metabolites, which are produced endogenously, can act as RORα/γ inverse agonists and modulate their activity and functions. These studies indicate that, in addition to VDR, RORs may provide an alternative pathway by which D3 and its metabolites regulate physiological processes. Previtamin D3 can also undergo photoisomerization to lumisterol (L3) under the influence of UVB [56], which then can undergo sequential hydroxylations by CYP11A1 producing 20(OH)L3, 22(OH)L3, and 20,22(OH)2L3 [55](Figure 3). Cell-based and in vitro analyses demonstrated that several hydroxylumisterols exhibit inverse agonist activity, while molecular modeling supported the hypothesis that they act as ligands of RORα and RORγ [55].
Figure 3. Hydroxylated derivatives of vitamin D function as weak RORα/RORγ inverse agonists.
After absorption of UVB the RORγ agonist, 7-dehydrocholesterol, is phototransformed to vitamin D (D3) and lumisterol (L3). Vitamin D is activated through sequential hydroxylation at C25 and C1α to produce 1,25 (OH)2D3 that, in addition of acting on vitamin D receptor (VDR), also serves as an inverse agonist of RORs. Vitamin D3 and lumisterol can also be hydroxylated by CYP11A1 to produce hydroxylumisterols or hydroxyderivatives of vitamin D that can further be hydroxylated by other CYPs to produce hydroxyderivatives that can also act as inverse agonists of RORs. Weak inverse agonists shown in red; 7DHC, RORγ agonist 7-dehydrocholesterol.
6. Disruption of cholesterol biosynthesis and RORα/γ activity
The discovery that various sterols function as ligands, predicted that alterations in sterol homeostasis that change the intracellular pool of ROR (inverse) agonists, would affect ROR transcriptional activity and as a consequence the physiological as well as pathological processes in which RORs are implicated. This hypothesis was strongly supported by studies examining the effect of disruption of cholesterol biosynthesis on RORγt-dependent regulation of Th17 differentiation and gene expression.
RORγt is required for the differentiation of naïve T cells into Th17 cells and for the transcriptional activation of Il17A/F, IL21, and Il23R (Figure 4A). Interestingly, during Th17 differentiation, the expression of several cholesterol biosynthetic genes (e.g., FDFT1, LSS, and DHCR24) are enhanced, whereas that of genes involved in cholesterol metabolism (e.g., CYP7A1, CYP27A1) and efflux (e.g., ABCA1, ABCG1) are decreased [8]. In addition, Th17 differentiation is accompanied with an increase in the expression of the sulfotransferase SULT2B1 and a decrease in sterol sulfatase (STS) expression [8]. A positive correlation between IL-17 levels and the expression of several cholesterol biosynthesis-related genes was also revealed by transcriptome analysis of RNA isolated from normal and psoriatic skin biopsies [58]. Together, these findings are consistent with the concept that increased cholesterol biosynthetic gene expression would enhance the availability of RORγt agonists and subsequently stimulate RORγt activity, Th17 differentiation, and IL-17 production, and consequently promote inflammatory disease.
Figure 4. Changes in cholesterol/sterol homeostasis alter RORγ transcriptional activity and subsequently the regulation of gene expression, physiological functions and disease, in which RORγ is involved.
A. The formation of RORγt agonists by the cholesterol biosynthetic pathway leads to activation of RORγt, which is required for Th17 differentiation and the expression of target genes, such as Il17, Il21, and Il23R. The HMG-CoA reductase inhibitors, statins, inhibit the generation of RORγt agonists resulting in reduced RORγt activation and Th17 differentiation. Statins also reduce RORγt expression and Th17 differentiation by inhibiting STAT3, which is required for RORγt transcription. The scavenger receptor CD5L is expressed at high levels in nonpathogenic Th17 cells, where it suppresses cholesterol biosynthesis and the formation of RORγt agonists. It becomes expressed at low levels in pathogenic Th17 cells leading to increased cholesterol biosynthesis, generation of RORγt agonists, and pathogenicity of Th17 cells. B. Inhibition of CYP51A1 by azoles, deficiency in CYP51A1 or FDFT1 inhibit cholesterol biosynthesis, thereby reducing the availability of RORγ agonists and RORγt activation. Statins and CD5L have a negative effect on cholesterol biosynthesis and RORγt activation. This subsequently reduces Th17 differentiation and Il17 expression, and suppresses the development of inflammatory disease. Other mechanisms that affects the generation of RORγt agonists are CYP27A1 deficiency, which reduces the generation of 27-hydroxycholesterol. Induction of ABCG1/A1 expression by LXRs increase cholesterol efflux and potentially reduce the availability of RORγt agonists and Il17 expression, whereas ApoE-deficiency and high fat/cholesterol diet (HFD) have the opposite effect by increasing cholesterol levels.
Several studies have demonstrated that disruption of the cholesterol biosynthesis pathway has a negative effect on RORγt activation and Th17 differentiation (Figure 4A and B). The cholesterol-lowering drugs, statins, bind to and inhibit 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase (HMGCR), which catalyzes the conversion of HMG-CoA to mevalonic acid, one of the first steps in cholesterol biosynthesis. Statins have been shown to inhibit Th17 differentiation and the expression of inflammatory cytokines [59]. This appears to involve several mechanisms including up-regulation of suppressor of cytokine secretion (SOCS3 and -7) and reduced activation of STAT3 and RORγt expression. In addition, the reduction in cholesterol biosynthesis by statins might diminish the generation of RORγt agonists, thereby reducing RORγt activation, RORγt-mediated Th17 differentiation, and activation of Il17 (Figure 4A). The formation of squalene by squalene synthase (Fdft1) is another early step in the cholesterol biosynthetic pathway. A study examining the effect of disruption of squalene synthesis on RORγt activation demonstrated that the RORγ-mediated transactivation of a RORE-driven reporter was greatly reduced in a squalene synthase (Fdft1)-deficient cell line, SXLT, while exogenous expression of Fdft1 or the addition of squalene restored RORγt-mediated transactivation of the reporter [11] (Figure 4B).
Disruption of 14α-demethylase cytochrome P450, Cyp51a1 (lanosterol 14α-demethylase), either in Cyp51a1-deficient mice or in cells treated with Cyp51a1 inhibitors, such as azole-type fungicides, resulted in decreased synthesis of zymosterol and desmosterol and a significant reduction in RORγ-mediated transactivation and consequently in Th17 differentiation and IL-17 expression [8,11,60](Figure 4B). Consistent with this are similarities in the phenotype between Cyp51a1- and RORγ-deficient mice. Branchial lymph node anlagen were absent in 75% of Cyp51a1−/− mice and the number of IL17RA+ and CD4+ Lti cells was reduced. A recent study of a hepatocyte-specific knockout of Cyp51a1 in mice, referred to as HCyp51−/−, showed development of progressive liver injury and fibrosis [61]. Hepatic cholesterol biosynthesis was disrupted in these mice leading to lower levels of hepatic and blood cholesterol, as well as several cholesterol intermediates, including zymosterol and desmosterol, which act as ROR agonists. This reduced agonist levels are likely responsible for the observed reduction in RORα and RORγ transcriptional activity. In addition, in some of alterations in hepatic gene expression in HCyp51−/− mice overlapped with ROR target genes consistent with the concept that some of these changes are due to reduced RORα/γ activation.
7. Regulation of RORγ activity by other sterol metabolic pathways
Cytochrome P450 enzymes play a key role in the regulation of various aspects of cholesterol homeostasis and many catalyze the formation of oxysterols, several of which act as (inverse) antagonists of RORγt. CYP27A1 catalyzes the formation of 27-hydroxysterols, including 27-hydroxycholesterol, 7α, 27- and 7β, 27-dihydroxycholesterol. These hydroxysterols, which were reported to be preferentially produced by mouse Th17 cells, all act as RORγt agonists and stimulate Th17 differentiation [10]. The production of 27-hydroxycholesterols in Cyp27a1-deficient Th17 cells was greatly diminished as was the number of IL-17 producing cells in Cyp27a1-deficient mice suggesting that 27-hydroxysterols are an important source of RORγt agonists in these cells (Figure 4B). However, these findings appear at odds with another study showing that enzymes involved in oxysterol formation are expressed at low levels during Th17 differentiation [8].
One might expect that hypercholesterolemic conditions lead to an increase in endogenous sterol levels and availability of ROR agonists. This would subsequently results in increased activation of RORγt and promote, for example, Th17 differentiation and the production of the inflammatory cytokine IL-17A. Low cholesterol diet might do the opposite. This hypothesis is supported by several reports. Apolipoprotein E (ApoE)-deficient mice have increased levels of total cholesterol and LDL-cholesterol. These mice spontaneously develop atherosclerosis and show an increased susceptibility to type II collagen-induced arthritis [62]. Since Th17 cells and the production of inflammatory cytokines, such as IL17A, play a critical role in the development of these pathologies, the increased susceptibility to these diseases might be in part related to the elevated blood levels of cholesterol that lead to an increase in RORγ agonists and Th17 cells [62](Figure 4B). A different study reported that patients with chronic hepatitis C, when placed on a normocaloric, low cholesterol diet showed a significant reduction in Th17 cells and IL-17 levels, which might at least in part be related to reduced RORγt activation [63].
CD5-like (CD5L/AIM), a member of the scavenger receptor cysteine-rich superfamily, is highly expressed in non-pathogenic Th17 cells, where it regulates lipid synthesis and modulates intracellular lipid homeostasis, but is down-regulated in pathogenic Th17 cells (Fig. 4A)[64]. CD5L reduces the content of polyunsaturated fatty acid (PUFA) in favor of saturated fatty acids (SFA) and limits cholesterol biosynthesis. The low expression of CD5L in pathogenic Th17 cells increases cholesterol biosynthesis and the generation of RORγt agonists, and promotes their pathogenicity. Similarly, deficiency in CD5L in mice leads to increased PUFA content and expression of several cholesterol biosynthetic enzymes, such as Cyp51a1 and Sc4mol, in Th17 cells leading to increased availability of RORγt agonists (Figure 4B). Consequently, this leads to RORγt-dependent transactivation of Il17 and Il23r promoters, thereby promoting a shift from non-pathogenic to pathogenic Th17 cells.
RORα/γ activity might be influenced by a number of other cholesterol regulatory pathways, including sterol regulatory element binding protein 2 (SREBP2) and the LXR nuclear receptors [45,65–67]. The transcription factor SREBP2 regulates cholesterol biosynthesis in part by increasing the transcription of several genes involved in cholesterol biosynthesis, including HMGCR and HMGCS, thereby enhancing the availability of RORγ agonists [66,67]. Activation of the T cell receptor (TCR) pathway results in activation of SREBP in favor of sterol-sulfate and cholesterol synthesis and might synergize with RORγ in promoting Th17 differentiation and IL-17 synthesis [8,68]. LXRs play a critical role in cholesterol homeostasis by regulating the expression of ABCA1 and ABCG1, which mediate cholesterol efflux [45,65]. Thus, LXR agonists reduce cholesterol levels in cells thereby reducing the availability of RORα/γ agonists (Figure 4B).
8. Regulation of sterol metabolism by RORs
In addition to being regulated by sterols, RORs themselves regulate the expression of a number of genes involved in cholesterol transport and sterol metabolism. The hepatic expression of ABCA1 and ABCG1, encoding reverse cholesterol transporters, is reduced in the liver of RORα-deficient mice [69]. In HUVEC endothelial cells and the monocytic cell line, THP-1, ABCA1 was found to be directly regulated by RORα [70]. Treatment with an inverse RORα agonist was shown to decrease plasma cholesterol levels and suppress atherosclerosis development in mice, which appeared in part to be related to increased ABCG5/G8 expression and cholesterol excretion by the intestine [71]. The hepatic expression of sterol 12α-hydroxylase, CYP8B1, and 7α hydroxylase, CYP7B1, which are involved in the conversion of cholesterol into bile acids and the regulation of hepatic and serum cholesterol levels, have been reported to be directly regulated by RORα and/or RORγ [22,25,72]. RORα regulates CYP8B1 by directly binding to an RORE in its promoter and through the recruitment of the co-activator, CBP [72].
In addition, RORα/γ have been implicated in the circadian regulation of several 3β-hydroxysteroid dehydrogenases and sterol sulfotransferases [22,25]. RORα/γ positively regulate the hepatic expression of the hydroxysteroid sulfotransferase, Sult1B1, and steroid/bile acid sulfotransferase, Sult2A1 [22,73,74]. Sult2A1 transcription was shown to be regulated directly by RORα and RORγ [73]. In macrophages, RORα regulates cholesterol 25-hydroxylase (Ch25h), which converts cholesterol to 25-hydroxycholesterol, and promotes phagocytosis [50]. Regulation of sterol metabolic genes by RORs may enhance or reduce the synthesis of agonists or inverse agonist thereby modulating RORγ activity, thereby creating a positive or negative feedback loop. Whether the regulation of sterol metabolic genes by RORs affects the availability of ROR (inverse) agonists and therefore ROR activation, needs further study.
9. Synthetic RORγ ligands
Genetic deficiency of RORγt in mice greatly affects Th17 cell differentiation and Il17 expression and protects mice against Th17-dependent inflammation in several experimental autoimmune disease models [75–78]. This, together with the discovery that RORs act as ligand-dependent transcription factors, prompted a search for synthetic ROR inverse agonists with therapeutic potential. Screens of chemical libraries for ROR modulators identified several series of synthetic small molecular weight molecules that function either as an agonist or inverse agonist [2,17,48,79–91]. Recently, a synthetic RORγ agonist, SR0987, was also described [92]. Chemical structures of several synthetic ROR inverse agonists are shown in Figure 5. For a more comprehensive overview, we refer to several recent reviews [2,4,12–14].
Figure 5. Chemical structures of several RORγ inverse agonists.
SR1001 (Scripps Institute) is a dual RORα/RORγ inverse agonist, while all others are RORγ-selective inverse agonists.
X-ray crystallography of the RORγ(LBD) in complex with various inverse agonists suggested the existence of distinct classes [16,93,94]. Most inverse agonists bind the canonical ligand binding pocket of RORγ; however, these interactions can be distinguished on basis of the formation of different bonds with RORγ. In one class, binding of the inverse agonist causes repositioning of H12 that disrupts interaction with co-activators. Binding of a second class of inverse agonists involves a “water trapping” mechanism, in which H12 remains in the active conformation and still allows interaction with co-activator peptides [16]. A different study revealed still another type of antagonism [93]. Indazole-type inverse agonists, such as MRL-871, do not interact with the canonical, orthosteric ligand binding pocket, but with a different, distal, allosteric binding site in RORγ(LBD). MRL-871 reorients H12 and actively blocks binding of cofactors. Molecular studies revealed two additional modes of action by which inverse agonists can inhibit RORγ transcriptional activity. The inverse agonists TMP778, TMP920, and GSK805 interact physically with the canonical ligand-binding pocket of RORγt and inhibit Th17 differentiation; however, interaction with TMP920 disrupts RORγt binding to DNA, whereas TMP778- and GSK805-bound RORγt are still associated with target genes and actually stabilized RORγt binding to a number of new genomic sites, suggesting that RORγ binding to DNA remains largely intact [17]. Similarly, the inverse agonist Cpd1 did not affect the binding of RORγ to ROREs (Guntermann, 2017 #1276}. Binding of RORγt to the Il17a and Il23r promoter was found to be associated with a more open chromatin structure as indicated by H3AcK9/K14 acetylation and H3K4me3 methylation at these loci. This histone methylation and acetylation is downregulated in the presence of Cpd1 suggesting a less open chromatin structure.
10. RORγ as therapeutic target
Treatment of cells and rodents with synthetic RORγ inverse agonists induces many of the same effects as observed in RORγ null mice, including inhibition of Th17 differentiation, RORE-mediated transactivation, activation of the Il17 transcription, and IL-17 production [2,4,17,19,20,95–98]. Moreover, as observed in RORγ null mice, RORγ inverse agonists greatly alleviate Th17-dependent inflammation in several experimental autoimmune disease models in rodents [17,51,76,89,96,98–102]. The dual RORα/RORγ inverse agonist SR1001 and the RORγ-selective inverse agonists, ursolic acid, digoxin, MRL-248, TMP778, TMP920, have been reported to ameliorate Th17-mediated experimental autoimmune encephalomyelitis (EAE) in mice and rats [17,51,103–106]. Treatment with inverse RORγ agonists Bio-0554019, GSK2981278, TMP778, VTP-43742, JNJ-54271074 attenuated skin inflammation in imiquimod-induced psoriatic model as well as in other psoriatic models, the IL-23-injection model and K5.Stat3C transgenic mice [98–100,105,107–109]. Administration of the inverse RORα/RORγ inverse agonist SR1001 to NOD mice, a model of type 1 diabetes, significantly reduced diabetes incidence and insulitis [101], while treatment of obese diabetic mice with SR1555 caused a significant reduction in fat mass and improved insulin sensitivity [110]. A different study showed that Cpd1 reduced hypersensitivity responses in a methylated bovine serum albumin–induced skin inflammation model in rats [102]. The inverse agonists, JNJ-54271074 and SR2211, have been reported to inhibit inflammation in a collagen-induced arthritis mouse model [89,99] and Cpd1 in antigen-induced arthritis in rats [102]. In an experimental murine model, treatment with TMP778 markedly alleviated chronic graft-versus-host disease (cGvHD), which represents a major complication of allogeneic stem cell transplantation, and may provide a new therapeutic approach in the management of cGvHD [111].
It is to be expected that treatment with RORγ inverse agonists would also affect the function of RORγ in other RORγ+ immune cells, including DP thymocytes, ILC3s, RORt+Tregs, and RORt+γδ. Treatment of mouse thymocytes with the inverse agonists was shown to down-regulate Bcl2l1 expression and to increase apoptosis, while treatment of mice or rats with, respectively, the RORγ inverse agonists MRL-248 or Cpd1, causes a reduction in the number of total thymocytes and perturbs DP thymocyte survival [97,105], as observed in RORγ null mice [19,20]. Moreover, administration of MRL-248 in mice resulted in skewed cell receptor alpha (TCRα) gene rearrangements and limited T cell repertoire diversity [105]. In a myelin oligodendrocyte glycoprotein (MOG)-induced EAE model, MRL-248 reduced the number of MOG-specific CD4+ T cells and delayed autoimmune EAE progression. Although ILC3s can be pro-inflammatory, RORγt+ ILC3s play a critical role in intestinal immunity and protect against infection-mediated inflammation [112]. In response to infection with Citrobacter rodentium, Th17 cell responses and intestinal inflammation were significantly inhibited in mice treated with the inverse agonist GSK805, while cytokine expression in ILC3s, including IL-17, was largely maintained [113]. Thus RORγ inverse agonists might provide an attractive strategy to treat inflammatory bowel disease.
The discovery that treatment with small molecular weight RORγ inverse agonists protects against autoimmune disease in several experimental rodent models initiated a number of clinical trials [109,114,115]. Vitae Pharmaceuticals/Allergan successfully finished its Phase 1 trial with VTP-43742 (Figure 5) and a Phase 2a trial showed statistical significant efficacy in psoriatic patients [115]. These studies suggest that RORγt inverse agonist might provide an attractive therapeutic alternative in the management of inflammatory autoimmune disease.
11. Lymphoma concerns
Ueda et al. [116] reported that RORγ-deficient mice develop thymic lymphomas within three months of age that was confirmed in a later study [117]. Knockout of RORγ in adult mice using conditional RORγ knockout mice also leads to thymic lymphoma development; however, in mice, in which the Rorc locus was incompletely deleted, RORγ-deficient thymocytes were rapidly replaced by wild type thymocytes [117]. These findings raised concerns whether therapeutic treatment with RORγ antagonists increases the risk of developing T cell lymphoma. Chronic treatment of rats with the RORγ inverse agonist Cpd1 was recently shown to cause progressive thymic changes that resemble those observed in RORγ-deficient mice [97]. This study further showed that one rat developed preneoplastic, hypertrophic lesions in the cortical thymus. Although this may not occur in humans, these studies point at an apparent risk of T cell lymphoma with chronic RORγ inverse agonist treatment at least in rodents.
Although these concerns may be real, the observed diversity in modes of interaction of inverse agonists with RORγ(LBD) provides hope that future studies might be able to identify inverse agonists that target certain biological processes more specifically thereby minimizing possible side effects of therapeutic treatment [16,17,93]. A recent study [42] showing that a two-amino-acid substitution in RORγt disrupts Th17 differentiation, but not thymocyte development, is consistent with this prospect.
12. Further considerations
Multiple T helper cell lineages and immune cell types contribute to immunopathology. As mentioned above RORγt is expressed in a number of immune cell subtypes with pathogenic as well as protective functions, including Th17, RORt+Tregs, RORt+γδ, and ILC3s. For example, Th17 cells have a pro-inflammatory role in the development of autoimmune disease, but are also key players in host defense against extracellular pathogens [78,118]. In the gut, a delicate balance exists between anti- and pro-inflammatory mechanisms that maintain the epithelial barrier integrity and control intestinal homeostasis and inflammation. This involves complex, mutual interactions between the gut microbiota and different immune cells, such as Th17, RORt+Tregs, RORt+γδ, and ILC3s (reviewed in [31,35,112,119]). How RORγt inverse agonists affect these interactions is not yet fully understood.
Adding to this complexity is that microbiota, metabolism, and immunity, are also regulated by the circadian clock [120–122] and in several tissues ROR expression is under the control of the circadian clock and expressed in a rhythmic pattern [4,123]. In addition, RORs themselves participate in the regulation of circadian clock genes [4,123–125]. Together, these findings suggest that timing of administering inverse agonists to treat autoimmune disease could be important and that the greatest therapeutic benefits might be obtained at the circadian time of optimal RORγt expression.
One also needs to take into account that changes in sterol homeostasis influences the activity of many other signaling pathways [45,66]. Cholesterol is critical for membrane fluidity of the plasma membrane and can interact with and regulate the activity of many other proteins, including G protein-coupled receptors, such as GPR183 [126] and Smoothened (SMO), a component of Hedgehog signaling [127], and several other nuclear receptors, such as LXRs [45,128]. Several (inverse) agonists have been reported to interact with both LXRs and RORs. It is therefore likely that the regulation of physiological processes by RORs intersects with many of the other cholesterol/sterol mediated regulatory signaling pathways.
13. Concluding remarks
An increasing number of studies have demonstrated that RORγt has multiple functions in immunity and immunopathology. Autoimmune disease is a complex disease in which metabolism, the microbiome, circadian clock, and multiple cell immune cells play mutually dependent roles. Further studies are needed to understand the precise molecular mechanisms underlying these interrelationships and how loss of RORγt function affects these interactions and immunopathologies. The discovery that loss of RORγt function protects against autoimmune disease and the identification of synthetic RORγ inverse agonists that protect against autoimmune disease in several experimental rodent models has made RORγ a conceivable therapeutic target. Successful phase 1 and phase 2 clinical trials suggest that RORγ inverse agonists may offer an attractive alternative therapy for autoimmune disease. To what extent the risk of developing lymphoma is a concern needs further study. Discovery of inverse agonists that target RORγt-regulated physiological processes more specifically may get around certain side effects. Like RORγt, RORα is also expressed in a number of immune cells, including Th17 and ILC2s; however, its role in immunity and immunopathology is less well understood. Identification of high affinity RORα (inverse) agonists will not only increase our understanding of the role of RORα in immunity and immune disease, but may also uncover its therapeutic potential.
Acknowledgments
The author would like to thank Drs. David Scoville and Caleb Sutherland for their comments on the manuscript. AMJ research was supported by the Intramural Research Program of the NIEHS, NIH (Z01-ES-101585) and partially by NIH grants R21AR066505 and 1R01AR071189–01A1 to AS.
Footnotes
Conflict of interest statement
Nothing declared
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Jetten AM. Retinoid-related orphan receptors (RORs): critical roles in development, immunity, circadian rhythm, and cellular metabolism. Nucl Recept Signal. 2009;7:e003. doi: 10.1621/nrs.07003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Solt LA, Burris TP. Action of RORs and their ligands in (patho)physiology. Trends Endocrinol Metab. 2012;23:619–627. doi: 10.1016/j.tem.2012.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Jetten AM, Kang HS, Takeda Y. Retinoic acid-related orphan receptors alpha and gamma: key regulators of lipid/glucose metabolism, inflammation, and insulin sensitivity. Front Endocrinol (Lausanne) 2013;4:1. doi: 10.3389/fendo.2013.00001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Cook DN, Kang HS, Jetten AM. Retinoic Acid-Related Orphan Receptors (RORs): Regulatory Functions in Immunity, Development, Circadian Rhythm, and Metabolism. Nucl Receptor Res. 2015;2 doi: 10.11131/2015/101185. pii: 101185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Giguere V, McBroom LD, Flock G. Determinants of target gene specificity for ROR alpha 1: monomeric DNA binding by an orphan nuclear receptor. Mol Cell Biol. 1995;15:2517–2526. doi: 10.1128/mcb.15.5.2517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Medvedev A, Yan ZH, Hirose T, Giguere V, Jetten AM. Cloning of a cDNA encoding the murine orphan receptor RZR/ROR gamma and characterization of its response element. Gene. 1996;181:199–206. doi: 10.1016/s0378-1119(96)00504-5. This study, together with a previous report by the same laboratory, describe the discovery of the mouse and human homologs of the third member of the retinoic acid-related orphan receptor subfamily, RORγ. The amino acid sequence of mouse RORγ exhibits an 88% overall identity to that of human RORγ. Analysis of the ROR gamma-response element (RORE) revealed that it binds as a monomer to response elements composed of a single core motif GGTCA preceded by a 6 bp AT-rich sequence. [DOI] [PubMed] [Google Scholar]
- 7.Kallen J, Schlaeppi JM, Bitsch F, Delhon I, Fournier B. Crystal structure of the human RORalpha Ligand binding domain in complex with cholesterol sulfate at 2. 2 A. J Biol Chem. 2004;279:14033–14038. doi: 10.1074/jbc.M400302200. In this study, the X-ray crystal structures of eight different classes natural and synthetic (inverse) agonists are analyzed. Several new molecular mechanisms for RORγt agonism and inverse agonism were proposed. One class of inverse agonists acts via steric clash that prevents co-activator binding and the other involving “water trapping”. These new mechanistic insights should prove useful for the design and optimization of further RORγt modulators. [DOI] [PubMed] [Google Scholar]
- 8.Hu X, Wang Y, Hao L-Y, Liu X, Lesch CA, et al. Sterol metabolism controls T(H)17 differentiation by generating endogenous RORγ agonists. Nature Chem Biol. 2015;11:141–147. doi: 10.1038/nchembio.1714. This report shows that during Th17 differentiation, cholesterol biosynthesis and uptake programs are induced, whereas their metabolism and efflux programs are suppressed, which result in increased accumulation of the cholesterol precursor, desmosterol, a potent endogenous RORγ agonist. Blocking cholesterol biosynthesis with inhibitors inhibits Th17 differentiation. The study shows that sterol synthesis, mobilization and metabolism play a critical role in the regulation of Th17 differentiation by regulating RORγ activation. [DOI] [PubMed] [Google Scholar]
- 9.Jin L, Martynowski D, Zheng S, Wada T, Xie W, et al. Structural basis for hydroxycholesterols as natural ligands of orphan nuclear receptor RORgamma. Mol Endocrinol. 2010;24:923–929. doi: 10.1210/me.2009-0507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Soroosh P, Wu J, Xue X, Song J, Sutton SW, et al. Oxysterols are agonist ligands of RORgammat and drive Th17 cell differentiation. Proc Natl Acad Sci U S A. 2014;111:12163–12168. doi: 10.1073/pnas.1322807111. Oxysterols elicit profound effects on immune and inflammatory responses. This study demonstrates that several naturally occurring oxysterols act as RORγt agonists by binding directly to recombinant RORγ ligand binding domain and promoting recruitment of a coactivators. The most potent and selective activator for RORγt, 7β, 27-dihydroxycholesterol, enhances the Th17 differentiation of murine and IL-17 production. Mice deficient in CYP27A1, a key enzyme in generating these oxysterols, showed significant reduction of IL-17-producing cells. CYP27A1 is shown to be important for the generation of these RORγt agonists and adaptive IL-17-dependent immune responses. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Santori FR, Huang P, van de Pavert SA, Douglass EF, Jr, Leaver DJ, et al. Identification of Natural RORgamma Ligands that Regulate the Development of Lymphoid Cells. Cell Metab. 2015;21:286–297. doi: 10.1016/j.cmet.2015.01.004. RORγt-deficient mice exhibit defective development of thymocytes, lymphoid organs, Th17 cells, and type 3 innate lymphoid cells. This study shows that several intermediates of the cholesterol biosynthesis, such as desmosterol and zymosterol, enhance RORγt activation and RORγ-dependent transcription. The results suggest that they function as RORγt ligands. They stabilized the RORγ ligand-binding domain and induced coactivator recruitment. The report suggests that these intermediates play a role in regulating lymphocyte development by binding RORγt. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Fauber BP, Magnuson S. Modulators of the nuclear receptor retinoic acid receptor-related orphan receptor-gamma (RORgamma or RORc) J Med Chem. 2014;57:5871–5892. doi: 10.1021/jm401901d. [DOI] [PubMed] [Google Scholar]
- 13.Kojetin DJ, Burris TP. REV-ERB and ROR nuclear receptors as drug targets. Nat Rev Drug Discov. 2014;13:197–216. doi: 10.1038/nrd4100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Cyr P, Bronner SM, Crawford JJ. Recent progress on nuclear receptor RORgamma modulators. Bioorg Med Chem Lett. 2016;26:4387–4393. doi: 10.1016/j.bmcl.2016.08.012. [DOI] [PubMed] [Google Scholar]
- 15.Slominski AT, Kim TK, Takeda Y, Janjetovic Z, Brozyna AA, et al. RORalpha and ROR gamma are expressed in human skin and serve as receptors for endogenously produced noncalcemic 20-hydroxy- and 20,23-dihydroxyvitamin D. FASEB J. 2014;28:2775–2789. doi: 10.1096/fj.13-242040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Kallen J, Izaac A, Be C, Arista L, Orain D, et al. Structural States of RORgammat: X-ray Elucidation of Molecular Mechanisms and Binding Interactions for Natural and Synthetic Compounds. ChemMedChem. 2017;12:1014–1021. doi: 10.1002/cmdc.201700278. [DOI] [PubMed] [Google Scholar]
- 17.Xiao S, Yosef N, Yang J, Wang Y, Zhou L, et al. Small-molecule RORgammat antagonists inhibit T helper 17 cell transcriptional network by divergent mechanisms. Immunity. 2014;40:477–489. doi: 10.1016/j.immuni.2014.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Giguere V, Tini M, Flock G, Ong E, Evans RM, et al. Isoform-specific amino-terminal domains dictate DNA-binding properties of ROR alpha, a novel family of orphan hormone nuclear receptors. Genes & Dev. 1994;8:538–553. doi: 10.1101/gad.8.5.538. [DOI] [PubMed] [Google Scholar]
- 19.Sun Z, Unutmaz D, Zou YR, Sunshine MJ, Pierani A, et al. Requirement for RORgamma in thymocyte survival and lymphoid organ development. Science. 2000;288:2369–2373. doi: 10.1126/science.288.5475.2369. [DOI] [PubMed] [Google Scholar]
- 20.Kurebayashi S, Ueda E, Sakaue M, Patel DD, Medvedev A, et al. Retinoid-related orphan receptor gamma (RORgamma) is essential for lymphoid organogenesis and controls apoptosis during thymopoiesis. Proc Natl Acad Sci USA. 2000;97:10132–10137. doi: 10.1073/pnas.97.18.10132. This study, together with that of Sun, Z., et al. (PNAS, 2000), identified for the time several immune functions of RORγ by generating and characterizing mice deficient in RORγ. RORγ-deficient mice lack peripheral and mesenteric lymph nodes and Peyer’s patches, indicating that RORγ expression is indispensable for lymph node organogenesis. Thymopoiesis is greatly impaired in RORγ-deficient mice due to accelerated apoptosis of the CD4(+)CD8(+) subpopulation. This was at least in part related to the greatly reduced level of expression of the anti-apoptotic gene Bcl-X(L). These two studies showed that RORγ is an critical regulator of several immune cells. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Hirose T, Smith RJ, Jetten AM. ROR gamma: the third member of ROR/RZR orphan receptor subfamily that is highly expressed in skeletal muscle. Biochem Biophys Res Commun. 1994;205:1976–1983. doi: 10.1006/bbrc.1994.2902. This is the first study describing the discovery of the RORγ gene. [DOI] [PubMed] [Google Scholar]
- 22.Kang HS, Angers M, Beak JY, Wu X, Gimble JM, et al. Gene expression profiling reveals a regulatory role for ROR alpha and ROR gamma in phase I and phase II metabolism. Physiol Genomics. 2007;31:281–294. doi: 10.1152/physiolgenomics.00098.2007. [DOI] [PubMed] [Google Scholar]
- 23.Schmidt SF, Madsen JG, Frafjord KO, Poulsen L, Salo S, et al. Integrative Genomics Outlines a Biphasic Glucose Response and a ChREBP-RORgamma Axis Regulating Proliferation in beta Cells. Cell Rep. 2016;16:2359–2372. doi: 10.1016/j.celrep.2016.07.063. [DOI] [PubMed] [Google Scholar]
- 24.Takeda Y, Kang HS, Freudenberg J, DeGraff LM, Jothi R, et al. Retinoic acid-related orphan receptor gamma (RORgamma): a novel participant in the diurnal regulation of hepatic gluconeogenesis and insulin sensitivity. PLoS Genet. 2014;10:e1004331. doi: 10.1371/journal.pgen.1004331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Takeda Y, Kang HS, Lih FB, Jiang H, Blaner WS, et al. Retinoid acid-related orphan receptor gamma, RORgamma, participates in diurnal transcriptional regulation of lipid metabolic genes. Nucleic Acids Res. 2014;42:10448–10459. doi: 10.1093/nar/gku766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Montaldo E, Juelke K, Romagnani C. Group 3 innate lymphoid cells (ILC3s): Origin, differentiation, and plasticity in humans and mice. Eur J Immunol. 2015;45:2171–2182. doi: 10.1002/eji.201545598. [DOI] [PubMed] [Google Scholar]
- 27.Eberl G, Littman DR. The role of the nuclear hormone receptor RORgammat in the development of lymph nodes and Peyer’s patches. Immunol Rev. 2003;195:81–90. doi: 10.1034/j.1600-065x.2003.00074.x. [DOI] [PubMed] [Google Scholar]
- 28.Ivanov II, Zhou L, Littman DR. Transcriptional regulation of Th17 cell differentiation. Semin Immunol. 2007;19:409–417. doi: 10.1016/j.smim.2007.10.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Bar-Ephraim YE, Mebius RE. Innate lymphoid cells in secondary lymphoid organs. Immunol Rev. 2016;271:185–199. doi: 10.1111/imr.12407. [DOI] [PubMed] [Google Scholar]
- 30.Melo-Gonzalez F, Hepworth MR. Functional and phenotypic heterogeneity of group 3 innate lymphoid cells. Immunology. 2017;150:265–275. doi: 10.1111/imm.12697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Ohnmacht C. Tolerance to the Intestinal Microbiota Mediated by ROR(gammat)(+) Cells. Trends Immunol. 2016;37:477–486. doi: 10.1016/j.it.2016.05.002. [DOI] [PubMed] [Google Scholar]
- 32.Ohnmacht C, Park JH, Cording S, Wing JB, Atarashi K, et al. The microbiota regulates type 2 immunity through RORgammat(+) T cells. Science. 2015;349:989–993. doi: 10.1126/science.aac4263. Together with Sefik et al. (Ref. 33) showed the complex interrelationships that exist between microbiota homeostasis, different RORγ-positive immune cell populations in the gut, host defence, and inflammatiry responses. [DOI] [PubMed] [Google Scholar]
- 33.Sefik E, Geva-Zatorsky N, Oh S, Konnikova L, Zemmour D, et al. Individual intestinal symbionts induce a distinct population of RORgamma(+) regulatory T cells. Science. 2015;349:993–997. doi: 10.1126/science.aaa9420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Cua DJ, Tato CM. Innate IL-17-producing cells: the sentinels of the immune system. Nat Rev Immunol. 2010;10:479–489. doi: 10.1038/nri2800. [DOI] [PubMed] [Google Scholar]
- 35.Burkett PR, Meyer zu Horste G, Kuchroo VK. Pouring fuel on the fire: Th17 cells, the environment, and autoimmunity. J Clin Invest. 2018 doi: 10.1172/JCI78085. In Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kim BS, Lu H, Ichiyama K, Chen X, Zhang YB, et al. Generation of RORgammat(+) Antigen-Specific T Regulatory 17 Cells from Foxp3(+) Precursors in Autoimmunity. Cell Rep. 2017;21:195–207. doi: 10.1016/j.celrep.2017.09.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Rutz S, Eidenschenk C, Kiefer JR, Ouyang W. Post-translational regulation of RORgammat-A therapeutic target for the modulation of interleukin-17-mediated responses in autoimmune diseases. Cytokine Growth Factor Rev. 2016;30:1–17. doi: 10.1016/j.cytogfr.2016.07.004. [DOI] [PubMed] [Google Scholar]
- 38.Ermisch M, Firla B, Steinhilber D. Protein kinase A activates and phosphorylates RORalpha4 in vitro and takes part in RORalpha activation by CaMK-IV. Biochem Biophys Res Commun. 2011;408:442–446. doi: 10.1016/j.bbrc.2011.04.046. [DOI] [PubMed] [Google Scholar]
- 39.Hwang EJ, Lee JM, Jeong J, Park JH, Yang Y, et al. SUMOylation of RORalpha potentiates transcriptional activation function. Biochem Biophys Res Commun. 2009;378:513–517. doi: 10.1016/j.bbrc.2008.11.072. [DOI] [PubMed] [Google Scholar]
- 40.Lim HW, Kang SG, Ryu JK, Schilling B, Fei M, et al. SIRT1 deacetylates RORgammat and enhances Th17 cell generation. J Exp Med. 2015;212:607–617. doi: 10.1084/jem.20132378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kathania M, Khare P, Zeng M, Cantarel B, Zhang H, et al. Itch inhibits IL-17-mediated colon inflammation and tumorigenesis by ROR-gammat ubiquitination. Nat Immunol. 2016;17:997–1004. doi: 10.1038/ni.3488. [DOI] [PubMed] [Google Scholar]
- 42.He Z, Ma J, Wang R, Zhang J, Huang Z, et al. A two-amino-acid substitution in the transcription factor RORgammat disrupts its function in TH17 differentiation but not in thymocyte development. Nat Immunol. 2017;18:1128–1138. doi: 10.1038/ni.3832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Kallen JA, Schlaeppi JM, Bitsch F, Geisse S, Geiser M, et al. X-ray structure of the hRORalpha LBD at 1. 63 A: structural and functional data that cholesterol or a cholesterol derivative is the natural ligand of RORalpha. Structure. 2002;10:1697–1707. doi: 10.1016/s0969-2126(02)00912-7. In this study, the X-ray crystal structures of eight different classes natural and synthetic (inverse) agonists are analyzed. Several new molecular mechanisms for RORγt agonism and inverse agonism were proposed. One class of inverse agonists acts via steric clash that prevents co-activator binding and the other involving “water trapping”. These new mechanistic insights should prove useful for the design and optimization of further RORγt modulators. [DOI] [PubMed] [Google Scholar]
- 44.Guillemot-Legris O, Mutemberezi V, Muccioli GG. Oxysterols in Metabolic Syndrome: From Bystander Molecules to Bioactive Lipids. Trends Mol Med. 2016;22:594–614. doi: 10.1016/j.molmed.2016.05.006. [DOI] [PubMed] [Google Scholar]
- 45.Mutemberezi V, Guillemot-Legris O, Muccioli GG. Oxysterols: From cholesterol metabolites to key mediators. Prog Lipid Res. 2016;64:152–169. doi: 10.1016/j.plipres.2016.09.002. [DOI] [PubMed] [Google Scholar]
- 46.Acimovic J, Goyal S, Kosir R, Golicnik M, Perse M, et al. Cytochrome P450 metabolism of the post-lanosterol intermediates explains enigmas of cholesterol synthesis. Sci Rep. 2016;6:28462. doi: 10.1038/srep28462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Wang Y, Kumar N, Solt LA, Richardson TI, Helvering LM, et al. Modulation of retinoic acid receptor-related orphan receptor alpha and gamma activity by 7-oxygenated sterol ligands. J Biol Chem. 2010;285:5013–5025. doi: 10.1074/jbc.M109.080614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Fauber BP, Rene O, Deng Y, DeVoss J, Eidenschenk C, et al. Discovery of 1-{4-[3-fluoro-4-((3s,6r)-3-methyl-1,1-dioxo-6-phenyl-[1,2]thiazinan-2-ylmethyl)- phenyl]-piperazin-1-yl}-ethanone (GNE-3500): a potent, selective, and orally bioavailable retinoic acid receptor-related orphan receptor C (RORc or RORgamma) inverse agonist. J Med Chem. 2015;58:5308–5322. doi: 10.1021/acs.jmedchem.5b00597. [DOI] [PubMed] [Google Scholar]
- 49.Wang Y, Kumar N, Crumbley C, Griffin PR, Burris TP. A second class of nuclear receptors for oxysterols: Regulation of RORalpha and RORgamma activity by 24S-hydroxycholesterol (cerebrosterol) Biochim Biophys Acta. 2010;1801:917–923. doi: 10.1016/j.bbalip.2010.02.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Tuong ZK, Lau P, Du X, Condon ND, Goode JM, et al. RORalpha and 25-Hydroxycholesterol Crosstalk Regulates Lipid Droplet Homeostasis in Macrophages. PLoS One. 2016;11:e0147179. doi: 10.1371/journal.pone.0147179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Xu T, Wang X, Zhong B, Nurieva RI, Ding S, et al. Ursolic acid suppresses interleukin-17 (IL-17) production by selectively antagonizing the function of RORgamma t protein. J Biol Chem. 2011;286:22707–22710. doi: 10.1074/jbc.C111.250407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Li X, Anderson M, Collin D, Muegge I, Wan J, et al. Structural studies unravel the active conformation of apo RORgammat nuclear receptor and a common inverse agonism of two diverse classes of RORgammat inhibitors. J Biol Chem. 2017;292:11618–11630. doi: 10.1074/jbc.M117.789024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Slominski AT, Kim TK, Hobrath JV, Oak ASW, Tang EKY, et al. Endogenously produced nonclassical vitamin D hydroxy-metabolites act as “biased” agonists on VDR and inverse agonists on RORalpha and RORgamma. J Steroid Biochem Mol Biol. 2017;173:42–56. doi: 10.1016/j.jsbmb.2016.09.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Slominski AT, Li W, Kim T, Semak I, Wang J, Zjawiony J, et al. Novel activities of CYP11A1 and their potential physiological significance. J Steroid Biochem Mol Biol. 2015;151:25–37. doi: 10.1016/j.jsbmb.2014.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Slominski AT, Kim TK, Hobrath JV, Janjetovic Z, Oak ASW, et al. Characterization of a new pathway that activates lumisterol in vivo to biologically active hydroxylumisterols. Sci Rep. 2017;7:11434. doi: 10.1038/s41598-017-10202-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Slominski AT, Kim TK, Shehabi HZ, Semak I, Tang EKY, et al. In vivo evidence for a novel pathway of vitamin D3 metabolism initiated by P450scc and modified by CYP27B1. FASEB J. 2012;26:3901–3915. doi: 10.1096/fj.12-208975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Slominski AT, Kim TK, Li W, Postlethwaite A, Thieu EW, et al. Detection of novel CYP11A1-derived secosteroids in the human epidermis and serum and pig adrenal gland. Sci Rep. 2015;5:14875. doi: 10.1038/srep14875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Varshney P, Narasimhan A, Mittal S, Malik G, Sardana K, et al. Transcriptome profiling unveils the role of cholesterol in IL-17A signaling in psoriasis. Sci Rep. 2016;6:19295. doi: 10.1038/srep19295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Ulivieri C, Baldari CT. Statins: from cholesterol-lowering drugs to novel immunomodulators for the treatment of Th17-mediated autoimmune diseases. Pharmacol Res. 2014;88:41–52. doi: 10.1016/j.phrs.2014.03.001. [DOI] [PubMed] [Google Scholar]
- 60.Kojima H, Muromoto R, Takahashi M, Takeuchi S, Takeda Y, et al. Inhibitory effects of azole-type fungicides on interleukin-17 gene expression via retinoic acid receptor-related orphan receptors alpha and gamma. Toxicol Appl Pharmacol. 2012;259:338–345. doi: 10.1016/j.taap.2012.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Urlep Z, Lorbek G, Perse M, Jeruc J, Juvan P, et al. Disrupting Hepatocyte Cyp51 from Cholesterol Synthesis Leads to Progressive Liver Injury in the Developing Mouse and Decreases RORC Signalling. Sci Rep. 2017;7:40775. doi: 10.1038/srep40775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Postigo J, Genre F, Iglesias M, Fernandez-Rey M, Buelta L, et al. Exacerbation of type II collagen-induced arthritis in apolipoprotein E-deficient mice in association with the expansion of Th1 and Th17 cells. Arthritis Rheum. 2011;63:971–980. doi: 10.1002/art.30220. [DOI] [PubMed] [Google Scholar]
- 63.Maggio R, Viscomi C, Andreozzi P, D’Ettorre G, Viscogliosi G, et al. Normocaloric low cholesterol diet modulates Th17/Treg balance in patients with chronic hepatitis C virus infection. PLoS One. 2014;9:e112346. doi: 10.1371/journal.pone.0112346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Wang C, Yosef N, Gaublomme J, Wu C, Lee Y, et al. CD5L/AIM Regulates Lipid Biosynthesis and Restrains Th17 Cell Pathogenicity. Cell. 2015;163:1413–1427. doi: 10.1016/j.cell.2015.10.068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Bovenga F, Sabba C, Moschetta A. Uncoupling nuclear receptor LXR and cholesterol metabolism in cancer. Cell Metab. 2015;21:517–526. doi: 10.1016/j.cmet.2015.03.002. [DOI] [PubMed] [Google Scholar]
- 66.Goldstein JL, DeBose-Boyd RA, Brown MS. Protein sensors for membrane sterols. Cell. 2006;124:35–46. doi: 10.1016/j.cell.2005.12.022. [DOI] [PubMed] [Google Scholar]
- 67.Shimano H, Sato R. SREBP-regulated lipid metabolism: convergent physiology - divergent pathophysiology. Nat Rev Endocrinol. 2017 doi: 10.1038/nrendo.2017.91. [DOI] [PubMed] [Google Scholar]
- 68.Kidani Y, Elsaesser H, Hock MB, Vergnes L, Williams KJ, et al. Sterol regulatory element-binding proteins are essential for the metabolic programming of effector T cells and adaptive immunity. Nat Immunol. 2013;14:489–499. doi: 10.1038/ni.2570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Lau P, Fitzsimmons RL, Raichur S, Wang SC, Lechtken A, et al. The orphan nuclear receptor, RORalpha, regulates gene expression that controls lipid metabolism: staggerer (SG/SG) mice are resistant to diet-induced obesity. J Biol Chem. 2008;283:18411–18421. doi: 10.1074/jbc.M710526200. [DOI] [PubMed] [Google Scholar]
- 70.Coban N, Gulec C, Ozsait-Selcuk B, Erginel-Unaltuna N. CYP19A1, MIF and ABCA1 genes are targets of the RORalpha in monocyte and endothelial cells. Cell Biol Int. 2017;41:163–176. doi: 10.1002/cbin.10712. [DOI] [PubMed] [Google Scholar]
- 71.Billon C, Sitaula S, Burris TP. Inhibition of RORalpha/gamma suppresses atherosclerosis via inhibition of both cholesterol absorption and inflammation. Mol Metab. 2016;5:997–1005. doi: 10.1016/j.molmet.2016.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Pathak P, Li T, Chiang JY. Retinoic acid-related orphan receptor alpha regulates diurnal rhythm and fasting induction of sterol 12alpha-hydroxylase in bile acid synthesis. J Biol Chem. 2013;288:37154–37165. doi: 10.1074/jbc.M113.485987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Ou Z, Shi X, Gilroy RK, Kirisci L, Romkes M, et al. Regulation of the human hydroxysteroid sulfotransferase (SULT2A1) by RORalpha and RORgamma and its potential relevance to human liver diseases. Mol Endocrinol. 2013;27:106–115. doi: 10.1210/me.2012-1145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Kang HS, Okamoto K, Takeda Y, Beak JY, Gerrish K, et al. Transcriptional profiling reveals a role for RORalpha in regulating gene expression in obesity-associated inflammation and hepatic steatosis. Physiol Genomics. 2011;43:818–828. doi: 10.1152/physiolgenomics.00206.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Nurieva RI, Chung Y, Hwang D, Yang XO, Kang HS, et al. Generation of T follicular helper cells is mediated by interleukin-21 but independent of T helper 1, 2, or 17 cell lineages. Immunity. 2008;29:138–149. doi: 10.1016/j.immuni.2008.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Yang XO, Pappu BP, Nurieva R, Akimzhanov A, Kang HS, et al. T helper 17 lineage differentiation is programmed by orphan nuclear receptors ROR alpha and ROR gamma. Immunity. 2008;28:29–39. doi: 10.1016/j.immuni.2007.11.016. In this study, the investigators report that Th17 cells also highly express another related nuclear receptor, RORα, that is induced by transforming growth factor-beta and interleukin-6 (IL-6), which is dependent on signal transducer and activator of transcription 3. Overexpression of RORα promotes Th17 differentiation. RORα-deficiency resulted in reduced IL-17 expression in vitro and in vivo. It was further shown that RORα and RORγ coexpression synergistically enhance Th17 differentiation, while RORα/RORγ double knockout mice exhibit a greater impaired Th17 generation and completely protected mice against experimental autoimmune encephalomyelitis. The study concludes that Th17 differentiation is directed by both nuclear receptors, RORα and RORγ. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, et al. The Orphan Nuclear Receptor RORgammat Directs the Differentiation Program of Proinflammatory IL-17(+) T Helper Cells. Cell. 2006;126:1121–1133. doi: 10.1016/j.cell.2006.07.035. IL-17-producing Th17 lymphocytes are major contributors to autoimmune disease. This report identifies RORγt as a key transcription factor of Th17 lineage differentiation. RORγt regulates the transcription of IL-17 and the related cytokine IL-17F in naïve T helper cells. RORγt-deficient mice T cells lack tissue-infiltration of Th17 cells and show an attenuated autoimmune disease. This study indicated for the first time that RORγt is a key regulator of immune homeostasis and might have potential as a therapeutic target in inflammatory diseases. [DOI] [PubMed] [Google Scholar]
- 78.Jetten AM. Immunology: A helping hand against autoimmunity. Nature. 2011;472:421–422. doi: 10.1038/472421a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Fauber BP, Rene O, Burton B, Everett C, Gobbi A, et al. Identification of tertiary sulfonamides as RORc inverse agonists. Bioorg Med Chem Lett. 2014;24:2182–2187. doi: 10.1016/j.bmcl.2014.03.038. [DOI] [PubMed] [Google Scholar]
- 80.Kumar N, Kojetin DJ, Solt LA, Kumar KG, Nuhant P, et al. Identification of SR3335 (ML-176): a synthetic RORalpha selective inverse agonist. ACS Chem Biol. 2011;6:218–222. doi: 10.1021/cb1002762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Kinzel O, Gege C, Steeneck C, Kleymann G, Hoffmann T. Seven-membered sulfonamides as modulators of RAR-related orphan receptor-gamma (RORγ, NR1F3) WO 2013/064231. 2013
- 82.Steeneck C, Kinzel O, Gege C, Kleymann G, Hoffmann T. Pyrrolo carboxamides as modulators of orphan nuclear receptor RAR-related orphan receptor-gamma (RORγ, NR1F3) activity and for the treatment of chronic inflammatory and autoimmune diseases. WO 2013/079223. 2013
- 83.Glick GD, Toogood PL, Romero AG, Vanhuis CA, Aicher TD, et al. WO 2012/064744. Tetrahydroquinoline and related bicyclic compounds for inhibition of RORγ activity and treatment of disease. 2012
- 84.Birault V, Campbell AJ, Harrison S, Le J. WO 2013/045431. Sulfonamide compounds and their use in the modulation of retinoid-related orphan receptor. 2013
- 85.Wang Y, Cai W, Zhang G, Yang T, Liu Q, et al. Discovery of novel N-(5-(arylcarbonyl)thiazol-2-yl)amides and N-(5-(arylcarbonyl)thiophen-2-yl)amides as potent RORgammat inhibitors. Bioorg Med Chem. 2014;22:692–702. doi: 10.1016/j.bmc.2013.12.021. [DOI] [PubMed] [Google Scholar]
- 86.van Niel MB, Fauber BP, Cartwright M, Gaines S, Killen JC, et al. A reversed sulfonamide series of selective RORc inverse agonists. Bioorg Med Chem Lett. 2014;24:5769–5776. doi: 10.1016/j.bmcl.2014.10.037. [DOI] [PubMed] [Google Scholar]
- 87.Doebelin C, Patouret R, Garcia-Ordonez RD, Chang MR, Dharmarajan V, et al. N-Arylsulfonyl Indolines as Retinoic Acid Receptor-Related Orphan Receptor gamma (RORgamma) Agonists. ChemMedChem. 2016;11:2607–2620. doi: 10.1002/cmdc.201600491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Olsson RI, Xue Y, von Berg S, Aagaard A, McPheat J, et al. Benzoxazepines Achieve Potent Suppression of IL-17 Release in Human T-Helper 17 (TH 17) Cells through an Induced-Fit Binding Mode to the Nuclear Receptor RORgamma. ChemMedChem. 2016;11:207–216. doi: 10.1002/cmdc.201500432. [DOI] [PubMed] [Google Scholar]
- 89.Chang MR, Lyda B, Kamenecka TM, Griffin PR. Pharmacologic repression of retinoic acid receptor-related orphan nuclear receptor gamma is therapeutic in the collagen-induced arthritis experimental model. Arthritis Rheumatol. 2014;66:579–588. doi: 10.1002/art.38272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Huang Z, Xie H, Wang R, Sun Z. Retinoid-related orphan receptor gamma t is a potential therapeutic target for controlling inflammatory autoimmunity. Expert Opin Ther Targets. 2007;11:737–743. doi: 10.1517/14728222.11.6.737. [DOI] [PubMed] [Google Scholar]
- 91.Hintermann S, Guntermann C, Mattes H, Carcache DA, Wagner J, et al. Synthesis and Biological Evaluation of New Triazolo- and Imidazolopyridine RORgammat Inverse Agonists. ChemMedChem. 2016;11:2640–2648. doi: 10.1002/cmdc.201600500. [DOI] [PubMed] [Google Scholar]
- 92.Chang MR, Dharmarajan V, Doebelin C, Garcia-Ordonez RD, Novick SJ, et al. Synthetic RORgammat Agonists Enhance Protective Immunity. ACS Chem Biol. 2016;11:1012–1018. doi: 10.1021/acschembio.5b00899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Scheepstra M, Leysen S, van Almen GC, Miller JR, Piesvaux J, et al. Identification of an allosteric binding site for RORgammat inhibition. Nat Commun. 2015;6:8833. doi: 10.1038/ncomms9833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Fujita-Sato S, Ito S, Isobe T, Ohyama T, Wakabayashi K, et al. Structural basis of digoxin that antagonizes RORgamma t receptor activity and suppresses Th17 cell differentiation and interleukin (IL)-17 production. J Biol Chem. 2011;286:31409–31417. doi: 10.1074/jbc.M111.254003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.de Wit J, Al-Mossawi MH, Huhn MH, Arancibia-Carcamo CV, Doig K, et al. RORgammat inhibitors suppress T(H)17 responses in inflammatory arthritis and inflammatory bowel disease. J Allergy Clin Immunol. 2016;137:960–963. doi: 10.1016/j.jaci.2015.09.048. [DOI] [PubMed] [Google Scholar]
- 96.Huh JR, Leung MW, Huang P, Ryan DA, Krout MR, et al. Digoxin and its derivatives suppress TH17 cell differentiation by antagonizing RORgammat activity. Nature. 2011;472:486–490. doi: 10.1038/nature09978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Guntermann C, Piaia A, Hamel ML, Theil D, Rubic-Schneider T, et al. Retinoic-acid-orphan-receptor-C inhibition suppresses Th17 cells and induces thymic aberrations. JCI Insight. 2017;2:e91127. doi: 10.1172/jci.insight.91127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Smith SH, Peredo CE, Takeda Y, Bui T, Neil J, et al. Development of a Topical Treatment for Psoriasis Targeting RORgamma: From Bench to Skin. PLoS One. 2016;11:e0147979. doi: 10.1371/journal.pone.0147979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Xue X, Soroosh P, De Leon-Tabaldo A, Luna-Roman R, Sablad M, et al. Pharmacologic modulation of RORgammat translates to efficacy in preclinical and translational models of psoriasis and inflammatory arthritis. Sci Rep. 2016;6:37977. doi: 10.1038/srep37977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Skepner J, Ramesh R, Trocha M, Schmidt D, Baloglu E, et al. Pharmacologic inhibition of RORgammat regulates Th17 signature gene expression and suppresses cutaneous inflammation in vivo. J Immunol. 2014;192:2564–2575. doi: 10.4049/jimmunol.1302190. [DOI] [PubMed] [Google Scholar]
- 101.Solt LA, Banerjee S, Campbell S, Kamenecka TM, Burris TP. ROR Inverse Agonist Suppresses Insulitis and Prevents Hyperglycemia in a Mouse Model of Type 1 Diabetes. Endocrinology. 2015:en20141677. doi: 10.1210/en.2014-1677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Guendisch U, Weiss J, Ecoeur F, Riker JC, Kaupmann K, et al. Pharmacological inhibition of RORgammat suppresses the Th17 pathway and alleviates arthritis in vivo. PLoS One. 2017;12:e0188391. doi: 10.1371/journal.pone.0188391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Solt LA, Kumar N, Nuhant P, Wang Y, Lauer JL, et al. Suppression of TH17 differentiation and autoimmunity by a synthetic ROR ligand. Nature. 2011;472:491–494. doi: 10.1038/nature10075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Wang T, Niwa S, Bouda K, Matsuura S, Homma T, et al. The effect of Am-80, one of retinoids derivatives on experimental allergic encephalomyelitis in rats. Life Sci. 2000;67:1869–1879. doi: 10.1016/s0024-3205(00)00776-1. [DOI] [PubMed] [Google Scholar]
- 105.Guo Y, MacIsaac KD, Chen Y, Miller RJ, Jain R, et al. Inhibition of RORgammaT Skews TCRalpha Gene Rearrangement and Limits T Cell Repertoire Diversity. Cell Rep. 2016;17:3206–3218. doi: 10.1016/j.celrep.2016.11.073. [DOI] [PubMed] [Google Scholar]
- 106.Wang Y, Cai W, Cheng Y, Yang T, Liu Q, et al. Discovery of Biaryl Amides as Potent, Orally Bioavailable, and CNS Penetrant RORgammat Inhibitors. ACS Med Chem Lett. 2015;6:787–792. doi: 10.1021/acsmedchemlett.5b00122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Takaishi M, Ishizaki M, Suzuki K, Isobe T, Shimozato T, et al. Oral administration of a novel RORgammat antagonist attenuates psoriasis-like skin lesion of two independent mouse models through neutralization of IL-17. J Dermatol Sci. 2017;85:12–19. doi: 10.1016/j.jdermsci.2016.10.001. [DOI] [PubMed] [Google Scholar]
- 108.Banerjee D, Zhao L, Wu L, Palanichamy A, Ergun A, et al. Small molecule mediated inhibition of RORgamma-dependent gene expression and autoimmune disease pathology in vivo. Immunology. 2016;147:399–413. doi: 10.1111/imm.12570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Gege C. Retinoid-related orphan receptor gamma t (RORgammat) inhibitors from Vitae Pharmaceuticals (WO2015116904) and structure proposal for their Phase I candidate VTP-43742. Expert Opin Ther Pat. 2016;26:737–744. doi: 10.1517/13543776.2016.1153066. [DOI] [PubMed] [Google Scholar]
- 110.Chang MR, He Y, Khan TM, Kuruvilla DS, Garcia-Ordonez R, et al. Antiobesity Effect of a Small Molecule Repressor of RORgamma. Mol Pharmacol. 2015;88:48–56. doi: 10.1124/mol.114.097485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Forcade E, Paz K, Flynn R, Griesenauer B, Amet T, et al. An activated Th17-prone T cell subset involved in chronic graft-versus-host disease sensitive to pharmacological inhibition. JCI Insight. 2017:2. doi: 10.1172/jci.insight.92111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Withers DR, Hepworth MR. Group 3 Innate Lymphoid Cells: Communications Hubs of the Intestinal Immune System. Front Immunol. 2017;8:1298. doi: 10.3389/fimmu.2017.01298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Withers DR, Hepworth MR, Wang X, Mackley EC, Halford EE, et al. Transient inhibition of ROR-gammat therapeutically limits intestinal inflammation by reducing TH17 cells and preserving group 3 innate lymphoid cells. Nat Med. 2016;22:319–323. doi: 10.1038/nm.4046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Bronner SM, Zbieg JR, Crawford JJ. RORgamma antagonists and inverse agonists: a patent review. Expert Opin Ther Pat. 2017;27:101–112. doi: 10.1080/13543776.2017.1236918. [DOI] [PubMed] [Google Scholar]
- 115.Gege C. RORgammat inhibitors as potential back-ups for the phase II candidate VTP-43742 from Vitae Pharmaceuticals: patent evaluation of WO2016061160 and US20160122345. Expert Opin Ther Pat. 2017;27:1–8. doi: 10.1080/13543776.2017.1262350. [DOI] [PubMed] [Google Scholar]
- 116.Ueda E, Kurebayashi S, Sakaue M, Backlund M, Koller B, et al. High incidence of T-cell lymphomas in mice deficient in the retinoid- related orphan receptor RORgamma. Cancer Res. 2002;62:901–909. In this study, the investigators show that changes in homeostasis in the thymus of RORγ-deficient mice are associated with a high incidence of T-cell lymphomas that metastasized frequently to spleen and liver. No other tumor types were detected in any of RORγ-deficient mice. Lymphoma formation was associated with increased cellular proliferation and an increase in the number of apoptotic cells. Analysis of the immunophenotype of the lymphoblastic cells showed that the CD4 and CD8 subpopulations varied substantially among different lymphomas. The established cell lines consisted mostly of CD44-CD25+CD4-CD8- cells. Thus, the loss of RORγ disturbs homeostasis in the thymus by enhancing apoptosis and cellular proliferation. The latter lead via an unknown mechanism to increased susceptibility to the development of T-cell lymphoma. [PubMed] [Google Scholar]
- 117.Liljevald M, Rehnberg M, Soderberg M, Ramnegard M, Borjesson J, et al. Retinoid-related orphan receptor gamma (RORgamma) adult induced knockout mice develop lymphoblastic lymphoma. Autoimmun Rev. 2016;15:1062–1070. doi: 10.1016/j.autrev.2016.07.036. [DOI] [PubMed] [Google Scholar]
- 118.Lee Y, Kuchroo V. Defining the functional states of Th17 cells. F1000Res. 2015;4:132. doi: 10.12688/f1000research.6116.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Thaiss CA, Zmora N, Levy M, Elinav E. The microbiome and innate immunity. Nature. 2016;535:65–74. doi: 10.1038/nature18847. [DOI] [PubMed] [Google Scholar]
- 120.Wang Y, Kuang Z, Yu X, Ruhn KA, Kubo M, et al. The intestinal microbiota regulates body composition through NFIL3 and the circadian clock. Science. 2017;357:912–916. doi: 10.1126/science.aan0677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Yu X, Rollins D, Ruhn KA, Stubblefield JJ, Green CB, et al. TH17 cell differentiation is regulated by the circadian clock. Science. 2013;342:727–730. doi: 10.1126/science.1243884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Man K, Loudon A, Chawla A. Immunity around the clock. Science. 2016;354:999–1003. doi: 10.1126/science.aah4966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Takeda Y, Jothi R, Birault V, Jetten AM. RORγ directly regulates the circadian expression of clock genes and downstream targets in vivo. Nucleic Acids Res. 2012;40:8519–8535. doi: 10.1093/nar/gks630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Solt LA, Kojetin DJ, Burris TP. The REV-ERBs and RORs: molecular links between circadian rhythms and lipid homeostasis. Future Med Chem. 2011;3:623–638. doi: 10.4155/fmc.11.9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Takeda Y, Kang HS, Angers M, Jetten AM. Retinoic acid-related orphan receptor gamma directly regulates neuronal PAS domain protein 2 transcription in vivo. Nucleic Acids Res. 2011;39:4769–4782. doi: 10.1093/nar/gkq1335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Hannedouche S, Zhang J, Yi T, Shen W, Nguyen D, et al. Oxysterols direct immune cell migration via EBI2. Nature. 2011;475:524–527. doi: 10.1038/nature10280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Nachtergaele S, Mydock LK, Krishnan K, Rammohan J, Schlesinger PH, et al. Oxysterols are allosteric activators of the oncoprotein Smoothened. Nat Chem Biol. 2012;8:211–220. doi: 10.1038/nchembio.765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Janowski BA, Grogan MJ, Jones SA, Wisely GB, Kliewer SA, et al. Structural requirements of ligands for the oxysterol liver X receptors LXRalpha and LXRbeta. Proc Natl Acad Sci U S A. 1999;96:266–271. doi: 10.1073/pnas.96.1.266. [DOI] [PMC free article] [PubMed] [Google Scholar]