Next Article in Journal
Cytokine Expression and Macrophage Localization in Xenograft and Allograft Tumor Models Stimulated with Lipopolysaccharide
Next Article in Special Issue
An aPPARent Functional Consequence in Skeletal Muscle Physiology via Peroxisome Proliferator-Activated Receptors
Previous Article in Journal
Clinicopathological Characteristics and Mutations Driving Development of Early Lung Adenocarcinoma: Tumor Initiation and Progression
Previous Article in Special Issue
Demyelination in Multiple Sclerosis: Reprogramming Energy Metabolism and Potential PPARγ Agonist Treatment Approaches
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of PPAR and Its Cross-Talk with CAR and LXR in Obesity and Atherosclerosis

1
Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
2
Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry, College of Life Sciences, Beijing Normal University, Beijing 100875, China
3
Department of Biology Science and Technology, Baotou Teacher’s College, Baotou 014030, China
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2018, 19(4), 1260; https://doi.org/10.3390/ijms19041260
Submission received: 26 March 2018 / Revised: 13 April 2018 / Accepted: 19 April 2018 / Published: 23 April 2018
(This article belongs to the Special Issue PPARs in Cellular and Whole Body Energy Metabolism)

Abstract

:
The prevalence of obesity and atherosclerosis has substantially increased worldwide over the past several decades. Peroxisome proliferator-activated receptors (PPARs), as fatty acids sensors, have been therapeutic targets in several human lipid metabolic diseases, such as obesity, atherosclerosis, diabetes, hyperlipidaemia, and non-alcoholic fatty liver disease. Constitutive androstane receptor (CAR) and liver X receptors (LXRs) were also reported as potential therapeutic targets for the treatment of obesity and atherosclerosis, respectively. Further clarification of the internal relationships between these three lipid metabolic nuclear receptors is necessary to enable drug discovery. In this review, we mainly summarized the cross-talk of PPARs-CAR in obesity and PPARs-LXRs in atherosclerosis.

Graphical Abstract

1. Introduction

Obesity is a lipid metabolic disturbance that has been growing across the world for nearly half a century. It is a global human health concern. In 2016, more than 1.9 billion adults (≥18 years old) were overweight and, of these, over 650 million were obese. Furthermore, 340 million children and adolescents (5–18 years old) and 41 million children (≤5 years old) were overweight or obese [1,2]. The body mass index (BMI), defined as a person’s weight in kilograms divided by the square of their height in meters, is a simple index used to classify overweight and obesity in adults. Obesity is associated with various metabolic disorders and cardiovascular diseases. A high BMI is considered to be an indicator of high body fatness that may lead to a high risk of cardiometabolic syndrome and atherosclerotic vascular disease [3,4,5]. Atherosclerosis, also known as arteriosclerosis, hardening of the arteries, is a disease in which fatty plaque deposits build up inside the arteries, narrowing them, leading to some serious problems, including coronary artery disease, stroke, or even death [6]. Obesity and atherosclerosis are common chronic lipid metabolic disorder diseases. The treatment and prevention of obesity and atherosclerosis are both major challenges, and studying this problem can help us live longer, healthier lives.
Nuclear receptors (NRs), a class of ligand-activated transcriptional factors, play significant roles in metabolic homeostasis. It is well known that there are 48 and 49 NR genes in humans (Homo sapiens) and mice (Mus musculus), respectively [7,8]. Most of the NRs contain six functional domains, such as the variable N-terminal regulatory domain (A–B), the conserved DNA-binding domain (DBD) (C), the variable hinge region (D), the conserved ligand binding domain (LBD) (E), and the variable C-terminal domain (F) (Figure 1a) [7,9]. The classical function of NRs is to transcriptionally regulate the expression of cognate target genes through the recruitment of coactivators or corepressors when ligands bind to the receptors [10,11] (Figure 1b). To perform the transcriptional activity, NRs either (1) act as monomers; (2) need to form dimeric complexes (homodimers); or (3) form complexes with the retinoid X receptor (RXR) (heterodimers) and bind to the DNA in the cell nucleus [9]. Recently, many studies have indicated the role of some NRs in the regulation of lipid metabolism. It has been recognized that peroxisome proliferator-activated receptors (PPARs) act as fatty acid sensors, regulating the multiple pathways involved in lipid and glucose metabolism and overall energy metabolism [12,13]. Furthermore, the constitutive androstane receptor (CAR), which was initially characterized as a xenosensor that controls xenobiotic responses, has been recently identified as a therapeutic target for obesity and its related metabolic disorders [14,15], whereas liver X receptors (LXRs) are sterol sensors that mainly regulate cholesterol, fatty acid and glucose homeostasis, they can inhibit atherosclerosis development, but promote lipogenesis in liver [16]. In this review, we briefly summarize the roles of PPARs, CAR and LXRs and their ligands in the treatment of metabolic diseases, obesity and atherosclerosis, and discuss the cross-talk of PPARs-CAR and PPARs-LXRs in lipid metabolism regulation.

2. The Initial Characterization of PPAR, CAR, and LXR

2.1. Fatty Acids Sensor PPARs

PPARs are molecular sensors of fatty acids and fatty acid derivatives and control energy homeostasis (carbohydrate, lipid, and protein) [17]. There are three types of PPARs which have been identified: PPARα (NR1C1, encoded by PPARA), PPARβ/δ (NR1C2, encoded by PPARD), and PPARγ (NR1C3, encoded by PPARG). They are all lipid sensors that transcriptionally regulate diverse aspects in response to nutritional inputs, and serving as effective therapeutic targets for some types of lipid metabolic syndrome, including obesity, atherosclerosis, dyslipidaemia, type 2 diabetes mellitus (T2DM), and nonalcoholic fatty liver disease (NAFLD) [12,18]. PPARα is highly active in liver, brown adipose tissue (BAT), kidney, heart, and muscle tissue [19], where it regulates the adaptive response to prolonged fasting by controlling the process of ketogenesis, fatty acid transport, fatty acid binding, fatty acid activation and mitochondrial fatty acid β-oxidation [20,21]. Genomic studies have indicated that PPARα, as a master regulator of lipid metabolism, has various target genes; the classical genes include acyl-CoA oxidase, thiolase, fatty acid transport protein (FATP), carnitine palmitoyltransferase I (CPT1), and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) [20,22]. The expression of PPARβ/δ is highest in adipose tissue, skeletal muscle, macrophages, brain, and skin, but is at low levels in the liver, where it mainly regulates fatty acid catabolism and the glycolytic-to-oxidative muscle fibre-type switching used in improving lipid homeostasis [23,24,25,26]. PPARα and PPARβ/δ have been shown to block lipid absorption by upregulating L-type fatty acid binding protein (L-FABP) and cluster of differentiation 36 (CD36) in the small intestine [27]. PPARγ function has mainly been characterized in adipose tissue, macrophages and the colon, and it has three forms: PPARγ1, PPARγ2, and PPARγ3 through alternative splicing [28,29,30]. PPARγ1 and PPARγ3 encode the same protein, and PPARγ3 is a splicing variant of PPARγ1. PPARγ2 has 28 additional amino acids at the variable N-terminal regulatory domain compared with PPARγ1 [31]. Furthermore, PPARγ1 has been found in nearly all tissues, except muscle, whereas PPARγ2 is mostly found in the adipose tissue and intestine, and PPARγ3 is mainly expressed white adipose tissue, colon, and macrophages [32]. PPARγ was initially known as an inducer during adipocyte differentiation [33,34], and its most famous role is in regulating lipogenic pathways. Genomic studies have revealed that PPARγ controls the expression of the early adipogenic differentiation factors CCAAT-enhancer-binding proteins (C/EBPs) and fatty acid binding protein 4 (FABP4), glucose homeostasis factors glucose transporter type 4 (GLUT4), and catabolite activator protein (CAP) genes. Moreover, PPARγ regulates some insulin sensitive adipokines, such as leptin, adiponectin, and tumour necrosis factor α (TNF-α) [35,36,37]. PPARγ is also involved in the metabolism of long-chain unsaturated fatty acid in the intestinal epithelium [38]. Although there are many similarities in lipid and glucose homeostasis, each of the PPAR isoforms has unique functions in vivo, probably due to their differential tissue distributions, the distinct ligands, and the inherent differences in biochemical characteristics [39].
Many endogenous agonists of PPARs have been identified, including polyunsaturated fatty acids, branched chain fatty acids, nitro/oxidized-fatty acids, phospholipids, eicosanoids, prostaglandin, oleoylethanolamide, carbaprostacyclin, 5HT metabolites, and so on [40,41,42,43]. In addition, many natural and synthetic PPAR ligands have been applied to treat lipid and glucose metabolic syndrome in pharmaceutical companies, as shown in Table 1. Fibrate drugs (including bezafibrate, clofibrate, fenofibrate, gemfibrozil, ronifibrate, etc.) are a class of classical PPARα agonists used to treat hyperlipidaemia and increase high-density lipoprotein cholesterol (HDL-c) in clinical settings. Moreover, pemafibrate [44] (approved in Japan in July 2017) and LY518674 [45] (phase II) are selective PPARα modulators used as anti-atherosclerosis agents in clinical trials. PPARβ/δ agonists are currently not used in clinical applications, but seladelpar (MBX-8025) is currently a promising activator for improving mixed dyslipidaemia and normalizing alkaline phosphatase levels, and is in phase 2 clinical development [46]. Additionally, KD-3010 is also a promising PPARβ/δ agonist for the potential treatment of diabetes and obesity in the phase I clinical trial. It shows the protective and anti-fibrotic effects in liver injury induced by carbon tetrachloride (CCl4) injection or bile duct ligation (BDL) [47]. Thiazolidinediones (generically marked as pioglitazone, rosiglitazone, and lobeglitazone) are potent agonists of PPARγ with powerful insulin sensitizing activity which can be used in the treatment of T2DM. However, they have some undesirable side effects, such as weight gain, osteoporosis, and congestive heart failure [39,48]. Some failed and non-marked thiazolidinediones include troglitazone (marked as Rezulin, which was withdrawn due to adverse liver effects), balaglitazone, ciglitazone, darglitazone, netoglitazone, and rivoglitazone, etc. Recently, several partial agonists of PPARγ have been reported to keep beneficial antidiabetic characteristics with few side effects. Honokiol is a natural compound purified from the bark of Magnolia officinalis in traditional Chinese medicine, which has been identified as a novel non-adipogenic partial PPAPγ ligand. It has an anti-hyperglycemic property but does not trigger adipogenesis in vitro and in vivo [48]. Amorfrutins, as selective PPARγ modulators, are also natural products derived from two legumes, Glycyrrhiza foetida and Amorpha fruticose. They were reported to improve insulin sensitivity and dyslipidemia and protect liver steatosis without a concomitant increase of body weight gain in diet-induced obese and db/db mice [49,50]. In our recent study, Danshensu Bingpian Zhi (DBZ) is a synthetic derivative of the natural compounds Danshensu (tanshinol) and Bingpian (borneol), which are used as “sovereign” and “courier” in the traditional Chinese medicine formula Fufang Danshen (FFDS). We found that DBZ is a putative PPARγ partial activator capable of preventing insulin resistance, obesity, and atherosclerosis in mice without significant unwanted effects [51,52]. Along with improving our understanding of the biological roles of PPARs, we suggest that further study of the selectively pleiotropic PPAR agonist is a promising approach for developing further therapies.

2.2. Xenobiotic Receptor CAR

CAR is a member of the NR1I3 family of nuclear receptors, initially serves as a xenobiotic nuclear receptor, responding to xenobiotics and drug stress [53,54]. Androstenol, and some isomers of androstanol, androstanes, have been found to be endogenous antagonists of CAR, and dehydroepiandrosterone (DHEA), also an androstane, is an endogenous agonist of CAR. Androstanes, despite acting as ligands, are the basis for the naming of this receptor. The name “constitutive androstane receptor” refers to the unusual, constitutively-active status of this receptor when not occupied by a ligand. CAR is primarily expressed in the liver and small intestine, but is also found in the kidney, heart, and brain [55], and we also detected it in the mammary gland, ovary, and uterus (our unpublished data). It, often along with the pregnane X receptor (PXR) and vitamin D receptor (VDR), regulates the phase I and II xenobiotic metabolizing enzymes (including cytochrome P450s, sulfotransferases, glutathione-S-transferases) and other multidrug-resistance associated proteins used to both modulate drug metabolism and bilirubin clearance and prevent hepatotoxicity [56,57,58]. More recently, CAR has been reported to regulate both lipid and glucose metabolism and has been a potential therapeutic target for several metabolic diseases, such as obesity [15,59], atherosclerosis [60,61], NAFLD [62,63], and T2DM [64,65], due to its ability to balance the endogenous homeostasis of components, including glucose, steroids, bile acids, bilirubin, and thyroid hormone.
Since CAR has a large hydrophobic LBD pocket, a variety of chemical xenobiotics can activate it, such as clinical drugs, insecticides, flavonoids, terpenoids, polyphenols, environmental chemicals, and others [66,67]. Interestingly, CAR exhibits arresting species specificity in the ligand binding recognition between human and rodent, though both species use the same DNA response element sequences to recruit CAR. For example, TCPOBOP (1,4-bis[2-(3,5-dichloropyridyloxy)]benzene), is a potent mouse CAR (mCAR) agonist which only activates mouse, but not human, CAR, whereas CITCO (6-(4-chlorophenyl) imidazo [2,1-β] [1,3] thiazole-5-carbaldehyde-O-(3,4-dichlorobenzyl) oxime) is only a human CAR (hCAR) agonist, having no effect on mouse CAR [68,69]. Thus, this specificity should be considered when choosing the animal model for studying pharmacologic effects or drug screens targeting CAR. Phenobarbital, also known as phenobarb or phenobarbitone, is the preferred antiepileptic and sedation medicine used clinically, and it can activate both human and mouse CAR. Some early studies have shown that phenobarbital can regulate energy mentalism and improve insulin sensitivity and hepatic lipid homeostasis in ob/ob mice and human patients [70,71,72]. Activation of CAR reduced sterol regulatory element-binding protein 1 (SREBP-1) levels by inducing the expression of insulin induced gene 1 protein (INSIG-1), a protein blocking the proteolytic activation of SREBPs [73]. In a previous study, we reported that activation of CAR inhibited lipogenesis by suppressing LXR ligand-responded recruitment of LXR to the LXR response element (LXRE) and the expression of LXR target genes, whereas activation of LXR inhibited the CAR ligand-induced recruitment of CAR to Cyp2b10 [74]. Although CAR is a potential therapeutic target for lipid metabolic disease, some barriers exist for the clinical use of its agonists: there are concerns around hepatic enlargement and carcinogenesis. CAR also interacts with PPAR and LXR in regulating lipid and glucose homeostasis. Better understanding of these mechanistic properties might help us overcome these barriers in the future.

2.3. Oxysterol Sensor LXRs

LXRs are well-known nuclear oxysterol receptors that have two isotypes: LXRα (NR1H3) and LXRβ (NR1H2). LXRα is highly active in the liver, intestines, kidneys, adipose tissue, lungs, macrophages, and adrenal glands. LXRβ, also named as a ubiquitous receptor, is expressed in almost all tissues and organs [75,76,77]. Both of them may control cholesterol, fatty acid, and glucose metabolism to protect against atherosclerosis, lipid disorders, diabetes, chronic inflammation, Alzheimer’s disease, and even cancer [78,79,80,81].
In cholesterol and lipid homeostasis, activation of LXR can stimulate reverse cholesterol transport and reduce the body’s cholesterol overload by inducing the sterol metabolism and transporter network, including cytochrome P450 family 7 subfamily A member 1 (CYP7A1), ATP-binding cassette sub-family A member 1 (ABCA1), ABCG1, ABCG5, ABCG8, and apolipoprotein E (ApoE) [82,83,84]. Furthermore, LXR activation also results in an increase in lipid synthesis in the liver through inducing the expression of SREBP-1c, fatty acid synthase (FAS), acetyl-CoA carboxylase 1 (ACC1), and stearoyl-CoA desaturase 1 (SCD-1) [85,86,87]. LXRs, as sterol sensors, have a variety of endogenous activators, most of which are oxidation products of cholesterol, such as 27-hydroxycholesterol, 22(R)-hydroxycholesterol, 20(S)-hydroxycholesterol, 24(S)-hydroxycholesterol and 24(S), and 25-epoxycholesterol [16,76,88]. Interestingly, these endogenous agonists, unlike natural synthetic LXR activators, do not activate the SREBP signal pathway [89,90,91]. Several studies have reported that mice treated with synthetic LXR activators, including GW3965 and TO901317, show enhanced hepatic and serous triglyceride levels, and have promoted very low-density lipoprotein (VLDL) secretion [86,92,93]. These shortcomings limit the use of LXR activators in clinical settings. LXRα is the major sensor of dietary cholesterol. Mice lacking LXRα cannot induce transcription of the gene encoding cholesterol 7α-hydroxylase (CYP7A), which is a rate-limiting enzyme in bile acid synthesis. LXRα−/− mice are healthy when fed with a normal chow (low cholesterol) diet. However, they develop enlarged fatty livers with high cholesterol levels, and lead to impaired hepatic function when fed a high-cholesterol diet [94]. LXR-623 (WAY-252623) is the first LXRα-partial/LXRβ-full agonist used for the treatment of atherosclerosis in animal models and has been tested in a phase I clinical trial. However, the trial was terminated due to adverse effects on the central nervous system [95,96]. Similar synthetic agonists, including CS8080, BMS-852927 (also named XL-041) have been terminated for undisclosed reasons, and only BMS-779788 (also named XL-652) has proved safe enough to continue with clinical trials [97,98], the detailed information as shown in Table 2. LXR activators can reduce cholesterol level in blood and liver. They also improve glucose tolerance in mice by decreasing insulin resistance. Human functional and genetic analysis showed that the common LXR promoter SNPs rs35463555 and rs17373080 may regulate sensibility to T2D [99]. We recently reported that DBZ inhibits foam cell formation and protects against atherosclerosis in ApoE−/− mice through activating LXRs [52,100]. DBZ also activates PPARγ and prevents high fat diet-induced obesity, insulin resistance and gut dysbiosis in mice [51]. By clarifying the cross-talk between PPARs and LXRs we may gain a better understanding of their synactic function in cholesterol and lipid homeostasis.

3. Cross-Talk of PPARs and CAR Links to Obesity

PPARs and CAR are both essential lipid metabolic nuclear receptors active in controlling obesity and its related metabolic disorders. PPARs are quite interesting. PPARα and PPARβ/δ are potential targets to prevent obesity [101,102,103], by the mechanism as mentioned above in Section 2.1. Contrarily, PPARγ is a master regulator of adipocyte differentiation both in vivo and in vitro [104]. A lack of PPARγ results in the inability to develop adipose tissue, as seen in PPARγ knockout mice [105,106]. Thiazolidinediones, as famous PPARγ activators, are a group of anti-diabetic drugs to treat T2MD, but can lead to serious side effects. Weight gain is an unwanted side effect: activation of PPARγ in adipose tissue stimulates the expression of genes leading to lipogenesis, including AP2, CD36, SCD-1, SREBP-1, and others, which promote lipid storage [18]. PPARα, as a key nutritional sensor, regulates the metabolism of lipids, carbohydrates, and amino acids [107]. It is a potential therapeutic target for the treatment of obesity, hypertriglyceridemia, NAFLD, and atherogenic dyslipidaemia [108,109,110]. Oestrogen inhibits the actions of PPARα on obesity and lipid metabolism through its effects on the PPARα-dependent regulation of target genes [111,112]. CAR, as a therapeutic target for obesity, was reported about ten years ago. Activation of CAR also increased faecal bile acid excretion and attenuated atherosclerosis in low-density lipoprotein receptor-deficient (LDLR−/−) and ApoE−/− mice by increasing reverse cholesterol transport [60,61]. Recently, we reported that activation of CAR with TCPOBOP inhibited lipogenesis and promoted fibrosis in the mammary gland of adolescent female mice [113]. The classical CAR agonist TCPOBOP has a robust anti-obesity phenotype in high-fat diet-induced obese mouse models. Mechanically, activation of CAR improves insulin sensitivity, inhibits lipogenesis and gluconeogenesis, and increases brown adipose tissue energy expenditure.
The cross-talk between PPARs and CAR in obesity can be achieved through their target gene PGC-1α. PGC-1α, as a transcriptional coactivator, interacts with nuclear receptor PPAR and controls energy metabolism through the regulation of mitochondrial biogenesis [114,115]. CAR regulates the degradation of PGC-1α by recruiting E3 ligase targeting PGC1α and promoting ubiquitination in the liver [116]. During fasting, the PPARα activator WY14643 induces both CAR and its target gene CYP2B expression in a PPARα-dependent manner in rat hepatocytes [117,118]. Meanwhile, Guo et al. reported that synthetic PPARα ligands ciprofibrate, clofibrate, and others drove adenoviral-enhanced green fluorescent protein-CAR into the hepatocyte nucleus in a PPARα- and PPARβ-independent manner in mouse liver in vivo. More interestingly, molecular docking assay showed that PPARα activators, Wy-14643 and ciprofibrate, could fit into the ligand binding pocket of CAR and their binding modes were similar with that of androstanol, an endogenic CAR inverse agonist. PPARα activators interfered with coactivator recruitment to the LBD of CAR and suppressed the constitutive transactivation of CAR. Mechanistically, the transcription coactivator PPAR-binding protein (PBP) plays a pivotal role in nuclear translocation of CAR in mouse liver, but not the PPAR-interacting protein (PRIP) [119,120]. These results indicated that activation of PPARα by some ligands induced nuclear translocation of CAR. β-oxidation is also controlled by both PPARs and CAR. PPARα regulates mitochondrial fatty acid β-oxidation by inducing the gene expression of CPT1, as previously mentioned. Conversely, the CAR ligand pentobarbital inhibits mitochondrial CPT1 expression and β-oxidation, resulting in increasing ketone production in serum [8,121]. However, in BAT, activation of CAR by TCPOBOP significantly increased expression of PGC-1α and β-oxidation [15]. Hence, the cross-talk between PPAR and CAR should be separately considered for different tissue types. Above all, the dual functions of PPAR activators have possible cross-talk with CAR through target gene PGC1α, coactivator recruitment, and mitochondrial fatty acid β-oxidation in different conditions in energy metabolism.

4. Cross-Talk of PPARS and LXRS in Atherosclerosis

There is a potential cross-talk or interaction between PPARs and LXRs in the prevention and treatment of atherosclerosis. Most nuclear receptors form heterodimers with RXR, including PPAR/RXR, LXR/RXR, CAR/RXR, and others. Ide et al. has elegantly reported that LXR-RXR-PPAR forms a network that regulates fatty acid metabolism and lipid degradation [122]. These compounds enhance binding to their respective target gene promoters. Unsaturated fatty acids increase the expression of LXRα, but not the LXRβ in rat liver cells, both in vivo and in vitro. This upregulated effect of LXRα is associated with the transcriptional rate and binding of PPARα to PPAR response element (PPRE). Meanwhile, a PPRE is found in the human LXRα flanking region [123]. SREBP-1c, as a direct target gene regulated by LXR, is crucial in both lipid and sterol biosynthesis. Luciferase assays have proven that the activation of PPARα and PPARγ reduces LXR-induced SREBP-1c promoter activity and gel shift assays have demonstrated that PPARs inhibit the binding of LXR/RXR to LXRE [124]. Thus, PPARs and LXRs play opposite roles in regulating triglyceride synthesis in the liver and serum. LXRα also inhibits peroxisome proliferator signalling through cross-talk with PPARα [125]. Moreover, Liduo Yue et al. reported that LXRs could bind to PPARs with different binding affinities in vitro using surface plasmon resonance technology and molecular dynamics simulation [126].
Despite the opposite roles in triglyceride homeostasis, PPARs and LXRs have some common ground in their anti-atherosclerotic effects. In foam cell macrophages, both PPARα and PPARγ (through the LXR-dependent ABC pathway) control cholesterol efflux [127,128], and activation of PPARα and PPARγ both prevent foam cell formation and atherosclerosis development in ApoE−/− and LDLR−/− mice [129,130]. Activation of LXRα also raises the expression of ABCA1 and ABCG1, which accelerate the reverse transport of cholesterol and then deposit in the liver [131]. PPAR-LXR-ABCA1 is an important pathway involved in cholesterol efflux and atherogenesis. In intestine tissue, the activation of LXR also increases the expression of ABCG5 and ABCG8 which regulate absorption of cholesterol and protect against atherosclerosis [79,132]. PPARs activation has performed similar acts inhibiting intestinal cholesterol absorption in rats and mice [133,134]. Taken together, both LXR and PPAR promote the movement of cholesterol from peripheral cells to the feces, which is referred to as reverse cholesterol transport (RCT).
Atherosclerosis is a chronic inflammatory disease; inflammation plays an important role in the pathogenesis and progression of atherosclerosis [135,136]. Recent studies have revealed the mechanism by which PPARs and LXRs regulate the inflammation process through some inflammatory target genes. Activation of PPARs and LXR can inhibit lipopolysaccharide- and cytokine-induced pro-inflammatory gene expression by repressing the toll-like receptor (TLR)-nuclear factor kappa B (NF-κB) signal pathway [137,138,139]. PPARα increases the expression of inhibitor of kappa B (IκB) to antagonize the NF-κB signalling pathway [140]. PPARβ/δ induces transforming growth factor beta (TGF-β) and inhibits the activation of NF-κB, thus regulating inflammatory processes [141]. Thiazolidinediones (TZDs) induced PPARγ activation also reduced the expression of inflammatory factors, including TNF-α and gelatinase B, in the aortic root, thus inhibiting the development of atherosclerosis [142]. All three PPAR isoforms regulate the immune response through different cell-signalling systems. LXRs repress inflammatory pathways through their transcriptional mechanisms [143,144]. LXRs and PPARγ control immunity by mediating proinflammatory gene transrepression through parallel small ubiquitin-like modifier (SUMO) ylation-dependent pathways [145]. PPARs and LXRs have been a critical interface for inflammation and cholesterol homeostasis. Concurrent activation of LXR and PPAR may have some beneficial effects. Activation of LXR by TO901317 and PPARα by fenofibrate in combination improves glucose tolerance, alleviates insulin resistance, and blocks TO901317-induced hyperlipidaemia, but aggravates hepatic steatosis in high fat diet-induced obese mice [146]. TO901317 and fenofibrate are both potent agonists. Concurrent partial agonists of LXR and PPAR may keep beneficial characteristics with few side effects. In our recently study, DBZ, as a promising therapeutic agent for atherogenesis and obesity in the mouse models, inhibits inflammation, macrophage migration, and foam cell formation, possibly through the partial activation of both PPARγ and LXRs.

5. Conclusions

PPARs, CAR, and LXRs are a part of nuclear hormone receptors that form heterodimers with RXR to regulate lipid metabolism. Ligand binding results in DNA binding and then triggers target gene expression. Obesity and atherosclerosis are both chronic lipid metabolic disorders, which were traditionally regarded as lipid deposition diseases, principally involving triglycerides in adipose tissue and cholesterol ester in arteries. Although they are distinct conditions, obesity is often associated with atherosclerosis. Recent findings have revealed the biological roles and mechanisms of these three NRs in obesity and atherosclerosis. These receptors have been potential therapeutic targets for drug discovery; further clarification and consideration of the internal relationship between them is necessary. In this study, we summarized the interaction of PPARs and CAR in lipid metabolism and obesity-related metabolic syndrome, and the cross-talk between PPARs and LXRs in cholesterol homeostasis and atherosclerosis (Figure 2). Concurrent activation of these NRs may have some beneficial effects in lipid metabolic disease. In recently study, we reported that DBZ prevented high fat diet-induced obesity and related metabolic disorders and attenuated atherosclerosis through concurrent partial activation of both PPARγ and LXRs. Moreover, it had no apparent side effects.
Beyond these cross-talks, more NRs, such as PXR, farnesoid X receptor (FXR), aryl hydrocarbon receptor (AhR), and retinoid-related orphan receptors (RORs), are being investigated. Future studies should focus on the complex network between these NRs and how that network affects their functions. We hope that by establishing a better understanding of nuclear receptor cross-talk between metabolic disorder diseases, we can reveal promising therapeutic targets for future research.

Acknowledgments

This study was supported by grants from the National Natural Science Foundation of China (Nos. 31571164 and 31271207 to Yonggong Zhai); the Natural Science Foundation of Inner Mongolia (No. 2017BS0801 to Jing Wang); the Program for Young Talents of Science and Technology in Universities of Inner Mongolia (No. NJYT-18-B29 to Jing Wang); and the Scientific Research Projection of Higher Schools of Inner Mongolia (No. NJZY17294 to Jing Wang).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. WHO. Available online: http://www.who.int/mediacentre/factsheets/fs311/en/ (accessed on 28 February 2018).
  2. Xu, P.; Wang, J.; Hong, F.; Wang, S.; Jin, X.; Xue, T.; Jia, L.; Zhai, Y. Melatonin prevents obesity through modulation of gut microbiota in mice. J. Pineal Res. 2017. [Google Scholar] [CrossRef] [PubMed]
  3. Lovren, F.; Teoh, H.; Verma, S. Obesity and atherosclerosis: Mechanistic insights. Can. J. Cardiol. 2015, 31, 177–183. [Google Scholar] [CrossRef] [PubMed]
  4. Xu, P.; Hong, F.; Wang, J.; Cong, Y.; Dai, S.; Wang, S.; Wang, J.; Jin, X.; Wang, F.; Liu, J.; et al. Microbiome remodeling via the montmorillonite adsorption-excretion axis prevents obesity-related metabolic disorders. EBioMedicine 2017, 16, 251–261. [Google Scholar] [CrossRef] [PubMed]
  5. Xu, P.F.; Dai, S.; Wang, J.; Zhang, J.; Liu, J.; Wang, F.; Zhai, Y.G. Preventive obesity agent montmorillonite adsorbs dietary lipids and enhances lipid excretion from the digestive tract. Sci. Rep. 2016, 6. [Google Scholar] [CrossRef] [PubMed]
  6. Bloomgarden, Z.T. International diabetes federation meeting, 1997, and other recent meetings. atherosclerosis and related topics. Diabetes Care 1998, 21, 1356–1363. [Google Scholar] [CrossRef] [PubMed]
  7. Zhao, Y.; Zhang, K.; Giesy, J.P.; Hu, J. Families of nuclear receptors in vertebrate models: Characteristic and comparative toxicological perspective. Sci. Rep. 2015, 5. [Google Scholar] [CrossRef] [PubMed]
  8. Xiao, L.; Wang, J.; Jiang, M.; Xie, W.; Zhai, Y. The emerging role of constitutive androstane receptor and its cross talk with liver X receptors and peroxisome proliferator-activated receptor A in lipid metabolism. Vitam. Horm. 2013, 91, 243–258. [Google Scholar] [PubMed]
  9. Germain, P.; Staels, B.; Dacquet, C.; Spedding, M.; Laudet, V. Overview of nomenclature of nuclear receptors. Pharmacol. Rev. 2006, 58, 685–704. [Google Scholar] [CrossRef] [PubMed]
  10. Horwitz, K.B.; Jackson, T.A.; Bain, D.L.; Richer, J.K.; Takimoto, G.S.; Tung, L. Nuclear receptor coactivators and corepressors. Mol. Endocrinol. 1996, 10, 1167–1177. [Google Scholar] [PubMed]
  11. Chai, X.; Zeng, S.; Xie, W. Nuclear receptors PXR and CAR: Implications for drug metabolism regulation, pharmacogenomics and beyond. Expert Opin. Drug. Metab. Toxicol. 2013, 9, 253–266. [Google Scholar] [CrossRef] [PubMed]
  12. Gross, B.; Pawlak, M.; Lefebvre, P.; Staels, B. PPARs in obesity-induced T2DM, dyslipidaemia and NAFLD. Nat. Rev. Endocrinol. 2017, 13, 36–49. [Google Scholar] [CrossRef] [PubMed]
  13. Wahli, W.; Michalik, L. PPARs at the crossroads of lipid signaling and inflammation. Trends Endocrinol. Metab. 2012, 23, 351–363. [Google Scholar] [CrossRef] [PubMed]
  14. Yan, J.; Chen, B.; Lu, J.; Xie, W. Deciphering the roles of the constitutive androstane receptor in energy metabolism. Acta Pharmacol. Sin. 2015, 36, 62–70. [Google Scholar] [CrossRef] [PubMed]
  15. Gao, J.; He, J.H.; Zhai, Y.G.; Wada, T.R.; Xie, W. The constitutive androstane receptor is an anti-obesity nuclear receptor that improves insulin sensitivity. J. Biol. Chem. 2009, 284, 25984–25992. [Google Scholar] [CrossRef] [PubMed]
  16. Xiao, L.; Xie, X.; Zhai, Y. Functional crosstalk of CAR-LXR and ROR-LXR in drug metabolism and lipid metabolism. Adv. Drug Deliv. Rev. 2010, 62, 1316–1321. [Google Scholar] [CrossRef] [PubMed]
  17. Grimaldi, P.A. Peroxisome proliferator-activated receptors as sensors of fatty acids and derivatives. Cell. Mol. Life Sci. 2007, 64, 2459–2564. [Google Scholar] [CrossRef] [PubMed]
  18. Evans, R.M.; Barish, G.D.; Wang, Y.X. PPARs and the complex journey to obesity. Nat. Med. 2004, 10, 355–361. [Google Scholar] [CrossRef] [PubMed]
  19. Tyagi, S.; Gupta, P.; Saini, A.S.; Kaushal, C.; Sharma, S. The peroxisome proliferator-activated receptor: A family of nuclear receptors role in various diseases. J. Adv. Pharm. Technol. Res. 2011, 2, 236–240. [Google Scholar] [CrossRef] [PubMed]
  20. Kersten, S. Integrated physiology and systems biology of PPARalpha. Mol. Metab. 2014, 3, 354–371. [Google Scholar] [CrossRef] [PubMed]
  21. Kersten, S.; Seydoux, J.; Peters, J.M.; Gonzalez, F.J.; Desvergne, B.; Wahli, W. Peroxisome proliferator-activated receptor alpha mediates the adaptive response to fasting. J. Clin. Investig. 1999, 103, 1489–1498. [Google Scholar] [CrossRef] [PubMed]
  22. Grygiel-Gorniak, B. Peroxisome proliferator-activated receptors and their ligands: Nutritional and clinical implications—A review. Nutr. J. 2014, 13. [Google Scholar] [CrossRef] [PubMed]
  23. Luquet, S.; Lopez-Soriano, J.; Holst, D.; Fredenrich, A.; Melki, J.; Rassoulzadegan, M.; Grimaldi, P.A. Peroxisome proliferator-activated receptor delta controls muscle development and oxidative capability. FASEB J. 2003, 17, 2299–2301. [Google Scholar] [CrossRef] [PubMed]
  24. Wang, Y.X.; Zhang, C.L.; Yu, R.T.; Cho, H.K.; Nelson, M.C.; Bayuga-Ocampo, C.R.; Ham, J.; Kang, H.; Evans, R.M. Regulation of muscle fiber type and running endurance by PPARdelta. PLoS Biol. 2004, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Schuler, M.; Ali, F.; Chambon, C.; Duteil, D.; Bornert, J.M.; Tardivel, A.; Desvergne, B.; Wahli, W.; Chambon, P.; Metzger, D. PGC1alpha expression is controlled in skeletal muscles by PPARbeta, whose ablation results in fiber-type switching, obesity, and type 2 diabetes. Cell Metab. 2006, 4, 407–414. [Google Scholar] [CrossRef] [PubMed]
  26. Palomer, X.; Barroso, E.; Pizarro-Delgado, J.; Pena, L.; Botteri, G.; Zarei, M.; Aguilar, D.; Montori-Grau, M.; Vazquez-Carrera, M. PPARbeta/delta: A key therapeutic target in metabolic disorders. Int. J. Mol. Sci. 2018, 19, 913. [Google Scholar] [CrossRef] [PubMed]
  27. Poirier, H.; Niot, I.; Monnot, M.C.; Braissant, O.; Meunier-Durmort, C.; Costet, P.; Pineau, T.; Wahli, W.; Willson, T.M.; Besnard, P. Differential involvement of peroxisome-proliferator-activated receptors alpha and delta in fibrate and fatty-acid-mediated inductions of the gene encoding liver fatty-acid-binding protein in the liver and the small intestine. Biochem. J. 2001, 355, 481–488. [Google Scholar] [CrossRef] [PubMed]
  28. Park, Y.K.; Wang, L.; Giampietro, A.; Lai, B.; Lee, J.E.; Ge, K. Distinct roles of transcription factors KLF4, Krox20, and peroxisome proliferator-activated receptor gamma in adipogenesis. Mol. Cell. Biol. 2017, 37. [Google Scholar] [CrossRef] [PubMed]
  29. Meirhaeghe, A.; Fajas, L.; Gouilleux, F.; Cottel, D.; Helbecque, N.; Auwerx, J.; Amouyel, P. A functional polymorphism in a STAT5B site of the human PPAR gamma 3 gene promoter affects height and lipid metabolism in a French population. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 289–294. [Google Scholar] [CrossRef] [PubMed]
  30. Fajas, L.; Auboeuf, D.; Raspe, E.; Schoonjans, K.; Lefebvre, A.M.; Saladin, R.; Najib, J.; Laville, M.; Fruchart, J.C.; Deeb, S.; et al. The organization, promoter analysis, and expression of the human PPARgamma gene. J. Biol. Chem. 1997, 272, 18779–18789. [Google Scholar] [CrossRef] [PubMed]
  31. Tachibana, K.; Yamasaki, D.; Ishimoto, K.; Doi, T. The role of PPARs in cancer. PPAR Res. 2008, 2008. [Google Scholar] [CrossRef] [PubMed]
  32. Lamichane, S.; Dahal Lamichane, B.; Kwon, S.M. Pivotal roles of peroxisome proliferator-activated receptors (PPARs) and their signal cascade for cellular and whole-body energy homeostasis. Int. J. Mol. Sci. 2018, 19, 949. [Google Scholar] [CrossRef] [PubMed]
  33. Tontonoz, P.; Spiegelman, B.M. Fat and beyond: The diverse biology of PPARgamma. Annu. Rev. Biochem. 2008, 77, 289–312. [Google Scholar] [CrossRef] [PubMed]
  34. Brun, R.P.; Tontonoz, P.; Forman, B.M.; Ellis, R.; Chen, J.; Evans, R.M.; Spiegelman, B.M. Differential activation of adipogenesis by multiple PPAR isoforms. Genes Dev. 1996, 10, 974–984. [Google Scholar] [CrossRef] [PubMed]
  35. Hollenberg, A.N.; Susulic, V.S.; Madura, J.P.; Zhang, B.; Moller, D.E.; Tontonoz, P.; Sarraf, P.; Spiegelman, B.M.; Lowell, B.B. Functional antagonism between CCAAT/Enhancer binding protein-alpha and peroxisome proliferator-activated receptor-gamma on the leptin promoter. J. Biol. Chem. 1997, 272, 5283–5290. [Google Scholar] [CrossRef] [PubMed]
  36. Hofmann, C.; Lorenz, K.; Braithwaite, S.S.; Colca, J.R.; Palazuk, B.J.; Hotamisligil, G.S.; Spiegelman, B.M. Altered gene expression for tumor necrosis factor-alpha and its receptors during drug and dietary modulation of insulin resistance. Endocrinology 1994, 134, 264–270. [Google Scholar] [CrossRef] [PubMed]
  37. Iwaki, M.; Matsuda, M.; Maeda, N.; Funahashi, T.; Matsuzawa, Y.; Makishima, M.; Shimomura, I. Induction of adiponectin, a fat-derived antidiabetic and antiatherogenic factor, by nuclear receptors. Diabetes 2003, 52, 1655–1663. [Google Scholar] [CrossRef] [PubMed]
  38. Duszka, K.; Oresic, M.; Le May, C.; Konig, J.; Wahli, W. PPARgamma modulates long chain fatty acid processing in the intestinal epithelium. Int. J. Mol. Sci. 2017, 18, 2559. [Google Scholar] [CrossRef] [PubMed]
  39. Ahmadian, M.; Suh, J.M.; Hah, N.; Liddle, C.; Atkins, A.R.; Downes, M.; Evans, R.M. PPARgamma signaling and metabolism: the good, the bad and the future. Nat. Med. 2013, 19, 557–566. [Google Scholar] [CrossRef] [PubMed]
  40. Xu, H.E.; Lambert, M.H.; Montana, V.G.; Parks, D.J.; Blanchard, S.G.; Brown, P.J.; Sternbach, D.D.; Lehmann, J.M.; Wisely, G.B.; Willson, T.M.; et al. Molecular recognition of fatty acids by peroxisome proliferator-activated receptors. Mol. Cell 1999, 3, 397–403. [Google Scholar] [CrossRef]
  41. Schupp, M.; Lazar, M.A. Endogenous ligands for nuclear receptors: digging deeper. J. Biol. Chem. 2010, 285, 40409–40415. [Google Scholar] [CrossRef] [PubMed]
  42. Poulsen, L.; Siersbaek, M.; Mandrup, S. PPARs: Fatty acid sensors controlling metabolism. Semin. Cell Dev. Biol. 2012, 23, 631–639. [Google Scholar] [CrossRef] [PubMed]
  43. Bensinger, S.J.; Tontonoz, P. Integration of metabolism and inflammation by lipid-activated nuclear receptors. Nature 2008, 454, 470–477. [Google Scholar] [CrossRef] [PubMed]
  44. Hennuyer, N.; Duplan, I.; Paquet, C.; Vanhoutte, J.; Woitrain, E.; Touche, V.; Colin, S.; Vallez, E.; Lestavel, S.; Lefebvre, P.; et al. The novel selective PPARalpha modulator (SPPARMalpha) pemafibrate improves dyslipidemia, enhances reverse cholesterol transport and decreases inflammation and atherosclerosis. Atherosclerosis 2016, 249, 200–208. [Google Scholar] [CrossRef] [PubMed]
  45. Khera, A.V.; Millar, J.S.; Ruotolo, G.; Wang, M.D.; Rader, D.J. Potent peroxisome proliferator-activated receptor-alpha agonist treatment increases cholesterol efflux capacity in humans with the metabolic syndrome. Eur. Heart J. 2015, 36, 3020–3022. [Google Scholar] [CrossRef] [PubMed]
  46. Jones, D.; Boudes, P.F.; Swain, M.G.; Bowlus, C.L.; Galambos, M.R.; Bacon, B.R.; Doerffel, Y.; Gitlin, N.; Gordon, S.C.; Odin, J.A.; et al. Seladelpar (MBX-8025), a selective PPAR-delta agonist, in patients with primary biliary cholangitis with an inadequate response to ursodeoxycholic acid: A double-blind, randomised, placebo-controlled, phase 2, proof-of-concept study. Lancet Gastroenterol. Hepatol. 2017, 2, 716–726. [Google Scholar] [CrossRef]
  47. Iwaisako, K.; Haimerl, M.; Paik, Y.H.; Taura, K.; Kodama, Y.; Sirlin, C.; Yu, E.; Yu, R.T.; Downes, M.; Evans, R.M.; et al. Protection from liver fibrosis by a peroxisome proliferator-activated receptor delta agonist. Proc. Natl. Acad. Sci. USA 2012, 109, E1369–E1376. [Google Scholar] [CrossRef] [PubMed]
  48. Atanasov, A.G.; Wang, J.N.; Gu, S.P.; Bu, J.; Kramer, M.P.; Baumgartner, L.; Fakhrudin, N.; Ladurner, A.; Malainer, C.; Vuorinen, A.; et al. Honokiol: A non-adipogenic PPARgamma agonist from nature. Biochim. Biophys. Acta 2013, 1830, 4813–4819. [Google Scholar] [CrossRef] [PubMed]
  49. Weidner, C.; de Groot, J.C.; Prasad, A.; Freiwald, A.; Quedenau, C.; Kliem, M.; Witzke, A.; Kodelja, V.; Han, C.T.; Giegold, S.; et al. Amorfrutins are potent antidiabetic dietary natural products. Proc. Natl. Acad. Sci. USA 2012, 109, 7257–7262. [Google Scholar] [CrossRef] [PubMed]
  50. Weidner, C.; Wowro, S.J.; Freiwald, A.; Kawamoto, K.; Witzke, A.; Kliem, M.; Siems, K.; Muller-Kuhrt, L.; Schroeder, F.C.; Sauer, S. Amorfrutin B is an efficient natural peroxisome proliferator-activated receptor gamma (PPARgamma) agonist with potent glucose-lowering properties. Diabetologia 2013, 56, 1802–1812. [Google Scholar] [CrossRef] [PubMed]
  51. Xu, P.; Hong, F.; Wang, J.; Wang, J.; Zhao, X.; Wang, S.; Xue, T.; Xu, J.; Zheng, X.; Zhai, Y. DBZ is a putative PPARgamma agonist that prevents high fat diet-induced obesity, insulin resistance and gut dysbiosis. Biochim. Biophys. Acta 2017, 1861, 2690–2701. [Google Scholar] [CrossRef] [PubMed]
  52. Wang, J.; Xu, P.; Xie, X.; Li, J.; Zhang, J.; Wang, J.; Hong, F.; Li, J.; Zhang, Y.; Song, Y.; et al. DBZ (Danshensu Bingpian Zhi), a novel natural compound derivative, attenuates atherosclerosis in apolipoprotein e-deficient mice. J. Am. Heart Assoc. 2017, 6. [Google Scholar] [CrossRef] [PubMed]
  53. Banerjee, M.; Robbins, D.; Chen, T. Targeting xenobiotic receptors PXR and CAR in human diseases. Drug. Discov. Today 2015, 20, 618–628. [Google Scholar] [CrossRef] [PubMed]
  54. Yan, J.; Xie, W. A brief history of the discovery of PXR and CAR as xenobiotic receptors. Acta Pharm. Sin. B 2016, 6, 450–452. [Google Scholar] [CrossRef] [PubMed]
  55. Choi, H.S.; Chung, M.; Tzameli, I.; Simha, D.; Lee, Y.K.; Seol, W.; Moore, D.D. Differential transactivation by two isoforms of the orphan nuclear hormone receptor CAR. J. Biol. Chem. 1997, 272, 23565–23571. [Google Scholar] [CrossRef] [PubMed]
  56. Tzameli, I.; Moore, D.D. Role reversal: New insights from new ligands for the xenobiotic receptor CAR. Trends Endocrinol. Metab. 2001, 12, 7–10. [Google Scholar] [CrossRef]
  57. Cherrington, N.J.; Hartley, D.P.; Li, N.; Johnson, D.R.; Klaassen, C.D. Organ distribution of multidrug resistance proteins 1, 2, and 3 (Mrp1, 2, and 3) mRNA and hepatic induction of Mrp3 by constitutive androstane receptor activators in rats. J. Pharmacol. Exp. Ther. 2002, 300, 97–104. [Google Scholar] [CrossRef] [PubMed]
  58. Ding, X.; Lichti, K.; Kim, I.; Gonzalez, F.J.; Staudinger, J.L. Regulation of constitutive androstane receptor and its target genes by fasting, cAMP, hepatocyte nuclear factor alpha, and the coactivator peroxisome proliferator-activated receptor gamma coactivator-1alpha. J. Biol. Chem. 2006, 281, 26540–26551. [Google Scholar] [CrossRef] [PubMed]
  59. Gao, J.; Xie, W. Pregnane X receptor and constitutive androstane receptor at the crossroads of drug metabolism and energy metabolism. Drug Metab. Dispos. 2010, 38, 2091–2095. [Google Scholar] [CrossRef] [PubMed]
  60. Sberna, A.L.; Assem, M.; Xiao, R.; Ayers, S.; Gautier, T.; Guiu, B.; Deckert, V.; Chevriaux, A.; Grober, J.; Le Guern, N.; et al. Constitutive androstane receptor activation decreases plasma apolipoprotein B-containing lipoproteins and atherosclerosis in low-density lipoprotein receptor-deficient mice. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 2232–2239. [Google Scholar] [CrossRef] [PubMed]
  61. Sberna, A.L.; Assem, M.; Gautier, T.; Grober, J.; Guiu, B.; Jeannin, A.; Pais de Barros, J.P.; Athias, A.; Lagrost, L.; Masson, D. Constitutive androstane receptor activation stimulates faecal bile acid excretion and reverse cholesterol transport in mice. J. Hepatol. 2011, 55, 154–161. [Google Scholar] [CrossRef] [PubMed]
  62. Dong, B.; Saha, P.K.; Huang, W.; Chen, W.; Abu-Elheiga, L.A.; Wakil, S.J.; Stevens, R.D.; Ilkayeva, O.; Newgard, C.B.; Chan, L.; et al. Activation of nuclear receptor CAR ameliorates diabetes and fatty liver disease. Proc. Natl. Acad. Sci. USA 2009, 106, 18831–18836. [Google Scholar] [CrossRef] [PubMed]
  63. Lynch, C.; Pan, Y.; Li, L.; Heyward, S.; Moeller, T.; Swaan, P.W.; Wang, H. Activation of the constitutive androstane receptor inhibits gluconeogenesis without affecting lipogenesis or fatty acid synthesis in human hepatocytes. Toxicol. Appl. Pharmacol. 2014, 279, 33–42. [Google Scholar] [CrossRef] [PubMed]
  64. Dong, B.; Qatanani, M.; Moore, D.D. Constitutive androstane receptor mediates the induction of drug metabolism in mouse models of type 1 diabetes. Hepatology 2009, 50, 622–629. [Google Scholar] [CrossRef] [PubMed]
  65. Jiang, M.; Xie, W. Role of the constitutive androstane receptor in obesity and type 2 diabetes: A case study of the endobiotic function of a xenobiotic receptor. Drug Metab. Rev. 2013, 45, 156–163. [Google Scholar] [CrossRef] [PubMed]
  66. Cherian, M.T.; Chai, S.C.; Chen, T. Small-molecule modulators of the constitutive androstane receptor. Expert Opin. Drug Metab. Toxicol. 2015, 11, 1099–1114. [Google Scholar] [CrossRef] [PubMed]
  67. Chai, S.C.; Cherian, M.T.; Wang, Y.M.; Chen, T. Small-molecule modulators of PXR and CAR. Biochim. Biophys. Acta 2016, 1859, 1141–1154. [Google Scholar] [CrossRef] [PubMed]
  68. Tzameli, I.; Pissios, P.; Schuetz, E.G.; Moore, D.D. The xenobiotic compound 1, 4-bis[2-(3,5-dichloropyridyloxy)]benzene is an agonist ligand for the nuclear receptor CAR. Mol. Cell. Biol. 2000, 20, 2951–2958. [Google Scholar] [CrossRef] [PubMed]
  69. Maglich, J.M.; Parks, D.J.; Moore, L.B.; Collins, J.L.; Goodwin, B.; Billin, A.N.; Stoltz, C.A.; Kliewer, S.A.; Lambert, M.H.; Willson, T.M.; et al. Identification of a novel human constitutive androstane receptor (CAR) agonist and its use in the identification of CAR target genes. J. Biol. Chem. 2003, 278, 17277–17283. [Google Scholar] [CrossRef] [PubMed]
  70. Lahtela, J.T.; Arranto, A.J.; Sotaniemi, E.A. Enzyme inducers improve insulin sensitivity in non-insulin-dependent diabetic subjects. Diabetes 1985, 34, 911–916. [Google Scholar] [CrossRef] [PubMed]
  71. Karvonen, I.; Stengard, J.H.; Huupponen, R.; Stenback, F.G.; Sotaniemi, E.A. Effects of enzyme induction therapy on glucose and drug metabolism in obese mice model of non-insulin dependent diabetes mellitus. Diabetes Res. 1989, 10, 85–92. [Google Scholar] [PubMed]
  72. Sotaniemi, E.A.; Karvonen, I. Glucose tolerance and insulin response to glucose load before and after enzyme inducing therapy in subjects with glucose intolerance and patients with NIDDM having hyperinsulinemia or relative insulin deficiency. Diabetes Res. 1989, 11, 131–139. [Google Scholar] [PubMed]
  73. Roth, A.; Looser, R.; Kaufmann, M.; Blattler, S.M.; Rencurel, F.; Huang, W.; Moore, D.D.; Meyer, U.A. Regulatory cross-talk between drug metabolism and lipid homeostasis: constitutive androstane receptor and pregnane X receptor increase Insig-1 expression. Mol. Pharmacol. 2008, 73, 1282–1289. [Google Scholar] [CrossRef] [PubMed]
  74. Zhai, Y.; Wada, T.; Zhang, B.; Khadem, S.; Ren, S.; Kuruba, R.; Li, S.; Xie, W. A functional cross-talk between liver X receptor-alpha and constitutive androstane receptor links lipogenesis and xenobiotic responses. Mol. Pharmacol. 2010, 78, 666–674. [Google Scholar] [CrossRef] [PubMed]
  75. Whitney, K.D.; Watson, M.A.; Goodwin, B.; Galardi, C.M.; Maglich, J.M.; Wilson, J.G.; Willson, T.M.; Collins, J.L.; Kliewer, S.A. Liver X receptor (LXR) regulation of the LXRalpha gene in human macrophages. J. Biol. Chem. 2001, 276, 43509–43515. [Google Scholar] [CrossRef] [PubMed]
  76. Janowski, B.A.; Grogan, M.J.; Jones, S.A.; Wisely, G.B.; Kliewer, S.A.; Corey, E.J.; Mangelsdorf, D.J. Structural requirements of ligands for the oxysterol liver X receptors LXRalpha and LXRbeta. Proc. Natl. Acad. Sci. USA 1999, 96, 266–271. [Google Scholar] [CrossRef] [PubMed]
  77. Chuu, C.P.; Kokontis, J.M.; Hiipakka, R.A.; Liao, S. Modulation of liver X receptor signaling as novel therapy for prostate cancer. J. Biomed. Sci. 2007, 14, 543–553. [Google Scholar] [CrossRef] [PubMed]
  78. Jakobsson, T.; Treuter, E.; Gustafsson, J.A.; Steffensen, K.R. Liver X receptor biology and pharmacology: New pathways, challenges and opportunities. Trends Pharmacol. Sci. 2012, 33, 394–404. [Google Scholar] [CrossRef] [PubMed]
  79. Bonamassa, B.; Moschetta, A. Atherosclerosis: Lessons from LXR and the intestine. Trends Endocrinol. Metab. 2013, 24, 120–128. [Google Scholar] [CrossRef] [PubMed]
  80. Lee, S.D.; Tontonoz, P. Liver X receptors at the intersection of lipid metabolism and atherogenesis. Atherosclerosis 2015, 242, 29–36. [Google Scholar] [CrossRef] [PubMed]
  81. Ma, Z.; Deng, C.; Hu, W.; Zhou, J.; Fan, C.; Di, S.; Liu, D.; Yang, Y.; Wang, D. Liver X receptors and their agonists: Targeting for cholesterol homeostasis and cardiovascular diseases. Curr. Issues Mol. Biol. 2017, 22, 41–64. [Google Scholar] [CrossRef] [PubMed]
  82. Repa, J.J.; Turley, S.D.; Lobaccaro, J.A.; Medina, J.; Li, L.; Lustig, K.; Shan, B.; Heyman, R.A.; Dietschy, J.M.; Mangelsdorf, D.J. Regulation of absorption and ABC1-mediated efflux of cholesterol by RXR heterodimers. Science 2000, 289, 1524–1529. [Google Scholar] [CrossRef] [PubMed]
  83. Repa, J.J.; Berge, K.E.; Pomajzl, C.; Richardson, J.A.; Hobbs, H.; Mangelsdorf, D.J. Regulation of ATP-binding cassette sterol transporters ABCG5 and ABCG8 by the liver X receptors alpha and beta. J. Biol. Chem. 2002, 277, 18793–18800. [Google Scholar] [CrossRef] [PubMed]
  84. Laffitte, B.A.; Repa, J.J.; Joseph, S.B.; Wilpitz, D.C.; Kast, H.R.; Mangelsdorf, D.J.; Tontonoz, P. LXRs control lipid-inducible expression of the apolipoprotein E gene in macrophages and adipocytes. Proc. Natl. Acad. Sci. USA 2001, 98, 507–512. [Google Scholar] [CrossRef] [PubMed]
  85. Repa, J.J.; Liang, G.; Ou, J.; Bashmakov, Y.; Lobaccaro, J.M.; Shimomura, I.; Shan, B.; Brown, M.S.; Goldstein, J.L.; Mangelsdorf, D.J. Regulation of mouse sterol regulatory element-binding protein-1c gene (SREBP-1c) by oxysterol receptors, LXRalpha and LXRbeta. Genes Dev. 2000, 14, 2819–2830. [Google Scholar] [CrossRef] [PubMed]
  86. Joseph, S.B.; Laffitte, B.A.; Patel, P.H.; Watson, M.A.; Matsukuma, K.E.; Walczak, R.; Collins, J.L.; Osborne, T.F.; Tontonoz, P. Direct and indirect mechanisms for regulation of fatty acid synthase gene expression by liver X receptors. J. Biol. Chem. 2002, 277, 11019–11025. [Google Scholar] [CrossRef] [PubMed]
  87. Talukdar, S.; Hillgartner, F.B. The mechanism mediating the activation of acetyl-coenzyme a carboxylase-alpha gene transcription by the liver X receptor agonist T0–901317. J. Lipid Res. 2006, 47, 2451–2461. [Google Scholar] [CrossRef] [PubMed]
  88. Janowski, B.A.; Willy, P.J.; Devi, T.R.; Falck, J.R.; Mangelsdorf, D.J. An oxysterol signalling pathway mediated by the nuclear receptor LXR alpha. Nature 1996, 383, 728–731. [Google Scholar] [CrossRef] [PubMed]
  89. Spencer, T.A.; Gayen, A.K.; Phirwa, S.; Nelson, J.A.; Taylor, F.R.; Kandutsch, A.A.; Erickson, S.K. 24(S), 25-Epoxycholesterol. Evidence consistent with a role in the regulation of hepatic cholesterogenesis. J. Biol. Chem. 1985, 260, 13391–13394. [Google Scholar] [PubMed]
  90. Fu, X.; Menke, J.G.; Chen, Y.; Zhou, G.; MacNaul, K.L.; Wright, S.D.; Sparrow, C.P.; Lund, E.G. 27-hydroxycholesterol is an endogenous ligand for liver X receptor in cholesterol-loaded cells. J. Biol. Chem. 2001, 276, 38378–38387. [Google Scholar] [CrossRef] [PubMed]
  91. Hong, C.; Tontonoz, P. Liver X receptors in lipid metabolism: Opportunities for drug discovery. Nat. Rev. Drug Discov. 2014, 13, 433–444. [Google Scholar] [CrossRef] [PubMed]
  92. Bradley, M.N.; Hong, C.; Chen, M.; Joseph, S.B.; Wilpitz, D.C.; Wang, X.; Lusis, A.J.; Collins, A.; Hseuh, W.A.; Collins, J.L.; et al. Ligand activation of LXR beta reverses atherosclerosis and cellular cholesterol overload in mice lacking LXR alpha and apoE. J. Clin. Investig. 2007, 117, 2337–2346. [Google Scholar] [CrossRef] [PubMed]
  93. Kiss, E.; Popovic, Z.; Bedke, J.; Wang, S.; Bonrouhi, M.; Gretz, N.; Stettner, P.; Teupser, D.; Thiery, J.; Porubsky, S.; et al. Suppression of chronic damage in renal allografts by Liver X receptor (LXR) activation relevant contribution of macrophage LXRalpha. Am. J. Pathol. 2011, 179, 92–103. [Google Scholar] [CrossRef] [PubMed]
  94. Peet, D.J.; Turley, S.D.; Ma, W.; Janowski, B.A.; Lobaccaro, J.M.; Hammer, R.E.; Mangelsdorf, D.J. Cholesterol and bile acid metabolism are impaired in mice lacking the nuclear oxysterol receptor LXR alpha. Cell 1998, 93, 693–704. [Google Scholar] [CrossRef]
  95. Katz, A.; Udata, C.; Ott, E.; Hickey, L.; Burczynski, M.E.; Burghart, P.; Vesterqvist, O.; Meng, X. Safety, pharmacokinetics, and pharmacodynamics of single doses of LXR-623, a novel liver X-receptor agonist, in healthy participants. J. Clin. Pharmacol. 2009, 49, 643–649. [Google Scholar] [CrossRef] [PubMed]
  96. Quinet, E.M.; Basso, M.D.; Halpern, A.R.; Yates, D.W.; Steffan, R.J.; Clerin, V.; Resmini, C.; Keith, J.C.; Berrodin, T.J.; Feingold, I.; et al. LXR ligand lowers LDL cholesterol in primates, is lipid neutral in hamster, and reduces atherosclerosis in mouse. J. Lipid Res. 2009, 50, 2358–2370. [Google Scholar] [CrossRef] [PubMed]
  97. Li, X.; Yeh, V.; Molteni, V. Liver X receptor modulators: A review of recently patented compounds (2007–2009). Expert Opin. Ther. Pat. 2010, 20, 535–562. [Google Scholar] [CrossRef] [PubMed]
  98. Loren, J.; Huang, Z.; Laffitte, B.A.; Molteni, V. Liver X receptor modulators: A review of recently patented compounds (2009–2012). Expert Opin. Ther. Pat. 2013, 23, 1317–1335. [Google Scholar] [CrossRef] [PubMed]
  99. Dahlman, I.; Nilsson, M.; Gu, H.F.; Lecoeur, C.; Efendic, S.; Ostenson, C.G.; Brismar, K.; Gustafsson, J.A.; Froguel, P.; Vaxillaire, M.; et al. Functional and genetic analysis in type 2 diabetes of liver X receptor alleles—A cohort study. BMC Med. Genet. 2009, 10. [Google Scholar] [CrossRef] [PubMed]
  100. Xie, X.; Wang, S.; Xiao, L.; Zhang, J.; Wang, J.; Liu, J.; Shen, X.; He, D.; Zheng, X.; Zhai, Y. DBZ blocks LPS-induced monocyte activation and foam cell formation via inhibiting nuclear factor-kB. Cell. Physiol. Biochem. 2011, 28, 649–662. [Google Scholar] [CrossRef] [PubMed]
  101. Wang, Y.X.; Lee, C.H.; Tiep, S.; Yu, R.T.; Ham, J.; Kang, H.; Evans, R.M. Peroxisome-proliferator-activated receptor delta activates fat metabolism to prevent obesity. Cell 2003, 113, 159–170. [Google Scholar] [CrossRef]
  102. Grimaldi, P.A. Regulatory functions of PPARbeta in metabolism: Implications for the treatment of metabolic syndrome. Biochim. Biophys. Acta 2007, 1771, 983–990. [Google Scholar] [CrossRef] [PubMed]
  103. Oliver, W.R., Jr.; Shenk, J.L.; Snaith, M.R.; Russell, C.S.; Plunket, K.D.; Bodkin, N.L.; Lewis, M.C.; Winegar, D.A.; Sznaidman, M.L.; Lambert, M.H.; et al. A selective peroxisome proliferator-activated receptor delta agonist promotes reverse cholesterol transport. Proc. Natl. Acad. Sci. USA 2001, 98, 5306–5311. [Google Scholar] [CrossRef] [PubMed]
  104. Rosen, E.D.; Sarraf, P.; Troy, A.E.; Bradwin, G.; Moore, K.; Milstone, D.S.; Spiegelman, B.M.; Mortensen, R.M. PPAR gamma is required for the differentiation of adipose tissue in vivo and in vitro. Mol. Cell 1999, 4, 611–617. [Google Scholar] [CrossRef]
  105. Barak, Y.; Nelson, M.C.; Ong, E.S.; Jones, Y.Z.; Ruiz-Lozano, P.; Chien, K.R.; Koder, A.; Evans, R.M. PPAR gamma is required for placental, cardiac, and adipose tissue development. Mol. Cell 1999, 4, 585–595. [Google Scholar] [CrossRef]
  106. Kubota, N.; Terauchi, Y.; Miki, H.; Tamemoto, H.; Yamauchi, T.; Komeda, K.; Satoh, S.; Nakano, R.; Ishii, C.; Sugiyama, T.; et al. PPAR gamma mediates high-fat diet-induced adipocyte hypertrophy and insulin resistance. Mol. Cell 1999, 4, 597–609. [Google Scholar] [CrossRef]
  107. Contreras, A.V.; Torres, N.; Tovar, A.R. PPAR-alpha as a key nutritional and environmental sensor for metabolic adaptation. Adv. Nutr. 2013, 4, 439–452. [Google Scholar] [CrossRef] [PubMed]
  108. Feng, X.; Gao, X.; Jia, Y.; Zhang, H.; Xu, Y.; Wang, G. PPAR-alpha agonist fenofibrate decreased RANTES levels in type 2 diabetes patients with hypertriglyceridemia. Med. Sci. Monit. 2016, 22, 743–751. [Google Scholar] [CrossRef] [PubMed]
  109. Lamers, C.; Schubert-Zsilavecz, M.; Merk, D. Therapeutic modulators of peroxisome proliferator-activated receptors (PPAR): A patent review (2008-present). Expert Opin. Ther. Pat. 2012, 22, 803–841. [Google Scholar] [CrossRef] [PubMed]
  110. Pawlak, M.; Lefebvre, P.; Staels, B. Molecular mechanism of PPARalpha action and its impact on lipid metabolism, inflammation and fibrosis in non-alcoholic fatty liver disease. J. Hepatol. 2015, 62, 720–733. [Google Scholar] [CrossRef] [PubMed]
  111. Yoon, M. The role of PPARalpha in lipid metabolism and obesity: Focusing on the effects of estrogen on PPARalpha actions. Pharmacol. Res. 2009, 60, 151–159. [Google Scholar] [CrossRef] [PubMed]
  112. Yoon, M. PPARalpha in obesity: Sex difference and estrogen involvement. PPAR Res. 2010, 2010. [Google Scholar] [CrossRef] [PubMed]
  113. Xu, P.; Hong, F.; Wang, J.; Dai, S.; Wang, J.; Zhai, Y. The CAR agonist TCPOBOP inhibits lipogenesis and promotes fibrosis in the mammary gland of adolescent female mice. Toxicol. Lett. 2018, 290, 29–35. [Google Scholar] [CrossRef] [PubMed]
  114. Valero, T. Mitochondrial biogenesis: Pharmacological approaches. Curr. Pharm. Des. 2014, 20, 5507–5509. [Google Scholar] [CrossRef] [PubMed]
  115. Sanchis-Gomar, F.; Garcia-Gimenez, J.L.; Gomez-Cabrera, M.C.; Pallardo, F.V. Mitochondrial biogenesis in health and disease. Molecular and therapeutic approaches. Curr. Pharm. Des. 2014, 20, 5619–5633. [Google Scholar] [CrossRef] [PubMed]
  116. Gao, J.; Yan, J.; Xu, M.; Ren, S.; Xie, W. CAR suppresses hepatic gluconeogenesis by facilitating the ubiquitination and degradation of PGC1alpha. Mol. Endocrinol. 2015, 29, 1558–1570. [Google Scholar] [CrossRef] [PubMed]
  117. Wieneke, N.; Hirsch-Ernst, K.I.; Kuna, M.; Kersten, S.; Puschel, G.P. PPARalpha-dependent induction of the energy homeostasis-regulating nuclear receptor NR1i3 (CAR) in rat hepatocytes: Potential role in starvation adaptation. FEBS Lett. 2007, 581, 5617–5626. [Google Scholar] [CrossRef] [PubMed]
  118. Wada, T.; Gao, J.; Xie, W. PXR and CAR in energy metabolism. Trends Endocrinol. Metab. 2009, 20, 273–279. [Google Scholar] [CrossRef] [PubMed]
  119. Guo, D.; Sarkar, J.; Suino-Powell, K.; Xu, Y.; Matsumoto, K.; Jia, Y.; Yu, S.; Khare, S.; Haldar, K.; Rao, M.S.; et al. Induction of nuclear translocation of constitutive androstane receptor by peroxisome proliferator-activated receptor alpha synthetic ligands in mouse liver. J. Biol. Chem. 2007, 282, 36766–36776. [Google Scholar] [CrossRef] [PubMed]
  120. Guo, D.; Sarkar, J.; Ahmed, M.R.; Viswakarma, N.; Jia, Y.; Yu, S.; Sambasiva Rao, M.; Reddy, J.K. Peroxisome proliferator-activated receptor (PPAR)-binding protein (PBP) but not PPAR-interacting protein (PRIP) is required for nuclear translocation of constitutive androstane receptor in mouse liver. Biochem. Biophys. Res. Commun. 2006, 347, 485–495. [Google Scholar] [CrossRef] [PubMed]
  121. Kiyosawa, N.; Tanaka, K.; Hirao, J.; Ito, K.; Niino, N.; Sakuma, K.; Kanbori, M.; Yamoto, T.; Manabe, S.; Matsunuma, N. Molecular mechanism investigation of phenobarbital-induced serum cholesterol elevation in rat livers by microarray analysis. Arch. Toxicol. 2004, 78, 435–442. [Google Scholar] [CrossRef] [PubMed]
  122. Ide, T.; Shimano, H.; Yoshikawa, T.; Yahagi, N.; Amemiya-Kudo, M.; Matsuzaka, T.; Nakakuki, M.; Yatoh, S.; Iizuka, Y.; Tomita, S.; et al. Cross-talk between peroxisome proliferator-activated receptor (PPAR) alpha and liver X receptor (LXR) in nutritional regulation of fatty acid metabolism. II. LXRs suppress lipid degradation gene promoters through inhibition of PPAR signaling. Mol. Endocrinol. 2003, 17, 1255–1267. [Google Scholar] [CrossRef] [PubMed]
  123. Parikh, M.; Patel, K.; Soni, S.; Gandhi, T. Liver X receptor: A cardinal target for atherosclerosis and beyond. J. Atheroscler. Thromb. 2014, 21, 519–531. [Google Scholar] [CrossRef] [PubMed]
  124. Yoshikawa, T.; Ide, T.; Shimano, H.; Yahagi, N.; Amemiya-Kudo, M.; Matsuzaka, T.; Yatoh, S.; Kitamine, T.; Okazaki, H.; Tamura, Y.; et al. Cross-talk between peroxisome proliferator-activated receptor (PPAR) alpha and liver X receptor (LXR) in nutritional regulation of fatty acid metabolism. I. PPARs suppress sterol regulatory element binding protein-1c promoter through inhibition of LXR signaling. Mol. Endocrinol. 2003, 17, 1240–1254. [Google Scholar] [PubMed]
  125. Miyata, K.S.; McCaw, S.E.; Patel, H.V.; Rachubinski, R.A.; Capone, J.P. The orphan nuclear hormone receptor LXR alpha interacts with the peroxisome proliferator-activated receptor and inhibits peroxisome proliferator signaling. J. Biol. Chem. 1996, 271, 9189–9192. [Google Scholar] [CrossRef] [PubMed]
  126. Yue, L.; Ye, F.; Gui, C.; Luo, H.; Cai, J.; Shen, J.; Chen, K.; Shen, X.; Jiang, H. Ligand-binding regulation of LXR/RXR and LXR/PPAR heterodimerizations: SPR technology-based kinetic analysis correlated with molecular dynamics simulation. Protein Sci. 2005, 14, 812–822. [Google Scholar] [CrossRef] [PubMed]
  127. Chinetti, G.; Lestavel, S.; Bocher, V.; Remaley, A.T.; Neve, B.; Torra, I.P.; Teissier, E.; Minnich, A.; Jaye, M.; Duverger, N.; et al. PPAR-alpha and PPAR-gamma activators induce cholesterol removal from human macrophage foam cells through stimulation of the ABCA1 pathway. Nat. Med. 2001, 7, 53–58. [Google Scholar] [CrossRef] [PubMed]
  128. Akiyama, T.E.; Sakai, S.; Lambert, G.; Nicol, C.J.; Matsusue, K.; Pimprale, S.; Lee, Y.H.; Ricote, M.; Glass, C.K.; Brewer, H.B., Jr.; et al. Conditional disruption of the peroxisome proliferator-activated receptor gamma gene in mice results in lowered expression of ABCA1, ABCG1, and apoE in macrophages and reduced cholesterol efflux. Mol. Cell. Biol. 2002, 22, 2607–2619. [Google Scholar] [CrossRef] [PubMed]
  129. Claudel, T.; Leibowitz, M.D.; Fievet, C.; Tailleux, A.; Wagner, B.; Repa, J.J.; Torpier, G.; Lobaccaro, J.M.; Paterniti, J.R.; Mangelsdorf, D.J.; et al. Reduction of atherosclerosis in apolipoprotein E knockout mice by activation of the retinoid X receptor. Proc. Natl. Acad. Sci. USA 2001, 98, 2610–2615. [Google Scholar] [CrossRef] [PubMed]
  130. Li, A.C.; Binder, C.J.; Gutierrez, A.; Brown, K.K.; Plotkin, C.R.; Pattison, J.W.; Valledor, A.F.; Davis, R.A.; Willson, T.M.; Witztum, J.L.; et al. Differential inhibition of macrophage foam-cell formation and atherosclerosis in mice by PPARalpha, beta/delta, and gamma. J. Clin. Investig. 2004, 114, 1564–1576. [Google Scholar] [CrossRef] [PubMed]
  131. Chawla, A.; Boisvert, W.A.; Lee, C.H.; Laffitte, B.A.; Barak, Y.; Joseph, S.B.; Liao, D.; Nagy, L.; Edwards, P.A.; Curtiss, L.K.; et al. A PPAR gamma-LXR-ABCA1 pathway in macrophages is involved in cholesterol efflux and atherogenesis. Mol. Cell 2001, 7, 161–171. [Google Scholar] [CrossRef]
  132. Lo Sasso, G.; Murzilli, S.; Salvatore, L.; D’Errico, I.; Petruzzelli, M.; Conca, P.; Jiang, Z.Y.; Calabresi, L.; Parini, P.; Moschetta, A. Intestinal specific LXR activation stimulates reverse cholesterol transport and protects from atherosclerosis. Cell Metab. 2010, 12, 187–193. [Google Scholar] [CrossRef] [PubMed]
  133. Umeda, Y.; Kako, Y.; Mizutani, K.; Iikura, Y.; Kawamura, M.; Seishima, M.; Hayashi, H. Inhibitory action of gemfibrozil on cholesterol absorption in rat intestine. J. Lipid Res. 2001, 42, 1214–1219. [Google Scholar] [PubMed]
  134. Valasek, M.A.; Clarke, S.L.; Repa, J.J. Fenofibrate reduces intestinal cholesterol absorption via PPARalpha-dependent modulation of NPC1L1 expression in mouse. J. Lipid Res. 2007, 48, 2725–2735. [Google Scholar] [CrossRef] [PubMed]
  135. Ross, R. Atherosclerosis—An inflammatory disease. N. Engl. J. Med. 1999, 340, 115–126. [Google Scholar] [CrossRef] [PubMed]
  136. Tuttolomondo, A.; Di Raimondo, D.; Pecoraro, R.; Arnao, V.; Pinto, A.; Licata, G. Atherosclerosis as an inflammatory disease. Curr. Pharm. Des. 2012, 18, 4266–4288. [Google Scholar] [CrossRef] [PubMed]
  137. Welch, J.S.; Ricote, M.; Akiyama, T.E.; Gonzalez, F.J.; Glass, C.K. PPARgamma and PPARdelta negatively regulate specific subsets of lipopolysaccharide and IFN-gamma target genes in macrophages. Proc. Natl. Acad. Sci. USA 2003, 100, 6712–6717. [Google Scholar] [CrossRef] [PubMed]
  138. Ogawa, S.; Lozach, J.; Benner, C.; Pascual, G.; Tangirala, R.K.; Westin, S.; Hoffmann, A.; Subramaniam, S.; David, M.; Rosenfeld, M.G.; et al. Molecular determinants of crosstalk between nuclear receptors and toll-like receptors. Cell 2005, 122, 707–721. [Google Scholar] [CrossRef] [PubMed]
  139. Fessler, M.B. The challenges and promise of targeting the Liver X Receptors for treatment of inflammatory disease. Pharmacol. Ther. 2018, 181, 1–12. [Google Scholar] [CrossRef] [PubMed]
  140. Delerive, P.; De Bosscher, K.; Vanden Berghe, W.; Fruchart, J.C.; Haegeman, G.; Staels, B. DNA binding-independent induction of IkappaBalpha gene transcription by PPARalpha. Mol. Endocrinol. 2002, 16, 1029–1039. [Google Scholar] [PubMed]
  141. Bishop-Bailey, D.; Bystrom, J. Emerging roles of peroxisome proliferator-activated receptor-beta/delta in inflammation. Pharmacol. Ther. 2009, 124, 141–150. [Google Scholar] [CrossRef] [PubMed]
  142. Li, A.C.; Brown, K.K.; Silvestre, M.J.; Willson, T.M.; Palinski, W.; Glass, C.K. Peroxisome proliferator-activated receptor gamma ligands inhibit development of atherosclerosis in LDL receptor-deficient mice. J. Clin. Investig. 2000, 106, 523–531. [Google Scholar] [CrossRef] [PubMed]
  143. Im, S.S.; Osborne, T.F. Liver x receptors in atherosclerosis and inflammation. Circ. Res. 2011, 108, 996–1001. [Google Scholar] [CrossRef] [PubMed]
  144. Steffensen, K.R.; Jakobsson, T.; Gustafsson, J.A. Targeting liver X receptors in inflammation. Expert Opin. Ther. Targets 2013, 17, 977–990. [Google Scholar] [CrossRef] [PubMed]
  145. Ghisletti, S.; Huang, W.; Ogawa, S.; Pascual, G.; Lin, M.E.; Willson, T.M.; Rosenfeld, M.G.; Glass, C.K. Parallel SUMOylation-dependent pathways mediate gene- and signal-specific transrepression by LXRs and PPARgamma. Mol. Cell 2007, 25, 57–70. [Google Scholar] [CrossRef] [PubMed]
  146. Gao, M.; Bu, L.; Ma, Y.; Liu, D. Concurrent activation of liver X receptor and peroxisome proliferator-activated receptor alpha exacerbates hepatic steatosis in high fat diet-induced obese mice. PLoS ONE 2013, 8. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic structure of NRs (nuclear receptors) and model of NR signalling. (a) General domain structure of NRs; and (b) the mechanism of general NR action. The ligands bind to the LBD (ligand-binding domain) of NRs in the cytoplasm, and translocate to the nucleus. Then the DBD (DNA-binding domain) of NRs bind to the XRE (xenobiotic responsive elements) forming dimeric complexes with RXR and the recruitment of co-activators or co-repressors. Finally, this leads to the transcription of the target genes. This model is applied to type II NRs, including PPARs, CAR, LXRs, and others. The colorful words just match the corresponding colorful shape. The dotted arrows mean different ligands can recruit coactivators or corepressors to form dimers, respectively.
Figure 1. Schematic structure of NRs (nuclear receptors) and model of NR signalling. (a) General domain structure of NRs; and (b) the mechanism of general NR action. The ligands bind to the LBD (ligand-binding domain) of NRs in the cytoplasm, and translocate to the nucleus. Then the DBD (DNA-binding domain) of NRs bind to the XRE (xenobiotic responsive elements) forming dimeric complexes with RXR and the recruitment of co-activators or co-repressors. Finally, this leads to the transcription of the target genes. This model is applied to type II NRs, including PPARs, CAR, LXRs, and others. The colorful words just match the corresponding colorful shape. The dotted arrows mean different ligands can recruit coactivators or corepressors to form dimers, respectively.
Ijms 19 01260 g001
Figure 2. Proposed model of the cross-talks between PPARs and CAR in obesity and PPARs and LXRs in atherosclerosis. Red arrows: promotion; green T-bar: inhibition; red up-arrows: up-regulation; black down-arrows: down-regulation.
Figure 2. Proposed model of the cross-talks between PPARs and CAR in obesity and PPARs and LXRs in atherosclerosis. Red arrows: promotion; green T-bar: inhibition; red up-arrows: up-regulation; black down-arrows: down-regulation.
Ijms 19 01260 g002
Table 1. Different PPAR ligands and their development status regarding the treatment of lipid and glucose metabolic syndrome.
Table 1. Different PPAR ligands and their development status regarding the treatment of lipid and glucose metabolic syndrome.
LigandsClassificationStructureIndicationCurrent Stage
BezafibratePPARα agonist Ijms 19 01260 i001HyperlipidemiaOn the market
ClofibratePPARα agonist Ijms 19 01260 i002HyperlipidemiaDiscontinued
FenofibratePPARα agonist Ijms 19 01260 i003Hypercholesterolemia, mixed dyslipidemiaOn the market
GemfibrozilPPARα agonist Ijms 19 01260 i004Hyperlipidemia, ischaemic disorderOn the market
PemafibratePPARα agonist Ijms 19 01260 i005Lipid modifying agentOn the market in Japan
LY518674PPARα agonist Ijms 19 01260 i006AtherosclerosisPhase II
Seladelpar (MBX-8025)PPARβ/δ agonist Ijms 19 01260 i007Dyslipidaemia, T2D, NASHPhase II
KD-3010PPARβ/δ agonist Ijms 19 01260 i008Diabetes, obesity, dyslipidemiaPhase I
TroglitazonePPARγ agonist Ijms 19 01260 i009T2DWithdrawn due to hepatotoxicity
RosiglitazonePPARγ agonist Ijms 19 01260 i010T2DWithdrawn due to risk of CV events
PioglitazonePPARγ agonist Ijms 19 01260 i011T2DOn the market
LobeglitazonePPARα/PPARγ agonist Ijms 19 01260 i012T2DOn the market in Korea
Balaglitazone (DRF-2593)PPARγ agonist Ijms 19 01260 i013T2DPhase III Discontinued
CiglitazonePPARγ agonist Ijms 19 01260 i014T2DPhase II Discontinued
DarglitazonePPARγ inhibitor Ijms 19 01260 i015T2DPhase I Discontinued
Netoglitazone (MCC-555)PPARα/PPARγ agonist Ijms 19 01260 i016T2DPhase II Discontinued
RivoglitazonePPARγ agonist Ijms 19 01260 i017T2DPhase III Discontinued
HonokiolPPARγ agonist Ijms 19 01260 i018Gingival diseases, anti-hyperglycemic propertyPhase III
Table 2. Different LXR ligands and their development status regarding anti-atherosclerosis.
Table 2. Different LXR ligands and their development status regarding anti-atherosclerosis.
LigandsClassificationStructureIndicationCurrent Stage
LXR-623 (WAY-252623)LXRα-partial LXRβ-full agonist Ijms 19 01260 i019AtherosclerosisPhase I Discontinued
BMS-852927 (XL-041)LXR modulator Ijms 19 01260 i020Atherosclerosis, hypercholesterolemiaPhase I Discontinued
BMS-779788 (XL-652)LXR agonist Ijms 19 01260 i021AtherosclerosisPhase I

Share and Cite

MDPI and ACS Style

Xu, P.; Zhai, Y.; Wang, J. The Role of PPAR and Its Cross-Talk with CAR and LXR in Obesity and Atherosclerosis. Int. J. Mol. Sci. 2018, 19, 1260. https://doi.org/10.3390/ijms19041260

AMA Style

Xu P, Zhai Y, Wang J. The Role of PPAR and Its Cross-Talk with CAR and LXR in Obesity and Atherosclerosis. International Journal of Molecular Sciences. 2018; 19(4):1260. https://doi.org/10.3390/ijms19041260

Chicago/Turabian Style

Xu, Pengfei, Yonggong Zhai, and Jing Wang. 2018. "The Role of PPAR and Its Cross-Talk with CAR and LXR in Obesity and Atherosclerosis" International Journal of Molecular Sciences 19, no. 4: 1260. https://doi.org/10.3390/ijms19041260

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop