Skip to main content
Liver Research logoLink to Liver Research
. 2024 Mar 12;8(1):34–45. doi: 10.1016/j.livres.2024.03.002

Targeting nuclear receptors for NASH/MASH: From bench to bedside

Rohit A Sinha 1
PMCID: PMC7615772  EMSID: EMS194792  PMID: 38544909

Abstract

The onset of metabolic dysfunction-associated steatohepatitis (MASH) or non-alcoholic steatohepatitis (NASH) represents a tipping point leading to liver injury and subsequent hepatic complications in the natural progression of what is now termed metabolic dysfunction-associated steatotic liver diseases (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD). With no pharmacological treatment currently available for MASH/NASH, the race is on to develop drugs targeting multiple facets of hepatic metabolism, inflammation, and pro-fibrotic events, which are major drivers of MASH. Nuclear receptors (NRs) regulate genomic transcription upon binding to lipophilic ligands and govern multiple aspects of liver metabolism and inflammation. Ligands of NRs may include hormones, lipids, bile acids, and synthetic ligands, which upon binding to NRs regulate the transcriptional activities of target genes. NR ligands are presently the most promising drug candidates expected to receive approval from the United States Food and Drug Administration as a pharmacological treatment for MASH. This review aims to cover the current understanding of NRs, including nuclear hormone receptors, non-steroid hormone receptors, circadian NRs, and orphan NRs, which are currently undergoing clinical trials for MASH treatment, along with NRs that have shown promising results in preclinical studies.

Keywords: Nuclear receptor (NR), Metabolic dysfunction-associated steatohepatitis (MASH), Metabolic dysfunction-associated steatotic liver disease (MASLD), Transcription factor, Liver, Drug

1. Introduction

Metabolic dysfunction-associated steatotic liver disease (MASLD)/non-alcoholic fatty liver disease (NAFLD) is one of the fastest-growing metabolic epidemics worldwide.1 A recent study published in 2023, shows an overall global prevalence of MASLD around 30.05% with an approximate increase of 50.40% from 25.26% in 1990–2006 to 38.20% in 2016–2019.2 Metabolic dysfunction-associated steatohepatitis (MASH) is a clinically advanced stage of MASLD, and is a risk factor for several end-stage liver diseases.3 MASH is now recognized as a major cause for liver transplantation and is often associated with diabetes and chronic kidney disease.4,5 It is triggered by decompensated steatosis in hepatocytes upon fatty acid and sugar influx, resulting in hepatocyte injury via a process termed “Lipotoxicity”.6 Decompensated steatosis results in the increased availability of free fatty acids in the hepatocyte cytosol, leading to lipotoxic injury by inducing mitochondrial damage, oxidative stress, endoplasmic reticulum stress, and eventually resulting in hepatocyte death. The injured or dying hepatocytes release several damage-associated molecular patterns (DAMPs), which upon binding to DAMP receptors on macrophages and hepatic stellate cells (HSCs), trigger these cells to adopt an activated phenotype, thereby leading to inflammation and fibrosis in the liver, which is a hallmark of MASH.7,8 Presently, there are no approved pharmacological treatments for MASH, and lifestyle modification remains the sole available option for its management. This review outlines the current understanding of nuclear receptors (NRs) function in the liver, their dysregulation in MASH, and the therapeutic targeting to counter MASH pathogenesis.

2. NRs pharmacology and its application in MASH treatment

Phenotypic alteration of hepatocytes in MASH is a result of dynamic changes in the transcriptome in response to extracellular cues, including nutrients, hormones, and cytokines. NRs, a ligand-regulated class of transcription factors, play a critical role as key mediators of hepatic transcriptome alteration in response to environmental stress. NRs act as ligand-activated transcriptional regulators that affect hepatic pathophysiology. In humans, 48 NRs have been defined by shared structural and functional features, including DNA-binding domains and ligand-binding domains. Among these, four classes of NRs play key roles in regulating liver metabolism. Targeting these receptors has shown preclinical and/or clinical efficacy in modulating MASH pathology.9 The first class of these NRs includes the classical hormone receptors, such as thyroid hormone receptor (THR), glucocorticoid receptor (GR), estrogen receptor (ER), vitamin D receptor (VDR), and retinoic acid receptor (RAR). The second class of NRs are non-steroid hormone receptors that are activated by lipids or their derivatives. It includes peroxisome proliferator-activated receptors (PPARs), farnesoid X receptor (FXR), liver X receptor (LXR), and pregnane X receptor (PXR), which mainly utilize dietary lipids as their ligands. The third group contains REV-ERBs and RAR-related orphan receptors (RORs) that regulate the temporal transcription of liver metabolic genes aligned with the circadian rhythm. Lastly, the fourth class of NRs includes the “orphan receptors” the endogenous ligands of which remain unidentified and include estrogen-related receptor (ERR), constitutive androstane receptor (CAR), small heterodimer partner (SHP), hepatocyte nuclear factor 4 alpha (HNF4α), and liver receptor homolog-1 (LRH-1/NR5A2). In the liver, several hormones and lipid-induced NRs heterodimerize with retinoid X receptors (RXRs) and mediate the epigenetic modulation of gene transcription upon binding to hormone response elements on the target gene promoter or enhancer region (Fig. 1).10 Besides their classical genomic action, several non-genomic actions of NRs have been described; however, their relative contribution to human physiology remains less elucidated.11 Several genes (e.g., PPARα, PPARγ, NRH14) involved in lipid and glucose metabolism, inflammation, and fibrosis are targets of NRs in the liver (Fig. 2). Therefore, NRs have been at the forefront of targeting strategies for MASH, targeting different stages of MASH progression by affecting lipid and bile metabolism, activating immune cell, and modulating pro-fibrotic signaling.

Fig. 1.

Fig. 1

Molecular mechanisms of NRaction. Nuclear receptor (NR) ligands which may include hormones, lipids, cholesterol derivatives, and xenobiotics may bind to either cytosolic or nuclear resident NRs which results in the binding of NRs to their cognate response elements such as hormone response elements (HREs) on the promoter/enhancer region of the target genes. Upon ligand binding NRs evoke a dynamic exchange of nuclear receptor corepressor (CoR) with nuclear receptor coactivator (CoA) complexes. The CoA has a histone acetylase activity which helps in the opening of the nucleosomes and initiation of RNA polymerase II (POL II) mediated transcription. The mRNA synthesized in response to NR activation further results in protein synthesis and alteration of cellular function. Besides the classical genomic action of NRs, cytosolic NRs may also induce non-genomic signaling via interaction with cytoplasmic proteins.

Fig. 2.

Fig. 2

Cell-specific NRs modulation of metabolism, inflammation, and fibrosis response in MASH. Multiple nuclear receptors (NRs) as shown in this schematic play both distinct and overlapping cellular roles in regulating diverse aspects of hepatic lipid metabolism, immunomodulation, and fibrosis response during metabolic dysfunction-associated steatohepatitis (MASH) pathogenesis via their distinct action on hepatocytes, immune cells (Kupffer cells & T cells), and hepatic stellate cells (HSCs). The green font color denotes NRs that negatively regulate the pathogenic processes of hepatosteatosis, immune cell activation (inflammation), and HSC activation (fibrosis), and the red font color denotes NRs that positively regulate them. Abbreviations: THRs, thyroid hormone receptors; GRs, glucocorticoid receptors; ERs, estrogen receptors; VDRs, vitamin D receptors; RARs, retinoic acid receptors; PPARα, peroxisome proliferator-activated receptor α; FXR, farnesoid X receptor; LXR, liver X receptor; PXR, pregnane X receptor; RORs, RAR-related orphan receptors; ERR, estrogen-related receptor; RXR, retinoid X receptor.

3. Nuclear hormone receptors and MASH

Hormone-responsive NRs regulate different aspects of hepatic lipid and glucose metabolism. Studies performed in both animals and humans have shown that alterations in hormones and their cognate NRs are associated with both the incidence and progression of MASLD and MASH. Furthermore, hormones and hormone mimetics have shown efficacy in resolving MASH severity in both preclinical and clinical studies as described below.

3.1. THRs

THRs are nuclear resident transcription factors that bind to thyroid hormone (TH) and triiodothyronine (T3), regulating the expression of several genes involved in lipogenesis, fat-oxidation, cholesterol transport, and gluconeogenesis in the liver.12 In humans and rodents, two THRs (THRalpha (THRα) and THRbeta (THRβ)) are expressed, with THRβ being the predominant form in the liver. Once inside the nuclei, T3 may activate or repress the expression of its target genes through the action of its receptor bound to the DNA.13 T3-induced genes harbor a cognate DNA sequence in their promoter/enhancer regions known as positive thyroid response elements (TRE), on which T3 bound THR complex is recruited either as a homodimer or as a heterodimer with RXR. The presence of T3 leads to the assembly of a THR-coactivator complex with an intrinsic histone acetylase activity resulting in nucleosome uncoiling and RNA POL II-mediated transcription.13 In the liver, THRs are expressed in hepatocytes, cholangiocytes, kupffer cells, and HSCs, and therefore, TH can influence almost all aspects of hepatic physiology.14, 15, 16, 17 Epidemiological studies have shown an increased incidence of MASLD with both overt and subclinical hypothyroidism in humans.18, 19, 20, 21, 22, 23 Furthermore, animal models of MASH have shown evidence of intra-hepatic TH insufficiency, suggesting deregulated TH metabolism in fatty liver.24 In rodent models of diet-induced MASH, both T3 and the liver-specific THRβ agonist significantly reduced hepatic steatosis and inflammation.25, 26, 27 At a molecular level, T3 decreases MASLD/MASH by increasing mitochondrial β-oxidation and promoting autophagy in the hepatocytes.12,28, 29, 30, 31, 32 Further evidence supporting the role of THRs in MASLD comes from a mouse model that expresses a dominant negative mutation in THRβ (THRβPV/PV). These mutant mice develop hepatosteatosis by 4–5 months of age.33 Similarly, humans with a similar mutation in THRβ, characterized as resistance to thyroid hormone (RTH) phenotype, also exhibit increased hepatic fat content.34 In human studies, low-dose TH as well as two liver-specific THRβ specific agonists VK-2809/MB-07811 and resmetirom (MGL-3196) have shown significant efficacy in resolving MASLD/MASH.35,36 The pivotal phase 3 MAESTRO-NASH clinical trial with Madrigal Pharmaceuticals’ oral MASH therapy, resmetirom, robustly demonstrated a significant reduction in hepatic fat and inflammation in patients with MASH. This treatment has received the United States Food and Drug Administration (FDA) breakthrough therapy designation.37, 38, 39, 40, 41

3.2. GRs

Hepatic genes related to glucose homeostasis, stress response, and inflammation, are regulated by GRs. GRs can be activated by both endogenous steroids (e.g., cortisol) and pharmaceutical ligands such as dexamethasone. At a molecular level, ligand binding to the cytoplasmic GRs elicits a conformational change in GRs and their interaction with the chaperone complex, which aids in GRs translocation into the nucleus.42 Upon entering the nucleus, GRs bind to DNA sequences known as glucocorticoid responsive elements (GRE) to activate or repress gene transcription. GRs can transactivate genes by binding to GRE as a dimer, but also as a monomer by binding to other transcription factors (TFs) through tethering or by binding to composite elements.42 GRs may also heterodimerize with other NRs such as cyclic adenosine monophosphate (cAMP) response element binding protein (CREB) and peroxisome proliferator-activated receptor α (PPARα) during fasting to increase the expression of gluconeogenic and ketogenic genes in the liver.43 Mouse models with liver-specific loss of GRs in obesity-prone animals (db/db mice) have shown amelioration of hepatic steatosis via derepression of the direct GR target gene, hairy enhancer of split 1 (Hes1).44 In contrast, the loss of GRs in liver macrophages aggravated liver inflammation in animal models of obesity via repression of the anti-inflammatory glucocorticoid-induced leucine zipper (GILZ) expression in monocytes/macrophages.45 However, steroids are known to induce fatty liver in humans, which has so far dampened any interest in targeting GRs for MASH.46 Nevertheless, given the potential of glucocorticoids in ameliorating inflammation in MASH, clinical studies targeting GRs as a single adjuvant therapy in advance of MASH, are warranted.

3.3. ERs

Estrogens are a group of hormones essential for the development and function of the female reproductive system. The classic estrogenic hormone, estradiol (E2) action is mediated by the ERs, which are expressed as ERα and ERβ isoforms.47 ERα is the major isoform expressed in the liver and regulates the expression of several lipogenic genes.48 Upon binding of the ligand, ERs dissociate from cytoplasmic heat shock protein 90 (HSP90) and translocate to the nucleus, where they bind to estrogen response elements (EREs) on the promoter region of the target genes.47 In the liver, several other NRs, including the SHP and signal transducer and activator of transcription 3 (STAT3), are direct targets of ERα. Beneficial effects of E2 on the liver, including the repression of lipid biosynthesis and gluconeogenesis, may be indirectly mediated by these secondary NRs.49,50 The loss of ERα is known to induce hepatosteatosis in both male and female mice; however, extrahepatic ER signaling also contributes to the overall beneficial effects of E2 on reducing hepatosteatosis.51, 52, 53, 54, 55 Furthermore, ERα is essential for the protective effect of E2 by inducing M1 to M2 macrophage polarization, thereby reducing MASH-associated inflammation in mice.56 In humans, premenopausal females are less likely to develop MASLD compared to males; however, the risk of developing MASLD/MASH significantly increases in postmenopausal females.57, 58, 59, 60 Estrogen replacement therapy has also been found to mitigate the development of MASLD in diabetic patients.61 These studies suggest a protective role of estrogen and ERα against MASLD development in humans.62 Additionally, ERα levels are shown to be reduced in the livers of MASH patients.63 While ERα is more widely studied and expressed in various tissues including the liver, ERβ agonists have also shown protective effects in preclinical models of MASH.64 Although clinical trials aimed at targeting ERs for MASH are currently lacking, preclinical studies are suggestive of perhaps testing this possibility in the future.

3.4. VDRs

While VDRs mediate the biological effects of vitamin D on the human skeletal system,65 the extra-skeletal effects of vitamin D, particularly on MASLD progression, have been increasingly recognized.66 The biologically active form of vitamin D is 1, 25-dihydroxyvitamin D3 (1,25(OH)2D3), which upon binding to VDR converts DNA-bound VDR homodimers into VDR-RXR heterodimers, which recruit corepressors or coactivators to regulate gene transcription.65 In rodent models, animals treated with 1,25(OH)2D3, displayed significant protection from diet-induced liver steatosis, inflammation, and fibrosis.67, 68, 69 However, animal studies investigating the impact of vitamin D and VDRs on the progression of MASLD and MASH have yielded conflicting results. While some studies reported that VDR-deficient mice are resistant to high-fat diet (HFD)-induced or ob/ob model-induced liver steatosis and VDR-dependent steatosis, other long-term studies reported that the absence of VDRs can actually exacerbate hepatic inflammation and fibrosis.70, 71, 72, 73 While some studies have suggested the potential benefits of vitamin D in mitigating hepatic steatosis, the evidence is not conclusive. Conflicting data also stem from the diverse and opposing effects of liver, adipose, and intestinal VDRs in the regulation of hepatic steatosis.73, 74, 75, 76 Although expressed at a low level in hepatocytes, VDRs are enriched in non-parenchymal liver cells such as cholangiocytes, kupffer cells, and HSCs.77 The VDR agonist 1,25(OH)2D3 can ameliorate transforming growth factor-β1(TGF-β1)-induced stellate cell activation in vitro, and HFD-induced liver steatosis and inflammation in vivo.73 Similarly, the activation of liver macrophage VDRs by vitamin D ameliorates liver inflammation, steatosis, and insulin resistance in mice.78 In humans, VDR polymorphisms and circulating vitamin D levels have been associated with the severity of MASLD.79, 80, 81, 82 Human studies suggest that although hepatic VDR expression is upregulated in benign steatosis, it is only modestly increased in individuals with MASH, indicating a temporal effect of VDR function in MASLD progression, in a tissue/cell-type specific manner.70 However, as the role of VDRs vis à vis that of vitamin D in MASH is still debatable and unclear, there are currently no clinical trials targeting VDR for the treatment of MASLD/MASH.

3.5. RARs

The RARs (RARα, RARβ, and RAR gamma (RARγ)) are ligand-activated transcription factors that are activated by both all-trans retinoic acid and 9-cis retinoic acid, retinoid active derivatives of vitamin A.83 Like other NRs, RARs also heterodimerize with the RXRs and bind to their cognate sequences on the target gene promoter regions, thereby regulating transcription.83 Endogenous retinoids regulate hepatic lipid and bile metabolism by binding to RARs.84,85 Animal studies have demonstrated a protective effect of both all-trans retinoic acid administration and RARα overexpression in reducing hepatic steatosis.86 Similarly, the overexpression of a dominant negative RARα, specifically in the liver, exhibits steatohepatitis and insulin resistance in mice.87 Furthermore, synthetic agonists for RARβ2 have shown efficacy in reducing hepatic lipid accumulation, activating HSCs, and alleviating insulin resistance in both genetic and diet-induced models of diabetes-associated MASLD.88, 89, 90 Although perturbed retinoic acid metabolism is observed in MASLD and MASH patients, clinical trials utilizing RAR-targeted ligands for MASLD/MASH treatment are still lacking.91,92 Intriguingly, in addition to retinoids, a novel synthetic RXR ligand, UAB126 (rexinoids), has shown positive results in preventing obesity-induced metabolic diseases, including MASLD, in animals fed obesogenic diets. Importantly, these effects were achieved without any adverse side effects including hypertriglyceridemia, hepatomegaly, and disturbances in the thyroid hormone axis.93 Thus, these results raise hopes of translating some promising leads obtained from retinoids and rexinoids into human clinical trials for MASLD.

4. Non-steroid hormone receptors

Non-steroid hormone NRs are activated by lipids, cholesterol, carbohydrates, and bile acids, and serve as a nutrient sensor within the cell. Many of these NRs, which belong to the class of non-steroid hormone receptors, are currently being considered, or already in clinical trials, to explore their efficacy in treating MASLD/MASH.94, 95, 96

4.1. PPARs

PPARs are lipid-regulated NRs that bind to PPAR response elements (PPREs) as heterodimers with RXRs, regulating the expression of genes involved in hepatic lipid and carbohydrate metabolism, inflammation, and cellular proliferation.97 There are three PPAR isotypes: PPARα (NR1C1), PPARβ/delta(δ) (NR1C2), and PPARγ (NR1C3). Several ligands of PPARs, such as elafibranor, are currently under clinical development for the treatment of MASH.98

4.1.1. PPARα (NR1C1)

PPARα is a fasting-induced NR that directly regulates the genes involved in fatty acid uptake, lipophagy, β-oxidation, and ketogenesis within hepatocytes.99, 100, 101 Additionally, some of the effects of PPARα are indirectly mediated via its induction of fibroblast growth factor 21 (FGF21), which increases lipid catabolism.102 PPARα fasting knock-out mice show impaired fatty acid oxidation and hepatic fat accumulation.103 Paradoxically, PPARα also increases the expression of lipogenic genes in the liver.104 This conflicting effect of PPARα may be attributed to its differential preference for promoting lipolytic gene expression during fasting and lipogenesis in the fed state. Studies in PPAR null mice fed an HFD exhibit massive hepatic lipid accumulation owing to the inhibition of fatty acid uptake and β-oxidation.105

Moreover, PPARα shows an anti-inflammatory activity in a murine model of systemic inflammation.106 In animal models of MASH, the PPARα agonist WY-14643 prevents hepatic steatosis and inflammation by reducing the number of activated macrophages and HSCs, ultimately facilitating the normalization of the histologic changes typical of MASH.107 Furthermore, PPARα inhibits the fibrotic and inflammatory gene expression induced by MASH diets through its physical interaction with nuclear factor-kappa B and activator protein-1 transcription factors.108,109 In humans, PPARα expression negatively correlates with MASH severity.110 A clinical trial using pemafibrate, a selective PPARα modulator, demonstrated improvement in magnetic resonance elastography-based liver stiffness and alanine transaminase (ALT) levels among patients with MASLD, but did not show a significant reduction in hepatic steatosis.111 Similarly, a dual PPARα/δ agonist, elafibranor, also showed promise as an anti-MASH therapy in earlier studies but failed in the later phase III trials due to safety concerns.112 Notably, pegozafermin which is an FGF-21 analogue has exhibited improvements in MASH-associated fibrosis during a recent phase II clinical trial, suggesting the potential of downstream PPARα signaling as a viable therapeutic area in MASH treatment.113

4.1.2. PPAR β/δ (NR1C2)

The activation of PPARβ/δ in hepatocytes increases the expression of the enzyme stearoyl-coenzyme A desaturase 1 (SCD1) that converts lipotoxic saturated fatty acids into mono-unsaturated fatty acids (MUFA), which can be easily stored as lipid droplets.114 Indeed, animals with liver-specific adenovirus-mediated PPARβ/δ overexpression exhibit lesser hepatic damage, despite increased lipid accumulation when fed a diet rich in saturated fat.115 A mechanism through which PPARβ/δ activation shows protection against MASLD is via the regulation of hepatic very low-density lipoprotein receptor (VLDLR).116 Consistent with animal studies, the expression of VLDLR correlates negatively with the abundance of PPARβ/δ in steatotic liver biopsy specimens.116 112

4.1.3. PPAR γ (NR1C3)

While expressed at a very low level in the liver, PPARγ is induced in the livers of animals upon feeding an HFD.117 Concurrently, hepatic ablation of PPARγ prevents the development of hepatic steatosis by reducing both hepatic fatty acid uptake and DNL.118,119 Paradoxically, the administration of PPARγ ligand rosiglitazone ameliorates the MASH phenotype in animal models.120 This paradoxical effect of PPARγ is likely mediated by its differential effect on HSCs vs. hepatocytes. Indeed, the activated pro-fibrotic phenotype of HSCs may be reversed to the quiescent ones upon binding PPARγ ligands, thus highlighting PPARγ ability to modulate pro-inflammatory and pro-fibrogenic gene expression in HSCs.121, 122, 123, 124 Additionally, PPARγ acts in liver macrophages to induce M2-type polarization, which is associated with decreased secretion of inflammatory cytokines and growth factors, thereby resulting in attenuated fibrosis.125, 126, 127 Indeed, macrophage-specific PPARγ deletion predisposes animals to develop diet-induced obesity and insulin resistance and worsen carbon tetrachloride-induced liver fibrosis.128 Similarly to rodents, PPARγ expression is also increased in the human steatotic liver.129 PPARγ agonists, including rosiglitazone and pioglitazone, have been evaluated in several clinical trials, showing efficacy in alleviating steatosis and inflammation, but with a modest reduction of fibrosis.130, 131, 132 However, a PPARα/γ dual agonist, saroglitazar, has exhibited significant efficacy in resolving MASH and is currently an approved drug for MASH treatment in India.133

Recently, a phase III clinical trial led by Inventiva Pharma is underway to evaluate the efficacy of a pan-PPAR agonist lanifibranor in MASH (ClinicalTrials.gov, NCT04849728). Results from the prior phase IIb trial showed a significant improvement in MASH resolution with lanifibranor.134

4.2. FXR

FXR is a bile acid receptor that regulates the metabolism of bile, lipids, and carbohydrates.135 FXR is highly expressed in the liver, brain, intestine, and kidney, and it exerts profound metabolic effects.136 FXR heterodimerizes with RXR and binds to inverted repeats with 1 nucleotide separating (IR1).135 The natural ligands of FXR, including chenodeoxycholic acid (CDCA) and cholic acid (CA), facilitate the recruitment of coactivators and the upregulation of transcription.135 The systemic activation of FXR by a synthetic agonist (GW4064) and obeticholic acid (OCA) improved glucose tolerance and ameliorated steatosis severity in mice fed with an HFD and high carbohydrate diet.137,138 In another mouse model of MASH, administration with FXR ligand WAY-362450 for 4 weeks, led to decreased levels of liver enzymes, reduced inflammatory cell infiltration, and alleviated hepatic steatosis, all of which were dependent on FXR expression.139 More recently, a study has demonstrated that the FXR agonist GSK2324 regulates hepatic lipids by reducing absorption and selectively decreasing fatty acid synthesis via its distinct effect on intestinal and hepatic FXRs.140 In the liver, FXR negatively regulates lipogenesis and positively enhances β-oxidation via its induction of SHP and PPARα.141,142 Furthermore, FXR sulfhydration affected by hepatic endogenous H2S promotes FXR activity and attenuates MASLD.143 Notably, although FXR expression is low in HSCs, its anti-fibrotic action is presumed to be executed by FGF-19/15, which is responsive to intestinal FXR activation.144, 145, 146

In a multicenter, randomized, phase III study, the FXR ligand OCA, demonstrated improved liver histology, including improvements in steatosis, inflammation, and fibrosis (measured as NAFLD activity score), in non-cirrhotic, non-alcoholic steatohepatitis patients within the farnesoid X receptor ligand obeticholic acid in NASH treatment (FLINT) trial; however, further clarification is needed regarding its long-term benefits and safety.147 Similarly, in a phase III, randomized, double-blind, placebo-controlled trial known as REGENERATE (ClinicalTrials.gov, NCT02548351), notable improvements in significantly improved fibrosis and key components of MASH disease activity were observed among patients with MASH.148 However, two recent trials, REVERSE (ClinicalTrials.gov, NCT03439254) and OCALIVA evaluating the efficacy of OCA failed to reach expected MASH endpoints in terms of statistical significance for histological improvement. Several nonsteroidal FXR agonists (tropifexor, nidufexor, and turofexorate) are also in the trial pipeline for MASH treatment. Recently, a phase II trial on GS-9674 (cilofexor), an FXR agonist showed improvement in hepatic steatosis, liver biochemistry, and serum bile acids, in patients with MASH (ClinicalTrials.gov, NCT02854605). Similarly, the reduction in ALT levels and hepatic fat induced by tropifexor was sustained up to week 48 (ClinicalTrials.gov, NCT02855164); however, dose-related pruritus was frequently observed.149 In spite of mixed success, FXR agonists remain an attractive choice for future MASH therapy.

4.3. LXRs

LXRs are cholesterol sensors that play a crucial role in regulating fatty acids, sphingolipids, cholesterol, and glucose metabolism, as well as inflammation.150, 151, 152 LXRs exist as two isotypes: LXRα is expressed mainly in the liver, adipose tissue, kidney, and macrophages, whereas LXRβ is expressed ubiquitously.151,152 Natural ligands of LXRs include oxysterols (cholesterol derivatives) such as 24(S),25-epoxycholesterol, 25-hydroxycholesterol, and 22(R)-hydroxycholesterol. Upon ligand binding, nuclear LXRs heterodimerize with RXRs to regulate the expression of hepatic genes involved in DNL (FASN, SREBP1c, SCD1) and cholesterol excretion (CYP7A1, ATP-binding cassette transporters A1(ABCA1) and ABCG1).151,152 Although the activation of LXRs in mouse hepatocytes increases hepatic steatosis when challenged with saturated fats, this process serves as a cytoprotective mechanism by converting saturated fats into MUFAs through the action of LXR target gene SCD1.153 Following LXR agonist treatment, LXRs exert anti-inflammatory functions via suppression of pro-inflammatory genes such as cyclooxygenase-2 (COX-2) and inducible NO synthase (iNOS).154 Additionally, LXR activation inhibits Toll-like receptor (TLR) ligand dependent inflammatory pathways through ABCA1 induction in macrophages.155 In mice deficient in LXRα, feeding a high-cholesterol diet promoted cholesterol accumulation and increased serum levels of ALT and aspartate aminotransferase (AST), as well as enhanced macrophage recruitment and Kupffer cell activation.156 Furthermore, LXRα/β double knockout mice show hepatic fibrosis, evidenced by the accumulation of hepatic lipid droplets and the induction of pro-fibrotic genes such as Acta2 and Col1a1.157 As LXR levels in humans positively correlate with MASLD severity and exhibit cell type specific pleiotropic effects on steatosis and inflammation.158 However, no clinical trials with either LXR antagonist or agonist are currently ongoing. Future research is needed to better understand the complex mechanisms of LXR signaling in MASLD and to optimize therapeutic strategies targeting LXRs.

4.4. PXR

The xenobiotic sensor PXR acts as a receptor for endobiotics, including bile acids, cholesterol, and steroid derivatives in the liver.159 Animal studies have shown that PXR activation in diet-induced MASH mouse models promotes a “fatty phenotype”.160,161 Consistently, a reduction in HFD-induced obesity was observed in PXR knockout mice and was correlated with an upregulation of FGF15 expression, which suppresses the synthesis of bile acids and reduces lipid absorption and triglycerides (TGs) in the liver.162 However, PXR levels have been shown to be down-regulated in human MASH.163 Therefore, given the discordance in the data regarding the role of PXR in human vs. mouse MASLD, further studies are still required to elucidate its utility in targeting MASH.164

5. Circadian NRs

5.1. REV-ERBs

The REV-ERB proteins, which exist in REV-ERBα and REV-ERBβ isoforms, mediate the negative feedback loop of the circadian clock in mammals.165 These NRs bind heme as their natural ligand and act as a transcriptional repressor of several metabolic genes in the liver.165 The combined loss of both REV-ERB isoforms results in hepatosteatosis in mice.166 Similarly, treatment with REV-ERB pan-agonists modifies both CLOCK and metabolic gene expression, reduces hepatic TG storage, and suppresses hepatic cholesterol synthesis in a diet-induced mouse MASLD model.167 Another study using a high-calorie diet fed genetically obese mice showed that REV-ERB pan-agonists inhibited fibrosis, suggesting a direct role of REV-ERBs in regulating MASLD/MASH pathogenesis.168,169 REV-ERBs also regulate different facets of liver inflammation, including inflammasome activation and T-helper 17 (Th17) cell activation, which are key players in MASH progression.168 Although a putative REV-ERB agonist SR9009 is available as a dietary supplement, there are no ongoing clinical trials targeting REV-ERB for MASLD/MASH therapy. Despite the potential benefits of targeting REV-ERBs in MASH, further research is needed to fully elucidate their mechanism of action and evaluate their efficacy and safety in clinical settings.

5.2. RORs

In contrast to REV-ERBs, RORs mostly act as transcriptional activators, and coordinate the circadian rhythms of lipid metabolism and inflammation in the liver.170 RORα and RORγ are the predominant RORs expressed in the liver and can be activated by cholesterol derivatives.171 The RORs play a critical role in orchestrating the circadian synchronization of hepatic lipid metabolism. Notably, a significant elevation in hepatic TG levels was observed in hepatic-specific RORα knockout mice fed an HFD.172, 173, 174, 175 The action of RORs on immune cells also regulates liver inflammation in MASH.176 However, given the puzzling results obtained from synthetic agonists and inverse agonists of RORs in mouse models of MASH, more work needs to be done before considering any clinical trial.176, 177, 178

6. Orphan NRs

By definition, an orphan NR is a protein that shares a structural similarity with identified receptors, but whose endogenous ligand remains unknown. Nevertheless, their genetic silencing and modulation by synthetic ligands demonstrate their vital role in regulating metabolic functions.

6.1. ERRs

ERRs are key regulators of energy metabolism and mediate mitochondrial oxidative metabolism.179 ERRα, the predominant isoform expressed in the liver induces or represses target gene expression by interaction with coregulators such as peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α).179 Metabolic crosstalk with other NRs such as PPARs, and THRs affect different aspects of ERRα metabolic signaling.180 Studies employing whole-body ERRα knockout models have shown resistance to HFD-induced weight gain.181 However, in contrast, the loss of ERRα aggravated the mammalian target of rapamycin inhibition induced fatty liver in mice.182 Similarly, the lack of ERRα exhibited differential effects on lipid-induced and fasting-induced hepatic steatosis in mice.183 Additionally, hepatic VLDL-TG secretion is blunted in ERRα liver-specific knockout mice, leading to hepatosteatosis, cellular stress, inflammation, and MASH development. Importantly, ERRα acts downstream of estrogen/ERα signaling, contributing to sex-based differences in MASLD/MASH.184 Recently, the utilization of small molecule inhibitors of ERRα has shown promise in diminishing hepatic lipid deposition and MASH development in both dietary and genetic models of MASLD.185 Collectively, there seems to be discordance in the results obtained from various studies, which may be attributed to the tissue-specific effect of ERRα. Further resolution of ERRα inhibition vs. activation using liver specificity of ERRα targeting modulators may pave the way for human trials in MASH.

6.2. CAR

Originally identified as an NR that regulates drug metabolism and detoxification, CAR has recently been recognized to be associated with energy metabolism and is found to be abundantly expressed in the liver.186 Studies conducted in mice have shown that CAR activation directly leads to liver steatosis, and its genetic ablation protects from diet-induced as well as toxicant-induced MASH.187, 188, 189, 190 However, in contrast, other studies have demonstrated that the loss of CAR actually worsens MASLD and MASH-induced fibrosis in mice.191,192 Additionally, CAR levels were found to be negatively correlated with MASH severity in humans.193 Thus, the relationship between CAR and MASLD/MASH should be further assessed, and there are currently no therapies targeting CAR in clinical trials.

6.3. SHP

Predominantly expressed in the liver, SHP is an atypical NR that lacks a DNA-binding domain.194 SHP acts as a repressor of NR action by competing for the coactivators required for NR action.195 In the liver, the role of SHP remains controversial, with studies showing totally contrasting effects of SHP modulation on the development of steatosis and inflammation in animals with pro-steatotic vs. anti-inflammatory effects.196, 197, 198, 199, 200, 201, 202 Furthermore, a notable reduction in SHP levels has been observed in patients at the advanced stages compared to those in mild MASLD, suggesting a stage-specific function of SHP during MASLD development.203 Given the unclear function of SHP in MASLD development, no clinical trials are currently underway.

6.4. HNF4α

HNF4α, unlike SHP, acts as a transcriptional activator that drives the transcription of a broad spectrum of genes related to essential liver functions involving lipid, glucose, drug, and bile acid metabolism, as well as inflammatory response. HNF4α activation has also been linked to improvements in MASH pathology, while the loss of HNF4α activity leads to hepatic steatosis and MASH progression.204, 205, 206, 207, 208, 209 Additionally, a chemical antagonist of HNF4α was shown to induce hepatic steatosis in mice.210 Given the beneficial role of HNF4α in MASH prevention and its down-regulation observed in human MASH, clinical trials involving HNF4α activators may be an attractive possibility for MASH treatment.211

6.5. LRH-1/NR5A2

LRH-1/NR5A2, originally identified in the liver as a regulator of bile acid and cholesterol homeostasis, regulates a multitude of other hepatic metabolic processes, encompassing glucose and lipid processing, methyl group sensing, and cellular stress responses.212 Studies performed in liver-specific LRH-1 null mice unveiled derangement in phospholipid composition, hepatic steatosis, and the development of MASH phenotype.213 Furthermore, LRH-1 levels were found to decrease as MASH progressed in humans. Given the encouraging results from preclinical studies, the development of a specific human LRH-1 agonist may be the way forward to test the efficacy of LRH-1 in reducing MASH-related complications in humans.

7. Conclusions and future perspectives

NRs-based pharmacological drugs are currently the most promising candidates to obtain FDA approval for MASH treatment. Owing to their wide coverage of the genomic landscape, NRs regulate several hepatic processes including lipid synthesis, lipolysis, mitochondrial function, as well as inflammatory and fibrotic signaling, all of which are deregulated at different stages of MASLD progression (Table 1). Understanding the contrasting effects of some NRs on hepatic physiology and pathophysiology is crucial, as these effects may be due to the differential expression of NRs within liver cell types together with their interaction with other transcription factors. Furthermore, a limitation of present NR therapeutics is the unwanted and often adverse extrahepatic activation of target NRs. Therefore, the liver-specificity of NR agonism/antagonism is vital to maximize the benefits of NR activation through synthetic ligands and will be a way ahead for more precise and tactical MASH therapies. Further translational evidence should accelerate the entry of more NRs in the trial pipeline, alongside genetic screening for pathogenic NR mutations that may predict MASH development and progression. Additionally, future trials should take into account the differential NR targeting for lean NASH vs. diabetes associated MASH. NR crosstalk is another important area that warrants increased attention while designing future NR-based therapeutics for MASLD/MASH. Novel ligands that precisely enhance or reduce NR factor crosstalk are expected to minimize side effects and resistance observed with endogenous NR ligands. Furthermore, unorthodox targeting concepts, such as dual NR ligands, allosteric ligands, and ligands targeting different NR domains or NR-related protein-protein interactions may add refinement to existing NR-based pharmacology for treating MASLD/MASH.

Table 1.

Effects of NRs targeting on liver pathology in MASLD/MASH.

NRs Agonist or antagonist Research methods Key findings NR activation/inhibition References
THRs Agonist (GC-1) Mouse/rat Anti-steatosis effects Activation 25,26
Thyroxine Mouse Anti-steatosis action and anti-inflammatory effects Activation 27
THRβ Liver-specific agonist (Resmetirom) Human Anti-steatosis, anti-inflammatory, and anti-fibrotic effects Activation (clinical trial: NCT03900429; active) 37, 38, 39, 40
GR Genetic silencing Mouse Anti-steatosis effects Inhibition (GR KO in hepatocytes) 43
Genetic silencing Mouse Pro-inflammatory effects Inhibition (GR KO in macrophage) 44
ER Genetic silencing Mouse Pro-steatosis effects Inhibition (ER KO) 50,54
ERα and ERβ agonists Mouse Anti-inflammatory effects Activation 55,63
VDR Agonist (vitamin D3) Mouse Anti-steatosis, anti-inflammatory, and anti-fibrotic effects Activation 67
RARα Agonist Mouse Anti-steatosis effects Activation 85
RARβ Agonist Mouse Anti-steatosis and anti-fibrotic effects Activation 87, 88, 89
RXR Agonist Mouse Anti-steatosis effects Activation 93
PPARα Agonist Mouse Anti-steatosis and anti-inflammatory effects Activation 107
Pemafibrate Human Improvement in liver stiffness and ALT levels Activation (clinical trial: NCT03350165; completed) 110
PPAR β/δ Agonist Mouse Reduction in liver injury Activation 115
PPAR γ Agonist Mouse Anti-inflammatory and anti-fibrotic effects Activation in macrophage and HSCs 121, 122, 123, 124, 125, 126, 127
PPARα/γ Agonist (Saroglitazar) Human Anti-steatosis, anti-inflammatory, and anti-fibrotic effects Activation (clinical trial: EVIDENCES II; completed) 133
FXR Agonist Mouse Anti-steatosis, anti-inflammatory, and anti-fibrotic effects Activation 136, 137, 138
REGENERATE
Obeticholic acid
GS-9674
Human Anti-steatosis, anti-inflammatory, and anti-fibrotic effects Activation (clinical trial: NCT02548351; completed), (clinical trial: NCT01265498; completed), (clinical trial: NCT02854605; completed) 146, 147, 148
LXR Agonist Mouse Pro-steatosis effects Activation (hepatocytes) 152
Agonist Mouse Anti-inflammatory effects Activation (macrophage) 154
PXR Agonist Mouse Pro-steatosis effects Activation 159,160
REV-ERB Agonist Mouse Anti-steatosis, anti-inflammatory, and anti-fibrotic effects Activation 166, 167, 168
RORα Genetic silencing Mouse Pro-steatosis effects Inhibition (liver-specific ROR KO) 171, 172, 173, 174
Overexpression Mouse Anti-inflammatory effects Activation 175
ERRα Antagonist Mouse Anti-steatosis effects Inhibition 185
CAR Agonist Mouse Pro-steatosis effects Activation 186, 187, 188
SHP Overexpression Mouse Anti-inflammatory effects Activation 195
HNF4α Antagonist Mouse Pro-steatotic effects Inhibition 210
LRH-1 Genetic silencing Mouse Pro-steatosis and pro-inflammatory effects Inhibition (LRH-1 KO) 213

Abbreviations: NRs, nuclear receptors; THR, thyroid hormone receptor; GR, glucocorticoid receptor; ER, estrogen receptor; VDR, vitamin D receptor; RAR, retinoic acid receptors; RXR, retinoid X receptors; PPARα, peroxisome proliferator-activated receptorα; HSCs, hepatic stellate cells; FXR, farnesoid X receptor; ALT, alanine transaminase; LXR, liver X receptors; PXR, pregnane X receptor; ROR, RAR-related orphan receptor; ERR, estrogen-related receptor; CAR, constitutive androstane receptor; SHP, small heterodimer partner; HNF4α, hepatocyte nuclear factor 4α; LRH-1, liver receptor homolog-1.

Author's contribution

Rohit A. Sinha: Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Project administration, Resources, Software, Supervision, Validation, Visualization, Writing – original draft, Writing – review & editing.

Declaration of competing interest

The author declares that there is no conflicts of interest.

Acknowledgements

This study was supported by the SERB (CRG/2022/002149) and Wellcome Trust/DBT India Alliance Fellowship [IA/I/16/2/502691].

Footnotes

Edited by Peiling Zhu.

References

  • 1.Younossi Z., Tacke F., Arrese M., et al. Global perspectives on nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Hepatology. 2019;69:2672–2682. doi: 10.1002/hep.30251. [DOI] [PubMed] [Google Scholar]
  • 2.Wong V.W., Ekstedt M., Wong G.L., Hagström H. Changing epidemiology, global trends and implications for outcomes of NAFLD. J Hepatol. 2023;79:842–852. doi: 10.1016/j.jhep.2023.04.036. [DOI] [PubMed] [Google Scholar]
  • 3.Chandrakumaran A., Siddiqui M.S. Implications of nonalcoholic steatohepatitis as the cause of end-stage liver disease before and after liver transplant. Gastroenterol Clin N Am. 2020;49:165–178. doi: 10.1016/j.gtc.2019.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Adams L.A., Anstee Q.M., Tilg H., Targher G. Non-alcoholic fatty liver disease and its relationship with cardiovascular disease and other extrahepatic diseases. Gut. 2017;66:1138–1153. doi: 10.1136/gutjnl-2017-313884. [DOI] [PubMed] [Google Scholar]
  • 5.Burra P., Becchetti C., Germani G. NAFLD and liver transplantation: disease burden, current management and future challenges. JHEP Rep. 2020;2 doi: 10.1016/j.jhepr.2020.100192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Sheka A.C., Adeyi O., Thompson J., Hameed B., Crawford P.A., Ikramuddin S. Nonalcoholic steatohepatitis: a review. JAMA. 2020;323:1175–1183. doi: 10.1001/jama.2020.2298. [DOI] [PubMed] [Google Scholar]
  • 7.Wallace S.J., Tacke F., Schwabe R.F., Henderson N.C. Understanding the cellular interactome of non-alcoholic fatty liver disease. JHEP Rep. 2022;4 doi: 10.1016/j.jhepr.2022.100524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Tewari A., Rajak S., Raza S., et al. Targeting extracellular RNA mitigates hepatic lipotoxicity and liver injury in NASH. Cells. 2023;12:1845. doi: 10.3390/cells12141845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Xiao Y., Kim M., Lazar M.A. Nuclear receptors and transcriptional regulation in non-alcoholic fatty liver disease. Mol Metab. 2021;50 doi: 10.1016/j.molmet.2020.101119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Evans R.M., Mangelsdorf D.J. Nuclear receptors, RXR, and the big bang. Cell. 2014;157:255–266. doi: 10.1016/j.cell.2014.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Arnal J.F., Fontaine C., Adlanmerini M., Lenfant F. Special issue on non-genomic actions of nuclear receptors: an evolutionary and physiological perspective. Mol Cell Endocrinol. 2023;564 doi: 10.1016/j.mce.2023.111884. [DOI] [PubMed] [Google Scholar]
  • 12.Sinha R.A., Singh B.K., Yen P.M. Direct effects of thyroid hormones on hepatic lipid metabolism. Nat Rev Endocrinol. 2018;14:259–269. doi: 10.1038/nrendo.2018.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Sinha R., Yen P.M. In: Feingold K.R., Anawalt B., Blackman M.R., et al., editors. MDText.com, Inc; South Dartmouth (MA): 2018. Cellular Action of Thyroid Hormone. [Google Scholar]
  • 14.Puymirat J., Gadbois P., Dussault L., Garceau L., Dussault J.H. Production of a specific polyclonal antibody against the rat beta thyroid receptor, using synthetic peptide as antigen. Acta Endocrinol (Copenh) 1991;125:397–400. doi: 10.1530/acta.0.1250397. [DOI] [PubMed] [Google Scholar]
  • 15.Fava G., Ueno Y., Glaser S., et al. Thyroid hormone inhibits biliary growth in bile duct-ligated rats by PLC/IP(3)/Ca(2+)-dependent downregulation of SRC/ERK1/2. Am J Physiol Cell Physiol. 2007;292:C1467–C1475. doi: 10.1152/ajpcell.00575.2006. [DOI] [PubMed] [Google Scholar]
  • 16.Tapia G., Santibáñez C., Farías J., et al. Kupffer-cell activity is essential for thyroid hormone rat liver preconditioning. Mol Cell Endocrinol. 2010;323:292–297. doi: 10.1016/j.mce.2010.03.014. [DOI] [PubMed] [Google Scholar]
  • 17.Manka P., Coombes J., Bechmann L., et al. Thyroid hormone receptor regulates hepatic stellate cell activation. J Hepatol. 2017;66:S582. doi: 10.1016/S0168-8278(17)31587-8. [DOI] [Google Scholar]
  • 18.Sheikhi V., Heidari Z. Association of subclinical hypothyroidism with nonalcoholic fatty liver disease in patients with type 2 diabetes mellitus: a cross-sectional study. Adv Biomed Res. 2022;11:124. doi: 10.4103/abr.abr_15_21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Kim D., Vazquez-Montesino L.M., Escober J.A., et al. Low thyroid function in nonalcoholic fatty liver disease is an independent predictor of all-cause and cardiovascular mortality. Am J Gastroenterol. 2020;115:1496–1504. doi: 10.14309/ajg.0000000000000654. [DOI] [PubMed] [Google Scholar]
  • 20.Kim D., Kim W., Joo S.K., Bae J.M., Kim J.H., Ahmed A. Subclinical hypothyroidism and low-normal thyroid function are associated with nonalcoholic steatohepatitis and fibrosis. Clin Gastroenterol Hepatol. 2018;16:123–131.e1. doi: 10.1016/j.cgh.2017.08.014. [DOI] [PubMed] [Google Scholar]
  • 21.Pagadala M.R., Zein C.O., Dasarathy S., Yerian L.M., Lopez R., McCullough A.J. Prevalence of hypothyroidism in nonalcoholic fatty liver disease. Dig Dis Sci. 2012;57:528–534. doi: 10.1007/s10620-011-2006-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Gardner C.J., Richardson P., Wong C., Polavarapu N., Kemp G.J., Cuthbertson D.J. Hypothyroidism in a patient with non-alcoholic fatty liver disease. BMJ. 2011;342:c7199. doi: 10.1136/bmj.c7199. [DOI] [PubMed] [Google Scholar]
  • 23.Kalaitzakis E. Fatigue in non-alcoholic fatty liver disease: is there a role for hypothyroidism. Gut. 2009;58:149–150. [PubMed] [Google Scholar]
  • 24.Bohinc B.N., Michelotti G., Xie G., et al. Repair-related activation of hedgehog signaling in stromal cells promotes intrahepatic hypothyroidism. Endocrinology. 2014;155:4591–4601. doi: 10.1210/en.2014-1302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Cable E.E., Finn P.D., Stebbins J.W., et al. Reduction of hepatic steatosis in rats and mice after treatment with a liver-targeted thyroid hormone receptor agonist. Hepatology. 2009;49:407–417. doi: 10.1002/hep.22572. [DOI] [PubMed] [Google Scholar]
  • 26.Perra A., Simbula G., Simbula M., et al. Thyroid hormone (T3) and TRbeta agonist GC-1 inhibit/reverse nonalcoholic fatty liver in rats. FASEB J. 2008;22:2981–2989. doi: 10.1096/fj.08-108464. [DOI] [PubMed] [Google Scholar]
  • 27.Zhou J., Tripathi M., Ho J.P., et al. Thyroid hormone decreases hepatic steatosis, inflammation, and fibrosis in a dietary mouse model of nonalcoholic steatohepatitis. Thyroid. 2022;32:725–738. doi: 10.1089/thy.2021.0621. [DOI] [PubMed] [Google Scholar]
  • 28.Iannucci L.F., Cioffi F., Senese R., et al. Metabolomic analysis shows differential hepatic effects of T2 and T3 in rats after short-term feeding with high fat diet. Sci Rep. 2017;7:2023. doi: 10.1038/s41598-017-02205-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Zhou J., Sinha R.A., Yen P.M. The roles of autophagy and thyroid hormone in the pathogenesis and treatment of NAFLD. Hepatoma Res. 2021;7:72. doi: 10.20517/2394-5079.2021.82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Sinha R.A., Singh B.K., Yen P.M. Reciprocal crosstalk between autophagic and endocrine signaling in metabolic homeostasis. Endocr Rev. 2017;38:69–102. doi: 10.1210/er.2016-1103. [DOI] [PubMed] [Google Scholar]
  • 31.Sinha R.A., Yen P.M. Thyroid hormone-mediated autophagy and mitochondrial turnover in NAFLD. Cell Biosci. 2016;6:46. doi: 10.1186/s13578-016-0113-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Sinha R.A., You S.H., Zhou J., et al. Thyroid hormone stimulates hepatic lipid catabolism via activation of autophagy. J Clin Invest. 2012;122:2428–2438. doi: 10.1172/JCI60580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Araki O., Ying H., Zhu X.G., Willingham M.C., Cheng S.Y. Distinct dysregulation of lipid metabolism by unliganded thyroid hormone receptor isoforms. Mol Endocrinol. 2009;23:308–315. doi: 10.1210/me.2008-0311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Chaves C., Bruinstroop E., Refetoff S., Yen P.M., Anselmo J. Increased hepatic fat content in patients with resistance to thyroid hormone beta. Thyroid. 2021;31:1127–1134. doi: 10.1089/thy.2020.0651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Bruinstroop E., Dalan R., Cao Y., et al. Low-dose levothyroxine reduces intrahepatic lipid content in patients with type 2 diabetes mellitus and NAFLD. J Clin Endocrinol Metab. 2018;103:2698–2706. doi: 10.1210/jc.2018-00475. [DOI] [PubMed] [Google Scholar]
  • 36.Li L., Song Y., Shi Y., Sun L. Thyroid hormone receptor-β agonists in NAFLD therapy: possibilities and challenges. J Clin Endocrinol Metab. 2023;108:1602–1613. doi: 10.1210/clinem/dgad072. [DOI] [PubMed] [Google Scholar]
  • 37.Harrison S.A., Bashir M.R., Guy C.D., et al. Resmetirom (MGL-3196) for the treatment of non-alcoholic steatohepatitis: a multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet. 2019;394:2012–2024. doi: 10.1016/S0140-673632517-6. [DOI] [PubMed] [Google Scholar]
  • 38.Harrison S.A., Bashir M., Moussa S.E., et al. Effects of resmetirom on noninvasive endpoints in a 36-week phase 2 active treatment extension study in patients with NASH. Hepatol Commun. 2021;5:573–588. doi: 10.1002/hep4.1657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Younossi Z.M., Stepanova M., Taub R.A., Barbone J.M., Harrison S.A. Hepatic fat reduction due to resmetirom in patients with nonalcoholic steatohepatitis is Associated with improvement of quality of life. Clin Gastroenterol Hepatol. 2022;20:1354–1361.e7. doi: 10.1016/j.cgh.2021.07.039. [DOI] [PubMed] [Google Scholar]
  • 40.Javanbakht M., Fishman J., Moloney E., Rydqvist P., Ansaripour A. Early cost-effectiveness and price threshold analyses of resmetirom: an investigational treatment for management of nonalcoholic steatohepatitis. Pharmacoecon Open. 2023;7:93–110. doi: 10.1007/s41669-022-00370-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Harrison S.A., Bedossa P., Guy C.D., et al. A phase 3, randomized, controlled trial of resmetirom in NASH with liver fibrosis. N Engl J Med. 2024;390:497–509. doi: 10.1056/NEJMoa2309000. [DOI] [PubMed] [Google Scholar]
  • 42.Frank F., Ortlund E.A., Liu X. Structural insights into glucocorticoid receptor function. Biochem Soc Trans. 2021;49:2333–2343. doi: 10.1042/BST20210419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Goldstein I., Baek S., Presman D.M., Paakinaho V., Swinstead E.E., Hager G.L. Transcription factor assisted loading and enhancer dynamics dictate the hepatic fasting response. Genome Res. 2017;27:427–439. doi: 10.1101/gr.212175.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Lemke U., Krones-Herzig A., Berriel Diaz M., et al. The glucocorticoid receptor controls hepatic dyslipidemia through Hes1. Cell Metab. 2008;8:212–223. doi: 10.1016/j.cmet.2008.08.001. [DOI] [PubMed] [Google Scholar]
  • 45.Robert O., Boujedidi H., Bigorgne A., et al. Decreased expression of the glucocorticoid receptor-GILZ pathway in Kupffer cells promotes liver inflammation in obese mice. J Hepatol. 2016;64:916–924. doi: 10.1016/j.jhep.2015.11.023. [DOI] [PubMed] [Google Scholar]
  • 46.Rahimi L., Rajpal A., Ismail-Beigi F. Glucocorticoid-induced fatty liver disease. Diabetes Metab Syndr Obes. 2020;13:1133–1145. doi: 10.2147/DMSO.S247379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Osborne C.K., Schiff R. Estrogen-receptor biology: continuing progress and therapeutic implications. J Clin Oncol. 2005;23:1616–1622. doi: 10.1200/JCO.2005.10.036. [DOI] [PubMed] [Google Scholar]
  • 48.Palmisano B.T., Zhu L., Stafford J.M. Role of estrogens in the regulation of liver lipid metabolism. Adv Exp Med Biol. 2017;1043:227–256. doi: 10.1007/978-3-319-70178-3_12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Gao H., Bryzgalova G., Hedman E., et al. Long-term administration of estradiol decreases expression of hepatic lipogenic genes and improves insulin sensitivity in ob/ob mice: a possible mechanism is through direct regulation of signal transducer and activator of transcription 3. Mol Endocrinol. 2006;20:1287–1299. doi: 10.1210/me.2006-0012. [DOI] [PubMed] [Google Scholar]
  • 50.Gao H., Fält S., Sandelin A., Gustafsson J.A., Dahlman-Wright K. Genome-wide identification of estrogen receptor alpha-binding sites in mouse liver. Mol Endocrinol. 2008;22:10–22. doi: 10.1210/me.2007-0121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Chow J.D., Jones M.E., Prelle K., Simpson E.R., Boon W.C. A selective estrogen receptor α agonist ameliorates hepatic steatosis in the male aromatase knockout mouse. J Endocrinol. 2011;210:323–334. doi: 10.1530/JOE-10-0462. [DOI] [PubMed] [Google Scholar]
  • 52.Guillaume M., Riant E., Fabre A., et al. Selective liver estrogen receptor α modulation prevents steatosis, diabetes, and obesity through the anorectic growth differentiation factor 15 hepatokine in mice. Hepatol Commun. 2019;3:908–924. doi: 10.1002/hep4.1363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Hart-Unger S., Arao Y., Hamilton K.J., et al. Hormone signaling and fatty liver in females: analysis of estrogen receptor α mutant mice. Int J Obes. 2017;41:945–954. doi: 10.1038/ijo.2017.50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Winn N.C., Jurrissen T.J., Grunewald Z.I., et al. Estrogen receptor-α signaling maintains immunometabolic function in males and is obligatory for exercise-induced amelioration of nonalcoholic fatty liver. Am J Physiol Endocrinol Metab. 2019;316:E156–E167. doi: 10.1152/ajpendo.00259.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Villa A., Della Torre S., Stell A., Cook J., Brown M., Maggi A. Tetradian oscillation of estrogen receptor α is necessary to prevent liver lipid deposition. Proc Natl Acad Sci U S A. 2012;109:11806–11811. doi: 10.1073/pnas.1205797109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Shu Z., Zhang G., Zhu X., Xiong W. Estrogen receptor α mediated M1/M2 macrophages polarization plays a critical role in NASH of female mice. Biochem Biophys Res Commun. 2022;596:63–70. doi: 10.1016/j.bbrc.2022.01.085. [DOI] [PubMed] [Google Scholar]
  • 57.DiStefano J.K. NAFLD and NASH in Postmenopausal women: implications for diagnosis and treatment. Endocrinology. 2020;161:bqaa134. doi: 10.1210/endocr/bqaa134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ryu S., Suh B.S., Chang Y., et al. Menopausal stages and non-alcoholic fatty liver disease in middle-aged women. Eur J Obstet Gynecol Reprod Biol. 2015;190:65–70. doi: 10.1016/j.ejogrb.2015.04.017. [DOI] [PubMed] [Google Scholar]
  • 59.Völzke H., Schwarz S., Baumeister S.E., et al. Menopausal status and hepatic steatosis in a general female population. Gut. 2007;56:594–595. doi: 10.1136/gut.2006.115345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Lonardo A., Nascimbeni F., Ballestri S., et al. Sex differences in nonalcoholic fatty liver disease: state of the art and identification of research gaps. Hepatology. 2019;70:1457–1469. doi: 10.1002/hep.30626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.McKenzie J., Fisher B.M., Jaap A.J., Stanley A., Paterson K., Sattar N. Effects of HRT on liver enzyme levels in women with type 2 diabetes: a randomized placebo-controlled trial. Clin Endocrinol (Oxf) 2006;65:40–44. doi: 10.1111/j.1365-2265.2006.02543.x. [DOI] [PubMed] [Google Scholar]
  • 62.Meda C., Barone M., Mitro N., et al. Hepatic ERα accounts for sex differences in the ability to cope with an excess of dietary lipids. Mol Metab. 2020;32:97–108. doi: 10.1016/j.molmet.2019.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Erkan G., Yilmaz G., Konca Degertekin C., Akyol G., Ozenirler S. Presence and extent of estrogen receptor-alpha expression in patients with simple steatosis and NASH. Pathol Res Pract. 2013;209:429–432. doi: 10.1016/j.prp.2013.04.010. [DOI] [PubMed] [Google Scholar]
  • 64.Ponnusamy S., Tran Q.T., Thiyagarajan T., Miller D.D., Bridges D., Narayanan R. An estrogen receptor β-selective agonist inhibits non-alcoholic steatohepatitis in preclinical models by regulating bile acid and xenobiotic receptors. Exp Biol Med (Maywood) 2017;242:606–616. doi: 10.1177/1535370216688569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Rochel N. Vitamin D and its receptor from a structural perspective. Nutrients. 2022;14:2847. doi: 10.3390/nu14142847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Bouillon R., Marcocci C., Carmeliet G., et al. Skeletal and extraskeletal actions of vitamin D: current evidence and outstanding questions. Endocr Rev. 2019;40:1109–1151. doi: 10.1210/er.2018-00126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Yin Y., Yu Z., Xia M., Luo X., Lu X., Ling W. Vitamin D attenuates high fat diet-induced hepatic steatosis in rats by modulating lipid metabolism. Eur J Clin Invest. 2012;42:1189–1196. doi: 10.1111/j.1365-2362.2012.02706.x. [DOI] [PubMed] [Google Scholar]
  • 68.Nakano T., Cheng Y.F., Lai C.Y., et al. Impact of artificial sunlight therapy on the progress of non-alcoholic fatty liver disease in rats. J Hepatol. 2011;55:415–425. doi: 10.1016/j.jhep.2010.11.028. [DOI] [PubMed] [Google Scholar]
  • 69.Raza S., Tewari A., Rajak S., Sinha R.A. Vitamins and non-alcoholic fatty liver disease: a molecular insight. Liver Res. 2021;5:62–71. doi: 10.1016/j.livres.2021.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Bozic M., Guzmán C., Benet M., et al. Hepatocyte vitamin D receptor regulates lipid metabolism and mediates experimental diet-induced steatosis. J Hepatol. 2016;65:748–757. doi: 10.1016/j.jhep.2016.05.031. [DOI] [PubMed] [Google Scholar]
  • 71.García-Monzón C., Petrov P.D., Rey E., et al. Angiopoietin-like protein 8 is a novel vitamin D receptor target gene involved in nonalcoholic fatty liver pathogenesis. Am J Pathol. 2018;188:2800–2810. doi: 10.1016/j.ajpath.2018.07.028. [DOI] [PubMed] [Google Scholar]
  • 72.Barchetta I., Cimini F.A., Chiappetta C., et al. Relationship between hepatic and systemic angiopoietin-like 3, hepatic vitamin D receptor expression and NAFLD in obesity. Liver Int. 2020;40:2139–2147. doi: 10.1111/liv.14554. [DOI] [PubMed] [Google Scholar]
  • 73.Ding N., Yu R.T., Subramaniam N., et al. A vitamin D receptor/SMAD genomic circuit gates hepatic fibrotic response. Cell. 2013;153:601–613. doi: 10.1016/j.cell.2013.03.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Zhang H., Shen Z., Lin Y., et al. Vitamin D receptor targets hepatocyte nuclear factor 4α and mediates protective effects of vitamin D in nonalcoholic fatty liver disease. J Biol Chem. 2020;295:3891–3905. doi: 10.1074/jbc.RA119.011487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Tao T., Kobelski M.M., Saini V., Demay M.B. Adipose-specific VDR deletion leads to hepatic steatosis in female mice fed a low-fat diet. Endocrinology. 2022;163:bqab249. doi: 10.1210/endocr/bqab249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Jahn D., Dorbath D., Schilling A.K., et al. Intestinal vitamin D receptor modulates lipid metabolism, adipose tissue inflammation and liver steatosis in obese mice. Biochim Biophys Acta, Mol Basis Dis. 2019;1865:1567–1578. doi: 10.1016/j.bbadis.2019.03.007. [DOI] [PubMed] [Google Scholar]
  • 77.Gascon-Barré M., Demers C., Mirshahi A., Néron S., Zalzal S., Nanci A. The normal liver harbors the vitamin D nuclear receptor in nonparenchymal and biliary epithelial cells. Hepatology. 2003;37:1034–1042. doi: 10.1053/jhep.2003.50176. [DOI] [PubMed] [Google Scholar]
  • 78.Dong B., Zhou Y., Wang W., et al. Vitamin D receptor activation in liver macrophages ameliorates hepatic inflammation, steatosis, and insulin resistance in mice. Hepatology. 2020;71:1559–1574. doi: 10.1002/hep.30937. [DOI] [PubMed] [Google Scholar]
  • 79.Tourkochristou E., Mouzaki A., Triantos C. Gene polymorphisms and biological effects of vitamin D receptor on nonalcoholic fatty liver disease development and progression. Int J Mol Sci. 2023;24:8288. doi: 10.3390/ijms24098288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Jaroenlapnopparat A., Suppakitjanusant P., Ponvilawan B., Charoenngam N. Vitamin D-related genetic variations and nonalcoholic fatty liver disease: a systematic review. Int J Mol Sci. 2022;23:9122. doi: 10.3390/ijms23169122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Gibson P.S., Quaglia A., Dhawan A., et al. Vitamin D status and associated genetic polymorphisms in a cohort of UK children with non-alcoholic fatty liver disease. Pediatr Obes. 2018;13:433–441. doi: 10.1111/ijpo.12293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Arai T., Atsukawa M., Tsubota A., et al. Association of vitamin D levels and vitamin D-related gene polymorphisms with liver fibrosis in patients with biopsy-proven nonalcoholic fatty liver disease. Dig Liver Dis. 2019;51:1036–1042. doi: 10.1016/j.dld.2018.12.022. [DOI] [PubMed] [Google Scholar]
  • 83.Petkovich M., Chambon P. Retinoic acid receptors at 35 years. J Mol Endocrinol. 2022;69:T13–T24. doi: 10.1530/JME-22-0097. [DOI] [PubMed] [Google Scholar]
  • 84.He Y., Gong L., Fang Y., et al. The role of retinoic acid in hepatic lipid homeostasis defined by genomic binding and transcriptome profiling. BMC Genomics. 2013;14:575. doi: 10.1186/1471-2164-14-575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Yang F., He Y., Liu H.X., et al. All-trans retinoic acid regulates hepatic bile acid homeostasis. Biochem Pharmacol. 2014;91:483–489. doi: 10.1016/j.bcp.2014.08.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Kim S.C., Kim C.K., Axe D., et al. All-trans-retinoic acid ameliorates hepatic steatosis in mice by a novel transcriptional cascade. Hepatology. 2014;59:1750–1760. doi: 10.1002/hep.26699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Tsuchiya H., Ikeda Y., Ebata Y., et al. Retinoids ameliorate insulin resistance in a leptin-dependent manner in mice. Hepatology. 2012;56:1319–1330. doi: 10.1002/hep.25798. [DOI] [PubMed] [Google Scholar]
  • 88.Trasino S.E., Tang X.H., Jessurun J., Gudas L.J. Retinoic acid receptor β2 agonists restore glycaemic control in diabetes and reduce steatosis. Diabetes Obes Metab. 2016;18:142–151. doi: 10.1111/dom.12590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Tang X.H., Melis M., Lu C., et al. A retinoic acid receptor β2 agonist attenuates transcriptome and metabolome changes underlying nonalcohol-associated fatty liver disease. J Biol Chem. 2021;297 doi: 10.1016/j.jbc.2021.101331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Trasino S.E., Tang X.H., Jessurun J., Gudas L.J. A retinoic acid receptor β2 agonist reduces hepatic stellate cell activation in nonalcoholic fatty liver disease. J Mol Med (Berl) 2016;94:1143–1151. doi: 10.1007/s00109-016-1434-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Liu Y., Chen H., Wang J., Zhou W., Sun R., Xia M. Association of serum retinoic acid with hepatic steatosis and liver injury in nonalcoholic fatty liver disease. Am J Clin Nutr. 2015;102:130–137. doi: 10.3945/ajcn.114.105155. [DOI] [PubMed] [Google Scholar]
  • 92.Saeed A., Dullaart R.P.F., Schreuder T.C.M.A., Blokzijl H., Faber K.N. Disturbed vitamin a metabolism in non-alcoholic fatty liver disease (NAFLD) Nutrients. 2017;10:29. doi: 10.3390/nu10010029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Ren G., Kim T., Kim H.S., et al. A small molecule, UAB126, reverses diet-induced obesity and its associated metabolic disorders. Diabetes. 2020;69:2003–2016. doi: 10.2337/db19-1001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Puengel T., Liu H., Guillot A., Heymann F., Tacke F., Peiseler M. Nuclear receptors linking metabolism, inflammation, and fibrosis in nonalcoholic fatty liver disease. Int J Mol Sci. 2022;23:2668. doi: 10.3390/ijms23052668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Königshofer P., Brusilovskaya K., Petrenko O., et al. Nuclear receptors in liver fibrosis. Biochim Biophys Acta, Mol Basis Dis. 2021;1867 doi: 10.1016/j.bbadis.2021.166235. [DOI] [PubMed] [Google Scholar]
  • 96.Cariello M., Piccinin E., Moschetta A. Transcriptional regulation of metabolic pathways via lipid-sensing nuclear receptors PPARs, FXR, and LXR in NASH. Cell Mol Gastroenterol Hepatol. 2021;11:1519–1539. doi: 10.1016/j.jcmgh.2021.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Wahli W., Michalik L. PPARs at the crossroads of lipid signaling and inflammation. Trends Endocrinol Metab. 2012;23:351–363. doi: 10.1016/j.tem.2012.05.001. [DOI] [PubMed] [Google Scholar]
  • 98.Staels B., Butruille L., Francque S. Treating NASH by targeting peroxisome proliferator-activated receptors. J Hepatol. 2023;79:1302–1316. doi: 10.1016/j.jhep.2023.07.004. [DOI] [PubMed] [Google Scholar]
  • 99.Sinha R.A., Rajak S., Singh B.K., Yen P.M. Hepatic lipid catabolism via PPARα-lysosomal crosstalk. Int J Mol Sci. 2020;21:2391. doi: 10.3390/ijms21072391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Frohnert B.I., Hui T.Y., Bernlohr D.A. Identification of a functional peroxisome proliferator-responsive element in the murine fatty acid transport protein gene. J Biol Chem. 1999;274:3970–3977. doi: 10.1074/jbc.274.7.3970. [DOI] [PubMed] [Google Scholar]
  • 101.Gulick T., Cresci S., Caira T., Moore D.D., Kelly D.P. The peroxisome proliferator-activated receptor regulates mitochondrial fatty acid oxidative enzyme gene expression. Proc Natl Acad Sci U S A. 1994;91:11012–11016. doi: 10.1073/pnas.91.23.11012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Lundåsen T., Hunt M.C., Nilsson L.M., et al. PPARalpha is a key regulator of hepatic FGF21. Biochem Biophys Res Commun. 2007;360:437–440. doi: 10.1016/j.bbrc.2007.06.068. [DOI] [PubMed] [Google Scholar]
  • 103.Montagner A., Polizzi A., Fouché E., et al. Liver PPARα is crucial for whole-body fatty acid homeostasis and is protective against NAFLD. Gut. 2016;65:1202–1214. doi: 10.1136/gutjnl-2015-310798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Fernández-Alvarez A., Alvarez M.S., Gonzalez R., Cucarella C., Muntané J., Casado M. Human SREBP1c expression in liver is directly regulated by peroxisome proliferator-activated receptor alpha (PPARalpha) J Biol Chem. 2011;286:21466–21477. doi: 10.1074/jbc.M110.209973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.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 Invest. 1999;103:1489–1498. doi: 10.1172/JCI6223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Mansouri R.M., Baugé E., Staels B., Gervois P. Systemic and distal repercussions of liver-specific peroxisome proliferator-activated receptor-alpha control of the acute-phase response. Endocrinology. 2008;149:3215–3223. doi: 10.1210/en.2007-1339. [DOI] [PubMed] [Google Scholar]
  • 107.Ip E., Farrell G., Hall P., Robertson G., Leclercq I. Administration of the potent PPARalpha agonist, Wy-14,643, reverses nutritional fibrosis and steatohepatitis in mice. Hepatology. 2004;39:1286–1296. doi: 10.1002/hep.20170. [DOI] [PubMed] [Google Scholar]
  • 108.Pawlak M., Baugé E., Bourguet W., et al. The transrepressive activity of peroxisome proliferator-activated receptor alpha is necessary and sufficient to prevent liver fibrosis in mice. Hepatology. 2014;60:1593–1606. doi: 10.1002/hep.27297. [DOI] [PubMed] [Google Scholar]
  • 109.Gervois P., Vu-Dac N., Kleemann R., et al. Negative regulation of human fibrinogen gene expression by peroxisome proliferator-activated receptor alpha agonists via inhibition of CCAAT box/enhancer-binding protein beta. J Biol Chem. 2001;276:33471–33477. doi: 10.1074/jbc.M102839200. [DOI] [PubMed] [Google Scholar]
  • 110.Francque S., Verrijken A., Caron S., et al. PPARα gene expression correlates with severity and histological treatment response in patients with non-alcoholic steatohepatitis. J Hepatol. 2015;63:164–173. doi: 10.1016/j.jhep.2015.02.019. [DOI] [PubMed] [Google Scholar]
  • 111.Nakajima A., Eguchi Y., Yoneda M., et al. Randomised clinical trial: pemafibrate, a novel selective peroxisome proliferator-activated receptor α modulator (SPPARMα), versus placebo in patients with non-alcoholic fatty liver disease. Aliment Pharmacol Ther. 2021;54:1263–1277. doi: 10.1111/apt.16596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Ratziu V., Harrison S.A., Francque S., et al. Elafibranor, an agonist of the peroxisome proliferator-activated receptor-α and -δ, induces resolution of nonalcoholic steatohepatitis without fibrosis worsening. Gastroenterology. 2016;150:1147–1159.e5. doi: 10.1053/j.gastro.2016.01.038. [DOI] [PubMed] [Google Scholar]
  • 113.Loomba R., Sanyal A.J., Kowdley K.V., et al. Randomized, controlled trial of the FGF21 analogue pegozafermin in NASH. N Engl J Med. 2023;389:998–1008. doi: 10.1056/NEJMoa2304286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Flowers M.T., Ntambi J.M. Role of stearoyl-coenzyme A desaturase in regulating lipid metabolism. Curr Opin Lipidol. 2008;19:248–256. doi: 10.1097/MOL.0b013e3282f9b54d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Liu S., Hatano B., Zhao M., et al. Role of peroxisome proliferator-activated receptor {delta}/{beta} in hepatic metabolic regulation. J Biol Chem. 2011;286:1237–1247. doi: 10.1074/jbc.M110.138115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Zarei M., Barroso E., Palomer X., et al. Hepatic regulation of VLDL receptor by PPARβ/δ and FGF21 modulates non-alcoholic fatty liver disease. Mol Metab. 2018;8:117–131. doi: 10.1016/j.molmet.2017.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Inoue M., Ohtake T., Motomura W., et al. Increased expression of PPARgamma in high fat diet-induced liver steatosis in mice. Biochem Biophys Res Commun. 2005;336:215–222. doi: 10.1016/j.bbrc.2005.08.070. [DOI] [PubMed] [Google Scholar]
  • 118.Morán-Salvador E., López-Parra M., García-Alonso V., et al. Role for PPARγ in obesity-induced hepatic steatosis as determined by hepatocyte- and macrophage-specific conditional knockouts. FASEB J. 2011;25:2538–2550. doi: 10.1096/fj.10-173716. [DOI] [PubMed] [Google Scholar]
  • 119.Matsusue K., Haluzik M., Lambert G., et al. Liver-specific disruption of PPARgamma in leptin-deficient mice improves fatty liver but aggravates diabetic phenotypes. J Clin Invest. 2003;111:737–747. doi: 10.1172/JCI17223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Nan Y.M., Fu N., Wu W.J., et al. Rosiglitazone prevents nutritional fibrosis and steatohepatitis in mice. Scand J Gastroenterol. 2009;44:358–365. doi: 10.1080/00365520802530861. [DOI] [PubMed] [Google Scholar]
  • 121.Hazra S., Miyahara T., Rippe R.A., Tsukamoto H. PPAR gamma and hepatic stellate cells. Comp Hepatol. 2004;Suppl 1:S7. doi: 10.1186/1476-5926-2-S1-S7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Ni X.X., Ji P.X., Chen Y.X., et al. Regulation of the macrophage-hepatic stellate cell interaction by targeting macrophage peroxisome proliferator-activated receptor gamma to prevent non-alcoholic steatohepatitis progression in mice. Liver Int. 2022;42:2696–2712. doi: 10.1111/liv.15441. [DOI] [PubMed] [Google Scholar]
  • 123.Ni X.X., Li X.Y., Wang Q., Hua J. Regulation of peroxisome proliferator-activated receptor-gamma activity affects the hepatic stellate cell activation and the progression of NASH via TGF-β1/Smad signaling pathway. J Physiol Biochem. 2021;77:35–45. doi: 10.1007/s13105-020-00777-7. [DOI] [PubMed] [Google Scholar]
  • 124.Panebianco C., Oben J.A., Vinciguerra M., Pazienza V. Senescence in hepatic stellate cells as a mechanism of liver fibrosis reversal: a putative synergy between retinoic acid and PPAR-gamma signalings. Clin Exp Med. 2017;17:269–280. doi: 10.1007/s10238-016-0438-x. [DOI] [PubMed] [Google Scholar]
  • 125.Luo W., Xu Q., Wang Q., Wu H., Hua J. Effect of modulation of PPAR-γ activity on Kupffer cells M1/M2 polarization in the development of non-alcoholic fatty liver disease. Sci Rep. 2017;7 doi: 10.1038/srep44612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Morán-Salvador E., Titos E., Rius B., et al. Cell-specific PPARγ deficiency establishes anti-inflammatory and anti-fibrogenic properties for this nuclear receptor in non-parenchymal liver cells. J Hepatol. 2013;59:1045–1053. doi: 10.1016/j.jhep.2013.06.023. [DOI] [PubMed] [Google Scholar]
  • 127.Wu H.M., Ni X.X., Xu Q.Y., Wang Q., Li X.Y., Hua J. Regulation of lipid-induced macrophage polarization through modulating peroxisome proliferator-activated receptor-gamma activity affects hepatic lipid metabolism via a Toll-like receptor 4/NF-κB signaling pathway. J Gastroenterol Hepatol. 2020;35:1998–2008. doi: 10.1111/jgh.15025. [DOI] [PubMed] [Google Scholar]
  • 128.Odegaard J.I., Ricardo-Gonzalez R.R., Goforth M.H., et al. Macrophage-specific PPARgamma controls alternative activation and improves insulin resistance. Nature. 2007;447:1116–1120. doi: 10.1038/nature05894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Pettinelli P., Videla L.A. Up-regulation of PPAR-gamma mRNA expression in the liver of obese patients: an additional reinforcing lipogenic mechanism to SREBP-1c induction. J Clin Endocrinol Metab. 2011;96:1424–1430. doi: 10.1210/jc.2010-2129. [DOI] [PubMed] [Google Scholar]
  • 130.Harrison S.A., Alkhouri N., Davison B.A., et al. Insulin sensitizer MSDC-0602K in non-alcoholic steatohepatitis: a randomized, double-blind, placebo-controlled phase IIb study. J Hepatol. 2020;72:613–626. doi: 10.1016/j.jhep.2019.10.023. [DOI] [PubMed] [Google Scholar]
  • 131.Aithal G.P., Thomas J.A., Kaye P.V., et al. Randomized, placebo-controlled trial of pioglitazone in nondiabetic subjects with nonalcoholic steatohepatitis. Gastroenterology. 2008;135:1176–1184. doi: 10.1053/j.gastro.2008.06.047. [DOI] [PubMed] [Google Scholar]
  • 132.Ratziu V., Giral P., Jacqueminet S., et al. Rosiglitazone for nonalcoholic steatohepatitis: one-year results of the randomized placebo-controlled Fatty Liver Improvement with Rosiglitazone Therapy (FLIRT) trial. Gastroenterology. 2008;135:100–110. doi: 10.1053/j.gastro.2008.03.078. [DOI] [PubMed] [Google Scholar]
  • 133.Gawrieh S., Noureddin M., Loo N., et al. Saroglitazar, a PPAR-α/γ agonist, for treatment of NAFLD: a randomized controlled double-blind phase 2 trial. Hepatology. 2021;74:1809–1824. doi: 10.1002/hep.31843. [DOI] [PubMed] [Google Scholar]
  • 134.Francque S.M., Bedossa P., Ratziu V., et al. A randomized, controlled trial of the pan-PPAR agonist lanifibranor in NASH. N Engl J Med. 2021;385:1547–1558. doi: 10.1056/NEJMoa2036205. [DOI] [PubMed] [Google Scholar]
  • 135.Panzitt K., Wagner M. FXR in liver physiology: multiple faces to regulate liver metabolism. Biochim Biophys Acta Mol Basis Dis. 2021;1867 doi: 10.1016/j.bbadis.2021.166133. [DOI] [PubMed] [Google Scholar]
  • 136.Chiang J.Y. Bile acid metabolism and bile acid receptor signaling in metabolic diseases and therapy. Liver Res. 2021;5:103–104. doi: 10.1016/j.livres.2021.08.002. [DOI] [Google Scholar]
  • 137.Ma Y., Huang Y., Yan L., Gao M., Liu D. Synthetic FXR agonist GW4064 prevents diet-induced hepatic steatosis and insulin resistance. Pharm Res. 2013;30:1447–1457. doi: 10.1007/s11095-013-0986-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Gai Z., Visentin M., Gui T., et al. Effects of farnesoid X receptor activation on arachidonic acid metabolism, NF-kB signaling, and hepatic inflammation. Mol Pharmacol. 2018;94:802–811. doi: 10.1124/mol.117.111047. [DOI] [PubMed] [Google Scholar]
  • 139.Zhang S., Wang J., Liu Q., Harnish D.C. Farnesoid X receptor agonist WAY-362450 attenuates liver inflammation and fibrosis in murine model of non-alcoholic steatohepatitis. J Hepatol. 2009;51:380–388. doi: 10.1016/j.jhep.2009.03.025. [DOI] [PubMed] [Google Scholar]
  • 140.Clifford B.L., Sedgeman L.R., Williams K.J., et al. FXR activation protects against NAFLD via bile-acid-dependent reductions in lipid absorption. Cell Metab. 2021;33:1671–1684.e4. doi: 10.1016/j.cmet.2021.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Watanabe M., Houten S.M., Wang L., et al. Bile acids lower triglyceride levels via a pathway involving FXR, SHP, and SREBP-1c. J Clin Invest. 2004;113:1408–1418. doi: 10.1172/JCI21025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Pineda Torra I., Claudel T., Duval C., Kosykh V., Fruchart J.C., Staels B. Bile acids induce the expression of the human peroxisome proliferator-activated receptor alpha gene via activation of the farnesoid X receptor. Mol Endocrinol. 2003;17:259–272. doi: 10.1210/me.2002-0120. [DOI] [PubMed] [Google Scholar]
  • 143.Xu W., Cui C., Cui C., et al. Hepatocellular cystathionine γ lyase/hydrogen sulfide attenuates nonalcoholic fatty liver disease by activating farnesoid X receptor. Hepatology. 2022;76:1794–1810. doi: 10.1002/hep.32577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Fickert P., Fuchsbichler A., Moustafa T., et al. Farnesoid X receptor critically determines the fibrotic response in mice but is expressed to a low extent in human hepatic stellate cells and periductal myofibroblasts. Am J Pathol. 2009;175:2392–2405. doi: 10.2353/ajpath.2009.090114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Schumacher J.D., Guo G.L. Pharmacologic modulation of bile acid-FXR-FGF15/FGF19 pathway for the treatment of nonalcoholic steatohepatitis. Handb Exp Pharmacol. 2019;256:325–357. doi: 10.1007/164_2019_228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Tian S., Chen M., Wang B., Han Y., Shang H., Chen J. Salvianolic acid B blocks hepatic stellate cell activation via FGF19/FGFR4 signaling. Ann Hepatol. 2021;20 doi: 10.1016/j.aohep.2020.07.013. [DOI] [PubMed] [Google Scholar]
  • 147.Neuschwander-Tetri B.A., Loomba R., Sanyal A.J., et al. Farnesoid X nuclear receptor ligand obeticholic acid for non-cirrhotic, non-alcoholic steatohepatitis (FLINT): a multicentre, randomised, placebo-controlled trial. Lancet. 2015;385:956–965. doi: 10.1016/S0140-673661933-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Sanyal A.J., Ratziu V., Loomba R., et al. Results from a new efficacy and safety analysis of the REGENERATE trial of obeticholic acid for treatment of pre-cirrhotic fibrosis due to non-alcoholic steatohepatitis. J Hepatol. 2023;79:1110–1120. doi: 10.1016/j.jhep.2023.07.014. [DOI] [PubMed] [Google Scholar]
  • 149.Sanyal A.J., Lopez P., Lawitz E.J., et al. Tropifexor for nonalcoholic steatohepatitis: an adaptive, randomized, placebo-controlled phase 2a/b trial. Nat Med. 2023;29:392–400. doi: 10.1038/s41591-022-02200-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Sherwin X., Singh B.K., Yen P.M., Sinha R.A. Activation of liver X receptors (LXRs) increases sphingolipid biosynthesis in hepatic cells. Matters Select. 2016;2:1–5. doi: 10.19185/MATTERS.201611000022. [DOI] [Google Scholar]
  • 151.Endo-Umeda K., Makishima M. Liver X receptors regulate cholesterol metabolism and immunity in hepatic nonparenchymal cells. Int J Mol Sci. 2019;20:5045. doi: 10.3390/ijms20205045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Liu Y., Qiu D.K., Ma X. Liver X receptors bridge hepatic lipid metabolism and inflammation. J Dig Dis. 2012;13:69–74. doi: 10.1111/j.1751-2980.2011.00554.x. [DOI] [PubMed] [Google Scholar]
  • 153.Sinha R.A., Singh B.K., Zhou J., et al. Loss of ULK1 increases RPS6KB1-NCOR1 repression of NR1H/LXR-mediated Scd1 transcription and augments lipotoxicity in hepatic cells. Autophagy. 2017;13:169–186. doi: 10.1080/15548627.2016.1235123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Venteclef N., Jakobsson T., Ehrlund A., et al. GPS2-dependent corepressor/SUMO pathways govern anti-inflammatory actions of LRH-1 and LXRbeta in the hepatic acute phase response. Genes Dev. 2010;24:381–395. doi: 10.1101/gad.545110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Ito A., Hong C., Rong X., et al. LXRs link metabolism to inflammation through Abca1-dependent regulation of membrane composition and TLR signaling. Elife. 2015;4 doi: 10.7554/eLife.08009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Endo-Umeda K., Nakashima H., Umeda N., Seki S., Makishima M. Dysregulation of Kupffer cells/macrophages and natural killer T cells in steatohepatitis in LXRα knockout male mice. Endocrinology. 2018;159:1419–1432. doi: 10.1210/en.2017-03141. [DOI] [PubMed] [Google Scholar]
  • 157.Beaven S.W., Wroblewski K., Wang J., et al. Liver X receptor signaling is a determinant of stellate cell activation and susceptibility to fibrotic liver disease. Gastroenterology. 2011;140:1052–1062. doi: 10.1053/j.gastro.2010.11.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Kim H., Park C., Kim T.H. Targeting liver X receptors for the treatment of non-alcoholic fatty liver disease. Cells. 2023;12:1292. doi: 10.3390/cells12091292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Kliewer S.A., Moore J.T., Wade L., et al. An orphan nuclear receptor activated by pregnanes defines a novel steroid signaling pathway. Cell. 1998;92:73–82. doi: 10.1016/s0092-8674(00)80900-9. [DOI] [PubMed] [Google Scholar]
  • 160.Roth A., Looser R., Kaufmann M., Meyer U.A. Sterol regulatory element binding protein 1 interacts with pregnane X receptor and constitutive androstane receptor and represses their target genes. Pharmacogenetics Genom. 2008;18:325–337. doi: 10.1097/FPC.0b013e3282f706e0. [DOI] [PubMed] [Google Scholar]
  • 161.Zhou J., Febbraio M., Wada T., et al. Hepatic fatty acid transporter Cd36 is a common target of LXR, PXR, and PPARgamma in promoting steatosis. Gastroenterology. 2008;134:556–567. doi: 10.1053/j.gastro.2007.11.037. [DOI] [PubMed] [Google Scholar]
  • 162.Zhao L.Y., Xu J.Y., Shi Z., Englert N.A., Zhang S.Y. Pregnane X receptor (PXR) deficiency improves high fat diet-induced obesity via induction of fibroblast growth factor 15 (FGF15) expression. Biochem Pharmacol. 2017;142:194–203. doi: 10.1016/j.bcp.2017.07.019. [DOI] [PubMed] [Google Scholar]
  • 163.Bitter A., Rümmele P., Klein K., et al. Pregnane X receptor activation and silencing promote steatosis of human hepatic cells by distinct lipogenic mechanisms. Arch Toxicol. 2015;89:2089–2103. doi: 10.1007/s00204-014-1348-x. [DOI] [PubMed] [Google Scholar]
  • 164.Sayaf K., Zanotto I., Russo F.P., Gabbia D., De Martin S. The nuclear receptor PXR in chronic liver disease. Cells. 2021;11:61. doi: 10.3390/cells11010061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Feng D., Liu T., Sun Z., et al. A circadian rhythm orchestrated by histone deacetylase 3 controls hepatic lipid metabolism. Science. 2011;331:1315–1319. doi: 10.1126/science.1198125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Zhang Y., Fang B., Emmett M.J., et al. GENE REGULATION. Discrete functions of nuclear receptor Rev-erbα couple metabolism to the clock. Science. 2015;348:1488–1492. doi: 10.1126/science.aab3021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Solt L.A., Wang Y., Banerjee S., et al. Regulation of circadian behaviour and metabolism by synthetic REV-ERB agonists. Nature. 2012;485:62–68. doi: 10.1038/nature11030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Griffett K., Hayes M.E., Boeckman M.P., Burris T.P. The role of REV-ERB in NASH. Acta Pharmacol Sin. 2022;43:1133–1140. doi: 10.1038/s41401-022-00883-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Griffett K., Bedia-Diaz G., Elgendy B., Burris T.P. REV-ERB agonism improves liver pathology in a mouse model of NASH. PLoS One. 2020;15 doi: 10.1371/journal.pone.0236000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Duez H., Staels B. The nuclear receptors Rev-erbs and RORs integrate circadian rhythms and metabolism. Diab Vasc Dis Res. 2008;5:82–88. doi: 10.3132/dvdr.2008.0014. [DOI] [PubMed] [Google Scholar]
  • 171.Solt L.A., Burris T.P. 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]
  • 172.Kang H.S., Okamoto K., Takeda Y., et al. Transcriptional profiling reveals a role for RORalpha in regulating gene expression in obesity-associated inflammation and hepatic steatosis. Physiol Genom. 2011;43:818–828. doi: 10.1152/physiolgenomics.00206.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Lau P., Fitzsimmons R.L., Raichur S., Wang S.C., Lechtken A., Muscat G.E. 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]
  • 174.Zhang Y., Papazyan R., Damle M., et al. The hepatic circadian clock fine-tunes the lipogenic response to feeding through RORα/γ. Genes Dev. 2017;31:1202–1211. doi: 10.1101/gad.302323.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Kim K., Boo K., Yu Y.S., et al. RORα controls hepatic lipid homeostasis via negative regulation of PPARγ transcriptional network. Nat Commun. 2017;8:162. doi: 10.1038/s41467-017-00215-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Han Y.H., Kim H.J., Na H., et al. RORα Induces KLF4-mediated M2 polarization in the liver macrophages that protect against nonalcoholic steatohepatitis. Cell Rep. 2017;20:124–135. doi: 10.1016/j.celrep.2017.06.017. [DOI] [PubMed] [Google Scholar]
  • 177.He B., Nohara K., Park N., et al. The small molecule nobiletin targets the molecular Oscillator to enhance circadian rhythms and protect against metabolic syndrome. Cell Metab. 2016;23:610–621. doi: 10.1016/j.cmet.2016.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Kumar N., Kojetin D.J., Solt L.A., et al. Identification of SR3335 (ML-176): a synthetic RORα selective inverse agonist. ACS Chem Biol. 2011;6:218–222. doi: 10.1021/cb1002762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Xia H., Dufour C.R., Giguère V. ERRα as a Bridge between transcription and function: role in liver metabolism and disease. Front Endocrinol(Lausanne) 2019;10:206. doi: 10.3389/fendo.2019.00206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Singh B.K., Sinha R.A., Tripathi M., et al. Thyroid hormone receptor and ERRα coordinately regulate mitochondrial fission, mitophagy, biogenesis, and function. Sci Signal. 2018;11 doi: 10.1126/scisignal.aam5855. [DOI] [PubMed] [Google Scholar]
  • 181.Luo J., Sladek R., Carrier J., Bader J.A., Richard D., Giguère V. Reduced fat mass in mice lacking orphan nuclear receptor estrogen-related receptor alpha. Mol Cell Biol. 2003;23:7947–7956. doi: 10.1128/MCB.23.22.7947-7956.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Chaveroux C., Eichner L.J., Dufour C.R., et al. Molecular and genetic crosstalks between mTOR and ERRα are key determinants of rapamycin-induced nonalcoholic fatty liver. Cell Metab. 2013;17:586–598. doi: 10.1016/j.cmet.2013.03.003. [DOI] [PubMed] [Google Scholar]
  • 183.B’chir W., Dufour C.R., Ouellet C., et al. Divergent role of estrogen-related receptor α in lipid- and fasting-induced hepatic steatosis in mice. Endocrinology. 2018;159:2153–2164. doi: 10.1210/en.2018-00115. [DOI] [PubMed] [Google Scholar]
  • 184.Yang M., Liu Q., Huang T., et al. Dysfunction of estrogen-related receptor alpha-dependent hepatic VLDL secretion contributes to sex disparity in NAFLD/NASH development. Theranostics. 2020;10:10874–10891. doi: 10.7150/thno.47037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Chen C.Y., Li Y., Zeng N., et al. Inhibition of estrogen-related receptor α blocks liver steatosis and steatohepatitis and attenuates triglyceride biosynthesis. Am J Pathol. 2021;191:1240–1254. doi: 10.1016/j.ajpath.2021.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Tian J., Marino R., Johnson C., Locker J. Binding of drug-activated CAR/Nr1i3 alters metabolic regulation in the liver. iScience. 2018;9:209–228. doi: 10.1016/j.isci.2018.10.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Wahlang B., Prough R.A., Falkner K.C., et al. Polychlorinated biphenyl-xenobiotic nuclear receptor interactions regulate energy metabolism, behavior, and inflammation in non-alcoholic-steatohepatitis. Toxicol Sci. 2016;149:396–410. doi: 10.1093/toxsci/kfv250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Marmugi A., Lukowicz C., Lasserre F., et al. Activation of the constitutive androstane receptor induces hepatic lipogenesis and regulates Pnpla3 gene expression in a LXR-independent way. Toxicol Appl Pharmacol. 2016;303:90–100. doi: 10.1016/j.taap.2016.05.006. [DOI] [PubMed] [Google Scholar]
  • 189.Dauwe Y., Mary L., Oliviero F., et al. Steatosis and metabolic disorders associated with synergistic activation of the CAR/RXR heterodimer by pesticides. Cells. 2023;12:1201. doi: 10.3390/cells12081201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Takizawa D., Kakizaki S., Horiguchi N., Yamazaki Y., Tojima H., Mori M. Constitutive active/androstane receptor promotes hepatocarcinogenesis in a mouse model of non-alcoholic steatohepatitis. Carcinogenesis. 2011;32:576–583. doi: 10.1093/carcin/bgq277. [DOI] [PubMed] [Google Scholar]
  • 191.Yamazaki Y., Kakizaki S., Horiguchi N., et al. The role of the nuclear receptor constitutive androstane receptor in the pathogenesis of non-alcoholic steatohepatitis. Gut. 2007;56:565–574. doi: 10.1136/gut.2006.093260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Dong B., Saha P.K., Huang W., et al. Activation of nuclear receptor CAR ameliorates diabetes and fatty liver disease. Proc Natl Acad Sci U S A. 2009;106:18831–18836. doi: 10.1073/pnas.0909731106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Elbel E.E., Lavine J.E., Downes M., et al. Hepatic nuclear receptor expression associates with features of histology in pediatric nonalcoholic fatty liver disease. Hepatol Commun. 2018;2:1213–1226. doi: 10.1002/hep4.1232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Seol W., Choi H.S., Moore D.D. An orphan nuclear hormone receptor that lacks a DNA binding domain and heterodimerizes with other receptors. Science. 1996;272:1336–1339. doi: 10.1126/science.272.5266.1336. [DOI] [PubMed] [Google Scholar]
  • 195.Boulias K., Katrakili N., Bamberg K., Underhill P., Greenfield A., Talianidis I. Regulation of hepatic metabolic pathways by the orphan nuclear receptor SHP. EMBO J. 2005;24:2624–2633. doi: 10.1038/sj.emboj.7600728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Benet M., Guzmán C., Pisonero-Vaquero S., et al. Repression of the nuclear receptor small heterodimer partner by steatotic drugs and in advanced nonalcoholic fatty liver disease. Mol Pharmacol. 2015;87:582–594. doi: 10.1124/mol.114.096313. [DOI] [PubMed] [Google Scholar]
  • 197.Kim Y.C., Qi M., Dong X., et al. Transgenic mice lacking FGF15/19-SHP phosphorylation display altered bile acids and gut bacteria, promoting nonalcoholic fatty liver disease. J Biol Chem. 2023;299 doi: 10.1016/j.jbc.2023.104946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Magee N., Zou A., Ghosh P., Ahamed F., Delker D., Zhang Y. Disruption of hepatic small heterodimer partner induces dissociation of steatosis and inflammation in experimental nonalcoholic steatohepatitis. J Biol Chem. 2020;295:994–1008. doi: 10.1074/jbc.RA119.010233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Myronovych A., Salazar-Gonzalez R.M., Ryan K.K., et al. The role of small heterodimer partner in nonalcoholic fatty liver disease improvement after sleeve gastrectomy in mice. Obesity (Silver Spring) 2014;22:2301–2311. doi: 10.1002/oby.20890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Zhou L.M., Fan J.H., Xu M.M., et al. Epiberberine regulates lipid synthesis through SHP (NR0B2) to improve non-alcoholic steatohepatitis. Biochim Biophys Acta Mol Basis Dis. 2023;1869 doi: 10.1016/j.bbadis.2023.166639. [DOI] [PubMed] [Google Scholar]
  • 201.Zou A., Magee N., Deng F., Lehn S., Zhong C., Zhang Y. Hepatocyte nuclear receptor SHP suppresses inflammation and fibrosis in a mouse model of nonalcoholic steatohepatitis. J Biol Chem. 2018;293:8656–8671. doi: 10.1074/jbc.RA117.001653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Huang J., Iqbal J., Saha P.K., et al. Molecular characterization of the role of orphan receptor small heterodimer partner in development of fatty liver. Hepatology. 2007;46:147–157. doi: 10.1002/hep.21632. [DOI] [PubMed] [Google Scholar]
  • 203.Bechmann L.P., Kocabayoglu P., Sowa J.P., et al. Free fatty acids repress small heterodimer partner (SHP) activation and adiponectin counteracts bile acid-induced liver injury in superobese patients with nonalcoholic steatohepatitis. Hepatology. 2013;57:1394–1406. doi: 10.1002/hep.26225. [DOI] [PubMed] [Google Scholar]
  • 204.Huang K.W., Reebye V., Czysz K., et al. Liver activation of hepatocellular nuclear factor-4α by small activating RNA rescues dyslipidemia and improves metabolic profile. Mol Ther Nucleic Acids. 2020;19:361–370. doi: 10.1016/j.omtn.2019.10.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Ren H., Hu F., Wang D., et al. Sirtuin 2 prevents liver steatosis and metabolic disorders by deacetylation of hepatocyte nuclear factor 4α. Hepatology. 2021;74:723–740. doi: 10.1002/hep.31773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Thymiakou E., Othman A., Hornemann T., Kardassis D. Defects in high density lipoprotein metabolism and hepatic steatosis in mice with liver-specific ablation of hepatocyte nuclear factor 4A. Metabolism. 2020;110 doi: 10.1016/j.metabol.2020.154307. [DOI] [PubMed] [Google Scholar]
  • 207.Xu Y., Hu S., Jadhav K., et al. Hepatocytic activating transcription factor 3 protects against steatohepatitis via hepatocyte nuclear factor 4α. Diabetes. 2021;70:2506–2517. doi: 10.2337/db21-0181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Xu Y., Zhu Y., Hu S., et al. Hepatocyte nuclear factor 4α prevents the steatosis-to-NASH progression by regulating p53 and bile acid signaling (in mice) Hepatology. 2021;73:2251–2265. doi: 10.1002/hep.31604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Yu D., Chen G., Pan M., et al. High fat diet-induced oxidative stress blocks hepatocyte nuclear factor 4α and leads to hepatic steatosis in mice. J Cell Physiol. 2018;233:4770–4782. doi: 10.1002/jcp.26270. [DOI] [PubMed] [Google Scholar]
  • 210.Kiselyuk A., Lee S.H., Farber-Katz S., et al. HNF4α antagonists discovered by a high-throughput screen for modulators of the human insulin promoter. Chem Biol. 2012;19:806–818. doi: 10.1016/j.chembiol.2012.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Gunewardena S., Huck I., Walesky C., Robarts D., Weinman S., Apte U. Progressive loss of hepatocyte nuclear factor 4 alpha activity in chronic liver diseases in humans. Hepatology. 2022;76:372–386. doi: 10.1002/hep.32326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Sun Y., Demagny H., Schoonjans K. Emerging functions of the nuclear receptor LRH-1 in liver physiology and pathology. Biochim Biophys Acta Mol Basis Dis. 2021;1867 doi: 10.1016/j.bbadis.2021.166145. [DOI] [PubMed] [Google Scholar]
  • 213.Miranda D.A., Krause W.C., Cazenave-Gassiot A., et al. LRH-1 regulates hepatic lipid homeostasis and maintains arachidonoyl phospholipid pools critical for phospholipid diversity. JCI Insight. 2018;3 doi: 10.1172/jci.insight.96151. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Liver Research are provided here courtesy of Third Affiliated Hospital of Sun Yat-sen University

RESOURCES