v
Search
Advanced

Publications > Journals > Journal of Clinical and Translational Hepatology> Article Full Text

  • OPEN ACCESS

Orphan Nuclear Receptors in Metabolic Dysfunction-associated Steatotic Liver Disease Development

  • Huan Li#,
  • Jian Chen#,
  • Ziyin Huang and
  • Mingkai Chen* 
 Author information 

Abstract

Metabolic dysfunction-associated fatty liver disease, representing a spectrum of liver disorders from simple steatosis to metabolic dysfunction-associated steatohepatitis, fibrosis, and cirrhosis, has emerged as one of the most prevalent chronic liver conditions globally, affecting an estimated approximately 30% of the world's population. Its pathogenesis is highly complex, involving intricate interactions between genetic predisposition, metabolic dysregulation, inflammation, and cellular stress responses. Within this complex landscape, orphan nuclear receptors (ONRs) have gained significant attention. Defined by the lack of identified endogenous ligands, ONRs function as master transcriptional regulators controlling diverse biological processes. Crucially, they play pivotal roles in the development and progression of numerous diseases, including metabolic disorders.This review specifically focuses on elucidating the critical contributions of various ONRs to the pathogenesis of metabolic dysfunction-associated fatty liver disease. We examined how these receptors modulate key pathological drivers: lipid metabolism, inflammation,and autophagy.

Graphical Abstract

Keywords

Metabolic dysfunction-associated steatotic liver disease, MASLD, Orphan nuclear receptors, ONRs, SHP, RORs, HNF4α, ERRs, LRH-1

Introduction

Metabolic dysfunction-associated steatotic liver disease (MASLD) has emerged as one of the most prevalent chronic liver conditions worldwide, affecting approximately 30% of the global population.1 MASLD encompasses metabolic dysfunction-associated steatotic liver and metabolic dysfunction-associated steatohepatitis (MASH), including their associated fibrotic stages.2 Research has established a strong correlation between MASLD and systemic metabolic disorders such as obesity, insulin resistance, and type 2 diabetes, indicating that this condition is not merely a localized hepatic pathology but rather a multisystem disease intricately linked to systemic metabolic dysregulation.3

Orphan nuclear receptors, one of the three major subfamilies of nuclear receptors, lack identified endogenous ligands. Among the 48 known nuclear receptors, 25 are classified as orphan nuclear receptors. They function as transcription factors or cofactors, regulating gene transcription and participating in various metabolic processes.4 A summary of orphan nuclear receptors is provided in Table 1.5–39 As core regulators of metabolic homeostasis, orphan nuclear receptors contribute to MASLD pathogenesis. For instance, chicken ovalbumin upstream promoter-transcription factor II (also known as NR2F2) is upregulated in fibrotic kidneys and livers in humans, and overexpression of NR2F2 inhibits the fatty acid oxidation pathway in fibroblasts.40 During fasting, estrogen-related receptor (ERR) γ (also known as NR3B3) undergoes O-GlcNAcylation by O-GlcNAc transferase, which increases its stability and enhances its recruitment to gluconeogenic gene promoters, thereby upregulating gluconeogenic gene expression in the liver.41 Small heterodimer partner (SHP, also known as NR0B2) inhibits the conversion of cholesterol to bile acids and plays a key role in maintaining cholesterol and bile acid homeostasis.42 The farnesoid X receptor (FXR) agonist GSK2324 reduces hepatic fatty acid uptake and synthesis to control liver lipid levels, while bile acid supplementation reverses this effect, suggesting that bile acid metabolism plays a critical role in hepatic lipid metabolism.43 Emerging evidence demonstrates that orphan nuclear receptors exert pleiotropic effects on MASLD progression through distinct yet interconnected mechanisms involving metabolic regulation, inflammation suppression, and fibrogenesis inhibition. However, systematic investigations remain limited, preventing comprehensive delineation of their integrative roles within the lipid metabolism–inflammation–fibrosis axis of MASLD pathogenesis.

Table 1

Information on orphan nuclear receptors

GeneCommon nameFull nameFunction on MASLD
NR0B1DAX-1Dosage-sensitive sex reversalNot or very lowly expressed
NR0B2SHPSmall heterodimer partnerInhibit fat synthesis,57 alleviate inflammation and fibrosis,8,9 and participate in the circadian rhythm regulation of the liver10
NR1D1REV-ERBαReverse strand of ERB-alphaInhibit fat synthesis,11 alleviate inflammation and fibrosis,12 and participate in the circadian rhythm regulation of the liver17,18
NR1D2REV-ERBβReverse strand of ERB-betaNot or very lowly expressed
NR1F1RORαRetinoid-related orphan nuclear receptor-alphaPromote fat breakdown and lipophagy,13,14 influence mitochondrial quality,15 and participate in the circadian rhythm regulation of the liver18
NR1F2RORβRetinoid-related orphan nuclear receptor-betaNot or very lowly expressed
NR1F3RORγRetinoid-related orphan nuclear receptor-gammaIncrease fat accumulation,16 and participate in the circadian rhythm regulation of the liver19
NR2A1HNF4αHepatocyte nuclear factor 4-alphaIncrease fatty acid oxidation and lipophagy,20,21 reduce fat synthesis,22 participate in lipid transport,23 and promote bile acid uptake and synthesis24
NR2A2HNF4γHepatocyte nuclear factor 4-gammaNot or very lowly expressed
NR2C1TR2Testicular nuclear receptors 2Not or very lowly expressed
NR2C2TR4Testicular nuclear receptors 4Not or very lowly expressed
NR2E1TLXNuclear receptor taillessPromote cholesterol and fatty acid synthesis while inhibiting fatty acid oxidation25
NR2E3PNRPhotoreceptor-specific nuclear receptorNot or very lowly expressed
NR2F1COUP-TFIChick ovalbumin upstream promoter-transcription factor-1Not or very lowly expressed
NR2F2COUP-TF-IIChick ovalbumin upstream promoter-transcription factor-2Increase fat accumulation and promote hepatic stellate cell activation26
NR2F6EAR2V-erb-related geneIncrease hepatic uptake of free fatty acids27
NR3B1ERRαEstrogen-related receptor alphaPromote fat synthesis29 and insulin resistance,30 inhibit autophagy activity31 and fat transport32
NR3B2ERRβEstrogen-related receptor betaNot or very lowly expressed
NR3B3ERRγEstrogen-related receptor gammaPromote fat synthesis28
NR4A1Nur77Nerve growth factor-1βInhibit liver inflammation,33 regulate mitochondrial function34
TR3
NGFIB
NR4A2Nurr1Nuclear receptor related-1Involved in fat degeneration,35 liver extracellular matrix formation36
RNR-1
TONOR
NR4A3Nor1Neuron-derived orphan receptor-1Not or very lowly expressed
NR5A1SF-1Steroidogenic factor-1Not or very lowly expressed
NR5A2LRH-1Liver receptor homolog-1Involved in liver lipid accumulation and phospholipid composition,37,38 reduce liver oxidative stress39
NR6A1GCNFGerm cell nuclear factorNot or very lowly expressed

Therefore, this article reviews the roles of orphan nuclear receptors in MASLD, aiming to elucidate their multidimensional regulatory characteristics, highlight their potential as novel therapeutic targets, and provide directions for future research.

NR0

The orphan nuclear receptor NR0 family consists of two members, NR0B1 (DAX-1) and NR0B2 (SHP). Both receptors lack the canonical DNA-binding domain and thus cannot directly regulate transcription. Instead, they function as coactivators or corepressors to modulate cellular activities.4 SHP is an inhibitory nuclear receptor primarily expressed in the liver, heart, pancreas, skeletal muscle, and adipose tissue. It plays a crucial role in regulating cholesterol metabolism, energy homeostasis, and bile acid metabolism.44 Studies have shown that increased SHP expression can alleviate liver lipid accumulation in mice subjected to a high-fat diet (HFD).45 Notably, a point mutation introducing T58A (ACC to GCA) in SHP (NM_011850.3) causes SHP-T58A to upregulate the expression of genes related to fatty acid synthesis and uptake, while downregulating genes associated with fatty acid oxidation. This results in elevated systemic triglyceride levels and renders SHP-T58A mice more susceptible to obesity.46 However, in the study by J. Huang et al., leptin and SHP double-mutant mice exhibited increased very low-density lipoprotein (VLDL) secretion and elevated expression of microsomal triglyceride transfer protein, thereby preventing liver steatosis.47 It remains unclear whether the primary effect is driven by the leptin mutation or the SHP mutation. Furthermore, endogenous ligands of FXR can promote SHP expression by activating FXR, which in turn inhibits sterol regulatory element-binding protein 1-c (SREBP-1c) expression, thereby correcting hypertriglyceridemia in mice fed a high-fat diet.48 Additionally, SHP can bind to liver X receptor α and suppress its expression, regulating multiple genes involved in cholesterol homeostasis.49

In the study by L. M. Zhou et al., SHP binds to SREBP1 and inhibits its transcription and translation, leading to downregulation of the de novo lipogenesis gene fatty acid synthase (Fasn) and suppression of de novo lipogenesis.5 However, Y. C. Kim et al. discovered that SHP inhibits Fasn expression through a different mechanism: SHP recruits DNA methyltransferase-3a to the Fasn promoter, inducing promoter methylation and subsequent epigenetic suppression. Co-expression of DNA methyltransferase-3a and SHP enhances SHP’s inhibitory effect on Fasn promoter activity, supporting this mechanism.6 Additionally, S. Seok found that SHP can bind to the miR-802 promoter to suppress its transcription, thereby reducing Fasn expression and increasing the expression of the fatty acid β-oxidation gene Macad, ultimately lowering triglyceride levels in the liver.7 Studies have also shown that hepatocyte-specific SHP deletion promotes secretion of chemokine (C-C motif) ligand 2 by hepatocytes, leading to macrophage M1 polarization and increased inflammatory responses.50 In a study by N. Magee et al., hepatocyte-specific SHP deficiency in mice fed a high-fat cholesterol and fructose diet further increased the expression of inflammatory and fibrotic genes, exacerbating liver inflammation and fibrosis.8 Conversely, Lee YK et al. found that double knockout of apolipoprotein E and SHP reduced the sensitivity to diet-induced inflammatory responses observed in apolipoprotein E single-knockout mice, without affecting fat accumulation. They suggested that the reduction in inflammatory responses due to SHP knockout is independent of fat accumulation.51 These seemingly contradictory results may arise from differences in dietary conditions or the fact that SHP is a rapidly degraded protein,42 potentially causing inconsistent detection across studies. Significantly, after liver steatosis develops, SHP knockout does not alter the degree of steatosis but increases liver inflammation and fibrosis.8 Additionally, SHP can inhibit the transcription of early growth response-1, attenuating bile duct ligation–induced liver fibrosis in mice.9 Furthermore, SHP knockout can reduce serum lipopolysaccharide and phenylacetic acid levels by modulating gut microbiota composition, thereby improving MASLD.52 Moreover, in the study by S. M. Lee et al., SHP was found to participate in hepatic lipoprotein metabolism related to circadian rhythms through its interaction with retinoic acid receptor-related orphan receptor (ROR) γ.10 In conclusion, SHP’s regulation of MASLD is multifaceted, and further research is needed to elucidate its overall impact on MASLD. The schematic diagram of the intracellular pathway of the NR0 nuclear orphan receptors is summarized in Figure 1, where red arrows represent activation/promotion, blue arrows represent inhibition, and black arrows indicate regulation.

Intracellular pathways of NR0.
Fig. 1  Intracellular pathways of NR0.

Created with BioRender.com. SREBP-1c, sterol regulatory element-binding protein 1c; DNMT3A, DNA methyltransferase 3A; miR-802, microRNA-802; Egr-1, early growth response protein 1; Fasn, fatty acid synthase; TG, triglycerides; KO, knockout; CCL2, C-C motif chemokine ligand 2; M1, classically activated macrophages; RORγ, retinoid-related orphan receptor gamma.

NR1

REV-ERBs (NR1D1/REV-ERBα, NR1D2/REV-ERBβ) and RORs belong to nuclear receptor subfamily 1 and play crucial roles in regulating liver lipid metabolism and circadian rhythms. Studies have found that, compared to wild-type mice, mice with a knockout of the DNA-binding domain (exons 3 and 4) of REV-ERBα exhibit more severe liver steatosis after being fed a high-fat diet,53 suggesting that REV-ERBα has a protective role against liver steatosis. Similarly, J. Guo et al. found that overexpression of REV-ERBα inhibited fatty acid synthesis genes and rescued liver steatosis caused by apolipoprotein A5 deficiency.11 Additionally, D. Zhong et al. demonstrated that increased REV-ERBα expression reduced cytochrome P450 4A14 and increased acyl-CoA thioesterase 4 levels, leading to reduced liver lipid accumulation and improvements in liver damage, inflammation, and fibrosis.12 Studies have also shown that increased expression of RORα promotes macrophage M2 polarization, protecting mice from high-fat diet-induced MASH.54 Y. H. Han et al. found that RORα enhances the transcription and translation of Patatin-like phospholipase domain-containing 3 (also known as adiponutrin), which has triacylglycerol (TAG) hydrolase activity, thereby promoting triglyceride (TG) hydrolysis and alleviating liver lipid accumulation.13 They also discovered that free fatty acids inhibit RORα expression via c-Jun N-terminal kinase, reducing Patatin-like phospholipase domain-containing 3 expression and increasing TAG accumulation.13 H. J. Kim et al. found that RORα is involved in the transcription of genes related to lysosomal function; hepatocyte-specific RORα knockout mice exhibited decreased lysosomal acidity and a significant reduction in the number of autolysosomes containing mitochondria and lipid droplets in the liver.14 Additionally, H. J. Kim et al. demonstrated that RORα knockout downregulated BCL2/adenovirus E1B 19 kDa interacting protein 3 and phosphorylated dynamin-related protein 1, leading to impaired mitochondrial fission in response to nutritional stimuli. This resulted in defects in hepatic mitochondrial quality control, exacerbating diet-induced MASH.15 Furthermore, perillartine, a compound derived from Perilla frutescens, improves lipid deposition and glucose homeostasis in hepatocytes by inhibiting the transcriptional activity of RORγ, suggesting RORγ as a potential therapeutic target for MASLD.16

It is worth mentioning that both REV-ERBα and RORs play important roles in regulating hepatic circadian rhythms.17,18,55 A high-fat diet disrupts the circadian rhythms of lipid metabolism, whereas the docosahexaenoic acid metabolite maresin 1 acts as an endogenous ligand for RORα, enhancing its transcriptional and translational activity.54 As a transcription factor, RORα promotes the transcription of brain and muscle ARNT-like protein 1, whereas REV-ERBα suppresses its transcription by binding to the peroxisome proliferator response element box in the Bmal1 promoter. The balance between RORα and REV-ERBα participates in regulating circadian genes.18 This balance also affects insulin-induced gene-2, a key molecule in lipid metabolism, thereby regulating downstream SREBP activation and lipid accumulation.18 In the study by D. Li et al., compared to HFD mice, high-sucrose diet mice exhibited increased circadian amplitude and median levels of RORγ, suggesting that RORγ may regulate the circadian rhythm system under high-sucrose diet conditions. Further research revealed that RORγ deficiency suppresses SREBP-1c gene expression and reduces hepatic lipid levels.19 The schematic diagram of the intracellular pathway of the NR1 nuclear orphan receptors is summarized in Figure 2.

Intracellular pathways of NR1.
Fig. 2  Intracellular pathways of NR1.

Created with BioRender.com. REV-ERBα, reverse strand of ERB-alpha; P450 4A14, cytochrome P450 family 4 subfamily A member 14; Fasn, fatty acid synthase; Scd1, stearoyl-CoA desaturase 1; BMAL1, brain and muscle ARNT-like 1; INSIG2, insulin induced gene 2; SREBP, sterol regulatory element-binding protein; RORα, retinoid-related orphan receptor alpha; M2, alternatively activated macrophages; PNPLA3, patatin-like phospholipase domain-containing protein 3; TG, triglycerides; KO, knockout; Bnip3, BCL2 interacting protein 3; p-Drp1, phosphorylated dynamin-related protein 1.

NR2

In current MASLD research, significant progress has been made in studying the nuclear orphan receptor NR2 family member NR2A1 (hepatocyte nuclear factor 4 alpha, HNF4α). HNF4α is indispensable for hepatocyte differentiation and is highly expressed in hepatocytes. It regulates genes involved in liver-specific functions such as nutrient metabolism, drug metabolism, urea clearance, bile acid synthesis, and coagulation factor production, all of which are crucial for maintaining liver health.56 HNF4α activity decreases significantly as the liver progresses from dysplasia to early cirrhosis and from cirrhosis to early hepatocellular carcinoma, marking a critical point of liver function decline.56

Studies have shown that liver-specific deletion of Ssu72 phosphatase downregulates HNF4α transcription and translation, leading to hepatocyte dedifferentiation and contributing to the progression of steatohepatitis-associated hepatocellular carcinoma.57 N-trans caffeoyltyramine and N-trans feruloyltyramine can activate HNF4α, increasing the expression of downstream genes such as Spinster homolog 2 and CYP26A1. This enhances dihydroceramide production and action, promoting lipid autophagy and alleviating lipid accumulation.58 Further studies have revealed that N-trans caffeoyltyramine increases mitochondrial mass by enhancing the activity of the PPARGC1A pathway, thereby promoting fatty acid oxidation, reducing mitochondrial stress, and slowing MASLD progression.20 D. H. Lee and colleagues found that the expression of autophagy-related genes is suppressed in fatty liver, while HNF4α can directly bind to the promoter region of UNC-51-like kinase 1, increasing its transcription in hepatocytes and improving autophagy activity.21 HNF4α is also a key regulator of triglyceride hydrolysis and fatty acid oxidation. Liver-specific overexpression of HNF4α can prevent MASH induced by a high-fat/cholesterol/fructose diet by inhibiting p53.59 Studies have shown that HNF4α increases the promoter activity of the murine carboxylesterase 2 gene by binding to DR-1 elements (direct repeats separated by one nucleotide) in the proximal promoter. This regulation modulates lipolysis, endoplasmic reticulum stress, and lipogenesis, thereby preventing hepatic steatosis.60 Thus, the reduction of carboxylesterase 2 in MASH patients is likely due to decreased HNF4α expression. The phosphatidylinositol 3-kinase signaling pathway also plays a crucial role in lipid metabolism. PIK3R3, the 62 kDa regulatory subunit of phosphatidylinositol 3-kinase, promotes hepatic fatty acid oxidation by inducing PPARα expression, thereby ameliorating hepatic steatosis in HFD-induced mice. However, this pathway requires HNF4α involvement. The deletion of HNF4α suppresses the PIK3R3-PPARα pathway, inhibiting fatty acid β-oxidation.61 Notably, C. I. Kasano-Camones found that in HNF4α liver-specific knockout mice fed a Western diet, despite decreased PPARα mRNA and protein levels, increased PPARγ coactivator 1α expression promoted PPARα transactivation, activating downstream target genes that led to steatosis and advanced MASLD progression.62 Additionally, J. Ahn et al. discovered that HNF4α is recruited to the upstream region of the miR-467b-3p transcription start site, mediating its upregulation. miR-467b-3p directly binds to the 3′ untranslated region of glycerol-3-phosphate acyltransferase-1, suppressing its expression, thereby reducing TG synthesis and alleviating steatosis.22 HNF4α is also involved in lipid transport; liver-specific knockout of HNF4α alters the structure and composition of HDL, reducing its antioxidant capacity and affecting genes involved in HDL maturation and modification. This leads to decreased levels of HDL-C, LDL-C, phospholipids, and TG in mice.23 It is generally accepted that a high-fat diet reduces HNF4α expression in hepatocytes.60,63 However, D. Yu and colleagues proposed that the effects of an HFD are not mediated by altering HNF4α expression but rather by increasing oxidative stress, which induces protein kinase C phosphorylation at Ser78. This phosphorylation inhibits HNF4α nuclear translocation, ultimately blocking apolipoprotein B transcription, reducing hepatic VLDL secretion, and promoting hepatic TAG accumulation.64,65 Therefore, the oxidative stress/protein kinase C/HNF4α/ApoB pathway may serve as a novel therapeutic target for HFD-induced fatty liver disease. Beyond lipid metabolism, HNF4α also plays a crucial role in bile acid metabolism. In a study by Y. J. Roh et al., HNF4α knockdown suppressed the expression of genes related to bile acid uptake and synthesis, reducing bile acid toxicity and alleviating steatohepatitis and liver fibrosis. However, while HNF4α knockdown decreases bile acid toxicity, it also inhibits the expression of genes involved in fatty acid oxidation (CPT1A and ACOX1) and fat export (ApoB and MTTP), leading to increased lipotoxicity.24 Thus, HNF4α appears to have opposing roles in lipid and bile acid metabolism.

Other orphan nuclear receptors in the NR2 family also influence MASLD progression. Q. Xiong et al. found that in mice with high-fat diet-induced liver steatosis, expression of nuclear receptor subfamily 2 group E member 1 (NR2E1, TLX) was decreased. Compared to wild-type mice, NR2E1 knockout mice showed increased expression of cholesterol synthesis-related genes (HMGCR, SREBP-1), fatty acid synthesis genes (FAS, ACC), and suppressed expression of genes related to lipid β-oxidation (CPT1A, PPARα). This led to increased cholesterol and fatty acid synthesis and aggravated liver steatosis.25 In contrast, high-fat diets increase the expression of NR2F2. NR2F2 knockout results in elevated adiponectin levels, which alleviate fat accumulation and steatosis in the liver. Additionally, NR2F2 knockout suppresses the expression of α-smooth muscle actin and Pnpla3, inhibiting the activation of hepatic stellate cells.26 It is worth noting that liver-specific overexpression of NR2F6 can increase hepatic triglyceride accumulation without affecting body weight or food intake. Further studies have shown that NR2F6 promotes liver triglyceride accumulation by binding to the CD36 promoter, increasing CD36 expression, and enhancing hepatic fatty acid uptake. This effect is not mediated by changes in circulating β-hydroxybutyrate levels or VLDL secretion rate, ultimately leading to steatosis.27 The schematic diagram of the intracellular pathway of NR2 nuclear orphan receptors is summarized in Figure 3.

Intracellular pathways of NR2.
Fig. 3  Intracellular pathways of NR2.

Created with BioRender.com. SPNS2, spinster homolog 2; CYP26A1, cytochrome P450 family 26 subfamily A member 1; Ulk1, unc-51 like autophagy activating kinase 1; CES2, carboxylesterase 2; miR-467b-3p, microRNA-467b-3p; GPAT1, glycerol-3-phosphate acyltransferase 1; TG, triglycerides; PIK3R3, phosphoinositide-3-kinase regulatory subunit 3; PPARα, peroxisome proliferator activated receptor alpha; β-oxidation, beta-oxidation; HDL, high-density lipoprotein; HDL-C, high-density lipoprotein cholesterol; HNF4α, hepatocyte nuclear factor 4-alpha.

NR3

ERRs α, β, and γ, members of nuclear receptor subfamily 3, are expressed in various tissues and play important roles in regulating bone homeostasis, energy metabolism, and cancer progression.66 Studies have shown that environmental pollutants triclocarban and triclosan can bind to ERRα and ERRγ, enhancing their transcriptional activity. This upregulates genes related to lipogenesis, thereby exacerbating hepatic lipid accumulation induced by a high-fat diet.28 In human MAFL/MASH cohorts, particularly at MASH stages F0–F2, ERRα expression positively correlates with MASH-related genes,67 suggesting that ERRα contributes to MASH progression. In mice fed a high-fat diet, ERRα expression is upregulated. However, treatment with the ERRα inhibitor ERR-PA suppresses the expression of two key enzymes involved in de novo lipogenesis, Fasn and acetyl-CoA carboxylase, indicating that ERRα promotes MASLD progression by influencing de novo lipogenesis.29 ERRα knockout mice exhibit resistance to HFD-induced MASLD, showing reduced white adipose tissue storage and impaired hepatic lipid accumulation. However, under fasting and refeeding conditions, ERRα knockout impairs the reversal of fasting-induced MASLD, indicating that ERRα deletion plays a preventive role in MASLD development rather than a therapeutic one.68 Moreover, ERRα can promote insulin resistance by downregulating oxidative phosphorylation genes, with insulin resistance being a key mechanism in MASLD pathogenesis.30 Interestingly, during the progression from steatosis to steatohepatitis in mice, the mRNA and protein levels of ERRα and Ribosomal large subunit protein 1 are downregulated, leading to reduced translation of autophagy-related proteins and inhibited autophagic activity.31 This suggests ERRα plays distinct roles at different MASLD stages via diverse mechanisms. Furthermore, intermittent fasting was found to promote the expression of ERRα and Ribosomal large subunit protein 1, providing valuable insight for rational dietary interventions in MASH.31 Comparative studies revealed that, in fasting conditions, female mice express higher levels of ERRα than male mice, which leads to sex differences in VLDL-TG assembly and secretion. Liver-specific ERRα deficiency in female mice increased hepatic fat accumulation and worsened MASLD progression, potentially explaining gender differences in MASLD pathogenesis.32 Additionally, ovariectomized mice showed downregulated expression of ERRα, ApoB, and MTTP, developing fatty liver. Overexpression of ERRα reversed these effects, suggesting that estrogen’s protective effect against MASLD is mediated through ERRα.32 The schematic diagram of the intracellular pathway of the NR3 nuclear orphan receptor family is summarized in Figure 4.

Intracellular pathways of NR3-5.
Fig. 4  Intracellular pathways of NR3-5.

Created with BioRender.com. ERRα, estrogen-related receptor alpha; ACC, acetyl-CoA carboxylase; GPAT4, glycerol-3-phosphate acyltransferase 4; OxPHOS, oxidative phosphorylation; Rplp1, ribosomal protein lateral stalk subunit P1; VLDL, very low-density lipoprotein; TG, triglycerides; NR4A1, nuclear receptor subfamily 4 group A member 1; NR4A2, nuclear receptor subfamily 4 group A member 2; M2, alternatively activated macrophages; p-DNA-PKcs, phosphorylated DNA-dependent protein kinase catalytic subunit; P-p53, phosphorylated tumor protein p53; Drp1, dynamin-related protein 1; SREBP-1α, sterol regulatory element-binding protein 1-alpha; Gbp5, guanylate binding protein 5; α-SMA, alpha smooth muscle actin; LRH-1, liver receptor homolog-1; K289R, lysine-289 to arginine mutation; Sod2, superoxide dismutase 2; ROS, reactive oxygen species; Osbpl3, oxysterol binding protein-like 3; KO, knockout; Fads2, fatty acid desaturase 2; Elovl5, ELOVL fatty acid elongase 5.

NR4

The NR4A subfamily of nuclear receptors (NR4A1/Nur77/TR3/NGFI-B, NR4A2/Nurr1, NR4A3/Nor1) are important regulatory proteins involved in various disease states and metabolic conditions, particularly in the liver and muscle.69,70 NR4A expression is upregulated in the adipose tissue of patients with severe obesity, and following weight loss surgery, NR4A levels return to those observed in normal lean controls.70 Bioinformatics analyses indicate that NR4A1 is downregulated in the livers of MASLD patients, where it is implicated in NOD-like receptor and JAK-STAT signaling pathways. NR4A1 plays a role in regulating mitochondrial autophagy and immune activation during MASLD progression, with its expression significantly negatively correlated with infiltration of pro-inflammatory M1 macrophages. Additionally, NR4A2 is dysregulated in MAFLD and may modulate immune responses in the disease, serving as a potential diagnostic marker for MASLD.71–75

J. He et al. demonstrated NR4A1’s role in macrophages, showing that hyperoside upregulates NR4A1 expression, promoting the conversion of pro-inflammatory M1 macrophages to anti-inflammatory M2 macrophages, thereby alleviating MASLD in HFD-fed mice.33 However, H. Zhou et al. reported that NR4A1 can phosphorylate the DNA-dependent protein kinase catalytic subunit. Phosphorylated DNA-dependent protein kinase catalytic subunit dissociates from Ku80 and phosphorylates p53, which increases the expression of dynamin-related protein 1, leading to mitochondrial fission. This mitochondrial dysfunction disrupts mitochondrial homeostasis in MASLD, potentially causing hepatocyte injury or death.34 This finding partially explains the conclusion from Y. Xu et al. that “the inhibitory effect of HNF4α on MASLD is partly mediated through the p53 pathway, as overexpression of p53 can partially counteract the inhibitory effect of HNF4α on steatohepatitis”.59 In addition, NR4A1 can attenuate homocysteine-induced steatosis both in vivo and in vitro, suggesting its potential as a therapeutic target for homocysteine-related MASLD.76 Studies have found that overexpression of NR4A2 upregulates genes related to lipogenesis, liver inflammation, and steatosis, exacerbating steatosis, liver inflammation, and fibrosis.35 Notably, upon stimulation of hepatic stellate cells with the strong inducer PDGF-BB, NR4A2 expression decreases alongside downregulation of phosphorylation levels of ERK1/2, P38, and c-Jun N-terminal kinase—key components of the MAPK pathway involved in liver fibrosis. Concurrently, the fibrosis marker α-smooth muscle actin significantly increases, suggesting that NR4A2 contributes to extracellular matrix accumulation in liver fibrosis through regulation of the MAPK pathway.36 The schematic diagram of the intracellular pathway of NR4 nuclear orphan receptors is summarized in Figure 4.

NR5

NR5A2 (liver receptor homolog-1 (LRH-1)), a member of the NR5A subfamily of nuclear receptors, is expressed in endoderm-derived tissues, including the intestine, liver, exocrine pancreas, and ovaries. It plays a critical role in development, reverse cholesterol transport, bile acid homeostasis, and steroidogenesis.77,78 N. Sahini and J. Borlak performed liver biopsies on healthy controls and MASLD patients and found that various transcription factors, including LRH-1, were suppressed in the livers of MASLD patients.79 Studies in diet-induced MASH mice have shown that phosphatidylcholine levels in the liver are decreased, and supplementation with phosphatidylcholine reduces MASH progression by mechanisms involving activation of B cell signaling via the LRH-1/PPARγ2/nuclear factor κ-light-chain enhancer pathway.80 LRH-1 plays a crucial role in hepatic lipid storage and phospholipid composition. Research has demonstrated that deletion of LRH-1 inhibits two key enzymes in arachidonic acid biosynthesis—long-chain fatty acid desaturase and elongase—resulting in decreased arachidonic acid–containing phospholipids. This disrupts liver phospholipid composition, leading to abnormal lipid accumulation and non-dietary-related hepatic steatosis.37 Notably, dilauroyl phosphatidylcholine (DLPC), an unusual phosphatidylcholine species with two saturated 12-carbon fatty acid acyl chains, acts as an in vitro agonist of LRH-1. DLPC induces bile acid biosynthetic enzymes in the liver, increases bile acid levels, reduces liver triglycerides and serum glucose, and alleviates hepatic steatosis. Furthermore, DLPC improves glucose homeostasis in two mouse models of insulin resistance, indicating that LRH-1 regulates bile acid metabolism and glucose homeostasis via the phosphatidylcholine pathway.81 Additionally, LRH-1 directly regulates the expression of Fasn and promotes the transcription factor ChREBP, which directly or indirectly controls all components of de novo lipogenesis.78 It is important to note that small ubiquitin-related modifier modification-deficient LRH-1—specifically the LRH-1 K289R mutant mouse—is more susceptible to MASLD when fed a high-fat diet. This mutation promotes Osbpl3 expression and subsequently induces SREBP-1 signaling at the transcriptional level, leading to increased Fasn expression and enhanced de novo lipogenesis.38 Additionally, LRH-1 binds to the promoter of superoxide dismutase 2, promoting its expression and reducing reactive oxygen species production induced by high palmitate concentrations, thereby alleviating oxidative stress in MASLD.82 It is noteworthy that SHP can transcriptionally repress LRH-1 by recruiting class III histone deacetylase SIRT1, which inhibits LRH-1 target gene promoter activity and mRNA levels.39 Conversely, LRH-1 can activate the SHP promoter, promoting SHP expression.83 This bidirectional interaction affects their respective roles in MASLD. The SHP-T58A mutant disrupts its interaction with LRH-1, resulting in derepression of SHP/LRH-1 target genes related to bile acid and lipid synthesis, thereby promoting MASLD progression.46 The schematic diagram of the intracellular pathway of the NR5 nuclear orphan receptor family is summarized in Figure 4.

Targeting nuclear orphan receptors for the treatment of MAFLD

Given the critical role of nuclear orphan receptors in lipid metabolism, inflammation, and fibrosis processes in MASLD, researchers have begun exploring their therapeutic potential. Several natural compounds have been identified that improve MASLD by modulating nuclear orphan receptors. For instance, hyperoside acts on SHP,84 6-gingerol targets HNF4α,22 and Angelica sinensis polysaccharide regulates ERRα.85 Additionally, Inonotus obliquus and its bioactive compound inotodiol exert anti-MASLD effects by modulating the FXR/SHP/SREBP-1c pathway.86 In high-fat diet-fed mouse models, long-term oral administration of HNF4α agonists (e.g., NCT) enhances mitochondrial quality and function, preventing weight gain and hepatic steatosis.20 The selective ERRα agonist JND003 enhances the transactivation of ERRα downstream target genes while improving insulin sensitivity and alleviating fatty liver symptoms.87 Dapagliflozin, a selective sodium-glucose cotransporter-2 inhibitor widely used to treat type 2 diabetes,88 reduces de novo lipogenesis in the livers of db/db mice by upregulating FXR/SHP and downregulating liver X receptor α/SREBP-1c.89 To date, no pharmacological agents targeting nuclear orphan receptors have been approved for MASLD therapy, highlighting a significant unmet clinical need. Given their regulatory role in the interconnected pathways of lipid metabolism, inflammation, and fibrosis, the development of novel drugs targeting these receptors represents a promising therapeutic strategy.

Conclusions

In summary, orphan nuclear receptors influence the onset and progression of MASLD, particularly in lipid metabolism, inflammation, autophagy, and cholesterol metabolism. These receptors may play either similar or opposing roles within the same pathological and physiological processes, forming an intricate network of interactions. Among them, SHP and HNF4α have been studied extensively. SHP participates in MASLD progression by influencing lipid synthesis, liver inflammation, fibrosis, and circadian rhythms, while also interacting with other orphan nuclear receptors such as RORγ and LRH-1. HNF4α, in contrast, influences MASLD progression primarily through its impact on lipid and bile acid metabolism. Although orphan nuclear receptors play important roles in MASLD, few drugs currently target these receptors for MASLD treatment. Therefore, the development of therapeutics aimed at orphan nuclear receptors is essential. Moreover, the fact that some orphan nuclear receptors exert opposing effects in the same physiological processes is both intriguing and worthy of further investigation. Additional research is necessary to elucidate these underlying mechanisms, thereby providing a theoretical foundation for the potential clinical use of orphan nuclear receptors as therapeutic targets in the future. In this review, we discuss the contributions of distinct nuclear orphan receptors to MASLD pathogenesis, evaluate emerging therapeutic strategies targeting these receptors, and highlight promising avenues for future MAFLD intervention.

Declarations

Funding

This work was supported by the National Key Research and Development Program of China (2023YFC2507405), and the Interdisciplinary Innovative Talents Foundation from Renmin Hospital of Wuhan University (JCRCFZ-2022-017).

Conflict of interest

The authors have no conflict of interests related to this publication.

Authors’ contributions

Study concept and design (HL), drafting of the manuscript (JC), critical revision of the manuscript for important intellectual content (ZH), administrative, technical, material support, and study supervision (MC). All authors have approved the final version and publication of the manuscript.

References

  1. Quek J, Chan KE, Wong ZY, Tan C, Tan B, Lim WH, et al. Global prevalence of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis in the overweight and obese population: a systematic review and meta-analysis. Lancet Gastroenterol Hepatol 2023;8(1):20-30 View Article PubMed/NCBI
  2. Huang DQ, Wong VWS, Rinella ME, Boursier J, Lazarus JV, Yki-Järvinen H, et al. Metabolic dysfunction-associated steatotic liver disease in adults. Nat Rev Dis Primers 2025;11(1):14 View Article PubMed/NCBI
  3. Targher G, Byrne CD, Tilg H. MASLD: a systemic metabolic disorder with cardiovascular and malignant complications. Gut 2024;73(4):691-702 View Article PubMed/NCBI
  4. Guzmán A, Hughes CHK, Murphy BD. Orphan nuclear receptors in angiogenesis and follicular development. Reproduction 2021;162(3):R35-r54 View Article PubMed/NCBI
  5. Zhou LM, Fan JH, Xu MM, Xiong MY, Wang QJ, Chai X, et al. Epiberberine regulates lipid synthesis through SHP (NR0B2) to improve non-alcoholic steatohepatitis. Biochim Biophys Acta Mol Basis Dis 2023;1869(4):166639 View Article PubMed/NCBI
  6. Kim YC, Seok S, Zhang Y, Ma J, Kong B, Guo G, et al. Intestinal FGF15/19 physiologically repress hepatic lipogenesis in the late fed-state by activating SHP and DNMT3A. Nat Commun 2020;11(1):5969 View Article PubMed/NCBI
  7. Seok S, Sun H, Kim YC, Kemper B, Kemper JK. Defective FXR-SHP Regulation in Obesity Aberrantly Increases miR-802 Expression, Promoting Insulin Resistance and Fatty Liver. Diabetes 2021;70(3):733-744 View Article PubMed/NCBI
  8. 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(4):994-1008 View Article PubMed/NCBI
  9. Zhang Y, Xu N, Xu J, Kong B, Copple B, Guo GL, et al. E2F1 is a novel fibrogenic gene that regulates cholestatic liver fibrosis through the Egr-1/SHP/EID1 network. Hepatology 2014;60(3):919-930 View Article PubMed/NCBI
  10. Lee SM, Zhang Y, Tsuchiya H, Smalling R, Jetten AM, Wang L. Small heterodimer partner/neuronal PAS domain protein 2 axis regulates the oscillation of liver lipid metabolism. Hepatology 2015;61(2):497-505 View Article PubMed/NCBI
  11. Guo J, Miao G, Zhang W, Shi H, Lai P, Xu Y, et al. Depletion of ApoA5 aggravates spontaneous and diet-induced nonalcoholic fatty liver disease by reducing hepatic NR1D1 in hamsters. Theranostics 2024;14(5):2036-2057 View Article PubMed/NCBI
  12. Zhong D, Cai J, Hu C, Chen J, Zhang R, Fan C, et al. Inhibition of mPGES-2 ameliorates NASH by activating NR1D1 via heme. Hepatology 2023;78(2):547-561 View Article PubMed/NCBI
  13. Han YH, Kim HJ, Lee MO. RORα regulates hepatic lipolysis by inducing transcriptional expression of PNPLA3 in mice. Mol Cell Endocrinol 2021;522:111122 View Article PubMed/NCBI
  14. Kim HJ, Han YH, Kim JY, Lee MO. RORα Enhances Lysosomal Acidification and Autophagic Flux in the Hepatocytes. Hepatol Commun 2021;5(12):2121-2138 View Article PubMed/NCBI
  15. Kim HJ, Han YH, Na H, Kim JY, Kim T, Kim HJ, et al. Liver-specific deletion of RORα aggravates diet-induced nonalcoholic steatohepatitis by inducing mitochondrial dysfunction. Sci Rep 2017;7(1):16041 View Article PubMed/NCBI
  16. Xiao Y, Xiao L, Li M, Liu S, Wang Y, Huang L, et al. Perillartine protects against metabolic associated fatty liver in high-fat diet-induced obese mice. Food Funct 2023;14(2):961-977 View Article PubMed/NCBI
  17. Zhao H, Han G, Jiang Z, Gao D, Zhang H, Yang L, et al. Identification of BMAL1-Regulated circadian genes in mouse liver and their potential association with hepatocellular carcinoma: Gys2 and Upp2 as promising candidates. Biochem Biophys Res Commun 2024;696:149422 View Article PubMed/NCBI
  18. Chen R, Zuo Z, Li Q, Wang H, Li N, Zhang H, et al. DHA substitution overcomes high-fat diet-induced disturbance in the circadian rhythm of lipid metabolism. Food Funct 2020;11(4):3621-3631 View Article PubMed/NCBI
  19. Li D, Ikaga R, Ogawa H, Yamazaki T. Different expressions of clock genes in fatty liver induced by high-sucrose and high-fat diets. Chronobiol Int 2021;38(5):762-778 View Article PubMed/NCBI
  20. Veeriah V, Lee SH, Levine F. Long-term oral administration of an HNF4α agonist prevents weight gain and hepatic steatosis by promoting increased mitochondrial mass and function. Cell Death Dis 2022;13(1):89 View Article PubMed/NCBI
  21. Lee DH, Park SH, Ahn J, Hong SP, Lee E, Jang YJ, et al. Mir214-3p and Hnf4a/Hnf4α reciprocally regulate Ulk1 expression and autophagy in nonalcoholic hepatic steatosis. Autophagy 2021;17(9):2415-2431 View Article PubMed/NCBI
  22. Ahn J, Lee H, Jung CH, Ha SY, Seo HD, Kim YI, et al. 6-Gingerol Ameliorates Hepatic Steatosis via HNF4α/miR-467b-3p/GPAT1 Cascade. Cell Mol Gastroenterol Hepatol 2021;12(4):1201-1213 View Article PubMed/NCBI
  23. 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;110:154307 View Article PubMed/NCBI
  24. Roh YJ, Kim Y, Lee JS, Oh JH, Lee SM, Yoon EL, et al. Regulation of Hepatocyte Nuclear Factor 4α Attenuated Lipotoxicity but Increased Bile Acid Toxicity in Non-Alcoholic Fatty Liver Disease. Life (Basel) 2022;12(11):1682 View Article PubMed/NCBI
  25. Xiong Q, Wu Y, Yang M, Wu G, Wang Y, Wang H, et al. Nr2e1 ablation impairs liver glucolipid metabolism and induces inflammation, high-fat diets amplify the damage. Biomed Pharmacother 2019;120:109503 View Article PubMed/NCBI
  26. Mao Z, Zhao J, Cui F, Li Z, Cao J, Zhou J, et al. STUB1 increases adiponectin expression by inducing ubiquitination and degradation of NR2F2, thereby reducing hepatic stellate cell activation and alleviating non-alcoholic fatty liver disease. Tissue Cell 2024;88:102345 View Article PubMed/NCBI
  27. Zhou B, Jia L, Zhang Z, Xiang L, Yuan Y, Zheng P, et al. The Nuclear Orphan Receptor NR2F6 Promotes Hepatic Steatosis through Upregulation of Fatty Acid Transporter CD36. Adv Sci (Weinh) 2020;7(21):2002273 View Article PubMed/NCBI
  28. Li X, Zhang JD, Xiao H, He S, He TT, Ren XM, et al. Triclocarban and triclosan exacerbate high-fat diet-induced hepatic lipid accumulation at environmental related levels: The potential roles of estrogen-related receptors pathways. Sci Total Environ 2023;858(Pt 3):160079 View Article PubMed/NCBI
  29. Chen CY, Li Y, Zeng N, He L, Zhang X, Tu T, et al. Inhibition of Estrogen-Related Receptor α Blocks Liver Steatosis and Steatohepatitis and Attenuates Triglyceride Biosynthesis. Am J Pathol 2021;191(7):1240-1254 View Article PubMed/NCBI
  30. Xia H, Dufour CR, Giguère V. ERRα as a Bridge Between Transcription and Function: Role in Liver Metabolism and Disease. Front Endocrinol (Lausanne) 2019;10:206 View Article PubMed/NCBI
  31. Tripathi M, Gauthier K, Sandireddy R, Zhou J, Guptta P, Sakthivel S, et al. Esrra regulates Rplp1-mediated translation of lysosome proteins suppressed in metabolic dysfunction-associated steatohepatitis and reversed by alternate day fasting. Mol Metab 2024;87:101997 View Article PubMed/NCBI
  32. Yang M, Liu Q, Huang T, Tan W, Qu L, Chen T, et al. Dysfunction of estrogen-related receptor alpha-dependent hepatic VLDL secretion contributes to sex disparity in NAFLD/NASH development. Theranostics 2020;10(24):10874-10891 View Article PubMed/NCBI
  33. Sun B, Zhang R, Liang Z, Fan A, Kang D. Hyperoside attenuates non-alcoholic fatty liver disease through targeting Nr4A1 in macrophages. Int Immunopharmacol 2021;94:107438 View Article PubMed/NCBI
  34. Zhou H, Zhu P, Wang J, Toan S, Ren J. DNA-PKcs promotes alcohol-related liver disease by activating Drp1-related mitochondrial fission and repressing FUNDC1-required mitophagy. Signal Transduct Target Ther 2019;4:56 View Article PubMed/NCBI
  35. Li B, Yang Z, Mao F, Gong W, Su Q, Yang J, et al. Downregulation of microRNA-145a-5p promotes steatosis-to-NASH progression through upregulation of Nr4a2. J Hepatol 2023;79(5):1096-1109 View Article PubMed/NCBI
  36. Chen P, Li J, Huo Y, Lu J, Wan L, Li B, et al. Orphan nuclear receptor NR4A2 inhibits hepatic stellate cell proliferation through MAPK pathway in liver fibrosis. PeerJ 2015;3:e1518 View Article PubMed/NCBI
  37. Miranda DA, Krause WC, Cazenave-Gassiot A, Suzawa M, Escusa H, Foo JC, et al. LRH-1 regulates hepatic lipid homeostasis and maintains arachidonoyl phospholipid pools critical for phospholipid diversity. JCI Insight 2018;3(5):e96151 View Article PubMed/NCBI
  38. Stein S, Lemos V, Xu P, Demagny H, Wang X, Ryu D, et al. Impaired SUMOylation of nuclear receptor LRH-1 promotes nonalcoholic fatty liver disease. J Clin Invest 2017;127(2):583-592 View Article PubMed/NCBI
  39. Chanda D, Xie YB, Choi HS. Transcriptional corepressor SHP recruits SIRT1 histone deacetylase to inhibit LRH-1 transactivation. Nucleic Acids Res 2010;38(14):4607-4619 View Article PubMed/NCBI
  40. Li L, Galichon P, Xiao X, Figueroa-Ramirez AC, Tamayo D, Lee JJ, et al. Orphan nuclear receptor COUP-TFII enhances myofibroblast glycolysis leading to kidney fibrosis. EMBO Rep 2021;22(6):e51169 View Article PubMed/NCBI
  41. Misra J, Kim DK, Jung YS, Kim HB, Kim YH, Yoo EK, et al. O-GlcNAcylation of Orphan Nuclear Receptor Estrogen-Related Receptor γ Promotes Hepatic Gluconeogenesis. Diabetes 2016;65(10):2835-2848 View Article PubMed/NCBI
  42. Miao J, Xiao Z, Kanamaluru D, Min G, Yau PM, Veenstra TD, et al. Bile acid signaling pathways increase stability of Small Heterodimer Partner (SHP) by inhibiting ubiquitin-proteasomal degradation. Genes Dev 2009;23(8):986-996 View Article PubMed/NCBI
  43. Clifford BL, Sedgeman LR, Williams KJ, Morand P, Cheng A, Jarrett KE, et al. FXR activation protects against NAFLD via bile-acid-dependent reductions in lipid absorption. Cell Metab 2021;33(8):1671-1684.e1674 View Article PubMed/NCBI
  44. Yang ZX, Shen W, Sun H. Effects of nuclear receptor FXR on the regulation of liver lipid metabolism in patients with non-alcoholic fatty liver disease. Hepatol Int 2010;4(4):741-748 View Article PubMed/NCBI
  45. Zheng Y, Ying H, Shi J, Li L, Zhao Y. Alanyl-Glutamine Dipeptide Attenuates Non-Alcoholic Fatty Liver Disease Induced by a High-Fat Diet in Mice by Improving Gut Microbiota Dysbiosis. Nutrients 2023;15(18):3988 View Article PubMed/NCBI
  46. Kim YC, Qi M, Dong X, Seok S, Sun H, Kemper B, 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(8):104946 View Article PubMed/NCBI
  47. Huang J, Iqbal J, Saha PK, Liu J, Chan L, Hussain MM, et al. Molecular characterization of the role of orphan receptor small heterodimer partner in development of fatty liver. Hepatology 2007;46(1):147-157 View Article PubMed/NCBI
  48. Watanabe M, Houten SM, Wang L, Moschetta A, Mangelsdorf DJ, Heyman RA, et al. Bile acids lower triglyceride levels via a pathway involving FXR, SHP, and SREBP-1c. J Clin Invest 2004;113(10):1408-1418 View Article PubMed/NCBI
  49. Brendel C, Schoonjans K, Botrugno OA, Treuter E, Auwerx J. The small heterodimer partner interacts with the liver X receptor alpha and represses its transcriptional activity. Mol Endocrinol 2002;16(9):2065-2076 View Article PubMed/NCBI
  50. 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(22):8656-8671 View Article PubMed/NCBI
  51. Lee YK, Park JE, Lee M, Mifflin R, Xu Y, Novak R, et al. Deletion of hepatic small heterodimer partner ameliorates development of nonalcoholic steatohepatitis in mice. J Lipid Res 2023;64(11):100454 View Article PubMed/NCBI
  52. Mifflin R, Park JE, Lee M, Jena PK, Wan YY, Barton HA, et al. Microbial products linked to steatohepatitis are reduced by deletion of nuclear hormone receptor SHP in mice. J Lipid Res 2023;64(12):100469 View Article PubMed/NCBI
  53. Na H, Lee H, Lee MH, Lim HJ, Kim HJ, Jeon Y, et al. Deletion of exons 3 and 4 in the mouse Nr1d1 gene worsens high-fat diet-induced hepatic steatosis. Life Sci 2016;166:13-19 View Article PubMed/NCBI
  54. Han YH, Shin KO, Kim JY, Khadka DB, Kim HJ, Lee YM, et al. A maresin 1/RORα/12-lipoxygenase autoregulatory circuit prevents inflammation and progression of nonalcoholic steatohepatitis. J Clin Invest 2019;129(4):1684-1698 View Article PubMed/NCBI
  55. Robarts DR, Paine-Cabrera D, Kotulkar M, Venneman KK, Gunewardena S, Foquet L, et al. Identifying novel mechanisms of per- and polyfluoroalkyl substance-induced hepatotoxicity using FRG humanized mice. Arch Toxicol 2024;98(9):3063-3075 View Article PubMed/NCBI
  56. 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(2):372-386 View Article PubMed/NCBI
  57. Kim HS, Yoon JS, Jeon Y, Park EJ, Lee JK, Chen S, et al. Ssu72-HNF4α signaling axis classify the transition from steatohepatitis to hepatocellular carcinoma. Cell Death Differ 2022;29(3):600-613 View Article PubMed/NCBI
  58. Lee SH, Veeriah V, Levine F. Liver fat storage is controlled by HNF4α through induction of lipophagy and is reversed by a potent HNF4α agonist. Cell Death Dis 2021;12(6):603 View Article PubMed/NCBI
  59. Xu Y, Zhu Y, Hu S, Xu Y, Stroup D, Pan X, et al. Hepatocyte Nuclear Factor 4α Prevents the Steatosis-to-NASH Progression by Regulating p53 and Bile Acid Signaling (in mice). Hepatology 2021;73(6):2251-2265 View Article PubMed/NCBI
  60. Li Y, Zalzala M, Jadhav K, Xu Y, Kasumov T, Yin L, et al. Carboxylesterase 2 prevents liver steatosis by modulating lipolysis, endoplasmic reticulum stress, and lipogenesis and is regulated by hepatocyte nuclear factor 4 alpha in mice. Hepatology 2016;63(6):1860-1874 View Article PubMed/NCBI
  61. Yang X, Fu Y, Hu F, Luo X, Hu J, Wang G. PIK3R3 regulates PPARα expression to stimulate fatty acid β-oxidation and decrease hepatosteatosis. Exp Mol Med 2018;50(1):e431 View Article PubMed/NCBI
  62. Kasano-Camones CI, Takizawa M, Ohshima N, Saito C, Iwasaki W, Nakagawa Y, et al. PPARα activation partially drives NAFLD development in liver-specific Hnf4a-null mice. J Biochem 2023;173(5):393-411 View Article PubMed/NCBI
  63. Jia N, Lin X, Ma S, Ge S, Mu S, Yang C, et al. Amelioration of hepatic steatosis is associated with modulation of gut microbiota and suppression of hepatic miR-34a in Gynostemma pentaphylla (Thunb.) Makino treated mice. Nutr Metab (Lond) 2018;15:86 View Article PubMed/NCBI
  64. Yu D, Chen G, Pan M, Zhang J, He W, Liu Y, 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(6):4770-4782 View Article PubMed/NCBI
  65. Song Y, Wu W, Sheng L, Jiang B, Li X, Cai K. Chrysin ameliorates hepatic steatosis induced by a diet deficient in methionine and choline by inducing the secretion of hepatocyte nuclear factor 4α-dependent very low-density lipoprotein. J Biochem Mol Toxicol 2020;34(7):e22497 View Article PubMed/NCBI
  66. Crevet L, Vanacker JM. Regulation of the expression of the estrogen related receptors (ERRs). Cell Mol Life Sci 2020;77(22):4573-4579 View Article PubMed/NCBI
  67. Xia H, Dufour CR, Medkour Y, Scholtes C, Chen Y, Guluzian C, et al. Hepatocyte FBXW7-dependent activity of nutrient-sensing nuclear receptors controls systemic energy homeostasis and NASH progression in male mice. Nat Commun 2023;14(1):6982 View Article PubMed/NCBI
  68. B’Chir W, Dufour CR, Ouellet C, Yan M, Tam IS, Andrzejewski S, et al. Divergent Role of Estrogen-Related Receptor α in Lipid- and Fasting-Induced Hepatic Steatosis in Mice. Endocrinology 2018;159(5):2153-2164 View Article PubMed/NCBI
  69. Liu D, Jia H, Holmes DI, Stannard A, Zachary I. Vascular endothelial growth factor-regulated gene expression in endothelial cells: KDR-mediated induction of Egr3 and the related nuclear receptors Nur77, Nurr1, and Nor1. Arterioscler Thromb Vasc Biol 2003;23(11):2002-2007 View Article PubMed/NCBI
  70. Veum VL, Dankel SN, Gjerde J, Nielsen HJ, Solsvik MH, Haugen C, et al. The nuclear receptors NUR77, NURR1 and NOR1 in obesity and during fat loss. Int J Obes (Lond) 2012;36(9):1195-1202 View Article PubMed/NCBI
  71. Yu X, Yang K, Fang Z, Liu C, Hui T, Guo Z, et al. Bioinformatics analysis of potential ferroptosis and non-alcoholic fatty liver disease biomarkers. Gen Physiol Biophys 2024;43(5):371-384 View Article PubMed/NCBI
  72. Luo Z, Yan S, Chao Y, Shen M. Unveiling the mitophagy puzzle in non-alcoholic fatty liver disease (NAFLD): Six hub genes for early diagnosis and immune modulatory roles. Heliyon 2024;10(7):e28935 View Article PubMed/NCBI
  73. Cao Y, Du Y, Jia W, Ding J, Yuan J, Zhang H, et al. Identification of biomarkers for the diagnosis of chronic kidney disease (CKD) with non-alcoholic fatty liver disease (NAFLD) by bioinformatics analysis and machine learning. Front Endocrinol (Lausanne) 2023;14:1125829 View Article PubMed/NCBI
  74. Yu R, Huang Y, Hu X, Chen J. Analysis of machine learning based integration to identify the crosslink between inflammation and immune response in non-alcoholic fatty liver disease through bioinformatic analysis. Heliyon 2024;10(14):e32783 View Article PubMed/NCBI
  75. Nazari E, Khalili-Tanha G, Asadnia A, Pourali G, Maftooh M, Khazaei M, et al. Bioinformatics analysis and machine learning approach applied to the identification of novel key genes involved in non-alcoholic fatty liver disease. Sci Rep 2023;13(1):20489 View Article PubMed/NCBI
  76. Liang H, Xie X, Song X, Huang M, Su T, Chang X, et al. Orphan nuclear receptor NR4A1 suppresses hyperhomocysteinemia-induced hepatic steatosis in vitro and in vivo. FEBS Lett 2019;593(10):1061-1071 View Article PubMed/NCBI
  77. Fayard E, Auwerx J, Schoonjans K. LRH-1: an orphan nuclear receptor involved in development, metabolism and steroidogenesis. Trends Cell Biol 2004;14(5):250-260 View Article PubMed/NCBI
  78. 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(8):166145 View Article PubMed/NCBI
  79. Sahini N, Borlak J. Genomics of human fatty liver disease reveal mechanistically linked lipid droplet-associated gene regulations in bland steatosis and nonalcoholic steatohepatitis. Transl Res 2016;177:41-69 View Article PubMed/NCBI
  80. Sánchez V, Baumann A, Brandt A, Wodak MF, Staltner R, Bergheim I. Oral Supplementation of Phosphatidylcholine Attenuates the Onset of a Diet-Induced Metabolic Dysfunction-Associated Steatohepatitis in Female C57BL/6J Mice. Cell Mol Gastroenterol Hepatol 2024;17(5):785-800 View Article PubMed/NCBI
  81. Lee JM, Lee YK, Mamrosh JL, Busby SA, Griffin PR, Pathak MC, et al. A nuclear-receptor-dependent phosphatidylcholine pathway with antidiabetic effects. Nature 2011;474(7352):506-510 View Article PubMed/NCBI
  82. Lee JS, Bae S, Kang HS, Im SS, Moon YA. Liver receptor homolog-1 regulates mouse superoxide dismutase 2. Biochem Biophys Res Commun 2017;489(3):299-304 View Article PubMed/NCBI
  83. Oiwa A, Kakizawa T, Miyamoto T, Yamashita K, Jiang W, Takeda T, et al. Synergistic regulation of the mouse orphan nuclear receptor SHP gene promoter by CLOCK-BMAL1 and LRH-1. Biochem Biophys Res Commun 2007;353(4):895-901 View Article PubMed/NCBI
  84. Wang S, Sheng F, Zou L, Xiao J, Li P. Hyperoside attenuates non-alcoholic fatty liver disease in rats via cholesterol metabolism and bile acid metabolism. J Adv Res 2021;34:109-122 View Article PubMed/NCBI
  85. Luo L, Zhang H, Chen W, Zheng Z, He Z, Wang H, et al. Angelica sinensis polysaccharide ameliorates nonalcoholic fatty liver disease via restoring estrogen-related receptor α expression in liver. Phytother Res 2023;37(11):5407-5417 View Article PubMed/NCBI
  86. Peng A, Liu S, Fang L, Zhu Z, Zhou Y, Yue S, et al. Inonotus obliquus and its bioactive compounds alleviate non-alcoholic fatty liver disease via regulating FXR/SHP/SREBP-1c axis. Eur J Pharmacol 2022;921:174841 View Article PubMed/NCBI
  87. Mao L, Peng L, Ren X, Chu Y, Nie T, Lin W, et al. Discovery of JND003 as a New Selective Estrogen-Related Receptor α Agonist Alleviating Nonalcoholic Fatty Liver Disease and Insulin Resistance. ACS Bio Med Chem Au 2022;2(3):282-296 View Article PubMed/NCBI
  88. Dhillon S. Dapagliflozin: A Review in Type 2 Diabetes. Drugs 2019;79(10):1135-1146 View Article PubMed/NCBI
  89. Qiao P, Jia Y, Ma A, He J, Shao C, Li X, et al. Dapagliflozin protects against nonalcoholic steatohepatitis in db/db mice. Front Pharmacol 2022;13:934136 View Article PubMed/NCBI

About this Article

Cite this article
Li H, Chen J, Huang Z, Chen M. Orphan Nuclear Receptors in Metabolic Dysfunction-associated Steatotic Liver Disease Development. J Clin Transl Hepatol. Published online: Jun 19, 2025. doi: 10.14218/JCTH.2025.00019.
Copy        Export to RIS        Export to EndNote
Article History
Received Revised Accepted Published
January 9, 2025 May 9, 2025 May 19, 2025 June 19, 2025
DOI http://dx.doi.org/10.14218/JCTH.2025.00019
  • Journal of Clinical and Translational Hepatology
  • pISSN 2225-0719
  • eISSN 2310-8819
Back to Top

Orphan Nuclear Receptors in Metabolic Dysfunction-associated Steatotic Liver Disease Development

Huan Li, Jian Chen, Ziyin Huang, Mingkai Chen
  • Reset Zoom
  • Download TIFF