Elsevier

Pharmacology & Therapeutics

Volume 179, November 2017, Pages 142-157
Pharmacology & Therapeutics

Associate editor: James Hardwick
Targeting nuclear receptors for the treatment of fatty liver disease

https://doi.org/10.1016/j.pharmthera.2017.05.011Get rights and content

Abstract

Ligand-activated nuclear receptors, including peroxisome proliferator-activated receptor alpha (PPARα), pregnane X receptor, and constitutive androstane receptor, were first identified as key regulators of the responses against chemical toxicants. However, numerous studies using mouse disease models and human samples have revealed critical roles for these receptors and others, such as PPARβ/δ, PPARγ, farnesoid X receptor (FXR), and liver X receptor (LXR), in maintaining nutrient/energy homeostasis in part through modulation of the gut-liver-adipose axis. Recently, disorders associated with disrupted nutrient/energy homeostasis, e.g., obesity, metabolic syndrome, and non-alcoholic fatty liver disease (NAFLD), are increasing worldwide. Notably, in NAFLD, a progressive subtype exists, designated as non-alcoholic steatohepatitis (NASH) that is characterized by typical histological features resembling alcoholic steatohepatitis (ASH), and NASH/ASH are recognized as major causes of hepatitis virus-unrelated liver cirrhosis and hepatocellular carcinoma. Since hepatic steatosis is basically caused by an imbalance between fat/energy influx and utilization, abnormal signaling of these nuclear receptors contribute to the pathogenesis of fatty liver disease. Standard therapeutic interventions have not been fully established for fatty liver disease, but some new agents that activate or inhibit nuclear receptor signaling have shown promise as possible therapeutic targets. In this review, we summarize recent findings on the roles of nuclear receptors in fatty liver disease and discuss future perspectives to develop promising pharmacological strategies targeting nuclear receptors for NAFLD/NASH.

Introduction

Liver is the largest solid organ in the body playing a crucial role in maintaining energy homeostasis through metabolism of various nutrients. For example, the main symptoms and signs of acute liver failure are jaundice (impaired bilirubin conjugation/excretion), bleeding tendency (impaired synthesis of coagulation factors), and consciousness disturbance (impaired detoxification of ammonia and other neurotoxic metabolites). Patients having liver cirrhosis often exhibit impaired glucose metabolism (insulin resistance and diabetes), protein/amino acid metabolism (decreased albumin and branched-chain amino acids and increased aromatic amino acids), and lipid metabolism (hypocholesterolemia). These clinical findings mirror a key role of the liver in whole-body metabolism.

Infection with hepatotropic viruses and parasites, autoimmunity, intake of ethanol and certain drugs/medications, and exposure to occupational and environmental toxicants cause liver damage. In Asia, hepatitis B virus (HBV) and hepatitis C virus (HCV) infections are among the main causes for chronic hepatitis, liver cirrhosis, and hepatocellular carcinoma (HCC). Recent advances in antiviral therapies, such as nucleoside analogues for HBV and pegylated interferon/ribavirin and direct-acting antiviral agents for HCV, have improved the quality of life and survival for HBV- or HCV-infected patients. Further improvements in preventive/therapeutic strategies will lead to reduced incidence and mortality of patients having hepatitis virus-related diseases.

Metabolic derangements also cause chronic liver disease, such as fatty liver disease, glycogen storage disease, and hemochromatosis. Fatty liver disease refers to a pathological spectrum ranging from lipid accumulation in hepatocytes (steatosis) to the development of accompanying hepatocyte degeneration (ballooning, Mallory-Denk body) and hepatic inflammation (steatohepatitis), eventually leading to liver fibrosis/cirrhosis, portal hypertension, decompensated liver failure, and HCC (Cohen, Horton, & Hobbs, 2011). Although the mechanism of fatty liver disease may vary according to the etiologies, the liver pathology is often indistinguishable. Therefore, fatty liver diseases are classified according to their etiology/cause, i.e., long-term excess ethanol consumption [alcoholic liver disease/steatohepatitis (ALD/ASH)], overnutrition, visceral obesity, and metabolic syndrome without ethanol intake [non-alcoholic fatty liver disease/steatohepatitis (NAFLD/NASH)], occupational/environmental chemical exposure [toxicant-associated fatty liver disease/steatohepatitis], and others (e.g., drug-induced steatohepatitis and steatohepatitis following gastrointestinal surgery).

The worldwide spread of sedentary lifestyle and diet westernization has increased a prevalence of NAFLD in many countries among wider generations. In Japan, approximately 30% of Japanese upon annual health checkups were found to have NAFLD (Kojima et al., 2003, Eguchi et al., 2012), which extrapolates to an estimated 20 million NAFLD patients nationwide. The prevalence of NAFLD in junior high school students was also estimated as approximately 4% in certain areas of Japan (Tsuruta et al., 2010). NAFLD is associated with obesity, insulin resistance, diabetes, hypertension, dyslipidemia, atherosclerosis, and systemic inflammation, representing hepatic manifestation of metabolic syndrome. Although it remains controversial whether NAFLD is a cause or a result of glucose intolerance and insulin resistance, a prospective study demonstrated higher risk of diabetes and cardiovascular events in non-diabetic humans with NAFLD compared with those without NAFLD (Heianza et al., 2014). Additionally, liver with significant steatosis is more susceptible for hepatotoxicants and retards/impairs regeneration following partial hepatectomy (Kele et al., 2013). Therefore, NAFLD is considered as a detrimental condition necessitating appropriate therapeutic interventions.

Dr. Ludwig, a pathologist in Mayo Clinic, proposed a term non-alcoholic steatohepatitis (NASH) in 1980 (Ludwig, Viggiano, McGill, & Oh, 1980). He described 20 non-alcoholic patients having histological findings as compatible with ASH, such as fatty changes, focal necrosis, ballooned hepatocytes with Mallory-Denk bodies, lobular inflammation, and perisinusoidal/perivenular fibrosis. Clinically, most of these patients were obese and 25% had diabetes. At present, NAFLD is classified into two categories according to liver pathology: non-alcoholic fatty liver (NAFL, previously designated as simple steatosis) and NASH. NASH is defined by the presence of hepatocyte ballooning, lobular inflammation, and/or fibrosis in addition to macrovesicular steatosis, and NAFL is characterized as macrovesicular steatosis without ballooned hepatocytes (Hashimoto, Tokushige, & Ludwig, 2015). This pathology-based classification stems from the concept that NASH can progress into advanced liver fibrosis and the prognosis is poorer than that of NAFL and exhibited the clinical outcome different from NAFL. Indeed, in our NASH cases with obesity, diabetes, hypertension, and dyslipidemia, ballooned hepatocytes were detected in the initial biopsied samples, in which liver fibrosis apparently progressed in 5 years (Fig. 1). Matteoni et al. identified that the outcomes of cirrhosis and liver-related death were more frequent in NAFLD patients with ballooned hepatocytes than in those without ballooned hepatocytes (Matteoni et al., 1999). Others reported that the survival of NASH patients, but not NAFL patients, was significantly lower than an age- and sex-matched reference population (Ekstedt et al., 2006). Based on these findings, the notion that NASH is a serious and progressive type of NAFLD has generally been accepted. The diagnosis of NASH and evaluation of histological severity of NAFLD are performed by the pathological findings of the liver, but liver biopsy is somewhat invasive and costly. Additionally, sampling errors and differences in diagnostic accuracy between independent pathologists can sometimes be problematic. Therefore, less invasive and more accurate strategies to discriminate between NAFL and NASH and predict actual steatosis/inflammation/fibrosis instead of liver biopsy have been evaluated (Fujimori et al., 2016, Hatta et al., 2010, Kitabatake et al., 2017, Matsubara et al., 2012, Tanaka et al., 2006, Tanaka et al., 2006, Tanaka et al., 2012, Tsutsui et al., 2010). Recent studies demonstrated that the presence of fibrosis, but not hepatocyte ballooning, was a determinant of poor prognosis in NAFLD patients (Angulo et al., 2015, Loomba and Chalasani, 2015). Indeed, such a case of NAFLD with careful 27-year follow-up was examined (Nagaya et al., 2008). This patient was diagnosed as having NAFL at the first liver biopsy but gradually developed into cirrhosis and HCC over 20 years. This case teaches us that NAFL is not always benign. Additionally, HCC may occur from NAFL regardless of the absence of advanced fibrosis, past HBV infection, and regular ethanol consumption (Kimura et al., 2017). Although key factors affecting clinical course and outcome of NAFLD and methods to predict fibrosis progression and HCC development have not been identified, attenuating steatosis, hepatic injury, and inflammation and inhibiting fibrosis progression are promising strategies to improve the prognosis of NAFLD/NASH patients.

Understanding NAFLD/NASH pathogenesis is mandatory for developing novel therapeutic intervention strategies. Insulin resistance and diabetes were reportedly associated with NAFLD with more advanced fibrosis, while impaired glucose metabolism and insulin signaling aggravated liver fibrogenesis and NAFLD, in turn, worsening diabetes and driving systemic inflammation. A ‘two-hit’ model has been proposed to explain why some, but not all, individuals with steatosis develop steatohepatitis (Day & James, 1998). Besides diabetes/insulin resistance, many other second-hit mechanisms, such as lipotoxicity due to saturated fatty acid (FA), free cholesterol, and ceramide, endotoxins from gut and gum, oxidative stress, mitochondrial dysfunction, endoplasmic reticulum stress, and iron overload, have been extensively reviewed. Additionally, contribution of other organs to the development of NAFLD/NASH should be considered (Jiang et al., 2015, Jiang et al., 2015, Tanaka et al., 2014). Recently, a term ‘multiple-hit theory’ has been widely accepted because of close interconnections among these hits (Buzzetti, Pinzani, & Tsochatzis, 2016).

More recently, the adverse outcome pathway framework has been used to contextualize the role of nuclear receptors in hepatosteatosis (Mellor et al., 2016, Willett et al., 2014). In humans, there are 48 nuclear receptors categorized into 7 subfamilies designated as NR0-NR6 (Evans & Mangelsdorf, 2014) (Table 1). Nuclear receptors are ligand-activated transcription factors regulating the expression of several genes through direct modulation of the transcriptional activities and epigenetic changes. Notably, nuclear receptors in the NR1 subfamily are associated with energy/nutrient control, which may play an important role in the pathogenesis of fatty liver disease. These NR1 nuclear receptors are NR1C1–3: peroxisome proliferator-activated receptor (PPAR) α, β/δ, and γ, NR1H2–3: liver X receptor (LXR) α and β, NR1H4: farnesoid X receptor (FXR), NR1I2: pregnane X receptor (PXR), and NR1I3: constitutive androstane receptor (CAR) (Table 1). These nuclear receptors are mainly activated by binding with ligands, form a heterodimer with retinoid X receptor (RXR) α, β, and γ (NR2B1–3), and exhibit their function as transcription factors. The ligands of NR1 subfamily include nuclear pore-permeable lipophilic endogenous substances mainly derived from nutrients [e.g., FAs, eicosanoids, oxysterols, and bile acids (BAs)] and exogenous chemicals. When the ligands are unbound, the activity of NR1 receptors is suppressed by binding to co-repressors. When either NR1 nuclear receptor or its heterodimeric partner RXR is liganded, these receptors release co-repressors and recruit co-activators. However, the transcriptional signal is amplified when both heterodimeric receptors are liganded. The boosting effect of liganded RXR allows these NR1 receptors to significantly increase their transcriptional activities. Not only the ligands of RXR, but also the presence of RXR itself are important for the function of NR1 receptors. Indeed, loss of hepatocyte-specific RXRα disrupts the basal functions of NR1 receptors and alters nutrient metabolism (Anderson et al., 2004).

To understand the physiological role of NR1 nuclear receptors, the concept of “energy vector” was proposed (Evans & Mangelsdorf, 2014; Fig. 2). In the fasting state, triacylglycerol (TAG) stored in white adipose tissue (WAT) is subjected to lipolysis and released into the circulation as FAs. FAs are used in many organs as an energy source. In the liver, FAs activate PPARα and enhance FA catabolism, resulting in the production of ATP, ketone bodies, and hepatokine fibroblast growth factor (FGF) 21. Ketone bodies are consumed as an energy source in the brain and FGF21 serves as a stress messenger to prepare other organs for energy deprivation. In the fed state, energy flux is reversed and FXR, LXR, PPARβ/δ and PPARγ are mainly involved in nutrient absorption from the gut and distribution from gut/liver to peripheral tissues, such as WAT and muscle. After meals, BAs activate intestinal FXR, promoting nutrient absorption and maintaining a barrier to the gut microbiome. Absorbed dietary lipids are transported into the circulation as chylomicron and its remnant. Hepatic FXR promotes post-prandial TAG-rich lipoprotein clearance. Excess cholesterol is removed from the body by reverse cholesterol transport under the control of the FXR-stimulated enterokine FGF19 (FGF15 in rodents) and/or activation of hepatic LXR by oxysterols. Fecal elimination of cholesterol is the last step in the reverse cholesterol transport pathway. FGF19 increases hydrophilicity of the bile salt pool and stimulates transintestinal cholesterol excretion (de Boer et al., 2017). FGF15/19 also attenuates post-prandial hyperglycemia through enhancing hepatic glycogenesis. Consequently, excess nutrients are either consumed in muscle or stored in WAT due to PPARβ/δ and PPARγ, respectively. Post-prandial hepatic activation of PXR and CAR promotes the clearance of toxic dietary metabolites and xenobiotics. Because abnormal energy/nutrient homeostasis is a major cause of NAFLD/NASH, the concept of “dysfunction of energy vectors on the gut-liver-adipose axis” may represent a mechanism on how dysregulated nuclear receptors contributes to NAFLD/NASH development (Fig. 2).

The following sections describe the roles of nuclear receptors, mainly focusing on NR1 receptors, in liver pathophysiology and possible therapeutic strategies for the prevention and treatment of fatty liver disease through targeting these key receptors.

Section snippets

PPAR overview

Since the administration of certain chemicals, such as Wy-14643, nafenopin, and fibrate derivatives, to mice induces hepatic peroxisome proliferation (increased peroxisome number and size) and hepatomegaly, these chemicals are called as peroxisome proliferators (PP) (Reddy & Krishnakantha, 1975). Furthermore, long-term PP administration results in HCC without accompanying hepatic fibrosis/inflammation (Reddy, Azarnoff, & Hignite, 1980). Since no apparent genetic mutations have been detected in

Hepatic steatosis and PPARβ/δ

PPARβ/δ is highly expressed in muscle, skin, adipose tissue, and liver. PPARβ/δ levels are far higher in muscle than the other two forms of PPAR in rodents and humans and its activation during the fed state or exercise increases fuel consumption in muscle mainly via enhancement of β-oxidation (Manickam & Wahli, 2017). In obese monkeys, the treatment with PPARβ/δ agonist GW501516 normalizes serum insulin and TAG concentrations, increases high-density lipoprotein (HDL) cholesterol, and decreases

Hepatic steatosis and PPARγ

PPARγ is highly expressed in adipose tissue and macrophage where it has an important role in energy storage and immune modulation, respectively (Ahmadian et al., 2013). PPARγ expression in hepatocytes is relatively low, but is increased in human steatotic livers and mouse NAFLD/NASH livers. Forced expression of PPARγ in hepatocytes by adenovirus induced hepatosteatosis, and hepatocyte-specific disruption of PPARγ in ob/ob mice attenuated fatty liver (Matsusue et al., 2003, Yu et al., 2003).

LXR overview

There are two LXR isoforms in humans, LXRα (NR1H3) and LXRβ (NR1H2). LXR activates hepatic TAG synthesis and export to peripheral tissues, and stimulates reverse cholesterol transport to moderate cholesterol toxicity in extrahepatic tissues (Hong & Tontonoz, 2014). LXRα is highly expressed in the liver, intestine, kidney and adipose tissue, while LXRβ is ubiquitously expressed (Shinar et al., 1994, Willy et al., 1995).

LXRs bind to DR4, direct repeats of the core sequence AGGTCA spaced by four

FXR overview

FXR was identified as a nuclear receptor activated by farnesol pyrophosphate in 1995 (Forman et al., 1995). FXR exists as two variants in humans, FXRα (NR1H4) and FXRβ (NR1H5), while the latter is a pseudogene (Evans and Mangelsdorf, 2014, Zhang et al., 2008). FXR is abundantly expressed in the organs involved in BA metabolism and transport, such as liver, intestine, and kidney, but also is present in adipose tissue and adrenal gland. Upon ligand activation, FXR binds to the transcriptional

PXR

PXR (NR1I2) is a xenobiotic-sensing nuclear receptor that regulates drug metabolism and detoxification. PXR is abundant in the liver and gut, and is expressed not only in hepatocytes, but also in HSC and Kupffer cells (Banerjee, Robbins, & Chen, 2015). PXR recognizes DR3 and DR4 sequences as well as everted repeats separated by 6 or 8 base pairs (Kliewer, Goodwin, & Willson, 2002). A typical target gene of PXR is cytochrome P450 (CYP) 3A4, but the spectrum of PXR targets has expanded to genes

Future directions of fatty liver disease treatment targeting nuclear receptors

Pharmacological activation of NR1 is expected to attenuate hepatic steatosis, inflammation, fibrosis, insulin resistance, dyslipidemia and obesity. However, some nuclear receptor agonists exhibited insufficient or paradoxical effects. For example, PPARα activation is basically beneficial for fatty liver disease, but the effects of fibrates for human NAFLD are limited. PPARγ activation improves adipocyte function, but enhances hepatocyte steatosis. Such unexpected findings may be derived from

Conclusion

We reviewed the role of NR1 for the development of fatty liver disease. Nuclear receptor dysregulation contributes to the pathogenesis of NAFLD/NASH by impacting the integrated control of energy/nutrient metabolism thorough the gut-liver-adipose axis and inflammatory signaling. Nuclear receptor-targeted therapies may be beneficial for fatty liver disease, but the effectiveness is still unsatisfactory. Novel pharmacological interventions, such as dual/triple agonists, combination of

Conflict of interest statement

The authors declare that there are no conflicts of interest.

Acknowledgment

The authors thank the following collaborators for a lot of helps, advice, instruction, and encouragement for the studies of fatty liver disease: Dr. Michiharu Komatsu, Dr. Tadanobu Nagaya, Dr. Takefumi Kimura, Dr. Naoyuki Fujimori, Dr. Ayumi Sugiura, Dr. Kenji Sano, Dr. Takero Nakajima, Dr. Xiao Hu, Dr. Xiaojing Wang, Dr. Wataru Okiyama, Dr. Goro Tsuruta, Dr. Kan Nakagawa, Dr. Hiroyuki Kitabatake, Prof. Masahide Yazaki, Dr. Yasunari Fujinaga, Dr. Akira Kobayashi, and Prof. Eiji Tanaka (Shinshu

References (217)

  • P. Delerive et al.

    Peroxisome proliferator-activated receptor alpha negatively regulates the vascular inflammatory gene response by negative cross-talk with transcription factors NF-kappaB and AP-1

    The Journal of Biological Chemistry

    (1999)
  • R.M. Evans et al.

    Nuclear receptors, RXR, and the big bang

    Cell

    (2014)
  • A. Fernández-Alvarez et al.

    Human SREBP1c expression in liver is directly regulated by peroxisome proliferator-activated receptor alpha (PPARalpha)

    The Journal of Biological Chemistry

    (2011)
  • C. Fernández-Miranda et al.

    A pilot trial of fenofibrate for the treatment of non-alcoholic fatty liver disease

    Digestive and Liver Disease

    (2008)
  • B.M. Forman et al.

    Identification of a nuclear receptor that is activated by farnesol metabolites

    Cell

    (1995)
  • S. Francque et al.

    PPARα gene expression correlates with severity and histological treatment response in patients with non-alcoholic steatohepatitis

    Journal of Hepatology

    (2015)
  • G. Gaich et al.

    The effects of LY2405319, an FGF21 analog, in obese human subjects with type 2 diabetes

    Cell Metabolism

    (2013)
  • J. Gao et al.

    The constitutive androstane receptor is an anti-obesity nuclear receptor that improves insulin sensitivity

    The Journal of Biological Chemistry

    (2009)
  • S. Gill et al.

    Sterol regulators of cholesterol homeostasis and beyond: The oxysterol hypothesis revisited and revised

    Progress in Lipid Research

    (2008)
  • F.J. Gonzalez et al.

    An intestinal microbiota-farnesoid X receptor axis modulates metabolic disease

    Gastroenterology

    (2016)
  • F.J. Gonzalez et al.

    PPARalpha: Mechanism of species differences and hepatocarcinogenesis of peroxisome proliferators

    Toxicology

    (2008)
  • M. Goudarzi et al.

    PPARβ/δ modulates ethanol-induced hepatic effects by decreasing pyridoxal kinase activity

    Toxicology

    (2013)
  • K. Griffett et al.

    The LXR inverse agonist SR9238 suppresses fibrosis in a model of non-alcoholic steatohepatitis

    Molecular Metabolism

    (2015)
  • S.C. Hasenfuss et al.

    Regulation of steatohepatitis and PPARγ signaling by distinct AP-1 dimers

    Cell Metabolism

    (2014)
  • T. Hashimoto et al.

    Defect in peroxisome proliferator-activated receptor alpha-inducible fatty acid oxidation determines the severity of hepatic steatosis in response to fasting

    The Journal of Biological Chemistry

    (2000)
  • X. Hu et al.

    PPARα protects against trans-fatty-acid-containing diet-induced steatohepatitis

    The Journal of Nutritional Biochemistry

    (2017)
  • N. Ijssennagger et al.

    Gene expression profiling in human precision cut liver slices in response to the FXR agonist obeticholic acid

    Journal of Hepatology

    (2016)
  • T. Inagaki et al.

    Endocrine regulation of the fasting response by PPARalpha-mediated induction of fibroblast growth factor 21

    Cell Metabolism

    (2007)
  • Y. Kamada et al.

    Hypoadiponectinemia accelerates hepatic tumor formation in a nonalcoholic steatohepatitis mouse model

    Journal of Hepatology

    (2007)
  • M.A. Kennedy et al.

    Characterization of the human ABCG1 gene: Liver X receptor activates an internal promoter that produces a novel transcript encoding an alternative form of the protein

    The Journal of Biological Chemistry

    (2001)
  • N.M. Kettner et al.

    Circadian homeostasis of liver metabolism suppresses hepatocarcinogenesis

    Cancer Cell

    (2016)
  • M. Komatsu et al.

    Steatogenesis in adult-onset type II citrullinemia is associated with down-regulation of PPARα

    Biochimica et Biophysica Acta

    (2015)
  • M. Komatsu et al.

    Citrin deficiency as a cause of chronic liver disorder mimicking non-alcoholic fatty liver disease

    Journal of Hepatology

    (2008)
  • J. Laurin et al.

    Ursodeoxycholic acid or clofibrate in the treatment of non-alcohol-induced steatohepatitis: A pilot study

    Hepatology

    (1996)
  • M. Ahmadian et al.

    PPARγ signaling and metabolism: The good, the bad and the future

    Nature Medicine

    (2013)
  • S.B. Ahn et al.

    Expression of liver X receptor correlates with intrahepatic inflammation and fibrosis in patients with nonalcoholic fatty liver disease

    Digestive Diseases and Sciences

    (2014)
  • S.P. Anderson et al.

    Overlapping transcriptional programs regulated by the nuclear receptors peroxisome proliferator-activated receptor alpha, retinoid X receptor, and liver X receptor in mouse liver

    Molecular Pharmacology

    (2004)
  • P. Angulo et al.

    Liver fibrosis, but no other histologic features, is associated with long-term outcomes of patients with nonalcoholic fatty liver disease

    Gastroenterology

    (2015)
  • M.A. Bae et al.

    Selective inhibition of activated stellate cells and protection from carbon tetrachloride-induced liver injury in rats by a new PPARgamma agonist KR62776

    Archives of Pharmacal Research

    (2010)
  • E. Barroso et al.

    The PPARβ/δ activator GW501516 prevents the down-regulation of AMPK caused by a high-fat diet in liver and amplifies the PGC-1α-Lipin 1-PPARα pathway leading to increased fatty acid oxidation

    Endocrinology

    (2011)
  • L.D. Beilke et al.

    Constitutive androstane receptor-mediated changes in bile acid composition contributes to hepatoprotection from lithocholic acid-induced liver injury in mice

    Drug Metabolism and Disposition

    (2009)
  • A. Bitter et al.

    Pregnane X receptor activation and silencing promote steatosis of human hepatic cells by distinct lipogenic mechanisms

    Archives of Toxicology

    (2015)
  • J.F. de Boer et al.

    Intestinal farnesoid X receptor controls transintestinal cholesterol excretion in mice

    Gastroenterology

    (2017)
  • D.R. Boverhof et al.

    Transgenic animal models in toxicology: Historical perspectives and future outlook

    Toxicological Sciences

    (2011)
  • C.N. Brocker et al.

    Hepatocyte-specific PPARA expression exclusively promotes agonist-induced cell proliferation without influence from non-parenchymal cells

    American Journal of Physiology. Gastrointestinal and Liver Physiology

    (2017)
  • L. Chen et al.

    Oleoylethanolamide, an endogenous PPAR-α ligand, attenuates liver fibrosis targeting hepatic stellate cells

    Oncotarget

    (2015)
  • J. Cheng et al.

    Chronic exposure to rifaximin causes hepatic steatosis in pregnane X receptor-humanized mice

    Toxicological Sciences

    (2012)
  • J. Cheng et al.

    Therapeutic role of rifaximin in inflammatory bowel disease: Clinical implication of human pregnane X receptor activation

    The Journal of Pharmacology and Experimental Therapeutics

    (2010)
  • J.C. Cohen et al.

    Human fatty liver disease: Old questions and new insights

    Science

    (2011)
  • J.C. Corton et al.

    Mode of action framework analysis for receptor-mediated toxicity: The peroxisome proliferator-activated receptor alpha (PPARα) as a case study

    Critical Reviews in Toxicology

    (2014)
  • Cited by (168)

    View all citing articles on Scopus
    View full text