Full length article
Hepatoprotective effects of ZLY16, a dual peroxisome proliferator-activated receptor α/δ agonist, in rodent model of nonalcoholic steatohepatitis

https://doi.org/10.1016/j.ejphar.2020.173300Get rights and content

Highlights

  • ZLY16 was identified by cyclization strategy based on GFT505.

  • ZLY16 is a novel PPARα/δ agonist with relative higher potency on PPARγ.

  • ZLY16 alleviates fatty liver in MCD-induced db/db mice.

  • ZLY16 inhibited liver inflammation and fibrosis by regulating related gene expression.

  • ZLY16 revealed greater improvement on liver lipid accumulation, fibrosis and inflammation than GFT505.

Abstract

Nonalcoholic fatty liver disease (NAFLD), a chronic progressive liver disease, covers a series of liver damage encompassing steatosis, nonalcoholic steatohepatitis (NASH), fibrosis and cirrhosis. However, there are no approved therapies for NAFLD. Herein, we characterize the pharmacological profile of ZLY16 ((E)-2-(4-(3-(2,3-dihydrobenzo[b]thiophen -5-yl)-3-oxoprop-1-en-1-yl)-2,6-dimethylphenoxy)-2-methylpropanoic acid), a novel highly potent PPARα/δ agonist with relative higher potency on PPARγ. The chronic effects of ZLY16 on NASH development were evaluated in MCD-induced db/db mice. ZLY16 revealed decreased liver injury biomarkers, hepatic steatosis, inflammation, ballooning, and oxidative stress. Further mechanism researches suggested that ZLY16 inhibited liver inflammation and fibrosis by regulating gene expression including COLIA1, TIMP, TGFβ, TNFα, and IL6. Moreover, ZLY16 offers more favorable effects in decreasing liver TC and TG accumulation, blocking liver fibrosis and inflammation than GFT505, the most advanced candidate of PPARα/δ agonist for the treatment of NASH. These results indicate that ZLY16 is a highly potent PPARα/δ agonist that provides great protection against NASH development, and may be useful for the treatment of NAFLD/NASH.

Graphical abstract

In NASH model, the long-term administration of PPARα/δ dual agonist ZLY16 has multiple benefits on improving liver lipid profiles, hepatic steatosis, inflammation, ballooning and oxidative stress.

Image 1
  1. Download : Download high-res image (99KB)
  2. Download : Download full-size image

Introduction

Nonalcoholic fatty liver disease (NAFLD) is regarded as the manifestations of hepatic disorders, ranging from simple hepatocellular steatosis through nonalcoholic steatohepatitis (NASH) to fibrosis, and irreversible cirrhosis (Chatterjee and Mitra, 2015; Younossi et al., 2015, 2016). With the prevalence of type 2 diabetes and obesity, NAFLD is now the most common liver metabolic disease in Western countries(Cheung and Sanyal, 2010). Although the incidence of NAFLD is rapidly growing, yet there are no approved pharmacotherapies for this disease (Younossi et al., 2018). Hence, efficacious therapeutic agents for the treatment of NAFLD are striking.

Peroxisome proliferator-activator receptors (PPARs) belong to the nuclear hormone receptor superfamily (Gross et al., 2017; Hong et al., 2019; Zhao et al., 2015). PPARs play crucial roles in the regulation of numerous physiological processes, including immune response, energy homeostasis, cell proliferation, migration and differentiation(Magadum and Engel, 2018). The PPAR family is composed of three primary isotypes: PPARα, PPARβ/δ and PPARγ—that share high levels of sequence homology but have various tissue distributions and ligand selectivity(Poulsen et al., 2012). PPARα, the target of anti-hyperlipidemic fibrates, is expressed high levels in the adipose tissue, heart, skeletal muscle, kidney and liver(Brown and Plutzky, 2007). In various animal models, the absence of PPARα in hepatocytes is linked with the aggravation hepatic steatosis(Kersten et al., 1999; Montagner et al., 2016). PPARδ, another member of the PPAR family, exhibits high expression levels not only in liver but also in skeletal muscle and macrophages(Bojic and Huff, 2013), and its activation ameliorates insulin-sensitivity, inhibits glucose production in the liver, decreases macrophage and Kupffer cell activation and increases fatty acid oxidation (Adhikary et al., 2015; Sprecher et al., 2007; Tanaka et al., 2003). In previous studies, we have identified several PPARδ agonists with high potential on the improvement of energy metabolism (Li et al., 2018, 2019a, 2019b, 2020). The agonist of PPARγ (rosiglitazone) has been widely used to the treatment of metabolic disease, though excessive activation of PPARγ may cause side effects (Ahmadian et al., 2013; Monsalve et al., 2013; Verges, 2004).

Based on the well-known functions of PPARα and PPARδ, the dual PPARα/δ agonist might have the potential to improve the complex pathogenesis of NASH. Elafibranor (GFT-505), a novel PPARα/δ agonist, aims to synergize the beneficial impact of PPARα and PPARδ agonist. Studies from human and animal studies suggest that GFT505 alleviates fatty liver with additional benefit on plasma lipid levels(Cariou et al., 2011; Hanf et al., 2014; Staels et al., 2013). The peripheral and hepatic insulin sensitivity was also improved by the administration of GFT505. Besides, GFT505 improves liver enzymes levels and decreases inflammation markers. Taken together, PPARα/δ was considered as attractive targets for the treatment of NAFLD.

Interestingly, the cyclization of GFT505 provided ZLY16, a potent PPARα/δ dual agonist with relative higher potency on PPARγ, which might be more favorable to the treatment of NASH while avoiding side effect induced by excessive activation of PPARγ. To explore whether the novel dual PPARα/δ agonist ZLY16 could improve NASH, we evaluated the long-term effects of ZLY16 on NASH in MCD diet-induced db/db mice.

Section snippets

Chemistry

All commercial available materials and reagents were used without purification unless otherwise indicated. Purification by column chromatography was carried out using silica gel (200–300 mesh). The NMR spectra (300 MHz for 1 H NMR) were detected on Bruker ACF-300Q instrument. Chemical shifts are showed as values relative to the internal standard (tetramethylsilane), and coupling constants (J values) were given in hertz (Hz).

1-(2,3-dihydrobenzo[b]thiophen-5-yl)ethan-1-one (2a). Dissolution of

Chemistry

ZLY16 was synthesized according to the synthetic route depicted in Scheme 1, purity >98%. The key intermediate 2a was prepared by Friedel-Crafts acylation of commercially available 1a and acetyl chloride. The commercially starting material 3a was treated with methyl 2-bromo-2-methylpropanoate to afford the intermediates 4a, which was further converted to target compounds ZLY16 by aldol condensation with 2a. Purification of reaction products was carried out by column chromatography using

Discussion

NAFLD, an emerging metabolic-related disease defined features as excess hepatic lipid accumulation, is becoming one of the most common liver metabolic diseases which is closely related to obesity. However, there is no convincing pharmacological treatment for NAFLD. Thus, there is a pressing need for studies on potential therapeutic interventions of NAFLD. Recently, PPARs have drawn interest as potent targets for the treatment of NAFLD. PPARα is highly expressed in liver and plays a dominant

Conclusion

In conclusion, the present study reported a novel potent PPARα/δ modulator named ZLY16 which provides powerful protection against MCD-induced fatty liver hepatitis in db/db mice. The chronic study demonstrated that ZLY16 could exert equivalent functions as GFT505 in decreasing liver injury biomarkers, improving NASH features such as steatosis, inflammation, ballooning and reducing liver oxidative stress. In addition, ZLY16 offers more favorable effects in decreasing liver TC and TG

CRediT authorship contribution statement

Zongtao Zhou: Writing - original draft. Liming Deng: Project administration. Lijun Hu: Project administration. Qiang Ren: Software, Resources. Zongyu Cai: Software, Resources. Bin Wang: Software, Resources. Zheng Li: Funding acquisition, Writing - review & editing. Luyong Zhang: Funding acquisition, Writing - review & editing.

Declaration of competing interest

The authors have no conflicts of interest to declare.

Acknowledgments

This study was supported by the Guangdong Basic and Applied Basic Research Foundation (Grant 2019A1515011036), Key Field R&D Plan Project of Guangdong province (No. 2019B020201002), the National Natural Science Foundation of China (Grant 81803341), the Natural Science Foundation of Guangdong Province, China (Grant 2018A030313445), the Innovative strong school project of Guangdong Pharmaceutical University (Grant 2018KTSCX111), the projects of Guangzhou key laboratory of construction and

References (56)

  • Z. Li et al.

    A novel FFA1 agonist, CPU025, improves glucose-lipid metabolism and alleviates fatty liver in obese-diabetic (ob/ob) mice

    Pharmacol. Res.

    (2020)
  • J.I. Odegaard et al.

    Alternative M2 activation of Kupffer cells by PPARdelta ameliorates obesity-induced insulin resistance

    Cell Metabol.

    (2008)
  • L. Poulsen et al.

    PPARs: fatty acid sensors controlling metabolism

    Semin. Cell Dev. Biol.

    (2012)
  • M.E. Rinella et al.

    Mechanisms of hepatic steatosis in mice fed a lipogenic methionine choline-deficient diet

    J. Lipid Res.

    (2008)
  • A.P. Rolo et al.

    Role of oxidative stress in the pathogenesis of nonalcoholic steatohepatitis

    Free Radic. Biol. Med.

    (2012)
  • M. Schieber et al.

    ROS function in redox signaling and oxidative stress

    Curr. Biol.

    (2014)
  • S. Seki et al.

    In situ detection of lipid peroxidation and oxidative DNA damage in non-alcoholic fatty liver diseases

    J. Hepatol.

    (2002)
  • R. Shiri-Sverdlov et al.

    Early diet-induced non-alcoholic steatohepatitis in APOE2 knock-in mice and its prevention by fibrates

    J. Hepatol.

    (2006)
  • B. Verges

    Clinical interest of PPARs ligands - particular benefit in type 2 diabetes and metabolic syndrome

    Diabetes Metab.

    (2004)
  • T. Adhikary et al.

    The transcriptional PPARbeta/delta network in human macrophages defines a unique agonist-induced activation state

    Nucleic Acids Res.

    (2015)
  • M. Ahmadian et al.

    PPAR gamma signaling and metabolism: the good, the bad and the future

    Nat. Med.

    (2013)
  • L.A. Bojic et al.

    Peroxisome proliferator-activated receptor delta: a multifaceted metabolic player

    Curr. Opin. Lipidol.

    (2013)
  • J.D. Brown et al.

    Peroxisome proliferator-activated receptors as transcriptional nodal points and therapeutic targets

    Circulation

    (2007)
  • B. Cariou et al.

    Effects of the new dual PPAR alpha/delta agonist GFT505 on lipid and glucose homeostasis in abdominally obese patients with combined dyslipidemia or impaired glucose metabolism

    Diabetes Care

    (2011)
  • O. Cheung et al.

    Recent advances in nonalcoholic fatty liver disease

    Curr. Opin. Gastroenterol.

    (2010)
  • D. Dibra et al.

    Mutant p53 in concert with an interleukin-27 receptor alpha deficiency causes spontaneous liver inflammation, fibrosis, and steatosis in mice

    Hepatology

    (2016)
  • G.C. Farrell et al.

    Nonalcoholic fatty liver disease: from steatosis to cirrhosis

    Hepatology

    (2006)
  • A.M. Gressner et al.

    Roles of TGF-beta in hepatic fibrosis

    Front. Biosci.

    (2002)
  • Cited by (11)

    • Privileged scaffolds in anti-diabetic drug discovery

      2023, Privileged Scaffolds in Drug Discovery
    • Trilobatin alleviates non-alcoholic fatty liver disease in high-fat diet plus streptozotocin-induced diabetic mice by suppressing NLRP3 inflammasome activation

      2022, European Journal of Pharmacology
      Citation Excerpt :

      Liver fragments were fixed in 10% formalin for 48h, dehydrated, embedded in paraffin, sectioned (4 μm thick), then stained with H&E and Sirius Red according to standard protocols, respectively. The NAFLD activity score (NAS) system including steatosis (0–3), lobular inflammation (0–3) and hepatocellular ballooning (0–3) was used to assess histological liver changes (Zhou et al., 2020). For the staining of Oil red O, liver tissues were embedded in optimal cutting temperature medium, sectioned, stained with Oil red and five views from each section were randomly selected to evaluate hepatic steatosis.

    • Discovery of new and highly effective quadruple FFA1 and PPARα/γ/δ agonists as potential anti-fatty liver agents

      2022, European Journal of Medicinal Chemistry
      Citation Excerpt :

      Hepatic inflammation acts as an important bridge between the initial metabolic load and subsequent hepatocyte death as well as trigger of fibrogenesis in NASH [47]. The activation of PPARδ exerts beneficial effects on inflammation and energy metabolism [41,48–50]. Interestingly, ZLY18 markedly diminished the inflammation-related genes expression, such as tumor necrosis factor alpha (TNFα) and interleukin-6 (IL-6).

    • Discovery of the first-in-class dual PPARδ/γ partial agonist for the treatment of metabolic syndrome

      2021, European Journal of Medicinal Chemistry
      Citation Excerpt :

      For C23H22O6: C, 70.04; H, 5.62; Found: C, 70.22; H, 5.43. The cell-based assays of PPARα, PPARγ and PPARδ were performed according to our previous reports [18–21]. After transfection for 24 h, test compounds were added and incubated for 18 h.

    • Design, synthesis and structure-activity relationship studies of novel partial FXR agonists for the treatment of fatty liver

      2020, Bioorganic Chemistry
      Citation Excerpt :

      Therefore, it is urgent demand to develop effective drugs for the treatment of NAFLD. Currently, a lot of researches come from institutes and companies have focused on this field, including farnesoid X receptor (FXR) agonist and peroxisome proliferators-activated receptors agonist [8–12]. FXR, a sensor of chenodeoxycholic acid, regulates the synthesis and transport of bile acid [13,14].

    View all citing articles on Scopus
    1

    These authors made equal contributions to this work.

    View full text