Abstract
Recent developments in understanding the role of bile acids (BAs) as signalling molecules in human metabolism and inflammation have opened new avenues in the field of hepatology research. BAs are no longer considered as simple molecules helping in fat digestion but as agents with real therapeutic value in treating complex autoimmune and metabolic liver diseases. BAs and their receptors such as farnesoid X receptor, transmembrane G protein-coupled receptor 5 and peroxisome proliferator-activated receptor have been identified as novel targets for drug development. Some of these novel pharmaceuticals are already in clinical evaluation with the most advanced drugs having reached phase III trials. Chronic liver diseases such as primary biliary cholangitis, primary sclerosing cholangitis and nonalcoholic fatty liver disease, for which there is no or limited pharmacotherapy, are most likely to gain from these developments. In this review we discuss recent and the most relevant basic and clinical research findings related to BAs and their implications for novel therapy for chronic liver diseases.
Keywords: bile acids, drug therapy, liver disease, nonalcoholic fatty liver disease, primary biliary cholangitis, primary sclerosing cholangitis
Introduction
The incidence and prevalence of liver diseases in general is rising and significantly contributing to the increasing burden on health care [Williams and Horton, 2013]. To deal with the challenge of treating patients with liver diseases, clinicians need effective therapies that target the underlying disease pathology as well as symptoms and complications associated with the disease. Whilst great success has been achieved in the last few years in the pharmacological treatment of viral hepatitis [Lam et al. 2015], the need for effective drug therapies in metabolic and cholestatic liver diseases has been only partially met. As obesity is becoming increasingly prevalent, there is a clear and pressing need for drug therapies in patients with nonalcoholic fatty liver diseases (NAFLDs) and nonalcoholic steatohepatitis (NASH). At the same time, patients with primary sclerosing cholangitis (PSC) and high-risk primary biliary cholangitis (PBC) are also in need of better second-line disease-modifying drugs as well as effective therapies to manage associated symptoms (e.g. pruritus).
Bile acids (BAs) have always been of interest to gastroenterologists and hepatologists with their traditional role in fat digestion known for more than 50 years [Borgstrom et al. 1957]. In recent years, there has been a growing interest in BAs as signalling molecules and they are emerging as the key players in the quest for novel drug therapies in liver diseases. Major developments achieved in the basic and clinical research related to BAs have augmented our interest in exploiting their physiological role for therapeutic benefit in liver diseases. This has effectively set the stage to identify novel targets for treating patients with PBC, PSC and NAFLD for which there is currently limited or no effective drug therapy.
An in-depth review of BAs with respect to their chemistry, synthesis, transport and regulation is beyond the scope of this article and has been extensively reviewed elsewhere [Dawson et al. 2009; Monte et al. 2009; Schaap et al. 2014]. Therefore, in the first part of this review we provide an overview of BA synthesis, transport, signalling and regulation. We then describe recent advances in the understanding of BA receptors in relation to cholestasis, glucose and lipid metabolism, immune function and antifibrotic actions. These relevant updates will help the reader to better understand the therapeutic benefits of BAs in chronic liver diseases covered in the later part of this paper. In this review of potential novel BA-based therapeutic agents we mainly focus on the treatment of patients with PBC, PSC and NAFLD.
BA synthesis and transport
BAs along with phospholipids and cholesterol are major constituents of bile. BAs are amphipathic molecules (i.e. with both hydrophilic and hydrophobic regions) with detergent-like actions and are synthesized from enzymatic catabolism of cholesterol by the hepatocytes [Monte et al. 2009]. BA synthesis is a complex process involving at least 17 different enzymes but can be summarized into three main steps: modification of the steroid ring, cleavage of the side chain and conjugation with glycine or taurine [Russell, 2003]. Two pathways exist for BA synthesis. The classical (‘neutral’) pathway is responsible for the production of cholic acid (CA) and chenodeoxycholic acid (CDCA) which accounts for 90% of primary BA synthesis in humans [Anderson et al. 1972]. The other 10% is produced by the alternative (‘acidic’) pathway which can only produce CDCA. Cytochrome P450 7A1 (CYP7A1) is the rate-limiting enzyme in BA synthesis. After their synthesis, unconjugated CA and CDCA are targeted to the peroxisomes where they are conjugated (amidation) with glycine and taurine that renders them more hydrophilic and more readily secretable in the bile.
In humans, predominant conjugated BAs are glycoconjugates and under physiological pH conditions these conjugated BAs exist as anionic salts and are therefore called ‘bile salts’ (BS). These BS are stored in the gallbladder and upon ingestion of a meal they are released into the intestinal lumen where they facilitate absorption of fat and fat-soluble vitamins. Conjugated primary BAs present in the intestinal lumen are modified by the intestinal bacteria by deconjugation, oxidation and dehydroxylation to produce secondary BAs: lithocholic acid (LCA) and deoxycholic acid (DCA) [Ridlon et al. 2006]. Human bile predominantly contains CDCA and DCA and a very small amount of ursodeoxycholic acid (UDCA). Hydrophobicity of the BAs is a determinant of their cytotoxicity which increases in the order of LCA > DCA > CDCA > CA > UDCA [Carey, 1983].
After their normal physiological function is completed in the intestine, BAs reach the ileum where most are reabsorbed efficiently via a sodium-dependent process. The apical sodium-dependent BA transporter (ASBT, gene symbol SLC10A2) expressed in the distal ileum is the predominant transporter mediating the ileal uptake of conjugated BAs [Craddock et al. 1998]. ASBT mediates active transfer of BAs across the luminal plasma membrane to an intracellular protein called ileal BA binding protein (IBABP) that facilitates intracellular diffusion of BAs to the basolateral membrane (Figure 1). Then, BAs exit the enterocyte across the basolateral plasma membrane mediated by the organic solute transporter (OSTα/β) and enter the portal bloodstream [Rao et al. 2008]. BAs circulating in the portal circulation are transported across the basolateral membranes of the hepatocytes via sodium taurocholate cotransporting polypeptide transporter (NTCP, gene symbol SLC10A1) [Hagenbuch and Meier, 1994]. Finally, BAs are transported across the canalicular plasma membrane of the hepatocytes via the bile salt export pump (BSEP) and secreted into bile. This efficient cycle between small intestine and liver is repeated several times a day to ensure 95% of BAs re-enter the liver, leaving only approximately 5% (or approximately 0.5 g/day) in the intestinal lumen [Hofmann, 1984].
Figure 1.

Enterohepatic circulation of bile acids (BAs) via enterocyte in terminal ileum. (1) Primary BAs synthesized in liver and excreted into duodenum as constituent of bile; (2) BAs avidly and actively reabsorbed in the terminal ileum via ASBT (apical sodium bile acid transporter); (3) BAs transported intracellularly by IBABP (intracytosolic bile acid binding protein); (4) BAs free to bind with nuclear receptor FXR (farnesoid X receptor); (5) BAs released into portal venous circulation via organic solute transporter (OST) α/β and circulated back to liver. SHP, small heterodimer partner.
BA regulation
BA homeostasis is under tight regulation mediated by the negative feedback effect of BAs on the activity and expression of CYP7A1 as well as signalling via farnesoid X receptor (FXR) and fibroblast growth factor (FGF19).
FXR
FXR was first identified in 1995 as an orphan receptor. It is a member of the nuclear receptor superfamily and acts as key regulator in a diverse range of cell functions including development, differentiation and metabolism [Mangelsdorf et al. 1995]. FXR was so named because a supraphysiological concentration of farnesol, an intermediate in the mevalonate pathway, was found to demonstrate weak agonism [Modica et al. 2010]. The discovery that primary BAs were the natural, endogenous ligands for FXR was reported at the turn of the century [Makishima et al. 1999; Parks et al. 1999]. Soon FXR was implicated in BA homeostasis following the discovery that FXR knockout (FXR–/–) mice showed diminished ability to downregulate CYP7A1 mRNA in response to BAs [Sinal et al. 2000]. Although FXR is expressed in the ileum, liver, adrenal glands and the kidneys, the intestine (mainly ileum) seems to have the most intense FXR expression [Inagaki et al. 2006]. Indeed, among all the nuclear hormone receptors, FXR is the most dedicated to BA signalling [Schaap et al. 2014]. The activation of FXR by primary BAs has several downstream effects on both hepatocytes and enterocytes. In the hepatocytes, FXR activation induces small heterodimer partner (SHP) that downregulates the synthesis of BAs by inhibiting CYP7A1 [Kir et al. 2012]. Also, FXR activation downregulates NTCP (reduced BA uptake) and upregulates BSEP (increased export of BAs) [Martinez-Augustin and Sanchez De Medina, 2008]. In the enterocytes, FXR activation reduces ASBT expression (inhibits BA absorption) and increases expression of IBABP and OSTα/β (prevents intracellular BA accumulation) [Martinez-Augustin and Sanchez De Medina, 2008]. FXR activation also prevents BA toxicity by transcriptional induction of detoxification enzymes and canalicular secretion of BAs via upregulation of BSEP.
FGF19
A second FXR-dependent mechanism to reduce primary BA synthesis is through the production of enterokine FGF15 (in rodents) or FGF19 (in humans) [Holt et al. 2003; Inagaki et al. 2005]. When there is a high BA load in the ileum, activated FXR induces transcription of the FGF19 in the ileum. FGF19 is able to travel in the bloodstream and bind to its receptor hepatocyte FGF receptor 4 (FGFR4) and initiate a SHP-independent downregulation of CYP7A1, resulting in inhibition of BA synthesis [Jones, 2012; Kir et al. 2012].
In summary, FXR and FGF19 activation reduces endogenous BA synthesis, protects hepatocytes from BA toxicity and promotes secretion of BAs. Therefore the ‘anticholestatic’ effects of the BA-FXR-FGF19 signalling cascade have potential therapeutic implications in cholestatic diseases.
Transmembrane G protein coupled receptor 5
Transmembrane G protein coupled receptor 5 (TGR5, also called Gpbar-1) is a G protein coupled BA receptor that was identified as the first cell surface receptor for BAs [Maruyama et al. 2002; Kawamata et al. 2003]. TGR5 is not expressed in the hepatocytes but found in the cholangiocytes as well as a variety of other cell types such as brown adipose tissue, brain, gall bladder epithelium, intestines, spleen, endothelial cells, Kupffer cells and CD14+ cells [Maruyama et al. 2002; Kawamata et al. 2003; Keitel et al. 2007]. Although many BAs are capable of activating TGR5, the most potent natural ligands are taurine-conjugated secondary BAs, such as taurolithocholate [Keitel et al. 2008]. It has been shown that BA-induced TGR5 activity plays a major role in glucose homeostasis, increased energy expenditure, oxygen consumption and gallbladder filling [Katsuma et al. 2005; Watanabe et al. 2006; Thomas et al. 2009; Li et al. 2011]. In addition, TGR5 activation improves hepatic steatosis and insulin sensitivity and protects biliary epithelium against the detergent effect of BAs. More recently, BA-mediated TGR5 activation has also been shown to have an anti-inflammatory role by reducing nuclear factor κB (NFκB) translocation [Li et al. 2011]. Due to its diverse and favourable effects, TGR5 is an emerging target for drug discovery with a potentially beneficial role of TGR5 agonists in the treatment of type 2 diabetes mellitus (T2DM) and inflammation-driven metabolic diseases such as NASH.
Scientific rationale for BA-based therapy
In addition to their key role in BA homeostasis, FXR activity and FGF19 signalling are involved in diverse biological pathways. Essentially, FXR exerts its functions by eliciting transcriptional alterations and controls a number of important metabolic pathways [Schaap et al. 2014]. In this section, we briefly review the key biological processes modulated by FXR and FGF19 with respect to their attractive therapeutic implications.
Glucose and lipid metabolism
Activation of FXR inhibits the expression of hepatic sterol regulatory element-binding protein 1c (SREBP-1c) [Watanabe et al. 2004]. SREBPs are transcription factors that act as master regulators of lipid metabolism. They act to control the biogenesis of cholesterol and also control the expression of genes involved in lipogenesis. In an animal model, activation of FXR by CA inhibited hepatic SREBP-1c expression in a SHP-dependent manner, leading to reduction in serum triglyceride levels [Watanabe et al. 2004]. In addition, FXR can induce the expression of apolipoprotein C-II [Houten et al. 2006] which is a coactivator for lipoprotein lipase that acts to clear serum triglyceride from the circulation. FGF19 is also shown to regulate key enzymes in hepatic lipid synthesis [Miyata et al. 2011].
FXR signalling is also essential to maintain glucose homeostasis. In an FXR–/– mouse model, elevated serum glucose and impaired glucose and insulin tolerance were demonstrated by Ma and colleagues [Ma et al. 2006]. This study also demonstrated that administration of CA repressed the expression of gluconeogenic genes and decreased serum glucose in wild-type mice. In addition, FGF19 is involved in glucose metabolism with its actions resembling that of insulin; that is, inhibition of gluconeogenesis and stimulation of glycogen synthesis [Potthoff et al. 2011].
Immune functions
It has long been recognized that BAs are bacteriostatic (but weakly bactericidal) and that decrease in BAs within the small bowel leads to bacterial overgrowth [Floch et al. 1971; Berg, 1995]. This has been confirmed recently by the experimental studies of obstructive cholestasis and cirrhosis that showed oral administration of BAs reduced bacterial overgrowth as well as maintained the intestinal barrier function and prevented endotoxaemia [Lorenzo-Zuniga et al. 2003; Ogata et al. 2003]. BA-FXR signalling has been proposed as the key mechanism by which BAs control bacterial overgrowth and maintain the epithelial barrier. A landmark study showed that BAs regulate an anti-inflammatory response via FXR in the terminal ileum [Inagaki et al. 2006]. In this study of a rodent model of cholestasis FXR agonist treatment protected the epithelial barrier by increasing the expression of several genes associated with intestinal mucosal defence pathways and decreased the number of bacteria isolated from mesenteric lymph nodes. Similarly, treatment with a FXR agonist has been shown to maintain the epithelial barrier in an animal model of colitis in wild type, but not FXR−/− mice [Gadaleta et al. 2011]. In addition, FXR activation rendered several different immune cell types refractory to stimulation with lipo-polysaccharide (LPS), a bacterial cell wall component. More recent evidence shows FXR agonism reduces the LPS-induced production of proinflammatory cytokines by macrophages, whilst maintaining the production of anti-inflammatory interleukin (IL)-10 [Haselow et al. 2013] and attenuates the chemoattractant IL-8 response to stimulation with tumour necrosis factor α (TNFα) [Speight et al. 2015]. Finally, FXR and FGF19 exert anti-inflammatory activity via suppression of NFκB, which is a key nuclear receptor in both acute and chronic inflammatory processes. This is supported by observations that FXR and NFκB mutually antagonize each other [Wang et al. 2008] and activation of FXR and FGF19 inhibit the expression of NFκB controlled inflammatory genes [Drafahl et al. 2010; Gadaleta et al. 2010; Zhou et al. 2014].
Taken together, these results suggest FXR agonists have anti-inflammatory actions and may have potential therapeutic utility in preventing bacterial translocation and reducing spontaneous bacterial peritonitis in patients with cholestasis and cirrhosis.
Liver fibrosis and carcinogenesis
Evidence shows that FGF19 increases proliferation of hepatocytes through activation of FGFR4, and BA-FXR-FGF19 signalling is essential for normal liver regenerative process [Huang et al. 2006; Wu et al. 2010; Zhang et al. 2012; Uriarte et al. 2013; Kong et al. 2014]. Therefore, inhibition of FGF19 signalling could be a potential therapy for hepatocellular carcinoma (HCC). Also the observation that FXR–/– mice have high incidence of HCC [Kim et al. 2007] suggests the potential regulatory role of FXR in tumour suppression. This has recently been corroborated by two animal studies in which FXR agonist treatment prevented development of liver cancer and reduced liver tumour size and metastasis [Deuschle et al. 2012; Jiang et al. 2013].
FXR activation is also implicated in the inhibition of fibrotic mechanisms within the liver via hepatic stellate cells (HSCs) [Fiorucci et al. 2005; Renga et al. 2011]. In a mouse model, administration of a FXR agonist for 12 weeks promoted resolution of liver fibrosis [Fiorucci et al. 2004]. However, a more recent study has contradicted this by showing a low level of FXR expression in HSCs in liver fibrosis and suggesting HSCs may not represent direct therapeutic targets for FXR ligands [Fickert et al. 2009]. Therefore, the current evidence on the antifibrotic effect of FXR is equivocal and merits further investigation.
Novel BA-based therapies
PBC, PSC and NASH represent complex, multifactorial diseases in which effective drug management remains an unmet clinical need. There is a clear need beyond UDCA in patients with PBC and PSC and beyond diet and lifestyle modifications in patients with NASH. In this section we review innovative BA-based therapeutic approaches being investigated for these diseases (summary in Table 1).
Table 1.
Novel bile acid based therapeutic approaches in chronic liver diseases.
| Class of molecule | Example molecules | Therapeutic rationale | Target disease | Phase of development | ClinicalTrials.gov identifier |
|---|---|---|---|---|---|
| FXR agonists | INT-747, INT-767, GW4064, GSK2324, PX-102, Way362450, fexaramine, LJN452 | ↓BA synthesis and promotion of BA excretion | PBC | Phase II completed | NCT00570765 |
| Phase II completed | NCT00550862 | ||||
| Phase III (POISE) | NCT01473524 | ||||
| Phase III | NCT02308111 | ||||
| Phase II | NCT02516605 | ||||
| Phase II | NCT02516605 | ||||
| PSC | Phase II (AESOP) | NCT02177136 | |||
| ↓lipogenesis, gluconeogenesis and liver inflammation | T2DM with NAFLD | Phase II completed | NCT00501592 | ||
| NASH | Phase II | NCT01265498 | |||
| ↑tumour suppression and ↓liver fibrosis | HCC | – | – | ||
| TGR5 agonist | INT-767, INT-777 | ↓BA synthesis, ↑bile flow, anti-inflammatory | PSC | ||
| ↑energy expenditure, anti-inflammatory | NASH | – | – | ||
| FGF-19 analogue | NGM-282 | ↓BA synthesis, anti-inflammatory, antifibrotic | PBC | Phase II | NCT02026401 |
| ASBT inhibitor | LUM-001, GSK2330672 | ↓enterohepatic circulation of BAs and ↑faecal excretion | Pruritus in PBC | Phase II (CLARITY) | NCT01904058 |
| Phase II | NCT01899703 | ||||
| PPAR agonists | Fenofibrate, bezafibrate | ↓BA synthesis and promotion of BA excretion, anti-inflammatory, may ↑FXR activity | PBC, PSC | Phase III (BEZURSO) | NCT01654731 |
| GFT505 | ↑insulin sensitivity and glucose homeostasis, hepatoprotective, anti-inflammatory | NASH | Phase II (GOLDEN) | NCT01694849 | |
| UDCA related | norUDCA | cholangioprotective, stabilizes ‘biliary bicarbonate umbrella’ | PSC | Phase II (NUC-3) | NCT01755507 |
| Tauroursodeoxycholate (TUDCA) | more hydrophilic than UDCA | PBC | Phase III | NCT01857284 | |
| Fatty acid–bile acid conjugate | Aramchol | Stearoyl coenzyme A desaturase 1 (SCD1) inhibitor, ↓liver fat | NASH | Phase II (ARREST) | NCT02279524 |
ASBT, apical sodium dependent BA transporter; BA, bile acid; FGF, fibroblast growth factor; FXR, farnesoid X receptor; HCC, hepatocellular carcinoma; NAFLD, nonalcoholic fatty liver disease; NASH, nonalcoholic steatohepatitis; PBC, primary biliary cholangitis; POISE, PBC OCA International Study of Efficacy; PPAR, peroxisome proliferator-activated receptor; PSC, primary sclerosing cholangitis; T2DM, type 2 diabetes mellitus; TGR, transmembrane G protein-coupled receptor; UDCA, ursodeoxycholic acid.
UDCA: ‘the current BA therapy’
UDCA is the only US Food and Drug Administration (FDA) approved drug for PBC and it is the current standard of care for patients with this condition. The mechanism of action of UDCA has been well established and comprehensively explained in an excellent recent review [Beuers et al. 2015]. In brief, UDCA has potent anticholestatic, antiapoptotic and anti-inflammatory properties. Notably, UDCA is a weak FXR and TGR5 ligand [Parks et al. 1999; Halilbasic et al. 2013]. The optimum dose of UDCA in treating patients with PBC is 13–15 mg/kg/day and guidelines recommend initiating treatment at a low dose and increasing it gradually to the optimum dose [Beuers et al. 2009; Lindor et al. 2009].
Multiple lines of evidence confirm that UDCA improves biochemical markers of cholestasis and may delay the progression of PBC [Beuers et al. 2009; Lindor et al. 2009]. However, unfortunately the response to UDCA is not universal and up to 40% of patients with PBC do not improve their liver biochemistry on UDCA. Indeed a substantial proportion of patients have disease progression despite UDCA therapy. Patients with incomplete biochemical response to UDCA are referred to as ‘UDCA nonresponders’ based on different treatment response biochemical criteria (e.g. Paris and Barcelona criteria) [Corpechot, 2012]. UDCA nonresponders are at higher risk of disease progression, symptom burden and poor prognosis compared with ‘UDCA responders’. In addition, the Global PBC Study Group recently demonstrated that serum levels of alkaline phosphatase (ALP) and bilirubin are surrogate endpoints of outcomes in PBC and patients with an ALP greater than 1.67 × upper limit of normal (ULN) or bilirubin greater than ULN have increased risk of transplantation or death [hazard ratio (95% confidence interval, CI): 2.83 (2.4–3.4); p < 1 × 10−34] [Lammers et al. 2014].
In PSC patients the conventional dose of UDCA (10–15 mg/kg/day) is safe but high dose (28–30 mg/kg/day) has been shown to be harmful [Lindor et al. 2009]. Moreover, the long term efficacy of UDCA therapy in PSC is unclear as the evidence suggests that UDCA improves liver biochemistry but has no significant effect in slowing disease progression [Poropat et al. 2011]. Therefore, the current guidelines recommend against the use of UDCA as medical therapy in PSC [Beuers et al. 2009; Chapman et al. 2010].
In PBC and PSC, UDCA is not effective in improving cholestasis-associated symptoms such as pruritus and fatigue. Due to these limitations of UDCA, there is an urgent need for novel therapy in PSC and second line therapies in patients with PBC with UDCA nonresponse status. The potential therapeutic role of UDCA in other but rare liver diseases is also being explored. Recent experimental models of polycystic liver disease (PLD) have shown that UDCA inhibits hepatic cystogenesis by inhibiting the proliferation of polycystic cholangiocytes [Munoz-Garrido et al. 2015]. Therefore, UDCA may be an effective therapeutic option in reducing liver volume in PLD and help to improve symptoms caused by the mass effect of polycystic liver. An international, multicentre, randomised controlled trial is currently recruiting patients to assess the efficacy of UDCA (15–20 mg/kg/day UDCA for 24 weeks) in reducing total liver volume in PLD patients (CURSOR study; NCT02021110).
FXR agonists
As noted above, among all nuclear receptors FXR has emerged as a prime therapeutic target due to its diverse functions in the regulation of BAs, metabolism of glucose and lipids, and anti-inflammatory activity. Several FXR agonists have been developed with two basic structures: small molecule, steroidal semisynthetic ligands and nonsteroidal, fully synthetic ligands. The most clinically advanced therapeutic FXR agonist is INT-747 [obeticholic acid (OCA), 6α-ethyl-chenodeoxycholic acid, Intercept Pharmaceuticals, New York, USA]. OCA is a steroidal semisynthetic BA in which CDCA has been modified by the addition of an alkyl group to form a more potent FXR agonist [Pellicciari et al. 2002].
GW4064 (GlaxoSmithKline, NC, USA) is a nonsteroidal fully synthetic FXR agonist first developed in 2000 [Crawley, 2010]. Animal studies have shown that GW4064 prevents diet-induced hepatic steatosis and insulin resistance, and attenuates endotoxin-induced hepatic inflammation by repressing macrophage activation [Ma et al. 2013; Yao et al. 2014]. As an FXR agonist, GW4064 has greater potency than CDCA but is currently not being evaluated for clinical use. It can be commercially obtained from Sigma Aldrich (St. Louis, USA) for experimental use.
PX-102 is a therapeutic nonsteroidal compound manufactured as a modification of GW4064 (Phenex Pharmaceuticals AG, Ludwigshafen, Germany). It has demonstrated some efficacy in the mouse models of NAFLD by decreasing levels of serum and liver cholesterol and triglyceride [Hambruch et al. 2013].
WAY-362450 (Exelixis Inc., California) is a fully synthetic agonist manufactured as an azepino derivative [Flatt et al. 2009]. Studies on animal models of NAFLD and NASH demonstrated that treatment with WAY-362450 reduced liver inflammation and fibrosis with an associated decrease in serum liver enzymes [Zhang et al. 2009].
Finally, fexaramine is another nonsteroidal FXR ligand shown to have distinct genomic targets and favourable metabolic effects in a mice model but currently is not being studied in humans [Downes et al. 2003].
FXR therapeutics in PBC
By virtue of its potent FXR agonist action and resulting effects on BAs, OCA is emerging as a promising second-line agent for treating patients with PBC who are ‘UDCA nonresponders’. Two phase II randomized placebo-controlled trials have evaluated OCA in PBC with improvement in liver enzymes as the primary endpoint and as a surrogate marker for patient outcome. The first study [ClinicalTrials.gov identifier: NCT00570765] showed that in comparison to placebo, monotherapy with OCA 10 or 50 mg daily for 12 weeks (n = 59) reduced ALP from a mean of 3.9 × ULN to 1.9 × ULN [Kowdley et al. 2011]. Significant improvements were also seen in the levels of serum γ-glutamyl transpeptidase (γ-GT), bilirubin, C-reactive protein, immunoglobulin M (IgM) and TNFα. More recently, in a phase II multicentre double-blind efficacy trial, 165 patients with PBC and inadequate response to UDCA (serum alkaline phosphatase levels > 1.5 × ULN) were randomly treated with 10 mg, 25 mg or 50 mg doses of OCA or placebo once daily for 3 months [ClinicalTrials.gov identifier: NCT00550862] [Hirschfield et al. 2015]. Compared with placebo, OCA significantly decreased ALP levels with average decline of 20–25% from baseline. Significant improvements were also seen in the levels of serum bilirubin, γ-GT and ALT. The biochemical benefit of OCA was maintained in 61 patients who had completed a 12-month open-label extension of the study. Currently two phase III studies of OCA in PBC are ongoing [ClinicalTrials.gov identifiers: NCT01473524, NCT02308111] with early results from the first phase III PBC OCA International Study of Efficacy (POISE) study showing clinically meaningful biochemical improvement [Nevens et al. 2014].
LJN452 (Novartis, Basel, Switzerland) is a non-BA FXR agonist currently entering a phase II trial to assess safety, tolerability and efficacy in patients with PBC [ClinicalTrials.gov identifier: NCT02516605].
FXR therapeutics in NAFLD
NAFLD is the most common cause of chronic liver disease in the developed world, affecting up to a third of the population. It is characterized by the accumulation of hepatic fat and presents as a spectrum from simple steatosis to NASH, fibrosis and eventually cirrhosis [Sattar et al. 2014]. NAFLD is strongly associated with T2DM and patients with NASH have an increased risk for the development of progressive fibrosis, cirrhosis and HCC. So far, OCA is the only FXR agonist studied in patients with NAFLD, with encouraging results from the FLINT (Farnesoid X nuclear receptor ligand obeticholic acid for non-cirrhotic, non-alcoholic steatohepatitis) study reported recently [Neuschwander-Tetri et al. 2015]. In this double-blind, randomized, placebo-controlled trial, patients with NASH without cirrhosis were randomized to receive either 25 mg of OCA (n = 141) or placebo (n = 142) for 72 weeks. The primary outcome measure was reduction in the histological score for fibrosis (NAFLD activity score) from baseline to the end of treatment. The trial was stopped early as 45% of patients in the OCA group had reached the primary endpoint compared with 21% in the placebo group (relative risk 1.9; 95% CI 1·3–2·8). The OCA intervention also significantly improved the serum ALT levels. These results suggest that the FXR agonist OCA may be beneficial in NAFLD and prevent progression of fibrosis in patients with NASH.
Safety of FXR therapy
Pruritus is the most common adverse event reported to be associated with OCA in PBC and NAFLD trials. In the phase II PBC trial, frequency of pruritus was 47% with 10 mg, 87% with 25 mg and 80% with 50 mg of OCA [Hirschfield et al. 2015]. Similarly in the FLINT study, 23% of patients in the OCA arm developed pruritus compared with 6% in the placebo group [Neuschwander-Tetri et al. 2015]. Data from both trials suggested that pruritus was directly related to the OCA dose. This issue is being investigated in the phase III (POISE) study, with early results suggesting the frequency of pruritus is lowest when OCA is started at 5 mg and titrated upwards [Nevens et al. 2014].
A potentially more concerning adverse event in the FLINT study was dyslipidaemia seen in patients treated with OCA. Increase in low-density lipoprotein and decrease in high-density lipoprotein (HDL) levels were observed in the OCA group. Although dyslipidaemia is a well-known cardiovascular risk factor, whether this is true for patients treated with an FXR agonist is unclear. Interestingly animal models of atherosclerosis have provided some evidence to suggest that FXR agonists are protective [Hartman et al. 2009]. Clearly more research is needed to assess the cardiovascular risk of FXR agonist therapy in NASH. Interestingly in the PBC study OCA treatment was associated with decrease in serum levels of total and HDL cholesterol [Hirschfield et al. 2015].
An unproven safety concern is the potential for FXR agonists to predispose people to the development of HCC induced by FXR overexpression and elevation of circulating FGF19 levels. FGF19 transgenic mice develop HCC [Nicholes et al. 2002] and overexpression of FGF19 has been associated with development of HCC in both animal models and potentially in humans [Lin and Desnoyers, 2012; Schaap et al. 2015]. Therefore, it is feasible that overpromotion of FXR with FXR agonist therapy could lead to high levels of FGF19, which in turn is carcinogenic. This concern merits further clinical evaluation. Similar procarcinogenic concern applies to FGF19 analogues. One proposal to overcome this undesirable effect is to engineer FGF19 variants that have lost the mitotic activity but still are effective in their metabolic activity [Wu et al. 2010].
INT-767
INT-767, a semisynthetic BA analogue, is a 23-sulphate derivative of OCA with dual FXR and TGR5 agonist actions but with a higher affinity to FXR [Rizzo et al. 2010]. Prominent features of INT-767 include inhibition of BA synthesis, simulation of bicarbonate-rich choleresis by enhancing biliary bicarbonate secretion and immune modulation via inhibition of NFκB. In the mouse model of sclerosing cholangitis, INT-767 reduced serum levels of ALT, ALP as well as liver inflammation and fibrosis [Baghdasaryan et al. 2011]. Interestingly these effects were shown to be mediated exclusively by FXR and not by TGR5. Therefore, INT-767 may be a potential therapeutic agent in treating patients with cholestasis and it is currently entering phase I clinical trials.
INT-777/TGR5 agonists
INT-777 [6α-ethyl-23(S)-methylcholic acid] is a potent, semisynthetic and selective TGR5 agonist [Pellicciari et al. 2009]. Animal studies have shown that INT-777 increases bile flow, produces significant reduction in weight gain and adiposity as well as improvement of liver function with concomitant reductions in steatosis and fibrosis [Pellicciari et al. 2009; Thomas et al. 2009]. These results suggest promising therapeutic potential for INT-777 in the treatment of obesity and related disorders such as NASH. A theoretical concern with the use of TGR5 agonists is the potential for aggravating pruritus, a common symptom in patients with cholestatic liver disease. Although animal studies have shown that activation of TGR5 can induce itch [Alemi et al. 2013; Lieu et al. 2014] it remains unknown whether therapeutic use of TGR5 agonists will have a similar effect on pruritus. Currently there are no ongoing clinical trials with INT-777.
NGM-282
NGM-282 (NGM Biopharmaceuticals, Inc., San Francisco, CA, USA), a biological drug, is a recombinant variant of FGF19 with potential anticholestatic properties. A phase II clinical trial evaluating the safety and tolerability of 28 days of treatment of NGM-282 with UDCA in patients with PBC has recently been completed [ClinicalTrials.gov identifier: NCT02026401] and results are awaited.
Norursodeoxycholic acid
24-norUrsodeoxycholic acid (norUDCA) is a synthetic, side-chain-shortened UDCA homologue. With its hepatocyte and cholangiocyte protective properties it has recently emerged as an attractive therapeutic candidate for cholestatic liver diseases, especially for PSC. NorUDCA differs from UDCA in metabolism and therapeutic mechanisms, with important clinical consequences. Like UDCA, norUDCA is not a direct FXR or TGR5 ligand but it is significantly more hydrophilic and less toxic than UDCA [Fickert et al. 2013]. NorUDCA is superior to UDCA in the treatment of sclerosing cholangitis, attributed largely to its ability to increase the hydrophilicity of biliary BAs, stimulate bile flow with flushing of injured bile ducts, and induce detoxification and elimination routes for BAs [Hofmann et al. 2005; Fickert et al. 2006]. Because of its shortened side chain norUDCA is not conjugated with taurine or glycine in the liver and is secreted into the bile in its unconjugated form. Human studies have shown norUDCA induces a sustained bicarbonate-rich hypercholeresis, the increased bile flow being attributed mainly to ‘cholehepatic shunting’ [Hofmann et al. 2005]. The cholangioprotective effect is mainly due to profound alkalinization of bile which stabilizes the ‘biliary bicarbonate umbrella’ [Hohenester et al. 2012] and in turn reduces ductular reaction, inflammation and fibrosis [Halilbasic et al. 2009; Fickert et al. 2013]. In addition to the anticholestatic and cholangioprotective mechanisms, rodent studies have suggested that norUDCA has potential antiproliferative, anti-inflammatory and antifibrotic properties [Fickert et al. 2006; Halilbasic et al. 2009] which could be beneficial in both cholestatic and noncholestatic conditions. Recent data also suggest norUDCA produces significant suppression of lipogenesis and normalization of BA metabolism through mechanisms involving crosstalk between CYP7A1 and SHP [Beraza et al. 2011].
Due to its multiple beneficial properties suggested by experimental data, norUDCA is a promising drug therapy to attenuate the progression of complex disorders such as PSC and NASH. Also the potential beneficial effects of combined therapy with norUDCA (bicarbonate-rich choleresis) and FXR agonist (suppression of BA synthesis) in cholestatic liver disease merits further exploration. Currently the optimal dose of norUDCA for therapeutic benefit is not known and a large, multicentre, double-blind, placebo-controlled, randomized dose-finding phase II trial (Dr Falk Pharma GmbH, Freiburg im Breisgau, Germany) is evaluating the efficacy of three different doses of norUDCA for the treatment of PSC [ClinicalTrials.gov identifier: NCT01755507].
Aramchol
Aramchol [(3β-arachidyl-amido, 7α-12α-dihydroxy, 5β-cholan-24-oic acid), Trima Israel Pharmaceutical Products, Maabarot, Israel] is a fatty acid–BA conjugate currently being investigated for NAFLD and NASH. Aramchol is a novel synthetic lipid molecule obtained by conjugating two natural components, CA and arachidic acid (saturated fatty acid). The main mechanism of action of Aramchol is via inhibition of the stearoyl coenzyme A desaturase 1 (SCD1) activity which is a key enzyme modulating fatty acid metabolism in the liver [Dobrzyn and Ntambi, 2005; Leikin-Frenkel et al. 2008]. SCD1 inhibition decreases the synthesis and increases β oxidation of fatty acids, resulting in decreased hepatic storage of triglycerides and fatty acid esters.
Aramchol has received a fast-track status from the FDA. In a phase IIa placebo-controlled trial of 58 patients [ClinicalTrials.gov identifier: NCT01094158], 3 months of treatment with single daily dose of aramchol (100 or 300 mg) was safe and well tolerated and produced significant and dose-dependent reduction in liver fat [Safadi et al. 2014]. A large multicentre, double-blind, placebo-controlled phase IIb study (ARREST trial) is currently evaluating the safety and efficacy of two aramchol doses in patients with NASH without cirrhosis [ClinicalTrials.gov identifier NCT02279524] with reduction in hepatic steatosis as the primary endpoint.
ASBT inhibitor
As noted above, ASBT is a key protein involved in the enterohepatic circulation of BAs and maintaining the BA pool. Physiological effects of ASBT inhibition include lack of ileal BA uptake, increased faecal BAs, reduced FXR stimulation and reduced FGF19 levels. In animal studies, SC-435 (an ASBT inhibitor) produced BA malabsorption (resulting in diarrhoea) and lowered plasma cholesterol [West et al. 2003]. As ileal BA uptake is upregulated in PBC [Lanzini et al. 2003], pharmacological inhibition of ASBT on the circulating levels of BAs in PBC has generated considerable interest. Recently in a bile duct ligated mice model, treatment with ASBT inhibitor A4250 was shown to attenuate BA-mediated cholestatic liver injury by reducing biliary BA output [Baghdasaryan et al. 2014].
Since BAs have been proposed as potential direct or indirect pruritogens in cholestasis, ASBT inhibitors may also have a role in treating pruritus. Recently two large multicentre, randomized phase II clinical trials evaluating the safety and efficacy of ASBT inhibitor drugs (GSK2330672 and LUM001) in patients with PBC and pruritus have completed recruitment [ClinicalTrials.gov identifiers: NCT01899703, NCT01904058]. The results of these studies are likely to inform the safety and therapeutic potential of ASBT inhibitors in the treatment of cholestatic pruritus.
Fibrates
Fenofibrate and bezafibrate are fibric acid derivatives that have been in use for over two decades primarily to treat hyperlipidaemia in patients with cardiovascular and metabolic diseases. Following the first study in 1993 that suggested fibrates improve liver biochemistry [Day et al. 1993], they have been actively pursued as potential adjuvants to UDCA therapy to improve cholestasis. The primary mechanism of anticholestatic effect of fibrates is through inhibition of BA synthesis mediated via nuclear receptor peroxisome proliferator-activated receptor (PPAR). Fenofibrate is a PPARα selective agonist and bezafibrate is a ‘pan-PPAR’ agonist as it activates all three isoforms (α, γ and δ). PPARα plays a key role in maintaining BA homeostasis by regulating genes responsible for BA synthesis and transport [Ghonem et al. 2015]. Therefore, by activating PPARα, fenofibrate and bezafibrate reduce BA synthesis (downregulate CYP7A1), decrease BA secretion into bile, facilitate elimination of toxic BA and increase biliary phospholipid output. Fibrates also have immune modulator function via inhibition of NFκB, increased apolipoprotein AII (inhibits lymphocyte activation), and suppression of lymphocyte proliferation [Vu-Dac et al. 1995; Schoonjans et al. 1996]. In addition, the crosstalk between PPARα and FXR may enhance PPARα transcription in HSCs, leading to decreased liver fibrosis [Pineda Torra et al. 2003].
To date, a number of studies (mostly uncontrolled and small case series) with bezafibrate and fenofibrate have consistently reported significant improvement in ALP, ALT and IgM in patients with PBC. Recent systematic reviews and meta-analyses also support the use of fibrates as adjuvant treatment in patients with PBC who are UDCA nonresponders despite treatment with the optimum dose of UDCA for 12 months [Grigorian et al. 2015; Zhang et al. 2015]. However, fibrates currently carry contraindications for use in patients with hepatic or severe renal dysfunction, including in patients with PBC. Therefore, safety and efficacy of fibrates need further evaluation in prospective studies before they can be used in routine clinical practice for PBC. Currently, a large multicentre, prospective, double-blind, placebo-controlled phase III study of bezafibrate in combination with UDCA (BEZURSO study) in PBC is recruiting [ClinicalTrials.gov identifier: NCT01654731] and the results are eagerly awaited to clarify the true effect of fibrates in cholestasis.
GFT505
GFT505 (GENFIT, Loos, France) a novel PPARα/δ agonist, is currently being developed as a novel therapy for NASH and has received FDA fast-track status. Preclinical studies showed hepatoprotective effects of GFT505 as it decreased hepatic lipid accumulation, improved liver dysfunction markers, and inhibited proinflammatory gene expression [Staels et al. 2013]. GFT505 treatment also decreased plasma concentrations of ALT, ALP and γ-GT. In addition, GFT505 has an insulin-sensitizing effect which in combination with hepatoprotective effects makes it a potential therapy for NAFLD [Cariou and Staels, 2014]. Currently an international placebo-controlled phase IIb study (GOLDEN trial) is investigating the safety and efficacy of GFT505 (80 and 120 mg once daily for 52 weeks) in patients with NASH without cirrhosis, with resolution of NASH without worsening of fibrosis as the primary endpoint [ClinicalTrials.gov identifier: NCT01694849].
BA therapy in malignancy
A few years ago BAs were proposed as potential shuttles to deliver chemotherapy agents to treat tumours such as HCC, cholangiocarcinoma (CCA) and colorectal carcinoma [Kramer et al. 1992]. The rationale for this proposal was that the tumours in the enterohepatic circulation maintain good expression of BA transporter proteins (especially ASBT) and the ‘BA-drug couple’ would be efficiently taken up by carrier proteins expressed in the tumour cells. Therefore specific organotropic cytostatic BA derivatives (called ‘Bamets’) were developed to enhance anti-tumour drug delivery and increase tumour sensitivity to chemotherapy [Criado et al. 1997]. Conjugates of cisplatin with glycocholate (Bamet-R2) and with UDCA (Bamet-UD2) have been proved useful in the chemotherapy of experimental models of HCC [Macias et al. 1998; Larena et al. 2002]. More recently, Bamet-UD2 has been shown to inhibit tumour growth in CCA cells expressing ASBT [Lozano et al. 2015]. Results of these experimental models are encouraging but the possibility of using BA derivatives to treat malignancies needs further clinical evaluation.
Conclusion
The landscape of treating chronic liver diseases is rapidly changing largely due to recent advances in unravelling the role of BAs as signalling molecules in metabolic and cholestatic diseases. The understanding of diverse role of BAs in biological pathways is not complete and is continuing to evolve. Novel BA-based pharmaceuticals have raised hope for availability of better therapies in the near future with several compounds already in clinical trials. The current and future development of drugs based on the therapeutic concept of BAs is most likely to benefit patients with PBC, PSC and NASH.
Acknowledgments
VSH, RAS and REE researched the literature and contributed equally in the preparation of the first draft of the manuscript. DEJJ provided intellectual input and made substantial contributions to discussion of content. All authors reviewed and approved the final manuscript.
VSH gratefully acknowledges the National Institute for Health Research (NIHR) Newcastle Biomedical Research Centre (BRC) for the clinical research fellowship grant.
Footnotes
Funding: This research received no specific grant from any funding agency in the public, commercial, or not for-profit sectors.
Conflict of interest statement: REE is partially funded by a research grant from Intercept Pharmaceuticals. DEJJ has research funding from Intercept, GSK, Lumena and Shire pharmaceuticals and has undertaken consultancy for GSK and Intercept in relation to bile acid therapeutics. VSH and RAS do not have any conflict of interest.
Contributor Information
Vinod S. Hegade, Clinical Research Fellow, Fourth Floor, Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK.
R. Alexander Speight, Institute of Cellular Medicine, Faculty of Medical Science, Newcastle University, Newcastle upon Tyne, UK.
Rachel E. Etherington, Institute of Cellular Medicine, Faculty of Medical Science, Newcastle University, Newcastle upon Tyne, UK
David E. J. Jones, Institute of Cellular Medicine, Faculty of Medical Science, Newcastle University, Newcastle upon Tyne, UK
References
- Alemi F., Kwon E., Poole D., Lieu T., Lyo V., Cattaruzza F., et al. (2013) The TGR5 receptor mediates bile acid-induced itch and analgesia. J Clin Invest 123: 1513–1530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Anderson K., Kok E., Javitt N. (1972) Bile acid synthesis in man: Metabolism of 7-hydroxycholesterol-14 C and 26-hydroxycholesterol-3 H. J Clin Invest 51: 112–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baghdasaryan A., Claudel T., Gumhold J., Silbert D., Adorini L., Roda A., et al. (2011) Dual farnesoid X receptor/TGR5 agonist INT-767 reduces liver injury in the MDR2–/– (ABCB4–/–) mouse cholangiopathy model by promoting biliary HCO-3 output. Hepatology 54: 1303–1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baghdasaryan A., Jha P., Müller M., Auer N., Deutschmann A., Zöhrer C., et al. (2014) O135 inhibition of intestinal bile acid absorption by ASBT inhibitor A4250 protects against bile acid-mediated cholestatic liver injury in mice. J Hepatol 60: S57. [Google Scholar]
- Beraza N., Ofner-Ziegenfuss L., Ehedego H., Boekschoten M., Bischoff S., Mueller M., et al. (2011) Nor-ursodeoxycholic acid reverses hepatocyte-specific nemo-dependent steatohepatitis. Gut 60: 387–396. [DOI] [PubMed] [Google Scholar]
- Berg R. (1995) Bacterial translocation from the gastrointestinal tract. Trends Microbiol 3: 149–154. [DOI] [PubMed] [Google Scholar]
- Beuers U. (2006) Drug insight: mechanisms and sites of action of ursodeoxycholic acid in cholestasis. Nat Clin Pract Gastroenterol Hepatol 3: 318–328. [DOI] [PubMed] [Google Scholar]
- Beuers U., Boberg K., Chapman R., Chazouilleres O., Invernizzi P., Jones D., et al. (2009) EASL clinical practice guidelines: management of cholestatic liver diseases. J Hepatol 51: 237–267. [DOI] [PubMed] [Google Scholar]
- Beuers U., Boyer J., Paumgartner G. (1998) Ursodeoxycholic acid in cholestasis: potential mechanisms of action and therapeutic applications. Hepatology 28: 1449–1453. [DOI] [PubMed] [Google Scholar]
- Borgstrom B., Dahlqvist A., Lundh G., Sjovall J. (1957) Studies of intestinal digestion and absorption in the human. J Clin Invest 36: 1521–1536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carey M. (1983) Measurement of the physical-chemical properties of bile salt solutions. In: Barbara L., Dowling R., Hofmann A., Roda E. (eds), Bile Acids in Gastroenterology. Lancaster: MTP Press. [Google Scholar]
- Cariou B., Staels B. (2014) GFT505 for the treatment of nonalcoholic steatohepatitis and type 2 diabetes. Expert Opin Investig Drugs 23: 1441–1448. [DOI] [PubMed] [Google Scholar]
- Chapman R., Fevery J., Kalloo A., Nagorney D., Boberg K., Shneider B., et al. (2010) Diagnosis and management of primary sclerosing cholangitis. Hepatology 51: 660–678. [DOI] [PubMed] [Google Scholar]
- Corpechot C. (2012) Primary biliary cirrhosis and bile acids. Clin Res Hepatol Gastroenterol 36(Suppl. 1): S13–S20. [DOI] [PubMed] [Google Scholar]
- Craddock A., Love M., Daniel R., Kirby L., Walters H., Wong M., et al. (1998) Expression and transport properties of the human ileal and renal sodium-dependent bile acid transporter. Am J Physiol 274: G157–G169. [DOI] [PubMed] [Google Scholar]
- Crawley M. (2010) Farnesoid X receptor modulators: a patent review. Expert Opin Ther Patents 20: 1047–1057. [DOI] [PubMed] [Google Scholar]
- Dawson P., Lan T., Rao A. (2009) Bile acid transporters. J Lipid Res 50: 2340–2357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Day A., Feher M., Chopra R., Mayne P. (1993) The effect of bezafibrate treatment on serum alkaline phosphatase isoenzyme activities. Metabolism 42: 839–842. [DOI] [PubMed] [Google Scholar]
- Deuschle U., Schuler J., Schulz A., Schluter T., Kinzel O., Abel U., et al. (2012) FXR controls the tumor suppressor NDRG2 and FXR agonists reduce liver tumor growth and metastasis in an orthotopic mouse xenograft model. PLoS One 7: e43044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dobrzyn A., Ntambi J. (2005) Stearoyl-CoA desaturase as a new drug target for obesity treatment. Obes Rev 6: 169–174. [DOI] [PubMed] [Google Scholar]
- Downes M., Verdecia M., Roecker A., Hughes R., Hogenesch J., Kast-Woelbern H., et al. (2003) A chemical, genetic, and structural analysis of the nuclear bile acid receptor FXR. Mol Cell 11: 1079–1092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Drafahl K., Mcandrew C., Meyer A., Haas M., Donoghue D. (2010) The receptor tyrosine kinase FGFR4 negatively regulates NF-kappaB signaling. PLoS One 5: e14412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fickert P., Fuchsbichler A., Moustafa T., Wagner M., Zollner G., Halilbasic E., et al. (2009) Farnesoid X receptor critically determines the fibrotic response in mice but is expressed to a low extent in human hepatic stellate cells and periductal myofibroblasts. Am J Pathol 175: 2392–2405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fickert P., Pollheimer M., Silbert D., Moustafa T., Halilbasic E., Krones E., et al. (2013) Differential effects of norUDCA and UDCA in obstructive cholestasis in mice. J Hepatol 58: 1201–1208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fickert P., Wagner M., Marschall H., Fuchsbichler A., Zollner G., Tsybrovskyy O., et al. (2006) 24-Norursodeoxycholic acid is superior to ursodeoxycholic acid in the treatment of sclerosing cholangitis in MDR2 (ABCB4) knockout mice. Gastroenterology 130: 465–481. [DOI] [PubMed] [Google Scholar]
- Fiorucci S., Antonelli E., Rizzo G., Renga B., Mencarelli A., Riccardi L., et al. (2004) The nuclear receptor SHP mediates inhibition of hepatic stellate cells by FXR and protects against liver fibrosis. Gastroenterology 127: 1497–1512. [DOI] [PubMed] [Google Scholar]
- Fiorucci S., Rizzo G., Antonelli E., Renga B., Mencarelli A., Riccardi L., et al. (2005) A farnesoid X receptor-small heterodimer partner regulatory cascade modulates tissue metalloproteinase inhibitor-1 and matrix metalloprotease expression in hepatic stellate cells and promotes resolution of liver fibrosis. J Pharmacol Exp Ther 314: 584–595. [DOI] [PubMed] [Google Scholar]
- Flatt B., Martin R., Wang T., Mahaney P., Murphy B., Gu X., et al. (2009) Discovery of Xl335 (WAY-362450), a highly potent, selective, and orally active agonist of the farnesoid X receptor (FXR). J Med Chem 52: 904–907. [DOI] [PubMed] [Google Scholar]
- Floch M., Gershengoren W., Elliott S., Spiro H. (1971) Bile acid inhibition of the intestinal microflora – a function for simple bile acids? Gastroenterology 61: 228–233. [PubMed] [Google Scholar]
- Gadaleta R., Van Erpecum K., Oldenburg B., Willemsen E., Renooij W., Murzilli S., et al. (2011) Farnesoid X receptor activation inhibits inflammation and preserves the intestinal barrier in inflammatory bowel disease. Gut 60: 463–472. [DOI] [PubMed] [Google Scholar]
- Gadaleta R., Van Mil S., Oldenburg B., Siersema P., Klomp L., Van Erpecum K. (2010) Bile acids and their nuclear receptor FXR: relevance for hepatobiliary and gastrointestinal disease. Biochim Biophys Acta 1801: 683–692. [DOI] [PubMed] [Google Scholar]
- Ghonem N., Assis D., Boyer J. (2015) On fibrates and cholestasis: a review. Hepatology 62: 635–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grigorian A., Mardini H., Corpechot C., Poupon R., Levy C. (2015) Fenofibrate is effective adjunctive therapy in the treatment of primary biliary cirrhosis: a meta-analysis. Clin Res Hepatol Gastroenterol 39: 296–306. [DOI] [PubMed] [Google Scholar]
- Hagenbuch B., Meier P. (1994) Molecular cloning, chromosomal localization, and functional characterization of a human liver Na+/bile acid cotransporter. J Clin Invest 93: 1326–1331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Halilbasic E., Baghdasaryan A., Trauner M. (2013) Nuclear receptors as drug targets in cholestatic liver diseases. Clin Liver Dis 17: 161–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Halilbasic E., Fiorotto R., Fickert P., Marschall H., Moustafa T., Spirli C., et al. (2009) Side chain structure determines unique physiologic and therapeutic properties of norursodeoxycholic acid in MDR2–/– mice. Hepatology 49: 1972–1981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hambruch E., Miyazaki M., Miyazaki-Anzia S., Hahn U., Kremoser C. (2013) FXR agonist PX-102 improves steatosis in NAFLD mouse models. Presented at APASL Liver Week, Singapore, Malaysia. [Google Scholar]
- Hartman H., Gardell S., Evans M. (2009) Activation of farnesoid X receptor prevents atherosclerotic lesion formation in LDLR–/– and ApoE–/– mice. J Lipid Res 50: 1090–1100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haselow K., Bode J., Wammers M., Ehlting C., Keitel V., Kleinebrecht L., et al. (2013) Bile acids PKA-dependently induce a switch of the IL-10/IL-12 ratio and reduce proinflammatory capability of human macrophages. J Leukoc Biol 94: 1253–1264. [DOI] [PubMed] [Google Scholar]
- Hirschfield G., Mason A., Luketic V., Lindor K., Gordon S., Mayo M., et al. (2015) Efficacy of obeticholic acid in patients with primary biliary cirrhosis and inadequate response to ursodeoxycholic acid. Gastroenterology 148: 751–761.e758. [DOI] [PubMed] [Google Scholar]
- Hofmann A. (1984) Chemistry and enterohepatic circulation of bile acids. Hepatology 4: 4S–14S. [DOI] [PubMed] [Google Scholar]
- Hofmann A., Zakko S., Lira M., Clerici C., Hagey L., Lambert K., et al. (2005) Novel biotransformation and physiological properties of norursodeoxycholic acid in humans. Hepatology 42: 1391–1398. [DOI] [PubMed] [Google Scholar]
- Hohenester S., Wenniger L., Paulusma C., Van Vliet S., Jefferson D., Elferink R., et al. (2012) A biliary HCO3– umbrella constitutes a protective mechanism against bile acid-induced injury in human cholangiocytes. Hepatology 55: 173–183. [DOI] [PubMed] [Google Scholar]
- Holt J., Luo G., Billin A., Bisi J., McNeill Y., Kozarsky K., et al. (2003) Definition of a novel growth factor-dependent signal cascade for the suppression of bile acid biosynthesis. Genes Dev 17: 1581–1591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Houten S., Watanabe M., Auwerx J. (2006) Endocrine functions of bile acids. EMBO 25: 1419–1425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang W., Ma K., Zhang J., Qatanani M., Cuvillier J., Liu J., et al. (2006) Nuclear receptor-dependent bile acid signaling is required for normal liver regeneration. Science 312: 233–236. [DOI] [PubMed] [Google Scholar]
- Inagaki T., Choi M., Moschetta A., Peng L., Cummins C., McDonald J., et al. (2005) Fibroblast growth factor 15 functions as an enterohepatic signal to regulate bile acid homeostasis. Cell Metabolism 2: 217–225. [DOI] [PubMed] [Google Scholar]
- Inagaki T., Moschetta A., Lee Y., Peng L., Zhao G., Downes M., et al. (2006) Regulation of antibacterial defense in the small intestine by the nuclear bile acid receptor. Proc Natl Acad Sci U S A 103: 3920–3925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang Y., Iakova P., Jin J., Sullivan E., Sharin V., Hong I., et al. (2013) Farnesoid X receptor inhibits gankyrin in mouse livers and prevents development of liver cancer. Hepatology 57: 1098–1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jones S. (2012) Physiology of FDF15/19. Adv Exp Med Biol 728: 171–182. [DOI] [PubMed] [Google Scholar]
- Katsuma S., Hirasawa A., Tsujimoto G. (2005) Bile acids promote glucagon-like peptide-1 secretion through TGR5 in a murine enteroendocrine cell line STC-1. Biochem Biophys Res Commun 329: 386–390. [DOI] [PubMed] [Google Scholar]
- Kawamata Y., Fujii R., Hosoya M., Harada M., Yoshida H., Miwa M., et al. (2003) A G protein-coupled receptor responsive to bile acids. J Biol Chem 278: 9435–9440. [DOI] [PubMed] [Google Scholar]
- Keitel V., Donner M., Winandy S., Kubitz R., Haussinger D. (2008) Expression and function of the bile acid receptor TGR5 in Kupffer cells. Biochem Biophys Res Commun 372: 78–84. [DOI] [PubMed] [Google Scholar]
- Keitel V., Reinehr R., Gatsios P., Rupprecht C., Görg B., Selbach O., et al. (2007) The G-protein coupled bile salt receptor TGR5 is expressed in liver sinusoidal endothelial cells. Hepatology 45: 695–704. [DOI] [PubMed] [Google Scholar]
- Kim I., Morimura K., Shah Y., Yang Q., Ward J., Gonzalez F. (2007) Spontaneous hepatocarcinogenesis in farnesoid X receptor-null mice. Carcinogenesis 28: 940–946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kir S., Zhang Y., Gerard R., Kliewer S., Mangelsdorf D. (2012) Nuclear receptors HNF4alpha and LRH-1 cooperate in regulating CYP7A1 in vivo. J Biol Chem 287: 41334–41341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kong B., Huang J., Zhu Y., Li G., Williams J., Shen S., et al. (2014) Fibroblast growth factor 15 deficiency impairs liver regeneration in mice. Am J Physiol Gastrointest Liver Physiol 306: G893–G902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kowdley K., Jones D., Luketic V., Shapiro D. (2011) An international study evaluating the farnesoid x receptor agonist obeticholic acid as monotherapy in PBC. J Hepatol 54: S13. [Google Scholar]
- Lam B., Jeffers T., Younoszai Z., Fazel Y., Younossi Z. (2015) The changing landscape of hepatitis C virus therapy: focus on interferon-free treatment. Ther Adv Gastroenterol 8: 298–312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lammers W., Van Buuren H., Hirschfield G., Janssen H., Invernizzi P., Mason A., et al. (2014) Levels of alkaline phosphatase and bilirubin are surrogate end points of outcomes of patients with primary biliary cirrhosis: an international follow-up study. Gastroenterology 147: 1338–1349 e1335; quiz e1315. [DOI] [PubMed] [Google Scholar]
- Lanzini A., De Tavonatti M., Panarotto B., Scalia S., Mora A., Benini F., et al. (2003) Intestinal absorption of the bile acid analogue 75SE-homocholic acid-taurine is increased in primary biliary cirrhosis, and reverts to normal during ursodeoxycholic acid administration. Gut 52: 1371–1375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leikin-Frenkel A., Goldiner I., Leikin-Gobbi D., Rosenberg R., Bonen H., Litvak A., et al. (2008) Treatment of preestablished diet-induced fatty liver by oral fatty acid-bile acid conjugates in rodents. Eur J Gastroenterol Hepatol 20: 1205–1213. [DOI] [PubMed] [Google Scholar]
- Li T., Holmstrom S., Kir S., Umetani M., Schmidt D., Kliewer S., et al. (2011) The G protein-coupled bile acid receptor, TGR5, stimulates gallbladder filling. Mol Endocrinol 25: 1066–1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lieu T., Jayaweera G., Zhao P., Poole D., Jensen D., Grace M., et al. (2014) The bile acid receptor TGR5 activates the TRPA1 channel to induce itch in mice. Gastroenterology 147: 1417–1428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin B., Desnoyers L. (2012) FGF19 and cancer. Adv Exp Med Biol 728: 183–194. [DOI] [PubMed] [Google Scholar]
- Lindor K., Gershwin M., Poupon R., Kaplan M., Bergasa N., Heathcote E., et al. (2009) Primary biliary cirrhosis. Hepatology 50: 291–308. [DOI] [PubMed] [Google Scholar]
- Lorenzo-Zuniga V., Bartoli R., Planas R., Hofmann A., Vinado B., Hagey L., et al. (2003) Oral bile acids reduce bacterial overgrowth, bacterial translocation, and endotoxemia in cirrhotic rats. Hepatology 37: 551–557. [DOI] [PubMed] [Google Scholar]
- Ma K., Saha P., Chan L., Moore D. (2006) Farnesoid X receptor is essential for normal glucose homeostasis. J Clin Invest 116: 1102–1109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma Y., Huang Y., Yan L., Gao M., Liu D. (2013) Synthetic FXR agonist GW4064 prevents diet-induced hepatic steatosis and insulin resistance. Pharm Res 30: 1447–1457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Makishima M., Okamoto A., Repa J., Tu H., Learned R. (1999) Identification of a nuclear receptor for bile acids. Science 21: 1362–1365. [DOI] [PubMed] [Google Scholar]
- Mangelsdorf D., Thummel C., Beato M., Herrlich P., Schutz G., Evans R. (1995) The nuclear receptor superfamily: the second decade. Cell 83: 835–839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martinez-Augustin O., Sanchez De Medina F. (2008) Intestinal bile acid physiology and pathophysiology. World J Gastroenterol 14: 5630–5640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maruyama T., Miyamoto Y., Nakamura T., Tamai Y., Okada H., Sugiyama E., et al. (2002) Identification of membrane-type receptor for bile acids (M-BAR). Biochem Biophys Res Commun 298: 714–719. [DOI] [PubMed] [Google Scholar]
- Miyata M., Sakaida Y., Matsuzawa H., Yoshinari K., Yamazoe Y. (2011) Fibroblast growth factor 19 treatment ameliorates disruption of hepatic lipid metabolism in farnesoid X receptor (FXR)-null mice. Biol Pharm Bull 34: 1885–1889. [DOI] [PubMed] [Google Scholar]
- Modica S., Gadaleta R., Moschetta A. (2010) Deciphering the nuclear bile acid receptor FXR paradigm. Nucl Recept Signal 8: e005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Monte M., Marin J., Antelo A., Vazquez-Tato J. (2009) Bile acids: chemistry, physiology, and pathophysiology. World J Gastroenterol 15: 804–816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Neuschwander-Tetri B., Loomba R., Sanyal A., Lavine J., Van Natta M., Abdelmalek M., et al. (2015) Farnesoid X nuclear receptor ligand obeticholic acid for non-cirrhotic, non-alcoholic steatohepatitis (FLINT): a multicentre, randomised, placebo-controlled trial. Lancet 385: 956–965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nevens F., Andreone P., Mazzella G., Strasser S., Bowlus C., Invernizzi P., et al. (2014) O168 the first primary biliary cirrhosis (PBC) phase 3 trial in two decades – an international study of the FXR agonist obeticholic acid in PBC patients. J Hepatol 60: S525–S526. [Google Scholar]
- Nicholes K., Guillet S., Tomlinson E., Hillan K., Wright B., Frantz G., et al. (2002) A mouse model of hepatocellular carcinoma: ectopic expression of fibroblast growth factor 19 in skeletal muscle of transgenic mice. Am J Pathol 160: 2295–2307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ogata Y., Nishi M., Nakayama H., Kuwahara T., Ohnishi Y., Tashiro S. (2003) Role of bile in intestinal barrier function and its inhibitory effect on bacterial translocation in obstructive jaundice in rats. J Surg Res 115: 18–23. [DOI] [PubMed] [Google Scholar]
- Parks D., Blanchard S., Bledsoe R., Chandra G. (1999) Bile acids: natural ligands for an orphan nuclear receptor. Science 21: 1365–1368. [DOI] [PubMed] [Google Scholar]
- Parks D., Blanchard S., Bledsoe R., Chandra G., Consler T., Kliewer S., et al. (1999) Bile acids: natural ligands for an orphan nuclear receptor. Science 284: 1365–1368. [DOI] [PubMed] [Google Scholar]
- Pellicciari R., Fiorucci S., Camaioni E., Clerici C. (2002) 6alpha-ethyl-chenodeoxycholic acid (6-ECDCA), a potent and selective FXR agonist endowed with anticholestatic activity. J Medic Chem 45: 3569–3572. [DOI] [PubMed] [Google Scholar]
- Pellicciari R., Gioiello A., Macchiarulo A., Thomas C., Rosatelli E., Natalini B., et al. (2009) Discovery of 6alpha-ethyl-23(S)-methylcholic acid (S-EMCA, INT-777) as a potent and selective agonist for the TGR5 receptor, a novel target for diabesity. J Med Chem 52: 7958–7961. [DOI] [PubMed] [Google Scholar]
- Pineda Torra I., Claudel T., Duval C., Kosykh V., Fruchart J., Staels B. (2003) Bile acids induce the expression of the human peroxisome proliferator-activated receptor alpha gene via activation of the farnesoid x receptor. Mol Endocrinol 17: 259–272. [DOI] [PubMed] [Google Scholar]
- Potthoff M., Boney-Montoya J., Choi M., He T., Sunny N., Satapati S., et al. (2011) FGF15/19 regulates hepatic glucose metabolism by inhibiting the CREB-PGC-1alpha pathway. Cell Metab 13: 729–738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rao A., Haywood J., Craddock A., Belinsky M., Kruh G., Dawson P. (2008) The organic solute transporter alpha-beta, OSTalpha-OSTbeta, is essential for intestinal bile acid transport and homeostasis. Proc Natl Acad Sci U S A 105: 3891–3896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Renga B., Mencarelli A., Migliorati M., Cipriani S., D’Amore C., Distrutti E., et al. (2011) SHP-dependent and -independent induction of peroxisome proliferator-activated receptor-gamma by the bile acid sensor farnesoid X receptor counter-regulates the pro-inflammatory phenotype of liver myofibroblasts. Inflamm Res 60: 577–587. [DOI] [PubMed] [Google Scholar]
- Ridlon J., Kang D., Hylemon P. (2006) Bile salt biotransformations by human intestinal bacteria. J Lipid Res 47: 241–259. [DOI] [PubMed] [Google Scholar]
- Rizzo G., Passeri D., De Franco F., Ciaccioli G., Donadio L., Rizzo G., et al. (2010) Functional characterization of the semisynthetic bile acid derivative INT-767, a dual farnesoid X receptor and TGR5 agonist. Mol Pharmacol 78: 617–630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Russell D. (2003) The enzymes, regulation, and genetics of bile acid synthesis. Ann Rev Biochem 72: 137–174. [DOI] [PubMed] [Google Scholar]
- Safadi R., Konikoff F., Mahamid M., Zelber-Sagi S., Halpern M., Gilat T., et al. (2014) The fatty acid-bile acid conjugate aramchol reduces liver fat content in patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 12: 2085–2091 e2081. [DOI] [PubMed] [Google Scholar]
- Sattar N., Forrest E., Preiss D. (2014) Non-alcoholic fatty liver disease. BMJ 349: g4596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schaap F., Jansen P., Olde Damink S. (2015) FXR, intestinal fixer of hepatocellular carcinoma? Hepatology 61: 21–23. [DOI] [PubMed] [Google Scholar]
- Schaap F., Trauner M., Jansen P. (2014) Bile acid receptors as targets for drug development. Nat Rev Gastroenterol Hepatol 11: 55–67. [DOI] [PubMed] [Google Scholar]
- Schoonjans K., Staels B., Auwerx J. (1996) Role of the peroxisome proliferator-activated receptor (PPAR) in mediating the effects of fibrates and fatty acids on gene expression. J Lipid Res 37: 907–925. [PubMed] [Google Scholar]
- Sinal C., Tohkin M., Miyata M., Ward J., Lambert G., Gonzalez F. (2000) Targeted disruption of the nuclear receptor FXR/BAR impairs bile acid and lipid homeostasis. Cell 102: 731–744. [DOI] [PubMed] [Google Scholar]
- Speight R., Barker C., Palmer J., Kirby J. (2015) Farnesoid X receptor agonism modulates gut epithelial innate immune response. Presented at UEGW, Barcelona, Spain. [Google Scholar]
- Staels B., Rubenstrunk A., Noel B., Rigou G., Delataille P., Millatt L., et al. (2013) Hepatoprotective effects of the dual peroxisome proliferator-activated receptor alpha/delta agonist, GFT505, in rodent models of nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. Hepatology 58: 1941–1952. [DOI] [PubMed] [Google Scholar]
- Thomas C., Gioiello A., Noriega L., Strehle A., Oury J., Rizzo G., et al. (2009) TGR5-mediated bile acid sensing controls glucose homeostasis. Cell Metab 10: 167–177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Uriarte I., Fernandez-Barrena M., Monte M., Latasa M., Chang H., Carotti S., et al. (2013) Identification of fibroblast growth factor 15 as a novel mediator of liver regeneration and its application in the prevention of post-resection liver failure in mice. Gut 62: 899–910. [DOI] [PubMed] [Google Scholar]
- Vu-Dac N., Schoonjans K., Kosykh V., Dallongeville J., Fruchart J., Staels B., et al. (1995) Fibrates increase human apolipoprotein A-II expression through activation of the peroxisome proliferator-activated receptor. J Clin Invest 96: 741–750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Y., Chen W., Wang M., Yu D., Forman B., Huang W. (2008) Farnesoid X receptor antagonizes nuclear factor kappaB in hepatic inflammatory response. Hepatology 48: 1632–1643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Watanabe M., Houten S., Mataki C., Christoffolete M., Kim B., Sato H., et al. (2006) Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation. Nature 439: 484–489. [DOI] [PubMed] [Google Scholar]
- Watanabe M., Houten S., Wang L., Moschetta A., Auwerx J. (2004) Bile acids lower triglyceride levels via a pathway involving FXR, SHP, and SREBP-1C. J Clin Invest 113: 1408–1418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- West K., Zern T., Butteiger D., Keller B., Fernandez M. (2003) SC-435, an ileal apical sodium co-dependent bile acid transporter (ASBT) inhibitor lowers plasma cholesterol and reduces atherosclerosis in guinea pigs. Atherosclerosis 171: 201–210. [DOI] [PubMed] [Google Scholar]
- Williams R., Horton R. (2013) Liver disease in the UK: a Lancet commission. Lancet 382: 1537–1538. [DOI] [PubMed] [Google Scholar]
- Wu X., Ge H., Lemon B., Vonderfecht S., Baribault H., Weiszmann J., et al. (2010) Separating mitogenic and metabolic activities of fibroblast growth factor 19 (FGF19). Proc Natl Acad Sci U S A 107: 14158–14163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu X., Ge H., Lemon B., Vonderfecht S., Weiszmann J., Hecht R., et al. (2010) FGF19-induced hepatocyte proliferation is mediated through FGFR4 activation. J Biol Chem 285: 5165–5170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yao J., Zhou C., Ma X., Fu B., Tao L., Chen M., et al. (2014) FXR agonist GW4064 alleviates endotoxin-induced hepatic inflammation by repressing macrophage activation. World J Gastroenterol 20: 14430–14441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang L., Wang Y., Chen W., Wang X., Lou G., Liu N., et al. (2012) Promotion of liver regeneration/repair by farnesoid X receptor in both liver and intestine in mice. Hepatology 56: 2336–2343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang S., Wang J., Liu Q., Harnish D. (2009) Farnesoid X receptor agonist WAY-362450 attenuates liver inflammation and fibrosis in murine model of non-alcoholic steatohepatitis. J Hepatol 51: 380–388. [DOI] [PubMed] [Google Scholar]
- Zhang Y., Li S., He L., Wang F., Chen K., Li J., et al. (2015) Combination therapy of fenofibrate and ursodeoxycholic acid in patients with primary biliary cirrhosis who respond incompletely to udca monotherapy: a meta-analysis. Drug Des Devel Ther 9: 2757–2766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou X., Cao L., Jiang C., Xie Y., Cheng X., Krausz K., et al. (2014) PPARalpha-UGT axis activation represses intestinal FXR-FGF15 feedback signalling and exacerbates experimental colitis. Nat Commun 5: 4573. [DOI] [PMC free article] [PubMed] [Google Scholar]
