37
DOI: 10.1039/c6tx00376a Improved hepatic physiology in hepatic cytochrome P450 reductase null (HRN™) mice dosed orally with fenclozic acid James A. Akingbasote a , Alison J. Foster * b , Huw B. Jones b , Rhiannon David b , Nigel J Gooderham c , Ian D. Wilson c , and J. Gerry Kenna d a MRC Centre for Drug Safety Science, University of Liverpool, Liverpool, L69 3GE, United Kingdom b Drug Safety and Metabolism, Unit 310 - Darwin Building, Cambridge Science Park, Milton Road, Cambridge, CB4 0WG. c Section of Computational and Systems Medicine, Department of Surgery and Cancer Faculty of Medicine, Imperial College London, South Kensington Campus, London SW7 2AZ United Kingdom. d Current affiliation: Drug Safety Consultant, Macclesfield, United Kingdom. 1

spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

Embed Size (px)

Citation preview

Page 1: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

DOI: 10.1039/c6tx00376a

Improved hepatic physiology in hepatic cytochrome P450 reductase null

(HRN™) mice dosed orally with fenclozic acid

James A. Akingbasotea, Alison J. Foster* b, Huw B. Jonesb, Rhiannon Davidb, Nigel J

Gooderhamc, Ian D. Wilsonc, and J. Gerry Kennad

a MRC Centre for Drug Safety Science, University of Liverpool, Liverpool, L69 3GE, United

Kingdom

bDrug Safety and Metabolism, Unit 310 - Darwin Building, Cambridge Science Park, Milton Road,

Cambridge, CB4 0WG.

c Section of Computational and Systems Medicine, Department of Surgery and Cancer

Faculty of Medicine, Imperial College London, South Kensington Campus, London

SW7 2AZ United Kingdom.

d Current affiliation: Drug Safety Consultant, Macclesfield, United Kingdom.

Email addresses: [email protected]; [email protected];

[email protected]; [email protected] ; [email protected];

[email protected]; [email protected]

Corresponding author: * Alison Foster: Drug Safety and Metabolism, Unit 310 - Darwin Building,

Cambridge Science Park, Milton Road, Cambridge, CB4 0WG; E-mail:

[email protected]; tel. : +441625234713

Running Title: HEPATIC EFFECTS OF FENCLOZIC ACID IN HRNTM MICE

1

Page 2: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

2

Page 3: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

Abstract

Hepatic NADPH-cytochrome P450 oxidoreductase null (HRN™) mice exhibit no functional

expression of hepatic cytochrome P450 (P450) when compared to wild type (WT) mice, but have

normal hepatic and extrahepatic expression of other biotransformation enzymes. We have assessed

the utility of HRN™ mice for investigation of the role of metabolic bioactivation in liver toxicity

caused by the nonsteroidal anti-inflammatory drug (NSAID) fenclozic acid. In vitro studies revealed

significant NADPH-dependent (i.e. P450-mediated) covalent binding of [14C]-fenclozic acid to liver

microsomes from WT mice and HRN™ mice, whereas no in vitro covalent binding was observed in

the presence of the UDP-glucuronyltransferase cofactor UDPGA. Oral fenclozic acid administration

did not alter the liver histopathology or elevate the plasma liver enzyme activities of WT mice, or

affect their hepatic miRNA contents. Livers from HRN™ mice exhibited abnormal liver

histopathology (enhanced lipid accumulation, bile duct proliferation, hepatocellular degeneration,

necrosis, inflammatory cell infiltration) and plasma clinical chemistry (elevated alanine

aminotransferase, glutamate dehydrogenase and alkaline phosphatase activities). Modest apparent

improvements in these abnormalities were observed when HRN™ mice dosed orally with fenclozic

acid for 7 days at 100 mg/kg/day. Previously we observed more marked effects on liver

histopathology and integrity in HRN™ mice dosed orally with the NSAID diclofenac for 7 days at 30

mg/kg/day. We conclude that HRN™ mice are valuable for assessing P450-related hepatic drug

biotransformation, but not for drug toxicity studies due to underlying liver dysfunction. Nonetheless,

HRN™ mice may provide novel insights into the role of inflammation in liver injury, thereby aiding

its treatment.

Key words: Fenclozic acid, hepatotoxicity, intestinal toxicity, biotransformation, covalent binding,

cytochrome P450.

3

Page 4: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

Introduction

HRN™ mice lack the gene encoding hepatic NADPH-cytochrome P450 oxidoreductase (CYP P450

reductase; POR; EC 1.6.2.4)1, which catalyses the transfer of electrons from cytochrome P450 (P450)

and other endoplasmic reticulum enzymes which use NADPH as co-factor.2,3 Hence HRN™ mice are

deficient in hepatic P450 activities, whereas the activities of other hepatic drug metabolizing enzymes

and extra-hepatic P450 enzymes are unaffected.1,4 In vivo biotransformation studies have shown that

HRN™ mice can provide valuable insights into the role played by hepatic P450s in xenobiotic

biotransformation. For example, formation of the toxic quinoneimine metabolite was not observed

when HRN™ mice were dosed with acetaminophen (paracetamol).1 This model may also prove

especially useful in circumstances where both oxidative P450-mediated metabolism and conjugation

reactions result in the production of reactive metabolites that might cause toxicity. For example, the

nonsteroidal anti-inflammatory drug (NSAID) diclofenac causes hepatotoxicity in patients and is

metabolised in vivo via both P450-mediated oxidative and UDP-glucuronyltransferase (UGT)-

mediated conjugative routes in wild type mice5, humans and other animal species.6 Currently it is

unclear whether oxidation or conjugation of diclofenac, or a combination of both, is responsible for

the hepatotoxicity caused by the drug in patients.

When administered to HRN™ mice, diclofenac was metabolized overwhelmingly to conjugates

(mainly to taurine and glucuronic acid), with little evidence of oxidative metabolism.7 In a previous

study8 we undertook toxicological studies in wild type (WT) and HRN™ mice dosed orally with

diclofenac to assess whether use of this model might provide novel insights into the possible role

played by conjugation of diclofenac to chemically reactive acyl glucuronides in the liver toxicity

observed in human patients. HRN™ mice treated with dose vehicle alone exhibited enhanced hepatic

lipid accumulation and degenerative liver histopathologies that primarily affected bile ducts and

periportal hepatocytes, plus markedly elevated activities in plasma of various liver-derived enzymes,

which were not present in livers from dose vehicle treated WT mice. Unexpectedly, the severities of

4

Page 5: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

the abnormal liver histopathology and plasma liver enzyme findings in HRN™ mice were reduced

markedly when HRN™ mice were dosed orally with diclofenac for 7 days at 30 mg/kg/day.8

A possible explanation for the positive effect of diclofenac administration to HRN™ mice on liver

pathophysiology is via a reduction/inhibition of hepatic inflammation.9,10 To gain further insight into

whether this is a plausible hypothesis, and determine whether the effects were unique to diclofenac or

were seen with other drugs in this class, in the present studies we have explored tissue histopathology

and plasma clinical chemistry in HRN™ and WT mice dosed orally for seven days with dose vehicle

alone or fenclozic acid (Myalex®). Fenclozic acid is a carboxylic acid NSAID with potent analgesic

and antipyretic activities11 whose clinical development was terminated due to liver injury observed in

clinical trials12 and which also is metabolised to chemically reactive metabolites.13,14 Recently it has

recently been reported that miRNA could be sensitive and informative biomarkers of hepatotoxicity;

for example, paracetamol induced toxicity in mice rapidly resulted in a decline in hepatic miRNAs

such as miR-122 and miR-192.15 This observation prompted us also to investigate whether altered

amounts of hepatic miRNAs were evident when HRN™ and WT mice were treated with fenclozic

acid.

Materials and methods

Chemicals

FluothaneTM, fenclozic acid and [14C]-fenclozic acid (purity >99%, specific activity 60.5 mCi/mmol)

were obtained from AstraZeneca. PierceTM BCA Protein Assay Kit was from Thermo Scientific

(Rockford, IL). Formic acid was from VWR International Ltd (Poole, UK). The EnzyChrom™

Alanine Transaminase Assay Kit was from BioAssay Systems (Hayward, CA). The bile acid reagent

was from Diazyme Laboratories (Dresden, Germany) and other clinical chemistry reagents were from

Roche Diagnostics (Burgess Hill, UK). All other reagents and solvents were from Sigma-Aldrich

5

Page 6: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

Company Ltd. (Poole, UK) or Fisher Scientific (Loughborough, UK) and were of HPLC grade or

higher.

In vivo studies

The experimentation, transportation and care of the mice were performed in accordance with

approved project license, the relevant national guidelines (UK Animals and Scientific Procedures Act,

1986) and in compliance with AstraZeneca regulations on the ethical treatment of animals. All

experiments were approved by AstraZeneca’s ethical committee. Statistical analysis was undertaken

to determine the minimum number of animals to give 80% power to detect fold changes of

approximately 2-3 fold in ALT/ALP via pairwise comparison of groups with 5% significance level,

using a t-test approach (2-sided) via ANOVA. The animals were individually housed in standard

conditions with freely available access to drinking water and food. All animals were thoroughly

examined before dosing commenced and at weekly intervals from the start of dosing through to

termination. They were also visually inspected at least once daily after dosing, paying particular

attention up to 3 hours after dosing.

The dose levels and route of administration of fenclozic acid were based on information available in

the literature. 12,16 Male C57/Bl6 WT mice (8-10 weeks old (21-25 g), Charles River, Margate, UK)

were treated daily for 7 days, by oral gavage, with dose vehicle alone (0.1 M sodium phosphate

buffer, pH 7.7; Group 1), or fenclozic acid at doses of 50 or 100 mg/kg/day (in the same vehicle), 10

mL/kg (Groups 2 and 3 respectively; n=5 per group). Male HRN™ mice (4–11 weeks old (20-27 g);

Taconic Farms Inc. Germantown, NY, USA) were treated by daily oral gavage for 7 days with

fenclozic acid at a dose of 100 mg/kg/day (Group 5; n=5), or with equivalent volumes of dose vehicle

alone (Group 4; n=4). The HRN™ mice had been developed using C57BL/6J background strain mice

[1]. Blood samples (50 L) were obtained from the animals by tail bleeding on days -1, 2 and 4 of

the study and plasma ALT activities were determined. At 24 h after the final administration of

fenclozic acid or dose vehicle, animals were euthanized under fluothaneTM anaesthesia. Terminal

blood samples were collected for immediate analysis of plasma alanine aminotransferase (ALT),

6

Page 7: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

alkaline phosphatase (ALP) and glutamate dehydrogenase (GLDH), which was undertaken using a

Roche Modular P800 Chemistry Analyser (Roche Diagnostics Ltd., Burgess Hill, UK). Additionally,

ALT activities of plasma samples were determined using an EnzyChrom™ Alanine Transaminase

Assay Kit (Bioassays Systems, Hayward, CA).

The liver, intestines, oesophagus and stomach/duodenum were examined for any abnormalities and

weighed and tissue samples were processed to paraffin blocks using standard histologic procedures.

Sections were cut and were stained with haematoxylin and eosin (H&E) for histopathologic

examination using light microscopy. The severities of tissue histopathology findings were scored

qualitatively, using the following scheme: Grade 1 = Minimal / very few / very small; Grade 2 =

Slight / few / small; Grade 3 = Moderate / moderate number / moderate size; Grade 4 = Marked /

many / large; Grade 5 = Massive / extensive number / extensive size.

Statistical analysis of data was performed by paired or unpaired t-testing, using GraphPad Prism

version 4.03 (GraphPad Software, San Diego, CA).

To quantify hepatic miRNAs, total RNA was extracted from liver tissue using the MiRVana PARIS

kit (Life Technologies, Paisley, UK) and was quantified using a NanoPhotometer (Implen GmbH,

Munchen, Germany). The ratios A260/A280 and A260/A230 were used to assess RNA quality. Total

RNA was reverse transcribed using the TaqMan MicroRNA Reverse Transcription Kit (Life

Technologies, Paisley, UK). Mature miRNA TaqMan assays (Life Technologies) were used to

generate cDNA, which was amplified using the Taqman 2x Universal PCR master mix, No AmpErase

UNG (Life Technologies), with each reaction performed in triplicate. The qPCR data were analysed

using the ABI 7500 Sequence Detection System (Life Technologies) and the comparative Ct Method

(ΔΔCT Method) [17]. Calibration was based on the expression of snoRNA, as per manufacturer’s

instructions.

Preparation of liver microsomes and determination of microsomal cytochrome c reductase activity

7

Page 8: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

The preparation of liver microsomes from both WT and HRN™ mouse livers was undertaken as

described by Akingbasote et al.8 Protein concentrations were determined using the Pierce BCA

Protein Assay Kit®.

In vitro covalent binding (CVB) of [14C]-fenclozic acid to liver microsomes

Determination of CVB for [14C]-fenclozic acid was performed as described by Day et al.18 Liver

microsomes (1 mg/mL final protein concentration) were incubated at 37 °C with 10 M [14C]-

fenclozic acid in 0.1 M phosphate buffer supplemented with 10 mM MgCl2, pH 7.0, in the presence or

absence of NADPH (2 mM) or UDPGA (4 mM). In some experiments, 1-aminobenzotriazole (ABT;

1 mM) was also added to the incubation mixture. The final assay volume was 500 μL. After

incubation for 0 and 60 min, aliquots (200 μL) were mixed with 300 μL of chilled acetone and then a

further 500 μL acetone was added. Precipitated proteins were collected onto filter disks using a

Brandel 96-sample cell harvester (Gaithersburg MD, USA), washed with 80% methanol, then

protein-bound [14C] was quantified by liquid scintillation counting as described by Foster et al. 19 Non-

specific binding (NSB) was determined by incubating microsomes in the absence of fenclozic acid

for 1 hour at 37 °C, then ‘back-adding’ 10 µM [14C]-fenclozic acid immediately prior to processing

the samples. Levels of CVB were expressed as pmol equivalents/mg protein.

Results

Abnormal liver histopathology and plasma clinical chemistry of vehicle treated HRN™ mice

WT mice which were dosed orally for 7 days with dose vehicle alone exhibited no abnormalities in

liver histology, other than minimal extents of non-periportal hepatocyte necrosis with inflammatory

cell infiltration and minimal mixed inflammatory cell aggregates (Figure 1A, Table 1). The

frequencies and severities of these findings were in line with those observed historically in WT

8

Page 9: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

control mice. As we have reported previously,8 a variety of histopathological findings were observed

in livers from dose vehicle treated HRN™ mice that were not evident in livers from WT mice.

Elevated numbers and sizes of hepatocellular fat vacuoles, increased extents of bile duct proliferation

and periportal hepatocyte necrosis with accompanying inflammatory cell infiltration were observed in

each of four livers from vehicle-treated HRN™ mice, while a few necrotic hepatocyte foci were also

observed in one liver (Figure 1A and C and Table 1). Furthermore, plasma from mice exhibited

markedly elevated ALT and GLDH activities and less markedly elevated ALP activity, when

compared with plasma from vehicle treated WT mice (Table 2).

Hepatic effects of oral administration of fenclozic acid to HRN™ and WT mice

Oral daily administration of fenclozic acid at doses of 50 or 100 mg/kg/day to WT mice did not result

in any compound-related alterations in liver histopathology (Figure 1C and Table 1), in hepatic

expression of miR-122 or in plasma ALT or GLDH activities (Table 2). However, a dose dependent

reduction in plasma ALP activity was observed in fenclozic acid treated WT mice (Table 2).

When compared with HRN™ mice treated with dose vehicle alone, HRN™ mice dosed orally for 7

days with fenclozic acid at 100 mg/kg/day exhibited less severe bile duct proliferation and periportal

hepatocyte necrosis with accompanying inflammatory cell infiltration (Figure 1, compare panels D

and C; Table 1). The modest apparent reduction in severity of liver histopathology in fenclozic acid

dosed HRN™ mice was consistent with plasma clinical chemistry data, which demonstrated a

statistically significant (p<0.05) reduction in plasma ALP activity and by reductions in plasma ALT

and GLDH activities that did not achieve statistical significance (Table 2). However, the relative

expression of hepatic miR-122 in these animals was unaffected by fenclozic acid treatment (Table 2).

Upper GI histopathology in fenclozic acid treated HRN™ and WT mice

Neither HRN™ nor WT mice dosed orally for 7 days with dose vehicle alone exhibited abnormalities

in upper gastrointestinal (GI) histopathology, apart from minimal or slight oesophageal muscle cell

alterations and minimal glandular epithelial cysts at the stomach limiting ridge in several animals

9

Page 10: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

(Table 3). The frequencies and severities of these upper GI findings in these animals were in line

with those observed historically in WT control mice.

No alterations in upper GI histopathology were observed in WT mice dosed with fenclozic acid at 50

mg/kg/day for 7 days. However, three of four WT mice dosed with fenclozic acid at 100 mg/kg/day

exhibited evidence of stomach pyloric erosion and pyloric gastritis was observed in one of these

animals (Table 3). Similar stomach lesions were observed in three of five HRN™ mice dosed with

fenclozic acid at 100 mg/kg/day (Table 3).

In vitro CVB of [14C]-fenclozic acid to liver microsomes from WT and HRN™ mice

CVB of radiolabel to proteins (>170 pmol/mg protein) was observed when [14C]-fenclozic acid was

incubated for 60 min with liver microsomes from WT mice that were supplemented with NADPH

(Table 4). In contrast, CVB to proteins was not observed when equivalent incubations were

undertaken either in the absence of NADPH, or in the presence of UDPGA in place of NADPH.

Similarly, no CVB to proteins was seen when the incubation with NADPH was undertaken for 0 min

or when [14C]-fenclozic acid was “back-added” to microsomes that had been incubated with NADPH

for 60 min. A marked and statistically significant reduction in the amount of NADPH-dependent

CVB to proteins (to <50 pmol/mg protein) was observed when [14C]-fenclozic acid was incubated for

60 min with WT liver microsomes in the presence of the CYP inhibitor ABT. Detectable CVB was

also observed when [14C]-fenclozic acid was incubated for 60 min with liver microsomes from

HRN™ mice in the presence of NADPH. However, the amount of CVB to HRN™ mouse liver

microsomes was approximately 6-fold lower than the amount of CVB to WT liver microsomes.

Furthermore, the CVB to HRN™ mouse liver microsomes was not altered in the presence of ABT. A

detectable amount of CVB was not observed when [14C]-fenclozic acid was incubated for 60 min with

liver microsomes from HRN™ mice in the presence of UDPGA (Table 4).

10

Page 11: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

Discussion

The clinical development of fenclozic acid was terminated due to liver injury in clinical trials, 12 which

was not observed in any of the animal species used in its preclinical safety testing. In order to gain

insight into the mechanisms responsible for the human-specific hepatotoxicity of this drug, previously

we investigated its potential for bioactivation in vitro and in vivo. The in vitro studies were

undertaken using isolated human hepatocytes13 and also rat, dog and human liver microsomes.14

These revealed relatively high levels of NADPH-dependent CVB to liver proteins, implying that

oxidative bioactivation had occurred. In addition, in vivo metabolite profiling studies identified a

variety of oxidized and glutathione-derived metabolites in urine and bile from fenclozic acid treated

rats20 and WT mice (Pickup et al., in preparation). These findings provided evidence that metabolic

bioactivation of fenclozic acid also occurs in vivo and support our suggestion13,14 this process could be

responsible for the hepatotoxicity caused by the drug in humans in vivo. Such oxidative metabolites

were not seen in urine or faeces obtained from HRN™ mice that had been dosed with fenclozic acid,

where its metabolism was via the carboxylic acid moiety and resulted in the formation of an acyl

glucuronide as well as glycine and taurine conjugates.7 These various metabolic pathways are

summarised in Figure 2.

The NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we

observed in our present investigations are consistent with these earlier studies. However, the small but

statistically significant amount of CVB to proteins in liver microsomes from HRN™ mice, which was

unaffected by addition of the P450 inhibitor ABT, was a novel and unexpected finding. A plausible

explanation is that fenclozic acid is bioactivated in livers from HRN™ mice by a novel mechanism,

which is distinct from the P450 mediated process evident in livers of rats and WT mice and shown in

Figure 2. Additional studies will be required to characterise the responsible enzyme, although a

possible candidate is flavin monoxygenase (FMO).21 Since we observed no evidence of compound

related liver toxicity following in vivo administration of fenclozic acid to either WT or HRN™ mice,

our current investigations were unable to provide insight into the possible relevance of metabolic

bioactivation to its human hepatotoxicity.

11

Page 12: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

Similar patterns and severities of adverse stomach pathology were observed when both WT and

HRNTM mice dosed orally with fenclozic acid for 7 days at 100 mg/kg/day, which was not observed in

the vehicle treated animals. The likely explanation for this toxicity is impaired wound healing which

arises as a consequence of cyclooxygenase inhibition, since such toxicity is a common finding in

animals and humans dosed with numerous other NSAIDs.22 However, whereas vehicle-treated

HRNTM mice exhibited no abnormalities in stomach and gastrointestinal pathology when compared

with WT mice, as we have noted previously8 these animals have a variety of underlying abnormalities

in liver pathology and function (resulting in markedly abnormal plasma clinical chemistry).

Therefore whilst HRNTM mice are useful for dissecting out the role of hepatic P450 dependent

processes in drug metabolism, they appear to be less well suited to investigations of mechanisms of

hepatic drug toxicity.

Interestingly, we found no changes in the relative expression of hepatic miR-122 in either mouse

strain, irrespective of treatment. Paracetamol-induced hepatotoxicity has been reported to reduce the

expression of the abundant hepatic enriched miR-122 with a concomitant release of miR-122 into the

plasma.15 This finding prompted the proposal that changes in miRNA expression could be sensitive

and informative biomarkers of drug-induced liver injury. Although the data reported herein fail to

support this hypothesis, there are significant experimental differences between our study and

paracetamol induced toxicity. Specifically, the acute response to paracetamol toxicity and the slower

onset of hepatotoxicity induced by fenclozic acid in humans.12 In addition, the hepatic miR-122 levels

were assessed after 7 days of fenclozic acid administration to mice. It is also pertinent to note that the

hepatic miR-122 levels were unaffected by fenclozic acid treatment of the HRNTM mice, despite the

apparent drug-induced improvement of hepatic histology; again hepatic levels of miR-122 were

assessed after 7 days of treatment. This lack of effect on hepatic miR-122 expression in both mouse

strains may be a reflection of the regulatory function of the miRs and their role in maintaining

homeostasis. In this context, it would be interesting to measure circulating and hepatic miR-122

expression during the first 24 h of treatment of mice with fenclozic acid.

12

Page 13: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

The abnormal and degenerative liver histopathology and high plasma activities of liver-derived

enzymes present in vehicle treated HRN™ mice are likely to be a consequence of impaired lipid

metabolism and bile acid homeostasis, which arise due to the absence of hepatic P450 activities in

these animals.1-3 Previously we reported a marked reduction in both degenerative liver

histopathology, and plasma liver-derived enzyme activities (ALT, GLDH and ALP), when HRN™

mice were dosed orally for 7 days with diclofenac.8 Our present results demonstrate more modest

apparent improvements in plasma liver-derived enzyme activities and liver histopathology when

HRN™ mice were dosed orally for 7 days with a second NSAID, fenclozic acid, at a dose of 100

mg/kg/day. Additional experiments are required to clarify whether the apparent effects observed in

HRN™ mice dosed with fenclozic acid can indeed be attributed to compound administration.

Nonetheless, both diclofenac and fenclozic acid have, by design, anti-inflammatory properties and are

potent inhibitors of the enzyme cyclooxygenase, which converts arachidonic acid to prostanoid

inflammatory mediators.23 Inflammatory responses in the liver have been found to mediate hepatic

injury caused by endotoxins and various other xenobiotics.24,25 Hepatic inflammation involves

activation of Kupffer cells, which release biologically active compounds (including cytokines, growth

factors, arachidonic-acid-derived inflammatory mediators and lysosomal enzymes) that cause

hepatocyte necrosis and liver inflammation. Inhibition of cyclooxygenase in Kupffer cells has been

found to impair their activation and it has been proposed that NSAIDs may provide a promising

strategy for mitigation of the liver inflammation, and its resulting liver pathologies, that occurs in

human non-alcoholic fatty liver disease (NAFLD)26,27 and other liver types of liver disease.28

Therefore our current findings are consistent with our previous proposal8 that inflammatory processes

may play important roles in the liver injury evident in HRN™ mice, and that the positive effects of

diclofenac on the liver histopathology and function of these animals could be due to the anti-

inflammatory activity of the drug. It is also conceivable that the liver abnormalities evident in

HRN™ mice might provide a useful model system for exploration of mechanisms of NAFLD and

other clinically important human liver diseases, which could aid the assessment of the potential

therapeutic value of NSAIDs or other novel strategies for treatment of liver inflammation.

13

Page 14: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

Acknowledgements

We thank Jonathan Greenall, Elizabeth Johnson and Peter Cotton for their excellent technical

assistance, James Noakes for his directing of the in vivo study and Marie South for her statistical

advice.

Conflicts of interest

The authors declare no conflicts of interest.

Abbreviations

HRNTM, hepatic cytochrome P450 reductase null mice; WT, wild type C57BL6J mice; P450,

cytochrome P450; NADPH, reduced nicotinamide adenine dinucleotide phosphate; UDPGA, uridine

5'-diphospho-glucuronic acid; ALT, alanine aminotransferase; AST, aspartate aminotransferase;

GLDH, glutamate dehydrogenase; ALP, alkaline phosphatase; NAFLD non-alcoholic fatty liver

disease, miR-122, liver specific miRNA 122; snoRNA, small nucleolar RNAs.

14

Page 15: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

References

1. C.J. Henderson, D.M. Otto, D. Carrie, M.A. Magnuson, A.W. Mclaren, I. Rosewell and

C.R. Wolf, Inactivation of the hepatic cytochrome P450 system by conditional deletion

of hepatic cytochrome P450 reductase, J. Biol. Chem., 2003, 278, 13480-6.

2. X.J. Wang, M. Chamberlain, O. Vassieva, C.J. Henderson and C.R. Wolf, Relationship

between hepatic phenotype and changes in gene expression in cytochrome P450

reductase (POR) null mice, Biochem J., 2005, 388, 857-67.

3. D.M. Mutch, B. Klocke, P. Morrison, C.A. Murray, C.J. Henderson, M. Seifert and G.

Williamson, The disruption of hepatic cytochrome p450 reductase alters mouse lipid

metabolism, J. Proteome Res., 2007, 6, 3976-84.

4. M. Stiborová, V.M. Arlt, C.J. Henderson, C.R. Wolf, V. Kotrbová, M. Moserová, J.

Hudecek, D.H. Phillips and E. Frei, Role of hepatic cytochromes P450 in bioactivation

of the anticancer drug ellipticine: studies with the hepatic NADPH:cytochrome P450

reductase null mouse, Toxicol. Appl. Pharmacol., 2008, 226(3), 318-27.

5. S. Sarda, C. Page, K. Pickup, T. Schulz-Utermoehl and I. Wilson, Diclofenac

metabolism in the mouse: Novel in vivo metabolites identified by high performance

liquid chromatography coupled to linear ion trap mass spectrometry, Xenobiotica, 2012,

42, 179-194.

6. U.A. Boelsterli, Diclofenac-induced liver injury: a paradigm of idiosyncratic drug

toxicity, Toxicol. Appl. Pharmacol., 2003, 192, 307-22.

7. K. Pickup, J. Wills, A. Rodrigues, H.B. Jones, C. Page, S. Martin, S. Sarda and I.

Wilson, The metabolic fate of [14C]-fenclozic acid in the hepatic reductase null (HRN)

mouse, Xenobiotica, 2014, 44, 164-73.

15

Page 16: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

8. J.A. Akingbasote, A.J. Foster, I. Wilson, S. Sarda, H.B. Jones and J.G. Kenna, Hepatic

effects of repeated oral administration of diclofenac to hepatic cytochrome P450

reductase null (HRN™) and wild-type mice, Arch. Toxicol., 2016, 90, 853–62.

9. N. Alkhouri, L.J. Dixon and A.E. Feldstein, Lipotoxicity in non-alcoholic fatty liver

disease: not all lipids are created equal, Expert Rev. Gastroenterol. and Hepatol., 2009,

3(4), 445-51.

10. V. Vaish and S.N. Sanyal, Chemopreventive effects of NSAIDs on cytokines and

transcription factors during the early stages of colorectal cancer. Pharmacol. Rep., 2011,

63(5), 1210-21.

11. T.M. Chalmers, J.H. Kellgren and D.S. Platt, Evaluation in man of fenclozic acid (I.C.I.

54,450: Myalex), a new anti-inflammatory agent. II. Clinical trial in patients with

rheumatoid arthritis, Ann. Rheum. Dis., 1969, 28(6), 595-601.

12. S. Alcock, An anti-inflammatory compound: non-toxic to animals but with an adverse

action in man. Proc. Eur. Soc. Study Drug Toxic, 1971, 12, 7.

13. R.A. Thompson, E.M. Isin, Y. Li, L. Weidolf, K. Page, I. Wilson, S. Swallow, B.

Middleton, S. Stahl, A.J. Foster, H. Dolgos, R. Weaver and J.G. Kenna, In Vitro

Approach to Assess the Potential for Risk of Idiosyncratic Adverse Reactions Caused

by Candidate Drugs, Chem. Res. Toxicol., 2012, 25(8), 1616-1632.

14. A.V. Rodrigues, H.E. Rollison, S. Martin, S. Sarda, T. Schulz-Utermoehl, S. Stahl, F.

Gustafsson, J. Eakins, J.G. Kenna and I.D. Wilson, In vitro exploration of potential

mechanisms of toxicity of the human hepatotoxic drug fenclozic acid, Archives of

Toxicology, 2013, 87, 1569-79.

15. K. Wang, S. Zhang, B. Marzolf, P. Troisch, A. Brightman, Z. Hu, L.E. Hood and D.J.

Galas, Circulating microRNAs, potential biomarkers for drug-induced liver injury,

Proceedings of the National Academy of Sciences USA, 2009, 106(11), 4402-4407.

16

Page 17: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

16. B.B. Newbould, The pharmacology of fenclozic acid (2-(4-chlorophenyl)-thiazol-4-

ylacetic acid; I.C.I. 54,450; ‘Myalex’); a new compound with anti-inflammatory,

analgesic and antipyretic activity, Br. J. Pharmacol., 1969, 35(3), 487-97.

17. K.J. Livak and T.D. Schmittgen, Analysis of relative gene expression data using real-

time quantitative PCR and the 2(-Delta Delta C(T)) Method, Methods, 2001, 25(4),

402-8.

18. S.H. Day, A. Mao, R. White, T. Schulz-Utermoehl, R. Miller and M.G. Beconi, A semi-

automated method for measuring the potential for protein covalent binding in drug

discovery, J. Pharmacol. Toxicol. Methods, 2005, 52, 278-285.

19. A.J. Foster, L.H. Prime, F. Gustafsson, D.G. Temesi, E.M. Isin, J. Midlöv, N.

Castagnoli Jr. and J.G. Kenna, Bioactivation of the cannabinoid receptor antagonist

rimonabant to a cytotoxic iminium ion metabolite, Chem. Res. Toxicol., 2013, 26(1),

124-35.

20. S. Martin, E.M. Lenz, W. Keene and M.R. Clench, Identification of the reactive

metabolites of fenclozic acid in bile duct cannulated rats, Anal. Chem., 2014, 86(22),

11281-9.

21. R.S. Foti and D.K. Dalvie, Cytochrome P450 and Non-Cytochrome P450 Oxidative

Metabolism: Contributions to the Pharmacokinetics, Safety, and Efficacy of

Xenobiotics, Drug Metab. Dispos., 2016, 44(8), 1229-45.

22. H. Mizuno H, C. Sakamoto, K. Matsuda, K. Wada, T. Uchida, H. Noguchi, T.

Akamatsu and M. Kasuga, Induction of cyclooxygenase 2 in gastric mucosal lesions

and its inhibition by the specific antagonist delays healing in mice, Gastroenterology,

1997, 112, 387-397.

23. T.J. Gan, Diclofenac: an update on its mechanism of action and safety profile, Curr.

Med. Res. Opin., 2010, 26, 1715-31.

17

Page 18: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

24. J. Claria and E. Titos, Kupffer cell. Gastroenterol. Hepatol., 2004, 27, 264-73.

25. M.D. Wheeler, Endotoxin and Kupffer cell activation in alcoholic liver disease, Alcohol

Res. Health, 2003, 27, 300-6.

26. A. Planaguma, J. Claria, R. Miquel, M. Lopez-Parra, E. Titos, J.L. Masferrer, V.

Arroyo, J. Rodes, The selective cyclooxygenase-2 inhibitor SC-236 reduces liver

fibrosis by mechanisms involving non-parenchymal cell apoptosis and PPARgamma

activation, Faseb J., 2005, 19(9), 1120-2.

27. B.J. Park, Y.J. Lee and H.R. Lee, Chronic liver inflammation: clinical implications

beyond alcoholic liver disease, World J. Gastroenterol., 2014, 20(9), 2168-75.

28. N. Ferre and J. Claria, New insights into the regulation of liver inflammation and

oxidative stress, Mini Rev. Med. Chem. 2006, 6, 1321-1330.

18

Page 19: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

Table 1. Summary of effects of fenclozic acid administration on liver pathology in wild type and HRN™ mice

Histopathology finding Severity Number of WT mice affected Number of HRN™ mice affected

Vehicle

control

(n=5)

50 mg/kg/day

fenclozic acid

(n=5)

100 mg/kg/day

fenclozic acid

(n=5)

Vehicle control

(n=4)

100 mg/kg/day

fenclozic acid

(n=5)

Hepatocellular fat vacuolation Grade 2 0 0 0 0 0

Grade 3 0 0 0 4 4

Grade 4 0 0 0 0 1

Multifocal hepatocellular

necrosis

Grade 2 0 0 0 1 1

Bile duct proliferation Grade 1 0 0 0 0 2

Grade 2 0 0 0 1 1

Grade 3 0 0 0 3 2

Periportal hepatocyte necrosis/

inflammatory cell infiltration

Grade 1 0 0 0 0 3

Grade 2 0 0 0 4 1

Hepatocyte necrosis with

inflammatory cell infiltration

Grade 1 1 0 0 2 4

Grade 2 0 0 0 0 1

Mixed inflammatory cell

aggregates

Grade 1 2 3 3 0 0

19

Page 20: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

Table 2. Plasma ALT, GLDH, ALP and miR-122 in wild type and HRN mice treated with dose

vehicle or fenclozic acid. Values are means ± SD.

Parameter WT mice treated with: HRN™ mice treated with:

Dose vehicle

(n=5)

Fenclozic acid,

100 mg/kg (n=5)

Dose vehicle

(n=4)

Fenclozic acid,

100 mg/kg (n=5)

ALT (IU/L) 37.6 ± 12.6 66.4 ± 25.8 1,764.0 ± 859.7 1,131.6 ± 1056.3

AST (IU/L) 56.0 ± 21.5 71.2 ± 22.3 1,153.0 ± 656.2 965.6 ± 1,022.7

ALP (IU/L) 162.4 ± 35.1 128.0 ± 24.7 444.0 ± 72.7 325.6 ± 31.1 *

GLDH (IU/L) 15.2 ± 9.8 16.8 ± 5.2 521.5 ± 332.08 174.4 ± 152.8

miR-122

(relative expression)

1.0 0.96 1.0 0.86

* p<0.05, compared with vehicle control value.

20

Page 21: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

Table 3. Effects of fenclozic acid administration on gastrointestinal histopathology of wild type and HRN™ mice

Organ Histopathology

finding

Severity Number of WT mice affected Number of HRN™ mice

affected

Vehicle

control

(n=5)

50 mg/kg/day

fenclozic acid

(n=5)

100 mg/kg/day

fenclozic acid

(n=5)

Vehicle

control

(n=4)

100 mg/kg/day

fenclozic acid

(n=5)

Oesophagus No abnormalities 0 2 0 4 3 3

Adjacent muscle

degeneration/

necrosis/inflammation

Grade 1 2 0 0 1 0

Grade 2 1 0 1 0 1

Stomach No abnormalities 4 4 1a 4 2

Pyloric ulceration Grade 2 0 0 0 0 1

Pyloric erosion Grade 1 0 0 2 0 0

Grade 2 0 0 1 0 2

Pyloric gastritis Grade 1 0 0 1 0 0

Grade 2 0 0 0 0 2

Glandular epithelial

cyst(s) at limiting ridge

Grade 2 1 1 0 0 0

21

Page 22: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

a Tissues from 4 animals were evaluated.

22

Page 23: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

Table 4. In vitro CVB of [14C]-fenclozic acid to liver microsomes from wild type mice (WT

microsomes) but not HRN™ mice (HRNTM microsomes). Incubations were undertaken for 0 min (T =

0) or 60 min (T = 60), in the absence (- NADPH) or presence (+ NADPH) of NADPH, in the presence

of NADPH plus ABT (+ NADPH, + ABT), or in the presence of UDPGA (+ UDPGA). Back added =

[14C]-fenclozic acid was added immediately prior to processing the samples. Mean values ± SD from

three independent experiments, each conducted in duplicate, are shown.

pmol equivalents/mg protein

WT microsomes HRNTM microsomes

Mean SD Mean SD

Mic -NADPH, T=0 7.7 2.3 6.0 0.8

Mic -NADPH, T=60 9.7 1.9 9.8 1.7

Mic +NADPH, T=0 19.4 4.6 9.0 2.9

Mic +NADPH, T=60 187.7 * 30.4 32.2 * 7.0

Mic + ABT, T=0 9.7 1.7 10.4 0.0

Mic + ABT, T=60 39.5 # 10.0 29.2 4.2

Mic +UDPGA, T=0 7.1 1.9 8.2 1.9

Mic +UDPGA, T=60 10.4 2.3 9.2 2.8

Back added 33.0 8.5 16.3 4.6

*significantly elevated vs. NADPH background value (+NADPH, T=0); p<0.001#significantly reduced vs. CVB in absence of ABT (+NADPH, T=60); p<0.001

23

Page 24: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

Figure 1. Liver histopathology in wild type (A,B) and HRN™ (B,D) mice treated for 7 days with

dose vehicle (A,B) or 100 mg/kg/day fenclozic acid (C,D). Representative photomicrographs from

formalin fixed and H&E stained liver sections at x 20 magnification are shown. The positions of

centrilobular (CL) and periportal (PP) hepatocytes are indicated.

PPCL

PPCL

C. HRN + vehicle

D. HRN + fenclozic acid

CLPP

CLPP

A. WT + vehicle

B. WT + fenclozic acid

24

Page 25: spiral.imperial.ac.uk · Web viewThe NADPH-dependent CVB of radiolabelled fenclozic acid to WT liver microsomal proteins that we observed in our present investigations are consistent

Figure 2. Metabolism of fenclozic acid. FMO, flavin monooxygenase; CYP, cytochrome P450; UGT,

UDP-glucuronyltransferase; GSH, reduced glutathione.

25