Urinary Elimination of Bile Acid Glucuronides under Severe ...

13
Research Article Urinary Elimination of Bile Acid Glucuronides under Severe Cholestatic Situations: Contribution of Hepatic and Renal Glucuronidation Reactions Martin Perreault, 1 Ewa Wunsch , 2 Andrzej BiaBek, 2 Jocelyn Trottier, 1 Mélanie Verreault, 1 Patrick Caron, 1 Guy G. Poirier, 3 Piotr Milkiewicz , 4 and Olivier Barbier 1 1 Laboratory of Molecular Pharmacology, CHU-Qu´ ebec Research Centre and the Faculty of Pharmacy, Laval University, Quebec City, QC, Canada 2 Translation Medicine Group, Pomeranian Medical University, Szczecin, Poland 3 Proteomics Platform, Qu´ ebec Genomic Center, CHU-Qu´ ebec Research Centre and Faculty of Medicine, Laval University, Quebec City, QC, Canada 4 Liver and Internal Medicine, Medical University of Warsaw, Warsaw, Poland Correspondence should be addressed to Olivier Barbier; [email protected] Received 6 December 2017; Accepted 15 February 2018; Published 11 April 2018 Academic Editor: Pascal Lapierre Copyright © 2018 Martin Perreault et al. is is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Biliary obstruction, a severe cholestatic complication, causes accumulation of toxic bile acids (BAs) in liver cells. Glucuronidation, catalyzed by UDP-glucuronosyltransferase (UGT) enzymes, detoxifies cholestatic BAs. Using liquid chromatography coupled to tandem mass spectrometry, 11 BA glucuronide (-G) species were quantified in prebiliary and postbiliary stenting serum and urine samples from 17 patients with biliary obstruction. Stenting caused glucuronide- and fluid-specific changes in BA-G levels and BA-G/BA metabolic ratios. In vitro glucuronidation assays with human liver and kidney microsomes revealed that even if renal enzymes generally displayed lower values, the two tissues shared similar glucuronidation capacities for BAs. By contrast, major differences between the two tissues were observed when four human BA-conjugating UGTs 1A3, 1A4, 2B4, and 2B7 were analyzed for mRNA and protein levels. Notably, the BA-24G producing UGT1A3 enzyme, abundant in the liver, was not detected in kidney microsomes. In conclusion, the circulating and urinary BA-G profiles are hugely impacted under severe cholestasis. e similar BA-glucuronidating abilities of hepatic and renal extracts suggest that both the liver and kidney may contribute to the urine BA-G pool. 1. Introduction Bile acids (BAs) play a major role in cholesterol homeostasis. In the liver, cholesterol is efficiently converted into the primary cholic (CA) and chenodeoxycholic (CDCA) acids for subsequent secretion into intestine via the bile [1]. In the duodenum, these acids act as natural detergents to facilitate the absorption of dietary lipids, liposoluble vitamins, and cholesterol [2]. Significant proportions of CDCA and CA are then converted in the respective secondary lithocholic acid (LCA) and deoxycholic acid (DCA) by resident bacteria [2]. Both primary and secondary acids are reabsorbed and return to the liver via the portal circulation. Back in the liver, LCA and CDCA sustain additional biotransformations into the 6-hydroxylated hyodeoxycholic acid (HDCA) and hyocholic acid (HCA), respectively [2, 3]. BAs are cytotoxic at elevated concentrations [4], and their accumulation in liver cells favors oxidative stress, inflammation, apoptosis, and subsequent damage to the liver parenchyma [2]. Such features are characteristic of cholestatic situations, where a reduction of the bile flow limits BA elimi- nation from hepatocytes [5]. A reduction of BA hepatic levels is therefore an important goal for anticholestatic strategies [5]. Glucuronidation, catalyzed by members of the UDP- glucuronosyltransferase (UGT) family, is a major elimination Hindawi Canadian Journal of Gastroenterology and Hepatology Volume 2018, Article ID 8096314, 12 pages https://doi.org/10.1155/2018/8096314

Transcript of Urinary Elimination of Bile Acid Glucuronides under Severe ...

Page 1: Urinary Elimination of Bile Acid Glucuronides under Severe ...

Research ArticleUrinary Elimination of Bile Acid Glucuronidesunder Severe Cholestatic Situations Contribution of Hepaticand Renal Glucuronidation Reactions

Martin Perreault1 EwaWunsch 2 Andrzej BiaBek2 Jocelyn Trottier1 Meacutelanie Verreault1

Patrick Caron1 Guy G Poirier3 Piotr Milkiewicz 4 and Olivier Barbier 1

1Laboratory of Molecular Pharmacology CHU-Quebec Research Centre and the Faculty of Pharmacy Laval UniversityQuebec City QC Canada2Translation Medicine Group Pomeranian Medical University Szczecin Poland3Proteomics Platform Quebec Genomic Center CHU-Quebec Research Centre and Faculty of Medicine Laval UniversityQuebec City QC Canada4Liver and Internal Medicine Medical University of Warsaw Warsaw Poland

Correspondence should be addressed to Olivier Barbier olivierbarbiercrchudequebeculavalca

Received 6 December 2017 Accepted 15 February 2018 Published 11 April 2018

Academic Editor Pascal Lapierre

Copyright copy 2018 Martin Perreault et alThis is an open access article distributed under theCreative CommonsAttribution Licensewhich permits unrestricted use distribution and reproduction in any medium provided the original work is properly cited

Biliary obstruction a severe cholestatic complication causes accumulation of toxic bile acids (BAs) in liver cells Glucuronidationcatalyzed by UDP-glucuronosyltransferase (UGT) enzymes detoxifies cholestatic BAs Using liquid chromatography coupled totandem mass spectrometry 11 BA glucuronide (-G) species were quantified in prebiliary and postbiliary stenting serum and urinesamples from 17 patients with biliary obstruction Stenting caused glucuronide- and fluid-specific changes in BA-G levels andBA-GBA metabolic ratios In vitro glucuronidation assays with human liver and kidney microsomes revealed that even if renalenzymes generally displayed lower119870119872 values the two tissues shared similar glucuronidation capacities for BAs By contrast majordifferences between the two tissues were observed when four human BA-conjugating UGTs 1A3 1A4 2B4 and 2B7 were analyzedfor mRNA and protein levels Notably the BA-24G producing UGT1A3 enzyme abundant in the liver was not detected in kidneymicrosomes In conclusion the circulating and urinary BA-G profiles are hugely impacted under severe cholestasis The similarBA-glucuronidating abilities of hepatic and renal extracts suggest that both the liver and kidney may contribute to the urine BA-Gpool

1 Introduction

Bile acids (BAs) play a major role in cholesterol homeostasisIn the liver cholesterol is efficiently converted into theprimary cholic (CA) and chenodeoxycholic (CDCA) acidsfor subsequent secretion into intestine via the bile [1] In theduodenum these acids act as natural detergents to facilitatethe absorption of dietary lipids liposoluble vitamins andcholesterol [2] Significant proportions of CDCA and CAare then converted in the respective secondary lithocholicacid (LCA) and deoxycholic acid (DCA) by resident bacteria[2] Both primary and secondary acids are reabsorbed andreturn to the liver via the portal circulation Back in the

liver LCA and CDCA sustain additional biotransformationsinto the 6120572-hydroxylated hyodeoxycholic acid (HDCA) andhyocholic acid (HCA) respectively [2 3]

BAs are cytotoxic at elevated concentrations [4] andtheir accumulation in liver cells favors oxidative stressinflammation apoptosis and subsequent damage to the liverparenchyma [2] Such features are characteristic of cholestaticsituations where a reduction of the bile flow limits BA elimi-nation from hepatocytes [5] A reduction of BA hepatic levelsis therefore an important goal for anticholestatic strategies[5]

Glucuronidation catalyzed by members of the UDP-glucuronosyltransferase (UGT) family is a major elimination

HindawiCanadian Journal of Gastroenterology and HepatologyVolume 2018 Article ID 8096314 12 pageshttpsdoiorg10115520188096314

2 Canadian Journal of Gastroenterology and Hepatology

mechanism for a variety of exogenous and endogenousmolecules [6] This enzymatic reaction consists in thetransfer of the glucuronosyl group from uridine 51015840-diphosphoglucuronic acid (UDPGA) to acceptor substratesIn humans 19 functional UGTs have been categorized intothree major UGT subfamilies UGT1A UGT2A and UGT2B[7] Among these enzymes hepatic UGT1A3 1A4 2B4 and2B7 isoforms have a remarkable capacity to convert BAsinto glucuronide in vitro [8ndash12] Bile acid glucuronidationinvolves either the 36-hydroxyl or 24-carboxyl groups of theBA steroid nucleus for the respective formation of ether 36Gor acylester 24G [8 9 11 12] UGT1A3 is the major enzymefor hepatic production of BA-24G while UGT1A4 2B4 and2B7 are themain producers of ether glucuronides [8 9 11 12]

An important consequence of glucuronidation is theintroduction of an additional negative charge to the BAmolecule which allows BA-G transport by conjugate-transporters such as the multidrug resistance related proteins(MRPs) 3 and 4 that are present at the basolateral mem-brane of hepatocytes [4] These transporters facilitate BA-G secretion into the blood followed by enhanced urinaryexcretion While increased BA-G urinary elimination hasbeen reported in patients with acute cholestasis the profileof these conjugates in both the circulation and urine hasonly been partially resolved [13ndash17] On the other hand thedogma that liver is the major site for glucuronidation hasbeen challenged by the recent evidences that human kidneyalso has significant drug glucuronidation capacity [18] Untilnow only few investigations were performed to determinethe contribution of renal UGTs to BA glucuronidation duringcholestasis [19] In the present study 11 BA-G species havebeen quantified in serum and urine samples from patientswith biliary obstruction obtained before and after biliarystenting In parallel experiments the BA-conjugating activ-ities of microsomal extracts from human liver and kidneywere compared

2 Materials and Methods

21 Ethics Statement All work has been conducted inaccordance with the declaration of Helsinki (1964) Thisstudy was approved by the appropriate clinical study reviewboards at the CHU-Quebec Research Centre Laval Univer-sity (ldquoComite drsquoethique de la recherche Clinique du CHULrdquoQuebec City QC Canada Projects 950514 and 970514)and PomeranianMedical University (Bioethics CommissionPomeranian Medical University in Szczecin Poland Reso-lution number BN-0014306) Experiments were conductedwith the human subjectsrsquo understanding and consent Allpatients had signed a written consent form before eachprocedure

22 Materials Bile acid glucuronides were obtained asdescribed [11 21] Commercially available microsomes fromhuman liver (pool of 26 men and 24 women HO620) andkidney (pool of 4 female and 4 male donors H0610R) werepurchased from Xenotech (Walkersville MD) Human liversamples were from kidney donors as previously described(livers 2 to 8) [10 12 22] Total RNA from individual

human liver and kidney tissues was purchased from Ambion(Streetsville Canada) (liver 1 and kidney 1) and Stratagene(Santa Clara CA) (kidney 2) Total RNA from a poolof 5 human kidney donors was purchased from Ambion(Abingdon UK) The Tri Reagent was from Molecu-lar Research Center Inc (Cincinnati OH) Protein assayreagents were obtained from Bio-Rad Laboratories Inc(Marnes-la-Coquette France) UGT antibodies were as pre-viously described [10 23 24] The secondary anti-rabbit IgGhorse antibody was from Amersham (Oakville Canada)

23 Patients with Biliary Obstruction Seventeen patients (8men and 9 women mean age 64 plusmn 10 years) with clinicaland biochemical features of cholestasis were recruited aspreviously reported [20] (Supplementary Material 1) Diag-nosis biliary tree dilatation evidences and biliary stentingprocedures were extensively described in a previous report[20] Stored urine was available from only 12 of these patients(6 men and 6 women) for BA-G measurement Informedconsent was obtained from each patient

24 Bile Acid Glucuronides Measurement Bile acid-glucu-ronide concentrations were determined from serum andurine samples (100 120583L) using liquid chromatography coupledto tandem mass spectrometry (LC-MSMS) with an elec-trospray interface as previously reported [11 12 21] Thechromatographic system consisted of an Alliance 2690HPLCapparatus (Waters Milford MA) and the tandem massspectrometry system was an API3200 mass spectrometer(Applied Biosystems Concord Canada)

25 Glucuronidation Assays All glucuronidation assays wereperformed for 2 hours in the presence of 10 120583g microsomalproteins in a previously reported assay buffer [11 12] Kineticparameters were assessed in liver and kidney samples usingsubstrate concentrations ranging from 1 to 350 120583M Theenzyme kinetic model was selected as recommended [25]using the Sigma Plot 112 assisted by Enzyme Kinetics 13program (SSI San Jose CA)

26 RNA Extraction Reverse Transcription and Real-TimePCR Analyses Total RNA was isolated according to the TriReagent acid phenol protocol as specified by the supplier(Molecular Research Center Inc) The reverse transcription(RT) and quantitative PCR reactions were performed aspreviously described [11 26] The real-time PCR ampli-fications were performed using an ABI Prism 7500FASTinstrument from Applied Biosystems (Foster City CA) Foreach reaction the final volume of 20 120583L comprised 10 120583Lof SYBR Green PCR Mix 2 120583L (4 120583M) of each previouslyreported primer [11] and 6 120583L of the indicated dilution ofRT products For each gene in each tissue the amplificationefficiency was tested using 2 to 5 log of cDNA produced fromliver- or kidney-purified RNA and sequential dilutions ofUGT cDNA constructs (00001 to 10 pg120583l) The differencebetween standard curve and sample efficiency was below10 as recommended [27] The amount of target geneswas derived from linear regression with standard curves ofthese UGT constructs according to reported protocols [28]

Canadian Journal of Gastroenterology and Hepatology 3

CA-24G

3000

2000

1000

CDCA-3G

5000

4000

3000

2000

1000

CDCA-24G

30

20

10

1800

1400

1000

600

200

LCA-3G

300

200

100lowast

40

30

20

10

LCA-24G DCA-3G

800

200

600

400

30

20

10

DCA-24G

ns

HDCA-6G

30

20

10

HDCA-24G

4500

3000

1500

HCA-6G

5001000

25002000

35004000

30

20

10

HCA-24G

400

800

1200

1600

2000

20

40

60

100

120

80

1000

2000

3000

5000

6000

4000

ns

4060

100

160180

120140

80

20

10

30

20

100

350

300250200150

50

100

250

200

150

50

3000

70006000

4000

1000

5000

200004

10

08

06

02

40000

80000

60000

20000

20

40

30

10

(a) Serum

(b) Urine

ns

ns

ns

ns

nsns

ns

ns

lowast

lowastlowast

lowast

lowast

lowastlowast

lowastlowast

lowastlowastlowast

lowastlowast

lowastlowast

lowastlowast

Bile

acid

-glu

curo

nide

le

vels

(nM

)Bi

le ac

id-g

lucu

roni

de

leve

ls (n

M)

Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post

Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post

Figure 1 Endoscopic stenting of the bile duct differentially affects the serum (a) and urine (b) bile acid-glucuronide profiles in patients sufferingfrom biliary obstruction Serum ((a) 119899 = 17) and urine ((b) 119899 = 12) samples drawn before (pre) and after (post) endoscopic biliary stentingfrom patients with stenosed bile ducts were analyzed for their concentrations in 11 bile acid-glucuronide species using LC-MSMS Resultsrepresent the mean concentration plusmn SEM and are expressed as nM 119901 values were determined by the Wilcoxon matched-pairs signed-ranktest lowast119901 lt 005 lowastlowast119901 lt 001 lowastlowastlowast119901 lt 0001 ns not significant

Messenger RNA levels were calculated using the averagemolecular weight of one base-paired nucleotide (660 gmol)and Avogadrorsquos constant (6022 times 1023molminus1)

27 UGT Protein Determination For Western blottingmicrosomes (5 120583g) were size-separated by 10 SDS-polyacrylamide gels and transferred onto nitrocellulosemembranes All antibodies were used at 1 2000 dilutions[10 23 24] The housekeeping protein 120573-actin was used toensure the equal protein loading in each lane An anti-rabbitIgG horse antibody (1 10000) conjugated with peroxidase(Amersham) was used as the second antibody and theresulting immunocomplexes were visualized as described[12 26]

Determination of UGT1A3 protein levels was alsoachieved using LC-MSMS quantification according to thepreviously reported analytical strategy [10]

28 Data Analyses Bile acid-glucuronide levels were calcu-lated asmeanplusmn standard error of themean (SEM) BA-G con-centrations did not satisfy the normal distribution accordingto the Shapiro-Wilk test thus the Wilcoxon matched-pairssigned-rank test was used for statistical analyses of theresponse to treatment Correlations were assessed by Spear-manrsquos rank correlation coefficient using the JMP StatisticalDiscovery V701 program (SAS Institute Cary NC)

3 Results

31 Biliary Stenting Modulates Circulating and Urinary BA-G Levels in Patients with Biliary Obstruction The circulatingand urinary BA-G profiles in prestenting and poststentingsamples from biliary stenosed patients are illustrated inFigure 1 In sera the species distribution was CDCA-3G

gt HCA-6G gt CA-24G gt DCA-3G = HDCA-6G gt LCA-3G ≫ HDCA-24G = HCA-24G gt DCA-24G = LCA-24G geCDCA-24G In urine the most abundant species was HCA-6G gt HDCA-6G = CA-24G gt CDCA-3G gt DCA-3G Asin serum other species were minor components Bile flowrestoration caused significant reduction in serum CDCA-3G(27-fold) CA-24G (25-fold) and LCA-3G (24-fold) contents(Figure 1(a)) By contrast CDCA-24G (24-fold) LCA-24G(30-fold) and HDCA-6G (30-fold) concentrations weresignificantly increased in poststenting versus prestenting sera(Figure 1(a)) Other changes failed to reach the statisticalsignificance As we previously reported [11] serum levels ofBA-G in noncholestatic volunteers varied from 07 plusmn 01 nMfor HDCA-24G to 635 plusmn 86 nM for HCA-6G As observedin the present study the most abundant species were HCA-6G CDCA-3G DCA-3G and HDCA-6G Biliary stentingalso changed urine BA-G levels (Figure 1(b)) CDCA-24G(41-fold) LCA-3G (35-fold) LCA-24G (65-fold) andDCA-24G (59-fold) were increased and CDCA-3G (43-fold) andHCA-24G (83-fold) were reduced

32 Biliary Stenting Differentially Affects the BA-G versusBA Metabolic Ratios in Serum and Urine from BiliaryStenosed Patients The serum and urine samples were pre-viously analyzed for their content in the 6 unconjugatedprecursors of the glucuronide derivatives analyzed here [20]These concentrations were used to calculate the metabolicratio (ie BA-GBA) for each glucuronide (Figure 2 andSupplementary Material 2) This parameter translates theglucuronide production capability from the available poolof unconjugated precursor thus ensuring that changes inblood or urine levels of BA-G species do not only reflectchanges in their precursors [11] In serum samples onlythe 65- 206- and 172-fold reductions in the respective

4 Canadian Journal of Gastroenterology and Hepatology

Serum UrineMetabolic ratios

Before stenting After stenting Before stenting After stenting

CDCA-3GCDCA 252 plusmn 43 69 plusmn 12 42 plusmn 08

CDCA-24GCDCA 01 plusmn 01 01 plusmn 01 02 plusmn 01 11 plusmn 03

CA-24GCA 102 plusmn 33 44 plusmn 12

LCA-3GLCA 14 plusmn 08 03 plusmn 01 NA NA

LCA-24GLCA 02 plusmn 01 01 plusmn 01 NA NA

DCA-3GDCA 64 plusmn 28 43 plusmn 28 21 plusmn 05

DCA-24GDCA 02 plusmn 01 01 plusmn 00 07 plusmn 06 17 plusmn 02

HDCA-6GHDCA 41 plusmn 40 49 plusmn 15 50 plusmn 25 414 plusmn 113

HDCA-24GHDCA 01 plusmn 01 01 plusmn 01 00 plusmn 00 00 plusmn 00

HCA-6GHCA 155 plusmn 56 198 plusmn 35 41591 plusmn 37459 2954 plusmn 2353

HCA-24GHCA 03 plusmn 01 03 plusmn 01 15 plusmn 07 01 plusmn 01

39 plusmn 28lowastlowastlowast

05 plusmn 02lowastlowast

04 plusmn 02lowastlowast

08 plusmn 03lowastlowast

(a)15

lowastlowast

10

5

0

minus5

minus35

minus30

minus25

minus20

minus15

minus10

CDCA

-24G

CDCA

-3G

CA-2

4G

LCA-

3G

LCA-

24G

DCA

-3G

DCA

-24G

HD

CA-6

G

HCA

-6G

HD

CA-2

4G

HCA

-24G

lowastlowastlowast

lowastlowast

lowastlowast

Urine

MR

BA-

GB

A(fo

ld ch

ange

s afte

r bili

ary

stent

ing)

Serum

(b)

Before stenting

p = 001

10

20

30

40

50

60

Seru

m C

A-24

GC

A ra

tio r2 = 052

4 6 8 10 12 142Urine CA-24GCA ratio

(c)

After stenting

01

02

03

04

05

06

07

Seru

m C

A-24

GC

A ra

tio

p = 002r2 = 045

2 3 41Urine CA-24GCA ratio

(d)

Response

minus50

minus40

minus30

minus20

minus10

0

10

20

30

Seru

m C

A-24

GC

A ra

tio

p = 002r2 = 044

minus8 minus4 0 4minus12Urine CA-24GCA ratio

(e)Figure 2 While bile flow restoration modulates metabolic ratios of bile acid glucuronides in a glucuronide- and fluid-dependent manner ((a)and (b)) the CA-24GCA ratio is unique in being significantly associated with serum and urine (cndashe) Serum and urine samples drawn beforeand after endoscopic biliary stenting from patients with stenosed bile ducts were analyzed for their concentrations in 11 bile acid-glucuronidespecies using LC-MSMS Concentrations of unconjugated acids were determined as reported [20] and themetabolic ratio for each species (a)was calculated as the ratio of glucuronide versus unconjugated precursor (b) Black (serum 119899 = 17) and white (urine 119899 = 12) bars representthe mean plusmn SEM of fold changes (posttreatment values divided by pretreatment values) in MRs Statistically significant differences betweenprestenting versus poststenting samples were determined using the Wilcoxon matched-pairs signed-rank test lowastlowast119901 lt 001 lowastlowastlowast119901 lt 0001Other changes failed to reach statistical significance The absence of unconjugated LCA in urine impaired such analysis for LCA-3 and -24G[20] while urine HDCA-24GHDCA was null (cndashe) Correlation analyses of paired (119899 = 12) prestenting (c) and poststenting (d) serum andurine CA-24GCA values or response to stenting determined as the difference between post- and pretreatment levels (e) were performedusing the Spearman rank-order correlation (1199032) and 119901 values for comparisons are indicated

Canadian Journal of Gastroenterology and Hepatology 5

Table 1 Summary of the correlation studies between serum and urine bile acid-glucuronide profiles

Urine levels Before stenting After stenting Response(1199032 119901 value) (1199032 119901 value) (1199032 119901 value)

CDCA-3G 010 030 002 067 025 010CDCA-24G 001 069 005 050 002 063CA-24G 032 005 051 001 007 042LCA-3G 000 097 019 015 001 081LCA-24G 014 022 047 001 042 002DCA-3G 082 lt00001 014 023 001 071DCA-24G 006 043 016 019 058 0004HDCA-6G 029 007 031 006 035 004HDCA-24G 000 100 005 047 003 059HCA-6G 029 007 015 020 067 0001HCA-24G 000 083 027 008 004 054Correlation analyses were performed using the Spearman rank-order correlation (1199032) and 119901 values for comparisons are indicated Response is calculated asthe difference between post- and prestenting values CA cholic acid CDCA chenodeoxycholic acid DCA deoxycholic acid HCA hyocholic acid HDCAhyodeoxycholic acid LCA lithocholic acid G glucuronide

CDCA-3GCDCA CA-24GCA and DCA-3GDCA ratioswere statistically significant (Figures 2(a) and 2(b)) Otherchanges even the reduction in DCA-24GDCA (117-fold)and LCA-3GLCA (53-fold) or the 25-fold increase of theHDCA-24GHDCA ratio failed to reach statistical signif-icance (Figure 2(b)) In urine (Supplementary Material 2)the unique significant change corresponded to the 52-foldreduction of theCA-24GCA ratio (Figure 2(b)) while the 14-and 15-fold reductions in HCA-6 and -24GMRs also failed toreach statistical significance

33 Serum and Urine BA-G Profiles Are Only Feebly Asso-ciated Correlation studies of circulating and urinary BA-Glevels in paired samples identified only few significant associ-ations (Table 1) In prestenting samples only the serumDCA-3G was significantly and positively correlated (1199032 = 082)to its urinary concentration while in poststenting fluids nocorrelation for this conjugate was detected Serum and urineCA-24G (1199032 = 051) and LCA-24G (1199032 = 047) levels weresignificantly correlated but only in samples obtained afterbile flow restoration (Table 1) When the response to stenting(determined as the difference between post- and prestentingvalues) was analyzed only changes for LCA-24G (1199032 = 042)DCA-24G (1199032 = 058) HDCA-6G (1199032 = 035) and HCA-6G (1199032 = 067) were positively and significantly associated(Table 1) Finally when similar analyses were performed forMRs the unique significant association in prestenting (1199032 =052) and poststenting (1199032 = 045) levels and response(1199032 = 044) was obtained with the ratio CA-24GCA (Figures2(c)ndash2(e))

34 Bile Acid Glucuronidation by Human Liver and KidneyExtracts The limited associations between circulating andurinary profiles observed above suggest a potential extra-hepatic origin for urinary levels of BA-G To test such ahypothesis we sought to compare the kinetic parameters ofBA glucuronidation by human liver (pool of 50 donors) and

kidney (pool of 8 donors) microsomes (Figures 3ndash5) Nodifferences in kinetic models were observed with the BA-specific glucuronidation reactions being distributed among 3models

(i) The sigmoidal pattern for CA-24G and CDCA-24 and-3G (Figure 3) DCA-3 and -24G (Figures 4(c) and 4(d)) andHDCA-6 and -24G (Figures 5(a) and 5(b)) production

(ii) The Michaelis-Menten model for the formation ofLCA-24G (Figure 4(b)) and HCA-6 and -24G (Figures 5(c)and 5(d))

(iii) The substrate-inhibition model for LCA-3G withrespective 119870119868 values of 2403 plusmn 535 and 2554 plusmn 699 120583M forliver and kidney extracts (Figure 4(a))

With the exception of CA-24G HDCA-24G and HCA-24G kidney UGTs generally displayed lower 119870119872 values Forexample kidney microsomes exhibited a 6-fold lower 119870119872value than liver ones for LCA-3G production (Figure 4(a))Another notable difference between liver and kidney micro-somes relates to the 10-fold higher 119881max value of HDCA-24Gproduction obtained with the hepatic microsomal prepara-tion (Figure 5(b)) Nevertheless the most efficient reactionswere obtained with the conversion of HDCA and HCA into6G derivatives that occurred at high velocities (ie 119881max)with both liver and kidney microsomes (Figures 5(a) and5(c)) In both tissue extracts the 6120572-hydroxylated acids weremore actively converted into 6-glucuronide when comparedto their 24-glucuronide counterparts (Figure 5) Interestinglythe opposite was observed for CDCA (Figures 3(a) and 3(b))and LCA (Figures 4(a) and 4(b)) The 119881max values for DCA-3and -24G formation were however similar (Figures 4(c) and4(d))

35 Differential BA-Conjugating UGT Expression in theHuman Liver and Kidney Results from kinetic experimentsindicate that the human kidney possesses an efficient BA-conjugating UGT system We next investigated whetherthe BA-conjugating UGTs 1A3 1A4 2B4 and 2B7 [8ndash11] are differentially expressed in human liver and kidney

6 Canadian Journal of Gastroenterology and Hepatology

02

04

06

08

10CD

CA-3

G fo

rmat

ion

(nm

olh

mg

prot

eins

)

100[CDCA] (M)

200 300 400

Liver nA = 17 plusmn 01

KMB = 821 plusmn 60

VGRC = 08 plusmn 01

Kidney nA = 19 plusmn 07KM

B = 443 plusmn 96VGR

C = 05 plusmn 01

(a)

10

20

30

CDCA

-24G

form

atio

n(n

mol

hm

g pr

otei

ns)

100[CDCA] (M)

200 300 400

Liver nA = 14 plusmn 01KM

B = 1682 plusmn 186VGR

C = 37 plusmn 02

Kidney nA = 13 plusmn 03KM

B = 897 plusmn 298VGR

C = 25 plusmn 04

(b)

05

10

15

20

CA-2

4G fo

rmat

ion

(nm

olh

mg

prot

eins

)

100[CA] (M)

200 300 400

Liver nA = 12 plusmn 01KM

B = 1141 plusmn 168VGR

C = 16 plusmn 01

Kidney nA = 10 plusmn 01KM

B = 1481 plusmn 397VGR

C = 11 plusmn 01

(c)

Figure 3 Kinetic analyses of CDCA-3G (a) CDCA-24G (b) and CA-24G (c) formation by microsomes from human liver and kidney Humanliver (black squares pool of 50 donors) and kidney (white diamonds pool of 8 donors) microsomes (10 120583g) were incubated in the presenceof increasing concentrations (1 to 350 120583M) of chenodeoxycholic acid (CDCA (a) and (b)) or cholic acid (CA (c)) for 2 hours at 37∘C Theformation of CDCA-3G (a) CDCA-24G (b) and CA-24G (c) was analyzed by LC-MSMS For each panel graphs represent the rate ofproduct formation (119910-axis) versus substrate concentration (119909-axis) of two experiments performed in triplicate The listed kinetic parameterswere determined as indicated in ldquoMaterials and Methodsrdquo A119899 Hill coefficient B119870119872 calculated Michaelis constant expressed as 120583M C119881maxcalculated maximal velocity expressed as nmolhmg proteins

Canadian Journal of Gastroenterology and Hepatology 7

200 300 400100[LCA] (M)

40

80

120

160

LCA-

3G fo

rmat

ion

(nm

olh

mg

prot

eins

)

LiverKM

B = 487 plusmn 90VGR

C = 171 plusmn 29

Kidney

KIA = 2403 plusmn 535

KMB = 82 plusmn 22

VGRC = 121 plusmn 11

KIA = 2554 plusmn 699

(a)

200 300 400100[LCA] (M)

60

120

180

240

300

LCA-

24G

form

atio

n (n

mol

hm

g pr

otei

ns)

LiverKM

B = 403 plusmn 67VGR

C = 321 plusmn 16

KidneyKM

B = 305 plusmn 56VGR

C = 162 plusmn 08

(b)

100 200 300 400[DCA] (M)

10

20

30

40

50

60

DCA

-3G

form

atio

n(n

mol

hm

g pr

otei

ns)

Liver nD 17 plusmn 03KM

B = 831 plusmn 113VGR

C = 31 plusmn 02

Kidney nD = 17 plusmn 03KM

B = 823 plusmn 136VGR

C = 36 plusmn 03

(c)

20

40

60

80

DCA

-24G

form

atio

n(n

mol

hm

g pr

otei

ns)

200 300 400100[DCA] (M)

Liver nD 12 plusmn 02KM

B = 711 plusmn 121VGR

C = 58 plusmn 05

Kidney nD = 17 plusmn 03KM

B = 582 plusmn 87VGR

C = 38 plusmn 03

(d)Figure 4Kinetic analyses of LCA-3G (a) LCA-24G (b) DCA-3G (c) and DCA-24G (d) formation by microsomes from human liver and kidneyHuman liver (black squares pool of 50 donors) and kidney (white diamonds pool of 8 donors) microsomes (10 120583g) were incubated in thepresence of increasing concentrations (1 to 350120583M) of lithocholic acid (LCA (a) and (b)) or deoxycholic acid (DCA (c) and (d)) for 2 hoursat 37∘C The formation of LCA-3G (a) LCA-24G (b) DCA-3G (c) and DCA-24G (d) was analyzed by LC-MSMS For each panel graphsrepresent the rate of product formation (119910-axis) versus substrate concentration (119909-axis) of two experiments performed in triplicateThe listedkinetic parameters were determined as indicated in ldquoMaterials and Methodsrdquo A119870119868 constant of inhibition expressed as 120583M B119870119872 calculatedMichaelis constant expressed as 120583M C119881max calculated maximal velocity expressed as nmolhmg proteins D119899 Hill coefficient

8 Canadian Journal of Gastroenterology and Hepatology

1750

3500

5250

7000

HD

CA-6

G fo

rmat

ion

(nm

olh

mg

prot

eins

)

200 300 400100[HDCA] (M)

Liver nA 29 plusmn 04KM

B = 551 plusmn 33VGR

C = 5287 plusmn 211

Kidney nA = 27 plusmn 05KM

B = 447 plusmn 38VGR

C = 3084 plusmn 156

(a)

100

200

300

400

500

HD

CA-2

4G fo

rmat

ion

(nm

olh

mg

prot

eins

)

200 300 400100[HDCA] (M)

Liver nA 38 plusmn 11KM

B = 808 plusmn 64VGR

C = 316 plusmn 22

Kidney nA = 23 plusmn 08KM

B = 939 plusmn 184VGR

C = 37 plusmn 05

(b)

800

1600

2400

3200

HCA

-6G

form

atio

n(n

mol

hm

g pr

otei

ns)

200 300 400100[HCA] (M)

LiverKM

B = 1202 plusmn 156VGR

C = 2285 plusmn 134

KidneyKM

B = 960 plusmn 192VGR

C = 2844 plusmn 241

(c)

20

40

60

HCA

-24G

form

atio

n(n

mol

hm

g pr

otei

ns)

200 300 400100[HCA] (M)

LiverKM

B = 617 plusmn 65VGR

C = 49 plusmn 02

KidneyKM

B = 978 plusmn 206VGR

C = 33 plusmn 03

(d)Figure 5 Kinetic analyses of HDCA-6G (a) HDCA-24G (b) HCA-6G (c) and HCA-24G (d) formation by microsomes from human liver andkidney Human liver (black squares pool of 50 donors) and kidney (white diamonds pool of 8 donors) microsomes (10 120583g) were incubatedin the presence of increasing concentrations (1 to 350120583M) of hyodeoxycholic acid (HDCA (a) and (b)) or hyocholic acid (HCA (c) and(d)) for 2 hours at 37∘CThe formation of HDCA-6G (a) HDCA-24G (b) HCA-6G (c) and HCA-24G (d) was analyzed by LC-MSMS Foreach panel graphs represent the rate of product formation (119910-axis) versus substrate concentration (119909-axis) of two experiments performedin triplicate The listed kinetic parameters were determined as indicated in ldquoMaterials and Methodsrdquo A119899 Hill coefficient B119870119872 calculatedMichaelis constant expressed as 120583M C119881max calculated maximal velocity expressed as nmolhmg proteins

Canadian Journal of Gastroenterology and Hepatology 9

Kid

ney

1A3

Kid

ney

Kid

ney

Kid

ney

Live

r

Live

r

Live

r

Live

r

1A4 2B4 2B7UGT

103

104

105

106

107U

GT

tran

scrip

ts (lo

g of

copy

num

ber

g RN

A)

(a)

1A3

2B42B7

2B7

1A

2B

LiverKidneyAnti-UGT kDa

55

55

55

55

55

40Anti-actin

(b)

Live

r

Kidn

ey

ND

5

10

15

20

UG

T1A

3 pr

otei

n le

vels

(pm

olm

g m

icro

som

es)

(c)

Figure 6 Differential expression of the bile acid-conjugating UGT enzymes in the human liver and kidney (a) UGTs 1A3 1A4 2B4 and 2B7mRNA levels in total RNA samples from human kidney (1 pool of 5 donors and 2 individual donors) and liver (8 individual donors) weredetermined through qRT-PCR analyses as described in ldquoMaterials andMethodsrdquo Results are expressed as log10 ofmRNA copy number per 120583gof total RNA Each symbol represents 1 donor with the exception of the white triangle that corresponds to a pool of the 5 kidney donors (b)The UGT protein contents in microsomal preparations (5120583g) from human kidney (pool of 4 female and 4 male donors) and liver (pool of 26male and 24 female donors) were determined after size separation on SDS-PAGE and hybridization with the anti-UGT1A3 anti-UGT2B47anti-UGT2B7 anti-UGT1A and anti-UGT2B antibodies (1 2000 dilution) The housekeeping protein 120573-actin was used to ensure the equalprotein loading in each lane Data presented are representative of two experiments (c) The UGT1A3 protein content in human liver andkidney microsomes was determined using LC-MSMS analyses as previously described [10] Data represent the mean plusmn SD of two differentexperiments performed in triplicate ND not detected

(Figure 6) With the exception of UGT2B4 transcripts thatwere detected only in 1 kidney mRNA sample other UGTmessengers were detected in all kidney and liver prepara-tions (Figure 6(a)) As expected [28 29] their mRNA levelssustained a strong interindividual variability Western blotanalyses evidenced however major differences between liverand kidney (Figure 6(b)) A clear UGT1A3 immunoreactivecomplex was obtained with liver extracts while this UGTprotein was not detected inmicrosomal proteins from kidney(Figure 6(b)) While the UGT2B7 protein was found atsimilar levels in both tissues the use of an anti-UGT2B42B7antibody indicated that the UGT2B4 protein may be moreabundant in liver than in kidney extracts (Figure 6(b))The lack of an accurate and specific anti-UGT1A4 antibody

impaired the quantification of this BA-conjugating UGThowever we were able to further confirm the absence ofthe UGT1A3 protein in kidney microsomes through the useof our previously established LC-MSMS-based proteomicmethod (Figure 6(c)) [10]

4 Discussion

Urinary and circulating levels of BA-Gs detected in thepresent study are similar to previous findings in terms ofboth glucuronide concentrations and species distribution [1314 16] However an important improvement of the presentinvestigations resides in our ability to discriminate betweenether and ester glucuronides of a single BA For example

10 Canadian Journal of Gastroenterology and Hepatology

while glucuronide conjugates of CDCA CA and DCA werepreviously identified as accumulating metabolites in serafrom patients undergoing bile drainage [20] the nature ofthese glucuronides has not been resolved until now Ourresults point out the ester CDCA-3G and DCA-3G andthe acyl CA-24G as the most abundant glucuronide speciesfound in patients with biliary obstruction Because acyl butnot ester glucuronides can be potentially toxic molecules(reviewed in [30]) such information may be critical inanticipating the consequences of their accumulation duringcholestasis

Interestingly serum levels of some of the bile acidglucuronides such as CDCA-3G CA-24G and LCA-3Gwere significantly reduced after biliary stenting while someothers like CDCA-24G LCA-24G and HDCA-6G weresignificantly increased While additional investigations arewarranted to decipher the mechanisms beyond such a glu-curonide species-dependent responsiveness one can spec-ulate that these changes may actually reflect the differen-tial manner in which the UGT enzymes andor the BA-Gtransporters may be altered upon biliary obstruction andafter stenting Therefore it would have been of interest tocompare how these proteins behave in tissues (ie liver andkidney) from cholestatic donors to validate such a hypothesisActually one limitation of the current study is that the liverand kidneymicrosomes used are not fromdonorswith biliaryobstruction When compared to other glucuronide conju-gates CA-24G presented a unique behavior (1) in contrastto other acyl BA-Gs that are only minor components of theurine and serum glucuronide pools CA-24G is the 3rd mostabundant glucuronidated acid in prestenting samples (2) thisspecies is the most spectacularly affected when poststentingversus prestenting sera profiles are compared (3) the serumCA-24GCA ratio sustains the stronger reduction after biliarystenting (4) this MR is also unique in being significantlyreduced in posttreatment urines and (5) only the CA-24GCA ratio exhibits positive serumurine association interms of prestenting and poststenting distribution as wellas treatment response These last observations support ahepatic origin for the formation of CA-24G and the strongCA-24G accumulation in prestenting fluids and its spec-tacular reduction after bile flow restoration suggests thatunder noncholestatic situations this compound formed inliver cells is normally secreted in bile This hypothesis isfurther supported by previous investigations in which theintravenous injections of cholate glucuronide to rats resultedin a rapid and efficient secretion in bile of the unchangedglucuronide conjugate [31] By contrast when the sameprocedure was applied to bile duct ligated animals 95 of theradioactivity was recovered in urine [31] The present studyindicates that similar rerouting of CA-24G elimination tourine may also occur in cholestatic patients since circulatinglevels detected in prestenting cholestatic donors were almost50-fold higher than those previously quantified in samplesfrom noncholestatic volunteers (sim2 nM [11]) Privilegingsecretion from liver cells into the blood for subsequenturinary elimination may actually protect the liver againstthe accumulation of such abundant and potentially toxicglucuronide species during cholestasis In the same vein the

improved formation of other conjugates such as CDCA-3Gand DCA-3G which we recently identified as nontoxic BAs[32] may also participate in the hepatic detoxification oftheir unconjugated CDCA and DCA precursors Consistentwith the above stated hypothesis circulating MRs for these2 conjugates (ie 252 and 64 for CDCA-3G and DCA-3Gresp) are spectacularly increased when compared to thosepreviously found in noncholestatic donors (06 and 01) [11]However the fact that circulating and urinary MRs for thesespecies were not associated and the fact that biliary stentingexerted only minor effects on these ratios in urine are alsoindicative of an extrahepatic origin for urine CDCA- andDCA-3G species at least after bile flow restoration

We next compared the human liver and kidney in termsof bile acid glucuronidation activity and BA-conjugatingUGT expression With 2 exceptions (ie the 6- and 10-fold difference in 119870119872 and 119881max values for LCA-3G andHDCA-24G formations resp) these experiments revealthe remarkable similarity of renal and hepatic bile acidglucuronidation processes and the clear preference of bothtissues in producing HDCA-6 and HCA-6G Such preferencemay actually explain the predominance of these conjugates invarious human fluids as previously described [11] Howeverthe most intriguing observation issuing from these analysesrelates to the UGT1A3 enzyme Indeed not only was thisenzyme detected in kidney extracts only at the mRNA(but not protein) level but also the absence of such anactive enzyme previously identified as the main isoform foracyl BA glucuronidation [8ndash10 12] is inconsistent with theelevated acyl glucuronide production capability of kidneymicrosomes A plausible explanation for the conflictingobservation on UGT1A3 mRNA and protein expression inkidney samples relates to the fact that protein and RNAextracts were from different sources and thus were notpaired It is therefore possible that only donors for mRNAexpress these isoforms in the kidney Meanwhile unlikelysuch a possibility is supported by the controversial nature ofthe UGT1A3 expression in the human kidney [29 33ndash35]Additional investigations are therefore warranted to clarifywhether this BA-conjugating protein is found or not in thehuman kidney

Nevertheless the present study clearly establishes themajor contributions that liver and kidney play in glu-curonidating bile acid during cholestasis Interestingly Zhouand colleagues reported that intestinal glucuronidation alsoplays an important role in controlling the local bile acidhomeostasis and the pathological development of colitis [36]These authors elegantly demonstrated that in the intestinebile acids glucuronidation leads to reduced intracellular levelsof ligands for the nuclear receptor FXR Reduced FXR activa-tion limits the secretion of FGF1519 in the portal circulationwhich in turns leads to an increase of de novo formationof bile acid in the liver [36] Thus in contrast to the liverand kidney where glucuronidation is thought to facilitate theremoval of bile acids the intestinal glucuronidation seems toincrease the delivery of bile acidsHowever it should be notedthat during obstructive cholestasis the delivery of hepaticbile acids to the intestine is compromised and thus onlyminor intestinal glucuronidation may occur

Canadian Journal of Gastroenterology and Hepatology 11

5 Conclusion

In conclusion the present study evidences the major impactthat bile flow restoration exerts on the circulating and urinarybile acid-glucuronide profiles and demonstrates the elevatedcapability of the human kidney to glucuronidate theseendogenous compounds Future investigations are requiredto clarify the mechanism allowing kidney extracts to convertbile acids into acyl glucuronides in the absence of theUGT1A3 protein

Abbreviations

BA Bile acidBO Biliary obstructionCA Cholic acidCDCA Chenodeoxycholic acidDCA Deoxycholic acidFXR Farnesoid X receptorG GlucuronideHDCA Hyodeoxycholic acidHCA Hyocholic acidLCA Lithocholic acidLC-MSMS Liquid chromatography-tandem mass

spectrometryUDPGA UDP-glucuronic acidUGT UDP-glucuronosyltransferaseMRPs Multidrug resistance related proteins

Conflicts of Interest

The authors declare that there are no conflicts of interest andthat there is no pertinent financial arrangement concerningthe work presented here

Acknowledgments

The authors wish to thank Dr Virginie Bocher for criticalreading of the manuscript This study was supported bygrants from theCanadian Institute ofHealth Research (CIHR119331) the Canadian Liver Foundation the Natural Sci-ences and Engineering Research Council of Canada (NSERC402213-2012) and the Canadian Foundation for Innovation(CFI 25712)Martin Perreault is holder of a scholarship fromthe ldquoFonds pour la Recherche en Sante du Quebecrdquo (FRSQ23444) Olivier Barbier is holder of a salary grant fromCIHR (New Investigator MSH95330) Piotr Milkiewiczwas supported by Grants DEC-201101BNZ504216 and201102ANZ500321 from the National Science Centre inPoland

Supplementary Materials

The supplementary materials present details of patient withbiliary obstruction (Supplementary Material 1) and thechanges in the urine profile of bile acid glucuronides and intheir metabolic ratios before and after a biliary stenting pro-cedure in patients with biliary obstruction (SupplementaryMaterial 2) Supplementary Material 1 patients with biliary

obstruction (adapted from [20]) Supplementary Material 2changes in the urine profile of bile acid glucuronides andin their metabolic ratios before and after a biliary stentingprocedure in patients with biliary obstruction (119899 = 12)(Supplementary Materials)

References

[1] D W Russell ldquoThe enzymes regulation and genetics of bileacid synthesisrdquo Annual Review of Biochemistry vol 72 pp 137ndash174 2003

[2] M J Monte J J G Marin A Antelo and J Vazquez-TatoldquoBile acids Chemistry physiology and pathophysiologyrdquoWorldJournal of Gastroenterology vol 15 no 7 pp 804ndash816 2009

[3] A F Hofmann ldquoDetoxification of lithocholic acid a toxicbile-acid relevance to drug hepatotoxicityrdquo Drug MetabolismReviews vol 36 no 3-4 pp 703ndash722 2004

[4] C Pauli-Magnus B Stieger Y Meier G A Kullak-Ublick andP J Meier ldquoEnterohepatic transport of bile salts and genetics ofcholestasisrdquo Journal of Hepatology vol 43 no 2 pp 342ndash3572005

[5] G Paumgartner ldquoPharmacotherapy of cholestatic liver dis-easesrdquo Journal of Digestive Diseases vol 11 no 3 pp 119ndash1252010

[6] M Verreault J Kaeding P Caron et al ldquoRegulation ofendobiotics glucuronidation by ligand-activated transcriptionfactors Physiological function and therapeutic potentialrdquoDrugMetabolism Reviews vol 42 no 1 pp 106ndash118 2010

[7] P I Mackenzie K W Bock B Burchell et al ldquoNomenclatureupdate for the mammalian UDP glycosyltransferase (UGT)gene superfamilyrdquo Pharmacogenetics and Genomics vol 15 no10 pp 677ndash685 2005

[8] M H Court S Hazarika S Krishnaswamy M Finel andJ A Williams ldquoNovel polymorphic human UDP-glucurono-syltransferase 2A3 Cloning functional characterization ofenzyme variants comparative tissue expression and geneinductionrdquoMolecular Pharmacology vol 74 no 3 pp 744ndash7542008

[9] W E Gall G Zawada B Mojarrabi et al ldquoDifferential glu-curonidation of bile acids androgens and estrogens by humanUGT1A3 and 2B7rdquo The Journal of Steroid Biochemistry andMolecular Biology vol 70 no 1ndash3 pp 101ndash108 1999

[10] J Trottier D El Husseini M Perreault et al ldquoThe humanUGT1A3 enzyme conjugates norursodeoxycholic acid into aC23-ester glucuronide in the liverrdquo The Journal of BiologicalChemistry vol 285 no 2 pp 1113ndash1121 2010

[11] J Trottier M Perreault I Rudkowska et al ldquoProfiling serumbile acid glucuronides in humans Gender divergences geneticdeterminants and response to fenofibraterdquo Clinical Pharmacol-ogy ampTherapeutics vol 94 no 4 pp 533ndash543 2013

[12] P Caron J Trottier M Verreault J Belanger J Kaeding andO Barbier ldquoEnzymatic production of bile acid glucuronidesused as analytical standards for liquid chromatography-massspectrometry analysesrdquo Molecular Pharmaceutics vol 3 no 3pp 293ndash302 2006

[13] B Alme and J Sjovall ldquoAnalysis of bile acid glucuronidesin urine identification of 3120572612057212120572-trihydroxy-5120573-cholanoicacidrdquoThe Journal of Steroid Biochemistry andMolecular Biologyvol 13 no 8 pp 907ndash916 1980

[14] A Stiehl R Raedsch G Rudolph P Czygan and S WalkerldquoAnalysis of bile acid glucuronides in urine group separation

12 Canadian Journal of Gastroenterology and Hepatology

on a lipophilic anion exchangerrdquo Clinica Chimica Acta vol 123no 3 pp 275ndash285 1982

[15] A Stiehl R Raedsch G Rudolph U Gundert-Remy and MSenn ldquoBiliary and urinary excretion of sulfated glucuronidatedand tetrahydroxylated bile acids in cirrhotic patientsrdquo Hepatol-ogy vol 5 no 3 pp 492ndash495 1985

[16] H Takikawa T Beppu and Y Seyama ldquoUrinary concentrationsof bile acid glucuronides and sulfates in hepatobiliary diseasesrdquoGastroenterologia Japonica vol 19 no 2 pp 104ndash109 1984

[17] H Wietholtz H Marschall R Reuschenbach H Matern andS Matern ldquoUrinary excretion of bile acid glucosides and glu-curonides in extrahepatic cholestasisrdquo Hepatology vol 13 no4 pp 656ndash662 1991

[18] K M Knights A Rowland and J O Miners ldquoRenal drugmetabolism in humans The potential for drug-endobioticinteractions involving cytochrome P450 (CYP) and UDP-glu-curonosyltransferase (UGT)rdquo British Journal of Clinical Phar-macology vol 76 no 4 pp 587ndash602 2013

[19] S Matern H Matern E H Farthmann and W GerokldquoHepatic and extrahepatic glucuronidation of bile acids in manCharacterization of bile acid uridine 5rsquo-diphosphate-glucu-ronosyltransferase in hepatic renal and intestinalmicrosomesrdquoThe Journal of Clinical Investigation vol 74 no 2 pp 402ndash4101984

[20] J Trottier A Białek P Caron R J Straka P Milkiewicz and OBarbier ldquoProfiling circulating and urinary bile acids in patientswith biliary obstruction before and after biliary stentingrdquo PLoSONE vol 6 no 7 Article ID e22094 2011

[21] M Perreault L Gauthier-Landry J Trottier et al ldquoThe humanUDP-glucuronosyltransferase UGT2A1 and UGT2A2 enzymesare highly active in bile acid glucuronidationrdquoDrugMetabolismand Disposition vol 41 no 9 pp 1616ndash1620 2013

[22] M E Campbell D M Grant T Inaba and W KalowldquoBiotransformation of caffeine paraxanthine theophylline andtheobromine by polycyclic aromatic hydrocarbon-induciblecytochrome(s) P-450 in human liver microsomesrdquo DrugMetabolism and Disposition vol 15 no 2 pp 237ndash249 1987

[23] J Lepine O Bernard M Plante et al ldquoSpecificity andregioselectivity of the conjugation of estradiol estrone andtheir catecholestrogen and methoxyestrogen metabolites byhuman uridine diphospho-glucuronosyltransferases expressedin endometriumrdquo The Journal of Clinical Endocrinology ampMetabolism vol 89 no 10 pp 5222ndash5232 2004

[24] J Thibaudeau J Lepine J Tojcic et al ldquoCharacterizationof common UGT1A8 UGT1A9 and UGT2B7 variants withdifferent capacities to inactivate mutagenic 4-hydroxylatedmetabolites of estradiol and estronerdquo Cancer Research vol 66no 1 pp 125ndash133 2006

[25] J O Miners P I MacKenzie and K M Knights ldquoTheprediction of drug-glucuronidation parameters in humansUDP-glucuronosyltransferase enzyme-selective substrate andinhibitor probes for reaction phenotyping and in vitroin vivoextrapolation of drug clearance and drug-drug interactionpotentialrdquo Drug Metabolism Reviews vol 42 no 1 pp 189ndash2012010

[26] M Verreault K Senekeo-Effenberger J Trottier et al ldquoTheliver X-receptor alpha controls hepatic expression of the humanbile acid-glucuronidating UGT1A3 enzyme in human cells andtransgenic micerdquo Hepatology vol 44 no 2 pp 368ndash378 2006

[27] R G Rutledge ldquoMathematics of quantitative kinetic PCR andthe application of standard curvesrdquo Nucleic Acids Research vol31 no 16 pp 93endash93 2003

[28] T Izukawa M Nakajima R Fujiwara et al ldquoQuantitative anal-ysis of UDP-glucuronosyltransferase (UGT) 1A and UGT2Bexpression levels in human liversrdquo Drug Metabolism and Dis-position vol 37 no 8 pp 1759ndash1768 2009

[29] S Ohno and S Nakajin ldquoDetermination of mRNA expressionof human UDP-glucuronosyltransferases and application forlocalization in various human tissues by real-time reversetranscriptase-polymerase chain reactionrdquoDrugMetabolism andDisposition vol 37 no 1 pp 32ndash40 2009

[30] S L Regan J L Maggs T G Hammond C Lambert D PWilliams and B K Park ldquoAcyl glucuronidesThe good the badand the uglyrdquo Biopharmaceutics amp Drug Disposition vol 31 no7 pp 367ndash395 2010

[31] J M Little M V Chari and R Lester ldquoExcretion of cholateglucuroniderdquo Journal of Lipid Research vol 26 no 5 pp 583ndash592 1985

[32] M Perreault A Białek J Trottier et al ldquoRole of glucuronidationfor hepatic detoxification and urinary elimination of toxic bileacids during biliary obstructionrdquo PLoS ONE vol 8 no 11Article ID e80994 2013

[33] H Girard E Levesque J Bellemare K Journault B Caillierand C Guillemette ldquoGenetic diversity at the UGT1 locus isamplified by a novel 31015840 alternative splicing mechanism leadingto nine additional UGT1A proteins that act as regulators ofglucuronidation activityrdquo Pharmacogenetics and Genomics vol17 no 12 pp 1077ndash1089 2007

[34] A Nakamura M Nakajima H Yamanaka R Fujiwara and TYokoi ldquoExpression of UGT1A and UGT2B mRNA in humannormal tissues and various cell linesrdquo Drug Metabolism andDisposition vol 36 no 8 pp 1461ndash1464 2008

[35] D E Harbourt J K Fallon S Ito et al ldquoQuantificationof human uridine-diphosphate glucuronosyl transferase 1Aisoforms in liver intestine and kidney using nanobore liq-uid chromatography-tandem mass spectrometryrdquo AnalyticalChemistry vol 84 no 1 pp 98ndash105 2012

[36] X Zhou L Cao C Jiang et al ldquoPPAR120572-UGT axis activationrepresses intestinal FXR-FGF15 feedback signalling and exacer-bates experimental colitisrdquoNature Communications vol 5 2014

Stem Cells International

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

MEDIATORSINFLAMMATION

of

EndocrinologyInternational Journal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Disease Markers

Hindawiwwwhindawicom Volume 2018

BioMed Research International

OncologyJournal of

Hindawiwwwhindawicom Volume 2013

Hindawiwwwhindawicom Volume 2018

Oxidative Medicine and Cellular Longevity

Hindawiwwwhindawicom Volume 2018

PPAR Research

Hindawi Publishing Corporation httpwwwhindawicom Volume 2013Hindawiwwwhindawicom

The Scientific World Journal

Volume 2018

Immunology ResearchHindawiwwwhindawicom Volume 2018

Journal of

ObesityJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Computational and Mathematical Methods in Medicine

Hindawiwwwhindawicom Volume 2018

Behavioural Neurology

OphthalmologyJournal of

Hindawiwwwhindawicom Volume 2018

Diabetes ResearchJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Research and TreatmentAIDS

Hindawiwwwhindawicom Volume 2018

Gastroenterology Research and Practice

Hindawiwwwhindawicom Volume 2018

Parkinsonrsquos Disease

Evidence-Based Complementary andAlternative Medicine

Volume 2018Hindawiwwwhindawicom

Submit your manuscripts atwwwhindawicom

Page 2: Urinary Elimination of Bile Acid Glucuronides under Severe ...

2 Canadian Journal of Gastroenterology and Hepatology

mechanism for a variety of exogenous and endogenousmolecules [6] This enzymatic reaction consists in thetransfer of the glucuronosyl group from uridine 51015840-diphosphoglucuronic acid (UDPGA) to acceptor substratesIn humans 19 functional UGTs have been categorized intothree major UGT subfamilies UGT1A UGT2A and UGT2B[7] Among these enzymes hepatic UGT1A3 1A4 2B4 and2B7 isoforms have a remarkable capacity to convert BAsinto glucuronide in vitro [8ndash12] Bile acid glucuronidationinvolves either the 36-hydroxyl or 24-carboxyl groups of theBA steroid nucleus for the respective formation of ether 36Gor acylester 24G [8 9 11 12] UGT1A3 is the major enzymefor hepatic production of BA-24G while UGT1A4 2B4 and2B7 are themain producers of ether glucuronides [8 9 11 12]

An important consequence of glucuronidation is theintroduction of an additional negative charge to the BAmolecule which allows BA-G transport by conjugate-transporters such as the multidrug resistance related proteins(MRPs) 3 and 4 that are present at the basolateral mem-brane of hepatocytes [4] These transporters facilitate BA-G secretion into the blood followed by enhanced urinaryexcretion While increased BA-G urinary elimination hasbeen reported in patients with acute cholestasis the profileof these conjugates in both the circulation and urine hasonly been partially resolved [13ndash17] On the other hand thedogma that liver is the major site for glucuronidation hasbeen challenged by the recent evidences that human kidneyalso has significant drug glucuronidation capacity [18] Untilnow only few investigations were performed to determinethe contribution of renal UGTs to BA glucuronidation duringcholestasis [19] In the present study 11 BA-G species havebeen quantified in serum and urine samples from patientswith biliary obstruction obtained before and after biliarystenting In parallel experiments the BA-conjugating activ-ities of microsomal extracts from human liver and kidneywere compared

2 Materials and Methods

21 Ethics Statement All work has been conducted inaccordance with the declaration of Helsinki (1964) Thisstudy was approved by the appropriate clinical study reviewboards at the CHU-Quebec Research Centre Laval Univer-sity (ldquoComite drsquoethique de la recherche Clinique du CHULrdquoQuebec City QC Canada Projects 950514 and 970514)and PomeranianMedical University (Bioethics CommissionPomeranian Medical University in Szczecin Poland Reso-lution number BN-0014306) Experiments were conductedwith the human subjectsrsquo understanding and consent Allpatients had signed a written consent form before eachprocedure

22 Materials Bile acid glucuronides were obtained asdescribed [11 21] Commercially available microsomes fromhuman liver (pool of 26 men and 24 women HO620) andkidney (pool of 4 female and 4 male donors H0610R) werepurchased from Xenotech (Walkersville MD) Human liversamples were from kidney donors as previously described(livers 2 to 8) [10 12 22] Total RNA from individual

human liver and kidney tissues was purchased from Ambion(Streetsville Canada) (liver 1 and kidney 1) and Stratagene(Santa Clara CA) (kidney 2) Total RNA from a poolof 5 human kidney donors was purchased from Ambion(Abingdon UK) The Tri Reagent was from Molecu-lar Research Center Inc (Cincinnati OH) Protein assayreagents were obtained from Bio-Rad Laboratories Inc(Marnes-la-Coquette France) UGT antibodies were as pre-viously described [10 23 24] The secondary anti-rabbit IgGhorse antibody was from Amersham (Oakville Canada)

23 Patients with Biliary Obstruction Seventeen patients (8men and 9 women mean age 64 plusmn 10 years) with clinicaland biochemical features of cholestasis were recruited aspreviously reported [20] (Supplementary Material 1) Diag-nosis biliary tree dilatation evidences and biliary stentingprocedures were extensively described in a previous report[20] Stored urine was available from only 12 of these patients(6 men and 6 women) for BA-G measurement Informedconsent was obtained from each patient

24 Bile Acid Glucuronides Measurement Bile acid-glucu-ronide concentrations were determined from serum andurine samples (100 120583L) using liquid chromatography coupledto tandem mass spectrometry (LC-MSMS) with an elec-trospray interface as previously reported [11 12 21] Thechromatographic system consisted of an Alliance 2690HPLCapparatus (Waters Milford MA) and the tandem massspectrometry system was an API3200 mass spectrometer(Applied Biosystems Concord Canada)

25 Glucuronidation Assays All glucuronidation assays wereperformed for 2 hours in the presence of 10 120583g microsomalproteins in a previously reported assay buffer [11 12] Kineticparameters were assessed in liver and kidney samples usingsubstrate concentrations ranging from 1 to 350 120583M Theenzyme kinetic model was selected as recommended [25]using the Sigma Plot 112 assisted by Enzyme Kinetics 13program (SSI San Jose CA)

26 RNA Extraction Reverse Transcription and Real-TimePCR Analyses Total RNA was isolated according to the TriReagent acid phenol protocol as specified by the supplier(Molecular Research Center Inc) The reverse transcription(RT) and quantitative PCR reactions were performed aspreviously described [11 26] The real-time PCR ampli-fications were performed using an ABI Prism 7500FASTinstrument from Applied Biosystems (Foster City CA) Foreach reaction the final volume of 20 120583L comprised 10 120583Lof SYBR Green PCR Mix 2 120583L (4 120583M) of each previouslyreported primer [11] and 6 120583L of the indicated dilution ofRT products For each gene in each tissue the amplificationefficiency was tested using 2 to 5 log of cDNA produced fromliver- or kidney-purified RNA and sequential dilutions ofUGT cDNA constructs (00001 to 10 pg120583l) The differencebetween standard curve and sample efficiency was below10 as recommended [27] The amount of target geneswas derived from linear regression with standard curves ofthese UGT constructs according to reported protocols [28]

Canadian Journal of Gastroenterology and Hepatology 3

CA-24G

3000

2000

1000

CDCA-3G

5000

4000

3000

2000

1000

CDCA-24G

30

20

10

1800

1400

1000

600

200

LCA-3G

300

200

100lowast

40

30

20

10

LCA-24G DCA-3G

800

200

600

400

30

20

10

DCA-24G

ns

HDCA-6G

30

20

10

HDCA-24G

4500

3000

1500

HCA-6G

5001000

25002000

35004000

30

20

10

HCA-24G

400

800

1200

1600

2000

20

40

60

100

120

80

1000

2000

3000

5000

6000

4000

ns

4060

100

160180

120140

80

20

10

30

20

100

350

300250200150

50

100

250

200

150

50

3000

70006000

4000

1000

5000

200004

10

08

06

02

40000

80000

60000

20000

20

40

30

10

(a) Serum

(b) Urine

ns

ns

ns

ns

nsns

ns

ns

lowast

lowastlowast

lowast

lowast

lowastlowast

lowastlowast

lowastlowastlowast

lowastlowast

lowastlowast

lowastlowast

Bile

acid

-glu

curo

nide

le

vels

(nM

)Bi

le ac

id-g

lucu

roni

de

leve

ls (n

M)

Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post

Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post

Figure 1 Endoscopic stenting of the bile duct differentially affects the serum (a) and urine (b) bile acid-glucuronide profiles in patients sufferingfrom biliary obstruction Serum ((a) 119899 = 17) and urine ((b) 119899 = 12) samples drawn before (pre) and after (post) endoscopic biliary stentingfrom patients with stenosed bile ducts were analyzed for their concentrations in 11 bile acid-glucuronide species using LC-MSMS Resultsrepresent the mean concentration plusmn SEM and are expressed as nM 119901 values were determined by the Wilcoxon matched-pairs signed-ranktest lowast119901 lt 005 lowastlowast119901 lt 001 lowastlowastlowast119901 lt 0001 ns not significant

Messenger RNA levels were calculated using the averagemolecular weight of one base-paired nucleotide (660 gmol)and Avogadrorsquos constant (6022 times 1023molminus1)

27 UGT Protein Determination For Western blottingmicrosomes (5 120583g) were size-separated by 10 SDS-polyacrylamide gels and transferred onto nitrocellulosemembranes All antibodies were used at 1 2000 dilutions[10 23 24] The housekeeping protein 120573-actin was used toensure the equal protein loading in each lane An anti-rabbitIgG horse antibody (1 10000) conjugated with peroxidase(Amersham) was used as the second antibody and theresulting immunocomplexes were visualized as described[12 26]

Determination of UGT1A3 protein levels was alsoachieved using LC-MSMS quantification according to thepreviously reported analytical strategy [10]

28 Data Analyses Bile acid-glucuronide levels were calcu-lated asmeanplusmn standard error of themean (SEM) BA-G con-centrations did not satisfy the normal distribution accordingto the Shapiro-Wilk test thus the Wilcoxon matched-pairssigned-rank test was used for statistical analyses of theresponse to treatment Correlations were assessed by Spear-manrsquos rank correlation coefficient using the JMP StatisticalDiscovery V701 program (SAS Institute Cary NC)

3 Results

31 Biliary Stenting Modulates Circulating and Urinary BA-G Levels in Patients with Biliary Obstruction The circulatingand urinary BA-G profiles in prestenting and poststentingsamples from biliary stenosed patients are illustrated inFigure 1 In sera the species distribution was CDCA-3G

gt HCA-6G gt CA-24G gt DCA-3G = HDCA-6G gt LCA-3G ≫ HDCA-24G = HCA-24G gt DCA-24G = LCA-24G geCDCA-24G In urine the most abundant species was HCA-6G gt HDCA-6G = CA-24G gt CDCA-3G gt DCA-3G Asin serum other species were minor components Bile flowrestoration caused significant reduction in serum CDCA-3G(27-fold) CA-24G (25-fold) and LCA-3G (24-fold) contents(Figure 1(a)) By contrast CDCA-24G (24-fold) LCA-24G(30-fold) and HDCA-6G (30-fold) concentrations weresignificantly increased in poststenting versus prestenting sera(Figure 1(a)) Other changes failed to reach the statisticalsignificance As we previously reported [11] serum levels ofBA-G in noncholestatic volunteers varied from 07 plusmn 01 nMfor HDCA-24G to 635 plusmn 86 nM for HCA-6G As observedin the present study the most abundant species were HCA-6G CDCA-3G DCA-3G and HDCA-6G Biliary stentingalso changed urine BA-G levels (Figure 1(b)) CDCA-24G(41-fold) LCA-3G (35-fold) LCA-24G (65-fold) andDCA-24G (59-fold) were increased and CDCA-3G (43-fold) andHCA-24G (83-fold) were reduced

32 Biliary Stenting Differentially Affects the BA-G versusBA Metabolic Ratios in Serum and Urine from BiliaryStenosed Patients The serum and urine samples were pre-viously analyzed for their content in the 6 unconjugatedprecursors of the glucuronide derivatives analyzed here [20]These concentrations were used to calculate the metabolicratio (ie BA-GBA) for each glucuronide (Figure 2 andSupplementary Material 2) This parameter translates theglucuronide production capability from the available poolof unconjugated precursor thus ensuring that changes inblood or urine levels of BA-G species do not only reflectchanges in their precursors [11] In serum samples onlythe 65- 206- and 172-fold reductions in the respective

4 Canadian Journal of Gastroenterology and Hepatology

Serum UrineMetabolic ratios

Before stenting After stenting Before stenting After stenting

CDCA-3GCDCA 252 plusmn 43 69 plusmn 12 42 plusmn 08

CDCA-24GCDCA 01 plusmn 01 01 plusmn 01 02 plusmn 01 11 plusmn 03

CA-24GCA 102 plusmn 33 44 plusmn 12

LCA-3GLCA 14 plusmn 08 03 plusmn 01 NA NA

LCA-24GLCA 02 plusmn 01 01 plusmn 01 NA NA

DCA-3GDCA 64 plusmn 28 43 plusmn 28 21 plusmn 05

DCA-24GDCA 02 plusmn 01 01 plusmn 00 07 plusmn 06 17 plusmn 02

HDCA-6GHDCA 41 plusmn 40 49 plusmn 15 50 plusmn 25 414 plusmn 113

HDCA-24GHDCA 01 plusmn 01 01 plusmn 01 00 plusmn 00 00 plusmn 00

HCA-6GHCA 155 plusmn 56 198 plusmn 35 41591 plusmn 37459 2954 plusmn 2353

HCA-24GHCA 03 plusmn 01 03 plusmn 01 15 plusmn 07 01 plusmn 01

39 plusmn 28lowastlowastlowast

05 plusmn 02lowastlowast

04 plusmn 02lowastlowast

08 plusmn 03lowastlowast

(a)15

lowastlowast

10

5

0

minus5

minus35

minus30

minus25

minus20

minus15

minus10

CDCA

-24G

CDCA

-3G

CA-2

4G

LCA-

3G

LCA-

24G

DCA

-3G

DCA

-24G

HD

CA-6

G

HCA

-6G

HD

CA-2

4G

HCA

-24G

lowastlowastlowast

lowastlowast

lowastlowast

Urine

MR

BA-

GB

A(fo

ld ch

ange

s afte

r bili

ary

stent

ing)

Serum

(b)

Before stenting

p = 001

10

20

30

40

50

60

Seru

m C

A-24

GC

A ra

tio r2 = 052

4 6 8 10 12 142Urine CA-24GCA ratio

(c)

After stenting

01

02

03

04

05

06

07

Seru

m C

A-24

GC

A ra

tio

p = 002r2 = 045

2 3 41Urine CA-24GCA ratio

(d)

Response

minus50

minus40

minus30

minus20

minus10

0

10

20

30

Seru

m C

A-24

GC

A ra

tio

p = 002r2 = 044

minus8 minus4 0 4minus12Urine CA-24GCA ratio

(e)Figure 2 While bile flow restoration modulates metabolic ratios of bile acid glucuronides in a glucuronide- and fluid-dependent manner ((a)and (b)) the CA-24GCA ratio is unique in being significantly associated with serum and urine (cndashe) Serum and urine samples drawn beforeand after endoscopic biliary stenting from patients with stenosed bile ducts were analyzed for their concentrations in 11 bile acid-glucuronidespecies using LC-MSMS Concentrations of unconjugated acids were determined as reported [20] and themetabolic ratio for each species (a)was calculated as the ratio of glucuronide versus unconjugated precursor (b) Black (serum 119899 = 17) and white (urine 119899 = 12) bars representthe mean plusmn SEM of fold changes (posttreatment values divided by pretreatment values) in MRs Statistically significant differences betweenprestenting versus poststenting samples were determined using the Wilcoxon matched-pairs signed-rank test lowastlowast119901 lt 001 lowastlowastlowast119901 lt 0001Other changes failed to reach statistical significance The absence of unconjugated LCA in urine impaired such analysis for LCA-3 and -24G[20] while urine HDCA-24GHDCA was null (cndashe) Correlation analyses of paired (119899 = 12) prestenting (c) and poststenting (d) serum andurine CA-24GCA values or response to stenting determined as the difference between post- and pretreatment levels (e) were performedusing the Spearman rank-order correlation (1199032) and 119901 values for comparisons are indicated

Canadian Journal of Gastroenterology and Hepatology 5

Table 1 Summary of the correlation studies between serum and urine bile acid-glucuronide profiles

Urine levels Before stenting After stenting Response(1199032 119901 value) (1199032 119901 value) (1199032 119901 value)

CDCA-3G 010 030 002 067 025 010CDCA-24G 001 069 005 050 002 063CA-24G 032 005 051 001 007 042LCA-3G 000 097 019 015 001 081LCA-24G 014 022 047 001 042 002DCA-3G 082 lt00001 014 023 001 071DCA-24G 006 043 016 019 058 0004HDCA-6G 029 007 031 006 035 004HDCA-24G 000 100 005 047 003 059HCA-6G 029 007 015 020 067 0001HCA-24G 000 083 027 008 004 054Correlation analyses were performed using the Spearman rank-order correlation (1199032) and 119901 values for comparisons are indicated Response is calculated asthe difference between post- and prestenting values CA cholic acid CDCA chenodeoxycholic acid DCA deoxycholic acid HCA hyocholic acid HDCAhyodeoxycholic acid LCA lithocholic acid G glucuronide

CDCA-3GCDCA CA-24GCA and DCA-3GDCA ratioswere statistically significant (Figures 2(a) and 2(b)) Otherchanges even the reduction in DCA-24GDCA (117-fold)and LCA-3GLCA (53-fold) or the 25-fold increase of theHDCA-24GHDCA ratio failed to reach statistical signif-icance (Figure 2(b)) In urine (Supplementary Material 2)the unique significant change corresponded to the 52-foldreduction of theCA-24GCA ratio (Figure 2(b)) while the 14-and 15-fold reductions in HCA-6 and -24GMRs also failed toreach statistical significance

33 Serum and Urine BA-G Profiles Are Only Feebly Asso-ciated Correlation studies of circulating and urinary BA-Glevels in paired samples identified only few significant associ-ations (Table 1) In prestenting samples only the serumDCA-3G was significantly and positively correlated (1199032 = 082)to its urinary concentration while in poststenting fluids nocorrelation for this conjugate was detected Serum and urineCA-24G (1199032 = 051) and LCA-24G (1199032 = 047) levels weresignificantly correlated but only in samples obtained afterbile flow restoration (Table 1) When the response to stenting(determined as the difference between post- and prestentingvalues) was analyzed only changes for LCA-24G (1199032 = 042)DCA-24G (1199032 = 058) HDCA-6G (1199032 = 035) and HCA-6G (1199032 = 067) were positively and significantly associated(Table 1) Finally when similar analyses were performed forMRs the unique significant association in prestenting (1199032 =052) and poststenting (1199032 = 045) levels and response(1199032 = 044) was obtained with the ratio CA-24GCA (Figures2(c)ndash2(e))

34 Bile Acid Glucuronidation by Human Liver and KidneyExtracts The limited associations between circulating andurinary profiles observed above suggest a potential extra-hepatic origin for urinary levels of BA-G To test such ahypothesis we sought to compare the kinetic parameters ofBA glucuronidation by human liver (pool of 50 donors) and

kidney (pool of 8 donors) microsomes (Figures 3ndash5) Nodifferences in kinetic models were observed with the BA-specific glucuronidation reactions being distributed among 3models

(i) The sigmoidal pattern for CA-24G and CDCA-24 and-3G (Figure 3) DCA-3 and -24G (Figures 4(c) and 4(d)) andHDCA-6 and -24G (Figures 5(a) and 5(b)) production

(ii) The Michaelis-Menten model for the formation ofLCA-24G (Figure 4(b)) and HCA-6 and -24G (Figures 5(c)and 5(d))

(iii) The substrate-inhibition model for LCA-3G withrespective 119870119868 values of 2403 plusmn 535 and 2554 plusmn 699 120583M forliver and kidney extracts (Figure 4(a))

With the exception of CA-24G HDCA-24G and HCA-24G kidney UGTs generally displayed lower 119870119872 values Forexample kidney microsomes exhibited a 6-fold lower 119870119872value than liver ones for LCA-3G production (Figure 4(a))Another notable difference between liver and kidney micro-somes relates to the 10-fold higher 119881max value of HDCA-24Gproduction obtained with the hepatic microsomal prepara-tion (Figure 5(b)) Nevertheless the most efficient reactionswere obtained with the conversion of HDCA and HCA into6G derivatives that occurred at high velocities (ie 119881max)with both liver and kidney microsomes (Figures 5(a) and5(c)) In both tissue extracts the 6120572-hydroxylated acids weremore actively converted into 6-glucuronide when comparedto their 24-glucuronide counterparts (Figure 5) Interestinglythe opposite was observed for CDCA (Figures 3(a) and 3(b))and LCA (Figures 4(a) and 4(b)) The 119881max values for DCA-3and -24G formation were however similar (Figures 4(c) and4(d))

35 Differential BA-Conjugating UGT Expression in theHuman Liver and Kidney Results from kinetic experimentsindicate that the human kidney possesses an efficient BA-conjugating UGT system We next investigated whetherthe BA-conjugating UGTs 1A3 1A4 2B4 and 2B7 [8ndash11] are differentially expressed in human liver and kidney

6 Canadian Journal of Gastroenterology and Hepatology

02

04

06

08

10CD

CA-3

G fo

rmat

ion

(nm

olh

mg

prot

eins

)

100[CDCA] (M)

200 300 400

Liver nA = 17 plusmn 01

KMB = 821 plusmn 60

VGRC = 08 plusmn 01

Kidney nA = 19 plusmn 07KM

B = 443 plusmn 96VGR

C = 05 plusmn 01

(a)

10

20

30

CDCA

-24G

form

atio

n(n

mol

hm

g pr

otei

ns)

100[CDCA] (M)

200 300 400

Liver nA = 14 plusmn 01KM

B = 1682 plusmn 186VGR

C = 37 plusmn 02

Kidney nA = 13 plusmn 03KM

B = 897 plusmn 298VGR

C = 25 plusmn 04

(b)

05

10

15

20

CA-2

4G fo

rmat

ion

(nm

olh

mg

prot

eins

)

100[CA] (M)

200 300 400

Liver nA = 12 plusmn 01KM

B = 1141 plusmn 168VGR

C = 16 plusmn 01

Kidney nA = 10 plusmn 01KM

B = 1481 plusmn 397VGR

C = 11 plusmn 01

(c)

Figure 3 Kinetic analyses of CDCA-3G (a) CDCA-24G (b) and CA-24G (c) formation by microsomes from human liver and kidney Humanliver (black squares pool of 50 donors) and kidney (white diamonds pool of 8 donors) microsomes (10 120583g) were incubated in the presenceof increasing concentrations (1 to 350 120583M) of chenodeoxycholic acid (CDCA (a) and (b)) or cholic acid (CA (c)) for 2 hours at 37∘C Theformation of CDCA-3G (a) CDCA-24G (b) and CA-24G (c) was analyzed by LC-MSMS For each panel graphs represent the rate ofproduct formation (119910-axis) versus substrate concentration (119909-axis) of two experiments performed in triplicate The listed kinetic parameterswere determined as indicated in ldquoMaterials and Methodsrdquo A119899 Hill coefficient B119870119872 calculated Michaelis constant expressed as 120583M C119881maxcalculated maximal velocity expressed as nmolhmg proteins

Canadian Journal of Gastroenterology and Hepatology 7

200 300 400100[LCA] (M)

40

80

120

160

LCA-

3G fo

rmat

ion

(nm

olh

mg

prot

eins

)

LiverKM

B = 487 plusmn 90VGR

C = 171 plusmn 29

Kidney

KIA = 2403 plusmn 535

KMB = 82 plusmn 22

VGRC = 121 plusmn 11

KIA = 2554 plusmn 699

(a)

200 300 400100[LCA] (M)

60

120

180

240

300

LCA-

24G

form

atio

n (n

mol

hm

g pr

otei

ns)

LiverKM

B = 403 plusmn 67VGR

C = 321 plusmn 16

KidneyKM

B = 305 plusmn 56VGR

C = 162 plusmn 08

(b)

100 200 300 400[DCA] (M)

10

20

30

40

50

60

DCA

-3G

form

atio

n(n

mol

hm

g pr

otei

ns)

Liver nD 17 plusmn 03KM

B = 831 plusmn 113VGR

C = 31 plusmn 02

Kidney nD = 17 plusmn 03KM

B = 823 plusmn 136VGR

C = 36 plusmn 03

(c)

20

40

60

80

DCA

-24G

form

atio

n(n

mol

hm

g pr

otei

ns)

200 300 400100[DCA] (M)

Liver nD 12 plusmn 02KM

B = 711 plusmn 121VGR

C = 58 plusmn 05

Kidney nD = 17 plusmn 03KM

B = 582 plusmn 87VGR

C = 38 plusmn 03

(d)Figure 4Kinetic analyses of LCA-3G (a) LCA-24G (b) DCA-3G (c) and DCA-24G (d) formation by microsomes from human liver and kidneyHuman liver (black squares pool of 50 donors) and kidney (white diamonds pool of 8 donors) microsomes (10 120583g) were incubated in thepresence of increasing concentrations (1 to 350120583M) of lithocholic acid (LCA (a) and (b)) or deoxycholic acid (DCA (c) and (d)) for 2 hoursat 37∘C The formation of LCA-3G (a) LCA-24G (b) DCA-3G (c) and DCA-24G (d) was analyzed by LC-MSMS For each panel graphsrepresent the rate of product formation (119910-axis) versus substrate concentration (119909-axis) of two experiments performed in triplicateThe listedkinetic parameters were determined as indicated in ldquoMaterials and Methodsrdquo A119870119868 constant of inhibition expressed as 120583M B119870119872 calculatedMichaelis constant expressed as 120583M C119881max calculated maximal velocity expressed as nmolhmg proteins D119899 Hill coefficient

8 Canadian Journal of Gastroenterology and Hepatology

1750

3500

5250

7000

HD

CA-6

G fo

rmat

ion

(nm

olh

mg

prot

eins

)

200 300 400100[HDCA] (M)

Liver nA 29 plusmn 04KM

B = 551 plusmn 33VGR

C = 5287 plusmn 211

Kidney nA = 27 plusmn 05KM

B = 447 plusmn 38VGR

C = 3084 plusmn 156

(a)

100

200

300

400

500

HD

CA-2

4G fo

rmat

ion

(nm

olh

mg

prot

eins

)

200 300 400100[HDCA] (M)

Liver nA 38 plusmn 11KM

B = 808 plusmn 64VGR

C = 316 plusmn 22

Kidney nA = 23 plusmn 08KM

B = 939 plusmn 184VGR

C = 37 plusmn 05

(b)

800

1600

2400

3200

HCA

-6G

form

atio

n(n

mol

hm

g pr

otei

ns)

200 300 400100[HCA] (M)

LiverKM

B = 1202 plusmn 156VGR

C = 2285 plusmn 134

KidneyKM

B = 960 plusmn 192VGR

C = 2844 plusmn 241

(c)

20

40

60

HCA

-24G

form

atio

n(n

mol

hm

g pr

otei

ns)

200 300 400100[HCA] (M)

LiverKM

B = 617 plusmn 65VGR

C = 49 plusmn 02

KidneyKM

B = 978 plusmn 206VGR

C = 33 plusmn 03

(d)Figure 5 Kinetic analyses of HDCA-6G (a) HDCA-24G (b) HCA-6G (c) and HCA-24G (d) formation by microsomes from human liver andkidney Human liver (black squares pool of 50 donors) and kidney (white diamonds pool of 8 donors) microsomes (10 120583g) were incubatedin the presence of increasing concentrations (1 to 350120583M) of hyodeoxycholic acid (HDCA (a) and (b)) or hyocholic acid (HCA (c) and(d)) for 2 hours at 37∘CThe formation of HDCA-6G (a) HDCA-24G (b) HCA-6G (c) and HCA-24G (d) was analyzed by LC-MSMS Foreach panel graphs represent the rate of product formation (119910-axis) versus substrate concentration (119909-axis) of two experiments performedin triplicate The listed kinetic parameters were determined as indicated in ldquoMaterials and Methodsrdquo A119899 Hill coefficient B119870119872 calculatedMichaelis constant expressed as 120583M C119881max calculated maximal velocity expressed as nmolhmg proteins

Canadian Journal of Gastroenterology and Hepatology 9

Kid

ney

1A3

Kid

ney

Kid

ney

Kid

ney

Live

r

Live

r

Live

r

Live

r

1A4 2B4 2B7UGT

103

104

105

106

107U

GT

tran

scrip

ts (lo

g of

copy

num

ber

g RN

A)

(a)

1A3

2B42B7

2B7

1A

2B

LiverKidneyAnti-UGT kDa

55

55

55

55

55

40Anti-actin

(b)

Live

r

Kidn

ey

ND

5

10

15

20

UG

T1A

3 pr

otei

n le

vels

(pm

olm

g m

icro

som

es)

(c)

Figure 6 Differential expression of the bile acid-conjugating UGT enzymes in the human liver and kidney (a) UGTs 1A3 1A4 2B4 and 2B7mRNA levels in total RNA samples from human kidney (1 pool of 5 donors and 2 individual donors) and liver (8 individual donors) weredetermined through qRT-PCR analyses as described in ldquoMaterials andMethodsrdquo Results are expressed as log10 ofmRNA copy number per 120583gof total RNA Each symbol represents 1 donor with the exception of the white triangle that corresponds to a pool of the 5 kidney donors (b)The UGT protein contents in microsomal preparations (5120583g) from human kidney (pool of 4 female and 4 male donors) and liver (pool of 26male and 24 female donors) were determined after size separation on SDS-PAGE and hybridization with the anti-UGT1A3 anti-UGT2B47anti-UGT2B7 anti-UGT1A and anti-UGT2B antibodies (1 2000 dilution) The housekeeping protein 120573-actin was used to ensure the equalprotein loading in each lane Data presented are representative of two experiments (c) The UGT1A3 protein content in human liver andkidney microsomes was determined using LC-MSMS analyses as previously described [10] Data represent the mean plusmn SD of two differentexperiments performed in triplicate ND not detected

(Figure 6) With the exception of UGT2B4 transcripts thatwere detected only in 1 kidney mRNA sample other UGTmessengers were detected in all kidney and liver prepara-tions (Figure 6(a)) As expected [28 29] their mRNA levelssustained a strong interindividual variability Western blotanalyses evidenced however major differences between liverand kidney (Figure 6(b)) A clear UGT1A3 immunoreactivecomplex was obtained with liver extracts while this UGTprotein was not detected inmicrosomal proteins from kidney(Figure 6(b)) While the UGT2B7 protein was found atsimilar levels in both tissues the use of an anti-UGT2B42B7antibody indicated that the UGT2B4 protein may be moreabundant in liver than in kidney extracts (Figure 6(b))The lack of an accurate and specific anti-UGT1A4 antibody

impaired the quantification of this BA-conjugating UGThowever we were able to further confirm the absence ofthe UGT1A3 protein in kidney microsomes through the useof our previously established LC-MSMS-based proteomicmethod (Figure 6(c)) [10]

4 Discussion

Urinary and circulating levels of BA-Gs detected in thepresent study are similar to previous findings in terms ofboth glucuronide concentrations and species distribution [1314 16] However an important improvement of the presentinvestigations resides in our ability to discriminate betweenether and ester glucuronides of a single BA For example

10 Canadian Journal of Gastroenterology and Hepatology

while glucuronide conjugates of CDCA CA and DCA werepreviously identified as accumulating metabolites in serafrom patients undergoing bile drainage [20] the nature ofthese glucuronides has not been resolved until now Ourresults point out the ester CDCA-3G and DCA-3G andthe acyl CA-24G as the most abundant glucuronide speciesfound in patients with biliary obstruction Because acyl butnot ester glucuronides can be potentially toxic molecules(reviewed in [30]) such information may be critical inanticipating the consequences of their accumulation duringcholestasis

Interestingly serum levels of some of the bile acidglucuronides such as CDCA-3G CA-24G and LCA-3Gwere significantly reduced after biliary stenting while someothers like CDCA-24G LCA-24G and HDCA-6G weresignificantly increased While additional investigations arewarranted to decipher the mechanisms beyond such a glu-curonide species-dependent responsiveness one can spec-ulate that these changes may actually reflect the differen-tial manner in which the UGT enzymes andor the BA-Gtransporters may be altered upon biliary obstruction andafter stenting Therefore it would have been of interest tocompare how these proteins behave in tissues (ie liver andkidney) from cholestatic donors to validate such a hypothesisActually one limitation of the current study is that the liverand kidneymicrosomes used are not fromdonorswith biliaryobstruction When compared to other glucuronide conju-gates CA-24G presented a unique behavior (1) in contrastto other acyl BA-Gs that are only minor components of theurine and serum glucuronide pools CA-24G is the 3rd mostabundant glucuronidated acid in prestenting samples (2) thisspecies is the most spectacularly affected when poststentingversus prestenting sera profiles are compared (3) the serumCA-24GCA ratio sustains the stronger reduction after biliarystenting (4) this MR is also unique in being significantlyreduced in posttreatment urines and (5) only the CA-24GCA ratio exhibits positive serumurine association interms of prestenting and poststenting distribution as wellas treatment response These last observations support ahepatic origin for the formation of CA-24G and the strongCA-24G accumulation in prestenting fluids and its spec-tacular reduction after bile flow restoration suggests thatunder noncholestatic situations this compound formed inliver cells is normally secreted in bile This hypothesis isfurther supported by previous investigations in which theintravenous injections of cholate glucuronide to rats resultedin a rapid and efficient secretion in bile of the unchangedglucuronide conjugate [31] By contrast when the sameprocedure was applied to bile duct ligated animals 95 of theradioactivity was recovered in urine [31] The present studyindicates that similar rerouting of CA-24G elimination tourine may also occur in cholestatic patients since circulatinglevels detected in prestenting cholestatic donors were almost50-fold higher than those previously quantified in samplesfrom noncholestatic volunteers (sim2 nM [11]) Privilegingsecretion from liver cells into the blood for subsequenturinary elimination may actually protect the liver againstthe accumulation of such abundant and potentially toxicglucuronide species during cholestasis In the same vein the

improved formation of other conjugates such as CDCA-3Gand DCA-3G which we recently identified as nontoxic BAs[32] may also participate in the hepatic detoxification oftheir unconjugated CDCA and DCA precursors Consistentwith the above stated hypothesis circulating MRs for these2 conjugates (ie 252 and 64 for CDCA-3G and DCA-3Gresp) are spectacularly increased when compared to thosepreviously found in noncholestatic donors (06 and 01) [11]However the fact that circulating and urinary MRs for thesespecies were not associated and the fact that biliary stentingexerted only minor effects on these ratios in urine are alsoindicative of an extrahepatic origin for urine CDCA- andDCA-3G species at least after bile flow restoration

We next compared the human liver and kidney in termsof bile acid glucuronidation activity and BA-conjugatingUGT expression With 2 exceptions (ie the 6- and 10-fold difference in 119870119872 and 119881max values for LCA-3G andHDCA-24G formations resp) these experiments revealthe remarkable similarity of renal and hepatic bile acidglucuronidation processes and the clear preference of bothtissues in producing HDCA-6 and HCA-6G Such preferencemay actually explain the predominance of these conjugates invarious human fluids as previously described [11] Howeverthe most intriguing observation issuing from these analysesrelates to the UGT1A3 enzyme Indeed not only was thisenzyme detected in kidney extracts only at the mRNA(but not protein) level but also the absence of such anactive enzyme previously identified as the main isoform foracyl BA glucuronidation [8ndash10 12] is inconsistent with theelevated acyl glucuronide production capability of kidneymicrosomes A plausible explanation for the conflictingobservation on UGT1A3 mRNA and protein expression inkidney samples relates to the fact that protein and RNAextracts were from different sources and thus were notpaired It is therefore possible that only donors for mRNAexpress these isoforms in the kidney Meanwhile unlikelysuch a possibility is supported by the controversial nature ofthe UGT1A3 expression in the human kidney [29 33ndash35]Additional investigations are therefore warranted to clarifywhether this BA-conjugating protein is found or not in thehuman kidney

Nevertheless the present study clearly establishes themajor contributions that liver and kidney play in glu-curonidating bile acid during cholestasis Interestingly Zhouand colleagues reported that intestinal glucuronidation alsoplays an important role in controlling the local bile acidhomeostasis and the pathological development of colitis [36]These authors elegantly demonstrated that in the intestinebile acids glucuronidation leads to reduced intracellular levelsof ligands for the nuclear receptor FXR Reduced FXR activa-tion limits the secretion of FGF1519 in the portal circulationwhich in turns leads to an increase of de novo formationof bile acid in the liver [36] Thus in contrast to the liverand kidney where glucuronidation is thought to facilitate theremoval of bile acids the intestinal glucuronidation seems toincrease the delivery of bile acidsHowever it should be notedthat during obstructive cholestasis the delivery of hepaticbile acids to the intestine is compromised and thus onlyminor intestinal glucuronidation may occur

Canadian Journal of Gastroenterology and Hepatology 11

5 Conclusion

In conclusion the present study evidences the major impactthat bile flow restoration exerts on the circulating and urinarybile acid-glucuronide profiles and demonstrates the elevatedcapability of the human kidney to glucuronidate theseendogenous compounds Future investigations are requiredto clarify the mechanism allowing kidney extracts to convertbile acids into acyl glucuronides in the absence of theUGT1A3 protein

Abbreviations

BA Bile acidBO Biliary obstructionCA Cholic acidCDCA Chenodeoxycholic acidDCA Deoxycholic acidFXR Farnesoid X receptorG GlucuronideHDCA Hyodeoxycholic acidHCA Hyocholic acidLCA Lithocholic acidLC-MSMS Liquid chromatography-tandem mass

spectrometryUDPGA UDP-glucuronic acidUGT UDP-glucuronosyltransferaseMRPs Multidrug resistance related proteins

Conflicts of Interest

The authors declare that there are no conflicts of interest andthat there is no pertinent financial arrangement concerningthe work presented here

Acknowledgments

The authors wish to thank Dr Virginie Bocher for criticalreading of the manuscript This study was supported bygrants from theCanadian Institute ofHealth Research (CIHR119331) the Canadian Liver Foundation the Natural Sci-ences and Engineering Research Council of Canada (NSERC402213-2012) and the Canadian Foundation for Innovation(CFI 25712)Martin Perreault is holder of a scholarship fromthe ldquoFonds pour la Recherche en Sante du Quebecrdquo (FRSQ23444) Olivier Barbier is holder of a salary grant fromCIHR (New Investigator MSH95330) Piotr Milkiewiczwas supported by Grants DEC-201101BNZ504216 and201102ANZ500321 from the National Science Centre inPoland

Supplementary Materials

The supplementary materials present details of patient withbiliary obstruction (Supplementary Material 1) and thechanges in the urine profile of bile acid glucuronides and intheir metabolic ratios before and after a biliary stenting pro-cedure in patients with biliary obstruction (SupplementaryMaterial 2) Supplementary Material 1 patients with biliary

obstruction (adapted from [20]) Supplementary Material 2changes in the urine profile of bile acid glucuronides andin their metabolic ratios before and after a biliary stentingprocedure in patients with biliary obstruction (119899 = 12)(Supplementary Materials)

References

[1] D W Russell ldquoThe enzymes regulation and genetics of bileacid synthesisrdquo Annual Review of Biochemistry vol 72 pp 137ndash174 2003

[2] M J Monte J J G Marin A Antelo and J Vazquez-TatoldquoBile acids Chemistry physiology and pathophysiologyrdquoWorldJournal of Gastroenterology vol 15 no 7 pp 804ndash816 2009

[3] A F Hofmann ldquoDetoxification of lithocholic acid a toxicbile-acid relevance to drug hepatotoxicityrdquo Drug MetabolismReviews vol 36 no 3-4 pp 703ndash722 2004

[4] C Pauli-Magnus B Stieger Y Meier G A Kullak-Ublick andP J Meier ldquoEnterohepatic transport of bile salts and genetics ofcholestasisrdquo Journal of Hepatology vol 43 no 2 pp 342ndash3572005

[5] G Paumgartner ldquoPharmacotherapy of cholestatic liver dis-easesrdquo Journal of Digestive Diseases vol 11 no 3 pp 119ndash1252010

[6] M Verreault J Kaeding P Caron et al ldquoRegulation ofendobiotics glucuronidation by ligand-activated transcriptionfactors Physiological function and therapeutic potentialrdquoDrugMetabolism Reviews vol 42 no 1 pp 106ndash118 2010

[7] P I Mackenzie K W Bock B Burchell et al ldquoNomenclatureupdate for the mammalian UDP glycosyltransferase (UGT)gene superfamilyrdquo Pharmacogenetics and Genomics vol 15 no10 pp 677ndash685 2005

[8] M H Court S Hazarika S Krishnaswamy M Finel andJ A Williams ldquoNovel polymorphic human UDP-glucurono-syltransferase 2A3 Cloning functional characterization ofenzyme variants comparative tissue expression and geneinductionrdquoMolecular Pharmacology vol 74 no 3 pp 744ndash7542008

[9] W E Gall G Zawada B Mojarrabi et al ldquoDifferential glu-curonidation of bile acids androgens and estrogens by humanUGT1A3 and 2B7rdquo The Journal of Steroid Biochemistry andMolecular Biology vol 70 no 1ndash3 pp 101ndash108 1999

[10] J Trottier D El Husseini M Perreault et al ldquoThe humanUGT1A3 enzyme conjugates norursodeoxycholic acid into aC23-ester glucuronide in the liverrdquo The Journal of BiologicalChemistry vol 285 no 2 pp 1113ndash1121 2010

[11] J Trottier M Perreault I Rudkowska et al ldquoProfiling serumbile acid glucuronides in humans Gender divergences geneticdeterminants and response to fenofibraterdquo Clinical Pharmacol-ogy ampTherapeutics vol 94 no 4 pp 533ndash543 2013

[12] P Caron J Trottier M Verreault J Belanger J Kaeding andO Barbier ldquoEnzymatic production of bile acid glucuronidesused as analytical standards for liquid chromatography-massspectrometry analysesrdquo Molecular Pharmaceutics vol 3 no 3pp 293ndash302 2006

[13] B Alme and J Sjovall ldquoAnalysis of bile acid glucuronidesin urine identification of 3120572612057212120572-trihydroxy-5120573-cholanoicacidrdquoThe Journal of Steroid Biochemistry andMolecular Biologyvol 13 no 8 pp 907ndash916 1980

[14] A Stiehl R Raedsch G Rudolph P Czygan and S WalkerldquoAnalysis of bile acid glucuronides in urine group separation

12 Canadian Journal of Gastroenterology and Hepatology

on a lipophilic anion exchangerrdquo Clinica Chimica Acta vol 123no 3 pp 275ndash285 1982

[15] A Stiehl R Raedsch G Rudolph U Gundert-Remy and MSenn ldquoBiliary and urinary excretion of sulfated glucuronidatedand tetrahydroxylated bile acids in cirrhotic patientsrdquo Hepatol-ogy vol 5 no 3 pp 492ndash495 1985

[16] H Takikawa T Beppu and Y Seyama ldquoUrinary concentrationsof bile acid glucuronides and sulfates in hepatobiliary diseasesrdquoGastroenterologia Japonica vol 19 no 2 pp 104ndash109 1984

[17] H Wietholtz H Marschall R Reuschenbach H Matern andS Matern ldquoUrinary excretion of bile acid glucosides and glu-curonides in extrahepatic cholestasisrdquo Hepatology vol 13 no4 pp 656ndash662 1991

[18] K M Knights A Rowland and J O Miners ldquoRenal drugmetabolism in humans The potential for drug-endobioticinteractions involving cytochrome P450 (CYP) and UDP-glu-curonosyltransferase (UGT)rdquo British Journal of Clinical Phar-macology vol 76 no 4 pp 587ndash602 2013

[19] S Matern H Matern E H Farthmann and W GerokldquoHepatic and extrahepatic glucuronidation of bile acids in manCharacterization of bile acid uridine 5rsquo-diphosphate-glucu-ronosyltransferase in hepatic renal and intestinalmicrosomesrdquoThe Journal of Clinical Investigation vol 74 no 2 pp 402ndash4101984

[20] J Trottier A Białek P Caron R J Straka P Milkiewicz and OBarbier ldquoProfiling circulating and urinary bile acids in patientswith biliary obstruction before and after biliary stentingrdquo PLoSONE vol 6 no 7 Article ID e22094 2011

[21] M Perreault L Gauthier-Landry J Trottier et al ldquoThe humanUDP-glucuronosyltransferase UGT2A1 and UGT2A2 enzymesare highly active in bile acid glucuronidationrdquoDrugMetabolismand Disposition vol 41 no 9 pp 1616ndash1620 2013

[22] M E Campbell D M Grant T Inaba and W KalowldquoBiotransformation of caffeine paraxanthine theophylline andtheobromine by polycyclic aromatic hydrocarbon-induciblecytochrome(s) P-450 in human liver microsomesrdquo DrugMetabolism and Disposition vol 15 no 2 pp 237ndash249 1987

[23] J Lepine O Bernard M Plante et al ldquoSpecificity andregioselectivity of the conjugation of estradiol estrone andtheir catecholestrogen and methoxyestrogen metabolites byhuman uridine diphospho-glucuronosyltransferases expressedin endometriumrdquo The Journal of Clinical Endocrinology ampMetabolism vol 89 no 10 pp 5222ndash5232 2004

[24] J Thibaudeau J Lepine J Tojcic et al ldquoCharacterizationof common UGT1A8 UGT1A9 and UGT2B7 variants withdifferent capacities to inactivate mutagenic 4-hydroxylatedmetabolites of estradiol and estronerdquo Cancer Research vol 66no 1 pp 125ndash133 2006

[25] J O Miners P I MacKenzie and K M Knights ldquoTheprediction of drug-glucuronidation parameters in humansUDP-glucuronosyltransferase enzyme-selective substrate andinhibitor probes for reaction phenotyping and in vitroin vivoextrapolation of drug clearance and drug-drug interactionpotentialrdquo Drug Metabolism Reviews vol 42 no 1 pp 189ndash2012010

[26] M Verreault K Senekeo-Effenberger J Trottier et al ldquoTheliver X-receptor alpha controls hepatic expression of the humanbile acid-glucuronidating UGT1A3 enzyme in human cells andtransgenic micerdquo Hepatology vol 44 no 2 pp 368ndash378 2006

[27] R G Rutledge ldquoMathematics of quantitative kinetic PCR andthe application of standard curvesrdquo Nucleic Acids Research vol31 no 16 pp 93endash93 2003

[28] T Izukawa M Nakajima R Fujiwara et al ldquoQuantitative anal-ysis of UDP-glucuronosyltransferase (UGT) 1A and UGT2Bexpression levels in human liversrdquo Drug Metabolism and Dis-position vol 37 no 8 pp 1759ndash1768 2009

[29] S Ohno and S Nakajin ldquoDetermination of mRNA expressionof human UDP-glucuronosyltransferases and application forlocalization in various human tissues by real-time reversetranscriptase-polymerase chain reactionrdquoDrugMetabolism andDisposition vol 37 no 1 pp 32ndash40 2009

[30] S L Regan J L Maggs T G Hammond C Lambert D PWilliams and B K Park ldquoAcyl glucuronidesThe good the badand the uglyrdquo Biopharmaceutics amp Drug Disposition vol 31 no7 pp 367ndash395 2010

[31] J M Little M V Chari and R Lester ldquoExcretion of cholateglucuroniderdquo Journal of Lipid Research vol 26 no 5 pp 583ndash592 1985

[32] M Perreault A Białek J Trottier et al ldquoRole of glucuronidationfor hepatic detoxification and urinary elimination of toxic bileacids during biliary obstructionrdquo PLoS ONE vol 8 no 11Article ID e80994 2013

[33] H Girard E Levesque J Bellemare K Journault B Caillierand C Guillemette ldquoGenetic diversity at the UGT1 locus isamplified by a novel 31015840 alternative splicing mechanism leadingto nine additional UGT1A proteins that act as regulators ofglucuronidation activityrdquo Pharmacogenetics and Genomics vol17 no 12 pp 1077ndash1089 2007

[34] A Nakamura M Nakajima H Yamanaka R Fujiwara and TYokoi ldquoExpression of UGT1A and UGT2B mRNA in humannormal tissues and various cell linesrdquo Drug Metabolism andDisposition vol 36 no 8 pp 1461ndash1464 2008

[35] D E Harbourt J K Fallon S Ito et al ldquoQuantificationof human uridine-diphosphate glucuronosyl transferase 1Aisoforms in liver intestine and kidney using nanobore liq-uid chromatography-tandem mass spectrometryrdquo AnalyticalChemistry vol 84 no 1 pp 98ndash105 2012

[36] X Zhou L Cao C Jiang et al ldquoPPAR120572-UGT axis activationrepresses intestinal FXR-FGF15 feedback signalling and exacer-bates experimental colitisrdquoNature Communications vol 5 2014

Stem Cells International

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

MEDIATORSINFLAMMATION

of

EndocrinologyInternational Journal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Disease Markers

Hindawiwwwhindawicom Volume 2018

BioMed Research International

OncologyJournal of

Hindawiwwwhindawicom Volume 2013

Hindawiwwwhindawicom Volume 2018

Oxidative Medicine and Cellular Longevity

Hindawiwwwhindawicom Volume 2018

PPAR Research

Hindawi Publishing Corporation httpwwwhindawicom Volume 2013Hindawiwwwhindawicom

The Scientific World Journal

Volume 2018

Immunology ResearchHindawiwwwhindawicom Volume 2018

Journal of

ObesityJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Computational and Mathematical Methods in Medicine

Hindawiwwwhindawicom Volume 2018

Behavioural Neurology

OphthalmologyJournal of

Hindawiwwwhindawicom Volume 2018

Diabetes ResearchJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Research and TreatmentAIDS

Hindawiwwwhindawicom Volume 2018

Gastroenterology Research and Practice

Hindawiwwwhindawicom Volume 2018

Parkinsonrsquos Disease

Evidence-Based Complementary andAlternative Medicine

Volume 2018Hindawiwwwhindawicom

Submit your manuscripts atwwwhindawicom

Page 3: Urinary Elimination of Bile Acid Glucuronides under Severe ...

Canadian Journal of Gastroenterology and Hepatology 3

CA-24G

3000

2000

1000

CDCA-3G

5000

4000

3000

2000

1000

CDCA-24G

30

20

10

1800

1400

1000

600

200

LCA-3G

300

200

100lowast

40

30

20

10

LCA-24G DCA-3G

800

200

600

400

30

20

10

DCA-24G

ns

HDCA-6G

30

20

10

HDCA-24G

4500

3000

1500

HCA-6G

5001000

25002000

35004000

30

20

10

HCA-24G

400

800

1200

1600

2000

20

40

60

100

120

80

1000

2000

3000

5000

6000

4000

ns

4060

100

160180

120140

80

20

10

30

20

100

350

300250200150

50

100

250

200

150

50

3000

70006000

4000

1000

5000

200004

10

08

06

02

40000

80000

60000

20000

20

40

30

10

(a) Serum

(b) Urine

ns

ns

ns

ns

nsns

ns

ns

lowast

lowastlowast

lowast

lowast

lowastlowast

lowastlowast

lowastlowastlowast

lowastlowast

lowastlowast

lowastlowast

Bile

acid

-glu

curo

nide

le

vels

(nM

)Bi

le ac

id-g

lucu

roni

de

leve

ls (n

M)

Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post

Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post Pre

Post

Figure 1 Endoscopic stenting of the bile duct differentially affects the serum (a) and urine (b) bile acid-glucuronide profiles in patients sufferingfrom biliary obstruction Serum ((a) 119899 = 17) and urine ((b) 119899 = 12) samples drawn before (pre) and after (post) endoscopic biliary stentingfrom patients with stenosed bile ducts were analyzed for their concentrations in 11 bile acid-glucuronide species using LC-MSMS Resultsrepresent the mean concentration plusmn SEM and are expressed as nM 119901 values were determined by the Wilcoxon matched-pairs signed-ranktest lowast119901 lt 005 lowastlowast119901 lt 001 lowastlowastlowast119901 lt 0001 ns not significant

Messenger RNA levels were calculated using the averagemolecular weight of one base-paired nucleotide (660 gmol)and Avogadrorsquos constant (6022 times 1023molminus1)

27 UGT Protein Determination For Western blottingmicrosomes (5 120583g) were size-separated by 10 SDS-polyacrylamide gels and transferred onto nitrocellulosemembranes All antibodies were used at 1 2000 dilutions[10 23 24] The housekeeping protein 120573-actin was used toensure the equal protein loading in each lane An anti-rabbitIgG horse antibody (1 10000) conjugated with peroxidase(Amersham) was used as the second antibody and theresulting immunocomplexes were visualized as described[12 26]

Determination of UGT1A3 protein levels was alsoachieved using LC-MSMS quantification according to thepreviously reported analytical strategy [10]

28 Data Analyses Bile acid-glucuronide levels were calcu-lated asmeanplusmn standard error of themean (SEM) BA-G con-centrations did not satisfy the normal distribution accordingto the Shapiro-Wilk test thus the Wilcoxon matched-pairssigned-rank test was used for statistical analyses of theresponse to treatment Correlations were assessed by Spear-manrsquos rank correlation coefficient using the JMP StatisticalDiscovery V701 program (SAS Institute Cary NC)

3 Results

31 Biliary Stenting Modulates Circulating and Urinary BA-G Levels in Patients with Biliary Obstruction The circulatingand urinary BA-G profiles in prestenting and poststentingsamples from biliary stenosed patients are illustrated inFigure 1 In sera the species distribution was CDCA-3G

gt HCA-6G gt CA-24G gt DCA-3G = HDCA-6G gt LCA-3G ≫ HDCA-24G = HCA-24G gt DCA-24G = LCA-24G geCDCA-24G In urine the most abundant species was HCA-6G gt HDCA-6G = CA-24G gt CDCA-3G gt DCA-3G Asin serum other species were minor components Bile flowrestoration caused significant reduction in serum CDCA-3G(27-fold) CA-24G (25-fold) and LCA-3G (24-fold) contents(Figure 1(a)) By contrast CDCA-24G (24-fold) LCA-24G(30-fold) and HDCA-6G (30-fold) concentrations weresignificantly increased in poststenting versus prestenting sera(Figure 1(a)) Other changes failed to reach the statisticalsignificance As we previously reported [11] serum levels ofBA-G in noncholestatic volunteers varied from 07 plusmn 01 nMfor HDCA-24G to 635 plusmn 86 nM for HCA-6G As observedin the present study the most abundant species were HCA-6G CDCA-3G DCA-3G and HDCA-6G Biliary stentingalso changed urine BA-G levels (Figure 1(b)) CDCA-24G(41-fold) LCA-3G (35-fold) LCA-24G (65-fold) andDCA-24G (59-fold) were increased and CDCA-3G (43-fold) andHCA-24G (83-fold) were reduced

32 Biliary Stenting Differentially Affects the BA-G versusBA Metabolic Ratios in Serum and Urine from BiliaryStenosed Patients The serum and urine samples were pre-viously analyzed for their content in the 6 unconjugatedprecursors of the glucuronide derivatives analyzed here [20]These concentrations were used to calculate the metabolicratio (ie BA-GBA) for each glucuronide (Figure 2 andSupplementary Material 2) This parameter translates theglucuronide production capability from the available poolof unconjugated precursor thus ensuring that changes inblood or urine levels of BA-G species do not only reflectchanges in their precursors [11] In serum samples onlythe 65- 206- and 172-fold reductions in the respective

4 Canadian Journal of Gastroenterology and Hepatology

Serum UrineMetabolic ratios

Before stenting After stenting Before stenting After stenting

CDCA-3GCDCA 252 plusmn 43 69 plusmn 12 42 plusmn 08

CDCA-24GCDCA 01 plusmn 01 01 plusmn 01 02 plusmn 01 11 plusmn 03

CA-24GCA 102 plusmn 33 44 plusmn 12

LCA-3GLCA 14 plusmn 08 03 plusmn 01 NA NA

LCA-24GLCA 02 plusmn 01 01 plusmn 01 NA NA

DCA-3GDCA 64 plusmn 28 43 plusmn 28 21 plusmn 05

DCA-24GDCA 02 plusmn 01 01 plusmn 00 07 plusmn 06 17 plusmn 02

HDCA-6GHDCA 41 plusmn 40 49 plusmn 15 50 plusmn 25 414 plusmn 113

HDCA-24GHDCA 01 plusmn 01 01 plusmn 01 00 plusmn 00 00 plusmn 00

HCA-6GHCA 155 plusmn 56 198 plusmn 35 41591 plusmn 37459 2954 plusmn 2353

HCA-24GHCA 03 plusmn 01 03 plusmn 01 15 plusmn 07 01 plusmn 01

39 plusmn 28lowastlowastlowast

05 plusmn 02lowastlowast

04 plusmn 02lowastlowast

08 plusmn 03lowastlowast

(a)15

lowastlowast

10

5

0

minus5

minus35

minus30

minus25

minus20

minus15

minus10

CDCA

-24G

CDCA

-3G

CA-2

4G

LCA-

3G

LCA-

24G

DCA

-3G

DCA

-24G

HD

CA-6

G

HCA

-6G

HD

CA-2

4G

HCA

-24G

lowastlowastlowast

lowastlowast

lowastlowast

Urine

MR

BA-

GB

A(fo

ld ch

ange

s afte

r bili

ary

stent

ing)

Serum

(b)

Before stenting

p = 001

10

20

30

40

50

60

Seru

m C

A-24

GC

A ra

tio r2 = 052

4 6 8 10 12 142Urine CA-24GCA ratio

(c)

After stenting

01

02

03

04

05

06

07

Seru

m C

A-24

GC

A ra

tio

p = 002r2 = 045

2 3 41Urine CA-24GCA ratio

(d)

Response

minus50

minus40

minus30

minus20

minus10

0

10

20

30

Seru

m C

A-24

GC

A ra

tio

p = 002r2 = 044

minus8 minus4 0 4minus12Urine CA-24GCA ratio

(e)Figure 2 While bile flow restoration modulates metabolic ratios of bile acid glucuronides in a glucuronide- and fluid-dependent manner ((a)and (b)) the CA-24GCA ratio is unique in being significantly associated with serum and urine (cndashe) Serum and urine samples drawn beforeand after endoscopic biliary stenting from patients with stenosed bile ducts were analyzed for their concentrations in 11 bile acid-glucuronidespecies using LC-MSMS Concentrations of unconjugated acids were determined as reported [20] and themetabolic ratio for each species (a)was calculated as the ratio of glucuronide versus unconjugated precursor (b) Black (serum 119899 = 17) and white (urine 119899 = 12) bars representthe mean plusmn SEM of fold changes (posttreatment values divided by pretreatment values) in MRs Statistically significant differences betweenprestenting versus poststenting samples were determined using the Wilcoxon matched-pairs signed-rank test lowastlowast119901 lt 001 lowastlowastlowast119901 lt 0001Other changes failed to reach statistical significance The absence of unconjugated LCA in urine impaired such analysis for LCA-3 and -24G[20] while urine HDCA-24GHDCA was null (cndashe) Correlation analyses of paired (119899 = 12) prestenting (c) and poststenting (d) serum andurine CA-24GCA values or response to stenting determined as the difference between post- and pretreatment levels (e) were performedusing the Spearman rank-order correlation (1199032) and 119901 values for comparisons are indicated

Canadian Journal of Gastroenterology and Hepatology 5

Table 1 Summary of the correlation studies between serum and urine bile acid-glucuronide profiles

Urine levels Before stenting After stenting Response(1199032 119901 value) (1199032 119901 value) (1199032 119901 value)

CDCA-3G 010 030 002 067 025 010CDCA-24G 001 069 005 050 002 063CA-24G 032 005 051 001 007 042LCA-3G 000 097 019 015 001 081LCA-24G 014 022 047 001 042 002DCA-3G 082 lt00001 014 023 001 071DCA-24G 006 043 016 019 058 0004HDCA-6G 029 007 031 006 035 004HDCA-24G 000 100 005 047 003 059HCA-6G 029 007 015 020 067 0001HCA-24G 000 083 027 008 004 054Correlation analyses were performed using the Spearman rank-order correlation (1199032) and 119901 values for comparisons are indicated Response is calculated asthe difference between post- and prestenting values CA cholic acid CDCA chenodeoxycholic acid DCA deoxycholic acid HCA hyocholic acid HDCAhyodeoxycholic acid LCA lithocholic acid G glucuronide

CDCA-3GCDCA CA-24GCA and DCA-3GDCA ratioswere statistically significant (Figures 2(a) and 2(b)) Otherchanges even the reduction in DCA-24GDCA (117-fold)and LCA-3GLCA (53-fold) or the 25-fold increase of theHDCA-24GHDCA ratio failed to reach statistical signif-icance (Figure 2(b)) In urine (Supplementary Material 2)the unique significant change corresponded to the 52-foldreduction of theCA-24GCA ratio (Figure 2(b)) while the 14-and 15-fold reductions in HCA-6 and -24GMRs also failed toreach statistical significance

33 Serum and Urine BA-G Profiles Are Only Feebly Asso-ciated Correlation studies of circulating and urinary BA-Glevels in paired samples identified only few significant associ-ations (Table 1) In prestenting samples only the serumDCA-3G was significantly and positively correlated (1199032 = 082)to its urinary concentration while in poststenting fluids nocorrelation for this conjugate was detected Serum and urineCA-24G (1199032 = 051) and LCA-24G (1199032 = 047) levels weresignificantly correlated but only in samples obtained afterbile flow restoration (Table 1) When the response to stenting(determined as the difference between post- and prestentingvalues) was analyzed only changes for LCA-24G (1199032 = 042)DCA-24G (1199032 = 058) HDCA-6G (1199032 = 035) and HCA-6G (1199032 = 067) were positively and significantly associated(Table 1) Finally when similar analyses were performed forMRs the unique significant association in prestenting (1199032 =052) and poststenting (1199032 = 045) levels and response(1199032 = 044) was obtained with the ratio CA-24GCA (Figures2(c)ndash2(e))

34 Bile Acid Glucuronidation by Human Liver and KidneyExtracts The limited associations between circulating andurinary profiles observed above suggest a potential extra-hepatic origin for urinary levels of BA-G To test such ahypothesis we sought to compare the kinetic parameters ofBA glucuronidation by human liver (pool of 50 donors) and

kidney (pool of 8 donors) microsomes (Figures 3ndash5) Nodifferences in kinetic models were observed with the BA-specific glucuronidation reactions being distributed among 3models

(i) The sigmoidal pattern for CA-24G and CDCA-24 and-3G (Figure 3) DCA-3 and -24G (Figures 4(c) and 4(d)) andHDCA-6 and -24G (Figures 5(a) and 5(b)) production

(ii) The Michaelis-Menten model for the formation ofLCA-24G (Figure 4(b)) and HCA-6 and -24G (Figures 5(c)and 5(d))

(iii) The substrate-inhibition model for LCA-3G withrespective 119870119868 values of 2403 plusmn 535 and 2554 plusmn 699 120583M forliver and kidney extracts (Figure 4(a))

With the exception of CA-24G HDCA-24G and HCA-24G kidney UGTs generally displayed lower 119870119872 values Forexample kidney microsomes exhibited a 6-fold lower 119870119872value than liver ones for LCA-3G production (Figure 4(a))Another notable difference between liver and kidney micro-somes relates to the 10-fold higher 119881max value of HDCA-24Gproduction obtained with the hepatic microsomal prepara-tion (Figure 5(b)) Nevertheless the most efficient reactionswere obtained with the conversion of HDCA and HCA into6G derivatives that occurred at high velocities (ie 119881max)with both liver and kidney microsomes (Figures 5(a) and5(c)) In both tissue extracts the 6120572-hydroxylated acids weremore actively converted into 6-glucuronide when comparedto their 24-glucuronide counterparts (Figure 5) Interestinglythe opposite was observed for CDCA (Figures 3(a) and 3(b))and LCA (Figures 4(a) and 4(b)) The 119881max values for DCA-3and -24G formation were however similar (Figures 4(c) and4(d))

35 Differential BA-Conjugating UGT Expression in theHuman Liver and Kidney Results from kinetic experimentsindicate that the human kidney possesses an efficient BA-conjugating UGT system We next investigated whetherthe BA-conjugating UGTs 1A3 1A4 2B4 and 2B7 [8ndash11] are differentially expressed in human liver and kidney

6 Canadian Journal of Gastroenterology and Hepatology

02

04

06

08

10CD

CA-3

G fo

rmat

ion

(nm

olh

mg

prot

eins

)

100[CDCA] (M)

200 300 400

Liver nA = 17 plusmn 01

KMB = 821 plusmn 60

VGRC = 08 plusmn 01

Kidney nA = 19 plusmn 07KM

B = 443 plusmn 96VGR

C = 05 plusmn 01

(a)

10

20

30

CDCA

-24G

form

atio

n(n

mol

hm

g pr

otei

ns)

100[CDCA] (M)

200 300 400

Liver nA = 14 plusmn 01KM

B = 1682 plusmn 186VGR

C = 37 plusmn 02

Kidney nA = 13 plusmn 03KM

B = 897 plusmn 298VGR

C = 25 plusmn 04

(b)

05

10

15

20

CA-2

4G fo

rmat

ion

(nm

olh

mg

prot

eins

)

100[CA] (M)

200 300 400

Liver nA = 12 plusmn 01KM

B = 1141 plusmn 168VGR

C = 16 plusmn 01

Kidney nA = 10 plusmn 01KM

B = 1481 plusmn 397VGR

C = 11 plusmn 01

(c)

Figure 3 Kinetic analyses of CDCA-3G (a) CDCA-24G (b) and CA-24G (c) formation by microsomes from human liver and kidney Humanliver (black squares pool of 50 donors) and kidney (white diamonds pool of 8 donors) microsomes (10 120583g) were incubated in the presenceof increasing concentrations (1 to 350 120583M) of chenodeoxycholic acid (CDCA (a) and (b)) or cholic acid (CA (c)) for 2 hours at 37∘C Theformation of CDCA-3G (a) CDCA-24G (b) and CA-24G (c) was analyzed by LC-MSMS For each panel graphs represent the rate ofproduct formation (119910-axis) versus substrate concentration (119909-axis) of two experiments performed in triplicate The listed kinetic parameterswere determined as indicated in ldquoMaterials and Methodsrdquo A119899 Hill coefficient B119870119872 calculated Michaelis constant expressed as 120583M C119881maxcalculated maximal velocity expressed as nmolhmg proteins

Canadian Journal of Gastroenterology and Hepatology 7

200 300 400100[LCA] (M)

40

80

120

160

LCA-

3G fo

rmat

ion

(nm

olh

mg

prot

eins

)

LiverKM

B = 487 plusmn 90VGR

C = 171 plusmn 29

Kidney

KIA = 2403 plusmn 535

KMB = 82 plusmn 22

VGRC = 121 plusmn 11

KIA = 2554 plusmn 699

(a)

200 300 400100[LCA] (M)

60

120

180

240

300

LCA-

24G

form

atio

n (n

mol

hm

g pr

otei

ns)

LiverKM

B = 403 plusmn 67VGR

C = 321 plusmn 16

KidneyKM

B = 305 plusmn 56VGR

C = 162 plusmn 08

(b)

100 200 300 400[DCA] (M)

10

20

30

40

50

60

DCA

-3G

form

atio

n(n

mol

hm

g pr

otei

ns)

Liver nD 17 plusmn 03KM

B = 831 plusmn 113VGR

C = 31 plusmn 02

Kidney nD = 17 plusmn 03KM

B = 823 plusmn 136VGR

C = 36 plusmn 03

(c)

20

40

60

80

DCA

-24G

form

atio

n(n

mol

hm

g pr

otei

ns)

200 300 400100[DCA] (M)

Liver nD 12 plusmn 02KM

B = 711 plusmn 121VGR

C = 58 plusmn 05

Kidney nD = 17 plusmn 03KM

B = 582 plusmn 87VGR

C = 38 plusmn 03

(d)Figure 4Kinetic analyses of LCA-3G (a) LCA-24G (b) DCA-3G (c) and DCA-24G (d) formation by microsomes from human liver and kidneyHuman liver (black squares pool of 50 donors) and kidney (white diamonds pool of 8 donors) microsomes (10 120583g) were incubated in thepresence of increasing concentrations (1 to 350120583M) of lithocholic acid (LCA (a) and (b)) or deoxycholic acid (DCA (c) and (d)) for 2 hoursat 37∘C The formation of LCA-3G (a) LCA-24G (b) DCA-3G (c) and DCA-24G (d) was analyzed by LC-MSMS For each panel graphsrepresent the rate of product formation (119910-axis) versus substrate concentration (119909-axis) of two experiments performed in triplicateThe listedkinetic parameters were determined as indicated in ldquoMaterials and Methodsrdquo A119870119868 constant of inhibition expressed as 120583M B119870119872 calculatedMichaelis constant expressed as 120583M C119881max calculated maximal velocity expressed as nmolhmg proteins D119899 Hill coefficient

8 Canadian Journal of Gastroenterology and Hepatology

1750

3500

5250

7000

HD

CA-6

G fo

rmat

ion

(nm

olh

mg

prot

eins

)

200 300 400100[HDCA] (M)

Liver nA 29 plusmn 04KM

B = 551 plusmn 33VGR

C = 5287 plusmn 211

Kidney nA = 27 plusmn 05KM

B = 447 plusmn 38VGR

C = 3084 plusmn 156

(a)

100

200

300

400

500

HD

CA-2

4G fo

rmat

ion

(nm

olh

mg

prot

eins

)

200 300 400100[HDCA] (M)

Liver nA 38 plusmn 11KM

B = 808 plusmn 64VGR

C = 316 plusmn 22

Kidney nA = 23 plusmn 08KM

B = 939 plusmn 184VGR

C = 37 plusmn 05

(b)

800

1600

2400

3200

HCA

-6G

form

atio

n(n

mol

hm

g pr

otei

ns)

200 300 400100[HCA] (M)

LiverKM

B = 1202 plusmn 156VGR

C = 2285 plusmn 134

KidneyKM

B = 960 plusmn 192VGR

C = 2844 plusmn 241

(c)

20

40

60

HCA

-24G

form

atio

n(n

mol

hm

g pr

otei

ns)

200 300 400100[HCA] (M)

LiverKM

B = 617 plusmn 65VGR

C = 49 plusmn 02

KidneyKM

B = 978 plusmn 206VGR

C = 33 plusmn 03

(d)Figure 5 Kinetic analyses of HDCA-6G (a) HDCA-24G (b) HCA-6G (c) and HCA-24G (d) formation by microsomes from human liver andkidney Human liver (black squares pool of 50 donors) and kidney (white diamonds pool of 8 donors) microsomes (10 120583g) were incubatedin the presence of increasing concentrations (1 to 350120583M) of hyodeoxycholic acid (HDCA (a) and (b)) or hyocholic acid (HCA (c) and(d)) for 2 hours at 37∘CThe formation of HDCA-6G (a) HDCA-24G (b) HCA-6G (c) and HCA-24G (d) was analyzed by LC-MSMS Foreach panel graphs represent the rate of product formation (119910-axis) versus substrate concentration (119909-axis) of two experiments performedin triplicate The listed kinetic parameters were determined as indicated in ldquoMaterials and Methodsrdquo A119899 Hill coefficient B119870119872 calculatedMichaelis constant expressed as 120583M C119881max calculated maximal velocity expressed as nmolhmg proteins

Canadian Journal of Gastroenterology and Hepatology 9

Kid

ney

1A3

Kid

ney

Kid

ney

Kid

ney

Live

r

Live

r

Live

r

Live

r

1A4 2B4 2B7UGT

103

104

105

106

107U

GT

tran

scrip

ts (lo

g of

copy

num

ber

g RN

A)

(a)

1A3

2B42B7

2B7

1A

2B

LiverKidneyAnti-UGT kDa

55

55

55

55

55

40Anti-actin

(b)

Live

r

Kidn

ey

ND

5

10

15

20

UG

T1A

3 pr

otei

n le

vels

(pm

olm

g m

icro

som

es)

(c)

Figure 6 Differential expression of the bile acid-conjugating UGT enzymes in the human liver and kidney (a) UGTs 1A3 1A4 2B4 and 2B7mRNA levels in total RNA samples from human kidney (1 pool of 5 donors and 2 individual donors) and liver (8 individual donors) weredetermined through qRT-PCR analyses as described in ldquoMaterials andMethodsrdquo Results are expressed as log10 ofmRNA copy number per 120583gof total RNA Each symbol represents 1 donor with the exception of the white triangle that corresponds to a pool of the 5 kidney donors (b)The UGT protein contents in microsomal preparations (5120583g) from human kidney (pool of 4 female and 4 male donors) and liver (pool of 26male and 24 female donors) were determined after size separation on SDS-PAGE and hybridization with the anti-UGT1A3 anti-UGT2B47anti-UGT2B7 anti-UGT1A and anti-UGT2B antibodies (1 2000 dilution) The housekeeping protein 120573-actin was used to ensure the equalprotein loading in each lane Data presented are representative of two experiments (c) The UGT1A3 protein content in human liver andkidney microsomes was determined using LC-MSMS analyses as previously described [10] Data represent the mean plusmn SD of two differentexperiments performed in triplicate ND not detected

(Figure 6) With the exception of UGT2B4 transcripts thatwere detected only in 1 kidney mRNA sample other UGTmessengers were detected in all kidney and liver prepara-tions (Figure 6(a)) As expected [28 29] their mRNA levelssustained a strong interindividual variability Western blotanalyses evidenced however major differences between liverand kidney (Figure 6(b)) A clear UGT1A3 immunoreactivecomplex was obtained with liver extracts while this UGTprotein was not detected inmicrosomal proteins from kidney(Figure 6(b)) While the UGT2B7 protein was found atsimilar levels in both tissues the use of an anti-UGT2B42B7antibody indicated that the UGT2B4 protein may be moreabundant in liver than in kidney extracts (Figure 6(b))The lack of an accurate and specific anti-UGT1A4 antibody

impaired the quantification of this BA-conjugating UGThowever we were able to further confirm the absence ofthe UGT1A3 protein in kidney microsomes through the useof our previously established LC-MSMS-based proteomicmethod (Figure 6(c)) [10]

4 Discussion

Urinary and circulating levels of BA-Gs detected in thepresent study are similar to previous findings in terms ofboth glucuronide concentrations and species distribution [1314 16] However an important improvement of the presentinvestigations resides in our ability to discriminate betweenether and ester glucuronides of a single BA For example

10 Canadian Journal of Gastroenterology and Hepatology

while glucuronide conjugates of CDCA CA and DCA werepreviously identified as accumulating metabolites in serafrom patients undergoing bile drainage [20] the nature ofthese glucuronides has not been resolved until now Ourresults point out the ester CDCA-3G and DCA-3G andthe acyl CA-24G as the most abundant glucuronide speciesfound in patients with biliary obstruction Because acyl butnot ester glucuronides can be potentially toxic molecules(reviewed in [30]) such information may be critical inanticipating the consequences of their accumulation duringcholestasis

Interestingly serum levels of some of the bile acidglucuronides such as CDCA-3G CA-24G and LCA-3Gwere significantly reduced after biliary stenting while someothers like CDCA-24G LCA-24G and HDCA-6G weresignificantly increased While additional investigations arewarranted to decipher the mechanisms beyond such a glu-curonide species-dependent responsiveness one can spec-ulate that these changes may actually reflect the differen-tial manner in which the UGT enzymes andor the BA-Gtransporters may be altered upon biliary obstruction andafter stenting Therefore it would have been of interest tocompare how these proteins behave in tissues (ie liver andkidney) from cholestatic donors to validate such a hypothesisActually one limitation of the current study is that the liverand kidneymicrosomes used are not fromdonorswith biliaryobstruction When compared to other glucuronide conju-gates CA-24G presented a unique behavior (1) in contrastto other acyl BA-Gs that are only minor components of theurine and serum glucuronide pools CA-24G is the 3rd mostabundant glucuronidated acid in prestenting samples (2) thisspecies is the most spectacularly affected when poststentingversus prestenting sera profiles are compared (3) the serumCA-24GCA ratio sustains the stronger reduction after biliarystenting (4) this MR is also unique in being significantlyreduced in posttreatment urines and (5) only the CA-24GCA ratio exhibits positive serumurine association interms of prestenting and poststenting distribution as wellas treatment response These last observations support ahepatic origin for the formation of CA-24G and the strongCA-24G accumulation in prestenting fluids and its spec-tacular reduction after bile flow restoration suggests thatunder noncholestatic situations this compound formed inliver cells is normally secreted in bile This hypothesis isfurther supported by previous investigations in which theintravenous injections of cholate glucuronide to rats resultedin a rapid and efficient secretion in bile of the unchangedglucuronide conjugate [31] By contrast when the sameprocedure was applied to bile duct ligated animals 95 of theradioactivity was recovered in urine [31] The present studyindicates that similar rerouting of CA-24G elimination tourine may also occur in cholestatic patients since circulatinglevels detected in prestenting cholestatic donors were almost50-fold higher than those previously quantified in samplesfrom noncholestatic volunteers (sim2 nM [11]) Privilegingsecretion from liver cells into the blood for subsequenturinary elimination may actually protect the liver againstthe accumulation of such abundant and potentially toxicglucuronide species during cholestasis In the same vein the

improved formation of other conjugates such as CDCA-3Gand DCA-3G which we recently identified as nontoxic BAs[32] may also participate in the hepatic detoxification oftheir unconjugated CDCA and DCA precursors Consistentwith the above stated hypothesis circulating MRs for these2 conjugates (ie 252 and 64 for CDCA-3G and DCA-3Gresp) are spectacularly increased when compared to thosepreviously found in noncholestatic donors (06 and 01) [11]However the fact that circulating and urinary MRs for thesespecies were not associated and the fact that biliary stentingexerted only minor effects on these ratios in urine are alsoindicative of an extrahepatic origin for urine CDCA- andDCA-3G species at least after bile flow restoration

We next compared the human liver and kidney in termsof bile acid glucuronidation activity and BA-conjugatingUGT expression With 2 exceptions (ie the 6- and 10-fold difference in 119870119872 and 119881max values for LCA-3G andHDCA-24G formations resp) these experiments revealthe remarkable similarity of renal and hepatic bile acidglucuronidation processes and the clear preference of bothtissues in producing HDCA-6 and HCA-6G Such preferencemay actually explain the predominance of these conjugates invarious human fluids as previously described [11] Howeverthe most intriguing observation issuing from these analysesrelates to the UGT1A3 enzyme Indeed not only was thisenzyme detected in kidney extracts only at the mRNA(but not protein) level but also the absence of such anactive enzyme previously identified as the main isoform foracyl BA glucuronidation [8ndash10 12] is inconsistent with theelevated acyl glucuronide production capability of kidneymicrosomes A plausible explanation for the conflictingobservation on UGT1A3 mRNA and protein expression inkidney samples relates to the fact that protein and RNAextracts were from different sources and thus were notpaired It is therefore possible that only donors for mRNAexpress these isoforms in the kidney Meanwhile unlikelysuch a possibility is supported by the controversial nature ofthe UGT1A3 expression in the human kidney [29 33ndash35]Additional investigations are therefore warranted to clarifywhether this BA-conjugating protein is found or not in thehuman kidney

Nevertheless the present study clearly establishes themajor contributions that liver and kidney play in glu-curonidating bile acid during cholestasis Interestingly Zhouand colleagues reported that intestinal glucuronidation alsoplays an important role in controlling the local bile acidhomeostasis and the pathological development of colitis [36]These authors elegantly demonstrated that in the intestinebile acids glucuronidation leads to reduced intracellular levelsof ligands for the nuclear receptor FXR Reduced FXR activa-tion limits the secretion of FGF1519 in the portal circulationwhich in turns leads to an increase of de novo formationof bile acid in the liver [36] Thus in contrast to the liverand kidney where glucuronidation is thought to facilitate theremoval of bile acids the intestinal glucuronidation seems toincrease the delivery of bile acidsHowever it should be notedthat during obstructive cholestasis the delivery of hepaticbile acids to the intestine is compromised and thus onlyminor intestinal glucuronidation may occur

Canadian Journal of Gastroenterology and Hepatology 11

5 Conclusion

In conclusion the present study evidences the major impactthat bile flow restoration exerts on the circulating and urinarybile acid-glucuronide profiles and demonstrates the elevatedcapability of the human kidney to glucuronidate theseendogenous compounds Future investigations are requiredto clarify the mechanism allowing kidney extracts to convertbile acids into acyl glucuronides in the absence of theUGT1A3 protein

Abbreviations

BA Bile acidBO Biliary obstructionCA Cholic acidCDCA Chenodeoxycholic acidDCA Deoxycholic acidFXR Farnesoid X receptorG GlucuronideHDCA Hyodeoxycholic acidHCA Hyocholic acidLCA Lithocholic acidLC-MSMS Liquid chromatography-tandem mass

spectrometryUDPGA UDP-glucuronic acidUGT UDP-glucuronosyltransferaseMRPs Multidrug resistance related proteins

Conflicts of Interest

The authors declare that there are no conflicts of interest andthat there is no pertinent financial arrangement concerningthe work presented here

Acknowledgments

The authors wish to thank Dr Virginie Bocher for criticalreading of the manuscript This study was supported bygrants from theCanadian Institute ofHealth Research (CIHR119331) the Canadian Liver Foundation the Natural Sci-ences and Engineering Research Council of Canada (NSERC402213-2012) and the Canadian Foundation for Innovation(CFI 25712)Martin Perreault is holder of a scholarship fromthe ldquoFonds pour la Recherche en Sante du Quebecrdquo (FRSQ23444) Olivier Barbier is holder of a salary grant fromCIHR (New Investigator MSH95330) Piotr Milkiewiczwas supported by Grants DEC-201101BNZ504216 and201102ANZ500321 from the National Science Centre inPoland

Supplementary Materials

The supplementary materials present details of patient withbiliary obstruction (Supplementary Material 1) and thechanges in the urine profile of bile acid glucuronides and intheir metabolic ratios before and after a biliary stenting pro-cedure in patients with biliary obstruction (SupplementaryMaterial 2) Supplementary Material 1 patients with biliary

obstruction (adapted from [20]) Supplementary Material 2changes in the urine profile of bile acid glucuronides andin their metabolic ratios before and after a biliary stentingprocedure in patients with biliary obstruction (119899 = 12)(Supplementary Materials)

References

[1] D W Russell ldquoThe enzymes regulation and genetics of bileacid synthesisrdquo Annual Review of Biochemistry vol 72 pp 137ndash174 2003

[2] M J Monte J J G Marin A Antelo and J Vazquez-TatoldquoBile acids Chemistry physiology and pathophysiologyrdquoWorldJournal of Gastroenterology vol 15 no 7 pp 804ndash816 2009

[3] A F Hofmann ldquoDetoxification of lithocholic acid a toxicbile-acid relevance to drug hepatotoxicityrdquo Drug MetabolismReviews vol 36 no 3-4 pp 703ndash722 2004

[4] C Pauli-Magnus B Stieger Y Meier G A Kullak-Ublick andP J Meier ldquoEnterohepatic transport of bile salts and genetics ofcholestasisrdquo Journal of Hepatology vol 43 no 2 pp 342ndash3572005

[5] G Paumgartner ldquoPharmacotherapy of cholestatic liver dis-easesrdquo Journal of Digestive Diseases vol 11 no 3 pp 119ndash1252010

[6] M Verreault J Kaeding P Caron et al ldquoRegulation ofendobiotics glucuronidation by ligand-activated transcriptionfactors Physiological function and therapeutic potentialrdquoDrugMetabolism Reviews vol 42 no 1 pp 106ndash118 2010

[7] P I Mackenzie K W Bock B Burchell et al ldquoNomenclatureupdate for the mammalian UDP glycosyltransferase (UGT)gene superfamilyrdquo Pharmacogenetics and Genomics vol 15 no10 pp 677ndash685 2005

[8] M H Court S Hazarika S Krishnaswamy M Finel andJ A Williams ldquoNovel polymorphic human UDP-glucurono-syltransferase 2A3 Cloning functional characterization ofenzyme variants comparative tissue expression and geneinductionrdquoMolecular Pharmacology vol 74 no 3 pp 744ndash7542008

[9] W E Gall G Zawada B Mojarrabi et al ldquoDifferential glu-curonidation of bile acids androgens and estrogens by humanUGT1A3 and 2B7rdquo The Journal of Steroid Biochemistry andMolecular Biology vol 70 no 1ndash3 pp 101ndash108 1999

[10] J Trottier D El Husseini M Perreault et al ldquoThe humanUGT1A3 enzyme conjugates norursodeoxycholic acid into aC23-ester glucuronide in the liverrdquo The Journal of BiologicalChemistry vol 285 no 2 pp 1113ndash1121 2010

[11] J Trottier M Perreault I Rudkowska et al ldquoProfiling serumbile acid glucuronides in humans Gender divergences geneticdeterminants and response to fenofibraterdquo Clinical Pharmacol-ogy ampTherapeutics vol 94 no 4 pp 533ndash543 2013

[12] P Caron J Trottier M Verreault J Belanger J Kaeding andO Barbier ldquoEnzymatic production of bile acid glucuronidesused as analytical standards for liquid chromatography-massspectrometry analysesrdquo Molecular Pharmaceutics vol 3 no 3pp 293ndash302 2006

[13] B Alme and J Sjovall ldquoAnalysis of bile acid glucuronidesin urine identification of 3120572612057212120572-trihydroxy-5120573-cholanoicacidrdquoThe Journal of Steroid Biochemistry andMolecular Biologyvol 13 no 8 pp 907ndash916 1980

[14] A Stiehl R Raedsch G Rudolph P Czygan and S WalkerldquoAnalysis of bile acid glucuronides in urine group separation

12 Canadian Journal of Gastroenterology and Hepatology

on a lipophilic anion exchangerrdquo Clinica Chimica Acta vol 123no 3 pp 275ndash285 1982

[15] A Stiehl R Raedsch G Rudolph U Gundert-Remy and MSenn ldquoBiliary and urinary excretion of sulfated glucuronidatedand tetrahydroxylated bile acids in cirrhotic patientsrdquo Hepatol-ogy vol 5 no 3 pp 492ndash495 1985

[16] H Takikawa T Beppu and Y Seyama ldquoUrinary concentrationsof bile acid glucuronides and sulfates in hepatobiliary diseasesrdquoGastroenterologia Japonica vol 19 no 2 pp 104ndash109 1984

[17] H Wietholtz H Marschall R Reuschenbach H Matern andS Matern ldquoUrinary excretion of bile acid glucosides and glu-curonides in extrahepatic cholestasisrdquo Hepatology vol 13 no4 pp 656ndash662 1991

[18] K M Knights A Rowland and J O Miners ldquoRenal drugmetabolism in humans The potential for drug-endobioticinteractions involving cytochrome P450 (CYP) and UDP-glu-curonosyltransferase (UGT)rdquo British Journal of Clinical Phar-macology vol 76 no 4 pp 587ndash602 2013

[19] S Matern H Matern E H Farthmann and W GerokldquoHepatic and extrahepatic glucuronidation of bile acids in manCharacterization of bile acid uridine 5rsquo-diphosphate-glucu-ronosyltransferase in hepatic renal and intestinalmicrosomesrdquoThe Journal of Clinical Investigation vol 74 no 2 pp 402ndash4101984

[20] J Trottier A Białek P Caron R J Straka P Milkiewicz and OBarbier ldquoProfiling circulating and urinary bile acids in patientswith biliary obstruction before and after biliary stentingrdquo PLoSONE vol 6 no 7 Article ID e22094 2011

[21] M Perreault L Gauthier-Landry J Trottier et al ldquoThe humanUDP-glucuronosyltransferase UGT2A1 and UGT2A2 enzymesare highly active in bile acid glucuronidationrdquoDrugMetabolismand Disposition vol 41 no 9 pp 1616ndash1620 2013

[22] M E Campbell D M Grant T Inaba and W KalowldquoBiotransformation of caffeine paraxanthine theophylline andtheobromine by polycyclic aromatic hydrocarbon-induciblecytochrome(s) P-450 in human liver microsomesrdquo DrugMetabolism and Disposition vol 15 no 2 pp 237ndash249 1987

[23] J Lepine O Bernard M Plante et al ldquoSpecificity andregioselectivity of the conjugation of estradiol estrone andtheir catecholestrogen and methoxyestrogen metabolites byhuman uridine diphospho-glucuronosyltransferases expressedin endometriumrdquo The Journal of Clinical Endocrinology ampMetabolism vol 89 no 10 pp 5222ndash5232 2004

[24] J Thibaudeau J Lepine J Tojcic et al ldquoCharacterizationof common UGT1A8 UGT1A9 and UGT2B7 variants withdifferent capacities to inactivate mutagenic 4-hydroxylatedmetabolites of estradiol and estronerdquo Cancer Research vol 66no 1 pp 125ndash133 2006

[25] J O Miners P I MacKenzie and K M Knights ldquoTheprediction of drug-glucuronidation parameters in humansUDP-glucuronosyltransferase enzyme-selective substrate andinhibitor probes for reaction phenotyping and in vitroin vivoextrapolation of drug clearance and drug-drug interactionpotentialrdquo Drug Metabolism Reviews vol 42 no 1 pp 189ndash2012010

[26] M Verreault K Senekeo-Effenberger J Trottier et al ldquoTheliver X-receptor alpha controls hepatic expression of the humanbile acid-glucuronidating UGT1A3 enzyme in human cells andtransgenic micerdquo Hepatology vol 44 no 2 pp 368ndash378 2006

[27] R G Rutledge ldquoMathematics of quantitative kinetic PCR andthe application of standard curvesrdquo Nucleic Acids Research vol31 no 16 pp 93endash93 2003

[28] T Izukawa M Nakajima R Fujiwara et al ldquoQuantitative anal-ysis of UDP-glucuronosyltransferase (UGT) 1A and UGT2Bexpression levels in human liversrdquo Drug Metabolism and Dis-position vol 37 no 8 pp 1759ndash1768 2009

[29] S Ohno and S Nakajin ldquoDetermination of mRNA expressionof human UDP-glucuronosyltransferases and application forlocalization in various human tissues by real-time reversetranscriptase-polymerase chain reactionrdquoDrugMetabolism andDisposition vol 37 no 1 pp 32ndash40 2009

[30] S L Regan J L Maggs T G Hammond C Lambert D PWilliams and B K Park ldquoAcyl glucuronidesThe good the badand the uglyrdquo Biopharmaceutics amp Drug Disposition vol 31 no7 pp 367ndash395 2010

[31] J M Little M V Chari and R Lester ldquoExcretion of cholateglucuroniderdquo Journal of Lipid Research vol 26 no 5 pp 583ndash592 1985

[32] M Perreault A Białek J Trottier et al ldquoRole of glucuronidationfor hepatic detoxification and urinary elimination of toxic bileacids during biliary obstructionrdquo PLoS ONE vol 8 no 11Article ID e80994 2013

[33] H Girard E Levesque J Bellemare K Journault B Caillierand C Guillemette ldquoGenetic diversity at the UGT1 locus isamplified by a novel 31015840 alternative splicing mechanism leadingto nine additional UGT1A proteins that act as regulators ofglucuronidation activityrdquo Pharmacogenetics and Genomics vol17 no 12 pp 1077ndash1089 2007

[34] A Nakamura M Nakajima H Yamanaka R Fujiwara and TYokoi ldquoExpression of UGT1A and UGT2B mRNA in humannormal tissues and various cell linesrdquo Drug Metabolism andDisposition vol 36 no 8 pp 1461ndash1464 2008

[35] D E Harbourt J K Fallon S Ito et al ldquoQuantificationof human uridine-diphosphate glucuronosyl transferase 1Aisoforms in liver intestine and kidney using nanobore liq-uid chromatography-tandem mass spectrometryrdquo AnalyticalChemistry vol 84 no 1 pp 98ndash105 2012

[36] X Zhou L Cao C Jiang et al ldquoPPAR120572-UGT axis activationrepresses intestinal FXR-FGF15 feedback signalling and exacer-bates experimental colitisrdquoNature Communications vol 5 2014

Stem Cells International

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

MEDIATORSINFLAMMATION

of

EndocrinologyInternational Journal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Disease Markers

Hindawiwwwhindawicom Volume 2018

BioMed Research International

OncologyJournal of

Hindawiwwwhindawicom Volume 2013

Hindawiwwwhindawicom Volume 2018

Oxidative Medicine and Cellular Longevity

Hindawiwwwhindawicom Volume 2018

PPAR Research

Hindawi Publishing Corporation httpwwwhindawicom Volume 2013Hindawiwwwhindawicom

The Scientific World Journal

Volume 2018

Immunology ResearchHindawiwwwhindawicom Volume 2018

Journal of

ObesityJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Computational and Mathematical Methods in Medicine

Hindawiwwwhindawicom Volume 2018

Behavioural Neurology

OphthalmologyJournal of

Hindawiwwwhindawicom Volume 2018

Diabetes ResearchJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Research and TreatmentAIDS

Hindawiwwwhindawicom Volume 2018

Gastroenterology Research and Practice

Hindawiwwwhindawicom Volume 2018

Parkinsonrsquos Disease

Evidence-Based Complementary andAlternative Medicine

Volume 2018Hindawiwwwhindawicom

Submit your manuscripts atwwwhindawicom

Page 4: Urinary Elimination of Bile Acid Glucuronides under Severe ...

4 Canadian Journal of Gastroenterology and Hepatology

Serum UrineMetabolic ratios

Before stenting After stenting Before stenting After stenting

CDCA-3GCDCA 252 plusmn 43 69 plusmn 12 42 plusmn 08

CDCA-24GCDCA 01 plusmn 01 01 plusmn 01 02 plusmn 01 11 plusmn 03

CA-24GCA 102 plusmn 33 44 plusmn 12

LCA-3GLCA 14 plusmn 08 03 plusmn 01 NA NA

LCA-24GLCA 02 plusmn 01 01 plusmn 01 NA NA

DCA-3GDCA 64 plusmn 28 43 plusmn 28 21 plusmn 05

DCA-24GDCA 02 plusmn 01 01 plusmn 00 07 plusmn 06 17 plusmn 02

HDCA-6GHDCA 41 plusmn 40 49 plusmn 15 50 plusmn 25 414 plusmn 113

HDCA-24GHDCA 01 plusmn 01 01 plusmn 01 00 plusmn 00 00 plusmn 00

HCA-6GHCA 155 plusmn 56 198 plusmn 35 41591 plusmn 37459 2954 plusmn 2353

HCA-24GHCA 03 plusmn 01 03 plusmn 01 15 plusmn 07 01 plusmn 01

39 plusmn 28lowastlowastlowast

05 plusmn 02lowastlowast

04 plusmn 02lowastlowast

08 plusmn 03lowastlowast

(a)15

lowastlowast

10

5

0

minus5

minus35

minus30

minus25

minus20

minus15

minus10

CDCA

-24G

CDCA

-3G

CA-2

4G

LCA-

3G

LCA-

24G

DCA

-3G

DCA

-24G

HD

CA-6

G

HCA

-6G

HD

CA-2

4G

HCA

-24G

lowastlowastlowast

lowastlowast

lowastlowast

Urine

MR

BA-

GB

A(fo

ld ch

ange

s afte

r bili

ary

stent

ing)

Serum

(b)

Before stenting

p = 001

10

20

30

40

50

60

Seru

m C

A-24

GC

A ra

tio r2 = 052

4 6 8 10 12 142Urine CA-24GCA ratio

(c)

After stenting

01

02

03

04

05

06

07

Seru

m C

A-24

GC

A ra

tio

p = 002r2 = 045

2 3 41Urine CA-24GCA ratio

(d)

Response

minus50

minus40

minus30

minus20

minus10

0

10

20

30

Seru

m C

A-24

GC

A ra

tio

p = 002r2 = 044

minus8 minus4 0 4minus12Urine CA-24GCA ratio

(e)Figure 2 While bile flow restoration modulates metabolic ratios of bile acid glucuronides in a glucuronide- and fluid-dependent manner ((a)and (b)) the CA-24GCA ratio is unique in being significantly associated with serum and urine (cndashe) Serum and urine samples drawn beforeand after endoscopic biliary stenting from patients with stenosed bile ducts were analyzed for their concentrations in 11 bile acid-glucuronidespecies using LC-MSMS Concentrations of unconjugated acids were determined as reported [20] and themetabolic ratio for each species (a)was calculated as the ratio of glucuronide versus unconjugated precursor (b) Black (serum 119899 = 17) and white (urine 119899 = 12) bars representthe mean plusmn SEM of fold changes (posttreatment values divided by pretreatment values) in MRs Statistically significant differences betweenprestenting versus poststenting samples were determined using the Wilcoxon matched-pairs signed-rank test lowastlowast119901 lt 001 lowastlowastlowast119901 lt 0001Other changes failed to reach statistical significance The absence of unconjugated LCA in urine impaired such analysis for LCA-3 and -24G[20] while urine HDCA-24GHDCA was null (cndashe) Correlation analyses of paired (119899 = 12) prestenting (c) and poststenting (d) serum andurine CA-24GCA values or response to stenting determined as the difference between post- and pretreatment levels (e) were performedusing the Spearman rank-order correlation (1199032) and 119901 values for comparisons are indicated

Canadian Journal of Gastroenterology and Hepatology 5

Table 1 Summary of the correlation studies between serum and urine bile acid-glucuronide profiles

Urine levels Before stenting After stenting Response(1199032 119901 value) (1199032 119901 value) (1199032 119901 value)

CDCA-3G 010 030 002 067 025 010CDCA-24G 001 069 005 050 002 063CA-24G 032 005 051 001 007 042LCA-3G 000 097 019 015 001 081LCA-24G 014 022 047 001 042 002DCA-3G 082 lt00001 014 023 001 071DCA-24G 006 043 016 019 058 0004HDCA-6G 029 007 031 006 035 004HDCA-24G 000 100 005 047 003 059HCA-6G 029 007 015 020 067 0001HCA-24G 000 083 027 008 004 054Correlation analyses were performed using the Spearman rank-order correlation (1199032) and 119901 values for comparisons are indicated Response is calculated asthe difference between post- and prestenting values CA cholic acid CDCA chenodeoxycholic acid DCA deoxycholic acid HCA hyocholic acid HDCAhyodeoxycholic acid LCA lithocholic acid G glucuronide

CDCA-3GCDCA CA-24GCA and DCA-3GDCA ratioswere statistically significant (Figures 2(a) and 2(b)) Otherchanges even the reduction in DCA-24GDCA (117-fold)and LCA-3GLCA (53-fold) or the 25-fold increase of theHDCA-24GHDCA ratio failed to reach statistical signif-icance (Figure 2(b)) In urine (Supplementary Material 2)the unique significant change corresponded to the 52-foldreduction of theCA-24GCA ratio (Figure 2(b)) while the 14-and 15-fold reductions in HCA-6 and -24GMRs also failed toreach statistical significance

33 Serum and Urine BA-G Profiles Are Only Feebly Asso-ciated Correlation studies of circulating and urinary BA-Glevels in paired samples identified only few significant associ-ations (Table 1) In prestenting samples only the serumDCA-3G was significantly and positively correlated (1199032 = 082)to its urinary concentration while in poststenting fluids nocorrelation for this conjugate was detected Serum and urineCA-24G (1199032 = 051) and LCA-24G (1199032 = 047) levels weresignificantly correlated but only in samples obtained afterbile flow restoration (Table 1) When the response to stenting(determined as the difference between post- and prestentingvalues) was analyzed only changes for LCA-24G (1199032 = 042)DCA-24G (1199032 = 058) HDCA-6G (1199032 = 035) and HCA-6G (1199032 = 067) were positively and significantly associated(Table 1) Finally when similar analyses were performed forMRs the unique significant association in prestenting (1199032 =052) and poststenting (1199032 = 045) levels and response(1199032 = 044) was obtained with the ratio CA-24GCA (Figures2(c)ndash2(e))

34 Bile Acid Glucuronidation by Human Liver and KidneyExtracts The limited associations between circulating andurinary profiles observed above suggest a potential extra-hepatic origin for urinary levels of BA-G To test such ahypothesis we sought to compare the kinetic parameters ofBA glucuronidation by human liver (pool of 50 donors) and

kidney (pool of 8 donors) microsomes (Figures 3ndash5) Nodifferences in kinetic models were observed with the BA-specific glucuronidation reactions being distributed among 3models

(i) The sigmoidal pattern for CA-24G and CDCA-24 and-3G (Figure 3) DCA-3 and -24G (Figures 4(c) and 4(d)) andHDCA-6 and -24G (Figures 5(a) and 5(b)) production

(ii) The Michaelis-Menten model for the formation ofLCA-24G (Figure 4(b)) and HCA-6 and -24G (Figures 5(c)and 5(d))

(iii) The substrate-inhibition model for LCA-3G withrespective 119870119868 values of 2403 plusmn 535 and 2554 plusmn 699 120583M forliver and kidney extracts (Figure 4(a))

With the exception of CA-24G HDCA-24G and HCA-24G kidney UGTs generally displayed lower 119870119872 values Forexample kidney microsomes exhibited a 6-fold lower 119870119872value than liver ones for LCA-3G production (Figure 4(a))Another notable difference between liver and kidney micro-somes relates to the 10-fold higher 119881max value of HDCA-24Gproduction obtained with the hepatic microsomal prepara-tion (Figure 5(b)) Nevertheless the most efficient reactionswere obtained with the conversion of HDCA and HCA into6G derivatives that occurred at high velocities (ie 119881max)with both liver and kidney microsomes (Figures 5(a) and5(c)) In both tissue extracts the 6120572-hydroxylated acids weremore actively converted into 6-glucuronide when comparedto their 24-glucuronide counterparts (Figure 5) Interestinglythe opposite was observed for CDCA (Figures 3(a) and 3(b))and LCA (Figures 4(a) and 4(b)) The 119881max values for DCA-3and -24G formation were however similar (Figures 4(c) and4(d))

35 Differential BA-Conjugating UGT Expression in theHuman Liver and Kidney Results from kinetic experimentsindicate that the human kidney possesses an efficient BA-conjugating UGT system We next investigated whetherthe BA-conjugating UGTs 1A3 1A4 2B4 and 2B7 [8ndash11] are differentially expressed in human liver and kidney

6 Canadian Journal of Gastroenterology and Hepatology

02

04

06

08

10CD

CA-3

G fo

rmat

ion

(nm

olh

mg

prot

eins

)

100[CDCA] (M)

200 300 400

Liver nA = 17 plusmn 01

KMB = 821 plusmn 60

VGRC = 08 plusmn 01

Kidney nA = 19 plusmn 07KM

B = 443 plusmn 96VGR

C = 05 plusmn 01

(a)

10

20

30

CDCA

-24G

form

atio

n(n

mol

hm

g pr

otei

ns)

100[CDCA] (M)

200 300 400

Liver nA = 14 plusmn 01KM

B = 1682 plusmn 186VGR

C = 37 plusmn 02

Kidney nA = 13 plusmn 03KM

B = 897 plusmn 298VGR

C = 25 plusmn 04

(b)

05

10

15

20

CA-2

4G fo

rmat

ion

(nm

olh

mg

prot

eins

)

100[CA] (M)

200 300 400

Liver nA = 12 plusmn 01KM

B = 1141 plusmn 168VGR

C = 16 plusmn 01

Kidney nA = 10 plusmn 01KM

B = 1481 plusmn 397VGR

C = 11 plusmn 01

(c)

Figure 3 Kinetic analyses of CDCA-3G (a) CDCA-24G (b) and CA-24G (c) formation by microsomes from human liver and kidney Humanliver (black squares pool of 50 donors) and kidney (white diamonds pool of 8 donors) microsomes (10 120583g) were incubated in the presenceof increasing concentrations (1 to 350 120583M) of chenodeoxycholic acid (CDCA (a) and (b)) or cholic acid (CA (c)) for 2 hours at 37∘C Theformation of CDCA-3G (a) CDCA-24G (b) and CA-24G (c) was analyzed by LC-MSMS For each panel graphs represent the rate ofproduct formation (119910-axis) versus substrate concentration (119909-axis) of two experiments performed in triplicate The listed kinetic parameterswere determined as indicated in ldquoMaterials and Methodsrdquo A119899 Hill coefficient B119870119872 calculated Michaelis constant expressed as 120583M C119881maxcalculated maximal velocity expressed as nmolhmg proteins

Canadian Journal of Gastroenterology and Hepatology 7

200 300 400100[LCA] (M)

40

80

120

160

LCA-

3G fo

rmat

ion

(nm

olh

mg

prot

eins

)

LiverKM

B = 487 plusmn 90VGR

C = 171 plusmn 29

Kidney

KIA = 2403 plusmn 535

KMB = 82 plusmn 22

VGRC = 121 plusmn 11

KIA = 2554 plusmn 699

(a)

200 300 400100[LCA] (M)

60

120

180

240

300

LCA-

24G

form

atio

n (n

mol

hm

g pr

otei

ns)

LiverKM

B = 403 plusmn 67VGR

C = 321 plusmn 16

KidneyKM

B = 305 plusmn 56VGR

C = 162 plusmn 08

(b)

100 200 300 400[DCA] (M)

10

20

30

40

50

60

DCA

-3G

form

atio

n(n

mol

hm

g pr

otei

ns)

Liver nD 17 plusmn 03KM

B = 831 plusmn 113VGR

C = 31 plusmn 02

Kidney nD = 17 plusmn 03KM

B = 823 plusmn 136VGR

C = 36 plusmn 03

(c)

20

40

60

80

DCA

-24G

form

atio

n(n

mol

hm

g pr

otei

ns)

200 300 400100[DCA] (M)

Liver nD 12 plusmn 02KM

B = 711 plusmn 121VGR

C = 58 plusmn 05

Kidney nD = 17 plusmn 03KM

B = 582 plusmn 87VGR

C = 38 plusmn 03

(d)Figure 4Kinetic analyses of LCA-3G (a) LCA-24G (b) DCA-3G (c) and DCA-24G (d) formation by microsomes from human liver and kidneyHuman liver (black squares pool of 50 donors) and kidney (white diamonds pool of 8 donors) microsomes (10 120583g) were incubated in thepresence of increasing concentrations (1 to 350120583M) of lithocholic acid (LCA (a) and (b)) or deoxycholic acid (DCA (c) and (d)) for 2 hoursat 37∘C The formation of LCA-3G (a) LCA-24G (b) DCA-3G (c) and DCA-24G (d) was analyzed by LC-MSMS For each panel graphsrepresent the rate of product formation (119910-axis) versus substrate concentration (119909-axis) of two experiments performed in triplicateThe listedkinetic parameters were determined as indicated in ldquoMaterials and Methodsrdquo A119870119868 constant of inhibition expressed as 120583M B119870119872 calculatedMichaelis constant expressed as 120583M C119881max calculated maximal velocity expressed as nmolhmg proteins D119899 Hill coefficient

8 Canadian Journal of Gastroenterology and Hepatology

1750

3500

5250

7000

HD

CA-6

G fo

rmat

ion

(nm

olh

mg

prot

eins

)

200 300 400100[HDCA] (M)

Liver nA 29 plusmn 04KM

B = 551 plusmn 33VGR

C = 5287 plusmn 211

Kidney nA = 27 plusmn 05KM

B = 447 plusmn 38VGR

C = 3084 plusmn 156

(a)

100

200

300

400

500

HD

CA-2

4G fo

rmat

ion

(nm

olh

mg

prot

eins

)

200 300 400100[HDCA] (M)

Liver nA 38 plusmn 11KM

B = 808 plusmn 64VGR

C = 316 plusmn 22

Kidney nA = 23 plusmn 08KM

B = 939 plusmn 184VGR

C = 37 plusmn 05

(b)

800

1600

2400

3200

HCA

-6G

form

atio

n(n

mol

hm

g pr

otei

ns)

200 300 400100[HCA] (M)

LiverKM

B = 1202 plusmn 156VGR

C = 2285 plusmn 134

KidneyKM

B = 960 plusmn 192VGR

C = 2844 plusmn 241

(c)

20

40

60

HCA

-24G

form

atio

n(n

mol

hm

g pr

otei

ns)

200 300 400100[HCA] (M)

LiverKM

B = 617 plusmn 65VGR

C = 49 plusmn 02

KidneyKM

B = 978 plusmn 206VGR

C = 33 plusmn 03

(d)Figure 5 Kinetic analyses of HDCA-6G (a) HDCA-24G (b) HCA-6G (c) and HCA-24G (d) formation by microsomes from human liver andkidney Human liver (black squares pool of 50 donors) and kidney (white diamonds pool of 8 donors) microsomes (10 120583g) were incubatedin the presence of increasing concentrations (1 to 350120583M) of hyodeoxycholic acid (HDCA (a) and (b)) or hyocholic acid (HCA (c) and(d)) for 2 hours at 37∘CThe formation of HDCA-6G (a) HDCA-24G (b) HCA-6G (c) and HCA-24G (d) was analyzed by LC-MSMS Foreach panel graphs represent the rate of product formation (119910-axis) versus substrate concentration (119909-axis) of two experiments performedin triplicate The listed kinetic parameters were determined as indicated in ldquoMaterials and Methodsrdquo A119899 Hill coefficient B119870119872 calculatedMichaelis constant expressed as 120583M C119881max calculated maximal velocity expressed as nmolhmg proteins

Canadian Journal of Gastroenterology and Hepatology 9

Kid

ney

1A3

Kid

ney

Kid

ney

Kid

ney

Live

r

Live

r

Live

r

Live

r

1A4 2B4 2B7UGT

103

104

105

106

107U

GT

tran

scrip

ts (lo

g of

copy

num

ber

g RN

A)

(a)

1A3

2B42B7

2B7

1A

2B

LiverKidneyAnti-UGT kDa

55

55

55

55

55

40Anti-actin

(b)

Live

r

Kidn

ey

ND

5

10

15

20

UG

T1A

3 pr

otei

n le

vels

(pm

olm

g m

icro

som

es)

(c)

Figure 6 Differential expression of the bile acid-conjugating UGT enzymes in the human liver and kidney (a) UGTs 1A3 1A4 2B4 and 2B7mRNA levels in total RNA samples from human kidney (1 pool of 5 donors and 2 individual donors) and liver (8 individual donors) weredetermined through qRT-PCR analyses as described in ldquoMaterials andMethodsrdquo Results are expressed as log10 ofmRNA copy number per 120583gof total RNA Each symbol represents 1 donor with the exception of the white triangle that corresponds to a pool of the 5 kidney donors (b)The UGT protein contents in microsomal preparations (5120583g) from human kidney (pool of 4 female and 4 male donors) and liver (pool of 26male and 24 female donors) were determined after size separation on SDS-PAGE and hybridization with the anti-UGT1A3 anti-UGT2B47anti-UGT2B7 anti-UGT1A and anti-UGT2B antibodies (1 2000 dilution) The housekeeping protein 120573-actin was used to ensure the equalprotein loading in each lane Data presented are representative of two experiments (c) The UGT1A3 protein content in human liver andkidney microsomes was determined using LC-MSMS analyses as previously described [10] Data represent the mean plusmn SD of two differentexperiments performed in triplicate ND not detected

(Figure 6) With the exception of UGT2B4 transcripts thatwere detected only in 1 kidney mRNA sample other UGTmessengers were detected in all kidney and liver prepara-tions (Figure 6(a)) As expected [28 29] their mRNA levelssustained a strong interindividual variability Western blotanalyses evidenced however major differences between liverand kidney (Figure 6(b)) A clear UGT1A3 immunoreactivecomplex was obtained with liver extracts while this UGTprotein was not detected inmicrosomal proteins from kidney(Figure 6(b)) While the UGT2B7 protein was found atsimilar levels in both tissues the use of an anti-UGT2B42B7antibody indicated that the UGT2B4 protein may be moreabundant in liver than in kidney extracts (Figure 6(b))The lack of an accurate and specific anti-UGT1A4 antibody

impaired the quantification of this BA-conjugating UGThowever we were able to further confirm the absence ofthe UGT1A3 protein in kidney microsomes through the useof our previously established LC-MSMS-based proteomicmethod (Figure 6(c)) [10]

4 Discussion

Urinary and circulating levels of BA-Gs detected in thepresent study are similar to previous findings in terms ofboth glucuronide concentrations and species distribution [1314 16] However an important improvement of the presentinvestigations resides in our ability to discriminate betweenether and ester glucuronides of a single BA For example

10 Canadian Journal of Gastroenterology and Hepatology

while glucuronide conjugates of CDCA CA and DCA werepreviously identified as accumulating metabolites in serafrom patients undergoing bile drainage [20] the nature ofthese glucuronides has not been resolved until now Ourresults point out the ester CDCA-3G and DCA-3G andthe acyl CA-24G as the most abundant glucuronide speciesfound in patients with biliary obstruction Because acyl butnot ester glucuronides can be potentially toxic molecules(reviewed in [30]) such information may be critical inanticipating the consequences of their accumulation duringcholestasis

Interestingly serum levels of some of the bile acidglucuronides such as CDCA-3G CA-24G and LCA-3Gwere significantly reduced after biliary stenting while someothers like CDCA-24G LCA-24G and HDCA-6G weresignificantly increased While additional investigations arewarranted to decipher the mechanisms beyond such a glu-curonide species-dependent responsiveness one can spec-ulate that these changes may actually reflect the differen-tial manner in which the UGT enzymes andor the BA-Gtransporters may be altered upon biliary obstruction andafter stenting Therefore it would have been of interest tocompare how these proteins behave in tissues (ie liver andkidney) from cholestatic donors to validate such a hypothesisActually one limitation of the current study is that the liverand kidneymicrosomes used are not fromdonorswith biliaryobstruction When compared to other glucuronide conju-gates CA-24G presented a unique behavior (1) in contrastto other acyl BA-Gs that are only minor components of theurine and serum glucuronide pools CA-24G is the 3rd mostabundant glucuronidated acid in prestenting samples (2) thisspecies is the most spectacularly affected when poststentingversus prestenting sera profiles are compared (3) the serumCA-24GCA ratio sustains the stronger reduction after biliarystenting (4) this MR is also unique in being significantlyreduced in posttreatment urines and (5) only the CA-24GCA ratio exhibits positive serumurine association interms of prestenting and poststenting distribution as wellas treatment response These last observations support ahepatic origin for the formation of CA-24G and the strongCA-24G accumulation in prestenting fluids and its spec-tacular reduction after bile flow restoration suggests thatunder noncholestatic situations this compound formed inliver cells is normally secreted in bile This hypothesis isfurther supported by previous investigations in which theintravenous injections of cholate glucuronide to rats resultedin a rapid and efficient secretion in bile of the unchangedglucuronide conjugate [31] By contrast when the sameprocedure was applied to bile duct ligated animals 95 of theradioactivity was recovered in urine [31] The present studyindicates that similar rerouting of CA-24G elimination tourine may also occur in cholestatic patients since circulatinglevels detected in prestenting cholestatic donors were almost50-fold higher than those previously quantified in samplesfrom noncholestatic volunteers (sim2 nM [11]) Privilegingsecretion from liver cells into the blood for subsequenturinary elimination may actually protect the liver againstthe accumulation of such abundant and potentially toxicglucuronide species during cholestasis In the same vein the

improved formation of other conjugates such as CDCA-3Gand DCA-3G which we recently identified as nontoxic BAs[32] may also participate in the hepatic detoxification oftheir unconjugated CDCA and DCA precursors Consistentwith the above stated hypothesis circulating MRs for these2 conjugates (ie 252 and 64 for CDCA-3G and DCA-3Gresp) are spectacularly increased when compared to thosepreviously found in noncholestatic donors (06 and 01) [11]However the fact that circulating and urinary MRs for thesespecies were not associated and the fact that biliary stentingexerted only minor effects on these ratios in urine are alsoindicative of an extrahepatic origin for urine CDCA- andDCA-3G species at least after bile flow restoration

We next compared the human liver and kidney in termsof bile acid glucuronidation activity and BA-conjugatingUGT expression With 2 exceptions (ie the 6- and 10-fold difference in 119870119872 and 119881max values for LCA-3G andHDCA-24G formations resp) these experiments revealthe remarkable similarity of renal and hepatic bile acidglucuronidation processes and the clear preference of bothtissues in producing HDCA-6 and HCA-6G Such preferencemay actually explain the predominance of these conjugates invarious human fluids as previously described [11] Howeverthe most intriguing observation issuing from these analysesrelates to the UGT1A3 enzyme Indeed not only was thisenzyme detected in kidney extracts only at the mRNA(but not protein) level but also the absence of such anactive enzyme previously identified as the main isoform foracyl BA glucuronidation [8ndash10 12] is inconsistent with theelevated acyl glucuronide production capability of kidneymicrosomes A plausible explanation for the conflictingobservation on UGT1A3 mRNA and protein expression inkidney samples relates to the fact that protein and RNAextracts were from different sources and thus were notpaired It is therefore possible that only donors for mRNAexpress these isoforms in the kidney Meanwhile unlikelysuch a possibility is supported by the controversial nature ofthe UGT1A3 expression in the human kidney [29 33ndash35]Additional investigations are therefore warranted to clarifywhether this BA-conjugating protein is found or not in thehuman kidney

Nevertheless the present study clearly establishes themajor contributions that liver and kidney play in glu-curonidating bile acid during cholestasis Interestingly Zhouand colleagues reported that intestinal glucuronidation alsoplays an important role in controlling the local bile acidhomeostasis and the pathological development of colitis [36]These authors elegantly demonstrated that in the intestinebile acids glucuronidation leads to reduced intracellular levelsof ligands for the nuclear receptor FXR Reduced FXR activa-tion limits the secretion of FGF1519 in the portal circulationwhich in turns leads to an increase of de novo formationof bile acid in the liver [36] Thus in contrast to the liverand kidney where glucuronidation is thought to facilitate theremoval of bile acids the intestinal glucuronidation seems toincrease the delivery of bile acidsHowever it should be notedthat during obstructive cholestasis the delivery of hepaticbile acids to the intestine is compromised and thus onlyminor intestinal glucuronidation may occur

Canadian Journal of Gastroenterology and Hepatology 11

5 Conclusion

In conclusion the present study evidences the major impactthat bile flow restoration exerts on the circulating and urinarybile acid-glucuronide profiles and demonstrates the elevatedcapability of the human kidney to glucuronidate theseendogenous compounds Future investigations are requiredto clarify the mechanism allowing kidney extracts to convertbile acids into acyl glucuronides in the absence of theUGT1A3 protein

Abbreviations

BA Bile acidBO Biliary obstructionCA Cholic acidCDCA Chenodeoxycholic acidDCA Deoxycholic acidFXR Farnesoid X receptorG GlucuronideHDCA Hyodeoxycholic acidHCA Hyocholic acidLCA Lithocholic acidLC-MSMS Liquid chromatography-tandem mass

spectrometryUDPGA UDP-glucuronic acidUGT UDP-glucuronosyltransferaseMRPs Multidrug resistance related proteins

Conflicts of Interest

The authors declare that there are no conflicts of interest andthat there is no pertinent financial arrangement concerningthe work presented here

Acknowledgments

The authors wish to thank Dr Virginie Bocher for criticalreading of the manuscript This study was supported bygrants from theCanadian Institute ofHealth Research (CIHR119331) the Canadian Liver Foundation the Natural Sci-ences and Engineering Research Council of Canada (NSERC402213-2012) and the Canadian Foundation for Innovation(CFI 25712)Martin Perreault is holder of a scholarship fromthe ldquoFonds pour la Recherche en Sante du Quebecrdquo (FRSQ23444) Olivier Barbier is holder of a salary grant fromCIHR (New Investigator MSH95330) Piotr Milkiewiczwas supported by Grants DEC-201101BNZ504216 and201102ANZ500321 from the National Science Centre inPoland

Supplementary Materials

The supplementary materials present details of patient withbiliary obstruction (Supplementary Material 1) and thechanges in the urine profile of bile acid glucuronides and intheir metabolic ratios before and after a biliary stenting pro-cedure in patients with biliary obstruction (SupplementaryMaterial 2) Supplementary Material 1 patients with biliary

obstruction (adapted from [20]) Supplementary Material 2changes in the urine profile of bile acid glucuronides andin their metabolic ratios before and after a biliary stentingprocedure in patients with biliary obstruction (119899 = 12)(Supplementary Materials)

References

[1] D W Russell ldquoThe enzymes regulation and genetics of bileacid synthesisrdquo Annual Review of Biochemistry vol 72 pp 137ndash174 2003

[2] M J Monte J J G Marin A Antelo and J Vazquez-TatoldquoBile acids Chemistry physiology and pathophysiologyrdquoWorldJournal of Gastroenterology vol 15 no 7 pp 804ndash816 2009

[3] A F Hofmann ldquoDetoxification of lithocholic acid a toxicbile-acid relevance to drug hepatotoxicityrdquo Drug MetabolismReviews vol 36 no 3-4 pp 703ndash722 2004

[4] C Pauli-Magnus B Stieger Y Meier G A Kullak-Ublick andP J Meier ldquoEnterohepatic transport of bile salts and genetics ofcholestasisrdquo Journal of Hepatology vol 43 no 2 pp 342ndash3572005

[5] G Paumgartner ldquoPharmacotherapy of cholestatic liver dis-easesrdquo Journal of Digestive Diseases vol 11 no 3 pp 119ndash1252010

[6] M Verreault J Kaeding P Caron et al ldquoRegulation ofendobiotics glucuronidation by ligand-activated transcriptionfactors Physiological function and therapeutic potentialrdquoDrugMetabolism Reviews vol 42 no 1 pp 106ndash118 2010

[7] P I Mackenzie K W Bock B Burchell et al ldquoNomenclatureupdate for the mammalian UDP glycosyltransferase (UGT)gene superfamilyrdquo Pharmacogenetics and Genomics vol 15 no10 pp 677ndash685 2005

[8] M H Court S Hazarika S Krishnaswamy M Finel andJ A Williams ldquoNovel polymorphic human UDP-glucurono-syltransferase 2A3 Cloning functional characterization ofenzyme variants comparative tissue expression and geneinductionrdquoMolecular Pharmacology vol 74 no 3 pp 744ndash7542008

[9] W E Gall G Zawada B Mojarrabi et al ldquoDifferential glu-curonidation of bile acids androgens and estrogens by humanUGT1A3 and 2B7rdquo The Journal of Steroid Biochemistry andMolecular Biology vol 70 no 1ndash3 pp 101ndash108 1999

[10] J Trottier D El Husseini M Perreault et al ldquoThe humanUGT1A3 enzyme conjugates norursodeoxycholic acid into aC23-ester glucuronide in the liverrdquo The Journal of BiologicalChemistry vol 285 no 2 pp 1113ndash1121 2010

[11] J Trottier M Perreault I Rudkowska et al ldquoProfiling serumbile acid glucuronides in humans Gender divergences geneticdeterminants and response to fenofibraterdquo Clinical Pharmacol-ogy ampTherapeutics vol 94 no 4 pp 533ndash543 2013

[12] P Caron J Trottier M Verreault J Belanger J Kaeding andO Barbier ldquoEnzymatic production of bile acid glucuronidesused as analytical standards for liquid chromatography-massspectrometry analysesrdquo Molecular Pharmaceutics vol 3 no 3pp 293ndash302 2006

[13] B Alme and J Sjovall ldquoAnalysis of bile acid glucuronidesin urine identification of 3120572612057212120572-trihydroxy-5120573-cholanoicacidrdquoThe Journal of Steroid Biochemistry andMolecular Biologyvol 13 no 8 pp 907ndash916 1980

[14] A Stiehl R Raedsch G Rudolph P Czygan and S WalkerldquoAnalysis of bile acid glucuronides in urine group separation

12 Canadian Journal of Gastroenterology and Hepatology

on a lipophilic anion exchangerrdquo Clinica Chimica Acta vol 123no 3 pp 275ndash285 1982

[15] A Stiehl R Raedsch G Rudolph U Gundert-Remy and MSenn ldquoBiliary and urinary excretion of sulfated glucuronidatedand tetrahydroxylated bile acids in cirrhotic patientsrdquo Hepatol-ogy vol 5 no 3 pp 492ndash495 1985

[16] H Takikawa T Beppu and Y Seyama ldquoUrinary concentrationsof bile acid glucuronides and sulfates in hepatobiliary diseasesrdquoGastroenterologia Japonica vol 19 no 2 pp 104ndash109 1984

[17] H Wietholtz H Marschall R Reuschenbach H Matern andS Matern ldquoUrinary excretion of bile acid glucosides and glu-curonides in extrahepatic cholestasisrdquo Hepatology vol 13 no4 pp 656ndash662 1991

[18] K M Knights A Rowland and J O Miners ldquoRenal drugmetabolism in humans The potential for drug-endobioticinteractions involving cytochrome P450 (CYP) and UDP-glu-curonosyltransferase (UGT)rdquo British Journal of Clinical Phar-macology vol 76 no 4 pp 587ndash602 2013

[19] S Matern H Matern E H Farthmann and W GerokldquoHepatic and extrahepatic glucuronidation of bile acids in manCharacterization of bile acid uridine 5rsquo-diphosphate-glucu-ronosyltransferase in hepatic renal and intestinalmicrosomesrdquoThe Journal of Clinical Investigation vol 74 no 2 pp 402ndash4101984

[20] J Trottier A Białek P Caron R J Straka P Milkiewicz and OBarbier ldquoProfiling circulating and urinary bile acids in patientswith biliary obstruction before and after biliary stentingrdquo PLoSONE vol 6 no 7 Article ID e22094 2011

[21] M Perreault L Gauthier-Landry J Trottier et al ldquoThe humanUDP-glucuronosyltransferase UGT2A1 and UGT2A2 enzymesare highly active in bile acid glucuronidationrdquoDrugMetabolismand Disposition vol 41 no 9 pp 1616ndash1620 2013

[22] M E Campbell D M Grant T Inaba and W KalowldquoBiotransformation of caffeine paraxanthine theophylline andtheobromine by polycyclic aromatic hydrocarbon-induciblecytochrome(s) P-450 in human liver microsomesrdquo DrugMetabolism and Disposition vol 15 no 2 pp 237ndash249 1987

[23] J Lepine O Bernard M Plante et al ldquoSpecificity andregioselectivity of the conjugation of estradiol estrone andtheir catecholestrogen and methoxyestrogen metabolites byhuman uridine diphospho-glucuronosyltransferases expressedin endometriumrdquo The Journal of Clinical Endocrinology ampMetabolism vol 89 no 10 pp 5222ndash5232 2004

[24] J Thibaudeau J Lepine J Tojcic et al ldquoCharacterizationof common UGT1A8 UGT1A9 and UGT2B7 variants withdifferent capacities to inactivate mutagenic 4-hydroxylatedmetabolites of estradiol and estronerdquo Cancer Research vol 66no 1 pp 125ndash133 2006

[25] J O Miners P I MacKenzie and K M Knights ldquoTheprediction of drug-glucuronidation parameters in humansUDP-glucuronosyltransferase enzyme-selective substrate andinhibitor probes for reaction phenotyping and in vitroin vivoextrapolation of drug clearance and drug-drug interactionpotentialrdquo Drug Metabolism Reviews vol 42 no 1 pp 189ndash2012010

[26] M Verreault K Senekeo-Effenberger J Trottier et al ldquoTheliver X-receptor alpha controls hepatic expression of the humanbile acid-glucuronidating UGT1A3 enzyme in human cells andtransgenic micerdquo Hepatology vol 44 no 2 pp 368ndash378 2006

[27] R G Rutledge ldquoMathematics of quantitative kinetic PCR andthe application of standard curvesrdquo Nucleic Acids Research vol31 no 16 pp 93endash93 2003

[28] T Izukawa M Nakajima R Fujiwara et al ldquoQuantitative anal-ysis of UDP-glucuronosyltransferase (UGT) 1A and UGT2Bexpression levels in human liversrdquo Drug Metabolism and Dis-position vol 37 no 8 pp 1759ndash1768 2009

[29] S Ohno and S Nakajin ldquoDetermination of mRNA expressionof human UDP-glucuronosyltransferases and application forlocalization in various human tissues by real-time reversetranscriptase-polymerase chain reactionrdquoDrugMetabolism andDisposition vol 37 no 1 pp 32ndash40 2009

[30] S L Regan J L Maggs T G Hammond C Lambert D PWilliams and B K Park ldquoAcyl glucuronidesThe good the badand the uglyrdquo Biopharmaceutics amp Drug Disposition vol 31 no7 pp 367ndash395 2010

[31] J M Little M V Chari and R Lester ldquoExcretion of cholateglucuroniderdquo Journal of Lipid Research vol 26 no 5 pp 583ndash592 1985

[32] M Perreault A Białek J Trottier et al ldquoRole of glucuronidationfor hepatic detoxification and urinary elimination of toxic bileacids during biliary obstructionrdquo PLoS ONE vol 8 no 11Article ID e80994 2013

[33] H Girard E Levesque J Bellemare K Journault B Caillierand C Guillemette ldquoGenetic diversity at the UGT1 locus isamplified by a novel 31015840 alternative splicing mechanism leadingto nine additional UGT1A proteins that act as regulators ofglucuronidation activityrdquo Pharmacogenetics and Genomics vol17 no 12 pp 1077ndash1089 2007

[34] A Nakamura M Nakajima H Yamanaka R Fujiwara and TYokoi ldquoExpression of UGT1A and UGT2B mRNA in humannormal tissues and various cell linesrdquo Drug Metabolism andDisposition vol 36 no 8 pp 1461ndash1464 2008

[35] D E Harbourt J K Fallon S Ito et al ldquoQuantificationof human uridine-diphosphate glucuronosyl transferase 1Aisoforms in liver intestine and kidney using nanobore liq-uid chromatography-tandem mass spectrometryrdquo AnalyticalChemistry vol 84 no 1 pp 98ndash105 2012

[36] X Zhou L Cao C Jiang et al ldquoPPAR120572-UGT axis activationrepresses intestinal FXR-FGF15 feedback signalling and exacer-bates experimental colitisrdquoNature Communications vol 5 2014

Stem Cells International

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

MEDIATORSINFLAMMATION

of

EndocrinologyInternational Journal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Disease Markers

Hindawiwwwhindawicom Volume 2018

BioMed Research International

OncologyJournal of

Hindawiwwwhindawicom Volume 2013

Hindawiwwwhindawicom Volume 2018

Oxidative Medicine and Cellular Longevity

Hindawiwwwhindawicom Volume 2018

PPAR Research

Hindawi Publishing Corporation httpwwwhindawicom Volume 2013Hindawiwwwhindawicom

The Scientific World Journal

Volume 2018

Immunology ResearchHindawiwwwhindawicom Volume 2018

Journal of

ObesityJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Computational and Mathematical Methods in Medicine

Hindawiwwwhindawicom Volume 2018

Behavioural Neurology

OphthalmologyJournal of

Hindawiwwwhindawicom Volume 2018

Diabetes ResearchJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Research and TreatmentAIDS

Hindawiwwwhindawicom Volume 2018

Gastroenterology Research and Practice

Hindawiwwwhindawicom Volume 2018

Parkinsonrsquos Disease

Evidence-Based Complementary andAlternative Medicine

Volume 2018Hindawiwwwhindawicom

Submit your manuscripts atwwwhindawicom

Page 5: Urinary Elimination of Bile Acid Glucuronides under Severe ...

Canadian Journal of Gastroenterology and Hepatology 5

Table 1 Summary of the correlation studies between serum and urine bile acid-glucuronide profiles

Urine levels Before stenting After stenting Response(1199032 119901 value) (1199032 119901 value) (1199032 119901 value)

CDCA-3G 010 030 002 067 025 010CDCA-24G 001 069 005 050 002 063CA-24G 032 005 051 001 007 042LCA-3G 000 097 019 015 001 081LCA-24G 014 022 047 001 042 002DCA-3G 082 lt00001 014 023 001 071DCA-24G 006 043 016 019 058 0004HDCA-6G 029 007 031 006 035 004HDCA-24G 000 100 005 047 003 059HCA-6G 029 007 015 020 067 0001HCA-24G 000 083 027 008 004 054Correlation analyses were performed using the Spearman rank-order correlation (1199032) and 119901 values for comparisons are indicated Response is calculated asthe difference between post- and prestenting values CA cholic acid CDCA chenodeoxycholic acid DCA deoxycholic acid HCA hyocholic acid HDCAhyodeoxycholic acid LCA lithocholic acid G glucuronide

CDCA-3GCDCA CA-24GCA and DCA-3GDCA ratioswere statistically significant (Figures 2(a) and 2(b)) Otherchanges even the reduction in DCA-24GDCA (117-fold)and LCA-3GLCA (53-fold) or the 25-fold increase of theHDCA-24GHDCA ratio failed to reach statistical signif-icance (Figure 2(b)) In urine (Supplementary Material 2)the unique significant change corresponded to the 52-foldreduction of theCA-24GCA ratio (Figure 2(b)) while the 14-and 15-fold reductions in HCA-6 and -24GMRs also failed toreach statistical significance

33 Serum and Urine BA-G Profiles Are Only Feebly Asso-ciated Correlation studies of circulating and urinary BA-Glevels in paired samples identified only few significant associ-ations (Table 1) In prestenting samples only the serumDCA-3G was significantly and positively correlated (1199032 = 082)to its urinary concentration while in poststenting fluids nocorrelation for this conjugate was detected Serum and urineCA-24G (1199032 = 051) and LCA-24G (1199032 = 047) levels weresignificantly correlated but only in samples obtained afterbile flow restoration (Table 1) When the response to stenting(determined as the difference between post- and prestentingvalues) was analyzed only changes for LCA-24G (1199032 = 042)DCA-24G (1199032 = 058) HDCA-6G (1199032 = 035) and HCA-6G (1199032 = 067) were positively and significantly associated(Table 1) Finally when similar analyses were performed forMRs the unique significant association in prestenting (1199032 =052) and poststenting (1199032 = 045) levels and response(1199032 = 044) was obtained with the ratio CA-24GCA (Figures2(c)ndash2(e))

34 Bile Acid Glucuronidation by Human Liver and KidneyExtracts The limited associations between circulating andurinary profiles observed above suggest a potential extra-hepatic origin for urinary levels of BA-G To test such ahypothesis we sought to compare the kinetic parameters ofBA glucuronidation by human liver (pool of 50 donors) and

kidney (pool of 8 donors) microsomes (Figures 3ndash5) Nodifferences in kinetic models were observed with the BA-specific glucuronidation reactions being distributed among 3models

(i) The sigmoidal pattern for CA-24G and CDCA-24 and-3G (Figure 3) DCA-3 and -24G (Figures 4(c) and 4(d)) andHDCA-6 and -24G (Figures 5(a) and 5(b)) production

(ii) The Michaelis-Menten model for the formation ofLCA-24G (Figure 4(b)) and HCA-6 and -24G (Figures 5(c)and 5(d))

(iii) The substrate-inhibition model for LCA-3G withrespective 119870119868 values of 2403 plusmn 535 and 2554 plusmn 699 120583M forliver and kidney extracts (Figure 4(a))

With the exception of CA-24G HDCA-24G and HCA-24G kidney UGTs generally displayed lower 119870119872 values Forexample kidney microsomes exhibited a 6-fold lower 119870119872value than liver ones for LCA-3G production (Figure 4(a))Another notable difference between liver and kidney micro-somes relates to the 10-fold higher 119881max value of HDCA-24Gproduction obtained with the hepatic microsomal prepara-tion (Figure 5(b)) Nevertheless the most efficient reactionswere obtained with the conversion of HDCA and HCA into6G derivatives that occurred at high velocities (ie 119881max)with both liver and kidney microsomes (Figures 5(a) and5(c)) In both tissue extracts the 6120572-hydroxylated acids weremore actively converted into 6-glucuronide when comparedto their 24-glucuronide counterparts (Figure 5) Interestinglythe opposite was observed for CDCA (Figures 3(a) and 3(b))and LCA (Figures 4(a) and 4(b)) The 119881max values for DCA-3and -24G formation were however similar (Figures 4(c) and4(d))

35 Differential BA-Conjugating UGT Expression in theHuman Liver and Kidney Results from kinetic experimentsindicate that the human kidney possesses an efficient BA-conjugating UGT system We next investigated whetherthe BA-conjugating UGTs 1A3 1A4 2B4 and 2B7 [8ndash11] are differentially expressed in human liver and kidney

6 Canadian Journal of Gastroenterology and Hepatology

02

04

06

08

10CD

CA-3

G fo

rmat

ion

(nm

olh

mg

prot

eins

)

100[CDCA] (M)

200 300 400

Liver nA = 17 plusmn 01

KMB = 821 plusmn 60

VGRC = 08 plusmn 01

Kidney nA = 19 plusmn 07KM

B = 443 plusmn 96VGR

C = 05 plusmn 01

(a)

10

20

30

CDCA

-24G

form

atio

n(n

mol

hm

g pr

otei

ns)

100[CDCA] (M)

200 300 400

Liver nA = 14 plusmn 01KM

B = 1682 plusmn 186VGR

C = 37 plusmn 02

Kidney nA = 13 plusmn 03KM

B = 897 plusmn 298VGR

C = 25 plusmn 04

(b)

05

10

15

20

CA-2

4G fo

rmat

ion

(nm

olh

mg

prot

eins

)

100[CA] (M)

200 300 400

Liver nA = 12 plusmn 01KM

B = 1141 plusmn 168VGR

C = 16 plusmn 01

Kidney nA = 10 plusmn 01KM

B = 1481 plusmn 397VGR

C = 11 plusmn 01

(c)

Figure 3 Kinetic analyses of CDCA-3G (a) CDCA-24G (b) and CA-24G (c) formation by microsomes from human liver and kidney Humanliver (black squares pool of 50 donors) and kidney (white diamonds pool of 8 donors) microsomes (10 120583g) were incubated in the presenceof increasing concentrations (1 to 350 120583M) of chenodeoxycholic acid (CDCA (a) and (b)) or cholic acid (CA (c)) for 2 hours at 37∘C Theformation of CDCA-3G (a) CDCA-24G (b) and CA-24G (c) was analyzed by LC-MSMS For each panel graphs represent the rate ofproduct formation (119910-axis) versus substrate concentration (119909-axis) of two experiments performed in triplicate The listed kinetic parameterswere determined as indicated in ldquoMaterials and Methodsrdquo A119899 Hill coefficient B119870119872 calculated Michaelis constant expressed as 120583M C119881maxcalculated maximal velocity expressed as nmolhmg proteins

Canadian Journal of Gastroenterology and Hepatology 7

200 300 400100[LCA] (M)

40

80

120

160

LCA-

3G fo

rmat

ion

(nm

olh

mg

prot

eins

)

LiverKM

B = 487 plusmn 90VGR

C = 171 plusmn 29

Kidney

KIA = 2403 plusmn 535

KMB = 82 plusmn 22

VGRC = 121 plusmn 11

KIA = 2554 plusmn 699

(a)

200 300 400100[LCA] (M)

60

120

180

240

300

LCA-

24G

form

atio

n (n

mol

hm

g pr

otei

ns)

LiverKM

B = 403 plusmn 67VGR

C = 321 plusmn 16

KidneyKM

B = 305 plusmn 56VGR

C = 162 plusmn 08

(b)

100 200 300 400[DCA] (M)

10

20

30

40

50

60

DCA

-3G

form

atio

n(n

mol

hm

g pr

otei

ns)

Liver nD 17 plusmn 03KM

B = 831 plusmn 113VGR

C = 31 plusmn 02

Kidney nD = 17 plusmn 03KM

B = 823 plusmn 136VGR

C = 36 plusmn 03

(c)

20

40

60

80

DCA

-24G

form

atio

n(n

mol

hm

g pr

otei

ns)

200 300 400100[DCA] (M)

Liver nD 12 plusmn 02KM

B = 711 plusmn 121VGR

C = 58 plusmn 05

Kidney nD = 17 plusmn 03KM

B = 582 plusmn 87VGR

C = 38 plusmn 03

(d)Figure 4Kinetic analyses of LCA-3G (a) LCA-24G (b) DCA-3G (c) and DCA-24G (d) formation by microsomes from human liver and kidneyHuman liver (black squares pool of 50 donors) and kidney (white diamonds pool of 8 donors) microsomes (10 120583g) were incubated in thepresence of increasing concentrations (1 to 350120583M) of lithocholic acid (LCA (a) and (b)) or deoxycholic acid (DCA (c) and (d)) for 2 hoursat 37∘C The formation of LCA-3G (a) LCA-24G (b) DCA-3G (c) and DCA-24G (d) was analyzed by LC-MSMS For each panel graphsrepresent the rate of product formation (119910-axis) versus substrate concentration (119909-axis) of two experiments performed in triplicateThe listedkinetic parameters were determined as indicated in ldquoMaterials and Methodsrdquo A119870119868 constant of inhibition expressed as 120583M B119870119872 calculatedMichaelis constant expressed as 120583M C119881max calculated maximal velocity expressed as nmolhmg proteins D119899 Hill coefficient

8 Canadian Journal of Gastroenterology and Hepatology

1750

3500

5250

7000

HD

CA-6

G fo

rmat

ion

(nm

olh

mg

prot

eins

)

200 300 400100[HDCA] (M)

Liver nA 29 plusmn 04KM

B = 551 plusmn 33VGR

C = 5287 plusmn 211

Kidney nA = 27 plusmn 05KM

B = 447 plusmn 38VGR

C = 3084 plusmn 156

(a)

100

200

300

400

500

HD

CA-2

4G fo

rmat

ion

(nm

olh

mg

prot

eins

)

200 300 400100[HDCA] (M)

Liver nA 38 plusmn 11KM

B = 808 plusmn 64VGR

C = 316 plusmn 22

Kidney nA = 23 plusmn 08KM

B = 939 plusmn 184VGR

C = 37 plusmn 05

(b)

800

1600

2400

3200

HCA

-6G

form

atio

n(n

mol

hm

g pr

otei

ns)

200 300 400100[HCA] (M)

LiverKM

B = 1202 plusmn 156VGR

C = 2285 plusmn 134

KidneyKM

B = 960 plusmn 192VGR

C = 2844 plusmn 241

(c)

20

40

60

HCA

-24G

form

atio

n(n

mol

hm

g pr

otei

ns)

200 300 400100[HCA] (M)

LiverKM

B = 617 plusmn 65VGR

C = 49 plusmn 02

KidneyKM

B = 978 plusmn 206VGR

C = 33 plusmn 03

(d)Figure 5 Kinetic analyses of HDCA-6G (a) HDCA-24G (b) HCA-6G (c) and HCA-24G (d) formation by microsomes from human liver andkidney Human liver (black squares pool of 50 donors) and kidney (white diamonds pool of 8 donors) microsomes (10 120583g) were incubatedin the presence of increasing concentrations (1 to 350120583M) of hyodeoxycholic acid (HDCA (a) and (b)) or hyocholic acid (HCA (c) and(d)) for 2 hours at 37∘CThe formation of HDCA-6G (a) HDCA-24G (b) HCA-6G (c) and HCA-24G (d) was analyzed by LC-MSMS Foreach panel graphs represent the rate of product formation (119910-axis) versus substrate concentration (119909-axis) of two experiments performedin triplicate The listed kinetic parameters were determined as indicated in ldquoMaterials and Methodsrdquo A119899 Hill coefficient B119870119872 calculatedMichaelis constant expressed as 120583M C119881max calculated maximal velocity expressed as nmolhmg proteins

Canadian Journal of Gastroenterology and Hepatology 9

Kid

ney

1A3

Kid

ney

Kid

ney

Kid

ney

Live

r

Live

r

Live

r

Live

r

1A4 2B4 2B7UGT

103

104

105

106

107U

GT

tran

scrip

ts (lo

g of

copy

num

ber

g RN

A)

(a)

1A3

2B42B7

2B7

1A

2B

LiverKidneyAnti-UGT kDa

55

55

55

55

55

40Anti-actin

(b)

Live

r

Kidn

ey

ND

5

10

15

20

UG

T1A

3 pr

otei

n le

vels

(pm

olm

g m

icro

som

es)

(c)

Figure 6 Differential expression of the bile acid-conjugating UGT enzymes in the human liver and kidney (a) UGTs 1A3 1A4 2B4 and 2B7mRNA levels in total RNA samples from human kidney (1 pool of 5 donors and 2 individual donors) and liver (8 individual donors) weredetermined through qRT-PCR analyses as described in ldquoMaterials andMethodsrdquo Results are expressed as log10 ofmRNA copy number per 120583gof total RNA Each symbol represents 1 donor with the exception of the white triangle that corresponds to a pool of the 5 kidney donors (b)The UGT protein contents in microsomal preparations (5120583g) from human kidney (pool of 4 female and 4 male donors) and liver (pool of 26male and 24 female donors) were determined after size separation on SDS-PAGE and hybridization with the anti-UGT1A3 anti-UGT2B47anti-UGT2B7 anti-UGT1A and anti-UGT2B antibodies (1 2000 dilution) The housekeeping protein 120573-actin was used to ensure the equalprotein loading in each lane Data presented are representative of two experiments (c) The UGT1A3 protein content in human liver andkidney microsomes was determined using LC-MSMS analyses as previously described [10] Data represent the mean plusmn SD of two differentexperiments performed in triplicate ND not detected

(Figure 6) With the exception of UGT2B4 transcripts thatwere detected only in 1 kidney mRNA sample other UGTmessengers were detected in all kidney and liver prepara-tions (Figure 6(a)) As expected [28 29] their mRNA levelssustained a strong interindividual variability Western blotanalyses evidenced however major differences between liverand kidney (Figure 6(b)) A clear UGT1A3 immunoreactivecomplex was obtained with liver extracts while this UGTprotein was not detected inmicrosomal proteins from kidney(Figure 6(b)) While the UGT2B7 protein was found atsimilar levels in both tissues the use of an anti-UGT2B42B7antibody indicated that the UGT2B4 protein may be moreabundant in liver than in kidney extracts (Figure 6(b))The lack of an accurate and specific anti-UGT1A4 antibody

impaired the quantification of this BA-conjugating UGThowever we were able to further confirm the absence ofthe UGT1A3 protein in kidney microsomes through the useof our previously established LC-MSMS-based proteomicmethod (Figure 6(c)) [10]

4 Discussion

Urinary and circulating levels of BA-Gs detected in thepresent study are similar to previous findings in terms ofboth glucuronide concentrations and species distribution [1314 16] However an important improvement of the presentinvestigations resides in our ability to discriminate betweenether and ester glucuronides of a single BA For example

10 Canadian Journal of Gastroenterology and Hepatology

while glucuronide conjugates of CDCA CA and DCA werepreviously identified as accumulating metabolites in serafrom patients undergoing bile drainage [20] the nature ofthese glucuronides has not been resolved until now Ourresults point out the ester CDCA-3G and DCA-3G andthe acyl CA-24G as the most abundant glucuronide speciesfound in patients with biliary obstruction Because acyl butnot ester glucuronides can be potentially toxic molecules(reviewed in [30]) such information may be critical inanticipating the consequences of their accumulation duringcholestasis

Interestingly serum levels of some of the bile acidglucuronides such as CDCA-3G CA-24G and LCA-3Gwere significantly reduced after biliary stenting while someothers like CDCA-24G LCA-24G and HDCA-6G weresignificantly increased While additional investigations arewarranted to decipher the mechanisms beyond such a glu-curonide species-dependent responsiveness one can spec-ulate that these changes may actually reflect the differen-tial manner in which the UGT enzymes andor the BA-Gtransporters may be altered upon biliary obstruction andafter stenting Therefore it would have been of interest tocompare how these proteins behave in tissues (ie liver andkidney) from cholestatic donors to validate such a hypothesisActually one limitation of the current study is that the liverand kidneymicrosomes used are not fromdonorswith biliaryobstruction When compared to other glucuronide conju-gates CA-24G presented a unique behavior (1) in contrastto other acyl BA-Gs that are only minor components of theurine and serum glucuronide pools CA-24G is the 3rd mostabundant glucuronidated acid in prestenting samples (2) thisspecies is the most spectacularly affected when poststentingversus prestenting sera profiles are compared (3) the serumCA-24GCA ratio sustains the stronger reduction after biliarystenting (4) this MR is also unique in being significantlyreduced in posttreatment urines and (5) only the CA-24GCA ratio exhibits positive serumurine association interms of prestenting and poststenting distribution as wellas treatment response These last observations support ahepatic origin for the formation of CA-24G and the strongCA-24G accumulation in prestenting fluids and its spec-tacular reduction after bile flow restoration suggests thatunder noncholestatic situations this compound formed inliver cells is normally secreted in bile This hypothesis isfurther supported by previous investigations in which theintravenous injections of cholate glucuronide to rats resultedin a rapid and efficient secretion in bile of the unchangedglucuronide conjugate [31] By contrast when the sameprocedure was applied to bile duct ligated animals 95 of theradioactivity was recovered in urine [31] The present studyindicates that similar rerouting of CA-24G elimination tourine may also occur in cholestatic patients since circulatinglevels detected in prestenting cholestatic donors were almost50-fold higher than those previously quantified in samplesfrom noncholestatic volunteers (sim2 nM [11]) Privilegingsecretion from liver cells into the blood for subsequenturinary elimination may actually protect the liver againstthe accumulation of such abundant and potentially toxicglucuronide species during cholestasis In the same vein the

improved formation of other conjugates such as CDCA-3Gand DCA-3G which we recently identified as nontoxic BAs[32] may also participate in the hepatic detoxification oftheir unconjugated CDCA and DCA precursors Consistentwith the above stated hypothesis circulating MRs for these2 conjugates (ie 252 and 64 for CDCA-3G and DCA-3Gresp) are spectacularly increased when compared to thosepreviously found in noncholestatic donors (06 and 01) [11]However the fact that circulating and urinary MRs for thesespecies were not associated and the fact that biliary stentingexerted only minor effects on these ratios in urine are alsoindicative of an extrahepatic origin for urine CDCA- andDCA-3G species at least after bile flow restoration

We next compared the human liver and kidney in termsof bile acid glucuronidation activity and BA-conjugatingUGT expression With 2 exceptions (ie the 6- and 10-fold difference in 119870119872 and 119881max values for LCA-3G andHDCA-24G formations resp) these experiments revealthe remarkable similarity of renal and hepatic bile acidglucuronidation processes and the clear preference of bothtissues in producing HDCA-6 and HCA-6G Such preferencemay actually explain the predominance of these conjugates invarious human fluids as previously described [11] Howeverthe most intriguing observation issuing from these analysesrelates to the UGT1A3 enzyme Indeed not only was thisenzyme detected in kidney extracts only at the mRNA(but not protein) level but also the absence of such anactive enzyme previously identified as the main isoform foracyl BA glucuronidation [8ndash10 12] is inconsistent with theelevated acyl glucuronide production capability of kidneymicrosomes A plausible explanation for the conflictingobservation on UGT1A3 mRNA and protein expression inkidney samples relates to the fact that protein and RNAextracts were from different sources and thus were notpaired It is therefore possible that only donors for mRNAexpress these isoforms in the kidney Meanwhile unlikelysuch a possibility is supported by the controversial nature ofthe UGT1A3 expression in the human kidney [29 33ndash35]Additional investigations are therefore warranted to clarifywhether this BA-conjugating protein is found or not in thehuman kidney

Nevertheless the present study clearly establishes themajor contributions that liver and kidney play in glu-curonidating bile acid during cholestasis Interestingly Zhouand colleagues reported that intestinal glucuronidation alsoplays an important role in controlling the local bile acidhomeostasis and the pathological development of colitis [36]These authors elegantly demonstrated that in the intestinebile acids glucuronidation leads to reduced intracellular levelsof ligands for the nuclear receptor FXR Reduced FXR activa-tion limits the secretion of FGF1519 in the portal circulationwhich in turns leads to an increase of de novo formationof bile acid in the liver [36] Thus in contrast to the liverand kidney where glucuronidation is thought to facilitate theremoval of bile acids the intestinal glucuronidation seems toincrease the delivery of bile acidsHowever it should be notedthat during obstructive cholestasis the delivery of hepaticbile acids to the intestine is compromised and thus onlyminor intestinal glucuronidation may occur

Canadian Journal of Gastroenterology and Hepatology 11

5 Conclusion

In conclusion the present study evidences the major impactthat bile flow restoration exerts on the circulating and urinarybile acid-glucuronide profiles and demonstrates the elevatedcapability of the human kidney to glucuronidate theseendogenous compounds Future investigations are requiredto clarify the mechanism allowing kidney extracts to convertbile acids into acyl glucuronides in the absence of theUGT1A3 protein

Abbreviations

BA Bile acidBO Biliary obstructionCA Cholic acidCDCA Chenodeoxycholic acidDCA Deoxycholic acidFXR Farnesoid X receptorG GlucuronideHDCA Hyodeoxycholic acidHCA Hyocholic acidLCA Lithocholic acidLC-MSMS Liquid chromatography-tandem mass

spectrometryUDPGA UDP-glucuronic acidUGT UDP-glucuronosyltransferaseMRPs Multidrug resistance related proteins

Conflicts of Interest

The authors declare that there are no conflicts of interest andthat there is no pertinent financial arrangement concerningthe work presented here

Acknowledgments

The authors wish to thank Dr Virginie Bocher for criticalreading of the manuscript This study was supported bygrants from theCanadian Institute ofHealth Research (CIHR119331) the Canadian Liver Foundation the Natural Sci-ences and Engineering Research Council of Canada (NSERC402213-2012) and the Canadian Foundation for Innovation(CFI 25712)Martin Perreault is holder of a scholarship fromthe ldquoFonds pour la Recherche en Sante du Quebecrdquo (FRSQ23444) Olivier Barbier is holder of a salary grant fromCIHR (New Investigator MSH95330) Piotr Milkiewiczwas supported by Grants DEC-201101BNZ504216 and201102ANZ500321 from the National Science Centre inPoland

Supplementary Materials

The supplementary materials present details of patient withbiliary obstruction (Supplementary Material 1) and thechanges in the urine profile of bile acid glucuronides and intheir metabolic ratios before and after a biliary stenting pro-cedure in patients with biliary obstruction (SupplementaryMaterial 2) Supplementary Material 1 patients with biliary

obstruction (adapted from [20]) Supplementary Material 2changes in the urine profile of bile acid glucuronides andin their metabolic ratios before and after a biliary stentingprocedure in patients with biliary obstruction (119899 = 12)(Supplementary Materials)

References

[1] D W Russell ldquoThe enzymes regulation and genetics of bileacid synthesisrdquo Annual Review of Biochemistry vol 72 pp 137ndash174 2003

[2] M J Monte J J G Marin A Antelo and J Vazquez-TatoldquoBile acids Chemistry physiology and pathophysiologyrdquoWorldJournal of Gastroenterology vol 15 no 7 pp 804ndash816 2009

[3] A F Hofmann ldquoDetoxification of lithocholic acid a toxicbile-acid relevance to drug hepatotoxicityrdquo Drug MetabolismReviews vol 36 no 3-4 pp 703ndash722 2004

[4] C Pauli-Magnus B Stieger Y Meier G A Kullak-Ublick andP J Meier ldquoEnterohepatic transport of bile salts and genetics ofcholestasisrdquo Journal of Hepatology vol 43 no 2 pp 342ndash3572005

[5] G Paumgartner ldquoPharmacotherapy of cholestatic liver dis-easesrdquo Journal of Digestive Diseases vol 11 no 3 pp 119ndash1252010

[6] M Verreault J Kaeding P Caron et al ldquoRegulation ofendobiotics glucuronidation by ligand-activated transcriptionfactors Physiological function and therapeutic potentialrdquoDrugMetabolism Reviews vol 42 no 1 pp 106ndash118 2010

[7] P I Mackenzie K W Bock B Burchell et al ldquoNomenclatureupdate for the mammalian UDP glycosyltransferase (UGT)gene superfamilyrdquo Pharmacogenetics and Genomics vol 15 no10 pp 677ndash685 2005

[8] M H Court S Hazarika S Krishnaswamy M Finel andJ A Williams ldquoNovel polymorphic human UDP-glucurono-syltransferase 2A3 Cloning functional characterization ofenzyme variants comparative tissue expression and geneinductionrdquoMolecular Pharmacology vol 74 no 3 pp 744ndash7542008

[9] W E Gall G Zawada B Mojarrabi et al ldquoDifferential glu-curonidation of bile acids androgens and estrogens by humanUGT1A3 and 2B7rdquo The Journal of Steroid Biochemistry andMolecular Biology vol 70 no 1ndash3 pp 101ndash108 1999

[10] J Trottier D El Husseini M Perreault et al ldquoThe humanUGT1A3 enzyme conjugates norursodeoxycholic acid into aC23-ester glucuronide in the liverrdquo The Journal of BiologicalChemistry vol 285 no 2 pp 1113ndash1121 2010

[11] J Trottier M Perreault I Rudkowska et al ldquoProfiling serumbile acid glucuronides in humans Gender divergences geneticdeterminants and response to fenofibraterdquo Clinical Pharmacol-ogy ampTherapeutics vol 94 no 4 pp 533ndash543 2013

[12] P Caron J Trottier M Verreault J Belanger J Kaeding andO Barbier ldquoEnzymatic production of bile acid glucuronidesused as analytical standards for liquid chromatography-massspectrometry analysesrdquo Molecular Pharmaceutics vol 3 no 3pp 293ndash302 2006

[13] B Alme and J Sjovall ldquoAnalysis of bile acid glucuronidesin urine identification of 3120572612057212120572-trihydroxy-5120573-cholanoicacidrdquoThe Journal of Steroid Biochemistry andMolecular Biologyvol 13 no 8 pp 907ndash916 1980

[14] A Stiehl R Raedsch G Rudolph P Czygan and S WalkerldquoAnalysis of bile acid glucuronides in urine group separation

12 Canadian Journal of Gastroenterology and Hepatology

on a lipophilic anion exchangerrdquo Clinica Chimica Acta vol 123no 3 pp 275ndash285 1982

[15] A Stiehl R Raedsch G Rudolph U Gundert-Remy and MSenn ldquoBiliary and urinary excretion of sulfated glucuronidatedand tetrahydroxylated bile acids in cirrhotic patientsrdquo Hepatol-ogy vol 5 no 3 pp 492ndash495 1985

[16] H Takikawa T Beppu and Y Seyama ldquoUrinary concentrationsof bile acid glucuronides and sulfates in hepatobiliary diseasesrdquoGastroenterologia Japonica vol 19 no 2 pp 104ndash109 1984

[17] H Wietholtz H Marschall R Reuschenbach H Matern andS Matern ldquoUrinary excretion of bile acid glucosides and glu-curonides in extrahepatic cholestasisrdquo Hepatology vol 13 no4 pp 656ndash662 1991

[18] K M Knights A Rowland and J O Miners ldquoRenal drugmetabolism in humans The potential for drug-endobioticinteractions involving cytochrome P450 (CYP) and UDP-glu-curonosyltransferase (UGT)rdquo British Journal of Clinical Phar-macology vol 76 no 4 pp 587ndash602 2013

[19] S Matern H Matern E H Farthmann and W GerokldquoHepatic and extrahepatic glucuronidation of bile acids in manCharacterization of bile acid uridine 5rsquo-diphosphate-glucu-ronosyltransferase in hepatic renal and intestinalmicrosomesrdquoThe Journal of Clinical Investigation vol 74 no 2 pp 402ndash4101984

[20] J Trottier A Białek P Caron R J Straka P Milkiewicz and OBarbier ldquoProfiling circulating and urinary bile acids in patientswith biliary obstruction before and after biliary stentingrdquo PLoSONE vol 6 no 7 Article ID e22094 2011

[21] M Perreault L Gauthier-Landry J Trottier et al ldquoThe humanUDP-glucuronosyltransferase UGT2A1 and UGT2A2 enzymesare highly active in bile acid glucuronidationrdquoDrugMetabolismand Disposition vol 41 no 9 pp 1616ndash1620 2013

[22] M E Campbell D M Grant T Inaba and W KalowldquoBiotransformation of caffeine paraxanthine theophylline andtheobromine by polycyclic aromatic hydrocarbon-induciblecytochrome(s) P-450 in human liver microsomesrdquo DrugMetabolism and Disposition vol 15 no 2 pp 237ndash249 1987

[23] J Lepine O Bernard M Plante et al ldquoSpecificity andregioselectivity of the conjugation of estradiol estrone andtheir catecholestrogen and methoxyestrogen metabolites byhuman uridine diphospho-glucuronosyltransferases expressedin endometriumrdquo The Journal of Clinical Endocrinology ampMetabolism vol 89 no 10 pp 5222ndash5232 2004

[24] J Thibaudeau J Lepine J Tojcic et al ldquoCharacterizationof common UGT1A8 UGT1A9 and UGT2B7 variants withdifferent capacities to inactivate mutagenic 4-hydroxylatedmetabolites of estradiol and estronerdquo Cancer Research vol 66no 1 pp 125ndash133 2006

[25] J O Miners P I MacKenzie and K M Knights ldquoTheprediction of drug-glucuronidation parameters in humansUDP-glucuronosyltransferase enzyme-selective substrate andinhibitor probes for reaction phenotyping and in vitroin vivoextrapolation of drug clearance and drug-drug interactionpotentialrdquo Drug Metabolism Reviews vol 42 no 1 pp 189ndash2012010

[26] M Verreault K Senekeo-Effenberger J Trottier et al ldquoTheliver X-receptor alpha controls hepatic expression of the humanbile acid-glucuronidating UGT1A3 enzyme in human cells andtransgenic micerdquo Hepatology vol 44 no 2 pp 368ndash378 2006

[27] R G Rutledge ldquoMathematics of quantitative kinetic PCR andthe application of standard curvesrdquo Nucleic Acids Research vol31 no 16 pp 93endash93 2003

[28] T Izukawa M Nakajima R Fujiwara et al ldquoQuantitative anal-ysis of UDP-glucuronosyltransferase (UGT) 1A and UGT2Bexpression levels in human liversrdquo Drug Metabolism and Dis-position vol 37 no 8 pp 1759ndash1768 2009

[29] S Ohno and S Nakajin ldquoDetermination of mRNA expressionof human UDP-glucuronosyltransferases and application forlocalization in various human tissues by real-time reversetranscriptase-polymerase chain reactionrdquoDrugMetabolism andDisposition vol 37 no 1 pp 32ndash40 2009

[30] S L Regan J L Maggs T G Hammond C Lambert D PWilliams and B K Park ldquoAcyl glucuronidesThe good the badand the uglyrdquo Biopharmaceutics amp Drug Disposition vol 31 no7 pp 367ndash395 2010

[31] J M Little M V Chari and R Lester ldquoExcretion of cholateglucuroniderdquo Journal of Lipid Research vol 26 no 5 pp 583ndash592 1985

[32] M Perreault A Białek J Trottier et al ldquoRole of glucuronidationfor hepatic detoxification and urinary elimination of toxic bileacids during biliary obstructionrdquo PLoS ONE vol 8 no 11Article ID e80994 2013

[33] H Girard E Levesque J Bellemare K Journault B Caillierand C Guillemette ldquoGenetic diversity at the UGT1 locus isamplified by a novel 31015840 alternative splicing mechanism leadingto nine additional UGT1A proteins that act as regulators ofglucuronidation activityrdquo Pharmacogenetics and Genomics vol17 no 12 pp 1077ndash1089 2007

[34] A Nakamura M Nakajima H Yamanaka R Fujiwara and TYokoi ldquoExpression of UGT1A and UGT2B mRNA in humannormal tissues and various cell linesrdquo Drug Metabolism andDisposition vol 36 no 8 pp 1461ndash1464 2008

[35] D E Harbourt J K Fallon S Ito et al ldquoQuantificationof human uridine-diphosphate glucuronosyl transferase 1Aisoforms in liver intestine and kidney using nanobore liq-uid chromatography-tandem mass spectrometryrdquo AnalyticalChemistry vol 84 no 1 pp 98ndash105 2012

[36] X Zhou L Cao C Jiang et al ldquoPPAR120572-UGT axis activationrepresses intestinal FXR-FGF15 feedback signalling and exacer-bates experimental colitisrdquoNature Communications vol 5 2014

Stem Cells International

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

MEDIATORSINFLAMMATION

of

EndocrinologyInternational Journal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Disease Markers

Hindawiwwwhindawicom Volume 2018

BioMed Research International

OncologyJournal of

Hindawiwwwhindawicom Volume 2013

Hindawiwwwhindawicom Volume 2018

Oxidative Medicine and Cellular Longevity

Hindawiwwwhindawicom Volume 2018

PPAR Research

Hindawi Publishing Corporation httpwwwhindawicom Volume 2013Hindawiwwwhindawicom

The Scientific World Journal

Volume 2018

Immunology ResearchHindawiwwwhindawicom Volume 2018

Journal of

ObesityJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Computational and Mathematical Methods in Medicine

Hindawiwwwhindawicom Volume 2018

Behavioural Neurology

OphthalmologyJournal of

Hindawiwwwhindawicom Volume 2018

Diabetes ResearchJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Research and TreatmentAIDS

Hindawiwwwhindawicom Volume 2018

Gastroenterology Research and Practice

Hindawiwwwhindawicom Volume 2018

Parkinsonrsquos Disease

Evidence-Based Complementary andAlternative Medicine

Volume 2018Hindawiwwwhindawicom

Submit your manuscripts atwwwhindawicom

Page 6: Urinary Elimination of Bile Acid Glucuronides under Severe ...

6 Canadian Journal of Gastroenterology and Hepatology

02

04

06

08

10CD

CA-3

G fo

rmat

ion

(nm

olh

mg

prot

eins

)

100[CDCA] (M)

200 300 400

Liver nA = 17 plusmn 01

KMB = 821 plusmn 60

VGRC = 08 plusmn 01

Kidney nA = 19 plusmn 07KM

B = 443 plusmn 96VGR

C = 05 plusmn 01

(a)

10

20

30

CDCA

-24G

form

atio

n(n

mol

hm

g pr

otei

ns)

100[CDCA] (M)

200 300 400

Liver nA = 14 plusmn 01KM

B = 1682 plusmn 186VGR

C = 37 plusmn 02

Kidney nA = 13 plusmn 03KM

B = 897 plusmn 298VGR

C = 25 plusmn 04

(b)

05

10

15

20

CA-2

4G fo

rmat

ion

(nm

olh

mg

prot

eins

)

100[CA] (M)

200 300 400

Liver nA = 12 plusmn 01KM

B = 1141 plusmn 168VGR

C = 16 plusmn 01

Kidney nA = 10 plusmn 01KM

B = 1481 plusmn 397VGR

C = 11 plusmn 01

(c)

Figure 3 Kinetic analyses of CDCA-3G (a) CDCA-24G (b) and CA-24G (c) formation by microsomes from human liver and kidney Humanliver (black squares pool of 50 donors) and kidney (white diamonds pool of 8 donors) microsomes (10 120583g) were incubated in the presenceof increasing concentrations (1 to 350 120583M) of chenodeoxycholic acid (CDCA (a) and (b)) or cholic acid (CA (c)) for 2 hours at 37∘C Theformation of CDCA-3G (a) CDCA-24G (b) and CA-24G (c) was analyzed by LC-MSMS For each panel graphs represent the rate ofproduct formation (119910-axis) versus substrate concentration (119909-axis) of two experiments performed in triplicate The listed kinetic parameterswere determined as indicated in ldquoMaterials and Methodsrdquo A119899 Hill coefficient B119870119872 calculated Michaelis constant expressed as 120583M C119881maxcalculated maximal velocity expressed as nmolhmg proteins

Canadian Journal of Gastroenterology and Hepatology 7

200 300 400100[LCA] (M)

40

80

120

160

LCA-

3G fo

rmat

ion

(nm

olh

mg

prot

eins

)

LiverKM

B = 487 plusmn 90VGR

C = 171 plusmn 29

Kidney

KIA = 2403 plusmn 535

KMB = 82 plusmn 22

VGRC = 121 plusmn 11

KIA = 2554 plusmn 699

(a)

200 300 400100[LCA] (M)

60

120

180

240

300

LCA-

24G

form

atio

n (n

mol

hm

g pr

otei

ns)

LiverKM

B = 403 plusmn 67VGR

C = 321 plusmn 16

KidneyKM

B = 305 plusmn 56VGR

C = 162 plusmn 08

(b)

100 200 300 400[DCA] (M)

10

20

30

40

50

60

DCA

-3G

form

atio

n(n

mol

hm

g pr

otei

ns)

Liver nD 17 plusmn 03KM

B = 831 plusmn 113VGR

C = 31 plusmn 02

Kidney nD = 17 plusmn 03KM

B = 823 plusmn 136VGR

C = 36 plusmn 03

(c)

20

40

60

80

DCA

-24G

form

atio

n(n

mol

hm

g pr

otei

ns)

200 300 400100[DCA] (M)

Liver nD 12 plusmn 02KM

B = 711 plusmn 121VGR

C = 58 plusmn 05

Kidney nD = 17 plusmn 03KM

B = 582 plusmn 87VGR

C = 38 plusmn 03

(d)Figure 4Kinetic analyses of LCA-3G (a) LCA-24G (b) DCA-3G (c) and DCA-24G (d) formation by microsomes from human liver and kidneyHuman liver (black squares pool of 50 donors) and kidney (white diamonds pool of 8 donors) microsomes (10 120583g) were incubated in thepresence of increasing concentrations (1 to 350120583M) of lithocholic acid (LCA (a) and (b)) or deoxycholic acid (DCA (c) and (d)) for 2 hoursat 37∘C The formation of LCA-3G (a) LCA-24G (b) DCA-3G (c) and DCA-24G (d) was analyzed by LC-MSMS For each panel graphsrepresent the rate of product formation (119910-axis) versus substrate concentration (119909-axis) of two experiments performed in triplicateThe listedkinetic parameters were determined as indicated in ldquoMaterials and Methodsrdquo A119870119868 constant of inhibition expressed as 120583M B119870119872 calculatedMichaelis constant expressed as 120583M C119881max calculated maximal velocity expressed as nmolhmg proteins D119899 Hill coefficient

8 Canadian Journal of Gastroenterology and Hepatology

1750

3500

5250

7000

HD

CA-6

G fo

rmat

ion

(nm

olh

mg

prot

eins

)

200 300 400100[HDCA] (M)

Liver nA 29 plusmn 04KM

B = 551 plusmn 33VGR

C = 5287 plusmn 211

Kidney nA = 27 plusmn 05KM

B = 447 plusmn 38VGR

C = 3084 plusmn 156

(a)

100

200

300

400

500

HD

CA-2

4G fo

rmat

ion

(nm

olh

mg

prot

eins

)

200 300 400100[HDCA] (M)

Liver nA 38 plusmn 11KM

B = 808 plusmn 64VGR

C = 316 plusmn 22

Kidney nA = 23 plusmn 08KM

B = 939 plusmn 184VGR

C = 37 plusmn 05

(b)

800

1600

2400

3200

HCA

-6G

form

atio

n(n

mol

hm

g pr

otei

ns)

200 300 400100[HCA] (M)

LiverKM

B = 1202 plusmn 156VGR

C = 2285 plusmn 134

KidneyKM

B = 960 plusmn 192VGR

C = 2844 plusmn 241

(c)

20

40

60

HCA

-24G

form

atio

n(n

mol

hm

g pr

otei

ns)

200 300 400100[HCA] (M)

LiverKM

B = 617 plusmn 65VGR

C = 49 plusmn 02

KidneyKM

B = 978 plusmn 206VGR

C = 33 plusmn 03

(d)Figure 5 Kinetic analyses of HDCA-6G (a) HDCA-24G (b) HCA-6G (c) and HCA-24G (d) formation by microsomes from human liver andkidney Human liver (black squares pool of 50 donors) and kidney (white diamonds pool of 8 donors) microsomes (10 120583g) were incubatedin the presence of increasing concentrations (1 to 350120583M) of hyodeoxycholic acid (HDCA (a) and (b)) or hyocholic acid (HCA (c) and(d)) for 2 hours at 37∘CThe formation of HDCA-6G (a) HDCA-24G (b) HCA-6G (c) and HCA-24G (d) was analyzed by LC-MSMS Foreach panel graphs represent the rate of product formation (119910-axis) versus substrate concentration (119909-axis) of two experiments performedin triplicate The listed kinetic parameters were determined as indicated in ldquoMaterials and Methodsrdquo A119899 Hill coefficient B119870119872 calculatedMichaelis constant expressed as 120583M C119881max calculated maximal velocity expressed as nmolhmg proteins

Canadian Journal of Gastroenterology and Hepatology 9

Kid

ney

1A3

Kid

ney

Kid

ney

Kid

ney

Live

r

Live

r

Live

r

Live

r

1A4 2B4 2B7UGT

103

104

105

106

107U

GT

tran

scrip

ts (lo

g of

copy

num

ber

g RN

A)

(a)

1A3

2B42B7

2B7

1A

2B

LiverKidneyAnti-UGT kDa

55

55

55

55

55

40Anti-actin

(b)

Live

r

Kidn

ey

ND

5

10

15

20

UG

T1A

3 pr

otei

n le

vels

(pm

olm

g m

icro

som

es)

(c)

Figure 6 Differential expression of the bile acid-conjugating UGT enzymes in the human liver and kidney (a) UGTs 1A3 1A4 2B4 and 2B7mRNA levels in total RNA samples from human kidney (1 pool of 5 donors and 2 individual donors) and liver (8 individual donors) weredetermined through qRT-PCR analyses as described in ldquoMaterials andMethodsrdquo Results are expressed as log10 ofmRNA copy number per 120583gof total RNA Each symbol represents 1 donor with the exception of the white triangle that corresponds to a pool of the 5 kidney donors (b)The UGT protein contents in microsomal preparations (5120583g) from human kidney (pool of 4 female and 4 male donors) and liver (pool of 26male and 24 female donors) were determined after size separation on SDS-PAGE and hybridization with the anti-UGT1A3 anti-UGT2B47anti-UGT2B7 anti-UGT1A and anti-UGT2B antibodies (1 2000 dilution) The housekeeping protein 120573-actin was used to ensure the equalprotein loading in each lane Data presented are representative of two experiments (c) The UGT1A3 protein content in human liver andkidney microsomes was determined using LC-MSMS analyses as previously described [10] Data represent the mean plusmn SD of two differentexperiments performed in triplicate ND not detected

(Figure 6) With the exception of UGT2B4 transcripts thatwere detected only in 1 kidney mRNA sample other UGTmessengers were detected in all kidney and liver prepara-tions (Figure 6(a)) As expected [28 29] their mRNA levelssustained a strong interindividual variability Western blotanalyses evidenced however major differences between liverand kidney (Figure 6(b)) A clear UGT1A3 immunoreactivecomplex was obtained with liver extracts while this UGTprotein was not detected inmicrosomal proteins from kidney(Figure 6(b)) While the UGT2B7 protein was found atsimilar levels in both tissues the use of an anti-UGT2B42B7antibody indicated that the UGT2B4 protein may be moreabundant in liver than in kidney extracts (Figure 6(b))The lack of an accurate and specific anti-UGT1A4 antibody

impaired the quantification of this BA-conjugating UGThowever we were able to further confirm the absence ofthe UGT1A3 protein in kidney microsomes through the useof our previously established LC-MSMS-based proteomicmethod (Figure 6(c)) [10]

4 Discussion

Urinary and circulating levels of BA-Gs detected in thepresent study are similar to previous findings in terms ofboth glucuronide concentrations and species distribution [1314 16] However an important improvement of the presentinvestigations resides in our ability to discriminate betweenether and ester glucuronides of a single BA For example

10 Canadian Journal of Gastroenterology and Hepatology

while glucuronide conjugates of CDCA CA and DCA werepreviously identified as accumulating metabolites in serafrom patients undergoing bile drainage [20] the nature ofthese glucuronides has not been resolved until now Ourresults point out the ester CDCA-3G and DCA-3G andthe acyl CA-24G as the most abundant glucuronide speciesfound in patients with biliary obstruction Because acyl butnot ester glucuronides can be potentially toxic molecules(reviewed in [30]) such information may be critical inanticipating the consequences of their accumulation duringcholestasis

Interestingly serum levels of some of the bile acidglucuronides such as CDCA-3G CA-24G and LCA-3Gwere significantly reduced after biliary stenting while someothers like CDCA-24G LCA-24G and HDCA-6G weresignificantly increased While additional investigations arewarranted to decipher the mechanisms beyond such a glu-curonide species-dependent responsiveness one can spec-ulate that these changes may actually reflect the differen-tial manner in which the UGT enzymes andor the BA-Gtransporters may be altered upon biliary obstruction andafter stenting Therefore it would have been of interest tocompare how these proteins behave in tissues (ie liver andkidney) from cholestatic donors to validate such a hypothesisActually one limitation of the current study is that the liverand kidneymicrosomes used are not fromdonorswith biliaryobstruction When compared to other glucuronide conju-gates CA-24G presented a unique behavior (1) in contrastto other acyl BA-Gs that are only minor components of theurine and serum glucuronide pools CA-24G is the 3rd mostabundant glucuronidated acid in prestenting samples (2) thisspecies is the most spectacularly affected when poststentingversus prestenting sera profiles are compared (3) the serumCA-24GCA ratio sustains the stronger reduction after biliarystenting (4) this MR is also unique in being significantlyreduced in posttreatment urines and (5) only the CA-24GCA ratio exhibits positive serumurine association interms of prestenting and poststenting distribution as wellas treatment response These last observations support ahepatic origin for the formation of CA-24G and the strongCA-24G accumulation in prestenting fluids and its spec-tacular reduction after bile flow restoration suggests thatunder noncholestatic situations this compound formed inliver cells is normally secreted in bile This hypothesis isfurther supported by previous investigations in which theintravenous injections of cholate glucuronide to rats resultedin a rapid and efficient secretion in bile of the unchangedglucuronide conjugate [31] By contrast when the sameprocedure was applied to bile duct ligated animals 95 of theradioactivity was recovered in urine [31] The present studyindicates that similar rerouting of CA-24G elimination tourine may also occur in cholestatic patients since circulatinglevels detected in prestenting cholestatic donors were almost50-fold higher than those previously quantified in samplesfrom noncholestatic volunteers (sim2 nM [11]) Privilegingsecretion from liver cells into the blood for subsequenturinary elimination may actually protect the liver againstthe accumulation of such abundant and potentially toxicglucuronide species during cholestasis In the same vein the

improved formation of other conjugates such as CDCA-3Gand DCA-3G which we recently identified as nontoxic BAs[32] may also participate in the hepatic detoxification oftheir unconjugated CDCA and DCA precursors Consistentwith the above stated hypothesis circulating MRs for these2 conjugates (ie 252 and 64 for CDCA-3G and DCA-3Gresp) are spectacularly increased when compared to thosepreviously found in noncholestatic donors (06 and 01) [11]However the fact that circulating and urinary MRs for thesespecies were not associated and the fact that biliary stentingexerted only minor effects on these ratios in urine are alsoindicative of an extrahepatic origin for urine CDCA- andDCA-3G species at least after bile flow restoration

We next compared the human liver and kidney in termsof bile acid glucuronidation activity and BA-conjugatingUGT expression With 2 exceptions (ie the 6- and 10-fold difference in 119870119872 and 119881max values for LCA-3G andHDCA-24G formations resp) these experiments revealthe remarkable similarity of renal and hepatic bile acidglucuronidation processes and the clear preference of bothtissues in producing HDCA-6 and HCA-6G Such preferencemay actually explain the predominance of these conjugates invarious human fluids as previously described [11] Howeverthe most intriguing observation issuing from these analysesrelates to the UGT1A3 enzyme Indeed not only was thisenzyme detected in kidney extracts only at the mRNA(but not protein) level but also the absence of such anactive enzyme previously identified as the main isoform foracyl BA glucuronidation [8ndash10 12] is inconsistent with theelevated acyl glucuronide production capability of kidneymicrosomes A plausible explanation for the conflictingobservation on UGT1A3 mRNA and protein expression inkidney samples relates to the fact that protein and RNAextracts were from different sources and thus were notpaired It is therefore possible that only donors for mRNAexpress these isoforms in the kidney Meanwhile unlikelysuch a possibility is supported by the controversial nature ofthe UGT1A3 expression in the human kidney [29 33ndash35]Additional investigations are therefore warranted to clarifywhether this BA-conjugating protein is found or not in thehuman kidney

Nevertheless the present study clearly establishes themajor contributions that liver and kidney play in glu-curonidating bile acid during cholestasis Interestingly Zhouand colleagues reported that intestinal glucuronidation alsoplays an important role in controlling the local bile acidhomeostasis and the pathological development of colitis [36]These authors elegantly demonstrated that in the intestinebile acids glucuronidation leads to reduced intracellular levelsof ligands for the nuclear receptor FXR Reduced FXR activa-tion limits the secretion of FGF1519 in the portal circulationwhich in turns leads to an increase of de novo formationof bile acid in the liver [36] Thus in contrast to the liverand kidney where glucuronidation is thought to facilitate theremoval of bile acids the intestinal glucuronidation seems toincrease the delivery of bile acidsHowever it should be notedthat during obstructive cholestasis the delivery of hepaticbile acids to the intestine is compromised and thus onlyminor intestinal glucuronidation may occur

Canadian Journal of Gastroenterology and Hepatology 11

5 Conclusion

In conclusion the present study evidences the major impactthat bile flow restoration exerts on the circulating and urinarybile acid-glucuronide profiles and demonstrates the elevatedcapability of the human kidney to glucuronidate theseendogenous compounds Future investigations are requiredto clarify the mechanism allowing kidney extracts to convertbile acids into acyl glucuronides in the absence of theUGT1A3 protein

Abbreviations

BA Bile acidBO Biliary obstructionCA Cholic acidCDCA Chenodeoxycholic acidDCA Deoxycholic acidFXR Farnesoid X receptorG GlucuronideHDCA Hyodeoxycholic acidHCA Hyocholic acidLCA Lithocholic acidLC-MSMS Liquid chromatography-tandem mass

spectrometryUDPGA UDP-glucuronic acidUGT UDP-glucuronosyltransferaseMRPs Multidrug resistance related proteins

Conflicts of Interest

The authors declare that there are no conflicts of interest andthat there is no pertinent financial arrangement concerningthe work presented here

Acknowledgments

The authors wish to thank Dr Virginie Bocher for criticalreading of the manuscript This study was supported bygrants from theCanadian Institute ofHealth Research (CIHR119331) the Canadian Liver Foundation the Natural Sci-ences and Engineering Research Council of Canada (NSERC402213-2012) and the Canadian Foundation for Innovation(CFI 25712)Martin Perreault is holder of a scholarship fromthe ldquoFonds pour la Recherche en Sante du Quebecrdquo (FRSQ23444) Olivier Barbier is holder of a salary grant fromCIHR (New Investigator MSH95330) Piotr Milkiewiczwas supported by Grants DEC-201101BNZ504216 and201102ANZ500321 from the National Science Centre inPoland

Supplementary Materials

The supplementary materials present details of patient withbiliary obstruction (Supplementary Material 1) and thechanges in the urine profile of bile acid glucuronides and intheir metabolic ratios before and after a biliary stenting pro-cedure in patients with biliary obstruction (SupplementaryMaterial 2) Supplementary Material 1 patients with biliary

obstruction (adapted from [20]) Supplementary Material 2changes in the urine profile of bile acid glucuronides andin their metabolic ratios before and after a biliary stentingprocedure in patients with biliary obstruction (119899 = 12)(Supplementary Materials)

References

[1] D W Russell ldquoThe enzymes regulation and genetics of bileacid synthesisrdquo Annual Review of Biochemistry vol 72 pp 137ndash174 2003

[2] M J Monte J J G Marin A Antelo and J Vazquez-TatoldquoBile acids Chemistry physiology and pathophysiologyrdquoWorldJournal of Gastroenterology vol 15 no 7 pp 804ndash816 2009

[3] A F Hofmann ldquoDetoxification of lithocholic acid a toxicbile-acid relevance to drug hepatotoxicityrdquo Drug MetabolismReviews vol 36 no 3-4 pp 703ndash722 2004

[4] C Pauli-Magnus B Stieger Y Meier G A Kullak-Ublick andP J Meier ldquoEnterohepatic transport of bile salts and genetics ofcholestasisrdquo Journal of Hepatology vol 43 no 2 pp 342ndash3572005

[5] G Paumgartner ldquoPharmacotherapy of cholestatic liver dis-easesrdquo Journal of Digestive Diseases vol 11 no 3 pp 119ndash1252010

[6] M Verreault J Kaeding P Caron et al ldquoRegulation ofendobiotics glucuronidation by ligand-activated transcriptionfactors Physiological function and therapeutic potentialrdquoDrugMetabolism Reviews vol 42 no 1 pp 106ndash118 2010

[7] P I Mackenzie K W Bock B Burchell et al ldquoNomenclatureupdate for the mammalian UDP glycosyltransferase (UGT)gene superfamilyrdquo Pharmacogenetics and Genomics vol 15 no10 pp 677ndash685 2005

[8] M H Court S Hazarika S Krishnaswamy M Finel andJ A Williams ldquoNovel polymorphic human UDP-glucurono-syltransferase 2A3 Cloning functional characterization ofenzyme variants comparative tissue expression and geneinductionrdquoMolecular Pharmacology vol 74 no 3 pp 744ndash7542008

[9] W E Gall G Zawada B Mojarrabi et al ldquoDifferential glu-curonidation of bile acids androgens and estrogens by humanUGT1A3 and 2B7rdquo The Journal of Steroid Biochemistry andMolecular Biology vol 70 no 1ndash3 pp 101ndash108 1999

[10] J Trottier D El Husseini M Perreault et al ldquoThe humanUGT1A3 enzyme conjugates norursodeoxycholic acid into aC23-ester glucuronide in the liverrdquo The Journal of BiologicalChemistry vol 285 no 2 pp 1113ndash1121 2010

[11] J Trottier M Perreault I Rudkowska et al ldquoProfiling serumbile acid glucuronides in humans Gender divergences geneticdeterminants and response to fenofibraterdquo Clinical Pharmacol-ogy ampTherapeutics vol 94 no 4 pp 533ndash543 2013

[12] P Caron J Trottier M Verreault J Belanger J Kaeding andO Barbier ldquoEnzymatic production of bile acid glucuronidesused as analytical standards for liquid chromatography-massspectrometry analysesrdquo Molecular Pharmaceutics vol 3 no 3pp 293ndash302 2006

[13] B Alme and J Sjovall ldquoAnalysis of bile acid glucuronidesin urine identification of 3120572612057212120572-trihydroxy-5120573-cholanoicacidrdquoThe Journal of Steroid Biochemistry andMolecular Biologyvol 13 no 8 pp 907ndash916 1980

[14] A Stiehl R Raedsch G Rudolph P Czygan and S WalkerldquoAnalysis of bile acid glucuronides in urine group separation

12 Canadian Journal of Gastroenterology and Hepatology

on a lipophilic anion exchangerrdquo Clinica Chimica Acta vol 123no 3 pp 275ndash285 1982

[15] A Stiehl R Raedsch G Rudolph U Gundert-Remy and MSenn ldquoBiliary and urinary excretion of sulfated glucuronidatedand tetrahydroxylated bile acids in cirrhotic patientsrdquo Hepatol-ogy vol 5 no 3 pp 492ndash495 1985

[16] H Takikawa T Beppu and Y Seyama ldquoUrinary concentrationsof bile acid glucuronides and sulfates in hepatobiliary diseasesrdquoGastroenterologia Japonica vol 19 no 2 pp 104ndash109 1984

[17] H Wietholtz H Marschall R Reuschenbach H Matern andS Matern ldquoUrinary excretion of bile acid glucosides and glu-curonides in extrahepatic cholestasisrdquo Hepatology vol 13 no4 pp 656ndash662 1991

[18] K M Knights A Rowland and J O Miners ldquoRenal drugmetabolism in humans The potential for drug-endobioticinteractions involving cytochrome P450 (CYP) and UDP-glu-curonosyltransferase (UGT)rdquo British Journal of Clinical Phar-macology vol 76 no 4 pp 587ndash602 2013

[19] S Matern H Matern E H Farthmann and W GerokldquoHepatic and extrahepatic glucuronidation of bile acids in manCharacterization of bile acid uridine 5rsquo-diphosphate-glucu-ronosyltransferase in hepatic renal and intestinalmicrosomesrdquoThe Journal of Clinical Investigation vol 74 no 2 pp 402ndash4101984

[20] J Trottier A Białek P Caron R J Straka P Milkiewicz and OBarbier ldquoProfiling circulating and urinary bile acids in patientswith biliary obstruction before and after biliary stentingrdquo PLoSONE vol 6 no 7 Article ID e22094 2011

[21] M Perreault L Gauthier-Landry J Trottier et al ldquoThe humanUDP-glucuronosyltransferase UGT2A1 and UGT2A2 enzymesare highly active in bile acid glucuronidationrdquoDrugMetabolismand Disposition vol 41 no 9 pp 1616ndash1620 2013

[22] M E Campbell D M Grant T Inaba and W KalowldquoBiotransformation of caffeine paraxanthine theophylline andtheobromine by polycyclic aromatic hydrocarbon-induciblecytochrome(s) P-450 in human liver microsomesrdquo DrugMetabolism and Disposition vol 15 no 2 pp 237ndash249 1987

[23] J Lepine O Bernard M Plante et al ldquoSpecificity andregioselectivity of the conjugation of estradiol estrone andtheir catecholestrogen and methoxyestrogen metabolites byhuman uridine diphospho-glucuronosyltransferases expressedin endometriumrdquo The Journal of Clinical Endocrinology ampMetabolism vol 89 no 10 pp 5222ndash5232 2004

[24] J Thibaudeau J Lepine J Tojcic et al ldquoCharacterizationof common UGT1A8 UGT1A9 and UGT2B7 variants withdifferent capacities to inactivate mutagenic 4-hydroxylatedmetabolites of estradiol and estronerdquo Cancer Research vol 66no 1 pp 125ndash133 2006

[25] J O Miners P I MacKenzie and K M Knights ldquoTheprediction of drug-glucuronidation parameters in humansUDP-glucuronosyltransferase enzyme-selective substrate andinhibitor probes for reaction phenotyping and in vitroin vivoextrapolation of drug clearance and drug-drug interactionpotentialrdquo Drug Metabolism Reviews vol 42 no 1 pp 189ndash2012010

[26] M Verreault K Senekeo-Effenberger J Trottier et al ldquoTheliver X-receptor alpha controls hepatic expression of the humanbile acid-glucuronidating UGT1A3 enzyme in human cells andtransgenic micerdquo Hepatology vol 44 no 2 pp 368ndash378 2006

[27] R G Rutledge ldquoMathematics of quantitative kinetic PCR andthe application of standard curvesrdquo Nucleic Acids Research vol31 no 16 pp 93endash93 2003

[28] T Izukawa M Nakajima R Fujiwara et al ldquoQuantitative anal-ysis of UDP-glucuronosyltransferase (UGT) 1A and UGT2Bexpression levels in human liversrdquo Drug Metabolism and Dis-position vol 37 no 8 pp 1759ndash1768 2009

[29] S Ohno and S Nakajin ldquoDetermination of mRNA expressionof human UDP-glucuronosyltransferases and application forlocalization in various human tissues by real-time reversetranscriptase-polymerase chain reactionrdquoDrugMetabolism andDisposition vol 37 no 1 pp 32ndash40 2009

[30] S L Regan J L Maggs T G Hammond C Lambert D PWilliams and B K Park ldquoAcyl glucuronidesThe good the badand the uglyrdquo Biopharmaceutics amp Drug Disposition vol 31 no7 pp 367ndash395 2010

[31] J M Little M V Chari and R Lester ldquoExcretion of cholateglucuroniderdquo Journal of Lipid Research vol 26 no 5 pp 583ndash592 1985

[32] M Perreault A Białek J Trottier et al ldquoRole of glucuronidationfor hepatic detoxification and urinary elimination of toxic bileacids during biliary obstructionrdquo PLoS ONE vol 8 no 11Article ID e80994 2013

[33] H Girard E Levesque J Bellemare K Journault B Caillierand C Guillemette ldquoGenetic diversity at the UGT1 locus isamplified by a novel 31015840 alternative splicing mechanism leadingto nine additional UGT1A proteins that act as regulators ofglucuronidation activityrdquo Pharmacogenetics and Genomics vol17 no 12 pp 1077ndash1089 2007

[34] A Nakamura M Nakajima H Yamanaka R Fujiwara and TYokoi ldquoExpression of UGT1A and UGT2B mRNA in humannormal tissues and various cell linesrdquo Drug Metabolism andDisposition vol 36 no 8 pp 1461ndash1464 2008

[35] D E Harbourt J K Fallon S Ito et al ldquoQuantificationof human uridine-diphosphate glucuronosyl transferase 1Aisoforms in liver intestine and kidney using nanobore liq-uid chromatography-tandem mass spectrometryrdquo AnalyticalChemistry vol 84 no 1 pp 98ndash105 2012

[36] X Zhou L Cao C Jiang et al ldquoPPAR120572-UGT axis activationrepresses intestinal FXR-FGF15 feedback signalling and exacer-bates experimental colitisrdquoNature Communications vol 5 2014

Stem Cells International

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

MEDIATORSINFLAMMATION

of

EndocrinologyInternational Journal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Disease Markers

Hindawiwwwhindawicom Volume 2018

BioMed Research International

OncologyJournal of

Hindawiwwwhindawicom Volume 2013

Hindawiwwwhindawicom Volume 2018

Oxidative Medicine and Cellular Longevity

Hindawiwwwhindawicom Volume 2018

PPAR Research

Hindawi Publishing Corporation httpwwwhindawicom Volume 2013Hindawiwwwhindawicom

The Scientific World Journal

Volume 2018

Immunology ResearchHindawiwwwhindawicom Volume 2018

Journal of

ObesityJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Computational and Mathematical Methods in Medicine

Hindawiwwwhindawicom Volume 2018

Behavioural Neurology

OphthalmologyJournal of

Hindawiwwwhindawicom Volume 2018

Diabetes ResearchJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Research and TreatmentAIDS

Hindawiwwwhindawicom Volume 2018

Gastroenterology Research and Practice

Hindawiwwwhindawicom Volume 2018

Parkinsonrsquos Disease

Evidence-Based Complementary andAlternative Medicine

Volume 2018Hindawiwwwhindawicom

Submit your manuscripts atwwwhindawicom

Page 7: Urinary Elimination of Bile Acid Glucuronides under Severe ...

Canadian Journal of Gastroenterology and Hepatology 7

200 300 400100[LCA] (M)

40

80

120

160

LCA-

3G fo

rmat

ion

(nm

olh

mg

prot

eins

)

LiverKM

B = 487 plusmn 90VGR

C = 171 plusmn 29

Kidney

KIA = 2403 plusmn 535

KMB = 82 plusmn 22

VGRC = 121 plusmn 11

KIA = 2554 plusmn 699

(a)

200 300 400100[LCA] (M)

60

120

180

240

300

LCA-

24G

form

atio

n (n

mol

hm

g pr

otei

ns)

LiverKM

B = 403 plusmn 67VGR

C = 321 plusmn 16

KidneyKM

B = 305 plusmn 56VGR

C = 162 plusmn 08

(b)

100 200 300 400[DCA] (M)

10

20

30

40

50

60

DCA

-3G

form

atio

n(n

mol

hm

g pr

otei

ns)

Liver nD 17 plusmn 03KM

B = 831 plusmn 113VGR

C = 31 plusmn 02

Kidney nD = 17 plusmn 03KM

B = 823 plusmn 136VGR

C = 36 plusmn 03

(c)

20

40

60

80

DCA

-24G

form

atio

n(n

mol

hm

g pr

otei

ns)

200 300 400100[DCA] (M)

Liver nD 12 plusmn 02KM

B = 711 plusmn 121VGR

C = 58 plusmn 05

Kidney nD = 17 plusmn 03KM

B = 582 plusmn 87VGR

C = 38 plusmn 03

(d)Figure 4Kinetic analyses of LCA-3G (a) LCA-24G (b) DCA-3G (c) and DCA-24G (d) formation by microsomes from human liver and kidneyHuman liver (black squares pool of 50 donors) and kidney (white diamonds pool of 8 donors) microsomes (10 120583g) were incubated in thepresence of increasing concentrations (1 to 350120583M) of lithocholic acid (LCA (a) and (b)) or deoxycholic acid (DCA (c) and (d)) for 2 hoursat 37∘C The formation of LCA-3G (a) LCA-24G (b) DCA-3G (c) and DCA-24G (d) was analyzed by LC-MSMS For each panel graphsrepresent the rate of product formation (119910-axis) versus substrate concentration (119909-axis) of two experiments performed in triplicateThe listedkinetic parameters were determined as indicated in ldquoMaterials and Methodsrdquo A119870119868 constant of inhibition expressed as 120583M B119870119872 calculatedMichaelis constant expressed as 120583M C119881max calculated maximal velocity expressed as nmolhmg proteins D119899 Hill coefficient

8 Canadian Journal of Gastroenterology and Hepatology

1750

3500

5250

7000

HD

CA-6

G fo

rmat

ion

(nm

olh

mg

prot

eins

)

200 300 400100[HDCA] (M)

Liver nA 29 plusmn 04KM

B = 551 plusmn 33VGR

C = 5287 plusmn 211

Kidney nA = 27 plusmn 05KM

B = 447 plusmn 38VGR

C = 3084 plusmn 156

(a)

100

200

300

400

500

HD

CA-2

4G fo

rmat

ion

(nm

olh

mg

prot

eins

)

200 300 400100[HDCA] (M)

Liver nA 38 plusmn 11KM

B = 808 plusmn 64VGR

C = 316 plusmn 22

Kidney nA = 23 plusmn 08KM

B = 939 plusmn 184VGR

C = 37 plusmn 05

(b)

800

1600

2400

3200

HCA

-6G

form

atio

n(n

mol

hm

g pr

otei

ns)

200 300 400100[HCA] (M)

LiverKM

B = 1202 plusmn 156VGR

C = 2285 plusmn 134

KidneyKM

B = 960 plusmn 192VGR

C = 2844 plusmn 241

(c)

20

40

60

HCA

-24G

form

atio

n(n

mol

hm

g pr

otei

ns)

200 300 400100[HCA] (M)

LiverKM

B = 617 plusmn 65VGR

C = 49 plusmn 02

KidneyKM

B = 978 plusmn 206VGR

C = 33 plusmn 03

(d)Figure 5 Kinetic analyses of HDCA-6G (a) HDCA-24G (b) HCA-6G (c) and HCA-24G (d) formation by microsomes from human liver andkidney Human liver (black squares pool of 50 donors) and kidney (white diamonds pool of 8 donors) microsomes (10 120583g) were incubatedin the presence of increasing concentrations (1 to 350120583M) of hyodeoxycholic acid (HDCA (a) and (b)) or hyocholic acid (HCA (c) and(d)) for 2 hours at 37∘CThe formation of HDCA-6G (a) HDCA-24G (b) HCA-6G (c) and HCA-24G (d) was analyzed by LC-MSMS Foreach panel graphs represent the rate of product formation (119910-axis) versus substrate concentration (119909-axis) of two experiments performedin triplicate The listed kinetic parameters were determined as indicated in ldquoMaterials and Methodsrdquo A119899 Hill coefficient B119870119872 calculatedMichaelis constant expressed as 120583M C119881max calculated maximal velocity expressed as nmolhmg proteins

Canadian Journal of Gastroenterology and Hepatology 9

Kid

ney

1A3

Kid

ney

Kid

ney

Kid

ney

Live

r

Live

r

Live

r

Live

r

1A4 2B4 2B7UGT

103

104

105

106

107U

GT

tran

scrip

ts (lo

g of

copy

num

ber

g RN

A)

(a)

1A3

2B42B7

2B7

1A

2B

LiverKidneyAnti-UGT kDa

55

55

55

55

55

40Anti-actin

(b)

Live

r

Kidn

ey

ND

5

10

15

20

UG

T1A

3 pr

otei

n le

vels

(pm

olm

g m

icro

som

es)

(c)

Figure 6 Differential expression of the bile acid-conjugating UGT enzymes in the human liver and kidney (a) UGTs 1A3 1A4 2B4 and 2B7mRNA levels in total RNA samples from human kidney (1 pool of 5 donors and 2 individual donors) and liver (8 individual donors) weredetermined through qRT-PCR analyses as described in ldquoMaterials andMethodsrdquo Results are expressed as log10 ofmRNA copy number per 120583gof total RNA Each symbol represents 1 donor with the exception of the white triangle that corresponds to a pool of the 5 kidney donors (b)The UGT protein contents in microsomal preparations (5120583g) from human kidney (pool of 4 female and 4 male donors) and liver (pool of 26male and 24 female donors) were determined after size separation on SDS-PAGE and hybridization with the anti-UGT1A3 anti-UGT2B47anti-UGT2B7 anti-UGT1A and anti-UGT2B antibodies (1 2000 dilution) The housekeeping protein 120573-actin was used to ensure the equalprotein loading in each lane Data presented are representative of two experiments (c) The UGT1A3 protein content in human liver andkidney microsomes was determined using LC-MSMS analyses as previously described [10] Data represent the mean plusmn SD of two differentexperiments performed in triplicate ND not detected

(Figure 6) With the exception of UGT2B4 transcripts thatwere detected only in 1 kidney mRNA sample other UGTmessengers were detected in all kidney and liver prepara-tions (Figure 6(a)) As expected [28 29] their mRNA levelssustained a strong interindividual variability Western blotanalyses evidenced however major differences between liverand kidney (Figure 6(b)) A clear UGT1A3 immunoreactivecomplex was obtained with liver extracts while this UGTprotein was not detected inmicrosomal proteins from kidney(Figure 6(b)) While the UGT2B7 protein was found atsimilar levels in both tissues the use of an anti-UGT2B42B7antibody indicated that the UGT2B4 protein may be moreabundant in liver than in kidney extracts (Figure 6(b))The lack of an accurate and specific anti-UGT1A4 antibody

impaired the quantification of this BA-conjugating UGThowever we were able to further confirm the absence ofthe UGT1A3 protein in kidney microsomes through the useof our previously established LC-MSMS-based proteomicmethod (Figure 6(c)) [10]

4 Discussion

Urinary and circulating levels of BA-Gs detected in thepresent study are similar to previous findings in terms ofboth glucuronide concentrations and species distribution [1314 16] However an important improvement of the presentinvestigations resides in our ability to discriminate betweenether and ester glucuronides of a single BA For example

10 Canadian Journal of Gastroenterology and Hepatology

while glucuronide conjugates of CDCA CA and DCA werepreviously identified as accumulating metabolites in serafrom patients undergoing bile drainage [20] the nature ofthese glucuronides has not been resolved until now Ourresults point out the ester CDCA-3G and DCA-3G andthe acyl CA-24G as the most abundant glucuronide speciesfound in patients with biliary obstruction Because acyl butnot ester glucuronides can be potentially toxic molecules(reviewed in [30]) such information may be critical inanticipating the consequences of their accumulation duringcholestasis

Interestingly serum levels of some of the bile acidglucuronides such as CDCA-3G CA-24G and LCA-3Gwere significantly reduced after biliary stenting while someothers like CDCA-24G LCA-24G and HDCA-6G weresignificantly increased While additional investigations arewarranted to decipher the mechanisms beyond such a glu-curonide species-dependent responsiveness one can spec-ulate that these changes may actually reflect the differen-tial manner in which the UGT enzymes andor the BA-Gtransporters may be altered upon biliary obstruction andafter stenting Therefore it would have been of interest tocompare how these proteins behave in tissues (ie liver andkidney) from cholestatic donors to validate such a hypothesisActually one limitation of the current study is that the liverand kidneymicrosomes used are not fromdonorswith biliaryobstruction When compared to other glucuronide conju-gates CA-24G presented a unique behavior (1) in contrastto other acyl BA-Gs that are only minor components of theurine and serum glucuronide pools CA-24G is the 3rd mostabundant glucuronidated acid in prestenting samples (2) thisspecies is the most spectacularly affected when poststentingversus prestenting sera profiles are compared (3) the serumCA-24GCA ratio sustains the stronger reduction after biliarystenting (4) this MR is also unique in being significantlyreduced in posttreatment urines and (5) only the CA-24GCA ratio exhibits positive serumurine association interms of prestenting and poststenting distribution as wellas treatment response These last observations support ahepatic origin for the formation of CA-24G and the strongCA-24G accumulation in prestenting fluids and its spec-tacular reduction after bile flow restoration suggests thatunder noncholestatic situations this compound formed inliver cells is normally secreted in bile This hypothesis isfurther supported by previous investigations in which theintravenous injections of cholate glucuronide to rats resultedin a rapid and efficient secretion in bile of the unchangedglucuronide conjugate [31] By contrast when the sameprocedure was applied to bile duct ligated animals 95 of theradioactivity was recovered in urine [31] The present studyindicates that similar rerouting of CA-24G elimination tourine may also occur in cholestatic patients since circulatinglevels detected in prestenting cholestatic donors were almost50-fold higher than those previously quantified in samplesfrom noncholestatic volunteers (sim2 nM [11]) Privilegingsecretion from liver cells into the blood for subsequenturinary elimination may actually protect the liver againstthe accumulation of such abundant and potentially toxicglucuronide species during cholestasis In the same vein the

improved formation of other conjugates such as CDCA-3Gand DCA-3G which we recently identified as nontoxic BAs[32] may also participate in the hepatic detoxification oftheir unconjugated CDCA and DCA precursors Consistentwith the above stated hypothesis circulating MRs for these2 conjugates (ie 252 and 64 for CDCA-3G and DCA-3Gresp) are spectacularly increased when compared to thosepreviously found in noncholestatic donors (06 and 01) [11]However the fact that circulating and urinary MRs for thesespecies were not associated and the fact that biliary stentingexerted only minor effects on these ratios in urine are alsoindicative of an extrahepatic origin for urine CDCA- andDCA-3G species at least after bile flow restoration

We next compared the human liver and kidney in termsof bile acid glucuronidation activity and BA-conjugatingUGT expression With 2 exceptions (ie the 6- and 10-fold difference in 119870119872 and 119881max values for LCA-3G andHDCA-24G formations resp) these experiments revealthe remarkable similarity of renal and hepatic bile acidglucuronidation processes and the clear preference of bothtissues in producing HDCA-6 and HCA-6G Such preferencemay actually explain the predominance of these conjugates invarious human fluids as previously described [11] Howeverthe most intriguing observation issuing from these analysesrelates to the UGT1A3 enzyme Indeed not only was thisenzyme detected in kidney extracts only at the mRNA(but not protein) level but also the absence of such anactive enzyme previously identified as the main isoform foracyl BA glucuronidation [8ndash10 12] is inconsistent with theelevated acyl glucuronide production capability of kidneymicrosomes A plausible explanation for the conflictingobservation on UGT1A3 mRNA and protein expression inkidney samples relates to the fact that protein and RNAextracts were from different sources and thus were notpaired It is therefore possible that only donors for mRNAexpress these isoforms in the kidney Meanwhile unlikelysuch a possibility is supported by the controversial nature ofthe UGT1A3 expression in the human kidney [29 33ndash35]Additional investigations are therefore warranted to clarifywhether this BA-conjugating protein is found or not in thehuman kidney

Nevertheless the present study clearly establishes themajor contributions that liver and kidney play in glu-curonidating bile acid during cholestasis Interestingly Zhouand colleagues reported that intestinal glucuronidation alsoplays an important role in controlling the local bile acidhomeostasis and the pathological development of colitis [36]These authors elegantly demonstrated that in the intestinebile acids glucuronidation leads to reduced intracellular levelsof ligands for the nuclear receptor FXR Reduced FXR activa-tion limits the secretion of FGF1519 in the portal circulationwhich in turns leads to an increase of de novo formationof bile acid in the liver [36] Thus in contrast to the liverand kidney where glucuronidation is thought to facilitate theremoval of bile acids the intestinal glucuronidation seems toincrease the delivery of bile acidsHowever it should be notedthat during obstructive cholestasis the delivery of hepaticbile acids to the intestine is compromised and thus onlyminor intestinal glucuronidation may occur

Canadian Journal of Gastroenterology and Hepatology 11

5 Conclusion

In conclusion the present study evidences the major impactthat bile flow restoration exerts on the circulating and urinarybile acid-glucuronide profiles and demonstrates the elevatedcapability of the human kidney to glucuronidate theseendogenous compounds Future investigations are requiredto clarify the mechanism allowing kidney extracts to convertbile acids into acyl glucuronides in the absence of theUGT1A3 protein

Abbreviations

BA Bile acidBO Biliary obstructionCA Cholic acidCDCA Chenodeoxycholic acidDCA Deoxycholic acidFXR Farnesoid X receptorG GlucuronideHDCA Hyodeoxycholic acidHCA Hyocholic acidLCA Lithocholic acidLC-MSMS Liquid chromatography-tandem mass

spectrometryUDPGA UDP-glucuronic acidUGT UDP-glucuronosyltransferaseMRPs Multidrug resistance related proteins

Conflicts of Interest

The authors declare that there are no conflicts of interest andthat there is no pertinent financial arrangement concerningthe work presented here

Acknowledgments

The authors wish to thank Dr Virginie Bocher for criticalreading of the manuscript This study was supported bygrants from theCanadian Institute ofHealth Research (CIHR119331) the Canadian Liver Foundation the Natural Sci-ences and Engineering Research Council of Canada (NSERC402213-2012) and the Canadian Foundation for Innovation(CFI 25712)Martin Perreault is holder of a scholarship fromthe ldquoFonds pour la Recherche en Sante du Quebecrdquo (FRSQ23444) Olivier Barbier is holder of a salary grant fromCIHR (New Investigator MSH95330) Piotr Milkiewiczwas supported by Grants DEC-201101BNZ504216 and201102ANZ500321 from the National Science Centre inPoland

Supplementary Materials

The supplementary materials present details of patient withbiliary obstruction (Supplementary Material 1) and thechanges in the urine profile of bile acid glucuronides and intheir metabolic ratios before and after a biliary stenting pro-cedure in patients with biliary obstruction (SupplementaryMaterial 2) Supplementary Material 1 patients with biliary

obstruction (adapted from [20]) Supplementary Material 2changes in the urine profile of bile acid glucuronides andin their metabolic ratios before and after a biliary stentingprocedure in patients with biliary obstruction (119899 = 12)(Supplementary Materials)

References

[1] D W Russell ldquoThe enzymes regulation and genetics of bileacid synthesisrdquo Annual Review of Biochemistry vol 72 pp 137ndash174 2003

[2] M J Monte J J G Marin A Antelo and J Vazquez-TatoldquoBile acids Chemistry physiology and pathophysiologyrdquoWorldJournal of Gastroenterology vol 15 no 7 pp 804ndash816 2009

[3] A F Hofmann ldquoDetoxification of lithocholic acid a toxicbile-acid relevance to drug hepatotoxicityrdquo Drug MetabolismReviews vol 36 no 3-4 pp 703ndash722 2004

[4] C Pauli-Magnus B Stieger Y Meier G A Kullak-Ublick andP J Meier ldquoEnterohepatic transport of bile salts and genetics ofcholestasisrdquo Journal of Hepatology vol 43 no 2 pp 342ndash3572005

[5] G Paumgartner ldquoPharmacotherapy of cholestatic liver dis-easesrdquo Journal of Digestive Diseases vol 11 no 3 pp 119ndash1252010

[6] M Verreault J Kaeding P Caron et al ldquoRegulation ofendobiotics glucuronidation by ligand-activated transcriptionfactors Physiological function and therapeutic potentialrdquoDrugMetabolism Reviews vol 42 no 1 pp 106ndash118 2010

[7] P I Mackenzie K W Bock B Burchell et al ldquoNomenclatureupdate for the mammalian UDP glycosyltransferase (UGT)gene superfamilyrdquo Pharmacogenetics and Genomics vol 15 no10 pp 677ndash685 2005

[8] M H Court S Hazarika S Krishnaswamy M Finel andJ A Williams ldquoNovel polymorphic human UDP-glucurono-syltransferase 2A3 Cloning functional characterization ofenzyme variants comparative tissue expression and geneinductionrdquoMolecular Pharmacology vol 74 no 3 pp 744ndash7542008

[9] W E Gall G Zawada B Mojarrabi et al ldquoDifferential glu-curonidation of bile acids androgens and estrogens by humanUGT1A3 and 2B7rdquo The Journal of Steroid Biochemistry andMolecular Biology vol 70 no 1ndash3 pp 101ndash108 1999

[10] J Trottier D El Husseini M Perreault et al ldquoThe humanUGT1A3 enzyme conjugates norursodeoxycholic acid into aC23-ester glucuronide in the liverrdquo The Journal of BiologicalChemistry vol 285 no 2 pp 1113ndash1121 2010

[11] J Trottier M Perreault I Rudkowska et al ldquoProfiling serumbile acid glucuronides in humans Gender divergences geneticdeterminants and response to fenofibraterdquo Clinical Pharmacol-ogy ampTherapeutics vol 94 no 4 pp 533ndash543 2013

[12] P Caron J Trottier M Verreault J Belanger J Kaeding andO Barbier ldquoEnzymatic production of bile acid glucuronidesused as analytical standards for liquid chromatography-massspectrometry analysesrdquo Molecular Pharmaceutics vol 3 no 3pp 293ndash302 2006

[13] B Alme and J Sjovall ldquoAnalysis of bile acid glucuronidesin urine identification of 3120572612057212120572-trihydroxy-5120573-cholanoicacidrdquoThe Journal of Steroid Biochemistry andMolecular Biologyvol 13 no 8 pp 907ndash916 1980

[14] A Stiehl R Raedsch G Rudolph P Czygan and S WalkerldquoAnalysis of bile acid glucuronides in urine group separation

12 Canadian Journal of Gastroenterology and Hepatology

on a lipophilic anion exchangerrdquo Clinica Chimica Acta vol 123no 3 pp 275ndash285 1982

[15] A Stiehl R Raedsch G Rudolph U Gundert-Remy and MSenn ldquoBiliary and urinary excretion of sulfated glucuronidatedand tetrahydroxylated bile acids in cirrhotic patientsrdquo Hepatol-ogy vol 5 no 3 pp 492ndash495 1985

[16] H Takikawa T Beppu and Y Seyama ldquoUrinary concentrationsof bile acid glucuronides and sulfates in hepatobiliary diseasesrdquoGastroenterologia Japonica vol 19 no 2 pp 104ndash109 1984

[17] H Wietholtz H Marschall R Reuschenbach H Matern andS Matern ldquoUrinary excretion of bile acid glucosides and glu-curonides in extrahepatic cholestasisrdquo Hepatology vol 13 no4 pp 656ndash662 1991

[18] K M Knights A Rowland and J O Miners ldquoRenal drugmetabolism in humans The potential for drug-endobioticinteractions involving cytochrome P450 (CYP) and UDP-glu-curonosyltransferase (UGT)rdquo British Journal of Clinical Phar-macology vol 76 no 4 pp 587ndash602 2013

[19] S Matern H Matern E H Farthmann and W GerokldquoHepatic and extrahepatic glucuronidation of bile acids in manCharacterization of bile acid uridine 5rsquo-diphosphate-glucu-ronosyltransferase in hepatic renal and intestinalmicrosomesrdquoThe Journal of Clinical Investigation vol 74 no 2 pp 402ndash4101984

[20] J Trottier A Białek P Caron R J Straka P Milkiewicz and OBarbier ldquoProfiling circulating and urinary bile acids in patientswith biliary obstruction before and after biliary stentingrdquo PLoSONE vol 6 no 7 Article ID e22094 2011

[21] M Perreault L Gauthier-Landry J Trottier et al ldquoThe humanUDP-glucuronosyltransferase UGT2A1 and UGT2A2 enzymesare highly active in bile acid glucuronidationrdquoDrugMetabolismand Disposition vol 41 no 9 pp 1616ndash1620 2013

[22] M E Campbell D M Grant T Inaba and W KalowldquoBiotransformation of caffeine paraxanthine theophylline andtheobromine by polycyclic aromatic hydrocarbon-induciblecytochrome(s) P-450 in human liver microsomesrdquo DrugMetabolism and Disposition vol 15 no 2 pp 237ndash249 1987

[23] J Lepine O Bernard M Plante et al ldquoSpecificity andregioselectivity of the conjugation of estradiol estrone andtheir catecholestrogen and methoxyestrogen metabolites byhuman uridine diphospho-glucuronosyltransferases expressedin endometriumrdquo The Journal of Clinical Endocrinology ampMetabolism vol 89 no 10 pp 5222ndash5232 2004

[24] J Thibaudeau J Lepine J Tojcic et al ldquoCharacterizationof common UGT1A8 UGT1A9 and UGT2B7 variants withdifferent capacities to inactivate mutagenic 4-hydroxylatedmetabolites of estradiol and estronerdquo Cancer Research vol 66no 1 pp 125ndash133 2006

[25] J O Miners P I MacKenzie and K M Knights ldquoTheprediction of drug-glucuronidation parameters in humansUDP-glucuronosyltransferase enzyme-selective substrate andinhibitor probes for reaction phenotyping and in vitroin vivoextrapolation of drug clearance and drug-drug interactionpotentialrdquo Drug Metabolism Reviews vol 42 no 1 pp 189ndash2012010

[26] M Verreault K Senekeo-Effenberger J Trottier et al ldquoTheliver X-receptor alpha controls hepatic expression of the humanbile acid-glucuronidating UGT1A3 enzyme in human cells andtransgenic micerdquo Hepatology vol 44 no 2 pp 368ndash378 2006

[27] R G Rutledge ldquoMathematics of quantitative kinetic PCR andthe application of standard curvesrdquo Nucleic Acids Research vol31 no 16 pp 93endash93 2003

[28] T Izukawa M Nakajima R Fujiwara et al ldquoQuantitative anal-ysis of UDP-glucuronosyltransferase (UGT) 1A and UGT2Bexpression levels in human liversrdquo Drug Metabolism and Dis-position vol 37 no 8 pp 1759ndash1768 2009

[29] S Ohno and S Nakajin ldquoDetermination of mRNA expressionof human UDP-glucuronosyltransferases and application forlocalization in various human tissues by real-time reversetranscriptase-polymerase chain reactionrdquoDrugMetabolism andDisposition vol 37 no 1 pp 32ndash40 2009

[30] S L Regan J L Maggs T G Hammond C Lambert D PWilliams and B K Park ldquoAcyl glucuronidesThe good the badand the uglyrdquo Biopharmaceutics amp Drug Disposition vol 31 no7 pp 367ndash395 2010

[31] J M Little M V Chari and R Lester ldquoExcretion of cholateglucuroniderdquo Journal of Lipid Research vol 26 no 5 pp 583ndash592 1985

[32] M Perreault A Białek J Trottier et al ldquoRole of glucuronidationfor hepatic detoxification and urinary elimination of toxic bileacids during biliary obstructionrdquo PLoS ONE vol 8 no 11Article ID e80994 2013

[33] H Girard E Levesque J Bellemare K Journault B Caillierand C Guillemette ldquoGenetic diversity at the UGT1 locus isamplified by a novel 31015840 alternative splicing mechanism leadingto nine additional UGT1A proteins that act as regulators ofglucuronidation activityrdquo Pharmacogenetics and Genomics vol17 no 12 pp 1077ndash1089 2007

[34] A Nakamura M Nakajima H Yamanaka R Fujiwara and TYokoi ldquoExpression of UGT1A and UGT2B mRNA in humannormal tissues and various cell linesrdquo Drug Metabolism andDisposition vol 36 no 8 pp 1461ndash1464 2008

[35] D E Harbourt J K Fallon S Ito et al ldquoQuantificationof human uridine-diphosphate glucuronosyl transferase 1Aisoforms in liver intestine and kidney using nanobore liq-uid chromatography-tandem mass spectrometryrdquo AnalyticalChemistry vol 84 no 1 pp 98ndash105 2012

[36] X Zhou L Cao C Jiang et al ldquoPPAR120572-UGT axis activationrepresses intestinal FXR-FGF15 feedback signalling and exacer-bates experimental colitisrdquoNature Communications vol 5 2014

Stem Cells International

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

MEDIATORSINFLAMMATION

of

EndocrinologyInternational Journal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Disease Markers

Hindawiwwwhindawicom Volume 2018

BioMed Research International

OncologyJournal of

Hindawiwwwhindawicom Volume 2013

Hindawiwwwhindawicom Volume 2018

Oxidative Medicine and Cellular Longevity

Hindawiwwwhindawicom Volume 2018

PPAR Research

Hindawi Publishing Corporation httpwwwhindawicom Volume 2013Hindawiwwwhindawicom

The Scientific World Journal

Volume 2018

Immunology ResearchHindawiwwwhindawicom Volume 2018

Journal of

ObesityJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Computational and Mathematical Methods in Medicine

Hindawiwwwhindawicom Volume 2018

Behavioural Neurology

OphthalmologyJournal of

Hindawiwwwhindawicom Volume 2018

Diabetes ResearchJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Research and TreatmentAIDS

Hindawiwwwhindawicom Volume 2018

Gastroenterology Research and Practice

Hindawiwwwhindawicom Volume 2018

Parkinsonrsquos Disease

Evidence-Based Complementary andAlternative Medicine

Volume 2018Hindawiwwwhindawicom

Submit your manuscripts atwwwhindawicom

Page 8: Urinary Elimination of Bile Acid Glucuronides under Severe ...

8 Canadian Journal of Gastroenterology and Hepatology

1750

3500

5250

7000

HD

CA-6

G fo

rmat

ion

(nm

olh

mg

prot

eins

)

200 300 400100[HDCA] (M)

Liver nA 29 plusmn 04KM

B = 551 plusmn 33VGR

C = 5287 plusmn 211

Kidney nA = 27 plusmn 05KM

B = 447 plusmn 38VGR

C = 3084 plusmn 156

(a)

100

200

300

400

500

HD

CA-2

4G fo

rmat

ion

(nm

olh

mg

prot

eins

)

200 300 400100[HDCA] (M)

Liver nA 38 plusmn 11KM

B = 808 plusmn 64VGR

C = 316 plusmn 22

Kidney nA = 23 plusmn 08KM

B = 939 plusmn 184VGR

C = 37 plusmn 05

(b)

800

1600

2400

3200

HCA

-6G

form

atio

n(n

mol

hm

g pr

otei

ns)

200 300 400100[HCA] (M)

LiverKM

B = 1202 plusmn 156VGR

C = 2285 plusmn 134

KidneyKM

B = 960 plusmn 192VGR

C = 2844 plusmn 241

(c)

20

40

60

HCA

-24G

form

atio

n(n

mol

hm

g pr

otei

ns)

200 300 400100[HCA] (M)

LiverKM

B = 617 plusmn 65VGR

C = 49 plusmn 02

KidneyKM

B = 978 plusmn 206VGR

C = 33 plusmn 03

(d)Figure 5 Kinetic analyses of HDCA-6G (a) HDCA-24G (b) HCA-6G (c) and HCA-24G (d) formation by microsomes from human liver andkidney Human liver (black squares pool of 50 donors) and kidney (white diamonds pool of 8 donors) microsomes (10 120583g) were incubatedin the presence of increasing concentrations (1 to 350120583M) of hyodeoxycholic acid (HDCA (a) and (b)) or hyocholic acid (HCA (c) and(d)) for 2 hours at 37∘CThe formation of HDCA-6G (a) HDCA-24G (b) HCA-6G (c) and HCA-24G (d) was analyzed by LC-MSMS Foreach panel graphs represent the rate of product formation (119910-axis) versus substrate concentration (119909-axis) of two experiments performedin triplicate The listed kinetic parameters were determined as indicated in ldquoMaterials and Methodsrdquo A119899 Hill coefficient B119870119872 calculatedMichaelis constant expressed as 120583M C119881max calculated maximal velocity expressed as nmolhmg proteins

Canadian Journal of Gastroenterology and Hepatology 9

Kid

ney

1A3

Kid

ney

Kid

ney

Kid

ney

Live

r

Live

r

Live

r

Live

r

1A4 2B4 2B7UGT

103

104

105

106

107U

GT

tran

scrip

ts (lo

g of

copy

num

ber

g RN

A)

(a)

1A3

2B42B7

2B7

1A

2B

LiverKidneyAnti-UGT kDa

55

55

55

55

55

40Anti-actin

(b)

Live

r

Kidn

ey

ND

5

10

15

20

UG

T1A

3 pr

otei

n le

vels

(pm

olm

g m

icro

som

es)

(c)

Figure 6 Differential expression of the bile acid-conjugating UGT enzymes in the human liver and kidney (a) UGTs 1A3 1A4 2B4 and 2B7mRNA levels in total RNA samples from human kidney (1 pool of 5 donors and 2 individual donors) and liver (8 individual donors) weredetermined through qRT-PCR analyses as described in ldquoMaterials andMethodsrdquo Results are expressed as log10 ofmRNA copy number per 120583gof total RNA Each symbol represents 1 donor with the exception of the white triangle that corresponds to a pool of the 5 kidney donors (b)The UGT protein contents in microsomal preparations (5120583g) from human kidney (pool of 4 female and 4 male donors) and liver (pool of 26male and 24 female donors) were determined after size separation on SDS-PAGE and hybridization with the anti-UGT1A3 anti-UGT2B47anti-UGT2B7 anti-UGT1A and anti-UGT2B antibodies (1 2000 dilution) The housekeeping protein 120573-actin was used to ensure the equalprotein loading in each lane Data presented are representative of two experiments (c) The UGT1A3 protein content in human liver andkidney microsomes was determined using LC-MSMS analyses as previously described [10] Data represent the mean plusmn SD of two differentexperiments performed in triplicate ND not detected

(Figure 6) With the exception of UGT2B4 transcripts thatwere detected only in 1 kidney mRNA sample other UGTmessengers were detected in all kidney and liver prepara-tions (Figure 6(a)) As expected [28 29] their mRNA levelssustained a strong interindividual variability Western blotanalyses evidenced however major differences between liverand kidney (Figure 6(b)) A clear UGT1A3 immunoreactivecomplex was obtained with liver extracts while this UGTprotein was not detected inmicrosomal proteins from kidney(Figure 6(b)) While the UGT2B7 protein was found atsimilar levels in both tissues the use of an anti-UGT2B42B7antibody indicated that the UGT2B4 protein may be moreabundant in liver than in kidney extracts (Figure 6(b))The lack of an accurate and specific anti-UGT1A4 antibody

impaired the quantification of this BA-conjugating UGThowever we were able to further confirm the absence ofthe UGT1A3 protein in kidney microsomes through the useof our previously established LC-MSMS-based proteomicmethod (Figure 6(c)) [10]

4 Discussion

Urinary and circulating levels of BA-Gs detected in thepresent study are similar to previous findings in terms ofboth glucuronide concentrations and species distribution [1314 16] However an important improvement of the presentinvestigations resides in our ability to discriminate betweenether and ester glucuronides of a single BA For example

10 Canadian Journal of Gastroenterology and Hepatology

while glucuronide conjugates of CDCA CA and DCA werepreviously identified as accumulating metabolites in serafrom patients undergoing bile drainage [20] the nature ofthese glucuronides has not been resolved until now Ourresults point out the ester CDCA-3G and DCA-3G andthe acyl CA-24G as the most abundant glucuronide speciesfound in patients with biliary obstruction Because acyl butnot ester glucuronides can be potentially toxic molecules(reviewed in [30]) such information may be critical inanticipating the consequences of their accumulation duringcholestasis

Interestingly serum levels of some of the bile acidglucuronides such as CDCA-3G CA-24G and LCA-3Gwere significantly reduced after biliary stenting while someothers like CDCA-24G LCA-24G and HDCA-6G weresignificantly increased While additional investigations arewarranted to decipher the mechanisms beyond such a glu-curonide species-dependent responsiveness one can spec-ulate that these changes may actually reflect the differen-tial manner in which the UGT enzymes andor the BA-Gtransporters may be altered upon biliary obstruction andafter stenting Therefore it would have been of interest tocompare how these proteins behave in tissues (ie liver andkidney) from cholestatic donors to validate such a hypothesisActually one limitation of the current study is that the liverand kidneymicrosomes used are not fromdonorswith biliaryobstruction When compared to other glucuronide conju-gates CA-24G presented a unique behavior (1) in contrastto other acyl BA-Gs that are only minor components of theurine and serum glucuronide pools CA-24G is the 3rd mostabundant glucuronidated acid in prestenting samples (2) thisspecies is the most spectacularly affected when poststentingversus prestenting sera profiles are compared (3) the serumCA-24GCA ratio sustains the stronger reduction after biliarystenting (4) this MR is also unique in being significantlyreduced in posttreatment urines and (5) only the CA-24GCA ratio exhibits positive serumurine association interms of prestenting and poststenting distribution as wellas treatment response These last observations support ahepatic origin for the formation of CA-24G and the strongCA-24G accumulation in prestenting fluids and its spec-tacular reduction after bile flow restoration suggests thatunder noncholestatic situations this compound formed inliver cells is normally secreted in bile This hypothesis isfurther supported by previous investigations in which theintravenous injections of cholate glucuronide to rats resultedin a rapid and efficient secretion in bile of the unchangedglucuronide conjugate [31] By contrast when the sameprocedure was applied to bile duct ligated animals 95 of theradioactivity was recovered in urine [31] The present studyindicates that similar rerouting of CA-24G elimination tourine may also occur in cholestatic patients since circulatinglevels detected in prestenting cholestatic donors were almost50-fold higher than those previously quantified in samplesfrom noncholestatic volunteers (sim2 nM [11]) Privilegingsecretion from liver cells into the blood for subsequenturinary elimination may actually protect the liver againstthe accumulation of such abundant and potentially toxicglucuronide species during cholestasis In the same vein the

improved formation of other conjugates such as CDCA-3Gand DCA-3G which we recently identified as nontoxic BAs[32] may also participate in the hepatic detoxification oftheir unconjugated CDCA and DCA precursors Consistentwith the above stated hypothesis circulating MRs for these2 conjugates (ie 252 and 64 for CDCA-3G and DCA-3Gresp) are spectacularly increased when compared to thosepreviously found in noncholestatic donors (06 and 01) [11]However the fact that circulating and urinary MRs for thesespecies were not associated and the fact that biliary stentingexerted only minor effects on these ratios in urine are alsoindicative of an extrahepatic origin for urine CDCA- andDCA-3G species at least after bile flow restoration

We next compared the human liver and kidney in termsof bile acid glucuronidation activity and BA-conjugatingUGT expression With 2 exceptions (ie the 6- and 10-fold difference in 119870119872 and 119881max values for LCA-3G andHDCA-24G formations resp) these experiments revealthe remarkable similarity of renal and hepatic bile acidglucuronidation processes and the clear preference of bothtissues in producing HDCA-6 and HCA-6G Such preferencemay actually explain the predominance of these conjugates invarious human fluids as previously described [11] Howeverthe most intriguing observation issuing from these analysesrelates to the UGT1A3 enzyme Indeed not only was thisenzyme detected in kidney extracts only at the mRNA(but not protein) level but also the absence of such anactive enzyme previously identified as the main isoform foracyl BA glucuronidation [8ndash10 12] is inconsistent with theelevated acyl glucuronide production capability of kidneymicrosomes A plausible explanation for the conflictingobservation on UGT1A3 mRNA and protein expression inkidney samples relates to the fact that protein and RNAextracts were from different sources and thus were notpaired It is therefore possible that only donors for mRNAexpress these isoforms in the kidney Meanwhile unlikelysuch a possibility is supported by the controversial nature ofthe UGT1A3 expression in the human kidney [29 33ndash35]Additional investigations are therefore warranted to clarifywhether this BA-conjugating protein is found or not in thehuman kidney

Nevertheless the present study clearly establishes themajor contributions that liver and kidney play in glu-curonidating bile acid during cholestasis Interestingly Zhouand colleagues reported that intestinal glucuronidation alsoplays an important role in controlling the local bile acidhomeostasis and the pathological development of colitis [36]These authors elegantly demonstrated that in the intestinebile acids glucuronidation leads to reduced intracellular levelsof ligands for the nuclear receptor FXR Reduced FXR activa-tion limits the secretion of FGF1519 in the portal circulationwhich in turns leads to an increase of de novo formationof bile acid in the liver [36] Thus in contrast to the liverand kidney where glucuronidation is thought to facilitate theremoval of bile acids the intestinal glucuronidation seems toincrease the delivery of bile acidsHowever it should be notedthat during obstructive cholestasis the delivery of hepaticbile acids to the intestine is compromised and thus onlyminor intestinal glucuronidation may occur

Canadian Journal of Gastroenterology and Hepatology 11

5 Conclusion

In conclusion the present study evidences the major impactthat bile flow restoration exerts on the circulating and urinarybile acid-glucuronide profiles and demonstrates the elevatedcapability of the human kidney to glucuronidate theseendogenous compounds Future investigations are requiredto clarify the mechanism allowing kidney extracts to convertbile acids into acyl glucuronides in the absence of theUGT1A3 protein

Abbreviations

BA Bile acidBO Biliary obstructionCA Cholic acidCDCA Chenodeoxycholic acidDCA Deoxycholic acidFXR Farnesoid X receptorG GlucuronideHDCA Hyodeoxycholic acidHCA Hyocholic acidLCA Lithocholic acidLC-MSMS Liquid chromatography-tandem mass

spectrometryUDPGA UDP-glucuronic acidUGT UDP-glucuronosyltransferaseMRPs Multidrug resistance related proteins

Conflicts of Interest

The authors declare that there are no conflicts of interest andthat there is no pertinent financial arrangement concerningthe work presented here

Acknowledgments

The authors wish to thank Dr Virginie Bocher for criticalreading of the manuscript This study was supported bygrants from theCanadian Institute ofHealth Research (CIHR119331) the Canadian Liver Foundation the Natural Sci-ences and Engineering Research Council of Canada (NSERC402213-2012) and the Canadian Foundation for Innovation(CFI 25712)Martin Perreault is holder of a scholarship fromthe ldquoFonds pour la Recherche en Sante du Quebecrdquo (FRSQ23444) Olivier Barbier is holder of a salary grant fromCIHR (New Investigator MSH95330) Piotr Milkiewiczwas supported by Grants DEC-201101BNZ504216 and201102ANZ500321 from the National Science Centre inPoland

Supplementary Materials

The supplementary materials present details of patient withbiliary obstruction (Supplementary Material 1) and thechanges in the urine profile of bile acid glucuronides and intheir metabolic ratios before and after a biliary stenting pro-cedure in patients with biliary obstruction (SupplementaryMaterial 2) Supplementary Material 1 patients with biliary

obstruction (adapted from [20]) Supplementary Material 2changes in the urine profile of bile acid glucuronides andin their metabolic ratios before and after a biliary stentingprocedure in patients with biliary obstruction (119899 = 12)(Supplementary Materials)

References

[1] D W Russell ldquoThe enzymes regulation and genetics of bileacid synthesisrdquo Annual Review of Biochemistry vol 72 pp 137ndash174 2003

[2] M J Monte J J G Marin A Antelo and J Vazquez-TatoldquoBile acids Chemistry physiology and pathophysiologyrdquoWorldJournal of Gastroenterology vol 15 no 7 pp 804ndash816 2009

[3] A F Hofmann ldquoDetoxification of lithocholic acid a toxicbile-acid relevance to drug hepatotoxicityrdquo Drug MetabolismReviews vol 36 no 3-4 pp 703ndash722 2004

[4] C Pauli-Magnus B Stieger Y Meier G A Kullak-Ublick andP J Meier ldquoEnterohepatic transport of bile salts and genetics ofcholestasisrdquo Journal of Hepatology vol 43 no 2 pp 342ndash3572005

[5] G Paumgartner ldquoPharmacotherapy of cholestatic liver dis-easesrdquo Journal of Digestive Diseases vol 11 no 3 pp 119ndash1252010

[6] M Verreault J Kaeding P Caron et al ldquoRegulation ofendobiotics glucuronidation by ligand-activated transcriptionfactors Physiological function and therapeutic potentialrdquoDrugMetabolism Reviews vol 42 no 1 pp 106ndash118 2010

[7] P I Mackenzie K W Bock B Burchell et al ldquoNomenclatureupdate for the mammalian UDP glycosyltransferase (UGT)gene superfamilyrdquo Pharmacogenetics and Genomics vol 15 no10 pp 677ndash685 2005

[8] M H Court S Hazarika S Krishnaswamy M Finel andJ A Williams ldquoNovel polymorphic human UDP-glucurono-syltransferase 2A3 Cloning functional characterization ofenzyme variants comparative tissue expression and geneinductionrdquoMolecular Pharmacology vol 74 no 3 pp 744ndash7542008

[9] W E Gall G Zawada B Mojarrabi et al ldquoDifferential glu-curonidation of bile acids androgens and estrogens by humanUGT1A3 and 2B7rdquo The Journal of Steroid Biochemistry andMolecular Biology vol 70 no 1ndash3 pp 101ndash108 1999

[10] J Trottier D El Husseini M Perreault et al ldquoThe humanUGT1A3 enzyme conjugates norursodeoxycholic acid into aC23-ester glucuronide in the liverrdquo The Journal of BiologicalChemistry vol 285 no 2 pp 1113ndash1121 2010

[11] J Trottier M Perreault I Rudkowska et al ldquoProfiling serumbile acid glucuronides in humans Gender divergences geneticdeterminants and response to fenofibraterdquo Clinical Pharmacol-ogy ampTherapeutics vol 94 no 4 pp 533ndash543 2013

[12] P Caron J Trottier M Verreault J Belanger J Kaeding andO Barbier ldquoEnzymatic production of bile acid glucuronidesused as analytical standards for liquid chromatography-massspectrometry analysesrdquo Molecular Pharmaceutics vol 3 no 3pp 293ndash302 2006

[13] B Alme and J Sjovall ldquoAnalysis of bile acid glucuronidesin urine identification of 3120572612057212120572-trihydroxy-5120573-cholanoicacidrdquoThe Journal of Steroid Biochemistry andMolecular Biologyvol 13 no 8 pp 907ndash916 1980

[14] A Stiehl R Raedsch G Rudolph P Czygan and S WalkerldquoAnalysis of bile acid glucuronides in urine group separation

12 Canadian Journal of Gastroenterology and Hepatology

on a lipophilic anion exchangerrdquo Clinica Chimica Acta vol 123no 3 pp 275ndash285 1982

[15] A Stiehl R Raedsch G Rudolph U Gundert-Remy and MSenn ldquoBiliary and urinary excretion of sulfated glucuronidatedand tetrahydroxylated bile acids in cirrhotic patientsrdquo Hepatol-ogy vol 5 no 3 pp 492ndash495 1985

[16] H Takikawa T Beppu and Y Seyama ldquoUrinary concentrationsof bile acid glucuronides and sulfates in hepatobiliary diseasesrdquoGastroenterologia Japonica vol 19 no 2 pp 104ndash109 1984

[17] H Wietholtz H Marschall R Reuschenbach H Matern andS Matern ldquoUrinary excretion of bile acid glucosides and glu-curonides in extrahepatic cholestasisrdquo Hepatology vol 13 no4 pp 656ndash662 1991

[18] K M Knights A Rowland and J O Miners ldquoRenal drugmetabolism in humans The potential for drug-endobioticinteractions involving cytochrome P450 (CYP) and UDP-glu-curonosyltransferase (UGT)rdquo British Journal of Clinical Phar-macology vol 76 no 4 pp 587ndash602 2013

[19] S Matern H Matern E H Farthmann and W GerokldquoHepatic and extrahepatic glucuronidation of bile acids in manCharacterization of bile acid uridine 5rsquo-diphosphate-glucu-ronosyltransferase in hepatic renal and intestinalmicrosomesrdquoThe Journal of Clinical Investigation vol 74 no 2 pp 402ndash4101984

[20] J Trottier A Białek P Caron R J Straka P Milkiewicz and OBarbier ldquoProfiling circulating and urinary bile acids in patientswith biliary obstruction before and after biliary stentingrdquo PLoSONE vol 6 no 7 Article ID e22094 2011

[21] M Perreault L Gauthier-Landry J Trottier et al ldquoThe humanUDP-glucuronosyltransferase UGT2A1 and UGT2A2 enzymesare highly active in bile acid glucuronidationrdquoDrugMetabolismand Disposition vol 41 no 9 pp 1616ndash1620 2013

[22] M E Campbell D M Grant T Inaba and W KalowldquoBiotransformation of caffeine paraxanthine theophylline andtheobromine by polycyclic aromatic hydrocarbon-induciblecytochrome(s) P-450 in human liver microsomesrdquo DrugMetabolism and Disposition vol 15 no 2 pp 237ndash249 1987

[23] J Lepine O Bernard M Plante et al ldquoSpecificity andregioselectivity of the conjugation of estradiol estrone andtheir catecholestrogen and methoxyestrogen metabolites byhuman uridine diphospho-glucuronosyltransferases expressedin endometriumrdquo The Journal of Clinical Endocrinology ampMetabolism vol 89 no 10 pp 5222ndash5232 2004

[24] J Thibaudeau J Lepine J Tojcic et al ldquoCharacterizationof common UGT1A8 UGT1A9 and UGT2B7 variants withdifferent capacities to inactivate mutagenic 4-hydroxylatedmetabolites of estradiol and estronerdquo Cancer Research vol 66no 1 pp 125ndash133 2006

[25] J O Miners P I MacKenzie and K M Knights ldquoTheprediction of drug-glucuronidation parameters in humansUDP-glucuronosyltransferase enzyme-selective substrate andinhibitor probes for reaction phenotyping and in vitroin vivoextrapolation of drug clearance and drug-drug interactionpotentialrdquo Drug Metabolism Reviews vol 42 no 1 pp 189ndash2012010

[26] M Verreault K Senekeo-Effenberger J Trottier et al ldquoTheliver X-receptor alpha controls hepatic expression of the humanbile acid-glucuronidating UGT1A3 enzyme in human cells andtransgenic micerdquo Hepatology vol 44 no 2 pp 368ndash378 2006

[27] R G Rutledge ldquoMathematics of quantitative kinetic PCR andthe application of standard curvesrdquo Nucleic Acids Research vol31 no 16 pp 93endash93 2003

[28] T Izukawa M Nakajima R Fujiwara et al ldquoQuantitative anal-ysis of UDP-glucuronosyltransferase (UGT) 1A and UGT2Bexpression levels in human liversrdquo Drug Metabolism and Dis-position vol 37 no 8 pp 1759ndash1768 2009

[29] S Ohno and S Nakajin ldquoDetermination of mRNA expressionof human UDP-glucuronosyltransferases and application forlocalization in various human tissues by real-time reversetranscriptase-polymerase chain reactionrdquoDrugMetabolism andDisposition vol 37 no 1 pp 32ndash40 2009

[30] S L Regan J L Maggs T G Hammond C Lambert D PWilliams and B K Park ldquoAcyl glucuronidesThe good the badand the uglyrdquo Biopharmaceutics amp Drug Disposition vol 31 no7 pp 367ndash395 2010

[31] J M Little M V Chari and R Lester ldquoExcretion of cholateglucuroniderdquo Journal of Lipid Research vol 26 no 5 pp 583ndash592 1985

[32] M Perreault A Białek J Trottier et al ldquoRole of glucuronidationfor hepatic detoxification and urinary elimination of toxic bileacids during biliary obstructionrdquo PLoS ONE vol 8 no 11Article ID e80994 2013

[33] H Girard E Levesque J Bellemare K Journault B Caillierand C Guillemette ldquoGenetic diversity at the UGT1 locus isamplified by a novel 31015840 alternative splicing mechanism leadingto nine additional UGT1A proteins that act as regulators ofglucuronidation activityrdquo Pharmacogenetics and Genomics vol17 no 12 pp 1077ndash1089 2007

[34] A Nakamura M Nakajima H Yamanaka R Fujiwara and TYokoi ldquoExpression of UGT1A and UGT2B mRNA in humannormal tissues and various cell linesrdquo Drug Metabolism andDisposition vol 36 no 8 pp 1461ndash1464 2008

[35] D E Harbourt J K Fallon S Ito et al ldquoQuantificationof human uridine-diphosphate glucuronosyl transferase 1Aisoforms in liver intestine and kidney using nanobore liq-uid chromatography-tandem mass spectrometryrdquo AnalyticalChemistry vol 84 no 1 pp 98ndash105 2012

[36] X Zhou L Cao C Jiang et al ldquoPPAR120572-UGT axis activationrepresses intestinal FXR-FGF15 feedback signalling and exacer-bates experimental colitisrdquoNature Communications vol 5 2014

Stem Cells International

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

MEDIATORSINFLAMMATION

of

EndocrinologyInternational Journal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Disease Markers

Hindawiwwwhindawicom Volume 2018

BioMed Research International

OncologyJournal of

Hindawiwwwhindawicom Volume 2013

Hindawiwwwhindawicom Volume 2018

Oxidative Medicine and Cellular Longevity

Hindawiwwwhindawicom Volume 2018

PPAR Research

Hindawi Publishing Corporation httpwwwhindawicom Volume 2013Hindawiwwwhindawicom

The Scientific World Journal

Volume 2018

Immunology ResearchHindawiwwwhindawicom Volume 2018

Journal of

ObesityJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Computational and Mathematical Methods in Medicine

Hindawiwwwhindawicom Volume 2018

Behavioural Neurology

OphthalmologyJournal of

Hindawiwwwhindawicom Volume 2018

Diabetes ResearchJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Research and TreatmentAIDS

Hindawiwwwhindawicom Volume 2018

Gastroenterology Research and Practice

Hindawiwwwhindawicom Volume 2018

Parkinsonrsquos Disease

Evidence-Based Complementary andAlternative Medicine

Volume 2018Hindawiwwwhindawicom

Submit your manuscripts atwwwhindawicom

Page 9: Urinary Elimination of Bile Acid Glucuronides under Severe ...

Canadian Journal of Gastroenterology and Hepatology 9

Kid

ney

1A3

Kid

ney

Kid

ney

Kid

ney

Live

r

Live

r

Live

r

Live

r

1A4 2B4 2B7UGT

103

104

105

106

107U

GT

tran

scrip

ts (lo

g of

copy

num

ber

g RN

A)

(a)

1A3

2B42B7

2B7

1A

2B

LiverKidneyAnti-UGT kDa

55

55

55

55

55

40Anti-actin

(b)

Live

r

Kidn

ey

ND

5

10

15

20

UG

T1A

3 pr

otei

n le

vels

(pm

olm

g m

icro

som

es)

(c)

Figure 6 Differential expression of the bile acid-conjugating UGT enzymes in the human liver and kidney (a) UGTs 1A3 1A4 2B4 and 2B7mRNA levels in total RNA samples from human kidney (1 pool of 5 donors and 2 individual donors) and liver (8 individual donors) weredetermined through qRT-PCR analyses as described in ldquoMaterials andMethodsrdquo Results are expressed as log10 ofmRNA copy number per 120583gof total RNA Each symbol represents 1 donor with the exception of the white triangle that corresponds to a pool of the 5 kidney donors (b)The UGT protein contents in microsomal preparations (5120583g) from human kidney (pool of 4 female and 4 male donors) and liver (pool of 26male and 24 female donors) were determined after size separation on SDS-PAGE and hybridization with the anti-UGT1A3 anti-UGT2B47anti-UGT2B7 anti-UGT1A and anti-UGT2B antibodies (1 2000 dilution) The housekeeping protein 120573-actin was used to ensure the equalprotein loading in each lane Data presented are representative of two experiments (c) The UGT1A3 protein content in human liver andkidney microsomes was determined using LC-MSMS analyses as previously described [10] Data represent the mean plusmn SD of two differentexperiments performed in triplicate ND not detected

(Figure 6) With the exception of UGT2B4 transcripts thatwere detected only in 1 kidney mRNA sample other UGTmessengers were detected in all kidney and liver prepara-tions (Figure 6(a)) As expected [28 29] their mRNA levelssustained a strong interindividual variability Western blotanalyses evidenced however major differences between liverand kidney (Figure 6(b)) A clear UGT1A3 immunoreactivecomplex was obtained with liver extracts while this UGTprotein was not detected inmicrosomal proteins from kidney(Figure 6(b)) While the UGT2B7 protein was found atsimilar levels in both tissues the use of an anti-UGT2B42B7antibody indicated that the UGT2B4 protein may be moreabundant in liver than in kidney extracts (Figure 6(b))The lack of an accurate and specific anti-UGT1A4 antibody

impaired the quantification of this BA-conjugating UGThowever we were able to further confirm the absence ofthe UGT1A3 protein in kidney microsomes through the useof our previously established LC-MSMS-based proteomicmethod (Figure 6(c)) [10]

4 Discussion

Urinary and circulating levels of BA-Gs detected in thepresent study are similar to previous findings in terms ofboth glucuronide concentrations and species distribution [1314 16] However an important improvement of the presentinvestigations resides in our ability to discriminate betweenether and ester glucuronides of a single BA For example

10 Canadian Journal of Gastroenterology and Hepatology

while glucuronide conjugates of CDCA CA and DCA werepreviously identified as accumulating metabolites in serafrom patients undergoing bile drainage [20] the nature ofthese glucuronides has not been resolved until now Ourresults point out the ester CDCA-3G and DCA-3G andthe acyl CA-24G as the most abundant glucuronide speciesfound in patients with biliary obstruction Because acyl butnot ester glucuronides can be potentially toxic molecules(reviewed in [30]) such information may be critical inanticipating the consequences of their accumulation duringcholestasis

Interestingly serum levels of some of the bile acidglucuronides such as CDCA-3G CA-24G and LCA-3Gwere significantly reduced after biliary stenting while someothers like CDCA-24G LCA-24G and HDCA-6G weresignificantly increased While additional investigations arewarranted to decipher the mechanisms beyond such a glu-curonide species-dependent responsiveness one can spec-ulate that these changes may actually reflect the differen-tial manner in which the UGT enzymes andor the BA-Gtransporters may be altered upon biliary obstruction andafter stenting Therefore it would have been of interest tocompare how these proteins behave in tissues (ie liver andkidney) from cholestatic donors to validate such a hypothesisActually one limitation of the current study is that the liverand kidneymicrosomes used are not fromdonorswith biliaryobstruction When compared to other glucuronide conju-gates CA-24G presented a unique behavior (1) in contrastto other acyl BA-Gs that are only minor components of theurine and serum glucuronide pools CA-24G is the 3rd mostabundant glucuronidated acid in prestenting samples (2) thisspecies is the most spectacularly affected when poststentingversus prestenting sera profiles are compared (3) the serumCA-24GCA ratio sustains the stronger reduction after biliarystenting (4) this MR is also unique in being significantlyreduced in posttreatment urines and (5) only the CA-24GCA ratio exhibits positive serumurine association interms of prestenting and poststenting distribution as wellas treatment response These last observations support ahepatic origin for the formation of CA-24G and the strongCA-24G accumulation in prestenting fluids and its spec-tacular reduction after bile flow restoration suggests thatunder noncholestatic situations this compound formed inliver cells is normally secreted in bile This hypothesis isfurther supported by previous investigations in which theintravenous injections of cholate glucuronide to rats resultedin a rapid and efficient secretion in bile of the unchangedglucuronide conjugate [31] By contrast when the sameprocedure was applied to bile duct ligated animals 95 of theradioactivity was recovered in urine [31] The present studyindicates that similar rerouting of CA-24G elimination tourine may also occur in cholestatic patients since circulatinglevels detected in prestenting cholestatic donors were almost50-fold higher than those previously quantified in samplesfrom noncholestatic volunteers (sim2 nM [11]) Privilegingsecretion from liver cells into the blood for subsequenturinary elimination may actually protect the liver againstthe accumulation of such abundant and potentially toxicglucuronide species during cholestasis In the same vein the

improved formation of other conjugates such as CDCA-3Gand DCA-3G which we recently identified as nontoxic BAs[32] may also participate in the hepatic detoxification oftheir unconjugated CDCA and DCA precursors Consistentwith the above stated hypothesis circulating MRs for these2 conjugates (ie 252 and 64 for CDCA-3G and DCA-3Gresp) are spectacularly increased when compared to thosepreviously found in noncholestatic donors (06 and 01) [11]However the fact that circulating and urinary MRs for thesespecies were not associated and the fact that biliary stentingexerted only minor effects on these ratios in urine are alsoindicative of an extrahepatic origin for urine CDCA- andDCA-3G species at least after bile flow restoration

We next compared the human liver and kidney in termsof bile acid glucuronidation activity and BA-conjugatingUGT expression With 2 exceptions (ie the 6- and 10-fold difference in 119870119872 and 119881max values for LCA-3G andHDCA-24G formations resp) these experiments revealthe remarkable similarity of renal and hepatic bile acidglucuronidation processes and the clear preference of bothtissues in producing HDCA-6 and HCA-6G Such preferencemay actually explain the predominance of these conjugates invarious human fluids as previously described [11] Howeverthe most intriguing observation issuing from these analysesrelates to the UGT1A3 enzyme Indeed not only was thisenzyme detected in kidney extracts only at the mRNA(but not protein) level but also the absence of such anactive enzyme previously identified as the main isoform foracyl BA glucuronidation [8ndash10 12] is inconsistent with theelevated acyl glucuronide production capability of kidneymicrosomes A plausible explanation for the conflictingobservation on UGT1A3 mRNA and protein expression inkidney samples relates to the fact that protein and RNAextracts were from different sources and thus were notpaired It is therefore possible that only donors for mRNAexpress these isoforms in the kidney Meanwhile unlikelysuch a possibility is supported by the controversial nature ofthe UGT1A3 expression in the human kidney [29 33ndash35]Additional investigations are therefore warranted to clarifywhether this BA-conjugating protein is found or not in thehuman kidney

Nevertheless the present study clearly establishes themajor contributions that liver and kidney play in glu-curonidating bile acid during cholestasis Interestingly Zhouand colleagues reported that intestinal glucuronidation alsoplays an important role in controlling the local bile acidhomeostasis and the pathological development of colitis [36]These authors elegantly demonstrated that in the intestinebile acids glucuronidation leads to reduced intracellular levelsof ligands for the nuclear receptor FXR Reduced FXR activa-tion limits the secretion of FGF1519 in the portal circulationwhich in turns leads to an increase of de novo formationof bile acid in the liver [36] Thus in contrast to the liverand kidney where glucuronidation is thought to facilitate theremoval of bile acids the intestinal glucuronidation seems toincrease the delivery of bile acidsHowever it should be notedthat during obstructive cholestasis the delivery of hepaticbile acids to the intestine is compromised and thus onlyminor intestinal glucuronidation may occur

Canadian Journal of Gastroenterology and Hepatology 11

5 Conclusion

In conclusion the present study evidences the major impactthat bile flow restoration exerts on the circulating and urinarybile acid-glucuronide profiles and demonstrates the elevatedcapability of the human kidney to glucuronidate theseendogenous compounds Future investigations are requiredto clarify the mechanism allowing kidney extracts to convertbile acids into acyl glucuronides in the absence of theUGT1A3 protein

Abbreviations

BA Bile acidBO Biliary obstructionCA Cholic acidCDCA Chenodeoxycholic acidDCA Deoxycholic acidFXR Farnesoid X receptorG GlucuronideHDCA Hyodeoxycholic acidHCA Hyocholic acidLCA Lithocholic acidLC-MSMS Liquid chromatography-tandem mass

spectrometryUDPGA UDP-glucuronic acidUGT UDP-glucuronosyltransferaseMRPs Multidrug resistance related proteins

Conflicts of Interest

The authors declare that there are no conflicts of interest andthat there is no pertinent financial arrangement concerningthe work presented here

Acknowledgments

The authors wish to thank Dr Virginie Bocher for criticalreading of the manuscript This study was supported bygrants from theCanadian Institute ofHealth Research (CIHR119331) the Canadian Liver Foundation the Natural Sci-ences and Engineering Research Council of Canada (NSERC402213-2012) and the Canadian Foundation for Innovation(CFI 25712)Martin Perreault is holder of a scholarship fromthe ldquoFonds pour la Recherche en Sante du Quebecrdquo (FRSQ23444) Olivier Barbier is holder of a salary grant fromCIHR (New Investigator MSH95330) Piotr Milkiewiczwas supported by Grants DEC-201101BNZ504216 and201102ANZ500321 from the National Science Centre inPoland

Supplementary Materials

The supplementary materials present details of patient withbiliary obstruction (Supplementary Material 1) and thechanges in the urine profile of bile acid glucuronides and intheir metabolic ratios before and after a biliary stenting pro-cedure in patients with biliary obstruction (SupplementaryMaterial 2) Supplementary Material 1 patients with biliary

obstruction (adapted from [20]) Supplementary Material 2changes in the urine profile of bile acid glucuronides andin their metabolic ratios before and after a biliary stentingprocedure in patients with biliary obstruction (119899 = 12)(Supplementary Materials)

References

[1] D W Russell ldquoThe enzymes regulation and genetics of bileacid synthesisrdquo Annual Review of Biochemistry vol 72 pp 137ndash174 2003

[2] M J Monte J J G Marin A Antelo and J Vazquez-TatoldquoBile acids Chemistry physiology and pathophysiologyrdquoWorldJournal of Gastroenterology vol 15 no 7 pp 804ndash816 2009

[3] A F Hofmann ldquoDetoxification of lithocholic acid a toxicbile-acid relevance to drug hepatotoxicityrdquo Drug MetabolismReviews vol 36 no 3-4 pp 703ndash722 2004

[4] C Pauli-Magnus B Stieger Y Meier G A Kullak-Ublick andP J Meier ldquoEnterohepatic transport of bile salts and genetics ofcholestasisrdquo Journal of Hepatology vol 43 no 2 pp 342ndash3572005

[5] G Paumgartner ldquoPharmacotherapy of cholestatic liver dis-easesrdquo Journal of Digestive Diseases vol 11 no 3 pp 119ndash1252010

[6] M Verreault J Kaeding P Caron et al ldquoRegulation ofendobiotics glucuronidation by ligand-activated transcriptionfactors Physiological function and therapeutic potentialrdquoDrugMetabolism Reviews vol 42 no 1 pp 106ndash118 2010

[7] P I Mackenzie K W Bock B Burchell et al ldquoNomenclatureupdate for the mammalian UDP glycosyltransferase (UGT)gene superfamilyrdquo Pharmacogenetics and Genomics vol 15 no10 pp 677ndash685 2005

[8] M H Court S Hazarika S Krishnaswamy M Finel andJ A Williams ldquoNovel polymorphic human UDP-glucurono-syltransferase 2A3 Cloning functional characterization ofenzyme variants comparative tissue expression and geneinductionrdquoMolecular Pharmacology vol 74 no 3 pp 744ndash7542008

[9] W E Gall G Zawada B Mojarrabi et al ldquoDifferential glu-curonidation of bile acids androgens and estrogens by humanUGT1A3 and 2B7rdquo The Journal of Steroid Biochemistry andMolecular Biology vol 70 no 1ndash3 pp 101ndash108 1999

[10] J Trottier D El Husseini M Perreault et al ldquoThe humanUGT1A3 enzyme conjugates norursodeoxycholic acid into aC23-ester glucuronide in the liverrdquo The Journal of BiologicalChemistry vol 285 no 2 pp 1113ndash1121 2010

[11] J Trottier M Perreault I Rudkowska et al ldquoProfiling serumbile acid glucuronides in humans Gender divergences geneticdeterminants and response to fenofibraterdquo Clinical Pharmacol-ogy ampTherapeutics vol 94 no 4 pp 533ndash543 2013

[12] P Caron J Trottier M Verreault J Belanger J Kaeding andO Barbier ldquoEnzymatic production of bile acid glucuronidesused as analytical standards for liquid chromatography-massspectrometry analysesrdquo Molecular Pharmaceutics vol 3 no 3pp 293ndash302 2006

[13] B Alme and J Sjovall ldquoAnalysis of bile acid glucuronidesin urine identification of 3120572612057212120572-trihydroxy-5120573-cholanoicacidrdquoThe Journal of Steroid Biochemistry andMolecular Biologyvol 13 no 8 pp 907ndash916 1980

[14] A Stiehl R Raedsch G Rudolph P Czygan and S WalkerldquoAnalysis of bile acid glucuronides in urine group separation

12 Canadian Journal of Gastroenterology and Hepatology

on a lipophilic anion exchangerrdquo Clinica Chimica Acta vol 123no 3 pp 275ndash285 1982

[15] A Stiehl R Raedsch G Rudolph U Gundert-Remy and MSenn ldquoBiliary and urinary excretion of sulfated glucuronidatedand tetrahydroxylated bile acids in cirrhotic patientsrdquo Hepatol-ogy vol 5 no 3 pp 492ndash495 1985

[16] H Takikawa T Beppu and Y Seyama ldquoUrinary concentrationsof bile acid glucuronides and sulfates in hepatobiliary diseasesrdquoGastroenterologia Japonica vol 19 no 2 pp 104ndash109 1984

[17] H Wietholtz H Marschall R Reuschenbach H Matern andS Matern ldquoUrinary excretion of bile acid glucosides and glu-curonides in extrahepatic cholestasisrdquo Hepatology vol 13 no4 pp 656ndash662 1991

[18] K M Knights A Rowland and J O Miners ldquoRenal drugmetabolism in humans The potential for drug-endobioticinteractions involving cytochrome P450 (CYP) and UDP-glu-curonosyltransferase (UGT)rdquo British Journal of Clinical Phar-macology vol 76 no 4 pp 587ndash602 2013

[19] S Matern H Matern E H Farthmann and W GerokldquoHepatic and extrahepatic glucuronidation of bile acids in manCharacterization of bile acid uridine 5rsquo-diphosphate-glucu-ronosyltransferase in hepatic renal and intestinalmicrosomesrdquoThe Journal of Clinical Investigation vol 74 no 2 pp 402ndash4101984

[20] J Trottier A Białek P Caron R J Straka P Milkiewicz and OBarbier ldquoProfiling circulating and urinary bile acids in patientswith biliary obstruction before and after biliary stentingrdquo PLoSONE vol 6 no 7 Article ID e22094 2011

[21] M Perreault L Gauthier-Landry J Trottier et al ldquoThe humanUDP-glucuronosyltransferase UGT2A1 and UGT2A2 enzymesare highly active in bile acid glucuronidationrdquoDrugMetabolismand Disposition vol 41 no 9 pp 1616ndash1620 2013

[22] M E Campbell D M Grant T Inaba and W KalowldquoBiotransformation of caffeine paraxanthine theophylline andtheobromine by polycyclic aromatic hydrocarbon-induciblecytochrome(s) P-450 in human liver microsomesrdquo DrugMetabolism and Disposition vol 15 no 2 pp 237ndash249 1987

[23] J Lepine O Bernard M Plante et al ldquoSpecificity andregioselectivity of the conjugation of estradiol estrone andtheir catecholestrogen and methoxyestrogen metabolites byhuman uridine diphospho-glucuronosyltransferases expressedin endometriumrdquo The Journal of Clinical Endocrinology ampMetabolism vol 89 no 10 pp 5222ndash5232 2004

[24] J Thibaudeau J Lepine J Tojcic et al ldquoCharacterizationof common UGT1A8 UGT1A9 and UGT2B7 variants withdifferent capacities to inactivate mutagenic 4-hydroxylatedmetabolites of estradiol and estronerdquo Cancer Research vol 66no 1 pp 125ndash133 2006

[25] J O Miners P I MacKenzie and K M Knights ldquoTheprediction of drug-glucuronidation parameters in humansUDP-glucuronosyltransferase enzyme-selective substrate andinhibitor probes for reaction phenotyping and in vitroin vivoextrapolation of drug clearance and drug-drug interactionpotentialrdquo Drug Metabolism Reviews vol 42 no 1 pp 189ndash2012010

[26] M Verreault K Senekeo-Effenberger J Trottier et al ldquoTheliver X-receptor alpha controls hepatic expression of the humanbile acid-glucuronidating UGT1A3 enzyme in human cells andtransgenic micerdquo Hepatology vol 44 no 2 pp 368ndash378 2006

[27] R G Rutledge ldquoMathematics of quantitative kinetic PCR andthe application of standard curvesrdquo Nucleic Acids Research vol31 no 16 pp 93endash93 2003

[28] T Izukawa M Nakajima R Fujiwara et al ldquoQuantitative anal-ysis of UDP-glucuronosyltransferase (UGT) 1A and UGT2Bexpression levels in human liversrdquo Drug Metabolism and Dis-position vol 37 no 8 pp 1759ndash1768 2009

[29] S Ohno and S Nakajin ldquoDetermination of mRNA expressionof human UDP-glucuronosyltransferases and application forlocalization in various human tissues by real-time reversetranscriptase-polymerase chain reactionrdquoDrugMetabolism andDisposition vol 37 no 1 pp 32ndash40 2009

[30] S L Regan J L Maggs T G Hammond C Lambert D PWilliams and B K Park ldquoAcyl glucuronidesThe good the badand the uglyrdquo Biopharmaceutics amp Drug Disposition vol 31 no7 pp 367ndash395 2010

[31] J M Little M V Chari and R Lester ldquoExcretion of cholateglucuroniderdquo Journal of Lipid Research vol 26 no 5 pp 583ndash592 1985

[32] M Perreault A Białek J Trottier et al ldquoRole of glucuronidationfor hepatic detoxification and urinary elimination of toxic bileacids during biliary obstructionrdquo PLoS ONE vol 8 no 11Article ID e80994 2013

[33] H Girard E Levesque J Bellemare K Journault B Caillierand C Guillemette ldquoGenetic diversity at the UGT1 locus isamplified by a novel 31015840 alternative splicing mechanism leadingto nine additional UGT1A proteins that act as regulators ofglucuronidation activityrdquo Pharmacogenetics and Genomics vol17 no 12 pp 1077ndash1089 2007

[34] A Nakamura M Nakajima H Yamanaka R Fujiwara and TYokoi ldquoExpression of UGT1A and UGT2B mRNA in humannormal tissues and various cell linesrdquo Drug Metabolism andDisposition vol 36 no 8 pp 1461ndash1464 2008

[35] D E Harbourt J K Fallon S Ito et al ldquoQuantificationof human uridine-diphosphate glucuronosyl transferase 1Aisoforms in liver intestine and kidney using nanobore liq-uid chromatography-tandem mass spectrometryrdquo AnalyticalChemistry vol 84 no 1 pp 98ndash105 2012

[36] X Zhou L Cao C Jiang et al ldquoPPAR120572-UGT axis activationrepresses intestinal FXR-FGF15 feedback signalling and exacer-bates experimental colitisrdquoNature Communications vol 5 2014

Stem Cells International

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

MEDIATORSINFLAMMATION

of

EndocrinologyInternational Journal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Disease Markers

Hindawiwwwhindawicom Volume 2018

BioMed Research International

OncologyJournal of

Hindawiwwwhindawicom Volume 2013

Hindawiwwwhindawicom Volume 2018

Oxidative Medicine and Cellular Longevity

Hindawiwwwhindawicom Volume 2018

PPAR Research

Hindawi Publishing Corporation httpwwwhindawicom Volume 2013Hindawiwwwhindawicom

The Scientific World Journal

Volume 2018

Immunology ResearchHindawiwwwhindawicom Volume 2018

Journal of

ObesityJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Computational and Mathematical Methods in Medicine

Hindawiwwwhindawicom Volume 2018

Behavioural Neurology

OphthalmologyJournal of

Hindawiwwwhindawicom Volume 2018

Diabetes ResearchJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Research and TreatmentAIDS

Hindawiwwwhindawicom Volume 2018

Gastroenterology Research and Practice

Hindawiwwwhindawicom Volume 2018

Parkinsonrsquos Disease

Evidence-Based Complementary andAlternative Medicine

Volume 2018Hindawiwwwhindawicom

Submit your manuscripts atwwwhindawicom

Page 10: Urinary Elimination of Bile Acid Glucuronides under Severe ...

10 Canadian Journal of Gastroenterology and Hepatology

while glucuronide conjugates of CDCA CA and DCA werepreviously identified as accumulating metabolites in serafrom patients undergoing bile drainage [20] the nature ofthese glucuronides has not been resolved until now Ourresults point out the ester CDCA-3G and DCA-3G andthe acyl CA-24G as the most abundant glucuronide speciesfound in patients with biliary obstruction Because acyl butnot ester glucuronides can be potentially toxic molecules(reviewed in [30]) such information may be critical inanticipating the consequences of their accumulation duringcholestasis

Interestingly serum levels of some of the bile acidglucuronides such as CDCA-3G CA-24G and LCA-3Gwere significantly reduced after biliary stenting while someothers like CDCA-24G LCA-24G and HDCA-6G weresignificantly increased While additional investigations arewarranted to decipher the mechanisms beyond such a glu-curonide species-dependent responsiveness one can spec-ulate that these changes may actually reflect the differen-tial manner in which the UGT enzymes andor the BA-Gtransporters may be altered upon biliary obstruction andafter stenting Therefore it would have been of interest tocompare how these proteins behave in tissues (ie liver andkidney) from cholestatic donors to validate such a hypothesisActually one limitation of the current study is that the liverand kidneymicrosomes used are not fromdonorswith biliaryobstruction When compared to other glucuronide conju-gates CA-24G presented a unique behavior (1) in contrastto other acyl BA-Gs that are only minor components of theurine and serum glucuronide pools CA-24G is the 3rd mostabundant glucuronidated acid in prestenting samples (2) thisspecies is the most spectacularly affected when poststentingversus prestenting sera profiles are compared (3) the serumCA-24GCA ratio sustains the stronger reduction after biliarystenting (4) this MR is also unique in being significantlyreduced in posttreatment urines and (5) only the CA-24GCA ratio exhibits positive serumurine association interms of prestenting and poststenting distribution as wellas treatment response These last observations support ahepatic origin for the formation of CA-24G and the strongCA-24G accumulation in prestenting fluids and its spec-tacular reduction after bile flow restoration suggests thatunder noncholestatic situations this compound formed inliver cells is normally secreted in bile This hypothesis isfurther supported by previous investigations in which theintravenous injections of cholate glucuronide to rats resultedin a rapid and efficient secretion in bile of the unchangedglucuronide conjugate [31] By contrast when the sameprocedure was applied to bile duct ligated animals 95 of theradioactivity was recovered in urine [31] The present studyindicates that similar rerouting of CA-24G elimination tourine may also occur in cholestatic patients since circulatinglevels detected in prestenting cholestatic donors were almost50-fold higher than those previously quantified in samplesfrom noncholestatic volunteers (sim2 nM [11]) Privilegingsecretion from liver cells into the blood for subsequenturinary elimination may actually protect the liver againstthe accumulation of such abundant and potentially toxicglucuronide species during cholestasis In the same vein the

improved formation of other conjugates such as CDCA-3Gand DCA-3G which we recently identified as nontoxic BAs[32] may also participate in the hepatic detoxification oftheir unconjugated CDCA and DCA precursors Consistentwith the above stated hypothesis circulating MRs for these2 conjugates (ie 252 and 64 for CDCA-3G and DCA-3Gresp) are spectacularly increased when compared to thosepreviously found in noncholestatic donors (06 and 01) [11]However the fact that circulating and urinary MRs for thesespecies were not associated and the fact that biliary stentingexerted only minor effects on these ratios in urine are alsoindicative of an extrahepatic origin for urine CDCA- andDCA-3G species at least after bile flow restoration

We next compared the human liver and kidney in termsof bile acid glucuronidation activity and BA-conjugatingUGT expression With 2 exceptions (ie the 6- and 10-fold difference in 119870119872 and 119881max values for LCA-3G andHDCA-24G formations resp) these experiments revealthe remarkable similarity of renal and hepatic bile acidglucuronidation processes and the clear preference of bothtissues in producing HDCA-6 and HCA-6G Such preferencemay actually explain the predominance of these conjugates invarious human fluids as previously described [11] Howeverthe most intriguing observation issuing from these analysesrelates to the UGT1A3 enzyme Indeed not only was thisenzyme detected in kidney extracts only at the mRNA(but not protein) level but also the absence of such anactive enzyme previously identified as the main isoform foracyl BA glucuronidation [8ndash10 12] is inconsistent with theelevated acyl glucuronide production capability of kidneymicrosomes A plausible explanation for the conflictingobservation on UGT1A3 mRNA and protein expression inkidney samples relates to the fact that protein and RNAextracts were from different sources and thus were notpaired It is therefore possible that only donors for mRNAexpress these isoforms in the kidney Meanwhile unlikelysuch a possibility is supported by the controversial nature ofthe UGT1A3 expression in the human kidney [29 33ndash35]Additional investigations are therefore warranted to clarifywhether this BA-conjugating protein is found or not in thehuman kidney

Nevertheless the present study clearly establishes themajor contributions that liver and kidney play in glu-curonidating bile acid during cholestasis Interestingly Zhouand colleagues reported that intestinal glucuronidation alsoplays an important role in controlling the local bile acidhomeostasis and the pathological development of colitis [36]These authors elegantly demonstrated that in the intestinebile acids glucuronidation leads to reduced intracellular levelsof ligands for the nuclear receptor FXR Reduced FXR activa-tion limits the secretion of FGF1519 in the portal circulationwhich in turns leads to an increase of de novo formationof bile acid in the liver [36] Thus in contrast to the liverand kidney where glucuronidation is thought to facilitate theremoval of bile acids the intestinal glucuronidation seems toincrease the delivery of bile acidsHowever it should be notedthat during obstructive cholestasis the delivery of hepaticbile acids to the intestine is compromised and thus onlyminor intestinal glucuronidation may occur

Canadian Journal of Gastroenterology and Hepatology 11

5 Conclusion

In conclusion the present study evidences the major impactthat bile flow restoration exerts on the circulating and urinarybile acid-glucuronide profiles and demonstrates the elevatedcapability of the human kidney to glucuronidate theseendogenous compounds Future investigations are requiredto clarify the mechanism allowing kidney extracts to convertbile acids into acyl glucuronides in the absence of theUGT1A3 protein

Abbreviations

BA Bile acidBO Biliary obstructionCA Cholic acidCDCA Chenodeoxycholic acidDCA Deoxycholic acidFXR Farnesoid X receptorG GlucuronideHDCA Hyodeoxycholic acidHCA Hyocholic acidLCA Lithocholic acidLC-MSMS Liquid chromatography-tandem mass

spectrometryUDPGA UDP-glucuronic acidUGT UDP-glucuronosyltransferaseMRPs Multidrug resistance related proteins

Conflicts of Interest

The authors declare that there are no conflicts of interest andthat there is no pertinent financial arrangement concerningthe work presented here

Acknowledgments

The authors wish to thank Dr Virginie Bocher for criticalreading of the manuscript This study was supported bygrants from theCanadian Institute ofHealth Research (CIHR119331) the Canadian Liver Foundation the Natural Sci-ences and Engineering Research Council of Canada (NSERC402213-2012) and the Canadian Foundation for Innovation(CFI 25712)Martin Perreault is holder of a scholarship fromthe ldquoFonds pour la Recherche en Sante du Quebecrdquo (FRSQ23444) Olivier Barbier is holder of a salary grant fromCIHR (New Investigator MSH95330) Piotr Milkiewiczwas supported by Grants DEC-201101BNZ504216 and201102ANZ500321 from the National Science Centre inPoland

Supplementary Materials

The supplementary materials present details of patient withbiliary obstruction (Supplementary Material 1) and thechanges in the urine profile of bile acid glucuronides and intheir metabolic ratios before and after a biliary stenting pro-cedure in patients with biliary obstruction (SupplementaryMaterial 2) Supplementary Material 1 patients with biliary

obstruction (adapted from [20]) Supplementary Material 2changes in the urine profile of bile acid glucuronides andin their metabolic ratios before and after a biliary stentingprocedure in patients with biliary obstruction (119899 = 12)(Supplementary Materials)

References

[1] D W Russell ldquoThe enzymes regulation and genetics of bileacid synthesisrdquo Annual Review of Biochemistry vol 72 pp 137ndash174 2003

[2] M J Monte J J G Marin A Antelo and J Vazquez-TatoldquoBile acids Chemistry physiology and pathophysiologyrdquoWorldJournal of Gastroenterology vol 15 no 7 pp 804ndash816 2009

[3] A F Hofmann ldquoDetoxification of lithocholic acid a toxicbile-acid relevance to drug hepatotoxicityrdquo Drug MetabolismReviews vol 36 no 3-4 pp 703ndash722 2004

[4] C Pauli-Magnus B Stieger Y Meier G A Kullak-Ublick andP J Meier ldquoEnterohepatic transport of bile salts and genetics ofcholestasisrdquo Journal of Hepatology vol 43 no 2 pp 342ndash3572005

[5] G Paumgartner ldquoPharmacotherapy of cholestatic liver dis-easesrdquo Journal of Digestive Diseases vol 11 no 3 pp 119ndash1252010

[6] M Verreault J Kaeding P Caron et al ldquoRegulation ofendobiotics glucuronidation by ligand-activated transcriptionfactors Physiological function and therapeutic potentialrdquoDrugMetabolism Reviews vol 42 no 1 pp 106ndash118 2010

[7] P I Mackenzie K W Bock B Burchell et al ldquoNomenclatureupdate for the mammalian UDP glycosyltransferase (UGT)gene superfamilyrdquo Pharmacogenetics and Genomics vol 15 no10 pp 677ndash685 2005

[8] M H Court S Hazarika S Krishnaswamy M Finel andJ A Williams ldquoNovel polymorphic human UDP-glucurono-syltransferase 2A3 Cloning functional characterization ofenzyme variants comparative tissue expression and geneinductionrdquoMolecular Pharmacology vol 74 no 3 pp 744ndash7542008

[9] W E Gall G Zawada B Mojarrabi et al ldquoDifferential glu-curonidation of bile acids androgens and estrogens by humanUGT1A3 and 2B7rdquo The Journal of Steroid Biochemistry andMolecular Biology vol 70 no 1ndash3 pp 101ndash108 1999

[10] J Trottier D El Husseini M Perreault et al ldquoThe humanUGT1A3 enzyme conjugates norursodeoxycholic acid into aC23-ester glucuronide in the liverrdquo The Journal of BiologicalChemistry vol 285 no 2 pp 1113ndash1121 2010

[11] J Trottier M Perreault I Rudkowska et al ldquoProfiling serumbile acid glucuronides in humans Gender divergences geneticdeterminants and response to fenofibraterdquo Clinical Pharmacol-ogy ampTherapeutics vol 94 no 4 pp 533ndash543 2013

[12] P Caron J Trottier M Verreault J Belanger J Kaeding andO Barbier ldquoEnzymatic production of bile acid glucuronidesused as analytical standards for liquid chromatography-massspectrometry analysesrdquo Molecular Pharmaceutics vol 3 no 3pp 293ndash302 2006

[13] B Alme and J Sjovall ldquoAnalysis of bile acid glucuronidesin urine identification of 3120572612057212120572-trihydroxy-5120573-cholanoicacidrdquoThe Journal of Steroid Biochemistry andMolecular Biologyvol 13 no 8 pp 907ndash916 1980

[14] A Stiehl R Raedsch G Rudolph P Czygan and S WalkerldquoAnalysis of bile acid glucuronides in urine group separation

12 Canadian Journal of Gastroenterology and Hepatology

on a lipophilic anion exchangerrdquo Clinica Chimica Acta vol 123no 3 pp 275ndash285 1982

[15] A Stiehl R Raedsch G Rudolph U Gundert-Remy and MSenn ldquoBiliary and urinary excretion of sulfated glucuronidatedand tetrahydroxylated bile acids in cirrhotic patientsrdquo Hepatol-ogy vol 5 no 3 pp 492ndash495 1985

[16] H Takikawa T Beppu and Y Seyama ldquoUrinary concentrationsof bile acid glucuronides and sulfates in hepatobiliary diseasesrdquoGastroenterologia Japonica vol 19 no 2 pp 104ndash109 1984

[17] H Wietholtz H Marschall R Reuschenbach H Matern andS Matern ldquoUrinary excretion of bile acid glucosides and glu-curonides in extrahepatic cholestasisrdquo Hepatology vol 13 no4 pp 656ndash662 1991

[18] K M Knights A Rowland and J O Miners ldquoRenal drugmetabolism in humans The potential for drug-endobioticinteractions involving cytochrome P450 (CYP) and UDP-glu-curonosyltransferase (UGT)rdquo British Journal of Clinical Phar-macology vol 76 no 4 pp 587ndash602 2013

[19] S Matern H Matern E H Farthmann and W GerokldquoHepatic and extrahepatic glucuronidation of bile acids in manCharacterization of bile acid uridine 5rsquo-diphosphate-glucu-ronosyltransferase in hepatic renal and intestinalmicrosomesrdquoThe Journal of Clinical Investigation vol 74 no 2 pp 402ndash4101984

[20] J Trottier A Białek P Caron R J Straka P Milkiewicz and OBarbier ldquoProfiling circulating and urinary bile acids in patientswith biliary obstruction before and after biliary stentingrdquo PLoSONE vol 6 no 7 Article ID e22094 2011

[21] M Perreault L Gauthier-Landry J Trottier et al ldquoThe humanUDP-glucuronosyltransferase UGT2A1 and UGT2A2 enzymesare highly active in bile acid glucuronidationrdquoDrugMetabolismand Disposition vol 41 no 9 pp 1616ndash1620 2013

[22] M E Campbell D M Grant T Inaba and W KalowldquoBiotransformation of caffeine paraxanthine theophylline andtheobromine by polycyclic aromatic hydrocarbon-induciblecytochrome(s) P-450 in human liver microsomesrdquo DrugMetabolism and Disposition vol 15 no 2 pp 237ndash249 1987

[23] J Lepine O Bernard M Plante et al ldquoSpecificity andregioselectivity of the conjugation of estradiol estrone andtheir catecholestrogen and methoxyestrogen metabolites byhuman uridine diphospho-glucuronosyltransferases expressedin endometriumrdquo The Journal of Clinical Endocrinology ampMetabolism vol 89 no 10 pp 5222ndash5232 2004

[24] J Thibaudeau J Lepine J Tojcic et al ldquoCharacterizationof common UGT1A8 UGT1A9 and UGT2B7 variants withdifferent capacities to inactivate mutagenic 4-hydroxylatedmetabolites of estradiol and estronerdquo Cancer Research vol 66no 1 pp 125ndash133 2006

[25] J O Miners P I MacKenzie and K M Knights ldquoTheprediction of drug-glucuronidation parameters in humansUDP-glucuronosyltransferase enzyme-selective substrate andinhibitor probes for reaction phenotyping and in vitroin vivoextrapolation of drug clearance and drug-drug interactionpotentialrdquo Drug Metabolism Reviews vol 42 no 1 pp 189ndash2012010

[26] M Verreault K Senekeo-Effenberger J Trottier et al ldquoTheliver X-receptor alpha controls hepatic expression of the humanbile acid-glucuronidating UGT1A3 enzyme in human cells andtransgenic micerdquo Hepatology vol 44 no 2 pp 368ndash378 2006

[27] R G Rutledge ldquoMathematics of quantitative kinetic PCR andthe application of standard curvesrdquo Nucleic Acids Research vol31 no 16 pp 93endash93 2003

[28] T Izukawa M Nakajima R Fujiwara et al ldquoQuantitative anal-ysis of UDP-glucuronosyltransferase (UGT) 1A and UGT2Bexpression levels in human liversrdquo Drug Metabolism and Dis-position vol 37 no 8 pp 1759ndash1768 2009

[29] S Ohno and S Nakajin ldquoDetermination of mRNA expressionof human UDP-glucuronosyltransferases and application forlocalization in various human tissues by real-time reversetranscriptase-polymerase chain reactionrdquoDrugMetabolism andDisposition vol 37 no 1 pp 32ndash40 2009

[30] S L Regan J L Maggs T G Hammond C Lambert D PWilliams and B K Park ldquoAcyl glucuronidesThe good the badand the uglyrdquo Biopharmaceutics amp Drug Disposition vol 31 no7 pp 367ndash395 2010

[31] J M Little M V Chari and R Lester ldquoExcretion of cholateglucuroniderdquo Journal of Lipid Research vol 26 no 5 pp 583ndash592 1985

[32] M Perreault A Białek J Trottier et al ldquoRole of glucuronidationfor hepatic detoxification and urinary elimination of toxic bileacids during biliary obstructionrdquo PLoS ONE vol 8 no 11Article ID e80994 2013

[33] H Girard E Levesque J Bellemare K Journault B Caillierand C Guillemette ldquoGenetic diversity at the UGT1 locus isamplified by a novel 31015840 alternative splicing mechanism leadingto nine additional UGT1A proteins that act as regulators ofglucuronidation activityrdquo Pharmacogenetics and Genomics vol17 no 12 pp 1077ndash1089 2007

[34] A Nakamura M Nakajima H Yamanaka R Fujiwara and TYokoi ldquoExpression of UGT1A and UGT2B mRNA in humannormal tissues and various cell linesrdquo Drug Metabolism andDisposition vol 36 no 8 pp 1461ndash1464 2008

[35] D E Harbourt J K Fallon S Ito et al ldquoQuantificationof human uridine-diphosphate glucuronosyl transferase 1Aisoforms in liver intestine and kidney using nanobore liq-uid chromatography-tandem mass spectrometryrdquo AnalyticalChemistry vol 84 no 1 pp 98ndash105 2012

[36] X Zhou L Cao C Jiang et al ldquoPPAR120572-UGT axis activationrepresses intestinal FXR-FGF15 feedback signalling and exacer-bates experimental colitisrdquoNature Communications vol 5 2014

Stem Cells International

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

MEDIATORSINFLAMMATION

of

EndocrinologyInternational Journal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Disease Markers

Hindawiwwwhindawicom Volume 2018

BioMed Research International

OncologyJournal of

Hindawiwwwhindawicom Volume 2013

Hindawiwwwhindawicom Volume 2018

Oxidative Medicine and Cellular Longevity

Hindawiwwwhindawicom Volume 2018

PPAR Research

Hindawi Publishing Corporation httpwwwhindawicom Volume 2013Hindawiwwwhindawicom

The Scientific World Journal

Volume 2018

Immunology ResearchHindawiwwwhindawicom Volume 2018

Journal of

ObesityJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Computational and Mathematical Methods in Medicine

Hindawiwwwhindawicom Volume 2018

Behavioural Neurology

OphthalmologyJournal of

Hindawiwwwhindawicom Volume 2018

Diabetes ResearchJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Research and TreatmentAIDS

Hindawiwwwhindawicom Volume 2018

Gastroenterology Research and Practice

Hindawiwwwhindawicom Volume 2018

Parkinsonrsquos Disease

Evidence-Based Complementary andAlternative Medicine

Volume 2018Hindawiwwwhindawicom

Submit your manuscripts atwwwhindawicom

Page 11: Urinary Elimination of Bile Acid Glucuronides under Severe ...

Canadian Journal of Gastroenterology and Hepatology 11

5 Conclusion

In conclusion the present study evidences the major impactthat bile flow restoration exerts on the circulating and urinarybile acid-glucuronide profiles and demonstrates the elevatedcapability of the human kidney to glucuronidate theseendogenous compounds Future investigations are requiredto clarify the mechanism allowing kidney extracts to convertbile acids into acyl glucuronides in the absence of theUGT1A3 protein

Abbreviations

BA Bile acidBO Biliary obstructionCA Cholic acidCDCA Chenodeoxycholic acidDCA Deoxycholic acidFXR Farnesoid X receptorG GlucuronideHDCA Hyodeoxycholic acidHCA Hyocholic acidLCA Lithocholic acidLC-MSMS Liquid chromatography-tandem mass

spectrometryUDPGA UDP-glucuronic acidUGT UDP-glucuronosyltransferaseMRPs Multidrug resistance related proteins

Conflicts of Interest

The authors declare that there are no conflicts of interest andthat there is no pertinent financial arrangement concerningthe work presented here

Acknowledgments

The authors wish to thank Dr Virginie Bocher for criticalreading of the manuscript This study was supported bygrants from theCanadian Institute ofHealth Research (CIHR119331) the Canadian Liver Foundation the Natural Sci-ences and Engineering Research Council of Canada (NSERC402213-2012) and the Canadian Foundation for Innovation(CFI 25712)Martin Perreault is holder of a scholarship fromthe ldquoFonds pour la Recherche en Sante du Quebecrdquo (FRSQ23444) Olivier Barbier is holder of a salary grant fromCIHR (New Investigator MSH95330) Piotr Milkiewiczwas supported by Grants DEC-201101BNZ504216 and201102ANZ500321 from the National Science Centre inPoland

Supplementary Materials

The supplementary materials present details of patient withbiliary obstruction (Supplementary Material 1) and thechanges in the urine profile of bile acid glucuronides and intheir metabolic ratios before and after a biliary stenting pro-cedure in patients with biliary obstruction (SupplementaryMaterial 2) Supplementary Material 1 patients with biliary

obstruction (adapted from [20]) Supplementary Material 2changes in the urine profile of bile acid glucuronides andin their metabolic ratios before and after a biliary stentingprocedure in patients with biliary obstruction (119899 = 12)(Supplementary Materials)

References

[1] D W Russell ldquoThe enzymes regulation and genetics of bileacid synthesisrdquo Annual Review of Biochemistry vol 72 pp 137ndash174 2003

[2] M J Monte J J G Marin A Antelo and J Vazquez-TatoldquoBile acids Chemistry physiology and pathophysiologyrdquoWorldJournal of Gastroenterology vol 15 no 7 pp 804ndash816 2009

[3] A F Hofmann ldquoDetoxification of lithocholic acid a toxicbile-acid relevance to drug hepatotoxicityrdquo Drug MetabolismReviews vol 36 no 3-4 pp 703ndash722 2004

[4] C Pauli-Magnus B Stieger Y Meier G A Kullak-Ublick andP J Meier ldquoEnterohepatic transport of bile salts and genetics ofcholestasisrdquo Journal of Hepatology vol 43 no 2 pp 342ndash3572005

[5] G Paumgartner ldquoPharmacotherapy of cholestatic liver dis-easesrdquo Journal of Digestive Diseases vol 11 no 3 pp 119ndash1252010

[6] M Verreault J Kaeding P Caron et al ldquoRegulation ofendobiotics glucuronidation by ligand-activated transcriptionfactors Physiological function and therapeutic potentialrdquoDrugMetabolism Reviews vol 42 no 1 pp 106ndash118 2010

[7] P I Mackenzie K W Bock B Burchell et al ldquoNomenclatureupdate for the mammalian UDP glycosyltransferase (UGT)gene superfamilyrdquo Pharmacogenetics and Genomics vol 15 no10 pp 677ndash685 2005

[8] M H Court S Hazarika S Krishnaswamy M Finel andJ A Williams ldquoNovel polymorphic human UDP-glucurono-syltransferase 2A3 Cloning functional characterization ofenzyme variants comparative tissue expression and geneinductionrdquoMolecular Pharmacology vol 74 no 3 pp 744ndash7542008

[9] W E Gall G Zawada B Mojarrabi et al ldquoDifferential glu-curonidation of bile acids androgens and estrogens by humanUGT1A3 and 2B7rdquo The Journal of Steroid Biochemistry andMolecular Biology vol 70 no 1ndash3 pp 101ndash108 1999

[10] J Trottier D El Husseini M Perreault et al ldquoThe humanUGT1A3 enzyme conjugates norursodeoxycholic acid into aC23-ester glucuronide in the liverrdquo The Journal of BiologicalChemistry vol 285 no 2 pp 1113ndash1121 2010

[11] J Trottier M Perreault I Rudkowska et al ldquoProfiling serumbile acid glucuronides in humans Gender divergences geneticdeterminants and response to fenofibraterdquo Clinical Pharmacol-ogy ampTherapeutics vol 94 no 4 pp 533ndash543 2013

[12] P Caron J Trottier M Verreault J Belanger J Kaeding andO Barbier ldquoEnzymatic production of bile acid glucuronidesused as analytical standards for liquid chromatography-massspectrometry analysesrdquo Molecular Pharmaceutics vol 3 no 3pp 293ndash302 2006

[13] B Alme and J Sjovall ldquoAnalysis of bile acid glucuronidesin urine identification of 3120572612057212120572-trihydroxy-5120573-cholanoicacidrdquoThe Journal of Steroid Biochemistry andMolecular Biologyvol 13 no 8 pp 907ndash916 1980

[14] A Stiehl R Raedsch G Rudolph P Czygan and S WalkerldquoAnalysis of bile acid glucuronides in urine group separation

12 Canadian Journal of Gastroenterology and Hepatology

on a lipophilic anion exchangerrdquo Clinica Chimica Acta vol 123no 3 pp 275ndash285 1982

[15] A Stiehl R Raedsch G Rudolph U Gundert-Remy and MSenn ldquoBiliary and urinary excretion of sulfated glucuronidatedand tetrahydroxylated bile acids in cirrhotic patientsrdquo Hepatol-ogy vol 5 no 3 pp 492ndash495 1985

[16] H Takikawa T Beppu and Y Seyama ldquoUrinary concentrationsof bile acid glucuronides and sulfates in hepatobiliary diseasesrdquoGastroenterologia Japonica vol 19 no 2 pp 104ndash109 1984

[17] H Wietholtz H Marschall R Reuschenbach H Matern andS Matern ldquoUrinary excretion of bile acid glucosides and glu-curonides in extrahepatic cholestasisrdquo Hepatology vol 13 no4 pp 656ndash662 1991

[18] K M Knights A Rowland and J O Miners ldquoRenal drugmetabolism in humans The potential for drug-endobioticinteractions involving cytochrome P450 (CYP) and UDP-glu-curonosyltransferase (UGT)rdquo British Journal of Clinical Phar-macology vol 76 no 4 pp 587ndash602 2013

[19] S Matern H Matern E H Farthmann and W GerokldquoHepatic and extrahepatic glucuronidation of bile acids in manCharacterization of bile acid uridine 5rsquo-diphosphate-glucu-ronosyltransferase in hepatic renal and intestinalmicrosomesrdquoThe Journal of Clinical Investigation vol 74 no 2 pp 402ndash4101984

[20] J Trottier A Białek P Caron R J Straka P Milkiewicz and OBarbier ldquoProfiling circulating and urinary bile acids in patientswith biliary obstruction before and after biliary stentingrdquo PLoSONE vol 6 no 7 Article ID e22094 2011

[21] M Perreault L Gauthier-Landry J Trottier et al ldquoThe humanUDP-glucuronosyltransferase UGT2A1 and UGT2A2 enzymesare highly active in bile acid glucuronidationrdquoDrugMetabolismand Disposition vol 41 no 9 pp 1616ndash1620 2013

[22] M E Campbell D M Grant T Inaba and W KalowldquoBiotransformation of caffeine paraxanthine theophylline andtheobromine by polycyclic aromatic hydrocarbon-induciblecytochrome(s) P-450 in human liver microsomesrdquo DrugMetabolism and Disposition vol 15 no 2 pp 237ndash249 1987

[23] J Lepine O Bernard M Plante et al ldquoSpecificity andregioselectivity of the conjugation of estradiol estrone andtheir catecholestrogen and methoxyestrogen metabolites byhuman uridine diphospho-glucuronosyltransferases expressedin endometriumrdquo The Journal of Clinical Endocrinology ampMetabolism vol 89 no 10 pp 5222ndash5232 2004

[24] J Thibaudeau J Lepine J Tojcic et al ldquoCharacterizationof common UGT1A8 UGT1A9 and UGT2B7 variants withdifferent capacities to inactivate mutagenic 4-hydroxylatedmetabolites of estradiol and estronerdquo Cancer Research vol 66no 1 pp 125ndash133 2006

[25] J O Miners P I MacKenzie and K M Knights ldquoTheprediction of drug-glucuronidation parameters in humansUDP-glucuronosyltransferase enzyme-selective substrate andinhibitor probes for reaction phenotyping and in vitroin vivoextrapolation of drug clearance and drug-drug interactionpotentialrdquo Drug Metabolism Reviews vol 42 no 1 pp 189ndash2012010

[26] M Verreault K Senekeo-Effenberger J Trottier et al ldquoTheliver X-receptor alpha controls hepatic expression of the humanbile acid-glucuronidating UGT1A3 enzyme in human cells andtransgenic micerdquo Hepatology vol 44 no 2 pp 368ndash378 2006

[27] R G Rutledge ldquoMathematics of quantitative kinetic PCR andthe application of standard curvesrdquo Nucleic Acids Research vol31 no 16 pp 93endash93 2003

[28] T Izukawa M Nakajima R Fujiwara et al ldquoQuantitative anal-ysis of UDP-glucuronosyltransferase (UGT) 1A and UGT2Bexpression levels in human liversrdquo Drug Metabolism and Dis-position vol 37 no 8 pp 1759ndash1768 2009

[29] S Ohno and S Nakajin ldquoDetermination of mRNA expressionof human UDP-glucuronosyltransferases and application forlocalization in various human tissues by real-time reversetranscriptase-polymerase chain reactionrdquoDrugMetabolism andDisposition vol 37 no 1 pp 32ndash40 2009

[30] S L Regan J L Maggs T G Hammond C Lambert D PWilliams and B K Park ldquoAcyl glucuronidesThe good the badand the uglyrdquo Biopharmaceutics amp Drug Disposition vol 31 no7 pp 367ndash395 2010

[31] J M Little M V Chari and R Lester ldquoExcretion of cholateglucuroniderdquo Journal of Lipid Research vol 26 no 5 pp 583ndash592 1985

[32] M Perreault A Białek J Trottier et al ldquoRole of glucuronidationfor hepatic detoxification and urinary elimination of toxic bileacids during biliary obstructionrdquo PLoS ONE vol 8 no 11Article ID e80994 2013

[33] H Girard E Levesque J Bellemare K Journault B Caillierand C Guillemette ldquoGenetic diversity at the UGT1 locus isamplified by a novel 31015840 alternative splicing mechanism leadingto nine additional UGT1A proteins that act as regulators ofglucuronidation activityrdquo Pharmacogenetics and Genomics vol17 no 12 pp 1077ndash1089 2007

[34] A Nakamura M Nakajima H Yamanaka R Fujiwara and TYokoi ldquoExpression of UGT1A and UGT2B mRNA in humannormal tissues and various cell linesrdquo Drug Metabolism andDisposition vol 36 no 8 pp 1461ndash1464 2008

[35] D E Harbourt J K Fallon S Ito et al ldquoQuantificationof human uridine-diphosphate glucuronosyl transferase 1Aisoforms in liver intestine and kidney using nanobore liq-uid chromatography-tandem mass spectrometryrdquo AnalyticalChemistry vol 84 no 1 pp 98ndash105 2012

[36] X Zhou L Cao C Jiang et al ldquoPPAR120572-UGT axis activationrepresses intestinal FXR-FGF15 feedback signalling and exacer-bates experimental colitisrdquoNature Communications vol 5 2014

Stem Cells International

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

MEDIATORSINFLAMMATION

of

EndocrinologyInternational Journal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Disease Markers

Hindawiwwwhindawicom Volume 2018

BioMed Research International

OncologyJournal of

Hindawiwwwhindawicom Volume 2013

Hindawiwwwhindawicom Volume 2018

Oxidative Medicine and Cellular Longevity

Hindawiwwwhindawicom Volume 2018

PPAR Research

Hindawi Publishing Corporation httpwwwhindawicom Volume 2013Hindawiwwwhindawicom

The Scientific World Journal

Volume 2018

Immunology ResearchHindawiwwwhindawicom Volume 2018

Journal of

ObesityJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Computational and Mathematical Methods in Medicine

Hindawiwwwhindawicom Volume 2018

Behavioural Neurology

OphthalmologyJournal of

Hindawiwwwhindawicom Volume 2018

Diabetes ResearchJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Research and TreatmentAIDS

Hindawiwwwhindawicom Volume 2018

Gastroenterology Research and Practice

Hindawiwwwhindawicom Volume 2018

Parkinsonrsquos Disease

Evidence-Based Complementary andAlternative Medicine

Volume 2018Hindawiwwwhindawicom

Submit your manuscripts atwwwhindawicom

Page 12: Urinary Elimination of Bile Acid Glucuronides under Severe ...

12 Canadian Journal of Gastroenterology and Hepatology

on a lipophilic anion exchangerrdquo Clinica Chimica Acta vol 123no 3 pp 275ndash285 1982

[15] A Stiehl R Raedsch G Rudolph U Gundert-Remy and MSenn ldquoBiliary and urinary excretion of sulfated glucuronidatedand tetrahydroxylated bile acids in cirrhotic patientsrdquo Hepatol-ogy vol 5 no 3 pp 492ndash495 1985

[16] H Takikawa T Beppu and Y Seyama ldquoUrinary concentrationsof bile acid glucuronides and sulfates in hepatobiliary diseasesrdquoGastroenterologia Japonica vol 19 no 2 pp 104ndash109 1984

[17] H Wietholtz H Marschall R Reuschenbach H Matern andS Matern ldquoUrinary excretion of bile acid glucosides and glu-curonides in extrahepatic cholestasisrdquo Hepatology vol 13 no4 pp 656ndash662 1991

[18] K M Knights A Rowland and J O Miners ldquoRenal drugmetabolism in humans The potential for drug-endobioticinteractions involving cytochrome P450 (CYP) and UDP-glu-curonosyltransferase (UGT)rdquo British Journal of Clinical Phar-macology vol 76 no 4 pp 587ndash602 2013

[19] S Matern H Matern E H Farthmann and W GerokldquoHepatic and extrahepatic glucuronidation of bile acids in manCharacterization of bile acid uridine 5rsquo-diphosphate-glucu-ronosyltransferase in hepatic renal and intestinalmicrosomesrdquoThe Journal of Clinical Investigation vol 74 no 2 pp 402ndash4101984

[20] J Trottier A Białek P Caron R J Straka P Milkiewicz and OBarbier ldquoProfiling circulating and urinary bile acids in patientswith biliary obstruction before and after biliary stentingrdquo PLoSONE vol 6 no 7 Article ID e22094 2011

[21] M Perreault L Gauthier-Landry J Trottier et al ldquoThe humanUDP-glucuronosyltransferase UGT2A1 and UGT2A2 enzymesare highly active in bile acid glucuronidationrdquoDrugMetabolismand Disposition vol 41 no 9 pp 1616ndash1620 2013

[22] M E Campbell D M Grant T Inaba and W KalowldquoBiotransformation of caffeine paraxanthine theophylline andtheobromine by polycyclic aromatic hydrocarbon-induciblecytochrome(s) P-450 in human liver microsomesrdquo DrugMetabolism and Disposition vol 15 no 2 pp 237ndash249 1987

[23] J Lepine O Bernard M Plante et al ldquoSpecificity andregioselectivity of the conjugation of estradiol estrone andtheir catecholestrogen and methoxyestrogen metabolites byhuman uridine diphospho-glucuronosyltransferases expressedin endometriumrdquo The Journal of Clinical Endocrinology ampMetabolism vol 89 no 10 pp 5222ndash5232 2004

[24] J Thibaudeau J Lepine J Tojcic et al ldquoCharacterizationof common UGT1A8 UGT1A9 and UGT2B7 variants withdifferent capacities to inactivate mutagenic 4-hydroxylatedmetabolites of estradiol and estronerdquo Cancer Research vol 66no 1 pp 125ndash133 2006

[25] J O Miners P I MacKenzie and K M Knights ldquoTheprediction of drug-glucuronidation parameters in humansUDP-glucuronosyltransferase enzyme-selective substrate andinhibitor probes for reaction phenotyping and in vitroin vivoextrapolation of drug clearance and drug-drug interactionpotentialrdquo Drug Metabolism Reviews vol 42 no 1 pp 189ndash2012010

[26] M Verreault K Senekeo-Effenberger J Trottier et al ldquoTheliver X-receptor alpha controls hepatic expression of the humanbile acid-glucuronidating UGT1A3 enzyme in human cells andtransgenic micerdquo Hepatology vol 44 no 2 pp 368ndash378 2006

[27] R G Rutledge ldquoMathematics of quantitative kinetic PCR andthe application of standard curvesrdquo Nucleic Acids Research vol31 no 16 pp 93endash93 2003

[28] T Izukawa M Nakajima R Fujiwara et al ldquoQuantitative anal-ysis of UDP-glucuronosyltransferase (UGT) 1A and UGT2Bexpression levels in human liversrdquo Drug Metabolism and Dis-position vol 37 no 8 pp 1759ndash1768 2009

[29] S Ohno and S Nakajin ldquoDetermination of mRNA expressionof human UDP-glucuronosyltransferases and application forlocalization in various human tissues by real-time reversetranscriptase-polymerase chain reactionrdquoDrugMetabolism andDisposition vol 37 no 1 pp 32ndash40 2009

[30] S L Regan J L Maggs T G Hammond C Lambert D PWilliams and B K Park ldquoAcyl glucuronidesThe good the badand the uglyrdquo Biopharmaceutics amp Drug Disposition vol 31 no7 pp 367ndash395 2010

[31] J M Little M V Chari and R Lester ldquoExcretion of cholateglucuroniderdquo Journal of Lipid Research vol 26 no 5 pp 583ndash592 1985

[32] M Perreault A Białek J Trottier et al ldquoRole of glucuronidationfor hepatic detoxification and urinary elimination of toxic bileacids during biliary obstructionrdquo PLoS ONE vol 8 no 11Article ID e80994 2013

[33] H Girard E Levesque J Bellemare K Journault B Caillierand C Guillemette ldquoGenetic diversity at the UGT1 locus isamplified by a novel 31015840 alternative splicing mechanism leadingto nine additional UGT1A proteins that act as regulators ofglucuronidation activityrdquo Pharmacogenetics and Genomics vol17 no 12 pp 1077ndash1089 2007

[34] A Nakamura M Nakajima H Yamanaka R Fujiwara and TYokoi ldquoExpression of UGT1A and UGT2B mRNA in humannormal tissues and various cell linesrdquo Drug Metabolism andDisposition vol 36 no 8 pp 1461ndash1464 2008

[35] D E Harbourt J K Fallon S Ito et al ldquoQuantificationof human uridine-diphosphate glucuronosyl transferase 1Aisoforms in liver intestine and kidney using nanobore liq-uid chromatography-tandem mass spectrometryrdquo AnalyticalChemistry vol 84 no 1 pp 98ndash105 2012

[36] X Zhou L Cao C Jiang et al ldquoPPAR120572-UGT axis activationrepresses intestinal FXR-FGF15 feedback signalling and exacer-bates experimental colitisrdquoNature Communications vol 5 2014

Stem Cells International

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

MEDIATORSINFLAMMATION

of

EndocrinologyInternational Journal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Disease Markers

Hindawiwwwhindawicom Volume 2018

BioMed Research International

OncologyJournal of

Hindawiwwwhindawicom Volume 2013

Hindawiwwwhindawicom Volume 2018

Oxidative Medicine and Cellular Longevity

Hindawiwwwhindawicom Volume 2018

PPAR Research

Hindawi Publishing Corporation httpwwwhindawicom Volume 2013Hindawiwwwhindawicom

The Scientific World Journal

Volume 2018

Immunology ResearchHindawiwwwhindawicom Volume 2018

Journal of

ObesityJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Computational and Mathematical Methods in Medicine

Hindawiwwwhindawicom Volume 2018

Behavioural Neurology

OphthalmologyJournal of

Hindawiwwwhindawicom Volume 2018

Diabetes ResearchJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Research and TreatmentAIDS

Hindawiwwwhindawicom Volume 2018

Gastroenterology Research and Practice

Hindawiwwwhindawicom Volume 2018

Parkinsonrsquos Disease

Evidence-Based Complementary andAlternative Medicine

Volume 2018Hindawiwwwhindawicom

Submit your manuscripts atwwwhindawicom

Page 13: Urinary Elimination of Bile Acid Glucuronides under Severe ...

Stem Cells International

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

MEDIATORSINFLAMMATION

of

EndocrinologyInternational Journal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Disease Markers

Hindawiwwwhindawicom Volume 2018

BioMed Research International

OncologyJournal of

Hindawiwwwhindawicom Volume 2013

Hindawiwwwhindawicom Volume 2018

Oxidative Medicine and Cellular Longevity

Hindawiwwwhindawicom Volume 2018

PPAR Research

Hindawi Publishing Corporation httpwwwhindawicom Volume 2013Hindawiwwwhindawicom

The Scientific World Journal

Volume 2018

Immunology ResearchHindawiwwwhindawicom Volume 2018

Journal of

ObesityJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Computational and Mathematical Methods in Medicine

Hindawiwwwhindawicom Volume 2018

Behavioural Neurology

OphthalmologyJournal of

Hindawiwwwhindawicom Volume 2018

Diabetes ResearchJournal of

Hindawiwwwhindawicom Volume 2018

Hindawiwwwhindawicom Volume 2018

Research and TreatmentAIDS

Hindawiwwwhindawicom Volume 2018

Gastroenterology Research and Practice

Hindawiwwwhindawicom Volume 2018

Parkinsonrsquos Disease

Evidence-Based Complementary andAlternative Medicine

Volume 2018Hindawiwwwhindawicom

Submit your manuscripts atwwwhindawicom