You are on page 1of 8

Aquatic Toxicology 109 (2012) 259–266

Contents lists available at SciVerse ScienceDirect

Aquatic Toxicology
journal homepage: www.elsevier.com/locate/aquatox

In vitro inhibition of cytochrome P450-mediated reactions by gemfibrozil,


erythromycin, ciprofloxacin and fluoxetine in fish liver microsomes
Emily M. Smith, Fathima I. Iftikar 1 , Sarah Higgins, Anam Irshad, Racquel Jandoc, Matthew Lee,
Joanna Y. Wilson ∗
Department of Biology, McMaster University, 1280 Main Street West, Hamilton, Ontario, Canada L8S 4K1

a r t i c l e i n f o a b s t r a c t

Article history: Inhibition of mammalian cytochrome P450 enzymes (CYPs) is well characterized; major hepatic CYPs can
Received 9 June 2011 be inhibited by drugs and other environmental contaminants. CYP function and inhibition has not yet been
Received in revised form 22 August 2011 well established in fish yet these studies are important for several reasons. First, such studies will provide
Accepted 25 August 2011
functional information for non-mammalian CYPs. Second, specific inhibitors can be used as a diagnos-
tic tool for studying CYP-mediated reactions. Lastly, pharmaceutical mixtures are found in the aquatic
Keywords:
environment and adverse effects associated with drug–drug interactions, including CYP inhibition by
Fluoxetine
pharmaceuticals may be of concern. Using liver microsomes from untreated and ␤-naphthoflavone (BNF)-
Ciprofloxacin
Gemfibrozil
treated rainbow trout, eight fluorescent CYP-mediated catalytic assays were used to assess in vitro CYP
Erythromycin inhibition by four pharmaceuticals: fluoxetine, ciprofloxacin, gemfibrozil and erythromycin. Expressed
Cytochrome P450 zebrafish CYP1 proteins (CYP1A, CYP1B1, CYP1C1 and CYP1C2) were assessed for inhibition with selected
Enzyme inhibition substrates. All pharmaceuticals decreased the metabolism of a number of substrates. Fluoxetine was
the strongest and most broad inhibitor of CYP-mediated reactions in liver microsomes. Zebrafish CYP1s
were strongly inhibited by erythromycin and fluoxetine. Although the pharmaceuticals are selective CYP
inhibitors in mammals, inhibition across a number of substrates suggests they are broad inhibitors in fish.
These data demonstrate that in vitro hepatic CYP inhibition by pharmaceuticals is possible in fish and the
patterns seen here are different than what would be expected based on CYP inhibition in mammals.
© 2011 Elsevier B.V. All rights reserved.

1. Introduction inhibition of CYP activity. Yet, pharmaceutical metabolism and CYP


inhibition has not been well studied in fish. Studies of CYP inhibition
The presence of pharmaceuticals within the aquatic environ- by environmentally relevant pharmaceuticals in fish will provide
ment was first reported in the mid-1970s (Hignite and Azarnoff, valuable information for assessing the risk of pharmaceuticals to
1977) and has since grown to be a major area of research in envi- aquatic species.
ronmental toxicology. Discharge from sewage treatment plants CYPs from families 1 through 4 are well associated with
has been identified as the primary source of pharmaceuticals to pharmaceutical metabolism and are highly expressed in mam-
the aquatic environment; it has been predicted that levels could malian hepatocytes. More specifically, CYP3A4, CYP2D6, CYP2C9
approach kilograms per year in the near future (Lindberg et al., and CYP1A2 are responsible for 50%, 25%, 15% and 5% of human drug
2005). metabolism, respectively (Hemeryck and Belpaire, 2002). These
In aquatic environments, pharmaceuticals are found in mixtures specific CYP isoforms are among the major hepatic CYPs in mam-
and therefore, drug–drug interactions in aquatic species are of con- mals. A review of the major CYP isoforms, their induction and
cern. Cytochrome P450 enzymes (CYP) are important mediators inhibition in mammals can be found in Martignoni et al. (2006).
of drug–drug interactions in mammals because CYPs are primarily Hepatic expression of multiple CYPs, including isoforms from
responsible for drug metabolism. Drug–drug interactions are clin- families 1–4, has been found in teleost fish (reviewed in Arellano-
ically important in humans and mediated by both induction and Aguilar et al., 2009; Stegeman, 1993). Yet, novel CYP isoforms are
found in fish and assuming homologous function between mam-
malian and actinopterygian CYPs can be problematic (Scornaienchi
et al., 2010a,b; Smith and Wilson, 2010). The evolution of the CYP2
∗ Corresponding author. Tel.: +1 905 525 9140x20075; fax: +1 905 522 6066.
gene family is quite different between mammals and fish; subfam-
E-mail address: joanna.wilson@mcmaster.ca (J.Y. Wilson).
1
Present address: School of Biological Sciences, University of Auckland, New ilies are typically distinct across the vertebrates and neither CYP2C
Zealand. nor CYP2D homologs exist in fish (Kirischian et al., 2011). Thus,

0166-445X/$ – see front matter © 2011 Elsevier B.V. All rights reserved.
doi:10.1016/j.aquatox.2011.08.022
260 E.M. Smith et al. / Aquatic Toxicology 109 (2012) 259–266

≥40% of human drugs must be metabolized by non-orthologous diluted in assay buffer just prior to use in assays. CYP inhibition
CYPs in fish. was determined using fluorescent-based catalytic activity assays
Laville et al. (2004) provided the first study of CYP inhibition by for eight substrates with inhibitor concentrations of 0, 10 and
pharmaceuticals using fish hepatocytes. Testing a broad range of 100 ␮M. The specificity of these substrates is known for mam-
drug classes in vitro, 24 h exposure to fibrates and anti-depressants, malian CYPs but not for non-mammalian CYPs. The substrates
in particular, caused potent inhibition of ethoxyresorufin-O- were chosen based on the catalytic capacity of unexposed and BNF
dealkylase (EROD), an indicator of CYP1 activity. Enrofloxacin induced trout liver for these substrates (Smith and Wilson, 2010).
inhibited a number of monooxygenase reactions, both in vivo and Alkoxyresorufin-O-dealkylase (AROD) activities were determined
in vitro in sea bass, Dicentrarchus labrax (Vaccaro et al., 2003). using ethoxyresorufin (ER), methoxyresorufin (MR), benzyloxyre-
Gemfibrozil and fluoxetine have been reported to cause both inhi- sorufin (BR) and pentoxyresorufin (PR; Sigma–Aldrich, St. Louis,
bition (Laville et al., 2004; Thibaut et al., 2006) and induction MO) according to previously published methods (Hahn et al., 1993).
of EROD activity (Thibaut and Porte, 2008). Inhibition of 7- ER, MR, BR and PR were run at substrate concentrations of 2, 2,
benzyloxy-4-trifluoromethyl-coumarin (BFC)-O-debenzyloxidase 3, and 7 ␮M, respectively. Benzyloxy-4-trifluoromethylcoumarin
has been reported by fluoxetine and gemfibrozil in carp liver micro- (BFC), methoxy-4-trifluoromethylcoumarin (MFC), and benzy-
somes (Thibaut et al., 2006). Erythromycin inhibited testosterone loxyquinoline (BQ; BD Biosciences, San Jose, CA), were used in
6␤-hydroxylation in intestinal microsomes from channel catfish assays optimized for fish (Smith and Wilson, 2010), based on meth-
(James et al., 2005). Collectively, these data suggest that pharma- ods published for mammalian microsomes (Crespi et al., 1997;
ceuticals that inhibit CYP mediated reactions in mammals are likely Miller et al., 2000; Stresser et al., 2000). Assay conditions and lim-
teleost CYP inhibitors. its of detection (LODs) were as previously published (Smith and
In this study, we assess the inhibition potential of four pharma- Wilson, 2010) with the exception of BFC and BQ. Assays with BFC
ceuticals (erythromycin, ciprofloxacin, gemfibrozil and fluoxetine) and BQ were modified from Smith and Wilson (2010) with the addi-
in rainbow trout liver microsomes and expressed zebrafish CYP1s tion of bovine serum albumin (1.1 mg mL−1 final concentration) and
using a panel of fluorescent CYP assays. These compounds have lower final substrate concentrations (100 ␮M BQ; 500 ␮M BFC). All
been selected because they are potent inhibitors of the domi- reactions were run using kinetic assays at room temperature for
nant CYPs responsible for pharmaceutical metabolism in mammals. 10 min in black 96-well plates and normalized for total protein con-
These selected compounds have been detected in sewage treat- tent. Between 4 and 10 replicate liver microsome samples were
ment effluent and surface waters (Corcoran et al., 2010; Daughton assayed for each substrate.
and Ternes, 1999; Hirsch et al., 1999; Kolpin et al., 2002; Metcalfe Pharmaceutical inhibition of ER, MR and BR metabolism was
et al., 2003) and are environmentally relevant pharmaceuticals. investigated using heterologously expressed CYP1 proteins from
This data will be useful for assessing the potential for drug–drug zebrafish (Danio rerio). Expressed CYP1A, 1B1, 1C1 and 1C2
interactions in aquatic species and for understanding important (Scornaienchi et al., 2010a) were used in catalytic assays with
differences between mammalian and fish CYP function. 0, 1, 10 and 100 ␮M ciprofloxacin, gemfibrozil and fluoxetine;
compounds with inhibition were assessed for inhibition kinetics.
2. Methods Reactions with expressed CYP proteins were normalized for total
CYP content, based on CO-reduction assays (Omura and Sato, 1964).
2.1. Animals A minimum of triplicate measures were taken for each expressed
protein at each concentration of inhibitor.
Juvenile rainbow trout (Oncorhynchus mykiss) were purchased
from Humber Springs trout hatchery (Mono Mills, ON, Canada).
2.4. Data and statistical analysis
Trout were held at 12–15 ◦ C in flow-through dechlorinated city
water and fed floating trout pellets 3 times per week. All fish were
Inhibition of CYP mediated reactions were determined using
held for a minimum of two weeks before use. Feeding was sus-
one-way repeated measures (RM) Analysis of Variance (ANOVA) on
pended 24 h before treatment.
log-transformed enzyme activity (pmol mg−1 min−1 ) at 0, 10 and
100 ␮M inhibitor concentrations, except for MFC where one-way
2.2. In vivo CYP induction
ANOVA was used. If log-transformation did not normalize the data,
Friedman repeated measures Analysis of Variance on ranks was
␤-Naphthoflavone (BNF) was purchased from Sigma–Aldrich
performed (BFC – both control and BNF fish, BQ – BNF fish only). All
(St. Louis, MO, USA). Trout were exposed to 50 mg kg−1 body
post hoc tests were completed using the Student–Newman–Keuls
weight of BNF dissolved in corn oil and administered via i.p. injec-
method. Statistical tests were completed using Sigmaplot 11.0 (Sys-
tion, under anesthetic (100 mg L−1 methanesulfonate salt, MS-222).
tat Software Inc.).
Injection volume did not exceeding 10 ␮L g−1 total body weight and
Inhibition of EROD, MROD and BROD activity with expressed
control fish were injected with corn oil carrier alone. BNF is a proto-
zebrafish CYP1s were assessed using GraphPad Prism version 5.0
typical CYP1A inducer and has been shown to induce many CYP
for Windows (GraphPad Software Inc., San Diego, CA). These assays
mediated reactions in rainbow trout liver at this dose (Smith and
were run with single preparations of expressed proteins (CYP1A,
Wilson, 2010). All fish were sacrificed 48-h after injection and liv-
1B1, 1C1, 1C2) run in triplicate with 1 ␮M, 10 ␮M and 100 ␮M
ers were collected and flash-frozen in liquid nitrogen. Tissues were
inhibitor. The inhibitor concentration at which 50% activity is
stored at −80 ◦ C until use. Liver microsomes were prepared as in
achieved (IC50) was calculated using the non-linear enzyme inhi-
Stegeman et al. (1995) and total protein content was determined
bition function (model: y = 100/(1 + 10(X-LogIC50) ) for normalized
using a bicinchoninic acid (BCA) kit, according to manufacturer’s
response) within the GraphPad Software.
protocols (Pierce protein research products, Rockford, IL, USA).

2.3. In vitro inhibition of catalytic activity 3. Results

Ciprofloxacin, gemfibrozil and fluoxetine were dissolved in The inhibition potential for four pharmaceuticals was deter-
DMSO and erythromycin was dissolved in methanol, to concen- mined for CYP mediated reactions in hepatic trout microsomes,
trated stock solutions (Sigma–Aldrich, St. Louis, MO). Stocks were from undosed and BNF-treated animals. CYP mediated metabolism
E.M. Smith et al. / Aquatic Toxicology 109 (2012) 259–266 261

Fig. 2. EROD inhibition by pharmaceuticals in un-treated and BNF exposed trout.


EROD activity is shown without inhibitor (baseline) and in the presence of
ciprofloxacin (cipro), erythromycin (erythro), and gemfibrozil (gem). Those bars
marked with an (*) are statistically different from baseline activities, n = 10.

inhibited 45% by 100 ␮M fluoxetine in untreated fish assays and


only 14% in BNF-treated microsomes (Fig. 1).
Ciprofloxacin, erythromycin and gemfibrozil showed more
modest inhibition, both in terms of degree of inhibition and number
of substrates inhibited (Figs. 2–4). BFC metabolism was inhibited by
all inhibitors tested, regardless whether untreated or BNF induced
trout microsomes were used to catalyze the reaction (Figs. 1 and 4).
In control fish, EROD was inhibited by 100 ␮M, but not 10 ␮M,
ciprofloxacin and gemfibrozil (40% and 45% reduction in activity,
respectively; Fig. 2). EROD assays with BNF-induced trout were
more susceptible to inhibition; EROD was inhibited with 100 ␮M
ciprofloxacin and erythromycin and both 10 and 100 ␮M gemfi-
Fig. 1. Inhibition of CYP mediated metabolism by fluoxetine. In vitro inhibition of brozil (Fig. 2).
microsomal reactions with (A) un-treated and (B) BNF-exposed fish. Baseline data
Contrary to BCF metabolism, which was widely inhibited, BQ
represents the level of activity without fluoxetine added. Those bars marked with
an (*) are statistically different from baseline activities, n = 10. metabolism was only inhibited by 100 ␮M ciprofloxacin, gem-
fibrozil and fluoxetine (Figs. 1 and 4). These reductions in BQ
metabolism were evident in both control and BNF-fish; in all cases,
was measured in fluorescent-based assays with ethoxyresorufin assays with microsomes from control fish showed 2–3 times the
(ER), methoxyresorufin (MR), benzyloxyresorufin (BR), pen- level of inhibition compared to BNF-treated fish (Figs. 1 and 4).
toxyresorufin (PR), benzyloxy-4-trifluoromethylcoumarin (BFC), No inhibitor showed an affect on BQ metabolism at 10 ␮M
methoxy-4-trifluoromethylcoumarin (MFC), or benzyloxyquino- concentration; erythromycin did not inhibit BQ metabolism at
line (BQ). The inhibitory effects of each pharmaceutical for either concentration tested (Fig. 4). BR metabolism, at least
CYP1 proteins was tested with bacterially expressed zebrafish
CYP 1 proteins (CYP1A, CYP1B1, CYP1C1 or CYP1C2) using the
resorufin-based substrates only.

3.1. Inhibition of CYP activity in trout liver microsomes

All compounds tested showed inhibition of a number of sub-


strates. Of these, fluoxetine showed the broadest inhibition across
substrates (Fig. 1). Fluoxetine (100 ␮M) was capable of inhibiting
the metabolism of all substrates catalyzed by both untreated and
BNF induced trout liver microsomes. The largest inhibitions by flu-
oxetine were seen at 100 ␮M fluoxetine with BFC (untreated liver
microsomes, 84% reduction in activity), MROD and BROD (BNF-
treated microsomes, both 82% inhibition) and MFC (control and BNF
microsomes, 80–81% inhibition; Fig. 1). The lower concentration
of fluoxetine (10 ␮M) caused significant inhibition (35–58% inhi-
bition) of all but MFC and BQ metabolism (Fig. 1). In general, the
BNF-treated microsomes showed similar or higher percent inhi-
bition compared to control microsomes at the same fluoxetine
Fig. 3. BFC inhibition by pharmaceuticals in un-treated and BNF exposed trout. BFC
concentration; BFC and BQ were both exceptions to this pattern.
activity is shown without inhibitor (baseline) and in the presence of ciprofloxacin
In reactions with BFC, higher inhibition was seen in untreated fish (cipro), erythromycin (erythro), and gemfibrozil (gem). Those bars marked with an
at both fluoxetine concentrations. Similarly, BQ metabolism was (*) are statistically different from baseline activities, n = 10.
262 E.M. Smith et al. / Aquatic Toxicology 109 (2012) 259–266

erythromycin for heterologously expressed zebrafish CYP1A, 1B1,


1C1 and 1C2; these substrates were metabolized well by zebrafish
CYP1s (Scornaienchi et al., 2010a,b). Contrary to inhibition of
trout liver microsomes, erythromycin showed potent inhibition
of expressed CYP1s (Figs. 5–7). Erythromycin (100 ␮M) inhib-
ited CYP 1A, 1B1, 1C1 and 1C2-mediated EROD by 75%, 79%, 57%
and 41%, respectively (Fig. 5). CYP1A-mediated EROD activity was
inhibited by all compounds except gemfibrozil (Fig. 5a), although
ciprofloxacin reduced activity by only 10% at the high concentra-
tion. The highest concentration (100 ␮M) of fluoxetine showed
strong inhibition of CYP1A (60% reduction in activity, Fig. 5a).
The addition of ciprofloxacin to CYP1C1 and 1C2-mediated EROD
assays caused a concentration-dependent increase in activity that
was similar for both proteins (13–35% increase; data not shown).
IC50s for ciprofloxacin and gemfibrozil inhibition of EROD activ-
ity were relatively high compared to fluoxetine and erythromycin
(Table 1).
Fig. 4. BQ inhibition by pharmaceuticals in un-treated and BNF exposed trout. BQ Inhibition of the MROD assay was seen for most inhibitors,
activity is shown without inhibitor (baseline) and in the presence of ciprofloxacin except gemfibrozil, with the CYP1 proteins (Fig. 6a and b). Gem-
(cipro), erythromycin (erythro), and gemfibrozil (gem). Those bars marked with an
(*) are statistically different from baseline activities, n = 10.
fibrozil was only able to inhibit CYP1C1 and 1C2-mediated MROD
at 100 ␮M (Fig. 6c and d). CYP 1C1 and 1C2-mediated MROD
showed amongst the highest levels of inhibition; 94% inhibition
in liver microsomes from BNF treated trout, was inhibited by was seen with 100 ␮M erythromycin for both proteins (Fig. 6c and
ciprofloxacin, erythromycin and gemfibrozil at both concentra- d). IC50s for erythromycin inhibition of MROD activity were con-
tions; MR metabolism in trout liver microsomes from BNF treated sistently lower than the other inhibitors for all expressed CYP1s
trout, was inhibited at the high concentration only (Supplementary (Table 1).
Table 1). Inhibition of CYP1-mediated BROD activity was fairly limited.
Surprisingly, ciprofloxacin, gemfibrozil and fluoxetine signifi- Only CYP1C1-mediated BROD showed a reduction in activity with
cantly elevated catalytic activity compared to baseline for several all compounds (Fig. 7c). 100 ␮M erythromycin was the most effec-
substrates. PROD, in particular, showed significantly higher rates tive inhibitor for CYP1A and 1B1-mediated BROD activity (75%
of metabolism in hepatic microsomes from BNF treated fish with and 95% reduction, respectively); fluoxetine was the most effec-
the addition of all three pharmaceuticals (Supplementary Table tive inhibitor for CYP1C1 and 1C2-mediated BROD (42% and 63%
1). Erythromycin increased the metabolism of MFC in assays with reduction, respectively; Fig. 7). Ciprofloxacin caused increased
microsomes from BNF treated fish (Supplementary Table 1). metabolism in CYP 1A, 1B1 and 1C2-mediated BROD (data not
shown). Although erythromycin caused inhibition for CYP 1A, 1B1
3.2. Inhibition of AROD activity in expressed CYP1s and 1C1-mediated BROD (Fig. 7), CYP1C2 activity was increased to
over twice the baseline metabolism with erythromycin (1 ␮M and
Three substrates (ER, MR and BR) were selected to test the 10 ␮M), the highest activation seen across all substrates tested with
inhibition potential of fluoxetine, gemfibrozil, ciprofloxacin and expressed CYP1s (data not shown).

Fig. 5. Inhibition of EROD activity mediated by heterologously expressed zebrafish CYP1s. Data are shown for (A) CYP1A, (B) CYP1B1, (C) CYP1C1, and (D) CYP1C2 as log
[Inhibitor, ␮M] versus % Activity. Pharmaceutical inhibitors were ciprofloxacin (CIPRO), erythromycin (ERYTH), fluoxetine (FLX), and gemfibrozil (GEM).
E.M. Smith et al. / Aquatic Toxicology 109 (2012) 259–266 263

Fig. 6. Inhibition of MROD activity mediated by heterologously expressed zebrafish CYP1s. Data are shown for (A) CYP1A, (B) CYP1B1, (C) CYP1C1, and (D) CYP1C2 as log
[Inhibitor, ␮M] versus % Activity. Pharmaceutical inhibitors were ciprofloxacin (CIPRO), erythromycin (ERYTH), fluoxetine (FLX), and gemfibrozil (GEM).

Fig. 7. Inhibition of BROD activity mediated by heterologously expressed zebrafish CYP1s. Data are shown for (A) CYP1A, (B) CYP1B1, (C) CYP1C1, and (D) CYP1C2 as log
[Inhibitor, ␮M] versus % Activity. Pharmaceutical inhibitors were ciprofloxacin (CIPRO), erythromycin (ERYTH), fluoxetine (FLX), and gemfibrozil (GEM).

Table 1
IC50s (␮M) for inhibition of CYP1 mediated metabolism of resorufin-based substrates. IC50s were calculated through nonlinear enzyme inhibition regression plots. CYP1
proteins were heterologously expressed zebrafish proteins (see Section 2 for details).

EROD MROD BROD

CYP1A CYP1B1 CYP1C1 CYP1C2 CYP1A CYP1B1 CYP1C1 CYP1C2 CYP1A CYP1B1 CYP1C1 CYP1C2

Fluoxetine 75 475 1000 180 115 300 30 140 200 280 125 70
Ciprofloxacin 850 >1000 n/a n/a 900 680 >1000 410 n/a n/a 180 n/a
Erythromycin 40 40 90 145 5 25 15 25 60 50 275 n/a
Gemfibrozil >1000 >2000 n/a n/a >1000 96 350 330 n/a n/a 170 n/a

n/a, reactions do not fit inhibition model and therefore IC50 could not be calculated.
264 E.M. Smith et al. / Aquatic Toxicology 109 (2012) 259–266

4. Discussion present study, lower concentrations of gemfibrozil elicit inhibi-


tion of EROD, with BNF-trout microsomes susceptible at the lowest
The inhibitors chosen represent environmentally relevant concentration (10 ␮M). Yet, EROD activity mediated by zebrafish
pharmaceuticals, which have specificity for the major drug metab- CYP1s was not hugely affected by gemfibrozil. Likewise, IC50s for
olizing CYPs in human, CYP1A2, CYP2D6, CYP2C9 and CYP3A4. ciprofloxacin inhibition of zebrafish CYP1s, based on EROD and
Ciprofloxacin is a selective inhibitor of CYP1A2 (Fuhr et al., 1992; MROD activity, were in excess of 850 and 410 ␮M, respectively,
Granfors et al., 2004). Gemfibrozil is a modest inhibitor of CYP1A2 in contrast to the large inhibition of EROD activity in rainbow trout
in humans (Wen et al., 2001). Fish do not have a CYP1A2 ortholog microsomal reactions. The reason for the incongruence between
and MR, a CYP1A2 substrate in mammals, is catalyzed by several the data for microsomes and expressed proteins is not clear.
CYP1s in fish including the single CYP1A protein (Scornaienchi et al., Certainly, species differences may be one plausible reason; the
2010b; Smith and Wilson, 2010), which metabolizes ER and MR microsomes were from rainbow trout and the expressed proteins
equally (Gooneratne et al., 1997). were from zebrafish.
Human CYP3A4 is strongly inhibited by erythromycin (Okudaira
et al., 2007; Yamazaki and Shimada, 1998) and fluoxetine (Jeppesen 4.2. Inhibition of BFC and BQ metabolism
et al., 1996). BFC and BQ are CYP3A substrates in mammals and
in most fish studies, such activity is attributed to CYP3A activity All four inhibitors, at both concentrations, were successful at
(Hegelund et al., 2004; Kashiwada et al., 2005). The assumption reducing the metabolism of BFC, in both undosed and BNF-treated
that these substrates are selectively catalyzed by CYP3A in fish may fish. BFC inhibition by gemfibrozil has been previously reported
be premature; BFC metabolism was higher with CYP1s than with in carp liver microsomes (Thibaut et al., 2006), although at much
CYP3A65 in zebrafish (Scornaienchi et al., 2010a,b). Interestingly, higher concentrations (1 mM) than used in this study. BFC is a
fish CYP3A isoforms are more closely related to CYP3B and CYP3C mammalian CYP3A substrate, metabolized selectively by CYP3A4
genes (Yan and Cai, 2010), suggesting that CYP3A genes are not in humans (Stresser et al., 2000). BQ, another mammalian CYP3A
orthologous across vertebrates. substrate, was inhibited by the high concentration of ciprofloxacin,
Gemfibrozil is a potent inhibitor of CYP2C9 and a modest gemfibrozil and fluoxetine, but not erythromycin. Previous data
inhibitor of CYP1A2 and 2C19 in humans (Wen et al., 2001). Fluox- suggested that different hepatic CYPs were responsible for the
etine effectively inhibits human CYP2D6 and 2C19 (Jeppesen et al., metabolism of BCF and BQ in fish (Scornaienchi et al., 2010b; Smith
1996). There are no CYP2C or CYP2D orthologs in fish, although and Wilson, 2010); a notion supported here because erythromycin
many CYP2 genes are present in fish genomes (Kirischian et al., was not capable of inhibiting the metabolism of both substrates.
2011). The actinopterygian CYP(s) responsible for metabolism of These microsomal data provide evidence that erythromycin
CYP2C9 and CYP2D6 mammalian substrates is unknown. inhibits fish CYPs, but to a minimal degree compared to other
The inhibition potential for pharmaceuticals was determined for tested compounds. The one substrate significantly inhibited at
CYP mediated reactions in hepatic trout microsomes, a widely used both concentrations in untreated and BNF treated trout micro-
aquatic toxicology model species that was large enough to allow the somes was BFC. In a study with catfish intestinal microsomes,
analyses on individual fish. CYP mediated metabolism was mea- erythromycin was less potent compared to other inhibitors, like
sured in fluorescent-based assays with substrates that have been ketoconazole (James et al., 2005). Erythromycin metabolism in
used previously with trout hepatic microsomes (Smith and Wilson, sea bass hepatic microsomes has been reported in the range
2010) such that reasonable catalytic activity was expected for all of 1000 pmol min−1 mg−1 protein (Vaccaro et al., 2003, 2007). If
substrates and we were determining inhibition for multiple hepatic hepatic CYPs in rainbow trout are capable of metabolizing ery-
CYPs. The inhibitory effects of pharmaceuticals for CYP1 proteins thromycin, erythromycin may be competitively inhibiting the
was tested with bacterially expressed zebrafish CYP 1 proteins metabolism of BCF. The metabolism of erythromycin in microsomal
(CYP1A, CYP1B1, CYP1C1 or CYP1C2) using the resorufin-based sub- preparations could significantly reduce erythromycin concentra-
strates only; catalytic activity was expected for most CYP1 proteins tions in the reaction and explain the significant inhibition of CYP1
with these substrates (Scornaienchi et al., 2010a,b). Collectively, proteins, which have no CYP3A, but minimal inhibition of EROD or
this data will identify if CYP inhibition by pharmaceuticals is widely MROD activity in microsomes where CYP1 and CYP3A isoforms are
expected, suggest which microsomal reactions are CYP1 mediated, co-expressed.
and determine whether the selectivity of mammalian inhibitors are
conserved in fish. 4.3. Specificity of CYP inhibition

4.1. Inhibition of EROD and MROD Our data does not support the finding that fish CYPs are
inhibited similarly to mammals. Fluoxetine, a CYP2D6 and 3A4
To our knowledge, no studies have previously determined the inhibitor in mammals, had the broadest inhibition, affecting all
inhibition potential of ciprofloxacin in fish. Ciprofloxacin is a potent substrates tested with rainbow trout microsomes. Fluoxetine has
CYP1A2 inhibitor in mammals (Fuhr et al., 1992; Granfors et al., been reported to inhibit EROD activity in fish hepatocytes and liver
2004). In comparison to other inhibitors, ciprofloxacin did not show microsomes (Laville et al., 2004; Thibaut et al., 2006). In our study,
preferential inhibition of ER or MR; this was evident in assays fluoxetine significantly inhibited EROD activity in trout micro-
with microsomes and expressed zebrafish CYP1A. In microsomes, somes and was particularly potent against the zebrafish CYP1A
EROD activity was reduced by over 60% by 100 ␮M ciprofloxacin protein. Yet, strong inhibition was seen with zebrafish CYP1C1-
from both BNF-treated and untreated (control) trout and yet MROD mediated MROD activity and CYP1C2-mediated BROD activity.
activity was inhibited by 100 ␮M ciprofloxacin in BNF treated trout Collectively, this data suggests fluoxetine is an inhibitor of multiple
only. Inhibition of a number of CYP-mediated reactions, including hepatic CYPs in fish including CYP1s.
EROD, have been reported for enrofloxacin, the parent compound Like fluoxetine, the other inhibitors did not have the specificity
of ciprofloxacin (Vaccaro et al., 2003, 2007). In vivo treatment with expected. Ciprofloxacin is a potent CYP1A2 inhibitor in mam-
enrofloxacin and in vitro exposures caused reduced EROD activity mals yet was neither a preferential inhibitor of zebrafish CYP1s
in sea bass liver microsomes (Vaccaro et al., 2003). nor a potent inhibitor of EROD or BROD activity in trout micro-
Thibaut et al. (Thibaut and Porte, 2008; Thibaut et al., 2006) somes. Erythromycin inhibits mammalian CYP3A4 but was capable
reported inhibition of EROD activity with 1 mM gemfibrozil. In the of nearly completely abolishing CYP1C mediated MROD activity;
E.M. Smith et al. / Aquatic Toxicology 109 (2012) 259–266 265

something no other inhibitor was capable of, even ciprofloxacin Crespi, C.L., Miller, V.P., Penman, B.W., 1997. Microtiter plate assays for inhi-
bition of human, drug-metabolizing cytochromes P450. Anal. Biochem. 248,
the mammalian CYP1 inhibitor. Gemfibrozil is a potent inhibitor of
188–190.
mammalian CYP2C9 and a modest inhibitor of CYP1A2, yet it was Daughton, C.G., Ternes, T.A., 1999. Pharmaceuticals and personal care products in
most potent in inhibiting BFC metabolism and BROD activity. Col- the environment: agents of subtle change? Environ. Health Perspect. 107 (Suppl.
lectively, mammalian CYP inhibitors appear to inhibit fish CYPs but 6), 907–938.
Fuhr, U., Anders, E.M., Mahr, G., Sorgel, F., Staib, A.H., 1992. Inhibitory potency of
do not share isoform specificity. quinolone antibacterial agents against cytochrome-P4501A2 activity in vivo and
in vitro. Antimicrob. Agents Chemother. 36, 942–948.
Gooneratne, R., Miranda, C.L., Henderson, M.C., Buhler, D.R., 1997. Beta-
4.4. Activation of CYP activity
naphthoflavone induced CYP1A1 and 1A3 proteins in the liver of rainbow trout
(Oncorhynchus mykiss). Xenobiotica 27, 175–187.
Surprisingly, several CYP mediated reactions were increased Granfors, M.T., Backman, J.T., Laitila, J., Neuvonen, P.J., 2004. Tizanidine is mainly
metabolized by cytochrome P450 1A2 in vitro. Brit. J. Clin. Pharmacol. 57,
with the addition of pharmaceuticals. PROD, in particular, showed
349–353.
significantly higher rates of metabolism in hepatic microsomes Hahn, M.E., Lamb, T.M., Schultz, M.E., Smolowitz, R.M., Stegeman, J.J., 1993.
from BNF treated fish with the addition of pharmaceuticals yet all Cytochrome P4501A induction and inhibition by 3,3 ,4,4 -tetrachlorobiphenyl
pharmaceuticals increased at least one CYP mediated reaction. Acti- in an Ah receptor-containing fish hepatoma cell line (PLHC-1). Aquat. Toxicol.
26, 185–208.
vation of CYP reactions has been reported with mammalian CYP3A4 Hegelund, T., Ottosson, K., Radinger, M., Tomberg, P., Celander, M.C., 2004. Effects
(Ludwig et al., 1999; Niwa et al., 2003) yet, to our knowledge, CYP of the antifungal imidazole ketoconazole on CYPIA and CYP3A in rainbow trout
activation has not been reported in fish. Until the CYP isoform pri- and killifish. Environ. Toxicol. Chem. 23, 1326–1334.
Hemeryck, A., Belpaire, F.M., 2002. Selective serotonin reuptake inhibitors and
marily responsible for hepatic PROD activity is identified, studies cytochrome P-450 mediated drug–drug interactions: an update. Curr. Drug
of how pharmaceuticals activate PROD are not possible. There are Metab. 3, 13–37.
several reported mechanisms by which CYP mediated metabolism Hignite, C., Azarnoff, D.L., 1977. Drugs and drug metabolites as environmental
contaminants – chlorophenoxyisobutyrate and salicylic-acid in sewage water
can be activated in mammals. effluent. Life Sci. 20, 337–341.
Hirsch, R., Ternes, T., Haberer, K., Kratz, K.L., 1999. Occurrence of antibiotics in the
4.5. Implications of CYP inhibition aquatic environment. Sci. Total Environ. 225, 109–118.
James, M.O., Lou, Z., Rowland-Faux, L., Celander, M.C., 2005. Properties and regional
expression of a CYP3A-like protein in channel catfish intestine. Aquat. Toxicol.
With the exception of fluoxetine and general BFC metabolism, 72, 361–371.
the lowest inhibitor concentration, 10 ␮M, did not significantly Jeppesen, U., Gram, L.F., Vistisen, K., Loft, S., Poulsen, H.E., Brosen, K., 1996.
Dose dependent inhibition of CYP1A2, CYP2C19 and CYP2D6 by citalo-
inhibit the majority of reactions in un-treated fish. The higher con- pram, fluoxetine, fluvoxamine and paroxetine. Eur. J. Clin. Pharmacol. 51,
centration chosen (100 ␮M) is likely orders of magnitude higher 73–78.
than what would be found in the environment. However, many Kashiwada, S., Hinton, D.E., Kullman, S.W., 2005. Functional characterization
of medaka CYP3A38 and CYP3A40: kinetics and catalysis by expres-
pharmaceuticals have been found to bioaccumulate in the tissues sion in a recombinant baculovirus system. Comp. Biochem. Phys. C 141,
of wild fish (Brooks et al., 2005; Nakamura et al., 2008). Additive 338–348.
and synergistic effects are something that should be considered Kirischian, N., McArthur, A.G., Jesuthasan, C., Krattenmacher, B., Wilson, J.Y., 2011.
Phylogenetic and functional analysis of the vertebrate cytochrome P450 2 fam-
particularly because these data suggest broad CYP inhibition by
ily. J. Mol. Evol. 72, 56–71.
the tested compounds. The environmental relevance of inhibition Kolpin, D.W., Furlong, E.T., Meyer, M.T., Thurman, E.M., Zaugg, S.D., Barber, L.B.,
by these compounds is therefore not straightforward. Clearly, our Buxton, H.T., 2002. Pharmaceuticals, hormones, and other organic wastewater
data suggests that inhibition of CYP mediated reactions is possible contaminants in U.S. streams, 1999–2000: a national reconnaissance. Environ.
Sci. Technol. 36, 1202–1211.
with environmentally relevant pharmaceuticals but the concentra- Laville, N., Ait-Aissa, S., Gomez, E., Casellas, C., Porcher, J.M., 2004. Effects of human
tions found in the environment may be too low to cause significant pharmaceuticals on cytotoxicity, EROD activity and ROS production in fish hep-
drug–drug interactions. atocytes. Toxicology 196, 41–55.
Lindberg, R.H., Wennberg, P., Johansson, M.I., Tysklind, M., Andersson, B.A.V., 2005.
Screening of human antibiotic substances and determination of weekly mass
Acknowledgements flows in five sewage treatment plants in Sweden. Environ. Sci. Technol. 39,
3421–3429.
Ludwig, E., Schmid, J., Beschke, K., Ebner, T., 1999. Activation of human cytochrome
This work was supported by funding from the Canadian Water P-450-catalyzed meloxicam 5’-methylhydroxylation by quinidine and hydro-
Network, the Ontario Ministry of the Environment Best in Sci- quinidine in vitro. J. Pharmacol. Exp. Ther. 290, 1–8.
Martignoni, M., Groothuis, G.M.M., de Kanter, R., 2006. Species differences between
ence program, and NSERC discovery (grant #328204) program. E.S
mouse, rat, dog, monkey and human CYP-mediated drug metabolism, inhibition
was partially supported by funding provided by the Department and induction. Expert Opin. Drug Metab. 2, 875–894.
of Biology, McMaster University. The authors wish to thank Dr. Metcalfe, C.D., Miao, X.S., Koenig, B.G., Struger, J., 2003. Distribution of acidic and
Malin Celander and Johanna Gräns for advice on modifying the BCF neutral drugs in surface waters near sewage treatment plants in the lower Great
Lakes, Canada. Environ. Toxicol. Chem. 22, 2881–2889.
assays. We thank Mr. James Harskamp for preparation of the CYP1 Miller, V.P., Stresser, D.M., Blanchard, A.P., Turner, S., Crespi, C.L., 2000. Fluorometric
expressed proteins used in this study. high-throughput screening for inhibitors of cytochrome P450. Ann. N.Y. Acad.
Sci. 919, 26–32.
Nakamura, Y., Yamamoto, H., Sekizawa, J., Kondo, T., Hirai, N., Tatarazako, N., 2008.
Appendix A. Supplementary data The effects of pH on fluoxetine in Japanese medaka (Oryzias latipes): acute
toxicity in fish larvae and bioaccumulation in juvenile fish. Chemosphere 70,
865–873.
Supplementary data associated with this article can be found, in
Niwa, T., Shiraga, T., Yamasaki, S., Ishibashi, K., Ohno, Y., Kagayama, A., 2003. In vitro
the online version, at doi:10.1016/j.aquatox.2011.08.022. activation of 7-benzyloxyresorufin O-debenzylation and nifedipine oxidation in
human liver microsomes. Xenobiotica 33, 717–729.
Okudaira, T., Kotegawa, T., Imai, H., Tsutsumi, K., Nakano, S., Ohashi, K., 2007. Effect
References of the treatment period with erythromycin on cytochrome P450 3A activity in
humans. J. Clin. Pharmacol. 47, 871–876.
Arellano-Aguilar, O., Montoya, R.M., Garcia, C.M., 2009. Endogenous functions and Omura, T., Sato, R., 1964. The carbon monoxide-binding pigment of liver micro-
expression of cytochrome P450 enzymes in teleost fish: a review. Rev. Fish. Sci. somes. J. Biol. Chem. 239, 2370–2378.
17, 541–556. Scornaienchi, M.L., Thornton, C., Willett, K.L., Wilson, J.Y., 2010a. Cytochrome P450-
Brooks, B.W., Chambliss, C.K., Stanley, J.K., Ramirez, A., Banks, K.E., Johnson, R.D., mediated 17beta-estradiol metabolism in zebrafish (Danio rerio). J. Endocrinol.
Lewis, R.J., 2005. Determination of select antidepressants in fish from an 206, 317–325.
effluent-dominated stream. Environ. Toxicol. Chem. 24, 464–469. Scornaienchi, M.L., Thornton, C., Willett, K.L., Wilson, J.Y., 2010b. Functional dif-
Corcoran, J., Winter, M.J., Tyler, C.R., 2010. Pharmaceuticals in the aquatic environ- ferences in the cytochrome P450 1 family enzymes from zebrafish (Danio
ment: a critical review of the evidence for health effects in fish. Crit. Rev. Toxicol. rerio) using heterologously expressed proteins. Arch. Biochem. Biophys. 502,
40, 287–304. 17–22.
266 E.M. Smith et al. / Aquatic Toxicology 109 (2012) 259–266

Smith, E.M., Wilson, J.Y., 2010. Assessment of cytochrome P450 fluoromet- endogenous and xenobiotic metabolizing enzymes in carp liver: an in vitro
ric substrates with rainbow trout and killifish exposed to dexametha- study. Environ. Sci. Technol. 40, 5154–5160.
sone, pregnenolone-16alpha-carbonitrile, rifampicin, and beta-naphthoflavone. Vaccaro, E., Giorgi, M., Longo, V., Mengozzi, G., Gervasi, P.G., 2003. Inhibition of
Aquat. Toxicol. 97, 324–333. cytochrome P450 enzymes by enrofloxacin in the sea bass (Dicentrarchus labrax).
Stegeman, J.J., 1993. Cytochrome P450 forms in fish. In: Schenkman, J.B., Greim, H.V. Aquat. Toxicol. 62, 27–33.
(Eds.), Handbook of Experimental Pharmacology. Springer, Berlin, pp. 279–291. Vaccaro, E., Salvetti, A., Del Carratore, R., Nencioni, S., Longo, V., Gervasi, P.G., 2007.
Stegeman, J.J., Hahn, M.E., Weisbrod, R., Woodin, B.R., Joy, J.S., Najibi, S., Cohen, R.A., Cloning, tissue expression, and inducibility of CYP 3A79 from sea bass (Dicen-
1995. Induction of cytochrome P4501A1 by aryl hydrocarbon receptor agonists trarchus labrax). J. Biochem. Mol. Toxicol. 21, 32–40.
in porcine aorta endothelial cells in culture and cytochrome P4501A1 activity Wen, X., Wang, J.S., Backman, J.T., Kivisto, K.T., Neuvonen, P.J., 2001. Gemfibrozil
in intact cells. Mol. Pharmacol. 47, 296–306. is a potent inhibitor of human cytochrome P4502C9. Drug Metab. Dispos. 29,
Stresser, D.M., Blanchard, A.P., Turner, S.D., Erve, J.C., Dandeneau, A.A., Miller, V.P., 1359–1361.
Crespi, C.L., 2000. Substrate-dependent modulation of CYP3A4 catalytic activity: Yamazaki, H., Shimada, T., 1998. Comparative studies of in vitro inhibition of
analysis of 27 test compounds with four fluorometric substrates. Drug Metab. cytochrome P450 3A4-dependent testosterone 6 beta-hydroxylation by rox-
Dispos. 28, 1440–1448. ithromycin and its metabolites, troleandomycin, and erythromycin. Drug Metab.
Thibaut, R., Porte, C., 2008. Effects of fibrates, anti-inflammatory drugs and antide- Dispos. 26, 1053–1057.
pressants in the fish hepatoma cell line PLHC-1: cytotoxicity and interactions Yan, J., Cai, Z., 2010. Molecular evolution and functional divergence of the
with cytochrome P450 1A. Toxicol. In Vitro 22, 1128–1135. cytochrome P450 3 (CYP3) family in actinopterygii (ray-finned fish). PLoS One
Thibaut, R., Schnell, S., Porte, C., 2006. The interference of pharmaceuticals with 5 (12), e14276.

You might also like