Вы находитесь на странице: 1из 12

Journal of Chromatography B, 1029 (2016) 145–156

Contents lists available at ScienceDirect

Journal of Chromatography B
journal homepage: www.elsevier.com/locate/chromb

Comparison of the in vitro metabolism of psoralidin among different


species and characterization of its inhibitory effect against UDP-
glucuronosyltransferase (UGT) or cytochrome p450 (CYP450)
enzymes
Xianbao Shi a,1 , Gang Zhang a,b,1 , Brianna Mackie b , Shuman Yang c , Jian Wang d ,
Lina Shan a,∗
a
Department of Pharmacy, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, China
b
Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
c
Department of Internal Medicine/Community Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
d
School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China

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

Article history: Psoralidin has shown a variety of biological and pharmacological activities such as anti-tumor anti-
Received 8 April 2016 oxidant, anti-bacterial, anti-depressant and anti-inflammatory activities. Herein, we reported the
Received in revised form 17 June 2016 metabolism of psoralidin among different species and its inhibitory effect against UGTs and CYP450s.
Accepted 18 June 2016
Liquid chromatography was used to investigate the inhibitory activity of psoralidin against ten different
Available online 30 June 2016
UGTs and eight distinct CYP450 isoforms. In addition, we characterized the CYP450 isoforms involved in
the psoralidin metabolism on the basis of chemical inhibition studies and screening assays with recom-
Keywords:
binant human cytochrome P450s. In vitro metabolic profiles and metabolites of psoralidin from varying
Psoralidin
Cytochrome P450
liver microsomes obtained from human (HLMs), monkey (MLMs), rat (RLMs), dog (DLMs), minipig (PLMs)
Glucuronosyltransferase (UGT) and rabbit (RAMs) were determined by LC–MS/MS. In vivo pharmacokinetic profiles were investigated by
Drug-drug interactions injecting psoralidin (2 mg/kg) into the tail vein of Wistar rats. Molecular modeling studies were carried
LC–MS/MS out in order to assess the binding profile and recognition motif between psoralidin and the enzymes.
Psoralidin showed potent and noncompetitive inhibition against UGT1A1, UGT1A7, CYP1A2 and CYP2C8
with IC50 values of 6.12, 0.38, 1.81, 0.28 ␮M, respectively. The metabolism of psoraldin exhibited signifi-
cant differences among humans, monkeys, dogs, minipigs, rabbits and rats; however, monkeys showed
the highest similarity to humans. Furthermore, eleven metabolites were observed among these species
and their structures were characterized by LC–MS/MS. CYP2C19 played a key role in the metabolism of
psorslidin in human liver microsomes. These findings could be used to advance the understanding of
psoralidin.
© 2016 Elsevier B.V. All rights reserved.

1. Introduction drugs to treat various ailments [4–7]. Previous studies have demon-
strated biological and pharmacological activities of psoralidin, but
Psoraleacorylifolia is a plant widely used in traditional Chinese pharmacokinetic evaluation has never been reported. Pharmacoki-
medicine, which exhibits anti-oxidative, anti-microbial, anti- netic information could be critical to further understand the clinical
inflammatory and anti-tumor activities [1–3]. One of the major efficiency and safety of psoralidin. Cytochrome p450 (CYP450)
components isolated from the seed of psoraleacorylifolia is psora- and UDP-glucuronosyltransferase (UGT) are the most important
lidin (Fig. 1), which is extensively used by itself or along with other enzyme systems of detoxication, and approximately 70%–80% of
the known Phase I and II metabolisms are attributed to them [8–10].
Inhibition of CYP450s and UGTs might increase the concentra-
tion of co-administered drugs in the blood or therapeutic targets
∗ Corresponding author at: The First Affiliated Hospital of Liaoning Medical Uni- and result in adverse effects [11–13]. These severe adverse effects
versity, No. 5 Renmin Street, Jinzhou 121001, China. have resulted in costly and late failures of drug development, and
E-mail address: shanlina123@163.com (L. Shan).
1 withdrawal of drugs already on the market [14,15]. Psoralen and
These authors have contributed equally to this work.

http://dx.doi.org/10.1016/j.jchromb.2016.06.031
1570-0232/© 2016 Elsevier B.V. All rights reserved.
146 X. Shi et al. / J. Chromatogr. B 1029 (2016) 145–156

2.2. Preparation and characterization of liver microsomes

Liver microsomes from humans (HLMs), monkeys (MLMs), rab-


bits (RAMs), rats (RLMs), dogs (DLMs), and minipigs (PLMs) used
in this study were provided by the Research Institute for Liver Dis-
ease Co. (Shanghai, China). The HLMs was prepared from eleven
individual human donor livers. MLMs, RAMs, RLMs, DLMs, and
PLMs were prepared from ten individual animal livers. Protein
concentration and enzyme activities of CYP1A2, CYP2C9, CYP2C8,
CYP2A6, CYP2D6, CYP2E1, CYP3A4 and CYP2C19 had been previ-
ously characterized by the Research Institute for Liver Disease Co. In
Fig. 1. The structure of psoralidin.
brief, the concentration of protein was determined by bicinchoninic
acid (BCA) Protein Assay Kit, which was previously described by
Smith et al. [21]. The characterization of above CYP450s was per-
formed by detecting the metabolites of their probe substrates
isopsoralen, two important compounds of psoralea, have shown the
using LC–MS/MS method, the procedure of which was described
inhibition activity against -CYP3A4 in a concentration-dependent
in Section 2.5. All the probe substrates, their metabolites and the
manner [16]. However, the effect of psoralidin on CYP450 isoforms
inhibitors used as positive control were presented in Table 1.
has never been reported. In recent years, in vitro methods have been
used to predict in vivo aspects of humans such as drug metabolism
2.3. Analytical instruments and conditions
and pharmacokinetics and received satisfactory results [17,18].
Although all members of the CYP450 or UGT super family pos-
The HPLC system (Shimadzu, Kyoto, Japan) consisted of a CBM-
sess highly conserved regions of amino acid residues, there are
20Alite system, two LC-20AB pumps, and an SPD-20A ultraviolet
a few differences in the primary amino acid sequence of CYP450
light (UV) detector. The chromatographic separation was achieved
and UGT across species. However, minor differences in the amino
using an Inertsil ODS-3 C18 column (4.6 mm × 150 mm, 5 ␮m,
acid sequences at the active sites of CYP450s can result in a wide
Shimadzu-GL Sciences Inc., Kyoto, Japan). The mobile phases con-
variation of substrate specificity and catalytic activity [19]. The
sisted of LC grade water containing 0.1% formic acid (A) and
differences in CYP450 or UGT isoforms are a major cause of the dif-
LC grade acetonitrile (B), with the following gradient profile:
ferences in drug metabolism across varying species [20]. Therefore,
0–12 min, 20%–90% B; 12–17 min, 90% B; 17–17.30 min, 90%–20%
understanding the differences in CYP450 or UGT function between
B; 17.30–22.00 min, 20% B. The flow rate was 1 mL/min and col-
animals and humans is critical.
umn temperature was set to 40 ◦ C. Psoralidin and its metabolites
We sought to examine the impact of psoralidin against ten
were detected at 347 nm. Due to the absence of authentic standards
UGTs and eight CYP450 isoforms. The plasma concentrations and
for metabolites of psoralidin, the standard curve for psoralidin was
pharmacokinetic parameters were also evaluated after intravenous
used to quantify P3 in the incubation mixtures. Method validation
administration of psoralidin. CYP450 isozymes involved in pso-
was provided in supporting materials (Tables S1–S2, Fig. S1). The
ralidin metabolism were confirmed by assays with recombinant
performance parameters including specificity, linearity, accuracy,
CYP450 isoforms and chemical inhibition experiments. In addi-
precision, decision limit, quantification limit and stability showed
tion, We determined the psoralidin metabolism characteristics in
the validated method was qualified for the determination of pso-
liver microsomes from six species, including rat, minipig, rabbit,
ralidin and its metabolites.
dog, monkey and human. This was to confirm which species best
LC–MS/MS analysis was performed on Ultra Performance Liquid
resembles the drug-metabolism capabilities of humans. Addition-
Chromatograph (UPLC) equipped with a Q-TOF SYNAPT G2-Si High
ally, molecular docking was employed to further understand the
Definition Mass Spectrometry (Waters Corporation, Milford, MA).
interactions between psoralidin and CYP450s.
The UPLC system was fitted with an Acquity UPLC BEH C18 column
(2.1mm × 100 mm, 1.7 ␮m; Waters Co., Ltd., USA). Mass detection
was performed in negative mode from m/z 100 to 1000. The optimal
2. Materials and methods parameters were as follows: Collision gas is Argon; trap collision
energies were 6 V (low energy) and 20–50 V (high energy); capil-
2.1. Chemicals and reagents lary and cone voltages were 2 kV and 40 V; source and desolvation
temperatures were 120 and 600 ◦ C; the cone and desolvation gas
Psoralidin was purchased from Shifeng Corp in Shanghai, China, flows were 50 and 800 L h−1 , respectively.
and its purity was above 98%. The substrates, metabolites and pos-
itive inhibitors of CYP450 isoforms are listed in Table 1. Probe sub- 2.4. CYP450 probe substrate assays
strates and positive inhibitors were purchased from Sigma-Aldrich
(St. Louis, MO, USA). Metabolites of probe substrates were provided The CYP450s activities were characterized based on their probe
by the Research Institute for Liver Disease Co. (Shanghai, China). reactions: CYP1A2 (phenacetin o-deethylation), CYP2A6 (coumarin
1-aminobenzotriazole (ABT), glucose-6-phosphate dehydroge- 7-hydroxylation), CYP2C8 (paclitaxel 6␣-hydroxylation), CYP2C9
nase, NADP+ , d-glucose-6-phosphate, 4-Methylumbelliferone(4- (diclofenac 4 -hydroxylation), CYP2D6 (dextromethorphan o-
MU), 4-methylumbelliferone-␤-d-glucuronide (4-MUG), Tris-HCl, demethylation), CYP2E1 (chlorzoxazone 6-hydroxylation), CYP3A4
7-hydroxycoumarin and uridine 5 -diphosphoglucuronic acid (testosterone 6␤-hydroxylation) and CYP2C19 (S-mephenytoin4 -
(UDPGA) (trisodium salt) werepurchased from Sigma-Aldrich. hydroxylation) [22].
Recombinant human supersomes (UGT1A1, UGT1A3, UGT1A6,
UGT1A7, UGT1A8, UGT1A9, UGT1A10, UGT2B4, UGT2B7, UGT2B15, 2.5. Incubation systems with liver microsomes or recombinant
CYP1A2, CYP2C9, CYP2D6, CYP2E1, CYP3A4 and CYP2C19) were CYP450 supersomes
obtained from BD Gentest Corp. (Woburn, MA, USA). All other
reagents were of HPLC grade or of the highest grade commercially The optimal incubation conditions of microsomes have been
available. reported [23,24]. In brief, the typical incubation system contained
X. Shi et al. / J. Chromatogr. B 1029 (2016) 145–156 147

Table 1
The substrates, inhibitors and metabolites of CYP450.

CYP isoforms Substrates Inhibitors metabolites

CYP1A2 phenacetin furafylline O-deethylation phenacetin


CYP2C9 diclofenac sulfaphenazole 4 -hydroxylation diclofenac
CYP2C8 paclitaxel quercetin 6␣-hydroxylation paclitaxel
CYP2A6 coumarin xanthotoxin 7-hydroxylation coumarin
CYP2D6 dextromethorphan quinidine O-demethylation dextromethorphan
CYP2E1 chlorzoxazone clomethiazole 6-hydroxylation chlorzoxazone
CYP3A4 testosterone ketoconazole 6␤-hydroxylation testosterone
CYP2C19 S-mephenytoin omeprazole 4 -hydroxylation S-mephenytoin

100 mM potassium phosphate buffer (pH 7.4), NADPH-generating UGT1A7, UGT1A8 and UGT1A9, respectively. The reactions were
system (1 mM NADP+ , 10 mM glucose-6-phosphate, 1 unit/mL quenched by 200 ␮L acetonitrile and 7-hydroxycoumarin (100 ␮M)
of glucose-6-phosphate dehydrogenase, and 4 mM MgCL2 ), the which was used as an internal standard. The mixture was cen-
appropriate concentration of liver microsomes or recombinant trifuged at 20,000g for 20 min and an aliquot of supernatant was
CYP450s supersomes, the corresponding probe substrate and pso- analyzed by HPLC with ultraviolet light (UV) detection.
ralidin (or positive inhibitor for different probe reactions) in a final
volume of 200 ␮L. According to preliminary experiments (data not 2.8. CYP450inhibition experiments
shown), the final protein concentration of 0.3 mg/mL in liver micro-
somes or 15 nM in recombinant human supersomes, and a 30 min Marker assays for CYP450s (CYP1A2, CYP3A4, CYP2D6, CYP2E1,
reaction time were selected to ensure linear formation of metabo- CYP2A6, CYP2C9, CYP2C19 and CYP2C8) were conducted using the
lites during the incubations. There was a 3 min preincubation at above incubation systems and conditions and consisted of liver
37 ◦ C before the reaction was initiated by adding the NADPH- microsomes, the NADPH-generating system, probed substrates at
generating system. The reaction was placed on ice and terminated the concentration of their approximate Km value, and psoralidin
by adding 200 ␮L acetonitrile and an internal standard. The mixture (or the control inhibitor). The positive control inhibitors of indi-
was centrifuged at 20,000g for 20 min and an aliquot of supernatant vidual CYP450 isoforms were as follows: furafylline (10 ␮M) for
was transferred for HPLC or LC–MS/MS analysis. CYP1A2, xanthotoxin (2.5 ␮M) for CYP2A6, quercetin (2 ␮M) for
CYP2C8, sulfaphenazole (10 ␮M) for CYP2C9, omeprazole (20 ␮M)
2.6. Pharmacokinetics study for CYP2C19, quinidine (10 ␮M) for CYP2D6, clomethiazole (50 ␮M)
for CYP2E1, ketoconazole for CYP3A4 and ABT (500 mM) for broad
Six male Wister rats (200–250 g) were obtained from the Lab- CYP450s [26,27]. Furthermore, kinetic analysis was performed for
oratory Animal Center of Liaoning Medical University. Approval the isoforms which had been inhibited by more than 90% total
from the Institutional Animal Ethics Committee was sought and activity. IC50 values were obtained by incubating various psora-
the study protocols were approved before the commencement of lidin concentrations (0–15 ␮M for CYP1A2, 0–5 ␮M for CYP2C8,
the studies. Psoralidin was administered intravenously into the tail 0–100 ␮M for UGT1A1, and 0–5 ␮M for UGT1A7) with the isoforms.
vein at a dose of 2 mg/kg. The blood samples were collected through Dixon and Lineweaver-Burk plots were generated to determine
orbital venous plexus at 0.083, 0.3, 0.75, 1, 1.5, 2, 3.5, 4.5, 6 and the inhibition type. A second graph was created by plotting the
8 h after intravenous administration, and centrifuged at 2000g for slopes from the Lineweaver-Burk plot versus the concentrations of
5 min at 4 ◦ C to obtain the plasma samples. The plasma (0.1 mL) psoralidin to calculate the Ki values.
was mixed with 200 ␮L acetonitrile and phenacetin (100 ␮M) was
used as an internal standard. After centrifugation of the mixture 2.9. Identification of metabolites in human, rat, monkey, minipig,
at 20,000g for 20 min at 4 ◦ C, the supernatant was collected and rabbit and dog liver microsomes
analyzed by HPLC.
In this study, in vitro metabolism of psoralidin among species
2.7. UGTs inhibition experiments was compared with respect to metabolites, metabolic profiles and
catalytic efficiency using liver microsomes from humans (HLMs),
4-MU and recombinant UGT isoforms were used as the nonspe- rabbits (RAMs), rats (RLMs), dogs (DLMs), minipigs (PLMs), and
cific probed substrates and enzyme source to assess the inhibitory monkeys (MLMs). Incubating conditions were conducted using the
effect of psoralidin against UGT isoforms. Incubation and ana- reaction system described above. Metabolites were determined by
lytical conditions were as described previously [25]. In brief, HPLC.
the mixture (200 ␮L total volume) contained recombinant UGTs
(final concentrations: 0.125, 0.05, 0.025, 0.05, 0.025, 0.05, 0.05, 2.10. Assay with recombinant P450s
0.25, 0.05 and 0.2 mg/mL for UGT1A1, UGT1A3, UGT1A6, UGT1A7,
UGT1A8, UGT1A9, UGT1A10, UGT2B4, UGT2B7 and 2B15, respec- Six cDNA-expressed human CYP isoforms (CYP1A2, CYP2C9,
tively), 5 mM UDPGA, 5 mM MgCL2 , 50 mM Tris-HCl buffer (pH CYP2C19, CYP2D6, CYP2E1 and CYP3A4) were used to screen the
7.4) and 4-MU in the absence or presence of different concen- involved isoform(s) for metabolites of psoralidin. The incubations
trations of psoralidin. The concentrations of 4-MU were 110 ␮M were carried out with each of the CYP450 isoforms using the proto-
for UGT1A1, 1200 ␮M for UGT1A3, 110 ␮M for UGT1A6, 15 ␮M col described above. Psoralidin (10 ␮M) was incubated with each of
for UGT1A7, 750 ␮M for UGT1A8, 30 ␮M for UGT1A9, 80 ␮M for the recombinant CYP450s (15 nM) at 37 ◦ C for 30 min and possible
UGT1A10, 1200 ␮M for UGT2B4, 350 ␮M for UGT2B7 and 250 ␮M metabolites were monitored by HPLC.
for UGT2B15. Psoralidin was dissolved in methanol and the final
concentration of methanol was 0.5% (v/v). After a 5 min prein- 2.11. Chemical inhibition assays
cubation at 37 ◦ C, UDPGA was added to the mixture to initiate
the reaction. Incubation time was 120 min for UGT1A1, UGT1A3, Initially, we used ABT (a broad-specificity P450 inactivator)
UGT1A10, UGT2B4, UGT2B7 and UGT2B15, 30 min for UGT1A6, to screen the P450 isoform(s) responsible for the formation of
148 X. Shi et al. / J. Chromatogr. B 1029 (2016) 145–156

Fig. 2. Representative HPLC profiles of psoralidin and its metabolites in cDNA-expressed human P450 isoforms liver microsomes from HLMs, DLMs, MLMs, PLMs, and RLMs.
Psoralidin (10 ␮M) was incubated with liver microsomes (0.3 mg/mL) from different species and an NADPH-generating system at 37 ◦ C for 30 min.

metabolites in HLMs. If ABT inhibited the formation of psora- 1000. The injections volume was 0.5 ␮L using the partial-loop
lidin metabolites, it suggested that CYP450s were responsible for mode, the column temperature was 55 ◦ C and the flow rate was
psoralidin metabolism in HLMs. Additionally, the metabolism of 3 ␮L/min. The mobile phases consisted of LC grade water with 0.1%
psoralidin in HLMs in the absence or presence of selective inhibitors formic acid (A) and LC grade acetonitrile (B) with the following
for CYP1A2, CYP2A6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1 gradient profile: 0.0–10.0 min from 30% to 50% B, 10–11 min from
and CYP3A4 were measured to explore the enzyme(s) involved in 50% to 90% B, 11–12 min 90% B, 12–13 min from 90% to 30% B, and
this biotransformation of psoralidin. 13–15 min at 30% B.

2.12. Enzymatic kinetic study 2.14. Determination of P3 structure

The enzyme kinetics of psoralidin metabolism among six species As the main metabolite, P3 in HLMs was determined by AB SCIEX
and the recombinant human CYP2C19 enzyme were investigated. 4000 Q-TrapTM (Applied Biosystems, Foster City, CA) interfaced
As the dominant metabolite of psoralidin, P3 was selected to con- online with an ekspertultra LC 100 system (Applied Biosystems)
duct the kinetic parameter evaluation in each species and CYP2C19. in negative mode. The separation of samples was achieved on a
Preliminary experiments were carried out to ensure that the for- HYPERSIL BDS C18 column (4.6 mm × 150 mm, 5 ␮m, Dalian Elite
mation of the metabolites was linear with the reaction time and the Analytical Instruments Co., Dalian, China). The mobile phases and
concentration of the protein in liver microsomes. Concentrations of chromatographic condition were described as above. Multiple-
the psoralidin dilutions were between 0.1 to 250 ␮M. Protein con- reaction monitoring (MRM) scans were used to detect and
tents of liver microsomes and recombinant human CYP2C19 were identifyP3 . MS analyses were conducted with ion transitions m/z
0.3 mg/mL and 15 nM, respectively. Incubation time was 30 min. 351 → 351. The optimized MS instrument parameters were set as
All incubations were carried out in three independent experi- follows: ion spray voltage, 4500 V; ion source temperature, 650 ◦ C;
ments. The apparent Km and Vmax values were calculated from curtain gas, 10 psi; gas 1, 50 psi; and gas 2, 50 psi.
nonlinear regression analysis of experimental data according to
the Michaelis-Menten equation, and the results were graphically 2.15. Docking study of psoralidin into the reported structures of
represented by Eadie-Currently Hofstee plots. The intrinsic clear- CYP1A2, CYP2C8 and CYP2C19
ance (CLint ) for formation of psoralidin metabolite was calculated
by Vmax /Km . Kinetic constants were estimated using origin 7.5 and Sybyl 6.9.1 software package was used for protein and ligand
their estimates are presented as mean ± standard deviation (SD). preparation. AutoDock 4.02 was used for molecular docking. Dis-
covery Studio Visualizer was used for graphic display.
2.13. Analysis of metabolites by LC–MS/MS The X-ray structures of CYP 1A2 (PDB code 2HI4), 2C8 (PDB code
2VN0) and 2C19 (PDB code 4GQS) were obtained from RCSB Pro-
Structure characterization of metabolites in HLMs, DLMs, PLMs, tein Databank (http://rcsb.org/). Heme moiety was considered as a
RAMs, RLMs and MLMs were performed on UPLC–MS/MS. Mass part of the protein and Tripos atom types were defined accordingly.
detection was performed in negative ion mode from m/z 100 to Polar hydrogens and partial charges were added for CYP proteins
X. Shi et al. / J. Chromatogr. B 1029 (2016) 145–156 149

Table 2
The pharmacokinetic parameters of psoralidin in rats after intravenous administra-
tion (n = 6).

Pharmacokinetic parameters Units Values (mean ± SD)

AUC (0-∞) mg/L/h 1.95 ± 0.50


t1/2 h 4.45 ± 0.95
V L/kg 6.22 ± 1.82
CL L/h/kg 1.12 ± 0.45
Cmax mg/L 2.19 ± 0.48

54.42% ± 4.29%, 140.74% ± 10.28% and 77.79% ± 5.23% of the human


levels. Metabolism of psoralidin exhibited significant differences
between species (P < 0.05).
Fig. 3. The plasma concentration profiles after intravenous administration of pso-
ralidin at a single dose of 2 mg/kg. Data represent mean ± SD of six rats. 3.2. Pharmacokinetics

using the Kollman United Atom charges with Sybyl 6.9.1, and After injecting psoralidin intravenously, the blood samples were
energy minimization was made employing both steepest descent collected at different time points. These samples were processed
and conjugate gradients protocols. as described earlier and analyzed by the presently developed HPLC
The psoralidin structure was built by the Sybyl 6.9.1 soft- method. The plasma concentration profile of psoralidin was shown
ware package (Tripos Associates: St. Louis, MO, 2003), assigned in Fig. 3. The pharmacokinetic (PK) parameters such as AUC(0-∞) ,
charges using the Gasteiger–Hückel method and minimized with t1/2 , V, CL and Cmax were calculated by anoncompartmental method
the Powell method (Tripos force field) with an energy gradient of using DAS 3.1 software package (Jiangxi University of Traditional
0.05 kcal/(mol Å). Flexible torsions in the ligands were assigned and Chinese Medicine, Nanchang, China) and the values were showed
all dihedral angles were allowed to rotate freely. in Table 2.
Automated docking studies were performed with AutoDock 4.0.
A 80 × 80 × 80 Å grid box with a grid spacing of 0.375 Å was gener- 3.3. Inhibition against UGTs and CYP450activities by psoralidin
ated for the receptor. Affinity grid fields were generated using the
auxiliary program AutoGrid 4.0. Many herbs have inhibitory activity towards CYP450s or
The Lamarckian genetic algorithm (LGA) was used to find the UGTs, which may affect the concentration of co-administered
appropriate binding positions, orientations, and conformations of drugs and result in adverse effects of the drugs. Thus, we
the ligands. The optimized AutoDocking parameters increased the determined the inhibitory potential of psoralidin against eight
maximum number of energy evaluations to 25,000,000 per run, set CYP450s and ten UGTs. 100 ␮M of psoralidin inhibited the
the iterations of Solis & Wets local search to 3000; set the number activities of UGT1A1, UGT1A7, CYP1A2 and CYP2C8 by more
of individuals in population to 300 and the number of generations than 90%. The strong inhibitory effects against the activities of
to 100. All other parameters were maintained as the default. Clus- UGT1A1, GT1A7, CYP1A2 and CYP2C8 were in a concentration-
ter analysis with AutoDock results was performed to determine if dependent manner. The IC50 values of UGT1A1, GT1A7, CYP1A2 and
different binding sites had been produced from multiple runs. CYP2C8 were 6.12 ± 0.43 ␮M, 0.38 ± 0.03 ␮M, 1.818 ± 0.22 ␮M and
0.28 ± 0.03 ␮M, respectively. The Ki values were 5.638 ± 0.46 ␮M,
2.16. Statistical analysis 0.248 ± 0.02 ␮M, 1.158 ± 0.13 ␮M and 0.25 ± 0.03 ␮M, respectively.
Dixon and Lineweaver-Burk plots showed that psoralidin non-
Statistical analysis was performed with SPSS 13.0 software for competitively inhibited UGT1A1, GT1A7, CYP1A2 and CYP2C8
Windows operation system (SPSS Inc, Chicago, IL, USA). All grouped (Supplemental Figs. S2–S5).
data were expressed as means ± SD. The identification of signifi-
cance between different groups was carried out with student t-test. 3.4. Identifying CYP450 involved in the metabolism of psoralidin
Two-sided P < 0.05 was considered to be statistically significant.
The enzymes involved in the formation of P3 were investigated
3. Results using cDNA-expressed human P450 isoforms. Quantities of P3 in
the CYP3A4, CYP2D6, CYP2E1, CYP2C9 and CYP1A2 incubation were
3.1. Analysis of psoralidin metabolites compared with its counterpart from CYP2C19 incubation which
was normalized to 100%. As shown in Fig. 4, the quantities of P3
After psoralidin was incubated with liver microsomes from generated by CYP3A4, CYP2D6, CYP2E1, CYP2C9 and CYP1A2 were
humans, monkeys, dogs, minipigs, rats or rabbits, five new 0.42%8 ± 0.15%, 13.58%8 ± 2.2%, 0.47%8 ± 0.11%, 6.94% ± 1.63% and
peaks (P1 –P5 ) were observed in RLMs and two new peaks 8.5%8 ± 1.32% of the CYP2C19 levels. CYP2C19 demonstrated a vast
(P1 , P3 ) were found in HLMs, MLMs, DLMs, PLMs and RAMs ability to catalyze the formation of P3 , while other enzymes dis-
by HPLC as compared to the control group (CTRL) (Fig. 2). played limited ability to metabolize psoralidin. The key role of
The formation of these metabolites was time-, NADPH-, and CYP2C19 in metabolizing psoralidin was also confirmed by the
microsome-dependent. P3 , the dominant metabolite in the human chemical inhibition assays. The formation of P3 could be potently
liver microsome, was also generated by all of the animal inhibited by ABT (a broad specificity CYP450 inactivator) and
liver microsomes. Therefore, P3 was selected to evaluate the omeprazole (a potent inhibitor of CYP2C19) to less than 10% activ-
metabolism differences among different species. Peak areas of ity, while selective inhibitors of other CYP450 isoforms had minor
P3 in the animal incubation were compared with its counter- effects on the formation of P3 (Fig. 5). According to the results of
part from human incubation, which was normalized to 100%. As the assays with recombinant CYP450 isoforms and chemical inhi-
a result, the peak areas of P3 generated by rats, dogs, minip- bition experiments, CYP2C19 was the main enzyme involved in the
igs, rabbits and monkeys were 214.38% ± 16.36%, 50.37% ± 3.82%, metabolism of psoralidin.
150 X. Shi et al. / J. Chromatogr. B 1029 (2016) 145–156

Fig. 4. Representative HPLC profiles of psoralidin and its metabolites in recombinant CYP450 supersomes. Psoralidin (10 ␮M) was incubated with recombinant CYP450
supersomes (15 nM) and an NADPH-generating system at 37 ◦ C for 30 min.

3.5. Kinetic characteristics of psoralidin metabolism 3.6. Structure characterization of metabolites by LC–MS/MS

To further characterize the kinetic differences among different In this study, psoralidin and eleven metabolites were identified
species and identify the optimal animal species to serve as a model from HLMs, DLMs, RLMs, PLMs, MLMs and RAMs based on MS and
of metabolism in humans, psoralidin (0.1–200 ␮M) was incubated MS/MS data by using UPLC/Q-TOF-MS/MS in negative mode of an
in HLMs, MLMs, DLMs, PLMs, RLM, RAMs and CYP2C19. Under the ESI mass spectra.Eight (M1 –M5 , M7 –M9 ), seven (M1 –M5 , M10 and
experimental conditions used, the metabolism of psoralidin obeyed M11 ), five (M1 , M2 , M3 , M5 and M9 ), nine (M1 –M5 , M8 –M11 ), ten
the typical monophasic Michaelis-Menten kinetics, as evidenced by (M1 –M4 , M6 –M11 ) and three (M1 , M3 and M8 ) metabolites of pso-
the Eadie-Hofstee plot (Fig. 6). The kinetic parameters (apparent Km ralidin were detected in HLMs, MLMs, PLMs, RAMs, RLMs and DLM,
and Vmax ) and calculated intrinsic clearance (CLint ) are summarized respectively. The data of psoralidin and its metabolites are listed
in Table 3. in Table 4, and the structural skeleton of psoralidin and its poten-
tial metabolic pathways are shown in Fig. 7. TOF/MS/MS spectra of
psoralidin and its metabolites are shown in Fig. 8. Psoralidin (m/z
335.0919) exhibited six characteristic fragment ions: m/z 319.0608
(loss of methyl), m/z 317.0447, m/z 280.0374 (loss of butenyl), m/z
260.8776, m/z 251.0349 (loss of butenyl and carbonyl) and m/z
146.9665 (Fig. 8A). Fragment ion at m/z 146.9665 was also found in
all mass spectra of metabolites and was inferred to be the parent
nucleus of psoralidin.
M1 . The molecular ion at m/z 353.1035 (M1 , [M+H2 O]) (Fig. 8B)
was detected at a retention time of 8.17 min, with the product ions
at m/z 293.0459, 280.0381, 251.0356 and 146.9666. The molecular
ion of M1 was 18 Da more than the m/z of the parent psoralidin;
therefore, it was identified as the hydration product of psoralidin.
According to the fragment ions of M1 and psoralidin at m/z 280,
251 and 146, and analyzing the fragment ion of M1 at m/z 293, a
hydration reaction occurred at the double bond between C-2 and
C-3 , and the hydroxyl group was bonded to C-3 .
M2 and M3 . M2 and M3 showed an m/z of 16 Da more than
the parent compound indicating that the two metabolites were
hydroxylated psoralidin. The cleaved pathways and fragment ions
Fig. 5. Inhibition assays of psoralidin metabolism by selective CYP450 inhibitors in
HLM. Results were mean ± SD at least 3 separate assays.
of M2 and M3 are shown in Fig. 8C and D.
X. Shi et al. / J. Chromatogr. B 1029 (2016) 145–156 151

Fig. 6. Michaelis-Menten plots for psoralidin in HLMs (A), RLMs (B), PLMs (C), MLMs (D), RAMs (E), DLMs (F) and CYP2C19 (G). The corresponding Eadie-Hofstee plots were
shown as insets. Each data point represents the mean of triplicate determinations.

Table 3
Kinetic parameters of P3 production from psoralidin in pooled human liver microsomes (HLMs), rat liver microsomes (RLMs), minipig liver microsomes (PLMs), monkey liver
microsomes (MLMs), rabbit liver microsomes (RAMs), dog liver microsomes (DLM) and recombinant CYP2C19.

Enzyme sources HLMa RLMa PLMa MLMa RAMa DLMa CYP2C19b

Vmax 16.27 ± 1.21 57.21 ± 3.18 40.42 ± 3.58 27.04 ± 1.56 34.76 ± 2.40 28.61 ± 2.1 3.01 ± 0.23
Km 5.171 ± 0.43 21.04 ± 1.53c 9.07 ± 0.65c 4.81 ± 0.43 4.41 ± 0.35 11.25 ± 0.93c 2.42 ± 0.15
CL(Vmax /Km ) 3.15 ± 0.22 2.72 ± 0.21 4.45 ± 0.31 5.61 ± 0.41 7.87 ± 0.59 2.54 ± 0.19 1.24 ± 0.14
a
Km , Vmax , and CLint were expressed in units of ␮M, nmol min−1 mg−1 protein, and mL min−1 mg−1 protein, respectively.
b
Km , Vmax , and CLint were expressed in units of ␮M, nmo min−1 pmol−1 CYP, and mL min−1 pmol−1 CYP, respectively.
c
Indicates Km value is statistical significant at P < 0.05 as compared to the Km value in HLM. All grouped data were expressed as means ± SD.

Fig. 7. Proposed chemical structures and major metabolic pathway of psoralidin in six species (including rat, monkey, dog, rabbit, minipig and human).
152 X. Shi et al. / J. Chromatogr. B 1029 (2016) 145–156

Fig. 8. Mass spectrum (MS/MS) of psoralidin and its metabolites.


X. Shi et al. / J. Chromatogr. B 1029 (2016) 145–156 153

M4 , M5 , M6 and M7 . M4 (tR = 6.82 min), M5 (tR = 7.39 min), M6

52.79
1.59

5.06

0.02
N.D.

N.D.
N.D.
N.D.
N.D.

N.D.
N.D.
N.D.
Dog
(tR = 7.52 min) and M7 (tR = 7.68 min) were isomers with the same
molecular formula of C20 H16 O7 (m/z 367.0818) and 32 Da more
than the parent compound psoralidin. Exact mass determination

Human

30.09
and the corresponding chemical composition showed the addition

2.29
0.34
6.80
0.16

0.24
0.03

0.03
0.01
N.D.

N.D.
N.D.
of two hydroxyl groups to the parent molecule. Structural formula
Rabbit and fragmentation pathway of M4 –M7 are shown in Fig. 8E–G.

42.56
M8 . The molecular formula of M8 was determined to be
2.09
0.28
5.80

0.11

0.12

0.11
0.40

0.05

0.06
N.D.
N.D.
C20 H18 O6 (m/z 353.1025) based on its MS data. According to its
molecular formula and MS/MS spectra, M8 was a reduction prod-
39.66
1.36

3.28
0.25

0.45

0.21
N.D.

N.D.
N.D.
N.D.

N.D.
N.D.
uct of M3 (2 Da more than M3 ), and two hydrogen were bonded to
Pig

C-2 and C-3 (Fig. 8H).


M9 . The mass spectrum of M9 (Fig. 8I) with a molecular ion at
Monkey

28.09

m/z 349.0712 was produced by a dehydrogenation reaction from


9.33
4.10

0.42
1.30
0.04

0.07
0.06
N.D.
N.D.
N.D.
N.D.
Peak area (%)

M3 . Psoralidin and M9 had the same fragment ions at m/z 321.0769


and 305.0464. Thus, the dehydrogenation reaction of M3 occurred
23.79

13.61

at C-4 and produced the M9 metabolite.


8.83

2.23

0.94
0.96
0.13
0.08

0.07
0.11
0.09
N.D.
Rat

M10 and M11 . M10 and M11 were isomers with the same molec-
ular formula (C20 H18 O7 ) but different retention times (6.30 and
Hydroxylation + Dehydrogenation

6.56 min). According to the MS/MS spectra and information on the


2 × Hydroxylation + Reduction
2 × Hydroxylation + Reduction

fragment ions, M10 and M11 were identified as the reduction and
Hydroxylation + Reduction

two hydroxylation products of psoralidin (Fig. 8J).


Metabolic reaction

2 × Hydroxylation
2 × Hydroxylation
2 × Hydroxylation
2 × Hydroxylation

3.7. The inference about P3 and M3


Hydroxylation
Hydroxylation

The HPLC, total ion and extracted ion chromatograms were


Hydration

showed in Fig. S6. P3 and P4 were observed in HPLC chromatogram,


parent
Accurate mass measurement for the protonated molecules of metabolites in rat, monkey, pig, rabbit, human and dog liver microsomes.

and M2 and M3 were found in total ion chromatogram. P3 , P4 ,


M2 and M3 were not found in control group. The retention time
of P3 (10.75 min) and P4 (11.02 min) in HPLC chromatogram was
319.0608, 317.0447, 280.0374, 279.0298, 260.8776, 146.9665
353.1035, 351.0880, 293.0459, 280.0381, 251.0359, 146.9666
333.0777, 321.0773, 317.0416, 305.0461, 280.0381, 146.9667

321.0767, 309.1137, 305.0455, 280.0383, 260.8782, 146.9667

same with M3 and M2 in total ion chromatogram, respectively.


321.0767,309.1137,305.0455,280.0383,260.8782,146.9667

The corresponding extracted ion chromatograms showed M2 and


M3 had same molecular ion at m/z 351. According to retention time
333.0776, 321.0766, 305.0454, 266.0236, 146.9664
349.0725, 317.0466, 307.0617, 260.8782, 146.9666

and peak area described in Table 4, the frontal peak in total ion
chromatogram was M3 and the latter peak was M2 . Thus, M3 was
inferred to be the metabolite P3 and M2 was inferred to be the
349.0725, 337.0726, 296.0322, 146.9667

294.0134, 280.0381, 279.0306, 146.9666


321.0769, 305.0464, 280,0373, 146.9664
325.1082, 280.0383, 240.0427, 146.9667
325.1082, 280.0383, 240.0427, 146.9667

metabolite P4 .

3.8. Molecular modelling

CYP2C19 is the enzyme involved in the metabolism of pso-


ralidin, and psoralidin showed strong inhibitory effects against
CYP1A2 and CYP2C8. Therefore, the interactions between pso-
ralidin and CYP1A2 or CYP2C8 at a molecular basis were
studied through molecular docking. Psoralidin bound within the
Fragment

hydrophobic pocket of CYP1A2 containing residues Val220, Phe226,


Phe260, Phe315, Gly316, Phe319, and Leu497 (Fig. 9A), within
the hydrogen-bonded and hydrophobic pocket of CYP2C8 con-
taining residues Ser103, Phe205, Ala297, Val366, Val477 and Ile
Error (ppm)

113 (Fig. 9B), and within the hydrogen-bonded and hydrophobic


pocket of CYP2C19 containing the residues Val113, Phe114, Ala297,
3.9
1.4
3.7

3.3
1.4

2.4
3.8
2.5
0.7
2.6
4.0

3.0

Thr301, Leu361, Ile362, Leu368, Phe476, and Ala477 (Fig. 9C).


335.0919
353.1039
351.0878
351.0871
367.0846

367.0846
367.0846
353.1035
349.0717
369.0841
369.0841
367.0808

4. Discussion
Mass

CYP450 and UGT are two imperative enzymes responsible for


the metabolism of most drugs. The inhibition of these two enzyme
C20 H16 O5
C20 H18 O6
C20 H16 O6
C20 H16 O6
C20 H16 O7
C20 H16 O7
C20 H16 O7
C20 H16 O7
C20 H18 O6
C20 H14 O6
C20 H18 O7
C20 H18 O7
Formula

systems could result in drug-drug interactions and severe adverse


side effects. As a conservative approach, the inhibitor Cmax at the
highest clinical dose expected should be used in the estimation of
AUC change. Both the Food and Drug Administration and European
tR (min)

Medicines Agency stated that an interaction would likely occur for


N.D., No detect.
8.76
8.17
7.74
7.24
6.82
7.39
7.52
7.68
7.74
7.82

6.56
6.30

reversible inhibition if the ratio of inhibitor Cmax /Ki was greater


than 1. Interactions are possible if the ratio is between 0.1 and 1 and
Table 4

M10
M11
NO.

remote if below 0.1 [28]. Using UGT1A1-overexpressing HeLa cells,


M0
M1
M2
M3
M4
M5
M6
M7
M8
M9

Sun et al. found that psoralidin showed strong inhibition against


154 X. Shi et al. / J. Chromatogr. B 1029 (2016) 145–156

Fig. 9. Binding modes of compound with CYP 1A2 (A), 2C8 (B) and 2C19 (C). The compounds were displayed in green sticks, residues were displayed in blue thin sticks,
hydrogen bonds were displayed in green dot lines, and hydrophobic interactions were displayed in pink dot line. (For interpretation of the references to colour in this figure
legend, the reader is referred to the web version of this article.)

UGT1A1 [29]. In this study, we found psoralidin strongly inhibited for drug metabolism and pharmacokinetics studies of psoralidin.
the activities of UGT1A1, UGT1A7, CYP1A2 and CYP2C8 with the This conclusion agrees well with previous publications in which
calculated Cmax /Ki values of1.15, 27.12, 5.67, and 26.21, respec- monkeys are reported to be closely related to humans and share
tively. The results of the in vitro-in vivo extrapolation were well a common ancestor from roughly 25 million years ago. Because
above the thresholds for clinical DDI evaluation as recommended they are genetically and physiologically similar to humans, mon-
in the regulatory guideline. This indicated that the most likely in keys are the most widely used nonhuman primate in basic and
vivo adverse effects are due to the large Cmax /Ki values of pso- applied biomedical research [37].
ralidin against UGT1A1, UGT1A7, CYP1A2 and CYP2C8. CYP1A2, Wang and Geng found that psoralidin produced 13 metabolites
CYP2C8, UGT1A1 and UGT1A7 are particularly significant enzymes by cyclization, dehydration, hydroxylation and glucuronidation
because they are involved in the metabolism of many important reaction [38,39], 9 of which metabolites were not identified in our
drugs and endogenous substances such as bilirubin, raltegravir, study. However, we identified 7 metabolites (M3, M5, M7–M11)
caffeine and paclitaxel [30–33]. Thus, potential drug-drug inter- that were not reported by them. The inconsistent between our
actions should be monitored when psoralidin is co-administered and previous studies may be caused by differences in the selec-
with clinical medications. tion of model.Compared with their studies using rats as model,
We identified the CYP450 isoforms involved in psoralidin livermicrosomes from different species were used to simulate the
metabolism by screening assays with recombinant CYP450 super- in vivo metabolism of psoralidin in our study which can identify
somes including CYP1A2, CYP3A4, CYP2E1, CYP2C9, CYP2D6 and the metabolic differences between different species. Additionally,
CYP2C19 and chemical inhibition studies. CYP2C19 played a vital Sun et al.resported that psoralidin generated one or two monoglu-
role in the metabolism of psoralidin, while all other CYP isoforms curonides in HLMs and UGT1A9 was responsible for 99.6% of
displayed an insignificant effect compared to CYP2C19. CYP2C19 is psoralidin-3-O-glucuronidation [40]. The monoglucuronides did
one of important CYP450 isoforms, which catalyzes the metabolic not be detected in our experiment. Due to the purpose of our study
reaction of many drugs such as R-warfarin [34]. In addition, the that more and non-target metabolites of psoralidin were expected
Km value of psoralidin in CYP2C19 was similar to its Km value in to explore, the data acquiring and ananlysis method developing
HLMs which further indicated that CYP2C19 was the major enzyme would not be most suitable for monoglucuronides of psoralidin. Our
involved in its metabolism. These results demonstrated that pso- results were valuable and helpful for understanding and depicting
ralidin was selectively catalyzed by CYP2C19 in humans. the metabolism profiles of psoralidin comprehensively. Further-
In preclinical studies to evaluate pharmacokinetics and toxic- more, structural skeleton and potential metabolic pathways of
ity, animal models have been commonly used to replace humans. psoralidin and its metabolites among different species were deter-
However, it should be understood that suitable animal models for mined and analyzed. Metabolites of psoralidin yielded in DLMs
in vivo drug metabolism and pharmacokinetics studies are impor- were less than those in other species, which was consistent with
tant and should have similar metabolic behaviors as compared to the results determined by HPLC-UV. M1 and M3 were found in
humans. The models should include identical metabolic profiles, all species and their peak areas were more than 1%, which indi-
equivalent or similar metabolic enzymes, and comparable catalytic cated that hydration and hydroxylation were the major metabolic
efficiency. Consequently, we distinguished the in vitro metabolism transformations of psoralidin. In particular, M3 had biggest peak
behaviors of psoralidinin HLMs, RLMs, DLMs, RAM, MLMs and PLMs. area among the metabolites. Furthermore, we inferred M3 was
The kinetic parameters including Km , Vmax , and CLint for the forma- the metabolite P3 described in Fig. 2. In this study, all metabolites
tion of P3 were determined and varied among species.Our findings were only measured by mass spectrometry, and more spectromet-
also revealed significant differences in the metabolism of psora- ric methods (i.e., NMR) are warranted in the future.
lidin between species. The Km values in DLMs, RLMs and PLMs Molecular modeling studies were carried out to explore the
were much higher than those in HLMs and all the differences are interactions between psoralidin and CYP1A2, CYP2C8 or CYP2C19
statistically significant (all P < 0.01). In comparison, Km values in to further understand the key residues involved in binding
MLMs were comparableto the Km values of HLMs. The Km values in and recognition between the substrate and enzymes. Psora-
HLMs and MLMs indicated comparable binding affinities towards lidin interacted with CYP1A2 exclusively through hydrophobic
the metabolic site and reflected species similarities [35,36]. Simul- interactions, while it interacted with CYP2C8 through hydrogen
taneously, the chromatogram and P3 peak area of monkeys shown bonds and hydrophobic interactions. The hydrogen bonds formed
in Fig. 2 was also similar to humans. These results indicated that between CYP2C8 and psoralidin imply that psoralidin exhibited
monkeys might be a suitable choice to serve as a surrogate model stronger interactions with CYP2C8 than CYP1A2. These molecular
X. Shi et al. / J. Chromatogr. B 1029 (2016) 145–156 155

modeling findings also agree with the above-mentioned experi- using liquid chromatography with tandem mass spectrometry, J. Chromatogr.
mental results in which psoralidin had lower IC50 and Ki values B Analyt. Technol. Biomed. Life Sci. 1020 (2016) 111–119.
[12] J.J. Kiser, J.R. Burton, G.T. Everson, Drug-drug interactions during antiviral
for CYP2C8 than CYP1A2. In addition, psoralidin also interacted therapy for chronic hepatitis C, Nat. Rev. Gastroenterol. Hepatol. 10 (2013)
with CYP2C19 through hydrogen bonding and hydrophobic inter- 596–606.
actions. These results support the high binding affinity between [13] V. Garg, R. van Heeswijk, J.E. Lee, K. Alves, P. Nadkarni, X. Luo, Effect of
telaprevir on the pharmacokinetics of cyclosporine and tacrolimus,
CYP2C19 and psoralidin and that CYP2C19 was the major enzyme Hepatology 54 (2011) 20–27.
participating in psoralidin metabolism. [14] C. Lu, A. Suri, W.C. Shyu, S. Prakash, Assessment of cytochrome P450-mediated
drug–drug interaction potential of orteronel and exposure changes in
patients with renal impairment using physiologically based pharmacokinetic
5. Conclusion modeling and simulation, Biopharm. Drug Dispos. 35 (2014) 543–552.
[15] C. Lecefel, P. Eloy, B. Chauvin, B. Wyplosz, V. Amilien, L. Massias, A.M. Taburet,
Psoralidin exhibited potent inhibition against UGT1A1, UGT1A7, H. Francois, Worsening pneumonitis due to a pharmacokinetic drug-drug
interaction between everolimus and voriconazole in a renal transplant
CYP1A2 and CYP2C8, which may lead to drug-drug interactions. patient, J. Clin. Pharm. Ther. 40 (2015) 119–120.
Eleven metabolites were observed in the liver microsome incu- [16] Y. Liu, T.J. Flynn, CYP3A4 inhibition by Psoraleacorylifolia and its major
bation system. CYP2C19 was the major isozyme involved in the components in human recombinant enzyme differentiated human hepatoma
HuH-7 and HepaRG cells, Toxicol. Rep. 2 (2015) 530–534.
metabolism of psoralidin. Monkeys possessed the metabolic capac-
[17] D. Gao, X. Chen, X. Yang, Q. Wu, F. Jin, H. Wen, Y. Jiang, H. Liu, Stable isotope
ity that is most similar to humans and could be used as the animal labeling strategy for curcumin metabolite study in human liver
selection in advanced toxicology or pharmacokinetic studies of pso- microsomesby Liquid Chromatography–Tandem Mass Spectrometry, J. Am.
ralidin in the future. Soc. Mass Spectrom. 26 (2015) 686–694.
[18] Z.Z. Fang, K.W. Krausz, K. Nagaoka, N. Tanaka, K. Gowda, S.G. Amin, G.H.
Perdew, F.J. Gonzalez, In vivo effects of the pure aryl hydrocarbon receptor
Conflict of interests antagonist GNF-351 after oral administration are limited to the
gastrointestinal tract, Br. J. Pharmacol. 171 (2014) 1735–1746.
[19] V. Dohnal, Q. Wu, K. Kuča, Metabolism of aflatoxins: key enzymes and
None. interindividual as well as interspecies differences, Arch. Toxicol. 88 (2014)
1635–1644.
[20] J. Zielinski, Inhibition of in vitro metabolism of testosterone in human, dog
Acknowledgment
and horse liver microsomes to investigate species differences, Toxicol. In
Vitro 29 (2015) 468–478.
We acknowledged that financially supported fromthe Natural [21] P.K. Smith, R.I. Krohn, G.T. Hermanson, A.K. Mallia, F.H. Gartner, M.D.
Science Foundation of Liaoning Province (Project No. 2014022005). Provenzano, E.K. Fujimoto, N.M. Goeke, B.J. Olson, D.C. Klenk, Measurement of
protein using bicinchoninic acid, Anal. Biochem. 75 (1976) 596–603.
[22] Y. Liu, H. Ma, J.W. Zhang, M.C. Den, L. Yang, Influence of ginsenoside Gh1 and
Appendix A. Supplementary data F1 on human cytochrome P450 enzymes, Planta Med. 72 (2006) 126–131.
[23] X.Y. Qi, S.C. Liang, G.B. Ge, Y. Liu, P.P. Dong, J.W. Zhang, A.X. Wang, J. Hou, L.L.
Zhu, L. Yang, C.X. Tu, Inhibitory effects of sanguinarine on human liver
Supplementary data associated with this article can be found, cytochrome P450 enzymes, Food Chem. Toxicol. 56 (2013) 392–397.
in the online version, at http://dx.doi.org/10.1016/j.jchromb.2016. [24] Z.Z. Fang, Y.Y. Zhang, G.B. Ge, H. Huo, S.C. Liang, L. Yang, Time-dependent
06.031. inhibition (TDI) of CYP3A4 and CYP2C9 by noscapine potentially explains
clinical noscapine-warfarin interaction, Br. J. Clin. Pharmacol. 69 (2010)
193–199.
References [25] H.Y. Ma, D.X. Sun, Y.F. Cao, C.Z. Ai, Y.Q. Qu, C.M. Hu, C. Jiang, P.P. Dong, X.Y.
Sun, M. Hong, N. Tanaka, F. Gonzalez, X.C. Ma, Z.Z. Fang, Herb-drug interaction
[1] N.S. Amit, M. Nutan, P.F. Devanand, Determination of isoflavone content and prediction based on the high specific inhibition of andrographolide
antioxidant activity in Psoraleacorylifolia L. callus cultures, Food Chem. 118 derivatives towards UDP-glucuronosyltransferase (UGT) 2B7, Toxicol. Appl.
(2010) 128–132. Pharmacol. 277 (2014) 86–94.
[2] H. Katsura, R.I. Tsukiyama, A. Suzuki, M. Kobayashi, In vitro antimicrobial [26] J.W. Zhang, Y. Liu, W. Li, D.C. Hao, L. Yang, Inhibitory effect of
activities of bakuchiol against oral microorganisms, Antimicrob. Agents medroxyprogesterone acetate on human liver cytochrome P450 enzymes,
Chemother. 45 (2001) 3009–3013. Eur. J. Clin. Pharmacol. 62 (2006) 497–502.
[3] P.G. Latha, D.A. Evans, K.R. Panikkar, K.K. Jayavardhanan, Immunomodulatory [27] D.X. Sun, Z.Z. Fang, Y.Y. Zhang, Y.F. Cao, L. Yang, J. Yin, Inhibtory effects of
and antitumour properties of Psoraleacorylifolia seeds, Fitoterapia 71 (2000) curcumenol on human liver cytochrome P450 enzymes, Phytother. Res. 24
223–231. (2010) 1213–1216.
[4] R. Kumar, S. Srinivasan, P. Pahari, J. Rohr, C. Damodaran, Activating [28] M. Jarratt, W.J. Siu, E. Yamakawa, N. Kodera, R. Pillai, K. Smith, Safety and
stress-activated protein kinase–mediated cell death and inhibiting epidermal pharmacokinetics of efinaconazole 10% solution in healthy volunteers and
growth factor receptor signaling: a promising therapeutic strategy for patients with severe onychomycosis, J. Drugs Dermatol. 12 (2013) 1010–1016.
prostate cancer, Mol. Cancer Ther. 9 (2010) 2488–2496. [29] H. Sun, X. Zhou, B. Wu, Accurate identification of
[5] Y.C. Chen, Y.T. Cheung, L.D. Kong, T.B. Ng, C.F. Qiao, S.F. Mo, H.X. Xu, H.F. Kung, UDP-glucuronosyltransferase 1A1 (UGT1A1) inhibitors using
Transcriptional regulation of corticotrophin releasing factor gene by UGT1A1-overexpressing HeLa cells, Xenobiotica 45 (2015) 945–953.
furocoumarins isolated from seeds of Psoraleacorylifolia, Life Sci. 82 (2008) [30] M.Y. Lee, M. Apellániz-Ruiz, I. Johansson, S. Vikingsson, T.K. Bergmann, K.
1117–1121. Brøsen, H. Green, C. Rodríguez-Antona, M. Ingelman-Sundberg, Role of
[6] M. Chino, K. Sato, T. Yamazaki, T. Maitani, Constituent of natural food additive cytochrome P450 2C8*3 (CYP2C8*3) in paclitaxel metabolism and
hokosshi extract and an analytical method for the additive in foods, Shokuhin paclitaxel-induced neurotoxicity, Pharmacogenomics 16 (2015) 929–937.
Eiseigaku Zasshi 43 (2002) 352–355. [31] E. Schievano, C. Finotello, L. Navarini, S. Mammi, Quantification of caffeine in
[7] L.T. Yi, Y.C. Li, Y. Pan, H.M. Li, Q. Xu, S.F. Mo, C.F. Qiao, F.X. Jiang, H.X. Xu, X.B. human saliva by nuclear magnetic resonance as an alternative method for
Lu, L.D. Kong, H.F. Kung, Antidpressant-like effects of psoralidin isolated from cytochrome CYP1A2 phenotyping, Talanta 140 (2015) 36–41.
the seeds of PsoraleaCorylifolia in the forced swimming test in mice, Prog. [32] J.B. Valenzuela, S.M. Gonzalez, O.V. Escudero, N.E. Martinez, R.C. Perez, L.J.
Neuropsychopharmacol. Biol. Psychiatry 32 (2008) 510–519. Rebollo, M.R. Gonzalez, R.J.J. Perez, Influence of genetic polymorphisms in
[8] W.Y. Foo, H.Y. Tay, E.C. Chan, A.J. Lau, Meclizine, a pregnane X receptor UGT1A1, UGT1A7 and UGT1A9 on the pharmacokynetics of irinotecan SN-38
agonist, is a direct inhibitor andmechanism-based inactivator of human and SN-38G, Farm. Hosp. 37 (2014) 111–127.
cytochrome P450 3A, Biochem. Pharmacol. 97 (2015) 320–330. [33] B.B. Xu, Song. Gao, B.J. Wu, T.H. Yin, M. Hu, Absolute quantification of UGT1A1
[9] L. Qiang, L.P. Wang, P.M. Dai, X.J. Zeng, X.X. Qi, L.J. Zhu, T.M. Yan, Y. Wang, L.L. in various tissues and cell lines using isotope label-free UPLC–MS/MS method
Lu, M. Hu, X.C. Wang, Z.Q. Liu, A combined strategy of mass fragmentation, determines its turnover number and correlates with its glucuronidation
post-column cobalt complexation and shift in ultraviolet absorption spectra activities, J. Pharm. Biomed. Anal. 88 (2014) 180–190.
to determine the uridine 5 -diphospho-glucuronosyltransferase metabolism [34] M. Utoh, T. Yoshikawa, Y. Hayashi, M. Shimizu, K. Iwasaki, Y. Uno, H. Yamazaki,
profiling of flavones after oral administration of a flavone mixture in rats, J. Slow R-warfarin 7-hydroxylation mediated by P450 2C19 geneticvariants in
Chromatogr. A 1395 (2015) 116–128. cynomolgus monkeys in vivo, Biochem. Pharmacol. 95 (2015) 110–114.
[10] V.B. Urlacher, M. Girhard, Cytochrome P450 monooxygenases: an update on [35] Y.Y. Zhang, Y. Liu, J.W. Zhang, G.B. Ge, H.X. Liu, L.M. Wang, J. Sun, L. Yang, C-7
perspectives for synthetic application, Trends Biotechnol. 30 (2012) 26–36. configuration as one of determinants in taxanes metabolism by human
[11] X. Liang, M. Van Parys, X. Ding, N. Zheng, L. Bi, D. Dorshort, J. McKnight, D. cytochrome P450 enzymes, Xenobiotica 39 (2009) 903–914.
Milanowski, J. Mao, Y. Chen, J.A. Ware, B. Dean, C.E. Hop, Y. Deng, [36] Y.F. Cao, Y.Y. Zhang, J. Li, G.B. Ge, D. Hu, H.X. Liu, T. Huang, Y.C. Wang, Z.Z. Fang,
Simultaneous determination of itraconazole, hydroxyitraconazole, keto D.X. Sun, H. Huo, J. Yin, L. Yang, CYP3A catalysesschizandrin biotransformation
itraconazole and N-desalkylitraconazole concentration in human plasma inhuman, minipig and rat liver microsomes, Xenobiotica 40 (2010) 38–47.
156 X. Shi et al. / J. Chromatogr. B 1029 (2016) 145–156

[37] R.A. Gibbs, J. Rogers, M.G. Katze, R. Bumgarner, G.M. Weinstock, E.R. Mardis, [39] H. Sun, Z. Ma, D. Lu, B. Wu, Regio- and isoform-specific glucuronidation of
et al., Evolutionary and biomedical insights from the rhesus macaque psoralidin: evaluation of 3-O-glucuronidation as a functional marker for
genome, Science 316 (2007) 222–234. UGT1A9, J. Pharm. Sci. 104 (2015) 2369–2377.
[38] J.L. Geng, Y. Dai, Z.H. Yao, Z.F. Qin, X.L. Wang, L. Qin, X.S. Yao, Metabolites [40] P.L. Wang, Z.H. Yao, F.X. Zhang, X.Y. Shen, Y. Dai, L. Qin, X.S. Yao, Identification
profile of Xian-Ling-Gu-Bao capsule a traditional Chinese medicine of metabolites of PSORALEAE FRUCTUS in rats by ultra performance liquid
prescription, in rats by ultra performance liquid chromatography coupled chromatography coupled with quadrupole time-of-flight tandem mass
with quadrupole time-of-flight tandem mass spectrometry analysis, J. Pharm. spectrometry analysis, J. Pharm. Biomed. Anal. 112 (2015) 23–35.
Biomed. Anal. 96 (2014) 90–103.

Вам также может понравиться