Subscriber access provided by Oregon Health & Science University Library
New Analytical Methods
Simultaneous determination of oleanolic acid and ursolic acid by in vivo microdialysis via UHPLC-MS/MS using magnetic dispersive solid phase extraction coupling with microwave-assisted derivatization and its application to a pharmacokinetic study of Arctiumlappa L. root extract in rats Zhenjia Zheng, Xian-En Zhao, Shuyun Zhu, Jun Dang, Xuguang Qiao, Zhichang Qiu, and Yanduo Tao J. Agric. Food Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jafc.7b06015 • Publication Date (Web): 21 Mar 2018 Downloaded from http://pubs.acs.org on March 21, 2018
Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.
is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.
Page 1 of 28
Journal of Agricultural and Food Chemistry
Simultaneous determination of oleanolic acid and ursolic acid by in vivo microdialysis via UHPLC-MS/MS using magnetic dispersive solid phase extraction coupling with microwave-assisted derivatization and its application to a pharmacokinetic study of Arctiumlappa L. root extract in rats Zhenjia Zheng1, Xian-En Zhao3*, Shuyun Zhu3*, Jun Dang2, Xuguang Qiao1**, Zhichang Qiu1, Yanduo Tao2 1
College of Food Science and Engineering, Shandong Agricultural University, 61 Daizong Street,
Taian 271018, Shandong, P.R. China; 2
Qinghai Provincial Key Laboratory of Tibetan Medicine Research & Key Laboratory of Tibetan
Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Science, Xining 810001, Qinghai, P.R. China; 3
College of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165,
Shandong, P.R. China
1
ACS Paragon Plus Environment
Journal of Agricultural and Food Chemistry
1
ABSTRACT
2
Simultaneous detection of oleanolic acid and ursolic acid in rat blood by in vivo microdialysis can
3
provide important pharmacokinetics information. Microwave-assisted derivatization coupled with
4
magnetic dispersive solid phase extraction was established for the determination of oleanolic acid
5
and ursolic acid by liquid chromatography tandem mass spectrometry. 2’-Carbonyl-piperazine
6
rhodamine B was firstly designed and synthesized as the derivatization reagent, which was easily
7
adsorbed onto the surface of Fe3O4/graphene oxide. Simultaneous derivatization and extraction of
8
oleanolic acid and ursolic acid were performed on Fe3O4/graphene oxide. The permanent positive
9
charge of the derivatization reagent significantly improved the ionization efficiencies. The limits
10
of detection were 0.025 and 0.020 ng/mL for oleanolic acid and ursolic acid, respectively. The
11
validated method was shown to be promising for sensitive, accurate and simultaneous
12
determination of oleanolic acid and ursolic acid. It was used for their pharmacokinetics study in
13
rat blood after oral administration of Arctiumlappa L. root extract.
14
KEYWORDS: In vivo microdialysis, triterpenic acid, microwave-assisted derivatization,
15
magnetic graphene oxide, pharmacokinetics, Arctiumlappa L. root.
16 17 18 19 20 21 22 23 24 25
2
ACS Paragon Plus Environment
Page 2 of 28
Page 3 of 28
Journal of Agricultural and Food Chemistry
26
INTRODUCTION
27
Oleanolic acid (OA) and ursolic acid (UA) belong to triterpenoid compounds that widely exist in
28
herbs and fruits. They have been reported to have important pharmacological properties, such as
29
anti-inflammatory,
30
antihyperlipidemic activities and so on.1,2 Therefore, a sensitive and rapid analytical method is
31
necessary and helpful for their pharmacokinetic study to better understand the pharmacological
32
activity of related foods and herbs.
hepatoprotective,
antiulcer,
antimicrobial,
antitumour,
anti-HIV,
33
OA and UA are triterpine isomers with exactly the same chemical structures, with the only
34
difference found in the position of a methyl group in the E ring (Figure 1), thus they are difficult
35
to separate and detect rapidly.3-5 A lot of methods have been reported for the determination of OA
36
and UA, such as gas chromatography (GC),6 thin-layer chromatography (TLC),7 capillary
37
electrophoresis (CE),8 high-performance liquid chromatography (HPLC) with UV,9 fluorescence
38
detection,3 mass spectrometry (MS) 4, 5 or nuclear magnetic resonance (NMR).10 Each method has
39
its own feature, but many of them show a limited enhancement on the sensitivity, accuracy and
40
specificity. In the past decade, ultra high performance liquid chromatography with tandem mass
41
spectrometry (UHPLC-MS/MS) in the multiple reaction monitoring mode (MRM) has aroused
42
wide attention in rapid pharmaceutical analysis and bioanalysis in different biological matrices.11
43
However, very low concentrations and strong matrix interferences in real samples usually make
44
difficult the sensitive and accurate determination of compounds. Therefore, sensitive, accurate,
45
rapid and simultaneous determination of OA and UA is still a challenging task. However, OA and
46
UA lack a chromophore and cannot easily gain a charge because of their carboxyl group, thus
47
they provide a very low sensitivity in relation to UV, fluorescence and MS detection. Under these
48
circumstances, sensitivity enhancement by chemical derivatization can solve problems.12, 13 Some
49
derivatization reagents have been reported for them by HPLC fluorescence detection
50
GC-MS.19 However, there are almost no synthesized derivatization reagents for the enhanced
3
ACS Paragon Plus Environment
14-18
and
Journal of Agricultural and Food Chemistry
51
UHPLC-MS/MS determination of OA and UA.
52
However, the UHPLC-MS/MS detection sensitivity is frequently compromised by the low
53
contents of analytes and serious matrix effect from real samples and derivatization procedure.
54
Therefore, efficient sample pretreatment procedure is necessary.20,21 Compared to the popular
55
liquid liquid extraction (LLE)
56
extraction (d-SPE) is time saving, easy to operate and low consumed of toxic organic solvents.
57
Especially, the use of magnetic sorbents for magnetic dispersive solid phase extraction (MDSPE)
58
has drawn significant attention because of their excellent dispersibility and ease of separation by
59
an external magnetic field.25,
60
surface area-to-volume ratio, strong π-π stacking interactions and high mechanical strength,25, 27,
61
28
62
analytes depending on the hydrogen bonding, hydrophobic interactions, van der Waals forces and
63
electrostatic forces.29
11,12,22
26
and solid phase extraction (SPE),23,24 dispersive solid-phase
Graphene oxide (GO), a carbon-based material with a high
presents enormous advantages in the separation and determination of small amount of organic
64
In vivo microdialysis sampling is a preeminent technique for neuroscience, pharmacokinetic
65
(PK), pharmacodynamic (PD) and clinical studies.30 It is commonly used for investigation of
66
PK/PD since concentration of the unbound drug is more correlated to the pharmacological effects.
67
By coupling with powerful analytical methods, in vivo microdialysis is able to overcome several
68
disadvantages of conventional pharmacokinetic techniques which include continuous sampling in
69
the same animal, minimizing the number of animals used and also minimizing inter-animal
70
variation. However, drug quantification in microdialysis technique remains a major challenge
71
because of the low concentration and the small volume of microdialysate. Therefore, a highly
72
sensitive, accurate and selective analytical method employing UHPLC-MS/MS (MRM) is always
73
recommended.31,32
74
In this study, a new method based on in vivo microdialysis by microwave-assisted
75
derivatization coupling with MDSPE (MAD-MDSPE) in a single step has been developed for the
76
simultaneous determination of OA and UA by UHPLC-MS/MS. 2’-Carbonyl-piperazine 4
ACS Paragon Plus Environment
Page 4 of 28
Page 5 of 28
Journal of Agricultural and Food Chemistry
77
rhodamine B (CPR) was designed and synthesized as derivatization reagent for the labeling of
78
carboxyl group of OA and UA. This method was used for the simultaneous pharmacokinetic
79
study of OA and UA in rat blood.
80 81
MATERIALS AND METHODS
82
Chemicals and Reagents. OA, UA and the internal standard (IS) betulinic acid were
83
purchased from National institute for the control of pharmaceutical and biological products
84
(Beijing, China). 1-Ethyl-3-(3-dimethylaminopropyl)-carbodiimide hydrochloride (EDC-HCl)
85
and the HPLC grade formic acid were purchased from Sigma Co. (St. Louis, MO, USA). HPLC
86
grade acetonitrile and methanol were purchased from Fisher Scientific Co. (Fair Lawn, NJ, USA).
87
N,N-dimethylformamide (DMF) and pyridine was of analytical grade and obtained from Tianjin
88
Guangcheng Chemical Reagent Co. (Tianjin, China). Pure water was obtained on a Millipore
89
system (Bedford, MA, USA). All other reagents used were of HPLC grade or at least of
90
analytical grade obtained commercially.
91
Stock solutions of OA (10.0 µmol/L), UA (10.0 µmol/L) and betulinic acid (IS, 10.0
92
µmol/L), and derivatization reagent CPR (100.0 µmol/L) were prepared by HPLC grade
93
acetonitrile. All working solutions with different concentrations were prepared by diluting
94
corresponding stock solutions with acetonitrile. Solution of 0.10 mol/L coupling reagent EDC
95
was prepared in HPLC acetonitrile. The quality control samples (QCs) containing OA (0.5, 5.0,
96
100 ng/mL) and UA (0.5, 5.0, 100 ng/mL) were prepared at three concentration levels by adding
97
appropriate working standard solutions to drug-free rat microdialysates. When not in use, all the
98
solutions were stored at 4 °C.
99
Instrumentation. The UHPLC-MS/MS system consisted of an Agilent 1290 UHPLC
100
system and an Agilent 6460 Triple Quadrupole MS/MS system (Agilent, USA). The
101
chromatographic separation was realized on an Agilent SB C18 column (2.1 mm × 50 mm, 1.8 5
ACS Paragon Plus Environment
Journal of Agricultural and Food Chemistry
102
µm) at 30 °C column temperature with 2.0 µL injection volumes. The flow rate was constant at
103
0.2 mL/min. Eluent A was 5% acetonitrile/water (0.1% formic acid) and B was acetonitrile (0.1%
104
formic acid). The linear binary gradient elution conditions were as follows: 65-82% B from 0 to 2
105
min; 82-98% B from 2 to 6 min; 98-100% B from 6 to 8 min. During 0-5.5 min after injection the
106
flow was diverted into waste to protect the mass spectrometer from potential contaminations
107
because there were no detectable analytes. The column was equilibrated using the initial mobile
108
phase for 1.5 min for each injection. The mass spectrometer was run in positive ion MRM mode
109
of electrospray source. The optimal MS conditions were the same as our recent report in 2016.11
110
The fragmentor voltage (FV) and collision energy (CE) were also optimized for the target
111
derivatives. Experimental conditions for the direct MRM detection of OA and UA were set
112
according to the literature.4 Transmission electron microscope (TEM) images were obtained
113
using the JEM-2100PLUS microscope (JEOL, Tokyo, Japan).
114
In vivo microdialysis sampling was accomplished using in vivo microdialysis system from
115
Sweden CMA Co., including a CMA 402 syringe pump (CMA, Solna, Sweden), a CMA 120
116
system (CMA, Solna, Sweden) for freely moving animals, and a microdialysis MAB6 probe
117
(Stockholm, Sweden). ASI stereotaxic flat skull coordinates were purchased from ASI
118
Instruments Inc. (MI, USA). The probe was perfused with Ringer’s solution (5 mmol/L) at a flow
119
rate of 2.0 µL/min.
120
Synthesis of CPR. The synthesis reaction schematic of CPR was shown in Figure 1.
121
The synthesis of raw material N-hydroxysuccinimidyl rhodamine B ester (RB-S) was carried out
122
as our previous report.33 The synthesis procedure was as follows: RB-S (1.5 g) and 0.5 g
123
piperazine were added into 50 mL of acetonitrile and 25 mL sodium bicarbonate buffer (pH 8.5),
124
the solution was heated to 45 oC with continued agitation for 2 h. After reduced-pressure
125
distillation of the solvent and recrystallization in dichloroethane/absolute ethanol (v:v, 1:1), CPR
126
was obtained as dark red crystals with a yield of 55%. 1HNMR (500 MHz, CDCl3/δ, ppm): 7.69 (t, 6
ACS Paragon Plus Environment
Page 6 of 28
Page 7 of 28
Journal of Agricultural and Food Chemistry
127
J = 9.7 Hz, 2H), 7.61 (d, J = 13.5 Hz, 1H), 7.34 (d, J = 6.9 Hz, 1H), 7.19 (d, J = 9.5 Hz, 2H), 6.98
128
(d, J = 9.1 Hz, 2H), 6.75 (s, 2H), 3.82 – 3.55 (m, 12H), 3.03 (d, J = 34.4 Hz, 3H), 1.33 (t, J = 7.0
129
Hz, 11H). HRMS: [M+H]+ 511.30692.
130
Preparation of Fe3O4/GO. The preparation of graphene oxide was described in
131
Supporting information according to the Hummers method with minor modifications.34
132
Fe3O4/GO was prepared based on chemical coprecipitation of Fe2+ and Fe3+ in alkaline media in
133
presence of GO as described in Supporting information.35
134
MDSPE-MAD procedure. Fe3O4/GO (8 mg) was put into a 1.5 mL vial and then 30
135
µL of mixed standards or microdialysates, 30 µL of EDC (0.1 mol/L), 200 µL of CPR reagent
136
were added. The vial was sealed and immersed in the ultrasound bath for 2.0 min to form a
137
homogeneous dispersed solution. This solution was radiated for 25 min in a microwave reactor
138
(450 W) at 60 °C to achieve complete MDSPE-MAD procedure. The derivatization reaction
139
scheme is shown in Figure 1. And then the magnetic materials were separated rapidly from the
140
derivatization solution by an external magnet. After that, CPR derivatives were eluted from the
141
magnetic materials by 300 µL of methanol (containing 1.0 % formic acid) under ultrasound for 1
142
min, and 2.0 µL of the solution was analyzed by UHPLC-MS/MS.
143
Preparation of Arctiumlappa L. root extract. Dried Arctiumlappa L. root was
144
purchased from China Beijing Tongrentang (Group) Co., Ltd. (Beijing). To 50 g of powdered raw
145
herb (250 µm) in a 500 mL flask, 300 mL of ethanol was added. The mixture was extracted 2
146
times by ethanol refluxing with 6 volume equivalents. The combined extracts were added with 20
147
mL of 30% β-cyclodextrin to increase the solubility, and then concentrated to 10 mL under
148
vacuum to obtain herb solution for rats. The OA and UA contents in the extract were
149
quantitatively determined by the developed method in this work. The contents of free OA and UA
150
in the extract were 673.8 and 526.4 µg/g. After saponification reaction with 10% potassium
151
hydroxide at 90 °C for 3 h, the conjugated OA and UA in the extract were determined and their 7
ACS Paragon Plus Environment
Journal of Agricultural and Food Chemistry
152
concentrations were 1438.5 and 1142.2 µg/g.
153
Pharmacokinetics of OA and UA in rats by in vivo microdialysis. Male
154
Sprague-Dawley rats (200-220 g, n=6) were purchased from Shandong Lukang Pharmaceutical
155
Co. Ltd. The care and use of animals were in accordance with the related principles of China.
156
Rats were narcotized with 20% urethane (1.2 g/kg, i.p.) before surgery, and kept anesthesis in the
157
whole surgery of MAB probe implantation. After recovering, rats were free drinking and food
158
intake for 24 h, and then fasted for 12 h with free drinking water before the pharmacokinetics test.
159
Rats were orally administrated with extracts at a dose of 1.0 g/kg body weight. In vivo
160
microdialysate samples from rat carotid artery were collected at 15, 30, 45, 60, 80, 100, 120, 180,
161
240, 360, 480 and 600 min after oral administration. Each 30 µL of microdialysates were used for
162
MAD-MDSPE procedure and UHPLC-MS/MS analysis.
163 164
RESULTS AND DISCUSSION
165
Optimization of Chromatography and MS Conditions. UHPLC and MS
166
conditions were optimized for obtaining the best chromatographic separation and maximum MS
167
sensitivity. The chromatographic conditions optimization was similar to our previous study.11 The
168
optimum conditions were described in the experimental section. The representative MRM
169
chromatograms of CPR-derivatives for standards and internal standard were shown in Figure 2A.
170
MS conditions were also optimized similar to our previous study.11 All the CPR derivatives
171
showed intense [M+H]+ ions. They were set as precursor ions. The most abundant product ions
172
for three CPR derivatives were m/z 398.8 and m/z 443.2. In ESI-MS/MS conditions, these two
173
specific product ions contained a permanent intramolecular positive charge. They brought
174
enhanced sensitivity by increasing the ionization efficiency in the electrospray ionization. The
175
proposed collision-induced dissociation pathways for the precursor ion of CPR-UA were shown
176
in Figure 2B. In this study, m/z 398.8 was used for quantitative analysis, and m/z 443.2 was used 8
ACS Paragon Plus Environment
Page 8 of 28
Page 9 of 28
Journal of Agricultural and Food Chemistry
177
for confirmation analysis. The optimal FVs and CEs for two transitions of OA, UA and betulinic
178
acid (IS) were shown in Table 1.
179
Optimization of MAD-MDSPE. Optimization of the volume of EDC solution.
180
Using EDC as the condensing agent, the carboxylic acid group of OA and UA can be selectively
181
labeled. A great advantage of this derivatization reaction was that a small amount of water was
182
allowed to be present in the EDC condensation derivatization system. Therefore, CPR coupling
183
with EDC has the great advantage for the determination of triterpenic acids in aqueous sample
184
(such as microdialyaste) and thus was selected in this work. The volumes of CPR solution were
185
optimized in the range of 10-60 µL as shown in Figure 3A. The peak area increased with the
186
volumes of EDC from 10 to 30 µL and then decreased. Thus, 30 µL of EDC (0.1 mol/L) was
187
selected.
188
Optimization of the volume of CPR solution. To study the effect of CPR amount, the
189
volume of CPR solution was optimized in the range of 50-350 µL as shown in Figure 3B. No less
190
than 200 µL of CPR solution was significantly excess and insured the thorough derivatization of
191
analytes. The excess CPR was purified by the MDSPE procedure. 200 µL of CPR was chosen for
192
the MAD-MDSPE procedure.
193
Optimization of derivatization time and temperature. The derivatization time was
194
optimized from 10 to 40 min in the MAD-MDSPE procedure (microwave 450 W). As shown in
195
Figure 3C, the derivatization reaction was completed rapidly under the microwave assistance.
196
There were no remarkable increases of peak areas when the derivatization reaction time was more
197
than 25 min. Therefore, 25 min was used to perform the derivatization. The derivatization
198
temperature was optimized from 40 to 70 °C while other conditions were kept constant as shown
199
in Figure 3D. Optimum peak areas were obtained when OA and UA standards were derivatized at
200
60 °C.
201
Optimization of sorbent amount. To evaluate the effect of sorbent amount on 9
ACS Paragon Plus Environment
Journal of Agricultural and Food Chemistry
202
extraction efficiency and derivatization efficiency of CPR derivatives, the amount of Fe3O4/GO
203
was optimized in the range of 4-16 mg. The incremental amounts of sorbent up to 8 mg possibly
204
helped the derivatization reaction by providing a sufficient surface for derivatives adsorption, but
205
in higher amounts of it, lower extraction efficiency was obtained. Therefore, the MAD-MDSPE
206
procedure was carried out with 8 mg of Fe3O4/GO.
207
Optimization of desorption conditions. To achieve good desorption efficiency of
208
CPR derivatives, many desorption solutions including acetonitrile, acetone and methanol (each
209
containing 1.0 % formic acid) were estimated. The results indicated that methanol had the
210
strongest desorption power of CPR derivatives (Figure 4A). Methanol was a stronger polar
211
organic solvent than acetonitrile and acetone. Moreover, GO can be easily dispersed in polar
212
solvents because of its polar groups on the surface. Therefore, methanol was selected to ensure
213
sufficient desorption of the derivatives.
214
The effect of the volumes of desorption solution on the desorption efficiency was also
215
evaluated. When the methanol volume was increased to 300 µL, desorption efficiency was
216
increased because of the high rate of Fe3O4/GO dispersion in methanol. The results showed that
217
300 µL of methanol (1.0% formic acid) was enough for the efficient desorption of the derivatives
218
(Figure 4B).
219
Desorption time was optimized in the range of 10-100 s (ultrasound 120 W, 40 KHz).
220
Significantly increased peak areas of the CPR-derivatives were detected in 10-60 s and no
221
significant increase was obtained with the enhance of desorption time in 60-100 s. In the end, 1.0
222
min was employed as the optimal desorption time.
223
Method validation. To investigate the applicability of the developed MAD-MDSPE
224
coupled to UHPLC-MS/MS, linearity ranges, limits of detection (LODs), quantification (LOQs),
225
repeatability, recovery, precision, accuracy and matrix effect (ME) were determined. The linearity
226
of this method was established using internal standard spiked calibration solutions at 7 10
ACS Paragon Plus Environment
Page 10 of 28
Page 11 of 28
Journal of Agricultural and Food Chemistry
227
concentration levels. To evaluate the dynamic ranges of this method, six batches of calibration
228
microdialysate samples were prepared and determined. The peak-area ratio between the analyte
229
and IS of each spiked microdialysate sample was determined. The calibration curves were then
230
constructed by plotting the peak-area ratio with the spiked concentrations using linear regression
231
for each analyte, respectively. The regression equations were y=9.836x+0.082 (R=0.991) for OA
232
and y=9.923x−0.051 (R=0.995) for UA, where y was the peak-area ratio of the analyte and IS
233
and x was the concentration of analyte (ng/mL). The calibration curves covering the
234
concentration range of 0.050-100 ng/mL showed good linearity with correlation coefficient R >
235
0.99. The LODs were 0.025 and 0.020 ng/mL for OA and UA (S/N > 3). The LOQs for OA and
236
UA in microdialysates were 0.090 and 0.080 ng/mL (S/N > 10).
237
As shown in Table 2, the precision was in the range of 2.20-6.01 %, and the accuracy was in
238
the range of 85.1-112.3% from the actual QCs. The intra- and inter-day accuracy and precision
239
were all within 15% by FDA. The matrix effect of the analytes ranged from 92.6-111.3% at 3
240
concentration levels. The repeatability of the method was in the range of 2.34-7.35%. The
241
recoveries of the OA and UA were in the range of 97.6-109.8% at 3 concentration levels. These
242
results indicated that the developed method could be well used for the sensitive, specific and
243
accurate determination of OA and UA in rat microdialysate samples.
244
Pharmacokinetics of OA and UA from in vivo rat blood microdialysates.
245
The developed MAD-MDSPE coupled to UHPLC-MS/MS method has been used to analyzing
246
microdialysate samples from rat blood after oral administration of Arctiumlappa L. root extract.
247
Typical MRM chromatograms of internal standard (IS) betulinic acid, OA and UA derivatives in
248
a rat blood microdialysate sample were shown in Figure 5. The mean plasma drug
249
concentration-time profiles were shown in Figure 6. Table 3 presented the pharmacokinetic
250
parameters including the maximum plasma concentration (Cmax), the time for reaching the
251
maximum concentration (Tmax), terminal half-life (t1/2), the area under the concentration-time 11
ACS Paragon Plus Environment
Journal of Agricultural and Food Chemistry
252
curve (AUC), mean residence time (MRT), the apparent volume of distribution (Vz/F) and
253
time-averaged total body clearance (CL/F). After oral administration, both OA and UA could be
254
absorbed into the blood. However, their systemic exposures were quite different. OA appeared to
255
be absorbed slightly quickly into the plasma with a Tmax of 45 min, while UA relatively absorbed
256
slowly with a Tmax of 50 min. However, the absolute bioavailability of UA was obviously better
257
than OA with Cmax of 40 vs 10 ng/mL. This finding is consistent with previous study,5 which will
258
be helpful for future pharmacology, pharmacodynamics and drug development.
259
In conclusion, we developed a rapid, selective and sensitive strategy based on MAD-MDSPE
260
coupled to UHPLC-MS/MS (MRM) for the simultaneous determination of OA and UA in the rat
261
blood microdialysates. The derivatization, extraction and purification of OA and UA occurred on
262
the surface of Fe3O4/GO and were integrated into one step. Furthermore, the validated method
263
was successfully used to the pharmacokinetics study.
264
Supporting Information
265
The Supporting Information is available free of charge on the ACS Publications website.
266
Preparation of Fe3O4/GO.
267
Corresponding Authors
268
*E-mail:
[email protected] (Zhao XE), Tel: +86-537-4456301, Fax: +86-537-4456305;
269
*E-mail:
[email protected] (Zhu SY);
270
**E-mail:
[email protected] (Qiao XG).
271
ORCID
272
Xian-En Zhao: 0000-0003-3500-9518;
273
Shuyun Zhu: 0000-0002-9632-8187;
274
Funding
275
This work was supported by the National Natural Science Foundation of China (Nos. 21775088,
276
21405094, and 81303179), the Special Fund for Agro-scientific Research in the Public Interest 12
ACS Paragon Plus Environment
Page 12 of 28
Page 13 of 28
Journal of Agricultural and Food Chemistry
277
(Grant No. 201503142), the Innovation Platform for the Development and Construction of
278
Special Project of Key Laboratory of Tibetan Medicine Research of Qinghai Province (No.
279
2017-ZJ-Y11), and the Open Projects Program of the Key Laboratory of Tibetan Medicine
280
Research of Chinese Academy of Sciences.
281
Notes
282
The authors declare that they have no conflict of interest.
283
REFERENCES
284
1. Gautam, R.; Jachak, S. M. Recent developments in anti-inflammatory natural products. Med.
285
Res. Rev. 2009, 29, 767-820.
286
2. Yin, M. C.; Lin, M. C.; Mong, M. C.; Lin, C. Y. Bioavailability, distribution, and antioxidative
287
effects of selected triterpenes in mice. J. Agric. Food Chem. 2012, 60, 7697-7701.
288
3. Wu, H. L.; Li, G. L.; Liu, S. C.; Liu, D.; Chen, G.; Hu, N.; Suo, Y. R. Simultaneous
289
determination of six triterpenic acids in some Chinese medicinal herbs using ultrasound-assisted
290
dispersive liquid-liquid microextraction and high-performance liquid chromatography with
291
fluorescence detection. J. Pharmaceut. Biomed. 2015, 107, 98-107.
292
4. Sanchez-Avila, N.; Priego-Capote, F.; Ruiz-Jimenez, J.; de Castro, M. D. L. Fast and selective
293
determination of triterpenic compounds in olive leaves by liquid chromatography-tandem mass
294
spectrometry with multiple reaction monitoring after microwave-assisted extraction. Talanta
295
2009, 78, 40-48.
296
5. Zhao, L.; Li, W. H.; Li, Y. Y.; Xu, X.; Lv, L.; Wang, X. X.; Chai, Y. F.; Zhang, G. Q.
297
Simultaneous Determination of oleanolic and ursolic acids in rat plasma by HPLC–MS:
298
application to a pharmacokinetic study after oral administration of different combinations of
299
qinggansanjie decoction extracts. J. Chromatogr. Sci. 2015, 53, 1185-1192.
300
6. Avila, N. S.; Capote, F. P.; de Castro, M. D. L. Ultrasound-assisted extraction and silylation
301
prior to gas chromatography–mass spectrometry for the characterization of the triterpenic fraction 13
ACS Paragon Plus Environment
Journal of Agricultural and Food Chemistry
302
in olive leaves. J. Chromatogr. A 2007, 1165, 158-165.
303
7. Banerjee, A.; T Sane, R.; Mangaonkar, K.; Shailajan, S.; Deshpande, A.; Gundi, G.
304
Quantitation of oleanolic acid in Oldenlandia corymbosa L. whole-plant powder by
305
high-performance thin-layer chromatography. JPC-J. Planar Chromat. 2006, 19, 68-72.
306
8. Gao, R. B. ; Wang, L. T.; Yang, Y.; Ni, J. M.; Zhao, L.; Dong, S. Q.; Guo, M. Simultaneous
307
determination of oleanolic acid, ursolic acid, quercetin and apigenin in Swertia mussotii Franch
308
by capillary zone electrophoresis with running buffer modifier. Biomed. Chromatogr. 2015, 29,
309
402-409.
310
9. Sowa, I.; Wojciak-Kosior, M.; Rokicka, K.; Kocjan, R.; Szymczak, G. Application of solid
311
phase extraction with the use of silica modified with polyaniline film for pretreatment of samples
312
from plant material before HPLC determination of triterpenicacids. Talanta 2014, 122, 51-57.
313
10. Dais, P.; Plessel, R.; Williamson, K.; Hatzakis, E. Complete H-1 and C-13 NMR assignment
314
and P-31 NMR determination of pentacyclic triterpenic acids. Anal. Methods 2017, 9, 949-957.
315
11. He, Y. R.; Zhao, X. E.; Wang, R. J.; Wei, N.; Sun, J.; Dang, J.; Chen, G.; Liu, Z. Q.; Zhu, S. Y.;
316
You, J. M. Simultaneous Determination of food-related biogenic amines and precursor amino
317
acids using in situ derivatization ultrasound-assisted dispersive liquid-liquid microextraction by
318
ultra-high-performance liquid chromatography tandem mass spectrometry. J. Agric. Food Chem.
319
2016, 64, 8225-8234.
320
12. Zhao, X.-E.; Lv, T.; Zhu, S. Y.; Qu, F.; Chen, G.; He, Y. R.; Wei, N.; Li, G. L.; Xia, L. ; Sun, Z.
321
W.; Zhang, S. J.; You, J. M.; Liu, S.; Liu, Z. G. ; Sun, J.; Liu, S. Y. Dual ultrasonic-assisted
322
dispersive liquid-liquid microextraction coupled with microwave-assisted derivatization for
323
simultaneous determination of 20(S)-protopanaxadiol and 20(S)-protopanaxatriol by ultra high
324
performance liquid chromatography-tandem mass spectrometry. J. Chromatogr. A 2016, 1437,
325
49-57.
326
13. Mung, D.; Li, L. Development of chemical isotope labeling LC-MS for milk metabolomics:
327
comprehensive and quantitative profiling of the amine/phenol submetabolome. Anal. Chem. 2017, 14
ACS Paragon Plus Environment
Page 14 of 28
Page 15 of 28
Journal of Agricultural and Food Chemistry
328
89, 4435-4443.
329
14. Li, G. L.; You, J. M.; Song, C. H.; Xia, L. A.; Zheng, J.; Suo, Y. R. Development of a new
330
HPLC method with precolumn fluorescent derivatization for rapid, selective and sensitive
331
detection of triterpenic acids in fruits. J. Agric. Food Chem. 2011, 59, 2972-2979.
332
15. Chen, G.; Li, J.; Song, C. H.; Suo, Y. R.; You, J. M. A sensitive and efficient method for
333
simultaneous trace detection and identification of triterpene acids and its application to
334
pharmacokinetic study. Talanta 2012, 98, 101-111.
335
16. Wang, Y. W.; Suo, Y. R.; Sun, Y. N.; You, J. M. Determination of triterpene acids from 37
336
different varieties of raspberry using pre-column derivatization and HPLC fluorescence detection.
337
Chromatographia 2016, 79, 1515-1525.
338
17. Wozniak, L.; Marszalek, K.; Skapska, S.; Jedrzejczak, R. Novel method for hplc analysis of
339
triterpenic acids using 9-anthryldiazomethane derivatization and fluorescence detection.
340
Chromatographia 2017, 80, 1527-1533.
341
18. You, J. M.; Wu, D.; Zhao, M.; Li, G. L.; Gong, P. W.; Wu, Y. Y.; Guo, Y.; Chen, G.; Zhao, X.
342
E.; Sun, Z. W.; Xia, L.; Wu, Y. N. Development of a facile and sensitive HPLC-FLD method via
343
fluorescence labeling for triterpenic acid bioavailability investigation. Biomed. Chromatogr. 2017,
344
31, UNSP e3894.
345
19. Jemmali, Z.; Chartier, A.; Dufresne, C.; Elfakir, C. Optimization of the derivatization protocol
346
of pentacyclic triterpenes prior to their gas chromatography-mass spectrometry analysis in plant
347
extracts. Talanta 2016, 147, 35-43.
348
20. Tang, S.; Zhang, H.; Lee, H. K. Advances in sample extraction. Anal. Chem. 2016, 88,
349
228-249.
350
21. Xu, L. N.; Qi, X. Y.; Li, X. J.; Bai, Y.; Liu, H. W. Recent advances in applications of
351
nanomaterials for sample preparation. Talanta 2016, 146, 714-726.
352
22. Zhao, X. E.; He, Y. R.; Li, M.; Chen, G.; Wei, N.; Wang, X.; Sun, J.; Zhu, S. Y.; You, J. M.
353
Analysis of amino acid and monoamine neurotransmitters and their metabolites in rat urine of 15
ACS Paragon Plus Environment
Journal of Agricultural and Food Chemistry
354
Alzheimer’s disease using in situ ultrasound-assisted derivatization dispersive liquid-liquid
355
microextraction with UHPLC–MS/MS. J. Pharmaceut. Biomed. 2017, 135, 186-198.
356
23. Cao, Z. Y.; Ma, Y. N.; Sun, L. H.; Mou, R. X.; Zhu, Z. W.; Chen, M. X. Direct determination
357
of six cytokinin nucleotide monophosphates in coconut flesh by reversed-phase liquid
358
chromatography-tandem mass spectrometry. J. Agric. Food Chem. 2017, 65, 9909–9915
359
24. Xiang, L.; Chen, L.; Xiao, T.; Mo, C. H.; Li, Y. W.; Cai, Q. Y.; Li, H.; Zhou, D. M.; Wong, M.
360
H. Determination of trace perfluoroalkyl carboxylic acids in edible crop matrices: matrix effect
361
and method development. J. Agric. Food Chem. 2017, 65, 8763-8772
362
25 Xu, J.; Wu, P.; Ye, E. C.; Yuan, B. F.; Feng, Y. Q. Metal oxides in sample pretreatment. Trends
363
Anal. Chem. 2016, 80, 41-56.
364
26. Wang, A. X.; Lu, H. Z.; Xu, S. F. Preparation of magnetic hollow molecularly imprinted
365
polymers for detection of triazines in food samples. J. Agric. Food Chem. 2016, 64, 5110-5116.
366
27 Wen, Y. Y.; Chen, L.; Li, J. H.; Liu, D. Y.; Chen, L. X. Recent advances in solid-phase sorbents
367
for sample preparation prior to chromatographic analysis. Trends Anal. Chem. 2014, 59, 26-41.
368
28 Pei, H. M.; Zhu, S. Y.; Yang, M. H.; Kong, R. M.; Zheng, Y. Q.; Qu, F. L. Graphene oxide
369
quantum dots@silver core-shell nanocrystals as turn-on fluorescent nanoprobe for ultrasensitive
370
detection of prostate specific antigen. Biosens. Bioelectronics 2015, 74, 909-914.
371
29 Ren, H.; Kukarni, D. D.; Kodiyath, R.; Xu, W. N.; Choi, I.; Tsukruk, V. V. Competitive
372
adsorption of dopamine and rhodamine 6g on the surface of graphene oxide. Appl. Mater.
373
Interfaces 2014, 6, 2459-2470.
374
30. Shannon, R. J.; Carpenter, K. L. H.; Guilfoyle, M. R.; Helmy, A.; Hutchinson, P. J. Cerebral
375
microdialysis in clinical studies of drugs: pharmacokinetic applications. J. Pharmacokinet. Phar.
376
2013, 40, 343-358.
377
31. Zhao, X. E.; He, Y. R.; Yan, P.; Wei, N.; Wang, R. J.; Sun, J.; Zheng, L. F.; Zhu, S. Y.; You, J.
378
M. Sensitive and accurate determination of neurotransmitters from in vivo rat brain
379
microdialysate of Parkinson's disease using in situ ultrasound-assisted derivatization dispersive 16
ACS Paragon Plus Environment
Page 16 of 28
Page 17 of 28
Journal of Agricultural and Food Chemistry
380
liquid–liquid microextraction by UHPLC-MS/MS. RSC Adv. 2016, 6, 108635-108644.
381
32 Kong, W. M.; Mohamed, Z.; Alshawsh, M. A.; Chik, Z. Evaluation of pharmacokinetics and
382
blood-brain barrier permeabilityof mitragynine using in vivo microdialysis technique. J.
383
Pharmaceut. Biomed. 2017, 143, 43-47
384
33. Zheng, L. F.; Zhao, X. E.; Ji, W. H.; Wang, X.; Tao, Y. D.; Sun, J.; Xu, Y. Q.; Wang, X.; Zhu,
385
S. Y.; You, J. M. Core-shell magnetic molecularly imprinted polymers used rhodamine B
386
hydroxyproline derivate as template combined with in situ derivatization for the specific
387
measurement of L-hydroxyproline. J. Chromatogr. A 2018, 1532, 30-39.
388
34. Anastassiades, M.; Lehotay, S. J.; Štajnbaher, D.; Schenck, F. J. Fast and easy multiresidue
389
method employing acetonitrile extraction/partitioning and dispersive solid-phase extraction for
390
the determination of pesticide residues in produce. J. AOAC Int. 2003, 86, 412-431.
391
35. Sun, J. P., Liang, Q. L., Han, Q., Zhang, X. Q., Ding, M. Y. One-step synthesis of magnetic
392
graphene oxide nanocomposite and its application in magnetic solid phase extraction of heavy
393
metal ions from biological samples. Talanta 2015, 132, 557-563.
394 395 396 397 398 399 400 401 402 403 404 405 17
ACS Paragon Plus Environment
Journal of Agricultural and Food Chemistry
406
FIGURE CAPTIONS
407
Figure 1. The synthesized of CPR and the derivatization reaction scheme of CPR with OA and
408
UA.
409
Figure 2. (A) The representative MRM chromatogram of CPR derivatives of internal standard
410
(IS) betulinic acid, OA and UA standards, (B) product ion spectrum and the proposed
411
fragmentation schematics of CPR-UA derivative.
412
Figure 3. Optimization of MAD conditions (n = 5), (A) volumes of EDC, (B) volumes of CPR
413
solution, (C) time (min), and (D) temperature (°C).
414
Figure 4. Optimization of desorption conditions (n = 5), (A) types of desorption solution, (B)
415
volumes of desorption solution.
416
Figure 5. Typical MRM chromatograms of internal standard (IS) betulinic acid, OA and UA
417
derivatives in a rat blood microdialysate sample.
418
Figure 6. Mean concentration-time curves of OA and UA in rat plasma microdialysates after oral
419
administration of Arctiumlappa L. root extract. Each point represents the mean ± standard error
420
(n=6).
421
18
ACS Paragon Plus Environment
Page 18 of 28
Page 19 of 28
Journal of Agricultural and Food Chemistry
Tables Table 1. MRM Parameters of OA, UA and Betulinic Acid (IS) Analytes
Fragmentor (V)
Quantitation Transition (m/z)
Collision Energy (eV)
Confirmation Transition (m/z)
Collision Energy (eV)
OA
240
949.6 > 398.8
82
949.6 > 443.2
78
UA
240
949.6 > 398.8
81
949.6 > 443.2
78
Betulinic Acid (IS)
250
949.6 > 398.8
79
949.6 > 443.2
76
19
ACS Paragon Plus Environment
Journal of Agricultural and Food Chemistry
Page 20 of 28
Table 2. Results of Recovery, Matrix Effect, Repeatability, Precision, and Accuracy for OA and UA (n = 6)
Analytes
OA
UA
Spiked Levels (ng/mL)
Recovery (%)
Matrix Effect (%)
Repeatability (RSD, %)
Intra-day Precision
Inter-day Precision
(RSD, %)
(RSD, %)
Accuracy (%)
Peak Area
Retention Time
Peak Area
Retention Time
Peak Area
Retention Time
Intra-day
Inter-day
0.50
98.4±4.2
98.6±6.5
7.35
3.14
3.59
3.35
4.80
3.50
93.0
92.8
5.0
97.6±3.1
94.6±6.1
6.62
2.34
5.81
2.57
3.45
3.21
99.1
112.3
50.0
104.1±5.1
103.3±7.0
4.23
4.50
3.87
2.90
2.33
3.89
101.8
87.2
0.50
98.7±5.1
111.3±6.5
4.75
3.70
4.41
4.20
5.58
2.20
101.4
85.1
5.0
109.8±4.3
103.7±6.9
5.52
2.88
5.27
3.77
6.01
5.74
98.0
103.4
50.0
103.5±3.6
92.6±4.5
6.26
2.70
5.70
3.29
5.12
3.10
96.4
110.5
20
ACS Paragon Plus Environment
Page 21 of 28
Journal of Agricultural and Food Chemistry
Table 3. The Plasma Pharmacokinetic Parameters after Oral Administration of Arctiumlappa L. Root Extract to Rats. Parameter
Unit
UA
OA
t1/2
min
294.67±83.66
333.89±107.88
Tmax
min
50±7.75
45±9.49
Cmax
ng/mL
40.55±4.11
10.51±3.26
AUC0−t
ng/mL*min
6897.44±428.33
1249.10±245.91
AUC0−∞
ng/mL*min
9491.76±953.31
1749.03±470.50
MRT
min
457.42±87.94
473.65±110.73
Vz/F
(ng)/(ng/mL)
36.60±7.35
228.31±40.28
CL/F
(ng)/(ng/mL)/min
0.09±0.009
0.50±0.12
21
ACS Paragon Plus Environment
Journal of Agricultural and Food Chemistry
Figure 1
22
ACS Paragon Plus Environment
Page 22 of 28
Page 23 of 28
Journal of Agricultural and Food Chemistry
Figure 2 (A, B)
23
ACS Paragon Plus Environment
Journal of Agricultural and Food Chemistry
Figure 3 (A, B, C, D)
24
ACS Paragon Plus Environment
Page 24 of 28
Page 25 of 28
Journal of Agricultural and Food Chemistry
Figure 4 (A, B)
25
ACS Paragon Plus Environment
Journal of Agricultural and Food Chemistry
Figure 5
26
ACS Paragon Plus Environment
Page 26 of 28
Page 27 of 28
Journal of Agricultural and Food Chemistry
Figure 6
27
ACS Paragon Plus Environment
Journal of Agricultural and Food Chemistry
Graphic for Table of Contents
28
ACS Paragon Plus Environment
Page 28 of 28