You are on page 1of 7

Chinese

Journal of
Natural
Chinese Journal of Natural Medicines 2015, 13(3): 0208−0214 Medicines

Determination of α-glucosidase inhibitors from Scutellaria baicalensis


using liquid chromatography with quadrupole time of flight tandem
mass spectrometry coupled with centrifugal ultrafiltration
YANG Jun-Ran, LUO Jian-Guang, KONG Ling-Yi*

State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nan-
jing 210009, China
Available online 20 Mar. 2015

[ABSTRACT] The present study aimed at identifying potential lead compounds for diabetes mellitus drug discovery. We developed a
novel method involving centrifugal ultrafiltration separation subsequent liquid chromatography with quadrupole time of flight tandem
mass spectrometry (LC-Q/TOF-MS/MS) determination to screen α-glucosidase inhibitors in complex Scutellaria baicalensis Georgi
(SBG) extract. By adding a second filter to the screening process, the level of non-specific binding of Compounds 1, 3, 10 and 11 was
significantly decreased, and the level of non-specific binding of Compounds 5 and 15 also was reduced. As a result, five flavonoids
identified as baicalein, baicalein, wogonin, chrysin, and oroxylin A, were rapidly found to interact with α-glucosidase and possess
potent anti-α-glucosidase activity in vitro. Specific binding of ligands to α-glucosidase was demonstrated though the proposed method
and the ligands could be ranked in order of affinity for α-glucosidase, which were corresponded to the order of inhibitory activity in
vitro. In conclusion, our results indicated that the developed method is a rapid and effective screening method for rat intestinal
α-glucosidase inhibitors from complex herbal medicines such as SBG.
[KEY WORDS] LC-Q/TOF-MS/MS; Centrifugal ultrafiltration; Flavonoids; Scutellaria baicalensis; α-Glucosidase inhibitors
[CLC Number] R917 [Document code] A [Article ID] 2095-6975(2015)03-0208-07

agents based on the interaction of the compounds with intes-


Introduction tinal α-glucosidase.
The radix of Scutellaria baicalensis Georgi (SBG), a α-Glucosidase (AGH, EC3.2.1.20), located in the brush-
perennial herb of the Labiatae family, is a well-known, widely border surface membrane of the small intestinal cells, is an
used medicinal plant in China. Its main chemical constituents exo-type carbohydrate enzyme that catalyzes the liberation of
are flavonoids which have been demonstrated to have anti- α-glucose from the non-reducing end of the carbohydrates. The
bacterial [1], anti-inflammatory [2], and radical scavenging inhibition of intestinal α-glucosidase can delay the digestion
activities [3]. Although anti-type 2 diabetes drugs can cure the and absorption of carbohydrates and, consequently, suppress
hyperglycemia-induced mitochondrial damage in streptozoto- postprandial hyperglycemia. Many studies have used a broad
cin-induced diabetes Wistar rats [4], no studies have been car- range of natural [5] or synthetic compounds [6] with in vitro
ried out to investigate the potentials of anti-α-glucosidase AGH inhibitory activity. However, most of these studies were
done by using microbial AGH, not mammalian AGH. Com-
[Received on] 30-Mar.-2014 mercially available AGHs may be divided into two families,
[Research funding] This work was supported by the National Key types I (microbial) and types II (mammals), on the basis of the
Scientific and Technological Special Projects (2012ZX09103- differences in primary structure [7]. Since microbial
101-007), the Program for Changjiang Scholars and Innovative Re- α-glucosidase inhibitors (AGIs) may not necessarily inhibit
search Team in University (PCSIRT-IRT1193), and the Project
mammalian AGH and vice versa [8], the study of AGIs from
Funded by the Priority Academic Program Development of Jiangsu
Higher Education Institutions (PAPD).
mammalian sources may be a better model to design and de-
velop anti-AGH agents.
[*Corresponding author] Tel/Fax: 86-25-83271405, E-mail: cpu_
The conventional methods for screening bioactive
lykong@126.com
compounds from Traditional Chinese Medicines (TCMs)
These authors have no conflict of interest to declare.
Published by Elsevier B.V. All rights reserved are both time-consuming and inefficient, requiring isola-

– 208 –
YANG Jun-Ran, et al. / Chin J Nat Med, 2015, 13(3): 208−214

tion and purification of the chemical constituents, fol- Acetonitrile was of HPLC grade and purchased from
lowed by evaluation of their bioactivity in vivo or in vitro Merck (Darmstadt, Germany) and ultrapure water was pre-
[9]
. Moreover, most of the screening techniques in vitro are pared by Thermo Scientific Barnstead D8611 Easypure II
based on optical or radioactive detection, which may be (Thermo, Waltham, USA). Rat intestine acetone powder was
affected by matrix interference, especially when complex purchased from Sigma Chemical Co. Ltd. (St. Louis, MO,
samples are analyzed. Therefore, the methods using spec- USA). Acarbose was obtained from the National Institute
trometry coupled with bioactivity screening have emerged for the Control of Pharmaceutical and Biological Products
as an important analytical tool in the identification and (Beijing, China). Glucose assay kit was obtained from Nan-
characterization of biologically active compounds for pro- jing Jiancheng Bioengineering Institute (Nanjing, China).
tein targets of interest [10-13]. Centrifugal ultrafiltration Maltose, KH2PO4, and Na2HPO4 used for α-glucosidase
hyphenated techniques (LC-MS or GC-MS) have attracted inhibitory activity assaywere all analytical grade reagents
much attention worldwide [14]. The centrifugal ultrafiltra- purchased from Nanjing Chemical Reagent Corporation
tion technique allows the removal of solutes of low mo- (Nanjing, China).
lecular weight from the chamber, retaining solutes of high LC-Q/TOF-MS/MS analysis
molecular weight (i.e., enzyme and enzyme-ligand), in- The LC analysis was performed on an Agilent 1200
cluding the analytes, after centrifugation through a suitable HPLC system (Agilent Technologies, Santa Clara, CA, USA),
molecular weight cutoff [15]. This procedure permits a equipped with a dual pump, a degasser, a column oven, an
minimal consumption of sample, and is a simple, rapid and autosampler and a diode array detector. The separations were
easily applicable technique in a routine analytical labora- carried out using a Zorbax SB C18 column (250 mm × 4.6 mm
tory, providing a satisfactory separation. However, one-filter i.d., 5 μm) and an Alltima C18 guard column (4.6 mm × 7.5
centrifugal ultrafiltration can result in high non-specific mm i.d., 5μm). The column temperature was kept at 25 °C.
binding that might interfere with the detection of specifi- The mobile phase was consisted of 0.2% aqueous acetic acid
cally bound ligands [16-17]. Consequently, there is still room (A) and acetonitrile (B); the eluting gradient was as follows:
for improvement for this technique. 0−5 min, 15% B; 5−15 min, 20% B; 15−20 min, 20%−25% B;
In the present study, a method for the analysis of AGIs in S. 20−35 min, 25% B; 35−50 min, 25%−33% B; 50−60 min,
baicalensis was developed using LC-Q/TOF-MS/MS coupled 37% B; 60−70 min, 33%−53% B; 70−80 min, 53% B. The
with two-filter centrifugal ultrafiltration. As a result, five poten- flow rate was set at 0.8 mL·min−1 and the injection volume
tial active candidate compounds, which could interact with rat was 15 μL. The chromatogram was acquired at 280 nm.
intestinal α-glucosidase, were detected and identified. Their The LC analysis of rat intestinal α-glucosidase ligands
inhibitory activity assays were subsequently confirmed in vitro. (baicalin, baicalein, wogonin, chrysin and oroxylin A) in
spiked samples was performed as described above, except for
Experimental the mobile phase conditions. The LC was carried out with an
isocratic solvent system consisting of 0.2% aqueous acetic
Chemicals and reagents
acid (A) and methanol (B) at a flow rate of 1 mL·min−1. Other
S. baicalensis roots were obtained from Xiansheng
parameters were adopted as follows: injection volume, 15 μL;
pharmacy store in Nanjing, Jiangsu Province, China, in July,
Column temperature, 30 °C; and monitored wavelength, 280
2011. The reference compounds baicalein, wogonin, chrysin,
nm. The isocratic solvents of 45% methanol were used for
oroxylin A, and baicalin were isolated by silica gel column
baicalin, and 65% methanol for baicalein, wogonin, chrysin,
chromatography and then by preparative RP-HPLC from S.
and oroxylin A.
baicalensis in our laboratory. The purity of these com-
The LC-Q/TOF-MS/MS analysis was performed on an
pounds was over 98%, and they were identified by spectral
Agilent 1200 Series LC system (Agilent Technologies)
data (UV, MS, NMR) compared those reported in litera-
equipped with a binary pump, an auto plate-sampler, a ther-
ture [18-19]. The chemical structures of the five compounds
mostatically controlled column compartment, a diode array
are shown in Fig. 1.
detector, and an Agilent G6520 quadrupole time-of-flight
tandem mass spectrometer. The negative ion ESI-MS and
ESI-MS/MS experiments were conducted using conditions as
follows: skimmer, 65 V; drying gas (N2) flow, 8 L·min−1; dry
temperature, 350 °C; and nebulizer (N2) pressure, 45 psi. Full
scan data acquisition was performed for m/z of 100−1 200 in
the negative ion mode. All of the operations, acquisition, and
analysis of data were monitored by Agilent LC-Q/TOF-MS
Mass Hunter Acquisition Software Version B.04.00 and op-
Fig. 1 Chemical structures of the five flavonoids identified erated under Mass Hunter Qualitative Analysis Software Ver-
from enzyme-ligands mixture sion B.04.00 (Agilent Technologies).

– 209 –
YANG Jun-Ran, et al. / Chin J Nat Med, 2015, 13(3): 208−214

Sample preparation − (ODsample − ODsample blank)] /(ODcontrol − ODcontrol blank)


The roots of S. baicalensis (100 g) were extracted with The IC50 value was defined as the concentration of
95% ethanol (500 mL) for 3 h at 85 °C and then filtered. The α-glucosidase inhibitor that inhibited 50% of α-glucosidase
filtrate was evaporated to afford a residue (5.3 g). The crude activity.
extract (20.0 mg, drying weight) was dissolved in methanol (1
mL), and filtered through a 0.45-µm microporous membrane.
Results and Discussion
The filtrate was then used for centrifugal ultrafiltration analy- Bio-affinity screening of ligands to a target enzyme is
sis. Five purified flavonoids (50 μmol·L−1, 100 μmol·L−1) mainly based on the interaction between the ligands and the
were dissolved in methanol, and filtered through a 0.45-µm active site of the enzyme. The ultrafiltration membrane retains
microporous membrane, respectively. The solutions were the α-glucosidase and the enzyme-ligands complexes, but
used for the relative ranking of binding affinity analysis by allows unbound, low molecular weight compounds to wash
LC-Q/TOF-MS/MS coupled with a centrifugal ultrafiltration away from the chamber. When specific binding to rat intesti-
method. nal α-glucosidase occurred, the peak area of the ligand was
Centrifugal ultrafiltration procedure greater in the chromatogram corresponding to incubation with
For the centrifugal ultrafiltration experiment, 2 μL of active rat intestinal α-glucosidase than that of sample incu-
20.0 mg·mL−1 SBG sample solution or 3.75 mmol·L−1 of a bated with inactive rat intestinal α-glucosidase. Background
purified compound was incubated for 2 h at 37 °C with 60 noise due to non-specific binding of compounds to the ul-
μL of α-glucosidase (1.07 mg·mL−1) in 10 mmol·L−1 am- trafiltration membrane was minimized by introducing a sec-
monium acetate buffer (pH 6.8), in a total volume of 150 ond filter before the ligand-enzyme dissociation step; the
μL. After incubation, each mixture was filtered through a specific binding ligands were then identified by LC-Q/TOF-
Microcon (Millipore, Bedford, MA, USA) YM-10 cen- MS/MS.
trifugal filter containing a regenerated cellulose ultrafiltra- Centrifugal ultrafiltration
tion membrane with 10 000 MW cutoff by centrifugation The use of the ultrafiltration technique for screening can
at 10 000 × g for 15 min at 4 °C. Using the modified result in high non-specific binding that might interfere with
method of Mulabagal et al [20], the ultrafiltration filter was the detection of specifically bound ligands. By replacing the
first washed three times by centrifugation with 200 μL of ultrafiltration filter before the ligand dissociation and elution
ammonium acetate buffer (pH 6.8) at 4 °C to remove the step with a second filter, the level of non-specific binding was
unbound compounds. Subsequently, the washed en- decreased. Fig. 2 shows the ultrafiltration a-glucosidase
zyme-ligand solution was transferred into a new filter, screening results for the extract of SBG obtained using
proceeding the same operation. Finally, the ultrafiltrate one-filter centrifugal ultrafiltration. As shown in Fig. 2, peaks
was dried under vacuum, and then reconstituted in metha- 1, 3, 10, and 11 were barely detected with centrifugal ultrafil-
nol (100 μL) for analysis. For comparison, the control tration in the chromatogram (solid line), and Compounds 5
experiments with denatured enzyme were carried out be- and 15 were also decreased. Therefore, the two-filter method
fore each screening experiment. was used as a tool to screen the ligands of a-glucosidase from
Rat intestinal α-glucosidase inhibitory activity SBG.
Rat intestinal α-glucosidase inhibitory activity was
measured as described previously with a slight modification
[21]
. In brief, 10 μL of the test sample in DMSO solution was
reconstituted in 100 μL of 0.1 mol·L−1 phosphate buffer (pH
6.8) in a 96-well microplate and 20 μL of the enzyme solution
added, and the mixture was incubated at 37 °C for 10 min.
The buffer and substrate (5 mmol·L−1 maltose in 0.1 mol·L−1
phosphate buffer) were added to the mixture and incubated at
37 °C for 20 min. The reaction mixtures heated for 3 min in a
boiling water bath to stop the reaction. The concentration of
glucose released from the reaction mixtures was determined
using a glucose assay kit based on the glucose oxidase me-
thod. Individual blanks for test samples were prepared to
correct for background absorbance where the substrate was
replaced with buffer. The control blank contained DMSO (10
Fig. 2 Comparison of specific and non-specific binding of
μL) in place of the test samples. Acarbose was used as a posi-
SBG constituents after incubation with active rat intestinal
tive control. Inhibitory activity was calculated by the follow- α-glucosidase using one-filter and two-filter ultrafiltration
ing equation: (solid line: two-filter ultrafiltration; dashed line: one-filter
Inhibitory activity (%) = 100 × [(ODcontrol − ODcontrol blank) ultrafiltration)

– 210 –
YANG Jun-Ran, et al. / Chin J Nat Med, 2015, 13(3): 208−214

Screening bioactive components from SBG


The chromatographic conditions were optimized to
achieve a complete separation of the nineteen SBG constitu-
ents in the extract; the baseline separations are shown in Fig.
3a. After the incubation with rat intestinal α-glucosidase and
two-filter affinity purification, the trapped ligands in SGB
were analyzed by LC-Q/TOF-MS/MS. The LC-chroma-
togram for the experiment is shown in Fig. 3b. Enhancement
of the HPLC peak areas in the experimental incubations (dash
lines) indicated the specific binding of ligands to rat intestinal
α-glucosidase. As shown in Fig. 3b, five trapped ligands
(Compounds 5, 13, 15, 16, and 18) from SBG showed spe-
cific binding to rat intestinal α-glucosidase. Compounds 1, 2,
3, 4, 6, 7, 8, 9, 10, 11, 14, 17, and 19 were not regarded as rat
intestinal α-glucosidase ligands because they could not be
distinguished from the control sample in two-filter centrifugal
ultrafiltration screening assay.
Identification of inhibitors in the S. baicalensis extract
The LC-Q/TOF-MS/MS analysis was carried out to
identify compounds that were bound in the rat intestinal
α-glucosidase assays. The mass spectral data of the SBG
extract and the ultrafiltrate were obtained in the negative
ion mode. Detailed fragmentation ions and their rela-
tionships with the molecular structure of each compound
were analyzed by high-resolution Q/TOF-MS/MS, and
the results are listed in Table 1 and Fig. 4. By comparing
the proposed product ions, UV spectra data, measured Fig. 3 LC chromatogram of the extract of S. baicalensis
monitored at 280 nm: (a) extract of S. baicalensis, (b) extract
and theoretical masses, measurement error and proposed
of S. baicalensis incubated with rat intestinal α-glucosidase
natural losses with those of the corresponding reference after performing ultrafiltration (solid lines: control experi-
compounds and the literature [18, 19] , the five ligands were ments incubated with denatured rat intestinal α-glucosidase;
identified as the flavonoids baicalin, baicalein, wogonin, dashed lines: experiments incubated with active rat intestinal
chrysin, and oroxylin A. α-glucosidase), (c) the amplification of Fig. 3b

Table 1 LC-Q/TOF-MS/MS accurate product ion data for the identification of five rat intestinal α-glucosidase ligands from SBG
Peak λmax (nm) Measured m/z Fragment ions Theory m/z Formula [M – H]– Error (ppm) Identificationa
445.077 7 [M – H]– 445.077 6 C21H17O11 –0.25
269.045 3 [M – H – Glc acid]– 269.045 5 C15H9O5 1.06 Baicalein-7-O-Glu acidb
5 222, 276, 316
241.051 0 [M – H – Glc acid – CO]– 241.050 6 C14H9O4 –1.39 (Baicalin)
223.040 6 [M – H – Glc acid – CO – H2O]– 223.040 1 C14H7O3 –2.38
269.045 7 [M – H]– 269.045 5 C15H9 O5 –0.68
251.034 3 [M – H – H2O]– 251.035 0 C15H7O4 2.54
5,6,7-Trihydroxy flavone
13 276, 323 241.050 1 [M – H – CO]– 241.050 6 C14H9O4 2.40
(Baicalein)
223.040 2 [M – H – H2O – CO]– 223.040 1 C14H7O3 –0.38
195.045 0 [M – H – H2O – 2CO]– 195.045 2 C13H7O2 0.69
283.061 7 [M – H]– 283.061 2 C16H11O5 –1.62
268.031 8 [M – H – CH3]– 268.031 7 C15H8O5 –0.26 5,7-Dihydroxy-8-methx
15 222, 274, 324
240.042 5 [M – H – CH3 – CO]– 240.042 8 C14H8O4 1.34 oy flavone (Wogonin)
163.003 8 [M – H – CH3 – C7H5O]– 163.003 7 C8H3O4 –0.86
253.050 7 [M – H]– 253.050 6 C15H9 O4 –0.45
5,7-Dihydroxy flavone
16 268, 315 209.061 0 [M – H – CO2]– 209.060 8 C14H9O2 –0.80
(Chrysin)
145.029 2 [M – H – C6H4O2]– 145.029 5 C9H5O2 2.42
283.061 4 [M – H]– 283.061 2 C16H11O5 –0.88
5,7-Dihydroxy-6-metho
18 222, 270, 318 268.038 1 [M – H – CH3]– 268.037 7 C15H8O5 –1.42
xy flavone (Oroxylin A)
239.035 7 [M – H – CH3 – COH]– 239.035 0 C14H7O4 –2.88
a
Identification results obtained by comparing the retention time, UV spectra, and MS data with authentic standards.
b
glu acid: glucuronic acid

– 211 –
YANG Jun-Ran, et al. / Chin J Nat Med, 2015, 13(3): 208−214

Fig. 4 Q/TOF-MS/MS in the negative ion mode spectra of Compounds 5 (a), 13(b), 15 (c), 16 (d), and 18 (e)

Compound 5 represented a main constituent of SBG. It fragments gave the ions consistent with baicalin. Thus, com-
exhibited [M − H]− ion at m/z 445.077 7, and its MS-MS pound 5 was identified as baicalin and this identification was

– 212 –
YANG Jun-Ran, et al. / Chin J Nat Med, 2015, 13(3): 208−214

confirmed by comparison with a pure standard. Compound 13 showed excellent linearity with correlation coefficients (r2)
displayed the [M − H]¯ ion at m/z 269.045 7. The MS-MS exceeding > 0.999 (Table 2). The enrichment values of the
fragments were similar to that of Compound 5, showing five compounds are shown in Table 3, and were used to rank
[M − H − H2O]¯at m/z 251.034 3 and [M − H − CO]¯at m/z the relative binding affinities of these flavonoids. As shown in
241.050 1. On comparison with the standard, Compound Table 3, the enrichment factors for baicalin, baicalein, wogo-
13 was confirmed as baicalein. Compound 16 yielded an nin, chrysin, and oroxylin A were 38.44% ± 2.91%, 62.07% ±
[M − H]¯ ion at m/z 253.050 7. The UV spectrum showed 3.50%, 46.99% ± 1.81%, 30.00% ± 3.43%, and 26.96% ±
maximum absorption bands at 268 and 315 nm. The 1.95% at a test concentration of 50 μmol·L−1, resepctively. At
MS-MS spectra gave ions of [M − H − 45]¯ at m/z 209.061 the concentration of 100 μmol·L−1 for each ligand, the
0 and [M − H − 108]¯ at m/z 145.029 2. The fragments and enrichment factors for baicalin, baicalein, wogonin, chrysin
retention time in HPLC of Compound 16 suggested a ten- and oroxylin A were 17.99% ± 1.95%, 37.24% ± 2.13%,
tative assignment as chrysin. Compounds 15 and 18 both 29.55% ± 1.61%, 15.75% ± 2.14% and 14.39% ± 3.20%,
exhibited [M − H]¯ ions at m/z 283.161. The MS fragments respectively. These results showed that enrichment factors
and retention time in HPLC of Compound 15 were consis- were dependent upon the relative concentrations of ligand and
tent with the known compound wogonin. The MS spectra enzyme. Additionally, the affinity rank order for binding to rat
of Compound 18 were almost identical to that of Com- intestinal α-glucosidase was baicalein > wogonin > baicalin >
pound 15. However, its retention time supported its possi- chrysin ≥ oroxylin A, which corresponded to the in vitro
ble identification as oroxylin A. enzyme assay results. This work showed that the devel-
Evaluation of relative binding affinity of the inhibitors oped method can be used to rapidly rank the order of the
To further investigate their binding affinity, the five puri- binding of the ligands with respect to their affinity for
fied compounds (baicalin, baicalein, wogonin, chrysin, and high throughput screening of rat intestinal α-glucosidase
oroxylin A) were evaluated for their inhibitory activity of rat inhibitors.
intestinal α-glucosidase, respectively. Incubations of rat intes- α-Glucosidase inhibitory activity of the ligands in vitro
tinal α-glucosidase were carried out with two equimolar con- The inhibitory activity of the five compounds (baicalin,
centrations of each compound (50 μmol·L−1, 100 μmol·L−1) baicalein, wogonin, chrysin, and oroxylin A) against rat
using LC-Q/TOF-MS/MS combined with centrifugal ultrafil- intestinal α-glucosidase was subsequently conducted in vitro.
tration based binding assay as described above. The relative As shown in Table 4, baicalein showed significantinhibitory
binding affinity of the ligands in the SBG coupled towards rat activity, and baicalin, and wogonin had moderate inhibitory
intestinal α-glucosidase was compared using the values of activity, while chrysin and oroxylin A also had a subtle ef-
“enrichment factors”, as defined by Liu et al.[16] The standard fects. Therefore, the inhibitory activity decreased in the
curves for the five ligands were obtained by LC-MS and following order: baicalein > wogonin > baicalin > chrysin>

Table 2 The calibration curves, the scope of linearity, LOQ and LOD of screening of Compounds 5, 13, 15, 16, and 18 from S. bai-
calensis
Sample Calibration curves Scope (μg·mL−1) r LOQ (μg·mL−1) LOD (μg·mL−1)
Baicalin y = 24.249 x + 12.528 1.45–66.9 0.999 9 1.43 0.49
Baicalein y = 62.211 x – 9.819 3 0.84–40.5 0.999 4 0.54 0.19
Wogonin y = 65.704 x + 9.120 2 1.78–42.6 0.999 4 0.26 0.11
Chrysin y = 62.780 x + 14.859 1.43–38.1 0.999 0 0.27 0.07
Oroxylin A y = 33.922 x – 4.362 1 0.57–42.6 0.999 6 0.57 0.14

Table 3 Enrichment factors a for ligands (5, 13, 15, 16, and 18) Table 4 Inhibitory activities of the screening ligands (Compounds
to rat intestinal α-glucosidase (mean ± SD, n = 3) 5, 13, 15, 16, and 18) against rat intestinal α-glucosidase (mean ± SD,
n = 3)
Peak Sample 50 μmol·L−1 100 μmol·L−1
Peak Samples Inhibition a (%) IC50 (μmol·L−1)
5 Baicalin 38.44 ± 2.91 17.99 ± 1.95
5 Baicalin 33.34 ± 2.92 98.04 ± 4.16
13 Baicalein 62.07 ± 3.50 37.24 ± 2.13
13 Baicalein 66.67 ± 1.37 38.19 ± 1.82
15 Wogonin 46.99 ± 1.81 29.55 ± 1.61 15 Wogonin 42.62 ± 2.19 78.04 ± 2.96
16 Chrysin 30.00 ± 3.43 15.75 ± 2.14 16 Chrysin 31.33 ± 3.29 164.07 ± 5.6
18 Oroxylin A 26.96 ± 1.95 14.39 ± 3.20 18 Oroxylin A 25.65 ± 3.95 269.8 ± 7.84
a
a
Enrichment factor = (amount of compound specifically bound) Inhibition by 50 μmol·L−1 flavonoids, the IC50 of positive drug
/ (total amount of compound incubation) × 100% (acarbose) was at concentration of 3.5 μmol·L−1

– 213 –
YANG Jun-Ran, et al. / Chin J Nat Med, 2015, 13(3): 208−214

oroxylin A. The above results indicated that the ligands from [8] Pluempanupat W, Adisakwattana S, Yibchok-Anun S, et al.
SBG identified by centrifugal ultrafiltration method were all Synthesis of N-phenylphthalimide derivatives as α-glucosidase
anti-α-glucosidase agents, and that the inhibitory activity of inhibitors [J]. Arch Pharmcal Res, 2007, 30(12): 1501-1506.
these flavonoids against rat intestinal α-glucosidase corre- [9] Miyazawa M, Takahashi T, Horibe I, et al. Two new aromatic
compounds and a new D-arabinitol ester from the mushroom
sponded to the results from the LC-Q/ TOF-MS/MS binding
Hericium erinaceum [J]. Tetrahedron, 2012, 68: 2007-2010.
affinity assay.
[10] Kool J, Giera M, Irth H, et al. Advances in mass spectrome-
Conclusions try-based post-column bioaffinity profiling of mixtures [J]. Anal
and Bioanal Chem, 2011, 399(8): 2655-2668.
An LC-Q/TOF-MS/MS method coupled with centrifugal [11] van Breemen RB, Tao Y, Li W. Cyclooxygenase-2 inhibitors in
ultrafiltration was developed and successfully applied to as- ginger (Zingiber officinale) [J]. Fitoterapia, 2011, 82(1): 38-43.
sess the presence of rat intestinal α-glucosidase inhibitors in a [12] Allen Annis D, Nickbarg E, Yang XS, et al. Affinity selec-
multicomponent S. baicalensis extract. This procedure pro- tion-mass spectrometry screening techniques for small molecule
drug discovery [J]. Curr Opin Chem Biol, 2007, 11: 518-526.
vided a better tool for rapid screening and identifying AGIs
[13] Liu S, Yan J, Xing JP, et al. Characterization of compounds and
from complex plant extract matrix, compared with the com-
potential neuraminidase inhibitors from the n-butanol extract of
monly used bioassay-guided column separation technique. Compound Indigowoad Root Granule using ultrafiltration and
The availability of such a system allows a rapid identification liquid chromatography-tandem mass spectrometry [J]. J Pharm
of natural α-glucosidase inhibitors for pharmacological inves- Biomed Anal, 2012, 59: 96-101.
tigation. [14] Zhou H, Xing JP, Liu S, et al. Screening and determination for
potential α-glucosidase inhibitors from leaves of Acanthopanax
References senticosus Harms by using UF-LC/MS and ESI-MSn [J]. Phy-
tochem Anal, 2012, 24: 315-323.
[1] Lu YJ, Joerger R, Wu CQ. Study of the chemical composition
[15] Luque-Garcia JL, Neubert TA. Sample preparation for se-
and antimicrobial activities of ethanolic extracts from roots of
rum/plasma profiling and biomarker identification by mass
Scutellaria baicalensis Georgi [J]. J Agric Food Chem, 2011,
spectrometry [J]. J Chromatogr A, 2007, 1153: 259-276.
59(20): 10934-10942.
[16] Liu DT, Guo J, Luo Y, et al. Screening for ligands of human
[2] Yoon SB, Lee YJ, Park SK, et al. Anti-inflammatory effects of
retinoid X receptor-α using ultrafiltration mass spectrometry [J].
Scutellaria baicalensis water extract on LPS-activated RAW
Anal Chem, 2007, 79(24): 9398-9402.
264.7 macrophages [J]. J Ethnopharmacol, 2009, 125: 286-290.
[17] Li HL, Song FR, Xing JP, et al. Screening and structural char-
[3] Wang R, Luo JG, Kong LY. Screening of radical scavengers in
acterization of a-glucosidase inhibitors from hawthorn leaf fla-
Scutellaria baicalensis using HPLC with diode array and che-
vonoids extract by ultrafiltration LC-DAD-MSn and SORI-CID-
miluminescence detection [J]. J Sep Sci, 2012, 35(17):
FTICR-MS [J]. J Am Soc Mass Spectrom, 2009, 20: 1496-1503.
2223-2227.
[18] Han J, Ye M, Xu M, et al. Characterization of flavonoids in the
[4] Waisundara VY, Hsu A, Tan BKH, et al. Baicalin reduces mi-
traditional Chinese herbal medicine-Huangqin by liquid
tochondrial damage in streptozotocin-induced diabetic Wistar
chromatography coupled with electrospray ionization mass
rats [J]. Diabetes Metab Res Rev, 2009, 25(7): 671-677.
spectrometry [J]. J Chromatogr B, 2007, 848(2): 355-362.
[5] Lai YC, Chen CK, Tsai SF, et al. Triterpenes as α-glucosidase
[19] Liu GZ, Rajesh N, Wang XS, et al. Identification of flavonoids
inhibitors from Fagus hayatae [J]. Phytochemisty, 2012, 74: in the stems and leaves of Scutellaria baicalensis Georgi [J]. J
206-211. Chromatogr B, 2011, 879: 1023-1028.
[6] Wang SJ, Yan JF, Wang XY, et al. Synthesis and evaluation of [20] Mulabagal V, Calderón A I. Development of binding assays to
the a-glucosidase inhibitory activity of 3-[4-(phenylsulfonamido) screen ligands for Plasmodium falciparum thioredoxin and
benzoyl]-2H-1-benzopyran-2-one derivatives [J]. Eur J Med glutathione reductases by ultrafiltration and liquid chromato-
Chem, 2010, 45(3): 1250-1255. graphy/mass spectrometry [J]. J Chromatogr B, 2010, 878:
[7] Hakamata W, Kurihara M, Okuda H, et al. Design and 987-993.
screening strategies for alpha-glucosidase inhibitors based on [21] Gunawan-Puteri MDPT, Kawabata J. Novel α-glucosidase
enzymological information [J]. Curr Top Med Chem, 2009, 9: inhibitors from Macaranga tanarius leaves [J]. Food Chem,
3-12. 2010, 123: 384-389.

Cite this article as: YANG Jun-Ran, LUO Jian-Guang, KONG Ling-Yi. Determination of α-glucosidase inhibi-
tors from Scutellaria baicalensis using liquid chromatography with quadrupole time of flight tandem mass spec-
trometry coupled with centrifugal ultrafiltration [J]. Chinese Journal of Natural Medicines, 2014, 13(3): 208-214

– 214 –

You might also like