Open Access Article
This Open Access Article is licensed under a Creative Commons Attribution-Non Commercial 3.0 Unported Licence

Rapid characterization of the chemical constituents of Sanhua decoction by UHPLC coupled with Fourier transform ion cyclotron resonance mass spectrometry

Yanhui Zhaoa, Miao Wangb, Lin Suna, Xue Jianga, Min Zhao*a and Chunjie Zhao*a
aSchool of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenhe District, Shenyang 110016, Liaoning Province, China. E-mail: lab433@163.com
bSchool of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, China

Received 21st March 2020 , Accepted 23rd May 2020

First published on 10th July 2020


Abstract

Sanhua decoction, a famous Chinese herbal formula has been widely used for the treatment of stroke. In our study, a rapid, swift and straightforward analytical method with the help of UHPLC-FT-ICR-MS/MS was successfully developed for the first time to separate and identify the chemical constituents of Sanhua decoction. Chromatography was performed on a Universal XB C18 column (150 mm × 2.1 mm, 1.8 μm) using a mobile phase containing 0.1% formic acid–water (A) and acetonitrile (B). A total of 137 compounds in Sanhua decoction were identified or tentatively characterized. The findings revealed the fact that Sanhua decoction mainly contains flavonoids (in Aurantii fructus immaturus and Rheum palmatum L.), anthraquinones (in Rheum palmatum L.), coumarins (in Notopterygii Rhizoma Et Radix), phenylpropanoid glycosides, alkaloids and lignans (in Magnoliae Officmalis Cortex), which made up the key ingredients existing in Sanhua decoction. This study is hoped to be meaningful for the characterization of components in other traditional Chinese medicines, and lay the foundation for research on the pharmacology of Sanhua decoction.


1. Introduction

Traditional Chinese medicines (TCMs) play an important role in Chinese medicine due to their wide application and efficacy, particularly in the management of chronic diseases.1 TCMs are composed of complex chemical constituents for multi-target treatment, and produce synergistic effects and reduce side effects. Although there has been a lot of research on TCMs, the active ingredients of most traditional Chinese medicine prescriptions remain unknown. Therefore, developing a fast and efficient analysis method to separate and identify complex constituents in TCMs is necessary for illustrating the pharmacological material basis of TCMs.

Sanhua decoction (SHD), a famous Chinese herbal formula, is recorded in the classic traditional Chinese medicine (TCM) book Suwen Bingji Qiyi Baomingji (Plain Questions: Discourse on Mechanism for Preserving Life). It consists of four crude herbs: Zhishi (Aurantii fructus immaturus), Dahuang (Rheum palmatum L.), Houpu (Magnoliae Officmalis Cortex) and Qianghuo (Notopterygii Rhizoma Et Radix). SHD is used widely for the treatment of stroke. It can improve ischemic cerebral edema and brain blood barrier (BBB) permeability.2 It has been documented that Zhishi has neuroprotective, antioxidant, anti inflammatory and anti-apoptotic effects.3 Dahuang can protect neurons from hypoxic-ischemic brain damage.4 Houpu can protect neural damage from cerebral ischemia and reperfusion by suppressing cerebral inflammation and improving BBB function.5 Qianghuo has a function of anti-thrombosis, increasing cerebral blood flow and improving cerebral blood circulation.6 However, the chemical material basis of SHD has not been studied in detail until now.

As the complexity of traditional Chinese medicine, it is very important to establish a sensitive and reliable detection method. Chromatography-mass spectrometry (LC-MS) is widely used for its high sensitivity and specificity. Ultra high performance liquid chromatography (UHPLC) coupled with Fourier transform ion cyclotron resonance mass spectrometry (FT-ICR-MS) as the most powerful instrument with high resolution, accuracy and sensitivity play a role in analyzing different kinds of complex samples.7–10 It provides an unambiguous elemental composition and information about the isotopic abundance of ions to help determine the compositions and structures of chemical constituents. By usage of this technology, Li et al. characterized 179 constituents in Rhodiola crenulata as well as 37 prototypes and 142 metabolites in rats.11 Guan et al. characterized 120 constituents in Sijunzi decoction12 and Liu et al. characterized 174 constituents in Gegenqinlian decoction as well as 107 prototypes and 67 metabolites in rats.13 The results show that this method is useful in detecting and indenting chemical constituents and metabolites by information about accurate molecular weight and MS2.

In this study, a rapid and simple approach by UHPLC-FT-ICR-MS was established for the systematical characterization of the constituents of SHD. This constituent characterization and structural elucidation provided significant information for a pharmacological study of SHD.

2. Material and methods

2.1 Chemicals and materials

The purchase of key ingredients including, Aurantii fructus immaturus (batch number: 170[thin space (1/6-em)]901; source: Sichuan China), Rheum palmatum L. (batch number: 20[thin space (1/6-em)]181[thin space (1/6-em)]101; source: Gansu China), Magnoliae Officmalis Cortex (batch number: 181[thin space (1/6-em)]001; source: Sichuan China) and Notopterygii Rhizoma Et Radix (batch number: 13[thin space (1/6-em)]112[thin space (1/6-em)]501; source: Sichuan China) was made from Guoda pharmacy (Shenyang, China), which followed identification by Professor Jingming Jia (Department of TCM, Shenyang Pharmaceutical University, Shenyang, China). The key source of the reference compounds (purity > 98%), including sennoside B, nodakenin, narirutin, sennoside A, neohesperidin, naringenin, hesperetin, nobiletin was Shanghai Yuanye Bio-Technology Co., Ltd. (Shanghai, China); naringin, hesperidin and honokiol was attained from the National Institute for the Control of Pharmaceutical and Biological Products (Beijing, China). In addition, acetonitrile of HPLC grade, together with formic acid of LC-MS grade was attained from Fisher Scientific (Fair Lawn, NJ, USA), followed by attaining the purified water from Wahaha (Hangzhou, China).

2.2 Preparation of SHD for analysis

Following the documentary records of SHD, four constituting herbs that included Aurantii fructus immaturus (40 g), Rheum palmatum L. (40 g), Magnoliae Officmalis Cortex (40 g) and Notopterygii Rhizoma Et Radix (40 g) were soaked in water for 60 min, then decocted two times in 80 mL water for a period of 1 h each time in a glass flask and the ensuing solution was mixed and dried with the help of lyophilization. Prior to the analysis, dissolution the 0.5 g of dried powder was dissoluted by 5 mL methanol-aqueous (1[thin space (1/6-em)]:[thin space (1/6-em)]1) solution, soaking for 15 min with 80 °C water.

2.3 Instrument and analytical conditions

The sample was analyzed by the UHPLC-DAD-FT-ICR-MS system contains an Agilent 1260 UHPLC system combined with a Bruker Solarix7.0T FT-ICR-MS system (Germany) and Bruker Compass-Hystar workstation (Bruker, Germany). Chromatography was carried out on a Universal XB C18 column (150 mm × 2.1 mm, 1.8 μm; Kromat, USA) at the temperature of 35 °C. The mobile phase contained 0.1% formic acid–water (A) and acetonitrile (B). The gradient conditions were as follows: 8–12% (B) in 0 to 10 min, 12–13% (B) in 10 to 12 min, 13–18% (B) in 13 to 18 min, 18–24% (B) in 17 to 28 min, 24–30% (B) in 28 to 31 min, 30–36% (B) in 31 to 35 min, 36–55% (B) in 35 to 37 min, 55–90% (B) in 37 to 47 min, which was delivered at a flow rate of 0.2 mL min−1 and injection volume was 2 μL.

The mass spectra were performed with positive as well as negative electrospray ionization (ESI) modes, followed by setting the optimized conditions are as follows: capillary voltage, 4.5 kV; plate offset, 500 v; nebulizer gas pressure, 4.0 bar; dry gas flow rate, 8 L min−1; dry gas temperature, 200 °C; ion accumulation time, 0.15 s, and flight time, 0.6 ms. Full scan mass spectrometry data were recorded between m/z 100 and 1200 amu and the collision energy was ranged from 10 eV to 30 eV for MS/MS experiments.

3. Results and discussion

The Fig. 1 reveals the base peak ion chromatograms (BPC) of SHD in positive as well as negative ion modes, together with the respective compounds. The extract ion chromatograms (EIC) of each molecule weight were correspondingly attained for detecting the associated compound followed by presenting in support information Fig. S1. Comparing with reference compounds, eleven peaks from SHD were accurately identified by the retention times and MS fragments in positive ion mode. The peak of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 in Fig. 1C were sennoside B, nodakenin, narirutin, naringin, sennoside A, hesperidin, neohesperidin, naringenin, hesperetin, honokiol, nobiletin, respectively. The others were predicted by Bruker workstation, whether or not the mass error values between the identified molecular weights with the measured molecular weights were below 3.0 ppm. If yes, it can infer the chemical structure of each component by the retention time and MS/MS data with published literature.14–28 In the end, a total of 137 compounds were identified and characterized, whereas their layouts were presented in support information Fig. S2.
image file: d0ra02264k-f1.tif
Fig. 1 The base peak ion chromatograms (BPC) of SHT in both positive (A) and negative (B) ion modes and the corresponding compounds (C). (1) Sennoside B, (2) nodakenin, (3) narirutin, (4) naringin, (5) sennoside A, (6) hesperidin, (7) neohesperidin, (8) naringenin, (9) hesperetin, (10) honokiol, (11) nobiletin.

The MS/MS spectra of representative compounds are shown in Fig. 2, and displaying deduced fragmentation pathways are shown in Fig. 3. Except that, the formula, retention time, MS data, calculated m/z, error, ion mode and MS/MS data of other detected components were listed in Table 1.


image file: d0ra02264k-f2.tif
Fig. 2 The MS/MS spectra of the typical compounds. (A) Hesperidin, (B) nomilin, (C) emodin-O-malonyl-glucose, (D) galloyl-glucoside, (E) epicatechin, (F) honokiol, (G) magnolol, (H) acteoside, (I) magnocurarine, (J) bergaptol.

image file: d0ra02264k-f3.tif
Fig. 3 The possible fragmentation pathways of the typical compounds. (A) Hesperidin, (B) nomilin, (C) emodin-O-malonyl-glucose, (D) galloyl-glucoside, (E) epicatechin, (F) honokiol, (G) magnolol, (H) acteoside, (I) magnocurarine, (J) bergaptol.
Table 1 UHPLC-FT-ICR-MS analysis of SHDa
No. tR (min) Identification Formula Monoisotopic mass (m/z) Detected mass (m/z) Ion mode ppm MS/MS (m/z) Main source
a Ps: C represented Aurantii fructus immaturus; R represented Rheum palmatum L.; M represented Magnoliae Officmalis Cortex; N represented Notopterygii Rhizoma Et Radix.
1 2.07 Quinic acid C7H12O6 193.07066 193.07024 [M + H]+ 2.21 147.04051, 129.05446 C
2 2.64 Galloyl-glucoside C13H16O10 331.06707 331.06759 [M − H] −1.57 313.05289, 271.04772, 211.02436, 169.01338, 125.00325 R
3 2.91 1-Galloyl-di-glucoside C19H26O15 493.11989 493.12035 [M − H] −0.93 373.07705, 331.06771, 313.05667, 271.04642 R
4 3.21 Gallic acid C7H6O5 169.01425 169.01447 [M − H] −1.30 151.00284, 125.02324 R
5 4.85 Catechin-3-O-glucoside C21H24O11 451.12459 451.12518 [M − H] −1.31 289.07014, 245.08287 R
6 5.59 Rebouoside C C20H30O12 461.16645 461.16674 [M − H] −0.64 299.11268, 137.05927 M
7 6.04 Chlorogenic acid C16H18O9 353.08781 353.08824 [M − H] −1.23 191.09726, 179.76826, 173.10064 N
8 7.02 Catechin-5-O-glucoside C21H24O11 451.12459 451.12504 [M − H] −1.01 289.07156, 245.08176, 203.07109 R
9 7.79 Magnocurarine C19H24NO3+ 314.17507 314.17473 [M + H]+ 1.07 269.11618, 237.08974, 175.07514, 107.04887 M
10 10.40 Catechin C15H14O6 289.07176 289.07211 [M − H] −1.22 245.08146, 205.04962, 203.06974 R
11 10.63 Cryptochlorogenic acid C16H18O9 353.08781 353.08825 [M − H] −1.25 191.10029, 179.14054, 173.10065 N
12 10.64 Syringin C17H24O9 395.13125 395.13094 [M + Na]+ 0.80 372.08647, 343.94804, 208.09042, 137.03937 M
13 13.55 Magnoflorine C20H24NO4+ 342.16998 342.16967 [M + H]+ 0.91 297.10956, 282.08696, 265.08507, 237.08972 M
14 16.29 (R)-Oblongine C19H24NO3+ 314.17507 314.17475 [M + H]+ 1.02 269.11625, 237.09024, 175.07485, 107.04861 M
15 16.62 Epicatechin C15H14O6 289.07176 289.07213 [M − H] −1.26 245.08235, 203.07153 R
16 18.49 Teupolioside C35H46O20 785.25097 785.24931 [M − H] 2.11 649.28262, 623.38565, 477.34832 M
17 19.22 Aloe-emodin-O-di-glucoside C27H30O15 593.15119 593.15142 [M − H] −0.38 431.09851, 269.04559, 239.03448 R
18 19.31 Apigenin-6,8-di-C-glucoside C27H30O15 595.16575 595.16502 [M + H]+ 1.23 577.15636, 505.13492, 475.12405 C
19 20.32 Xanthoplanine C21H25O4N+ 356.18563 356.18528 [M + H]+ 1.01 311.12628, 296.10825, 280.10864 M
20 20.74 Asimilobine C17H17O2N 268.13321 268.13294 [M + H]+ 0.99 251.10647, 236.08474, 219.08076, 191.08617 M
21 21.26 Echinacoside C35H46O20 785.25097 785.24959 [M − H] 1.76 649.28357, 623.38647, 477.34187 M
22 21.76 Chysoeriol-6,8-di-C-glucoside C28H32O16 625.17631 625.17579 [M + H]+ 0.84 607.16748, 535.14662, 505.13284 C
23 21.95 1-Cinnamoyl-6-O-(glucosyl)-galloyl-glucoside C28H32O16 623.16176 623.16247 [M − H] −1.15 461.10814, 331.06702, 313.05827, 211.02305 R
24 22.31 Scopoletin C10H8O4 193.04954 193.04935 [M + H]+ 0.95 178.02482, 165.05373, 122.03784 C
25 22.45 2-O-Cinnamyl-β-D-glucoside C15H18O7 309.09798 309.09841 [M − H] −1.39 237.07692, 201.01967, 146.96590 R
26 22.55 Emodin-O-glucuronic acid methyl ester C22H20O11 459.09329 459.09358 [M − H] −0.65 269.04543, 241.05106, 225.05490, 213.05525 R
27 23.60 Decuroside V C20H20O10 447.12617 447.12576 [M + Na]+ 0.91 263.08937, 245.08472 N
28 23.73 Xanthotoxol C11H6O4 203.03389 203.03379 [M + H]+ 0.45 173.07564, 145.04536, 117.05273 C
29 23.73 Bergaptol C11H6O4 201.01933 201.01957 [M − H] −1.17 185.02456, 157.17256, 129.10451, 101.96320 N
30 23.76 1-O-Cinnamyl-β-D-glucoside C15H18O7 309.09798 309.09843 [M − H] −1.47 248.95978, 161.04574, 146.96419 R
31 23.77 Bergaptol-O-β-D-glucopyranoside C17H16O9 365.08671 365.08641 [M + H]+ 0.81 201.03287, 159.17264, 131.10514 N
32 24.94 Natsudaidain-3-O-glucoside C27H32O14 581.18648 581.18599 [M + H]+ 0.85 419.13174, 273.07487, 153.01846 C
33 24.97 Narirutin-4′-O-glucoside C33H42O19 743.23931 743.23875 [M + H]+ 0.75 581.18682, 435.12794, 273.07526 C
34 25.16 Eriocitrin C27H32O15 597.18140 597.18096 [M + H]+ 0.73 435.12791, 289.06947, 153.01849 C
35 25.35 Resveratrol-4′-O-glucoside C20H22O8 389.12419 389.12479 [M − H] −0.83 245.08196, 227.07138, 199.07567, 135.04384 R
36 25.47 Isolindleyin C23H26O11 477.14024 477.14067 [M − H] −0.90 313.05691, 211.02328, 169.01307, 125.02387 R
37 25.79 Lindleyin C23H26O11 477.14024 477.14071 [M − H] −1.12 313.05659, 211.02454, 169.01340, 125.02321 R
38 26.19 Limonin C26H30O8 471.20134 471.20060 [M + H]+ 0.83 425.19343, 339.19426, 161.05897 C
39 26.19 Rhein-8-O-β-D-glucoside C21H18O11 445.07763 445.07816 [M − H] −1.21 285.07559, 267.03326, 257.07879, 241.07041 R
40 26.28 Nicotiflorin C27H30O15 595.16575 595.16515 [M + H]+ 1.00 419.13403, 390.09214, 287.07785 C
41 26.28 Naringenin-7-O-glucoside C21H22O10 435.12857 435.12833 [M + H]+ 0.56 272.07462, 227.18968, 177.22102, 165.13608 C
42 26.29 Eriodictyol C15H12O6 289.07066 289.07051 [M + H]+ 0.53 153.01692, 135.06538, 107.04796 C
43 26.42 Limonin-17-β-D-glucoside C32H42O14 649.25018 649.25108 [M − H] −1.39 487.19725, 425.18954, 339.18671 C
44 26.49 Neoeriocitrin C27H32O15 597.18140 597.18134 [M + H]+ 0.52 435.12786, 289.06814, 153.01794 C
45 26.55 Isosakuranetin C16H14O5 287.09140 287.09138 [M + H]+ 0.38 153.01718, 133.065095 C
46 27.07 Alhagidin C34H44O20 771.23532 771.23656 [M − H] −1.61 635.18786, 609.19823, 161.02394 M
47 27.17 Cinnamoyl-6-O-galloyl-diglucoside C28H32O16 623.16176 623.16185 [M − H] −1.50 475.10891, 331.06765, 313.05664 R
48 27.25 Nodakenin C20H24O9 407.13476 407.13536 [M − H] −1.48 247.04625, 229.14536, 175.41575 N
49 27.29 Marmesin C14H14O4 247.09649 247.09655 [M + H]+ −0.28 153.01714 C
50 27.38 Veronicastroside C27H30O15 595.16575 595.16502 [M + H]+ 0.84 577.15147, 457.10763, 295.05903 C
51 27.40 Sennoside B C42H38O20 861.18837 861.18831 [M − H] −1.71 699.13514, 655.14385, 389.08791 R
52 27.44 Emodin-O-di-glucoside C27H30O15 593.15119 593.15112 [M − H] −1.24 269.04549, 240.04318, 225.05493 R
53 28.01 Cassiachromone C13H12O4 231.06628 231.06653 [M − H] −1.09 269.04549, 240.04321 R
54 28.96 Narirutin C27H32O14 581.18648 581.18594 [M + H]+ 0.94 435.12779, 419.13317, 273.07528 C
55 29.56 Isorhoifolin C27H30O14 579.17083 579.17056 [M + H]+ 0.47 449.14346, 303.08368, 153.01728 C
56 29.64 Chrysophanol-1,8-O-di-glucoside C27H30O14 577.15628 577.15658 [M − H] −0.52 415.10387, 253.04962, 225.05395 R
57 30.04 Acteoside C29H36O15 623.19814 623.19902 [M − H] −0.36 461.16314, 161.02265 M
58 30.15 1-Dihydro-p-coumaroyl-6-O-galloyl-glucoside C22H24O12 479.11950 479.12006 [M − H] −1.16 461.10874, 313.05591, 169.01294, 125.02296 R
59 30.21 Prunin C21H22O10 435.12857 435.12887 [M + H]+ −0.68 273.07329, 153.01674 C
60 30.22 Natsudaidai C21H22O9 419.13366 419.13349 [M + H]+ 0.39 390.09212, 389.09134, 371.08154 C
61 30.36 Naringin C27H32O14 581.18648 581.18605 [M + H]+ 0.75 435.12862, 419.13274, 273.07564 C
62 30.80 Sennoside A C42H38O20 885.18486 885.18538 [M + Na]+ −0.59 701.13594, 657.14538, 391.08807, 270.03807 R
63 30.94 Isoacteoside C29H36O15 623.19814 623.19925 [M − H] −1.77 461.16741, 161.02568 M
64 30.98 Laccaic acid-D-O-glucoside C22H20O12 475.08740 475.08820 [M − H] −0.91 431.09851, 269.03452, 253.05063, 240.04252 R
65 31.45 Rhoifolin C27H30O14 579.17083 579.17057 [M + H]+ 0.57 433.10764, 271.05967, 153.02389 C
66 31.45 Neodiosmin C28H32O15 609.18140 609.18107 [M + H]+ 0.77 301.07091, 286.04659 C
67 31.48 Isosakuranin C22H24O10 449.14422 449.14408 [M + H]+ 0.96 301.06877, 287.09012, 286.04485 C
68 31.59 Hesperidin C28H34O15 611.19705 611.19650 [M + H]+ 0.10 449.14312, 303.08581, 288.09732, 153.02175, C
69 32.03 Nodakenetin C14H14O4 245.08193 245.08221 [M − H] −1.14 229.10047, 211.64807, 175.42487 N
70 32.36 Physcion-1-O-glucuronic acid C22H20O11 459.09329 459.09368 [M − H] −0.86 283.06124, 268.03815, 212.04737, 184.05123 R
71 32.78 Diosmin C28H32O15 607.16684 607.16710 [M − H] −0.42 299.10025, 284.29568 C
609.18140 609.18081 [M + H]+ 0.96 301.06863[thin space (1/6-em)]286.04525
72 32.89 Neohesperidin C28H34O15 611.19705 611.19637 [M + H]+ 0.53 449.14228, 303.08417, 153.01734 C
73 32.89 Hesperetin-7-O-glucoside C22H24O11 465.13914 465.13958 [M + H]+ −0.92 303.08652, 288.03284, 153.01532 C
74 32.90 Sakuranetin C22H24O10 449.14422 449.14477 [M + H]+ −1.22 301.06945, 287.08765, 286.045473 C
75 32.97 Magnoloside E C28H34O15 609.18249 609.18261 [M − H] 0.19 447.15213, 315.11019, 161.02613 M
76 33.05 Homoeriodictyol C16H14O6 303.08631 303.08630 [M + H]+ 0.06 288.04856, 153.01712, 117.03375 C
77 33.18 Limocitrin-3-O-(3-hydroxy-3-methylglutarate)-glucoside C29H32O17 653.17123 653.17394 [M + H]+ −0.73 539.13891, 347.07494 C
78 33.40 1-Dihydrocinnamoyl-6-O-galloyl-glucoside C22H24O11 463.12459 463.12503 [M − H] −0.97 331.06693, 313.05656, 211.02564, 169.01332 R
79 34.30 Magnolignan B C18H20O5 315.12380 315.12417 [M − H] −0.95 267.10167, 249.09178, 221.09658, 133.06619 M
80 34.51 Meranzin C15H16O4 261.11214 261.11203 [M + H]+ 0.42 243.10142, 159.04375, 103.05456 C
81 34.59 Hesperidone hydrate C15H18O5 296.14925 296.14925 [M + HH4]+ 0.00 403.12656, 418.12351, 165.05352 C
82 35.89 Diosmetin-7-O-(6′′-O-acetyl) neohesperidoside C30H34O17 667.18688 667.18653 [M + H]+ −0.26 301.07182, 286.05412 C
83 36.14 Natsudaidain-3-O-(3-hydroxy-3-methylglutarate)-glucoside C33H40O18 725.22874 725.22824 [M + H]+ 0.47 581.18621, 419.13224 C
84 36.29 Magnolignan A C18H20O4 299.12888 299.12924 [M − H] −1.18 239.10865, 221.10746 M
85 37.01 Physcion-8-O-glucuronic acid C22H20O11 459.09329 459.09369 [M − H] −0.89 283.06138, 268.03804, 239.03501, 224.04837 R
86 38.08 Cinnamoyl-6-O-galloyl-glucoside C22H22O11 461.10894 461.10948 [M − H] −1.18 401.08794, 313.05874, 285.03987, 271.04612 R
87 39.00 Didymin C28H34O14 595.20213 595.20196 [M + H]+ 0.70 433.14783, 287.09001, 153.01794 C
88 39.08 Beta-hydroxyacteoside C29H36O16 639.19306 639.19315 [M − H] −0.14 503.14287, 477.16173, 161.02408, 133.03174 M
89 39.39 6-Methoxy-8-O-β-D-glucoside C20H24O9 407.13476 407.13521 [M − H] −1.12 253.04989, 201.09011, 146.96598 R
90 39.47 Magnoloside A C29H36O15 623.19705 623.19645 [M + H]+ 0.95 461.16607, 161.02574 M
91 39.51 Chrysophanol isomer C15H10O4 253.05063 253.05093 [M − H] −1.23 225.05517, 210.03072, 197.06054 R
92 39.52 Chrysophanol-1-O-glucoside C21H20O9 415.10346 415.10402 [M − H] −1.35 253.04884, 225.05627, 209.05942 R
93 39.70 Emodin-O-glucoside C21H20O10 431.09837 431.09867 [M − H] −0.7 269.04562, 241.04997, 225.05524 R
94 39.74 3,5,6,7,8,3′,4′-Heptamethoxy flavone C22H24O9 433.14931 433.14910 [M + H]+ 0.49 418.12356, 403.12656, 165.05354 C
95 39.79 1-P-Coumaroyl-6-O-galloyl-di-glucoside C29H36O16 639.19306 639.19367 [M − H] −0.96 477.13972, 313.05688, 211.02412, 141.03624 R
96 39.87 Sakuranin C16H14O5 287.09142 287.09127 [M + H]+ 0.46 153.01721, 133.06492 C
97 39.88 Poncirin C28H34O9 595.20213 595.20168 [M + H]+ 0.73 433.14812, 287.09110, 153.16712 C
98 39.93 Magnoloside M C29H36O15 623.19705 623.19643 [M + H]+ 0.99 461.16597, 161.02576 M
99 39.98 6′-O-trans-Feruloylnodakenin C30H32O12 583.18213 583.18148 [M − H] 1.07 247.10278, 229.44072, 175.28676 N
607.17865 607.17861 [M + Na]+ −0.02 249.11947, 177.29759
100 40.24 Chrysophanol C15H10O4 253.05063 253.05094 [M − H] −1.21 225.05537, 210.03104, 202.88598 R
101 40.93 Marmin C19H24O5 333.16965 333.16942 [M + H]+ 0.70 163.03837, 107.08582 C
102 41.15 2-(2′-hydroxypropyl)-5-methyl-7-hydroxychromone C13H14O4 233.08193 233.08217 [M − H] −1.01 189.05568, 146.96597, 129.97617 R
103 41.29 Magnolignan C C18H20O4 299.12888 299.12928 [M − H] −1.33 258.09247, 239.10852, 197.06244 M
104 41.55 Naringenin C15H12O5 273.07575 273.07562 [M + H]+ 0.48 153.17823, 133.06712 C
105 42.12 Procyanidin B C30H26O12 577.13515 577.13535 [M − H] −0.49 425.08747, 407.07637, 289.07107 R
106 42.26 Physcion-8-O-glucuronic acid methyl ester C23H22O11 473.10894 473.10946 [M − H] −1.11 283.06094, 268.03811, 240.04325 R
107 42.27 Emodin-O-malonyl-glucose C24H22O13 517.09876 517.09935 [M − H] −1.15 473.10884, 431.09157, 269.04531, 241.05131 R
108 42.51 Chrysophanol-8-O-glucoside C21H20O9 415.10346 415.10400 [M − H] −1.30 253.04984, 225.05618, 209.05959 R
109 42.51 Emodin C15H10O5 269.04555 269.04588 [M − H] −1.22 241.05016, 225.05524, 197.06027 R
110 42.53 Aloe-emodin-O-glucoside C21H20O10 431.09885 431.09886 [M − H] −1.14 269.04462, 239.03647, 211.02524 R
111 42.61 Physcion-1-O-glucoside C22H22O10 445.11402 445.11460 [M − H] −1.30 427.03257, 283.06205, 269.04607 R
112 42.61 Physcion C16H12O5 283.06120 283.06162 [M − H] −1.49 283.07696, 268.04557, 240.04266 R
113 42.62 Hesperetin C16H14O6 303.08631 303.08608 [M + H]+ 0.77 153.01741, 110.04783 C
114 42.85 Magnatriol C15H14O3 241.08702 241.08730 [M − H] −1.17 223.07597, 197.09716, 133.06719 M
115 42.97 Magnolignan E C18H18O4 297.11323 297.11365 [M − H] −1.39 225.09132, 184.05786, 183.04759 M
116 43.29 5-Hydroxy-6,7,3′,4′,5′-pentamethoxyflavone C20H20O8 389.12309 389.12283 [M + H]+ 0.67 374.09631, 359.08221, 356.08174, 328.07931 C
117 43.54 5-o-Demethylnobiletin C20H20O7 373.12818 373.12781 [M + H]+ 0.73 358.10001, 343.07224, 325.06554, 297.06973 C
118 43.78 Demethylnobiletin C20H20O8 389.12309 389.12282 [M + H]+ 0.79 359.08112, 360.07831, 341.06984 C
119 44.32 Obovatol C18H18O3 281.11832 281.11866 [M − H] −1.20 267.10174, 249.08513 M
120 44.54 Sinensetin C20H20O7 373.12818 373.12790 [M + H]+ 0.74 175.01549, 147.03194, 119.01593 C
121 44.59 Honokiol C18H18O2 265.12340 265.12413 [M − H] −0.42 224.08126, 223.07514, 197.05923 M
122 44.59 O-Prenyl-umbelliferone C14H14O3 229.08072 229.08726 [M − H] −1.04 160.01707, 142.17814 N
123 44.74 Aloe-emodin C15H10O5 269.04555 269.04598 [M − H] −1.60 253.05121, 240.04247, 211.03968 R
124 44.94 Magnaldehyde D C16H14O3 253.08702 253.08735 [M − H] −1.32 235.07963, 207.08634, 194.04012 M
125 45.13 Phelloptein C17H16O5 323.08899 323.08875 [M + Na]+ 0.76 269.07627, 231.01796 N
126 45.25 Notopterol C21H22O5 353.13945 353.13988 [M − H] −1.21 203.17578, 159.09786, 147.63478 N
377.13594 377.13559 [M + Na]+ 0.95 205.20652, 161.11072
127 45.38 Rhein C15H8O6 283.02481 283.02504 [M − H] −0.82 257.04507, 239.03461, 211.03958 R
128 45.38 Danthron C14H8O4 239.03498 239.03530 [M − H] −1.33 186.73096, 141.51394, 130.71196 R
129 45.58 Nobiletin C21H22O8 403.13874 403.13819 [M + H]+ 1.41 373.09359, 355.08584, 327.08476 C
130 45.69 Nomilin C28H34O9 515.22756 515.22753 [M + H]+ 0.05 469.21806, 455.20675, 411.21462, 161.05854 C
131 45.88 Notoptol C21H22O5 353.13945 353.13987 [M − H] −1.19 225.17472, 207.13978, 189.14075 N
377.13594 377.13563 [M + Na]+ 0.97 227.19076, 209.14289
132 46.17 Magnaldehyde E C16H14O3 253.08702 253.08735 [M + H]+ −1.32 225.09424, 184.05624, 183.05074 M
133 46.17 Magnolol C18H18O2 265.12340 265.12394 [M − H] −0.39 247.10096, 245.09517, 223.07491 M
134 46.19 p-Hydroxyphenethyl anisate C16H16O4 295.09408 295.09386 [M + Na]+ 0.73 153.07368, 138.08654 N
135 46.54 Obovaaldehyde C16H14O4 269.08193 269.08215 [M − H] −0.80 152.01478, 124.02018 M
136 46.70 Isosinensetin C20H20O7 373.12818 373.12783 [M + H]+ 0.94 358.10398, 343.08158, 315.08276 C
137 46.70 Tangeretin C20H20O7 395.11012 395.10998 [M + Na]+ 0.35 278.54947, 276.55982, 243.12708 C


3.1 Characterization of flavonoids in SHD

Flavonoids have a typical cyclohexene structure in the C ring, which is unstable and easy to break it for Diels–Alder reaction (RDA), and get the characteristic fragment ions 1,3A and 1,3B. Flavonoid glycosides have methoxy group, firstly losing the glycosyl groups, followed by losing the methyl groups, at last cleavage of the C ring. The mass spectral fragmentation of flavonoids by hesperidin as an example was described. In positive ion mode, adduct ion at m/z 611.19650 ([M + H]+) indicating an elemental composition of C28H34O15. The MS/MS spectrum (Fig. 3A) is presented that the main fragment ions are m/z 449.14312, 303.08581, 288.09732, 153.02175 as well as 136.04821. Among them, m/z 449.14312 and 303.08581 are inferred by cleaving of the glycosidic linkage and losing C6H10O5 (162 Da) or C12H20O9 (308 Da) from the ion of [M − H]+, m/z 288.09732 is the result of losing CH3 (15 Da) from m/z 303.08581, and m/z 153.02175 and m/z 136.04821 was due to the Diels–Alder reaction from m/z 288.09732. The characterization of other flavonoids in SHD was performed based on the fragmentation patterns and related literatures.14–18

3.2 Characterization of triterpenoids in SHD

Triterpenoids are prone to lose H2O (18 Da), CO (28 Da) and CO2 (44 Da), due to the presence of lactone rings. Nomilin was selected to illustrate the fragmentation pathways. In positive ion mode, adduct ion at m/z 515.22753 ([M + H]+) indicating an elemental composition of C28H34O9. The MS/MS spectrum (Fig. 3B) is presented that the main fragment ions are m/z 469.21806, 455.20675, 411.21462, 161.05854. Among them, m/z 469.21806 is inferred by losing CO and H2O (46 Da) from [M + H]+, m/z 455.20675 is presumed by losing C2H4O2 (60 Da) from [M + H]+, m/z 411.21462 is the result of losing CO2 (44 Da) from m/z 455.20675, and m/z 161.05854 is estimated by losing C18H26O7 (354 Da) from [M + H]+. The characterization of other triterpenoids in SHD was performed based on the fragmentation patterns and related literatures.15,19

3.3 Characterization of anthraquinones in SHD

The common characteristic of MS2 in anthraquinones is the continuous loss of CO (28 Da). Anthraquinone glycosides can lose glycosyl and CO, continually. Aglycones can be judged by [aglycone − H]. Anthraquinone glycosides can lose methyl-glucuronide (190 Da), cinnamoyl (148 Da), malonyl (86 Da), acetyl-glucose (204 Da), and glucose (162 Da). Emodin-O-malonylglucose was selected to illustrate the fragmentation pathways. In negative ion mode, adduct ion at m/z 517.09935 ([M − H]) indicating an elemental composition of C24H22O13. The mass spectrum (Fig. 3C) shows that the main fragment ions are m/z 473.10884, 431.09157, 269.04531 and 241.05131. Among them, m/z 473.10884 is presumed by losing of CO2 (44 Da) from [M − H], m/z 431.09157 is inferred by losing of the C3H2O3 (86 Da) from [M − H], and m/z 269.04531 is speculated by cleaving of the glycosidic linkage and losing C6H10O5 (162 Da) from m/z 431.09157, m/z 241.05131 is the result of losing CO (28 Da) at m/z 269.04531. The characterization of anthraquinone compounds in SHD is inferred from this cleavage rule and related literatures.20,21

3.4 Characterization of glucosides in SHD

Glucosides are formed by the esterification of acid with glucose. According to the type of acid, it can be divided into cinnamoyl-glucose, dihydrocinnamoyl-glucose, p-cinnamoyl-glucose, dihydro-p-cinnamoyl glucose, galloyl-glucose, and resveratrol-galloyl-glucose in rhubarb. They are vulnerable to loss of neutral fragments 148 Da, 150 Da, 164 Da, 166 Da, 170 Da, and 228 Da, respectively. Galloyl-glucose was selected to illustrate the fragmentation pathways. In negative ion mode, adduct ion at m/z 331.06759 ([M − H]) indicating an elemental composition of C13H16O10. The mass spectrum (Fig. 3D) shows that the main fragment ions are m/z 313.05289, 271.04772, 211.02436, 169.01338, and 125.00325. Among them, m/z 313.05289 is presumed by losing H2O (18 Da) from [M − H], m/z 271.04772 is inferred by losing C2H4O2 (60 Da) from [M − H], m/z 211.02436 is the result of losing C4H8O4 (120 Da) from [M − H], m/z 169.01338 is speculated by cleaving of the glycosidic linkage and losing glucose C6H10O5 (162 Da) from [M − H], and m/z 125.00325 is deduced by losing CO2 (44 Da) from m/z 169.01338. The characterization of glucosides in SHD is concluded by referring to the cleavage rule and related literature.21

3.5 Characterization of epicatechin in SHD

Epicatechin was selected to illustrate the fragmentation pathways for catechins. The adduct ion at 289.07213 ([M − H]) indicating an elemental composition of C15H14O6, and the MS/MS spectrum (Fig. 3E) is presented that the main fragment ions are m/z 245.08235, 203.07153. Among them, m/z 245.08235 is presumed by losing CO2 (44 Da) from [M − H], and m/z 203.07153 is inferred by losing of C2H2O (42 Da) from m/z 245.08235. The characterization of catechins in SHD is concluded by referring to the cleavage rule and related literatures.21–23

3.6 Characterization of lignans in SHD

Most of the lignans have the presence of allyl and hydroxyl groups, which relative positions are different, their cleavage methods will be different. Honokiol and magnolol were selected to illustrate the fragmentation pathways. Magnolol showed m/z 265.12394 ([M − H]) in the negative ion mode, which indicated the elemental composition of C18H18O2. The mass spectrum (Fig. 3F) shows that the main fragment ions are m/z 247.10096, 245.09517, 223.07491. Among them, m/z 247.10096 is presumed by losing H2O (18 Da) from [M − H], m/z 245.09517 is inferred by losing H2 (2 Da) from m/z 247.10096, and m/z 223.07491 is obtained by losing C3H6 (42 Da) from [M − H]. Honokiol showed m/z 265.12413 ([M − H]) in the negative ion mode, which indicated the elemental composition of C18H18O2. The mass spectrum (Fig. 3G) shows that the main fragment ions are m/z 224.08126, 223.07514, 197.05923. Among them, m/z 224.08126 is presumed by losing C3H5 (41 Da) from [M − H], m/z 223.07514 is inferred by losing C3H6 (42 Da) from [M − H], and m/z 197.05923 is obtained by losing of C2H2 (26 Da) by m/z 223.07514. The characterization of lignans in SHD is concluded by referring to the cleavage rule and related literatures.24,25

3.7 Characterization of phenylpropanoid glycosides in SHD

The phenylpropanoid glycosides are composed of glucose and phenethyl alcohol, which mainly based on the loss of coffee groups (162 Da). Verbascoside is chosen as an example to explain the cleavage pathway. It showed m/z 623.19814 ([M − H]) in the negative ion, which indicated an elemental composition of C29H36O15. The mass spectrum (Fig. 3H) shows that its main fragment ion is m/z 461.16314, 161.02265. Among them, m/z 461.16314 is presumed by losing C9H6O3 (162 Da) from [M − H], and m/z 161.02265 is inferred by losing C20H30O12 (462 Da) from [M − H]. The characterization of phenylpropanoid glycosides in SHD is concluded by referring to the cleavage rule and related literatures.24,25

3.8 Characterization of alkaloids in SHD

The main rupture method of alkaloids is the loss of N. (R)-Oblongine is chosen as an example to explain the cleavage pathway. It showed an m/z 314.17473 ([M + H]+) in the positive ion mode, which indicated the elemental composition of C19H24NO3+. The mass spectrum (Fig. 3I) shows that the main fragment ions are m/z 269.11625, 237.09024, 175.07485, 107.04861. Among them, m/z 269.11625 is presumed by losing C2H6NH (45 Da) from [M + H]+, m/z 237.09024 is inferred by losing CH4O (32 Da) at m/z 269.11625, m/z 175.07485 is obtained by losing C6H6O (94 Da) from m/z 269.11625, and m/z 107.04861 is speculated by losing C12H18NO2 (207 Da) from [M + H]+. The characterization of alkaloids in SHD is concluded by referring to the cleavage rule and related literature.25

3.9 Characterization of coumarins in SHD

The main cleavage mode of coumarins is loss of CO and CO2, while coumarin glycosides lose glucose firstly, and then break by the above-mentioned cleavage law. Bergaptol is chosen as an example to explain the cleavage pathway. It showed an m/z 201.01957 ([M − H]) in the negative ion mode, which indicated an elemental composition of C11H6O4. The mass spectrum (Fig. 3J) shows that the main fragment ions are m/z 185.02456, 157.17256, 129.10451, 101.96320. Among them, m/z 185.02456 is presumed by losing OH (17 Da) from [M − H], m/z 157.17256 is presumed by losing CO (28 Da) from m/z 185.02456, m/z 129.10451 is inferred by losing CO (28 Da) from m/z 157.17256, and m/z 101.96320 is obtained by losing CO (28 Da) from m/z 129.10451. The characterization of coumarins in SHD is concluded by referring to the cleavage rule and related literatures.26–28

4. Conclusions

In our study, a rapid, swift and straightforward analytical method with the help of UHPLC-FT-ICR-MS/MS was successfully developed for the first time to separate and identify the chemical constituents of SHD. A total of 137 compounds in SHD were identified or tentatively characterized, and 11 compounds were accurately identified by reference. The research indicated that SHD mainly contains 42 flavonoids (in Aurantii fructus immaturus and Rheum palmatum L.), 3 triterpenoids (in Aurantii fructus immaturus), 23 anthraquinones (in Rheum palmatum L.), 14 glucosides, 5 catechins and 2 anthranone (in Rheum palmatum L.), 11 phenylpropanoid glycosides, 5 alkaloids and 11 lignans (in Magnoliae Officmalis Cortex), 16 coumarins (in Notopterygii Rhizoma Et Radix and in Aurantii fructus immaturus),which has laid the foundation for prospective research associated with SHD. And it is hoped to be useful to identify and characterize the components in other TCMs. And it provided a basis for finding the prototype components and metabolites in the serum of containing SHD.

In this study, the structure of the compound, except for the comparison of the reference, was obtained through the combination of retention time, accurate primary and secondary mass spectrometry information, cracking rules of the same compound and literature comparison. The identification of compound structure by this method is limited to known compounds, because this method only mass spectrometry for identification.

Abbreviations

TCMsTraditional Chinese medicines
SHDSanhua decoction
ZhishiAurantii fructus immaturus
DahuangRheum palmatum L
HoupuMagnoliae Officmalis Cortex
QianghuoNotopterygii Rhizoma Et Radix
BBBBrain blood barrier
UHPLC-FT-ICR-MS/MSUltra high performance liquid chromatography coupled with Fourier transform ion cyclotron resonance mass spectrometry
BPCBase peak ion chromatograms
EICExtract ion chromatograms
RDADiels–Alder reaction

Conflicts of interest

The authors declare that there are no conflicts of interest.

Acknowledgements

The authors gratefully acknowledge the facilities and assistance provided by Dr Fei Han of the Bruker Corporation.

References

  1. L. Zhang, J. B. Yan, X. M. Liu, Z. G. Ye, X. H. Yang, R. Meyboom, K. Chan, D. Shaw and P. Duez, J. Ethnopharmacol., 2012, 140, 519–525,  DOI:10.1016/j.jep.2012.01.058.
  2. C. Zhang, G. Q. Zheng and H. J. Huang, Chin. J. Integr. Tradit. West. Med. Intensive Crit. Care, 2007, 6, 352–356,  DOI:10.3321/j.issn:1008-9691.2007.06.009.
  3. J. Bi, H. Zhang, J. Lu and W. Lei, Mol. Med. Rep., 2016, 14, 5408–5414,  DOI:10.3892/mmr.2016.5919.
  4. Y. Ma, X. Xia, J. M. Cheng and Y. Q. Kuang, Neurochem. Res., 2014, 39, 1809–1816,  DOI:10.1007/s11064-014-1395-y.
  5. X. Liu, X. Chen, Y. Zhu, K. Wang and Y. Wang, Metab. Brain Dis., 2017, 32, 1109–1118,  DOI:10.1007/s11011-017-0004-6.
  6. X. Wu, Y. B. Zhang, L. Zhang and X. W. Yang, J. Chromatogr. B: Anal. Technol. Biomed. Life Sci., 2018, 1092, 244–251,  DOI:10.1016/j.jchromb.2018.06.006.
  7. S. Forcisi, F. Moritz, B. Kanawati, D. Tziotis, R. Lehmann and P. Schmitt-Kopplin, J. Chromatogr. A, 2013, 1292, 51–56,  DOI:10.1016/j.chroma.2013.04.017.
  8. D. F. Smith, A. Kiss, F. E. Leach, E. W. Robinson, L. PasaTolic and R. M. Heeren, Anal. Bioanal. Chem., 2013, 405, 6069–6076,  DOI:10.1007/s00216-013-7048-1.
  9. Y. N. Wang, M. Zhao, Y. B. Yu, M. Wang and C. J. Zhao, RSC Adv., 2016, 6, 39642–39651,  10.1039/c6ra01428c.
  10. Y. N. Wang, M. Zhao, Y. F. Ou, B. W. Zeng, X. Y. Lou, M. Wang and C. J. Zhao, J. Chromatogr. B: Anal. Technol. Biomed. Life Sci., 2016, 1020, 120–128,  10.1039/c6ra01428c.
  11. Y. Li, Y. Y. Zhao, X. Li, T. F. Liu, X. W. Jiang and F. Han, J. Pharm. Biomed. Anal., 2017, 149, 318–328,  DOI:10.1016/j.jpba.2017.10.032.
  12. Z. Guan, M. Wang, Y. Cai, H. M. Yang, M. Zhao and C. J. Zhao, J. Chromatogr. B: Anal. Technol. Biomed. Life Sci., 2018, 1086, 11–22,  DOI:10.1016/j.jchromb.2018.04.009.
  13. T. Liu, X. M. Tian, Z. Q. Li, F. Han, B. Ji, Y. L. Zhao and Z. G. Yu, J. Chromatogr. B: Anal. Technol. Biomed. Life Sci., 2018, 1079, 69–84,  DOI:10.1016/j.jchromb.2018.02.001.
  14. H. F. Chen, W. G. Zhang, J. B. Yuan, Y. G. Li, S. L. Yang and W. L. Yang, J. Pharm. Biomed. Anal., 2012, 59, 90–95,  DOI:10.1016/j.jpba.2011.10.013.
  15. Y. He, Z. Li, W. Wang, S. Sooranna, Y. Shi, Y. Chen, Y. Chen, C. Q. Wu, J. G. Zeng, Q. Tang and H. Q. Xie, Molecules, 2018, 23, 2189,  DOI:10.3390/molecules23092189.
  16. R. Tong, M. Peng, C. Tong, K. K. Guo and S. Y. Shi, J. Chromatogr. B: Anal. Technol. Biomed. Life Sci., 2018, 1077–1078, 1–6,  DOI:10.1016/j.jchromb.2018.01.031.
  17. W. Y. Liu, C. Zhou and C. M. Yan, Chin. J. Nat. Med., 2012, 10, 456–463,  DOI:10.1016/S1875-5364(12)60087-9.
  18. X. J. Zhao, Q. Q. Liu and T. Xing, J. Southwest Jiaot. Univ., 2018, 40, 20–29,  DOI:10.13718/j.cnki.xdzk.2018.11.003.
  19. Y. Jiang, R. Liu, J. Chen, M. Liu, B. Liu, L. Z. Yi and S. Liu, J. Chromatogr. A, 2019, 1600, 197–208,  DOI:10.1016/j.chroma.2019.04.051.
  20. Y. Liu, L. Li, Y. Q. Xiao, J. Q. Yao, P. Y. Li, D. R. Yu and Y. L. Ma, Food Chem., 2016, 192, 531–540,  DOI:10.1016/j.foodchem.2015.07.013.
  21. L. Zhang, H. Y. Liu, L. L. Qin, Z. X. Zhang, Q. Wang, Q. Q. Zhang, Z. W. Lu, S. L. Wei, X. Y. Gao and P. F. Tu, J. Sep. Sci., 2015, 38, 511–522,  DOI:10.1002/jssc.201400971.
  22. J. Han, M. Ye, M. Xu, X. Qiao, H. B. Chen, B. R. Wang, J. H. Zheng and D. A. Guo, Planta Med., 2008, 74, 873–879,  DOI:10.1097/BCR.0b013e31818480b8.
  23. Y. Li, M. Wang, Q. Zhang, Y. Tian, F. G. Xu and Z. J. Zhang, Anal. Methods, 2014, 6, 1720–1727,  10.1039/c3ay41986j.
  24. K. Guo, C. Tong, Q. Fu, J. Xu, S. Shi and Y. Xiao, J. Pharm. Biomed. Anal., 2019, 170, 153–160,  DOI:10.1016/j.jpba.2019.03.044.
  25. K. P. Sun, K. M. Qin, W. D. Li, S. Y. Peng, J. Jin, B. Yang and B. C. Cai, World Chin. Med., 2019, 14, 287–291,  DOI:10.3969/j.issn.1673-7202.2019.02.007.
  26. Y. H. Li, S. Y. Jiang, Y. L. Guan, X. Liu, L. M. Zhou, S. X. Huang, H. D. Sun, S. L. Peng, Y. Zhou, 2006, 64, 405–411.  DOI:10.1365/s10337-006-0052-2.
  27. K. Xu, S. Jiang, Y. Zhou, B. Xia, X. Xu, Y. Zhou, Y. Li, M. Wang and L. Ding, J. Pharm. Biomed. Anal., 2011, 56, 1089–1093,  DOI:10.1016/j.jpba.2011.07.034.
  28. X. Liu, S. Jiang, K. Xu, H. Sun, Y. Zhou, X. Xu, J. Yi, Y. Gu and L. S. Ding, J. Ethnopharmacol., 2009, 126, 474–479,  DOI:10.1016/j.jep.2009.09.011.

Footnote

Electronic supplementary information (ESI) available. See DOI: 10.1039/d0ra02264k

This journal is © The Royal Society of Chemistry 2020