Skip to main content
Erschienen in: Chinese Medicine 1/2019

Open Access 01.12.2019 | Research

Rapid discovery of chemical constituents and absorbed components in rat serum after oral administration of Fuzi-Lizhong pill based on high-throughput HPLC-Q-TOF/MS analysis

verfasst von: Zhen Zhang, Maoyuan Jiang, Xinyi Wei, Jinfeng Shi, Zhao Geng, Shasha Yang, Chaomei Fu, Li Guo

Erschienen in: Chinese Medicine | Ausgabe 1/2019

Abstract

Background

Fuzi-Lizhong pill (FZLZP), which was first recorded in the Classic–“Taiping Huimin Heji Ju Fang” of the Song Dynasty, has been widely used to treat gastrointestinal disease in clinic for thousands of years in China. However, an in-depth understanding of the chemical constituents of FZLZP and its potential bioactive constituents is lacking.

Methods

A simple, sensitive and selective method of high-performance liquid chromatography coupled with quadrupole-time-of-flight high-definition mass spectrometry (HPLC-Q-TOF/MS) and automated data analysis (Agilent MassHunter Qualitative Analysis B.06.00 Workstation Software) was developed to simultaneously identify the chemical constituents of FZLZP and the absorbed prototypes as well as the metabolites in rat serum after the oral administration of FZLZP.

Results

Sixty-seven compounds, including alkaloids, flavonoids, triterpenes, gingerols, phenylpropanoids and volatile oil, in the FZLZP extract were tentatively characterized by comparing the retention time and mass spectrometry data and retrieving the reference literatures. Additionally, 23 prototype compounds and 3 metabolites in the rat serum samples were identified after oral administration of FZLZP, which might be the potential active components in vivo. In addition, the absorption of alkaloids decreased when Aconitum carmichaeli Debx. was in the form of combined application as a prescription compared to when it was in the form of herb powder.

Conclusions

Herein, the chemical constituent in vitro and the absorbed compounds in the serum of a traditional Chinese formula, Fuzi-Lizhong pill, were fully characterized using a rapid and comprehensive analysis approach based on high-performance liquid chromatography combined with quadrupole time-of-flight mass spectrometry coupled to MassHunter Qualitative Analysis software data processing approach. The results provide helpful chemical information on FZLZP for further pharmacology and active mechanism research. In view of the bioactive constitutes that basically were derived from these absorbed compounds in vivo, this work could provide a useful strategy to explore the bioactive substances of traditional Chinese medicine.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s13020-019-0227-z) contains supplementary material, which is available to authorized users.
ZZ and MJ contributed equally to this work.
Abkürzungen
HPLC-QTOF-MS
high-performance liquid chromatography–electrospray ionization/quadrupole-time-of-flight high-definition mass spectrometry
FZLZP
Fuzi Lizhong Pill
FZP
Fuzi powder
Group A
FZLZP group for dosed rat serum
Group B
Fuzi powder group for dosed rat serum
Group C
control group for blank rat serum

Background

Fuzi-Lizhong pill (FZLZP) is a popular Traditional Chinese medicine pill that was originally described in the Classic “Taiping Huimin Heji Ju F/ang” of the Song Dynasty (year 1102 by the Western calendar). It is composed of five herbal medicines, including Aconitum carmichaeli Debx. (Fuzi), Codonopsis pilosula (Franch.) Nannf. (Dangshen), Atractylodes macrocephala Koidz. (Baizhu), Glycyrrhiza uralensis Fisch. (Gancao) and Zingiber officinale Rosc. (Ganjiang). FZLZP is famous for warming the middle-jiao and tonifying the spleen and is used to treat spleen yang deficiency syndrome including enteritis, chronic diarrhoea, irritable bowel syndrome, abdominal pain, vomiting and spasm, peripheral chill, etc. [17]. Modern pharmacological research shows that FZLZP possesses a variety of pharmacological activities, including an increase in adaptive thermogenesis, pain relief, anti-inflammation, and spasmolytic benefits [815]. Although pharmacological activities of FZLZP have been extensively studied, very little is known about its systematic chemical constituents, and the bioactive compounds that account for its therapeutic effects remain unclear.
In our previous research, we focused on the dissolution behaviour of FZLZP in vitro and the results showed that some constituents in Aconitum carmichaeli Debx. and Glycyrrhiza uralensis Fisch., such as benzoylaconine, liquiritin and glycyrrhizic acid, were dissolved well in vitro [1618]. While FZLZP has the so-called active ingredients, there are no empirical data to prove their effectiveness as bioactive compounds. According to the theory of serum pharmacochemistry, while there are multiple components in herbs, only compounds that are absorbed into the blood have the possibility of showing pharmacological bioactivities [1924]. Therefore, simultaneous identification of systematic chemical constituents in vitro and potential active components in the blood of FZLZP are indispensable.
It was reported that the main components in Aconitum carmichaeli Debx. are monoester diterpenoid alkaloids (MDAs) and diester diterpenoid alkaloids (DDAs), which are toxicity and efficacy compounds and should be highly concerned [21]. Due to the toxicity, Fuzi is usually used in combination with other herbs as a prescription. Some researcher considered the combination to cause the reduction of the absorption of toxic compounds [21, 25]. As a typical combination, however, there are no detailed studies of this mechanism and the compound variations of FZLZP. The strategy of serum thermochemistry can provide us the accurate qualitative and the preliminary quantitative information for exploring the quantitative change of alkaloids and toxicity reducing mechanism.
Currently, LC–MS is widely applied for the analysis of herbal constituents in vitro and in vivo because of its superior sensitivity, selectivity and ability to conclusively identify the compounds [2629]. In this study, an approach of high-performance liquid chromatography (HPLC) quadrupole time-of-flight mass spectrometry (QTOF-MS) based on serum pharmacochemistry was developed to identify the phytochemical constituents of FZLZP and multiple absorbed components in rat serum.

Methods

The Minimum Standards of Reporting Checklist contains details of the experimental design, and statistics, and resources used in this study (Additional file 1).

Chemicals and materials

Nine reference compounds were obtained from Sichuan Victor Biological Technology Co. Ltd. (Chengdu China). HPLC grade Ethanol, formic acid and methanol were obtained from Fisher (ThermoFisher Scientific Inc, Waltham, MA, USA). Deionised water (18 MΩ) was prepared by distilled water through a Milli-Q system (Millipore, Milford, MA, USA). Fuzi (No. 1703003), Dangshen (No. 1705003), Baizhu (No. 1704088), Ganjiang (No. 1703060) and Gancao (No. 1703034) were purchased from Sichuan Neautus Traditional Chinese Medicine Co., Ltd. (Chengdu China) and were authenticated by Prof. Jin Pei, Department of Pharmacognosy of Chengdu University of Chinese Medicine.

Preparation of FZLZP

Fuzi, Ganjiang, Dangshen, Baizhu and Gancao were ground into fine powers and weighed according to the instructions recorded in Chinese Pharmacopoeia (2015 edition) and mixed well. Honey was heated at 116–118 °C until bright yellow uniform bubbles appeared on the surface and the honey became sticky. Mixed power and thermal refined honey were mixed at a ratio of 1:0.8 and were made into FZLZP (there is 0.153 kg crude aconite for every 1 kg FZLZP).

Preparation of FZLZP extract samples for LC/MS analysis

FZLZP (1.5 g) was weighed and reflux-extracted with 50 mL 70% ethanol for 1 h. Then, the filtered supernatant sample was rotary evaporated at 40 °C to a concentration of 15 mL, and was centrifuged at 5000 revolutions/min (rpm) for 5 min. The solution was filtered through a 0.22-μm membrane for further analysis.

Animal handling and serum sample preparation

Eighteen male Sprague–Dawley rats (200 ± 20 g) were obtained from the Sichuan Dashuo Biotechnology Co., Ltd. and were randomly divided into three groups of 6 rats each (group A, FZLZP group for dosed rat serum; group B, Fuzi powder (FZP) group for dosed rat serum; group C, control group for blank rat serum). The animal facilities and protocols conformed to the Care and Use of Laboratory Animals published by the National Institutes of Health. The experiment was approved by the ethical committee of Chengdu University of TCM (No.20161105). The rats were housed in an animal room with a controlled environment (20–25 °C, 65–69% relative humidity, 12 h dark–light cycle), and were given water and fed normal food for 1 week before the experiment. All animals were fasted overnight before the experiments and had free access to water.
The FZLZP was dissolved in 0.5% CMC-Na and were grinded to prepare the FZLZP suspension (150 mg crude drug/mL). Fuzi powder was dissolved in 0.5% CMC-Na to prepare the FZPsuspension (23 mg crude drug/mL, the concentration of FuZi was calculated by the ratio in FZLZP). Group A was intragastric administration 1.5 g/kg body weight of FZLZP suspension for 3 days. Group B was intragastric administration 0.23 g/kg body weight of FZP suspension for 3 days. Group C was intragastric administration with an equivalent volume of 0.5% CMC-Na. Blood samples were collected from the abdominal aorta 45 min after oral administration on the 3rd day and were placed at room temperature for 1 h until solidification. Then, samples were centrifuged at 3000 rpm for 10 min at 4 °C. All samples were stored at − 80 °C until analysis. Three times methanol was added to the 2 mL serum samples, vortexed and then, centrifuged at 12,000 rpm for 20 min. The supernatant was dried with nitrogen gas. The residue was redissolved in 50 μL methanol, vortexed and then, centrifuged at 12,000 rpm for 20 min, and the filtrate was used as the LC/MS sample. 10 µL aliquot was injected for HPLC/MS analysis.

HPLC-QTOF-MS analysis condition

Chromatographic analysis was performed in an Agilent 1290 HPLC system controlled with MassHunter Workstation Software (V B.05.00, Agilent Technologies Inc, Santa Clara, CA, USA). Samples were separated on an Agilent HC-C18 column (4.6 × 250 mm, 5.0 μm, Agilent Technologies Inc.) held at 35 °C and the flow rate was 1.0 mL/min with the injection volume of 10 μL. The mobile phase consisted of 0.1% formic acid–water (v/v, A) and methanol (B). The optimal gradient elution programme was as follows: 0–15 min, 95–70% A; 15–30 min, 70–48% A; 30–45 min, 48–25% A; 45–48 min, 25–15% A; 48–55 min, 15–2% A; and 55–65 min, 2–2% A.

Mass spectrometry conditions

Mass spectrometry was performed using an Agilent 6540 QTOF–MS (Agilent Corp., USA) equipped with a Dual AJS electrospray ionization (ESI) source, and the following operating parameters were used: positive mode, drying gas (nitrogen, N2); flow rate, 8.0 L/min; gas temperature, 325 °C; nebulizer, 40 psig; sheath gas temperature, 350 °C; sheath gas flow, 11 L/min; capillary voltage, 4000 V; skimmer, 65 V; OCT 1 RF Vpp, 750 V; fragmentor, 110 V. The sample collision energy was set at 10, 20, 30 and 40 V. All the operations, acquisition, and analyses of data were controlled by Agilent LCMS-QTOF Mass Hunter Acquisition Software Ver. B.06.00 (Agilent Technologies Inc.) and operated under Mass Hunter Workstation Software Version B.06.00 (Agilent Technologies Inc.).

Establishment of FZLZP database

By searching databases, such as PubMed of the US National Library Medicine and the National Institutes of Health, SciFinder Scholar of American Chemical Society and the Chinese National Knowledge Infrastructure (CNKI) of Tsinghua University, all components reported in the literature on Aconitum carmichaeli Debx., Codonopsis pilosula (Franch.) Nannf., Atractylodes macrocephala Koidz., Glycyrrhiza uralensis Fisch. and Zingiber officinale Rosc. were summarized in an Agilent PCDL software Ver. B.06.00 (Agilent Technologies Inc.) to establish a database, which includes the name, molecular formula, chemical structure and literatures of each published known compound.

Results

Characterization of chemical constituents from FZLZP

Using the optimal conditions described above, all information on the MS data that was obtained from the robust HPLC-TOF-MS analysis, indicated the retention time and precise molecular mass and provided the MS/MS data. The protonated molecular weights of all target compounds were calculated within an error of 5 ppm. The base peak chromatogram (BPC) of the FZLZP extract sample in positive and negative ion modes are shown in Fig. 1A, and the data were processed by the Agilent MassHunter Qualitative Analysis B.06.00 Workstation Software with the “find compounds by molecular formula” tool. A total of 73 peaks were obtained, and 67 compounds were identified or tentatively characterized by comparing the tR values and the MS fragment characteristics of the compounds.
The reference standards are summarized in Table 1 and their fragmentation mechanism are proposed in Fig. 2. The compounds in FZLZP which are identified by the reference standards are summarized and marked in Table 2. For example, reference standards (RS) 1 liquiritigenin in Table 1 were detected in the positive ion mode at the Rt in 24.843 min with the m/z of 257.0809 (C15H13O4). Its MS/MS data were shown as m/z of 239.0698[M + H–H2O]+, 137.0234 [C7H4O3 + H]+, 121.0293[C8H8O + H]+ and 120.0721 [C7H4O3 + H–OH]+. And the compound 29 in Table 2 were detected in the positive ion mode at the Rt in 24.785 min with the m/z of 257.0819 (C15H13O4), 239.0707[M + H–H2O]+ and 137.0235 [C7H4O3 + H]+. Then compound 29 were characterized as liquiritigenin. Similar to the identification process above, among 67 compounds, 9 compounds were identified as benzoylaconine, benzoylmesaconine, benzoylhypaconine, mesaconitine, liquiritigenin, isoliquiritigenin, glycyrrhizic acid, glycyrrhetinic acid and atractylenolide II. The MS data of the (+) ESI–MS spectra are shown in Table 2.
Table 1
Retention time, m/z values of ions of reference standards
Peak no.
Rt (min)
Systematic name
Molecular formula
[M + H]+
[M + Na]+
Fragmentations (m/z)
Measured mass (m/z)
Error (ppm)
Measured mass (m/z)
Error (ppm)
1
24.843
Liquiritigenin
C15H12O4
257.0809
0.3890
  
257.0809[M + H]+, 239.0698[M + H–H2O]+, 137.0234 [C7H4O3 + H]+, 121.0293 [C8H8O + H]+, 120.0721 [C7H4O3 + H–OH]+
2
27.507
Benzoylmesaconine
C31H43NO10
590.2952
− 1.3553
590.2952[M + H]+, 572.2832[M + H–H2O]+, 558.2683[M + H-CH3OH]+, 540.2580[M + H–CH3OH–H2O]+
3
28.228
Benzoylaconine
C32H45NO10
604.3130
2.3167
604.3130[M + H]+, 586.2995[M + H–H2O]+, 572.2852[M + H–CH3OH]+
554.2735[M + H–2H2O]+, 540.2577[M + H–CH3OH]+, 522.2475[M + H–2CH3OH–H2O]+
4
29.152
Benzoylhypaconine
C31H43NO9
574.3003
− 1.3930
574.3003[M + H] + , 542.2741[M + H–CH3OH]+, 524.2615[M + H–CH3OH–H2O]+, 510.2477[M + H–2CH3OH]+
5
31.663
Mesaconitine
C33H45NO11
632.3064
− 0.1582
632.3064[M + H]+, 600.2787[M + H–CH3OH]+, 572.2853[M + H–AcOH]+, 540.2594[M + H–AcOH–CH3OH]+, 512.2637[M + H–AcOH–CH3OH–CO]+,
6
39.648
Isoliquiritigenin
C15H12O4
257.0809
0.3890
  
257.0809[M + H]+, 239.0692[M + H–H2O]+, 137.0235[C7H4O3 + H]+, 121.0287 [C8H8O + H]+, 120.0514 [C7H4O3 + H–OH]+
7
48.854
Atractylenolide II
C15H20O2
233.1538
0.8578
  
233.1538[M + Na]+, 215.1440[M + Na–H2O]+, 187.1484[M + Na–CH2O2]+, 159.1165[M + Na–CH2O2–C2H4]+, 145.101 [M + Na–CH2O2–C3H6]+
131.0856[M + Na–CH2O2–C4H8]+, 105.0702[M + Na–CH2O2–C4H8–C2H2]+,
8
49.134
Glycyrrhizic acid
C42H62O16
  
845.3947
2.0109
845.3947[M + Na]+, 669.3614[M + Na–(GluA–H2O)]+
9
55.125
Glycyrrhetinic acid
C30H46O4
471.3458
− 2.3337
  
471.3458[M + H]+, 453.3349[M + H–H2O]+, 435.3244[M + H-2H2O]+
Table 2
Identification information of constituents in vitro of FZLZP by HPLC-ESI/QTOF/MS
Peak no.
Rt (min)
Systematic name
Molecular formula
Molecular weight
[M + H]+
[M + Na]+
Fragmentations (m/z)
Source
Measured mass (m/z)
Error (ppm)
Measured mass (m/z)
Error (ppm)
1
5.091
l-Pyroglutamic acid
C5H7NO3
129.0426
130.0505
4.6136
  
130.0505[M + H]+, 112.0123[M + H–H2O]+, 84.0449[M + H–HCOOH]+
Dangshen
2
8.051
Codonopsine
C14H21NO4
267.1471
268.1543
0
  
268.1543[M + H]+, 250.1451[M + H–H2O]+,
205.0863[M + H–2CH3OH]+
Dangshen
3
9.229
5-hydroxymethyfurfural
C6H6O3
126.0317
127.0394
3.1486
  
127.0394[M + H]+, 109.0291[M + H–H2O]+
Dangshen
4
9.398
Karakolidine
C22H35NO5
393.2515
394.2590
0.5072
  
394.2590[M + H]+, 376.2489[M + H–H2O]+, 358.2371[M + H–2H2O]+
Fuzi
5
10.142
Phenylalanine
C9H11NO2
165.0790
166.0872
5.4188
  
166.0872[M + H]+, 120.0817[M + H–HCOOH]+
Dangshen
6
11.288
Senbusine A
C23H37NO6
423.2621
424.2696
0.4713
  
424.2696[M + H]+, 406.2579 [M + H–H2O]+
Fuzi
7
11.407
9-OH-senbusine A
C23H37NO7
439.2570
440.2635
− 1.8170
  
440.2635[M + H]+, 422.2531[M + H–H2O]+, 408.2318[M + H–CH3OH]+
Fuzi
8
12.042
16-β-hydroxycardiopetaline
C21H33NO4
363.2410
364.2480
− 0.5490
  
364.2480[M + H]+, 346.2372[M + H–H2O]+, 328.2273[M + H–2H2O]+
Fuzi
9
12.389
Mesaconine
C24H39NO9
485.2625
486.2697
− 0.2056
  
486.2697 M + H]+, 468.2573[M + H–H2O]+, 436.2323[M + H–H2O–CH3OH]
Fuzi
10
12.578
Songorine
C22H31NO3
357.2304
358.2382
1.3957
  
358.2382[M + H]+, 340.2267[M + H–H2O]+
Fuzi
11
12.908
Karakoline
C22H35NO4
377.2566
378.2639
0
  
378.2639[M + H]+, 360.2533[M + H–H2O]+
Fuzi
12
13.081
Isotalatizidine
C23H37NO5
407.2672
408.2743
− 0.2449
  
408.2743[M + H]+, 390.2630[M + H–H2O]+, 372.2517[M + H–2H2O]+, 358.2374[M + H–H2O–CH3OH]+
Fuzi
13
13.109
Senbusine B
C23H37NO6
423.2621
424.2707
3.0640
  
424.2707[M + H]+, 406.2584 [M + H–H2O]+
Fuzi
14
13.937
14-Acetylkarakoline
C24H37NO5
419.2672
420.2750
1.4276
  
420.2750[M + H]+, 402.1695[M + H–H2O]+,
356.1122[M + H–H2O–2CH3OH]+,
Fuzi
15
14.091
Aconine
C25H41NO9
499.2781
500.2850
− 0.7995
  
500.2850[M + H]+, 482.2741[M + H–H2O]+, 468.2564[M + H–CH3OH]+, 450.2478[M + H–H2O–CH3OH]+, 436.2309[M + H–2CH3OH]+, 418.2209[M + H–H2O–2CH3OH]+
Fuzi
16
14.380
Hetisine
C20H27NO3
329.1991
330.2064
0
  
330.2064[M + H]+, 312.1951[M + H–H2O]+
Fuzi
17
15.319
Hypaconine
C24H39NO8
469.2676
470.2744
− 0.8506
  
470.2744[M + H]+, 453.2301[M + H–OH]+, 438.2474[M + H–CH3OH]+, 406.2212[M + H–2CH3OH]+, 374.1941[M + H–3CH3OH]+
Fuzi
18
15.810
Fuzitine
C20H23NO4
341.1627
342.1697
− 0.8767
  
342.1697[M + H]+, 324.1026[M + H–H2O]+
Fuzi
19
16.070
Fuziline
C24H39NO7
453.2727
454.2800
0.2201
  
454.2800[M + H]+, 436.2677[M + H–H2O]+, 418.2583[M + H–2H2O]+, 404.2443[M + H–H2O–CH3OH]+, 386.2295[M + H–2H2O–CH3OH]+, 354.2069[M + H–2H2O–2CH3OH]+
Fuzi
20
16.248
Tau-cadinol
C15H26O
222.1984
245.1852
− 9.7884
  
245.1852[M + H]+, 213.0195[M + H–CH3OH]+, 199.1252[M + H–CH3OH–CH3]+, 184.9885[M + H–CH3OH–2CH3]+,
169.0055[M + H–CH3OH–3CH3]+,
Ganjiang
21
16.573
Neoline
C24H39NO6
437.2777
438.2848
− 0.4563
  
438.2848 M + H]+, 420.2756[M + H–H2O]+, 388.2478[M + H–H2O–CH3OH]+, 370.2365[M + H–2H2O–CH3OH]+, 356.2213[M + H–H2O–2CH3OH]+
Fuzi
22
16.743
Talatisamine
C24H39NO5
421.2828
422.2899
0.4736
  
422.2899[M + H]+, 390.2621[M + H–CH3OH]+, 358.2349[M + H–2CH3OH]+
Fuzi
23
18.651
Chasmanine
C25H41NO6
451.2934
452.3008
02210
  
452.3008[M + H]+, 420.2737[M + H–CH3OH]+
Fuzi
24
19.739
Geranial
C10H16O
152.1201
153.1275
0.6530
  
153.1275[M + H]+, 135.1162[M + H–H2O]+, 125.0940[M + H–CO]+
Ganjiang
25
20.390
14-Acetyltalatizamine
C26H41NO6
463.2934
464.3014
1.5076
  
464.3014[M + H]+, 432.2753[M + H–CH3OH]+, 414.2645[M + H–CH3OH–H2O]+, 400.2486[M + H–2CH3OH]+, 372.2522[M + H–CH3OH–AcOH]+
Fuzi
26
21.828
7-hydroxycoumarin
C9H6O3
162.0317
163.0395
3.0667
  
163.0395[M + H]+, 145.0627[M + H–H2O]+
Baizhu
27
23.891
Schaftoside
C26H28O14
564.1479
565.1542
− 1.7694
  
565.1542[M + H]+, 547.1434[M + H–H2O]+, 529.1303[M + H–2H2O]+, 511.1220[M + H–3H2O]+
Gancao
28
24.041
Scopoletin
C10H8O4
192.0423
193.0500
2.5900
  
193.0500[M + H]+, 161.0603[M + H–CH3OH]+
Baizhu
29#
24.785
Liquiritigenin
C15H12O4
256.0736
257.0819
4.2788
  
257.0819[M + H]+, 239.0707[M + H–H2O]+, 137.0235[C7H4O3 + H]+, 121.0280[C8H8O + H]+, 120.0525 [C7H4O3 + H–OH]+
Gancao
30#
27.065
Benzoylmesaconine
C31H43NO10
589.2887
590.2959
− 0.1694
590.2959[M + H]+, 572.2826[M + H–H2O]+, 558.2663[M + H–CH3OH]+ 540.2573[M + H–CH3OH–H2O]+
Fuzi
31
27.325
Isoviolanthin
C27H30O14
578.1636
579.1700
− 1.3812
  
579.1700[M + H]+, 561.1588[M + H–H2O]+, 543.1485[M + H–2H2O]+, 525.1382[M + H–3H2O]+
Gancao
32#
27.614
Benzoylaconine
C32H45NO10
603.3043
604.3114
− 0.3309
  
604.3114[M + H]+, 587.2801[M + H–OH]+, 554.2711[M + H–2CH3OH]+
Fuzi
33#
28.595
Benzoylhypaconine
C31H43NO9
573.2938
574.3011
0
  
574.3011[M + H]+, 542.2745[M + H–CH3OH]+,,510.2457[M + H–2CH3OH]+
Fuzi
34
28.748
Lobetyolinin
C26H38O13
558.2312
  
581.2203
− 0.3441
581.2203[M + Na]+, 419.1709[M + Na–C6H10O5]+
Dangshen
35
31.019
Liquiritin apioside or Isoliquiritin apioside
C26H30O13
550.1686
551.1751
− 1.4514
  
551.1751[M + H]+, 419.1333[M + H–(Apiose–H2O)]+, 257.0830[M + H–(Apiose–H2O)–(Glc–H2O)]+
Gancao
36#
31.163
Mesaconitine
C33H45NO11
631.2993
632.3067
0.3163
632.3067[M + H]+, 614.1110[M + H–H2O]+,
600.2748[M + H–CH3OH]+, 572.2834[M + H–AcOH]+
Fuzi
37
31.423
7-methoxy-liquiritin
C22H22O9
430.1264
431.1332
− 1.1597
  
431.1332[M + H]+, 269.0811[M + H–(Glc–H2O)]+
Gancao
38
31.646
14-Benzoylneoline
C31H43NO7
541.3040
542.3135
4.2411
  
542.3135[M + H]+, 524.3010[M + H–H2O]+,
510.2731[M + H–CH3OH]+, 492.2733[M + H–H2O–CH3OH]+
Fuzi
39
31.659
Dehydrated benzoylhypaconine
C31H41NO8
555.2832
556.2906
0.1798
  
556.2906[M + H]+, 524.2647[M + H–CH3OH]+, 492.2381[M + H–2CH3OH]+
Fuzi
40
31.683
Liquiritin or Isoliquiritin
C21H22O9
418.1264
419.1335
0.4771
  
419.1335[M + H]+, 257.0811[M + H–(Glc–H2O)]+
Gancao
41
31.921
Aconifine
C34H47NO12
661.3098
662.3172
0.1509
  
662.3172[M + H]+, 644.3095[M + H–H2O]+,
626.1346 [M + H–2H2O]+
Fuzi
42
32.100
Hypaconitine
C33H45NO10
615.3043
616.3116
0
  
616.3116[M + H]+, 584.2843[M + H–CH3OH]+
556.2899[M + H–C2H4O2]+, 524.2533[M + H–C2H4O2–CH3OH]+, 496.2678[M + H–C2H4O2–CH3OH–CO]+
Fuzi
43
32.245
Formononetin
C16H12O4
268.0736
269.0814
2.2298
  
269.0814[M + H]+, 254.0580[M + H–CH3]+, 237.0536[M + H–CH3OH]+, 225.0554[M + H–CH3–CO]+, 213.0908[M + H–C2O2]+, 181.0666[M + H–C2O2–CH3OH]+
Gancao
44
32.528
Aconitine
C34H47NO11
645.3149
646.3216
− 0.9283
  
646.3216[M + H]+, 628.3140[M + H–H2O]+,
596.2849[M + H–H2O–CH3OH]+
Fuzi
45
33.241
Deoxyaconitine
C34H47NO10
629.3200
630.3273
0
  
630.3273[M + H]+, 598.3070[M + H–CH3OH]+
Fuzi
46
36.853
Echinatin
C16H14O4
270.0892
271.0963
− 0.7377
  
271.0963[M + H]+, 253.0850[M + H–H2O]+
Gancao
47
38.085
Benzoic acid
C7H6O2
122.0368
123.0447
4.8763
  
123.0447[M + H]+, 77.0379[M + H–HCOOH]+
Baizhu
48#
39.763
Isoliquiritigenin
C15H12O4
256.0736
257.0814
2.334
  
257.0814[M + H]+, 239.0704[M + H–H2O]+, 137.0235[C7H4O3 + H]+, 121.0277[C8H8O + H]+, 120.0527 [C7H4O3 + H–OH]+
Gancao
49
40.720
Glycycoumarin
C21H20O6
368.1260
369.1345
3.2508
  
369.1345[M + H]+, 333.2235[M + H–2H2O]+,
313.1057 [M + H–C4H8]+,
Gancao
50
41.513
6-gingerdione
C17H24O4
292.1675
293.1736
− 2.7520
  
293.1736[M + H]+, 275.1650[M + H–H2O]+
257.1517[M + H–2H2O]+
Ganjiang
51
42.593
Kumatakenin
C17H14O6
314.0790
315.0859
− 1.2694
  
315.0859[M + H]+, 298.2146[M + H–OH]+,
279.0782[M + H–2H2O]+
Ganjiang
52
43.486
6-gingerol
C17H26O4
294.1831
  
317.1737
4.4140
317.1771[M + Na]+, 299.2546[M + Na-H2O]+
Ganjiang
53
43.507
Gingerenone-A
C21H24O5
356.1624
357.1710
3.6397
  
357.1710[M + H]+, 339.2718[M + H–H2O]+, 321.2612[M + H–2H2O]+
Ganjiang
54
43.544
6-shogaol
C17H24O3
276.1725
277.1795
1.0823
  
277.1795[M + H]+, 259.1694[M + Na–H2O]+,
Ganjiang
55
45.779
lupiwighteone
C20H18O5
338.1154
339.1239
3.5385
  
339.1239[M + H]+, 321.2818[M + H–H2O]+
Gancao
56
46.339
Atractylenolide III
C15H20O3
248.1412
245.1485
0
  
249.1485[M + H]+, 231.1389[M + H–H2O]+, 175.0751[M + H–H2O–2CO]+, 163.0756[M + H–H2O–C5H8]+
Baizhu
57
48.364
Gancaonin L
C20H18O6
354.1103
355.1189
3.6607
  
355.1189[M + H]+, 337.2536[M + H–H2O]+
Gancao
58
48.398
Licoricesaponin G2
C42H62O17
838.3987
839.4076
1.9061
  
839.4076[M + H]+, 663.3722[M + H–(GluA–H2O)]+, 469.3308[M + H–2 (GluA–H2O)–H2O]+
Gancao
59#
48.887
Atractylenolide II
C15H20O2
232.1463
233.1541
2.1445
  
233.1541[M + Na]+, 187.1485[M + Na–CH2O2]+,
159.0806[M + Na–CH2O2–C2H4]+, 145.1013 [M + Na–CH2O2–C3H6]+, 131.0857[M + Na–CH2O2–C4H8]+, 105.0703[M + Na–CH2O2–C4H8–C2H2]+
Baizhu
60#
49.296
Glycyrrhizic acid
C42H62O16
822.4038
823.4130
2.3075
  
823.4130 [M + H]+, 647.3793[M + H–(GluA–H2O)]+
Gancao
61
49.667
Farnesal
C15H24O
220.1827
221.1907
3.1647
  
221.1907 M + H]+, 192.9740[M + H–CO]+
Ganjiang
62#
49.841
Glycyrrhetinic acid
C30H46O4
470.3396
471.3488
4.031
  
471.3488[M + H]+, 453.3354[M + H–H2O]+, 435.3224[M + H–2H2O]+, 425.3378[M + H–HCOOH]+
Gancao
63
50.671
Licorice saponin B2
C42H64O15
808.4245
  
831.4151
1.6838
831.4151 [M + Na]+, 655.3825[M + Na–(GluA–H2O)]+,
479.3547[M + Na–2 (GluA–H2O)]+
Gancao
64
51.232
Licoricone
C22H22O6
382.1416
383.1502
3.3929
  
383.1502[M + H]+, 355.1587[M + H–C2H4]+
Gancao
65
51.390
Atractylenolide I
C15H18O2
230.1307
231.1383
1.2979
  
231.1383[M + H]+, 185.1326[M + H–HCOOH]+,
157.1012[M + H–HCOOH–C2H4]+, 105.0701 [M + H–HCOOH–2C2H4–2C]+
Baizhu
66
52.950
Neoglycyrol
C21H18O6
366.1103
367.1165
− 0.5447
  
367.1165[M + H]+, 349.2239[M + H–H2O]+,
335.2389[M + H–CH3OH]+,
317.2283[M + H–2H2O–CH3OH]+
Gancao
67
54.310
Licorice-saponin J2
C42H64O16
824.4194
825.4286
2.3018
  
825.4286[M + H]+, 649.3906 [M + H–(GluA–H2O)]+, 455.3537[M + H-2 (GluA–H2O)–H2O]+, 437.3435 [M + H-2 (GluA–H2O)–2H2O]+
Gancao
#Indicates compounds identified by comparing with the reference standards
The remaining 58 compounds were tentatively characterized based on their chromatographic and spectrometric data, referring to previous literature [25, 3033]. For example, MS2 spectra of compound 4 (molecular ion at m/z [M + H]+ 394.2590) in Table 2 gave characteristic fragment ions of [M + H–H2O]+ at m/z 376.2489 and [M + H–2H2O]+ at m/z 358.2371. Thus, it corresponded to Karakolidine by comparison with literature data [30]. Moreover, MS2 spectra of compound 12 (molecular ion at m/z [M + H]+ 408.2743) in Table 2 gave characteristic fragment ions of [M + H–H2O]+ at m/z 390.2630, 372.2517[M + H–2H2O]+ and [M + H–CH3OH]+ at m/z 358.2374. Then it was identified as Isotalatizidine. All the MS data of the (+) ESI–MS spectra are shown in Table 2. Besides, all the structures of the compounds identified are shown in Figs. 3 and 4. The deriving herb for each compound was also assigned. The majority of constituents are identified as alkaloids, flavonoids, triterpenes, gingerols, phenylpropanoids and volatile oil.

Characterization of the absorbed chemical constituents in rat serum

Identification of the bioactive chemical prototype constituents in rat serum

As the results of constituents in rat serum show in Table 3, by comparing the tR values and MS fragment characteristics between compounds in serum and compounds in FZLZP extract, 10 alkaloid components sourced from Aconitum carmichaeli Debx. were identified, including benzoylaconine, benzoylmesaconine, benzoylhypaconine, mesaconitine, Hypaconitine, fuziline, neoline, talatisamine, chasmanine, and 14-acetyltalatizamine. These constituents have been reported as parts of the main constituents with significant effects of analgesia, anti-inflammation, thermogenesis and increasing blood oxygen in Fuzi [34, 35]. The MS data of the (+) ESI–MS spectra are shown in Table 3. For example, MS2 spectra of compound 19 in Table 2 was detected at the Rt in 16.070 min with the molecular ion at m/z 454.2800[M + H]+ and gave characteristic fragment ions of [M + H–H2O]+ at m/z 436.2677. Similarly, MS2 spectra of compound 2 in Table 3 was detected at the Rt in 16.615 min with the molecular ion at m/z 454.2808[M + H]+ and gave characteristic fragment ions of [M + H–H2O]+ at m/z 436.0243. Thus, compound 2 in Table 3 was identified as the absorbed prototype of Fuziline in rat serum. The other alkaloid components were identified in a similar way.
Table 3
Characterization of chemical constituents in vivo and metabolites of FZLZP by HPLC-ESI/QTOF/MS
Peak no.
Rt (min)
Systematic name
Molecular formula
Molecular weight (Da)
[M + H]+
[M + Na]+
Fragmentations (m/z)
Source/prototype
Measured value (Da)
Error (ppm)
Measured value (Da)
Error (ppm)
1
4.841
l-Pyroglutamic acid
C5H7NO3
129.0426
130.0498
− 0.7689
  
130.0498[M + H]+, 112.9741[M + H–H2O]+
Dangshen
2
16.615
Fuziline
C24H39NO7
453.2727
454.2808
1.981
  
454.2808[M + H]+, 436.0243[M + H–H2O]+
Fuzi
3
17.021
Talatisamine
C24H39NO5
421.2828
422.2905
0.9472
  
422.2905[M + H]+, 390.2651[M + H–CH3OH]+, 359.3263[M + H–CH2OH–CH3OH]+
Fuzi
4*
24.357
Glucuronide conjugation metabolite
C21H20O10
432.1056
433.1132
0.6927
  
433.1132[M + H]+, 257.0843[M + H–(GluA–H2O)]+
Liquiritigenin
5
25.811
Liquiritigenin
C15H12O4
256.0736
257.0819
4.2788
  
257.0819 [M + H]+, 239.0713[M + H–H2O]+, 137.0237[C7H4O3 + H]+
Gancao
6
27.236
Benzoylmesaconine
C31H43NO10
589.2887
590.2948
− 2.033
  
590.2948[M + H]+, 558.2657[M + H–CH3OH]+ 540.2537[M + H–CH3OH–H2O]+, 508.2218[M + H-2CH3OH–H2O]+
Fuzi
7
27.520
Benzoylaconine
C32H45NO10
603.3043
604.3134
2.97
  
604.3134[M + H]+, 540.6158[M + H-2CH3OH]+, 508.8095[M + H-3CH3OH]+
Fuzi
8
28.379
Liquiritin or Isoliquiritin
C21H22O9
418.1264
  
441.1144
− 2.72
441.1144 [M + Na]+, 424.0979 [M + Na–OH]+, 350.8191[M + Na–C6H3O]+
Gancao
9
28.595
Benzoylhypaconine
C31H43NO9
573.2938
574.3025
0
574.3025[M + H]+, 443.8613[M + H-3CH3OH–H2O–HO]+
Fuzi
10
31.405
Mesaconitine
C33H45NO11
631.2993
632.3079
2.2141
  
632.3079[M + H]+, 599.9372[M + H–CH3OH]+, 540.2653[M + H–AcOH–CH3OH]+
Fuzi
11
32.453
Hypaconitine
C33H45NO10
615.3043
616.3089
− 4.381
  
616.3089[M + H]+, 597.8211 [M + H–H2O]+, 556.2792[M + H–C2H4O2]+
Fuzi
12*
33.299
Glucuronide conjugation metabolite
C30H47NO13
629.3047
630.3295
27.7640
  
630.3295 [M + H]+, 454.8397[M + H–(GluA–H2O)]+
Fuziline
13*
33.165
Glucuronide conjugation metabolite
C21H20O10
432.1056
433.1145
3.6942
  
433.1145[M + H]+, 257.0829[M + H–(GluA–H2O)]+
Isoliquiritigenin
14
40.710
Isoliquiritigenin
C15H12O4
256.0736
257.0807
− 0.3889
  
257.0807[M + H]+, 239.1624[M + H–H2O]+
Gancao
15
42.275
6-gingerdione
C17H24O4
292.1675
293.1734
− 4.4342
  
293.1734[M + H]+, 275.1586[M + H–H2O]+
Ganjiang
16
42.514
Formononetin
C16H12O4
268.0736
269.0799
− 3.3447
  
269.0799[M + H]+, 181.0511[M + H–C2O2–CH3OH]+
Gancao
17
44.584
14-Acetyltalatizamine
C26H41NO6
463.2934
464.3015
1.7230
  
464.3015[M + H]+, 446.2652[M + H– H2O]+, 432.6414[M + H–CH3OH]+
Fuzi
18
46.555
6-gingerol
C17H26O4
294.1831
295.1905
0.3388
  
295.1905[M + H]+, 263.1618[M + H–CH3OH]+, 179.1028[M + H–C7H15O]+
Ganjiang
19
46.980
6-shogaol
C17H24O3
276.1725
277.1781
− 6.1332
  
277.1794[M + H]+, 260.1816[M + Na–OH]+, 245.1533[M + H–CH3OH]+
Ganjiang
20
47.690
Atractylenolide II
C15H20O2
232.1463
233.1533
− 1.2867
  
233.1533[M + Na]+, 187.1487[M + Na–CH2O2]+,
159.1179[M + Na–CH2O2–C2H4]+, 145.1005[M + Na–CH2O2–C3H6]+
Baizhu
21
48.102
Chasmanine
C25H41NO6
451.2934
  
474.2841
3.1627
474.2841[M + H]+, 442.0836[M + H–CH3OH]+
Fuzi
22
49.895
Glycyrrhizic acid
C42H62O16
822.4038
823.4094
− 2.0646
  
823.4094[M + H]+, 647.3792[M + H–(GluA–H2O)]+
Gancao
23
50.826
Atractylenolide I
C15H18O2
230.1307
231.1382
0.8653
  
231.1382[M + H]+, 105.9823[M + H–HCOOH–2C2H4–2C]+
Baizhu
24
51.095
Neoline
C24H39NO6
437.2777
  
460.2669
− 0.2173
460.2669 [M + Na]+, 442.2666[M + Na–H2O]+
Fuzi
25
54.144
7-hydroxycoumarin
C9H6O3
162.0317
163.0396
3.6801
  
163.0396[M + H]+, 145.5012[M + H–H2O]+
Baizhu
26
56.004
Glycyrrhetinic acid
C30H46O4
470.3396
471.3479
− 2.122
  
471.3479[M + H]+, 453.4285[M + H–H2O]+
Gancao
* Indicates metabolites
Six compounds sourced from Glycyrrhiza uralensis Fisch. were identified, including 3 flavonoids, namely, liquiritigenin, isoliquiritigenin, and formononetin and 2 triterpenes, namely, glycyrrhetinic acid and glycyrrhizic acid. The MS data of the (+)ESI–MS spectra are shown in Table 3. For example, MS2 spectra of compound 48 in Table 2 was detected at the Rt in 39.763 min with the molecular ion at m/z 257.0814[M + H]+ and gave characteristic fragment ions of 239.0704[M + H–H2O]+, 137.0235[C7H4O3 + H]+, 121.0277[C8H8O + H]+, 120.0527 [C7H4O3 + H–OH]+. Similarly, MS2 spectra of compound 14 in Table 3 was detected at the Rt in 40.710 min with the molecular ion at m/z 257.0807[M + H]+ and gave characteristic fragment ions of [M + H–H2O]+ at m/z 239.1624. Thus, compound 14 in Table 3 was identified as the absorbed prototype of Isoliquiritigenin in rat serum. Furthermore, liquiritin or isoliquiritin may also have been found, but further comparison with reference compounds is needed to identify these isomers. The flavonoids and triterpenes in Glycyrrhiza uralensis Fisch. have been reported as having significant anti-inflammatory, abirritation and immunoregulation effects [3638].
7-Hydroxycoumarin, atractylenolide I and atractylenolide II have been identified as bioactive chemical constituents sourced form Atractylodes macrocephala Koidz. (Baizhu) and were found as the main institutes with the effect of anti-inflammatory, antitumor and gastrointestinal regulation in Baizhu [3942]. The MS data of the (+) ESI–MS spectra are shown in Table 3. For example, MS2 spectra of compound 26 in Table 2 was detected with the molecular ion at m/z 163.0395 [M + H]+ and gave characteristic fragment ions of 145.0627[M + H–H2O]+. Similarly, MS2 spectra of compound 25 in Table 3 was detected with the molecular ion at m/z 163.0396[M + H]+ and gave characteristic fragment ions of [M + H–H2O]+ at m/z 145.5012. Thus, compound 25 in Table 3 was identified as the absorbed prototype of 7-hydroxycoumarin in rat serum.
6-Gingerdione, 6-gingerol and 6-shogaol sourced from Zingiber officinale Rosc (Ganjiang) were identified and were reported as having obvious antioxidant, anti-inflammatory, gastrointestinal protective and antitumor effects [43, 44]. The MS data of the (+) ESI–MS spectra are shown in Table 3. For example, MS2 spectra of compound 50 in Table 2 was detected with the molecular ion at m/z 293.1736[M + H]+ and gave characteristic fragment ions of 275.1650[M + H–H2O]+, 257.1517[M + H–2H2O]+. Similarly, MS2 spectra of compound 15 in Table 3 was detected with the molecular ion at m/z 293.1734[M + H]+ and gave characteristic fragment ions of [M + H–H2O]+ at m/z 257.1586. Thus, compound 15 in Table 3 was identified as the absorbed prototype of 6-gingerdione in rat serum.
One compound was sourced from Codonopsis pilosula (Franch.) Nannf. (Dangshen) and was identified as l-pyroglutamic acid. MS2 spectra of compound 1 in Table 2 was detected with the molecular ion at m/z 130.0505[M + H]+ and gave characteristic fragment ions of 112.0123[M + H–H2O]+, 84.0449[M + H–HCOOH]+. Similarly, MS2 spectra of compound 1 in Table 3 was detected with the molecular ion at m/z 130.0498[M + H]+ and gave characteristic fragment ions of [M + H–H2O]+ at m/z 112.9741. Thus, compound 1 in Table 3 was identified as the absorbed prototype of l-pyroglutamic acid in rat serum.

Identification of the bioactive metabolites in rat serum

Based on a comparison of the information for ions, 8 peaks were detected only in dosed serum and were assigned to metabolites. Detailed information about the elemental compositions, retention times, and the characteristic fragment ions of metabolites are shown in Table 3. Alkaloid-, phenylpropanoids- and gingerols-related metabolites are the main metabolic constituents of FZPLP absorbed in vivo, and the main metabolic pathways in vivo were glucuronide conjugation and glucuronide. Identification of the corresponding fragment ions was obvious. For example, compound 4 (24.357 min) in Table 3 produced [M + H] + at m/z 433 and MS2 yielded a major ion at m/z 257 (− 176, Da with the loss of C6H8O6) in the positive ion mode, combined with the retention time of the reference standard 1 in Table 1 and compound 29 in Table 2. Therefore, the peak was identified tentatively as a glucuronide conjugation metabolite of liquiritigenin. Similarly, compound 13 (the tR 33.165 min) in Table 3 has the similar retention time compared with the reference standard 6 in Table 1 and compound 48 in Table 2. And it produced [M + H] + at m/z 433 and MS2 yielded a major ion at m/z 257 (− 176, Da with the loss of C6H8O6) in the positive ion mode. Therefore, the peak was identified tentatively as a glucuronide conjugation metabolite of isoliquiritigenin. The possible structures of metabolites were elucidated as described above. All of the structures of metabolites were identified, and the MS data of the (+) ESI–MS spectra are shown in Table 3. This article reports these metabolites of FZLZP for the first time. The bioactivities are the subject of ongoing research.

Alkaloids difference between Group A and Group B

As the result shows in Fig. 5a, 10 kinds of alkaloids were detected in Group A. Most of them were trace amounts in vivo, which indicated the alkaloids’ poor absorption in the prescription. Conversely, unlike Group A, the amount of the alkaloids in vivo increased obviously in Group B (Fig. 5b). The difference indicated that the absorption amount of alkaloids in the prescription can be decreased compared to the absorption amount of alkaloids in the herb powder.

Discussion

To obtain LC chromatograms of lower pressure, greater baseline stability, better resolution and higher ionization efficiency, methanol and acetonitrile and series of concentrations of aqueous formic acid solution were prepared for analysis. The best result was achieved when the mobile phase consisted of 0.1% formic acid aqueous solution and methanol. Both positive and negative modes were investigated, and the results showed that the positive ion mode was more sensitive and could provide more information for both extract samples and serum samples analyses.
FZLZP is a formula composed under the guidance of traditional Chinese medicine theory. According to TCM theory, Aconitum carmichaeli Debx. is the “monarch drug” and the main herb in FZLZP recipe to warm middle jiao and eliminate cold. This was confirmed in this research with 10 constituents among 23 prototype components sourced from Aconitum carmichaeli Debx., which maintains the maximum bioactive compounds. Glycyrrhiza uralensis Fisch. is frequently prescribed in combination with other herbs to decrease toxicity and to increase efficacy. In this recipe, it is the “envoy drug” and is considered to be the paramount assistant herb, which can detoxify the toxicity of aconitum. In this study, we found that Glycyrrhiza uralensis Fisch. was the second most-absorbed herb. The results that some compounds absorbed well in vivo derived from Aconitum carmichaeli Debx. and Glycyrrhiza uralensis Fisch. are consistent with our previous studies that they were dissolved very well in vitro [16].
Alkaloids in Fuzi herb are the toxicity as well as the efficacy compounds. The prescriptions which contains Fuzi herb should be highly concerned. In our study, the results on the differences in alkaloids between Group A and Group B show that the amount of absorption of bioactive constituents in Fuzi can be significantly reduced when this herb is used as part of a prescription rather than used alone. We think there are two reasons. Firstly, according to the TCM theory, the toxicity of Fuzi can be reduced in combination with Gancao [25]. This should be further confirmed by researching the relationship and differences in the chemistry constituents between Fuzi-Gancao herb pairs in FZLZP. Secondly, the pill form is the embryonic form of sustained-release preparations. As a TCM classic says: only pill among all dosage forms can reduce the toxicity of toxic drugs. The toxic herb was usually made into a pill form to reduce the toxicity in TCM [17]. And it can be further confirmed by researching differences in the chemistry constituents between FZLZP and the Fuzi pill that made from Aconitum carmichaeli Debx. powder.

Conclusions

This study describes a simple, sensitive and selective HPLC-QTOF-MS method for structural characterization of chemical constituents in FZLZP and bioactive components in rat serum following oral administration of FZLZP. As a result, in vitro, a total of 67 compounds were successfully identified, and 23 prototype compounds that were absorbed in vivo were identified for the first time. In addition, 3 metabolites of the bioactive compounds were tentatively identified. In this prescription, the majority of compounds absorbed in vivo derived from Fuzi and Gancao. The results provide helpful chemical information for FZLZP for further pharmacological and active mechanism research. In addition, it helped to classify the material basis responsible for the therapeutic effects of FZLZP. Furthermore, the HPLC-QTOF-MS was a potentially powerful strategy for simultaneously achieving screening and analysis of multiple bioactive compounds in FZLZP.

Authors’ contributions

ZZ and MJ carried out the screening experiments, ZZ wrote the manuscript and analyzed the data, MJ, XW, SY, JS graphed the picture, GZ revised the manuscript, CF and LG conceived of the study, contributed to the design and interpretation of the research. All authors read and approved the final manuscript.

Acknowledgements

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

The dataset supporting the conclusions of this article is included within the article.
Not applicable.
Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (No. 81803742), Key Project of Natural Science Fund of Sichuan Province (No. 18ZA0187), Pre-research National Natural Science Foundation of Chengdu University of Traditional Chinese Medicine (No. ZRYY1718).

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Yang XY. Investigation and analysis of clinical use of fuzi lizhong pills. Chin J Clinic Ratio Drug Use. 2018;11(10):98–100. Yang XY. Investigation and analysis of clinical use of fuzi lizhong pills. Chin J Clinic Ratio Drug Use. 2018;11(10):98–100.
2.
Zurück zum Zitat Yang LL, Xu YQ. Fuzi Lizhong Pill in the treatment of yang deficiency of spleen and kidney infantile anorexia. J GM Chin Med. 2018;33(12):1699–701. Yang LL, Xu YQ. Fuzi Lizhong Pill in the treatment of yang deficiency of spleen and kidney infantile anorexia. J GM Chin Med. 2018;33(12):1699–701.
3.
Zurück zum Zitat Xu L, Zhao D. A randomized controlled study of fuzi lizhong pill combined with moxibustion in the treatment of severe enteral nutritional diarrhea. Cardi Disea J Integr Tradi Chin West Med. 2018;6(19):152–3. Xu L, Zhao D. A randomized controlled study of fuzi lizhong pill combined with moxibustion in the treatment of severe enteral nutritional diarrhea. Cardi Disea J Integr Tradi Chin West Med. 2018;6(19):152–3.
4.
Zurück zum Zitat Hu XL, Zhou H, Ning QY, Zhao ZX, Xu XS. Observation on the curative effect of the treatment of chronic terminal ileus of spleen and kidney Yang deficiency by the combined treatment of Fuzi-Lizhong Pill and heavy moxibustion. Guid J Tradit Chin Med Pharm. 2012;18(1):33–4. Hu XL, Zhou H, Ning QY, Zhao ZX, Xu XS. Observation on the curative effect of the treatment of chronic terminal ileus of spleen and kidney Yang deficiency by the combined treatment of Fuzi-Lizhong Pill and heavy moxibustion. Guid J Tradit Chin Med Pharm. 2012;18(1):33–4.
5.
Zurück zum Zitat Liu HL, Huang LX. Treating IBS with the Fuzi Lizhong wan plus bifico, aroxetine. Clin J Chin Med. 2015;7(13):71–3. Liu HL, Huang LX. Treating IBS with the Fuzi Lizhong wan plus bifico, aroxetine. Clin J Chin Med. 2015;7(13):71–3.
6.
Zurück zum Zitat Hu LQ, Zhang LX, Zhang LF. Observation and analysis of the curative effect of fuzi lizhong pill combined with wuweita oral solution in the treatment of chronic ulcerative colitis. Med Innov Chin J. 2012;9(3):123–4. Hu LQ, Zhang LX, Zhang LF. Observation and analysis of the curative effect of fuzi lizhong pill combined with wuweita oral solution in the treatment of chronic ulcerative colitis. Med Innov Chin J. 2012;9(3):123–4.
7.
Zurück zum Zitat Ye SH. The treatment of 62 cases of diarrhea irritable bowel syndrome by sishen pill combine with fuzi lizhong pill. SD J Tradi Chin Med. 2010;29(5):310–1. Ye SH. The treatment of 62 cases of diarrhea irritable bowel syndrome by sishen pill combine with fuzi lizhong pill. SD J Tradi Chin Med. 2010;29(5):310–1.
8.
Zurück zum Zitat Zhao X, Yang SJ, Zhang WT, Zu CZ, Tang BH, Zhang BC, et al. Fuzi-Lizhong pill compensates hypothyroid-hypothermia via ghrelin release. J Ethnopharmacol. 2013;149:707–12.CrossRef Zhao X, Yang SJ, Zhang WT, Zu CZ, Tang BH, Zhang BC, et al. Fuzi-Lizhong pill compensates hypothyroid-hypothermia via ghrelin release. J Ethnopharmacol. 2013;149:707–12.CrossRef
9.
Zurück zum Zitat Dong LY, Cheng BF, Luo Y, Zhang N, Duan HQ, Jiang M, et al. Identification of nuclear factor-κB inhibitors and β2 adrenergic receptor agonists in Chinese medicinal preparation Fuzilizhong Pills using UPLC with quadrupole time-of-flight MS. Phytochem Anal. 2014;25:113–21.CrossRef Dong LY, Cheng BF, Luo Y, Zhang N, Duan HQ, Jiang M, et al. Identification of nuclear factor-κB inhibitors and β2 adrenergic receptor agonists in Chinese medicinal preparation Fuzilizhong Pills using UPLC with quadrupole time-of-flight MS. Phytochem Anal. 2014;25:113–21.CrossRef
10.
Zurück zum Zitat Zhao X, Wang Y, Yang SJ, Zhang WT, Zu ZC, Zhang TT, et al. Underlying mechanism of Aconitum Lizhong acting on experimental hypothermia with indigestion in rats: role of ghrelin. Evid Based Complement Alternat Med. 2012;2012:542461.PubMedPubMedCentral Zhao X, Wang Y, Yang SJ, Zhang WT, Zu ZC, Zhang TT, et al. Underlying mechanism of Aconitum Lizhong acting on experimental hypothermia with indigestion in rats: role of ghrelin. Evid Based Complement Alternat Med. 2012;2012:542461.PubMedPubMedCentral
11.
Zurück zum Zitat Zhang WT, Tang HQ, Wang Y, Lu AN, Zhao X, Song M, et al. Aconitum Lizhong Pill strenthening adaptive thermogenesis in rats with spleen yang deficient syndrome. Chin Pharmac. 2010;13(7):918–21. Zhang WT, Tang HQ, Wang Y, Lu AN, Zhao X, Song M, et al. Aconitum Lizhong Pill strenthening adaptive thermogenesis in rats with spleen yang deficient syndrome. Chin Pharmac. 2010;13(7):918–21.
12.
Zurück zum Zitat Zhang WT, Tang HQ, Lu AN, Wang XY, Luo J, Yang MJ, et al. Fuzi Lizhong Pills regulating myosin ATPase activity of skeletal muscle in rats with spleen yang deficient syndrome. Chin J Tradi Chin Med Pharm. 2011;26(3):490–4. Zhang WT, Tang HQ, Lu AN, Wang XY, Luo J, Yang MJ, et al. Fuzi Lizhong Pills regulating myosin ATPase activity of skeletal muscle in rats with spleen yang deficient syndrome. Chin J Tradi Chin Med Pharm. 2011;26(3):490–4.
13.
Zurück zum Zitat Tang HQ, Zhang WT, Lu A, Wang Y, Wang XY, Yang Y, et al. Effects of Aconitum Lizhong Pills on energy charge in skeletal muscle of rats with spleen yang deficiency syndrome. Chin Pharmac. 2010;13(12):1691–4. Tang HQ, Zhang WT, Lu A, Wang Y, Wang XY, Yang Y, et al. Effects of Aconitum Lizhong Pills on energy charge in skeletal muscle of rats with spleen yang deficiency syndrome. Chin Pharmac. 2010;13(12):1691–4.
14.
Zurück zum Zitat Zhang WT, Tang HQ, Lu AN, Zhao X, Li GZ, Jiang YF, et al. Treatment with Aconitum Lizhong Pill down-regulates liver energy charge in rats with spleen yang deficiency syndrome. World Chin J Digest. 2010;18(35):3782–6.CrossRef Zhang WT, Tang HQ, Lu AN, Zhao X, Li GZ, Jiang YF, et al. Treatment with Aconitum Lizhong Pill down-regulates liver energy charge in rats with spleen yang deficiency syndrome. World Chin J Digest. 2010;18(35):3782–6.CrossRef
15.
Zurück zum Zitat Lu L, Tang HQ, Li XH, Zhu XY. Effects of Fuzi Lizhong Decoction on content of ANP, Expression of pGC mRNA and gastrointestinal dynamics in rats with spleen yang deficiency syndrome. Chin J Exp Tradit Med Form. 2013;19(24):264–6. Lu L, Tang HQ, Li XH, Zhu XY. Effects of Fuzi Lizhong Decoction on content of ANP, Expression of pGC mRNA and gastrointestinal dynamics in rats with spleen yang deficiency syndrome. Chin J Exp Tradit Med Form. 2013;19(24):264–6.
16.
Zurück zum Zitat Jiang MY, Zhang Z, Shi JF, Zhang JM, Fu CM, Lin X, et al. Dissolution behavior of Fuzi Lizhong pill based on simultaneous determination of two components in Glycyrrhizae Radix et Rhizoma. Chin J Chin Mater Med. 2018;43(5):952–4. Jiang MY, Zhang Z, Shi JF, Zhang JM, Fu CM, Lin X, et al. Dissolution behavior of Fuzi Lizhong pill based on simultaneous determination of two components in Glycyrrhizae Radix et Rhizoma. Chin J Chin Mater Med. 2018;43(5):952–4.
17.
Zurück zum Zitat Zhang Z, Gao TH, Fu CM, Zhang JM, Shi JF, He Y, et al. Analysis on dosage form theory and current application situation of traditional Chinese medicine pill. Chin J Chin Mater Med. 2017;42(12):2048. Zhang Z, Gao TH, Fu CM, Zhang JM, Shi JF, He Y, et al. Analysis on dosage form theory and current application situation of traditional Chinese medicine pill. Chin J Chin Mater Med. 2017;42(12):2048.
18.
Zurück zum Zitat Gao TH, Zhang Z, Zhang JM, Fu S, Fu CM. Research progress and comprehensive analysis of “toxicity and effect” of Fuzi Lizhong Pill. Pharm Clin Chin Mater Med. 2017;8(2):74–6. Gao TH, Zhang Z, Zhang JM, Fu S, Fu CM. Research progress and comprehensive analysis of “toxicity and effect” of Fuzi Lizhong Pill. Pharm Clin Chin Mater Med. 2017;8(2):74–6.
19.
Zurück zum Zitat Sun H, Wu F, Zhang A, Wei W, Han Y, Wang X. Profiling and identification of the absorbed constituents and metabolites of schisandra lignans by ultra-performance liquid chromatography coupled to mass spectrometry. Biomed Chromatogr. 2013;27:1511–9.CrossRef Sun H, Wu F, Zhang A, Wei W, Han Y, Wang X. Profiling and identification of the absorbed constituents and metabolites of schisandra lignans by ultra-performance liquid chromatography coupled to mass spectrometry. Biomed Chromatogr. 2013;27:1511–9.CrossRef
20.
Zurück zum Zitat Wang P, Yin QW, Zhang AH, Sun H, Wu XH, Wang XJ. Preliminary identification of the absorbed bioactive components and metabolites in rat plasma after oral administration of Shaoyao-Gancao decoction by ultra-performance liquid chromatography with electrospray ionization tandem mass spectrometry. Pharmacogn Mag. 2014;10(40):497–502.CrossRef Wang P, Yin QW, Zhang AH, Sun H, Wu XH, Wang XJ. Preliminary identification of the absorbed bioactive components and metabolites in rat plasma after oral administration of Shaoyao-Gancao decoction by ultra-performance liquid chromatography with electrospray ionization tandem mass spectrometry. Pharmacogn Mag. 2014;10(40):497–502.CrossRef
21.
Zurück zum Zitat He JL, Zhao JW, Ma ZC, Wang YG, Liang QD, Tan HL, et al. Serum pharmacochemistry analysis using UPLC-Q-TOF/MS after oral administration to rats of Shenfu decoction. Evid Based Complement Alternat Med. 2015;2015:973930.PubMedPubMedCentral He JL, Zhao JW, Ma ZC, Wang YG, Liang QD, Tan HL, et al. Serum pharmacochemistry analysis using UPLC-Q-TOF/MS after oral administration to rats of Shenfu decoction. Evid Based Complement Alternat Med. 2015;2015:973930.PubMedPubMedCentral
22.
Zurück zum Zitat Sun H, Wu F, Zhang A, Wei W, Han Y, Wang X. Pharmacokinetic study of schisandrin, schisandrol B, schisantherin A, deoxyschisandrin, and schisandrin B in rat plasma after oral administration of Shengmaisan formula by UPLC-MS. J Sep Sci. 2013;36:485–91.CrossRef Sun H, Wu F, Zhang A, Wei W, Han Y, Wang X. Pharmacokinetic study of schisandrin, schisandrol B, schisantherin A, deoxyschisandrin, and schisandrin B in rat plasma after oral administration of Shengmaisan formula by UPLC-MS. J Sep Sci. 2013;36:485–91.CrossRef
23.
Zurück zum Zitat Zeng HJ, Yang R, Guo C, Wang QW, Qu LB, Li JJ. Pharmacokinetic study of six flavones in rat plasma and tissues after oral administration of “JiangYaBiFeng” using SPE-HPLC-DAD. J Pharm Biomed Anal. 2011;56(4):815–9.CrossRef Zeng HJ, Yang R, Guo C, Wang QW, Qu LB, Li JJ. Pharmacokinetic study of six flavones in rat plasma and tissues after oral administration of “JiangYaBiFeng” using SPE-HPLC-DAD. J Pharm Biomed Anal. 2011;56(4):815–9.CrossRef
24.
Zurück zum Zitat Zhang A, Sun H, Wang X, Jiao G, Yuan Y, Sun W. Simultaneous in vivo RP-HPLC-DAD quantification of multiple-component and drug-drug interaction by pharmacokinetics, using 6,7-dimethylesculetin, geniposide and rhein as examples. Biomed Chromatogr. 2012;26:844–50.CrossRef Zhang A, Sun H, Wang X, Jiao G, Yuan Y, Sun W. Simultaneous in vivo RP-HPLC-DAD quantification of multiple-component and drug-drug interaction by pharmacokinetics, using 6,7-dimethylesculetin, geniposide and rhein as examples. Biomed Chromatogr. 2012;26:844–50.CrossRef
25.
Zurück zum Zitat Zhang JM, Li L, Gao F, Li Y, He Y, Fu CM. Chemical ingredient analysis of sediments from both single Radix Aconiti Lateralis decoction and Radix Aconiti Lateralis—Radix Glycyrrhizae decoction by HPLC-MS. Acta Pharm Sin. 2012;47(11):1527–33.CrossRef Zhang JM, Li L, Gao F, Li Y, He Y, Fu CM. Chemical ingredient analysis of sediments from both single Radix Aconiti Lateralis decoction and Radix Aconiti Lateralis—Radix Glycyrrhizae decoction by HPLC-MS. Acta Pharm Sin. 2012;47(11):1527–33.CrossRef
26.
Zurück zum Zitat Xue CS, Zhang AH, Sun H, Han Y, Zou D, Wang YY, et al. An improved ultra-performance liquid chromatography–electrospray ionization/quadrupole-time-of-flight high-definition mass spectrometry method for determining ingredients of herbal Fructus corni in blood samples. Pharmacogn Mag. 2014;10(40):422–9.CrossRef Xue CS, Zhang AH, Sun H, Han Y, Zou D, Wang YY, et al. An improved ultra-performance liquid chromatography–electrospray ionization/quadrupole-time-of-flight high-definition mass spectrometry method for determining ingredients of herbal Fructus corni in blood samples. Pharmacogn Mag. 2014;10(40):422–9.CrossRef
27.
Zurück zum Zitat Dong W, Wang P, Meng XC, Sun H, Zhang AH, Wang WM, et al. Ultra-performance liquid chromatography-high-definition mass spectrometry analysis of constituents in the root of Radix Stemonae and those absorbed in blood after oral administration of the extract of the crude drug. Phytochem Anal. 2012;23(6):657–67.CrossRef Dong W, Wang P, Meng XC, Sun H, Zhang AH, Wang WM, et al. Ultra-performance liquid chromatography-high-definition mass spectrometry analysis of constituents in the root of Radix Stemonae and those absorbed in blood after oral administration of the extract of the crude drug. Phytochem Anal. 2012;23(6):657–67.CrossRef
28.
Zurück zum Zitat Wang SP, Zhang JZ, Zhang ZD, Gao WY, Yan YN, Li X, et al. Identification of chemical constituents in the extract and rat serum from Ziziphus Jujube Mill. by HPLC-PDA-ESI-MSn. Iran J Pharm Res. 2014;13(3):1055–63.PubMedPubMedCentral Wang SP, Zhang JZ, Zhang ZD, Gao WY, Yan YN, Li X, et al. Identification of chemical constituents in the extract and rat serum from Ziziphus Jujube Mill. by HPLC-PDA-ESI-MSn. Iran J Pharm Res. 2014;13(3):1055–63.PubMedPubMedCentral
29.
Zurück zum Zitat Li XN, Sun H, Zhang AH, Liu ZD, Zou D, Song YH, et al. High-throughput LC-MS method for the rapid characterization of multiple chemical constituents and metabolites of Da-Bu-Yin-Wan. J Sep Sci. 2017;40:4102–12.CrossRef Li XN, Sun H, Zhang AH, Liu ZD, Zou D, Song YH, et al. High-throughput LC-MS method for the rapid characterization of multiple chemical constituents and metabolites of Da-Bu-Yin-Wan. J Sep Sci. 2017;40:4102–12.CrossRef
30.
Zurück zum Zitat Tan GG. Chemome and Metabonomics studied of traditional Chinese medicine Sini decoction. Second military medical university. Shanghai, China. 2012. Tan GG. Chemome and Metabonomics studied of traditional Chinese medicine Sini decoction. Second military medical university. Shanghai, China. 2012.
31.
Zurück zum Zitat Huang XF, Ou YH, Li JM, Lu YJ, Li W, Gong QF. Identification of characteristic constituents in Atractylodis Macrocephalae Rhizoma from different regions by UPLC-Q-TOF-MS/MS. Chin J Exp Tradit Med Form. 2017;23(23):27–33. Huang XF, Ou YH, Li JM, Lu YJ, Li W, Gong QF. Identification of characteristic constituents in Atractylodis Macrocephalae Rhizoma from different regions by UPLC-Q-TOF-MS/MS. Chin J Exp Tradit Med Form. 2017;23(23):27–33.
32.
Zurück zum Zitat Hikino H, Konno C, Takata H, Yamada Y, Yamada C, Ohizumi Y, et al. Anti-inflammatory principles of Aconitum roots. J Pharm Dyn. 1980;3(10):514–25.CrossRef Hikino H, Konno C, Takata H, Yamada Y, Yamada C, Ohizumi Y, et al. Anti-inflammatory principles of Aconitum roots. J Pharm Dyn. 1980;3(10):514–25.CrossRef
33.
Zurück zum Zitat Zhang JM, Fu CM, He YX, Lu JR, Gao F, Wang JS. Comparison on chemical components in sediments of Aconiti Lateralis Radix Preparata-Glycyrrhizae Radix et Rhizoma before and after their compatibility. Chin Trad Herb Drug. 2013;44(2):165–9. Zhang JM, Fu CM, He YX, Lu JR, Gao F, Wang JS. Comparison on chemical components in sediments of Aconiti Lateralis Radix Preparata-Glycyrrhizae Radix et Rhizoma before and after their compatibility. Chin Trad Herb Drug. 2013;44(2):165–9.
34.
Zurück zum Zitat Makino T, Kato K, Mizukami H. Processed Aconite root prevents cold-stress-induced hypothermia and immune-suppression in mice. Biol Pharm Bull. 2009;32(10):1741–8.CrossRef Makino T, Kato K, Mizukami H. Processed Aconite root prevents cold-stress-induced hypothermia and immune-suppression in mice. Biol Pharm Bull. 2009;32(10):1741–8.CrossRef
35.
Zurück zum Zitat Chen S, Li WH, Chen XF, Liu M, Zhu ZY, Chai YF. Research progress of components and quality control analysis of alkaloids in Fuzi. Chin J Pharm Anal. 2014;34(10):1709–17. Chen S, Li WH, Chen XF, Liu M, Zhu ZY, Chai YF. Research progress of components and quality control analysis of alkaloids in Fuzi. Chin J Pharm Anal. 2014;34(10):1709–17.
36.
Zurück zum Zitat Takami M, Tomohiro Y, Eiji T, Hiroaki S, Ikuo S, Tadato T. Pharmaceutical evaluation of liquorice before and after roasting in mice. J Pharm Pharmacol. 2010;56(5):589–95. Takami M, Tomohiro Y, Eiji T, Hiroaki S, Ikuo S, Tadato T. Pharmaceutical evaluation of liquorice before and after roasting in mice. J Pharm Pharmacol. 2010;56(5):589–95.
37.
Zurück zum Zitat Kim KR, Jeong CK, Park KK, Choi JH, Park JHY, Lim SS, et al. Anti-inflammatory effects of Licorice and roasted Licorice extracts on TPA-induced acute inflammation and collagen-induced arthritis in mice. J Biomed Biotechnol. 2010;2010:709378.PubMedPubMedCentral Kim KR, Jeong CK, Park KK, Choi JH, Park JHY, Lim SS, et al. Anti-inflammatory effects of Licorice and roasted Licorice extracts on TPA-induced acute inflammation and collagen-induced arthritis in mice. J Biomed Biotechnol. 2010;2010:709378.PubMedPubMedCentral
38.
Zurück zum Zitat Ryoko S, Maiko O, Hiroko T, Kenzo O. Inhibitory effect of glycyrrhizin on the phosphorylation and DNA-binding abilities of high mobility group proteins 1 and 2. Biol Pharm Bull. 2001;24(8):906–11.CrossRef Ryoko S, Maiko O, Hiroko T, Kenzo O. Inhibitory effect of glycyrrhizin on the phosphorylation and DNA-binding abilities of high mobility group proteins 1 and 2. Biol Pharm Bull. 2001;24(8):906–11.CrossRef
39.
Zurück zum Zitat Li CQ, He LC, Dong HY, Jin JQ. Screening for the anti-inflammatory activity of fractions and compounds from Atractylodes macrocephala koidz. J Ethnopharmacol. 2007;114(2):212–7.CrossRef Li CQ, He LC, Dong HY, Jin JQ. Screening for the anti-inflammatory activity of fractions and compounds from Atractylodes macrocephala koidz. J Ethnopharmacol. 2007;114(2):212–7.CrossRef
40.
Zurück zum Zitat Wang CH, Duan HJ, He LC. Inhibitory effect of atractylenolide I on angiogenesis in chronic inflammation in vivi and in vitro. Eur J Pharmacol. 2009;612(1):143–52.CrossRef Wang CH, Duan HJ, He LC. Inhibitory effect of atractylenolide I on angiogenesis in chronic inflammation in vivi and in vitro. Eur J Pharmacol. 2009;612(1):143–52.CrossRef
41.
Zurück zum Zitat Li CQ, He LC, Jin JQ. Atractylenolide I and Atractylenolide III inhibit lipopolysaccharide-induced TNF-α and no production in macrophages. Phytother Res. 2010;21(4):347–53.CrossRef Li CQ, He LC, Jin JQ. Atractylenolide I and Atractylenolide III inhibit lipopolysaccharide-induced TNF-α and no production in macrophages. Phytother Res. 2010;21(4):347–53.CrossRef
42.
Zurück zum Zitat Yang E, Zhong YM, Feng YF. Advance on the chemical constituents and pharmacological effects of Atractylodes macrocephala Koidz. J Guangdong Pharm Univ. 2012;28(2):219–21. Yang E, Zhong YM, Feng YF. Advance on the chemical constituents and pharmacological effects of Atractylodes macrocephala Koidz. J Guangdong Pharm Univ. 2012;28(2):219–21.
43.
Zurück zum Zitat Yuki M, Hiroe K, Masashi H, Nobuji N. Antioxidant properties of gingerol related compounds from ginger. BioFactors. 2004;21:293–6.CrossRef Yuki M, Hiroe K, Masashi H, Nobuji N. Antioxidant properties of gingerol related compounds from ginger. BioFactors. 2004;21:293–6.CrossRef
44.
Zurück zum Zitat Chrubasika S, Pittler MH, Roufogalis BD. Zingiberis rhizome: a comprehensive review on the ginger effect and efficacy profiles. Phytomedicine. 2005;12(9):684–701.CrossRef Chrubasika S, Pittler MH, Roufogalis BD. Zingiberis rhizome: a comprehensive review on the ginger effect and efficacy profiles. Phytomedicine. 2005;12(9):684–701.CrossRef
Metadaten
Titel
Rapid discovery of chemical constituents and absorbed components in rat serum after oral administration of Fuzi-Lizhong pill based on high-throughput HPLC-Q-TOF/MS analysis
verfasst von
Zhen Zhang
Maoyuan Jiang
Xinyi Wei
Jinfeng Shi
Zhao Geng
Shasha Yang
Chaomei Fu
Li Guo
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Chinese Medicine / Ausgabe 1/2019
Elektronische ISSN: 1749-8546
DOI
https://doi.org/10.1186/s13020-019-0227-z

Weitere Artikel der Ausgabe 1/2019

Chinese Medicine 1/2019 Zur Ausgabe