Next Article in Journal
Association of Indoleamine 2,3-Dioxygenase (IDO) Activity with Outcome after Cardiac Surgery in Adult Patients
Previous Article in Journal
Explainable AI to Facilitate Understanding of Neural Network-Based Metabolite Profiling Using NMR Spectroscopy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Metabolite Profiling Analysis of the Tongmai Sini Decoction in Rats after Oral Administration through UHPLC-Q-Exactive-MS/MS

1
The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
2
Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China
*
Author to whom correspondence should be addressed.
Metabolites 2024, 14(6), 333; https://doi.org/10.3390/metabo14060333
Submission received: 23 April 2024 / Revised: 28 May 2024 / Accepted: 3 June 2024 / Published: 14 June 2024
(This article belongs to the Special Issue LC-MS/MS Analysis for Plant Secondary Metabolites)

Abstract

:
Tongmai Sini decoction (TSD), the classical prescriptions of traditional Chinese medicine, consisting of three commonly used herbal medicines, has been widely applied for the treatment of myocardial infarction and heart failure. However, the absorbed components and their metabolism in vivo of TSD still remain unknown. In this study, a reliable and effective method using ultra-performance liquid chromatography coupled with hybrid quadrupole-Orbitrap mass spectrometry (UHPLC-Q-Exactive-MS/MS) was employed to identify prototype components and metabolites in vivo (rat plasma and urine). Combined with mass defect filtering (MDF), dynamic background subtraction (DBS), and neutral loss filtering (NLF) data-mining tools, a total of thirty-two major compounds were selected and investigated for their metabolism in vivo. As a result, a total of 82 prototype compounds were identified or tentatively characterized in vivo, including 41 alkaloids, 35 phenolic compounds, 6 saponins. Meanwhile, A total of 65 metabolites (40 alkaloids and 25 phenolic compounds) were tentatively identified. The metabolic reactions were mainly hydrogenation, demethylation, hydroxylation, hydration, methylation, deoxylation, and sulfation. These findings will be beneficial for an in-depth understanding of the pharmacological mechanism and pharmacodynamic substance basis of TSD.

1. Introduction

The classical prescriptions of traditional Chinese medicine (TCM) have originated from the fixed combination of certain kinds of herbal medicines recorded in the ancient classics, which have been still widely used in East Asia and exhibited precise clinical efficacy, with obvious characteristics and advantages [1]. For a long time, some classic prescriptions have been developed into modern Chinese medicinal preparations through the optimization of preparation technology and drug development research [2,3]. Most classical prescriptions have existed for at least hundreds of years, and with time, classic prescriptions may have changed to some extent, while their core characteristics (e.g., composition of herbs, proportion of herbs, etc.) have not changed significantly [4,5]. The reasons for the inheritance of classical prescriptions to the present day can be attributed to the high safety of the prescriptions and their proven efficacy due to a large number of clinical applications.
Tongmai Sini decoction (TSD) is a classic formula from the Chinese medical masterpiece “The Treatise on Typhoid Fever”, written 1800 years ago. It consists of three herbal concoctions of Radix Aconiti Lateralis Preparata (RALP), Rhizoma Zingiberis (RZ), and Radix Glycyrrhizae Preparata (RGP) and is commonly used in modern times for myocardial infarction and heart failure, atherosclerosis, shock, diarrhea, etc. [6,7]. TSD has the effects of raising blood pressure, strengthening the heart, anti-hypoxia, anti-shock, anti-thrombosis, anti-myocardial ischemia, anti-slowing arrhythmia, and so on [8,9]. The main chemical constituents of TSD include alkaloids (from RALP), phenolic acids and saponins (from RGP), and volatile oils (from RZ). At present, chemical composition [10], pharmacological, pharmacokinetic [11,12,13], and metabolomics [8,9,14] studies have been preliminarily conducted on TSD, especially on its cardiovascular activities. Most of the studies on TDS concentrated on the pharmacokinetics of diterpene alkaloids after oral administration of TDS, and some of the studies focused on the changes in the in vivo metabolome or lipidome profile against myocardial ischemia, heart failure, hypothyroidism. There is a lack of systematic and in-depth in vivo chemical and metabolite studies of TDS.
The components of different botanicals enter the body and produce metabolites, which exert therapeutic effects through multiple pathways [15]. To fully understand the therapeutic components, it is necessary to first analyze the blood-entering components and their metabolites, as well as to study the metabolites distributed in plasma, urine, feces, and tissues, which is conducive to analysis of the potential components and pathways of action in the body [16,17].
High-resolution mass spectrometry (HRMS), in combination with chromatography technology, has provided useful structural information about chemical components, offering strong support for the characterization of in vivo and in vitro metabolic components of botanicals [18,19,20]. In recent years, in order to improve the sensitivity and selectivity of obtaining MS/MS or MSn data for trace components in vivo, some acquisition and identification strategies have been developed, combined, and applied, for instance, the extracted ion chromatogram (EIC), mass defect filter (MDF), dynamic background subtraction (DBS) and neutral loss filter (NLF) [21,22,23].
In this paper, the established UHPLC-Q-Exactive-MS/MS methods have great advantages for the qualitative analysis of bioactive samples in rats after oral doses of TSD, and a variety of post-data processing techniques, including EIC, MDF, DBS, and NLF was applied for quickly screen and systematically identify the metabolites. These metabolic studies can provide the chemical foundation and an in-depth understanding of metabolic transformation for further research on effective substances and the action mechanism of TSD.

2. Materials and Methods

2.1. Chemicals and Plant Materials

Aconitine, mesaconitine, hypaconitine, liquiritin, liquiritigenin, ononin, and formononetin, with purities greater than 98%, were purchased from Shanghai Yuanye Biological Technology Co., Ltd. (Shanghai, China). HPLC-grade acetonitrile, methanol, and formic acid were purchased from Sigma Aldrich (St. Louis, MO, USA). Ultrapure Water (18.2 MΏ) was produced by a Milli-Q water system (Millipore, Bedford, MA, USA). The herbal pieces of RALP, RZ, and RGP were purchased from Kangmei Pharmaceutical Co. Ltd. Of Guangdong, China, and identified by Prof. Zhi-hai Huang, The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China.

2.2. Plant Extract Preparation

According to the documentary records of TSD, the TSD pieces that included RALP (30 g), RZ (20 g), and RGP (30 g) were soaked with 8 times the amount of water for 30 min and decocted to boil (100 °C) for 2 h. The filtrate was collected, and the residue was decocted in 8 times the amount of water for 1.5 h again. The hot filtrate was combined and concentrated to 80 mL (1 g herbal pieces/1 mL aqueous solution). The obtained TSD extract was stored at −20 °C before use.

2.3. Animal and Drug Administration

Male Sprague–Dawley rats (220–260 g) were obtained from Guangdong Provincial Medical Laboratory Animal Center (Guangdong, China). All animal experiments were performed at the SPF animal laboratory [experimental animals license number SYXK (Guangdong, China) 2008–0094]. The Institutional Animal Ethics Committee of Guangdong Provincial Hospital of Chinese Medicine approved all experimental protocols (No. 2023131).
Six SD rats were randomly divided into two groups (Urine and plasma groups) and adapted to the metabolic cage for a week before the experiment. Blank urine and plasma samples were collected under abrosia state ahead of gastric gavage. The rats were fasted for 14 h with water ad libitum before oral administration of TSD extract and underwent 4 h of water deprivation after that. TSD extract was orally administered to rats of urine and plasma groups twice at an interval of 1 h, and the dosage was 2 mL per 100 g bodyweight per time.

2.4. Sample Collection and Pretreatment

Urine samples from 0 to 24 h after the second dosing were collected and stored at −80 °C prior to analysis. Plasma samples were obtained at 1, 2, 4, 8, and 12 h after the second administration in heparinized 1.5 mL polythene tubes under diethyl ether anesthesia, respectively. All plasma samples were centrifuged at 4000 rpm for 10 min, and the plasma supernatants were then merged in equal volume and frozen at −80 °C prior to analysis.
The collected urine and plasma samples (200 μL) were added with 4× the volume of acetonitrile-methanol (3:1) to precipitate protein, respectively. All separate supernatants were dried under N2 flow, and the residues were resuspended in 200 μL acetonitrile and centrifuged at 15,000× g for 8 min. Finally, a 5 μL sample was injected into the UHPLC-Q-Exactive-Orbitrap MS system for further analysis.

2.5. Instrumentation and Conditions

LC analyses were conducted on a Thermo UltiMate 3000 UHPLC system (Thermo Fisher Scientific, San Jose, CA, USA) equipped with a quaternary pump, a cooling autosampler, and a thermostatically controlled column oven. An ACQUITY UPLC HSS T3 Column (2.1 × 100 mm, 1.8 μm) was used. The mobile solvents were composed of acetonitrile (A) and water with 0.02% formic acid (B), and the gradient elution profile was employed as follows: 5% A, 0 min; 16% A, 12 min; 55% A, 23 min; 90% A, 35 min; 95% A, 40 min; returning to initial conditions in 4 min at a flow rate of 200 μL/min at room temperature. The injection volume was 5 μL. The temperatures of the sample tray and the column oven were set at 4 and 35 °C, respectively.
A Q-Exactive hybrid quadrupole-orbitrap mass spectrometer was connected to an LC system via an electrospray ionization source as an interface. Data acquisition and processing were calculated using Compound Discoverer 3.2 software. The optimized parameters for MS analysis were as follows: the mass spectrometer parameters were positive (PI) and negative (NI) ion mode; the resolution of the Orbitrap mass analyzer was set as 30,000; ion spray voltage was −3.8 kV; the capillary temperature was 325 °C; the sheath gas flow rate was 40 psi; the auxiliary gas flow rate was 8 psi; and the mass range was m/z 150–1500. The properties of data-dependent MS2 scanning (DDS) parameters and events were as follows: resolution, 17,500; HCD, 35 eV; repeat count, 2; exclusion list, 50; repeat duration, 5 s; and exclusion duration, 30 s. The mass error for molecular ions of all compounds identified was within ±5 ppm.

3. Results and Discussion

3.1. Systematic Analytical Strategy for Online Metabolite Analysis

Based on our previous research on the cleavage patterns of components in RALP and RGP and a review of the literature [24,25,26,27,28], the metabolite profiling of TSD was systematically investigated by UHPLC-Q-Exactive-MS/MS methods. The workflow of the analytic procedure was carried out and shown in Figure 1. Figures S1 and S2 (Supplementary Materials) displayed the detailed workflow for the identification of prototype components and metabolites, respectively.
The strategy consisted of the following steps: (1) First, the chemical database (Table S1, Supplementary Materials) was constructed, including mass weights, elemental compositions, and structure information of chemical compositions originating from RALP, RGP, and RZ based on our previous research and the related literature [24,25,26,27,28,29]. (2) Then, an online full-scan and MS/MS data acquisition was processed in both negative and positive modes based on the DBS and DDS techniques for potential metabolite detection. (3) Next, the data files were imported into the Compound Discoverer 3.2 software, and the data-mining tools of EIC, NLF, and MDF were applied to screen the possible metabolites of TSD. Table S2 (Supplementary Materials) showed the detailed parameters of data processing. The main compounds with mass spectral peak areas greater than 108 in the decoction (shown in Table 1) were used as parent compound templates for MDF data screening (±50 mDa) (4) Next, based on the chemical database, acquired accurate mass data, retention time, and characteristic fragment ions, the identification of prototype components was elucidated (shown in Table 2). In addition, the Clog p values calculated by ChemDraw 14.0 were used to distinguish isomers at different retention times. (5) Finally, the mass information of potential metabolites, as well as their possible biotransformation pathways and composition change given by Compound Discoverer 3.2, were compared by the data of prototype components and the related literature to verify the metabolites and their metabolic pathways (shown in Table 3).

3.2. Identification of Prototype Components

An in-house database has been established for each compound involved in RALP, RGP, and RZ based on our previous experimental data and the related literature for the investigation of their chemical constituents. The database consisted of the compound name, molecular formula, accurate molecular mass, chemical structure, MS2 mass spectra, and related product ion information. The total ion chromatograms (BPIs) of TSD and the urine and plasma samples after oral administration by UHPLC-Q-Exactive-MS/MS in positive and negative ion modes are presented in Figure 2. It is found that the majority of alkaloids responded well in the positive mode, and the majority of phenolic compounds and saponins responded well in the negative mode. A total of 82 prototype compounds were identified or tentatively characterized, including 41 alkaloids, 35 phenolic compounds, and 6 saponins (shown in Table 2) by comparing the EICs among TSD, drugged, and blank samples and by comparison with reference standards, internal database, and the literature. Figure S3 (Supplementary Materials) displayed MS/MS spectra of major prototype compounds in the urine samples.

3.2.1. Identification of Alkaloid Components

Metabolites for alkaloids obtained in this study could be classified into three subtypes, namely, diester-diterpenoid alkaloids (DDAs), monoester-diterpenoid alkaloids (MDAs), and amine-diterpenoid alkaloids (ADAs) [30]. We conducted an in-depth study of the chemical constituents of alkaloids of Aconitum carmichaeli in previous research [24,29], in which we carried out detailed mass fragmentation analysis of DDAs, MDAs, and ADAs, and a total of 42 DDAs and 120 diterpenoid alkaloids were identified, respectively.
In the MS2 spectra of DDAs, the most abundant ion yielded from the loss of a molecule of AcOH at the C8 site, which could be a diagnostic neutral loss for the differentiation of DDAs from MDAs and ADAs [29]. Thus, Compounds 29, 33, 35–38, and 40–41 were extracted by NLF for 60 Da in MS spectra for the urine sample, showing their molecular weight between 600 and 650 Da. Among them, Compounds 33, 36, and 37 were unambiguously identified as mesaconitine (MA), aconitine (AC), and hypaconitine (HA), respectively, by comparing their tR values and mass spectra data with those of reference compounds. Apart from the ion of [M+H-60(AcOH)]+ (m/z 572.2844, 586.3002, 556.2903), the ions of [M+H-60-32(MeOH)]+ (m/z 554.2727, 524.2634, 540.2551) and [M+H-60-32(MeOH)-28(carbonyl group)]+ (m/z 526.2797, 496.2750, 522.2487) of the three compounds, respectively, suggested the active elimination of MeOH occurred at C16 site and a neutral molecule of CO, which could also be regarded as characteristic fragments for identification of the DDAs. Compounds 29, 35, 38, and 40–41 were tentatively identified as 10-OH-mesaconitine, dehydrohypaconitine, secoyunaconitine, 3-deoxyaconitine, and chasmaconitine by comparing their acquired accurate mass data, characteristic fragment ions with those of compounds in our previous research [29].
In the MS spectra for the urine sample, by extraction of NLF for both 32 Da and 18 Da with limitation of molecular weight ranging from 500 to 620 Da, ten peaks were found. Neutral losses of 32, 18, and 122 Da, corresponding to the elimination of acetic acid, methanol, and benzoic acid, or combinations of these, could be considered diagnostic fragment ions for MDAs [31]. However, fragment peaks formed by the loss of the typical substituent group as BzOH (122 Da) were hardly detected for MDAs in this study. Thus, Compounds 22–28 and 30–32 were identified as MDAs accordingly by comparing the accurate mass data and diagnostic fragment ions with those of the compounds in our previous research [24].
A total of 21 prototype compounds were identified as ADAs, most of which possessed molecular weight between 390 and 500 Da and were eluted within the initial 16 min. The substitutions of C1 and C3 sites of ADAs were relatively active sites and could be easily cleaved, yielding major peaks [M+H-H2O]+ or [M+H-CH3OH]+ in MS2 spectra as the diagnostic ion accordingly. Fragmentation pathways of differently substituted ADAs included different diagnostic ions. Compounds 1, 4, 6, 7, 8, 13, 14, as ADAs with C1-OH substitution, firstly fragmented into [M+H-H2O]+ as diagnostic fragment ions and followed by losses of typical substituent groups (CH3OH and H2O) in their MS2 spectra. By comparing their accurate mass data with our chemical database and the literature [24,32], they were identified as karakolidine, senbusine A, senbusine B, karakoline, isotalatizidine, fuziline, and neoline, respectively. For Compounds 18 (talatizamine), the most prominent fragmentation ions were designated as 390.2696 ([M+H-CH3OH]+), suggesting its C1 site with -OCH3 substitutions. It also yielded 372.2517 ([M+H-CH3OH-H2O]+), 358.2379 ([M+H-CH3OH-CH3OH]+), and 340.2238 ([M+H-CH3OH-CH3OH-H2O]+), and its characteristic fragmentation patterns are shown in Figure 3.

3.2.2. Identification of Phenolic Compounds

In addition to alkaloids from RALP, the main prototype compounds identified in vivo included flavonoids, isoflavonoids, coumarins, and saponins from RGP, and volatile oils from RZ, as shown in Table 2. The MS data of these compounds were compared with those of reference standards, internal databases, and the literature, while isomers could be initially identified by comparing their ClogP.
Flavonoids are important active components of RGP, among which four components, namely liquiritigenin, isoliquiritigenin, iquiritin, and isoliquiritin, have the highest content and are regarded as the indicator components of RGP, which were identified by comparing mass data with those of the reference standards. Compound 47, as reference compound liquiritin, formed the [M-H]-based peak at m/z 417.11890 (C21H21O9), for which furtherly formed fragmentation ion m/z 255.0662 [M-H-glu] of the aglycone element in the MS/MS spectrum, accompanied by three characteristic fragments at m/z 135.0074 (C7H3O3), 119.0488 (C8H7O), and 91.0173 (C6H3O), which can be used for the identification of the same type of licorice flavonoids.
Compound 50 formed the [M+H]+ molecular ion peak at m/z 431.13280 and further removed one molecule of glucose residues to form the aglycone at m/z 269.08121, which was identified as ononin, the main isoflavone of RGP. Its aglycone formed the same ion at m/z 269.08170 at the retention time of 26.58 min and was fragmented into the fragments of m/z 253.0497, 237.0554, and 213.0911, which is identified as formononetin, and the two prototypes are the most important isoflavonoid components in RGP.
The elemental compositions of other types of licorice flavonoid constituents determined by LC-MS were compared with the data of existing database compounds. Compounds 44, 53, 54, 67, and 68 were preliminarily identified as 5-hydroxyliquiritin, licochalcone B, dihydroxyflavone, licoflavone A, and isolicoflanonol. Similarly, other types of phenolic compounds, such as coumarins, were identified or preliminarily identified, including Compounds 60, 66, 64, 70, 73, and 74, which were identified as glycycoum-arim, licocoumarione, licopyranocoumarin, glycyrin isoglycyrol, and glycyrol, correspondingly. A few other phenolic components observed in vivo of TSD were derived from RZ, while compounds 56, 63, and 75 were tentatively identified as 6-gingerol, 6-shogaol, and 10-shogaol, respectively, with fragment ions m/z 177.09 and 137.06 as their characteristic fragment ions in PI mode, which is consistent with the literature [33].

3.2.3. Identification of Saponins

From the LC-MS/MS profiles, six saponin components were found as absorbed prototype components, all of which were derived from RGP. The saponins (Compounds 77, 78, 79, 81, and 82) were within the retention time of 14–21 min and had both mass spectral response in NI and in PI mode.
As a general rule for triterpenoid saponins in MS/MS spectra, the fragmentation reactions undergone by activated saponin ions almost occur within the glycan part of the saponin ions, and the sugar chains can be eliminated successively from end to inner and finally to obtain an aglycone ion [34]. Through glycosidic cleavages or cross-ring cleavages, the parent ion obtained a series of ions retaining the charge at the reducing terminus were termed Y and Z (glycosidic cleavages) and X (cross-ring cleavages), whereas those ions retaining the charge at the non-reducing terminus are termed B, C (glycoside cleavages), and A (cross-ring cleavages) [35].
The MS cleavage pathways of saponins from RGP, however, were incompletely abided by this rule. Take glycyrrhizic acid as an example; in MS spectra of PI mode, the ions of [M-H] were obtained, accompanied by the fragment ions of m/z 647.37744 [M+H-β-D-glucuronopyronosyl (glcA)]+ and m/z 453.33554 [aglycottne (agl)+H-H2O]+, which were similarly for the other detected saponins and has not been reported up to present. More interestingly, in the MS/MS spectra of the detected saponins, the ions of [agl+H-H2O]+ rather than [agl+H]+ were observed as the base peaks, namely, m/z 453.34 (C30H45O3+), 469.33 (C30H45O4+), and 511.34 (C30H45O4+), corresponding to the aglycone of enoxolone, hydroxyenoxolone, and acetoxyenoxolone, respectively.
The produced ions obtained in NI mode were quite different from those in PI mode. The fragment ions of glycosidic cleavages or cross-ring cleavages, as well as the aglycone, were hardly detected in NI mode. The ions of m/z 351.05 (C12H15O12), 193.03 (C6H9O7), 175.02 (C6H7O6), and 113.02 (C5H5O3) were observed, corresponding to the successive loss of two glucuronopyranosyls. Thus, the identification information for aglycone s and sugar chains of licorice saponins can be obtained from PI and NI ion modes, respectively.

3.3. Identification of Metabolites

Prototypes and metabolites exist simultaneously in plasma and urine samples. Thirty-two major prototypes, including 11 alkaloids from RALP, as well as 21 phenolic and saponin compounds from RGP and RZ, were selected as MDF templates for metabolite screening. The 32 compounds contained a wide range of chemical structure types with relatively high content in TDS. A total of 40 alkaloids and 25 phenolic compounds were identified or tentatively characterized by comparing the mass data with those of prototype compounds and metabolic pathways reported by the literature [36,37,38,39,40].
After prototypes are absorbed into the body, some of them are excreted as prototypes, and some of them can be converted into other metabolites. DDAs were ester hydrolyzed to MDAs in rats; for example, MA, HA, and AC could be ester hydrolyzed to 14-Benzoylmesaconine (BM), 14-Benzoylhypaconine (BH), and 14-Benzoylaconitine (BA) during the process of metabolism in rat, while BM, BH, and BA themselves could be metabolized to mesaconine, hypaconine, and aconine [36]. Therefore, certain prototypes are themselves metabolites and metabolized from other prototypes in rats.

3.3.1. Identification of Alkaloid Metabolites

For diterpenoid alkaloids, most metabolites from hydroxylation, deoxylation, demethylation, deethylation, dehydrogenation, ester hydrolysis, and demethylation with deoxylation have been found in vivo. Metabolites of alkaloids were identified or tentatively identified based on their metabolic pathways, as reported in the literature [37].
The metabolites for major alkaloids were found in the urine and plasma samples, as displayed in Table 3. Most metabolites observed were mainly metabolized from karakolidine, songorine, karakoline, talatizamine, hypaconitine, mesaconitine, neoline, and fuziline. These results manifested that alkaloids mainly underwent oxidation, dehydrogenation, demethylation, N-deethylation, hydrolysis, demethylation with deoxidation, and dehydrogenation with demethylation, etc.
After oral administration of TSD, eight related metabolites of talatizamine (18) were identified in urine samples. Metabolite M18 and M19 showed [M+H]+ ion at m/z 408.27386 and 408.27393 (giving formula C23H37NO5), 14 Da (CH2) less than the parent compound. In the MS2 spectra, characteristic ions at m/z 376.25 ([M+H-CH3OH]+), 358.24 ([M+H-CH3OH-H2O]+), and 326.21 ([M+H-CH3OH-H2O-CH3OH]+), suggesting its C1 site with -OCH3 substitutions. Those characteristic ions were different from the characteristic ions of the prototype component, isotalatizidine (Compound 8), although they shared the same elemental composition (C23H37NO5). Isotalatizidine, with -OH substitutions at the C1 site, first yielded 390.2631 ([M+H-H2O]+) by loss of H2O at the C1 site. The fragmentation pathways of demethyl talatizamine and isotalatizidine can be compared in Figure 3. The methyl group of the C16 site or C18 site could easily be metabolized instead of that of the C1 site for M18 and M19. The Clog p values of 18-O-demethyl talatizamine and 16-O-demethyl talatizamine were −0.78 and −0.74, calculated by ChemDraw 14.0. Hence, M18 and M19 were tentatively determined as 18-O-demethyl talatizamine and 16-O-demethyl talatizamine.
M4 was confirmed as hydroxylated talatizamine for the [M+H]+ ion at m/z 438.28433 (formula C24H39NO6), 16 Da (O) more than talatizamine, and the fragment ions at m/z 406.2588, 388.2476, 374.230 and 356.2226 were all 16 Da less than those of talatizamine. Therefore, M4 was deduced as 10-Hydroxy Talatizamine, as for the C10 site in diterpenoid alkaloids prone to be hydroxylated by the literature [38].
Apart from these three metabolites, other metabolites (M13, M26, M36, M39, and M40) of talatizamine were produced through the reaction of dehydrogenation, demethylation, N-deethylation, and deoxidation. The proposed metabolic pathways of talatizamine are shown in Figure 4. The other metabolites of alkaloids were deduced accordingly by their acquired accurate mass data, retention time, and characteristic fragment ions, as well as the Clog p values, and biotransformation pathways information and composition change calculated by ChemDraw 14.0 and Compound Discoverer 3.2.

3.3.2. Identification of Phenolic Compound Metabolites

Metabolites of phenolic compounds, mainly from hydroxylation, oxylation, methylation, dehydrogenation, hydration, methylation with oxylation, dehydrogenation with oxylation, and sulfation, have been observed in vivo. They were identified or tentatively identified by comparing their accurate mass data with prototypes and their metabolic pathways reported by the literature [39,40]. The metabolites for major phenolic compounds found in the urine and plasma samples were exhibited in Table 4.
Metabolites of phenolic compounds observed in vivo were mainly derived from the metabolism of liquiritigenin, isoliquiritigenin, and 6-gingerol, which were the most important aglycones from RGP and RZ in TSD. According to the MS data and the metabolic pathways reported in the literature, eleven related metabolites were identified in urine and plasma samples after the absorption of liquiritigenin and isoliquiritigenin. M60 and M61 showed [M-H] ion at m/z 285.0765 (C16H13O5+), 30 Da (CH2+O) heavier than parent compounds. In the MS/MS spectra, characteristic ions at m/z 270.05 [M-H-CH2] indicated the methyl substitution, and m/z 135.01 or 119.05 were used for the characterization of liquiritigenin or isoliquiritigenin derivatives. According to the polarity of liquiritigenin and isoliquiritigenin, M60 and M61 were confirmed as methyl-hydroxy liquiritigenin and methyl-hydroxy isoliquiritigenin. In vivo, liquiritigenin and isoliquiritigenin could be metabolized to a series of metabolites (M41, M42, M50, M53, M54, M56, M57, M60, M61, M63, and M64) by reaction of hydrogenation, dehydrogenation, hydroxylation, oxylation, hydration, methylation, and sulfation. In vivo, seven metabolites (M43, M47, M48, M52, M55, M58, and M59) of 6-gingerol were produced through the reaction of hydrogenation, methylation, hydration, hydroxylation, and dehydrogenation, with characteristic ions at m/z 163.08 or 137.06.

3.4. Difference between Urine and Plasma Samples

Xenobiotics usually vary at trace levels and are interfered with endogenous components. Comparative analysis of metabolites between plasma and urine samples was carried out by the same LC-MS/MS method. Most prototype components and metabolites possessed suitable signal responses in urine samples, mainly as metabolites from phase I metabolism referring to dehydrogenation, demethylation, hydroxylation, deoxylation, and deethylation. A few phase II metabolites were detected in the urine, including sulfate conjugates of liquiritigenin, isoliquiritigenin, and formononetin.
Metabolites of TSD detected in the plasma samples are fewer than those in the urine samples. As for plasma samples, 10 prototype components (eight phenolic compounds and two alkaloids) were detected and tentatively identified, most of which were flavonoid aglycones. Fifteen metabolites derived from neoline, talatizamine, karakoline songorine, and fuziline, as well as sixteen metabolites derived from liquiritigenin, isoliquiritigenin, formononetin, gancaonin M, and 6-gingerol, respectively, were found in plasma samples, which indicated there were fewer metabolites identified in plasma samples. These results are reasonable due to their relatively lower concentration and higher matrix interference in plasma than in urine samples.
In the present study, ADAs and their metabolites from RALP were mainly detected in rats after oral administration of TSD. DDAs are the most toxic but chemically unstable alkaloids in RALP, and the alkaloidal composition changed during concocting and decocting, with DDAs changing to MDAs, and both transformed further to ADAs while the toxicity gradually diminished. ADAs, such as fuziline and neoline, showed activity against pentobarbital sodium-induced cardiomyocyte damage by obviously recovering beating rhythm and increasing the cell viability [41]. Mesaconine and hypaconine showed strong cardiac actions on the isolated perfused bullfrog heart. Moreover, mesaconine has protective effects, including improved inotropic effect and left ventricular diastolic function, on myocardial ischemia-reperfusion injury in rats [42].
Metabolites of licorice flavonoids and 6-gingerol were also mainly detected. Liquiritigenin offers cytoprotective effects against various cardiac injuries, and it could protect against myocardial ischemic injury by antioxidation, antiapoptosis, counteraction mitochondrial dysfunction, and damping intracellular Ca2+ [43]. 6-Gingerol was identified as a novel angiotensin II type 1 receptor antagonist for cardiovascular disease by high-throughput screening, which partially clarified the mechanism of ginger regulating blood pressure and strengthening the heart [44]. 6-gingerol administration protected I/R-induced cardiomyocyte apoptosis via the JNK/NF-κB pathway in the regulation of HMGB2 [45].
The results of the in vivo metabolite study of TSD in this study suggested that in the following pharmacokinetic, pharmacological, and efficacy studies, attention should be paid primarily to the ADAs alkaloids, licorice flavonoids, gingerol-6, and their metabolites

4. Conclusions

A total of 82 compounds, including 41 alkaloids, 35 phenolic compounds, and 6 saponins, were identified or tentatively characterized in TSD by UHPLC-Q-Exactive-MS/MS. Among them, 32 representative compounds with relatively high mass spectral peak areas and different core structures were selected as parent compound templates for further investigation of their metabolic profiles in rats. In total, 65 metabolites were screened out and tentatively characterized in rats’ urine and plasma based on their MS characteristic fragmentation patterns and information. The main metabolic reactions involved hydrogenation, demethylation, hydroxylation, hydration, methylation, deoxylation, and sulfation. This is a systematic study of in vivo metabolism of TSD, and it will be beneficial for further understanding of the pharmacological and pharmacokinetic study of TSD.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/metabo14060333/s1, Figure S1: Workflow for the identification of prototype components, Figure S2: Workflow for the identification of metabolites, Figure S3: MS/MS spectra of major prototype compounds in the urine samples. Table S1: Compound information of in-house database, Table S2: Parameters of data processing by Compound Discoverer software.

Author Contributions

Conceptualization, G.D.; Methodology, W.X.; Formal analysis, X.Z., Y.Z. and M.P.; Data curation, X.Z. and Y.Z.; Writing—original draft, X.Z. and M.P.; Writing—review & editing, X.Z., W.X. and G.D.; Supervision, G.D.; Project administration, G.D.; Funding acquisition, G.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research is supported by special funds for international cooperation from the Guangdong Provincial Hospital of Chinese Medicine (YN2024RD01) and the Scientific Research Project of Guangdong Provincial Bureau of Traditional Chinese Medicine (20231144).

Institutional Review Board Statement

All animal experiments were performed at the SPF animal laboratory [experimental animals license number SYXK (Guangdong, China) 2008–0094]. The Institutional Animal Ethics Committee of Guangdong Provincial Hospital of Chinese Medicine approved all experimental protocols (approval code: No. 2023131, approval date: 21 December 2023).

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author. The data are not publicly available due to confidentiality.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. He, L.Y.; Kang, Q.M.; Zhang, Y.; Chen, M.; Wang, Z.F.; Wu, Y.H.; Gao, H.T.; Zhong, Z.F.; Tan, W. Glycyrrhizae Radix et Rhizoma: The popular occurrence of herbal medicine applied in classical prescriptions. Phytother. Res. 2023, 37, 3135–3160. [Google Scholar] [CrossRef] [PubMed]
  2. Lu, C.C.; Zhang, S.Y.; Lei, S.S.; Wang, D.N.; Peng, B.; Shi, R.P.; Chong, C.M.; Zhong, Z.F.; Wang, Y.T. A comprehensive review of the classical prescription Yiguan Jian: Phytochemistry, quality control, clinical applications, pharmacology, and safety profile. J. Ethnopharmacol. 2024, 319, 117230. [Google Scholar] [CrossRef] [PubMed]
  3. Zhang, Q.; Han, X.-X.; Mao, C.-Q.; Xie, H.; Chen, L.-H.; Mao, J.; Lu, T.-L.; Yan, G.-J. Opportunities and challenges in development of compound preparations of traditional Chinese medicine: Problems and countermeasures in research of ancient classical prescriptions. China J. Chin. Mater. Medica 2019, 44, 4300–4308. [Google Scholar]
  4. Chen, Z.K.; Wang, X.N.; Li, Y.Y.; Wang, Y.H.; Tang, K.L.; Wu, D.F.; Zhao, W.Y.; Ma, Y.M.; Liu, P.; Cao, Z.W. Comparative network pharmacology analysis of classical TCM prescriptions for chronic liver disease. Front. Pharmacol. 2019, 10, 1353. [Google Scholar] [CrossRef] [PubMed]
  5. Chu, X.Y.; Wei, X.H.; Wu, X.F.; Chen, J.; Xia, H.; Xia, G.Y.; Lin, S.; Shang, H.C. Pharmacological research progress of five classical prescriptions in treatment of chronic heart failure. China J. Chin. Mater. Medica 2023, 48, 6324–6333. [Google Scholar]
  6. Zhou, Q.; Meng, P.; Zhang, Y.; Chen, P.; Wang, H.B.; Tan, G.G. The compatibility effects of sini decoction against doxorubicin-induced heart failure in rats revealed by mass spectrometry-based serum metabolite profiling and computational analysis. J. Ethnopharmacol. 2020, 252, 112618. [Google Scholar] [CrossRef] [PubMed]
  7. Chen, Q.; Xiao, S.; Li, Z.H.; Ai, N.; Fan, X.H. Chemical and Metabolic Profiling of Si-Ni Decoction Analogous Formulae by High performance Liquid Chromatography-Mass Spectrometry. Sci. Rep. 2015, 5, 11638. [Google Scholar] [CrossRef]
  8. Chen, S.; Wu, S.; Li, W.H.; Chen, X.F.; Dong, X.; Tan, G.G.; Zhang, H.; Hong, Z.Y.; Zhu, Z.Y.; Chai, Y.F. Investigation of the therapeutic effectiveness of active components in Sini decoction by a comprehensive GC/LC-MS based metabolomics and network pharmacology approaches. Mol. Biosyst. 2014, 10, 3310–3321. [Google Scholar] [CrossRef] [PubMed]
  9. Tan, G.G.; Wang, X.; Liu, K.; Dong, X.; Liao, W.T.; Wu, H. Correlation of drug-induced and drug-related ultra-high performance liquid chromatography-mass spectrometry serum metabolomic profiles yields discovery of effective constituents of Sini decoction against myocardial ischemia in rats. Food Funct. 2018, 9, 5528–5535. [Google Scholar] [CrossRef]
  10. Hu, Q.; Chen, M.; Yan, M.M.; Wang, P.L.; Lei, H.M.; Xue, H.Y.; Ma, Q. Comprehensive analysis of Sini decoction and investigation of acid-base self-assembled complexes using cold spray ionization mass spectrometry. Microchem. J. 2020, 173, 107008. [Google Scholar] [CrossRef]
  11. Zhang, H.; Liu, M.; Zhang, W.; Chen, J.; Zhu, Z.Y.; Cao, H.; Chai, Y.F. Comparative pharmacokinetics of three monoester-diterpenoid alkaloids after oral administration of Acontium carmichaeli extract and its compatibility with other herbal medicines in Sini Decoction to rats. Biomed. Chromatogr. 2015, 29, 1076–1083. [Google Scholar] [CrossRef]
  12. Zhou, Q.; Meng, P.; Wang, H.B.; Dong, X.; Tan, G.G. Pharmacokinetics of monoester-diterpenoid alkaloids in myocardial infarction and normal rats after oral administration of Sini decoction by microdialysis combined with liquid chromatography-tandem mass spectrometry. Biomed. Chromatogr. 2019, 33, e4406. [Google Scholar] [CrossRef] [PubMed]
  13. Sun, S.; Chen, Q.S.; Ge, J.Y.; Liu, X.; Wang, X.X.; Zhan, Q.; Zhang, H.; Zhang, G.Q. Pharmacokinetic interaction of aconitine, liquiritin and 6-gingerol in a traditional Chinese herbal formula. Sini Decoction 2018, 48, 45–52. [Google Scholar] [CrossRef]
  14. Zhou, J.; Ma, X.Q.; Shi, M.; Chen, C.W.; Sun, Y.; Li, J.J.; Xiong, Y.X.; Chen, J.J.; Li, F.Z. Serum metabolomics analysis reveals that obvious cardioprotective effects of low dose Sini decoction against isoproterenol-induced myocardial injury in rats. Phytomedicine 2017, 31, 18–31. [Google Scholar] [CrossRef] [PubMed]
  15. Bai, S.S.; Luo, D.W.; Zhong, G.Y.; Yang, S.L.; Ouyang, H.; Rao, X.Y.; Feng, Y.L. Exploration of plant metabolomics variation and absorption characteristics of water-extracted Rheum tanguticum and ethanol-extracted Rheum tanguticum by UHPLC-Q-TOF-MS/MS. Phytochem. Anal. 2024, 35, 288–307. [Google Scholar] [CrossRef]
  16. Ye, L.H.; He, X.X.; Yan, M.Z.; Chang, Q. Identification of in vivo components in rats after oral administration of lotus leaf flavonoids using ultra fast liquid chromatography with tandem mass spectrometry. Anal. Methods 2014, 6, 6088–6094. [Google Scholar] [CrossRef]
  17. Tao, J.H.; Zhao, M.; Jiang, S.; Zhang, W.; Xu, B.H.; Duan, J.A. UPLC-Q-TOF/MS-based metabolic profiling comparison of four major bioactive components in normal and CKD rat plasma, urine and feces following oral administration of Cornus officinalis Sieb and Rehmannia glutinosa Libosch herb couple extract. J. Pharm. Biomed. Anal. 2018, 161, 254–261. [Google Scholar] [CrossRef]
  18. Deng, F.; Li, X.M.; Gong, Q.Q.; Zheng, Z.X.; Zeng, L.; Zhang, M.J.; Duan, T.Y.; Liu, X.; Zhang, M.Z.; Guo, D.L. Identification of in vivo metabolites of Citri Sarcodactylis Fructus by UHPLC-Q/Orbitrap HRMS. Phytochem. Anal. 2023, 34, 938–949. [Google Scholar] [CrossRef] [PubMed]
  19. Zhang, X.J.; Liu, S.; Xing, J.P.; Pi, Z.F.; Liu, Z.Q.; Song, F.R. Systematic study on metabolism and activity evaluation of Radix Scutellaria extract in rat plasma using UHPLC with quadrupole time-of-flight mass spectrometry and microdialysis intensity-fading mass spectrometry. J. Sep. Sci. 2018, 41, 1704–1710. [Google Scholar] [CrossRef]
  20. Huang, J.; Zhang, J.P.; Bai, J.Q.; Wei, M.J.; Zhang, J.; Huang, Z.H.; Qu, G.H.; Xu, W.; Qiu, X.H. Chemical profiles and metabolite study of raw and processed Polygoni Multiflori Radix in rats by UPLC-LTQ-Orbitrap MSn spectrometry. Chin. J. Nat. Med. 2018, 16, 375–400. [Google Scholar]
  21. Su, C.Y.; Wang, J.H.; Chang, T.Y.; Shih, C.L. Mass defect filter technique combined with stable isotope tracing for drug metabolite identification using high-resolution mass spectrometry. Anal. Chim. Acta 2022, 1208, 339814. [Google Scholar] [CrossRef] [PubMed]
  22. Zhou, X.H.; Chen, X.; Yin, X.M.; Wang, M.Y.; Zhao, J.J.; Ren, Y. A strategy integrating parent ions list-modified mass defect filtering-diagnostic product ions for rapid screening and systematic characterization of flavonoids in Scutellaria barbata using hybrid quadrupole-orbitrap high-resolution mass spectrometry. J. Chromatogr. A 2022, 1674, 463149. [Google Scholar] [CrossRef]
  23. Wang, B.L.; Lu, Y.M.; Hu, X.L.; Feng, J.H.; Shen, W.; Wang, R.; Wang, H. Systematic Strategy for Metabolites of Amentoflavone In Vivo and In Vitro Based on UHPLC-Q-TOF-MS/MS Analysis. J. Agric. Food Chem. 2020, 68, 14808–14823. [Google Scholar] [CrossRef] [PubMed]
  24. Xu, W.; Zhang, J.; Zhu, D.Y.; Huang, J.; Huang, Z.H.; Bai, J.Q.; Qiu, X.H. Rapid separation and characterization of diterpenoid alkaloids in processed roots of Aconitum carmichaeli using ultra high performance liquid chromatography coupled with hybrid linear ion trap-Orbitrap tandem mass spectrometry. J. Sep. Sci. 2014, 37, 2864–2873. [Google Scholar] [CrossRef] [PubMed]
  25. Cai, X.F.; XU, Y.; Liu, H.P.; Shang, Q.; Qiu, J.Q.; Xu, W. Chemical analysis of classical prescription Qianghuo Shengshi standard decoction by UHPLC-Q Exactive Orbitrap MS. China J. Chin. Mater. Medica 2022, 47, 343–357. [Google Scholar]
  26. Lu, F.Y.; Cai, H.; Li, S.M.; Xie, W.; Sun, R.J. The Chemical Signatures of Water Extract of Zingiber officinale Rosc. Molecules 2022, 27, 7818. [Google Scholar] [CrossRef] [PubMed]
  27. Meng, X.Y.; Li, H.L.; Song, F.R.; Liu, C.M.; Liu, Z.Q.; Liu, S.Y. Studies on Triterpenoids and Flavones in Glycyrrhiza uralensis Fisch by HPLC-ESI-MSn and FT-ICR-MSn. Chin. J. Chem. 2009, 27, 299–305. [Google Scholar] [CrossRef]
  28. Avula, B.; Bae, J.Y.; Chittiboyina, A.G.; Wang, Y.H.; Wang, M.; Zhao, J.P.; Ali, Z.; Brinckmann, J.A.; Li, J.; Wu, C. Chemometric analysis and chemical characterization for the botanical identification of Glycyrrhiza species (G. glabra, G. ura-lensis, G. inflata, G. echinata and G. lepidota) using liquid chromatography-quadrupole time of flight mass spectrometry (LC-QToF) Bharathi. J. Food Compos. Anal. 2022, 112, 104679. [Google Scholar]
  29. Zhang, J.; Huang, Z.H.; Qiu, X.H.; Yang, Y.M.; Zhu, D.Y.; Xu, W. Neutral fragment filtering for rapid identification of new diester-diterpenoid alkaloids in roots of Aconitum carmichaeli by ultra-high-pressure liquid chromatography coupled with linear ion trap-Orbitrap mass spectrometry. PLoS ONE 2012, 7, e52352. [Google Scholar] [CrossRef]
  30. He, G.N.; Wang, X.X.; Liu, W.R.; Li, Y.L.; Shao, Y.M.; Liu, W.D.; Liang, X.D.; Bao, X. Chemical constituents, pharmacological effects, toxicology, processing and compatibility of Fuzi (lateral root of Aconitum carmichaelii Debx): A review. J. Ethnopharmacol. 2023, 307, 116160. [Google Scholar] [CrossRef]
  31. Chen, X.; Tan, P.; He, R.; Liu, Y.G. Study on the fragmentation pathway of the aconitine-type alkaloids under electrospray ionization tandem mass spectrometry utilizing quantum chemistry. J. Pharm. Innov. 2013, 8, 83–89. [Google Scholar] [CrossRef]
  32. Hu, R.; Zhao, J.; Qi, L.W.; Li, P.; Jing, S.L.; Li, H.J. Structural characterization and identification of C19- and C20-diterpenoid alkaloids in roots of Aconitum carmichaeli by rapid-resolution liquid chromatography coupled with time-of-flight mass spectrometry. Rapid Commun. Mass Spectrom. 2009, 23, 1619–1635. [Google Scholar] [CrossRef] [PubMed]
  33. Lv, L.S.; Soroka, D.; Chen, X.X.; Leung, T.C.; Sang, S.M. 6-Gingerdiols as the major metabolites of 6-gingerol in cancer cells and in mice and their cytotoxic effects on human cancer cells. Agric Food Chem. 2012, 60, 11372–11377. [Google Scholar] [CrossRef] [PubMed]
  34. Pham, H.N.; Tran, C.A.; Trinh, T.D.; Thi, N.L.N.; Phan, H.N.; Le, V.N.; Le, N.H.; Phung, V. UHPLC-Q-TOF-MS/MS Dereplication to identify chemical constituents of Hedera helix leaves in Vietnam. J. Anal. Methods Chem. 2022, 2022, 1167265. [Google Scholar] [CrossRef] [PubMed]
  35. Savarino, P.; Demeyer, M.; Decroo, C.; Colson, E.; Gerbaux, P. Mass spectrometry analysis of saponins. Mass Spectrom. Rev. 2023, 42, 954–983. [Google Scholar] [CrossRef] [PubMed]
  36. Zhang, Z.; Jiang, M.Y.; Wei, X.Y.; Shi, J.F.; Geng, Z.; Yang, S.S.; Fu, C.M.; Guo, L. 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. Chin. Med. 2019, 14, 6. [Google Scholar] [CrossRef] [PubMed]
  37. Cao, Y.; Chen, X.F.; Lu, D.Y.; Dong, X.; Zhang, G.Q.; Chai, Y.F. Using cell membrane chromatography and HPLC-TOF/MS method for in vivo study of active components from roots of Aconitum carmichaeli. J. Pharm. Anal. 2011, 1, 125–134. [Google Scholar] [PubMed]
  38. Zhang, M.; Peng, C.; Li, X.B. In vivo and in vitro metabolites from the main diester and monoester diterpenoid alkaloids in a traditional Chinese herb, the Aconitum species. Evid. Based Complement. Altern. Med. 2015, 2015, 252434. [Google Scholar]
  39. Zhang, L.; Wang, C.X.; Wu, J.; Wang, T.Y.; Zhong, Q.Q.; Du, Y.; Ji, S.; Wang, L.; Guo, M.Z.; Xu, S.Q. Metabolic profiling of mice plasma, bile, urine and feces after oral administration of two licorice flavonones. J. Ethnopharmacol. 2020, 257, 112892. [Google Scholar] [CrossRef]
  40. Li, Y.Y.; Yang, L.; Chai, X.; Yang, J.J.; Wang, Y.F.; Zhu, Y. Four major urinary metabolites of liquiritigenin in rats and their anti-platelet aggregation activity. Chem. Nat. Compd. 2018, 54, 443–446. [Google Scholar] [CrossRef]
  41. Xiong, L.; Peng, C.; Xie, X.F.; Guo, L.; He, C.J.; Geng, Z.; Wan, F.; Dai, O.; Zhou, Q.M. Alkaloids Isolated from the Lateral Root of Aconitum carmichaelii. Molecules 2012, 17, 9939–9946. [Google Scholar] [CrossRef] [PubMed]
  42. Liu, X.X.; Jian, X.X.; Cai, X.F.; Chao, R.B.; Chen, Q.H.; Chen, D.L.; Wang, X.L.; Wang, F.P. Cardioactive C19-diterpenoid alkaloids from the lateral roots of Aconitum carmichaeli “Fu Zi”. Chem. Pharm. Bull. 2012, 60, 144–149. [Google Scholar] [CrossRef] [PubMed]
  43. Zhang, M.Q.; Qi, J.Y.; He, Q.Q.; Ma, D.L.; Li, J.; Chu, X.; Zuo, S.J.; Zhang, Y.X.; Li, L.; Chu, L. Liquiritigenin protects against myocardial ischemic by inhibiting oxidative stress, apoptosis, and L-type Ca2+ channels. Phytother. Res. 2022, 36, 3619–3631. [Google Scholar] [CrossRef] [PubMed]
  44. Liu, Q.; Liu, J.J.; Guo, H.L.; Sun, S.N.; Wang, S.F.; Zhang, Y.L.; Li, S.Y.; Qiao, Y.J. [6]-Gingerol: A Novel AT1 Antagonist for the Treatment of Cardiovascular Disease. Planta Medica 2013, 79, 322–326. [Google Scholar] [CrossRef]
  45. Zhang, W.Y.; Liu, X.Y.; Jiang, Y.P.; Wang, N.N.; Li, F.; Xin, H.L. 6-Gingerol attenuates ischemia-reperfusion-induced cell apoptosis in human AC16 cardiomyocytes through HMGB2-JNK1/2-NF-κB pathway. Evid. Based Complement. Altern. 2019, 2019, 8798653. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Workflow of the analytic strategy for the metabolite identification of TSD.
Figure 1. Workflow of the analytic strategy for the metabolite identification of TSD.
Metabolites 14 00333 g001
Figure 2. Total ion chromatograms (TIC) of TSD and the urine and plasma samples after oral administration by UHPLC-Q-Exactive-MS/MS ((A): Tongmai Sini decoction; (B): blank urine samples; (C): urine samples; (D): blank plasma samples; (E): plasma samples.).
Figure 2. Total ion chromatograms (TIC) of TSD and the urine and plasma samples after oral administration by UHPLC-Q-Exactive-MS/MS ((A): Tongmai Sini decoction; (B): blank urine samples; (C): urine samples; (D): blank plasma samples; (E): plasma samples.).
Metabolites 14 00333 g002
Figure 3. Probable fragmentation pathways of talatizamine, isotalatizidine, and 16-O-demethyl talatizamine.
Figure 3. Probable fragmentation pathways of talatizamine, isotalatizidine, and 16-O-demethyl talatizamine.
Metabolites 14 00333 g003
Figure 4. Proposed metabolic pathways of talatizamine in vivo.
Figure 4. Proposed metabolic pathways of talatizamine in vivo.
Metabolites 14 00333 g004
Table 1. Main prototype components as parent compound templates for MDF data screening. (mass spectral peak areas greater than 108 in the decoction).
Table 1. Main prototype components as parent compound templates for MDF data screening. (mass spectral peak areas greater than 108 in the decoction).
Alkaloids (from RALP)Phenolic and Saponin Compounds (from RGP and RZ)
KarakolidineLiquiritigeninFormononetin
FuzilineIsoliquiritigeninOnonin
NeolineLiquiritinGlycyrrhizic Acid
SongorineLicochalcone BGlycyrrhetinic Acid
14-BenzoylhypaconineLicochalcone CUralsaponin C
TalatizamineLicochalcone DLicoricesaponin G2
KarakolineLicoflavone CGlycycoum-Arim
14-BenzoylmesaconineLicoflavone AGlycyrol
MesaconitineLicoricidinGlycyrin
HypaconitineLicoleafol6-Gingerol
AconitineGancaonin M
Table 2. Prototype compounds identified or tentatively characterized in the urine and plasma samples after oral administration of TSD.
Table 2. Prototype compounds identified or tentatively characterized in the urine and plasma samples after oral administration of TSD.
ID[M+H]+(m/z)FormulatR
(min)
Error
(ppm)
ms/msIdentificationClogPArea
Urine Plasma
Alkaloids
1.394.25839C22H35NO53.51−1.58376.2474, 358.2376, 344.2229, 326.2116, 243.2516Karakolidine +++
2.394.25820C22H35NO54.02−1.42376.2476, 358.2367, 340.2268, 328.2260, 307.4473, 218.6333Chuanfumine +++
3.439.25229[M-H]−C23H37NO74.250.56392.2438, 344.2226, 295.8235, 193.8604, 146.93759-Hydroxysenbusine A +
4.424.26871C23H37NO66.69−1.55406.2584, 388.2478, 356.2207, 154.1227Senbusine A−2.70+++
5.486.26941C24H39NO97.01−0.72454.2438, 436.2322, 404.2069, 378.1887, 372.1793, 319.9836Mesaconine ++
6.424.26871C23H37NO67.74−1.55406.2581, 388.2472, 356.2210, 154.1231Senbusine B0.16++
7.378.26306C22H35NO48.20−2.18360.2524, 342.2431, 328.2268, 242.3140Karakoline ++
8.408.27371C23H37NO58.26−1.81390.2631, 372.2533, 358.2367, 340.2271Isotalatizidine ++
9.358.23691C22H31NO39.17−2.13340.2265Songorine +
10.360.25293C22H33NO39.18−1.09-Napelline ++
11.330.20569C20H27NO39.70−2.07236.8785, 170.7432, 152.4712Hetisine ++
12.470.27435C24H39NO810.51−1.05-Hypaconine ++
13.454.27933C24H39NO711.33−1.26436.2685, 418.2609, 404.2422, 154.1227Fuziline +++
14.438.28445C24H39NO611.58−1.28420.2736, 402.2617, 388.2472, 356.2214, 278.6899Neoline +++
15.420.27390C24H37NO512.31−1.32402.2632, 384.2512, 370.2359, 342.2414, 324.2322, 251.139614-Acetylkarakoline +
16.484.28937C25H41NO812.41−2.33-Deoxyaconine +
17.342.16931C20H23NO412.89−1.96297.1120, 282.0887, 237.0910, 219.0804, 191.0860N-Methyl-laurotetanine +++
18.422.28931C24H39NO513.64−1.88390.2629, 372.2517, 358.2379, 340.2238, 98.0970Talatizamine ++++++
19.420.23825C23H33NO615.48−0.86402.2268, 370.1989, 293.7002, 154.1224Giraldine F ++
20.452.29996C25H41NO615.54−1.57420.2740, 388.2465, 356.2219, 209.1644, 154.1228, 114.0916Chasmanine ++++
21.464.30038C26H41NO616.933−0.60432.2740, 414.2626, 400.2474, 372.2535, 265.1608, 235.1487, 154.122514-Acetyltalatizamine ++++++
22.606.28992C31H43NO1117.26−1.60574.2627, 556.2545, 524.2269, 506.2188, 492.1945, 261.0641, 173.0955, 105.034114-Benzoyl-10-OH-mesaconine ++
23.544.28955C30H41NO819.47−0.95512.2635, 494.2548, 480.2364, 462.2258, 390.2286, 270.0846, 105.0340Gadenine +
24.590.29490C31H43NO1019.51−1.78558.2616, 540.2575, 419.7593, 307.8019, 246.8854, 105.033914-Benzoylmesaconine ++
25.540.29486C31H41NO720.01−1.33504.2730, 462.2614, 382.2463, 340.2256, 322.2149, 304.2042Aconicarchamine B +
26.604.31060C32H45NO1020.53−1.68572.2811, 554.2750, 522.2495, 490.2176, 340.3151, 105.034114-Benzoylaconine ++
27.574.30010C31H43NO921.20−1.65542.2744, 510.2461, 304.5384, 198.1281, 105.033914-Benzoylhypaconine +++
28.618.29210[M-H]−C32H45NO1121.210.31384.9167, 351.8983, 270.7405, 190.926714-Benzoyl-10-OH-aconine ++
29.648.30023C33H45NO1221.93−1.98588.2775, 556.2513, 455.3509, 370.1645, 105.034010-OH-mesaconitine ++
30.558.30530C31H43NO822.07−1.52526.2800, 508.2674, 232.0710182.0626, 105.034114-Benzoyl-doxyhypaconine ++
31.588.31561C32H45NO922.32−1.24556.2905, 524.2639, 506.2443, 346.4250, 253.7027, 154.1226, 105.034114-Benzoyldeoxyaconine +
32.542.31061C31H43NO723.18−1.15510.2846, 492.2735, 482.2483, 460.2504, 154.123114-Benzoylneoline +
33.632.30591C33H45NO1123.18−0.63572.2844, 540.2551, 522.2487, 508.2299, 354.1694, 105.0341Mesaconitine * +++
34.662.31683C34H47NO1223.39−0.27-Aconifine ++
35.614.29553C33H43NO1024.17−0.72554.2743, 494.2534, 372.2162, 344.21622, 203.5583, 105.03412,3-didehydrohypaconitine +
36.646.32135C34H47NO1124.58−0.84586.3002, 554.2727, 526.2797, 494.2520, 368.1843, 105.0340Aconitine * ++
37.616.31079C33H45NO1024.61−0.83556.2903, 524.2634, 496.2750, 464.2434, 338.1741, 310.1812, 105.0341Hypaconitine * ++++
38.600.31592C33H45NO924.96−1.32540.2948, 508.2683, 480.2747, 476.2424, 448.2475, 354.2031, 254.4337, 105.0339Secoyunaconitine +
39.572.32117C32H45NO824.95−1.10484.2688, 456.2745, 382.2002, 322.1798, 294.1857, 158.096414-O-Anisoylneoline +
40.630.32635C34H47NO1026.07−1.36570.3046, 538.2788, 510.2882, 506.2528, 478.2571, 352.1898, 314.5361, 105.03413-Deoxyaconitine +++
41.614.33173C34H47NO927.60−0.53-Chasmaconitine ++
Phenolic compounds
42.209.04474[M-H]−C10H10O58.56−3.85165.0545, 121.0281, 103.9187, 87.9238, 59.0123Hydroxyferulic acid +++++
43.433.13394[M-H]−C18H24O129.880.08161.0442, 125.0230, 99.0436Asperulosidic acid ++
44.433.11407[M-H]−C21H22O1013.87−0.19271.0615, 151.00245-Hydroxyliquiritin ++
45.593.15137[M-H]−C27H30O1515.510.29473.1098, 383.9785, 353.0774Vitexin II +++
46.563.14055[M-H]−C26H28O1415.83−0.51473.1089, 443.0985, 383.0769, 253.0502, 146.9367Vitexin I +
47.417.11890[M-H]−C21H22O916.74−0.32255.0662, 153.0182, 135.0074, 119.0488Liquiritin * ++++
48.505.13339C24H24O1218.72−0.89257.0809, 137.0234Malonyl liquiritin +
49.505.13358C24H24O1218.99−0.07257.0810, 137.0234Malonyl liquiritin +
50.431.13280C22H22O920.26−1.97269.0809Ononin ++++
51.417.11908[M-H]−C21H22O920.39−1.01255.0662, 153.0180, 135.0072, 119.0481Neoliquiritin0.75+++
52.417.11900[M-H]−C21H22O920.74−2.47255.0662, 153.01816, 135.0074, 119.0488Isoliquiritin1.28++
53.285.07670[M-H]−C16H14O521.23−0.31270.0536, 253.0505, 177.0182, 150.0310, 108.0203Licochalcone B ++
54.255.06560C15H10O421.300.10227.0704, 199.0754, 145.0286, 137.0234Dihydroxyflavone ++++
55.255.06580[M-H]−C15H12O421.70−0.36153.0180, 135.0073, 119.0487, 91.0173Liquiritigenin * +++++++
56.295.19040C17H26O423.34−0.22177.0914, 163.0755, 137.0598, 131.0493, 99.08096-Gingerol ++++++
57.269.04530[M-H]−C15H10O524.340.38233.1537, 181.0644Genistein +++
58.255.06586[M-H]−C15H12O426.29−0.49153.0179, 135.0073, 119.0487, 91.0174Isoliquiritigenin * +++++++
59.269.08170C16H12O426.581.04253.0497, 237.0554, 213.0911, 118.0418, 107.0497Formononetin * +++++++
60.367.11790[M-H]−C21H20O627.75−1.99352.0944, 309.0400, 298.0476, 283.0247Glycycoum-arim/Licocoumarione +++
61.271.09565C16H14O428.561.19254.2579, 161.0599, 137.0598, 123.04440, 100.0763Echinatin ++
62.355.11835[M-H]−C20H20O628.60−1.07328.1265, 269.11820, 269.11820, 178.9975, 125.02308-Dimethylallyleriodictyol/6-Dimethylallyleriodictyol ++
63.277.18008C17H24O328.842.28177.0912, 145.0649, 137.05986-Shogaol +++++
64.355.15480[M-H]−C21H24O529.81−1.06323.1284, 233.1176, 207.1017, 135.0438, 125.0230, 109.0280Isopentadienyl glycyrrhizoflavone ++
65.367.11790[M-H]−C21H20O629.53−2.00309.0400, 297.0400, 284.0325, 203.0702Glycycoum-arim/Licocoumarione +++
66.321.11262[M-H]−C20H18O430.07−1.93306.0892, 174.9549Licoflavone A +
67.353.10290[M-H]−C20H18O630.17−1.33339.1187, 321.1126, 295.0613, 283.0614, 270.0535Isolicoflanonol +++++
68.353.13782C21H20O530.22−1.59299.0906, 297.0857, 267.0653, 199.0758, 147.0441, 135.0441Gancaonin M ++
69.383.11273[M-H]−C21H20O730.39−2.33338.2439, 247.1310, 227.0704, 207.1015, 155.0337, 140.0101Licopyranocoumarin +
70.383.14828C22H22O630.66−1.50327.0859, 299.0913, 191.0704Glycyrin ++++
71.355.15320C21H22O530.94−2.01289.0549, 287.0553, 191.1067, 153.0548, 69.0708Licobenzofuran/liconeolignan +++
72.337.10780[M-H]−C20H18O531.02−1.07314.0428, 282.0531Licoflavone C ++
73.365.10239[M-H]−C21H18O630.17−1.63307.0244, 295.0245, 282.0169Isoglycyrol4.84++
74.365.10236[M-H]−C21H18O631.12−1.99307.0242, 295.0243, 282.0167Glycyrol5.04+++
75.333.24170C21H32O334.17−1.99177.0911, 145.0649, 137.059810-Shogaol ++
76.279.23264[M-H]−C18H32O238.271.13261.2219, 199.8500Linoleic acid ++
Saponins
77.879.40173[M-H]− 881.41516
705.38361[M+H-glcA]+
511.34122[agl+H-H2O]+
C44H64O1824.011−0.67
−1.38
(−) 351.0557, 193.0342, 113.0229
(+) 511.3408, 493.3279, 451.3188, 141.0183
Uralsaponin M ++
78.837.39105[M-H]−
839.40466
469.33072[gal+H-H2O]+
C42H62O1725.49−0.79
−1.32
(−) 351.05603, 289.05652, 193.03430, 175.02340, 113.02294
(+) 469.3304, 487.3415, 451.3209, 141.0184
Yunganoside K2 ++
79.837.39178[M-H]−
839.40491
469.33084[agl+H-H2O]+
C42H62O1726.07−0.84
−1.07
(−) 351.05557, 289.05621, 193.03413, 175.02360, 113.02285
(+) 469.3304, 487.3413, 451.3198, 141.0183
Licoricesaponin G2 +
80.471.34613C30H46O426.62−1.41453.33508, 425.34262, 317.21100, 235.16887, 189.16374Glycyrrhetinic acid (enoxolone) * +
81.821.39630[M-H]−
823.40936
647.37744[M+H-glcA]+
453.33554[agl+H-H2O]+
C42H62O1626.641.08
−1.70
(−) 351.05573, 193.03406, 175.02338, 113.02288
(+) 453.3354, 471.3451, 435.3259
Glycyrrhizic acid * ++++
82.821.39612[M-H]−
823.40936
647.37787[M+H-glcA]+
453.33585[agl+H-H2O]+
C42H62O1627.650.86
−0.47
(−) 351.05640, 193.03404, 175.02319, 113.02289
(+) 453.3354, 435.3257
Uralsaponin B or Licoricesaponine K2/H2 ++
Note: * Compounds identified by comparing with reference standards; glcA: β-D-glucuronopyronosyl; agl: aglycone; +, response area below 106; ++, response area between 106 and 107; +++, response area between 107 and 108; ++++, response area above 108.
Table 3. Metabolites of major alkaloids found in the urine and plasma samples.
Table 3. Metabolites of major alkaloids found in the urine and plasma samples.
ID[M+H]+
(m/z)
FormulatR
(min)
Error
(ppm)
ms/msComposition ChangeIdentificationClogPArea
UrinePlasma
M1.410.25302C22H35NO63.72−1.69392.2425, 374.2317, 360.2165, 342.2054+OHydroxy karakolidine ++
M2.374.23212C22H31NO47.55−1.25356.2212, 338.2106, 198.1122+OHydroxy songorine ++
M3.394.25812C22H35NO57.76−1.85376.2476, 358.2362, 98.0971, 58.0611+OHydroxy karakoline ++
M4.438.28433C24H39NO610.760.67406.2588, 388.2476, 374.230, 356.2226+O10-Hydroxy talatizamine ++
M5.632.30560C33H45NO1122.61−1.50572.2853, 540.2590, 512.2641, 508.2310, 480.2390, 358.2004, 354.1703, 105.0341+OHydroxy hypaconitine ++
M6.392.24268C22H33NO58.28−1.21374.2315, 344.2221, 312.1962, 114.0916-H2Dehydrogenated karakolidine +++
M7.392.24249C22H33NO58.95−2.15374.2325, 344.2240-H2Dehydrogenated karakolidine +++
M8.452.26361C24H37NO79.85−1.48434.2529, 416.2419, 204.2270, 384.2155-H2Dehydrogenated fuziline +++
M9.376.24780C22H33NO410.38−1.82358.2373, 98.0969-H2Dehydrogenated karakoline ++
M10.376.24756C22H33NO410.96−1.15358.2375, 234.0137, 98.0970-H2Dehydrogenated karakoline +++
M11.436.26895C24H37NO610.24−0.95418.2581, 400.2475, 386.2315, 358.2355, 340.2265-H2Dehydrogenated neoline ++++
M12.436.26907C24H37NO610.64−0.67418.2585, 400.2473, 386.2303, 358.2383-H2Dehydrogenated neoline +++
M13.420.27393C24H37NO513.60−1.33388.2477, 370.2375, 98.0972-H214-Dehydrogenated talatizamine +++
M14.364.24744C21H33NO47.39−2.20346.2374, 328.2268-CH2Demethyl karakoline ++++
M15.364.24740C21H33NO47.86−2.20346.2370, 328.2266-CH2Demethyl karakoline +++++
M16.424.26892C23H37NO610.68−1.05406.2583, 374.2327, 356.2211, 342.2069, 154.1226-CH2Demethyl neoline ++++
M17.424.26890C23H37NO611.08−0.98406.2581, 374.2317, 356.2222, 342.2076, 154.1228-CH2Demethyl neoline ++++
M18.408.27386C23H37NO59.87−1.44376.2475, 358.2365, 326.2136-CH218-o-Demethyl talatizamine −0.78++++++
M19.408.27393C23H37NO511.17−1.29376.2478, 358.2373, 326.2129-CH216-o-Demethyl talatizamine−0.73++++++
M20.602.29486C32H43NO1021.31−1.70542.2742, 510.2477, 482.2540, 478.2212, 324.1592, 105.0339-CH2Demethyl hypaconitine +++++
M21.618.28992C32H43NO1122.17−1.22558.2684, 526.2423, 508.2394, 354.1695, 105.0341-CH2Demethyl mesaconitine ++
M22.330.20581C20H27NO38.19−1.70312.1954-C2H4Deethyl songorine +++
M23.350.23181C20H31NO49.05−2.21332.2215, 314.2106, 300.1958, 234.9901, 158.9743-C2H4Deethyl karakoline +++
M24.410.25314C22H35NO610.97−1.40392.2423, 378.2271, 360.2163, 328.1906-C2H4Deethyl neoline ++
M25.408.27374C23H37NO58.96−1.74390.2631, 372.2537, 358.2369-CH2ODemethyl-deoxy neoline ++++++
M26.392.27896C23H37NO413.38−1.46360.2527, 342.2436, 328.2265-CH2O16-O-Demethyl-14-deoxy Talatizamine +++
M27.602.29529C32H43NO1023.87−1.20542.2773, 510.2486, 478.2222, 324.1592, 105.0341-CH2ODemethyl-deoxy mesaconitine +++
M28.360.25272C22H33NO39.38−1.68342.2422, 324.2325, 121.0651-H2ODehydrated karakoline ++++
M29.360.25250C22H33NO39.92−2.11342.2422, 324.2307-H2ODehydrated karakoline +++++
M30.614.2517C33H43NO1024.17−1.57544.2743, 522.2518, 494.2534, 372.2162, 344.2215-H2ODehydrated mesaconitine ++
M31.380.24277C22H33NO58.81−1.01362.2316, 344.2046, 330.2065-CH2+ODemethyl-hydroxy karakoline ++
M32.438.24811C23H35NO712.09−1.06420.2374, 402.2265, 392.2440, 374.2317-CH2-H2Dehydrogenated-demethyl fuziline +++
M33.438.24890C23H35NO712.210.49420.2367, 402.2283, 392.2442, 374.2323-CH2-H2Dehydrogenated-demethyl fuziline +++
M34.362.23172C21H31NO48.15−0.45344.2223, 185.0710-CH2-H2Dehydrogenated-demethyl karakoline +++
M35.422.25327C23H35NO610.99−1.07390.2268, 406.2597, 390.2268, 374.2324-CH2-H2Dehydrogenated-demethyl neoline ++++
M36.406.25839C23H35NO510.70−1.01388.2477, 370.2368, 328.2266-CH2-H214-Dehydrogenated-16-O-demethyl talatizamine +++
M37.346.20090C20H27NO47.79−1.12328.1904, 296.1645, 268.1701, 251.1437-C2H4+ON-Deethyl-hydroxy songorine +++
M38.346.20071C20H27NO48.49−1.65328.1903, 296.1650, 268.1699, 251.1429-C2H4+ON-Deethyl-hydroxy songorine +++
M39.378.26337C22H35NO410.98−1.43346.2371, 328.2279-C2H4-ON-Deethyl-14-deoxy talatizamine0.88++++
M40.378.26334C22H35NO411.23−1.45346.2371, 328.2267-C2H4-ON-Deethyl-8-deoxy talatizamine1.15+++
Note: +, response area below 106; ++, response area between 106 and 107; +++, response area between 107 and 108; ++++, response area above 108.
Table 4. Metabolites of phenolic compounds found in the urine and plasma samples.
Table 4. Metabolites of phenolic compounds found in the urine and plasma samples.
ID[M+H]+
(m/z)
FormulatR
(min)
Error
(ppm)
ms/msComposition ChangeIdentificationArea
Urine Plasma
M41.259.09701C15H14O417.572.53153.0548, 135.0441, 107.0496+H2Hydrogenated liquiritigenin++++++
M42.259.09689C15H14O420.781.83153.0549, 107.0497+H2Hydrogenated isoliquiritigenin+++++++
M43.297.20602C17H28O422.11−0.23177.0912, 163.0755, 137.0598, 131.0494+H2Hydrogenated 6-gingerol++++++
M44.269.08170[M-H]−C16H14O426.82−0.03254.0582, 153.0178, 135.0073, 91.0173+H2Hydrogenated formononetin++++++
M45.367.11792[M-H]−C21H20O629.54−2.01352.0936, 309.0400, 310.0434, 284.0325+H2Hydrogenated glycyrol++++
M46.355.15311C21H22O530.71−2.57337.1065, 299.0912, 189.0911, 177.0546, 151.0393+H2Hydrogenated gancaonin M++
M47.309.20578C18H28O426.17−0.70163.0756, 137.0599, 131.0494+CH2Methyl 6-gingerol++
M48.309.20572C18H28O426.80−0.92179.0704, 150.068, 137.0598, 83.0864+CH2Methyl 6-gingerol++
M49.285.07587C16H12O518.490.51270.0525, 253.0499, 299.0866, 225.0546, 123.0443+OHydroxy formononetin+++
M50.273.07593C15H12O519.060.72255.066, 179.0339, 153.0184, 147.0442, 123.044, 119.0496+OHydroxy liquiritigenin/isoliquiritigenin+++++
M51.369.13266C21H20O629.18−1.71351.1222, 229.0860, 193.0497, 165.0548, 151.0389+OHydroxy gancaonin M++++
M52.313.20038C17H28O515.07−0.96203.1066, 163.0754, 137.0598+H2OHydrated 6-gingerol+++
M53.273.07629[M-H]−C15H14O520.08−1.36255.0661, 167.0337, 109.0279+H2OHydrated liquiritigenin+++
M54.273.07660[M-H]−C15H14O523.05−0.68255.0655, 151.0387, 135.0072, 109.0280+H2OHydrated isoliquiritigenin+++
M55.293.17447C17H24O416.57−0.91163.0756, 137.0598, 99.0811-H2Dehydrogenated 6-gingerol+++
M56.255.06552C15H10O418.490.41227.0703, 199.0756, 137.0234-H2Dehydrogenated liquiritigenin++++
M57.255.06550C15H10O421.29−0.16227.0699, 199.0755, 137.0234-H2Dehydrogenated isoliquiritigenin+++++
M58.307.15466[M-H]−C17H24O526.17−1.41275.1288, 171.1014, 153.0907, 121.0280, 111.0799-H2+ODehydrogenated-hydroxy 6-gingerol++
M59.277.18039C17H24O328.852.07189.0914, 177.09123, 145.05493, 137.0597-H2-ODehydrated 6-gingerol++++++
M60.285.07648[M-H]−C16H14O522.03−0.35270.0533, 153.0180, 149.0594, 135.0073, 134.0358, 91.0174+CH2+OMethyl-hydroxy liquiritigenin++++
M61.285.07645[M-H]−C16H14O526.88−1.06270.0535, 153.0180, 149.0595, 135.0073, 91.0174+CH2+OMethyl-hydroxy isoliquiritigenin++
M62.299.09170C17H14O526.990.69284.0680, 243.1061, 166.0268+CH2+OMethyl-hydroxy formononetin++++
M63.335.02261[M-H]−C15H12O7S18.62−1.33255.0661, 199.0064, 135.0073, 119.0487+SO3Liquiritigenin sulfate+++++++
M64.335.02271[M-H]−C15H12O7S23.32−0.94255.0663, 199.0055, 135.0073, 119.0486+SO3Isoliquiritigenin sulfate++++++++
M65.347.02263[M-H]−C16H12O7S23.56−1.41267.0664, 252.0427+SO3Formononetin sulfate+++++++
Note: +, response area below 106; ++, response area between 106 and 107; +++, response area between 107 and 108; ++++, response area above 108.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zheng, X.; Zhan, Y.; Peng, M.; Xu, W.; Deng, G. Metabolite Profiling Analysis of the Tongmai Sini Decoction in Rats after Oral Administration through UHPLC-Q-Exactive-MS/MS. Metabolites 2024, 14, 333. https://doi.org/10.3390/metabo14060333

AMA Style

Zheng X, Zhan Y, Peng M, Xu W, Deng G. Metabolite Profiling Analysis of the Tongmai Sini Decoction in Rats after Oral Administration through UHPLC-Q-Exactive-MS/MS. Metabolites. 2024; 14(6):333. https://doi.org/10.3390/metabo14060333

Chicago/Turabian Style

Zheng, Xianhui, Yingying Zhan, Mengling Peng, Wen Xu, and Guanghai Deng. 2024. "Metabolite Profiling Analysis of the Tongmai Sini Decoction in Rats after Oral Administration through UHPLC-Q-Exactive-MS/MS" Metabolites 14, no. 6: 333. https://doi.org/10.3390/metabo14060333

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop