Next Article in Journal
Recent Advances in the Semisynthesis, Modifications and Biological Activities of Ocotillol-Type Triterpenoids
Previous Article in Journal
Mucuna pruriens Administration Minimizes Neuroinflammation and Shows Anxiolytic, Antidepressant and Slimming Effects in Obese Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Exploring the In Vivo Existence Forms (23 Original Constituents and 147 Metabolites) of Astragali Radix Total Flavonoids and Their Distributions in Rats Using HPLC-DAD-ESI-IT-TOF-MSn

State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No. 38 Xueyuan Road, Beijing 100191, China
*
Authors to whom correspondence should be addressed.
Molecules 2020, 25(23), 5560; https://doi.org/10.3390/molecules25235560
Submission received: 1 November 2020 / Revised: 22 November 2020 / Accepted: 23 November 2020 / Published: 26 November 2020

Abstract

:
Astragali Radix total flavonoids (ARTF) is one of the main bioactive components of Astragali Radix (AR), and has many pharmacological effects. However, its metabolism and effective forms remains unclear. The HPLC-DAD-ESI-IT-TOF-MSn technique was used to screen and tentatively identify the in vivo original constituents and metabolites of ARTF and to clarify their distribution in rats after oral administration. In addition, modern chromatographic methods were used to isolate the main metabolites from rat urine and NMR spectroscopy was used to elucidate their structures. As a result, 170 compounds (23 original constituents and 147 metabolites) were tentatively identified as forms existing in vivo, 13 of which have the same pharmacological effect with ARTF. Among 170 compounds, three were newly detected original constituents in vivo and 89 were new metabolites of ARTF, from which 12 metabolites were regarded as new compounds. Nineteen original constituents and 65 metabolites were detected in 10 organs. Four metabolites were isolated and identified from rat urine, including a new compound (calycoisn-3’-O-glucuronide methyl ester), a firstly-isolated metabolite (astraisoflavan-7-O-glucoside-2’-O-glucuronide), and two known metabolites (daidzein-7-O-sulfate and calycosin-3’-O-glucuronide). The original constituents and metabolites existing in vivo may be material basis for ARTF efficacy, and these findings are helpful for further clarifying the effective forms of ARTF.

1. Introduction

Astragali Radix total flavonoids (ARTF) is one of the main bioactive components of Astragali Radix (AR, Huangqi in Chinese) [1]. Many studies have proven its cardiovascular protective effect, owing that it exhibited a protective effect on an ischemia-reperfusion model by effectively inhibiting the free radical spectrum [2,3], and exhibited vasorelaxant and endothelial-protective effect via the Akt/eNOS signaling pathway [4]. ARTF has an obvious protective effect on the inflammatory response in brain tissue of a natural aging rat by reducing the expression level of the downstream inflammatory factors [5]. ARTF also has immunostimulatory and anti-inflammatory effects via regulating MAPK (Mitogen-Activated Protein Kinase) and NF-κB signaling pathways [6,7]. ARTF has a protective effect against hepatic damage induced by paracetamol [8] or reperfusion [9] as well. In a word, ARTF has a wide range of pharmacological actions.
Up to now, besides studies on the pharmacological actions of AR, many investigations have been conducted in the field of phytochemistry. Over 70 flavonoid compounds have been isolated and identified from AR by modern chromatographic and spectroscopic methods [10,11,12], and 421 flavonoids have been detected and characterized from AR (the roots of Astragalus membranaceus (Fisch.) Bge. var. mongholicus (Bge.) Hsiao) by HPLC-MS (High Performance Liquid Chromatography-Mass Spectrometry) technology [13]. All these findings indicate that many kinds of constituents exist in ARTF. The metabolism of some high content flavonoid compounds in ARTF have been reported by our research group. Forty-one and 21 metabolites have been identified from the urine of rats after administration of calycosin-7-O-glucoside [14] and ononin [15], respectively. Twenty-six and 14 metabolites have been identified when calycosin [14] and formononetin [15] were incubated with rat liver S9. Relatively, the metabolic studies of isoflavan and pterocarpan are pretty few, and only our research group has done several before. Twenty-one and 20 metabolites of astrapterocarpan-3-O-glucoside, astraisoflavan-7-O-glucoside have been identified in rats, respectively [15]. Forty and 19 metabolites of astrapterocarpan [16] and astraisoflavan [15] have been detected when incubated with rat liver S9, respectively. And many other research groups also have identified some metabolites of these four main isoflavones (calycosin [17], calycosin-7-O-glucoside [18], formononetin [19,20], ononin [21]) in different test systems, such as human gut microbiota [18], zebrafish larvae [17], sheep [19], human liver microsome [20,21], and so on.
However, the forms existing in vivo (i.e., absorbed constituents and metabolites) of ARTF remains unclear. Our research group had done a study on ARTF metabolism before, and identified 127, 43, and 22 compounds in the urine, plasma, and feces, respectively [22]. But in that study, no glucuronides of ARTF were found, which should be main metabolites of flavonoid compounds; the distribution of ARTF was not investigated either. And the dose schedule of ARTF was once per day for seven days. As is well known, it usually needs a long dose period for traditional Chinese medicines to treat diseases in clinical practice. As for AR, it can be used for a long time [23]. Therefore, to better simulate the long dose period of AR and understand the existence forms in vivo of ARTF under long-term administration situation, this study was performed. The rats were orally administrated with ARTF twice a day for 31 days, and the samples of urine, plasma, feces, and organs were collected to identify original constituents and metabolites that existed in vivo, and to study the distribution of the existence forms in vivo. After that, the ARTF-containing urine was used to isolate some major metabolites.

2. Results and Discussion

To know which compounds exist in vivo, which is one of the prerequisites for the compounds to be effective forms, the compounds in the bio-samples including urine, plasma, feces, and organs of the rats after oral administration of ARTF were analyzed. In total, one hundred and seventy compounds (23 original constituents and 147 metabolites) were identified, among which 12 were regarded as new compounds (they are all metabolites) by retrieving information from the SciFinder database, three were newly detected original constituents, and 89 were new metabolites of ARTF. In 147 metabolites, nine were phase I and 138 were phase II metabolites. Among 138 phase II metabolites, ninety-two sulfates, twenty-six glucuronides, thirteen both sulfuric acid, and glucuronic acid conjugates and seven methyl conjugates were included, which indicated that sulfates were the most important existence forms of ARTF. To better understand the distribution of compounds, ten different organs of the rat were analyzed as well. Nineteen original constituents including six ones detected only in the organs (F18F23) and 65 metabolites containing three ones detected only in the organs (M145M147) were identified. Three, 5, 4, 3, 5, 17, 7, 7, 6 original constituents together with 3, 18, 5, 6, 22, 30, 46, 21, 6 metabolites were identified in the heart, liver, spleen, lungs, kidneys, stomach, small intestine, colon intestine, and thymus, respectively. However, no compounds were found in the brain. Four metabolites were isolated from the ARTF-containing urine and identified by NMR. To better understand potential effective forms among those compounds in vivo, we retrieved over 40 compounds which have specific structures from SciFinder, and 13 compounds including six original constituents, and seven metabolites were found to possess the same pharmacological effects as ARTF. Most of the phase Ⅱ metabolites were not found to possess bioactivity, perhaps owing that this kind of material was difficult to gain and to study.

2.1. Identification of Original Constituents and Metabolites of ARTF in Rats

2.1.1. Identification of Original Constituents

The peaks which appeared at the same position in LC-MS chromatograms of both the administrated bio-samples and the ARTF but did not exist in the blank bio-samples were regarded as original constituents. By comparison of their extracted ion chromatograms (EICs) and base peak chromatograms (BPCs), twenty-three original constituents were identified (Table 1, F1F23, and F18F23 only detected in the organs). They were composed of 15 flavones or isoflavones, namely calycosin and its isomers (F1F3), calycosin-7-O-glucoside (F4), ononin (F5), formononetin (F6), isomers of pratensein (F7, F8), daidzein (F11), trihydroxyisoflavone/flavone (F13), isomer of odoratin (F14), dihydroxy dimethoxyisoflavone/flavone (F16), naringin (F18), pratensein glucoside and its isomer (F22, F23); four pterocarpans and isoflavanes, namely, astraisoflavane isomer (F15), astraptercarpan (F17), astrapterocarpan pentose glucoside (F20), 3,10-dihydroxy-9-methoxypterocarpan (F21); four dihydroisoflavones/flavones and chalcones, namely trihydroxy-dihydroisoflavone/flavone (F9), trihydroxy-tetrahydroisoflavone/flavone (F10), trihydroxychalcone (F12), dihydrocalycosin pentose glucoside (F19). Among the 23 original constituents, three were newly found original constituents of ARTF, namely F3, F7, and F8. Seventeen original constituents (F1F17) were found in the urine (Figure 1); F4F6 (Figure S1) were detected in the plasma; F1, F6, F12, and F17 (Figure S2) existed in the feces. The remaining six original constituents (F18F23; Table 1) were detected only in the organs.

2.1.2. Identification of Metabolites

The peaks only appearing in LC-MS chromatograms of ARTF-treated rat bio-samples, but not existing in either blank bio-samples or ARTF were regarded as metabolites. By comparing the EICs and BPCs of them, 147 peaks were assigned as metabolites (M1M147; Table 2). M145M147 were only found in the organs. One hundred and six, 64, and 17 metabolites were identified in the urine (Figure 2), plasma (Figure S3), and feces (Figure S4), respectively. Among the 147 metabolites, 89 were new metabolites of ARTF, from which 12 were regarded as new compounds by searching information from SciFinder database (their MS information was shown in Table S1 and Figures S5–S16). Eighty-nine new metabolites included the sulfates of the ring cleavage products of flavone, sulfates of oxidized, reduced, methylated astraisoflavan, and all the glucuronides as well as disulfates. And 12 potential new compounds were sulfates, disulfates, glucuronides, diglucuronides of tetrahydrocalycosin. By analyzing the structures of 147 metabolites, we speculated they were mainly derived from calycosin and its glycoside (maybe the sources of M17M72, M131M139), formononetin and its glycosides (maybe the sources of M73M78, M131M139), astrapterocarpan-3-O-glucoside (maybe the sources of M79M84), astraisoflavan-7-O-glucisode (maybe the sources of M85M106, M147) and many other low content constituents such as astrapterocarpan (maybe one of the sources of M79M84), astraisoflavan (maybe one of the sources of M85M106, M147), daidzein (maybe one source of M107M117, M131M139), genistein (maybe one source of M118M130), and so on. Hence, they were speculated to be the main sources of existence forms of ARTF. These metabolites could be classified into 9 phase I metabolites and 138 phase II metabolites. A hundred and thirty-eight phase II metabolites consisted of 7 methyl conjugates, 92 sulfates, 26 glucuronides, and 13 both sulfuric acid and glucuronic acid conjugates, which indicated that sulfates were the most important existence forms of ARTF. The structural elucidation process of some representative metabolites was described as follows.

Identification of the Sulfates of the Ring Cleavage Products of Flavone (M1M16)

A total of 16 compounds were assigned as metabolites originating from flavone, which underwent ring cleavage, then conjugation with sulfuric acid, and all of them were new metabolites of ARTF. M1 showed [M − H] at m/z 200.99 and its molecular formula was predicted as C7H6O5S, and the fragment ion at m/z 121.03 was formed by the neutral loss of 79.95 Da (SO3) in its MS2 spectra, so it was determined as hydroxyl-benzaladehyde sulfate or other isomers. M2 showed [M − H] at m/z 219.00 and its molecular formula was predicted as C7H8O6S, and the fragment ion generated by loss 79.95 Da could be detected at m/z 139.04, which was predicted as C7H8O3 and was one CH2 more than pyrogallol, so it was tentatively identified as methyl pyrogallol sulfate [24] or other isomers. M3 and M4 showed [M − H] at m/z 231.00, which indicated that their molecular formulae were C8H8O6S. In the MS2 spectra, fragment ions at m/z 151.04, 137.03 were formed by sequential losses of SO3 and CH2. According to literature, they were identified as hydroxy phenylacetic acid sulfate [25]. M5 and M6 showed [M − H] at m/z 243.00, which indicated that their molecular formulae were C9H8O6S, and according to the fragment ions at m/z 163.04, 119.05 and previous report, they were identified as hydroxycinnamic acid sulfate [26]. M7 showed [M − H] at m/z 243.03 and was predicted as C10H12O5S. And according to the fragment ions at m/z 163.08, 148.05 in MS2 spectra, it was regarded as eugenol sulfate [27]. M8 showed [M − H] at m/z 247.00 and was predicted as C8H8O7S, and yielded a fragment ion at m/z 167.03 by neutral loss of 79.95 Da (SO3), so it was determined as vanillic acid sulfate [28]. M9 showed [M − H] at m/z 247.03, which indicated that their molecular formulae were C9H12O6S, and according to the fragment ions at m/z 167.07, 153.09, 137.05 and previous report, it was identified as homovanillyl alcohol sulfate [29]. M10 showed [M − H] at m/z 273.00, which indicated that its molecular formula was C10H10O7S, and according to the fragment ions at m/z 178.03, 134.04, it was identified as ferulic acid sulfate [24]. M11 and M12 showed [M − H] at m/z 273.04, which indicated that their molecular formulae were C11H14O6S. And the fragment ions at m/z 193.09, 178.06, 163.03 were observed in MS2 spectra, which were 30.01 Da (OCH2) lager than that of the aglycon of M7. Therefore, they were tentatively determined as methoxyeugenol sulfate. M16 showed [M − H] at m/z 303.02, which indicated that its molecular formula was C11H12O8S, and the fragment ions at m/z 223.06, 208.04, 164.05, 149.02, were similar to the fragment ions of 4-hydroxy-3,5-dimethoxycinnamic acid, so it was determined as 4-hydroxy-3,5-dimethoxycinnamic acid sulfate [30].

Identification of the Calycosin-Related Metabolites (M17M72, M146)

M17 showed [M − H] at m/z 283.06 and its molecular formula was predicted as C16H12O5. The fragment ions at m/z 269.04, 268.04, 195.04 were detected in MS2, which were like those of calycosin, so it was identified as calycosin isomer. M18–M20 were identified as calycosin isomer owing that their predicted molecular formulae and fragment ions were like those of calycosin in the positive ion mode. M21 showed [M + H]+ at 301.06 and its molecule formula was predicted as C16H12O6, which had one more oxygen atom than that of calycosin, and the fragment ions at m/z 270.08, 197.08 were similar to those of calycosin in positive ion mode, so it was determined as hydroxycalycosin. The fragment ion of [aglycon−H] at m/z 283.06 could be detected in the MS2 spectra of M25–M34, which was predicted as C16H12O5, and other fragment ions were like those of calycosin, so they were determined as calycosin metabolites. M25M30 were predicted as C16H12O8S according to [M − H] at m/z 363.02, and the [aglycon − H] formed by the neutral loss of 79.95 Da (SO3) was detected. Therefore, they were determined as calycosin sulfate and its isomers. In the same way, M30 was determined as calycosin-7,3’-O-disulfate. M31 and M146 showed [M − H] at m/z 459.09 and their molecular formulae were predicted as C22H20O11, and the fragment ion of [aglycon − H] were formed by the neutral loss of 176.03 Da (C6H8O6) in the MS2 spectra, so they were determined as calycosin glucuronide. There are two positions (C-7-OH and C-3’-OH) in calycosin that could be linked with glucuronic acid, and C-3’-OH was the major position according to a previous study [31]. In addition, M31 was the main metabolite, hence it was determined as calycosin-3’-O-glucuronide, and M146 was determined as calycosin-7-O-glucuronide.
In the MS2 spectra of M35M39, the fragment ion of [aglycon − H] at m/z 299.05 can be detected, which were predicted as C16H11O6 and was the same as M21, so they were tentatively determined as metabolites of hydroxycalycosin. And according to molecule formulae and characteristic neutral losses, they were tentatively determined as hydroxycalycosin sulfate or glucuronide, respectively.
M42 showed [M − H] at m/z 621.15 and its molecular formula was predicted as C28H30O16. The fragment ion at m/z 283.06 predicted as C16H11O5 was detected in MS2 which was generated by sequential loss of 162.05 Da (C6H10O5), and 176.03 Da (C6H8O6). Since calycosin-7-O-glucoside was the main constituent of ARTF, so it was determined as calycosin-7-O-glucoside-3’-O-glucuronide.
The fragment ion of [aglycon − H] at m/z 285.08 could be detected in the MS2 spectra of M43M46, which was predicted as C16H13O5 and had 2H (2.01 Da) more than that of calycosin, so they were tentatively regarded as metabolites of dihydrocalycosin. The fragment ion of [aglycon − H] at m/z 287.09 could be detected in the MS2 spectra of M54M63, which were predicted as C16H15O5 and had 4H more than that of calycosin, so they were tentatively regarded as metabolites of tetrahydrocalycosin. In the MS2 spectra of M64M69, the fragment ion of [aglycon − H] at m/z 303.08 could be detected, which were predicted as C16H15O6 and had one more oxygen atom than that of tetrahydrocalycosin, so they were tentatively regarded as hydroxy tetrahydrocalycosin metabolites.

Identification of the Formononetin-Related Metabolites (M73M78)

M73 showed [M − H] at m/z 347.02 and its molecular formula was predicted as C16H12O7S. The fragment ion of m/z 267.07 in the MS2 spectra was formed by the neutral loss of 79.95 (SO3), which was predicted as C16H11O4, and its fragment ion m/z 252.03 was like that of formononetin. Because only C-7-OH could be sulfated, so M73 was determined as formononetin-7-O-sulfate. In the same way, M74 was determined as formononetin-7-O-glucuronide. M75M77 showed [M − H] at m/z 351.05 and their molecular formulae were predicted as C16H16O7S. The fragment ions at m/z 271.09 formed by neutral loss of 79.95 Da (SO3) was determined as C16H15O4, which had 4H more than that of formononetin, so they were tentatively determined as tetrahydroformononetin sulfate.

Identification of the Astrapterocarpan-Related Metabolites (M79M84)

M79 showed [M − H] at m/z 379.05 and its molecular formula was predicted as C17H16O8S. The fragment ions m/z 299.08 formed by a natural loss of 176.03 Da (C6H8O6) was predicted as C17H15O5, which was the same to that of astrapterocarpan. Owing that only C-3-OH of astrapterocarpan could be linked to sulfuric acid, it was determined as astrapterocarpan-3-O-sulfate. In the same way, M80 was determined as astrapterocarpan-3-O-glucuronide.
M82M84 showed [M − H] at m/z 491.12 and their molecular formulae were predicted as C23H24O12. The fragment ions at m/z 315.09 generated by neutral loss of 176.03 Da (C6H8O6) was predicted as C17H15O6, and had one more O than that of astrapterocarpan, so they were tentatively determined as hydroxyastrapterocarpan glucuronide.

Identification of the Astraisoflavan-Related Metabolites (M85M106, M147)

M85 showed [M + H]+ at m/z 303.13 and its molecular formula was predicted as C17H18O5. The fragment ions at m/z 167.10, 149.09, and 125.07 could be detected in MS2 spectra and were like those of astraisoflavan in positive ion mode. So, M85 was determined as astraisoflavan isomer.
M102M103 showed [M − H] at m/z 477.14 and their molecular formulae were predicted as C23H26O11. The fragment ions at m/z 301.10 in MS2 spectra formed by a neutral loss of 176.03 Da (C6H8O6) was the same to that of astraisoflavan. Therefore, they were determined as astraisoflavan glucuronide. Because there are only two glucuronidation sites (C-7-OH, C-2’-OH) in astraisoflavan, and a larger CLogP value means a lower polarity and a larger retention time in reversed phase HPLC, M102 (CLogP = 3.6083, tR = 58.702 min) was determined as astraisoflavan-7-O-glucuronide and M103 (CLogP = 3.2673, tR = 57.802 min) was determined as astraisoflavan-2’-O-glucuronide.

Identification of the Daidzein-Related Metabolites (M107M117)

M107 and M108 showed [M − H] at m/z 333.00 and their molecular formulae were predicted as C15H10O7S. The fragment ion at m/z 253.05 in MS2 spectra formed by the neutral loss of 79.95 Da (SO3), and it was predicted as daidzein owing that the fragment was predicted as C15H9O4 and m/z 225.05, 197.06, 135.01 were detected in MS3 spectra. Because there are two sulfation sites (C-7-OH, C-4’-OH) in daidzein, and a larger CLogP value means a lower polarity and a larger retention time in reversed phase HPLC, M107 (CLogP = 0.4985, tR = 41.557 min) was determined as daidzein-4’-O-sulfate and M108 (CLogP = 0.3050, tR = 28.925 min) was determined as daidzein-7-O-sulfate.
M112 showed [M − H] at m/z 335.02 and its molecular formula was predicted as C15H12O7S. In MS2 spectra, the fragment ion at m/z 255.06 was formed by the neutral loss of 79.95 Da (SO3), which was predicted as C15H11O4 and had 2H more than that of daidzein. Therefore, it was tentatively determined as dihydrodaidzein sulfate.
M113M116 showed [M − H] at m/z 337.04 and their molecular formulae were predicted as C15H14O7S. The fragment ion at m/z 257.08 in MS2 spectra formed by the neutral loss of 79.95 Da (SO3), which was predicted as C15H13O4 and had 4H more than that of daidzein. Therefore, they were tentatively determined as tetrahydrodaidzein sulfate.

Identification of the Genistein-Related Metabolites (M118M130)

M118 showed [M + H]+ at m/z 271.06 and its molecule formula was predicted as C15H10O5. The fragment ions at m/z 253.01, 225.06, 215.07 which were like those of genistein in reported literature [32], so it was determined as genistein.
M123M125 showed [M − H] at m/z 351.02 and their molecular formulae were predicted as C15H12O8S. The fragment ion at m/z 271.06 was formed by a neutral loss 79.95 Da (SO3), which was predicted as C15H11O5 and had 2H more than that of genistein, so they were tentatively determined as dihydrogenistein sulfate.
M126 showed [M − H] at m/z 273.07 and its molecular formula was predicted as C15H14O5, which was 4H more than that of genistein, so it was tentatively determined as tetrahydrogenistein. M130 showed [M − H] at m/z 515.09 and its molecules formula was predicted as C21H24O13S. The fragment ion at m/z 273.07 which was predicted as C15H13O5 formed by a neutral loss of 162.05 Da (C6H10O5) and 79.95 Da (SO3), so it was tentatively determined as tetrahydrogenistin sulfate.

Identification of the Equol-Related Metabolites (M131M139)

In the MS2 spectra of M131M138, the fragment ion of [aglycon − H] at m/z 241.09 could be detected, which were predicted as C15H13O3 and its fragment ions of m/z 135.05, 121.04, 119.06 were like those of equol [33], so they were regarded as equol metabolites. M136M138 showed [M − H] at m/z 497.07 and their molecules formulae were predicted as C21H22O12S, and the [aglycon − H] formed by the neutral loss of 176.03 Da (C6H8O6), 79.97 Da (SO3). Therefore, they were determined as equol glucuronide sulfate. M139 showed [M − H] at m/z 323.06 and its molecular formula was predicted as C15H16O6S. The fragment ion at m/z 243.10 formed by a neutral loss of 79.95 Da (SO3), which was predicted as C15H15O3 and 2H (2.01Da) more than that of equol. Hence, it was tentatively determined as dihydroequol sulfate.

Identification of the other Metabolites (M140M145)

M140 showed [M + H]+ at m/z 303.09 and its molecular formula was predicted as C16H14O6, which had 2H more than that of pratensein. Therefore, it was tentatively determined as dihydropratensein. M141 and M145 showed [M − H] at m/z 555.05 and their molecular formulae were predicted as C22H20O15S. The fragment ions at m/z 299.05 predicted as C16H11O6 was formed by a neutral loss of 176.03 Da (C6H8O6) and 79.95 Da (SO3). Therefore, it was tentatively determined as pratensein glucuronide sulfate.

2.2. Distribution of Original Constituents and Metabolites of ARTF in Rats Organs

2.2.1. Distribution of Original Constituents

Nineteen original constituents were detected in the organs, with zero in brain, three in heart, five in liver, four in spleen, three in lung, five in kidney, seventeen in stomach, seven in small intestine, seven in colon intestine, and six in thymus, respectively (Table S2, Figure S17a–S25a). Six (F18F23) were only detected in the organs and were not detected in the urine, plasma, and feces. Calycosin (F1), formononetin (F6), daidzein (F11), and naringin (F18) were widely distributed, which could be detected in seven and even more organs, and these compounds may be important material basis for the efficacy of ARTF.

2.2.2. Distribution of Metabolites

Sixty-five metabolites were identified in the organs, of which three metabolites (M145M147) were only detected in the organs, and 0, 3, 18, 5, 6, 22, 30, 46, 21, and 6 were identified in the brain, heart, liver, spleen, lung, kidney, stomach, small intestine, colon, and thymus (Table S3, Figures S17b–S25b), respectively. Twelve metabolites containing seven sulfates and five glucuronides were distributed widely in five and even more tissues. Seven sulfates were calycosin sulfate (M26), tetrahydrocalycosin sulfate (M57, M58), hydroxyastraisoflavan sulfate (M96), daidzein-4’-O-sulfate (M107), equol sulfate (M132, M133), respectively. Five glucuronides consisted of calycosin-3’-O-glucuronide (M32), dimethoxy hydroxytetrahydrocalycosin glucuronide (M72), astrapterocarpan-3-O-glucuronide (M80), astraisoflavan-2’-O-glucuronide (M103), and astraisoflavan-7-O-glucoside-2’-O-glucuronide (M106). These widely distributed metabolites may play an important role in the efficacies of ARTF.

2.3. Identification of Metabolites Isolated from Rat Urine

MI-1 (M108) was obtained as a white powder and assigned a molecular formula of C15H10O7S based on its HR-ESI-MS mass spectrum, which showed a quasi-molecular ion peak [M − H] at m/z 333.0076 (calcd. for C15H10O7S 333.0069). The main fragment ion was m/z 253.0505 [M − SO3 − H] in MS2 spectra, so it was regarded as a sulfate. MI-1 (M108):13C-NMR (DMSO-d6, 100MHz) ppm: 153.6 (C-2), 122.5 (C-3), 175.0 (C-4), 129.7 (C-5), 118.0 (C-6), 158.1 (C-7), 107.1 (C-8), 156.6 (C-9), 119.0 (C-10), 123.7 (C-1’), 130.2 (C-2’, C-6’), 115.08 (C-3’, C-5’), 157.3 (C-4’), which were in consistent with daidzein [34]. 1H-NMR spectra of MI-1 (M108) (DMSO-d6, 400MHz) ppm: 8.38 (1H, s, H-2), 8.03 (1H, d, J = 8.8Hz, H-5), 7.43 (1H, d, J = 2.1Hz, H-8), 7.40 (2H, d, J = 8.3Hz, H-2’ and H-6’), 7.25 (1H, dd, J = 8.8Hz, 2.1Hz, H-6), 6.81 (2H, d, J = 8.3Hz, H-3’ and H-5’), which were like those of daidzein-7-O-sulfate reported in literature [35].Given all of this, MI-1 was determined as daidzein-7-O-sulfate. Its structure and NMR spectroscopy were shown in Figure 3a and Figure S26, respectively.
MI-2 (M32) was obtained as a white powder and assigned a molecular formula of C22H20O11 based on its HR-ESI-MS mass spectrum, which showed a quasi-molecular ion peak [M − H] t m/z 459.0943 (calcd. for C22H20O11 459.0935). The main fragment ion was m/z 283.0609 [M − C6H8O6 − H] in MS2 spectra, so it was regarded as a glucuronide. MI-2 (M32): 1H-NMR spectra (DMSO-d6, 400MHz) ppm: 8.31 (1H, s, H-2), 7.97 (1H, d, J = 8.8Hz, H-5), 6.94 (1H, dd, J = 8.8Hz, 2.2Hz, H-6), 6.87 (1H, d, J = 2.2Hz, H-8), 7.29 (1H, d, J = 2.0Hz, H-2’), 7.04 (1H, d, J = 8.5Hz, H-5’), 7.23 (1H, dd, J = 8.5Hz, 2.0Hz, H-6’), 3.79 (3H, s, C-4’-OCH3), 10.88 (1H, s, C-7-OH). 13C-NMR (DMSO-d6, 100MHz) ppm: 153.5 (C-2), 123.3 (C-3), 174.7 (C-4), 127.4 (C-5), 115.4 (C-6), 162.7 (C-7), 102.3 (C-8), 157.5 (C-9), 116.4 (C-10), 124.5 (C-1’), 116.7 (C-2’), 145.7 (C-3’), 149.2 (C-4’), 112.6 (C-5’), 123.3 (C-6’), 55.9 (C-4’-OCH3), which were similar to 13C-NMR of calycosin reported in literature [36]. The characteristic signals of six carbons in glucuronide were 100.2 (C-1’’), 73.1 (C-2’’), 76.3 (C-3’’), 71.6 (C-4’’), 75.6 (C-5’’), 170.2 (C-6’’), and all were consisted with calycosin-3’-O-glucuronide [31]. Based on the above analysis, MI-2 (M32) was determined as calycosin-3’-O-glucuronide. Its structure and NMR spectroscopy were shown in Figure 3b and Figure S27, respectively.
MI-3 was obtained as a white powder and assigned a molecular formula of C23H22O11 based on its HR-ESI-MS mass spectrum, which showed a quasi-molecular ion peak [M − H] at m/z 473.1113 (calcd. for C23H22O11 473.1084). The main fragment ion was m/z 283.0601 [M − C7H10O6 − H] in MS2 spectra, so it was predicted as a glucuronide methyl ester. 1H-NMR (DMSO-d6, 400MHz) ppm: 8.31 (1H, s, H-2), 7.97 (1H, d, J = 8.7Hz, H-5), 6.94 (1H, dd, J = 8.7Hz, 2.2Hz, H-6), 6.87 (1H, d, J = 2.2Hz, H-8), 7.29 (1H, d, J = 2.0Hz, H-2’), 7.05 (1H, d, J = 8.5Hz, H-5’), 7.23 (1H, dd, J = 8.5Hz, 2.0Hz, H-6’), 3.79 (3H, s, C-4’-OCH3), 10.77 (1H, s, C-7-OH), 3.62 (3H, s, C-6’’-OCH3). 13C-NMR (DMSO-d6, 100MHZ) ppm: 153.4 (C-2), 123.2 (C-3), 174.5 (C-4), 127.3 (C-5), 115.3 (C-6), 162.8 (C-7), 102.1 (C-8), 157.4 (C-9), 116.2 (C-10), 124.5 (C-1’), 116.6 (C-2’), 145.5 (C-3’), 149.0 (C-4’), 112.4 (C-5’), 123.0 (C-6’), 55.8 (C-4’-OCH3), were carbon signals of calycosin [31], 99.9 (C-1’’), 73.0 (C-2’’), 75.8 (C-3’’), 71.4 (C-4’’), 75.2 (C-5’’), 169.2 (C-6’’) were carbon symbols of glucuronide, which were similar to those of calycosin-3’-O-glucuronide [31]. Compared with that, an additional methoxy carbon signal at δ52.0 was observed, and H signal of this methoxy at δ3.62 (3H, s, C-6’’-OCH3) was in correlated with carbonyl carbon signal of glucuronide at δ169.2 (C-6’’) in HMBC spectra, indicating that the methoxy was linked to the carbonyl of glucuronide. According to all above analysis, MI-3 was identified as calycoisn-3’-O-glucuronide methyl ester (Figure 3c). It was a new compound. And its NMR spectroscopy was shown in Figure S28. But unfortunately, MI-3 could not be detected in the bio-samples using the HPLC-DAD-ESI-IT-TOF-MSn technique. Therefore, MI-3 was maybe produced during the isolation process.
MI-4 (M106) was obtained as a faint yellow powder and assigned a molecular formula of C29H36O16 based on its HR-ESI-MS mass spectrum, which showed a quasi-molecular ion peak [M − H] at m/z 639.1959 (calcd. for C22H20O11 639.1925). The main fragment ion was m/z 463.1650 [M − C6H10O5 − H], 301.1066 [M − C6H10O5 − C6H8O8 − H] in MS2 spectra, so it was regarded as glucoside and glucuronide. 1H-NMR (DMSO-d6, 400MHz) ppm: 3.86 (1H, t, H-2a), 4.23 (1H, d, J = 8.0Hz, H-2b), 3.59 (1H, m, H-3), 2.74 (1H, m, H-4a), 2.84 (1H, m, H-4b), 6.98 (1H, d, J = 8.4Hz, H-5), 6.54 (1H, dd, J = 8.4 Hz, 2.4Hz, H-6), 6.48 (1H, d, J = 2.4Hz, H-8), 6.81 (1H, d, J = 8.8Hz, H-5’), 6.92 (1H, d, J = 8.8Hz, H-6’), 3.72 (3H, s, C-3’-OCH3), 3.77 (3H, s, C-4’-OCH3). 13C-NMR (DMSO-d6, 100MHZ) ppm: 69.8 (C-2), 30.0 (C-3), 30.9 (C-4), 130.0 (C-5), 108.8 (C-6), 156.8 (C-7), 104.0 (C-8), 115.9 (C-9), 154.6 (C-10), 128.4 (C-1’), 147.2 (C-2’), 141.1 (C-3’), 152.1 (C-4’), 121.8 (C-5’), 103.2 (C-6’), 60.5 (C-3’-OCH3), 55.8 (C-4’-OCH3), 100.8 (C-1’’), 73.7 (C-2’’), 76.6 (C-3’’), 69.5 (C-4’’), 77.1 (C-5’’), 60.8 (C-6’’) were carbon signal of astraisoflavan-7-O-glucoside [36]. And 100.9 (C-1’’’), 73.3 (C-2’’’), 75.8 (C-3’’’), 71.5 (C-4’’’), 75.7 (C-5’’’), 170.1 (C-6’’’) were carbon signal of glucuronide. According to HMBC spectra, δ4.90, which was the terminal hydrogen signal of glucuronide, related to the δ147.2 (C-2’), which indicated that glucuronide was linked to C-2’. At the same time, the terminal hydrogen of glucoside δ4.79 was in correlated with glucoside carbon at δ156.8 (C-7), which indicated that glucoside was linked to C-7. Considering all of the above, MI-4 (M106) was determined as astraisoflavan-7-O-glucoside-2’-O-glucuronide (Figure 3d). It was a compound that isolated and identified by NMR for the first time. And its NMR spectroscopy was shown in Figure S29.

2.4. ARTF-Related Pharmacological Effect of Compounds In Vivo

The pharmacological literature of over 40 existence forms of ARTF which have specific or potential structure were retrieved from SciFinder and then analyzed. We found that 13 existence forms showed related pharmacological effect to ARTF, such as cardiovascular protective, neuroprotective, anti-inflammatory, and so on (Table S4). Six of them were original constituents, namely calycosin (F1), calycosin-7-O-glucoside (F4), ononin (F5), formononetin (F6), daidzein (F11), naringin (F18); seven of them were metabolites, namely daidzein-4’-O-sulfate (M107), daidzein-7-O-sulfate (M108), daidzein-7-O-glucuronide or daidzein-4’-O-glucuronide (M110), genistein (M118), genistein-7-O-sulfate and genistein-4′-O-sulfate (two of M119–M121), and equol-7-O-sulfate (M132 or M133). From Table S4, we could find that the metabolites in Table S4, especially phase Ⅱ metabolites, could activate estrogen receptor (ER). ER activation is associated with cardiovascular protective [37] and anti-inflammatory [38] effects, which is the main pharmacological effect of ARTF. In addition, we predicted that sulfate of flavonoids might have an effect on ER by molecular docking technique (data not shown). Furthermore, phase Ⅱ metabolites, especially sulfates, were the main existence forms of ARTF, so it could be speculated that some existence forms in vivo might be the material bases of the efficacies of ARTF, i.e., its effective forms.

3. Materials and Methods

3.1. Chemicals and Materials

ARTF (lot: 20170730) was obtained from Shanxi Baoji Herbest Biotech Co., Ltd. (Baoji, Shanxi, China) in August 2017 and its content was 62.7%, and the content of six main flavonoids namely calycosin-7-O-glucoside, calycosin, ononin, formononetin, astrapterocarpan-3-O-glucoside, and asisoflavan-7-O-glucoside was 11.3, 6.3, 5.8, 5.4, 7.2, and 1.6%, respectively, which was detected by HPLC-DAD-ELSD and calculated by using area normalization of ELSD (Evaporative Light Scattering Detector) chromatogram (Figure S30). The analysis of its constituents was conducted by HPLC-DAD-ESI-IT-TOF-MSn, and 69 constituents had been identified (Table S5).
Calycosin (lot: MUST-16031110), calycosin-7-O-glucoside (lot: MUST-16031205) were brought from Chengdu Must Biotech Co., Ltd. (Chengdu, Sichuang, China). Formononetin (lot: LS60Q22) was supplied by Beijing J&K Scientific Co., Ltd. (Beijing, China). Astrapterocarpan (lot: PRF7042221), astrapterocarpan-3-O-glucoside (lot: PRF7043003), astraisoflavan (lot: PRF7120221) were provided by Chengdu Biopurify Phytochemicals Ltd. (Chengdu, Sichuang, China). Astraisoflavan-7-O-glucoside (lot: 160217) was purchased from Chengdu Pufei De Biotech Co., Ltd. (Chengdu, Sichuang, China). Ononin was isolated in our laboratory [36]. The purities for all reference compounds were over 98%.
Acetonitrile (HPLC grade, lot: 184866), formic acid (LC/MS grade, lot: 182088) were purchased from Thermo Fisher Scientific (Waltham, MA, USA). Methanol was supplied by Tianjing Damao Co., Ltd. (Tianjing, China). Sodium carboxymethyl cellulose (CMC-Na, Analytical grade) was purchased from Tianjing Guangfu Fine Chemical Research Institute (Tianjin, China). XAD-2 macroporous resins (lot: 94664), ODS (lot:9833), sephadex LH-20 were applied by Supelco (Bellefonte, PA, USA), YMC (Kyoto, Japan), and Amersham Biociences (Boston, MA, USA), respectively. Ultra-pure water was obtained by a Millipore Milli-Q Integral 3 Ultrapure water system (Billerica, MA, USA).

3.2. Animals and ARTF Administration

Fifty male Sprague-Dawley (SD) rats (250–300g) were obtained from the Experimental Animal Center of Peking University Health Science Center (Beijing, China) and 40 of them were kept in metabolic cages (Type: DXL-DL, Suzhou Fengshi Laboratory Animal Equipment Co. Ltd. (Suzhou, Jiangsu, China)) with two rats in each cage, and the other 10 rats were kept in normal cages, for free water and food twice a day. All the animals were maintained in an environmentally controlled breeding room for two days. Then, the next two days, urine and feces samples were collected twice a day and combined as blank samples. After that, the 40 rats in metabolic cages were orally administered with ARTF at 200 mg/kg (the suspension of ARTF at 25 mg/kg was prepared with 0.5% CMC-Na in an ultrasonic bath), twice a day (8:00 am and 8:00 pm) for 31 days, and the 10 rats in normal cages weren’t administered anything except for water and food. The animal experiments were approved by the Biomedical Ethical Committee of Peking University (approval No. LA2016205).

3.3. Bio-Samples Collection and Pre-Treatment

3.3.1. Urine Collection and Pre-Treatment

Urine samples were collected twice a day (8:00 am and 8:00 pm) after administration of ARTF from metabolic cages and merged together, then filtered to remove impurities such as hair and dried in vacuum at 50 °C using a Heidolph Laborota 4001 rotatory evaporator (Heidolph Instruments GmhH & Co., Schwabach, Germany). After that, at a ratio of 1.0 g, dried samples were reconstituted in 10 mL methanol followed by 30 min ultrasonic extraction and filtered, then the filtrate was dried in vacuum (ca 25 g/day obtained) and 1.0 g sample was added 1 mL methanol to resuspended and stored at −20 °C. After 31 days, all the processed urine samples were mixed together as one sample and dried in vacuum. g). 1.0 g of the urine extract was taken out and redissolved in 5 mL methanol, then centrifuged at 15,000 rpm for 15 min and the supernatant was filtered through 0.45 μm nylon filter (Tianjin jinteng Experiment Co. Ltd., Tianjin, China). Finally, the filtrate was transferred into sample injection vial waiting for LC/MS analysis.

3.3.2. Feces Collection and Pre-Treatment

Feces samples were collected twice a day (8:00 am and 8:00 pm) from metabolic cages. Feces samples of each day were dried in 50 °C for 48 h and were crushed into powder. A pulverized sample of 1.0 g was extracted with 5 mL methanol for 30 min in an ultrasonic bath three times. Afterward, the filtrates were combined and concentrated to dryness, then redissolved in 10 folds methanol to store at −20 °C. All feces samples were combined after processing, and concentrated to dryness as ARTF-containing feces extract; 0.3 g was taken out to resuspended in 3 mL methanol and centrifuged at 15,000 rpm for 15 min, and filtered through 0.45 μm nylon filter before LC/MS analysis.

3.3.3. Plasma Collection and Pre-Treatment

At 32nd day, blood samples from rats in metabolic cages were collected by heart puncture technique under anesthesia at 10 min, 30 min, 1 h, 2 h, and 4 h (eight rats were sacrificed at each time point), and centrifuged at 5000 rpm for 15 min to obtain ARTF-containing plasma. Blank plasma was collected and processed in the same way from the rats in normal cages. All ARTF-containing and blank plasma were merged respectively, and 10 folds volume of methanol was added, and ultrasonically vibrated for 30 min to precipitate protein. Then, the mixture was centrifuged at 5000 rpm for 15 min, and the supernatant was condensed and dissolved in methanol, and the volume of methanol was 1% of the initial volume of plasma, and then centrifuged at 15,000 rpm for 15 min, finally filtered through 0.45 μm nylon filter before LC/MS analysis.

3.3.4. Organs Collection and Pre-Treatment

After blood collection, the heart, liver, spleen, lung, kidney, stomach, small intestine, colon, thymus, and brain were quickly removed and flushed clearly with stroke-physiological saline solution until there was no obvious blood or content in the surface or cavity. All the organs were stored at –80℃. All the organs were shredded and suspended in deionized water at a ratio of 1.0 g to 4 mL, then homogenized by ultrasound homogenizer (Ultra-Turrax T8, Ika-werke Gmbh & Co. KG, Staufen, Germany). After that, 8 mL homogenates were extracted with 10-folds volume methanol in an ultrasonic bath for 30 min. The mixture was centrifuged at 5000 rpm for 15 min, and the supernatant was separated, dried, and resuspended in 2 mL methanol and filtered through 0.45 μm nylon filter before LC/MS analysis.

3.4. Isolation and Identification of Metabolites from ARTF-Containing Urine

Isolation procedure: ARTF-containing urine extract obtained in Section 3.3 (ca. 750 g), was dissolved in 1.5 L deionized water, filtered, and then subjected to XAD-2 macroporous resins column chromatography. Water, 20% methanol-water, 60% methanol-water, and 100% methanol were used to elute the column and get Fraction 1 to Fraction 4, respectively. The four metabolites, MI-1 (M108) (2.67 mg), MI-2 (M32) (818.30 mg), MI-3 (16.66 mg), MI-4 (M106) (34.62 mg), were isolated and purified from Fraction-3 by ODS column chromatography, Sephadex LH-20 column chromatography, and a Shimadzu preparative HPLC system sequentially, and their purity was above 90% determined by an Agilent 1200 HPLC.
Structure identification using NMR: MI-1 and the other three metabolites were dissolved in 0.15 mL and 0.5 mL DMSO-d6, respectively. Their 1H, 13C, heteronuclear singular quantum correlation (HSQC), heteronuclear multiple bond correlation (HMBC). NMR spectra were recorded on a Bruker DRX-400 NMR spectrometer (Bruker, Rheinstetten, Germany), using tetramethylsilane (TMS) as internal standard. All chemical shifts were reported in parts per million (ppm, δ), and coupling constants (J) in Hertz. UV spectra (200–400 nm) and HRMS data were recorded on the LC-MS-IT-TOF instrument with a PDA detector.

3.5. Instruments and Conditions

HPLC-DAD-ESI-IT-TOF-MSn analyses were performed on a Shimadzu HPLC instrument (two LC-20AD pumps, an SIL-20AC autosampler, a CTO-20A column oven, an SPD-M20A PDA detector, a CBM-20A system controller) coupled with an IT-TOF mass spectrometer (Shimadzu, Kyoto, Japan) through an ESI interface. All data were processed by Shimadzu software, specifically, LCMS solution Version 3.60, Formula Predictor Version 1.2. and Accurate Mass Calculator. The chromatography separations were performed on an Industries Epic C18 column (250mm × 4.6 mm, 5 μm) (New Brunswick, NJ, USA) protected with an Agilent ZORBAX SB C18 column (12.5 mm × 4.6 mm, 5 μm) (Santa Clara, CA, USA). The mobile phase consisted of water-formic acid (100:0.1, v/v) (A) and acetonitrile (B) at a flow rate of 10,000 mL/min. A gradient elution program was adopted, specifically as 5% B at 0–10 min, 5–16% B at 10–12 min, 16–20% B at 12–25 min, 20–22% B at 25–45 min, 22–35% B at 45–60 min, 35–60% B at 60–85 min, 60–100% at 85–90 min. At the end of each run, 100% B was used to flush the column for 20 min. For mass detection, the mass spectrometer was programmed to carry out a full scan over m/z 100–1500 (MS1) with 30 ms accumulation time and m/z 50–1500 (MS2 and MS3) with 20 ms accumulation in both positive ion (PI) and negative ion (NI) detection mode; the flow rate was 0.2000 mL/min; the heat block and curved desolvation line temperature was 200 °C; the nebulizing nitrogen gas flow was 1.5 L/min; the interface voltage was (+), 4.5 kV; (−), 3.5 kV; the detector voltage was 1.7 kV; the relative collision-induced dissociation energy was 50%.

4. Conclusions

In summary, 170 kinds of compound (23 original constituents and 147 metabolites) were identified after administration of ARTF to rats, which included three newly detected original constituents and 89 new metabolites of ARTF, and 12 were regarded as new compounds (they are all metabolites) by retrieving information from the Sci inder database. Nineteen original constituents and 65 metabolites were detected and characterized in 10 organs. Four metabolites, including a new compound (calycoisn-3’-O-glucuronide methyl ester) and a first-isolated compound (astraisoflavan-7-O-glucoside-2’-O-glucuronide), along with two known compounds (daidzein-7-O-sulfate and calycosin-3’-O-glucuronide) were isolated from ARTF-containing urine and identified by NMR. Although the bioactivity studies of phase Ⅱ metabolites were little, 13 compounds (six original constituents, one phase I metabolite, six phase Ⅱ metabolites) in vivo were reported to possess similar pharmacological effects with ARTF, which indicated that they were effective forms of ARTF, and phase II metabolites might contribute to the efficacies of ARTF in vivo.
In the future, firstly, more kinds of phase I and phase II metabolites of ARTF should be obtained by synthesis or biotransformation. Then, the bioactivities of these metabolites should be determined to clarify the effective forms of ARTF. After that, the action mechanism of the effective forms can be studied. Finally, a new strategy to evaluate and control the quality of AR can be established.

Supplementary Materials

The following are available online. Detailed information on the determination of the contents of ARTF and its major constituents and the isolation procedure of compounds from urine, with Tables S1–S5 and Figures S1–S35 are available. References [39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69] are cited in the Supplementary Materials.

Author Contributions

Conceptualization, S.-Q.C. and F.X.; methodology, F.X., H.-F.L.; validation, S.-Q.C., F.X.; formal analysis, L.-J.L.; investigation, L.-J.L., H.-Y.W., Y.-F.Z.; writing—original draft preparation, L.-J.L; writing—review and editing, L.-J.L., H.-F.L., F.X., G.-X.L., M.-Y.S., X.W., S.-Q.C.; supervision, S-Q.C., F.X.; project administration, S-Q.C., F.X., L.-J.L.; funding acquisition, S.-Q.C., F.X. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China (NO. 81673595) and the National Science and Technology Major Project for Significant New Drugs Development (2019ZX09201004).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jing, L.; Zhao, Z.Z.; Chen, H.B. Review of Astragali Radix. Chin. Herb. Med. 2011, 3, 90–105. [Google Scholar]
  2. Wang, D.Q.; Zhuang, Y.; Tian, Y.P.; Thomas, G.N.; Ying, M.Z.; Tomlinson, B. Study of the effects of total flavonoids of Astragalus on atherosclerosis formation and potential mechanisms. Oxid. Med. Cell Longev. 2012, 2012, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Wang, D.Q.; Ding, B.G.; Tomas, G.N.; Tian, Y.P.; Wang, C.B.; Tomlinson, B. Effects of total flavonoids of astragalus on early atheroselerosis in rabbits with diet-induced hyPereholesterolemia. Chin. Pharmacol. Bull. 2003, 19, 637–639. [Google Scholar]
  4. Liu, Q.; Zhang, L.; Shan, Q.; Ding, Y.; Zhang, Z.; Zhu, M.; Mao, Y. Total flavonoids from Astragalus alleviate endothelial dysfunction by activating the Akt/eNOS pathway. J. Int. Med. Res. 2018, 46, 2096–2103. [Google Scholar] [CrossRef] [Green Version]
  5. Li, Q. Effects of Total Flavonoids of Astragalus on Inflammatory Response in Brain Tissue of Natural Aging Rats and Its Related Mechanism. J. Beihua Univ. (Nat. Sci.) 2018, 19, 745–749. [Google Scholar]
  6. Guo, Z.; Xu, H.Y.; Xu, L.; Wang, S.S.; Zhang, X.M. In Vivo and in Vitro Immunomodulatory and Anti-Inflammatory Effects of Total Flavonoids of Astragalus. Afr. J. Tradit. Complement. Altern. Med. 2016, 13, 60–73. [Google Scholar] [CrossRef] [Green Version]
  7. Li, J.; Xu, L.; Sang, R.; Yu, Y.; Ge, B.; Zhang, X. Immunomodulatory and anti-inflammatory effects of total flavonoids of Astragalus by regulating NF-κB and MAPK signalling pathways in RAW 264.7 macrophages. Pharmazie 2018, 73, 589–593. [Google Scholar]
  8. Wang, D.Q.; Ding, B.G.; Ma, Y.Q.; Zhao, H.L.; Thomas, G.N.; Tomlinson, B.; Tian, Y.P.; Wang, C.B.; Critchley, J.A. Studies on Protective Effect of Total Flavonoids of Astragalus on Liver Damage Paracetamol-Induced by. Chin. J. Chin. Mater. Med. 2001, 26, 483–486. [Google Scholar]
  9. Shen, W.M.; Wang, C.B.; Wang, D.Q.; Tian, Y.P.; Yan, G.T.; Hao, X.H. The protective effects of TFA on reperfusion induced hepatic injury in hemorrhagic shock. Chin. Pharmacol. Bull. 1997, 13, 532–534. [Google Scholar]
  10. Lee, E.J.; Yean, M.H.; Jung, H.S.; Kim, J.S.; Kang, S.S. Phytochemical Studies on Astragalus Root (2); Flavonoids and a Lignan. Nat. Prod. Sci. 2008, 14, 131–137. [Google Scholar]
  11. Yang, L.P.; Shen, J.G.; Xu, W.C.; Li, J.; Jiang, J.Q. Secondary metabolites of the genus Astragalus: Structure and biological-activity update. Chem. Biodivers. 2013, 10, 1004–1054. [Google Scholar] [CrossRef] [PubMed]
  12. Zhang, L.J.; Liu, H.K.; Hsiao, P.C.; Kuo, L.M.Y.; Lee, I.J.; Wu, T.S.; Chiou, W.F.; Kuo, Y.H. New Isoflavonoid Glycosides and Related Constituents from Astragali Radix (Astragalus membranaceus) and Their Inhibitory Activity on Nitric Oxide Production. J. Agr. Food Chem. 2011, 59, 1131–1137. [Google Scholar] [CrossRef] [PubMed]
  13. Yan, Z.; Lin, G.; Ye, Y.; Wang, Y.; Yan, R. A generic multiple reaction monitoring based approach for plant flavonoids profiling using a triple quadrupole linear ion trap mass spectrometry. J. Am. Soc. Mass. Spectr. 2014, 25, 955–965. [Google Scholar] [CrossRef]
  14. Zhang, Y.Z.; Xu, F.; Yang, D.H.; Shang, M.Y.; Yang, D.H.; Wang, X.; Cai, S.Q. Profiling and identification of the metabolites of calycosin in rat hepatic 9000× g supernatant incubation system and the metabolites of calycosin-7-O-β-D-glucoside in rat urine by HPLC-DAD-ESI-IT-TOF-MSn technique. J. Pharm. Biomed. Anal. 2012, 70, 425–439. [Google Scholar] [CrossRef] [PubMed]
  15. Zhang, Y.Z. Metabolic research of 8 isoflavone in Astragalus. Ph.D. Thesis, Beijing University, Beijing, China, 2011. [Google Scholar]
  16. Zhang, Y.Z.; Xu, F.; Dong, J.; Liang, J.; Hashi, Y.; Liu, G.X.; Li, Y.L.; Shang, M.Y.; Wang, X.; Cai, S.Q. Profiling the metabolites of astrapterocarpan in rat hepatic 9000g supernatant. Chin. J. Nat. Med. 2019, 17, 842–857. [Google Scholar] [CrossRef]
  17. Hu, G.; Siu, S.O.; Li, S.; Chu, I.K.; Kwan, Y.W.; Chan, S.W.; Leung, G.P.H.; Yan, R.; Lee, S.M.Y. Metabolism of calycosin, an isoflavone from Astragali Radix, in zebrafish larvae. Xenobiotica 2012, 42, 294–303. [Google Scholar] [CrossRef]
  18. Zhang, W.; Jiang, S.; Qian, D.W.; Shang, E.X.; Duan, J.A. Analysis of interaction property of calycosin-7-O-β-D-glucoside with human gut microbiota. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2014, 963, 16–23. [Google Scholar] [CrossRef]
  19. Davies, H.L.; Hill, J.L. The Effect of Diet on the Metabolism in Sheep of the Tritiated Isoflavones Formononetin and Biochanin-A. Aust. J. Agr. Res. 1989, 40, 157–163. [Google Scholar] [CrossRef]
  20. Tolleson, W.H.; Doerge, D.R.; Churchwell, M.I.; Marques, M.M.; Roberts, D.W. Metabolism of biochanin A and formononetin by human liver microsomes in vitro. J. Agr. Food Chem. 2002, 50, 4783–4790. [Google Scholar] [CrossRef]
  21. Zhang, W.; Jiang, S.; Qian, D.W.; Shang, E.X.; Guan, H.L.; Ren, H.; Zhu, Z.H.; Duan, J.A. The interaction between ononin and human intestinal bacteria. Acta Pharm. Sinica 2014, 49, 1162–1168. [Google Scholar]
  22. Li, H.F. Studies on the Metabolism of Astragali Radix In Vivo. Ph.D. Thesis, Shenyang Pharmaceutical University, Shenyang, China, 2017. [Google Scholar]
  23. Sun, J. Progress and Application of Astragalus and its classic prescription. Chin. Hosp. Pharm. J. 2019, 39, 1311–1314. [Google Scholar]
  24. Yang, P. Study on the In Vivo Metabolism of the Rhizomes of Smilax Glabra and Screening of Its Bioactive Components In Vivo. Ph.D. Thesis, Beijing University, Beijing, China, 2016. [Google Scholar]
  25. Liang, J.; Xu, F.; Zhang, Y.Z.; Zang, X.Y.; Wang, D.; Shang, M.Y.; Wang, X.; Chui, D.H.; Cai, S.Q. The profiling and identification of the metabolites of (+)-catechin and study on their distribution in rats by HPLC-DAD-ESI-IT-TOF-MS(n) technique. Biomed. Chromatogr. 2014, 28, 401–411. [Google Scholar] [CrossRef] [PubMed]
  26. Liang, J.; Xu, F.; Zhang, Y.Z.; Huang, S.; Zang, X.Y.; Zhao, X.; Zhang, L.; Shang, M.Y.; Yang, D.H.; Wang, X.; et al. The profiling and identification of the absorbed constituents and metabolites of Paeoniae Radix Rubra decoction in rat plasma and urine by the HPLC-DAD-ESI-IT-TOF-MS(n) technique: A novel strategy for the systematic screening and identification of absorbed constituents and metabolites from traditional Chinese medicines. J. Pharm. Biomed. Anal. 2013, 83, 108–121. [Google Scholar] [PubMed]
  27. Gao, X.; Mu, J.; Li, Q.; Guan, S.; Liu, R.; Du, Y.; Zhang, H.; Bi, K. Comprehensive Identification of Guan-Xin-Shu-Tong Capsule via a Mass Defect and Fragment Filtering Approach by High Resolution Mass Spectrometry: In Vitro and In Vivo Study. Molecules 2017, 22, 1007. [Google Scholar] [CrossRef]
  28. Zhao, X.; Yang, D.H.; Xu, F.; Huang, S.; Zhang, L.; Liu, G.X.; Cai, S.Q. The in vivo absorbed constituents and metabolites of Danshen decoction in rats identified by HPLC with electrospray ionization tandem ion trap and time-of-flight mass spectrometry. Biomed. Chromatogr. 2015, 29, 285–304. [Google Scholar] [CrossRef]
  29. Li, W.L.; Sun, X.M.; Song, H.; Ding, J.X.; Bai, J.; Chen, Q. HPLC/Q-TOF-MS-Based Identification of Absorbed Constituents and Their Metabolites in Rat Serum and Urine after Oral Administration of Cistanche deserticola Extract. J. Food Sci. 2015, 80, H2079–H2087. [Google Scholar] [CrossRef]
  30. Hamed, A.I.; Ben Said, R.; Kontek, B.; Al-Ayed, A.S.; Kowalczyk, M.; Moldoch, J.; Stochmal, A.; Olas, B. LC-ESI-MS/MS profile of phenolic and glucosinolate compounds in samh flour (Mesembryanthemum forsskalei Hochst. ex Boiss) and the inhibition of oxidative stress by these compounds in human plasma. Food Res. Int. 2016, 85, 282–290. [Google Scholar] [CrossRef]
  31. Ren, X.L. Study on the Effective Substances of Buyang Huanwu Decoction—Study on the Chemical Constituents of Buyang Huanwu Decoction in Pigs Urine. Master’s Thesis, Beijing University, Beijing, China, 2013. [Google Scholar]
  32. Fang, G.; Zhang, P.; Ye, X.L. Elecron spray ion trap mass spectrometry of isoflavones and isoflavone aglycones of Semen Sojae Praeparatum. Acad. J. Second Mil. Med. Univ. 2013, 34, 1108–1115. [Google Scholar] [CrossRef]
  33. Antignac, J.P.; Cariou, R.; Le Bizec, B.; Andre, F. New data regarding phytoestrogens content in bovine milk. Food Chem. 2004, 87, 275–281. [Google Scholar] [CrossRef]
  34. Murthy, M.S.R.; Rao, E.V.; Ward, R.S. Carbon-13 nuclear magnetic resonance spectra of isoflavones. Magn. Reson. Chem. 1986, 24, 225–230. [Google Scholar] [CrossRef]
  35. Nakano, H.; Ogura, K.; Takahashi, E.; Harada, T.; Nishiyama, T.; Muro, K.; Hiratsuka, A.; Kadota, S.; Watabe, T. Regioselective monosulfation and disulfation of the phytoestrogens daidzein and genistein by human liver sulfotransferases. Drug Metab. Pharmacol. 2004, 19, 216–226. [Google Scholar] [CrossRef] [PubMed]
  36. Zhang, Y.Z.; Xu, F.; Liang, J.; Tang, J.S.; Shang, M.Y.; Wang, X.; Cai, S.Q. Isoflavonoids from roots of Astragalus membranaceus var. mongholicus. Chin. J. Chin. Mater. Med. 2012, 37, 3243–3248. [Google Scholar]
  37. Knowlton, A.A.; Lee, A.R. Estrogen and the cardiovascular system. Pharmacol. Ther. 2012, 135, 54–70. [Google Scholar] [CrossRef] [Green Version]
  38. Straub, R.H. The complex role of estrogens in inflammation. Endocr. Rev. 2007, 28, 521–574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Bai, F.; Makino, T.; Kono, K.; Nagatsu, A.; Ono, T.; Mizukami, H. Calycosin and formononetin from astragalus root enhance dimethylarginine dimethylaminohydrolase 2 and nitric oxide synthase expressions in Madin Darby Canine Kidney II cells. J. Nat. Med. 2013, 67, 782–789. [Google Scholar] [CrossRef]
  40. Yu, D.H.; Bao, Y.M.; An, L.J.; Yang, M. Protection of PC12 Cells against Superoxide-induced Damage by Isoflavonoids from Astragalus mongholicus. Biomed. Environ. Sci. 2009, 22, 50–54. [Google Scholar] [CrossRef]
  41. Ma, R.; Yuan, F.; Wang, S.; Liu, Y.; Fan, T.; Wang, F. Calycosin alleviates cerulein-induced acute pancreatitis by inhibiting the inflammatory response and oxidative stress via the p38 MAPK and NF-kappaB signal pathways in mice. Biomed. Pharmacother. 2018, 105, 599–605. [Google Scholar] [CrossRef]
  42. Tian, J.; Wang, Y.; Zhang, X.; Ren, Q.Y.; Li, R.; Huang, Y.; Lu, H.L.; Chen, J. Calycosin inhibits the in vitro and in vivo growth of breast cancer cells through WDR7-7-GPR30 Signaling. J. Exp. Clin. Canc. Res. 2017, 36, 153–166. [Google Scholar] [CrossRef]
  43. Duan, X.; Meng, Q.; Wang, C.; Liu, Z.; Sun, H.; Huo, X.; Sun, P.; Ma, X.; Peng, J.; Liu, K. Effects of calycosin against high-fat diet-induced nonalcoholic fatty liver disease in mice. J. Gastroenterol Hepatol 2018, 33, 533–542. [Google Scholar] [CrossRef]
  44. Ren, M.; Wang, X.; Du, G.; Tian, J.; Liu, Y. Calycosin-7-O-β-D-glucoside attenuates ischemia-reperfusion injury in vivo via activation of the PI3K/Akt pathway. Mol. Med. Rep. 2015, 13, 633–640. [Google Scholar] [CrossRef] [Green Version]
  45. Fu, S.P.; Gu, Y.; Jiang, J.Q.; Chen, X.; Xu, M.J.; Chen, X.M.; Shen, J.G. Calycosin-7-O-beta-D-glucoside regulates nitric oxide/caveolin-1/matrix metalloproteinases pathway and protects blood-brain barrier integrity in experimental cerebral ischemia-reperfusion injury. J. Ethnopharmacol. 2014, 155, 692–701. [Google Scholar] [CrossRef] [PubMed]
  46. Choi, S.I.; Heo, T.R.; Min, B.H.; Cui, J.H.; Choi, B.H.; Park, S.R. Alleviation of osteoarthritis by calycosin-7-O-β-D-glucopyranoside (CG) isolated from Astragali radix (AR) in rabbit osteoarthritis (OA) model. Osteoarthritis Cartilage 2007, 15, 1086–1092. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Zhang, D.M. Effect of calycosin-7-O-beta-D-glucoside on cell apoptosis in cervical cancer HeLa cell and expression of Bcl-2/Bax. Chin. Tradit. Herbal Drugs 2015, 46, 1498–1502. [Google Scholar]
  48. Zhu, J.T.T.; Choi, R.C.Y.; Chu, G.K.Y.; Cheung, A.W.H.; Gao, Q.T.; Li, J.; Jiang, Z.Y.; Dong, T.T.X.; Tsim, K.W.K. Flavonoids possess neuroprotective effects on cultured pheochromocytoma PC12 cells: A comparison of different flavonoids in activating estrogenic effect and in preventing β-amyloid-induced cell death. J. Agr Food Chem. 2007, 55, 2438–2445. [Google Scholar] [CrossRef]
  49. Dong, L.; Yin, L.; Zhang, Y.; Fu, X.; Lu, J. Anti-inflammatory effects of ononin on lipopolysaccharide-stimulated RAW 264.7 cells. Mol. Immunol. 2017, 83, 46–51. [Google Scholar] [CrossRef]
  50. Jun, M.; Hong, J.; Jeong, W.S.; Ho, C.T. Suppression of arachidonic acid metabolism and nitric oxide formation by kudzu isoflavones in murine macrophages. Mol. Nutr. Food Res. 2005, 49, 1154–1159. [Google Scholar] [CrossRef]
  51. Jin, Y.M.; Xu, T.M.; Zhao, Y.H.; Wang, Y.C.; Cui, M.H. In vitro and in vivo anti-cancer activity of formononetin on human cervical cancer cell line HeLa. Tumour Biol. 2014, 35, 2279–2284. [Google Scholar] [CrossRef]
  52. Yang, S.; Wei, L.; Xia, R.; Liu, L.; Chen, Y.; Zhang, W.; Li, Q.; Feng, K.; Yu, M.; Zhang, W.; et al. Formononetin ameliorates cholestasis by regulating hepatic SIRT1 and PPARalpha. Biochem. Biophys. Res. Commun. 2019, 512, 770–778. [Google Scholar] [CrossRef]
  53. Applova, L.; Karlickova, J.; Riha, M.; Filipsky, T.; Macakova, K.; Spilkova, J.; Mladenka, P. The isoflavonoid tectorigenin has better antiplatelet potential than acetylsalicylic acid. Phytomedicine 2017, 35, 11–17. [Google Scholar] [CrossRef]
  54. Valeri, A.; Fiorenzani, P.; Rossi, R.; Aloisi, A.M.; Valoti, M.; Pessina, F. The soy phytoestrogens genistein and daidzein as neuroprotective agents against anoxia-glucopenia and reperfusion damage in rat urinary bladder. Pharmacol Res. 2012, 66, 309–316. [Google Scholar] [CrossRef]
  55. He, Y.; Wu, X.; Cao, Y.; Hou, Y.; Chen, H.; Wu, L.; Lu, L.; Zhu, W.; Gu, Y. Daidzein exerts anti-tumor activity against bladder cancer cells via inhibition of FGFR3 pathway. Neoplasma 2016, 63, 523–531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Wong, M.C.; Portmann, B.; Sherwood, R.; Niemela, O.; Koivisto, H.; Parkkila, S.; Trick, K.; L’Abbe, M.R.; Wilson, J.; Dash, P.R.; et al. The cytoprotective effect of alpha-tocopherol and daidzein against d-galactosamine-induced oxidative damage in the rat liver. Metabolism 2007, 56, 865–875. [Google Scholar] [CrossRef] [PubMed]
  57. Rajadurai, M.; Prince, P.S. Preventive effect of naringin on isoproterenol-induced cardiotoxicity in Wistar rats: An in vivo and in vitro study. Toxicology 2007, 232, 216–225. [Google Scholar] [CrossRef] [PubMed]
  58. Golechha, M.; Chaudhry, U.; Bhatia, J.; Saluja, D.; Arya, D.S. Naringin protects against kainic acid-induced status epilepticus in rats: Evidence for an antioxidant, anti-inflammatory and neuroprotective intervention. Biol. Pharm. Bull. 2011, 34, 360–365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Li, J.; Dong, Y.; Hao, G.; Wang, B.; Wang, J.; Liang, Y.; Liu, Y.; Zhen, E.; Feng, D.; Liang, G. Naringin suppresses the development of glioblastoma by inhibiting FAK activity. J. Drug Target. 2017, 25, 41–48. [Google Scholar] [CrossRef] [PubMed]
  60. Zhou, C.; Lai, Y.; Huang, P.; Xie, L.; Lin, H.; Zhou, Z.; Mo, C.; Deng, G.; Yan, W.; Gao, Z.; et al. Naringin attenuates alcoholic liver injury by reducing lipid accumulation and oxidative stress. Life Sci. 2019, 216, 305–312. [Google Scholar] [CrossRef]
  61. Guo, D.; Wang, J.; Wang, X.; Luo, H.; Zhang, H.; Cao, D.; Chen, L.; Huang, N. Double directional adjusting estrogenic effect of naringin from Rhizoma drynariae (Gusuibu). J. Ethnopharmacol. 2011, 138, 451–457. [Google Scholar] [CrossRef]
  62. Pugazhendhi, D.; Watson, K.A.; Mills, S.; Botting, N.; Pope, G.S.; Darbre, P.D. Effect of sulphation on the oestrogen agonist activity of the phytoestrogens genistein and daidzein in MCF-7 human breast cancer cells. J. Endocrinol. 2008, 197, 503–515. [Google Scholar] [CrossRef]
  63. Kgomotso, T.; Chiu, F.; Ng, K. Genistein- and daidzein 7-O-beta-D-glucuronic acid retain the ability to inhibit copper-mediated lipid oxidation of low density lipoprotein. Mol. Nutr. Food Res. 2008, 52, 1457–1466. [Google Scholar] [CrossRef]
  64. Morito, K.; Aomori, T.; Hirose, T.; Kinjo, J.; Hasegawa, J.; Ogawa, S.; Inoue, S.; Muramatsu, M.; Masamune, Y. Interaction of phytoestrogens with estrogen receptors α and β (II). Biol. Pharm. Bull. 2002, 25, 48–52. [Google Scholar] [CrossRef] [Green Version]
  65. Farina, H.G.; Pomies, M.; Alonso, D.F.; Gomez, D.E. Antitumor and antiangiogenic activity of soy isoflavone genistein in mouse models of melanoma and breast cancer. Oncol. Rep. 2006, 16, 885–891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Huang, Q.; Huang, R.; Zhang, S.; Lin, J.; Wei, L.; He, M.; Zhuo, L.; Lin, X. Protective effect of genistein isolated from Hydrocotyle sibthorpioides on hepatic injury and fibrosis induced by chronic alcohol in rats. Toxicol. Lett. 2013, 217, 102–110. [Google Scholar] [CrossRef] [PubMed]
  67. Schrader, C.; Ernst, I.M.; Sinnecker, H.; Soukup, S.T.; Kulling, S.E.; Rimbach, G. Genistein as a potential inducer of the anti-atherogenic enzyme paraoxonase-1: Studies in cultured hepatocytes in vitro and in rat liver in vivo. J. Cell Mol. Med. 2012, 16, 2331–2341. [Google Scholar] [CrossRef] [PubMed]
  68. Wan, Y.F.; XU, Y.J.; YUAN, B.; WANG, L.L.; Lin, L.Q.; Xu, H.Y. Synthesis of genistein sulfates and effects of genistein and its sulfates on proliferation of breast cancer cells. Shizhen Guoyi Guoyao 2011, 22, 284–286. [Google Scholar]
  69. Rimbach, G.; Weinberg, P.D.; de Pascual-Teresa, S.; Alonso, M.G.; Ewins, B.A.; Turner, R.; Minihane, A.M.; Botting, N.; Fairley, B.; Matsugo, S.; et al. Sulfation of genistein alters its antioxidant properties and its effect on platelet aggregation and monocyte and endothelial function. Biochim. Biophys. Acta. 2004, 1670, 229–237. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The extracted ion chromatograms (EICs) of original constituents (F1F17) in rat urine after administration of Astragali Radix total flavonoid (ARTF).
Figure 1. The extracted ion chromatograms (EICs) of original constituents (F1F17) in rat urine after administration of Astragali Radix total flavonoid (ARTF).
Molecules 25 05560 g001
Figure 2. The EICs of 106 metabolites in rat urine after administration of ARTF. (a) EICs of M2M41; (b) EICs of M43M93; (c) EICs of M96M144.
Figure 2. The EICs of 106 metabolites in rat urine after administration of ARTF. (a) EICs of M2M41; (b) EICs of M43M93; (c) EICs of M96M144.
Molecules 25 05560 g002
Figure 3. The structures of isolated metabolites from drug-containing urine. (a) MI-1 (M108); (b) MI-2 (M32); (c) MI-3; (d) MI-4 (M106).
Figure 3. The structures of isolated metabolites from drug-containing urine. (a) MI-1 (M108); (b) MI-2 (M32); (c) MI-3; (d) MI-4 (M106).
Molecules 25 05560 g003
Table 1. Original constituents in vivo after administration of ARTF to rats.
Table 1. Original constituents in vivo after administration of ARTF to rats.
No.tR
(min)
Formula (M)IonMeas. (m/z)Pred. (m/z)Diff (ppm)DBEIdentificationPlasmaUrineFeces
F1 58.602C16H12O5[M − H]283.0623283.06123.8911Calycosin
F277.588C16H12O5[M + H]+285.0753285.0758−1.7511Calycosin isomer 1
F3 *56.485C16H12O5[M − H]283.0608283.0612−1.4111Calycosin isomer 2
F4 27.200C22H22O10[M + HCOO]491.1215491.11954.0712Calycosin-7-O-glucoside
F5 47.743C22H22O9[M + H]+431.1322431.1337−3.4812Ononin
F6 71.750C16H12O4[M − H]267.0673267.06633.7411Formononetin
F7 *66.802C16H12O6[M − H]299.0569299.05612.6811Pratensein/Rhamnocitrin/5,7,4’-trihydroxy-3’-methoxyisoflavone
F8 *68.197C16H12O6[M − H]299.0576299.05615.0211Pratensein/Rhamnocitrin/5,7,4’-trihydroxy-3’-methoxyisoflavone
F965.018C15H12O5[M − H]271.0622271.06123.6910Trihydroxy-dihydroisoflavone/flavone
F1044.750C15H14O5[M − H]273.0777273.07683.309Trihydroxy-tetrahydroisoflavone/flavone
F1154.377C15H10O4[M − H]253.0514253.05063.1611Daidzein
F1255.485C15H12O4[M − H]255.0675255.06634.7010Trihydroxychalcone
F1365.627C15H10O5[M − H]269.0452269.0455−1.1211Trihydroxyisoflavone/flavone
F1460.827C17H14O6[M + H]+315.0882315.08636.0311Odoratin isomer
F1557.460C17H18O5[M + H]+303.1216303.1227−3.639Astraisoflavane isomer
F1665.852C17H16O6[M + H]+317.1040317.10206.3110Dihydroxy dimethoxyisoflavone/flavone
F17 73.328C17H16O5[M + H]+301.1052301.1071−6.3110Astraptercarpan
F1835.440C27H32O14[M − H]579.1748579.17195.0112Naringin
F1934.635C28H36O13[M − H]579.2103579.20833.4511Dihydrocalycosin pentose glucoside
F2051.837C28H34O14[M − H]593.1899593.18763.8812Astrapterocarpan pentose glucoside
F2163.302C16H14O5[M + H]+287.0927287.09144.53103,10-dihydroxy-9-methoxypterocarpan
F2260.812C22H22O11[M − H]461.1114461.10895.4212Pratensein glucoside/5,7’,4’-trihydroxy-3’-methoxyisoflavone glucoside
F2322.332C22H22O11[M − H]461.1112461.10894.9912Pratensein glucoside/5,7’,4’-trihydroxy-3’-methoxyisoflavone glucoside
Sum3174
Note: tR: Retention time; Meas.: measured; Pred.: predicted; Diff: difference; DBE: double bone equivalents. These constituents were identified by comparison with reference compounds; * New original constituents found in vivo after administration of ARTF. ▲ Detected.
Table 2. Metabolites in vivo after administration of ARTF to rats.
Table 2. Metabolites in vivo after administration of ARTF to rats.
NO.tR(min)Formula (M)IonMeas.
(m/z)
Pred.
(m/z)
Diff (ppm)DBEIdentificationUrinePlasmaFeces
M1 19.500C7H6O5S[M − H]200.9862200.9863−0.505Hydroxyl-benzaladehyde sulfate or isomer
M2 10.948C7H8O6S[M − H]218.9967218.9969−0.914Methyl pyrogallol sulfate or isomer
M3 20.550C8H8O6S[M − H]230.9961230.9969−3.465Hydroxyphenylacetic acid sulfate
M4 28.350C8H8O6S[M − H230.9963230.9969−2.65Hydroxyphenylacetic acid sulfate isomer
M5 21.008C9H8O6S[M − H]242.9978242.99693.76Hydroxycinnamic acid sulfate 1
M6 23.533C9H8O6S[M − H]242.9960242.9969−3.706Hydroxycinnamic acid sulfate 2
M7 55.018C10H12O5S[M − H]243.0345243.03334.945Eugenol sulfate
M8 36.100C8H8O7S[M − H]246.9921246.99181.215Vanillic acid sulfate
M9 19.617C9H12O6S[M − H]247.0281247.0282−0.404Homovanillyl alcohol sulfate
M10 20.383C10H10O7S[M − H]273.0080273.00742.206Ferulic Acid sulfate
M11 58.818C11H14O6S[M − H]273.0436273.0438−0.735Methoxyeugenol sulfate 1
M12 55.593C11H14O6S[M − H]273.0433273.0438−1.835Methoxyeugenol sulfate 2
M13 43.617C11H12O7S[M − H]287.0237287.02312.096C11H12O4 sulfate
M14 23.750C11H14O7S[M − H]289.0393289.03872.085Ethylhomovanillic acid sulfate 1
M15 26.642C11H14O7S[M − H]289.0397289.03873.465Ethylhomovanillic acid sulfate 2
M16 27.558C11H12O8 S[M − H]303.0173303.018−2.3163’,5’-dimethoxy-4’-hydroxycinnamic acid sulfate
M1780.230C16H12O5[M − H]283.0604283.0612−2.8311Calycosin isomer 1
M18 65.833C16H12O5[M + H]+285.0767285.07583.1611Calycosin isomer 2
M19 31.508C16H12O5[M + H]+285.0738285.0758−7.0211Calycosin isomer 3
M20 79.592C16H12O5[M + H]+285.0777285.07586.6611Calycosin isomer 4
M2151.610C16H12O6[M + H]+301.0688301.0707−6.3111Hydroxycalycosin
M2254.902C17H14O6[M + H]+315.0883315.08636.3511Methoxycalycosin 1
M2334.517C17H14O6[M + H]+315.0836315.0863−8.5711Methoxycalycosin 2
M2461.477C17H14O6[M + H]+315.0840315.0863−7.311Methoxycalycosin 3
M2531.483C16H12O8S[M − H]-363.0178363.0180−0.5511Calycosin sulfate 1
M2647.512C16H12O8S[M − H]363.018363.0180011Calycosin sulfate 2
M2751.168C16H12O8S[M − H]363.0187363.01801.9311Calycosin sulfate isomer 1
M28 67.248C16H12O8S[M − H]363.0174363.0180−1.6511Calycosin sulfate isomer 2
M29 44.867C16H12O8S[M − H]363.0189363.01802.4811Calycosin sulfate isomer 3
M30 79.697C16H12O8S[M − H]363.0193363.01803.5811Calycosin sulfate isomer 4
M31 33.158C16H12O11S2[M − H]442.9767442.97484.2911Calycosin-7,3’-O-disulfate
M32 36.133C22H20O11[M − H]459.0952459.09334.1413Calycosin-3’-O-glucuronide
M33 23.417C22H20O14S[M − H]539.0538539.05016.8613Calycosin glucuronide sulfate 1
M34 31.245C22H20O14S[M − H]539.0519539.05013.3413Calycosin glucuronide sulfate 2
M3571.298C16H12O9S[M − H]379.0137379.01292.1111Hydroxycalycosin sulfate 1
M3657.743C16H12O9S[M − H]-379.0153379.01296.3311Hydroxycalycosin sulfate 2
M3761.243C16H12O9S[M − H]379.0151379.01295.8011Hydroxycalycosin sulfate 3
M38 26.125C22H20O12[M − H]475.0847475.0882−7.3713Hydroxycalycosin glucuronide 1
M39 51.610C22H20O12[M − H]475.0918475.08827.5813Hydroxycalycosin glucuronide 2
M40 39.042C23H22O12[M − H]489.1063489.10385.1113Methoxycalycosin glucuronide
M41 33.123C23H22O15S[M − H]569.0634569.06074.7413Methoxycalycosin glucuronide sulfate
M42 20.108C28H30O16[M − H]621.1451621.1461−1.6114Calycosin-7-O-glucoside-3’-O-glucuronide
M4348.258C16H14O8S[M − H]365.0341365.03371.110Dihydrocalycosin sulfate 1
M4443.285C16H14O8S[M − H]365.0346365.03372.4710Dihydrocalycosin sulfate 2
M4545.652C16H14O8S[M − H]365.0356365.03375.210Dihydrocalycosin sulfate 3
M46 38.098C22H22O11[M − H]461.1111461.10894.7712Dihydrocalycosin glucuronide
M4757.285C16H14O9S[M − H]381.0282381.0286−1.0510Hydroxy dihydrocalycosin sulfate 1
M4893.980C16H14O9S[M − H]381.0296381.02862.6210Hydroxy dihydrocalycosin sulfate 2
M4949.422C16H14O9S[M − H]381.0300381.02863.6710Hydroxy dihydrocalycosin sulfate 3
M5039.792C18H18O8S[M − H]393.0665393.0653.8210Dimethyl dihydrocalycosin sulfate
M51 52.118C17H16O9S[M − H]395.0434395.0442−2.0310Methoxy dihydrocalycosin sulfate 1
M52 68.430C17H16O9S[M − H]395.0457395.04423.8010Methoxy dihydrocalycosin sulfate 2
M5366.472C16H14O10S[M − H]397.0251397.02354.0310Dihydroxyl dihydrocalycosin sulfate
M5425.252C16H16O8S[M − H]367.0477367.0493−4.369Tetrahydrocalycosin sulfate 1
M5519.342C16H16O8S[M − H]367.0496367.04930.829Tetrahydrocalycosin sulfate 2
M5621.443C16H16O8S[M − H]367.0500367.04931.919Tetrahydrocalycosin sulfate 3
M5750.593C16H16O8S[M − H]367.0516367.04936.279Tetrahydrocalycosin sulfate 4
M5867.630C16H16O8S[M − H]367.0517367.04936.549Tetrahydrocalycosin sulfate 5
M5957.860C16H16O8S[M − H]367.0518367.04936.819Tetrahydrocalycosin sulfate 6
M60 34.182C22H24O11[M − H]463.1257463.12462.3811Tetrahydrocalycosin glucuronide 1
M61 54.027C22H24O11[M − H]463.1269463.12464.9711Tetrahydrocalycosin glucuronide 2
M62 △,26.975C22H24O14S[M − H]543.0792543.0814−4.0511Tetrahydrocalycosin glucuronide sulfate
M63 △,32.350C28H32O17[M − H]639.1607639.15676.2613Tetrahydrocalycosin diglucuronide
M6448.375C16H16O9S[M − H]383.0449383.04421.839Hydroxy tetrahydrocalycosin sulfate 1
M6525.200C16H16O9S[M − H]383.046383.04424.709Hydroxy tetrahydrocalycosin sulfate 2
M6664.843C16H16O9S[M − H]383.0462383.04425.229Hydroxy tetrahydrocalycosin sulfate 3
M6740.192C16H16O9S[M − H]383.0467383.04426.539Hydroxy tetrahydrocalycosin sulfate 4
M6830.608C16H16O9S[M − H]383.0444383.04420.529Hydroxy tetrahydrocalycosin sulfate 5
M69 △,27.617C16H16O12S2[M − H]463.0031463.00104.759Hydroxy tetrahydrocalycosin disulfate
M70 △,45.733C18H20O10S[M − H]427.0716427.07042.819Dihydroxy dihydrocalycosin sulfate
M71 56.593C24H28O12[M − H]507.1535507.15085.3211Dimethyl hydroxy tetrahydrocalycosin glucuronide 1
M72 54.727C24H28O12[M − H]507.1539507.15086.1111Dimethyl hydroxy tetrahydrocalycosin glucuronide 2
M73 71.300C16H12O7S[M − H]347.0228347.0231−0.8611Formononetin-7-O-sulfate
M74 49.357C22H20O10[M − H]-443.0977443.0984−1.5813Formononetin-7-O-glucuronide
M7521.960C16H16O7S[M − H]351.0537351.0544−1.999Tetrahydroformononetin sulfate 1
M7659.352C16H16O7S[M − H]351.0554351.05442.859Tetrahydroformononetin sulfate 2
M7759.860C16H16O7S[M − H]351.0564351.05445.709Tetrahydroformononetin sulfate 3
M78 49.257C22H24O10[M − H]447.1322447.12975.5911Tetrahydroformononetin glucuronide
M79 69.797C17H16O8S[M − H]379.0509379.04934.2210Astrapterocarpan-3-O-sulfate
M80 56.043C23H24O11[M − H]475.1276475.12466.3112Astrapterocarpan-3-O-glucuronide
M8131.018C18H18O6[M − H]329.1027329.1031−1.2210Methoxyastrapterocarpan
M82 53.593C23H24O12[M − H]491.1225491.11956.1112Hydroxyastrapterocarpan glucuronide 1
M83 40.915C23H24O12[M − H]491.1194491.1195−0.2012Hydroxyastrapterocarpan glucuronide 2
M84 43.618C23H24O12[M − H]491.1173491.1195−4.4812Hydroxyastrapterocarpan glucuronide 3
M85 35.667C17H18O5[M +H]+303.1205303.1227−7.269Astraisoflavan isomer
M86 32.868C18H20O5[M − H]315.1228315.1238−3.179Methoxyastraisoflavan
M87 37.802C18H20O5[M − H]315.1235315.1238−0.959Methoxyastraisoflavan isomer
M88 34.368C19H22O6[M − H]-345.1360345.13444.649Hydroxy dimethoxyastraisoflavan
M8959.918C17H18O8S[M − H]381.0639381.0650−2.899Astraisoflavan-7-O-sulfate
M9062.510C17H18O8S[M − H]381.0660381.06502.629Astraisoflavan-2’-O-sulfate
M9134.427C17H18O8S[M − H]381.0662381.06503.159Astraisoflavan sulfate isomer
M92 34.993C18H20O8S[M − H]395.0795395.0806−2.789Methyoxyastraisoflavan sulfate 1
M93 30.460C18H20O8S[M − H]395.0819395.08063.299Methyoxyastraisoflavan sulfate 2
M9420.252C17H18O9S[M − H]397.0602397.05990.769Hydroxyastraisoflavan sulfate 1
M9549.543C17H18O9S[M − H]397.0603397.05991.019Hydroxyastraisoflavan sulfate 2
M9644.008C17H18O9S[M − H]397.0608397.05992.279Hydroxyastraisoflavan sulfate 3
M97 51.435C17H18O9S[M − H]397.0614397.05993.789Hydroxyastraisoflavan sulfate 4
M98 68.372C17H18O9S[M − H]397.0620397.05995.299Hydroxyastraisoflavan sulfate 5
M99 18.817C17H18O9S[M − H]397.0622397.05995.799Hydroxyastraisoflavan sulfate 6
M100 62.393C18H20O9S[M − H]411.0743411.0755−2.929Methoxyastraisoflavan sulfate 1
M101 60.210C18H20O9S[M − H]411.0758411.07550.739Methoxyastraisoflavan sulfate 2
M102 58.702C23H26O11[M − H]477.1429477.14025.6611Astraisoflavan-7-O-glucuronide
M103 56.868C23H26O11[M − H]477.1430477.14025.8711Astraisoflavan-2’-O-glucuronide
M104 △,38.250C23H26O14S[M − H]557.0997557.09704.8511Astraisoflavan glucuronide sulfate 1
M105 △,52.510C23H26O14S[M − H]557.1001557.09705.5611Astraisoflavan glucuronide sulfate 2
M106 35.633C29H36O16[M − H]639.1949639.19312.8212Astraisoflavan-7-O-glucoside-2’-O-glucuronide
M10741.557C15H10O7S[M − H]333.0074333.0074011Daidzein-4’-O-sulfate
M10828.925C15H10O7S[M − H]333.0080333.00741.8011Daidzein-7-O-sulfate
M109 28.400C15H10O10S2[M − H]412.9658412.96433.6311Daidzein-7,4’-O-disulfate
M110 23.817C21H18O10[M − H]429.0835429.08271.8613Daidzein glucuronide
M111 20.167C21H18O13S[M − H]509.0403509.03951.5713Daidzein glucuronide sulfate
M112 44.292C15H12O7S[M − H]335.0212335.0231−5.6710Dihydrodaidzein sulfate
M11353.852C15H14O7S[M − H]337.0389337.03870.599Tetrahydrodaidzein sulfate 1
M11439.333C15H14O7S[M − H]337.0400337.03873.869Tetrahydrodaidzein sulfate 2
M11557.002C15H14O7S[M − H]337.0402337.03874.459Tetrahydrodaidzein sulfate 3
M11660.560C15H14O7S[M − H]337.0409337.03876.539Tetrahydrodaidzein sulfate 4
M117 34.458C15H14O10S2[M − H]416.9988416.99567.679Tetrahydrodaidzein disulfate
M11840.017C15H10O5[M + H]+271.0617271.06015.9011Gensitein
M11941.833C15H10O8S[M − H]349.0033349.00242.5811Genistein sulfate 1
M12057.918C15H10O8S[M − H]349.0039349.00244.3011Genistein sulfate 2
M12137.233C15H10O8S[M − H]349.0030349.00241.7211Genistein sulfate 3
M122 25.042C21H18O14S[M − H]525.0367525.0370−0.579Genistein glucuronide sulfate
M12333.658C15H12O8S[M − H]351.0180351.0180010Dihydrogenistein sulfate 1
M124 74.098C15H12O8S[M − H]351.0184351.01801.1410Dihydrogenistein sulfate 2
M125 51.688C15H12O8S[M − H]351.0189351.0182.5610Dihydrogenistein sulfate 3
M12644.758C15H14O5[M − H]273.0757273.0768−4.039Tetrahydrogenistein
M12763.985C15H14O8S[M − H]353.0359353.03376.239Tetrahydrogenistein sulfate 1
M12865.410C15H14O8S[M − H]353.0360353.03376.519Tetrahydrogenistein sulfate 2
M129 △,49.090C20H22O12S[M − H]485.0787485.07595.7710Tetrahydrogenistein pentose sulfate
M130 △,39.850C21H24O13S[M − H]515.0911515.08658.9310Tetrahydrogenistin sulfate
M13162.235C15H14O6S[M − H]321.0439321.04380.319Equol sulfate isomer
M13256.152C15H14O6S[M − H]321.0444321.04381.879Equol sulfate 1
M13358.943C15H14O6S[M − H]321.0454321.04384.989Equol sulfate 2
M134 46.017C21H22O9[M − H]417.1185417.1191−1.4411Equol glucuronide 1
M135 47.975C21H22O9[M − H]417.1209417.11914.3211Equol glucuronide 2
M136 △,32.293C21H22O12S[M − H]497.0786497.07595.4311Equol glucuronide sulfate 1
M137 △,28.058C21H22O12S[M − H]497.0797497.07597.6411Equol glucuronide sulfate 2
M138 37.317C21H22O12S[M − H]497.0757497.0759-0.411Equol glucuronide sulfate 3
M139 57.977C15H16O6S[M − H]323.0616323.05956.58Dihydroequol sulfate
M14028.817C16H14O6[M + H]+303.0878303.08634.9510Dihydropratensein
M141 24.925C22H20O15S[M − H]555.0487555.04506.6713Pratensein glucuronide sulfate
M142 △,26.300C27H30O17[M − H]625.1462625.14108.3213Tetrahydro trihydroxyisoflavone diglucuronide 1
M143 △,26.867C27H30O17[M − H]625.1467625.14109.1213Tetrahydro trihydroxyisoflavone diglucuronide 2
M144 41.650C15H14O9S[M − H]369.0303369.02864.619Tetrahydro-tetrahydroxyisoflavone sulfate
M145 41.930C22H20O15S[M − H]555.0478555.04506.7613Pratensein glucuronide sulfate
M146 61.012C22H20O11[M − H]459.0925459.0933−1.2313Calycosin-7-O-glucuronide
M147 69.988C17H18O8S[M − H]381.0652381.06500.529Astraisoflavan sulfate isomer
Sum1066417
Note: tR: Retention time; Meas.: measured; Pred.: predicted; Diff: difference; DBE: double bone equivalents. New metabolites found in vivo after administration of ARTF; Potential New compound by retrieving information from SciFinder database. ▲ Detected.

Sample Availability

Samples of the compounds are not available from the authors.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Liu, L.-J.; Li, H.-F.; Xu, F.; Wang, H.-Y.; Zhang, Y.-F.; Liu, G.-X.; Shang, M.-Y.; Wang, X.; Cai, S.-Q. Exploring the In Vivo Existence Forms (23 Original Constituents and 147 Metabolites) of Astragali Radix Total Flavonoids and Their Distributions in Rats Using HPLC-DAD-ESI-IT-TOF-MSn. Molecules 2020, 25, 5560. https://doi.org/10.3390/molecules25235560

AMA Style

Liu L-J, Li H-F, Xu F, Wang H-Y, Zhang Y-F, Liu G-X, Shang M-Y, Wang X, Cai S-Q. Exploring the In Vivo Existence Forms (23 Original Constituents and 147 Metabolites) of Astragali Radix Total Flavonoids and Their Distributions in Rats Using HPLC-DAD-ESI-IT-TOF-MSn. Molecules. 2020; 25(23):5560. https://doi.org/10.3390/molecules25235560

Chicago/Turabian Style

Liu, Li-Jia, Hong-Fu Li, Feng Xu, Hong-Yan Wang, Yi-Fan Zhang, Guang-Xue Liu, Ming-Ying Shang, Xuan Wang, and Shao-Qing Cai. 2020. "Exploring the In Vivo Existence Forms (23 Original Constituents and 147 Metabolites) of Astragali Radix Total Flavonoids and Their Distributions in Rats Using HPLC-DAD-ESI-IT-TOF-MSn" Molecules 25, no. 23: 5560. https://doi.org/10.3390/molecules25235560

Article Metrics

Back to TopTop