Skip to main content

Identifying the active components of Baihe–Zhimu decoction that ameliorate depressive disease by an effective integrated strategy: a systemic pharmacokinetics study combined with classical depression model tests

A Correction to this article was published on 02 April 2020

This article has been updated

Abstract

Background

Modern pharmacological studies have demonstrated that Baihe–Zhimu decoction (BZD) has antidepressant effects. However, the complex composition and lack of clear evaluation standards for BZD make it less likely to be understood and accepted than evidence-based active natural compounds.

Methods

In this study, an effective method for the identification of antidepressant components was demonstrated and applied to BZD. The first step was to evaluate the efficacy of BZD by the forced swimming test (FST) and the tail suspension test (TST), followed by successive quantitative analyses of the absorbed constituents at different stages, such as before hepatic disposition, liver distribution, after hepatic disposition and brain distribution after the oral administration of BZD. Finally, the compounds detected in the brain were confirmed by activity testing.

Results

Our investigation observed that timosaponin BII and timosaponin BIII were accurately determined in the brain after oral administration of BZD, and they were further confirmed to reduce the immobility time in the FST and TST. As described above, timosaponin BII and timosaponin BIII were used to scientifically and reasonably explain the effective chemical basis of the effect of BZD on depression.

Conclusions

This research affords an effective method to discover lead molecules for antidepressants from traditional Chinese medicine.

Background

Many Chinese medicine formulae have been used for the treatment of diseases in China and are also regarded as alternative therapeutic agents in other Asian countries [1, 2]. However, the chemical constituents in Chinese medicine formulae are so complex and diverse that some of them may be effective and others may not throughout the course of therapy. Therefore, the identification of the main effective components from Chinese medicine formulae will benefit the discovery of lead compounds and the optimization of new drug development. At present, the conventional phytochemical approach remains the main method for the discovery of effective components or effective component groups. Although they exhibit various bioactivities in vivo, some components may display extremely low bioavailability [3, 4]. Thus, they cannot be regarded as the main effective components of Chinese medicine formulae, because efficacy is considered to rely on bioactive compounds with sufficient exposure in the plasma or target organs [5, 6]. Hence, it is necessary to develop a strategy for screening bioactive components with high exposure from Chinese medicine formulae.

Our group used an effective method combining classical tests for specific activity and pharmacokinetics studies based on high-performance liquid chromatography coupled to triple quadrupole mass spectrometry (HPLC-QQQ MS) to screen for the main effective compounds in traditional Chinese medicine (TCM) [7,8,9]. The first step was to evaluate the efficacy of crude extracts and perform quantitative analysis of their major chemical constituents. Then, the absorbed constituents were quantitatively analyzed in successive stages in vivo after the oral administration of crude extracts, such as before hepatic disposition (in the portal vein plasma), during liver distribution, after hepatic disposition (in the systemic plasma) and during target organ distribution. Finally, the compounds detected in the target organ were confirmed by activity testing and further mechanistic research.

Baihe–Zhimu decoction (BZD), consisting of two herbs, Baihe (Lilium brownii var. viridulum) and Zhimu (Rhizoma anemarrhenae), is a traditional prescription to treat lily disease, which exhibits similar symptoms to depression. Modern pharmacological studies have demonstrated that BZD has antidepressant effects in animal models [10, 11]. However, the complex composition of BZD and the lack of clear evaluation standards make its use less likely to be understood and accepted than that of evidence-based active natural compounds. Therefore, we have previously characterized a total of 39 compounds in the BZD to understand its chemical basis [12]. Among them, flavonoids and steroid saponins are considered the most important bioactive constituents [13,14,15]. Moreover, we found that the levels of mangiferin, neomangiferin, timosaponin BII, timosaponin BIII and timosaponin AIII in the portal vein plasma and systemic plasma were all above the detection limit of HPLC-QQQ MS [16], which could contribute to the in vivo process of BZD.

In this study, we tried to discover the effective components of BZD for the treatment of depression using the strategy described above. After evaluation of the antidepressant activity of BZD, each individual herb and the corresponding fractions, the major components in BZD were quantified by HPLC-QQQ MS. Then, the absorbed constituents were determined in several stages after oral administration of BZD. The brain is the main target organ for mental disease, but little information related to brain disposition after oral administration of BZD in rats was available. Therefore, the pharmacokinetic behavior in the brain was finally tested, and the antidepressant activity of the detected components in the brain was validated. A flowchart of this study is shown in Fig. 1. This research provided an effective method for the discovery of the main effective components in Chinese medicine formulae.

Fig. 1
figure 1

Procedure for the discovery of antidepressant components in BZD

Methods

Materials and reagents

Baihe (Lilium brownii var. viridulum) and Zhimu (Rhizoma anemarrhenae) were purchased from Shanghai Kangqiao Chinese Medicine Tablet Co., Ltd. (Shanghai China) and authenticated by Dr. Zhixiong Li (Shanghai Research Center for Modernization of TCM, Shanghai Institute of Materia Medica, Shanghai, China). Mangiferin, neomangiferin, timosaponin BII, timosaponin BIII and timosaponin AIII (purity > 98%) were supplied by Chengdu Biopurify Phytochemicals Co., Ltd. (Chengdu, China), and fluoxetine hydrochloride was provided by Eli Lilly & Co., Ltd. (Indianapolis, USA). HPLC-grade agents, including methanol, acetonitrile and formic acid, were obtained from Merck & Co., Inc. (Darmstadt, Germany). All other chemicals were purchased from Sinopharm Chemical Reagent Co., Ltd. (Shanghai, China) and were of analytical grade. The deionized water was purified by a Milli-Q system (Millipore, Billerica, MA, USA).

Preparation of Baihe–Zhimu decoction, individual herbs and related fractions

Baihe (300 g) and Zhimu (150 g) were cut into slices and mixed together at a weight ratio of 2:1. Then, the mixture (450 g) was extracted twice with boiling water (4500 mL) for 2 h each time. After filtration, the supernatant was condensed to 300 mL under reduced pressure to obtain Baihe–Zhimu decoction (BZD). The chemical characteristics of BZD were further investigated by HPLC-QQQ MS to ensure the chemical consistency of the tested sample. Five major components, including timosaponin BII, timosaponin BIII, mangiferin, neomangiferin and timosaponin AIII, were identified by comparing the retention times with those of the chemical standards, and their concentrations were determined to be 8979.80, 4191.10, 2649.02, 1624.27 and 442.91 μg/g BZD, respectively. In addition, each herb in BZD was individually extracted with boiling water using the same method as motioned above. The prepared Zhimu decoction (ZD) was fractionated by microporous resin to obtain three fractions, including polysaccharides of Zhimu (PZ, elution with water), xanthones of Zhimu (XZ, elution with 20% EtOH) and saponins of Zhimu (SZ, elution with 60% EtOH).

HPLC-QQQ MS conditions

The HPLC-QQQ MS method was developed according to a previous study in our group [16]. Briefly, five compounds were simultaneously determined in plasma, liver and brain samples using a 1260 Series liquid chromatography system (Agilent Technologies, Palo Alto, CA, USA) coupled to a 6460 triple quadrupole mass spectrometer with an electrospray ionization source (Agilent Technologies, Palo Alto, CA, USA). Chromatographic separation was performed on an ACE Super C18 column (100 mm × 2.1 mm, 3.0 µm, Advanced Chromatography Technologies Ltd. Aberdeen, Scotland) at a temperature of 40 °C. The mobile phase consisted of water containing 0.1% formic acid (A) and acetonitrile containing 0.1% formic acid (B). The gradient program was performed as follows: 0–3 min, 92% A; 3.5–4.0 min, 88–60% A; 5.5–6 min, 60–55% A; 6.6–7 min, 55–5% A; 11–11.01 min, 5–92% A; 11.01–13.5 min, 92% A. The flow rate was set to 0.35 mL/min in the time range of 0–6.6 min and kept at 0.45 mL/min between 7 and 13.5 min. Five components were monitored in negative multiple reaction monitoring (MRM) mode. The parameters of the mass spectrometer were as follows: capillary voltage, 3500 V; nebulizer, 45 psi; gas temperature, 350 °C; gas flow rate, 12 L/min; sheath gas temperature, 400 °C; sheath gas flow rate, 8 L/min. MassHunter Workstation software (Agilent Technologies, Palo Alto, CA, USA) was used for system operation and data analysis.

Animals and treatments

All Sprague–Dawley (SD) rats and ICR mice were purchased from Shanghai SLAC Laboratory Animal Co., Ltd. and kept in the breeding room at 22 ± 2 °C and 50 ± 10% humidity in Shanghai Institute of Materia Medica (SIMM). The experimental protocols were approved by the Institutional Animal Care and Use Committee in SIMM. After 1 week of acclimation, all the SD rats were orally administered BZD (15 g/kg). The biological samples were collected from the rats after fasting for 12 h. After anaesthetization, the rats were dissected at 5 min, 15 min, 30 min, 1 h, 2 h, 4 h, 7 h, 10 h, 20 h and 40 h (n = 5 for each time-point). The hepatic portal vein plasma (2 mL) and the systemic plasma (6–8 mL) were subsequently harvested, and finally the liver and brain tissue samples were collected. The plasma sample were centrifuged at 9500×g for 5 min, and the tissue samples were washed with saline solution. All samples were stored at − 80 °C until analysis. The samples were prepared according to the method described in the previous study by our group [16].

The antidepressant activity of the drugs was evaluated by classic behavioral measurements, such as the forced swimming test (FST) and tail suspension test (TST). All the ICR mice were randomly divided into several groups (n = 8) as follows: a control group with stimulation and different test groups subjected to FST and TST after the oral administration of different drugs for 8 days, including fluoxetine (10 mg/kg), BZD (3 g/kg), ZD (3 g/kg), Baihe decoction (BD, 3 g/kg), PZ (21.2 mg/kg), XZ (21.2 mg/kg), SZ (21.2 mg/kg), timosaponin BII (10 mg/kg) and timosaponin BIII (10 mg/kg).

Behavioral assessment

The FST was performed according to a previously described method with minor modifications [17]. Briefly, each mouse was individually placed in an open water-filled cylinder (H: 50 cm; Ø: 20 cm; water depth: 35 cm; temperature: 23–25 °C) and allowed to swim for a period of 6 min. The total time of immobility was recorded during the last 4 min of the testing duration. Immobility is defined as the mice floating in the water without movement. The TST was conducted as described in the literature [18]. Briefly, the mice were suspended 20 cm above the floor for 6 min. The time of immobility was recorded after the first 2 min. Mice in a completely motionless state were considered immobile.

Determination of monoamine neurotransmitter levels

The contents of dopamine (DA) and serotonin (5-HT) in the plasma were determined using ELISA Kits (Shanghai Jianglai Biotech Co., Ltd., Shanghai, China) according to the manufacturer’s instructions.

Data analysis

A noncompartmental analysis was carried out using WinNonlin software (Pharsight 6.2, NC, USA) to calculate the PK parameters. The significance of the results in behavior assessment was analyzed by unpaired Student’s t-tests. A P value less than 0.05 was considered significant. The liver extraction ratio (ER) indicated the fraction of hepatic clearance and first-pass effect, and the calculation formula was as reported in a previous study by our group [16].

Results

The forced swimming test (FST) and the tail suspension test (TST) together were performed to evaluate the antidepressant effect of BZD. As described in Table 1, BZD and fluoxetine both markedly attenuated the immobility time in the FST (P < 0.05) and TST (P < 0.01) by comparison with that of control mice, suggesting that BZD could alleviate depressive disorder. To further evaluate the contribution of the individual herbs in BZD in relieving depressive symptoms, the extracts of each herb were similarly applied in FST and TST. The results showed that the Zhimu decoction (ZD) also caused a significant reduction in immobility time in both tests compared with that of control mice (P < 0.05), indicating that the individual herbal medicine Zhimu could ameliorate depressive-like behavior. Meanwhile, there was no significant decrease in the duration of immobility after treatment with Baihe decoction (BD). Thus, the individual herb Zhimu may play a major role in relieving depression. Based on the analysis of the components of BZD, the extract of Zhimu was further separated into different fractions by microporous resin, including the polysaccharides of Zhimu (PZ), xanthones of Zhimu (XZ) and saponins of Zhimu (SZ), for screening for antidepressant activity. As shown in Table 2, the immobility time of the SZ-treated group was significantly decreased compared with that of the control group. No significant differences were observed in the PZ-treated and XZ-treated groups. These results indicated that SZ should be considered the main active fraction.

Table 1 Effects of BZD, ZD and BD on the immobility time of ICR mice in FST and TST
Table 2 Effects of different fractions of Zhimu on the immobility time of ICR mice in FST and TST

Pharmacokinetic study of five major components after oral administration of BZD

The HPLC-QQQ MS method has already been developed according to the previous study in our group [16]. In brief, full validation of the selectivity, linearity, accuracy, precision, matrix effect, extraction recovery, and stability, was performed for the simultaneous determination of two xanthones (neomangiferin and mangiferin) and three saponins (timosaponin BII, timosaponin BIII and timosaponin AIII) in the biological matrix by HPLC-QQQ MS. These results confirmed that two xanthones and three saponins in the biological matrix could be simultaneously determined by developed method [16]. Based on the developed method, five major components, including timosaponin BII, timosaponin BIII, mangiferin, neomangiferin and timosaponin AIII, were selected for simultaneous determination and pharmacokinetic analysis in the portal vein plasma, liver tissue, systemic plasma and brain tissue. The related pharmacokinetic parameters are summarized in Table 3.

Table 3 The PK parameters of five major components of Zhimu in the portal vein plasma, liver, systemic plasma and brain (containing hippocampus and cerebellum) after oral administration of BZD at 15 g/kg in rats (n = 5)

Pharmacokinetic study before hepatic disposition

Portal vein plasma is the site after gut absorption but before hepatic disposition. As displayed in Fig. 2, five compounds were accurately quantified in the portal vein plasma after oral administration of BZD. Figure 2 shows that timosaponin BII, timosaponin BIII, mangiferin and timosaponin AIII exhibited the double-peak phenomenon in the concentration–time curves, which may be caused by enterohepatic recirculation. The plasma concentrations of timosaponin BII, timosaponin BIII, mangiferin, neomangiferin and timosaponin AIII reached a maximum plasma concentration (Cmax) at 880.38 ± 159.95 ng/mL, 226.39 ± 43.92 ng/mL, 1757.12 ± 219.18 ng/mL, 64.64 ± 12.00 ng/mL and 61.79 ± 25.59 ng/mL, respectively. The Tmax values of timosaponin BII, timosaponin BIII and neomangiferin were greater than those of mangiferin and timosaponin AIII, and the rank order of t1/2 was timosaponin BII > timosaponin BIII > timosaponin AIII > mangiferin > neomangiferin. This result revealed that timosaponin BII was eliminated more slowly. The area under the concentration–time curve (AUC) is usually regarded as the objective marker for exposure to chemical components and as predictive of pharmacological responses [19]. The AUC values of timosaponin BII, timosaponin BIII, mangiferin, neomangiferin and timosaponin AIII were 1094.92 ± 183.89 ng/mL, 369.20 ± 74.63 ng/mL, 4020.89 ± 397.02 ng/mL, 34.36 ± 2.60 ng/mL and 582.66 ± 104.67 ng/mL, respectively. The large AUC values of the four compounds other than neomangiferin indicated good absorption and utility in the portal vein plasma.

Fig. 2
figure 2

Mean concentration–time curves of the major chemical constituents in the portal vein plasma after the oral administration of BZD at 15 g/kg in rats (n = 5)

Liver distribution

As displayed in Fig. 3, five compounds were determined accurately in the liver after oral administration of BZD. In the liver, timosaponin AIII had the maximum exposure, and the AUCs of timosaponin BII, timosaponin BIII, mangiferin, neomangiferin and timosaponin AIII were 6518.05 ± 411.22 ng/g, 859.42 ± 120.72 ng/g, 2520.63 ± 118.68 ng/g, 751.52 ± 87.96 ng/g and 199,598.41 ± 7787.31 ng/g, respectively. These values were different from those in the portal vein plasma, especially the highest value of timosaponin AIII. These results suggested that timosaponin AIII largely accumulated in the liver.

Fig. 3
figure 3

Mean concentration–time curves of the major chemical constituents in the liver tissue after the oral administration of BZD at 15 g/kg in rats (n = 5)

Pharmacokinetic study after hepatic disposition

After hepatic disposition, timosaponin BII, timosaponin BIII, mangiferin, neomangiferin and timosaponin AIII were transported to the systemic plasma. Figure 4 shows the similar pharmacokinetic properties of the five compounds detected in the systemic plasma and the portal vein plasma. Mangiferin showed the maximum AUC, followed by timosaponin BII, timosaponin BIII, timosaponin AIII and neomangiferin, with values of 4217.27 ± 177.38 ng/mL, 975.80 ± 253.95 ng/mL, 326.47 ± 54.29 ng/mL, 156.12 ± 21.63 ng/mL and 78.15 ± 5.66 ng/mL, respectively. Except for mangiferin and neomangiferin, the AUCs of the other compounds were much higher in the portal vein plasma than in the systemic plasma, corresponding to the effective recovery (ER) of timosaponin BII, timosaponin BIII and timosaponin AIII at 10.88%, 11.57% and 73.21%, respectively. In contrast, the ER values of mangiferin and neomangiferin were − 4.88% and − 127.44%, respectively. These results indicated that some other components may be converted to mangiferin and neomangiferin after hepatic metabolism in vivo.

Fig. 4
figure 4

Mean concentration–time curves of the major chemical constituents in the systemic plasma after the oral administration of BZD at 15 g/kg in rats (n = 5)

Brain distribution

As shown in Fig. 5, only two components were accurately quantified in the cerebellum and hippocampus after oral administration of BZD. Similar to those in the systemic blood, timosaponin BII and timosaponin BIII exhibited an obvious double-peak phenomenon in the time-concentration curves in the cerebellum. However, a multiple-peak phenomenon occurred in the hippocampus. This observation may be attributed to the multiple sites of intestinal absorption. The Cmax and AUC of timosaponin BII were 371.90 ± 153.80 ng/g and 9093.43 ± 1034.21 ng/g, respectively, with a larger Tmax at 10 h than that of timosaponin BIII (1 h). The Cmax and AUC of timosaponin BIII were 198.23 ± 122.31 ng/g and 2559.67 ± 928.52 ng/g, respectively. These results indicated that timosaponin BII and timosaponin BIII, which achieved exposure in the brain tissue, may be the main effective components of BZD.

Fig. 5
figure 5

Mean concentration–time curves of timosaponin BII (a) and timosaponin BIII (b) in the hippocampus tissue and of timosaponin BII (c) and timosaponin BIII (d) in the cerebellum tissue after the oral administration of BZD at 15 g/kg in rats (n = 5)

Antidepressant activity of chemical components detected in brain

To verify the screening results of the method, the FST and the TST were employed to assess the antidepressant effects of the chemical compounds detected in the brain. As shown in Table 4, both timosaponin BII and BIII markedly attenuated the time of immobility compared with that of control mice in both the FST (P < 0.05) and TST (P < 0.01), suggesting that they could alleviate depressive disorder. Furthermore, the timosaponins BII and BIII were used to screen for the active mechanism by using 5-HT and DA assays. As shown in Table 5, the levels of 5-HT in the timosaponin BII-treated group and the timosaponin BIII-treated group were both significantly decreased compared with that of the control group. These results further confirmed that timosaponins BII and BIII should be considered effective components of BZD.

Table 4 Effects of timosaponin BII and timosaponin BIII on the immobility time of ICR mice in FST and TST
Table 5 Effects of timosaponin BII and timosaponin BIII on the content of 5-HT and DA

Discussion

Depression is a common psychiatric disorder that affects mental and physical health and involves a number of symptoms, including low mood, lack of happiness and attention, sleep disorders and fatigue, and feelings of guilt [20,21,22]. Although many synthetic antidepressant drugs have been used to treat depression, the therapeutic effects are unsatisfactory due to numerous side effects, such as insomnia, headache, and anxiety [23,24,25]. Therefore, it is urgent to identify promising alternative agents with greater efficacy and fewer undesirable effects.

In China, many Chinese medicine formulae, which are also regarded as alternative therapeutic agents in other Asian countries, have been used for the treatment of depression [26, 27]. Baihe–Zhimu decoction (BZD) is a classical prescription used to treat depression. However, the complex composition of BZD and lack of clear evaluation standards make it less well understood and accepted than better-studied natural active compounds. There are multiple ingredients in Chinese medicine formulae, but sufficient absorption into the plasma or target organs is required for them to be considered effective compounds. Therefore, as shown in Fig. 1, we developed an integrated method combining classical tests for specific activity and pharmacokinetic studies based on HPLC-QQQ MS to screen for BZD components that were effective against depression.

The forced swimming test (FST) and the tail suspension test (TST) are commonly used to investigate antidepressant activity. Our data showed that BZD and Zhimu decoction (ZD, aqueous extract of Zhimu) elicited a significant reduction in the immobility time in both tests, indicating that the single herb Zhimu could obviously improve depressive-like behaviors, whereas the single herb Baihe could not. Thus, the single herb Zhimu may play a more important role in relieving depression. The effective chemical base is regarded as a bioactive compound with adequate exposure in the plasma and liver tissue. Therefore, as described above, five major components from Zhimu, namely, timosaponin BII, timosaponin BIII, mangiferin, neomangiferin and timosaponin AIII, were selected for further study according to their bioactivities and higher content in the plasma and liver tissue [15, 16]. These compounds were then selected for quantitative analysis and pharmacokinetic study to evaluate their distribution in the brain after oral administration of BZD. Finally, two components detected in the brain, timosaponin BII and timosaponin BIII, were further validated by FST and TST to scientifically and reasonably explain the chemical basis of the effect of BZD on depression.

The results showed that timosaponin BII and timosaponin BIII clearly improved depressive-like behaviors. Moreover, Lu et al. confirmed the significant antidepressant effect of timosaponin BII in depressive rats, which was likely related to the content of 5-HT in the brain [28]. Zhang et al. had already demonstrated that timosaponin BIII significantly ameliorated depressive effects in rats by regulating inflammatory cytokines, BNDF signaling and synaptic plasticity [29]. We found that YY-21 and YY-23, two modified derivatives of timosaponin BIII, also exhibited antidepressant activity in the animal model [30, 31]. The validated results regarding the antidepressant activities of timosaponin BII and timosaponin BIII were consistent with previous literature reports. Thus, given their high brain exposure and obvious antidepressant effects in vivo, timosaponin BII and timosaponin BIII are probably predominantly responsible for the antidepressant effect of orally administered BZD.

Conclusions

This investigation found that BZD, the individual herb Zhimu, and the saponins of Zhimu clearly ameliorated depressive-like behaviors evaluated by the forced swimming test (FST) and the tail suspension test (TST), whereas the individual herb Baihe did not, indicating that Zhimu may play a more important role in relieving depression. Based on the bioactivities and level of exposure in the plasma and liver tissue, five components, timosaponin BII, timosaponin BIII, mangiferin, neomangiferin and timosaponin AIII, were selected for quantitative analysis and pharmacokinetic characterization. Timosaponin BII and timosaponin BIII were accurately quantified in the brain after oral administration of BZD, and they were confirmed to improve depressive-like behaviors in the FST and TST. The levels of serotonin (5-HT) were also decreased after oral administration of timosaponin BII and timosaponin BIII. As described above, timosaponin BII and timosaponin BIII provided a rational explanation of the chemical basis for the effect of BZD on depression. The results of this study provide an effective method to discover lead compounds for depression therapy in traditional Chinese medicine.

Availability of data and materials

The datasets used in the current study are available from the corresponding author on reasonable request.

Change history

  • 02 April 2020

    In the original publication of this article [1], another affiliation (Affiliation 3: University of Chinese Academy of Sciences, Beijing 100049, People���s Republic of China.) for the author Ziqiong Guo is missing due to the carelessness during the author proof. In addition, in Affiliation 2 ���Chinese Academy of Science��� should be changed to ���Chinese Academy of Sciences���.

Abbreviations

FST:

forced swimming test

TST:

tail suspension test

DA:

dopamine

5-HT:

serotonin

HPLC-QQQ MS:

high-performance liquid chromatography coupled to triple quadrupole mass spectrometry

PK:

pharmacokinetics

ER:

extraction ratio

BZD:

Baihe–Zhimu decoction

ZD:

Zhimu decoction

BD:

Baihe decoction

PZ:

polysaccharides of Zhimu

XZ:

xanthones of Zhimu

SZ:

saponins of Zhimu

References

  1. Yeung WF, Chung KF, Ng KY, Yu YM, Ziea ET, Ng BF. A systematic review on the efficacy, safety and types of Chinese herbal medicine for depression. J Psychiatr Res. 2014;57:165–75.

    Article  Google Scholar 

  2. Ren Y, Zhu CJ, Wu JJ, Zheng RW, Cao HJ. Comparison between herbal medicine and fluoxetine for depression: a systematic review of randomized controlled trials. Complement Ther Med. 2015;23(5):674–84.

    Article  Google Scholar 

  3. Han DD, Chen CJ, Zhang C, Zhang Y, Tang X. Determination of mangiferin in rat plasma by liquid–liquid extraction with UPLC-MS/MS. J Pharm Biomed Anal. 2010;51(1):260–3.

    Article  CAS  Google Scholar 

  4. Zhang NT, Cheng C, Olaleye OE, Sun Y, Li L, Huang YH, Du FF, Yang JL, Wang FQ, Shi YH, Xu F, Li YF, Wen Q, Zhang NX, Li C. Pharmacokinetics-based identification of potential therapeutic phthalides from XueBiJing, a chinese herbal injection used in sepsis management. Drug Metab Dispos. 2018;46(6):823–34.

    Article  CAS  Google Scholar 

  5. Tian XT, Gao Y, Xu Z, Lian S, Ma YJ, Guo XZ, Hu P, Li ZX, Huang CG. Pharmacokinetics of mangiferin and its metabolite-Norathyriol, Part 1: systemic evaluation of hepatic first-pass effect in vitro and in vivo. BioFactors. 2016;42(5):533–44.

    Article  CAS  Google Scholar 

  6. Hu ZY, Yang JL, Cheng C, Huang YH, Du FF, Wang FQ, Niu W, Xu F, Jiang RR, Gao XM, Li C. Combinatorial metabolism notably affects human systemic exposure to ginsenosides from orally administered extract of Panax notoginseng roots (Sanqi). Drug Metab Dispos. 2013;41(7):1457–69.

    Article  CAS  Google Scholar 

  7. Tian XT, Liu F, Li ZX, Lin YF, Liu H, Hu P, Chen MC, Sun ZL, Xu Z, Zhang YT, Han L, Zhang YY, Pan GY, Huang CG. Enhanced anti-diabetic effect of berberine combined with timosaponin B2 in Goto-Kakizaki rats, associated with increased variety and exposure of effective substances through intestinal absorption. Front Pharmacol. 2019;10:19.

    Article  CAS  Google Scholar 

  8. Tian XT, Xu Z, Chen MC, Hu P, Liu F, Sun ZL, Liu H, Guo XZ, Li ZX, Huang CG. Simultaneous determination of eight bioactive compounds by LC-MS/MS and its application to the pharmacokinetics, liver first-pass effect, liver and brain distribution of orally administrated Gouteng-Baitouweng (GB) in rats. J Chromatogr B. 2018;1084:122–31.

    Article  CAS  Google Scholar 

  9. Tian XT, Liu H, Qiao SD, Yin H, Chen MC, Hu P, Wang YY, Peng HG, Liu F, Pan GY, Huang CG. Exploration of the hepatoprotective chemical base of an orally administered herbal formulation (YCHT) in normal and CCl4-intoxicated liver injury rats. Part 2: hepatic disposition in vivo and hepatoprotective activity in vitro. J Ethnopharmacol. 2019;236:161–72.

    Article  CAS  Google Scholar 

  10. Du HL, Wang KQ, Su L, Zhao HX, Gao SY, Lin QS, Ma XF, Zhu BK, Dong X, Lou ZY. Metabonomic identification of the effects of the Zhimu–Baihe saponins on a chronic unpredictable mild stress-induced rat model of depression. J Pharm Biomed. 2016;128:469–79.

    Article  CAS  Google Scholar 

  11. Yang B, Liu ZR, Wang Q, Chai YF, Xia PY. Pharmacokinetic comparison of seven major bioactive components in normal and depression model rats after oral administration of Baihe Zhimu decoction by liquid chromatography-tandem mass spectrometry. J Pharm Biomed. 2018;148:119–27.

    Article  CAS  Google Scholar 

  12. Jiang WX, Liu ZY, Wu B, Huang CG. Characterization and identification of major constituents in Baihe Zhimu decoction by HPLC-MSn. Asian J Chem. 2013;25(16):8976–80.

    Article  CAS  Google Scholar 

  13. Xie QM, Zhao HX, Li N, Su L, Xu X, Hong ZY. Protective effects of timosaponin BII on oxidative stress damage in PC12 cells based on metabolomics. Biomed Chromatogr. 2018;32:e4321.

    Article  Google Scholar 

  14. Sekar V, Mani S, Malarvizhi R, Nithya P, Vasanthi HR. Antidiabetic effect of mangiferin in combination with oral hypoglycemic agents metformin and gliclazide. Phytomedicine. 2019;59:152901.

    Article  CAS  Google Scholar 

  15. Li GL, Tang ZS, Yang J, Duan JN, Qian DW, Guo JM, Zhu ZH, Liu HB. Simultaneous determination of five components in rat plasma by UPLC-MS/MS and its application to a comparative pharmacokinetic study in Baihe Zhimu Tang and Zhimu extract. Molecules. 2015;20(4):6700–14.

    Article  CAS  Google Scholar 

  16. Xie Y, Zhou X, Pei H, Chen MC, Sun ZL, Xue YR, Tian XT, Huang CG. Metabolism, pharmacokinetics, and hepatic disposition of xanthones and saponins on Zhimu treatments for exploratively interpreting the discrepancy between the herbal safety and timosaponin A3-induced hepatotoxicity. Acta Pharmacol Sin. 2018;39(12):1923–34.

    Article  CAS  Google Scholar 

  17. Bourin M, Mocaër E, Porsolt R. Antidepressant-like activity of S 20098 (agomelatine) in the forced swimming test in rodents: involvement of melatonin and serotonin receptors. J Psychiatry Neurosci. 2004;29(2):126–33.

    PubMed  PubMed Central  Google Scholar 

  18. Steru L, Chermat R, Thierry B, Simon P. The tail suspension test: a new method for screening antidepressant in mice. Psychopharmacology. 1985;85:367–70.

    Article  CAS  Google Scholar 

  19. Wallemacq P, Armstrong VW, Brunet M, Haufroid V, Holt DW, Johnston A, Kuypers D, Le Meur Y, Marquet P, Oellerich M, Thervet E, Toenshoff B, Undre N, Weber LT, Westley IS, Mourad M. Opportunities to optimize tacrolimus therapy in solid organ transplantation: report of the European consensus conference. Ther Drug Monit. 2009;31(2):139–52.

    Article  CAS  Google Scholar 

  20. Serretti A, Mandelli L, Lattuada E, Smeraldi E. Depressive syndrome in major psychoses: a study on 1351 subjects. Psychiatry Res. 2004;127(1–2):85–99.

    Article  Google Scholar 

  21. Petschner P, Gonda X, Baksa D, Eszlari N, Trivaks M, Juhasz G, Bagdy G. Genes linking mitochondrial function, cognitive impairment and depression are associated with endophenotypes serving precision medicine. Neuroscience. 2018;370:207–17.

    Article  CAS  Google Scholar 

  22. Felger JC, Treadway MT. Inflammation effects on motivation and motor activity. Role of dopamine. Neuropsychopharmacology. 2017;42(1):216–41.

    Article  CAS  Google Scholar 

  23. Monteggia LM. Depression: the best way forward. Nature. 2014;515:200–1.

    Article  CAS  Google Scholar 

  24. Wang YF, Huang M, Lu XY, Wei RZ, Xu JY. Ziziphi spinosae lily powder suspension in the treatment of depression-like behaviors in rats. BMC Complement Altern Med. 2017;17:238.

    Article  Google Scholar 

  25. de Oliveira MR, Chenet AL, Duarte AR, Scaini G, Quevedo J. Molecular mechanisms underlying the anti-depressant effects of resveratrol: a review. Mol Neurobiol. 2018;55(6):4543–59.

    Article  Google Scholar 

  26. Pan QX, Wu JJ, Liu YY, Li XJ, Chen JX. Involvement of hepatic SHIP2 and PI3K/Akt signalling in the regulation of plasma insulin by Xiaoyaosan in chronic immobilization-stressed rats. Molecules. 2019;24:480.

    Article  Google Scholar 

  27. Li Y, Peng Y, Ma P, Yang HL, Xiong HY, Wang MY, Peng CS, Tu PF, Li XB. Antidepressant-like effects of Cistanche tubulosa extract on chronic unpredictable stress rats through restoration of gut microbiota homeostasis. Front Pharmacol. 2018;9:967.

    Article  Google Scholar 

  28. Lu MZ, Zhang ZZ, Yi J, Chen WS, Hou ZH, Li TJ. Study on the effect and mechanisms of timosaponin BII on antidepressant. J Pharm Pract. 2010;28(4):283–7.

    CAS  Google Scholar 

  29. Zhang XL, Wang L, Xiong L, Huang FH, Xue H. Timosaponin B-III exhibits antidepressive activity in a mouse model of postpartum depression by the regulation of inflammatory cytokines, BNDF signaling and synaptic plasticity. Exp Ther Med. 2017;14(4):3856–61.

    Article  CAS  Google Scholar 

  30. Guo F, Zhang B, Fu ZW, Ma YQ, Gao Y, Shen FY, Huang CG, Li Y. The rapid antidepressant and anxiolytic-like effects of YY-21 involve enhancement of excitatory synaptic transmission via activation of mTOR signaling in the mPFC. Eur Neuropsychopharm. 2016;26(7):1087–98.

    Article  CAS  Google Scholar 

  31. Zhang Q, Guo F, Fu ZW, Zhang B, Huang CG, Li Y. Timosaponin derivative YY-23 acts as a non-competitive NMDA receptor antagonist and exerts a rapid antidepressant-like effect in mice. Acta Pharmacol Sin. 2016;37(2):166–76.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Many thanks to Zhou Xu, Zhaolin Sun, Pei Hu, Yang Xie and Qiang Tian for their valuable ideas and comments.

Funding

This work was supported by the National Science and Technology Major Project of the Ministry of Science and Technology of China (Nos. 2018ZX09731016-003 and 2018ZX09201001-001-008), a grant from the Focus on research and development plan in Shandong Province, China (2018GSF119006), the National Natural Science Foundation of China for Young Scientists (No. 81803828), the General Financial Grant from the China Postdoctoral Science Foundation (No. 2018M642123), and the Youth Innovation Promotion Association of the Chinese Academy of Sciences (No. 2019280).

Author information

Authors and Affiliations

Authors

Contributions

FL, ZQG, CGH and XTT conceived and designed the study; FL and MZ wrote the main manuscript text; XTT, SJC, ZPS, ZXL, CMC and GHW performed the animal experiments; XTT, CMC, MNZ, HGG, MZ and GPZ analyzed the data and prepared the figures; FL, ZQG, CGH, XTT and MZ revised the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Fang Liu or Chenggang Huang.

Ethics declarations

Ethics approval and consent to participate

The animal care and experimental procedures used in the current study were approved by the Institutional Animal Care and Use Committee of Shanghai Institute of Materia Medica, Chinese Academy of Sciences (2018-01-HCG-27).

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhong, M., Tian, X., Chen, S. et al. Identifying the active components of Baihe–Zhimu decoction that ameliorate depressive disease by an effective integrated strategy: a systemic pharmacokinetics study combined with classical depression model tests. Chin Med 14, 37 (2019). https://doi.org/10.1186/s13020-019-0254-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13020-019-0254-9

Keywords