Skip to main content

REVIEW article

Front. Pharmacol., 11 March 2022
Sec. Drug Metabolism and Transport

A Comprehensive Review of the Main Lignan Components of Schisandra chinensis (North Wu Wei Zi) and Schisandra sphenanthera (South Wu Wei Zi) and the Lignan-Induced Drug-Drug Interactions Based on the Inhibition of Cytochrome P450 and P-Glycoprotein Activities

Feng Zhang,&#x;Feng Zhang1,2Jianxiu Zhai&#x;Jianxiu Zhai3Nan WengNan Weng3Jie Gao,,Jie Gao1,2,4Jun Yin
Jun Yin3*Wansheng Chen,,
Wansheng Chen1,2,5*
  • 1Department of Pharmacy, Changzheng Hospital, Navl Medical University (Second Military Medical University), Shanghai, China
  • 2Shanghai Key Laboratory for Pharmaceutical Metabolite Research, Shanghai, China
  • 3School of Traditional Chinese Material, Shenyang Pharmaceutical University, Shenyang, China
  • 4School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
  • 5School of Pharmacy, Research and Development Center of Chinese Medicine Resources and Biotechnology, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China

Wu Wei Zi is the dried fruit of Schisandra chinensis (Turcz.) Baill. or Schisandra sphenanthera Rehd. et Wils. (family Magnoliaceae). As a homology of medicine and food, it has been widely used in China for thousands of years, to tonify the kidney, and ameliorate neurological, cardiovascular, liver, and gastrointestinal disorders. As its increasing health benefits and pharmacological value, many literatures have reported that the combination of Wu Wei Zi in patients has led to fluctuations in the blood level of the combined drug. Therefore, it is extremely important to evaluate its safety concern such as drug-drug interactions (DDIs) when patients are under the poly-therapeutic conditions. This review summarized the effects of Wu Wei Zi extract and its major lignan components on cytochrome P450 and P-glycoprotein activities, the change of which could induce metabolic DDIs. Our review also elaborated on the differences of the major lignan components of the two Schisandra species, as well as the absorption, distribution, metabolism, and elimination of the major lignans. In conclusion, these results would enhance our understanding of the DDI mechanisms involving Wu Wei Zi, and may potentially untangle some differing and conflicting results in the future.

Introduction

“Wu Wei Zi” refers to the ripe fruits of two plants in the genus Schisandra, S. chinensis (Turcz.) Baill. (North Wu Wei Zi) or S. sphenanthera Rehd. et Wils (South Wu Wei Zi). It has been long used as nutritional and fatigue-fighting food supplement in China, Japan, Korea, and Russia. Records suggest that Russian hunters would eat Wu Wei Zi to alleviate fatigue, improve night vision, and replenish body fluids, while Ainu used Wu Wei Zi to protect against seasickness and cold weather (Panossian and Wikman, 2008). Both North Wu Wei Zi and South Wu Wei Zi were described with functions of astringency, reinforcing qi to generate body fluids, and tonifying the kidney to calm the mind (Chinese Pharmacopoeia Commission, 2020), along with hepatoprotective and anti-diabetic activities (Mu et al., 2006; Liu et al., 2017). Additionally, the two species also host unique properties. For instance, South Wu Wei Zi is more suitable for the treatment against chronic cough, mild breathlessness, and skin inflammation (Huyke et al., 2007), while North Wu Wei Zi has better cardiovascular-protective, neuroprotective, and tonic activities (Panossian and Wikman, 2008; Chun et al., 2014; Chen et al., 2015). More than 100 traditional Chinese medicine prescriptions composed of North Wu Wei Zi and South Wu Wei Zi are documented in the Pharmacopoeia of the People’s Republic of China (Ali et al., 2018), with various pharmacological activities. With their increasing consumption, it is extremely important to evaluate the safety concern such as drug-drug interactions (DDIs) when patients are under the poly-therapeutic conditions.

Recent studies revealed that both North Wu Wei Zi and South Wu Wei Zi are enriched in dibenzocyclooctadiene lignan components (Schisandra lignans, Figure 1), which are responsible for the major bioactivities of the two species (Sowndhararajan et al., 2018). However, the schisandra lignans are found to interfere with cytochrome P450 isoenzymes (CYPs) and P-glycoprotein (P-gp) activities, thus affecting the absorption, transportation, or metabolism of these or other drugs (Zhou et al., 2005; Qin et al., 2014a; Wang Cai-Ping et al., 2014), resulting metabolic DDIs. Depending on different medication circumstances, the “clinically relevant” DDI could lead to risk of adverse events or could be an advantageous for the therapy appropriateness processes (Palleria et al., 2020).

FIGURE 1
www.frontiersin.org

FIGURE 1. Structure of the major lignan components of Wu Wei Zi.

In recent years, the researchers paid more attention to the review of the botany, phytochemistry, chemical composition, in-depth in vivo pharmacological study, and clinical application of North Wu Wei Zi or South Wu Wei Zi (Szopa et al., 2017; Li et al., 2018; Huang et al., 2021), and only one review covered both North Wu Wei Zi and South Wu Wei Zi (Yang et al., 2022). However, little information was presented about their metabolic DDIs. In this review, we mainly focus on the main components of North Wu Wei Zi and South Wu Wei Zi, and identified CYP- and P-gp-interfering effects of the schisandra lignans, in order to better understand the quality control and potential DDIs of Wu Wei Zi products. By creating a full picture of the potential impacts of Wu Wei Zi, this review might facilitate investigations on DDIs for new chemical products and traditional Chinese medicines.

Literature Search Strategy

Literature research was conducted in May 2021 for articles published from 2000 to 2021, without restriction to regions and publication type. Eligible literatures for inclusion are written in English or Chinese. Electronic databases such as PubMed, Web of Science, ScienceDirect, Springer, Wiley, China National Knowledge Infrastructure (CNKI) and WANFANG Data were searched by two authors independently. Detailed PubMed strategy was listed as follows: key words were composed of Wu Wei Zi/Schisandra chinensis/Schisandra sphenanthera/schisandra lignans and contents/pharmacokinetics/DDI/CYP/P-gp. Articles dealing with the geographical, history, botany, ethnomedicine were excluded. Most articles were from PubMed (85 articles), supplemented by Science Direct (2 articles), Web of Science (2 articles), CNKI (26 articles, 6 dissertations), WANFANG Data (4 articles).

The Main Lignan Components of North Wu Wei Zi and South Wu Wei Zi

Schisandra lignans have a basic dibenzocyclooctadiene structure (Figure 1). They showed different contents in North Wu Wei Zi and South North Wu Wei Zi. The quantity and quality determinations of schisandra lignans to date are based on high-performance liquid chromatography (HPLC) methods such as HPLC-ultraviolet (UV), HPLC-capillary electrophoresis (CE), and HPLC-mass spectrometry (MS), or on gas chromatography-mass spectrometry (GC-MS) (Table 1). HPLC-UV has become the predominant method used to study Wu Wei Zi as there is a strong UV absorbance of schisandra lignans between 230 and 255 nm, resulting in high sensitivity and specificity (Zhang Hai et al., 2009).

TABLE 1
www.frontiersin.org

TABLE 1. Contents of major schisandra lignans of Wu Wei Zi.

The major schisandra lignans in North Wu Wei Zi were found to be schisandrin B (Sch B), schisandrin (SCH) and gomisin A (Gom A) (Ren and Zhang, 2020), while South Wu Wei Zi contains higher levels of anwulignan, schisandrin A (Sch A) and gomisin C (Gom C) (Sun et al., 2014). As such, SCH and Gom C are defined as the quality markers of North Wu Wei Zi and South Wu Wei Zi, respectively, according to the Chinese pharmacopoeia, where the SCH content is required to be above 0.40% (North Wu Wei Zi), and Gom C content should be higher than above 0.20% (South Wu Wei Zi).

In addition to species, the place of origin and associated climatic conditions could also affect the lignan contents. It was found that North Wu Wei Zi has a higher enrichment with schisandra lignans when collected from Zhongtiao Mountain, south and east of Qinling mountain, and south of Taihang mountain than from other locations. Similarly, the content of Gom C decreases as rainfall increases, and anwulignan content was reduced by higher maximum temperatures (Sun et al., 2014). It has also been reported that cultivated North Wu Wei Zi that were planted in a north-south orientation had higher lignan accumulation, with SCH, Sch A, and Sch B content in north-south planted North Wu Wei Zi 10.2, 25.8, and 26.8% higher than in those planted in an east-west orientation (Yang, 2011).

The “ADME” of the Primary Schisandra Lignans

Absorption

Pharmacokinetic studies have found that the lignans of Wu Wei Zi are readily absorbed into the bloodstream after oral ingestion. Following intragastric (i.g.) administration of Wuzhi capsule (a Chinese patent medicine composed of the ethanol extract of South Wu Wei Zi), Sch A, SCH, Gom A, Gom C, and schisanhenol were rapidly absorbed into the blood and were subsequently metabolized fully before elimination (Wei et al., 2010b; Wei et al., 2014; Wang et al., 2016).

Previous studies showed that SCH, the main lignan in North Wu Wei Zi, could be quickly absorpted after oral administration, with Tmax less lower than 1 h (Wang et al., 2003; Yan et al., 2006). Further study revealed that it is primarily absorbed in the duodenum and jejunum, followed by the colon and rectum, with absorption percentage of 37.4, 36.1, 24.9 and 31.4%, respectively (Zhang W et al., 2009). In contrast, Sch A and Sch B showed relative slower absorption. For example, the Tmax of Sch A was found to be between 2 and 4 h after i.g. of South Wu Wei Zi ethanol extract oil in rats (Wei et al., 2010a; Li H. L. et al., 2013), and a secondary absorption could be observed around 8 h (Li WL. et al., 2013). Sch B exhibited the first plasma concentration peak at 5.5–6.0 h, closely followed by a second absorption concentration peak 0.5 h after the first peak. Currently available data showed that the absorption rates are much faster in mice compared with rats, with observed secondary absorption of Sch C, Gom A, Gom B, and Gom C appearing at 8–25 h after North Wu Wei Zi extract administration. Meanwhile, the secondary Cmax was often lower that the primary one (Wei Hua et al., 2013; Liu et al., 2017). It is suggested that the secondary peak may be caused either by enterohepatic circulation or by the metabolic transformation from other schisandra lignans.

The majority of the schisandra lignans could change their absorption characteristics when Wu Wei Zi was administrated along with other medical materials. For example, the absorption rate of SCH decreased slightly (Tmax 0.32∼1.00 h) when North Wu Wei Zi was administrated in the prescription “Shengmai Powder” which also contains Radix Ophipogonis and Radix et Rhizoma Ginseng (Wang et al., 2003; Li et al., 2005; Guo et al., 2012). Moreover, SCH showed a significantly decreased Tmax at 7.0 h when administrated in the prescription “Longlingchun mixture”, which is composed of North Wu Wei Zi and other ten Chinese medicines (Zhao et al., 2006).

Distribution

After absorption, or after tail vein injection, the highest concentrations of the main schisandra lignans (such as SCH, Sch A, and Sch B) were presented in the lungs and liver, followed by the heart, kidneys, and lastly the spleen (Zhou et al., 2013). Sch A and SCH were also both found in rat brain at a very low concentration level, suggesting that they were able to cross the blood-brain barrier (Hu, 2012). Within the different rat brain regions, SCH showed the highest concentrations in the hypothalamus, striatum, and hippocampus, followed by the brain stem, and the lowest concentration of which was found in the cerebral cortex and cerebellum. The high distribution in hypothalamus, striatum, and hippocampus may explain the neuro-protective effects of SCH and North Wu Wei Zi on mice with Alzheimer’s disease and depression (Wang et al., 2013; Wang Baolian et al., 2014).

Metabolism

The major phase I metabolism pathways of schisandra lignans are demethylation and hydroxylation (Zhang et al., 2014; Zhang et al., 2017a), including a total of 33 metabolites characterized from six lignans following administration of a South Wu Wei Zi preparation. In detail, the enzyme CYP3A4 catalyzes Sch A to SCH in the first step (Cao, 2010). Secondly, SCH formed SCH-M1, mediated by CYP3A4 again at hydroxylation of C8 position, and then biotransformed by demethylation of C2 position (SCH-M2a) or C3 position (SCH-M2b) (Figure 2). The production of SCH-M2b was characterizes two-thirds of that of SCH-M2a in the rat liver microsomes study. The main metabolic pathways of Sch B have been revealed as the mono-oxygenation of C4 or C11 position, the demethylation of the methoxy group on C12 position, or demethylation at the methylenedioxy group connected with C2 and C3 positions which opens the five-member ring, followed by glucuronidation on C3 position (Qian et al., 2015).

FIGURE 2
www.frontiersin.org

FIGURE 2. The metabolism of schisandrin A (Sch A) and schisandrin (SCH).

Elimination

Wu et al. (2014a) reported that the main schisandra lignans SCH, Sch A, and Sch B, were fully eliminated within 25 h in male rats following South Wu Wei Zi extract administration. Basically, schisandra lignans and their metabolites are gradually excreted through feces and urine (Qian et al., 2015).

Wu Wei Zi Induced Drug-Drug Interactions

The Biphasic Effect of Schisandra Lignans on CYPs

DDIs have been implicated in some studies for both North Wu Wei Zi and South Wu Wei Zi. Interestingly, recent studies have identified seemingly contradictory inductive and inhibitory effects of their water or ethanol extracts, and more specifically schisandra lignans, on CYPs and P-gp.

Both Wu et al. (2007a) and Mao and Tay (2016) found that North Wu Wei Zi extract inhibited CYP3A in human liver microsomes (HLMs; in vitro) and in healthy volunteers (in vivo). In contrast, Hu (2011) reported that North Wu Wei Zi extract increased CYP450 contents in Kunming mice after 7-day-oral administration. The bi-directional effect on CYPs was first identified by Cheng et al., and it was found that CYP3A activity in rats was inhibited within 12 h after North Wu Wei Zi water extract administration yet was increased after 6-day-oral administration. The phenomenon was further discussed by Chen et al. (2010), suggesting that the bi-directional effect of North Wu Wei Zi extracts on CYPs depend on the pre- or co-treatment time period. The inhibition effect could be observed immediately after administration, while the induction effect could only be monitored till the episode of DNA transcription and protein synthesis. Besides, a few schisandra lignans showed contradictory effects on CYP activities when involving individually in different experimental conditions. For example, Sch B was identified as a CYP inducer when administrated to mice in vivo (Liu, 1988), yet could inhibit CYP3A activity when incubated with rat liver microsomes in vitro (Li et al., 2010). In general, both North Wu Wei Zi and South Wu Wei Zi extracts, Sch A, Sch B, and SCH are recognized as activators when administered in vivo with long-term administration (>6 days), and as inhibitors when incubated with liver microsomes or CYP isoforms in vitro (Tables 2, 3).

TABLE 2
www.frontiersin.org

TABLE 2. Biphasic effect on regulating CYP activities by Wu Wei Zi extract.

TABLE 3
www.frontiersin.org

TABLE 3. Inhibitory of main CYP450 isoform activity by the major schisandra lignans.

Zhai et al. (2015) refined the theory, suggesting that Wu Wei Zi extracts or their major schisandra lignans exhibit inhibitory or inductive effect on CYPs activities depending on the experimental conditions. Specifically, it is suspected that the main schisandra lignans could induce CYP DNA expression and inhibit CYP protein activity at the same time; however, the CYP inhibition would be observed immediately, explaining CYP-inhibitory effects were only seen in in vitro experiments when using liver microsomes or CYP isoforms, which did not rely on DNA transcription and translation. On the other hand, the lignan-induced CYP expression could only occur after DNA transcription and translation had taken place, a process of which apparently could take 5 days or more in vivo. The modified theory has been supported by other studies. For example, North Wu Wei Zi water extract inhibited CYP3A activity with single-dose administration and induced CYP3A activity with multiple administration in rats (1–3 g/kg, qd for 3 days) (Chen et al., 2010) (Table 2). This study also showed that the CYP3A induction was caused by an increase of mRNA transcription with Sch B and Gom A.

The inhibition or induction of Wu Wei Zi and its major schisandra lignans is summarized in Tables 2–4. For ease of comparison, the dose of Wu Wei Zi in the in vivo studies were converted into human dosage according to an average body weight of 54 kg.

The Inhibitory Effects of Wu Wei Zi and its Major Lignans on CYP Enzymes

Wu Wei Zi extracts were found with extensive inhibitory effects on CYPs (Table 2). CYP inhibition can generally be classified as reversible or irreversible interactions (Berg et al., 2012), with reversible CYP inhibitions further divided into competitive, non-competitive, and un-competitive interactions according to the interaction between inhibitors and CYP enzymes. For irreversible inhibition, inhibitors are first catalyzed by CYPs into reactive intermediates which then form metabolite-inhibitor complexes with CYP that causes deactivation. As a result, irreversible inhibitions are time-dependent and NADPH-dependent process when the inhibitors exposed to the CYP. This phenomenon could be reinforced by multiple dosing of the inhibitors in vivo.

Wang et al. (2011) reported that CYP1A2, CYP2D2, CYP2C6, CYP2C11, CYP2E1, and CYP3A activities could be inhibited by North Wu Wei Zi water/ethanol extract in rat liver microsomes (RLMs). The activities of the CYPs were reduced by 50–90% at a concentration of 120 μg/ml when incubated with North Wu Wei Zi ethanol extract, which was recognized as a stronger CYP inhibitor than North Wu Wei Zi water extract. Moreover, the activities of CYP2D2 and CYP2C11 remained inhibited 1 week after North Wu Wei Zi ethanol administration (i.g., 1.5 g/kg). South Wu Wei Zi was also found to be a potent CYP3A inhibitor in rat hepatic or intestinal microsomes, the lignan extract of which showed IC50 values of 25–40 μM. It was further confirmed that Gom C, the most abundant lignan in South Wu Wei Zi, exhibited a strong inhibitory effect in a time-dependent manner, indicating a mechanism-based inhibition (Lai et al., 2009).

Both North and South Wu Wei Zi and their major lignan components have been investigated as CYP inhibitors in DDI studies with clinical-practiced medicines for financial burden reducing, toxicity-alleviating, or pharmacodynamics-enhancing purposes. Take tacrolimus (FK506), a natural immunosuppressant as an example, which is metabolized by CYP3A (Harding et al., 1989; Chen et al., 2020). Its blood concentration can be largely increased by the combination of Wuzhi tablet, a commercially available medicine composed of South Wu Wei Zi ethanol extract, owing to the schisandra lignans’ higher affinity to CYP3A than that of FK506 (Qin et al., 2010a; Qin et al., 2014b; Li et al., 2017; Qin et al., 2020). Thus, Wuzhi tablet is recognized to reduce the oral dosage of FK506 as well as relieve FK506-induced hepatotoxicity, which is similar to Wuzhi capsule (Wei et al., 2014; Wang et al., 2016; Cheng et al., 2021). Many clinical practices have evidenced that both Wuzhi tablet and Wuzhi capsule could effectively increase the blood exposure of FK506 in renal, heart and liver transplanted patients (Jiang et al., 2010; Xin et al., 2011; Liu et al., 2012; Zhou et al., 2017). Similarly, as csyclosporin A was a substrate of CYP3A and P-gp, Wuzhi Tablet could also dramatically increase its Cmax and the blood exposure of by 84.1 and 293.1%, respectively (Xue et al., 2013). What’s more, Wuzhi tablet was also effective in increasing the blood exposure of paclitaxel by 1.9˗fold in rats, while Gom A could effectively increase the Cmax of oral-administered paclitaxel by 2.8˗fold at an oral dose of 25 mg/kg (Jin et al., 2011). Meanwhile, the North Wu Wei ethyl acetate extract was found to block the CYP3A-mediated toxic metabolic pathway of cyclophosphamide, and thus reduce the metabolite related liver, kidney, and brain toxicity in rats (Zhai et al., 2018).

With further investigation, it was found that the CYP-inhibitory efficacy comes from several major lignans of south and north Wu Wei Zi. Zhai et al. have identified Gom A as an efficient CYP3A blocker (KI = 0.35 µM, kinact = 1.96 min−1) (Zhai et al., 2017), while Iwata et al. reported that the CYP3A-inhibiting efficacy of Gom C was found stronger than that of ketoconazole (Iwata et al., 2004). Additionally, other schisandra lignans also take roles as CYP inhibitors (Table 3). For example, Sch A and Sch B have both been found to decrease CYP3A activity with IC50 ranging from 6 to 70 μM (Li Ruidong et al., 2012; Wu et al., 2014), and the inhibition effect was not reversed by activation after 3 days of continuous administration (Li Wei-Liang et al., 2012; Li WL. et al., 2013).

So far, the major schisandra lignans were found working as CYP-inhibitiors with different mechanisms. The in vivo evidences reported that Sch A and B could dose-dependently inhibited hepatic microsomal CYP3A activity as noncompetitive inhibitors (Ki Sch A = 30.67 mg/kg; Ki Sch B = 16.64 mg/kg) (Li Ruidong et al., 2012; Li H. L. et al., 2013). Meanwhile, Gom A was found as a mix-type CYP3A inhibitor, which showed characteristics of competitive as well as time- and NADPH- dependent inhibition (Zhai et al., 2017), while Gom C could irreversibly inactivate CYP3A by forming metabolite-inhibitor complexes (Iwata et al., 2004). It is suspected that the potent inhibition on CYP3A may be caused by the metabolite-intermediate complexes formed by the methylenedioxyphenyl structure with P450 enzyme (Iwata et al., 2004; Lai et al., 2009; Seo et al., 2021). In all, lignans with one methylenedioxyphenyl group such as Gom A, Gom B, Gom C, and Sch B most strongly inhibited CYP3A activity, with IC50 values as low as 0.19–0.28 μM, which was much lower than those of the other P450 isoforms. For these schisandra lignans, some researches obtained larger IC50 values, the discrepancy of which could be due to differences in incubation conditions including CYP3A probe substrates and/or the enzyme source. Among those schisandra lignans, most of them showed potent or moderate inhibition of on CYP2C19, with IC50 values ≤16.3 μM, except Gom B (>50 μM). Sch C with two methylenedioxyphenyl groups showed most inhibitory effect on CYP2C19, with a IC50 value of 2.7 μM. Only SCH, containing none methylenedioxyphenyl group but one hydroxyl group, moderately inhibited CYP2E1(IC50 value 4.2 μM).

The Inductive Effects of Wu Wei Zi and its Major Lignans on CYP Enzyme Activity

Since the late 1960s, both North Wu Wei Zi and South Wu Wei Zi extracts have shown hepatic-protective effects in clinical practice (Zuo et al., 2019). Apart from the alleviation of inflammatory response, it was found that North Wu Wei Zi extract significantly increased the smooth endoplasmic reticulum in hepatocytes of rats after continuous ingestion (≥3 days, qd), which was consistent with the increased liver CYP content and the metabolism acceleration of toxic components in experimental animals (Qiu et al., 2018). More studies found that multiple prolonged administration of Wu Wei Zi extract (≥3 days, qd) could lead to CYP induction, increasing activities of CYP3A, CYP2C, and CYP2E (Chen et al., 2010; Mu et al., 2006) (Table 4), and its administration at high dose could be more likely to cause CYP induction (≥1 g/kg/d for rats, which is equivalent to≥8.6 g/d for human, Table 2). Besides, continuous ingestion of Wu Wei Zi extract at low dose (0.15 g/kg/d for rats, which is equivalent to 1.3 g/d for human) for 2 weeks was also found to cause CYP3A induction in rats (Lai et al., 2009).

TABLE 4
www.frontiersin.org

TABLE 4. Induction of CYP3A activity by the major schisandra lignans.

Mu et al. (2006) further suggested that the mechanism of CYP induction of Wu Wei Zi extract may be caused by the activation of the pregnane X receptor, with Sch A, Sch B, and Gom B as major responsible agonists. Interestingly, recent investigations revealed that pregnane X receptor might also be the key target for the hepato-protective efficacy of Gom A. By activating the pregnane X receptor, Gom A could accelerate the bile acid metabolism, promote bile acid efflux and induce hepatic expression of Cyp3a in mice (Zeng et al., 2017).

Effects of Wu Wei Zi and Their Major Lignans on P-Glycoprotein Activity

As well as the CYP-dependent metabolism changes, phytochemical-mediated alterations in P-glycoprotein (P-gp) activity may also produce DDIs by altering drug absorption, distribution, and elimination, which could be achieved either by decreasing P-gp expression or by inhibiting P-gp activity. For example, North Wu Wei Zi extract could effectively increase the blood exposure of P-gp substrate taninolol by 47% in healthy volunteers (Fan et al., 2009). A number of studies have shown that Gom A, Gom C, SCH, Sch A, and Sch B could reverse P-gp-mediated multidrug resistance (MDR) and decrease the efflux of P-gp substrate in cancer cells (Wan et al., 2006; Fong et al., 2007; Li et al., 2007; Yoo et al., 2007; Qin et al., 2010b).

Interestingly, a subsequent transport study using Caco-2 cells also found that Gom A, Gom C, Sch A, and SCH were not substrates of P-gp (Qin et al., 2014a). It was found that instead of directly inhibiting P-gp activity, the underlying mechanism of the schisandra lignans may be intervening with the substrate-P-gp complex and blocking the active P-gp transport sites (Fong et al., 2007). What’s more, continuous treatment with schisandra lignan extract for 10 days (or >10 days) would cause massive P-gp expression decrease in rat intestinal and brain tissues (Liang et al., 2013).

Discussion and Conclusion

In clinical setting, combination with traditional Chinese medicine is usually used to enhance the efficacy and/or reduce adverse reactions of modern medicines. Over the past few years, there have been many reports concerning the pharmacokinetic interactions involving the components of Wu Wei Zi in the form of herb extract, herb decoction, or herb preparation (Feng et al., 2017). Therefore, Table 5 summarized some DDI in animal experiments and clinical researches. It was found that Wu Wei Zi and its preparations have the capacity of affecting the blood concentration/exposure and pharmacokinetic profiles of other drugs. For example, the blood concentration of FK506 in vivo mostly contributed to the inhibition of CYP3A4 and/or P-gp via schisandra lignans when co-administration both in single and multiple doses. Meanwhile, the blood concentrations of the schisandra lignans were decreased because of their higher affinity to CYP3A. The schisandra lignans could also enhance the blood exposure of the compounds which were metabolized by CYP2D6 and CYP2D2, or transported by P-gp. The same compound showed different increases in the Cmax or AUC when the schisandra lignans co-administrated in different situation, indicating the occurrence of DDI was due to be the overall effect of all of the complex chemical components rather than the activity of an individual component. Considering the similarity of the chemical structure of the major schisandra lignans and their transformation during the episode of their metabolism, it is possible that the CYP3A/P-gp inhibitory activity of Wu Wei Zi should be considered as an overall and comprehensive effect of all schisandra lignans and their metabolites. It is important to point out that although schisandra lignans exhibit the potential for other CYPs-associated DDI based mostly on in vitro experimental results (Tables 2-4), “clinically relevant” DDI mediated by these CYPs (such as 2C19 and CYP2E1) are limited.

TABLE 5
www.frontiersin.org

TABLE 5. Reported DDIs in animal experiments and clinical researches.

Nowadays, a few in vitro studies have evaluated the modulatory effects of individual schisandra lignans on CYP3A/P-gp activities, the interactions and synergistic impact of schisandra lignans on CYP3A/P-gp activities are still remains to be further investigated. Besides, the discovery of a family of nuclear receptors such as pregnane X receptor (PXR), constitutive androstane receptor (CAR) and glucocorticoid receptor (GR) had given insight into the molecular explanation of CYP3A induction by xenobiotics (Qin and Wang, 2019). Regarding that Sch A, Sch B, SCH and Gom C could activate human PXR and induce the CYP3A4 reporter gene assay (Zhang et al., 2020), whether the same phenomenon could be observed for CAR and GR was not known. Therefore, studies should begin to explore the potential of these nuclear receptors as targets for those schisandra lignans to elucidate the ability to modulate the expression of CYP/P-gp whether they directly or through the transcriptional activation of nuclear receptors.

According to “Pharmacopoeia of the People’s Republic of China”, the suggested daily dosage of Wu Wei Zi is 2∼6 g. Based on the above discussion, the dosage is unlikely to affect the CYP activity according to Table 2 (the equivalent CYP inhibitory/inductive dosage of North- and South-Wu Wei Zi extract for human were annotated by “EDH”). Even so, the risk of CYP inhibition is high when patients are taking other CYP-blocking chemical drugs at the same time (especially CYP3A), such as nefazolone, ketoconazole, erythromycin and clarithromycin (Manikandan and Nagini, 2018). On the other hand, long-term use of Wu Wei Zi along with CYP-inducers (such as nevirapine and rifampin) could cause significant CYP induction (Finkelstein et al., 2010). In the above cases, the clinicians should put extra caution in applying drugs with narrow theraputic windows, like theophylline, tizanidine, warfarin, phenytoin, tacrolimus and quinidine (Schutte-Nutgen et al., 2018; Patocka et al., 2020). Meanwhile, clinicians must be aware of the contents and pharmacokinetic features of the schisandra lignans in Wu Wei Zi, which is helpful to understand the potential change of chemical drug efficacy with Wu Wei Zi co-administration, as well as the opposite after discontinuation of Wu Wei Zi. Therefore, this paper aimed to provide enough published data about DDIs induced by Wu Wei Zi and the schisandra lignans, and expected to provide guidance for rational use of chemical drugs or herbal medicines in order to avoid the occurrence of adverse side effects via CYP3A or P-gp inhibition and/or induction. Of note, the information and knowledge that were given in this review were the present practical methods and rules for screening and characterization of CYP3A/P-gp inhibitors and inductors and for facilitating the investigations on CYP3A/P-gp mediated DDI interactions.

Author Contributions

FZ and JZ wrote this manuscript. JZ and NW designed tables and figures. JG checked this manuscript. WC and JY supervised the project and critically revised the manuscript.

Funding

This project was supported by Innovative Clinical Research Funding Project (2020YLCYJ-Y25), the Three-year Action Plan for the Development of Traditional Chinese Medicine of Shanghai (ZY (2021-2023)-0215), Shanghai Municipal Health Commission (ZYCC2019018), Jin-Zi-Ta Talent project (0806 and 1016), Science and Technology Commission of Shanghai Municipality (22S21901900), National Natural Science Foundation of China (81830109, 81573793) and Shanghai Science and Technology Development Funds (19QB1404500).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Ali, M., Khan, T., Fatima, K., Ali, Q. U. A., Ovais, M., Khalil, A. T., et al. (2018). Selected Hepatoprotective Herbal Medicines: Evidence from Ethnomedicinal Applications, Animal Models, and Possible Mechanism of Actions. Phytother. Res. 32 (2), 199–215. doi:10.1002/ptr.5957

PubMed Abstract | CrossRef Full Text | Google Scholar

Avula, B., Choi, Y. W., Srinivas, P. V., and Khan, I. A. (2005). Quantitative Determination of Lignan Constituents from Schisandra Chinensis by Liquid Chromatyography. Chromatographia 61 (9-10), 515–518. doi:10.1365/s10337-005-0547-2

CrossRef Full Text | Google Scholar

Berg, J., Tymoczko, J., and Stryer, L. (2012). Editor W. H. Freeman (Basingstoke.Biochemistry

Google Scholar

Cao, Y. (2010)., (Vol Doctor). Shenyang: Shenyang Pharmaceitical Univiersity.In Vitro metabolic Properties of Schisandrae LignansSch. traditional Chin. materia

Google Scholar

Chen, L., Xiong, X., Hou, X., Wei, H., Zhai, J., Xia, T., et al. (2019). Wuzhi Capsule Regulates Chloroacetaldehyde Pharmacokinetics Behaviour and Alleviates High-Dose Cyclophosphamide-Induced Nephrotoxicity and Neurotoxicity in Rats. Basic Clin. Pharmacol. Toxicol. 125 (2), 142–151. doi:10.1111/bcpt.13211

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, L., He, L., Yu, C., and Chen, J. (2015). Research Progress of Neuroprotective Effects of Schisandra Chinensis Baill Lignans. J. Beihua Univ. (Natural Science),

Google Scholar

Chen, Q., Wu, Y. J., Cheng, N. N., Li, Y. L., and Wang, Y. M. (2010). Dual Effects of Extract of Schisandra Chinensis Baill on Rat Hepatic CYP3A. Yao Xue Xue Bao 45 (9), 1194–1198.

PubMed Abstract | Google Scholar

Chen, X., Wang, Y., Ma, N., Tian, J., Shao, Y., Zhu, B., et al. (2020). Target Identification of Natural Medicine with Chemical Proteomics Approach: Probe Synthesis, Target Fishing and Protein Identification. Signal. Transduct. Target. Ther. 5 (1), 72. doi:10.1038/s41392-020-0186-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, X., Zhao, Z., Chen, Y., Gou, X., Zhou, Z., Zhong, G., et al. (2016). Mechanistic Understanding of the Effect of Dengzhan Shengmai Capsule on the Pharmacokinetics of Clopidogrel in Rats. J. Ethnopharmacol. 192, 362–369. doi:10.1016/j.jep.2016.07.066

CrossRef Full Text | Google Scholar

Cheng, F., Li, Q., Wang, J., Zeng, F., and Zhang, Y. (2021). Effects and Safety Evaluation of Wuzhi Capsules Combined with Tacrolimus for the Treatment of Kidney Transplantation Recipients. J. Clin. Pharm. Ther. 46 (6), 1636–1649. doi:10.1111/jcpt.13493

CrossRef Full Text | Google Scholar

Chinese Pharmacopoeia Commission (2020). Pharmacopoeia of the People’s Republic of China, (Vol. 1). Beijing: People’s Medical Publishing House.

Google Scholar

Chun, J. N., Cho, M., So, I., and Jeon, J. H. (2014). The Protective Effects of Schisandra Chinensis Fruit Extract and its Lignans against Cardiovascular Disease: a Review of the Molecular Mechanisms. Fitoterapia 97, 224–233. doi:10.1016/j.fitote.2014.06.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Cui, Y., Li, Y., Fan, L., An, J., Wang, X., Fu, R., et al. (2021). UPLC-MS/MS Method for the Determination of Lenvatinib in Rat Plasma and its Application to Drug-Drug Interaction Studies. J. Pharm. Biomed. Anal. 206, 114360. doi:10.1016/j.jpba.2021.114360

CrossRef Full Text | Google Scholar

Deng, X., Chen, X., Cheng, W., Shen, Z., and Bi, K. (2008). Simultaneous LC-MS Quantification of 15 Lignans in Schisandra Chinensis (Turcz.) Baill. Fruit. Chroma 67 (s7-8), 559–566. doi:10.1365/s10337-008-0589-3

CrossRef Full Text | Google Scholar

Dou, Z. H., An, L. P., Chen, M., Miao, Y. X., and Feng, Y. (2014). Simultaneous Determination of Six Lignanoids in Schisandra Chinensis by Quantitative Analysis of Multi-Components by Single Marker. Chin. Pharm. J. (Chinese) 49 (2), 147–151. doi:10.11669/cpj.2014.02.016

CrossRef Full Text | Google Scholar

Fan, L., Mao, X. Q., Tao, G. Y., Wang, G., Jiang, F., Chen, Y., et al. (2009). Effect of Schisandra Chinensis Extract and Ginkgo Biloba Extract on the Pharmacokinetics of Talinolol in Healthy Volunteers. Xenobiotica 39 (3), 249–254. doi:10.1080/00498250802687657

PubMed Abstract | CrossRef Full Text | Google Scholar

Feng, G., Zhai, J. X., Chen, W. S., Xiong, X. J., Gao, S. H., Yang, Y., et al. (2017). Study Progress of the Interactions between the Schisandra Chinensis and the Schisandra Sphenanthera as Well as Thier Preparations with Other Drugs. Chin. J. Hosp. Pharm. (Chinese) 37 (21), 2206–2208.

Google Scholar

Finkelstein, Y., Aks, S. E., Hutson, J. R., Juurlink, D. N., Nguyen, P., Dubnov-Raz, G., et al. (2010). Colchicine Poisoning: the Dark Side of an Ancient Drug. Clin. Toxicol. (Phila) 48 (5), 407–414. doi:10.3109/15563650.49534810.3109/15563650.2010.495348

PubMed Abstract | CrossRef Full Text | Google Scholar

Fong, W. F., Wan, C. K., Zhu, G. Y., Chattopadhyay, A., Dey, S., Zhao, Z., et al. (2007). Schisandrol A from Schisandra Chinensis Reverses P-Glycoprotein-Mediated Multidrug Resistance by Affecting Pgp-Substrate Complexes. Planta Med. 73 (3), 212–220. doi:10.1055/s-2007-967120

PubMed Abstract | CrossRef Full Text | Google Scholar

Fu, R., Wang, X. N., Guo, C. H., Li, Y., Ding, C. Y., Li, Y. J., et al. (2021). Wuzhi Capsule Increased Systemic Exposure to Methotrexate by Inhibiting the Expression of OAT1/3 and P-Gp. Ann. Transl. Med. 9 (10), 845. doi:10.21037/atm-21-1303

PubMed Abstract | CrossRef Full Text | Google Scholar

Gao, J. P., Wang, Y. H., and Yu, Y. Q. (2003). Determination by HPLC and Variation Regularity of Lignan Constituents in Chinese Crude Drug Fructus Schisandrae Sphenantherae. Chin. J. Nat. Medicines (Chinese) 1, 89–93.

Google Scholar

Geng, L. C., and Yang, Z. L. (2007). Determination of Lignanoids in Fructus Schisandrae from Different Regions by HPLC. Acta Chin. Med. Pharmacol. 35 (2), 47–48. doi:10.3969/j.issn.1002-2392.2007.02.017

CrossRef Full Text | Google Scholar

Gu, W., Wei, N., and Wang, Z. (2008). LC analysis of lignans from Schisandra sphenanthera Rehd. et Wils. Chroma 67 (11-12), 979–983. doi:10.1365/s10337-008-0621-7

CrossRef Full Text | Google Scholar

Guo, W. J., Shao, Q., Zhang, Y. F., and Fan, X. H. (2012). Pharmacokinetics Interaction Among Three Major Active Compounds of Shengmai Formula in Rats. Zhejiang Da Xue Xue Bao Yi Xue Ban 41 (1), 6–12. doi:10.3785/j.issn.1008-9292.2012.01.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Halstead, C. W., Lee, S., Khoo, C. S., Hennell, J. R., and Bensoussan, A. (2007). Validation of a Method for the Simultaneous Determination of Four Schisandra Lignans in the Raw Herb and Commercial Dried Aqueous Extracts of Schisandra Chinensis (Wu Wei Zi) by RP-LC with DAD. J. Pharm. Biomed. Anal. 45 (1), 30–37. doi:10.1016/j.jpba.2007.05.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Harding, M. W., Galat, A., Uehling, D. E., and Schreiber, S. L. (1989). A Receptor for the Immunosuppressant FK506 Is a Cis-Trans Peptidyl-Prolyl Isomerase. Nature 341, 758–760. doi:10.1038/341758a0

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, D., Han, N., Yao, X., Liu, Z., Wang, Y., Yang, J., et al. (2014). Structure-activity Relationship Study of Dibenzocyclooctadiene Lignans Isolated from Schisandra Chinensis on Lipopolysaccharide-Induced Microglia Activation. Planta Med. 80 (08/09), 671–675. doi:10.1055/s-0034-1368592

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, D. (2012). Studies on Active Lignans from Schisandra Chinensis for Anti-alzheimer's Disease. Shenyang: Shenyang Pharmaceitical Univiersity (Vol Doctor).

Google Scholar

Hu, F. (2011). The Effect of Vinegar-Preparation on the Schisandra Chinensis Lignan Contents and its Effect on CYP450 Activity, (Vol Doctor). Nanjing: Nanjing University of Chinese Medicine.

Google Scholar

Huang, S., Zhang, D., Li, Y., Fan, H., Liu, Y., Huang, W., et al. (2021). Schisandra Sphenanthera: A Comprehensive Review of its Botany, Phytochemistry, Pharmacology, and Clinical Applications. Am. J. Chin. Med. 49 (7), 1577–1622. doi:10.1142/S0192415X21500749

CrossRef Full Text | Google Scholar

Huyke, C., Engel, K., Simon-Haarhaus, B., Quirin, K. W., and Schempp, C. M. (2007). Composition and Biological Activity of Different Extracts from Schisandra Sphenanthera and Schisandra Chinensis. Planta Med. 73 (10), 1116–1126. doi:10.1055/s-2007-981559

PubMed Abstract | CrossRef Full Text | Google Scholar

Iwata, H., Tezuka, Y., Kadota, S., Hiratsuka, A., and Watabe, T. (2004). Identification and Characterization of Potent CYP3A4 Inhibitors in Schisandra Fruit Extract. Drug Metab. Dispos. 32 (12), 1351–1358. doi:10.1124/dmd.104.000646

PubMed Abstract | CrossRef Full Text | Google Scholar

Jiang, W., Wang, X., Xu, X., and Kong, L. (2010). Effect of Schisandra Sphenanthera Extract on the Concentration of Tacrolimus in the Blood of Liver Transplant Patients. Int. J. Clin. Pharmacol. Ther. 48 (3), 224–229. doi:10.5414/cpp48224

CrossRef Full Text | Google Scholar

Jin, J., Bi, H., Hu, J., Zeng, H., Zhong, G., Zhao, L., et al. (2011). Effect of Wuzhi Tablet (Schisandra Sphenanthera Extract) on the Pharmacokinetics of Paclitaxel in Rats. Phytother. Res. 25 (8), 1250–1253. doi:10.1002/ptr.3407

PubMed Abstract | CrossRef Full Text | Google Scholar

Jin, J., Bi, H., Hu, J., Zhong, G., Zhao, L., Huang, Z., et al. (2010). Enhancement of Oral Bioavailability of Paclitaxel after Oral Administration of Schisandrol B in Rats. Biopharm. Drug Dispos. 31 (4), 264–268. doi:10.1002/bdd.705

PubMed Abstract | CrossRef Full Text | Google Scholar

Lai, L., Hao, H., Wang, Q., Zheng, C., Zhou, F., Liu, Y., et al. (2009). Effects of Short-Term and Long-Term Pretreatment of Schisandra Lignans on Regulating Hepatic and Intestinal CYP3A in Rats. Drug Metab. Dispos. 37 (12), 2399–2407. doi:10.1124/dmd.109.027433

PubMed Abstract | CrossRef Full Text | Google Scholar

Lai, Q., Wei, J., Mahmoodurrahman, M., Zhang, C., Quan, S., Li, T., et al. (2015). Pharmacokinetic and Nephroprotective Benefits of Using Schisandra Chinensis Extracts in a Cyclosporine A-Based Immune-Suppressive Regime. Drug Des. Devel. Ther. 9, 4997–5018. doi:10.2147/DDDT.S89876

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, H. J., and Kim, C. Y. (2010). Simultaneous Determination of Nine Lignans Using Pressurized Liquid Extraction and HPLC-DAD in the Fruits of Schisandra Chinensis. Food Chem. 120 (4), 1224–1228. doi:10.1016/j.foodchem.2009.11.068

CrossRef Full Text | Google Scholar

Li, C. L., Hsieh, C. H., and Tsai, T. H. (2020). Preclinical Pharmacokinetics of Lamivudine and its Interaction with Schisandra Chinensis Extract in Rats. ACS Omega 5 (4), 1997–2004. doi:10.1021/acsomega.9b03922

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, H. L., Ji, D., Mao, C. Q., Lu, T. L., and Wang, J. (2013). Rapid Determination of Three Lignans in the Fruits of Schisandra Chinensis by LC-MS Methods and Analysis of Their Pharmacokinetics in Rat Plasma. Chin. Pharmacol. Bull. 29 (9), 1313–1317. doi:10.3969/j.issn.1001-1978.2013.09.031

CrossRef Full Text | Google Scholar

Li, J., Chen, S., Qin, X., Fu, Q., Bi, H., Zhang, Y., et al. (2017). Wuzhi Tablet (Schisandra Sphenanthera Extract) Is a Promising Tacrolimus-Sparing Agent for Renal Transplant Recipients Who Are CYP3A5 Expressers: a Two-phase Prospective Study. Drug Metab. Dispos. 45 (11), 1114–1119. doi:10.1124/dmd.117.076737

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, L., Pan, Q., Sun, M., Lu, Q., and Hu, X. (2007). Dibenzocyclooctadiene Lignans: a Class of Novel Inhibitors of Multidrug Resistance-Associated Protein 1. Life Sci. 80 (8), 741–748. doi:10.1016/j.lfs.2006.11.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, R., Guo, W., Fu, Z., Ding, G., Wang, Z., and Fu, H. (2012). A Study about Drug Combination Therapy of Schisandra Sphenanthera Extract and Rapamycin in Healthy Subjects. Can. J. Physiol. Pharmacol. 90 (7), 941–945. doi:10.1139/y2012-080

CrossRef Full Text | Google Scholar

Li, W-L., Xin, H. W., and Su, M. W. (2012). Inhibitory Effects of Continuous Ingestion of Schisandrin A on CYP3A in the Rat. Basic Clin. Pharmacol. Toxicol. 110 (2), 187–192. doi:10.1111/j.1742-7843.2011.00787.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, W. L., Xin, H. W., Yu, A. R., and Wu, X. C. (2013). In Vivo effect of Schisandrin B on Cytochrome P450 Enzyme Activity. Phytomedicine 20 (8-9), 760–765. doi:10.1016/j.phymed.2013.02.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, W. L., Xin, H. W., and Su, M. W. (2010). Investigation on the Effect of Schisandrin B in Rat Liver Microsomal CYP3A Activity and its Inhibition Mechanism in. Vitro. Chin. Pharmacol. Bull. (Chinese) 26 (12), 1616–1621. CNKI: SUN:YAOL.0.2010-12-021.

Google Scholar

Li, Y. H., Yang, Y. H., and Liu, X. (2005). Study on Comparison between Shengmai Ultramicro Granule and Shengmai Powders. China J. Traditional Chin. Med. Pharm. (Chinese) 20, 588–590. doi:10.2217/nnm.14.169

CrossRef Full Text | Google Scholar

Li, Z., He, X., Liu, F., Wang, J., and Feng, J. (2018). A Review of Polysaccharides from Schisandra Chinensis and Schisandra Sphenanthera: Properties, Functions and Applications. Carbohydr. Polym. 184, 178–190. doi:10.1016/j.carbpol.2017.12.058

PubMed Abstract | CrossRef Full Text | Google Scholar

Liang, Y., Zhou, Y., Zhang, J., Liu, Y., Guan, T., Wang, Y., et al. (2013). In Vitro to In Vivo Evidence of the Inhibitor Characteristics of Schisandra Lignans toward P-Glycoprotein. Phytomedicine 20 (11), 1030–1038. doi:10.1016/j.phymed.2013.04.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Liang, Y., Zhou, Y., Zhang, J., Rao, T., Zhou, L., Xing, R., et al. (2014). Pharmacokinetic Compatibility of Ginsenosides and Schisandra Lignans in Shengmai-San: from the Perspective of P-Glycoprotein. PloS One 9 (6), e98717. doi:10.1371/journal.pone.0098717

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, G. T. (1988). Biochemical and Pharmacological Effects of Schisandra chinensis and its Components on Animal Liver. Prog. Physiol. Sci. (Chinese) 19 (3), 197–203.

Google Scholar

Liu, H. S., Zhang, Y. M., Wang, S. S., Shen, Z. Y., and Liu, H. L. (2015). Determination of Deoxyschizandrin, Schisandrin B, Schisandrin C, and Ursolic Acid in Baogan Pill by RP-HPLC. Chin. Traditional Herbal Drugs (Chinese) 46 (24), 3691–3694. doi:10.7501/j.issn.0253-2670.2015.24.014

CrossRef Full Text | Google Scholar

Liu, K., Song, Y., Liu, Y., Peng, M., Li, H., Li, X., et al. (2017). An Integrated Strategy Using UPLC-QTOF-MSE and UPLC-QTOF-MRM (Enhanced Target) for Pharmacokinetics Study of Wine Processed Schisandra Chinensis Fructus in Rats. J. Pharm. Biomed. Anal. 139, 165–178. doi:10.1016/j.jpba.2017.02.043

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, L. S., Wang, C. X., Qian, F. U., Chen, S. Y., Li, J. L., Li, J., et al. (2012). Effect of Wuzhi Tablet on Pharmacokinetics of Tacrolimus and Long-Term Clinical Outcome in Renal Transplant Patients. Organ. Transplant. (Chinese) 3 (1), 33–36. doi:10.1016/j.jep.2008.04.020

CrossRef Full Text | Google Scholar

Manikandan, P., and Nagini, S. (2018). Cytochrome P450 Structure, Function and Clinical Significance: a Review. Curr. Drug Targets 19 (1), 38–54. doi:10.2174/1389450118666170125144557

PubMed Abstract | CrossRef Full Text | Google Scholar

Mao, J., Tay, S., Khojasteh, C. S., Chen, Y., Hop, C. E. C. A., Kenny, J. R., et al. (2016). Revaluation of Time Dependent Inhibition Assays for Marketed Oncology Drugs: Comparison of Human Hepatocytes and Liver Microsomes in the Presence and Absence of Human Plasma. Pharm. Res. 33 (5), 1204–1219. doi:10.1007/s11095-016-1865-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Mu, Y., Zhang, J., Zhang, S., Zhou, H. H., Toma, D., Ren, S., et al. (2006). Traditional Chinese Medicines Wu Wei Zi (Schisandra Chinensis Baill) and Gan Cao (Glycyrrhiza Uralensis Fisch) Activate Pregnane X Receptor and Increase Warfarin Clearance in Rats. J. Pharmacol. Exp. Ther. 316 (3), 1369–1377. doi:10.1124/jpet.105.094342

PubMed Abstract | CrossRef Full Text | Google Scholar

Palleria, C., Roberti, R., Iannone, L. F., Tallarico, M., Barbieri, M. A., Vero, A., et al. (2020). Clinically Relevant Drug Interactions between Statins and Antidepressants. J. Clin. Pharm. Ther. 45 (2), 227–239. doi:10.1111/jcpt.13058

CrossRef Full Text | Google Scholar

Panossian, A., and Wikman, G. (2008). Pharmacology of Schisandra Chinensis Bail.: an Overview of Russian Research and Uses in Medicine. J. Ethnopharmacol. 118 (2), 183–212. doi:10.1016/j.jep.2008.04.020

PubMed Abstract | CrossRef Full Text | Google Scholar

Patocka, J., Wu, Q., Nepovimova, E., and Kuca, K. (2020). Phenytoin - an Anti-seizure Drug: Overview of its Chemistry, Pharmacology and Toxicology. Food Chem. Toxicol. 142, 111393. doi:10.1016/j.fct.2020.111393

PubMed Abstract | CrossRef Full Text | Google Scholar

Qian, T., Leong, P. K., Ko, K. M., and Chan, W. (2015). Investigation of In Vitro and In Vivo Metabolism of Schisandrin B from Schisandrae Fructus by Liquid Chromatography Coupled Electrospray Ionization Tandem Mass Spectrometry. Pp 06 (8), 363–373. doi:10.4236/pp.2015.68037

CrossRef Full Text | Google Scholar

Qin, X., and Wang, X. (2019). Role of Vitamin D Receptor in the Regulation of CYP3A Gene Expression. Acta Pharm. Sin. B 9 (6), 1087–1098. doi:10.1016/j.apsb.2019.03.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Qin, X. L., Bi, H. C., Wang, C. X., Li, J. L., Wang, X. D., Liu, L. S., et al. (2010b). Study of the Effect of Wuzhi Tablet (Schisandra Sphenanthera Extract) on Tacrolimus Tissue Distribution in Rat by Liquid Chromatography Tandem Mass Spectrometry Method. Biomed. Chromatogr. 24 (4), 399–405. doi:10.1002/bmc.1305

PubMed Abstract | CrossRef Full Text | Google Scholar

Qin, X. L., Bi, H. C., Wang, X. D., Li, J. L., Wang, Y., Xue, X. P., et al. (2010a). Mechanistic Understanding of the Different Effects of Wuzhi Tablet (Schisandra Sphenanthera Extract) on the Absorption and First-Pass Intestinal and Hepatic Metabolism of Tacrolimus (FK506). Int. J. Pharm. 389 (1-2), 114–121. doi:10.1016/j.ijpharm.2010.01.025

PubMed Abstract | CrossRef Full Text | Google Scholar

Qin, X. L., Chen, X., Wang, Y., Xue, X. P., Wang, Y., Li, J. L., et al. (2014a). In Vivo to In Vitro Effects of Six Bioactive Lignans of Wuzhi Tablet (Schisandra Sphenanthera Extract) on the CYP3A/P-Glycoprotein-Mediated Absorption and Metabolism of Tacrolimus. Drug Metab. Dispos. 42 (1), 193–199. doi:10.1124/dmd.113.053892

CrossRef Full Text | Google Scholar

Qin, X. L., Chen, X., Zhong, G. P., Fan, X. M., Wang, Y., Xue, X. P., et al. (2014b). Effect of Tacrolimus on the Pharmacokinetics of Bioactive Lignans of Wuzhi Tablet (Schisandra Sphenanthera Extract) and the Potential Roles of CYP3A and P-Gp. Phytomedicine 21 (5), 766–772. doi:10.1016/j.phymed.2013.12.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Qin, X. L., Li, J. L., Wang, S. H., Chen, X., Huang, M., and Bi, H. C. (2020). Co-administration of Wuzhi Tablet (Schisandra Sphenanthera Extract) Alters Tacrolimus Pharmacokinetics in a Dose- and Time-dependent Manner in Rats. J. Ethnopharmacol. 263, 113233. doi:10.1016/j.jep.2020.113233

CrossRef Full Text | Google Scholar

Qin, X. L., Yu, T., Li, L. J., Wang, Y., Gu, H. M., Wang, Y. T., et al. (2013). Effect of Long-Term Co-administration of Wuzhi Tablet (Schisandra Sphenanthera Extract) and Prednisone on the Pharmacokinetics of Tacrolimus. Phytomedicine 20 (3-4), 375–379. doi:10.1016/j.phymed.2012.11.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Qiu, B. X., Liu, K., Zou, L., Zhu, H. F., and Feng, S. (2018). Effects of Schisandrae Chinensis Fructus Induced CYPs and Nrf2 Activation on Acute Liver Injury Induced by Acetaminophen. Zhongguo Zhong Yao Za Zhi 43 (24), 4908–4915. doi:10.19540/j.cnki.cjcmm.20180625.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Ren, W., and Zhang, C. (2019). Research Progress of Schisandra chinensis and Prediction Analysis of Q-Maker. Chin. Trad. Herbal Drugs 51 (11), 3110–3116.

Google Scholar

Schutte-Nutgen, K., Tholking, G., Suwelack, B., and Reuter, S. (2018). Tacrolimus - Pharmacokinetic Considerations for Clinicians. Curr. Drug Metab. 19 (4), 342–350. doi:10.2174/1389200219666180101104159

PubMed Abstract | CrossRef Full Text | Google Scholar

Seo, H. J., Ji, S. B., Kim, S. E., Lee, G. M., Park, S. Y., Wu, Z., et al. (2021). Inhibitory Effects of Schisandra Lignans on Cytochrome P450s and Uridine 5'-Diphospho-Glucuronosyl Transferases in Human Liver Microsomes. Pharmaceutics 13 (3), 371. doi:10.3390/pharmaceutics13030371

PubMed Abstract | CrossRef Full Text | Google Scholar

Song, H. X. (2004). Studies on HPLC–MS Analysis on the Lignans of Fructus Schisandrae Chinensis and Clustering Analysis on the Chief Active Constituents of Chinese Herbal Medicine, Vol Master. Changsha: Hunan Normal University.

Google Scholar

Sowndhararajan, K., Deepa, P., Kim, M., Park, S. J., and Kim, S. (2018). An Overview of Neuroprotective and Cognitive Enhancement Properties of Lignans from Schisandra Chinensis. Biomed. Pharmacother. 97, 958–968. doi:10.1016/j.biopha.2017.10.145

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, Y. X., Cong, Y. L., Liu, Y., Jin, B., Si, L., Wang, A. B., et al. (2014). Schisandrin A and B Affect Subventricular Zone Neurogenesis in Mouse. Eur. J. Pharmacol. 740, 552–559. doi:10.1016/j.ejphar.2014.06.032

CrossRef Full Text | Google Scholar

Szopa, A., Ekiert, R., and Ekiert, H. (2017). Current Knowledge of Schisandra Chinensis (Turcz.) Baill. (Chinese magnolia Vine) as a Medicinal Plant Species: a Review on the Bioactive Components, Pharmacological Properties, Analytical and Biotechnological Studies. Phytochem. Rev. 16 (2), 195–218. doi:10.1007/s11101-016-9470-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Tian, F., Dou, D. Q., Cheng, D. R., Wang, L. N., Li, M., Qin, Y., et al. (2007). Comparison of Lignan Content in Crude and Processed Fructus. Schisandrae Chinensis. Mod. Chin. Med. (Chinese) 9 (9), 7–8. doi:10.3969/j.issn.1673-4890.2007.09.003

CrossRef Full Text | Google Scholar

Wan, C. K., Zhu, G. Y., Shen, X. L., Chattopadhyay, A., Dey, S., and Fong, W. F. (2006). Gomisin A Alters Substrate Interaction and Reverses P-Glycoprotein-Mediated Multidrug Resistance in HepG2-DR Cells. Biochem. Pharmacol. 72 (7), 824–837. doi:10.1016/j.bcp.2006.06.036

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, B., Yang, S., Hu, J., and Li, Y. (2014). Multifaceted Interaction of the Traditional Chinese Medicinal Herb Schisandra Chinensis with Cytochrome P450-Mediated Drug Metabolism in Rats. J. Ethnopharmacol. 155 (3), 1473–1482. doi:10.1016/j.jep.2014.07.026

CrossRef Full Text | Google Scholar

Wang, B., Hu, J., Sheng, L., and Li, Y. (2011). The Effects of Schisandra Chinensis on the Isozyme of Cytochrome P450 in Rat Liver Determined by Mixed Probe Substrate Method. Acta Pharmaceutica Sinica (Chinese) 46 (8), 922–927.

Google Scholar

Wang, C-P., Li, G. C., Shi, Y. W., Zhang, X. C., Li, J. L., Wang, Z. W., et al. (2014). Neuroprotective Effect of Schizandrin A on Oxygen and Glucose Deprivation/reperfusion-Induced Cell Injury in Primary Culture of Rat Cortical Neurons. J. Physiol. Biochem. 70 (3), 735–747. doi:10.1007/s13105-014-0342-3

CrossRef Full Text | Google Scholar

Wang, L. J., Liu, S. M., Mao, C. Q., Tu-Lin, L. U., Zhou, Y., Xiao-Nan, S. U., et al. (2015). Simultaneous Determination of 8 Lignans in Schisandra Chinensis by Quantitative Analysis of Multi-Components by Single Marker. Chin. J. Pharm. Anal. (Chinese) 35 (7), 1191–1197.

Google Scholar

Wang, S., Zhao, H., Huangpu, M., and Li, X. (2003). Pharmacokinetic Effect of the Combination of Fructus Schisandrae and Radix Salvia Miltiorrhizae on Schizandrol A and Schizandrin B. Chin. Traditional Patent Med. (Chinese) 26 (12), 987–991.

Google Scholar

Wang, W., Zhu, S., Guo, W., Fei, T., Wei, H., and Chen, W. (2016). Simultaneous Determination of Tacrolimus and Five Main Compounds of Wuzhi Capsule in Liver Transplant Patients' Whole Blood by LC-MS/MS. Chin. J. Mod. Appl. Pharm. (Chinese) 33 (7), 854–859. doi:10.13748/j.cnki.issn1007-7693.2016.07.005

CrossRef Full Text | Google Scholar

Wang, Y. C., Ren, K., Shen, N., Huang, X. D., and Fan, H. Y. (2013). Improvement of Impaired Memory in Mice by Schisandrin. Amr 750-752, 1533–1538. doi:10.4028/www.scientific.net/amr.750-752.1533

CrossRef Full Text | Google Scholar

Wang, Z., Jin, L., Ma, Y., Cui, Z., Li, Q., and Huang, D. (2017). Quality Evaluation of Schisandra Sphenanthera Based on the Content of Gomisin C. China J. Chin. Materia Med. (Chinese) 42 (19), 3728–3732.

Google Scholar

Wei, B., Li, Q., Fan, R., Su, D., Ou, X., Chen, K., et al. (2013). UFLC-MS/MS Method for Simultaneous Determination of Six Lignans of Schisandra Chinensis (Turcz.) Baill. In normal and Insomniac Rats Brain Microdialysates and Homogenate Samples: towards an In-Depth Study for its Sedative-Hypnotic Activity. J. Mass. Spectrom. 48 (4), 448–458. doi:10.1002/jms.3176

PubMed Abstract | CrossRef Full Text | Google Scholar

Wei, B. B., Chen, X. H., Su, D., Qian, Z. Z., Yuan, R. H., and Bi, K. S. (2010). Pharmacokinetics of Deoxyschizandrin and γ-Schizandrin in Schisandra Chinensis Oil in Rat Plasma. Chin. J. Hosp. Pharm. (Chinese) 9 012.

Google Scholar

Wei, H., Miao, H., Yun, Y., Li, J., Qian, X., Wu, R., et al. (2014). Validation of an LC-MS/MS Method for Quantitative Analysis of the 5 Bioactive Components of Wuzhi Capsule in Human Plasma Samples. Ther. Drug Monit. 36 (6), 781–788. doi:10.1097/FTD.0000000000000079

PubMed Abstract | CrossRef Full Text | Google Scholar

Wei, H. (2010). Study on the Pharmacokinetics of Wuzhi-Capsule and its Herb-Drug Interaction with Tacrolimus, Vol Doctor. Shanghai: Second Military Medical University.

Google Scholar

Wei, H., Sun, L., Tai, Z., Gao, S., Xu, W., and Chen, W. (2010a). A Simple and Sensitive HPLC Method for the Simultaneous Determination of Eight Bioactive Components and Fingerprint Analysis of Schisandra Sphenanthera. Anal. Chim. Acta 662 (1), 97–104. doi:10.1016/j.aca.2009.12.039

PubMed Abstract | CrossRef Full Text | Google Scholar

Wei, H., Tao, X., Di, P., Yang, Y., Li, J., Qian, X., et al. (2013). Effects of Traditional Chinese Medicine Wuzhi Capsule on Pharmacokinetics of Tacrolimus in Rats. Drug Metab. Dispos. 41 (7), 1398–1403. doi:10.1124/dmd.112.050302

PubMed Abstract | CrossRef Full Text | Google Scholar

Wei, H., Xu, W., Cai, F., Zhao, G., Feng, J., Sun, L., et al. (2010b). Simultaneous Determination of Five Lignan Constituents of Wuzhi Capsule in Rat Plasma by LC-MS/MS: Application to Pharmacokinetic Study. J. Pharm. Biomed. Anal. 52 (5), 741–746. doi:10.1016/j.jpba.2010.02.003

CrossRef Full Text | Google Scholar

Wu, J., Cao, Y., Zhang, Y., Liu, Y., Hong, J. Y., Zhu, L., et al. (2014). Deoxyschizandrin, a Naturally Occurring Lignan, Is a Specific Probe Substrate of Human Cytochrome P450 3A. Drug Metab. Dispos. 42 (1), 94–104. doi:10.1124/dmd.113.053884

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, X., Zhou, Y., Yin, F., Dai, G., Li, L., Xu, B., et al. (2014a). Comparative Pharmacokinetics and Tissue Distribution of Schisandrin, Deoxyschisandrin and Schisandrin B in Rats after Combining Acupuncture and Herb Medicine (Schisandra Chinensis). Biomed. Chromatogr. 28 (8), 1075–1083. doi:10.1002/bmc.3122

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, Y. J., Cheng, N. N., Hu, Z. H., and Chen, B. Y. (2007a). Effects of Schisandra Chinensis Baill and Lignans on Enzyme Activity of Hepatic CYP3A. Chin. J. Clin. Pharm. (Chinese) 5, 267–271. doi:10.3969/j.issn.1007-4406.2007.05.001

CrossRef Full Text | Google Scholar

Xia, Y.-G., Yang, B.-Y., Liang, J., Wang, J.-S., and Kuang, H.-X. (2014a). Simultaneous Quantification of Five Dibenzocyclooctadiene Lignans in Schisandra Chinensis by HPLC Separation and Fluorescence Detection. Anal. Methods 6 (15), 5981–5985. doi:10.1039/c4ay00017j

CrossRef Full Text | Google Scholar

Xia, Y. G., Yang, B. Y., Liang, J., Yang, Q., Wang, D., and Kuang, H. X. (2014b). Quantitative Analysis and Fingerprint Profiles for Quality Control of Fructus Schisandrae by Gas Chromatography: Mass Spectrometry. ScientificWorldJournal 2014, 806759–806768. doi:10.1155/2014/806759

PubMed Abstract | CrossRef Full Text | Google Scholar

Xin, H. W., Li, Q., Wu, X. C., He, Y., Yu, A. R., Xiong, L., et al. (2011). Effects of Schisandra Sphenanthera Extract on the Blood Concentration of Tacrolimus in Renal Transplant Recipients. Eur. J. Clin. Pharmacol. 67 (12), 1309–1311. doi:10.1007/s00228-011-1075-7

CrossRef Full Text | Google Scholar

Xin, H. W., Wu, X. C., Li, Q., Yu, A. R., Zhu, M., Shen, Y., et al. (2007). Effects of Schisandra Sphenanthera Extract on the Pharmacokinetics of Tacrolimus in Healthy Volunteers. Br. J. Clin. Pharmacol. 64 (4), 469–475. doi:10.1111/j.1365-2125.2007.02922.x

CrossRef Full Text | Google Scholar

Xue, X. P., Qin, X. L., Xu, C., Zhong, G. P., Wang, Y., Huang, M., et al. (2013). Effect of Wuzhi Tablet (Schisandra Sphenanthera Extract) on the Pharmacokinetics of Cyclosporin A in Rats. Phytother Res. 27 (8), 1255–1259. doi:10.1002/ptr.4849

PubMed Abstract | CrossRef Full Text | Google Scholar

Yan, J., Ma, Y., Wang, T., and Ji, L. (2006). Pharmacokinetics Study of Schisandrin in Shengmai Granule. Traditional Chin. Drug Res. Clin. Pharmacol. (Chinese) 1 012.

Google Scholar

Yang, G. (2011). The Key Technique Research of the High Quality and High Yield of Schisandra Chinensis, Vol Master. Haerbin: Heilongjiang University of Chinese Medicine.

Google Scholar

Yang, J., Duan, J. A., Li, G. L., Zhu, Z. H., Zhu, T. L., Qian, D. W., et al. (2014). Determination of Lignans in Schisandrae Sphenantherae Fructus from Different Regions. Zhongguo Zhong Yao Za Zhi 39 (23), 4647–4652.

PubMed Abstract | Google Scholar

Yang, K., Qiu, J., Huang, Z., Yu, Z., Wang, W., Hu, H., et al. (2022). A comprehensive review of ethnopharmacology, phytochemistry, pharmacology, and pharmacokinetics of Schisandra chinensis (Turcz.) Baill. and Schisandra sphenanthera Rehd. et Wils. J. Ethnopharmacol. 284, 114759. doi:10.1016/j.jep.2021.114759

CrossRef Full Text | Google Scholar

Yoo, H. H., Lee, M., Lee, M. W., Lim, S. Y., Shin, J., and Kim, D. H. (2007). Effects of Schisandra Lignans on P-Glycoprotein-Mediated Drug Efflux in Human Intestinal Caco-2. Planta Med. 73 (5), 444–450. doi:10.1055/s-2007-967178

PubMed Abstract | CrossRef Full Text | Google Scholar

Yuan, G., Liu, Y., Li, T., Wang, Y., Sheng, Y., and Guan, M. (2011). Simultaneous and Rapid Determination of Main Lignans in Different Parts of Schisandra Sphenanthera by Micellar Electrokinetic Capillary Chromatography. Molecules 16 (5), 3713–3722. doi:10.3390/molecules16053713

PubMed Abstract | CrossRef Full Text | Google Scholar

Zeng, H., Jiang, Y., Chen, P., Fan, X., Li, D., Liu, A., et al. (2017). Schisandrol B Protects against Cholestatic Liver Injury through Pregnane X Receptors. Br. J. Pharmacol. 174 (8), 672–688. doi:10.1111/bph.13729

CrossRef Full Text | Google Scholar

Zhai, J., Zhang, F., Gao, S., Chen, L., Feng, G., Yin, J., et al. (2018). Schisandra Chinensis Extract Decreases Chloroacetaldehyde Production in Rats and Attenuates Cyclophosphamide Toxicity in Liver, Kidney and Brain. J. Ethnopharmacol. 210, 223–231. doi:10.1016/j.jep.2017.08.020

CrossRef Full Text | Google Scholar

Zhai, J., Zhang, F., Gao, S., Chen, L., Feng, G., Yin, J., et al. (2017). Time- and NADPH-dependent Inhibition on CYP3A by Gomisin A and the Pharmacokinetic Interactions between Gomisin A and Cyclophosphamide in Rats. Molecules 22 (8), 1298. doi:10.3390/molecules22081298

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhai, J. X., Liu, Z. H., Han, N., and Yin, J. (2015). The Study of the CYP-Interfering Effects of Schisandra Chinensis and its Mechanism. Modernization Traditional Chin. Med. Materia Materia-World Sci. Tech. (Chinese) 1, 52–55. doi:10.11842/wst.2015.01.008

CrossRef Full Text | Google Scholar

Zhan, S., Guo, W., Shao, Q., Fan, X., Li, Z., and Cheng, Y. (2014). A Pharmacokinetic and Pharmacodynamic Study of Drug-Drug Interaction between Ginsenoside Rg1, Ginsenoside Rb1 and Schizandrin after Intravenous Administration to Rats. J. Ethnopharmacol. 152 (2), 333–339. doi:10.1016/j.jep.2014.01.014

CrossRef Full Text | Google Scholar

Zhang, H., Jiang, Y., Wu, J., Zheng, C., Ran, X., Li, D., et al. (2017b). Metabolic Mapping of Schisandra Sphenanthera Extract and its Active Lignans Using a Metabolomic Approach Based on Ultra High Performance Liquid Chromatography with High-Resolution Mass Spectrometry. J. Sep. Sci. 40 (2), 574–586. doi:10.1002/jssc.201600954

CrossRef Full Text | Google Scholar

Zhang, H., Bu, F., Li, L., Jiao, Z., Ma, G., Cai, W., et al. (2017a). Prediction of Drug-Drug Interaction between Tacrolimus and Principal Ingredients of Wuzhi Capsule in Chinese Healthy Volunteers Using Physiologically-Based Pharmacokinetic Modelling. Basic Clin. Pharmacol. Toxicol. 122, 331–340. doi:10.1111/bcpt.12914

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, H., Zhang, G., Zhu, Z., Zhao, L., Fei, Y., Jing, J., et al. (2009). Determination of Six Lignans in Schisandra Chinensis (Turcz.) Baill. Fruits and Related Chinese Multiherb Remedies by HPLC. Food Chem. 115 (2), 735–739. doi:10.1016/j.foodchem.2008.12.010

CrossRef Full Text | Google Scholar

Zhang, H., Zhou, Y.-n., Sun, S., Sun, F.-f., Chai, Y.-f., and Zhang, G.-q. (2014). Metabolic Rates and Metabolites of Three Lignans in Schisandra Chinensis (Turcz.) Baill in Rat Liver Microsomes. Acad. J. Second Mil. Med. Univ. 35 (4), 426–431. doi:10.3724/sp.j.1008.2014.00426

CrossRef Full Text | Google Scholar

Zhang, W., Zhu, Y. D., Zhang, Q. Y., Ma, L. J., Yang, L., Guo, W. Z., et al. (2020). Research Progress in Application and Mechanism of Schisandrae Chinensis Fructus for Prevention and Treatment of Liver Diseases. Zhongguo Zhong Yao Za Zhi 45 (16), 3759–3769. doi:10.19540/j.cnki.cjcmm.20200513.601

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, W., Wang, Y., and Zou, G. (2009). Study on Gastrointestinal Dynamics of Schisandrin. J. Liaoning Univ. Traditional Chin. Med. (Chinese) 36, 1965–1966.

Google Scholar

Zhao, H. Q., Zhao, R. S., and Zhang, C. (2006). Pharmacokinetic Studies of Schisandrin in Rat Plasma. Chin. J. Pharm. Anal. (Chinese) 26, 605–608.

Google Scholar

Zhou, H., Zhang, J., Wu, S., Huang, Y., Shi, S., Zhang, Y., et al. (2017). Effects of Wuzhi Capsules on Tacrolimus Trough Concentration in Heart Transplant Recipients. Herald Med. (Chinese) 36 (2), 158–162. doi:10.1080/00926230290001565

CrossRef Full Text | Google Scholar

Zhou, S., Feng, X., Kestell, P., Paxton, J. W., Baguley, B. C., and Chan, E. (2005). Transport of the Investigational Anti-cancer Drug 5,6-Dimethylxanthenone-4-Acetic Acid and its Acyl Glucuronide by Human Intestinal Caco-2 Cells. Eur. J. Pharm. Sci. 24 (5), 513–524. doi:10.1016/j.ejps.2005.01.006

CrossRef Full Text | Google Scholar

Zhou, Y., Mao, C. Q., and Hu, J. Y. (2013). Tissue Distribution of Schisandrin, Deoxyschizandrin and Schisandrin B in Rat Determined by HPLC-MS. Chin. J. Pharm. Anal. (Chinese) 33, 1121–1126. doi:10.1056/NEJMoa1307361

CrossRef Full Text | Google Scholar

Zhu, M., Chen, X.-s., and Wang, K.-x. (2007). Variation of the lignan content of Schisandra chinensis (Turcz.) Baill. and Schisandra sphenanthera Rehd. et Wils. Chroma 66 (1-2), 125–128. doi:10.1365/s10337-007-0261-3

CrossRef Full Text | Google Scholar

Zuo, X. B., Sun, X. M., Wang, C. Y., Liu, J., Xiao, X. H., and Bai, Z. F. (2019). China J. Chin. Materia Med. (Chinese) 44 (6), 1238–1245. doi:10.19540/j.cnki.cjcmm.20181210.001

CrossRef Full Text

Keywords: Schisandra sphenanthera, Schisandraceae, lignans, drug-drug interactions, ADME, Schisandra chinensis

Citation: Zhang F, Zhai J, Weng N, Gao J, Yin J and Chen W (2022) A Comprehensive Review of the Main Lignan Components of Schisandra chinensis (North Wu Wei Zi) and Schisandra sphenanthera (South Wu Wei Zi) and the Lignan-Induced Drug-Drug Interactions Based on the Inhibition of Cytochrome P450 and P-Glycoprotein Activities. Front. Pharmacol. 13:816036. doi: 10.3389/fphar.2022.816036

Received: 16 November 2021; Accepted: 14 February 2022;
Published: 11 March 2022.

Edited by:

Yan Liu, Heilongjiang University of Chinese Medicine, China

Reviewed by:

Xiao Chen, China Pharmaceutical University, China
Subrata Deb, Larkin University, United States

Copyright © 2022 Zhang, Zhai, Weng, Gao, Yin and Chen. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Wansheng Chen, chenwansheng@smmu.edu.cn; Jun Yin, yinjun826@sina.com

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.