Skip to main content
Saudi Pharmaceutical Journal : SPJ logoLink to Saudi Pharmaceutical Journal : SPJ
. 2018 Jun 1;26(8):1069–1072. doi: 10.1016/j.jsps.2018.05.016

Novel plant inducers of PXR-dependent cytochrome P450 3A4 expression in HepG2 cells

Mohammed S Al-Dosari 1,, Mohammad K Parvez 1,
PMCID: PMC6260467  PMID: 30532626

Abstract

The cytochrome P450 3A4 (CYP3A4) is the most abundant CYP450 enzyme involved in the metabolism of endogenous products and xenobiotics, including prescription drugs and herbals. Modulation of hepatic CYP3A4 gene expression via nuclear receptors, like pregnane X receptor (PXR), is a major cause of adverse effects like drug-unresponsiveness and toxicity. In the present study, ethanol extracts of 58 medicinal plants, belonging to 27 families, were evaluated for potential activities in CYP3A4 induction in HepG2 cells by reporter gene assay. For PXR-mediated CYP3A4 induction, a 50 μg/ml concentration was used for all non-cytotoxic plants extracts. Rifampicin (10 μM) and DMSO (0.1%) were used as standard inducer and untreated (negative) control, respectively. The comparative fold-induction of CYP34A by the plant extracts in relation to the untreated control was determined. As a result, Dodonaea angustifolia (2.62 fold; P < 0.0001) was found to be the most promising inducer of CYP3A4, followed by Euphorbia tirucalli (1.95 fold; P = 0.0004), Alternanthera pungens (1.74 fold, P = 0.0035), and Ficus palmata (1.65 fold; P = 0.0097). Further phytochemical characterizations of the active plants are therefore, warranted.

Keywords: Pregnane X receptor, Cytochrome P450, CYP3A4, Plant extracts, Luciferase

1. Introduction

Cytochrome P450 (CYP) is a superfamily of drug-metabolizing enzymes that is involved in the metabolism of endogenous compounds, xenobiotics and pharmaceuticals (Anzenbacher and Anzenbacherová, 2001). The major CYP involved in the hepatic metabolism of most of the drugs include CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4 (Anzenbacher and Anzenbacherová, 2001, Al-Dosari and Parvez, 2016). Since CYP is involved in phase-1 metabolism of >70% of prescription drugs, modulation of its expressions is a major cause of adverse drug-drug interactions, including decreased drug efficacy (Tyagi et al., 2010, Zhang et al., 2010). Expression of CYP3A4 is markedly induced both in vivo and in cultured human hepatocytes in vitro in response to a variety of xenobiotics (eg., dexamethasone and rifampicin) as well as medicinal herbs like, St. John’s wort (Hypericum perforatum) (Kolars et al., 1992, Schuetz and Guzelian, 1984, Zhou and Lai, 2008, Harmsen et al., 2008, Quatrochi and Guzelian, 2001, Moore et al., 2000).

Further, gene expression of CYP is regulated by a set of nuclear receptors in response to a wide spectrum of xenobiotics (Pelkonen et al., 1998, Quatrochi and Guzelian, 2001, Al-Dosari and Parvez, 2016). Of these, the pregnane X receptor (PXR), also known as xenobiotic or pregnane-activated receptor regulates CYP3A4 gene induction (Matic et al., 2007, Hustert et al., 2001). The most common clinical implication for the PXR activation is the drug-drug interactions, mediated by the upregulation of CYP3A4. Notably, PXR itself is activated by rifampicin and other xenobiotics (Lehmann et al., 1998) as well as plant secondary metabolites like, hyperforin from H. perforatum (Zhou and Lai, 2008, Moore et al., 2000). In addition to this, PXR and its target genes also play an important role in maintaining normal physiological function and homeostasis. For example, artemisinin (Artemisia annua), piperine (Piper nigrum) and notoginsenoside (Panax notoginseng), the known activators of PXR, have been shown to prevent severity of colonic inflammatory bowel disease by inducing CYP3A4 expression (Hu et al., 2015, Hu et al., 2014, Zhang et al., 2015).

Hepatic PXR has broad substrate specificity and thus may be activated by a large number of chemically-diverse secondary metabolites found in dietary supplements and therapeutic herbs. Since such natural products are often orally consumed, the high concentration of their phytoconstituents in gut and liver may potentially affect the CYP activity. In this report, we therefore, intended to screen the novel PXR-dependent CYP3A4 activation potential of 58 medicinal plants of 27 families using cultured hepatocytes and reporter gene assay.

2. Experimental methods

2.1. Plant materials and extraction

The studied plant extracts included pre-identified, non-cytotoxic medicinal plants (Table 1) with their traditionally known or published therapeutic values (Arbab et al., 2017). The dried plant parts were ground to a coarse powder using mortar-pestle and extracted with 80% ethanol (Merck, Germany) for three days with periodic shaking and filtered using Whatman No. 1 paper (Sigma, Germany). After removal of the solvent under reduced pressure using rotary evaporator (4 °C) and complete drying, their yield percentage were calculated. Stock of each extract (100 mg/ml) was prepared by dissolving in dimethyl sulfoxide (DMSO, Sigma, USA), and stored at -20 °C.

Table 1.

List of medicinal plants (n = 58) screened for PXR-mediated CYP3A4 induction activity.

No. Specimen no. Plants name Family Plant part used
1 16011 Achyranthe aspera Amaranthaceae Shoots
2 16391 Alternanthera pungens Shoots
3 16189 Amaranthus alba Shoots
4 16196 Avera Javanica Shoots
5 16198 Flaveria trineriva Asteraceae Shoots
6 16083 Pulicaria crispa Shoots
7 16075 Pergularia tomentosa Asclepiadaceae Shoots
8 16318 Eruca sativa Brassicaceae Leaves, Stems
9 15841 Capparis decidua Capparaceae Stems
10 16195 Atriplex suberecta Chenopodiaceae Shoots
11 15496 Combretum molle Combretaceae Bark
12 798 Guiera senegalensis Leaves
13 16075 Ipomoea cairica Convolvulaceae Shoots
14 16179 Juniperus phonicea Cupressaceae Leaves, Stems
15 16194 Juniperus procera Leaves, Stems
16 15830 Cleome droserifolia Crassulaceae Shoots
17 16275 Coccinia grandis Cucurbitaceae Leaves, Stems
18 16393 Corallocarpus epigeus Leaves
19 16395 Momordica balsamina Leaves
20 16181 Chenopodium ambrosioides Arial parts
21 16197 Chenopodium glaucum Leaves, Stem
22 16172 Euphorbia tirucalli Euphorbiaceae Stems
23 16084 Euphorbia hirta Shoots
24 15189 Jatropha curcas Seeds
25 14005 Ricinus communis Leaves
26 16281 Acacia mellifera Fabaceae Leaves
27 16221 Acacia hamulosa Leaves, Stems
28 16387 Acacia asak Leaves
29 16385 Acacia ehrenbergiana Stems
30 16390 Acacia laeta Stems
31 16389 Acacia oerfota Stems
32 15007 Acacia salicina Leaves
33 14977 Acacia tortilis Stems
34 16182 Albizia procera Leaves
35 16035 Delonix elata Leaves
36 16183 Delonix regia Leaves
37 16392 Indigofera coerulea Shoots
38 16390 Indigofera tinctoria Shoots
39 160322 Senna obtusifolia Fruits
40 155009 Senna occidentalis Fruits
41 16245 Senna alexandrina Leaves
42 16301 Fumaria parviflora Fumariaceae Leaves, Stems
43 16043 Marrubium vulgare Labiatae Shoots
44 15716 Cassytha filiformis Lauraceae Stems
45 16082 Abutilon figarianum Malvaceae Leaves
46 16080 Ficus benghalensis Moraceae Leaves, Stems
47 15448 Ficus palmata Leaves
48 16085 Psidium guajava Myrtaceae Leaves
49 16184 Boerhavia diffusa Nyctaginaceae Leaves
50 16177 Bougainvillea spectabilis Leaves
51 16185 Argemone ochroleuca Papaveraceae Shoots
52 16186 Rumex dentatus Polygonaceae Shoots
53 16173 Citrus maxima Rutaceae Leaves
54 15787 Dodonea angustifolia Sapindaceae Leaves
55 15604 Daturai noxia Solanaceae Leaves
56 16386 Solanum surrattense Leaves
57 12788 Clerodendrum inerme Verbenaceae Leaves, Stems
58 560 Balanites aegyptiaca Zygophyllaceae Bark

2.2. Cell culture and reagents

Human hepatoblastoma cell line, HepG2 (López-Terrada et al., 2009) was maintained in T75 culture flask (Corning, USA) in RPMI-1640 medium (Gibco, Thermo Fisher Scientific, Inc., MA, USA), supplemented with 10% heat-inactivated bovine serum (Gibco, MA, USA), 1× penicillin-streptomycin (Gibco, MA, USA) and 1× sodium pyruvate (GE Healthcare Life Sci., UT, USA) in an incubator at 37 °C with 5% CO2 supply. Dimethyl sulphoxide (DMSO; Sigma, Germany) was used as carrier to prepare stocks of plants extracts or compounds as well as negative control. Rifampicin (Sigma, Germany) was used as standard PXR-mediated CYP3A4 inducer or positive control.

2.3. Plasmid DNA preparations

The nuclear receptor expression vector pCDG-hPXR and CYP3A4 firefly-luciferase reporter construct pGL3-CYP3A4-XREM were kind gifts from Dr. Ron Evans (The Salk Institute for Biological Studies, La Jolla, USA) and Dr. Richard Kim (Department of Physiology and Pharmacology, University of Western Ontario, London, Canada), respectively. The renilla-luciferase expression plasmid (pRL-TK; Promega, USA) served as internal control. All plasmid DNA were transformed into DH5α XL competent cells (Invitrogen, USA) by the heat-shock method and plated on ampicillin (50 μg/ml) containing agar plates. Following an overnight incubation at 37 °C, bacterial colonies were picked and plasmids (Qiagen Plasmid Mini-prep Kit, Germany) were screened by restriction digestion. Further, DNA stocks were prepared (Qiagen Plasmid Maxi-prep Kit, Germany), quantified (Nanodrop 3300) and stored at −20 °C.

2.4. Transient transfection

HepG2 cells were seeded in 24-well culture plates (Corning, USA) and incubated overnight to reach up to 60–70% confluency. Next day, cells were co-transfected with pGL3-CYP3A4-XREM (CYP, 400 ng), pCDG-hPXR (PXR, 400 ng) and pRL-TK (200 ng), using transfection reagent FuGENE6 (Promega, USA) per well. A mock transfection control (negative, without plasmid) was also included. For a 24-well plate (200 μl media/well), the amount of FuGENE6 to DNA per well (3:1) was optimized as per the FuGENE6 manual. After 24 h, the medium was removed and 200 μl/well of fresh medium containing DMSO (0.1%) or rifampicin (10 μM) or plant extracts (50 μg/ml) was added. The treated cells were further incubated for 24 h at 37 °C. The cells were transfected in triplicate for all samples, including controls.

2.5. Luciferase reporter gene assay

After 24 h of treatment (48 h post-transfection), the reporter activities of firefly-luciferase and renilla-luciferase were measured with the Dual-Luciferase Reporter Assay System (Promega, USA) according to the manufacturer’s manual. Briefly, reagents were brought to room temperature (RT), and reconstituted. Meanwhile, media were discarded and cells were carefully washed with 1× PBS (200 μl/well). The passive lysis buffer (1× PLB; 60 μl/well) was added and cells were allowed to lyse for 15 min at RT by gentle rocking (Heidolph DuoMax 1030, Heidolph instruments, GmBH, Germany). Total cell lysates were properly mixed and carefully harvested into pre-labeled 1.5 ml Eppendorf tubes. Lysates were quickly cleared at 10000 rpm for 30 sec (Eppendorf 5415D, USA) and placed in ice bath. The assay was instantly performed in round bottom high-clarity polypropylene tubes (5 ml; Falcon, USA) with 100 μl Luciferase Assay Reagent II or Stop & Glo Reagent and 10 μl lysate, using an illuminometer (Berthhold Lumat LB9507, Berthhold Technologies, USA). The assay was performed in triplicate for all samples and repeated. The firefly-luciferase signal was normalized to renilla-luciferase signal for each sample. To calculate fold-inductions, the ratios of all tested extracts were compared with normalized signals of the control. To determine the potency of the active extracts, concentrations <50 μg/ml were also tested. All tests were performed in triplicate and repeated twice. Data was analyzed and represented as bar graph (Excel 2010; Microsoft, OK, USA).

2.6. Statistical analysis

All experiments were performed in triplicate and data were presented as the mean ± standard error, and were analyzed by One Way ANOVA using GraphPad Prism 7.04. The statistical differences between the control (CYP + PXR only) and treatment groups were carried out using Dunnett’s Test (P value <0.05).

3. Results

3.1. PXR-dependent CYP3A4 induction by plant extracts

For PXR-dependent CYP3A4 induction, a 50 μg/ml concentration was used for all non-cytotoxic plants ethanol extracts. Of the 58 plants extracts screened, four showed induction of PXR-mediated CYP3A4 expression. The comparative fold-induction of CYP34A by the plant extracts in relation to the untreated (DMSO) control was determined (Fig. 1). The induction activities of the tested plants were in the order, Dodonaea angustifolia (2.62 fold, P < 0.0001), Euphorbia tirucalli (1.95 fold, P = 0.0004), Alternanthera pungens (1.74 fold, P = 0.0035), Ficus palmata (1.65 fold, P = 0.0097); and the rifampicin mediated fold of induction was 1.42 (P = 0.1209).

Fig. 1.

Fig. 1

Reporter gene assay, showing fold-activation of PXR-mediated CYP3A4 expression in HepG2 cells upon treatment with rifampicin (10 μM) and different plant extracts (50 μg/ml). Data are presented as the mean ± standard error (n = 3). **P < 0.01, ***P < 0.001 vs. CYP + PXR only (DMSO control) group. PXR: pCDG-hPXR; CYP: pGL3-CYP3A4-XREM; F. palmate: Ficus palmate; A. pungens: Alternanthera pungens; E. tirucalli: Euphorbia tirucalli; D. angustifolia: Dodonaea angustifolia.

4. Discussion

The PXR-mediated CYP3A4 expression is markedly induced in cultured human hepatocytes in response to a variety of xenobiotics and drugs, including some bioactive plant products. In this study, we have therefore, screened ethanol extracts of 58 medicinal plants using HepG2 cell culture and dual-luciferase assay for their PXR-mediated CYP3A4 activation potential. All the non-toxic extracts were tested at the safe concentration 50 μg/ml as compared to a similar study where extracts at 100 μg/ml doses were used (Mooiman et al., 2013).

Dodonaea angustifolia (sand olive) occurs naturally in Arabia and southern Africa, including Australia and New Zealand. An important traditional medicine of Africa, its leaves decoction is used for fever, colds, flu, stomachache, measles, tuberculosis and skin rashes (van Heerden et al., 2000). However, very limited studies have been published on Dodonaea angustifolia and none on its chemical constituents compared to its other species. A single study has reported presence of flavonoids, reducing sugars, alkaloids, saponins and tannin in its leaves (Amabeoku et al., 2001). Here, we have for the first time, demonstrated its very promising activation potential of PXR-mediated CYP3A4 expression in HepG2 cells.

Euphorbia tirucalli (firestick or milk bush) has a wide distribution in Arabia and Africa, including many other tropical regions. It is a hydrocarbon plant that produces a poisonous latex, possibly convertible to biofuel (Hastilestari et al., 2013). It is used in traditional medicine for cancer, excrescence, tumors, warts, asthma, cough, earache, neuralgia and rheumatism (Duke, 1983). In this study, we report a novel PXR-mediated CYP3A4 activation property of Euphorbia tirucalli in HepG2 cells.

Alternanthera pungens (Kunth) is a ruderal plant of roadsides, path verges and waste places. Though a native of South America, it is also reported from other tropical countries including India (Jakhar and Dahiya, 2017). Compared to its other species, Alternanthera pungens is poorly studied. In a very recent study, its crude extract is shown to have a wide spectrum antibacterial activity as well as good antioxidant potential (Jakhar and Dahiya, 2017). Here, we have demonstrated the novel PXR-mediated CYP3A4 activation potential Alternanthera pungens in HepG2 cells.

Ficus palmata (Fegra or Wild Himalayan Fig) occurs in North West India, Afghanistan, Iran, Arabia and Africa (Joshi et al., 2014). It is used as hypoglycemic, anti-tumour, anti-ulcer, anti-diabetic, lipid lowering and antifungal remedy, including nephro-hepatoprotective effect (Joshi et al., 2014). In this report, we have shown a novel PXR-mediated CYP3A4 inducing activity of Ficus palmata in HepG2 cells.

5. Conclusion

Our screening of ethanol extracts of 58 medicinal plants using HepG2 cells and reporter gene assay, has demonstrated the novel PXR-mediated CYP3A4 gene induction potential of four plants. Of these, Dodonaea angustifolia was found to be the most promising CYP3A4 activator, followed by Euphorbia tirucalli, Alternanthera pungens, and Ficus palmata. Further phytochemical characterizations, including isolation of active principles are therefore, warranted.

Footnotes

Peer review under responsibility of King Saud University.

Contributor Information

Mohammed S. Al-Dosari, Email: mdosari@ksu.edu.

Mohammad K. Parvez, Email: mohkhalid@ksu.edu.sa.

References

  1. Amabeoku G.J., Eagles P., Scott G., Mayeng I., Springfield E. Analgesic and antipyretic effects of Dodonaea angustifolia and Salvia africana-lutea. J. Ethnopharmacol. 2001;75:117–124. doi: 10.1016/s0378-8741(00)00395-0. [DOI] [PubMed] [Google Scholar]
  2. Anzenbacher P., Anzenbacherová E. Cytochromes P450 and metabolism of xenobiotics. Cell. Mol. Life Sci. 2001;58:737–747. doi: 10.1007/PL00000897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Arbab A.H., Parvez M.K., Al-Dosari M.S., Al-Rehaily A.J. In vitro evaluation of novel antiviral activities of 60 medicinal plants extract against hepatitis B virus. Exp. Ther. Med. 2017;14:626–634. doi: 10.3892/etm.2017.4530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Al-Dosari M.S., Parvez M.K. Genetic polymorphisms of drug eliminating enzymes and transporters. Biomed. Genet. Genom. 2016;1:44–50. [Google Scholar]
  5. Duke, J., 1983. Euphorbia tirucalli L., Handbook of Energy Crops. Purdue University Centre for New Crops and Plant Products. Available at: <www.hort.purdue.edu>.
  6. Hastilestari B.R., Mudersbach M., Tomala F., Vogt H., Biskupek-Korell B., Van Damme P., Guretzki S., Papenbrock J. Euphorbia tirucalli L.-comprehensive characterization of a drought tolerant plant with a potential as biofuel source. PLoS One. 2013;8 doi: 10.1371/journal.pone.0063501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Harmsen S., Koster A.S., Beijnen J.H., Schellens J.H., Meijerman I. Comparison of two immortalized human cell lines to study nuclear receptor-mediated CYP3A4 induction. Drug Metab. Dispos. 2008;36:1166–1171. doi: 10.1124/dmd.107.017335. [DOI] [PubMed] [Google Scholar]
  8. Hustert E., Zibat A., Presecan-Siedel E., Eiselt R., Mueller R., Fuss C., Brehm I., Brinkmann U., Eichelbaum M., Wojnowski L., Burk O. Natural protein variants of pregnane X receptor with altered transactivation activity toward CYP3A4. Drug Metab. Dispos. 2001;29:1454–1459. [PubMed] [Google Scholar]
  9. Hu D., Wang Y., Chen Z., Ma Z., You Q., Zhang X., Liang Q., Tan H., Xiao C., Tang X., Gao Y. The protective effect of piperine on dextran sulfate sodium induced inflammatory bowel disease and its relation with pregnane X receptor activation. J. Ethnopharmacol. 2015;169:109–123. doi: 10.1016/j.jep.2015.04.006. [DOI] [PubMed] [Google Scholar]
  10. Hu D., Wang Y., Chen Z., Ma Z., You Q., Zhang X., Zhou T., Xiao Y., Liang Q., Tan H., Xiao C., Tang X., Zhang B., Gao Y. Artemisinin protects against dextran sulfate-sodium-induced inflammatory bowel disease, which is associated with activation of the pregnane X receptor. Eur. J. Pharmacol. 2014;738:273–284. doi: 10.1016/j.ejphar.2014.04.050. [DOI] [PubMed] [Google Scholar]
  11. Jakhar S., Dahiya P. Antimicrobial, antioxidant and phytochemical potential of Alternanthera pungens HB&K. J. Pharm. Sci. Res. 2017;9:1305–1311. [Google Scholar]
  12. Joshi Y., Joshi A.K., Prasad N., Juyal D. A review on Ficus palmata (Wild Himalayan Fig) J. Phytopharmacol. 2014;3:374–377. [Google Scholar]
  13. Kolars J.C., Schmiedlin-Ren P., Schuetz J.D., Fang C., Watkins P.B. Identification of rifampin-inducible P450IIIA4 (CYP3A4) in human small bowel enterocytes. J. Clin. Invest. 1992;90:1871–1878. doi: 10.1172/JCI116064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Lehmann J.M., McKee D.D., Watson M.A., Willson T.M., Moore J.T., Kliewer S.A. The human orphan nuclear receptor PXR is activated by compounds that regulate CYP3A4 gene expression and cause drug interactions. Proc. Natl. Acad. Sci. USA. 1998;95:12208–12213. doi: 10.1172/JCI3703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Matic M., Mahns A., Tsoli M., Corradin A., Polly P., Robertson G.R. Pregnane X receptor: promiscuous regulator of detoxification pathways. Int. J. Biochem. Cell Biol. 2007;39:478–483. doi: 10.1016/j.biocel.2006.08.017. [DOI] [PubMed] [Google Scholar]
  16. Moore L.B., Goodwin B., Jones S.A., Wisely G.B., Serabjit-Singh C.J., Willson T.M., Collins J.L., Kliewer S.A. St. John's wort induces hepatic drug metabolism through activation of the pregnane X receptor. Proc. Natl. Acad. Sci. USA. 2000;97:7500–7502. doi: 10.1073/pnas.130155097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Mooiman K.D., Maas-Bakker R.F., Moret E.E., Beijnen J.H., Schellens J.H., Meijerman I. Milk thistle's active components silybin and isosilybin: novel inhibitors of PXR-mediated CYP3A4 induction. Drug Metab. Dispos. 2013;41:1494–1504. doi: 10.1124/dmd.113.050971. [DOI] [PubMed] [Google Scholar]
  18. López-Terrada D., Cheung S.W., Finegold M.J., Knowles B.B. HepG2 is a hepatoblastoma-derived cell line. Hum. Pathol. 2009;40:1512–1515. doi: 10.1016/j.humpath.2009.07.003. [DOI] [PubMed] [Google Scholar]
  19. Pelkonen O., Mäenpää J., Taavitsainen P., Rautio A., Raunio H. Inhibition and induction of human cytochrome P450 (CYP) enzymes. Xenobiotica. 1998;28:1203–1253. doi: 10.1080/004982598238886. [DOI] [PubMed] [Google Scholar]
  20. Quatrochi L.C., Guzelian P.S. CYP3A regulation: from pharmacology to nuclear receptors. Drug Met. Disposit. 2001;29:615–622. [PubMed] [Google Scholar]
  21. Schuetz E.G., Guzelian P.S. Induction of cytochrome P450 by glucocorticoids in rat liver. II. Evidence that glucocorticoids regulate induction of cytochrome P450 by a nonclassical receptor mechanism. J. Biol. Chem. 1984;259:2007–2012. [PubMed] [Google Scholar]
  22. Tyagi L.K., Singh M., Singh V., Singh N., Tyagi S.N., Kori M.L. Herb-drug interactions: emerging threat and their management. Bot. Res. Int. 2010;3:1–13. [Google Scholar]
  23. van Heerden F.R., Viljoen A.M., van Wyk B.E. The major flavonoid of Dodonaea angustifolia. Fitoterapia. 2000;71:602–604. doi: 10.1016/s0367-326x(00)00201-x. [DOI] [PubMed] [Google Scholar]
  24. Zhang L., Reynolds K.S., Zhao P., Huang S.M. Drug interactions evaluation: an integrated part of risk assessment of therapeutics. Toxicol. Appl. Pharmacol. 2010;243:134–145. doi: 10.1016/j.taap.2009.12.016. [DOI] [PubMed] [Google Scholar]
  25. Zhang J., Ding L., Wang B., Ren G., Sun A., Deng C., Wei X., Mani S., Wang Z., Dou W. Notoginsenoside R1 attenuates experimental inflammatory bowel disease via pregnane X receptor activation. J. Pharmacol. Exp. Ther. 2015;352:315–324. doi: 10.1124/jpet.114.218750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Zhou S.F., Lai X. An update on clinical drug interactions with the herbal antidepressant St. John’s wort. Curr. Drug Metab. 2008;9:394–409. doi: 10.2174/138920008784746391. [DOI] [PubMed] [Google Scholar]

Articles from Saudi Pharmaceutical Journal : SPJ are provided here courtesy of Springer

RESOURCES