Home | Register | Login | Inquiries | Alerts | Sitemap |  


Advanced Search
JKM > Volume 35(2); 2014 > Article
Jin, Ha, Jeong, and Shin: Effects of Korean traditional herbal formula for common cold on the activities of human CYP450 isozymes

Abstract

Objectives:

Most drug interactions are attributed to the inhibition or induction of the activity of cytochrome P450s (CYP450). Although the regulation of CYP450s by drugs has been widely reported, there have been few studies on influence of traditional herbal formulas on the drug-metabolizing enzymes. Because herbal formulas have been used traditionally to treat various diseases and because herb-drug interactions are crucial factors determining therapeutic efficacies, a systematic evaluation of the effects of herbal formulas is important.

Methods:

The effects of Galgeun-tang (GGT, gegen tang), Gumiganghwal-tang (GMGHT, jiuweiqianghuo tang), Insampaedok-san (ISPDS, renshenbaidu powder), Samsoeum (SSE, shensu drink), Socheongryong-tang (SCRT, xiaoqinglong-tang) and Sosiho-tang (SSHT, xiaochaihu tang) that are traditional herbal formulas used to treat common cold, on drug-metabolizing enzymes were evaluated through an in vitro CYP3A4, CYP2D6, CYP2C19 and CYP2E1 inhibition assay to assess its interaction potential with synthetic drugs. The inhibitory effects of herbal formulas were characterized with IC50 values.

Results:

These six herbal formulas inhibited the activities of CYP3A4, 2C19, 2D6 and 2E1, in a concentration-dependent manner. Among the six herbal formulas, GGT critically inhibited CYP2C19, CYP2D6 and CYP2E1. GMGHT also inhibited CYP2D6 and CYP2E1 to a greater extent than the other CYP450 isozymes. Additionally, SSE and SSHT may change the effects of medicines that depend primarily on the CYP2C19 and CYP2E1 pathways. On the other hand, ISPDS and SCRT were not inhibited CYP3A4, CYP2C19, CYP2D6 and CYP2E1-mediated metabolism.

Conclusions:

These findings provide useful information regarding the safety and effectiveness of herbal formulas.

Introduction

Cytochrome P450s (CYP450), the most important drug-metabolizing enzyme system in humans, are expressed predominantly in the liver.1) Since herbal formulas modulate the metabolism of drugs by controlling the activities of specific CYP450 isozymes, herb-drug interactions can occur when herbal formulas and synthetic drugs are co-administered.2) In recent years, many studies reported that co-administration of herbal medicine and synthetic drug can lead to herb-drug interactions through regulation of the drug-metabolizing enzymes.3) Although several studies have evaluated the risk and safety for combination of herb and drug, study on effect of herbal formulas on the drug-metabolizing enzyme CYP450s are still rare.
Among the traditional herbal formulas, Galgeun-tang (GGT, gegen tang, kakkon-to), Gumiganghwal-tang (GMGHT, jiuweiqianghuo tang, kumi-kyokatsu-to), Insampaedok-san (ISPDS, renshenbaidu powder, ninjin-haidoku-san), Samsoeum (SSE, shensu drink, jinsoin), Socheongryong-tang (SCRT, xiaoqinglong-tang, sho-seiryu-to) and Sosiho-tang (SSHT, xiaochaihu tang, shosaika-to) are frequently used to treat common cold in Korea.4,5) These herbal formulas composed of different crude herbs (Table 1) and commonly exhibits various biochemical activities such antipyretic611) and anti-inflammatory12) effects in common. In addition, GGT has antioxidant13) and antivirus14) effects, and SSE and SCRT have antiallergic15) and antiasthma16) effects. SCRT also exhibits biological activities such as antiobesity and improvement of lung damage16) as well as induction of CYP2B, CYP2C and CYP3B activation effects.17) SSHT has immunoregulation, anti-hepatitis, antivirus and antioxidant effects.18) These herbal formulas can be used in combination with conventional drugs, and thus herb-drug interactions should be considered for proper administration of herb and drug. Therefore, we investigated the possible effects of six herbal formulas on the four major human CYP450 isozymes (CYP3A4, CYP2D6, CYP2C19 and CYP2E1) by an in vitro CYP450 isozyme inhibition assay.

Materials and methods

1. Chemicals and materials

Vivid®CYP450 Screening Kits (Vivid® CYP3A4 Green, Vivid® CYP2D6 Blue, Vivid® CYP2C19 Blue, and Vivid® CYP2E1 Blue) were purchased from Invitrogen Co. (Camarillo, CA, USA). The kits use di(benzyloxymethoxy)fluorescein (DBOMF) as a substrate for CYP3A4 and 7-ethoxy-methyloxy-3-cyanocoumarin (EOMCC) as a substrate for CYP2D6, CYP2C19 and CYP2E1. Ketoconazole, quinidine, miconazole, and sodium diethyldithiocarbamate trihydrate were obtained from Sigma Chemical Co. (St. Louis, MO, USA). All other chemicals were of analytical grade.

2. Preparation of herbal formula extracts

The herbs of each herbal formulation (Table 1) were chopped, mixed and extracted in water at 10 0°C for 120 min in an herb extractor (COSMOS660, Kyung Seo Machine Co., Incheon, Korea) and then filtered. All herbs were taxonomically confirmed by Professor Je-Hyeun Lee, Dongguk University, Korea. Voucher specimens have been deposited at the Herbal Medicine Formulation Research Group, Korea Institute of Oriental Medicine. The extracts were lyophilized and powdered extracts were stored at 4°C. The yields of herbal formula extracts are shown in Table 1. The HPLC profile of each extract was is same as in the previous study announced (Table 2).1823)

3. Cytochrome P450 isozyme assay

The assays were performed using the Vivid® CYP450 Screening Kits with the protocol provided by the manufacturer. The Vivid® CYP450 Screening Kits are designed to assess the metabolic activity and inhibition of the predominant human CYP450 isozymes involved in hepatic drug metabolism: CYP3A4, CYP2D6, CYP2C19 and CYP2E1. Vivid® Substrate and Fluorescent Standards were reconstituted, and a standard curve was prepared. A test sample of 40 μL diluted in solvent, a positive inhibition control (a compound that inhibits the respective CYP450 isozyme) or a solvent control was added to each well. The solutions were mixed after adding 50 μL of Master Pre-Mix containing P450 BACULOSOMES in Vivid® CYP450 Reaction Buffer and Regeneration System (consisting of glucose-6-phosphate and glucose-6-phosphate dehydrogenase), and the plate was incubated for 20 min to allow the samples to interact with the CYP enzymes. After pre-incubation, the reaction was started by adding 10 μL of Vivid® Substrate and NADP+. The Regeneration System converts NADP+ into NADPH, which is required to start the CYP450 reaction. The enzymatic reaction is initiated by the addition of a mixture of NADP+ and the appropriate Vivid® Substrate. The fluorescence intensity was measured using an EnVision2103 Multilabel Reader (PerkinElmer Inc., MA, USA) for 15 min at excitation and emission wavelengths of 485 and 535 nm for CYP3A4. For CYP2D6, CYP2C19 and CYP2E1, the fluorescence intensity was measured for 60 min at excitation and emISPDSion wavelengths of 405 and 450 nm, respectively.
The percent inhibition (%) was obtained by the following equation: % Inhibition = [1 − (S1−S0)/(C1−C0)] 100, where C1 is the fluorescence of the control after incubation, C0 is the initial fluorescence of the control, S1 is the fluorescence of the test sample after incubation, and S0 is the initial fluorescence of the test sample in the linear section.
The background fluorescence of herbal formulas was corrected by subtracting the values obtained from incubation without substrates. The CYP450s inhibition of each sample was in term of IC50, as calculated from the log-dose inhibition curve. Data were expressed as the meanSEM (n = 3). Ketoconazole, miconazole, quinidine and sodium diethyldithiocarbamate trihydrate were used as positive controls for CYP3A4, CYP2C19, CYP2D6 and CYP2E1, respectively.

Results

To evaluate whether six Korean traditional herbal formulas (GGT, GMGHT, ISPDS, SSE, SCRT and SSHT) can influence activities of CYP3A4, CYP2C19, CYP2D6 and CYP2E1, CYP450 isozyme inhibition assay was conducted. Ketoconazole, miconazole, quinidine and diethyldithiocarbamate used as positive controls inhibited CYP3A4, CYP2C19, CYP2D6 and CYP2E1 in a dose-dependent manner, with IC50 values of 1.17 to 2.70 nM, 0.25 to 1.54 M, 4.33 to 7.90 nM and 5.87 to 9.83 μM, respectively.

1. Effects of GGT on the activities of CYP450s

As demonstrated by the data in Fig. 1 and Table 3, GGT exhibited the most potent inhibition of CYP2E1, with an IC50 value of 20.57 μg/mL. In contrast, GGT exerted inhibition of CYP3A4, CYP2C19 and CYP2D6, with IC50 values of 259.6, 105.6 and 151.0 μg/mL, respectively.

2. Effects of GMGHT on the activities of CYP450s

GMGHT showed the most potent inhibition of CYP2D6, with an IC50 value of 80.15 μg/mL (Fig. 2 and Table 3). Additionally, GMGHT inhibited CYP3A4, CYP2C19 and CYP2E1, with respective IC50 values of 334.0, 255.8, and 106.0 μg/mL.

3. Effects of ISPDS on the activities of CYP450s

As shown in Fig. 3 and Table 3, ISPDS inhibited of CYP3A4 and CYP2C19, with similar IC50 values of 286.6 and 231.6 μg/mL, respectively. Additionally, ISPDS relatively weak inhibition of CYP2D6 and CYP2E1, with IC50 values of 695.5 and 958.1 μg/mL, respectively.

4. Effects of SSE on the activities of CYP450s

As presented in Fig. 4 and Table 3, SSE potently repressed the activity of inhibited CYP2C19, with an IC50 value of 84.38 μg/mL, while having no effect on CYP2D6, with an IC50 value of over 1000 μg/mL. For CYP3A4 and CYP2E1, SSE exhibited inhibitory activities, with IC50 values of 308.9 and 137.0 μg/mL, respectively.

5. Effects of SCRT on the activities of CYP450s

As demonstrated by the data in Fig. 5 and Table 3, SCRT showed the most potent inhibition of CYP2E1 (IC50 = 270.2 μg/mL), followed by CYP2D6 (IC50 = 378.2 μg/mL). In addition, SCRT displayed similar inhibition against CYP3A4 and CYP2C19, with IC50 values of 596.3 and 578.9 μg/mL, respectively.

6. Effects of SSHT on the activities of CYP450s

As shown in Fig. 6 and Table 3, SSHT exhibited the maximum inhibition of CYP2C19, with an IC50 value of 35.44 μg/mL. SSHT also inhibited CYP3A4, CYP2D6 and CYP2E1, with IC50 values of 373.8, 497.4 and 193.1 μg/mL, respectively.

Discussion

In recent years, many researchers have shown an increasing interest in herbal medicines, especially traditional herbal formulas, a combination of several different herbs. Although herbal formulas have been used continuously and extensively, there have been few clinical and preclinical data generated on the interactions between herbal formulas and synthetic drugs. CYP450s, drug-metabolizing enzymes in the liver were determined to be heme proteins, can be regulated by a wide variety of xenobiotics, including multiple active constituents of herbs.2426) The inhibition of CYP450s results in elevated drug concentrations in tissues, leading to various adverse reactions, particularly for drugs with a low therapeutic index.27,28) Therefore, it is important to identify the potential herb-drug interactions to prevent adverse effects in patients taking combinations of herbal formulas and conventional drugs. The major CYP450 isozymes that are important in human drug metabolisms are CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP2E1 and CYP3A4.25)
GGT, GMGHT, ISPDS, SSE, SCRT and SSHT are traditional herbal formulas used to treat common cold. These herbal formulas are consisted of respective different herbs (Table 1), and contain various active components. To date, several studies reported the influences of herbal extracts or chemicals in herbal formulas on the CYP450s activities. Puerariae Radix and its component puerarin of GGT and SSE were reported to inactivate the human CYP3A, CYP2B1 and CYP2E1.29) In addition, pseudoephedrine in Ephedrae Herba of GGT and SCRT has inhibitory effects on CYP1A1/2 and CYP2E1 of rat liver microsomes,30) and ginsenoside Rg1 and Rb1 in Ginseng Radix of ISPDS, SSE and SSHT have inductive effect on CYP1A1 expression in HepG2 cells.31) Furthermore, GGT and SSHT of the six formulas inhibited the activities of CYP3A4 in a dose-dependent manner but inhibition at 1000 g/mL did not reach 50%.32) Among the marker compounds of these herbal formulas, glycyrrhizin in GGT, GMGHT, ISPDS, SSE and SSHT have been reported to inhibit the human CYP1A2.33) Baicalin, containing of GMGHT and SSHT, was reported to increase the liver microsomal CYP1A1, CYP2B1 and CYP2C11 in mice.34) According to Ueng et al.,35) baicalein and wogonin in GMGHT have inhibitory effects on hepatic CYP3A4 and CYP2E1 in mice. In addition, it has been reported that the hepatic CYP1A2 protein level was increased by baicalein in mice,35) and the activity of CYP1A2 and CYP2C19 were inhibited by wogonin in human liver microsomes.36) Naringin of ISPDS and SSE reduce the CYP1A2 protein level in mice.37) However, no studies have been conducted on the effects of six traditional herbal formulas used to treat common cold on the activities of human CYP450. Therefore, the present study focused on the effects of GGT, GMGHT, ISPDS, SSE, SCRT and SSHT, six traditional herbal formulas, on the activities of major human CYP450 isozymes: CYP3A4, CYP2D6, CYP2C19 and CYP2E1. Our results showed that GGT containing of Puerariae Radix, Cinnamomi Ramulus, Ephedrae Herba and Paeoniae Radix showed the most potent inhibition of CYP2E1. These results might be related to the inhibitory effect of Puerariae Radix on CYP2E1 based on Guerra et al’s paper.29) GMGHT likely interacts with drugs metabolized by CYP2D6 and CYP2E1 rather than the other isozymes. In contrast, ISPDS and SCRT were weak inhibited the activities of CYP3A4, CYP2C19, CYP2D6 and CYP2E1. These results suggest that ISPDS and SCRT are unlikely to inhibit the metabolism of drugs metabolized by CYP3A4, CYP2C19, CYP2D6 and CYP2E1. However, SSE and SSHT inhibited CYP2C19 and CYP2E1 at low concentrations. In addition, it is necessary to pay attention to herb-drug interactions when SSE or SSHT is used in combination with other cold medicine metabolized by CYP2C19.

Conclusions

By now the regulation of CYP450s by drugs has been widely reported, however there have been few studies on influence of traditional herbal formulas on the drug-metabolizing enzymes. Therefore, we investigated influence of six herbal formulas on the activities of CYP3A4, CYP2C19, CYP2D and CYP2E1 by in vitro CYP450 isozyme assay. As a result, the possibility of herb-drug interactions should be considered if a patient with colds consumes GGT, GMGHT, SSE or SSHT. Further studies regarding the importance of herb-drug interactions will be required because these reports provide limited information of herbal formulas on the CYP450 activities.

ACKNOWLEDGMENTS

This Research was supported by the grant ‘Construction of Scientific Evidences for Herbal Medicine Formulas (K14030)’ from the Korea Institute of Oriental Medicine (KIOM).

Fig. 1.
Effects of Galgeun-tang on the CYP3A4 (A), CYP2C19 (B), CYP2D6 (C) and CYP2E1 (D).
The fluorescence-based enzyme assays on CYP450 isozymes were established in vitro. Ketoconazole, miconazole, quinidine and diethyldithiocarbamate was used as positive controls, respectively. Data are presented as mean ± SEM (n = 3).
jkm-35-2-47f1.tif
Fig. 2.
Effects of Gumiganghwal-tang on the CYP3A4 (A), CYP2C19 (B), CYP2D6 (C) and CYP2E1 (D).
The fluorescence-based enzyme assays on CYP450 isozymes were established in vitro. Ketoconazole, miconazole, quinidine and diethyldithiocarbamate was used as positive controls, respectively. Data are presented as mean ± SEM (n = 3).
jkm-35-2-47f2.tif
Fig. 3.
Effects of Insampaedok-san on the CYP3A4 (A), CYP2C19 (B), CYP2D6 (C) and CYP2E1 (D).
The fluorescence-based enzyme assays on CYP450 isozymes were established in vitro. Ketoconazole, miconazole, quinidine and diethyldithiocarbamate was used as positive controls, respectively. Data are presented as mean ± SEM (n = 3).
jkm-35-2-47f3.tif
Fig. 4.
Effects of Samsoeum on the CYP3A4 (A), CYP2C19 (B), CYP2D6 (C) and CYP2E1 (D).
The fluorescence-based enzyme assays on CYP450 isozymes were established in vitro. Ketoconazole, miconazole, quinidine and diethyldithiocarbamate was used as positive controls, respectively. Data are presented as mean ± SEM (n = 3).
jkm-35-2-47f4.tif
Fig. 5.
Effects of Socheongryong-tang on the CYP3A4 (A), CYP2C19 (B), CYP2D6 (C) and CYP2E1 (D).
The fluorescence-based enzyme assays on CYP450 isozymes were established in vitro. Ketoconazole, miconazole, quinidine and diethyldithiocarbamate was used as positive controls, respectively. Data are presented as mean ± SEM (n = 3).
jkm-35-2-47f5.tif
Fig. 6.
Effects of Sosiho-tang on the CYP3A4 (A), CYP2C19 (B), CYP2D6 (C) and CYP2E1 (D).
The fluorescence-based enzyme assays on CYP450 isozymes were established in vitro. Ketoconazole, miconazole, quinidine and diethyldithiocarbamate was used as positive controls, respectively. Data are presented as mean ± SEM (n = 3).
jkm-35-2-47f6.tif
Table 1.
The Compositions of Herbal Formulas
Crude drugs Amount (g)
Galgeun-tang Gumiganghwal-tang Insampaedok-san Samsoeum Socheongryong-tang Sosiho-tang
Puerariae Radix 9.00 3.75
Cinnamomi Ramulus 6.00 3.75
Ephedrae Herba 6.00 5.63
Paeoniae Radix alba 6.00 5.63
Glycyrrhizae Radix 6.00 1.88 3.75 2.81 3.75 1.88
Zingiberis Rhizoma 6.00 3.75 3.75 3.75
Zizyphi Fructus 5.00 3.75 3.75
Osterici Radix 5.63 3.75
Saposhnikoviae Radix 5.63
Cnidii Rhizoma 4.50 3.75
Angelicae Dahuricae Radix 4.50
Atractylodis Rhizoma 4.50
Scutellariae Radix 4.50 3.75
Rehmanniae Radix 4.50
Asiasari Radix et Rhizoma 1.88 3.75
Bupleuri Radix 3.75 11.25
Ginseng Radix 3.75 3.75 3.75
Pinelliae Tuber 3.75 5.63 7.50
Angelicae Gigantis Radix
Citri Unshius Pericarpium 2.81
Magnoliae Cortex
Platycodonis Radix 3.75 2.81
Aurantii Fructus 2.81
Zingiberis Rhizoma 3.75
Poria Sclerotium 3.75 3.75
Cinnamomi Cortex
Perillae Herba 3.75
Anthrisci Radix 3.75 3.75
Schizandrae Fructus 5.63
Araliae Cordatae Radix 3.75
Menthae Herba 3.75

Total (g) 44.00 37.52 41.25 41.24 37.52 35.63
Yield (%) 12.60 22.76 24.30 12.60 21.70 16.37
Table 2.
The Contents of Marker Compounds in Herbal Formulas
Compound Contents in extract (mg/g)
Galgeun-tang Gumiganghwal-tang Insampaedok-san Samsoeum Socheongryong-tang Sosiho-tang
Puerarin 10.26 : 0.030 5.30 : 0.026
Albiflorin 2.54 : 0.010 2.123 : 0.010
Daidzin 1.94 : 0.012 1.25 : 0.012
Paeoniflorin 3.54 : 0.012 12.224 : 0.016
Liquiritin 6.60 : 0.084 1.78 : 0.007 2.33 : 0.022 2.30 : 0.023 3.131 : 0.034 2.80 : 0.04
Glycyrrhizin 12.13 : 0.045 1.87 : 0.001 3.28 : 0.008 4.00 : 0.040 2.025 : 0.006 2.30 : 0.010
Cinnamic acid 2.00 : 0.021 0.338 : 0.000
Cinnamaldehyde 7.26 : 0.055 0.236 : 0.002
Ferulic acid 1.69 : 0.001 0.72 : 0.002
Baicalin 37.90 : 0.267 60.90 : 0.040
Baicalein 0.26 : 0.007
Wogonin 0.03 : 0.000
Naringin 9.88 : 0.033 10.45 : 0.076
Hesperidin 1.42 : 0.034 5.69 : 0.081
Neohesperidin 6.82 : 0.028 5.98 : 0.031
Gallic acid 0.993 : 0.004
Benzoic acid 0.974 : 0.013
Isoliquiritin 0.240 : 0.000
Coumarin 0.215 : 0.000
6-Gingerol 0.456 : 0.001
Schizandrin 0.596 : 0.003
Methyl eugenol 0.123 : 0.000

The data are presented as means±SD from three independent experiments in triplicate.

Table 3.
IC50 Values (μg/mL) of Herbal Formulas on CYP450 Isozyme Activities
Herbal formula CYP3A4 CYP2C19 CYP2D6 CYP2E1
Galgeun-tang 259.6 105.6 151.0 20.57
Gumiganghwal-tang 334.0 255.8 80.15 106.0
Insampaedok-san 286.6 231.6 695.5 958.1
Samsoeum 308.9 84.38 > 1000 137.0
Socheongryong-tang 596.3 578.9 378.2 270.2
Sosiho-tang 373.8 35.44 497.4 193.1

Positive Control 1.17−2.70 nM 0.25−1.54 μ M 4.33−7.90 nM 5.87−9.83 μ M

Ketoconazole, miconazole, quinidine and diethyldithiocarbamate were used as positive controls for CYP3A4, CYP2C19, CYP2D6 and CYP2E1, respectively.

References

1.. Watkins PB. Drug metabolism by cytochromes P450 in the liver and small bowel. Gastroenterol Clin North Am. 1992; 21:3. 51126
pmid

2.. Hellum BH, Hu Z, Nilsen OG. The induction of CYP1A2, CYP2D6 and CYP3A4 by six trade herbal products in cultured primary human hepatocytes. Basic Clin Pharmacol Toxicol. 2007; 100:1. 2330
crossref pmid

3.. Liu R, Qin M, Hang P, Liu Y, Zhang Z, Liu G. Effects of Panax notoginseng saponins on the activities of CYP1A2, CYP2C9, CYP2D6 and CYP3A4 in rats in vivo. Phytotherapy Res. 2011; 26:8. 11138
crossref

4.. Seo CS, Huang DS, Lee JK, Ha HK, Chun JM, Um YR, et al. Concentration of heavy metals, residual pesticides and sulfur dioxide of before/after a decoction. Kor J Herbology. 2008; 23:4. 518


5.. Kim JH, Shin HK. Analysis of biological experiment on Socheongryong-tang (Xiaoqinglongang). J Korean Oriental Med. 2012; 33:3. 5473


6.. Yang TK, Kim YB, Chae BY. An experimental study on the anti-allergic effects, anti-inflammatory action, anti-pyretic action and analgesic action of Galgeun-tang, Gamigalgeun-tang and Geomahwanggalgeun-ang. J Korean Oriental Med Ophthalmol tolaryngol Dermatol. 2002; 15:1. 7695


7.. Moon YH, Go JJ, Park JY. The anti-inflammatory and analgesic activities of Gumiganghwaltang. Korean J Pharmacognosy. 1999; 30:1. 1824


8.. Shim JY, Kim BW. A study of effects of Insampaedok-san and Kammiinsampaedok san on the analgesia, antipyresis and thioacetamide induced liver damage in rats and mice. Kyung-Hee Univ Orient Med J. 1984; 7:1. 32334


9.. Choi SB, Rhee HK. A study of effects of Samsoeum on the analgesia, antipyresis and O3 poisoned lung in rats and mice. Kyung-Hee Univ Orient Med J. 1985; 8:13948


10.. Yamahara J, Yamada T, Kimura H, Sawada T, Fujimura H. Biologically active principles of crude drugs. II. Anti-allergic principles in “Shoseiryu-To” anti-inflammatory properties of paeoniflorin and its derivatives. J Pharmacobiodyn. 1982; 5:11. 9219
crossref pmid

11.. Woo HJ, Kim DH, Kim BW. A experimental and clinical study on effects of Soshiho-tang. Kyung-Hee Univ Orient Med J. 1990; 6:1. 97107


12.. Lee JA, Ha H, Jung DY, Lee H, Lee JK, Huang DS, et al. Comparative study of 25 herbal formulas on anti-inflammatory effect. J Orient Gynecol. 2010; 23:3. 10111


13.. Shin JM, Kim YO, Baek SH. Free radical scavenging activity and kinetic behavior of the Galgeuntang water extract. Kyung-Hee Univ Orient Med J. 2008; 8:1. 3238
crossref

14.. Wu MS, Yen HR, Chang CW, Peng TY, Heish CF, Chen CJ, et al. Mechanism of action of the suppression of influenza virus replication by Ko-Ken Tang through inhibition of the phosphatidylinositol 3-kinase/Akt signaling pathway and viral RNP nuclear export. J Ethnopharmacol. 2011; 134:3. 61423
pmid

15.. Jeong HJ, Hong MC, Shin MK, Bae HS. Sochungyong-tang, tradititional Korean medicine, suppresses Th2 lineage development. Korean J Orient Physiol Pathol. 2005; 19:2. 3808


16.. Kim JH, Shin HK. Analysis of biological experiment on Socheongryong-tang. J Korean Orient Med. 2012; 33:3. 5473


17.. Lee JW, Jung HJ, Jung SG, Lee HG. The effects of Socheoryong-tang on lymphocytes in BALF of rat. Orient Pharm Exp Med. 2001; 17:2. 24253


18.. Seo CS, Lim JH, Shin HK. Simultaneous determination of albiflorin, cinnamaldehyde, cinnamic acid, daidzin, glycyrrhizin, liquiritin, paeoniflorin and puerarin in Galgeun-tang by HPLC-PDA. J Korean Orient Med. 2010; 31:6. 815


19.. Shin IS, Seo CS, Lee MY, Ha HK, Huh JI, Shin HK. In vitro and in vivo evaluation of the genotoxicity of Gumiganghwal-tang, a traditional herbal prescription. J Ethnopharmacol. 2012; 141:1. 3506
crossref pmid

20.. Seo CS, Kim JH, Shin HK. Simultaneous determination of six major constituents of the herbal formula Insampaedok-san using HPLC-PDA. ISRN Anal Chem. 2012; Article ID 609587.
crossref

21.. Seo CS, Kim JH, Huang DS, Shin HK. Simultaneous determination of seven compounds in Samsoeum by HPLC-PDA. Korean J Orient Med Prescription. 2010; 18:1. 95103


22.. Kim JH, Jeon WY, Lee MY, Seo CS, Lim HS, Shin HK. Inhibitory activity of Socheongryongtang and its constituent components against the production of RANTES, eotaxin, eotaxin-3, and MMP-9 from BEAS2B-cells. Spandidos Publ;2014. (in press).


23.. Shin IS, Lee MY, Kim Y, Seo CS, Kim JH, Shin HK. Subacute toxicity and stability of Soshihotang, a traditional herbal formula, in Sprague-Dawley rats. BMC Complement & Altern Med. 2012; 12:26675
pmid pmc

24.. Goshman L, Roller K. Clinically significant cytochrome P450 drug interactions. J Pharm Soc Wis. 1999; 3:2338


25.. Lee HW, Kim DW, Phapale PB, Lim MS, Park J, Seo JJ, et al. In vitro inhibitory effects of Wen-pi-tang-Hab-Wu-ling-san on human cytochrome P450 isoforms. J Clin Pharm Ther. 2011; 36:4. 496503
crossref pmid

26.. Xu C, Li CY, Kong AN. Induction of phase I, II and III drug metabolism/transport by xenobiotics. Arch Pharm Res. 2005; 28:3. 24968
pmid

27.. Saxena A, Tripathi KP, Roy S, Khan F, Sharma A. Pharmacovigilance: effects of herbal components on human drugs interactions involving cytochrome P450. Bioinformation. 2008; 3:5. 198204
crossref pmid pmc

28.. Foti RS, Rock DA, Wienkers LC, Wahlstrom JL. Selection of alternative CYP3A4 probe substrates for clinical drug interaction studies using in vitro data and in vivo simulation. Drug Metabolism Disposition. 2010; 38:6. 9817
crossref pmid

29.. Guerra MC, Speroni E, Broccoli M, Cangini M, Pasini P, Minghett A, et al. Comparison between chinese medical herb Pueraria lobata crude extract and its main isoflavone puerarin antioxidant properties and effects on rat liver CYP-catalysed drug metabolism. Life Sci. 2000; 67:24. 29973006
crossref

30.. Wu W, Liu L, Han F, Chen Y. Effect of pseudoephedrine and ephedrine on the activities of cytochrome P450 enzymes in rat liver microsomes. China J Trad Med Pharm. 2011; 08.


31.. Wang Y, Ye X, Ma Z, Liang Q, Lu B, Tan H, et al. Induction of cytochrome P450 1A1 expression by ginsenoside Rg1 and Rb1 in HepG2 cells. European J Pharmacol. 2008; 601:13. 738
crossref pmid

32.. Takahashi K, Uejima E, Morisaki T, Takahashi K, Kurokawa N, Azuma J. In vitro inhibitory effects of Kampo medicines on metabolic reactions catalyzed by human liver microsomes. J Clin Pharm Ther. 2003; 28:4. 31927
crossref pmid

33.. Park JH, Park JY, Ju YS. Inhibitory effects of licorice ethanol extracts and glycyrrhizin on Cytochrome P450 drug-metabolizing enzymes in human liver microsomes. Korean J Orient Prev Med Soc. 2003; 7:2. 6574


34.. Hou YN, Zhu XY, Cheng GF. Effects of baicalin on liver microsomal cytochrome P450 system. Acta Pharmaceutica Sinica. 2000; 35:12. 890–2.
pmid

35.. Ueng YF, Shyu CC, Lin YL, Park SS, Liao JF, Chen CF. Effects of baicalein and wogonin on drug-metabolizing enzymes in C57BL/6J mice. Life Sci. 2000; 67:18. 2189–200.
crossref pmid

36.. Li T, Li N, Guo Q, Ji H, Zhao D, Xie S, et al. Inhibitory effects of wogonin on catalytic activity of cytochrome P450 enzyme in human liver microsomes. Eur J Drug Metab Pharmacokinet. 2011; 36:4. 249–56.
crossref pmid

37.. Ueng YF, Chang YL, Oda Y, Park SS, Liao JF, Lin MF, et al. In vitro and in vivo effects of naringin on cytochrome P450-dependent monooxygenase in mouse liver. Life Sci. 1999; 65:24. 2591–602.
crossref pmid

TOOLS
PDF Links  PDF Links
Full text via DOI  Full text via DOI
PubReader  PubReader
Download Citation  Download Citation
  Print
Share:      
METRICS
4
Crossref
7,193
View
132
Download
Editorial office contact information
3F, #26-27 Gayang-dong, Gangseo-gu Seoul, 157-200 Seoul, Korea
The Society of Korean Medicine
Tel : +82-2-2658-3627   Fax : +82-2-2658-3631   E-mail : skom1953.journal@gmail.com
About |  Browse Articles |  Current Issue |  For Authors and Reviewers
Developed in M2PI