荷叶代谢性药物相互作用及体内成分研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
细胞色素P450(cytochromeP450,CYP450)酶系统是机体重要的代谢酶,它参与了许多内源性和外源性物质的代谢。在药物的代谢过程中,CYP2C19、CYP2D6. CYP3A4、CYP2E1和CYP2C9是最主要的亚型,其参与了市场上90%左右药物的代谢。当两种药物合并使用时,一种药物对另一种药物的代谢酶产生抑制或诱导作用都可能产生药物相互作用,导致产生毒性或治疗失败,增加临床治疗的风险。在临床过程中,抑制作用的危害要远大于诱导作用的危害。抑制P450酶的活性可能会引起血药浓度升高而导致毒性作用,有时甚至会危及生命。很多药物因发生严重的代谢性药物相互作用而被撤出市场。
     中药是来自大自然的天然产物,其常被认为是“安全的,无毒副作用”的药物,从而受到人们的青睐,被广泛使用。然而,近年来的研究发现多种中(草)药及其制剂也会对P450酶产生抑制或诱导作用,从而引起代谢性药物相互作用。这种药物间的相互作用已经引起人们的关注。因此,对一些常用中药进行潜在药物相互作用研究,以预测其可能发生的药物相互作用,是保证临床用药安全有效的重要途径,具有极其重要的意义。
     荷叶(Nelumbinis Folium)为睡莲科植物Nelumbo nucifera Gaertn.的干燥叶,为药食两用的中药。其具有清暑化湿、升发清阳,凉血止血的功效,常用于暑热烦渴、暑湿泄泻、脾虚泄泻、血热吐衄、便血崩漏等症。现代药理研究表明,荷叶黄酮和/或生物碱成分具有降血脂,减肥,抗氧化,抗HIV,抗菌和降糖等多种生物活性。作为传统中药,荷叶是多种降脂复方制剂的重要组成部分;作为天然食用资源,目前已被开发成多种减肥茶或其他保健饮品。因此,荷叶及其制剂与其他药物,特别是降脂减肥类药物共同使用的几率较大。然而,到目前为止,有关荷叶及其制剂在使用过程中是否会对P450酶的活性产生影响,是否会导致药物相互作用发生的研究报道仍然缺乏。因此,本文将从P450酶方面来探讨荷叶潜在的药物相互作用。本研究采用液相色谱-质谱联用(LC-MS/MS)技术,建立体外、体内酶活性的评价方法。首先经过体外模型预测荷叶潜在的药物相互作用,然后再通过体内实验进行验证,最后再通过分析荷叶在大鼠体内的药物成分及其在组织中的分布,进一步的从物质基础方面来阐释荷叶潜在药物相互作用的发生原因。此外,还初步探讨了荷叶提取物对转运体P-糖蛋白(P-gp)功能的影响。本论文完成的研究工作主要有以下几个方面:
     1.以奥美拉唑、右美沙芬、睾丸酮、氯唑沙宗和甲苯磺丁脲分别作为CYP2C19、CYP2D6、CYP3A4、CYP2E1、CYP2C9的探针底物,将这5个探针底物与人肝微粒体共同孵育,并利用LC-MS/MS分析技术检测代谢产物的生成量,建立同时测定人肝微粒体P450酶5种亚型活性的cocktail探针底物体外评价方法;并运用所建立的方法研究12味常用中药(荷叶、酸枣仁、决明子、五味子、山楂、枸杞子、莲子心、人参、菊花、黄精、茯苓和金银花)对人肝微粒体P450同工酶活性的影响。研究结果显示,所建立的方法具有灵敏度高、特异性强、重现性好和准确度高的优点,能满足体外代谢性药物相互作用研究的要求。12味中药的水提取物对P450同工酶的抑制作用较弱或没有抑制作用;而乙醇提取物则有不同程度的抑制作用(除枸杞子外),而且这种抑制作用存在浓度依赖性。其中荷叶乙醇提取物对CYP2C19、CYP2D6、 CYP3A4、CYP2E1和CYP2C9半数抑制浓度IC50值分别为77.38、12.05、98.01、61.43和83.46μg/mL;菊花乙醇提取物对CYP2C9活性抑制的IC50值为45.25μg/mL;五味子乙醇提取物对CYP3A4和CYP2C19活性抑制的IC50值分别为47.24和64.42μg/mL。上述研究结果提示,日常服用这些中药时,尤其是在长期服用这些中药经乙醇提取后制备所得的制剂时,应注意其潜在的药物相互作用。
     2.采用cocktail探针底物法分别研究荷叶总黄酮和总生物碱及其主要化学成分对人肝微粒体P450酶各亚型活性的影响。结果表明荷叶总黄酮对肝微粒体P450酶活性的抑制作用相对较弱,其IC50值均在100μg/mL左右;总生物碱对P450酶活性的抑制作用则较强,其对CYP2C19、CYP2D6、CYP3A4、CYP2E1(?)CYP2C9的半数抑制浓度IC50值分别为40.79、0.96、44.87、63.84和52.58μg/mL,其中对CYP2D6同工酶的抑制作用最强。总生物碱中的主要成分荷叶碱、N-去甲荷叶碱和2-羟基-1-甲氧基阿朴啡通过竞争性抑制方式抑制CYP2D6酶的活性,其抑制常数Ki值分别为1.88、2.34和1.56μM。但这三者对CYP2D6酶活性的抑制不存在时间依赖性和NADPH依赖性的不可逆抑制作用。
     采用Caco-2细胞模型研究荷叶乙醇提取物、总黄酮和总生物碱对P-糖蛋白(P-gp)功能的影响。结果表明,荷叶乙醇提取物和总生物碱对P-gp存在浓度依赖性的抑制作用,在浓度为100μg/mL时,Caco-2细胞内的荧光强度明显增加,即Caco-2细胞对Rho-123的摄取量增加,有显著的抑制作用(P<0.05);荷叶总黄酮对P-gP有一定的抑制作用,但与空白对照组相比较,无显著差异(P>0.05)。
     上述研究显示,荷叶对P450酶的活性有抑制作用,尤其是对CYP2D6亚型有较强的抑制作用;对P-gp功能也有一定的抑制作用。这些抑制作用主要与荷叶所含的生物碱成分有关。提示荷叶在与其他药物合并使用时,可能存在药物相互作用风险。
     3.采用右美沙芬和美托洛尔作为CYP2D6酶的探针底物,以LC-MS/MS技术为分析手段,研究荷叶总生物碱对这两种探针底物在体内的代谢产物右菲烷和α-羟基美托洛尔药代动力学的影响;给大鼠连续灌胃总生物碱(50mg/kg)7天,并于末次给药后2或12h时间点给大鼠静脉注射右美沙芬(5mg/kg)或美托洛尔(10mg/kg),观察其代谢产物血药浓度的变化。并通过蛋白质印迹法(western blot)研究总生物碱对大鼠体内CYP2D6酶蛋白表达水平的影响。结果表明,总生物碱能显著降低右菲烷和α-羟基美托洛尔的血药浓度。在末次给总生物碱后2或12h时间点给大鼠静脉注射右美沙芬或美托洛尔,总生物碱使右菲烷的AUCall从167.27h-ng/mL分别下降到43.13h-ng/mL(P<0.01)(给生物碱后2h时间点)和62.25h-ng/mL(P<0.05)(给生物碱后12h时间点);使α-羟基美托洛尔的AUCall从347.68h·ng/mL下降到223.24h·ng/mL(P<0.05)(给生物碱后12h时间点)。连续给予大鼠总生物碱7天后,大鼠体内CYP2D6酶蛋白的表达水平并无明显改变。上述研究结果提示,总生物碱在体内亦对大鼠CYP2D6酶有明显的抑制作用,与体外结果相吻合;但这种抑制作用并不影响CYP2D6蛋白的表达水平。
     4.采用LC-MS/MS分析方法,研究给大鼠灌胃荷叶总黄酮(50mg/kg)或总生物碱(50mg/kg)后的体内药物成分,以及大鼠静脉注射荷叶总生物碱(50mg/kg)后,其主要生物碱成分荷叶碱、N-去甲荷叶碱和2-羟基-1-甲氧基阿朴啡在大鼠组织中的分布情况。结果从大鼠血液和尿液中鉴定出37个黄酮成分,主要包括原型化合物和这些化合物与葡萄糖醛酸和硫酸等结合而形成的代谢产物,表明荷叶黄酮成分在体内的代谢途径主要为Ⅱ相代谢;从大鼠血液和尿液中鉴定出17个生物碱成分,包括生物碱原型化合物及其经氧化、甲基化等的代谢产物,和葡萄糖醛酸和硫酸等的结合物,表明荷叶生物碱在体内代谢广泛,其代谢途径包括Ⅰ相和Ⅱ相代谢。给大鼠静脉注射总生物碱后,三个主要生物碱成分荷叶碱、N-去甲荷叶碱和2-羟基-1-甲氧基阿朴啡能迅速(5min)分布到各组织,其中在肝脏、肺、脾脏和肾脏等组织中具有较高的浓度,并在体内维持较长时间。通过上述研究结果可初步推测荷叶对P450酶活性的抑制与其在体内的药物成分及其组织分布情况有关。
     总之,本文系统地研究了荷叶乙醇提取物、荷叶总黄酮和总生物碱及其主要化学成分对P450酶活性的影响,尤其是总生物碱对CYP2D6酶活性的抑制作用方面进行了深入研究。同时,对荷叶乙醇提取物及总黄酮和总生物碱对P-gp功能的影响进行了初步研究;最后,对给药后的大鼠进行体内药物成分分析和鉴定,并对总生物碱的主要成分在大鼠组织中的分布情况进行了研究。结果提示荷叶可通过抑制P450酶的活性或P-gp的功能,引起药物相互作用;而且,荷叶对P450酶的活性和P-gp功能的影响主要与其所含生物碱成分有关。本研究为荷叶在临床上的安全、合理应用提供了新信息;为荷叶成分的药理、毒理研究和开发利用提供了新思路。
Cytochrome P450(CYP) is a major metabolizing enzyme system in the body and plays an important role in the metabolism of many endogenous and exogenous substances. Five P450isoenzymes, namely, CYP2C19, CYP2D6, CYP3A4, CYP2E1and CYP2C9are particularly important because they are responsible for the metabolism of approximately90%of the currently known therapeutic drugs. Drug-drug interactions are frequently occurrence when drugs are co-administered and one drug modifies the metabolic clearance of the other drugs by inhibition or induction of some P450isoenzymes. In general, inhibition is much more harmful than induction in clinic settings. The inhibition of P450activity may cause an increase in the blood concentrations of the drug that may lead to toxic symptoms and sometimes even fatal interactions. Many drugs have been withdrawn from the market due to their significant inhibition of some isoenzymes that resulted in serious clinical damages.
     Traditional Chinese medicines (TCMs) are often considered as safe and harmless natural products and are conventionally used with prescription drugs. However, many different side effects have been reported for some herbs and/or their formulations recently. Many TCMs have been reported to exhibit inhibitory or induction effects on the P450enzymes, this cause to herb-drug interactions (HDI) and have attracted much attention. Thus, it is extremely important to evaluate the potential HDI of the commonly used herbs for their safe and effective use.
     Nelumbinis Folium, the dried leaves of lotus (Nelumbo nucifera Gaertn), is utilized not only as a dietary staple but also as a TCM to treat sunstroke, assuage thirst, and cure both diarrhea and fever in China. Modern pharmacological studies have demonstrated that the flavonoids and/or alkaloids of the herb exhibit various pharmacological effects, such as anti-hyperlipidemia, anti-obesity, anti-oxidant, anti-diabetic, anti-microbial, and anti-HIV activities. Currently, the herb is becoming more popular in China as a "tea drink"'or as a main ingredient of some herbal formulations, which implies that the herb and/or its products are now more likely to be concurrently administered with conventional medicines for losing body weight and reducing blood lipids. However, information on the leaf extract and its active compounds that cause HDI by inhibition of P450is limited. In the present study, we investigated the potential HDI of lotus leaves that mediated by P450through in vitro or in vivo models. Firsty, the inhibitory effects on the CYP activities in human liver microsomes were evaluated by in vitro model, and later were verified by in vivo model. Finally, the analysis of chemical constituents of lotus leaves in rats and their tissue distribution were investigated to interpret the mechanism of lotus leaves leading to the potential HDI. In addition, the P-gp function was preliminary investgated by Caco-2cell model. The main contents of this paper are as follows:
     1. By using LC-MS/MS techniques, an in vitro cocktail method was established for simultaneous determination of five major cytochrome P450isoenzymes activities by monitoring the metabolites of probe substrates, including omeprazole (CYP2C19), dextromethorphan (CYP2D6), testosterone (CYP3A4), chlorzoxazone (CYP2E1), tolbutamide (CYP2C9), in incubation human liver microsomes with mixed the five probe substrates. Inhibition activities of12TCMs including Nelumbinis Folium, Ziziphi Spinosae Semen, Cassiae Semen, Schisandrae Chinensis Fructus, Crataegi Fructus, Lycii Fructus, Nelumbinis Plumula, Ginseng Radix Et Rhizoma, Chrysanthemi Flos, Polygonati Rhizoma, Poria and Lonicerae Japonicae Flos on the five isoenzymes were evaluated using established method. The results indicated that the method possessed a good linearity and sensitivity, as well as the excellent precession and accuracy, and can be used for the high throughput screening of P450enzyme activities. In addition, aqueous extracts of12TCMs showed a weak or no inhibitory effects on P450enzymes, however, their ethanolic extracts possessed more potent and concentration-dependent inhibition effects (except for the Lycii Fructus). Among them, the lotus leaf ethanolic extract strongly inhibited the CYP2C19, CYP2D6, CYP3A4, CYP2E1and CYP2C9activities with IC50values of77.38,12.05,98.01,61.43and83.46μg/mL, respectively; chrysanthemum Flos ethanolic extract inhibited the CYP2C9activity with IC50value of45.25μg/mL; Schisandrae Chinensis Fructus ethanolic extract inhibited CYP3A4and CYP2C19activities with IC50values of47.24and64.42μg/mL, respectively. These results suggest that the possible HDI between the herb ethanolic extracts and their preparations with conventional medicines should thus be taken into account.
     2. The inhibitory effects of the total flavonoids and total alkaloids of lotus leaves, as well as its main alkaloid compounds on the activities of human liver microsomal P450enzymes were investigated by the established cocktail probe substrate method. The results showed that the total flavonoids weakly inhibited P450enzyme activities, with IC50values around100μg/mL; while, the total alkaloids strongly inhibited the activities of CYP2C19, CYP2D6, CYP3A4, CYP2E1and CYP2C9with IC50value of40.79,0.96, 44.87,63.84and52.58μg/mL, respectively. The individual alkaloids, namely, nuciferine (NF), N-nornuciferine (N-NF), and2-hydroxy-l-methoxyaporphine (HMA) competitively inhibited CYP2D6activity with Ki values of1.88,2.34, and1.56μM, respectively. And the three alkaloids are not mechanism-based inhibitors of CYP2D6.
     The effects of lotus leaf ethanolic extract, total flavonoids and total alkaloids on the function of P-glycoprotein (P-gp) were investigated by using Caco-2cell model and a P-gp substrate Rhodamine-123(Rho-123). The result showed that the ethanolic extract and total alkaloids could significantly increase the uptake of Rho-123by Caco-2cells at the concentration of100μg/mL (P<0.05), this indicates that they can inhibit the function of P-gp. The total flavonoids showed a weak inhibition effect on P-gp, but no significant difference comparing with negative control (P>0.05).
     All these results suggest that lotus leaves have inhibitory effects on P450enzyme activities and P-gp function and the effects may probable related with its alkaloid components. There are potential drug interactions between total alkaloids and other therapeutic drugs.
     3. The in vivo inhibition effects of lotus leaf total alkaloids on CYP2D6activity was investigated in rats using dextromethorphan and metoprolol as probe substrates. After being pretreated with total alkaloids (50mg/kg) for continuous7days, the rats was intravenously administrated with dextromethorphan (5mg/kg) or metoprolol (10mg/kg) at2or12h time point of the last alkaloid dosing. The plasma concentrations of the substrate metabolites, dextrorphan and a-hydroxymetoprolol were determined by LC-MS/MS method, for comparing their pharmacokinetic properties between the treated and untreated rats, and protein expression of CYP2D6in rat liver was also determined by western blot. The results showed total alkaloids significantly decreased the plasma concentrations of the two probe metabolites, AUCall of dextrorphan was significantly decreased from167.27to43.13h-ng/mL (P<0.01)(given at2h after the last dose of alkaloids) and62.25h-ng/mL (P<0.05)(given at12h after the last dose of alkaloids), respectively; and AUCall of a-hydroxy-metoprolol were also significantly decreased from347.68to223.24h-ng/mL (P<0.05)(given at2h after the last dose of alkaloids). The total alkaloids had no effect on the protein expression of CYP2D6in rat liver. These results suggest that the total alkaloids have significant inhibition on rat CYP2D6enzyme, which was consistent with the in vitro results.
     4. Based on developed LC-MS/MS methods, the in vivo components of lotus leaf extracts were identified after oral administration of total flavonoids and total alkaloids to rats. In addition, the tissue distribution of alkaloids was also studied after intravenous administration to rats. A total of37flavonoid compounds, including parent flavonoids and their glucuronidation and sulfation metabolites, were identified in rat plasma and urine. This indicates that flavonoids are mainly biotransformed by phase II metabolism pathway. A total of18alkaloids were identified, including parent alkaloids and their oxidation, methylation, glucuronidation and sulfation metabolites, were also identified in rat plasma and urine. This indicates that the alkaloids were biotransformed by both phase I and phase II metabolism pathways. The studies on tissue distribution of alkaloids showed that NF, N-NF and HMA could quickly (within5min) distribute to various tissues, especially in the liver, lungs, spleens and kidneys with a relative high concentrations, and could remained in the body for a relative long period. It is speculated that the inhibitory effects of lotus leaves on P450enzyme activities are likely to relate to these in vivo components.
     In conclusion, the present study provides systematic investigations on the in vitro inhibitory effects of lotus leaf ethanolic extracts, and its main two fractions total flavonoids and total alkaloids on P450enzyme activities, particularly the CYP2D6isoenzyme, which was further confirmed by an in vivo study. Meanwhile, the effect of lotus leaf ethanolic extract and the two fractions on the function of P-gp was preliminary evaluated by using a Caco-2cell model. In addition, the in vivo components of lotus leave flavonoids and alkaloids, as well as the tissue distribution of alkaloids in rats were also investigated. The results reveals that the lotus leaves can inhibit the activities of P450enzymes, especially for CYP2D6, and inhibit P-gp function. These effects are mainly related to the alkaloids existed in lotus leaves, this might cause the potential HDI and should pay much attentions when used in clinic. The present studies provide new information for safe and appropriate use of lotus leaves, and also provide ideas for further investigation of pharmacology and toxicology, and further development and utilization of the herb.
引文
[1]CHAN E, TAN M, XIN J, et al. Interactions between traditional Chinese medicines and Western therapeutics [J]. Current opinion in drug discovery & development,2010, 13(1):50-65.
    [2]FOSTER B C, ARNASON J T, BRIGGS C J. Natural health products and drug disposition [J]. Annu Rev Pharmacol Toxicol,2005,45:203-226.
    [3]BENT S. Herbal medicine in the United States:review of efficacy, safety, and regulation:grand rounds at University of California, San Francisco Medical Center [J]. J Gen Intern Med,2008,23(6):854-859.
    [4]KENNEDY J, WANG C C, WU C H. Patient Disclosure about Herb and Supplement Use among Adults in the US [J]. Evidence-based complementary and alternative medicine:eCAM,2008,5(4):451-456.
    [5]BARANCZEWSKI P, STANCZAK A, KAUTIAINEN A, et al. Introduction to early in vitro identification of metabolites of new chemical entities in drug discovery and development. [J]. Pharmacol Rep,2006,58(3):314-352.
    [6]LEUNG L, KALGUTKAR A S, OBACH R S. Metabolic activation in drug-induced liver injury [J]. Drug Metab Rev,2012,44(1):18-33.
    [7]ZHOU S, GAO Y, JIANG W, et al. Interactions of herbs with cytochrome P450 [J]. Drug Metab Rev,2003,35(1):35-98.
    [8]LEUCUTA S E, VLASE L. Pharmacokinetics and metabolic drug interactions [J]. Current clinical pharmacology,2006,1(1):5-20.
    [9]BENZ F W, NERLAND D E. Effect of cytochrome P450 inhibitors and anticonvulsants on the acute toxicity of acrylonitrile [J]. Arch Toxicol,2005,79(10): 610-614.
    [10]冷欣夫,邱星辉.细胞色素P450酶系的结构功能与应用前景[M].北京:科学出版社,2001.
    [11]SELISKAR M, ROZMAN D. Mammalian cytochromes P450--importance of tissue specificity [J]. Biochimica et biophysica acta,2007,1770(3):458-466.
    [12]NEBERT D W, NELSON D R, COON M J, et al. The P450 superfamily:update on new sequences, gene mapping, and recommended nomenclature [J]. Dna Cell Biol,1991, 10(1):1-14.
    [13]NELSON D R, KOYMANS L, KAMATAKI T, et al. P450 superfamily:update on new sequences, gene mapping, accession numbers and nomenclature [J]. Pharmacogenetics,1996,6(1):1-42.
    [14]SHIMADA T, YAMAZAKI H, MIMURA M, et al. Interindividual variations in human liver cytochrome P-450 enzymes involved in the oxidation of drugs, carcinogens and toxic chemicals:studies with liver microsomes of 30 Japanese and 30 Caucasians [J]. J Pharmacol Exp Ther,1994,270(1):414-423.
    [15]XIAO Z S, GOLDSTEIN J A, XIE H G, et al. Differences in the incidence of the CYP2C19 polymorphism affecting the S-mephenytoin phenotype in Chinese Han and Bai populations and identification of a new rare CYP2C19 mutant allele [J]. J Pharmacol Exp Ther,1997,281(1):604-609.
    [16]崔颖,张永旺.P450酶的研究进展[J].中国新技术新产品,2009,2(16):7-8.
    [17]MCNAUGHTON S A, MISHRA G D, PAUL A A, et al. Supplement use is associated with health status and health-related behaviors in the 1946 British birth cohort [J]. J Nutr,2005,135(7):1782-1789.
    [18]WILSON K M, KLEIN J D, SESSELBERG T S, et al. Use of complementary medicine and dietary supplements among U.S. adolescents [J]. The Journal of adolescent health:official publication of the Society for Adolescent Medicine,2006,38(4):385-394.
    [19]SINGH S R, LEVINE M A. Potential interactions between pharmaceuticals and natural health products in Canada [J]. J Clin Pharmacol,2007,47(2):249-258.
    [20]SANDSON N. Drug-drug interactions:the silent epidemic [J]. Psychiatr Serv,2005, 56(1):22-24.
    [21]张洁,谭浩.利福平与其他药物相互作用研究进展[J].天津药学,2009,21(1):69-72.
    [22]张石革,宋菲.回顾若干处方药的淘汰史,重视药物的临床再评价[J].中国医院用药评价与分析,2002,2(4):195-198.
    [23]郭晓昕,杜晓曦,薛松林.从美国药品撤市看药品安全风险管理[J].中国药物警戒,2011,8(4):212-216.
    [24]ALFARO C L. Emerging role of drug interaction studies in drug development:the good, the bad, and the unknown [J]. Psychopharmacology bulletin,2001,35(4):80-93.
    [25]陈旺青,张伟.撤市药物的遗传药理学[J].中国临床药理学与治疗学,2011,16(9):1065-1071.
    [26]王爱平,李若瑜.特比萘芬、伊曲康唑和氟康唑不良反应和药物相互作用评价[J].中国真菌学杂志,2010,15(1):44-47.
    [27]周权,张仲苗,周宏民,等.2种他汀类药物代谢性相互作用处方分析[J].中国药学杂志,2008,43(2):156-158.
    [28]刘彦卿,洪燕君,曾苏.代谢性药物-药物相互作用的研究进展[J].浙江大学学报(医学版),2009,38(2):215-224.
    [29]IZZO A A, DI CARLO G, BORRELLI F, et al. Cardiovascular pharmacotherapy and herbal medicines:the risk of drug interaction [J]. Int J Cardiol,2005,98(1):1-14.
    [30]YANG L J, FAN L, LIU Z Q, et al. Effects of allicin on CYP2C19 and CYP3A4 activity in healthy volunteers with different CYP2C19 genotypes [J]. Eur J Clin Pharmacol,2009,65(6):601-608.
    [31]董宇,王阶,杨庆,等.CYP450酶与中药代谢相互作用关系研究概况[J].中国中医药信息杂志,2011,18(1):100-103.
    [32]艾常虹,孙汉雄,李桦,等.中药有效成分对细胞色素P450酶的抑制活性评价[J].中国药理学通报,2011,27(4):519-523.
    [33]LIU L, XIAO J, PENG Z H, et al. In vitro metabolism of glycyrrhetic acid by human cytochrome P450 [J]. Yao xue xue bao= Acta pharmaceutica Sinica,2011,46(1):81-87.
    [34]杨静,彭仁琇,孔锐,等.18α-甘草酸二铵对大鼠肝脏细胞色素P450和Ⅱ相酶的影响[J].药学学报,2001,36(5):321-324.
    [35]FDA. Guidance for Industry Drug Interaction Studies-Study Design, Data Analysis, and Implications for Dosing and Labelinge. Draft Guidance [S]. http://wwwfdagov/cder/guidance/indexhtm.2006.
    [36]BJORNSSON T D, CALLAGHAN J T, EINOLF H J, et al. The conduct of in vitro and in vivo drug-drug interaction studies:a Pharmaceutical Research and Manufacturers of America (PhRMA) perspective [J]. Drug Metab Dispos,2003,31(7):815-832.
    [37]胡云珍.地非三唑在鼠肝微粒体中的体外代谢以及对细胞色素P450的体内诱导和体外抑制作用[D];浙江大学,2004.
    [38]BREIMER D D, SCHELLENS J H. A'cocktail'strategy to assess in vivo oxidative drug metabolism in humans [J]. Trends Pharmacol Sci.1990,11(6):223-225.
    [39]KIM M J, KIM H, CHA I J, et al. High-throughput screening of inhibitory potential of nine cytochrome P450 enzymes in vitro using liquid chromatography/tandem mass spectrometry [J]. Rapid communications in mass spectrometry,2005,19(18):2651-2658.
    [40]FANG Z Z, ZHANG Y Y, GE G B, et al. Time-dependent inhibition (TDI) of CYP3A4 and CYP2C9 by noscapine potentially explains clinical noscapine-warfarin interaction [J]. Br J Clin Pharmacol,2010,69(2):193-199.
    [41]KARJALAINEN M J, NEUVONEN P J, BACKMAN J T. Rofecoxib is a potent, metabolism-dependent inhibitor of CYP1A2:implications for in vitro prediction of drug interactions [J]. Drug Metab Dispos,2006,34(12):2091-2096.
    [42]BROWN H S, ITO K, GALETIN A, et al. Prediction of in vivo drug-drug interactions from in vitro data:impact of incorporating parallel pathways of drug elimination and inhibitor absorption rate constant [J]. Br J Clin Pharmacol,2005,60(5): 508-518.
    [43]曹国颖,武秀忠,胡欣,等.那格列奈在大鼠游离肝细胞中的代谢特点[J].中国新药杂志,2004,13(8):699-703.
    [44]BORN S L, HU J K, LEHMAN-MCKEEMAN L D. o-hydroxyphenylacetaldehyde is a hepatotoxic metabolite of coumarin [J]. Drug Metab Dispos,2000,28(2):218-223.
    [45]张丽芳,胡晓,王萍,等.蛇床子素在大鼠肝细胞代谢的体外研究[J].药学学报,2009,44(10):1131-1135.
    [46]ZHANG W, RAMAMOORTHY Y, KILICARSLAN T, et al. Inhibition of cytochromes P450 by antifungal imidazole derivatives [J]. Drug Metab Dispos,2002, 30(3):314-318.
    [47]CHING M S, BICHARA N, BLAKE C L, et al. Propranolol 4- and 5-hydroxylation and N-desisopropylation by cloned human cytochrome P4501A1 and P4501A2 [J]. Drug Metab Dispos,1996,24(6):692-694.
    [48]CHU V, EINOLF H J, EVERS R, et al. In vitro and in vivo induction of cytochrome p450:a survey of the current practices and recommendations:a pharmaceutical research and manufacturers of america perspective [J]. Drug Metab Dispos,2009,37(7): 1339-1354.
    [49]EKINS S, RING B J, GRACE J, et al. Present and future in vitro approaches for drug metabolism [J]. J Pharmacol Toxicol Methods,2000,44(1):313-324.
    [50]何益军,石苏英,金科涛,等.甘草与大戟甘遂芫花配伍对大鼠肝脏细胞色素P4501A2酶活性的影响[J].中国药物与临床,2007,7(4):278-280.
    [51]王宇光.基于药物代谢酶的中药相互作用研究[D];中国人民解放军军事医学科学院,2006.
    [52]夏成云,高月,周京国,等.大戟配伍甘草对大鼠肝功能及肝脏微粒体中CYP3A2的影响[J].中国中医急症,2006,15(9):1013-1015+1066.
    [53]王宇光,高月,柴彪新,等.人参、藜芦合用对大鼠肝P450酶活性及mRNA表达的调控作用[J].中国中药杂志,2004,29(4):82-86.
    [54]石苏英,金科涛,王宇光,等.白蔹配伍乌头对大鼠肝脏CYP450的调节作用[J].医药导报,2007,26(9):975-979.
    [55]和凡,钟国平,赵立子,等.丹参酮ⅡA对大鼠细胞色素P450酶的诱导作用[J].中草药,2009,40(6):938-942.
    [56]刘高峰,甄立棉,黄丽军.丹参注射液对家兔体内CYP1A2、CYP2D6和CYP3A4活性的影响[J].中国临床药理学与治疗学,2010,15(7):775-780.
    [57]樊慧蓉.“Cocktail"探针药物法评价人参皂苷Re对肝细胞色素P450酶亚型的影响[D];天津医科大学,2004.
    [58]韩晓文.“Cocktail"探针药物法评价苦参对大鼠细胞色素P450不同亚型体内代谢活性的影响[D];天津医科大学,2011.
    [59]工军升,周宏灏.CYP3A基因表达与调控的分子机制[J].生理科学进展,1998,29(2):79-82.
    [60]SOMOGYI A, MUIRHEAD M. Pharmacokinetic interactions of cimetidine 1987 [J]. Clin Pharmacokinet,1987,12(5):321-366.
    [61]WAGNER F, KALUSCHE D, TRENK D, et al. Drug interaction between propafenone and metoprolol [J]. Br J clin Pharmac,1987,24:213-220.
    [62]AHMAD S. Metoprolol-induced delirium perpetuated by propafenone [J]. Am Fam Physician,1991,44(4):1142-1144.
    [63]刘勇.早期药物代谢研究在中药科学性评价中的应用[D];中国科学院研究生院(大连化学物理研究所),2006.
    [64]VON MOLTKE L L, GREENBLATT D J, SCHMIDER J, et al. In vitro approaches to predicting drug interactions in vivo [J]. Biochem Pharmacol,1998,55(2):113-122.
    [65]TRACY T S. Atypical cytochrome p450 kinetics:implications for drug discovery [J]. Drugs in R&D,2006,7(6):349-363.
    [66]MAD AN A, USUKI E, BURTON LA, et al. In Vitro Approaches for Studying the Inhibition of Drug-Metabolizing Enzymes and Identifying the Drug-Metabolizing Enzymes Responsible for the Metabolism of Drugs (Reaction Phenotyping) with Emphasis on Cytochrome P450. In:Rodrigues AD, editor. [M]. Drug-Drug Interactions New York:Marcel Dekker.2002 p.217-294.
    [67]TAKAHASHI N, SUBEHAN, KADOTA S, et al. Mechanism-based CYP2D6 inactivation by acridone alkaloids of Indonesian medicinal plant Lunasia amara [J]. Fitoterapia,2012,83(4):774-779.
    [68]SUBEHAN, USIA T, IWATA H, et al. Mechanism-based inhibition of CYP3A4 and CYP2D6 by Indonesian medicinal plants [J]. J Ethnopharmacol,2006,105(3):449-455.
    [69]AMUNUGAMA H T, ZHANG H, HOLLENBERG P F. Mechanism-based inactivation of cytochrome P450 2B6 by methadone through destruction of prosthetic heme [J]. Drug Metab Dispos,2012,40(9):1765-1770.
    [70]LU W J, BIES R, KAMDEN L K, et al. Methadone: a substrate and mechanism-based inhibitor of CYP19 (aromatase) [J]. Drug Metab Dispos,2010,38(8): 1308-1313.
    [71]TASSANEEYAKUL W, GUO L Q, FUKUDA K, et al. Inhibition selectivity of grapefruit juice components on human cytochromes P450 [J]. Arch Biochem Biophys, 2000,378(2):356-363.
    [72]CHUN Y J, RYU S Y, JEONG T C, et al. Mechanism-based inhibition of human cytochrome P450 1A1 by rhapontigenin [J]. Drug Metab Dispos,2001,29(4 Pt 1): 389-393.
    [73]CHAN W K, DELUCCHI A B. Resveratrol, a red wine constituent, is a mechanism-based inactivator of cytochrome P450 3A4 [J]. Life Sci,2000,67(25): 3103-3112.
    [74]SURH Y J, LEE S S. Capsaicin, a double-edged sword:toxicity, metabolism, and chemopreventive potential [J]. Life Sci,1995,56(22):1845-1855.
    [75]OBACH R S, WALSKY R L, VENKATAKRISHNAN K. Mechanism-Based Inactivation of Human Cytochrome P450 Enzymes and the Prediction of Drug-Drug Interactions [J]. Drug Metab Dispos,2006,35(2):246-255.
    [76]OKEY A B. Enzyme induction in the cytochrome P-450 system [J]. Pharmacol Ther, 1990,45(2):241-298.
    [77]HEWITT N J, LECLUYSE E L, FERGUSON S S. Induction of hepatic cytochrome P450 enzymes:methods, mechanisms, recommendations, and in vitro-in vivo correlations [J]. Xenobiotica,2007,37(10-11):1196-1224.
    [78]USIA T, IWATA H, HIRATSUKA A, et al. CYP3A4 and CYP2D6 inhibitory activities of Indonesian medicinal plants [J]. Phytomedicine,2006,13(1-2):67-73.
    [79]SEVIOR D K, HOKKANEN J, TOLONEN A, et al. Rapid screening of commercially available herbal products for the inhibition of major human hepatic cytochrome P450 enzymes using the N-in-one cocktail [J]. Xenobiotica,2010,40(4): 245-254.
    [80]MASIMIREMBWA C M, THOMPSON R, ANDERSSON T B. In vitro high throughput screening of compounds for favorable metabolic properties in drug discovery [J]. Comb Chem High Throughput Screen,2001,4(3):245-263.
    [81]DIERKS E A, STAMS K R, LIM H K, et al. A method for the simultaneous evaluation of the activities of seven major human drug-metabolizing cytochrome P450s using an in vitro cocktail of probe substrates and fast gradient liquid chromatography tandem mass spectrometry [J]. Drug Metab Dispos,2001,29(1):23-29.
    [82]THUMMEL K E. In vitro and in vivo drug interactions involving human CYP3A4 [J]. Annu Rev Pharmacol Toxicol,1998,38:389-430.
    [83]FDA. Guidance for Industry Drug Interaction Studie—Study Design, Data Analysis, Implications for Dosing, and Labeling Recommendations [S]. http://wwwfdagov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/default htm.2012.
    [84]HU Z Y, PARKER R B, LAIZURE S C. In vivo information-guided prediction approach for assessing the risks of drug-drug interactions associated with circulating inhibitory metabolites [J]. Drug Metab Dispos,2012,40(8):1487-1494.
    [85]LIN H Y, KUO Y H, LIN Y L, et al. Antioxidative effect and active components from leaves of Lotus (Nelumbo nucifera) [J]. J Agric Food Chem,2009,57(15): 6623-6629.
    [86]ONO Y, HATTORI E, FUKAYA Y, et al. Anti-obesity effect of Nelumbo nucifera leaves extract in mice and rats [J]. J Ethnopharmacol,2006,106(2):238-244.
    [87]方建国,李艳艳,万进,等.反相高效液相色谱法测定荷叶口服液中荷叶碱的含量[J].医药导报,2010,29(3):367-369.
    [88]陈锐.荷叶丸临床应用解析[J].中国社区医师,2012,9(9):13.
    [89]CHEN J, MA X, GAO K, et al. The active ingredients of Jiang-Zhi-Ning:study of the Nelumbo nucifera alkaloids and their main bioactive metabolites [J]. Molecules,2012, 17(8):9855-9867.
    [90]张婉婷,王登良.荷叶茶的研究进展[J].广东茶业,2010,10(5):19-22.
    [91]刘淑萍,樊淑彦,侯海妮,等.荷叶化学成分及药理作用研究进展[J].河北医科大学学报,2004,25(4):254-256.
    [92]王福刚,曹娟,刘斌,等.荷叶的化学成分及其药理作用研究进展[J].时珍国医国药,2010,21(9):2339-2340.
    [93]张国庆,冯习坤,曾为驰,等.荷叶的化学和药理研究进展[J].药学实践杂志,2010,5(28):328-330.
    [94]CHEN S, FANG L, XI H, et al. Simultaneous qualitative assessment and quantitative analysis of flavonoids in various tissues of lotus (Nelumbo nucifera) using high performance liquid chromatography coupled with triple quad mass spectrometry [J]. Anal Chim Acta,2012.724:127-135.
    [95]赵小亮.荷叶化学成分和黄山药皂苷类化学成分及生物活性的研究[D];北京协和医学院,2011.
    [96]王玲玲,刘斌,石任兵.荷叶的化学成分研究[J].天然产物研究与开发,2009,21(3):416-419.
    [97]肖桂青,卢向阳,田云,等.荷叶中生物碱类成分的研究进展[J].化学与生物工程,2006,5(23):1-2+8.
    [98]何晓曦,孔令提,张泽生,等.不同产地和不同采收期荷叶中生物碱的含量变化[J].天然产物研究与开发,2013,05):652-655+661.
    [99]曾虹燕,苏杰龙,方芳,等.不同方法提取的荷叶挥发油化学成分分析[J].西北植物学报,2005,25(3):578-582.
    [100]缪存铅,张赞彬.超临界CO2萃取与水蒸气蒸馏提取荷叶挥发油的比较研究[J].食品科技,2009,11:216-220.
    [101]尹慧晶,钱一帆,濮存海.均匀设计法优化荷叶超临界CO2萃取工艺及萃取物GC-MS分析[J].中药材,2007,4(30):464-466.
    [102]曾虹燕,冯波,方芳,等.超临界CO2萃取荷叶挥发油工艺的研究[J].中南林学院学报,2005,2(25):42-45.
    [103]CHAUDHURI P K, SINGH D. A new triterpenoid from the rhizomes of Nelumbo nucifera [J]. Natural product research,2013,27(6):532-536.
    [104]关章顺,吴俊,喻泽兰,等.荷叶水提物对人体高脂血症的降脂效果研究[J].郴州医学高等专科学校学报,2003,5(3):3-6.
    [105]朱晓青,吕敬慈,霍世欣,等.荷叶生物碱对胰脂肪酶的抑制作用[J].上海大学学报(自然科学版),2007,17(1):85-87+93.
    [106]陈希平,杨鹏,张阳德.荷叶中生物碱的分离纯化及脂肪酶活性抑制作用研究[J].农业现代化研究,2009,6(30):748-751+760.
    [107]ZHANG A, ZHANG Y, BRANFMAN A R, et al. Advances in development of dopaminergic aporphinoids [J]. J Med Chem,2007,50(2):171-181.
    [108]KASHIWADA Y, AOSHIMA A, IKESHIRO Y, et al. Anti-HIV benzylisoquinoline alkaloids and flavonoids from the leaves of Nelumbo nucifera, and structure-activity correlations with related alkaloids [J]. Bioorg Med Chem,2005,13(2):443-448.
    [109]BOUSTIE J, STIGLIANI J L, MONTANHA J, et al. Antipoliovirus structure-activity relationships of some aporphine alkaloids [J]. J Nat Prod,1998,61(4): 480-484.
    [110]WU Y B, ZHENG L J, WU J G, et al. Antioxidant activities of extract and fractions from receptaculum nelumbinis and related flavonol glycosides [J]. International journal of molecular sciences,2012,13(6):7163-7173.
    [111]赵奇志,赵毅民.阿朴菲类生物碱生物活性研究进展[J].天然产物研究与开发,2006,18(2):316-324.
    [112]蒋益虹.荷叶抑菌活性成分的研究[D];浙江大学,2007.
    [113]LI M, XU Z. Quercetin in a lotus leaves extract may be responsible for antibacterial activity [J]. Arch Pharm Res,2008,31(5):640-644.
    [114]纪丽莲.荷叶中抑菌成分的提取及其抑菌活性的研究[J].食品科学,1999,20(8):64-66.
    [115]陈健芬,何卫华,钱伏刚.荷叶提取物口腔抑菌有效成分的定性定量分析[J].日用化学工业,2003,33(1):49-51+63.
    [116]MCLENNAN H, WHEAL H V. The specificity of action of three possible antagonists of amino acid-induced neuronal excitations [J]. Neuropharmacology,1976, 15(11):709-712.
    [117]FELIX D, FRANGI U. Dimethoxyaporphine as an antagonist of chemical excitation in the pigeon optic tectum [J]. Neurosci Lett,1977,4(6):347-350.
    [118]BHATTACHARYA S K, BOSE R, GHOSH P, et al. Psychopharmacological studies on (--)-nuciferine and its Hofmann degradation product atherosperminine [J]. Psychopharmacology (Berl),1978,59(1):29-33.
    [119]MARTINEZ-VAZQUEZ M, ESTRADA-REYES R, ARAUJO ESCALONA A G, et al. Antidepressant-like effects of an alkaloid extract of the aerial parts of Annona cherimolia in mice [J]. J Ethnopharmacol,2012,139(1):164-170.
    [120]YANG M, CHANG Y, CHAN K, et al. Flavonoid-enriched extracts from Nelumbo nucifera leaves inhibits proliferation of breast cancer in vitro and in vivo [J]. European Journal of Integrative Medicine,2011,3(3):e153-e163.
    [121]WU Y C, YAMAGISHI T, LEE K H. Cytotoxic isoquinoline alkaloids from Xanthorhiza simplicissima [J]. Gaoxiong yi xue ke xue za zhi= The Kaohsiung journal of medical sciences,1989,5(7):409-411.
    [122]JUNG H A, JUNG Y J, YOON N Y, et al. Inhibitory effects of Nelumbo nucifera leaves on rat lens aldose reductase, advanced glycation endproducts formation, and oxidative stress [J]. Food Chem Toxicol,2008,46(12):3818-3826.
    [123]HUANG C F, CHEN Y W. YANG C Y, et al. Extract of lotus leaf (Nelumbo nucifera) and its active constituent catechin with insulin secretagogue activity [J]. J Agric Food Chem,2011,59(4):1087-1094.
    [124]PAN Y, CAI B, WANG K, et al. Neferine enhances insulin sensitivity in insulin resistant rats [J]. J Ethnopharmacol,2009,124(1):98-102.
    [125JNGUYEN K H, TA T N, PHAM T H, et al. Nuciferine stimulates insulin secretion from beta cells-an in vitro comparison with glibenclamide [J]. J Ethnopharmacol,2012, 142(2):488-495.
    [126]QIAN J Q. Cardiovascular pharmacological effects of bisbenzylisoquinoline alkaloid derivatives [J]. Acta Pharmacol Sin,2002,23(12):1086-1092.
    [127]肖华,陈爱华,季爱民,等.前荷叶碱对人脐静脉内皮细胞一氧化氮及一氧化氮合酶的影响[J].中药材,2005,28(6):503-505.
    [128]JAIN A, SONI M, DEB L, et al. Antioxidant and hepatoprotective activity of ethanolic and aqueous extracts of Momordica dioica Roxb. leaves [J]. J Ethnopharmacol, 2008,115(1):61-66.
    [129]AGNIHOTRI V K, ELSOHLY H N, KHAN S I, et al. Constituents of Nelumbo nucifera leaves and their antimalarial and antifungal activity [J]. Phytochemistry Letters, 2008,1(2):89-93.
    [130]刘虹,郝或,朱晓月,等.HPLC测定血浆中荷叶碱的浓度及其在大鼠体内的药代动力学研究[J].中国实验方剂学杂志,2010,16(12):165-168.
    [131]刘淑萍.荷叶中生物碱的制备及其药理学活性研究[D];河北医科大学,2004.
    [132]谢水林,王翌尧,谭玉溪,等.荷叶碱在大鼠体内的绝对生物利用度研究[J].广东药学院学报,2010,26(1):17-19.
    [133]李晓宇,周尔君,顾圣莹,等.荷叶碱大鼠肠道吸收特性研究[J].中国药学杂志,2011,46(18):1417-1420.
    [134]王玉霞,刘斌,石任兵,等.RP-HPLC法测定Beagle犬血浆中荷叶碱的浓度及药动学研究[J].药物分析杂志,2008,28(9):1418-1421.
    [135]吴昊,刘斌,王伟,等.LC-MSn鉴定2-羟基-1-甲氧基阿朴啡在Beagle犬体内的主要代谢产物[J].中国药学杂志,2009,44(20):1574-1577.
    [136]HU L, XU W, ZHANG X, et al. In-vitro and in-vivo evaluations of cytochrome P450 1A2 interactions with nuciferine [J]. J Pharm Pharmacol,2010,62(5):658-662.
    [137]国家药典委员会.中国药典[S].北京;中国医药科技出版社.2010.
    [138]陈建维,饶印保,文皓颞.HPLC法测定荷叶调脂茶中荷叶碱的含量[J].江西中医药,2012,43(12):65-66.
    [139]CHEN J, ZHAO H, YANG Y, et al. Lipid-lowering and antioxidant activities of Jiang-Zhi-Ning in Traditional Chinese Medicine [J]. J Ethnopharmacol,2011,134(3): 919-930.
    [140]许鸿超.降脂Ⅰ号Ⅱ号胶囊治疗高脂血症106例临床观察[J].河北中医,1996,18(4):16-17.
    [141]何嘉琳.荷叶露药用谈[J].浙江中医杂志,1993,28(6):267.
    [142]崔冬生.鲜荷叶在儿科疾病中的应用[J].四川中医,1996,14(11):23.
    [143]高继红.HPLC法测定脂定宁中荷叶碱的含量[J].光明中医,2008,23(10):1449-1450.
    [144]汪应梅.荷叶可作畜禽饲料[J].四川农业科技,2006,7:36.
    [145]陈俊鹏,王劫,郭福有,等.日粮中添加荷叶提取物对黄羽肉鸡生长性能和肉品质的影响[J].养禽与禽病防治,2010,11:8-11.
    [146]梁晓慧,徐新娟.肥胖并发高血压的机制及治疗进展[J].心血管病学进展,2006,27(5):608-611.
    [147]王晓健.肥胖与糖尿病的关系及发病机制的研究进展[J].中国疗养医学,2011,20(8):723-725.
    [148]卢盛华,肇玉明.肥胖病因及其药物治疗研究进展[J].中国药理学通报,2001, 17(6):614-616.
    [1]ERNST E. The role of complementary and alternative medicine [J]. BMJ,2000, 321(7269):1133-1135.
    [2]FOSTER B C, ARNASON J T, BRIGGS C J. Natural health products and drug disposition [J]. Annu Rev Pharmacol Toxicol,2005,45:203-226.
    [3]MCNAUGHTON S A, MISHRA G D, PAUL A A, et al. Supplement use is associated with health status and health-related behaviors in the 1946 British birth cohort
    [J]. J Nutr,2005,135(7):1782-1789. [4] WILSON K M, KLEIN J D, SESSELBERG T S, et al. Use of complementary medicine and dietary supplements among U.S. adolescents [J]. The Journal of adolescent health:official publication of the Society for Adolescent Medicine,2006,38(4): 385-394.
    [5]SINGH S R, LEVINE M A. Potential interactions between pharmaceuticals and natural health products in Canada [J]. J Clin Pharmacol,2007,47(2):249-258.
    [6]CHANG G W, KAM P C. The physiological and pharmacological roles of cytochrome P450 isoenzymes [J]. Anaesthesia,1999,54(1):42-50.
    [7]LEUCUTA S E, VLASE L. Pharmacokinetics and metabolic drug interactions [J]. Current clinical pharmacology,2006,1(1):5-20.
    [8]SANDSON N. Drug-drug interactions:the silent epidemic [J]. Psychiatr Serv,2005, 56(1):22-24.
    [9]ZHOU S, GAO Y, JIANG W, et al. Interactions of herbs with cytochrome P450 [J]. Drug Metab Rev,2003,35(1):35-98.
    [10]CHAN E, TAN M, XIN J, et al. Interactions between traditional Chinese medicines and Western therapeutics [J]. Current opinion in drug discovery & development,2010, 13(1):50-65.
    [11]IZZO A A, DI CARLO G, BORRELLI F, et al. Cardiovascular pharmacotherapy and herbal medicines:the risk of drug interaction [J]. Int J Cardiol,2005.98(1):1-14.
    [12]YANG L J, FAN L, LIU Z Q, et al. Effects of allicin on CYP2C19 and CYP3A4 activity in healthy volunteers with different CYP2C19 genotypes [J]. Eur J Clin Pharmacol,2009,65(6):601-608.
    [13]CHAURET N, GAUTHIER A, NICOLL-GRIFFITH D A. Effect of common organic solvents on in vitro cytochrome P450-mediated metabolic activities in human liver microsomes [J]. Drug Metab Dispos,1998,26(1):1-4.
    [14]BUSBY W F, JR., ACKERMANN J M, CRESPI C L. Effect of methanol, ethanol, dimethyl sulfoxide, and acetonitrile on in vitro activities of cDNA-expressed human cytochromes P-450 [J]. Drug Metab Dispos,1999,27(2):246-249.
    [15]HE F, BI H C, XIE Z Y, et al. Rapid determination of six metabolites from multiple cytochrome P450 probe substrates in human liver microsome by liquid chromatography/mass spectrometry:application to high-throughput inhibition screening of terpenoids [J]. Rapid communications in mass spectrometry,2007,21(5):635-643.
    [16]TURPEINEN M, UUSITALO J, JALONEN J, et al. Multiple P450 substrates in a single run:rapid and comprehensive in vitro interaction assay [J]. Eur J Pharm Sci,2005, 24(1):123-132.
    [17]WU J, HUGHES C S, PICARD P, et al. High-throughput cytochrome P450 inhibition assays using laser diode thermal desorption-atmospheric pressure chemical ionization-tandem mass spectrometry [J]. Anal Chem,2007,79(12):4657-4665.
    [18]SHADER R I, GRANDA B W, VON MOLTKE L L, et al. Inhibition of human cytochrome P450 isoforms in vitro by zafirlukast [J]. Biopharm Drug Dispos,1999, 20(8):385-388.
    [19]TESTINO S A, JR., PATONAY G. High-throughput inhibition screening of major human cytochrome P450 enzymes using an in vitro cocktail and liquid chromatography-tandem mass spectrometry [J]. J Pharm Biomed Anal,2003,30(5): 1459-1467.
    [20]USIA T, IWATA H, HIRATSUKA A, et al. CYP3A4 and CYP2D6 inhibitory activities of Indonesian medicinal plants [J]. Phytomedicine,2006,13(1-2):67-73.
    [21]SEVIOR D K, HOKKANEN J, TOLONEN A, et al. Rapid screening of commercially available herbal products for the inhibition of major human hepatic cytochrome P450 enzymes using the N-in-one cocktail [J]. Xenobiotica,2010,40(4): 245-254.
    [22]IWATA H, TEZUKA Y, KADOTA S, et al. Identification and characterization of potent CYP3A4 inhibitors in Schisandra fruit extract [J]. Drug Metab Dispos,2004, 32(12):1351-1358.
    [23]MUKHERJEE P K, MUKHERJEE D, MAJI A K, et al. The sacred lotus (Nelumbo nucifera)-phytochemical and therapeutic profile [J]. J Pharm Pharmacol,2009,61(4): 407-422.
    [24]张婉婷,王登良.荷叶茶的研究进展[J].广东茶业,2010,10(5):19-22.
    [25]赵莉,唐生安.UPLC法同时测定荷丹片中8种指标成分[J].中草药,2013,44(16):2257-2260.
    [26]CHEN J, MA X, GAO K, et al. The active ingredients of Jiang-Zhi-Ning:study of the Nelumbo nucifera alkaloids and their main bioactive metabolites [J]. Molecules,2012, 17(8):9855-9867.
    [27]HU L, XU W, ZHANG X, et al. In-vitro and in-vivo evaluations of cytochrome P450 1A2 interactions with nuciferine [J]. J Pharm Pharmacol,2010,62(5):658-662.
    [28]LI W L, XIN H W, SU M W, et al. Inhibitory effects of schisandrin A and schisandrin B on CYP3A activity [J]. Methods Find Exp Clin Pharmacol,2010,32(3):163-169.
    [29]刘治军,傅得兴,孙春华,等.体内药物相互作用研究进展[J].药物不良反应杂志,2006,8(1):33-38.
    [30]刘彦卿,洪燕君,曾苏.代谢性药物-药物相互作用的研究进展[J].浙江大学学报(医学版),2009,38(2):215-224.
    [31]杨少林,李焕德,彭珍珍,等.由P-糖蛋白介导的药物相互作用研究进展[J].中国医院用药评价与分析,2011,11(6):494-497.
    [32]陈旺青,张伟.撤市药物的遗传药理学[J].中国临床药理学与治疗学,2011,16(9):1065-1071.
    [33]ALFARO C L. Emerging role of drug interaction studies in drug development:the good, the bad, and the unknown [J]. Psychopharmacology bulletin,2001,35(4):80-93.
    [34]鄢琳,程鲁榕,兰奋,等.国外药物撤销、限制使用的情况对我国药品评价的启示[J].中国临床药理学杂志,2002,18(6):470-474.
    [35]张伟霞,周宏灏.P-糖蛋白介导的药代动力学及其药物相互作用[J].中国临床药理学杂志,2004,20(2):139-143.
    [36]BALIMANE P V, PATEL K, MARINO A, et al. Utility of 96 well Caco-2 cell system for increased throughput of P-gp screening in drug discovery [J]. Eur J Pharm Biopharm,2004,58(1):99-105.
    [37]PAVEK P, STAUD F, FENDRICH Z, et al. Examination of the functional activity of P-glycoprotein in the rat placental barrier using rhodamine 123 [J]. J Pharmacol Exp Ther, 2003,305(3):1239-1250.
    [38]LIN H Y, KUO Y H, LIN Y L, et al. Antioxidative effect and active components from leaves of Lotus (Nelumbo nucifera) [J]. J Agric Food Chem,2009,57(15): 6623-6629.
    [39]ONO Y, HATTORI E, FUKAYA Y, et al. Anti-obesity effect of Nelumbo nucifera leaves extract in mice and rats [J]. J Ethnopharmacol,2006,106(2):238-244.
    [40]CHEN S, WU B H, FANG J B, et al. Analysis of flavonoids from lotus (Nelumbo nucifera) leaves using high performance liquid chromatography/photodiode array detector tandem electrospray ionization mass spectrometry and an extraction method optimized by orthogonal design [J]. J Chromatogr A,2012,1227:145-153.
    [41]WU Y B, ZHENG L J, WU J G, et al. Antioxidant activities of extract and fractions from receptaculum nelumbinis and related flavonol glycosides [J]. International journal of molecular sciences,2012,13(6):7163-7173.
    [42]LI M, XU Z. Quercetin in a lotus leaves extract may be responsible for antibacterial activity [J]. Arch Pharm Res,2008,31(5):640-644.
    [43]KASHIWADA Y, AOSHIMA A, IKESHIRO Y, et al. Anti-HIV benzylisoquinoline alkaloids and flavonoids from the leaves of Nelumbo nucifera, and structure-activity correlations with related alkaloids [J]. Bioorg Med Chem,2005,13(2):443-448.
    [44]YANG M, CHANG Y, CHAN K, et al. Flavonoid-enriched extracts from Nelumbo nucifera leaves inhibits proliferation of breast cancer in vitro and in vivo [J]. European Journal of Integrative Medicine,2011,3(3):e153-e163.
    [45]范婷婷,法鲁克,方芳,等.荷叶总生物碱降脂减肥作用的体内外试验[J].浙江大学学报(农业与生命科学版),2013,29(2):141-148.
    [46]KASHIWADA Y, AOSHIMA A, IKESHIRO Y, et al. Anti-HIV benzylisoquinoline alkaloids and flavonoids from the leaves of Nelumbo nucifera, and structure-activity correlations with related alkaloids [J]. Bioorg Med Chem,2005,13(2):443-448.
    [47]NGUYEN K H, TA T N, PHAM T H, et al. Nuciferine stimulates insulin secretion from beta cells-an in vitro comparison with glibenclamide [J]. J Ethnopharmacol,2012, 142(2):488-495.
    [48]PAN Y, CAI B, WANG K, et al. Neferine enhances insulin sensitivity in insulin resistant rats [J]. J Ethnopharmacol,2009,124(1):98-102.
    [49]陈爱华,肖华,李志梁,等.前荷叶碱对血管紧张素Ⅱ诱导人脐静脉内皮细胞凋亡的影响[J].中草药,2006,37(7):1045-1048.
    [50]肖华,陈爱华,季爱民,等.前荷叶碱对人脐静脉内皮细胞一氧化氮及一氧化氮合酶的影响[J].中药材,2005,28(6):503-505.
    [51]CHEN S, FANG L, XI H, et al. Simultaneous qualitative assessment and quantitative analysis of flavonoids in various tissues of lotus (Nelumbo nucifera) using high performance liquid chromatography coupled with triple quad mass spectrometry [J]. Anal Chim Acta,2012,724:127-135.
    [52]周永刚,刘畅,毛飞,等.荷叶化学成分的HPLC-TOF/MS分析[J].药学实践杂志,2011,29(5):342-346.
    [53]王玲玲,刘斌,石任兵.荷叶的化学成分研究[J].天然产物研究与开发,2009,21(3):416-419.
    [54]刘斌,石任兵,王伟,等.荷叶总生物碱提取物及其制备方法[P].中国专利:101085041,2007-12-12.
    [55]陈剑锋.一种采用阳离子交换树脂分离纯化荷叶碱的方法[P].中国专利:101139321,2008-03-12.
    [56]陈剑锋.从荷叶中分离制备荷叶碱和荷叶黄酮的方法[P].中国专利:101139320,2008-03-12.
    [57]邓泽元,单斌,范亚苇.同步分离提取与纯化荷叶中黄酮、生物碱和多糖的方法[P].中国专利:101402627,2009-04-08.
    [58]何晓曦,孔令提,张泽生,等.不同产地和不同采收期荷叶中生物碱的含量变化[J].天然产物研究与开发,2013,5(25):652-655+661.
    [59]PANG C Y, MAK J W, ISMAIL R, et al. In vitro modulatory effects of flavonoids on human cytochrome P450 2C8 (CYP2C8) [J]. Naunyn-Schmiedeberg's archives of pharmacology,2012,385(5):495-502.
    [60]LI J K, HE F, BI H C, et al. Inhibition of human cytochrome P-450 CYP1A2 by flavonoids:a quantitative structure-activity relationship study [J]. Acta Pharmaceutica Sinica (药学学报),2008,43(12):1198-1204.
    [61]KIMURA Y, ITO H, OHNISHI R, et al. Inhibitory effects of polyphenols on human cytochrome P450 3A4 and 2C9 activity [J]. Food Chem Toxicol,2010,48(1):429-435.
    [62]TAKAHASHI N, SUBEHAN, KADOTA S, et al. Mechanism-based CYP2D6 inactivation by acridone alkaloids of Indonesian medicinal plant Lunasia amara [J]. Fitoterapia,2012,83(4):774-779.
    [63]SUBEHAN, USIA T, IWATA H, et al. Mechanism-based inhibition of CYP3A4 and CYP2D6 by Indonesian medicinal plants [J]. J Ethnopharmacol,2006,105(3):449-455.
    [64]BROWN H S, ITO K, GALETIN A, et al. Prediction of in vivo drug-drug interactions from in vitro data:impact of incorporating parallel pathways of drug elimination and inhibitor absorption rate constant [J]. Br J Clin Pharmacol,2005,60(5): 508-518.
    [65]HAN Y, MENG X, LI D, et al. In Vitro Inhibition of Six Cytochrome P450 Isoforms in Human Liver Microsomes by Qingkailing Injection and Its Two Active Components (Baicalin and Geniposide) [J]. Chin Pharm J (中国药学杂志),2011,46(19):1486-1490.
    [66]刘勇.早期药物代谢研究在中药科学性评价中的应用[D];中国科学院研究生院(大连化学物理研究所),2006.
    [67]MASIMIREMBWA C M, THOMPSON R, ANDERSSON T B. In vitro high throughput screening of compounds for favorable metabolic properties in drug discovery [J]. Comb Chem High Throughput Screen,2001,4(3):245-263.
    [68]DIERKS E A, STAMS K R, LIM H K, et al. A method for the simultaneous evaluation of the activities of seven major human drug-metabolizing cytochrome P450s using an in vitro cocktail of probe substrates and fast gradient liquid chromatography tandem mass spectrometry [J]. Drug Metab Dispos,2001,29(1):23-29.
    [69]THUMMEL K E. In vitro and in vivo drug interactions involving human CYP3A4 [J]. Annu Rev Pharmacol Toxicol,1998,38:389-430.
    [70]CERMAK R, WEIN S, WOLFFRAM S, et al. Effects of the flavonol quercetin on the bioavailability of simvastatin in pigs [J]. Eur J Pharm Sci,2009,38(5):519-524.
    [71]PAULKE A, ECKERT G P, SCHUBERT-ZSILAVECZ M, et al. Isoquercitrin provides better bioavailability than quercetin:comparison of quercetin metabolites in body tissue and brain sections after six days administration of isoquercitrin and quercetin [J]. Pharmazie,2012,67(12):991-996.
    [72]KRIZKOVA J, BURDOVA K, STIBOROVA M, et al. The effects of selected flavonoids on cytochromes P450 in rat liver and small intestine [J]. Interdisciplinary toxicology,2009,2(3):201-204.
    [73]WAGNER F, KALUSCHE D, TRENK D, et al. Drug interaction between propafenone and metoprolol [J]. Br J clin Pharmac,1987,24(2):213-220.
    [74]AHMAD S. Metoprolol-induced delirium perpetuated by propafenone [J]. Am Fam Physician,1991,44(4):1142,1144.
    [75]AMUNUGAMA H T, ZHANG H, HOLLENBERG P F. Mechanism-based inactivation of cytochrome P450 2B6 by methadone through destruction of prosthetic heme [J]. Drug Metab Dispos,2012,40(9):1765-1770.
    [76]LU W J, BIES R, KAMDEN L K, et al. Methadone:a substrate and mechanism-based inhibitor of CYP19 (aromatase) [J]. Drug Metab Dispos,2010,38(8): 1308-1313.
    [77]TASSANEEYAKUL W, GUO L Q, FUKUDA K, et al. Inhibition selectivity of grapefruit juice components on human cytochromes P450 [J]. Arch Biochem Biophys, 2000,378(2):356-363.
    [78]CHUN Y J, RYU S Y, JEONG T C, et al. Mechanism-based inhibition of human cytochrome P450 1A1 by rhapontigenin [J]. Drug Metab Dispos,2001,29(4 Pt 1): 389-393.
    [79]CHAN W K, DELUCCHI A B. Resveratrol, a red wine constituent, is a mechanism-based inactivator of cytochrome P450 3A4 [J]. Life Sci,2000,67(25): 3103-3112.
    [80]SURH Y J, LEE S S. Capsaicin, a double-edged sword:toxicity, metabolism, and chemopreventive potential [J]. Life Sci,1995,56(22):1845-1855.
    [81]OBACH R S, WALSKY R L, VENKATAKRISHNAN K. Mechanism-Based Inactivation of Human Cytochrome P450 Enzymes and the Prediction of Drug-Drug Interactions [J]. Drug Metab Dispos,2006,35(2):246-255.
    [82]DONG W, WANG P, MENG X, et al. Ultra-performance liquid chromatography-high-definition mass spectrometry analysis of constituents in the root of Radix Stemonae and those absorbed in blood after oral administration of the extract of the crude drug [J]. Phytochem Anal,2012,23(6):657-667.
    [83]LIU Y, ZHANG J W, LI W, et al. Ginsenoside metabolites, rather than naturally occurring ginsenosides, lead to inhibition of human cytochrome P450 enzymes [J]. Toxicol Sci,2006,91(2):356-364.
    [84]SHIRASAKA Y, KAWASAKI M, SAKANE T, et al. Induction of human P-glycoprotein in Caco-2 cells:development of a highly sensitive assay system for P-glycoprotein-mediated drug transport [J]. Drug Metab Pharmacokinet,2006,21(5): 414-423.
    [85]阎祖炜,朱欣,李闻文.MTT法、CPE观察法用于药物细胞毒性实验的比较与分析[J].实用预防医学,2007,14(5):1552-1554.
    [86]颜苗.甘草酸18位差向异构体及其水解产物对P-糖蛋白影响的研究[D];中南大学,2010.
    [87]刘宁宁,杨秀萍.生物碱逆转肿瘤多药耐药性的研究进展[J].中国现代中药,2009,11(8):7-10.
    [88]ZHANG S, MORRIS M E. Effects of the flavonoids biochanin A, morin, phloretin, and silymarin on P-glycoprotein-mediated transport [J]. J Pharmacol Exp Ther,2003, 304(3):1258-1267.
    [89]LOHNER K, SCHNABELE K, DANIEL H, et al. Flavonoids alter P-gp expression in intestinal epithelial cells in vitro and in vivo [J]. Molecular nutrition & food research, 2007,51(3):293-300.
    [90]张韶瑜.痹祺胶囊组方配伍对细胞色素P450同工酶活性的影响[D];天津大学,2009.
    [91]郭晓昕,杜晓曦,薛松林.从美国药品撤市看药品安全风险管理[J].中国药物警戒,2011,8(4):212-216.
    [92]张石革,宋菲.回顾若干处方药的淘汰史,重视药物的临床再评价[J].中国医院用药评价与分析,2002,2(04):195-198.
    [93]FDA. Guidance for Industry Drug Interaction Studie—Study Design, Data Analysis, Implications for Dosing, and Labeling Recommendations [S]. http://wwwfdagov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/default htm.2012.
    [94]刘虹,郝或,朱晓月,等.HPLC测定血浆中荷叶碱的浓度及其在大鼠体内的药代动力学研究[J].中国实验方剂学杂志,2010,16(12):165-168.
    [95]何晓曦,孔令提,叶林虎,等.荷叶生物碱在大鼠血和脑中的药代动力学研究;2012年中国药学大会暨第十二届中国药师周[C],中国江苏南京,2012.
    [96]OTTON S V, CREWE H K, LENNARD M S, et al. Use of quinidine inhibition to define the role of the sparteine/debrisoquine cytochrome P450 in metoprolol oxidation by human liver microsomes [J]. J Pharmacol Exp Ther,1988,247(1):242-247.
    [97]REN X, MAO X, SI L, et al. Pharmaceutical excipients inhibit cytochrome P450 activity in cell free systems and after systemic administration [J]. Eur J Pharm Biopharm, 2008,70(1):279-288.
    [98]REN X, MAO X, CAO L, et al. Nonionic surfactants are strong inhibitors of cytochrome P450 3A biotransformation activity in vitro and in vivo [J]. Eur J Pharm Sci, 2009,36(4-5):401-411.
    [99]和凡,钟国平,赵立子,等.丹参酮ⅡA对大鼠细胞色素P450酶的诱导作用[J].中草药,2009,40(6):938-942.
    [100]石苏英,金科涛,王宇光,等.白蔹配伍乌头对大鼠肝脏CYP450的调节作用[J].医药导报,2007,26(9):975-979.
    [101]孙韬.外源物对细胞色素P450同工酶表达的基因调控[J].国外医学药学分册,2001,28(5):279-281.
    [102]王宇光.基于药物代谢酶的中药相互作用研究[D];中国人民解放军军事医学科学院,2006.
    [103]姚新生.中药天然药物活性成分的研究方法[J].药学服务与研究,2003,3(4):205-209.
    [104]闫广利,韩莹,王喜军.基于LC-MS分析的中药体内成分辨识技术[J].中国中药杂志,2012,37(12):1765-1770.
    [105]MULLEN W, BOITIER A, STEWART A J, et al. Flavonoid metabolites in human plasma and urine after the consumption of red onions:analysis by liquid chromatography with photodiode array and full scan tandem mass spectrometric detection [J]. J Chromatogr A,2004,1058(1-2):163-168.
    [106]吴昊,刘斌,王伟,等.LC-MSn鉴定2-羟基-1-甲氧基阿朴啡在Beagle犬体内的主要代谢产物[J].中国药学杂志,2009,44(20):1574-1577.
    [107]LU Y L, HE Y Q, WANG M, et al. Characterization of nuciferine metabolism by P450 enzymes and uridine diphosphate glucuronosyltransferases in liver microsomes from humans and animals [J]. Acta Pharmacol Sin,2010,31(12):1635-1642.
    [108]ZHANG H, ZHANG D, RAY K, et al. Mass defect filter technique and its applications to drug metabolite identification by high-resolution mass spectrometry [J]. J Mass Spectrom,2009,44(7):999-1016.
    [109]LIU D Q, HOP C E. Strategies for characterization of drug metabolites using liquid chromatography-tandem mass spectrometry in conjunction with chemical derivatization and on-line H/D exchange approaches [J]. J Pharm Biomed Anal,2005,37(1):1-18.
    [110]LIN P, QIAN W, WANG X, et al. The biotransformation of oleuropein in rats [J]. Biomed Chromatogr,2013,27(9):1162-1167.
    [111]HAN Y Q, WANG J, CUI Q X, et al. Absorption, metabolism and effect of compatibility on absorption of qishenyiqi dropping pill [J]. Biomed Chromatogr,2013, 28(4):554-563.
    [112]MULLEN W, EDWARDS C A, CROZIER A. Absorption, excretion and metabolite profiling of methyl-, glucuronyl-, glucosyl- and sulpho-conjugates of quercetin in human plasma and urine after ingestion of onions [J]. Brit J Nutr,2007,96(1):107-116.
    [113]LI A C, ALTON D. BRYANT M S, et al. Simultaneously quantifying parent drugs and screening for metabolites in plasma pharmacokinetic samples using selected reaction monitoring information-dependent acquisition on a QTrap instrument [J]. Rapid communications in mass spectrometry:RCM,2005,19(14):1943-1950.
    [114]CHANG Q, ZUO Z, CHOW M S, et al. Difference in absorption of the two structurally similar flavonoid glycosides, hyperoside and isoquercitrin, in rats [J]. Eur J Pharm Biopharm,2005,59(3):549-555.
    [115]HAO W, BIN L, WEI W, et al. Identification of Major Metabolites of 2-Hydroxy-1-Methoxyaporphine in Beagle Dogs Plasma by Liquid Chromatography/Ion Trap Mass Spectrometry [J]. Chin Pharm J,2009,44(20):1574-1577.
    [116]王玉霞,刘斌,石任兵,等RP-HPLC法测定Beagle犬血浆中荷叶碱的浓度及药动学研究[J].药物分析杂志,2008,28(9):1418-1421.
    [117]BARANCZEWSKI P, STANCZAK A, KAUTIAINEN A, et al. Introduction to early in vitro identification of metabolites of new chemical entities in drug discovery and development. [J]. Pharmacol Rep,2006,58(3):314-352.
    [118]MCLENNAN H, WHEAL H V. The specificity of action of three possible antagonists of amino acid-induced neuronal excitations [J]. Neuropharmacology,1976, 15(11):709-712.
    [119]FELIX D, FRANGI U. Dimethoxyaporphine as an antagonist of chemical excitation in the pigeon optic tectum [J]. Neurosci Lett,1977,4(6):347-350.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700