百里酚及异百里酚的体外代谢研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
百里酚和异百里酚是百里香属、薄荷香属等药用植物中广泛分布的单酚类化合物,它们在结构上互为同分异构体。作为百里香属、薄荷香属植物的主要活性成分,百里酚和异百里酚显示了良好的抗氧化、抗炎、抗菌及抗肿瘤等药理学活性,是多味中药产生疗效的物质基础。此外,这两个化合物还作为食品添加剂(FDA和欧盟批准)被广泛应用于食品工业中。借助体外技术体系开展中药活性成分在人体及动物组织的吸收、分布、代谢及排泄(ADME)等性质研究,不仅有助于揭示中药的活性物质基础及其在人体及动物体内的处置进程,进而选择适宜动物模型来开展整体药代、药理及毒理研究;还对科学解释中药体内作用机制以及中草药药物的相互作用具有重大作用。百里酚和异百里酚作为多种药食同源中草药的主要活性成分,其在人体内的代谢转化及其对代谢酶活性的影响对临床安全用药至关重要。然而,到目前为止,对上述两个单酚类化合物的体内代谢途径尚不清楚,其代谢稳定性及代谢行为、以及是否引发药物药物相互作用(DDⅠ)等科学问题至今未得到很好的研究,而这些性质对中药自身以及与其共服药物的有效性和安全性均会产生重大影响。同时,百里酚和异百里酚在多种食物和中草药中大量存在,在人们食用富含百里酚和异百里酚的食物时其与其它食物和药物的相互作用,非常值得我们关注,尤其是对患者在临床用药中的药物膳食结构更应引起我们的注意。
     酚类化合物在中草药中广泛存在且结构多样、而且多具有抗氧化、抗炎、抗菌等活性,其不但为新药研发提供了广阔的资源,同时其代谢产物又进一步丰富了化合物的结构多样性。药物代谢性质的研究,对先导化合物的优化、中药作用机理的发现及药物的安全有效使用均具有重要意义。同时多数酚类化合物均是Ⅱ相代谢酶的天然底物,包括磺酸化酶,甲基化酶,葡萄糖醛酸化酶(UGT)等Ⅱ相代谢酶;但作为人体主要代谢酶的细胞色素氧化酶P450更倾向于代谢含有疏水基的化合物,然而既含有酚羟基又具有疏水基的物质其在人体内代谢行为,及在特定化合物中CYP与UGT代谢的重要性至今鲜有报道。本研究利用人肝及动物肝微粒体等体外孵育体系,对百里酚和异百里酚的体外代谢稳定性、代谢酶动力学、种属差异、药物相互作用以及化合物结构对药物代谢行为的影响进行了系统研究。
     本论文首先建立了百里酚和异百里酚的高效液相检测方法;所建立的方法具有很好的特异性;精密度和准确度及稳定性等研究均符合方法学的要求;经过方法学验证,该方法可以满足百里酚和异百里酚两种化合物在后续研究中的定量要求。在此基础上,论文着重对百里酚和异百里酚进行了体外Ⅰ、Ⅱ相酶代谢研究,阐明了两化合物的代谢途径;同时考察了两化合物代谢的种属差异及药物相互作用的效应。
     本论文系统研究了百里酚和异百里酚体外Ⅰ相代谢,阐明了两化合物在人肝中的Ⅰ相代谢途径。在人肝微粒体孵育体系中,百里酚在能生成一个羟化代谢产物(M 1),异百里酚能够生成两个羟化代谢产物(M 1,M 2)。进一步研究证实,CYP1A2、CYP2A6和CYP2B6参与了百里酚的Ⅰ相代谢,CYP1A2、CYP2A6参与了异百里酚Ⅰ相代谢M 1的生成,CYP1A2、CYP2A6和CYP2B6则参与介导了M 2的生成。在不同种属间的代谢能力表现出较大的差异,依据研究的不同目的,可以选择不同的实验动物作为模型。在研究原形的毒性时,可能要考虑清除比较慢的动物。在研究代谢产物的毒性时,可能要考虑代谢转化较快的动物。各种属对百里酚和异百里酚CYP代谢能力的差异,也提示在未来工作中,深入研究代谢物的活性及毒性是非常有必要的。
     本论文同时研究了百里酚和异百里酚体外Ⅱ相代谢,阐明了两化合物在人肝和人肠中的Ⅱ相代谢途径。在人肝和人肠微粒体孵育体系中,百里酚和异百里酚都能生成一个单葡萄糖醛酸结合物。进一步研究证实,两化合物的葡萄糖醛酸结合代谢都是多酶参与的过程,其中UGT1A3、UGT1A7、UGT1A8、UGT1A9和UGT2B7能催化百里酚的葡萄糖醛酸化代谢,UGT1A3,UGT1A6,UGT1A7,UGT1A9和UGT2B7能催化异百里酚的葡萄糖醛酸化代谢。通过抑制剂研究发现,UGT1A9在人肝脏代谢中起主要作用,UGT1A7在人肠代谢中起主要代谢作用。两个化合物在不同种属间的葡萄糖醛酸化代谢酶谱尽管没有区别,但是代谢活性还是存在着较大的差异,需要在后续研究中依照不同的研究目的选择合适的动物代谢模型。
     通过比较百里酚和异百里酚的Ⅰ、Ⅱ相代谢发现,异百里酚无论在Ⅰ相还是Ⅱ相酶代谢过程中,均表现出了比百里酚较高的代谢活性和相对高的代谢清除率。尤其是在Ⅱ相葡萄糖醛酸化代谢中表现更为明显。同时发现,在百里酚的Ⅰ相、Ⅱ相代谢比较中,CYP代谢路径的代谢清除率占有更大优势,可能是其主要的代谢途径。在异百里酚的代谢中,UGT代谢所起的作用更加重要,暗示疏水集团和酚羟基的空间距离在决定药物代谢途径和代谢快慢上具有重要作用。
     本论文还考察了百里酚和异百里酚引发DDⅠ的可能性,研究发现异百里酚能够抑制UGT1A9的活性。该抑制表现出了明显依赖于酶或底物的特点。通过异百里酚对UGT特异性探针底物4 MU和异丙酚抑制作用的能力来评价其导致DDⅠ的可能性。结果发现,异百里酚对4 MU在UGT1A9代谢中产生了竞争性的抑制作用(Ki=5.7 M),而在对异丙酚在UGT1A9单酶代谢中产生了非竞争性的抑制作用(Ki=25.0 M)。所以在临床或者食用异百里酚的过程中需要注意DDI的可能性作用。
     比较两种化合物的结构并结合国内外已有文献,发现疏水基远离酚羟基的化合物更趋向于发生Ⅱ相化代谢;同时其代谢酶谱也产生一定的差异,代谢能力也发生很大的变化;这可能是由于疏水基空间位阻的作用产生了上述的影响。以上发现提示小分子化合物结构上的微小变化,都可能会导致代谢途径的巨大差异。在模式实验动物选择中应选择与人具有相似代谢行为和代谢能力的动物来开展整体动物实验,若利用了不合适的动物模型进行临床前研究可能会产生较大差异、从而影响临床实验的设计及数据的可靠性。
     本研究对百里酚和异百里酚的体外代谢系统研究,首次报道揭示了它们的代谢通路,有助于对该类化合物在药物早期研究中的‘成药性’进行评估。同时丰富了单酚类化合物的药代动力学研究,对此类化合物的后续研究提供了理论依据和实验支持。也对临床药物膳食结构具有一定的指导意义。
Thymol and carvacrol are isomers in structure, which are predominant monoterpenic phenol widely distributed in thyme and oregano. Thymol and carvacrol are the major component of thyme and oregano essential oils, which have many diverse pharmacology activities such as antioxidant, anti inflammatory, antibacterial and anti tumor. In addition, the two compounds are also widely used as a food additive in the food industry. The study of absorption, distribution, metabolism and excretion (ADME) properties in the human body and animals tissues of the main active ingredients of Chinese medicine, which carry out in vitro technology system, not only helps to reveal the material basis of the activity of traditional Chinese medicine and its disposal of human and animal body process, then select the appropriate animal model to carry out the overall pharmacokinetics, pharmacology and toxicology; also have a significant role to explain the mechanism of traditional Chinese medicine in the human body and Chinese herbal medicine drug interactions. Thymol and carvacrol are the main active ingredients of Chinese herbal medicine homology as many different variety of food and medicine; and which the contribution of its metabolic transformation in the human body and its effects on the metabolic enyme activity is essential for the clinical safety of drugs. However, so far, metabolic pathway of the two monoterpene in vivo is not clear, the metabolic stability and metabolic behavior, and whether the cause of drug drug interactions (DDI) and other scientific issues has not been well study; and these properties for the safety and effectiveness of the Chinese medicine itself and co administer drug will have a significant impact.
     Phenolics compounds is widespread in Chinese herbal medicine and structurally diverse, and most have antioxidant, anti inflammatory, antibacterial activity; which not only furnish the extensive resources for the new drug development, and its metabolites further enrich the structure of various compounds. The study is to investigate the metabolic stability in vitro, enzyme pharmacokinetics, species differences, drug drug interaction and structure pharmacokinetics of thymol and carvacrol, which use human liver microsomes, animal liver microsomes and other in vitro incubation systems
     The different methods of high performance liquid detection for thymol and carvacrol were established at first. The method has good specificity, which the precision & accuracy and stability of it are in line with methodology requirements; and the method can meet the quantitative requirements for the follow up study. On this basis, the paper focuses on in vitro phase I andⅡmetabolism of thymol and carvacrol, and the metabolic pathways of the two compounds have been elucidation.
     In vitro phase I of thymol and carvacrol were investigated in this study, and the phase I pathway of the two compounds in HLMs were illustrated. There were one hydroxylated metabolite (M 1) found in the metabolic reaction of thymol and two hydroxylated metabolites (M 1, M 2) generated in the metabolic reaction of carvacrol. Throughout the investigation of recombine CYP and inhibitory effects, we found CYP1A2, CYP2A6 and CYP2B6 were involved in the metabolism of thymol in the hydroxylation reaction; CYP1A2, CYP2A6 involved in the formation of M 1, and CYP1A2, CYP2A6 and CYP2B6 were involved in the formation of M 2. The diversity of the ability of metabolism was shown in different species, so we can select animal model according as the object of the study. For the toxicity study of former drug, the animal which have the comparatively slow clear rate was the better choice; but for the metabolite, the quicker the better. The diversity of the CYP in the different species prompt us the study of pharmacodynamics and toxicity of metabolite were very essential
     Meanwhile, the investigation of the in vitro phaseⅡof thymol and carvarol were carried out, the phaseⅡpathyway of it in HLMs and HIMs were clarified. There were one glucuronidation metabolite found in the HLMs and HIMs. The study also validated there were more enzymes involoved the reaction. Chmical inhibition studies and related screening of recombinant human enzyme were experimented, and found UGT1A3, UGT1A7, UGT1A8, UGT1A9, and UGT2B7 involoved in the thymol glucuronidation metabolic response, UGT1A3, UGT1A6, UGT1A7, UGT1A9 and UGT2B7 in carvacrol. Furthermore, we indentified UGT1A9 as the major isozyme responsible for carvacrol glucuronidation in HLMs; and the UGT1A7 as the major isozyme responsible in HIMs. In dispite of there is not much different in zymogram between the different spices, the ability of metabolism have shown the huge diversity. So, the selection of the animal model should be in line with the orders.
     The kinetic paraments showed that carvacrol have own the relatively high metabolic activities and clearance rate than thymol in terms of phase I or II metabolic reactions, especially in the glucuronidation of carvacrol. Meanwhile, the higher clearance rate of thymol hydroxylation in the phase I and II shown that the CYP might be the main metabolic pathway of thymol in human; and the result of the carvacrol in the phase I and II indicate the CYP and UGT metabolism might have the identical contribution in human body. The results shown that the interspace between the hydrophobic group and hydroxyl group have the significant effects in determining the pathyway and speed of metabolism.
     The study also extremely investigated the inhibitory effects of carvacrol and thymol on major CYP and UGT isoformes, and the carvacrol have the ability of inhibition on UGT1A9 isoformes. The probe substrate for all tested UGT isoforms is 4 MU which is a nonselective substrate of UGTs, and the Propofol, mainly metabolized by UGT1A9, could be used as a probe substrate for UGT1A9. The results showed that carvacrol could inhibit the activity of UGT1A9 with negligible effects on other UGT isoforms. The carvacrol competitively inhibit UGT1A9 mediated 4 MU glucuronidation reaction (Ki=5.7 M), but the noncompetitively inhibiton found in carvacrol towards UGT1A9 mediated propofol glucuronidation(Ki=25.0 M). To date, mechanism of substrate dependent inhibition of drug metabolizing enzymes was still unclear. Therefore, in vitro data on the inhibition of UGT1A9 by carvacrol should be interpreted with caution.
     We found the compounds that the hydrophobic group far from the phenolic hydroxyl group have the tendency of occurrence of phase II, through compare the structure between the two compounds and analysis the literature at home and abroad. Meanwhile, there were some different on the metabolic enzyme and metabolic capability, which may be due to the stereospecific blockade effects of hydrophobic group. These findings suggest that small changes in the structure of small molecule compounds, may lead to a huge difference in the metabolic pathways. It should be selected with the people with similar metabolic behavior and metabolic capability to carry out the whole animal experiments, if the inappropriate animal models were be use, the pre clinical study may generate vary significantly, thus affecting the design of clinical trials and data reliability.
     In this study, the in vitro metabolism of thymol and carvacrol were carried out, and the metabolism pathway of the two compounds were first clarified, and the consequence is very useful to asses the‘medicine’for the candidate compounds in the early development. However, the result enriches the pharmacokinetic study of monoterpenic phenol, and provide a theoretical basis and experimental support for such compounds in the follow up study.
引文
[1] Wienkes LC, Health TG. Predicting in vivo drug interactions from in vitro drug discovery data. Nature Reviews Drug Discovery. 2005. 4:825 833.
    [2] Josephy PD, Guengerich FP, Miners JO, et.al.“Phase I and Phase II”Drug Metabolism: Terminology that we should Phase out? Drug Metabolism Reviews. 2005. 37(4): 575 580.
    [3] Yengi LG, Leung L, Kao J. The evolving role of drug metabolism in drug discovery and development. Pharmcology Research. 2007. 24(5): 842 858.
    [4] Lynch T, Price A. the effect of cytochrome P450 metabolism on drug response, interactions, and adverse effects. American Family Physician. 2007. 76(3): 391 396.
    [5] Mackenzie PI, Bock KW, Burchell B, Guillemette C, Ikushiro S, Iyanagi T, Miners JO, Owens IS, Nebert DW. Nomenclature update for the mammalian UDP glycosyltransferase (UGT) gene superfamily. Pharmacogenet Genomics. 2005. 15(10): 677 685.
    [6] Williams JA, Hyland R, Jones BC, Smith DA, Hurst S, Goosen TC, Peterkin V, Koup JR, Ball SE. Drug drug interactions for UDP glucuronosyltransferase substrates: a pharmacokinetic explanation for typically observed low exposure (AUCi/AUC) ratios. Drug Metabolism and Disposition. 2004. 32(11):1201 1208.
    [7] Testa B, Kramer SD, The biochemistry of drug metabolism an introduction: part 1. Principles and overview. Chemistry & Biodiversity. 2006.3(10):1053 1101.
    [8] Smith DA, Waterbeemd H. Pharmacokinetics and metabolism in early drug discovery. Current Opinion in Chemical Biology. 1999. 3(4):373 378.
    [9] Thompson TN. Optimization of Metabolic Stability as a Goal of Modern Drug Design. Medicinal Research Reviews. 2001. 21(5): 412 449.
    [10] Ariens EJ, Simonis AM. Optimization of Pharmacokinetics, an Essential Aspect of Drug Development, by‘Metabolic Stabilization’. In Strategy in Drug Research (Pharmacochemistry Library), 1982; pp.165–178, Elsevier.
    [11] Walsky RL, Obach RS. Validated assays for human cytochrome P450 activities. Drug Metabolism and Disposition. 2004. 32(6): 647 660.
    [12] Kiang TKL, Ensom MHH, Chang TKH. UDP glucurosyltransferases and clinical drug drug interactions. Pharmacology & Terapeutics. 2005. 106: 97 132.
    [13] Abu La? S, Odeh I, Dewik H, et al. Thymol and carvacrol production from leaves of wild Palestinian Majorana syriaca. Bioresource Technology. 2008. 99: 3914–3918.
    [14] Edward E, Langenau. The determination of thymol and carvacrol in Spanish thyme oils. Journal of the Science of Food and Agriculture. 1949. 38:261–266.
    [15] Cimanga K, Apers S, de Bruyne T, et al. Chemical composition and antifungal activity of essential oils of some aromatic medicinal plants growing in the Democratic Republic of Congo. Journal of Essential Oil Research. 2002. 14(5): 382 387.
    [16] Baser KHC. Biological and Pharmacological Activities of Carvacrol and Carvacrol Bearing Essential Oils. Current Pharmaceutical Design. 2008. 29: 3106 3119.
    [17] Azirak S, Rencuzogullari E. The in vivo genotoxic effects of carvacrol and thymol in rat bone marrow cells. Environment Toxicology. 2008. 23: 728–735.
    [18] Passreiter CM, Wilson J, Andersen R, et al. Metabolism of Thymol and trans Anethole in Larvae of Spodoptera litura and Trichoplusia ni (Lepidoptera: Noctuidae). Journal of Agriculture and Food Chemistry. 2004. 52: 2549 2551.
    [19] Shimoda K, Kondo Y, Nishida T, et al. Biotransformation of thymol, carvacrol, and eugenol by cultured cells of Eucalyptus perriniana. Phytochemistry. 2006. 67: 2256–2261.
    [20] Austgulen LT, Solheim E, Scheline RR. Metabolism in rats of p cymene derivatives: carvacrol and thymol. Pharmacology & Toxicology. 1987. 61: 98 102.
    [21] Kohlert C, Schindler G, M?rz RW, et al. Systemic Availability and Pharmacokinetics of Thymol in Humans. Journal of Clinical Pharmacology. 2002. 42: 731 737.
    [22] SENAFI SB, CLARKE DJ, Burchell B. Investigation of the substrate specificity of a cloned expressed human bilirubin UDP glucuronosyltransferase: UDP sugar specificity and involvement in steroid and xenobiotic glucuronidation. Biochemcal Journal.1994. 303: 233 240.
    [23] Jedlitschky G, Cassidy AJ, Sales M, et al. Cloning and characterization of a novel human olfactory UDP glucuronosyltransferase. Biochemcal Journal. 1999. 340: 837–843.
    [24] Turgeon D, Carrier J S, Chouinard S, et al. Glucuronidation activity of the UGT2B17 enzyme toward xenobiotics. Drug Metabolism and Disposition. 2003.31: 670 676.
    [25] Basu NK, Ciotti M, Hwang MS, et al. Differential and Special Properties of the Major Human UGT1 encoded Gastrointestinal UDP glucuronosyltransferases Enhance Potential to Control Chemical Uptake. The Journal of Biological Chemistry. 2004. 279: 1429–1441.
    [26] Smith PA, Sorich MJ, McKinnon RA, et al. In silico Insights: chmical and structural characteristics associated with uridine diphosphate glucuronosyltransferase substrate selectivity. Clinical and Experimental Pharmacology and Physiology. 2003. 30: 836–840.
    [27] Smith PA, Sorich MJ, McKinnon RA, et al. Pharmacophore and Quantitative Structure Activity Relationship Modeling: Complementary Approaches for the Rationalization and Prediction of UDP Glucuronosyltransferase 1A4 Substrate Selectivity. Journal of Medicinal Chemistry. 2003. 46: 1617 1626.
    [28] Jukic M, Politeo O, Maksimovic M, et al. In Vitro Acetylcholinesterase Inhibitory Properties of Thymol, Carvacrol and their Derivatives Thymoquinone and Thymohydroquinone. Phytotherapy Research. 2007. 21: 259 261.
    [1] Wienk?tter N, Begrow F, Kinzinger U, et al. The Effect of Thyme Extract onβ2 Receptors and Mucociliary Clearance. Planta Medica. 2007. 73 (7): 629–635.
    [2] Priestley, Caroline M, Williamson E, et al. Thymol, a constituent of thyme essential oil, is a positive allosteric modulator of human GABAA receptors and a homo oligomeric GABA receptor from Drosophila melanogaster. British Journal of Pharmacology. 2003. 140: 1363–1372.
    [3] Xu J, Zhou F, Ji BP, et al. The antibacterial mechanism of carvacrol and thymol against Escherichia coli. Letters in Applied Microbiology. 2008, 47 (3): 174–179.
    [4] Rivas L, McDonnell MJ, Burgess CM, et al. Inhibition of verocytotoxigenic Escherichia coli in model broth and rumen systems by carvacrol and thymol". International Journal of Food Microbiology. 2010. 139 (1 2): 70–78.
    [5] Ettayebi K, El Yamani J, Rossi Hassani B. Synergistic effects of nisin and thymol on antimicrobial activities in Listeria monocytogenes and Bacillus subtilis. FEMS Microbiology Letters.2006. 183(1): 191–195.
    [6] Nebert DW, Dalton TP. The role of cytochrome P450 enzymes in endogenous signalling pathways and environmental carcinogenesis. Nature. 2006. 6: 947 960.
    [7] Bachmann KA, Ghosh R. The use of in vitro methods to predict in vivo pharmacokinetics and drug interactions. Current Drug Metabolism. 2001. 2: 299–314.
    [8] Howgate EM, Rowland Yeo K, Proctor NJ, et al. Prediction of in vivo drug clearance from in vitro data. I: impact of inter individual variability. Xenobiotica. 2006. 36: 473–97.
    [9] Obach RS, Walsky RL, Venkatakrishnan K, et al. The utility of in vitro cytochrome P450 inhibition data in the prediction of drug drug interactions. Journal of Pharmacology and Experimental Therapeutics. 2006. 316: 336–48.
    [10] Sanderink G, Bournique B, Stevens J, et al. Involvement of human CYP1A isoenzymes in the metabolism and drug interactions of riluzole in vitro. Journal of Pharmacology and Experimental Therapeutics. 1997. 282: 1465–72.
    [11] Lowry OH, Rosebrough NJ, Farr AL, et al. Protein measurement with the Folin phenol reagent. Journal of Biological Chemistry. 1951. 193: 265–75.
    [12] Bjornsson Td, Callaghan Jt, Einolf Hj, et al. The conduct of in vitro and in vivo drug drug interaction studies: a phrma perspective. Drug Metabolism and Disposition. 2003. 31: 815–32.
    [13] Huang SM, Temple R, Throckmorton DC,et al. Drug interaction studies: study design, data analysis, and implications for dosing and labeling. Clinical Pharmacology Therapeutic. 2007. 81: 298–304.
    [14] Harris JW, Rahman A, Kim BR, et al. Metabolism of taxol by human hepatic microsomes and liver slices: participation of cytochrome P450 3A4 and an unknown P450 enzyme. Cancer Research.1994. 54: 4026–4035.
    [1] Stammati A, Bonsi P, Zucco F, et al. Toxicity of selected plant volatiles in microbial and mammalian short term assays. Food and Chemical Toxicology. 1999. 37: 813 823.
    [2] Fabian D, Sabol M, Domaracka K,et al. Essential oils their antimicrobial activity against Escherichia coli and effect on intestinal cell viability. Toxicology in Vitro. 2006. 20: 1435 1445.
    [3] Austgulen LT, Solhein E, Scheline RR. Metabollism in rats of para cymene derivatives carvacrol and thymol. Pharmacology & Toxicology. 1987. 61(2): 98 102.
    [4] Narimatsu, S; Yonemoto, R; Masuda, K, et al.Oxidation of 5 methoxy N, N diisopropyltryptamine in rat liver microsomes and recombinant cytochrome P450 enzymes. Biochemical Pharmacology.. 2008. 75(3): 752 760.
    [5] Sauer C, Peters FT, Schwaninger AE, et al. Identification of Cytochrome P450 Enzymes Involved in the Metabolism of the Designer Drugs N (1 Phenylcycloh exyl) 3 ethoxypropanamine and N (1 Phenylcyclohexyl) 3 methoxypropanamine. Chemical Research Toxicology. 2008. 21(10): 1949 1955.
    [6] Nebert DW, Russell DW. Clinical importance of the cytochromes P450. Lancet 2002. 360(9340): 1155 1162.
    [7] Lin JH, Lu AYH. Interindividual variability in inhibition and induction of cytochrome P450 enzymes. Annual Revieew of Pharmacology and Toxicology. 2001 .41: 535–567.
    [8] Lewis DF, Ioannides C, Parke DV, et al. Quantitative structure–activity relationships in a series of endogenous and synthetic steroids exhibiting induction of CYP3A activity and hepatomegaly associated with increased DNA synthesis. The Journal of Steroid Biochemistry and Molecular Biology. 2000. 74(4): 179–185.
    [9] Evans WE, McLeod HL. Pharmacogenomics drug disposition, drug targets, and side effects. The New England Journal of Medicine. 2003. 348(6): 538–549.
    [1] Radominska Pandya A, Czernik PJ, Little JA, et al. Structural and functional studies of UDP glucuronosyltransferases. Drug Metabolism and Reviewes. 1999. 31: 817–899.
    [2] Tukey RH, Strassburg CP. Human UDP glucuronosyltransferases: metabolism, expression, and disease. Annual Review Pharmacology and Toxicology. 2000. 40: 581–616.
    [3] Ritter JK. Roles of glucuronidation and UDP glucuronosyltransferases in xenobiotic bioactivation reactions. Chem Biol Interact. 2000. 129: 171–193.
    [4] Soars MG, Burchell B, Riley RJ. In vitro analysis of human drug glucuronidation and prediction of in vivo metabolic clearance. Journal of Pharmacology and Experimental Therapeutics. 2002. 301: 382–390.
    [5] Williams JA, Hyland R, Jones BC, et al. Drug–drug interactions for UDP glucuronosyltransferase substrates: a pharmacokinetic explanation fortypically observed low exposure (AUCi/AUC) ratios. Drug Metabolism and Disposition.. 2004. 32(11): 1201–1208.
    [6] Uchaipichat V, Mackenzie PI, Elliot DJ, et al. Selectivity of substrate (trifuoperazine) and inhibitor (amitriptyline, androsterone, canrenoic acid, hecogenin, phenylbutazone, quinidine, quinine, and sulfnpyrazone)“probes”for human UDP glucuronosyltransferases. Drug Metabolism and Disposition. 2006. 34: 449–456.
    [7] Zhang YY, Liu Y, Zhang JW, et al. Characterization of human cytochrome P450 isoforms involved in the metabolism of 7 epi paclitaxel. Xenobiotica. 2009 39: 283–292.
    [8] Liu Y, Ramirez J, House L, et al. Comparison of the drug drug interactions potential of erlotinib and gefitinib via inhibition of UDP glucuronosyltransferases. Drug Metabolism and Disposition. 2010. 38: 32 39.
    [9] Huang T, Fang ZZ, Yang L. Strong inhibitory effect of medroxyprogesterone acetate (MPA) on UDP glucurosyltransferase (UGT) 2B7 might induce drug drug interactions. Pharmazie. 2010. 65: 919 921.
    [10] Austgulen LT, Solheim E, Scheline RR. Metabolism in rats of p cymene derivatives: carvacrol and thymol. Pharmacology & Toxicology. 1987. 61: 98 102.
    [11] Kohlert C, Schindler G, M?rz RW, et al. Systemic Availability and Pharmacokinetics of Thymol in Humans. Journal of Clinical Pharmacology. 2002. 42: 731 737.
    [1] King CD, Rios GR, Green MD, et al. UDP glucuronosyltransferases. Current Drug Metabolism. 2000. 1: 143–161.
    [2] Mackenzie PI, Owens IS, Burchell B, et al. The UDP glycosyltransferase gene superfamily: Recommended nomenclature update based on evolutionary divergence. Pharmacogenetics. 1997. 7(4): 255–269.
    [3] Mackenzie PI, Bock KW, Burchell B, et al. Nomenclature update for the mammalian UDP glycosyltransferase (UGT) gene superfamily. Pharmacogenet Genomics. 2005. 15: 677–685.
    [4] Court MH. Isoform selective probe substrates for in vitro studies of human UDP glucuronosyltransferases. Methods Enzymol. 2005. 400:104–116.
    [5] Miners JO, Knights KM, Houston JB, et al. In vitro in vivo correlation for drugsand other compounds eliminated by glucuronidation in humans: pitfalls and promises. Biochemcal Pharmacology. 2006. 71: 1531–1539.
    [6] Nakamura A, Nakajima M, Yamanaka H, et al. Expression of UGT1A and UGT2B mRNA in human normal tissues and various cell lines. Drug Metabolism and Disposition. 2008. 8: 1461 1464.
    [7] Kirimer N., Baser KHC, Tumen G. Carvacrol rich plants in Turkey. Khimiya Prirodnykh Soedinenii. 1995. 31: 49–54.
    [8] Baser KHC. Biological and Pharmacological Activities of Carvacrol and Carvacrol Bearing Essential Oils. Current Pharmaceutical Design. 2008. 29: 3106 3119.
    [9] Jamroz D, Wertecki T, Houszka M, et al. In?uence of diet type on the inclusion of plant origin active substances on morphological and histochemical characteristics of the stomach and jejunum walls in chicken. Journal of Animal Physiology and Animal Nutrition. 2006. 90(5 6): 255–268.
    [10] Lagouri V, Bleskas G, Tsimidou M, et al. Composition and antioxidant activity of essential oils from Oregano plants grown wild in Greece. Zeitschrift Fur Lebensmitteluntersuchung Und Forschung A. 1993. 197: 20–23.
    [11] Antonio L, Grillasca JP, Taskinen J, et al. Characterization of catechol glucuronidation in rat liver. Drug Metabolism and Disposition. 2002. 30: 199–207.
    [12] Miners JO, Mackenzie PI, Knights KM. The prediction of drug glucuronidation parameters in humans: UDP glucuronosyltransferase enzyme selective substrate and inhibitor probes for reaction phenotyping and in vitro in vivo extrapolation of drug clearance and drug drug interaction potential. Drug Metabolism Reviews. 2010. 42: 196 208.
    [13] Liang SC, Ge GB, Liu HX, et al. Identification and Characterization of Human UDP Glucuronosyltransferases responsible for the in vitro Glucuronidation of Daphnetin. Drug Metabolism and Disposition. 2010. 38(6): 973 980.
    [14] Krishnaswamy S, Duan SX, von Moltke LL, et al. Validation of serotonin (5 hydroxytryptamine) as an in vitro substrate probe for human UDP glucuronos yltransferase(UGT)1A6. Drug Metabolism and Disposition. 2003. 31: 133–139.
    [15] Krishnaswamy S, Hao Q, von Moltke LL, et al. Evaluation of 5 hydroxytrypto phol and other endogenous serotonin (5 hydroxytryptamine) analogs as substrates for UDP glucuronosyltransferase 1A6. Drug Metablolism and Disposition. 2004. 32(8): 862 869.
    [16] Court MH. Interindividual variability in hepatic drug glucuronidation: studiesinto the role of age, sex, enzyme inducers, and genetic polymorphism using the human liver bank as a model system. Drug Metabolism Reviews. 2010. 42: 209 224.
    [17] Ockenga J, Vogel A, Teich N, et al. UDP glucuronosyltransferase (UGT1A7) gene polymorphisms increase the risk of chronic pancreatitis and pancreatic cancer. Gastroenterology. 2003. 124: 1802 1808.
    [1] Lin JH, Lu AYH. Role of pharmacokinetics and metabolism in drug discovery and didevelopment. Pharmacological Reviews.1997. 49 (4): 403 449.
    [2] Michalets EL. Update: clinically significant cytochrome P450 drug interactions. Pharmacotherapy 1998.18: 84 112.
    [3] Tanaka E. Clinically important pharmacokinetic drug drug interactions: role of cytochrome P450 enzymes. Journal of Clinical Pharmacy and Therapeutics. 1998. 23 (6): 403 416.
    [4] Fang ZZ, Zhang YY, Ge GB, et al. Time dependent inhibition of CYP3A4 and CYP2C9 by noscapine potentially explains clinical noscapine warfarin interaction. British Journal of Clinical Pharmacology. 2010. 69 (2): 193 199.
    [5] Evans WE, Relling MV. Pharmacogenomics: translating functional genomics into rational therapeutics. Science.1999. 286: 487 491.
    [6] Ritter JK. Roles of glucuronidation and UDP glucuronosyltransferases in xenobiotic bioactivation reactions. Chemico Biological Interactions. 2000. 129: 171 193.
    [7] Strassburg CP, Kneip S, Topp J, Obermayer Straub P, et al. Polymorphic gene regulation and interindividual variation of UDP glucuronosyltransferase activity in human small intestine. Journal of Biological Chemistry. 2000. 275: 36164–36171.
    [8] Liang SC, Ge GB, Liu HX, et al. Determination of propofol UDP glucuronos yltransferase (UGT) activities in hepatic microsomes from different species by UFLC ESI MS. Journal of Pharmqceutical and Biomedical Analysis. 2011. 54(1): 236 241.
    [9] Bjornsson TD, Callaghan JT, Einolf HJ, et al. The conduct of in vitro and in vivo drug drug interaction studies: a pharmaceutical research and manufacturers of America (PhRMA) perspective. Drug Metabolism and Disposition. 2003. 31:815 832.
    [10] Bresalier RS, Sandler RS, Quan H, et al. Cardiovascular events associated with rofecoxib in a colorectal adenoma chemoprevention trial. New England Journal of Medicine. 2005. 352: 1092–1102.
    [11] Pratico, D; Dogne, JM. Selective cyclooxygenase 2 inhibitors development in cardiovascular medicine. Circulation. 2005. 112(7): 1073 1079.
    [12] Kiang TKL, Ensom MHH, Chang TKH. UDP glucurosyltransferases and clinical drug drug interactions. Pharmacology & Therapeutics. 2005. 106: 97 132.
    [13] Radominska Pandya A, Bratton S, Little JM. A historical overview of the heterologous expression of mammalian UDP glucuronosyltransferase isoforms over the past twenty years. Current Drug Metabolism. 2005. 6: 141 160.
    [14] Hao M, Zhao Y, Chen P, et al. Structure activity relationship and substrate dependent phenomena in effects of ginsenosides on activities of drug metabolizing P450 enzymes. Plos One.2008. 3: 1 10.
    [15] Stresser DM, Blanchard AP, Turner SD, et al. Substrate dependent modulation of CYP3A4 catalytic activity: analysis of 27 test compounds with four fluorometric substrates. Drug Metabolism and Disposition. 2000. 28: 1440 1448.
    [1] Borges, F; Roleira, F; Milhazes, N, et al. Simple coumarins and analogues in medicinal chemistry: Occurrence, synthesis and biological activity. Current Medicinal Chemistry. 2005. 12(8): 887 916.
    [2] Sardari, S; Mori, Y; Horita, K, et al. Synthesis and antifungal activity of coumarins and angular furanocoumarins. Biddrganic & Medicinal Chemistry. 1999. 7(9): 1933 1940.
    [3] Yang, EB; Zhao, YN; Zhang, K, et al. Daphnetin, one of coumarin derivatives, is a protein kinase inhibitor. Biochemical and Biophysical Research Commmunication. 1999. 260(3): 682 685.
    [1] King A, Young G. Characteristics and occurrence of phenolic phytochemicals. Journal of the American Dietetic Association. 1999. 99:213 218.
    [2] Boudet AM. Evolution and current status of research in phenolic compounds. Phytochemistry, 2007. 68:2722 2735.
    [3] Rice Evans CA, Miller NJ, Paganga G.. Antioxidant properties of phenolic compounds. Trends In Plant Science. 1997. 2:152–159.
    [4] Liang SC, Ge GB, Liu HX, et al. Indentification and characterization of human UDP glucuronosyltransferases responsible for the in vitro glucuronidation of daphnetin. Drug Metab Disp, 2010. 38:973–80,
    [5] Bang KH, Kim. YK, Min. BS, et al. Antifungal activity of magnolol and honokiol. Archives of Pharmacal Research. 2000. 23(1): 46 49
    [6] Summers CB, Felton GW. Prooxidant effects of phenolic acids on the generalist herbivore Helicoverpa zea(Lepidoptera: Noctuidae): potential mode of action for phenolic compounds in plant anti herbivore chemistry. Insect Biochemistry and Molecular Biology. 1994. 24(9): 943–953.
    [7] Yamanaka N, Oda O, Nagao S. Prooxidant activity of caffeic acid, dietary non ?avonoid phenolic acid, on Cu2+ induced low density lipoprotein oxidation. FEBS Lett. 1997. 405:186–190.
    [8] Sugihara N, Arakawa T, Ohnishi M, et al. Anti and pro oxidative effects of ?avonoids on metal in duced lipid hydroperoxide dependent lipid peroxidation in cultured hepatocytes loaded with alpha linolenic acid. Free Radical Biology Medicine. 1999. 27:1313–1323.
    [9] Decker EA. Phenolics: prooxidants or antioxidants? Nutrition Reviews. 1997. 55(11): 396–407.
    [10] Sakihama Y, Cohen MF, Grace SC, et al. Plant phenolic antioxidant and prooxidant activities: phenolics induced oxidative damage mediated by metals in plants. Toxicology. 2002. 177(1): 67–80.
    [11] Chen L, Lee MJ, Li H, et al. Absorption, distribution and elimination of tea polyphenols in rats. Drug Metabolism and Disposition. 1997. 25(9): 1045 1050.
    [12] Yang CS, Chen LS, Lee MJ, et al. Blood and urine levels of tea catechins after ingestion of different amounts of green tea by human volunteers. Cancer Epidemiology Biomarkers & Prevention. 1998. 79(4):351 354.
    [13] Van Amelavoort JM, Van Hof KH, Mathot JN, et al. Plasma concentration of individual tea catechins after a single oral dose in humans. Xenobiotica, 2001. 31(12): 891 901.
    [14] Yang CS, Lee MJ, Chen LS, et al. Human salivary tea catechin levels and catechin esterase activities: impliucation in human cancer prevention studies. Cancer Epidemiology Biomarkers & Prevention. 1999. 8(1):83 89.
    [15] Ogata N, Matsushima N, Shibata T. Pharmacokinetics of Wood Creosote: Glucuronic Acid and Sulfate Conjugation of Phenolic Compounds. Pharmacology. 1995. 51:195 204.
    [16] Wu X, Cao G, Prior RL. Absorption and metabolism of anthocyanins in elderly women after consumption of elderberry or blue berry. Journal of Nutrition. 2002. 132(7): 18652 1871.
    [17] Felgines C, Talavera S, Gonthier MP, et al. Strawberry anthocyanins are recovered in urine as glucuro and sulfo conjugates in humans. Journal of Nutrition. 2003, 133(5): 12962 13011.
    [18] Regev Shoshani G, Shoseyov O, Bilkis I, et al. Glycosylation of resveratrol protects it from enzymic oxidati on. Biochemical Journal. 2003, 374(Pt 1): 1572 1631.
    [19] Morand C, Manach C, Crespy V, et al. Quercetin 3 Oβglucoside is better absorbed than other quercetin form and is not present in rat plasma. Free Radic Research. 2000. 33(5): 6672 6761.
    [20] Halliwell B, Zhao K, Whiteman M. The gastrointestinal tract: a major site of antioxidant action. Free Radic Research. 2000. 33(6): 8192 8301.
    [21] Henning SM, Aronson W, Niu Y, et al. Tea polyphenols and theaflavins are present in prostate tissue of humans and mice after green and black tea consumption. Journal of Nutrition. 2006. 136(7): 18392 18431.
    [22] Garcea G, Berry DP, Jones DJ, et al. Consumption of the putative chemopreventive agent curcumin by cancer patients: assessment of curcumin levels in the colorectum and their pharmacodynamic consequences. Cancer Epidemiology Biomarkers & Prevention. 2005. 14(1): 1202 1251.
    [23] Maubach J, Bracke ME, Heyerick A, et al. Quantitati on of soy derived phytoestrogens in human breast tissue and biological fluids by high performance liquid chromatography. Journal of Chromatography B Analytical Technologies in the Biomedical and Life Sciences. 2003. 784(1): 137 144.
    [24] Boersma MG, VanderWoude H, Bogaards J, et al. Regioselectivity of phase II metabolism of luteolin and quercetin by UDP glucuronosyl transferases. Chemical Research in Toxicology. 2002. 15(5): 662 670.
    [25] Day AJ, Bao YP, Morgan MRA, et al. Conjugation positi on of quercetin glucuronides and effect on biological activity. Free Radical Biology and Medicine. 2000. 29(12): 1234 1243.
    [26] Setchell KD, Faughnan MS, Avades T, et al. Comparing the pharmacokinetics of daidzein and genisteinwith the use of 13C labeled tracers in premenopausal women. American Journal of Clinical Nutrition. 2003. 77(2): 411 419.
    [27] Graefe EU, Wittig J, Mueller S, et al. Pharmacokinetics and bioavailability of quercetin glycosides in humans. Journal of Clinical Pharmacology. 2001. 41(5): 492 499.
    [28] Donovan JL, Kasim Karakas S, German JB, et al. Urinary excreti on of catechin metabolites by human subjects after red wine consumption. British Journal of Nutrition. 2002. 87(1): 31 37.
    [29] Bravo L. Polyphenols: Chemistry, Dietary Sources, Metabolism, and Nutritional Significance. Nutrition Reviews. 1998. 56(11): 317–333.
    [30] Abu La? S, Odeh I, Dewik H, et al. Thymol and carvacrol production from leaves of wild Palestinian Majorana syriaca. Bioresource Technology. 2008. 99: 3914–3918.
    [31] Edward E, Langenau. The determination of thymol and carvacrol in Spanish thyme oils. Journal of the Science of Food and Agriculture. 1949. 38:261–266.
    [32] Cimanga K, Apers S, de Bruyne T, et al. Chemical composition and antifungal activity of essential oils of some aromatic medicinal plants growing in the Democratic Republic of Congo. Journal of Essential Oil Research. 2002. 14(5): 382 387.
    [33] Center for Food Safety and Applied Nutrition. 2006. EAFUS: a food additive database. Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Washington, DC. http://www.cfsan.fda.gov/dms /eafus.html. Consulted 22 May 2006.
    [34] Commission of the European Communities. 1999. Commission decision of 23 February 1999 adopting a register of ?avouring substances used in or on foodstuffs drawn up in application of regulation (EC) no. 2232/96 of the European Parliament and of the Council of 28 October 1996 (noti?ed under number C(1999)399) (text with EEA relevance) (1999/217/EC), p. 1–137. Of?cial Journal of the European Communities L084, 27/03/1999. Commission of the European Communities, Brussels, Belgium.
    [35] Baser KHC. Biological and Pharmacological Activities of Carvacrol and Carvacrol Bearing Essential Oils. Current Pharmaceutical Design. 2008. 29: 3106 3119.
    [36] Liolios CC, Gortzi O, Lalas S,et al. Liposomal incorporation of carvacrol and thymol isolated from the essential oil of Origanum dictamnus L. and in vitro antimicrobial activity. Food Chemistry. 2009. 112:77–83.
    [37] Iten F, Saller R, Abel G, et al. Additive Antmicrobial Effects of the Active Components of the Essential Oil of Thymus vulgaris Chemotype Carvacrol. Planta Medica. 2009, 75(11): 1231 1236
    [38] Chami F, Chami N, Bennis S, et al. Oregano and clove essential oils induce surface alteration of Saccharomyces Cerevisiae. Phytotherapy Research. 2005. 19(5): 405 408.
    [39] Jeong EY, Lim JH, Kim HG, et al. Acaricidal activity of Thymus vulgaris oil and its main components against Tyrophagus putrescentiae, a stored food mite. Journal of Food Protection. 2008. 71(2): 351 355.
    [40] Kordali S, Cakir A, Ozer H, et al. Antifungal, phytotoxic and insecticidal properties of essential oil isolated from Turkish Origanum acutidens and its three components, carvacrol, thymol and p cymene. Bioresource Technology. 2008. 99(18): 8788 8795.
    [41] Lambert RJW, Skandamis PN, Coote PJ, et al. A study of the minimum inhibitory concentration and mode of action of oregano essential oil, thymol and carvacrol. Journal of Applied Microbiology. 2001. 91(3): 453 462.
    [42] Veldhuizen EJA, Tjeerdsma Van Bokhoven JLM, Zweijtzer C, et al. Structural requirements for the antimicrobial activity of carvacrol. Journal of Agricultural and Food Chemistry. 2006. 54(5): 1874 1879.
    [43] Nostro A, Marino A, Blanco AR, et al. In vitro activity of carvacrol against staphylococcal preformed biofilm by liquid and vapour contact. Journal of Medical Microbiology. 2009. 58: 791–797
    [44] Palaniappan K, Holley RA. Use of natural antimicrobials to increase antibiotic susceptibility of drug resistant bacteria. International Journal of Food Microbiology. 2010.140: 164 168.
    [45] Rattanachaikunsopon P, Phumkhachorn P. In vitro study of synergistic antimicrobial effect of carvacrol and cymene on drug resistant Salmonella typhi. African Journal of Microbiology Research. 2009. 3: 978 980.
    [46] Zhou F, Ji BP, Zhang H, et al. Synergistic effect of thymol and carvacrol combined with chelators and organic acids against Salmonella typhimurium. Journal of Food Protection. 2007. 70: 1704 1709.
    [47] Chami N, Bennis S, Chami F, et al. Study of anticandidal activity of carvacrol and eugenol in vitro and in vivo. Oral Microbiology Immunology. 2005. 20: 106–111.
    [48] Pei RS, Zhou F, Ji BP,et al. Evaluation of Combined Antibacterial Effects of Eugenol, Cinnamaldehyde, Thymol, and Carvacrol against E coli with an Improved Method. Journal of Food Science. 2009, 74: 379 383.
    [49] Obaidat MM, Frank JF. Inactivation of Salmonella and Escherichia coli O157:H7 on Sliced and Whole Tomatoes by Allyl Isothiocyanate, Carvacrol, and Cinnamaldehyde in Vapor Phase. Journal of Food Protection. 2009. 72: 315 324.
    [50] Veldhuizen EJA, Creutzberg TO, Burt SA, et al. Low temperature and binding to food components inhibit the antibacterial activity of carvacrol against Listeria monocytogenes in steak tartare. Journal of Food Protection. 2007. 70: 2127 2132.
    [51] Ultee A, Kets EPW, Alberda M et al. Adaptation of the food borne pathogen Bacillus cereus to carvacrol. Archives of Microbiology. 2000. 174: 233 238.
    [52] Rattanachaikunsopon P, Phumkhachorn P. Assessment of synergistic efficacy of carvacrol and cymene against Edwardsiella tarda in vitro and in Tilapia (Oreochromis niloticus). African Journal of Microbiology Research. 2010. 4: 420 425.
    [53] Michiels J, Missotten J, Fremaut D, et al. In vitro dose–response of carvacrol, thymol, eugenol and trans cinnamaldehyde and interaction of combinations for the antimicrobial activity against the pig gut flora. Livestock Science. 2007. 109: 157–160.
    [54] Kordali S, Cakir A, Ozer H,et al. Antifungal, phytotoxic and insecticidal properties of essential oil isolated from Turkish Origanum acutidens and its three components, carvacrol, thymol and p cymene. Bioresource Technology. 2008. 99: 8788 8795.
    [55] Del Nobile MA, Conte A. Incoronato AL, et al. Antimicrobial ef?cacy and release kinetics of thymol from zein ?lms. Journal of Food Engineering. 2008. 89:57–63.
    [56] Martínez Romero D, Guillén F, Valverde JM, et al. Influence of carvacrol on survival of Botrytis cinerea inoculated in table grapes. International Journal of Food Microbiology. 2007. 115: 144 148.
    [57] Akgul A, Kivanc M, Sert S. Effect of carvacrol on growth and toxin production by aspergillus flavus and asperigillus parasiticus. Sciences Des Aliments. 1991. 11: 361 370.
    [58] Menniti AM, Gregori R, Neri F. Activity of natural compounds on Fusarium verticillioides and fumonisin production in stored maize kernels. International Journal of Food Microbiology. 2010. 136: 304 309.
    [59] Michiels J, Missotten J, Van Hoorick A, et al. Effects of dose and formulation of carvacrol and thymol on bacteria and some functional traits of the gut in piglets after weaning. Archives of Animal Nutrition. 2010. 64: 136 154.
    [60] Undeger U, Basaran A, Degen GH, et al. Antioxidant activities of major thyme ingredients and lack of (oxidative) DNA damage in V79 Chinese hamster lung fibroblast cells at low levels of carvacrol and thymol. Food and Chemical Toxicology. 2009. 47: 2037 2043.
    [61] Luna A, Labaque MC, Zygadlo JA, et al. Effects of thymol and carvacrol feed supplementation on lipid oxidation in broiler meat. Poultry Science. 2010. 89: 366 370.
    [62] Liolios CC, Gortzi O, Lalas S, et al. Liposomal incorporation of carvacrol and thymol isolated from the essential oil of Origanum dictamnus L. and in vitro antimicrobial activity. Food Chemistry. 2009. 112: 77 83.
    [63] Baca P, Clavero J, Baca AP, et al. Effect of chlorhexidine thymol varnish on root caries in a geriatric population: A randomized double blind clinical trial. Journal of Dentistry. 2009. 37: 679–685.
    [64] Botelho MA, Martins JG, Ruela RS, et al. Protective Effect of Locally Applied Carvacrol Gel on Ligature induced Periodontitis in Rats: A Tapping Mode AFM Study. Phytotherapy Research. 2009, 23: 1439 1448.
    [65] Landa P, Kokoska L, Pribylova M, et al. In vitro Anti inflammatory Activity of Carvacrol: Inhibitory Effect on COX 2 Catalyzed Prostaglandin E2 Biosynthesis. Archives of Pharmacal Research. 2009. 32: 75 78.
    [66] Kimura K, Yamaoka M, Kamisaka Y. Inhibition of Lipid Accumulation and Lipid Body Formation in Oleaginous Yeast by Effective Components in Spices,Carvacrol, Eugenol, Thymol, and Piperine. Journal of agriculture and food chemistry. 2006. 54: 3528 3534.
    [67] Aristatile B, Al Numair KS, Veeramani C, et al. Antihyperlipidemic effect of Carvacrol on D galactosamine induced Hepatotoxic Rats. Journal of Basic and Clinical Physiology and Pharmacology. 2009. 20(1): 15 27.
    [68] Aristatilea B, Al Numair KS, Veeramani C, et al. Effect of carvacrol on hepatic marker enzymes and antioxidant status in D galactosamine induced hepatotoxicity in rats. Fundamental & Clinical Pharmacology. 2009. 23: 757 765.
    [69] Case GL, He L, Mo HB, et al. Induction of Geranyl Pyrophosphate Pyrophosphatase Activity by Cholesterol Suppressive Isoprenoids. Lipids. 1995. 30(4): 357 359.
    [70] Uyanoglua M, Canbek M, Aral E, et al. Effects of carvacrol upon the liver of rats undergoing partial hepatectomy. Phytomedicine. 2008. 15: 226–229.
    [71] Canbek M, Uyanoglu M, Bayramoglu G, et al. Effects of carvacrol on defects of ischemia reperfusion in the rat liver. Phytomedicine. 2008. 15: 447–452.
    [72] Karkabounas S, Kostoula OK, Daskalou T, et al. Anticarcinogenic and antiplatelet effects of carvacrol. Experimental Oncology. 2006. 2: 121 125.
    [73] Chantasart D, Pongjanyakul T, Higuchi WI, et al. Effects of Oxygen Containing Terpenes as Skin Permeation Enhancers on the Lipoidal Pathways of Human Epidermal Membrane. Journal of Pharmaceutical Sciences. 2009. 98: 3617 3632.
    [74] Xu J, Zhou F, Ji BP, et al. The antibacterial mechanism of carvacrol and thymol against Escherichia coli. Letters in Applied Microbiology. 2008. 47: 174 179.
    [75] Ultee A, Slump RA, Steging G, et al. Antimicrobial activity of carvacrol toward Bacillus cereus on rice. Journal of Food Protection. 2000. 63(5): 620 624.
    [76] Ultee A, Kets EPW, Smid EJ, et al. Mechanisms of action of carvacrol on the food borne pathogen Bacillus cereus. Applied and Enviromental Microbiology. 1999. 65(10): 4606 4610.
    [77] Gill AO, Holley RA. Inhibition of membrane bound ATPases of Escherichia coli and Listeria monocytogenes by plant oil aromatics. International Journal of Food Microbiology. 2006. 111(2): 170 174.
    [78] Huang TC, Lin YT, Chuang KP. Carvacrol has the priming effects of reactive oxygen species (ROS) production in C6 glioma cells. Food and Agricultural Immunology. 2010. 21: 47 55.
    [79] Burt SA., Zee Rvd, Koets AP, et al. Carvacrol Induces Heat Shock Protein 60 andInhibits Synthesis of Flagellin in Escherichia coli O157:H7. Applied and Environmental Microbiology. 2007. 14: 4484–4490
    [80] Lee J, Jung E, Yu H, et al. Mechanisms of carvacrol induced expression of type I collagen gene. Journal of Dermatological Science. 2008. 52: 160 169.
    [81] Arfa AB, Combes S, Preziosi Belloy L, et al. Antimicrobial activity of carvacrol related to its chemical structure. Letters in Applied Microbiology. 2006. 43: 149–154.
    [82] Veldhuizen EJA, Bokhoven JLMT V, Zweijtzer C, et al. Structural Requirements for the Antimicrobial Activity of Carvacrol. Journal of agriculture and food chemistry. 2006. 54: 1874 1879.
    [83] Ben Arfa A, Combes S, Preziosi Belloy L, et al. Antimicrobial activity of carvacrol related to its chemical structure. Letters in Applied Microbiology. 2006. 43(2): 149 154.
    [84] Mariko H, Rieko N, Michiko K,et al. Carvacrol, a component of thyme oil, activates PPAR alpha and gamma and suppresses COX 2 expression. Journal of Lipid Research. 2010. 51: 132 139.
    [85] Lee SP, Buber MT, Yang Q,et al. Thymol and related alkyl phenols activate the hTRPA1 channel. British Journal of Pharmacology. 2008. 153: 1739–1749.
    [86] Moshe P, Maximilian P, Daniela D, et al. Carvacrol is a novel inhibitor of Drosophila TRPL and mammalian TRPM7 channels. Cell Calcium. 2009. 45: 300 309.
    [87] Deighton N, Glidewell SM, Deans SG, et al. Identification by epr spectroscopy of carvacrol and thymol as the major sources of free radicals in the oxidation of plant essential oils. Journal of the Science of Food and Agriculture. 1993. 2: 221–225.
    [88] Na?si Sh, Hajiakhoondi A, Yektadoost A. Thymol and Carvacrol Binding to DNA: Model for Drug–DNA Interaction Biopolymers.2004. 74: 345–351.
    [89] Slamenova D, Horvathova E, Sramkova M,et al. DNA protective effects of two components of essential plant oils carvacrol and thymol on mammalian cells cultured in vitro. Neoplasma. 2007. 54: 108 112.
    [90] http://www.epa.gov/fedrgstr/EPA PEST/2009/March/Day 25/p6262.pdf
    [91] Jenner PM, Hagan EC, Taylor JM, et al. Food flavourings and compounds of related structure I. Acute oral toxicity. Food and Cosmetics Toxicology. 1964. 2: 327 343.
    [92] von Angerer E, Kranzfelder G, Taneja AK, et al. N,N' Dialkylbis(dichlorophenyl)ethylenedi amines and imidazolidines: relationship between structure and estradiol receptor affinity. Journal of Medicinal Chemistry. 1980, 23(12): 1347 50.
    [93] Azirak S, Rencuzogullari E. The in vivo genotoxic effects of carvacrol and thymol in rat bone marrow cells. Environment Toxicology. 2008. 23: 728–735.
    [94] Bimczok D, Rau H, Sewekowa E, et al. In?uence of carvacrol on proliferation and survival of porcine lymphocytes and intestinal epithelial cells in vitro. Toxicology in Vitro. 2008. 22: 652–658
    [95] Kim DK, Lillehoj HS, Lee SH, et al. High throughput gene expression analysis of intestinal intraepithelial lymphocytes after oral feeding of carvacrol, cinnamaldehyde, or Capsicum oleoresin. Poultry Science. 2010, 89: 68 81.
    [96] Monzote L, Stamberg W, Staniek K, et al. Toxic effects of carvacrol, caryophyllene oxide, and ascaridole from essential oil of Chenopodium ambrosioides on mitochondria. Toxicology and Applied Pharmacology. 2009. 240: 337–347.
    [97] Ternes W, Gronemeyer M, Schwarz K. Determination of p eymene 2,3 diol, thymol, and earvaerol in different foodstuffs. Zeitschrift Fur Lebensmittelunter suchung Und Forschung A. 1995. 201(6): 544 547
    [98] Alekseeva LI. Determining thymol and carvacrol by reversed phase high performance liquid chromatography. Pharmaceutical Chemistry Journal. 2008. 43(12): 665 667.
    [99] Heil TP, Lindsayb RC. A method for quantitative analysis of flavor tainting alkylphenols and aromatic thiols in fish. Journal of Environmental Science and Health. 1988. 23: 475 488.
    [100] Kohlert C, Abel G, Schmid E, et al. Determination of thymol in human plasma by automated headspace solid phase microextraction–gas chromatographic analysis. Journal of Chromatography B. 2002. 767: 11–18.
    [101] Vincenzi MD, Stammati A, Vincenzi AD, et al. Constituents of aromatic plants: carvacrol. Fitoterapia. 2004. 75: 801–804.
    [102] Wang Q, Gong J, Huang X, et al. In vitro evaluation of the activity of microencapsulated carvacrol against Escherichia coli with K88 pili. Journal of Applied Microbiology. 2009. 107: 1781 1788.
    [103] Michiels J, Missotten J, Dierick N, et al. In vitro degradation and in vivo passage kinetics of carvacrol, thymol, eugenol and trans cinnamaldehyde along the gastrointestinal tract of piglets. Journal of the Science of Food and Agriculture.2008. 88: 2371–2381
    [104] Passreiter CM, Wilson J, Andersen R, et al. Metabolism of Thymol and trans Anethole in Larvae of Spodoptera litura and Trichoplusia ni (Lepidoptera: Noctuidae). Journal of Agriculture and Food Chemistry. 2004. 52: 2549 2551.
    [105] Shimoda K, Kondo Y, Nishida T, et al. Biotransformation of thymol, carvacrol, and eugenol by cultured cells of Eucalyptus perriniana. Phytochemistry. 2006. 67: 2256–2261.
    [106] Chamberlain EM,Dagley S. The Metabolism of Thymol by a Pseudomonas. Biochemical Journal. 1968. 110: 755 764.
    [107] Jiang YY, He X, de Montellano PRO. Radical Intermediates in the Catalytic Oxidation of Hydrocarbons by Bacterial and Human Cytochrome P450 Enzymes. 2006. 45(2): 533 542.
    [108] Dudai N, Larkov O, Mayer AM, et al. Metabolism of essential oils during inhibition of wheat seed germination. Seed Biology: Advances and Applications. 2000.18: 315 320.
    [109] Varel VH. Carvacrol and Thymol Reduce Swine Waste Odor and Pathogens:Stability of Oils. Current Microbiology. 2002. 44: 38 43.
    [110] Austgulen LT, Solheim E, Scheline RR. Metabolism in rats of p cymene derivatives: carvacrol and thymol. Pharmacology & Toxicology. 1987. 61: 98 102.
    [111] Takada M, Agata I, Sakamoto M, et al. On the metabolic detoxication of thymol in rabbit and man. The Journal of Toxicological Sciences. 1979. 4: 341 350.
    [112] Kohlert C, Schindler G, M?rz RW, et al. Systemic Availability and Pharmacokinetics of Thymol in Humans. Journal of Clinical Pharmacology. 2002. 42: 731 737.
    [113] Abid A, Bouchon I, Siest G, et al. Glucuronidation in the Caco 2 Human Intestinal Cell Line: Induction of UDP Glucuronosyltransferase 1*6. Biochemical Pharmacology. 1995. 50: 557 561.
    [114] SENAFI SB, CLARKE DJ, Burchell B. Investigation of the substrate specificity of a cloned expressed human bilirubin UDP glucuronosyltransferase: UDP sugar specificity and involvement in steroid and xenobiotic glucuronidation. Biochemcal Journal.1994. 303: 233 240.
    [115] Jedlitschky G, Cassidy AJ, Sales M, et al. Cloning and characterization of a novel human olfactory UDP glucuronosyltransferase. Biochemcal Journal. 1999. 340: 837–843.
    [116] Turgeon D, Carrier J S, Chouinard S, et al. Glucuronidation activity of the UGT2B17 enzyme toward xenobiotics. Drug Metabolism and Disposition. 2003. 31: 670 676.
    [117] Basu NK, Ciotti M, Hwang MS, et al. Differential and Special Properties of the Major Human UGT1 encoded Gastrointestinal UDP glucuronosyltransferases Enhance Potential to Control Chemical Uptake. The Journal of Biological Chemistry. 2004. 279: 1429–1441.
    [118] Smith PA, Sorich MJ, McKinnon RA, et al. In silico Insights: chmical and structural characteristics associated with uridine diphosphate glucuronosyltransf erase substrate selectivity. Clinical and Experimental Pharmacology and Physiology. 2003. 30: 836–840.
    [119] Smith PA, Sorich MJ, McKinnon RA, et al. Pharmacophore and Quantitative Structure Activity Relationship Modeling: Complementary Approaches for the Rationalization and Prediction of UDP Glucuronosyltransferase 1A4 Substrate Selectivity. Journal of Medicinal Chemistry. 2003. 46: 1617 1626.
    [120] Guellec CL, Lacarelle B, Villard P H, et al.Glucuronidation of Propofol in Microsomal Fractions from Various Tissues and Species Including Humans: Effect of Different Drugs. Anesthesia and Analgesia. 1995. 81: 855 61.
    [121] Jukic M, Politeo O, Maksimovic M, et al. In Vitro Acetylcholinesterase Inhibitory Properties of Thymol, Carvacrol and their Derivatives Thymoquinone and Thymohydroquinone. Phytotherapy Research. 2007. 21: 259 261.

© 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