147-Pharmacology-Chen-BB
参一胶囊对恶性肿瘤患者血清血管内皮生长因子影响的Meta分析

参一胶囊对恶性肿瘤患者血清血管内皮生长因子影响的Meta分析成远;陈超;陈歆妮;华海清【期刊名称】《现代肿瘤医学》【年(卷),期】2015(023)006【摘要】目的:系统评价参一胶囊对恶性肿瘤患者血清VEGF的影响.方法:利用计算机和手工方式检索Pubmed、Cochrane图书馆、中国生物医学文献数据库、中国期刊全文数据库、中文科技期刊数据库、万方数据数、中国中医药数据库.检索参一胶囊治疗恶性肿瘤的随机对照临床研究.3位研究者对文献质量进行独立的评价,用Cochrane Collaboration's software RevMan5.2进行Meta分析.结果:共有20项研究被纳入本系统评价,纳入病例1299例.Meta分析显示参一胶囊治疗能够显著降低血清VEGF[SMD=-1.75,95% CI(-2.32,-1.17),P<0.00001],参一胶囊治疗12周较治疗4周能进一步降低血清VEGF[SMD=2.35,95%CI(0.76,3.95),P=0.004)].结论:参一胶囊能够降低血清VEGF,这一证据需要更大规模的多中心随机临床对照研究进一步验证.【总页数】6页(P844-849)【作者】成远;陈超;陈歆妮;华海清【作者单位】南京中医药大学附属南京八一医院全军肿瘤中心内科,江苏南京210002;南京中医药大学附属南京八一医院全军肿瘤中心内科,江苏南京210002;南京中医药大学附属南京八一医院全军肿瘤中心内科,江苏南京210002;南京中医药大学附属南京八一医院全军肿瘤中心内科,江苏南京210002【正文语种】中文【中图分类】R730.58【相关文献】1.妇科恶性肿瘤患者血清中血管内皮生长因子测定的临床意义 [J], 彭小萍;李俊东;李孟达;叶鲜明;颜婉嫦2.重组人血管内皮抑制素联合化疗对中晚期恶性肿瘤患者化疗效果及血管内皮生长因子水平的影响 [J], 张秦;陈华飞;朱有才3.消化道恶性肿瘤患者血清肿瘤特异性生长因子与血管内皮生长因子水平检测及意义 [J], 张家明;洪流;卓少贤;温本;谢朝阳4.国内恶性肿瘤患者营养不良影响因素Meta分析 [J], 张艳华;李晓玲;李增宁5.叙事疗法对恶性肿瘤患者负性情绪影响的Meta分析 [J], 李炫桦;胡燕琪;唐红梅因版权原因,仅展示原文概要,查看原文内容请购买。
periodical__zglcylxzz__zglc2009__0903pdf__090317

ChinJClinPharmacoI257V01.25No.3May2009(SerialNo.119)中药脑脊液药理学方法研究进展AdvanceinsearchforcerebrospinalfluidpharmacologyinChinesemateriamedica闰晨,康立源(天津巾医药大学中疾药研究院省部共建方剂学重点实验室,天津300193)YANChen,KANGLi—yuan(The研LaboratoryforPrescriptionofNationalEducationalMinistryofChi—n口.TraditionalChineeMedicineRe.searchCenter,TianjinUniversityofTra—ditionalChineseMedicine.死onjin300193.China)收稿日期:2008—03一10修回日期:2008—10—21资金项目:天津市高等学校科技发展计划基金资助项日(2006305)作者简介:闰晨(1985一),女。
硕士研究生。
主要从事lfI药防治脑血管疾病研究通讯作者:康立源,副研究员Tel:(022)23051183E・mail:klyzm@163.tom摘要:随着中医药研究的不断深入以及心脑血管疾病防治的需要,反映体内真实血药浓度下的中药作用物质基础和作用机制研究,已成为药理方法学研究的重点。
血清药理学方法的广泛应用,已为中药体外研究提供了一种科学方法;但存在中药复方应用于离体实验时,体内外环境不完全一致等不足。
中药脑脊液药理学方法的提出,初步解决了这一难点,为中医药研究中选择适宜的细胞离体培养体系提供了新途径,更为中药复方作用的物质基础和作用机制研究提供了新思路。
关键词:血清药理学;中药脑脊液药理学;离体培养中图分类号:R285.6;R285.1文献标识码:A文章编号:1001—6821(2009)03—0257一03Abstract:AlongwiththedevelopingofresearchonChinesemedicine,aswellascardiovascularandeerebrovasculardiseasesneedtoprevent,reflectmaterialbasisandmechanismofpharmacologicalmethodsofherbsinvivoplasmaconcentrationreallybecomethefocusofstudy.Serophar-macologymethodprovidesabasisforChinesemedicineinvitrostudy,butthereissomeshortcomingsinvitroexperiments.Therefore,thedevel-opofcerebrospinalfluidpharmacologyofChinesemateriamedicaprelim—inaryprovidesanewideaforthestudyoftraditionalChinesemedicine.Keywords:sero-pharmacology;eerebrospinalfluidpharmacologyofChinesemateriamedica;cultureinvitro中药复方作为祖国医药宝库的重要组成部分,是中医治疗法则在组方用药上的具体应用,是中医药防治疾病的重要手段。
一种卡培他滨药用组合物及其制备方法[发明专利]
![一种卡培他滨药用组合物及其制备方法[发明专利]](https://img.taocdn.com/s3/m/a4a728a2b307e87100f6966b.png)
专利名称:一种卡培他滨药用组合物及其制备方法专利类型:发明专利
发明人:程雪翔
申请号:CN201110189145.7
申请日:20110707
公开号:CN102266303A
公开日:
20111207
专利内容由知识产权出版社提供
摘要:本发明公开了一种卡培他滨片剂,所述片剂按重量份组成为:卡培他滨100-600,微晶纤维素10-80,乳糖10-70,交联羧甲基纤维素钠30-100,1%羟丙甲纤维素50%乙醇溶液10-70,硬脂酸镁1-10,纯化水适量。
申请人:程雪翔
地址:100101 北京市朝阳区小营路13号亚非大厦9008室
国籍:CN
代理机构:北京品源专利代理有限公司
代理人:陈慧珍
更多信息请下载全文后查看。
2024贝伐珠单抗长期治疗诱导胶质母细胞瘤侵袭转移的研究进展要点(全文)

2024贝伐珠单抗长期治疗诱导胶质母细胞瘤侵袭转移的研究进展要点(全文)胶质母细胞瘤(glioblasto ma,GBM)是最常见和最具侵袭性的原发性脑肿瘤。
尽管对GBM进行手术、放疗和化疗,但其复发仍不可避免。
目前标准治疗方案是最大程度地安全切除,然后进行放化疗(CRT)。
放射治疗总剂量60Gy,在6周内分30次完成,同时每日使用替莫嗤胺,后续辅助替莫嗤胺治疗6个月。
诊断和治疗后的中位生存期为12~15个月。
美国目前GBM5年生存率约为5%。
在替莫嗤胺之外,美国食品药品监督管理局千2009年快速批准贝伐株单抗(bevacizumab,BVZ)用千治疗GBM。
BVZ是一种靶向抑制血管内皮生长因子(vascular endothelial growth factor, VEG F)的特异性抗体,其试图阻止肿瘤血管生成,从而减少肿瘤血液供应,减缓肿瘤细胞扩散。
然而随着研究不断深入,研究发现:B VZ 对胶质瘤仅发挥轻微的抗肿瘤作用,主要用千症状控制,在总生存率方面无显著益处,反而会增强肿瘤侵袭性。
本文就BV Z治疗GBM诱发侵袭转移的机制,以及用千预测BVZ治疗反应的特异性标志物展开论述。
1.BVZ在G BM的应用BVZ治疗GBM的首次临床试验是2009年的“AVF3708g/BRAIN"和“NCI06-C-0064E二期试验。
在试验中,BVZ单药或联合伊立替康治疗GBM的客观有效率为28%~40%,6个月无进展生存率为40%~50%,与较高的历史对照组相比改善显著,但总体生存率为38%~40%并无改善。
随后,2014年完成的两项田期临床试验评估在原发GBM中应用BV Z 辅助标准放化疗方案的价值,研究结果显示:应用BVZ联合标准放化疗治疗的病人与仅采用标准放化疗方案的病人相比,无进展生存期(progression-free survival, PFS)有显著改善(10.6个月VS6.2个月),但总生存期(o verall survival, OS)并无显著差异(16.7个月VS 16.8个月)。
红景天苷对Lewis肺癌小鼠恶性胸腔积液的抑制作用及其对免疫功能的影响

红景天苷对Lewis肺癌小鼠恶性胸腔积液的抑制作用及其对免疫功能的影响王松海;凡瑾;陈捷【期刊名称】《陕西医学杂志》【年(卷),期】2024(53)1【摘要】目的:探究红景天苷(SAL)对肺癌小鼠恶性胸腔积液生成的抑制作用及对其免疫功能的影响。
方法:通过胸腔注射Lewis肺癌细胞(LLC)建立C57BL/6小鼠恶性胸腔积液(MPE)模型;建模后随机分为正常组、模型组、SAL组、顺铂组(DDP 组)、红景天苷+顺铂联合组(SAL+DDP组);实验结束后,收集各组小鼠胸水并测量胸水体积、生存周期。
酶联免疫吸附法(ELISA)检测MPE小鼠血清中肿瘤坏死因子-α(TNF-α)、低氧诱导因子-1α(HIF-1α)、血管内皮生长因子(VEGF)水平;流式细胞仪定量检测T细胞亚群CD3+、CD4+、CD8+、CD4+/CD8+及自然杀伤(NK)细胞百分比。
结果:MPE模型小鼠经治疗后,与模型组比较,DDP组、联合组小鼠生存周期显著延长(均P<0.01);SAL组、DDP组和联合组MPE体积均显著减少(均P<0.01);SAL组、DDP组和联合组外周血中TNF-α水平显著升高,HIF-1α、VEGF 水平显著降低(均P<0.05);SAL组、DDP组和联合组CD3+均显著升高(均P<0.05);SAL组、联合组CD4+显著升高(均P<0.05),DDP组升高无统计学意义(P>0.05);SAL组、联合组CD8+、CD4+/CD8+、NK均显著升高(均P<0.05),DDP组降低无统计学意义(P>0.05)。
结论:SAL联合顺铂胸腔灌注抑制肺癌小鼠MPE生成效果显著,可能与HIF-1α-VEGF分子通路抑制以及免疫功能激活有关。
【总页数】5页(P37-41)【作者】王松海;凡瑾;陈捷【作者单位】陕西省中医医院肿瘤科;陕西中医药大学【正文语种】中文【中图分类】R734.2【相关文献】1.三叶青乙酸乙酯提取物对小鼠Lewis肺癌移植性肿瘤的抑制作用及免疫功能的影响2.通关藤苷H对小鼠Lewis肺癌的抑制作用及对荷瘤小鼠免疫功能的影响3.苏铁总黄酮对Lewis肺癌模型小鼠的抑制作用及免疫功能的影响4.红景天苷对Lewis荷瘤小鼠调节性T细胞的抑制作用5.红景天苷对Lewis肺癌小鼠恶性胸腔积液生成的影响因版权原因,仅展示原文概要,查看原文内容请购买。
Rho激酶抑制剂Y27632促进人诱导多能干细胞来源原始神经上皮细胞向多巴胺能神经前体细胞的转化

参考文献:[1]Virani SS,Alonso A,Aparicio HJ,et al.Heart disease and strokestatistics-2021update:a report from the American heart association [J ].Circulation,2021,143(8):e254-743.[2]李天伦,张中,赵蓓,等.急性心肌梗死患者血浆白细胞介素22水平与冠状动脉病变程度和预后的关系[J ].海军军医大学学报,2022,43(4):398-405.[3]Yeh KC,Lee CJ,Song JS,et al.Protective effect of CXCR4antagonist DBPR807against ischemia-reperfusion injury in a rat and porcine model of myocardial infarction:potential adjunctive therapy for percutaneous coronary intervention [J ].Int J Mol Sci,2022,23(19):11730.[4]Zhou ML,Yu YF,Luo XX,et al.Myocardial ischemia-reperfusioninjury:therapeutics from a mitochondria-centric perspective [J ].I/R+CCC I/R+CCC+DSMPSOI/R图7秋水仙碱通过激活AMPK 逆转I/R 手术导致的小鼠心功能下降和心脏损伤Fig.7Colchicine reverses reduced cardiac function and cardiac damage in mice induced by I/R by activating AMPK.A :Representative M-mode ultrasound images of the mice in each group.B -D :Quantitative analysis of the LVEF,LVFS and HR (n =4).E :Representative TTC staining images of the mice in each group.F -H :Quantitative analysis of the infarct area and serum cTnT and LDH levels (n =4).*P <0.05vs SO group;#P <0.05vs I/R group;&P <0.05vs I/R+colchicine group.B CEF ADG H I /RS O I /R+C C C I /R +C CC +D SM P L V E F (%)#*&100.090.080.070.060.050.040.0I /RS O I /R +CC CI /R +C C C +D SM P L V F S (%)SO I/RI/R+CCC I/R+CCC+DSMP60.050.040.030.020.010.00.0H R (B P M )500.0400.0300.0200.0100.00.0I /RS O I /R +CC C I /R +C C C +D SM P I /RS O I /R +C C C I /R +C CC +D SM P I /RS O I /R +C C C I /R +C C C +D SM P I /RS O I /R+C C C I /R +C CC +D SM P I n f a r c a r e a (%)70.060.050.040.030.020.010.00.0L D H i n s e r u m (U /L )1600.01400.01200.01000.0800.0600.0400.0200.00.0c T n T i n s e r u m (p g /m L )450.0400.0350.0300.0250.0200.0150.0100.050.00.0#*&#*&#*&#*&J South Med Univ,2024,44(2):226-235··234Cardiology,2021,146(6):781-92.[5]Chen MX,Li XP,Yang H,et al.Hype or hope:Vagus nerve stimulation against acute myocardial ischemia-reperfusion injury [J].Trends Cardiovasc Med,2020,30(8):481-8.[6]Liu X,Xu L,Wu J,et al.Down-regulation of SIK2expression alleviates myocardial ischemia-reperfusion injury in rats by inhibiting autophagy through the mTOR-ULK1signaling pathway [J].J South Med Univ,2022,42(7):1082-8.[7]Lu CH,Guo X,He XH,et al.Cardioprotective effects of sinomenine in myocardial ischemia/reperfusion injury in a rat model[J].Saudi Pharm J,2022,30(6):669-78.[8]Cadenas S.ROS and redox signaling in myocardial ischemia-reperfusion injury and cardioprotection[J].Free Radic Biol Med, 2018,117:76-89.[9]Maximilian Buja L.Mitochondria in ischemic heart disease[J].Adv Exp Med Biol,2017,982:127-40.[10]Peoples JN,Saraf A,Ghazal N,et al.Mitochondrial dysfunction and oxidative stress in heart disease[J].Exp Mol Med,2019,51(12):1-13.[11]Zou RJ,Shi WT,Qiu JX,et al.Empagliflozin attenuates cardiac microvascular ischemia/reperfusion injury through improving mitochondrial homeostasis[J].Cardiovasc Diabetol,2022,21(1): 106.[12]Tong DC,Wilson AM,Layland J.Colchicine in cardiovascular disease:an ancient drug with modern tricks[J].Heart,2016,102(13):995-1002.[13]Elshafei MN,El-Bardissy A,Khalil A,et al.Colchicine use might be associated with lower mortality in COVID-19patients:a meta-analysis[J].Eur J Clin Invest,2021,51(9):e13645.[14]Deftereos SG,Beerkens FJ,Shah B,et al.Colchicine in cardio-vascular disease:In-depth review[J].Circulation,2022,145(1):61-78.[15]Wang LR,Shan YL,Chen L,et al.Colchicine protects rat skeletal muscle from ischemia/reperfusion injury by suppressing oxidative stress and inflammation[J].Iran J Basic Med Sci,2016,19(6):670-5.[16]Tang YJ,Shi CY,Qin YY,et work pharmacology-based investigation and experimental exploration of the antiapoptotic mechanism of colchicine on myocardial ischemia reperfusion injury [J].Front Pharmacol,2021,12:804030.[17]李晨霏,樊迪,杨政,等.AMPK在心肌纤维化相关疾病中的作用及机制研究进展[J].解放军医学杂志,2021,46(12):1239-44.[18]胡淼,童旭辉,黄杰,等.基于铁死亡探讨AMPK抗小鼠脑缺血/再灌注损伤的作用及机制[J].华中科技大学学报:医学版,2021,50(4):418-23.[19]Zhang Y,Wang Y,Xu JN,et al.Melatonin attenuates myocardial ischemia-reperfusion injury via improving mitochondrial fusion/ mitophagy and activating the AMPK-OPA1signaling pathways[J].J Pineal Res,2019,66(2):e12542.[20]曾菲,李强,曾昪,等.氢溴酸加兰他敏介导AMPKα1/Nrf2/HO-1通路对大鼠心肌缺血再灌注损伤的保护作用[J].四川大学学报:医学版,2020,51(3):337-43.[21]Wang Y,Viollet B,Terkeltaub R,et al.AMP-activated protein kinase suppresses urate crystal-induced inflammation and transduces colchicine effects in macrophages[J].Ann Rheum Dis,2016,75(1): 286-94.[22]Lu YY,Chen YC,Kao YH,et al.Colchicine modulates calcium homeostasis and electrical property of HL-1cells[J].J Cell MolMed,2016,20(6):1182-90.[23]Liu HQ,Mo HQ,Yang CB,et al.A novel function of ATF3in suppression of ferroptosis in mouse heart suffered ischemia/reperfusion[J].Free Radic Biol Med,2022,189:122-35.[24]Akodad M,Fauconnier J,Sicard P,et al.Interest of colchicine in the treatment of acute myocardial infarct responsible for heart failure ina mouse model[J].Int J Cardiol,2017,240:347-53.[25]Yu HL,Liu Q,Chen GD,et al.SIRT3-AMPK signaling pathway as a protective target in endothelial dysfunction of early sepsis[J].IntImmunopharmacol,2022,106:108600.[26]Lv DY,Luo MH,Cheng Z,et al.Tubeimoside I ameliorates myocardial ischemia-reperfusion injury through SIRT3-dependentregulation of oxidative stress and apoptosis[J].Oxid Med CellLongev,2021,2021:5577019.[27]Xiang M,Lu YD,Xin LY,et al.Role of oxidative stress in reperfusion following myocardial iIschemia and Its treatments[J].Oxid Med Cell Longev,2021,2021:6614009.[28]Yue HH,Liang WT,Zhan YJ,et al.Colchicine:emerging therapeutic effects on atrial fibrillation by alleviating myocardial fibrosis in a ratmodel[J].Biomedecine Pharmacother,2022,154:113573.[29]Yang MY,Lv H,Liu Q,et al.Colchicine alleviates cholesterol crystal-induced endothelial cell pyroptosis through activating AMPK/SIRT1pathway[J].Oxid Med Cell Longev,2020,2020:9173530.[30]Xin T,Lu CZ.SirT3activates AMPK-related mitochondrial biogenesis and ameliorates sepsis-induced myocardial injury[J].Aging,2020,12(16):16224-37.[31]Feng LF,Ren JL,Li YF,et al.Resveratrol protects against isoproterenol induced myocardial infarction in rats through VEGF-B/AMPK/eNOS/NO signalling pathway[J].Free Radic Res,2019,53(1):82-93.[32]Tian L,Cao WJ,Yue RJ,et al.Pretreatment with Tilianin improves mitochondrial energy metabolism and oxidative stress in rats withmyocardial ischemia/reperfusion injury via AMPK/SIRT1/PGC-1alpha signaling pathway[J].J Pharmacol Sci,2019,139(4):352-60.[33]吴志林,朱轶.右美托咪定通过Trx1/AMPK通路减轻心肌缺血再灌注损伤中的氧化应激[J].华中科技大学学报:医学版,2020,49(4):404-7.[34]Wu SN,Zou MH.AMPK,mitochondrial function,and cardio-vascular disease[J].Int J Mol Sci,2020,21(14):4987.[35]秦秀男,秦溱,冉珂,等.七氟醚预处理通过线粒体NAD+-SIRT3通路减轻大鼠心肌缺血再灌注损伤[J].中南大学学报:医学版,2022,47(8):1108-19.[36]韦亚忠,薛晓梅,何斌.活性氧介导心肌缺血再灌注损伤的研究进展[J].上海交通大学学报:医学版,2021,41(6):826-9.[37]Paradies G,Paradies V,Ruggiero FM,et al.Mitochondrial bioenergetics and cardiolipin alterations in myocardial ischemia-reperfusion injury:implications for pharmacological cardiopro-tection[J].Am J Physiol Heart Circ Physiol,2018,315(5):H1341-52.[38]Brenner D,Mak TW.Mitochondrial cell death effectors[J].Curr Opin Cell Biol,2009,21(6):871-7.(编辑:经媛) J South Med Univ,2024,44(2):226-235··235帕金森病(PD )是一种进行性神经退行性疾病,是60岁以上人群中第2常见的神经退行性疾病,其主要原因是黑质致密部(SNc )多巴胺能(DA )神经元的死亡和含α-突触核蛋白的路易体的形成[1]。
碧云天生物技术一氧化氮检测试剂盒说明书

碧云天生物技术/Beyotime Biotechnology 订货热线:400-1683301或800-8283301 订货e-mail :******************技术咨询:*****************网址:碧云天网站 微信公众号一氧化氮检测试剂盒产品编号 产品名称包装 S0021S 一氧化氮检测试剂盒 500次 S0021M一氧化氮检测试剂盒2500次产品简介:碧云天生产的一氧化氮检测试剂盒采用了经典的Griess Reagent ,并对其测定的溶液体系进行了优化,使检测下限达到1µM ,在1-100µM 范围内有非常完美的线性关系。
检测速度极快,完成一条标准曲线或5-10个样品的测定只需3分钟。
样品范围广,可以检测细胞或组织及其培养液中的一氧化氮的含量,酚红和10%血清均对测定无明显干扰,也可以检测血清、血浆和尿液中一氧化氮的含量。
包装清单:产品编号 产品名称 包装 S0021S-1 1M NaNO 2 1ml S0021S-2 Griess Reagent I 25ml S0021S-3 Griess Reagent II25ml —说明书1份产品编号 产品名称 包装 S0021M-1 1M NaNO 2 1ml S0021M-2 Griess Reagent I 125ml S0021M-3Griess Reagent II125ml —说明书1份保存条件:-20ºC 避光保存,一年有效。
4ºC 避光保存,半年有效。
注意事项:本产品对人体有害,操作时请小心,并注意有效防护以避免直接接触人体或吸入体内。
如保存不当导致溶液变色或沉淀,则说明该溶液已经失效,请购买新的试剂盒。
不建议使用RIPA 裂解液对细胞或者组织进行裂解,使用RIPA 裂解液可能在后续反应中产生沉淀,影响测试。
推荐使用碧云天的细胞与组织裂解液(一氧化氮检测用)(S3090)或Western 及IP 细胞裂解液(P0013)。
李时珍的英语作文

李时珍的英语作文Li Shizhen, also known as Li Shih-Chen in the Western world, was a Chinese physician and pharmacologist who lived in the Ming dynasty. 李时珍,也被称为李时珍在西方世界,是一位生活在明朝的中国医生和药剂师。
He is best known for his significant contribution to Chinese medicine through his renowned work, the Compendium of Materia Medica (Bencao Gangmu). This comprehensive medical text contains detailed information about over 1,800 different kinds of drugs and their uses, as well as observations and experiences from over 3,000 herbal medicine practitioners.他以他著名的作品《本草纲目》对中国医学做出了重要贡献。
这部综合性的医学文本包含了对1800多种不同药物及其用途的详细信息,以及来自3000多名草药医药从业者的观察和经验。
Li Shizhen's dedication to the field of medicine and his tireless efforts to understand and document the rich knowledge of traditional Chinese medicine has earned him a well-deserved reputation as a pioneer in the history of Chinese pharmacology. His work has notonly had a profound impact on traditional Chinese medicine, but it has also contributed to the development and broadening of the understanding of natural medicine and pharmacology worldwide. 李时珍对医学领域的奉献以及他对了解和记录传统中医知识的不懈努力使他在中国药理学史上赢得了当之无愧的先驱声誉。
- 1、下载文档前请自行甄别文档内容的完整性,平台不提供额外的编辑、内容补充、找答案等附加服务。
- 2、"仅部分预览"的文档,不可在线预览部分如存在完整性等问题,可反馈申请退款(可完整预览的文档不适用该条件!)。
- 3、如文档侵犯您的权益,请联系客服反馈,我们会尽快为您处理(人工客服工作时间:9:00-18:30)。
E-Mail karger@ O riginal Paper P harmacology 2015;95:145–153D O I:10.1159/000380883 Effects of Methoxychlor and 2,2-bis ( p -Hydroxyphenyl)-1,1,1-Trichloroethane on Cytochrome P450 Enzyme Activities in Human and Rat LiversB ingbing Chen a Peipei Pan c Li Wang a Menchun Chen a Yaoyao Dong a Ren-Shan Ge b Guo-Xin Hu aaS chool of Pharmacy, and b T he 2nd Affiliated Hospital, Wenzhou Medical University, W enzhou , and cT aizhou Central Hospital, T aizhou , ChinaBoth chemicals had no effect on human CYP3A4 and ratC YP3A1 activity. In summary, MXC and HPTE are potent in-hibitors of some human and rat CYPs. © 2015 S. Karger AG, BaselI ntroduction Cytochrome P450 (CYP) enzymes are a superfamily of hemoproteins. They are involved in the metabolism of endogenous and exogenous compounds, including ste-roids, fatty acids, bile acids, lipid-soluble vitamins, drugs,and environmental pollutants[1–3] . Most CYPs are ex-pressed in the liver, but some are localized in extrahepat-ic tissues. In humans, CYP2C9, CYP2C19, CYP2D6, and CYP3A4 are the major drug-metabolizing CYPs in the liver, which are homologous with CYP2C11, CYP2C6,CYP2D2, and CYP3A1, respectively, in rats[4–6].These CYPs are responsible for the metabolism of many drugs. CYP induction or inhibition can alter the biotransforma-tion and bioavailability of many drugs. However, directK ey WordsC ytochrome P450 enzyme · Enzyme inhibition · Methoxychlor · 2,2-bis( p -Hydroxyphenyl)-1,1,1-trichloroethane · HPTEA b stract C ytochrome P450 (CYP) enzymes are involved in the metab-olism of endogenous and exogenous compounds. Human and rat liver microsomes were used to investigate the inhib-itory effects of methoxychlor (MXC) and its metabolite 2,2-bis( p -hydroxyphenyl)-1,1,1-trichloroethane (HPTE) on the activities of corresponding human and rat CYPs. Probe drugs were used to test the inhibitory effects of MXC and HPTE on human and rat CYPs. The results showed that MXC and HPTE inhibited both human CYP2C9 and rat liver CYP2C11 activity, with half-maximal inhibitory concentra-tion (IC 50 ) values of 15.47 ± 0.36 (MXC) and 8.87 ± 0.53 μmol/l (HPTE) for human CYP2C9, and of 22.45 ± 1.48 (MXC) and 24.63 ± 1.35 μmol/l (HPTE) for rat CYP2C11. MXC and HPTE had no effects on human CYP2C19 activity but inhibited rat CYP2C6 activity with IC 50 values of 14.84 ± 0.04 (MXC) and 8.72 ± 0.25 μmol/l (HPTE). With regard to human CYP2D6 and rat CYP2D2 activity, only HPTE potently inhibited human C YP2D6 activity, with an IC 50 value of 16.56 ± 0.69 μmol/l.Received: December 12, 2014A ccepted after revision: February 1, 2015P ublished online: March 31, 2015 G uo-Xin HuS chool of PharmacyW enzhou Medical UniversityC hashan District, Wenzhou, Zhejiang 325035 (China) E -Mail hgx @ w w /phaR .-S. Ge and G.-X. Hu contributed equally to this work. w n l o a d e d b y : n z h o u M e d i c a l C o l l e g e .107.222.187 - 5/27/2015 8:27:47 A MC hen/Pan/Wang/Chen/Dong/Ge/HuP harmacology 2015;95:145–153DOI: 10.1159/000380883146inhibition of these enzymes by environmental pollutantshas been investigated far less.O ne of the environmental pollutants is methoxychlor (MXC), an organochlorine pesticide. MXC replaced di-chlorodiphenyltrichloroethane (DDT) for wide use in ag-riculture, since DDT has a half-life of elimination (T 1/2)about 8 years and is easily absorbed in animal bodies[7].MXC is considered the preferred alternative to DDT for reasons of its much lower toxicity and shorter eliminationrate (T 1/2 is about 6 months)[8] . After ingestion, MXC is predominantly metabolized into 2,2-bis( p -hydro x y p he-nyl)-1,1,1-trichloroethane (HPTE)[9] . Both MXC and HPTE were characterized to be endocrine disruptors. A previous observation indicates that MXC is a reproduc-tive toxicant [10] . Interestingly, HPTE was found to exert a greater reproductive toxicity than the parent compoundMXC in both sexes[8, 9] . Further study indicated that both MXC and HPTE bind to human estrogen receptor-α, with the latter being more potent than the parent com-pound [11] . In addition, MXC and HPTE may have other toxicities, such as interfering with the metabolism of en-dogenous compounds or exogenous chemicals.O ur previous study has shown that MXC and HPTE suppressed the activities of some steroidogenic enzymes of both human and rat testes in vitro, including 3β-hydroxysteroid dehydrogenase (HSD), 17β-HSD3,and 11β-HSD[8, 12] . Recently, we have demonstrated that they are also potent inhibitors of 17α-hydrox-ylase/17,20-lyase (CYP17A1)[13] . However, sinceC YP17A1 belongs to the family of CYP enzymes, it is presently unclear whether both MXC and its metabolite HPTE effect direct inhibition of other CYP enzymes. In this study, we compared the potencies of MXC and HPTE in inhibiting the activities of four major types of CYP en-zyme in the human or rat liver.M aterials and Methods M ateri alsC ytochrome enzyme substrates and their metabolites includ-ing diclofenac and 4 ′ -hydroxydiclofenac for homologous CYP2C9 and CYP2C11, omeprazole and 5 ′ -hydroxy omeprazole for ho-mologous CYP2C19 and CYP2C6, dextromethorphan and dex-trorphan for homologous CYP2D6 and CYP2D2, midazolam and 1 ′ -hydroxy midazolam for homologous CYP3A4 and CYP3A1, as well as internal control diazepam, bisoprolol, and carbamazepine were all purchased from the National Institute for the Control of Pharmaceutical and Biological Products (Beijing, China). MXC and HPTE were purchased from Sigma (St. Louis, Mo., USA). All the other chemicals and solvents used were of analytical grade. MXC and HPTE were prepared using ethanol as a solvent.M ale Sprague-Dawley rats (250–300 g) were purchased from the Animal Center of Wenzhou Medical University. All animal procedures were carried out in strict accordance with the recom-mendations of the Guide for the Care and Use of Laboratory Ani-mals of Wenzhou Medical University. The protocol was approved by the Committee on the Ethics of Animal Experimentation of Wenzhou Medical University (permit No. 2014-0055). All surger-ies were performed under sodium pentobarbital anesthesia, and all efforts were made to minimize suffering. Human liver microsomes were purchased from Gentest (Woburn, Mass., USA).P reparation of Microsomal ProteinM icrosomal preparations of rat livers were made as described previously [8, 14] . Briefly, liver samples from rats were homoge-nized in cold 0.01 mmol/l PBS buffer containing 0.25 mmol/l of sucrose and centrifuged at 700 g for 30 min. The supernatants were transferred to new tubes and centrifuged at 10,000 g for an addi-tional 30 min. Then, the they were centrifuged at 105,000 g for 1 h twice. The pellets were resuspended with cold 0.01 mmol/l PBS, and protein concentrations were measured using the Bio-Rad Pro-tein Assay Kit according to the manufacturer’s protocol. The mi-crosomes were used for the measurement of CYP activity.E nzyme Inhibition Assays for CYPs in Human and Rat LiverT he inhibition assays for the respective human and rat enzyme pairs CYP2C9 and CYP2C11, CYP2C19 and CYP2C6, CYP2D6 and CYP2D2, and CYP3A4 and CYP3A1 were performed as de-scribed previously [15] . Preliminary experiments were performed to determine whether the metabolite formation rate was linear with the time of incubation and microsomal protein concentration, and the result showed that the metabolite formation rate was linear within 30 min. 285-μl incubation mixtures were prepared, contain-ing microsomes (0.25 mg/ml of liver microsomes), 100 mmol/l Tris-HCl buffer (pH 7.4), 10 μmol/l substrate, and different con-centrations of MXC or HPTE. After 5 min of preincubation at 37 °C , the abovementioned incubation mixture was initiated by ad-dition of 15 μl of 1 mmol/l NADPH. The substrate concentration was selected based on the K m of the substrate for human CYPs [16] .T o determine the effects of MXC or HPTE on CYP2C9 and CYP2C11, the inhibition assays were performed as follows. Diclo-fenac (10 μmol/l) was used as the common probe substrate for de-tecting human CYP2C9 or rat CYP2C11 activity. Varying concen-trations (0.1, 1.0, 5.0, 10, 25, 50, and 100 μmol/l) of MXC or HPTE were added to an incubation mixture to determine the half-maxi-mal inhibitory concentration (IC 50 ). The conversion rate of diclo-fenac (10 μmol/l) to 4 ′ -hydroxydiclofenac was measured to evalu-ate the inhibition in the presence of 100 μmol/l inhibitor (MXC or HPTE) in human and rat liver microsomes. When CYP2C9 and CYP2C11 were obviously inhibited by MXC or HPTE, the inhibi-tory modes of MXC and HPTE were further investigated.I n a separate study, different final concentrations of NADPH (20, 50, 100, and 200 μmol/l) were incubated together with the reac-tion mixture containing 0.25 mg/ml liver microsomes and 0–50μmol/l MXC or HPTE to determine whether MXC or HPTE com-petes with the cofactor NADPH. All reactions were terminated by adding 30 μl cold HCl (0.1 mol/l). Then, 50 μl diazepam (10 μg/ml) internal standard and 1 ml ethyl acetate were added. After mixing thoroughly for 2 min, the samples were centrifuged at 13,000 rpm for 5 min, and the organic layer was transferred to a clean glass tubeand dried under nitrogen. The organic phase was evaporated under w n l o a d e d b y : n z h o u M e d i c a l C o l l e g e .107.222.187 - 5/27/2015 8:27:47 A ME ffects of MXC and HPTE on Series CYPs P harmacology 2015;95:145–153DOI: 10.1159/000380883147a nitrogen stream at 40 °C . The residue was dissolved in 100 μl of mobile phase for measuring the conversion of diclofenac into 4 ′-hy-droxydiclofenac by HPLC, and 20 μl was injected into the HPLC system. The percent inhibitory activity of CYP2C9 or CYP2C11 was calculated depending on the percent conversion between the en-zyme substrate and its metabolite. The inhibition assays for ther espective human and rat enzyme pairs CYP2C19 and CYP2C6, CYP2D6 and CYP2D2, and CYP3A4 and CYP3A1 were performed as mentioned above with the corresponding common substrates omeprazole, dextromethorphan, and midazolam, respectively; the concentration of the corresponding substrate selected was based on the K m [17, 18] . A different internal standard was added to the assay mixture for determination of the activity of the above enzymes.D etermination of Substrates and Products of CYPs by HPLCT he HPLC system (Agilent 1100) was equipped with a quater-nary pump, on-line vacuum degasser, autosampler, thermostat-controlled column compartment, fluorescence detector, diodearray detector, and Agilent ChemStation Rev A.10.02 (Agilent, Santa Clara, Calif., USA). The Agilent ZORBAX SB-C18 column (4.6 × 150 mm, ID 5 μm) was used for the separation of substrates and their metabolites; the methods have been described previous-ly[15, 19–21] . Diazepam, midazolam, bisoprolol, and carbamaze-pine were used as an internal control for determination of diclofe-nac, omeprazole, dextromethorphan, and midazolam, respective-ly. The percent conversion of the substrate into its metabolite was used to calculate CYP activity.S tati sti csT he percent conversion of the probe substrate into its metabo-lite was calculated to render a value for CYP activity by HPLC. The positive control was chosen as 100% activity of the enzyme. The inhibition was repeated 5 times. The IC 50 value was calculatedu sing GraphPad (version 5; GraphPad Software Inc., San Diego, Calif., USA), and nonlinear regression of curve fit with one-site competition was performed. Lineweaver-Burk analysis was used to determine the mode of inhibition. The data were subjected to analysis by one-way ANOVA followed by Duncan’s multiple com-parisons to identify significant differences between groups when 3 and more groups were analyzed. All data are expressed as means ± SEM. Differences were regarded as significant at p < 0.05.R esults Effects of MXC and HPTE on CYP2C9 Activity in Human and on CYP2C11 Activity in Rat Liver Microsomes A s shown in f igure 1 , when the maximum concentra-tion of MXC or HPTE (100 μmol/l) was used, MXC and HPTE inhibited human CYP2C9 by 72.24 and 100%, re-spectively, while they inhibited rat CYP2C11 by 81.91 and 85.76%, respectively (p < 0.05). F urthermore, the IC 50 values for the inhibition of human CYP2C9 activity by MXC and HPTE were 15.47 ± 0.36 and 8.87 ± 0.53 μmol/l, while those for inhibition of rat CYP2C11 activ-ity by MXC and HPTE were 22.45 ± 1.48 and 24.63 ± 1.35 μmol/l, respectively ( t able 1 ). These data indicate that MXC and HPTE are inhibitors of CYP2C9 or CYP2C11, and that they inhibit human CYP2C9 more potently than rat CYP2C11.F ig. 1. Effects of MXC and HPTE on human CYP2C9 ( a ) and rat CYP2C11 activity ( b ) in liver microsomes. The activity was mea-sured by the conversion rate of diclofenac (10 μmol/l) to 4 ′-hy-droxydiclofenac in the presence of 100 μmol/l inhibitor (MXC or HPTE). The conversion rate was 21.87 ± 1.19% (%CON, MXC) and 0% (%CON, HPTE) for human liver microsomes and 20.99 ± 1.82% (%CON, MXC) and 14.70 ± 0.76% (%CON, HPTE) for rat liver microsomes. Means ± SEM (n = 5). * * *Significant difference compared with the control at p < 0.05. CON = Control.T ab le 1. I C 50 values (μmol/l) for human and rat CYPsMXC 15.47 ± 0.3622.45 ± 1.48ND 14.84 ± 0.04NDND HPTE8.87±0.5324.63±1.35ND8.72±0.2516.56±0.69NDV alues are means ± SEM (n = 5). ND = Not determined.w n l o a d e d b y : n z h o u M e d i c a l C o l l e g e .107.222.187 - 5/27/2015 8:27:47 A MC hen/Pan/Wang/Chen/Dong/Ge/HuP harmacology 2015;95:145–153DOI: 10.1159/000380883148M echanism Explaining the Effect of MXC and HPTE on Human CYP2C9 ActivityT he mechanisms of the inhibitory effect of MXC and HPTE on CYP2C9 activity were investigated. Lineweav-er-Burk plot analysis showed that both MXC and HPTEwere competitive inhibitors of human CYP2C9 activityagainst the substrate diclofenac ( f ig. 2 a , b). This indicatesthat they inhibit human CYP2C9 enzyme activity by competing with the substrate diclofenac at its enzyme-binding site. However, when NADPH was investigated, Lineweaver-Burk analysis showed that MXC and HPTE inhibited human CYP2C9 activity in an uncompetitive mode against the cofactor NADPH ( f ig. 2 c , d), indicating that both MXC and HPTE bind to the CYP2C9-NADPH-P450-reductase complex.E ffects of MXC and HPTE on CYP2C19 Activity in Human and on CYP2C6 Activity in Rat Liver MicrosomesO meprazole (10 μmol/l) was used as the common probe substrate for detecting human CYP2C19 or ratC YP2C6 activity. The conversion rate of omeprazole (10 μmol/l) to 5 ′ -hydroxy omeprazole was measured to eval-F ig. 2. Lineweaver-Burk plots of human CYP2C9 kinetics in the presence of MXC or HPTE. CYP2C9 activity was measured by the conversion rate of diclofenac (10 μmol/l) to 4 ′ -hydroxydiclofenac in human liver microsomes for 30 min in the pres-ence of varying concentrations of the sub-strate diclofenac or the cofactor NADPH. The experiments were repeated 5 times. a ,b Effects of MXC ( a ) and HPTE ( b )on human CYP2C9 activity against diclofe-nac. c ,d Effects of MXC ( c ) and HPTE ( d )on human CYP2C9 activity against NADPH. pro = Protein.F ig. 3. Effects of MXC and HPTE on human CYP2C19 ( a ) and ratCYP2C6 activity ( b ) in liver microsomes. The activity was mea-sured by the conversion rate of omeprazole (10 μmol/l) to 5 ′-hy-droxy omeprazole in the presence of 100 μmol/l inhibitor (MXC orHPTE). The conversion rate was 70.67 ± 0.06% (%CON, MXC) and 18.57 ± 0.07% (%CON, HPTE) for human liver microsomes and 4.09 ± 0.24% (%CON, MXC) and 1.16 ± 0.11% (%CON, HPTE) for rat liver microsomes. Means ± SEM (n = 5). * * *Significant differ-ence compared with the control at p < 0.05. CON = Control.w n l o a d e d b y : n z h o u M e d i c a l C o l l e g e .107.222.187 - 5/27/2015 8:27:47 A ME ffects of MXC and HPTE on Series CYPs P harmacology 2015;95:145–153DOI: 10.1159/000380883149uate the inhibition in the presence of 100 μmol/l inhibitor (MXC or HPTE) in human and rat liver microsomes. Both MXC and HPTE inhibited human CYP2C19 and rat CYP2C6 activity in the liver microsomes when compared to controls (p < 0.05). MXC showed 29.33 ± 0.06% inhibi-tion of the conversion of omeprazole to 5 ′-hydroxy omeprazole by CYP2C19 in human liver microsomes, while HPTE showed 81.43 ± 0.07% inhibition of the h uman enzyme activity ( f ig. 3 a ). MXC and HPTE also showed 95.91 ± 0.24 and 98.84 ± 0.11% inhibition of rat CYP2C6 activity compared to the control activity, respec-tively ( f ig. 3 b ). The IC 50 values for the inhibition of rat CYP2C6 activity were 14.84 ± 0.041 μmol/l for MXC and 8.72 ± 0.25 μmol/l for HPTE, respectively ( t able 1 ). These data indicate that MXC and HPTE induce a more potent inhibition of rat CYP2C6 than of human CYP2C19 activ-ity, and that HPTE is a more potent inhibitor of both hu-man CYP2C19 and rat CYP2C6 activity than its parent compound MXC.M echanism Explaining the Effect of MXC and HPTE on Rat CYP2C6 ActivityT he mechanisms of the inhibitory effect of MXC and HPTE on rat CYP2C6 activity were investigated. Line-weaver-Burk plot analysis showed that both MXC and HPTE were noncompetitive inhibitors of rat CYP2C6 a ctivity ( f ig. 4 a , b). However, when NADPH was used for the measurement of CYP2C6 activity in the rat liver mi-crosomes, Lineweaver-Burk analysis showed that MXC and HPTE were in a competitive mode ( f ig. 4 c , d). These results indicate that MXC and HPTE inhibited the C YP2C6 enzyme activity by competing with the substrate NADPH at its enzyme-binding site of P450 reductase.E ffects of MXC and HPTE on CYP2D6 Activity in Human and on CYP2D2 Activity in Rat Liver MicrosomesD extromethorphan (10 μmol/l) was used as the com-mon probe substrate for detecting human CYP2D6 or ratF ig. 4. Lineweaver-Burk plots of ratC YP2C6 kinetics in the presence of MXC or HPTE. CYP2C6 activity was measured by the conversion rate of omeprazole to 5 ′ -hydroxy omeprazole in rat liver micro-somes for 30 min in the presence of varying concentrations of the substrate omeprazole or the cofactor NADPH. The experiments were repeated 5 times. a ,b Effects of MXC ( a ) and HPTE ( b ) on rat CYP2C6 activity against omeprazole. c ,d Effects of MXC ( c )and HPTE ( d ) on human CYP2C6 activity against NADPH.pro =Protein.w n l o a d e d b y : n z h o u M e d i c a l C o l l e g e .107.222.187 - 5/27/2015 8:27:47 A MC hen/Pan/Wang/Chen/Dong/Ge/HuP harmacology 2015;95:145–153DOI: 10.1159/000380883150CYP2D2 activity. The conversion rate of dextrometho-rphan (10 μmol/l) to dextrorphan was measured to evalu-ate the inhibition in the presence of 100 μmol/l inhibitor (MXC or HPTE) in human and rat liver microsomes. MXC inhibited human CYP2D6 activity by 19.70 ± 8.29%, while HPTE inhibited the activity of this enzyme by 66.31 ± 1.05% compared to the control ( f ig. 5 a ). MXC inhibited rat CYP2D2 activity by 21.34 ± 2.45%, whereas HPTE in-hibited the enzyme activity by 57.18 ± 6.90% ( f ig. 5 b ). The IC 50 value for the inhibition of human CYP2D6 activity by HPTE was 16.56 ± 0.69 μmol/l ( t able 1 ). These data indicate that MXC induces weak inhibition of both hu-man CYP2D6 and rat CYP2D2 activity, and that HPTE is a more potent inhibitor of these enzymes than its parent compound MXC. Meanwhile, HPTE exhibited a more potent inhibition of human CYP2D6 than of rat CYP2D2 activity.M echanism Explaining the Effect of HPTE on Human CYP2D6 ActivityT he mechanisms of the inhibitory effect of HPTE on human CYP2D6 activity were investigated. Lineweaver-Burk plot analysis showed that HPTE was a mixed-type inhibitor of human CYP2D6 activity ( f ig. 6 a ). However, when NADPH was used for the measurement of human CYP2D6 activity, Lineweaver-Burk analysis showed thatHPTE was in an uncompetitive mode ( f ig. 6 b ). The resultsindicate that HPTE inhibits human CYP2D6 enzyme ac-tivity by competing with the substrate dextromethorphan at its enzyme-binding site.E ffects of MXC and HPTE on CYP3A4 Activity in Human and on CYP3A1 Activity in Rat Liver MicrosomesM idazolam (10 μmol/l) was used as the common probe substrate for detecting human CYP3A4 or rat CYP3A1 activity. The conversion rate of midazolam (10 μmol/l) to 1 ′ -hydroxy midazolam was measured to evaluate the in-hibition in the presence of 100 μmol/l inhibitor (MXC or HPTE) in human and rat liver microsomes. MXC and HPTE induced almost no inhibition of human CYP3A4 and rat CYP3A1 activity ( f ig. 7 ).F ig. 5. Effects of MXC and HPTE on human CYP2D6 ( a ) and ratCYP2D2 activity (b ) in liver microsomes. The activity was mea-sured by the conversion rate of dextromethorphan (10 μmol/l) to N-dextromethorphan in the presence of 100 μmol/l inhibitor (MXC or HPTE). The conversion rate was 81.3 ± 8.29% (%CON, MXC) and 33.69 ± 1.05% (%CON, HPTE) for human liver micro-somes and 78.66 ± 2.45% (%CON, MXC) and 42.82 ± 6.9% (%CON, HPTE) for rat liver microsomes. Means ± SEM (n = 5). * * * Significant difference compared with the control at p < 0.05. CON =Control.F ig. 6. Lineweaver-Burk plots of human CYP2D6 kinetics in the presence of HPTE. CYP2D6 activity was measured by the conver-sion rate of dextromethorphan (10 μmol/l) to N-dextromethor-phan in human liver microsomes for 30 min in the presence of varying concentrations of the inhibitor or the cofactor NADPH. The experiments were repeated 5 times. Effects of HPTE on humanCYP2D6 activity against dextromethorphan (a ) and NADPH (b ). w n l o a d e d b y : n z h o u M e d ic a l C o l l e g e .107.222.187 - 5/27/2015 8:27:47 A ME ffects of MXC and HPTE on Series CYPs P harmacology 2015;95:145–153DOI: 10.1159/000380883151D iscussion MXC is an organochlorine pesticide which is widely used in agriculture[22] . Thus, it may pose a health risk not only to agricultural workers but also to the general long-term-exposed population. In the present study, we investigated the dose-dependent inhibition of human liv-er CYP2C9, CYP2C19, CYP2D6, and CYP3A4 activity as well as of the homologous rat liver CYP2C11, CYP2C6, CYP2D2, and CYP3A1 activity by MXC and its metabo-lite HPTE.C YP2C9, CYP2C19, CYP2D6, and CYP3A4 are the main members of the CYP group of enzymes; they are localized to human liver microsomes and are homolo-gous with CYP2C11, CYP2C6, CYP2D2, and CYP3A1 in the rat liver, respectively [16, 23–25] . These enzymes play a critical role in the metabolism of a variety of endoge-nous and exogenous compounds, including steroids,drugs, and environmental pollutants[3, 25].Diclofenac,omeprazole, dextromethorphan, and midazolam were used as the common substrates of human CYP2C9 and rat CYP2C11, human CYP2C19 and rat CYP2C6, hu-man CYP2D6 and CYP2D2, and human CYP3A4 and CYP3A1, respectively. These substrates were used as the probe drugs for measuring the activity of the above CYPs by calculating the percent conversion of the substrates totheir metabolites[16, 23–25] .O ur results indicate that of these homologous CYPs, MXC and HPTE can completely inhibit human CYP2C9 and rat CYP2C11 activity, and that the inhibition of hu-man CYP2C9 activity was more potent than that of rat CYP2C11 activity. The same concentration of inhibitor (MXC and HPTE) also had an obvious inhibitory effect on both human CYP2C19 and rat CYP2C6 activity, but the inhibition was more sensitive to the rat than theh uman enzyme. However, for human CYP2D6 and rat CYP2D2 activity, MXC showed weak inhibition, but HPTE had a more obvious inhibitory effect on these en-zymes than the parent compound MXC. Different from the abovementioned CYPs, MXC and HPTE induced no inhibition of human CYP3A4 and rat CYP3A1 activity.O ur study also showed that MXC and HPTE were competitive inhibitors of human CYP2C9 and ratC YP2C11 activity, noncompetitive inhibitors of human CYP2C19 and rat CYP2C6 activity, and mixed inhibitors of human CYP2D6 and rat CYP2D2 activity against their respective substrates. We investigated whether MXC or HPTE competed with the cofactor NADPH. The present results indicate that MXC and HPTE were noncompeti-tive inhibitors of human CYP2C9 and rat CYP2C11 ac-tivity, competitive inhibitors of human CYP2C19 and rat CYP2C6 activity, and noncompetitive inhibitors of hu-man CYP2D6 and rat CYP2D2 activity against NADPH. Thus, it is reasonable to infer that MXC and HPTE show different modes of inhibition of these enzymes. MXC and HPTE also possibly interfere with the CYP2C9 (CYP2C11)-NADPH-P450 reductase complex. It sug-gests that the inhibition caused by MXC and HPTE may affect CYP catalysis of endogenous and exogenous com-pounds. Among CYPs, CYP2C9, CYP2C19, and CYP2D6 are responsible for the metabolism of 10, 5, and 30%, re-spectively, of the drugs metabolized by the cytochromeenzyme family in humans[26] . MXC and HPTE expo-sure can decrease the rate of hepatic biotransformation of the drugs, causing increased serum concentrations andtoxicity by inhibiting these CYP enzymes[27] . If there is a narrow therapeutic index for the drugs, the elevated se-rum levels may elicit unwanted toxicities. For example, CYP2C9 is responsible for the metabolism of ibuprofen and tolbutamide, among other drugs. Inhibition ofC YP2C9 by MXC and HPTE can cause an increasing hy-poglycemia risk and other adverse reactions. Drugs me-tabolized by CYP2C19 include omeprazole, diazepam, and lansoprazole. More than 80 drugs in the clinical set-ting are metabolized by CYP2D6, including several anti-psychotics and antidepressants; thus, long-time MXC or HPTE exposure is a risk factor for postural hypotensionF ig. 7. Effects of MXC and HPTE on human CYP3A4 ( a ) and rat CYP3A1 activity ( b ) in liver microsomes. The activity was mea-sured by the conversion rate of midazolam (10μmol/l)to 1′-hy-droxy midazolam in the presence of 100 μmol/l inhibitor (MXC or HPTE). The conversion rate was 96.17 ± 3.00% (%CON, MXC) and 97.82 ± 4.31% (%CON, HPTE) for human liver microsomes and 92.97 ± 5.22% (%CON, MXC) and 80.99 ± 1.65% (%CON, HPTE) for rat liver microsomes. Means ± SEM (n = 5). CON = Control.w n l o a d e d b y : n z h o u M e d i c a l C o l l e g e .107.222.187 - 5/27/2015 8:27:47 A MC hen/Pan/Wang/Chen/Dong/Ge/HuP harmacology 2015;95:145–153DOI: 10.1159/000380883152and extrapyramidal side effects [26] . MXC and HPTE in-hibit hepatic CYPs, reducing presystemic drug metabo-lism, resulting in an increase in oral bioavailability of the drug absorbed.B ecause CYP2C9 is mainly involved in the metabolism of estrogen [28] , the inhibition of CYP2C9 by MXC or its metabolite HPTE may be a factor contributing to the es-trogen-like effect of MXC and HPTE. When estrogen me-tabolism is reduced, the estrogen level is increased. This can cause many male reproductive diseases such as hypo-spadias, cryptorchidism, adult testiculoma, breast carci-noma, and carcinoma of the prostate[29] . In this study, we demonstrated that concentrations as low as 5 μmol/l of MXC or HPTE suppressed human CYP2C9 activity. This indicates that this inhibition may be involved in the reduced metabolism of estrogen. The level of MXC instream water can reach levels as high as 128 μg/l[30].Thus, these observations are relevant to public health, and such concentrations of MXC or HPTE have the po-tential to affect CYP2C9 activity. A heightened estrogen level in the human body may increase estrogen activity via binding to estrogen receptor activity. It is perhaps rea-sonable to infer that exposure to low concentrations of MXC and HPTE will increase the estrogen level. The re-sults presented herein identify additional mechanisms of action of MXC and HPTE, i.e. causing estrogen-like ac-tion, affecting normal endocrine function. An identifica-tion of the mechanisms of action of hormonally active agents present in the environment is required to facilitate the process of risk assessment in a population.C onclusion The present study demonstrates that MXC and HPTE inhibited human CYP2C9 and rat CYP2C11 as well as human CYP2C19 and rat CYP2C6 activity but had no ef-fects on human CYP3A4 and rat CYP3A1 activity. For CYP2D6 and CYP2D2, only HPTE potently inhibited the activity of this enzyme. MXC and HPTE showed com-petitive inhibition of human CYP2C9 and rat CYP2C11 activity against their substrate, but they displayed non-competitive inhibition of human CYP2C19 and ratC YP2C6 activity against their substrate. HPTE showed noncompetitive inhibition of human CYP2D6 and rat CYP2D2 activity against its substrate. MXC and HPTE are potent disruptors of endogenous and exogenous c ompound metabolism.D isclosure Statement T he authors report no conflicts of interest.R eferences 1 Gonzalez FJ, Lee YH: Constitutive expressionof hepatic cytochrome p450 genes. FASEB J 1996; 10:1112–1117. 2 Girennavar B, Jayaprakasha GK, Patil BS: Po-tent inhibition of human cytochrome P450 3A4, 2D6, and 2C9 isoenzymes by grapefruit juice and its furocoumarins. J Food Sci 2007; 72:C417–C421. 3 Belic A, Temesvari M, Kohalmy K, Vrzal R,Dvorak Z, Rozman D, Monostory K: Investi-gation of the CYP2C9 induction profile in hu-man hepatocytes by combining experimental and modelling approaches. Curr Drug Metab 2009; 10:1066–1074. 4 Ohkura T, Ohta K, Nagao T, Kusumoto K,Koeda A, Ueda T, Jomura T, Ikeya T, Ozeki E, Wada K, Naitoh K, Inoue Y, Takahashi N, Iwai H, Arakawa H, Ogihara T: Evaluation of human hepatocytes cultured by three-dimen-sional spheroid systems for drug metabolism. Drug Metab Pharmacokinet 2014; 29:373–378.5 Wang Z, Sun W, Huang CK, Wang L, Ia MM,Cui X, Hu GX, Wang ZS: Inhibitory effects of curcumin on activity of cytochrome P450 2C9 enzyme in human and 2C11 in rat liver mi c rosomes. Drug Dev Ind Pharm 2014, Epub ahead of print.6 Videau O, Pitarque S, Troncale S, Hery P,Thevenot E, Delaforge M, Benech H: Can a cocktail designed for phenotyping pharmaco-kinetics and metabolism enzymes in human be used efficiently in rat? Xenobiotica 2012; 42:349–354. 7 Kapoor IP, Metcalf RL, Nyström RF, SanghaGK: Comparative metabolism of methoxy-chlor, methiochlor, and DDT in mouse, in-sects, and in a model ecosystem. J Agric Food Chem 1970; 18:1145–1152. 8 Hu GX, Zhao B, Chu Y, Li XH, AkingbemiBT, Zheng ZQ, Ge RS: Effects of methoxy-chlor and 2,2-bis( p -hydroxyphenyl)-1,1,1-trichloroethane on 3β-hydroxysteroid dehy-drogenase and 17β-hydroxysteroid dehydro-genase-3 activities in human and rat testes. Int J Androl 2011; 34:138–144. 9 Akingbemi BT, Ge RS, Klinefelter GR, Gun-salus GL, Hardy MP: A metabolite of me-thoxychlor, 2,2-bis( p -hydroxyphenyl)-1,1,1-trichloroethane, reduces testosterone biosyn-thesis in rat Leydig cells through suppression of steady-state messenger ribonucleic acid levels of the cholesterol side-chain cleavage enzyme. Biol Reprod 2000; 62:571–578.10 Lafuente A, Marquez N, Pousada Y, Pazo D, Esquifino AI: Possible estrogenic and/or anti-androgenic effects of methoxychlor on pro-lactin release in male rats. Arch Toxicol 2000; 74:270–275.11 Paulose T, Hernandez-Ochoa I, Basavarajap-pa MS, Peretz J, Flaws JA: Increased sensitiv-ity of estrogen receptor alpha overexpressing antral follicles to methoxychlor and its me-tabolites. Toxicol Sci 2012; 120:447–459.12 Guo J, Deng H, Li H, Zhu Q, Zhao B, Chen B, Chu Y, Ge RS: Effects of methoxychlor and its metabolite 2,2-bis( p -hydroxyphenyl)-1,1,1-trichloroethane on 11β-hydroxysteroid dehy-drogenase activities in vitro. Toxicol Lett 2013; 218:18–23.w n l o a d e d b y : n z h o u M e d i c a l C o l l e g e .107.222.187 - 5/27/2015 8:27:47 A M。