15-Redox properties of cytochrome P450BM3 measured by direct methods

合集下载

nnk是烟草中特有的一种物质

nnk是烟草中特有的一种物质

nnk是烟草中特有的一种物质,可以诱导肺癌的发生。

卷烟燃烧可产生4000多种化学物质,其中40余种有明确的诱变/致癌性。

主要的致癌物有烟草特有亚硝胺(TSNA)、苯并(a)芘、多环芳烃(PAH),芳香胺、苯、二嗯英、儿茶酚及致癌的醌、肼类等。

TSNA是尼古丁被亚硝化的产物,4-(甲基亚硝氨基)-3-吡啶-1-丁酮(NNK)是已知7种TSNA中最强的致癌源。

NNK在主流、侧流烟气及不燃烧的烟草中均大量存在。

NNK是卷烟致癌的主要标志物,尼古丁是吸烟成瘾的主要原因。

Efficient Bioelectronic Actuation of the Natural Catalytic Pathway ofHuman Metabolic Cytochrome P450sSadagopan Krishnan,†Dhanuka Wasalathanthri,†Linlin Zhao,†John B. Schenkman,‡and James F. Rusling*,†,‡Herein, we describe fabrication of LbL films made by combining pure cyt P450s with CPR microsomes on electrodes to achieve a large ratio of cyt P450 to CPR (Figure 1), as in the human liver.1,2,40 Electrons are injected into the film from the electrode to accurately mimic the natural cyt P450 catalytic cycle at high catalytic turnover.We provide unambiguous evidence for electron transfer from electrode to CPR to cyt P450 from measured redox potentials, electron transfer rates, enzyme turnover rates, and carbon monoxide (CO) binding. Results suggest dynamic participation of a CPR-cyt P450 complex in a key equilibrium redox process facilitating efficient catalytic turnover of the excess cyt P450s. In addition, the electrode-driven turnover rate for a model oxidation reaction was as good as or better than when NADPH was utilized.这里我们主要叙述了层层膜结构通过将纯的P450酶和P450还原酶连接到电极上来实现像人体中从P450酶到P450还原酶一个较大差异,电子从电极中被送到膜中,准确模拟再高催化转化情况下的p450的催化过程。

昆虫抗药性相关细胞色素P450基因的表达调控机制

昆虫抗药性相关细胞色素P450基因的表达调控机制

http : ! www. insect. ora. cndol : 10.16380/j.kcxb.2021.01.0121 月 Januara2021, 64(1): 109 - 120昆虫学报ACTA ENTOMOLOGICA SINICA昆虫抗药性相关细胞色素P45Y 基因的表达调控机制朱江1>2,邱星辉1!(1.中国科学院动物研究所,农业虫害鼠害综合治理研究国家重点实验室,北京100101; 2-中国科学院大学,北京100049)摘要:杀虫剂的频繁持续使用,必然导致昆虫产生抗药性。

大量研究事例表明参与杀虫剂解毒的细胞色素P450(简称P450)过量表达是昆'对不同类型杀'剂产生抗性的重要原因,但目前人们对P450基因过量表达机制的认识还非常有限。

近十年来,随着生命科学与相关研究技术的发展, 有关昆'P450基因表达调控机制的研究取得了实质性的进展$本文综述了这一研究领域的重要发现。

除了基因重复或基因扩增导致的P450基因拷贝数增加外,P450基因在转录层面的上调表达是P450介导抗药性的普遍且重要的机制。

P450基因的转录上调由顺式调控元件与反式作用因 子相互作用得以实现。

现已发现了几种不同类型的转录因子(CncC , CREB 和核受体等)对昆' P450表达的直接调控,也鉴定了间接调控P450表达的作用因子如G 蛋白偶联受体及其下游效应 子。

CncC :Maf/Keap1是抗药性相关P450基因表达的重要而普遍的调控途径。

越来越多的事例表明小RNA 在昆'P450的表达调控中起重要作用。

现有的研究结果揭示了昆'P450基因调控因子和信号转导通路的多 及调控机制的 $关键词:杀'剂抗性;细胞色素P450;转录因子;调控通路;顺式调控元件;小RNA中图分类号:Q965.9文献标识码:A 文章编号:0454-6296(2021)01-0109-12Molecular mechanisms of expression regulation of insecS cytochrome P450 genes involveC in insecticine resistanccZHU Jian/1,2, QIU Xinv-Hui 1," (1. Staiv Key Laboratoc of Inteyrated Mana/emeni of Pvt Insects andRodents , InstituteoeZoooogy , ChineseAcademyoeSciences , Beieing100101, China ; 2. Uni eeasity o eChineseAcademyoeSciences , Bei eing 100049 , China )Abstraci : Tha frequent and extensiva usa of insecticidas leads to tha evolution of insecticiVa resistanco.Itiswe o aecogniyed thatoeeasetpae s ion o ecytoch aome P450s in eo oeed in insecticidedetotieication isone common and maeoamechanism oeaesistancetoeaaiouscoa s esoeinsecticidesin insects.Howeeea , themooecuoaamechanismseoaoeeaetpae s ion oeinsecticideaesistancesaeoated P450 genesin insectshaeenot been we o identieied untionow.In theaecentdecade , with thedeeeoopmentoebiooogicaoscienceandtechnooogy , manysignieicantadeanceshaeebeen achieeed in this diaection. In thisaaticoe , an oeeaeiew oethecu a entknowoedgewith aespecttoetpae s ion aeguoation oeinsectcytochaomeP450 geneswas paoeided.In addition togenedupoication and ampoieication , upsaeguoation oeP450satthetaanscaiptionaooeeeocan beascaibed totheinteaaction between the ts saeguoatoayeoementsand the ooans sactingeactoas.Taanscaiption eactoassuch asCncC , CREBand AhR diaectoyaeguoatetheetpae s ion oeP450s.TheGs paoteinscoupoed aeceptoas and theiadownstaeam e e ctoas poay indiaectaooes in the taanscaiptionao基金项目:国家自然科学基金项目(31672371)作者简介:朱江,女,1994年11月生,湖南衡阳人,博士研究生,研究方向为细胞色素P450的功能与调控,E-mail : zhujiang2016@ioe . -c . -n* 通讯作者 Corresponding author , E-mail : qiuxh@ ioz. -c - cn收稿日期 Received : 2020-07-S0;接受日期 Accepted : 2020-10-S9110昆虫学扌艮Acta Sipick64卷regulation of P450genes.The universality of the ala of CncC:Maf/Keap1V the expression regulation of P450s is suppo eted bystudieson aeaeietyofinsectspecies.Inceeasingeepoetsconfiemtheineoaeementof miceoRNAsin theeeguaation ofP450expee s ion.Cu e ntfindingseeeeaathedieeesityofeeguaatoeyfactoes and signal transduction pathways,and the complexity of underlying mechanisms V the regulation of P450 expee s ion.Key woJ'ds:Insecticide resistance;cy/chama P450;transciption fac/r;regulatoa pathway;cis-regulatoa element;mvaRNA人类为防治害虫,相继研发了有机氯、有机磷、氨基甲酸酯、拟除虫菊酯、新烟碱以及二酰胺类等杀虫药剂。

细胞色素P450酶生物转化及新催化反应的最新进展

细胞色素P450酶生物转化及新催化反应的最新进展

细胞色素P450酶生物转化及新催化反应的最新进展潘丽霞;朱婧;王青艳;杨登峰【摘要】Cytochrome P450 enzymes(P450s or CYPs)are a widespread large super family of mono-oxygenase that catalyzes the different substrates via heme in life beings.On the one hand,P450s involves in the key step of high-active natural product biosynthetic pathway,its biotransformation studies will help to increase the yield and activity of active molecules.On the other hand,its catalysis involves in metal elements and cofactors,new breakthroughs have been made in the protein modification and new catalytic reaction of P450s.In this article the latest advances in these two aspects of cytochrome P450 enzymes have been summarized and dis-cussed.%细胞色素P450 酶(P450s或CYPs)是一类广泛存在于生命体中,依赖于亚铁血红素催化多种底物的单加氧酶.一方面,它涉及许多高活性天然产物生物合成的关键步骤,对其生物转化研究有助于提高活性分子的产率和活性;另一方面,其催化涉及金属元素以及辅助因子,其蛋白质改造和新催化反应的研发也不断取得新突破.本文就细胞色素P450 酶这两方面的最近研究进展进行了总结与论述.【期刊名称】《广西科学》【年(卷),期】2018(025)003【总页数】4页(P258-261)【关键词】酶工程;细胞色素P450酶;生物转化;新催化反应【作者】潘丽霞;朱婧;王青艳;杨登峰【作者单位】广西科学院,非粮生物质酶解国家重点实验室,国家非粮生物质能源工程技术研究中心,广西生物质产业化工程院,广西生物炼制重点实验室,广西南宁530007;广西科学院,非粮生物质酶解国家重点实验室,国家非粮生物质能源工程技术研究中心,广西生物质产业化工程院,广西生物炼制重点实验室,广西南宁 530007;广西科学院,非粮生物质酶解国家重点实验室,国家非粮生物质能源工程技术研究中心,广西生物质产业化工程院,广西生物炼制重点实验室,广西南宁 530007;广西科学院,广西北部湾海洋研究中心,广西海洋天然产物与组合合成生物化学重点实验室,广西南宁 530007【正文语种】中文【中图分类】R978.10 引言细胞色素P450酶(P450s或者CYPs)是一类依赖于亚铁血红素,催化多种复杂氧化反应的多功能酶。

心血管药物相互作用与细胞色素P450酶关系的探讨

心血管药物相互作用与细胞色素P450酶关系的探讨

・进展述评・心血管药物相互作用与细胞色素P450酶关系的探讨万 超① 朱 宁①摘要:细胞色素P450(cytochrome P450,CYP450)酶系,是人体中与药物代谢密切相关的重要酶系之一,90%以上的药物的氧化是由P450酶催化的。

多药合用对机体P450酶的影响,能造成临床上显著的药物相互作用。

基于P450独特的功能和特点,可以预测、控制药物的相互作用,并可能解释某患者对特定治疗方案的反应。

该文从细胞色素P450分类及其各自的催化特点,心血管药物相互作用与细胞色素P450酶的关系方面进行探讨,为指导临床安全、合理的用药,增加患者依从性提供参考。

关键词:心血管药物,细胞色素P450,药物相互作用中图分类号:R54 文献标识码:A 文章编号:1002-0772(2007)12-0055-03R elationship betw een Cytochrome P450and Drug2drug I nteraction Among C ardiovascular Drugs W A N Chao,ZHU N ing.Car2 diovascular Depart ment of the Second A f f iliated Hospital of Dalian Medical U niversity,Dalian116027,ChinaAbstract:Cytochrome P450(CYP450)is one of the most important enzyme systems that involves in the metabolism of drugs in vi2 vo.There are about90%of drugs are oxidized by CYP450.The effects of multidrug on CYP450may induce notable clinical drug -drug interactions(DDI).Based on the unique function and character of Cytochrome P450,drug-drug interaction could be pre2 dicted and controlled.Furthermore,a patient’s response to specified therapeutic regimen could also be explained.In this article, researches on the classification and respectively catalytic characteristic of CYP450and drug-drug interaction among cardiovascular drugs induced by CYP450were investigated for safety and rationality of clinical medication and for the advancement of patients’compliance.K ey Words:cardiovascular drugs,cytochrome P450,drug-drug interaction 心血管药物是临床上常用的药物,由于药物种类的多样性和临床应用的长期性,使发生药物相互作用的可能性也随之增加。

细胞色素P450研究进展

细胞色素P450研究进展

1.1细胞色素P450研究进展1.1.1细胞色素P450细胞色素P450(cytochrome P450或CYP, 简称P450)是一个古老的以血红素为辅基的B族细胞色素蛋白酶基因超家族,广泛存在于细菌、真菌、植物以及动物等各种生物体内[1],通常与质体、线粒体、内质网、高尔基体等细胞器膜结合。

还原态P450与CO结合后在450nm处能检测到最大吸收峰,故命名为P450。

因其能使疏水性分子插入一个氧原子而变得更具有亲水性或者活性,因此又称之为单加氧酶(mixed-function oxidase, 简称MFO)[2]。

P450酶系作为自然界中生物催化剂,它所催化的反应类型多样,最典型的反应是把分子氧还原为水的同时,将其中一个氧原子转移至底物形成产物,催化反应为[3]:RH+O2+NADPH+H+ ROH+H2O+NADP+1958年,在大鼠肝微粒体中第一次发现P450。

D.S Frear于1969年首次在棉花(Gossypium hirsutum L.)中发现了它的存在[4]。

此后,大量的研究表明在拟南芥(Arabidopsis thaliana L.)[5]、小麦(Triticum aestivum L.)[6]、苜蓿(Medicago sativa L.)[7]、蓖麻(Ricinus communis L.)[8]等许多植物中也均有P450存在。

P450酶系在植物中参与多种代谢反应,发挥重要的催化作用。

[1] Omura T (1999). Forty years of cytochrome P450. BiochemBiophys Res Commun, 266 (3): 690~698.[2] Nelson D R, Kaymans L, Kamataki T, et al. P450superfamily:update on new sequence, gene mapping, accession numbers andnomenclature [J]. Pharmacogenetics, 1996, 6:1-42.[3] Ortiz de Montellano PR. Cytochrome P450: structure,mechanism, and biochemistry [M], 3rd ed. Kluwer Academic/Plenum Press, New York, 2005, 183-245.[4]Frear DS, Swanson HR, Tanaka FS. N-Demethylation ofsubstituted 3-(phenyl)-1-methylureas: isolation and characterization of a microsomal mixed function oxidase from cotton. Phytochemistry, 1969, 8(11): 2157–2169. [5] Paquette SM, Bak S, Feyereisen R. Intron-exon organizationand phylogeny in a large superfamily, the paralogous cytochrome P450 genes of Arabidopsis thaliana. DNA Cell Biol, 2000, 19(5): 307–317.[6] Murphy PJ, West CA. The role of mixed function oxidases inkaurene metabolism in Echinocystis macrocarpa Greene endosperm. Arch Biochem Biophys, 1969, 133(2): 395–407.[7] Li LY, Cheng H, Gai JY, Yu DY. Genome-wide identifycationand characterization of putative cytochrome P450 genes in the model legume Medicago truncatula. Planta, 2007, 226(1): 109–123.[8] Lew FL, West CA. (-)-kaur-16-en-7β-ol-19-oic acid, anintermediate in gibberellin biosynthesis. Phytochemistry, 1971, 10(9): 2065–2076.1.1.2细胞色素P450结构特征在细胞色素P450超基因家族中,不同成员之间在氨基酸序列上具有高度的变异性,但其空间结构上却保持较高的相似性,P450蛋白三级结构主要由C端的α-螺旋结构和N端的β-折叠结构组成[1,2]。

细胞色素P450酶系总活性荧光定量检测试剂盒产品说明书

细胞色素P450酶系总活性荧光定量检测试剂盒产品说明书

细胞色素P450酶系总活性荧光定量检测试剂盒产品说明书(中文版)要紧用途细胞色素P450酶系(CYP-ECOD)总活性荧光定量检测试剂是一种旨在通过乙氧基香豆素脱乙基酶反映系统中乙氧基香豆素转化为羟基香豆素后荧光峰值的转变,即采纳荧光法来测定样品中酶系活性的权威而经典的技术方式。

该技术通过精心研制、成功实验证明的。

其适用于各类细胞或组织裂解萃取液样品(动物、人体)或纯化微粒体样品细胞色素P450酶系的总活性检测。

产品严格无菌,即到即用,操作简捷,性能稳固。

技术背景细胞色素P450酶是肝细胞微粒体复合功能单加氧化酶系统的总称。

其分成五十多个亚酶:CYP1至CYP51。

作用在于体内外源化合物(xenobiotics),包括药物、致癌剂、化学污染物的氧化代谢,即单加氧化作用(monooxygenation)和羟化作用(hydroxylation)。

乙氧基香豆素脱乙基酶(7-ethoxycoumarinO-deethylase;ECOD)的活性是细胞色素P450酶系的诊断标记,其基于ECOD普遍性催化细胞色素P450亚酶的活性。

乙氧基香豆素(7-ethoxycoumarin)在乙氧基香豆素脱乙基酶的催化下,转化为羟基香豆素(7-hydroxycoumarin)后荧光峰值的转变(激发波长368nm,散发波长456nm),来定量测定细胞色素P450酶系的活性。

乙氧基香豆素脱乙基酶反映系统为:ECOD7-ethoxycoumarin + NADPH →7-hydroxycoumarin + CH3CHO + NADP+产品内容缓冲液(Reagent A) 5毫升反映液(Reagent B) 500微升底物液(Reagent C) 125微升终止液(Reagent D) 2毫升标准液(Reagent E) 100微升产品说明书 1份保留方式保留在-20℃冰箱里,反映液(Reagent B)、底物液(Reagent C)和标准液(Reagent E)幸免光照,终止液(Reagent D)具有侵蚀性,注意操作平安;有效保证6月用户自备毫升离心管:用于标准样品配制和反映的容器培育箱:用于孵育反映200微升1厘米光径比色皿或黑色96孔板:用于荧光分析的容器荧光分光光度仪过荧光酶标仪:用于荧光分析实验步骤实验开始前,将-20℃冰箱里的试剂置入冰槽里融化。

细胞色素p450超家族蛋白

细胞色素p450超家族蛋白全文共四篇示例,供读者参考第一篇示例:细胞色素P450超家族蛋白(Cytochrome P450 superfamily)是一类重要的酶蛋白家族,广泛存在于细胞内的内质网膜和线粒体膜上,参与了许多生物体内的代谢过程。

这一超家族包含了大约57个不同的基因家族,每个家族都有多个亚型,共计有数千种不同的细胞色素P450酶。

细胞色素P450超家族蛋白的命名来源于其光谱特性,其吸收波长为450纳米,同时也是一种铁含量丰富的蛋白质。

这种酶蛋白在细胞内的代谢过程中扮演了至关重要的角色,包括药物代谢、激素合成、胆固醇合成和维生素D代谢等。

细胞色素P450超家族蛋白在药物代谢中起着非常重要的作用。

人体内大约70%的药物是通过细胞色素P450酶代谢和排泄的。

这些酶可以将药物转化为更易排出体外的水溶性代谢物,同时也可以将药物转化为有毒代谢产物,导致药物不良反应。

细胞色素P450酶对于药物的疗效和毒性具有重要影响。

细胞色素P450超家族蛋白还参与了激素合成的过程。

胆固醇合成的关键酶就是细胞色素P450酶。

这一过程是肝脏中胆固醇的主要合成途径,胆固醇不仅是细胞膜的重要组成成分,还是合成类固醇激素和维生素D的前体物质。

细胞色素P450超家族蛋白还在维生素D代谢中发挥着关键作用。

维生素D是一个重要的营养素,参与了骨骼的形成和维持、钙磷代谢等多个生理过程。

细胞色素P450酶可以将维生素D转化为其活性形式,从而发挥生理功能。

细胞色素P450超家族蛋白在人体内的生理代谢中扮演着不可或缺的角色。

它们通过参与药物代谢、激素合成、胆固醇合成和维生素D代谢等多个重要生物过程,维持了身体内各种代谢平衡和功能活动。

对细胞色素P450超家族蛋白的研究不仅有助于理解人体内代谢的机制,还能为疾病的治疗和药物的开发提供重要的理论基础。

第二篇示例:细胞色素P450超家族蛋白是一类重要的酶蛋白,广泛存在于生物体内,包括植物、动物以及微生物等。

细胞色素P450研究进展

1.1细胞色素P450研究进展1.1.1细胞色素P450细胞色素P450(cytochrome P450或CYP, 简称P450)是一个古老的以血红素为辅基的B族细胞色素蛋白酶基因超家族,广泛存在于细菌、真菌、植物以及动物等各种生物体内[1],通常与质体、线粒体、内质网、高尔基体等细胞器膜结合。

还原态P450与CO结合后在450nm处能检测到最大吸收峰,故命名为P450。

因其能使疏水性分子插入一个氧原子而变得更具有亲水性或者活性,因此又称之为单加氧酶(mixed-function oxidase, 简称MFO)[2]。

P450酶系作为自然界中生物催化剂,它所催化的反应类型多样,最典型的反应是把分子氧还原为水的同时,将其中一个氧原子转移至底物形成产物,催化反应为[3]:RH+O2+NADPH+H+ ROH+H2O+NADP+1958年,在大鼠肝微粒体中第一次发现P450。

D.S Frear于1969年首次在棉花(Gossypium hirsutum L.)中发现了它的存在[4]。

此后,大量的研究表明在拟南芥(Arabidopsis thaliana L.)[5]、小麦(Triticum aestivum L.)[6]、苜蓿(Medicago sativa L.)[7]、蓖麻(Ricinus communis L.)[8]等许多植物中也均有P450存在。

P450酶系在植物中参与多种代谢反应,发挥重要的催化作用。

[1] Omura T (1999). Forty years of cytochrome P450. Biochem BiophysRes Commun, 266 (3): 690~698.[2] Nelson D R, Kaymans L, Kamataki T, et al. P450 superfamily:updateon new sequence, gene mapping, accession numbers andnomenclature [J]. Pharmacogenetics, 1996, 6:1-42.[3] Ortiz de Montellano PR. Cytochrome P450: structure, mechanism,and biochemistry [M], 3rd ed. Kluwer Academic/Plenum Press, New York, 2005, 183-245.[4]Frear DS, Swanson HR, Tanaka FS. N-Demethylation of substituted3-(phenyl)-1-methylureas: isolation and characterization of a microsomal mixed function oxidase from cotton. Phytochemistry, 1969, 8(11): 2157–2169.[5] Paquette SM, Bak S, Feyereisen R. Intron-exon organization andphylogeny in a large superfamily, the paralogous cytochrome P450 genes of Arabidopsis thaliana. DNA Cell Biol, 2000, 19(5): 307–317.[6] Murphy PJ, West CA. The role of mixed function oxidases in kaurenemetabolism in Echinocystis macrocarpa Greene endosperm. Arch Biochem Biophys, 1969, 133(2): 395–407.[7] Li LY, Cheng H, Gai JY, Yu DY. Genome-wide identifycation andcharacterization of putative cytochrome P450 genes in the model legume Medicago truncatula. Planta, 2007, 226(1): 109–123.[8] Lew FL, West CA. (-)-kaur-16-en-7β-ol-19-oic acid, an intermediatein gibberellin biosynthesis. Phytochemistry, 1971, 10(9): 2065–2076.1.1.2细胞色素P450结构特征在细胞色素P450超基因家族中,不同成员之间在氨基酸序列上具有高度的变异性,但其空间结构上却保持较高的相似性,P450蛋白三级结构主要由C端的α-螺旋结构和N端的β-折叠结构组成[1,2]。

色氨酸代谢研究进展

㊀㊀基金项目:国家自然科学基金资助项目(82074083);上海市宝山区科技创新专项基金资助项目(18-E -13)作者单位:201999㊀上海市宝山区中西医结合医院(苟小军㊁杨艳㊁杨晓露㊁曹姗);712046㊀西安,陕西中医药大学药学院(高珊珊)通讯作者:高珊珊,电子信箱:2392059931@;曹姗,电子信箱:caoshan -33@色氨酸代谢研究进展苟小军㊀杨㊀艳㊀杨晓露㊀曹㊀姗㊀高珊珊摘㊀要㊀色氨酸(tryptophan,Trp)是人体无法合成的必需氨基酸,对人体的新陈代谢至关重要,通过多种途径进行广泛的代谢,产生多种生物活性代谢产物,对生理过程产生重要影响㊂它在人体的多个器官例如脑㊁胃㊁肠道㊁肝脏㊁免疫细胞等具有重要的代谢作用,本文综述了历年来人们对Trp 的代谢生理作用的研究,阐述了Trp 在人体发挥的重要作用及近年来Trp 的研究进展㊂关键词㊀Trp㊀生理作用㊀代谢作用㊀研究进展中图分类号㊀R575.5㊀㊀㊀㊀文献标识码㊀A㊀㊀㊀㊀DOI ㊀10.11969/j.issn.1673-548X.2021.03.030㊀㊀Trp 也称为α-氨基-β-吲哚丙酸,自从1910年阐明其作用以来,受到了越来越多的关注㊂近几十年来,人们对Trp 进行了大量研究,国外不仅在Trp 的合成㊁营养㊁代谢及其生理生化特性方面进行了大量研究,而且在生产㊁医药㊁健康等方面也被广泛应用[1]㊂Trp 作为一种营养物质,是唯一通过非共价键与血清蛋白结合的氨基酸,这种结合与其分子结构有关[2]㊂Trp 是细胞激活和增殖所必需的氨基酸㊂Trp 沿犬尿氨酸(kynurenine,KYN)途径分解的代谢产物有3-羟基犬尿氨酸㊁邻氨基苯甲酸㊁3-HAA㊁哇琳酸(quinolinic acid,QA)㊁KYN 等㊂在这种特殊的微环境下,导致必需氨基酸的功能减弱,其分解可抑制T 细胞的增殖[3]㊂Trp 是一种必需的氨基酸,在蛋白质合成中起着重要作用,但其比例非常低(<1%)㊂此外,Trp 及其代谢产物在其他生物功能中也有重要作用,包括产生5-羟色胺(5-HT)㊁褪黑激素等生物活性分子㊂研究证明含有5-HT㊁褪黑激素㊁酪氨酸(tyrosine,KYNA)㊁烟酰胺腺嘌呤二核苷酸(nicotin-amide adenine dinucleotide,NAD)㊁烟酰胺腺嘌呤二核苷酸磷酸酯(nicotinamide adenine dinucleotide phos-phate,NADP)等的Trp 代谢物对正常代谢和器官功能至关重要[4]㊂一㊁Trp 的生理作用Trp 是一种芳香族氨基酸代谢物,在调节生长和摄食㊁情绪和行为以及免疫反应等方面具有广泛的生理功能,它是人体必需的氨基酸之一,人体不能合成,必须从日常饮食(植物和细菌)中获取[5]㊂Trp 代谢的KYNA 途径激活导致潜在的抑制性Trp 分解代谢产物的增加和Trp 合成5-HT 的可用性降低[6]㊂另有研究发现,与氨基酸生物合成相关的基因中至少有5%是Trp 基因,这也证明了Trp 在人体生理功能中的作用[7]㊂KYN 途径中的第1步和限速步骤是Trp吲哚环的氧化裂解,形成了正甲酰犬尿氨酸,后者会自发分解为KYN,由干扰素(interferon,IFN)-γ19诱导的吲哚胺2,3-双加氧酶-1(indoleamine -2,3-dioxygenase,IDO -1)催化该步骤,Trp 失调可能在谷氨酸(glutamate,Glu)系统中也具有极其重要的作用㊂Trp 途径中的关键代谢产物KYN 与精神分裂症有密切关系,Trp 可以通过两种途径进行代谢,即甲氧吲哚和犬尿氨酸酶(kynurenase,KYNs)途径[8]㊂Trp 沿甲氧吲哚途径进行代谢,形成了5-HT /血清素和5-甲氧基-N -乙酰色胺(褪黑素),KYN 途径是Trp 的主要降解代谢途径,是免疫应答的关键调节因子[9]㊂色氨酸羟化酶(tryptophan hydroxylase,TPH)亚型分为两种,即TPH1和TPH2,TPH1主要在胃肠道表达,TPH2仅在神经细胞表达,除大脑外,肠嗜铬细胞(en-terochromaffin cells,EC)和肥大细胞中也产生大量5-HT,并储存在血小板中[10]㊂Trp 衍生的尿毒症毒素具有促氧化㊁促炎性㊁促凝血作用以及促凋亡作用[11]㊂二㊁Trp 在人体器官中的代谢作用1.Trp 在肝中代谢:Trp 的生理功能有多个,其中一个重要的生理功能是用来合成蛋白质,除了合成蛋白质外,Trp 还是许多重要生理代谢产物的前体,饮㊃621㊃㊀㊃综述与进展㊃J Med Res,March 2021,Vol.50No.3㊀㊀食中有1%的Trp用于蛋白质合成,合成的蛋白质可以补偿降解的蛋白质㊂因此,大多数膳食Trp通过4条途径代谢,其中最重要的是肝氧化KP,大于Trp降解部分的95%以上,色氨酸2,3-双加氧酶(TDO)是该途径的第一个限速酶[12]㊂Trp氧化成N-甲酰犬尿氨酸(N-formylkynurenine,N-KYN)的主要途径存在于肝脏中,并由TDO催化㊂Trp分解代谢的肝途径依赖于磷酸吡哆醛(pyridoxal phosphate,PLP)的双功能酶犬尿氨酸酶(kynureninease,KYNU)和犬尿氨酸氨基转移酶(kynurenine aminotransferase,KAT),并受到维生素B6缺乏的影响[13],这两种酶都能通过复杂的代谢途径催化Trp转化为轧花碱㊁QA和轧花酸,通过TDO途径增加的Trp消耗增加了抑制T细胞反应并导致具有耐受性树突状细胞发育的KYNA的产生;缺乏Trp的饮食会增加血浆皮质酮水平,降低慢性应激大鼠中缝背侧和正中的血浆5-HT水平[14]㊂Trp通过转运体从肠腔转运到血液循环,Trp输送到肝脏,以及Trp参与肌肉和大脑的循环交换[15]㊂2.Trp在肠道参与的代谢作用:Trp作为某些微生物(如大肠杆菌和肺炎克雷伯菌)生长的氮源,是其重要的生理功能之一,因此,血清Trp水平的变化是肠道微生物群发酵的标志[5]㊂Trp及其内源性代谢产物是哺乳动物体内必需的营养物质,参与肠道免疫稳态和多种免疫疾病㊂血浆中Trp和Trp代谢物的浓度可以通过控制肠道微生物成分来调节[16]㊂在肠道和中枢神经系统中,细菌对Trp的代谢与5-HT 合成所必需的Trp之间保持平衡,肠道内的Trp和5-HT由宿主菌群直接和间接调节㊂肠道微生物群对Trp代谢和5-HT合成的间接调节主要通过KY-NA途径实现,如前所述,合成的Trp约占Trp代谢的90%,肠内细菌的Trp代谢产物I3P有助于抗生素和富含Trp饮食引起的体重增加的变化[17]㊂Trp不仅可以通过内源性合成的媒介物如KYNA和5-HT发挥生物学效应,还可以通过Trp衍生的肠道细菌产物,即吲哚类发挥生物学效应[18]㊂3.Trp在胃中代谢作用:Trp刺激血浆胆囊收缩素和幽门压力,减缓胃排空,胃排空调节餐后血糖㊂据报道,Trp可以减少能量的摄入,如胃内Trp对混合营养饮料的血糖反应㊁胃排空以及随后的能量摄入的影响[19]㊂5-HT作为Trp的前体,是一种单胺类激素和神经递质,是一种必需的胃肠调节剂,能调节肠道的生理,如蠕动和运动㊁分泌㊁营养吸收㊂5-HT在中枢神经系统和胃肠道中合成,由肠内分泌细胞分泌,其生物合成受TPH的两种亚型调节,其中TPH1主要定位于胃肠道内分泌细胞[20]㊂4.Trp在脑中代谢:研究表明,调节Trp代谢和血清素系统的KYN途径被认为在介导促炎性细胞因子对大脑的影响方面有重要作用㊂在炎性状态下,促炎性细胞因子上调了IDO的表达,从而激活了KYN的另一个代谢途径㊂在这种情况下,KYN很可能被代谢为喹啉酸(quinolinic acid,QA),QA是一种神经毒性代谢物[21]㊂L-Trp在扣带皮质的默认模式网络和双侧岛叶的显著性网络中具有更高的连接性㊂大脑区域调节食欲的活动受不同营养素的影响,L-Trp 可能是一种关键的氨基酸,增加了控制个体代谢状态区域的大脑连接性[22]㊂Trp缺乏和脑内Trp代谢不平衡在很大程度上与重度抑郁症有关,Trp是5-HT 和KYN的前体,转运体介导的血浆中必需氨基酸Trp 的摄取是脑内Trp代谢的决定因素㊂血浆Trp水平降低总是伴随着大脑Trp利用率和5-HT合成的不足,Trp-KYN和Trp-5-HT途径是抑郁症的主要Trp代谢途径,5%的Trp通过TPH2的5-HT途径代谢[23]㊂5.Trp在免疫细胞中的代谢作用:免疫系统的T 细胞通常识别并破坏异常细胞,包括癌组织和移植组织,这个过程需要氨基酸Trp㊂一个L-氨基酸转运蛋白(L-amino acid transporter,LAT1)交换Trp作为其KYNA降解产物,LAT1与IDO形成一个代谢小循环,将Trp饥饿与KYNA诱导的细胞死亡结合起来,提供相邻细胞双管齐下的失活,IDO1消耗Trp,同时产生Trp代谢产物(如KYNA),从而诱导免疫T淋巴细胞发生凋亡[24]㊂在免疫抑制酶IDO1介导下的Trp 耗竭和Trp代谢产物的积累,触发了免疫细胞的凋亡,炎症信号诱导的IDO1不仅通过Trp耗竭改变炎症过程,而且通过形成具有免疫调节作用的蛋白代谢产物,如KYNA和黄嘌呤酸通过限制免疫细胞产生IFNγ来减少炎症[25]㊂三、展㊀㊀望Trp作为人体必需氨基酸之一,人体本身不能合成,必须通过食物来获得㊂它在人体的情绪㊁免疫细胞反应㊁生理代谢以及氧化应激的炎性反应中具有重要作用㊂研究发现,IDO激活和刺激KYN途径发挥免疫功能可能是损害抗肿瘤免疫反应的调节机制[26]㊂Trp自身的代谢产物也会对人体的器官产生一定的影响,5-HT是调节肠道蠕动的重要介质㊂Trp在人体中具有不可替代的作用,对人体的器官以㊃721㊃㊀㊀医学研究杂志㊀2021年3月㊀第50卷㊀第3期㊃综述与进展㊃㊀及组织都具有重要的生理功能作用㊂综上所述,对于Trp的作用应进行深层次的研究以及验证,例如Trp 对抑郁症的治疗,Trp与肝硬化疾病的相关关系等都需要开展进一步的研究,为将来治疗肝硬化㊁抑郁症等疾病奠定基础㊂参考文献1㊀景寒松,徐淼,杨桂芹.色氨酸的来源㊁代谢途径及其在家禽生产上的应用[J].动物营养学报,2018,30(12):4813-4820 2㊀Stevens EA,Mezrich JD,Bradfield CA.The aryl hydrocarbon recep-tor:a perspective on potential roles in the immune system[J].Immu-nology,2009,127(3):299-3113㊀郑洪友,杨瑞利,苏光森.血清L-犬尿氨酸浓度作为R-CHOP方案治疗弥漫大B细胞淋巴瘤的预后因素[J].中国医药指南,2011,9(9):266-2684㊀KangX,Liu HN,Bai MM,et al.Redox properties of tryptophan metabolism and the concept of tryptophan use in pregnancy[J].Int J Mol Sci,2017,18(7):15955㊀Chen TL,Zheng XJ,Huang FJ,et al.Tryptophan predicts the risk for future type2diabetes[J].PLoS One,2016,11(9):e0162192 6㊀Neupane SP,Lien L,Martinez P,et al.The relationship of alcohol -use disorders and depressive symptoms to tryptophan metabolism: cross-sectional data from a Nepalese alcohol treatment sample[J]. Alcohol Clin Exp Res,2015,39(3):514-5217㊀Juliana K,Oded B,Jonathan CK,et al.The tryptophan pathway genes of the sargasso sea metagenome:new operon structures and the prevalence of non-operon organization[J].Genome Biol,2008,9 (1):R208㊀Yao JK,Reddy RD,Dougherty GG,et al.Altered interactions of tryptophan metabolites in first-episode neuroleptic-naive patients with schizophrenia[J].Mol Psychiatry,2010,15(9):938-953 9㊀Monic W,Elizabeth N,Sujatha K,et al.Maternal inflammation results in altered tryptophan metabolism in rabbit placenta and fetal brain[J].Dev Neurosci,2017,39(5):399-41210㊀Katsuhiko F.Etiological classification of depression based on the en-zymes of tryptophan metabolism[J].BMC Psychiatry,2014,14: 37211㊀Laetitia D,Claire C,Stephane P,et al.The aryl hydrocarbon re-ceptor-activating effect of uremic toxins from tryptophan metabolism: a new concept to understand cardiovascular complications of chronic kidney disease[J].Toxins(Basel),2014,6(3):934-949 12㊀Badawy AAB.Tryptophan metabolism,disposition and utilization in pregnancy[J].Biosci Rep,2015,35(5):1-1613㊀Luisa RA,Maria AR,Susan SP,et al.Metabolite profile analysis reveals functional effects of28-day vitamin B-6restriction on one-carbon metabolism and tryptophan catabolic pathways in healthy men and women[J].J Nutr,2013,143(11):1719-172714㊀Luana GB,Beatriz SB,Matheus JC,et al.Exogenous tryptophan promotes cutaneous wound healing of chronically stressed mice throughinhibition of TNF-αand IDO activation[J].PLoS One,2015,10 (6):1-1915㊀Rios-Avila L,Nijhout HF,Reed MC,et al.A mathematical mod-el of tryptophan metabolism via the kynurenine pathway provides in-sights into the effects of vitamin B-6deficiency,tryptophan loading, and induction of tryptophan2,3-dioxygenase on tryptophan metabo-lites[J].J Nutr,2013,143(9):1509-151916㊀Gao J,Xu K,Liu G,et al.Impact of the gut microbiota on intesti-nal immunity mediated by tryptophan metabolism[J].Front Cell In-fect Microbiol,2018,8:1317㊀Trisha A Jenkins,Jason CD Nguyen,Kate E Polglaze,et al.Influ-ence of tryptophan and serotonin on mood and cognition with a possible role of the gut-brain axis[J].Nutrients,2016,8(1):5618㊀Piotr K,Marek K,Marta GK,et al.Indole-3-propionicAcid,a tryptophan-derived bacterial metabolite,reduces weight gain in rats [J].Nutrients,2019,11(3):59119㊀Ullrich SS,Fitzgerald CE,Giesbertz P,et al.Effects of intragastric administration of tryptophan on the blood glucose response to a nutrient drink and energy intake,in lean and obese men[J].Nutrients, 2018,10(4):46320㊀Wang X,Zhang C,Zheng MY,et al.Metabolomics analysis of L-arginine induced gastrointestinal motility disorder in rats using UPLC-MS after magnolol treatment[J].Front Pharmacol,2019, 10:18321㊀Liu FH,Chen ZW,Zheng JJ,et al.Role of the indoleamine-2, 3-dioxygenase/kynurenine pathway of tryptophan metabolism in be-havioral alterations in a hepatic encephalopathy rat model?[J].J Neuroinflammation,2018,15(1):322㊀Claudia S,Stefan B,Katharina J,et al.Differential effects of L-tryptophan and L-leucine administration on brain resting state func-tional networks and plasma hormone levels[J].Sci Rep,2016, 1020(6):3572723㊀Li H,Xia X,Zhang JB,et al.Therapeutic duration and extent af-fect the effect of moxibustion on depression-like behaviour in rats via regulating the brain tryptophan transport and metabolism[J].Evid Based Complement Alternat Med,2019,2019(7592124):1-10 24㊀Thijs K,Loren LL,Hitomi T,et al.Nanosensor detection of an im-munoregulatory tryptophan influx/kynurenine efflux cycle[J].PLoS Biol,2007,5(10):e25725㊀Deac OM,Mills JJ,Gardiner CM,et al.Serum immune system bi-omarkers neopterin and interleukin-10are strongly related to trypto-phan metabolism in healthy young adults[J].J Nutr,2016,146 (9):1801-180626㊀Lukas L,Patricia K,Eva ME,et al.Inflammation-induced trypto-phan breakdown is related with anemia,fatigue,and depression in cancer[J].Front Immunol,2020,11(249):1-21(收稿日期:2020-09-07)(修回日期:2020-09-29)㊃821㊃㊀㊃综述与进展㊃J Med Res,March2021,Vol.50No.3㊀㊀。

细胞色素P450及其药物代谢研究

细胞色素P450及其药物代谢研究IntroductionHuman beings are exposed to countless types of chemicals, including drugs, environmental pollutants, and food additives, every day. These compounds have different chemical structures and pharmacological activities, and they must be metabolized and eliminated from the body to avoid toxicity. The cytochrome P450 (CYP) enzymes play a vital role in drug metabolism and the metabolism of other xenobiotics. This article focuses on the structure, function, and regulation of the CYP enzymes in drug metabolism.Structure of CYP ProteinsThe CYP proteins are a group of heme-containing monooxygenases that catalyze the oxidation of a vast array of endogenous and exogenous substrates. They are found in a wide range of organisms, including bacteria, fungi, plants, and animals, reflecting their evolutionarily conserved role as detoxification enzymes. In humans, there are 57 functional CYP genes and 18 pseudogenes identified to date, which are classified into 18 families based on their amino acid sequence homology.All CYP proteins share a conserved structural fold, consisting of a structural core of α-helices and a heme prosthetic group buried within the protein structure. The heme cofactors of CYP enzymes are involved in the catalytic cycle, in which an oxygen atom is inserted into the target molecule, resulting in hydroxylation or epoxidation, and the reduced heme (Fe2+) is regenerated by molecular oxygen binding. The CYP enzymes also require a source of electrons to reduce the ferric heme, which is supplied by the NADPH-dependent electron transport chain via the cytochrome P450 reductase (CPR).Function of CYP ProteinsThe CYP enzymes are involved in various physiological and pathological processes, including steroid biosynthesis, vitamin D metabolism, bile acid synthesis, and drug metabolism. In particular, the CYP3A subfamily is responsible for the metabolism ofmore than 50% of clinically used drugs, making it the most important drug-metabolizing enzyme family.Drug metabolism by CYP enzymes is a complex process that involves multiple enzymes and metabolic pathways. Generally, drugs are metabolized by Phase I and Phase II reactions. Phase I reactions convert lipophilic (fat-soluble) drugs into more polar (water-soluble) metabolites by introducing a hydroxyl group or other functional group. Phase II reactions conjugate the metabolite with another molecule, such as glucuronic acid or sulfate, to increase its water solubility and facilitate excretion.The CYP enzymes are responsible for the majority of Phase I drug metabolism, including oxidation, reduction, and hydrolysis. The oxidation reactions include N-, S-, and O-dealkylation, aromatic hydroxylation, and aliphatic hydroxylation. The reduction reactions involve nitro reduction and aromatic reduction. The hydrolysis reactions involve ester hydrolysis. The products of these reactions are often reactive or toxic intermediates, which can be further metabolized by Phase II enzymes. However, some drugs can cause drug-drug interactions by inhibiting or inducing the activity of CYP enzymes, leading to altered drug concentrations and pharmacological effects.Regulation of CYP ProteinsThe expression and activity of CYP enzymes are regulated by multiple factors, including genetic variation, environmental factors, and drug-drug interactions. The genetic variation can lead to interindividual differences in CYP enzyme activity and drug metabolism, which can affect the efficacy and safety of drug therapy. For example,CYP2D6 is subject to genetic polymorphisms that affect its expression and activity, resulting in poor metabolizers, intermediate metabolizers, or ultrarapid metabolizers, depending on the specific allele combination.Environmental factors, such as diet, smoking, and alcohol consumption, can also modulate the expression and activity of CYP enzymes. For instance, the induction of CYP1A2 by smoking can increase the metabolism of certain drugs, such as caffeine and theophylline, leading to reduced drug efficacy. On the other hand, the inhibition ofCYP3A4 by grapefruit juice can increase the bioavailability of some drugs, such as felodipine and simvastatin, leading to increased drug toxicity.Drug-drug interactions are common in clinical practice and can affect the pharmacokinetics and pharmacodynamics of co-administered drugs. Some drugs can act as inhibitors or inducers of CYP enzymes, leading to altered drug concentrations and therapeutic outcomes. For example, ketoconazole is a potent inhibitor of CYP3A4 and can increase the plasma concentrations of various drugs, such as cyclosporine and tacrolimus, leading to increased toxicity. Conversely, rifampicin is a potent inducer of CYP3A4 and can decrease the plasma concentrations of several drugs, such as nifedipine and oral contraceptives, leading to reduced efficacy.ConclusionThe CYP enzymes play a crucial role in the metabolism of xenobiotics, including drugs, and are subject to various genetic, environmental, and pharmacological factors that modulate their expression and activity. The understanding of CYP enzymes and their regulation is essential for the rational use of drugs and the management of drug interactions and toxicity. Further research is needed to elucidate the structure and function of CYP enzymes and their contributions to human physiology and pathophysiology.。

  1. 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. 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. 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)。

Redox properties of cytochrome P450BM3measured by direct methodsBarry D.Fleming1,Yanni Tian1,Stephen G.Bell1,Luet-Lok Wong1,Vlada Urlacher2and H.Allen O.Hill1 1Department of Chemistry,Inorganic Chemistry Laboratory,University of Oxford,Oxford,UK;2Institute of Technical Biochemistry, University of Stuttgart,Stuttgart,GermanyCytochrome P450BM3is a self-sufficient fatty acid mono-oxygenase consisting of a diflavin(FAD/FMN)reductase domain and a heme domain fused together in a single polypeptide chain.The multidomain structure makes it an ideal model system for studying the mechanism of electron transfer and for understanding P450systems in general. Here we report the redox properties of the cyto-chrome P450BM3wild-type holoenzyme,and its isolated FAD reductase and P450heme domains,when immobilized in a didodecyldimethylammonium bromidefilm cast on an edge-plane graphite electrode.The holoenzyme showed cyclic voltammetric peaks originating from both theflavin reductase domain and the Fe III/Fe II redox couple contained in the heme domain,with formal potentials of)0.388and )0.250V with respect to a saturated calomel electrode, respectively.When measured in buffer solutions containing the holoenzyme or FAD-reductase domain,the reductase response could be maintained for several hours as a result of protein reorganization and refreshing at the didodecyldi-methylammonium modified surface.When measured in buffer solution alone,the cyclic voltammetric peaks from the reductase domain rapidly diminished in favour of the heme response.Electron transfer from the electrode to the heme was measured directly and at a similarly fast rate (k s¢¼221s)1)to natural biological rates.The redox potential of the Fe III/Fe II couple increased when carbon monoxide was bound to the reduced heme,but when in the presence of substrate(s)no shift in potential was observed. The reduced heme rapidly catalysed the reduction of oxygen to hydrogen peroxide.Keywords:cytochrome P450BM3;redox properties;electro-chemistry.The cytochrome P450group of enzymes comprises a variety of heme-containing monooxygenases that are present in the majority of prokaryotic and eukaryotic organisms[1].The primary reaction catalysed by these enzymes is the hydroxylation of carbonÆhydrogen bonds. Substrate hydroxylation requires the activation of dioxy-gen.The two-electron reducing agent in biological systems is almost exclusively NAD(P)H,and this reductive activa-tion of oxygen by P450occurs in two separate one-electron additions.Thefirst electron reduces the substrate-bound ferric heme,facilitating the rapid binding of dioxygen and the formation of the ferrous–dioxygen intermediate.The second electron addition,followed by protonation,forms the ferric hydroperoxy complex.The O–O bond is cleaved, with one O inserted into the substrate and the other reduced to form water.Cytochrome P450BM3is a self-sufficient fatty acid mono-oxygenase found in Bacillus megaterium[2,3].The native function of wild-type P450BM3is to oxidize long-chain fatty acids,but it has also been shown to oxidize many other substrates[4,5].The119-kDa molecular mass holoenzyme has its diflavin(FAD/FMN)reductase domain and heme domain fused together in a single polypeptide chain,making the transfer of electrons highly efficient[6].The multi-domain structure has made it an ideal model system for studying the mechanism of electron transfer and for understanding P450systems in general[7–10].Independent expression of the two domains has permitted their study in isolation[11].Redox potentiometric studies have shown the electron flow for P450BM3to follow the path NADPHfiFADfiFMNfiheme[12].The electronflow to the heme centre in P450BM3was presumed to be regulated by a substrate-dependent increase(>100mV)in the redox potential of the heme,with theÔsuitabilityÕof the substrate for catalytic transformation being reflected in the magnitude of the increase in potential.Thermodynamic arguments recently presented by Honeychurch et al.[13],suggested that the binding of dioxygen to P450cam,another essential step in the P450catalytic process,would be sufficient to enable electron cycling,regardless of whether camphor is present.Research on the electrochemistry of enzymes is driven partly by the desire to understand the details of electron transport in proteins and partly by the great potential uses of enzymes in electrochemically based biosensors and bioreactors.Electron transfer between an electrode and protein was initially accomplished in the pioneering work of Eddowes&Hill[14]and Yeh&Kuwana[15].They showed that the problem of slow electron transfer betweenCorrespondence to B.D.Fleming,Department of Chemistry,Inorganic Chemistry Laboratory,University of Oxford,South Parks Road,Oxford OX13QR,UK.Fax:+441865272690,Tel.:+441865275902,E-mail:barry.fleming@Abbreviations:DDAB,didodecyldimethylammonium bromide;EPG,edge-plane pyrolytic graphite;SCE,saturated calomel electrode.Enzymes:cytochrome P450(CYP102,P450BM3)(EC1.14.14.1).(Received14July2003,revised19August2003,accepted21August2003)Eur.J.Biochem.270,4082–4088(2003)ÓFEBS2003doi:10.1046/j.1432-1033.2003.03799.xan electrode and a metalloprotein could be overcome by use of an electron shuttle or mediator.Since then,much effort has been directed into developing suitably mediated or modified electrode systems that facilitate biological electrochemistry.One such approach is the recently documented technique of using synthetic surfactant-based biomimetic membranes[16].These castfilms have been shown to be useful in attaining direct electrochemistry of heme-containing proteins[17–22].In the present work we employed this method to obtain the direct, quasi-reversible electrochemistry of the wild-type cyto-chrome P450BM3holoenzyme and its isolated FAD-reduc-tase and heme domains.Materials and methodsEnzymes and chemicalsWild-type cytochrome P450BM3was overexpressed in the Escherichia coli strain,DH5a(that contained the gene of the wild-type cytochrome P450BM3)and the bacterial growth and protein purification were carried out using published procedures[23].The FAD-reductase domain was cloned, expressed and purified according to published procedures [24].The P450BM3heme domain was cloned and expressed, using standard procedures,to encompass amino acid residues1–481of P450BM3.The P450BM3heme domain was purified by DEAE Sepharose and Source-Q anion-exchange chromatography,as described previously[23]. After purification,fractions with an A417/A280of>1were collected and stored.All samples were stored at)20°C in 40m M phosphate buffer,pH7.4,containing50%v/v glycerol.Glycerol was removed immediately prior to experiments by gelfiltration on a Amersham-Pharmacia PD-10column equilibrated with40m M phosphate buffer, pH7.4.Chemicals and solvents were of reagent grade and used without further purification unless stated otherwise.Dido-decyldimethylammonium bromide(DDAB)from Aldrich was prepared and used as a0.1-M stock solution in chloroform.Apparatus and proceduresDC cyclic voltammetry experiments were performed at room temperature in a standard two-compartment glass cell with a working volume of0.5mL.The working compart-ment housed the platinum gauze counter electrode in addition to the edge-plane pyrolytic graphite(EPG)work-ing electrode.A saturated calomel electrode(SCE)was used as a reference in a sidearm that connected to the working compartment via a Luggin capillary.All potentials were referred to the SCE.An Autolab potentiostat(Eco Chemie, Utrecht,the Netherlands)was used to record and control the potential of the working electrode.All measurements were made in40m M potassium phosphate buffer,pH7.4. Voltammograms were taken in solutions that had been deoxygenated by purified argon.For buffered protein solutions this was accomplished by blowing argon over the solution for several hours.An argon atmosphere was maintained over the solution during the experiment,unless stated otherwise.Preparation of P450/DDAB/EPG electrodesDDABfilms were made by spreading5l L of the stock solution onto a freshly polished EPG electrode.The chloroform was allowed to evaporate in air at room temperature for%1h.To incorporate the enzyme into the DDAB-modified electrode,the electrode was either placed into a solution of protein(%10l M)for1h at4°C, or directly into the electrochemical cell containing buffered protein solution at room temperature.ResultsA typical cyclic voltammogram,recorded for a P450BM3–DDAB-modified EPG electrode in oxygen-free phosphate buffer,pH7.4,is shown in Fig.1.Three cyclic voltammo-grams are shown:the initial scan soon after electrode immersion,and then scans after30and60min.This series of scans highlights the evolution of the response over a period of1h.Initially,the electrode response was domin-ated by a well-defined reversible couple centred at)0.388V, with a broader,less intense couple observed at)0.250V. With time the response at the more negative potential diminished,whilst the intensity of the other increased,until after1h the current response of the couples,relative to each other,remained stable(over longer periods of time both couples decreased at approximately the same rate).A significant response at)0.250V could still be observed for up to1week for electrodes maintained in buffer solution at 4°C.A different result was observed for voltammograms measured using a DDAB-modified EPG electrode in a buffered solution of the holoenzyme(Fig.2).The intensity of the couple at)0.388V could be maintained for longer time-periods(up to3h in this case),with a slight increase in the signal centred on)0.250V.When the electrode was removed from the enzyme solution and placed into enzyme-free buffer solution,the electrode response observedwas Fig.1.Cyclic voltammetry of wild-type P450BM3holoenzyme immobi-lized at a didodecyldimethylammonium bromide(DDAB)-modified edge-plane pyrolytic graphite(EPG)electrode.Cyclic voltammograms were recorded in deoxygenated phosphate-buffer solution(pH7.4),at a scan rate of0.1VÆs)1,for wild-type P450BM3holoenzyme immobilized at a DDAB-modified EPG electrode after being initially immersed (darkest line)and then30and60min(lightest line)later.The arrows indicate the direction of current change for the respective peaks over the duration of the experiment.ÓFEBS2003Redox properties of cytochrome P450BM3(Eur.J.Biochem.270)4083similar to that shown in Fig.1.If,after removing the electrode from the enzyme solution it was stored in buffer for any length of time,then the response was essentially dominated by the signal at )0.250V.To help identify the origins of the cyclic voltammetric peaks observed with the holoenzyme,the individual reduc-tase and heme domains were tested separately.Figure 3shows that when the FAD-reductase domain was immobi-lized at a DDAB-coated EPG electrode,quasi-reversible electrochemistry in buffer was possible.The mid-point potential of the reductase domain was )0.405V.As with the holoenzyme,the reductase domain response diminished quite rapidly over a period of 1h when in buffer solution,but could be maintained for a longer duration when in protein solution.A typical cyclic voltammogram recorded for the heme-domain DDAB-modified EPG electrode in buffer is shown in Fig.4.The response consisted of one Faradaic couple centred on )0.244V,probably from the Fe III /Fe II redoxcouple.This strong response could be maintained for many hours and for up to 1week when the electrode was maintained in buffer solution at 4°C.The cyclic voltam-metric peaks were approximately symmetrical,having equal areas under both the reductive and oxidative cycle,and showed a linear current dependence with scan rate from 0.01to 1V Æs )1,as expected for thin films of electroactive ing these peak areas,the concen-tration of the electroactive enzyme at the electrode surface was calculated to be of the order of nmoles Æcm )2.The peak separation was measured as a function of scan rate for the Fe III /Fe II redox couple in both the holoenzyme and the heme domain.The Ôtrumpet Õplots,so-formed in this case,are shown in Fig.5.Significant peak separation was only observed when the scan rate exceeded %10V Æs )1.From these data,a value for the apparent average electron-transfer rate constant (k s ¢)could be calculated.Generally,k s ¢values are obtained from the peak (E pa )to peak (E pc )potential separation values in cyclic voltammograms based on Laviron’s approach for diffusionless thin-layer voltam-metry [25].The k s ¢value for the Fe III /Fe II redox coupleinFig.2.Cyclic voltammetry of wild-type P450BM3holoenzyme in solu-tionusin g a DDAB-modified EPG elec trode.Cyclic voltammograms were recorded in a pH 7.4-buffered solution of wild-type P450BM3holoenzyme at a scan rate of 0.1V Æs )1for a DDAB-modified EPG electrode after being immersed for 1(darkest line),2or 3h (lightest line),respectively.The arrows indicate the direction of current change for the respective peaks over the duration of theexperiment.Fig.3.Cyclic voltammetry of the P450BM3FAD-reductase domain immobilized at a DDAB-modified EPG electrode.The cyclic vol-tammogram was recorded in deoxygenated phosphate-buffer solution,pH 7.4,at a scan rate of 0.1V Æs )1for the P450BM3reductase domain immobilized at a DDAB-modified EPGelectrode.Fig.4.Cyclic voltammetry of the P450BM3heme domainimmobilized at a DDAB-modified EPG electrode.The cyclic voltammogram was recorded in deoxygenated phosphate-buffer solution,pH 7.4,at a scan rate of 0.1V Æs )1for the P450BM3heme domain immobilized at a DDAB-modified EPGelectrode.Fig.5.‘Trumpet’plots for the heme-domainrespon se.The response from the P450BM3holoenzyme (j )and the heme domain (m )are shown.The reductive and oxidative peak potentials are plotted against the scan rate.This type of plot can be used to calculate the electron transfer rate constant.4084B.D.Fleming et al.(Eur.J.Biochem.270)ÓFEBS 2003the holoenzyme and heme domain were determined to be 138and 221s )1,respectively.The influence of pH on the heme redox potential,measured by cyclic voltammetry,is shown in Fig.6.These data are representative for the heme response from both the holoenzyme and isolated heme domain.The mid-point potential became increas-ingly more negative as the pH was increased from 3to 10.Two linear regions of different slopes were observed,one between pH 3and pH 8,and the other between pH 8and pH 10.The slope of these regions was )33and )126mV ÆpH unit )1,respectively.After bubbling the buffer solution with CO for 15min,the mid-point potential of the heme domain was positively shifted by 50mV (Fig.7).When purged with argon,the original formal potential returned.The effect of substrate binding on the redox potential of the heme domain was also investigated.When any of the substrates lauric acid,palmitic acid or octane were added to the buffer solution,there was typically no change in the cyclic oltammetric peaks.The heme redox couple was very sensitive to the presence of molecular oxygen.Figure 8shows the effect,on the cyclic voltammogram for the heme domain DDAB-modified EPG electrode,of adding 1,3or 5mL of air into the buffer solution.A new couple,at a potential slightly positive of the Fe III /Fe II couple,was observed.The reduction of O 2by Ôbare Õor DDAB-coated EPG electrodes occurred at more negative potentials ()0.5to )0.7V).Thus,the presence of the heme significantly lowers the overpotential required for O 2reduction.The magnitude of the reduction peak was related to the amount of O 2added.The oxidation peak height was less intense than the reduction peak,which is characteristic of a mechanism involving the rapid electrocatalytic reduction of O 2to H 2O 2by the reduced heme-containing films as per Eqn (1)and Eqn (2):P450Fe II þO 2!P450Fe II ÀO 2ð1ÞP450Fe II ÀO 2þ2H þþ2e !P450Fe II þH 2O 2ð2ÞDiscussionThe isolated FAD reductase and heme domains of P450BM3have been useful in identifying the voltammetric response observed with the P450BM3holoenzyme.It was clear that the Faradaic couples centred on )0.388and )0.250V had their origin in the reductase and heme components,respectively.The peak identification was aided by the fact that there was no significant shift in potential for the individual electroactive components compared with when they were fused together in the holoenzyme.A close similarity in the redox potential has also been measured by redox potentiometry for FAD and FMN in the isolated domains or the P450BM3holoenzyme [12].These solution measurements were explained in terms of there being no significant change in the domain environments whether isolated or fused together.The same could also be said here of the reductase and heme domains when they are incorporated in the DDAB film at the electrodesurface.Fig.6.Influence of pH on the heme redox potential.Cyclic vol-tammograms at 0.1V Æs )1for wild-type P450BM3holoenzyme were measured at different pH values.The mid-point potentials observed for each voltammogram were plotted against pH.A similar trend was observed for the isolated hemedomain.Fig.7.The effect of CO binding on the P450BM3heme redox potential.Cyclic voltammograms were recorded in deoxygenated phosphate-buffer solution (at pH 7.4),before and after bubbling with CO for 15min,at a scan rate of 0.1V Æs )1for the P450BM3heme domain immobilized at a didodecyldimethylammonium bromide (DDAB)-modified EPG electrode.Peaks shifted to the right in the presence ofCO.Fig.8.The effect of O 2binding on the electrochemistry of the P450BM3heme domain.Cyclic voltammograms were recorded in a phosphate-buffer solution,pH 7.4,after adding 0,1,3or 5mL of air,at a scan rate of 0.1V Æs )1for the P450BM3heme domain immobilized at a DDAB-modified EPG electrode.The height of the reduction peak increased with the amount of air injected into the solution.ÓFEBS 2003Redox properties of cytochrome P450BM3(Eur.J.Biochem.270)4085The change in the peak intensities for the holoenzyme, when measured in enzyme-free buffer solution,is of particular interest and several explanations are considered. First,some reorganization at the electrode/solution inter-face,presumably within the DDABfilm,had taken place. The initially large response from the reductase domain indicates that it is preferentially bound/oriented closest to the electrode surface.The further development of the electrode response,lowering of the reductase peak vs. increase of the heme peak,indicates that this orientation may be reversed,with the enzyme seemingly rotating to allow the heme domain to take up a more favourable electron transfer position near the electrode surface.It is also possible that the short-lived response in buffer is caused by denaturation at the electrode surface.This is suggested by the results observed with the isolated reductase domain. Its response,both in the presence of protein and in buffer only,was similar to that for the holoenzyme(except for the heme component).When the reductase domain is present in solution,it is plausible that the surface can be refreshed, effectively replacing the denatured protein.This could account for the longer duration of the large Faradaic currents observed.When the protein is not present in solution,denaturation takes place with noÔrefreshmentÕof the electrode surface and hence the relatively rapid decrease in current.With bare EPG electrodes,the direct electron transfer to P450BM3is slow and often not observed.The heme group is deep within the protein structure and favourable orientation at the electrode surface must occur to ensure electron transfer.When incorporated into the surfactant layer,the direct,rapid and quasi-reversible electron transfer between the P450heme and electrode was observed.The P450heme redox potential measured here(in both the holoenzyme and the heme domain only)is of a similar value to previous measurements for other P450enzymes and heme-containing proteins incorporated in DDABfilms[16,17,21,22].The broadness of the heme peak is considered to be caused by dispersion of E°values resulting from slight variations in protein orientation at the electrode surface[17].The differences in broadness between the reductive and oxidative waves,evident in Figs3and4,is typically the result of nonideality observed with thin-film systems[26].The potential of the heme Fe III/Fe II redox couple()0.252V) was much more positive than that measured in solution ()0.609V)[12].This type of behaviour has been reported for all cases where DDAB has been present as the biomimetic membrane.This is the result of interactions between the protein and surfactant and/or surfactant-related electrical double-layer effects on electrode potential. This effect was also evident,albeit to a lesser extent,with the reductase response.Our results show that the process of immobilization provides a very favourable environment for electron transfer to the heme to occur.The k s¢values calculated for the P450 heme domain(%200s)1)were similar in magnitude to the k s¢value measured for the natural electron transfer process–that between the FMN and heme for the P450BM3 holoenzyme in solution(223s)1with myristate)[7].These electron transfer results were dependent on the nature of the substrate–with the less favoured substrate lauric acid showing a lower k s¢value of130s)1.It has also been shown that it takes more energy to transfer an electron to the P450 heme when no substrate is bound[12].Our results are all the more interesting given that no substrate was present.Other k s¢values for heme-containing proteins,measured using electrode systems,are well below those reported here for the P450BM3heme domain.For example,P450cam in a DDAB film on a PG electrode was25s)1[17],whilst a variety of modified PG electrodes containing myoglobin showed k s¢values ranging from27to86s)1[27].The pH-dependent potential change observed in Fig.6 has been shown previously for other heme-containing proteins immobilized at electrode surfaces[18,19,22,27–30].However,in contrast to these previously published data, the slopes measured in this study for the linear regions were both quite different to the)59mVÆpH unit)1expected for a reversible one-electron transfer coupled to a single proton transfer.A similar low-slope region has also been reported for myoglobin and hemoglobin in polyacrylamidefilms,but this was for pH values of<5,a region where protein integrity might be questioned[27,28].Previous redox potentiometry experiments on P450BM3 showed that the presence of a suitable substrate results in an anodic(or positive)shift in redox potential in excess of 100mV[12].A similar substrate-dependent anodic shift was reported for P450cam from electrochemical data[31]. Our results indicate that no shift occurs in the formal potential of heme when in the presence of the substrates lauric acid,palmitic acid or octane.Similar results,based on cyclic voltammetric data,were reported recently for P450cam and P450cin in the presence of their natural substrates,camphor and cineole,respectively[22,32].The work with P450cin employed a similar procedure for enzyme immobilization as reported in this work[22]. Interestingly,redox potentiometric data for P450cin also showed no indication of a substrate-dependent anodic shift.There are reasonable thermodynamic arguments to suggest that substrate binding is not the only or main consideration in determining whether electron transfer to P450will occur[13].CO is known to rapidly bind specifically as a sixth ligand to the reduced heme iron of P450BM3[33].The fact that addition of CO to the buffer solution in our electrochemical experiments resulted in a peak shift of50mV confirmed that the observed response is from the heme domain. Similar results were obtained for P450cam immobilized at a DDAB-modified PG electrode[17]and a glassy carbon electrode modified with sodium montmorillonite[32].For catalytic reactions involving P450enzymes,the reduction of molecular oxygen to reactive oxygen species, such as H2O2,is typically an unwanted occurrence which dramatically reduces the efficiency of the desired catalytic process.As shown in Fig.8,once generated,the ferrous heme rapidly binds dioxygen,but unfortunately catalytic reduction to H2O2usually quickly follows.The real challenge then,in any development of electrode-based bioreactors designed to utilize the monooxygenase capabi-lities of P450,is getting the second electron to be used in peroxoiron complex formation and not in H2O2dissoci-ation.Several groups have recognized the difficulties associated with overcoming this problem and have attemp-ted to utilize mediator-promoted and H2O2-driven pathways to achieve their desired oxidation reactions4086B.D.Fleming et al.(Eur.J.Biochem.270)ÓFEBS2003[30,34,35].Given the fast electron transfer rates and low potentials necessary for thefirst electron reduction of the P450BM3heme domain in this surfactant-electrode ensem-ble,it follows that it should be the subject of ongoing study, as in our laboratory.However,it remains to be seen whether an effective electrochemically driven bioreactor,fully utili-zing Nature’s enzyme technology,can be achieved. ConclusionsWe determined the redox properties of cyto-chrome P450BM3by direct electrochemistry.The holo-enzyme response at a DDAB-modified EPG electrode was characterized by redox couples at)0.388V and)0.250V. These were identified as being direct electron transfer to the individualflavin reductase domain and P450heme domain, respectively.We have also shown that,although electron transfer in the biological system is from FAD/FMN to the heme,electron transfer can occur directly from the electrode to the heme under electrochemical conditions.The rate of this electron transfer is very rapid,of the order of the rates observed in the natural donor system.The redox potential of the heme did not appear to be affected by substrate binding,but it was possible that substrate inclusion in the surfactant layer may alter the surrounding charge environ-ment experienced by the protein.The reduction of mole-cular oxygen was readily catalysed by the P450BM3 heme domain immobilized at the DDAB-modified elec-trode.AcknowledgementsWe would like to thank the European Union for the collaborative grant.We also thank ECEnzymes,BASF and Dr T.Habicher. References1.OrtiZ de Montellano,P.R.(1995)Cytochrome P450:Structure,Mechanism and Biochemistry,2nd edn.Plenum Press,New York.2.Narhi,L.O.&Fulco,A.J.(1986)Characterization of a catalyti-cally self-sufficient119,000-dalton cytochrome P-450mono-oxygenase induced by barbiturates in Bacillus megaterium.J.Biol.Chem.261,7160–7169.3.Ravichandran,K.G.,Boddupalli,S.S.,Hasemann,C.A.,Peter-son,J.A.&Deisenhofer,J.(1993)Crystal structure of hemopro-tein domain of P450BM-3,a prototype for microsomal P450s.Science261,731–736.4.Guengerich,F.P.(2001)Common and uncommon cytochromeP450reactions related to metabolism and chemical toxicology.Chem.Res.Toxicol.14,611–650.5.Guengerich, F.P.(2002)Cytochrome P450enzymes in thegeneration of commercial products.Nat.Rev.Drug Discov.1, 359–366.6.Narhi,L.O.&Fulco,A.J.(1987)Identification and character-ization of two functional domains in cytochrome P-450BM-3,a catalytically self-sufficient monooxygenase induced by barbitu-rates in Bacillus megaterium.J.Biol.Chem.262,6683–6690.7.Munro, A.W.,Daff,S.,Coggins,J.R.,Lindsay,J.G.&Chapman,S.K.(1996)Probing electron transfer inflavocyto-chrome P-450BM3and its component domains.Eur.J.Biochem.239,403–409.8.Sevrioukova,I.F.,Hazzard,J.T.,Tollin,G.&Poulos,T.L.(1999)The FMN to heme electron transfer in cytochrome P450BM-3.J.Biol.Chem.274,36097–36106.9.Sevrioukova,I.F.,Li,H.,Zhang,H.,Peterson,J.A.&Poulos,T.L.(1999)Structure of a cytochrome P450-redox partner electron-transfer complex.Proc.Natl A96,1863–1868.10.Munro,A.W.,Leys,D.G.,McLean,K.J.,Marshall,K.R.,Ost,T.W.B.,Daff,S.,Miles, C.S.,Chapman,S.K.,Lysek, D.A., Moser,C.C.,Page,C.C.&Leslie Dutton,P.(2002)P450BM3:the very model of a modernflavocytochrome.Trends Biochem.Sci.27,250–257.es,J.S.,Munro,A.W.,Rospendowski,B.N.,Smith,W.E.,McKnights,J.&Thomson,A.J.(1992)Domains of the catalyti-cally self-sufficient cytochrome P-450BM-3.Genetic construction, overexpression,purification and spectroscopic characterisation.Biochem.J.288,503–509.12.Daff,S.N.,Chapman,S.K.,Turner,K.L.,Holt,R.A.,Govinda-raj,S.,Poulos,T.L.&Munro,A.W.(1997)Redox control of the catalytic cycle offlavocytochrome P-450BM3.Biochemistry36, 13816–13823.13.Honeychurch,M.J.,Hill,H.A.O.&Wong,L.-L.(1999)Thethermodynamics and kinetics of electron transfer in the cyto-chrome P450cam enzyme system.FEBS Lett.451,351–353. 14.Eddowes,M.J.&Hill,H.A.O.(1977)Novel method for theinvestigation of the electrochemistry of metalloproteins:cyto-chrome c.J.Chem.Soc.,mun.21,771–772.15.Yeh,P.&Kuwana,T.(1977)Reversible electrode reaction ofcytochrome c.Chem.Lett.10,1145–1148.16.Rusling,J.F.&Nassar,A.-E.F.(1993)Enhanced electron transferfor myoglobin in surfactantfilms on electrodes.J.Am.Chem.Soc.115,11891–11897.17.Zhang,Z.,Nassar,A.-E.F.,Lu,Z.,Schenkman,J.B.&Rusling,J.F.(1997)Direct electron injection from electrodes to cytochrome P450cam biomembrane-likefilms.J.Chem.Soc.,Faraday Trans.93,1769–1774.18.Rusling,J.F.(1998)Enzyme bioelectrochemistry in cast bio-membrame-likefilms.Acc.Chem.Res.31,363–369.19.Chen,X.,Hu,N.,Zeng,Y.,Rusling,J.F.&Yang,J.(1999)Ordered electrochemically activefilms of hemoglobin, didodecylmethylammonium ions,and ngmuir15,7022–7030.20.Boussaad,S.&Tao,N.J.(1999)Electron transfer and adsorptionof myoglobin on self-assembled surfactantfilms:an electro-chemical tapping-mode AFM study.J.Am.Chem.Soc.121, 4510–4515.21.Koo,L.S.,Immos,C.E.,Cohen,M.S.,Farmer,P.J.&OrtiZ deMontellano,P.R.(2002)Enhanced electron transfer and lauric acid hydroxylation by site-directed mutagenesis of CYP119.J.Am.Chem.Soc.124,5684–5691.22.Aguey-Zinsou,K.-F.,Bernhardt,P.V.,De Voss,J.J.&Slessor,K.E.(2003)Electrochemistry of P450cin:new insights into P450 electron mun.3,418–419.23.Carmichael,A.B.&Wong,L.-L.(2001)Protein engineering ofBacillus megaterium CYP102.The oxidation of polycyclic aromatic hydrocarbons.Eur.J.Biochem.268,3117–3125.24.Sevrioukova,I.,Truan,G.&Peterson,J.A.(1996)Theflavo-protein domain of P450BM-3:expression,purification,and properties of theflavin adenine dinucleotide-andflavin mono-nucleotide-binding subdomains.Biochemistry35,7528–7535. viron,E.(1979)General expression of the linear potential sweepvoltammogram in the case of diffusionless electrochemical sys-tems.J.Electroanal.Chem.101,19–28.26.Armstrong,F.,Heering,H.A.&Hirst,J.(1997)Reactions ofcomplex metalloproteins studied by protein-film voltammetry.Chem.Soc.Rev.26,169–179.27.Shen,L.,Huang,R.&Hu,N.(2002)Myoglobin in poly-acrylamide hydrogelfilms:direct electrochemistry and electro-chemical catalysis.Talanta56,1131–1139.ÓFEBS2003Redox properties of cytochrome P450BM3(Eur.J.Biochem.270)4087。

相关文档
最新文档