用户名: 密码: 验证码:
工业蛋白质构效关系的计算生物学解析
详细信息    查看全文 | 推荐本文 |
  • 英文篇名:Computational analysis of structure-activity relationship of industrial enzymes
  • 作者:陈琦 ; 李春秀 ; 郑高伟 ; 郁惠蕾 ; 许建和
  • 英文作者:Qi Chen;Chunxiu Li;Gaowei Zheng;Huilei Yu;Jianhe Xu;School of Biotechnology, East China University of Science and Technology;
  • 关键词:工业蛋白质 ; ; 结构-功能关系 ; 分子模拟 ; 理性设计
  • 英文关键词:industrial protein;;enzyme;;structure-activity relationship;;molecular simulation;;rational design
  • 中文刊名:生物工程学报
  • 英文刊名:Chinese Journal of Biotechnology
  • 机构:华东理工大学生物工程学院;
  • 出版日期:2019-10-25
  • 出版单位:生物工程学报
  • 年:2019
  • 期:10
  • 基金:上海市自然科学基金(No.19ZR1472900);; 国家自然科学基金(Nos.31971380,21536004,21672063,21776085)资助~~
  • 语种:中文;
  • 页:34-47
  • 页数:14
  • CN:11-1998/Q
  • ISSN:1000-3061
  • 分类号:TQ426.97
摘要
工业催化用酶已经成为现代生物制造技术的核心"芯片"。不断设计和研发新型高效的酶催化剂是发展工业生物技术的关键。工业催化剂创新设计的科学基础是对酶与底物的相互作用、结构与功能关系及其调控机制的深入剖析。随着生物信息学和智能计算技术的发展,可以通过计算的方法解析酶的催化反应机理,进而对其结构的特定区域进行理性重构,实现酶催化性能的定向设计与改造,促进其工业应用。聚焦工业酶结构-功能关系解析的计算模拟和理性设计,已成为工业酶高效创制改造不可或缺的关键技术。本文就各种计算方法和设计策略以及未来发展趋势进行简要介绍和讨论。
        Industrial enzymes have become the core "chip" for bio-manufacturing technology. Design and development of novel and efficient enzymes is the key to the development of industrial biotechnology. The scientific basis for the innovative design of industrial catalysts is an in-depth analysis of the structure-activity relationship between enzymes and substrates, as well as their regulatory mechanisms. With the development of bioinformatics and computational technology, the catalytic mechanism of the enzyme can be solved by various calculation methods. Subsequently, the specific regions of the structure can be rationally reconstructed to improve the catalytic performance, which will further promote the industrial application of the target enzyme. Computational simulation and rational design based on the analysis of the structure-activity relationship have become the crucial technology for the preparation of high-efficiency industrial enzymes. This review provides a brief introduction and discussion on various calculation methods and design strategies as well as future trends.
引文
[1] Privett HK, Kiss G, Lee TM, et al. Iterative approach to computational enzyme design. Proc Natl Acad Sci USA,2012, 109(10):3790–3795.
    [2] Verma R, Mitchell-Koch K. In silico studies of small molecule interactions with enzymes reveal aspects of catalytic function. Catalysts, 2017, 7(7):212.
    [3] van der Kamp MW, Mulholland AJ. Combined quantum mechanics/molecular mechanics(QM/MM)methods in computational enzymology. Biochemistry, 2013, 52(16):2708–2728.
    [4] Grajciar L, Heard CJ, Bondarenko AA, et al. Towards operando computational modeling in heterogeneous catalysis. Chem Soc Rev, 2018, 47(22):8307–8348.
    [5] Zhu CR, Gao DQ, Ding J, et al. TMD-based highly efficient electrocatalysts developed by combined computational and experimental approaches. Chem Soc Rev, 2018, 47(12):4332–4356.
    [6] Huang PS, Ban YEA, Richter F, et al. RosettaRemodel:a generalized framework for flexible backbone protein design. PLoS ONE, 2011, 6(8):e24109.
    [7] Addington TA, Mertz RW, Siegel JB, et al. Janus:prediction and ranking of mutations required for functional interconversion of enzymes. J Mol Biol, 2013,425(8):1378–1389.
    [8] Bendl J, Stourac J, Sebestova E, et al. HotSpot Wizard2.0:automated design of site-specific mutations and smart libraries in protein engineering. Nucleic Acids Res,2016, 44(W1):W479–W487.
    [9] Malisi C, Schumann M, Toussaint NC, et al. Binding pocket optimization by computational protein design.PLoS ONE, 2012, 7(12):e52505.
    [10] Chovancova E, Pavelka A, Benes P, et al. CAVER 3.0:a tool for the analysis of transport pathways in dynamic protein structures. PLoS Comput Biol, 2012, 8(10):e1002708.
    [11] Durrant JD, Votapka L, S?rensen J, et al. POVME 2.0:an enhanced tool for determining pocket shape and volume characteristics. J Chem Theory Comput, 2014,10(11):5047–5056.
    [12] Wijma HJ, Floor RJ, Bjelic S, et al. Enantioselective enzymes by computational design and in silico screening.Angew Chem Int Ed, 2015, 54(12):3726–3730.
    [13] Wijma HJ, Floor RJ, Jekel PA, et al. Computationally designed libraries for rapid enzyme stabilization. Protein Eng Des Sel, 2014, 27(2):49–58.
    [14] Park SB, Bae DW, Clavio NAB, et al. Structural and biochemical characterization of the curcumin-reducing activity of CurA from Vibrio vulnificus. J Agric Food Chem, 2018, 66(40):10608–10616.
    [15] Ren WW, Pengelly R, Farren-Dai M, et al. Revealing the mechanism for covalent inhibition of glycoside hydrolases by carbasugars at an atomic level. Nat Commun, 2018, 9(1):3243.
    [16] Fan FF, Chen NH, Wang YH, et al. QM/MM and MM MD simulations on the pyrimidine-specific nucleoside hydrolase:a comprehensive understanding of enzymatic hydrolysis of uridine. J Phys Chem B, 2018, 122(3):1121–1131.
    [17] Kulkarni YS, Liao QH, Petrovi?D, et al. Enzyme architecture:modeling the operation of a hydrophobic clamp in catalysis by triosephosphate isomerase. J Am Chem Soc, 2017, 139(30):10514–10525.
    [18] Schrepfer P, Buettner A, Goerner C, et al. Identification of amino acid networks governing catalysis in the closed complex of class I terpene synthases. Proc Natl Acad Sci USA, 2016, 113(8):E958–E967.
    [19] Mhashal AR, Vardi-Kilshtain A, Kohen A, et al. The role of the Met20 loop in the hydride transfer in Escherichia coli dihydrofolate reductase. J Biol Chem, 2017,292(34):14229–14239.
    [20] Ling BP, Li H, Yan LJ, et al. Conversion mechanism of enoyl thioesters into acyl thioesters catalyzed by2-enoyl-thioester reductases from Candida Tropicalis.Phys Chem Chem Phys, 2019, 21(19):10105–10113.
    [21] Driller R, Janke S, Fuchs M, et al. Towards a comprehensive understanding of the structural dynamics of a bacterial diterpene synthase during catalysis. Nat Commun, 2018, 9:3971.
    [22] Karuppiah V, Ranaghan KE, Leferink NGH, et al.Structural basis of catalysis in the bacterial monoterpene synthases linalool synthase and 1,8-Cineole synthase.ACS Catal, 2017, 7(9):6268–6282.
    [23] Haatveit KC, Garcia-Borràs M, Houk KN.Computational protocol to understand P450 mechanisms and design of efficient and selective biocatalysts. Front Chem, 2019, 6:663.
    [24] Faponle AS, Quesne MG, De Visser SP. Origin of the regioselective fatty-acid hydroxylation versus decarboxylation by a cytochrome P450 peroxygenase:what drives the reaction to biofuel production?Chemistry, 2016, 22(16):5478–5483.
    [25] Zhou HY, Wang BJ, Wang F, et al. Chemo-and regioselective dihydroxylation of benzene to hydroquinone enabled by engineered cytochrome P450monooxygenase. Angew Chem Int Ed, 2019, 58(3):764–768.
    [26]?widerek K, Tu?ón I, Williams IH, et al. Insights on the origin of catalysis on glycine N-methyltransferase from computational modeling. J Am Chem Soc, 2018,140(12):4327–4334.
    [27] Marques SM, Dunajova Z, Prokop Z, et al. Catalytic cycle of haloalkane dehalogenases toward unnatural substrates explored by computational modeling. J Chem Inf Model, 2017, 57(8):1970–1989.
    [28] Wang BJ, Cao ZX, Rovira C, et al. Fenton-derived OH radicals enable the MPnS enzyme to convert2-hydroxyethylphosphonate to methylphosphonate:insights from Ab initio QM/MM MD simulations. J Am Chem Soc, 2019, 141(23):9284–9291.
    [29] Sheng X, Plasch K, Payer SE, et al. Reaction mechanism and substrate specificity of Iso-orotate decarboxylase:a combined theoretical and experimental study. Front Chem, 2018, 6:608.
    [30] Behiry EM, Ruiz-Pernia JJ, Luk L, et al. Isotope substitution of promiscuous alcohol dehydrogenase reveals the origin of substrate preference in the transition state. Angew Chem Int Ed, 2018, 57(12):3128–3131.
    [31] Liu J, Wu SK, Li Z. Recent advances in enzymatic oxidation of alcohols. Curr Opin Chem Biol, 2018, 43:77–86.
    [32] Kong XD, Yuan SG, Li L, et al. Engineering of an epoxide hydrolase for efficient bioresolution of bulky pharmaco substrates. Proc Natl Acad Sci USA, 2014,111(44):15717–15722.
    [33] Herter S, Medina F, Wagschal S, et al. Mapping the substrate scope of monoamine oxidase(MAO-N)as a synthetic tool for the enantioselective synthesis of chiral amines. Bioorg Med Chem, 2018, 26(7):1338–1346.
    [34] Li GY, Yao PY, Gong R, et al. Simultaneous engineering of an enzyme’s entrance tunnel and active site:the case of monoamine oxidase MAO-N. Chem Sci, 2017, 8(5):4093–4099.
    [35] Collazo L, Klinman JP. Control of the position of oxygen delivery in soybean lipoxygenase-1 by amino acid side chains within a gas migration channel. J Biol Chem, 2016, 291(17):9052–9059.
    [36] Han SW, Kim J, Cho HS, et al. Active site engineering ofω-transaminase guided by docking orientation analysis and virtual activity screening. ACS Catal, 2017,7(6):3752–3762.
    [37] Mader SL, Br?uer A, Groll M, et al. Catalytic mechanism and molecular engineering of quinolone biosynthesis in dioxygenase AsqJ. Nat Commun, 2018,9(1):1168.
    [38] Dijkman WP, Binda C, Fraaije MW, et al. Structurebased enzyme tailoring of 5-hydroxymethylfurfural oxidase. ACS Catal, 2015, 5(3):1833–1839.
    [39] Ferrari AR, Lee M, Fraaije MW. Expanding the substrate scope of chitooligosaccharide oxidase from Fusarium graminearum by structure-inspired mutagenesis.Biotechnol Bioeng, 2015, 112(6):1074–1080.
    [40] Zheng GW, Liu YY, Chen Q, et al. Preparation of structurally diverse chiral alcohols by engineering ketoreductase CgKR1. ACS Catal, 2017, 7(10):7174–7181.
    [41] Chen Q, Luan ZJ, Cheng XL, et al. Molecular dynamics investigation of the substrate binding mechanism in carboxylesterase. Biochemistry, 2015, 54(9):1841–1848.
    [42] Chen FF, Zheng GW, Liu L, et al. Reshaping the active pocket of amine dehydrogenases for asymmetric synthesis of bulky aliphatic amines. ACS Catal, 2018,8(3):2622–2628.
    [43] Maria-Solano MA, Serrano-Hervás E, Romero-Rivera A,et al. Role of conformational dynamics in the evolution of novel enzyme function. Chem Commun(Camb),2018, 54(50):6622–6634.
    [44] Romero-Rivera A, Garcia-Borràs M, Osuna S.Computational tools for the evaluation of laboratory-engineered biocatalysts. Chem Commun(Camb), 2016, 53(2):284–297.
    [45] Wijma HJ, Marrink SJ, Janssen DB. Computationally efficient and accurate enantioselectivity modeling by clusters of molecular dynamics simulations. J Chem Inf Model, 2014, 54(7):2079–2092.
    [46] Maria-Solano MA, Romero-Rivera A, Osuna S.Exploring the reversal of enantioselectivity on a zinc-dependent alcohol dehydrogenase. Org Biomol Chem, 2017, 15(19):4122–4129.
    [47] Serapian SA, van der Kamp MW. Unpicking the cause of stereoselectivity in actinorhodin ketoreductase variants with atomistic simulations. ACS Catal, 2019,9(3):2381–2394.
    [48] Sun ZT, Salas PT, Siirola E, et al. Exploring productive sequence space in directed evolution using binary patterning versus conventional mutagenesis strategies.Bioresour Bioprocess, 2016, 3:44.
    [49] Sun ZT, Wu L, Bocola M, et al. Structural and computational insight into the catalytic mechanism of limonene epoxide hydrolase mutants in stereoselective transformations. J Am Chem Soc, 2018, 140(1):310–318.
    [50] Yang B, Wang HJ, Song W, et al. Engineering of the conformational dynamics of lipase to increase enantioselectivity. ACS Catal, 2017, 7(11):7593–7599.
    [51] Li FL, Kong XD, Chen Q, et al. Regioselectivity engineering of epoxide hydrolase:near-perfect enantioconvergence through a single site mutation. ACS Catal, 2018, 8(9):8314–8317.
    [52] Reetz MT, Soni P, Acevedo JP, et al. Creation of an amino acid network of structurally coupled residues in the directed evolution of a thermostable enzyme. Angew Chem Int Ed Engl, 2009, 48(44):8268–8272.
    [53] Blum JK, Ricketts MD, Bommarius AS. Improved thermostability of AEH by combining B-FIT analysis and structure-guided consensus method. J Biotechnol,2012, 160(3/4):214–221.
    [54] Gong XM, Qin Z, Li FL, et al. Development of an engineered ketoreductase with simultaneously improved thermostability and activity for making a bulky Atorvastatin precursor. ACS Catal, 2019, 9(1):147–153.
    [55] Hao JJ, Berry A. A thermostable variant of fructose bisphosphate aldolase constructed by directed evolution also shows increased stability in organic solvents.Protein Eng Des Sel, 2004, 17(9):689–697.
    [56] Reetz MT, Soni P, Fernández L, et al. Increasing the stability of an enzyme toward hostile organic solvents by directed evolution based on iterative saturation mutagenesis using the B-FIT method. Chem Commun,2010, 46(45):8657–8658.
    [57] Vazquez-Figueroa E, Yeh V, Broering JM, et al.Thermostable variants constructed via the structure-guided consensus method also show increased stability in salts solutions and homogeneous aqueous-organic media. Protein Eng Des Sel, 2008,21(11):673–680.
    [58] Bommarius AS. Biocatalysis:a status report. Annu Rev Chem Biomol Eng, 2015, 6:319–345.
    [59] Jiang W, Yang RN, Lin P, et al. Bioinspired genetic engineering of supramolecular assembled formate dehydrogenase with enhanced biocatalysis activities. J Biotechnol, 2019, 292:50–56.
    [60] van der Kamp MW, Prentice EJ, Kraakman KL, et al.Dynamical origins of heat capacity changes in enzyme-catalysed reactions. Nat Commun, 2018, 9:1177.
    [61] Thompson MP, Turner NJ. Two-enzyme hydrogen-borrowing amination of alcohols enabled by a cofactor-switched alcohol dehydrogenase. ChemCatChem,2017, 9(20):3833–3836.
    [62] Cahn JKB, Werlang CA, Baumschlager A, et al. A general tool for engineering the NAD/NADP cofactor preference of oxidoreductases. ACS Synth Biol, 2017,6(2):326–333.
    [63] You ZN, Chen Q, Shi SC, et al. Switching cofactor dependence of 7β-Hydroxysteroid dehydrogenase for cost-effective production of ursodeoxycholic acid. ACS Catal, 2019, 9(1):466–473.
    [64] Tomi?S, Koji?-Prodi?BA. Quantitative model for predicting enzyme enantioselectivity:application to Burkholderia cepacia lipase and 3-(aryloxy)-1,2-propanediol derivatives. J Mol Graph Model, 2001,21(3):241–252.
    [65] Paier J, Stockner T, Steinreiber A, et al.Enantioselectivity of epoxide hydrolase catalysed oxirane ring opening:a 3D QSAR study. J Comput Aided Mol Des, 2003, 17(1):1–11.
    [66] Braiuca P, Boscarol L, Ebert C, et al. 3D-QSAR applied to the quantitative prediction of penicillin G amidase selectivity. Adv Synth Catal, 2006, 348(6):773–780.
    [67] Gu JL, Liu J, Yu HW. Quantitative prediction of enantioselectivity of Candida antarctica lipase B by combining docking simulations and quantitative structure-activity relationship(QSAR)analysis. J Mol Catal B:Enzym, 2011, 72(3/4):238–247.
    [68] Cheng Q, Chen Q, Xu JH, et al. A 3D-QSAR assisted activity prediction strategy for expanding substrate spectra of an aldehyde ketone reductase. Mol Catalys,2018, 455:224–232.
    [69] Gao N, Liang T, Yuan Y, et al. Exploring the mechanism of F282L mutation-caused constitutive activity of GPCR by a computational study. Phys Chem Chem Phys, 2016,18(42):29412–29422.
    [70] Romero-Rivera A, Garcia-Borràs M, Osuna S. Role of conformational dynamics in the evolution of retro-aldolase activity. ACS Catal, 2017, 7(12):8524–8532.
    [71] Chen Q, Yu HL, Cheng XL, et al. Structural investigation of the enantioselectivity and thermostability mechanisms of esterase Rh Est1. J Mol Graph Model, 2018, 85:182–189.
    [72] Lian P, Yuan CM, Xu Q, et al. Thermostability mechanism for the hyperthermophilicity of extremophile cellulase Tm Cel12A:Implied from molecular dynamics simulation. J Phys Chem B, 2016, 120(30):7346–7352.
    [73] Yang KK, Wu Z, Arnold FH. Machine-learning-guided directed evolution for protein engineering. Nat Methods,2019, 16(8):687–694.
    [74] Saito Y, Oikawa M, Nakazawa H, et al.Machine-learning-guided mutagenesis for directed evolution of fluorescent proteins. ACS Synth Biol, 2018,7(9):2014–2022.
    [75] Wu Z, Kan SBJ, Lewis RD, et al. Machine learning-assisted directed protein evolution with combinatorial libraries. Proc Natl Acad Sci USA, 2019,116(18):8852–8858.
    [76] Bonk BM, Weis JW, Tidor B. Machine learning identifies chemical characteristics that promote enzyme catalysis. J Am Chem Soc, 2019, 141(9):4108–4118.
    [77] Jiang L, Althoff EA, Clemente FR, et al. De novo computational design of retro-aldol enzymes. Science,2008, 319(5868):1387–1391.
    [78] R?thlisberger D, Khersonsky O, Wollacott AM, et al.Kemp elimination catalysts by computational enzyme design. Nature, 2008, 453(7192):190–195.
    [79] Siegel JB, Zanghellini A, Lovick HM, et al.Computational design of an enzyme catalyst for a stereo selective bimolecular Diels-Alder reaction. Science,2010, 329(5989):309–313.
    [80] Lu PL, Min DY, DiMaio F et al. Accurate computational design of multipass transmembrane proteins. Science,2018, 359(6379):1042–1046.
    [81] Dou JY, Vorobieva AA, Sheffler W, et al. De novo design of a fluorescence-activatingβ-barrel. Nature,2018, 561(7724):485–491.
    [82] Shen H, Fallas JA, Lynch E, et al. De novo design of self-assembling helical protein filaments. Science, 2018,362(6415):705–709.
    [83] Silva DA, Yu S, Ulge UY, et al. De novo design of potent and selective mimics of IL-2 and IL-15. Nature,2019, 565(7738):186–191.
    [84] Boyken SE, Benhaim MA, Busch F, et al. De novo design of tunable, pH-driven conformational changes.Science, 2019, 364(6441):658–664.
    [85] Langan RA, Boyken SE, Ng AH, et al. De novo design of bioactive protein switches. Nature, 2019, 572(7768):205–210.
    [86] Evans R, Jumper J, Kirkpatrick J, et al. De novo structure prediction with deep-learning based scoring//Thirteenth Critical Assessment of Techniques for Protein Structure Prediction(Abstracts). Iberostar,Paraiso, 2018.

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

地址:北京市海淀区学院路29号 邮编:100083

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