Abstract

Cytochrome P450 enzymes (P450s) are broadly distributed among living organisms and play crucial roles in natural product biosynthesis, degradation of xenobiotics, steroid biosynthesis, and drug metabolism. P450s are considered as the most versatile biocatalysts in nature because of the vast variety of substrate structures and the types of reactions they catalyze. In particular, P450s can catalyze regio- and stereoselective oxidations of nonactivated C–H bonds in complex organic molecules under mild conditions, making P450s useful biocatalysts in the production of commodity pharmaceuticals, fine or bulk chemicals, bioremediation agents, flavors, and fragrances. Major efforts have been made in engineering improved P450 systems that overcome the inherent limitations of the native enzymes. In this review, we focus on recent progress of different strategies, including protein engineering, redox-partner engineering, substrate engineering, electron source engineering, and P450-mediated metabolic engineering, in efforts to more efficiently produce pharmaceuticals and other chemicals. We also discuss future opportunities for engineering and applications of the P450 systems. Cytochrome P450 enzymes (P450s) are broadly distributed among living organisms and play crucial roles in natural product biosynthesis, degradation of xenobiotics, steroid biosynthesis, and drug metabolism. P450s are considered as the most versatile biocatalysts in nature because of the vast variety of substrate structures and the types of reactions they catalyze. In particular, P450s can catalyze regio- and stereoselective oxidations of nonactivated C–H bonds in complex organic molecules under mild conditions, making P450s useful biocatalysts in the production of commodity pharmaceuticals, fine or bulk chemicals, bioremediation agents, flavors, and fragrances. Major efforts have been made in engineering improved P450 systems that overcome the inherent limitations of the native enzymes. In this review, we focus on recent progress of different strategies, including protein engineering, redox-partner engineering, substrate engineering, electron source engineering, and P450-mediated metabolic engineering, in efforts to more efficiently produce pharmaceuticals and other chemicals. We also discuss future opportunities for engineering and applications of the P450 systems. Cytochrome P450 enzymes (P450s) 3The abbreviations used are: P450 or CYPcytochrome P450FdRferredoxin reductaseFdxferredoxinAdxadrenodoxinAdRadrenodoxin reductaseCPRcytochrome P450 reductaseCpd 0I, and II, compound 0, I, and IIepPCRerror-prone polymerase chain reaction1α,25(OH)2D31α,25-dihydroxyvitamin D3PDBProtein Data BankSRSsubstrate recognition site. are a superfamily of heme-thiolate–containing proteins named for the characteristic state of the reduced, carbon monoxide (CO)-bound complex displaying a maximum UV-visible absorption band at 450 nm, due to the heme iron group being linked to the apoprotein via an axial conserved cysteine (1Guengerich F.P. Mechanisms of cytochrome P450-catalyzed oxidations.ACS Catal. 2018; 8 (31105987): 10964-1097610.1021/acscatal.8b03401Crossref PubMed Scopus (0) Google Scholar, 2Meunier B. de Visser S.P. Shaik S. Mechanism of oxidation reactions catalyzed by cytochrome P450 enzymes.Chem. Rev. 2004; 104 (15352783): 3947-398010.1021/cr020443gCrossref PubMed Scopus (1611) Google Scholar). cytochrome P450 ferredoxin reductase ferredoxin adrenodoxin adrenodoxin reductase cytochrome P450 reductase I, and II, compound 0, I, and II error-prone polymerase chain reaction 1α,25-dihydroxyvitamin D3 Protein Data Bank substrate recognition site. Since the first discovery of P450 as a pigment (the P denoting “pigment”) in rat liver microsomes in 1958 (3Klingenberg M. Pigments of rat liver microsomes.Arch. Biochem. Biophys. 1958; 75 (13534720): 376-38610.1016/0003-9861(58)90436-3Crossref PubMed Google Scholar), more than 370,000 P450 sequences have been released (UniProt), which are found in human, animals, plants, microbes, and even viruses, demonstrating their incredible and significant diversity in nature (4Nelson D.R. Cytochrome P450 diversity in the tree of life.Biochim. Biophys. Acta Proteins Proteom. 2018; 1866 (28502748): 141-15410.1016/j.bbapap.2017.05.003Crossref PubMed Scopus (0) Google Scholar). P450s play important roles in biosynthetic pathways for natural products, degradation of xenobiotics, biosynthesis of steroid hormones, and drug metabolism (5Zhang X. Li S. Expansion of chemical space for natural products by uncommon P450 reactions.Nat. Prod. Rep. 2017; 34 (28770915): 1061-108910.1039/C7NP00028FCrossref PubMed Google Scholar, 6Guengerich F.P. Common and uncommon cytochrome P450 reactions related to metabolism and chemical toxicity.Chem. Res. Toxicol. 2001; 14 (11409933): 611-65010.1021/tx0002583Crossref PubMed Google Scholar). P450s are considered to be the most versatile biocatalysts in nature (7Coon M.J. Cytochrome P450: nature's most versatile biological catalyst.Annu. Rev. Pharmacol. Toxicol. 2005; 45 (15832443): 1-2510.1146/annurev.pharmtox.45.120403.100030Crossref PubMed Scopus (261) Google Scholar) and are involved in more than 20 different types of chemical oxidation reactions, including hydroxylation, epoxidation, decarboxylation, N- and O-dealkylation, nitration, and C–C bond coupling or cleavage, to name a few (5Zhang X. Li S. Expansion of chemical space for natural products by uncommon P450 reactions.Nat. Prod. Rep. 2017; 34 (28770915): 1061-108910.1039/C7NP00028FCrossref PubMed Google Scholar, 8Guengerich F.P. Munro A.W. Unusual cytochrome P450 enzymes and reactions.J. Biol. Chem. 2013; 288 (23632016): 17065-1707310.1074/jbc.R113.462275Abstract Full Text Full Text PDF PubMed Scopus (184) Google Scholar) (plus some reductions). Furthermore, the substrate diversity of P450s covers almost all classes of organic structures found in nature (e.g. terpenoids, polyketides, fatty acids, alkaloids, and polypeptides) (5Zhang X. Li S. Expansion of chemical space for natural products by uncommon P450 reactions.Nat. Prod. Rep. 2017; 34 (28770915): 1061-108910.1039/C7NP00028FCrossref PubMed Google Scholar, 9Podust L.M. Sherman D.H. Diversity of P450 enzymes in the biosynthesis of natural products.Nat. Prod. Rep. 2012; 29 (22820933): 1251-126610.1039/c2np20020aCrossref PubMed Scopus (156) Google Scholar, 10Rudolf J.D. Chang C.Y. Ma M. Shen B. Cytochromes P450 for natural product biosynthesis in Streptomyces: sequence, structure, and function.Nat. Prod. Rep. 2017; 34 (28758170): 1141-117210.1039/C7NP00034KCrossref PubMed Google Scholar). The ubiquitous distribution and the multiplicity of reactions and substrates demonstrate the plasticity of P450 enzyme systems, providing a limitless space for mining, engineering, and designing P450 systems for practical catalysis. Among diverse functionalities, the most important is that P450s are capable of catalyzing the regio- and stereoselective oxidation of inert C–H bonds in complex molecular scaffolds under mild conditions, making them superior to many chemical catalysts and of great interest for pharmaceutical, chemical, and biotechnological applications. However, the narrow substrate scope of some P450s, low catalytic efficiency, low stability, dependence on redox partners, high cost of cofactors, and electron uncoupling have limited the industrial applications of P450s (11Sakaki T. Practical application of cytochrome P450.Biol. Pharm. Bull. 2012; 35 (22687473): 844-84910.1248/bpb.35.844Crossref PubMed Google Scholar, 12Bernhardt R. Urlacher V.B. Cytochromes P450 as promising catalysts for biotechnological application: chances and limitations.Appl. Microbiol. Biotechnol. 2014; 98 (24848420): 6185-620310.1007/s00253-014-5767-7Crossref PubMed Scopus (201) Google Scholar). More recently, innovative P450 systems have been developed to fuel industrial projects with the use of a number of new engineering strategies (e.g. interactions of essential elements, including P450 itself, redox partner, substrate, and cofactor). These include the powerful directed evolution approach pioneered by the Nobel Laureate Frances H. Arnold, used to build unnatural but more robust P450 systems (13Arnold F.H. Design by directed evolution.Acc. Chem. Res. 1998; 31: 125-13110.1021/ar960017fCrossref Google Scholar). Several excellent reviews have covered the diversity, functions, novel chemistry, and applications of P450s (5Zhang X. Li S. Expansion of chemical space for natural products by uncommon P450 reactions.Nat. Prod. Rep. 2017; 34 (28770915): 1061-108910.1039/C7NP00028FCrossref PubMed Google Scholar, 10Rudolf J.D. Chang C.Y. Ma M. Shen B. Cytochromes P450 for natural product biosynthesis in Streptomyces: sequence, structure, and function.Nat. Prod. Rep. 2017; 34 (28758170): 1141-117210.1039/C7NP00034KCrossref PubMed Google Scholar, 14Urlacher V.B. Girhard M. Cytochrome P450 monooxygenases in biotechnology and synthetic biology.Trends Biotechnol. 2019; 37 (30739814): 882-89710.1016/j.tibtech.2019.01.001Abstract Full Text Full Text PDF PubMed Scopus (71) Google Scholar, 15Xu L.H. Du Y.L. Rational and semi-rational engineering of cytochrome P450s for biotechnological applications.Synth. Syst. Biotechnol. 2018; 3 (30533540): 283-29010.1016/j.synbio.2018.10.001Crossref PubMed Scopus (6) Google Scholar, 16Wei Y. Ang E.L. Zhao H. Recent developments in the application of P450 based biocatalysts.Curr. Opin. Chem. Biol. 2018; 43 (29100098): 1-710.1016/j.cbpa.2017.08.006Crossref PubMed Scopus (46) Google Scholar, 17Schmitz L.M. Rosenthal K. Lütz S. Recent advances in heme biocatalysis engineering.Biotechnol. Bioeng. 2019; 116 (31483477): 3469-347510.1002/bit.27156Crossref PubMed Scopus (5) Google Scholar). For more insight into intriguing P450-related mechanisms and to deeply understand the strategies related to the practical application of P450 catalysis, we will focus on recent advances in P450 protein engineering, particularly engineering strategies for optimization of the interaction between P450s and redox partners. We will also consider substrate engineering, cofactor (NAD(P)H) regeneration, and several atypical strategies for engineering the electron transport system. Finally, a brief summary of P450-related metabolic engineering will be provided. In general, a P450 catalytic system includes four components: the substrate, a P450 enzyme for substrate binding and oxidative catalysis, the redox partner(s) that functions as an electron transfer shuttle, and the cofactor (NAD(P)H), which provides the reducing equivalents. Most P450s share a common sophisticated catalytic cycle (Fig. 1) (2Meunier B. de Visser S.P. Shaik S. Mechanism of oxidation reactions catalyzed by cytochrome P450 enzymes.Chem. Rev. 2004; 104 (15352783): 3947-398010.1021/cr020443gCrossref PubMed Scopus (1611) Google Scholar, 5Zhang X. Li S. Expansion of chemical space for natural products by uncommon P450 reactions.Nat. Prod. Rep. 2017; 34 (28770915): 1061-108910.1039/C7NP00028FCrossref PubMed Google Scholar, 18Jiang Y. Li S. Catalytic function and application of cytochrome P450 enzymes in biosynthesis and organic synthesis.Chinese J. Org. Chem. 2018; 38: 2307-232310.6023/cjoc201805055Crossref Scopus (4) Google Scholar), using the typical hydroxylation reaction as a paradigm, as shown in Fig. 1. The ferric resting state (generally) of a P450 (A) first accepts a substrate (RH), which displaces an active-site water molecule but does not bond directly to the iron. The ferric iron (FeIII) of the high-spin, substrate-bound complex (B) is then reduced to ferrous iron (FeII) (C) by one electron, transferred via a redox partner. Next, binding of dioxygen to FeII results in the [FeII O2] complex (D). The complex D is reduced by the second electron to form complex E, which uses a proton from solvent to generate a ferric hydroperoxo species [FeIII–OOH] (F), referred as to Compound 0 (Cpd 0). The O–O bond of Cpd 0 is cleaved upon the addition of the second proton and releases a molecule of water to generate the high-valent porphyrin π radical cation tetravalent iron [FeIV=O] (i.e. Compound I (Cpd I; G)). This highly reactive complex abstracts a hydrogen atom from the substrate, leading to the formation of the ferryl-hydroxo compound II (Cpd II; H). Subsequently, the hydroxylated product (R-OH) is formed by the reaction of the substrate radical with the hydroxyl group of Cpd II and released from the active site of complex I. Finally, a molecule of water returns to coordinate with FeIII, restoring the resting state A. The same catalytic cycle is initiated repeatedly as substrate molecules bind to the heme-centered active site of P450. It is worth noting that some P450s are capable of directly utilizing H2O2 as the sole electron and proton donor to form Cpd 0 and do catalysis via the so-called peroxide shunt pathway (Fig. 1, dashed arrows). However, this shunt pathway is greatly limited by the low efficiency and the low H2O2 tolerance of most P450s, except P450 peroxygenases (e.g. CYP152 subfamily) (19Matthews S. Belcher J.D. Tee K.L. Girvan H.M. McLean K.J. Rigby S.E. Levy C.W. Leys D. Parker D.A. Blankley R.T. Munro A.W. Catalytic determinants of alkene production by the cytochrome P450 peroxygenase OleTJE.J. Biol. Chem. 2017; 292 (28053093): 5128-514310.1074/jbc.M116.762336Abstract Full Text Full Text PDF PubMed Scopus (40) Google Scholar). The well-studied and established catalytic cycle provides a theoretical basis and roadmap to understand and manipulate this P450 peroxygenase subfamily by protein and substrate engineering. Maintenance of the P450 catalytic cycle relies on continuous electron transport to the heme-iron by redox partners, which are complicated electron-transfer systems. Based on the types of redox partners and the P450-redox partner interaction relationships, P450 systems can be divided into five main classes (10Rudolf J.D. Chang C.Y. Ma M. Shen B. Cytochromes P450 for natural product biosynthesis in Streptomyces: sequence, structure, and function.Nat. Prod. Rep. 2017; 34 (28758170): 1141-117210.1039/C7NP00034KCrossref PubMed Google Scholar, 11Sakaki T. Practical application of cytochrome P450.Biol. Pharm. Bull. 2012; 35 (22687473): 844-84910.1248/bpb.35.844Crossref PubMed Google Scholar, 15Xu L.H. Du Y.L. Rational and semi-rational engineering of cytochrome P450s for biotechnological applications.Synth. Syst. Biotechnol. 2018; 3 (30533540): 283-29010.1016/j.synbio.2018.10.001Crossref PubMed Scopus (6) Google Scholar) (Fig. 2). The Class I P450 system present in most bacterial and mitochondrial P450s has a two-component redox partner system, comprised of an FAD-containing ferredoxin reductase (FdR) and a small iron-sulfur-containing ferredoxin (Fdx) (20Sevrioukova I.F. Poulos T.L. Structural biology of redox partner interactions in P450cam monooxygenase: a fresh look at an old system.Arch. Biochem. Biophys. 2011; 507 (20816746): 66-7410.1016/j.abb.2010.08.022Crossref PubMed Scopus (42) Google Scholar, 21Kido T. Kimura T. The formation of binary and ternary complexes of cytochrome P-450scc with adrenodoxin and adrenodoxin reductase·adrenodoxin complex: the implication in ACTH function.J. Biol. Chem. 1979; 254 (227881): 11806-11815Abstract Full Text PDF PubMed Google Scholar). The Class II P450 system employed by eukaryotic organisms has a single-component redox partner, which is a membrane-bound protein containing both an FAD and an FMN domain, termed cytochrome P450 reductase (CPR). Class III P450 systems have a eukaryotic-like CPR naturally fused to the C terminus of the P450 domain through a flexible linker, represented by Bacillus megaterium P450BM3 (CYP102A1) (22Whitehouse C.J.C. Bell S.G. Wong L.-L. P450BM3(CYP102A1): connecting the dots.Chem. Soc. Rev. 2012; 41 (22008827): 1218-126010.1039/C1CS15192DCrossref PubMed Google Scholar). Class IV P450 systems are exemplified by P450 RhF from Rhodococcus sp. NCIMB 9784, whose FMN/Fe2S2-containing reductase domain forms a natural fusion with the P450 domain (23Roberts G.A. Celik A. Hunter D.J. Ost T.W. White J.H. Chapman S.K. Turner N.J. Flitsch S.L. A self-sufficient cytochrome P450 with a primary structural organization that includes a flavin domain and a [2Fe-2S] redox center.J. Biol. Chem. 2003; 278 (14514666): 48914-4892010.1074/jbc.M309630200Abstract Full Text Full Text PDF PubMed Scopus (90) Google Scholar). Interestingly, a few P450s can directly interact with their electron donors and are independent of additional redox partner proteins to accomplish the catalytic reactions; these Class V P450s include P450Nor (24Daiber A. Shoun H. Ullrich V. Nitric oxide reductase (P450nor) from Fusarium oxysporum.J. Inorg. Biochem. 2005; 99 (15598501): 185-19310.1016/j.jinorgbio.2004.09.018Crossref PubMed Scopus (91) Google Scholar) and P450 TxA (25Hsu P.-Y. Tsai A.-L. Kulmacz R.J. Wang L.-H. Expression, purification, and spectroscopic characterization of human thromboxane synthase.J. Biol. Chem. 1999; 274 (9873013): 762-76910.1074/jbc.274.2.762Abstract Full Text Full Text PDF PubMed Scopus (38) Google Scholar). Class III–V P450s are independent of redox partner proteins and are often called self-sufficient P450s. Notably, these single-component P450 systems provide very desirable scaffolds for engineering P450 systems, due to their self-sufficiency and hence the significantly increased electron transport efficiency. It is worth noting that other classification systems also exist: Munro et al. (26Munro A.W. Girvan H.M. McLean K.J. Cytochrome P450-redox partner fusion enzymes.Biochim. Biophys. Acta. 2007; 1770 (17023115): 345-35910.1016/j.bbagen.2006.08.018Crossref PubMed Scopus (145) Google Scholar) have categorized five other novel P450-fused redox partner systems in addition to the classical Class I and Class II types, and Bernhardt et al. (27Hannemann F. Bichet A. Ewen K.M. Bernhardt R. Cytochrome P450 systems—biological variations of electron transport chains.Biochim. Biophys. Acta. 2007; 1770 (16978787): 330-34410.1016/j.bbagen.2006.07.017Crossref PubMed Scopus (482) Google Scholar) classified 10 types for P450s based on the topology of protein components involved in the electron transfer chains of P450 enzymes. The incomparable diversity of P450s regarding substrates and reaction types provides nearly limitless application potential for production of chemicals and pharmaceuticals (28Paddon C.J. Westfall P.J. Pitera D.J. Benjamin K. Fisher K. McPhee D. Leavell M.D. Tai A. Main A. Eng D. Polichuk D.R. Teoh K.H. Reed D.W. Treynor T. Lenihan J. et al.High-level semi-synthetic production of the potent antimalarial artemisinin.Nature. 2013; 496 (23575629): 528-53210.1038/nature12051Crossref PubMed Scopus (1076) Google Scholar, 29McLean K.J. Hans M. Meijrink B. van Scheppingen W.B. Vollebregt A. Tee K.L. van der Laan J.M. Leys D. Munro A.W. van den Berg M.A. Single-step fermentative production of the cholesterol-lowering drug pravastatin via reprogramming of Penicillium chrysogenum.Proc. Natl. Acad. Sci. U.S.A. 2015; 112 (25691737): 2847-285210.1073/pnas.1419028112Crossref PubMed Scopus (59) Google Scholar), biosensor-based analysis (30Bistolas N. Wollenberger U. Jung C. Scheller F.W. Cytochrome P450 biosensors—a review.Biosens. Bioelectron. 2005; 20 (15854816): 2408-242310.1016/j.bios.2004.11.023Crossref PubMed Scopus (175) Google Scholar), chemoenzymatic synthesis (31Lowell A.N. DeMars 2nd, M.D. Slocum S.T. Yu F. Anand K. Chemler J.A. Korakavi N. Priessnitz J.K. Park S.R. Koch A.A. Schultz P.J. Sherman D.H. Chemoenzymatic total synthesis and structural diversification of tylactone-based macrolide antibiotics through late-stage polyketide assembly, tailoring, and C–H functionalization.J. Am. Chem. Soc. 2017; 139 (28525276): 7913-792010.1021/jacs.7b02875Crossref PubMed Scopus (34) Google Scholar), and pollutant biodegradation (32Du L. Dong S. Zhang X. Jiang C. Chen J. Yao L. Wang X. Wan X. Liu X. Wang X. Huang S. Cui Q. Feng Y. Liu S.J. Li S. Selective oxidation of aliphatic C–H bonds in alkylphenols by a chemomimetic biocatalytic system.Proc. Natl. Acad. Sci. U.S.A. 2017; 114 (28607077): E5129-E513710.1073/pnas.1702317114PubMed Google Scholar). For instance, the Saccharopolyspora erythraea EryF and EryK P450s are involved in the production of the antibacterial agent erythromycin (Fig. 3, compound 1) (33Minas W. Brünker P. Kallio P.T. Bailey J.E. Improved erythromycin production in a genetically engineered industrial strain of Saccharopolyspora erythraea.Biotechnol. Prog. 1998; 14 (9694676): 561-56610.1021/bp980055tCrossref PubMed Scopus (0) Google Scholar); Streptomyces fradiae TylI and TylHI P450s are involved in the biosynthesis of the antimycoplasma drug tylosin (Fig. 3, compound 2) (34Bate N. Cundliffe E. The mycinose-biosynthetic genes of Streptomyces fradiae, producer of tylosin.J. Ind. Microbiol. Biotechnol. 1999; 23 (10510490): 118-12210.1038/sj.jim.2900707Crossref PubMed Scopus (0) Google Scholar); and Aspergillus terreus LovA is responsible for biosynthesizing monacolin J acid (Fig. 3, compound 3), the precursor of a series of cholesterol-lowering statin drugs (35Barriuso J. Nguyen D.T. Li J.W. Roberts J.N. MacNevin G. Chaytor J.L. Marcus S.L. Vederas J.C. Ro D.K. Double oxidation of the cyclic nonaketide dihydromonacolin L to monacolin J by a single cytochrome P450 monooxygenase, LovA.J. Am. Chem. Soc. 2011; 133 (21495633): 8078-808110.1021/ja201138vCrossref PubMed Scopus (44) Google Scholar, 36Huang X. Tang S. Zheng L. Teng Y. Yang Y. Zhu J. Lu X. Construction of an efficient and robust Aspergillus terreus cell factory for monacolin J production.ACS Synth. Biol. 2019; 8 (30856313): 818-82510.1021/acssynbio.8b00489Crossref PubMed Scopus (4) Google Scholar). The production of high value-added chemical intermediates from phenolic environmental pollutants has been achieved with P450 biodegradation systems (32Du L. Dong S. Zhang X. Jiang C. Chen J. Yao L. Wang X. Wan X. Liu X. Wang X. Huang S. Cui Q. Feng Y. Liu S.J. Li S. Selective oxidation of aliphatic C–H bonds in alkylphenols by a chemomimetic biocatalytic system.Proc. Natl. Acad. Sci. U.S.A. 2017; 114 (28607077): E5129-E513710.1073/pnas.1702317114PubMed Google Scholar, 37Sulistyaningdyah W.T. Ogawa J. Li Q.S. Maeda C. Yano Y. Schmid R.D. Shimizu S. Hydroxylation activity of P450 BM-3 mutant F87V towards aromatic compounds and its application to the synthesis of hydroquinone derivatives from phenolic compounds.Appl. Microbiol. Biotechnol. 2005; 67 (15549292): 556-56210.1007/s00253-004-1761-9Crossref PubMed Scopus (49) Google Scholar, 38Du L. Ma L. Qi F. Zheng X. Jiang C. Li A. Wan X. Liu S.J. Li S. Characterization of a unique pathway for 4-cresol catabolism initiated by phosphorylation in Corynebacterium glutamicum.J. Biol. Chem. 2016; 291 (26817843): 6583-659410.1074/jbc.M115.695320Abstract Full Text Full Text PDF PubMed Scopus (0) Google Scholar), and soluble P450s have been used in bacterial cell libraries to mimic human P450 drug metabolic profiles (39Otey C.R. Bandara G. Lalonde J. Takahashi K. Arnold F.H. Preparation of human metabolites of propranolol using laboratory-evolved bacterial cytochromes P450.Biotechnol. Bioeng. 2006; 93 (16224788): 494-49910.1002/bit.20744Crossref PubMed Scopus (125) Google Scholar, 40Parikh A. Gillam E.M. Guengerich F.P. Drug metabolism by Escherichia coli expressing human cytochromes P450.Nat. Biotechnol. 1997; 15 (9255795): 784-78810.1038/nbt0897-784Crossref PubMed Google Scholar). Genome mining and high-throughput screening of P450s have proven to be effective and successful strategies for seeking suitable and robust biocatalysts in industry. P450sca-2 (CYP105A3), screened from Streptomyces carbophilus, is able to catalyze the 6β-hydroxylation of compactin produced by Penicillium citrinum, generating the cholesterol-lowering drug pravastatin (Fig. 3, compound 4) (41Hosobuchi M. Kurosawa K. Yoshikawa H. Application of computer to monitoring and control of fermentation process: microbial conversion of ML-236B Na to pravastatin.Biotechnol. Bioeng. 1993; 42 (18613128): 815-82010.1002/bit.260420705Crossref PubMed Scopus (32) Google Scholar), considered to be one of the most successful instances of practical P450 catalysis in industry (11Sakaki T. Practical application of cytochrome P450.Biol. Pharm. Bull. 2012; 35 (22687473): 844-84910.1248/bpb.35.844Crossref PubMed Google Scholar, 42Yasuda K. Sugimoto H. Hayashi K. Takita T. Yasukawa K. Ohta M. Kamakura M. Ikushiro S. Shiro Y. Sakaki T. Protein engineering of CYP105s for their industrial uses.Biochim. Biophys. Acta Proteins Proteom. 2018; 1866 (28583351): 23-3110.1016/j.bbapap.2017.05.014Crossref PubMed Scopus (5) Google Scholar). The bioconversion of 11-deoxycortisol into hydrocortisol by the P450lun-containing fungus Curvularia lunata has been launched by Bayer on an industrial scale (11Sakaki T. Practical application of cytochrome P450.Biol. Pharm. Bull. 2012; 35 (22687473): 844-84910.1248/bpb.35.844Crossref PubMed Google Scholar, 43Suzuki K. Sanga K. Chikaoka Y. Itagaki E. Purification and properties of cytochrome P-450 (P-450lun) catalyzing steroid 11β-hydroxylation in Curvularia lunata.Biochim. Biophys. Acta. 1993; 1203 (8268203): 215-22310.1016/0167-4838(93)90086-7Crossref PubMed Scopus (49) Google Scholar) (Table 1 and Fig. 3, compound 5). The industrially relevant P450 VD25 (CYP105A2) from Amycolata autotrophica (later renamed as Pseudonocardia autotrophica) is capable of transforming vitamin D3 into its most bioactive form, 1α,25-dihydroxyvitamin D3 (1α,25(OH)2D3) (44Kawauchi H. Sasaki J. Adachi T. Hanada K. Beppu T. Horinouchi S. Cloning and nucleotide sequence of a bacterial cytochrome P-450VD25 gene encoding vitamin D-3 25-hydroxylase.Biochim. Biophys. Acta. 1994; 1219 (8086461): 179-18310.1016/0167-4781(94)90266-6Crossref PubMed Scopus (20) Google Scholar) (Table 1 and Fig. 3, compound 6). The P450 hydroxylase CYP-sb21 from the rare actinomycetes Sebekia benihana and CYP-pa1 from P. autotrophica are candidate biocatalysts for site-selective hydroxylation of the immunosuppressive drug cyclosporin A to two hair-stimulating agents with significantly decreased immunosuppressant activity, γ-hydroxy-N-methyl-l-Leu4-cyclosporin A and γ-hydroxy-N-methyl-l-Leu9-cyclosporin A, respectively (45Ma L. Du L. Chen H. Sun Y. Huang S. Zheng X. Kim E.S. Li S. Reconstitution of the in vitro activity of the cyclosporine-specific P450 hydroxylase from Sebekia benihana and development of a heterologous whole-cell biotransformation system.Appl. Environ. Microbiol. 2015; 81 (26150455): 6268-627510.1128/AEM.01353-15Crossref PubMed Scopus (21) Google Scholar, 46Ban J.G. Woo M.W. Lee B.R. Lee M.J. Choi S.S. Kim E.S. A novel regiospecific cyclosporin hydroxylase gene revealed through the genome mining of Pseudonocardia autotrophica.J. Ind. Microbiol. Biotechnol. 2014; 41 (24659179): 879-88610.1007/s10295-014-1432-5Crossref PubMed Scopus (4) Google Scholar, 47Sun Y. Ma L. Han D. Du L. Qi F. Zhang W. Sun J. Huang S. Kim E.S. Li S. In vitro reconstitution of the cyclosporine specific P450 hydroxylases using heterologous redox partner proteins.J. Ind. Microbiol. Biotechnol. 2017; 44 (27888364): 161-16610.1007/s10295-016-1875-yCrossref PubMed Scopus (7) Google Scholar, 48Lee M.-J. Kim H.-B. Yoon Y.J. Han K. Kim E.-S. Identification of a cyclosporine-specific P450 hydroxylase gene through targeted cytochrome P450 complement (CYPome) disruption in Sebekia benihana.Appl. Environ. Microbiol. 2013; 79 (23354713): 2253-226210.1128/AEM.03722-12Crossref PubMed Scopus (12) Google Scholar) (Table 1 and Fig. 3, compounds 7–9).Table 1Selected P450s involved in production of pharmaceuticals and chemical intermediatesP450PDB codeOriginWT/mutantFunctionSubstrateReferenceEryF (CYP107A1)1OXASaccharopolyspora erythraeaWT6-Hydroxylation6-Deoxyerythronolide B33Minas W. Brünker P. Kallio P.T. Bailey J.E. Improved erythromycin production in a genetically engineered industrial strain of Saccharopolyspora erythraea.Biotechnol. Prog. 1998; 14 (9694676): 561-56610.1021/bp980055tCrossref PubMed Scopus (0) Google ScholarEryK (CYP113A1)2JJNS. erythraeaWT12-HydroxylationErythromycin D33Minas W. Brünker P. Kallio P.T. Bailey J.E. Improved erythromycin production in a genetically engineered industrial strain of Saccharopolyspora erythraea.Biotechnol. Prog. 1998; 14 (9694676): 561-56610.1021/bp980055tCrossref PubMed Scopus (0) Google ScholarTylIStreptomyces fradiaeWTC20 hydroxylation/dehydrogenation5-Mycaminosyl tylacone34Bate N. Cundliffe E. The mycinose-biosynthetic genes of Streptomyces fradiae, producer of tylosin.J. Ind. Microbiol. Biotechnol. 1999; 23 (10510490): 118-12210.1038/sj.jim.2900707Crossref PubMed Scopus (0) Google ScholarTylHI6B11S. fradiaeWTC23 Hydroxylation23-Deoxy-5-omycaminosyl-tylonolide34Bate N. Cundliffe E. The mycinose-biosynthetic genes of Streptomyces fradiae, producer of tylosin.J. Ind. Microbiol. Biotechnol. 1999; 23 (10510490): 118-12210.1038/sj.jim.2900707Crossref PubMed Scopus (0) Google ScholarLovAAspergillus terreusWT4a/5-Dehydrogenation, 8-hydroxylationDihydromonacolin L35Barriuso J. Nguyen D.T. Li J.W. Roberts J.N. MacNevin G. Chaytor J.L. Marcus S.L. Vederas J.C. Ro D.K. Double oxidation of the cyclic nonaketide dihydromonacolin L to monacolin J by a single cytochrome P450 monooxygenase, LovA.J. Am. Chem. Soc. 2011; 133 (21495633): 8078-808110.1021/ja201138vCrossref PubMed Scopus (44) Google ScholarCYP71AV1Artemisia annuaWT12-CarboxylationAmorphadiene49Ro D.K. Paradise E.M. Ouellet M. Fisher K.J. Newman K.L. Ndungu J.M. Ho K.A. Eachus R.A. Ham T.S. Kirby J. Chang M.C. Withers S.T. Shiba Y. Sarpong R. Keasling J.D. Production of the antimalarial drug precursor artemisinic acid in engineered yeast.Nature. 2006; 440 (16612385): 940-94310.1038/nature04640Crossref PubMed Scopus (1925) Google ScholarP450sca-2 (CYP105A3)Streptomyces carbophilusWT6β-HydroxylationCompactin41Hosobuchi M. Kur

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call