Please wait a minute...

中国生物工程杂志

China Biotechnology
China Biotechnology  2016, Vol. 36 Issue (11): 83-89    DOI: 10.13523/j.cb.20161112
    
Progress and Prospect of Heterologous Biosynthesis of Ttriterpenoids in Engineered Escherichia coli
ZHANG Qiang, LI Da shuai, LU Wen yu
Department of Biological Engineering, School of Chemical Engineering and Technology, Tianjin University, Key Laboratory of Systems Bioengineering, Ministry of Education, SynBio Research Platform, Collaborative Innovation Center of Chemical Science and Engineering, Tianjin 300072, China
Download: HTML   PDF(582KB) HTML
Export: BibTeX | EndNote (RIS)      

Abstract  

Triterpenoids are the major pharmacologically active constituents of many medicinal plants, which exhibit potential economic value. Currently, triterpenoids are mainly produced through their extraction from plants which is complicated, time-consuming, labour intensive and produce relatively low yields. Escherichia coli as a commonly used recombinant microbial system has the ability to generate valuable natural products, so, heterologous biosynthesis of triterpenoids or precursors in E. coli presents an attractive system. Research progress of heterologous biosynthesis of triterpenoids is summarized, including metabolic pathway, key enzymes and optimization of E. coli chassis and introduction of functional modules. Basic problems on efficient synthesis of triterpenoids in E. coli are discussed and a prospect of using E. coli as a new chassis of biosynthesis of triterpenoids is analyzed.



Key wordsSynthetic biology      Heterologous biosynthesis      Escherichia coli      Triterpenoids     
Received: 21 April 2016      Published: 25 November 2016
ZTFLH:  Q93-91  
Cite this article:

ZHANG Qiang, LI Da shuai, LU Wen yu. Progress and Prospect of Heterologous Biosynthesis of Ttriterpenoids in Engineered Escherichia coli. China Biotechnology, 2016, 36(11): 83-89.

URL:

https://manu60.magtech.com.cn/biotech/10.13523/j.cb.20161112     OR     https://manu60.magtech.com.cn/biotech/Y2016/V36/I11/83

[1] Phillips D R, Rasbery J M, Bartel B, et al. Biosynthetic diversity in plant triterpene cyclization. Current Opinion in Plant Biology, 2006, 9(3):305-314.
[2] Watanabe K. Proceedings:effective use of heterologous hosts for characterization of biosynthetic enzymes allows production of natural products and promotes new natural product discovery. The Japanese journal of antibiotics, 2015, 68(1):55-67.
[3] Thimmappa R, Geisler K, Louveau T, et al. Triterpene biosynthesis in plants. Annual Review of Plant Biology, 2014, 65(1):225-257.
[4] Hoshino T,Sato T.Squalene-hopene cyclase:catalytic mechanism and substrate recognition. Chemical Communications, 2002,33(4):291-301.
[5] Augustin J M, Kuzina V, Andersen S B, et al. Molecular activities, biosynthesis and evolution of triterpenoid saponins. Phytochemistry, 2011, 72(6):435-457.
[6] Facchini P J, Bohlmann J, Covello P S, et al. Synthetic biosystems for the production of high-value plant metabolites. Trends in Biotechnology, 2012, 30(3):127-131.
[7] Thimmappa R, Geisler K, Louveau T, et al. Triterpene biosynthesis in plants. Annual Review of Plant Biology, 2014, 65(1):225-257.
[8] Dai Z, Liu Y, Zhang X, et al. Metabolic engineering of Saccharomyces cerevisiae for production of ginsenosides. Metabolic Engineering, 2013, 20(5):146-156.
[9] Dai Z, Wang B, Liu Y, et al. Producing aglycons of ginsenosides in bakers' yeast. Scientific Reports, 2014, 4(4):3698.
[10] Zhao F, Bai P, Liu T, et al. Optimization of a cytochrome P450 oxidation system for enhancing protopanaxadiol production in Saccharomyces cerevisiae. Biotechnology and Bioengineering, 2016,113(8):1787-1795.
[11] Yan X, Fan Y, Wei W, et al. Production of bioactive ginsenoside compound K in metabolically engineered yeast. Cell Research, 2014, 24(6):770-773.
[12] Wang P, Wei Y, Fan Y, et al. Production of bioactive ginsenosides Rh2 and Rg3 by metabolically engineered yeasts. Metabolic Engineering, 2015, 29:97-105.
[13] Moses T, Pollier J, Almagro L, et al. Combinatorial biosynthesis of sapogenins and saponins in Saccharomyces cerevisiae using a C-16α hydroxylase from Bupleurum falcatum. Proceedings of the National Academy of Sciences, 2014, 111(4):1634-1639.
[14] Davidovich-Rikanati R, Shalev L, Baranes N, et al. Recombinant yeast as a functional tool for understanding bitterness and cucurbitacin biosynthesis in watermelon (Citrullus spp.). Yeast, 2015, 32(1):103-114.
[15] Huang L, Li J, Ye H, et al. Molecular characterization of the pentacyclic triterpenoid biosynthetic pathway in Catharanthus roseus. Planta, 2012, 236(5):1571-1581.
[16] Li D, Zhang Q, Zhou Z, et al. Heterologous biosynthesis of triterpenoid dammarenediol-Ⅱ in engineered Escherichia coli. Biotechnology Letters, 2016, 38(4):1-7.
[17] Katabami A, Li L, Iwasaki M, et al. Production of squalene by squalene synthases and their truncated mutants in Escherichia coli. Journal of Bioscience and Bioengineering, 2015, 119(2):165-171.
[18] Laden B P, Tang Y,Porter T D. Cloning, heterologous expression, and enzymological characterization of human squalene monooxygenase. Archives of Biochemistry and Biophysics, 2000, 374(2):381-388.
[19] Jiang M, Stephanopoulos G, Pfeifer B A. Toward biosynthetic design and implementation of Escherichia coli-derived paclitaxel and other heterologous polyisoprene compounds. Applied and Environmental Microbiology, 2012, 78(8):2497-2504.
[20] Leonard E,Koffas M G.Engineering of artificial plant cytochrome P450 enzymes for synthesis of iIsoflavones by Escherichia coli. Applied and Environmental Microbiology, 2007, 73(22):7246-7251.
[21] Ajikumar P K, Xiao W H, Tyo K E, et al. Isoprenoid pathway optimization for Taxol precursor overproduction in Escherichia coli. Science, 2010, 330(6000):70-74.
[22] Tsuruta H, Paddon C J, Eng D, et al. High-level production of amorpha-4, 11-diene, a precursor of the antimalarial agent artemisinin, in Escherichia coli. PLoS One, 2009, 4(2):e4489.
[23] Tansakul P, Shibuya M, Kushiro T, et al. Dammarenediol-Ⅱ synthase, the first dedicated enzyme for ginsenoside biosynthesis, in Panax ginseng. FEBS Letters, 2006, 580(22):5143-5149.
[24] Li J,Zhang Y.Increase of betulinic acid production in Saccharomyces cerevisiae by balancing fatty acids and betulinic acid forming pathways. Applied Microbiology and Biotechnology, 2014, 98(7):3081-3089.
[25] Fukushima E O, Seki H, Ohyama K, et al. CYP716A subfamily members are multifunctional oxidases in triterpenoid biosynthesis. Plant and Cell Physiology, 2011, 52(12):2050-2061.
[26] Shibuya M, Zhang H, Endo A, et al. Two branches of the lupeol synthase gene in the molecular evolution of plant oxidosqualene cyclases. European Journal of Biochemistry, 1999, 266(1):302-307.
[27] Castillo D A, Kolesnikova M D, Matsuda S P. An effective strategy for exploring unknown metabolic pathways by genome mining. Journal of the American Chemical Society, 2013, 135(15):5885-5894.
[28] Field B, Fiston-Lavier AS, Kemen A, et al. Formation of plant metabolic gene clusters within dynamic chromosomal regions. Proceedings of the National Academy of Sciences, 2011, 108(38):16116-16121.
[29] Fukushima E O, Seki H, Ohyama K, et al. CYP716A subfamily members are multifunctional oxidases in triterpenoid biosynthesis. Plant and Cell Physiology, 2011, 52(12):2050-2061.
[30] Ba L, Li P, Zhang H, et al. Semi-rational engineering of cytochrome P450sca-2 in a hybrid system for enhanced catalytic activity:Insights into the important role of electron transfer. Biotechnology and Bioengineering, 2013, 110(11):2815-2825.
[31] Fasan R, Crook N C, Peters M W, et al. Improved product-per-glucose yields in P450-dependent propane biotransformations using engineered Escherichia coli. Biotechnology and Bioengineering, 2011, 108(3):500-510.
[32] Li S, Podust L M,Sherman D H.Engineering and analysis of a self-sufficient biosynthetic cytochrome P450 PikC fused to the RhFRED reductase domain. Journal of the American Chemical Society, 2007, 129(43):12940-12941.
[33] Shen A L, Porter T, Wilson T, et al. Structural analysis of the FMN binding domain of NADPH-cytochrome P-450 oxidoreductase by site-directed mutagenesis. Journal of Biological Chemistry, 1989, 264(13):7584-7589.
[34] Venkateswarlu K, Lamb D C, Kelly D E, et al. The N-terminal membrane domain of yeast NADPH-cytochrome P450(CYP) oxidoreductase is not required for catalytic activity in sterol biosynthesis or in reconstitution of CYP activity. Journal of Biological Chemistry, 1998, 273(8):4492-4496.
[35] Sadeghi S J,Gilardi G.Chimeric P450 enzymes:activity of artificial redox fusions driven by different reductases for biotechnological applications. Biotechnology and Applied Biochemistry, 2013, 60(1):102-110.
[36] Meesapyodsuk D, Balsevich J, Reed D W, et al. Saponin biosynthesis in Saponaria vaccaria cDNAs encoding β-amyrin synthase and a triterpene carboxylic acid glucosyltransferase. Plant Physiology, 2007, 143(2):959-969.
[37] Naoumkina M A, Modolo L V, Huhman D V, et al. Genomic and coexpression analyses predict multiple genes involved in triterpene saponin biosynthesis in Medicago truncatula. The Plant Cell, 2010, 22(3):850-866.
[38] Wang J, Li S, Xiong Z, et al. Pathway mining-based integration of critical enzyme parts for de novo biosynthesis of steviolglycosides sweetener in Escherichia coli. Cell Research, 2015,26(2):1-4.
[39] Peralta-Yahya P P, Ouellet M, Chan R, et al. Identification and microbial production of a terpene-based advanced biofuel. Nature Communications, 2011, 2(9):2749-2763.
[40] Sliva A, Yang H, Boeke J D, et al. Freedom and responsibility in synthetic genomics:The Synthetic yeast project. Genetics, 2015, 200(4):1021-1028.

[1] MA Ning,WANG Han-jie. Advances of Optogenetics in the Regulation of Bacterial Production[J]. China Biotechnology, 2021, 41(9): 101-109.
[2] HUANG Huan-bang,WU Yang,YANG You-hui,WANG Zhao-guan,QI Hao. Progress in Incorporation of Non-canonical Amino Acid Based on Archaeal Tyrosyl-tRNA Synthetase[J]. China Biotechnology, 2021, 41(9): 110-125.
[3] GUO Man-man,TIAN Kai-ren,QIAO Jian-jun,LI Yan-ni. Application of Phage Recombinase Systems in Synthetic Biology[J]. China Biotechnology, 2021, 41(8): 90-102.
[4] DONG Shu-xin,QIN Lei,LI Chun,LI Jun. Transcription Factor Engineering Harnesses Metabolic Networks to Meet Efficient Production in Cell Factories[J]. China Biotechnology, 2021, 41(4): 55-63.
[5] ZHENG Yi,GUO Shi-ying,SUI Feng-xiang,YANG Qi-yu,WEI Ya-xuan,LI Xiao-yan. Applications of Quorum Sensing Systems in Synthetic Biology[J]. China Biotechnology, 2021, 41(11): 100-109.
[6] CHA Ya-ping, ZHU Mu-zi, LI Shuang. Research Progress on In Vivo Continuous Directed Evolution[J]. China Biotechnology, 2021, 41(1): 42-51.
[7] GUO Er-peng, ZHANG Jian-zhi, SI Tong. Recent Advances in the High-throughput Engineering of Lanthipeptides[J]. China Biotechnology, 2021, 41(1): 30-41.
[8] CHANG Lu, HUANG Jiao-fang, DONG Hao, ZHOU Bin-hui, ZHU Xiao-juan, ZHUANG Ying-ping. A Review on Bioremediation and Detection of Heavy Metal Pollution by Synthetic Biological Engineered Microorganisms and Biofilms[J]. China Biotechnology, 2021, 41(1): 62-71.
[9] RAO Hai-mi,LIANG Dong-mei,LI Wei-guo,QIAO Jian-jun,CAI YIN Qing-ge-le. Advances in Synthetic Biology of Fungal Aromatic Polyketides[J]. China Biotechnology, 2020, 40(9): 52-61.
[10] ZHANG Yu-ting,LI Wei-guo,LIANG Dong-mei,QIAO Jian-jun,CAI YIN Qing-ge-le. Research Progress in Synthetic Biology of P450s in Terpenoid Synthesis[J]. China Biotechnology, 2020, 40(8): 84-96.
[11] WANG Zhen,LI Xia,YUAN Ying-jin. Advances in Production of Caffeic Acid and Its Ester Derivatives in Heterologous Microbes[J]. China Biotechnology, 2020, 40(7): 91-99.
[12] SUN Qing,LIU De-hua,CHEN Zhen. Research Progress of Methanol Utilization and Bioconversion[J]. China Biotechnology, 2020, 40(10): 65-75.
[13] LIU Jin-cong,LIU Xue,YU Hong-jian,ZHAO Guang-rong. Recent Advances in Microbial Production of Phloretin and Its Glycosides[J]. China Biotechnology, 2020, 40(10): 76-84.
[14] Shuo XU,Wen-yu LU. Progress of Heterologous Biosynthesis of Terpenoids in Engineered Corynebacterium glutamicum[J]. China Biotechnology, 2019, 39(6): 91-96.
[15] Hua-ling XIE,Dong-qiao LI,Pei-juan CHI,Yan-ping YANG. An Analysis on the Competition of Patents in Synthetic Biology[J]. China Biotechnology, 2019, 39(4): 114-123.