•  
  •  
 

Bulletin of Chinese Academy of Sciences (Chinese Version)

Keywords

plant-derived natural products; synthetic biology; cell factories; terpenoids

Document Type

Article

Abstract

Plant-derived natural products (PNPs) have been widely used in pharmaceutical and neutracuetical fields. Currently, extracting PNPs from their original plants is the main method to produce them. However, this extracting method is environmentally unfriendly, unsafe, and inefficient. Based on synthetic biology, construction of artificial cell factories for production of PNPs provides an alternative way. In this review, we focus on research progress for microbial production of terpenoids, alkaloids, and phenylpropanoids to give a brief introduction of construction of artificial cell factories for production of PNPs.

First page

1228

Last Page

1238

Language

Chinese

Publisher

Bulletin of Chinese Academy of Sciences

References

程锦锥, 朱恒鹏.中国药品市场报告.北京:社会科学文献出版社, 2012.

刘夺, 张莹, 周晓, 等.合成生物技术生产甾体激素中间体的研究展望.生命科学, 2013(10):958-965.

王冬, 戴住波, 张学礼.酵母人工合成细胞生产植物源天然产物.微生物学报, 2016, 56(3):516-529.

Kuboyama T, Yokoshima S, Tokuyama H, et al. Stereocontrolled total synthesis of (+)-vincristine. PNAS, 2004, 101(33):11966-11970.

Wang P P, Wei Y J, Fan Y, et al. Production of bioactive ginsenosides Rh2 and Rg3 by metabolically engineered yeasts. Metabolic Engineering, 2015, 29:97-105.

Paddon C J, Westfall P J, Pitera D J, et al. High-level semisynthetic production of the potent antimalarial artemisinin. Nature, 2013, 496(7446):528-529.

Dai Z B, Liu Y, Huang L Q, et al. Production of miltiradiene by metabolically engineered Saccharomyces cerevisiae. Biotechnology and Bioengineering, 2012, 109(11):2845-2853.

Xie W P, Lv X M, Ye L D, et al. Construction of lycopene-overproducing Saccharomyces cerevisiae by combining directed evolution and metabolic engineering. Metabolic Engineering, 2015, 30:69-78.

DeLoache W C, Russ Z N, Narcross L, et al. An enzyme-coupled biosensor enables (S)-reticuline production in yeast from glucose. Nature Chemical Biology, 2015, 11(7):465-471.

Farrow S C, Hagel J M, Beaudoin G A W, et al. Stereochemical inversion of (S)-reticuline by a cytochrome P450 fusion in opium poppy. Nature Chemical Biology, 2015, 11(9):728-732.

Fossati E, Narcross L, Ekins A, et al. Synthesis of Morphinan Alkaloids in Saccharomyces cerevisiae. PLoS One, 2015, 10(4):15.

Winzer T, Kern M, King A J, et al. Morphinan biosynthesis in opium poppy requires a P450-oxidoreductase fusion protein. Science, 2015, 349(6245):309-312.

Zhu M, Wang C, Sun W, et al. Boosting 11-oxo-β-amyrin and glycyrrhetinic acid synthesis in Saccharomyces cerevisiae via pairing novel oxidation and reduction system from legume plants. Metabolic Engineering, 2017, 45:43.

Guo J, Zhou Y J, Hillwig M L, et al. CYP76AH1 catalyzes turnover of miltiradiene in tanshinones biosynthesis and enables heterologous production of ferruginol in yeasts. PNAS, 2013, 110(29):12108-12113.

Shang Y, Ma Y S, Zhou Y, et al. Biosynthesis, regulation, and domestication of bitterness in cucumber. Science, 2014, 346(6213):1084-1088.

Itkin M, Davidovich-Rikanati R, Cohen S, et al. The biosynthetic pathway of the nonsugar, high-intensity sweetener mogroside Ⅴ from Siraitia grosvenorii. PNAS, 2016, 113(47):7619-7628.

Lau W, Sattely E S. Six enzymes from mayapple that complete the biosynthetic pathway to the etoposide aglycone. Science, 2015, 349(6253):1224-1228.

Caputi L, Franke J, Farrow S C, et al. Missing enzymes in the biosynthesis of the anticancer drug vinblastine in Madagascar periwinkle. Science, 360(6394):1235-1239.

Zhang X, Li S. Expansion of chemical space for natural products by uncommon P450 reactions. Natural Product Reports, 2017, 34(9):1061-1089.

Du L, Ma L, Qi F, et al. Characterization of a unique pathway for 4-cresol catabolism initiated by phosphorylation in corynebacterium glutamicum. Journal of Biological Chemistry, 2016, 291(12):6583-6594.

Du L, Dong S, Zhang X, et al. Selective oxidation of aliphatic C-H bonds in alkylphenols by a chemomimetic biocatalytic system. PNAS, 2017, 114(26):5129-5137.

Petschacher B, Nidetzky B. Altering the coenzyme preference of xylose reductase to favor utilization of NADH enhances ethanol yield from xylose in a metabolically engineered strain of Saccharomyces cerevisiae. Microbial Cell Factories, 2008, 7:12.

Wang C W, Liao J C. Alteration of product specificity of Rhodobacter sphaeroides phytoene desaturase by directed evolution. Journal of Biological Chemistry, 2001, 276(44):41161-41164.

Dueber J E, Wu G C, Malmirchegini G R, et al. Synthetic protein scaffolds provide modular control over metabolic flux. Nature Biotechnology, 2009, 27(8):753-U107.

Lenihan J R, Tsuruta H, Diola D, et al. Developing an industrial artemisinic acid fermentation process to support the cost-effective production of antimalarial artemisinin-based combination therapies. Biotechnology Progress, 2008, 24(5):1026-1032.

Li J, Zhang Y S. 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.

Liu D, Xiao Y, Evans B S, et al. Negative feedback regulation of fatty acid production based on a malonyl-CoA sensor-actuator. Acs Synthetic Biology, 2015, 4(2):132-140.

Xu P, Wang W, Li L, et al. Design and kinetic analysis of a hybrid promoter-regulator system for malonyl-coa sensing in Escherichia coli. ACS Chemical Biology, 2014, 9(2):451-458.

Farhi M, Marhevka E, Masci T, et al. Harnessing yeast subcellular compartments for the production of plant terpenoids. Metabolic Engineering, 2011, 13(5):474-481.

Thodey K, Galanie S, Smolke C D. A microbial biomanufacturing platform for natural and semisynthetic opioids. Nature Chemical Biology, 2014, 10(10):837-844.

Wu T, Ye L, Zhao D, et al. Membrane engineering-A novel strategy to enhance the production and accumulation of beta-carotene in Escherichia coli. Metabolic Engineering, 2017, 43(Pt A):85-91.

Arendt P, Miettinen K, Pollier J, et al. An endoplasmic reticulum-engineered yeast platform for overproduction of triterpenoids. Metabolic Engineering, 2017, 40:165-175.

Shaw A J, Lam F H, Hamilton M, et al. Metabolic engineering of microbial competitive advantage for industrial fermentation processes. Science, 2016, 353(6299):583-586.

Galanie S, Thodey K, Trenchard I J, et al. Complete biosynthesis of opioids in yeast. Science, 2015, 349(6252):1095-1100.

Ajikumar P K, Xiao W-H, Tyo K E J, et al. Isoprenoid pathway optimization for taxol precursor overproduction in Escherichia coli. Science, 2010, 330(6000):70-74.

Xue Z, Sharpe P L, Hong S P, et al. Production of omega-3 eicosapentaenoic acid by metabolic engineering of Yarrowia lipolytica. Nature Biotechnology, 2013, 31(8):734-740.

Chang M C Y, Eachus R A, Trieu W, et al. Engineering Escherichia coli for production of functionalized terpenoids using plant P450s. Nature Chemical Biology, 2007, 3(5):274-277.

张学礼, 黄璐琦, 戴住波, 等.一种重组菌及其用途: 中国, PCT/CN2017/109029P. 2017-11-02.

Gao W, Hillwig M L, Huang L, et al. A functional genomics approach to tanshinone biosynthesis provides stereochemical insights. Organic Letters, 2009, 11(22):5170-5173.

Zhou Y J, Gao W, Rong Q, et al. Modular pathway engineering of diterpenoid synthases and the mevalonic acid pathway for miltiradiene production. Journal of the American Chemical Society, 2012, 134(6):3234-3241.

Ulschan S, Wolfgang B, Andrea P, et al. Elucidation of the biosynthesis of carnosic acid and its reconstitution in yeast. Nature Communications, 2016, 7:12942.

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, 2016, 26(2):258-261.

Sun Y, Chen Z, Li J, et al. Diterpenoid UDP-Glycosyltransferases from Chinese sweet tea and ashitaba complete the biosynthesis of rubusoside. Mol Plant, 2018, 11(10):1308-1311.

Lee S J, Ji S L, Lee E, et al. The ginsenoside metabolite compound K inhibits hormone-independent breast cancer through downregulation of cyclin D1. Journal of Functional Foods, 2018, 46:159-166.

Liao L M, Zhang Y, Lin S F, et al. Enzymatic transformation from protopanaxadiol ginsenoside Rb1 into rare ginsenoside C-K and its anti-cancer activity. Advanced Materials Research, 2013, 641-642:752-755.

Lee K T, Jung T W, Lee H J, et al. The antidiabetic effect of ginsenoside Rb2 via activation of AMPK. Archives of Pharmacal Research, 2011, 34(7):1201-1208.

Dai Z, Liu Y, Zhang X, et al. Metabolic engineering of Saccharomyces cerevisiae for production of ginsenosides. Metabolic Engineering, 2013, 20(5):146-156.

王冬, 刘怡, 许骄阳, 等.创建酿酒酵母细胞工厂高效生产人参皂苷前体达玛烯二醇Ⅱ.药学学报, 2018, 53(8):1233-1241.

Dai Z, Wang B, Liu Y, et al. Producing aglycons of ginsenosides in bakers' yeast. Scientific Reports, 2014, 4(3698):3698.

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.

Pingping, Wang, Yongjun, et al. Characterization of panax ginseng UDP-Glycosyltransferases catalyzing protopanaxatriol and biosyntheses of bioactive ginsenosides F1 and Rhl in metabolically engineered yeasts. Molecular Plant, 2015, 8(9):1412-1424.

Xue B, Zhang Y I, Jiang H, et al. Effects of maslinic acid on the proliferation and apoptosis of A549 lung cancer cells. Molecular Medicine Reports, 2016, 13(1):117-122.

Miura T, Ueda N, Yamada K, et al. Antidiabetic effects of corosolic acid in KK-Ay diabetic mice. Biological & Pharmaceutical Bulletin, 2006, 29(3):585-587.

Bonte F, Dumas M, Chaudagne C, et al. Influence of asiatic acid, madecassic acid, and asiaticoside on human collagen Ⅰ synthesis. Planta Medica, 1994, 60(2):133-135.

张学礼, 戴住波, 刘芸, 等.三萜2位α-羟化酶MAA45及其相关生物材料与它们在制备山楂酸和科罗索酸中的应用: 中国, CN201610236283.9. 2016-04-14.

Dai Z, Liu Y, Sun Z, et al. Identification of a novel cytochrome P450 enzyme that catalyzes the C-2α hydroxylation of pentacyclic triterpenoids and its application in yeast cell factories. Metabolic Engineering. 2019, 51:70-78.

Zhao J, Li Q, Sun T, et al. Engineering central metabolic modules of Escherichia coli for improving β-carotene production. Metabolic Engineering, 2013, 17(17):42-50.

Li Q, Fan F, Gao X, et al. Balanced activation of IspG and IspH to eliminate MEP intermediate accumulation and improve isoprenoids production in Escherichia coli. Metabolic Engineering, 2017, 44:13-21.

Bai Y, Yin H, Bi H, et al. De novo biosynthesis of gastrodin in Escherichia coli. Metabolic Engineering, 2016, 35:138-147.

钟贝芬, 杜磊, 李众, 等.基于细胞色素P450单加氧酶介导的4-甲酚氧化降解途径的天麻素生物合成.湖南师范大学自然科学学报, 2018, 41(4):33-40.

Liu X, Cheng J, Zhang G, et al. Engineering yeast for the production of breviscapine by genomic analysis and synthetic biology approaches. Nature Communications, 2018, 9(1):448.

Yao Y F, Wang C S, Qiao J, et al. Metabolic engineering of Escherichia coli for production of salvianic acid A via an artificial biosynthetic pathway. Metabolic Engineering, 2013, 19(5):79-87.

Share

COinS