전체메뉴
검색
Article Search

JMB Journal of Microbiolog and Biotechnology

QR Code QR Code

Review


References

  1. Zhu L, Chen L. 2019. Progress in research on paclitaxel and tumor immunotherapy. Cell. Mol. Biol. Lett. 24: 40.
    Pubmed PMC CrossRef
  2. Li ZM, Xu SW, Liu PQ. 2018. Salvia miltiorrhizaBurge (Danshen): a golden herbal medicine in cardiovascular therapeutics. Acta Pharm. Sinica 39: 802-824.
    Pubmed PMC CrossRef
  3. Yaro P, Nie J, Xu M, Zeng K, He H, Yao J, et al. 2019. Influence of organic anion transporter 1/3 on the pharmacokinetics and renal excretion of ginkgolides and bilobalide. J. Ethnopharmacol. 243: 112098.
    Pubmed CrossRef
  4. Arumugam B, Subramaniam A, Alagaraj P. 2020. Stevia as a natural sweetener: A review. Cardiovasc. Hematol. Agents Med. Chem. 18: 94-103.
    Pubmed CrossRef
  5. Rizza A, Jones AM. 2019. The makings of a gradient: spatiotemporal distribution of gibberellins in plant development. Curr. Opin. Plant Biol. 47: 9-15.
    Pubmed PMC CrossRef
  6. Guo X, Zhang RR, Sun JY, Liu Y, Yuan XS, Chen YY, et al. 2024. The molecular mechanism of action for the potent antitumor component extracted using supercritical fluid extraction from Croton crassifolius root. J. Ethnopharmacol. 327: 117835.
    Pubmed CrossRef
  7. Zhu H, Ren X, Huang Y, Su T, Yang L. 2023. Chemical constituents of Euphorbia stracheyi boiss (Euphorbiaceae). Metabolites 13: 852.
    Pubmed PMC CrossRef
  8. Ren X, Yuan X, Jiao SS, He XP, Hu H, Kang JJ, et al. 2023. Clerodane diterpenoids from the Uygur medicine Salvia deserta with immunosuppressive activity. Phytochemistry 214: 113823.
    Pubmed CrossRef
  9. Ajikumar PK, Xiao WH, Tyo KE, Wang Y, Simeon F, Leonard E, et al. 2010. Isoprenoid pathway optimization for Taxol precursor overproduction in Escherichia coli. Science 330: 70-74.
    Pubmed PMC CrossRef
  10. Zhang P, Lee Y, Wei X, Wu J, Liu Q, Wan S. 2018. Enhanced production of tanshinone IIA in endophytic fungi Emericella foeniculicola by genome shuffling. Pharm. Biol. 56: 357-362.
    Pubmed PMC CrossRef
  11. Moon JH, Lee K, Lee JH, Lee PC. 2020. Redesign and reconstruction of a steviol-biosynthetic pathway for enhanced production of steviol in Escherichia coli. Microb. Cell Fact. 19: 20.
    Pubmed PMC CrossRef
  12. Gutbrod K, Romer J, Dörmann P. 2019. Phytol metabolism in plants. Progress Lipid Res. 74: 1-17.
    Pubmed CrossRef
  13. Lin YP, Charng YY. 2021. Chlorophyll dephytylation in chlorophyll metabolism: a simple reaction catalyzed by various enzymes. Plant Sci. 302: 110682.
    Pubmed CrossRef
  14. Xue S, Zhang P, Tang P, Wang C, Kong L, Luo J. 2020. Acyclic diterpene and norsesquiterpene from the seed of Aphanamixis polystachya. Fitoterapia 142: 104518.
    Pubmed CrossRef
  15. Ibrahim SRM, Mohamed GAA. 2017. Tagetones A and B, new cytotoxic monocyclic diterpenoids from flowers of Tagetes minuta. Chin. J. Nat. Med. 15: 546-549.
    Pubmed CrossRef
  16. Zhang J, Chen S, Sun P, Liu Y, Jiang J, Guo C, et al. 2024. Ginkgolides with anti-PAF activity from Ginkgo biloba L. Fitoterapia 175: 105915-105915.
    Pubmed CrossRef
  17. Yu Y, Wang Y, Wang GC, Tan CY, Wang Y, Liu JS, et al. 2023. Andropanilides A-C, the novel labdane-type diterpenoids from Andrographis paniculata and their anti-inflammation activity. Nat. Prod. Bioprospect. 13: 31.
    Pubmed PMC CrossRef
  18. Thekkeveedu RP, Devasya RP. 2024. Biotechnological interventions for the production of forskolin, an active compound from the medicinal plant, Coleus forskohlii. Physiol. Mol. Biol. Plants 30: 213-226.
    Pubmed CrossRef
  19. Li R, Morris-Natschke SL, Lee KH. 2016. Clerodane diterpenes: sources, structures, and biological activities. Nat. Prod. Rep. 33: 1166-1226.
    Pubmed PMC CrossRef
  20. Wang M, Ma C, Chen Y, Li X, Chen J. 2019. Cytotoxic neo-clerodane diterpenoids from Scutellaria barbata D.Don. Chem. Biodivers. 16: e1800499.
    Pubmed CrossRef
  21. Ledoux A, Hamann C, Bonnet O, Jullien K, Quetin-Leclercq J, Tchinda A, et al. 2023. Bioactive clerodane diterpenoids from the leaves of Casearia coriacea vent. Molecules 28: 1197.
    Pubmed PMC CrossRef
  22. Zhang C, Liu JX, Sun MQ, Mian L, Sun L. 2018. Study on fragmentation pathways of three ginkgolides by mass spectrometry. China J. Chin. Mater. Medica 43: 4093-4096.
  23. Liao HJ, Zheng YF, Hong-Yang LI, Peng GP. 2012. Study on antinplatelet aggregation activity induced by PAF of Ginkgolide compounds. J. Liaoning University of Traditional Chinese Medicine.
  24. Hao M, Xu J, Wen H, Du J, Zhang S, Lv M, et al. 2022. Recent advances on biological activities and structural modifications of dehydroabietic acid. Toxins 14: 632.
    Pubmed PMC CrossRef
  25. González MA, Correa-Royero J, Agudelo L, Mesa A, Betancur-Galvis L. 2009. Synthesis and biological evaluation of abietic acid derivatives. Eur. J. Med. Chem. 44: 2468-2472.
    Pubmed CrossRef
  26. Xu J, Zhi X, Zhang Y, Ding R. 2024. Tanshinone IIA alleviates IL-1β-induced chondrocyte apoptosis and inflammation by regulating FBXO11 expression. Clinics 79: 100365.
    Pubmed PMC CrossRef
  27. Habtemariam S. 2023. Anti-inflammatory therapeutic mechanisms of natural products: Insight from rosemary diterpenes, carnosic acid and carnosol. Biomedicines 11: 545.
    Pubmed PMC CrossRef
  28. Yin XW, Bian XX, Chen YF, Zhang M, Wu L, Ren FC, et al. 2023. Structure-diversified terpenoids from Salvia prattii and their protective activity against alcoholic liver diseases. Phytochemistry 214: 113819.
    Pubmed CrossRef
  29. Chen W, Li X, Guo S, Song N, Wang J, Jia L, et al. 2019. Tanshinone IIA harmonizes the crosstalk of autophagy and polarization in macrophages via miR-375/KLF4 pathway to attenuate atherosclerosis. Int. Immunopharmacol. 70: 486-497.
    Pubmed CrossRef
  30. Guan Z, Chen J, Li X, Dong N. 2020. Tanshinone IIA induces ferroptosis in gastric cancer cells through p53-mediated SLC7A11 down-regulation. Biosci. Rep. 40: BSR20201807.
    Pubmed PMC CrossRef
  31. Sun M, Guo B, Xu M, Zhao M, Onakpa MM, Wu Z, et al. 2021. (9βH)- and 17-Nor-Pimaranes from Icacina oliviformis. J. Nat. Prod. 84: 949-955.
    Pubmed PMC CrossRef
  32. Jongsomjainuk O, Boonsombat J, Thongnest S, Prawat H, Batsomboon P, Charoensutthivarakul S, et al. 2023. Kaemtakols A-D, highly oxidized pimarane diterpenoids with potent anti-inflammatory activity from Kaempferia takensis. Nat. Prod. Bioprospect. 13: 55.
    Pubmed PMC CrossRef
  33. Kim C, Le D, Lee M. 2021. Diterpenoids isolated from Podocarpus macrophyllus inhibited the inflammatory mediators in LPSinduced HT-29 and RAW 264.7 cells. Molecules (Basel, Switzerland) 26: 4326.
    Pubmed PMC CrossRef
  34. Zhao X, Cacherat B, Hu Q, Ma D. 2022. Recent advances in the synthesis of ent-kaurane diterpenoids. Nat. Prod. Rep. 39: 119-138.
    Pubmed CrossRef
  35. Liu M, Wang WG, Sun HD, Pu JX. 2017. Diterpenoids from Isodon species: an update. Nat. Prod. Rep. 34: 1090-1140.
    Pubmed CrossRef
  36. Camara MC, Vandenberghe LPS, Rodrigues C, de Oliveira J, Faulds C, Bertrand E, et al. 2018. Current advances in gibberellic acid (GA(3)) production, patented technologies and potential applications. Planta 248: 1049-1062.
    Pubmed CrossRef
  37. Binenbaum J, Weinstain R, Shani E. 2018. Gibberellin localization and transport in plants. Trends Plant Sci. 23: 410-421.
    Pubmed CrossRef
  38. Kildegaard KR, Arnesen JA, Adiego-Pérez B, Rago D, Kristensen M, Klitgaard AK, et al. 2021. Tailored biosynthesis of gibberellin plant hormones in yeast. Metab. Eng. 66: 1-11.
    Pubmed PMC CrossRef
  39. Drummond GJ, Grant PS, Brimble MA. 2021. ent-Atisane diterpenoids: isolation, structure and bioactivity. Nat. Prod. Rep. 38: 330-345.
    Pubmed CrossRef
  40. Wang B, Wei Y, Zhao X, Tian X, Ning J, Zhang B, et al. 2018. Unusual ent-atisane type diterpenoids with 2-oxopropyl skeleton from the roots of Euphorbia ebracteolata and their antiviral activity against human rhinovirus 3 and enterovirus 71. Bioorg. Chem. 81: 234-240.
    Pubmed CrossRef
  41. Maji D, Barnawal D, Gupta A, King S, Singh AK, Kalra A. 2013. A natural plant growth promoter calliterpenone from a plant Callicarpa macrophylla Vahl improves the plant growth promoting effects of plant growth promoting rhizobacteria (PGPRs). World J. Microbiol. Biotechnol. 29: 833-839.
    Pubmed CrossRef
  42. Li CH, Zhang JY, Zhang XY, Li SH, Gao JM. 2019. An overview of grayanane diterpenoids and their biological activities from the Ericaceae family in the last seven years. Eur. J. Med. Chem. 166: 400-416.
    Pubmed CrossRef
  43. Sun N, Zheng G, He M, Feng Y, Liu J, Wang M, et al. 2019. Grayanane diterpenoids from the leaves of Rhododendron auriculatum and their analgesic activities. J. Nat. Prod. 82: 1849-1860.
    Pubmed CrossRef
  44. Leonelli F, Migneco LM, Valletta A, Marini Bettolo R. 2021. Stemodane diterpenes and diterpenoids: Isolation, structure elucidation, biogenesis, biosynthesis, biological activity, biotransformations, metabolites and derivatives biological activity, rearrangements. Molecules 26: 2761.
    Pubmed PMC CrossRef
  45. Leonelli F, Valletta A, Migneco LM, Marini Bettolo R. 2019. Stemarane diterpenes and diterpenoids. Int. J. Mol. Sci. 20: 2627.
    Pubmed PMC CrossRef
  46. Schneider F, Pan L, Ottenbruch M, List T, Gaich T. 2021. The Chemistry of Nonclassical Taxane Diterpene. Acc. Chem. Res. 54: 2347-2360.
    Pubmed CrossRef
  47. Wani MC, Taylor HL, Wall ME, Coggon P, McPhail AT. 1971. Plant antitumor agents. VI. The isolation and structure of taxol, a novel antileukemic and antitumor agent from Taxus brevifolia. J. Am. Chem. Soc. 93: 2325-2327.
    Pubmed CrossRef
  48. Gallego-Jara J, Lozano-Terol G, Sola-Martínez RA, Cánovas-Díaz M, de Diego Puente T. 2020. A compressive review about Taxol(R): History and future challenges. Molecules 25: 5986.
    Pubmed PMC CrossRef
  49. Bouchet BP, Galmarini CM. 2010. Cabazitaxel, a new taxane with favorable properties. Drugs Today 46: 735-742.
    Pubmed CrossRef
  50. Singer JW. 2005. Paclitaxel poliglumex (XYOTAX, CT-2103): a macromolecular taxane. J. Control. Release 109: 120-126.
    Pubmed CrossRef
  51. Abbas MG, Muhammad R, Syed S-u-H, Zafar PR, Zafar PU. 2015. Prostratin: An overview. Mini Rev. Med. Chem. 15: 1122-1130.
    Pubmed CrossRef
  52. Hou P, Zeng Y, Ma B, Bi K, Chen X. 2013. A new cytotoxic cembrane diterpene from the roots of Euphorbia pekinensis Rupr. Fitoterapia 90: 10-13.
    Pubmed CrossRef
  53. Wang K, Yu H, Wu H, Wang X, Pan Y, Chen Y, et al. 2015. A new casbane diterpene from Euphorbia pekinensis. Nat. Prod. Res. 29: 1456-1460.
    Pubmed CrossRef
  54. Fattahian M, Ghanadian M, Ali Z, Khan IA. 2020. Jatrophane and rearranged jatrophane-type diterpenes: biogenesis, structure, isolation, biological activity and SARs (1984-2019). Phytochem. Rev. 19: 265-336.
    Pubmed PMC CrossRef
  55. Wang YF, Su XH, Li LG, Wang W, Zhang ML, Huo CH, et al. 2009. Verticillane derivatives from natural sources. Chem. Biodiver. 6: 1661-1673.
    Pubmed CrossRef
  56. Bai X, Qu H, Zhang J, Li L, Zhang C, Li S, et al. 2024. Effect of steviol glycosides as natural sweeteners on glucose metabolism in adult participants. Food Funct. 15: 2908-3919.
    Pubmed CrossRef
  57. Roy S, Ghosh A, Majie A, Karmakar V, Das S, Dinda SC, et al. 2024. Terpenoids as potential phytoconstituent in the treatment of diabetes: From preclinical to clinical advancement. Phytomedicine 129: 155638.
    Pubmed CrossRef
  58. Grodsky L, Wilson M, Rathinasabapathy T, Komarnytsky S. 2024. Triptolide administration alters immune responses to mitigate insulin resistance in obese states. Biomolecules 14: 395.
    Pubmed PMC CrossRef
  59. Bidart JE, Pertino MW, Schmeda-Hirschmann G, Alché LE, Petrera E. 2023. Antiviral effect of natural and semisynthetic diterpenoids against adenovirus infection in vitro. Planta Med. 89: 1001-1009.
    Pubmed CrossRef
  60. Sadeghi Z, Cerulli A, Marzocco S, Moridi Farimani M, Masullo M, Piacente S. 2023. Anti-inflammatory activity of tanshinonerelated diterpenes from Perovskia artemisioides roots. J. Nat. Prod. 86: 812-821.
    Pubmed PMC CrossRef
  61. Tamuli R, Nguyen T, Macdonald JR, Pierens GK, Fisher GM, Andrews KT, et al. 2023. Isolation and in vitro and in vivo activity of secondary metabolites from Clerodendrum polycephalum baker against Plasmodium Malaria parasites. J. Nat. Prod. 86: 2661-2671.
    Pubmed CrossRef
  62. Deng QD, Lei XP, Zhong YH, Chen MS, Ke YY, Li Z, et al. 2021. Triptolide suppresses the growth and metastasis of non-small cell lung cancer by inhibiting β-catenin-mediated epithelial-mesenchymal transition. Acta Pharmacol. Sinica 42: 1486-1497.
    Pubmed PMC CrossRef
  63. Sarwar MS, Xia YX, Liang ZM, Tsang SW, Zhang HJ. 2020. Mechanistic pathways and molecular targets of plant-derived anticancer ent-Kaurane diterpenes. Biomolecules 10: 144.
    Pubmed PMC CrossRef
  64. Bailly C. 2023. Yuexiandajisu diterpenoids from Euphorbia ebracteolata Hayata (Langdu roots): An overview. Phytochemistry 213: 113784.
    Pubmed CrossRef
  65. Wang X, Wang Y, Liu Z, Zhao H, Yao GD, Liu Q, et al. 2024. New daphnane diterpenoidal 1,3,4-oxdiazole derivatives as potential anti-hepatoma agents: synthesis, biological evaluation and molecular modeling studies. Bioorg. Chem. 145: 107208.
    Pubmed CrossRef
  66. Yazdiniapour Z, Sohrabi MH, Motinia N, Zolfaghari B, Mehdifar P, Ghanadian M, et al. 2023. Diterpenoids from Euphorbia gedrosiaca as potential anti-proliferative agents against breast cancer cells. Metabolites 13: 225.
    Pubmed PMC CrossRef
  67. Wang X, Yang Y, Liu X, Gao X. 2020. Pharmacological properties of tanshinones, the natural products from Salvia miltiorrhiza. Adv. Pharmacol. 87: 43-70.
    CrossRef
  68. Wang W, Wu Y, Chen X, Zhang P, Li H, Chen L. 2019. Synthesis of new ent-labdane diterpene derivatives from andrographolide and evaluation of their anti-inflammatory activities. Eur. J. Med. Chem. 162: 70-79.
    Pubmed CrossRef
  69. Wang Y, Song Z, Guo Y, Xie H, Zhang Z, Sun D, et al. 2021. Diterpenoids from the seeds of Euphorbia lathyris and their antiinflammatory activity. Bioorg. Chem. 112: 104944.
    Pubmed CrossRef
  70. Kim J, Nguyen TTH, Jin J, Septiana I, Son GM, Lee GH, et al. 2019. Anti-cariogenic characteristics of rubusoside. Biotechnol. Bioprocess Eng. 24: 282-287.
    Pubmed PMC CrossRef
  71. Siddique H, Pendry B, Rahman MM. 2019. Terpenes from Zingiber montanum and heir Screening against Multi-Drug Resistant and Methicillin Resistant Staphylococcus aureus. Molecules 24: 385.
    Pubmed PMC CrossRef
  72. Wardana AP, Aminah NS, Rosyda M, Abdjan MI, Kristanti AN, Tun KNW, et al. 2021. Potential of diterpene compounds as antivirals, a review. Heliyon 7: e07777.
    Pubmed PMC CrossRef
  73. Wu Z, Xu H, Xu Y, Fan W, Yao H, Wang Y, et al. 2020. Andrographolide promotes skeletal muscle regeneration after acute injury through epigenetic modulation. Eur. J. Pharmacol. 888: 173470.
    Pubmed CrossRef
  74. Zhang F, Koh GY, Hollingsworth J, Russo PS, Stout RW, Liu Z. 2012. Reformulation of etoposide with solubility-enhancing rubusoside. Int. J. Pharm. 434: 453-459.
    Pubmed CrossRef
  75. Zhang M, Dai T, Feng N. 2017. A novel solubility-enhanced rubusoside-based micelles for increased cancer therapy. Nanoscale Res. Llett. 12: 274.
    Pubmed PMC CrossRef
  76. Shi JB, Wang N, Zhou H, Xu QH, Yan GT. 2019. The role of gibberellin synthase gene GhGA2ox1 in upland cotton (Gossypium hirsutum L.) responses to drought and salt stress. Biotechnol. Appl. Biochem. 66: 298-308.
    Pubmed CrossRef
  77. Li J, Meng X, Yin C, Zhang L, Lin B, Liu P, et al. 2023. Antimalarial and neuroprotective ent-abietane diterpenoids from the aerial parts of Phlogacanthus curviflorus. Chin. J. Nat. Med. 21: 619-630.
    Pubmed CrossRef
  78. Mirza FJ, Zahid S, Holsinger RMD. 2023. Neuroprotective effects of carnosic acid: Insight into its mechanisms of action. Molecules 28: 2306.
    Pubmed PMC CrossRef
  79. Vranová E, Coman D, Gruissem W. 2013. Network analysis of the MVA and MEP pathways for isoprenoid synthesis. Ann. Rev. Plant Biol. 64: 665-700.
    Pubmed CrossRef
  80. Bergman ME, Davis B, Phillips MA. 2019. Medically useful plant terpenoids: biosynthesis, occurrence, and mechanism of action. Molecules 24: 3961.
    Pubmed PMC CrossRef
  81. Zerbe P, Hamberger B, Yuen MM, Chiang A, Sandhu HK, Madilao LL, et al. 2013. Gene discovery of modular diterpene metabolism in nonmodel systems. Plant Physiol. 162: 1073-1091.
    Pubmed PMC CrossRef
  82. Pateraki I, Andersen-Ranberg J, Hamberger B, Heskes AM, Martens HJ, Zerbe P, et al. 2014. Manoyl oxide (13R), the biosynthetic precursor of forskolin, is synthesized in specialized root cork cells in Coleus forskohlii. Plant Physiol. 164: 1222-1236.
    Pubmed PMC CrossRef
  83. Pateraki I, Andersen-Ranberg J, Jensen NB, Wubshet SG, Heskes AM, Forman V, et al. 2017. Total biosynthesis of the cyclic AMP booster forskolin from Coleus forskohlii. ELife 6: e23001.
    Pubmed PMC CrossRef
  84. Mastan A, Rane D, Dastager SG, Vivek Babu CS. 2020. Plant probiotic bacterial endophyte, Alcaligenes faecalis, modulates plant growth and forskolin biosynthesis in Coleus forskohlii. Probiotics Antimicrob. Proteins 12: 481-493.
    Pubmed CrossRef
  85. Gao W, Hillwig ML, Huang L, Cui G, Wang X, Kong J, et al. 2009. A functional genomics approach to tanshinone biosynthesis provides stereochemical insights. Org. Lett. 11: 5170-5173.
    Pubmed PMC CrossRef
  86. Guo J, Zhou YJ, Hillwig ML, Shen Y, Yang L, Wang Y, et al. 2013. CYP76AH1 catalyzes turnover of miltiradiene in tanshinones biosynthesis and enables heterologous production of ferruginol in yeasts. Proc. Natl. Acad. Sci. USA 110: 12108-12113.
    Pubmed PMC CrossRef
  87. Guo J, Ma X, Cai Y, Ma Y, Zhan Z, Zhou YJ, et al. 2016. Cytochrome P450 promiscuity leads to a bifurcating biosynthetic pathway for tanshinones. New Phytol. 210: 525-534.
    Pubmed PMC CrossRef
  88. Mao Y, Ma Y, Chen T, Ma X, Xu Y, Bu J, et al. 2020. Functional integration of two CYP450 genes involved in biosynthesis of tanshinones for improved diterpenoid production by synthetic biology. ACS Synth. Biol. 9: 1763-1770.
    Pubmed CrossRef
  89. Ma Y, Cui G, Chen T, Ma X, Wang R, Jin B, et al. 2021. Expansion within the CYP71D subfamily drives the heterocyclization of tanshinones synthesis in Salvia miltiorrhiza. Nat. Commun. 12: 685.
    Pubmed PMC CrossRef
  90. Deng C, Hao X, Shi M, Fu R, Wang Y, Zhang Y, et al. 2019. Tanshinone production could be increased by the expression of SmWRKY2 in Salvia miltiorrhiza hairy roots. Plant Sci. 284: 1-8.
    Pubmed CrossRef
  91. Zheng H, Jing L, Jiang X, Pu C, Zhao S, Yang J, et al. 2021. The ERF-VII transcription factor SmERF73 coordinately regulates tanshinone biosynthesis in response to stress elicitors in Salvia miltiorrhiza. New Phytol. 231: 1940-1955.
    Pubmed CrossRef
  92. Lv X, Zhang W, Chu S, Zhang H, Wu Y, Zhu Y, et al. 2024. Endophytic fungus Penicillium steckii DF33 promoted tanshinones biosynthesis in Salvia miltiorrhiza by regulating the expression of CYP450 genes. Gene 899: 148094.
    Pubmed CrossRef
  93. Li X, Lin Y, Qin Y, Han G, Wang H, Yan Z. 2024. Beneficial endophytic fungi improve the yield and quality of Salvia miltiorrhiza by performing different ecological functions. PeerJ. 12: e16959.
    Pubmed PMC CrossRef
  94. Xu Y, Wang X, Zhang C, Zhou X, Xu X, Han L, et al. 2022. De novo biosynthesis of rubusoside and rebaudiosides in engineered yeasts. Nat. Commun. 13: 3040.
    Pubmed PMC CrossRef
  95. Wang Y, Sun X, Jia X, Zhu L, Yin H. 2021. Comparative transcriptomic of Stevia rebaudiana provides insight into rebaudioside D and rebaudioside M biosynthesis. Plant Physiol. Biochem. PPB. 167: 541-549.
    Pubmed CrossRef
  96. Zhang J, Tang M, Chen Y, Ke D, Zhou J, Xu X, et al. 2021. Catalytic flexibility of rice glycosyltransferase OsUGT91C1 for the production of palatable steviol glycosides. Nat. Commun. 12: 7030.
    Pubmed PMC CrossRef
  97. Dewitte G, Walmagh M, Diricks M, Lepak A, Gutmann A, Nidetzky B, et al. 2016. Screening of recombinant glycosyltransferases reveals the broad acceptor specificity of stevia UGT-76G1. J. Biotechnol. 233: 49-55.
    Pubmed CrossRef
  98. Chau M, Croteau R. 2004. Molecular cloning and characterization of a cytochrome P450 taxoid 2alpha-hydroxylase involved in Taxol biosynthesis. Arch. Biochem. Biophys. 427: 48-57.
    Pubmed CrossRef
  99. Chau M, Jennewein S, Walker K, Croteau R. 2004. Taxol biosynthesis: Molecular cloning and characterization of a cytochrome P450 taxoid 7 beta-hydroxylase. Chem. Biol. 11: 663-672.
    CrossRef
  100. Schoendorf A, Rithner CD, Williams RM, Croteau RB. 2001. Molecular cloning of a cytochrome P450 taxane 10 beta-hydroxylase cDNA from Taxus and functional expression in yeast. Proc. Natl. Acad. Sci. USA 98: 1501-1506.
    Pubmed PMC CrossRef
  101. Jennewein S, Long RM, Williams RM, Croteau R. 2004. Cytochrome P450 taxadiene 5alpha-hydroxylase, a mechanistically unusual monooxygenase catalyzing the first oxygenation step of taxol biosynthesis. Chem. Biol. 11: 379-387.
    Pubmed CrossRef
  102. Jennewein S, Rithner CD, Williams RM, Croteau RB. 2001. Taxol biosynthesis: taxane 13 alpha-hydroxylase is a cytochrome P450-dependent monooxygenase. Proc. Natl. Acad. Sci. USA 98: 13595-13600.
    Pubmed PMC CrossRef
  103. Walker K, Schoendorf A, Croteau R. 2000. Molecular cloning of a taxa-4(20),11(12)-dien-5alpha-ol-O-acetyl transferase cDNA from Taxus and functional expression in Escherichia coli. Arch. Biochem. Biophys. 374: 371-380.
    Pubmed CrossRef
  104. Walker K, Croteau R. 2000. Molecular cloning of a 10-deacetylbaccatin III-10-O-acetyl transferase cDNA from Taxus and functional expression in Escherichia coli. Proc. Natl. Acad. Sci. USA 97: 583-587.
    Pubmed PMC CrossRef
  105. Walker K, Croteau R. 2000. Taxol biosynthesis: molecular cloning of a benzoyl-CoA:taxane 2alpha-O-benzoyltransferase cDNA from Taxus and functional expression in Escherichia coli. Proc. Natl. Acad. Sci. USA 97: 13591-13596.
    Pubmed PMC CrossRef
  106. Bartsch S, Wybenga GG, Jansen M, Heberling MM, Wu B, Dijkstra BW, et al. 2013. Redesign of a phenylalanine aminomutase into a phenylalanine ammonia lyase. ChemCatChem 5: 1797-1802.
    CrossRef
  107. Walker KD, Klettke K, Akiyama T, Croteau R. 2004. Cloning, heterologous expression, and characterization of a phenylalanine aminomutase involved in Taxol biosynthesis. J. Biol. Chem. 279: 53947-53954.
    Pubmed CrossRef
  108. Walker K, Fujisaki S, Long R, Croteau R. 2002. Molecular cloning and heterologous expression of the C-13 phenylpropanoid side chain-CoA acyltransferase that functions in Taxol biosynthesis. Proc. Natl. Acad. Sci. USA 99: 12715-12720.
    Pubmed PMC CrossRef
  109. Walker K, Long R, Croteau R. 2002. The final acylation step in taxol biosynthesis: cloning of the taxoid C13-side-chain Nbenzoyltransferase from Taxus. Proc. Natl. Acad. Sci. USA 99: 9166-9171.
    Pubmed PMC CrossRef
  110. Zhang Y, Wiese L, Fang H, Alseekh S, Perez de Souza L, Scossa F, et al. 2023. Synthetic biology identifies the minimal gene set required for paclitaxel biosynthesis in a plant chassis. Mol. Plant 16: 1951-1961.
    Pubmed CrossRef
  111. Paddon CJ, Westfall PJ, Pitera DJ, Benjamin K, Fisher K, McPhee D, et al. 2013. High-level semi-synthetic production of the potent antimalarial artemisinin. Nature 496: 528-532.
    Pubmed CrossRef
  112. Dai Z, Wang B, Liu Y, Shi M, Wang D, Zhang X, et al. 2014. Producing aglycons of ginsenosides in bakers' yeast. Sci. Rep. 4: 3698.
    Pubmed PMC CrossRef
  113. Zhang B, Yao Z, Liu Z, Zheng Y. 2021. Metabolic engineering of Escherichia coli for L-homoserine production. Sheng wu gong cheng xue bao Chin. J. Biotechnol. 37: 1287-1297.
  114. Kong MK, Kang HJ, Kim JH, Oh SH, Lee PC. 2015. Metabolic engineering of the Stevia rebaudiana ent-kaurene biosynthetic pathway in recombinant Escherichia coli. J. Biotechnol. 214: 95-102.
    Pubmed CrossRef
  115. Sun Y, Chen Z, Wang G, Lv H, Mao Y, Ma K, et al. 2022. De novo production of versatile oxidized kaurene diterpenes in Escherichia coli. Metab. Eng. 73: 201-213.
    Pubmed CrossRef
  116. Dong H, Chen S, Zhu J, Gao K, Zha W, Lin P, et al. 2020. Enhance production of diterpenoids in yeast by overexpression of the fused enzyme of ERG20 and its mutant mERG20. J. Biotechnol. 307: 29-34.
    Pubmed CrossRef
  117. Hu T, Zhou J, Tong Y, Su P, Li X, Liu Y, et al. 2020. Engineering chimeric diterpene synthases and isoprenoid biosynthetic pathways enables high-level production of miltiradiene in yeast. Metab. Eng. 60: 87-96.
    Pubmed CrossRef
  118. Forman V, Bjerg-Jensen N, Dyekjær JD, Møller BL, Pateraki I. 2018. Engineering of CYP76AH15 can improve activity and specificity towards forskolin biosynthesis in yeast. Microb. Cell Fact. 17: 181.
    Pubmed PMC CrossRef
  119. Ju H, Zhang C, He S, Nan W, Lu W. 2022. Construction and optimization of Saccharomyces cerevisiae for synthesizing forskolin. Appl. Microbiol. Biotechnol. 106: 1933-1944.
    Pubmed CrossRef
  120. Wei P, Zhang C, Bian X, Lu W. 2022. Metabolic engineering of Saccharomyces cerevisiae for heterologous carnosic acid production. Front. Bioeng. Biotechnol. 10: 916605.
    Pubmed PMC CrossRef
  121. Einhaus A, Steube J, Freudenberg RA, Barczyk J, Baier T, Kruse O. 2022. Engineering a powerful green cell factory for robust photoautotrophic diterpenoid production. Metab. Eng. 73: 82-90.
    Pubmed CrossRef
  122. Khalid A, Takagi H, Panthee S, Muroi M, Chappell J, Osada H, et al. 2017. Development of a terpenoid-production platform in Streptomyces reveromyceticus SN-593. ACS Synth. Biol. 6: 2339-2349.
    Pubmed CrossRef
  123. Yamada Y, Arima S, Nagamitsu T, Johmoto K, Uekusa H, Eguchi T, et al. 2015. Novel terpenes generated by heterologous expression of bacterial terpene synthase genes in an engineered Streptomyces host. J. Antibiotics 68: 385-394.
    Pubmed PMC CrossRef
  124. Wang J, Li S, Xiong Z, Wang Y. 2015. Pathway mining-based integration of critical enzyme parts for de novo biosynthesis of steviolglycosides sweetener in Escherichia coli. Cell Res. 26: 258-261.
    Pubmed PMC CrossRef
  125. Xuan C, Wei Y, Yu C, Shan Y, K ZZ, Jens N, et al. 2022. Engineering yeast for high-level production of diterpenoid sclareol. Metab. Eng. 75: 19-28.
    Pubmed CrossRef
  126. Norkin M, Ordóñez-Morán P, Huelsken J. 2021. High-content, targeted RNA-seq screening in organoids for drug discovery in colorectal cancer. Cell Rep. 35: 109026.
    Pubmed CrossRef
  127. Dong Y, Zhao Q, Wang Y. 2021. Network pharmacology-based investigation of potential targets of astragalus membranaceousangelica sinensis compound acting on diabetic nephropathy. Sci. Rep. 11: 19496.
    Pubmed PMC CrossRef
  128. Murthy BN, Sinha S, Surolia A, Indi SS, Jayaraman N. 2008. SPR and ITC determination of the kinetics and the thermodynamics of bivalent versus monovalent sugar ligand-lectin interactions. Glycoconj. J. 25: 313-321.
    Pubmed CrossRef
  129. Ferreira LG, Dos Santos RN, Oliva G, Andricopulo AD. 2015. Molecular docking and structure-based drug design strategies. Molecules 20: 13384-13421.
    Pubmed PMC CrossRef
  130. Xu X, Yuan H, Yu X, Huang S, Sun Y, Zhang T, et al. 2021. The chromosome-level Stevia genome provides insights into steviol glycoside biosynthesis. Hortic. Res. 8: 129.
    Pubmed PMC CrossRef

Related articles in JMB

More Related Articles

Article

Review

J. Microbiol. Biotechnol. 2024; 34(8): 1563-1579

Published online August 28, 2024 https://doi.org/10.4014/jmb.2402.02014

Copyright © The Korean Society for Microbiology and Biotechnology.

Advances in the Structures, Pharmacological Activities, and Biosynthesis of Plant Diterpenoids

Leilei Li1, Jia Fu2, and Nan Liu1*

1School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
2State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China

Correspondence to:Liu Nan,       liunanlnn@tjutcm.edu.cn

Received: February 8, 2024; Revised: May 25, 2024; Accepted: May 28, 2024

Abstract

More and more diterpenoids have attracted extensive attention due to the diverse chemical structures and excellent biological activities, and have been developed into clinical drugs or consumer products. The vast majority of diterpenoids are derived from plants. With the long-term development of plant medicinal materials, the natural resources of many plant diterpenoids are decreasing, and the biosynthetic mechanism of key active components has increasingly become a research hotspot. Using synthetic biology to engineer microorganisms into "cell factories" to produce the desired compounds is an essential means to solve these problems. In this review, we depict the plant-derived diterpenoids from chemical structure, biological activities, and biosynthetic pathways. We use representative plant diterpenes as examples to expound the research progress on their biosynthesis, and summarize the heterologous production of plant diterpenoids in microorganisms in recent years, hoping to lay the foundation for the development and application of plant diterpenoids in the future.

Keywords: Plant diterpenoids, chemical structures, pharmacological activities, biosynthetic pathways, synthetic biology, heterologous production

Introduction

Diterpenes are widely distributed in plants, fungi, and marine organisms. The backbones of diterpenes are composed of four isoprene units that join head-to-tail. The complexity and diversity of diterpenoids make them rich in biological activities. Several plant diterpenes have been developed into clinical drugs to treat various diseases. For example, paclitaxel from Taxus is currently used for treating ovarian and breast cancer in clinical practice [1]. The tanshinones extracted from Salvia miltiorrhiza, a traditional Chinese medicine (TCM), are mainly used for treating cardiovascular diseases [2]. Ginkgolides, the active ingredients extracted from Ginkgo biloba, have medicinal effects in preventing and treating cardiovascular and cerebrovascular diseases [3]. In addition to medicinal values, diterpenoids have applications in industry and agriculture. For example, stevioside extracted from Stevia is 300 times sweeter than sucrose. It contains no calories and is regarded as the “third source of sugar in the world” [4]. Gibberellins are widely used to regulate the growth and development of plants in agriculture [5].

Currently, various diterpenoids have been discovered in plants, showcasing higher diversity and content compared to animals and microorganisms. It seems that plant-derived diterpenes have higher purity and biological activity [6-8]. Therefore, the primary sources of diterpenoids rely on plant extraction. However, the low content of diterpenoids in plants, the long growth cycle of plants, and the complicated extraction and purification processes for diterpenes do not satisfy the requirements for green and sustainable diterpenoid production. In recent years, the chemical synthesis of many diterpenoids has been accomplished. However, due to the diverse structure of diterpenoids, the yield of total synthesis is often low and is not practical for industrial production. Therefore, exploring the biosynthetic pathways for plant diterpenoids and using the synthetic biology strategy to design and engineer microbial strains to yield plant diterpenoids has been recognized as a promising method to produce these crucial compounds (Fig. 1).

Figure 1. Comprehensive research strategies for plant diterpene resources, including their structures, biosynthetic pathway, heterologous production and biological activity (Adapted from [96, 126-130]).

With the explosion of genome sequencing data and the increasing characterization of biosynthetic enzymes, the biosynthetic pathways of many diterpenoids have been elucidated, which lays the foundation for the heterologous production of diterpenoid products. Through metabolic engineering, synthetic biology, and fermentation engineering, researchers have developed environmentally friendly biosynthetic strategies and designed efficient cell factories to produce high-value diterpenoids (Fig. 1). Various microorganisms, including Escherichia coli and yeasts, have been exploited as chassis cells to produce diterpenoids. In recent years, a few plant diterpenoids or their structural analogs, such as taxadiene [9], tanshinone IIA [10], and steviol [11], have been produced in these microbial cell factories.

This review first describes the chemical structures and pharmacological activities of plant diterpenoids. Then we summarize the research progress of the biosynthetic pathways of a few diterpenes and the heterologous production of plant diterpenoids in microbial cell factories. We predict the prospects and development trends of synthetic biology in producing plant diterpenoids, aiming to provide more insights for future research and application of diterpenoids.

Classification of Plant Diterpenoids

According to the number of skeleton rings, plant diterpenoids can be divided into acyclic, monocyclic, dicyclic, tricyclic, tetracyclic, and macrocyclic subfamilies. Besides sesquiterpenoids, diterpenoids have the most abundant structure types in terpenoids, which can be generated through the fracture, carbon bond shift, and skeleton rearrangement (Fig. 2). In this paper, we classify diterpenoids in plants based on the skeleton ring system.

Figure 2. The different skeleton types of plant diterpenoids and the corresponding compounds. Acyclic and monocyclic diterpenoids (cyan), bicyclic diterpenoids (blue), tricyclic diterpenoids (yellow), tetracyclic diterpenoids (pink), and macrocyclic diterpenoids(green).

Acyclic and Monocyclic Diterpenoids

Acyclic and monocyclic diterpenoids are relatively rare in plants. Still, they have essential biological functions. Phytol is an acyclic diterpene commonly found in the leaves of green plants [12]. It acts as a side chain and binds to magnesium-containing porphyrins to form chlorophyll. At the same time, phytol is used as a precursor for the biosynthesis of vitamins E and K1 [13]. Recently, three new acyclic diterpenoids with strong anti-inflammatory activity, aphanamoxenes A, B, and C, were isolated from the seed of Aphanamixis polystachya [14] (Fig. 2). Ibrahim et al. identified two new monocyclic diterpenoids, tagetones A and B, from the flowers of Tagetes minuta [15] (Fig. 2).

Bicyclic Diterpenoids

Bicyclic diterpenoids are mainly divided into the labdane and clerodane families. In addition, bicyclic diterpenoids can combine with other structure types to form new compounds, such as ginkgolides composed of sesquiterpene and diterpene structures [16].

Labdanes. Labdanes have naphthane as the parent nucleus and possess various structures formed by rearranging and C–C bond cleavage of the parent nucleus. They are divided into the labdane (β-CH3 at C-10 and α-H at C-5 and C-9) and ent-labdane (α-CH3 at C-10, β-H at C-5 and C-9) subfamilies (Fig. 2). Andrographolide and its derivatives isolated from Andrographis paniculate of the family Acanthaceae [17] and forskolin from Coleus forskohlii are two representative compounds in this class [18].

Clerodanes. Clerodanes have attracted attention because of their insect antifeedant activity. Most of them were found in Angiosperm. The clerodanes can be divided into two classes, clerodane and ent-clerodane [19]. Various clerodanes with antitumor activity have been found in Scutellaria barbata of the genus Scutellaria in Labiatae, such as scutebata A. It has cytotoxic activities against a variety of cancer cells, LoVo (colon cancer), MCF-7 (breast cancer), SMMC-7721 (hepatoma cancer), and HCT-116 (colon cancer) cells [20] (Fig. 2). Besides, six undescribed clerodanes were isolated from a traditional Uygur medicine, Salvia deserta, which can inhibit the secretion of cytokines TNF-α and IL-6 in macrophages RAW264.7, displaying immunosuppressive activity likely attributed to their possession of a terminal, α-unsaturated-β-lactone moiety [8, 21]. Furthermore, the clerodanes diterpenoids separated from levels of Casearia coriacea possess toxicity to malaria parasite with selectivity.

Others. Ginkgolides were isolated from the Ginkgo biloba of the Ginkgoaceae family. Ginkgolides include ginkgolides A, B, C, J, M, K, L, and bilobalide. Different from the other seven diterpenes, bilobalide is a sesquiterpene. Ginkgolides have a unique twelve carbons skeleton structure embedded with a tert-butyl group and six 5-membered cyclic, which include a spiro [4.4] nonane, tetrahydrofuran, and three lactonic rings [22](Fig. 2). Ginkgolides specifically inhibit the platelet-activating factor receptor (PAFR). It was found that the position and number of hydroxyl groups have a significant impact on the activity of ginkgolides, such as the hydroxyls of ginkgolides C and J are at the C-7 position, their activity are weaker than ginkgolide B whose hydroxyl is located on the C-1 position, and it has the highest activity [23].

Tricyclic Diterpenoids

Tricyclic diterpenoids are abundant in nature and rich in biological activities, such as anti-tumor, anti-inflammatory, and antibacterial. Tricyclic diterpenoids mainly include abietane, pimarane, rosane, cassane, totarane, and spongian. They can be divided into two types: aromatic and non-aromatic, according to the structural characteristics of their C-ring skeleton.

Abietanes. In the 19th century, people found the first abietane diterpene from colophony, abietic acid. It has antitumor, antioxidant, and other activities [24]. Studies have shown that methyl abietate, abietinol, and abietinal can be obtained by modifying the carboxyl at the C-18 position through esterification, reduction, and oxidation. Through assessing their cytotoxicity, found that among them, methyl abietate showed the highest cytotoxicity, and abietinol presented weaker cytotoxicity [25]. Carnosic acid is also one of the representative tricyclic diterpenes, which was isolated from Rosmarinus officinalis of the family Lamiaceae. It is mainly used in the food processing industry (Fig. 2). This compound is recognized for its antioxidant, anti-inflammatory and cardiovascular protective effects [26-28]. Tanshinone IIA and tanshinone is an active component extracted from S. miltiorrhiza, a traditional Chinese medicine (Fig. 2). It can alleviate atherosclerosis by regulating macrophage polarization and has antitumor activity [29]. It was reported that introducing amino acids in the D-ring of tanshinone IIA could enhance its cytotoxicity against tumor cells [30].

Pimaranes. Like abietanes, pimaranes were also originally isolated from colophony. They can be divided into four types, namely, pimarane, isopimarane, ent-pimarane and ent-isopimarane. The difference between isopimarane and pimarane is in the configuration of the methyl group at C-17. Isopimarane has α-CH3 while pimarane has β-CH3. Pimaric acid is the first pimarane diterpene isolated from Pinus sylvestris of the genus Pinus in Pinaceae in 1983 (Fig. 2). Icacinol, a pimarane-type diterpene, was isolated from the tubers of Icacina oliviformis [31]. Four highly oxidized pimarane diterpeniods isolated from the rhizomes of Kaempferia takensis can inhibit the generation of nitric oxide (NO) in RAW264.7 macrophage cells and inhibiting NF-κB production in HaCat human skin cells [32].

Others. In addition to the two tricyclic diterpenes described above, there are several other types, such as rosane, cassane, totarane, and spongian. Totarol is a representative totarane diterpene isolated from the duramen of Podocarpus macrophyllus (Fig. 2) [33]. At present, many daily chemical products and cosmetics contain totarol. Triptolide is mainly extracted from Tripterygium wilfordii, its potency is 100-200 times higher than that of tripterygium glycosides, and its therapeutic window is narrow. Triptolide is currently the main component of the quality control specification of T. wilfordii preparations. (Fig. 2).

Tetracyclic Diterpenoids

Tetracyclic diterpenoids can be classified into two types. The tetracyclic diterpenes with a C-8 bridging ring include kaurane, ent-gibberellin, phyllocladane, atisane, beyerene, and grayanane. Their biosynthetic intermediate is ent-CPP. The tetracyclic diterpenes with C-9 bridging ring include aphidicolane, stemodane, stamarane, ent-stamarane, and scopadulane, among which aphidicolane was only found in fungi, and the intermediate is syn-CPP.

Kauranes. Kaurene diterpenoids were initially discovered from the essential oil of the leaves of Agathis australis, a plant of the Araucariaceae family from New Zealand. More than 1000 kauranes have been identified [34], and most of them are from plants of the Isodon and Asteraceae families [35]. Steviol glycosides (STE-G) are representative kauranes isolated from Stevia rebaudiana. They are currently used as food sweeteners. Various STE-G can be formed by substituting the C-13 hydroxyl or C-19 carboxyl of steviol with different types and numbers of glycosyl groups. Rubusoside possesses one molecule of glucose at the C-13 and C-19 positions of steviol, respectively. Stevioside contains additional glucose at the C-13 position of rubusoside. Continuing to add glucose by the glycosyltransferase gives rebaudioside A, D, and M (Fig. 2).

Gibberellins. Gibberellins (GAs) are a family of important hormones that control plant growth and development. GAs share the gibberellane backbone and differ by changing the number and position of double bonds and hydroxyl groups. GAs were initially isolated from Gibberella fujikuroi, a rice pathogen, and were later found to exist in the plant and bacteria kingdoms. More than 130 GAs have been found [36]. According to the number of carbon atoms, GAs can be divided into 20C ent-gibberellane and 19C ent-gibberellane, and the latter is relatively more varied. GA3, GA7, and GA30, which possess similar structures to their A-ring, showed better activity. Because the content of GA3 in plants is too low to extract, it can be achieved by using a synthetic biology strategy, for example, Kildegaard et al. reconstructed the biosynthetic pathway of gibberellins in Yarrowia lipolytica to produce GA3 and GA4, successfully [37, 38] (Fig. 2).

Atisanes. Atisane diterpenoids mainly exist in nature as ent-atisanes. Nearly half of the atisanes were isolated from Euphorbia. Other genera that contain atisanes include Isodon, Xylopia, and Spiraea. Drummond et al. first isolated atisanes from the tropical tree Erythroxylon monogynum in 1965 [39]. Oxidative modification of these compounds often occurs at the C-3, C-16, and C-17 positions of the parent nucleus (Fig. 2). Wang et al. discovered two novel ent-atisane ebractenone A and ebractenone B, which possess the unusual 2-oxopropyl moiety, from the roots of Euphorbia ebracteolata [40].

Others. Phyllocladanes are rare in nature, and their structures are similar to kauranes. Calliterpenone and its analogs isolated from Callicarpa macrophylla have the activity of promoting plant growth [41]. Grayananes are derived from kaurene skeleton rearrangement in nature and are found only in Ericaceae plants [42]. These compounds share a unique 5/7/6/5 tetracyclic structure. Sun et al. isolated 24 grayanane diterpenoids from Rhododendron auriculatum, six of which exhibited significant analgesic effects at 5.0 mg/Kg [43]. Stemodin and stemodinone are the first reported stemodanes derived from Stemodia maritima of the genus Stemodia in the family Scrophulariaceae [44] (Fig. 2). Two years later, another compound with a new configuration, stemarin, was isolated from S. maritima [45].

Macrocyclic Diterpenoids

Taxanes. At present, more than 500 taxanes have been found. They can be divided into four subfamilies, i.e., taxanes, secotaxanes, abeotaxanes, and cyclotaxanes [46] (Fig. 2). Taxanes diterpenoids were mainly isolated from the Taxus of the Taxaceae family. Paclitaxel, one of the most famous anticancer drugs, was initially discovered in the screening of antitumor drugs, and its structure was first reported in 1971 [47]. In 1994, the total synthesis of paclitaxel was reported. However, because of the complexity and the high cost of total synthesis, it is unrealistic to produce paclitaxel by this method. And semi-synthesis and microbial fermentation are the most promising alternative methods [48].

Due to the side effects and poor water solubility of paclitaxel, plenty of research has been focused on its structure modification in recent years. Bouchet et al. modified the benzoylphenyl and acetyl groups of paclitaxel to alkoxy and hydroxyl groups, respectively, to afford docetaxel. Docetaxel binds to tubulin with twice the affinity of paclitaxel because of its smaller space steric hindrance and stronger hydrophilicity brought by the polar groups [49]. Singer et al. found that adding hydrophilic and non-pharmacological groups to paclitaxel can improve the antitumor efficiency and reduce the side effects, as exemplified by xyotax [50].

Others. Macrocyclic diterpenes are a class of compounds whose molecular skeleton consists of four isoprene units and has a ring structure of more than six members. Their structures are complex, and generally have variety of functional groups, with unique biological activities and pharmacological effects, such as cytotoxicity, anti-tumor, antiviral and anti-inflammatory activities. These compounds are mainly found in plants of Euphorbiaceae and Thymelaeceae [7]. Such as cembranes, tigliane, casbanes, and jatrophanes, which have become the research hotspot due to their different skeletal structures and extensive biological activities (Fig. 2). For example, the ingenol extracted from Euphorbia kansui or Leptochloa chinensis has significant anti-tumor and antiviral activity. Prostratin, a macrocyclic diterpenoid isolated from small shrubs of New Zealand, can inhibit the HIV-1 infection and reduce its incubation period [51].

Cembranes can be divided into isopropyl, five-, six-, seven-, and eight-membered lactone rings, ring-opening, reduced-carbon type, etc. A new cembrane diterpene, named (-)-(1S)-15-hydroxy-18-carboxycembrene, was isolated from the roots of Euphorbia pekinensis Rupr, which had inhibitory effects on five tumor cells, including Hela, PC-3, HT1080, A375-S2 and MDA231 [52]. Pekinenin G is isolated from Euphorbia, which belongs to casbanes. It showed different cytotoxicity against four kinds of tumor cells, including BGC-823, HT-29, MCF-7, and A549 [53]. Jatrophanes have a bicyclic 15-carbon skeleton and are a new class of P-glycoprotein inhibitors [54]. Verticillol, the first isolated verticillane compound (Fig. 2), was isolated from Sciadopitys verticillate in 1962. It is mainly distributed in Taxu and is considered to be the precursor of taxanes [55].

Pharmacological Activities of Plant Diterpenoids

Diterpenoids have a wide range of biological activities due to their complex and diverse structure. They are commonly used to treat cardiovascular diseases, hypertension[56], and diabetes [57, 58]. These compounds also have antineoplastic, anti-inflammatory, antibacterial, antiviral [59], antioxidative [60], and antimalarial activities [61]. Herein, our paper systematically summarizes the pharmacological studies on diterpenoids.

Antineoplastic Activity

The antitumor effect is one of the most interesting pharmacological activities of diterpenes. Paclitaxel, triptolide, and ent-kaurene are star compounds for treating cancer. Paclitaxel has a variety of antineoplastic activities, especially for ovarian cancer, endometrial carcinoma, and breast cancer, which have a high incidence. It has been reported that paclitaxel can exert antitumor effects by promoting tubulin polymerization and blocking the progression of mitosis. At the same time, it can also promote the proliferation and differentiation of T-lymphocyte and increase the number of nature killers [1].

It is well known that non-small cell lung cancer (NSCLC) is one of the most severe diseases, it is characterized by a high incidence of metastasis and poor survival, and epithelial-to-mesenchymal transition (EMT) is a major factor inducing tumor metastasis. Deng et al. found that triptolide could inhibit the EMT of NCL-H1299 cells in a concentration-dependent manner and suppress β-catenin expression in NCL-H1299 and NCL-H460 cells [62].

In recent years, ent-kaurane has also been shown to have high anticancer activity. For example, oridonin, a natural compound isolated from the genus Isodon in the Labiatae, is currently in phase I clinical trial in China. This compound has anticancer effects on various human cell lines, such as large intestine cancer, acute myeloid leukemia, and BxPC-3 [63]. Overall, diterpenoids offer diverse avenues for cancer treatment, for example, they can inhibit the growth and migration of cancer cells by suppressing the AKT signaling of the key proteins implicated in diseases [64, 65], or controlling cell cycle to induce senescence and death of cancer cells [65]. Furthermore, altering the structure of diterpenoids enables the control of toxicity selectivity, premyrsinanes isolated from Euphorbia gedrosiaca show safety against breast cancer cells [66].

Anti-Inflammatory Activity

The pathogenesis of numerous diseases is closely linked to inflammation. Diterpenes exhibit significant anti-inflammatory properties, suggesting their potential therapeutic efficacy in a range of conditions including neuroinflammatory diseases, atherosclerosis, liver damage, and colitis. The inflammatory response induced by cytokines and chemokines can cause various inflammatory diseases, and anti-inflammatory drugs are an important therapeutic strategy. Tanshinone IIA and cryptotanshinone have significant anti-inflammation activities. The research showed that tanshinone IIA plays an anti-inflammatory role by inhibiting the expression of inflammatory cytokines and TLR2, NF-kappa B, and ICAM-1 [67].

(1R,2R,4aS,5R)-1-(hydroxymethyl)-1,4a-dimethyl-6-methylene-5-(2-(4-nitrophenoxy) ethyl) decahydronaphthalen-2-ol is a derivative of andrographolide. Wang et al. examined its inhibitory activity for LPS-induced NO production in the RAW264.7 macrophage. It was shown that its IC50 value (3.38 ± 1.03 μM) was higher than that of andrographolide (8.81 ± 1.03 μM). They found that andrographolide and its derivatives exert anti-inflammatory effects by down-regulating the expression of COX-2, iNOS, and NF-κB and inhibiting the signal pathway [68].

Wang et al. isolated twelve new diterpenoids from the leaves of Euphorbia lathylris and verified their anti-inflammatory effects. They found that euplarisan A showed anti-inflammatory effects with an IC50 value of 7.50 ± 1.45 μM. It could inhibit the expression of inflammatory cytokines in a dose-dependent manner, including iNOS, COX-2, and p-IκBα [69].

Antibacterial Activity

Some diterpenoids have a broad antimicrobial spectrum, low toxicity, and high antibacterial activity. For instance, Streptococcus mutans has been recognized as the most important oral cariogenic pathogen. It was reported that rubusoside (50 mM) showed the 97% inhibitory activity against the mutansucrase in S. mutans with 500 mM sucrose. Therefore, it has particular research value and application prospects in preventing and treating caries [70].

Siddique et al. isolated a new labdane diterpene, named (E)-8(17), 12-labdadiene-15, 16-dial, from Zingiber montanum, it has potential antibacterial activity against a series of multi-drug and methicillin-resistant Staphylococcus aureus (MRSA) with minimum inhibitory concentrations (MIC) of 32–128 μg/ml. It has exomethylene at C-8, an olefine at C-12, and two aldehyde groups at C-16 and C-17. These are the reasons for its significant antibacterial activity against MRSA strains [71].

Others

In addition to the above common activities, diterpenoids have numerous other biological activities. Such as andrographolide, 14-dehydroxyandrographolide-12-sulfonic acid sodium salt, and 14-α-lipoyl andrographolide have been found to inhibit several influenza viruses, including H5N1, H1N1, and H9N2. Among them, 14-α-lipoyl andrographolide inhibited the adsorption of the virus to erythrocytes. This suggested that the compound can interfere with viral hemagglutinin [72]. Andrographolide can also promote the regeneration of the skeletal muscle of mice. The experimental results show that it might up-regulate the transcription of myogenic differentiation genes to facilitate the differentiation and fusion of muscle cells by enhancing the methylation of histone H3K4, thereby promoting the regeneration of skeletal muscle [73].

Due to its high sweetness and low calorie, rubusoside is used as a sweetener. It also can combine with resveratrol, curcumin, and other anticancer drugs to increase solubility, such as by 33-fold of resveratrol and 60-fold of curcumin [74, 75].

GAs is a large group of endogenous hormones that can regulate plant vegetative and reproductive growth. They play important roles in promoting seed germination, root growth, stem and leaf growth, flowering, and fruit ripening [76]. Many diterpenoids showed noteworthy neuroprotective effects in cell injury models induced by H2O2 and MPP+ [77], they can activate the autophagia and trigger the parkin/IKK/p65 pathway to increase the OPA1. They can also show a neuroprotective effect by inhibiting the COX-2 pathway and MAPK pathway [78].

Studies on the Biosynthesis of Plant Diterpenoids

In plants, all terpenoids are formed by the condensation of five-carbon unit isopentenyl pyrophosphate (IPP) and dimethylallyl pyrophosphate (DMAPP) to form the precursor geranyl diphosphate (GPP), farnesyl diphosphate (FPP), and (E, E, E)-geranylgeranyl diphosphate (GGPP), finally form a terpenoid skeleton through the post-modification reaction. The five-carbon structure in plants can be obtained from the mevalonic acid (MVA) pathway in the cytoplasm and the 2-C-methyl-D-erythritol-4-phosphate (MEP) pathway in the plastid. The products of these two pathways can be interchanged through the plasma membrane. IPP and DMAPP can also undergo tautomerization under the action of isopentenyl pyrophosphate isomerase (IDI) (Fig. 3A). The MVA pathway and the MEP pathway have been extensively reported and will not be detailed in this review [79, 80].

Figure 3. (A) Biosynthesis of the IPP and DMAPP via the MVA pathway (purple) and MEP pathway (green). Enzyme abbreviations are as follows; AACT, acetoactl-CoA thiolase. HMGS, hydroxymethylglutaryl-CoA synthase. HMGR, hydroxymethylglutaryl-CoA reductase. MVK, mevalonate kinase. PMK, phosphmevalonate kinase. MVD, mevalonate 5- phosphate decarboxylase. IDI, isopentenyl diphosphate isomerase. DXS, 1-deoxy-D-xylulose 5-phosphate synthase. DXR, 1- deoxy-D-xylulose 5-phosphate reductoisomerase. MCT, 2C-methyl-D-erythritol 4-phosphate cytidyltransferase. CMK, 4- (cytidine 5’-diphospho)-2C-methyl-D-erythritol kinase. MDS, 2C-methyl-D-erythritol-2,4’-cyclodiphosphate synthase. HDS, 4-hydroxy-3-methylbut-2-enyl diphosphate synthase. HDR, 4-hydroxy-3-methylbut-2-enyl diphosphate reductase. For intermediates, GAP, D-glyceralfehyde-3-phosphate. DXP, 1-deoxy-D-xylulose 5-phosphate. MEP, 2C-methyl-D-erythritol 4- phosphate. CDP-cytidyl diphosphate. MEcDP, 2C-methyl D-erythritol-2,4-cyclodiphosphate. HMBDP-1-hydroxy-2-methyl- 2-(E)-butenyl-4-diphosphate. OP and OPP signify mono- and diphosphate moieties, respectively. Pi represents inorganic phosphate (B) Biosynthetic pathway of forskolin.

After synthesizing IPP and DMAPP, geranylgeranyl pyrophosphate synthase (GGPPS) catalyzes the synthesis of GGPP, a precursor common to all diterpenoids, by three molecules of IPP and one molecule of DMAPP. GGPP could form the skeletons of various diterpenes under the action of different diterpene synthases. Then these skeletons are converted into different compounds through a series of enzymatic reactions, such as hydroxylation, peroxidation, methylation, glycosylation, and cleavage rearrangement. According to the functional domains of diterpene synthases, they can be divided into Class I, Class II, and Class I/ Class II diterpene synthases, Class I and Class II diterpene synthases contain DDXXD and DXDD domains, respectively. Class I/ Class II diterpene synthases contain both types of characteristic domains, having the characteristic of catalyzing two reactions at the same time. For example, abietadiene synthase isolated from Gymnosperms can catalyze GGPP to produce copalyl diphosphate (CPP), (+)-CPP, or ent-CPP, and then further be converted into abietadiene and ent-kaurene. Many related studies have been reported and will not be discussed here.

Biosynthesis of Forskolin

Forskolin is a diterpenoid belonging to the labdane. It is produced uniquely to the root species of C. forskohlii, a genus of Coleus in the Labiaceae family. It mainly exists in the cork layer of the root of C. forskohlii, but the content is very low in plants. Forskolin can activate intracellular adenylyl cyclase to increase the concentration of cyclic adenosine monophosphates (cAMP). It has the effect of preventing platelet aggregation and antihypertension. It has been approved for treating asthma, glaucoma, hypertension, and other diseases.

In plants, GGPP is synthesized through the MEP pathway. The diterpene synthases are used to subsequently generate the basic diterpene skeleton, and finally, forskolin is obtained under a series of oxidases and acetyltransferases (Fig. 3B). Zerbe et al. performed transcriptome sequencing of C. forskohlii [81]. They identified six candidate diterpene synthases (CfTPS1, CfTPS2, CfTPS3, CfTPS4, CfTPS14, and CfTPS15) from the transcriptomic data. CfTPS2 belongs to the Class II diterpene synthase. It can convert GGPP to form 8α-hydroxy-CPP and then cyclize into 13R‐manoyl oxide (13R-MO) by the Class I diterpene synthase CfTPS3. However, in the presence of CfTPS1 and CfTPS3, or CfTPS4, GGPP can be catalyzed to consecutively produce CPP and miltiradiene, a key hub in the biosynthesis of tricyclic diterpenes [82] (Fig. 4). In 2017, Pateraki et al. characterized the downstream enzymes in the forskolin biosynthetic pathway utilizing an agrobacterium-mediated transient expression system in tobacco. The results showed that CfCYP76AH15, CfCYP76AH8, and CfCYP76AH17 could catalyze the formation of 11-oxo-13R-manoyl oxide from 13R-MO. CfCYP76AH15 showed the highest activity and specificity among these three enzymes. CfCYP76AH11 can convert 11-oxo-13R-manoyl oxide into 9-deoxy-7-deacetylforskolin, which is subsequently catalyzed to form 7-deacetylforskolin by CfCYP76AH16. By co-expressing CfDXS, CfGGPPS, CfTPS2, CfTPS3, CfCYP76AH15, CfCYP76AH11, and CfCYP76AH16, the intermediate of forskolin, 7-deacetylforskolin, was obtained. In addition, CfACT1-6 and CfACT1-8 were selected as candidate enzymes from ten acetyltransferases. It was found that CfACT1-8 has better specificity and could effectively convert 7‐deacetylforskolin into forskolin [83]. It was found that CFRB1 (Alcaligenes faecalis), a plant endophyte of C. forskohlii, could promote the synthesis of forskolin in plants by up-regulating the expression of the genes mentioned above [84].

Figure 4. Biosynthetic pathway of tanshinones. Dash arrowhead indicates that the enzymes have not been identified.

Biosynthesis of Tanshinones

Tanshinones are a class of lipid-soluble abietane found in S. miltiorrhiza, mainly distributed in the root and stem of S. miltiorrhiza. Up to now, more than 50 tanshinones have been isolated, including tanshinone I, tanshinone IIA, tanshinone IIB, dihydrotanshinone I, etc. Studies have proved that the phenanthrene-quinone structure of tanshinones is the basis of its cytotoxic effect, which can be combined with the DNA molecules to kill tumor cells [67].

In S. miltiorrhiza, GGPP is cyclized to miltiradiene under the diterpenoid synthases SmCPS1 and SmKSL1. Then various tanshinones are generated by different CYP450 enzymes, dehydrogenases, and demethylases (Fig. 4). Guo et al. identified six candidate CYP450s by analyzing the transcriptomic data of the root and the stem of S. miltiorrhiza [85]. They demonstrated that SmCYP76AH1 could catalyze the generation of ferruginol from miltiradiene [86]. Guo et al. identified SmCYP76AH3 and SmCYP76AK1 that can sequentially catalyze multi-step oxidation reactions to ferruginol, generating two intermediates, 11,20-hydroxy ferruginol and 11,20-hydroxyl sugiol [87]. Mao et al. found that the sequence homology of SmCYP76AH1 and SmCYP76AH3 exceeded 80%, so they designed a series of SmCYP76AH1 mutants to integrate the functions of the two enzymes based on homology modeling. Finally, CYP76AH1D301E and CYP76AH1V479F were certified to have the functions of both enzymes, and this strategy could improve the catalytic efficiency by reducing the transformation of intermediates [88]. Through the analysis and mining of genomic data, Ma et al. discovered the CYP71D subfamily in the genome of S. miltiorrhiza, and further studies demonstrated that CYP71D373 and CYP71D375 were involved in tanshinone biosynthesis [89].

There are many unknowns in the biosynthetic pathways of tanshinones and their intermediates. In recent years, many reports have paid more attention to the regulatory factors in the biosynthetic pathway. For example, Deng et al. found that SmWRKY2 could increase the production of tanshinones by upregulating the expression of SmDXS2 and SmCPS [90]. SmERF73, a stress response transcription factor that belongs to the VII ethylene response factor family, plays a role in regulating gene expression related to tanshinones biosynthesis. When SmERF73 is overexpressed, seven genes involved in biosynthesis are up-regulated to increase the yield of tanshinones. Conversely, the transcript levels of these genes and the production of tanshinones will be down-regulated [91]. Besides, endophytic fungi can also enhance the synthesis of secondary metabolites in medical plants, Penicillium steckii DF33 can up-regulate the expression of key enzyme genes like GGPPS, CPS1, KSL1 [92], the yield of tanshinone was increased, and the resistance of plants was also improved [93].

Biosynthesis of Steviol Glycosides

STE-G are high-sweetness and zero-calorie sweeteners isolated from S. rebaudiana of the genus Stevia in the Asteraceae family. The synthesis of these compounds in plants occurs in chloroplasts. GGPP is also produced through the MEP pathway by using 13C-labeled glucose. GGPP is cyclized to ent-kaurene under the catalysis of class I terpene synthases, ent-copalyl diphosphate synthases (ent-CPPS), and class II terpene synthases, ent-kaurene synthases (ent-KS). The synthesized ent-kaurene is transported to the endoplasmic reticulum, where it is converted to ent-kaurenoic acid (KA) by the multifunctional CYP450 enzyme, ent-kaurene oxidase (KO).

KA is converted to steviol by adding a hydroxyl at the C13 under the action of ent‐kaurenoic acid 13‐hydroxylase (KA13H). Steviol is the common precursor of all STE-G. Under the catalysis of glycosyltransferase SrUGT73E1, one molecule of glucose is added to the carboxyl at C-19 of steviol to obtain steviol glycoside. Alternatively, under the coaction of SrUGT85C2 and SrUGT73E1, the glucosides at C-13 and C-19 are attached to form rubusoside. Then, stevioside can be achieved under the catalysis of glycosyltransferase SrUGT91D2 that can form a 1,2-β-D-glycosidic bond on the glycosyl at C-13 of rubusoside. Moreover, with the catalyst of SrUGT76G1 and SrUGT91D2, stevioside is converted to rebaudioside A (Reb A) and rebaudioside E (Reb E), respectively. Rebaudioside D (Reb D) could be synthesized under the continuous catalysis of the above two enzymes [94](Fig. 5).

Figure 5. Biosynthetic pathway of steviol glycosides.

The most widely used STE-G on the market is Reb A, which has high sweetness and safety and has passed the safety certification of the United States Food and Drug Administration. However, Reb A has a bitter aftertaste. Compared to Reb A, Reb D and Reb M have one or two more glucose molecules attaching to the C-19 glycosyl, so the bitterness is significantly reduced. Wang et al. analyzed the transcriptomic data of S. rebaudiana and identified seven UDP-glycosyltransferases and 76 transcription factors that may be involved in the biosynthetic pathway of Reb D and Reb M [95]. Zhang et al. identified the glycosyltransferase OsUGT91C1 in rice. It can catalyze the conversion of Reb A to Reb D. Moreover, its F208M/F379A mutant reduced the formation of β-1,6 glycosidic bonds [96]. Dewitte et al. randomly mutated SrUGT76G1 from S. rebaudiana and found that UGT76G1T146G and UGT76G1H155L could effectively reduce the production of by-products and increase the yield of Reb D or Reb M [97].

Biosynthesis of Paclitaxel

Paclitaxel is mainly isolated from the bark of Taxus. The demand for paclitaxel is increasing, but it is in short supply due to its low content in plants and the scarcity of the Taxus species. There is a consensus that constructing a cell factory to produce taxol by fermentation is the key to solve this problem. The biosynthetic process of paclitaxel has been divided into three parts: (1) biosynthesis of baccatin III, the parent nucleus of taxane, (2) biosynthesis of the phenylisoserine side chain, and (3) linkage of the side chain to the taxane skeleton (Fig. 6).

Figure 6. Biosynthetic pathway of paclitaxel. Blue font indicates that the enzymes have not been identified.

Table 1 . Overview of production of plant diterpenoids in different hosts..

HostDiterpeneMetabolic engineering strategiesTiter (mg/l)Reference
E. colient-kaurane◆ Screening of optimal GGPPS and E. coli host
◆ Overexpression of three key enzymes in MEP pathway
◆ Culturing strain in a bioreactor
578[114]
E. coliSteviol◆ Optimization of the upstream pathway
◆ Screening of proper E. coli host and kaurene oxidase
◆ Truncating the N terminus-modified mutants of kaurene oxidase and attachment of soluble tags
◆ Codon optimization and increasing the copy number of CPR
◆ Introduction of cytochrome b5 (CYB5)
◆ Site-directed mutation of AtCYP714A2
1073.8[115]
E. coliTaxadiene◆ Modularization of the taxadiene metabolic pathway
◆ Using systematic multivariate analysis to achieve a balance of the two modules
1020[9]
S. cerevisiaeMiltiradiene◆ Overexpression of the pathway genes
◆ Downregulation of Erg9
◆ Knocked out transcriptional regulators
◆ Optimization of the medium
◆ Fusion of CfTPS1 and SmKSL1
◆ Protein modification of SmKSL1
3500[117]
S. cerevisiaeForskolin◆ Fusion of BTS1 and ERG20F96C
◆ Overexpression of HMG1
◆ Truncating the N terminus of CfCPR
◆ Fusion of CfCYP76AHs and tR~tB
◆ Regulation of copy numbers of the target genes, amplification of the endoplasmic reticulum (ER) area and cofactor metabolism enhancement
◆ Fed-batch fermentation
21.47[119]
S. cerevisiaeCarnosic acid◆ Overexpression of BTS1-GGGS-ERG20F96Cp
◆ Codon-optimization, N-terminus truncation, and fusion of tSmCPS and tSmKSL
◆ Used SmCPR from S. miltiorrhiza
◆ Co-expression of SmCPR~t28SpCytb5 fusion protein and CYP76AH1
◆ Overexpression of ScCTA1 and ScCTT1
◆ Overexpression of INO2, the HEM3 (heme synthase) gene, and the NADH kinase gene (POS5)
◆ Batch and fed-batch fermentation
24.65[120]
S. cerevisiaeRubusoside◆ KS from G. fujikuroi, KO, CPR1, UGT74G1, and UGT85C2 from S. rebaudiana, KAH from Arabidopsis thaliana
◆ Overexpression of tHMG1 and IDI1
◆ Site-directed mutation of FPSF112A
◆ Replacing promoter of INO2 with a stronger one
◆ Overexpression of the efflux-pump PDR11 and the stress-response factor MSN4
◆ Knocking out GAL7 and overexpression of PGM2
1368.6[94]
Y. lipolyticaGibberellin◆ Downregulating the endogenous squalene synthase gene
◆ Choosing AtCPS, AtKS, AtKO, AtATR2 ang YlCyb5 to producing ent-KA
◆ Codon-optimization of all genes
◆ Gene AtC20ox and AtC3ox were expressed under the control of the strong promotors PrExp and PrTefintron, respectively
◆ N-terminus truncation of AtCPS, AtKS, and AtKO and fusion of CPS and KS
GA4 17.29
GA3 2.93
[38]
C. reinhardtiiSclareol◆ Codon optimization of terpene synthase
◆ All transgenes were driven by the PSAD promoter and FDX1 terminator
◆ Using GGPPS from C. reinhardtii and sclareol synthase from S. sclarea
656[121]


GGPP is first cyclized to taxadiene by the taxadiene synthase (TASY), a Class I diterpene synthase. Then taxadiene undergoes a series of hydroxylation at the C-1, C-2, C-4, C-5, C-7, C-9, C-10, and C-13 positions. The enzymes catalyzing hydroxylation at five positions of taxadiene have been successfully identified [98-100]. Stefan et al. found that taxadiene‐5α‐hydroxylase (T5αOH) can introduce a hydroxyl group at the C-5 position of taxadiene. The hydroxylation promotes the migration of double bond to form taxa‐4(20),11(12)‐dien‐5α‐ol [101]. Taxane 13α-hydroxylase (T13αOH) can convert taxane-5α-ol to taxadiene-5α, 13α-diol [102]. In addition, multi-step formylation and acetylation are involved in the formation of paclitaxel. The taxane-2α-ol-O-aectyltransferase (TAT) was identified from the Taxus, and it can catalyze the acylation of 5α-hydroxyl [103]. Walker et al. reported that taxane-2α-benzoyl transferase could convert the semi-synthetic substrate 2-debenzoyl-7,13-diacetylbaccatin III to form 7,13-diacetylbaccatin III. They also discovered that the 10-deacetylbaccatin III-10-O-acetyl transferase (DBAT) from Taxus chinensis could catalyze the reaction of 10-deacetylbaccatin III with acetyl-CoA to form bacatine III [104, 105] (Fig. 6).

Formation of the side chain of paclitaxel is the rate-limiting step in paclitaxel biosynthesis. With L-α-Phe as the precursor, β-Phe is obtained under the action of phenylalanine aminomutase (PAM) [106]. β-Phe is converted to β-Phe-CoA by β-phenylalanine-CoA ligase (PCL) [107]. The β-phenylpropanoyl-CoA is attached to the taxane scaffold at C-13 by taxoid C-13 O-phenylpropanoyltransferase (BAPT), the 2' position of the side chain is subsequently hydroxylated to form 3'-N-debenzoyltaxol by CYP450 hydroxylase [108]. Finally, the benzoylation occurs at the C3' position under the 3'-N-debenzoyl-2'-deoxytaxol-N-benzoyltransferase (DBTNBT) to complete the synthesis of paclitaxel [109] (Fig. 6). Zhang et al. identified a new C4β-C20 epoxidase and utilized it in conjunction with oxomutases / epoxidases, taxane 1β-hydroxylase, taxane 9α-hydroxylase, taxane 9α-dioxygenase, and phenylalanine-CoA ligase to successfully biosynthesize the key intermediate baccatin III and to convert baccatin III into paclitaxel in Nicotiana benthamiana, establishing a metabolic route to taxoid biosynthesis [110]. Through persistent efforts, the intricate biosynthetic pathways of taxanes have been gradually resolved.

By analyzing the functions of the enzymes identified so far, we can see that the hydroxylation and acylation on the parent nucleus are alternate, and the order of most reactions still needs to be determined. There are about eight CYP450 enzymes involved in paclitaxel biosynthesis, but only five have been identified. The corresponding CYP450 hydroxylases catalyzing reactions at the C-1, C-4, and C-9 positions are still unknown. Moreover, the enzymes involved in the biosynthesis of propylene oxide remain unknown. More efforts are needed to elucidate the complete paclitaxel biosynthetic pathway.

Heterologous Production of Plant Diterpenoids

Using synthetic biology technology to produce active ingredients of medicinal plants in heterologous expression systems has great advantages, including short production cycles, independence from environmental factors, easy separation and purification, and easy to perform large-scale fermentation.

A variety of pharmaceutical ingredients have been produced de novo in chassis organisms. Keasling’s group has constructed a high-yield yeast engineering strain for the synthesis of the precursor of artemisinin, artemisinic acid. Then artemisinin can be obtained through a series of simple chemical reactions. The yield of artemisinic acid in a 100 m3 fermenter is equal to that of 8,000 acres of agricultural cultivation [111]. Chinese researchers have cooperated to construct a "Ginseng-Yeast" cell factory that can simultaneously produce oleanolic acid, protopanaxadiol, and protopanaxatriol, which enables the yeast to synthesize sapogenin [112]. L-homoserine is used as an important intermediate and additive in the industry. Zhang et al. achieved a yield of 11.1 g/l in E. coli through metabolic engineering [113].

Heterologous Production in E. coli

E. coli has been a popular host in synthetic biology research because of its clear genetic background, rapid growth and metabolism, and genetic amenability. E. coli is often used as a chassis for producing terpenes. However, its IPP and DMAPP precursors cannot meet the needs of high-titer terpenoid production. Genetic modification is needed to enhance the metabolic flux in terpenoid biosynthesis, thus improving the yield of target products. Kong et al. overexpressed the 1-deoxyxylulose-5-phosphate synthase (DXS), farnesyl diphosphate synthase (IspA), and IDI in the MEP pathway to improve the titer of isoprenoid precursor. At the same time, they screened six GGPPSs from different species and eight E. coli hosts to determine the best combination for ent-kaurene production. Finally, ent-kaurene yield could reach 578 mg/l when expressing the GGPPS of Rhodobacter sphaeroides in E. coli MG1655 [114]. Using the strain optimization and precursor-enhancing strategies, Sun et al. selected E. coli K-12-derived strains with an enhanced MVA pathway as the host, followed by screening KO and adding protein fusion tags, the yield of KA could reach 250 mg/l. Subsequently, they used several methods, including N-terminal truncation, codon optimization, copy number increase, molecular docking, and site-directed mutagenesis, to improve the catalytic efficiency of oxidase KA13H. Finally, the titer of steviol reached 1.07 g/l [115].

Ajikumar et al. used multivariate-modular metabolic engineering in E. coli to partition the metabolic pathway of taxadiene into two modules: the upstream module of the endogenous MEP pathway for supplying IPP and the heterologous downstream module for terpene biosynthesis. By overexpressing four genes in the upstream module and two genes in the downstream module by changing the plasmid copy number and regulating the promoter strength, the yield of taxadiene reached 1.02 ± 0.08 g/l in the fed‐batch fermentations [9].

Heterologous Production in Yeasts

Yeasts are also popular hosts for reconstituting the biosynthetic pathways of plant secondary metabolites. Many membrane-bound enzymes involved in plant metabolism, especially CYP450s, are relatively easier to be functionally expressed in yeast. Saccharomyces cerevisiae was the first yeast host for exogenous gene expression and reconstituting heterologous biosynthetic pathways. Dong et al. integrated tHMG1 encoding the catalytic domain of 3-hydroxy-3-methylglutaryl coenzyme A reductase and UPC2-1, the G888D mutant of the transcription factor UPC2, into the chromosome of yeast BY4741 to improve the expression levels of ERG13, ERG12, and ERG8 in the MVA pathway. In addition, they used the weak promotor PHXT1 to replace the native promotor of ERG9 to reduce the flux to the squalene by-product, and the quantitative analysis showed that the content of GGOH reached 3.97 mg/l. Through site-directed mutagenesis to obtain mERG20, which can function as a GGPPS, then the fusion of ERG20 and mERG20 could enhance the titer of GGOH, which can be achieved at 12.54 mg/l [116].

Hu et al. used different approaches, such as overexpression of key enzymes in the MVA pathway, down-regulating the expression of genes in the competing metabolic pathways, and knocking down the transcription factors that repress terpenoids biosynthesis. Finally, the titer of GGPP reached 2.1 g/l. Then they used this strain to evaluate different miltiradiene synthases. It was found that the fusion protein composed of TPS1 from C. forskohlii and KSL1 from S. miltiorrhiza could produce 3.5 g/l of miltiradiene in a 5-L bioreactor [117]. The site-directed mutagenesis of CYP76AH15A99I resulted in the enhanced production of 11-oxo-manoyl oxide, the intermediate of forskolin, by 5.6 folds [118]. Based on this, Ju et al. optimized the adaptations between CfCYP76AHs, t66CfCPR, and t30AaCYB5, and the yield of forskolin reached 759.42 μg/l. Moreover, multiple metabolic engineering strategies, including regulation of the gene copy numbers, amplification of the endoplasmic reticulum (ER) area, and enhancement of cofactor metabolism, were implemented to improve the metabolic flux of 13R-MO during forskolin generation. The yield of forskolin reached 21.47 mg/l in the shake flask and 79.33 mg/l in a 5-L fermenter [119].

Wei et al. improved carnosic acid biosynthesis efficiency in yeast through the co-expression of CYP450s and CPR enzymes, generating fusion protein, utilizing endoplasmic reticulum engineering, and increasing the supply of cofactors to improve the carnosic acid yield to 24.65 mg/l in shake flasks [120]. Xu et al. remodeled the complex metabolic networks of rubusoside by modular engineering. By overexpressing the rate-limiting enzymes and using the FPSF112A mutant to reduce the generation of shunt products, they increased the yield of ent-kaurene by 33.9 times. In addition, the combination of different strategies, including regulating the cellular stress response, activating the efflux pump, and knocking out bypass pathways, resulted in the production of 1368.6 mg/L rubusoside in a 15-L bioreactor [94].

Kildegaard et al. used the oleaginous yeast Y. lipolytica to construct the biosynthetic pathway for GAs. The production of KA, the GA precursor, reached 3.75 mg/l by optimizing the MVA pathway and overexpressing the rate-limiting enzymes in the pathway. Then they generated a GA4-producing strain by introducing the enzymes involved in GAs biosynthesis, resulting in the production of 17.29 mg/l GA4 [38].

Other Chassis Cells

In addition to E. coli and yeast, other microorganisms were also used as chassis for diterpenoids biosynthesis. Green microalgae can convert CO2 into valuable products. The chloroplasts of the photoautotrophic green microalgae (C. reinhardtii) contain large amounts of GGPP. Therefore, it holds tremendous potential as an efficient and sustainable heterologous chassis for the biosynthesis of plant diterpenoids. By combining synthetic biology and metabolic engineering strategies, Einhaus et al. constructed an efficient and stable microalgae chassis to produce sclareol. Then they evaluated and fused GGPP synthase and sclareol synthase from different sources. The yield of sclareol reached 656 ± 9.5 mg/l by optimizing the fermentation conditions [121].

Streptomyces is an important source of natural products. With the development of genome sequencing technology, a variety of terpene synthases have been discovered in Streptomyces. Therefore, Streptomyces are also good chassis to produce diterpenes. Khalid et al. constructed a terpenoid-biosynthetic platform by enhancing precursor supply and screening promoters in Streptomyces reveromyceticus SN-593 [122]. Streptomyces avermitilis SUKA22 is a model strain for gene expression. Yamada et al. expressed seven different diterpene synthases silencing in the original strains and produced 11 new terpenoids, among them, odyverdienes A and B each displaying a novel diterpene skeleton [123].

Summary and Scope

With their amazing structural diversity and broad biological activities, natural products have provided many pharmaceutical agents and intermediates for human beings. Terpenoids are a large group of natural products, among which diterpenoids are rich in structure and have attracted human attention because of their extensive biological activities like antineoplastic activities, anti-inflammatory activities, antibacterial abilities, and many other strong functions. However, their complex and varied structures pose challenges in terms of separation and purification. With the increased demand for bioactive diterpenes, extracting diterpenes from plants is far from satisfying human needs. By analyzing the biosynthetic pathways for plant diterpenes, the yield of diterpenoids can be effectively improved by rational construction and optimization of the biosynthetic pathway in heterologous hosts. The biosynthetic pathways of several diterpenes have been completely or partially resolved in recent years. However, due to the complex structures of diterpenes and the discontinuity of gene clusters in plants, it is a great challenge to elucidate the diterpene biosynthetic pathways. The development of next-generation sequencing techniques and bioinformatic analysis can effectively reduce the candidate genes involved in the biosynthesis of plant diterpenoids. By screening candidate genes in well-established chassis, such as E. coli and yeast, the yield of diterpenoids in the heterologous hosts can be improved efficiently.

Significant progress has been made in the production of diterpenoids in microorganisms recently. Representative studies such as: through the pathway mining, analyzing and assembling the biosynthesis of steviol glycosides, the heterologous synthesis of plant-derived steviol glycosides was realized in E. coli chassis [124]. The natural fragrance sclareol is the raw material of ambergris, whose efficient synthesis was realized through the analysis of its synthesis pathway, reconstruction and optimization in S. cerevisiae, and the global regulation of central metabolic pathway [125]. The field holds immense promise and is of paramount importance for resource development, medicine, and the chemical industry. In early studies, characterizing and expressing key enzymes involved in biosynthesis were an important strategy to increase the yields of diterpenoids. However, the yield improvement was often far from satisfactory. More and more research focus on improving the catalytic efficiency of enzymes by site-directed evolution and truncation or fusion of enzymes to improve the substrate delivery efficiency. In the heterologous hosts, knocking out the competitive pathways or suppressor genes or knocking down their expression by replacing the native promoters with weaker promoters can also reduce the production of by-products. Some diterpenes are cytotoxic, inhibiting the growth of the host, and limiting the accumulation of products. Therefore, enhancing the efflux of compounds and the tolerance of strains are also helpful in improving the yield of compounds. In addition, introducing key genes in the biosynthetic pathways into different chassis may also generate new products, thereby enriching the types of diterpenoids and contributing to the discovery and development of new diterpenes.

In conclusion, obtaining genes related to the diterpenoid biosynthetic pathway from plant genomes is the basis for producing active components utilizing synthetic biology. The metabolic pathway of engineering bacteria was redesigned and constructed using synthetic biology technology, and functional gene elements were assembled into chassis organisms to build efficient cell factories. As research on diterpene components deepens, the demand for diterpenes in medical and natural environment outpaces supply. This comprehensive strategy provides ideas and methods not only for the biosynthesis of diterpenoids but also for other kinds of natural products. With the unlocking of the bottleneck and limiting factors in the biosynthesis of diterpenes, the production and utilization of diterpenoids will also make more significant progress.

Abbreviations

TCM: traditional Chinese medicine. GGPP: (E, E, E)-geranylgeranyl diphosphate. STE-G: steviol glycosides. GAs: gibberellins. NSCLC: non-small cell lung cancer. EMT: epithelial-to-mesenchymal transition. MRSA: methicillin resistant Staphylococcus aureus. IPP: isopentenylpyrophosphate. DMAPP: dimethylallyl pyrophosphate. GPP: geranyl diphosphate. FPP: farnesyl diphosphate. MVA pathway: mevalonic acid pathway. MEP pathway: 2-C-methyl-D-erythritol-4-phosphate pathway. IDI: isopentenyl pyrophosphate isomerase. GGPPS: geranylgeranyl pyrophosphate synthase. cAMP: cyclic adenosine monophosphates. 13R-MO: 13R‐manoyl oxide. ent-CPPS: ent-copalyl diphosphate synthases. ent-KS: ent-kaurene synthases. KA: ent-kaurenoic acid. KO: ent-kaurene oxidase. Reb A: rebaudioside A. Reb D: rebaudioside D. Reb M: rebaudioside M. Reb N: rebaudioside N. TASY: taxadiene synthase. T5αOH: taxadiene‐5α‐hydroxylase. T13αOH: taxane 13α-hydroxylase. TAT: taxane-2α-ol-O-aectyltransferase. DBAT: 10-deacetylbaccatin III-10-O-acetyl transferase. PAM: phenylalanine aminomutase. PCL: β-phenylalanine-CoA ligase. BAPT: taxoid C-13 O-phenylpropanoyltransferase. DBTNBT: 3'-N-debenzoyl-2'-deoxytaxol-N-benzoyltransferase. DXS: 1-deoxyxylulose-5-phosphate synthase. IspA: farnesyl diphosphate synthase.

Conflict of Interest

The authors have no financial conflicts of interest to declare.

Fig 1.

Figure 1.Comprehensive research strategies for plant diterpene resources, including their structures, biosynthetic pathway, heterologous production and biological activity (Adapted from [96, 126-130]).
Journal of Microbiology and Biotechnology 2024; 34: 1563-1579https://doi.org/10.4014/jmb.2402.02014

Fig 2.

Figure 2.The different skeleton types of plant diterpenoids and the corresponding compounds. Acyclic and monocyclic diterpenoids (cyan), bicyclic diterpenoids (blue), tricyclic diterpenoids (yellow), tetracyclic diterpenoids (pink), and macrocyclic diterpenoids(green).
Journal of Microbiology and Biotechnology 2024; 34: 1563-1579https://doi.org/10.4014/jmb.2402.02014

Fig 3.

Figure 3.(A) Biosynthesis of the IPP and DMAPP via the MVA pathway (purple) and MEP pathway (green). Enzyme abbreviations are as follows; AACT, acetoactl-CoA thiolase. HMGS, hydroxymethylglutaryl-CoA synthase. HMGR, hydroxymethylglutaryl-CoA reductase. MVK, mevalonate kinase. PMK, phosphmevalonate kinase. MVD, mevalonate 5- phosphate decarboxylase. IDI, isopentenyl diphosphate isomerase. DXS, 1-deoxy-D-xylulose 5-phosphate synthase. DXR, 1- deoxy-D-xylulose 5-phosphate reductoisomerase. MCT, 2C-methyl-D-erythritol 4-phosphate cytidyltransferase. CMK, 4- (cytidine 5’-diphospho)-2C-methyl-D-erythritol kinase. MDS, 2C-methyl-D-erythritol-2,4’-cyclodiphosphate synthase. HDS, 4-hydroxy-3-methylbut-2-enyl diphosphate synthase. HDR, 4-hydroxy-3-methylbut-2-enyl diphosphate reductase. For intermediates, GAP, D-glyceralfehyde-3-phosphate. DXP, 1-deoxy-D-xylulose 5-phosphate. MEP, 2C-methyl-D-erythritol 4- phosphate. CDP-cytidyl diphosphate. MEcDP, 2C-methyl D-erythritol-2,4-cyclodiphosphate. HMBDP-1-hydroxy-2-methyl- 2-(E)-butenyl-4-diphosphate. OP and OPP signify mono- and diphosphate moieties, respectively. Pi represents inorganic phosphate (B) Biosynthetic pathway of forskolin.
Journal of Microbiology and Biotechnology 2024; 34: 1563-1579https://doi.org/10.4014/jmb.2402.02014

Fig 4.

Figure 4.Biosynthetic pathway of tanshinones. Dash arrowhead indicates that the enzymes have not been identified.
Journal of Microbiology and Biotechnology 2024; 34: 1563-1579https://doi.org/10.4014/jmb.2402.02014

Fig 5.

Figure 5.Biosynthetic pathway of steviol glycosides.
Journal of Microbiology and Biotechnology 2024; 34: 1563-1579https://doi.org/10.4014/jmb.2402.02014

Fig 6.

Figure 6.Biosynthetic pathway of paclitaxel. Blue font indicates that the enzymes have not been identified.
Journal of Microbiology and Biotechnology 2024; 34: 1563-1579https://doi.org/10.4014/jmb.2402.02014

Table 1 . Overview of production of plant diterpenoids in different hosts..

HostDiterpeneMetabolic engineering strategiesTiter (mg/l)Reference
E. colient-kaurane◆ Screening of optimal GGPPS and E. coli host
◆ Overexpression of three key enzymes in MEP pathway
◆ Culturing strain in a bioreactor
578[114]
E. coliSteviol◆ Optimization of the upstream pathway
◆ Screening of proper E. coli host and kaurene oxidase
◆ Truncating the N terminus-modified mutants of kaurene oxidase and attachment of soluble tags
◆ Codon optimization and increasing the copy number of CPR
◆ Introduction of cytochrome b5 (CYB5)
◆ Site-directed mutation of AtCYP714A2
1073.8[115]
E. coliTaxadiene◆ Modularization of the taxadiene metabolic pathway
◆ Using systematic multivariate analysis to achieve a balance of the two modules
1020[9]
S. cerevisiaeMiltiradiene◆ Overexpression of the pathway genes
◆ Downregulation of Erg9
◆ Knocked out transcriptional regulators
◆ Optimization of the medium
◆ Fusion of CfTPS1 and SmKSL1
◆ Protein modification of SmKSL1
3500[117]
S. cerevisiaeForskolin◆ Fusion of BTS1 and ERG20F96C
◆ Overexpression of HMG1
◆ Truncating the N terminus of CfCPR
◆ Fusion of CfCYP76AHs and tR~tB
◆ Regulation of copy numbers of the target genes, amplification of the endoplasmic reticulum (ER) area and cofactor metabolism enhancement
◆ Fed-batch fermentation
21.47[119]
S. cerevisiaeCarnosic acid◆ Overexpression of BTS1-GGGS-ERG20F96Cp
◆ Codon-optimization, N-terminus truncation, and fusion of tSmCPS and tSmKSL
◆ Used SmCPR from S. miltiorrhiza
◆ Co-expression of SmCPR~t28SpCytb5 fusion protein and CYP76AH1
◆ Overexpression of ScCTA1 and ScCTT1
◆ Overexpression of INO2, the HEM3 (heme synthase) gene, and the NADH kinase gene (POS5)
◆ Batch and fed-batch fermentation
24.65[120]
S. cerevisiaeRubusoside◆ KS from G. fujikuroi, KO, CPR1, UGT74G1, and UGT85C2 from S. rebaudiana, KAH from Arabidopsis thaliana
◆ Overexpression of tHMG1 and IDI1
◆ Site-directed mutation of FPSF112A
◆ Replacing promoter of INO2 with a stronger one
◆ Overexpression of the efflux-pump PDR11 and the stress-response factor MSN4
◆ Knocking out GAL7 and overexpression of PGM2
1368.6[94]
Y. lipolyticaGibberellin◆ Downregulating the endogenous squalene synthase gene
◆ Choosing AtCPS, AtKS, AtKO, AtATR2 ang YlCyb5 to producing ent-KA
◆ Codon-optimization of all genes
◆ Gene AtC20ox and AtC3ox were expressed under the control of the strong promotors PrExp and PrTefintron, respectively
◆ N-terminus truncation of AtCPS, AtKS, and AtKO and fusion of CPS and KS
GA4 17.29
GA3 2.93
[38]
C. reinhardtiiSclareol◆ Codon optimization of terpene synthase
◆ All transgenes were driven by the PSAD promoter and FDX1 terminator
◆ Using GGPPS from C. reinhardtii and sclareol synthase from S. sclarea
656[121]

References

  1. Zhu L, Chen L. 2019. Progress in research on paclitaxel and tumor immunotherapy. Cell. Mol. Biol. Lett. 24: 40.
    Pubmed KoreaMed CrossRef
  2. Li ZM, Xu SW, Liu PQ. 2018. Salvia miltiorrhizaBurge (Danshen): a golden herbal medicine in cardiovascular therapeutics. Acta Pharm. Sinica 39: 802-824.
    Pubmed KoreaMed CrossRef
  3. Yaro P, Nie J, Xu M, Zeng K, He H, Yao J, et al. 2019. Influence of organic anion transporter 1/3 on the pharmacokinetics and renal excretion of ginkgolides and bilobalide. J. Ethnopharmacol. 243: 112098.
    Pubmed CrossRef
  4. Arumugam B, Subramaniam A, Alagaraj P. 2020. Stevia as a natural sweetener: A review. Cardiovasc. Hematol. Agents Med. Chem. 18: 94-103.
    Pubmed CrossRef
  5. Rizza A, Jones AM. 2019. The makings of a gradient: spatiotemporal distribution of gibberellins in plant development. Curr. Opin. Plant Biol. 47: 9-15.
    Pubmed KoreaMed CrossRef
  6. Guo X, Zhang RR, Sun JY, Liu Y, Yuan XS, Chen YY, et al. 2024. The molecular mechanism of action for the potent antitumor component extracted using supercritical fluid extraction from Croton crassifolius root. J. Ethnopharmacol. 327: 117835.
    Pubmed CrossRef
  7. Zhu H, Ren X, Huang Y, Su T, Yang L. 2023. Chemical constituents of Euphorbia stracheyi boiss (Euphorbiaceae). Metabolites 13: 852.
    Pubmed KoreaMed CrossRef
  8. Ren X, Yuan X, Jiao SS, He XP, Hu H, Kang JJ, et al. 2023. Clerodane diterpenoids from the Uygur medicine Salvia deserta with immunosuppressive activity. Phytochemistry 214: 113823.
    Pubmed CrossRef
  9. Ajikumar PK, Xiao WH, Tyo KE, Wang Y, Simeon F, Leonard E, et al. 2010. Isoprenoid pathway optimization for Taxol precursor overproduction in Escherichia coli. Science 330: 70-74.
    Pubmed KoreaMed CrossRef
  10. Zhang P, Lee Y, Wei X, Wu J, Liu Q, Wan S. 2018. Enhanced production of tanshinone IIA in endophytic fungi Emericella foeniculicola by genome shuffling. Pharm. Biol. 56: 357-362.
    Pubmed KoreaMed CrossRef
  11. Moon JH, Lee K, Lee JH, Lee PC. 2020. Redesign and reconstruction of a steviol-biosynthetic pathway for enhanced production of steviol in Escherichia coli. Microb. Cell Fact. 19: 20.
    Pubmed KoreaMed CrossRef
  12. Gutbrod K, Romer J, Dörmann P. 2019. Phytol metabolism in plants. Progress Lipid Res. 74: 1-17.
    Pubmed CrossRef
  13. Lin YP, Charng YY. 2021. Chlorophyll dephytylation in chlorophyll metabolism: a simple reaction catalyzed by various enzymes. Plant Sci. 302: 110682.
    Pubmed CrossRef
  14. Xue S, Zhang P, Tang P, Wang C, Kong L, Luo J. 2020. Acyclic diterpene and norsesquiterpene from the seed of Aphanamixis polystachya. Fitoterapia 142: 104518.
    Pubmed CrossRef
  15. Ibrahim SRM, Mohamed GAA. 2017. Tagetones A and B, new cytotoxic monocyclic diterpenoids from flowers of Tagetes minuta. Chin. J. Nat. Med. 15: 546-549.
    Pubmed CrossRef
  16. Zhang J, Chen S, Sun P, Liu Y, Jiang J, Guo C, et al. 2024. Ginkgolides with anti-PAF activity from Ginkgo biloba L. Fitoterapia 175: 105915-105915.
    Pubmed CrossRef
  17. Yu Y, Wang Y, Wang GC, Tan CY, Wang Y, Liu JS, et al. 2023. Andropanilides A-C, the novel labdane-type diterpenoids from Andrographis paniculata and their anti-inflammation activity. Nat. Prod. Bioprospect. 13: 31.
    Pubmed KoreaMed CrossRef
  18. Thekkeveedu RP, Devasya RP. 2024. Biotechnological interventions for the production of forskolin, an active compound from the medicinal plant, Coleus forskohlii. Physiol. Mol. Biol. Plants 30: 213-226.
    Pubmed CrossRef
  19. Li R, Morris-Natschke SL, Lee KH. 2016. Clerodane diterpenes: sources, structures, and biological activities. Nat. Prod. Rep. 33: 1166-1226.
    Pubmed KoreaMed CrossRef
  20. Wang M, Ma C, Chen Y, Li X, Chen J. 2019. Cytotoxic neo-clerodane diterpenoids from Scutellaria barbata D.Don. Chem. Biodivers. 16: e1800499.
    Pubmed CrossRef
  21. Ledoux A, Hamann C, Bonnet O, Jullien K, Quetin-Leclercq J, Tchinda A, et al. 2023. Bioactive clerodane diterpenoids from the leaves of Casearia coriacea vent. Molecules 28: 1197.
    Pubmed KoreaMed CrossRef
  22. Zhang C, Liu JX, Sun MQ, Mian L, Sun L. 2018. Study on fragmentation pathways of three ginkgolides by mass spectrometry. China J. Chin. Mater. Medica 43: 4093-4096.
  23. Liao HJ, Zheng YF, Hong-Yang LI, Peng GP. 2012. Study on antinplatelet aggregation activity induced by PAF of Ginkgolide compounds. J. Liaoning University of Traditional Chinese Medicine.
  24. Hao M, Xu J, Wen H, Du J, Zhang S, Lv M, et al. 2022. Recent advances on biological activities and structural modifications of dehydroabietic acid. Toxins 14: 632.
    Pubmed KoreaMed CrossRef
  25. González MA, Correa-Royero J, Agudelo L, Mesa A, Betancur-Galvis L. 2009. Synthesis and biological evaluation of abietic acid derivatives. Eur. J. Med. Chem. 44: 2468-2472.
    Pubmed CrossRef
  26. Xu J, Zhi X, Zhang Y, Ding R. 2024. Tanshinone IIA alleviates IL-1β-induced chondrocyte apoptosis and inflammation by regulating FBXO11 expression. Clinics 79: 100365.
    Pubmed KoreaMed CrossRef
  27. Habtemariam S. 2023. Anti-inflammatory therapeutic mechanisms of natural products: Insight from rosemary diterpenes, carnosic acid and carnosol. Biomedicines 11: 545.
    Pubmed KoreaMed CrossRef
  28. Yin XW, Bian XX, Chen YF, Zhang M, Wu L, Ren FC, et al. 2023. Structure-diversified terpenoids from Salvia prattii and their protective activity against alcoholic liver diseases. Phytochemistry 214: 113819.
    Pubmed CrossRef
  29. Chen W, Li X, Guo S, Song N, Wang J, Jia L, et al. 2019. Tanshinone IIA harmonizes the crosstalk of autophagy and polarization in macrophages via miR-375/KLF4 pathway to attenuate atherosclerosis. Int. Immunopharmacol. 70: 486-497.
    Pubmed CrossRef
  30. Guan Z, Chen J, Li X, Dong N. 2020. Tanshinone IIA induces ferroptosis in gastric cancer cells through p53-mediated SLC7A11 down-regulation. Biosci. Rep. 40: BSR20201807.
    Pubmed KoreaMed CrossRef
  31. Sun M, Guo B, Xu M, Zhao M, Onakpa MM, Wu Z, et al. 2021. (9βH)- and 17-Nor-Pimaranes from Icacina oliviformis. J. Nat. Prod. 84: 949-955.
    Pubmed KoreaMed CrossRef
  32. Jongsomjainuk O, Boonsombat J, Thongnest S, Prawat H, Batsomboon P, Charoensutthivarakul S, et al. 2023. Kaemtakols A-D, highly oxidized pimarane diterpenoids with potent anti-inflammatory activity from Kaempferia takensis. Nat. Prod. Bioprospect. 13: 55.
    Pubmed KoreaMed CrossRef
  33. Kim C, Le D, Lee M. 2021. Diterpenoids isolated from Podocarpus macrophyllus inhibited the inflammatory mediators in LPSinduced HT-29 and RAW 264.7 cells. Molecules (Basel, Switzerland) 26: 4326.
    Pubmed KoreaMed CrossRef
  34. Zhao X, Cacherat B, Hu Q, Ma D. 2022. Recent advances in the synthesis of ent-kaurane diterpenoids. Nat. Prod. Rep. 39: 119-138.
    Pubmed CrossRef
  35. Liu M, Wang WG, Sun HD, Pu JX. 2017. Diterpenoids from Isodon species: an update. Nat. Prod. Rep. 34: 1090-1140.
    Pubmed CrossRef
  36. Camara MC, Vandenberghe LPS, Rodrigues C, de Oliveira J, Faulds C, Bertrand E, et al. 2018. Current advances in gibberellic acid (GA(3)) production, patented technologies and potential applications. Planta 248: 1049-1062.
    Pubmed CrossRef
  37. Binenbaum J, Weinstain R, Shani E. 2018. Gibberellin localization and transport in plants. Trends Plant Sci. 23: 410-421.
    Pubmed CrossRef
  38. Kildegaard KR, Arnesen JA, Adiego-Pérez B, Rago D, Kristensen M, Klitgaard AK, et al. 2021. Tailored biosynthesis of gibberellin plant hormones in yeast. Metab. Eng. 66: 1-11.
    Pubmed KoreaMed CrossRef
  39. Drummond GJ, Grant PS, Brimble MA. 2021. ent-Atisane diterpenoids: isolation, structure and bioactivity. Nat. Prod. Rep. 38: 330-345.
    Pubmed CrossRef
  40. Wang B, Wei Y, Zhao X, Tian X, Ning J, Zhang B, et al. 2018. Unusual ent-atisane type diterpenoids with 2-oxopropyl skeleton from the roots of Euphorbia ebracteolata and their antiviral activity against human rhinovirus 3 and enterovirus 71. Bioorg. Chem. 81: 234-240.
    Pubmed CrossRef
  41. Maji D, Barnawal D, Gupta A, King S, Singh AK, Kalra A. 2013. A natural plant growth promoter calliterpenone from a plant Callicarpa macrophylla Vahl improves the plant growth promoting effects of plant growth promoting rhizobacteria (PGPRs). World J. Microbiol. Biotechnol. 29: 833-839.
    Pubmed CrossRef
  42. Li CH, Zhang JY, Zhang XY, Li SH, Gao JM. 2019. An overview of grayanane diterpenoids and their biological activities from the Ericaceae family in the last seven years. Eur. J. Med. Chem. 166: 400-416.
    Pubmed CrossRef
  43. Sun N, Zheng G, He M, Feng Y, Liu J, Wang M, et al. 2019. Grayanane diterpenoids from the leaves of Rhododendron auriculatum and their analgesic activities. J. Nat. Prod. 82: 1849-1860.
    Pubmed CrossRef
  44. Leonelli F, Migneco LM, Valletta A, Marini Bettolo R. 2021. Stemodane diterpenes and diterpenoids: Isolation, structure elucidation, biogenesis, biosynthesis, biological activity, biotransformations, metabolites and derivatives biological activity, rearrangements. Molecules 26: 2761.
    Pubmed KoreaMed CrossRef
  45. Leonelli F, Valletta A, Migneco LM, Marini Bettolo R. 2019. Stemarane diterpenes and diterpenoids. Int. J. Mol. Sci. 20: 2627.
    Pubmed KoreaMed CrossRef
  46. Schneider F, Pan L, Ottenbruch M, List T, Gaich T. 2021. The Chemistry of Nonclassical Taxane Diterpene. Acc. Chem. Res. 54: 2347-2360.
    Pubmed CrossRef
  47. Wani MC, Taylor HL, Wall ME, Coggon P, McPhail AT. 1971. Plant antitumor agents. VI. The isolation and structure of taxol, a novel antileukemic and antitumor agent from Taxus brevifolia. J. Am. Chem. Soc. 93: 2325-2327.
    Pubmed CrossRef
  48. Gallego-Jara J, Lozano-Terol G, Sola-Martínez RA, Cánovas-Díaz M, de Diego Puente T. 2020. A compressive review about Taxol(R): History and future challenges. Molecules 25: 5986.
    Pubmed KoreaMed CrossRef
  49. Bouchet BP, Galmarini CM. 2010. Cabazitaxel, a new taxane with favorable properties. Drugs Today 46: 735-742.
    Pubmed CrossRef
  50. Singer JW. 2005. Paclitaxel poliglumex (XYOTAX, CT-2103): a macromolecular taxane. J. Control. Release 109: 120-126.
    Pubmed CrossRef
  51. Abbas MG, Muhammad R, Syed S-u-H, Zafar PR, Zafar PU. 2015. Prostratin: An overview. Mini Rev. Med. Chem. 15: 1122-1130.
    Pubmed CrossRef
  52. Hou P, Zeng Y, Ma B, Bi K, Chen X. 2013. A new cytotoxic cembrane diterpene from the roots of Euphorbia pekinensis Rupr. Fitoterapia 90: 10-13.
    Pubmed CrossRef
  53. Wang K, Yu H, Wu H, Wang X, Pan Y, Chen Y, et al. 2015. A new casbane diterpene from Euphorbia pekinensis. Nat. Prod. Res. 29: 1456-1460.
    Pubmed CrossRef
  54. Fattahian M, Ghanadian M, Ali Z, Khan IA. 2020. Jatrophane and rearranged jatrophane-type diterpenes: biogenesis, structure, isolation, biological activity and SARs (1984-2019). Phytochem. Rev. 19: 265-336.
    Pubmed KoreaMed CrossRef
  55. Wang YF, Su XH, Li LG, Wang W, Zhang ML, Huo CH, et al. 2009. Verticillane derivatives from natural sources. Chem. Biodiver. 6: 1661-1673.
    Pubmed CrossRef
  56. Bai X, Qu H, Zhang J, Li L, Zhang C, Li S, et al. 2024. Effect of steviol glycosides as natural sweeteners on glucose metabolism in adult participants. Food Funct. 15: 2908-3919.
    Pubmed CrossRef
  57. Roy S, Ghosh A, Majie A, Karmakar V, Das S, Dinda SC, et al. 2024. Terpenoids as potential phytoconstituent in the treatment of diabetes: From preclinical to clinical advancement. Phytomedicine 129: 155638.
    Pubmed CrossRef
  58. Grodsky L, Wilson M, Rathinasabapathy T, Komarnytsky S. 2024. Triptolide administration alters immune responses to mitigate insulin resistance in obese states. Biomolecules 14: 395.
    Pubmed KoreaMed CrossRef
  59. Bidart JE, Pertino MW, Schmeda-Hirschmann G, Alché LE, Petrera E. 2023. Antiviral effect of natural and semisynthetic diterpenoids against adenovirus infection in vitro. Planta Med. 89: 1001-1009.
    Pubmed CrossRef
  60. Sadeghi Z, Cerulli A, Marzocco S, Moridi Farimani M, Masullo M, Piacente S. 2023. Anti-inflammatory activity of tanshinonerelated diterpenes from Perovskia artemisioides roots. J. Nat. Prod. 86: 812-821.
    Pubmed KoreaMed CrossRef
  61. Tamuli R, Nguyen T, Macdonald JR, Pierens GK, Fisher GM, Andrews KT, et al. 2023. Isolation and in vitro and in vivo activity of secondary metabolites from Clerodendrum polycephalum baker against Plasmodium Malaria parasites. J. Nat. Prod. 86: 2661-2671.
    Pubmed CrossRef
  62. Deng QD, Lei XP, Zhong YH, Chen MS, Ke YY, Li Z, et al. 2021. Triptolide suppresses the growth and metastasis of non-small cell lung cancer by inhibiting β-catenin-mediated epithelial-mesenchymal transition. Acta Pharmacol. Sinica 42: 1486-1497.
    Pubmed KoreaMed CrossRef
  63. Sarwar MS, Xia YX, Liang ZM, Tsang SW, Zhang HJ. 2020. Mechanistic pathways and molecular targets of plant-derived anticancer ent-Kaurane diterpenes. Biomolecules 10: 144.
    Pubmed KoreaMed CrossRef
  64. Bailly C. 2023. Yuexiandajisu diterpenoids from Euphorbia ebracteolata Hayata (Langdu roots): An overview. Phytochemistry 213: 113784.
    Pubmed CrossRef
  65. Wang X, Wang Y, Liu Z, Zhao H, Yao GD, Liu Q, et al. 2024. New daphnane diterpenoidal 1,3,4-oxdiazole derivatives as potential anti-hepatoma agents: synthesis, biological evaluation and molecular modeling studies. Bioorg. Chem. 145: 107208.
    Pubmed CrossRef
  66. Yazdiniapour Z, Sohrabi MH, Motinia N, Zolfaghari B, Mehdifar P, Ghanadian M, et al. 2023. Diterpenoids from Euphorbia gedrosiaca as potential anti-proliferative agents against breast cancer cells. Metabolites 13: 225.
    Pubmed KoreaMed CrossRef
  67. Wang X, Yang Y, Liu X, Gao X. 2020. Pharmacological properties of tanshinones, the natural products from Salvia miltiorrhiza. Adv. Pharmacol. 87: 43-70.
    CrossRef
  68. Wang W, Wu Y, Chen X, Zhang P, Li H, Chen L. 2019. Synthesis of new ent-labdane diterpene derivatives from andrographolide and evaluation of their anti-inflammatory activities. Eur. J. Med. Chem. 162: 70-79.
    Pubmed CrossRef
  69. Wang Y, Song Z, Guo Y, Xie H, Zhang Z, Sun D, et al. 2021. Diterpenoids from the seeds of Euphorbia lathyris and their antiinflammatory activity. Bioorg. Chem. 112: 104944.
    Pubmed CrossRef
  70. Kim J, Nguyen TTH, Jin J, Septiana I, Son GM, Lee GH, et al. 2019. Anti-cariogenic characteristics of rubusoside. Biotechnol. Bioprocess Eng. 24: 282-287.
    Pubmed KoreaMed CrossRef
  71. Siddique H, Pendry B, Rahman MM. 2019. Terpenes from Zingiber montanum and heir Screening against Multi-Drug Resistant and Methicillin Resistant Staphylococcus aureus. Molecules 24: 385.
    Pubmed KoreaMed CrossRef
  72. Wardana AP, Aminah NS, Rosyda M, Abdjan MI, Kristanti AN, Tun KNW, et al. 2021. Potential of diterpene compounds as antivirals, a review. Heliyon 7: e07777.
    Pubmed KoreaMed CrossRef
  73. Wu Z, Xu H, Xu Y, Fan W, Yao H, Wang Y, et al. 2020. Andrographolide promotes skeletal muscle regeneration after acute injury through epigenetic modulation. Eur. J. Pharmacol. 888: 173470.
    Pubmed CrossRef
  74. Zhang F, Koh GY, Hollingsworth J, Russo PS, Stout RW, Liu Z. 2012. Reformulation of etoposide with solubility-enhancing rubusoside. Int. J. Pharm. 434: 453-459.
    Pubmed CrossRef
  75. Zhang M, Dai T, Feng N. 2017. A novel solubility-enhanced rubusoside-based micelles for increased cancer therapy. Nanoscale Res. Llett. 12: 274.
    Pubmed KoreaMed CrossRef
  76. Shi JB, Wang N, Zhou H, Xu QH, Yan GT. 2019. The role of gibberellin synthase gene GhGA2ox1 in upland cotton (Gossypium hirsutum L.) responses to drought and salt stress. Biotechnol. Appl. Biochem. 66: 298-308.
    Pubmed CrossRef
  77. Li J, Meng X, Yin C, Zhang L, Lin B, Liu P, et al. 2023. Antimalarial and neuroprotective ent-abietane diterpenoids from the aerial parts of Phlogacanthus curviflorus. Chin. J. Nat. Med. 21: 619-630.
    Pubmed CrossRef
  78. Mirza FJ, Zahid S, Holsinger RMD. 2023. Neuroprotective effects of carnosic acid: Insight into its mechanisms of action. Molecules 28: 2306.
    Pubmed KoreaMed CrossRef
  79. Vranová E, Coman D, Gruissem W. 2013. Network analysis of the MVA and MEP pathways for isoprenoid synthesis. Ann. Rev. Plant Biol. 64: 665-700.
    Pubmed CrossRef
  80. Bergman ME, Davis B, Phillips MA. 2019. Medically useful plant terpenoids: biosynthesis, occurrence, and mechanism of action. Molecules 24: 3961.
    Pubmed KoreaMed CrossRef
  81. Zerbe P, Hamberger B, Yuen MM, Chiang A, Sandhu HK, Madilao LL, et al. 2013. Gene discovery of modular diterpene metabolism in nonmodel systems. Plant Physiol. 162: 1073-1091.
    Pubmed KoreaMed CrossRef
  82. Pateraki I, Andersen-Ranberg J, Hamberger B, Heskes AM, Martens HJ, Zerbe P, et al. 2014. Manoyl oxide (13R), the biosynthetic precursor of forskolin, is synthesized in specialized root cork cells in Coleus forskohlii. Plant Physiol. 164: 1222-1236.
    Pubmed KoreaMed CrossRef
  83. Pateraki I, Andersen-Ranberg J, Jensen NB, Wubshet SG, Heskes AM, Forman V, et al. 2017. Total biosynthesis of the cyclic AMP booster forskolin from Coleus forskohlii. ELife 6: e23001.
    Pubmed KoreaMed CrossRef
  84. Mastan A, Rane D, Dastager SG, Vivek Babu CS. 2020. Plant probiotic bacterial endophyte, Alcaligenes faecalis, modulates plant growth and forskolin biosynthesis in Coleus forskohlii. Probiotics Antimicrob. Proteins 12: 481-493.
    Pubmed CrossRef
  85. Gao W, Hillwig ML, Huang L, Cui G, Wang X, Kong J, et al. 2009. A functional genomics approach to tanshinone biosynthesis provides stereochemical insights. Org. Lett. 11: 5170-5173.
    Pubmed KoreaMed CrossRef
  86. Guo J, Zhou YJ, Hillwig ML, Shen Y, Yang L, Wang Y, et al. 2013. CYP76AH1 catalyzes turnover of miltiradiene in tanshinones biosynthesis and enables heterologous production of ferruginol in yeasts. Proc. Natl. Acad. Sci. USA 110: 12108-12113.
    Pubmed KoreaMed CrossRef
  87. Guo J, Ma X, Cai Y, Ma Y, Zhan Z, Zhou YJ, et al. 2016. Cytochrome P450 promiscuity leads to a bifurcating biosynthetic pathway for tanshinones. New Phytol. 210: 525-534.
    Pubmed KoreaMed CrossRef
  88. Mao Y, Ma Y, Chen T, Ma X, Xu Y, Bu J, et al. 2020. Functional integration of two CYP450 genes involved in biosynthesis of tanshinones for improved diterpenoid production by synthetic biology. ACS Synth. Biol. 9: 1763-1770.
    Pubmed CrossRef
  89. Ma Y, Cui G, Chen T, Ma X, Wang R, Jin B, et al. 2021. Expansion within the CYP71D subfamily drives the heterocyclization of tanshinones synthesis in Salvia miltiorrhiza. Nat. Commun. 12: 685.
    Pubmed KoreaMed CrossRef
  90. Deng C, Hao X, Shi M, Fu R, Wang Y, Zhang Y, et al. 2019. Tanshinone production could be increased by the expression of SmWRKY2 in Salvia miltiorrhiza hairy roots. Plant Sci. 284: 1-8.
    Pubmed CrossRef
  91. Zheng H, Jing L, Jiang X, Pu C, Zhao S, Yang J, et al. 2021. The ERF-VII transcription factor SmERF73 coordinately regulates tanshinone biosynthesis in response to stress elicitors in Salvia miltiorrhiza. New Phytol. 231: 1940-1955.
    Pubmed CrossRef
  92. Lv X, Zhang W, Chu S, Zhang H, Wu Y, Zhu Y, et al. 2024. Endophytic fungus Penicillium steckii DF33 promoted tanshinones biosynthesis in Salvia miltiorrhiza by regulating the expression of CYP450 genes. Gene 899: 148094.
    Pubmed CrossRef
  93. Li X, Lin Y, Qin Y, Han G, Wang H, Yan Z. 2024. Beneficial endophytic fungi improve the yield and quality of Salvia miltiorrhiza by performing different ecological functions. PeerJ. 12: e16959.
    Pubmed KoreaMed CrossRef
  94. Xu Y, Wang X, Zhang C, Zhou X, Xu X, Han L, et al. 2022. De novo biosynthesis of rubusoside and rebaudiosides in engineered yeasts. Nat. Commun. 13: 3040.
    Pubmed KoreaMed CrossRef
  95. Wang Y, Sun X, Jia X, Zhu L, Yin H. 2021. Comparative transcriptomic of Stevia rebaudiana provides insight into rebaudioside D and rebaudioside M biosynthesis. Plant Physiol. Biochem. PPB. 167: 541-549.
    Pubmed CrossRef
  96. Zhang J, Tang M, Chen Y, Ke D, Zhou J, Xu X, et al. 2021. Catalytic flexibility of rice glycosyltransferase OsUGT91C1 for the production of palatable steviol glycosides. Nat. Commun. 12: 7030.
    Pubmed KoreaMed CrossRef
  97. Dewitte G, Walmagh M, Diricks M, Lepak A, Gutmann A, Nidetzky B, et al. 2016. Screening of recombinant glycosyltransferases reveals the broad acceptor specificity of stevia UGT-76G1. J. Biotechnol. 233: 49-55.
    Pubmed CrossRef
  98. Chau M, Croteau R. 2004. Molecular cloning and characterization of a cytochrome P450 taxoid 2alpha-hydroxylase involved in Taxol biosynthesis. Arch. Biochem. Biophys. 427: 48-57.
    Pubmed CrossRef
  99. Chau M, Jennewein S, Walker K, Croteau R. 2004. Taxol biosynthesis: Molecular cloning and characterization of a cytochrome P450 taxoid 7 beta-hydroxylase. Chem. Biol. 11: 663-672.
    CrossRef
  100. Schoendorf A, Rithner CD, Williams RM, Croteau RB. 2001. Molecular cloning of a cytochrome P450 taxane 10 beta-hydroxylase cDNA from Taxus and functional expression in yeast. Proc. Natl. Acad. Sci. USA 98: 1501-1506.
    Pubmed KoreaMed CrossRef
  101. Jennewein S, Long RM, Williams RM, Croteau R. 2004. Cytochrome P450 taxadiene 5alpha-hydroxylase, a mechanistically unusual monooxygenase catalyzing the first oxygenation step of taxol biosynthesis. Chem. Biol. 11: 379-387.
    Pubmed CrossRef
  102. Jennewein S, Rithner CD, Williams RM, Croteau RB. 2001. Taxol biosynthesis: taxane 13 alpha-hydroxylase is a cytochrome P450-dependent monooxygenase. Proc. Natl. Acad. Sci. USA 98: 13595-13600.
    Pubmed KoreaMed CrossRef
  103. Walker K, Schoendorf A, Croteau R. 2000. Molecular cloning of a taxa-4(20),11(12)-dien-5alpha-ol-O-acetyl transferase cDNA from Taxus and functional expression in Escherichia coli. Arch. Biochem. Biophys. 374: 371-380.
    Pubmed CrossRef
  104. Walker K, Croteau R. 2000. Molecular cloning of a 10-deacetylbaccatin III-10-O-acetyl transferase cDNA from Taxus and functional expression in Escherichia coli. Proc. Natl. Acad. Sci. USA 97: 583-587.
    Pubmed KoreaMed CrossRef
  105. Walker K, Croteau R. 2000. Taxol biosynthesis: molecular cloning of a benzoyl-CoA:taxane 2alpha-O-benzoyltransferase cDNA from Taxus and functional expression in Escherichia coli. Proc. Natl. Acad. Sci. USA 97: 13591-13596.
    Pubmed KoreaMed CrossRef
  106. Bartsch S, Wybenga GG, Jansen M, Heberling MM, Wu B, Dijkstra BW, et al. 2013. Redesign of a phenylalanine aminomutase into a phenylalanine ammonia lyase. ChemCatChem 5: 1797-1802.
    CrossRef
  107. Walker KD, Klettke K, Akiyama T, Croteau R. 2004. Cloning, heterologous expression, and characterization of a phenylalanine aminomutase involved in Taxol biosynthesis. J. Biol. Chem. 279: 53947-53954.
    Pubmed CrossRef
  108. Walker K, Fujisaki S, Long R, Croteau R. 2002. Molecular cloning and heterologous expression of the C-13 phenylpropanoid side chain-CoA acyltransferase that functions in Taxol biosynthesis. Proc. Natl. Acad. Sci. USA 99: 12715-12720.
    Pubmed KoreaMed CrossRef
  109. Walker K, Long R, Croteau R. 2002. The final acylation step in taxol biosynthesis: cloning of the taxoid C13-side-chain Nbenzoyltransferase from Taxus. Proc. Natl. Acad. Sci. USA 99: 9166-9171.
    Pubmed KoreaMed CrossRef
  110. Zhang Y, Wiese L, Fang H, Alseekh S, Perez de Souza L, Scossa F, et al. 2023. Synthetic biology identifies the minimal gene set required for paclitaxel biosynthesis in a plant chassis. Mol. Plant 16: 1951-1961.
    Pubmed CrossRef
  111. Paddon CJ, Westfall PJ, Pitera DJ, Benjamin K, Fisher K, McPhee D, et al. 2013. High-level semi-synthetic production of the potent antimalarial artemisinin. Nature 496: 528-532.
    Pubmed CrossRef
  112. Dai Z, Wang B, Liu Y, Shi M, Wang D, Zhang X, et al. 2014. Producing aglycons of ginsenosides in bakers' yeast. Sci. Rep. 4: 3698.
    Pubmed KoreaMed CrossRef
  113. Zhang B, Yao Z, Liu Z, Zheng Y. 2021. Metabolic engineering of Escherichia coli for L-homoserine production. Sheng wu gong cheng xue bao Chin. J. Biotechnol. 37: 1287-1297.
  114. Kong MK, Kang HJ, Kim JH, Oh SH, Lee PC. 2015. Metabolic engineering of the Stevia rebaudiana ent-kaurene biosynthetic pathway in recombinant Escherichia coli. J. Biotechnol. 214: 95-102.
    Pubmed CrossRef
  115. Sun Y, Chen Z, Wang G, Lv H, Mao Y, Ma K, et al. 2022. De novo production of versatile oxidized kaurene diterpenes in Escherichia coli. Metab. Eng. 73: 201-213.
    Pubmed CrossRef
  116. Dong H, Chen S, Zhu J, Gao K, Zha W, Lin P, et al. 2020. Enhance production of diterpenoids in yeast by overexpression of the fused enzyme of ERG20 and its mutant mERG20. J. Biotechnol. 307: 29-34.
    Pubmed CrossRef
  117. Hu T, Zhou J, Tong Y, Su P, Li X, Liu Y, et al. 2020. Engineering chimeric diterpene synthases and isoprenoid biosynthetic pathways enables high-level production of miltiradiene in yeast. Metab. Eng. 60: 87-96.
    Pubmed CrossRef
  118. Forman V, Bjerg-Jensen N, Dyekjær JD, Møller BL, Pateraki I. 2018. Engineering of CYP76AH15 can improve activity and specificity towards forskolin biosynthesis in yeast. Microb. Cell Fact. 17: 181.
    Pubmed KoreaMed CrossRef
  119. Ju H, Zhang C, He S, Nan W, Lu W. 2022. Construction and optimization of Saccharomyces cerevisiae for synthesizing forskolin. Appl. Microbiol. Biotechnol. 106: 1933-1944.
    Pubmed CrossRef
  120. Wei P, Zhang C, Bian X, Lu W. 2022. Metabolic engineering of Saccharomyces cerevisiae for heterologous carnosic acid production. Front. Bioeng. Biotechnol. 10: 916605.
    Pubmed KoreaMed CrossRef
  121. Einhaus A, Steube J, Freudenberg RA, Barczyk J, Baier T, Kruse O. 2022. Engineering a powerful green cell factory for robust photoautotrophic diterpenoid production. Metab. Eng. 73: 82-90.
    Pubmed CrossRef
  122. Khalid A, Takagi H, Panthee S, Muroi M, Chappell J, Osada H, et al. 2017. Development of a terpenoid-production platform in Streptomyces reveromyceticus SN-593. ACS Synth. Biol. 6: 2339-2349.
    Pubmed CrossRef
  123. Yamada Y, Arima S, Nagamitsu T, Johmoto K, Uekusa H, Eguchi T, et al. 2015. Novel terpenes generated by heterologous expression of bacterial terpene synthase genes in an engineered Streptomyces host. J. Antibiotics 68: 385-394.
    Pubmed KoreaMed CrossRef
  124. Wang J, Li S, Xiong Z, Wang Y. 2015. Pathway mining-based integration of critical enzyme parts for de novo biosynthesis of steviolglycosides sweetener in Escherichia coli. Cell Res. 26: 258-261.
    Pubmed KoreaMed CrossRef
  125. Xuan C, Wei Y, Yu C, Shan Y, K ZZ, Jens N, et al. 2022. Engineering yeast for high-level production of diterpenoid sclareol. Metab. Eng. 75: 19-28.
    Pubmed CrossRef
  126. Norkin M, Ordóñez-Morán P, Huelsken J. 2021. High-content, targeted RNA-seq screening in organoids for drug discovery in colorectal cancer. Cell Rep. 35: 109026.
    Pubmed CrossRef
  127. Dong Y, Zhao Q, Wang Y. 2021. Network pharmacology-based investigation of potential targets of astragalus membranaceousangelica sinensis compound acting on diabetic nephropathy. Sci. Rep. 11: 19496.
    Pubmed KoreaMed CrossRef
  128. Murthy BN, Sinha S, Surolia A, Indi SS, Jayaraman N. 2008. SPR and ITC determination of the kinetics and the thermodynamics of bivalent versus monovalent sugar ligand-lectin interactions. Glycoconj. J. 25: 313-321.
    Pubmed CrossRef
  129. Ferreira LG, Dos Santos RN, Oliva G, Andricopulo AD. 2015. Molecular docking and structure-based drug design strategies. Molecules 20: 13384-13421.
    Pubmed KoreaMed CrossRef
  130. Xu X, Yuan H, Yu X, Huang S, Sun Y, Zhang T, et al. 2021. The chromosome-level Stevia genome provides insights into steviol glycoside biosynthesis. Hortic. Res. 8: 129.
    Pubmed KoreaMed CrossRef