전체메뉴
검색
Article Search

JMB Journal of Microbiolog and Biotechnology

QR Code QR Code

Review

References

  1. Ohnishi Y, Ishikawa J, Hara H, Suzuki H, Ikenoya M, Ikeda H, et al. 2008. Genome sequence of the streptomycin-producing microorganism Streptomyces griseus IFO 13350. J. Bacteriol. 190: 4050-4060.
    Pubmed PMC CrossRef
  2. Khan ST, Komaki H, Motohashi K, Kozone I, Mukai A, Takagi M, et al. 2011. Streptomyces associated with a marine sponge Haliclona sp.; biosynthetic genes for secondary metabolites and products. Environ. Microbiol. 13: 391-403.
    Pubmed CrossRef
  3. Onaka H. 2017. Novel antibiotic screening methods to awaken silent or cryptic secondary metabolic pathways in actinomycetes. J. Antibiot. (Tokyo) 70: 865-870.
    Pubmed CrossRef
  4. Lim FY, Sanchez JF, Wang CC, Keller NP. 2012. Toward awakening cryptic secondary metabolite gene clusters in filamentous fungi. Methods Enzymol. 517: 303-324.
    Pubmed PMC CrossRef
  5. Medema MH, Alam MT, Breitling R, Takano E. 2011. The future of industrial antibiotic production: from random mutagenesis to synthetic biology. Bioeng. Bugs. 2: 230-233.
    Pubmed CrossRef
  6. Nguyen QT, Merlo ME, Medema MH, Jankevics A, Breitling R, Takano E. 2012. Metabolomics methods for the synthetic biology of secondary metabolism. FEBS Lett. 586: 2177-2183.
    Pubmed CrossRef
  7. Yang YH, Song E, Lee BR, Kim EJ, Park SH, Kim YG, et al. 2010. Rapid functional screening of Streptomyces coelicolor regulators by use of a pH indicator and application to the MarR-like regulator AbsC. Appl. Environ. Microbiol. 76: 3645-3656.
    Pubmed PMC CrossRef
  8. Myronovskyi M, Luzhetskyy A. 2016. Native and engineered promoters in natural product discovery. Nat. Prod. Rep. 33: 1006-1019.
    Pubmed CrossRef
  9. Ziemert N, Alanjary M, Weber T. 2016. The evolution of genome mining in microbes -a review. Nat. Prod. Rep. 33: 988-1005.
    Pubmed CrossRef
  10. Harrison J, Studholme DJ. 2014. Recently published Streptomyces genome sequences. Microb. Biotechnol. 7: 373-380.
    Pubmed PMC CrossRef
  11. Bentley SD, Chater KF, Cerdeno-Tarraga AM, Challis GL, Thomson NR, James KD, et al. 2002. Complete genome sequence of the model actinomycete Streptomyces coelicolor A3(2). Nature 417: 141-147.
    Pubmed CrossRef
  12. Komaki H, Sakurai K, Hosoyama A, Kimura A, Igarashi Y, Tamura T. 2018. Diversity of nonribosomal peptide synthetase and polyketide synthase gene clusters among taxonomically close Streptomyces strains. Sci. Rep. 8: 6888.
    Pubmed PMC CrossRef
  13. Starcevic A, Zucko J, Simunkovic J, Long PF, Cullum J, Hranueli D. 2008. ClustScan: an integrated program package for the semi-automatic annotation of modular biosynthetic gene clusters and in silico prediction of novel chemical structures. Nucleic Acids Res. 36: 6882-6892.
    Pubmed PMC CrossRef
  14. Li MH, Ung PM, Zajkowski J, Garneau-Tsodikova S, Sherman DH. 2009. Automated genome mining for natural products. BMC Bioinformatics 10: 185.
    Pubmed PMC CrossRef
  15. Skinnider MA, Dejong CA, Rees PN, Johnston CW, Li H, Webster AL, et al. 2015. Genomes to natural products prediction informatics for secondary metabolomes (PRISM). Nucleic Acids Res. 43: 9645-9662.
    Pubmed PMC CrossRef
  16. Blin K, Wolf T, Chevrette MG, Lu X, Schwalen CJ, Kautsar SA, et al. 2017. antiSMASH 4.0-improvements in chemistry prediction and gene cluster boundary identification. Nucleic Acids Res. 45: W36-W41.
    Pubmed PMC CrossRef
  17. Weber T, Kim HU. 2016. The secondary metabolite bioinformatics portal: computational tools to facilitate synthetic biology of secondary metabolite production. Synth. Syst. Biotechnol. 1: 69-79.
    Pubmed PMC CrossRef
  18. Low ZJ, Pang LM, Ding Y, Cheang QW, Le Mai Hoang K, Thi Tran H, et al. 2018. Identification of a biosynthetic gene cluster for the polyene macrolactam sceliphrolactam in a Streptomyces strain isolated from mangrove sediment. Sci. Rep. 8: 1594.
    Pubmed PMC CrossRef
  19. Jia N, Ding MZ, Luo H, Gao F, Yuan YJ. 2017. Complete genome sequencing and antibiotics biosynthesis pathways analysis of Streptomyces lydicus 103. Sci. Rep. 7: 44786.
  20. Zhang G, Yu D, Sang B, Feng J, Han L, Zhang X. 2017. Genome-wide analysis reveals the secondary metabolome in Streptomyces kanasensis ZX01. Genes (Basel) 8: 346.
    Pubmed PMC CrossRef
  21. Ansari MZ, Yadav G, Gokhale RS, Mohanty D. 2004. NRPS-PKS: a knowledge-based resource for analysis of NRPS/PKS megasynthases. Nucleic Acids Res. 32: W405-413.
    Pubmed PMC CrossRef
  22. Kim MS, Cho WJ, Song MC, Park SW, Kim K, Kim E, et al. 2017. Engineered biosynthesis of milbemycins in the avermectin high-producing strain Streptomyces avermitilis. Microb. Cell Fact. 16: 9.
    Pubmed PMC CrossRef
  23. Ikeda H, Ishikawa J, Hanamoto A, Shinose M, Kikuchi H, Shiba T, et al. 2003. Complete genome sequence and comparative analysis of the industrial microorganism Streptomyces avermitilis. Nat. Biotechnol. 21: 526-531.
    Pubmed CrossRef
  24. Bode HB, Bethe B, Hofs R, Zeeck A. 2002. Big effects from small changes: possible ways to explore nature’s chemical diversity. Chembiochem. 3: 619-627.
  25. Marmann A, Aly AH, Lin W, Wang B, Proksch P. 2014. Co-cultivation--a powerful emerging tool for enhancing the chemical diversity of microorganisms. Mar. Drugs. 12: 1043-1065.
    Pubmed PMC CrossRef
  26. Baral B, Akhgari A, Metsa-Ketela M. 2018. Activation of microbial secondary metabolic pathways: Avenues and challenges. Synth. Syst. Biotechnol. 3: 163-178.
    Pubmed PMC CrossRef
  27. King AA, Chater KF. 1986. The expression of the Escherichia coli lacZ gene in Streptomyces. J. Gen. Microbiol. 132: 1739-1752.
    Pubmed CrossRef
  28. Lussier FX, Denis F, Shareck F. 2010. Adaptation of the highly productive T7 expression system to Streptomyces lividans. Appl. Environ. Microbiol. 76: 967-970.
    Pubmed PMC CrossRef
  29. Mitra A, Angamuthu K, Jayashree HV, Nagaraja V. 2009. Occurrence, divergence and evolution of intrinsic terminators across eubacteria. Genomics 94: 110-116.
    Pubmed CrossRef
  30. Bibb MJ, White J, Ward JM, Janssen GR. 1994. The mRNA for the 23S rRNA methylase encoded by the ermE gene of Saccharopolyspora erythraea is translated in the absence of a conventional ribosome-binding site. Mol. Microbiol. 14: 533-545.
    Pubmed CrossRef
  31. Bibb MJ, Janssen GR, Ward JM. 1985. Cloning and analysis of the promoter region of the erythromycin resistance gene (ermE) of Streptomyces erythraeus. Gene 38: 215-226.
    Pubmed CrossRef
  32. Kieser T, Bibb M, Buttner M, Chater K, Hopwood D. 2000. Practical Streptomyces Genetics. John Innes Foundation, Norwich, England.
  33. Wang W, Li X, Wang J, Xiang S, Feng X, Yang K. 2013. An engineered strong promoter for Streptomycetes. Appl. Environ. Microbiol. 79: 4484-4492.
    Pubmed PMC CrossRef
  34. Labes G, Bibb M, Wohlleben W. 1997. Isolation and characterization of a strong promoter element from the Streptomyces ghanaensis phage I19 using the gentamicin resistance gene (aacC1) of Tn 1696 as reporter. Microbiology 143: 1503-1512.
    Pubmed CrossRef
  35. Shao Z, Rao G, Li C, Abil Z, Luo Y, Zhao H. 2013. Refactoring the silent spectinabilin gene cluster using a plug-and-play scaffold. ACS Synth. Biol. 2: 662-669.
    Pubmed PMC CrossRef
  36. Sohoni SV, Fazio A, Workman CT, Mijakovic I, Lantz AE. 2014. Synthetic promoter library for modulation of actinorhodin production in Streptomyces coelicolor A3(2). PLoS One 9: e99701.
    Pubmed PMC CrossRef
  37. Seghezzi N, Amar P, Koebmann B, Jensen PR, Virolle MJ. 2011. The construction of a library of synthetic promoters revealed some specific features of strong Streptomyces promoters. Appl. Microbiol. Biotechnol. 90: 615-623.
    Pubmed CrossRef
  38. Luo Y, Zhang L, Barton KW, Zhao H. 2015. Systematic identification of a panel of strong constitutive promoters from Streptomyces albus. ACS Synth. Biol. 4: 1001-1010.
    Pubmed CrossRef
  39. Li S, Wang J, Li X, Yin S, Wang W, Yang K. 2015. Genome-wide identification and evaluation of constitutive promoters in Streptomycetes. Microb. Cell Fact. 14: 172.
    Pubmed PMC CrossRef
  40. Murakami T, Holt TG, Thompson CJ. 1989. Thiostrepton-induced gene expression in Streptomyces lividans. J. Bacteriol. 171: 1459-1466.
    Pubmed PMC CrossRef
  41. Takano E, White J, Thompson CJ, Bibb MJ. 1995. Construction of thiostrepton-inducible, high-copy-number expression vectors for use in Streptomyces spp. Gene 166: 133-137.
    Pubmed CrossRef
  42. Huang H, Zheng G, Jiang W, Hu H, Lu Y. 2015. One-step high-efficiency CRISPR/Cas9-mediated genome editing in Streptomyces. Acta Biochim. Biophys. Sin (Shanghai) 47: 231-243.
    Pubmed CrossRef
  43. Rodriguez-Garcia A, Combes P, Perez-Redondo R, Smith MC, Smith MC. 2005. Natural and synthetic tetracycline-inducible promoters for use in the antibiotic-producing bacteria Streptomyces. Nucleic Acids Res. 33: e87.
    Pubmed PMC CrossRef
  44. Horbal L, Fedorenko V, Luzhetskyy A. 2014. Novel and tightly regulated resorcinol and cumate-inducible expression systems for Streptomyces and other actinobacteria. Appl. Microbiol. Biotechnol. 98: 8641-8655.
    Pubmed CrossRef
  45. Herai S, Hashimoto Y, Higashibata H, Maseda H, Ikeda H, Omura S, et al. 2004. Hyper-inducible expression system for Streptomycetes. Proc. Natl. Acad. Sci. USA 101: 14031-14035.
    Pubmed PMC CrossRef
  46. Noguchi Y, Kashiwagi N, Uzura A, Ogino C, Kondo A, Ikeda H, et al. 2018. Development of a strictly regulated xylose-induced expression system in Streptomyces. Microb. Cell Fact. 17: 151.
    Pubmed PMC CrossRef
  47. Hindle Z, Smith CP. 1994. Substrate induction and catabolite repression of the Streptomyces coelicolor glycerol operon are mediated through the GylR protein. Mol. Microbiol. 12: 737-745.
    Pubmed CrossRef
  48. Ward JM, Janssen GR, Kieser T, Bibb MJ, Buttner MJ, Bibb MJ. 1986. Construction and characterisation of a series of multi-copy promoter-probe plasmid vectors for Streptomyces using the aminoglycoside phosphotransferase gene from Tn5 as indicator. Mol. Gen. Genet. 203: 468-478.
    Pubmed CrossRef
  49. Pulido D, Jimenez A, Salas M, Mellado RP. 1987. A Bacillus subtilis phage phi 29 transcription terminator is efficiently recognized in Streptomyces lividans. Gene 56: 277-282.
    Pubmed CrossRef
  50. Cobb RE, Wang Y, Zhao H. 2015. High-efficiency multiplex genome editing of Streptomyces species using an engineered CRISPR/Cas system. ACS Synth. Biol. 4: 723-728.
    Pubmed PMC CrossRef
  51. West S, Proudfoot NJ. 2009. Transcriptional termination enhances protein expression in human cells. Mol. Cell. 33: 354-364.
    Pubmed PMC CrossRef
  52. Mapendano CK, Lykke-Andersen S, Kjems J, Bertrand E, Jensen TH. 2010. Crosstalk between mRNA 3’ end processing and transcription initiation. Mol. Cell. 40: 410-422.
  53. Dar D, Shamir M, Mellin JR, Koutero M, Stern-Ginossar N, Cossart P, et al. 2016. Term-seq reveals abundant ribo-regulation of antibiotics resistance in bacteria. Science 352: aad9822.
    Pubmed PMC CrossRef
  54. Lu P, Vogel C, Wang R, Yao X, Marcotte EM. 2007. Absolute protein expression profiling estimates the relative contributions of transcriptional and translational regulation. Nat. Biotechnol. 25: 117-124.
    Pubmed CrossRef
  55. Jeong Y, Kim JN, Kim MW, Bucca G, Cho S, Yoon YJ, et al. 2016. The dynamic transcriptional and translational landscape of the model antibiotic producer Streptomyces coelicolor A3(2). Nat. Commun. 7: 11605.
    Pubmed PMC CrossRef
  56. Makrides SC. 1996. Strategies for achieving high-level expression of genes in Escherichia coli. Microbiol. Rev. 60: 512-538.
    Pubmed PMC
  57. Na D, Lee D. 2010. RBSDesigner: software for designing synthetic ribosome binding sites that yields a desired level of protein expression. Bioinformatics 26: 2633-2634.
    Pubmed CrossRef
  58. Bai C, Zhang Y, Zhao X, Hu Y, Xiang S, Miao J, et al. 2015. Exploiting a precise design of universal synthetic modular regulatory elements to unlock the microbial natural products in Streptomyces. Proc. Natl. Acad. Sci. USA 112: 12181-12186.
    Pubmed PMC CrossRef
  59. Yi JS, Kim MW, Kim M, Jeong Y, Kim EJ, Cho BK, et al. 2017. A novel approach for gene expression optimization through native promoter and 5’ UTR combinations based on RNA-seq, Ribo-seq, and TSS-seq of Streptomyces coelicolor. ACS Synth. Biol. 6: 555-565.
  60. Janssen GR, Bibb MJ. 1990. Tandem promoters, tsrp1 and tsrp2, direct transcription of the thiostrepton resistance gene (tsr) of Streptomyces azureus: transcriptional initiation from tsrp2 occurs after deletion of the -35 region. Mol. Gen. Genet. 221: 339-346.
    Pubmed CrossRef
  61. Sohaskey CD, Im H, Nelson AD, Schauer AT. 1992. Tn4556 and luciferase: synergistic tools for visualizing transcription in Streptomyces. Gene 115: 67-71.
    Pubmed CrossRef
  62. Ingram C, Brawner M, Youngman P, Westpheling J. 1989. xylE functions as an efficient reporter gene in Streptomyces spp.: use for the study of galP1, a catabolite-controlled promoter. J. Bacteriol. 171: 6617-6624.
    Pubmed PMC CrossRef
  63. Myronovskyi M, Welle E, Fedorenko V, Luzhetskyy A. 2011. Beta-glucuronidase as a sensitive and versatile reporter in actinomycetes. Appl. Environ Microbiol. 77: 5370-5383.
    Pubmed PMC CrossRef
  64. Flores FJ, Rincon J, Martin JF. 2003. Characterization of the iron-regulated desA promoter of Streptomyces pilosus as a system for controlled gene expression in actinomycetes. Microb. Cell Fact. 2: 5.
    Pubmed PMC CrossRef
  65. Willemse J, van Wezel GP. 2009. Imaging of Streptomyces coelicolor A3(2) with reduced autofluorescence reveals a novel stage of FtsZ localization. PLoS One 4: e4242.
    Pubmed PMC CrossRef
  66. Santos-Beneit F, Errington J. 2017. Green fluorescent protein as a reporter for the spatial and temporal expression of actIII in Streptomyces coelicolor. Arch. Microbiol. 199: 875-880.
    Pubmed PMC CrossRef
  67. Nguyen KD, Au-Young SH, Nodwell JR. 2007. Monomeric red fluorescent protein as a reporter for macromolecular localization in Streptomyces coelicolor. Plasmid 58: 167-173.
    Pubmed CrossRef
  68. Phelan RM, Sachs D, Petkiewicz SJ, Barajas JF, Blake-Hedges JM, Thompson MG, et al. 2017. Development of next generation synthetic biology tools for use in Streptomyces venezuelae. ACS Synth. Biol. 6: 159-166.
    Pubmed CrossRef
  69. Siegl T, Luzhetskyy A. 2012. Actinomycetes genome engineering approaches. Antonie Van Leeuwenhoek. 102: 503-516.
    Pubmed CrossRef
  70. Herrmann S, Siegl T, Luzhetska M, Petzke L, Jilg C, Welle E, et al. 2012. Site-specific recombination strategies for engineering actinomycete genomes. Appl. Environ. Microbiol. 78: 1804-1812.
    Pubmed PMC CrossRef
  71. Khodakaramian G, Lissenden S, Gust B, Moir L, Hoskisson PA, Chater KF, et al. 2006. Expression of Cre recombinase during transient phage infection permits efficient marker removal in Streptomyces. Nucleic Acids Res. 34: e20.
    Pubmed PMC CrossRef
  72. Zelyas N, Tahlan K, Jensen SE. 2009. Use of the native flp gene to generate in-frame unmarked mutations in Streptomyces spp. Gene 443: 48-54.
    Pubmed CrossRef
  73. Barrangou R, Fremaux C, Deveau H, Richards M, Boyaval P, Moineau S, et al. 2007. CRISPR provides acquired resistance against viruses in prokaryotes. Science 315: 1709-1712.
    Pubmed CrossRef
  74. Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, et al. 2013. Multiplex genome engineering using CRISPR/Cas systems. Science 339: 819-823.
    Pubmed PMC CrossRef
  75. DiCarlo JE, Norville JE, Mali P, Rios X, Aach J, Church GM. 2013. Genome engineering in Saccharomyces cerevisiae using CRISPR-Cas systems. Nucleic Acids Res. 41: 4336-4343.
    Pubmed PMC CrossRef
  76. Friedland AE, Tzur YB, Esvelt KM, Colaiacovo MP, Church GM, Calarco JA. 2013. Heritable genome editing in C. elegans via a CRISPR-Cas9 system. Nat. Methods 10: 741-743.
    Pubmed PMC CrossRef
  77. Jiang W, Bikard D, Cox D, Zhang F, Marraffini LA. 2013. RNA-guided editing of bacterial genomes using CRISPR-Cas systems. Nat. Biotechnol. 31: 233-239.
    Pubmed PMC CrossRef
  78. Li JF, Norville JE, Aach J, McCormack M, Zhang D, Bush J, et al. 2013. Multiplex and homologous recombination-mediated genome editing in Arabidopsis and Nicotiana benthamiana using guide RNA and Cas9. Nat. Biotechnol. 31: 688-691.
    Pubmed PMC CrossRef
  79. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. 2012. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337: 816-821.
    Pubmed PMC CrossRef
  80. Bibikova M, Beumer K, Trautman JK, Carroll D. 2003. Enhancing gene targeting with designed zinc finger nucleases. Science 300: 764.
    Pubmed CrossRef
  81. Christian M, Cermak T, Doyle EL, Schmidt C, Zhang F, Hummel A, et al. 2010. Targeting DNA double-strand breaks with TAL effector nucleases. Genetics 186: 757-761.
    Pubmed PMC CrossRef
  82. Rouet P, Smih F, Jasin M. 1994. Introduction of double-strand breaks into the genome of mouse cells by expression of a rare-cutting endonuclease. Mol. Cell Biol. 14: 8096-8106.
    Pubmed PMC CrossRef
  83. Doudna JA, Charpentier E. 2014. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science 346: 1258096.
    Pubmed CrossRef
  84. Anders C, Niewoehner O, Duerst A, Jinek M. 2014. Structural basis of PAM-dependent target DNA recognition by the Cas9 endonuclease. Nature 513: 569-573.
    Pubmed PMC CrossRef
  85. Mojica FJ, Diez-Villasenor C, Garcia-Martinez J, Almendros C. 2009. Short motif sequences determine the targets of the prokaryotic CRISPR defence system. Microbiology 155: 733-740.
    Pubmed CrossRef
  86. Tong Y, Charusanti P, Zhang L, Weber T, Lee SY. 2015. CRISPR-Cas9 based engineering of actinomycetal genomes. ACS Synth. Biol. 4: 1020-1029.
    Pubmed CrossRef
  87. Zhang MM, Wong FT, Wang Y, Luo S, Lim YH, Heng E, et al. 2017. CRISPR-Cas9 strategy for activation of silent Streptomyces biosynthetic gene clusters. Nat. Chem. Biol. 13: 607-609.
    Pubmed PMC CrossRef
  88. Zeng H, Wen S, Xu W, He Z, Zhai G, Liu Y, et al. 2015. Highly efficient editing of the actinorhodin polyketide chain length factor gene in Streptomyces coelicolor M145 using CRISPR/Cas9-CodA(sm) combined system. Appl. Microbiol. Biotechnol. 99: 10575-10585.
    Pubmed CrossRef
  89. Li L, Zheng G, Chen J, Ge M, Jiang W, Lu Y. 2017. Multiplexed site-specific genome engineering for over-producing bioactive secondary metabolites in actinomycetes. Metab. Eng. 40: 80-92.
    Pubmed CrossRef
  90. Meng J, Feng R, Zheng G, Ge M, Mast Y, Wohlleben W, et al. 2017. Improvement of pristinamycin I (PI) production in Streptomyces pristinaespiralis by metabolic engineering approaches. Synth. Syst. Biotechnol. 2: 130-136.
    Pubmed PMC CrossRef
  91. Qin Z, Munnoch JT, Devine R, Holmes NA, Seipke RF, Wilkinson KA, et al. 2017. Formicamycins, antibacterial polyketides produced by Streptomyces formicae isolated from African Tetraponera plant-ants. Chem. Sci. 8: 3218-3227.
    Pubmed PMC CrossRef
  92. Wang Y, Cobb RE, Zhao H. 2016. High-efficiency genome editing of Streptomyces species by an engineered CRISPR/Cas System. Methods Enzymol. 575: 271-284.
    Pubmed CrossRef
  93. Jia H, Zhang L, Wang T, Han J, Tang H, Zhang L. 2017. Development of a CRISPR/Cas9-mediated gene-editing tool in Streptomyces rimosus. Microbiology 163: 1148-1155.
    Pubmed CrossRef
  94. Tao W, Yang A, Deng Z, Sun Y. 2018. CRISPR/Cas9-based editing of Streptomyces for discovery, characterization, and production of natural products. Front Microbiol. 9: 1660.
    Pubmed PMC CrossRef
  95. Liu Y, Tao W, Wen S, Li Z, Yang A, Deng Z, et al. 2015. In vitro CRISPR/Cas9 system for efficient targeted DNA editing. MBio 6: e01714-01715.
    Pubmed PMC CrossRef
  96. Jiang W, Zhao X, Gabrieli T, Lou C, Ebenstein Y, Zhu TF. 2015. Cas9-assisted targeting of chromosome segments CATCH enables one-step targeted cloning of large gene clusters. Nat. Commun. 6: 8101.
    Pubmed PMC CrossRef
  97. Kang HS, Charlop-Powers Z, Brady SF. 2016. Multiplexed CRISPR/Cas9-and TAR-mediated promoter engineering of natural product biosynthetic gene clusters in yeast. ACS Synth. Biol. 5: 1002-1010.
    Pubmed PMC CrossRef
  98. Tao W, Yurkovich ME, Wen S, Lebe KE, Samborskyy M, Liu Y, et al. 2016. A genomics-led approach to deciphering the mechanism of thiotetronate antibiotic biosynthesis. Chem. Sci. 7: 376-385.
    Pubmed PMC CrossRef
  99. Li L, Wei K, Liu X, Wu Y, Zhensg G, Chen S, et al. 2019. aMSGE: advanced multiplex site-specific genome engineering with orthogonal modular recombinases in actinomycetes. Metab. Eng. 52: 7.
    Pubmed CrossRef
  100. Qi LS, Larson MH, Gilbert LA, Doudna JA, Weissman JS, Arkin AP, et al. 2013. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 152: 1173-1183.
    Pubmed PMC CrossRef
  101. Zhao Y, Li L, Zheng G, Jiang W, Deng Z, Wang Z, et al. 2018. CRISPR/dCas9-mediated multiplex gene repression in Streptomyces. Biotechnol. J. 13: e1800121.
    Pubmed CrossRef
  102. Xu Z, Wang Y, Chater KF, Ou HY, Xu HH, Deng Z, et al. 2017. Large-scale transposition mutagenesis of Streptomyces coelicolor identifies hundreds of genes influencing antibiotic biosynthesis. Appl. Environ. Microbiol. 83: e02889-02816.
    Pubmed PMC CrossRef
  103. Komatsu M, Uchiyama T, Omura S, Cane DE, Ikeda H. 2010. Genome-minimized Streptomyces host for the heterologous expression of secondary metabolism. Proc. Natl. Acad. Sci. USA 107: 2646-2651.
    Pubmed PMC CrossRef
  104. Gomez-Escribano JP, Bibb MJ. 2011. Engineering Streptomyces coelicolor for heterologous expression of secondary metabolite gene clusters. Microb. Biotechnol. 4: 207-215.
    Pubmed PMC CrossRef
  105. Zetsche B, Gootenberg JS, Abudayyeh OO, Slaymaker IM, Makarova KS, Essletzbichler P, et al. 2015. Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system. Cell 163: 759-771.
    Pubmed PMC CrossRef
  106. Li L, Wei K, Zheng G, Liu X, Chen S, Jiang W, et al. 2018. CRISPR-Cpf1-assisted multiplex genome editing and transcriptional repression in Streptomyces. Appl. Environ. Microbiol. 84: e00827-00818.
    Pubmed PMC CrossRef
  107. Xu M, Wright GD. 2018. Heterologous expression-facilitated natural products’ discovery in actinomycetes. J. Ind. Microbiol. Biotechnol. 46: 415-431.
  108. Liu R, Deng Z, Liu T. 2018. Streptomyces species: Ideal chassis for natural product discovery and overproduction. Metab. Eng. 50: 74-84.
    Pubmed CrossRef
  109. Nah HJ, Pyeon HR, Kang SH, Choi SS, Kim ES. 2017. Cloning and heterologous expression of a large-sized natural product biosynthetic gene cluster in Streptomyces Species. Front. Microbiol. 8: 394.
    Pubmed PMC CrossRef
  110. Rutledge PJ, Challis GL. 2015. Discovery of microbial natural products by activation of silent biosynthetic gene clusters. Nat. Rev. Microbiol. 13: 509-523.
    Pubmed CrossRef
  111. Katz M, Hover BM, Brady SF. 2016. Culture-independent discovery of natural products from soil metagenomes. J. Ind. Microbiol Biotechnol. 43: 129-141.
    Pubmed CrossRef
  112. Pyeon HR, Nah HJ, Kang SH, Choi SS, Kim ES. 2017. Heterologous expression of pikromycin biosynthetic gene cluster using Streptomyces artificial chromosome system. Microb. Cell Fact. 16: 96.
    Pubmed PMC CrossRef
  113. Nah HJ, Woo MW, Choi SS, Kim ES. 2015. Precise cloning and tandem integration of large polyketide biosynthetic gene cluster using Streptomyces artificial chromosome system. Microb. Cell Fact. 14: 140.
    Pubmed PMC CrossRef
  114. Du D, Wang L, Tian Y, Liu H, Tan H, Niu G. 2015. Genome engineering and direct cloning of antibiotic gene clusters via phage varphiBT1 integrase-mediated site-specific recombination in Streptomyces. Sci Rep. 5: 8740.
  115. Greunke C, Duell ER, Glockle A, Lamm K, Gulder TAM, D’Agostino PM. 2018. Direct pathway cloning (DiPaC) to unlock natural product biosynthetic potential. Metab. Eng. 47: 334-345.
    Pubmed CrossRef
  116. Shao Z, Luo Y, Zhao H. 2011. Rapid characterization and engineering of natural product biosynthetic pathways via DNA assembler. Mol. Biosyst. 7: 1056-1059.
    Pubmed PMC CrossRef
  117. Fu J, Bian X, Hu S, Wang H, Huang F, Seibert PM, et al. 2012. Full-length RecE enhances linear-linear homologous recombination and facilitates direct cloning for biopro-specting. Nat. Biotechnol. 30: 440-446.
    Pubmed CrossRef
  118. Yamanaka K, Reynolds KA, Kersten RD, Ryan KS, Gonzalez DJ, Nizet V, et al. 2014. Direct cloning and refactoring of a silent lipopeptide biosynthetic gene cluster yields the antibiotic taromycin A. Proc. Natl. Acad. Sci. USA 111: 1957-1962.
    Pubmed PMC CrossRef
  119. Orr-Weaver TL, Szostak JW, Rothstein RJ. 1981. Yeast transformation: a model system for the study of recombination. Proc. Natl. Acad. Sci USA 78: 6354-6358.
    Pubmed PMC CrossRef
  120. Lee NC, Larionov V, Kouprina N. 2015. Highly efficient CRISPR/Cas9-mediated TAR cloning of genes and chromosomal loci from complex genomes in yeast. Nucleic Acids Res. 43: e55.
    Pubmed PMC CrossRef
  121. Bu QT, Yu P, Wang J, Li ZY, Chen XA, Mao XM, et al. 2019. Rational construction of genome-reduced and high-efficient industrial Streptomyces chassis based on multiple comparative genomic approaches. Microb. Cell Fact. 18: 16.
    Pubmed PMC CrossRef
  122. Myronovskyi M, Rosenkranzer B, Nadmid S, Pujic P, Normand P, Luzhetskyy A. 2018. Generation of a cluster-free Streptomyces albus chassis strains for improved heterologous expression of secondary metabolite clusters. Metab. Eng. 49: 316-324.
    Pubmed CrossRef
  123. Xu M, Wang Y, Zhao Z, Gao G, Huang SX, Kang Q, et al. 2016. Functional genome mining for metabolites encoded by large gene clusters through heterologous expression of a whole-genome bacterial artificial chromosome library in Streptomyces spp. Appl. Environ. Microbiol. 82: 5795-5805.
    Pubmed PMC CrossRef
  124. Liu Q, Xiao L, Zhou Y, Deng K, Tan G, Han Y, et al. 2016. Development of Streptomyces sp. FR-008 as an emerging chassis. Synth. Syst. Biotechnol. 1: 207-214.
    Pubmed PMC CrossRef
  125. Komatsu M, Komatsu K, Koiwai H, Yamada Y, Kozone I, Izumikawa M, et al. 2013. Engineered Streptomyces avermitilis host for heterologous expression of biosynthetic gene cluster for secondary metabolites. ACS Synth. Biol. 2: 384-396.
    Pubmed PMC CrossRef
  126. Daniels W, Bouvin J, Busche T, Kalinowski J, Bernaerts K. 2016. Finding targets for genome reduction in Streptomyces lividans TK24 using flux balance analysis. IFAC-PapersOnLine 49: 252-257.
  127. Toro L, Pinilla L, Avignone-Rossa C, Rios-Estepa R. 2018. An enhanced genome-scale metabolic reconstruction of Streptomyces clavuligerus identifies novel strain improvement strategies. Bioprocess Biosyst. Eng. 41: 657-669.
    Pubmed CrossRef
  128. Kallifidas D, Jiang G, Ding Y, Luesch H. 2018. Rational engineering of Streptomyces albus J1074 for the overexpression of secondary metabolite gene clusters. Microb. Cell Fact. 17: 25.
    Pubmed PMC CrossRef
  129. Machado H, Tuttle RN, Jensen PR. 2017. Omics-based natural product discovery and the lexicon of genome mining. Curr. Opin. Microbiol. 39: 136-142.
    Pubmed PMC CrossRef
  130. Ran FA, Cong L, Yan WX, Scott DA, Gootenberg JS, Kriz AJ, et al. 2015. In vivo genome editing using Staphylococcus aureus Cas9. Nature 520: 186-191.
    Pubmed PMC CrossRef
  131. Liu JJ, Orlova N, Oakes BL, Ma E, Spinner HB, Baney KLM, et al. 2019. CasX enzymes comprise a distinct family of RNA-guided genome editors. Nature 566: 218-223.
    Pubmed PMC CrossRef
  132. Bikard D, Jiang W, Samai P, Hochschild A, Zhang F, Marraffini LA. 2013. Programmable repression and activation of bacterial gene expression using an engineered CRISPR-Cas system. Nucleic Acids Res. 41: 7429-7437.
    Pubmed PMC CrossRef
  133. Siu KH, Chen W. 2019. Riboregulated toehold-gated gRNA for programmable CRISPR-Cas9 function. Nat. Chem. Biol. 15: 217-220.
    Pubmed CrossRef
  134. Bunet R, Song L, Mendes MV, Corre C, Hotel L, Rouhier N, et al. 2011. Characterization and manipulation of the pathway-specific late regulator AlpW reveals Streptomyces ambofaciens as a new producer of Kinamycins. J. Bacteriol. 193: 1142-1153.
    Pubmed PMC CrossRef
  135. Miao V, Coeffet-Legal MF, Brian P, Brost R, Penn J, Whiting A, et al. 2005. Daptomycin biosynthesis in Streptomyces roseosporus: cloning and analysis of the gene cluster and revision of peptide stereochemistry. Microbiology 151: 1507-1523.
    Pubmed CrossRef
  136. Myronovskyi M, Rosenkranzer B, Luzhetskyy A. 2014. Iterative marker excision system. Appl. Microbiol. Biotechnol. 98: 4557-4570.
    Pubmed CrossRef
  137. Thapa LP, Oh TJ, Lee HC, Liou K, Park JW, Yoon YJ, et al. 2007. Heterologous expression of the kanamycin biosynthetic gene cluster (pSKC2) in Streptomyces venezuelae YJ003. Appl. Microbiol. Biotechnol. 76: 1357-1364.
    Pubmed CrossRef
  138. Jones AC, Gust B, Kulik A, Heide L, Buttner MJ, Bibb MJ. 2013. Phage p1-derived artificial chromosomes facilitate heterologous expression of the FK506 gene cluster. PLoS One 8: e69319.
    Pubmed PMC CrossRef
  139. Steffensky M, Muhlenweg A, Wang ZX, Li SM, Heide L. 2000. Identification of the novobiocin biosynthetic gene cluster of Streptomyces spheroides NCIB 11891. Antimicrob. Agents Chemother. 44: 1214-1222.
    Pubmed PMC CrossRef
  140. Zhao Z, Shi T, Xu M, Brock NL, Zhao YL, Wang Y, et al. 2016. Hybrubins: Bipyrrole tetramic acids obtained by crosstalk between a truncated undecylprodigiosin pathway and heterologous tetramic acid biosynthetic genes. Org. Lett. 18: 572-575.
    Pubmed CrossRef

Related articles in JMB

More Related Articles

Article

Review

J. Microbiol. Biotechnol. 2019; 29(5): 667-686

Published online May 28, 2019 https://doi.org/10.4014/jmb.1904.04015

Copyright © The Korean Society for Microbiology and Biotechnology.

Synthetic Biology Tools for Novel Secondary Metabolite Discovery in Streptomyces

Namil Lee 1, Soonkyu Hwang 1, Yongjae Lee 1, Suhyung Cho 1, Bernhard Palsson 3, 4, 5 and Byung-Kwan Cho 1, 2*

1Department of Biological Sciences and KI for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
2Intelligent Synthetic Biology Center, Daejeon 34141, Republic of Korea
3Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA 4Department of Pediatrics, University of California San Diego, La Jolla, CA, 92093, USA
5Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, 2800, Denmark

Correspondence to:Byung-Kwan  Cho
bcho@kaist.ac.kr

Received: April 10, 2019; Accepted: May 11, 2019

Abstract

Streptomyces are attractive microbial cell factories that have industrial capability to produce a wide array of bioactive secondary metabolites. However, the genetic potential of the Streptomyces species has not been fully utilized because most of their secondary metabolite biosynthetic gene clusters (SM-BGCs) are silent under laboratory culture conditions. In an effort to activate SM-BGCs encoded in Streptomyces genomes, synthetic biology has emerged as a robust strategy to understand, design, and engineer the biosynthetic capability of Streptomyces secondary metabolites. In this regard, diverse synthetic biology tools have been developed for Streptomyces species with technical advances in DNA synthesis, sequencing, and editing. Here, we review recent progress in the development of synthetic biology tools for the production of novel secondary metabolites in Streptomyces, including genomic elements and genome engineering tools for Streptomyces, the heterologous gene expression strategy of designed biosynthetic gene clusters in the Streptomyces chassis strain, and future directions to expand diversity of novel secondary metabolites.

Keywords: Streptomyces, secondary metabolites, biosynthetic gene cluster, antibiotics, synthetic biology, genome editing, CRISPR/Cas9, heterologous expression

Introduction

Streptomyces species are Gram-positive bacteria that produce more than two-thirds of the medically and agriculturally important secondary metabolites, including antibiotic, anticancer, antifungal, antiparasitic, and immuno-suppressive compounds [1, 2]. Recent genome sequencing efforts have revealed that individual Streptomyces species have a wealth of genetic potential to produce novel secondary metabolites. However, most secondary metabolite biosynthetic gene clusters (SM-BGCs) are silent under laboratory culture conditions, limiting effective use of Streptomyces [3]. Therefore, activation of silent SM-BGCs is one of the most promising approaches to discover novel bioactive secondary metabolites from Streptomyces.

To activate silent SM-BGCs, various strategies have been applied, including culture media modifications, chemical or antibiotic treatments, heterologous gene expression in different hosts, and co-culture with cohabiting microbes [4]. However, these methods are untargeted, resulting in non-directed activation of silent SM-BGCs in Streptomyces. To overcome this limitation, a synthetic biology approach has been proposed to redesign and reconstruct target SM-BGCs [5, 6]. Generally, a main obstacle in activating cryptic SM-BGCs is the difficulty in bypassing the complex native regulation. Expression of SM-BGCs is tightly governed by multi-layered regulatory networks, which are often triggered by environmental signals [7]. Using synthetic genetic parts, such as promoters, ribosome binding sites (RBS), and terminators, which are not controlled by the host’s regulatory system, expression of target SM-BGC can be achieved through bypassing the native regulatory networks. In this respect, various synthetic genetic parts and genome engineering tools are required to regulate gene expression and redesign target SM-BGCs in Streptomyces. Even though high GC content and highly interconnected regulatory networks challenge the application of synthetic genetic parts that are widely used in other bacterial systems to Streptomyces, enabling synthetic biology tools have been recently developed for Streptomyces engineering [8]. Here, we review synthetic biology strategies applied in the discovery of novel secondary metabolites in Streptomyces. As synthetic biology is defined by an iterative “design-build-test” cycle in engineering biology, we have classified the strategy into three steps (Fig. 1). First, for the design step, discovery of novel SM-BGCs via genome mining is described, followed by designing of SM-BGCs using various synthetic genetic parts developed for Streptomyces. Second, for the build step, the process of building the re-designed SM-BGCs is discussed, including genome engineering of original host and SM-BGC assembly into plasmids for heterologous expression. Further, Streptomyces chassis strains are suggested as optimal heterologous expression hosts for reconstructing target SM-BGCs. Third, for the test step, high-throughput testing methods are described to measure activation of target SM-BGCs for the next “design-build-test” cycle.

Figure 1. Overview of synthetic biology strategy to produce novel secondary metabolite from Streptomyces. Abbreviations: BGC, biosynthetic gene cluster; CDS, coding sequence; 5’ UTR, 5’ untranslated region; RBS, ribosome binding site; WT, wild type; LA, left homology arm; RA, right homology arm.

Mining of Secondary Metabolite Biosynthetic Gene Clusters

In the pre-genome mining era, most of the secondary metabolites in Streptomyces were discovered via chemistry-based methods of changing culture conditions and mass spectrometry or NMR biosynthetic identification [9]. Due to the biochemical complexity of SM-BGCs, however, the discovery of novel secondary metabolites has been challenging. This limitation has been overcome through a genome mining approach, in which recent advances in DNA sequencing technology caused a rapid increase in the number of high-quality Streptomyces genome sequences [10, 11]. To date, about 500 scaffold-level and 84 complete-level genome sequences of Streptomyces strains are available in the NCBI database. These large numbers of genome sequences encode invaluable resources for novel secondary metabolite discovery [12]. To identify SM-BGCs from these genome sequences, several genome-mining tools have been developed, such as ClustSCAN, NP.searcher, GNP/PRISM, and antiSMASH [13-16]. Among them, antiSMASH is the most comprehensive and widely used software pipeline for genome mining, offering a user-friendly web interface and prediction of the broad spectrum of SM-BGCs [16]. antiSMASH has detected 45 different classes of SM-BGCs based on a rule-based cluster detection approach and further predicted the modular domain structures of genes within these SM-BGCs [17].

In general, each Streptomyces genome encodes approximately 30 SM-BGCs, which are diverse and differ between species, indicating that Streptomyces strains are an immeasurable source of novel secondary metabolites [18-20]. Information about SM-BGCs mined from genome sequences is not only essential data for novel secondary metabolite discovery, but also a resource to facilitate rational design of SM-BGCs based on the synthetic biology approach. In particular, polyketides (PK) and nonribosomal peptides (NRP) can be redesigned using this approach as they are synthesized by serially connected modular enzymes that recognize module-specific CoAs or amino acids, respectively [21]. For example, replacement of AveA1 and module 7 of AveA3 in avermectin BGC of Streptomyces avermitilis with MilA1 and MilA3 in milbemycin BGC of S. hygroscopicus resulted in milbemycin production in S. avermitilis [22]. Taken together, a genome mining approach can accelerate secondary metabolite discovery at an unprecedented rate with an ever-growing number of Streptomyces genome sequences.

Genetic Parts for Streptomyces Synthetic Biology

Genomic information on Streptomyces strains has revealed the great potential of Streptomyces to produce novel secondary metabolites. However, most SM-BGCs in Streptomyces are inactive under general laboratory culture conditions. For example, although S. coelicolor, S. griseus, and S. avermitilis genomes encode more than 30 SM-BGCs, respectively, only 3–5 secondary metabolites have been detected [1, 11, 23]. Although many efforts have been made to activate silent SM-BGCs in Streptomyces strains, including the One Strain-Many Compounds (OSMAC) strategy and co-cultivation methods [24, 25], these approaches resulted in non-directed activation of silent BGCs. For specific activation of targeted SM-BGCs, the following methods have been implemented: (1) promoter replacement, (2) overexpression or repression of regulatory genes, (3) heterologous expression in different hosts, and (4) refactoring of targeted SM-BGCs [26]. To this end, the number of genetic parts, such as promoters, ribosome-binding sites (RBS), and terminators available for Streptomyces species, has dramatically increased as summarized in Table 1.

Table 1 . Genetic parts for Streptomyces..

Genetic partsFeatureReference
Constitutive promoters
ermE* promoterMutation at the promoter of the erythromycin resistance gene of Streptomyces erythraeus[31]
SF14P promoterGenome of Streptomyces ghanaensis phage I19[34]
kasOP promoterPromoter of SARP family regulator in Stretpomyces coelicolor A3[33]
gapdh promoterPromoter of glyceraldehyde-3-phosphate dehydrogenase in Streptomyces griseus[35]
rpsL promoterPromoter of 30S ribosomal protein S12 in Streptomyces griseus[35]
195 native or synthetic promotersHigh-throughput screening in S. venezueale[58]
32 native promotersTranscriptome data-based selection in S. albus[38]
166 native promotersTranscriptome data-based selection in S. coelicolor[39]
2 native promotersMulti-omics data-based selection in S. coelicolor[59]
Inducible promoters
tipA promoterThiostrepton-induced promoter[40]
nitA promoterε-caprolactam-induced promoter[45]
xylA promoterXylose-induced promoter[46]
tcp830Tetracycline-induced promoter[43]
PA3-rolOResorcinol-induced promoter[44]
P21-cmtCumate-induced promoter[44]
Terminators
FdBidirectional transcription termination originated from E. coli phage fd[48]
TD1Bidirectional transcription termination originated from Bacillus subtilis phage Φ29[49]
RBS
AAAGGAGGTypical RBS sequence of S. coelicolor[134]
192 native or synthetic RBSsHigh-throughput screening in S. venezueale[58]
4 native RBSsMulti-omics data-based selection in S. coelicolor[59]
Reporter genes
luxAB cassetten-Decanal as substrate; absorbance at 490 nm wavelength[61]
amy geneSoluble starch with 3,5-dinitrosalycilic acid (DNS) as substrate; absorbance at 540 nm wavelength[64]
xylE geneCatecol as substrate; absorbance at 375 nm wavelength[62]
gusA genep-Nitrophenyl-β-D-glucuronide as substrate; absorbance at 415 nm wavelength[63]
eGFPGreen fluorescent protein; excitation wavelength 470-490 nm and emission wavelength 515 nm[65]
sfGFPGreen fluorescent protein; excitation wavelength 488 nm and emission wavelength 500~550 nm[58]
mRFPRed fluorescent protein; excitation wavelength 584 nm and emission wavelength 607 nm[67]
mCherryRed fluorescent protein; excitation wavelength 587 nm and emission wavelength 610 nm[68]


Genetic Parts for Transcriptional Regulation in Streptomyces

In bacterial cells, a transcription unit is defined as a basic unit of regulation and is composed of several genes and accessory genetic elements, including promoters, transcrip-tion start sites (TSS), RBSs, and terminators. Design and utilization of these genetic parts, at appropriate strengths, is critical for precise transcriptional and translational regulation of targeted gene expression. In transcriptional regulation, the most important genetic part is the promoter, which is the binding site of RNA polymerase to initiate transcription. However, widely used promoters for bacterial genetic engineering, such as lacZ and T7 promoters, are not directly applicable in Streptomyces [27, 28], thus only a few promoters have been used for gene expression in Streptomyces. The terminator is also important in preventing transcriptional read-through to downstream genes [29]. Particularly, when redesigning SM-BGCs in Streptomyces, due to their operon-like genomic structure, precise transcriptional termination is required between transcription units. In this section, we described genetic parts that have been developed and used to regulate the transcription of Streptomyces.

First, constitutive promoters, which generate constant gene expression levels regardless of growth phases, are extensively used for SM-BGC expression in Streptomyces. In particular, ermE* promoter is the most commonly used strong constitutive promoter in Streptomyces, which is a derivative of the ermE promoter, containing a trinucleotide deletion in the ermEp1 region of the erythromycin resistance gene in S. erythraeus [30-32]. Further, SF14P and kasOP, discovered from the S. ghanaensis phage I19 genome and promoter of the SARP family regulator in S. coelicolor A3, respectively, constitutively transcribe gene expression similarly or more strongly than the ermE* promoter [33, 34]. Additionally, two strong constitutive promoters, gapdhP and rpsLP, which have higher activity than the ermE* promoter, were obtained from the promoter region of housekeeping genes in the S. griseus genome [35]. In addition, several strategies have been applied to develop strong constitutive promoters. One approach screens strong synthetic promoters from a randomized promoter library. For example, the randomized sequence library of the kasO promoter was used to generate synthetic promoters with promoter strength ranging from 0.95% to 187.5%, compared to the parental kasO promoter [33]. Similar approaches have been conducted on the ermE and actII-orf4 promoters of S. coelicolor [36]. Several strong promoters were screened through this approach; however, all promoters were weaker than the ermE* promoter [37]. Another rational strategy identifies strong promoters using gene expression data. In S. albus, promoter sequences of highly expressed genes were selected based on transcrip-tional profiling data, and the strength of each promoter was tested, resulting in the selection of ten promoters that were stronger than the ermE* promoter [38]. A similar study was performed on S. coelicolor, which identified 166 potentially constitutive promoters across the Streptomyces genus [39].

Second, expression of SM-BGC genes under constitutive promoters sometimes causes growth retardation. Therefore, it is desirable to establish a controllable gene expression system in Streptomyces. To date, the most widely used inducible promoter in Streptomyces is the tipA promoter, which is induced by thiostrepton treatment [40, 41]. The basal expression level of the tipA promoter is considerable, which limits precise regulation of the targeted gene expression. However, basal expression of the tipA promoter is sometimes used to maintain low expression levels of toxic genes [42]. The tetracycline-induced strong promoter, tcp830, was constructed by combining conserved sequences of the ermE promoter and Tn10 tetR/tetO systems [43]. However, like tipA promoter, a major disadvantage of this promoter is its basal expression level. To overcome this limitation, inducible promoters with low leaky expression, such as PA3-rolO and P21-cmt promoters, have been synthesized. The PA3-rolO promoter is a resorcinol-induced promoter synthesized by combining the rolO operator and synthetic promoter PA3. The P21-cmt promoter is a cumate-induced expression system synthesized by fusing the operator of the Pseudomonas putida F1 cumate degradation operon to the P21 synthetic promoter [44]. Other inducible systems used in Streptomyces are nitA and xylA promoters. The nitA promoter, which originates from the nitrilase promoter of Rhodococcus rhodochrous, is induced by a complex of ε-caprolactam and the transcription regulator NitR [45]. The recently developed xylA promoter is a strictly regulated xylose-induced expression promoter [46]. Additionally, several glycerol-inducible systems have been developed for Streptomyces; however, these systems have not been utilized because glycerol treatment may alter intrinsic cellular metabolism [47].

Further, only a limited number of terminator sequences is available in Streptomyces strains. Two bidirectional transcription terminators, Fd originated from Escherichia coli phage fd and TD1 originated from Bacillus subtilis phage ϕ29, are efficiently recognized in Streptomyces [48, 49]. Although lambda T0 and T7 terminators have been used in several Streptomyces vectors, these terminators have not been systematically validated in Streptomyces for their effects on gene expression levels [50]. While most studies have focused on promoter strength as a determinant of gene expression levels, transcription terminators also have crucial roles in recycling transcription complexes and ultimately gene expression level [51, 52]. To expand the repertoire of transcription terminators in Streptomyces, understanding transcription termination and identifying native terminator sequences in Streptomyces are required. In this regard, Term-seq, which is a recent RNA sequencing method that enables genome-wide determination of transcript 3’ end positions, is a suitable technique for screening terminator sequences in Streptomyces genomes [53].

Taken together, many efforts in molecular biology of Streptomyces have provided genetic parts to control gene expression, including constitutive promoters, inducible promoters, and terminators. However, to flexibly and precisely control secondary metabolite production in Streptomyces, more generalized and validated genetic parts for transcription, which can be used in various Streptomyces species and at different strengths, have to be developed.

Genetic Parts for Translational Regulation in Streptomyces

Because cellular protein level does not directly correlate with mRNA abundance, yet depends on translational efficiency, transcriptional regulation alone is not sufficient to design an efficient gene expression system in Streptomyces [54, 55]. Translational efficiency is primarily determined by 5’ untranslated regions (5’-UTR), RBS, and codon usage of target genes [56]. For example, the RBS contains the Shine Dalgarno (SD) sequence, which includes a complementary sequence with the 3’ end of the 16s rRNA region of the 30S ribosomal subunit. Sequence diversity and accessibility of the SD sequence influence binding affinity with the ribosome, determining translational efficiency [57]. In this regard, several studies have measured the strength of the 5’-UTR and RBS in Streptomyces species to expand genetic parts for Streptomyces engineering. In this section, we will provide two examples of translational regulatory genetic part screenings in Streptomyces genomes.

First, in S. venezuelae, sequences of the strongest RBS among 15 native RBSs were selected and randomized. After comparing the RBS strength, 177 synthetic RBSs with activity over 200-fold compared to their parental RBSs were collected. Furthermore, seven promoters were combined with nine RBSs in a pairwise manner to screen the most optimal promoter-RBS set for gene expression [58]. Second, two promoters and four 5’-UTR sequences were selected from S. coelicolor based on multi-omics data, including TSS-seq, RNA-seq, and Ribo-seq. Pairwise sets of promoters and 5’-UTR sequences showed strength in a range of 0.03-to 2.4-fold, compared to the ermE* promoter with the SD sequence of the nitA gene [59]. Although development of a translational regulatory genetic part is at the beginning stage, as compared to transcription, ultimately transcriptional and translational genetic parts with various strengths have to be combined and utilized to design and control enzyme stoichiometry in SM-BGCs for enhancing secondary metabolite production.

Reporter Systems for High-Throughput Screening

For high-throughput characterization of genetic parts developed for Streptomyces, reporter systems that rapidly represent gene expression level with minimal influence to the cell physiology are required. Although many antibiotic resistance genes have been used as conventional markers for gene expression, their effects on cellular metabolism and narrow dynamic range limit their suitability for gene expression quantification [32, 60]. To quantify gene expression, colorimetric methods are more appropriate because gene expression levels can be quantified by measuring the absorbance of a specific wavelength of light. To date, various colorimetric methods, including luxAB, amy, xylE, and gusA, have been employed in Streptomyces strains [61-64]. In particular, gusA is the most widely used colorimetric reporter system in Streptomyces strains [44, 59]. However, colorimetric reporter systems are based on an enzymatic reaction that requires an additional substrate treatment, which may affect cellular metabolism (Table 1). For example, catechol dioxygenase, encoded by xylE, produces hydroxymuconic semialdehyde with a yellow color from catechol as a substrate [62]. In contrast, fluorescent proteins do not require any supplemental reagents and thus are suitable for high-throughput screening using fluorescence-activated cell sorting (FACS). However, since Streptomyces have relatively high levels of autofluorescence, several studies have argued that fluorescent proteins are not appropriate for high-throughput screening of Streptomyces strains [63]. Despite this limitation, several efforts have been made to apply fluorescent proteins in Streptomyces [65]. Among the fluorescent proteins, GFP derivatives, such as eGFP and sfGFP, are widely used in Streptomyces, and mRFP has been implemented to enable multi-color fluorescence-based studies [58, 65-67]. To test the available fluorescent proteins in S. venezuelae, seven fluorescent proteins (mTagBFP, mCerulean, mTFP, sfGFP, mCherry, mKate, and mCardinal) were expressed, and the fluorescence intensity of each fluorescent protein was compared. Among the seven fluorescent proteins, mCherry protein showed the most significant reduction in signal-to-background noise level and was used for further studies to characterize genetic parts of S. venezuelae [68].

CRISPR/Cas-Based Genome-Engineering Tools for Streptomyces

To utilize genetic parts and reconstruct metabolic pathways for secondary metabolite production, efficient genome engineering tools are required. Conventional genome engineering of Streptomyces is heavily dependent on either single or double crossover of a plasmid, which is laborious and time-consuming to obtain the desired clones [32]. Furthermor, the use of selection markers, such as antibiotics resistance genes, is often required to avoid reversion of the engineered genotype to wild type and may confer undesired effects, including polar effects. To overcome this limitation, site-specific recombination strategies, including Cre/loxP, Dre/rox and Flp/FRT, have been exploited in Streptomyces [69-72]. However, these approaches retain recombinase recognition sites in the chromosome and may limit successive applications for multiple genetic manipulations.

Recently, CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats and CRISPR-associated protein 9) has emerged as a promising tool for genome engineering of Streptomyces strains [50, 73-78]. Briefly, the Cas9 endo-nuclease forms a complex with guide RNA (gRNA) and is guided to a protospacer sequence, which is complementary to a spacer sequence of gRNA [79]. The guided Cas9 induces a double-strand break (DSB) in the genome, followed by repairing the DSB via native non-homologous end joining repair (NHEJ) or homology directed repair (HDR) mecha-nisms which facilitate genome engineering. CRISPR/Cas9 systems are superior to simple homologous recombination or site-specific recombination systems as they facilitate unmarked genome engineering with reduced time and labor. Even though other genome engineering strategies, such as zinc finger nuclease and transcription activator-like effector nuclease, have been developed, the protein-mediated recognition of target DNA sequence requires design of appropriate proteins for individual target sequences and thus limits their applications [80-82]. Therefore, the CRISPR/Cas9 system has become a dominant genome engineering tool, outpacing their performance (Table 2) [83].

Table 2 . Application of CRISPR/Cas9-mediated engineering in Streptomyces..

CasTargetStrategyVectorRepairOrganismRelated secondary metaboliteRemarkRef
SpCas9actI-orf1DisruptionpCRISPR-Cas9NHEJS. coelicolorACTReconstituted NHEJ with ligD expression[86]
actVBDisruptionpCRISPR-Cas9NHEJS. coelicolorACTReconstituted NHEJ with ligD expression
actI-orf2DisruptionpWHUNHEJS. coelicolorACTcodA(sm)-based screening system for plasmid-cured strain[88]
zwf2DisruptionpCRISPomycesNHEJS. rimosus-Oxytetracycline production enhancement by disruption of competitive gene[93]
devBDisruptionpCRISPomycesNHEJS. rimosus-Oxytetracycline production enhancement by disruption of competitive gene
sshg_00040 - sshg_00050DeletionpCRISPomycesHDR S. albusLanthipeptide-[50]
sshg_05713DeletionpCRISPomycesHDR S. albusPolycylic tetramic acid macrolactam-
Formicamycin clusterDeletionpCRISPomycesHDRS. formicaeFormicamycin-[91]
forVDeletionpCRISPomycesHDRS. formicaeFormicamycin-
actVA-orf5DeletionpCRISPomycesHDRS. lividansACT-[50]
redD-redFDeletionpCRISPomycesHDRS. lividansRED-
redNDeletionpCRISPomycesHDRS. lividansRED-
actVA-orf5 and redNDeletionpCRISPomycesHDRS. lividansACT and REDMultiplexed editing
devBDeletionpCRISPomycesHDRS. rimosus-Oxytetracycline production enhancement by disruption of competitive gene[93]
zwf2DeletionpCRISPomycesHDRS. rimosus-Oxytetracycline production enhancement by disruption of competitive gene
phpDDeletionpCRISPomycesHDRS. viridochromogenesPhosphinothricin tripeptide-[50]
phpMDeletionpCRISPomycesHDRS. viridochromogenesPhosphinothricin tripeptide-
sceNDeletionpCRISPR-Cas9HDRStreptomyces sp.SD85BGC11 (sceliphrolactam)-[18]
sceQ-sceR fusionDeletionpCRISPR-Cas9HDRStreptomyces sp.SD85BGC11 (sceliphrolactam)Fusion of sceQ and sce R by deleting stop codon of sceQ, intergenic region between sceQ and sceR, and start codon of sceR
actI-orf1DeletionpCRISPR-Cas9HDRS. coelicolorACT-[86]
actVBDeletionpCRISPR-Cas9HDRS. coelicolorACT-
SpCas9ACT clusterDeletionpKCCas9HDRS. coelicolorACT-[42]
actII-orf4DeletionpKCCas9HDRS. coelicolorACT-
actII-orf4 and redDDeletionpKCCas9HDRS. coelicolorACT and REDMultiplexed editing
CDA clusterDeletionpKCCas9HDRS. coelicolorCDA-
glnRDeletionpKCCas9HDRS. coelicolor--
RED clusterDeletionpKCCas9HDRS. coelicolorRED-
redDDeletionpKCCas9HDRS. coelicolorRED-
papR3DeletionpKCCas9HDRS. pristinaespiralispristinamycin-[90]
snaE1 and snaE2DeletionpKCCas9HDRS. pristinaespiralispristinamycin-
actI-orf2DeletionpWHUHDRS. coelicolorACTDevelopment of codA(sm)-based selection system for screening plasmid-cured strain[88]
rpsLPoint mutationpKCCas9HDRS. coelicolor-Lys88Glu mutation[42]
ACT, CDA, CPK, RED deleted regionReplacementpKCCas9HDRS. coelicolor M1146, M1152-ΦC31 attB integration[89]
Non-target BGCsReplacementpKCCas9HDRS. pristinaespiralisBGC2, 3, 5, 13, and 15Non-target BGC replacement with ΦC31 attB or ΦBT1 attB site
indC-like indigoidine synthaseInsertionpCRISPomycesHDR S. albusIndigoidineKasO* promoter knock-in to activate silent BGCs[87]
redDInsertionpCRISPomycesHDRS. lividansREDKasO* promoter knock-in to activate silent BGCs
actII-orf4InsertionpCRISPomycesHDRS. lividansACTKasO* promoter knock-in to activate silent BGCs
frbD operon and frbC homologInsertionpCRISPomycesHDRS. roseosporusFR-900098KasO* promoter knock-in to activate silent BGCs
main synthase geneInsertionpCRISPomycesHDRS. roseosporusBGC3 (T1pks)KasO* promoter knock-in to activate silent BGCs
luxR-type regulatorInsertionpCRISPomycesHDRS. roseosporusBGC18 (T1pks)KasO* promoter knock-in to activate silent BGCs
SSGG_RS0133915InsertionpCRISPomycesHDRS. roseosporusBGC24 (Nrps-t1pks)KasO* promoter knock-in to activate silent BGCs
rppA and cytochrome P450InsertionpCRISPomycesHDRS. venezuelaeBGC16 (T3pks)KasO* promoter knock-in to activate silent BGCs
SSQG_RS26895-RS26920 operonInsertionpCRISPomycesHDRS. viridochromogenesBGC22 (T2pks)KasO* promoter knock-in to activate silent BGCs
rkDCloning---RK-682ICE[95]
homECloning---HolomycinICE
stuE~stuF2Cloning---Tü 3010ICE[98]
stuD1, stuD2Cloning---Tü 3010ICE
SpCas9Tetarimycin BGCCloning---TetarimycinmCRISTAR[97]
spr1 region (pglE - snbC)Cloning---PristinamycinmCRISTAR[90]
5-oxomilbemycin BGCCloning---5-oxomilbemycinmCRISTAR[99]
Jadomycin and chlortetracycline BGCCloning---Jadomycin, and chlortetracyclineCATCH[96]
Chloramphenicol, YM-216391, and pristinamycin II BGCsCloning---Chloramphenicol, YM-216391, and pristinamycinCRISPR/Cas9 cleavage and Gibson assembly[89]
SpdCas9actI-orf1CRISPRipCRISPR-dCas9-S. coelicolorACT-[86]
actI-orf1CRISPRipSET-dCas9-S. coelicolorACT-[101]
actII-orf4CRISPRipSET-dCas9-S. coelicolorACT-
cdaPS1CRISPRipSET-dCas9-S. coelicolorCDA-
cpkACRISPRipSET-dCas9-S. coelicolorCPK-
redQCRISPRipSET-dCas9-S. coelicolorRED-
actI-orf1 and cdaPS1CRISPRipSET-dCas9-S. coelicolorACT, CDAMultiplexed editing
actI-orf1 and cdaPS1, cpkACRISPRipSET-dCas9-S. coelicolorACT, CDA, and CPKMultiplexed editing
actI-orf1, cdaPS1, and cpkA, redQCRISPRipSET-dCas9-S. coelicolorACT, RED, CDA, and CPKMultiplexed editing
Proteins with AmiR and NasR Transcriptional Antiterminator Regulator domain (ANTAR)CRISPRipSET-dCas9-S. coelicolor-Gene essentiality test
FnCpf1actI-orf1DisruptionpKCCpf1NHEJS. coelicolorACT-[106]
actI-orf1DisruptionpKCCpf1NHEJS. coelicolorACTReconstituted NHEJ with ligD and Ku expression
redXDisruptionpKCCpf1NHEJS. coelicolorRED-
redXDisruptionpKCCpf1NHEJS. coelicolorRED Reconstituted NHEJ with ligD and Ku expression
redX, redGDeletionpKCCpf1NHEJS. coelicolorREDDeletion by reconstituted NHEJ with ligD and Ku expression at two cleavage sites
actI-orfIDeletionpKCCpf1HDRS. coelicolorACT-
redXDeletionpKCCpf1HDRS. coelicolorRED-
actI-orf1, redXDeletionpKCCpf1HDRS. coelicolorACT and REDMultiplexed editing
SBI00792DeletionpKCCpf1HDRS. hygroscopicusAdjacent to 5-oxomilbemycin-
FnddCpf1actI-orf1CRISPRipSETddCpf1-S. coelicolorACT-
redXCRISPRipSETddCpf1-S. coelicolorRED-
cpkACRISPRipSETddCpf1-S. coelicolorCPK-
redX, actI-orf1, and cpkACRISPRipSETddCpf1-S. coelicolorRED, ACT, and CPKMultiplexed editing


Editing Streptomyces Genomes and Secondary Metabolite Biosynthetic Gene Clusters

The DSB by Cas9 occurs only if a specific sequence motif, called a protospacer adjacent motif (PAM), is present next to the protospacer [84]. All applications of the CRISPR/Cas9 system in Streptomyces strains utilize Cas9 from Streptococcus pyogenes (SpCas9), whose cognate PAM sequence is 5’-NGG [85]. As a large portion of Streptomyces genome is typically composed of G and C, SpCas9 offers plenty of potential target sites for CRISPR/Cas9-based genome engineering. Using the CRISPR/Cas9 system, genetic manipulations, including deletion, insertion, and point mutation, have been executed in various Streptomyces species [18, 42, 50, 86-94].

CRISPR/Cas9-based approaches can be divided by the type of DSB repair, NHEJ and HDR. NHEJ-mediated genome engineering generates random mutation, insertion or deletion of a few nucleotides, to disrupt a gene of interest [86, 88, 93]. Further, it has been applied to S. coelicolor and S. rimosus to disrupt actinorhodin synthesis and the pentose phosphate pathway, respectively. However, this technology was inefficient since the NHEJ system of most Streptomyces species is incomplete [86]. To enhance NHEJ-based genome engineering efficiency, LigD, the lacking component of NHEJ, was simultaneously introduced into S. coelicolor with CRISPR/Cas9 [86]. However, to avoid this limitation, CRISPR/Cas9-mediated genome engineering approaches in Streptomyces utilize HDR. For HDR-mediated genome engineering, template DNA for homologous recombination is introduced with CRISPR/Cas9 to enhance production of secondary metabolites and activate cryptic SM-BGCs in Streptomyces [87, 89, 90, 93]. For example, oxytetracycline production was increased in S. rimosus by deleting zwf2 and devB, and redirecting oxygen and NADPH to oxytetracycline biosynthesis [93]. In addition, knock-in of a strong promoter upstream of pathway-specific transcriptional activators or secondary metabolite biosynthesis genes increased secondary metabolite production and activated silent SM-BGCs [87].

In addition to the in vivo genome engineering, CRISPR/Cas9 can be utilized for cloning and refactoring of large SM-BGCs [89, 95-99]. Restriction enzymes or PCR-based cloning strategies are not suitable for manipulation of large-sized DNA fragments due to the limited restriction sites and DNA amplification errors. The high-resolution site-specific cleavage activity of the CRISPR/Cas9 system enables efficient in vitro manipulation of large SM-BGCs, up to 100 kb [96]. In addition, multiplexed refactoring of promoters in a SM-BGC has been facilitated by using a CRISPR/Cas9 system with transformation-associated recom-bination (TAR) in yeast [97]. Therefore, these in vitro SM-BGC engineering tools will provide an efficient strategy for cloning and repurposing SM-BGCs to enhance secondary metabolite production and activate silent SM-BGCs.

CRISPR/Cas9-Based Transcriptional Repression and Activation

Cas9 contains two nuclease domains, RuvC1 and HNH, which are responsible for DSB formation at the target DNA sequence [79]. Introduction of two silencing mutations to the RuvC1 and HNH nuclease domains (D10A and H840A) generates catalytically dead Cas9 (dCas9), which lacks nuclease activity yet retains DNA binding activity. By guiding dCas9 to the promoter region or coding region of the target gene, transcription initiation or transcription elongation can be blocked, respectively [100]. The CRISPR/dCas9-based transcriptional repression system, called CRISPR interference (CRISPRi), has been exploited for transcriptional repression of genes in S. coelicolor [86, 101]. Further, transcriptional repression of genes within SM-BGC by the CRISPRi resulted in decreased production of secondary metabolites [86, 101]. In addition, transcriptional repression by CRISPRi system can be utilized to screen functional genes [101]. Although a high-throughput functional gene screening system based on transposon sequencing (Tn-seq) has been implemented in Streptomyces, the resolution of Tn mutagenesis is relatively low, and thus insufficient for screening of essential Streptomyces genes [102]. The CRISPRi-based screening will enable high-throughput identification of essential genes and provide invaluable resources for the construction of genome-reduced chassis for efficient production of secondary metabolites via heterologous expression [103, 104].

Cpf1 as an Alternative to Cas9

Since SpCas9 recognizes 5’-NGG as the PAM sequence, there are considerable target sites in the GC-rich Streptomyces genome [85]. However, AT-rich regions in the Streptomyces genome are not targetable by the CRISPR/SpCas9 system, limiting precise engineering of the desired genomic locus. To overcome this limitation, another Cas enzyme from Francisella novicida, FnCpf1, has been introduced to Streptomyces as an alternative genome engineering tool. FnCpf1 recognizes 5’-TTV as the PAM sequence, and thus is suitable for targeting AT-rich regions, further elevating the potential of CRISPR/Cas-based genome engineering in Streptomyces [105]. Based on CRISPR/Cpf1, successful applications, including NHEJ, reconstituted NHEJ and HDR-mediated genome editing, have been made in Streptomyces [106]. In addition to genome editing, transcriptional repression based on DNase-deactivated Cpf1 (ddCpf1) has been applied in Streptomyces, successfully covering all applications of CRISPR/Cas9 for Streptomyces [106]. Cpf1 not only expands the potential targets for CRISPR/Cas mediated genome engineering, but it also broadens the range of CRISPR/Cas applicable organisms with less cellular toxicity compared to Cas9 [106]. However, in vitro applications of the CRISPR/Cpf1 system have not been described for SM-BGC cloning in Streptomyces. Unlike Cas9, Cpf1 generates a DSB with sticky ends, which may enable efficient directional cloning with simple ligation [105].

Heterologous Expression of Secondary Metabolite Biosynthetic Gene Clusters

Compared to secondary metabolite production in the native host, heterologous expression has several advantages, including: (1) it enables SM-BGCs expression of unculturable or slow-growing native host strains, (2) it overcomes the difficult genetic manipulation of the native host, and (3) it bypasses the innate regulatory network of the native host [107]. In fact, the Streptomyces species is the most suitable host for heterologous expression of SM-BGCs, as compared to other organisms such as E. coli, bacillus, or yeast, because of its (1) abundant precursors, cofactors, and enzymes for secondary metabolite biosynthesis; (2) sophisticated post-modification system for secondary metabolites such as phosphorylation, acetylation, farnesylation, and glycosylation;(3) broad antibiotic resistance and tolerance; (4) proper protein folding for the functionality of multi-enzyme complexes; and (5) other cellular environments, including pH and redox potential [108]. Over the past few decades, approximately 100 SM-BGCs have been heterologously expressed in Streptomyces, particularly in S. coelicolor, S. lividans, S. avermitilis, and S. albus [109]. Although heterologous expression has been successfully used for secondary metabolite production, several limitations still remain. First, the large size of SM-BGC hampers the efficiency of genetic manipulation [110]. Further, even if the transfer of SM-BGCs to the heterologous expression host was successful, their expression may be insignificant due to differences in the precursor pool from that of the native host and metabolic competitions between the target SM-BGC and other endogenous SM-BGCs in the expression host [108]. In this subsection, we discuss cloning strategies for large-size SM-BGCs and optimization of the heterologous expression host.

There are four steps to express the SM-BGC in the heterologous expression host, which include (1) acquisition of the target SM-BGC from the native host genome, (2) ligation or assembly of the SM-BGC to the vector, (3) transfer of the SM-BGC-encoded vector to the heterologous expression host, and (4) target of secondary metabolite production (Table 3).

Table 3 . Different strategy for BGC cloning..

BGC cloning stepsStrategiesRepresentative examplesRef
Acquisition of the target BGC from the native host genomeGenomic libraryCosmid, fosmid, BAC, and PAC[135]
Cut off both ends of target BGCRestriction: pSBAC[113]
Integrase: IR[114]
CRISPR: CATCH, mCRISTAR, and CRISPR-TAR[96, 97, 120]
PCR amplificationDNA assembler[116]
Ligation or assembly of the target BGC to the vectorIn vitroSticky end ligation: pSBAC[113]
Blunt end ligation: ICE[95]
Gibson assembly: CATCH and MSGE[89, 96]
In vivoRecombination in native host: IR[114]
Recombination in E. coli: LLHR[117]
Recombination in yeast: TAR, DNA assembler, DiPac, and mCRISTAR[97, 115, 116, 118]
Transferring BGC vector to the expression hostConjugationpUWLcre[136]
Protoplast transformationpSKC2 and pOJ446[137]
Target secondary metabolite production by expression of the BGC vectorIntegrativepSET152, pCAP01, and pESAC[118, 138]
ReplicativepSKC2 and pUWL201[139]


The most frequently used method for acquisition of the target SM-BGC is the genomic library construction using cosmid, fosmid, BAC, and PAC vectors [109]. This method can be applied to a broad range of Streptomyces genomes, in which the full sequence is unknown. It is particularly effective to discover novel secondary metabolites from the metagenome, including unculturable bacteria [111]. The second method is to cut both ends of the target SM-BGC from the genomic DNA. However, restriction sites are not generally available at both ends of target SM-BGCs. Thus, unique restriction sites can be introduced using a suicide plasmid, which contains a homologous sequence of one of the end sites, and integrated into both ends of target SM-BGCs by single crossover. Streptomyces bacterial artificial chromosome system (pSBAC) is a successful example for some SM-BGCs, such as tautomycetin (80 kb) and pikromycin (60 kb) [112, 113]. Another strategy is the integrase-mediated recombination (IR) system that introduces integration sites, such as attB6 and attB9, at both ends by single crossover homologous recombination of the vector, such as pKC1139 [114]. Φ-integrase-mediated excision of the target SM-BGC is subsequently performed to obtain the SM-BGC vector in vivo. Recently, in vitro site-specific digestion of genomic DNA using the CRISPR/Cas9 system was used as an alternative strategy for the acquisition of target SM-BGCs [94-97, 115]. The third method for acquisition of target SM-BGCs is PCR amplification [116], in which each fragment has homologous arms at both ends added by PCR primers to self-assemble or assemble together with genetic parts such as promoters and RBSs. As all three SM-BGC acquisition methods have their own advantages and limitations, they should be applied according to specific situations.

Ligation or assembly methods of target SM-BGC to the heterologous expression vector can be largely divided into two groups, in vitro and in vivo. In vitro cloning of the target SM-BGCs involves (1) ligation of cognate sticky ends formed by restriction enzyme digestion or two CRISPR/Cas9 digestion using T4 DNA ligase [95, 109] and (2) Gibson assembly using 5’ exonuclease [89, 96]. Ligation of two fragments by DNA ligase is simple and efficient, but preparation of cognate ends for ligation is required. Conversely, the Gibson assembly method is theoretically universal for any fragments by introduction of homologous sequence at their ends through PCR amplification. In vivo cloning of SM-BGCs exploits the homologous recombination system in the native host [114], E. coli [117], and yeast [118]. For example, integrase-mediated recombination (IR) directly obtains the SM-BGC-containing vector from the genomic DNA of the native host [114]. Linear-linear homologous recombination (LLHR) using λ-Red system or RecET system in E. coli is one of the most widely used in vivo recombination systems with high cloning efficiency, but their reported size limit is about 50 kb [107, 109, 117]. Unlike bacteria, yeast have their own efficient homologous recombination system, such that more than two large fragments can be assembled by transformation of all fragments simultaneously into yeast, which is named as transformation-associated recombination (TAR) [119]. The TAR cloning method has shown relatively higher efficiency, size capacity, and number of fragments in many strategies such as CRISPR-TAR, mCRISTAR, and DNA assembler, compared to other in vivo cloning methods [97, 116, 120]. Thus, TAR and in vitro Gibson assembly are considered as the most high-throughput and efficient strategies for the preparation of Streptomyces SM-BGCs vectors.

In most cases, constructed SM-BGC vectors have been transformed to the heterologous expression host through conjugation between E. coli and Streptomyces strains [109]. Otherwise, the vector can be directly transformed to the heterologous expression host by the protoplast method [32], which depends on the vector components such as oriT (essential for conjugation) and the Streptomyces species. After transfer of the SM-BGC vector to the expression host, the SM-BGC vector can be integrated into the host genome or remain as a replicative plasmid. Most heterologous expression vectors in previous studies were integrative vectors, which are more stable after serial generations [109]. Genetic stability is very important for the further fermentation process of secondary metabolites [121]. However, the copy number of the integrative SM-BGC vector is only one compared to multi-copy replicative vectors. Therefore, multi-copy integration of the SM-BGC vector into genome or promoter refactoring of SM-BGC genes should be additionally performed to increase productivity [89]. Thus far, many SM-BGC cloning strategies for heterologous expression have been developed. However, cloning of a large size and number of SM-BGC fragments is a common limitation. Therefore, a high-throughput, stepwise, systematic strategy for efficient cloning of SM-BGCs for heterologous expression should be considered.

Streptomyces Chassis Strains for Heterologous Gene Expression

To improve yields of secondary metabolites, Streptomyces hosts were genetically modified by removing endogenous SM-BGCs, nonessential genes, and genomic regions and engineering genes with pleiotropic functions. Representative examples of optimized Streptomyces as heterologous expression hosts are shown in Table 4. Removing SM-BGCs resulted in a “reduced Streptomyces genome,” which can conserve energy and other building blocks in addition to the specific precursor pool. In other words, the nucleotide and energy for replication of the reduced genome will be decreased, and this redundant energy can be used for the target metabolite production. Indeed, several engineered strains of S. coelicolor (4 SM-BGCs deletion), S. lividans (3 SM-BGCs deletion), and S. albus (15 SM-BGCs deletion) have shown improved target secondary metabolite production and reduced background chemical profiles [104, 122-124].

Table 4 . Representative examples of Streptomyces chassis strain for optimal heterologous expression..

Heterologous hostEngineeringTarget genes or regionsDeletion methodExpressed BGCBGC vectorEffectLimitationRef
Streptomyces coelicolor M145BGC deletion and Pleiotropic gene engineeringDeletion of four BGCs (ACT, RED, CPK, and CDA) Point mutations of rpoB and rpsL.Homologous recombination by double crossover of the plasmidShlorampheniocol and congocidineCosmidImproved production, clean profile of background metabolitesLow fitness[104]
Streptomyces sp. FR-008BGC deletionDeletion of three BGCs (candicidin, type III PKS, and type I PKS)Homologous recombination by double crossover of the plasmidNoneNoneImproved fitness, sporulation, and clean profile of background metabolitesHeterologous expression was not tested[124]
Streptomyces lividans TK24BGC deletionDeletion of three BGCs (ACT, RED, and CDA) One copy integration of AfsRS by attB integraseHomologous recombination by double crossover of the plasmidStreptothiricins, borrelidin, and linear lipopeptidesBACHigh-throughput functional genome mining of Streptomyces rocheiLow fitness, laborious screening of BAC libraries[123]
Streptomyces lividans TK24BGC deletionDeletion of three BGCs (ACT, RED, and CDA) Additional copies integration of AfsRS by attB integraseHomologous recombination by double crossover of the plasmidHybrubinsBACHigh-throughput functional genome mining of Streptomyces variabilis Pathway crosstalk between incompletely deleted RED cluster.Low fitness[140]
Streptomyces albus J1074BGC deletionDeletion of fifteen BGCs (Frontalamide, Paulomycin, Geosmin, Lantibiotic, carotenoid, flaviolin, candicidin, antimycin, 2 PKSNRPS, and 4 NRPS)Homologous recombination by double crossover of the plasmid using λ-red systemTunicamycin B2, moenomycin M, griseorhodin A, pyridinopyrone A, bhimamycin A, didesmethylmensacarcin, didemethoxyaranciamycino ne, aloesaponarin II, and cinnamycin, fralnimycinFosmid and BACImproved production, clean profile of background metabolitesMoenomycin M productivity was reduced.[122]
Streptomyces avermitilisNonessential region deletion and BGC deletionDeletion of 1.48 Mb left arm determined by comparative genomicsHomologous recombination by double crossover of the plasmid using λ-red system Cre/loxP systemStreptomycin, cephamycin C, and pladienolideCosmidand BACImproved production by additional introduction of regulatory gene and optimization of codon usageLow conjugation efficiency[103]
Streptomyces avermitilisNonessential region deletionDeletion of 1.48 Mb left arm and some regions determined by comparative genomicsHomologous recombination by double crossover of the plasmid using λ-red system Cre/loxP systemStreptomycin, ribostamycin, kasugamycin, pholipomycin, oxytetracycline, resistomycin, pladienolide B, erythromycin A, bafilimycin B1, nemadectin α, aureothin, leptomycin, cephamycin C, holomycin, lactacystin, clavulanic acid, rebeccamycin, novobiocin, chloramphenicol, 2-methylisoborneol, pentalenolactone, amorpha-1,4-diene, taxa-4,11-diene, levopimaradiene, and abietatrieneCosmid and BACImproved production, fitness, clean profile of background metabolites. Broad precursor capacity (sugar, polyketide, peptide, shikimate, and MVA or MEP)Ribostamycin, oxytetracycline productivity were reduced[125]
Streptomyces chattanoogensis L10Nonessential region deletionDeletion of 1.3 Mb and 0.7 Mb nonessential arms determined by comparative genomics and prediction toolsCre/loxP recombinationACTpMM1Improved production, fitness, ATP, NADPH, transformation efficiency, and genetic stability. Dispersed morphology.1.3 Mb deleted strain was detrimental due to deletion of some unknown genes[121]
Streptomyces albus J1074Pleiotropic gene engineering and BGC deletionDeletion of pfk, wblA, overexpression of cpk, and deletion of one BGC (paulomycin)Homologous recombination by double crossover of the plasmid using λ-red systemACTFosmidImproved production, fitness, and NADPH.Undesirable effects might be incurred due to the global change of transcriptome[128]


Nonessential genomic regions (NGR) are usually located at the ends of linear chromosomes, which are not conserved in all species and dispensable for cell growth. They include genomic islands (GI), IS elements, and endogenous CRISPR array regions that decrease genomic stability. For example, NGRs in S. avermitilis (1.48 Mb) and S. chattanoogenisis (0.7 Mb) were selected, based on comparative genomics of Streptomyces genomes, and deleted by λ-Red system or Cre/loxP recombination system [121, 125]. As expected, deletion of NGRs increased the fitness level of the engineered strain relative to the wild-type strain with beneficial effects on morphology, ATP level, NADPH level, transformation efficiency, and genetic stability in S. chattanoogenisis [121]. However, large deletion of NGRs may cause undesired deleterious effects on cell growth due to the unknown essential function of genes and synthetic lethality of more than two abundant essential genes. Therefore, the systems level of functional genomic studies should be followed to determine the nonessential regions more precisely. Integration of these functional studies with multi-omics data and experimental validations might allow construction of a highly efficient Streptomyces chassis strain for heterologous expression of SM-BGCs [126, 127].

Genes with pleiotropic functions can be additionally engineered to improve productivity of target secondary metabolites. For instance, deletion of phosphofructokinase gene pfk and global transcriptional regulator gene wblA and overexpression of global transcriptional regulator gene crp in S. albus increased secondary metabolite production, redox potential, and fitness by changing the global transcriptional status [128]. Further, point mutations of RNA polymerase gene rpoB and ribosomal protein gene rpsL in S. coelicolor showed increased fitness and secondary metabolite production by altering regulation at both the transcriptional and translational levels [104]. As pleiotropic functions usually include undesired phenotypes for secondary metabolite production, the engineering target should be selected carefully through rational design based on systematic information.

Future Perspective

In this review, we summarized a synthetic biology strategy to produce novel secondary metabolites in Streptomyces. Accumulation of genetic information and SM-BGCs aided by recent advances in Next-Generation Sequencing (NGS) has revealed the enormous potential of Streptomyces as a reservoir for novel bioactive compounds and is far outpacing our capacity to explore SM-BGCs and their products [129]. To fully harness Streptomyces’ ability to produce valuable secondary metabolites, rational design and efficient synthetic biology tools for Streptomyces are essential. To date, however, in silico SM-BGC prediction tools, such as antiSMASH, still need to be optimized for precise mining capability. Further, synthetic biology tools for Streptomyces are limited to fulfill the precise designs for novel secondary metabolite production. To this end, integration of massive omics data and vigorous functional studies can elevate the fidelity of SM-BGC mining and increase genetic parts for Streptomyces engineering. Specifically, construction of genetic part libraries based on transcriptome and translatome data by screening using a high-throughput reporter system will deliver a universal set of genetic parts for Streptomyces.

Although genetic tools based on the CRISPR/Cas system have offered diverse strategies to enhance secondary metabolite production and activate silent SM-BGCs [87, 89, 90, 93, 99], further optimization of the CRISPR/Cas system for Streptomyces is still required. An example is that the toxicity of Cas nucleases is disadvantageous for multiplexed applications and CRISPR/Cas bearing systems. In addition, expansion of PAM recognition is required to enable efficient genome engineering. These limitations can be overcome by modulating Cas expression and exploiting Cas variants [106, 130, 131]. Particularly, CRISPR activation (CRISPRa) has not been applied to Streptomyces [132]. The CRISPRa approach can serve as an efficient strategy to investigate silent SM-BGCs without labor-intensive genome editing efforts. Multiplexing the precise transcriptional regulation through integration of both CRISPRa and CRISPRi is expected to reconstruct the metabolic network to enhance the precursor supply, reducing flux toward competing pathways or unwanted by-products, bypassing gene expression regulation, and expressing the SM-BGCs. Recently, toehold-gated gRNA was developed, which links endogenous signals to activation of the CRISPR/Cas system [133]. As the production of secondary metabolites requires sufficient accumulation of precursors and activation of biosynthetic genes, the combination of toehold-gated gRNA and CRISPRa and CRISPRi strategies will suggest a new metabolic engineering approach, linking the production of precursors to production of secondary metabolites.

Development of synthetic biology tools can also be exploited to construct the Streptomyces chassis for heterologous expression of novel SM-BGCs. So far, S. albus J1074 with deletion of 15 BGCs and S. chattanoogensis L10 with deletion of 0.7 Mb of a nonessential arm seem to be the best Streptomyces chassis for heterologous expression [121, 122]. A further challenge might be the construction of the “superhost Streptomyces chassis” by removing all endogenous SM-BGCs, nonessential genes, and genomic regions and adding all precursor synthetic genes. By heterologous expression of the target SM-BGC in this superhost chassis, productivity of target secondary metabolites will be further improved. In addition, novel secondary metabolites will be discovered from a wide array of silent SM-BGCs in a high-throughput manner. However, construction of a superhost for all secondary metabolites might not be feasible due to precursor differences in Streptomyces species. Indeed, all five types of secondary metabolites were produced in the S. albus J1074 strain; however, some metabolites were not produced significantly [122]. Therefore, construction of several “specialized Streptomyces chassis hosts” for each type of secondary metabolite might be a better choice [108]. This “design-build-test (DBT)” cycle, which is a rational design based on in silico SM-BGC mining, the build of a SM-BGC expression in Streptomyces chassis, and the high-throughput test of secondary metabolite production will be iterated to learn and optimize production of novel secondary metabolites (Fig. 1). This synthetic biology strategy will ultimately expand the productivity and diversity of available novel secondary metabolites as potential biopharmaceuticals.

Acknowledgments

This work was supported by a grant from the Novo Nordisk Foundation (grant number NNF10CC1016517). This research was also supported by the Basic Science Research Program (2018R1A1A3A04079196 to S.C.), the Basic Core Technology Development Program for the Oceans and the Polar Regions (2016M1A5A1027458 to B.-K.C.), and the Bio & Medical Technology Development Program (2018M3A9F3079664 to B.-K.C.) through the National Research Foundation (NRF) funded by the Ministry of Science and ICT.

Conflict of Interest


The authors have no financial conflicts of interest to declare.

Fig 1.

Figure 1.Overview of synthetic biology strategy to produce novel secondary metabolite from Streptomyces. Abbreviations: BGC, biosynthetic gene cluster; CDS, coding sequence; 5’ UTR, 5’ untranslated region; RBS, ribosome binding site; WT, wild type; LA, left homology arm; RA, right homology arm.
Journal of Microbiology and Biotechnology 2019; 29: 667-686https://doi.org/10.4014/jmb.1904.04015

Table 1 . Genetic parts for Streptomyces..

Genetic partsFeatureReference
Constitutive promoters
ermE* promoterMutation at the promoter of the erythromycin resistance gene of Streptomyces erythraeus[31]
SF14P promoterGenome of Streptomyces ghanaensis phage I19[34]
kasOP promoterPromoter of SARP family regulator in Stretpomyces coelicolor A3[33]
gapdh promoterPromoter of glyceraldehyde-3-phosphate dehydrogenase in Streptomyces griseus[35]
rpsL promoterPromoter of 30S ribosomal protein S12 in Streptomyces griseus[35]
195 native or synthetic promotersHigh-throughput screening in S. venezueale[58]
32 native promotersTranscriptome data-based selection in S. albus[38]
166 native promotersTranscriptome data-based selection in S. coelicolor[39]
2 native promotersMulti-omics data-based selection in S. coelicolor[59]
Inducible promoters
tipA promoterThiostrepton-induced promoter[40]
nitA promoterε-caprolactam-induced promoter[45]
xylA promoterXylose-induced promoter[46]
tcp830Tetracycline-induced promoter[43]
PA3-rolOResorcinol-induced promoter[44]
P21-cmtCumate-induced promoter[44]
Terminators
FdBidirectional transcription termination originated from E. coli phage fd[48]
TD1Bidirectional transcription termination originated from Bacillus subtilis phage Φ29[49]
RBS
AAAGGAGGTypical RBS sequence of S. coelicolor[134]
192 native or synthetic RBSsHigh-throughput screening in S. venezueale[58]
4 native RBSsMulti-omics data-based selection in S. coelicolor[59]
Reporter genes
luxAB cassetten-Decanal as substrate; absorbance at 490 nm wavelength[61]
amy geneSoluble starch with 3,5-dinitrosalycilic acid (DNS) as substrate; absorbance at 540 nm wavelength[64]
xylE geneCatecol as substrate; absorbance at 375 nm wavelength[62]
gusA genep-Nitrophenyl-β-D-glucuronide as substrate; absorbance at 415 nm wavelength[63]
eGFPGreen fluorescent protein; excitation wavelength 470-490 nm and emission wavelength 515 nm[65]
sfGFPGreen fluorescent protein; excitation wavelength 488 nm and emission wavelength 500~550 nm[58]
mRFPRed fluorescent protein; excitation wavelength 584 nm and emission wavelength 607 nm[67]
mCherryRed fluorescent protein; excitation wavelength 587 nm and emission wavelength 610 nm[68]

Table 2 . Application of CRISPR/Cas9-mediated engineering in Streptomyces..

CasTargetStrategyVectorRepairOrganismRelated secondary metaboliteRemarkRef
SpCas9actI-orf1DisruptionpCRISPR-Cas9NHEJS. coelicolorACTReconstituted NHEJ with ligD expression[86]
actVBDisruptionpCRISPR-Cas9NHEJS. coelicolorACTReconstituted NHEJ with ligD expression
actI-orf2DisruptionpWHUNHEJS. coelicolorACTcodA(sm)-based screening system for plasmid-cured strain[88]
zwf2DisruptionpCRISPomycesNHEJS. rimosus-Oxytetracycline production enhancement by disruption of competitive gene[93]
devBDisruptionpCRISPomycesNHEJS. rimosus-Oxytetracycline production enhancement by disruption of competitive gene
sshg_00040 - sshg_00050DeletionpCRISPomycesHDR S. albusLanthipeptide-[50]
sshg_05713DeletionpCRISPomycesHDR S. albusPolycylic tetramic acid macrolactam-
Formicamycin clusterDeletionpCRISPomycesHDRS. formicaeFormicamycin-[91]
forVDeletionpCRISPomycesHDRS. formicaeFormicamycin-
actVA-orf5DeletionpCRISPomycesHDRS. lividansACT-[50]
redD-redFDeletionpCRISPomycesHDRS. lividansRED-
redNDeletionpCRISPomycesHDRS. lividansRED-
actVA-orf5 and redNDeletionpCRISPomycesHDRS. lividansACT and REDMultiplexed editing
devBDeletionpCRISPomycesHDRS. rimosus-Oxytetracycline production enhancement by disruption of competitive gene[93]
zwf2DeletionpCRISPomycesHDRS. rimosus-Oxytetracycline production enhancement by disruption of competitive gene
phpDDeletionpCRISPomycesHDRS. viridochromogenesPhosphinothricin tripeptide-[50]
phpMDeletionpCRISPomycesHDRS. viridochromogenesPhosphinothricin tripeptide-
sceNDeletionpCRISPR-Cas9HDRStreptomyces sp.SD85BGC11 (sceliphrolactam)-[18]
sceQ-sceR fusionDeletionpCRISPR-Cas9HDRStreptomyces sp.SD85BGC11 (sceliphrolactam)Fusion of sceQ and sce R by deleting stop codon of sceQ, intergenic region between sceQ and sceR, and start codon of sceR
actI-orf1DeletionpCRISPR-Cas9HDRS. coelicolorACT-[86]
actVBDeletionpCRISPR-Cas9HDRS. coelicolorACT-
SpCas9ACT clusterDeletionpKCCas9HDRS. coelicolorACT-[42]
actII-orf4DeletionpKCCas9HDRS. coelicolorACT-
actII-orf4 and redDDeletionpKCCas9HDRS. coelicolorACT and REDMultiplexed editing
CDA clusterDeletionpKCCas9HDRS. coelicolorCDA-
glnRDeletionpKCCas9HDRS. coelicolor--
RED clusterDeletionpKCCas9HDRS. coelicolorRED-
redDDeletionpKCCas9HDRS. coelicolorRED-
papR3DeletionpKCCas9HDRS. pristinaespiralispristinamycin-[90]
snaE1 and snaE2DeletionpKCCas9HDRS. pristinaespiralispristinamycin-
actI-orf2DeletionpWHUHDRS. coelicolorACTDevelopment of codA(sm)-based selection system for screening plasmid-cured strain[88]
rpsLPoint mutationpKCCas9HDRS. coelicolor-Lys88Glu mutation[42]
ACT, CDA, CPK, RED deleted regionReplacementpKCCas9HDRS. coelicolor M1146, M1152-ΦC31 attB integration[89]
Non-target BGCsReplacementpKCCas9HDRS. pristinaespiralisBGC2, 3, 5, 13, and 15Non-target BGC replacement with ΦC31 attB or ΦBT1 attB site
indC-like indigoidine synthaseInsertionpCRISPomycesHDR S. albusIndigoidineKasO* promoter knock-in to activate silent BGCs[87]
redDInsertionpCRISPomycesHDRS. lividansREDKasO* promoter knock-in to activate silent BGCs
actII-orf4InsertionpCRISPomycesHDRS. lividansACTKasO* promoter knock-in to activate silent BGCs
frbD operon and frbC homologInsertionpCRISPomycesHDRS. roseosporusFR-900098KasO* promoter knock-in to activate silent BGCs
main synthase geneInsertionpCRISPomycesHDRS. roseosporusBGC3 (T1pks)KasO* promoter knock-in to activate silent BGCs
luxR-type regulatorInsertionpCRISPomycesHDRS. roseosporusBGC18 (T1pks)KasO* promoter knock-in to activate silent BGCs
SSGG_RS0133915InsertionpCRISPomycesHDRS. roseosporusBGC24 (Nrps-t1pks)KasO* promoter knock-in to activate silent BGCs
rppA and cytochrome P450InsertionpCRISPomycesHDRS. venezuelaeBGC16 (T3pks)KasO* promoter knock-in to activate silent BGCs
SSQG_RS26895-RS26920 operonInsertionpCRISPomycesHDRS. viridochromogenesBGC22 (T2pks)KasO* promoter knock-in to activate silent BGCs
rkDCloning---RK-682ICE[95]
homECloning---HolomycinICE
stuE~stuF2Cloning---Tü 3010ICE[98]
stuD1, stuD2Cloning---Tü 3010ICE
SpCas9Tetarimycin BGCCloning---TetarimycinmCRISTAR[97]
spr1 region (pglE - snbC)Cloning---PristinamycinmCRISTAR[90]
5-oxomilbemycin BGCCloning---5-oxomilbemycinmCRISTAR[99]
Jadomycin and chlortetracycline BGCCloning---Jadomycin, and chlortetracyclineCATCH[96]
Chloramphenicol, YM-216391, and pristinamycin II BGCsCloning---Chloramphenicol, YM-216391, and pristinamycinCRISPR/Cas9 cleavage and Gibson assembly[89]
SpdCas9actI-orf1CRISPRipCRISPR-dCas9-S. coelicolorACT-[86]
actI-orf1CRISPRipSET-dCas9-S. coelicolorACT-[101]
actII-orf4CRISPRipSET-dCas9-S. coelicolorACT-
cdaPS1CRISPRipSET-dCas9-S. coelicolorCDA-
cpkACRISPRipSET-dCas9-S. coelicolorCPK-
redQCRISPRipSET-dCas9-S. coelicolorRED-
actI-orf1 and cdaPS1CRISPRipSET-dCas9-S. coelicolorACT, CDAMultiplexed editing
actI-orf1 and cdaPS1, cpkACRISPRipSET-dCas9-S. coelicolorACT, CDA, and CPKMultiplexed editing
actI-orf1, cdaPS1, and cpkA, redQCRISPRipSET-dCas9-S. coelicolorACT, RED, CDA, and CPKMultiplexed editing
Proteins with AmiR and NasR Transcriptional Antiterminator Regulator domain (ANTAR)CRISPRipSET-dCas9-S. coelicolor-Gene essentiality test
FnCpf1actI-orf1DisruptionpKCCpf1NHEJS. coelicolorACT-[106]
actI-orf1DisruptionpKCCpf1NHEJS. coelicolorACTReconstituted NHEJ with ligD and Ku expression
redXDisruptionpKCCpf1NHEJS. coelicolorRED-
redXDisruptionpKCCpf1NHEJS. coelicolorRED Reconstituted NHEJ with ligD and Ku expression
redX, redGDeletionpKCCpf1NHEJS. coelicolorREDDeletion by reconstituted NHEJ with ligD and Ku expression at two cleavage sites
actI-orfIDeletionpKCCpf1HDRS. coelicolorACT-
redXDeletionpKCCpf1HDRS. coelicolorRED-
actI-orf1, redXDeletionpKCCpf1HDRS. coelicolorACT and REDMultiplexed editing
SBI00792DeletionpKCCpf1HDRS. hygroscopicusAdjacent to 5-oxomilbemycin-
FnddCpf1actI-orf1CRISPRipSETddCpf1-S. coelicolorACT-
redXCRISPRipSETddCpf1-S. coelicolorRED-
cpkACRISPRipSETddCpf1-S. coelicolorCPK-
redX, actI-orf1, and cpkACRISPRipSETddCpf1-S. coelicolorRED, ACT, and CPKMultiplexed editing

Table 3 . Different strategy for BGC cloning..

BGC cloning stepsStrategiesRepresentative examplesRef
Acquisition of the target BGC from the native host genomeGenomic libraryCosmid, fosmid, BAC, and PAC[135]
Cut off both ends of target BGCRestriction: pSBAC[113]
Integrase: IR[114]
CRISPR: CATCH, mCRISTAR, and CRISPR-TAR[96, 97, 120]
PCR amplificationDNA assembler[116]
Ligation or assembly of the target BGC to the vectorIn vitroSticky end ligation: pSBAC[113]
Blunt end ligation: ICE[95]
Gibson assembly: CATCH and MSGE[89, 96]
In vivoRecombination in native host: IR[114]
Recombination in E. coli: LLHR[117]
Recombination in yeast: TAR, DNA assembler, DiPac, and mCRISTAR[97, 115, 116, 118]
Transferring BGC vector to the expression hostConjugationpUWLcre[136]
Protoplast transformationpSKC2 and pOJ446[137]
Target secondary metabolite production by expression of the BGC vectorIntegrativepSET152, pCAP01, and pESAC[118, 138]
ReplicativepSKC2 and pUWL201[139]

Table 4 . Representative examples of Streptomyces chassis strain for optimal heterologous expression..

Heterologous hostEngineeringTarget genes or regionsDeletion methodExpressed BGCBGC vectorEffectLimitationRef
Streptomyces coelicolor M145BGC deletion and Pleiotropic gene engineeringDeletion of four BGCs (ACT, RED, CPK, and CDA) Point mutations of rpoB and rpsL.Homologous recombination by double crossover of the plasmidShlorampheniocol and congocidineCosmidImproved production, clean profile of background metabolitesLow fitness[104]
Streptomyces sp. FR-008BGC deletionDeletion of three BGCs (candicidin, type III PKS, and type I PKS)Homologous recombination by double crossover of the plasmidNoneNoneImproved fitness, sporulation, and clean profile of background metabolitesHeterologous expression was not tested[124]
Streptomyces lividans TK24BGC deletionDeletion of three BGCs (ACT, RED, and CDA) One copy integration of AfsRS by attB integraseHomologous recombination by double crossover of the plasmidStreptothiricins, borrelidin, and linear lipopeptidesBACHigh-throughput functional genome mining of Streptomyces rocheiLow fitness, laborious screening of BAC libraries[123]
Streptomyces lividans TK24BGC deletionDeletion of three BGCs (ACT, RED, and CDA) Additional copies integration of AfsRS by attB integraseHomologous recombination by double crossover of the plasmidHybrubinsBACHigh-throughput functional genome mining of Streptomyces variabilis Pathway crosstalk between incompletely deleted RED cluster.Low fitness[140]
Streptomyces albus J1074BGC deletionDeletion of fifteen BGCs (Frontalamide, Paulomycin, Geosmin, Lantibiotic, carotenoid, flaviolin, candicidin, antimycin, 2 PKSNRPS, and 4 NRPS)Homologous recombination by double crossover of the plasmid using λ-red systemTunicamycin B2, moenomycin M, griseorhodin A, pyridinopyrone A, bhimamycin A, didesmethylmensacarcin, didemethoxyaranciamycino ne, aloesaponarin II, and cinnamycin, fralnimycinFosmid and BACImproved production, clean profile of background metabolitesMoenomycin M productivity was reduced.[122]
Streptomyces avermitilisNonessential region deletion and BGC deletionDeletion of 1.48 Mb left arm determined by comparative genomicsHomologous recombination by double crossover of the plasmid using λ-red system Cre/loxP systemStreptomycin, cephamycin C, and pladienolideCosmidand BACImproved production by additional introduction of regulatory gene and optimization of codon usageLow conjugation efficiency[103]
Streptomyces avermitilisNonessential region deletionDeletion of 1.48 Mb left arm and some regions determined by comparative genomicsHomologous recombination by double crossover of the plasmid using λ-red system Cre/loxP systemStreptomycin, ribostamycin, kasugamycin, pholipomycin, oxytetracycline, resistomycin, pladienolide B, erythromycin A, bafilimycin B1, nemadectin α, aureothin, leptomycin, cephamycin C, holomycin, lactacystin, clavulanic acid, rebeccamycin, novobiocin, chloramphenicol, 2-methylisoborneol, pentalenolactone, amorpha-1,4-diene, taxa-4,11-diene, levopimaradiene, and abietatrieneCosmid and BACImproved production, fitness, clean profile of background metabolites. Broad precursor capacity (sugar, polyketide, peptide, shikimate, and MVA or MEP)Ribostamycin, oxytetracycline productivity were reduced[125]
Streptomyces chattanoogensis L10Nonessential region deletionDeletion of 1.3 Mb and 0.7 Mb nonessential arms determined by comparative genomics and prediction toolsCre/loxP recombinationACTpMM1Improved production, fitness, ATP, NADPH, transformation efficiency, and genetic stability. Dispersed morphology.1.3 Mb deleted strain was detrimental due to deletion of some unknown genes[121]
Streptomyces albus J1074Pleiotropic gene engineering and BGC deletionDeletion of pfk, wblA, overexpression of cpk, and deletion of one BGC (paulomycin)Homologous recombination by double crossover of the plasmid using λ-red systemACTFosmidImproved production, fitness, and NADPH.Undesirable effects might be incurred due to the global change of transcriptome[128]

References

  1. Ohnishi Y, Ishikawa J, Hara H, Suzuki H, Ikenoya M, Ikeda H, et al. 2008. Genome sequence of the streptomycin-producing microorganism Streptomyces griseus IFO 13350. J. Bacteriol. 190: 4050-4060.
    Pubmed KoreaMed CrossRef
  2. Khan ST, Komaki H, Motohashi K, Kozone I, Mukai A, Takagi M, et al. 2011. Streptomyces associated with a marine sponge Haliclona sp.; biosynthetic genes for secondary metabolites and products. Environ. Microbiol. 13: 391-403.
    Pubmed CrossRef
  3. Onaka H. 2017. Novel antibiotic screening methods to awaken silent or cryptic secondary metabolic pathways in actinomycetes. J. Antibiot. (Tokyo) 70: 865-870.
    Pubmed CrossRef
  4. Lim FY, Sanchez JF, Wang CC, Keller NP. 2012. Toward awakening cryptic secondary metabolite gene clusters in filamentous fungi. Methods Enzymol. 517: 303-324.
    Pubmed KoreaMed CrossRef
  5. Medema MH, Alam MT, Breitling R, Takano E. 2011. The future of industrial antibiotic production: from random mutagenesis to synthetic biology. Bioeng. Bugs. 2: 230-233.
    Pubmed CrossRef
  6. Nguyen QT, Merlo ME, Medema MH, Jankevics A, Breitling R, Takano E. 2012. Metabolomics methods for the synthetic biology of secondary metabolism. FEBS Lett. 586: 2177-2183.
    Pubmed CrossRef
  7. Yang YH, Song E, Lee BR, Kim EJ, Park SH, Kim YG, et al. 2010. Rapid functional screening of Streptomyces coelicolor regulators by use of a pH indicator and application to the MarR-like regulator AbsC. Appl. Environ. Microbiol. 76: 3645-3656.
    Pubmed KoreaMed CrossRef
  8. Myronovskyi M, Luzhetskyy A. 2016. Native and engineered promoters in natural product discovery. Nat. Prod. Rep. 33: 1006-1019.
    Pubmed CrossRef
  9. Ziemert N, Alanjary M, Weber T. 2016. The evolution of genome mining in microbes -a review. Nat. Prod. Rep. 33: 988-1005.
    Pubmed CrossRef
  10. Harrison J, Studholme DJ. 2014. Recently published Streptomyces genome sequences. Microb. Biotechnol. 7: 373-380.
    Pubmed KoreaMed CrossRef
  11. Bentley SD, Chater KF, Cerdeno-Tarraga AM, Challis GL, Thomson NR, James KD, et al. 2002. Complete genome sequence of the model actinomycete Streptomyces coelicolor A3(2). Nature 417: 141-147.
    Pubmed CrossRef
  12. Komaki H, Sakurai K, Hosoyama A, Kimura A, Igarashi Y, Tamura T. 2018. Diversity of nonribosomal peptide synthetase and polyketide synthase gene clusters among taxonomically close Streptomyces strains. Sci. Rep. 8: 6888.
    Pubmed KoreaMed CrossRef
  13. Starcevic A, Zucko J, Simunkovic J, Long PF, Cullum J, Hranueli D. 2008. ClustScan: an integrated program package for the semi-automatic annotation of modular biosynthetic gene clusters and in silico prediction of novel chemical structures. Nucleic Acids Res. 36: 6882-6892.
    Pubmed KoreaMed CrossRef
  14. Li MH, Ung PM, Zajkowski J, Garneau-Tsodikova S, Sherman DH. 2009. Automated genome mining for natural products. BMC Bioinformatics 10: 185.
    Pubmed KoreaMed CrossRef
  15. Skinnider MA, Dejong CA, Rees PN, Johnston CW, Li H, Webster AL, et al. 2015. Genomes to natural products prediction informatics for secondary metabolomes (PRISM). Nucleic Acids Res. 43: 9645-9662.
    Pubmed KoreaMed CrossRef
  16. Blin K, Wolf T, Chevrette MG, Lu X, Schwalen CJ, Kautsar SA, et al. 2017. antiSMASH 4.0-improvements in chemistry prediction and gene cluster boundary identification. Nucleic Acids Res. 45: W36-W41.
    Pubmed KoreaMed CrossRef
  17. Weber T, Kim HU. 2016. The secondary metabolite bioinformatics portal: computational tools to facilitate synthetic biology of secondary metabolite production. Synth. Syst. Biotechnol. 1: 69-79.
    Pubmed KoreaMed CrossRef
  18. Low ZJ, Pang LM, Ding Y, Cheang QW, Le Mai Hoang K, Thi Tran H, et al. 2018. Identification of a biosynthetic gene cluster for the polyene macrolactam sceliphrolactam in a Streptomyces strain isolated from mangrove sediment. Sci. Rep. 8: 1594.
    Pubmed KoreaMed CrossRef
  19. Jia N, Ding MZ, Luo H, Gao F, Yuan YJ. 2017. Complete genome sequencing and antibiotics biosynthesis pathways analysis of Streptomyces lydicus 103. Sci. Rep. 7: 44786.
  20. Zhang G, Yu D, Sang B, Feng J, Han L, Zhang X. 2017. Genome-wide analysis reveals the secondary metabolome in Streptomyces kanasensis ZX01. Genes (Basel) 8: 346.
    Pubmed KoreaMed CrossRef
  21. Ansari MZ, Yadav G, Gokhale RS, Mohanty D. 2004. NRPS-PKS: a knowledge-based resource for analysis of NRPS/PKS megasynthases. Nucleic Acids Res. 32: W405-413.
    Pubmed KoreaMed CrossRef
  22. Kim MS, Cho WJ, Song MC, Park SW, Kim K, Kim E, et al. 2017. Engineered biosynthesis of milbemycins in the avermectin high-producing strain Streptomyces avermitilis. Microb. Cell Fact. 16: 9.
    Pubmed KoreaMed CrossRef
  23. Ikeda H, Ishikawa J, Hanamoto A, Shinose M, Kikuchi H, Shiba T, et al. 2003. Complete genome sequence and comparative analysis of the industrial microorganism Streptomyces avermitilis. Nat. Biotechnol. 21: 526-531.
    Pubmed CrossRef
  24. Bode HB, Bethe B, Hofs R, Zeeck A. 2002. Big effects from small changes: possible ways to explore nature’s chemical diversity. Chembiochem. 3: 619-627.
  25. Marmann A, Aly AH, Lin W, Wang B, Proksch P. 2014. Co-cultivation--a powerful emerging tool for enhancing the chemical diversity of microorganisms. Mar. Drugs. 12: 1043-1065.
    Pubmed KoreaMed CrossRef
  26. Baral B, Akhgari A, Metsa-Ketela M. 2018. Activation of microbial secondary metabolic pathways: Avenues and challenges. Synth. Syst. Biotechnol. 3: 163-178.
    Pubmed KoreaMed CrossRef
  27. King AA, Chater KF. 1986. The expression of the Escherichia coli lacZ gene in Streptomyces. J. Gen. Microbiol. 132: 1739-1752.
    Pubmed CrossRef
  28. Lussier FX, Denis F, Shareck F. 2010. Adaptation of the highly productive T7 expression system to Streptomyces lividans. Appl. Environ. Microbiol. 76: 967-970.
    Pubmed KoreaMed CrossRef
  29. Mitra A, Angamuthu K, Jayashree HV, Nagaraja V. 2009. Occurrence, divergence and evolution of intrinsic terminators across eubacteria. Genomics 94: 110-116.
    Pubmed CrossRef
  30. Bibb MJ, White J, Ward JM, Janssen GR. 1994. The mRNA for the 23S rRNA methylase encoded by the ermE gene of Saccharopolyspora erythraea is translated in the absence of a conventional ribosome-binding site. Mol. Microbiol. 14: 533-545.
    Pubmed CrossRef
  31. Bibb MJ, Janssen GR, Ward JM. 1985. Cloning and analysis of the promoter region of the erythromycin resistance gene (ermE) of Streptomyces erythraeus. Gene 38: 215-226.
    Pubmed CrossRef
  32. Kieser T, Bibb M, Buttner M, Chater K, Hopwood D. 2000. Practical Streptomyces Genetics. John Innes Foundation, Norwich, England.
  33. Wang W, Li X, Wang J, Xiang S, Feng X, Yang K. 2013. An engineered strong promoter for Streptomycetes. Appl. Environ. Microbiol. 79: 4484-4492.
    Pubmed KoreaMed CrossRef
  34. Labes G, Bibb M, Wohlleben W. 1997. Isolation and characterization of a strong promoter element from the Streptomyces ghanaensis phage I19 using the gentamicin resistance gene (aacC1) of Tn 1696 as reporter. Microbiology 143: 1503-1512.
    Pubmed CrossRef
  35. Shao Z, Rao G, Li C, Abil Z, Luo Y, Zhao H. 2013. Refactoring the silent spectinabilin gene cluster using a plug-and-play scaffold. ACS Synth. Biol. 2: 662-669.
    Pubmed KoreaMed CrossRef
  36. Sohoni SV, Fazio A, Workman CT, Mijakovic I, Lantz AE. 2014. Synthetic promoter library for modulation of actinorhodin production in Streptomyces coelicolor A3(2). PLoS One 9: e99701.
    Pubmed KoreaMed CrossRef
  37. Seghezzi N, Amar P, Koebmann B, Jensen PR, Virolle MJ. 2011. The construction of a library of synthetic promoters revealed some specific features of strong Streptomyces promoters. Appl. Microbiol. Biotechnol. 90: 615-623.
    Pubmed CrossRef
  38. Luo Y, Zhang L, Barton KW, Zhao H. 2015. Systematic identification of a panel of strong constitutive promoters from Streptomyces albus. ACS Synth. Biol. 4: 1001-1010.
    Pubmed CrossRef
  39. Li S, Wang J, Li X, Yin S, Wang W, Yang K. 2015. Genome-wide identification and evaluation of constitutive promoters in Streptomycetes. Microb. Cell Fact. 14: 172.
    Pubmed KoreaMed CrossRef
  40. Murakami T, Holt TG, Thompson CJ. 1989. Thiostrepton-induced gene expression in Streptomyces lividans. J. Bacteriol. 171: 1459-1466.
    Pubmed KoreaMed CrossRef
  41. Takano E, White J, Thompson CJ, Bibb MJ. 1995. Construction of thiostrepton-inducible, high-copy-number expression vectors for use in Streptomyces spp. Gene 166: 133-137.
    Pubmed CrossRef
  42. Huang H, Zheng G, Jiang W, Hu H, Lu Y. 2015. One-step high-efficiency CRISPR/Cas9-mediated genome editing in Streptomyces. Acta Biochim. Biophys. Sin (Shanghai) 47: 231-243.
    Pubmed CrossRef
  43. Rodriguez-Garcia A, Combes P, Perez-Redondo R, Smith MC, Smith MC. 2005. Natural and synthetic tetracycline-inducible promoters for use in the antibiotic-producing bacteria Streptomyces. Nucleic Acids Res. 33: e87.
    Pubmed KoreaMed CrossRef
  44. Horbal L, Fedorenko V, Luzhetskyy A. 2014. Novel and tightly regulated resorcinol and cumate-inducible expression systems for Streptomyces and other actinobacteria. Appl. Microbiol. Biotechnol. 98: 8641-8655.
    Pubmed CrossRef
  45. Herai S, Hashimoto Y, Higashibata H, Maseda H, Ikeda H, Omura S, et al. 2004. Hyper-inducible expression system for Streptomycetes. Proc. Natl. Acad. Sci. USA 101: 14031-14035.
    Pubmed KoreaMed CrossRef
  46. Noguchi Y, Kashiwagi N, Uzura A, Ogino C, Kondo A, Ikeda H, et al. 2018. Development of a strictly regulated xylose-induced expression system in Streptomyces. Microb. Cell Fact. 17: 151.
    Pubmed KoreaMed CrossRef
  47. Hindle Z, Smith CP. 1994. Substrate induction and catabolite repression of the Streptomyces coelicolor glycerol operon are mediated through the GylR protein. Mol. Microbiol. 12: 737-745.
    Pubmed CrossRef
  48. Ward JM, Janssen GR, Kieser T, Bibb MJ, Buttner MJ, Bibb MJ. 1986. Construction and characterisation of a series of multi-copy promoter-probe plasmid vectors for Streptomyces using the aminoglycoside phosphotransferase gene from Tn5 as indicator. Mol. Gen. Genet. 203: 468-478.
    Pubmed CrossRef
  49. Pulido D, Jimenez A, Salas M, Mellado RP. 1987. A Bacillus subtilis phage phi 29 transcription terminator is efficiently recognized in Streptomyces lividans. Gene 56: 277-282.
    Pubmed CrossRef
  50. Cobb RE, Wang Y, Zhao H. 2015. High-efficiency multiplex genome editing of Streptomyces species using an engineered CRISPR/Cas system. ACS Synth. Biol. 4: 723-728.
    Pubmed KoreaMed CrossRef
  51. West S, Proudfoot NJ. 2009. Transcriptional termination enhances protein expression in human cells. Mol. Cell. 33: 354-364.
    Pubmed KoreaMed CrossRef
  52. Mapendano CK, Lykke-Andersen S, Kjems J, Bertrand E, Jensen TH. 2010. Crosstalk between mRNA 3’ end processing and transcription initiation. Mol. Cell. 40: 410-422.
  53. Dar D, Shamir M, Mellin JR, Koutero M, Stern-Ginossar N, Cossart P, et al. 2016. Term-seq reveals abundant ribo-regulation of antibiotics resistance in bacteria. Science 352: aad9822.
    Pubmed KoreaMed CrossRef
  54. Lu P, Vogel C, Wang R, Yao X, Marcotte EM. 2007. Absolute protein expression profiling estimates the relative contributions of transcriptional and translational regulation. Nat. Biotechnol. 25: 117-124.
    Pubmed CrossRef
  55. Jeong Y, Kim JN, Kim MW, Bucca G, Cho S, Yoon YJ, et al. 2016. The dynamic transcriptional and translational landscape of the model antibiotic producer Streptomyces coelicolor A3(2). Nat. Commun. 7: 11605.
    Pubmed KoreaMed CrossRef
  56. Makrides SC. 1996. Strategies for achieving high-level expression of genes in Escherichia coli. Microbiol. Rev. 60: 512-538.
    Pubmed KoreaMed
  57. Na D, Lee D. 2010. RBSDesigner: software for designing synthetic ribosome binding sites that yields a desired level of protein expression. Bioinformatics 26: 2633-2634.
    Pubmed CrossRef
  58. Bai C, Zhang Y, Zhao X, Hu Y, Xiang S, Miao J, et al. 2015. Exploiting a precise design of universal synthetic modular regulatory elements to unlock the microbial natural products in Streptomyces. Proc. Natl. Acad. Sci. USA 112: 12181-12186.
    Pubmed KoreaMed CrossRef
  59. Yi JS, Kim MW, Kim M, Jeong Y, Kim EJ, Cho BK, et al. 2017. A novel approach for gene expression optimization through native promoter and 5’ UTR combinations based on RNA-seq, Ribo-seq, and TSS-seq of Streptomyces coelicolor. ACS Synth. Biol. 6: 555-565.
  60. Janssen GR, Bibb MJ. 1990. Tandem promoters, tsrp1 and tsrp2, direct transcription of the thiostrepton resistance gene (tsr) of Streptomyces azureus: transcriptional initiation from tsrp2 occurs after deletion of the -35 region. Mol. Gen. Genet. 221: 339-346.
    Pubmed CrossRef
  61. Sohaskey CD, Im H, Nelson AD, Schauer AT. 1992. Tn4556 and luciferase: synergistic tools for visualizing transcription in Streptomyces. Gene 115: 67-71.
    Pubmed CrossRef
  62. Ingram C, Brawner M, Youngman P, Westpheling J. 1989. xylE functions as an efficient reporter gene in Streptomyces spp.: use for the study of galP1, a catabolite-controlled promoter. J. Bacteriol. 171: 6617-6624.
    Pubmed KoreaMed CrossRef
  63. Myronovskyi M, Welle E, Fedorenko V, Luzhetskyy A. 2011. Beta-glucuronidase as a sensitive and versatile reporter in actinomycetes. Appl. Environ Microbiol. 77: 5370-5383.
    Pubmed KoreaMed CrossRef
  64. Flores FJ, Rincon J, Martin JF. 2003. Characterization of the iron-regulated desA promoter of Streptomyces pilosus as a system for controlled gene expression in actinomycetes. Microb. Cell Fact. 2: 5.
    Pubmed KoreaMed CrossRef
  65. Willemse J, van Wezel GP. 2009. Imaging of Streptomyces coelicolor A3(2) with reduced autofluorescence reveals a novel stage of FtsZ localization. PLoS One 4: e4242.
    Pubmed KoreaMed CrossRef
  66. Santos-Beneit F, Errington J. 2017. Green fluorescent protein as a reporter for the spatial and temporal expression of actIII in Streptomyces coelicolor. Arch. Microbiol. 199: 875-880.
    Pubmed KoreaMed CrossRef
  67. Nguyen KD, Au-Young SH, Nodwell JR. 2007. Monomeric red fluorescent protein as a reporter for macromolecular localization in Streptomyces coelicolor. Plasmid 58: 167-173.
    Pubmed CrossRef
  68. Phelan RM, Sachs D, Petkiewicz SJ, Barajas JF, Blake-Hedges JM, Thompson MG, et al. 2017. Development of next generation synthetic biology tools for use in Streptomyces venezuelae. ACS Synth. Biol. 6: 159-166.
    Pubmed CrossRef
  69. Siegl T, Luzhetskyy A. 2012. Actinomycetes genome engineering approaches. Antonie Van Leeuwenhoek. 102: 503-516.
    Pubmed CrossRef
  70. Herrmann S, Siegl T, Luzhetska M, Petzke L, Jilg C, Welle E, et al. 2012. Site-specific recombination strategies for engineering actinomycete genomes. Appl. Environ. Microbiol. 78: 1804-1812.
    Pubmed KoreaMed CrossRef
  71. Khodakaramian G, Lissenden S, Gust B, Moir L, Hoskisson PA, Chater KF, et al. 2006. Expression of Cre recombinase during transient phage infection permits efficient marker removal in Streptomyces. Nucleic Acids Res. 34: e20.
    Pubmed KoreaMed CrossRef
  72. Zelyas N, Tahlan K, Jensen SE. 2009. Use of the native flp gene to generate in-frame unmarked mutations in Streptomyces spp. Gene 443: 48-54.
    Pubmed CrossRef
  73. Barrangou R, Fremaux C, Deveau H, Richards M, Boyaval P, Moineau S, et al. 2007. CRISPR provides acquired resistance against viruses in prokaryotes. Science 315: 1709-1712.
    Pubmed CrossRef
  74. Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, et al. 2013. Multiplex genome engineering using CRISPR/Cas systems. Science 339: 819-823.
    Pubmed KoreaMed CrossRef
  75. DiCarlo JE, Norville JE, Mali P, Rios X, Aach J, Church GM. 2013. Genome engineering in Saccharomyces cerevisiae using CRISPR-Cas systems. Nucleic Acids Res. 41: 4336-4343.
    Pubmed KoreaMed CrossRef
  76. Friedland AE, Tzur YB, Esvelt KM, Colaiacovo MP, Church GM, Calarco JA. 2013. Heritable genome editing in C. elegans via a CRISPR-Cas9 system. Nat. Methods 10: 741-743.
    Pubmed KoreaMed CrossRef
  77. Jiang W, Bikard D, Cox D, Zhang F, Marraffini LA. 2013. RNA-guided editing of bacterial genomes using CRISPR-Cas systems. Nat. Biotechnol. 31: 233-239.
    Pubmed KoreaMed CrossRef
  78. Li JF, Norville JE, Aach J, McCormack M, Zhang D, Bush J, et al. 2013. Multiplex and homologous recombination-mediated genome editing in Arabidopsis and Nicotiana benthamiana using guide RNA and Cas9. Nat. Biotechnol. 31: 688-691.
    Pubmed KoreaMed CrossRef
  79. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. 2012. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337: 816-821.
    Pubmed KoreaMed CrossRef
  80. Bibikova M, Beumer K, Trautman JK, Carroll D. 2003. Enhancing gene targeting with designed zinc finger nucleases. Science 300: 764.
    Pubmed CrossRef
  81. Christian M, Cermak T, Doyle EL, Schmidt C, Zhang F, Hummel A, et al. 2010. Targeting DNA double-strand breaks with TAL effector nucleases. Genetics 186: 757-761.
    Pubmed KoreaMed CrossRef
  82. Rouet P, Smih F, Jasin M. 1994. Introduction of double-strand breaks into the genome of mouse cells by expression of a rare-cutting endonuclease. Mol. Cell Biol. 14: 8096-8106.
    Pubmed KoreaMed CrossRef
  83. Doudna JA, Charpentier E. 2014. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science 346: 1258096.
    Pubmed CrossRef
  84. Anders C, Niewoehner O, Duerst A, Jinek M. 2014. Structural basis of PAM-dependent target DNA recognition by the Cas9 endonuclease. Nature 513: 569-573.
    Pubmed KoreaMed CrossRef
  85. Mojica FJ, Diez-Villasenor C, Garcia-Martinez J, Almendros C. 2009. Short motif sequences determine the targets of the prokaryotic CRISPR defence system. Microbiology 155: 733-740.
    Pubmed CrossRef
  86. Tong Y, Charusanti P, Zhang L, Weber T, Lee SY. 2015. CRISPR-Cas9 based engineering of actinomycetal genomes. ACS Synth. Biol. 4: 1020-1029.
    Pubmed CrossRef
  87. Zhang MM, Wong FT, Wang Y, Luo S, Lim YH, Heng E, et al. 2017. CRISPR-Cas9 strategy for activation of silent Streptomyces biosynthetic gene clusters. Nat. Chem. Biol. 13: 607-609.
    Pubmed KoreaMed CrossRef
  88. Zeng H, Wen S, Xu W, He Z, Zhai G, Liu Y, et al. 2015. Highly efficient editing of the actinorhodin polyketide chain length factor gene in Streptomyces coelicolor M145 using CRISPR/Cas9-CodA(sm) combined system. Appl. Microbiol. Biotechnol. 99: 10575-10585.
    Pubmed CrossRef
  89. Li L, Zheng G, Chen J, Ge M, Jiang W, Lu Y. 2017. Multiplexed site-specific genome engineering for over-producing bioactive secondary metabolites in actinomycetes. Metab. Eng. 40: 80-92.
    Pubmed CrossRef
  90. Meng J, Feng R, Zheng G, Ge M, Mast Y, Wohlleben W, et al. 2017. Improvement of pristinamycin I (PI) production in Streptomyces pristinaespiralis by metabolic engineering approaches. Synth. Syst. Biotechnol. 2: 130-136.
    Pubmed KoreaMed CrossRef
  91. Qin Z, Munnoch JT, Devine R, Holmes NA, Seipke RF, Wilkinson KA, et al. 2017. Formicamycins, antibacterial polyketides produced by Streptomyces formicae isolated from African Tetraponera plant-ants. Chem. Sci. 8: 3218-3227.
    Pubmed KoreaMed CrossRef
  92. Wang Y, Cobb RE, Zhao H. 2016. High-efficiency genome editing of Streptomyces species by an engineered CRISPR/Cas System. Methods Enzymol. 575: 271-284.
    Pubmed CrossRef
  93. Jia H, Zhang L, Wang T, Han J, Tang H, Zhang L. 2017. Development of a CRISPR/Cas9-mediated gene-editing tool in Streptomyces rimosus. Microbiology 163: 1148-1155.
    Pubmed CrossRef
  94. Tao W, Yang A, Deng Z, Sun Y. 2018. CRISPR/Cas9-based editing of Streptomyces for discovery, characterization, and production of natural products. Front Microbiol. 9: 1660.
    Pubmed KoreaMed CrossRef
  95. Liu Y, Tao W, Wen S, Li Z, Yang A, Deng Z, et al. 2015. In vitro CRISPR/Cas9 system for efficient targeted DNA editing. MBio 6: e01714-01715.
    Pubmed KoreaMed CrossRef
  96. Jiang W, Zhao X, Gabrieli T, Lou C, Ebenstein Y, Zhu TF. 2015. Cas9-assisted targeting of chromosome segments CATCH enables one-step targeted cloning of large gene clusters. Nat. Commun. 6: 8101.
    Pubmed KoreaMed CrossRef
  97. Kang HS, Charlop-Powers Z, Brady SF. 2016. Multiplexed CRISPR/Cas9-and TAR-mediated promoter engineering of natural product biosynthetic gene clusters in yeast. ACS Synth. Biol. 5: 1002-1010.
    Pubmed KoreaMed CrossRef
  98. Tao W, Yurkovich ME, Wen S, Lebe KE, Samborskyy M, Liu Y, et al. 2016. A genomics-led approach to deciphering the mechanism of thiotetronate antibiotic biosynthesis. Chem. Sci. 7: 376-385.
    Pubmed KoreaMed CrossRef
  99. Li L, Wei K, Liu X, Wu Y, Zhensg G, Chen S, et al. 2019. aMSGE: advanced multiplex site-specific genome engineering with orthogonal modular recombinases in actinomycetes. Metab. Eng. 52: 7.
    Pubmed CrossRef
  100. Qi LS, Larson MH, Gilbert LA, Doudna JA, Weissman JS, Arkin AP, et al. 2013. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 152: 1173-1183.
    Pubmed KoreaMed CrossRef
  101. Zhao Y, Li L, Zheng G, Jiang W, Deng Z, Wang Z, et al. 2018. CRISPR/dCas9-mediated multiplex gene repression in Streptomyces. Biotechnol. J. 13: e1800121.
    Pubmed CrossRef
  102. Xu Z, Wang Y, Chater KF, Ou HY, Xu HH, Deng Z, et al. 2017. Large-scale transposition mutagenesis of Streptomyces coelicolor identifies hundreds of genes influencing antibiotic biosynthesis. Appl. Environ. Microbiol. 83: e02889-02816.
    Pubmed KoreaMed CrossRef
  103. Komatsu M, Uchiyama T, Omura S, Cane DE, Ikeda H. 2010. Genome-minimized Streptomyces host for the heterologous expression of secondary metabolism. Proc. Natl. Acad. Sci. USA 107: 2646-2651.
    Pubmed KoreaMed CrossRef
  104. Gomez-Escribano JP, Bibb MJ. 2011. Engineering Streptomyces coelicolor for heterologous expression of secondary metabolite gene clusters. Microb. Biotechnol. 4: 207-215.
    Pubmed KoreaMed CrossRef
  105. Zetsche B, Gootenberg JS, Abudayyeh OO, Slaymaker IM, Makarova KS, Essletzbichler P, et al. 2015. Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system. Cell 163: 759-771.
    Pubmed KoreaMed CrossRef
  106. Li L, Wei K, Zheng G, Liu X, Chen S, Jiang W, et al. 2018. CRISPR-Cpf1-assisted multiplex genome editing and transcriptional repression in Streptomyces. Appl. Environ. Microbiol. 84: e00827-00818.
    Pubmed KoreaMed CrossRef
  107. Xu M, Wright GD. 2018. Heterologous expression-facilitated natural products’ discovery in actinomycetes. J. Ind. Microbiol. Biotechnol. 46: 415-431.
  108. Liu R, Deng Z, Liu T. 2018. Streptomyces species: Ideal chassis for natural product discovery and overproduction. Metab. Eng. 50: 74-84.
    Pubmed CrossRef
  109. Nah HJ, Pyeon HR, Kang SH, Choi SS, Kim ES. 2017. Cloning and heterologous expression of a large-sized natural product biosynthetic gene cluster in Streptomyces Species. Front. Microbiol. 8: 394.
    Pubmed KoreaMed CrossRef
  110. Rutledge PJ, Challis GL. 2015. Discovery of microbial natural products by activation of silent biosynthetic gene clusters. Nat. Rev. Microbiol. 13: 509-523.
    Pubmed CrossRef
  111. Katz M, Hover BM, Brady SF. 2016. Culture-independent discovery of natural products from soil metagenomes. J. Ind. Microbiol Biotechnol. 43: 129-141.
    Pubmed CrossRef
  112. Pyeon HR, Nah HJ, Kang SH, Choi SS, Kim ES. 2017. Heterologous expression of pikromycin biosynthetic gene cluster using Streptomyces artificial chromosome system. Microb. Cell Fact. 16: 96.
    Pubmed KoreaMed CrossRef
  113. Nah HJ, Woo MW, Choi SS, Kim ES. 2015. Precise cloning and tandem integration of large polyketide biosynthetic gene cluster using Streptomyces artificial chromosome system. Microb. Cell Fact. 14: 140.
    Pubmed KoreaMed CrossRef
  114. Du D, Wang L, Tian Y, Liu H, Tan H, Niu G. 2015. Genome engineering and direct cloning of antibiotic gene clusters via phage varphiBT1 integrase-mediated site-specific recombination in Streptomyces. Sci Rep. 5: 8740.
  115. Greunke C, Duell ER, Glockle A, Lamm K, Gulder TAM, D’Agostino PM. 2018. Direct pathway cloning (DiPaC) to unlock natural product biosynthetic potential. Metab. Eng. 47: 334-345.
    Pubmed CrossRef
  116. Shao Z, Luo Y, Zhao H. 2011. Rapid characterization and engineering of natural product biosynthetic pathways via DNA assembler. Mol. Biosyst. 7: 1056-1059.
    Pubmed KoreaMed CrossRef
  117. Fu J, Bian X, Hu S, Wang H, Huang F, Seibert PM, et al. 2012. Full-length RecE enhances linear-linear homologous recombination and facilitates direct cloning for biopro-specting. Nat. Biotechnol. 30: 440-446.
    Pubmed CrossRef
  118. Yamanaka K, Reynolds KA, Kersten RD, Ryan KS, Gonzalez DJ, Nizet V, et al. 2014. Direct cloning and refactoring of a silent lipopeptide biosynthetic gene cluster yields the antibiotic taromycin A. Proc. Natl. Acad. Sci. USA 111: 1957-1962.
    Pubmed KoreaMed CrossRef
  119. Orr-Weaver TL, Szostak JW, Rothstein RJ. 1981. Yeast transformation: a model system for the study of recombination. Proc. Natl. Acad. Sci USA 78: 6354-6358.
    Pubmed KoreaMed CrossRef
  120. Lee NC, Larionov V, Kouprina N. 2015. Highly efficient CRISPR/Cas9-mediated TAR cloning of genes and chromosomal loci from complex genomes in yeast. Nucleic Acids Res. 43: e55.
    Pubmed KoreaMed CrossRef
  121. Bu QT, Yu P, Wang J, Li ZY, Chen XA, Mao XM, et al. 2019. Rational construction of genome-reduced and high-efficient industrial Streptomyces chassis based on multiple comparative genomic approaches. Microb. Cell Fact. 18: 16.
    Pubmed KoreaMed CrossRef
  122. Myronovskyi M, Rosenkranzer B, Nadmid S, Pujic P, Normand P, Luzhetskyy A. 2018. Generation of a cluster-free Streptomyces albus chassis strains for improved heterologous expression of secondary metabolite clusters. Metab. Eng. 49: 316-324.
    Pubmed CrossRef
  123. Xu M, Wang Y, Zhao Z, Gao G, Huang SX, Kang Q, et al. 2016. Functional genome mining for metabolites encoded by large gene clusters through heterologous expression of a whole-genome bacterial artificial chromosome library in Streptomyces spp. Appl. Environ. Microbiol. 82: 5795-5805.
    Pubmed KoreaMed CrossRef
  124. Liu Q, Xiao L, Zhou Y, Deng K, Tan G, Han Y, et al. 2016. Development of Streptomyces sp. FR-008 as an emerging chassis. Synth. Syst. Biotechnol. 1: 207-214.
    Pubmed KoreaMed CrossRef
  125. Komatsu M, Komatsu K, Koiwai H, Yamada Y, Kozone I, Izumikawa M, et al. 2013. Engineered Streptomyces avermitilis host for heterologous expression of biosynthetic gene cluster for secondary metabolites. ACS Synth. Biol. 2: 384-396.
    Pubmed KoreaMed CrossRef
  126. Daniels W, Bouvin J, Busche T, Kalinowski J, Bernaerts K. 2016. Finding targets for genome reduction in Streptomyces lividans TK24 using flux balance analysis. IFAC-PapersOnLine 49: 252-257.
  127. Toro L, Pinilla L, Avignone-Rossa C, Rios-Estepa R. 2018. An enhanced genome-scale metabolic reconstruction of Streptomyces clavuligerus identifies novel strain improvement strategies. Bioprocess Biosyst. Eng. 41: 657-669.
    Pubmed CrossRef
  128. Kallifidas D, Jiang G, Ding Y, Luesch H. 2018. Rational engineering of Streptomyces albus J1074 for the overexpression of secondary metabolite gene clusters. Microb. Cell Fact. 17: 25.
    Pubmed KoreaMed CrossRef
  129. Machado H, Tuttle RN, Jensen PR. 2017. Omics-based natural product discovery and the lexicon of genome mining. Curr. Opin. Microbiol. 39: 136-142.
    Pubmed KoreaMed CrossRef
  130. Ran FA, Cong L, Yan WX, Scott DA, Gootenberg JS, Kriz AJ, et al. 2015. In vivo genome editing using Staphylococcus aureus Cas9. Nature 520: 186-191.
    Pubmed KoreaMed CrossRef
  131. Liu JJ, Orlova N, Oakes BL, Ma E, Spinner HB, Baney KLM, et al. 2019. CasX enzymes comprise a distinct family of RNA-guided genome editors. Nature 566: 218-223.
    Pubmed KoreaMed CrossRef
  132. Bikard D, Jiang W, Samai P, Hochschild A, Zhang F, Marraffini LA. 2013. Programmable repression and activation of bacterial gene expression using an engineered CRISPR-Cas system. Nucleic Acids Res. 41: 7429-7437.
    Pubmed KoreaMed CrossRef
  133. Siu KH, Chen W. 2019. Riboregulated toehold-gated gRNA for programmable CRISPR-Cas9 function. Nat. Chem. Biol. 15: 217-220.
    Pubmed CrossRef
  134. Bunet R, Song L, Mendes MV, Corre C, Hotel L, Rouhier N, et al. 2011. Characterization and manipulation of the pathway-specific late regulator AlpW reveals Streptomyces ambofaciens as a new producer of Kinamycins. J. Bacteriol. 193: 1142-1153.
    Pubmed KoreaMed CrossRef
  135. Miao V, Coeffet-Legal MF, Brian P, Brost R, Penn J, Whiting A, et al. 2005. Daptomycin biosynthesis in Streptomyces roseosporus: cloning and analysis of the gene cluster and revision of peptide stereochemistry. Microbiology 151: 1507-1523.
    Pubmed CrossRef
  136. Myronovskyi M, Rosenkranzer B, Luzhetskyy A. 2014. Iterative marker excision system. Appl. Microbiol. Biotechnol. 98: 4557-4570.
    Pubmed CrossRef
  137. Thapa LP, Oh TJ, Lee HC, Liou K, Park JW, Yoon YJ, et al. 2007. Heterologous expression of the kanamycin biosynthetic gene cluster (pSKC2) in Streptomyces venezuelae YJ003. Appl. Microbiol. Biotechnol. 76: 1357-1364.
    Pubmed CrossRef
  138. Jones AC, Gust B, Kulik A, Heide L, Buttner MJ, Bibb MJ. 2013. Phage p1-derived artificial chromosomes facilitate heterologous expression of the FK506 gene cluster. PLoS One 8: e69319.
    Pubmed KoreaMed CrossRef
  139. Steffensky M, Muhlenweg A, Wang ZX, Li SM, Heide L. 2000. Identification of the novobiocin biosynthetic gene cluster of Streptomyces spheroides NCIB 11891. Antimicrob. Agents Chemother. 44: 1214-1222.
    Pubmed KoreaMed CrossRef
  140. Zhao Z, Shi T, Xu M, Brock NL, Zhao YL, Wang Y, et al. 2016. Hybrubins: Bipyrrole tetramic acids obtained by crosstalk between a truncated undecylprodigiosin pathway and heterologous tetramic acid biosynthetic genes. Org. Lett. 18: 572-575.
    Pubmed CrossRef