Synthetic biology and metabolic engineering of actinomycetes for natural product discovery.
暂无分享,去创建一个
Yaojun Tong | Tilmann Weber | Sang Yup Lee | Emilia Palazzotto | S. Lee | Y. Tong | T. Weber | E. Palazzotto
[1] Qian Liu,et al. Development of Streptomyces sp. FR-008 as an emerging chassis☆ , 2016, Synthetic and systems biotechnology.
[2] Françoise Gilard,et al. Carbon-Flux Distribution within Streptomyces coelicolor Metabolism: A Comparison between the Actinorhodin-Producing Strain M145 and Its Non-Producing Derivative M1146 , 2013, PloS one.
[3] O. Genilloud. Actinomycetes: still a source of novel antibiotics. , 2017, Natural product reports.
[4] Kai Blin,et al. antiSMASH 4.0—improvements in chemistry prediction and gene cluster boundary identification , 2017, Nucleic Acids Res..
[5] Hao Wang,et al. RAVEN 2.0: A versatile toolbox for metabolic network reconstruction and a case study on Streptomyces coelicolor , 2018, bioRxiv.
[6] R. H. Baltz,et al. Gene transfer and transposition mutagenesis in Streptomyces roseosporus: mapping of insertions that influence daptomycin or pigment production. , 1996, Microbiology.
[7] Z. Deng,et al. A Cellulose Synthase-Like Protein Involved in Hyphal Tip Growth and Morphological Differentiation in Streptomyces , 2008, Journal of bacteriology.
[8] P. Dorrestein,et al. Molecular Basis for Chloronium-mediated Meroterpene Cyclization , 2007, Journal of Biological Chemistry.
[9] Rosa Alduina,et al. Artificial Chromosomes to Explore and to Exploit Biosynthetic Capabilities of Actinomycetes , 2012, Journal of biomedicine & biotechnology.
[10] Hyun Uk Kim,et al. Current state and applications of microbial genome-scale metabolic models , 2017 .
[11] S. Donadio,et al. Large inserts for big data: artificial chromosomes in the genomic era. , 2018, FEMS microbiology letters.
[12] M. Marahiel. A structural model for multimodular NRPS assembly lines. , 2016, Natural product reports.
[13] A. Scaloni,et al. Differential proteomic analysis reveals novel links between primary metabolism and antibiotic production in Amycolatopsis balhimycina , 2010, Proteomics.
[14] Yunkun Liu,et al. In Vitro CRISPR/Cas9 System for Efficient Targeted DNA Editing , 2015, mBio.
[15] B. Ye,et al. Lysine acetylproteome analysis suggests its roles in primary and secondary metabolism in Saccharopolyspora erythraea , 2014, Applied Microbiology and Biotechnology.
[16] Min Woo Kim,et al. 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. , 2017, ACS synthetic biology.
[17] A. Luzhetskyy,et al. Native and engineered promoters in natural product discovery. , 2016, Natural product reports.
[18] Kira J Weissman,et al. Genetic engineering of modular PKSs: from combinatorial biosynthesis to synthetic biology. , 2016, Natural product reports.
[19] Huimin Zhao,et al. High-Efficiency Multiplex Genome Editing of Streptomyces Species Using an Engineered CRISPR/Cas System , 2014, ACS synthetic biology.
[20] Yinhua Lu,et al. Multiplexed site-specific genome engineering for overproducing bioactive secondary metabolites in actinomycetes. , 2017, Metabolic engineering.
[21] J. Kalinowski,et al. Development of a Biosensor Concept to Detect the Production of Cluster-Specific Secondary Metabolites. , 2017, ACS synthetic biology.
[22] Tyler W. H. Backman,et al. ClusterCAD: a computational platform for type I modular polyketide synthase design , 2017, Nucleic Acids Res..
[23] P. Bruheim,et al. High-yield actinorhodin production in fed-batch culture by a Streptomyces lividans strain overexpressing the pathway-specific activator gene actII-ORF4 , 2002, Journal of Industrial Microbiology and Biotechnology.
[24] J. Suh,et al. Rapid construction of a Bacterial Artificial Chromosomal (BAC) expression vector using designer DNA fragments. , 2014, Plasmid.
[25] J. Hashimoto,et al. Reprogramming of the antimycin NRPS-PKS assembly lines inspired by gene evolution , 2018, Nature Communications.
[26] Hal S Alper,et al. Enabling tools for high-throughput detection of metabolites: Metabolic engineering and directed evolution applications. , 2017, Biotechnology advances.
[27] Q. Tu,et al. Direct cloning and heterologous expression of the salinomycin biosynthetic gene cluster from Streptomyces albus DSM41398 in Streptomyces coelicolor A3(2) , 2015, Scientific Reports.
[28] J. Davies. Are antibiotics naturally antibiotics? , 2006, Journal of Industrial Microbiology and Biotechnology.
[29] Tilmann Weber,et al. Characterization of the ‘pristinamycin supercluster’ of Streptomyces pristinaespiralis , 2011, Microbial biotechnology.
[30] Hyun Uk Kim,et al. Metabolic engineering of microorganisms: general strategies and drug production. , 2009, Drug discovery today.
[31] Shana Topp,et al. Random Walks to Synthetic Riboswitches—A High‐Throughput Selection Based on Cell Motility , 2008, Chembiochem : a European journal of chemical biology.
[32] M. Abdelfattah,et al. Isolation, Characterization, and Heterologous Expression of the Biosynthesis Gene Cluster for the Antitumor Anthracycline Steffimycin , 2006, Applied and Environmental Microbiology.
[33] Yinhua Lu,et al. One-step high-efficiency CRISPR/Cas9-mediated genome editing in Streptomyces. , 2015, Acta biochimica et biophysica Sinica.
[34] Frank Buchholz,et al. A new logic for DNA engineering using recombination in Escherichia coli , 1998, Nature Genetics.
[35] J. Nodwell,et al. Genome Context as a Predictive Tool for Identifying Regulatory Targets of the TetR Family Transcriptional Regulators , 2012, PloS one.
[36] K. Chater,et al. Engineering of Primary Carbohydrate Metabolism for Increased Production of Actinorhodin in Streptomyces coelicolor , 2006, Applied and Environmental Microbiology.
[37] John A McLean,et al. Comparative mass spectrometry-based metabolomics strategies for the investigation of microbial secondary metabolites. , 2017, Natural product reports.
[38] Lin Wang,et al. Comparative metabolomics reveals the mechanism of avermectin production enhancement by S-adenosylmethionine , 2017, Journal of Industrial Microbiology & Biotechnology.
[39] Michael G Thomas,et al. Biosynthesis of polyketide synthase extender units. , 2009, Natural product reports.
[40] H. Ikeda,et al. Development of a strictly regulated xylose-induced expression system in Streptomyces , 2018, Microbial Cell Factories.
[41] W. Tao,et al. CRISPR/Cas9-Based Editing of Streptomyces for Discovery, Characterization, and Production of Natural Products , 2018, Front. Microbiol..
[42] K. Chater,et al. Heterologous Expression of Novobiocin and Clorobiocin Biosynthetic Gene Clusters , 2005, Applied and Environmental Microbiology.
[43] Mingzi M. Zhang,et al. Auroramycin: A Potent Antibiotic from Streptomyces roseosporus by CRISPR‐Cas9 Activation , 2018, Chembiochem : a European journal of chemical biology.
[44] Jay D Keasling,et al. Comprehensive in Vitro Analysis of Acyltransferase Domain Exchanges in Modular Polyketide Synthases and Its Application for Short-Chain Ketone Production. , 2017, ACS synthetic biology.
[45] Rolf Müller,et al. Full-length RecE enhances linear-linear homologous recombination and facilitates direct cloning for bioprospecting , 2012, Nature Biotechnology.
[46] Hyun Uk Kim,et al. Systems biology and biotechnology of Streptomyces species for the production of secondary metabolites. , 2014, Biotechnology advances.
[47] Tilmann Weber,et al. Metabolic engineering with systems biology tools to optimize production of prokaryotic secondary metabolites. , 2016, Natural product reports.
[48] S. Lee,et al. Systems strategies for developing industrial microbial strains , 2015, Nature Biotechnology.
[49] Tilmann Weber,et al. Polyketide Bioderivatization Using the Promiscuous Acyltransferase KirCII. , 2017, ACS synthetic biology.
[50] Yawei Zhao,et al. A stepwise increase in pristinamycin II biosynthesis by Streptomyces pristinaespiralis through combinatorial metabolic engineering. , 2015, Metabolic engineering.
[51] Transcriptome-guided identification of SprA as a pleiotropic regulator in Streptomyces chattanoogensis , 2015, Applied Microbiology and Biotechnology.
[52] T. Eguchi,et al. Protein-protein interactions in polyketide synthase-nonribosomal peptide synthetase hybrid assembly lines. , 2018, Natural product reports.
[53] Andriy Luzhetskyy,et al. A set of synthetic versatile genetic control elements for the efficient expression of genes in Actinobacteria , 2018, Scientific Reports.
[54] Huanhuan Liu,et al. Combining metabolomics and network analysis to improve tacrolimus production in Streptomyces tsukubaensis using different exogenous feedings , 2017, Journal of Industrial Microbiology & Biotechnology.
[55] Á. Manteca,et al. ArgR of Streptomyces coelicolor Is a Pleiotropic Transcriptional Regulator: Effect on the Transcriptome, Antibiotic Production, and Differentiation in Liquid Cultures , 2018, Front. Microbiol..
[56] F. Kopp,et al. Macrocyclization strategies in polyketide and nonribosomal peptide biosynthesis. , 2007, Natural product reports.
[57] Kai Blin,et al. Recent development of antiSMASH and other computational approaches to mine secondary metabolite biosynthetic gene clusters , 2017, Briefings Bioinform..
[58] Chunbo Lou,et al. Exploiting a precise design of universal synthetic modular regulatory elements to unlock the microbial natural products in Streptomyces , 2015, Proceedings of the National Academy of Sciences.
[59] J. Westpheling,et al. Branched-chain amino acid catabolism provides precursors for the Type II polyketide antibiotic, actinorhodin, via pathways that are nutrient dependent , 2008, Journal of Industrial Microbiology & Biotechnology.
[60] V. Kaberdin,et al. Translation initiation and the fate of bacterial mRNAs. , 2006, FEMS microbiology reviews.
[61] C. Consolandi,et al. Time-Resolved Transcriptomics and Constraint-Based Modeling Identify System-Level Metabolic Features and Overexpression Targets to Increase Spiramycin Production in Streptomyces ambofaciens , 2017, Front. Microbiol..
[62] Seung-Hoon Kang,et al. Cloning and Heterologous Expression of a Large-sized Natural Product Biosynthetic Gene Cluster in Streptomyces Species , 2017, Front. Microbiol..
[63] R. Pérez-Redondo,et al. Natural and synthetic tetracycline-inducible promoters for use in the antibiotic-producing bacteria Streptomyces , 2005, Nucleic acids research.
[64] S. Brady,et al. Functional analysis of environmental DNA-derived type II polyketide synthases reveals structurally diverse secondary metabolites , 2011, Proceedings of the National Academy of Sciences.
[65] R. Reid,et al. Chalcomycin Biosynthesis Gene Cluster from Streptomyces bikiniensis: Novel Features of an Unusual Ketolide Produced through Expression of the chm Polyketide Synthase in Streptomyces fradiae , 2004, Antimicrobial Agents and Chemotherapy.
[66] Jesus F. Barajas,et al. Engineered polyketides: Synergy between protein and host level engineering , 2017, Synthetic and systems biotechnology.
[67] Fuzhong Zhang,et al. Applications and advances of metabolite biosensors for metabolic engineering. , 2015, Metabolic engineering.
[68] Jeong Wook Lee,et al. Systems metabolic engineering of microorganisms for natural and non-natural chemicals. , 2012, Nature chemical biology.
[69] B. Ye,et al. Dissecting and engineering of the TetR family regulator SACE_7301 for enhanced erythromycin production in Saccharopolyspora erythraea , 2014, Microbial Cell Factories.
[70] Barrie Wilkinson,et al. Biosynthesis of the Novel Macrolide Antibiotic Anthracimycin. , 2015, ACS chemical biology.
[71] Kira J Weissman,et al. The structural biology of biosynthetic megaenzymes. , 2015, Nature chemical biology.
[72] R. Kolter,et al. Antibiotics as signal molecules. , 2011, Chemical reviews.
[73] J. Rohr,et al. Properties of lanK-based regulatory circuit involved in landomycin biosynthesis in Streptomyces cyanogenus S136 , 2010, Russian Journal of Genetics.
[74] M. Bibb,et al. Identification and Heterologous Expression of the Chaxamycin Biosynthesis Gene Cluster from Streptomyces leeuwenhoekii , 2015, Applied and Environmental Microbiology.
[75] Philippe Normand,et al. Generation of a cluster-free Streptomyces albus chassis strains for improved heterologous expression of secondary metabolite clusters. , 2018, Metabolic engineering.
[76] Tilmann Weber,et al. The evolution of genome mining in microbes - a review. , 2016, Natural product reports.
[77] Huimin Zhao,et al. CRISPR-Cas9 strategy for activation of silent Streptomyces biosynthetic gene clusters , 2017, Nature chemical biology.
[78] Geon-Woo Kim,et al. Generation of New Complestatin Analogues by Heterologous Expression of the Complestatin Biosynthetic Gene Cluster from Streptomyces chartreusis AN1542 , 2016, Chembiochem : a European journal of chemical biology.
[79] J. Bérdy. Thoughts and facts about antibiotics: Where we are now and where we are heading , 2012, The Journal of Antibiotics.
[80] M. Bibb,et al. Heterologous expression of natural product biosynthetic gene clusters in Streptomyces coelicolor: from genome mining to manipulation of biosynthetic pathways , 2014, Journal of Industrial Microbiology & Biotechnology.
[81] B. Moore,et al. Direct Capture and Heterologous Expression of Salinispora Natural Product Genes for the Biosynthesis of Enterocin , 2014, Journal of natural products.
[82] Hui Hong,et al. Evidence for an iterative module in chain elongation on the azalomycin polyketide synthase , 2016, Beilstein journal of organic chemistry.
[83] M. Staver,et al. Acyltransferase domain substitutions in erythromycin polyketide synthase yield novel erythromycin derivatives , 1997, Journal of bacteriology.
[84] Richard H. Baltz,et al. Streptomyces and Saccharopolyspora hosts for heterologous expression of secondary metabolite gene clusters , 2010, Journal of Industrial Microbiology & Biotechnology.
[85] J. Engels,et al. Promoter constructions for efficient secretion expression in Streptomyces lividans , 2004, Applied Microbiology and Biotechnology.
[86] James E. DiCarlo,et al. RNA-Guided Human Genome Engineering via Cas9 , 2013, Science.
[87] V. Fedorenko,et al. Novel and tightly regulated resorcinol and cumate-inducible expression systems for Streptomyces and other actinobacteria , 2014, Applied Microbiology and Biotechnology.
[88] B. Shen,et al. Engineered production of iso-migrastatin in heterologous Streptomyces hosts. , 2009, Bioorganic & medicinal chemistry.
[89] C. Méndez,et al. Improving production of bioactive secondary metabolites in actinomycetes by metabolic engineering. , 2008, Metabolic engineering.
[90] Paula Y. Calle,et al. Cloning large natural product gene clusters from the environment: Piecing environmental DNA gene clusters back together with TAR , 2010, Biopolymers.
[91] I. Brikun,et al. Engineering of the methylmalonyl-CoA metabolite node of Saccharopolyspora erythraea for increased erythromycin production. , 2007, Metabolic engineering.
[92] Dong In Kim,et al. Applications of genome-scale metabolic network model in metabolic engineering , 2015, Journal of Industrial Microbiology & Biotechnology.
[93] Yaojun Tong,et al. CRISPR/Cas-based genome engineering in natural product discovery. , 2019, Natural product reports.
[94] J. Nielsen,et al. Increased glycopeptide production after overexpression of shikimate pathway genes being part of the balhimycin biosynthetic gene cluster. , 2010, Metabolic engineering.
[95] S. Horinouchi,et al. AdpA, a Central Transcriptional Regulator in the A-Factor Regulatory Cascade That Leads to Morphological Development and Secondary Metabolism in Streptomyces griseus , 2005, Bioscience, biotechnology, and biochemistry.
[96] K. Ochi,et al. Induction of actinorhodin production by rpsL (encoding ribosomal protein S12) mutations that confer streptomycin resistance in Streptomyces lividans and Streptomyces coelicolor A3(2) , 1996, Journal of bacteriology.
[97] D. Cane,et al. Enhancing the modularity of the modular polyketide synthases: transacylation in modular polyketide synthases catalyzed by malonyl-CoA:ACP transacylase. , 2003, Journal of the American Chemical Society.
[98] S. Lee,et al. Filling the Gaps in the Kirromycin Biosynthesis: Deciphering the Role of Genes Involved in Ethylmalonyl-CoA Supply and Tailoring Reactions , 2018, Scientific Reports.
[99] Qian Wang,et al. Pathway optimization and key enzyme evolution of N-acetylneuraminate biosynthesis using an in vivo aptazyme-based biosensor. , 2017, Metabolic engineering.
[100] Jacob L. Mueller,et al. CRISPR-mediated isolation of specific megabase segments of genomic DNA , 2017, Nucleic acids research.
[101] M. Bibb. Understanding and manipulating antibiotic production in actinomycetes. , 2013, Biochemical Society transactions.
[102] S. Nam,et al. Engineered biosynthesis of milbemycins in the avermectin high-producing strain Streptomyces avermitilis , 2017, Microbial Cell Factories.
[103] A. Luzhetskyy,et al. Dual control system – A novel scaffolding architecture of an inducible regulatory device for the precise regulation of gene expression , 2016, Metabolic engineering.
[104] Shuangjun Lin,et al. Sparsomycin Biosynthesis Highlights Unusual Module Architecture and Processing Mechanism in Non-ribosomal Peptide Synthetase. , 2015, ACS chemical biology.
[105] R. Barrangou,et al. CRISPR Provides Acquired Resistance Against Viruses in Prokaryotes , 2007, Science.
[106] A. Scaloni,et al. Tryptophan promotes morphological and physiological differentiation in Streptomyces coelicolor , 2015, Applied Microbiology and Biotechnology.
[107] Hee-Ju Nah,et al. Precise cloning and tandem integration of large polyketide biosynthetic gene cluster using Streptomyces artificial chromosome system , 2015, Microbial Cell Factories.
[108] Huimin Zhao,et al. Systematic Identification of a Panel of Strong Constitutive Promoters from Streptomyces albus. , 2015, ACS synthetic biology.
[109] V. Larionov,et al. Specific isolation of human rDNA genes by TAR cloning. , 1997, Gene.
[110] Satoshi Omura,et al. Genome-minimized Streptomyces host for the heterologous expression of secondary metabolism , 2010, Proceedings of the National Academy of Sciences.
[111] Yuan Lu. Cell-free synthetic biology: Engineering in an open world , 2017, Synthetic and systems biotechnology.
[112] Eugene V Koonin,et al. Diversity, classification and evolution of CRISPR-Cas systems. , 2017, Current opinion in microbiology.
[113] Gang Liu,et al. Molecular Regulation of Antibiotic Biosynthesis in Streptomyces , 2013, Microbiology and Molecular Reviews.
[114] W. Wohlleben,et al. Identification and functional characterization of phenylglycine biosynthetic genes involved in pristinamycin biosynthesis in Streptomyces pristinaespiralis. , 2011, Journal of biotechnology.
[115] B. Suess,et al. Conditional control of gene expression by synthetic riboswitches in Streptomyces coelicolor. , 2015, Methods in enzymology.
[116] C. Méndez,et al. An ABC transporter is essential for resistance to the antitumor agent mithramycin in the producerStreptomyces argillaceus , 1996, Molecular and General Genetics MGG.
[117] Jörn Piel,et al. Evolution-guided engineering of nonribosomal peptide synthetase adenylation domains , 2013 .
[118] Charles B. Larson,et al. PCR-Independent Method of Transformation-Associated Recombination Reveals the Cosmomycin Biosynthetic Gene Cluster in an Ocean Streptomycete. , 2017, Journal of natural products.
[119] J. Hashimoto,et al. Novel thioviridamide derivative—JBIR-140: heterologous expression of the gene cluster for thioviridamide biosynthesis , 2015, The Journal of Antibiotics.
[120] Keshav K. Nepal,et al. Streptomycetes: Surrogate hosts for the genetic manipulation of biosynthetic gene clusters and production of natural products. , 2019, Biotechnology advances.
[121] J. Davies,et al. The world of subinhibitory antibiotic concentrations. , 2006, Current opinion in microbiology.
[122] Jong Hyun Choi,et al. Repurposing type III polyketide synthase as a malonyl-CoA biosensor for metabolic engineering in bacteria , 2018, Proceedings of the National Academy of Sciences.
[123] A. Luzhetskyy,et al. Cloning and Heterologous Expression of the Grecocycline Biosynthetic Gene Cluster , 2016, PloS one.
[124] Kay Nieselt,et al. The dynamic architecture of the metabolic switch in Streptomyces coelicolor , 2010, BMC Genomics.
[125] Y. Hayakawa,et al. Identification of a prodigiosin cyclization gene in the roseophilin producer and production of a new cyclized prodigiosin in a heterologous host , 2016, The Journal of Antibiotics.
[126] Jay D Keasling,et al. Development of biosensors and their application in metabolic engineering. , 2015, Current opinion in chemical biology.
[127] Wenjun Jiang,et al. Cas9-Assisted Targeting of CHromosome segments CATCH enables one-step targeted cloning of large gene clusters , 2015, Nature Communications.
[128] M. Bibb,et al. 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. , 1997, Microbiology.
[129] J. Ju,et al. Identification of the grincamycin gene cluster unveils divergent roles for GcnQ in different hosts, tailoring the L-rhodinose moiety. , 2013, Organic letters.
[130] Gavin J. Williams. Engineering polyketide synthases and nonribosomal peptide synthetases. , 2013, Current opinion in structural biology.
[131] C. Thompson,et al. Thiostrepton-induced gene expression in Streptomyces lividans , 1989, Journal of bacteriology.
[132] Beatrix Suess,et al. Synthetic riboswitches for the conditional control of gene expression in Streptomyces coelicolor. , 2013, Microbiology.
[133] Yong-Quan Li,et al. Gamma-Butyrolactone Regulatory System of Streptomyces chattanoogensis Links Nutrient Utilization, Metabolism, and Development , 2011, Applied and Environmental Microbiology.
[134] T. Weber,et al. Omics and multi-omics approaches to study the biosynthesis of secondary metabolites in microorganisms. , 2018, Current opinion in microbiology.
[135] K. Ochi,et al. A possible mechanism for lincomycin induction of secondary metabolism in Streptomyces coelicolor A3(2) , 2018, Antonie van Leeuwenhoek.
[136] Satoshi Yuzawa,et al. Role of a conserved arginine residue in linkers between the ketosynthase and acyltransferase domains of multimodular polyketide synthases. , 2012, Biochemistry.
[137] Hang Wu,et al. Engineering of an Lrp family regulator SACE_Lrp improves erythromycin production in Saccharopolyspora erythraea. , 2017, Metabolic engineering.
[138] N. Kelleher,et al. Directed evolution of the nonribosomal peptide synthetase AdmK generates new andrimid derivatives in vivo. , 2011, Chemistry & biology.
[139] E. Graziani,et al. Rapid cloning and heterologous expression of the meridamycin biosynthetic gene cluster using a versatile Escherichia coli-streptomyces artificial chromosome vector, pSBAC. , 2009, Journal of natural products.
[140] T. Gulder,et al. Direct Pathway Cloning (DiPaC) to unlock natural product biosynthetic potential. , 2018, Metabolic engineering.
[141] M. Bibb,et al. Transformation of plasmid DNA into Streptomyces at high frequency , 1978, Nature.
[142] Z. Qin,et al. Sequential deletion of all the polyketide synthase and nonribosomal peptide synthetase biosynthetic gene clusters and a 900-kb subtelomeric sequence of the linear chromosome of Streptomyces coelicolor. , 2012, FEMS microbiology letters.
[143] T. Kieser. Practical streptomyces genetics , 2000 .
[144] X. Chen,et al. Specific cloning of human DNA as yeast artificial chromosomes by transformation-associated recombination. , 1996, Proceedings of the National Academy of Sciences of the United States of America.
[145] P. Dorrestein,et al. Direct cloning and refactoring of a silent lipopeptide biosynthetic gene cluster yields the antibiotic taromycin A , 2014, Proceedings of the National Academy of Sciences.
[146] Frank Wesche,et al. De novo design and engineering of non-ribosomal peptide synthetases. , 2018, Nature chemistry.
[147] Young Ji Yoo,et al. Enhanced FK506 production in Streptomyces clavuligerus CKD1119 by engineering the supply of methylmalonyl-CoA precursor , 2009, Journal of Industrial Microbiology & Biotechnology.
[148] Xiaoqiang Jia,et al. Genome-scale metabolic network guided engineering of Streptomyces tsukubaensis for FK506 production improvement , 2013, Microbial Cell Factories.
[149] K. Ochi,et al. Lincomycin at Subinhibitory Concentrations Potentiates Secondary Metabolite Production by Streptomyces spp , 2015, Applied and Environmental Microbiology.
[150] M. Tarry,et al. Piecing together nonribosomal peptide synthesis. , 2018, Current opinion in structural biology.
[151] L. Bai,et al. Engineering validamycin production by tandem deletion of γ-butyrolactone receptor genes in Streptomyces hygroscopicus 5008. , 2015, Metabolic engineering.
[152] S. Lee,et al. Genome‐Scale Metabolic Reconstruction of Actinomycetes for Antibiotics Production , 2018, Biotechnology journal.
[153] Ying Wen,et al. Increasing Avermectin Production in Streptomyces avermitilis by Manipulating the Expression of a Novel TetR-Family Regulator and Its Target Gene Product , 2015, Applied and Environmental Microbiology.
[154] M. Sánchez-Hidalgo,et al. Genome Mining of Streptomyces sp. Tü 6176: Characterization of the Nataxazole Biosynthesis Pathway , 2015, Chembiochem : a European journal of chemical biology.
[155] C. Workman,et al. Synthetic Promoter Library for Modulation of Actinorhodin Production in Streptomyces coelicolor A3(2) , 2014, PloS one.
[156] Courtney C Aldrich,et al. Engineering the substrate specificity of the DhbE adenylation domain by yeast cell surface display. , 2013, Chemistry & biology.
[157] Paul S Freemont,et al. Streptomyces venezuelae TX-TL - a next generation cell-free synthetic biology tool. , 2017, Biotechnology journal.
[158] C. Khosla,et al. Cloning and heterologous expression of the epothilone gene cluster. , 2000, Science.
[159] M. Metsä-Ketelä,et al. Targeted activation of silent natural product biosynthesis pathways by reporter-guided mutant selection. , 2015, Metabolic engineering.
[160] Z. Deng,et al. Activity of a Streptomyces transcriptional terminator in Escherichia coli. , 1987, Nucleic acids research.
[161] Tilmann Weber,et al. Genome‐scale metabolic representation of Amycolatopsis balhimycina , 2012, Biotechnology and bioengineering.
[162] E. Bossi,et al. Artificial chromosomes for antibiotic-producing actinomycetes , 2000, Nature Biotechnology.
[163] S. Horinouchi,et al. Regulation of secondary metabolism and cell differentiation in Streptomyces: A-factor as a microbial hormone and the AfsR protein as a component of a two-component regulatory system. , 1992, Gene.
[164] L. Tang,et al. Construction of the co-expression plasmids of fostriecin polyketide synthases and heterologous expression in Streptomyces , 2015, Pharmaceutical biology.
[165] L. Quek,et al. Reconstruction of the Saccharopolyspora erythraea genome-scale model and its use for enhancing erythromycin production , 2012, Antonie van Leeuwenhoek.
[166] D. Cane,et al. Dissecting the role of acyltransferase domains of modular polyketide synthases in the choice and stereochemical fate of extender units. , 1999, Biochemistry.
[167] Neil L Kelleher,et al. A Roadmap for Natural Product Discovery Based on Large-Scale Genomics and Metabolomics , 2014, Nature chemical biology.
[168] M. Bibb,et al. 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 , 1994, Molecular microbiology.
[169] M. V. Mendes,et al. Secondary metabolites overproduction through transcriptional gene cluster refactoring. , 2018, Metabolic engineering.
[170] Yinhua Lu,et al. Improvement of pristinamycin I (PI) production in Streptomyces pristinaespiralis by metabolic engineering approaches , 2017, Synthetic and systems biotechnology.
[171] J. Anné,et al. Streptomyces lividans as host for heterologous protein production. , 1993, FEMS microbiology letters.
[172] János Bérdy,et al. Bioactive microbial metabolites. , 2005, The Journal of antibiotics.
[173] D. Hopwood,et al. Cloning and expression in a heterologous host of the complete set of genes for biosynthesis of the Streptomyces coelicolor antibiotic undecylprodigiosin. , 1990, Gene.
[174] J. Nielsen,et al. The influence of carbon sources and morphology on nystatin production by Streptomyces noursei. , 2002, Journal of biotechnology.
[175] S. K. Desai,et al. A high-throughput screen for synthetic riboswitches reveals mechanistic insights into their function. , 2007, Chemistry & biology.
[176] Arwa Al-Dilaimi,et al. The secreted metabolome of Streptomyces chartreusis and implications for bacterial chemistry , 2018, Proceedings of the National Academy of Sciences.
[177] Hahk-Soo Kang,et al. Library of Synthetic Streptomyces Regulatory Sequences for Use in Promoter Engineering of Natural Product Biosynthetic Gene Clusters. , 2018, ACS synthetic biology.
[178] Genome Sequences of Three Tunicamycin-Producing Streptomyces Strains, S. chartreusis NRRL 12338, S. chartreusis NRRL 3882, and S. lysosuperificus ATCC 31396 , 2011, Journal of bacteriology.
[179] M. Bibb,et al. Cloning and analysis of the promoter region of the erythromycin resistance gene (ermE) of Streptomyces erythraeus. , 1985, Gene.
[180] Guojun Wang,et al. Dramatic Activation of Antibiotic Production in Streptomyces coelicolor by Cumulative Drug Resistance Mutations , 2008, Applied and Environmental Microbiology.
[181] Jae Sung Cho,et al. A comprehensive metabolic map for production of bio-based chemicals , 2019, Nature Catalysis.
[182] Seung-Hoon Kang,et al. Heterologous expression of pikromycin biosynthetic gene cluster using Streptomyces artificial chromosome system , 2017, Microbial Cell Factories.
[183] S. Brady,et al. Multiplexed CRISPR/Cas9- and TAR-Mediated Promoter Engineering of Natural Product Biosynthetic Gene Clusters in Yeast. , 2016, ACS synthetic biology.
[184] H. Tan,et al. Coordinative Modulation of Chlorothricin Biosynthesis by Binding of the Glycosylated Intermediates and End Product to a Responsive Regulator ChlF1* , 2016, The Journal of Biological Chemistry.
[185] M. Nishiyama,et al. Biosynthesis of Versipelostatin: Identification of an Enzyme-Catalyzed [4+2]-Cycloaddition Required for Macrocyclization of Spirotetronate-Containing Polyketides , 2014, Journal of the American Chemical Society.
[186] Feng Zhang,et al. Crystal Structure of Cas9 in Complex with Guide RNA and Target DNA , 2014, Cell.
[187] D. Court,et al. Recombineering: a homologous recombination-based method of genetic engineering , 2009, Nature Protocols.
[188] Carmen Méndez,et al. Engineering precursor metabolite pools for increasing production of antitumor mithramycins in Streptomyces argillaceus. , 2013, Metabolic engineering.
[189] Dennis Claessen,et al. Morphogenesis of Streptomyces in submerged cultures. , 2014, Advances in applied microbiology.
[190] P. Leadlay,et al. A hybrid modular polyketide synthase obtained by domain swapping. , 1996, Chemistry & biology.
[191] Wenyu Lu,et al. Genome-scale metabolic network reconstruction of Saccharopolyspora spinosa for Spinosad Production improvement , 2014, Microbial Cell Factories.
[192] Katharine R. Watts,et al. Comparative Analysis of the Substrate Specificity of trans- versus cis-Acyltransferases of Assembly Line Polyketide Synthases , 2014, Biochemistry.
[193] Tiangang Liu,et al. Heterologous Biosynthesis of Spinosad: An Omics-Guided Large Polyketide Synthase Gene Cluster Reconstitution in Streptomyces. , 2017, ACS synthetic biology.
[194] U. Rix,et al. Mithramycin SK, a novel antitumor drug with improved therapeutic index, mithramycin SA, and demycarosyl-mithramycin SK: three new products generated in the mithramycin producer Streptomyces argillaceus through combinatorial biosynthesis. , 2003, Journal of the American Chemical Society.
[195] Junko Hashimoto,et al. Engineered Streptomyces avermitilis host for heterologous expression of biosynthetic gene cluster for secondary metabolites. , 2013, ACS synthetic biology.
[196] León F. Toro,et al. An enhanced genome-scale metabolic reconstruction of Streptomyces clavuligerus identifies novel strain improvement strategies , 2018, Bioprocess and Biosystems Engineering.
[197] X. He,et al. Development of a Genetic System for Combinatorial Biosynthesis of Lipopeptides in Streptomyces fradiae and Heterologous Expression of the A54145 Biosynthesis Gene Cluster , 2010, Applied and Environmental Microbiology.
[198] B. Moore,et al. Biosynthetic Pathway Connects Cryptic Ribosomally Synthesized Posttranslationally Modified Peptide Genes with Pyrroloquinoline Alkaloids. , 2016, Cell chemical biology.
[199] Xianpu Ni,et al. Modulation of kanamycin B and kanamycin A biosynthesis in Streptomyces kanamyceticus via metabolic engineering , 2017, PloS one.
[200] Youming Zhang,et al. RecE/RecT and Redα/Redβ initiate double-stranded break repair by specifically interacting with their respective partners , 2000, Genes & Development.
[201] Christopher J. Silva,et al. Daptomycin biosynthesis in Streptomyces roseosporus: cloning and analysis of the gene cluster and revision of peptide stereochemistry. , 2005, Microbiology.
[202] Beatrix Suess,et al. Engineered riboswitches: Expanding researchers' toolbox with synthetic RNA regulators , 2012, FEBS letters.
[203] Z. Qin,et al. Development of a gene cloning system in a fast-growing and moderately thermophilic Streptomyces species and heterologous expression of Streptomyces antibiotic biosynthetic gene clusters , 2011, BMC Microbiology.
[204] Juan Wang,et al. An Engineered Strong Promoter for Streptomycetes , 2013, Applied and Environmental Microbiology.
[205] Jens Nielsen,et al. Antibiotic Overproduction in Streptomyces coelicolor A3(2) Mediated by Phosphofructokinase Deletion* , 2008, Journal of Biological Chemistry.
[206] Kira S. Makarova,et al. Classification and evolution of type II CRISPR-Cas systems , 2014, Nucleic acids research.
[207] Huimin Zhao,et al. Breaking the silence: new strategies for discovering novel natural products. , 2017, Current opinion in biotechnology.
[208] M. Bibb,et al. Phage P1-Derived Artificial Chromosomes Facilitate Heterologous Expression of the FK506 Gene Cluster , 2013, PloS one.
[209] B Wilkinson,et al. Engineering broader specificity into an antibiotic-producing polyketide synthase. , 1998, Science.
[210] M. Bibb,et al. Engineering Streptomyces coelicolor for heterologous expression of secondary metabolite gene clusters , 2011, Microbial biotechnology.
[211] Justin R Klesmith,et al. The Interrelationship between Promoter Strength, Gene Expression, and Growth Rate , 2014, PloS one.
[212] S. Shah,et al. Cloning, characterization and heterologous expression of a polyketide synthase and P-450 oxidase involved in the biosynthesis of the antibiotic oleandomycin. , 2000, The Journal of antibiotics.
[213] Cees M. J. Sagt,et al. Systems metabolic engineering in an industrial setting , 2013, Applied Microbiology and Biotechnology.
[214] Guoping Zhao,et al. Morphology engineering of Streptomyces coelicolor M145 by sub-inhibitory concentrations of antibiotics , 2017, Scientific Reports.
[215] Wan-Qiu Liu,et al. Cell-free synthetic biology for in vitro biosynthesis of pharmaceutical natural products , 2018, Synthetic and systems biotechnology.
[216] D. Pierson,et al. Growth of Streptomyces hygroscopicus in rotating-wall bioreactor under simulated microgravity inhibits rapamycin production , 2000, Applied Microbiology and Biotechnology.
[217] Z. Deng,et al. Streptomyces species: Ideal chassis for natural product discovery and overproduction. , 2018, Metabolic engineering.
[218] R. Süssmuth,et al. Nonribosomal Peptide Synthesis-Principles and Prospects. , 2017, Angewandte Chemie.
[219] S. Lee,et al. Metabolic engineering of antibiotic factories: new tools for antibiotic production in actinomycetes. , 2015, Trends in biotechnology.
[220] Carlos Olano,et al. Activation and identification of five clusters for secondary metabolites in Streptomyces albus J1074 , 2014, Microbial biotechnology.
[221] M. Cooper,et al. Antibiotics in the clinical pipeline at the end of 2015 , 2016, The Journal of Antibiotics.
[222] C. Thompson,et al. Construction of thiostrepton-inducible, high-copy-number expression vectors for use in Streptomyces spp. , 1995, Gene.
[223] Andriy Luzhetskyy,et al. Design, construction and characterisation of a synthetic promoter library for fine-tuned gene expression in actinomycetes. , 2013, Metabolic engineering.
[224] Sang Yup Lee,et al. Systems metabolic engineering for chemicals and materials. , 2011, Trends in biotechnology.
[225] Min Woo Kim,et al. The dynamic transcriptional and translational landscape of the model antibiotic producer Streptomyces coelicolor A3(2) , 2016, Nature Communications.
[226] Ryan A McClure,et al. In Vitro Reconstruction of Nonribosomal Peptide Biosynthesis Directly from DNA Using Cell-Free Protein Synthesis. , 2017, ACS synthetic biology.
[227] Alistair S Brown,et al. Structural, functional and evolutionary perspectives on effective re-engineering of non-ribosomal peptide synthetase assembly lines. , 2018, Natural product reports.
[228] Sang Yup Lee,et al. Metabolic Engineering of Microorganisms for the Production of Natural Compounds , 2018, Advanced Biosystems.
[229] Jay D Keasling,et al. Balancing a heterologous mevalonate pathway for improved isoprenoid production in Escherichia coli. , 2007, Metabolic engineering.
[230] Huanhuan Liu,et al. Enhancement of rapamycin production by metabolic engineering in Streptomyces hygroscopicus based on genome-scale metabolic model , 2017, Journal of Industrial Microbiology & Biotechnology.
[231] W. Wohlleben,et al. Acyltransferases as Tools for Polyketide Synthase Engineering , 2018, Antibiotics.
[232] Tong Un Chae,et al. Recent advances in systems metabolic engineering tools and strategies. , 2017, Current opinion in biotechnology.
[233] A. Keatinge-Clay,et al. The structures of type I polyketide synthases. , 2012, Natural product reports.
[234] E. Takano. Gamma-butyrolactones: Streptomyces signalling molecules regulating antibiotic production and differentiation. , 2006, Current opinion in microbiology.
[235] K. Reynolds,et al. Genes encoding acyl-CoA dehydrogenase (AcdH) homologues from Streptomyces coelicolor and Streptomyces avermitilis provide insights into the metabolism of small branched-chain fatty acids and macrolide antibiotic production. , 1999, Microbiology.
[236] Minsuk Kim,et al. Production of pikromycin using branched chain amino acid catabolism in Streptomyces venezuelae ATCC 15439 , 2018, Journal of Industrial Microbiology & Biotechnology.