Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
2.
ACS Synth Biol ; 12(10): 3114-3123, 2023 10 20.
Article in English | MEDLINE | ID: mdl-37722085

ABSTRACT

Streptomycetes have a strong ability to produce a vast array of bioactive natural products (NPs) widely used in agriculture and veterinary/human medicine. The recently developed CRISPR/Cas9-based genome editing tools have greatly facilitated strain improvement for target NP overproduction as well as novel NP discovery in Streptomyces. However, CRISPR/Cas9 shows high toxicity to the host, limiting its application in many Streptomyces strains with a low DNA transformation efficiency. In this study, we developed a low-toxicity CRISPR/Cas9D10A nickase (nCas9)-based genome editing tool in the model strain Streptomyces coelicolor M145. We showed that in the presence of both targeting sgRNA and Cas proteins, utilization of nCas9 instead of Cas9 significantly reduced the toxicity to the host and greatly enhanced cell survival. Using this tool, we achieved deletion of single genes and gene clusters with efficiencies of 87-100 and 63-87%, and simultaneous deletion of two genes or gene clusters with efficiencies of 47 and 43%, respectively. The editing efficiency of nCas9 is comparable to that of the Cas9-mediated editing tool. Finally, the nCas9-based editing tool was successfully applied for genome editing in the industrial rapamycin-producing strain Streptomyces rapamycinicus, in which CRISPR/Cas9 cannot work well. We achieved the deletion of three tested genes with an efficiency of 27.2-30%. Collectively, the CRISPR/nCas9-based editing tool offers a convenient and efficient genetic modification system for the engineering of streptomycetes, particularly those with low DNA transformation efficiency.


Subject(s)
Actinomycetales , Streptomyces , Humans , Gene Editing , CRISPR-Cas Systems/genetics , Deoxyribonuclease I/genetics , RNA, Guide, CRISPR-Cas Systems , Streptomyces/genetics , Streptomyces/metabolism , DNA , Actinomycetales/metabolism
3.
Nat Commun ; 14(1): 1827, 2023 04 01.
Article in English | MEDLINE | ID: mdl-37005419

ABSTRACT

Several groups of bacteria have complex life cycles involving cellular differentiation and multicellular structures. For example, actinobacteria of the genus Streptomyces form multicellular vegetative hyphae, aerial hyphae, and spores. However, similar life cycles have not yet been described for archaea. Here, we show that several haloarchaea of the family Halobacteriaceae display a life cycle resembling that of Streptomyces bacteria. Strain YIM 93972 (isolated from a salt marsh) undergoes cellular differentiation into mycelia and spores. Other closely related strains are also able to form mycelia, and comparative genomic analyses point to gene signatures (apparent gain or loss of certain genes) that are shared by members of this clade within the Halobacteriaceae. Genomic, transcriptomic and proteomic analyses of non-differentiating mutants suggest that a Cdc48-family ATPase might be involved in cellular differentiation in strain YIM 93972. Additionally, a gene encoding a putative oligopeptide transporter from YIM 93972 can restore the ability to form hyphae in a Streptomyces coelicolor mutant that carries a deletion in a homologous gene cluster (bldKA-bldKE), suggesting functional equivalence. We propose strain YIM 93972 as representative of a new species in a new genus within the family Halobacteriaceae, for which the name Actinoarchaeum halophilum gen. nov., sp. nov. is herewith proposed. Our demonstration of a complex life cycle in a group of haloarchaea adds a new dimension to our understanding of the biological diversity and environmental adaptation of archaea.


Subject(s)
Halobacteriaceae , Streptomyces , Hyphae/genetics , Proteomics , Phylogeny , RNA, Ribosomal, 16S/genetics , Streptomyces/genetics , Halobacteriaceae/genetics , Spores , Cell Differentiation , Sequence Analysis, DNA , China
4.
J Biol Chem ; 299(4): 104573, 2023 04.
Article in English | MEDLINE | ID: mdl-36870685

ABSTRACT

Sideromycins are a unique subset of siderophores comprising of a siderophore conjugated to an antimicrobial agent. The "Trojan horse" antibiotic albomycins are unique sideromycins consisting of a ferrichrome-type siderophore conjugated to a peptidyl nucleoside antibiotic. They exhibit potent antibacterial activities against many model bacteria and a number of clinical pathogens. Earlier studies have provided significant insight into the biosynthetic pathway of the peptidyl nucleoside moiety. We herein decipher the biosynthetic pathway of the ferrichrome-type siderophore in Streptomyces sp. ATCC 700974. Our genetic studies suggested that abmA, abmB, and abmQ are involved in the formation of the ferrichrome-type siderophore. Additionally, we performed biochemical studies to demonstrate that a flavin-dependent monooxygenase AbmB and an N-acyltransferase AbmA catalyze sequential modifications of L-ornithine to generate N5-acetyl-N5-hydroxyornithine. Three molecules of N5-acetyl-N5-hydroxyornithine are then assembled to generate the tripeptide ferrichrome through the action of a nonribosomal peptide synthetase AbmQ. Of special note, we found out that orf05026 and orf03299, two genes scattered elsewhere in the chromosome of Streptomyces sp. ATCC 700974, have functional redundancy for abmA and abmB, respectively. Interestingly, both orf05026 and orf03299 are situated within gene clusters encoding putative siderophores. In summary, this study provided new insight into the siderophore moiety of albomycin biosynthesis and shed light on the contingency of multiple siderophores in albomycin-producing Streptomyces sp. ATCC 700974.


Subject(s)
Siderophores , Streptomyces , Siderophores/metabolism , Ferrichrome/chemistry , Ferrichrome/metabolism , Streptomyces/genetics , Streptomyces/metabolism , Biosynthetic Pathways , Nucleosides/metabolism , Anti-Bacterial Agents/metabolism
5.
Appl Microbiol Biotechnol ; 106(5-6): 2147-2159, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35218390

ABSTRACT

Rapamycin is an important macrocyclic antibiotic produced by Streptomyces rapamycinicus. In the rapamycin biosynthetic gene cluster (BGC), there are up to five regulatory genes, which have been shown to play important roles in the regulation of rapamycin biosynthesis. Here, we demonstrated that the rapamycin BGC-situated LAL family regulator RapH co-ordinately regulated the biosynthesis of both rapamycin and elaiophylin. We showed that rapH overexpression not only resulted in enhanced rapamycin production but also led to increased synthesis of another type I polyketide antibiotic, elaiophylin. Consistent with this, rapH deletion resulted in decreased production of both antibiotics. Through real-time RT-PCR combined with ß-glucuronidase reporter assays, four target genes controlled by RapH, including rapL (encoding a lysine cyclodeaminase)/rapH in the rapamycin BGC and ela3 (encoding a LuxR family regulator)/ela9 (encoding a hypothetical protein) in the elaiophylin BGC, were identified. A relatively conserved signature sequence recognized by RapH, which comprises two 4-nt inverted repeats separated by 8-nt, 5'-GTT/AC-N8-GTAC-3', was defined. Taken together, our findings demonstrated that RapH was involved in co-ordinated regulation of two disparate BGCs specifying two unrelated antibiotics, rapamycin and elaiophylin. These results further expand our knowledge of the regulation of antibiotic biosynthesis in S. rapamycinicus. KEY POINTS: • The cluster-situated regulator RapH controlled the synthesis of two antibiotics. • Four promoter regions recognized by RapH were identified. • A 16-nt signature DNA sequence essential for RapH regulation was defined.


Subject(s)
Sirolimus , Streptomyces , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Macrolides , Multigene Family , Sirolimus/metabolism , Streptomyces/genetics , Streptomyces/metabolism
6.
Appl Microbiol Biotechnol ; 105(14-15): 5905-5914, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34287659

ABSTRACT

In the model actinomycete strain, Streptomyces coelicolor, an orphan histidine kinase (HK) named OhkA (encoded by SCO1596), which belongs to bacterial two-component regulatory systems (TCSs), has been identified as being involved in the regulation of both antibiotic biosynthesis and morphological development. However, its cognate response regulator (RR) remains unknown due to its isolated genetic location on the genome, which impedes the elucidation of the mechanism underlying OhkA-mediated regulation. Here, we identified the orphan RR OrrA (encoded by SCO3008) as the cognate RR of OhkA according to mutant phenotypic changes, transcriptomics analysis, and bacterial two-hybrid experiment. Considering that the partner RR of the orphan HK is also orphan, a library of mutants with in-frame individual deletion of these functionally unknown orphan RR-encoding genes were generated. Through phenotypic analysis, it was found that the ∆orrA mutant exhibited similar phenotypic changes as that of the ∆ohkA mutant, showing increased production of actinorhodin (ACT) and undecylprodigiosin (RED), and pink colony surface. Further transcriptomics analysis showed these two mutants exhibited highly similar transcriptomics profiles. Finally, the direct interaction between OhkA and OrrA was revealed by bacterial two-hybrid system. The identification of the partner RR of OhkA lays a good foundation for an in-depth elucidation of the molecular mechanism underlying OhkA-mediated regulation of development and antibiotic biosynthesis in Streptomyces. KEY POINTS: • OrrA was identified as the partner RR of the orphan histidine kinase OhkA. • The ∆orrA and ∆ohkA mutants showed similar phenotype and transcriptomic profiling. • Specific interaction of OrrA and OhkA was revealed by bacterial two-hybrid system.


Subject(s)
Streptomyces coelicolor , Streptomyces , Anti-Bacterial Agents , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Histidine Kinase/genetics , Histidine Kinase/metabolism , Secondary Metabolism/genetics , Streptomyces/genetics , Streptomyces coelicolor/genetics , Streptomyces coelicolor/metabolism
7.
Front Bioeng Biotechnol ; 9: 692797, 2021.
Article in English | MEDLINE | ID: mdl-34327194

ABSTRACT

Microbial natural products (NPs) are a major source of pharmacological agents. Most NPs are synthesized from specific biosynthetic gene clusters (BGCs). With the rapid increase of sequenced microbial genomes, large numbers of NP BGCs have been discovered, regarded as a treasure trove of novel bioactive compounds. However, many NP BGCs are silent in native hosts under laboratory conditions. In order to explore their therapeutic potential, a main route is to activate these silent NP BGCs in heterologous hosts. To this end, the first step is to accurately and efficiently capture these BGCs. In the past decades, a large number of effective technologies for cloning NP BGCs have been established, which has greatly promoted drug discovery research. Herein, we describe recent advances in strategies for BGC cloning, with a focus on the preparation of high-molecular-weight DNA fragment, selection and optimization of vectors used for carrying large-size DNA, and methods for assembling targeted DNA fragment and appropriate vector. The future direction into novel, universal, and high-efficiency methods for cloning NP BGCs is also prospected.

8.
Microbiol Res ; 233: 126411, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31981905

ABSTRACT

In Streptomyces pristinaespiralis, the orphan histidine kinase (HK) PdtaS-p (encoded by SSDG_02492), which belongs to proteins of two-component systems (TCSs), plays an important role in both morphological differentiation and antibiotic biosynthesis. Owing to the isolated genetic organization of pdtaS-p, it is a challenge to identify its cognate response regulator (RR) and hampers the efforts to elucidate the regulation mechanism of PdtaS-p. In this study, based on bioinformatics analysis, we identify the cognate RR PdtaR-p (encoded by SSDG_04087) of PdtaS-p by phenotype similarity of gene deletion mutants as well as in vitro phosphor-transfer assay. We show that the mutants (ΔpdtaR-p and ΔpdtaS-p) exhibit almost the same phenotypical changes, showing a bald phenotype on MS agar and reduced pristinamycin biosynthesis. Further phosphor-transfer assay indicates that the phosphoryl group of HK PdtaS-p can be specifically transferred to RR PdtaR-p. Compared with the majority of RRs that harbor DNA-binding domains, PdtaR-p contains a putative ANTAR RNA-binding domain involved in controlling gene expression at the post-transcription level. Finally, we demonstrate that their ortholog from the model strain Streptomyces coelicolor, PdtaS-c/PdtaR-c, also regulates both morphological differentiation and antibiotics biosynthesis, suggesting that PdtaS-p/PdtaR-p-mediated molecular regulation may be conserved in the genus Streptomyces. To our knowledge, this is the first report describing the functional identification of ANTAR RNA-binding regulators in Streptomyces.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Bacterial Proteins/metabolism , Histidine Kinase/metabolism , Streptomyces/enzymology , Bacterial Proteins/genetics , Computational Biology , Gene Deletion , Gene Expression Regulation, Bacterial , Histidine Kinase/genetics , Mutation , Phenotype , RNA Recognition Motif Proteins/genetics , Streptomyces/genetics
9.
Sci China Life Sci ; 63(7): 1053-1062, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31872379

ABSTRACT

CRISPR/Cas-mediated genome editing has greatly facilitated the study of gene function in Streptomyces. However, it could not be efficiently employed in streptomycetes with low homologous recombination (HR) ability. Here, a deaminase-assisted base editor dCas9-CDA-ULstr was developed in Streptomyces, which comprises the nuclease-deficient Cas9 (dCas9), the cytidine deaminase from Petromyzon marinus (PmCDA1), the uracil DNA glycosylase inhibitor (UGI) and the protein degradation tag (LVA tag). Using dCas9-CDA-ULstr, we achieved single-, double- and triple-point mutations (cytosine-to-thymine substitutions) at target sites in Streptomyces coelicolor with efficiency up to 100%, 60% and 20%, respectively. This base editor was also demonstrated to be highly efficient for base editing in the industrial strain, Streptomyces rapamycinicus, which produces the immunosuppressive agent rapamycin. Compared with base editors derived from the cytidine deaminase rAPOBEC1, the PmCDA1-assisted base editor dCas9-CDA-ULstr could edit cytosines preceded by guanosines with high efficiency, which is a great advantage for editing Streptomyces genomes (with high GC content). Collectively, the base editor dCas9-CDA-ULstr could be employed for efficient multiplex genome editing in Streptomyces. Since the dCas9-CDA-ULstr-based genome editing is independent of HR-mediated DNA repair, we believe this technology will greatly facilitate functional genome research and metabolic engineering in Streptomyces strains with weak HR ability.


Subject(s)
CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems/genetics , Cytidine Deaminase/genetics , Gene Editing/methods , Recombinant Proteins/genetics , Streptomyces/genetics , Gene Expression Regulation, Bacterial , Genetic Engineering , Genome, Bacterial/genetics , Guanosine/metabolism , Immunosuppressive Agents/metabolism , Point Mutation/genetics , Promoter Regions, Genetic , Sirolimus/metabolism
10.
Sci China Life Sci ; 62(11): 1492-1505, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31228045

ABSTRACT

Cyclic dimeric GMP (c-di-GMP) has emerged as the nucleotide second messenger regulating both development and antibiotic production in high-GC, Gram-positive streptomycetes. Here, a diguanylate cyclase (DGC), CdgD, encoded by SCO5345 from the model strain Streptomyces coelicolor, was functionally identified and characterized to be involved in c-di-GMP synthesis through genetic and biochemical analysis. cdgD overexpression resulted in significantly reduced production of actinorhodin and undecylprodigiosin, as well as completely blocked sporulation or aerial mycelium formation on two different solid media. In the cdgD-overexpression strain, intracellular c-di-GMP levels were 13-27-fold higher than those in the wild-type strain. In vitro enzymatic assay demonstrated that CdgD acts as a DGC, which could efficiently catalyze the synthesis of c-di-GMP from two GTP molecules. Heterologous overproduction of cdgD in two industrial Streptomyces strains could similarly impair developmental transitions as well as antibiotic biosynthesis. Collectively, our results combined with previously reported data clearly demonstrated that c-di-GMP-mediated signalling pathway plays a central and universal role in the life cycle as well as secondary metabolism in streptomycetes.


Subject(s)
Anti-Bacterial Agents/metabolism , Bacterial Proteins/genetics , Cyclic GMP/genetics , Escherichia coli Proteins/genetics , Phosphorus-Oxygen Lyases/genetics , Streptomyces coelicolor/metabolism , Anthraquinones/metabolism , Anti-Bacterial Agents/biosynthesis , Bacterial Proteins/metabolism , CRISPR-Cas Systems , Cyclic GMP/metabolism , DNA-Binding Proteins , Escherichia coli Proteins/metabolism , Fermentation , Gene Editing , Gene Expression Regulation, Bacterial , Mutation , Phosphorus-Oxygen Lyases/metabolism , Prodigiosin/analogs & derivatives , Prodigiosin/metabolism , Transfection
11.
Metab Eng ; 52: 153-167, 2019 03.
Article in English | MEDLINE | ID: mdl-30529239

ABSTRACT

Chromosomal integration of genes and pathways is of particular importance for large-scale and long-term fermentation in industrial biotechnology. However, stable, multi-copy integration of long DNA segments (e.g., large gene clusters) remains challenging. Here, we describe a plug-and-play toolkit that allows for high-efficiency, single-step, multi-locus integration of natural product (NP) biosynthetic gene clusters (BGCs) in actinomycetes, based on the innovative concept of "multiple integrases-multiple attB sites". This toolkit consists of 27 synthetic modular plasmids, which contain single- or multi-integration modules (from two to four) derived from five orthogonal site-specific recombination (SSR) systems. The multi-integration modules can be readily ligated into plasmids containing large BGCs by Gibson assembly, which can be simultaneously inserted into multiple native attB sites in a single step. We demonstrated the applicability of this toolkit by performing stabilized amplification of acetyl-CoA carboxylase genes to facilitate actinorhodin biosynthesis in Streptomyces coelicolor. Furthermore, using this toolkit, we achieved a 185.6% increase in 5-oxomilbemycin titers (from 2.23 to 6.37 g/L) in Streptomyces hygroscopicus via the multi-locus integration of the entire 5-oxomilbemycin BGC (72 kb) (up to four copies). Compared with previously reported methods, the advanced multiplex site-specific genome engineering (aMSGE) method does not require the introduction of any modifications into host genomes before the amplification of target genes or BGCs, which will drastically simplify and accelerate efforts to improve NP production. Considering that SSR systems are widely distributed in a variety of industrial microbes, this novel technique also promises to be a valuable tool for the enhanced biosynthesis of other high-value bioproducts.


Subject(s)
Actinobacteria/genetics , Actinobacteria/metabolism , Metabolic Engineering/methods , Recombinases/genetics , Genetic Vectors , Metabolic Networks and Pathways/genetics , Multigene Family/genetics , Plasmids/genetics , Recombination, Genetic , Streptomyces/genetics , Streptomyces/metabolism
12.
Appl Environ Microbiol ; 84(18)2018 09 15.
Article in English | MEDLINE | ID: mdl-29980561

ABSTRACT

Streptomyces has a strong capability for producing a large number of bioactive natural products and remains invaluable as a source for the discovery of novel drug leads. Although the Streptococcus pyogenes CRISPR-Cas9-assisted genome editing tool has been developed for rapid genetic engineering in Streptomyces, it has a number of limitations, including the toxicity of SpCas9 expression in some important industrial Streptomyces strains and the need for complex expression constructs when targeting multiple genomic loci. To address these problems, in this study, we developed a high-efficiency CRISPR-Cpf1 system (from Francisella novicida) for multiplex genome editing and transcriptional repression in Streptomyces Using an all-in-one editing plasmid with homology-directed repair (HDR), our CRISPR-Cpf1 system precisely deletes single or double genes at efficiencies of 75 to 95% in Streptomyces coelicolor When no templates for HDR are present, random-sized DNA deletions are achieved by FnCpf1-induced double-strand break (DSB) repair by a reconstituted nonhomologous end joining (NHEJ) pathway. Furthermore, a DNase-deactivated Cpf1 (ddCpf1)-based integrative CRISPRi system is developed for robust, multiplex gene repression using a single customized crRNA array. Finally, we demonstrate that FnCpf1 and SpCas9 exhibit different suitability in tested industrial Streptomyces species and show that FnCpf1 can efficiently promote HDR-mediated gene deletion in the 5-oxomilbemycin-producing strain Streptomyces hygroscopicus SIPI-KF, in which SpCas9 does not work well. Collectively, FnCpf1 is a powerful and indispensable addition to the Streptomyces CRISPR toolbox.IMPORTANCE Rapid, efficient genetic engineering of Streptomyces strains is critical for genome mining of novel natural products (NPs) as well as strain improvement. Here, a novel and high-efficiency Streptomyces genome editing tool is established based on the FnCRISPR-Cpf1 system, which is an attractive and powerful alternative to the S. pyogenes CRISPR-Cas9 system due to its unique features. When combined with HDR or NHEJ, FnCpf1 enables the creation of gene(s) deletion with high efficiency. Furthermore, a ddCpf1-based integrative CRISPRi platform is established for simple, multiplex transcriptional repression. Of importance, FnCpf1-based genome editing proves to be a highly efficient tool for genetic modification of some important industrial Streptomyces strains (e.g., S. hygroscopicus SIPI-KF) that cannot utilize the SpCRISPR-Cas9 system. We expect the CRISPR-Cpf1-assisted genome editing tool to accelerate discovery and development of pharmaceutically active NPs in Streptomyces as well as other actinomycetes.


Subject(s)
Bacterial Proteins/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Endonucleases/metabolism , Gene Editing/methods , Genome, Bacterial , Streptomyces/genetics , DNA End-Joining Repair , Francisella tularensis/enzymology , Genetic Engineering , Streptomyces coelicolor/genetics , Transcription, Genetic
13.
Biotechnol J ; 13(9): e1800121, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29862648

ABSTRACT

Streptomycetes are Gram-positive bacteria with the capacity to produce copious bioactive secondary metabolites, which are the main source of medically and industrially relevant drugs. However, genetic manipulation of Streptomyces strains is much more difficult than other model microorganisms like Escherichia coli and Saccharomyces cerevisiae. Recently, CRISPR/Cas9 or dCas9-mediated genetic manipulation tools have been developed and facilitated Streptomyces genome editing. However, till now, CRISPR/dCas9-based interference system (CRISPRi) is only designed to repress single gene expression. Herein, the authors developed a novel CRISPRi system for multiplex gene repression in the model strain Streptomyces coelicolor. In this system, the integrative plasmid pSET152 is used as the backbone for the expression of the dCas9/sgRNA complex and both dCas9 and sgRNAs are designed to be under the control of constitutive promoters. Using the integrative CRISPRi system, the authors achieved efficient repression of multiple genes simultaneously; the mRNA levels of four targets are reduced to 2-32% of the control. Furthermore, it is successfully employed for functional gene screening, and an orphan response regulator (RR) (encoded by SCO2013) containing an RNA-binding ANTAR domain is identified being involved in bacterial growth. Collectively, this integrative CRISPRi system is very effective for multiplex gene repression in S. coelicolor, which could be extended to other Streptomyces strains for functional gene screening as well as for metabolic engineering.


Subject(s)
Down-Regulation , Gene Editing/methods , Streptomyces coelicolor/growth & development , Bacterial Proteins/genetics , CRISPR-Cas Systems , Gene Expression Regulation, Bacterial , Metabolic Engineering , Promoter Regions, Genetic , Streptomyces coelicolor/genetics
14.
Front Microbiol ; 9: 264, 2018.
Article in English | MEDLINE | ID: mdl-29515546

ABSTRACT

In Streptomyces coelicolor, amtB transcription is promptly regulated by the global nitrogen regulator GlnR. Although the GlnR binding cis-element has been characterized in amtB promoter, consisting of three GlnR boxes of a3-b3, a1-b1, and a2-b2, its role in GlnR-mediated transcriptional regulation remains unclear. Here, we showed that GlnR had different binding affinity against each pair of GlnR binding sites in amtB promoter (i.e., a3-b3, a1-b1, and a2-b2 sites), and GlnR was able to bind a3-b3 and a1-b1, respectively, but not a2-b2 alone. Consistently, a2 was not a typical GlnR binding site and further experiments showed that a2 was non-essential for GlnR-mediated binding in vitro and transcriptional regulation in vivo. To uncover the physiological role of the three GlnR boxes, we then mutated the wild-type amtB promoter to a typical GlnR-binding motif containing two GlnR boxes (a3-b3-a2-b2), and found although the transcription of the mutated promoter could still be activated by GlnR, its increasing rate was less than that of the wild-type. Based on these findings, one could conclude that the three GlnR boxes assisted GlnR in more promptly activating amtB transcription in response to nitrogen limitation, facilitating bacterial growth under nitrogen stresses.

15.
Synth Syst Biotechnol ; 2(2): 130-136, 2017 Jun.
Article in English | MEDLINE | ID: mdl-29062970

ABSTRACT

Pristinamycin, produced by Streptomyces pristinaespiralis, which is a streptogramin-like antibiotic consisting of two chemically unrelated components: pristinamycin I (PI) and pristinamycin II (PII), shows potent activity against many antibiotic-resistant pathogens. However, so far pristinamycin production titers are still quite low, particularly those of PI. In this study, we constructed a PI single component producing strain by deleting the PII biosynthetic genes (snaE1 and snaE2). Then, two metabolic engineering approaches, including deletion of the repressor gene papR3 and chromosomal integration of an extra copy of the PI biosynthetic gene cluster (BGC), were employed to improve PI production. The final engineered strain ΔPIIΔpapR3/PI produced a maximum PI level of 132 mg/L, with an approximately 2.4-fold higher than that of the parental strain S. pristinaespiralis HCCB10218. Considering that the PI biosynthetic genes are clustered in two main regions in the 210 kb "supercluster" containing the PI and PII biosynthetic genes as well as a cryptic polyketide BGC, these two regions were cloned separately and then were successfully assembled into the PI BGC by the transformation-associated recombination (TAR) system. Collectively, the metabolic engineering approaches employed is very efficient for strain improvement in order to enhance PI titer.

16.
Sci China Life Sci ; 60(9): 948-957, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28785950

ABSTRACT

Bacterial prodigiosins are red-colored secondary metabolites with multiple activities, such as anticancer, antimalarial and immunosuppressive, which hold great potential for medical applications. In this study, dramatically enhanced prodigiosins (RED) production in Streptomyces coelicolor was achieved by combinatorial metabolic engineering, including inactivation of the repressor gene ohkA, deletion of the actinorhodin (ACT) and calcium-dependent antibiotic (CDA) biosynthetic gene clusters (BGCs) and multi-copy chromosomal integration of the RED BGC. The results showed that ohkA deletion led to a 1-fold increase of RED production over the wild-type strain M145. Then, the ACT and CDA BGCs were deleted successively based on the ΔohkA mutant (SBJ101). To achieve multi-copy RED BGC integration, artificial ΦC31 attB site(s) were inserted simultaneously at the position where the ACT and CDA BGCs were deleted. The resulting strains SBJ102 (with a single deletion of the ACT BGC and insertion of one artificial attB site) and SBJ103 (with the deletion of both BGCs and insertion of two artificial attB sites) produced 1.9- and 6-fold higher RED titers than M145, respectively. Finally, the entire RED BGC was introduced into mutants from SBJ101 to SBJ103, generating three mutants (from SBJ104 to SBJ106) with chromosomal integration of one to three copies of the RED BGC. The highest RED yield was from SBJ106, which produced a maximum level of 96.8 mg g-1 cell dry weight, showing a 12-fold increase relative to M145. Collectively, the metabolic engineering strategies employed in this study are very efficient for the construction of high prodigiosin-producing strains.


Subject(s)
Industrial Microbiology/methods , Metabolic Engineering , Multigene Family/genetics , Prodigiosin/biosynthesis , Streptomyces coelicolor/genetics , Streptomyces coelicolor/metabolism , Attachment Sites, Microbiological/genetics , Biosynthetic Pathways/genetics , Gene Deletion , Gene Expression Regulation, Bacterial/genetics , Genetic Enhancement , Genome, Bacterial/genetics , Metabolic Networks and Pathways/genetics
17.
Metab Eng ; 40: 80-92, 2017 03.
Article in English | MEDLINE | ID: mdl-28088540

ABSTRACT

Actinomycetes produce a large variety of pharmaceutically active compounds, yet production titers often require to be improved for discovery, development and large-scale manufacturing. Here, we describe a new technique, multiplexed site-specific genome engineering (MSGE) via the 'one integrase-multiple attB sites' concept, for the stable integration of secondary metabolite biosynthetic gene clusters (BGCs). Using MSGE, we achieved five-copy chromosomal integration of the pristinamycin II (PII) BGC in Streptomyces pristinaespiralis, resulting in the highest reported PII titers in flask and batch fermentations (2.2 and 2g/L, respectively). Furthermore, MSGE was successfully extended to develop a panel of powerful Streptomyces coelicolor heterologous hosts, in which up to four copies of the BGCs for chloramphenicol or anti-tumour compound YM-216391 were efficiently integrated in a single step, leading to significantly elevated productivity (2-23 times). Our multiplexed approach holds great potential for robust genome engineering of industrial actinomycetes and novel drug discovery by genome mining.


Subject(s)
Chloramphenicol/biosynthesis , Genetic Enhancement/methods , Genome, Bacterial/genetics , Multigene Family/genetics , Peptides, Cyclic/biosynthesis , Secondary Metabolism/genetics , Streptomyces/physiology , Biosynthetic Pathways/genetics , Chloramphenicol/isolation & purification , Metabolic Engineering/methods , Metabolic Networks and Pathways/genetics , Oxazoles/isolation & purification , Peptides, Cyclic/genetics , Peptides, Cyclic/isolation & purification , Up-Regulation/genetics
18.
J Biol Chem ; 291(51): 26443-26454, 2016 Dec 16.
Article in English | MEDLINE | ID: mdl-27875313

ABSTRACT

GlnR, an OmpR-like orphan two-component system response regulator, is a master regulator of nitrogen metabolism in the genus Streptomyces In this work, evidence that GlnR is also directly involved in the regulation of antibiotic biosynthesis is provided. In the model strain Streptomyces coelicolor M145, an in-frame deletion of glnR resulted in markedly increased actinorhodin (ACT) production but reduced undecylprodigiosin (RED) biosynthesis when exposed to R2YE culture medium. Transcriptional analysis coupled with DNA binding studies revealed that GlnR represses ACT but activates RED production directly via the pathway-specific activator genes actII-ORF4 and redZ, respectively. The precise GlnR-binding sites upstream of these two target genes were defined. In addition, the direct involvement of GlnR in antibiotic biosynthesis was further identified in Streptomyces avermitilis, which produces the important anthelmintic agent avermectin. We found that S. avermitilis GlnR (GlnRsav) could stimulate avermectin but repress oligomycin production directly through the respective pathway-specific activator genes, aveR and olmRI/RII To the best of our knowledge, this report describes the first experimental evidence demonstrating that GlnR regulates antibiotic biosynthesis directly through pathway-specific regulators in Streptomyces Our results suggest that GlnR-mediated regulation of antibiotic biosynthesis is likely to be universal in streptomycetes. These findings also indicate that GlnR is not only a master nitrogen regulator but also an important controller of secondary metabolism, which may help to balance nitrogen metabolism and antibiotic biosynthesis in streptomycetes.


Subject(s)
Bacterial Proteins/metabolism , Ivermectin/analogs & derivatives , Streptomyces/metabolism , Trans-Activators/metabolism , Bacterial Proteins/genetics , Ivermectin/metabolism , Streptomyces/genetics , Trans-Activators/genetics
19.
FEMS Microbiol Lett ; 363(15)2016 08.
Article in English | MEDLINE | ID: mdl-27313101

ABSTRACT

We previously demonstrated that in Streptomyces coelicolor two-component system AfsQ1/Q2 activates the production of the yellow-colored coelimycin P2 (also named as yCPK) on glutamate-supplemented minimal medium, and the response regulator AfsQ1 could specifically bind to the intergenic region between two structural genes, cpkA and cpkD Here, a more in-depth investigation was performed to elucidate the mechanism underlying the role of AfsQ1/Q2 in regulating coelimycin P2 biosynthesis. Deletion of afsQ1/Q2 resulted in markedly decreased expression of the whole coelimycin P2 biosynthetic gene cluster. Electrophoretic mobility shift assays revealed that AfsQ1 bound only to the target site identified previously, but not to any other promoters in the gene cluster. Mutations of AfsQ1-binding motif only resulted in drastically reduced transcription of the cpkA/B/C operon (encoding three type I polyketide synthases) and intriguingly, led to enhanced expression of some coelimcyin P2 genes, particularly accA1 and scF These results suggested the direct role of AfsQ1/Q2 in regulating coelimycin production, which is directly mediated by the structural genes, but not the cluster-situated regulatory genes, and also implied that other unknown mechanisms may be involved in AfsQ1/Q2-mediated regulation of coelimycin P2 biosynthesis.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Streptomyces coelicolor/genetics , Streptomyces coelicolor/metabolism , Electrophoretic Mobility Shift Assay , Genes, Regulator , Operon , Polyketide Synthases/genetics , Promoter Regions, Genetic
20.
J Biotechnol ; 214: 45-6, 2015 Nov 20.
Article in English | MEDLINE | ID: mdl-26376468

ABSTRACT

Streptomyces pristinaespiralis produces the streptogramin-like antibiotic pristinamycin, which is a mixture of two structurally different components: pristinamycin I (PI) and pristinamycin II (PII). Herein, we report the complete genome sequence of a high pristinamycin-producing strain HCCB10218 (8.5 Mb) obtained by using PacBio RSII combined with Illumina HiSeq 2500 sequencing system. The genome sequence presented here provides clues for the mechanism underlying the higher pristinamycin production of HCCB10218.


Subject(s)
Genome, Bacterial/genetics , Pristinamycin/metabolism , Streptomyces/genetics , Streptomyces/metabolism , DNA, Bacterial/analysis , DNA, Bacterial/genetics , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...