Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 929
Filter
1.
Nature ; 629(8010): 165-173, 2024 May.
Article in English | MEDLINE | ID: mdl-38632398

ABSTRACT

Streptomyces are a genus of ubiquitous soil bacteria from which the majority of clinically utilized antibiotics derive1. The production of these antibacterial molecules reflects the relentless competition Streptomyces engage in with other bacteria, including other Streptomyces species1,2. Here we show that in addition to small-molecule antibiotics, Streptomyces produce and secrete antibacterial protein complexes that feature a large, degenerate repeat-containing polymorphic toxin protein. A cryo-electron microscopy structure of these particles reveals an extended stalk topped by a ringed crown comprising the toxin repeats scaffolding five lectin-tipped spokes, which led us to name them umbrella particles. Streptomyces coelicolor encodes three umbrella particles with distinct toxin and lectin composition. Notably, supernatant containing these toxins specifically and potently inhibits the growth of select Streptomyces species from among a diverse collection of bacteria screened. For one target, Streptomyces griseus, inhibition relies on a single toxin and that intoxication manifests as rapid cessation of vegetative hyphal growth. Our data show that Streptomyces umbrella particles mediate competition among vegetative mycelia of related species, a function distinct from small-molecule antibiotics, which are produced at the onset of reproductive growth and act broadly3,4. Sequence analyses suggest that this role of umbrella particles extends beyond Streptomyces, as we identified umbrella loci in nearly 1,000 species across Actinobacteria.


Subject(s)
Antibiosis , Bacterial Proteins , Bacterial Toxins , Streptomyces , Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Antibiosis/drug effects , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/pharmacology , Bacterial Proteins/ultrastructure , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Bacterial Toxins/pharmacology , Cryoelectron Microscopy , Lectins/chemistry , Lectins/genetics , Lectins/metabolism , Lectins/ultrastructure , Microbial Sensitivity Tests , Models, Molecular , Streptomyces/chemistry , Streptomyces/drug effects , Streptomyces/genetics , Streptomyces/growth & development , Streptomyces coelicolor/chemistry , Streptomyces coelicolor/genetics , Streptomyces coelicolor/metabolism , Streptomyces griseus/drug effects , Streptomyces griseus/genetics , Streptomyces griseus/growth & development , Streptomyces griseus/metabolism
2.
Nature ; 590(7846): 463-467, 2021 02.
Article in English | MEDLINE | ID: mdl-33536618

ABSTRACT

Actinobacteria produce numerous antibiotics and other specialized metabolites that have important applications in medicine and agriculture1. Diffusible hormones frequently control the production of such metabolites by binding TetR family transcriptional repressors (TFTRs), but the molecular basis for this remains unclear2. The production of methylenomycin antibiotics in Streptomyces coelicolor A3(2) is initiated by the binding of 2-alkyl-4-hydroxymethylfuran-3-carboxylic acid (AHFCA) hormones to the TFTR MmfR3. Here we report the X-ray crystal structure of an MmfR-AHFCA complex, establishing the structural basis for hormone recognition. We also elucidate the mechanism for DNA release upon hormone binding through the single-particle cryo-electron microscopy structure of an MmfR-operator complex. DNA binding and release assays with MmfR mutants and synthetic AHFCA analogues define the role of individual amino acid residues and hormone functional groups in ligand recognition and DNA release. These findings will facilitate the exploitation of actinobacterial hormones and their associated TFTRs in synthetic biology and in the discovery of new antibiotics.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Furans/metabolism , Streptomyces coelicolor/metabolism , Apoproteins/chemistry , Apoproteins/metabolism , Apoproteins/ultrastructure , Bacterial Proteins/chemistry , Bacterial Proteins/classification , Bacterial Proteins/metabolism , Bacterial Proteins/ultrastructure , Cryoelectron Microscopy , Crystallography, X-Ray , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA/ultrastructure , Furans/chemistry , Hormones/chemistry , Hormones/classification , Hormones/metabolism , Ligands , Models, Molecular , Peptides/metabolism , Repressor Proteins/chemistry , Repressor Proteins/classification , Repressor Proteins/metabolism , Repressor Proteins/ultrastructure , Signal Transduction , Streptomyces coelicolor/chemistry , Streptomyces coelicolor/genetics , Structure-Activity Relationship
3.
Nucleic Acids Res ; 52(8): 4185-4197, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38349033

ABSTRACT

Zur is a Fur-family metalloregulator that is widely used to control zinc homeostasis in bacteria. In Streptomyces coelicolor, Zur (ScZur) acts as both a repressor for zinc uptake (znuA) gene and an activator for zinc exporter (zitB) gene. Previous structural studies revealed three zinc ions specifically bound per ScZur monomer; a structural one to allow dimeric architecture and two regulatory ones for DNA-binding activity. In this study, we present evidence that Zur contains a fourth specific zinc-binding site with a key histidine residue (H36), widely conserved among actinobacteria, for regulatory function. Biochemical, genetic, and calorimetric data revealed that H36 is critical for hexameric binding of Zur to the zitB zurbox and further binding to its upstream region required for full activation. A comprehensive thermodynamic model demonstrated that the DNA-binding affinity of Zur to both znuA and zitB zurboxes is remarkably enhanced upon saturation of all three regulatory zinc sites. The model also predicts that the strong coupling between zinc binding and DNA binding equilibria of Zur drives a biphasic activation of the zitB gene in response to a wide concentration change of zinc. Similar mechanisms may be pertinent to other metalloproteins, expanding their response spectrum through binding multiple regulatory metals.


Subject(s)
Bacterial Proteins , Streptomyces coelicolor , Zinc , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Binding Sites , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/chemistry , Gene Expression Regulation, Bacterial , Histidine/metabolism , Histidine/chemistry , Protein Binding , Repressor Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/chemistry , Streptomyces coelicolor/genetics , Streptomyces coelicolor/metabolism , Zinc/metabolism
4.
Nucleic Acids Res ; 52(8): 4604-4626, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38348908

ABSTRACT

Bacteria have evolved structured RNAs that can associate with RNA polymerase (RNAP). Two of them have been known so far-6S RNA and Ms1 RNA but it is unclear if any other types of RNAs binding to RNAP exist in bacteria. To identify all RNAs interacting with RNAP and the primary σ factors, we have established and performed native RIP-seq in Bacillus subtilis, Corynebacterium glutamicum, Streptomyces coelicolor, Mycobacterium smegmatis and the pathogenic Mycobacterium tuberculosis. Besides known 6S RNAs in B. subtilis and Ms1 in M. smegmatis, we detected MTS2823, a homologue of Ms1, on RNAP in M. tuberculosis. In C. glutamicum, we discovered novel types of structured RNAs that associate with RNAP. Furthermore, we identified other species-specific RNAs including full-length mRNAs, revealing a previously unknown landscape of RNAs interacting with the bacterial transcription machinery.


Subject(s)
Bacterial Proteins , DNA-Directed RNA Polymerases , RNA, Bacterial , Sigma Factor , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Corynebacterium glutamicum/genetics , Corynebacterium glutamicum/metabolism , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/genetics , Gene Expression Regulation, Bacterial , Mycobacterium smegmatis/genetics , Mycobacterium smegmatis/metabolism , Mycobacterium smegmatis/enzymology , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Nucleic Acid Conformation , RNA, Bacterial/metabolism , RNA, Bacterial/genetics , RNA, Untranslated , Sigma Factor/metabolism , Sigma Factor/genetics , Streptomyces coelicolor/genetics , Streptomyces coelicolor/metabolism , Transcription, Genetic
5.
Proc Natl Acad Sci U S A ; 120(11): e2222045120, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36877856

ABSTRACT

The soil-dwelling filamentous bacteria, Streptomyces, is widely known for its ability to produce numerous bioactive natural products. Despite many efforts toward their overproduction and reconstitution, our limited understanding of the relationship between the host's chromosome three dimension (3D) structure and the yield of the natural products escaped notice. Here, we report the 3D chromosome organization and its dynamics of the model strain, Streptomyces coelicolor, during the different growth phases. The chromosome undergoes a dramatic global structural change from primary to secondary metabolism, while some biosynthetic gene clusters (BGCs) form special local structures when highly expressed. Strikingly, transcription levels of endogenous genes are found to be highly correlated to the local chromosomal interaction frequency as defined by the value of the frequently interacting regions (FIREs). Following the criterion, an exogenous single reporter gene and even complex BGC can achieve a higher expression after being integrated into the chosen loci, which may represent a unique strategy to activate or enhance the production of natural products based on the local chromosomal 3D organization.


Subject(s)
Biological Products , Streptomyces coelicolor , Streptomyces coelicolor/genetics , Chromosome Structures , DNA Packaging , Multigene Family/genetics
6.
J Biol Chem ; 300(1): 105507, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38029966

ABSTRACT

Cystargolides are natural products originally isolated from Kitasatospora cystarginea NRRL B16505 as inhibitors of the proteasome. They are composed of a dipeptide backbone linked to a ß-lactone warhead. Recently, we identified the cystargolide biosynthetic gene cluster, but systematic genetic analyses had not been carried out because of the lack of a heterologous expression system. Here, we report the discovery of a homologous cystargolide biosynthetic pathway in Streptomyces durhamensis NRRL-B3309 by genome mining. The gene cluster was cloned via transformation-associated recombination and heterologously expressed in Streptomyces coelicolor M512. We demonstrate that it contains all genes necessary for the production of cystargolide A and B. Single gene deletion experiments reveal that only five of the eight genes from the initially proposed gene cluster are essential for cystargolide synthesis. Additional insights into the cystargolide pathway could be obtained from in vitro assays with CysG and chemical complementation of the respective gene knockout. This could be further supported by the in vitro investigation of the CysG homolog BelI from the belactosin biosynthetic gene cluster. Thereby, we confirm that CysG and BelI catalyze a cryptic SAM-dependent transfer of a methyl group that is critical for the construction of the cystargolide and belactosin ß-lactone warheads.


Subject(s)
Dipeptides , Methyltransferases , Streptomycetaceae , Biosynthetic Pathways , Dipeptides/metabolism , Lactones/metabolism , Methyltransferases/chemistry , Methyltransferases/genetics , Methyltransferases/metabolism , Multigene Family , Streptomyces coelicolor/genetics , Streptomycetaceae/enzymology , Streptomycetaceae/genetics
7.
J Bacteriol ; 206(3): e0042823, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38353530

ABSTRACT

In Streptomyces, multiple paralogs of SsgA-like proteins (SALPs) are involved in spore formation from aerial hyphae. However, the functions of SALPs have not yet been elucidated in other actinobacterial genera. Here, we report the primary function of an SsgB ortholog (AmSsgB) in Actinoplanes missouriensis, which develops terminal sporangia on the substrate mycelia via short sporangiophores. Importantly, AmSsgB is the sole SALP in A. missouriensis. The transcription of AmssgB was upregulated during sporangium formation, consistent with our previous findings that AmssgB is a member of the AmBldD regulon. The AmssgB null mutant (ΔAmssgB) strain formed non-globose irregular structures on the substrate mycelium. Transmission electron microscopy revealed that the irregular structures contained abnormally septate hypha-like cells, without an intrasporangial matrix. These phenotypic changes were restored by complementation with AmssgB. Additionally, analysis of the heterologous expression of seven SALP-encoding genes from Streptomyces coelicolor A3(2) (ssgA-G) in the ΔAmssgB strain revealed that only ssgB could compensate for AmSsgB deficiency. This indicated that SsgB of S. coelicolor A3(2) and AmSsgB have comparable functions in A. missouriensis. In contrast to the ΔAmssgB strain, the ftsZ-disrupted strain showed a severe growth defect and produced small sporangium-like structures that swelled to some extent. These findings indicate that AmSsgB is crucial for the early stages of sporangium formation, not for spore septum formation in the late stages. We propose that AmSsgB is involved in sporangium formation by promoting the expansion of the "presporangium" structures formed on the tips of the substrate hyphae. IMPORTANCE: SsgB has been proposed as an archetypical SsgA-like protein with an evolutionarily conserved function in the morphological development of spore-forming actinomycetes. SsgB in Streptomyces coelicolor A3(2) is involved in spore septum formation. However, it is unclear whether this is the primary function of SsgBs in actinobacteria. This study demonstrated that the SsgB ortholog (AmSsgB) in Actinoplanes missouriensis is essential for sporangium expansion, which does not seem to be related to spore septum formation. However, the heterologous expression of ssgB from S. coelicolor A3(2) restored morphological abnormalities in the ΔAmssgB mutant. We propose that the primary function of SsgB is to initiate sporulation in differentiating cells (e.g., aerial hyphae in Streptomyces and "presporangium" cells in A. missouriensis) although its molecular mechanism remains unknown.


Subject(s)
Actinobacteria , Actinoplanes , Streptomyces coelicolor , Streptomyces , Sporangia/metabolism , Streptomyces/genetics , Streptomyces coelicolor/genetics , Streptomyces coelicolor/metabolism , Actinobacteria/metabolism , Bacterial Proteins/metabolism , Spores, Bacterial/genetics , Spores, Bacterial/metabolism
8.
Microb Cell Fact ; 23(1): 149, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38790014

ABSTRACT

BACKGROUND: Streptomyces is renowned for its robust biosynthetic capacity in producing medically relevant natural products. However, the majority of natural products biosynthetic gene clusters (BGCs) either yield low amounts of natural products or remain cryptic under standard laboratory conditions. Various heterologous production hosts have been engineered to address these challenges, and yet the successful activation of BGCs has still been limited. In our search for a valuable addition to the heterologous host panel, we identified the strain Streptomyces sp. A4420, which exhibited rapid initial growth and a high metabolic capacity, prompting further exploration of its potential. RESULTS: We engineered a polyketide-focused chassis strain based on Streptomyces sp. A4420 (CH strain) by deleting 9 native polyketide BGCs. The resulting metabolically simplified organism exhibited consistent sporulation and growth, surpassing the performance of most existing Streptomyces based chassis strains in standard liquid growth media. Four distinct polyketide BGCs were chosen and expressed in various heterologous hosts, including the Streptomyces sp. A4420 wild-type and CH strains, alongside Streptomyces coelicolor M1152, Streptomyces lividans TK24, Streptomyces albus J1074, and Streptomyces venezuelae NRRL B-65442. Remarkably, only the Streptomyces sp. A4420 CH strain demonstrated the capability to produce all metabolites under every condition outperforming its parental strain and other tested organisms. To enhance visualization and comparison of the tested strains, we developed a matrix-like analysis involving 15 parameters. This comprehensive analysis unequivocally illustrated the significant potential of the new strain to become a popular heterologous host. CONCLUSION: Our engineered Streptomyces sp. A4420 CH strain exhibits promising attributes for the heterologous expression of natural products with a focus on polyketides, offering an alternative choice in the arsenal of heterologous production strains. As genomics and cloning strategies progress, establishment of a diverse panel of heterologous production hosts will be crucial for expediting the discovery and production of medically relevant natural products derived from Streptomyces.


Subject(s)
Biological Products , Metabolic Engineering , Multigene Family , Polyketides , Streptomyces , Streptomyces/genetics , Streptomyces/metabolism , Biological Products/metabolism , Polyketides/metabolism , Streptomyces coelicolor/genetics , Streptomyces coelicolor/metabolism , Streptomyces lividans/genetics , Streptomyces lividans/metabolism , Biosynthetic Pathways/genetics
9.
Appl Microbiol Biotechnol ; 108(1): 306, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38656376

ABSTRACT

The Streptomyces genus comprises Gram-positive bacteria known to produce over two-thirds of the antibiotics used in medical practice. The biosynthesis of these secondary metabolites is highly regulated and influenced by a range of nutrients present in the growth medium. In Streptomyces coelicolor, glucose inhibits the production of actinorhodin (ACT) and undecylprodigiosin (RED) by a process known as carbon catabolite repression (CCR). However, the mechanism mediated by this carbon source still needs to be understood. It has been observed that glucose alters the transcriptomic profile of this actinobacteria, modifying different transcriptional regulators, including some of the one- and two-component systems (TCSs). Under glucose repression, the expression of one of these TCSs SCO6162/SCO6163 was negatively affected. We aimed to study the role of this TCS on secondary metabolite formation to define its influence in this general regulatory process and likely establish its relationship with other transcriptional regulators affecting antibiotic biosynthesis in the Streptomyces genus. In this work, in silico predictions suggested that this TCS can regulate the production of the secondary metabolites ACT and RED by transcriptional regulation and protein-protein interactions of the transcriptional factors (TFs) with other TCSs. These predictions were supported by experimental procedures such as deletion and complementation of the TFs and qPCR experiments. Our results suggest that in the presence of glucose, the TCS SCO6162/SCO6163, named GarR/GarS, is an important negative regulator of the ACT and RED production in S. coelicolor. KEY POINTS: • GarR/GarS is a TCS with domains for signal transduction and response regulation • GarR/GarS is an essential negative regulator of the ACT and RED production • GarR/GarS putatively interacts with and regulates activators of ACT and RED.


Subject(s)
Bacterial Proteins , Gene Expression Regulation, Bacterial , Streptomyces coelicolor , Anthraquinones/metabolism , Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Benzoisochromanequinones , Catabolite Repression , Glucose/metabolism , Prodigiosin/analogs & derivatives , Prodigiosin/biosynthesis , Prodigiosin/metabolism , Secondary Metabolism/genetics , Streptomyces coelicolor/metabolism , Streptomyces coelicolor/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Int J Mol Sci ; 25(6)2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38542100

ABSTRACT

The marine bacterium Streptomyces sp. HNS054 shows promise as a platform for producing natural products. Isolated from a marine sponge, HNS054 possesses several desirable traits for bioengineering: rapid growth, salt tolerance, and compatibility with genetic tools. Its genome contains 21 potential biosynthetic gene clusters, offering a rich source of natural products. We successfully engineered HNS054 to increase the production of aborycin and actinorhodin by 4.5-fold and 1.2-fold, respectively, compared to S. coelicolor M1346 counterparts. With its unique features and amenability to genetic manipulation, HNS054 emerges as a promising candidate for developing novel marine-derived drugs and other valuable compounds.


Subject(s)
Actinobacteria , Biological Products , Streptomyces coelicolor , Streptomyces , Actinobacteria/genetics , Synthetic Biology , Streptomyces/genetics , Genomics , Biological Products/pharmacology , Multigene Family , Streptomyces coelicolor/genetics
11.
J Bacteriol ; 205(7): e0015323, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37347176

ABSTRACT

Streptomyces bacteria have been studied for more than 80 years thanks to their ability to produce an incredible array of antibiotics and other specialized metabolites and their unusual fungal-like development. Their antibiotic production capabilities have ensured continual interest from both academic and industrial sectors, while their developmental life cycle has provided investigators with unique opportunities to address fundamental questions relating to bacterial multicellular growth. Much of our understanding of the biology and metabolism of these fascinating bacteria, and many of the tools we use to manipulate these organisms, have stemmed from investigations using the model species Streptomyces coelicolor and Streptomyces venezuelae. Here, we explore the pioneering work in S. coelicolor that established foundational genetic principles relating to specialized metabolism and development, alongside the genomic and cell biology developments that led to the emergence of S. venezuelae as a new model system. We highlight key discoveries that have stemmed from studies of these two systems and discuss opportunities for future investigations that leverage the power and understanding provided by S. coelicolor and S. venezuelae.


Subject(s)
Streptomyces coelicolor , Streptomyces , Anti-Bacterial Agents/metabolism , Streptomyces coelicolor/genetics , Streptomyces/metabolism , Bacterial Proteins/genetics
12.
Biochemistry ; 62(15): 2301-2313, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37449555

ABSTRACT

The class I sesquiterpene cyclase epi-isozizaene synthase from Streptomyces coelicolor (EIZS) catalyzes the transformation of linear farnesyl diphosphate (FPP) into the tricyclic hydrocarbon epi-isozizaene in the biosynthesis of albaflavenone antibiotics. The active site cavity of EIZS is largely framed by four aromatic residues - F95, F96, F198, and W203 - that form a product-shaped contour, serving as a template to chaperone conformations of the flexible substrate and multiple carbocation intermediates leading to epi-isozizaene. Remolding the active site contour by mutagenesis can redirect the cyclization cascade away from epi-isozizaene biosynthesis to generate alternative sesquiterpene products. Here, we present the biochemical and structural characterization of four EIZS mutants in which aromatic residues have been substituted with polar residues (F95S, F96H, F198S, and F198T) to generate alternative cyclization products. Most notably, F95S EIZS generates a mixture of monocyclic sesquiterpene precursors of bisabolane, a D2 diesel fuel substitute. X-ray crystal structures of the characterized mutants reveal subtle changes in the active site contour showing how each aromatic residue influences the chemistry of a different carbocation intermediate in the cyclization cascade. We advance that EIZS may serve as a robust platform for the development of designer cyclases for the generation of high-value sesquiterpene products ranging from pharmaceuticals to biofuels in synthetic biology approaches.


Subject(s)
Alkyl and Aryl Transferases , Sesquiterpenes , Streptomyces coelicolor , Terpenes/chemistry , Cyclization , Sesquiterpenes/chemistry , Streptomyces coelicolor/genetics , Monocyclic Sesquiterpenes , Alkyl and Aryl Transferases/genetics
13.
Mol Microbiol ; 117(1): 179-192, 2022 01.
Article in English | MEDLINE | ID: mdl-34687261

ABSTRACT

WhiB7/WblC is a transcriptional factor of actinomycetes conferring intrinsic resistance to multiple translation-inhibitory antibiotics. It positively autoregulates its own transcription in response to the same antibiotics. The presence of a uORF and a potential Rho-independent transcription terminator in the 5' leader region has suggested a possibility that the whiB7/wblC gene is regulated via a uORF-mediated transcription attenuation. However, experimental evidence for the molecular mechanism to explain how antibiotic stress suppresses the attenuator, if any, and induces transcription of the whiB7/wblC gene has been lacking. Here we report that the 5' leader sequences of the whiB7/wblC genes in sub-clades of actinomycetes include conserved antiterminator RNA structures. We confirmed that the putative antiterminator in the whiB7/wblC leader sequences of both Streptomyces and Mycobacterium indeed suppresses Rho-independent transcription terminator and facilitates transcription readthrough, which is required for WhiB7/WblC-mediated antibiotic resistance. The antibiotic-mediated suppression of the attenuator can be recapitulated by amino acid starvation, indicating that translational inhibition of uORF by multiple signals is a key to induce whiB7/wblC expression. Our findings of a mechanism leading to intrinsic antibiotic resistance could provide an alternative to treat drug-resistant mycobacteria.


Subject(s)
5' Untranslated Regions/genetics , Actinobacteria/genetics , Anti-Bacterial Agents/pharmacokinetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Mycobacterium/genetics , Streptomyces coelicolor/genetics , Actinobacteria/physiology , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Drug Resistance, Bacterial/genetics , Mycobacterium/physiology , Ribosomes/metabolism , Streptomyces coelicolor/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
14.
Microbiology (Reading) ; 169(7)2023 07.
Article in English | MEDLINE | ID: mdl-37418299

ABSTRACT

CutRS was the first two-component system to be identified in Streptomyces species and is highly conserved in this genus. It was reported >25 years ago that deletion of cutRS increases the production of the antibiotic actinorhodin in Streptomyces coelicolor. However, despite this early work, the function of CutRS has remained enigmatic until now. Here we show that deletion of cutRS upregulates the production of the actinorhodin biosynthetic enzymes up to 300-fold, explaining the increase in actinorhodin production. However, while ChIP-seq identified 85 CutR binding sites in S. coelicolor none of these are in the actinorhodin biosynthetic gene cluster, meaning the effect is indirect. The directly regulated CutR targets identified in this study are implicated in extracellular protein folding, including two of the four highly conserved HtrA-family foldases: HtrA3 and HtrB, and a putative VKOR enzyme, which is predicted to recycle DsbA following its catalysis of disulphide bond formation in secreted proteins. Thus, we tentatively propose a role for CutRS in sensing and responding to protein misfolding outside the cell. Since actinorhodin can oxidise cysteine residues and induce disulphide bond formation in proteins, its over production in the ∆cutRS mutant may be a response to protein misfolding on the extracellular face of the membrane.


Subject(s)
Streptomyces coelicolor , Streptomyces , Streptomyces coelicolor/genetics , Streptomyces coelicolor/metabolism , Transcription Factors/genetics , Streptomyces/metabolism , Anti-Bacterial Agents/pharmacology , Disulfides/metabolism , Gene Expression Regulation, Bacterial
15.
Biochem Biophys Res Commun ; 645: 79-87, 2023 02 19.
Article in English | MEDLINE | ID: mdl-36680940

ABSTRACT

Bacterial cytokinesis starts with the polymerization of the tubulin-like FtsZ, which forms the cell division scaffold. SepF aligns FtsZ polymers and also acts as a membrane anchor for the Z-ring. While in most bacteria cell division takes place at midcell, during sporulation of Streptomyces many septa are laid down almost simultaneously in multinucleoid aerial hyphae. The genomes of streptomycetes encode two additional SepF paralogs, SflA and SflB, which can interact with SepF. Here we show that the sporogenic aerial hyphae of sflA and sflB mutants of Streptomyces coelicolor frequently branch, a phenomenon never seen in the wild-type strain. The branching coincided with ectopic localization of DivIVA along the lateral wall of sporulating aerial hyphae. Constitutive expression of SflA and SflB largely inhibited hyphal growth, further correlating SflAB activity to that of DivIVA. SflAB localized in foci prior to and after the time of sporulation-specific cell division, while SepF co-localized with active septum synthesis. Foci of FtsZ and DivIVA frequently persisted between adjacent spores in spore chains of sflA and sflB mutants, at sites occupied by SflAB in wild-type cells. Taken together, our data show that SflA and SflB play an important role in the control of growth and cell division during Streptomyces development.


Subject(s)
Streptomyces coelicolor , Streptomyces , Streptomyces coelicolor/genetics , Streptomyces coelicolor/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Cell Division , Cytokinesis , Streptomyces/metabolism , Spores, Bacterial/genetics , Spores, Bacterial/metabolism
16.
Appl Environ Microbiol ; 89(12): e0167423, 2023 12 21.
Article in English | MEDLINE | ID: mdl-37982622

ABSTRACT

IMPORTANCE: Central metabolism plays a key role in the control of growth and antibiotic production in streptomycetes. Specifically, aminosugars act as signaling molecules that affect development and antibiotic production, via metabolic interference with the global repressor DasR. While aminosugar metabolism directly connects to other major metabolic routes such as glycolysis and cell wall synthesis, several important aspects of their metabolism are yet unresolved. Accumulation of N-acetylglucosamine 6-phosphate or glucosamine 6-phosphate is lethal to many bacteria, a yet unresolved phenomenon referred to as "aminosugar sensitivity." We made use of this concept by selecting for suppressors in genes related to glucosamine toxicity in nagB mutants, which showed that the gene pair of rok-family regulatory gene rokL6 and major facilitator superfamily transporter gene sco1448 forms a cryptic rescue mechanism. Inactivation of rokL6 resulted in the expression of sco1448, which then prevents the toxicity of amino sugar-derived metabolites in Streptomyces. The systems biology of RokL6 and its transcriptional control of sco1448 shed new light on aminosugar metabolism in streptomycetes and on the response of bacteria to aminosugar toxicity.


Subject(s)
Streptomyces coelicolor , Streptomyces , Streptomyces coelicolor/genetics , Streptomyces coelicolor/metabolism , Glucosamine/metabolism , Streptomyces/genetics , Amino Sugars/metabolism , Anti-Bacterial Agents , Genes, Regulator , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial
17.
BMC Microbiol ; 23(1): 285, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37798648

ABSTRACT

BACKGROUND: Previous studies have revealed a nitric oxide (NO) metabolic cycle in which NO, nitrate (NO3-), and nitrite (NO2-) circulate. The NO produced in this cycle serves as a signalling molecule that regulates actinorhodin (ACT) production via the DevS/DevR NO-dependent two-component system (TCS) in Streptomyces coelicolor A3(2) M145. However, the mechanisms involved in the regulation of NO signalling in S. coelicolor have not yet been elucidated. Mycothiol (MSH), a thiol molecule produced by Actinomyces, is involved in the defence mechanisms against oxidative stress. Therefore, this study focused on the correlation between intracellular NO and MSH levels. RESULTS: To investigate the interaction of MSH with endogenously produced NO, we generated an S. coelicolor A3(2) strain deficient in MSH biosynthesis. This mutant strain exhibited a decrease in low-molecular-weight S-nitrosothiols and intracellular NO levels during culture compared to those of the wild-type strain. Moreover, the mutant strain exhibited reduced activity of the DevS/DevR TCS, a regulator of NO homeostasis and ACT production, from the early stage of culture, along with a decrease in ACT production compared to those of the wild-type strain. CONCLUSIONS: This study suggests that MSH maintains intracellular NO homeostasis by forming S-nitrosomycothiol, which induces NO signalling. Finally, we propose a metabolic model in which MSH from endogenously produced NO facilitates the maintenance of both NO homeostasis and signalling in S. coelicolor A3(2) M145.


Subject(s)
Streptomyces coelicolor , Streptomyces coelicolor/genetics , Streptomyces coelicolor/metabolism , Nitric Oxide/metabolism , Cysteine/metabolism , Homeostasis , Gene Expression Regulation, Bacterial , Anthraquinones/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Anti-Bacterial Agents/pharmacology
18.
Chemistry ; 29(32): e202300775, 2023 Jun 07.
Article in English | MEDLINE | ID: mdl-36951650

ABSTRACT

2-Methylisoborneol is a widespread musty odourant that is produced by many bacteria including actinomycetes, cyanobacteria and myxobacteria. Two 2-methylisoborneol synthases (MIBS) that are phylogenetically distant to the known enzyme from Streptomyces coelicolor were found to be highly active for 2-methylisoborneol biosynthesis. Based on the enzyme structure and on an amino acid sequence alignment, the MIBS from S. coelicolor was extensively studied through site-directed mutagenesis.


Subject(s)
Streptomyces coelicolor , Amino Acid Sequence , Streptomyces coelicolor/genetics , Streptomyces coelicolor/metabolism , Camphanes/chemistry , Camphanes/metabolism , Mutagenesis, Site-Directed
19.
Biotechnol Bioeng ; 120(7): 2039-2044, 2023 07.
Article in English | MEDLINE | ID: mdl-37042226

ABSTRACT

Streptomyces are important industrial bacteria that produce pharmaceutically valuable polyketides. However, mass production on an industrial scale is limited by low productivity, which can be overcome through metabolic engineering and the synthetic biology of the host strain. Recently, the introduction of an auto-inducible expression system depending on microbial physiological state has been suggested as an important tool for the industrial-scale production of polyketides. In this study, titer improvement by enhancing the pool of CoA-derived precursors required for polyketide production was driven in a quorum sensing (QS)-dependent manner. A self-sustaining and inducer-independent regulatory system, named the QS-based metabolic engineering of precursor pool (QMP) system, was constructed, wherein the expression of genes involved in precursor biosynthesis was regulated by the QS-responsive promoter, scbAp. The QMP system was applied for neoaureothin production in a heterologous host, Streptomyces coelicolor M1152, and productivity increased by up to 4-fold. In particular, the engineered hyperproducers produced high levels of neoaureothin without adversely affecting cell growth. Overall, this study showed that self-regulated metabolic engineering mediated by QS has the potential to engineer strains for polyketide titer improvement.


Subject(s)
Polyketides , Streptomyces coelicolor , Streptomyces , Streptomyces coelicolor/genetics , Streptomyces coelicolor/metabolism , Metabolic Engineering , Quorum Sensing/genetics , Streptomyces/genetics , Polyketides/metabolism
20.
Arch Microbiol ; 206(1): 8, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38038757

ABSTRACT

The development of practices that enhance the potential of actinomycetes as major antibiotic producers is a challenge in discovering new secondary metabolites. Light, an essential external stimulus for most microorganisms, could be exploited to manipulate their physiological processes. However, the effects of monochromatic green light on the production of secondary metabolites in actinomycetes have not yet been reported. In this paper, we report a novel and simple method that uses high-intensity monochromatic green light to potentially induce the production of cryptic secondary metabolites in the model actinomycete Streptomyces coelicolor A3(2). Using actinorhodin (ACT), a blue-pigmented antibiotic, and undecylprodigiosin (RED), a red-pigmented antibiotic, as indicators, we found that irradiation with high-intensity monochromatic green light-emitting diodes promoted sporulation, significantly decreased RED production, and increased ACT production. Semi-quantitative reverse transcription-polymerase chain reaction and western blot analyses revealed, for the first time, that stimulation with green light accelerated the expression of ActII-ORF4, a pathway-specific regulator of ACT biosynthesis in S. coelicolor A3(2). This approach of stimulating secondary metabolite biosynthesis pathways in actinomycetes by irradiation with high-intensity monochromatic green light is expected to facilitate the discovery of cryptic antibiotics that are not typically produced under conventional dark culture conditions. However, the effective intensity and duration of irradiation with green light that are required to activate these metabolite pathways may vary markedly among actinomycetes.


Subject(s)
Streptomyces coelicolor , Streptomyces coelicolor/genetics , Biosynthetic Pathways , Anti-Bacterial Agents/metabolism , Anthraquinones/metabolism , Gene Expression Regulation, Bacterial , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL