Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Microbiol Spectr ; 12(7): e0042324, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38864648

RESUMO

Clorobiocin is a well-known, highly effective inhibitor of DNA gyrase belonging to the aminocoumarin antibiotics. To identify potentially novel derivatives of this natural product, we conducted an untargeted investigation of clorobiocin biosynthesis in the known producer Streptomyces roseochromogenes DS 12.976 using LC-MSE, molecular networking, and analysis of fragmentation spectra. Previously undescribed clorobiocin derivatives uncovered in this study include bromobiocin, a variant halogenated with bromine instead of chlorine, hydroxylated clorobiocin, carrying an additional hydroxyl group on its 5-methyl-pyrrole 2-carboxyl moiety, and two other derivatives with modifications on their 3-dimethylallyl 4-hydroxybenzoate moieties. Furthermore, we identified several compounds not previously considered clorobiocin pathway products, which provide new insights into the clorobiocin biosynthetic pathway. By supplementing the medium with different concentrations of potassium bromide, we confirmed that the clorobiocin halogenase can utilize bromine instead of chlorine. The reaction, however, is impeded such that non-halogenated clorobiocin derivatives accumulate. Preliminary assays indicate that the antibacterial activity of bromobioin against Bacillus subtilis and efflux-impaired Escherichia coli matches that of clorobiocin. Our findings emphasize that yet unexplored compounds can be discovered from established strains and biosynthetic gene clusters by means of metabolomics analysis and highlight the utility of LC-MSE-based methods to contribute to unraveling natural product biosynthetic pathways. IMPORTANCE: The aminocoumarin clorobiocin is a well-known gyrase inhibitor produced by the gram-positive bacterium Streptomyces roseochromogenes DS 12.976. To gain a deeper understanding of the biosynthetic pathway of this complex composite of three chemically distinct entities and the product spectrum, we chose a metabolite-centric approach. Employing high-resolution LC-MSE analysis, we investigated the pathway products in extracted culture supernatants of the natural producer. Novel pathway products were identified that expand our understanding of three aspects of the biosynthetic pathway, namely the modification of the noviose, transfer and methylation of the pyrrole 2-carboxyl moiety, and halogenation. For the first time, brominated products were detected. Their levels and the levels of non-halogenated products increased in medium supplemented with KBr. Based on the presented data, we propose that the enzyme promiscuity contributes to a broad product spectrum.


Assuntos
Antibacterianos , Vias Biossintéticas , Metabolômica , Novobiocina , Streptomyces , Streptomyces/metabolismo , Streptomyces/genética , Antibacterianos/farmacologia , Antibacterianos/biossíntese , Antibacterianos/metabolismo , Antibacterianos/química , Novobiocina/análogos & derivados , Novobiocina/biossíntese , Novobiocina/farmacologia , Novobiocina/metabolismo , Cromatografia Líquida
2.
PLoS One ; 12(3): e0174665, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28355308

RESUMO

Acceptor substrate specificity of Streptomyces roseochromogenes prenyltransferase SrCloQ was investigated using different non-genuine phenolic compounds. RP-UHPLC-UV-MSn was used for the tentative annotation and quantification of the prenylated products. Flavonoids, isoflavonoids and stilbenoids with different types of substitution were prenylated by SrCloQ, although with less efficiency than the genuine substrate 4-hydroxyphenylpyruvate. The isoflavan equol, followed by the flavone 7,4'-dihydroxyflavone, were the best non-genuine acceptor substrates. B-ring C-prenylation was in general preferred over A-ring C-prenylation (ratio 5:1). Docking studies of non-genuine acceptor substrates with the B-ring oriented towards the donor substrate dimethylallyl pyrophosphate, showed that the carbonyl group of the C-ring was able to make stabilizing interactions with the residue Arg160, which might determine the preference observed for B-ring prenylation. No reaction products were formed when the acceptor substrate had no phenolic hydroxyl groups. This preference can be explained by the essential hydrogen bond needed between a phenolic hydroxyl group and the residue Glu281. Acceptor substrates with an additional hydroxyl group at the C3' position (B-ring), were mainly O3'-prenylated (> 80% of the reaction products). This can be explained by the proximity of the C3' hydroxyl group to the donor substrate at the catalytic site. Flavones were preferred over isoflavones by SrCloQ. Docking studies suggested that the orientation of the B-ring and of the phenolic hydroxyl group at position C7 (A-ring) of flavones towards the residue Tyr233 plays an important role in this observed preference. Finally, the insights obtained on acceptor substrate specificity and regioselectivity for SrCloQ were extended to other prenyltransferases from the CloQ/NhpB family.


Assuntos
Proteínas de Bactérias/metabolismo , Dimetilaliltranstransferase/metabolismo , Flavonoides/metabolismo , Isoflavonas/metabolismo , Streptomyces/enzimologia , Proteínas de Bactérias/química , Domínio Catalítico , Dimetilaliltranstransferase/química , Equol/química , Equol/metabolismo , Flavonoides/química , Ligação de Hidrogênio , Isoflavonas/química , Cinética , Simulação de Acoplamento Molecular , Estrutura Molecular , Novobiocina/análogos & derivados , Novobiocina/biossíntese , Novobiocina/química , Fenóis/química , Fenóis/metabolismo , Ácidos Fenilpirúvicos/química , Ácidos Fenilpirúvicos/metabolismo , Prenilação , Ligação Proteica , Estrutura Terciária de Proteína , Estilbenos/química , Estilbenos/metabolismo , Streptomyces/metabolismo , Especificidade por Substrato
3.
ACS Chem Biol ; 12(2): 374-379, 2017 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-28068060

RESUMO

C-methylation of aromatic small molecules by C-methyltransferases (C-MTs) is an important biological transformation that involves C-C bond formation using S-adenosyl-l-methionine (SAM) as the methyl donor. Here, two advances in the mechanistic understanding of C-methylation of the 8-position of coumarin substrates catalyzed by the C-MT NovO from Streptomyces spheroides are described. First, a crystal structure of NovO reveals the Arg116-Asn117 and His120-Arg121 motifs are essential for coumarin substrate binding. Second, the active-site His120 is responsible for deprotonation of the phenolic 7-hydroxyl group on the coumarin substrate, activating the rate-determining methyl transfer step from SAM. This work expands our mechanistic knowledge of C-MTs, which could be used in the downstream development of engineered biocatalysts for small molecule C-alkylations.


Assuntos
Cumarínicos/metabolismo , Catálise , Cristalografia por Raios X , Metilação , Metiltransferases/metabolismo , Estrutura Molecular , Novobiocina/biossíntese , Novobiocina/química , Streptomyces/metabolismo , Especificidade por Substrato
4.
Appl Microbiol Biotechnol ; 100(10): 4495-509, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26795961

RESUMO

Understanding the regulation of a heterologously expressed gene cluster in a host organism is crucial for activation of silent gene clusters or overproduction of the corresponding natural product. In this study, Streptomyces coelicolor M512(nov-BG1) containing the novobiocin biosynthetic gene cluster from Streptomyces niveus NCIMB 11891 was chosen as a model. An improved DNA affinity capturing assay (DACA), combined with semi-quantitative mass spectrometry, was used to identify proteins binding to the promoter regions of the novobiocin gene cluster. Altogether, 2475 proteins were identified in DACA studies with the promoter regions of the pathway-specific regulators novE (PnovE) and novG (PnovG), of the biosynthetic genes novH-W (PnovH) and of the vegetative σ-factor hrdB (PhrdB) as a negative control. A restrictive classification for specific binding reduced this number to 17 proteins. Twelve of them were captured by PnovH, among them, NovG, two were captured by PnovE, and three by PnovG. Unexpectedly some well-known regulatory proteins, such as the global regulators NdgR, AdpA, SlbR, and WhiA were captured in similar intensities by all four tested promoter regions. Of the 17 promoter-specific proteins, three were studied in more detail by deletion mutagenesis and by overexpression. Two of them, BxlRSc and BxlR2Sc, could be identified as positive regulators of novobiocin production in S. coelicolor M512. Deletion of a third gene, sco0460, resulted in reduced novobiocin production, while overexpression had no effect. Furthermore, binding of BxlRSc to PnovH and to its own promoter region was confirmed via surface plasmon resonance spectroscopy.


Assuntos
Regulação Bacteriana da Expressão Gênica , Genes Bacterianos , Família Multigênica , Novobiocina/biossíntese , Streptomyces coelicolor/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Meios de Cultura , DNA Bacteriano/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Deleção de Genes , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Plasmídeos/genética , Regiões Promotoras Genéticas , Fator sigma/genética , Fator sigma/metabolismo , Streptomyces coelicolor/metabolismo
5.
J Struct Funct Genomics ; 13(2): 125-33, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22644392

RESUMO

Aminocoumarin antibiotics are natural products of soil-dwelling bacteria called Streptomycetes. They are potent inhibitors of DNA gyrase, an essential bacterial enzyme and validated drug target, and thus have attracted considerable interest as potential templates for drug development. To date, aminocoumarins have not seen widespread clinical application on account of their poor pharmacological properties. Through studying the structures and mechanisms of enzymes from their biosynthetic pathways we will be better informed to redesign these compounds through rational pathway engineering. Novobiocin, the simplest compound, requires at least seventeen gene products to convert primary metabolites into the mature antibiotic. We have solved the crystal structures of four diverse biosynthetic enzymes from the novobiocin pathway, and used these as three-dimensional frameworks for the interpretation of functional and mechanistic data, and to speculate about how they might have evolved. The structure determinations have ranged from the routine to the challenging, necessitating a variety of different approaches.


Assuntos
Antibacterianos/biossíntese , Novobiocina/biossíntese , Streptomyces/química , Antibacterianos/química , Proteínas de Bactérias/química , Carboidratos Epimerases/química , Dimetilaliltranstransferase/química , Ferroproteínas não Heme/química , Novobiocina/química , Proteína O-Metiltransferase/química , Estrutura Secundária de Proteína , Streptomyces/enzimologia , Relação Estrutura-Atividade , Especificidade por Substrato
6.
Chem Biol ; 18(3): 304-13, 2011 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-21439475

RESUMO

An artificial operon was synthesized, consisting of the genes for chorismate pyruvate-lyase of E. coli and for 4-hydroxybenzoate 3-hydroxylase of Corynebacterium cyclohexanicum. This operon, directing the biosynthesis of 3,4-dihdroxybenzoate, was expressed in the heterologous expression host Streptomyces coelicolor M512, together with a modified biosynthetic gene cluster for the aminocoumarin antibiotic clorobiocin. The resulting strain produced a clorobiocin derivative containing a 3,4-dihdroxybenzoyl moiety. Its structure was confirmed by MS and NMR analysis, and it was found to be a potent inhibitor of the gyrases from Escherichia coli and Staphylococcus aureus. Bioassays against different E. coli mutants suggested that this compound is actively imported by catechol siderophore transporters in the cell envelope. This study provides an example of the structure of a natural product that can be rationally modified by synthetic biology.


Assuntos
Antibacterianos/biossíntese , Proteínas da Membrana Bacteriana Externa/metabolismo , Hidroxibenzoatos/metabolismo , Novobiocina/análogos & derivados , Receptores de Superfície Celular/metabolismo , 4-Hidroxibenzoato-3-Mono-Oxigenase/genética , Antibacterianos/química , Antibacterianos/farmacologia , Corynebacterium/genética , DNA Girase/metabolismo , Testes de Sensibilidade a Antimicrobianos por Disco-Difusão , Escherichia coli/enzimologia , Escherichia coli/genética , Hidroxibenzoatos/química , Família Multigênica , Novobiocina/biossíntese , Novobiocina/química , Novobiocina/farmacologia , Oxo-Ácido-Liases/genética , Streptomyces coelicolor/genética , Streptomyces coelicolor/metabolismo , Inibidores da Topoisomerase II
7.
J Mol Biol ; 404(4): 611-26, 2010 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-20946900

RESUMO

CloQ is an aromatic prenyltransferase from the clorobiocin biosynthetic pathway of Streptomyces roseochromogenes var. oscitans. It is involved in the synthesis of the prenylated hydroxybenzoate moiety of the antibiotic, specifically catalyzing the attachment of a dimethylallyl moiety to 4-hydroxyphenylpyruvate. Herein, we report the crystal structure of CloQ and use it as a framework for interpreting biochemical data from both wild-type and variant proteins. CloQ belongs to the aromatic prenyltransferase family, which is characterized by an unusual core fold comprising five consecutive ααßß elements that form a central 10-stranded anti-parallel ß-barrel. The latter delineates a solvent-accessible cavity where substrates bind and catalysis takes place. This cavity has well-defined polar and nonpolar regions, which have distinct roles in substrate binding and facilitate a Friedel-Crafts-type mechanism. We propose that the juxtaposition of five positively charged residues in the polar region circumvents the necessity for a Mg(2+), which, by contrast, is a strict requirement for the majority of prenyltransferases characterized to date. Our structure of CloQ complexed with 4-hydroxyphenylpyruvate reveals the formation of a covalent link between the substrate and Cys215 to yield a thiohemiketal species. Through site-directed mutagenesis, we show that this link is not essential for enzyme activity in vitro. Furthermore, we demonstrate that CloQ will accept alternative substrates and, therefore, has the capacity to generate a range of prenylated compounds. Since prenylation is thought to enhance the bioactivity of many natural products, CloQ offers considerable promise as a biocatalyst for the chemoenzymatic synthesis of novel compounds with therapeutic potential.


Assuntos
Vias Biossintéticas/genética , Dimetilaliltranstransferase/química , Dimetilaliltranstransferase/metabolismo , Novobiocina/análogos & derivados , Streptomyces/enzimologia , Substituição de Aminoácidos/genética , Coenzimas/metabolismo , Cristalografia por Raios X , Dimetilaliltranstransferase/genética , Magnésio/metabolismo , Modelos Moleculares , Mutagênese Sítio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Novobiocina/biossíntese , Ácidos Fenilpirúvicos/metabolismo , Estrutura Terciária de Proteína
8.
Biopolymers ; 93(9): 823-32, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20578003

RESUMO

The biosynthetic gene clusters of the aminocoumarin antibiotics clorobiocin and coumermycin A(1) and of the liponucleoside antibiotic caprazamycin were stably integrated into the genomes of different host strains derived from Streptomyces coelicolor A3(2). For the heterologous expression of clorobiocin derivatives in a chemically defined medium, inclusion of 0.6% of the siloxylated ethylene oxide/propylene oxide copolymer Q2-5247 into the growth medium proved to result in a 4.8-fold increase of productivity. Presumably, this copolymer acts as an oxygen carrier. The additional inclusion of cobalt chloride (0.2-2 mg l(-1)) dramatically increased the percentage of the desired compound clorobiocin within the total produced clorobiocin derivatives. This is very likely due to a stimulation of a cobalamin-dependent methylation reaction catalyzed by the enzyme CloN6 of clorobiocin biosynthesis. All three investigated host strains (S. coelicolor M512, M1146 and M1154) gave similar production rates of total clorobiocin derivatives (on average, 158 mg l(-1) in the presence of 0.6% Q2-5247 and 0.2 mg l(-1) CoCl(2)). In contrast, heterologous production of caprazamycin derivatives was optimal in strain M1154 (amounts of 152 mg l(-1) on average).


Assuntos
Azepinas/metabolismo , Expressão Gênica , Genes Bacterianos , Família Multigênica , Novobiocina/análogos & derivados , Streptomyces coelicolor/metabolismo , Aminocumarinas/metabolismo , Novobiocina/biossíntese , Streptomyces coelicolor/genética
9.
Appl Microbiol Biotechnol ; 87(1): 261-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20127238

RESUMO

The biosynthetic gene cluster of the aminocoumarin antibiotic novobiocin comprises 20 coding sequences. Sixteen of them code for essential enzymes for novobiocin formation, transcribed in the form of a single 18-kb polycistronic mRNA. In the present study, we replaced the genuine promoter of this operon by the tetracycline-inducible promoter tcp830 and at the same time deleting the two pathway-specific positive regulator genes of novobiocin biosynthesis. The heterologous producer Streptomyces coelicolor M512 harboring the modified gene cluster produced, upon addition of 2 mg L(-1) of the inducer compound anhydrotetracyline, 3.4-fold more novobiocin than strains carrying the unmodified cluster. A second tcp830 promoter was inserted in the middle of the 18-kb operon in order to ensure adequate transcription of the rearmost genes. However, this did not lead to a further increase of novobiocin formation, showing that a single tcp830 promoter was sufficient to achieve high transcription of all 16 genes of the operon. A high induction of novobiocin formation was achieved within a wide range of anhydrotetracyline concentrations (0.25-2.0 mg L(-1)). Growth of the strains was not affected by these concentrations. The inducer compound could be added either at the time of inoculation or at any other time up to mid-growth phase, always achieving a similar final antibiotic production. Therefore, the tcp830 promoter presents a robust, easy-to-use system for the inducible expression of biosynthetic gene clusters in heterologous hosts, independent from the genuine regulatory network.


Assuntos
Antibacterianos/biossíntese , Novobiocina/biossíntese , Regiões Promotoras Genéticas , Streptomyces coelicolor/genética , Streptomyces coelicolor/metabolismo , Engenharia Genética
10.
Arch Microbiol ; 192(4): 289-97, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20177662

RESUMO

The production of antibiotics in different Streptomyces strains has been reported to be stimulated by the external addition of S-adenosylmethionine (SAM) and by overexpression of the SAM synthetase gene metK. We investigated the influence of SAM addition, and of the expression of SAM biosynthetic genes, on the production of the aminocoumarin antibiotic novobiocin in the heterologous producer strain Streptomyces coelicolor M512 (nov-BG1). External addition of SAM did not influence novobiocin accumulation. However, overexpression of a SAM synthase gene stimulated novobiocin formation, concomitant with an increase of the intracellular SAM concentration. Streptomyces genomes contain orthologs of all genes required for the SAM cycle known from mammals. In contrast, most other bacteria use a different cycle for SAM regeneration. Three secondary metabolic gene clusters, coding for the biosynthesis of structurally very different antibiotics in different Streptomyces strains, were found to contain an operon comprising all five putative genes of the SAM cycle. We cloned one of these operons into an expression plasmid, under control of a strong constitutive promoter. However, transformation of the heterologous novobiocin producer strain with this plasmid did not stimulate novobiocin production, but rather showed a detrimental effect on cell viability in the stationary phase and strongly reduced novobiocin accumulation.


Assuntos
Antibacterianos/biossíntese , Novobiocina/biossíntese , S-Adenosilmetionina/metabolismo , Streptomyces coelicolor/metabolismo , Clonagem Molecular , DNA Bacteriano/genética , Regulação Bacteriana da Expressão Gênica , Genes Bacterianos , Metionina Adenosiltransferase/genética , Metionina Adenosiltransferase/metabolismo , Família Multigênica , Plasmídeos , Análise de Sequência de DNA , Streptomyces coelicolor/genética , Streptomyces coelicolor/crescimento & desenvolvimento
11.
J Biosci Bioeng ; 109(3): 230-4, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20159569

RESUMO

Highly reproducible production values of the aminocoumarin antibiotic novobiocin were achieved by cultivation of a heterologous Streptomyces producer strain in commercially available square deepwell plates consisting of 24 wells of 3 ml culture volume each. Between parallel cultivation batches in the deepwell plates, novobiocin accumulation showed standard deviations of 4-9%, compared to 39% in baffled Erlenmeyer flasks. Mycelia used as inoculum could be frozen in the presence of 20% peptone and stored at -70 degrees C, allowing repeated cultivations from the same batch of inoculum over extended periods of time. Originally, novobiocin titers in the deepwell plate (5-12 mg l(-1)) were lower than in Erlenmeyer flasks (24 mg l(-1)). Optimization of the inoculation procedure as well as addition of a siloxylated ethylene oxide/propylene oxide copolymer, acting as oxygen carrier, to the production medium increased novobiocin production to 54 mg l(-1). The additional overexpression of the pathway-specific positive regulator gene novG increased novobiocin production to 163 mg l(-1). Harvesting the precultures in a defined section of growth phase greatly reduced variability between different batches of inoculum. The use of deepwell plates may considerably reduce the workload and cost of investigations of antibiotic biosynthesis in streptomycetes and other microorganisms due to the high reproducibility and the low requirement for shaker space and culture medium.


Assuntos
Antibacterianos/biossíntese , Reatores Biológicos/microbiologia , Técnicas de Cultura de Células/instrumentação , Novobiocina/biossíntese , Streptomyces/metabolismo , Desenho de Equipamento , Análise de Falha de Equipamento , Streptomyces/citologia
12.
J Mol Biol ; 395(2): 390-407, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19857499

RESUMO

NovP is an S-adenosyl-l-methionine-dependent O-methyltransferase that catalyzes the penultimate step in the biosynthesis of the aminocoumarin antibiotic novobiocin. Specifically, it methylates at 4-OH of the noviose moiety, and the resultant methoxy group is important for the potency of the mature antibiotic: previous crystallographic studies have shown that this group interacts directly with the target enzyme DNA gyrase, which is a validated drug target. We have determined the high-resolution crystal structure of NovP from Streptomyces spheroides as a binary complex with its desmethylated cosubstrate S-adenosyl-l-homocysteine. The structure displays a typical class I methyltransferase fold, in addition to motifs that are consistent with a divalent-metal-dependent mechanism. This is the first representative structure of a methyltransferase from the TylF superfamily, which includes a number of enzymes implicated in the biosynthesis of antibiotics and other therapeutics. The NovP structure reveals a number of distinctive structural features that, based on sequence conservation, are likely to be characteristic of the superfamily. These include a helical 'lid' region that gates access to the cosubstrate binding pocket and an active center that contains a 3-Asp putative metal binding site. A further conserved Asp likely acts as the general base that initiates the reaction by deprotonating the 4-OH group of the noviose unit. Using in silico docking, we have generated models of the enzyme-substrate complex that are consistent with the proposed mechanism. Furthermore, these models suggest that NovP is unlikely to tolerate significant modifications at the noviose moiety, but could show increasing substrate promiscuity as a function of the distance of the modification from the methylation site. These observations could inform future attempts to utilize NovP for methylating a range of glycosylated compounds.


Assuntos
Proteína O-Metiltransferase/química , Sequência de Aminoácidos , Domínio Catalítico , Cristalografia por Raios X , Modelos Moleculares , Dados de Sequência Molecular , Novobiocina/biossíntese , Novobiocina/química , Conformação Proteica , Multimerização Proteica , Proteína O-Metiltransferase/genética , Proteína O-Metiltransferase/metabolismo , S-Adenosil-Homocisteína/metabolismo , Homologia de Sequência de Aminoácidos , Streptomyces/enzimologia , Streptomyces/genética , Homologia Estrutural de Proteína
14.
Microbiology (Reading) ; 155(Pt 12): 4025-4035, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19762445

RESUMO

The aminocoumarin antibiotic novobiocin is a gyrase inhibitor formed by a Streptomyces strain. The biosynthetic gene cluster of novobiocin spans 23.4 kb and contains 20 coding sequences, among them the two regulatory genes novE and novG. We investigated the location of transcriptional promoters within this cluster by insertion of transcriptional terminator cassettes and RT-PCR analysis of the resulting mutants. The cluster was found to contain eight DNA regions with promoter activity. The regulatory protein NovG binds to a previously identified binding site within the promoter region located upstream of novH, but apparently not to any of the other seven promoters. Quantitative real-time PCR was used to compare the number of transcripts in a strain carrying an intact novobiocin cluster with strains carrying mutated clusters. Both in-frame deletion of the regulatory gene novG and insertion of a terminator cassette into the biosynthetic gene novH led to a strong reduction of the number of transcripts of the genes located between novH and novW. This suggested that these 16 biosynthetic genes form a single operon. Three internal promoters are located within this operon but appear to be of minor importance, if any, under our experimental conditions. Transcription of novG was found to depend on the presence of NovE, suggesting that the two regulatory genes, novE and novG, act in a cascade-like mechanism. The resistance gene gyrB(R), encoding an aminocoumarin-resistant gyrase B subunit, may initially be co-transcribed with the genes from novH to novW. However, when the gyrase inhibitor novobiocin accumulates in the cultures, gyrB(R) is transcribed from its own promoter. Previous work has suggested that this promoter is controlled by the superhelical density of chromosomal DNA.


Assuntos
Genes Bacterianos , Família Multigênica , Novobiocina/biossíntese , Streptomyces/genética , Streptomyces/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sequência de Bases , Sítios de Ligação/genética , DNA Girase/genética , DNA Girase/metabolismo , DNA Bacteriano/genética , Regulação Bacteriana da Expressão Gênica , Vetores Genéticos , Modelos Biológicos , Dados de Sequência Molecular , Novobiocina/química , Plasmídeos/genética , Regiões Promotoras Genéticas , RNA Bacteriano/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica
15.
Methods Enzymol ; 459: 437-55, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19362650

RESUMO

The aminocoumarin antibiotics novobiocin, clorobiocin and coumermycin A(1) are formed by different Streptomyces strains and are potent inhibitors of bacterial gyrase. Their biosynthetic gene clusters have been analyzed in detail by genetic and biochemical investigations. Heterologous expression of these gene clusters by site-specific integration into the genome of the fully sequenced host Streptomyces coelicolor A3(2) readily results in an accumulation of the antibiotics in yields similar to the wildtype strains. In recent years, the aminocoumarins have developed into a model system for the generation of new antibiotics by genetic methods. Prior to heterologous expression in S. coelicolor, cosmids containing the complete biosynthetic clusters can be manipulated in Escherichia coli by lambda RED-mediated recombination, creating single or multiple gene replacements or gene deletions. Thereby, mutant strains are generated which are blocked in the synthesis of certain intermediates or in specific tailoring reactions. For instance, mutasynthetic experiments can subsequently be carried out to generate aminocoumarin antibiotics that contain modified acyl moieties attached to the aminocoumarin core, and chemoenzymatic synthesis can be employed for the acylation of the deoxysugar moiety of structural analogues of the aminocoumarin antibiotics. Metabolic engineering-the combination of gene deletions and foreign gene expression via replicative expression vectors-can be used to generate further structural variants of these antibiotics. These methods can be combined, allowing the generation of a wide variety of new compounds. This chapter may provide general pointers for the use of genetic methods in the generation of new antibiotics.


Assuntos
Aminocumarinas/síntese química , Aminocumarinas/metabolismo , Amida Sintases/metabolismo , Aminocumarinas/química , Modelos Genéticos , Estrutura Molecular , Novobiocina/análogos & derivados , Novobiocina/biossíntese , Novobiocina/síntese química , Novobiocina/química
16.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 64(Pt 11): 1000-2, 2008 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-18997325

RESUMO

Crystals of recombinant NovN, an O-carbamoyltransferase from Streptomyces spheroides, were grown by vapour diffusion. The protein crystallized in two different crystal forms. Crystal form I belonged to space group C2 and native data were collected to 2.9 A resolution in-house. Crystal form II had I-centred orthorhombic symmetry and native data were recorded to a resolution of 2.3 A at a synchrotron. NovN catalyses the final step in the biosynthesis of the aminocoumarin antibiotic novobiocin that targets the essential bacterial enzyme DNA gyrase.


Assuntos
Proteínas de Bactérias/química , Carboxil e Carbamoil Transferases/química , Novobiocina/biossíntese , Streptomyces/enzimologia , Proteínas de Bactérias/genética , Carboxil e Carbamoil Transferases/genética , Cristalização , Cristalografia por Raios X , Inibidores Enzimáticos/metabolismo , Dados de Sequência Molecular , Família Multigênica
17.
Arch Microbiol ; 190(5): 509-19, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18568336

RESUMO

The biosynthetic gene cluster of the aminocoumarin antibiotic novobiocin contains two putative regulatory genes, i.e., novE and novG. Functional proof for the role of NovG as a positive regulator of novobiocin biosynthesis had been provided previously, and we now investigated the role of novE. Heterologous expression experiments with the novobiocin biosynthetic gene cluster showed that the entire putative promoter region of novE is required to achieve optimal novobiocin production. Overexpression of novE, using a replicative vector, resulted in an increase of novobiocin formation. In contrast, inactivation of novE by in frame deletion resulted in a strong reduction of novobiocin biosynthesis. Novobiocin production could be restored by an intact copy of novE, but also by the regulatory gene novG. These observations suggest that novE is a positive regulator of novobiocin biosynthesis. NovE was expressed in E. coli and purified. However, in contrast to parallel experiments with NovG, no DNA-binding properties could be shown for NovE. RT-PCR experiments showed that expression of novG was detectable in the absence of NovE, and also that expression of novE occurred in absence of NovG.


Assuntos
Antibacterianos/biossíntese , Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica , Novobiocina/biossíntese , Streptomyces/fisiologia , Proteínas de Bactérias/genética , Clonagem Molecular , DNA Bacteriano/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Escherichia coli/genética , Deleção de Genes , Dosagem de Genes , Expressão Gênica , Perfilação da Expressão Gênica , Teste de Complementação Genética , Família Multigênica , Mutagênese Insercional , Ligação Proteica , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Streptomyces/genética
18.
J Antibiot (Tokyo) ; 60(8): 504-10, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17827661

RESUMO

Three new aminocoumarin antibiotics, termed ferulobiocin, 3-chlorocoumarobiocin and 8'-dechloro-3-chlorocoumarobiocin, were isolated from the culture broth of a Streptomyces coelicolor M512 strain expressing a modified clorobiocin biosynthetic gene cluster. Structural analysis showed that these new aminocoumarins were very similar to clorobiocin, with a substituted 4-hydroxycinnamoyl moieties instead of the prenylated 4-hydroxybenzoyl moiety of clorobiocin. The possible biosynthetic origin of these moieties is discussed.


Assuntos
Aminocumarinas/metabolismo , Antibacterianos/biossíntese , Ácidos Cumáricos/metabolismo , Genes Bacterianos/genética , Família Multigênica/genética , Novobiocina/análogos & derivados , Streptomyces coelicolor/genética , Streptomyces coelicolor/metabolismo , Aminocumarinas/química , Aminocumarinas/isolamento & purificação , Antibacterianos/química , Antibacterianos/isolamento & purificação , Meios de Cultura/metabolismo , Expressão Gênica , Testes de Sensibilidade Microbiana , Novobiocina/biossíntese , Propionatos , Streptomyces coelicolor/crescimento & desenvolvimento
19.
Biochemistry ; 46(28): 8462-71, 2007 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-17580964

RESUMO

The aminocoumarin antibiotics novobiocin, clorobiocin, and coumermycin A1 are highly potent inhibitors of the bacterial type II topoisomerase DNA gyrase. The key pharmacophore of both clorobiocin and coumermycin A1, the 5-methyl-2-pyrrolylcarbonyl moiety, targets the ATP-binding site of GyrB. The 5-methyl-2-pyrrolylcarbonyl group is transferred by the acyltransferases Clo/CouN7 from the carrier proteins Clo/CouN1 to the 3'-hydroxyl of the l-noviosyl scaffold during the late steps of clorobiocin and coumermycin A1 biosynthesis. We first examined the substrate specificity of the purified thiolation domain protein CouN1 in becoming primed by the phosphopantetheinyltransferase Sfp using a variety of synthetic CoA analogues of the 5-methyl-2-pyrrolylcarbonyl moiety. The acyl-S-CouN1 thioesters were then assayed as donors to the 3'-OH group of descarbamoylnovobiocin by the acyltransferase CouN7, resulting in 21 novel variants with heterocyclic acyl groups installed on the noviosyl moiety of the aminocoumarin scaffold. Scaleup of a 5-methylthiophene derivative yielded a compound with activity against both Gram-negative and Gram-positive bacteria. The minimal inhibitory concentration found for the Gram-positive bacteria was comparable to that of novobiocin.


Assuntos
Aciltransferases/metabolismo , Antibacterianos/biossíntese , Proteínas de Transporte/metabolismo , Novobiocina/análogos & derivados , Acil Coenzima A/síntese química , Acil Coenzima A/química , Acil Coenzima A/metabolismo , Antibacterianos/química , Antibacterianos/farmacologia , Apoenzimas/metabolismo , Cromatografia Líquida , Espectrometria de Massas , Novobiocina/biossíntese , Novobiocina/farmacologia
20.
Microbiology (Reading) ; 153(Pt 5): 1413-1423, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17464055

RESUMO

In the biosynthetic gene cluster of the aminocoumarin antibiotic clorobiocin, the small ORF cloY encodes a 71 aa protein which shows significant sequence similarity to mbtH from the mycobactin biosynthetic gene cluster of Mycobacterium tuberculosis. mbtH-like genes are frequently found in the biosynthetic gene clusters of peptide antibiotics and siderophores, but their function has remained enigmatic. In a recent publication it has been suggested that these genes may have no function for secondary metabolite biosynthesis. An in-frame deletion of cloY in the clorobiocin cluster has now been carried out. When the modified cluster was expressed in the heterologous host Streptomyces coelicolor M512, clorobiocin was still formed. However, when the two further mbtH-like genes from elsewhere in the host genome were inactivated as well, clorobiocin formation was reduced dramatically. Complementation with cloY or with any of three other mbtH-like genes restored clorobiocin formation. This is the first report proving the requirement of an mbtH-like gene for secondary metabolite formation, and the first proof that different mbtH-like genes can functionally replace each other. Feeding of an mbtH-defective triple mutant strain with an intact 3-amino-4,7-dihydroxy-coumarin moiety restored antibiotic production, showing that cloY is specifically required for the formation of this moiety of the clorobiocin molecule.


Assuntos
Proteínas de Bactérias/fisiologia , Deleção de Genes , Novobiocina/análogos & derivados , Streptomyces coelicolor/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Cromatografia Líquida de Alta Pressão , Cumarínicos/metabolismo , Teste de Complementação Genética , Dados de Sequência Molecular , Estrutura Molecular , Família Multigênica , Mycobacterium tuberculosis/genética , Novobiocina/biossíntese , Fases de Leitura Aberta , Alinhamento de Sequência , Streptomyces coelicolor/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA