Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Beilstein J Org Chem ; 15: 2922-2929, 2019.
Article in English | MEDLINE | ID: mdl-31839838

ABSTRACT

The argyrins are secondary metabolites from myxobacteria with antibiotic activity against Pseudomonas aeruginosa. Studying their structure-activity relationship is hampered by the complexity of the chemical total synthesis. Mutasynthesis is a promising approach where simpler and fully synthetic intermediates of the natural product's biosynthesis can be biotechnologically incorporated. Here, we report the synthesis of a series of tripeptide thioesters as mutasynthons containing the native sequence with a dehydroalanine (Dha) Michael acceptor attached to a sarcosine (Sar) and derivatives. Chemical synthesis of the native sequence ᴅ-Ala-Dha-Sar thioester required revision of the sequential peptide synthesis into a convergent strategy where the thioester with sarcosine was formed before coupling to the Dha-containing dipeptide.

2.
Nat Commun ; 10(1): 4055, 2019 09 06.
Article in English | MEDLINE | ID: mdl-31492836

ABSTRACT

Long-chain polyunsaturated fatty acids (LC-PUFAs), particularly the omega-3 LC-PUFAs eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA), and docosahexaenoic acid (DHA), have been associated with beneficial health effects. Consequently, sustainable sources have to be developed to meet the increasing demand for these PUFAs. Here, we demonstrate the design and construction of artificial PUFA biosynthetic gene clusters (BGCs) encoding polyketide synthase-like PUFA synthases from myxobacteria adapted for the oleaginous yeast Yarrowia lipolytica. Genomic integration and heterologous expression of unmodified or hybrid PUFA BGCs yielded different yeast strains with specific LC-PUFA production profiles at promising yield and thus valuable for the biotechnological production of distinct PUFAs. Nutrient screening revealed a strong enhancement of PUFA production, when cells were phosphate limited. This represents, to the best of our knowledge, highest concentration of DHA (16.8 %) in total fatty acids among all published PUFA-producing Y. lipolytica strains.


Subject(s)
Bacterial Proteins/metabolism , Fatty Acid Synthases/metabolism , Fatty Acids, Unsaturated/biosynthesis , Myxococcales/enzymology , Yarrowia/metabolism , Bacterial Proteins/genetics , Biotechnology/methods , Docosahexaenoic Acids/metabolism , Fatty Acid Synthases/genetics , Fatty Acids, Omega-3/metabolism , Fatty Acids, Unsaturated/metabolism , Metabolic Engineering/methods , Myxococcales/genetics , Reproducibility of Results
3.
Metab Eng ; 55: 201-211, 2019 09.
Article in English | MEDLINE | ID: mdl-31340171

ABSTRACT

Corallopyronins (COR) are α-pyrone antibiotics from myxobacteria representing highly promising lead structures for the development of antibacterial therapeutic agents. Their ability to inhibit RNA polymerase through interaction with the "switch region", a novel target, distant from binding sites of previously characterized RNA polymerase inhibitors (e.g. rifampicin), makes them particularly promising as antibiotic candidates. Corallopyronin A is currently also investigated as a lead compound for the treatment of lymphatic filariasis because of its superb activity against the nematode symbiont Wolbachia. As total synthesis is not a valid production option biotechnological optimization of compound supply is of utmost importance to further develop this highly potent compound class. Here we describe decisive improvements of the previously reported heterologous COR production and engineering platform yielding production of ~100 mg/L COR A. Furthermore, we provide a revised model of COR biosynthesis shedding light on the function of several biosynthetic proteins, including an unusual ECH-like enzyme providing dehydration functionality in trans and an uncharacterized protein conferring COR self-resistance in the myxobacterial heterologous host Myxococcus xanthus DK1622. We also report two new COR derivatives, COR D and oxyCOR A discovered in genetically engineered strains.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Filaricides/metabolism , Lactones/metabolism , Microorganisms, Genetically-Modified , Myxococcus xanthus , Microorganisms, Genetically-Modified/genetics , Microorganisms, Genetically-Modified/metabolism , Myxococcus xanthus/genetics , Myxococcus xanthus/metabolism
4.
ACS Synth Biol ; 8(5): 1121-1133, 2019 05 17.
Article in English | MEDLINE | ID: mdl-30995838

ABSTRACT

Argyrins represent a family of cyclic octapeptides exhibiting promising antimicrobial, antitumorigenic and immunosuppressant activities. They derive from a nonribosomal peptide synthetase pathway, which was identified and characterized in this study from the myxobacterial producer strain Cystobacter sp. SBCb004. Using the native biosynthetic gene cluster (BGC) sequence as template synthetic BGC versions were designed and assembled from gene synthesis fragments. A heterologous expression system was established after chromosomal deletion of a well-expressed lipopeptide pathway from the host strain Myxococcus xanthus DK1622. Different approaches were applied to engineer and improve heterologous argyrin production, which was finally increased to 160 mg/L, around 20-fold higher yields compared to the native producer. Heterologous production platform also led to identification of several novel argyrin derivatives (A2, F3, G3, I, J, K, and L). The optimized production system provides a versatile platform for future supply of argyrins and novel derivatives thereof.


Subject(s)
Peptides, Cyclic/biosynthesis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Metabolic Engineering/methods , Multigene Family , Myxococcus xanthus/metabolism , Peptide Synthases/genetics , Peptide Synthases/metabolism
5.
Chem Sci ; 9(38): 7510-7519, 2018 Oct 14.
Article in English | MEDLINE | ID: mdl-30319751

ABSTRACT

Synthetic biology techniques coupled with heterologous secondary metabolite production offer opportunities for the discovery and optimisation of natural products. Here we developed a new assembly strategy based on type IIS endonucleases and elaborate synthetic DNA platforms, which could be used to seamlessly assemble and engineer biosynthetic gene clusters (BGCs). By applying this versatile tool, we designed and assembled more than thirty different artificial myxochromide BGCs, each around 30 kb in size, and established heterologous expression platforms using a derivative of Myxococcus xanthus DK1622 as a host. In addition to the five native types of myxochromides (A, B, C, D and S), novel lipopeptide structures were produced by combinatorial exchange of nonribosomal peptide synthetase (NRPS) encoding genes from different myxochromide BGCs. Inspired by the evolutionary diversification of the native myxochromide megasynthetases, the ancestral A-type NRPS was engineered by inactivation, deletion, or duplication of catalytic domains and successfully converted into functional B-, C- and D-type megasynthetases. The constructional design approach applied in this study enables combinatorial engineering of complex synthetic BGCs and has great potential for the exploitation of other natural product biosynthetic pathways.

6.
Metab Eng ; 44: 160-170, 2017 11.
Article in English | MEDLINE | ID: mdl-29030273

ABSTRACT

Myxopyronins (MXN) and corallopyronins (COR) are structurally related α-pyrone antibiotics from myxobacteria that represent a highly promising compound class for the development of broad-spectrum antibacterial therapeutic agents. Their ability to inhibit RNA polymerase through interaction with the "switch region", a novel target, distant from previously characterized RNA polymerase inhibitors (e.g. rifampicin), makes them particularly promising candidates for further research. To improve compound supply for further investigation of MXN, COR and novel derivatives of these antibacterial agents, establishment of an efficient and versatile microbial production platform for myxobacterial α-pyrone antibiotics is highly desirable. Here we describe design, construction and expression of a heterologous production and engineering platforms for MXN and COR to facilitate rational structure design and yield improvement approaches in the myxobacterial host strain Myxococcus xanthus DK1622. Optimization of the cultivation medium yielded significantly higher production titers of MXN A at around 41-fold increase and COR A at around 25-fold increase, compared to the standard CTT medium.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Lactones/metabolism , Metabolic Engineering , Myxococcus xanthus , Pyrones/metabolism , Myxococcus xanthus/genetics , Myxococcus xanthus/metabolism
7.
Metab Eng ; 44: 213-222, 2017 11.
Article in English | MEDLINE | ID: mdl-28951265

ABSTRACT

Coronatine (COR) represents a phytotoxin produced by several pathovars of Pseudomonas syringae. It mediates multiple virulence activities by mimicking the plant stress hormone jasmonoyl-l-isoleucine. Structurally, COR consists of a bicyclic polyketide moiety, coronafacic acid (CFA), which is linked via an amide bond to an unusual ethylcyclopropyl amino acid moiety, coronamic acid (CMA). In our studies, we aimed at establishing and engineering of heterologous COR and CFA production platforms using P. putida KT2440 as host. Based on genetic information of the native producer P. syringae pv. tomato DC3000 a COR biosynthetic gene cluster was designed and reconstituted from synthetic DNA fragments. The applied constructional design facilitated versatile pathway modifications and the generation of various expression constructs, which were evaluated for the production of CFA, COR and its derivatives. By modifications of the gene cluster composition production profiles were directed towards target compounds and valuable information about the function and impact of selected pathway proteins on COR biosynthesis were obtained. Additional engineering of expression vector features, including the use of the constitutive PrpsH promoter and a p15Aori-based transposon backbone, led to the development of an expression strain with promising CFA production yields of > 90mg/l.


Subject(s)
Amino Acids , Indenes , Metabolic Engineering , Pseudomonas putida , Pseudomonas syringae/genetics , Synthetic Biology , Amino Acids/biosynthesis , Amino Acids/genetics , Pseudomonas putida/genetics , Pseudomonas putida/metabolism , Pseudomonas syringae/metabolism
8.
ACS Chem Biol ; 12(3): 779-786, 2017 03 17.
Article in English | MEDLINE | ID: mdl-28128551

ABSTRACT

Analysis of 122 myxobacterial genome sequences suggested 16 strains as producers of the myxochromide lipopeptide family. Detailed sequence comparison of the respective mch biosynthetic gene clusters informed a genome-mining approach, ultimately leading to the discovery and chemical characterization of four novel myxochromide core types. The myxochromide megasynthetase is subject to evolutionary diversification, resulting in considerable structural diversity of biosynthesis products. The observed differences are due to the number, type, sequence, and configuration of the incorporated amino acids. The analysis revealed molecular details on how point mutations and recombination events led to structural diversity. It also gave insights into the evolutionary scenarios that have led to the emergence of mch clusters in different strains and genera of myxobacteria.


Subject(s)
Genomics , Lipopeptides/metabolism , Myxococcales/genetics , Multigene Family , Myxococcales/metabolism
9.
Metab Eng ; 33: 98-108, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26617065

ABSTRACT

Long-chain polyunsaturated fatty acids (LC-PUFAs) can be produced de novo via polyketide synthase-like enzymes known as PUFA synthases, which are encoded by pfa biosynthetic gene clusters originally discovered from marine microorganisms. Recently similar gene clusters were detected and characterized in terrestrial myxobacteria revealing several striking differences. As the identified myxobacterial producers are difficult to handle genetically and grow very slowly we aimed to establish heterologous expression platforms for myxobacterial PUFA synthases. Here we report the heterologous expression of the pfa gene cluster from Aetherobacter fasciculatus (SBSr002) in the phylogenetically distant model host bacteria Escherichia coli and Pseudomonas putida. The latter host turned out to be the more promising PUFA producer revealing higher production rates of n-6 docosapentaenoic acid (DPA) and docosahexaenoic acid (DHA). After several rounds of genetic engineering of expression plasmids combined with metabolic engineering of P. putida, DHA production yields were eventually increased more than threefold. Additionally, we applied synthetic biology approaches to redesign and construct artificial versions of the A. fasciculatus pfa gene cluster, which to the best of our knowledge represents the first example of a polyketide-like biosynthetic gene cluster modulated and synthesized for P. putida. Combination with the engineering efforts described above led to a further increase in LC-PUFA production yields. The established production platform based on synthetic DNA now sets the stage for flexible engineering of the complex PUFA synthase.


Subject(s)
Amide Synthases/metabolism , Docosahexaenoic Acids/biosynthesis , Fatty Acids, Unsaturated/metabolism , Metabolic Engineering/methods , Myxococcales/enzymology , Pseudomonas putida/enzymology , Amide Synthases/genetics , Cloning, Molecular/methods , Docosahexaenoic Acids/genetics , Docosahexaenoic Acids/isolation & purification , Fatty Acids, Unsaturated/genetics , Myxococcales/genetics , Pseudomonas putida/genetics , Recombinant Proteins/metabolism
10.
Angew Chem Int Ed Engl ; 54(51): 15560-4, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26514647

ABSTRACT

The bengamides, sponge-derived natural products that have been characterized as inhibitors of methionine aminopeptidases (MetAPs), have been intensively investigated as anticancer compounds. We embarked on a multidisciplinary project to supply bengamides by fermentation of the terrestrial myxobacterium M. virescens, decipher their biosynthesis, and optimize their properties as drug leads. The characterization of the biosynthetic pathway revealed that bacterial resistance to bengamides is conferred by Leu 154 of the myxobacterial MetAP protein, and enabled transfer of the entire gene cluster into the more suitable production host M. xanthus DK1622. A combination of semisynthesis of microbially derived bengamides and total synthesis resulted in an optimized derivative that combined high cellular potency in the nanomolar range with high metabolic stability, which translated to an improved half-life in mice and antitumor efficacy in a melanoma mouse model.


Subject(s)
Azepines/metabolism , Biological Products/metabolism , Marine Biology , Myxococcales/metabolism , Porifera/metabolism , Animals , Area Under Curve , Azepines/pharmacokinetics , Azepines/pharmacology , Biological Products/pharmacokinetics , Biological Products/pharmacology , Female , Half-Life , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Structure-Activity Relationship
11.
Science ; 348(6239): 1106-12, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-26045430

ABSTRACT

The discovery of Streptomyces-produced streptomycin founded the age of tuberculosis therapy. Despite the subsequent development of a curative regimen for this disease, tuberculosis remains a worldwide problem, and the emergence of multidrug-resistant Mycobacterium tuberculosis has prioritized the need for new drugs. Here we show that new optimized derivatives from Streptomyces-derived griselimycin are highly active against M. tuberculosis, both in vitro and in vivo, by inhibiting the DNA polymerase sliding clamp DnaN. We discovered that resistance to griselimycins, occurring at very low frequency, is associated with amplification of a chromosomal segment containing dnaN, as well as the ori site. Our results demonstrate that griselimycins have high translational potential for tuberculosis treatment, validate DnaN as an antimicrobial target, and capture the process of antibiotic pressure-induced gene amplification.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Molecular Targeted Therapy , Mycobacterium tuberculosis/drug effects , Peptides, Cyclic/pharmacology , Tuberculosis, Multidrug-Resistant/drug therapy , Animals , Antitubercular Agents/chemistry , Antitubercular Agents/therapeutic use , Cell Line, Tumor , Crystallography, X-Ray , DNA-Directed DNA Polymerase , Disease Models, Animal , Drug Design , Humans , Mice , Microbial Sensitivity Tests , Molecular Sequence Data , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/enzymology , Mycobacterium tuberculosis/enzymology , Peptides, Cyclic/chemistry , Peptides, Cyclic/therapeutic use , Protein Structure, Secondary , Streptomyces/chemistry , Streptomyces/drug effects , Streptomyces/metabolism , Tuberculosis, Multidrug-Resistant/microbiology
12.
Chembiochem ; 16(6): 946-53, 2015 Apr 13.
Article in English | MEDLINE | ID: mdl-25757034

ABSTRACT

Myxopyronin is a natural α-pyrone antibiotic from the soil bacterium Myxococcus fulvus Mx f50. Myxopyronin inhibits bacterial RNA polymerase (RNAP) by binding to a part of the enzyme not targeted by the clinically used rifamycins. This mode of action makes myxopyronins promising molecules for the development of novel broad-spectrum antibacterials. We describe the derivatization of myxopyronins by an advanced mutasynthesis approach as a first step towards this goal. Site-directed mutagenesis of the biosynthetic machinery was used to block myxopyronin biosynthesis at different stages. The resulting mutants were fed with diverse precursors that mimic the biosynthetic intermediates to restore production. Mutasynthon incorporation and production of novel myxopyronin derivatives were analyzed by HPLC-MS/MS. This work sets the stage for accessing numerous myxopyronin derivatives, thus significantly expanding the chemical space of f α-pyrone antibiotics.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Biological Products/metabolism , Mutation , Myxococcus/genetics , Myxococcus/metabolism , Pyrones/metabolism , Anti-Bacterial Agents/chemistry , Biological Products/chemistry , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Models, Molecular , Protein Conformation , Pyrones/chemistry , Thermus thermophilus/enzymology
13.
J Biotechnol ; 191: 54-63, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25102237

ABSTRACT

Enormous progress in the field of polyketide biosynthesis has led to the establishment of rules for general text book biosynthetic logic and consequently to the assumption that biosynthetic genes can be easily correlated with the corresponding natural products. However, non-textbook examples of polyketide assembly continue to be discovered suggesting the gene to product and product to gene predictions need improvement, especially as they are increasingly used in the post-genomic era. Here, we analyzed the genomic blueprint of a myxobacterial multi-producer of secondary metabolites, Stigmatella aurantiaca DW4/3-1, for its biosynthetic potential by genome-mining. In addition to the five polyketide synthase and/or nonribosomal peptide synthetase gene clusters of known function we identified a further 13 genomic regions exemplifying the enormous genetic potential for the production of additional chemical diversity by this strain. We show by gene inactivation and heterologous expression of the newly identified biosynthetic pathway for dawenol that the biosynthesis of this known polyene does not follow text book biosynthetic logic. Intriguingly, a genomic locus encoding an unusual polyketide synthase exhibiting similarity to gene loci involved in the formation of polyunsaturated fatty acids and secondary lipids was identified.


Subject(s)
Polyenes/metabolism , Polyketide Synthases/chemistry , Stigmatella aurantiaca/enzymology , Amino Acid Sequence/genetics , Biosynthetic Pathways , Multienzyme Complexes/metabolism , Peptide Synthases/genetics , Polyenes/chemistry , Polyketide Synthases/genetics , Polyketide Synthases/isolation & purification
14.
ACS Synth Biol ; 3(10): 759-72, 2014 Oct 17.
Article in English | MEDLINE | ID: mdl-23654254

ABSTRACT

Natural products of microbial origin continue to be an important source of pharmaceuticals and agrochemicals exhibiting potent activities and often novel modes of action. Due to their inherent structural complexity chemical synthesis is often hardly possible, leaving fermentation as the only viable production route. In addition, the pharmaceutical properties of natural products often need to be optimized for application by sophisticated medicinal chemistry and/or biosynthetic engineering. The latter requires a detailed understanding of the biosynthetic process and genetic tools to modify the producing organism that are often unavailable. Consequently, heterologous expression of complex natural product pathways has been in the focus of development over recent years. However, piecing together existing DNA cloned from natural sources and achieving efficient expression in heterologous circuits represent several limitations that can be addressed by synthetic biology. In this work we have redesigned and reassembled the 56 kb epothilone biosynthetic gene cluster from Sorangium cellulosum for expression in the high GC host Myxococcus xanthus. The codon composition was adapted to a modified codon table for M. xanthus, and unique restriction sites were simultaneously introduced and others eliminated from the sequence in order to permit pathway assembly and future interchangeability of modular building blocks from the epothilone megasynthetase. The functionality of the artificial pathway was demonstrated by successful heterologous epothilone production in M. xanthus at significant yields that have to be improved in upcoming work. Our study sets the stage for future engineering of epothilone biosynthesis and production optimization using a highly flexible assembly strategy.


Subject(s)
Epothilones/biosynthesis , Genes, Synthetic , Polyketide Synthases/genetics , Polyketide Synthases/metabolism , Algorithms , Biosynthetic Pathways/genetics , Biotechnology , Codon/genetics , Epothilones/chemistry , Epothilones/genetics , Genetic Engineering , Multigene Family , Myxococcales/genetics , Myxococcales/metabolism , Myxococcus xanthus/genetics , Myxococcus xanthus/metabolism , Polyketides/metabolism , Synthetic Biology
15.
Chembiochem ; 14(13): 1581-9, 2013 Sep 02.
Article in English | MEDLINE | ID: mdl-23983106

ABSTRACT

Myxopyronins and corallopyronins are structurally related α-pyrone antibiotics from myxobacteria. They are thought to represent a highly promising compound class for the development of broad-spectrum antibacterial therapeutic agents, because of their ability to inhibit RNA polymerase through interaction with the "switch region", a recently identified novel drug target. Here we describe the identification and characterization of the myxopyronin biosynthetic pathway from Myxococcus fulvus Mx f50. A detailed comparison with the recently identified corallopyronin biosynthetic pathway revealed the genetic and biochemical basis, thus explaining the observed structural differences between the two natural product families. Directed mutagenesis procedures for M. fulvus Mx f50 were developed to enable functional studies and pathway modifications. Our work provided new insights into myxopyronin biosynthesis and led to the production of a novel and unexpected myxopyronin derivative.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/chemistry , Lactones/metabolism , Myxococcus/chemistry , Lactones/chemistry , Molecular Conformation , Myxococcus/metabolism , Pyrones
16.
Chem Biol ; 19(10): 1278-87, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-23021914

ABSTRACT

Bottromycins represent a promising class of antibiotics binding to the therapeutically unexploited A-site of the bacterial ribosome. By inhibiting translation they are active against clinically important pathogens, such as vancomycin-resistant Enterococci. Structurally, bottromycins are heavily modified peptides exhibiting various unusual biosynthetic features. To set the stage for compound modification and yield optimization, we identified the biosynthetic gene cluster, used synthetic biotechnology approaches to establish and improve heterologous production, and generated analogs by pathway genetic engineering. We unambiguously identified three radical SAM methyltransferase-encoding genes required for various methylations at unactivated carbons yielding tert-butyl valine, methyl-proline, and ß-methyl-phenylalanine residues, plus a gene involved in aspartate methyl-ester formation. Evidence for the formation of the exo-thiazole unit and for a macrocyclodehydration mechanism leading to amidine ring formation is provided.


Subject(s)
Ribosomes/metabolism , Amidines/chemistry , Base Sequence , Biotechnology , Cyclization , Enterococcus/genetics , Enterococcus/metabolism , Genetic Engineering , Methyltransferases/metabolism , Molecular Sequence Data , Multigene Family , Peptides, Cyclic/biosynthesis , S-Adenosylmethionine/metabolism , Streptomyces/genetics , Streptomyces/metabolism
18.
Mol Biosyst ; 5(6): 567-74, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19462013

ABSTRACT

In this article, we briefly review the potential of myxobacteria as 'natural product factories' by highlighting results from the recently sequenced myxobacterial model strain Myxococcus xanthus. We will focus on the production of polyketides, non-ribosomally-made peptides, and their hybrids, and discuss the evaluation of biosynthetic potential using genome-based methods, as well as biosynthetic process engineering.


Subject(s)
Industrial Microbiology/methods , Myxococcales/genetics , Myxococcales/metabolism , Biosynthetic Pathways , Epothilones/biosynthesis , Epothilones/chemistry , Gene Expression Regulation, Bacterial , Genetic Engineering , Macrolides/chemistry , Macrolides/metabolism , Myxococcales/growth & development , Myxococcus xanthus/genetics , Myxococcus xanthus/metabolism
19.
Curr Opin Drug Discov Devel ; 12(2): 220-30, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19333867

ABSTRACT

Myxobacteria produce an immense variety of natural products with useful biological activities. This review highlights recent advances to evaluate and further explore their biosynthetic potential for drug discovery, with particular focus on polyketide synthase and non-ribosomal peptide synthetase-derived secondary metabolites.


Subject(s)
Biological Products/biosynthesis , Drug Discovery , Myxococcales/enzymology , Animals , Anti-Infective Agents/metabolism , Antineoplastic Agents/metabolism , Biological Products/chemistry , Biological Products/genetics , Biological Products/pharmacology , Gene Expression Regulation, Bacterial , Gene Regulatory Networks , Humans , Myxococcales/genetics , Peptide Synthases/metabolism , Polyketide Synthases/metabolism , Structure-Activity Relationship
20.
Chembiochem ; 9(16): 2711-21, 2008 Nov 03.
Article in English | MEDLINE | ID: mdl-18972512

ABSTRACT

Kendomycin is a bioactive polyketide that is produced by various Streptomyces strains. It displays strong antibiotic activities against a wide range of bacteria and exhibits remarkable cytotoxic effects on the growth of several human cancer cell lines. In this study we cloned the corresponding biosynthetic locus from the producer Streptomyces violaceoruber (strain 3844-33C). Our analysis shows that a mixed type I/type III polyketide synthase pathway is responsible for the formation of the fully carbogenic macrocyclic scaffold of kendomycin, which is unprecedented among all of the ansa compounds that have been isolated so far. Heterologous expression of a gene set in Streptomyces coelicolor shows that 3,5-dihydroxybenzoic acid is an intermediate in the starter unit biosynthesis that is initiated by the type III polyketide synthase. The identification of the kendomycin biosynthetic gene cluster sets the stage to study a novel chain termination mechanism by a type I PKS that leads to carbocycle formation and provides the starting material for the heterologous expression of the entire pathway, and the production of novel derivatives by genetic engineering.


Subject(s)
Acyltransferases/metabolism , Polyketide Synthases/metabolism , Rifabutin/analogs & derivatives , Streptomyces/enzymology , Base Sequence , Drug Resistance, Bacterial/genetics , Gene Expression Regulation, Bacterial , Gene Library , Gene Silencing , Hydroxybenzoates/metabolism , Multigene Family/genetics , Resorcinols , Rifabutin/metabolism , Sequence Analysis, DNA , Streptomyces/genetics , Streptomyces/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...