Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Chem Biol ; 18(4): 794-802, 2023 04 21.
Article in English | MEDLINE | ID: mdl-37005433

ABSTRACT

Pseudouridimycin is a microbial C-nucleoside natural product that specifically inhibits bacterial RNA polymerases by binding to the active site and competing with uridine triphosphate for the nucleoside triphosphate (NTP) addition site. Pseudouridimycin consists of 5'-aminopseudouridine and formamidinylated, N-hydroxylated Gly-Gln dipeptide moieties to allow Watson-Crick base pairing and to mimic protein-ligand interactions of the triphosphates of NTP, respectively. The metabolic pathway of pseudouridimycin has been studied in Streptomyces species, but no biosynthetic steps have been characterized biochemically. Here, we show that the flavin-dependent oxidase SapB functions as a gate-keeper enzyme selecting pseudouridine (KM = 34 µM) over uridine (KM = 901 µM) in the formation of pseudouridine aldehyde. The pyridoxal phosphate (PLP)-dependent SapH catalyzes transamination, resulting in 5'-aminopseudouridine with a preference for arginine, methionine, or phenylalanine as cosubstrates as amino group donors. The binary structure of SapH in complex with pyridoxamine-5'-phosphate and site-directed mutagenesis identified Lys289 and Trp32 as key residues for catalysis and substrate binding, respectively. The related C-nucleoside oxazinomycin was accepted as a substrate by SapB with moderate affinity (KM = 181 µM) and was further converted by SapH, which opens possibilities for metabolic engineering to generate hybrid C-nucleoside pseudouridimycin analogues in Streptomyces.


Subject(s)
Nucleosides , Pseudouridine , Biosynthetic Pathways , DNA-Directed RNA Polymerases/metabolism , Nucleosides/metabolism , Pseudouridine/biosynthesis , Pseudouridine/metabolism , Pyridoxal Phosphate/chemistry , Streptomyces/chemistry , Streptomyces/metabolism
2.
Eur J Med Chem ; 237: 114342, 2022 Jul 05.
Article in English | MEDLINE | ID: mdl-35439612

ABSTRACT

Showdomycin produced by Streptomyces showdoensis ATCC 15227 is a C-nucleoside microbial natural product with antimicrobial and cytotoxic properties. The unique feature of showdomycin in comparison to other nucleosides is its maleimide base moiety, which has the distinct ability to alkylate nucleophilic thiol groups by a Michael addition reaction. In order to understand structure-activity relationships of showdomycin, we synthesized a series of derivatives with modifications in the maleimide ring at the site of alkylation to moderate its reactivity. The showdomycin congeners were designed to retain the planarity of the base ring system to allow Watson-Crick base pairing and preserve the nucleosidic character of the compounds. Consequently, we synthesized triphosphates of showdomycin derivatives and tested their activity against RNA polymerases. Bromo, methylthio, and ethylthio derivatives of showdomycin were incorporated into RNA by bacterial and mitochondrial RNA polymerases and somewhat less efficiently by the eukaryotic RNA polymerase II. Showdomycin derivatives acted as uridine mimics and delayed further extension of the RNA chain by multi-subunit, but not mitochondrial RNA polymerases. Bioactivity profiling indicated that the mechanism of action of ethylthioshowdomycin was altered, with approximately 4-fold reduction in both cytotoxicity against human embryonic kidney cells and antibacterial activity against Escherichia coli. In addition, the ethylthio derivative was not inactivated by medium components or influenced by addition of uridine in contrast to showdomycin. The results explain how both the maleimide ring and the nucleoside nature contribute to the bioactivity of showdomycin and demonstrates for the first time that the two activities can be separated.


Subject(s)
Nucleosides , Showdomycin , Anti-Bacterial Agents/pharmacology , Humans , Maleimides/pharmacology , RNA , Showdomycin/pharmacology , Structure-Activity Relationship , Uridine
3.
Chembiochem ; 21(21): 3062-3066, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32557994

ABSTRACT

Nogalamycin is an anthracycline anti-cancer agent that intercalates into the DNA double helix. The binding is facilitated by two carbohydrate units, l-nogalose and l-nogalamine, that interact with the minor and major grooves of DNA, respectively. However, recent investigations have shown that nogalamycin biosynthesis proceeds through the attachment of l-rhodosamine (2''-deoxy-4''-epi-l-nogalamine) to the aglycone. Herein, we demonstrate that the Rieske enzyme SnoT catalyzes 2''-hydroxylation of l-rhodosamine as an initial post-glycosylation step. Furthermore, we establish that the reaction order continues with 2-5'' carbocyclization and 4'' epimerization by the non-heme iron and 2-oxoglutarate-dependent enzymes SnoK and SnoN, respectively. These late-stage tailoring steps are important for the bioactivity of nogalamycin due to involvement of the 2''- and 4''-hydroxy groups of l-nogalamine in hydrogen bonding interactions with DNA.


Subject(s)
Amines/metabolism , Nogalamycin/biosynthesis , Oxygenases/metabolism , Amines/chemistry , Biocatalysis , Glycosylation , Hydroxylation , Models, Molecular , Molecular Conformation , Nogalamycin/chemistry
4.
Nucleic Acids Res ; 47(19): 10296-10312, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31495891

ABSTRACT

Oxazinomycin is a C-nucleoside antibiotic that is produced by Streptomyces hygroscopicus and closely resembles uridine. Here, we show that the oxazinomycin triphosphate is a good substrate for bacterial and eukaryotic RNA polymerases (RNAPs) and that a single incorporated oxazinomycin is rapidly extended by the next nucleotide. However, the incorporation of several successive oxazinomycins or a single oxazinomycin in a certain sequence context arrested a fraction of the transcribing RNAP. The addition of Gre RNA cleavage factors eliminated the transcriptional arrest at a single oxazinomycin and shortened the nascent RNAs arrested at the polythymidine sequences suggesting that the transcriptional arrest was caused by backtracking of RNAP along the DNA template. We further demonstrate that the ubiquitous C-nucleoside pseudouridine is also a good substrate for RNA polymerases in a triphosphorylated form but does not inhibit transcription of the polythymidine sequences. Our results collectively suggest that oxazinomycin functions as a Trojan horse substrate and its inhibitory effect is attributable to the oxygen atom in the position corresponding to carbon five of the uracil ring.


Subject(s)
DNA-Directed RNA Polymerases/chemistry , RNA/chemistry , Transcription, Genetic/drug effects , Uridine/analogs & derivatives , DNA-Directed RNA Polymerases/genetics , Escherichia coli/genetics , Oxygen/chemistry , Pseudomonas/chemistry , RNA/genetics , RNA Cleavage/drug effects , Streptomyces/chemistry , Substrate Specificity , Thymidine/chemistry , Thymidine/genetics , Transcription, Genetic/genetics , Transcriptional Elongation Factors/genetics , Uracil/chemistry , Uridine/chemical synthesis , Uridine/chemistry , Uridine/pharmacology
5.
Sci Rep ; 9(1): 8935, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31222036

ABSTRACT

Pseudouridimycin (PUM), a selective inhibitor of bacterial RNA polymerase has been previously detected in microbial-extracts of two strains of Streptomyces species (strain ID38640 and ID38673). Here, we isolated PUM and its deoxygenated analogue desoxy-pseudouridimycin (dPUM) from Streptomyces albus DSM 40763, previously reported to produce the metabolite strepturidin (STU). The isolated compounds were characterized by HRMS and spectroscopic techniques and they selectively inhibited transcription by bacterial RNA polymerase as previously reported for PUM. In contrast, STU could not be detected in the cultures of S. albus DSM 40763. As the reported characteristics reported for STU are almost identical with that of PUM, the existence of STU was questioned. We further sequenced the genome of S. albus DSM 40763 and identified a gene cluster that contains orthologs of all PUM biosynthesis enzymes but lacks the enzymes that would conceivably allow biosynthesis of STU as an additional product.


Subject(s)
Anti-Infective Agents/chemistry , Nucleosides/analogs & derivatives , Nucleosides/chemistry , Streptomyces/chemistry , Anti-Infective Agents/isolation & purification , Anti-Infective Agents/pharmacology , Genes, Bacterial , Multigene Family , Nucleosides/isolation & purification , Nucleosides/pharmacology , Streptomyces/genetics
6.
ACS Chem Biol ; 12(6): 1472-1477, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28418235

ABSTRACT

Nucleoside antibiotics are a large class of pharmaceutically relevant chemical entities, which exhibit a broad spectrum of biological activities. Most nucleosides belong to the canonical N-nucleoside family, where the heterocyclic unit is connected to the carbohydrate through a carbon-nitrogen bond. However, atypical C-nucleosides were isolated from Streptomyces bacteria over 50 years ago, but the molecular basis for formation of these metabolites has been unknown. Here, we have sequenced the genome of S. showdoensis ATCC 15227 and identified the gene cluster responsible for showdomycin production. Key to the detection was the presence of sdmA, encoding an enzyme of the pseudouridine monophosphate glycosidase family, which could catalyze formation of the C-glycosidic bond. Sequence analysis revealed an unusual combination of biosynthetic genes, while inactivation and subsequent complementation of sdmA confirmed the involvement of the locus in showdomycin formation. The study provides the first steps toward generation of novel C-nucleosides by pathway engineering.


Subject(s)
Antibiotics, Antineoplastic/biosynthesis , Multigene Family , Showdomycin/biosynthesis , Streptomyces/genetics , Bacterial Proteins/genetics , Biocatalysis , Biosynthetic Pathways , Genome, Bacterial/genetics , Glycoside Hydrolases/genetics , Glycoside Hydrolases/physiology , Nucleosides , Sequence Analysis, DNA , Streptomyces/enzymology
7.
Chem Biol ; 15(10): 1046-57, 2008 Oct 20.
Article in English | MEDLINE | ID: mdl-18940666

ABSTRACT

Alnumycin is closely related to the benzoisochromanequinone (BIQ) polyketides such as actinorhodin. Exceptional structural features include differences in aglycone tailoring that result in the unique alnumycin chromophore and the existence of an unusual 4-hydroxymethyl-5-hydroxy-1,3-dioxan moiety. Cloning and sequencing of the alnumycin gene cluster from Streptomyces sp. CM020 revealed expected biosynthesis genes for polyketide assembly, but several genes encoding subsequent tailoring enzymes were highly atypical. Heterologous expression studies confirmed that all of the genes required for alnumycin biosynthesis resided within the sequenced clone. Inactivation of genes aln4 and aln5 showed that the mechanism of pyran ring formation differs from actinorhodin and granaticin pathways. Further inactivation studies identified two genes, alnA and alnB, involved in the synthesis and attachment of the dioxan moiety, and resulted in the production of the polyketide prealnumycin.


Subject(s)
Dioxanes/chemistry , Dioxanes/metabolism , Multigene Family/genetics , Naphthoquinones/chemistry , Naphthoquinones/metabolism , Pyrans/chemistry , Pyrans/metabolism , Cloning, Molecular , Gene Expression , Genome, Fungal/genetics , Macrolides/chemistry , Macrolides/metabolism , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Molecular Structure , Streptomyces/chemistry , Streptomyces/genetics , Streptomyces/metabolism
8.
Chembiochem ; 8(13): 1577-84, 2007 Sep 03.
Article in English | MEDLINE | ID: mdl-17654627

ABSTRACT

Genome-sequencing projects have revealed that Streptomyces bacteria have the genetic potential to produce considerably larger numbers of natural products than can be observed under standard laboratory conditions. Cryptic angucycline-type aromatic polyketide gene clusters are particularly abundant. Sequencing of two such clusters from Streptomyces sp. PGA64 and H021 revealed the presence of several open reading frames that could be involved in processing the basic angucyclic carbon skeleton. The pga gene cluster contains one putative FAD-dependant monooxygenase (pgaE) and a putatively bifunctional monooxygenase/short chain alcohol reductase (pgaM), whereas the cab cluster contains two similar monooxygenases (cabE and cabM) and an independent reductase (cabV). In this study we have reconstructed the biosynthetic pathways for aglycone synthesis by cloning and sequentially expressing the angucycline tailoring genes with genes required for the synthesis of the unmodified angucycline metabolite-UWM6-in Streptomyces lividans TK24. The expression studies unequivocally showed that, after the production of UWM6, the pathways proceed through the action of the similar monooxygenases PgaE and CabE, followed by reactions catalysed by PgaM and CabMV. Analysis of the metabolites produced revealed that addition of pgaE and cabE genes directs both pathways to a known shunt product, rabelomycin, whereas expression of all genes from a given pathway results in the production of the novel angucycline metabolites gaudimycin A and B. However, one of the end products is most probably further modified by endogenous S. lividans TK24 enzymes. These experiments demonstrate that genes that are either inactive or cryptic in their native host can be used as biosynthetic tools to generate new compounds.


Subject(s)
Anti-Bacterial Agents/metabolism , Biosynthetic Pathways , Genes, Bacterial , Mixed Function Oxygenases/metabolism , Quinones/metabolism , Streptomyces/genetics , Anthraquinones/chemistry , Anthraquinones/metabolism , Antioxidants/pharmacology , Chromatography, Liquid , Cloning, Molecular , Flavin-Adenine Dinucleotide/metabolism , Gene Expression Regulation , Macrolides/metabolism , Magnetic Resonance Spectroscopy , Mass Spectrometry , Multigene Family , Open Reading Frames , Oxygenases/metabolism , Phylogeny , Quinones/chemistry , Telomerase/antagonists & inhibitors
9.
Antimicrob Agents Chemother ; 47(4): 1291-6, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12654660

ABSTRACT

The biosynthesis pathways of two anthracyclines, nogalamycin and aclacinomycin, were directed toward angucyclines by using an angucycline-specific cyclase, pgaF, isolated from a silent antibiotic biosynthesis gene cluster. Addition of pgaF to a gene cassette that harbored the early biosynthesis genes of nogalamycin resulted in the production of two known angucyclinone metabolites, rabelomycin and its precursor, UWM6. Substrate flexibility of pgaF was demonstrated by replacement of the nogalamycin minimal polyketide synthase genes in the gene cassette with the equivalent aclacinomycin genes together with aknE2 and aknF, which specify the unusual propionate starter unit in aclacinomycin biosynthesis. This modification led to the production of a novel angucyclinone, MM2002, in which the expected ethyl side chain was incorporated into the fourth ring.


Subject(s)
Aclarubicin/analogs & derivatives , Aclarubicin/biosynthesis , Anti-Bacterial Agents/biosynthesis , Antibiotics, Antineoplastic/biosynthesis , Nogalamycin/biosynthesis , Streptomyces/metabolism , Genetic Engineering , Multigene Family
SELECTION OF CITATIONS
SEARCH DETAIL
...