Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Proc Natl Acad Sci U S A ; 112(32): 9866-71, 2015 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-26216966

RESUMEN

Bacterial secondary metabolic pathways are responsible for the biosynthesis of thousands of bioactive natural products. Many enzymes residing in these pathways have evolved to catalyze unusual chemical transformations, which is facilitated by an evolutionary pressure promoting chemical diversity. Such divergent enzyme evolution has been observed in S-adenosyl-L-methionine (SAM)-dependent methyltransferases involved in the biosynthesis of anthracycline anticancer antibiotics; whereas DnrK from the daunorubicin pathway is a canonical 4-O-methyltransferase, the closely related RdmB (52% sequence identity) from the rhodomycin pathways is an atypical 10-hydroxylase that requires SAM, a thiol reducing agent, and molecular oxygen for activity. Here, we have used extensive chimeragenesis to gain insight into the functional differentiation of RdmB and show that insertion of a single serine residue to DnrK is sufficient for introduction of the monooxygenation activity. The crystal structure of DnrK-Ser in complex with aclacinomycin T and S-adenosyl-L-homocysteine refined to 1.9-Å resolution revealed that the inserted serine S297 resides in an α-helical segment adjacent to the substrate, but in a manner where the side chain points away from the active site. Further experimental work indicated that the shift in activity is mediated by rotation of a preceding phenylalanine F296 toward the active site, which blocks a channel to the surface of the protein that is present in native DnrK. The channel is also closed in RdmB and may be important for monooxygenation in a solvent-free environment. Finally, we postulate that the hydroxylation ability of RdmB originates from a previously undetected 10-decarboxylation activity of DnrK.


Asunto(s)
Antraciclinas/metabolismo , Vías Biosintéticas , Evolución Molecular , Oxigenasas de Función Mixta/genética , S-Adenosilmetionina/metabolismo , Aclarubicina/química , Aclarubicina/metabolismo , Secuencia de Aminoácidos , Antraciclinas/química , Biocatálisis , Dominio Catalítico , Cromatografía Líquida de Alta Presión , Ingeniería Genética , Hidroxilación , Metiltransferasas/metabolismo , Oxigenasas de Función Mixta/química , Oxigenasas de Función Mixta/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Familia de Multigenes , Proteínas Mutantes/metabolismo , Filogenia , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Espectrometría de Masa por Ionización de Electrospray , Electricidad Estática
2.
Proc Natl Acad Sci U S A ; 110(4): 1291-6, 2013 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-23297194

RESUMEN

Alnumycin A is an exceptional aromatic polyketide that contains a carbohydrate-like 4'-hydroxy-5'-hydroxymethyl-2',7'-dioxane moiety attached to the aglycone via a carbon-carbon bond. Recently, we have identified the D-ribose-5-phosphate origin of the dioxane unit and demonstrated that AlnA and AlnB are responsible for the overall C-ribosylation reaction. Here, we provide direct evidence that AlnA is a natural C-glycosynthase, which catalyzes the attachment of D-ribose-5-phosphate to prealnumycin by formation of the C(8)-C(1') bond as demonstrated by the structure of the intermediate alnumycin P. This compound is subsequently dephosphorylated by AlnB, an enzyme of the haloacid dehalogenase superfamily. Structure determination of the native trimeric AlnA to 2.1-Å resolution revealed a highly globular fold encompassing an α/ß/α sandwich. The crystal structure of the complex with D-ribose-5-phosphate indicated that the phosphosugar is bound in the open-chain configuration. Identification of residues E29, K86, and K159 near the C-1 carbonyl of the ligand led us to propose that the carbon-carbon bond formation proceeds through a Michael-type addition. Determination of the crystal structure of the monomeric AlnB in the open conformation to 1.25-Å resolution showed that the protein consists of core and cap domains. Modeling of alnumycin P inside the cap domain positioned the phosphate group next to a Mg(2+) ion present at the junction of the domains. Mutagenesis data were consistent with the canonical reaction mechanism for this enzyme family revealing the importance of residues D15 and D17 for catalysis. The characterization of the prealnumycin C-ribosylation illustrates an alternative means for attachment of carbohydrates to natural products.


Asunto(s)
Antibacterianos/biosíntesis , Antibacterianos/química , Naftoquinonas/química , Naftoquinonas/metabolismo , Secuencia de Aminoácidos , Vías Biosintéticas , Catálisis , Cristalografía por Rayos X , Glicosilación , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Mutagénesis Sitio-Dirigida , Péptidos/química , Péptidos/genética , Péptidos/metabolismo , Policétidos/química , Policétidos/metabolismo , Ribosamonofosfatos/metabolismo , Homología de Secuencia de Aminoácido , Electricidad Estática
3.
Proc Natl Acad Sci U S A ; 109(16): 6024-9, 2012 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-22474343

RESUMEN

Carbohydrate moieties are important components of natural products, which are often imperative for the solubility and biological activity of the compounds. The aromatic polyketide alnumycin A contains an extraordinary sugar-like 4'-hydroxy-5'-hydroxymethyl-2',7'-dioxane moiety attached via a carbon-carbon bond to the aglycone. Here we have extensively investigated the biosynthesis of the dioxane unit through (13)C labeling studies, gene inactivation experiments and enzymatic synthesis. We show that AlnA and AlnB, members of the pseudouridine glycosidase and haloacid dehalogenase enzyme families, respectively, catalyze C-ribosylation conceivably through Michael-type addition of d-ribose-5-phosphate and dephosphorylation. The ribose moiety may be attached both in furanose (alnumycin C) and pyranose (alnumycin D) forms. The C(1')-C(2') bond of alnumycin C is subsequently cleaved and the ribose unit is rearranged into an unprecedented dioxolane (cis-bicyclo[3.3.0]-2',4',6'-trioxaoctan-3'ß-ol) structure present in alnumycin B. The reaction is catalyzed by Aln6, which belongs to a previously uncharacterized enzyme family. The conversion was accompanied with consumption of O(2) and formation of H(2)O(2), which allowed us to propose that the reaction may proceed via hydroxylation of C1' followed by retro-aldol cleavage and acetal formation. Interestingly, no cofactors could be detected and the reaction was also conducted in the presence of metal chelating agents. The last step is the conversion of alnumycin B into the final end-product alnumycin A catalyzed by Aln4, an NADPH-dependent aldo-keto reductase. This characterization of the dioxane biosynthetic pathway sets the basis for the utilization of C-C bound ribose, dioxolane and dioxane moieties in the generation of improved biologically active compounds.


Asunto(s)
Vías Biosintéticas , Carbohidratos/química , Dioxanos/química , Naftoquinonas/química , Proteínas Bacterianas/metabolismo , Carbono/química , Isótopos de Carbono , Dioxanos/metabolismo , Electroforesis en Gel de Poliacrilamida , Glicósido Hidrolasas/metabolismo , Peróxido de Hidrógeno/química , Peróxido de Hidrógeno/metabolismo , Hidrolasas/metabolismo , Hidroxilación , Espectroscopía de Resonancia Magnética , Estructura Molecular , Naftoquinonas/metabolismo , Oxígeno/química , Oxígeno/metabolismo , Seudouridina/metabolismo , Ribosa/química , Ribosa/metabolismo , Ribosamonofosfatos/química , Ribosamonofosfatos/metabolismo , Streptomyces/genética , Streptomyces/metabolismo
4.
ACS Synth Biol ; 13(5): 1523-1536, 2024 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-38662967

RESUMEN

Streptomyces spp. are "nature's antibiotic factories" that produce valuable bioactive metabolites, such as the cytotoxic anthracycline polyketides. While the anthracyclines have hundreds of natural and chemically synthesized analogues, much of the chemical diversity stems from enzymatic modifications to the saccharide chains and, to a lesser extent, from alterations to the core scaffold. Previous work has resulted in the generation of a BioBricks synthetic biology toolbox in Streptomyces coelicolor M1152ΔmatAB that could produce aklavinone, 9-epi-aklavinone, auramycinone, and nogalamycinone. In this work, we extended the platform to generate oxidatively modified analogues via two crucial strategies. (i) We swapped the ketoreductase and first-ring cyclase enzymes for the aromatase cyclase from the mithramycin biosynthetic pathway in our polyketide synthase (PKS) cassettes to generate 2-hydroxylated analogues. (ii) Next, we engineered several multioxygenase cassettes to catalyze 11-hydroxylation, 1-hydroxylation, 10-hydroxylation, 10-decarboxylation, and 4-hydroxyl regioisomerization. We also developed improved plasmid vectors and S. coelicolor M1152ΔmatAB expression hosts to produce anthracyclinones. This work sets the stage for the combinatorial biosynthesis of bespoke anthracyclines using recombinant Streptomyces spp. hosts.


Asunto(s)
Antraciclinas , Sintasas Poliquetidas , Streptomyces coelicolor , Sintasas Poliquetidas/metabolismo , Sintasas Poliquetidas/genética , Antraciclinas/metabolismo , Streptomyces coelicolor/metabolismo , Streptomyces coelicolor/genética , Streptomyces/metabolismo , Streptomyces/genética , Vías Biosintéticas/genética , Hidroxilación , Antibacterianos/biosíntesis , Antibacterianos/metabolismo , Antibacterianos/química
5.
Biochemistry ; 52(26): 4507-16, 2013 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-23731237

RESUMEN

Two functionally distinct homologous flavoprotein hydroxylases, PgaE and JadH, have been identified as branching points in the biosynthesis of the polyketide antibiotics gaudimycin C and jadomycin A, respectively. These evolutionarily related enzymes are both bifunctional and able to catalyze the same initial reaction, C-12 hydroxylation of the common angucyclinone intermediate prejadomycin. The enzymes diverge in their secondary activities, which include hydroxylation at C-12b by PgaE and dehydration at C-4a/C-12b by JadH. A further difference is that the C-12 hydroxylation is subject to substrate inhibition only in PgaE. Here we have identified regions associated with the C-12b hydroxylation in PgaE by extensive chimeragenesis, focusing on regions surrounding the active site. The results highlight the importance of a hairpin-ß motif near the dimer interface, with two nonconserved residues, P78 and I79 (corresponding to Q89 and F90, respectively, in JadH), and invariant residue H73 playing key roles. Kinetic characterization of PgaE variants demonstrates that the secondary C-12b hydroxylation and substrate inhibition by prejadomycin are likely to be interlinked. The crystal structure of the PgaE P78Q/I79F variant at 2.4 Å resolution confirms that the changes do not alter the conformation of the ß-strand secondary structure and that the side chains of these residues in effect point away from the active site toward the dimer interface. The results support a catalytic model for PgaE containing two binding modes for C-12 and C-12b hydroxylations, where binding of prejadomycin in the orientation for C-12b hydroxylation leads to substrate inhibition. The presence of an allosteric network is evident based on enzyme kinetics.


Asunto(s)
Antraquinonas/química , Cristalografía por Rayos X , Oxigenasas de Función Mixta/química , Poligalacturonasa/química , Streptomyces/enzimología , Dominio Catalítico , Evolución Molecular , Hidroxilación , Oxigenasas de Función Mixta/genética , Mutagénesis , Poligalacturonasa/antagonistas & inhibidores , Poligalacturonasa/genética , Conformación Proteica , Streptomyces/genética , Relación Estructura-Actividad , Especificidad por Sustrato
6.
Biochemistry ; 52(31): 5304-14, 2013 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-23848284

RESUMEN

Angucyclines are biologically active natural products constructed around a common benz[a]anthraquinone carbon frame. One key branching point in the biosynthesis of angucyclines is the ketoreduction at C-6, which results in the opposite stereochemistry of landomycins and urdamycins/gaudimycins. Here we present the 1.65 Å resolution crystal structure of LanV from Streptomyces cyanogenus S136 that is responsible for the 6R stereochemistry of landomycins. The enzyme displays the common architectural fold of short-chain alcohol dehydrogenases/reductases and contains bound nicotinamide adenine dinucleotide phosphate. Determination of the structure of LanV in complex with 11-deoxylandomycinone at 2.0 Å resolution indicated that substrate binding does not induce large conformational changes and that substrate recognition occurs mainly through hydrophobic interactions. Analysis of the electron density map of the ternary complex revealed that the catalytic reaction had most likely proceeded backward in the crystal, because the data could be best fit with a compound harboring a carbonyl group at C-6. A coordinated water molecule was atypically identified between the ligand and the conserved Tyr160 residue, which was confirmed to be critical for the catalytic activity by site-directed mutagenesis. A catalytic triad of Ser147, Tyr160, and Lys164 could be recognized on the basis of the crystal structure, and stereoselective labeling studies demonstrated that the transfer of hydride from reduced nicotinamide adenine dinucleotide phosphate to the substrate occurs from the 4-pro-S side of the cosubstrate. Importantly, Ser192 was identified as being involved in controlling the stereochemistry of the reaction, as assays with single mutant Ser192Ile led to accumulation of gaudimycin C with 6S stereochemistry as a minor product.


Asunto(s)
Aminoglicósidos/biosíntesis , Antraquinonas/química , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Glicosiltransferasas/química , Glicosiltransferasas/metabolismo , Streptomyces/enzimología , Secuencias de Aminoácidos , Aminoglicósidos/química , Antraquinonas/metabolismo , Proteínas Bacterianas/genética , Glicosiltransferasas/genética , Estructura Molecular , Streptomyces/química , Streptomyces/genética , Especificidad por Sustrato
7.
PNAS Nexus ; 2(2): pgad009, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36874276

RESUMEN

Streptomyces soil bacteria produce hundreds of anthracycline anticancer agents with a relatively conserved set of genes. This diversity depends on the rapid evolution of biosynthetic enzymes to acquire novel functionalities. Previous work has identified S-adenosyl-l-methionine-dependent methyltransferase-like proteins that catalyze 4-O-methylation, 10-decarboxylation, or 10-hydroxylation, with additional differences in substrate specificities. Here we focused on four protein regions to generate chimeric enzymes using sequences from four distinct subfamilies to elucidate their influence in catalysis. Combined with structural studies we managed to depict factors that influence gain-of-hydroxylation, loss-of-methylation, and substrate selection. The engineering expanded the catalytic repertoire to include novel 9,10-elimination activity, and 4-O-methylation and 10-decarboxylation of unnatural substrates. The work provides an instructive account on how the rise of diversity of microbial natural products may occur through subtle changes in biosynthetic enzymes.

8.
ACS Chem Biol ; 18(4): 794-802, 2023 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-37005433

RESUMEN

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.


Asunto(s)
Nucleósidos , Seudouridina , Vías Biosintéticas , ARN Polimerasas Dirigidas por ADN/metabolismo , Nucleósidos/metabolismo , Seudouridina/biosíntesis , Seudouridina/metabolismo , Fosfato de Piridoxal/química , Streptomyces/química , Streptomyces/metabolismo
9.
J Bacteriol ; 194(11): 2829-36, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22467789

RESUMEN

Alnumycin A is an aromatic polyketide with a strong resemblance to related benzoisochromanequinone (BIQ) antibiotics, such as the model antibiotic actinorhodin. One intriguing difference between these metabolites is that the positions of the benzene and quinone rings are reversed in alnumycin A in comparison to the BIQ polyketides. In this paper we demonstrate that inactivation of either the monooxygenase alnT gene or the flavin reductase alnH gene results in the accumulation of a novel nonquinoid metabolite, thalnumycin A (ThA), in the culture medium. Additionally, two other previously characterized metabolites, K1115 A and 1,6-dihydroxy-8-propylanthraquinone (DHPA), were identified, which had oxidized into quinones putatively nonenzymatically at the incorrect position in the central ring. None of the compounds isolated contained correctly formed pyran rings, which suggests that on the alnumycin pathway quinone biosynthesis occurs prior to third ring cyclization. The regiochemistry of the two-component monooxygenase system AlnT/AlnH was finally confirmed in vitro by using ThA, FMN, and NADH in enzymatic synthesis, where the reaction product, thalnumycin B (ThB), was verified to contain the expected p-hydroquinone structure in the lateral ring.


Asunto(s)
Proteínas Bacterianas/metabolismo , Oxigenasas de Función Mixta/metabolismo , Naftoquinonas/metabolismo , Quinonas/metabolismo , Streptomyces/enzimología , Proteínas Bacterianas/genética , Vías Biosintéticas , Ciclización , Oxigenasas de Función Mixta/genética , Estructura Molecular , Naftoquinonas/química , Oxidación-Reducción , Quinonas/química , Streptomyces/genética , Streptomyces/metabolismo
10.
Biochemistry ; 50(24): 5535-43, 2011 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-21595438

RESUMEN

A simplified model system composed of a NADPH-dependent flavoprotein hydroxylase PgaE and a short-chain alcohol dehydrogenase/reductase (SDR) CabV was used to dissect a multistep angucycline modification redox cascade into several subreactions in vitro. We demonstrate that the two enzymes are sufficient for the conversion of angucycline substrate 2,3-dehydro-UWM6 to gaudimycin C. The flavoenzyme PgaE is shown to be responsible for two consecutive NADPH- and O(2)-dependent reactions, consistent with the enzyme-catalyzed incorporation of oxygen atoms at C-12 and C-12b in gaudimycin C. The two reactions do not significantly overlap, and the second catalytic cycle is initiated only after the original substrate 2,3-dehydro-UWM6 is nearly depleted. This allowed us to isolate the product of the first reaction at limiting NADPH concentrations and allowed the study of the qualitative and kinetic properties of the separated reactions. Dissection of the reaction cascade also allowed us to establish that the SDR reductase CabV catalyzes the final biosynthetic step, which is closely coupled to the second PgaE reaction. In the absence of CabV, the complete PgaE reaction leads invariably to product degradation, whereas in its presence, the reaction yields the final product, gaudimycin C. The result implies that the C-6 ketoreduction step catalyzed by CabV is required for stabilization of a reactive intermediate. The close relationship between PgaE and CabV would explain previous in vivo observations: why the absence of a reductase gene may result in the lack of C-12b-oxygenated species and, vice versa, why all C-12b-oxygenated angucyclines appear to have undergone reduction at position C-6.


Asunto(s)
Antraquinonas/metabolismo , Oxigenasas de Función Mixta/metabolismo , Alcohol Deshidrogenasa/metabolismo , Oxidorreductasas de Alcohol/metabolismo , Secuencia de Bases , ADN Bacteriano/genética , Flavoproteínas/metabolismo , Cinética , Oxígeno/metabolismo , Streptomyces/genética , Streptomyces/metabolismo , Especificidad por Sustrato
11.
Biochemistry ; 49(5): 934-44, 2010 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-20052967

RESUMEN

SnoaB is a cofactor-independent monooxygenase that catalyzes the conversion of 12-deoxynogalonic acid to nogalonic acid in the biosynthesis of the aromatic polyketide nogalamycin in Streptomyces nogalater. In vitro (18)O(2) experiments establish that the oxygen atom incorporated into the substrate is derived from molecular oxygen. The crystal structure of the enzyme was determined in two different space groups to 1.7 and 1.9 A resolution, respectively. The enzyme displays the ferredoxin fold, with the characteristic beta-strand exchange at the dimer interface. The crystal structures reveal a putative catalytic triad involving two asparagine residues, Asn18 and Asn63, and a water molecule, which may play important roles in the enzymatic reaction. Site-directed mutagenesis experiments, replacing the two asparagines individually by alanine, led to a 100-fold drop in enzymatic activity. Replacement of an invariant tryptophan residue in the active site of the enzyme by phenylalanine also resulted in an enzyme variant with about 1% residual activity. Taken together, our findings are most consistent with a carbanion mechanism where the deprotonated substrate reacts with molecular oxygen via one electron transfer and formation of a caged radical.


Asunto(s)
Coenzimas/química , Oxigenasas de Función Mixta/química , Oxigenasas de Función Mixta/metabolismo , Nogalamicina/biosíntesis , Streptomyces/enzimología , Secuencia de Aminoácidos , Catálisis , Cristalografía por Rayos X , Estabilidad de Enzimas , Cinética , Macrólidos/química , Oxigenasas de Función Mixta/genética , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Nogalamicina/química , Streptomyces/genética , Especificidad por Sustrato
12.
Org Biomol Chem ; 8(4): 886-95, 2010 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-20135048

RESUMEN

Various commercial lyophilized and immobilized preparations of lipase A from Candida antarctica (CAL-A) were studied for their ability to catalyze the hydrolysis of amide bonds in N-acylated alpha-amino acids, 3-butanamidobutanoic acid (beta-amino acid) and its ethyl ester. The activity toward amide bonds is highly untypical of lipases, despite the close mechanistic analogy to amidases which normally catalyze the corresponding reactions. Most CAL-A preparations cleaved amide bonds of various substrates with high enantioselectivity, although high variations in substrate selectivity and catalytic rates were detected. The possible role of contaminant protein species on the hydrolytic activity toward these bonds was studied by fractionation and analysis of the commercial lyophilized preparation of CAL-A (Cat#ICR-112, Codexis). In addition to minor impurities, two equally abundant proteins were detected, migrating on SDS-PAGE a few kDa apart around the calculated size of CAL-A. Based on peptide fragment analysis and sequence comparison both bands shared substantial sequence coverage with CAL-A. However, peptides at the C-terminal end constituting a motile domain described as an active-site flap were not identified in the smaller fragment. Separated gel filtration fractions of the two forms of CAL-A both catalyzed the amide bond hydrolysis of ethyl 3-butanamidobutanoate as well as the N-acylation of methyl pipecolinate. Hydrolytic activity towards N-acetylmethionine was, however, solely confined to the fractions containing the truncated form of CAL-A. These fractions were also found to contain a trace enzyme impurity identified in sequence analysis as a serine carboxypeptidase. The possible role of catalytic impurities versus the function of CAL-A in amide bond hydrolysis is further discussed in the paper.


Asunto(s)
Amidas/metabolismo , Candida/enzimología , Carboxipeptidasas/metabolismo , Lipasa/metabolismo , Metionina/análogos & derivados , Acilación , Sitios de Unión , Catálisis , Electroforesis en Gel de Poliacrilamida/métodos , Hidrólisis , Metionina/química , Modelos Químicos , Modelos Moleculares , Especificidad por Sustrato
13.
Chem Biol ; 15(2): 157-66, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18291320

RESUMEN

Tailoring steps in aromatic polyketide antibiotic biosynthesis are an important source of structural diversity and, consequently, an intriguing focal point for enzymological studies. PgaE and PgaM from Streptomyces sp. PGA64 are representatives of flavoenzymes catalyzing early post-PKS reactions in angucycline biosynthesis. This in vitro study illustrates that the chemoenzymatic conversion of UWM6 into the metabolite, gaudimycin C, requires multiple closely coupled reactions to prevent intermediate degradation. The NMR structure of gaudimycin C confirms that the reaction cascade involves C12- and C12b-hydroxylation, C2,3-dehydration, and stereospecific ketoreduction at C6. Enzymatic 18O incorporation studies verify that the oxygens at C12 and C12b derive from O2 and H2O, respectively. The results indicate that PgaM deviates mechanistically from flavoprotein monooxygenases, and suggest an alternative catalytic mechanism involving a quinone methide intermediate.


Asunto(s)
Antraciclinas/metabolismo , Antraquinonas/metabolismo , Antibacterianos/biosíntesis , Flavoproteínas/metabolismo , Oxigenasas de Función Mixta/metabolismo , Streptomyces/enzimología , Antraquinonas/química , Espectroscopía de Resonancia Magnética , Oxígeno/metabolismo , Agua/metabolismo
14.
Chem Biol ; 15(10): 1046-57, 2008 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-18940666

RESUMEN

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.


Asunto(s)
Dioxanos/química , Dioxanos/metabolismo , Familia de Multigenes/genética , Naftoquinonas/química , Naftoquinonas/metabolismo , Piranos/química , Piranos/metabolismo , Clonación Molecular , Expresión Génica , Genoma Fúngico/genética , Macrólidos/química , Macrólidos/metabolismo , Espectroscopía de Resonancia Magnética , Datos de Secuencia Molecular , Estructura Molecular , Streptomyces/química , Streptomyces/genética , Streptomyces/metabolismo
15.
Artículo en Inglés | MEDLINE | ID: mdl-19255477

RESUMEN

12-deoxy-nogalonic acid oxygenase (SnoaB) catalyzes the oxygenation of 12-deoxy-nogalonic acid at position 12 to yield nogalonic acid, which is one of the steps in the biosynthesis of the polyketide nogalamycin in Streptomyces nogalater. SnoaB belongs to a family of small cofactor-free oxygenases which carry out oxygenation reactions without the aid of any prosthetic group, cofactor or metal ion. Recombinant SnoaB was crystallized in space group P2(1)2(1)2, with unit-cell parameters a = 58.8, b = 114.1, c = 49.5 A, and these crystals diffracted to 2.4 A resolution. Recombinant SnoaB does not contain any methionine residues and three double mutants were designed and produced for the preparation of selenomethionine-substituted samples. The selenomethionine-substituted mutant F40M/L89M crystallized in the same space group as the native enzyme.


Asunto(s)
Coenzimas/metabolismo , Oxigenasas de Función Mixta/aislamiento & purificación , Oxigenasas de Función Mixta/metabolismo , Nogalamicina/biosíntesis , Streptomyces/enzimología , Secuencia de Aminoácidos , Catálisis , Cristalización , Cristalografía por Rayos X , Oxigenasas de Función Mixta/química , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Nogalamicina/química , Selenometionina/metabolismo , Alineación de Secuencia
16.
ACS Chem Biol ; 14(5): 850-856, 2019 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-30995392

RESUMEN

Microbial natural products are an important source of chemical entities for drug discovery. Recent advances in understanding the biosynthesis of secondary metabolites has revealed how this rich chemical diversity is generated through functional differentiation of biosynthetic enzymes. For instance, investigations into anthracycline anticancer agents have uncovered distinct S-adenosyl methionine (SAM)-dependent proteins: DnrK is a 4-O-methyltransferase involved in daunorubicin biosynthesis, whereas RdmB (52% sequence identity) from the rhodomycin pathway catalyzes 10-hydroxylation. Here, we have mined unknown anthracycline gene clusters and discovered a third protein subclass catalyzing 10-decarboxylation. Subsequent isolation of komodoquinone B from two Streptomyces strains verified the biological relevance of the decarboxylation activity. Phylogenetic analysis inferred two independent routes for the conversion of methyltransferases into hydroxylases, with a two-step process involving loss-of-methylation and gain-of-hydroxylation presented here. Finally, we show that simultaneously with the functional differentiation, the evolutionary process has led to alterations in substrate specificities.


Asunto(s)
Antraciclinas/metabolismo , Evolución Biológica , Metiltransferasas/metabolismo , Genes Bacterianos , Filogenia , S-Adenosilmetionina/metabolismo , Streptomyces/enzimología , Streptomyces/genética , Streptomyces/metabolismo , Especificidad por Sustrato
17.
Sci Rep ; 9(1): 8935, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31222036

RESUMEN

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.


Asunto(s)
Antiinfecciosos/química , Nucleósidos/análogos & derivados , Nucleósidos/química , Streptomyces/química , Antiinfecciosos/aislamiento & purificación , Antiinfecciosos/farmacología , Genes Bacterianos , Familia de Multigenes , Nucleósidos/aislamiento & purificación , Nucleósidos/farmacología , Streptomyces/genética
18.
J Mol Biol ; 372(3): 633-48, 2007 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-17669423

RESUMEN

Angucyclines are aromatic polyketides produced in Streptomycetes via complex enzymatic biosynthetic pathways. PgaE and CabE from S. sp PGA64 and S. sp. H021 are two related homo-dimeric FAD and NADPH dependent aromatic hydroxylases involved in the early steps of the angucycline core modification. Here we report the three-dimensional structures of these two enzymes determined by X-ray crystallography using multiple anomalous diffraction and molecular replacement, respectively, to resolutions of 1.8 A and 2.7 A. The enzyme subunits are built up of three domains, a FAD binding domain, a domain involved in substrate binding and a C-terminal thioredoxin-like domain of unknown function. The structure analysis identifies PgaE and CabE as members of the para-hydroxybenzoate hydroxylase (pHBH) fold family of aromatic hydroxylases. In contrast to phenol hydroxylase and 3-hydroxybenzoate hydroxylase that utilize the C-terminal domain for dimer formation, this domain is not part of the subunit-subunit interface in PgaE and CabE. Instead, dimer assembly occurs through interactions of their FAD binding domains. FAD is bound non-covalently in the "in"-conformation. The active sites in the two enzymes differ significantly from those of other aromatic hydroxylases. The volumes of the active site are significantly larger, as expected in view of the voluminous tetracyclic angucycline substrates. The structures further suggest that substrate binding and catalysis may involve dynamic rearrangements of the middle domain relative to the other two domains. Site-directed mutagenesis studies of putative catalytic groups in the active site of PgaE argue against enzyme-catalyzed substrate deprotonation as a step in catalysis. This is in contrast to pHBH, where deprotonation/protonation of the substrate has been suggested as an essential part of the enzymatic mechanism.


Asunto(s)
Oxigenasas de Función Mixta/química , Compuestos Policíclicos/metabolismo , Streptomyces/enzimología , 4-Hidroxibenzoato-3-Monooxigenasa/química , Secuencia de Aminoácidos , Sitios de Unión , Catálisis , Cristalografía por Rayos X , Flavina-Adenina Dinucleótido/metabolismo , Oxigenasas de Función Mixta/aislamiento & purificación , Modelos Moleculares , Datos de Secuencia Molecular , Oxidación-Reducción , Compuestos Policíclicos/química , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Subunidades de Proteína/química , Alineación de Secuencia , Electricidad Estática , Especificidad por Sustrato
19.
J Mol Biol ; 357(1): 210-20, 2006 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-16414075

RESUMEN

AknH is a small polyketide cyclase that catalyses the closure of the fourth carbon ring in aclacinomycin biosynthesis in Streptomyces galilaeus, converting aklanonic acid methyl ester to aklaviketone. The crystal structure analysis of this enzyme, in complex with substrate and product analogue, showed that it is closely related in fold and mechanism to the polyketide cyclase SnoaL that catalyses the corresponding reaction in the biosynthesis of nogalamycin. Similarity is also apparent at a functional level as AknH can convert nogalonic acid methyl ester, the natural substrate of SnoaL, to auraviketone in vitro and in constructs in vivo. Despite the conserved structural and mechanistic features between these enzymes, the reaction products of AknH and SnoaL are stereochemically distinct. Supplied with the same substrate, AknH yields a C9-R product, like most members of this family of polyketide cyclases, whereas the product of SnoaL has the opposite C9-S stereochemistry. A comparison of high-resolution crystal structures of the two enzymes combined with in vitro mutagenesis studies revealed two critical amino acid substitutions in the active sites, which contribute to product stereoselectivity in AknH. Replacement of residues Tyr15 and Asn51 of AknH, located in the vicinity of the main catalytic residue Asp121, by their SnoaL counter-parts phenylalanine and leucine, respectively, results in a complete loss of product stereoselectivity.


Asunto(s)
Proteínas Bacterianas/química , Isomerasas/química , Estructura Terciaria de Proteína , Streptomyces/enzimología , Aclarubicina/análogos & derivados , Aclarubicina/biosíntesis , Aclarubicina/química , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Catálisis , Cristalografía por Rayos X , Isomerasas/genética , Isomerasas/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Mutagénesis Sitio-Dirigida , Nogalamicina/biosíntesis , Nogalamicina/química , Unión Proteica , Estructura Cuaternaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Especificidad por Sustrato
20.
J Mol Biol ; 359(3): 728-40, 2006 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-16650858

RESUMEN

SnoaL2 and AclR are homologous enzymes in the biosynthesis of the aromatic polyketides nogalamycin in Streptomyces nogalater and cinerubin in Streptomyces galilaeus, respectively. Evidence obtained from gene transfer experiments suggested that SnoaL2 catalyzes the hydroxylation of the C-1 carbon atom of the polyketide chain. Here we show that AclR is also involved in the production of 1-hydroxylated anthracyclines in vivo. The three-dimensional structure of SnoaL2 has been determined by multi-wavelength anomalous diffraction to 2.5A resolution, and that of AclR to 1.8A resolution using molecular replacement. Both enzymes are dimers in solution and in the crystal. The fold of the enzyme subunits consists of an alpha+beta barrel. The dimer interface is formed by packing of the beta-sheets from the two subunits against each other. In the interior of the alpha+beta barrel a hydrophobic cavity is formed that most likely binds the substrate and harbors the active site. The subunit fold and the architecture of the active site in SnoaL2 and AclR are similar to that of the polyketide cyclases SnoaL and AknH; however, they show completely different quaternary structures. A comparison of the active site pockets of the putative hydroxylases AclR and SnoaL2 with those of bona fide polyketide cyclases reveals distinct differences in amino acids lining the cavity that might be responsible for the switch in chemistry. The moderate degree of sequence similarity and the preservation of the three-dimensional fold of the polypeptide chain suggest that these enzymes are evolutionary related. Members of this enzyme family appear to have evolved from a common protein scaffold by divergent evolution to catalyze reactions chemically as diverse as aldol condensation and hydroxylation.


Asunto(s)
Antraciclinas/metabolismo , Antibacterianos/biosíntesis , Proteínas Bacterianas/química , Isomerasas/química , Oxigenasas de Función Mixta/metabolismo , Modelos Moleculares , Streptomyces/enzimología , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Dimerización , Isomerasas/genética , Isomerasas/metabolismo , Oxigenasas de Función Mixta/química , Oxigenasas de Función Mixta/genética , Datos de Secuencia Molecular , Mutación , Nogalamicina/biosíntesis , Estructura Cuaternaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA