Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
ACS Synth Biol ; 13(5): 1523-1536, 2024 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-38662967

RESUMO

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.


Assuntos
Antraciclinas , Policetídeo Sintases , Streptomyces coelicolor , Policetídeo Sintases/metabolismo , Policetídeo Sintases/genética , Antraciclinas/metabolismo , Streptomyces coelicolor/metabolismo , Streptomyces coelicolor/genética , Streptomyces/metabolismo , Streptomyces/genética , Vias Biossintéticas/genética , Hidroxilação , Antibacterianos/biossíntese , Antibacterianos/metabolismo , Antibacterianos/química
2.
PNAS Nexus ; 2(2): pgad009, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36874276

RESUMO

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.

3.
ACS Chem Biol ; 14(5): 850-856, 2019 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-30995392

RESUMO

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.


Assuntos
Antraciclinas/metabolismo , Evolução Biológica , Metiltransferases/metabolismo , Genes Bacterianos , Filogenia , S-Adenosilmetionina/metabolismo , Streptomyces/enzimologia , Streptomyces/genética , Streptomyces/metabolismo , Especificidade por Substrato
4.
ACS Chem Biol ; 12(6): 1472-1477, 2017 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-28418235

RESUMO

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.


Assuntos
Antibióticos Antineoplásicos/biossíntese , Família Multigênica , Showdomicina/biossíntese , Streptomyces/genética , Proteínas de Bactérias/genética , Biocatálise , Vias Biossintéticas , Genoma Bacteriano/genética , Glicosídeo Hidrolases/genética , Glicosídeo Hidrolases/fisiologia , Nucleosídeos , Análise de Sequência de DNA , Streptomyces/enzimologia
5.
Proc Natl Acad Sci U S A ; 112(32): 9866-71, 2015 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-26216966

RESUMO

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.


Assuntos
Antraciclinas/metabolismo , Vias Biossintéticas , Evolução Molecular , Oxigenases de Função Mista/genética , S-Adenosilmetionina/metabolismo , Aclarubicina/química , Aclarubicina/metabolismo , Sequência de Aminoácidos , Antraciclinas/química , Biocatálise , Domínio Catalítico , Cromatografia Líquida de Alta Pressão , Engenharia Genética , Hidroxilação , Metiltransferases/metabolismo , Oxigenases de Função Mista/química , Oxigenases de Função Mista/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Família Multigênica , Proteínas Mutantes/metabolismo , Filogenia , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Espectrometria de Massas por Ionização por Electrospray , Eletricidade Estática
6.
Proc Natl Acad Sci U S A ; 110(4): 1291-6, 2013 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-23297194

RESUMO

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.


Assuntos
Antibacterianos/biossíntese , Antibacterianos/química , Naftoquinonas/química , Naftoquinonas/metabolismo , Sequência de Aminoácidos , Vias Biossintéticas , Catálise , Cristalografia por Raios X , Glicosilação , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Mutagênese Sítio-Dirigida , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Policetídeos/química , Policetídeos/metabolismo , Ribosemonofosfatos/metabolismo , Homologia de Sequência de Aminoácidos , Eletricidade Estática
7.
J Mol Biol ; 375(5): 1212-21, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18076902

RESUMO

The gene pgaM is involved in the biosynthesis of an angucycline-type polyketide antibiotic in Streptomyces sp. PGA64. It encodes a two-domain polypeptide consisting of an N-terminal flavoprotein oxygenase and a C-terminal short-chain alcohol dehydrogenase/reductase, which are fused together at the translational level as a result of end codon deletion. Here we show that translation also initiates at an internal start codon that enables independent expression of a separate reductase subunit, PgaMred. This confirms that the gene exhibits a rare viral-like arrangement of two overlapping reading frames that allows simultaneous expression of two alternative forms of the protein. Together, these two proteins associate to form a stable non-covalent complex, the native form of PgaM. The reductase subunit PgaMred is shown to provide enzyme stability and to affect the redox state of the oxygenase domain FAD. Finally, a model for the quaternary structure of the complex that explains the necessity for a nested gene system and the unusual behaviour of the protein subunits in vitro is presented.


Assuntos
Genes Bacterianos , Genes Inseridos , Estrutura Quaternária de Proteína/genética , Proteínas/genética , Streptomyces/genética , Álcool Desidrogenase/química , Sequência de Aminoácidos , Sequência de Bases , Catálise , Códon de Iniciação , Dimerização , Escherichia coli/genética , Isoenzimas , Oxigenases de Função Mista/química , Modelos Moleculares , Dados de Sequência Molecular , Família Multigênica , Fases de Leitura Aberta , Oxirredução , Plasmídeos , Policetídeo Sintases/biossíntese , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Análise de Sequência de Proteína
8.
Proc Natl Acad Sci U S A ; 104(15): 6170-5, 2007 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-17395717

RESUMO

Aclacinomycin (Acl) oxidoreductase (AknOx) catalyzes the last two steps in the biosynthesis of polyketide antibiotics of the Acl group, the oxidation of the terminal sugar moiety rhodinose to l-aculose. We present the crystal structure of AknOx with bound FAD and the product AclY, refined to 1.65-A resolution. The overall fold of AknOx identifies the enzyme as a member of the p-cresol methylhydroxylase superfamily. The cofactor is bicovalently attached to His-70 and Cys-130 as 8alpha-Ndelta1-histidyl, 6-S-cysteinyl FAD. The polyketide ligand is bound in a deep cleft in the substrate-binding domain, with the tetracyclic ring system close to the enzyme surface and the three-sugar chain extending into the protein interior. The terminal sugar residue packs against the isoalloxazine ring of FAD and positions the carbon atoms that are oxidized close to the N5 atom of FAD. The structure and site-directed mutagenesis data presented here are consistent with a mechanism where the two different reactions of AknOx are catalyzed in the same active site but by different active site residues. Tyr-450 is responsible for proton removal from the C-4 hydroxyl group in the first reaction, the oxidation of rhodinose to cinerulose A. Tyr-378 acts as a catalytic base involved in proton abstraction from C3 of cinerulose A in the second reaction, for formation L-aculose. Replacement of this residue, however, does not impair the conversion of rhodinose to cinerulose A.


Assuntos
Aclarubicina/análogos & derivados , Oxigenases de Função Mista/química , Oxigenases de Função Mista/genética , Modelos Moleculares , Streptomyces/enzimologia , Aclarubicina/biossíntese , Sequência de Aminoácidos , Catálise , Biologia Computacional , Cristalografia , Escherichia coli , Dados de Sequência Molecular , Estrutura Molecular , Monossacarídeos/metabolismo , Mutagênese Sítio-Dirigida , Conformação Proteica
9.
J Mol Biol ; 359(3): 728-40, 2006 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-16650858

RESUMO

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.


Assuntos
Antraciclinas/metabolismo , Antibacterianos/biossíntese , Proteínas de Bactérias/química , Isomerases/química , Oxigenases de Função Mista/metabolismo , Modelos Moleculares , Streptomyces/enzimologia , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Dimerização , Isomerases/genética , Isomerases/metabolismo , Oxigenases de Função Mista/química , Oxigenases de Função Mista/genética , Dados de Sequência Molecular , Mutação , Nogalamicina/biossíntese , Estrutura Quaternária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
10.
J Biol Chem ; 280(5): 3636-44, 2005 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-15548527

RESUMO

Aclacinomycin 10-hydroxylase is a methyltransferase homologue that catalyzes a S-adenosyl-L-methionine (AdoMet)-dependent hydroxylation of the C-10 carbon atom of 15-demethoxy-epsilon-rhodomycin, a step in the biosynthesis of the polyketide antibiotic beta-rhodomycin. S-Adenosyl-L-homocysteine is an inhibitor of the enzyme, whereas the AdoMet analogue sinefungin can act as cofactor, indicating that a positive charge is required for catalysis. 18O2 experiments show that the hydroxyl group is derived from molecular oxygen. The reaction further requires thiol reagents such as glutathione or dithiothreitol. Incubation of the enzyme with substrate in the absence of reductant leads to the accumulation of an intermediate with a molecular mass consistent with a perhydroxy compound. This intermediate is turned into product upon addition of glutathione. The crystal structure of an abortive enzyme-AdoMet product ternary complex reveals large conformational changes consisting of a domain rotation leading to active site closure upon binding of the anthracycline ligand. The data suggest a mechanism where decarboxylation of the substrate results in the formation of a carbanion intermediate, which is stabilized by resonance through the aromatic ring system of the anthracycline substrate. The delocalization of the electrons is facilitated by the positive charge of the cofactor AdoMet. The activation of oxygen and formation of a hydroxyperoxide intermediate occurs in a manner similar to that observed in flavoenzymes. Aclacinomycin-10-hydroxylase is the first example of a AdoMet-dependent hydroxylation reaction, a novel function for this cofactor. The enzyme lacks methyltransferase activity due to the positioning of the AdoMet methyl group unfavorable for a SN2-type methyl transfer to the substrate.


Assuntos
Adenosina/análogos & derivados , Oxigenases de Função Mista/química , Oxigenases de Função Mista/metabolismo , S-Adenosilmetionina/metabolismo , Streptomyces/enzimologia , Adenosina/química , Antraciclinas/análise , Antraciclinas/metabolismo , Antimaláricos/química , Cromatografia Líquida de Alta Pressão , Cromatografia Líquida , Coenzimas/metabolismo , Hidroxilação , Espectrometria de Massas , Oxigenases de Função Mista/genética , Mutagênese , Oxigênio/metabolismo , Estrutura Terciária de Proteína , Especificidade por Substrato
11.
J Biol Chem ; 279(39): 41149-56, 2004 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-15273252

RESUMO

One of the final steps in the biosynthesis of the widely used anti-tumor drug daunorubicin in Streptomyces peucetius is the methylation of the 4-hydroxyl group of the tetracyclic ring system. This reaction is catalyzed by the S-adenosyl-L-methionine-dependent carminomycin 4-O-methyltransferase DnrK. The crystal structure of the ternary complex of this enzyme with the bound products S-adenosyl-L-homocysteine and 4-methoxy-epsilon-rhodomycin T has been determined to a 2.35-angstroms resolution. DnrK is a homodimer, and the subunit displays the typical fold of small molecule O-methyltransferases. The structure provides insights into the recognition of the anthracycline substrate and also suggests conformational changes as part of the catalytic cycle of the enzyme. The position and orientation of the bound ligands are consistent with an SN2 mechanism of methyl transfer. Mutagenesis experiments on a putative catalytic base confirm that DnrK most likely acts as an entropic enzyme in that rate enhancement is mainly due to orientational and proximity effects. This contrasts the mechanism of DnrK with that of other O-methyltransferases where acid/base catalysis has been demonstrated to be an essential contribution to rate enhancement.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Proteínas de Bactérias/química , Daunorrubicina/biossíntese , Metiltransferases/química , Streptomyces/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Catálise , Cromatografia Líquida de Alta Pressão , Cristalografia por Raios X , Dimerização , Elétrons , Histidina/química , Ligantes , Metilação , Modelos Químicos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese , Ligação Proteica , Conformação Proteica , Proteínas Recombinantes/química , Homologia de Sequência de Aminoácidos , Software
12.
Acta Crystallogr D Biol Crystallogr ; 60(Pt 6): 1118-20, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15159574

RESUMO

Nogalonic acid methyl ester cyclase (SnoaL) catalyzes the last ring-closure step in the biosynthesis of the polyketide antibiotic nogalamycin. Crystals of a complex of SnoaL with the substrate nogalonic acid methyl ester have been obtained using PEG 4000 as precipitant. The crystals are orthorhombic, space group I222, with unit-cell parameters a = 69.1, b = 72.0, c = 65.4 angstroms. They diffract to 1.35 angstroms resolution using synchrotron radiation. A Matthews coefficient of 2.0 angstroms3 Da(-1) suggests one subunit in the asymmetric unit. Diffraction data for an isomorphous uranium derivative were collected and a difference Patterson map showed strong peaks which allowed determination of the position of the uranium ions.


Assuntos
Cristalografia por Raios X/métodos , Isomerases/química , Nogalamicina/biossíntese , Streptomyces/enzimologia , Sequência de Aminoácidos , Antibióticos Antineoplásicos/química , Proteínas de Bactérias , Escherichia coli/metabolismo , Concentração de Íons de Hidrogênio , Íons , Modelos Químicos , Dados de Sequência Molecular , Polietilenoglicóis/química , Especificidade por Substrato , Síncrotrons , Urânio/química , Urânio/metabolismo
13.
EMBO J ; 23(9): 1911-21, 2004 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-15071504

RESUMO

SnoaL belongs to a family of small polyketide cyclases, which catalyse ring closure steps in the biosynthesis of polyketide antibiotics produced in Streptomyces. Several of these antibiotics are among the most used anti-cancer drugs currently in use. The crystal structure of SnoaL, involved in nogalamycin biosynthesis, with a bound product, has been determined to 1.35 A resolution. The fold of the subunit can be described as a distorted alpha+beta barrel, and the ligand is bound in the hydrophobic interior of the barrel. The 3D structure and site-directed mutagenesis experiments reveal that the mechanism of the intramolecular aldol condensation catalysed by SnoaL is different from that of the classical aldolases, which employ covalent Schiff base formation or a metal ion cofactor. The invariant residue Asp121 acts as an acid/base catalyst during the reaction. Stabilisation of the enol(ate) intermediate is mainly achieved by the delocalisation of the electron pair over the extended pi system of the substrate. These polyketide cyclases thus form of family of enzymes with a unique catalytic strategy for aldol condensation.


Assuntos
Aldeídos/metabolismo , Antibacterianos/biossíntese , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Isomerases/química , Isomerases/metabolismo , Modelos Moleculares , Nogalamicina/biossíntese , Streptomyces/enzimologia , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Biologia Computacional , Cristalografia por Raios X , Isomerases/genética , Espectrometria de Massas , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Oligonucleotídeos , Conformação Proteica , Alinhamento de Sequência
14.
J Mol Biol ; 334(2): 269-80, 2003 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-14607118

RESUMO

Anthracyclines are aromatic polyketide antibiotics, and several of these compounds are widely used as anti-tumor drugs in chemotherapy. Aclacinomycin-10-hydroxylase (RdmB) is one of the tailoring enzymes that modify the polyketide backbone in the biosynthesis of these metabolites. RdmB, a S-adenosyl-L-methionine-dependent methyltransferase homolog, catalyses the hydroxylation of 15-demethoxy-epsilon-rhodomycin to beta-rhodomycin, one step in rhodomycin biosynthesis in Streptomyces purpurascens. The crystal structure of RdmB, determined by multiwavelength anomalous diffraction to 2.1A resolution, reveals that the enzyme subunit has a fold similar to methyltransferases and binds S-adenosyl-L-methionine. The N-terminal domain, which consists almost exclusively of alpha-helices, is involved in dimerization. The C-terminal domain contains a typical alpha/beta nucleotide-binding fold, which binds S-adenosyl-L-methionine, and several of the residues interacting with the cofactor are conserved in O-methyltransferases. Adjacent to the S-adenosyl-L-methionine molecule there is a large cleft extending to the enzyme surface of sufficient size to bind the substrate. Analysis of the putative substrate-binding pocket suggests that there is no enzymatic group in proximity of the substrate 15-demethoxy-epsilon-rhodomycin, which could assist in proton abstraction and thus facilitate methyl transfer. The lack of a suitably positioned catalytic base might thus be one of the features responsible for the inability of the enzyme to act as a methyltransferase.


Assuntos
Antraciclinas/metabolismo , Oxigenases de Função Mista/química , S-Adenosilmetionina/metabolismo , Streptomyces/química , Sequência de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X , Oxigenases de Função Mista/metabolismo , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Streptomyces/metabolismo
15.
Acta Crystallogr D Biol Crystallogr ; 59(Pt 9): 1637-9, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12925797

RESUMO

Two enzymes participating in the biosynthesis of anthracyclines in Streptomyces purpurascens, aclacinomycin-10-methyl esterase (RdmC) and aclacinomycin-10-hydroxylase (RdmB), have been crystallized. RdmB is a S-adenosyl-methionine-dependent hydroxylase and RdmC hydrolyses the carboxymethyl group of the aglycone skeleton of aclacinomycin. Crystals of RdmB obtained in the presence of S-adenosyl-L-methionine were orthorhombic, space group C222(1), with unit-cell parameters a = 63.2, b = 92.2, c = 115.3 A; diffraction data were collected to 2.1 A resolution. RdmC was crystallized as a complex with the substrate, aclacinomycin T. These crystals diffracted to 1.45 A resolution and belonged to space group P2(1), with unit-cell parameters a = 38.2, b = 84.7, c = 44.3 A, beta = 99.9 degrees.


Assuntos
Hidrolases de Éster Carboxílico/química , Cristalização/métodos , Oxigenases de Função Mista/química , Streptomyces/enzimologia , Antraciclinas/metabolismo , Difração de Raios X
16.
FEMS Microbiol Lett ; 208(1): 117-22, 2002 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-11934504

RESUMO

The rdm genes B, C and E from Streptomyces purpurascens encode enzymes that tailor aklavinone and aclacinomycins. We report that in addition to hydroxylation of aklavinone to epsilon-rhodomycinone, RdmE (aklavinone-11-hydroxylase) hydroxylated 11-deoxy-beta-rhodomycinone to beta-rhodomycinone both in vivo and in vitro. 15-Demethoxyaklavinone and decarbomethoxyaklavinone did not serve as substrates. RdmC (aclacinomycin methyl esterase) converted aclacinomycin T (AcmT) to 15-demethoxyaclacinomycin T, which was in turn converted to 10-decarbomethoxyaclacinomycin T and then to rhodomycin B by RdmB (aclacinomycin-10-hydroxylase). RdmC and RdmB were most active on AcmT, the one-sugar derivative, with their activity decreasing by 70-90% on two- and three-sugar aclacinomycins. Aclacinomycin A competitively inhibited the AcmT modifications at C-10. The results presented here suggest that in vivo the modifications at C-10 take place principally after addition of the first sugar.


Assuntos
Aclarubicina/análogos & derivados , Aclarubicina/metabolismo , Antraciclinas/metabolismo , Antibióticos Antineoplásicos/metabolismo , Proteínas de Bactérias/metabolismo , Naftacenos/metabolismo , Streptomyces/metabolismo , Aclarubicina/química , Hidrocarboneto de Aril Hidroxilases/genética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Proteínas de Bactérias/genética , Hidrolases de Éster Carboxílico/genética , Hidrolases de Éster Carboxílico/metabolismo , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/metabolismo , Mutação , Naftacenos/química , Streptomyces/genética , Streptomyces/crescimento & desenvolvimento , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA