Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Nat Chem Biol ; 19(5): 633-640, 2023 05.
Article in English | MEDLINE | ID: mdl-36702957

ABSTRACT

Genome mining of biosynthetic pathways with no identifiable core enzymes can lead to discovery of the so-called unknown (biosynthetic route)-unknown (molecular structure) natural products. Here we focused on a conserved fungal biosynthetic pathway that lacks a canonical core enzyme and used heterologous expression to identify the associated natural product, a highly modified cyclo-arginine-tyrosine dipeptide. Biochemical characterization of the pathway led to identification of a new arginine-containing cyclodipeptide synthase (RCDPS), which was previously annotated as a hypothetical protein and has no sequence homology to non-ribosomal peptide synthetase or bacterial cyclodipeptide synthase. RCDPS homologs are widely encoded in fungal genomes; other members of this family can synthesize diverse cyclo-arginine-Xaa dipeptides, and characterization of a cyclo-arginine-tryptophan RCDPS showed that the enzyme is aminoacyl-tRNA dependent. Further characterization of the biosynthetic pathway led to discovery of new compounds whose structures would not have been predicted without knowledge of RCDPS function.


Subject(s)
Biological Products , Dipeptides/genetics , Bacteria/genetics , Multigene Family , Biosynthetic Pathways/genetics , Genome, Bacterial
2.
Angew Chem Int Ed Engl ; 61(12): e202116928, 2022 03 14.
Article in English | MEDLINE | ID: mdl-35075754

ABSTRACT

The fungal natural product CJ-12662 is a structurally complex terpene-amino acid hybrid, and is a potent anthelmintic compound. The biosynthetic pathway of CJ-12662 is elucidated based on metabolite analysis from heterologous expression. We demonstrate the terpene portion is derived from successive P450-catalyzed oxidations of amorpha-4,11-diene, while three flavin-dependent enzymes are involved in morphing the esterified tryptophan into a chlorinated pyrrolobenzoxazine, utilizing a cascaded [1,2]-Meisenheimer rearrangement.


Subject(s)
Alkyl and Aryl Transferases , Biological Products , Benzoxazines , Pyrroles , Terpenes/chemistry
3.
ACS Synth Biol ; 10(9): 2159-2166, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34415146

ABSTRACT

The microbial synthesis of cannabinoids and related molecules requires access to the intermediate olivetolic acid (OA). Whereas plant enzymes have been explored for E. coli and yeast biosynthesis, moderate yields and shunt product formation are major hurdles. Here, based on the chemical logic to form 2,4-dihydroxybenzoate-containing natural products, we discovered a set of fungal tandem polyketide synthases that can produce OA and the related octanoyl-primed derivative sphaerophorolcarboxylic acid in high titers using the model organism Aspergillus nidulans. This new set of enzymes will enable new synthetic biology strategies to access microbial cannabinoids.


Subject(s)
Aspergillus nidulans/metabolism , Biosynthetic Pathways/genetics , Salicylates/metabolism , Aspergillus nidulans/chemistry , Aspergillus nidulans/genetics , Cannabinoids/chemistry , Cannabinoids/metabolism , Metabolic Engineering/methods , Multigene Family , Polyketide Synthases/genetics , Polyketide Synthases/metabolism , Salicylates/chemistry
4.
J Am Chem Soc ; 2021 Jun 16.
Article in English | MEDLINE | ID: mdl-34132537

ABSTRACT

Acetohydroxyacid synthase (AHAS) is the first enzyme in the branched-chain amino acid biosynthetic pathway and is a validated target for herbicide and fungicide development. Here we report harzianic acid (HA, 1) produced by the biocontrol fungus Trichoderma afroharzianum t-22 (Tht22) as a natural product inhibitor of AHAS. The biosynthetic pathway of HA was elucidated with heterologous reconstitution. Guided by a putative self-resistance enzyme in the genome, HA was biochemically demonstrated to be a selective inhibitor of fungal AHAS, including those from phytopathogenic fungi. In addition, HA can inhibit a common resistant variant of AHAS in which the active site proline is mutated. Structural analysis of AHAS complexed with HA revealed the molecular basis of competitive inhibition, which differs from all known commercial AHAS inhibitors. The alternative binding mode also rationalizes the selectivity of HA, as well as effectiveness toward resistant mutants. A proposed role of HA biosynthesis by Tht22 in the rhizosphere is discussed based on the data.

5.
Org Biomol Chem ; 19(9): 1985-1990, 2021 03 11.
Article in English | MEDLINE | ID: mdl-33570538

ABSTRACT

Trichoderma harzianum is a widely used biocontrol agent in agriculture. Obtaining a full inventory of the small molecules that can be biosynthesized from the encoded biosynthetic gene clusters (BGCs) is therefore useful for understanding associated plant-microbe and microbe-microbe interactions. Here we heterologously reconstituted a polyketide synthase-nonribosomal peptide synthetase (PKS-NRPS) encoding gene cluster from T. harzianum t-22 in Aspergillus nidulans A1145. Six new tetronate natural products trihazone A-F (1-6) were isolated and elucidated by HRESIMS and 1D and 2D NMR data. Three of the products contain an exocyclic olefin, which is derived from the oxidative decarboxylation of an α-ketoglutarate-dependent dioxygenase ThnC as shown by biochemical assays.


Subject(s)
Biological Products/analysis , Hypocreales/chemistry , Multigene Family , Peptide Synthases/chemistry , Polyketide Synthases/chemistry , 4-Butyrolactone/analogs & derivatives , 4-Butyrolactone/analysis , Biological Products/chemistry , Fungal Proteins/genetics , Genomics/methods , Hypocreales/genetics , Mass Spectrometry/methods , Peptide Synthases/genetics , Polyketide Synthases/genetics
6.
Chembiochem ; 22(2): 317-318, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33174653

ABSTRACT

Biocatalysis is a continuously expanding subfield in chemical biology. Herein, I describe two categories of biocatalysts, the LEGO-brick-like and game-console-like type, both of which can streamline the synthetic routes to therapeutics. A multi-disciplinary approach to expand the biocatalytic toolkit will open up opportunities to develop new therapeutics.


Subject(s)
Imines/metabolism , Oxidoreductases/metabolism , Biocatalysis , Imines/chemistry , Molecular Structure , Oxidoreductases/chemistry
7.
J Am Chem Soc ; 142(46): 19668-19677, 2020 11 18.
Article in English | MEDLINE | ID: mdl-33155797

ABSTRACT

Fusaric acid (FA) is a well-known mycotoxin that plays an important role in plant pathology. The biosynthetic gene cluster for FA has been identified, but the biosynthetic pathway remains unclarified. Here, we elucidated the biosynthesis of FA, which features a two-enzyme catalytic cascade, a pyridoxal 5'-phosphate (PLP)-dependent enzyme (Fub7), and a flavin mononucleotide (FMN)-dependent oxidase (Fub9) in synthesizing the picolinic acid scaffold. FA biosynthesis also involves an off-line collaboration between a highly reducing polyketide synthase (HRPKS, Fub1) and a nonribosomal peptide synthetase (NRPS)-like carboxylic acid reductase (Fub8) in making an aliphatic α,ß-unsaturated aldehyde. By harnessing the stereoselective C-C bond-forming activity of Fub7, we established a chemoenzymatic route for stereoconvergent synthesis of a series of 5-alkyl-, 5,5-dialkyl-, and 5,5,6-trialkyl-l-pipecolic acids of high diastereomeric ratio.


Subject(s)
Fusaric Acid/biosynthesis , Mycotoxins/biosynthesis , Oxidoreductases/metabolism , Peptide Synthases/metabolism , Pipecolic Acids/chemistry , Polyketide Synthases/metabolism , Aldehydes/chemistry , Aspergillus nidulans/enzymology , Aspergillus nidulans/metabolism , Biosynthetic Pathways , Flavin Mononucleotide/chemistry , Multigene Family , Picolinic Acids/chemistry , Stereoisomerism
8.
J Am Chem Soc ; 142(23): 10506-10515, 2020 06 10.
Article in English | MEDLINE | ID: mdl-32434326

ABSTRACT

Pyridoxal phosphate (PLP)-dependent enzymes can catalyze transformations of l-amino acids at α, ß, and γ positions. These enzymes are frequently involved in the biosynthesis of nonproteinogenic amino acids as building blocks of natural products and are attractive biocatalysts. Here, we report the discovery of a two-step enzymatic synthesis of (2S,6S)-6-methyl pipecolate 1, from the biosynthetic pathway of citrinadin. The key enzyme CndF is PLP-dependent and catalyzes the synthesis of (S)-2-amino-6-oxoheptanoate 3 that is in equilibrium with the cyclic Schiff base. The second enzyme CndE is a stereoselective imine reductase that gives 1. Biochemical characterization of CndF showed this enzyme performs γ-elimination of O-acetyl-l-homoserine to generate the vinylglycine ketimine, which is subjected to nucleophilic attack by acetoacetate to form the new Cγ-Cδ bond in 3 and complete the γ-substitution reaction. CndF displays promiscuity toward different ß-keto carboxylate and esters. With use of an Aspergillus strain expressing CndF and CndE, feeding various alkyl-ß-keto esters led to the biosynthesis of 6-substituted l-pipecolates. The discovery of CndF expands the repertoire of reactions that can be catalyzed by PLP-dependent enzymes.


Subject(s)
Amino Acids/metabolism , Carbon-Oxygen Lyases/metabolism , Oxidoreductases/metabolism , Pipecolic Acids/metabolism , Pyridoxal Phosphate/metabolism , Amino Acids/chemistry , Biocatalysis , Carbon-Oxygen Lyases/chemistry , Molecular Structure , Oxidoreductases/chemistry , Pipecolic Acids/chemistry , Pyridoxal Phosphate/chemistry
9.
J Am Chem Soc ; 142(2): 710-714, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31885262

ABSTRACT

Biosynthetic pathways containing multiple core enzymes have potential to produce structurally complex natural products. Here we mined a fungal gene cluster that contains two predicted terpene cyclases (TCs) and a nonribosomal peptide synthetase (NRPS). We showed the flv pathway produces flavunoidine 1, an alkaloidal terpenoid. The core of 1 is a tetracyclic, cage-like, and oxygenated sesquiterpene that is connected to dimethylcadaverine via a C-N bond and is acylated with 5,5-dimethyl-l-pipecolate. The roles of all flv enzymes are established on the basis of metabolite analysis from heterologous expression.


Subject(s)
Alkaloids/chemistry , Genome , Peptides/chemistry , Terpenes/chemistry , Ribosomes/chemistry
10.
Proc Natl Acad Sci U S A ; 116(12): 5499-5504, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30842286

ABSTRACT

Fungi have the potential to produce a large repertoire of bioactive molecules, many of which can affect the growth and development of plants. Genomic survey of sequenced biofertilizer fungi showed many secondary metabolite gene clusters are anchored by iterative polyketide synthases (IPKSs), which are multidomain enzymes noted for generating diverse small molecules. Focusing on the biofertilizer Trichoderma harzianum t-22, we identified and characterized a cryptic IPKS-containing cluster that synthesizes tricholignan A, a redox-active ortho-hydroquinone. Tricholignan A is shown to reduce Fe(III) and may play a role in promoting plant growth under iron-deficient conditions. The construction of tricholignan by a pair of collaborating IPKSs was investigated using heterologous reconstitution and biochemical studies. A regioselective methylation step is shown to be a key step in formation of the ortho-hydroquinone. The responsible methyltransferase (MT) is fused with an N-terminal pseudo-acyl carrier protein (ψACP), in which the apo state of the ACP is essential for methylation of the growing polyketide chain. The ψACP is proposed to bind to the IPKS and enable the trans MT to access the growing polyketide. Our studies show that a genome-driven approach to discovering bioactive natural products from biofertilizer fungi can lead to unique compounds and biosynthetic knowledge.


Subject(s)
Arabidopsis/metabolism , Iron/metabolism , Polyketides/metabolism , Trichoderma/genetics , Arabidopsis/enzymology , Metabolic Networks and Pathways/genetics , Methylation , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Multigene Family/genetics , Polyketide Synthases/genetics , Polyketide Synthases/metabolism , Trichoderma/enzymology , Trichoderma/metabolism
11.
Nature ; 549(7673): 502-506, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28902839

ABSTRACT

Pericyclic reactions-which proceed in a concerted fashion through a cyclic transition state-are among the most powerful synthetic transformations used to make multiple regioselective and stereoselective carbon-carbon bonds. They have been widely applied to the synthesis of biologically active complex natural products containing contiguous stereogenic carbon centres. Despite the prominence of pericyclic reactions in total synthesis, only three naturally existing enzymatic examples (the intramolecular Diels-Alder reaction, and the Cope and the Claisen rearrangements) have been characterized. Here we report a versatile S-adenosyl-l-methionine (SAM)-dependent enzyme, LepI, that can catalyse stereoselective dehydration followed by three pericyclic transformations: intramolecular Diels-Alder and hetero-Diels-Alder reactions via a single ambimodal transition state, and a retro-Claisen rearrangement. Together, these transformations lead to the formation of the dihydropyran core of the fungal natural product, leporin. Combined in vitro enzymatic characterization and computational studies provide insight into how LepI regulates these bifurcating biosynthetic reaction pathways by using SAM as the cofactor. These pathways converge to the desired biosynthetic end product via the (SAM-dependent) retro-Claisen rearrangement catalysed by LepI. We expect that more pericyclic biosynthetic enzymatic transformations remain to be discovered in naturally occurring enzyme 'toolboxes'. The new role of the versatile cofactor SAM is likely to be found in other examples of enzyme catalysis.


Subject(s)
Aspergillus nidulans/enzymology , Biocatalysis , Biological Products/metabolism , Biosynthetic Pathways , Coenzymes/metabolism , S-Adenosylmethionine/metabolism , Aspergillus nidulans/genetics , Benzopyrans/chemistry , Benzopyrans/metabolism , Biological Products/chemistry , Chromatography, High Pressure Liquid , Cycloaddition Reaction , Escherichia coli/genetics , Pyrans/chemistry , Pyrans/metabolism , Pyridones/chemistry , Pyridones/metabolism
12.
Biochemistry ; 56(14): 2010-2023, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28362483

ABSTRACT

Terpenoid synthases catalyze isoprenoid cyclization reactions underlying the generation of more than 80,000 natural products. Such dramatic chemodiversity belies the fact that these enzymes generally consist of only three domain folds designated as α, ß, and γ. Catalysis by class I terpenoid synthases occurs exclusively in the α domain, which is found with α, αα, αß, and αßγ domain architectures. Here, we explore the influence of domain architecture on catalysis by taxadiene synthase from Taxus brevifolia (TbTS, αßγ), fusicoccadiene synthase from Phomopsis amygdali (PaFS, (αα)6), and ophiobolin F synthase from Aspergillus clavatus (AcOS, αα). We show that the cyclization fidelity and catalytic efficiency of the α domain of TbTS are severely compromised by deletion of the ßγ domains; however, retention of the ß domain preserves significant cyclization fidelity. In PaFS, we previously demonstrated that one α domain similarly influences catalysis by the other α domain [ Chen , M. , Chou , W. K. W. , Toyomasu , T. , Cane , D. E. , and Christianson , D. W. ( 2016 ) ACS Chem. Biol. 11 , 889 - 899 ]. Here, we show that the hexameric quaternary structure of PaFS enables cluster channeling. We also show that the α domains of PaFS and AcOS can be swapped so as to make functional chimeric αα synthases. Notably, both cyclization fidelity and catalytic efficiency are altered in all chimeric synthases. Twelve newly formed and uncharacterized C20 diterpene products and three C25 sesterterpene products are generated by these chimeras. Thus, engineered αßγ and αα terpenoid cyclases promise to generate chemodiversity in the greater family of terpenoid natural products.


Subject(s)
Alkyl and Aryl Transferases/chemistry , Aspergillus/genetics , Isomerases/chemistry , Mutant Chimeric Proteins/chemistry , Saccharomycetales/genetics , Taxus/genetics , Alkyl and Aryl Transferases/genetics , Alkyl and Aryl Transferases/metabolism , Aspergillus/enzymology , Cyclization , Diterpenes/metabolism , Gene Expression , Isomerases/genetics , Isomerases/metabolism , Kinetics , Models, Molecular , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/metabolism , Protein Domains , Protein Engineering , Protein Structure, Secondary , Saccharomycetales/enzymology , Sesterterpenes/biosynthesis , Taxus/enzymology
13.
Curr Opin Struct Biol ; 41: 27-37, 2016 12.
Article in English | MEDLINE | ID: mdl-27285057

ABSTRACT

Crystal structures of terpenoid cyclases reveal assemblies of three basic domains designated α, ß, and γ. While the biosynthesis of cyclic monoterpenes (C10) and sesquiterpenes (C15) most often involves enzymes with α or αß domain architecture, the biosynthesis of cyclic diterpenes (C20), sesterterpenes (C25), and triterpenes (C30) can involve enzymes with α, αα, ßγ, or αßγ domain architecture. Indeed, some enzymes of terpenoid biosynthesis are bifunctional, with distinct active sites that catalyze sequential reactions. Interestingly, some of these enzymes oligomerize to form dimers, tetramers, and hexamers. Not only can such assemblies enable enzyme regulation by allostery, but they can also provide a modest enhancement of terpenoid product flux through proximity channeling or cluster channeling. The mixing and matching of functional terpenoid cyclase domains through tertiary and/or quaternary structure may also comprise an evolutionary strategy for facile product diversification.


Subject(s)
Lyases/chemistry , Lyases/metabolism , Terpenes/metabolism , Biocatalysis , Humans , Protein Domains , Protein Multimerization , Substrate Specificity
14.
Biochemistry ; 55(20): 2864-74, 2016 05 24.
Article in English | MEDLINE | ID: mdl-27172425

ABSTRACT

Aristolochene synthase (ATAS) is a high-fidelity terpenoid cyclase that converts farnesyl diphosphate exclusively into the bicyclic hydrocarbon aristolochene. Previously determined crystal structures of ATAS complexes revealed trapped active site water molecules that could potentially interact with catalytic intermediates: water "w" hydrogen bonds with S303 and N299, water molecules "w1" and "w2" hydrogen bond with Q151, and a fourth water molecule coordinates to the Mg(2+)C ion. There is no obvious role for water in the ATAS mechanism because the enzyme exclusively generates a hydrocarbon product. Thus, these water molecules are tightly controlled so that they cannot react with carbocation intermediates. Steady-state kinetics and product distribution analyses of eight ATAS mutants designed to perturb interactions with active site water molecules (S303A, S303H, S303D, N299A, N299L, N299A/S303A, Q151H, and Q151E) indicate relatively modest effects on catalysis but significant effects on sesquiterpene product distributions. X-ray crystal structures of S303A, N299A, N299A/S303A, and Q151H mutants reveal minimal perturbation of active site solvent structure. Seven of the eight mutants generate farnesol and nerolidol, possibly resulting from addition of the Mg(2+)C-bound water molecule to the initially formed farnesyl cation, but no products are generated that would suggest enhanced reactivity of other active site water molecules. However, intermediate germacrene A tends to accumulate in these mutants. Thus, apart from the possible reactivity of Mg(2+)C-bound water, active site water molecules in ATAS are not directly involved in the chemistry of catalysis but instead contribute to the template that governs the conformation of the flexible substrate and carbocation intermediates.


Subject(s)
Aspergillus/enzymology , Fungal Proteins/chemistry , Isomerases/chemistry , Sesquiterpenes/chemistry , Water/chemistry , Amino Acid Substitution , Aspergillus/genetics , Catalytic Domain , Crystallography, X-Ray , Fungal Proteins/genetics , Fungal Proteins/metabolism , Isomerases/genetics , Isomerases/metabolism , Mutation, Missense , Sesquiterpenes/metabolism , Water/metabolism
15.
Biochemistry ; 55(14): 2112-21, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26998816

ABSTRACT

The sesquiterpene synthase germacradiene-4-ol synthase (GdolS) from Streptomyces citricolor is one of only a few known high-fidelity terpene synthases that convert farnesyl diphosphate (FDP) into a single hydroxylated product. Crystals of unliganded GdolS-E248A diffracted to 1.50 Å and revealed a typical class 1 sesquiterpene synthase fold with the active site in an open conformation. The metal binding motifs were identified as D(80)DQFD and N(218)DVRSFAQE. Some bound water molecules were evident in the X-ray crystal structure, but none were obviously positioned to quench a putative final carbocation intermediate. Incubations in H2(18)O generated labeled product, confirming that the alcohol functionality arises from nucleophilic capture of the final carbocation by water originating from solution. Site-directed mutagenesis of amino acid residues from both within the metal binding motifs and without identified by sequence alignment with aristolochene synthase from Aspergillus terreus generated mostly functional germacradien-4-ol synthases. Only GdolS-N218Q generated radically different products (∼50% germacrene A), but no direct evidence of the mechanism of incorporation of water into the active site was obtained. Fluorinated FDP analogues 2F-FDP and 15,15,15-F3-FDP were potent noncompetitive inhibitors of GdolS. 12,13-DiF-FDP generated 12,13-(E)-ß-farnesene upon being incubated with GdolS, suggesting stepwise formation of the germacryl cation during the catalytic cycle. Incubation of GdolS with [1-(2)H2]FDP and (R)-[1-(2)H]FDP demonstrated that following germacryl cation formation a [1,3]-hydride shift generates the final carbocation prior to nucleophilic capture. The stereochemistry of this shift is not defined, and the deuteron in the final product was scrambled. Because no clear candidate residue for binding of a nucleophilic water molecule in the active site and no significant perturbation of product distribution from the replacement of active site residues were observed, the final carbocation may be captured by a water molecule from bulk solvent.


Subject(s)
Alkyl and Aryl Transferases/metabolism , Bacterial Proteins/metabolism , Hydro-Lyases/metabolism , Models, Molecular , Naphthols/metabolism , Streptomyces/enzymology , Water/metabolism , Alkyl and Aryl Transferases/antagonists & inhibitors , Alkyl and Aryl Transferases/chemistry , Alkyl and Aryl Transferases/genetics , Amino Acid Substitution , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Biocatalysis/drug effects , Catalytic Domain , Crystallography, X-Ray , Dimerization , Enzyme Inhibitors/pharmacology , Halogenation , Hydro-Lyases/antagonists & inhibitors , Hydro-Lyases/chemistry , Hydro-Lyases/genetics , Hydroxylation/drug effects , Mutation , Polyisoprenyl Phosphates/chemistry , Polyisoprenyl Phosphates/metabolism , Polyisoprenyl Phosphates/pharmacology , Protein Conformation , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sesquiterpenes/chemistry , Sesquiterpenes/metabolism , Sesquiterpenes/pharmacology
16.
ACS Chem Biol ; 11(4): 889-99, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26734760

ABSTRACT

Fusicoccin A is a diterpene glucoside phytotoxin generated by the fungal pathogen Phomopsis amygdali that causes the plant disease constriction canker, first discovered in New Jersey peach orchards in the 1930s. Fusicoccin A is also an emerging new lead in cancer chemotherapy. The hydrocarbon precursor of fusicoccin A is the tricyclic diterpene fusicoccadiene, which is generated by a bifunctional terpenoid synthase. Here, we report X-ray crystal structures of the individual catalytic domains of fusicoccadiene synthase: the C-terminal domain is a chain elongation enzyme that generates geranylgeranyl diphosphate, and the N-terminal domain catalyzes the cyclization of geranylgeranyl diphosphate to form fusicoccadiene. Crystal structures of each domain complexed with bisphosphonate substrate analogues suggest that three metal ions and three positively charged amino acid side chains trigger substrate ionization in each active site. While in vitro incubations reveal that the cyclase domain can utilize farnesyl diphosphate and geranyl diphosphate as surrogate substrates, these shorter isoprenoid diphosphates are mainly converted into acyclic alcohol or hydrocarbon products. Gel filtration chromatography and analytical ultracentrifugation experiments indicate that full-length fusicoccadiene synthase adopts hexameric quaternary structure, and small-angle X-ray scattering data yield a well-defined molecular envelope illustrating a plausible model for hexamer assembly.


Subject(s)
Diterpenes/metabolism , Ligases/metabolism , Catalysis , Crystallography, X-Ray , Ligases/chemistry , Structure-Activity Relationship
17.
Biochemistry ; 52(32): 5441-53, 2013 Aug 13.
Article in English | MEDLINE | ID: mdl-23905850

ABSTRACT

Aristolochene synthase, a metal-dependent sesquiterpene cyclase from Aspergillus terreus, catalyzes the ionization-dependent cyclization of farnesyl diphosphate (FPP) to form the bicyclic eremophilane (+)-aristolochene with perfect structural and stereochemical precision. Here, we report the X-ray crystal structure of aristolochene synthase complexed with three Mg(2+) ions and the unreactive substrate analogue farnesyl-S-thiolodiphosphate (FSPP), showing that the substrate diphosphate group is anchored by metal coordination and hydrogen bond interactions identical to those previously observed in the complex with three Mg(2+) ions and inorganic pyrophosphate (PPi). Moreover, the binding conformation of FSPP directly mimics that expected for productively bound FPP, with the exception of the precise alignment of the C-S bond with regard to the C10-C11 π system that would be required for C1-C10 bond formation in the first step of catalysis. We also report crystal structures of aristolochene synthase complexed with Mg(2+)3-PPi and ammonium or iminium analogues of bicyclic carbocation intermediates proposed for the natural cyclization cascade. Various binding orientations are observed for these bicyclic analogues, and these orientations appear to be driven by favorable electrostatic interactions between the positively charged ammonium group of the analogue and the negatively charged PPi anion. Surprisingly, the active site is sufficiently flexible to accommodate analogues with partially or completely incorrect stereochemistry. Although this permissiveness in binding is unanticipated, based on the stereochemical precision of catalysis that leads exclusively to the (+)-aristolochene stereoisomer, it suggests the ability of the active site to enable controlled reorientation of intermediates during the cyclization cascade. Taken together, these structures illuminate important aspects of the catalytic mechanism.


Subject(s)
Isomerases/chemistry , Aspergillus/enzymology , Aspergillus/metabolism , Binding Sites , Catalysis , Catalytic Domain , Cations, Divalent , Diphosphates/chemistry , Diphosphates/metabolism , Hydrogen Bonding , Isomerases/metabolism , Magnesium/chemistry , Magnesium/metabolism , Models, Molecular , Polyisoprenyl Phosphates/chemistry , Polyisoprenyl Phosphates/metabolism , Quaternary Ammonium Compounds/chemistry , Sesquiterpenes/chemistry , Sesquiterpenes/metabolism , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL
...