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










Publication year range
1.
Chem Sci ; 6(8): 5027-5033, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26366283

ABSTRACT

Polyketide synthase (PKS) ß-processing domains are responsible for much of the stereochemical complexity of polyketide natural products. Although the importance of ß-processing domains has been well noted and significantly explored, key stereochemical details pertaining to cryptic stereochemistry and the impact of remote stereogenic centers have yet to be fully discerned. To uncover the inner workings of ketoreductases (KR) and dehydratases (DH) from the tylosin pathway a didomain composed of TylDH3-KR3 was recombinantly expressed and interrogated with full-length tetraketide substrates to probe the impact of vicinal and distal stereochemistry. In vitro product isolation analysis revealed the products of the cryptic KR as d-alcohols and of the DH as trans-olefins. Steady-state kinetic analysis of the dehydration reaction demonstrated a strict stereochemical tolerance at the ß-position as d-configured substrates were processed more than 100 times more efficiently than l-alcohols. Unexpectedly, the kcat/KM values were diminished 14- to 45-fold upon inversion of remote ε- and ζ-stereocenters. This stereochemical discrimination is predicted to be driven by a combination of allylic A1,3 strain that likely disfavors binding of the ε-epimer and a loss of electrostatic interactions with the ζ-epimer. Our results strongly suggest that dehydratases may play a role in refining the stereochemical outcomes of preceding modules through their substrate stereospecificity, honing the configurational purity of the final PKS product.

2.
J Am Chem Soc ; 137(22): 7003-6, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-26027428

ABSTRACT

Metabolic engineering of polyketide synthase (PKS) pathways represents a promising approach to natural products discovery. The dehydratase (DH) domains of PKSs, which generate an α,ß-unsaturated bond through a dehydration reaction, have been poorly studied compared with other domains, likely because of the simple nature of the chemical reaction they catalyze and the lack of a convenient assay to measure substrate turnover. Herein we report the first steady-state kinetic analysis of a PKS DH domain employing LC-MS/MS analysis for product quantitation. PikDH2 was selected as a model DH domain. Its substrate specificity and mechanism were interrogated with a systematic series of synthetic triketide substrates containing a nonhydrolyzable thioether linkage as well as by site-directed mutagenesis, evaluation of the pH dependence of the catalytic efficiency (V(max)/K(M)), and kinetic characterization of a mechanism-based inhibitor. These studies revealed that PikDH2 converts d-alcohol substrates to trans-olefin products. The reaction is reversible with equilibrium constants ranging from 1.2 to 2. Moreover, the enzyme activity is robust, and PikDH2 was used on a preparative scale for the chemoenzymatic synthesis of unsaturated triketide products. PikDH2 was shown to possess remarkably strict substrate specificity and is unable to turn over substrates that are epimeric at the ß-, γ-, or δ-position. We also demonstrated that PikDH2 has a key ionizable group with a pK(a) of 7.0 and can be irreversibly inactivated through covalent modification by a mechanism-based inhibitor, which provides a foundation for future structural studies to elucidate substrate-protein interactions.


Subject(s)
Hydro-Lyases/metabolism , Macrolides/metabolism , Polyketide Synthases/metabolism , Hydro-Lyases/chemistry , Kinetics , Polyketide Synthases/chemistry , Tandem Mass Spectrometry
3.
Nat Prod Rep ; 32(5): 654-62, 2015 May.
Article in English | MEDLINE | ID: mdl-25677951

ABSTRACT

Since their first report in 2000, tubulysins have sparked great interest for development as anti-cancer agents due to their exceptionally potent antiproliferative activity. Progress in the discovery and development of tubulysins, especially tubulysin conjugates, has quickly advanced despite limitations in their availability from Nature. In this Highlight, the key research on the isolation and structure determination, biosynthesis, bioactivity, structure-activity relationships (SAR), synthesis, and conjugates of tubulysins is presented.


Subject(s)
Antimitotic Agents/chemistry , Antimitotic Agents/pharmacology , Oligopeptides/chemistry , Oligopeptides/pharmacology , Humans , Molecular Structure , Structure-Activity Relationship
4.
ACS Chem Biol ; 9(12): 2914-22, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25299319

ABSTRACT

Among natural product families, polyketides have shown the most promise for combinatorial biosynthesis of natural product-like libraries. Though recent research in the area has provided many mechanistic revelations, a basic-level understanding of kinetic and substrate tolerability is still needed before the full potential of combinatorial biosynthesis can be realized. We have developed a novel set of chemical probes for the study of ketoreductase domains of polyketide synthases. This chemical tool-based approach was validated using the ketoreductase of pikromycin module 2 (PikKR2) as a model system. Triketide substrate mimics 12 and 13 were designed to increase stability (incorporating a nonhydrolyzable thioether linkage) and minimize nonessential functionality (truncating the phosphopantetheinyl arm). PikKR2 reduction product identities as well as steady-state kinetic parameters were determined by a combination of LC-MS/MS analysis of synthetic standards and a NADPH consumption assay. The d-hydroxyl product is consistent with bioinformatic analysis and results from a complementary biochemical and molecular biological approach. When compared to widely employed substrates in previous studies, diketide 63 and trans-decalone 64, substrates 12 and 13 showed 2-10 fold lower K(M) values (2.4 ± 0.8 and 7.8 ± 2.7 mM, respectively), indicating molecular recognition of intermediate-like substrates. Due to an abundance of the nonreducable enol-tautomer, the k(cat) values were attenuated by as much as 15-336 fold relative to known substrates. This study reveals the high stereoselectivity of PikKR2 in the face of gross substrate permutation, highlighting the utility of a chemical probe-based approach in the study of polyketide ketoreductases.


Subject(s)
Biomimetic Materials/chemistry , Molecular Probes/chemistry , Polyketide Synthases/chemistry , Polyketides/chemistry , Streptomyces/chemistry , Amino Acid Sequence , Biomimetic Materials/chemical synthesis , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Kinetics , Models, Molecular , Molecular Probes/chemical synthesis , Molecular Sequence Data , NADP/chemistry , Polyketide Synthases/genetics , Polyketides/chemical synthesis , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Stereoisomerism , Streptomyces/enzymology , Substrate Specificity
5.
Bioorg Med Chem Lett ; 20(19): 5939-42, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20801036

ABSTRACT

A general strategy to enzymatically label acyl carrier proteins (ACPs) of polyketide synthases has been developed. Incorporation of a chloromethyl ketone or vinyl ketone moiety into polyketide chain elongation intermediate mimics allows for the synthesis of CoA adducts. These CoA adducts undergo enzymatic reaction with Sfp, a phosphopantetheinyl transferase, to afford labeled CurB carrier proteins.


Subject(s)
Acyl Carrier Protein/chemistry , Polyketide Synthases/chemistry , Bacterial Proteins/metabolism , Coenzyme A/chemistry , Ketones/chemistry , Polyketide Synthases/metabolism , Protein Structure, Tertiary , Substrate Specificity , Transferases (Other Substituted Phosphate Groups)/metabolism
6.
J Med Chem ; 52(2): 238-40, 2009 Jan 22.
Article in English | MEDLINE | ID: mdl-19102699

ABSTRACT

A stereoselective total synthesis of the cytotoxic natural products tubulysin U, tubulysin V, and its unnatural epimer epi-tubulysin V, is reported. Simplified analogues containing N,N-dimethyl-D-alanine as a replacement for the N-terminal N-Me-pipecolinic acid residue of the tubulysins are also disclosed. Biological evaluation of these natural products and analogues provided key information with regard to structural and stereochemical requirements for antiproliferative activity and tubulin polymerization inhibition.


Subject(s)
Oligopeptides/chemical synthesis , Oligopeptides/pharmacology , Pipecolic Acids/chemical synthesis , Pipecolic Acids/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatography, High Pressure Liquid , Crystallography, X-Ray , Humans , Magnetic Resonance Spectroscopy , Oligopeptides/chemistry , Pipecolic Acids/chemistry , Stereoisomerism , Structure-Activity Relationship
7.
Bioorg Med Chem Lett ; 18(9): 2996-9, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18411048

ABSTRACT

A series of tubulysin analogs in which one of the stereogenic centers of tubuphenylalanine was eliminated were synthesized. All compounds were tested for antiproliferative activity towards ovarian cancer cells and for inhibition of tubulin polymerization. The dimethyl analogs were generally more active than the desmethyl analogs, and four analogs have tubulin polymerization IC(50) values similar to combretastatin A4 and the hemiasterlin analog HTI-286.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Ovarian Neoplasms/pathology , Phenylalanine/pharmacology , Tubulin Modulators/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Female , Humans , Inhibitory Concentration 50 , Models, Chemical , Oligopeptides/pharmacology , Phenylalanine/analogs & derivatives , Phenylalanine/chemical synthesis , Stilbenes/pharmacology , Structure-Activity Relationship , Tubulin Modulators/chemical synthesis
8.
J Med Chem ; 51(6): 1530-3, 2008 Mar 27.
Article in English | MEDLINE | ID: mdl-18314944

ABSTRACT

An efficient route for the synthesis of the tubulysin family of antimitotic peptides was developed. Simplified tubulysin analogues were synthesized to define the minimum pharmacophore required for cytotoxicity. Simplified tubulysin analogues retain significant cytotoxicity and reveal important preliminary structure-activity relationships.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Oligopeptides/chemical synthesis , Oligopeptides/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Design , Drug Screening Assays, Antitumor , Humans , Molecular Conformation , Oligopeptides/chemistry , Structure-Activity Relationship , Time Factors
10.
J Org Chem ; 71(26): 9853-6, 2006 Dec 22.
Article in English | MEDLINE | ID: mdl-17168607

ABSTRACT

An improved total synthesis of narbonolide and its biotransformation to pikromycin is reported. This total synthesis utilized an intramolecular Nozaki-Hiyama-Kishi coupling that significantly improved macrocyclization yields (90-96%) and allowed for differentiation of the C3- and C5-oxidation states. A pikAI deletion mutant of Streptomyces venezuelae was used to biotransform synthetic narbonolide to pikromycin by glycosylation and oxidation in vivo. This integration of synthetic chemistry and engineered biotransformations holds great promise for the synthesis of novel macrolide analogues of biological interest.


Subject(s)
Macrolides/chemical synthesis , Macrolides/metabolism , Biotransformation , Chromatography, High Pressure Liquid , Macrolides/chemistry , Molecular Structure , Sensitivity and Specificity , Stereoisomerism , Streptomyces/chemistry , Streptomyces/metabolism
11.
Nat Chem Biol ; 2(10): 537-42, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16969372

ABSTRACT

Polyketides are a class of biologically active microbial and plant-derived metabolites that possess a high degree of structural and functional diversity and include many human therapeutics, among them anti-infective and anti-cancer drugs, growth promoters and anti-parasitic agents. The macrolide antibiotics, characterized by a glycoside-linked macrolactone, constitute an important class of polyketides, including erythromycin and the natural ketolide anti-infective agent pikromycin. Here we describe new mechanistic details of macrolactone ring formation catalyzed by the pikromycin polyketide synthase thioesterase domain from Streptomyces venezuelae. A pentaketide phosphonate mimic of the final pikromycin linear chain-elongation intermediate was synthesized and shown to be an active site affinity label. The crystal structures of the affinity-labeled enzyme and of a 12-membered-ring macrolactone product complex suggest a mechanism for cyclization in which a hydrophilic barrier in the enzyme and structural restraints of the substrate induce a curled conformation to direct macrolactone ring formation.


Subject(s)
Macrolides/chemistry , Thiolester Hydrolases/chemistry , Binding Sites/drug effects , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , Catalysis , Crystallization , Cyclization , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Macrolides/metabolism , Models, Molecular , Molecular Conformation , Organophosphonates/chemistry , Organophosphonates/pharmacology , Protein Conformation , Protein Structure, Tertiary , Stereoisomerism , Streptomyces/enzymology , Structure-Activity Relationship , Thiolester Hydrolases/antagonists & inhibitors , Thiolester Hydrolases/metabolism , X-Ray Diffraction
12.
Nat Chem Biol ; 2(10): 531-6, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16969373

ABSTRACT

Polyketides are a diverse class of natural products having important clinical properties, including antibiotic, immunosuppressive and anticancer activities. They are biosynthesized by polyketide synthases (PKSs), which are modular, multienzyme complexes that sequentially condense simple carboxylic acid derivatives. The final reaction in many PKSs involves thioesterase-catalyzed cyclization of linear chain elongation intermediates. As the substrate in PKSs is presented by a tethered acyl carrier protein, introduction of substrate by diffusion is problematic, and no substrate-bound type I PKS domain structure has been reported so far. We describe the chemical synthesis of polyketide-based affinity labels that covalently modify the active site serine of excised pikromycin thioesterase from Streptomyces venezuelae. Crystal structures reported here of the affinity label-pikromycin thioesterase adducts provide important mechanistic insights. These results suggest that affinity labels can be valuable tools for understanding the mechanisms of individual steps within multifunctional PKSs and for directing rational engineering of PKS domains for combinatorial biosynthesis.


Subject(s)
Macrolides/chemistry , Thiolester Hydrolases/chemistry , Binding Sites , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , Catalysis , Crystallization , Cyclization , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Macrolides/metabolism , Models, Molecular , Molecular Conformation , Organophosphonates/chemistry , Organophosphonates/pharmacology , Protein Conformation , Protein Structure, Tertiary , Stereoisomerism , Streptomyces/enzymology , Structure-Activity Relationship , Thiolester Hydrolases/antagonists & inhibitors , Thiolester Hydrolases/metabolism , X-Ray Diffraction
13.
Curr Opin Drug Discov Devel ; 8(6): 741-7, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16312149

ABSTRACT

The discovery of the clinical effectiveness of erythromycin and azithromycin in inflammatory airway diseases has inspired the discovery and development of macrolides with selective immunomodulatory activity. Erythromycin degradation continues to be a source of novel macrolides with a variety of selective biological activities. New technologies for drug discovery based in the emerging field of combinatorial biosynthesis provide the medicinal chemist with novel approaches toward the discovery of novel macrolides. Recent efforts to integrate synthetic organic medicinal chemistry with combinatorial biosynthesis have expanded the number of techniques available for macrolide synthesis.


Subject(s)
Cystic Fibrosis/drug therapy , Drug Design , Erythromycin/analogs & derivatives , Immunologic Factors/chemical synthesis , Macrolides/chemical synthesis , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Azithromycin/chemical synthesis , Azithromycin/therapeutic use , Combinatorial Chemistry Techniques , Cystic Fibrosis/immunology , Erythromycin/chemical synthesis , Erythromycin/therapeutic use , Humans , Immunologic Factors/therapeutic use , Inflammation/drug therapy , Inflammation/immunology , Macrolides/therapeutic use
14.
J Org Chem ; 70(18): 7267-72, 2005 Sep 02.
Article in English | MEDLINE | ID: mdl-16122247

ABSTRACT

[reaction: see text] An improved synthesis of (3S)-3-dihydronarbonolide is reported that constitutes a formal total synthesis of the 14-membered macrolactone antibiotic narbonolide. The key step was an intramolecular Nozaki-Hiyama-Kishi coupling to accomplish macrocyclization in improved yield. The high level of convergence will also allow us to rapidly synthesize narbonolide analogues for the study of enzymes in the pikromycin biosynthetic pathway.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Macrolides/chemical synthesis , Lactones/chemical synthesis
15.
J Am Chem Soc ; 127(25): 8910-1, 2005 Jun 29.
Article in English | MEDLINE | ID: mdl-15969542

ABSTRACT

The polyketide synthase-derived pikromycin thioesterase (Pik TE) is unique in its ability to catalyze the cyclization of 12- and 14-membered macrolactones. In this investigation, the total synthesis of the natural hexaketide chain elongation intermediate as its N-acetyl cysteamine (NAC) thioester has been achieved, and its reaction with Pik TE demonstrated the ability of Pik TE to catalyze its macrolactonization to the natural product 10-deoxymethynolide. A steady-state kinetic analysis of the hexaketide chain intermediate with Pik TE was done. A preliminary substrate specificity study with unnatural hexaketide analogues was accomplished, demonstrating the importance of total synthesis in obtaining access to advanced polyketide intermediates. The results show the sensitivity of Pik TE to minor substrate modifications, and illustrate the potential use of thioesterases as versatile macrolactonization catalysts.


Subject(s)
Lactones/chemical synthesis , Macrolides/chemical synthesis , Multienzyme Complexes/chemistry , Thiolester Hydrolases/chemistry , Catalysis , Lactones/chemistry , Macrolides/chemistry , Molecular Structure
16.
J Am Chem Soc ; 127(23): 8441-52, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15941278

ABSTRACT

The unique ability of the pikromycin (Pik) polyketide synthase to generate 12- and 14-membered ring macrolactones presents an opportunity to explore the fundamental processes underlying polyketide synthesis, specifically the mechanistic details of chain extension, keto group processing, acyl chain release, and macrocyclization. We have synthesized the natural pentaketide and hexaketide chain elongation intermediates as N-acetyl cysteamine (NAC) thioesters and have used them as substrates for in vitro conversions with engineered PikAIII+TE and in combination with native PikAIII (module 5) and PikAIV (module 6) multifunctional proteins. This investigation demonstrates directly the remarkable ability of these monomodules to catalyze one or two chain extension reactions, keto group processing steps, acyl-ACP release, and cyclization to generate 10-deoxymethynolide and narbonolide. The results reveal the enormous preference of Pik monomodules for their natural polyketide substrates and provide an important comparative analysis with previous studies using unnatural diketide NAC thioester substrates.


Subject(s)
Macrolides/metabolism , Polyketide Synthases/metabolism , Amino Acid Sequence , Cysteamine/analogs & derivatives , Cysteamine/metabolism , Kinetics , Molecular Sequence Data , Polyketide Synthases/chemistry
17.
J Med Chem ; 48(6): 1849-56, 2005 Mar 24.
Article in English | MEDLINE | ID: mdl-15771430

ABSTRACT

Fatty acid amide hydrolase (FAAH) degrades neuromodulating fatty acid amides including anandamide (endogenous cannabinoid agonist) and oleamide (sleep-inducing lipid) at their sites of action and is intimately involved in their regulation. Herein we report the discovery of a potent, selective, and efficacious class of reversible FAAH inhibitors that produce analgesia in animal models validating a new therapeutic target for pain intervention. Key to the useful inhibitor discovery was the routine implementation of a proteomics-wide selectivity screen against the serine hydrolase superfamily ensuring selectivity for FAAH coupled with systematic in vivo examinations of candidate inhibitors.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Analgesics/chemical synthesis , Ketones/chemical synthesis , Oxazoles/chemical synthesis , Pyridines/chemical synthesis , Amidohydrolases/chemistry , Analgesics/chemistry , Analgesics/pharmacology , Animals , COS Cells , Chlorocebus aethiops , Humans , Ketones/chemistry , Ketones/pharmacology , Models, Molecular , Oxazoles/chemistry , Oxazoles/pharmacology , Proteomics , Pyridines/chemistry , Pyridines/pharmacology , Rats , Recombinant Proteins/antagonists & inhibitors , Serine Endopeptidases/chemistry , Structure-Activity Relationship
18.
J Med Chem ; 48(4): 1229-36, 2005 Feb 24.
Article in English | MEDLINE | ID: mdl-15715489

ABSTRACT

In pursuit of an apparent literature anomaly, S- and R-6-methyl-6,7-dihydro-2H-benzo[a]quinolizin-2-one-3-carboxylic acids (12 and 22) were synthesized by an unambiguous route from optically active norephedrines, and their antibacterial potencies were measured. Against Gram-negative microorganisms and DNA gyrase a preference for S-absolute configuration was found whereas R-absolute stereochemistry was more active against Gram-positives. These results are in partial conflict with an earlier report. In an attempt to enhance potency, racemic 10-fluoro-9-piperazinyl (35) and related analogues were synthesized by a novel route. The latter analogues were surprisingly unimproved in potency. The implications of these findings are briefly discussed.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , DNA Gyrase/chemistry , Piperazines/chemical synthesis , Quinolizines/chemical synthesis , Topoisomerase II Inhibitors , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Binding Sites , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Piperazines/chemistry , Piperazines/pharmacology , Quinolizines/chemistry , Quinolizines/pharmacology , Stereoisomerism , Structure-Activity Relationship
19.
J Am Chem Soc ; 125(41): 12551-7, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-14531700

ABSTRACT

The unique ability of the pikromycin (Pik) polyketide synthase to generate 12- and 14-membered ring macrolactones presents an opportunity to explore the fundamental processes underlying polyketide synthesis, specifically the mechanistic details of the chain extension process. We have overexpressed and purified PikAIII (module 5) and PikAIV (module 6) and assessed the ability of these proteins to generate tri- and tetraketide lactone products using N-acetylcysteamine-activated diketides and (14)C-methylmalonyl-CoA as substrates. Comparison of the stereochemical specificities for PikAIII and PikAIV and the reported values for the DEBS modules reveals significant differences between these systems.


Subject(s)
Macrolides/metabolism , Multienzyme Complexes/metabolism , Bacillus subtilis/enzymology , Bacillus subtilis/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Kinetics , Multienzyme Complexes/biosynthesis , Multienzyme Complexes/genetics , Multienzyme Complexes/isolation & purification , Streptomyces/enzymology
20.
J Am Chem Soc ; 125(16): 4682-3, 2003 Apr 23.
Article in English | MEDLINE | ID: mdl-12696866

ABSTRACT

The unique ability of the pikromycin polyketide synthase (Pik PKS) to generate 12- and 14-membered ring macrolactones presents an opportunity to explore the fundamental processes of polyketide synthesis, specifically, the mechanistic details of the chain extension process. We have overexpressed and purified PikAIII and PikAIV and demonstrated the ability of these proteins to generate triketide lactone products using (14)C-methylmalonyl-CoA as the sole substrate. Monomodular PikAIII generates TKL (1) when reacted alone, and synthesizes TKL (2) upon reaction in combination with PikAIV. Product formation remains dependent on the enzymatic decarboxylation of methylmalonyl-CoA and transfer of the acyl chain within the enzyme rather than acylation by propionyl-CoA from spontaneous decarboxylation. We propose that synthesis of TKL (1) by PikAIII involves iterative assembly of the triketide chain within a PikAIII homodimer analogous to the nonmodular type I PKS systems.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Macrolides , Multienzyme Complexes/metabolism , Lactones/metabolism , Multienzyme Complexes/chemistry , Streptomyces/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...