Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 225
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Chem Biol ; 20(4): 530-540, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38355722

ABSTRACT

The biosynthetic dogma of ribosomally synthesized and posttranslationally modified peptides (RiPP) involves enzymatic intermolecular modification of core peptide motifs in precursor peptides. The plant-specific BURP-domain protein family, named after their four founding members, includes autocatalytic peptide cyclases involved in the biosynthesis of side-chain-macrocyclic plant RiPPs. Here we show that AhyBURP, a representative of the founding Unknown Seed Protein-type BURP-domain subfamily, catalyzes intramolecular macrocyclizations of its core peptide during the sequential biosynthesis of monocyclic lyciumin I via glycine-tryptophan crosslinking and bicyclic legumenin via glutamine-tyrosine crosslinking. X-ray crystallography of AhyBURP reveals the BURP-domain fold with two type II copper centers derived from a conserved stapled-disulfide and His motif. We show the macrocyclization of lyciumin-C(sp3)-N-bond formation followed by legumenin-C(sp3)-O-bond formation requires dioxygen and radical involvement based on enzyme assays in anoxic conditions and isotopic labeling. Our study expands enzymatic intramolecular modifications beyond catalytic moiety and chromophore biogenesis to RiPP biosynthesis.


Subject(s)
Lignans , Protein Biosynthesis , Protein Processing, Post-Translational , Amino Acid Sequence , Peptides/chemistry , Plants/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism
2.
Proc Natl Acad Sci U S A ; 120(15): e2218248120, 2023 04 11.
Article in English | MEDLINE | ID: mdl-37014851

ABSTRACT

Controlling the selectivity of a reaction is critical for target-oriented synthesis. Accessing complementary selectivity profiles enables divergent synthetic strategies, but is challenging to achieve in biocatalytic reactions given enzymes' innate preferences of a single selectivity. Thus, it is critical to understand the structural features that control selectivity in biocatalytic reactions to achieve tunable selectivity. Here, we investigate the structural features that control the stereoselectivity in an oxidative dearomatization reaction that is key to making azaphilone natural products. Crystal structures of enantiocomplementary biocatalysts guided the development of multiple hypotheses centered on the structural features that control the stereochemical outcome of the reaction; however, in many cases, direct substitutions of active site residues in natural proteins led to inactive enzymes. Ancestral sequence reconstruction (ASR) and resurrection were employed as an alternative strategy to probe the impact of each residue on the stereochemical outcome of the dearomatization reaction. These studies suggest that two mechanisms are active in controlling the stereochemical outcome of the oxidative dearomatization reaction: one involving multiple active site residues in AzaH and the other dominated by a single Phe to Tyr switch in TropB and AfoD. Moreover, this study suggests that the flavin-dependent monooxygenases (FDMOs) adopt simple and flexible strategies to control stereoselectivity, which has led to stereocomplementary azaphilone natural products produced by fungi. This paradigm of combining ASR and resurrection with mutational and computational studies showcases sets of tools for understanding enzyme mechanisms and provides a solid foundation for future protein engineering efforts.


Subject(s)
Biological Products , Mixed Function Oxygenases , Mixed Function Oxygenases/metabolism , Oxidation-Reduction , Flavins/metabolism , Proteins/metabolism , Biocatalysis , Organic Chemicals , Biological Products/chemistry
3.
PLoS Genet ; 18(12): e1010564, 2022 12.
Article in English | MEDLINE | ID: mdl-36574412

ABSTRACT

DNA replication is essential for all living organisms. Several events can disrupt replication, including DNA damage (e.g., pyrimidine dimers, crosslinking) and so-called "roadblocks" (e.g., DNA-binding proteins or transcription). Bacteria have several well-characterized mechanisms for repairing damaged DNA and then restoring functional replication forks. However, little is known about the repair of stalled or arrested replication forks in the absence of chemical alterations to DNA. Using a library of random transposon insertions in Bacillus subtilis, we identified 35 genes that affect the ability of cells to survive exposure to an inhibitor that arrests replication elongation, but does not cause chemical alteration of the DNA. Genes identified include those involved in iron-sulfur homeostasis, cell envelope biogenesis, and DNA repair and recombination. In B. subtilis, and many bacteria, two nucleases (AddAB and RecJ) are involved in early steps in repairing replication forks arrested by chemical damage to DNA and loss of either nuclease causes increased sensitivity to DNA damaging agents. These nucleases resect DNA ends, leading to assembly of the recombinase RecA onto the single-stranded DNA. Notably, we found that disruption of recJ increased survival of cells following replication arrest, indicating that in the absence of chemical damage to DNA, RecJ is detrimental to survival. In contrast, and as expected, disruption of addA decreased survival of cells following replication arrest, indicating that AddA promotes survival. The different phenotypes of addA and recJ mutants appeared to be due to differences in assembly of RecA onto DNA. RecJ appeared to promote too much assembly of RecA filaments. Our results indicate that in the absence of chemical damage to DNA, RecA is dispensable for cells to survive replication arrest and that the stable RecA nucleofilaments favored by the RecJ pathway may lead to cell death by preventing proper processing of the arrested replication fork.


Subject(s)
DNA Damage , DNA Repair , DNA Repair/genetics , DNA Damage/genetics , DNA Replication/genetics , DNA , DNA-Binding Proteins/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Rec A Recombinases/genetics , Rec A Recombinases/metabolism
4.
Mol Microbiol ; 118(4): 426-442, 2022 10.
Article in English | MEDLINE | ID: mdl-36053906

ABSTRACT

DNA replication is highly regulated and primarily controlled at the step of initiation. In bacteria, the replication initiator DnaA and the origin of replication oriC are the primary targets of regulation. Perturbations that increase or decrease replication initiation can cause a decrease in cell fitness. We found that multiple mechanisms, including an increase in replication elongation and a decrease in replication initiation, can compensate for lethal over-initiation. We found that in Bacillus subtilis, under conditions of rapid growth, loss of yabA, a negative regulator of replication initiation, caused a synthetic lethal phenotype when combined with the dnaA1 mutation that also causes replication over-initiation. We isolated several classes of suppressors that restored viability to dnaA1 ∆yabA double mutants. Some suppressors (relA, nrdR) stimulated replication elongation. Others (dnaC, cshA) caused a decrease in replication initiation. One class of suppressors decreased replication initiation in the dnaA1 ∆yabA mutant by causing a decrease in the amount of the replicative helicase, DnaC. We found that decreased levels of helicase in otherwise wild-type cells were sufficient to decrease replication initiation during rapid growth, indicating that the replicative helicase is limiting for replication initiation. Our results highlight the multiple mechanisms cells use to regulate DNA replication.


Subject(s)
Bacterial Proteins , DNA-Binding Proteins , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Bacterial Proteins/genetics , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , DNA Replication , DNA Helicases/genetics , DNA Helicases/metabolism , Replication Origin
5.
J Am Chem Soc ; 144(42): 19326-19336, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36223664

ABSTRACT

Prenyltransfer is an early-stage carbon-hydrogen bond (C-H) functionalization prevalent in the biosynthesis of a diverse array of biologically active bacterial, fungal, plant, and metazoan diketopiperazine (DKP) alkaloids. Toward the development of a unified strategy for biocatalytic construction of prenylated DKP indole alkaloids, we sought to identify and characterize a substrate-permissive C2 reverse prenyltransferase (PT). As the first tailoring event within the biosynthesis of cytotoxic notoamide metabolites, PT NotF catalyzes C2 reverse prenyltransfer of brevianamide F. Solving a crystal structure of NotF (in complex with native substrate and prenyl donor mimic dimethylallyl S-thiolodiphosphate (DMSPP)) revealed a large, solvent-exposed active site, intimating NotF may possess a significantly broad substrate scope. To assess the substrate selectivity of NotF, we synthesized a panel of 30 sterically and electronically differentiated tryptophanyl DKPs, the majority of which were selectively prenylated by NotF in synthetically useful conversions (2 to >99%). Quantitative representation of this substrate library and development of a descriptive statistical model provided insight into the molecular origins of NotF's substrate promiscuity. This approach enabled the identification of key substrate descriptors (electrophilicity, size, and flexibility) that govern the rate of NotF-catalyzed prenyltransfer, and the development of an "induced fit docking (IFD)-guided" engineering strategy for improved turnover of our largest substrates. We further demonstrated the utility of NotF in tandem with oxidative cyclization using flavin monooxygenase, BvnB. This one-pot, in vitro biocatalytic cascade enabled the first chemoenzymatic synthesis of the marine fungal natural product, (-)-eurotiumin A, in three steps and 60% overall yield.


Subject(s)
Biological Products , Dimethylallyltranstransferase , Animals , Dimethylallyltranstransferase/chemistry , Diketopiperazines , Data Science , Indole Alkaloids/chemistry , Protein Engineering , Flavins/metabolism , Mixed Function Oxygenases/metabolism , Solvents , Carbon , Substrate Specificity
6.
J Synchrotron Radiat ; 29(Pt 2): 393-399, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-35254302

ABSTRACT

Algorithms and procedures to fully automate retuning of synchrotron radiation beamlines over wide energy ranges are discussed. The discussion is based on the implementation at the National Institute of General Medical Sciences and the National Cancer Institute Structural Biology Facility at the Advanced Photon Source. When a user selects a new beamline energy, software synchronously controls the beamline monochromator and undulator to maintain the X-ray beam flux after the monochromator, preserves beam attenuation by determining a new set of attenuator foils, changes, as needed, mirror reflecting stripes and the undulator harmonic, preserves beam focal distance of compound refractive lens focusing by changing the In/Out combination of lenses in the transfocator, and, finally, restores beam position at the sample by on-the-fly scanning of either the Kirkpatrick-Baez mirror angles or the transfocator up/down and inboard/outboard positions. The sample is protected from radiation damage by automatically moving it out of the beam during the energy change and optimization.


Subject(s)
Lenses , Synchrotrons , Photons , Software , X-Rays
7.
J Virol ; 95(20): e0084421, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34346770

ABSTRACT

Dengue virus (DENV) and West Nile virus (WNV) are arthropod-transmitted flaviviruses that cause systemic vascular leakage and encephalitis syndromes, respectively, in humans. However, the viral factors contributing to these specific clinical disorders are not completely understood. Flavivirus nonstructural protein 1 (NS1) is required for replication, expressed on the cell surface, and secreted as a soluble glycoprotein, reaching high levels in the blood of infected individuals. Extracellular DENV NS1 and WNV NS1 interact with host proteins and cells, have immune evasion functions, and promote endothelial dysfunction in a tissue-specific manner. To characterize how differences in DENV NS1 and WNV NS1 might function in pathogenesis, we generated WNV NS1 variants with substitutions corresponding to residues found in DENV NS1. We discovered that the substitution NS1-P101K led to reduced WNV infectivity in the brain and attenuated lethality in infected mice, although the virus replicated efficiently in cell culture and peripheral organs and bound at wild-type levels to brain endothelial cells and complement components. The P101K substitution resulted in reduced NS1 antigenemia in mice, and this was associated with reduced WNV spread to the brain. Because exogenous administration of NS1 protein rescued WNV brain infectivity in mice, we conclude that circulating WNV NS1 facilitates viral dissemination into the central nervous system and impacts disease outcomes. IMPORTANCE Flavivirus NS1 serves as an essential scaffolding molecule during virus replication but also is expressed on the cell surface and is secreted as a soluble glycoprotein that circulates in the blood of infected individuals. Although extracellular forms of NS1 are implicated in immune modulation and in promoting endothelial dysfunction at blood-tissue barriers, it has been challenging to study specific effects of NS1 on pathogenesis without disrupting its key role in virus replication. Here, we assessed WNV NS1 variants that do not affect virus replication and evaluated their effects on pathogenesis in mice. Our characterization of WNV NS1-P101K suggests that the levels of NS1 in the circulation facilitate WNV dissemination to the brain and affect disease outcomes. Our findings facilitate understanding of the role of NS1 during flavivirus infection and support antiviral strategies for targeting circulating forms of NS1.


Subject(s)
Viral Nonstructural Proteins/metabolism , West Nile virus/metabolism , Animals , Brain/metabolism , Brain/virology , Dengue Virus/drug effects , Dengue Virus/immunology , Dengue Virus/metabolism , Endothelial Cells , Female , Flavivirus/pathogenicity , Immune Evasion , Male , Mice , Mice, Inbred C57BL , Viral Nonstructural Proteins/analysis , Viral Nonstructural Proteins/blood , Viral Nonstructural Proteins/genetics , Virus Replication/genetics , Virus Replication/physiology , West Nile Fever/immunology , West Nile virus/drug effects , West Nile virus/immunology
8.
Proc Natl Acad Sci U S A ; 116(14): 6775-6783, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30872475

ABSTRACT

Fatty acid biosynthesis in α- and γ-proteobacteria requires two functionally distinct dehydratases, FabA and FabZ. Here, mechanistic cross-linking facilitates the structural characterization of a stable hexameric complex of six Escherichia coli FabZ dehydratase subunits with six AcpP acyl carrier proteins. The crystal structure sheds light on the divergent substrate selectivity of FabA and FabZ by revealing distinct architectures of the binding pocket. Molecular dynamics simulations demonstrate differential biasing of substrate orientations and conformations within the active sites of FabA and FabZ such that FabZ is preorganized to catalyze only dehydration, while FabA is primed for both dehydration and isomerization.


Subject(s)
Acyl Carrier Protein/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/chemistry , Fatty Acid Synthase, Type II/chemistry , Fatty Acids/chemistry , Hydro-Lyases/chemistry , Molecular Dynamics Simulation , Multienzyme Complexes/chemistry , Acyl Carrier Protein/genetics , Acyl Carrier Protein/metabolism , Catalysis , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Fatty Acid Synthase, Type II/genetics , Fatty Acid Synthase, Type II/metabolism , Fatty Acids/biosynthesis , Fatty Acids/genetics , Hydro-Lyases/genetics , Hydro-Lyases/metabolism , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism
9.
Proc Natl Acad Sci U S A ; 116(48): 24303-24309, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31719195

ABSTRACT

Infection of animal cells by numerous viruses is detected and countered by a variety of means, including recognition of nonself nucleic acids. The zinc finger antiviral protein (ZAP) depletes cytoplasmic RNA that is recognized as foreign in mammalian cells by virtue of its elevated CG dinucleotide content compared with endogenous mRNAs. Here, we determined a crystal structure of a protein-RNA complex containing the N-terminal, 4-zinc finger human (h) ZAP RNA-binding domain (RBD) and a CG dinucleotide-containing RNA target. The structure reveals in molecular detail how hZAP is able to bind selectively to CG-rich RNA. Specifically, the 4 zinc fingers create a basic patch on the hZAP RBD surface. The highly basic second zinc finger contains a pocket that selectively accommodates CG dinucleotide bases. Structure guided mutagenesis, cross-linking immunoprecipitation sequencing assays, and RNA affinity assays show that the structurally defined CG-binding pocket is not required for RNA binding per se in human cells. However, the pocket is a crucial determinant of high-affinity, specific binding to CG dinucleotide-containing RNA. Moreover, variations in RNA-binding specificity among a panel of CG-binding pocket mutants quantitatively predict their selective antiviral activity against a CG-enriched HIV-1 strain. Overall, the hZAP RBD RNA structure provides an atomic-level explanation for how ZAP selectively targets foreign, CG-rich RNA.


Subject(s)
GC Rich Sequence , RNA, Viral/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Binding Sites , Crystallography, X-Ray , Fluorescence Polarization , HEK293 Cells , HIV-1/genetics , Humans , Models, Molecular , Mutagenesis , Mutation , Protein Domains , RNA, Viral/chemistry , RNA-Binding Proteins/genetics , Repressor Proteins/genetics , Zinc Fingers
10.
J Am Chem Soc ; 142(5): 2244-2252, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31904957

ABSTRACT

The paraherquamides are potent anthelmintic natural products with complex heptacyclic scaffolds. One key feature of these molecules is the spiro-oxindole moiety that lends a strained three-dimensional architecture to these structures. The flavin monooxygenase PhqK was found to catalyze spirocycle formation through two parallel pathways in the biosynthesis of paraherquamides A and G. Two new paraherquamides (K and L) were isolated from a ΔphqK strain of Penicillium simplicissimum, and subsequent enzymatic reactions with these compounds generated two additional metabolites, paraherquamides M and N. Crystal structures of PhqK in complex with various substrates provided a foundation for mechanistic analyses and computational studies. While it is evident that PhqK can react with various substrates, reaction kinetics and molecular dynamics simulations indicated that the dioxepin-containing paraherquamide L is the favored substrate. Through this effort, we have elucidated a key step in the biosynthesis of the paraherquamides and provided a rationale for the selective spirocyclization of these powerful anthelmintic agents.

11.
J Virol ; 93(24)2019 12 15.
Article in English | MEDLINE | ID: mdl-31578294

ABSTRACT

APOBEC3 proteins APOBEC3F (A3F), APOBEC3G (A3G), and APOBEC3H (A3H) are host restriction factors that inhibit HIV-1 through DNA cytidine deaminase-dependent and -independent mechanisms and have either one (A3H) or two (A3F and A3G) zinc-binding domains. A3H antiviral activity encompasses multiple molecular functions, all of which depend on recognition of RNA or DNA. A3H crystal structures revealed an unusual interaction with RNA wherein an RNA duplex mediates dimerization of two A3H proteins. In this study, we sought to determine the importance of RNA-binding amino acids in the antiviral and biochemical properties of A3H. We show that the wild-type A3H-RNA interaction is essential for A3H antiviral activity and for two deaminase-independent processes: encapsidation into viral particles and inhibition of reverse transcription. Furthermore, an extensive mutagenesis campaign revealed distinct roles for two groups of amino acids at the RNA binding interface. C-terminal helix residues exclusively bind RNA, and loop 1 residues play a dual role in recognition of DNA substrates and in RNA binding. Weakening the interface between A3H and RNA allows DNA substrates to bind with greater affinity and enhances deamination rates, suggesting that RNA binding must be disrupted to accommodate DNA. Intriguingly, we demonstrate that A3H can deaminate overhanging DNA strands of RNA/DNA heteroduplexes, which are early intermediates during reverse transcription and may represent natural A3H substrates. Overall, we present a mechanistic model of A3H restriction and a step-by-step elucidation of the roles of RNA-binding residues in A3H activity, particle incorporation, inhibition of reverse transcriptase inhibition, and DNA cytidine deamination.IMPORTANCE APOBEC3 proteins are host factors that protect the integrity of the host genome by inhibiting retroelements as well as retroviruses, such as HIV-1. To do this, the APOBEC3H protein has evolved unique interactions with structured RNAs. Here, we studied the importance of these interactions in driving antiviral activity of APOBEC3H. Our results provide a clear picture of how RNA binding drives the ability of APOBEC3H to infiltrate new viruses and prevent synthesis of viral DNA. We also explore how RNA binding by APOBEC3H influences recognition and deamination of viral DNA and describe two possible routes by which APOBEC3H might hypermutate the HIV-1 genome. These results highlight how one protein can sense many nucleic acid species for a variety of antiviral activities.


Subject(s)
Aminohydrolases/metabolism , Aminohydrolases/pharmacology , Antiviral Agents/pharmacology , HIV-1/drug effects , HIV-1/metabolism , APOBEC Deaminases/metabolism , Aminohydrolases/chemistry , Aminohydrolases/genetics , Cell Line , DNA, Viral/drug effects , DNA, Viral/metabolism , HIV-1/genetics , Humans , Models, Molecular , Protein Binding , Protein Conformation , RNA Recognition Motif Proteins , RNA-Binding Proteins/chemistry , Reverse Transcription , Virion
12.
Nat Chem Biol ; 14(4): 345-351, 2018 04.
Article in English | MEDLINE | ID: mdl-29531360

ABSTRACT

Hapalindole alkaloids are a structurally diverse class of cyanobacterial natural products defined by their varied polycyclic ring systems and diverse biological activities. These complex metabolites are generated from a common biosynthetic intermediate by the Stig cyclases in three mechanistic steps: a rare Cope rearrangement, 6-exo-trig cyclization, and electrophilic aromatic substitution. Here we report the structure of HpiC1, a Stig cyclase that catalyzes the formation of 12-epi-hapalindole U in vitro. The 1.5-Å structure revealed a dimeric assembly with two calcium ions per monomer and with the active sites located at the distal ends of the protein dimer. Mutational analysis and computational methods uncovered key residues for an acid-catalyzed [3,3]-sigmatropic rearrangement, as well as specific determinants that control the position of terminal electrophilic aromatic substitution, leading to a switch from hapalindole to fischerindole alkaloids.


Subject(s)
Alkaloids/chemistry , Cyanobacteria/enzymology , Indoles/chemistry , Calcium/chemistry , Catalysis , Catalytic Domain , Cloning, Molecular , Cyclization , DNA Mutational Analysis , Dimerization , Indole Alkaloids/chemistry , Ions , Molecular Conformation , Molecular Dynamics Simulation , Molecular Structure , Protein Binding , Quantum Theory , Recombinant Proteins/chemistry , Stereoisomerism
13.
Nature ; 510(7506): 512-7, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24965652

ABSTRACT

Polyketide natural products constitute a broad class of compounds with diverse structural features and biological activities. Their biosynthetic machinery, represented by type I polyketide synthases (PKSs), has an architecture in which successive modules catalyse two-carbon linear extensions and keto-group processing reactions on intermediates covalently tethered to carrier domains. Here we used electron cryo-microscopy to determine sub-nanometre-resolution three-dimensional reconstructions of a full-length PKS module from the bacterium Streptomyces venezuelae that revealed an unexpectedly different architecture compared to the homologous dimeric mammalian fatty acid synthase. A single reaction chamber provides access to all catalytic sites for the intramodule carrier domain. In contrast, the carrier from the preceding module uses a separate entrance outside the reaction chamber to deliver the upstream polyketide intermediate for subsequent extension and modification. This study reveals for the first time, to our knowledge, the structural basis for both intramodule and intermodule substrate transfer in polyketide synthases, and establishes a new model for molecular dissection of these multifunctional enzyme systems.


Subject(s)
Polyketide Synthases/chemistry , Polyketide Synthases/ultrastructure , Streptomyces/enzymology , Biocatalysis , Catalytic Domain , Cryoelectron Microscopy , Fatty Acid Synthases/chemistry , Macrolides/metabolism , Models, Molecular , Polyketide Synthases/metabolism
14.
Nature ; 510(7506): 560-4, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24965656

ABSTRACT

The polyketide synthase (PKS) mega-enzyme assembly line uses a modular architecture to synthesize diverse and bioactive natural products that often constitute the core structures or complete chemical entities for many clinically approved therapeutic agents. The architecture of a full-length PKS module from the pikromycin pathway of Streptomyces venezuelae creates a reaction chamber for the intramodule acyl carrier protein (ACP) domain that carries building blocks and intermediates between acyltransferase, ketosynthase and ketoreductase active sites (see accompanying paper). Here we determine electron cryo-microscopy structures of a full-length pikromycin PKS module in three key biochemical states of its catalytic cycle. Each biochemical state was confirmed by bottom-up liquid chromatography/Fourier transform ion cyclotron resonance mass spectrometry. The ACP domain is differentially and precisely positioned after polyketide chain substrate loading on the active site of the ketosynthase, after extension to the ß-keto intermediate, and after ß-hydroxy product generation. The structures reveal the ACP dynamics for sequential interactions with catalytic domains within the reaction chamber, and for transferring the elongated and processed polyketide substrate to the next module in the PKS pathway. During the enzymatic cycle the ketoreductase domain undergoes dramatic conformational rearrangements that enable optimal positioning for reductive processing of the ACP-bound polyketide chain elongation intermediate. These findings have crucial implications for the design of functional PKS modules, and for the engineering of pathways to generate pharmacologically relevant molecules.


Subject(s)
Biocatalysis , Polyketide Synthases/chemistry , Polyketide Synthases/metabolism , Streptomyces/enzymology , Acyl Carrier Protein/chemistry , Acyl Carrier Protein/metabolism , Acyl Carrier Protein/ultrastructure , Acyltransferases/chemistry , Acyltransferases/metabolism , Acyltransferases/ultrastructure , Alcohol Oxidoreductases/chemistry , Alcohol Oxidoreductases/metabolism , Alcohol Oxidoreductases/ultrastructure , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/ultrastructure , Catalytic Domain , Cryoelectron Microscopy , Macrolides/metabolism , Models, Molecular , Polyketide Synthases/ultrastructure , Protein Structure, Tertiary
15.
Linacre Q ; 87(4): 484-485, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33100396

ABSTRACT

We tend to think that "dignity" means that someone is sophisticated and well-mannered or that human dignity points to great achievements. There is, though, a special dignity in those who are vulnerable for they provide a focal point for love and generally respond enthusiastically to that love; they draw love out of others.

16.
J Biol Chem ; 293(8): 2675-2686, 2018 02 23.
Article in English | MEDLINE | ID: mdl-29348167

ABSTRACT

Rhodanese domains are structural modules present in the sulfurtransferase superfamily. These domains can exist as single units, in tandem repeats, or fused to domains with other activities. Despite their prevalence across species, the specific physiological roles of most sulfurtransferases are not known. Mammalian rhodanese and mercaptopyruvate sulfurtransferase are perhaps the best-studied members of this protein superfamily and are involved in hydrogen sulfide metabolism. The relatively unstudied human thiosulfate sulfurtransferase-like domain-containing 1 (TSTD1) protein, a single-domain cytoplasmic sulfurtransferase, was also postulated to play a role in the sulfide oxidation pathway using thiosulfate to form glutathione persulfide, for subsequent processing in the mitochondrial matrix. Prior kinetic analysis of TSTD1 was performed at pH 9.2, raising questions about relevance and the proposed model for TSTD1 function. In this study, we report a 1.04 Å resolution crystal structure of human TSTD1, which displays an exposed active site that is distinct from that of rhodanese and mercaptopyruvate sulfurtransferase. Kinetic studies with a combination of sulfur donors and acceptors reveal that TSTD1 exhibits a low Km for thioredoxin as a sulfane sulfur acceptor and that it utilizes thiosulfate inefficiently as a sulfur donor. The active site exposure and its interaction with thioredoxin suggest that TSTD1 might play a role in sulfide-based signaling. The apical localization of TSTD1 in human colonic crypts, which interfaces with sulfide-releasing microbes, and the overexpression of TSTD1 in colon cancer provide potentially intriguing clues as to its role in sulfide metabolism.


Subject(s)
Models, Molecular , NADP/metabolism , Neoplasm Proteins/metabolism , Thioredoxin-Disulfide Reductase/metabolism , Thioredoxins/metabolism , Thiosulfate Sulfurtransferase/metabolism , Amino Acid Substitution , Animals , Biocatalysis , Catalytic Domain , Colon/enzymology , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Crystallography, X-Ray , Databases, Protein , Humans , Intestinal Mucosa/enzymology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mutation , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Protein Conformation , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Substrate Specificity , Thioredoxin-Disulfide Reductase/chemistry , Thioredoxins/chemistry , Thioredoxins/genetics , Thiosulfate Sulfurtransferase/chemistry , Thiosulfate Sulfurtransferase/genetics
17.
J Am Chem Soc ; 141(46): 18551-18559, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31692339

ABSTRACT

Selective access to a targeted isomer is often critical in the synthesis of biologically active molecules. Whereas small-molecule reagents and catalysts often act with anticipated site- and stereoselectivity, this predictability does not extend to enzymes. Further, the lack of access to catalysts that provide complementary selectivity creates a challenge in the application of biocatalysis in synthesis. Here, we report an approach for accessing biocatalysts with complementary selectivity that is orthogonal to protein engineering. Through the use of a sequence similarity network (SSN), a number of sequences were selected, and the corresponding biocatalysts were evaluated for reactivity and selectivity. With a number of biocatalysts identified that operate with complementary site- and stereoselectivity, these catalysts were employed in the stereodivergent, chemoenzymatic synthesis of azaphilone natural products. Specifically, the first syntheses of trichoflectin, deflectin-1a, and lunatoic acid A were achieved. In addition, chemoenzymatic syntheses of these azaphilones supplied enantioenriched material for reassignment of the absolute configuration of trichoflectin and deflectin-1a based on optical rotation, CD spectra, and X-ray crystallography.


Subject(s)
Benzopyrans/chemical synthesis , Biological Products/chemical synthesis , Pigments, Biological/chemical synthesis , Benzopyrans/chemistry , Biocatalysis , Biological Products/chemistry , Pigments, Biological/chemistry , Stereoisomerism
18.
Proc Natl Acad Sci U S A ; 113(37): 10316-21, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27573844

ABSTRACT

Alkyl branching at the ß position of a polyketide intermediate is an important variation on canonical polyketide natural product biosynthesis. The branching enzyme, 3-hydroxy-3-methylglutaryl synthase (HMGS), catalyzes the aldol addition of an acyl donor to a ß-keto-polyketide intermediate acceptor. HMGS is highly selective for two specialized acyl carrier proteins (ACPs) that deliver the donor and acceptor substrates. The HMGS from the curacin A biosynthetic pathway (CurD) was examined to establish the basis for ACP selectivity. The donor ACP (CurB) had high affinity for the enzyme (Kd = 0.5 µM) and could not be substituted by the acceptor ACP. High-resolution crystal structures of HMGS alone and in complex with its donor ACP reveal a tight interaction that depends on exquisite surface shape and charge complementarity between the proteins. Selectivity is explained by HMGS binding to an unusual surface cleft on the donor ACP, in a manner that would exclude the acceptor ACP. Within the active site, HMGS discriminates between pre- and postreaction states of the donor ACP. The free phosphopantetheine (Ppant) cofactor of ACP occupies a conserved pocket that excludes the acetyl-Ppant substrate. In comparison with HMG-CoA (CoA) synthase, the homologous enzyme from primary metabolism, HMGS has several differences at the active site entrance, including a flexible-loop insertion, which may account for the specificity of one enzyme for substrates delivered by ACP and the other by CoA.


Subject(s)
Acyl Carrier Protein/chemistry , Hydroxymethylglutaryl-CoA Synthase/chemistry , Polyketide Synthases/chemistry , Polyketides/chemistry , Acyl Carrier Protein/genetics , Acyl Coenzyme A/chemistry , Acyl Coenzyme A/metabolism , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Cyclopropanes/chemistry , Hydroxymethylglutaryl-CoA Synthase/genetics , Polyketide Synthases/genetics , Streptomyces/genetics , Substrate Specificity , Thiazoles/chemistry
19.
J Biol Chem ; 292(34): 14026-14038, 2017 08 25.
Article in English | MEDLINE | ID: mdl-28684420

ABSTRACT

Hydrogen sulfide (H2S) is a signaling molecule that is toxic at elevated concentrations. In eukaryotes, it is cleared via a mitochondrial sulfide oxidation pathway, which comprises sulfide quinone oxidoreductase, persulfide dioxygenase (PDO), rhodanese, and sulfite oxidase and converts H2S to thiosulfate and sulfate. Natural fusions between the non-heme iron containing PDO and rhodanese, a thiol sulfurtransferase, exist in some bacteria. However, little is known about the role of the PDO-rhodanese fusion (PRF) proteins in sulfur metabolism. Herein, we report the kinetic properties and the crystal structure of a PRF from the Gram-negative endophytic bacterium Burkholderia phytofirmans The crystal structures of wild-type PRF and a sulfurtransferase-inactivated C314S mutant with and without glutathione were determined at 1.8, 2.4, and 2.7 Å resolution, respectively. We found that the two active sites are distant and do not show evidence of direct communication. The B. phytofirmans PRF exhibited robust PDO activity and preferentially catalyzed sulfur transfer in the direction of thiosulfate to sulfite and glutathione persulfide; sulfur transfer in the reverse direction was detectable only under limited turnover conditions. Together with the kinetic data, our bioinformatics analysis reveals that B. phytofirmans PRF is poised to metabolize thiosulfate to sulfite in a sulfur assimilation pathway rather than in sulfide stress response as seen, for example, with the Staphylococcus aureus PRF or sulfide oxidation and disposal as observed with the homologous mammalian proteins.


Subject(s)
Bacterial Proteins/metabolism , Burkholderiaceae/enzymology , Models, Molecular , Mutant Chimeric Proteins/metabolism , Quinone Reductases/metabolism , Thiosulfate Sulfurtransferase/metabolism , Amino Acid Substitution , Apoenzymes/chemistry , Apoenzymes/genetics , Apoenzymes/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Biocatalysis , Catalytic Domain , Computational Biology , Crystallography, X-Ray , Cysteine/chemistry , Disulfides/metabolism , Enzyme Stability , Glutathione/analogs & derivatives , Glutathione/chemistry , Glutathione/metabolism , Hydrogen Sulfide/metabolism , Mutant Chimeric Proteins/chemistry , Mutant Chimeric Proteins/genetics , Mutation , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Quinone Reductases/chemistry , Quinone Reductases/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Thiosulfate Sulfurtransferase/chemistry , Thiosulfate Sulfurtransferase/genetics , Thiosulfates/metabolism
20.
J Am Chem Soc ; 140(41): 13205-13208, 2018 10 17.
Article in English | MEDLINE | ID: mdl-30282455

ABSTRACT

A sophisticated intracellular trafficking pathway in humans is used to tailor vitamin B12 into its active cofactor forms, and to deliver it to two known B12-dependent enzymes. Herein, we report an unexpected strategy for cellular retention of B12, an essential and reactive cofactor. If methylmalonyl-CoA mutase is unavailable to accept the coenzyme B12 product of adenosyltransferase, the latter catalyzes homolytic scission of the cobalt-carbon bond in an unconventional reversal of the nucleophilic displacement reaction that was used to make it. The resulting homolysis product binds more tightly to adenosyltransferase than does coenzyme B12, facilitating cofactor retention. We have trapped, and characterized spectroscopically, an intermediate in which the cobalt-carbon bond is weakened prior to being broken. The physiological relevance of this sacrificial catalytic activity for cofactor retention is supported by the significantly lower coenzyme B12 concentration in patients with dysfunctional methylmalonyl-CoA mutase but normal adenosyltransferase activity.


Subject(s)
Cobamides/metabolism , Alkyl and Aryl Transferases/chemistry , Alkyl and Aryl Transferases/metabolism , Carbon/chemistry , Catalytic Domain , Cobalt/chemistry , Cobamides/chemistry , Fibroblasts/metabolism , Humans , Methylmalonyl-CoA Mutase/metabolism , Molecular Structure
SELECTION OF CITATIONS
SEARCH DETAIL