Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Commun Biol ; 5(1): 515, 2022 05 30.
Article in English | MEDLINE | ID: mdl-35637276

ABSTRACT

In SCN2A-related disorders, there is an urgent demand to establish efficient methods for determining the gain- (GoF) or loss-of-function (LoF) character of variants, to identify suitable candidates for precision therapies. Here we classify clinical phenotypes of 179 individuals with 38 recurrent SCN2A variants as early-infantile or later-onset epilepsy, or intellectual disability/autism spectrum disorder (ID/ASD) and assess the functional impact of 13 variants using dynamic action potential clamp (DAPC) and voltage clamp. Results show that 36/38 variants are associated with only one phenotypic group (30 early-infantile, 5 later-onset, 1 ID/ASD). Unexpectedly, we revealed major differences in outcome severity between individuals with the same variant for 40% of early-infantile variants studied. DAPC was superior to voltage clamp in predicting the impact of mutations on neuronal excitability and confirmed GoF produces early-infantile phenotypes and LoF later-onset phenotypes. For one early-infantile variant, the co-expression of the α1 and ß2 subunits of the Nav1.2 channel was needed to unveil functional impact, confirming the prediction of 3D molecular modeling. Neither DAPC nor voltage clamp reliably predicted phenotypic severity of early-infantile variants. Genotype, phenotypic group and DAPC are accurate predictors of the biophysical impact of SCN2A variants, but other approaches are needed to predict severity.


Subject(s)
Autism Spectrum Disorder , Epilepsy , Intellectual Disability , Autism Spectrum Disorder/genetics , Epilepsy/genetics , Humans , Intellectual Disability/genetics , NAV1.2 Voltage-Gated Sodium Channel/genetics , Phenotype
2.
J Med Chem ; 62(17): 7788-7805, 2019 09 12.
Article in English | MEDLINE | ID: mdl-31442049

ABSTRACT

Among the >120 modified ribonucleosides in the prokaryotic epitranscriptome, many tRNA modifications are critical to bacterial survival, which makes their synthetic enzymes ideal targets for antibiotic development. Here we performed a structure-based design of inhibitors of tRNA-(N1G37) methyltransferase, TrmD, which is an essential enzyme in many bacterial pathogens. On the basis of crystal structures of TrmDs from Pseudomonas aeruginosa and Mycobacterium tuberculosis, we synthesized a series of thienopyrimidinone derivatives with nanomolar potency against TrmD in vitro and discovered a novel active site conformational change triggered by inhibitor binding. This tyrosine-flipping mechanism is uniquely found in P. aeruginosa TrmD and renders the enzyme inaccessible to the cofactor S-adenosyl-l-methionine (SAM) and probably to the substrate tRNA. Biophysical and biochemical structure-activity relationship studies provided insights into the mechanisms underlying the potency of thienopyrimidinones as TrmD inhibitors, with several derivatives found to be active against Gram-positive and mycobacterial pathogens. These results lay a foundation for further development of TrmD inhibitors as antimicrobial agents.


Subject(s)
Enzyme Inhibitors/pharmacology , Pyrimidines/pharmacology , Tyrosine/pharmacology , tRNA Methyltransferases/antagonists & inhibitors , Binding Sites/drug effects , Dose-Response Relationship, Drug , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Models, Molecular , Molecular Structure , Pseudomonas aeruginosa/enzymology , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship , Tyrosine/chemistry , tRNA Methyltransferases/metabolism
3.
Nat Microbiol ; 2: 17104, 2017 Jun 30.
Article in English | MEDLINE | ID: mdl-28665414

ABSTRACT

Multidrug-resistant (MDR) bacterial infections are a serious threat to public health. Among the most alarming resistance trends is the rapid rise in the number and diversity of ß-lactamases, enzymes that inactivate ß-lactams, a class of antibiotics that has been a therapeutic mainstay for decades. Although several new ß-lactamase inhibitors have been approved or are in clinical trials, their spectra of activity do not address MDR pathogens such as Acinetobacter baumannii. This report describes the rational design and characterization of expanded-spectrum serine ß-lactamase inhibitors that potently inhibit clinically relevant class A, C and D ß-lactamases and penicillin-binding proteins, resulting in intrinsic antibacterial activity against Enterobacteriaceae and restoration of ß-lactam activity in a broad range of MDR Gram-negative pathogens. One of the most promising combinations is sulbactam-ETX2514, whose potent antibacterial activity, in vivo efficacy against MDR A. baumannii infections and promising preclinical safety demonstrate its potential to address this significant unmet medical need.


Subject(s)
Acinetobacter baumannii/drug effects , Azabicyclo Compounds/chemistry , Azabicyclo Compounds/pharmacology , Gram-Negative Bacteria/drug effects , beta-Lactamase Inhibitors/chemistry , beta-Lactamase Inhibitors/pharmacology , Acinetobacter Infections/drug therapy , Acinetobacter Infections/microbiology , Animals , Azabicyclo Compounds/therapeutic use , Azabicyclo Compounds/toxicity , Carbapenems/pharmacology , Dogs , Drug Design , Drug Evaluation, Preclinical , Drug Resistance, Multiple, Bacterial , Enterobacteriaceae/drug effects , Gram-Negative Bacterial Infections/drug therapy , Humans , Mice , Models, Molecular , Penicillin-Binding Proteins/antagonists & inhibitors , Rats , Sulbactam/chemistry , Sulbactam/pharmacology , beta-Lactamase Inhibitors/therapeutic use , beta-Lactamase Inhibitors/toxicity , beta-Lactamases/metabolism , beta-Lactams/pharmacology
4.
Proteins ; 83(9): 1706-19, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26177919

ABSTRACT

UDP-3-O-acyl-N-acetylglucosamine deacetylase (LpxC) is the first committed step to form lipid A, an essential component of the outer membrane of Gram-negative bacteria. As it is essential for the survival of many pathogens, LpxC is an attractive target for antibacterial therapeutics. Herein, we report the product-bound co-crystal structure of LpxC from the acheal Aquifex aeolicus solved to 1.6 Å resolution. We identified interactions by hydroxyl and hydroxymethyl substituents of the product glucosamine ring that may enable new insights to exploit waters in the active site for structure-based design of LpxC inhibitors with novel scaffolds. By using this product structure, we have performed quantum mechanical modeling on the substrate in the active site. Based on our results and published experimental data, we propose a new mechanism that may lead to a better understanding of LpxC catalysis and inhibition.


Subject(s)
Amidohydrolases/chemistry , Bacteria/enzymology , Bacterial Proteins/chemistry , Protein Structure, Tertiary , Acetylation , Amidohydrolases/metabolism , Bacterial Proteins/metabolism , Biocatalysis , Catalytic Domain , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Glucosamine/chemistry , Glucosamine/metabolism , Lipid A/biosynthesis , Models, Molecular , Molecular Structure , Protein Binding , Quantum Theory , Substrate Specificity , Zinc/chemistry , Zinc/metabolism
5.
Proc Natl Acad Sci U S A ; 111(46): 16274-9, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25368144

ABSTRACT

Negamycin is a natural product with broad-spectrum antibacterial activity and efficacy in animal models of infection. Although its precise mechanism of action has yet to be delineated, negamycin inhibits cellular protein synthesis and causes cell death. Here, we show that single point mutations within 16S rRNA that confer resistance to negamycin are in close proximity of the tetracycline binding site within helix 34 of the small subunit head domain. As expected from its direct interaction with this region of the ribosome, negamycin was shown to displace tetracycline. However, in contrast to tetracycline-class antibiotics, which serve to prevent cognate tRNA from entering the translating ribosome, single-molecule fluorescence resonance energy transfer investigations revealed that negamycin specifically stabilizes near-cognate ternary complexes within the A site during the normally transient initial selection process to promote miscoding. The crystal structure of the 70S ribosome in complex with negamycin, determined at 3.1 Å resolution, sheds light on this finding by showing that negamycin occupies a site that partially overlaps that of tetracycline-class antibiotics. Collectively, these data suggest that the small subunit head domain contributes to the decoding mechanism and that small-molecule binding to this domain may either prevent or promote tRNA entry by altering the initial selection mechanism after codon recognition and before GTPase activation.


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , RNA, Bacterial/drug effects , RNA, Ribosomal, 16S/drug effects , Ribosomes/drug effects , Amino Acids, Diamino/pharmacology , Anti-Bacterial Agents/metabolism , Base Pairing , Binding Sites , Binding, Competitive , Crystallography, X-Ray , Drug Resistance, Multiple, Bacterial/genetics , Escherichia coli/genetics , Minocycline/analogs & derivatives , Minocycline/pharmacology , Models, Molecular , Nucleic Acid Conformation , Point Mutation , Protein Biosynthesis/drug effects , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , RNA, Bacterial/physiology , RNA, Ribosomal, 16S/chemistry , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/physiology , RNA, Transfer/metabolism , Ribosomes/ultrastructure , Tetracycline Resistance/genetics , Tetracyclines/metabolism , Tetracyclines/pharmacology , Tigecycline
6.
ACS Med Chem Lett ; 5(11): 1213-8, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25408833

ABSTRACT

In the search for novel Gram-negative agents, we performed a comprehensive search of the AstraZeneca collection and identified a tetrahydropyran-based matrix metalloprotease (MMP) inhibitor that demonstrated nanomolar inhibition of UDP-3-O-(acyl)-N-acetylglucosamine deacetylase (LpxC). Crystallographic studies in Aquifex aeolicus LpxC indicated the tetrahydropyran engaged in the same hydrogen bonds and van der Waals interactions as other known inhibitors. Systematic optimization of three locales on the scaffold provided compounds with improved Gram-negative activity. However, the optimization of LpxC activity was not accompanied by reduced inhibition of MMPs. Comparison of the crystal structure of the native product, UDP-3-O-(acyl)-glucosamine, in Aquifex aeolicus to the structure of a tetrahydropyran-based inhibitor indicates pathways for future optimization.

7.
Protein Expr Purif ; 104: 57-64, 2014 12.
Article in English | MEDLINE | ID: mdl-25240855

ABSTRACT

In Gram-negative bacteria, the cell wall is surrounded by an outer membrane, the outer leaflet of which is comprised of charged lipopolysaccharide (LPS) molecules. Lipid A, a component of LPS, anchors this molecule to the outer membrane. UDP-3-O-(R-3-hydroxymyristoyl)-N-acetylglucosamine deacetylase (LpxC) is a zinc-dependent metalloamidase that catalyzes the first committed step of biosynthesis of Lipid A, making it a promising target for antibiotic therapy. Formation of soluble aggregates of Pseudomonas aeruginosa LpxC protein when overexpressed in Escherichia coli has limited the availability of high quality protein for X-ray crystallography. Expression of LpxC in the presence of an inhibitor dramatically increased protein solubility, shortened crystallization time and led to a high-resolution crystal structure of LpxC bound to the inhibitor. However, this approach required large amounts of compound, restricting its use. To reduce the amount of compound needed, an overexpression strain of E. coli was created lacking acrB, a critical component of the major efflux pump. By overexpressing LpxC in the efflux deficient strain in the presence of LpxC inhibitors, several structures of P. aeruginosa LpxC in complex with different compounds were solved to accelerate structure-based drug design.


Subject(s)
Amidohydrolases/metabolism , Bacterial Proteins/metabolism , Escherichia coli Proteins/genetics , Multidrug Resistance-Associated Proteins/genetics , Pseudomonas aeruginosa/enzymology , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/genetics , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Catalysis , Chromatography, Liquid , Crystallography, X-Ray , Escherichia coli , Gene Expression , Mass Spectrometry , Protein Conformation , Zinc/chemistry , Zinc/metabolism
9.
Antimicrob Agents Chemother ; 58(9): 5269-79, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24957822

ABSTRACT

Streptogramin antibiotics are divided into types A and B, which in combination can act synergistically. We compared the molecular interactions of the streptogramin combinations Synercid (type A, dalfopristin; type B, quinupristin) and NXL 103 (type A, flopristin; type B, linopristin) with the Escherichia coli 70S ribosome by X-ray crystallography. We further analyzed the activity of the streptogramin components individually and in combination. The streptogramin A and B components in Synercid and NXL 103 exhibit synergistic antimicrobial activity against certain pathogenic bacteria. However, in transcription-coupled translation assays, only combinations that include dalfopristin, the streptogramin A component of Synercid, show synergy. Notably, the diethylaminoethylsulfonyl group in dalfopristin reduces its activity but is the basis for synergy in transcription-coupled translation assays before its rapid hydrolysis from the depsipeptide core. Replacement of the diethylaminoethylsulfonyl group in dalfopristin by a nonhydrolyzable group may therefore be beneficial for synergy. The absence of general streptogramin synergy in transcription-coupled translation assays suggests that the synergistic antimicrobial activity of streptogramins can occur independently of the effects of streptogramin on translation.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Protein Biosynthesis/drug effects , Streptogramins/therapeutic use , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Crystallography, X-Ray , Drug Combinations , Drug Synergism , Enterococcus faecalis/drug effects , Escherichia coli/drug effects , Haemophilus influenzae/drug effects , Microbial Sensitivity Tests , Ribosomes/drug effects , Ribosomes/ultrastructure , Staphylococcus aureus/drug effects , Streptococcus pneumoniae/drug effects , Streptogramin A/administration & dosage , Streptogramin A/pharmacology , Streptogramin A/therapeutic use , Streptogramin B/administration & dosage , Streptogramin B/pharmacology , Streptogramin B/therapeutic use , Streptogramins/administration & dosage , Streptogramins/chemistry , Streptogramins/pharmacology , Virginiamycin/administration & dosage , Virginiamycin/pharmacology , Virginiamycin/therapeutic use
10.
J Med Chem ; 57(11): 4584-97, 2014 Jun 12.
Article in English | MEDLINE | ID: mdl-24828090

ABSTRACT

Thymidylate kinase (TMK), an essential enzyme in bacterial DNA biosynthesis, is an attractive therapeutic target for the development of novel antibacterial agents, and we continue to explore TMK inhibitors with improved potency, protein binding, and pharmacokinetic potential. A structure-guided design approach was employed to exploit a previously unexplored region in Staphylococcus aureus TMK via novel interactions. These efforts produced compound 39, with 3 nM IC50 against S. aureus TMK and 2 µg/mL MIC against methicillin-resistant S. aureus (MRSA). This compound exhibits a striking inverted chiral preference for binding relative to earlier compounds and also has improved physical properties and pharmacokinetics over previously published compounds. An example of this new series was efficacious in a murine S. aureus infection model, suggesting that compounds like 39 are options for further work toward a new Gram-positive antibiotic by maintaining a balance of microbiological potency, low clearance, and low protein binding that can result in lower efficacious doses.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Bacterial Proteins/antagonists & inhibitors , Gram-Positive Bacteria/drug effects , Nucleoside-Phosphate Kinase/antagonists & inhibitors , Piperidines/chemical synthesis , Pyrimidinones/chemical synthesis , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Binding Sites , Crystallography, X-Ray , Drug Resistance, Bacterial , Gram-Positive Bacteria/enzymology , Hydrophobic and Hydrophilic Interactions , Mice , Microbial Sensitivity Tests , Models, Molecular , Piperidines/chemistry , Piperidines/pharmacology , Protein Conformation , Pyrimidinones/chemistry , Pyrimidinones/pharmacology , Staphylococcal Infections/drug therapy , Staphylococcus aureus/drug effects , Staphylococcus aureus/enzymology , Stereoisomerism , Structure-Activity Relationship
11.
J Med Chem ; 56(18): 7278-88, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-23981144

ABSTRACT

The tRNA-(N(1)G37) methyltransferase (TrmD) is essential for growth and highly conserved in both Gram-positive and Gram-negative bacterial pathogens. Additionally, TrmD is very distinct from its human orthologue TRM5 and thus is a suitable target for the design of novel antibacterials. Screening of a collection of compound fragments using Haemophilus influenzae TrmD identified inhibitory, fused thieno-pyrimidones that were competitive with S-adenosylmethionine (SAM), the physiological methyl donor substrate. Guided by X-ray cocrystal structures, fragment 1 was elaborated into a nanomolar inhibitor of a broad range of Gram-negative TrmD isozymes. These compounds demonstrated no activity against representative human SAM utilizing enzymes, PRMT1 and SET7/9. This is the first report of selective, nanomolar inhibitors of TrmD with demonstrated ability to order the TrmD lid in the absence of tRNA.


Subject(s)
Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Haemophilus influenzae/enzymology , tRNA Methyltransferases/antagonists & inhibitors , Adenosine/metabolism , Amines/chemical synthesis , Amines/chemistry , Amines/metabolism , Amines/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Haemophilus influenzae/drug effects , Humans , Methionine/metabolism , Microbial Sensitivity Tests , Models, Molecular , Protein Structure, Tertiary , RNA, Transfer/chemistry , RNA, Transfer/metabolism , Structure-Activity Relationship , Substrate Specificity , tRNA Methyltransferases/chemistry , tRNA Methyltransferases/metabolism
12.
J Am Chem Soc ; 135(12): 4580-3, 2013 Mar 27.
Article in English | MEDLINE | ID: mdl-23480637

ABSTRACT

We report a general method for light-assisted control of interactions of PDZ domain binding motifs with their cognate domains by the incorporation of a photolabile caging group onto the essential C-terminal carboxylate binding determinant of the motif. The strategy was implemented and validated for both simple monovalent and biomimetic divalent ligands, which have recently been established as powerful tools for acute perturbation of native PDZ domain-dependent interactions in live cells.


Subject(s)
Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , PDZ Domains , Peptides/chemistry , Peptides/metabolism , Amino Acid Sequence , Animals , Fluorescence , Fluorescent Dyes/chemical synthesis , Ligands , Light , Models, Molecular , Molecular Sequence Data , Peptides/chemical synthesis , Photolysis , Protein Binding , Protein Interaction Mapping , Rats
13.
Bioorg Med Chem Lett ; 23(1): 169-73, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23206863

ABSTRACT

Thymidylate kinase (TMK) is an essential enzyme for DNA synthesis in bacteria, phosphorylating deoxythymidine monophosphate (dTMP) to deoxythymidine diphosphate (dTDP), and thus is a potential new antibacterial drug target. Previously, we have described the first potent and selective inhibitors of Gram-positive TMK, leading to in vivo validation of the target. Here, a structure-guided design approach based on the initial series led to the discovery of novel sulfonylpiperidine inhibitors of TMK. Formation of hydrogen bonds with Arg48 in Staphylococcus aureus TMK was key to obtaining excellent enzyme affinity, as verified by protein crystallography. Replacement of a methylene linker in the series by a sulfonamide was accomplished with retention of binding conformation. Further optimization of logD yielded phenol derivative 11, a potent inhibitor of TMK showing excellent MICs against a broad spectrum of Gram-positive bacteria and >10(5) selectivity versus the human TMK homologue.


Subject(s)
Anti-Bacterial Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Nucleoside-Phosphate Kinase/antagonists & inhibitors , Piperidines/chemistry , Staphylococcus aureus/enzymology , Sulfonamides/chemistry , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Gram-Positive Bacteria/drug effects , Microbial Sensitivity Tests , Molecular Docking Simulation , Nucleoside-Phosphate Kinase/metabolism , Piperidines/chemical synthesis , Piperidines/pharmacology , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/pharmacology
14.
J Med Chem ; 55(22): 10010-21, 2012 Nov 26.
Article in English | MEDLINE | ID: mdl-23043329

ABSTRACT

Thymidylate kinase (TMK) is an essential enzyme in bacterial DNA synthesis. The deoxythymidine monophosphate (dTMP) substrate binding pocket was targeted in a rational-design, structure-supported effort, yielding a unique series of antibacterial agents showing a novel, induced-fit binding mode. Lead optimization, aided by X-ray crystallography, led to picomolar inhibitors of both Streptococcus pneumoniae and Staphylococcus aureus TMK. MICs < 1 µg/mL were achieved against methicillin-resistant S. aureus (MRSA), S. pneumoniae, and vancomycin-resistant Enterococcus (VRE). Log D adjustments yielded single diastereomers 14 (TK-666) and 46, showing a broad antibacterial spectrum against Gram-positive bacteria and excellent selectivity against the human thymidylate kinase ortholog.


Subject(s)
Anti-Bacterial Agents/pharmacology , Benzoates/pharmacology , Enterococcus/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Nucleoside-Phosphate Kinase/antagonists & inhibitors , Staphylococcus aureus/drug effects , Streptococcus pneumoniae/drug effects , Thymine/analogs & derivatives , Vancomycin Resistance/drug effects , Anti-Bacterial Agents/chemical synthesis , Benzoates/chemical synthesis , Catalytic Domain , Crystallography, X-Ray , Humans , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Nucleoside-Phosphate Kinase/metabolism , Structure-Activity Relationship , Thymine/chemical synthesis , Thymine/pharmacology
15.
ACS Chem Biol ; 7(11): 1866-72, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-22908966

ABSTRACT

There is an urgent need for new antibacterials that pinpoint novel targets and thereby avoid existing resistance mechanisms. We have created novel synthetic antibacterials through structure-based drug design that specifically target bacterial thymidylate kinase (TMK), a nucleotide kinase essential in the DNA synthesis pathway. A high-resolution structure shows compound TK-666 binding partly in the thymidine monophosphate substrate site, but also forming new induced-fit interactions that give picomolar affinity. TK-666 has potent, broad-spectrum Gram-positive microbiological activity (including activity against methicillin-resistant Staphylococcus aureus and vancomycin-resistant Enterococcus), bactericidal action with rapid killing kinetics, excellent target selectivity over the human ortholog, and low resistance rates. We demonstrate in vivo efficacy against S. aureus in a murine infected-thigh model. This work presents the first validation of TMK as a compelling antibacterial target and provides a rationale for pursuing novel clinical candidates for treating Gram-positive infections through TMK.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/enzymology , Nucleoside-Phosphate Kinase/antagonists & inhibitors , Enterococcus/drug effects , Enterococcus/enzymology , Gram-Positive Bacterial Infections/drug therapy , Humans , Models, Molecular , Nucleoside-Phosphate Kinase/metabolism , Staphylococcal Infections/drug therapy , Staphylococcus aureus/drug effects , Staphylococcus aureus/enzymology
16.
Nat Chem Biol ; 7(2): 81-91, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21186349

ABSTRACT

The interactions of the AMPA receptor (AMPAR) auxiliary subunit Stargazin with PDZ domain-containing scaffold proteins such as PSD-95 are critical for the synaptic stabilization of AMPARs. To investigate these interactions, we have developed biomimetic competing ligands that are assembled from two Stargazin-derived PSD-95/DLG/ZO-1 (PDZ) domain-binding motifs using 'click' chemistry. Characterization of the ligands in vitro and in a cellular FRET-based model revealed an enhanced affinity for the multiple PDZ domains of PSD-95 compared to monovalent peptides. In cultured neurons, the divalent ligands competed with transmembrane AMPAR regulatory protein (TARP) for the intracellular membrane-associated guanylate kinase resulting in increased lateral diffusion and endocytosis of surface AMPARs, while showing strong inhibition of synaptic AMPAR currents. This provides evidence for a model in which the TARP-containing AMPARs are stabilized at the synapse by engaging in multivalent interactions. In light of the prevalence of PDZ domain clusters, these new biomimetic chemical tools could find broad application for acutely perturbing multivalent complexes.


Subject(s)
Biomimetics , Receptors, AMPA/metabolism , Synapses/metabolism , Ligands , Models, Molecular
17.
Biochemistry ; 49(33): 7227-37, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20604544

ABSTRACT

In recent years, the opportunistic pathogen Pseudomonas aeruginosa has emerged as a major source of hospital-acquired infections. Effective treatment has proven increasingly difficult due to the spread of multidrug resistant strains and thus requires a deeper understanding of the biochemical mechanisms of pathogenicity. The central carbohydrate of the P. aeruginosa PAO1 (O5) B-band O-antigen, ManNAc(3NAc)A, has been shown to be critical for virulence and is produced in a stepwise manner by five enzymes in the Wbp pathway (WbpA, WbpB, WbpE, WbpD, and WbpI). Herein, we present the crystal structure of the aminotransferase WbpE from P. aeruginosa PAO1 in complex with the cofactor pyridoxal 5'-phosphate (PLP) and product UDP-GlcNAc(3NH(2))A as the external aldimine at 1.9 A resolution. We also report the structures of WbpE in complex with PMP alone as well as the PLP internal aldimine and show that the dimeric structure of WbpE observed in the crystal structure is confirmed by analytical ultracentrifugation. Analysis of these structures reveals that the active site of the enzyme is composed of residues from both subunits. In particular, we show that a key residue (Arg229), which has previously been implicated in direct interactions with the alpha-carboxylate moiety of alpha-ketoglutarate, is also uniquely positioned to bestow specificity for the 6''-carboxyl group of GlcNAc(3NH(2))A through a salt bridge. This finding is intriguing because while an analogous basic residue is present in WbpE homologues that do not process 6''-carboxyl-modified saccharides, recent structural studies reveal that this side chain is retracted to accommodate a neutral C6'' atom. This work represents the first structural analysis of a nucleotide sugar aminotransferase with a bound product modified at the C2'', C3'', and C6'' positions and provides insight into a novel target for treatment of P. aeruginosa infection.


Subject(s)
Nitrogenous Group Transferases/chemistry , Pseudomonas Infections/enzymology , Pseudomonas aeruginosa/enzymology , Pyridoxal Phosphate/metabolism , Schiff Bases/metabolism , Uridine Diphosphate Glucuronic Acid/analogs & derivatives , Alanine/genetics , Crystallography, X-Ray , Models, Molecular , Mutation , Nitrogenous Group Transferases/genetics , Nitrogenous Group Transferases/metabolism , O Antigens/metabolism , Protein Binding , Pyridoxal Phosphate/chemistry , Pyridoxamine/analogs & derivatives , Pyridoxamine/chemistry , Pyridoxamine/metabolism , Schiff Bases/chemistry , Uridine Diphosphate Glucuronic Acid/chemistry , Uridine Diphosphate Glucuronic Acid/metabolism
18.
Nat Methods ; 6(7): 532-7, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19525958

ABSTRACT

Visualizing conformational dynamics in proteins has been difficult, and the atomic-scale motions responsible for the behavior of most allosteric proteins are unknown. Here we report that fluorescence resonance energy transfer (FRET) between a small fluorescent dye and a nickel ion bound to a dihistidine motif can be used to monitor small structural rearrangements in proteins. This method provides several key advantages over classical FRET, including the ability to measure the dynamics of close-range interactions, the use of small probes with short linkers, a low orientation dependence, and the ability to add and remove unique tunable acceptors. We used this 'transition metal ion FRET' approach along with X-ray crystallography to determine the structural changes of the gating ring of the mouse hyperpolarization-activated cyclic nucleotide-regulated ion channel HCN2. Our results suggest a general model for the conformational switch in the cyclic nucleotide-binding site of cyclic nucleotide-regulated ion channels.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Protein Conformation , Proteins/chemistry , Amino Acid Sequence , Animals , Binding Sites , Cations, Divalent , Crystallography, X-Ray , Cyclic AMP/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/chemistry , Ion Channels/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Nickel/chemistry , Peptides/chemistry , Potassium Channels , Protein Binding , Protein Structure, Secondary
19.
Biochim Biophys Acta ; 1788(9): 1939-49, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19524546

ABSTRACT

Investigation of the mechanistic bases and physiological importance of cAMP regulation of HCN channels has exploited an arginine to glutamate mutation in the nucleotide-binding fold, an approach critically dependent on the mutation selectively lowering the channel's nucleotide affinity. In apparent conflict with this, in intact Xenopus oocytes, HCN and HCN-RE channels exhibit qualitatively and quantitatively distinct responses to the tyrosine kinase inhibitor, genistein -- the estrogenic isoflavonoid strongly depolarizes the activation mid-point of HCN1-R538E, but not HCN1 channels (+9.8 mV + or - 0.9 versus +2.2 mV + or - 0.6) and hyperpolarizes gating of HCN2 (-4.8 mV + or - 1.0) but depolarizes gating of HCN2-R591E (+13.2 mV + or - 2.1). However, excised patch recording, X-ray crystallography and modeling reveal that this is not due to either a fundamental effect of the mutation on channel gating per se or of genistein acting as a mutation-sensitive partial agonist at the cAMP site. Rather, we find that genistein equivalently moves both HCN and HCN-RE channels closer to the open state (rendering the channels inherently easier to open but at a cost of decreasing the coupling energy of cAMP) and that the anomaly reflects a balance of these energetic effects with the isoform-specific inhibition of activation by the nucleotide gating ring and relief of this by endogenous cAMP. These findings have specific implications with regard to findings based on HCN-RE channels and kinase antagonists and general implications with respect to interpretation of drug effects in mutant channel backgrounds.


Subject(s)
Cyclic AMP/physiology , Cyclic Nucleotide-Gated Cation Channels/drug effects , Cyclic Nucleotide-Gated Cation Channels/physiology , Genistein/pharmacology , Ion Channel Gating/physiology , Potassium Channels/drug effects , Potassium Channels/physiology , Animals , Cyclic Nucleotide-Gated Cation Channels/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channel Gating/drug effects , Mice , Potassium Channels/genetics , Protein Structure, Tertiary
20.
J Biol Chem ; 283(41): 27937-27946, 2008 Oct 10.
Article in English | MEDLINE | ID: mdl-18667421

ABSTRACT

The carbohydrate 2, 4-diacetamido-2, 4, 6-trideoxy-alpha-D-glucopyranose (BacAc(2)) is found in a variety of eubacterial pathogens. In Campylobacter jejuni, PglD acetylates the C4 amino group on UDP-2-acetamido-4-amino-2, 4, 6-trideoxy-alpha-D-glucopyranose (UDP-4-amino-sugar) to form UDP-BacAc(2). Sequence analysis predicts PglD to be a member of the left-handed beta helix family of enzymes. However, poor sequence homology between PglD and left-handed beta helix enzymes with existing structural data precludes unambiguous identification of the active site. The co-crystal structures of PglD in the presence of citrate, acetyl coenzyme A, or the UDP-4-amino-sugar were solved. The biological assembly is a trimer with one active site formed between two protomers. Residues lining the active site were identified, and results from functional assays on alanine mutants suggest His-125 is critical for catalysis, whereas His-15 and His-134 are involved in substrate binding. These results are discussed in the context of implications for proteins homologous to PglD in other pathogens.


Subject(s)
Acetylglucosamine/analogs & derivatives , Acetyltransferases/chemistry , Campylobacter jejuni/enzymology , Acetylglucosamine/biosynthesis , Acetylglucosamine/chemistry , Acetyltransferases/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites/physiology , Catalysis , Crystallography, X-Ray , Protein Structure, Quaternary/physiology , Protein Structure, Secondary/physiology , Structural Homology, Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...