Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 9(5): e93593, 2014.
Article in English | MEDLINE | ID: mdl-24806882

ABSTRACT

The emergence of antibiotic-resistant bacterial strains underscores the importance of identifying new drug targets and developing new antimicrobial compounds. Lysine and meso-diaminopimelic acid are essential for protein production and bacterial peptidoglycan cell wall remodeling and are synthesized in bacteria by enzymes encoded within dap operon. Therefore dap enzymes may serve as excellent targets for developing a new class of antimicrobial agents. The dapE-encoded N-succinyl-L,L-diaminopimelic acid desuccinylase (DapE) converts N-succinyl-L,L-diaminopimelic acid to L,L-diaminopimelic acid and succinate. The enzyme is composed of catalytic and dimerization domains, and belongs to the M20 peptidase family. To understand the specific role of each domain of the enzyme we engineered dimerization domain deletion mutants of DapEs from Haemophilus influenzae and Vibrio cholerae, and characterized these proteins structurally and biochemically. No activity was observed for all deletion mutants. Structural comparisons of wild-type, inactive monomeric DapE enzymes with other M20 peptidases suggest that the dimerization domain is essential for DapE enzymatic activity. Structural analysis and molecular dynamics simulations indicate that removal of the dimerization domain increased the flexibility of a conserved active site loop that may provide critical interactions with the substrate.


Subject(s)
Amidohydrolases/chemistry , Amidohydrolases/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Amidohydrolases/genetics , Bacterial Proteins/genetics , Catalysis , Catalytic Domain , Crystallography, X-Ray , Kinetics , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Mutation , Vibrio cholerae/enzymology
2.
Microbiologyopen ; 2(5): 766-77, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23897711

ABSTRACT

Anaeromyxobacter dehalogenans is a δ-proteobacterium found in diverse soils and sediments. It is of interest in bioremediation efforts due to its dechlorination and metal-reducing capabilities. To gain an understanding on A. dehalogenans' abilities to adapt to diverse environments we analyzed its signal transduction proteins. The A. dehalogenans genome codes for a large number of sensor histidine kinases (HK) and methyl-accepting chemotaxis proteins (MCP); among these 23 HK and 11 MCP proteins have a sensor domain in the periplasm. These proteins most likely contribute to adaptation to the organism's surroundings. We predicted their three-dimensional folds and determined the structures of two of the periplasmic sensor domains by X-ray diffraction. Most of the domains are predicted to have either PAS-like or helical bundle structures, with two predicted to have solute-binding protein fold, and another predicted to have a 6-phosphogluconolactonase like fold. Atomic structures of two sensor domains confirmed the respective fold predictions. The Adeh_2942 sensor (HK) was found to have a helical bundle structure, and the Adeh_3718 sensor (MCP) has a PAS-like structure. Interestingly, the Adeh_3718 sensor has an acetate moiety bound in a binding site typical for PAS-like domains. Future work is needed to determine whether Adeh_3718 is involved in acetate sensing by A. dehalogenans.


Subject(s)
Bacterial Proteins/chemistry , Membrane Proteins/chemistry , Myxococcales/chemistry , Periplasm/chemistry , Protein Kinases/chemistry , Acetic Acid/chemistry , Adaptation, Physiological , Bacterial Proteins/genetics , Binding Sites , Escherichia coli/genetics , Escherichia coli/metabolism , Histidine Kinase , Membrane Proteins/genetics , Methyl-Accepting Chemotaxis Proteins , Models, Molecular , Myxococcales/genetics , Myxococcales/metabolism , Periplasm/genetics , Periplasm/metabolism , Protein Folding , Protein Kinases/genetics , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Signal Transduction , Structural Homology, Protein
3.
Proteins ; 81(10): 1709-26, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23606130

ABSTRACT

Lignin comprises 15-25% of plant biomass and represents a major environmental carbon source for utilization by soil microorganisms. Access to this energy resource requires the action of fungal and bacterial enzymes to break down the lignin polymer into a complex assortment of aromatic compounds that can be transported into the cells. To improve our understanding of the utilization of lignin by microorganisms, we characterized the molecular properties of solute binding proteins of ATP-binding cassette transporter proteins that interact with these compounds. A combination of functional screens and structural studies characterized the binding specificity of the solute binding proteins for aromatic compounds derived from lignin such as p-coumarate, 3-phenylpropionic acid and compounds with more complex ring substitutions. A ligand screen based on thermal stabilization identified several binding protein clusters that exhibit preferences based on the size or number of aromatic ring substituents. Multiple X-ray crystal structures of protein-ligand complexes for these clusters identified the molecular basis of the binding specificity for the lignin-derived aromatic compounds. The screens and structural data provide new functional assignments for these solute-binding proteins which can be used to infer their transport specificity. This knowledge of the functional roles and molecular binding specificity of these proteins will support the identification of the specific enzymes and regulatory proteins of peripheral pathways that funnel these compounds to central metabolic pathways and will improve the predictive power of sequence-based functional annotation methods for this family of proteins.


Subject(s)
ATP-Binding Cassette Transporters/chemistry , Coumaric Acids/chemistry , ATP-Binding Cassette Transporters/classification , ATP-Binding Cassette Transporters/metabolism , Acids, Carbocyclic/chemistry , Acids, Carbocyclic/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Calorimetry , Coumaric Acids/metabolism , Lignin/chemistry , Phylogeny , Propionates , Protein Conformation , Rhodopseudomonas , Spectrometry, Fluorescence
4.
J Mol Biol ; 423(4): 555-75, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-22925578

ABSTRACT

In vitro growth experiments have demonstrated that aromatic compounds derived from lignin can be metabolized and represent a major carbon resource for many soil bacteria. However, the proteins that mediate the movement of these metabolites across the cell membrane have not been thoroughly characterized. To address this deficiency, we used a library representative of lignin degradation products and a thermal stability screen to determine ligand specificity for a set of solute-binding proteins (SBPs) from ATP-binding cassette (ABC) transporters. The ligand mapping process identified a set of proteins from Alphaproteobacteria that recognize various benzoate derivatives. Seven high-resolution crystal structures of these proteins in complex with four different aromatic compounds were obtained. The protein-ligand complexes provide details of molecular recognition that can be used to infer binding specificity. This structure-function characterization provides new insight for the biological roles of these ABC transporters and their SBPs, which had been previously annotated as branched-chain amino-acid-binding proteins. The knowledge derived from the crystal structures provides a foundation for development of sequence-based methods to predict the ligand specificity of other uncharacterized transporters. These results also demonstrate that Alphaproteobacteria possess a diverse set of transport capabilities for lignin-derived compounds. Characterization of this new class of transporters improves genomic annotation projects and provides insight into the metabolic potential of soil bacteria.


Subject(s)
ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/metabolism , Benzoates/metabolism , Biological Transport, Active , Cell Membrane/metabolism , Alphaproteobacteria/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites , Cell Membrane/enzymology , Crystallography, X-Ray , Ligands , Lignin , Protein Binding , Protein Folding , Protein Structure, Tertiary , Structure-Activity Relationship
5.
Biochemistry ; 51(31): 6148-63, 2012 Aug 07.
Article in English | MEDLINE | ID: mdl-22788966

ABSTRACT

Inosine 5'-monophosphate dehydrogenase (IMPDH) catalyzes the first unique step of the GMP branch of the purine nucleotide biosynthetic pathway. This enzyme is found in organisms of all three kingdoms. IMPDH inhibitors have broad clinical applications in cancer treatment, as antiviral drugs and as immunosuppressants, and have also displayed antibiotic activity. We have determined three crystal structures of Bacillus anthracis IMPDH, in a phosphate ion-bound (termed "apo") form and in complex with its substrate, inosine 5'-monophosphate (IMP), and product, xanthosine 5'-monophosphate (XMP). This is the first example of a bacterial IMPDH in more than one state from the same organism. Furthermore, for the first time for a prokaryotic enzyme, the entire active site flap, containing the conserved Arg-Tyr dyad, is clearly visible in the structure of the apoenzyme. Kinetic parameters for the enzymatic reaction were also determined, and the inhibitory effect of XMP and mycophenolic acid (MPA) has been studied. In addition, the inhibitory potential of two known Cryptosporidium parvum IMPDH inhibitors was examined for the B. anthracis enzyme and compared with those of three bacterial IMPDHs from Campylobacter jejuni, Clostridium perfringens, and Vibrio cholerae. The structures contribute to the characterization of the active site and design of inhibitors that specifically target B. anthracis and other microbial IMPDH enzymes.


Subject(s)
Bacillus anthracis/enzymology , IMP Dehydrogenase/chemistry , IMP Dehydrogenase/metabolism , Inosine Monophosphate/metabolism , Ribonucleotides/metabolism , Amino Acid Sequence , Apoenzymes/antagonists & inhibitors , Apoenzymes/chemistry , Apoenzymes/metabolism , Benzimidazoles/chemistry , Benzimidazoles/metabolism , Benzimidazoles/pharmacology , Catalytic Domain , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , IMP Dehydrogenase/antagonists & inhibitors , Models, Molecular , Molecular Sequence Data , Mycophenolic Acid/metabolism , NAD/metabolism , Protein Binding , Triazoles/chemistry , Triazoles/metabolism , Triazoles/pharmacology , Xanthine
6.
Chem Res Toxicol ; 20(12): 1752-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18001056

ABSTRACT

We have recently reported on the development of a La assay to detect reactive molecules by nuclear magnetic resonance (ALARM NMR) to detect reactive false positive hits from high-throughput screening, in which we observed a surprisingly large number of compounds that can oxidize or form covalent adducts with protein thiols groups. In the vast majority of these cases, the covalent interactions are largely nonspecific (e.g., affect many protein targets) and therefore unsuitable for drug development. However, certain thiol-reactive species do appear to inhibit the target of interest in a specific manner. The question then arises as to the potential toxicology risks of developing a drug that can react with protein thiol groups. Here, we report on the evaluation of a large set of ALARM-reactive and -nonreactive compounds against a panel of additional proteins (aldehyde dehydrogenase, superoxide dismutase, and three cytochrome P450 enzymes). It was observed that ALARM-reactive compounds have significantly increased risks of interacting with one or more of these enzymes in vitro. Thus, ALARM NMR seems to be a sensitive tool to rapidly identify compounds with an enhanced risk of producing side effects in humans, including alcohol intolerance, the formation of reactive oxygen species, and drug-drug interactions. In conjunction with other toxicology assays, ALARM NMR should be a valuable tool for prioritizing compounds for lead optimization and animal testing.


Subject(s)
Aldehyde Dehydrogenase/chemistry , Autoantigens/chemistry , Cytochrome P-450 Enzyme Inhibitors , Pharmaceutical Preparations , Ribonucleoproteins/chemistry , Sulfhydryl Compounds/chemistry , Superoxide Dismutase/chemistry , Aldehyde Dehydrogenase/metabolism , Drug Design , Drug-Related Side Effects and Adverse Reactions , Humans , Magnetic Resonance Spectroscopy , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Molecular Structure , Pharmaceutical Preparations/analysis , Protein Binding , Sulfhydryl Compounds/metabolism , Superoxide Dismutase/metabolism , SS-B Antigen
7.
Chem Biol Drug Des ; 70(1): 1-12, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17630989

ABSTRACT

The molecular chaperone HSP90 has been shown to facilitate cancer cell survival by stabilizing key proteins responsible for a malignant phenotype. We report here the results of parallel fragment-based drug design approaches in the design of novel HSP90 inhibitors. Initial aminopyrimidine leads were elaborated using high-throughput organic synthesis to yield nanomolar inhibitors of the enzyme. Second site leads were also identified which bound to HSP90 in two distinct conformations, an 'open' and 'closed' form. Intriguingly, linked fragment approaches targeting both of these conformations were successful in producing novel, micromolar inhibitors. Overall, this study shows that, with only a few fragment hits, multiple lead series can be generated for HSP90 due to the inherent flexibility of the active site. Thus, ample opportunities exist to use these lead series in the development of clinically useful HSP90 inhibitors for the treatment of cancers.


Subject(s)
Drug Design , Enzyme Inhibitors , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Peptide Fragments , Aminopyridines/chemistry , Aminopyridines/metabolism , Crystallography, X-Ray , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Conformation
8.
Chem Biol Drug Des ; 69(6): 395-404, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17581233

ABSTRACT

As part of a fully integrated and comprehensive strategy to discover novel antibacterial agents, NMR- and mass spectrometry-based affinity selection screens were performed to identify compounds that bind to protein targets uniquely found in bacteria and encoded by genes essential for microbial viability. A biphenyl acid lead series emerged from an NMR-based screen with the Haemophilus influenzae protein HI0065, a member of a family of probable ATP-binding proteins found exclusively in eubacteria. The structure-activity relationships developed around the NMR-derived biphenyl acid lead were consistent with on-target antibacterial activity as the Staphylococcus aureus antibacterial activity of the series correlated extremely well with binding affinity to HI0065, while the correlation of binding affinity with B-cell cytotoxicity was relatively poor. Although further studies are needed to conclusively establish the mode of action of the biphenyl series, these compounds represent novel leads that can serve as the basis for the development of novel antibacterial agents that appear to work via an unprecedented mechanism of action. Overall, these results support the genomics-driven hypothesis that targeting bacterial essential gene products that are not present in eukaryotic cells can identify novel antibacterial agents.


Subject(s)
Adenosine Triphosphatases/metabolism , Anti-Bacterial Agents/chemistry , Bacterial Proteins/metabolism , Chemistry, Pharmaceutical/methods , Haemophilus influenzae/metabolism , Amino Acid Sequence , Animals , B-Lymphocytes/metabolism , Drug Design , Genome, Bacterial , Genomics , Magnetic Resonance Spectroscopy , Mass Spectrometry , Molecular Sequence Data , Protein Binding , Structure-Activity Relationship
9.
J Am Chem Soc ; 126(8): 2390-8, 2004 Mar 03.
Article in English | MEDLINE | ID: mdl-14982445

ABSTRACT

An NMR-based alternative to traditional X-ray crystallography and NMR methods for structure-based drug design is described that enables the structure determination of ligands complexed to virtually any biomolecular target regardless of size, composition, or oligomeric state. The method utilizes saturation transfer difference (STD) NMR spectroscopy performed on a ligand complexed to a series of target samples that have been deuterated everywhere except for specific amino acid types. In this way, the amino acid composition of the ligand-binding site can be defined, and, given the three-dimensional structure of the protein target, the three-dimensional structure of the protein-ligand complex can be determined. Unlike earlier NMR methods for solving the structures of protein-ligand complexes, no protein resonance assignments are necessary. Thus, the approach has broad potential applications--especially in cases where X-ray crystallography and traditional NMR methods have failed to produce structural data. The method is called SOS-NMR for structural information using Overhauser effects and selective labeling and is validated on two protein-ligand complexes: FKBP complexed to 2-(3'-pyridyl)-benzimidazole and MurA complexed to uridine diphosphate N-acetylglucosamine.


Subject(s)
Alkyl and Aryl Transferases/chemistry , Nuclear Magnetic Resonance, Biomolecular/methods , Tacrolimus Binding Proteins/chemistry , Benzimidazoles/chemistry , Binding Sites , Crystallography, X-Ray , Models, Molecular , Protein Conformation , Thermodynamics , Uridine Diphosphate N-Acetylglucosamine/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL