Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Protein Sci ; 28(4): 794-799, 2019 04.
Article in English | MEDLINE | ID: mdl-30687968

ABSTRACT

Cellulases play a significant role in the degradation of complex carbohydrates. In the human gut, anaerobic bacteria are essential to the well-being of the host by producing these essential enzymes that convert plant polymers into simple sugars that can then be further metabolized by the host. Here, we report the 2.08 Å resolution structure of HLB5, a chemically verified cellulase that was identified previously from an anaerobic gut bacterium and that has no structural cellulase homologues in PDB nor possesses any conserved region typical for glycosidases. We anticipate that the information presented here will facilitate the identification of additional cellulases for which no homologues have been identified to date and enhance our understanding how these novel cellulases bind and hydrolyze their substrates.


Subject(s)
Bacterial Proteins/chemistry , Bacteroidetes/chemistry , Cellulase/chemistry , Binding Sites , Crystallography, X-Ray , Hydrolysis , Models, Molecular , Protein Conformation
2.
Proc Natl Acad Sci U S A ; 112(41): 12693-8, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26420866

ABSTRACT

Acyltransferase (AT)-less type I polyketide synthases (PKSs) break the type I PKS paradigm. They lack the integrated AT domains within their modules and instead use a discrete AT that acts in trans, whereas a type I PKS module minimally contains AT, acyl carrier protein (ACP), and ketosynthase (KS) domains. Structures of canonical type I PKS KS-AT didomains reveal structured linkers that connect the two domains. AT-less type I PKS KSs have remnants of these linkers, which have been hypothesized to be AT docking domains. Natural products produced by AT-less type I PKSs are very complex because of an increased representation of unique modifying domains. AT-less type I PKS KSs possess substrate specificity and fall into phylogenetic clades that correlate with their substrates, whereas canonical type I PKS KSs are monophyletic. We have solved crystal structures of seven AT-less type I PKS KS domains that represent various sequence clusters, revealing insight into the large structural and subtle amino acid residue differences that lead to unique active site topologies and substrate specificities. One set of structures represents a larger group of KS domains from both canonical and AT-less type I PKSs that accept amino acid-containing substrates. One structure has a partial AT-domain, revealing the structural consequences of a type I PKS KS evolving into an AT-less type I PKS KS. These structures highlight the structural diversity within the AT-less type I PKS KS family, and most important, provide a unique opportunity to study the molecular evolution of substrate specificity within the type I PKSs.


Subject(s)
Evolution, Molecular , Polyketide Synthases/chemistry , Crystallography, X-Ray , Polyketide Synthases/genetics , Protein Structure, Tertiary , Substrate Specificity
3.
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
4.
Methods Mol Biol ; 1140: 189-200, 2014.
Article in English | MEDLINE | ID: mdl-24590719

ABSTRACT

The growth of diffraction-quality single crystals is of primary importance in protein X-ray crystallography. Chemical modification of proteins can alter their surface properties and crystallization behavior. The Midwest Center for Structural Genomics (MCSG) has previously reported how reductive methylation of lysine residues in proteins can improve crystallization of unique proteins that initially failed to produce diffraction-quality crystals. Recently, this approach has been expanded to include ethylation and isopropylation in the MCSG protein crystallization pipeline. Applying standard methods, 180 unique proteins were alkylated and screened using standard crystallization procedures. Crystal structures of 12 new proteins were determined, including the first ethylated and the first isopropylated protein structures. In a few cases, the structures of native and methylated or ethylated states were obtained and the impact of reductive alkylation of lysine residues was assessed. Reductive methylation tends to be more efficient and produces the most alkylated protein structures. Structures of methylated proteins typically have higher resolution limits. A number of well-ordered alkylated lysine residues have been identified, which make both intermolecular and intramolecular contacts. The previous report is updated and complemented with the following new data; a description of a detailed alkylation protocol with results, structural features, and roles of alkylated lysine residues in protein crystals. These contribute to improved crystallization properties of some proteins.


Subject(s)
Crystallography, X-Ray , Molecular Biology/methods , Proteins/chemistry , Alkylation , Computational Biology , Crystallization , High-Throughput Screening Assays/methods
5.
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
6.
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
7.
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
8.
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
9.
Bioorg Med Chem Lett ; 20(22): 6587-91, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20870405

ABSTRACT

The Bcl-2 family of proteins plays a major role in the regulation of apoptosis, or programmed cell death. Overexpression of the anti-apoptotic members of this family (Bcl-2, Bcl-x(L), and Mcl-1) can render cancer cells resistant to chemotherapeutic agents and therefore these proteins are important targets for the development of new anti-cancer agents. Here we describe the discovery of a potent, highly selective, Bcl-2 inhibitor using SAR by NMR and structure-based drug design which could serve as a starting point for the development of a Bcl-2 selective anti-cancer agent. Such an agent would potentially overcome the Bcl-x(L) mediated thrombocytopenia observed with ABT-263.


Subject(s)
Magnetic Resonance Spectroscopy/methods , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Models, Molecular , Structure-Activity Relationship
10.
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
11.
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
12.
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
13.
J Biomol NMR ; 34(4): 221-7, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16645812

ABSTRACT

Fluorine atoms are often incorporated into drug molecules as part of the lead optimization process in order to improve affinity or modify undesirable metabolic and pharmacokinetic profiles. From an NMR perspective, the abundance of fluorinated drug leads provides an exploitable niche for structural studies using 19F NMR in the drug discovery process. As 19F has no interfering background signal from biological sources, 19F NMR studies of fluorinated drugs bound to their protein receptors can yield easily interpretable and unambiguous structural constraints. 19F can also be selectively incorporated into proteins to obtain additional constraints for structural studies. Despite these advantages, 19F NMR has rarely been exploited for structural studies due to its broad lines in macromolecules and their ligand complexes, leading to weak signals in 1H/19F heteronuclear NOE experiments. Here we demonstrate several different experimental strategies that use 19F NMR to obtain ligand-protein structural constraints for ligands bound to the anti-apoptotic protein Bcl-xL, a drug target for anti-cancer therapy. These examples indicate the applicability of these methods to typical structural problems encountered in the drug development process.


Subject(s)
Magnetic Resonance Spectroscopy , Nuclear Magnetic Resonance, Biomolecular/methods , bcl-X Protein/chemistry , Fluorine Compounds/chemistry , Fluorine Radioisotopes , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy/methods , Models, Molecular , Multiprotein Complexes/chemistry , Protein Binding , Protein Conformation
14.
J Am Chem Soc ; 126(43): 13948-53, 2004 Nov 03.
Article in English | MEDLINE | ID: mdl-15506755

ABSTRACT

A novel approach for detection of ligand binding to a protein in solid samples is described. Hydrated precipitates of the anti-apoptotic protein Bcl-xL show well-resolved (13)C-(13)C 2D solid-state NMR spectra that allow site-specific assignment of resonances for many residues in uniformly (13)C-enriched samples. Binding of a small peptide or drug-like organic molecule leads to changes in the chemical shift of resonances from multiple residues in the protein that can be monitored to characterize binding. Differential chemical shifts can be used to distinguish between direct protein-ligand contacts and small conformational changes of the protein induced by ligand binding. The agreement with prior solution-state NMR results indicates that the binding pocket in solid and liquid samples is similar for this protein. Advantages of different labeling schemes involving selective (13)C enrichment of methyl groups of Ala, Val, Leu, and Ile (Cdelta1) for characterizing protein-ligand interactions are also discussed. It is demonstrated that high-resolution solid-state NMR spectroscopy on uniformly or extensively (13)C-enriched samples has the potential to screen proteins of moderate size ( approximately 20 kDa) for ligand binding as hydrated solids. The results presented here suggest the possibility of using solid-state NMR to study ligand binding in proteins not amenable to solution NMR.


Subject(s)
Nuclear Magnetic Resonance, Biomolecular/methods , Proto-Oncogene Proteins c-bcl-2/chemistry , Binding Sites , Carbon Isotopes , Ligands , Nitrogen Isotopes , Protein Conformation , Proto-Oncogene Proteins c-bcl-2/metabolism , Reproducibility of Results , bcl-X Protein
15.
J Med Chem ; 47(20): 4851-7, 2004 Sep 23.
Article in English | MEDLINE | ID: mdl-15369388

ABSTRACT

Reversal of aberrant gene expression that is induced by the proto-oncogene c-myc is likely to be effective for treating a variety of tumors that rely on this pathway for growth. One strategy to down-regulate the c-myc pathway is to target transcription factors that regulate its own expression. A host of proteins act in coordination to regulate c-myc expression and any one of them are theoretical targets for small-molecule therapy. Experimentally, it has been shown that the far upstream element (FUSE) binding protein (FBP) is essential for c-myc expression, and reductions in FBP levels both reduce c-myc expression and correlate with slower cell growth. FBP binds to ssDNA by capturing exposed DNA bases in a hydrophobic pocket. This suggests that a small molecule could be designed to occupy this pocket and inhibit FBP function. Using a variety of screening methodologies, we have identified ligands that bind to the DNA binding pockets of the KH domains of FBP. Gel shift analyses using full length FBP and a related transcription factor confirm that a small-molecule lead compound inhibits DNA binding in a specific manner. The benzoylanthranilic acid compounds described here represent leads in the design of FBP inhibitors that can serve as useful tools in the study of c-myc regulation and in the development of therapeutics that target the c-myc pathway.


Subject(s)
Combinatorial Chemistry Techniques/methods , DNA-Binding Proteins/antagonists & inhibitors , Genes, myc , Magnetic Resonance Spectroscopy , Promoter Regions, Genetic , Binding Sites , DNA Helicases , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Drug Design , Humans , Inhibitory Concentration 50 , Ligands , Models, Molecular , Protein Conformation , Protein Structure, Tertiary , Proto-Oncogene Mas , RNA-Binding Proteins , Repetitive Sequences, Amino Acid , Structure-Activity Relationship
16.
Biochemistry ; 43(20): 6059-63, 2004 May 25.
Article in English | MEDLINE | ID: mdl-15147189

ABSTRACT

The severe acute respiratory syndrome (SARS) virus belongs to the Coronaviridea family of viruses. Its virion encodes several proteins including a replicase and four structural proteins. Here we describe the three-dimensional structure of the N-terminal domain of the SARS coronavirus (CoV) nucleocapsid protein. The protein consists of a five-stranded beta sheet with a folding topology distinct from other RNA-binding proteins. Single-stranded RNAs bind to the protein surface at the junction between a flexible, positively charged beta hairpin and the core structure. NMR-based screening was used to identify low molecular weight compounds that bind to this site.


Subject(s)
Nucleocapsid Proteins/chemistry , Protein Structure, Tertiary , Severe acute respiratory syndrome-related coronavirus/chemistry , Animals , Binding Sites , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Protein Folding , RNA/metabolism
17.
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
18.
Protein Sci ; 12(11): 2613-21, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14573872

ABSTRACT

Streptococcus pneumoniae is a major human pathogen that causes high mortality and morbidity and has developed resistance to many antibiotics. We show that the gene product from SP1603, identified from S. pneumoniae TIGR4, is a CMP kinase that is essential for bacterial growth. It represents an attractive drug target for the development of a novel antibiotic to overcome the problems of drug resistance development for this organism. Here we describe the three-dimensional solution structure of the S. pneumoniae CMP kinase as determined by NMR spectroscopy. The structure consists of eight alpha-helices and two beta-sheets that fold into the classical core domain, the substrate-binding domain, and the LID domain. The three domains of the protein pack together to form a central cavity for substrate-binding and enzymatic catalysis. The S. pneumoniae CMP kinase resembles the fold of the Escherichia coli homolog. An insertion of one residue is observed at the beta-turn in the substrate-binding domain of the S. pneumoniae CMP kinase when compared with the E. coli homolog. Chemical shift perturbations caused by the binding of CMP, CDP, and ATP revealed that CMP or CDP binds to the junction between the core and substrate-binding domains, whereas ATP binds to the junction between the core and LID domains. From NMR relaxation studies, we determined that the loops in the LID domain are highly mobile. These mobile loops could aid in the closing/opening of the LID domain during enzyme catalysis.


Subject(s)
Bacterial Proteins/metabolism , Nucleoside-Phosphate Kinase/chemistry , Streptococcus pneumoniae/enzymology , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Escherichia coli/enzymology , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Nucleoside-Phosphate Kinase/genetics , Nucleoside-Phosphate Kinase/metabolism , Protein Conformation , Protein Structure, Tertiary , Sequence Alignment , Solutions , Streptococcus pneumoniae/genetics
19.
J Biomol Screen ; 7(5): 429-32, 2002 Oct.
Article in English | MEDLINE | ID: mdl-14599358

ABSTRACT

A strategy is described for the development of high-throughput screening assays against targets of unknown function that involves the use of nuclear magnetic resonance (NMR) spectroscopy. Using this approach, molecules that bind to the protein target are identified from an NMR-based screen of a library of substrates, cofactors, and other compounds that are known to bind to many proteins and enzymes. Once a ligand has been discovered, a fluorescent or radiolabeled analog of the ligand is synthesized that can be used in a high-throughput screen. The approach is illustrated in the development of a high-throughput screening assay against HI-0033, a conserved protein from Haemophilus influenzae whose function is currently unknown. Adenosine was found to bind to HI-0033 by NMR, and fluorescent analogs were rapidly identified that bound to HI-0033 in the submicromolar range. Using these fluorescent compounds, a fluorescence polarization assay was developed that is suitable for high-throughput screening and obtaining detailed structure-activity relationships for lead optimization.


Subject(s)
Bacterial Proteins/analysis , Biological Assay/methods , Combinatorial Chemistry Techniques/methods , Magnetic Resonance Spectroscopy/methods , Bacterial Proteins/metabolism , Haemophilus influenzae/chemistry , Haemophilus influenzae/metabolism , Ligands , Spectrometry, Fluorescence/methods
SELECTION OF CITATIONS
SEARCH DETAIL