Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Chem Inf Model ; 61(6): 2937-2956, 2021 06 28.
Article in English | MEDLINE | ID: mdl-34101460

ABSTRACT

Three-dimensional (3D) maps of the hydropathic environments of protein amino acid residues are information-rich descriptors of preferred conformations, interaction types and energetics, and solvent accessibility. The interactions made by each residue are the primary factor for rotamer selection and the secondary, tertiary, and even quaternary protein structure. Our evolving basis set of environmental data for each residue type can be used to understand the protein structure. This work focuses on the aromatic residues phenylalanine, tyrosine, and tryptophan and their structural roles. We calculated and analyzed side chain-to-environment 3D maps for over 70,000 residues of these three types that reveal, with respect to hydrophobic and polar interactions, the environment around each. After binning with backbone ϕ/ψ and side chain χ1, we clustered each bin by 3D similarities between map-map pairs. For each of the three residue types, four bins were examined in detail: one in the ß-pleat, two in the right-hand α-helix, and one in the left-hand α-helix regions of the Ramachandran plot. For high degrees of side chain burial, encapsulation of the side chain by hydrophobic interactions is ubiquitous. The more solvent-exposed side chains are more likely to be involved in polar interactions with their environments. Evidence for π-π interactions was observed in about half of the residues surveyed [phenylalanine (PHE): 53.3%, tyrosine (TYR): 34.1%, and tryptophan (TRP): 55.7%], but on an energy basis, this contributed to only ∼4% of the total. Evidence for π-cation interactions was observed in 14.1% of PHE, 8.3% of TYR, and 26.8% of TRP residues, but on an energy basis, this contributed to only ∼1%. This recognition of even these subtle interactions in the 3D hydropathic environment maps is key support for our interaction homology paradigm of protein structure elucidation and possibly prediction.


Subject(s)
Phenylalanine , Tyrosine , Cations , Proteins , Tryptophan
2.
Acta Crystallogr D Struct Biol ; 75(Pt 4): 437-450, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30988260

ABSTRACT

Although the HcpR regulator plays a vital step in initiation of the nitrosative stress response in many Gram-negative anaerobic bacteria, the molecular mechanisms that it uses to mediate gas sensing are not well understood. Here, a 2.6 Šresolution crystal structure of the N-terminal sensing domain of the anaerobic periodontopathogen Porphyromonas gingivalis HcpR is presented. The protein has classical features of the regulators belonging to the FNR-CRP family and contains a hydrophobic pocket in its N-terminal sensing domain. It is shown that heme bound to HcpR exhibits heme iron as a hexacoordinate system in the absence of nitric oxide (NO) and that upon nitrosylation it transitions to a pentacoordinate system. Finally, small-angle X-ray scattering experiments on full-length HcpR reveal that the C-terminal DNA-binding domain of HcpR has a high degree of interdomain flexibility.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Heme/metabolism , Nitric Oxide/metabolism , Nitrosative Stress , Porphyromonas gingivalis/metabolism , Amino Acid Sequence , Animals , Catalytic Domain , Crystallography, X-Ray/methods , Models, Molecular , Molecular Structure , Porphyromonas gingivalis/physiology , Protein Conformation , Sequence Homology
3.
Nucleic Acids Res ; 43(6): 3100-13, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25753662

ABSTRACT

The MBD2-NuRD (Nucleosome Remodeling and Deacetylase) complex is an epigenetic reader of DNA methylation that regulates genes involved in normal development and neoplastic diseases. To delineate the architecture and functional interactions of the MBD2-NuRD complex, we previously solved the structures of MBD2 bound to methylated DNA and a coiled-coil interaction between MBD2 and p66α that recruits the CHD4 nucleosome remodeling protein to the complex. The work presented here identifies novel structural and functional features of a previously uncharacterized domain of MBD2 (MBD2IDR). Biophysical analyses show that the MBD2IDR is an intrinsically disordered region (IDR). However, despite this inherent disorder, MBD2IDR increases the overall binding affinity of MBD2 for methylated DNA. MBD2IDR also recruits the histone deacetylase core components (RbAp48, HDAC2 and MTA2) of NuRD through a critical contact region requiring two contiguous amino acid residues, Arg(286) and Leu(287). Mutating these residues abrogates interaction of MBD2 with the histone deacetylase core and impairs the ability of MBD2 to repress the methylated tumor suppressor gene PRSS8 in MDA-MB-435 breast cancer cells. These findings expand our knowledge of the multi-dimensional interactions of the MBD2-NuRD complex that govern its function.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/chemistry , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Amino Acid Sequence , Animals , Cell Line , DNA Methylation , DNA-Binding Proteins/genetics , Epigenesis, Genetic , Gene Knockdown Techniques , HEK293 Cells , Humans , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/metabolism , Kinetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
4.
Nucleic Acids Res ; 42(17): 11218-32, 2014.
Article in English | MEDLINE | ID: mdl-25183517

ABSTRACT

Unlike other members of the methyl-cytosine binding domain (MBD) family, MBD4 serves as a potent DNA glycosylase in DNA mismatch repair specifically targeting mCpG/TpG mismatches arising from spontaneous deamination of methyl-cytosine. The protein contains an N-terminal MBD (MBD4MBD) and a C-terminal glycosylase domain (MBD4GD) separated by a long linker. This arrangement suggests that the MBD4MBD either directly augments enzymatic catalysis by the MBD4GD or targets the protein to regions enriched for mCpG/TpG mismatches. Here we present structural and dynamic studies of MBD4MBD bound to dsDNA. We show that MBD4MBD binds with a modest preference for mCpG as compared to mismatch, unmethylated and hydroxymethylated DNA. We find that while MBD4MBD exhibits slow exchange between molecules of DNA (intermolecular exchange), the domain exhibits fast exchange between two sites in the same molecule of dsDNA (intramolecular exchange). Introducing a single-strand defect between binding sites does not greatly reduce the intramolecular exchange rate, consistent with a local hopping mechanism for moving along the DNA. These results support a model in which the MBD4MBD4 targets the intact protein to (m)CpG islands and promotes scanning by rapidly exchanging between successive mCpG sites which facilitates repair of nearby mCpG/TpG mismatches by the glycosylase domain.


Subject(s)
Base Pair Mismatch , CpG Islands , DNA Methylation , DNA/chemistry , Endodeoxyribonucleases/chemistry , Binding Sites , DNA/metabolism , Endodeoxyribonucleases/metabolism , Models, Molecular , Protein Structure, Tertiary , Sodium Chloride/chemistry
5.
J Biol Chem ; 289(3): 1294-302, 2014 Jan 17.
Article in English | MEDLINE | ID: mdl-24307175

ABSTRACT

Although highly homologous to other methylcytosine-binding domain (MBD) proteins, MBD3 does not selectively bind methylated DNA, and thus the functional role of MBD3 remains in question. To explore the structural basis of its binding properties and potential function, we characterized the solution structure and binding distribution of the MBD3 MBD on hydroxymethylated, methylated, and unmethylated DNA. The overall fold of this domain is very similar to other MBDs, yet a key loop involved in DNA binding is more disordered than previously observed. Specific recognition of methylated DNA constrains the structure of this loop and results in large chemical shift changes in NMR spectra. Based on these spectral changes, we show that MBD3 preferentially localizes to methylated and, to a lesser degree, unmethylated cytosine-guanosine dinucleotides (CpGs), yet does not distinguish between hydroxymethylated and unmethylated sites. Measuring residual dipolar couplings for the different bound states clearly shows that the MBD3 structure does not change between methylation-specific and nonspecific binding modes. Furthermore, residual dipolar couplings measured for MBD3 bound to methylated DNA can be described by a linear combination of those for the methylation and nonspecific binding modes, confirming the preferential localization to methylated sites. The highly homologous MBD2 protein shows similar but much stronger localization to methylated as well as unmethylated CpGs. Together, these data establish the structural basis for the relative distribution of MBD2 and MBD3 on genomic DNA and their observed occupancy at active and inactive CpG-rich promoters.


Subject(s)
Avian Proteins/chemistry , CpG Islands/physiology , DNA-Binding Proteins/chemistry , DNA/chemistry , Animals , Avian Proteins/genetics , Avian Proteins/metabolism , Chickens , DNA/genetics , DNA/metabolism , DNA Methylation/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Protein Binding/physiology , Protein Structure, Tertiary
6.
Comput Biol Chem ; 47: 126-41, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24076743

ABSTRACT

The importance of protein-protein interactions (PPIs) is becoming increasingly appreciated, as these interactions lie at the core of virtually every biological process. Small molecule modulators that target PPIs are under exploration as new therapies. One of the greatest obstacles faced in crystallographically determining the 3D structures of proteins is coaxing the proteins to form "artificial" PPIs that lead to uniform crystals suitable for X-ray diffraction. This work compares interactions formed naturally, i.e., "biological", with those artificially formed under crystallization conditions or "non-biological". In particular, a detailed analysis of water molecules at the interfaces of high-resolution (≤2.30 Å) X-ray crystal structures of protein-protein complexes, where 140 are biological protein-protein complex structures and 112 include non-biological protein-protein interfaces, was carried out using modeling tools based on the HINT forcefield. Surprisingly few and relatively subtle differences were observed between the two types of interfaces: (i) non-biological interfaces are more polar than biological interfaces, yet there is better organized hydrogen bonding at the latter; (ii) biological associations rely more on water-mediated interactions with backbone atoms compared to non-biological associations; (iii) aromatic/planar residues play a larger role in biological associations with respect to water, and (iv) Lys has a particularly large role at non-biological interfaces. A support vector machines (SVMs) classifier using descriptors from this study was devised that was able to correctly classify 84% of the two interface types.


Subject(s)
Proteins/chemistry , Water/chemistry , Crystallization , Models, Molecular , Protein Binding , Proteins/metabolism , Water/metabolism
7.
Biomed Res Int ; 2013: 458571, 2013.
Article in English | MEDLINE | ID: mdl-23956983

ABSTRACT

Serine hydroxymethyltransferase catalyzes the reversible interconversion of L-serine and glycine with transfer of one-carbon groups to and from tetrahydrofolate. Active site residue Thr254 is known to be involved in the transaldimination reaction, a crucial step in the catalytic mechanism of all pyridoxal 5'-phosphate- (PLP-) dependent enzymes, which determines binding of substrates and release of products. In order to better understand the role of Thr254, we have expressed, characterized, and determined the crystal structures of rabbit cytosolic serine hydroxymethyltransferase T254A and T254C mutant forms, in the absence and presence of substrates. These mutants accumulate a kinetically stable gem-diamine intermediate, and their crystal structures show differences in the active site with respect to wild type. The kinetic and crystallographic data acquired with mutant enzymes permit us to infer that conversion of gem-diamine to external aldimine is significantly slowed because intermediates are trapped into an anomalous position by a misorientation of the PLP ring, and a new energy barrier hampers the transaldimination reaction. This barrier likely arises from the loss of the stabilizing hydrogen bond between the hydroxymethyl group of Thr254 and the ε -amino group of active site Lys257, which stabilizes the external aldimine intermediate in wild type SHMTs.


Subject(s)
Catalysis , Catalytic Domain/genetics , Glycine Hydroxymethyltransferase/chemistry , Structure-Activity Relationship , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , Gene Expression , Glycine Hydroxymethyltransferase/metabolism , Hydrogen Bonding , Kinetics , Mutagenesis, Site-Directed , Mutation , Pyridoxal Phosphate/chemistry , Pyridoxal Phosphate/metabolism , Rabbits
8.
Biochemistry ; 51(1): 466-74, 2012 Jan 10.
Article in English | MEDLINE | ID: mdl-22142337

ABSTRACT

The KsgA methyltransferase is universally conserved and plays a key role in regulating ribosome biogenesis. KsgA has a complex reaction mechanism, transferring a total of four methyl groups onto two separate adenosine residues, A1518 and A1519, in the small subunit rRNA. This means that the active site pocket must accept both adenosine and N(6)-methyladenosine as substrates to catalyze formation of the final product N(6),N(6)-dimethyladenosine. KsgA is related to DNA adenosine methyltransferases, which transfer only a single methyl group to their target adenosine residue. We demonstrate that part of the discrimination between mono- and dimethyltransferase activity lies in a single residue in the active site, L114; this residue is part of a conserved motif, known as motif IV, which is common to a large group of S-adenosyl-L-methionine-dependent methyltransferases. Mutation of the leucine to a proline mimics the sequence found in DNA methyltransferases. The L114P mutant of KsgA shows diminished overall activity, and its ability to methylate the N(6)-methyladenosine intermediate to produce N(6),N(6)-dimethyladenosine is impaired; this is in contrast to a second active site mutation, N113A, which diminishes activity to a level comparable to L114P without affecting the methylation of N(6)-methyladenosine. We discuss the implications of this work for understanding the mechanism of KsgA's multiple catalytic steps.


Subject(s)
Escherichia coli K12/enzymology , Escherichia coli Proteins/chemistry , Methyltransferases/chemistry , Adenosine/chemistry , Adenosine/genetics , Adenosine/metabolism , Amino Acid Sequence , Catalytic Domain/genetics , Crystallography, X-Ray , DNA Methylation , Escherichia coli K12/genetics , Escherichia coli Proteins/genetics , Humans , Methyltransferases/deficiency , Methyltransferases/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding/genetics , Ribosome Subunits, Small, Bacterial/enzymology , Ribosome Subunits, Small, Bacterial/genetics , Substrate Specificity/genetics
9.
PLoS One ; 6(9): e24712, 2011.
Article in English | MEDLINE | ID: mdl-21961043

ABSTRACT

BACKGROUND: There is a great interest in understanding and exploiting protein-protein associations as new routes for treating human disease. However, these associations are difficult to structurally characterize or model although the number of X-ray structures for protein-protein complexes is expanding. One feature of these complexes that has received little attention is the role of water molecules in the interfacial region. METHODOLOGY: A data set of 4741 water molecules abstracted from 179 high-resolution (≤ 2.30 Å) X-ray crystal structures of protein-protein complexes was analyzed with a suite of modeling tools based on the HINT forcefield and hydrogen-bonding geometry. A metric termed Relevance was used to classify the general roles of the water molecules. RESULTS: The water molecules were found to be involved in: a) (bridging) interactions with both proteins (21%), b) favorable interactions with only one protein (53%), and c) no interactions with either protein (26%). This trend is shown to be independent of the crystallographic resolution. Interactions with residue backbones are consistent for all classes and account for 21.5% of all interactions. Interactions with polar residues are significantly more common for the first group and interactions with non-polar residues dominate the last group. Waters interacting with both proteins stabilize on average the proteins' interaction (-0.46 kcal mol(-1)), but the overall average contribution of a single water to the protein-protein interaction energy is unfavorable (+0.03 kcal mol(-1)). Analysis of the waters without favorable interactions with either protein suggests that this is a conserved phenomenon: 42% of these waters have SASA ≤ 10 Å(2) and are thus largely buried, and 69% of these are within predominantly hydrophobic environments or "hydrophobic bubbles". Such water molecules may have an important biological purpose in mediating protein-protein interactions.


Subject(s)
Models, Molecular , Protein Structure, Tertiary , Proteins/chemistry , Water/chemistry , Algorithms , Amino Acid Motifs , Amino Acids/chemistry , Amino Acids/classification , Amino Acids/metabolism , Cluster Analysis , Crystallography, X-Ray , Humans , Hydrogen Bonding , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Models, Chemical , Molecular Structure , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Proteins/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/chemistry , Receptors, Transforming Growth Factor beta/metabolism , Solvents/chemistry , Surface Properties , Thermodynamics , Transforming Growth Factor beta3/chemistry , Transforming Growth Factor beta3/metabolism , Water/metabolism
10.
Biochemistry ; 50(32): 6973-82, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21736375

ABSTRACT

The serine protease thrombin plays multiple roles in many important physiological processes, especially coagulation, where it functions as both a pro- and anticoagulant. The polyanionic glycosaminoglycan heparin modulates thrombin's activity through binding at exosite II. Sucrose octasulfate (SOS) is often used as a surrogate for heparin, but it is not known whether it is an effective heparin mimic in its interaction with thrombin. We have characterized the interaction of SOS with thrombin in solution and determined a crystal structure of their complex. SOS binds thrombin with a K(d) of ~1.4 µM, comparable to that of the much larger polymeric heparin measured under the same conditions. Nonionic (hydrogen bonding) interactions make a larger contribution to thrombin binding of SOS than to heparin. SOS binding to exosite II inhibits thrombin's catalytic activity with high potency but with low efficacy. Analytical ultracentrifugation shows that bovine and human thrombins are monomers in solution in the presence of SOS, in contrast to their complexes with heparin, which are dimers. In the X-ray crystal structure, two molecules of SOS are bound nonequivalently to exosite II portions of a thrombin dimer, in contrast to the 1:2 stoichiometry of the heparin-thrombin complex, which has a different monomer association mode in the dimer. SOS and heparin binding to exosite II of thrombin differ on both chemical and structural levels and, perhaps most significantly, in thrombin inhibition. These differences may offer paths to the design of more potent exosite II binding, allosteric small molecules as modulators of thrombin function.


Subject(s)
Sucrose/analogs & derivatives , Thrombin/chemistry , Animals , Carbohydrate Sequence , Cattle , Crystallography, X-Ray , Heparin, Low-Molecular-Weight/chemistry , Humans , Models, Molecular , Molecular Sequence Data , Protein Binding , Spectrometry, Fluorescence , Sucrose/chemistry , Thermodynamics , Ultracentrifugation
11.
Nucleic Acids Res ; 39(15): 6741-52, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21531701

ABSTRACT

The epigenetic code of DNA methylation is interpreted chiefly by methyl cytosine binding domain (MBD) proteins which in turn recruit multiprotein co-repressor complexes. We previously isolated one such complex, MBD2-NuRD, from primary erythroid cells and have shown it contributes to embryonic/fetal ß-type globin gene silencing during development. This complex has been implicated in silencing tumor suppressor genes in a variety of human tumor cell types. Here we present structural details of chicken MBD2 bound to a methylated DNA sequence from the ρ-globin promoter to which it binds in vivo and mediates developmental transcriptional silencing in normal erythroid cells. While previous studies have failed to show sequence specificity for MBD2 outside of the symmetric mCpG, we find that this domain binds in a single orientation on the ρ-globin target DNA sequence. Further, we show that the orientation and affinity depends on guanine immediately following the mCpG dinucleotide. Dynamic analyses show that DNA binding stabilizes the central ß-sheet, while the N- and C-terminal regions of the protein maintain mobility. Taken together, these data lead to a model in which DNA binding stabilizes the MBD2 structure and that binding orientation and affinity is influenced by the DNA sequence surrounding the central mCpG.


Subject(s)
Avian Proteins/chemistry , DNA Methylation , DNA-Binding Proteins/chemistry , DNA/chemistry , Animals , Base Sequence , Chickens , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Structure, Tertiary
12.
Proc Natl Acad Sci U S A ; 108(18): 7487-92, 2011 May 03.
Article in English | MEDLINE | ID: mdl-21490301

ABSTRACT

Nucleosome remodeling complexes comprise several large families of chromatin modifiers that integrate multiple epigenetic control signals to play key roles in cell type-specific transcription regulation. We previously isolated a methyl-binding domain protein 2 (MBD2)-containing nucleosome remodeling and deacetylation (NuRD) complex from primary erythroid cells and showed that MBD2 contributes to DNA methylation-dependent embryonic and fetal ß-type globin gene silencing during development in vivo. Here we present structural and biophysical details of the coiled-coil interaction between MBD2 and p66α, a critical component of the MBD2-NuRD complex. We show that enforced expression of the isolated p66α coiled-coil domain relieves MBD2-mediated globin gene silencing and that the expressed peptide interacts only with a subset of components of the MBD2-NuRD complex that does not include native p66α or Mi-2. These results demonstrate the central importance of the coiled-coil interaction and suggest that MBD2-dependent DNA methylation-driven gene silencing can be disrupted by selectively targeting this coiled-coil complex.


Subject(s)
DNA-Binding Proteins/metabolism , Epigenesis, Genetic/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Models, Molecular , Repressor Proteins/metabolism , Blotting, Western , DNA Methylation/genetics , DNA Primers/genetics , Gene Silencing , Humans , Immunoprecipitation , RNA Interference
13.
PLoS One ; 6(1): e15920, 2011 Jan 05.
Article in English | MEDLINE | ID: mdl-21246043

ABSTRACT

BACKGROUND: The quality of X-ray crystallographic models for biomacromolecules refined from data obtained at high-resolution is assured by the data itself. However, at low-resolution, >3.0 Å, additional information is supplied by a forcefield coupled with an associated refinement protocol. These resulting structures are often of lower quality and thus unsuitable for downstream activities like structure-based drug discovery. METHODOLOGY: An X-ray crystallography refinement protocol that enhances standard methodology by incorporating energy terms from the HINT (Hydropathic INTeractions) empirical forcefield is described. This protocol was tested by refining synthetic low-resolution structural data derived from 25 diverse high-resolution structures, and referencing the resulting models to these structures. The models were also evaluated with global structural quality metrics, e.g., Ramachandran score and MolProbity clashscore. Three additional structures, for which only low-resolution data are available, were also re-refined with this methodology. RESULTS: The enhanced refinement protocol is most beneficial for reflection data at resolutions of 3.0 Å or worse. At the low-resolution limit, ≥4.0 Å, the new protocol generated models with Cα positions that have RMSDs that are 0.18 Å more similar to the reference high-resolution structure, Ramachandran scores improved by 13%, and clashscores improved by 51%, all in comparison to models generated with the standard refinement protocol. The hydropathic forcefield terms are at least as effective as Coulombic electrostatic terms in maintaining polar interaction networks, and significantly more effective in maintaining hydrophobic networks, as synthetic resolution is decremented. Even at resolutions ≥4.0 Å, these latter networks are generally native-like, as measured with a hydropathic interactions scoring tool.


Subject(s)
Crystallography, X-Ray/methods , Hydrophobic and Hydrophilic Interactions , Crystallography, X-Ray/standards , Macromolecular Substances , Models, Molecular
14.
Biochemistry ; 49(12): 2697-704, 2010 Mar 30.
Article in English | MEDLINE | ID: mdl-20163168

ABSTRACT

The KsgA/Dim1 family of proteins is intimately involved in ribosome biogenesis in all organisms. These enzymes share the common function of dimethylating two adenosine residues near the 3'-OH end of the small subunit rRNA; orthologs in the three kingdoms, along with eukaryotic organelles, have evolved additional functions in rRNA processing, ribosome assembly, and, surprisingly, transcription in mitochondria. The methyltransferase reaction is intriguingly elaborate. The enzymes can bind to naked small subunit rRNA but cannot methylate their target bases until a subset of ribosomal proteins have bound and the nascent subunit has reached a certain level of maturity. Once this threshold is reached, the enzyme must stabilize two adenosines into the active site at separate times and two methyl groups must be transferred to each adenosine, with concomitant exchanges of the product S-adenosyl-l-homocysteine and the methyl donor substrate S-adenosyl-l-methionine. A detailed molecular understanding of this mechanism is currently lacking. Structural analysis of the interactions between the enzyme and substrate will aid in this understanding. Here we present the structure of KsgA from Methanocaldococcus jannaschii in complex with several ligands, including the first structure of S-adenosyl-l-methionine bound to a KsgA/Dim1 enzyme in a catalytically productive way. We also discuss the inability thus far to determine a structure of a target adenosine bound in its active site.


Subject(s)
Adenosine/chemistry , Fatty Acids/pharmacology , Methyltransferases/chemistry , Protein Conformation , RNA, Ribosomal/chemistry , Base Sequence , Binding Sites/genetics , Catalytic Domain , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors , Methionine/analogs & derivatives , Methionine/chemistry , Methionine/metabolism , Methyltransferases/antagonists & inhibitors , Methyltransferases/metabolism , Models, Molecular , Molecular Sequence Data , RNA, Ribosomal/metabolism , RNA, Ribosomal, 16S , S-Adenosylmethionine/metabolism , Structure-Activity Relationship , Substrate Specificity
15.
J Comput Aided Mol Des ; 23(9): 621-32, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19554265

ABSTRACT

A public web server performing computational titration at the active site in a protein-ligand complex has been implemented. This calculation is based on the Hydropathic interaction noncovalent force field. From 3D coordinate data for the protein, ligand and bridging waters (if available), the server predicts the best combination of protonation states for each ionizable residue and/or ligand functional group as well as the Gibbs free energy of binding for the ionization-optimized protein-ligand complex. The 3D structure for the modified molecules is available as output. In addition, a graph depicting how this energy changes with acidity, i.e., as a function of added protons, can be obtained. This data may prove to be of use in preparing models for virtual screening and molecular docking. A few illustrative examples are presented. In beta secretase (2va7) computational titration flipped the amide groups of Gln12 and Asn37 and protonated a ligand amine yielding an improvement of 6.37 kcal mol(-1) in the protein-ligand binding score. Protonation of Glu139 in mutant HIV-1 reverse transcriptase (2opq) allows a water bridge between the protein and inhibitor that increases the protein-ligand interaction score by 0.16 kcal mol(-1). In human sialidase NEU2 complexed with an isobutyl ether mimetic inhibitor (2f11) computational titration suggested that protonating Glu218, deprotonating Arg237, flipping the amide bond on Tyr334, and optimizing the positions of several other polar protons would increase the protein-ligand interaction score by 0.71 kcal mol(-1).


Subject(s)
Algorithms , Computational Biology/methods , Internet , Models, Chemical , Molecular Structure , Protein Binding , Protons , Thermodynamics , Amino Acids/chemistry , Amyloid Precursor Protein Secretases/chemistry , Amyloid Precursor Protein Secretases/metabolism , Computer Graphics , Crystallography, X-Ray , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/metabolism , Humans , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Ligands , Neuraminidase/antagonists & inhibitors , Neuraminidase/chemistry , Neuraminidase/metabolism , Solvents/chemistry , Titrimetry
16.
J Mol Biol ; 391(5): 884-93, 2009 Sep 04.
Article in English | MEDLINE | ID: mdl-19520088

ABSTRACT

The enzymes of the KsgA/Dim1 family are universally distributed throughout all phylogeny; however, structural and functional differences are known to exist. The well-characterized function of these enzymes is to dimethylate two adjacent adenosines of the small ribosomal subunit in the normal course of ribosome maturation, and the structures of KsgA from Escherichia coli and Dim1 from Homo sapiens and Plasmodium falciparum have been determined. To this point, no examples of archaeal structures have been reported. Here, we report the structure of Dim1 from the thermophilic archaeon Methanocaldococcus jannaschii. While it shares obvious similarities with the bacterial and eukaryotic orthologs, notable structural differences exist among the three members, particularly in the C-terminal domain. Previous work showed that eukaryotic and archaeal Dim1 were able to robustly complement for KsgA in E. coli. Here, we repeated similar experiments to test for complementarity of archaeal Dim1 and bacterial KsgA in Saccharomyces cerevisiae. However, neither the bacterial nor the archaeal ortholog could complement for the eukaryotic Dim1. This might be related to the secondary, non-methyltransferase function that Dim1 is known to play in eukaryotic ribosomal maturation. To further delineate regions of the eukaryotic Dim1 critical to its function, we created and tested KsgA/Dim1 chimeras. Of the chimeras, only one constructed with the N-terminal domain from eukaryotic Dim1 and the C-terminal domain from archaeal Dim1 was able to complement, suggesting that eukaryotic-specific Dim1 function resides in the N-terminal domain also, where few structural differences are observed between members of the KsgA/Dim1 family. Future work is required to identify those determinants directly responsible for Dim1 function in ribosome biogenesis. Finally, we have conclusively established that none of the methyl groups are critically important to growth in yeast under standard conditions at a variety of temperatures.


Subject(s)
Archaeal Proteins/chemistry , Archaeal Proteins/metabolism , Methanococcus/enzymology , Methyltransferases/chemistry , Methyltransferases/metabolism , Protein Structure, Tertiary , Amino Acid Sequence , Animals , Archaeal Proteins/genetics , Crystallography, X-Ray , Genetic Complementation Test , Humans , Methyltransferases/genetics , Models, Molecular , Molecular Sequence Data , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment
17.
Chem Biol ; 15(4): 402-12, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18420147

ABSTRACT

Mycobacterium tuberculosis FabH initiates type II fatty acid synthase-catalyzed formation of the long chain (C(16)-C(22)) acyl-coenzyme A (CoA) precursors of mycolic acids, which are major constituents of the bacterial cell envelope. Crystal structures of M. tuberculosis FabH (mtFabH) show the substrate binding site to be a buried, extended L-shaped channel with only a single solvent access portal. Entrance of an acyl-CoA substrate through the solvent portal would require energetically unfavorable reptational threading of the substrate to its reactive position. Using a class of FabH inhibitors, we have tested an alternative hypothesis that FabH exists in an "open" form during substrate binding and product release, and a "closed" form in which catalysis and intermediate steps occur. This hypothesis is supported by mass spectrometric analysis of the product profile and crystal structures of complexes of mtFabH with these inhibitors.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/chemistry , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Mycobacterium tuberculosis/enzymology , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/genetics , Acyl Coenzyme A/antagonists & inhibitors , Acyl Coenzyme A/metabolism , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Cysteine/metabolism , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Hydrophobic and Hydrophilic Interactions , Ligands , Models, Molecular , Mutation , Mycolic Acids/metabolism , Protein Binding , Solvents/chemistry , Spectrometry, Mass, Electrospray Ionization
18.
Chem Biodivers ; 4(11): 2578-92, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18027372

ABSTRACT

The similarity in structure of potassium (K(+)) channels from different families has been revealed by only recently available crystallographic 3D structural data. The hydropathic analysis presented in this work illuminates whether homologous residues perform the same functions in channels that use different gating mechanisms. We calculated and compared the hydropathic profiles of two K(+) channels, KcsA and Kv1.2 (the latter a member of the Shaker family), at their pore-forming domain. Quantitative information describing important interactions stabilizing the protein beyond obvious secondary-structure elements was extracted from the analysis and applied as a template for subsequent molecular-dynamics (MD) analyses. For example, two key groups of interactions, defining the turns that connect the transmembrane helices and responsible for the orientation of the pore helix, were identified. Our results also indicate that Asp(80) and Asp(379) play a similar role in stabilizing the P-loop of KcsA and Kv1.2, respectively, but to significantly different extents.


Subject(s)
Bacterial Proteins/chemistry , Hydrophobic and Hydrophilic Interactions , Potassium Channels/chemistry , Shaker Superfamily of Potassium Channels/chemistry , Amino Acid Sequence , Amino Acid Substitution/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Humans , Kv1.2 Potassium Channel/chemistry , Kv1.2 Potassium Channel/metabolism , Models, Biological , Molecular Sequence Data , Potassium Channels/genetics , Potassium Channels/metabolism , Protein Binding , Sequence Homology, Amino Acid , Shaker Superfamily of Potassium Channels/genetics , Shaker Superfamily of Potassium Channels/metabolism , Thermodynamics
19.
Chem Biol ; 13(5): 539-48, 2006 May.
Article in English | MEDLINE | ID: mdl-16720275

ABSTRACT

Noncovalent interactions are ubiquitous in ternary systems involving metal ions, DNA/RNA, and proteins and represent a structural motif for design of selective inhibitors of biological function. This contribution shows that small molecules containing platinated purine nucleobases mimic the natural DNA(RNA)-tryptophan recognition interaction of zinc finger peptides, specifically the C-terminal finger of HIV NCp7 protein. Interaction with platinum results in Zn ejection from the peptide accompanied by loss of tertiary structure. Targeting the NCp7-DNA interaction for drug design represents a conceptual advance over electrophiles designed for chemical attack on the zinc finger alone. These results demonstrate examples of a new platinum structural class targeting specific biological processes, distinct from the bifunctional DNA-DNA binding of cytotoxic agents like cisplatin. The results confirm the validity of a chemical biological approach for metallodrug design for selective ternary DNA(RNA)-protein interactions.


Subject(s)
DNA/metabolism , Zinc Fingers , Amino Acid Sequence , Capsid Proteins/metabolism , Drug Design , Gene Products, gag/metabolism , Nuclear Magnetic Resonance, Biomolecular , Platinum/metabolism , Protein Binding , Spectrometry, Fluorescence , Spectrometry, Mass, Electrospray Ionization , Viral Proteins/metabolism , gag Gene Products, Human Immunodeficiency Virus
20.
Structure ; 14(2): 331-43, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16472752

ABSTRACT

X29, a 25 kDa Nudix hydrolase from Xenopus laevis that cleaves 5' caps from U8 snoRNA, crystallizes as a homodimeric apoenzyme. Manganese binds crystals of apo-X29 to form holo-X29 only in the presence of nucleot(s)ide. Structural changes in X29 on nucleo-t(s)ide-assisted Mn(+2) uptake account for the observed cooperativity of metal binding. Structures of X29 with GTP or m7GpppA show a different mode of ligand binding from that of other cap binding proteins and suggest a possible three- or four-metal Nudix reaction mechanism. The X29 dimer has no known RNA binding motif, but its striking surface dipolarity and unique structural features create a plausible RNA binding channel on the positive face of the protein. Because U8 snoRNP is essential for accumulation of mature 5.8S and 28S rRNA in vertebrate ribosome biogenesis, and cap structures are required for U8 stability in vivo, X29 could profoundly influence this fundamental cellular pathway.


Subject(s)
Manganese/chemistry , Models, Molecular , Pyrophosphatases/chemistry , RNA Caps/chemistry , RNA, Small Nuclear/chemistry , RNA, Small Nucleolar/chemistry , Xenopus Proteins/chemistry , Amino Acid Sequence , Apoenzymes/chemistry , Binding Sites , Crystallography, X-Ray , Dimerization , Guanosine Triphosphate/chemistry , Molecular Sequence Data , Pyrophosphatases/metabolism , RNA Cap Analogs/chemistry , RNA Cap Analogs/metabolism , RNA Cap-Binding Proteins/chemistry , RNA, Small Nuclear/metabolism , Sequence Alignment , Substrate Specificity , Xenopus Proteins/metabolism , Nudix Hydrolases
SELECTION OF CITATIONS
SEARCH DETAIL
...