49 results on '"Scarsdale JN"'
Search Results
2. Recent Advances in Structural Analysis of Gangliosides: Primary and Secondary Structures
- Author
-
Peter C. Demou, James H. Prestegard, Theodore A. W. Koerner, Scarsdale Jn, and Robert K. Yu
- Subjects
Bond length ,chemistry.chemical_classification ,Molecular geometry ,Computational chemistry ,Chemistry ,Molecule ,Moiety ,Sequence (biology) ,Nuclear Overhauser effect ,Nuclear magnetic resonance spectroscopy ,Oligosaccharide - Abstract
The growing interest in the biological function of cell surface glycosphingolipids (GSLs) has stimulated the constant search for new methods for analyzing their primary and secondary structures. To determine the primary structure of the oligosaccharide moiety of a GSL, it is necessary to establish the composition and configuration of its sugar residues, and the sequence and linkage sites of the oligosaccharide chain. This information has traditionally been obtained by application of a combination of such procedures as compositional analysis by gas-liquid chromatography, mass spectrometry, permethylation studies, Smith degradation, partial acid or enzyme hydrolysis, optical rotation measurements, etc. However, these techniques are relatively time-consuming, frequently require elaborate derivatization of the intact molecules, and access to many different instruments. Furthermore, these procedures do not afford secondary structural information (conformation) which is important in determining the biological activities of these compounds. Although x-ray crystallography has been used to provide accurate information about the conformation of a carbohydrate by measurement of bond lengths, bond angles, and interatomic distances, it is not certain whether the conformation that exists in a crystalline state is the one that is preferred in solution.
- Published
- 1984
- Full Text
- View/download PDF
3. Accumulation of a globo-series glycolipid having Gal alpha 1-3Gal in PC12h pheochromocytoma cells
- Author
-
Tadashi Miyatake, H. Kitagawa, Robert K. Yu, Scarsdale Jn, Mari Suzuki, Toshio Ariga, and Yoichiro Kuroda
- Subjects
Magnetic Resonance Spectroscopy ,Globoside ,Globosides ,Chemistry ,Adrenal Gland Neoplasms ,Alpha (ethology) ,Pheochromocytoma ,Fast atom bombardment ,Biochemistry ,Glycosphingolipids ,Mass Spectrometry ,Cell Line ,Rats ,carbohydrates (lipids) ,Glycolipid ,Exoglycosidase ,Proton NMR ,Animals ,lipids (amino acids, peptides, and proteins) ,Glycolipids ,Beta (finance) ,Two-dimensional nuclear magnetic resonance spectroscopy - Abstract
In a previous paper, we reported the presence of globoside as a major neutral glycolipid in PC12 pheochromocytoma cells [Ariga, T., Macala, L. J., Saito, M., Margolis, R. K., Greene, L. A., Margolis, R. U., & Yu, R. K. (1988) Biochemistry 27, 52-58]. Recently, we found that subcloned PC12h cells accumulated another unusual neutral glycolipid. In order to characterize this glycolipid, PC12h cells were subcutaneously transplanted into rats. The induced tumor tissue accumulated two major neutral glycolipids, which were purified by Iatrobeads column and preparative thin-layer chromatographies. One of the glycolipids was found to be globoside, and the other had a globotriaosyl structure with an additional terminal Gal alpha 1-3 residue. Its structure was determined by fast atom bombardment mass spectrometry, two-dimensional proton nuclear magnetic resonance spectrometry (2D NMR), permethylation study, sequential degradation with exoglycosidase, and mild acid hydrolysis to be Gal(alpha 1-3)Gal(alpha 1-4)Gal(beta 1-4)Glc(beta 1-1')Cer.
- Published
- 1988
4. 3D Interaction Homology: Hydropathic Analyses of the "π-Cation" and "π-π" Interaction Motifs in Phenylalanine, Tyrosine, and Tryptophan Residues.
- Author
-
Al Mughram MH, Catalano C, Bowry JP, Safo MK, Scarsdale JN, and Kellogg GE
- Subjects
- Cations, Proteins, Tryptophan, Phenylalanine, Tyrosine
- 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 χ
1 , 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 β-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.- Published
- 2021
- Full Text
- View/download PDF
5. Nitrosative stress sensing in Porphyromonas gingivalis: structure of and heme binding by the transcriptional regulator HcpR.
- Author
-
Belvin BR, Musayev FN, Burgner J, Scarsdale JN, Escalante CR, and Lewis JP
- Subjects
- Amino Acid Sequence, Animals, Catalytic Domain, Crystallography, X-Ray methods, Models, Molecular, Molecular Structure, Porphyromonas gingivalis physiology, Protein Conformation, Sequence Homology, Bacterial Proteins chemistry, Bacterial Proteins metabolism, Heme metabolism, Nitric Oxide metabolism, Nitrosative Stress, Porphyromonas gingivalis metabolism
- 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 Å resolution 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.
- Published
- 2019
- Full Text
- View/download PDF
6. An intrinsically disordered region of methyl-CpG binding domain protein 2 (MBD2) recruits the histone deacetylase core of the NuRD complex.
- Author
-
Desai MA, Webb HD, Sinanan LM, Scarsdale JN, Walavalkar NM, Ginder GD, and Williams DC Jr
- Subjects
- 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, DNA-Binding Proteins chemistry, DNA-Binding Proteins metabolism, Mi-2 Nucleosome Remodeling and Deacetylase Complex chemistry, Mi-2 Nucleosome Remodeling and Deacetylase Complex metabolism
- 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., (© The Author(s) 2015. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2015
- Full Text
- View/download PDF
7. Probing the dynamic distribution of bound states for methylcytosine-binding domains on DNA.
- Author
-
Cramer JM, Scarsdale JN, Walavalkar NM, Buchwald WA, Ginder GD, and Williams DC Jr
- Subjects
- 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, Avian Proteins chemistry, CpG Islands physiology, DNA chemistry, DNA-Binding Proteins chemistry
- 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.
- Published
- 2014
- Full Text
- View/download PDF
8. Solution structure and intramolecular exchange of methyl-cytosine binding domain protein 4 (MBD4) on DNA suggests a mechanism to scan for mCpG/TpG mismatches.
- Author
-
Walavalkar NM, Cramer JM, Buchwald WA, Scarsdale JN, and Williams DC Jr
- Subjects
- Binding Sites, DNA metabolism, Endodeoxyribonucleases metabolism, Models, Molecular, Protein Structure, Tertiary, Sodium Chloride chemistry, Base Pair Mismatch, CpG Islands, DNA chemistry, DNA Methylation, Endodeoxyribonucleases chemistry
- 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., (© The Author(s) 2014. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2014
- Full Text
- View/download PDF
9. Unintended consequences? Water molecules at biological and crystallographic protein-protein interfaces.
- Author
-
Ahmed MH, Habtemariam M, Safo MK, Scarsdale JN, Spyrakis F, Cozzini P, Mozzarelli A, and Kellogg GE
- Subjects
- Crystallization, Models, Molecular, Protein Binding, Proteins metabolism, Water metabolism, Proteins chemistry, Water chemistry
- 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., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
10. Structure-based mechanism for early PLP-mediated steps of rabbit cytosolic serine hydroxymethyltransferase reaction.
- Author
-
Di Salvo ML, Scarsdale JN, Kazanina G, Contestabile R, Schirch V, and Wright HT
- Subjects
- 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, Catalysis, Catalytic Domain genetics, Glycine Hydroxymethyltransferase chemistry, Structure-Activity Relationship
- 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.
- Published
- 2013
- Full Text
- View/download PDF
11. Control of substrate specificity by a single active site residue of the KsgA methyltransferase.
- Author
-
O'Farrell HC, Musayev FN, Scarsdale JN, and Rife JP
- Subjects
- 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, Escherichia coli K12 enzymology, Escherichia coli Proteins chemistry, Methyltransferases chemistry
- 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.
- Published
- 2012
- Full Text
- View/download PDF
12. Interaction of thrombin with sucrose octasulfate.
- Author
-
Desai BJ, Boothello RS, Mehta AY, Scarsdale JN, Wright HT, and Desai UR
- Subjects
- 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, Sucrose analogs & derivatives, Thrombin chemistry
- 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.
- Published
- 2011
- Full Text
- View/download PDF
13. Solution structure and dynamic analysis of chicken MBD2 methyl binding domain bound to a target-methylated DNA sequence.
- Author
-
Scarsdale JN, Webb HD, Ginder GD, and Williams DC Jr
- Subjects
- Animals, Base Sequence, Chickens, Models, Molecular, Nuclear Magnetic Resonance, Biomolecular, Protein Binding, Protein Structure, Tertiary, Avian Proteins chemistry, DNA chemistry, DNA Methylation, DNA-Binding Proteins chemistry
- 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 β-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 β-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.
- Published
- 2011
- Full Text
- View/download PDF
14. p66Alpha-MBD2 coiled-coil interaction and recruitment of Mi-2 are critical for globin gene silencing by the MBD2-NuRD complex.
- Author
-
Gnanapragasam MN, Scarsdale JN, Amaya ML, Webb HD, Desai MA, Walavalkar NM, Wang SZ, Zu Zhu S, Ginder GD, and Williams DC Jr
- Subjects
- Blotting, Western, DNA Methylation genetics, DNA Primers genetics, Gene Silencing, Humans, Immunoprecipitation, RNA Interference, DNA-Binding Proteins metabolism, Epigenesis, Genetic genetics, Mi-2 Nucleosome Remodeling and Deacetylase Complex metabolism, Models, Molecular, Repressor Proteins metabolism
- 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 β-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.
- Published
- 2011
- Full Text
- View/download PDF
15. Applying an empirical hydropathic forcefield in refinement may improve low-resolution protein X-ray crystal structures.
- Author
-
Koparde VN, Scarsdale JN, and Kellogg GE
- Subjects
- Crystallography, X-Ray standards, Macromolecular Substances, Models, Molecular, Crystallography, X-Ray methods, Hydrophobic and Hydrophilic Interactions
- 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.
- Published
- 2011
- Full Text
- View/download PDF
16. Bound water at protein-protein interfaces: partners, roles and hydrophobic bubbles as a conserved motif.
- Author
-
Ahmed MH, Spyrakis F, Cozzini P, Tripathi PK, Mozzarelli A, Scarsdale JN, Safo MA, and Kellogg GE
- Subjects
- 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, Models, Molecular, Protein Structure, Tertiary, Proteins chemistry, Water chemistry
- 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.
- Published
- 2011
- Full Text
- View/download PDF
17. Binding of adenosine-based ligands to the MjDim1 rRNA methyltransferase: implications for reaction mechanism and drug design.
- Author
-
O'Farrell HC, Musayev FN, Scarsdale JN, and Rife JP
- Subjects
- 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, Adenosine chemistry, Fatty Acids pharmacology, Methyltransferases chemistry, Protein Conformation, RNA, Ribosomal chemistry
- 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.
- Published
- 2010
- Full Text
- View/download PDF
18. Structural and functional divergence within the Dim1/KsgA family of rRNA methyltransferases.
- Author
-
Pulicherla N, Pogorzala LA, Xu Z, O Farrell HC, Musayev FN, Scarsdale JN, Sia EA, Culver GM, and Rife JP
- Subjects
- 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, Archaeal Proteins chemistry, Archaeal Proteins metabolism, Methanococcus enzymology, Methyltransferases chemistry, Methyltransferases metabolism, Protein Structure, Tertiary
- 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.
- Published
- 2009
- Full Text
- View/download PDF
19. Web application for studying the free energy of binding and protonation states of protein-ligand complexes based on HINT.
- Author
-
Bayden AS, Fornabaio M, Scarsdale JN, and Kellogg GE
- Subjects
- 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, Algorithms, Computational Biology methods, Internet, Models, Chemical, Molecular Structure, Protein Binding, Protons, Thermodynamics
- 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).
- Published
- 2009
- Full Text
- View/download PDF
20. Separate entrance and exit portals for ligand traffic in Mycobacterium tuberculosis FabH.
- Author
-
Sachdeva S, Musayev FN, Alhamadsheh MM, Scarsdale JN, Wright HT, and Reynolds KA
- Subjects
- 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, 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase chemistry, 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase metabolism, Mycobacterium tuberculosis enzymology
- 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.
- Published
- 2008
- Full Text
- View/download PDF
21. Hydropathic analysis and comparison of KcsA and Shaker potassium channels.
- Author
-
Peng Y, Scarsdale JN, and Kellogg GE
- Subjects
- 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, Bacterial Proteins chemistry, Hydrophobic and Hydrophilic Interactions, Potassium Channels chemistry, Shaker Superfamily of Potassium Channels chemistry
- 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.
- Published
- 2007
- Full Text
- View/download PDF
22. Targeting retroviral Zn finger-DNA interactions: a small-molecule approach using the electrophilic nature of trans-platinum-nucleobase compounds.
- Author
-
Anzellotti AI, Liu Q, Bloemink MJ, Scarsdale JN, and Farrell N
- Subjects
- 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, DNA metabolism, Zinc Fingers
- 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.
- Published
- 2006
- Full Text
- View/download PDF
23. Crystal structures of U8 snoRNA decapping nudix hydrolase, X29, and its metal and cap complexes.
- Author
-
Scarsdale JN, Peculis BA, and Wright HT
- Subjects
- 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, Manganese chemistry, Models, Molecular, Pyrophosphatases chemistry, RNA Caps chemistry, RNA, Small Nuclear chemistry, RNA, Small Nucleolar chemistry, Xenopus Proteins chemistry
- 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.
- Published
- 2006
- Full Text
- View/download PDF
24. Crystal structure of a substrate complex of Mycobacterium tuberculosis beta-ketoacyl-acyl carrier protein synthase III (FabH) with lauroyl-coenzyme A.
- Author
-
Musayev F, Sachdeva S, Scarsdale JN, Reynolds KA, and Wright HT
- Subjects
- 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase metabolism, Binding Sites, Catalysis, Crystallography, X-Ray, Cysteine chemistry, Cysteine genetics, Models, Molecular, Mutation, Protein Conformation, Substrate Specificity, 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase chemistry, Acyl Coenzyme A metabolism, Mycobacterium tuberculosis enzymology, Mycolic Acids metabolism
- Abstract
Beta-ketoacyl-acyl carrier protein synthase III (FabH) catalyzes a two step reaction that initiates the pathway of fatty acid biosynthesis in plants and bacteria. In Mycobacterium tuberculosis, FabH catalyzes extension of lauroyl, myristoyl and palmitoyl groups from which cell wall mycolic acids of the bacterium are formed. The first step of the reaction is an acyl group transfer from acyl-coenzyme A to the active-site cysteine of the enzyme; the second step is acyl chain extension by two carbon atoms through Claisen condensation with malonyl-acyl carrier protein. We have previously determined the crystal structure of a type II, dissociated M.tuberculosis FabH, which catalyzes extension of lauroyl, myristoyl and palmitoyl groups. Here we describe the first long-chain Michaelis substrate complex of a FabH, that of lauroyl-coenzyme A with a catalytically disabled Cys-->Ala mutant of M.tuberculosis FabH. An elongated channel extending from the mutated active-site cysteine defines the acyl group binding locus that confers unique acyl substrate specificity on M.tuberculosis FabH. CoA lies in a second channel, bound primarily through interactions of its nucleotide group at the enzyme surface. The apparent weak association of CoA in this complex may play a role in the binding and dissociation of long chain acyl-CoA substrates and products and poses questions pertinent to the mechanism of this enzyme.
- Published
- 2005
- Full Text
- View/download PDF
25. Crystals of X29, a Xenopus laevis U8 snoRNA-binding protein with nuclear decapping activity.
- Author
-
Peculis BA, Scarsdale JN, and Wright HT
- Subjects
- Animals, Cell Nucleolus metabolism, Crystallization, DNA, Complementary biosynthesis, DNA, Complementary genetics, Pyrophosphatases biosynthesis, RNA, Ribosomal biosynthesis, RNA, Small Nuclear metabolism, X-Ray Diffraction, Xenopus Proteins biosynthesis, Cell Nucleus metabolism, Pyrophosphatases chemistry, Xenopus Proteins chemistry, Xenopus laevis metabolism
- Abstract
Eukaryotic ribosome biosynthesis requires modification (methylation, pseudouridylation) and nucleolytic processing of precursor ribosomal RNAs in the nucleolus. The RNA components of the small nucleolar RNPs (snoRNAs) are essential for many of these events. One snoRNP, called U8, is necessary for maturation of 5.8S and 28S rRNA in vertebrates. In Xenopus laevis, U8 snoRNA was found to bind specifically and with high affinity to a protein called X29. X29 is a Nudix hydrolase, a nucleotide diphosphatase that removes the m(7)G and m(227)G caps from U8 and other RNAs. X29 requires an RNA as substrate and cap analogues are not substrates or inhibitors of cleavage. To study the determinants of X29 activity and its specificity for U8 RNA substrate, X29 was crystallized in an orthorhombic crystal form that diffracts to 2.1 A resolution.
- Published
- 2004
- Full Text
- View/download PDF
26. Crystal structure of KsgA, a universally conserved rRNA adenine dimethyltransferase in Escherichia coli.
- Author
-
O'Farrell HC, Scarsdale JN, and Rife JP
- Subjects
- Amino Acid Sequence, Crystallography, X-Ray, Methyltransferases metabolism, Models, Molecular, Molecular Sequence Data, Protein Conformation, Sequence Homology, Amino Acid, Substrate Specificity, Escherichia coli enzymology, Methyltransferases chemistry
- Abstract
The bacterial enzyme KsgA catalyzes the transfer of a total of four methyl groups from S-adenosyl-l-methionine (S-AdoMet) to two adjacent adenosine bases in 16S rRNA. This enzyme and the resulting modified adenosine bases appear to be conserved in all species of eubacteria, eukaryotes, and archaebacteria, and in eukaryotic organelles. Bacterial resistance to the aminoglycoside antibiotic kasugamycin involves inactivation of KsgA and resulting loss of the dimethylations, with modest consequences to the overall fitness of the organism. In contrast, the yeast ortholog, Dim1, is essential. In yeast, and presumably in other eukaryotes, the enzyme performs a vital role in pre-rRNA processing in addition to its methylating activity. Another ortholog has been discovered recently, h-mtTFB in human mitochondria, which has a second function; this enzyme is a nuclear-encoded mitochondrial transcription factor. The KsgA enzymes are homologous to another family of RNA methyltransferases, the Erm enzymes, which methylate a single adenosine base in 23S rRNA and confer resistance to the MLS-B group of antibiotics. Despite their sequence similarity, the two enzyme families have strikingly different levels of regulation that remain to be elucidated. We have crystallized KsgA from Escherichia coli and solved its structure to a resolution of 2.1A. The structure bears a strong similarity to the crystal structure of ErmC' from Bacillus stearothermophilus and a lesser similarity to sc-mtTFB, the Saccharomyces cerevisiae version of h-mtTFB. Comparison of the three crystal structures and further study of the KsgA protein will provide insight into this interesting group of enzymes.
- Published
- 2004
- Full Text
- View/download PDF
27. Crystallization and preliminary X-ray diffraction analysis of KsgA, a universally conserved RNA adenine dimethyltransferase in Escherichia coli.
- Author
-
O'Farrell HC, Musayev FN, Scarsdale JN, Wright HT, and Rife JP
- Subjects
- Cloning, Molecular, Crystallization, Models, Chemical, Recombinant Proteins chemistry, X-Ray Diffraction, Escherichia coli enzymology, Methyltransferases chemistry
- Abstract
The bacterial enzyme KsgA catalyzes the transfer of a total of four methyl groups from S-adenosylmethionine (SAM) to two adjacent adenosines in 16S rRNA. These modified adenosines are universally conserved in all species of eubacteria, eukaryotes and archaebacteria studied. Recombinant KsgA from Escherichia coli was overexpressed as a His-tagged fusion protein and purified. The recombinant protein was crystallized using PEG 4000 as a precipitant. The crystals belong to space group C2 and diffract X-rays to a resolution of 1.9 A. The unit-cell parameters are a = 173.9, b = 38.4, c = 83.0 A, beta = 90.0 degrees. Structure determination using the molecular-replacement method is at the early stages of refinement.
- Published
- 2003
- Full Text
- View/download PDF
28. Hydropathic analysis of the free energy differences in anthracycline antibiotic binding to DNA.
- Author
-
Cashman DJ, Scarsdale JN, and Kellogg GE
- Subjects
- Base Pairing, Base Sequence, Binding Sites, DNA chemistry, Doxorubicin analogs & derivatives, Doxorubicin chemistry, Doxorubicin metabolism, Models, Molecular, Antibiotics, Antineoplastic chemistry, Antibiotics, Antineoplastic metabolism, DNA metabolism
- Abstract
Molecular models of six anthracycline antibiotics and their complexes with 32 distinct DNA octamer sequences were created and analyzed using HINT (Hydropathic INTeractions) to describe binding. The averaged binding scores were then used to calculate the free energies of binding for comparison with experimentally determined values. In parsing our results based on specific functional groups of doxorubicin, our calculations predict a free energy contribution of -3.6 +/- 1.1 kcal x mol(-1) (experimental -2.5 +/- 0.5 kcal x mol(-1)) from the groove binding daunosamine sugar. The net energetic contribution of removing the hydroxyl at position C9 is -0.7 +/- 0.7 kcal x mol(-1) (-1.1 +/- 0.5 kcal x mol(-1)). The energetic contribution of the 3' amino group in the daunosamine sugar (when replaced with a hydroxyl group) is -3.7 +/- 1.1 kcal x mol(-1) (-0.7 +/- 0.5 kcal x mol(-1)). We propose that this large discrepancy may be due to uncertainty in the exact protonation state of the amine. The energetic contribution of the hydroxyl group at C14 is +0.4 +/- 0.6 kcal x mol(-1) (-0.9 +/- 0.5 kcal x mol(-1)), largely due to unfavorable hydrophobic interactions between the hydroxyl oxygen and the methylene groups of the phosphate backbone of the DNA. Also, there appears to be considerable conformational uncertainty in this region. This computational procedure calibrates our methodology for future analyses where experimental data are unavailable.
- Published
- 2003
- Full Text
- View/download PDF
29. Biosynthetic origin of hygromycin A.
- Author
-
Habib el-SE, Scarsdale JN, and Reynolds KA
- Subjects
- Anti-Bacterial Agents chemistry, Cinnamates chemistry, Enzyme Inhibitors pharmacology, Fermentation, Glycine pharmacology, Hygromycin B chemistry, Glyphosate, Anti-Bacterial Agents biosynthesis, Glycine analogs & derivatives, Hygromycin B analogs & derivatives, Hygromycin B biosynthesis, Streptomyces metabolism
- Abstract
Hygromycin A, an antibiotic produced by Streptomyces hygroscopicus, is an inhibitor of bacterial ribosomal peptidyl transferase. The antibiotic binds to the ribosome in a distinct but overlapping manner with other antibiotics and offers a different template for generation of new agents effective against multidrug-resistant pathogens. Reported herein are the results from a series of stable-isotope-incorporation studies demonstrating the biosynthetic origins of the three distinct structural moieties which comprise hygromycin A. Incorporation of [1-(13)C]mannose and intact incorporation of D-[1,2-(13)C(2)]glucose into the 6-deoxy-5-keto-D-arabino-hexofuranose moiety are consistent with a pathway in which mannose is converted to an activated L-fucose, via a 4-keto-6-deoxy-D-mannose intermediate, with a subsequent unusual mutation of the pyranose to the corresponding furanose. The aminocyclitol moiety was labeled by D-[1,2-(13)C(2)]glucose in a manner consistent with formation of myo-inositol and a subsequent unprecedented oxidation and transamination of the C-2 hydroxyl group to generate neo-inosamine-2. Incorporation of [carboxy-(13)C]-4-hydroxybenzoic acid and intact incorporation of [2,3-(13)C(2)]propionate are consistent with a polyketide synthase-type decarboxylation condensation to generate the 3,4-dihydroxy-alpha-methylcinnamic acid moiety of hygromycin A. No labeling of hygromycin A was observed when [3-(13)C]tyrosine, [3-(13)C]phenylalanine, or [carboxy-(13)C]benzoic acid was used, suggesting that the 4-hydroxybenzoic acid is derived directly from chorismic acid. Consistent with this hypothesis was the observation that hygromycin A titers could be reduced by addition of N-(phosphonomethyl)-glycine (an inhibitor of chorismic acid biosynthesis) and restored by coaddition of 4-hydroxybenzoic acid. The convergent biosynthetic pathway established for hygromycin A offers significant versatility for applying the techniques of combinatorial and directed biosynthesis to production of new antibiotics which target the ribosomal peptidyl transferase activity.
- Published
- 2003
- Full Text
- View/download PDF
30. Crystal structures of r(GGUCACAGCCC)2.
- Author
-
Kacer V, Scaringe SA, Scarsdale JN, and Rife JP
- Subjects
- Aminoglycosides chemistry, Crystallization, Crystallography, X-Ray, Hydrogen Bonding, Indicators and Reagents, Metals chemistry, Models, Molecular, Nucleic Acid Conformation, Oligoribonucleotides chemistry
- Abstract
Crystals of small RNAs, which regularly diffract to very high resolution, can often be readily obtained. Unfortunately, for some RNAs the conformations adopted in the crystalline form are different from those found in solution. For example, short RNAs that form hairpins in solution virtually never crystallize thus; rather, they form duplexes. Nevertheless, these unintended structures have contributed greatly to the understanding of RNA structure. In a similar occurrence, the homodimer r(GGUCACAGCCC)(2) has been crystallized from an 11-mer/12-mer heteroduplex, r(GGCUGAAGUCCG)/r(GGUCACAGCCC). This surprising phenomenon was observed under a variety of crystallization conditions. The structure of the homoduplex was determined from crystals that differed in the precipitant used and the type of metal present. In all cases, the resulting homoduplexes contain ten base pairings, of which the central six are non-canonical pairings. In two of the variants, ordered metal-binding sites were observed: two equivalent octacoordinate Tl(+) sites in one and two equivalent nanocoordinate Ba(2+) sites in another.
- Published
- 2003
- Full Text
- View/download PDF
31. Location of the pteroylpolyglutamate-binding site on rabbit cytosolic serine hydroxymethyltransferase.
- Author
-
Fu TF, Scarsdale JN, Kazanina G, Schirch V, and Wright HT
- Subjects
- Animals, Binding Sites, Cations, Crystallography, X-Ray, Glutamic Acid metabolism, Glutamine chemistry, Glycine Hydroxymethyltransferase metabolism, Kinetics, Ligands, Lysine chemistry, Models, Molecular, Mutagenesis, Site-Directed, Mutation, Protein Binding, Protein Conformation, Rabbits, Cytosol enzymology, Glycine Hydroxymethyltransferase chemistry, Pteroylpolyglutamic Acids chemistry
- Abstract
Serine hydroxymethyltransferase (SHMT; EC 2.1.2.1) catalyzes the reversible interconversion of serine and glycine with transfer of the serine side chain one-carbon group to tetrahydropteroylglutamate (H(4)PteGlu), and also the conversion of 5,10-methenyl-H(4)PteGlu to 5-formyl-H(4)PteGlu. In the cell, H(4)PteGlu carries a poly-gamma-glutamyl tail of at least 3 glutamyl residues that is required for physiological activity. This study combines solution binding and mutagenesis studies with crystallographic structure determination to identify the extended binding site for tetrahydropteroylpolyglutamate on rabbit cytosolic SHMT. Equilibrium binding and kinetic measurements of H(4)PteGlu(3) and H(4)PteGlu(5) with wild-type and Lys --> Gln or Glu site mutant homotetrameric rabbit cytosolic SHMTs identified lysine residues that contribute to the binding of the polyglutamate tail. The crystal structure of the enzyme in complex with 5-formyl-H(4)PteGlu(3) confirms the solution data and indicates that the conformation of the pteridine ring and its interactions with the enzyme differ slightly from those observed in complexes of the monoglutamate cofactor. The polyglutamate chain, which does not contribute to catalysis, exists in multiple conformations in each of the two occupied binding sites and appears to be bound by the electrostatic field created by the cationic residues, with only limited interactions with specific individual residues.
- Published
- 2003
- Full Text
- View/download PDF
32. Crystal structure of the Mycobacterium tuberculosis beta-ketoacyl-acyl carrier protein synthase III.
- Author
-
Scarsdale JN, Kazanina G, He X, Reynolds KA, and Wright HT
- Subjects
- 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase metabolism, Base Sequence, Catalysis, Crystallography, X-Ray, DNA Primers, Models, Molecular, Protein Conformation, 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase chemistry, Mycobacterium tuberculosis enzymology, Mycolic Acids metabolism
- Abstract
Mycolic acids (alpha-alkyl-beta-hydroxy long chain fatty acids) cover the surface of mycobacteria, and inhibition of their biosynthesis is an established mechanism of action for several key front-line anti-tuberculosis drugs. In mycobacteria, long chain acyl-CoA products (C(14)-C(26)) generated by a type I fatty-acid synthase can be used directly for the alpha-branch of mycolic acid or can be extended by a type II fatty-acid synthase to make the meromycolic acid (C(50)-C(56)))-derived component. An unusual Mycobacterium tuberculosis beta-ketoacyl-acyl carrier protein (ACP) synthase III (mtFabH) has been identified, purified, and shown to catalyze a Claisen-type condensation between long chain acyl-CoA substrates such as myristoyl-CoA (C(14)) and malonyl-ACP. This enzyme, presumed to play a key role in initiating meromycolic acid biosynthesis, was crystallized, and its structure was determined at 2.1-A resolution. The mtFabH homodimer is closely similar in topology and active-site structure to Escherichia coli FabH (ecFabH), with a CoA/malonyl-ACP-binding channel leading from the enzyme surface to the buried active-site cysteine residue. Unlike ecFabH, mtFabH contains a second hydrophobic channel leading from the active site. In the ecFabH structure, this channel is blocked by a phenylalanine residue, which constrains specificity to acetyl-CoA, whereas in mtFabH, this residue is a threonine, which permits binding of longer acyl chains. This same channel in mtFabH is capped by an alpha-helix formed adjacent to a 4-amino acid sequence insertion, which limits bound acyl chain length to 16 carbons. These observations offer a molecular basis for understanding the unusual substrate specificity of mtFabH and its probable role in regulating the biosynthesis of the two different length acyl chains required for generation of mycolic acids. This mtFabH presents a new target for structure-based design of novel antimycobacterial agents.
- Published
- 2001
- Full Text
- View/download PDF
33. Crystal structure at 2.4 A resolution of E. coli serine hydroxymethyltransferase in complex with glycine substrate and 5-formyl tetrahydrofolate.
- Author
-
Scarsdale JN, Radaev S, Kazanina G, Schirch V, and Wright HT
- Subjects
- Amino Acid Sequence, Animals, Aspartate Aminotransferases chemistry, Binding Sites, Crystallization, Crystallography, X-Ray, Dimerization, Glycine chemistry, Glycine Hydroxymethyltransferase genetics, Humans, Leucovorin chemistry, Models, Molecular, Molecular Sequence Data, Osmolar Concentration, Protein Structure, Quaternary, Pteroylpolyglutamic Acids metabolism, Pyridoxal Phosphate metabolism, Rabbits, Sequence Alignment, Solvents, Structure-Activity Relationship, Tetrahydrofolates metabolism, Escherichia coli enzymology, Glycine metabolism, Glycine Hydroxymethyltransferase chemistry, Glycine Hydroxymethyltransferase metabolism, Leucovorin metabolism
- Abstract
Serine hydroxymethyltransferase (EC 2.1.2.1), a member of the alpha-class of pyridoxal phosphate enzymes, catalyzes the reversible interconversion of serine and glycine, changing the chemical bonding at the C(alpha)-C(beta) bond of the serine side-chain mediated by the pyridoxal phosphate cofactor. Scission of the C(alpha)-C(beta) bond of serine substrate produces a glycine product and most likely formaldehyde, which reacts without dissociation with tetrahydropteroylglutamate cofactor. Crystal structures of the human and rabbit cytosolic serine hydroxymethyltransferases (SHMT) confirmed their close similarity in tertiary and dimeric subunit structure to each other and to aspartate aminotransferase, the archetypal alpha-class pyridoxal 5'-phosphate enzyme. We describe here the structure at 2.4 A resolution of Escherichia coli serine hydroxymethyltransferase in ternary complex with glycine and 5-formyl tetrahydropteroylglutamate, refined to an R-factor value of 17.4 % and R(free) value of 19.6 %. This structure reveals the interactions of both cofactors and glycine substrate with the enzyme. Comparison with the E. coli aspartate aminotransferase structure shows the distinctions in sequence and structure which define the folate cofactor binding site in serine hydroxymethyltransferase and the differences in orientation of the amino terminal arm, the evolution of which was necessary for elaboration of the folate binding site. Comparison with the unliganded rabbit cytosolic serine hydroxymethyltransferase structure identifies changes in the conformation of the enzyme, similar to those observed in aspartate aminotransferase, that probably accompany the binding of substrate. The tetrameric quaternary structure of liganded E. coli serine hydroxymethyltransferase also differs in symmetry and relative disposition of the functional tight dimers from that of the unliganded eukaryotic enzymes. SHMT tetramers have surface charge distributions which suggest distinctions in folate binding between eukaryotic and E. coli enzymes. The structure of the E. coli ternary complex provides the basis for a thorough investigation of its mechanism through characterization and structure determination of site mutants., (Copyright 2000 Academic Press.)
- Published
- 2000
- Full Text
- View/download PDF
34. Crystal structure of rabbit cytosolic serine hydroxymethyltransferase at 2.8 A resolution: mechanistic implications.
- Author
-
Scarsdale JN, Kazanina G, Radaev S, Schirch V, and Wright HT
- Subjects
- Amino Acid Sequence, Amino Sugars chemistry, Animals, Binding Sites, Crystallization, Crystallography, X-Ray, Dimerization, Humans, Hydrogen Bonding, Models, Molecular, Molecular Sequence Data, Pyridoxal Phosphate chemistry, Rabbits, Sequence Homology, Amino Acid, Sheep, Structure-Activity Relationship, Substrate Specificity, Cytosol enzymology, Glycine Hydroxymethyltransferase chemistry
- Abstract
Serine hydroxymethyltransferase (SHMT) catalyzes the reversible cleavage of serine to form glycine and single carbon groups that are essential for many biosynthetic pathways. SHMT requires both pyridoxal phosphate (PLP) and tetrahydropteroylpolyglutamate (H4PteGlun) as cofactors, the latter as a carrier of the single carbon group. We describe here the crystal structure at 2.8 A resolution of rabbit cytosolic SHMT (rcSHMT) in two forms: one with the PLP covalently bound as an aldimine to the Nepsilon-amino group of the active site lysine and the other with the aldimine reduced to a secondary amine. The rcSHMT structure closely resembles the structure of human SHMT, confirming its similarity to the alpha-class of PLP enzymes. The structures reported here further permit identification of changes in the PLP group that accompany formation of the geminal diamine complex, the first intermediate in the reaction pathway. On the basis of the current mechanism derived from solution studies and the properties of site mutants, we are able to model the binding of both the serine substrate and the H4PteGlun cofactor. This model explains the properties of several site mutants of SHMT and offers testable hypotheses for a more detailed mechanism of this enzyme.
- Published
- 1999
- Full Text
- View/download PDF
35. Identification and hydropathic characterization of structural features affecting sequence specificity for doxorubicin intercalation into DNA double-stranded polynucleotides.
- Author
-
Kellogg GE, Scarsdale JN, and Fornari FA Jr
- Subjects
- Base Pairing, Binding Sites, Hydrogen Bonding, Intercalating Agents chemistry, Ligands, Models, Molecular, Oligodeoxyribonucleotides chemistry, Substrate Specificity, Thermodynamics, DNA Adducts chemistry, Doxorubicin chemistry, Doxorubicin metabolism, Intercalating Agents metabolism, Oligodeoxyribonucleotides metabolism
- Abstract
The computer molecular modeling program HINT (Hydropathic INTeractions), an empirical hydropathic force field function that includes hydrogen bonding, coulombic and hydrophobic terms, was used to study sequence-selective doxorubicin binding/intercalation in the 64 unique CAxy, CGxy, TAxy, TGxy base pair quartet combinations. The CAAT quartet sequence is shown to have the highest binding score of the 64 combinations. Of the two regularly alternating polynucleotides, d(CGCGCG)2and d(TATATA)2, the HINT calculated binding scores reveal doxorubicin binds preferentially to d(TATATA)2. Although interactions of the chromophore with the DNA base pairs defining the intercalation site [I-1] [I+1] and the neighboring [I+2] base pair are predominant, the results obtained with HINT indicate that the base pair [I+3] contributes significantly to the sequence selectivity of doxorubicin by providing an additional hydrogen bonding opportunity for the N3' ammonium of the daunosamine sugar moiety in approximately 25% of the sequences. This observation, that interactions involving a base pair [I+3] distal to the intercalation site play a significant role in stabilizing/destabilizing the intercalation of doxorubicin into the various DNA sequences, has not been previously reported. In general terms, this work shows that molecular modeling and careful analysis of molecular interactions can have a significant role in designing and evaluating nucleotides and antineoplastic agents.
- Published
- 1998
- Full Text
- View/download PDF
36. Structural basis for serpin inhibitor activity.
- Author
-
Wright HT and Scarsdale JN
- Subjects
- Amino Acid Sequence, Binding Sites, Computer Graphics, Hydrogen Bonding, Models, Chemical, Models, Molecular, Molecular Sequence Data, Mutation, Plasminogen Activator Inhibitor 1 chemistry, Protein Binding, Protein Structure, Secondary, Serine Endopeptidases metabolism, Serine Proteinase Inhibitors genetics, Serine Proteinase Inhibitors metabolism, Serpins genetics, Serpins metabolism, alpha 1-Antitrypsin chemistry, alpha 1-Antitrypsin genetics, alpha 1-Antitrypsin metabolism, alpha-2-Antiplasmin chemistry, alpha-2-Antiplasmin genetics, alpha-2-Antiplasmin metabolism, Serine Proteinase Inhibitors chemistry, Serpins chemistry
- Abstract
The mechanism of formation and the structures of serpin-inhibitor complexes are not completely understood, despite detailed knowledge of the structures of a number of cleaved and uncleaved inhibitor, noninhibitor, and latent serpins. It has been proposed from comparison of inhibitor and noninhibitor serpins in the cleaved and uncleaved forms that insertion of strand s4A into preexisting beta-sheet A is a requirement for serpin inhibitor activity. We have investigated the role of this strand in formation of serpin-proteinase complexes and in serpin inhibitor activity through homology modeling of wild type inhibitor, mutant substrate, and latent serpins, and of putative serpin-proteinase complexes. These models explain the high stability of the complexes and provide an understanding of substrate behavior in serpins with point mutations in s4A and of latency in plasminogen activator inhibitor I.
- Published
- 1995
- Full Text
- View/download PDF
37. Accumulation of isogloboside and ganglio-N-tetraosyl ceramide having blood group B determinant in the hepatomas of female LEC rats.
- Author
-
Ariga T, Kasai N, Miyoshi I, Yamawaki M, Scarsdale JN, Yu RK, Kasama T, and Taki T
- Subjects
- Animals, Carbohydrate Sequence, Epitopes analysis, Female, G(M1) Ganglioside immunology, Globosides immunology, Glycolipids isolation & purification, Male, Molecular Sequence Data, Rats, ABO Blood-Group System, Biomarkers, Tumor analysis, Carcinoma, Hepatocellular metabolism, G(M1) Ganglioside metabolism, Globosides metabolism, Glycolipids metabolism, Liver Neoplasms metabolism
- Abstract
We have studied the neutral glycolipid composition of spontaneous hepatomas in LEC female rats. Neutral lipid fractions were isolated and purified by column chromatographies on DEAE-Toyopearl 650(M) and Iatrobeads. The neutral glycolipid fraction contained 3.2 to 4.4 micrograms lipid-bound glucose (Glc) per mg protein, and consisted of isogloboside (iso-Gb4, 50.8% of total neutral glycolipids) and IV3Gal, IV2Fuc, GgOse4Cer (asialo-BGM1, 13.5%) as the major neutral glycolipids and Gb3 and iso-Gb3 (9.2%), GlcCer (7.2%), LacCer (6.1%) as the other species. The structure of iso-Gb4 was elucidated by gas-liquid chromatography (GLC), permethylation study, liquid secondary ion (LSI) mass spectrometry, and nuclear Overhauser enhancement spectroscopy (NOESY) and that for asialo-BGM1 by GLC, LSI mass spectrometry, and high-performance thin-layer chromatography (HPTLC)-overlay method using anti-asialo-BGM1 antibody. Isogloboside and asialo-BGM1 which are found in negligible amounts in normal liver tissues may represent excellent markers for studying tumor metastasis and cellular adhesion.
- Published
- 1995
- Full Text
- View/download PDF
38. Conformational analysis of synthetic peptides encompassing the factor XI and prekallikrein overlapping binding domains of high molecular weight kininogen.
- Author
-
You JL, Page JD, Scarsdale JN, Colman RW, and Harris RB
- Subjects
- Amino Acid Sequence, Binding Sites physiology, Calorimetry, Differential Scanning, Ligands, Molecular Sequence Data, Molecular Weight, Protein Conformation, Protein Structure, Tertiary, Sequence Homology, Amino Acid, Spectrometry, Fluorescence, Titrimetry, Factor XI chemistry, Kininogens chemistry, Peptides chemistry, Prekallikrein chemistry
- Abstract
High molecular weight kininogen, a plasma glycoprotein, circulates as a noncovalent complex with either prekallikrein or factor XI, two other plasma glycoproteins. The binding domain for factor XI within kininogen, Pro556-Met613 (58 residues), wholly contains the binding domain for prekallikrein, Ser565-Lys595 (31 residues), but Trp569-Lys595 (27 residues) retains some ability to bind prekallikrein. Complex formation between these proteins is mediated by recognition between complementary domains. The 58-residue factor XI peptide domain has now been prepared following a strategy of condensation of long-chain peptide fragments prepared using orthogonal chemistry protocols. The 58-, 31-, and 27-residue peptides assume very different structures in aqueous solution as revealed by differential scanning calorimetry, intrinsic fluorescence emission, and circular dichroism spectroscopies. Thus, the 31-residue peptide shows a broad endothermic transition in differential scanning calorimetry (DSC), but the 58-mer undergoes a well-defined, two-state transition (Tm 43 degrees C; transition enthalpy approximately 30 kcal/mol). The 58- and 27-residue peptides continuously lose structure with increasing temperature, but the 31-mer retains significant structure even at temperatures approaching 90 degrees C. Lys595 plays a critical role in maintaining structure through electrostatic contacts, probably with Asp572 in the N-terminal segment of the 31-residue sequence. Isothermal ligand titration calorimetry was used to directly assess the ability of the 31-, 27-, and 58-residue peptides to bind prekallikrein. The 31-residue peptide binds prekallikrein with 25-fold higher affinity (Kd = 1.0 x 10(-6) M) than the 58-residue peptide and with 5.4-fold higher affinity than the 27-residue peptide. Hence, the essential features of the 31-residue peptide domain required for binding prekallikrein are absent in the 58-residue peptide, which is optimized for binding factor XI. The results suggest that a conformational change may occur within kininogen that causes expression of one domain structure in preference to the other.
- Published
- 1993
- Full Text
- View/download PDF
39. O-acetylated gangliosides in bovine buttermilk. Characterization of 7-O-acetyl, 9-O-acetyl, and 7,9-di-O-acetyl GD3.
- Author
-
Ren S, Scarsdale JN, Ariga T, Zhang Y, Klein RA, Hartmann R, Kushi Y, Egge H, and Yu RK
- Subjects
- Acetylation, Animals, Antibodies, Monoclonal immunology, Carbohydrate Sequence, Chromatography, Thin Layer, Gangliosides chemistry, Gangliosides immunology, Mass Spectrometry, Molecular Sequence Data, Molecular Structure, Butter analysis, Gangliosides analysis, Milk analysis
- Abstract
Three O-acetylated gangliosides, G1, G2, and G3, were purified from bovine buttermilk by using chloroform/methanol extraction, Folch partitioning, chromatography on DEAE-Sephadex A-25, and Iatrobeads columns. The final yields of gangliosides G1, G2, and G3 were 2 mg, 37 mg, and 40 mg per 1.7 kg of the buttermilk powder, respectively. On the basis of immunostaining on high performance thin layer chromatography with specific monoclonal antibodies, mild alkaline treatment, gas-liquid chromatographic analysis, fast atom bombardment mass spectrometry, and proton nuclear magnetic resonance studies, G1 and G2 are characterized as O-acetylated GD3 and G3 as O-acetylated GT3, and the structures of these gangliosides are as follows: [formula: see text] The major fatty acids of these gangliosides were C18:0, C22:0, C23:0, and C24:0, and the long chain base was C18-sphingosine.
- Published
- 1992
40. Calorimetric and spectroscopic examination of the solution phase structures of prekallikrein binding domain peptides of high molecular weight kininogen.
- Author
-
You JL, Scarsdale JN, and Harris RB
- Subjects
- Amino Acid Sequence, Binding Sites, Calorimetry, Differential Scanning, Circular Dichroism, Drug Stability, Hot Temperature, Kininogens chemistry, Magnetic Resonance Spectroscopy, Molecular Sequence Data, Solutions, Spectrometry, Fluorescence, Thermodynamics, Kininogens metabolism, Peptide Fragments metabolism, Prekallikrein metabolism
- Abstract
Unique sequence-binding sites are exposed on the surface of high molecular weight kininogen which complex prekallikrein or factor XI with high affinity and specificity. A sequence comprising 31 residues of the mature kininogen molecule (Asp565-Lys595) retains full binding activity for prekallikrein (KD = 20 nM) and assumes a complex folded structure in solution which is stabilized by long-range interactions between N- and C-terminal residues. The sequence Trp569-Lys595 (27 residues) shows only 28% of this binding affinity and lacks the key structural features required for protein recognition (Scarsdale, J. N., and Harris, R. B., J. Prot. Chem. 9, 647-659, 1990). We were thus able to predict that N- or C-terminal truncations of the binding-site sequence would disrupt the conformational integrity required for binding. Two new peptides of 20- and 22- residues have now been synthesized and their solution phase structures examined. These peptides are N- and C-terminal truncations, respectively, of the 27-residue sequence and correspond to the sequences Asp576-Lys595 and Trp569-Asp590 of high molecular weight kininogen. The results of fluorescence emission and circular dichroism (CD) spectroscopies in the range 25-90 degrees C and from differential scanning calorimetry (DSC) all substantiate the idea that the C-terminal truncation peptide binds prekallikrein 35-fold poorer than the 31-residue peptide because it is relatively unordered and possesses a less stable structure. Surprisingly, the N-terminal truncation peptide (20-mer) shows structural stability even at elevated temperatures and, like the 31-residue peptide, undergoes cold-induced denaturation observable in the DSC. 2D-NMR analysis of the 20-residue peptide revealed two distinct structures; one conformer possesses a more compact, folded structure than the other. However, the predicted structures assumed by either conformer are very different from those of either the 31- or 27-residue peptides. Hence, the binding affinity of the 20-residue peptide is 60-fold poorer than that for the 31-residue peptide because it assumes a nonproductive binding conformation(s).
- Published
- 1991
- Full Text
- View/download PDF
41. NMR and computational studies of interactions between remote residues in gangliosides.
- Author
-
Scarsdale JN, Prestegard JH, and Yu RK
- Subjects
- Alcohol Oxidoreductases genetics, Aldehyde Oxidoreductases genetics, Benzaldehyde Dehydrogenase (NADP+), Carbohydrate Conformation, Carbohydrate Sequence, G(M1) Ganglioside chemistry, Hydrogen Bonding, Magnetic Resonance Spectroscopy, Models, Molecular, Molecular Sequence Data, N-Acetylneuraminic Acid, Sialic Acids chemistry, G(M1) Ganglioside analogs & derivatives, Gangliosides chemistry
- Abstract
Conformational preferences of the gangliosides GM1, GM1b, and GD1a have been investigated by using a systematic combination of NMR distance constraints and molecular mechanics calculations. These gangliosides share a common four-sugar core but differ in the number or placement of sialic acid residues attached to the core. Placement of the sialic acid residues is shown to influence the preferred core conformation. The origin of these effects is postulated to be intramolecular interactions of the sialic acid residues with other remote residues. In the case of GM1, hydrogen bonds between the internal sialic acid and an N-acetyl group on GalNAc are suggested. In the case of GD1a, a hydrogen-bonding network between the terminal and internal sialic acids is suggested to play a role.
- Published
- 1990
- Full Text
- View/download PDF
42. Solution phase conformation studies of the prekallikrein binding domain of high molecular weight kininogen.
- Author
-
Scarsdale JN and Harris RB
- Subjects
- Amino Acid Sequence, Binding Sites physiology, Circular Dichroism, Fluorescence Polarization, Kininogens metabolism, Magnetic Resonance Spectroscopy, Molecular Sequence Data, Molecular Weight, Peptide Fragments chemical synthesis, Protein Conformation, Solutions, Spectrometry, Fluorescence, Kininogens chemistry, Prekallikrein metabolism
- Abstract
High molecular weight kininogen is a cofactor of the surface-dependent phase of the blood-clotting cascade. Unique sequence-binding sites are exposed on the surface of this glycoprotein which complex prekallikrein or factor XI with high affinity and specificity (Tait and Fujikawa, 1987). A sequence comprising 31-residues (residues 565-595 of the mature kininogen molecule) retains full binding activity for prekallikrein but the sequence 569-595 (27 residues) shows only 25% of this binding affinity (Vogel et al., 1990). Thus, the key structural features required for protein recognition reside in the 31-residue sequence but these features are likely compromised (or absent) in the 27-residue sequence. To determine the conformation of the prekallikrein-binding domain, peptides comprising the 31- and 27-residue sequences were prepared by solid-phase methods and their structures determined by circular dichroism, fluorescence polarization, and 2D-NMR techniques. Fluorescence emission spectra, polarization, and anisotropy measurements of the single Trp residue present in both peptides show that the 31-residue peptide contains an ordered microenvironment at its amino terminus, which is not present in the 27-residue peptide. This structural ordering is characterized by movement of the Trp residue into a more polar environment. Further, the 31-residue peptide possesses a higher limit anisotropy, longer rotational relaxation time, and shows a higher polarization value even at elevated temperatures. Circular dichroic spectra of both peptides in the far UV region are essentially identical and indicate that both peptides contain predominantly beta-turn elements, but also contain some alpha-helix, beta-sheet, and random coil character. The structural elements of both peptides are unchanged in urea solution, but the negative ellipticity absorption band in the near UV region assignable to Trp is eliminated in acid solution upon protonation of the neighboring-Asp-Asp-Asp- triplet. In the two peptides, the spin system of each amino acid has been assigned through 2D-1H scalar coupling correlated experiments; pure absorption NOESY experiments were used to determine through-space connectivities. The results are entirely consistent with the previous experiments in that both peptides contain predominantly beta-turn elements and the amino terminus of the 31-residue peptide is highly ordered in comparison with the 27-mer; in fact, this region is likely to be helical in nature.(ABSTRACT TRUNCATED AT 400 WORDS)
- Published
- 1990
- Full Text
- View/download PDF
43. Accumulation of a globo-series glycolipid having Gal alpha 1-3Gal in PC12h pheochromocytoma cells.
- Author
-
Ariga T, Yu RK, Scarsdale JN, Suzuki M, Kuroda Y, Kitagawa H, and Miyatake T
- Subjects
- Animals, Cell Line, Magnetic Resonance Spectroscopy, Mass Spectrometry, Rats, Adrenal Gland Neoplasms metabolism, Globosides metabolism, Glycolipids metabolism, Glycosphingolipids metabolism, Pheochromocytoma metabolism
- Abstract
In a previous paper, we reported the presence of globoside as a major neutral glycolipid in PC12 pheochromocytoma cells [Ariga, T., Macala, L. J., Saito, M., Margolis, R. K., Greene, L. A., Margolis, R. U., & Yu, R. K. (1988) Biochemistry 27, 52-58]. Recently, we found that subcloned PC12h cells accumulated another unusual neutral glycolipid. In order to characterize this glycolipid, PC12h cells were subcutaneously transplanted into rats. The induced tumor tissue accumulated two major neutral glycolipids, which were purified by Iatrobeads column and preparative thin-layer chromatographies. One of the glycolipids was found to be globoside, and the other had a globotriaosyl structure with an additional terminal Gal alpha 1-3 residue. Its structure was determined by fast atom bombardment mass spectrometry, two-dimensional proton nuclear magnetic resonance spectrometry (2D NMR), permethylation study, sequential degradation with exoglycosidase, and mild acid hydrolysis to be Gal(alpha 1-3)Gal(alpha 1-4)Gal(beta 1-4)Glc(beta 1-1')Cer.
- Published
- 1988
- Full Text
- View/download PDF
44. Analysis of complex carbohydrate primary and secondary structure via two-dimensional proton nuclear magnetic resonance spectroscopy.
- Author
-
Koerner TA, Yu RK, Scarsdale JN, Demou PC, and Prestegard JH
- Subjects
- Magnetic Resonance Spectroscopy, Molecular Sequence Data, Oligosaccharides, Carbohydrate Conformation, Carbohydrate Sequence
- Published
- 1988
- Full Text
- View/download PDF
45. High resolution proton NMR studies of gangliosides. Structure of two types of GD3 lactones and their reactivity with monoclonal antibody R24.
- Author
-
Ando S, Yu RK, Scarsdale JN, Kusunoki S, and Prestegard JH
- Subjects
- Animals, Antibodies, Monoclonal, Carbohydrate Conformation, Cattle, Chemical Phenomena, Chemistry, Chromatography, Ion Exchange, Chromatography, Thin Layer, Esters, Immunoassay, N-Acetylneuraminic Acid, Sialic Acids, Gangliosides chemical synthesis, Gangliosides isolation & purification, Magnetic Resonance Spectroscopy
- Abstract
Ganglioside GD3 was converted at room temperature to two stable lactones, denoted as GD3 lactones I and II. The reaction sequence was presumed to be GD3----GD3 lactone I----GD3 lactone II based on the time course of their production. Lactone I behaved as a monosialoganglioside and lactone II as a neutral species. The two lactones were isolated by DEAE-Sephadex column chromatography. The positions of the inner ester linkages were investigated by two-dimensional J-correlated proton NMR spectroscopy. An ester linkage was most likely formed between the carboxyl group of the external sialic acid residue and C9-OH of the internal sialic acid residue in lactone I. In addition to this ester linkage, a second ester linkage between the carboxyl group of the internal sialic acid and C2-OH of the galactose residue was likely formed in lactone II. The structural changes induced by lactonization were further examined by their reactivity with the monoclonal antibody R24 (Puckel, C. S., Lloyd, K. O., Travassos, L. R., Dippold, W. G., Oettgen, H. F., and Old, L. J. (1982) J. Exp. Med. 155, 1133-1147), which reacted with GD3. R24 was found to bind weakly to GD3 lactone I, but not to GD3 lactone II. The results suggest that the monoclonal antibody requires both sialic acid residues for high affinity binding, and the complete lactonization results in a loss of negative charges and/or a change in the overall conformation of the oligosaccharide moiety which may account for the loss of binding.
- Published
- 1989
46. Recent advances in structural analysis of gangliosides: primary and secondary structures.
- Author
-
Yu RK, Koerner TA Jr, Demou PC, Scarsdale JN, and Prestegard JH
- Subjects
- Animals, Carbohydrate Conformation, Carbohydrate Sequence, G(M2) Ganglioside, Globosides, Glycosides analysis, Humans, Magnetic Resonance Spectroscopy methods, Models, Molecular, Oligosaccharides analysis, Gangliosides
- Abstract
High-field (500 MHz) proton NMR has been used to elucidate the primary and secondary structures of glycosphingolipids (GSLs). Using 2-D J-correlated spectroscopy (2-D SECSY) which establishes scalar couplings of protons, the monosaccharide composition, anomeric configuration and aglycon structures of a GSL can be established. 2-D nuclear Overhauser effect spectroscopy (2-D NOE) then establishes through-space intra- and inter-residue couplings of cross-relaxing protons. We have found that each anomeric proton is involved in NOE couplings with inter- and intra-residue protons. The inter-residue coupling, resulting from interaction of protons across the glycosidic linkage, establishes the n-1 sugar residue and specific glycosidation site to which the n-residue is linked. When such information is known for each residue and is combined, the sequence of the core oligosaccharide is obtained. The sialylation-induced glycosidation shift is then used to establish the site of sialic acid residue attachment in a ganglioside molecule. We have also observed that the anomeric proton inter-residue NOE couplings can be used to suggest the preferred conformation of an oligosaccharide. We have found that the oligosaccharide residue of globoside exists in a unique and rather rigid conformation which could be stabilized by hydrogen bonds and van der Waals interactions. Since GSLs are known to have a receptor role and are implicated in cell-cell recognition, enzyme-substrate interaction and antigen-antibody interaction, the determination of their conformation should be useful in understanding their biological functions.
- Published
- 1984
- Full Text
- View/download PDF
47. Ab initio study on the molecular structure of the naphthalene metabolite, trans-1,2-dihydroxy-1,2-dihydronaphthalene.
- Author
-
Beland FA, Melchior WB Jr, Klimkowski VJ, Scarsdale JN, Van Alsenoy C, and Schäfer L
- Subjects
- Models, Molecular, Naphthols metabolism
- Abstract
The molecular geometries of three conformations of trans-1,2-dihydroxy-1,2-dihydronaphthalene have been refined by an ab initio gradient procedure at the 4-21G level to determine the effect of dihydrodiol conformation on arene structure. The preferred conformation is an equatorial form similar to the most stable conformation of ethylene glycol. All the structures investigated have similar arene geometries. The effect of the various conformations on metabolism of dihydrodiols to dihydrodiol epoxides is considered.
- Published
- 1984
- Full Text
- View/download PDF
48. Elucidation of glycolipid structure by proton nuclear magnetic resonance spectroscopy.
- Author
-
Yu RK, Koerner TA, Scarsdale JN, and Prestegard JH
- Subjects
- Carbohydrate Conformation, Carbohydrate Sequence, Gangliosides, Magnetic Resonance Spectroscopy, Oligosaccharides, Glycolipids
- Abstract
The primary structure of the oligosaccharide moiety of a glycosphingolipid can be elucidated by employing high-field proton nuclear magnetic resonance (NMR) spectroscopy. Information with respect to the composition and configuration of its sugar residues, and the sequence and linkage sites of the oligosaccharide chain can be obtained by employing a variety of one- and two-dimensional techniques. The latter include both scalar and dipolar correlated two-dimensional NMR spectroscopy. These techniques are also useful in establishing the solution conformation (secondary structure) of the oligosaccharide moiety. Examples in utilizing these techniques in elucidating the primary and secondary structures of glycolipids are presented.
- Published
- 1986
- Full Text
- View/download PDF
49. 1H-2D-nuclear magnetic resonance applied to the primary structure determination of a novel octasaccharide glycolipid isolated from the spermatozoa of bivalves.
- Author
-
Scarsdale JN, Prestegard JH, Ando S, Hori T, and Yu RK
- Subjects
- Animals, Carbohydrate Conformation, Carbohydrate Sequence, Magnetic Resonance Spectroscopy methods, Male, Mollusca, Glycolipids isolation & purification, Oligosaccharides isolation & purification, Spermatozoa analysis
- Abstract
High resolution, two-dimensional 1H-n.m.r. spectroscopy has been used to confirm a proposed primary structure of a glycolipid having an octasaccharide head-group. Pure absorption and relay experiments were found to be particularly useful in establishing connectivities in poorly resolved regions of the spectrum. The spectral assignments, which indicate novel linkages including an internal fucopyranosyl residue as well as terminal xylosyl and 4-O-methylglucopyranosyluronic acid groups, add to a growing data base for structural characterization through n.m.r. spectroscopy.
- Published
- 1986
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.