27 results on '"Yanchunas J"'
Search Results
2. Inhibition of Bovine Brain Nitric-Oxide Synthase by α-Amino and α-Carboxyl Derivatives of NG-Allyl-L-Arginine
- Author
-
Robertson, J.G., primary, Bernatowicz, M.S., additional, Dhalla, A.M., additional, Muhoberac, B.B., additional, Yanchunas, J., additional, Matsueda, G.R., additional, and Villafranca, J.J., additional
- Published
- 1995
- Full Text
- View/download PDF
3. Dimerization of Native and C-Terminally Proteolyzed p56lck Tyrosine Kinase
- Author
-
Robertson, J.G., primary, Yanchunas, J., additional, and Villafranca, J.J., additional
- Published
- 1995
- Full Text
- View/download PDF
4. Dimerization of Native and C-Terminally Proteolyzed p56lck Tyrosine Kinase
- Author
-
Robertson, J.G., Yanchunas, J., and Villafranca, J.J.
- Abstract
Recombinant p56
lck tyrosine kinase was purified to near homogeneity from a baculovirus/insect cell expression system. Treatment with thrombin proteolytically removed the C-terminal 54 amino acids from p56lck . Processed enzyme migrated on sodium dodecyl sulfate (SDS) gels with a Mr ≍ 6,000 lower than intact enzyme. Analytical ultracentrifugation of intact and processed p56lck gave Mr's of 62,600 and 56,200, respectively, confirming that the thrombin treated enzyme existed in solution as a processed polypeptide and that there was no anomalous migration in SDS gels due to thrombin treatment. Simultaneous multispeed analysis of sedimentation equilibrium data demonstrated that both intact and processed enzyme can dimerize with a weak binding constant in the range of 200-300 µM. Purified intact p56lck incorporated 2 mol of [32 P]Pi per mole of enzyme. Purified processed p56lck incorporated only 1 mol of [32 P]Pi per mole of enzyme. The loss of 1 mol of [32 P]Pi per mole of enzyme after thrombin deletion of the C-terminus demonstrates that p56lck undergoes autophosphorylation at the C-terminus. The data are consistent with autophosphorylation at tyrosine 505, which has previously been thought to be a regulatory phosphorylation site, but which now must also be considered as an autophosphorylation site.Copyright 1995, 1999 Academic Press, Inc.- Published
- 1995
- Full Text
- View/download PDF
5. Inhibition of Bovine Brain Nitric-Oxide Synthase by α-Amino and α-Carboxyl Derivatives of NG-Allyl-L-Arginine
- Author
-
Robertson, J.G., Bernatowicz, M.S., Dhalla, A.M., Muhoberac, B.B., Yanchunas, J., Matsueda, G.R., and Villafranca, J.J.
- Abstract
Three derivatives of the mechanism-based inhibitor N
G -allyl-L-arginine, designed to eliminate the effect of charge on the α-functional groups, were synthesized and tested as inhibitors of purified bovine brain nitric oxide synthase. The inhibitory properties of NG -allyl-L-arginine, NG -allyl-L-arginine methyl ester, Nα -acetyl-NG -allyl-L-arginine, and Nα -acetyl-NG -allyl-L-arginine methyl ester were determined in steady-state kinetic assays. The Kis of the four compounds were 7 ± 1, 11 ± 1, 147 ± 13, and 480 ± 45 µM, respectively. These results demonstrate that conversion of the α-carboxylgroup of NG -allyl-L-arginine to a methyl ester had only a small effect on its inhibitory properties, whereas acetylation of the α-amino group increased the Ki by more than an order of magnitude. Modification of both the α-amino and α-carboxyl groups increased the Ki more dramatically from 7 to 480 µM. Derivatization of the α-amino and α-carboxyl groups of NG -allyl-L-arginine would not be expected to alter the chemistry of inactivation by the NG -allyl guanidine moiety, and therefore the increased Kis of the derivatives are probably due solely to changes in binding specificity. These data suggest that the arginine binding pocket of brain nitric oxide synthase prefers the unmodified α-amino group of arginine for binding, but that it can accommodate a modified α-carboxylate. Thus, conservative modification at the alpha-carboxyl may represent a starting point for the design and synthesis of other inhibitors targeted at nitric oxide synthase.Copyright 1995, 1999 Academic Press, Inc.- Published
- 1995
- Full Text
- View/download PDF
6. Crystallization and preliminary crystallographic analysis of E. coli uridine 5'-diphospho- N-acetylenolpyruvylglucosamine reductase in two new crystal forms.
- Author
-
Ohringer, S. L., Chang, C. Y., Sheriff, S., Klei, H. E., Sack, J. S., Jacobson, B. L., Yanchunas, J., Lavoie, T., Dhalla, A. M., Robertson, J. G., and Einspahr, H. M.
- Published
- 1996
- Full Text
- View/download PDF
7. Long-term removal and retention of iron and manganese from acidic mine drainage by wetlands. Volume 1. Methods, results, and appendices. Final report, July 1987-January 1990
- Author
-
Yanchunas, J
- Published
- 1990
8. Long-term removal and retention of iron and manganese from acidic mine drainage by wetlands. Volume 2. Tarutis thesis. Final report, July 1987-January 1990
- Author
-
Yanchunas, J
- Published
- 1990
9. Biologic-like In Vivo Efficacy with Small Molecule Inhibitors of TNFα Identified Using Scaffold Hopping and Structure-Based Drug Design Approaches.
- Author
-
Xiao HY, Li N, Duan JJ, Jiang B, Lu Z, Ngu K, Tino J, Kopcho LM, Lu H, Chen J, Tebben AJ, Sheriff S, Chang CY, Yanchunas J Jr, Calambur D, Gao M, Shuster DJ, Susulic V, Xie JH, Guarino VR, Wu DR, Gregor KR, Goldstine CB, Hynes J Jr, Macor JE, Salter-Cid L, Burke JR, Shaw PJ, and Dhar TGM
- Subjects
- Animals, Arthritis, Experimental drug therapy, Arthritis, Rheumatoid drug therapy, Drug Design, Female, Humans, Mice, Inbred C57BL, Microsomes, Liver metabolism, Molecular Structure, Naphthyridines chemical synthesis, Naphthyridines pharmacokinetics, Naphthyridines therapeutic use, Proof of Concept Study, Quinolines chemical synthesis, Quinolines pharmacokinetics, Quinolines therapeutic use, Structure-Activity Relationship, Tumor Necrosis Factor-alpha metabolism, Naphthyridines pharmacology, Quinolines pharmacology, Tumor Necrosis Factor-alpha antagonists & inhibitors
- Abstract
Scaffold hopping and structure-based drug design were employed to identify substituted 4-aminoquinolines and 4-aminonaphthyridines as potent, small molecule inhibitors of tumor necrosis factor alpha (TNFα). Structure-activity relationships in both the quinoline and naphthyridine series leading to the identification of compound 42 with excellent potency and pharmacokinetic profile are discussed. X-ray co-crystal structure analysis and ultracentrifugation experiments clearly demonstrate that these inhibitors distort the TNFα trimer upon binding, leading to aberrant signaling when the trimer binds to TNF receptor 1 (TNFR1). Pharmacokinetic-pharmacodynamic activity of compound 42 in a TNF-induced IL-6 mouse model and in vivo activity in a collagen antibody-induced arthritis model, where it showed biologic-like in vivo efficacy, will be discussed.
- Published
- 2020
- Full Text
- View/download PDF
10. Small molecule receptor protein tyrosine phosphatase γ (RPTPγ) ligands that inhibit phosphatase activity via perturbation of the tryptophan-proline-aspartate (WPD) loop.
- Author
-
Sheriff S, Beno BR, Zhai W, Kostich WA, McDonnell PA, Kish K, Goldfarb V, Gao M, Kiefer SE, Yanchunas J, Huang Y, Shi S, Zhu S, Dzierba C, Bronson J, Macor JE, Appiah KK, Westphal RS, O'Connell J, and Gerritz SW
- Subjects
- Amino Acid Sequence, Catalytic Domain, Crystallography, X-Ray, Humans, Hydrophobic and Hydrophilic Interactions, Ligands, Molecular Sequence Data, Protein Binding, Protein Conformation, Receptor-Like Protein Tyrosine Phosphatases, Class 5 chemistry, Structure-Activity Relationship, Thiophenes chemical synthesis, Models, Molecular, Receptor-Like Protein Tyrosine Phosphatases, Class 5 antagonists & inhibitors, Thiophenes chemistry
- Abstract
Protein tyrosine phosphatases (PTPs) catalyze the dephosphorylation of tyrosine residues, a process that involves a conserved tryptophan-proline-aspartate (WPD) loop in catalysis. In previously determined structures of PTPs, the WPD-loop has been observed in either an "open" conformation or a "closed" conformation. In the current work, X-ray structures of the catalytic domain of receptor-like protein tyrosine phosphatase γ (RPTPγ) revealed a ligand-induced "superopen" conformation not previously reported for PTPs. In the superopen conformation, the ligand acts as an apparent competitive inhibitor and binds in a small hydrophobic pocket adjacent to, but distinct from, the active site. In the open and closed WPD-loop conformations of RPTPγ, the side chain of Trp1026 partially occupies this pocket. In the superopen conformation, Trp1026 is displaced allowing a 3,4-dichlorobenzyl substituent to occupy this site. The bound ligand prevents closure of the WPD-loop over the active site and disrupts the catalytic cycle of the enzyme.
- Published
- 2011
- Full Text
- View/download PDF
11. Aroylguanidine-based factor Xa inhibitors: the discovery of BMS-344577.
- Author
-
Shi Y, Li C, O'Connor SP, Zhang J, Shi M, Bisaha SN, Wang Y, Sitkoff D, Pudzianowski AT, Huang C, Klei HE, Kish K, Yanchunas J Jr, Liu EC, Hartl KS, Seiler SM, Steinbacher TE, Schumacher WA, Atwal KS, and Stein PD
- Subjects
- Anticoagulants pharmacology, Cytochrome P-450 CYP3A, Cytochrome P-450 CYP3A Inhibitors, Drug Discovery, Guanidines pharmacology, Humans, Inhibitory Concentration 50, Serine Proteinase Inhibitors pharmacology, Structure-Activity Relationship, Anticoagulants chemistry, Factor Xa Inhibitors, Guanidines chemistry, Serine Proteinase Inhibitors chemistry
- Abstract
We report the design and synthesis of a novel class of N,N'-disubstituted aroylguanidine-based lactam derivatives as potent and orally active FXa inhibitors. The structure-activity relationships (SAR) investigation led to the discovery of the nicotinoyl guanidine 22 as a potent FXa inhibitor (FXa IC(50)=4 nM, EC(2xPT)=7 microM). However, the potent CYP3A4 inhibition activity (IC(50)=0.3 microM) of 22 precluded its further development. Detailed analysis of the X-ray crystal structure of compound 22 bound to FXa indicated that the substituent at the 6-position of the nicotinoyl group of 22 would be solvent-exposed, suggesting that efforts to attenuate the unwanted CYP activity could focus at this position without affecting FXa potency significantly. Further SAR studies on the 6-substituted nicotinoyl guanidines resulted in the discovery of 6-(dimethylcarbamoyl) nicotinoyl guanidine 36 (BMS-344577, IC(50)=9 nM, EC(2xPT)=2.5 microM), which was found to be a selective, orally efficacious FXa inhibitor with an excellent in vitro liability profile, favorable pharmacokinetics and pharmacodynamics in animal models.
- Published
- 2009
- Full Text
- View/download PDF
12. Assessing compound binding to the Eg5 motor domain using a thermal shift assay.
- Author
-
McDonnell PA, Yanchunas J, Newitt JA, Tao L, Kiefer SE, Ortega M, Kut S, Burford N, Goldfarb V, Duke GJ, Shen H, Metzler W, Doyle M, Chen Z, Tarby C, Borzilleri R, Vaccaro W, Gottardis M, Lu S, Crews D, Kim K, Lombardo L, and Roussell DL
- Subjects
- Adenosine Diphosphate metabolism, Biophysical Phenomena, Calorimetry, Cell Line, Tumor, Circular Dichroism, Humans, Kinesins metabolism, Magnetic Resonance Spectroscopy, Models, Molecular, Protein Binding, Protein Denaturation, Protein Folding, Protein Structure, Tertiary, Temperature, Biochemistry methods, Kinesins chemistry, Pyrroles analysis, Pyrroles chemistry, Triazines analysis, Triazines chemistry
- Abstract
Eg5 is a kinesin whose inhibition leads to cycle arrest during mitosis, making it a potential therapeutic target in cancers. Circular dichroism and isothermal titration calorimetry of our pyrrolotriazine-4-one series of inhibitors with Eg5 motor domain revealed enhanced binding in the presence of adenosine 5'-diphosphate (ADP). Using this information, we studied the interaction of this series with ADP-Eg5 complexes using a thermal shift assay. We measured up to a 7 degrees C increase in the thermal melting (T(m)) of Eg5 for an inhibitor that produced IC(50) values of 60 and 130 nM in microtubule-dependent adenosine triphosphatase (ATPase) and cell-based cytotoxicity assays, respectively. In general, the inhibitor potency of the pyrrolotriazine-4-one series in in vitro biological assays correlated with the magnitude of the thermal stability enhancement of ADP-Eg5. The thermal shift assay also confirmed direct binding of Eg5 inhibitors identified in a high-throughput screen and demonstrated that the thermal shift assay is applicable to a range of chemotypes and can be useful in evaluating both potent (nM) and relatively weakly binding (microM) leads. Overall, the thermal shift assay was found to be an excellent biophysical method for evaluating direct binding of a large number of compounds to Eg5, and it complemented the catalytic assay screens by providing an alternative determination of inhibitor potency.
- Published
- 2009
- Full Text
- View/download PDF
13. Cyanoguanidine-based lactam derivatives as a novel class of orally bioavailable factor Xa inhibitors.
- Author
-
Shi Y, Zhang J, Shi M, O'Connor SP, Bisaha SN, Li C, Sitkoff D, Pudzianowski AT, Chong S, Klei HE, Kish K, Yanchunas J Jr, Liu EC, Hartl KS, Seiler SM, Steinbacher TE, Schumacher WA, Atwal KS, and Stein PD
- Subjects
- Administration, Oral, Animals, Antithrombin III chemistry, Benzofurans chemistry, Chemistry, Pharmaceutical methods, Crystallography, X-Ray methods, Dogs, Haplorhini, Humans, Inhibitory Concentration 50, Kinetics, Lactams pharmacology, Ligands, Models, Chemical, Rats, Structure-Activity Relationship, Thiourea chemistry, Antithrombin III pharmacology, Benzofurans pharmacology, Guanidines chemistry, Lactams chemistry
- Abstract
The N,N'-disubstituted cyanoguanidine is an excellent bioisostere of the thiourea and ketene aminal functional groups. We report the design and synthesis of a novel class of cyanoguanidine-based lactam derivatives as potent and orally active FXa inhibitors. The SAR studies led to the discovery of compound 4 (BMS-269223, K(i)=6.5nM, EC(2xPT)=32muM) as a selective, orally bioavailable FXa inhibitor with an excellent in vitro liability profile, favorable pharmacokinetics and pharmacodynamics in animal models. The X-ray crystal structure of 4 bound in FXa is presented and key ligand-protein interactions are discussed.
- Published
- 2009
- Full Text
- View/download PDF
14. Design, structure-activity relationships, X-ray crystal structure, and energetic contributions of a critical P1 pharmacophore: 3-chloroindole-7-yl-based factor Xa inhibitors.
- Author
-
Shi Y, Sitkoff D, Zhang J, Klei HE, Kish K, Liu EC, Hartl KS, Seiler SM, Chang M, Huang C, Youssef S, Steinbacher TE, Schumacher WA, Grazier N, Pudzianowski A, Apedo A, Discenza L, Yanchunas J, Stein PD, and Atwal KS
- Subjects
- Animals, Binding Sites drug effects, Computer Simulation, Crystallography, X-Ray, Dose-Response Relationship, Drug, Factor Xa drug effects, Humans, Hydrogen Bonding, Mice, Models, Chemical, Models, Molecular, Structure-Activity Relationship, Survival Analysis, Venoms pharmacology, Venous Thrombosis drug therapy, Venous Thrombosis enzymology, Drug Design, Enzyme Inhibitors chemical synthesis, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Factor Xa Inhibitors, Indoles chemical synthesis, Indoles chemistry, Indoles pharmacology, Quantum Theory
- Abstract
An indole-based P1 moiety was incorporated into a previously established factor Xa inhibitor series. The indole group was designed to hydrogen-bond with the carbonyl of Gly218, while its 3-methyl or 3-chloro substituent was intended to interact with Tyr228. These interactions were subsequently observed in the X-ray crystal structure of compound 18. SAR studies led to the identification of compound 20 as the most potent FXa inhibitor in this series (IC(50) = 2.4 nM, EC(2xPT) = 1.2 microM). An in-depth energetic analysis suggests that the increased binding energy of 3-chloroindole-versus 3-methylindole-containing compounds in this series is due primarily to (a) the more hydrophobic nature of chloro- versus methyl-containing compounds and (b) an increased interaction of 3-chloroindole versus 3-methylindole with Gly218 backbone. The stronger hydrophobicity of chloro- versus methyl-substituted aromatics may partly explain the general preference for chloro- versus methyl-substituted P1 groups in FXa, which extends beyond the current series.
- Published
- 2008
- Full Text
- View/download PDF
15. Involvement of DPP-IV catalytic residues in enzyme-saxagliptin complex formation.
- Author
-
Metzler WJ, Yanchunas J, Weigelt C, Kish K, Klei HE, Xie D, Zhang Y, Corbett M, Tamura JK, He B, Hamann LG, Kirby MS, and Marcinkeviciene J
- Subjects
- Adamantane chemistry, Adamantane metabolism, Binding Sites, Catalytic Domain, Crystallography, X-Ray, Dipeptides metabolism, Dipeptidyl Peptidase 4 metabolism, Dipeptidyl-Peptidase IV Inhibitors, Enzyme Inhibitors chemistry, Enzyme Inhibitors metabolism, Humans, Hydrogen Bonding, Mutant Proteins chemistry, Mutant Proteins metabolism, Nuclear Magnetic Resonance, Biomolecular, Protein Structure, Quaternary, Adamantane analogs & derivatives, Dipeptides chemistry, Dipeptidyl Peptidase 4 chemistry
- Abstract
The inhibition of DPP-IV by saxagliptin has been proposed to occur through formation of a covalent but reversible complex. To evaluate further the mechanism of inhibition, we determined the X-ray crystal structure of the DPP-IV:saxagliptin complex. This structure reveals covalent attachment between S630 and the inhibitor nitrile carbon (C-O distance <1.3 A). To investigate whether this serine addition is assisted by the catalytic His-Asp dyad, we generated two mutants of DPP-IV, S630A and H740Q, and assayed them for ability to bind inhibitor. DPP-IV H740Q bound saxagliptin with an approximately 1000-fold reduction in affinity relative to DPP-IV WT, while DPP-IV S630A showed no evidence for binding inhibitor. An analog of saxagliptin lacking the nitrile group showed unchanged binding properties to the both mutant proteins, highlighting the essential role S630 and H740 play in covalent bond formation between S630 and saxagliptin. Further supporting mechanism-based inhibition by saxagliptin, NMR spectra of enzyme-saxagliptin complexes revealed the presence of three downfield resonances with low fractionation factors characteristic of short and strong hydrogen bonds (SSHB). Comparison of the NMR spectra of various wild-type and mutant DPP-IV:ligand complexes enabled assignment of a resonance at approximately 14 ppm to H740. Two additional DPP-IV mutants, Y547F and Y547Q, generated to probe potential stabilization of the enzyme-inhibitor complex by this residue, did not show any differences in inhibitor binding either by ITC or NMR. Together with the previously published enzymatic data, the structural and binding data presented here strongly support a histidine-assisted covalent bond formation between S630 hydroxyl oxygen and the nitrile group of saxagliptin.
- Published
- 2008
- Full Text
- View/download PDF
16. Molecular basis for increased susceptibility of isolates with atazanavir resistance-conferring substitution I50L to other protease inhibitors.
- Author
-
Yanchunas J Jr, Langley DR, Tao L, Rose RE, Friborg J, Colonno RJ, and Doyle ML
- Subjects
- Amino Acid Substitution, Atazanavir Sulfate, Binding, Competitive drug effects, Calorimetry, Differential Scanning, Catalysis, Drug Resistance, Viral, HIV Protease chemistry, Hot Temperature, Models, Molecular, Models, Structural, Temperature, HIV Protease genetics, HIV Protease Inhibitors pharmacology, HIV-1 drug effects, HIV-1 genetics, Oligopeptides pharmacology, Pyridines pharmacology
- Abstract
Protease inhibitors (PIs) are highly effective drugs against the human immunodeficiency virus (HIV), yet long-term therapeutic use is limited by emergence of HIV type 1 (HIV-1) protease substitutions that confer cross-resistance to multiple protease inhibitor drugs. Atazanavir is a highly potent HIV protease inhibitor with a distinct resistance profile that includes effectiveness against most HIV-1 isolates resistant to one or two PIs. The signature resistance substitution for atazanavir is I50L, and it is frequently (53%) accompanied by a compensatory A71V substitution that helps restore viability and increases atazanavir resistance levels. We measured the binding affinities of wild-type (WT) and I50L/A71V HIV-1 proteases to atazanavir and other currently approved PIs (ritonavir, lopinavir, saquinavir, nelfinavir, indinavir, and amprenavir) by isothermal titration calorimetry. Remarkably, we find that all of the PIs have 2- to 10-fold increased affinities for I50L/A71V protease, except for atazanavir. The results are also manifested by thermal stability measures of affinity for WT and I50L/A71V proteases. Additional biophysical and enzyme kinetics experiments show I50L/A71V protease is a stable enzyme with catalytic activity that is slightly reduced (34%) relative to the WT. Computational modeling reveals that the unique resistance phenotype of I50L/A71V protease likely originates from bulky tert-butyl groups at P2 and P2' (specific to atazanavir) that sterically clash with methyl groups on residue L50. The results of this study provide a molecular understanding of the novel hypersusceptibility of atazanavir-resistant I50L/A71V-containing clinical isolates to other currently approved PIs.
- Published
- 2005
- Full Text
- View/download PDF
17. Structural and functional characterization of CFE88: evidence that a conserved and essential bacterial protein is a methyltransferase.
- Author
-
Constantine KL, Krystek SR, Healy MD, Doyle ML, Siemers NO, Thanassi J, Yan N, Xie D, Goldfarb V, Yanchunas J, Tao L, Dougherty BA, and Farmer BT 2nd
- Subjects
- Amino Acid Sequence, Molecular Sequence Data, Protein Structure, Tertiary, Bacterial Proteins chemistry, Methyltransferases chemistry, Streptococcus pneumoniae enzymology, Structural Homology, Protein
- Abstract
CFE88 is a conserved essential gene product from Streptococcus pneumoniae. This 227-residue protein has minimal sequence similarity to proteins of known 3D structure. Sequence alignment models and computational protein threading studies suggest that CFE88 is a methyltransferase. Characterization of the conformation and function of CFE88 has been performed by using several techniques. Backbone atom and limited side-chain atom NMR resonance assignments have been obtained. The data indicate that CFE88 has two domains: an N-terminal domain with 163 residues and a C-terminal domain with 64 residues. The C-terminal domain is primarily helical, while the N-terminal domain has a mixed helical/extended (Rossmann) fold. By aligning the experimentally observed elements of secondary structure, an initial unrefined model of CFE88 has been constructed based on the X-ray structure of ErmC' methyltransferase (Protein Data Bank entry 1QAN). NMR and biophysical studies demonstrate binding of S-adenosyl-L-homocysteine (SAH) to CFE88; these interactions have been localized by NMR to the predicted active site in the N-terminal domain. Mutants that target this predicted active site (H26W, E46R, and E46W) have been constructed and characterized. Overall, our results both indicate that CFE88 is a methyltransferase and further suggest that the methyltransferase activity is essential for bacterial survival.
- Published
- 2005
- Full Text
- View/download PDF
18. Decrypting the biochemical function of an essential gene from Streptococcus pneumoniae using ThermoFluor technology.
- Author
-
Carver TE, Bordeau B, Cummings MD, Petrella EC, Pucci MJ, Zawadzke LE, Dougherty BA, Tredup JA, Bryson JW, Yanchunas J Jr, Doyle ML, Witmer MR, Nelen MI, DesJarlais RL, Jaeger EP, Devine H, Asel ED, Springer BA, Bone R, Salemme FR, and Todd MJ
- Subjects
- Amino Acid Sequence, Benzopyrans metabolism, Dimerization, Furans metabolism, Ligands, Molecular Sequence Data, Pyridoxal Phosphate metabolism, Pyridoxamine metabolism, Streptococcus pneumoniae enzymology, Bacterial Proteins physiology, Genes, Essential physiology, Nucleoside Diphosphate Sugars metabolism, Streptococcus pneumoniae genetics, Transaminases physiology
- Abstract
The protein product of an essential gene of unknown function from Streptococcus pneumoniae was expressed and purified for screening in the ThermoFluor affinity screening assay. This assay can detect ligand binding to proteins of unknown function. The recombinant protein was found to be in a dimeric, native-like folded state and to unfold cooperatively. ThermoFluor was used to screen the protein against a library of 3000 compounds that were specifically selected to provide information about possible biological functions. The results of this screen identified pyridoxal phosphate and pyridoxamine phosphate as equilibrium binding ligands (K(d) approximately 50 pM, K(d) approximately 2.5 microM, respectively), consistent with an enzymatic cofactor function. Several nucleotides and nucleotide sugars were also identified as ligands of this protein. Sequence comparison with two enzymes of known structure but relatively low overall sequence homology established that several key residues directly involved in pyridoxal phosphate binding were strictly conserved. Screening a collection of generic drugs and natural products identified the antifungal compound canescin A as an irreversible covalent modifier of the enzyme. Our investigation of this protein indicates that its probable biological role is that of a nucleoside diphospho-keto-sugar aminotransferase, although the preferred keto-sugar substrate remains unknown. These experiments demonstrate the utility of a generic affinity-based ligand binding technology in decrypting possible biological functions of a protein, an approach that is both independent of and complementary to existing genomic and proteomic technologies.
- Published
- 2005
- Full Text
- View/download PDF
19. Correlation of high-throughput pregnane X receptor (PXR) transactivation and binding assays.
- Author
-
Zhu Z, Kim S, Chen T, Lin JH, Bell A, Bryson J, Dubaquie Y, Yan N, Yanchunas J, Xie D, Stoffel R, Sinz M, and Dickinson K
- Subjects
- Cells, Cultured, Culture Media, Ligands, Molecular Weight, Pharmaceutical Preparations metabolism, Pregnane X Receptor, Protein Binding, Regression Analysis, Reproducibility of Results, Radioligand Assay methods, Receptors, Cytoplasmic and Nuclear metabolism, Receptors, Steroid metabolism, Transcriptional Activation
- Abstract
Pregnane X receptor (PXR) transactivation and binding assays have been developed into high-throughput assays, which are robust and reproducible (Z' > 0.5). For most compounds, there was a good correlation between the results of the transactivation and binding assays. EC(50) values of compounds in the transactivation assay correlated reasonably well with their IC(50) values in the binding assay. However, there were discrepancies with some compounds showing high binding affinity in the binding assay translated into low transactivation. The most likely cause for these discrepancies was an agonist-dependent relationship between binding affinity and transactivation response. In general, compounds that bound to human PXR and transactivated PXR tended to be large hydrophobic molecules.
- Published
- 2004
- Full Text
- View/download PDF
20. Comparative studies of active site-ligand interactions among various recombinant constructs of human beta-amyloid precursor protein cleaving enzyme.
- Author
-
Kopcho LM, Ma J, Marcinkeviciene J, Lai Z, Witmer MR, Cheng J, Yanchunas J, Tredup J, Corbett M, Calambur D, Wittekind M, Paruchuri M, Kothari D, Lee G, Ganguly S, Ramamurthy V, Morin PE, Camac DM, King RW, Lasut AL, Ross OH, Hillman MC, Fish B, Shen K, Dowling RL, Kim YB, Graciani NR, Collins D, Combs AP, George H, Thompson LA, and Copeland RA
- Subjects
- Alzheimer Disease metabolism, Amyloid Precursor Protein Secretases, Animals, Aspartic Acid Endopeptidases metabolism, Binding Sites, CHO Cells, Catalysis, Catalytic Domain, Cell Line, Cell Membrane metabolism, Chromatography, High Pressure Liquid, Cricetinae, Dose-Response Relationship, Drug, Drosophila, Endopeptidases, Escherichia coli metabolism, Glycosylation, Humans, Inhibitory Concentration 50, Kinetics, Ligands, Light, Lipid Bilayers metabolism, Peptides chemistry, Protein Binding, Protein Structure, Tertiary, Recombinant Proteins metabolism, Scattering, Radiation, Time Factors, Aspartic Acid Endopeptidases chemistry, Recombinant Proteins chemistry
- Abstract
Amyloid precursor protein (APP) cleaving enzyme (BACE) is the enzyme responsible for beta-site cleavage of APP, leading to the formation of the amyloid-beta peptide that is thought to be pathogenic in Alzheimer's disease (AD). Hence, BACE is an attractive pharmacological target, and numerous research groups have begun searching for potent and selective inhibitors of this enzyme as a potential mechanism for therapeutic intervention in AD. The mature enzyme is composed of a globular catalytic domain that is N-linked glycosylated in mammalian cells, a single transmembrane helix that anchors the enzyme to an intracellular membrane, and a short C-terminal domain that extends outside the phospholipid bilayer of the membrane. Here we have compared the substrate and active site-directed inhibitor binding properties of several recombinant constructs of human BACE. The constructs studied here address the importance of catalytic domain glycosylation state, inclusion of domains other than the catalytic domain, and incorporation into a membrane bilayer on the interactions of the enzyme active site with peptidic ligands. We find no significant differences in ligand binding properties among these various constructs. These data demonstrate that the nonglycosylated, soluble catalytic domain of BACE faithfully reflects the ligand binding properties of the full-length mature enzyme in its natural membrane environment. Thus, the use of the nonglycosylated, soluble catalytic domain of BACE is appropriate for studies aimed at understanding the determinants of ligand recognition by the enzyme active site.
- Published
- 2003
- Full Text
- View/download PDF
21. Evaluation of the kinetic mechanism of Escherichia coli uridine diphosphate-N-acetylmuramate:L-alanine ligase.
- Author
-
Emanuele JJ Jr, Jin H, Yanchunas J Jr, and Villafranca JJ
- Subjects
- Adenosine Triphosphate metabolism, Alanine metabolism, Kinetics, Peptide Synthases antagonists & inhibitors, Peptidoglycan metabolism, Uridine Diphosphate N-Acetylmuramic Acid analogs & derivatives, Uridine Diphosphate N-Acetylmuramic Acid metabolism, Escherichia coli enzymology, Peptide Synthases metabolism
- Abstract
Initial velocity methods were used to probe the kinetic mechanism of Escherichia coli uridine diphosphate-N-acetylmuramate:L-alanine ligase (UNAM:L-Ala ligase). When the activity (in the forward direction) versus substrate concentration data were plotted in double-reciprocal form, all line patterns were intersecting. The best fit of these data was to the equation for an ordered mechanism with the following parameters: k(cat), 1000 +/- 100 min(-1); Kma, 210 +/- 40 microM; Kmb, 84 +/- 20 microM; Kmc, 70 +/- 15 microM; Kia, 180 +/- 50 microM; Kib, 68 +/- 24 microM. Initial velocity line patterns were also determined when the concentration of one substrate was varied at different fixed concentrations of a second substrate while the third substrate was held at a concentration more than 100 times its Km value. Reciprocal plots of data collected with either ATP or L-alanine present at more than 100 times their Km values resulted in intersecting line patterns. Data collected with UNAM present at 100 times its Km value gave a set of parallel lines. These data are consistent with UNAM binding as the second substrate in an ordered mechanism. ADP, uridine diphosphate-N-acetylmuramoyl-L-alanine (UNAMA), and phosphate were tested as product inhibitors versus substrates. None of the products were competitive inhibitors versus L-alanine or UNAM, while the only observed competitive inhibition was ADP versus ATP. These results are consistent with an ordered kinetic mechanism wherein ATP binds first, UNAM binds second, and ADP is the last product released. Rapid quench experiments were performed in the presence of all three substrates or in the presence of ATP and UNAM. The production of acid-labile phosphate as a function of time is characterized by a burst phase followed by a slower linear phase with the rate close to k(cat) in the presence of all three substrates. Only a burst phase was observed for the time course of the reaction in the presence of ATP and UNAM. In both cases, the burst rate was identical. These observations are consistent with L-alanine being the third substrate to bind in a sequential mechanism involving a putative acyl-phosphate intermediate.
- Published
- 1997
- Full Text
- View/download PDF
22. Characterization of NADP+ binding to perdeuterated MurB: backbone atom NMR assignments and chemical-shift changes.
- Author
-
Constantine KL, Mueller L, Goldfarb V, Wittekind M, Metzler WJ, Yanchunas J Jr, Robertson JG, Malley MF, Friedrichs MS, and Farmer BT 2nd
- Subjects
- Amino Acid Sequence, Bacterial Proteins chemistry, Bacterial Proteins metabolism, Carbohydrate Dehydrogenases chemistry, Carbon Isotopes, Deuterium, Magnetic Resonance Spectroscopy methods, Models, Molecular, Molecular Sequence Data, NADP chemistry, Nitrogen Isotopes, Protein Structure, Secondary, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Carbohydrate Dehydrogenases metabolism, NADP metabolism
- Abstract
Backbone-atom resonances have been assigned for both the substrate-free and the NADP+-complexed forms of UDP-N-acetylenolpyruvylglucosamine reductase (MurB), a monomeric, 347-residue (38.5 kDa) flavoenzyme essential for bacterial cell-wall biosynthesis. NMR studies were performed using perdeuterated, uniformly 13C/15N-labeled samples of MurB. In the case of substrate-free MurB, one or more backbone atoms have been assigned for 334 residues (96%). The assigned backbone atoms include 309 1HN and 15N atoms (94%), 315 13CO atoms (91%), 331 13C(alpha) atoms (95%), and 297 13C(beta) atoms (93%). For NADP+-complexed MurB, one or more backbone atoms have been assigned for 313 residues (90%); these include 283 1HN and 15N atoms (86%), 305 13CO atoms (88%), 310 13C(alpha) atoms (89%), and 269 13C(beta) atoms (84%). The strategies used for obtaining resonance assignments are described in detail. Information on the secondary structure in solution for both the substrate-free and NADP+-complexed forms of the enzyme has been derived both from 13C(alpha) and 13C(beta) chemical-shift deviations from random-coil values and from 1HN-1HN NOEs. These data are compared to X-ray crystallographic structures of substrate-free MurB and MurB complexed with the UDP-N-acetylglucosamine enolpyruvate (UNAGEP) substrate. NADP+ binding induces significant chemical-shift changes in residues both within the known UNAGEP and FAD binding pockets and within regions known to undergo conformational changes upon UNAGEP binding. The NMR data indicate that NADP+ and UNAGEP utilize the same binding pocket and, furthermore, that the binding of NADP+ induces structural changes in MurB. Finally, many of the residues within the UNAGEP/NADP+ binding pocket were difficult to assign due to dynamic processes which weaken and/or broaden the respective resonances. Overall, our results are consistent with MurB having a flexible active site.
- Published
- 1997
- Full Text
- View/download PDF
23. Spectroscopic properties of Escherichia coli UDP-N-acetylenolpyruvylglucosamine reductase.
- Author
-
Axley MJ, Fairman R, Yanchunas J Jr, Villafranca JJ, and Robertson JG
- Subjects
- Anaerobiosis, Carbohydrate Dehydrogenases metabolism, Circular Dichroism, Deuterium Oxide, Flavin-Adenine Dinucleotide metabolism, Hydrogen Bonding, Hydrogen-Ion Concentration, Kinetics, NADP metabolism, Oxidation-Reduction, Photochemistry, Protein Denaturation, Protein Folding, Protons, Solvents, Spectrophotometry, Spectrophotometry, Ultraviolet, Substrate Specificity, Uridine Diphosphate N-Acetylglucosamine analogs & derivatives, Uridine Diphosphate N-Acetylglucosamine chemistry, Uridine Diphosphate N-Acetylglucosamine metabolism, Carbohydrate Dehydrogenases chemistry, Escherichia coli enzymology
- Abstract
Purified uridine diphosphate N-acetylenolpyruvylglucosamine reductase (E.C. 1.1.1.158) was analyzed by circular dichroism (CD) and UV-visible spectroscopy to establish the spectral properties of its tightly bound flavin adenine dinucleotide (FAD) cofactor. The polypeptide backbone displayed a single circular dichroic minimum at 208 nm and a single maximum at 193 nm. The CD spectrum of bound flavin exhibited a single major negative Cotton peak at 364 nm and two minor negative Cotton peaks at 464 and 495 nm. The protein was reversibly unfolded in 9.8 M urea and refolded in buffer in the presence of excess FAD. The refolded enzyme incorporated FAD and catalyzed full activity. The bound FAD displayed an absorption maximum at 464 nm with an extinction coefficient of epsilon 464 = 11700 M-1 cm-1. Anaerobic reduction with dithionite was complete at 1 equiv. Anaerobic reduction with nicotinamide adenine dinucleotide phosphate, reduced form (NADPH), also was essentially complete at 1 equiv and produced a long-wavelength absorbance band characteristic of an FAD-pyridine nucleotide charge transfer complex. Photochemical bleaching in the presence of ethylenediaminetetraacetic acid (EDTA) followed exponential kinetics. None of the anaerobic reductive titrations produced a spectral intermediate characteristic of a flavin semiquinone, and all reduced enzyme species could be fully reoxidized by oxygen, with full recovery of catalytic activity. Photochemically reduced enzyme was reoxidized by titration with either NADP+ or uridine diphospho N-acetylglucosamine enolpyruvate (UNAGEP). Reoxidation by NADP+ reached a chemical equilibrium, whereas reoxidation by UNAGEP was stoichiometric. Binding of NADP+ or UNAGEP to the oxidized form of the enzyme produced a dead-end complex that could be titrated by following a 10-nm red shift in the absorption spectrum of the bound FAD. The Kd of NADP+ for oxidized enzyme was 0.7 +/- 0.3 microM and the Kd of UNAGEP was 2.7 +/- 0.3 microM. Solvent deuterium isotope effects on binding were observed for both NADP+ and UNAGEP, depending on the pH. At pH 8.5, the HKd/DKd was 2.2 for NADP+ and 3.9 for UNAGEP. No spectral changes were observed in the presence of a 40-fold excess of uridine diphospho N-acetylmuramic acid (UNAM) either aerobically or anaerobically. These studies have identified spectral signals for five steps in the kinetic mechanism, have indicated that product formation is essentially irreversible, and have indicated that hydrogen bonding or protonation contributes significantly to ground-state complex formation with the physiological substrate.
- Published
- 1997
- Full Text
- View/download PDF
24. Localizing the NADP+ binding site on the MurB enzyme by NMR.
- Author
-
Farmer BT 2nd, Constantine KL, Goldfarb V, Friedrichs MS, Wittekind M, Yanchunas J Jr, Robertson JG, and Mueller L
- Subjects
- Binding Sites, Escherichia coli enzymology, Magnetic Resonance Spectroscopy, Models, Molecular, Protein Conformation, Carbohydrate Dehydrogenases chemistry, NADP chemistry
- Published
- 1996
- Full Text
- View/download PDF
25. Steady-state kinetic mechanism of Escherichia coli UDP-N-acetylenolpyruvylglucosamine reductase.
- Author
-
Dhalla AM, Yanchunas J Jr, Ho HT, Falk PJ, Villafranca JJ, and Robertson JG
- Subjects
- Anaerobiosis, Carbohydrate Dehydrogenases biosynthesis, Carbohydrate Dehydrogenases isolation & purification, Cloning, Molecular, Escherichia coli genetics, Flavin-Adenine Dinucleotide metabolism, Genes, Bacterial, Glucosamine analogs & derivatives, Glucosamine metabolism, Kinetics, Mathematics, NADP metabolism, Recombinant Proteins isolation & purification, Ribonucleotides pharmacology, Uridine Diphosphate analogs & derivatives, Uridine Diphosphate metabolism, Carbohydrate Dehydrogenases metabolism, Escherichia coli enzymology, Recombinant Proteins metabolism, Uridine Diphosphate N-Acetylglucosamine analogs & derivatives
- Abstract
The Escherichia coli MurB gene encoding UDP-N-acetylenolpyruvylglucosamine reductase was expressed to a level of approximately 100 mg/L as a fusion construct with maltose binding protein. Rapid affinity purification, proteolysis, and anion exchange chromatography yielded homogeneous enzyme containing 1 mol/mol bound FAD. Enzyme was maximally activated by K+, NH4+, and Rb+ at cation concentrations between 10 and 50 mM. Steady-state enzyme kinetics at pH 8.0 and 37 degrees C revealed weak and strong substrate inhibition by NADPH and UDP-N-acetylenolpyruvylglucosamine, respectively, where the KiS were 910 microM and 73 microM. Substrate inhibition was pH dependent for both substrates. Initial velocity measurements as a function of both substrates produced patterns consistent with a ping pong bi bi double competitive substrate inhibition mechanism. Data at pH 8.0 yielded kinetic constants corresponding to Km,UNAGEP = 24 +/- 3 microM, Ki,UNAGEP = 73 +/- 19 microM, Km,NADPH = 17 +/- 3 microM, Ki,NADPH = 910 +/- 670 microM, and kcat = 62 +/- 3 s-1. A slow anaerobic exchange reaction with thio-NADP+ provided evidence for release of NADP+ in the absence of UNAGEP. Alternate reduced nicotinamide dinucleotides, including NHXDPH, 3'-NADPH, and alpha-NADPH, were substrates, whereas NADH was not. Several nucleotides, including ADP and UDP, were weak inhibitors of the enzyme with inhibition constants between 5 and 97 mM. Various analogs of NADP+, including 3'-NADP+, thio-NADP+, APADP+, NEthDP+, and NHXDP+, were inhibitors of the enzyme with respect to NADPH and yielded inhibition constants in the range of 110-1100 microM. Analogs without the 2'- or 3'-phosphate of NADPH or NADP+ were not substrates or inhibitors. Double inhibition experiments with varied APADP+ and UNAG produced inhibition patterns consistent with mutually exclusive inhibitor binding. The data suggest that NADPH and UNAGEP share a subsite that prevents both molecules from binding at once.
- Published
- 1995
- Full Text
- View/download PDF
26. Supramolecular self-assembly of Escherichia coli glutamine synthetase: characterization of dodecamer stacking and high order association.
- Author
-
Yanchunas J Jr, Dabrowski MJ, Schurke P, and Atkins WM
- Subjects
- Cations, Divalent, Cobalt pharmacology, Copper pharmacology, Half-Life, Histidine chemistry, Hydrogen-Ion Concentration, Light, Macromolecular Substances, Microscopy, Electron, Osmolar Concentration, Potassium Chloride pharmacology, Scattering, Radiation, Temperature, Zinc pharmacology, Escherichia coli enzymology, Glutamate-Ammonia Ligase chemistry
- Abstract
Dodecameric glutamine synthetase (GS) from bacteria is formed from two face-to-face hexameric rings of identical subunits. These highly symmetrical aggregates from some bacteria, including Escherichia coli, "stack" in the presence of Zn2+ and other divalent ions to generate protein tubes (phase I) and subsequently associate side-to-side to yield "cables" and nonspecific aggregates (phase II). In order to understand the molecular mechanisms of recognition leading to this macromolecular self-assembly, the effects of solution conditions on the kinetics of these processes have been studied. These reactions have been monitored by changes in light scattering and by electron microscopy. Conditions have been established for isolation of phases I and II. At 0.04 mg of GS/mL, pH 7.0, 100 mM KCl, and 1 mM Mn2+, 25 degrees C, minimal side-to-side aggregation occurs, and the stacking reaction follows second-order kinetics, with respect to GS, at low extent of reaction. The second-order rate constants determined for phase I, initiated by Zn2+ or Co2+, demonstrate a pH optimum at 7.0-7.25, whereas phase II is favored at pHs below 6.5. The pH profile for the stacking reaction suggests that His residues are involved, and modification of 2-3 histidines/subunit with diethyl pyrocarbonate (DEPC) is sufficient to completely inhibit metal-dependent dodecamer stacking. The effect of ionic strength on GS stacking was also studied. Although hydrophobic interactions have previously been assumed to dominate this protein-protein association, both phase I and phase II of the assembly are inhibited by KCl and NaCl, suggesting that ionic interactions also play an essential role.(ABSTRACT TRUNCATED AT 250 WORDS)
- Published
- 1994
- Full Text
- View/download PDF
27. Supramolecular self-assembly of glutamine synthetase: mutagenesis of a novel intermolecular metal binding site required for dodecamer stacking.
- Author
-
Dabrowski MJ, Yanchunas J Jr, Villafranca BC, Dietze EC, Schurke P, and Atkins WM
- Subjects
- Binding Sites, Cobalt metabolism, Cobalt pharmacology, Computer Simulation, Copper metabolism, Glutamate-Ammonia Ligase genetics, Macromolecular Substances, Magnesium metabolism, Manganese metabolism, Metals pharmacology, Microscopy, Electron, Models, Molecular, Molecular Structure, Protein Structure, Secondary, Structure-Activity Relationship, Zinc metabolism, Escherichia coli enzymology, Glutamate-Ammonia Ligase chemistry, Metals metabolism, Mutagenesis, Site-Directed
- Abstract
Dodecameric glutamine synthetase (GS) from Escherichia coli assembles into highly ordered supramolecular protein tubes in the presence of several divalent metal ions. The molecular mechanism for this metal-induced self-assembly of the E. coli GS has been studied by molecular modeling and site-directed mutagenesis. The X-ray crystal structure of the nearly identical Salmonella typhimurium GS has been used to construct a model of the "stacked" complex between two dodecamers. A complementary fit, based on steric constraints, reveals a possible interaction between the N-terminal helices from adjacent dodecamers. The amino acid side chains of His and Met residues within the helices from each of the subunits of one face of a dodecamer lie within approximately 3.5 A of the analogous side chains in the subunits from the adjacent dodecamer in the stacked complex. His-4, Met-8, and His-12 from adjacent helices provide potential ligands for a binuclear metal binding site. Replacement of each of these surface residues with aliphatic amino acids has negligible effects on the enzymatic activity, the regulation of activity via adenylylation, and gross dodecameric structure. However, the rate and extent of metal ion-mediated self-assembly of GS tubules are reduced to < 2% of the wild-type protein in the single mutants H4A, H12L, and H12D. The M8L mutant demonstrates a 3-fold decrease in the bimolecular rate constant for stacking, but electron microscopy indicates that this mutant does form stacked tubes. The cysteine-containing mutants H4C, M8C, and H12C were also constructed.(ABSTRACT TRUNCATED AT 250 WORDS)
- Published
- 1994
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.