31 results on '"Kavana M"'
Search Results
2. Discriminatory Interaction Behavior of Lipid Vesicles toward Diversely Emissive Carbon Dots Synthesized from Ortho, Meta, and Para Isomeric Carbon Precursors
- Author
-
Kavana M, Anjan Chakraborty, and Nishu Kanwa
- Subjects
Photoluminescence ,Biocompatibility ,Cellular process ,Vesicle ,chemistry.chemical_element ,Biological membrane ,02 engineering and technology ,Surfaces and Interfaces ,010402 general chemistry ,021001 nanoscience & nanotechnology ,Condensed Matter Physics ,01 natural sciences ,0104 chemical sciences ,Dynamic light scattering ,chemistry ,Electrochemistry ,Biophysics ,General Materials Science ,Lipid vesicle ,0210 nano-technology ,Carbon ,Spectroscopy - Abstract
Photoluminescent carbon dots (C-dots) are widely used for bioimaging techniques to study different cellular processes. However, biocompatibility of C-dots is crucial because the wrong selection of C-dots may lead to an adverse effect on a particular cellular process. Herein, we investigate the interaction of zwitterionic lipid vesicles with photoluminescent C-dots derived from different isomeric (ortho, meta, and para) precursors of phenylenediamine (PDA) by spectroscopic and microscopic imaging techniques as well as dynamic light scattering methods. The study reveals that interaction of lipid vesicles with C-dots is highly dependent on the properties of the isomeric precursors. We find that vesicles retain their morphology upon interaction with ortho C-dots (oCD). The microscopic images reveal that oCD are selectively embedded in the lipid vesicles and can effectively be used for imaging purpose. On the contrary, meta and para C-dots (mCD and pCD) being located on the interfacial region induce aggregation in the vesicles. We explain the observation in terms of the location of the C-dots on the lipid vesicles, their electrostatic attraction at the vesicle interface, possible cross-linking with other vesicles and different hydration features of the isomeric precursors of the C-dots. The study may be helpful in understanding the interactions and attachment processes of C-dots at the interface of biological membranes.
- Published
- 2020
- Full Text
- View/download PDF
3. Intramolecular CH Insertion Reactions of (Pentamethylcyclopentadienyl)Rhenium Alkynylcarbene Complexes
- Author
-
Casey, C. P., Kraft, S., and Kavana, M.
- Abstract
Thermolysis of the alkynylcarbene complex Cp*(CO)
2 Re&dbd;C(Ph)13C&tbd1;13CTol (5 ) at 120 °C resulted in rapid equilibration (t1/2 = 33 min) to a 1:1 mixture of5 and Cp*(CO)2 Re&dbd;13C(Tol)13C&tbd1;CPh (7 ) via a [1,3]-rhenium shift. Extended thermolysis of this mixture provided the CH insertion products {η5:η2-[C5 (CH3 )4 CH2 13CH(Tol)13C&tbd1;CPh]}Re(CO)2 (8 ) and {η5:η2-[C5 (CH3 )4 CH2 CH(Ph)13C&tbd1;13CTol]}Re(CO)2 (9 ). Thermolysis of the symmetrically substituted alkynylcarbene complex Cp*(CO)2 Re&dbd;C(Ph)C&tbd1;CPh (6 ) produced the CH insertion product {η5:η2-[C5 (CH3 )4 CH2 CH(Ph)C&tbd1;CPh]}Re(CO)2 (10 ). The CH insertion of Cp*(CO)2 Re&dbd;C(Ph)C&tbd1;CC6 D5 (6-HD ) was monitored at low conversion before complete equilibration with Cp*(CO)2 Re&dbd;C(C6 D5 )C&tbd1;CPh (6-DH ) occurred. An excess of {η5:η2-[C5 (CH3 )4 CH2 CH(C6 D5 )C&tbd1;CPh]}Re(CO)2 (13-HD ) over {η5:η2-[C5 (CH3 )4 CH2 CH(Ph)C&tbd1;CC6 D5 ]}Re(CO)2 (13-DH ) provides evidence for site-selective CH insertion of the remote alkyne carbon into the CH bond of a Cp* methyl group.- Published
- 2001
4. Reaction of rhenium alkynyl carbene complexes with tertiary phosphines produces dihydrophospholium rhenium complexes by a formal CH insertion process
- Author
-
Casey, C. P., Kraft, S., Powell, D. R., and Kavana, M.
- Published
- 2001
- Full Text
- View/download PDF
5. Static versus dynamic Jahn-Teller distortions in octahedral-like copper(II) complexes of 1,4,7-triazacyclononane and 1-oxa-4,7-diazacyclononane: implications for hydrolytic reactivity
- Author
-
Kavana, M., Powell, D.R., and Burstyn, J.N.
- Published
- 2000
- Full Text
- View/download PDF
6. High-Throughput Covalent Modifier Screening with Acoustic Ejection Mass Spectrometry.
- Author
-
Wen X, Liu C, Tovar K, Curran P, Richards M, Agrawal S, Johnstone R, Loy RE, Methot JL, Mansueto MS, Koglin M, Wildey MJ, Burton L, Covey TR, Bateman KP, Kavana M, and McLaren DG
- Subjects
- Small Molecule Libraries chemistry, Humans, Acoustics, Drug Discovery methods, Ligands, Mass Spectrometry methods, High-Throughput Screening Assays methods
- Abstract
Interests in covalent drugs have grown in modern drug discovery as they could tackle challenging targets traditionally considered "undruggable". The identification of covalent binders to target proteins typically involves directly measuring protein covalent modifications using high-resolution mass spectrometry. With a continually expanding library of compounds, conventional mass spectrometry platforms such as LC-MS and SPE-MS have become limiting factors for high-throughput screening. Here, we introduce a prototype high-resolution acoustic ejection mass spectrometry (AEMS) system for the rapid screening of a covalent modifier library comprising ∼10,000 compounds against a 50 kDa-sized target protein─Werner syndrome helicase. The screening samples were arranged in a 1536-well format. The sample buffer containing high-concentration salts was directly analyzed without any cleanup steps, minimizing sample preparation efforts and ensuring protein stability. The entire AEMS analysis process could be completed within a mere 17 h. An automated data analysis tool facilitated batch processing of the sample data and quantitation of the formation of various covalent protein-ligand adducts. The screening results displayed a high degree of fidelity, with a Z ' factor of 0.8 and a hit rate of 2.3%. The identified hits underwent orthogonal testing in a biochemical activity assay, revealing that 75% were functional antagonists of the target protein. Notably, a comparative analysis with LC-MS showcased the AEMS platform's low risk of false positives or false negatives. This innovative platform has enabled robust high-throughput covalent modifier screening, featuring a 10-fold increase in library size and a 10- to 100-fold increase in throughput when compared with similar reports in the existing literature.
- Published
- 2024
- Full Text
- View/download PDF
7. From Screening to Targeted Degradation: Strategies for the Discovery and Optimization of Small Molecule Ligands for PCSK9.
- Author
-
Petrilli WL, Adam GC, Erdmann RS, Abeywickrema P, Agnani V, Ai X, Baysarowich J, Byrne N, Caldwell JP, Chang W, DiNunzio E, Feng Z, Ford R, Ha S, Huang Y, Hubbard B, Johnston JM, Kavana M, Lisnock JM, Liang R, Lu J, Lu Z, Meng J, Orth P, Palyha O, Parthasarathy G, Salowe SP, Sharma S, Shipman J, Soisson SM, Strack A, Youm H, Zhao K, Zink DL, Zokian H, Addona GH, Akinsanya K, Tata JR, Xiong Y, and Imbriglio JE
- Published
- 2021
- Full Text
- View/download PDF
8. From Screening to Targeted Degradation: Strategies for the Discovery and Optimization of Small Molecule Ligands for PCSK9.
- Author
-
Petrilli WL, Adam GC, Erdmann RS, Abeywickrema P, Agnani V, Ai X, Baysarowich J, Byrne N, Caldwell JP, Chang W, DiNunzio E, Feng Z, Ford R, Ha S, Huang Y, Hubbard B, Johnston JM, Kavana M, Lisnock JM, Liang R, Lu J, Lu Z, Meng J, Orth P, Palyha O, Parthasarathy G, Salowe SP, Sharma S, Shipman J, Soisson SM, Strack AM, Youm H, Zhao K, Zink DL, Zokian H, Addona GH, Akinsanya K, Tata JR, Xiong Y, and Imbriglio JE
- Subjects
- Humans, Ligands, Models, Molecular, Molecular Structure, Serine Proteinase Inhibitors chemistry, Small Molecule Libraries chemistry, Drug Discovery, Drug Evaluation, Preclinical, Proprotein Convertase 9 metabolism, Proteolysis drug effects, Serine Proteinase Inhibitors pharmacology, Small Molecule Libraries pharmacology
- Abstract
Proprotein convertase substilisin-like/kexin type 9 (PCSK9) is a serine protease involved in a protein-protein interaction with the low-density lipoprotein (LDL) receptor that has both human genetic and clinical validation. Blocking this protein-protein interaction prevents LDL receptor degradation and thereby decreases LDL cholesterol levels. Our pursuit of small-molecule direct binders for this difficult to drug PPI target utilized affinity selection/mass spectrometry, which identified one confirmed hit compound. An X-ray crystal structure revealed that this compound was binding in an unprecedented allosteric pocket located between the catalytic and C-terminal domain. Optimization of this initial hit, using two distinct strategies, led to compounds with high binding affinity to PCSK9. Direct target engagement was demonstrated in the cell lysate with a cellular thermal shift assay. Finally, ligand-induced protein degradation was shown with a proteasome recruiting tag attached to the high-affinity allosteric ligand for PCSK9., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
9. Label-Free, LC-MS-Based Assays to Quantitate Small-Molecule Antagonist Binding to the Mammalian BLT1 Receptor.
- Author
-
Chen X, Stout S, Mueller U, Boykow G, Visconti R, Siliphaivanh P, Spencer K, Presland J, Kavana M, Basso AD, McLaren DG, and Myers RW
- Abstract
We have developed and validated label-free, liquid chromatography-mass spectrometry (LC-MS)-based equilibrium direct and competition binding assays to quantitate small-molecule antagonist binding to recombinant human and mouse BLT1 receptors expressed in HEK 293 cell membranes. Procedurally, these binding assays involve (1) equilibration of the BLT1 receptor and probe ligand, with or without a competitor; (2) vacuum filtration through cationic glass fiber filters to separate receptor-bound from free probe ligand; and (3) LC-MS analysis in selected reaction monitoring mode for bound probe ligand quantitation. Two novel, optimized probe ligands, compounds 1 and 2 , were identified by screening 20 unlabeled BLT1 antagonists for direct binding. Saturation direct binding studies confirmed the high affinity, and dissociation studies established the rapid binding kinetics of probe ligands 1 and 2 . Competition binding assays were established using both probe ligands, and the affinities of structurally diverse BLT1 antagonists were measured. Both binding assay formats can be executed with high specificity and sensitivity and moderate throughput (96-well plate format) using these approaches. This highly versatile, label-free method for studying ligand binding to membrane-associated receptors should find broad application as an alternative to traditional methods using labeled ligands.
- Published
- 2017
- Full Text
- View/download PDF
10. Novel, highly potent systemic glucokinase activators for the treatment of Type 2 Diabetes Mellitus.
- Author
-
Xu J, Lin S, Myers RW, Addona G, Berger JP, Campbell B, Chen HS, Chen Z, Eiermann GJ, Elowe NH, Farrer BT, Feng W, Fu Q, Kats-Kagan R, Kavana M, Malkani S, McMasters DR, Mitra K, Pachanski MJ, Tong X, Trujillo ME, Xu L, Zhang B, Zhang F, Zhang R, and Parmee ER
- Subjects
- Allosteric Regulation drug effects, Animals, Blood Glucose analysis, Blood Glucose metabolism, Diabetes Mellitus, Type 2 blood, Diabetes Mellitus, Type 2 metabolism, Drug Design, Enzyme Activation drug effects, Enzyme Activators pharmacokinetics, Enzyme Activators pharmacology, Humans, Hypoglycemic Agents pharmacokinetics, Hypoglycemic Agents pharmacology, Indoles pharmacokinetics, Indoles pharmacology, Insulin blood, Insulin metabolism, Mice, Mice, Inbred C57BL, Diabetes Mellitus, Type 2 drug therapy, Enzyme Activators chemistry, Enzyme Activators therapeutic use, Glucokinase metabolism, Hypoglycemic Agents chemistry, Hypoglycemic Agents therapeutic use, Indoles chemistry, Indoles therapeutic use
- Abstract
Glucokinase (GK, hexokinase IV) is a unique hexokinase that plays a central role in mammalian glucose homeostasis. Glucose phosphorylation by GK in the pancreatic β-cell is the rate-limiting step that controls glucose-stimulated insulin secretion. Similarly, GK-mediated glucose phosphorylation in hepatocytes plays a major role in increasing hepatic glucose uptake and metabolism and possibly lowering hepatic glucose output. Small molecule GK activators (GKAs) have been identified that increase enzyme activity by binding to an allosteric site. GKAs offer a novel approach for the treatment of Type 2 Diabetes Mellitus (T2DM) and as such have garnered much attention. We now report the design, synthesis, and biological evaluation of a novel series of 2,5,6-trisubstituted indole derivatives that act as highly potent GKAs. Among them, Compound 1 was found to possess high in vitro potency, excellent physicochemical properties, and good pharmacokinetic profile in rodents. Oral administration of Compound 1 at doses as low as 0.03mg/kg led to robust blood glucose lowering efficacy in 3week high fat diet-fed mice., (Copyright © 2016. Published by Elsevier Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
11. Discovery of orally active hepatoselective glucokinase activators for treatment of Type II Diabetes Mellitus.
- Author
-
Xu J, Lin S, Myers RW, Trujillo ME, Pachanski MJ, Malkani S, Chen HS, Chen Z, Campbell B, Eiermann GJ, Elowe N, Farrer BT, Feng W, Fu Q, Kats-Kagan R, Kavana M, McMasters DR, Mitra K, Tong X, Xu L, Zhang F, Zhang R, Addona GH, Berger JP, Zhang B, and Parmee ER
- Subjects
- Animals, Diabetes Mellitus, Type 2 blood, Diabetes Mellitus, Type 2 metabolism, Drug Discovery, Enzyme Activators chemistry, Enzyme Activators pharmacokinetics, Enzyme Activators therapeutic use, Humans, Hypoglycemic Agents chemistry, Hypoglycemic Agents pharmacokinetics, Hypoglycemic Agents therapeutic use, Insulin blood, Liver drug effects, Liver metabolism, Mice, Mice, Inbred C57BL, Pancreas drug effects, Pancreas metabolism, Pyridines chemistry, Pyridines pharmacokinetics, Pyridines therapeutic use, Diabetes Mellitus, Type 2 drug therapy, Enzyme Activators pharmacology, Glucokinase metabolism, Hypoglycemic Agents pharmacology, Pyridines pharmacology
- Abstract
Systemically acting glucokinase activators (GKA) have been demonstrated in clinical trials to effectively lower blood glucose in patients with type II diabetes. However, mechanism-based hypoglycemia is a major adverse effect that limits the therapeutic potential of these agents. We hypothesized that the predominant mechanism leading to hypoglycemia is GKA-induced excessive insulin secretion from pancreatic β-cells at (sub-)euglycemic levels. We further hypothesized that restricting GK activation to hepatocytes would maintain glucose-lowering efficacy while significantly reducing hypoglycemic risk. Here we report the discovery of a novel series of carboxylic acid substituted GKAs based on pyridine-2-carboxamide. These GKAs exhibit preferential distribution to the liver versus the pancreas in mice. SAR studies led to the identification of a potent and orally active hepatoselective GKA, compound 6. GKA 6 demonstrated robust glucose lowering efficacy in high fat diet-fed mice at doses ⩾10mpk, with ⩾70-fold liver:pancreas distribution, minimal effects on plasma insulin levels, and significantly reduced risk of hypoglycemia., (Copyright © 2016. Published by Elsevier Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
12. Quantitation of wall teichoic acid in Staphylococcus aureus by direct measurement of monomeric units using LC-MS/MS.
- Author
-
Berejnaia O, Wang H, Labroli M, Yang C, Gill C, Xiao J, Hesk D, DeJesus R, Su J, Tan CM, Sheth PR, Kavana M, and McLaren DG
- Subjects
- Chromatography, Liquid methods, Methicillin-Resistant Staphylococcus aureus chemistry, Teichoic Acids chemistry, Tunicamycin pharmacology, Mass Spectrometry methods, Methicillin-Resistant Staphylococcus aureus metabolism, Teichoic Acids metabolism
- Abstract
The emergence of methicillin-resistant Staphylococcus aureus (MRSA) has created an urgent need for new therapeutic agents capable of combating this threat. We have previously reported on the discovery of novel inhibitors targeting enzymes involved in the biosynthesis of wall teichoic acid (WTA) and demonstrated that these agents can restore β-lactam efficacy against MRSA. In those previous reports pathway engagement of inhibitors was demonstrated by reduction in WTA levels measured by polyacrylamide gel electrophoresis. To enable a more rigorous analysis of these inhibitors we sought to develop a quantitative method for measuring whole-cell reductions in WTA. Herein we describe a robust methodology for hydrolyzing polymeric WTA to the monomeric component ribitol-N-acetylglucosamine coupled with measurement by LC-MS/MS. Critical elements of the protocol were found to include the time and temperature of hydrofluoric acid-mediated hydrolysis of polymeric WTA and optimization of these parameters is fully described. Most significantly, the assay enabled accurate and reproducible measurement of depletion EC
50s for tunicamycin and representatives from the novel class of TarO inhibitors, the tarocins. The method described can readily be adapted to quantifying levels of WTA in tissue homogenates from a murine model of infection, highlighting the applicability for both in vitro and in vivo characterizations., (Copyright © 2016 Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
13. In Vitro Assays for the Discovery of PCSK9 Autoprocessing Inhibitors.
- Author
-
Salowe SP, Zhang L, Zokian HJ, Gesell JJ, Zink DL, Wiltsie J, Ai X, Kavana M, and Pinto S
- Subjects
- Escherichia coli genetics, Fluorescence Resonance Energy Transfer methods, Hep G2 Cells, Humans, Hypercholesterolemia genetics, Kinetics, Lactones antagonists & inhibitors, Mass Spectrometry methods, PCSK9 Inhibitors, Proprotein Convertase 9 genetics, Protein Conformation drug effects, Receptors, LDL genetics, Drug Delivery Systems methods, High-Throughput Screening Assays methods, Hypercholesterolemia drug therapy, Proprotein Convertase 9 chemistry
- Abstract
PCSK9 plays a significant role in regulating low-density lipoprotein (LDL) cholesterol levels and has become an important drug target for treating hypercholesterolemia. Although a member of the serine protease family, PCSK9 only catalyzes a single reaction, the autocleavage of its prodomain. The maturation of the proprotein is an essential prerequisite for the secretion of PCSK9 to the extracellular space where it binds the LDL receptor and targets it for degradation. We have found that a construct of proPCSK9 where the C-terminal domain has been truncated has sufficient stability to be expressed and purified from Escherichia coli for the in vitro study of autoprocessing. Using automated Western analysis, we demonstrate that autoprocessing exhibits the anticipated first-order kinetics. A high-throughput time-resolved fluorescence resonance energy transfer assay for autocleavage has been developed using a PCSK9 monoclonal antibody that is sensitive to the conformational changes that occur upon maturation of the proprotein. Kinetic theory has been developed that describes the behavior of both reversible and irreversible inhibitors of autocleavage. The analysis of an irreversible lactone inhibitor validates the expected relationship between potency and the reaction end point. An orthogonal liquid chromatography-mass spectrometry assay has also been implemented for the confirmation of hits from the antibody-based assays.
- Published
- 2016
- Full Text
- View/download PDF
14. Moderate to high throughput in vitro binding kinetics for drug discovery.
- Author
-
Zhang R, Barbieri CM, Garcia-Calvo M, Myers RW, McLaren D, and Kavana M
- Subjects
- Biosensing Techniques methods, Enzyme Activators chemistry, Enzyme Activators pharmacology, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Pharmacokinetics, Surface Plasmon Resonance methods, Tandem Mass Spectrometry, Drug Discovery methods, Enzyme Activators pharmacokinetics, Enzyme Inhibitors pharmacokinetics, High-Throughput Screening Assays methods
- Abstract
This review provides a concise summary for state of the art, moderate to high throughput in vitro technologies being employed to study drug-target binding kinetics. These technologies cover a wide kinetic timescale spanning up to nine orders of magnitude from milliseconds to days. Automated stopped flow measures transient and (pre)steady state kinetics from milliseconds to seconds. For seconds to hours timescale kinetics we discuss surface plasmon resonance-based biosensor, global progress curve analysis for high throughput kinetic profiling of enzyme inhibitors and activators, and filtration plate-based radioligand or fluorescent binding assays for receptor binding kinetics. Jump dilution after pre-incubation is the preferred method for very slow kinetics lasting for days. The basic principles, best practices and simulated data for these technologies are described. Finally, the application of a universal label-free technology, liquid chromatography coupled tandem mass spectrometry (LC/MS/MS), is briefly reviewed. Select literature references are highlighted for in-depth understanding. A new reality is dawning wherein binding kinetics is an integral and routine part of mechanism of action elucidation and translational, quantitative pharmacology for drug discovery.
- Published
- 2016
- Full Text
- View/download PDF
15. Bridging Model-Dependent Receptor Agonism and Allosterism Parameterization.
- Author
-
Zhang R and Kavana M
- Subjects
- Allosteric Regulation, Humans, Allosteric Site
- Published
- 2016
- Full Text
- View/download PDF
16. Quantitative Measure of Receptor Agonist and Modulator Equi-Response and Equi-Occupancy Selectivity.
- Author
-
Zhang R and Kavana M
- Subjects
- Algorithms, Allosteric Regulation, Animals, Drug Design, Humans, Structure-Activity Relationship, Receptors, G-Protein-Coupled agonists
- Abstract
G protein-coupled receptors (GPCRs) are an important class of drug targets. Quantitative analysis by global curve fitting of properly designed dose-dependent GPCR agonism and allosterism data permits the determination of all affinity and efficacy parameters based on a general operational model. We report here a quantitative and panoramic measure of receptor agonist and modulator equi-response and equi-occupancy selectivity calculated from these parameters. The selectivity values help to differentiate not only one agonist or modulator from another, but on-target from off-target receptor or functional pathway as well. Furthermore, in conjunction with target site free drug concentrations and endogenous agonist tones, the allosterism parameters and selectivity values may be used to predict in vivo efficacy and safety margins.
- Published
- 2016
- Full Text
- View/download PDF
17. Quantitative analysis of receptor allosterism and its implication for drug discovery.
- Author
-
Zhang R and Kavana M
- Subjects
- Allosteric Regulation drug effects, Allosteric Site, Drug Discovery methods, Humans, Receptors, G-Protein-Coupled metabolism, Structure-Activity Relationship, Drug Design, Models, Biological, Receptors, G-Protein-Coupled drug effects
- Abstract
Introduction: G protein-coupled receptors represent the largest class of druggable targets and are known to be modulated by both orthosteric agonists and positive/negative allosteric modulators (PAMs/NAMs). Proper experimental design and data analysis for the dose matrix between an agonist and PAM or NAM are critical to elucidate the key parameters for understanding molecular mechanism and structure-activity relationship (SAR) in drug discovery., Areas Covered: The authors provide an overview and best practice recommendations on the quantitative analysis of receptor allosterism. The authors propose a simple classification system for receptor modulators on the basis of their efficacy and affinity modifiers. The authors also outline the optimal assay designs for both fixed dose screening and dose matrix study of receptor modulators., Expert Opinion: The authors recommend the global curve fitting approach to reliably yield system- and modulator-specific parameters for SAR ranking. Furthermore, the authors suggest that the uncertainty in maximal system response has insignificant impact on SAR ranking. The authors anticipate that systems pharmacology models integrating both binding kinetics and functional allosterism will be needed to address the inherent limitations of current allosterism models.
- Published
- 2015
- Full Text
- View/download PDF
18. Biochemical characterization of cholesteryl ester transfer protein inhibitors.
- Author
-
Ranalletta M, Bierilo KK, Chen Y, Milot D, Chen Q, Tung E, Houde C, Elowe NH, Garcia-Calvo M, Porter G, Eveland S, Frantz-Wattley B, Kavana M, Addona G, Sinclair P, Sparrow C, O'Neill EA, Koblan KS, Sitlani A, Hubbard B, and Fisher TS
- Subjects
- Amides, Animals, Anticholesteremic Agents metabolism, Blood Proteins metabolism, Esters, Humans, Mice, Molecular Structure, Oxazolidinones metabolism, Quinolines metabolism, Sulfhydryl Compounds metabolism, Anticholesteremic Agents chemistry, Cholesterol Ester Transfer Proteins antagonists & inhibitors, Oxazolidinones chemistry, Quinolines chemistry, Sulfhydryl Compounds chemistry
- Abstract
Cholesteryl ester transfer protein (CETP) has been identified as a novel target for increasing HDL cholesterol levels. In this report, we describe the biochemical characterization of anacetrapib, a potent inhibitor of CETP. To better understand the mechanism by which anacetrapib inhibits CETP activity, its biochemical properties were compared with CETP inhibitors from distinct structural classes, including torcetrapib and dalcetrapib. Anacetrapib and torcetrapib inhibited CETP-mediated cholesteryl ester and triglyceride transfer with similar potencies, whereas dalcetrapib was a significantly less potent inhibitor. Inhibition of CETP by both anacetrapib and torcetrapib was not time dependent, whereas the potency of dalcetrapib significantly increased with extended preincubation. Anacetrapib, torcetrapib, and dalcetrapib compete with one another for binding CETP; however anacetrapib binds reversibly and dalcetrapib covalently to CETP. In addition, dalcetrapib was found to covalently label both human and mouse plasma proteins. Each CETP inhibitor induced tight binding of CETP to HDL, indicating that these inhibitors promote the formation of a complex between CETP and HDL, resulting in inhibition of CETP activity.
- Published
- 2010
- Full Text
- View/download PDF
19. PCSK9 is not involved in the degradation of LDL receptors and BACE1 in the adult mouse brain.
- Author
-
Liu M, Wu G, Baysarowich J, Kavana M, Addona GH, Bierilo KK, Mudgett JS, Pavlovic G, Sitlani A, Renger JJ, Hubbard BK, Fisher TS, and Zerbinatti CV
- Subjects
- Amyloid beta-Protein Precursor metabolism, Animals, Brain anatomy & histology, HEK293 Cells, Humans, Male, Mice, Mice, Knockout, Proprotein Convertase 9, Proprotein Convertases, Protein Binding, Serine Endopeptidases genetics, Amyloid Precursor Protein Secretases metabolism, Aspartic Acid Endopeptidases metabolism, Brain metabolism, LDL-Receptor Related Proteins metabolism, Receptors, LDL metabolism, Serine Endopeptidases metabolism
- Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a secreted protein that regulates hepatic low-density lipoprotein receptor (LDLR) levels in humans. PCSK9 has also been shown to regulate the levels of additional membrane-bound proteins in vitro, including the very low-density lipoprotein receptor (VLDLR), apolipoprotein E receptor 2 (ApoER2) and the beta-site amyloid precursor protein (APP)-cleaving enzyme 1 (BACE1), which are all highly expressed in the CNS and have been implicated in Alzheimer's disease. To better understand the role of PCSK9 in regulating these additional target proteins in vivo, their steady-state levels were measured in the brain of wild-type, PCSK9-deficient, and human PCSK9 overexpressing transgenic mice. We found that while PCSK9 directly bound to recombinant LDLR, VLDLR, and apoER2 protein in vitro, changes in PCSK9 expression did not alter the level of these receptors in the mouse brain. In addition, we found no evidence that PCSK9 regulates BACE1 levels or APP processing in the mouse brain. In conclusion, our results suggest that while PCSK9 plays an important role in regulating circulating LDL cholesterol levels by reducing the number of hepatic LDLRs, it does not appear to modulate the levels of LDLR and other membrane-bound proteins in the adult mouse brain.
- Published
- 2010
- Full Text
- View/download PDF
20. Investigation of functionally liver selective glucokinase activators for the treatment of type 2 diabetes.
- Author
-
Bebernitz GR, Beaulieu V, Dale BA, Deacon R, Duttaroy A, Gao J, Grondine MS, Gupta RC, Kakmak M, Kavana M, Kirman LC, Liang J, Maniara WM, Munshi S, Nadkarni SS, Schuster HF, Stams T, St Denny I, Taslimi PM, Vash B, and Caplan SL
- Subjects
- Animals, Blood Glucose drug effects, Diabetes Mellitus, Type 2 drug therapy, Glucose Tolerance Test, Hypoglycemic Agents pharmacology, Liver drug effects, Liver metabolism, Mice, Structure-Activity Relationship, Sulfonamides therapeutic use, Glucokinase drug effects, Sulfonamides pharmacology
- Abstract
Type 2 diabetes is a polygenic disease which afflicts nearly 200 million people worldwide and is expected to increase to near epidemic levels over the next 10-15 years. Glucokinase (GK) activators are currently under investigation by a number of pharmaceutical companies with only a few reaching early clinical evaluation. A GK activator has the promise of potentially affecting both the beta-cells of the pancreas, by improving glucose sensitive insulin secretion, as well as the liver, by reducing uncontrolled glucose output and restoring post-prandial glucose uptake and storage as glycogen. Herein, we report our efforts on a sulfonamide chemotype with the aim to generate liver selective GK activators which culminated in the discovery of 3-cyclopentyl-N-(5-methoxy-thiazolo[5,4-b]pyridin-2-yl)-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionamide (17c). This compound activated the GK enzyme (alphaK(a) = 39 nM) in vitro at low nanomolar concentrations and significantly reduced glucose levels during an oral glucose tolerance test in normal mice.
- Published
- 2009
- Full Text
- View/download PDF
21. Characterization of recombinant human acetyl-CoA carboxylase-2 steady-state kinetics.
- Author
-
Kaushik VK, Kavana M, Volz JM, Weldon SC, Hanrahan S, Xu J, Caplan SL, and Hubbard BK
- Subjects
- Acetyl-CoA Carboxylase antagonists & inhibitors, Chromatography, Liquid, Humans, Kinetics, Recombinant Proteins antagonists & inhibitors, Recombinant Proteins chemistry, Tandem Mass Spectrometry, Acetyl-CoA Carboxylase chemistry
- Abstract
Acetyl-CoA carboxylase (ACC) catalyzes the carboxylation of acetyl-CoA to form malonyl-CoA, a key metabolite in the fatty acid synthetic and oxidation pathways. The present study describes the steady-state kinetic analysis of a purified recombinant human form of the enzyme, namely ACC2, using a novel LC/MS/MS assay to directly measure malonyl-CoA formation. Four dimensional matrices, in which bicarbonate (HCO(3)(-)), ATP, acetyl-CoA, and citrate were varied, and global data fitting to appropriate steady-state equations were used to generate kinetic constants. Product inhibition studies support the notion that the enzyme proceeds through a hybrid (two-site) random Ter Ter mechanism, one that likely involves a two-step reaction at the biotin carboxylase domain. Citrate, a known activator of animal forms of ACC, activates both by increasing k(cat) and k(cat)/K(M) for ATP and acetyl-CoA.
- Published
- 2009
- Full Text
- View/download PDF
22. Synthesis and characterization of a BODIPY-labeled derivative of Soraphen A that binds to acetyl-CoA carboxylase.
- Author
-
Raymer B, Kavana M, Price A, Wang B, Corcoran L, Kulathila R, Groarke J, and Mann T
- Subjects
- Acetyl-CoA Carboxylase metabolism, Binding Sites, Boron Compounds chemical synthesis, Crystallography, X-Ray, Macrolides chemical synthesis, Protein Structure, Tertiary, Acetyl-CoA Carboxylase antagonists & inhibitors, Boron Compounds chemistry, Macrolides chemistry
- Abstract
BODIPY-labeled Soraphen A derivative 4 was synthesized and characterized as an acetyl-CoA carboxylase (ACC) binder. Biophysical measurements indicate that the molecule binds in the biotin carboxylase domain where Soraphen A has been shown to bind. The fluorescent label of the BODIPY can be used to biophysically identify a compound that binds to the Soraphen A site of the biotin carboxylase domain versus the carboxytransferase domain of ACC.
- Published
- 2009
- Full Text
- View/download PDF
23. Resveratrol inhibits firefly luciferase.
- Author
-
Bakhtiarova A, Taslimi P, Elliman SJ, Kosinski PA, Hubbard B, Kavana M, and Kemp DM
- Subjects
- Animals, Cell Line, Genes, Reporter, Luciferases, Firefly genetics, Luciferases, Firefly metabolism, Mice, Resveratrol, Antioxidants pharmacology, Luciferases, Firefly antagonists & inhibitors, Stilbenes pharmacology
- Abstract
The potential therapeutic value of resveratrol in age-related disease settings including cancer, diabetes, and Alzheimer's has emerged from a rapidly growing body of experimental evidence. Protection from oxidative stress appears to be a common feature of resveratrol that may be mediated through SirT1, though more specific molecular mechanisms by which resveratrol mediates its effects remain unclear. This has prompted an upsurge in cell-based mechanistic studies, often incorporating reporter assays for pathway elucidation in response to resveratrol treatment. Here, we report that resveratrol potently inhibits firefly luciferase with a K(i) value of 2microM, and caution that this confounding element may lead to compromised data interpretation.
- Published
- 2006
- Full Text
- View/download PDF
24. Spectroscopic and electronic structure studies of the role of active site interactions in the decarboxylation reaction of alpha-keto acid-dependent dioxygenases.
- Author
-
Neidig ML, Brown CD, Kavana M, Choroba OW, Spencer JB, Moran GR, and Solomon EI
- Subjects
- 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide) chemistry, Binding Sites, Circular Dichroism, Kinetics, Models, Molecular, Protein Conformation, Substrate Specificity, 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide) metabolism, Carboxylic Acids chemistry
- Abstract
The alpha-ketoglutate (alpha-KG)-dependent dioxygenases are a large class of mononuclear non-heme iron enzymes that require Fe(II), alpha-KG and dioxygen for catalysis, with the alpha-KG cosubstrate supplying the two additional electrons required for dioxygen activation. A sub-class of these enzymes exists in which the alpha-keto acid is covalently attached to the substrate, including (4-hydroxy)mandelate synthase (HmaS) and (4-hydroxyphenyl)pyruvate dioxygenase (HPPD) which utilize the same substrate but exhibit two different general reactivities (H-atom abstraction and electrophilic attack). Previous kinetic studies of Streptomyces avermitilis HPPD have shown that the substrate analog phenylpyruvate (PPA), which only differs from the normal substrate (4-hydroxyphenyl)pyruvate (HPP) by the absence of a para-hydroxyl group on the aromatic ring, does not induce a reaction with dioxygen. While an Fe(IV)O intermediate is proposed to be the reactive species in converting substrate to product, the key step utilizing O(2) to generate this species is the decarboxylation of the alpha-keto acid. It has been generally proposed that the two requirements for decarboxylation are bidentate coordination of the alpha-keto acid to Fe(II) and the presence of a 5C Fe(II) site for the O(2) reaction. Circular dichroism and magnetic circular dichroism studies have been performed and indicate that both enzyme complexes with PPA are similar with bidentate alpha-KG coordination and a 5C Fe(II) site. However, kinetic studies indicate that while HmaS reacts with PPA in a coupled reaction similar to the reaction with HPP, HPPD reacts with PPA in an uncoupled reaction at an approximately 10(5)-fold decreased rate compared to the reaction with HPP. A key difference is spectroscopically observed in the n-->pi( *) transition of the HPPD/Fe(II)/PPA complex which, based upon correlation to density functional theory calculations, is suggested to result from H-bonding between a nearby residue and the carboxylate group of the alpha-keto acid. Such an interaction would disfavor the decarboxylation reaction by stabilizing electron density on the carboxylate group such that the oxidative cleavage to yield CO(2) is disfavored.
- Published
- 2006
- Full Text
- View/download PDF
25. Spectroscopic and electronic structure studies of aromatic electrophilic attack and hydrogen-atom abstraction by non-heme iron enzymes.
- Author
-
Neidig ML, Decker A, Choroba OW, Huang F, Kavana M, Moran GR, Spencer JB, and Solomon EI
- Subjects
- 4-Hydroxyphenylpyruvate Dioxygenase chemistry, Circular Dichroism, Computational Biology, Spectrum Analysis, Thermodynamics, Hydrogen chemistry, Nonheme Iron Proteins chemistry
- Abstract
(4-Hydroxy)mandelate synthase (HmaS) and (4-hydroxyphenyl)pyruvate dioxygenase (HPPD) are two alpha-keto acid dependent mononuclear non-heme iron enzymes that use the same substrate, (4-hydroxyphenyl)pyruvate, but exhibit two different general reactivities. HmaS performs hydrogen-atom abstraction to yield benzylic hydroxylated product (S)-(4-hydroxy)mandelate, whereas HPPD utilizes an electrophilic attack mechanism that results in aromatic hydroxylated product homogentisate. These enzymes provide a unique opportunity to directly evaluate the similarities and differences in the reaction pathways used for these two reactivities. An Fe(II) methodology using CD, magnetic CD, and variable-temperature, variable-field magnetic CD spectroscopies was applied to HmaS and compared with that for HPPD to evaluate the factors that affect substrate interactions at the active site and to correlate these to the different reactivities exhibited by HmaS and HPPD to the same substrate. Combined with density functional theory calculations, we found that HmaS and HPPD have similar substrate-bound complexes and that the role of the protein pocket in determining the different reactivities exhibited by these enzymes (hydrogen-atom abstraction vs. aromatic electrophilic attack) is to properly orient the substrate, allowing for ligand field geometric changes along the reaction coordinate. Elongation of the Fe(IV) O bond in the transition state leads to dominant Fe(III) O(*-) character, which significantly contributes to the reactivity with either the aromatic pi-system or the C H sigma-bond.
- Published
- 2006
- Full Text
- View/download PDF
26. Spectroscopic and computational studies of NTBC bound to the non-heme iron enzyme (4-hydroxyphenyl)pyruvate dioxygenase: active site contributions to drug inhibition.
- Author
-
Neidig ML, Decker A, Kavana M, Moran GR, and Solomon EI
- Subjects
- 4-Hydroxyphenylpyruvate Dioxygenase metabolism, Binding Sites drug effects, Binding Sites physiology, Catalytic Domain, Circular Dichroism, Computational Biology methods, Cyclohexanones metabolism, Enzyme Inhibitors chemistry, Enzyme Inhibitors metabolism, Iron metabolism, Streptomyces enzymology, Thermodynamics, 4-Hydroxyphenylpyruvate Dioxygenase antagonists & inhibitors, 4-Hydroxyphenylpyruvate Dioxygenase chemistry, Cyclohexanones chemistry, Iron chemistry
- Abstract
(4-Hydroxyphenyl)pyruvate dioxygenase (HPPD) is an alpha-keto-acid-dependent dioxygenase which catalyzes the conversion of (4-hydroxyphenyl)pyruvate (HPP) to homogentisate as part of tyrosine catabolism. While several di- and tri-ketone alkaloids are known as inhibitors of HPPD and used commercially as herbicides, one such inhibitor, [2-nitro-4-(trifluoromethyl)benzoyl]-1,3-cyclohexanedione (NTBC), has also been used therapeutically to treat type I tyrosinemia and alkaptonuria in humans. To gain further insight into the mechanism of inhibition by NTBC, a combination of CD/MCD spectroscopy and DFT calculations of HPPD/Fe(II)/NTBC has been performed to evaluate the contribution of the Fe(II)-NTBC bonding interaction to the high affinity of this drug for the enzyme. The results indicate that the bonding of NTBC to Fe(II) is very similar to that for HPP, both involving similar pi-backbonding interactions between NTBC/HPP and Fe(II). Combined with the result that the calculated binding energy of NTBC is, in fact, approximately 3 kcal/mol less than that for HPP, the bidentate coordination of NTBC to Fe(II) is not solely responsible for its extremely high affinity for the enzyme. Thus, the pi-stacking interactions between the aromatic rings of NTBC and two phenyalanine residues, as observed in the crystallography of the HPPD/Fe(II)/NTBC complex, appear to be responsible for the observed high affinity of drug binding.
- Published
- 2005
- Full Text
- View/download PDF
27. Accumulation of multiple intermediates in the catalytic cycle of (4-hydroxyphenyl)pyruvate dioxygenase from Streptomyces avermitilis.
- Author
-
Johnson-Winters K, Purpero VM, Kavana M, and Moran GR
- Subjects
- 3,4-Dihydroxyphenylacetic Acid chemistry, 3,4-Dihydroxyphenylacetic Acid metabolism, Catalysis, Chromatography, High Pressure Liquid, Deuterium Exchange Measurement, Ferrous Compounds chemistry, Ferrous Compounds metabolism, Homogentisic Acid chemistry, Homogentisic Acid metabolism, Kinetics, Spectrophotometry, Substrate Specificity, 4-Hydroxyphenylpyruvate Dioxygenase chemistry, 4-Hydroxyphenylpyruvate Dioxygenase metabolism, Streptomyces enzymology
- Abstract
(4-Hydroxyphenyl)pyruvate dioxygenase (HPPD) catalyzes the conversion of (4-hydroxyphenyl)pyruvate (HPP) to homogentisate (HG). This reaction involves decarboxylation, substituent migration, and aromatic oxygenation in a single catalytic cycle. HPPD is a unique member of the alpha-keto acid dependent oxygenases that require Fe(II) and an alpha-keto acid substrate to oxygenate or oxidize an organic molecule. We have examined the reaction coordinate of HPPD from Streptomyces avermitilis using rapid mixing pre-steady-state methods in conjunction with steady-state kinetic analyses. Acid quench reactions and product analysis of homogentisate indicate that HPPD as isolated is fully active and that experiments limited in dioxygen concentration with respect to that of the enzyme do involve a single turnover. These experiments indicate that during the course of one turnover the concentration of homogentisate is stoichiometric with enzyme concentration by approximately 200 ms, well before the completion of the catalytic cycle. Subsequent single turnover reactions were monitored spectrophotometrically under pseudo-first-order and matched concentration reactant conditions. Three spectrophotometrically distinct intermediates are observed to accumulate. The first of these is a relatively strongly absorbing species with maxima at 380 and 480 nm that forms with a rate constant (k(1)) of 7.4 x 10(4) M(-)(1) s(-)(1) and then decays to a second intermediate with a rate constant (k(2)) of 74 s(-)(1). The rate constant for the decay of the second intermediate (k(3)) is 13 s(-)(1) and is concomitant with the formation of the product, homogentisate, based on rapid quench and pre-steady-state fluorescence measurements. The rate constant for this process decreases to 7.6 s(-)(1) when deuterons are substituted for protons in the aromatic ring of the substrate. The release of product from the enzyme is rate limiting and occurs at 1.6 s(-)(1). This final event exhibits a kinetic isotope effect of 2 with deuterium oxide as the solvent, consistent with a solvent isotope effect on V(max) of 2.6 observed in steady-state experiments.
- Published
- 2005
- Full Text
- View/download PDF
28. CD and MCD studies of the non-heme ferrous active site in (4-hydroxyphenyl)pyruvate dioxygenase: correlation between oxygen activation in the extradiol and alpha-KG-dependent dioxygenases.
- Author
-
Neidig ML, Kavana M, Moran GR, and Solomon EI
- Subjects
- 4-Hydroxyphenylpyruvate Dioxygenase metabolism, Binding Sites, Circular Dichroism methods, Ferrous Compounds metabolism, Ketoglutaric Acids metabolism, Oxygen metabolism, 4-Hydroxyphenylpyruvate Dioxygenase chemistry, Ferrous Compounds chemistry, Ketoglutaric Acids chemistry, Oxygen chemistry
- Abstract
(4-Hydroxyphenyl)pyruvate dioxygenase (HPPD) is an unusual alpha-keto acid-dependent non-heme iron dioxygenase as it incorporates both atoms of dioxygen into a single substrate, paralleling the extradiol dioxygenases. CD/MCD studies of the catalytically active ferrous site and its interaction with substrate reveal a geometic and electronic structure and mechanistic approach to oxygen activation which bridges those of the alpha-KG-dependent and the extradiol dioxygenases.
- Published
- 2004
- Full Text
- View/download PDF
29. Interaction of (4-hydroxyphenyl)pyruvate dioxygenase with the specific inhibitor 2-[2-nitro-4-(trifluoromethyl)benzoyl]-1,3-cyclohexanedione.
- Author
-
Kavana M and Moran GR
- Subjects
- 4-Hydroxyphenylpyruvate Dioxygenase chemistry, Cyclohexanones chemistry, Electron Spin Resonance Spectroscopy, Enzyme Inhibitors chemistry, Enzyme Inhibitors metabolism, Enzyme Inhibitors pharmacology, Enzyme Stability, Ferrous Compounds chemistry, Holoenzymes chemistry, Holoenzymes metabolism, Kinetics, Magnetic Resonance Spectroscopy methods, Nitric Oxide chemistry, Nitrobenzoates chemistry, Oxidation-Reduction, Oxygen chemistry, Protein Binding, Spectrophotometry, Ultraviolet, Streptomyces enzymology, 4-Hydroxyphenylpyruvate Dioxygenase antagonists & inhibitors, 4-Hydroxyphenylpyruvate Dioxygenase metabolism, Cyclohexanones metabolism, Cyclohexanones pharmacology, Nitrobenzoates metabolism, Nitrobenzoates pharmacology
- Abstract
(4-Hydroxyphenyl)pyruvate dioxygenase (HPPD) is a non-heme Fe(II) enzyme that catalyzes the conversion of (4-hydroxyphenyl)pyruvate (HPP) to homogentisate as part of the tyrosine catabolism pathway. Inhibition of HPPD by the triketone 2-[2-nitro-4-(trifluoromethyl)benzoyl]-1,3-cyclohexanedione (NTBC) is used to treat type I tyrosinemia, a rare but fatal defect in tyrosine catabolism. Although triketones have been used for many years as HPPD inhibitors for both medical and herbicidal purposes, the mechanism of inhibition is not well understood. The following work provides mechanistic insight into NTBC binding. The tautomeric population of NTBC in aqueous solution is dominated by a single enol as determined by NMR spectroscopy. NTBC preferentially binds to the complex of HPPD and FeII [HPPD.Fe(II)] as evidenced by a visible absorbance feature centered at 450 nm. The binding of NTBC to HPPD.Fe(II) was observed using a rapid mixing method and was shown to occur in two phases and comprise three steps. A hyperbolic dependence of the first observable process with NTBC concentration indicates a pre-equilibrium binding step followed by a limiting rate (K(1) = 1.25 +/- 0.08 mM, k(2) = 8.2 +/- 0.2 s(-1)), while the second phase (k(3) = 0.76 +/- 0.02 s(-1)) had no dependence on NTBC concentration. Neither K(1),k(2), nor k(3) was influenced by pH in the range of 6.0-8.0. Isotope effects on both k(2) and k(3) were observed when D(2)O is used as the solvent (for k(2), k(h)/k(d) = 1.3; for k(3), k(h)/k(d) = 3.2). It is therefore proposed that the bidentate association of NTBC with the active site metal ion (k(2)) precedes the Lewis acid-assisted conversion of the bound enol to the enolate (k(3)). Although the native enzyme without substrate reacts with molecular oxygen to form the oxidized holoenzyme, the HPPD.Fe(II).NTBC complex does not. When the complex is exposed to atmospheric oxygen, the absorbance feature associated with NTBC binding does not diminish over the course of 2 days. This means not only that the HPPD.Fe(II).NTBC complex does not oxidize but also that the dissociation rate constant for NTBC is essentially zero because any HPPD.Fe(II) that formed would readily oxidize in the presence of dioxygen. Consistent with this observation, EPR spectroscopy has shown that only 2% of the HPPD.Fe(II).NTBC complex forms an NO complex as compared to the holoenzyme.
- Published
- 2003
- Full Text
- View/download PDF
30. (4-Hydroxyphenyl)pyruvate dioxygenase from Streptomyces avermitilis: the basis for ordered substrate addition.
- Author
-
Johnson-Winters K, Purpero VM, Kavana M, Nelson T, and Moran GR
- Subjects
- 4-Hydroxyphenylpyruvate Dioxygenase genetics, 4-Hydroxyphenylpyruvate Dioxygenase isolation & purification, Cloning, Molecular, Ferrous Compounds chemistry, Gene Expression Regulation, Bacterial, Ketoglutaric Acids chemistry, Kinetics, Oxidation-Reduction, Oxygen chemistry, Phenylpyruvic Acids chemistry, Pyruvic Acid chemistry, Recombinant Proteins biosynthesis, Recombinant Proteins chemistry, Recombinant Proteins isolation & purification, Streptomyces genetics, Substrate Specificity genetics, Triazines chemistry, 4-Hydroxyphenylpyruvate Dioxygenase chemistry, Streptomyces enzymology
- Abstract
(4-hydroxyphenyl)pyruvate dioxygenase (HPPD) catalyzes the second step in the pathway for the catabolism of tyrosine, the conversion of (4-hydroxyphenyl)pyruvate (HPP) to homogentisate (HG). This reaction involves decarboxylation, substituent migration, and aromatic oxygenation. HPPD is a member of the alpha-keto acid dependent oxygenases that require Fe(II) and an alpha-keto acid substrate to oxygenate an organic molecule. We have examined the binding of ligands to HPPD from Streptomyces avermitilis. Our data show that HPP binds to the apoenzyme and that the apo-HPPD.HPP complex does not bind Fe(II) to generate active holoenzyme. The binding of HPP, phenylpyruvate (PPA), and pyruvate to the holoenzyme produces a weak ligand charge-transfer band at approximately 500 nm that is indicative of bidentate binding of the 1-carboxylate and 2-keto pyruvate oxygen atoms to the active site metal ion. For HPPD from this organism the 4-hydroxyl group of (4-hydroxyphenyl)pyruvate is a requirement for catalysis; no turnover is observed in the presence of phenylpyruvate. The rate constant for the dissociation of Fe(II) from the holoenzyme is 0.0006 s(-)(1) and indicates that this phenomenon is not significantly relevant in steady-state turnover. The addition of HPP and molecular oxygen to the holoenzyme is formally random. The basis of the ordered bi bi steady-state kinetic mechanism previously observed by Rundgren (Rundgren, M. (1977) J. Biol. Chem. 252, 5094-9) is the 3600-fold increase in oxygen reactivity when holo-HPPD is in complex with HPP. This complex reacts with molecular oxygen with a second-order rate constant of 1.4 x 10(5) M(-)(1) s(-)(1) inducing the formation of an intermediate that decays at the catalytically relevant rate of 7.8 s(-)(1).
- Published
- 2003
- Full Text
- View/download PDF
31. Hydrogen transfer to carbonyls and imines from a hydroxycyclopentadienyl ruthenium hydride: evidence for concerted hydride and proton transfer.
- Author
-
Casey CP, Singer SW, Powell DR, Hayashi RK, and Kavana M
- Subjects
- Kinetics, Protons, Aldehydes chemistry, Hydrogen chemistry, Imines chemistry, Ketones chemistry, Ruthenium Compounds chemistry
- Abstract
Reaction of ([2,5-Ph(2)-3,4-Tol(2)(eta(5)-C(4)CO)](2)H)Ru(2)(CO)(4)(mu-H) (6) with H(2) formed [2,5-Ph(2)-3,4-Tol(2)(eta(5)-C(4)COH)Ru(CO)(2)H] (8), the active species in catalytic carbonyl reductions developed by Shvo. Kinetic studies of the reduction of PhCHO by 8 in THF at -10 degrees C showed second-order kinetics with Delta H(double dagger) = 12.0 kcal mol(-1) and Delta S(double dagger) = -28 eu. The rate of reduction was not accelerated by CF(3)CO(2)H, and was not inhibited by CO. Selective deuteration of the RuH and OH positions in 8 gave individual kinetic isotope effects k(RuH)/k(RuD) = 1.5 +/- 0.2 and k(OH)/k(OD) = 2.2 +/- 0.1 for PhCHO reduction at 0 degrees C. Simultaneous deuteration of both positions in 8 gave a combined kinetic isotope effect of k(OHRuH)/k(ODRuD) = 3.6 +/- 0.3. [2,5-Ph(2)-3,4-Tol(2)(eta(5)-C(4)COSiEt(3))Ru(CO)(2)H] (12) and NEt(4)(+)[2,5-Ph(2)-3,4-Tol(2)(eta(4)-C(4)CO)Ru(CO)(2)H](-) (13) were unreactive toward PhCHO under conditions where facile PhCHO reduction by 8 occurred. PhCOMe was reduced by 8 30 times slower than PhCHO; MeN=CHPh was reduced by 8 26 times faster than PhCHO. Cyclohexene was reduced to cyclohexane by 8 at 80 degrees C only in the presence of H(2.) Concerted transfer of a proton from OH and hydride from Ru of 8 to carbonyls and imines is proposed.
- Published
- 2001
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.