24 results on '"Jean M. Severin"'
Search Results
2. Magnetic Resonance Imaging Detection and Time Course of Cerebral Microhemorrhages during Passive Immunotherapy in Living Amyloid Precursor Protein Transgenic Mice
- Author
-
Gerard B. Fox, Mark Day, Feng Luo, Ulrich Ebert, Vincent Hradil, Terese Seifert, Bryan F. Cox, Beate Roesner, Jean M. Severin, Heinz Hillen, Daniel A. Llano, and Nathan R. Rustay
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Time Factors ,medicine.drug_class ,Mice, Transgenic ,Monoclonal antibody ,Amyloid beta-Protein Precursor ,Mice ,Alzheimer Disease ,In vivo ,Amyloid precursor protein ,medicine ,Animals ,Humans ,Longitudinal Studies ,Cerebral Hemorrhage ,Cerebral Cortex ,Pharmacology ,Amyloid beta-Peptides ,biology ,medicine.diagnostic_test ,business.industry ,Magnetic resonance microscopy ,Immunization, Passive ,Antibodies, Monoclonal ,Magnetic resonance imaging ,Magnetic Resonance Imaging ,Mice, Inbred C57BL ,Disease Models, Animal ,Hemosiderin ,biology.protein ,Molecular Medicine ,Biomarker (medicine) ,Female ,Antibody ,business - Abstract
In recent years immunotherapy-based approaches for treating Alzheimer's disease have become the subject of intensive research. However, an important mechanistic-related safety concern is exacerbation of the risk of microhemorrhage that may be associated with fast removal of amyloid-β (Aβ) deposits found in blood vessels or brain parenchyma. Rapid in vivo detection of microhemorrhages in living amyloid precursor protein transgenic mice has not been described, and histological analysis can take several months before this risk is assessed. Aged transgenic mice were divided into two groups that would undergo longitudinal passive immunotherapy for 12 or 18 weeks. 6G1, a nonselective anti-Aβ monoclonal antibody, and 8F5, a more selective antioligomeric Aβ monoclonal antibody, were examined in both longitudinal studies. High-resolution T2*-weighted magnetic resonance microscopy (100 × 100 × 400 μm) was used for microhemorrhage detection in vivo. Cerebral microhemorrhages by magnetic resonance imaging were compared with histological hemosiderin staining in each animal; results showed that T2*-weighted magnetic resonance microscopy can reliably detect microhemorrhages of ≥60 μm in diameter at baseline and after 12 to 18 weeks of treatment in the same animals in vivo. This correlated significantly with histological readings. This new imaging safety biomarker can be readily applied to preclinical antibody screening in a longitudinal manner. 6G1 and 8F5, however, both increased microhemorrhage incidence in aged amyloid precursor protein transgenic mice compared with their baseline and vehicle treatment. A highly selective antibody for soluble Aβ is needed to address the question of whether antibodies that do not bind to deposited Aβ have microhemorrhage liability.
- Published
- 2010
- Full Text
- View/download PDF
3. Variants in Apaf-1 segregating with major depression promote apoptosome function
- Author
-
D A Egan, P Metzger, Jennifer Potter, Yuan Chen, S Dorwin, M Gopalakrishnan, J P Sullivan, Chris Neff, A Hahn, M Lake, Donna Shattuck, Karl A. Walter, R Mueller, Victor Abkevich, T Olsen, Audrey Beck, T. F. Holzman, B B Spear, Mark H. Skolnick, Jean M. Severin, J.E. Harlan, R W Johnson, C Davenport, E. Gubbins, K Voelp, David A. Katz, J-M Roch, U.S. Ladror, and D N Halbert
- Subjects
Caspase-9 ,Genetics ,Depressive Disorder, Major ,Polymorphism, Genetic ,biology ,Intracellular Signaling Peptides and Proteins ,Proteins ,Apoptosis ,Phenotype ,Cellular and Molecular Neuroscience ,Psychiatry and Mental health ,Apoptotic Protease-Activating Factor 1 ,biology.protein ,Humans ,Genetic Predisposition to Disease ,Apoptosome ,APAF1 ,Heterologous expression ,Allele ,Molecular Biology ,Gene ,Alleles ,Chromosome 12 - Abstract
APAF1, encoding the protein apoptosis protease activating factor 1 (Apaf-1), has recently been established as a chromosome 12 gene conferring predisposition to major depression in humans. The molecular phenotypes of Apaf-1 variants were determined by in vitro reconstruction of the apoptosome complex in which Apaf-1 activates caspase 9 and thus initiates a cascade of proteolytic events leading to apoptotic destruction of the cell. Cellular phenotypes were measured using a yeast heterologous expression assay in which human Apaf-1 and other proteins necessary to constitute a functional apoptotic pathway were overexpressed. Apaf-1 variants encoded by APAF1 alleles that segregate with major depression in families linked to chromosome 12 shared a common gain-of-function phenotype in both assay systems. In contrast, other Apaf-1 variants showed neutral or loss-of-function phenotypes. The depression-associated alleles thus have a common phenotype that is distinct from that of non-associated variants. This result suggests an etiologic role for enhanced apoptosis in major depression.
- Published
- 2005
- Full Text
- View/download PDF
4. Preparation and characterization of nitrilotriacetic-acid-terminated self-assembled monolayers on gold surfaces for matrix-assisted laser desorption ionization-time of flight-mass spectrometry analysis of proteins and peptides
- Author
-
Andrew Vogt, Richard S. Smith, T. F. Holzman, Jianwei Shen, Robert E. Johnson, Jieyi Wang, John E. Harlan, Tanveer F. Ahmed, Sally Dorwin, and Jean M. Severin
- Subjects
Nitrilotriacetic Acid ,MALDI imaging ,Chromatography ,Protein mass spectrometry ,Surface Properties ,Chemistry ,Biophysics ,Nitrilotriacetic acid ,Proteins ,Self-assembled monolayer ,Cell Biology ,Mass spectrometry ,Biochemistry ,Recombinant Proteins ,Sample preparation in mass spectrometry ,chemistry.chemical_compound ,Matrix-assisted laser desorption/ionization ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,Histidine ,Sample preparation ,Gold ,Peptides ,Molecular Biology - Abstract
On-target affinity capture, enrichment and purification of biomolecules improve detection of specific analytes from complex biological samples in matrix-assisted laser desorption ionization-time of flight-mass spectrometry (MALDI-TOF-MS) analysis. In this paper, we report a simple method for preparation of a self-assembled nitrilotriacetic acid (NTA) monolayer on gold surface which can be used as a MALDI-TOF-MS sample target specifically for recombinant oligohistidine-tagged proteins/peptides and phosphorylated peptides. The NTA functional groups are immobilized to the gold surface via the linkage of 1,8-octanedithiol which forms a self-assembled monolayer on gold. Characterization by X-ray photoelectron spectroscopy and MALDI analysis of the modified surface are described. The chemically modified surface shows strong affinity toward the analytes of interest, which allows effective removal of the common interferences, e.g. salts and detergents, and therefore leads to improved signal/noise ratio and detection limit. The use of the modified surface simplifies the sample preparation for MALDI analysis of these targeted analytes.
- Published
- 2005
- Full Text
- View/download PDF
5. Discovery of Potent Inhibitors of Dihydroneopterin Aldolase Using CrystaLEAD High-Throughput X-ray Crystallographic Screening and Structure-Directed Lead Optimization
- Author
-
Stephen F. Betz, Kevin Ronald Condroski, John E. Harlan, Bruce A. Beutel, Jean M. Severin, Sean M. Merrick, Vicki L. Nienaber, Susan J. Swanson, Rolf Wagner, Vincent S. Stoll, Karl A. Walter, J. Owen McCall, Peter Magdalinos, Claude G. Lerner, William J. Sanders, Geoffrey F. Stamper, Clarissa G. Jakob, and Robert P. Meadows
- Subjects
Models, Molecular ,Guanine ,Databases, Factual ,Molecular model ,Stereochemistry ,Dihydroneopterin aldolase ,Crystallography, X-Ray ,Benzoates ,Neopterin ,Structure-Activity Relationship ,Drug Discovery ,Structure–activity relationship ,Enzyme Inhibitors ,Aldehyde-Lyases ,chemistry.chemical_classification ,Binding Sites ,Molecular Structure ,biology ,Chemistry ,Aldolase A ,Active site ,Triazoles ,Lyase ,Crystallography ,Pyrimidines ,Enzyme ,Purines ,Enzyme inhibitor ,biology.protein ,Molecular Medicine - Abstract
Potent inhibitors of 7,8-dihydroneopterin aldolase (DHNA; EC 4.1.2.25) have been discovered using CrystaLEAD X-ray crystallographic high-throughput screening followed by structure-directed optimization. Screening of a 10 000 compound random library provided several low affinity leads and their corresponding X-ray crystal structures bound to the enzyme. The presence of a common structural feature in each of the leads suggested a strategy for the construction of a directed library of approximately 1000 compounds that were screened for inhibitory activity in a traditional enzyme assay. Several lead compounds with IC(50) values of about 1 microM against DHNA were identified, and crystal structures of their enzyme-bound complexes were obtained by cocrystallization. Structure-directed optimization of one of the leads thus identified afforded potent inhibitors with submicromolar IC(50) values.
- Published
- 2004
- Full Text
- View/download PDF
6. Design of Adenosine Kinase Inhibitors from the NMR-Based Screening of Fragments
- Author
-
Erol K. Bayburt, Andrew F. Stewart, Stanley Didomenico, Stephen W. Fesik, Thomas F. Holzman, Marlon D. Cowart, Larry Solomon, Michael F. Jarvis, Philip J. Hajduk, Jean M. Severin, Karl A. Walter, Elizabeth A. Kowaluk, Arthur Gomtsyan, Steve McGaraughty, and Richard D. Smith
- Subjects
Male ,Models, Molecular ,Magnetic Resonance Spectroscopy ,Databases, Factual ,Adenosine kinase ,Ligands ,Chemical synthesis ,Cell Line ,Rats, Sprague-Dawley ,In vivo ,Drug Discovery ,Animals ,Enzyme Inhibitors ,Fragmentation (cell biology) ,Adenosine Kinase ,chemistry.chemical_classification ,Analgesics ,biology ,Bicyclic molecule ,Chemistry ,In vitro ,Rats ,Enzyme ,Biochemistry ,Enzyme inhibitor ,biology.protein ,Molecular Medicine ,Protein Binding - Abstract
A strategy is described for designing high-affinity ligands using information derived from the NMR-based screening of fragments. The method involves the fragmentation of an existing lead molecule, identification of suitable replacements for the fragments, and incorporation of the newly identified fragments into the original scaffold. Using this technique, novel substituents were rapidly identified and incorporated into lead inhibitors of adenosine kinase that exhibited potent in vitro and in vivo activities. This approach is a valuable strategy for modifying existing leads to improve their potency, bioavailability, or toxicity profile and thus represents a useful technique for lead optimization.
- Published
- 2000
- Full Text
- View/download PDF
7. The Structure of VanX Reveals a Novel Amino-Dipeptidase Involved in Mediating Transposon-Based Vancomycin Resistance
- Author
-
Leonard Katz, Dirksen E. Bussiere, Steven D. Pratt, Chang H. Park, Jean M. Severin, and Thomas F. Holzman
- Subjects
Models, Molecular ,Dipeptidase ,Transposable element ,Dipeptidases ,animal structures ,Protein Conformation ,Enterococcus faecium ,Molecular Sequence Data ,High resolution ,Carboxypeptidases ,Phosphinate ,Crystallography, X-Ray ,Mice ,Structure-Activity Relationship ,Organophosphorus Compounds ,Bacterial Proteins ,Vancomycin ,Hydrolase ,Animals ,Hedgehog Proteins ,Amino Acid Sequence ,Enzyme Inhibitors ,Molecular Biology ,Vancomycin resistance ,Alanine ,Binding Sites ,Sequence Homology, Amino Acid ,biology ,Proteins ,Active site ,Drug Resistance, Microbial ,Dipeptides ,Cell Biology ,Serine-Type D-Ala-D-Ala Carboxypeptidase ,Carboxypeptidase ,Biochemistry ,DNA Transposable Elements ,Trans-Activators ,biology.protein ,bacteria ,Propionates ,Sequence Alignment ,Copper - Abstract
VanX is a zinc-dependent D-alanyl-D-alanine dipeptidase that is a critical component in a system that mediates transposon-based vancomycin resistance in enterococci. It is also a key drug target in circumventing clinical vancomycin resistance. The structure of VanX from E. faecium has been solved by X-ray crystallography and reveals a Zn 2+ -dipeptidase with a unique overall fold and a well-defined active site confined within a cavity of limited size. The crystal structures of VanX, the VanX:D-alanyl-D-alanine complex, the VanX:D-alanine complex, and VanX in complex with phosphonate and phosphinate transition-state analog inhibitors, are also presented at high resolution. Structural homology searches of known structures revealed that the fold of VanX is similar to those of two proteins: the N-terminal fragment of murine Sonic hedgehog and the Zn 2+ -dependent N-acyl-D-alanyl-D-alanine carboxypeptidase of S. albus G .
- Published
- 1998
- Full Text
- View/download PDF
8. Discovery of Potent Nonpeptide Inhibitors of Stromelysin Using SAR by NMR
- Author
-
E. Gubbins, D. W. Morgan, Edward T. Olejniczak, Jean M. Severin, S. K. Davidsen, G. Sheppard, G. M. Carrera, Robert L. Simmer, D. H. Steinman, David G. Nettesheim, R. P. Meadows, T. F. Holzman, and J. B. Summers, H. Smith, P. A. Marcotte, Suzanne B. Shuker, Philip J. Hajduk, Karl A. Walter, and Stephen W. Fesik
- Subjects
chemistry.chemical_classification ,Drug discovery ,Stereochemistry ,Acetohydroxamic acid ,A protein ,General Chemistry ,Matrix metalloproteinase ,Biochemistry ,Combinatorial chemistry ,Chemical synthesis ,Catalysis ,Colloid and Surface Chemistry ,Enzyme ,chemistry ,Yield (chemistry) ,medicine ,IC50 ,medicine.drug - Abstract
With the use of an NMR-based method, potent (IC50 < 25 nM) nonpeptide inhibitors of the matrix metalloproteinase stromelysin (MMP-3) were discovered. The method, called SAR by NMR (for structure−activity relationships by nuclear magnetic resonance), involves the identification, optimization, and linking of compounds that bind to proximal sites on a protein. Using this technique, two ligands that bind weakly to stromelysin (acetohydroxamic acid, KD = 17 mM; 3-(cyanomethyl)-4‘-hydroxybiphenyl, KD = 0.02 mM) were identified. On the basis of NMR-derived structural information, the two fragments were connected to produce a 15 nM inhibitor of this enzyme. This compound was rapidly discovered (less than 6 months) and required only a minimal amount of chemical synthesis. These studies indicate that the SAR by NMR method can be effectively applied to enzymes to yield potent lead inhibitorsan important part of the drug discovery process.
- Published
- 1997
- Full Text
- View/download PDF
9. ChemInform Abstract: Discovery of Potent Nonpeptide Inhibitors of Stromelysin Using SAR by NMR
- Author
-
Philip J. Hajduk, G. M. Jun. Carrera, Stephen W. Fesik, Patrick A. Marcotte, Edward T. Olejniczak, T. F. Holzman, E. Gubbins, Jean M. Severin, Suzanne B. Shuker, R. P. Meadows, Douglas W. Morgan, Karl A. Walter, David G. Nettesheim, H. Smith, D. H. Steinman, S. K. Davidsen, G. S. Sheppard, James B. Summers, and Robert L. Simmer
- Subjects
Chemistry ,General Medicine ,Combinatorial chemistry - Published
- 2010
- Full Text
- View/download PDF
10. High-level expression of recombinant human FK-binding protein from a fusion precursor
- Author
-
Stephen W. Fesik, David A. Egan, Neal S. Burres, Steven D. Pratt, Earl G. Gubbins, Jean M. Severin, Andrew M. Petros, Thomas F. Holzman, Tami J. Pilot-Matias, and Rohinton Edalji
- Subjects
Magnetic Resonance Spectroscopy ,Recombinant Fusion Proteins ,Proteolysis ,Molecular Sequence Data ,Gene Expression ,Hydroxylamine ,Isomerase ,Biology ,Hydroxylamines ,Biochemistry ,Tacrolimus ,law.invention ,Tacrolimus Binding Proteins ,law ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Cloning, Molecular ,Protein Precursors ,Amino Acid Isomerases ,chemistry.chemical_classification ,Molecular mass ,medicine.diagnostic_test ,Hydrolysis ,Binding protein ,Peptidylprolyl Isomerase ,Nucleotidyltransferases ,Fusion protein ,Molecular biology ,Recombinant Proteins ,FKBP ,Enzyme ,chemistry ,Recombinant DNA ,Cattle ,Electrophoresis, Polyacrylamide Gel ,Carrier Proteins - Abstract
The human peptidyl-prolyl isomerase FK-binding protein (FKBP) was cloned as a fusion partner with CMP-KDO synthetase (CKS), and the resultant construct was characterized as an improved high-expression source for FKBP. The CKS-FKBP fusion was expressed as a soluble protein at levels approaching 1 gm/L in Escherichia coli fermentations. The fusion protein was purified to near homogeneity by a one-step ammonium sulfate fractionation of whole cell lysate. After selective cleavage, the fusion precursor produced yields approaching 300 mg of purified FKBP per liter of harvested culture, a approximately 30 to 60-fold increase over that observed for a nonfusion construct. Selective cleavage of the fusion partners was accomplished using either hydroxylamine or specific, limited proteolysis. Once separated from the CKS fusion partner, the FKBP was isolated in a single step by either reversed-phase HPLC or chromatography on Q-Sepharose. For comparison of physical and chemical properties, a nonfusion construct of recombinant human FKBP was expressed in E. coli and isolated. The purified FKBPs exhibited expected SDS-PAGE molecular weights and N-terminal sequences. The proteins had similar proton NMR spectra and binding to [3H]FK-506. The fusion construct, CKS-FKBP, was also found to bind [3H]FK-506. These data indicate that FKBP fused to the C-terminus of CKS folds independently of the fusion partner and suggests the fused FKBP adopts a conformation resembling that of the native protein.
- Published
- 1992
- Full Text
- View/download PDF
11. Non-peptide entry inhibitors of HIV-1 that target the gp41 coiled coil pocket
- Author
-
Steve Anderson, Akhter Molla, Robert J Carrick, Jeffrey R. Huth, Karl A. Walter, Leo W. Barrett, Robert P. Meadows, Kent D. Stewart, Dale J. Kempf, Paul L. Richardson, Clarence J. Maring, Vincent S. Stoll, Edmund D. Matayoshi, Teresa I. Ng, Edward T. Olejniczak, William E. Kohlbrenner, Renaldo Mendoza, Jean M. Severin, Hongmei Mo, Keith F. McDaniel, and Rebecca Hutchinson
- Subjects
Models, Molecular ,Enfuvirtide ,Magnetic Resonance Spectroscopy ,Anti-HIV Agents ,Clinical Biochemistry ,Pharmaceutical Science ,Gp41 ,Crystallography, X-Ray ,Biochemistry ,chemistry.chemical_compound ,Structure-Activity Relationship ,Viral envelope ,HIV Fusion Inhibitors ,Drug Discovery ,medicine ,Humans ,Benzamide ,Molecular Biology ,Coiled coil ,Cell fusion ,Organic Chemistry ,HIV Envelope Protein gp41 ,Peptide Fragments ,Ectodomain ,chemistry ,Benzamides ,Molecular Medicine ,Heteronuclear single quantum coherence spectroscopy ,medicine.drug ,Protein Binding - Abstract
The ectodomain of HIV-1 gp41 mediates the fusion of viral and host cellular membranes. The peptide-based drug Enfuvirtide 1 is precedent that antagonists of this fusion activity may act as anti HIV-agents. Here, NMR screening was used to discover non-peptide leads against this target and resulted in the discovery of a new benzamide 1 series. This series is non-peptide, low molecular weight, and analogs have activity in a cell fusion assay with EC50 values ranging 3–41 μM. Structural work on the gp41/benzamide 1 complex was determined by NMR spectroscopy using a designed model peptide system that mimics an open pocket of the fusogenic form of the protein.
- Published
- 2009
12. Toxicological evaluation of thiol-reactive compounds identified using a la assay to detect reactive molecules by nuclear magnetic resonance
- Author
-
Karl A. Walter, Jeffrey R. Huth, Renaldo Mendoza, Sarah A. Dorwin, Jamey Mack, Uri S. Ladror, Danying Song, Philip J. Hajduk, Jean M. Severin, Diane Bartley, and Candice L. Black-Schaefer
- Subjects
Magnetic Resonance Spectroscopy ,Drug-Related Side Effects and Adverse Reactions ,Aldehyde dehydrogenase ,Toxicology ,Autoantigens ,Superoxide dismutase ,Nuclear magnetic resonance ,Cytochrome P-450 Enzyme Inhibitors ,Humans ,Sulfhydryl Compounds ,chemistry.chemical_classification ,Reactive oxygen species ,biology ,Molecular Structure ,Superoxide Dismutase ,Cytochrome P450 ,General Medicine ,Nuclear magnetic resonance spectroscopy ,Aldehyde Dehydrogenase ,Enzyme ,chemistry ,Biochemistry ,Pharmaceutical Preparations ,Ribonucleoproteins ,Drug Design ,biology.protein ,Thiol ,Microsomes, Liver ,Protein Binding - Abstract
We have recently reported on the development of a La assay to detect reactive molecules by nuclear magnetic resonance (ALARM NMR) to detect reactive false positive hits from high-throughput screening, in which we observed a surprisingly large number of compounds that can oxidize or form covalent adducts with protein thiols groups. In the vast majority of these cases, the covalent interactions are largely nonspecific (e.g., affect many protein targets) and therefore unsuitable for drug development. However, certain thiol-reactive species do appear to inhibit the target of interest in a specific manner. The question then arises as to the potential toxicology risks of developing a drug that can react with protein thiol groups. Here, we report on the evaluation of a large set of ALARM-reactive and -nonreactive compounds against a panel of additional proteins (aldehyde dehydrogenase, superoxide dismutase, and three cytochrome P450 enzymes). It was observed that ALARM-reactive compounds have significantly increased risks of interacting with one or more of these enzymes in vitro. Thus, ALARM NMR seems to be a sensitive tool to rapidly identify compounds with an enhanced risk of producing side effects in humans, including alcohol intolerance, the formation of reactive oxygen species, and drug-drug interactions. In conjunction with other toxicology assays, ALARM NMR should be a valuable tool for prioritizing compounds for lead optimization and animal testing.
- Published
- 2007
13. Discovery and design of novel HSP90 inhibitors using multiple fragment-based design strategies
- Author
-
Stephen W. Fesik, Sarah A. Dorwin, Diane Bartley, Karl A. Walter, Steven W. Elmore, Uri S. Ladror, Jun Chen, Jamey Mack, Sha Jin, Kunzer Aaron R, Russell A. Judge, Paul L. Richardson, Christopher L. Lynch, Philip J. Hajduk, Jean M. Severin, Chang Park, Stephen K. Tahir, Michael D. Wendt, Edward D. Matayoshi, Andrew M. Petros, Jeffrey R. Huth, Wang Xilu, and Kerry M. Swift
- Subjects
Models, Molecular ,Protein Conformation ,Molecular Sequence Data ,Aminopyridines ,Crystallography, X-Ray ,Ligands ,Biochemistry ,Protein structure ,Heat shock protein ,Drug Discovery ,Humans ,HSP90 Heat-Shock Proteins ,Enzyme Inhibitors ,Nuclear Magnetic Resonance, Biomolecular ,Malignant phenotype ,Pharmacology ,chemistry.chemical_classification ,biology ,Molecular Structure ,Organic Chemistry ,Active site ,Hsp90 ,Peptide Fragments ,Enzyme ,chemistry ,Chaperone (protein) ,Drug Design ,biology.protein ,Molecular Medicine - Abstract
The molecular chaperone HSP90 has been shown to facilitate cancer cell survival by stabilizing key proteins responsible for a malignant phenotype. We report here the results of parallel fragment-based drug design approaches in the design of novel HSP90 inhibitors. Initial aminopyrimidine leads were elaborated using high-throughput organic synthesis to yield nanomolar inhibitors of the enzyme. Second site leads were also identified which bound to HSP90 in two distinct conformations, an 'open' and 'closed' form. Intriguingly, linked fragment approaches targeting both of these conformations were successful in producing novel, micromolar inhibitors. Overall, this study shows that, with only a few fragment hits, multiple lead series can be generated for HSP90 due to the inherent flexibility of the active site. Thus, ample opportunities exist to use these lead series in the development of clinically useful HSP90 inhibitors for the treatment of cancers.
- Published
- 2007
14. Crystal structures of human adenosine kinase inhibitor complexes reveal two distinct binding modes
- Author
-
Clarissa L. Jakob, Chih-Hung Lee, Haixia Yu, Elizabeth A. Kowaluk, Shripad S. Bhagwat, Richard A. Smith, Mark A. Matulenko, Jean M. Severin, Andrew O. Stewart, Michael F. Jarvis, Arthur Gomtsyan, Marlon D. Cowart, and Steven W. Muchmore
- Subjects
Adenosine monophosphate ,Models, Molecular ,Stereochemistry ,Protein Conformation ,Morpholines ,Adenosine kinase ,Crystallography, X-Ray ,Tubercidin ,chemistry.chemical_compound ,Drug Discovery ,medicine ,Animals ,Humans ,Binding site ,Enzyme Inhibitors ,Adenosine Kinase ,chemistry.chemical_classification ,Antiinfective agent ,Binding Sites ,biology ,Molecular Structure ,Adenosine ,Enzyme ,Pyrimidines ,chemistry ,Cyclic nucleotide-binding domain ,biology.protein ,Molecular Medicine ,Adenosine triphosphate ,Toxoplasma ,medicine.drug ,Protein Binding - Abstract
Adenosine kinase (AK) is an enzyme responsible for converting endogenous adenosine (ADO) to adenosine monophosphate (AMP) in an adenosine triphosphate- (ATP-) dependent manner. The structure of AK consists of two domains, the first a large alpha/beta Rossmann-like nucleotide binding domain that forms the ATP binding site, and a smaller mixed alpha/beta domain, which, in combination with the larger domain, forms the ADO binding site and the site of phosphoryl transfer. AK inhibitors have been under investigation as antinociceptive, antiinflammatory, and anticonvulsant as well as antiinfective agents. In this work, we report the structures of AK in complex with two classes of inhibitors: the first, ADO-like, and the second, a novel alkynylpyrimidine series. The two classes of structures, which contain structurally similar substituents, reveal distinct binding modes in which the AK structure accommodates the inhibitor classes by a 30 degrees rotation of the small domain relative to the large domain. This change in binding mode stabilizes an open and a closed intermediate structural state and provide structural insight into the transition required for catalysis. This results in a significant rearrangement of both the protein active site and the orientation of the alkynylpyrimidine ligand when compared to the observed orientation of nucleosidic inhibitors or substrates.
- Published
- 2006
15. Design and characterization of an engineered gp41 protein from human immunodeficiency virus-1 as a tool for drug discovery
- Author
-
Akhter Molla, Leo W. Barrett, Stephen F. Betz, Kevin Steffy, Kent D. Stewart, Jeffrey R. Huth, Paul L. Richardson, Jean M. Severin, Steve Muchmore, John E. Harlan, Dale J. Kempf, Kevin S. Harris, Edmund D. Matayoshi, Renaldo R. Mendoza, Vincent S. Stoll, Karl A. Walter, Kerry M. Swift, and Emily E. Gramling-Evans
- Subjects
Binding Sites ,Base Sequence ,Drug discovery ,Protein Conformation ,Protein design ,Molecular Sequence Data ,Protein engineering ,Tripeptide ,Circular permutation in proteins ,Biology ,Gp41 ,Protein Engineering ,HIV Envelope Protein gp41 ,Computer Science Applications ,Ectodomain ,Biochemistry ,Drug Design ,Drug Discovery ,HIV-1 ,Humans ,Amino Acid Sequence ,Physical and Theoretical Chemistry ,Binding site - Abstract
Two new proteins of approximately 70 amino acids in length, corresponding to an unnaturally-linked N- and C-helix of the ectodomain of the gp41 protein from the human immunodeficiency virus (HIV) type 1, were designed and characterized. A designed tripeptide links the C-terminus of the C-helix with the N-terminus of the N-helix in a circular permutation so that the C-helix precedes the N-helix in sequence. In addition to the artificial peptide linkage, the C-helix is truncated at its N-terminus to expose a region of the N-helix known as the “Trp-Trp-Ile” binding pocket. Sedimentation, crystallographic, and nuclear magnetic resonance studies confirmed that the protein had the desired trimeric structure with an unoccupied binding site. Spectroscopic and centrifugation studies demonstrated that the engineered protein had ligand binding characteristics similar to previously reported constructs. Unlike previous constructs which expose additional, shallow, non-conserved, and undesired binding pockets, only the single deep and conserved Trp-Trp-Ile pocket is exposed in the proteins of this study. This engineered version of gp41 protein will be potentially useful in research programs aimed at discovery of new drugs for therapy of HIV-infection in humans.
- Published
- 2006
16. A high throughput fluorescent assay for measuring the activity of fatty acid amide hydrolase
- Author
-
Paul L. Richardson, Usha Warrior, Jean M. Severin, Carol S. Surowy, Victoria E. Scott, Ana Pereda-Lopez, Connie R. Faltynek, Melissa H. Vos, Thomas H. Lubben, Larry R. Solomon, Karen Kage, Karl A. Walter, Linda Traphagen, Diane Bartley, Rachel Davis-Taber, John E. Harlan, and Thomas F. Holzman
- Subjects
chemistry.chemical_classification ,Cannabinoid receptor ,Chemistry ,General Neuroscience ,High-throughput screening ,Fatty acid ,Reproducibility of Results ,Anandamide ,Decanoic acid ,Endocannabinoid system ,Amidohydrolases ,chemistry.chemical_compound ,Automation ,Enzyme ,nervous system ,Biochemistry ,Fatty acid amide hydrolase ,Coumarins ,Humans ,lipids (amino acids, peptides, and proteins) ,Biological Assay ,Indicators and Reagents ,psychological phenomena and processes ,Fluorescent Dyes - Abstract
Fatty acid amide hydrolase (FAAH) is the enzyme responsible for the rapid degradation of fatty acid amides such as the endocannabinoid anandamide. Inhibition of FAAH activity has been suggested as a therapeutic approach for the treatment of chronic pain, depression and anxiety, through local activation of the cannabinoid receptor CB1. We have developed a high throughput screening assay for identification of FAAH inhibitors using a novel substrate, decanoyl 7-amino-4-methyl coumarin (D-AMC) that is cleaved by FAAH to release decanoic acid and the highly fluorescent molecule 7-amino-4-methyl coumarin (AMC). This assay gives an excellent signal window for measuring FAAH activity and, as a continuous assay, inherently offers improved sensitivity and accuracy over previously reported endpoint assays. The assay was validated using a panel of known FAAH inhibitors and purified recombinant human FAAH, then converted to a 384 well format and used to screen a large library of compounds (>600,000 compounds) to identify FAAH inhibitors. This screen identified numerous novel FAAH inhibitors of diverse chemotypes. These hits confirmed using a native FAAH substrate, anandamide, and had very similar rank order potency to that obtained using the D-AMC substrate. Collectively these data demonstrate that D-AMC can be successfully used to rapidly and effectively identify novel FAAH inhibitors for potential therapeutic use.
- Published
- 2006
17. Chkl binds and phosphorylates BAD protein
- Author
-
Edward Kyu-ho, Han, Chris, Butler, Haichao, Zhang, Jean M, Severin, Wenying, Qin, Tom F, Holzman, Earl J, Gubbins, Robert L, Simmer, Saul, Rosenberg, Vincent L, Giranda, Shi-Chung, Ng, and Y, Luo
- Subjects
Polyisoprenyl Phosphates ,Checkpoint Kinase 1 ,Molecular Sequence Data ,Humans ,bcl-Associated Death Protein ,Amino Acid Sequence ,Carrier Proteins ,Embryo, Mammalian ,Kidney ,Protein Kinases ,Cell Line ,Protein Binding - Abstract
Chk1 (checkpoint kinase 1) is a serine-threonine kinase that is critical for G2/M arrest in response to DNA damage. Chk1 phosphorylates Cdc25C at serine-216, a major regulatory site, in response to DNA damage. Furthermore, Chk1 also phosphorylates Cdc25A on serine 123 which accelerates its degradation through the ubiquitin-proteasome pathway and arrests cells in late G2-phase after DNA damage. In the present study, we demonstrated that Chk1 phosphorylates pro-apoptotic protein BAD (Bcl-2/Bcl-XL-Antagonist, causing cell Death) in vitro. In vitro phosphorylation analysis with various mouse BAD peptides has revealed two phosphorylation sites for Chk1 at serine-155 and serine-170. When wild-type and mutant BAD (S155A) constructs were transfected into 293T cells, an association between BAD and Chk1 was observed by co-immunoprecipitation. In addition, there was an increase in the phosphorylation of serine-155 following DNA damage by adriamycin treatment. Our results suggest that Chk1 associates with BAD and phosphorylates the BAD protein at serine-155. Taken together, our results suggest that Chk1 may inactivate BAD by associating with and phosphorylating residues critical for BAD function in response to DNA damage.
- Published
- 2005
18. Identification of novel inhibitors of urokinase via NMR-based screening
- Author
-
Philip J. Hajduk, Richard D. Smith, Jean M. Severin, Stephen W. Fesik, Boyd Steven A, Vicki L. Nienaber, David G. Nettesheim, Don J. Davidson, and Todd W. Rockway
- Subjects
Urokinase ,Models, Molecular ,Magnetic Resonance Spectroscopy ,Bicyclic molecule ,biology ,Molecular model ,Blood clotting ,Chemistry ,Stereochemistry ,Crystallography, X-Ray ,Urokinase-Type Plasminogen Activator ,Inhibitory potency ,Structure-Activity Relationship ,Biochemistry ,Enzyme inhibitor ,Drug Discovery ,biology.protein ,medicine ,Molecular Medicine ,Moiety ,Benzimidazoles ,Enzyme Inhibitors ,Electrostatic interaction ,medicine.drug - Abstract
Using an NMR-based screen, a novel class of urokinase inhibitors were identified that contain a 2-aminobenzimidazole moiety. The inhibitory potency of this family of inhibitors is similar to that of inhibitors containing a guanidine or amidine group. However, unlike previously described guanidino- or amidino-based inhibitors which have pK(a) values greater than 9.0, urokinase inhibitors containing a 2-aminobenzimidazole have pK(a) values of 7.5. Thus, 2-aminobenzimidazoles may have improved pharmacokinetic properties which could increase the bioavailability of inhibitors which contain this moiety. A crystal structure of one of the lead inhibitors, 2-amino-5-hydroxybenzimidazole, complexed with urokinase reveals the electrostatic and hydrophobic interactions that stabilize complex formation and suggests nearby subsites that may be accessed to increase the potency of this new series of urokinase inhibitors.
- Published
- 2000
19. Solution structure of an rRNA methyltransferase (ErmAM) that confers macrolide-lincosamide-streptogramin antibiotic resistance
- Author
-
Ping Zhong, Liping Yu, Arndt Schnuchel, Jean M. Severin, Karl A. Walter, Andrew M. Petros, Stephen W. Fesik, and Thomas F. Holzman
- Subjects
Models, Molecular ,Protein Folding ,Methyltransferase ,Magnetic Resonance Spectroscopy ,medicine.drug_class ,Protein Conformation ,Molecular Sequence Data ,Biology ,Biochemistry ,Ribosome ,Virginiamycin ,Protein structure ,Structural Biology ,23S ribosomal RNA ,Genetics ,medicine ,Amino Acid Sequence ,Binding site ,Enzyme Inhibitors ,Lincosamides ,chemistry.chemical_classification ,Binding Sites ,Sequence Homology, Amino Acid ,Drug Resistance, Microbial ,Methyltransferases ,Ribosomal RNA ,Anti-Bacterial Agents ,Enzyme ,chemistry ,Drug Design ,Macrolides - Abstract
The Erm family of methyltransferases is responsible for the development of resistance to the macrolide-lincosamide-streptogramin type B (MLS) antibiotics. These enzymes methylate an adenine of 23S ribosomal RNA that prevents the MLS antibiotics from binding to the ribosome and exhibiting their antibacterial activity. Here we describe the three-dimensional structure of an Erm family member, ErmAM, as determined by NMR spectroscopy. The catalytic domain of ErmAM is structurally similar to that found in other methyltransferases and consists of a seven-stranded beta-sheet flanked by alpha-helices and a small two-stranded beta-sheet. In contrast to the catalytic domain, the substrate binding domain is different from other methyltransferases and adopts a novel fold that consists of four alpha-helices.
- Published
- 1997
20. Three-dimensional structure of the FK506 binding protein/ascomycin complex in solution by heteronuclear three- and four-dimensional NMR
- Author
-
Robert P. Meadows, Edward T. Olejniczak, David G. Nettesheim, Andrew M. Petros, Harriet T. Smith, Earl J. Gubbins, Stephen W. Fesik, Jean M. Severin, Robert X. Xu, and Thomas F. Holzman
- Subjects
Models, Molecular ,Magnetic Resonance Spectroscopy ,Analytical chemistry ,Molecular Conformation ,Nuclear Overhauser effect ,Crystal structure ,Dihedral angle ,Ligands ,Biochemistry ,Protein Structure, Secondary ,Tacrolimus ,Tacrolimus Binding Proteins ,medicine ,Humans ,Ascomycin ,Heat-Shock Proteins ,Chemistry ,Hydrogen bond ,Water ,Hydrogen Bonding ,Nuclear magnetic resonance spectroscopy ,Recombinant Proteins ,Solutions ,Crystallography ,FKBP ,Heteronuclear molecule ,Carrier Proteins ,medicine.drug - Abstract
A high-resolution three-dimensional solution structure of the FKBP/ascomycin complex has been determined using heteronuclear multidimensional nuclear magnetic resonance spectroscopy (NMR) and a distance geometry/simulated annealing protocol. A total of 43 structures of the complex, including 3 tightly bound water molecules, were obtained using 1958 experimental restraints consisting of 1724 nuclear Overhauser effect (NOE) derived distances, 66 chi 1 and 46 phi angular restraints, and 122 hydrogen bond restraints. The root mean square (rms) deviations between the 43 FKBP/ascomycin solution structures and the mean atomic coordinates were 0.43 +/- 0.08 A for the backbone heavy atoms and 0.80 +/- 0.08 A for all non-hydrogen atoms. Angular order parameters for the family of 43 conformations were calculated to determine dihedral convergence. Order parameters for phi, psi, and chi 1 angles exhibited mean values of 0.98, 0.97, and 0.95, respectively, while the mean of the chi 2 order parameter was 0.63. Comparisons were made between the FKBP/ascomycin complex and two NMR-derived solution structures of unbound FKBP and the X-ray crystal structure of an FKBP/FK506 complex. Differences were observed between the FKBP/ascomycin complex and uncomplexed FKBP for residues 33-45 and 78-92. In contrast, the NMR-derived solution structure of the FKBP/ascomycin complex and the X-ray crystal structure of the FKBP/FK506 complex were very similar. Differences between the two complexes were mainly observed in the conformations of some highly solvent exposed side chains.
- Published
- 1993
21. Toxicological Evaluation of Thiol-Reactive Compounds Identified Using a La Assay To Detect Reactive Molecules by Nuclear Magnetic Resonance.
- Author
-
Jeffrey R. Huth, Danying Song, Renaldo R. Mendoza, Candice L. Black-Schaefer, Jamey C. Mack, Sarah A. Dorwin, Uri S. Ladror, Jean M. Severin, Karl A. Walter, Diane M. Bartley, and Philip J. Hajduk
- Published
- 2007
- Full Text
- View/download PDF
22. 1H,13C and 15N backbone assignments of cyclophilin when bound to cyclosporin A (CsA) and preliminary structural characterization of the CsA binding site
- Author
-
Gerd Gemmecker, Jean M. Severin, Edward T. Olejniczak, Stephen W. Fesik, Thomas F. Holzman, Robert L. Simmer, Timothy M. Logan, Rosalind Helfrich, David G. Nettesheim, Placido Neri, and Robert P. Meadows
- Subjects
Models, Molecular ,Magnetic Resonance Spectroscopy ,Nitrogen ,Protein Conformation ,Stereochemistry ,Molecular Sequence Data ,Biophysics ,Biochemistry ,Structural Biology ,Cyclosporin a ,Genetics ,Humans ,Amino Acid Sequence ,Cloning, Molecular ,Binding site ,Molecular Biology ,Cyclophilin ,Amino Acid Isomerases ,Peptidylprolyl isomerase ,Carbon Isotopes ,Binding Sites ,Chemistry ,Cell Biology ,Peptidylprolyl Isomerase ,Recombinant Proteins ,NMR ,Cyclosporin A ,FKBP ,Heteronuclear molecule ,Cyclosporine ,Carrier Proteins ,Two-dimensional nuclear magnetic resonance spectroscopy ,Heteronuclear single quantum coherence spectroscopy ,Hydrogen - Abstract
The backbone 1H, 13C and 15N chemical shifts of cyclophilin (CyP) when bound to cyclosporin A (CsA) have been assigned from heteronuclear two- and three-dimensional NMR experiments involving selectively 15N- and uniformly 15N- and 15N,13C-labeled cyclophilin. From an analysis of the 1H and 15N chemical shifts of CyP that change upon binding to CsA and from CyP/CsA NOEs, we have determined the regions of cyclophilin involved in binding to CsA.
- Full Text
- View/download PDF
23. Structure-directed discovery of potent non-peptidic inhibitors of human urokinase that access a novel binding subsite
- Author
-
Karl A. Walter, Richard A. Smith, Todd W. Rockway, Robert A. Mantei, Sean M. Merrick, Rohinton Edalji, Michael D. Wendt, Vicki L. Nienaber, Moshe Weitzberg, Jieyi Wang, Jack Henkin, Vincent L. Giranda, Peter Magdalinos, Kent D. Stewart, Vered Klinghofer, Jean M. Severin, Xumiao Zhao, and Donald J. Davidson
- Subjects
Models, Molecular ,Proteases ,Macromolecular Substances ,medicine.medical_treatment ,Naphthalenes ,Protein degradation ,Crystallography, X-Ray ,Drug design ,Substrate Specificity ,Protein structure ,Thrombin ,Structural Biology ,medicine ,Humans ,Enzyme Inhibitors ,Urokinase ,Molecular Biology ,X-ray crystallography ,Binding Sites ,Protease ,Chemistry ,Inhibitors ,Urokinase-Type Plasminogen Activator ,Protein Structure, Tertiary ,Urokinase receptor ,Biochemistry ,Plasminogen activator ,Tumor metastasis ,medicine.drug - Abstract
Background: Human urokinase-type plasminogen activator has been implicated in the regulation and control of basement membrane and interstitial protein degradation. Because of its role in tissue remodeling, urokinase is a central player in the disease progression of cancer, making it an attractive target for design of an anticancer clinical agent. Few urokinase inhibitors have been described, which suggests that discovery of such a compound is in the early stages. Towards integrating structural data into this process, a new human urokinase crystal form amenable to structure-based drug design has been used to discover potent urokinase inhibitors. Results: On the basis of crystallographic data, 2-naphthamidine was chosen as the lead scaffold for structure-directed optimization. This co-crystal structure shows the compound binding at the primary specificity pocket of the trypsin-like protease and at a novel binding subsite that is accessible from the 8-position of 2-napthamidine. This novel subsite was characterized and used to design two compounds with very different 8-substituents that inhibit urokinase with K i values of 30–40 nM. Conclusions: Utilization of a novel subsite yielded two potent urokinase inhibitors even though this site has not been widely used in inhibitor optimization with other trypsin-like proteases, such as those reported for thrombin or factor Xa. The extensive binding pockets present at the substrate-binding groove of these other proteins are blocked by unique insertion loops in urokinase, thus necessitating the utilization of additional binding subsites. Successful implementation of this strategy and characterization of the novel site provides a significant step towards the discovery of an anticancer clinical agent.
- Full Text
- View/download PDF
24. Stimulation of fibroblast proliferation and prostaglandin production by purified recombinant murine interleukin 1
- Author
-
Steven B. Mizel, Shikego Yamazaki, Steven J. White, Jean M. Severin, and Mitchell Dukovich
- Subjects
Immunology ,Clone (cell biology) ,Biology ,medicine.disease_cause ,Dinoprostone ,Pathology and Forensic Medicine ,law.invention ,Mice ,law ,medicine ,Immunology and Allergy ,Animals ,Humans ,Prostaglandin E2 ,Fibroblast ,Escherichia coli ,Expression vector ,Dose-Response Relationship, Drug ,Prostaglandins E ,Interleukin ,Biological activity ,Fibroblasts ,Recombinant Proteins ,Molecular Weight ,medicine.anatomical_structure ,Biochemistry ,Recombinant DNA ,Cell Division ,medicine.drug ,Interleukin-1 - Abstract
Recombinant murine interleukin 1 (IL-1) obtained from a clone of Escherichia coli containing an IL-1 expression plasmid was purified to homogeneity using a sequential extraction procedure and gel filtration chromatography. The purified recombinant IL-1 exhibited a pI of approximately 5.2 and a sp act of 6 × 10 6 units/mg. These values are in agreement with those obtained with natural murine IL-1. The purified recombinant IL-1 enhanced the proliferation of human HEL and WI-38 fibroblasts in a serum-free medium. In addition, IL-1 stimulated fibroblast PGE 2 5- to 30-fold over a 24-hr period. The effects of IL-1 on fibroblast activation were obtained with the same concentrations of IL-1 that are effective in the mouse thymocyte assay. These results unequivocally establish the ability of IL-1 to modulate fibroblast proliferation and function.
- Published
- 1986
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.