Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem Lett ; 80: 129084, 2023 01 15.
Article in English | MEDLINE | ID: mdl-36423823

ABSTRACT

In the treatment of non-small cell lung cancer (NSCLC), patients harboring exon 20 insertion mutations in the epidermal growth factor receptor (EGFR) gene (EGFR) have few effective therapies because this subset of mutants is generally resistant to most currently approved EGFR inhibitors. This report describes the structure-guided design of a novel series of potent, irreversible inhibitors of EGFR exon 20 insertion mutations, including the V769_D770insASV and D770_N771insSVD mutants. Extensive structure-activity relationship (SAR) studies led to the discovery of mobocertinib (compound 21c), which inhibited growth of Ba/F3 cells expressing the ASV insertion with a half-maximal inhibitory concentration of 11 nM and with selectivity over wild-type EGFR. Daily oral administration of mobocertinib induced tumor regression in a Ba/F3 ASV xenograft mouse model at well-tolerated doses. Mobocertinib was approved in September 2021 for the treatment of adult patients with advanced NSCLC with EGFR exon 20 insertion mutations with progression on or after platinum-based chemotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Mice , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutagenesis, Insertional , Mutation , ErbB Receptors , Exons , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
2.
J Med Chem ; 65(9): 6775-6802, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35482677

ABSTRACT

d-Serine is a coagonist of the N-methyl d-aspartate (NMDA) receptor, a key excitatory neurotransmitter receptor. In the brain, d-serine is synthesized from its l-isomer by serine racemase and is metabolized by the D-amino acid oxidase (DAO, DAAO). Many studies have linked decreased d-serine concentration and/or increased DAO expression and enzyme activity to NMDA dysfunction and schizophrenia. Thus, it is feasible to employ DAO inhibitors for the treatment of schizophrenia and other indications. Powered by the Schrödinger computational modeling platform, we initiated a research program to identify novel DAO inhibitors with the best-in-class properties. The program execution leveraged an hDAO FEP+ model to prospectively predict compound potency. A new class of DAO inhibitors with desirable properties has been discovered from this endeavor. Our modeling technology on this program has not only enhanced the efficiency of structure-activity relationship development but also helped to identify a previously unexplored subpocket for further optimization.


Subject(s)
N-Methylaspartate , Schizophrenia , D-Amino-Acid Oxidase/metabolism , Humans , Receptors, N-Methyl-D-Aspartate/metabolism , Serine/metabolism , Structure-Activity Relationship
3.
Elife ; 82019 05 13.
Article in English | MEDLINE | ID: mdl-31081496

ABSTRACT

Elucidating the conformational heterogeneity of proteins is essential for understanding protein function and developing exogenous ligands. With the rapid development of experimental and computational methods, it is of great interest to integrate these approaches to illuminate the conformational landscapes of target proteins. SETD8 is a protein lysine methyltransferase (PKMT), which functions in vivo via the methylation of histone and nonhistone targets. Utilizing covalent inhibitors and depleting native ligands to trap hidden conformational states, we obtained diverse X-ray structures of SETD8. These structures were used to seed distributed atomistic molecular dynamics simulations that generated a total of six milliseconds of trajectory data. Markov state models, built via an automated machine learning approach and corroborated experimentally, reveal how slow conformational motions and conformational states are relevant to catalysis. These findings provide molecular insight on enzymatic catalysis and allosteric mechanisms of a PKMT via its detailed conformational landscape.


Subject(s)
Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/metabolism , Allosteric Regulation , Crystallography, X-Ray , Molecular Dynamics Simulation , Protein Conformation
4.
Bioorg Med Chem Lett ; 23(6): 1779-85, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23414845

ABSTRACT

In order to develop potent and selective focal adhesion kinase (FAK) inhibitors, synthetic studies on pyrazolo[4,3-c][2,1]benzothiazines targeted for the FAK allosteric site were carried out. Based on the X-ray structural analysis of the co-crystal of the lead compound, 8-(4-ethylphenyl)-5-methyl-1,5-dihydropyrazolo[4,3-c][2,1]benzothiazine 4,4-dioxide 1 with FAK, we designed and prepared 1,5-dimethyl-1,5-dihydropyrazolo[4,3-c][2,1]benzothiazin derivatives which selectively inhibited kinase activity of FAK without affecting seven other kinases. The optimized compound, N-(4-tert-butylbenzyl)-1,5-dimethyl-1,5-dihydropyrazolo[4,3-c][2,1]benzothiazin-8-amine 4,4-dioxide 30 possessed significant FAK kinase inhibitory activities both in cell-free (IC50=0.64µM) and in cellular assays (IC50=7.1µM). These results clearly demonstrated a potential of FAK allosteric inhibitors as antitumor agents.


Subject(s)
Antineoplastic Agents/chemistry , Cyclic S-Oxides/chemistry , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Heterocyclic Compounds, 3-Ring/chemistry , Protein Kinase Inhibitors/chemistry , Thiazines/chemistry , Allosteric Site , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Binding Sites , Crystallography, X-Ray , Cyclic S-Oxides/chemical synthesis , Cyclic S-Oxides/metabolism , Drug Evaluation, Preclinical , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Heterocyclic Compounds, 3-Ring/chemical synthesis , Heterocyclic Compounds, 3-Ring/metabolism , Molecular Docking Simulation , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Structure-Activity Relationship , Thiazines/chemical synthesis , Thiazines/metabolism
5.
Eur J Med Chem ; 61: 49-60, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22819505

ABSTRACT

Focal adhesion kinase (FAK) regulates cell survival and proliferation pathways. Here we report the discovery of a highly selective series of 1,5-dihydropyrazolo[4,3-c][2,1]benzothiazines that demonstrate a novel mode of allosteric inhibition of FAK. These compounds showed slow dissociation from unphosphorylated FAK and were noncompetitive with ATP after long preincubation. Co-crystal structural analysis revealed that the compounds target a novel allosteric site within the C-lobe of the kinase domain, which induces disruption of ATP pocket formation leading to the inhibition of kinase activity. The potency of allosteric inhibition was reduced by phosphorylation of FAK. Coupled SAR analysis revealed that N-substitution of the fused pyrazole is critical to achieve allosteric binding and high selectivity among kinases.


Subject(s)
Drug Discovery , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Thiazines/pharmacology , Allosteric Regulation/drug effects , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Structure-Activity Relationship , Thiazines/chemical synthesis , Thiazines/chemistry
6.
Bioorg Med Chem Lett ; 22(21): 6628-31, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23025999

ABSTRACT

Dipeptidyl peptidase IV (DPP-4) inhibitors have been shown to enhance GLP-1 levels and thereby improve hyperglycemia in type II diabetes. From a small fragment hit, using structure-based design, we have discovered a new class of non-covalent, potent and selective DPP-4 inhibitors.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors/chemistry , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Drug Design , Pyrimidinones/chemistry , Pyrimidinones/pharmacology , Catalytic Domain , Crystallography, X-Ray , Dipeptidyl-Peptidase IV Inhibitors/chemical synthesis , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Humans , Inhibitory Concentration 50 , Models, Molecular , Pyrimidinones/chemical synthesis
7.
J Med Chem ; 54(2): 510-24, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21186796

ABSTRACT

The discovery of two classes of heterocyclic dipeptidyl peptidase IV (DPP-4) inhibitors, pyrimidinones and pyrimidinediones, is described. After a single oral dose, these potent, selective, and noncovalent inhibitors provide sustained reduction of plasma DPP-4 activity and lowering of blood glucose in animal models of diabetes. Compounds 13a, 27b, and 27j were selected for development.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors/chemical synthesis , Pyrimidinones/chemical synthesis , Animals , Binding Sites , Biological Availability , Crystallography, X-Ray , Cytochrome P-450 Enzyme Inhibitors , Dipeptidyl-Peptidase IV Inhibitors/chemistry , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Dogs , Macaca fascicularis , Models, Molecular , Pyrimidinones/chemistry , Pyrimidinones/pharmacology , Rats , Stereoisomerism , Structure-Activity Relationship
8.
Bioorg Med Chem Lett ; 20(10): 3138-41, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20392637

ABSTRACT

A series of N-hydroxy-3-[3-(1-substituted-1H-benzoimidazol-2-yl)-phenyl]-acrylamides (5a-5ab) and N-hydroxy-3-[3-(1,4,5-trisubstituted-1H-imidazol-2-yl)-phenyl]-acrylamides (12a-s) were designed, synthesized, and found to be nanomolar inhibitors of human histone deacetylases. Multiple compounds bearing an N1-piperidine demonstrate EC(50)s of 20-100 nM in human A549, HL60, and PC3 cells, in vitro and in vivo hyperacetylation of histones H3 and H4, and induction of p21(waf). Compound 5x displays efficacy in human tumor xenograft models.


Subject(s)
Benzimidazoles/chemistry , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/chemistry , Imidazoles/chemistry , Acetylation , Animals , Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacology , Cell Line, Tumor , HL-60 Cells , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Imidazoles/chemical synthesis , Imidazoles/pharmacology , Mice , Mice, Nude , Structure-Activity Relationship , Xenograft Model Antitumor Assays
9.
Bioorg Med Chem Lett ; 18(7): 2362-7, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18346892

ABSTRACT

A novel series of non-covalent, benzimidazole-based inhibitors of DPP-4 has been developed from a small fragment hit using structure-based drug design. A highly versatile synthetic route was created for the development of SAR, which led to the discovery of potent and selective inhibitors with excellent pharmaceutical properties.


Subject(s)
Benzimidazoles/pharmacology , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Drug Design , Hypoglycemic Agents/pharmacology , Animals , Benzimidazoles/chemical synthesis , Crystallography, X-Ray , Dipeptidyl-Peptidase IV Inhibitors/chemical synthesis , Hypoglycemic Agents/chemical synthesis , Models, Chemical , Rats , Structure-Activity Relationship
10.
Structure ; 15(7): 793-805, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17637340

ABSTRACT

Higher eukaryote tRNA synthetases have expanded functions that come from enlarged, more differentiated structures that were adapted to fit aminoacylation function. How those adaptations affect catalytic mechanisms is not known. Presented here is the structure of a catalytically active natural splice variant of human tryptophanyl-tRNA synthetase (TrpRS) that is a potent angiostatic factor. This and related structures suggest that a eukaryote-specific N-terminal extension of the core enzyme changed substrate recognition by forming an active site cap. At the junction of the extension and core catalytic unit, an arginine is recruited to replace a missing landmark lysine almost 200 residues away. Mutagenesis, rapid kinetic, and substrate binding studies support the functional significance of the cap and arginine recruitment. Thus, the enzyme function of human TrpRS has switched more to the N terminus of the sequence. This switch has the effect of creating selective pressure to retain the N-terminal extension for functional expansion.


Subject(s)
Angiogenesis Inhibitors/chemistry , Models, Molecular , Protein Folding , Tryptophan-tRNA Ligase/chemistry , Acetylation , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Crystallography, X-Ray , Geobacillus stearothermophilus/metabolism , Humans , Molecular Sequence Data , Mutation , Protein Structure, Tertiary , RNA Splicing , Substrate Specificity , Tryptophan-tRNA Ligase/genetics
11.
J Med Chem ; 50(10): 2297-300, 2007 May 17.
Article in English | MEDLINE | ID: mdl-17441705

ABSTRACT

Alogliptin is a potent, selective inhibitor of the serine protease dipeptidyl peptidase IV (DPP-4). Herein, we describe the structure-based design and optimization of alogliptin and related quinazolinone-based DPP-4 inhibitors. Following an oral dose, these noncovalent inhibitors provide sustained reduction of plasma DPP-4 activity and a lowering of blood glucose in animal models of diabetes. Alogliptin is currently undergoing phase III trials in patients with type 2 diabetes.


Subject(s)
Dipeptidyl Peptidase 4/chemistry , Dipeptidyl-Peptidase IV Inhibitors , Hypoglycemic Agents/chemical synthesis , Piperidines/chemical synthesis , Pyrimidinones/chemical synthesis , Quinazolinones/chemical synthesis , Uracil/analogs & derivatives , Animals , Binding Sites , Blood Glucose/analysis , Cytochrome P-450 Enzyme Inhibitors , Diabetes Mellitus, Experimental/drug therapy , Dogs , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/drug effects , Female , Glucose Tolerance Test , Haplorhini , Humans , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Models, Molecular , Piperidines/pharmacokinetics , Piperidines/pharmacology , Pyrimidinones/pharmacokinetics , Pyrimidinones/pharmacology , Quinazolinones/pharmacokinetics , Quinazolinones/pharmacology , Rats , Rats, Wistar , Structure-Activity Relationship , Uracil/chemical synthesis , Uracil/pharmacokinetics , Uracil/pharmacology
12.
EMBO J ; 25(12): 2919-29, 2006 Jun 21.
Article in English | MEDLINE | ID: mdl-16724112

ABSTRACT

Aminoacylation of tRNA is the first step of protein synthesis. Here, we report the co-crystal structure of human tryptophanyl-tRNA synthetase and tRNATrp. This enzyme is reported to interact directly with elongation factor 1alpha, which carries charged tRNA to the ribosome. Crystals were generated from a 50/50% mixture of charged and uncharged tRNATrp. These crystals captured two conformations of the complex, which are nearly identical with respect to the protein and a bound tryptophan. They are distinguished by the way tRNA is bound. In one, uncharged tRNA is bound across the dimer, with anticodon and acceptor stem interacting with separate subunits. In this cross-dimer tRNA complex, the class I enzyme has a class II-like tRNA binding mode. This structure accounts for biochemical investigations of human TrpRS, including species-specific charging. In the other conformation, presumptive aminoacylated tRNA is bound only by the anticodon, the acceptor stem being free and having space to interact precisely with EF-1alpha, suggesting that the product of aminoacylation can be directly handed off to EF-1alpha for the next step of protein synthesis.


Subject(s)
Nucleic Acid Conformation , Protein Biosynthesis , RNA, Transfer, Trp/chemistry , RNA, Transfer, Trp/metabolism , Tryptophan-tRNA Ligase/chemistry , Tryptophan-tRNA Ligase/metabolism , Amino Acid Sequence , Anticodon/genetics , Crystallization , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Peptide Elongation Factor 1/metabolism , Protein Binding , Protein Conformation , Sequence Alignment , Tryptophan/genetics
13.
J Biol Chem ; 280(6): 4639-48, 2005 Feb 11.
Article in English | MEDLINE | ID: mdl-15513927

ABSTRACT

Human 11beta-hydroxysteroid dehydrogenase type I (11beta-HSD1) is an ER-localized membrane protein that catalyzes the interconversion of cortisone and cortisol. In adipose tissue, excessive cortisol production through 11beta-HSD1 activity has been implicated in the pathogenesis of type II diabetes and obesity. We report here biophysical, kinetic, mutagenesis, and structural data on two ternary complexes of 11beta-HSD1. The combined results reveal flexible active site interactions relevant to glucocorticoid recognition and demonstrate how four 11beta-HSD1 C termini converge to form an as yet uncharacterized tetramerization motif. A C-terminal Pro-Cys motif is localized at the center of the tetramer and forms reversible enzyme disulfides that alter enzyme activity. Conformational flexibility at the tetramerization interface is coupled to structural changes at the enzyme active site suggesting how the central Pro-Cys motif may regulate enzyme activity. Together, the crystallographic and biophysical data provide a structural framework for understanding 11beta-HSD1 activities and will ultimately facilitate the development of specific inhibitors.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenases/chemistry , Glucocorticoids/chemistry , Amino Acid Motifs , Binding Sites , Cloning, Molecular , Cortisone/chemistry , Crystallography, X-Ray , Cysteine/chemistry , Diabetes Mellitus, Type 2/metabolism , Dimerization , Disulfides/chemistry , Endoplasmic Reticulum/metabolism , Enzyme Inhibitors/pharmacology , Escherichia coli/metabolism , Humans , Hydrocortisone/chemistry , Kinetics , Mass Spectrometry , Models, Molecular , Mutagenesis , Obesity/metabolism , Oxygen/metabolism , Point Mutation , Proline/chemistry , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Recombinant Proteins/chemistry
14.
Structure ; 12(7): 1325-34, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15242608

ABSTRACT

Modulation of the acetylation state of histones plays a pivotal role in the regulation of gene expression. Histone deacetylases (HDACs) catalyze the removal of acetyl groups from lysines near the N termini of histones. This reaction promotes the condensation of chromatin, leading to repression of transcription. HDAC deregulation has been linked to several types of cancer, suggesting a potential use for HDAC inhibitors in oncology. Here we describe the first crystal structures of a human HDAC: the structures of human HDAC8 complexed with four structurally diverse hydroxamate inhibitors. This work sheds light on the catalytic mechanism of the HDACs, and on differences in substrate specificity across the HDAC family. The structure also suggests how phosphorylation of Ser39 affects HDAC8 activity.


Subject(s)
Histone Deacetylases/chemistry , Repressor Proteins/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Histone Deacetylases/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Molecular Structure , Protein Conformation , Repressor Proteins/metabolism , Substrate Specificity
15.
J Bacteriol ; 186(12): 3903-10, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15175304

ABSTRACT

We have determined the 1.35- and 1.45-A structures, respectively, of closed and open iron-loaded forms of Mannheimia haemolytica ferric ion-binding protein A. M. haemolytica is the causative agent in the economically important and fatal disease of cattle termed shipping fever. The periplasmic iron-binding protein of this gram-negative bacterium, which has homologous counterparts in many other pathogenic species, performs a key role in iron acquisition from mammalian host serum iron transport proteins and is essential for the survival of the pathogen within the host. The ferric (Fe(3+)) ion in the closed structure is bound by a novel asymmetric constellation of four ligands, including a synergistic carbonate anion. The open structure is ligated by three tyrosyl residues and a dynamically disordered solvent-exposed anion. Our results clearly implicate the synergistic anion as the primary mediator of global protein conformation and provide detailed insights into the molecular mechanisms of iron binding and release in the periplasm.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Iron/metabolism , Mannheimia haemolytica/metabolism , Periplasm/metabolism , Animals , Bacterial Outer Membrane Proteins , Cattle , Crystallization , Ferric Compounds/metabolism , Iron-Binding Proteins , Mannheimia haemolytica/chemistry , Models, Molecular , Molecular Sequence Data , Periplasmic Binding Proteins , Protein Conformation , X-Ray Diffraction
16.
J Biol Chem ; 279(30): 31655-63, 2004 Jul 23.
Article in English | MEDLINE | ID: mdl-15123710

ABSTRACT

The activity of the c-Kit receptor protein-tyrosine kinase is tightly regulated in normal cells, whereas deregulated c-Kit kinase activity is implicated in the pathogenesis of human cancers. The c-Kit juxtamembrane region is known to have an autoinhibitory function; however the precise mechanism by which c-Kit is maintained in an autoinhibited state is not known. We report the 1.9-A resolution crystal structure of native c-Kit kinase in an autoinhibited conformation and compare it with active c-Kit kinase. Autoinhibited c-Kit is stabilized by the juxtamembrane domain, which inserts into the kinase-active site and disrupts formation of the activated structure. A 1.6-A crystal structure of c-Kit in complex with STI-571 (Imatinib or Gleevec) demonstrates that inhibitor binding disrupts this natural mechanism for maintaining c-Kit in an autoinhibited state. Together, these results provide a structural basis for understanding c-Kit kinase autoinhibition and will facilitate the structure-guided design of specific inhibitors that target the activated and autoinhibited conformations of c-Kit kinase.


Subject(s)
Enzyme Inhibitors/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-kit/chemistry , Pyrimidines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Amino Acid Motifs , Amino Acid Sequence , Aspartic Acid/chemistry , Benzamides , Catalytic Domain , Conserved Sequence , Crystallography, X-Ray , Enzyme Activation , Humans , Imatinib Mesylate , In Vitro Techniques , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Conformation , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sequence Homology, Amino Acid , Static Electricity
17.
Mol Cell ; 13(6): 829-41, 2004 Mar 26.
Article in English | MEDLINE | ID: mdl-15053876

ABSTRACT

Early work on aminoacylation of alanine-specific tRNA (tRNA(Ala)) by alanyl-tRNA synthetase (AlaRS) gave rise to the concept of an early "second genetic code" imbedded in the acceptor stems of tRNAs. A single conserved and position-specific G:U base pair in the tRNA acceptor stem is the key identity determinant. Further understanding has been limited due to lack of a crystal structure of the enzyme. We determined a 2.14 A crystal structure of the 453 amino acid catalytic fragment of Aquifex aeolicus AlaRS. It contains the catalytic domain characteristic of class II synthetases, a helical domain with a hairpin motif critical for acceptor-stem recognition, and a C-terminal domain of a mixed alpha/beta fold. Docking of tRNA(Ala) on AlaRS shows critical contacts with the three domains, consistent with previous mutagenesis and functional data. It also suggests conformational flexibility within the C domain, which might allow for the positional variation of the key G:U base pair seen in some tRNA(Ala)s.


Subject(s)
Alanine-tRNA Ligase/chemistry , Bacterial Proteins/chemistry , Alanine-tRNA Ligase/genetics , Amino Acid Motifs , Amino Acid Sequence , Bacterial Proteins/genetics , Base Pairing , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/enzymology , Escherichia coli/genetics , Hydrophobic and Hydrophilic Interactions , Kinetics , Models, Molecular , Protein Structure, Secondary , Protein Structure, Tertiary , RNA, Bacterial/metabolism , Sequence Homology, Amino Acid , Substrate Specificity
18.
Protein Sci ; 13(2): 412-21, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14718659

ABSTRACT

Dipeptidyl peptidase IV (DPPIV) is a member of the prolyl oligopeptidase family of serine proteases. DPPIV removes dipeptides from the N terminus of substrates, including many chemokines, neuropeptides, and peptide hormones. Specific inhibition of DPPIV is being investigated in human trials for the treatment of type II diabetes. To understand better the molecular determinants that underlie enzyme catalysis and substrate specificity, we report the crystal structures of DPPIV in the free form and in complex with the first 10 residues of the physiological substrate, Neuropeptide Y (residues 1-10; tNPY). The crystal structure of the free form of the enzyme reveals two potential channels through which substrates could access the active site-a so-called propeller opening, and side opening. The crystal structure of the DPPIV/tNPY complex suggests that bioactive peptides utilize the side opening unique to DPPIV to access the active site. Other structural features in the active site such as the presence of a Glu motif, a well-defined hydrophobic S1 subsite, and minimal long-range interactions explain the substrate recognition and binding properties of DPPIV. Moreover, in the DPPIV/tNPY complex structure, the peptide is not cleaved but trapped in a tetrahedral intermediate that occurs during catalysis. Conformational changes of S630 and H740 between DPPIV in its free form and in complex with tNPY were observed and contribute to the stabilization of the tetrahedral intermediate. Our results facilitate the design of potent, selective small molecule inhibitors of DPPIV that may yield compounds for the development of novel drugs to treat type II diabetes.


Subject(s)
Dipeptidyl Peptidase 4/chemistry , Dipeptidyl Peptidase 4/metabolism , Neuropeptide Y/chemistry , Oligopeptides/chemistry , Oligopeptides/metabolism , Animals , Binding Sites , Catalytic Domain , Cells, Cultured , Crystallization , Crystallography, X-Ray , Humans , Insecta , Models, Molecular , Protein Structure, Tertiary , Substrate Specificity
19.
Proc Natl Acad Sci U S A ; 100(26): 15376-80, 2003 Dec 23.
Article in English | MEDLINE | ID: mdl-14671330

ABSTRACT

Early forms of the genetic code likely generated "statistical" proteins, with similar side chains occupying the same sequence positions at different ratios. In this scenario, groups of related side chains were treated by aminoacyl-tRNA synthetases as a single molecular species until a discrimination mechanism developed that could separate them. The aromatic amino acids tryptophan, tyrosine, and phenylalanine likely constituted one of these groups. A crystal structure of human tryptophanyl-tRNA synthetase was solved at 2.1 A with a tryptophanyl-adenylate bound at the active site. A cocrystal structure of an active fragment of human tyrosyl-tRNA synthetase with its cognate amino acid analog was also solved at 1.6 A. The two structures enabled active site identifications and provided the information for structure-based sequence alignments of approximately 45 orthologs of each enzyme. Two critical positions shared by all tyrosyl-tRNA synthetases and tryptophanyl-tRNA synthetases for amino acid discrimination were identified. The variations at these two positions and phylogenetic analyses based on the structural information suggest that, in contrast to many other amino acids, discrimination of tyrosine from tryptophan occurred late in the development of the genetic code.


Subject(s)
Genetic Code , Tryptophan-tRNA Ligase/chemistry , Tryptophan-tRNA Ligase/genetics , Adenosine Monophosphate , Amino Acid Sequence , Binding Sites , Conserved Sequence , Crystallography, X-Ray , Geobacillus stearothermophilus/enzymology , Humans , Models, Molecular , Molecular Sequence Data , Phylogeny , Protein Conformation , Sensitivity and Specificity , Sequence Alignment , Sequence Homology, Amino Acid , Tryptophan
20.
Biochemistry ; 42(41): 11908-14, 2003 Oct 21.
Article in English | MEDLINE | ID: mdl-14556621

ABSTRACT

The periplasmic iron binding protein plays an essential role in the iron uptake pathway of Gram-negative pathogenic bacteria from the Pasteurellaceae and Neisseriaceae families and is critical for survival of these pathogens within the host. In this study, we report the crystal structures of two mutant forms of ferric ion-binding protein A (FbpA) from Haemophilus influenzae with bound multinuclear oxo-metal clusters. Crystals of site-directed mutants in the metal or anion binding ligands contain protein in the open conformation, and two mutant FbpAs, H9A and N175L, contain different cluster arrangements in the iron-binding pocket. The iron clusters are anchored by binding to the two tyrosine ligands (Tyr195 and Tyr196) positioned at the vertex of the iron-binding pocket but are not coordinated by the other metal binding ligands. Our results suggest that the metal clusters may have formed in situ, suggesting that the mutant FbpAs may serve as a simple model for protein-mediated mineralization.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Ferric Compounds/chemistry , Haemophilus influenzae/chemistry , Mutagenesis, Site-Directed , Alanine/genetics , Asparagine/genetics , Bacterial Outer Membrane Proteins , Crystallization , Crystallography, X-Ray , Haemophilus influenzae/genetics , Histidine/genetics , Iron-Binding Proteins , Leucine/genetics , Periplasmic Binding Proteins , Protein Binding/genetics , Solutions , Transferrin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...