Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Commun Biol ; 5(1): 346, 2022 04 11.
Article in English | MEDLINE | ID: mdl-35410329

ABSTRACT

Human serine racemase (hSR) catalyses racemisation of L-serine to D-serine, the latter of which is a co-agonist of the NMDA subtype of glutamate receptors that are important in synaptic plasticity, learning and memory. In a 'closed' hSR structure containing the allosteric activator ATP, the inhibitor malonate is enclosed between the large and small domains while ATP is distal to the active site, residing at the dimer interface with the Tyr121 hydroxyl group contacting the α-phosphate of ATP. In contrast, in 'open' hSR structures, Tyr121 sits in the core of the small domain with its hydroxyl contacting the key catalytic residue Ser84. The ability to regulate SR activity by flipping Tyr121 from the core of the small domain to the dimer interface appears to have evolved in animals with a CNS. Multiple X-ray crystallographic enzyme-fragment structures show Tyr121 flipped out of its pocket in the core of the small domain. Data suggest that this ligandable pocket could be targeted by molecules that inhibit enzyme activity.


Subject(s)
Racemases and Epimerases , Tyrosine , Adenosine Triphosphate/chemistry , Animals , Catalysis , Racemases and Epimerases/genetics , Serine
2.
Chembiochem ; 23(1): e202100441, 2022 01 05.
Article in English | MEDLINE | ID: mdl-34726826

ABSTRACT

STE20/SPS1-related proline/alanine-rich kinase (SPAK) and oxidative stress responsive 1 (OSR1) kinase are two serine/threonine protein kinases that regulate the function of ion co-transporters through phosphorylation. The highly conserved C-terminal (CCT) domains of SPAK and OSR1 bind to RFx[V/I] peptide sequences from their upstream 'With No Lysine Kinases (WNKs), facilitating their activation via phosphorylation. Thus, the inhibition of SPAK and OSR1 binding, via their CCT domains, to WNK kinases is a plausible strategy for inhibiting SPAK and OSR1 kinases. To facilitate structure-guided drug design of such inhibitors, we expressed and purified human SPAK and OSR1 CCT domains and solved their crystal structures. Interestingly, these crystal structures show a highly conserved primary pocket adjacent to a flexible secondary pocket. We also employed a biophysical strategy and determined the affinity of SPAK and OSR1 CCT domains to short peptides derived from WNK4 and NKCC1. Together, this work provides a platform that facilitates the design of CCT domain specific small molecule binders that inhibit SPAK- and OSR1-activation by WNK kinases, and these could be useful in treating hypertension and ischemic stroke.


Subject(s)
Protein Serine-Threonine Kinases/chemistry , Crystallography, X-Ray , Humans , Models, Molecular , Protein Domains , Protein Serine-Threonine Kinases/metabolism
3.
Acta Crystallogr F Struct Biol Commun ; 76(Pt 10): 469-476, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-33006574

ABSTRACT

Inositol monophosphatase (IMPase) is inhibited by lithium, which is the most efficacious treatment for bipolar disorder. Several therapies have been approved, or are going through clinical trials, aimed at the replacement of lithium in the treatment of bipolar disorder. One candidate small molecule is ebselen, a selenium-containing antioxidant, which has been demonstrated to produce lithium-like effects both in a murine model and in clinical trials. Here, the crystallization and the first structure of human IMPase covalently complexed with ebselen, a 1.47 Šresolution crystal structure (PDB entry 6zk0), are presented. In the complex with human IMPase, ebselen in a ring-opened conformation is covalently attached to Cys141, a residue located away from the active site. IMPase is a dimeric enzyme and in the crystal structure two adjacent dimers share four ebselen molecules, creating a tetramer with approximate 222 symmetry. In the crystal structure presented in this publication, the active site in the tetramer is still accessible, suggesting that ebselen may function as an allosteric inhibitor or may block the binding of partner proteins.


Subject(s)
Antioxidants/metabolism , Azoles/metabolism , Crystallography, X-Ray/methods , Cysteine/metabolism , Organoselenium Compounds/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Cysteine/chemistry , Humans , Isoindoles , Protein Binding , Protein Conformation , Protein Domains
4.
Acta Crystallogr F Struct Biol Commun ; 76(Pt 2): 65-73, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-32039887

ABSTRACT

Serine racemase (SR) is a pyridoxal 5'-phosphate (PLP)-containing enzyme that converts L-serine to D-serine, an endogenous co-agonist for the N-methyl-D-aspartate receptor (NMDAR) subtype of glutamate ion channels. SR regulates D-serine levels by the reversible racemization of L-serine to D-serine, as well as the catabolism of serine by α,ß-elimination to produce pyruvate. The modulation of SR activity is therefore an attractive therapeutic approach to disorders associated with abnormal glutamatergic signalling since it allows an indirect modulation of NMDAR function. In the present study, a 1.89 Šresolution crystal structure of the human SR holoenzyme (including the PLP cofactor) with four subunits in the asymmetric unit is described. Comparison of this new structure with the crystal structure of human SR with malonate (PDB entry 3l6b) shows an interdomain cleft that is open in the holo structure but which disappears when the inhibitor malonate binds and is enclosed. This is owing to a shift of the small domain (residues 78-155) in human SR similar to that previously described for the rat enzyme. This domain movement is accompanied by changes within the twist of the central four-stranded ß-sheet of the small domain, including changes in the φ-ψ angles of all three residues in the C-terminal ß-strand (residues 149-151). In the malonate-bound structure, Ser84 (a catalytic residue) points its side chain at the malonate and is preceded by a six-residue ß-strand (residues 78-83), but in the holoenzyme the ß-strand is only four residues (78-81) and His82 has φ-ψ values in the α-helical region of the Ramachandran plot. These data therefore represent a crystallographic platform that enables the structure-guided design of small-molecule modulators for this important but to date undrugged target.


Subject(s)
Protein Conformation , Racemases and Epimerases/chemistry , Serine/chemistry , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Humans , Models, Molecular , Protein Domains
5.
J Mol Biol ; 431(18): 3427-3449, 2019 08 23.
Article in English | MEDLINE | ID: mdl-31301408

ABSTRACT

Type II topoisomerases regulate DNA topology by making a double-stranded break in one DNA duplex, transporting another DNA segment through this break and then resealing it. Bacterial type IIA topoisomerase inhibitors, such as fluoroquinolones and novel bacterial topoisomerase inhibitors, can trap DNA cleavage complexes with double- or single-stranded cleaved DNA. To study the mode of action of such compounds, 21 crystal structures of a "gyraseCORE" fusion truncate of Staphyloccocus aureus DNA gyrase complexed with DNA and diverse inhibitors have been published, as well as 4 structures lacking inhibitors. These structures have the DNA in various cleavage states and appear to track trajectories along the catalytic paths of the DNA cleavage/religation steps. The various conformations sampled by these multiple "gyraseCORE" structures show rigid body movements of the catalytic GyrA WHD and GyrB TOPRIM domains across the dimer interface. Conformational changes common to all compound-bound structures suggest common mechanisms for DNA cleavage-stabilizing compounds. The structures suggest that S. aureus gyrase uses a single moving-metal ion for cleavage and that the central four base pairs need to be stretched between the two catalytic sites, in order for a scissile phosphate to attract a metal ion to the A-site to catalyze cleavage, after which it is "stored" in another coordination configuration (B-site) in the vicinity. We present a simplified model for the catalytic cycle in which capture of the transported DNA segment causes conformational changes in the ATPase domain that push the DNA gate open, resulting in stretching and cleaving the gate-DNA in two steps.


Subject(s)
Anti-Bacterial Agents/pharmacology , DNA Cleavage/drug effects , DNA Topoisomerases, Type I/drug effects , DNA/chemistry , Topoisomerase Inhibitors/pharmacology , Catalytic Domain , DNA/metabolism , DNA Gyrase/chemistry , DNA Gyrase/genetics , DNA Topoisomerases, Type I/chemistry , DNA Topoisomerases, Type II/chemistry , DNA Topoisomerases, Type II/drug effects , Fluoroquinolones/pharmacology , Metals , Models, Molecular , Protein Conformation , Quinolones , Staphylococcus aureus/enzymology , Topoisomerase II Inhibitors/pharmacology
6.
Bioorg Med Chem Lett ; 29(11): 1407-1412, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30962087

ABSTRACT

A series of DNA gyrase inhibitors were designed based on the X-ray structure of a parent thiophene scaffold with the objective to improve biochemical and whole-cell antibacterial activity, while reducing cardiac ion channel activity. The binding mode and overall design hypothesis of one series was confirmed with a co-crystal structure with DNA gyrase. Although some analogs retained both biochemical activity and whole-cell antibacterial activity, we were unable to significantly improve the activity of the series and analogs retained activity against the cardiac ion channels, therefore we stopped optimization efforts.


Subject(s)
Acinetobacter baumannii/drug effects , Anti-Bacterial Agents/pharmacology , DNA Gyrase/metabolism , Drug Design , Escherichia coli/drug effects , Topoisomerase II Inhibitors/pharmacology , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Cell Line , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Mice , Mice, Knockout , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/chemistry
7.
Nucleic Acids Res ; 46(8): 4114-4128, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29538767

ABSTRACT

Imidazopyrazinones (IPYs) are a new class of compounds that target bacterial topoisomerases as a basis for their antibacterial activity. We have characterized the mechanism of these compounds through structural/mechanistic studies showing they bind and stabilize a cleavage complex between DNA gyrase and DNA ('poisoning') in an analogous fashion to fluoroquinolones, but without the requirement for the water-metal-ion bridge. Biochemical experiments and structural studies of cleavage complexes of IPYs compared with an uncleaved gyrase-DNA complex, reveal conformational transitions coupled to DNA cleavage at the DNA gate. These involve movement at the GyrA interface and tilting of the TOPRIM domains toward the scissile phosphate coupled to capture of the catalytic metal ion. Our experiments show that these structural transitions are involved generally in poisoning of gyrase by therapeutic compounds and resemble those undergone by the enzyme during its adenosine triphosphate-coupled strand-passage cycle. In addition to resistance mutations affecting residues that directly interact with the compounds, we characterized a mutant (D82N) that inhibits formation of the cleavage complex by the unpoisoned enzyme. The D82N mutant appears to act by stabilizing the binary conformation of DNA gyrase with uncleaved DNA without direct interaction with the compounds. This provides general insight into the resistance mechanisms to antibiotics targeting bacterial type II topoisomerases.


Subject(s)
Anti-Bacterial Agents/chemistry , DNA Gyrase/chemistry , Pyrazines/chemistry , Topoisomerase II Inhibitors/chemistry , Adenosine Triphosphate/metabolism , Anti-Bacterial Agents/classification , Ciprofloxacin/chemistry , DNA/chemistry , DNA Cleavage , DNA Gyrase/metabolism , Drug Resistance, Bacterial , Fluoroquinolones/chemistry , Magnesium/chemistry , Models, Molecular , Mutation
9.
ACS Med Chem Lett ; 8(10): 1093-1098, 2017 Oct 12.
Article in English | MEDLINE | ID: mdl-29057057

ABSTRACT

Selective inhibitors of phosphoinositide 3-kinase delta are of interest for the treatment of inflammatory diseases. Initial optimization of a 3-substituted indazole hit compound targeting the kinase PIM1 focused on improving selectivity over GSK3ß through consideration of differences in the ATP binding pockets. Continued kinase cross-screening showed PI3Kδ activity in a series of 4,6-disubstituted indazole compounds, and subsequent structure-activity relationship exploration led to the discovery of an indole-containing lead compound as a potent PI3Kδ inhibitor with selectivity over the other PI3K isoforms.

10.
Proc Natl Acad Sci U S A ; 114(22): E4492-E4500, 2017 05 30.
Article in English | MEDLINE | ID: mdl-28507124

ABSTRACT

A paucity of novel acting antibacterials is in development to treat the rising threat of antimicrobial resistance, particularly in Gram-negative hospital pathogens, which has led to renewed efforts in antibiotic drug discovery. Fluoroquinolones are broad-spectrum antibacterials that target DNA gyrase by stabilizing DNA-cleavage complexes, but their clinical utility has been compromised by resistance. We have identified a class of antibacterial thiophenes that target DNA gyrase with a unique mechanism of action and have activity against a range of bacterial pathogens, including strains resistant to fluoroquinolones. Although fluoroquinolones stabilize double-stranded DNA breaks, the antibacterial thiophenes stabilize gyrase-mediated DNA-cleavage complexes in either one DNA strand or both DNA strands. X-ray crystallography of DNA gyrase-DNA complexes shows the compounds binding to a protein pocket between the winged helix domain and topoisomerase-primase domain, remote from the DNA. Mutations of conserved residues around this pocket affect activity of the thiophene inhibitors, consistent with allosteric inhibition of DNA gyrase. This druggable pocket provides potentially complementary opportunities for targeting bacterial topoisomerases for antibiotic development.


Subject(s)
Anti-Bacterial Agents , DNA Cleavage , DNA Gyrase , Thiophenes , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Crystallography, X-Ray , DNA Gyrase/chemistry , DNA Gyrase/metabolism , Drug Discovery , Models, Molecular , Thiophenes/chemistry , Thiophenes/metabolism
11.
Nat Commun ; 6: 10048, 2015 Dec 07.
Article in English | MEDLINE | ID: mdl-26640131

ABSTRACT

New antibacterials are needed to tackle antibiotic-resistant bacteria. Type IIA topoisomerases (topo2As), the targets of fluoroquinolones, regulate DNA topology by creating transient double-strand DNA breaks. Here we report the first co-crystal structures of the antibacterial QPT-1 and the anticancer drug etoposide with Staphylococcus aureus DNA gyrase, showing binding at the same sites in the cleaved DNA as the fluoroquinolone moxifloxacin. Unlike moxifloxacin, QPT-1 and etoposide interact with conserved GyrB TOPRIM residues rationalizing why QPT-1 can overcome fluoroquinolone resistance. Our data show etoposide's antibacterial activity is due to DNA gyrase inhibition and suggests other anticancer agents act similarly. Analysis of multiple DNA gyrase co-crystal structures, including asymmetric cleavage complexes, led to a 'pair of swing-doors' hypothesis in which the movement of one DNA segment regulates cleavage and religation of the second DNA duplex. This mechanism can explain QPT-1's bacterial specificity. Structure-based strategies for developing topo2A antibacterials are suggested.


Subject(s)
Anti-Bacterial Agents/chemistry , Antineoplastic Agents/chemistry , DNA Gyrase/chemistry , Etoposide/chemistry , Fluoroquinolones/chemistry , Staphylococcus aureus/enzymology , Topoisomerase II Inhibitors/chemistry , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , DNA Gyrase/genetics , DNA Gyrase/metabolism , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Drug Resistance, Bacterial , Etoposide/pharmacology , Fluoroquinolones/pharmacology , Models, Molecular , Molecular Structure , Moxifloxacin , Staphylococcus aureus/chemistry , Staphylococcus aureus/drug effects , Topoisomerase II Inhibitors/pharmacology
12.
Acta Crystallogr F Struct Biol Commun ; 71(Pt 10): 1242-6, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26457513

ABSTRACT

Fluoroquinolone drugs such as moxifloxacin kill bacteria by stabilizing the normally transient double-stranded DNA breaks created by bacterial type IIA topoisomerases. Previous crystal structures of Staphylococcus aureus DNA gyrase with asymmetric DNAs have had static disorder (with the DNA duplex observed in two orientations related by the pseudo-twofold axis of the complex). Here, 20-base-pair DNA homoduplexes were used to obtain crystals of covalent DNA-cleavage complexes of S. aureus DNA gyrase. Crystals with QPT-1, moxifloxacin or etoposide diffracted to between 2.45 and 3.15 Šresolution. A G/T mismatch introduced at the ends of the DNA duplexes facilitated the crystallization of slightly asymmetric complexes of the inherently flexible DNA-cleavage complexes.


Subject(s)
DNA Cleavage , DNA Gyrase/chemistry , Etoposide/chemistry , Fluoroquinolones/chemistry , Heterocyclic Compounds, 4 or More Rings/chemistry , Spiro Compounds/chemistry , Staphylococcus aureus/enzymology , Base Sequence , Crystallization , Crystallography, X-Ray , Molecular Sequence Data , Moxifloxacin
13.
Nat Chem Biol ; 11(3): 189-91, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25622091

ABSTRACT

PAD4 has been strongly implicated in the pathogenesis of autoimmune, cardiovascular and oncological diseases through clinical genetics and gene disruption in mice. New selective PAD4 inhibitors binding a calcium-deficient form of the PAD4 enzyme have validated the critical enzymatic role of human and mouse PAD4 in both histone citrullination and neutrophil extracellular trap formation for, to our knowledge, the first time. The therapeutic potential of PAD4 inhibitors can now be explored.


Subject(s)
Benzimidazoles/pharmacology , Enzyme Inhibitors/pharmacology , Hydrolases/antagonists & inhibitors , Neutrophils/drug effects , Animals , Benzimidazoles/chemical synthesis , Binding, Competitive , Calcium/metabolism , Citrulline/metabolism , Enzyme Inhibitors/chemical synthesis , HEK293 Cells , Histones/metabolism , Humans , In Vitro Techniques , Mice , Models, Molecular , Protein-Arginine Deiminase Type 4 , Protein-Arginine Deiminases , Small Molecule Libraries , Substrate Specificity
14.
ACS Chem Biol ; 10(4): 1043-53, 2015 Apr 17.
Article in English | MEDLINE | ID: mdl-25621824

ABSTRACT

Protein arginine deiminases (PADs) are calcium-dependent histone-modifying enzymes whose activity is dysregulated in inflammatory diseases and cancer. PAD2 functions as an Estrogen Receptor (ER) coactivator in breast cancer cells via the citrullination of histone tail arginine residues at ER binding sites. Although an attractive therapeutic target, the mechanisms that regulate PAD2 activity are largely unknown, especially the detailed role of how calcium facilitates enzyme activation. To gain insights into these regulatory processes, we determined the first structures of PAD2 (27 in total), and through calcium-titrations by X-ray crystallography, determined the order of binding and affinity for the six calcium ions that bind and activate this enzyme. These structures also identified several PAD2 regulatory elements, including a calcium switch that controls proper positioning of the catalytic cysteine residue, and a novel active site shielding mechanism. Additional biochemical and mass-spectrometry-based hydrogen/deuterium exchange studies support these structural findings. The identification of multiple intermediate calcium-bound structures along the PAD2 activation pathway provides critical insights that will aid the development of allosteric inhibitors targeting the PADs.


Subject(s)
Calcium/metabolism , Hydrolases/chemistry , Hydrolases/metabolism , Amino Acid Sequence , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Catalytic Domain , Crystallography, X-Ray , Cysteine/chemistry , Cysteine/metabolism , Deuterium Exchange Measurement , Enzyme Activation , Enzyme Inhibitors/chemistry , Humans , Hydrolases/antagonists & inhibitors , Molecular Docking Simulation , Molecular Sequence Data , Protein Conformation , Protein-Arginine Deiminase Type 2 , Protein-Arginine Deiminases
15.
Cryst Growth Des ; 14(4): 2034-2047, 2014 Apr 02.
Article in English | MEDLINE | ID: mdl-24803849

ABSTRACT

The lipidic mesophase or in meso method for crystallizing membrane proteins has several high profile targets to its credit and is growing in popularity. Despite its success, the method is in its infancy as far as rational crystallogenesis is concerned. Consequently, significant time, effort, and resources are still required to generate structure-grade crystals, especially with a new target type. Therefore, a need exists for crystallogenesis protocols that are effective with a broad range of membrane protein types. Recently, a strategy for crystallizing a prokaryotic α-helical membrane protein, diacylglycerol kinase (DgkA), by the in meso method was reported (Cryst. Growth. Des.2013, 14, 2846-2857). Here, we describe its application to the human α-helical microsomal prostaglandin E2 synthase 1 (mPGES1). While the DgkA strategy proved useful, significant modifications were needed to generate structure-quality crystals of this important therapeutic target. These included protein engineering, using an additive phospholipid in the hosting mesophase, performing multiple rounds of salt screening, and carrying out trials at 4 °C in the presence of a tight binding ligand. The crystallization strategy detailed here should prove useful for generating structures of other integral membrane proteins by the in meso method.

16.
Article in English | MEDLINE | ID: mdl-23722853

ABSTRACT

Mycobacterium tuberculosis DNA gyrase, a nanomachine involved in the regulation of DNA topology, is the only type II topoisomerase present in this organism and hence is the sole target of fluoroquinolones in the treatment of tuberculosis. The ATPase domain provides the energy required for catalysis by ATP hydrolysis. Two constructs corresponding to this 43 kDa domain, Mtb-GyrB47(C1) and Mtb-GyrB47(C2), have been overproduced, purified and crystallized. Diffraction data were collected from three crystal forms. The crystals belonged to space groups P1 and P21 and diffracted to resolutions of 2.9 and 3.3 Å, respectively.


Subject(s)
Adenosine Triphosphatases/chemistry , Bacterial Proteins/chemistry , DNA Gyrase/chemistry , Mycobacterium tuberculosis/enzymology , Adenosine Triphosphatases/isolation & purification , Bacterial Proteins/isolation & purification , Crystallization , Crystallography, X-Ray , DNA Gyrase/isolation & purification
17.
J Med Chem ; 54(1): 78-94, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-21128618

ABSTRACT

A novel series of AMPAR positive modulators is described that were identified by high throughput screening. The molecules of the series have been optimized from a high quality starting point hit to afford excellent developability, tolerability, and efficacy profiles, leading to identification of a clinical candidate. Unusually for an ion channel target, this optimization was integrated with regular generation of ligand-bound crystal structures and uncovered a novel chemotype with a unique and highly conserved mode of interaction via a trifluoromethyl group.


Subject(s)
Indazoles/chemical synthesis , Receptors, AMPA/physiology , Allosteric Regulation , Animals , Calcium/metabolism , Crystallography, X-Ray , Dogs , High-Throughput Screening Assays , Humans , In Vitro Techniques , Indazoles/pharmacokinetics , Indazoles/pharmacology , Ligands , Macaca fascicularis , Male , Microsomes, Liver/metabolism , Models, Molecular , Molecular Conformation , Patch-Clamp Techniques , Protein Multimerization , Rats , Rats, Sprague-Dawley , Receptors, AMPA/chemistry , Recombinant Proteins/chemistry , Solubility , Structure-Activity Relationship , Swine , Swine, Miniature
18.
Nature ; 466(7309): 935-40, 2010 Aug 19.
Article in English | MEDLINE | ID: mdl-20686482

ABSTRACT

Despite the success of genomics in identifying new essential bacterial genes, there is a lack of sustainable leads in antibacterial drug discovery to address increasing multidrug resistance. Type IIA topoisomerases cleave and religate DNA to regulate DNA topology and are a major class of antibacterial and anticancer drug targets, yet there is no well developed structural basis for understanding drug action. Here we report the 2.1 A crystal structure of a potent, new class, broad-spectrum antibacterial agent in complex with Staphylococcus aureus DNA gyrase and DNA, showing a new mode of inhibition that circumvents fluoroquinolone resistance in this clinically important drug target. The inhibitor 'bridges' the DNA and a transient non-catalytic pocket on the two-fold axis at the GyrA dimer interface, and is close to the active sites and fluoroquinolone binding sites. In the inhibitor complex the active site seems poised to cleave the DNA, with a single metal ion observed between the TOPRIM (topoisomerase/primase) domain and the scissile phosphate. This work provides new insights into the mechanism of topoisomerase action and a platform for structure-based drug design of a new class of antibacterial agents against a clinically proven, but conformationally flexible, enzyme class.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , DNA Gyrase/chemistry , Quinolines/chemistry , Quinolines/pharmacology , Staphylococcus aureus/enzymology , Topoisomerase II Inhibitors , Anti-Bacterial Agents/metabolism , Apoenzymes/chemistry , Apoenzymes/metabolism , Arginine/metabolism , Aspartic Acid/metabolism , Binding Sites , Catalytic Domain , Ciprofloxacin/chemistry , Ciprofloxacin/metabolism , Crystallography, X-Ray , DNA/chemistry , DNA/metabolism , DNA Cleavage , DNA Gyrase/metabolism , DNA, Superhelical/chemistry , DNA, Superhelical/metabolism , Drug Design , Drug Resistance , Escherichia coli/enzymology , Manganese/metabolism , Models, Molecular , Protein Conformation , Quinolines/metabolism , Quinolones/chemistry , Quinolones/metabolism , Structure-Activity Relationship
19.
Nat Struct Mol Biol ; 17(9): 1152-3, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20802486

ABSTRACT

Quinolone antibacterials have been used to treat bacterial infections for over 40 years. A crystal structure of moxifloxacin in complex with Acinetobacter baumannii topoisomerase IV now shows the wedge-shaped quinolone stacking between base pairs at the DNA cleavage site and binding conserved residues in the DNA cleavage domain through chelation of a noncatalytic magnesium ion. This provides a molecular basis for the quinolone inhibition mechanism, resistance mutations and invariant quinolone antibacterial structural features.


Subject(s)
Acinetobacter baumannii/enzymology , DNA Topoisomerase IV/chemistry , Enzyme Inhibitors/chemistry , Quinolones/chemistry , DNA Topoisomerase IV/pharmacology , Enzyme Inhibitors/pharmacology , Models, Molecular , Protein Structure, Quaternary , Protein Structure, Tertiary , Quinolones/pharmacology
20.
J Med Chem ; 53(15): 5801-12, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-20614889

ABSTRACT

A series of AMPA receptor positive allosteric modulators has been optimized from poorly penetrant leads to identify molecules with excellent preclinical pharmacokinetics and CNS penetration. These discoveries led to 17i, a potent, efficacious CNS penetrant molecule with an excellent pharmacokinetic profile across preclinical species, which is well tolerated and is also orally bioavailable in humans.


Subject(s)
Indenes/chemical synthesis , Pyridines/chemical synthesis , Receptors, AMPA/physiology , Sulfonamides/chemical synthesis , Administration, Oral , Allosteric Regulation , Animals , Biological Availability , Blood Proteins/metabolism , Blood-Brain Barrier/metabolism , Callithrix , Cell Line , Crystallography, X-Ray , Dogs , Humans , Indenes/pharmacokinetics , Indenes/pharmacology , Macaca fascicularis , Male , Microsomes, Liver/metabolism , Models, Molecular , Protein Binding , Protein Structure, Tertiary , Pyridines/pharmacokinetics , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Species Specificity , Stereoisomerism , Structure-Activity Relationship , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL