Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Med Chem ; 65(2): 1567-1584, 2022 01 27.
Article in English | MEDLINE | ID: mdl-34931844

ABSTRACT

The serine/threonine kinase SGK1 is an activator of the ß-catenin pathway and a powerful stimulator of cartilage degradation that is found to be upregulated under genomic control in diseased osteoarthritic cartilage. Today, no oral disease-modifying treatments are available and chronic treatment in this indication sets high requirements for the drug selectivity, pharmacokinetic, and safety profile. We describe the identification of a highly selective druglike 1H-pyrazolo[3,4-d]pyrimidine SGK1 inhibitor 17a that matches both safety and pharmacokinetic requirements for oral dosing. Rational compound design was facilitated by a novel hSGK1 co-crystal structure, and multiple ligand-based computer models were applied to guide the chemical optimization of the compound ADMET and selectivity profiles. Compounds were selected for subchronic proof of mechanism studies in the mouse femoral head cartilage explant model, and compound 17a emerged as a druglike SGK1 inhibitor, with a highly optimized profile suitable for oral dosing as a novel, potentially disease-modifying agent for osteoarthritis.


Subject(s)
Arthritis, Experimental/drug therapy , Disease Models, Animal , Immediate-Early Proteins/antagonists & inhibitors , Microsomes, Liver/drug effects , Osteoarthritis/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrimidines/chemistry , Animals , Arthritis, Experimental/enzymology , Arthritis, Experimental/pathology , Ligands , Male , Mice , Mice, Inbred C57BL , Osteoarthritis/enzymology , Osteoarthritis/pathology , Protein Kinase Inhibitors/chemistry , Rats , Rats, Sprague-Dawley
2.
J Chem Inf Model ; 59(12): 5135-5147, 2019 12 23.
Article in English | MEDLINE | ID: mdl-31697501

ABSTRACT

We here report on nonequilibrium targeted molecular dynamics simulations as a tool for the estimation of protein-ligand unbinding kinetics. Correlating simulations with experimental data from SPR kinetics measurements and X-ray crystallography on two small molecule compound libraries bound to the N-terminal domain of the chaperone Hsp90, we show that the mean nonequilibrium work computed in an ensemble of trajectories of enforced ligand unbinding is a promising predictor for ligand unbinding rates. We furthermore investigate the molecular basis determining unbinding rates within the compound libraries. We propose ligand conformational changes and protein-ligand nonbonded interactions to impact on unbinding rates. Ligands may remain longer at the protein if they exhibit strong electrostatic and/or van der Waals interactions with the target. In the case of ligands with a rigid chemical scaffold that exhibit longer residence times, transient electrostatic interactions with the protein appear to facilitate unbinding. Our results imply that understanding the unbinding pathway and the protein-ligand interactions along this path is crucial for the prediction of small molecule ligands with defined unbinding kinetics.


Subject(s)
Molecular Dynamics Simulation , Proteins/metabolism , Kinetics , Ligands , Protein Binding , Protein Conformation , Proteins/chemistry , Static Electricity
3.
Nat Commun ; 9(1): 4420, 2018 10 24.
Article in English | MEDLINE | ID: mdl-30356040

ABSTRACT

Understanding the structural biology of the insulin receptor and how it signals is of key importance in the development of insulin analogs to treat diabetes. We report here a cryo-electron microscopy structure of a single insulin bound to a physiologically relevant, high-affinity version of the receptor ectodomain, the latter generated through attachment of C-terminal leucine zipper elements to overcome the conformational flexibility associated with ectodomain truncation. The resolution of the cryo-electron microscopy maps is 3.2 Å in the insulin-binding region and 4.2 Å in the membrane-proximal region. The structure reveals how the membrane proximal domains of the receptor come together to effect signalling and how insulin's negative cooperativity of binding likely arises. Our structure further provides insight into the high affinity of certain super-mitogenic insulins. Together, these findings provide a new platform for insulin analog investigation and design.


Subject(s)
Receptor, Insulin/chemistry , Receptor, Insulin/ultrastructure , Cryoelectron Microscopy , Crystallography, X-Ray , Humans , Protein Binding , Protein Conformation , Protein Structure, Secondary , Receptor, Insulin/metabolism , Signal Transduction/physiology
4.
J Chem Theory Comput ; 14(7): 3859-3869, 2018 Jul 10.
Article in English | MEDLINE | ID: mdl-29768913

ABSTRACT

Drug-target residence time (τ), one of the main determinants of drug efficacy, remains highly challenging to predict computationally and, therefore, is usually not considered in the early stages of drug design. Here, we present an efficient computational method, τ-random acceleration molecular dynamics (τRAMD), for the ranking of drug candidates by their residence time and obtaining insights into ligand-target dissociation mechanisms. We assessed τRAMD on a data set of 70 diverse drug-like ligands of the N-terminal domain of HSP90α, a pharmaceutically important target with a highly flexible binding site, obtaining computed relative residence times with an accuracy of about 2.3τ for 78% of the compounds and less than 2.0τ within congeneric series. Analysis of dissociation trajectories reveals features that affect ligand unbinding rates, including transient polar interactions and steric hindrance. These results suggest that τRAMD will be widely applicable as a computationally efficient aid to improving drug residence times during lead optimization.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Binding Sites , Drug Discovery , HSP90 Heat-Shock Proteins/chemistry , Humans , Kinetics , Ligands , Molecular Dynamics Simulation , Protein Binding , Protein Domains
5.
Biochem Pharmacol ; 151: 166-179, 2018 05.
Article in English | MEDLINE | ID: mdl-29102677

ABSTRACT

While equilibrium binding affinities and in vitro functional antagonism of CB1 receptor antagonists have been studied in detail, little is known on the kinetics of their receptor interaction. In this study, we therefore conducted kinetic assays for nine 1-(4,5-diarylthiophene-2-carbonyl)-4-phenylpiperidine-4-carboxamide derivatives and included the CB1 antagonist rimonabant as a comparison. For this we newly developed a dual-point competition association assay with [3H]CP55940 as the radioligand. This assay yielded Kinetic Rate Index (KRI) values from which structure-kinetics relationships (SKR) of hCB1 receptor antagonists could be established. The fast dissociating antagonist 6 had a similar receptor residence time (RT) as rimonabant, i.e. 19 and 14 min, respectively, while the slowest dissociating antagonist (9) had a very long RT of 2222 min, i.e. pseudo-irreversible dissociation kinetics. In functional assays, 9 displayed insurmountable antagonism, while the effects of the shortest RT antagonist 6 and rimonabant were surmountable. Taken together, this study shows that hCB1 receptor antagonists can have very divergent RTs, which are not correlated to their equilibrium affinities. Furthermore, their RTs appear to define their mode of functional antagonism, i.e. surmountable vs. insurmountable. Finally, based on the recently resolved hCB1 receptor crystal structure, we propose that the differences in RT can be explained by a different binding mode of antagonist 9 from short RT antagonists that is able to displace unfavorable water molecules. Taken together, these findings are of importance for future design and evaluation of potent and safe hCB1 receptor antagonists.


Subject(s)
Cannabinoid Receptor Antagonists , Receptor, Cannabinoid, CB1/metabolism , Animals , Binding, Competitive , CHO Cells , Cannabinoid Receptor Antagonists/chemical synthesis , Cannabinoid Receptor Antagonists/chemistry , Cannabinoid Receptor Antagonists/metabolism , Cricetulus , Cyclohexanols/metabolism , Kinetics , Ligands , Protein Binding , Radioligand Assay , Structure-Activity Relationship
6.
J Am Chem Soc ; 135(15): 5819-27, 2013 Apr 17.
Article in English | MEDLINE | ID: mdl-23565800

ABSTRACT

A key component to success in structure-based drug design is reliable information on protein-ligand interactions. Recent development in NMR techniques has accelerated this process by overcoming some of the limitations of X-ray crystallography and computational protein-ligand docking. In this work we present a new scoring protocol based on NMR-derived interligand INPHARMA NOEs to guide the selection of computationally generated docking modes. We demonstrate the performance in a range of scenarios, encompassing traditionally difficult cases such as docking to homology models and ligand dependent domain rearrangements. Ambiguities associated with sparse experimental information are lifted by searching a consensus solution based on simultaneously fitting multiple ligand pairs. This study provides a previously unexplored integration between molecular modeling and experimental data, in which interligand NOEs represent the key element in the rescoring algorithm. The presented protocol should be widely applicable for protein-ligand docking also in a different context from drug design and highlights the important role of NMR-based approaches to describe intermolecular ligand-receptor interactions.


Subject(s)
Drug Design , Molecular Docking Simulation , Animals , Cricetinae , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Ligands , Magnetic Resonance Spectroscopy , Protein Conformation , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology
7.
J Biol Chem ; 285(34): 26628-40, 2010 Aug 20.
Article in English | MEDLINE | ID: mdl-20547770

ABSTRACT

Fibroblast growth factor (FGF) signaling regulates mammalian development and metabolism, and its dysregulation is implicated in many inherited and acquired diseases, including cancer. Heparan sulfate glycosaminoglycans (HSGAGs) are essential for FGF signaling as they promote FGF.FGF receptor (FGFR) binding and dimerization. Using novel organic synthesis protocols to prepare homogeneously sulfated heparin mimetics (HM), including hexasaccharide (HM(6)), octasaccharide (HM(8)), and decasaccharide (HM(10)), we tested the ability of these HM to support FGF1 and FGF2 signaling through FGFR4. Biological assays show that both HM(8) and HM(10) are significantly more potent than HM(6) in promoting FGF2-mediated FGFR4 signaling. In contrast, all three HM have comparable activity in promoting FGF1.FGFR4 signaling. To understand the molecular basis for these differential activities in FGF1/2.FGFR4 signaling, we used NMR spectroscopy, isothermal titration calorimetry, and size-exclusion chromatography to characterize binding interactions of FGF1/2 with the isolated Ig-domain 2 (D2) of FGFR4 in the presence of HM, and binary interactions of FGFs and D2 with HM. Our data confirm the existence of both a secondary FGF1.FGFR4 interaction site and a direct FGFR4.FGFR4 interaction site thus supporting the formation of the symmetric mode of FGF.FGFR dimerization in solution. Moreover, our results show that the observed higher activity of HM(8) relative to HM(6) in stimulating FGF2.FGFR4 signaling correlates with the higher affinity of HM(8) to bind and dimerize FGF2. Notably FGF2.HM(8) exhibits pronounced positive binding cooperativity. Based on our findings we propose a refined symmetric FGF.FGFR dimerization model, which incorporates the differential ability of HM to dimerize FGFs.


Subject(s)
Fibroblast Growth Factors/metabolism , Heparin/analogs & derivatives , Oligosaccharides/pharmacology , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Animals , Binding Sites , Cell Line , Humans , Mice , Multiprotein Complexes/biosynthesis , Oligosaccharides/chemistry , Protein Binding , Protein Multimerization , Structure-Activity Relationship
9.
Biochemistry ; 47(16): 4683-91, 2008 Apr 22.
Article in English | MEDLINE | ID: mdl-18373353

ABSTRACT

Glycogen phosphorylase (GP) is a validated target for the treatment of type 2 diabetes. Here we describe highly potent GP inhibitors, AVE5688, AVE2865, and AVE9423. The first two compounds are optimized members of the acyl urea series. The latter represents a novel quinolone class of GP inhibitors, which is introduced in this study. In the enzyme assay, both inhibitor types compete with the physiological activator AMP and act synergistically with glucose. Isothermal titration calorimetry (ITC) shows that the compounds strongly bind to nonphosphorylated, inactive GP (GPb). Binding to phosphorylated, active GP (GPa) is substantially weaker, and the thermodynamic profile reflects a coupled transition to the inactive (tense) conformation. Crystal structures confirm that the three inhibitors bind to the AMP site of tense state GP. These data provide the first direct evidence that acyl urea and quinolone compounds are allosteric inhibitors that selectively bind to and stabilize the inactive conformation of the enzyme. Furthermore, ITC reveals markedly different thermodynamic contributions to inhibitor potency that can be related to the binding modes observed in the cocrystal structures. For AVE5688, which occupies only the lower part of the bifurcated AMP site, binding to GPb (Kd = 170 nM) is exclusively enthalpic (Delta H = -9.0 kcal/mol, TDelta S = 0.3 kcal/mol). The inhibitors AVE2865 (Kd = 9 nM, Delta H = -6.8 kcal/mol, TDelta S = 4.2 kcal/mol) and AVE9423 (Kd = 24 nM, Delta H = -5.9 kcal/mol, TDelta S = 4.6 kcal/mol) fully exploit the volume of the binding pocket. Their pronounced binding entropy can be attributed to the extensive displacement of solvent molecules as well as to ionic interactions with the phosphate recognition site.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Glycogen Phosphorylase/antagonists & inhibitors , Glycogen Phosphorylase/metabolism , Thermodynamics , Allosteric Regulation/drug effects , Animals , Buffers , Calorimetry , Glucose/metabolism , Glycogen Phosphorylase/chemistry , Kinetics , Models, Molecular , Molecular Structure , Protein Binding , Rabbits , Temperature , Titrimetry
10.
Acta Crystallogr D Biol Crystallogr ; 60(Pt 9): 1679-82, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15333953

ABSTRACT

APRV (Automatic Processing, Refinement and Visualization) is a new program that enables high-throughput batch processing of crystallographic data. The program combines processing of raw diffraction images, initial structure refinement and visual inspection of resulting electron density into a seamless one-step procedure, during which all relevant parameters are refined automatically. It is controlled by a user-friendly graphical interface, facilitating operation by non-experts.


Subject(s)
Crystallography, X-Ray/statistics & numerical data , Software , Computer Graphics , Crystallization , Crystallography, X-Ray/methods , Electronic Data Processing , Electrons , Internet , Models, Molecular , User-Computer Interface
11.
Structure ; 10(2): 195-204, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11839305

ABSTRACT

The crystal structure of the hypothetical protein MJ1247 from Methanococccus jannaschii at 2 A resolution, a detailed sequence analysis, and biochemical assays infer its molecular function to be 3-hexulose-6-phosphate isomerase (PHI). In the dissimilatory ribulose monophosphate (RuMP) cycle, ribulose-5-phosphate is coupled to formaldehyde by the 3-hexulose-6-phosphate synthase (HPS), yielding hexulose-6-phosphate, which is then isomerized to fructose-6-phosphate by the enzyme 3-hexulose-6-phosphate isomerase. MJ1247 is an alpha/beta structure consisting of a five-stranded parallel beta sheet flanked on both sides by alpha helices, forming a three-layered alpha-beta-alpha sandwich. The fold represents the nucleotide binding motif of a flavodoxin type. MJ1247 is a tetramer in the crystal and in solution and each monomer has a folding similar to the isomerase domain of glucosamine-6-phosphate synthase (GlmS).


Subject(s)
Archaeal Proteins/chemistry , Archaeal Proteins/metabolism , Glucose-6-Phosphate Isomerase/metabolism , Methanococcus/enzymology , Amino Acid Sequence , Bacterial Proteins/chemistry , Binding Sites , Crystallography, X-Ray , Escherichia coli/enzymology , Glucose-6-Phosphate Isomerase/chemistry , Hexosephosphates/metabolism , Models, Molecular , Molecular Sequence Data , Protein Structure, Quaternary , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Stereoisomerism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...