Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
2.
Nat Commun ; 15(1): 2130, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38503739

ABSTRACT

The Epidermal Growth Factor Receptor (EGFR) is frequently found to be mutated in non-small cell lung cancer. Oncogenic EGFR has been successfully targeted by tyrosine kinase inhibitors, but acquired drug resistance eventually overcomes the efficacy of these treatments. Attempts to surmount this therapeutic challenge are hindered by a poor understanding of how and why cancer mutations specifically amplify ligand-independent EGFR auto-phosphorylation signals to enhance cell survival and how this amplification is related to ligand-dependent cell proliferation. Here we show that drug-resistant EGFR mutations manipulate the assembly of ligand-free, kinase-active oligomers to promote and stabilize the assembly of oligomer-obligate active dimer sub-units and circumvent the need for ligand binding. We reveal the structure and assembly mechanisms of these ligand-free, kinase-active oligomers, uncovering oncogenic functions for hitherto orphan transmembrane and kinase interfaces, and for the ectodomain tethered conformation of EGFR. Importantly, we find that the active dimer sub-units within ligand-free oligomers are the high affinity binding sites competent to bind physiological ligand concentrations and thus drive tumor growth, revealing a link with tumor proliferation. Our findings provide a framework for future drug discovery directed at tackling oncogenic EGFR mutations by disabling oligomer-assembling interactions.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Ligands , ErbB Receptors/metabolism , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Drug Resistance, Neoplasm/genetics
3.
J Chem Inf Model ; 64(1): 205-218, 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38150388

ABSTRACT

A metadynamics protocol is presented to characterize the binding and unbinding of peptide ligands to class A G-protein-coupled receptors (GPCRs). The protocol expands on the one previously presented for binding and unbinding small-molecule ligands to class A GPCRs and accounts for the more demanding nature of the peptide binding-unbinding process. It applies to almost all class A GPCRs. Exemplary simulations are described for subtypes Y1R, Y2R, and Y4R of the neuropeptide Y receptor family, vasopressin binding to the vasopressin V2 receptor (V2R), and oxytocin binding to the oxytocin receptor (OTR). Binding free energies and the positions of alternative binding sites are presented and, where possible, compared with the experiment.


Subject(s)
Receptors, G-Protein-Coupled , Vasopressins , Receptors, G-Protein-Coupled/chemistry , Vasopressins/metabolism , Receptors, Oxytocin/chemistry , Receptors, Oxytocin/metabolism , Oxytocin/metabolism , Binding Sites , Ligands
4.
J Am Chem Soc ; 144(2): 935-950, 2022 01 19.
Article in English | MEDLINE | ID: mdl-34989224

ABSTRACT

The cationic porphyrin TMPyP4 is a well-established DNA G-quadruplex (G4) binding ligand that can stabilize different topologies via multiple binding modes. However, TMPyP4 can have both a stabilizing and destabilizing effect on RNA G4 structures. The structural mechanisms that mediate RNA G4 unfolding remain unknown. Here, we report on the TMPyP4-induced RNA G4 unfolding mechanism studied by well-tempered metadynamics (WT-MetaD) with supporting biophysical experiments. The simulations predict a two-state mechanism of TMPyP4 interaction via a groove-bound and a top-face-bound conformation. The dynamics of TMPyP4 stacking on the top tetrad disrupts Hoogsteen H-bonds between guanine bases, resulting in the consecutive TMPyP4 intercalation from top-to-bottom G-tetrads. The results reveal a striking correlation between computational and experimental approaches and validate WT-MetaD simulations as a powerful tool for studying RNA G4-ligand interactions.


Subject(s)
G-Quadruplexes , Ligands , Porphyrins/chemistry , Cations/chemistry , Hydrogen Bonding , Intercalating Agents/chemistry , Molecular Dynamics Simulation , Nucleic Acid Conformation , Thermodynamics
5.
Chem Sci ; 12(27): 9318-9327, 2021 Jul 14.
Article in English | MEDLINE | ID: mdl-34349901

ABSTRACT

Human histone deacetylase 8 (HDAC8) is a key hydrolase in gene regulation and an important drug-target. High-resolution structures of HDAC8 in complex with substrates or inhibitors are available, which have provided insights into the bound state of HDAC8 and its function. Here, using long all-atom unbiased molecular dynamics simulations and Markov state modelling, we show a strong correlation between the conformation of aromatic side chains near the active site and opening and closing of the surrounding functional loops of HDAC8. We also investigated two mutants known to allosterically downregulate the enzymatic activity of HDAC8. Based on experimental data, we hypothesise that I19S-HDAC8 is unable to release the product, whereas both product release and substrate binding are impaired in the S39E-HDAC8 mutant. The presented results deliver detailed insights into the functional dynamics of HDAC8 and provide a mechanism for the substantial downregulation caused by allosteric mutations, including a disease causing one.

6.
J Chem Theory Comput ; 16(7): 4641-4654, 2020 Jul 14.
Article in English | MEDLINE | ID: mdl-32427471

ABSTRACT

Calculating absolute binding free energies is challenging and important. In this paper, we test some recently developed metadynamics-based methods and develop a new combination with a Hamiltonian replica-exchange approach. The methods were tested on 18 chemically diverse ligands with a wide range of different binding affinities to a complex target; namely, human soluble epoxide hydrolase. The results suggest that metadynamics with a funnel-shaped restraint can be used to calculate, in a computationally affordable and relatively accurate way, the absolute binding free energy for small fragments. When used in combination with an optimal pathlike variable obtained using machine learning or with the Hamiltonian replica-exchange algorithm SWISH, this method can achieve reasonably accurate results for increasingly complex ligands, with a good balance of computational cost and speed. An additional benefit of using the combination of metadynamics and SWISH is that it also provides useful information about the role of water in the binding mechanism.


Subject(s)
Epoxide Hydrolases/chemistry , Machine Learning , Algorithms , Drug Design , Epoxide Hydrolases/metabolism , Humans , Ligands , Molecular Dynamics Simulation , Protein Binding , Protein Structure, Tertiary , Thermodynamics
7.
Acc Chem Res ; 53(3): 654-661, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32134250

ABSTRACT

This Account highlights recent advances and discusses major challenges in investigations of cryptic (hidden) binding sites by molecular simulations. Cryptic binding sites are not visible in protein targets crystallized without a ligand and only become visible crystallographically upon binding events. These sites have been shown to be druggable and might provide a rare opportunity to target difficult proteins. However, due to their hidden nature, they are difficult to find through experimental screening. Computational methods based on atomistic molecular simulations remain one of the best approaches to identify and characterize cryptic binding sites. However, not all methods are equally efficient. Some are more apt at quickly probing protein dynamics but do not provide thermodynamic or druggability information, while others that are able to provide such data are demanding in terms of time and resources. Here, we review the recent contributions of mixed-solvent simulations, metadynamics, Markov state models, and other enhanced sampling methods to the field of cryptic site identification and characterization. We discuss how these methods were able to provide precious information on the nature of the site opening mechanisms, to predict previously unknown sites which were used to design new ligands, and to compute the free energy landscapes and kinetics associated with the opening of the sites and the binding of the ligands. We highlight the potential and the importance of such predictions in drug discovery, especially for difficult ("undruggable") targets. We also discuss the major challenges in the field and their possible solutions.


Subject(s)
Molecular Dynamics Simulation , Binding Sites , Drug Discovery , Ligands , Markov Chains , Solvents/chemistry
8.
J Chem Inf Model ; 59(6): 2830-2836, 2019 06 24.
Article in English | MEDLINE | ID: mdl-31125224

ABSTRACT

Adenosine receptors are involved in many pathological conditions and are thus promising drug targets. However, developing drugs that target this GPCR subfamily is a challenging task. A number of drug candidates fail due to lack of selectivity which results in unwanted side effects. The extensive structural similarity of adenosine receptors complicates the design of selective ligands. The problem of selective targeting is a general concern in GPCRs, and in this respect adenosine receptors are a prototypical example. Here we use enhanced sampling simulations to decipher the determinants of selectivity of ligands in A2a and A1 adenosine receptors. Our model shows how small differences in the binding pocket and in the water network around the ligand can be leveraged to achieve selectivity.


Subject(s)
Molecular Dynamics Simulation , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Ligands , Protein Binding , Protein Conformation , Substrate Specificity
9.
Cells ; 8(4)2019 04 05.
Article in English | MEDLINE | ID: mdl-30959819

ABSTRACT

The epidermal growth factor receptor (EGFR) is historically the prototypical receptor tyrosine kinase, being the first cloned and the first where the importance of ligand-induced dimer activation was ascertained. However, many years of structure determination has shown that EGFR is not completely understood. One challenge is that the many structure fragments stored at the PDB only provide a partial view because full-length proteins are flexible entities and dynamics play a key role in their functionality. Another challenge is the shortage of high-resolution data on functionally important higher-order complexes. Still, the interest in the structure/function relationships of EGFR remains unabated because of the crucial role played by oncogenic EGFR mutants in driving non-small cell lung cancer (NSCLC). Despite targeted therapies against EGFR setting a milestone in the treatment of this disease, ubiquitous drug resistance inevitably emerges after one year or so of treatment. The magnitude of the challenge has inspired novel strategies. Among these, the combination of multi-disciplinary experiments and molecular dynamic (MD) simulations have been pivotal in revealing the basic nature of EGFR monomers, dimers and multimers, and the structure-function relationships that underpin the mechanisms by which EGFR dysregulation contributes to the onset of NSCLC and resistance to treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Lung Neoplasms/metabolism , Animals , Glycosylation , Humans , Molecular Dynamics Simulation , Structure-Activity Relationship
10.
J Phys Chem Lett ; 10(8): 1928-1934, 2019 Apr 18.
Article in English | MEDLINE | ID: mdl-30933516

ABSTRACT

Whether recent updates and new releases of atomistic force fields can model the structural and dynamical properties of proteins containing both folded and partially disordered domains is still unclear. To address this fundamental question, we tested eight recently released force fields against our set of nuclear magnetic resonance (NMR) observables for a complex and medically relevant system, the major factor VIII binding region on the von Willebrand factor. This biomedically important region comprises both a folded and a partially structured domain. By using an enhanced sampling technique (temperature replica-exchange molecular dynamics simulations), we find that some force fields indeed rise to the challenge and capture the structural and dynamical features of the NMR ensemble and, therefore, are the appropriate choice for simulations of proteins with partially structured domains. What is more, we show that only such force fields can qualitatively capture the effects of a pathogenic mutation on the structural ensemble.

11.
J Chem Theory Comput ; 15(1): 25-32, 2019 Jan 08.
Article in English | MEDLINE | ID: mdl-30468578

ABSTRACT

Path Collective Variables (PCVs) are a set of path-like variables that have been successfully used to investigate complex chemical and biological processes and compute their associated free energy surfaces and kinetics. Their current implementation relies on general, but at times inefficient, metrics (such as RMSD or DRMSD) to evaluate the distance between the instantaneous conformational state during the simulation and the reference coordinates defining the path. In this work, we present a new algorithm to construct optimal PCVs metrics as linear combinations of different CVs weighted through a spectral gap optimization procedure. The method was tested first on a simple model, trialanine peptide, in vacuo and then on a more complex path of an anticancer inhibitor binding to its pharmacological target. We also compared the results to those obtained with other path-based algorithms. We find that not only our proposed approach is able to automatically select relevant CVs for the PCVs metric but also that the resulting PCVs allow for reconstructing the associated free energy very efficiently. What is more, at difference with other path-based methods, our algorithm is able to explore nonlocally the reaction path space.


Subject(s)
Models, Chemical , Algorithms , Antineoplastic Agents/chemistry , CSK Tyrosine-Protein Kinase , Dasatinib/chemistry , Molecular Conformation , Molecular Dynamics Simulation , Oligopeptides/chemistry , Protein Binding , Protein Kinase Inhibitors/chemistry , Thermodynamics , src-Family Kinases/chemistry
12.
Nat Commun ; 9(1): 4325, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30337523

ABSTRACT

Our current understanding of epidermal growth factor receptor (EGFR) autoinhibition is based on X-ray structural data of monomer and dimer receptor fragments and does not explain how mutations achieve ligand-independent phosphorylation. Using a repertoire of imaging technologies and simulations we reveal an extracellular head-to-head interaction through which ligand-free receptor polymer chains of various lengths assemble. The architecture of the head-to-head interaction prevents kinase-mediated dimerisation. The latter, afforded by mutation or intracellular treatments, splits the autoinhibited head-to-head polymers to form stalk-to-stalk flexible non-extended dimers structurally coupled across the plasma membrane to active asymmetric tyrosine kinase dimers, and extended dimers coupled to inactive symmetric kinase dimers. Contrary to the previously proposed main autoinhibitory function of the inactive symmetric kinase dimer, our data suggest that only dysregulated species bear populations of symmetric and asymmetric kinase dimers that coexist in equilibrium at the plasma membrane under the modulation of the C-terminal domain.


Subject(s)
ErbB Receptors/antagonists & inhibitors , ErbB Receptors/chemistry , Protein Multimerization , Animals , CHO Cells , Cell Membrane/metabolism , Cricetinae , Cricetulus , Extracellular Matrix/metabolism , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Ligands , Models, Biological , Models, Molecular , Photobleaching , Polymers/chemistry , Protein Domains , Protein Kinases/chemistry , Protein Kinases/metabolism
13.
Sci Rep ; 8(1): 12916, 2018 08 27.
Article in English | MEDLINE | ID: mdl-30150677

ABSTRACT

The rise of multi-drug resistance in bacterial pathogens is one of the grand challenges facing medical science. A major concern is the speed of development of ß-lactamase-mediated resistance in Gram-negative species, thus putting at risk the efficacy of the most recently approved antibiotics and inhibitors, including carbapenems and avibactam, respectively. New strategies to overcome resistance are urgently required, which will ultimately be facilitated by a deeper understanding of the mechanisms that regulate the function of ß-lactamases such as the Klebsiella Pneumoniae carbapenemases (KPCs). Using enhanced sampling computational methods together with site-directed mutagenesis, we report the identification of two "hydrophobic networks" in the KPC-2 enzyme, the integrity of which has been found to be essential for protein stability and corresponding resistance. Present throughout the structure, these networks are responsible for the structural integrity and allosteric signaling. Disruption of the networks leads to a loss of the KPC-2 mediated resistance phenotype, resulting in restored susceptibility to different classes of ß-lactam antibiotics including carbapenems and cephalosporins. The "hydrophobic networks" were found to be highly conserved among class-A ß-lactamases, which implies their suitability for exploitation as a potential target for therapeutic intervention.


Subject(s)
Anti-Bacterial Agents/pharmacology , beta-Lactamases/pharmacology , Azabicyclo Compounds/pharmacology , Carbapenems/pharmacology , Drug Resistance, Microbial , Klebsiella pneumoniae/drug effects , Microbial Sensitivity Tests , Protein Structure, Secondary
14.
Nature ; 561(7721): 63-69, 2018 09.
Article in English | MEDLINE | ID: mdl-30158707

ABSTRACT

Glutamine synthetase, encoded by the gene GLUL, is an enzyme that converts glutamate and ammonia to glutamine. It is expressed by endothelial cells, but surprisingly shows negligible glutamine-synthesizing activity in these cells at physiological glutamine levels. Here we show in mice that genetic deletion of Glul in endothelial cells impairs vessel sprouting during vascular development, whereas pharmacological blockade of glutamine synthetase suppresses angiogenesis in ocular and inflammatory skin disease while only minimally affecting healthy adult quiescent endothelial cells. This relies on the inhibition of endothelial cell migration but not proliferation. Mechanistically we show that in human umbilical vein endothelial cells GLUL knockdown reduces membrane localization and activation of the GTPase RHOJ while activating other Rho GTPases and Rho kinase, thereby inducing actin stress fibres and impeding endothelial cell motility. Inhibition of Rho kinase rescues the defect in endothelial cell migration that is induced by GLUL knockdown. Notably, glutamine synthetase palmitoylates itself and interacts with RHOJ to sustain RHOJ palmitoylation, membrane localization and activation. These findings reveal that, in addition to the known formation of glutamine, the enzyme glutamine synthetase shows unknown activity in endothelial cell migration during pathological angiogenesis through RHOJ palmitoylation.


Subject(s)
Endothelial Cells/enzymology , Endothelial Cells/pathology , Glutamate-Ammonia Ligase/metabolism , Glutamine/biosynthesis , Neovascularization, Pathologic , Actins/metabolism , Animals , Cell Movement , Endothelial Cells/metabolism , Female , Glutamate-Ammonia Ligase/deficiency , Glutamate-Ammonia Ligase/genetics , Glutamate-Ammonia Ligase/physiology , HEK293 Cells , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lipoylation , Mice , Palmitic Acid/metabolism , Protein Processing, Post-Translational , Stress Fibers/metabolism , rho GTP-Binding Proteins/chemistry , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism
15.
Biochem J ; 475(11): 1909-1937, 2018 06 06.
Article in English | MEDLINE | ID: mdl-29626155

ABSTRACT

In all living organisms, coenzyme A (CoA) is an essential cofactor with a unique design allowing it to function as an acyl group carrier and a carbonyl-activating group in diverse biochemical reactions. It is synthesized in a highly conserved process in prokaryotes and eukaryotes that requires pantothenic acid (vitamin B5), cysteine and ATP. CoA and its thioester derivatives are involved in major metabolic pathways, allosteric interactions and the regulation of gene expression. A novel unconventional function of CoA in redox regulation has been recently discovered in mammalian cells and termed protein CoAlation. Here, we report for the first time that protein CoAlation occurs at a background level in exponentially growing bacteria and is strongly induced in response to oxidizing agents and metabolic stress. Over 12% of Staphylococcus aureus gene products were shown to be CoAlated in response to diamide-induced stress. In vitro CoAlation of S. aureus glyceraldehyde-3-phosphate dehydrogenase was found to inhibit its enzymatic activity and to protect the catalytic cysteine 151 from overoxidation by hydrogen peroxide. These findings suggest that in exponentially growing bacteria, CoA functions to generate metabolically active thioesters, while it also has the potential to act as a low-molecular-weight antioxidant in response to oxidative and metabolic stress.


Subject(s)
Antioxidants/metabolism , Bacterial Proteins/metabolism , Coenzyme A/metabolism , Staphylococcus aureus/metabolism , Bacterial Proteins/genetics , Coenzyme A/genetics , Diamide/pharmacology , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Oxidation-Reduction , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics
16.
Elife ; 62017 04 26.
Article in English | MEDLINE | ID: mdl-28445123

ABSTRACT

p38α is a Ser/Thr protein kinase involved in a variety of cellular processes and pathological conditions, which makes it a promising pharmacological target. Although the activity of the enzyme is highly regulated, its molecular mechanism of activation remains largely unexplained, even after decades of research. By using state-of-the-art molecular dynamics simulations, we decipher the key elements of the complex molecular mechanism refined by evolution to allow for a fine tuning of p38α kinase activity. Our study describes for the first time the molecular effects of different regulators of the enzymatic activity, and provides an integrative picture of the activation mechanism that explains the seemingly contradictory X-ray and NMR data.


Subject(s)
Enzyme Activation , Molecular Dynamics Simulation , p38 Mitogen-Activated Protein Kinases/chemistry , p38 Mitogen-Activated Protein Kinases/metabolism , Protein Binding , Protein Conformation
17.
J Am Chem Soc ; 138(43): 14257-14263, 2016 11 02.
Article in English | MEDLINE | ID: mdl-27726386

ABSTRACT

Cryptic pockets, that is, sites on protein targets that only become apparent when drugs bind, provide a promising alternative to classical binding sites for drug development. Here, we investigate the nature and dynamical properties of cryptic sites in four pharmacologically relevant targets, while comparing the efficacy of various simulation-based approaches in discovering them. We find that the studied cryptic sites do not correspond to local minima in the computed conformational free energy landscape of the unliganded proteins. They thus promptly close in all of the molecular dynamics simulations performed, irrespective of the force-field used. Temperature-based enhanced sampling approaches, such as Parallel Tempering, do not improve the situation, as the entropic term does not help in the opening of the sites. The use of fragment probes helps, as in long simulations occasionally it leads to the opening and binding to the cryptic sites. Our observed mechanism of cryptic site formation is suggestive of an interplay between two classical mechanisms: induced-fit and conformational selection. Employing this insight, we developed a novel Hamiltonian Replica Exchange-based method "SWISH" (Sampling Water Interfaces through Scaled Hamiltonians), which combined with probes resulted in a promising general approach for cryptic site discovery. We also addressed the issue of "false-positives" and propose a simple approach to distinguish them from druggable cryptic pockets. Our simulations, whose cumulative sampling time was more than 200 µs, help in clarifying the molecular mechanism of pocket formation, providing a solid basis for the choice of an efficient computational method.


Subject(s)
Molecular Dynamics Simulation , Proteins/chemistry , Proteins/metabolism , Binding Sites , Ligands , Molecular Targeted Therapy , Protein Conformation
18.
Cell Chem Biol ; 23(10): 1193-1205, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27693059

ABSTRACT

Allostery is a phenomenon observed in many proteins where binding of a macromolecular partner or a small-molecule ligand at one location leads to specific perturbations at a site not in direct contact with the region where the binding occurs. The list of proteins under allosteric regulation includes AGC protein kinases. AGC kinases have a conserved allosteric site, the phosphoinositide-dependent protein kinase 1 (PDK1)-interacting fragment (PIF) pocket, which regulates protein ATP-binding, activity, and interaction with substrates. In this study, we identify small molecules that bind to the ATP-binding site and affect the PIF pocket of AGC kinase family members, PDK1 and Aurora kinase. We describe the mechanistic details and show that although PDK1 and Aurora kinase inhibitors bind to the conserved ATP-binding site, they differentially modulate physiological interactions at the PIF-pocket site. Our work outlines a strategy for developing bidirectional small-molecule allosteric modulators of protein kinases and other signaling proteins.


Subject(s)
Adenosine Triphosphate/metabolism , Allosteric Regulation/drug effects , Indazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Allosteric Site/drug effects , Aurora Kinases/antagonists & inhibitors , Aurora Kinases/chemistry , Aurora Kinases/metabolism , Binding Sites/drug effects , HEK293 Cells , Humans , Indazoles/chemistry , Molecular Docking Simulation , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Pyrimidines/chemistry , Pyruvate Dehydrogenase Acetyl-Transferring Kinase
19.
Angew Chem Int Ed Engl ; 55(28): 8008-12, 2016 07 04.
Article in English | MEDLINE | ID: mdl-27184628

ABSTRACT

Molecular-dynamics simulations with metadynamics enhanced sampling reveal three distinct binding sites for arginine vasopressin (AVP) within its V2 -receptor (V2 R). Two of these, the vestibule and intermediate sites, block (antagonize) the receptor, and the third is the orthosteric activation (agonist) site. The contacts found for the orthosteric site satisfy all the requirements deduced from mutagenesis experiments. Metadynamics simulations for V2 R and its V1a R-analog give an excellent correlation with experimental binding free energies by assuming that the most stable binding site in the simulations corresponds to the experimental binding free energy in each case. The resulting three-site mechanism separates agonists from antagonists and explains subtype selectivity.


Subject(s)
Antidiuretic Hormone Receptor Antagonists/pharmacology , Receptors, Vasopressin/agonists , Receptors, Vasopressin/metabolism , Antidiuretic Hormone Receptor Antagonists/chemistry , Arginine Vasopressin/chemistry , Arginine Vasopressin/pharmacology , Binding Sites , Humans , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Receptors, Vasopressin/chemistry , Thermodynamics
20.
PLoS Comput Biol ; 11(11): e1004578, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26606374

ABSTRACT

Due to its inhibition of the Abl kinase domain in the BCR-ABL fusion protein, imatinib is strikingly effective in the initial stage of chronic myeloid leukemia with more than 90% of the patients showing complete remission. However, as in the case of most targeted anti-cancer therapies, the emergence of drug resistance is a serious concern. Several drug-resistant mutations affecting the catalytic domain of Abl and other tyrosine kinases are now known. But, despite their importance and the adverse effect that they have on the prognosis of the cancer patients harboring them, the molecular mechanism of these mutations is still debated. Here by using long molecular dynamics simulations and large-scale free energy calculations complemented by in vitro mutagenesis and microcalorimetry experiments, we model the effect of several widespread drug-resistant mutations of Abl. By comparing the conformational free energy landscape of the mutants with those of the wild-type tyrosine kinases we clarify their mode of action. It involves significant and complex changes in the inactive-to-active dynamics and entropy/enthalpy balance of two functional elements: the activation-loop and the conserved DFG motif. What is more the T315I gatekeeper mutant has a significant impact on the binding mechanism itself and on the binding kinetics.


Subject(s)
Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/physiology , Fusion Proteins, bcr-abl/chemistry , Fusion Proteins, bcr-abl/genetics , Imatinib Mesylate/pharmacology , Computational Biology , Drug Resistance, Neoplasm/drug effects , Fusion Proteins, bcr-abl/metabolism , Humans , Imatinib Mesylate/chemistry , Imatinib Mesylate/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL
...