Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Med Chem ; 257: 115419, 2023 Sep 05.
Article in English | MEDLINE | ID: mdl-37301076

ABSTRACT

Development of subtype-selective leads is essential in drug discovery campaigns targeting G protein-coupled receptors (GPCRs). Herein, a structure-based virtual screening approach to rationally design subtype-selective ligands was applied to the A1 and A2A adenosine receptors (A1R and A2AR). Crystal structures of these closely related subtypes revealed a non-conserved subpocket in the binding sites that could be exploited to identify A1R selective ligands. A library of 4.6 million compounds was screened computationally against both receptors using molecular docking and 20 A1R selective ligands were predicted. Of these, seven antagonized the A1R with micromolar activities and several compounds displayed slight selectivity for this subtype. Twenty-seven analogs of two discovered scaffolds were designed, resulting in antagonists with nanomolar potency and up to 76-fold A1R-selectivity. Our results show the potential of structure-based virtual screening to guide discovery and optimization of subtype-selective ligands, which could facilitate the development of safer drugs.


Subject(s)
Adenosine , Purinergic P1 Receptor Antagonists , Purinergic P1 Receptor Antagonists/pharmacology , Purinergic P1 Receptor Antagonists/chemistry , Molecular Docking Simulation , Ligands , Binding Sites , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A1/metabolism , Adenosine A2 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/chemistry
2.
Angew Chem Int Ed Engl ; 62(22): e202218959, 2023 05 22.
Article in English | MEDLINE | ID: mdl-36914577

ABSTRACT

G-protein-coupled receptors (GPCRs) play important roles in physiological processes and are modulated by drugs that either activate or block signaling. Rational design of the pharmacological efficacy profiles of GPCR ligands could enable the development of more efficient drugs, but is challenging even if high-resolution receptor structures are available. We performed molecular dynamics simulations of the ß2 adrenergic receptor in active and inactive conformations to assess if binding free energy calculations can predict differences in ligand efficacy for closely related compounds. Previously identified ligands were successfully classified into groups with comparable efficacy profiles based on the calculated shift in ligand affinity upon activation. A series of ligands were then predicted and synthesized, leading to the discovery of partial agonists with nanomolar potencies and novel scaffolds. Our results demonstrate that free energy simulations enable design of ligand efficacy and the same approach can be applied to other GPCR drug targets.


Subject(s)
Receptors, G-Protein-Coupled , Signal Transduction , Ligands , Receptors, G-Protein-Coupled/metabolism , Molecular Dynamics Simulation , Receptors, Adrenergic , Receptors, Adrenergic, beta-2/chemistry , Protein Conformation
3.
ACS Chem Biol ; 17(10): 2744-2752, 2022 10 21.
Article in English | MEDLINE | ID: mdl-36149353

ABSTRACT

Recently determined structures of class C G protein-coupled receptors (GPCRs) revealed the location of allosteric binding sites and opened new opportunities for the discovery of novel modulators. In this work, molecular docking screens for allosteric modulators targeting the metabotropic glutamate receptor 5 (mGlu5) were performed. The mGlu5 receptor is activated by the main excitatory neurotransmitter of the nervous central system, L-glutamate, and mGlu5 receptor activity can be allosterically modulated by negative or positive allosteric modulators. The mGlu5 receptor is a promising target for the treatment of psychiatric and neurodegenerative diseases, and several allosteric modulators of this GPCR have been evaluated in clinical trials. Chemical libraries containing fragment- (1.6 million molecules) and lead-like (4.6 million molecules) compounds were docked to an allosteric binding site of mGlu5 identified in X-ray crystal structures. Among the top-ranked compounds, 59 fragments and 59 lead-like compounds were selected for experimental evaluation. Of these, four fragment- and seven lead-like compounds were confirmed to bind to the allosteric site with affinities ranging from 0.43 to 8.6 µM, corresponding to a hit rate of 9%. The four compounds with the highest affinities were demonstrated to be negative allosteric modulators of mGlu5 signaling in functional assays. The results demonstrate that virtual screens of fragment- and lead-like chemical libraries have complementary advantages and illustrate how access to high-resolution structures of GPCRs in complex with allosteric modulators can accelerate lead discovery.


Subject(s)
Receptor, Metabotropic Glutamate 5 , Small Molecule Libraries , Receptor, Metabotropic Glutamate 5/metabolism , Allosteric Regulation , Molecular Docking Simulation , Small Molecule Libraries/pharmacology , Ligands , Glutamic Acid , Allosteric Site , Receptors, G-Protein-Coupled
4.
Pharmacol Rev ; 73(4): 527-565, 2021 10.
Article in English | MEDLINE | ID: mdl-34907092

ABSTRACT

G protein-coupled receptors (GPCRs) constitute the largest family of membrane proteins in the human genome and are important therapeutic targets. During the last decade, the number of atomic-resolution structures of GPCRs has increased rapidly, providing insights into drug binding at the molecular level. These breakthroughs have created excitement regarding the potential of using structural information in ligand design and initiated a new era of rational drug discovery for GPCRs. The molecular docking method is now widely applied to model the three-dimensional structures of GPCR-ligand complexes and screen for chemical probes in large compound libraries. In this review article, we first summarize the current structural coverage of the GPCR superfamily and the understanding of receptor-ligand interactions at atomic resolution. We then present the general workflow of structure-based virtual screening and strategies to discover GPCR ligands in chemical libraries. We assess the state of the art of this research field by summarizing prospective applications of virtual screening based on experimental structures. Strategies to identify compounds with specific efficacy and selectivity profiles are discussed, illustrating the opportunities and limitations of the molecular docking method. Our overview shows that structure-based virtual screening can discover novel leads and will be essential in pursuing the next generation of GPCR drugs. SIGNIFICANCE STATEMENT: Extraordinary advances in the structural biology of G protein-coupled receptors have revealed the molecular details of ligand recognition by this large family of therapeutic targets, providing novel avenues for rational drug design. Structure-based docking is an efficient computational approach to identify novel chemical probes from large compound libraries, which has the potential to accelerate the development of drug candidates.


Subject(s)
Receptors, G-Protein-Coupled , Signal Transduction , Binding Sites , Humans , Ligands , Molecular Docking Simulation , Protein Binding , Receptors, G-Protein-Coupled/metabolism
5.
Angew Chem Int Ed Engl ; 60(33): 18022-18030, 2021 08 09.
Article in English | MEDLINE | ID: mdl-33904641

ABSTRACT

Many diseases are polygenic and can only be treated efficiently with drugs that modulate multiple targets. However, rational design of compounds with multi-target profiles is rarely pursued because it is considered too difficult, in particular if the drug must enter the central nervous system. Here, a structure-based strategy to identify dual-target ligands of G-protein-coupled receptors is presented. We use this approach to design compounds that both antagonize the A2A adenosine receptor and activate the D2 dopamine receptor, which have excellent potential as antiparkinson drugs. Atomic resolution models of the receptors guided generation of a chemical library with compounds designed to occupy orthosteric and secondary binding pockets in both targets. Structure-based virtual screens identified ten compounds, of which three had affinity for both targets. One of these scaffolds was optimized to nanomolar dual-target activity and showed the predicted pharmacodynamic effect in a rat model of Parkinsonism.


Subject(s)
Antiparkinson Agents/pharmacology , Drug Design , Receptor, Adenosine A2A/metabolism , Receptors, Dopamine D2/metabolism , Small Molecule Libraries/pharmacology , Animals , Antiparkinson Agents/chemical synthesis , Antiparkinson Agents/chemistry , Drug Evaluation, Preclinical , Humans , Ligands , Molecular Structure , Rats , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry
6.
Bioinformatics ; 35(5): 874-876, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30124779

ABSTRACT

MOTIVATION: Three-dimensional protein structures are important starting points for elucidating protein function and applications like drug design. Computational methods in this area rely on high quality validation datasets which are usually manually assembled. Due to the increase in published structures as well as the increasing demand for specially tailored validation datasets, automatic procedures should be adopted. RESULTS: StructureProfiler is a new tool for automatic, objective and customizable profiling of X-ray protein structures based on the most frequently applied selection criteria currently in use to assemble benchmark datasets. As examples, four dataset configurations (Astex, Iridium, Platinum, combined), all results of the combined tests and the list of all PDB Ids passing the combined criteria set are attached in the Supplementary Material. AVAILABILITY AND IMPLEMENTATION: StructureProfiler is available as part of the ProteinsPlus web service http://proteins.plus and as standalone tool in the NAOMI ChemBio Suite. Dataset updates together with the tool can be found on http://www.zbh.uni-hamburg.de/structureprofiler. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Software , Computational Biology , Drug Design , Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...