ABSTRACT
G-protein-coupled receptors (GPCRs) are essential components of the signalling network throughout the body. To understand the molecular mechanism of G-protein-mediated signalling, solved structures of receptors in inactive conformations and in the active conformation coupled to a G protein are necessary. Here we present the structure of the adenosine A(2A) receptor (A(2A)R) bound to an engineered G protein, mini-Gs, at 3.4 Å resolution. Mini-Gs binds to A(2A)R through an extensive interface (1,048 Å2) that is similar, but not identical, to the interface between Gs and the ß2-adrenergic receptor. The transition of the receptor from an agonist-bound active-intermediate state to an active G-protein-bound state is characterized by a 14 Å shift of the cytoplasmic end of transmembrane helix 6 (H6) away from the receptor core, slight changes in the positions of the cytoplasmic ends of H5 and H7 and rotamer changes of the amino acid side chains Arg3.50, Tyr5.58 and Tyr7.53. There are no substantial differences in the extracellular half of the receptor around the ligand binding pocket. The A(2A)R-mini-Gs structure highlights both the diversity and similarity in G-protein coupling to GPCRs and hints at the potential complexity of the molecular basis for G-protein specificity.
Subject(s)
Heterotrimeric GTP-Binding Proteins/metabolism , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/metabolism , Adenosine A2 Receptor Agonists/metabolism , Amino Acid Sequence , Binding Sites , Crystallization , Crystallography, X-Ray , Cytoplasm/metabolism , Heterotrimeric GTP-Binding Proteins/chemistry , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Protein Conformation , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/metabolism , Substrate SpecificityABSTRACT
The activation process of G protein-coupled receptors (GPCRs) has been extensively studied, both experimentally and computationally. In particular, Molecular Dynamics (MD) simulations have proven useful in exploring GPCR conformational space. The typical behaviour of class A GPCRs, when subjected to unbiased MD simulations from their crystallized inactive state, is to fluctuate between inactive and intermediate(s) conformations, even with bound agonist. Fully active conformation(s) are rarely stabilized unless a G protein is also bound. Despite several crystal structures of the adenosine A2a receptor (A2aR) having been resolved in complex with co-crystallized agonists and Gs protein, its agonist-mediated activation process is still not completely understood. In order to thoroughly examine the conformational landscape of A2aR activation, we performed unbiased microsecond-length MD simulations in quadruplicate, starting from the inactive conformation either in apo or with bound agonists: endogenous adenosine or synthetic NECA, embedded in two homogeneous phospholipid membranes: 1,2-dioleoyl-sn-glycerol-3-phosphoglycerol (DOPG) or 1,2-dioleoyl-sn-glycerol-3-phosphocholine (DOPC). In DOPC with bound adenosine or NECA, we observe transition to an intermediate receptor conformation consistent with the known adenosine-bound crystal state. In apo state in DOPG, two different intermediate conformations are obtained. One is similar to that observed with bound adenosine in DOPC, while the other is closer to the active state but not yet fully active. Exclusively, in DOPG with bound adenosine or NECA, we reproducibly identify receptor conformations with fully active features, which are able to dock Gs protein. These different receptor conformations can be attributed to the action/absence of agonist and phospholipid-mediated allosteric effects on the intracellular side of the receptor.
Subject(s)
Adenosine A2 Receptor Agonists , Phospholipids , Receptor, Adenosine A2A , Adenosine/chemistry , Adenosine/metabolism , Adenosine A2 Receptor Agonists/chemistry , Adenosine A2 Receptor Agonists/metabolism , Binding Sites , Humans , Molecular Dynamics Simulation , Phosphatidylcholines , Phosphatidylglycerols , Phospholipids/chemistry , Phospholipids/metabolism , Protein Conformation , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/metabolismABSTRACT
G-protein coupled receptors (GPCRs) are known for their low stability and large conformational changes upon transitions between multiple states. A widely used method for stabilizing these receptors is to make chimeric receptors by fusing soluble proteins (i.e., fusion partner proteins) into the intracellular loop 3 (ICL3) connecting the transmembrane helices 5 and 6 (TM5 and TM6). However, this fusion approach requires experimental trial and error to identify appropriate soluble proteins, residue positions, and linker lengths for making the fusion. Moreover, this approach has not provided state-targeting stabilization of GPCRs. Here, to rationally stabilize a class A GPCR, adenosine A2A receptor (A2AR) in a target state, we carried out the custom-made de novo design of α-helical fusion partner proteins, which can fix the conformation of TM5 and TM6 to that in an inactive state of A2AR through straight helical connections without any kinks or intervening loops. The chimeric A2AR fused with one of the designs (FiX1) exhibited increased thermal stability. Moreover, compared with the wild type, the binding affinity of the chimera against the agonist NECA was significantly decreased, whereas that against the inverse agonist ZM241385 was similar, indicating that the inactive state was selectively stabilized. Our strategy contributes to the rational state-targeting stabilization of GPCRs.
Subject(s)
Adenosine A2 Receptor Agonists/metabolism , Proteins/metabolism , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/metabolism , Recombinant Fusion Proteins/metabolism , Adenosine/metabolism , Adenosine A2 Receptor Agonists/chemistry , Humans , Ligands , Models, Molecular , Molecular Dynamics Simulation , Protein Conformation, alpha-Helical , Proteins/chemistry , Recombinant Fusion Proteins/chemistryABSTRACT
Adenosine A2A receptor (A2AR)-dopamine D2 receptor (D2R) heteromers are key modulators of striatal neuronal function. It has been suggested that the psychostimulant effects of caffeine depend on its ability to block an allosteric modulation within the A2AR-D2R heteromer, by which adenosine decreases the affinity and intrinsic efficacy of dopamine at the D2R. We describe novel unsuspected allosteric mechanisms within the heteromer by which not only A2AR agonists, but also A2AR antagonists, decrease the affinity and intrinsic efficacy of D2R agonists and the affinity of D2R antagonists. Strikingly, these allosteric modulations disappear on agonist and antagonist coadministration. This can be explained by a model that considers A2AR-D2R heteromers as heterotetramers, constituted by A2AR and D2R homodimers, as demonstrated by experiments with bioluminescence resonance energy transfer and bimolecular fluorescence and bioluminescence complementation. As predicted by the model, high concentrations of A2AR antagonists behaved as A2AR agonists and decreased D2R function in the brain.
Subject(s)
Corpus Striatum/metabolism , Protein Multimerization , Receptor, Adenosine A2A/metabolism , Receptors, Dopamine D2/metabolism , Adenosine A2 Receptor Agonists/metabolism , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/metabolism , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Binding, Competitive/drug effects , Bioluminescence Resonance Energy Transfer Techniques , CHO Cells , Cricetinae , Cricetulus , Dopamine Agonists/metabolism , Dopamine Agonists/pharmacology , Dopamine D2 Receptor Antagonists/metabolism , Dopamine D2 Receptor Antagonists/pharmacology , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Kinetics , Male , Microscopy, Confocal , Protein Binding/drug effects , Rats, Sprague-Dawley , Receptor, Adenosine A2A/chemistry , Receptors, Dopamine D2/chemistry , Sheep , Time FactorsABSTRACT
Adenosine receptors and ß-adrenoceptors are G-protein-coupled receptors (GPCRs) that activate intracellular G proteins on binding the agonists adenosine or noradrenaline, respectively. GPCRs have similar structures consisting of seven transmembrane helices that contain well-conserved sequence motifs, indicating that they are probably activated by a common mechanism. Recent structures of ß-adrenoceptors highlight residues in transmembrane region 5 that initially bind specifically to agonists rather than to antagonists, indicating that these residues have an important role in agonist-induced activation of receptors. Here we present two crystal structures of the thermostabilized human adenosine A(2A) receptor (A(2A)R-GL31) bound to its endogenous agonist adenosine and the synthetic agonist NECA. The structures represent an intermediate conformation between the inactive and active states, because they share all the features of GPCRs that are thought to be in a fully activated state, except that the cytoplasmic end of transmembrane helix 6 partially occludes the G-protein-binding site. The adenine substituent of the agonists binds in a similar fashion to the chemically related region of the inverse agonist ZM241385 (ref. 8). Both agonists contain a ribose group, not found in ZM241385, which extends deep into the ligand-binding pocket where it makes polar interactions with conserved residues in H7 (Ser 277(7.42) and His 278(7.43); superscripts refer to Ballesteros-Weinstein numbering) and non-polar interactions with residues in H3. In contrast, the inverse agonist ZM241385 does not interact with any of these residues and comparison with the agonist-bound structures indicates that ZM241385 sterically prevents the conformational change in H5 and therefore it acts as an inverse agonist. Comparison of the agonist-bound structures of A(2A)R with the agonist-bound structures of ß-adrenoceptors indicates that the contraction of the ligand-binding pocket caused by the inward motion of helices 3, 5 and 7 may be a common feature in the activation of all GPCRs.
Subject(s)
Adenosine A2 Receptor Agonists/metabolism , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/metabolism , Adenosine/chemistry , Adenosine/metabolism , Adenosine/pharmacology , Adenosine A2 Receptor Agonists/pharmacology , Adenosine-5'-(N-ethylcarboxamide)/chemistry , Adenosine-5'-(N-ethylcarboxamide)/metabolism , Adenosine-5'-(N-ethylcarboxamide)/pharmacology , Animals , Binding Sites , CHO Cells , Cricetinae , Cricetulus , Crystallography, X-Ray , Drug Inverse Agonism , Humans , Ligands , Models, Molecular , Molecular Conformation , Triazines/metabolism , Triazines/pharmacology , Triazoles/metabolism , Triazoles/pharmacologyABSTRACT
Rheumatoid arthritis (RA), ankylosing spondylitis (AS) and psoriatic arthritis (PsA) are chronic inflammatory rheumatic diseases that affect joints, causing debilitating pain and disability. Adenosine receptors (ARs) play a key role in the mechanism of inflammation, and the activation of A2A and A3AR subtypes is often associated with a reduction of the inflammatory status. The aim of this study was to investigate the involvement of ARs in patients suffering from early-RA (ERA), RA, AS and PsA. Messenger RNA (mRNA) analysis and saturation binding experiments indicated an upregulation of A2A and A3ARs in lymphocytes obtained from patients when compared with healthy subjects. A2A and A3AR agonists inhibited nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) activation and reduced inflammatory cytokines release, such as tumor necrosis factor-α (TNF-α), interleukin (IL)-1ß and IL-6. Moreover, A2A and A3AR activation mediated a reduction of metalloproteinases (MMP)-1 and MMP-3. The effect of the agonists was abrogated by selective antagonists demonstrating the direct involvement of these receptor subtypes. Taken together, these data confirmed the involvement of ARs in chronic autoimmune rheumatic diseases highlighting the possibility to exploit A2A and A3ARs as therapeutic targets, with the aim to limit the inflammatory responses usually associated with RA, AS and PsA.
Subject(s)
Arthritis, Psoriatic/pathology , Arthritis, Rheumatoid/pathology , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A3/metabolism , Spondylitis, Ankylosing/pathology , Adenosine/analogs & derivatives , Adenosine/chemistry , Adenosine/metabolism , Adenosine A2 Receptor Agonists/chemistry , Adenosine A2 Receptor Agonists/metabolism , Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/metabolism , Adenosine A3 Receptor Agonists/chemistry , Adenosine A3 Receptor Agonists/metabolism , Adenosine A3 Receptor Antagonists/chemistry , Adenosine A3 Receptor Antagonists/metabolism , Arthritis, Psoriatic/metabolism , Arthritis, Rheumatoid/metabolism , Case-Control Studies , Cytokines/metabolism , Female , Humans , Kinetics , Lymphocytes/metabolism , Male , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 3/metabolism , Middle Aged , NF-kappa B/metabolism , Phenethylamines/chemistry , Phenethylamines/metabolism , Pyrazoles/chemistry , Pyrazoles/metabolism , Pyrimidines/chemistry , Pyrimidines/metabolism , RNA, Messenger/metabolism , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A3/genetics , Spondylitis, Ankylosing/metabolismABSTRACT
Dynamics and functions of G-protein coupled receptors (GPCRs) are accurately regulated by the type of ligands that bind to the orthosteric or allosteric binding sites. To glean the structural and dynamical origin of ligand-dependent modulation of GPCR activity, we performed total ~ 5 µsec molecular dynamics simulations of A2A adenosine receptor (A2AAR) in its apo, antagonist-bound, and agonist-bound forms in an explicit water and membrane environment, and examined the corresponding dynamics and correlation between the 10 key structural motifs that serve as the allosteric hotspots in intramolecular signaling network. We dubbed these 10 structural motifs "binary switches" as they display molecular interactions that switch between two distinct states. By projecting the receptor dynamics on these binary switches that yield 2(10) microstates, we show that (i) the receptors in apo, antagonist-bound, and agonist-bound states explore vastly different conformational space; (ii) among the three receptor states the apo state explores the broadest range of microstates; (iii) in the presence of the agonist, the active conformation is maintained through coherent couplings among the binary switches; and (iv) to be most specific, our analysis shows that W246, located deep inside the binding cleft, can serve as both an agonist sensor and actuator of ensuing intramolecular signaling for the receptor activation. Finally, our analysis of multiple trajectories generated by inserting an agonist to the apo state underscores that the transition of the receptor from inactive to active form requires the disruption of ionic-lock in the DRY motif.
Subject(s)
Adenosine A2 Receptor Agonists/metabolism , Adenosine A2 Receptor Antagonists/metabolism , Molecular Dynamics Simulation , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/metabolism , Adenosine A2 Receptor Agonists/chemistry , Adenosine A2 Receptor Antagonists/chemistry , Hydrogen Bonding , Protein SubunitsABSTRACT
Sepsis is a generalized infection accompanied by response of the body that manifests in a clinical and laboratory syndrome, namely, in the systemic inflammatory response syndrome (SIRS) from the organism to the infection. Although sepsis is a widespread and life-threatening disease, the assortment of drugs for its treatment is mostly limited by antibiotics. Therefore, the search for new cellular targets for drug therapy of sepsis is an urgent task of modern medicine and pharmacology. One of the most promising targets is the adenosine A(2A) receptor (A(2A)AR). The activation of this receptor, which is mediated by extracellular adenosine, manifests in almost all types of immune cells (lymphocytes, monocytes, macrophages, and dendritic cells) and results in reducing the severity of inflammation and reperfusion injury in various tissues. The activation of adenosine A(2A) receptor inhibits the proliferation of T cells and production of proinflammatory cytokines, which contributes to the activation of the synthesis of anti-inflammatory cytokines, thereby suppressing the systemic response. For this reason, various selective A(2A)AR agonists and antagonists may be considered to be drug candidates for sepsis pharmacotherapy. Nevertheless, they remain only efficient ligands and objects of pre-clinical and clinical trials. This review examines the molecular mechanisms of inflammatory response in sepsis and the structure and functions of A(2A)AR and its role in the pathogenesis of sepsis, as well as examples of using agonists and antagonists of this receptor for the treatment of SIRS and sepsis.
Subject(s)
Adenosine A2 Receptor Agonists/metabolism , Molecular Targeted Therapy , Receptor, Adenosine A2A/metabolism , Sepsis/drug therapy , Adenosine/therapeutic use , Adenosine A2 Receptor Agonists/therapeutic use , Cell Proliferation/drug effects , Humans , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Lymphocytes/drug effects , Lymphocytes/immunology , Monocytes/drug effects , Monocytes/immunology , Sepsis/genetics , Sepsis/pathologyABSTRACT
The determination of accurate binding affinities is critical in drug discovery and development. Several techniques are available for characterizing the binding of small molecules to soluble proteins. The situation is different for integral membrane proteins. Isothermal chemical denaturation has been shown to be a valuable biophysical method to determine, in a direct and label-free fashion, the binding of ligands to soluble proteins. In this study, the application of isothermal chemical denaturation was applied to an integral membrane protein, the A2a G-protein coupled receptor. Binding affinities for a set of 19 small molecule agonists/antagonists of the A2a receptor were determined and found to be in agreement with data from surface plasmon resonance and radioligand binding assays previously reported in the literature. Therefore, isothermal chemical denaturation expands the available toolkit of biophysical techniques to characterize and study ligand binding to integral membrane proteins, specifically G-protein coupled receptors in vitro.
Subject(s)
Biophysics/methods , Protein Denaturation/drug effects , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/metabolism , Temperature , Adenosine A2 Receptor Agonists/metabolism , Adenosine A2 Receptor Antagonists/metabolism , Guanidine/pharmacology , Ligands , Protein BindingABSTRACT
Adenosine receptors (ARs) have emerged as new drug targets. The majority of data on affinity/potency and selectivity of AR ligands described in the literature has been obtained for the human species. However, preclinical studies are mostly performed in mouse or rat, and standard AR agonists and antagonists are frequently used for studies in rodents without knowing their selectivity in the investigated species. In the present study, we selected a set of frequently used standard AR ligands, 8 agonists and 16 antagonists, and investigated them in radioligand binding studies at all four AR subtypes, A1, A2A, A2B, and A3, of three species, human, rat, and mouse. Recommended, selective agonists include CCPA (for A1AR of rat and mouse), CGS-21680 (for A2A AR of rat), and Cl-IB-MECA (for A3AR of all three species). The functionally selective partial A2B agonist BAY60-6583 was found to additionally bind to A1 and A3AR and act as an antagonist at both receptor subtypes. The antagonists PSB-36 (A1), preladenant (A2A), and PSB-603 (A2B) displayed high selectivity in all three investigated species. MRS-1523 acts as a selective A3AR antagonist in human and rat, but is only moderately selective in mouse. The comprehensive data presented herein provide a solid basis for selecting suitable AR ligands for biological studies.
Subject(s)
Receptors, Purinergic P1/drug effects , Adenosine A1 Receptor Agonists/metabolism , Adenosine A1 Receptor Agonists/pharmacology , Adenosine A1 Receptor Antagonists/metabolism , Adenosine A1 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Agonists/metabolism , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/metabolism , Adenosine A2 Receptor Antagonists/pharmacology , Adenosine A3 Receptor Agonists/metabolism , Adenosine A3 Receptor Agonists/pharmacology , Adenosine A3 Receptor Antagonists/metabolism , Adenosine A3 Receptor Antagonists/pharmacology , Animals , Arrestin/metabolism , Binding, Competitive/drug effects , CHO Cells , Cell Membrane/drug effects , Cell Membrane/metabolism , Cricetinae , Cricetulus , Cyclic AMP/metabolism , DNA, Complementary/drug effects , DNA, Complementary/genetics , Humans , Mice , Rats , Receptor, Adenosine A2A/drug effects , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A2B/drug effects , Receptor, Adenosine A2B/genetics , Receptor, Adenosine A2B/metabolism , Receptors, Purinergic P1/genetics , Receptors, Purinergic P1/metabolism , Species Specificity , Structure-Activity RelationshipABSTRACT
Crystal structures of G protein-coupled receptors (GPCRs) have recently revealed the molecular basis of ligand binding and activation, which has provided exciting opportunities for structure-based drug design. The A2A adenosine receptor (A2AAR) is a promising therapeutic target for cardiovascular diseases, but progress in this area is limited by the lack of novel agonist scaffolds. We carried out docking screens of 6.7 million commercially available molecules against active-like conformations of the A2AAR to investigate whether these structures could guide the discovery of agonists. Nine out of the 20 predicted agonists were confirmed to be A2AAR ligands, but none of these activated the ARs. The difficulties in discovering AR agonists using structure-based methods originated from limited atomic-level understanding of the activation mechanism and a chemical bias toward antagonists in the screened library. In particular, the composition of the screened library was found to strongly reduce the likelihood of identifying AR agonists, which reflected the high ligand complexity required for receptor activation. Extension of this analysis to other pharmaceutically relevant GPCRs suggested that library screening may not be suitable for targets requiring a complex receptor-ligand interaction network. Our results provide specific directions for the future development of novel A2AAR agonists and general strategies for structure-based drug discovery.
Subject(s)
Adenosine A2 Receptor Agonists/chemistry , Drug Discovery/methods , Molecular Docking Simulation , Structure-Activity Relationship , Adenosine A2 Receptor Agonists/metabolism , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/pharmacology , Animals , CHO Cells/drug effects , Cricetulus , Drug Design , Drug Evaluation, Preclinical/methods , Humans , Ligands , Prospective Studies , Protein Conformation , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolismABSTRACT
The plasma level of the regulatory metabolite adenosine increases during the activation of coagulation and inflammation. Here we investigated the effect of adenosine on modulation of thrombin-mediated proinflammatory responses in HUVECs. We found that adenosine inhibits the barrier-disruptive effect of thrombin in HUVECs by a concentration-dependent manner. Analysis of cell surface expression of adenosine receptors revealed that A2A and A2B are expressed at the highest level among the four receptor subtypes (A2B > A2A > A1 > A3 ) on HUVECs. The barrier-protective effect of adenosine in response to thrombin was recapitulated by the A2A specific agonist, CGS 21680, and abrogated both by the siRNA knockdown of the A2A receptor and by the A2A -specific antagonists, ZM-241385 and SCH-58261. The thrombin-induced RhoA activation and its membrane translocation were both inhibited by adenosine in a cAMP-dependent manner, providing a molecular mechanism through which adenosine exerts a barrier-protective function. Adenosine also inhibited thrombin-mediated activation of NF-κB and decreased adhesion of monocytic THP-1 cells to stimulated HUVECs via down-regulation of expression of cell surface adhesion molecules, VCAM-1, ICAM-1, and E-selectin. Moreover, adenosine inhibited thrombin-induced elevated expression of proinflammatory cytokines, IL-6 and HMGB-1; and chemokines, MCP-1, CXCL-1, and CXCL-3. Taken together, these results suggest that adenosine may inhibit thrombin-mediated proinflammatory signaling responses, thereby protecting the endothelium from injury during activation of coagulation and inflammation.
Subject(s)
Adenosine A2 Receptor Agonists/metabolism , Adenosine/metabolism , Blood Coagulation , Human Umbilical Vein Endothelial Cells/metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism , Signal Transduction , Thrombin/metabolism , Adenosine/pharmacology , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Blood Coagulation/drug effects , Capillary Permeability , Cell Adhesion , Cells, Cultured , Coculture Techniques , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , E-Selectin/metabolism , Enzyme Activation , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Humans , Inflammation/blood , Inflammation/immunology , Intercellular Adhesion Molecule-1/metabolism , NF-kappa B/metabolism , Protein Transport , RNA Interference , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A2B/metabolism , Signal Transduction/drug effects , Thrombin/pharmacology , Transfection , Vascular Cell Adhesion Molecule-1/metabolism , rhoA GTP-Binding Protein/metabolismABSTRACT
G protein-coupled receptors (GPCRs) represent the largest family of cell-surface receptors and about one-third of the actual targets of clinically used drugs. Following the progress made in the field of GPCRs structural determination, docking-based screening for novel potent and selective ligands is becoming an increasingly adopted strategy in the drug discovery process. However, this methodology is not yet able to anticipate the "bioactive" binding mode and discern it among other conformations. In the present work, we present a novel approach consisting in the integration of molecular docking and membrane MD simulations with the aim to merge the rapid sampling of ligand poses into in the binding site, typical of docking algorithms, with the thermodynamic accuracy of MD simulations in describing, at the molecular level, the stability a GPCR-ligand complex embedded into explicit lipid-water environment. To validate our approach, we have chosen as a key study the human A(2A) adenosine receptor (hA(2A) AR) and selected four receptor-antagonist complexes and one receptor-agonist complex that have been recently crystallized. In light of the obtained results, we believe that our novel strategy can be extended to other GPCRs and might represent a valuable tool to anticipate the "bioactive" conformation of high-affinity ligands.
Subject(s)
Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/metabolism , Adenosine A2 Receptor Agonists/chemistry , Adenosine A2 Receptor Agonists/metabolism , Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/metabolism , Adenosine-5'-(N-ethylcarboxamide)/chemistry , Adenosine-5'-(N-ethylcarboxamide)/metabolism , Algorithms , Binding Sites , Caffeine/chemistry , Caffeine/metabolism , Computational Biology , Computer Simulation , Crystallography, X-Ray , Humans , Ligands , Models, Molecular , Molecular Dynamics Simulation , Protein Conformation , Static Electricity , Structural Homology, ProteinABSTRACT
We leveraged variable-temperature 19F-NMR spectroscopy to compare the conformational equilibria of the human A2A adenosine receptor (A2AAR), a class A G protein-coupled receptor (GPCR), across a range of temperatures ranging from lower temperatures typically employed in 19F-NMR experiments to physiological temperature. A2AAR complexes with partial agonists and full agonists showed large increases in the population of a fully active conformation with increasing temperature. NMR data measured at physiological temperature were more in line with functional data. This was pronounced for complexes with partial agonists, where the population of active A2AAR was nearly undetectable at lower temperature but became evident at physiological temperature. Temperature-dependent behavior of complexes with either full or partial agonists exhibited a pronounced sensitivity to the specific membrane mimetic employed. Cellular signaling experiments correlated with the temperature-dependent conformational equilibria of A2AAR in lipid nanodiscs but not in some detergents, underscoring the importance of the membrane environment in studies of GPCR function.
Subject(s)
Receptor, Adenosine A2A , Humans , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A2A/chemistry , Temperature , Protein Binding , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Agonists/chemistry , Adenosine A2 Receptor Agonists/metabolism , Nuclear Magnetic Resonance, Biomolecular , Models, Molecular , Protein Conformation , HEK293 CellsABSTRACT
A series of 2-hydrazinyladenosine derivatives was synthesized and investigated in radioligand binding studies for their affinity at the adenosine receptor subtypes with the goal to obtain potent and A(2A)AR selective agonists and to explore the structure-activity relationships of this class of compounds at A(2A)AR. Modifications included introduction of a second sugar moiety at position 2 of adenosine to form new bis-sugar nucleosides and/or modifications of the 2-position linker in different ways. The performed modifications were found to produce compounds with relatively high A(2A)AR affinity and very high selectivity toward A(2A)AR. The most potent bis-sugar nucleoside was obtained with the D-galactose derivative 16 which exhibited a K(i) value of 329 nM at A(2A)AR with marked selectivity against the other AR subtypes. In another set of compounds, compound 3 was modified via replacement of its cyclic structure with mono- and disubstituted phenyl moieties and the resulting hydrazones 10-14 were found to have low nanomolar affinity for A(2A)AR. In addition to 3, compounds 10, 11 and 13 have been identified as the most potent compounds in the present series with K(i) values of 16.1, 24.4, and 12.0 nM, respectively, at rat A(2A)AR. Species differences were tested and found to exist in different rates. Functional properties of the most potent compounds 10, 11, 13 and 16 were assessed showing that the compounds acted as agonists at A(2A)AR.
Subject(s)
Adenosine A2 Receptor Agonists/chemical synthesis , Adenosine/analogs & derivatives , Hydrazones/chemistry , Receptor, Adenosine A2A/metabolism , Adenosine/chemical synthesis , Adenosine/metabolism , Adenosine A2 Receptor Agonists/chemistry , Adenosine A2 Receptor Agonists/metabolism , Animals , Brain/metabolism , Humans , Kinetics , Ligands , Protein Binding , Rats , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/genetics , Recombinant Proteins/agonists , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Structure-Activity RelationshipABSTRACT
Adenosine A(2) receptors have been suggested to modulate tubuloglomerular feedback (TGF) responses by counteracting adenosine A(1) receptor-mediated vasoconstriction, but the mechanisms are unclear. We tested the hypothesis that A(2A) receptor activation blunts TGF by release of nitric oxide in the juxtaglomerular apparatus (JGA). Maximal TGF responses were measured in male Sprague-Dawley rats as changes in proximal stop-flow pressure (ΔP(SF)) in response to increased perfusion of the loop of Henle (0 to 40 nl/min) with artificial tubular fluid (ATF). The maximal TGF response was studied after 5 min intratubular perfusion (10 nl/min) with ATF or ATF + A(2A) receptor agonist (CGS-21680; 10(-7) mol/l). The interaction with nitric oxide synthase (NOS) isoforms was tested by perfusion with a nonselective NOS inhibitor [N(ω)-nitro-L-arginine methyl ester hydrochloride (L-NAME); 10(-3) mol/l] or a selective neuronal NOS (nNOS) inhibitor [N(ω)-propyl-L-arginine (L-NPA); 10(-6) mol/l] alone, and with the A(2A) agonist. Blood pressure, urine flow, and P(SF) at 0 nl/min were similar among the groups. The maximal TGF response (ΔP(SF)) with ATF alone (12.3 ± 0.6 mmHg) was attenuated by selective A(2A) stimulation (9.5 ± 0.4 mmHg). L-NAME enhanced maximal TGF responses (18.9 ± 0.4 mmHg) significantly more than L-NPA (15.2 ± 0.7 mmHg). Stimulation of A(2A) receptors did not influence maximal TGF response during nonselective NOS inhibition (19.0 ± 0.4) but attenuated responses during nNOS inhibition (10.3 ± 0.4 mmHg). In conclusion, adenosine A(2A) receptor activation attenuated TGF responses by stimulation of endothelial NOS (eNOS), presumably in the afferent arteriole. Moreover, NO derived from both eNOS and nNOS in the JGA may blunt TGF responses.
Subject(s)
Kidney Glomerulus/physiology , Kidney Tubules/physiology , Nitric Oxide Synthase Type III/metabolism , Receptor, Adenosine A2A/physiology , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine A2 Receptor Agonists/metabolism , Animals , Arginine/analogs & derivatives , Arginine/metabolism , Arterioles/drug effects , Arterioles/physiology , Blood Pressure/drug effects , Enzyme Inhibitors/metabolism , Feedback, Physiological , Male , NG-Nitroarginine Methyl Ester/metabolism , Nitric Oxide/metabolism , Nitric Oxide/physiology , Phenethylamines/metabolism , Rats , Rats, Sprague-Dawley , Urination/drug effects , Urination/physiologyABSTRACT
Limonene is a major aromatic compound in essential oils extracted from citrus rind. The application of limonene, especially in aromatherapy, has expanded significantly, but its potential effects on cellular metabolism have been elusive. We found that limonene directly binds to the adenosine A(2A) receptor, which may induce sedative effects. Results from an in vitro radioligand binding assay showed that limonene exhibits selective affinity to A(2A) receptors. In addition, limonene increased cytosolic cAMP concentration and induced activation of protein kinase A and phosphorylation of cAMP-response element-binding protein in Chinese hamster ovary cells transfected with the human adenosine A(2A) receptor gene. Limonene also increased cytosolic calcium concentration, which can be achieved by the activation of adenosine A(2A) receptors. These findings suggest that limonene can act as a ligand and an agonist for adenosine A(2A) receptors.
Subject(s)
Adenosine A2 Receptor Agonists/chemistry , Adenosine A2 Receptor Agonists/pharmacology , Cyclohexenes/chemistry , Cyclohexenes/pharmacology , Receptor, Adenosine A2A/chemistry , Terpenes/chemistry , Terpenes/pharmacology , Adenosine A2 Receptor Agonists/metabolism , Animals , Brain/metabolism , CHO Cells , Cricetinae , Cricetulus , Cyclohexenes/metabolism , Humans , Ligands , Limonene , Radioligand Assay , Rats , Receptor, Adenosine A2A/metabolism , Terpenes/metabolismABSTRACT
In this study, we determined the crystal structure of an engineered human adenosine A2A receptor bound to a partial agonist and compared it to structures cocrystallized with either a full agonist or an antagonist/inverse agonist. The interaction between the partial agonist, belonging to a class of dicyanopyridines, and amino acids in the ligand binding pocket inspired us to develop a small library of derivatives and assess their affinity in radioligand binding studies and potency and intrinsic activity in a functional, label-free, intact cell assay. It appeared that some of the derivatives retained the partial agonist profile, whereas other ligands turned into inverse agonists. We rationalized this remarkable behavior with additional computational docking studies.
Subject(s)
Adenosine A2 Receptor Agonists/metabolism , Aminopyridines/metabolism , Pyrimidines/metabolism , Receptor, Adenosine A2A/metabolism , Aminopyridines/chemical synthesis , Animals , Binding Sites , CHO Cells , Cricetulus , Crystallography, X-Ray , Drug Inverse Agonism , Drug Partial Agonism , HEK293 Cells , Humans , Ligands , Molecular Docking Simulation , Protein Binding , Pyrimidines/chemical synthesis , Small Molecule Libraries/metabolismABSTRACT
Molecular dynamics (MD) is the common computational technique for assessing efficacy of GPCR-bound ligands. Agonist efficacy measures the capability of the ligand-bound receptor of reaching the active state in comparison with the free receptor. In this respect, agonists, neutral antagonists and inverse agonists can be considered. A collection of MD simulations of both the ligand-bound and the free receptor are needed to provide reliable conclusions. Variability in the trajectories needs quantification and proper statistical tools for meaningful and non-subjective conclusions. Multiple-factor (time, ligand, lipid) ANOVA with repeated measurements on the time factor is proposed as a suitable statistical method for the analysis of agonist-dependent GPCR activation MD simulations. Inclusion of time factor in the ANOVA model is consistent with the time-dependent nature of MD. Ligand and lipid factors measure agonist and lipid influence on receptor activation. Previously reported MD simulations of adenosine A2a receptor (A2aR) are reanalyzed with this statistical method. TM6-TM3 and TM7-TM3 distances are selected as dependent variables in the ANOVA model. The ligand factor includes the presence or absence of adenosine whereas the lipid factor considers DOPC or DOPG lipids. Statistical analysis of MD simulations shows the efficacy of adenosine and the effect of the membrane lipid composition. Subsequent application of the statistical methodology to NECA A2aR agonist, with resulting P values in consistency with its pharmacological profile, suggests that the method is useful for ligand comparison and potentially for dynamic structure-based virtual screening.
Subject(s)
Adenosine A2 Receptor Agonists/metabolism , Adenosine/metabolism , Molecular Dynamics Simulation , Protein Conformation , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/metabolism , Binding Sites , Humans , Ligands , Protein BindingABSTRACT
Acute lung injury (ALI) is characterized by disruption of the alveolar-capillary membrane resulting in pulmonary edema and accumulation of associated proteinaceous alveolar exudate. Initiation of ALI upregulates tumor necrosis factor-α (TNF-α), which activates nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinases (MAPK) that induce various pro-inflammatory mediators. Polydexyribonucleotide (PDRN) is an adenosine A2A receptor agonist that exerts anti-inflammatory effects by suppressing the production of pro-inflammatory cytokines and apoptosis. We investigated the therapeutic efficiency of PDRN on ALI induced by lipopolysaccharide (LPS) in rats. ALI was induced by intratracheal instillation of LPS (5 mg/kg) in 200 µL saline. The PDRN treatment group received a single intraperitoneal injection of 500 µL saline including PDRN (8 mg/kg) 1 h after ALI induction. To confirm the involvement of the adenosine A2A receptor in PDRN, 8 mg/kg 7-dimethyl-1-propargylxanthine (DMPX) was applied with PDRN treatment. Rats were then sacrificed 12 h after PDRN and DMPX treatments. Intratracheal administration of LPS caused lung tissue damage and significantly increased the lung injury scores and levels of pro-inflammatory cytokines, and apoptotic factors. In addition, MAPK/NF-κB signaling factors were increased by ALI initiation. PDRN treatment potently suppressed expressions of MAPK/NF-κB signaling factors compared to the PDRN + DMPX co-treated group. These alterations led to a reduction of pro-inflammatory cytokines, apoptotic factors, and NF-κB and MAPK signaling, which promoted the recovery of damaged lung tissue. PDRN therapy demonstrated therapeutic effects for LPS-induced ALI compared to the non-treated and DMPX-treated groups. Therefore, PDRN may be used as a therapy for initial treatment of ALI.