Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
J Chem Phys ; 160(15)2024 Apr 21.
Article in English | MEDLINE | ID: mdl-38639317

ABSTRACT

Enhanced sampling algorithms are indispensable when working with highly disconnected multimodal distributions. An important application of these is the conformational exploration of particular internal degrees of freedom of molecular systems. However, despite the existence of many commonly used enhanced sampling algorithms to explore these internal motions, they often rely on system-dependent parameters, which negatively impact efficiency and reproducibility. Here, we present fully adaptive simulated tempering (FAST), a variation of the irreversible simulated tempering algorithm, which continuously optimizes the number, parameters, and weights of intermediate distributions to achieve maximally fast traversal over a space defined by the change in a predefined thermodynamic control variable such as temperature or an alchemical smoothing parameter. This work builds on a number of previously published methods, such as sequential Monte Carlo, and introduces a novel parameter optimization procedure that can, in principle, be used in any expanded ensemble algorithms. This method is validated by being applied on a number of different molecular systems with high torsional kinetic barriers. We also consider two different soft-core potentials during the interpolation procedure and compare their performance. We conclude that FAST is a highly efficient algorithm, which improves simulation reproducibility and can be successfully used in a variety of settings with the same initial hyperparameters.

2.
J Med Chem ; 66(7): 5223-5241, 2023 04 13.
Article in English | MEDLINE | ID: mdl-36972104

ABSTRACT

The NLRP3 inflammasome is a critical component of innate immunity that senses diverse pathogen- and host-derived molecules. However, its aberrant activation has been associated with the pathogenesis of multiple diseases, including cancer. In this study, we designed and synthesized a series of aryl sulfonamide derivatives (ASDs) to inhibit the NLRP3 inflammasome. Among these, compounds 6c, 7n, and 10 specifically inhibited NLRP3 activation at nanomolar concentrations without affecting the activation of the NLRC4 and AIM2 inflammasomes. Furthermore, we demonstrated that these compounds reduce interleukin-1ß (IL-1ß) production in vivo and attenuate melanoma tumor growth. Moreover, metabolic stability in liver microsomes of 6c, 7n, and 10 was studied along with plasma exposure in mice of the most interesting compound 6c. Therefore, we generated potent NLRP3 inflammasome inhibitors, which can be considered in future medicinal chemistry and pharmacological studies aimed at developing a new therapeutic approach for NLRP3 inflammasome-driven cancer.


Subject(s)
Inflammasomes , Neoplasms , Mice , Animals , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Immunity, Innate , Interleukin-1beta/metabolism , Mice, Inbred C57BL
3.
J Chem Theory Comput ; 18(6): 3894-3910, 2022 Jun 14.
Article in English | MEDLINE | ID: mdl-35588256

ABSTRACT

The sampling problem is one of the most widely studied topics in computational chemistry. While various methods exist for sampling along a set of reaction coordinates, many require system-dependent hyperparameters to achieve maximum efficiency. In this work, we present an alchemical variation of adaptive sequential Monte Carlo (SMC), an irreversible importance resampling method that is part of a well-studied class of methods that have been used in various applications but have been underexplored in computational biophysics. Afterward, we apply alchemical SMC on a variety of test cases, including torsional rotations of solvated ligands (butene and a terphenyl derivative), translational and rotational movements of protein-bound ligands, and protein side chain rotation coupled to the ligand degrees of freedom (T4-lysozyme, protein tyrosine phosphatase 1B, and transforming growth factor ß). We find that alchemical SMC is an efficient way to explore targeted degrees of freedom and can be applied to a variety of systems using the same hyperparameters to achieve a similar performance. Alchemical SMC is a promising tool for preparatory exploration of systems where long-timescale sampling of the entire system can be traded off against short-timescale sampling of a particular set of degrees of freedom over a population of conformers.


Subject(s)
Proteins , Ligands , Monte Carlo Method
4.
ACS Cent Sci ; 7(11): 1847-1862, 2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34841058

ABSTRACT

Targeting G protein-coupled receptors (GPCRs) through allosteric sites offers advantages over orthosteric sites in identifying drugs with increased selectivity and potentially reduced side effects. In this study, we developed a probe confined dynamic mapping protocol that allows the prediction of allosteric sites at both the GPCR extracellular and intracellular sides, as well as at the receptor-lipid interface. The applied harmonic wall potential enhanced sampling of probe molecules in a selected area of a GPCR while preventing membrane distortion in molecular dynamics simulations. The specific probes derived from GPCR allosteric ligand structures performed better in allosteric site mapping compared to commonly used cosolvents. The M2 muscarinic, ß2 adrenergic, and P2Y1 purinergic receptors were selected for the protocol's retrospective validation. The protocol was next validated prospectively to locate the binding site of [5-fluoro-4-(hydroxymethyl)-2-methoxyphenyl]-(4-fluoro-1H-indol-1-yl)methanone at the D2 dopamine receptor, and subsequent mutagenesis confirmed the prediction. The protocol provides fast and efficient prediction of key amino acid residues surrounding allosteric sites in membrane proteins and facilitates the structure-based design of allosteric modulators.

5.
J Chem Theory Comput ; 17(3): 1806-1821, 2021 Mar 09.
Article in English | MEDLINE | ID: mdl-33534995

ABSTRACT

Binding free energy calculations using alchemical free energy (AFE) methods are widely considered to be the most rigorous tool in the computational drug discovery arsenal. Despite this, the calculations suffer from accuracy, precision, and reproducibility issues. In this publication, we perform a high-throughput study of more than a thousand AFE calculations, utilizing over 220 µs of total sampling time, on three different protein systems to investigate the impact of the initial crystal structure on the resulting binding free energy values. We also consider the influence of equilibration time and discover that the initial crystal structure can have a significant effect on free energy values obtained at short timescales that can manifest itself as a free energy difference of more than 1 kcal/mol. At longer timescales, these differences are largely overtaken by important rare events, such as torsional ligand motions, typically resulting in a much higher uncertainty in the obtained values. This work emphasizes the importance of rare event sampling and long-timescale dynamics in free energy calculations even for routinely performed alchemical perturbations. We conclude that an optimal protocol should not only concentrate computational resources on achieving convergence in the alchemical coupling parameter (λ) space but also on longer simulations and multiple repeats.

6.
J Comput Aided Mol Des ; 33(11): 983-996, 2019 11.
Article in English | MEDLINE | ID: mdl-31758354

ABSTRACT

We investigated the Gi-coupled A3 adenosine receptor (A3AR) activation mechanism by running 7.2 µs of molecular dynamics (MD) simulations. Based on homology to G protein-coupled receptor (GPCR) structures, three constitutively active mutant (CAM) and the wild-type (WT) A3ARs in the apo form were modeled. Conformational signatures associated with three different receptor states (inactive R, active R*, and bound to Gi protein mimic) were predicted by analyzing and comparing the CAMs with WT receptor and by considering site-directed mutagenesis data. Detected signatures that were correlated with receptor state included: Persistent salt-bridges involving key charged residues for activation (including a novel, putative ionic lock), rotameric state of conserved W6.48, and Na+ ions and water molecules present. Active-coupled state signatures similar to the X-ray structures of ß2 adrenergic receptor-Gs protein and A2AAR-mini-Gs and the recently solved cryo-EM A1AR-Gi complexes were found. Our MD analysis suggests that constitutive activation might arise from the D1073.49-R1083.50 ionic lock destabilization in R and the D1073.49-R1113.53 ionic lock stabilization in R* that presumably lowers the energy barrier associated with an R to R* transition. This study provides new opportunities to understand the underlying interactions of different receptor states of other Gi protein-coupled GPCRs.


Subject(s)
Receptor, Adenosine A3/metabolism , Humans , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Mutation , Protein Conformation , Receptor, Adenosine A3/chemistry , Receptor, Adenosine A3/genetics
7.
J Med Chem ; 62(3): 1502-1522, 2019 02 14.
Article in English | MEDLINE | ID: mdl-30605331

ABSTRACT

(N)-Methanocarba ([3.1.0]bicyclohexyl) adenosines and corresponding ribosides were synthesized to identify novel A1 adenosine receptor (A1AR) agonists for CNS or peripheral applications. Human and mouse AR binding was determined to assess the constrained ring system's A1AR compatibility. N6-Dicyclobutylmethyl ribose agonist (9, MRS7469, >2000-fold selective for A1AR) and known truncated N6-dicyclopropylmethyl methanocarba 7 (MRS5474) were drug-like. The pure diastereoisomer of known riboside 4 displayed high hA1AR selectivity. Methanocarba modification reduced A1AR selectivity of N6-dicyclopropylmethyl and endo-norbornyladenosines but increased ribavirin selectivity. Most analogues tested (ip) were inactive or weak in inducing mouse hypothermia, despite mA1AR full agonism and variable mA3AR efficacy, but strong hypothermia by 9 depended on A1AR, which reflects CNS activity (determined using A1AR or A3AR null mice). Conserved hA1AR interactions were preserved in modeling of 9 and methanocarba equivalent 24 (∼400-fold A1AR-selective). Thus, we identified, and characterized in vivo, ribose and methanocarba nucleosides, including with A1AR-enhancing N6-dicyclobutylmethyl-adenine and 1,2,4-triazole-3-carboxamide (40, MRS7451) nucleobases.


Subject(s)
Adenosine A1 Receptor Agonists/pharmacology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Bridged Bicyclo Compounds/pharmacology , Adenosine/chemical synthesis , Adenosine A1 Receptor Agonists/chemical synthesis , Adenosine A1 Receptor Agonists/pharmacokinetics , Animals , Bridged Bicyclo Compounds/chemical synthesis , Bridged Bicyclo Compounds/pharmacokinetics , CHO Cells , Cricetulus , Drug Design , HEK293 Cells , Humans , Macaca fascicularis , Male , Mice, Inbred C57BL , Molecular Docking Simulation , Molecular Structure , Receptor, Adenosine A1/metabolism , Structure-Activity Relationship
8.
Medchemcomm ; 9(11): 1920-1932, 2018 Nov 01.
Article in English | MEDLINE | ID: mdl-30568760

ABSTRACT

Recognition of nucleosides at adenosine receptors (ARs) is supported by multiple X-ray structures, but the structure of an adenine complex is unknown. We examined the selectivity of predicted A1AR and A3AR adenine antagonists that incorporated known agonist affinity-enhancing N 6 and C2 substituents. Adenines with A1AR-favoring N 6-alkyl, cycloalkyl and arylalkyl substitutions combined with an A3AR-favoring 2-((5-chlorothiophen-2-yl)ethynyl) group were human (h) A3AR-selective, e.g. MRS7497 17 (∼1000-fold over A1AR). In addition, binding selectivity over hA2AAR and hA2BAR and functional A3AR antagonism were demonstrated. 17 was subjected to computational docking and molecular dynamics simulation in a hA3AR homology model to predict interactions. The SAR of nucleoside AR agonists was not recapitulated in adenine AR antagonists, and modeling suggested an alternative, inverted binding mode with the key N2506.55 H-bonding to the adenine N 3 and N 9, instead of N 6 and N 7 as in adenosine agonists.

9.
ACS Omega ; 3(10): 12658-12678, 2018 Oct 31.
Article in English | MEDLINE | ID: mdl-30411015

ABSTRACT

While screening off-target effects of rigid (N)-methanocarba-adenosine 5'-methylamides as A3 adenosine receptor (AR) agonists, we discovered µM binding hits at the δ-opioid receptor (DOR) and translocator protein (TSPO). In an effort to increase OR and decrease AR affinity by structure activity analysis of this series, antagonist activity at κ-(K)OR appeared in 5'-esters (ethyl 24 and propyl 30), which retained TSPO interaction (µM). 7-Deaza modification of C2-(arylethynyl)-5'-esters but not 4'-truncation enhanced KOR affinity (MRS7299 28 and 29, K i ≈ 40 nM), revealed µ-OR and DOR binding, and reduced AR affinity. Molecular docking and dynamics simulations located a putative KOR binding mode consistent with the observed affinities, placing C7 in a hydrophobic region. 3-Deaza modification permitted TSPO but not OR binding, and 1-deaza was permissive to both; ribose-restored analogues were inactive at both. Thus, we have repurposed a known AR nucleoside scaffold for OR antagonism, with a detailed hypothesis for KOR recognition.

10.
J Med Chem ; 61(11): 4860-4882, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29767967

ABSTRACT

The P2Y14 receptor (P2Y14R) mediates inflammatory activity by activating neutrophil motility, but few classes of antagonists are known. We have explored the structure-activity relationship of a 3-(4-phenyl-1 H-1,2,3-triazol-1-yl)-5-(aryl)benzoic acid antagonist scaffold, assisted by docking and molecular dynamics (MD) simulation at a P2Y14R homology model. A computational pipeline using the High Throughput MD Python environment guided the analogue design. Selection of candidates was based upon ligand-protein shape and complementarity and the persistence of ligand-protein interactions over time. Predictions of a favorable substitution of a 5-phenyl group with thiophene and an insertion of a three-methylene spacer between the 5-aromatic and alkyl amino moieties were largely consistent with empirical results. The substitution of a key carboxylate group on the core phenyl ring with tetrazole or truncation of the 5-aryl group reduced affinity. The most potent antagonists, using a fluorescent assay, were a primary 3-aminopropyl congener 20 (MRS4458) and phenyl p-carboxamide 30 (MRS4478).


Subject(s)
Drug Design , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Purinergic P2 Receptor Antagonists/chemistry , Purinergic P2 Receptor Antagonists/pharmacology , Receptors, Purinergic P2/metabolism , Amines/chemistry , Animals , CHO Cells , Cricetulus , Heterocyclic Compounds/metabolism , Heterocyclic Compounds/pharmacokinetics , Humans , Molecular Dynamics Simulation , Protein Conformation , Purinergic P2 Receptor Antagonists/metabolism , Purinergic P2 Receptor Antagonists/pharmacokinetics , Receptors, Purinergic P2/chemistry , Structure-Activity Relationship
11.
Methods Mol Biol ; 1705: 45-72, 2018.
Article in English | MEDLINE | ID: mdl-29188558

ABSTRACT

Recent crystallographic structures of G protein-coupled receptors (GPCRs) have greatly advanced our understanding of the recognition of their diverse agonist and antagonist ligands. We illustrate here how this applies to A2A adenosine receptors (ARs) and to P2Y1 and P2Y12 receptors (P2YRs) for ADP. These X-ray structures have impacted the medicinal chemistry aimed at discovering new ligands for these two receptor families, including receptors that have not yet been crystallized but are closely related to the known structures. In this Chapter, we discuss recent structure-based drug design projects that led to the discovery of: (a) novel A3AR agonists based on a highly rigidified (N)-methanocarba scaffold for the treatment of chronic neuropathic pain and other conditions, (b) fluorescent probes of the ARs and P2Y14R, as chemical tools for structural probing of these GPCRs and for improving assay capabilities, and (c) new more drug-like antagonists of the inflammation-related P2Y14R. We also describe the computationally enabled molecular recognition of positive (for A3AR) and negative (P2Y1R) allosteric modulators that in some cases are shown to be consistent with structure-activity relationship (SAR) data. Thus, computational modeling has become an essential tool for the design of purine receptor ligands.


Subject(s)
Computer Simulation , Drug Design , Ligands , Models, Molecular , Receptors, G-Protein-Coupled/chemistry , Allosteric Regulation/drug effects , Chemistry, Pharmaceutical , Crystallography, X-Ray , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Receptors, G-Protein-Coupled/metabolism , Structure-Activity Relationship
12.
Med Res Rev ; 38(4): 1031-1072, 2018 07.
Article in English | MEDLINE | ID: mdl-28682469

ABSTRACT

The A3 adenosine receptor (A3 AR) subtype is a novel, promising therapeutic target for inflammatory diseases, such as rheumatoid arthritis (RA) and psoriasis, as well as liver cancer. A3 AR is coupled to inhibition of adenylyl cyclase and regulation of mitogen-activated protein kinase (MAPK) pathways, leading to modulation of transcription. Furthermore, A3 AR affects functions of almost all immune cells and the proliferation of cancer cells. Numerous A3 AR agonists, partial agonists, antagonists, and allosteric modulators have been reported, and their structure-activity relationships (SARs) have been studied culminating in the development of potent and selective molecules with drug-like characteristics. The efficacy of nucleoside agonists may be suppressed to produce antagonists, by structural modification of the ribose moiety. Diverse classes of heterocycles have been discovered as selective A3 AR blockers, although with large species differences. Thus, as a result of intense basic research efforts, the outlook for development of A3 AR modulators for human therapeutics is encouraging. Two prototypical selective agonists, N6-(3-Iodobenzyl)adenosine-5'-N-methyluronamide (IB-MECA; CF101) and 2-chloro-N6-(3-iodobenzyl)-adenosine-5'-N-methyluronamide (Cl-IB-MECA; CF102), have progressed to advanced clinical trials. They were found safe and well tolerated in all preclinical and human clinical studies and showed promising results, particularly in psoriasis and RA, where the A3 AR is both a promising therapeutic target and a biologically predictive marker, suggesting a personalized medicine approach. Targeting the A3 AR may pave the way for safe and efficacious treatments for patient populations affected by inflammatory diseases, cancer, and other conditions.


Subject(s)
Adenosine A3 Receptor Agonists/pharmacology , Arthritis, Rheumatoid/drug therapy , Carcinoma, Hepatocellular/drug therapy , Inflammation/drug therapy , Liver Neoplasms/drug therapy , Psoriasis/drug therapy , Receptor, Adenosine A3/metabolism , Allosteric Site , Animals , Clinical Trials as Topic , Crystallography, X-Ray , Humans , Immune System , Mice , Molecular Dynamics Simulation , Rats , Structure-Activity Relationship
13.
Medchemcomm ; 8(8): 1659-1667, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-29250307

ABSTRACT

A pyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidin-5-amine antagonist of the A2A adenosine receptor (AR) was functionalized as amine congeners, fluorescent conjugates and a sulfonate, and the A2AAR binding modes were predicted computationally. The optimal n-butyl spacer was incorporated into the following A2AAR-selective (Ki, nM) conjugates: BODIPY630/650 derivative 11 (MRS7396, 24.6) and AlexaFluor488 derivative 12 (MRS7416, 30.3). Flow cytometry of 12 in hA2AAR-expressing HEK-293 cells displayed saturable binding (low nonspecific) and inhibition by known A2AAR antagonists. Water-soluble sulfonate 13 was a highly potent (Ki = 6.2 nM) and selective A2AAR antagonist based on binding and functional assays. Docking and molecular dynamics simulations predicted the regions of interaction of the distal portions of these chain-extended ligands with the A2AAR. The BODIPY630/650 fluorophore of 11 was buried in a hydrophobic interhelical (TM1/TM7) region, while AlexaFluor488 of 12 associated with the hydrophilic extracellular loops. In conclusion, we have identified novel high affinity antagonist probes for A2AAR drug discovery and characterization.

14.
J Chem Inf Model ; 57(12): 3104-3123, 2017 12 26.
Article in English | MEDLINE | ID: mdl-29182323

ABSTRACT

We performed a molecular modeling analysis of 100 nucleotide-like bisphosphates and 46 non-nucleotide arylurea derivatives previously reported as P2Y1R binders using the recently solved hP2Y1R structures. We initially docked the compounds at the X-ray structures and identified the binding modes of representative compounds highlighting key patterns in the structure-activity relationship (SAR). We subsequently subjected receptor complexes with selected key agonists (2MeSADP and MRS2268) and antagonists (MRS2500 and BPTU) to membrane molecular dynamics (MD) simulations (at least 200 ns run in triplicate, simulation time 0.6-1.6 µs per ligand system) while considering alternative protonation states of nucleotides. Comparing the temporal evolution of the ligand-protein interaction patterns with available site-directed mutagenesis (SDM) data and P2Y1R apo state simulation provided further SAR insights and suggested reasonable explanations for loss/gain of binding affinity as well as the most relevant charged species for nucleotide ligands. The MD analysis also predicted local conformational changes required for the receptor inactive state to accommodate nucleotide agonists.


Subject(s)
Purinergic P2Y Receptor Agonists/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y1/metabolism , Drug Discovery , Humans , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Purinergic P2Y Receptor Agonists/chemistry , Purinergic P2Y Receptor Antagonists/chemistry , Receptors, Purinergic P2Y1/chemistry , Structure-Activity Relationship
15.
Drug Discov Today ; 22(12): 1782-1791, 2017 12.
Article in English | MEDLINE | ID: mdl-28781163

ABSTRACT

A single molecular scaffold can be adapted to interact with diverse targets, either separately or simultaneously. Nucleosides and nucleotides in which ribose is substituted with bicyclo[3.1.0]hexane are an example of a versatile drug-like scaffold for increasing selectivity at their classical targets: kinases, polymerases, adenosine and P2 receptors. Also, by applying structure-based functional group manipulations, rigidified adenosine derivatives can be repurposed to satisfy pharmacophoric requirements of various GPCRs, ion channels, enzymes and transporters, initially detected as off-target activities. Recent examples include 5HT2B serotonin receptor antagonists and novel dopamine transporter allosteric modulators. This directable target diversity establishes rigid nucleosides as privileged scaffolds.


Subject(s)
Nucleosides/pharmacology , Animals , Humans , Ligands , Molecular Conformation , Nucleosides/chemistry , Polypharmacology
16.
Molecules ; 22(3)2017 Mar 11.
Article in English | MEDLINE | ID: mdl-28287473

ABSTRACT

Adenosine is an endogenous modulator exerting its functions through the activation of four adenosine receptor (AR) subtypes, termed A1, A2A, A2B and A3, which belong to the G protein-coupled receptor (GPCR) superfamily. The human A3AR (hA3AR) subtype is implicated in several cytoprotective functions. Therefore, hA3AR modulators, and in particular agonists, are sought for their potential application as anti-inflammatory, anticancer, and cardioprotective agents. Structure-based molecular modeling techniques have been applied over the years to rationalize the structure-activity relationships (SARs) of newly emerged A3AR ligands, guide the subsequent lead optimization, and interpret site-directed mutagenesis (SDM) data from a molecular perspective. In this review, we showcase selected modeling-based and guided strategies that were applied to elucidate the binding of agonists to the A3AR and discuss the challenges associated with an accurate prediction of the receptor extracellular vestibule through homology modeling from the available X-ray templates.


Subject(s)
Adenosine A3 Receptor Agonists/chemical synthesis , Adenosine/chemical synthesis , Anti-Inflammatory Agents/chemical synthesis , Antineoplastic Agents/chemical synthesis , Cardiotonic Agents/chemical synthesis , Receptor, Adenosine A3/chemistry , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A3 Receptor Agonists/pharmacology , Adenosine A3 Receptor Antagonists/chemical synthesis , Adenosine A3 Receptor Antagonists/pharmacology , Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , Cardiotonic Agents/pharmacology , Drug Design , Humans , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Protein Binding , Protein Conformation , Receptor, Adenosine A3/genetics , Receptor, Adenosine A3/metabolism , Structural Homology, Protein , Structure-Activity Relationship
17.
Medchemcomm ; 8(10): 1897-1908, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-29423136

ABSTRACT

Both agonists and antagonists of the UDP-activated P2Y6 receptor (P2Y6R) have been proposed for therapeutic use, in conditions such as cancer, inflammation, neurodegeneration and diabetes. Uracil nucleotides containing a South-bicyclo[3.1.0]hexane ((S)-methanocarba) ring system in place of the ribose ring were synthesized and shown to be potent P2Y6R agonists in a calcium mobilization assay. The (S)-methanocarba modification was compatible with either a 5-iodo or 4-methoxyimino group on the pyrimidine, but not with a α,ß-methylene 5´-diphosphate. (S)-Methanocarba dinucleotide potency was compatible with a N4-methoxy modification on the proximal nucleoside that is assumed to bind at the P2Y6R similarly to UDP; (N)-methanocarba was preferred on the distal nucleoside moiety. This suggests that the distal dinucleotide P2Y6R binding site prefers a ribose-like group that can attain a (N) conformation, rather than (S). Dinucleotide binding was modeled by homology modeling, docking and molecular dynamics simulations, which suggested the same ribose conformational preferences found empirically.

18.
J Med Chem ; 59(24): 11006-11026, 2016 12 22.
Article in English | MEDLINE | ID: mdl-27933810

ABSTRACT

Adenosine derivatives developed to activate adenosine receptors (ARs) revealed micromolar activity at serotonin 5HT2B and 5HT2C receptors (5HTRs). We explored the structure-activity relationship at 5HT2Rs and modeled receptor interactions in order to optimize affinity and simultaneously reduce AR affinity. Depending on N6 substitution, small 5'-alkylamide modification maintained 5HT2BR affinity, which was enhanced upon ribose substitution with rigid bicyclo[3.1.0]hexane (North (N)-methanocarba), e.g., N6-dicyclopropylmethyl 4'-CH2OH derivative 14 (Ki 11 nM). 5'-Methylamide 23 was 170-fold selective as antagonist for 5HT2BR vs 5HT2CR. 5'-Methyl 25 and ethyl 26 esters potently antagonized 5HT2Rs with moderate selectivity in comparison to ARs; related 6-N,N-dimethylamino analogue 30 was 5HT2R-selective. 5' position flexibility of substitution was indicated in 5HT2BR docking. Both 5'-ester and 5'-amide derivatives displayed in vivo t1/2 of 3-4 h. Thus, we used G protein-coupled receptor modeling to repurpose nucleoside scaffolds in favor of binding at nonpurine receptors as novel 5HT2R antagonists, with potential for cardioprotection, liver protection, or central nervous system activity.


Subject(s)
Adenosine/pharmacology , Receptors, Serotonin/metabolism , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Adenosine/chemical synthesis , Adenosine/chemistry , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Serotonin 5-HT2 Receptor Antagonists/chemical synthesis , Serotonin 5-HT2 Receptor Antagonists/chemistry , Structure-Activity Relationship
19.
Mol Inform ; 35(8-9): 440-8, 2016 09.
Article in English | MEDLINE | ID: mdl-27546048

ABSTRACT

In this review, we present a survey of the recent advances carried out by our research groups in the field of ligand-GPCRs recognition process simulations recently implemented at the Molecular Modeling Section (MMS) of the University of Padova. We briefly describe a platform of tools we have tuned to aid the identification of novel GPCRs binders and the better understanding of their binding mechanisms, based on two extensively used computational techniques such as molecular docking and MD simulations. The developed methodologies encompass: (i) the selection of suitable protocols for docking studies, (ii) the exploration of the dynamical evolution of ligand-protein interaction networks, (iii) the detailed investigation of the role of water molecules upon ligand binding, and (iv) a glance at the way the ligand might go through prior reaching the binding site.


Subject(s)
Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Binding Sites/physiology , Humans , Ligands , Models, Molecular , Molecular Docking Simulation/methods , Molecular Dynamics Simulation , Protein Binding/physiology
20.
J Med Chem ; 59(14): 6860-77, 2016 07 28.
Article in English | MEDLINE | ID: mdl-27410258

ABSTRACT

Adenosine kinase (AdK) inhibitors raise endogenous adenosine levels, particularly in disease states, and have potential for treatment of seizures, neurodegeneration, and inflammation. On the basis of the South (S) ribose conformation and molecular dynamics (MD) analysis of nucleoside inhibitors bound in AdK X-ray crystallographic structures, (S)- and North (N)-methanocarba (bicyclo[3.1.0]hexane) derivatives of known inhibitors were prepared and compared as human (h) AdK inhibitors. 5'-Hydroxy (34, MRS4202 (S); 55, MRS4380 (N)) and 5'-deoxy 38a (MRS4203 (S)) analogues, containing 7- and N(6)-NH phenyl groups in 7-deazaadenine, robustly inhibited AdK activity (IC50 ∼ 100 nM), while the 5'-hydroxy derivative 30 lacking the phenyl substituents was weak. Docking in the hAdK X-ray structure and MD simulation suggested a mode of binding similar to 5'-deoxy-5-iodotubercidin and other known inhibitors. Thus, a structure-based design approach for further potency enhancement is possible. The potent AdK inhibitors in this study are ready to be further tested in animal models of epilepsy.


Subject(s)
Adenosine Kinase/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Tubercidin/pharmacology , Adenosine Kinase/metabolism , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Conformation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , Tubercidin/chemical synthesis , Tubercidin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...