Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
J Chem Inf Model ; 59(6): 2729-2740, 2019 06 24.
Article in English | MEDLINE | ID: mdl-31144815

ABSTRACT

Cyclic nucleotide phosphodiesterases (PDE's) are metalloenzymes that play a key role in regulating the levels of the ubiquitous second messengers, cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). In humans, 11 PDE protein families mediate numerous biochemical pathways throughout the body and are effective drug targets for the treatment of diseases ranging from central nervous system disorders to heart and pulmonary diseases. PDE's also share a highly conserved catalytic site (about 50%), thus making the design of selective drug candidates very challenging with classical structure-based design approaches given also the lack of publicly available co-crystal structures of pairs of PDE's with consistent biological data to be compared, as we show in our work. In this retrospective study, we apply free energy perturbation (FEP+) to predict the selectivity of inhibitors that bind two pairs of closely related PDE families: PDE9/1 and PDE5/6 where only 1 co-crystal structure per pair is publicly available. As another challenge, the p Ka of the PDE5/6 inhibitor is close to the experimental pH, making unclear the exact protonation state that should be used in the computational workflow. We demonstrate that running FEP+ on homology models constructed for these metalloenzymes accurately reproduces experimentally observed selectivity profiles also addressing the unclear protonation state to be used during computation with our recently developed p Ka-correction method. Based on these data, we conclude that FEP+ is a robust method for prediction of selectivity for this target class and may be helpful to address related lead optimization challenges in drug discovery.


Subject(s)
Drug Discovery , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Binding Sites/drug effects , Catalytic Domain/drug effects , Drug Discovery/methods , Humans , Ligands , Molecular Docking Simulation , Phosphoric Diester Hydrolases/chemistry , Thermodynamics
2.
ACS Infect Dis ; 4(5): 771-787, 2018 05 11.
Article in English | MEDLINE | ID: mdl-29465985

ABSTRACT

The success of Mycobacterium tuberculosis (Mtb) as a pathogen depends on the redundant and complex mechanisms it has evolved for resisting nitrosative and oxidative stresses inflicted by host immunity. Improving our understanding of these defense pathways can reveal vulnerable points in Mtb pathogenesis. In this study, we combined genetic, structural, computational, biochemical, and biophysical approaches to identify a novel enzyme class represented by Rv2466c. We show that Rv2466c is a mycothiol-dependent nitroreductase of Mtb and can reduce the nitro group of a novel mycobactericidal compound using mycothiol as a cofactor. In addition to its function as a nitroreductase, Rv2466c confers partial protection to menadione stress.


Subject(s)
Cysteine/metabolism , Glycopeptides/metabolism , Inositol/metabolism , Mycobacterium tuberculosis/enzymology , Nitroreductases/genetics , Nitroreductases/metabolism , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites , Cysteine/chemistry , Disease Models, Animal , Enzyme Activation , Female , Glycopeptides/chemistry , Inositol/chemistry , Mice , Models, Molecular , Mutation , Mycobacterium tuberculosis/classification , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Nitroreductases/chemistry , Oxidation-Reduction , Oxidative Stress , Phylogeny , Protein Binding , Protein Conformation , Structure-Activity Relationship , Tuberculosis/microbiology
3.
Curr Opin Pharmacol ; 30: 69-75, 2016 10.
Article in English | MEDLINE | ID: mdl-27490828

ABSTRACT

G protein-coupled receptors (GPCRs) constitute a major class of drug targets and modulating their signaling can produce a wide range of pharmacological outcomes. With the growing number of high-resolution GPCR crystal structures, we have the unprecedented opportunity to leverage structure-based drug design techniques. Here, we discuss a number of advanced molecular dynamics (MD) techniques that have been applied to GPCRs, including long time scale simulations, enhanced sampling techniques, water network analyses, and free energy approaches to determine relative binding free energies. On the basis of the many success stories, including those highlighted here, we expect that MD techniques will be increasingly applied to aid in structure-based drug design and lead optimization for GPCRs.


Subject(s)
Drug Design , Molecular Dynamics Simulation , Receptors, G-Protein-Coupled/metabolism , Humans , Models, Molecular , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/metabolism , Protein Binding , Receptors, G-Protein-Coupled/chemistry , Signal Transduction/drug effects
4.
ACS Omega ; 1(2): 293-304, 2016 Aug 31.
Article in English | MEDLINE | ID: mdl-30023478

ABSTRACT

The rapid growth of structural information for G-protein-coupled receptors (GPCRs) has led to a greater understanding of their structure, function, selectivity, and ligand binding. Although novel ligands have been identified using methods such as virtual screening, computationally driven lead optimization has been possible only in isolated cases because of challenges associated with predicting binding free energies for related compounds. Here, we provide a systematic characterization of the performance of free-energy perturbation (FEP) calculations to predict relative binding free energies of congeneric ligands binding to GPCR targets using a consistent protocol and no adjustable parameters. Using the FEP+ package, first we validated the protocol, which includes a full lipid bilayer and explicit solvent, by predicting the binding affinity for a total of 45 different ligands across four different GPCRs (adenosine A2AAR, ß1 adrenergic, CXCR4 chemokine, and δ opioid receptors). Comparison with experimental binding affinity measurements revealed a highly predictive ranking correlation (average spearman ρ = 0.55) and low root-mean-square error (0.80 kcal/mol). Next, we applied FEP+ in a prospective project, where we predicted the affinity of novel, potent adenosine A2A receptor (A2AR) antagonists. Four novel compounds were synthesized and tested in a radioligand displacement assay, yielding affinity values in the nanomolar range. The affinity of two out of the four novel ligands (plus three previously reported compounds) was correctly predicted (within 1 kcal/mol), including one compound with approximately a tenfold increase in affinity compared to the starting compound. Detailed analyses of the simulations underlying the predictions provided insights into the structural basis for the two cases where the affinity was overpredicted. Taken together, these results establish a protocol for systematically applying FEP+ to GPCRs and provide guidelines for identifying potent molecules in drug discovery lead optimization projects.

5.
Proc Natl Acad Sci U S A ; 112(15): E1898-907, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25827233

ABSTRACT

Next-generation sequencing is transforming our understanding of human genetic variation but assessing the functional impact of novel variants presents challenges. We analyzed missense variants in the integrin αIIbß3 receptor subunit genes ITGA2B and ITGB3 identified by whole-exome or -genome sequencing in the ThromboGenomics project, comprising ∼32,000 alleles from 16,108 individuals. We analyzed the results in comparison with 111 missense variants in these genes previously reported as being associated with Glanzmann thrombasthenia (GT), 20 associated with alloimmune thrombocytopenia, and 5 associated with aniso/macrothrombocytopenia. We identified 114 novel missense variants in ITGA2B (affecting ∼11% of the amino acids) and 68 novel missense variants in ITGB3 (affecting ∼9% of the amino acids). Of the variants, 96% had minor allele frequencies (MAF) < 0.1%, indicating their rarity. Based on sequence conservation, MAF, and location on a complete model of αIIbß3, we selected three novel variants that affect amino acids previously associated with GT for expression in HEK293 cells. αIIb P176H and ß3 C547G severely reduced αIIbß3 expression, whereas αIIb P943A partially reduced αIIbß3 expression and had no effect on fibrinogen binding. We used receiver operating characteristic curves of combined annotation-dependent depletion, Polyphen 2-HDIV, and sorting intolerant from tolerant to estimate the percentage of novel variants likely to be deleterious. At optimal cut-off values, which had 69-98% sensitivity in detecting GT mutations, between 27% and 71% of the novel αIIb or ß3 missense variants were predicted to be deleterious. Our data have implications for understanding the evolutionary pressure on αIIbß3 and highlight the challenges in predicting the clinical significance of novel missense variants.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Mutation, Missense , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Thrombasthenia/genetics , Alleles , Databases, Nucleic Acid , Exome/genetics , Fibrinogen/chemistry , Fibrinogen/metabolism , Gene Frequency , HEK293 Cells , Humans , Immunoblotting , Models, Molecular , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Thrombasthenia/metabolism
6.
Arterioscler Thromb Vasc Biol ; 34(10): 2321-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25147334

ABSTRACT

OBJECTIVE: Treatment of myocardial infarction within the first 1 to 2 hours with a thrombolytic agent, percutaneous coronary intervention, or an αIIbß3 antagonist decreases mortality and the later development of heart failure. We previously reported on a novel small molecule αIIbß3 antagonist, RUC-2, that has a unique mechanism of action. We have now developed a more potent and more soluble congener of RUC-2, RUC-4, designed to be easily administered intramuscularly by autoinjector to facilitate its use in the prehospital setting. Here, we report the properties of RUC-4 and the antiplatelet and antithrombotic effects of RUC-2 and RUC-4 in animal models. APPROACH AND RESULTS: RUC-4 was ≈ 20% more potent than RUC-2 in inhibiting human ADP-induced platelet aggregation and much more soluble in aqueous solutions (60-80 mg/mL). It shared RUC-2's specificity for αIIbß3 versus αVß3, did not prime the receptor to bind fibrinogen, or induce changes in ß3 identified by a conformation-specific monoclonal antibody. Both RUC-2 and RUC-4 prevented FeCl3-induced thrombotic occlusion of the carotid artery in mice and decreased microvascular thrombi in response to laser injury produced by human platelets infused into transgenic mice containing a mutated von Willebrand factor that reacts with human but not mouse platelets. Intramuscular injection of RUC-4 in nonhuman primates at 1.9 and 3.85 mg/kg led to complete inhibition of platelet aggregation within 15 minutes, with dose-dependent return of platelet aggregation after 4.5 to 24 hours. CONCLUSIONS: RUC-4 has favorable biochemical, pharmacokinetic, pharmacodynamic, antithrombotic, and solubility properties as a prehospital therapy of myocardial infarction, but the possibility of increased bleeding with therapeutic doses remains to be evaluated.


Subject(s)
Blood Platelets/drug effects , Carotid Stenosis/prevention & control , Emergency Medical Services , Fibrinolytic Agents/pharmacology , Myocardial Infarction/drug therapy , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Pyrimidinones/pharmacology , Thiadiazoles/pharmacology , Thrombosis/prevention & control , Animals , Binding Sites , Blood Platelets/metabolism , Carotid Stenosis/blood , Carotid Stenosis/chemically induced , Chlorides , Disease Models, Animal , Ferric Compounds , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/metabolism , Fibrinolytic Agents/pharmacokinetics , Humans , Macaca fascicularis , Male , Mice , Mice, Transgenic , Molecular Dynamics Simulation , Myocardial Infarction/blood , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/metabolism , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Binding , Protein Conformation , Pyrimidinones/chemistry , Pyrimidinones/metabolism , Pyrimidinones/pharmacokinetics , Solubility , Thiadiazoles/chemistry , Thiadiazoles/metabolism , Thiadiazoles/pharmacokinetics , Thrombosis/blood , Thrombosis/chemically induced , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
7.
Proteins ; 82(12): 3231-3240, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24677266

ABSTRACT

Platelet aggregation is the consequence of the binding of extracellular bivalent ligands such as fibrinogen and von Willebrand factor to the high affinity, active state of integrin αIIbß3. This state is achieved through a so-called "inside-out" mechanism characterized by the membrane-assisted formation of a complex between the F2 and F3 subdomains of intracellular protein talin and the integrin ß3 tail. Here, we present the results of multi-microsecond, all-atom molecular dynamics simulations carried on the complete transmembrane (TM) and C-terminal (CT) domains of αIIbß3 integrin in an explicit lipid-water environment, and in the presence or absence of the talin-1 F2 and F3 subdomains. These large-scale simulations provide unprecedented molecular-level insights into the talin-driven inside-out activation of αIIbß3 integrin. Specifically, they suggest a preferred conformation of the complete αIIbß3 TM/CT domains in a lipid-water environment, and testable hypotheses of key intermolecular interactions between αIIbß3 integrin and the F2/F3 domains of talin-1. Notably, not only do these simulations give support to a stable left-handed reverse turn conformation of the αIIb juxtamembrane motif rather than a helical turn, but they raise the question as to whether TM helix separation is required for talin-driven integrin activation.


Subject(s)
Cell Membrane/metabolism , Models, Biological , Platelet Activation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Talin/metabolism , Cell Membrane/chemistry , Databases, Protein , Humans , Kinetics , Molecular Dynamics Simulation , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Protein Conformation , Protein Interaction Domains and Motifs , Protein Refolding , Protein Stability , Protein Transport , Signal Transduction , Surface Properties , Talin/chemistry
8.
Bioorg Med Chem Lett ; 24(4): 1148-53, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24461295

ABSTRACT

A collection of αIIbß3 integrin receptor antagonists possessing a unique MIDAS metal ion displacement mechanism of action is presented. Insight into these agents' structure-activity relationships, binding modality, and pharmacokinetic and pharmacodynamic profiles highlight the potential of these small molecule ion displacement ligands as attractive candidates for clinical development.


Subject(s)
Blood Proteins/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Blood Proteins/chemical synthesis , Blood Proteins/chemistry , Dose-Response Relationship, Drug , Humans , Ions/chemistry , Ligands , Models, Molecular , Molecular Conformation , Platelet Aggregation/drug effects , Structure-Activity Relationship
9.
Biopolymers ; 101(1): 21-7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23564013

ABSTRACT

Delta-opioid (DOP) receptors are members of the G protein-coupled receptor (GPCR) sub-family of opioid receptors, and are evolutionarily related, with homology exceeding 70%, to cognate mu-opioid (MOP), kappa-opioid (KOP), and nociceptin opioid (NOP) receptors. DOP receptors are considered attractive drug targets for pain management because agonists at these receptors are reported to exhibit strong antinociceptive activity with relatively few side effects. Among the most potent analgesics targeting the DOP receptor are the linear and cyclic enkephalin analogs known as DADLE (Tyr-D-Ala-Gly-Phe-D-Leu) and DPDPE (Tyr-D-Pen-Gly-Phe-D-Pen), respectively. Several computational and experimental studies have been carried out over the years to characterize the conformational profile of these penta-peptides with the ultimate goal of designing potent peptidomimetic agonists for the DOP receptor. The computational studies published to date, however, have investigated only a limited range of timescales and used over-simplified representations of the solvent environment. We provide here a thorough exploration of the conformational space of DADLE and DPDPE in an explicit solvent, using microsecond-scale molecular dynamics and bias-exchange metadynamics simulations. Free-energy profiles derived from these simulations point to a small number of DADLE and DPDPE conformational minima in solution, which are separated by relatively small energy barriers. Candidate bioactive forms of these peptides are selected from identified common spatial arrangements of key pharmacophoric points within all sampled conformations.


Subject(s)
Enkephalin, D-Penicillamine (2,5)- , Receptors, Opioid, delta , Enkephalins , Receptors, Opioid , Receptors, Opioid, mu , Rubiaceae
10.
PLoS One ; 8(12): e81609, 2013.
Article in English | MEDLINE | ID: mdl-24349096

ABSTRACT

Structural and functional analyses of integrin αIIbß3 has implicated swing-out motion of the ß3 hybrid domain in αIIbß3 activation and ligand binding. Using data from targeted molecular dynamics (TMD) simulations, we engineered two disulfide-bonded mutant receptors designed to limit swing-out (XS-O). XS-O mutants cannot bind the high Mr ligand fibrinogen in the presence of an activating mAb or after introducing mutations into the αIIb subunit designed to simulate inside-out signaling. They also have reduced capacity to be "primed" to bind fibrinogen by pretreatment with eptifibatide. They can, however, bind the small RGD venom protein kistrin. Despite their inability to bind soluble fibrinogen, the XS-O mutants can support adhesion to immobilized fibrinogen, although such adhesion does not initiate outside-in signaling leading to normal cytoskeletal reorganization. Collectively, our data further define the biologic role of ß3 hybrid domain swing-out in both soluble and immobilized high Mr ligand binding, as well as in priming and outside-in signaling. We also infer that swing-out is likely to be a downstream effect of receptor extension.


Subject(s)
Cytoskeleton/metabolism , Fibrinogen/chemistry , Immobilized Proteins/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Binding Sites , Blood Platelets/cytology , Blood Platelets/metabolism , Cytoskeleton/ultrastructure , Eptifibatide , Fibrinogen/metabolism , Gene Expression , HEK293 Cells , Humans , Immobilized Proteins/metabolism , Ligands , Molecular Dynamics Simulation , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptides/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Snakes
11.
Proc Natl Acad Sci U S A ; 110(29): 12072-7, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23818586

ABSTRACT

G protein-coupled receptors play a pivotal role in many physiological signaling pathways. Mounting evidence suggests that G protein-coupled receptors, including opioid receptors, form dimers, and dimerization is necessary for receptor maturation, signaling, and trafficking. However, the physiological role of dimerization in vivo has not been well-explored because of the lack of tools to study these dimers in endogenous systems. To address this problem, we previously generated antibodies to µ-δ opioid receptor (µOR-δOR) dimers and used them to study the pharmacology and signaling by this heteromer. We also showed that the heteromer exhibits restricted distribution in the brain and that its abundance is increased in response to chronic morphine administration. Thus, the µOR-δOR heteromer represents a potentially unique target for the development of therapeutics to treat pain. Here, we report the identification of compounds targeting µOR-δOR heteromers through high-throughput screening of a small-molecule library. These compounds exhibit activity in µOR-δOR cells but not µOR or δOR cells alone. Among them, CYM51010 was found to be a µOR-δOR-biased ligand, because its activity is blocked by the µOR-δOR heteromer antibody. Notably, systemic administration of CYM51010 induced antinociceptive activity similar to morphine, and chronic administration of CYM51010 resulted in lesser antinociceptive tolerance compared with morphine. Taken together, these results suggest that CYM51010, a µOR-δOR-biased ligand, could serve as a scaffold for the development of a unique type (heteromer-biased) of drug that is more potent and without the severe side effects associated with conventional clinical opioids.


Subject(s)
Analgesics/pharmacology , Brain/metabolism , Piperidines/pharmacology , Receptors, Opioid, delta/agonists , Receptors, Opioid, mu/agonists , Analgesics/metabolism , Analysis of Variance , Animals , Antibodies, Monoclonal/metabolism , Cell Line , Dimerization , Drug Tolerance/physiology , High-Throughput Screening Assays , Male , Mice , Mice, Inbred C57BL , Piperidines/metabolism , Radioligand Assay , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Small Molecule Libraries
13.
J Chem Inf Model ; 53(3): 521-6, 2013 Mar 25.
Article in English | MEDLINE | ID: mdl-23461591

ABSTRACT

Kappa-opioid (KOP) receptor agonists exhibit analgesic effects without activating reward pathways. In the search for nonaddictive opioid therapeutics and novel chemical tools to study physiological functions regulated by the KOP receptor, we screened in silico its recently released inactive crystal structure. A selective novel KOP receptor agonist emerged as a notable result and is proposed as a new chemotype for the study of the KOP receptor in the etiology of drug addiction, depression, and/or pain.


Subject(s)
Drug Discovery/methods , Receptors, Opioid, kappa/agonists , Arrestin/metabolism , Crystallography, X-Ray , Cyclic AMP/metabolism , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Databases, Factual , GTP-Binding Proteins/metabolism , Genetic Vectors , HEK293 Cells , High-Throughput Screening Assays , Humans , Models, Molecular , Molecular Conformation , Receptors, Opioid, kappa/genetics , Stereoisomerism , Structure-Activity Relationship , Transfection , User-Computer Interface
14.
J Comput Aided Mol Des ; 26(9): 1005-15, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22893377

ABSTRACT

Integrin αIIbß3 has emerged as an important therapeutic target for thrombotic vascular diseases owing to its pivotal role in mediating platelet aggregation through interaction with adhesive ligands. In the search for effective anti-thrombotic agents that can be administered orally without inducing the high-affinity ligand binding state, we recently discovered via high-throughput screening of 33,264 compounds a novel, αIIbß3-selective inhibitor (RUC-1) of adenosine-5'-diphosphate (ADP) -induced platelet aggregation that exhibits a different chemical scaffold and mode of binding with respect to classical Arg-Gly-Asp (RGD)-mimicking αIIbß3 antagonists. Most importantly, RUC-1 and its higher-affinity derivative, RUC-2, do not induce major conformational changes in the protein ß3 subunit or prime the receptor to bind ligand. To identify additional αIIbß3-selective chemotypes that inhibit platelet aggregation through similar mechanisms, we screened in silico over 2.5 million commercially available, 'lead-like' small molecules based on complementarity to the predicted binding mode of RUC-2 into the RUC-1-αIIbß3 crystal structure. This first reported structure-based virtual screening application to the αIIbß3 integrin led to the identification of 2 αIIbß3-selective antagonists out of 4 tested, which compares favorably with the 0.003 % "hit rate" of our previous high-throughput chemical screening study. The newly identified compounds, like RUC-1 and RUC-2, showed specificity for αIIbß3 compared to αVß3 and did not prime the receptor to bind ligand. They thus may hold promise as αIIbß3 antagonist therapeutic scaffolds.


Subject(s)
Blood Platelets/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Small Molecule Libraries , Crystallization , Humans , Ligands , Models, Molecular , Molecular Docking Simulation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Binding
15.
Sci Transl Med ; 4(125): 125ra32, 2012 Mar 14.
Article in English | MEDLINE | ID: mdl-22422993

ABSTRACT

An integrin found on platelets, α(IIb)ß(3) mediates platelet aggregation, and α(IIb)ß(3) antagonists are effective antithrombotic agents in the clinic. Ligands bind to integrins in part by coordinating a magnesium ion (Mg(2+)) located in the ß subunit metal ion-dependent adhesion site (MIDAS). Drugs patterned on the integrin ligand sequence Arg-Gly-Asp have a basic moiety that binds the α(IIb) subunit and a carboxyl group that coordinates the MIDAS Mg(2+) in the ß(3) subunits. They induce conformational changes in the ß(3) subunit that may have negative consequences such as exposing previously hidden epitopes and inducing the active conformation of the receptor. We recently reported an inhibitor of α(IIb)ß(3) (RUC-1) that binds exclusively to the α(IIb) subunit; here, we report the structure-based design and synthesis of RUC-2, a RUC-1 derivative with a ~100-fold higher affinity. RUC-2 does not induce major conformational changes in ß(3) as judged by monoclonal antibody binding, light scattering, gel chromatography, electron microscopy, and a receptor priming assay. X-ray crystallography of the RUC-2-α(IIb)ß(3) headpiece complex in 1 mM calcium ion (Ca(2+))/5 mM Mg(2+) at 2.6 Å revealed that RUC-2 binds to α(IIb) the way RUC-1 does, but in addition, it binds to the ß(3) MIDAS residue glutamic acid 220, thus displacing Mg(2+) from the MIDAS. When the Mg(2+) concentration was increased to 20 mM, however, Mg(2+) was identified in the MIDAS and RUC-2 was absent. RUC-2's ability to inhibit ligand binding and platelet aggregation was diminished by increasing the Mg(2+) concentration. Thus, RUC-2 inhibits ligand binding by a mechanism different from that of all other α(IIb)ß(3) antagonists and may offer advantages as a therapeutic agent.


Subject(s)
Magnesium/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Animals , Binding Sites , Blood Platelets/drug effects , Blood Platelets/metabolism , Cell Adhesion/physiology , Collagen/metabolism , Crystallography, X-Ray , Fibrinogen/metabolism , Humans , Mice , Microscopy, Electron , Oligopeptides , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/metabolism , Platelet Aggregation Inhibitors/pharmacology , Protein Binding/drug effects , Protein Binding/physiology , Rats , Vitronectin/metabolism
16.
Org Biomol Chem ; 10(8): 1543-52, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22222915

ABSTRACT

Mitomycin C (MMC) is a potent antitumour agent that forms a covalent bond with the 2-amino group of selected guanines in the minor groove of double-stranded DNA following intracellular reduction of its quinone ring and opening of its aziridine moiety. At some 5'-CG-3' (CpG) steps the resulting monofunctional adduct can evolve towards a more deleterious bifunctional lesion, which is known as an interstrand crosslink (ICL). MMC reactivity is enhanced when the cytosine bases are methylated (5 MC) and decreased when they are replaced with 5-F-cytosine (5FC) whereas the stereochemical preference of alkylation changes upon decarbamoylation. We have studied three duplex oligonucleotides of general formula d(CGATAAXGCTAACG) in which X stands for C, 5MC or 5FC. Using a combination of molecular dynamics simulations in aqueous solution, quantum mechanics and continuum electrostatics, we have been able to (i) obtain a large series of snapshots that facilitate an understanding in atomic detail of the distinct stereochemistry of monoadduct and ICL formation by MMC and its decarbamoylated analogue, (ii) provide an explanation for the altered reactivity of MMC towards DNA molecules containing 5MC or 5FC, and (iii) show the distinct accommodation in the DNA minor groove of the different covalent modifications, particularly the most cytotoxic C1α and C1ß ICLs.


Subject(s)
CpG Islands , Cytosine/chemistry , DNA/chemistry , Mitomycin/chemistry , Models, Molecular , Stereoisomerism
17.
PLoS Comput Biol ; 7(10): e1002193, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22022248

ABSTRACT

Extensive experimental information supports the formation of ligand-specific conformations of G protein-coupled receptors (GPCRs) as a possible molecular basis for their functional selectivity for signaling pathways. Taking advantage of the recently published inactive and active crystal structures of GPCRs, we have implemented an all-atom computational strategy that combines different adaptive biasing techniques to identify ligand-specific conformations along pre-determined activation pathways. Using the prototypic GPCR ß2-adrenergic receptor as a suitable test case for validation, we show that ligands with different efficacies (either inverse agonists, neutral antagonists, or agonists) modulate the free-energy landscape of the receptor by shifting the conformational equilibrium towards active or inactive conformations depending on their elicited physiological response. Notably, we provide for the first time a quantitative description of the thermodynamics of the receptor in an explicit atomistic environment, which accounts for the receptor basal activity and the stabilization of different active-like states by differently potent agonists. Structural inspection of these metastable states reveals unique conformations of the receptor that may have been difficult to retrieve experimentally.


Subject(s)
Models, Theoretical , Receptors, G-Protein-Coupled/metabolism , Ligands , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Thermodynamics
18.
Res Dev Disabil ; 32(1): 217-21, 2011.
Article in English | MEDLINE | ID: mdl-20956067

ABSTRACT

The study of heart rate variability is an important tool for a noninvasive evaluation of the neurocardiac integrity. The present study aims to evaluate the autonomic heart rate modulation in supine and standing positions in 12 children diagnosed with cerebral palsy and 16 children with typical motor development (control group), as well as to relate the level of motor impairment in children with cerebral palsy, as classified by to the Gross Motor Function Classification System, to the heart rate variability indices. The heart rate variability was analyzed by linear model in the frequency domain, at low and high frequency bands in normalized units and low and high frequency ratio. The results indicate that children with cerebral palsy present lower heart rate variability indices, indicating sympathovagal imbalance. The decrease of heart rate variability in children with cerebral palsy is related to the motor impairment level.


Subject(s)
Autonomic Nervous System Diseases/physiopathology , Cerebral Palsy/physiopathology , Heart Rate/physiology , Motor Skills Disorders/physiopathology , Child , Cross-Sectional Studies , Female , Hemiplegia/physiopathology , Humans , Linear Models , Male , Quadriplegia/physiopathology , Supine Position , Vagus Nerve/physiopathology
19.
Org Biomol Chem ; 9(3): 892-901, 2011 Feb 07.
Article in English | MEDLINE | ID: mdl-21127790

ABSTRACT

Based on the presumed binding mode of an earlier identified inhibitor, we herein report new 3'-modified nucleosides as potent and selective inhibitors of mitochondrial thymidine kinase (TK2). A series of thirteen 3'-amino-, 3'-guanidino- and 3'-tetrazole-containing nucleosides were synthesized and evaluated for their TK2 inhibitory activity. Within the tetrazole series, compounds with nanomolar inhibitory activity were identified. A homology model of TK2 allowed to elucidate the observed activities. Introduction of a 2-bromovinyl group on C-5 of the pyrimidine base of the most promising 3'-derivative further improved the inhibitory activity, and caused a significant increase in the selectivity for TK2 versus TK1. Interestingly, for the current series of analogues, a strong correlation was observed between TK2 and Drosophila melanogaster dNK inhibition, further substantiating the phylogenetic relationship between these two nucleoside kinases.


Subject(s)
Amines/chemistry , Azoles/chemistry , Thymidine Kinase/antagonists & inhibitors , Thymidine/chemical synthesis , Animals , Drosophila melanogaster/drug effects , Drosophila melanogaster/enzymology , Humans , Models, Molecular , Molecular Structure , Thymidine/pharmacology , Thymidine Kinase/chemistry
20.
Blood ; 116(23): 5050-9, 2010 Dec 02.
Article in English | MEDLINE | ID: mdl-20679525

ABSTRACT

The platelet integrin α(IIb)ß(3) is essential for hemostasis and thrombosis through its binding of adhesive plasma proteins. We have determined crystal structures of the α(IIb)ß(3) headpiece in the absence of ligand and after soaking in RUC-1, a novel small molecule antagonist. In the absence of ligand, the α(IIb)ß(3) headpiece is in a closed conformation, distinct from the open conformation visualized in presence of Arg-Gly-Asp (RGD) antagonists. In contrast to RGD antagonists, RUC-1 binds only to the α(IIb) subunit. Molecular dynamics revealed nearly identical binding. Two species-specific residues, α(IIb) Y190 and α(IIb) D232, in the RUC-1 binding site were confirmed as important by mutagenesis. In sharp contrast to RGD-based antagonists, RUC-1 did not induce α(IIb)ß(3) to adopt an open conformation, as determined by gel filtration and dynamic light scattering. These studies provide insights into the factors that regulate integrin headpiece opening, and demonstrate the molecular basis for a novel mechanism of integrin antagonism.


Subject(s)
Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Animals , Binding Sites , Fibrinogen/metabolism , Humans , Mutagenesis, Site-Directed , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Binding , Protein Conformation , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL
...