Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Science ; 364(6446): 1184-1188, 2019 06 21.
Article in English | MEDLINE | ID: mdl-31221859

ABSTRACT

Cystic fibrosis is a fatal disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR). Two main categories of drugs are being developed: correctors that improve folding of CFTR and potentiators that recover the function of CFTR. Here, we report two cryo-electron microscopy structures of human CFTR in complex with potentiators: one with the U.S. Food and Drug Administration (FDA)-approved drug ivacaftor at 3.3-angstrom resolution and the other with an investigational drug, GLPG1837, at 3.2-angstrom resolution. These two drugs, although chemically dissimilar, bind to the same site within the transmembrane region. Mutagenesis suggests that in both cases, hydrogen bonds provided by the protein are important for drug recognition. The molecular details of how ivacaftor and GLPG1837 interact with CFTR may facilitate structure-based optimization of therapeutic compounds.


Subject(s)
Aminophenols/chemistry , Chloride Channel Agonists/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Drugs, Investigational/chemistry , Pyrans/chemistry , Pyrazoles/chemistry , Quinolones/chemistry , Aminophenols/pharmacology , Binding Sites , Chloride Channel Agonists/pharmacology , Chloride Channel Agonists/therapeutic use , Cryoelectron Microscopy , Cystic Fibrosis/drug therapy , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Drugs, Investigational/pharmacology , Drugs, Investigational/therapeutic use , HEK293 Cells , Humans , Hydrogen Bonding , Mutagenesis , Protein Domains , Protein Folding/drug effects , Pyrans/pharmacology , Pyrans/therapeutic use , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Quinolones/pharmacology
2.
Nature ; 566(7743): 224-229, 2019 02.
Article in English | MEDLINE | ID: mdl-30728502

ABSTRACT

Despite intense interest in expanding chemical space, libraries containing hundreds-of-millions to billions of diverse molecules have remained inaccessible. Here we investigate structure-based docking of 170 million make-on-demand compounds from 130 well-characterized reactions. The resulting library is diverse, representing over 10.7 million scaffolds that are otherwise unavailable. For each compound in the library, docking against AmpC ß-lactamase (AmpC) and the D4 dopamine receptor were simulated. From the top-ranking molecules, 44 and 549 compounds were synthesized and tested for interactions with AmpC and the D4 dopamine receptor, respectively. We found a phenolate inhibitor of AmpC, which revealed a group of inhibitors without known precedent. This molecule was optimized to 77 nM, which places it among the most potent non-covalent AmpC inhibitors known. Crystal structures of this and other AmpC inhibitors confirmed the docking predictions. Against the D4 dopamine receptor, hit rates fell almost monotonically with docking score, and a hit-rate versus score curve predicted that the library contained 453,000 ligands for the D4 dopamine receptor. Of 81 new chemotypes discovered, 30 showed submicromolar activity, including a 180-pM subtype-selective agonist of the D4 dopamine receptor.


Subject(s)
Dopamine Agonists/chemistry , Dopamine Agonists/isolation & purification , Molecular Docking Simulation/methods , Small Molecule Libraries/chemistry , Small Molecule Libraries/isolation & purification , beta-Lactamase Inhibitors/chemistry , beta-Lactamase Inhibitors/isolation & purification , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Crystallography, X-Ray , Humans , Ligands , Machine Learning , Observation , Receptors, Dopamine D4/agonists , Receptors, Dopamine D4/chemistry , Receptors, Dopamine D4/metabolism , beta-Lactamases/chemistry
3.
Front Mol Biosci ; 5: 84, 2018.
Article in English | MEDLINE | ID: mdl-30255025

ABSTRACT

[This corrects the article DOI: 10.3389/fmolb.2018.00009.].

4.
ACS Chem Biol ; 13(9): 2522-2533, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30095890

ABSTRACT

Unrelated ligands, often found in drug discovery campaigns, can bind to the same receptor, even with the same protein residues. To investigate how this might occur, and whether it might be typically possible to find unrelated ligands for the same drug target, we sought examples of topologically unrelated ligands that bound to the same protein in the same site. Seventy-six pairs of ligands, each bound to the same protein (152 complexes total), were considered, classified into three groups. In the first (31 pairs of complexes), unrelated ligands interacted largely with the same pocket residues through different functional groups. In the second group (39 pairs), the unrelated ligand in each pair engaged different residues, though still within the same pocket. The smallest group (6 pairs) contained ligands with different scaffolds but with shared functional groups interacting with the same residues. We found that there are multiple chemically unrelated but physically similar functional groups that can complement any given local protein pocket; when these functional group substitutions are combined within a single molecule, they lead to topologically unrelated ligands that can each well-complement a site. It may be that many active and orthosteric sites can recognize topologically unrelated ligands.


Subject(s)
Drug Discovery/methods , Proteins/metabolism , Binding Sites , Databases, Protein , Humans , Ligands , Molecular Docking Simulation , Protein Binding , Proteins/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
5.
Biochim Biophys Acta Gen Subj ; 1862(10): 2162-2173, 2018 10.
Article in English | MEDLINE | ID: mdl-30009876

ABSTRACT

BACKGROUND: In humans, bitterness perception is mediated by ~25 bitter taste receptors present in the oral cavity. Among these receptors three, TAS2R10, TAS2R14 and TAS2R46, exhibit extraordinary wide agonist profiles and hence contribute disproportionally high to the perception of bitterness. Perhaps the most broadly tuned receptor is the TAS2R14, which may represent, because of its prominent expression in extraoral tissues, a receptor of particular importance for the physiological actions of bitter compounds beyond taste. METHODS: To investigate how the architecture and composition of the TAS2R14 binding pocket enables specific interactions with a complex array of chemically diverse bitter agonists, we carried out homology modeling and ligand docking experiments, subjected the receptor to point-mutagenesis of binding site residues and performed functional calcium mobilization assays. RESULTS: In total, 40 point-mutated receptor constructs were generated to investigate the contribution of 19 positions presumably located in the receptor's binding pocket to activation by 7 different TAS2R14 agonists. All investigated positions exhibited moderate to pronounced agonist selectivity. CONCLUSIONS: Since numerous modifications of the TAS2R14 binding pocket resulted in improved responses to individual agonists, we conclude that this bitter taste receptor might represent a suitable template for the engineering of the agonist profile of a chemoreceptive receptor. GENERAL SIGNIFICANCE: The detailed structure-function analysis of the highly promiscuous and widely expressed TAS2R14 suggests that this receptor must be considered as potentially frequent target for known and novel drugs including undesired off-effects.


Subject(s)
Aristolochic Acids/metabolism , Monoterpenes/metabolism , Picrotoxin/analogs & derivatives , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Taste/physiology , Amino Acid Sequence , Aristolochic Acids/chemistry , Bicyclic Monoterpenes , Binding Sites , Humans , Ligands , Models, Molecular , Molecular Docking Simulation , Monoterpenes/chemistry , Mutagenesis, Site-Directed , Mutation , Picrotoxin/chemistry , Picrotoxin/metabolism , Protein Binding , Protein Conformation , Protein Engineering , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Sesterterpenes
6.
Front Mol Biosci ; 5: 9, 2018.
Article in English | MEDLINE | ID: mdl-29552563

ABSTRACT

The 25 human bitter taste receptors (hT2Rs) recognize thousands of structurally and chemically diverse bitter substances. The binding modes of human bitter taste receptors hT2R10 and hT2R46, which are responsible for strychnine recognition, were previously established using site-directed mutagenesis, functional assays, and molecular modeling. Here we construct a phylogenetic tree and reconstruct ancestral sequences of the T2R10 and T2R46 clades. We next analyze the binding sites in view of experimental data to predict their ability to recognize strychnine. This analysis suggests that the common ancestor of hT2R10 and hT2R46 is unlikely to bind strychnine in the same mode as either of its two descendants. Estimation of relative divergence times shows that hT2R10 evolved earlier than hT2R46. Strychnine recognition was likely acquired first by the earliest common ancestor of the T2R10 clade before the separation of primates from other mammals, and was highly conserved within the clade. It was probably independently acquired by the common ancestor of T2R43-47 before the homo-ape speciation, lost in most T2Rs within this clade, but enhanced in the hT2R46 after humans diverged from the rest of primates. Our findings suggest hypothetical strychnine T2R receptors in several species, and serve as an experimental guide for further study. Improved understanding of how bitter taste receptors acquire the ability to be activated by particular ligands is valuable for the development of sensors for bitterness and for potential toxicity.

7.
Nature ; 555(7695): 269-273, 2018 03 08.
Article in English | MEDLINE | ID: mdl-29466326

ABSTRACT

Dopamine is a neurotransmitter that has been implicated in processes as diverse as reward, addiction, control of coordinated movement, metabolism and hormonal secretion. Correspondingly, dysregulation of the dopaminergic system has been implicated in diseases such as schizophrenia, Parkinson's disease, depression, attention deficit hyperactivity disorder, and nausea and vomiting. The actions of dopamine are mediated by a family of five G-protein-coupled receptors. The D2 dopamine receptor (DRD2) is the primary target for both typical and atypical antipsychotic drugs, and for drugs used to treat Parkinson's disease. Unfortunately, many drugs that target DRD2 cause serious and potentially life-threatening side effects due to promiscuous activities against related receptors. Accordingly, a molecular understanding of the structure and function of DRD2 could provide a template for the design of safer and more effective medications. Here we report the crystal structure of DRD2 in complex with the widely prescribed atypical antipsychotic drug risperidone. The DRD2-risperidone structure reveals an unexpected mode of antipsychotic drug binding to dopamine receptors, and highlights structural determinants that are essential for the actions of risperidone and related drugs at DRD2.


Subject(s)
Antipsychotic Agents/chemistry , Antipsychotic Agents/metabolism , Receptors, Dopamine D2/chemistry , Receptors, Dopamine D2/metabolism , Risperidone/chemistry , Risperidone/metabolism , Binding Sites , Crystallography, X-Ray , Drug Design , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Ligands , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation , Receptors, Dopamine D2/genetics , Receptors, Dopamine D3/chemistry , Receptors, Dopamine D3/metabolism , Receptors, Dopamine D4/chemistry , Receptors, Dopamine D4/metabolism
8.
Science ; 358(6361): 381-386, 2017 10 20.
Article in English | MEDLINE | ID: mdl-29051383

ABSTRACT

Dopamine receptors are implicated in the pathogenesis and treatment of nearly every neuropsychiatric disorder. Although thousands of drugs interact with these receptors, our molecular understanding of dopaminergic drug selectivity and design remains clouded. To illuminate dopamine receptor structure, function, and ligand recognition, we determined crystal structures of the D4 dopamine receptor in its inactive state bound to the antipsychotic drug nemonapride, with resolutions up to 1.95 angstroms. These structures suggest a mechanism for the control of constitutive signaling, and their unusually high resolution enabled a structure-based campaign for new agonists of the D4 dopamine receptor. The ability to efficiently exploit structure for specific probe discovery-rapidly moving from elucidating receptor structure to discovering previously unrecognized, selective agonists-testifies to the power of structure-based approaches.


Subject(s)
Dopamine Agonists/chemistry , Receptors, Dopamine D4/chemistry , Allosteric Site , Antipsychotic Agents/chemistry , Benzamides/chemistry , Dopamine Agonists/isolation & purification , Humans , Protein Conformation , Receptors, Dopamine D4/ultrastructure , Structure-Activity Relationship
9.
Cell ; 168(3): 377-389.e12, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28129538

ABSTRACT

The prototypical hallucinogen LSD acts via serotonin receptors, and here we describe the crystal structure of LSD in complex with the human serotonin receptor 5-HT2B. The complex reveals conformational rearrangements to accommodate LSD, providing a structural explanation for the conformational selectivity of LSD's key diethylamide moiety. LSD dissociates exceptionally slow from both 5-HT2BR and 5-HT2AR-a major target for its psychoactivity. Molecular dynamics (MD) simulations suggest that LSD's slow binding kinetics may be due to a "lid" formed by extracellular loop 2 (EL2) at the entrance to the binding pocket. A mutation predicted to increase the mobility of this lid greatly accelerates LSD's binding kinetics and selectively dampens LSD-mediated ß-arrestin2 recruitment. This study thus reveals an unexpected binding mode of LSD; illuminates key features of its kinetics, stereochemistry, and signaling; and provides a molecular explanation for LSD's actions at human serotonin receptors. PAPERCLIP.


Subject(s)
Lysergic Acid Diethylamide/chemistry , Receptor, Serotonin, 5-HT2B/chemistry , Arrestin/chemistry , Crystallography, X-Ray , Humans , Kinetics , Models, Chemical , Molecular Dynamics Simulation
10.
Nature ; 537(7619): 185-190, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27533032

ABSTRACT

Morphine is an alkaloid from the opium poppy used to treat pain. The potentially lethal side effects of morphine and related opioids-which include fatal respiratory depression-are thought to be mediated by µ-opioid-receptor (µOR) signalling through the ß-arrestin pathway or by actions at other receptors. Conversely, G-protein µOR signalling is thought to confer analgesia. Here we computationally dock over 3 million molecules against the µOR structure and identify new scaffolds unrelated to known opioids. Structure-based optimization yields PZM21-a potent Gi activator with exceptional selectivity for µOR and minimal ß-arrestin-2 recruitment. Unlike morphine, PZM21 is more efficacious for the affective component of analgesia versus the reflexive component and is devoid of both respiratory depression and morphine-like reinforcing activity in mice at equi-analgesic doses. PZM21 thus serves as both a probe to disentangle µOR signalling and a therapeutic lead that is devoid of many of the side effects of current opioids.


Subject(s)
Analgesics, Opioid/adverse effects , Analgesics, Opioid/chemistry , Drug Discovery , Receptors, Opioid, mu/agonists , Thiophenes/chemistry , Thiophenes/pharmacology , Urea/analogs & derivatives , Analgesia/methods , Analgesics, Opioid/pharmacology , Animals , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Docking Simulation , Pain/drug therapy , Receptors, Opioid, mu/deficiency , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Spiro Compounds/pharmacology , Structure-Activity Relationship , Thiophenes/adverse effects , Urea/adverse effects , Urea/chemistry , Urea/pharmacology
11.
Methods Cell Biol ; 132: 401-27, 2016.
Article in English | MEDLINE | ID: mdl-26928553

ABSTRACT

G protein-coupled receptors (GPCRs) are seven transmembrane (TM) proteins that play a key role in human physiology. The GPCR superfamily comprises about 800 members, classified into several classes, with rhodopsin-like Class A being the largest and most studied thus far. A huge component of the human repertoire consists of the chemosensory GPCRs, including ∼400 odorant receptors, 25 bitter taste receptors (TAS2Rs), which are thought to guard the organism from consuming poisons, and sweet and umami TAS1R heteromers, which indicate the nutritive value of food. The location of the binding site of TAS2Rs is similar to that of Class A GPCRs. However, most of the known bitter ligands are agonists, with only a few antagonists documented thus far. The agonist-to-antagonist ratios of Class A GPCRs vary, but in general are much lower than for TAS2Rs. For a set of well-studied GPCRs, a gradual change in agonists-to-antagonists ratios is observed when comparing low (10 µM)- and high (10 nM)-affinity ligand sets from ChEMBL and the DrugBank set of drugs. This shift reflects pharmaceutical bias toward the therapeutically desirable pharmacology for each of these GPCRs, while the 10 µM sets possibly represent the native tendency of the receptors toward either agonists or antagonists. Analyzing ligand-GPCR interactions in 56 X-ray structures representative of currently available structural data, we find that the N-terminus, TM1 and TM2 are more involved in binding of antagonists than of agonists. On the other hand, ECL2 tends to be more involved in binding of agonists. This is of interest, since TAS2Rs harbor variations on the typical Class A sequence motifs, including the absence of the ECL2-TM3 disulfide bridge. This suggests an alternative mode of regulation of conformational states for TAS2Rs, with potentially less stabilized inactive state. The comparison of TAS2Rs and Class A GPCRs structural features and the pharmacology of the their ligands highlights the intricacies of GPCR architecture and provides a framework for rational design of new ligands.


Subject(s)
Receptors, G-Protein-Coupled/chemistry , Binding Sites , Humans , Ligands , Models, Molecular , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Taste/drug effects
12.
J Chem Inf Model ; 54(1): 184-94, 2014 Jan 27.
Article in English | MEDLINE | ID: mdl-24328091

ABSTRACT

G protein-coupled receptors (GPCRs) represent a large family of signaling proteins that includes many therapeutic targets. GPCR ligands include odorants, tastants, and neurotransmitters and vary in size and properties. Dramatic chemical diversity may occur even among ligands of the same receptor. Our goal is to unravel the structural and chemical features that determine GPCRs' promiscuity toward their ligands. We perform statistical analysis using more than 30 descriptors related to the sequence, physicochemical, structural, and energetic properties of the GPCR binding sites-we find that the chemical variability of antagonists significantly correlates with the binding site hydrophobicity and anticorrelates with the number of hydrogen bond donors in the binding site. The number of disulfide bridges in the extracellular region of a receptor anticorrelates with the range of molecular weights of its antagonists, highlighting the role of the entrance pathway in determining the size selectivity for GPCR antagonists. The predictive capability of the model is successfully validated using a separate set of GPCRs, using either X-ray structures or homology models.


Subject(s)
Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Animals , Artificial Intelligence , Binding Sites , Computational Biology , Crystallography, X-Ray , Databases, Protein , Humans , Hydrophobic and Hydrophilic Interactions , Least-Squares Analysis , Ligands , Linear Models , Models, Molecular , Principal Component Analysis , Protein Conformation , Receptors, G-Protein-Coupled/antagonists & inhibitors
13.
FASEB J ; 28(3): 1181-97, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24285091

ABSTRACT

Bitter taste receptors (TAS2Rs) mediate aversive response to toxic food, which is often bitter. These G-protein-coupled receptors are also expressed in extraoral tissues, and emerge as novel targets for therapeutic indications such as asthma and infection. Our goal was to identify ligands of the broadly tuned TAS2R14 among clinical drugs. Molecular properties of known human bitter taste receptor TAS2R14 agonists were incorporated into pharmacophore- and shape-based models and used to computationally predict additional ligands. Predictions were tested by calcium imaging of TAS2R14-transfected HEK293 cells. In vitro testing of the virtual screening predictions resulted in 30-80% success rates, and 15 clinical drugs were found to activate the TAS2R14. hERG potassium channel, which is predominantly expressed in the heart, emerged as a common off-target of bitter drugs. Despite immense chemical diversity of known TAS2R14 ligands, novel ligands and previously unknown polypharmacology of drugs were unraveled by in vitro screening of computational predictions. This enables rational repurposing of traditional and standard drugs for bitter taste signaling modulation for therapeutic indications.


Subject(s)
Receptors, G-Protein-Coupled/agonists , HEK293 Cells , Humans , Models, Biological , Structure-Activity Relationship
14.
J Neurosci ; 33(1): 201-13, 2013 Jan 02.
Article in English | MEDLINE | ID: mdl-23283334

ABSTRACT

Bitter taste is a basic taste modality, required to safeguard animals against consuming toxic substances. Bitter compounds are recognized by G-protein-coupled bitter taste receptors (TAS2Rs). The human TAS2R10 responds to the toxic strychnine and numerous other compounds. The mechanism underlying the development of the broad tuning of some TAS2Rs is not understood. Using comparative modeling, site-directed mutagenesis, and functional assays, we identified residues involved in agonist-induced activation of TAS2R10, and investigated the effects of different substitutions on the receptor's response profile. Most interestingly, mutations in S85(3.29) and Q175(5.40) have differential impact on stimulation with different agonists. The fact that single point mutations lead to improved responses for some agonists and to decreased activation by others indicates that the binding site has evolved to optimally accommodate multiple agonists at the expense of reduced potency. TAS2R10 shares the agonist strychnine with TAS2R46, another broadly tuned receptor. Engineering the key determinants for TAS2R46 activation by strychnine in TAS2R10 caused a loss of response to strychnine, indicating that these paralog receptors display different strychnine-binding modes, which suggests independent acquisition of agonist specificities. This implies that the gene duplication event preceding primate speciation was accompanied by independent evolution of the strychnine-binding sites.


Subject(s)
Receptors, G-Protein-Coupled/genetics , Taste Buds/metabolism , Taste/genetics , Biological Evolution , Chloramphenicol/pharmacology , Humans , Ligands , Mutation , Papaverine/pharmacology , Quaternary Ammonium Compounds/pharmacology , Receptors, G-Protein-Coupled/metabolism , Santonin/pharmacology , Strychnine/pharmacology
15.
Methods Mol Biol ; 914: 179-205, 2012.
Article in English | MEDLINE | ID: mdl-22976029

ABSTRACT

G protein-coupled receptors (GPCRs) are important mediators of cell signaling and a major family of drug targets. Despite recent breakthroughs, experimental elucidation of GPCR structures remains a formidable challenge. Homology modeling of 3D structures of GPCRs provides a practical tool for elucidating the structural determinants governing the interactions of these important receptors with their ligands. The working model of the binding site can then be used for virtual screening of additional ligands that may fit this site, for determining and comparing specificity profiles of related receptors, and for structure-based design of agonists and antagonists. The current review presents the protocol and enumerates the steps for modeling and validating the residues involved in ligand binding. The main stages include (a) modeling the receptor structure using an automated fragment-based approach, (b) predicting potential binding pockets, (c) docking known binders, (d) analyzing predicted interactions and comparing with positions that have been shown to bind ligands in other receptors, (e) validating the structural model by mutagenesis.


Subject(s)
Computational Biology/methods , Models, Molecular , Receptors, G-Protein-Coupled/chemistry , Structural Homology, Protein , Amino Acid Sequence , Binding Sites , Humans , Molecular Sequence Data , Mutagenesis/genetics , Mutant Proteins/chemistry , Sequence Alignment
16.
Appl Biochem Biotechnol ; 166(7): 1703-10, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22354633

ABSTRACT

The innate immunity utilizes a battery of broad-spectrum antibacterial cationic polypeptides (3-5 kDa), among them defensins. In humans, defensins are the first line of defense against pathogens and their expression has been implicated in several diseases. The antibacterial activity of defensins is generally ascribed to their overall positive charge, which enables them to disrupt bacterial membrane integrity and function, but their active surface has not been fully elucidated. To perform structural and functional assays, an efficient, high-yield, easy-to-use expression and purification system must be established. Up to now, most efforts to obtain larger quantities of active recombinant defensins have been only moderately successful. Herein, we report the establishment of an efficient, high-yield expression and purification system for human defensin 5 (HD-5). Using site-directed mutagenesis, we pinpoint several arginine residues and Y27 as important for HD-5 antibacterial activity. Our expression and purification system can be harnessed for structure/activity relationship studies of defensins in particular and small polypeptides in general.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gene Expression , alpha-Defensins/chemistry , Amino Acid Sequence , Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/chemistry , Arginine/genetics , Chromatography, High Pressure Liquid , Cloning, Molecular , Escherichia coli , Humans , Microbial Viability/drug effects , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Plasmids , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Static Electricity , Structure-Activity Relationship , alpha-Defensins/genetics , alpha-Defensins/pharmacology
17.
Nucleic Acids Res ; 40(Database issue): D413-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21940398

ABSTRACT

Basic taste qualities like sour, salty, sweet, bitter and umami serve specific functions in identifying food components found in the diet of humans and animals, and are recognized by proteins in the oral cavity. Recognition of bitter taste and aversion to it are thought to protect the organism against the ingestion of poisonous food compounds, which are often bitter. Interestingly, bitter taste receptors are expressed not only in the mouth but also in extraoral tissues, such as the gastrointestinal tract, indicating that they may play a role in digestive and metabolic processes. BitterDB database, available at http://bitterdb.agri.huji.ac.il/bitterdb/, includes over 550 compounds that were reported to taste bitter to humans. The compounds can be searched by name, chemical structure, similarity to other bitter compounds, association with a particular human bitter taste receptor, and so on. The database also contains information on mutations in bitter taste receptors that were shown to influence receptor activation by bitter compounds. The aim of BitterDB is to facilitate studying the chemical features associated with bitterness. These studies may contribute to predicting bitterness of unknown compounds, predicting ligands for bitter receptors from different species and rational design of bitterness modulators.


Subject(s)
Databases, Factual , Taste , Humans , Ligands , Molecular Structure , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Zebrafish Proteins/chemistry
18.
PLoS One ; 6(11): e27990, 2011.
Article in English | MEDLINE | ID: mdl-22132188

ABSTRACT

BACKGROUND AND MOTIVATION: The Prokineticin receptor (PKR) 1 and 2 subtypes are novel members of family A GPCRs, which exhibit an unusually high degree of sequence similarity. Prokineticins (PKs), their cognate ligands, are small secreted proteins of ∼80 amino acids; however, non-peptidic low-molecular weight antagonists have also been identified. PKs and their receptors play important roles under various physiological conditions such as maintaining circadian rhythm and pain perception, as well as regulating angiogenesis and modulating immunity. Identifying binding sites for known antagonists and for additional potential binders will facilitate studying and regulating these novel receptors. Blocking PKRs may serve as a therapeutic tool for various diseases, including acute pain, inflammation and cancer. METHODS AND RESULTS: Ligand-based pharmacophore models were derived from known antagonists, and virtual screening performed on the DrugBank dataset identified potential human PKR (hPKR) ligands with novel scaffolds. Interestingly, these included several HIV protease inhibitors for which endothelial cell dysfunction is a documented side effect. Our results suggest that the side effects might be due to inhibition of the PKR signaling pathway. Docking of known binders to a 3D homology model of hPKR1 is in agreement with the well-established canonical TM-bundle binding site of family A GPCRs. Furthermore, the docking results highlight residues that may form specific contacts with the ligands. These contacts provide structural explanation for the importance of several chemical features that were obtained from the structure-activity analysis of known binders. With the exception of a single loop residue that might be perused in the future for obtaining subtype-specific regulation, the results suggest an identical TM-bundle binding site for hPKR1 and hPKR2. In addition, analysis of the intracellular regions highlights variable regions that may provide subtype specificity.


Subject(s)
Models, Molecular , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/chemistry , Receptors, Peptide/metabolism , Small Molecule Libraries/metabolism , Amino Acid Sequence , Amino Acids/metabolism , Binding Sites , Drug Evaluation, Preclinical , Humans , Hydrogen Bonding , Ligands , Molecular Sequence Data , Protein Binding , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, Peptide/antagonists & inhibitors , Structural Homology, Protein , Structure-Activity Relationship , User-Computer Interface
19.
Curr Opin Drug Discov Devel ; 13(3): 317-25, 2010 May.
Article in English | MEDLINE | ID: mdl-20443165

ABSTRACT

GPCRs are key components of signal transduction pathways and are important drug targets. Recently determined GPCR structures provide opportunities for advancements in GPCR modeling. This review focuses on the choice of experimental templates, the treatment of extracellular loops and the description of ligand-binding sites in GPCR modeling. Four important conclusions are reached in this review: (i) multi-template models may produce better structures than single-template models, although inferior models may also be generated by multi-template approaches, warranting the development and application of improved model assessment methods; (ii) cautious incorporation of knowledge-based constraints can improve the quality of models and docking; (iii) molecular dynamics simulations account for structural features not observed in X-ray structures and may refine docking poses; and (iv) while progress in de novo methods for long loop prediction is ongoing, loopless models provide a practical alternative for docking and virtual screening applications.


Subject(s)
Crystallography, X-Ray/methods , Models, Structural , Receptors, G-Protein-Coupled/chemistry , Sequence Homology, Amino Acid , Binding Sites , Computer Simulation , Drug Discovery/methods , Ligands , Molecular Dynamics Simulation , Protein Conformation
20.
Endocrinology ; 151(4): 1914-22, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20176726

ABSTRACT

The pattern and regulation of endothlin-2 (EDN2) expression and its putative roles in bovine ovaries were investigated. EDN2 mRNA was determined in corpus luteum (CL) and during folliculoluteal transition induced by GnRH in vivo. EDN2 was elevated only in the early CL and was not present in older CL. In the young CL, EDN2 mRNA was identified mainly in luteal cells but not endothelial cells that expressed the EDN1 gene. Similarly, in preovulatory follicles, EDN2 was expressed in the granulosa cells (GCs) and not in the vascular theca interna. LH and hypoxia are two major stimulants of CL formation. Therefore, GCs were cultured with bovine LH, under hypoxic conditions. GCs incubated with bovine LH resulted in increased EDN2 mRNA 42 h later. CoCl2, a hypoxia-mimicking agent, elevated EDN2 in GCs in a dose-dependent manner. Incubation of the human GC line (Simian virus 40 large T antigen) under low oxygen tension (1%) augmented EDN2 6 and 24 h later. In these two cell types, along with EDN2, hypoxia augmented VEGF. EDN2 induced in GCs changes that characterize the developing CL: cell proliferation as well as up-regulation of vascular endothelial growth factor and cyclooxygenase-2 (mRNA and protein levels). Human chorionic gonadotropin also up-regulated these two genes. Small interfering RNA targeting EDN-converting enzyme-1 effectively reduced its mRNA levels. This treatment, expected to lower the mature EDN2 peptide production, inhibited VEGF mRNA levels and GC numbers. Together these data suggest that elevated EDN2 in the early bovine CL, triggered by LH surge and hypoxia, may facilitate CL formation by promoting angiogenesis, cell proliferation, and differentiation.


Subject(s)
Corpus Luteum/growth & development , Endothelin-2/metabolism , Granulosa Cells/metabolism , Hypoxia/metabolism , Luteinizing Hormone/metabolism , Analysis of Variance , Animals , Blotting, Western , Cattle , Cell Proliferation/drug effects , Cells, Cultured , Cobalt/pharmacology , Corpus Luteum/drug effects , Corpus Luteum/metabolism , Dose-Response Relationship, Drug , Endothelin-2/genetics , Female , Gene Expression Regulation , Gene Silencing , Gonadotropin-Releasing Hormone/pharmacology , Granulosa Cells/drug effects , Luteinizing Hormone/pharmacology , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...