Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 249
Filter
1.
Cell ; 187(14): 3712-3725.e34, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38810646

ABSTRACT

The cystic fibrosis transmembrane conductance regulator (CFTR) is a crucial ion channel whose loss of function leads to cystic fibrosis, whereas its hyperactivation leads to secretory diarrhea. Small molecules that improve CFTR folding (correctors) or function (potentiators) are clinically available. However, the only potentiator, ivacaftor, has suboptimal pharmacokinetics and inhibitors have yet to be clinically developed. Here, we combine molecular docking, electrophysiology, cryo-EM, and medicinal chemistry to identify CFTR modulators. We docked ∼155 million molecules into the potentiator site on CFTR, synthesized 53 test ligands, and used structure-based optimization to identify candidate modulators. This approach uncovered mid-nanomolar potentiators, as well as inhibitors, that bind to the same allosteric site. These molecules represent potential leads for the development of more effective drugs for cystic fibrosis and secretory diarrhea, demonstrating the feasibility of large-scale docking for ion channel drug discovery.


Subject(s)
Aminophenols , Cystic Fibrosis Transmembrane Conductance Regulator , Cystic Fibrosis , Molecular Docking Simulation , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Humans , Cystic Fibrosis/drug therapy , Cystic Fibrosis/metabolism , Aminophenols/pharmacology , Aminophenols/chemistry , Aminophenols/therapeutic use , Drug Discovery , Cryoelectron Microscopy , Quinolones/pharmacology , Quinolones/chemistry , Quinolones/therapeutic use , Allosteric Site/drug effects , Animals , Ligands
2.
Cell ; 186(10): 2160-2175.e17, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37137306

ABSTRACT

The serotonin transporter (SERT) removes synaptic serotonin and is the target of anti-depressant drugs. SERT adopts three conformations: outward-open, occluded, and inward-open. All known inhibitors target the outward-open state except ibogaine, which has unusual anti-depressant and substance-withdrawal effects, and stabilizes the inward-open conformation. Unfortunately, ibogaine's promiscuity and cardiotoxicity limit the understanding of inward-open state ligands. We docked over 200 million small molecules against the inward-open state of the SERT. Thirty-six top-ranking compounds were synthesized, and thirteen inhibited; further structure-based optimization led to the selection of two potent (low nanomolar) inhibitors. These stabilized an outward-closed state of the SERT with little activity against common off-targets. A cryo-EM structure of one of these bound to the SERT confirmed the predicted geometry. In mouse behavioral assays, both compounds had anxiolytic- and anti-depressant-like activity, with potencies up to 200-fold better than fluoxetine (Prozac), and one substantially reversed morphine withdrawal effects.


Subject(s)
Ibogaine , Selective Serotonin Reuptake Inhibitors , Serotonin Plasma Membrane Transport Proteins , Small Molecule Libraries , Animals , Mice , Fluoxetine/pharmacology , Ibogaine/chemistry , Ibogaine/pharmacology , Molecular Conformation , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/ultrastructure , Selective Serotonin Reuptake Inhibitors/pharmacology , Small Molecule Libraries/pharmacology
3.
Cell ; 182(6): 1574-1588.e19, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32946782

ABSTRACT

Hallucinogens like lysergic acid diethylamide (LSD), psilocybin, and substituted N-benzyl phenylalkylamines are widely used recreationally with psilocybin being considered as a therapeutic for many neuropsychiatric disorders including depression, anxiety, and substance abuse. How psychedelics mediate their actions-both therapeutic and hallucinogenic-are not understood, although activation of the 5-HT2A serotonin receptor (HTR2A) is key. To gain molecular insights into psychedelic actions, we determined the active-state structure of HTR2A bound to 25-CN-NBOH-a prototypical hallucinogen-in complex with an engineered Gαq heterotrimer by cryoelectron microscopy (cryo-EM). We also obtained the X-ray crystal structures of HTR2A complexed with the arrestin-biased ligand LSD or the inverse agonist methiothepin. Comparisons of these structures reveal determinants responsible for HTR2A-Gαq protein interactions as well as the conformational rearrangements involved in active-state transitions. Given the potential therapeutic actions of hallucinogens, these findings could accelerate the discovery of more selective drugs for the treatment of a variety of neuropsychiatric disorders.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , Hallucinogens/chemistry , Receptor, Serotonin, 5-HT2A/chemistry , Receptor, Serotonin, 5-HT2A/metabolism , Animals , Cryoelectron Microscopy , Crystallography, X-Ray , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Gene Expression , HEK293 Cells , Hallucinogens/pharmacology , Hallucinogens/therapeutic use , Humans , Ligands , Lysergic Acid Diethylamide/chemistry , Lysergic Acid Diethylamide/pharmacology , Methiothepin/chemistry , Methiothepin/metabolism , Models, Chemical , Mutation , Protein Conformation, alpha-Helical , Receptor, Serotonin, 5-HT2A/genetics , Recombinant Proteins , Serotonin/metabolism , Spodoptera
4.
Cell ; 174(3): 505-520, 2018 07 26.
Article in English | MEDLINE | ID: mdl-30053424

ABSTRACT

Although gene discovery in neuropsychiatric disorders, including autism spectrum disorder, intellectual disability, epilepsy, schizophrenia, and Tourette disorder, has accelerated, resulting in a large number of molecular clues, it has proven difficult to generate specific hypotheses without the corresponding datasets at the protein complex and functional pathway level. Here, we describe one path forward-an initiative aimed at mapping the physical and genetic interaction networks of these conditions and then using these maps to connect the genomic data to neurobiology and, ultimately, the clinic. These efforts will include a team of geneticists, structural biologists, neurobiologists, systems biologists, and clinicians, leveraging a wide array of experimental approaches and creating a collaborative infrastructure necessary for long-term investigation. This initiative will ultimately intersect with parallel studies that focus on other diseases, as there is a significant overlap with genes implicated in cancer, infectious disease, and congenital heart defects.


Subject(s)
Chromosome Mapping/methods , Neurodevelopmental Disorders/genetics , Systems Biology/methods , Gene Regulatory Networks/genetics , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study/methods , Genomics/methods , Humans , Neurobiology/methods , Neuropsychiatry
5.
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
6.
Nature ; 610(7932): 582-591, 2022 10.
Article in English | MEDLINE | ID: mdl-36171289

ABSTRACT

There is considerable interest in screening ultralarge chemical libraries for ligand discovery, both empirically and computationally1-4. Efforts have focused on readily synthesizable molecules, inevitably leaving many chemotypes unexplored. Here we investigate structure-based docking of a bespoke virtual library of tetrahydropyridines-a scaffold that is poorly sampled by a general billion-molecule virtual library but is well suited to many aminergic G-protein-coupled receptors. Using three inputs, each with diverse available derivatives, a one pot C-H alkenylation, electrocyclization and reduction provides the tetrahydropyridine core with up to six sites of derivatization5-7. Docking a virtual library of 75 million tetrahydropyridines against a model of the serotonin 5-HT2A receptor (5-HT2AR) led to the synthesis and testing of 17 initial molecules. Four of these molecules had low-micromolar activities against either the 5-HT2A or the 5-HT2B receptors. Structure-based optimization led to the 5-HT2AR agonists (R)-69 and (R)-70, with half-maximal effective concentration values of 41 nM and 110 nM, respectively, and unusual signalling kinetics that differ from psychedelic 5-HT2AR agonists. Cryo-electron microscopy structural analysis confirmed the predicted binding mode to 5-HT2AR. The favourable physical properties of these new agonists conferred high brain permeability, enabling mouse behavioural assays. Notably, neither had psychedelic activity, in contrast to classic 5-HT2AR agonists, whereas both had potent antidepressant activity in mouse models and had the same efficacy as antidepressants such as fluoxetine at as low as 1/40th of the dose. Prospects for using bespoke virtual libraries to sample pharmacologically relevant chemical space will be considered.


Subject(s)
Antidepressive Agents , Pyrrolidines , Receptor, Serotonin, 5-HT2A , Animals , Mice , Antidepressive Agents/pharmacology , Cryoelectron Microscopy , Fluoxetine/administration & dosage , Fluoxetine/pharmacology , Hallucinogens/administration & dosage , Hallucinogens/pharmacology , Ligands , Pyrrolidines/administration & dosage , Pyrrolidines/pharmacology , Receptor, Serotonin, 5-HT2A/metabolism , Small Molecule Libraries
7.
Nature ; 600(7890): 759-764, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34880501

ABSTRACT

The σ2 receptor has attracted intense interest in cancer imaging1, psychiatric disease2, neuropathic pain3-5 and other areas of biology6,7. Here we determined the crystal structure of this receptor in complex with the clinical candidate roluperidone2 and the tool compound PB288. These structures templated a large-scale docking screen of 490 million virtual molecules, of which 484 compounds were synthesized and tested. We identified 127 new chemotypes with affinities superior to 1 µM, 31 of which had affinities superior to 50 nM. The hit rate fell smoothly and monotonically with docking score. We optimized three hits for potency and selectivity, and achieved affinities that ranged from 3 to 48 nM, with up to 250-fold selectivity versus the σ1 receptor. Crystal structures of two ligands bound to the σ2 receptor confirmed the docked poses. To investigate the contribution of the σ2 receptor in pain, two potent σ2-selective ligands and one potent σ1/σ2 non-selective ligand were tested for efficacy in a mouse model of neuropathic pain. All three ligands showed time-dependent decreases in mechanical hypersensitivity in the spared nerve injury model9, suggesting that the σ2 receptor has a role in nociception. This study illustrates the opportunities for rapid discovery of in vivo probes through structure-based screens of ultra large libraries, enabling study of underexplored areas of biology.


Subject(s)
Neuralgia , Receptors, sigma , Animals , Ligands , Mice , Neuralgia/drug therapy , Receptors, sigma/metabolism , Structure-Activity Relationship
8.
Nature ; 600(7887): 170-175, 2021 12.
Article in English | MEDLINE | ID: mdl-34789874

ABSTRACT

The MRGPRX family of receptors (MRGPRX1-4) is a family of mas-related G-protein-coupled receptors that have evolved relatively recently1. Of these, MRGPRX2 and MRGPRX4 are key physiological and pathological mediators of itch and related mast cell-mediated hypersensitivity reactions2-5. MRGPRX2 couples to both Gi and Gq in mast cells6. Here we describe agonist-stabilized structures of MRGPRX2 coupled to Gi1 and Gq in ternary complexes with the endogenous peptide cortistatin-14 and with a synthetic agonist probe, respectively, and the development of potent antagonist probes for MRGPRX2. We also describe a specific MRGPRX4 agonist and the structure of this agonist in a complex with MRGPRX4 and Gq. Together, these findings should accelerate the structure-guided discovery of therapeutic agents for pain, itch and mast cell-mediated hypersensitivity.


Subject(s)
Cryoelectron Microscopy , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/chemistry , Pruritus/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Neuropeptide/chemistry , Drug Inverse Agonism , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/ultrastructure , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/ultrastructure , Humans , Models, Molecular , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/ultrastructure , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/ultrastructure , Receptors, Neuropeptide/metabolism , Receptors, Neuropeptide/ultrastructure
9.
Nat Chem Biol ; 2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38528119

ABSTRACT

The µ-opioid receptor (µOR) represents an important target of therapeutic and abused drugs. So far, most understanding of µOR activity has focused on a subset of known signal transducers and regulatory molecules. Yet µOR signaling is coordinated by additional proteins in the interaction network of the activated receptor, which have largely remained invisible given the lack of technologies to interrogate these networks systematically. Here we describe a proteomics and computational approach to map the proximal proteome of the activated µOR and to extract subcellular location, trafficking and functional partners of G-protein-coupled receptor (GPCR) activity. We demonstrate that distinct opioid agonists exert differences in the µOR proximal proteome mediated by endocytosis and endosomal sorting. Moreover, we identify two new µOR network components, EYA4 and KCTD12, which are recruited on the basis of receptor-triggered G-protein activation and might form a previously unrecognized buffering system for G-protein activity broadly modulating cellular GPCR signaling.

10.
Nature ; 579(7800): 609-614, 2020 03.
Article in English | MEDLINE | ID: mdl-32040955

ABSTRACT

The neuromodulator melatonin synchronizes circadian rhythms and related physiological functions through the actions of two G-protein-coupled receptors: MT1 and MT2. Circadian release of melatonin at night from the pineal gland activates melatonin receptors in the suprachiasmatic nucleus of the hypothalamus, synchronizing the physiology and behaviour of animals to the light-dark cycle1-4. The two receptors are established drug targets for aligning circadian phase to this cycle in disorders of sleep5,6 and depression1-4,7-9. Despite their importance, few in vivo active MT1-selective ligands have been reported2,8,10-12, hampering both the understanding of circadian biology and the development of targeted therapeutics. Here we docked more than 150 million virtual molecules to an MT1 crystal structure, prioritizing structural fit and chemical novelty. Of these compounds, 38 high-ranking molecules were synthesized and tested, revealing ligands with potencies ranging from 470 picomolar to 6 micromolar. Structure-based optimization led to two selective MT1 inverse agonists-which were topologically unrelated to previously explored chemotypes-that acted as inverse agonists in a mouse model of circadian re-entrainment. Notably, we found that these MT1-selective inverse agonists advanced the phase of the mouse circadian clock by 1.3-1.5 h when given at subjective dusk, an agonist-like effect that was eliminated in MT1- but not in MT2-knockout mice. This study illustrates the opportunities for modulating melatonin receptor biology through MT1-selective ligands and for the discovery of previously undescribed, in vivo active chemotypes from structure-based screens of diverse, ultralarge libraries.


Subject(s)
Circadian Rhythm/physiology , Ligands , Receptors, Melatonin/agonists , Receptors, Melatonin/metabolism , Animals , Circadian Rhythm/drug effects , Darkness , Drug Evaluation, Preclinical , Drug Inverse Agonism , Female , Humans , Light , Male , Mice , Mice, Knockout , Molecular Docking Simulation , Receptor, Melatonin, MT1/agonists , Receptor, Melatonin, MT1/deficiency , Receptor, Melatonin, MT1/genetics , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/agonists , Receptor, Melatonin, MT2/deficiency , Receptor, Melatonin, MT2/genetics , Receptor, Melatonin, MT2/metabolism , Receptors, Melatonin/deficiency , Receptors, Melatonin/genetics , Small Molecule Libraries/pharmacology , Substrate Specificity/genetics
11.
Proc Natl Acad Sci U S A ; 120(2): e2212931120, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36598939

ABSTRACT

The nonstructural protein 3 (NSP3) of the severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) contains a conserved macrodomain enzyme (Mac1) that is critical for pathogenesis and lethality. While small-molecule inhibitors of Mac1 have great therapeutic potential, at the outset of the COVID-19 pandemic, there were no well-validated inhibitors for this protein nor, indeed, the macrodomain enzyme family, making this target a pharmacological orphan. Here, we report the structure-based discovery and development of several different chemical scaffolds exhibiting low- to sub-micromolar affinity for Mac1 through iterations of computer-aided design, structural characterization by ultra-high-resolution protein crystallography, and binding evaluation. Potent scaffolds were designed with in silico fragment linkage and by ultra-large library docking of over 450 million molecules. Both techniques leverage the computational exploration of tangible chemical space and are applicable to other pharmacological orphans. Overall, 160 ligands in 119 different scaffolds were discovered, and 153 Mac1-ligand complex crystal structures were determined, typically to 1 Å resolution or better. Our analyses discovered selective and cell-permeable molecules, unexpected ligand-mediated conformational changes within the active site, and key inhibitor motifs that will template future drug development against Mac1.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Crystallography , Pandemics , Ligands , Molecular Docking Simulation , Protease Inhibitors/pharmacology , Antiviral Agents/pharmacology , Antiviral Agents/chemistry
12.
Nat Chem Biol ; 19(6): 712-718, 2023 06.
Article in English | MEDLINE | ID: mdl-36646956

ABSTRACT

Recently, 'tangible' virtual libraries have made billions of molecules readily available. Prioritizing these molecules for synthesis and testing demands computational approaches, such as docking. Their success may depend on library diversity, their similarity to bio-like molecules and how receptor fit and artifacts change with library size. We compared a library of 3 million 'in-stock' molecules with billion-plus tangible libraries. The bias toward bio-like molecules in the tangible library decreases 19,000-fold versus those 'in-stock'. Similarly, thousands of high-ranking molecules, including experimental actives, from five ultra-large-library docking campaigns are also dissimilar to bio-like molecules. Meanwhile, better-fitting molecules are found as the library grows, with the score improving log-linearly with library size. Finally, as library size increases, so too do rare molecules that rank artifactually well. Although the nature of these artifacts changes from target to target, the expectation of their occurrence does not, and simple strategies can minimize their impact.


Subject(s)
Libraries, Digital , Molecular Docking Simulation
13.
Nat Chem Biol ; 19(4): 416-422, 2023 04.
Article in English | MEDLINE | ID: mdl-36302898

ABSTRACT

The human MAS-related G protein-coupled receptor X1 (MRGPRX1) is preferentially expressed in the small-diameter primary sensory neurons and involved in the mediation of nociception and pruritus. Central activation of MRGPRX1 by the endogenous opioid peptide fragment BAM8-22 and its positive allosteric modulator ML382 has been shown to effectively inhibit persistent pain, making MRGPRX1 a promising target for non-opioid pain treatment. However, the activation mechanism of MRGPRX1 is still largely unknown. Here we report three high-resolution cryogenic electron microscopy structures of MRGPRX1-Gαq in complex with BAM8-22 alone, with BAM8-22 and ML382 simultaneously as well as with a synthetic agonist compound-16. These structures reveal the agonist binding mode for MRGPRX1 and illuminate the structural requirements for positive allosteric modulation. Collectively, our findings provide a molecular understanding of the activation and allosteric modulation of the MRGPRX1 receptor, which could facilitate the structure-based design of non-opioid pain-relieving drugs.


Subject(s)
Pain , Receptors, G-Protein-Coupled , Humans , Ligands , Receptors, G-Protein-Coupled/metabolism , Allosteric Regulation , Allosteric Site
14.
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
15.
J Chem Inf Model ; 64(2): 425-434, 2024 01 22.
Article in English | MEDLINE | ID: mdl-38191997

ABSTRACT

Discovering ligands for amyloid fibrils, such as those formed by the tau protein, is an area of great current interest. In recent structures, ligands bind in stacks in the tau fibrils to reflect the rotational and translational symmetry of the fibril itself; in these structures, the ligands make few interactions with the protein but interact extensively with each other. To exploit this symmetry and stacking, we developed SymDOCK, a method to dock molecules that follow the protein's symmetry. For each prospective ligand pose, we apply the symmetry operation of the fibril to generate a self-interacting and fibril-interacting stack, checking that doing so will not cause a clash between the original molecule and its image. Absent a clash, we retain that pose and add the ligand-ligand van der Waals energy to the ligand's docking score (here using DOCK3.8). We can check these geometries and energies using an implementation of ANI, a neural-network-based quantum-mechanical evaluation of the ligand stacking energies. In retrospective calculations, symmetry docking can reproduce the poses of three tau PET tracers whose structures have been determined. More convincingly, in a prospective study, SymDOCK predicted the structure of the PET tracer MK-6240 bound in a symmetrical stack to AD PHF tau before that structure was determined; the docked pose was used to determine how MK-6240 fit the cryo-EM density. In proof-of-concept studies, SymDOCK enriched known ligands over property-matched decoys in retrospective screens without sacrificing docking speed and can address large library screens that seek new symmetrical stackers. Future applications of this approach will be considered.


Subject(s)
Proteins , Prospective Studies , Ligands , Retrospective Studies , Proteins/chemistry , Molecular Docking Simulation , Protein Binding , Binding Sites
16.
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
17.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Article in English | MEDLINE | ID: mdl-34475217

ABSTRACT

Protein flexibility remains a major challenge in library docking because of difficulties in sampling conformational ensembles with accurate probabilities. Here, we use the model cavity site of T4 lysozyme L99A to test flexible receptor docking with energy penalties from molecular dynamics (MD) simulations. Crystallography with larger and smaller ligands indicates that this cavity can adopt three major conformations: open, intermediate, and closed. Since smaller ligands typically bind better to the cavity site, we anticipate an energy penalty for the cavity opening. To estimate its magnitude, we calculate conformational preferences from MD simulations. We find that including a penalty term is essential for retrospective ligand enrichment; otherwise, high-energy states dominate the docking. We then prospectively docked a library of over 900,000 compounds for new molecules binding to each conformational state. Absent a penalty term, the open conformation dominated the docking results; inclusion of this term led to a balanced sampling of ligands against each state. High ranked molecules were experimentally tested by Tm upshift and X-ray crystallography. From 33 selected molecules, we identified 18 ligands and determined 13 crystal structures. Most interesting were those bound to the open cavity, where the buried site opens to bulk solvent. Here, highly unusual ligands for this cavity had been predicted, including large ligands with polar tails; these were confirmed both by binding and by crystallography. In docking, incorporating protein flexibility with thermodynamic weightings may thus access new ligand chemotypes. The MD approach to accessing and, crucially, weighting such alternative states may find general applicability.


Subject(s)
Proteins/metabolism , Binding Sites , Crystallography, X-Ray , Kinetics , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Protein Conformation , Proteins/chemistry , Retrospective Studies , Thermodynamics
18.
Proc Natl Acad Sci U S A ; 117(27): 16009-16018, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32571913

ABSTRACT

Food and drug products contain diverse and abundant small-molecule additives (excipients) with unclear impacts on human physiology, drug safety, and response. Here, we evaluate their potential impact on intestinal drug absorption. By screening 136 unique compounds for inhibition of the key intestinal transporter OATP2B1 we identified and validated 24 potent OATP2B1 inhibitors, characterized by higher molecular weight and hydrophobicity compared to poor or noninhibitors. OATP2B1 inhibitors were also enriched for dyes, including 8 azo (R-N=N-R') dyes. Pharmacokinetic studies in mice confirmed that FD&C Red No. 40, a common azo dye excipient and a potent inhibitor of OATP2B1, decreased the plasma level of the OATP2B1 substrate fexofenadine, suggesting that FD&C Red No. 40 has the potential to block drug absorption through OATP2B1 inhibition in vivo. However, the gut microbiomes of multiple unrelated healthy individuals as well as diverse human gut bacterial isolates were capable of inactivating the identified azo dye excipients, producing metabolites that no longer inhibit OATP2B1 transport. These results support a beneficial role for the microbiome in limiting the unintended effects of food and drug additives in the intestine and provide a framework for the data-driven selection of excipients. Furthermore, the ubiquity and genetic diversity of gut bacterial azoreductases coupled to experiments in conventionally raised and gnotobiotic mice suggest that variations in gut microbial community structure may be less important to consider relative to the high concentrations of azo dyes in food products, which have the potential to saturate gut bacterial enzymatic activity.


Subject(s)
Bacteria/metabolism , Excipients/metabolism , Food Additives/metabolism , Food , Gastrointestinal Microbiome/physiology , Intestinal Absorption/physiology , Organic Anion Transporters/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Anti-Allergic Agents/metabolism , Anti-Allergic Agents/pharmacokinetics , Azo Compounds , Bacteria/isolation & purification , Excipients/pharmacokinetics , Female , Food Additives/pharmacokinetics , Histamine H1 Antagonists, Non-Sedating/metabolism , Histamine H1 Antagonists, Non-Sedating/pharmacokinetics , Humans , Intestinal Absorption/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Terfenadine/analogs & derivatives , ATP-Binding Cassette Sub-Family B Member 4
19.
Nat Chem Biol ; 16(7): 749-755, 2020 07.
Article in English | MEDLINE | ID: mdl-32483378

ABSTRACT

Most drugs acting on G-protein-coupled receptors target the orthosteric binding pocket where the native hormone or neurotransmitter binds. There is much interest in finding allosteric ligands for these targets because they modulate physiologic signaling and promise to be more selective than orthosteric ligands. Here we describe a newly developed allosteric modulator of the ß2-adrenergic receptor (ß2AR), AS408, that binds to the membrane-facing surface of transmembrane segments 3 and 5, as revealed by X-ray crystallography. AS408 disrupts a water-mediated polar network involving E1223.41 and the backbone carbonyls of V2065.45 and S2075.46. The AS408 binding site is adjacent to a previously identified molecular switch for ß2AR activation formed by I3.40, P5.50 and F6.44. The structure reveals how AS408 stabilizes the inactive conformation of this switch, thereby acting as a negative allosteric modulator for agonists and positive allosteric modulator for inverse agonists.


Subject(s)
Adrenergic beta-2 Receptor Agonists/chemistry , Adrenergic beta-Antagonists/chemistry , Alprenolol/chemistry , Norepinephrine/chemistry , Receptors, Adrenergic, beta-2/chemistry , Salmeterol Xinafoate/chemistry , Adrenergic beta-2 Receptor Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Allosteric Regulation , Allosteric Site , Alprenolol/pharmacology , HEK293 Cells , Humans , Kinetics , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Norepinephrine/pharmacology , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Receptors, Adrenergic, beta-2/metabolism , Salmeterol Xinafoate/pharmacology , Thermodynamics , Water/chemistry
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL