Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
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
2.
Cell ; 178(3): 748-761.e17, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31280962

ABSTRACT

Directed evolution, artificial selection toward designed objectives, is routinely used to develop new molecular tools and therapeutics. Successful directed molecular evolution campaigns repeatedly test diverse sequences with a designed selective pressure. Unicellular organisms and their viral pathogens are exceptional for this purpose and have been used for decades. However, many desirable targets of directed evolution perform poorly or unnaturally in unicellular backgrounds. Here, we present a system for facile directed evolution in mammalian cells. Using the RNA alphavirus Sindbis as a vector for heredity and diversity, we achieved 24-h selection cycles surpassing 10-3 mutations per base. Selection is achieved through genetically actuated sequences internal to the host cell, thus the system's name: viral evolution of genetically actuating sequences, or "VEGAS." Using VEGAS, we evolve transcription factors, GPCRs, and allosteric nanobodies toward functional signaling endpoints each in less than 1 weeks' time.


Subject(s)
Directed Molecular Evolution/methods , Allosteric Regulation , Amino Acid Sequence , Animals , Fluorescence Resonance Energy Transfer , Genetic Vectors/genetics , Genetic Vectors/metabolism , HEK293 Cells , Humans , Mutation , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Sequence Alignment , Sindbis Virus/genetics , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/genetics , Single-Domain Antibodies/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Cell ; 172(4): 719-730.e14, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29398112

ABSTRACT

Drugs frequently require interactions with multiple targets-via a process known as polypharmacology-to achieve their therapeutic actions. Currently, drugs targeting several serotonin receptors, including the 5-HT2C receptor, are useful for treating obesity, drug abuse, and schizophrenia. The competing challenges of developing selective 5-HT2C receptor ligands or creating drugs with a defined polypharmacological profile, especially aimed at G protein-coupled receptors (GPCRs), remain extremely difficult. Here, we solved two structures of the 5-HT2C receptor in complex with the highly promiscuous agonist ergotamine and the 5-HT2A-C receptor-selective inverse agonist ritanserin at resolutions of 3.0 Å and 2.7 Å, respectively. We analyzed their respective binding poses to provide mechanistic insights into their receptor recognition and opposing pharmacological actions. This study investigates the structural basis of polypharmacology at canonical GPCRs and illustrates how understanding characteristic patterns of ligand-receptor interaction and activation may ultimately facilitate drug design at multiple GPCRs.


Subject(s)
Ergotamine/chemistry , Receptor, Serotonin, 5-HT2C/chemistry , Ritanserin/chemistry , Serotonin 5-HT2 Receptor Agonists/chemistry , Serotonin 5-HT2 Receptor Antagonists/chemistry , HEK293 Cells , Humans , Obesity/drug therapy , Obesity/metabolism , Protein Domains , Receptor, Serotonin, 5-HT2C/metabolism , Schizophrenia/drug therapy , Schizophrenia/metabolism , Structure-Activity Relationship , Substance-Related Disorders/drug therapy , Substance-Related Disorders/metabolism
4.
Cell ; 172(1-2): 55-67.e15, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29307491

ABSTRACT

The κ-opioid receptor (KOP) mediates the actions of opioids with hallucinogenic, dysphoric, and analgesic activities. The design of KOP analgesics devoid of hallucinatory and dysphoric effects has been hindered by an incomplete structural and mechanistic understanding of KOP agonist actions. Here, we provide a crystal structure of human KOP in complex with the potent epoxymorphinan opioid agonist MP1104 and an active-state-stabilizing nanobody. Comparisons between inactive- and active-state opioid receptor structures reveal substantial conformational changes in the binding pocket and intracellular and extracellular regions. Extensive structural analysis and experimental validation illuminate key residues that propagate larger-scale structural rearrangements and transducer binding that, collectively, elucidate the structural determinants of KOP pharmacology, function, and biased signaling. These molecular insights promise to accelerate the structure-guided design of safer and more effective κ-opioid receptor therapeutics.


Subject(s)
Molecular Docking Simulation , Receptors, Opioid, kappa/chemistry , Analgesics/chemistry , Analgesics/pharmacology , Animals , Binding Sites , HEK293 Cells , Humans , Molecular Dynamics Simulation , Morphinans/chemistry , Morphinans/pharmacology , Protein Binding , Protein Stability , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/metabolism , Sf9 Cells , Spodoptera
5.
Cell ; 170(3): 414-427, 2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28753422

ABSTRACT

G protein-coupled receptors (GPCRs), which are modulated by a variety of endogenous and synthetic ligands, represent the largest family of druggable targets in the human genome. Recent structural and molecular studies have both transformed and expanded classical concepts of receptor pharmacology and have begun to illuminate the distinct mechanisms by which structurally, chemically, and functionally diverse ligands modulate GPCR function. These molecular insights into ligand engagement and action have enabled new computational methods and accelerated the discovery of novel ligands and tool compounds, especially for understudied and orphan GPCRs. These advances promise to streamline the development of GPCR-targeted medications.


Subject(s)
Molecular Targeted Therapy , Receptors, G-Protein-Coupled/metabolism , Animals , Drug Discovery , Humans , Ligands , Models, Molecular , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , Signal Transduction
6.
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
7.
Nature ; 630(8015): 237-246, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38720072

ABSTRACT

Psychedelic substances such as lysergic acid diethylamide (LSD) and psilocybin show potential for the treatment of various neuropsychiatric disorders1-3. These compounds are thought to mediate their hallucinogenic and therapeutic effects through the serotonin (5-hydroxytryptamine (5-HT)) receptor 5-HT2A (ref. 4). However, 5-HT1A also plays a part in the behavioural effects of tryptamine hallucinogens5, particularly 5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT), a psychedelic found in the toxin of Colorado River toads6. Although 5-HT1A is a validated therapeutic target7,8, little is known about how psychedelics engage 5-HT1A and which effects are mediated by this receptor. Here we map the molecular underpinnings of 5-MeO-DMT pharmacology through five cryogenic electron microscopy (cryo-EM) structures of 5-HT1A, systematic medicinal chemistry, receptor mutagenesis and mouse behaviour. Structure-activity relationship analyses of 5-methoxytryptamines at both 5-HT1A and 5-HT2A enable the characterization of molecular determinants of 5-HT1A signalling potency, efficacy and selectivity. Moreover, we contrast the structural interactions and in vitro pharmacology of 5-MeO-DMT and analogues to the pan-serotonergic agonist LSD and clinically used 5-HT1A agonists. We show that a 5-HT1A-selective 5-MeO-DMT analogue is devoid of hallucinogenic-like effects while retaining anxiolytic-like and antidepressant-like activity in socially defeated animals. Our studies uncover molecular aspects of 5-HT1A-targeted psychedelics and therapeutics, which may facilitate the future development of new medications for neuropsychiatric disorders.


Subject(s)
5-Methoxytryptamine , Anti-Anxiety Agents , Antidepressive Agents , Methoxydimethyltryptamines , Receptor, Serotonin, 5-HT1A , Receptor, Serotonin, 5-HT2A , Animals , Humans , Male , Mice , 5-Methoxytryptamine/analogs & derivatives , 5-Methoxytryptamine/chemistry , 5-Methoxytryptamine/pharmacology , 5-Methoxytryptamine/therapeutic use , Anti-Anxiety Agents/chemistry , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Antidepressive Agents/chemistry , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Cryoelectron Microscopy , Hallucinogens , Lysergic Acid Diethylamide/chemistry , Lysergic Acid Diethylamide/pharmacology , Methoxydimethyltryptamines/chemistry , Methoxydimethyltryptamines/pharmacology , Methoxydimethyltryptamines/therapeutic use , Models, Molecular , Receptor, Serotonin, 5-HT1A/chemistry , Receptor, Serotonin, 5-HT1A/genetics , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT1A/ultrastructure , Receptor, Serotonin, 5-HT2A/chemistry , Receptor, Serotonin, 5-HT2A/genetics , Receptor, Serotonin, 5-HT2A/metabolism , Receptor, Serotonin, 5-HT2A/ultrastructure , Serotonin Receptor Agonists/chemistry , Serotonin Receptor Agonists/pharmacology , Serotonin Receptor Agonists/therapeutic use , Structure-Activity Relationship
9.
J Biol Chem ; 299(9): 105176, 2023 09.
Article in English | MEDLINE | ID: mdl-37599003

ABSTRACT

Substance abuse is on the rise, and while many people may use illicit drugs mainly due to their rewarding effects, their societal impact can range from severe, as is the case for opioids, to promising, as is the case for psychedelics. Common with all these drugs' mechanisms of action are G protein-coupled receptors (GPCRs), which lie at the center of how these drugs mediate inebriation, lethality, and therapeutic effects. Opioids like fentanyl, cannabinoids like tetrahydrocannabinol, and psychedelics like lysergic acid diethylamide all directly bind to GPCRs to initiate signaling which elicits their physiological actions. We herein review recent structural studies and provide insights into the molecular mechanisms of opioids, cannabinoids, and psychedelics at their respective GPCR subtypes. We further discuss how such mechanistic insights facilitate drug discovery, either toward the development of novel therapies to combat drug abuse or toward harnessing therapeutic potential.


Subject(s)
Illicit Drugs , Receptors, G-Protein-Coupled , Humans , Analgesics, Opioid/metabolism , Analgesics, Opioid/pharmacology , Cannabinoids/metabolism , Cannabinoids/pharmacology , Hallucinogens/metabolism , Hallucinogens/pharmacology , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Illicit Drugs/metabolism , Illicit Drugs/pharmacology , Models, Molecular , Receptors, Serotonin/metabolism , Drug Development/standards
10.
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
11.
Mol Pharmacol ; 103(1): 1-8, 2023 01.
Article in English | MEDLINE | ID: mdl-36310031

ABSTRACT

Opioid analgesics exert their therapeutic and adverse effects by activating µ opioid receptors (MOPR); however, functional responses to MOPR activation are modulated by distinct signal transduction complexes within the brain. The ventrolateral periaqueductal gray (vlPAG) plays a critical role in modulation of nociception and analgesia, but the exact intracellular pathways associated with opioid responses in this region are not fully understood. We previously showed that knockout of the signal transduction modulator Regulator of G protein Signaling z1 (RGSz1) enhanced analgesic responses to opioids, whereas it decreased the rewarding efficacy of morphine. Here, we applied viral mediated gene transfer methodology and delivered adeno-associated virus (AAV) expressing Cre recombinase to the vlPAG of RGSz1fl\fl mice to demonstrate that downregulation of RGSz1 in this region decreases sensitivity to morphine in the place preference paradigm, under pain-free as well as neuropathic pain states. We also used retrograde viral vectors along with flippase-dependent Cre vectors to conditionally downregulate RGSz1 in vlPAG projections to the ventral tegmental area (VTA) and show that downregulation of RGSz1 prevents the development of place conditioning to low morphine doses. Consistent with the role for RGSz1 as a negative modulator of MOPR activity, RGSz1KO enhances opioid-induced cAMP inhibition in periaqueductal gray (PAG) membranes. Furthermore, using a new generation of bioluminescence resonance energy transfer (BRET) sensors, we demonstrate that RGSz1 modulates Gαz but not other Gαi family subunits and selectively impedes MOPR-mediated Gαz signaling events invoked by morphine and other opioids. Our work highlights a regional and circuit-specific role of the G protein-signaling modulator RGSz1 in morphine reward, providing insights on midbrain intracellular pathways that control addiction-related behaviors. SIGNIFICANCE STATEMENT: This study used advanced genetic mouse models to highlight the role of the signal transduction modulator named RGSz1 in responses to clinically used opioid analgesics. We show that RGSz1 controls the rewarding efficacy of opioids by actions in ventrolateral periaqueductal gray projections to the ventral tegmental area, a key component of the midbrain dopamine pathway. These studies highlight novel mechanisms by which pain-modulating structures control the rewarding efficacy of opioids.


Subject(s)
Analgesics, Opioid , Morphine , Mice , Animals , Morphine/pharmacology , Morphine/metabolism , Analgesics, Opioid/pharmacology , Analgesics, Opioid/metabolism , Periaqueductal Gray/metabolism , Signal Transduction , GTP-Binding Proteins/metabolism , Reward , Receptors, Opioid, mu/metabolism
12.
Med Res Rev ; 41(3): 1427-1473, 2021 05.
Article in English | MEDLINE | ID: mdl-33295676

ABSTRACT

Neurological disorders significantly outnumber diseases in other therapeutic areas. However, developing drugs for central nervous system (CNS) disorders remains the most challenging area in drug discovery, accompanied with the long timelines and high attrition rates. With the rapid growth of biomedical data enabled by advanced experimental technologies, artificial intelligence (AI) and machine learning (ML) have emerged as an indispensable tool to draw meaningful insights and improve decision making in drug discovery. Thanks to the advancements in AI and ML algorithms, now the AI/ML-driven solutions have an unprecedented potential to accelerate the process of CNS drug discovery with better success rate. In this review, we comprehensively summarize AI/ML-powered pharmaceutical discovery efforts and their implementations in the CNS area. After introducing the AI/ML models as well as the conceptualization and data preparation, we outline the applications of AI/ML technologies to several key procedures in drug discovery, including target identification, compound screening, hit/lead generation and optimization, drug response and synergy prediction, de novo drug design, and drug repurposing. We review the current state-of-the-art of AI/ML-guided CNS drug discovery, focusing on blood-brain barrier permeability prediction and implementation into therapeutic discovery for neurological diseases. Finally, we discuss the major challenges and limitations of current approaches and possible future directions that may provide resolutions to these difficulties.


Subject(s)
Artificial Intelligence , Central Nervous System Diseases , Algorithms , Central Nervous System Diseases/drug therapy , Drug Discovery , Humans , Machine Learning
13.
Proc Natl Acad Sci U S A ; 114(31): 8223-8228, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28716900

ABSTRACT

Monoclonal antibodies provide an attractive alternative to small-molecule therapies for a wide range of diseases. Given the importance of G protein-coupled receptors (GPCRs) as pharmaceutical targets, there has been an immense interest in developing therapeutic monoclonal antibodies that act on GPCRs. Here we present the 3.0-Å resolution structure of a complex between the human 5-hydroxytryptamine 2B (5-HT2B) receptor and an antibody Fab fragment bound to the extracellular side of the receptor, determined by serial femtosecond crystallography with an X-ray free-electron laser. The antibody binds to a 3D epitope of the receptor that includes all three extracellular loops. The 5-HT2B receptor is captured in a well-defined active-like state, most likely stabilized by the crystal lattice. The structure of the complex sheds light on the mechanism of selectivity in extracellular recognition of GPCRs by monoclonal antibodies.


Subject(s)
Immunoglobulin Fab Fragments/chemistry , Receptor, Serotonin, 5-HT2B/chemistry , Receptor, Serotonin, 5-HT2B/immunology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Binding, Competitive , Catalytic Domain , Crystallography, X-Ray , Epitopes/chemistry , Epitopes/metabolism , Ergotamine/chemistry , Ergotamine/metabolism , Humans , Immunoglobulin Fab Fragments/metabolism , Models, Molecular , Protein Conformation , Receptor, Serotonin, 5-HT2B/metabolism , Serotonin Receptor Agonists/chemistry , Serotonin Receptor Agonists/metabolism
15.
Nature ; 499(7459): 444-9, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23863937

ABSTRACT

Binding of the glucagon peptide to the glucagon receptor (GCGR) triggers the release of glucose from the liver during fasting; thus GCGR plays an important role in glucose homeostasis. Here we report the crystal structure of the seven transmembrane helical domain of human GCGR at 3.4 Å resolution, complemented by extensive site-specific mutagenesis, and a hybrid model of glucagon bound to GCGR to understand the molecular recognition of the receptor for its native ligand. Beyond the shared seven transmembrane fold, the GCGR transmembrane domain deviates from class A G-protein-coupled receptors with a large ligand-binding pocket and the first transmembrane helix having a 'stalk' region that extends three alpha-helical turns above the plane of the membrane. The stalk positions the extracellular domain (~12 kilodaltons) relative to the membrane to form the glucagon-binding site that captures the peptide and facilitates the insertion of glucagon's amino terminus into the seven transmembrane domain.


Subject(s)
Receptors, Glucagon/chemistry , Receptors, Glucagon/classification , Amino Acid Sequence , Binding Sites , Cell Membrane/metabolism , Crystallography, X-Ray , Glucagon/chemistry , Glucagon/metabolism , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Protein Structure, Tertiary , Receptors, CXCR4/chemistry , Receptors, CXCR4/classification , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism
16.
Nature ; 485(7398): 327-32, 2012 Mar 21.
Article in English | MEDLINE | ID: mdl-22437504

ABSTRACT

Opioid receptors mediate the actions of endogenous and exogenous opioids on many physiological processes, including the regulation of pain, respiratory drive, mood, and--in the case of κ-opioid receptor (κ-OR)--dysphoria and psychotomimesis. Here we report the crystal structure of the human κ-OR in complex with the selective antagonist JDTic, arranged in parallel dimers, at 2.9 Å resolution. The structure reveals important features of the ligand-binding pocket that contribute to the high affinity and subtype selectivity of JDTic for the human κ-OR. Modelling of other important κ-OR-selective ligands, including the morphinan-derived antagonists norbinaltorphimine and 5'-guanidinonaltrindole, and the diterpene agonist salvinorin A analogue RB-64, reveals both common and distinct features for binding these diverse chemotypes. Analysis of site-directed mutagenesis and ligand structure-activity relationships confirms the interactions observed in the crystal structure, thereby providing a molecular explanation for κ-OR subtype selectivity, and essential insights for the design of compounds with new pharmacological properties targeting the human κ-OR.


Subject(s)
Piperidines/chemistry , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, kappa/chemistry , Tetrahydroisoquinolines/chemistry , Binding Sites , Crystallography, X-Ray , Diterpenes, Clerodane/chemistry , Diterpenes, Clerodane/metabolism , Diterpenes, Clerodane/pharmacology , Guanidines/chemistry , Humans , Models, Molecular , Morphinans/chemistry , Mutagenesis, Site-Directed , Naltrexone/analogs & derivatives , Naltrexone/chemistry , Naltrexone/metabolism , Piperidines/pharmacology , Protein Conformation , Receptors, Adrenergic, beta-2/chemistry , Receptors, CXCR4/chemistry , Receptors, CXCR4/metabolism , Receptors, Opioid, kappa/genetics , Receptors, Opioid, kappa/metabolism , Structure-Activity Relationship , Tetrahydroisoquinolines/pharmacology
18.
Nat Commun ; 15(1): 108, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38168118

ABSTRACT

The human trace amine-associated receptor 1 (hTAAR1, hTA1) is a key regulator of monoaminergic neurotransmission and the actions of psychostimulants. Despite preclinical research demonstrating its tractability as a drug target, its molecular mechanisms of activation remain unclear. Moreover, poorly understood pharmacological differences between rodent and human TA1 complicate the translation of findings from preclinical disease models into novel pharmacotherapies. To elucidate hTA1's mechanisms on the molecular scale and investigate the underpinnings of its divergent pharmacology from rodent orthologs, we herein report the structure of the human TA1 receptor in complex with a Gαs heterotrimer. Our structure reveals shared structural elements with other TAARs, as well as with its closest monoaminergic orthologue, the serotonin receptor 5-HT4R. We further find that a single mutation dramatically shifts the selectivity of hTA1 towards that of its rodent orthologues, and report on the effects of substituting residues to those found in serotonin and dopamine receptors. Strikingly, we also discover that the atypical antipsychotic medication and pan-monoaminergic antagonist asenapine potently and efficaciously activates hTA1. Together our studies provide detailed insight into hTA1 structure and function, contrast its molecular pharmacology with that of related receptors, and uncover off-target activities of monoaminergic drugs at hTA1.


Subject(s)
Antipsychotic Agents , Central Nervous System Stimulants , Humans , Receptors, G-Protein-Coupled/chemistry , Synaptic Transmission
19.
Sci Adv ; 10(16): eadk4855, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38630816

ABSTRACT

Serotonin [5-hydroxytryptamine (5-HT)] acts via 13 different receptors in humans. Of these receptor subtypes, all but 5-HT1eR have confirmed roles in native tissue and are validated drug targets. Despite 5-HT1eR's therapeutic potential and plausible druggability, the mechanisms of its activation remain elusive. To illuminate 5-HT1eR's pharmacology in relation to the highly homologous 5-HT1FR, we screened a library of aminergic receptor ligands at both receptors and observe 5-HT1eR/5-HT1FR agonism by multicyclic drugs described as pan-antagonists at 5-HT receptors. Potent agonism by tetracyclic antidepressants mianserin, setiptiline, and mirtazapine suggests a mechanism for their clinically observed antimigraine properties. Using cryo-EM and mutagenesis studies, we uncover and characterize unique agonist-like binding poses of mianserin and setiptiline at 5-HT1eR distinct from similar drug scaffolds in inactive-state 5-HTR structures. Together with computational studies, our data suggest that these binding poses alongside receptor-specific allosteric coupling in 5-HT1eR and 5-HT1FR contribute to the agonist activity of these antidepressants.


Subject(s)
Mianserin , Serotonin , Humans , Mianserin/pharmacology , Antidepressive Agents , Receptors, Serotonin/metabolism , Signal Transduction
20.
bioRxiv ; 2023 Sep 07.
Article in English | MEDLINE | ID: mdl-37986760

ABSTRACT

The human trace amine-associated receptor 1 (hTAAR1, hTA1) is a key regulator of monoaminergic neurotransmission and the actions of psychostimulants. Despite preclinical research demonstrating its tractability as a drug target, its molecular mechanisms of activation remain unclear. Moreover, poorly understood pharmacological differences between rodent and human TA1 complicate the translation of findings from preclinical disease models into novel pharmacotherapies. To elucidate hTA1's mechanisms on the molecular scale and investigate the underpinnings of its divergent pharmacology from rodent orthologs, we herein report the structure of the human TA1 receptor in complex with a Gαs heterotrimer. Our structure reveals shared structural elements with other TAARs, as well as with its closest monoaminergic ortholog, the serotonin receptor 5-HT4R. We further find that a single mutation dramatically shifts the selectivity of hTA1 towards that of its rodent orthologs, and report on the effects of substituting residues to those found in serotonin and dopamine receptors. Strikingly, we also discover that the atypical antipsychotic medication and pan-monoaminergic antagonist asenapine potently and efficaciously activates hTA1. Together our studies provide detailed insight into hTA1 structure and function, contrast its molecular pharmacology with that of related receptors, and uncover off-target activities of monoaminergic drugs at hTA1.

SELECTION OF CITATIONS
SEARCH DETAIL