Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
Nature ; 589(7842): 474-479, 2021 01.
Article in English | MEDLINE | ID: mdl-33299186

ABSTRACT

The psychedelic alkaloid ibogaine has anti-addictive properties in both humans and animals1. Unlike most medications for the treatment of substance use disorders, anecdotal reports suggest that ibogaine has the potential to treat addiction to various substances, including opiates, alcohol and psychostimulants. The effects of ibogaine-like those of other psychedelic compounds-are long-lasting2, which has been attributed to its ability to modify addiction-related neural circuitry through the activation of neurotrophic factor signalling3,4. However, several safety concerns have hindered the clinical development of ibogaine, including its toxicity, hallucinogenic potential and tendency to induce cardiac arrhythmias. Here we apply the principles of function-oriented synthesis to identify the key structural elements of the potential therapeutic pharmacophore of ibogaine, and we use this information to engineer tabernanthalog-a water-soluble, non-hallucinogenic, non-toxic analogue of ibogaine that can be prepared in a single step. In rodents, tabernanthalog was found to promote structural neural plasticity, reduce alcohol- and heroin-seeking behaviour, and produce antidepressant-like effects. This work demonstrates that, through careful chemical design, it is possible to modify a psychedelic compound to produce a safer, non-hallucinogenic variant that has therapeutic potential.


Subject(s)
Behavior, Addictive/drug therapy , Drug Design , Ibogaine/analogs & derivatives , Ibogaine/adverse effects , Alcoholism/drug therapy , Animals , Antidepressive Agents/pharmacology , Arrhythmias, Cardiac/chemically induced , Chemistry Techniques, Synthetic , Depression/drug therapy , Disease Models, Animal , Female , Hallucinogens/adverse effects , Heroin Dependence/drug therapy , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity/drug effects , Patient Safety , Receptor, Serotonin, 5-HT2A/metabolism , Serotonin 5-HT2 Receptor Agonists/pharmacology , Substance-Related Disorders/drug therapy , Swimming , Tabernaemontana/chemistry
7.
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
8.
J Biol Chem ; 300(1): 105549, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38072064

ABSTRACT

G protein-coupled receptors (GPCRs) are leading druggable targets for several medicines, but many GPCRs are still untapped for their therapeutic potential due to poor understanding of specific signaling properties. The complement C3a receptor 1 (C3aR1) has been extensively studied for its physiological role in C3a-mediated anaphylaxis/inflammation, and in TLQP-21-mediated lipolysis, but direct evidence for the functional relevance of the C3a and TLQP-21 ligands and signal transduction mechanisms are still limited. In addition, C3aR1 G protein coupling specificity is still unclear, and whether endogenous ligands, or drug-like compounds, show ligand-mediated biased agonism is unknown. Here, we demonstrate that C3aR1 couples preferentially to Gi/o/z proteins and can recruit ß-arrestins to cause internalization. Furthermore, we showed that in comparison to C3a63-77, TLQP-21 exhibits a preference toward Gi/o-mediated signaling compared to ß-arrestin recruitment and internalization. We also show that the purported antagonist SB290157 is a very potent C3aR1 agonist, where antagonism of ligand-stimulated C3aR1 calcium flux is caused by potent ß-arrestin-mediated internalization. Finally, ligand-mediated signaling bias impacted cell function as demonstrated by the regulation of calcium influx, lipolysis in adipocytes, phagocytosis in microglia, and degranulation in mast cells. Overall, we characterize C3aR1 as a Gi/o/z-coupled receptor and demonstrate the functional relevance of ligand-mediated signaling bias in key cellular models. Due to C3aR1 and its endogenous ligands being implicated in inflammatory and metabolic diseases, these results are of relevance toward future C3aR1 drug discovery.


Subject(s)
Calcium , Complement C3a , beta-Arrestin 1/metabolism , beta-Arrestins/metabolism , Calcium/metabolism , Complement C3a/metabolism , Ligands , Receptors, G-Protein-Coupled/metabolism , Humans , Animals , Mice , Cell Line
9.
10.
Nature ; 569(7755): 289-292, 2019 05.
Article in English | MEDLINE | ID: mdl-31019305

ABSTRACT

The human MT1 and MT2 melatonin receptors1,2 are G-protein-coupled receptors (GPCRs) that help to regulate circadian rhythm and sleep patterns3. Drug development efforts have targeted both receptors for the treatment of insomnia, circadian rhythm and mood disorders, and cancer3, and MT2 has also been implicated in type 2 diabetes4,5. Here we report X-ray free electron laser (XFEL) structures of the human MT2 receptor in complex with the agonists 2-phenylmelatonin (2-PMT) and ramelteon6 at resolutions of 2.8 Å and 3.3 Å, respectively, along with two structures of function-related mutants: H2085.46A (superscripts represent the Ballesteros-Weinstein residue numbering nomenclature7) and N862.50D, obtained in complex with 2-PMT. Comparison of the structures of MT2 with a published structure8 of MT1 reveals that, despite conservation of the orthosteric ligand-binding site residues, there are notable conformational variations as well as differences in [3H]melatonin dissociation kinetics that provide insights into the selectivity between melatonin receptor subtypes. A membrane-buried lateral ligand entry channel is observed in both MT1 and MT2, but in addition the MT2 structures reveal a narrow opening towards the solvent in the extracellular part of the receptor. We provide functional and kinetic data that support a prominent role for intramembrane ligand entry in both receptors, and suggest that there might also be an extracellular entry path in MT2. Our findings contribute to a molecular understanding of melatonin receptor subtype selectivity and ligand access modes, which are essential for the design of highly selective melatonin tool compounds and therapeutic agents.


Subject(s)
Electrons , Lasers , Models, Molecular , Receptor, Melatonin, MT2/chemistry , Receptor, Melatonin, MT2/metabolism , Crystallization , Diabetes Mellitus, Type 2/genetics , Humans , Indenes/chemistry , Indenes/metabolism , Ligands , Melatonin/analogs & derivatives , Melatonin/chemistry , Melatonin/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Mutation , Receptor, Melatonin, MT1/chemistry , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/genetics , Structure-Activity Relationship , Substrate Specificity
11.
Nature ; 569(7755): 284-288, 2019 05.
Article in English | MEDLINE | ID: mdl-31019306

ABSTRACT

Melatonin (N-acetyl-5-methoxytryptamine) is a neurohormone that maintains circadian rhythms1 by synchronization to environmental cues and is involved in diverse physiological processes2 such as the regulation of blood pressure and core body temperature, oncogenesis, and immune function3. Melatonin is formed in the pineal gland in a light-regulated manner4 by enzymatic conversion from 5-hydroxytryptamine (5-HT or serotonin), and modulates sleep and wakefulness5 by activating two high-affinity G-protein-coupled receptors, type 1A (MT1) and type 1B (MT2)3,6. Shift work, travel, and ubiquitous artificial lighting can disrupt natural circadian rhythms; as a result, sleep disorders affect a substantial population in modern society and pose a considerable economic burden7. Over-the-counter melatonin is widely used to alleviate jet lag and as a safer alternative to benzodiazepines and other sleeping aids8,9, and is one of the most popular supplements in the United States10. Here, we present high-resolution room-temperature X-ray free electron laser (XFEL) structures of MT1 in complex with four agonists: the insomnia drug ramelteon11, two melatonin analogues, and the mixed melatonin-serotonin antidepressant agomelatine12,13. The structure of MT2 is described in an accompanying paper14. Although the MT1 and 5-HT receptors have similar endogenous ligands, and agomelatine acts on both receptors, the receptors differ markedly in the structure and composition of their ligand pockets; in MT1, access to the ligand pocket is tightly sealed from solvent by extracellular loop 2, leaving only a narrow channel between transmembrane helices IV and V that connects it to the lipid bilayer. The binding site is extremely compact, and ligands interact with MT1 mainly by strong aromatic stacking with Phe179 and auxiliary hydrogen bonds with Asn162 and Gln181. Our structures provide an unexpected example of atypical ligand entry for a non-lipid receptor, lay the molecular foundation of ligand recognition by melatonin receptors, and will facilitate the design of future tool compounds and therapeutic agents, while their comparison to 5-HT receptors yields insights into the evolution and polypharmacology of G-protein-coupled receptors.


Subject(s)
Electrons , Lasers , Models, Molecular , Receptor, Melatonin, MT1/chemistry , Receptor, Melatonin, MT1/metabolism , Acetamides/chemistry , Acetamides/metabolism , Amino Acid Sequence , Antidepressive Agents/chemistry , Antidepressive Agents/metabolism , Crystallization , Humans , Indenes/chemistry , Indenes/metabolism , Ligands , Melatonin/analogs & derivatives , Melatonin/chemistry , Molecular Docking Simulation , Mutation , Receptor, Melatonin, MT1/agonists , Receptor, Melatonin, MT1/genetics , Receptor, Serotonin, 5-HT2C/chemistry , Structure-Activity Relationship , Substrate Specificity
12.
J Neurosci ; 43(45): 7472-7482, 2023 11 08.
Article in English | MEDLINE | ID: mdl-37940583

ABSTRACT

Serotonergic psychedelics, such as psilocybin and LSD, have garnered significant attention in recent years for their potential therapeutic effects and unique mechanisms of action. These compounds exert their primary effects through activating serotonin 5-HT2A receptors, found predominantly in cortical regions. By interacting with these receptors, serotonergic psychedelics induce alterations in perception, cognition, and emotions, leading to the characteristic psychedelic experience. One of the most crucial aspects of serotonergic psychedelics is their ability to promote neuroplasticity, the formation of new neural connections, and rewire neuronal networks. This neuroplasticity is believed to underlie their therapeutic potential for various mental health conditions, including depression, anxiety, and substance use disorders. In this mini-review, we will discuss how the 5-HT2A receptor activation is just one facet of the complex mechanisms of action of serotonergic psychedelics. They also interact with other serotonin receptor subtypes, such as 5-HT1A and 5-HT2C receptors, and with neurotrophin receptors (e.g., tropomyosin receptor kinase B). These interactions contribute to the complexity of their effects on perception, mood, and cognition. Moreover, as psychedelic research advances, there is an increasing interest in developing nonhallucinogenic derivatives of these drugs to create safer and more targeted medications for psychiatric disorders by removing the hallucinogenic properties while retaining the potential therapeutic benefits. These nonhallucinogenic derivatives would offer patients therapeutic advantages without the intense psychedelic experience, potentially reducing the risks of adverse reactions. Finally, we discuss the potential of psychedelics as substrates for post-translational modification of proteins as part of their mechanism of action.


Subject(s)
Hallucinogens , Humans , Hallucinogens/pharmacology , Serotonin , Receptor, Serotonin, 5-HT2A , Psilocybin , Anxiety
13.
Mol Pharmacol ; 106(2): 92-106, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38821630

ABSTRACT

Bipolar disorder impacts millions of patients in the United States but the mechanistic understanding of its pathophysiology and therapeutics is incomplete. Atypical antipsychotic serotonin2A (5-HT2A) receptor antagonists, such as quetiapine and olanzapine, and mood-stabilizing voltage-gated sodium channel (VGSC) blockers, such as lamotrigine, carbamazepine, and valproate, show therapeutic synergy and are often prescribed in combination for the treatment of bipolar disorder. Combination therapy is a complex task for clinicians and patients, often resulting in unexpected difficulties with dosing, drug tolerances, and decreased patient compliance. Thus, an unmet need for bipolar disorder treatment is to develop a therapeutic agent that targets both 5-HT2A receptors and VGSCs. Toward this goal, we developed a novel small molecule that simultaneously antagonizes 5-HT2A receptors and blocks sodium current. The new compound, N-(4-bromo-2,5-dimethoxyphenethyl)-6-(4-phenylbutoxy)hexan-1-amine (XOB) antagonizes 5-HT-stimulated, Gq-mediated, calcium flux at 5-HT2A receptors at low micromolar concentrations while displaying negligible affinity and activity at 5-HT1A, 5-HT2B, and 5-HT2C receptors. At similar concentrations, XOB administration inhibits sodium current in heterologous cells and results in reduced action potential (AP) firing and VGSC-related AP properties in mouse prefrontal cortex layer V pyramidal neurons. Thus, XOB represents a new, proof-of-principle tool that can be used for future preclinical investigations and therapeutic development. This polypharmacology approach of developing a single molecule to act upon two targets, which are currently independently targeted by combination therapies, may lead to safer alternatives for the treatment of psychiatric disorders that are increasingly being found to benefit from the simultaneous targeting of multiple receptors. SIGNIFICANCE STATEMENT: The authors synthesized a novel small molecule (XOB) that simultaneously antagonizes two key therapeutic targets of bipolar disorder, 5-HT2A receptors and voltage-gated sodium channels, in heterologous cells, and inhibits the intrinsic excitability of mouse prefrontal cortex layer V pyramidal neurons in brain slices. XOB represents a valuable new proof-of-principle tool for future preclinical investigations and provides a novel molecular approach to the pharmacological treatment of complex neuropsychiatric disease, which often requires a combination of therapeutics for sufficient patient benefit.


Subject(s)
Receptor, Serotonin, 5-HT2A , Animals , Mice , Receptor, Serotonin, 5-HT2A/metabolism , Receptor, Serotonin, 5-HT2A/drug effects , Humans , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Voltage-Gated Sodium Channels/metabolism , Voltage-Gated Sodium Channels/drug effects , Male , Mice, Inbred C57BL , HEK293 Cells , Cricetulus
14.
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
15.
J Neurochem ; 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898705

ABSTRACT

3,4-Methylenedioxymethamphetamine (MDMA, 'ecstasy') is re-emerging in clinical settings as a candidate for the treatment of specific neuropsychiatric disorders (e.g. post-traumatic stress disorder) in combination with psychotherapy. MDMA is a psychoactive drug, typically regarded as an empathogen or entactogen, which leads to transporter-mediated monoamine release. Despite its therapeutic potential, MDMA can induce dose-, individual-, and context-dependent untoward effects outside safe settings. In this study, we investigated whether three new methylenedioxy bioisosteres of MDMA improve its off-target profile. In vitro methods included radiotracer assays, transporter electrophysiology, bioluminescence resonance energy transfer and fluorescence-based assays, pooled human liver microsome/S9 fraction incubations, metabolic stability studies, isozyme mapping, and liquid chromatography coupled to high-resolution mass spectrometry. In silico methods included molecular docking. Compared with MDMA, all three MDMA bioisosteres (ODMA, TDMA, and SeDMA) showed similar pharmacological activity at human serotonin, dopamine, and norepinephrine transporters (hSERT, hDAT, and hNET, respectively) but decreased agonist activity at 5-HT2A/2B/2C receptors. Regarding their hepatic metabolism, they differed from MDMA, with N-demethylation being the only metabolic route shared, and without forming phase II metabolites. In addition, TDMA showed an enhanced intrinsic clearance in comparison to its congeners. Additional screening for their interaction with human organic cation transporters (hOCTs) and plasma membrane monoamine transporter (hPMAT) revealed a weaker interaction of the MDMA analogs with hOCT1, hOCT2, and hPMAT. Our findings suggest that these new MDMA bioisosteres might constitute appealing therapeutic alternatives to MDMA, sparing the primary pharmacological activity at hSERT, hDAT, and hNET, but displaying a reduced activity at 5-HT2A/2B/2C receptors and alternative hepatic metabolism. Whether these MDMA bioisosteres may pose lower risk alternatives to the clinically re-emerging MDMA warrants further studies.

16.
Am J Physiol Cell Physiol ; 324(2): C573-C587, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36622066

ABSTRACT

The anthelmintic drug praziquantel (PZQ) causes contraction of parasitic schistosomes as well as constriction of blood vessels within the mesenteric vasculature of the host where the adult blood flukes reside. The contractile action of PZQ on the vasculature is mediated by the activation of host serotonergic 5-HT2B receptors (5-HT2BRs). However, the molecular basis for PZQ interaction with these targets and the location of these 5-HT2B receptors in the vessel wall have not been experimentally defined. Evaluation of a PZQ docking pose within the 5-HT2BR orthosteric site, using both Ca2+ reporter and bioluminescence resonance energy transfer (BRET) assays, identified residues F3406.51 and F3416.52 (transmembrane helix 6, TM6) as well as L209EL2 (extracellular loop 2) as critical for PZQ-mediated agonist activity. A key determinant of PZQ selectivity for the 5-HT2B receptor over the 5-HT2A/2C receptors was determined by M2185.39 in transmembrane helix 5 (TM5) of the orthosteric site. Mutation of this residue to valine (M218V), as found in 5-HT2A and 5-HT2C, decreased PZQ agonist activity, whereas the reciprocal mutation (V215M) in 5-HT2C increased PZQ activity. Two-photon imaging in intact mesenteric arterial strips visualized PZQ-evoked Ca2+ transients within the smooth muscle cells of the vessel wall. PZQ also triggered cytoplasmic Ca2+ signals in arterial smooth muscle cells in primary culture that were isolated from mesenteric blood vessels. These data define the molecular basis for PZQ action on 5-HT2B receptors localized in vascular smooth muscle.


Subject(s)
Anthelmintics , Praziquantel , Praziquantel/pharmacology , Serotonin , Anthelmintics/therapeutic use , Arteries
17.
Nat Chem Biol ; 16(8): 841-849, 2020 08.
Article in English | MEDLINE | ID: mdl-32367019

ABSTRACT

G-protein-coupled receptors (GPCRs) remain major drug targets, despite our incomplete understanding of how they signal through 16 non-visual G-protein signal transducers (collectively named the transducerome) to exert their actions. To address this gap, we have developed an open-source suite of 14 optimized bioluminescence resonance energy transfer (BRET) Gαßγ biosensors (named TRUPATH) to interrogate the transducerome with single pathway resolution in cells. Generated through exhaustive protein engineering and empirical testing, the TRUPATH suite of Gαßγ biosensors includes the first Gα15 and GαGustducin probes. In head-to-head studies, TRUPATH biosensors outperformed first-generation sensors at multiple GPCRs and in different cell lines. Benchmarking studies with TRUPATH biosensors recapitulated previously documented signaling bias and revealed new coupling preferences for prototypic and understudied GPCRs with potential in vivo relevance. To enable a greater understanding of GPCR molecular pharmacology by the scientific community, we have made TRUPATH biosensors easily accessible as a kit through Addgene.


Subject(s)
Biosensing Techniques/instrumentation , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/physiology , Biosensing Techniques/methods , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Protein Engineering/methods , Signal Transduction
18.
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
19.
J Biol Chem ; 295(40): 13927-13939, 2020 10 02.
Article in English | MEDLINE | ID: mdl-32788219

ABSTRACT

The human chemokine family consists of 46 protein ligands that induce chemotactic cell migration by activating a family of 23 G protein-coupled receptors. The two major chemokine subfamilies, CC and CXC, bind distinct receptor subsets. A sequence motif defining these families, the X position in the CXC motif, is not predicted to make significant contacts with the receptor, but instead links structural elements associated with binding and activation. Here, we use comparative analysis of chemokine NMR structures, structural modeling, and molecular dynamic simulations that suggested the X position reorients the chemokine N terminus. Using CXCL12 as a model CXC chemokine, deletion of the X residue (Pro-10) had little to no impact on the folded chemokine structure but diminished CXCR4 agonist activity as measured by ERK phosphorylation, chemotaxis, and Gi/o-mediated cAMP inhibition. Functional impairment was attributed to over 100-fold loss of CXCR4 binding affinity. Binding to the other CXCL12 receptor, ACKR3, was diminished nearly 500-fold. Deletion of Pro-10 had little effect on CXCL12 binding to the CXCR4 N terminus, a major component of the chemokine-GPCR interface. Replacement of the X residue with the most frequent amino acid at this position (P10Q) had an intermediate effect between WT and P10del in each assay, with ACKR3 having a higher tolerance for this mutation. This work shows that the X residue helps to position the CXCL12 N terminus for optimal docking into the orthosteric pocket of CXCR4 and suggests that the CC/CXC motif contributes directly to receptor selectivity by orienting the chemokine N terminus in a subfamily-specific direction.


Subject(s)
Chemokine CXCL12/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Receptors, CXCR4/chemistry , Receptors, CXCR/chemistry , Amino Acid Motifs , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Humans , Receptors, CXCR/genetics , Receptors, CXCR/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Structure-Activity Relationship
20.
Nat Chem Biol ; 14(2): 126-134, 2018 02.
Article in English | MEDLINE | ID: mdl-29227473

ABSTRACT

Development of biased ligands targeting G protein-coupled receptors (GPCRs) is a promising approach for current drug discovery. Although structure-based drug design of biased agonists remains challenging even with an abundance of GPCR crystal structures, we present an approach for translating GPCR structural data into ß-arrestin-biased ligands for aminergic GPCRs. We identified specific amino acid-ligand contacts at transmembrane helix 5 (TM5) and extracellular loop 2 (EL2) responsible for Gi/o and ß-arrestin signaling, respectively, and targeted those residues to develop biased ligands. For these ligands, we found that bias is conserved at other aminergic GPCRs that retain similar residues at TM5 and EL2. Our approach provides a template for generating arrestin-biased ligands by modifying predicted ligand interactions that block TM5 interactions and promote EL2 interactions. This strategy may facilitate the structure-guided design of arrestin-biased ligands at other GPCRs, including polypharmacological biased ligands.


Subject(s)
Ligands , Receptors, G-Protein-Coupled/chemistry , beta-Arrestin 1/chemistry , Aripiprazole/chemistry , Crystallography, X-Ray , Cyclic AMP/chemistry , Drug Design , Drug Discovery , HEK293 Cells , Humans , Hydrogen Bonding , Indoles/chemistry , Kinetics , Molecular Dynamics Simulation , Mutation , Protein Binding , Protein Conformation , Serine/chemistry , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL