Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
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
2.
Cell ; 152(3): 385-6, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23374334

ABSTRACT

Recent advances in G-protein-coupled receptor structural biology have provided only limited insight into the active conformations of these key signaling molecules. A paper from Nygaard et al. reveals the dynamic nature of GPCRs along the activation pathway by complementing NMR experiments with ultralong-timescale molecular dynamics simulations.

3.
J Pharmacol Exp Ther ; 375(2): 357-366, 2020 11.
Article in English | MEDLINE | ID: mdl-32848074

ABSTRACT

Fibrosis or accumulation of extracellular matrix is an evolutionarily conserved mechanism adopted by an organism as a response to chronic injury. Excessive fibrosis, however, leads to disruption of organ homeostasis and is a common feature of many chronic diseases. G protein-coupled receptors (GPCRs) are important cell signaling mediators and represent molecular targets for many Food and Drug Administration-approved drugs. To identify new targets for fibrosis, we used a synthetic GPCR system named designed receptors exclusively activated by designer drugs (DREADDs) to probe signaling pathways essential for fibrotic response. We found that upon expression in human lung fibroblasts, activation of Gq- and Gs-DREADDs abrogated the induction of TGFß-induced fibrosis marker genes. Genome-wide transcriptome analysis identified dysregulation of multiple GPCRs in lung fibroblasts treated with TGFß To investigate endogenous GPCR modulating TGFß signaling, we selected 13 GPCRs that signal through Gq or Gs and activated them by using specific agonists. We examined the impact of each agonist and how activation of endogenous GPCR affects TGFß signaling. Among the agonists examined, prostaglandin receptor agonists demonstrated the strongest inhibitory effect on fibrosis. Together, we have demonstrated that the DREADDs system is a valuable tool to identify beneficial GPCR signaling for fibrosis. This study in fibroblasts has served as a proof of concept and allowed us to further develop in vivo models for fibrosis GPCR discovery. SIGNIFICANCE STATEMENT: Fibrosis is the hallmark of many end-stage cardiometabolic diseases, and there is an unmet medical need to discover new antifibrotic therapies, reduce disease progression, and bring clinically meaningful efficacy to patients. Our work utilizes designed receptors exclusively activated by designer drug chemogenetic tools to identify beneficial GPCR signaling for fibrosis, providing new insights into GPCR drug discovery.


Subject(s)
Designer Drugs/pharmacology , Receptors, G-Protein-Coupled/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Fibrosis , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Gene Expression Regulation/drug effects , Humans , Receptors, Prostaglandin/metabolism , Signal Transduction/drug effects
4.
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
5.
Nature ; 485(7398): 395-9, 2012 May 16.
Article in English | MEDLINE | ID: mdl-22596163

ABSTRACT

Members of the opioid receptor family of G-protein-coupled receptors (GPCRs) are found throughout the peripheral and central nervous system, where they have key roles in nociception and analgesia. Unlike the 'classical' opioid receptors, δ, κ and µ (δ-OR, κ-OR and µ-OR), which were delineated by pharmacological criteria in the 1970s and 1980s, the nociceptin/orphanin FQ (N/OFQ) peptide receptor (NOP, also known as ORL-1) was discovered relatively recently by molecular cloning and characterization of an orphan GPCR. Although it shares high sequence similarity with classical opioid GPCR subtypes (∼60%), NOP has a markedly distinct pharmacology, featuring activation by the endogenous peptide N/OFQ, and unique selectivity for exogenous ligands. Here we report the crystal structure of human NOP, solved in complex with the peptide mimetic antagonist compound-24 (C-24) (ref. 4), revealing atomic details of ligand-receptor recognition and selectivity. Compound-24 mimics the first four amino-terminal residues of the NOP-selective peptide antagonist UFP-101, a close derivative of N/OFQ, and provides important clues to the binding of these peptides. The X-ray structure also shows substantial conformational differences in the pocket regions between NOP and the classical opioid receptors κ (ref. 5) and µ (ref. 6), and these are probably due to a small number of residues that vary between these receptors. The NOP-compound-24 structure explains the divergent selectivity profile of NOP and provides a new structural template for the design of NOP ligands.


Subject(s)
Biomimetic Materials/chemistry , Opioid Peptides/chemistry , Piperidines/chemistry , Piperidines/metabolism , Receptors, Opioid/chemistry , Receptors, Opioid/metabolism , Spiro Compounds/chemistry , Spiro Compounds/metabolism , Binding Sites , Biomimetic Materials/metabolism , Biomimetic Materials/pharmacology , Crystallography, X-Ray , HEK293 Cells , Humans , Ligands , Models, Molecular , Narcotic Antagonists , Opioid Peptides/metabolism , Opioid Peptides/pharmacology , Piperidines/pharmacology , Protein Conformation , Receptors, Opioid, kappa/chemistry , Receptors, Opioid, kappa/metabolism , Spiro Compounds/pharmacology , Substrate Specificity , Nociceptin Receptor
6.
J Neurosci ; 36(11): 3281-94, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26985037

ABSTRACT

In many human alcoholics, abstinence is self-imposed because of the negative consequences of excessive alcohol use, and relapse is often triggered by exposure to environmental contexts associated with prior alcohol drinking. We recently developed a rat model of this human condition in which we train alcohol-preferring P rats to self-administer alcohol in one context (A), punish the alcohol-reinforced responding in a different context (B), and then test for relapse to alcohol seeking in Contexts A and B without alcohol or shock. Here, we studied the role of projections to nucleus accumbens (NAc) shell from ventral subiculum (vSub), basolateral amygdala, paraventricular thalamus, and ventral medial prefrontal cortex in context-induced relapse after punishment-imposed abstinence. First, we measured double-labeling of the neuronal activity marker Fos with the retrograde tracer cholera toxin subunit B (injected in NAc shell) and demonstrated that context-induced relapse is associated with selective activation of the vSub→NAc shell projection. Next, we reversibly inactivated the vSub with GABA receptor agonists (muscimol+baclofen) before the context-induced relapse tests and provided evidence for a causal role of vSub in this relapse. Finally, we used a dual-virus approach to restrict expression of the inhibitory κ opioid-receptor based DREADD (KORD) in vSub→NAc shell projection neurons. We found that systemic injections of the KORD agonist salvinorin B, which selectively inhibits KORD-expressing neurons, decreased context-induced relapse to alcohol seeking. Our results demonstrate a critical role of vSub in context-induced relapse after punishment-imposed abstinence and further suggest a role of the vSub→NAc projection in this relapse. SIGNIFICANCE STATEMENT: In many human alcoholics, abstinence is self-imposed because of the negative consequences of excessive use, and relapse is often triggered by exposure to environmental contexts associated with prior alcohol use. Until recently, an animal model of this human condition did not exist. We developed a rat model of this human condition in which we train alcohol-preferring P rats to self-administer alcohol in one context (A), punish the alcohol-reinforced responding in a different context (B), and test for relapse to alcohol seeking in Contexts A and B. Here, we used neuroanatomical, neuropharmacological, and chemogenetic methods to demonstrate a role of ventral subiculum and potentially its projections to nucleus accumbens in context-induced relapse after punishment-imposed abstinence.


Subject(s)
Alcohol Abstinence/psychology , Alcohol Drinking/psychology , Conditioning, Operant/physiology , Extinction, Psychological/physiology , Nucleus Accumbens/physiopathology , Punishment , Alcohol Drinking/physiopathology , Animals , Cholera Toxin/metabolism , Conditioning, Operant/drug effects , Diterpenes/pharmacology , Diterpenes, Clerodane , Ethanol/administration & dosage , Extinction, Psychological/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Neurons/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Nucleus Accumbens/pathology , Oncogene Proteins v-fos/genetics , Oncogene Proteins v-fos/metabolism , Rats , Receptors, Opioid, kappa/metabolism , Recurrence , Reinforcement, Psychology , Self Administration , Transduction, Genetic
7.
J Biol Chem ; 288(48): 34470-83, 2013 Nov 29.
Article in English | MEDLINE | ID: mdl-24121503

ABSTRACT

The crystal structures of opioid receptors provide a novel platform for inquiry into opioid receptor function. The molecular determinants for activation of the κ-opioid receptor (KOR) were studied using a combination of agonist docking, functional assays, and site-directed mutagenesis. Eighteen positions in the putative agonist binding site of KOR were selected and evaluated for their effects on receptor binding and activation by ligands representing four distinct chemotypes: the peptide dynorphin A(1-17), the arylacetamide U-69593, and the non-charged ligands salvinorin A and the octahydroisoquinolinone carboxamide 1xx. Minimally biased docking of the tested ligands into the antagonist-bound KOR structure generated distinct binding modes, which were then evaluated biochemically and pharmacologically. Our analysis identified two types of mutations: those that affect receptor function primarily via ligand binding and those that primarily affect function. The shared and differential mechanisms of agonist binding and activation in KOR are further discussed. Usually, mutations affecting function more than binding were located at the periphery of the binding site and did not interact strongly with the various ligands. Analysis of the crystal structure along with the present results provide fundamental insights into the activation mechanism of the KOR and suggest that "functional" residues, along with water molecules detected in the crystal structure, may be directly involved in transduction of the agonist binding event into structural changes at the conserved rotamer switches, thus leading to receptor activation.


Subject(s)
Analgesics, Opioid/chemistry , Binding Sites/genetics , Receptors, Opioid, kappa/chemistry , Receptors, Opioid, kappa/genetics , Structure-Activity Relationship , Analgesics, Opioid/metabolism , Crystallography, X-Ray , Dynorphins/chemistry , Dynorphins/metabolism , HEK293 Cells , Humans , Ligands , Molecular Docking Simulation , Mutagenesis, Site-Directed , Mutation/genetics , Protein Conformation , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/metabolism
8.
Bioorg Med Chem Lett ; 23(10): 2860-2, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23587424

ABSTRACT

Salvinorin A, the active ingredient of the hallucinogenic plant Salvia divinorum is the most potent known naturally occurring hallucinogen and is a selective κ-opioid receptor agonist. To better understand the ligand-receptor interactions, a series of dicarboxylic ester-type of salvinorin A derivatives were synthesized and evaluated for their binding affinity at κ-, δ- and µ-opioid receptors. Most of the analogues show high affinity to the κ-opioid receptor. Methyl malonyl derivative 4 shows the highest binding affinity (Ki=2nM), analogues 5, 7, and 14 exhibit significant affinity for the κ-receptor (Ki=21, 36 and 39nM).


Subject(s)
Diterpenes, Clerodane/pharmacology , Drugs, Chinese Herbal/pharmacology , Receptors, Opioid, kappa/agonists , Camphanes , Diterpenes, Clerodane/chemistry , Diterpenes, Clerodane/isolation & purification , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/isolation & purification , HEK293 Cells , Humans , Molecular Conformation , Panax notoginseng , Salvia/chemistry , Salvia miltiorrhiza , Structure-Activity Relationship
9.
Proc Natl Acad Sci U S A ; 107(11): 4908-13, 2010 Mar 16.
Article in English | MEDLINE | ID: mdl-20194783

ABSTRACT

The Gibbs free energy difference between native and unfolded states ("stability") is one of the fundamental characteristics of a protein. By exploiting the thermodynamic linkage between ligand binding and stability, interactions of a protein with small molecules, nucleic acids, or other proteins can be detected and quantified. Determination of protein stability can therefore provide a universal monitor of biochemical function. Yet, the use of stability measurements as a functional probe is underutilized, because such experiments traditionally require large amounts of protein and special instrumentation. Here we present the quantitative cysteine reactivity (QCR) technique to determine protein stabilities rapidly and accurately using only picomole quantities of material and readily accessible laboratory equipment. We demonstrate that QCR-derived stabilities can be used to measure ligand binding over a wide range of ligand concentrations and affinities. We anticipate that this technique will have broad applications in high-throughput protein engineering experiments and functional genomics.


Subject(s)
Cysteine/metabolism , Nanotechnology/methods , Protein Stability , Proteins/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Ligands , Micrococcal Nuclease/chemistry , Micrococcal Nuclease/metabolism , Protein Conformation , Protein Folding , Proteins/metabolism , Temperature
10.
Neuropsychopharmacology ; 41(2): 402-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26019014

ABSTRACT

In the past decade, novel methods using engineered receptors have enabled researchers to manipulate neuronal activity with increased spatial and temporal specificity. One widely used chemogenetic method in mice and rats is the DREADD (designer receptors exclusively activated by designer drugs) system in which a mutated muscarinic G protein-coupled receptor is activated by an otherwise inert synthetic ligand, clozapine-N-oxide (CNO). Recently, the Roth laboratory developed a novel inhibitory DREADD in which a mutated kappa-opioid receptor (KORD) is activated by the pharmacologically inert drug salvinorin B (SalB; Vardy et al, 2015). They demonstrated the feasibility of using KORD to study brain circuits involved in motivated behavior in mice. Here, we used behavioral, electrophysiological, and neuroanatomical methods to demonstrate the feasibility of using the novel KORD to study brain circuits involved in motivated behavior in rats. In Exp. 1, we show that SalB dose-dependently decreased spontaneous and cocaine-induced locomotor activity in rats expressing KORD to midbrain (ventral tegmental area/substantia nigra). In Exp. 2, we show that SalB completely inhibited tonic firing in KORD-expressing putative dopamine neurons in midbrain. In Exp. 3, we used a 'retro-DREADD' dual-virus approach to restrict expression of KORD in ventral subiculum neurons that project to nucleus accumbens shell. We show that KORD activation selectively decreased novel context-induced Fos expression in this projection. Our results indicate that the novel KORD is a promising tool to selectively inactivate brain areas and neural circuits in rat studies of motivated behavior.


Subject(s)
Gene Transfer Techniques , Mesencephalon/physiology , Receptors, Opioid, kappa/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Central Nervous System Agents/pharmacology , Cocaine/pharmacology , Dependovirus/genetics , Diterpenes/pharmacology , Diterpenes, Clerodane , Dopamine Uptake Inhibitors/pharmacology , Feasibility Studies , Genetic Engineering , Genetic Vectors , Male , Mesencephalon/cytology , Mesencephalon/drug effects , Motor Activity/drug effects , Motor Activity/physiology , Mutation , Neurons/cytology , Neurons/drug effects , Neurons/physiology , Rats, Sprague-Dawley , Receptors, Opioid, kappa/genetics
11.
Chem Biol ; 22(6): 764-75, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26091169

ABSTRACT

It has been suggested that the evolution of vertebrate opioid receptors (ORs) follow a vector of increased functionality. Here, we test this idea by comparing human and frog ORs. Interestingly, some of the most potent opioid peptides known have been isolated from amphibian skin secretions. Here we show that such peptides (dermorphin and deltorphin) are highly potent in the human receptors and inactive in frog ORs. The molecular basis for the insensitivity of the frog ORs to these peptides was studied using chimeras and molecular modeling. The insensitivity of the delta OR (DOR) to deltorphin was due to variation of a single amino acid, Trp7.35, which is a leucine in mammalian DORs. Notably, Trp7.35 is completely conserved in all known DOR sequences from lamprey, fish, and amphibians. The deltorphin-insensitive phenotype was verified in fish. Our results provide a molecular explanation for the species selectivity of skin-derived opioid peptides.


Subject(s)
Amphibians/metabolism , Analgesics, Opioid/metabolism , Peptides/metabolism , Skin/metabolism , Amino Acid Sequence , Analgesics, Opioid/chemistry , Animals , Behavior, Animal/drug effects , Binding Sites , Humans , Kinetics , Molecular Dynamics Simulation , Molecular Sequence Data , Oligopeptides/chemistry , Oligopeptides/metabolism , Opioid Peptides/chemistry , Opioid Peptides/metabolism , Peptides/chemistry , Peptides/pharmacology , Protein Structure, Tertiary , Receptors, Opioid/chemistry , Receptors, Opioid/metabolism , Sequence Alignment , Species Specificity , Zebrafish/physiology
12.
Neuropsychopharmacology ; 40(11): 2614-22, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25881115

ABSTRACT

The OPRM1 A118G polymorphism is the most widely studied µ-opioid receptor (MOR) variant. Although its involvement in acute alcohol effects is well characterized, less is known about the extent to which it alters responses to opioids. Prior work has shown that both electrophysiological and analgesic responses to morphine but not to fentanyl are moderated by OPRM1 A118G variation, but the mechanism behind this dissociation is not known. Here we found that humanized mice carrying the 118GG allele (h/mOPRM1-118GG) were less sensitive than h/mOPRM1-118AA littermates to the rewarding effects of morphine and hydrocodone but not those of other opioids measured with intracranial self-stimulation. Reduced morphine reward in 118GG mice was associated with decreased dopamine release in the nucleus accumbens and reduced effects on GABA release in the ventral tegmental area that were not due to changes in drug potency or efficacy in vitro or receptor-binding affinity. Fewer MOR-binding sites were observed in h/mOPRM1-118GG mice, and pharmacological reduction of MOR availability unmasked genotypic differences in fentanyl sensitivity. These findings suggest that the OPRM1 A118G polymorphism decreases sensitivity to low-potency agonists by decreasing receptor reserve without significantly altering receptor function.


Subject(s)
Analgesics, Opioid/pharmacology , Nucleus Accumbens/metabolism , Receptors, Opioid, mu/metabolism , Reward , Ventral Tegmental Area/metabolism , Animals , Disease Models, Animal , Dopamine/metabolism , HEK293 Cells , Humans , Male , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/physiology , Narcotic Antagonists/pharmacology , Nucleus Accumbens/drug effects , Polymorphism, Single Nucleotide , Receptors, Opioid, mu/genetics , Self Stimulation , Tissue Culture Techniques , Ventral Tegmental Area/drug effects , gamma-Aminobutyric Acid/metabolism
13.
Neuron ; 86(4): 936-946, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25937170

ABSTRACT

DREADDs are chemogenetic tools widely used to remotely control cellular signaling, neuronal activity, and behavior. Here we used a structure-based approach to develop a new Gi-coupled DREADD using the kappa-opioid receptor as a template (KORD) that is activated by the pharmacologically inert ligand salvinorin B (SALB). Activation of virally expressed KORD in several neuronal contexts robustly attenuated neuronal activity and modified behaviors. Additionally, co-expression of the KORD and the Gq-coupled M3-DREADD within the same neuronal population facilitated the sequential and bidirectional remote control of behavior. The availability of DREADDs activated by different ligands provides enhanced opportunities for investigating diverse physiological systems using multiplexed chemogenetic actuators.


Subject(s)
Behavior, Animal/drug effects , Diterpenes/pharmacology , Neurons/drug effects , Receptors, Opioid, kappa/metabolism , Signal Transduction/drug effects , Animals , Diterpenes, Clerodane , Ligands , Mice, Inbred C57BL
14.
Protein Sci ; 13(7): 1832-40, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15215526

ABSTRACT

The structures of membrane transporters are still mostly unsolved. Only recently, the first two high-resolution structures of transporters of the major facilitator superfamily (MFS) were published. Despite the low sequence similarity of the two proteins involved, lactose permease and glycerol-3-phosphate transporter, the reported structures are highly similar. This leads to the hypothesis that all members of the MFS share a similar structure, regardless of their low sequence identity. To test this hypothesis, we generated models of two other members of the MFS, the Tn10-encoded metal-tetracycline/H(+) antiporter (TetAB) and the rat vesicular monoamine transporter (rVMAT2). The models are based on the two MFS structures and on experimental data. The models for both proteins are in good agreement with the data available and support the notion of a shared fold for all MFS proteins.


Subject(s)
Antiporters/chemistry , Bacterial Proteins/chemistry , Computer Simulation , Membrane Glycoproteins/chemistry , Membrane Transport Proteins/chemistry , Structural Homology, Protein , Animals , Rats , Vesicular Biogenic Amine Transport Proteins , Vesicular Monoamine Transport Proteins
15.
Eur J Med Chem ; 85: 818-29, 2014 Oct 06.
Article in English | MEDLINE | ID: mdl-25193297

ABSTRACT

The neoclerodane diterpenoid salvinorin A is a major secondary metabolite isolated from the psychoactive plant Salvia divinorum. Salvinorin A has been shown to have high affinity and selectivity for the κ-opioid receptor (KOR). To study the ligand-receptor interactions that occur between salvinorin A and the KOR, a new series of salvinorin A derivatives bearing potentially reactive Michael acceptor functional groups at C-2 was synthesized and used to probe the salvinorin A binding site. The κ-, δ-, and µ-opioid receptor (KOR, DOR and MOR, respectively) binding affinities and KOR efficacies were measured for the new compounds. Although none showed wash-resistant irreversible binding, most of them showed high affinity for the KOR, and some exhibited dual affinity to KOR and MOR. Molecular modeling techniques based on the recently-determined crystal structure of the KOR combined with results from mutagenesis studies, competitive binding, functional assays and structure-activity relationships, and previous salvinorin A-KOR interaction models were used to identify putative interaction modes of the new compounds with the KOR and MOR.


Subject(s)
Diterpenes, Clerodane/chemistry , Diterpenes, Clerodane/metabolism , Drug Design , Receptors, Opioid, kappa/metabolism , Diterpenes, Clerodane/chemical synthesis , HEK293 Cells , Humans , Ligands , Models, Molecular , Protein Binding , Protein Conformation , Receptors, Opioid, kappa/chemistry
16.
Nat Commun ; 5: 4355, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-25008467

ABSTRACT

The Smoothened receptor (SMO) mediates signal transduction in the hedgehog pathway, which is implicated in normal development and carcinogenesis. SMO antagonists can suppress the growth of some tumours; however, mutations at SMO have been found to abolish their antitumour effects, a phenomenon known as chemoresistance. Here we report three crystal structures of human SMO bound to the antagonists SANT1 and Anta XV, and the agonist, SAG1.5, at 2.6-2.8 Å resolution. The long and narrow cavity in the transmembrane domain of SMO harbours multiple ligand binding sites, where SANT1 binds at a deeper site as compared with other ligands. Distinct interactions at D473(6.54f) elucidated the structural basis for the differential effects of chemoresistance mutations on SMO antagonists. The agonist SAG1.5 induces a conformational rearrangement of the binding pocket residues, which could contribute to SMO activation. Collectively, these studies reveal the structural basis for the modulation of SMO by small molecules.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/physiology , Models, Molecular , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/drug effects , Crystallography , Cyclohexylamines/pharmacology , Hedgehog Proteins/physiology , Humans , Lysine/analogs & derivatives , Lysine/pharmacology , Phthalazines/pharmacology , Piperazines/pharmacology , Pyrazoles/pharmacology , Receptors, G-Protein-Coupled/physiology , Signal Transduction/physiology , Smoothened Receptor , Thiophenes/pharmacology
17.
J Med Chem ; 56(21): 8826-33, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24144404

ABSTRACT

In previous studies we reported that addition of 7α-acylamino groups to N-phenylpropyl-4ß-methyl-5-(3-hydroxyphenyl)morphan (4) led to compounds that were pure opioid receptor antagonists. In contrast to these findings we report in this study that addition of a 7α-amino (5a), 7α-alkylamino (5b-e), or 7α-dialkylamino (5f-h) group to 4 leads to opioid receptor ligands with varying degrees of agonist/antagonist activity. The 7α-amino and 7α-methylamino analogues were full agonists at the µ and δ receptors and antagonists at the κ receptor. The 7α-cyclopropylmethylamino analogue 5h was a full agonist at the µ receptor with weaker agonist activity at the δ and κ receptors. Whereas the addition of a 7α-acylamino group to the pure nonselective opioid receptor antagonist N-phenylpropyl-4ß-methyl-5-(3-hydroxyphenyl)morphan (4) led to κ selective pure opioid receptor antagonist, the addition of a 7α-amino, 7α-alkylamino, or 7α-dialkylamino group to 4 leads to opioid ligands that are largely µ or δ agonist with mixed agonist/antagonist properties.


Subject(s)
Azabicyclo Compounds/pharmacology , Narcotic Antagonists , Receptors, Opioid/agonists , Azabicyclo Compounds/chemical synthesis , Azabicyclo Compounds/chemistry , Dose-Response Relationship, Drug , Humans , Molecular Structure , Structure-Activity Relationship
18.
Science ; 340(6132): 615-9, 2013 May 03.
Article in English | MEDLINE | ID: mdl-23519215

ABSTRACT

Drugs active at G protein-coupled receptors (GPCRs) can differentially modulate either canonical or noncanonical signaling pathways via a phenomenon known as functional selectivity or biased signaling. We report biochemical studies showing that the hallucinogen lysergic acid diethylamide, its precursor ergotamine (ERG), and related ergolines display strong functional selectivity for ß-arrestin signaling at the 5-HT2B 5-hydroxytryptamine (5-HT) receptor, whereas they are relatively unbiased at the 5-HT1B receptor. To investigate the structural basis for biased signaling, we determined the crystal structure of the human 5-HT2B receptor bound to ERG and compared it with the 5-HT1B/ERG structure. Given the relatively poor understanding of GPCR structure and function to date, insight into different GPCR signaling pathways is important to better understand both adverse and favorable therapeutic activities.


Subject(s)
Ergotamine/metabolism , Receptor, Serotonin, 5-HT1B/metabolism , Receptor, Serotonin, 5-HT2B/chemistry , Receptor, Serotonin, 5-HT2B/metabolism , Amino Acid Motifs , Amino Acid Sequence , Arrestin/metabolism , Arrestins/metabolism , Binding Sites , Crystallography, X-Ray , Ergolines/chemistry , Ergolines/metabolism , Ergotamine/chemistry , HEK293 Cells , Humans , Ligands , Lysergic Acid Diethylamide/chemistry , Lysergic Acid Diethylamide/metabolism , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Structure, Secondary , Receptor, Serotonin, 5-HT1B/chemistry , Receptors, Serotonin/chemistry , Receptors, Serotonin/metabolism , Signal Transduction , beta-Arrestins
19.
Science ; 340(6132): 610-4, 2013 May 03.
Article in English | MEDLINE | ID: mdl-23519210

ABSTRACT

Serotonin or 5-hydroxytryptamine (5-HT) regulates a wide spectrum of human physiology through the 5-HT receptor family. We report the crystal structures of the human 5-HT1B G protein-coupled receptor bound to the agonist antimigraine medications ergotamine and dihydroergotamine. The structures reveal similar binding modes for these ligands, which occupy the orthosteric pocket and an extended binding pocket close to the extracellular loops. The orthosteric pocket is formed by residues conserved in the 5-HT receptor family, clarifying the family-wide agonist activity of 5-HT. Compared with the structure of the 5-HT2B receptor, the 5-HT1B receptor displays a 3 angstrom outward shift at the extracellular end of helix V, resulting in a more open extended pocket that explains subtype selectivity. Together with docking and mutagenesis studies, these structures provide a comprehensive structural basis for understanding receptor-ligand interactions and designing subtype-selective serotonergic drugs.


Subject(s)
Dihydroergotamine/metabolism , Ergotamine/metabolism , Receptor, Serotonin, 5-HT1B/chemistry , Receptor, Serotonin, 5-HT1B/metabolism , Serotonin 5-HT1 Receptor Agonists/chemistry , Serotonin 5-HT1 Receptor Agonists/metabolism , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Dihydroergotamine/chemistry , Ergotamine/chemistry , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Ligands , Lysergic Acid Diethylamide/chemistry , Lysergic Acid Diethylamide/metabolism , Models, Molecular , Molecular Docking Simulation , Molecular Sequence Data , Mutagenesis , Norfenfluramine/chemistry , Norfenfluramine/metabolism , Pindolol/analogs & derivatives , Pindolol/chemistry , Pindolol/metabolism , Propranolol/chemistry , Propranolol/metabolism , Protein Conformation , Protein Folding , Protein Structure, Secondary , Receptor, Serotonin, 5-HT1B/genetics , Tryptamines/chemistry , Tryptamines/metabolism
20.
Protein Eng Des Sel ; 22(10): 641-8, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19643977

ABSTRACT

Predicting mutations that enhance protein-protein affinity remains a challenging task, especially for high-affinity complexes. To test our capability to improve the affinity of such complexes, we studied interaction of acetylcholinesterase with the snake toxin, fasciculin. Using the program ORBIT, we redesigned fasciculin's sequence to enhance its interactions with Torpedo californica acetylcholinesterase. Mutations were predicted in 5 out of 13 interfacial residues on fasciculin, preserving most of the polar inter-molecular contacts seen in the wild-type toxin/enzyme complex. To experimentally characterize fasciculin mutants, we developed an efficient strategy to over-express the toxin in Escherichia coli, followed by refolding to the native conformation. Despite our predictions, a designed quintuple fasciculin mutant displayed reduced affinity for the enzyme. However, removal of a single mutation in the designed sequence produced a quadruple mutant with improved affinity. Moreover, one designed mutation produced 7-fold enhancement in affinity for acetylcholinesterase. This led us to reassess our criteria for enhancing affinity of the toxin for the enzyme. We observed that the change in the predicted inter-molecular energy, rather than in the total energy, correlates well with the change in the experimental free energy of binding, and hence may serve as a criterion for enhancement of affinity in protein-protein complexes.


Subject(s)
Acetylcholinesterase/metabolism , Elapid Venoms/metabolism , Protein Binding/genetics , Protein Engineering/methods , Recombinant Proteins/metabolism , Acetylcholinesterase/chemistry , Amino Acid Sequence , Elapid Venoms/chemistry , Elapid Venoms/genetics , Escherichia coli/genetics , Kinetics , Models, Molecular , Molecular Sequence Data , Mutation , Protein Interaction Domains and Motifs/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL