Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
1.
Cell ; 184(7): 1659-1660, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33798438

ABSTRACT

Partial agonism describes the relative efficacy of a drug compared to one that produces a greater response in a particular system; the designation is dependent upon the comparator and the system. In this issue of Cell, Huang et al. describe biophysical approaches to define the signature of GPCR partial agonists, providing direct measures of varying intrinsic efficacy.


Subject(s)
Caffeine
2.
Cell ; 180(4): 655-665.e18, 2020 02 20.
Article in English | MEDLINE | ID: mdl-32004463

ABSTRACT

Human endocannabinoid systems modulate multiple physiological processes mainly through the activation of cannabinoid receptors CB1 and CB2. Their high sequence similarity, low agonist selectivity, and lack of activation and G protein-coupling knowledge have hindered the development of therapeutic applications. Importantly, missing structural information has significantly held back the development of promising CB2-selective agonist drugs for treating inflammatory and neuropathic pain without the psychoactivity of CB1. Here, we report the cryoelectron microscopy structures of synthetic cannabinoid-bound CB2 and CB1 in complex with Gi, as well as agonist-bound CB2 crystal structure. Of important scientific and therapeutic benefit, our results reveal a diverse activation and signaling mechanism, the structural basis of CB2-selective agonists design, and the unexpected interaction of cholesterol with CB1, suggestive of its endogenous allosteric modulating role.


Subject(s)
Cannabinoid Receptor Agonists/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , Receptor, Cannabinoid, CB1/chemistry , Receptor, Cannabinoid, CB2/chemistry , Signal Transduction , Allosteric Regulation , Allosteric Site , Animals , CHO Cells , Cannabinoid Receptor Agonists/chemistry , Cannabinoids/chemistry , Cannabinoids/pharmacology , Cell Line, Tumor , Cholesterol/chemistry , Cholesterol/pharmacology , Cricetinae , Cricetulus , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Molecular Dynamics Simulation , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Sf9 Cells , Spodoptera
3.
Cell ; 176(3): 459-467.e13, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30639103

ABSTRACT

The cannabinoid receptor CB2 is predominately expressed in the immune system, and selective modulation of CB2 without the psychoactivity of CB1 has therapeutic potential in inflammatory, fibrotic, and neurodegenerative diseases. Here, we report the crystal structure of human CB2 in complex with a rationally designed antagonist, AM10257, at 2.8 Å resolution. The CB2-AM10257 structure reveals a distinctly different binding pose compared with CB1. However, the extracellular portion of the antagonist-bound CB2 shares a high degree of conformational similarity with the agonist-bound CB1, which led to the discovery of AM10257's unexpected opposing functional profile of CB2 antagonism versus CB1 agonism. Further structural analysis using mutagenesis studies and molecular docking revealed the molecular basis of their function and selectivity for CB2 and CB1. Additional analyses of our designed antagonist and agonist pairs provide important insight into the activation mechanism of CB2. The present findings should facilitate rational drug design toward precise modulation of the endocannabinoid system.


Subject(s)
Receptor, Cannabinoid, CB2/metabolism , Receptor, Cannabinoid, CB2/ultrastructure , Animals , Cannabinoid Receptor Antagonists/pharmacology , Cannabinoids/pharmacology , Drug Design , Endocannabinoids , Humans , Ligands , Molecular Docking Simulation , Protein Binding , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB2/chemistry , Receptors, Cannabinoid/chemistry , Receptors, Cannabinoid/metabolism , Receptors, Cannabinoid/ultrastructure , Receptors, G-Protein-Coupled/metabolism , Sf9 Cells , Structure-Activity Relationship
4.
Cell ; 171(5): 1165-1175.e13, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29149605

ABSTRACT

Biased agonism has been proposed as a means to separate desirable and adverse drug responses downstream of G protein-coupled receptor (GPCR) targets. Herein, we describe structural features of a series of mu-opioid-receptor (MOR)-selective agonists that preferentially activate receptors to couple to G proteins or to recruit ßarrestin proteins. By comparing relative bias for MOR-mediated signaling in each pathway, we demonstrate a strong correlation between the respiratory suppression/antinociception therapeutic window in a series of compounds spanning a wide range of signaling bias. We find that ßarrestin-biased compounds, such as fentanyl, are more likely to induce respiratory suppression at weak analgesic doses, while G protein signaling bias broadens the therapeutic window, allowing for antinociception in the absence of respiratory suppression.


Subject(s)
Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Receptors, Opioid, mu/agonists , Animals , Fentanyl/administration & dosage , GTP-Binding Proteins/metabolism , Mice , Morphine/administration & dosage , Receptors, Opioid, mu/chemistry , Respiratory System/drug effects , Signal Transduction , beta-Arrestins/metabolism
5.
Cell ; 167(3): 750-762.e14, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27768894

ABSTRACT

Cannabinoid receptor 1 (CB1) is the principal target of Δ9-tetrahydrocannabinol (THC), a psychoactive chemical from Cannabis sativa with a wide range of therapeutic applications and a long history of recreational use. CB1 is activated by endocannabinoids and is a promising therapeutic target for pain management, inflammation, obesity, and substance abuse disorders. Here, we present the 2.8 Å crystal structure of human CB1 in complex with AM6538, a stabilizing antagonist, synthesized and characterized for this structural study. The structure of the CB1-AM6538 complex reveals key features of the receptor and critical interactions for antagonist binding. In combination with functional studies and molecular modeling, the structure provides insight into the binding mode of naturally occurring CB1 ligands, such as THC, and synthetic cannabinoids. This enhances our understanding of the molecular basis for the physiological functions of CB1 and provides new opportunities for the design of next-generation CB1-targeting pharmaceuticals.


Subject(s)
Cannabinoid Receptor Antagonists/chemistry , Morpholines/chemistry , Pyrazoles/chemistry , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/chemistry , Binding Sites , Cannabinoids/pharmacology , Cannabis/chemistry , Crystallography, X-Ray , Dronabinol/pharmacology , Endocannabinoids/pharmacology , Humans , Ligands , Morpholines/chemical synthesis , Protein Binding , Protein Conformation, alpha-Helical , Pyrazoles/chemical synthesis
6.
Proc Natl Acad Sci U S A ; 118(48)2021 11 30.
Article in English | MEDLINE | ID: mdl-34819362

ABSTRACT

The ability of a ligand to preferentially promote engagement of one signaling pathway over another downstream of GPCR activation has been referred to as signaling bias, functional selectivity, and biased agonism. The presentation of ligand bias reflects selectivity between active states of the receptor, which may result in the display of preferential engagement with one signaling pathway over another. In this study, we provide evidence that the G protein-biased mu opioid receptor (MOR) agonists SR-17018 and SR-14968 stabilize the MOR in a wash-resistant yet antagonist-reversible G protein-signaling state. Furthermore, we demonstrate that these structurally related biased agonists are noncompetitive for radiolabeled MOR antagonist binding, and while they stimulate G protein signaling in mouse brains, partial agonists of this class do not compete with full agonist activation. Importantly, opioid antagonists can readily reverse their effects in vivo. Given that chronic treatment with SR-17018 does not lead to tolerance in several mouse pain models, this feature may be desirable for the development of long-lasting opioid analgesics that remain sensitive to antagonist reversal of respiratory suppression.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Receptors, Opioid, mu/metabolism , Signal Transduction/drug effects , Analgesics, Opioid/pharmacology , Animals , Benzimidazoles/pharmacology , GTP-Binding Proteins/metabolism , Ligands , Male , Mice , Mice, Inbred C57BL , Narcotic Antagonists/pharmacology , Piperidines/pharmacology , Receptors, G-Protein-Coupled/physiology , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/physiology , Signal Transduction/physiology , beta-Arrestin 2/metabolism
7.
Biochem Soc Trans ; 51(4): 1533-1543, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37646476

ABSTRACT

The cannabinoid receptors CB1 and CB2 mediate a variety of physiological processes and continue to be explored as desirable drug targets. Both receptors are activated by the endogenous endocannabinoids and the psychoactive components of marijuana. Over the years, many efforts have been made to make selective ligands; however, the high degree of homology between cannabinoid receptor subtypes introduces challenges in studying either receptor in isolation. Recent advancements in structure biology have resulted in a surge of high-resolution structures, enriching our knowledge and understanding of receptor structure and function. In this review, of recent cannabinoid receptor structures, key features of the inactive and active state CB1 and CB2 are presented. These structures will provide additional insight into the modulation and signaling mechanism of cannabinoid receptors CB1 and CB2 and aid in the development of future therapeutics.


Subject(s)
Receptors, G-Protein-Coupled , Signal Transduction , Drug Delivery Systems , Endocannabinoids , Receptors, Cannabinoid
8.
Nature ; 547(7664): 468-471, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28678776

ABSTRACT

The cannabinoid receptor 1 (CB1) is the principal target of the psychoactive constituent of marijuana, the partial agonist Δ9-tetrahydrocannabinol (Δ9-THC). Here we report two agonist-bound crystal structures of human CB1 in complex with a tetrahydrocannabinol (AM11542) and a hexahydrocannabinol (AM841) at 2.80 Å and 2.95 Å resolution, respectively. The two CB1-agonist complexes reveal important conformational changes in the overall structure, relative to the antagonist-bound state, including a 53% reduction in the volume of the ligand-binding pocket and an increase in the surface area of the G-protein-binding region. In addition, a 'twin toggle switch' of Phe2003.36 and Trp3566.48 (superscripts denote Ballesteros-Weinstein numbering) is experimentally observed and appears to be essential for receptor activation. The structures reveal important insights into the activation mechanism of CB1 and provide a molecular basis for predicting the binding modes of Δ9-THC, and endogenous and synthetic cannabinoids. The plasticity of the binding pocket of CB1 seems to be a common feature among certain class A G-protein-coupled receptors. These findings should inspire the design of chemically diverse ligands with distinct pharmacological properties.


Subject(s)
Cannabinoid Receptor Agonists/chemistry , Dronabinol/analogs & derivatives , Droperidol/analogs & derivatives , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/chemistry , Binding Sites , Cannabinoid Receptor Agonists/chemical synthesis , Cannabinoid Receptor Agonists/pharmacology , Crystallography, X-Ray , Dronabinol/chemical synthesis , Dronabinol/chemistry , Dronabinol/pharmacology , Droperidol/chemical synthesis , Droperidol/chemistry , Droperidol/pharmacology , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Ligands , Molecular Docking Simulation , Protein Binding , Protein Conformation , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism
9.
Biochemistry ; 61(18): 1923-1935, 2022 09 20.
Article in English | MEDLINE | ID: mdl-34468132

ABSTRACT

In a recent report in Science Signaling (Gillis, A., et al. Low intrinsic efficacy for G protein activation can explain the improved side effect profiles of new opioid agonists. Sci. Signaling 2020, 13, eaaz3140 10.1126/scisignal.aaz3140), it was suggested that low intrinsic agonism, and not biased agonism, leads to an improvement in the separation of potency in opioid-induced respiratory suppression versus antinociception. Although many of the compounds that were tested have been shown to display G protein signaling bias in prior publications, the authors conclude that because they cannot detect biased agonism in their cellular signaling studies the compounds are therefore not biased agonists. Rather, they conclude that it is low intrinsic efficacy that leads to the therapeutic window improvement. Intrinsic efficacy is the extent to which an agonist can stimulate a G protein-coupled receptor response in a system, while biased agonism takes into consideration not only the intrinsic efficacy but also the potency of an agonist in an assay. Herein, we have reanalyzed the data presented in the published work (10.1126/scisignal.aaz3140) [including the recent Erratum (10.1126/scisignal.abf9803)] to derive intrinsic efficacy and bias factors as ΔΔlog(τ/KA) and ΔΔlog(Emax/EC50), respectively. On the basis of this reanalysis, the data support the conclusion that biased agonism, favoring G protein signaling, was observed. Moreover, a conservation of rank order intrinsic efficacy was not observed upon comparing responses in each assay, further suggesting that multiple active receptor states were present. These observations agree with prior studies in which oliceridine, PZM21, and SR-17018 were first described as biased agonists with improvement in antinociception over respiratory suppression in mice. Therefore, the data in the Science Signaling paper provide strong corroborating evidence that G protein signaling bias may be a means of improving opioid analgesia while avoiding certain undesirable side effects.


Subject(s)
Analgesics, Opioid , Receptors, G-Protein-Coupled , Analgesics, Opioid/adverse effects , Animals , Benzimidazoles , GTP-Binding Proteins/metabolism , Mice , Piperidines , Receptors, G-Protein-Coupled/metabolism , Receptors, Opioid, mu/agonists , Signal Transduction
10.
J Am Chem Soc ; 143(40): 16320-16325, 2021 10 13.
Article in English | MEDLINE | ID: mdl-34596399

ABSTRACT

Due to the lack of genetically encoded probes for fluorine-19 nuclear magnetic resonance spectroscopy (19F NMR), its utility for probing eukaryotic membrane protein dynamics is limited. Here we report an efficient method for the genetic incorporation of an unnatural amino acid (UAA), 3'-trifluoromenthyl-phenylalanine (mtfF), into cannabinoid receptor 1 (CB1) in the Baculovirus Expression System. The probe can be inserted at any environmentally sensitive site, while causing minimal structural perturbation to the target protein. Using 19F NMR and X-ray crystallography methods, we discovered that the allosteric modulator Org27569 and agonists synergistically stabilize a previously unrecognized pre-active state. An allosteric modulation model is proposed to explain Org27569's distinct behavior. We demonstrate that our site-specific 19F NMR labeling method is a powerful tool in decoding the mechanism of GPCR allosteric modulation. This new method should be broadly applicable for uncovering conformational states for many important eukaryotic membrane proteins.


Subject(s)
Indoles , Piperidines
11.
Mol Psychiatry ; 25(9): 2086-2100, 2020 09.
Article in English | MEDLINE | ID: mdl-30120413

ABSTRACT

The dopamine (DA) D2 receptor (D2R) is an important target for the treatment of neuropsychiatric disorders such as schizophrenia and Parkinson's disease. However, the development of improved therapeutic strategies has been hampered by our incomplete understanding of this receptor's downstream signaling processes in vivo and how these relate to the desired and undesired effects of drugs. D2R is a G protein-coupled receptor (GPCR) that activates G protein-dependent as well as non-canonical arrestin-dependent signaling pathways. Whether these effector pathways act alone or in concert to facilitate specific D2R-dependent behaviors is unclear. Here, we report on the development of a D2R mutant that recruits arrestin but is devoid of G protein activity. When expressed virally in "indirect pathway" medium spiny neurons (iMSNs) in the ventral striatum of D2R knockout mice, this mutant restored basal locomotor activity and cocaine-induced locomotor activity in a manner indistinguishable from wild-type D2R, indicating that arrestin recruitment can drive locomotion in the absence of D2R-mediated G protein signaling. In contrast, incentive motivation was enhanced only by wild-type D2R, signifying a dissociation in the mechanisms that underlie distinct D2R-dependent behaviors, and opening the door to more targeted therapeutics.


Subject(s)
Arrestin , Locomotion , Motivation , Receptors, Dopamine D2 , Animals , Cocaine , Corpus Striatum/metabolism , Mice , Mice, Knockout , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism
12.
Behav Pharmacol ; 32(5): 453-458, 2021 08 01.
Article in English | MEDLINE | ID: mdl-33883450

ABSTRACT

G-protein-biased mu-opioid receptor (GPB-MOR) agonists are an emerging class of compounds being evaluated as candidate analgesics and agonist medications for opioid use disorder. Most of the basic pharmacology of GPB-MOR agonists has been conducted in rodents and much less is known how the basic behavioral pharmacology of these compounds translates to nonhuman primates. The present study determined the antinociceptive potency and time course of three putative GPB-MOR agonists: (+)-oliceridine (i.e. TRV130), SR14968, and SR17018 in male rhesus monkeys (n = 3). In addition, the respiratory effects of these compounds were also indirectly determined using a pulse oximeter to measure percent peripheral oxygen saturation (%SpO2). The largest intramuscular oliceridine dose (3.2 mg/kg) produced significant antinociception at 50°C, but not 54°C, and peak effects were between 10 and 30 min. Oliceridine also decreased SpO2 below the 90% threshold that would be clinically categorized as hypoxia in two out of three monkeys. The largest intramuscular SR14968 dose (0.32 mg/kg) produced 100% MPE at 50°C, but not 54°C, in two out of three monkeys, and peak effects were between 30 and 100 min. The largest intravenous SR17018 dose (1 mg/kg) produced 100% MPE at 50°C, but not 54°C, in the same two out of three monkeys, and peak effects were between 30 and 100 min. Solubility limitations for both SR14968 and SR17018 impaired our ability to determine in-vivo potency and effectiveness on antinociceptive and %SpO2 measures for these two compounds.


Subject(s)
GTP-Binding Proteins/metabolism , Opioid-Related Disorders/drug therapy , Pain/drug therapy , Receptors, Opioid, mu , Respiration/drug effects , Spiro Compounds/pharmacology , Thiophenes/pharmacology , Analgesics/pharmacology , Animals , Behavior, Animal , Drug Evaluation/methods , Macaca mulatta , Male , Oximetry/methods , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism
13.
Mol Pharmacol ; 96(5): 619-628, 2019 11.
Article in English | MEDLINE | ID: mdl-31515283

ABSTRACT

Cannabinoid receptor 1 (CB1) is a potential therapeutic target for the treatment of pain, obesity and obesity-related metabolic disorders, and addiction. The crystal structure of human CB1 has been determined in complex with the stabilizing antagonist AM6538. In the present study, we characterize AM6538 as a tight-binding/irreversible antagonist of CB1, as well as two derivatives of AM6538 (AM4112 and AM6542) as slowly dissociating CB1 antagonists across binding simulations and cellular signaling assays. The long-lasting nature of AM6538 was explored in vivo wherein AM6538 continues to block CP55,940-mediated behaviors in mice up to 5 days after a single injection. In contrast, the effects of SR141716A abate in mice 2 days after injection. These studies demonstrate the functional outcome of CB1 antagonist modification and open the path for development of long-lasting CB1 antagonists.


Subject(s)
Cannabinoid Receptor Antagonists/metabolism , Cannabinoid Receptor Antagonists/pharmacology , Nitrates/metabolism , Nitrates/pharmacology , Piperidines/metabolism , Piperidines/pharmacology , Pyrazoles/metabolism , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Animals , Binding Sites/drug effects , Binding Sites/physiology , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Protein Binding/drug effects , Protein Binding/physiology , Protein Structure, Secondary , Receptor, Cannabinoid, CB1/chemistry
14.
Bioorg Med Chem Lett ; 28(16): 2770-2772, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29426768

ABSTRACT

Salvinorin A (SalA) is a potent and selective agonist of the kappa-opioid receptor (KOR), but its instability has frustrated medicinal chemistry efforts. Treatment of SalA with weak bases like DBU leads to C8 epimerization with loss of receptor affinity and signaling potency. Here we show that replacement of C20 with H and replacement of O6 with CH2 stabilizes the SalA scaffold relative to its C8 epimer, so much so that epimerization is completely supressed. This new compound, O6C-20-nor-SalA, retains high potency for agonism of KOR.


Subject(s)
Carbon/pharmacology , Diterpenes, Clerodane/pharmacology , Oxygen/pharmacology , Receptors, Opioid, kappa/agonists , Carbon/chemistry , Diterpenes, Clerodane/chemical synthesis , Diterpenes, Clerodane/chemistry , Humans , Molecular Conformation , Oxygen/chemistry , Stereoisomerism , Structure-Activity Relationship
15.
J Am Chem Soc ; 139(16): 5865-5869, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28414442

ABSTRACT

Stolonidiol, a marine natural product, has been reported to potentiate the activity of choline acetyltransferase (ChAT), the enzyme that produces the neurotransmitter acetylcholine. Here we report the total synthesis of stolonidiol starting from (R)-(+)-limonene. To identify the mechanism by which ChAT activity is increased, we sought to identify the biological target of stolonidiol. We show that stolonidiol binds to the phorbol ester binding site of protein kinase C (PKC), induces translocation of PKC to the cell membrane, and activates kinase activity. Furthermore, we confirmed the increase in ChAT activity observed upon treatment of cells with stolonidiol and show that this effect is mediated by PKC. Collectively, our data strongly suggest that PKC activation by stolonidiol is responsible for the resulting potentiation of ChAT activity.


Subject(s)
Choline O-Acetyltransferase/metabolism , Diterpenes/pharmacology , Crystallography, X-Ray , Diterpenes/chemical synthesis , Diterpenes/chemistry , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , HEK293 Cells , Humans , Models, Molecular , Molecular Structure , Structure-Activity Relationship
16.
J Org Chem ; 81(21): 10538-10550, 2016 11 04.
Article in English | MEDLINE | ID: mdl-27399050

ABSTRACT

The quinoxaline and quinoxalinone family of nitrogen heterocycles is present in molecules of therapeutic relevance for diverse applications ranging from infectious diseases to neuroscience targets. Here, we describe a general synthetic sequence to afford pyrrolo[1,2-α]quinoxalinones from commercially available starting materials and their use in preparing potential kappa opioid receptor antagonists. The biological data obtained from the latter set of compounds is briefly presented and discussed.


Subject(s)
Pyrroles/chemistry , Quinoxalines/chemical synthesis , Receptors, Opioid, kappa/antagonists & inhibitors , Carbon-13 Magnetic Resonance Spectroscopy , Oxidation-Reduction , Proton Magnetic Resonance Spectroscopy , Quinoxalines/chemistry , Spectrometry, Mass, Electrospray Ionization
17.
Bioorg Med Chem Lett ; 26(2): 241-250, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26707396

ABSTRACT

G protein coupled receptors have historically been one of the most druggable classes of cellular proteins. The members of this large receptor gene family couple to primary effectors, G proteins, that have built in mechanisms for regeneration and amplification of signaling with each engagement of receptor and ligand, a kinetic event in itself. In recent years GPCRs, have been found to interact with arrestin proteins to initiate signal propagation in the absence of G protein interactions. This pinnacle observation has changed a previously held notion of the linear spectrum of GPCR efficacy and uncovered a new paradigm in GPCR research and drug discovery that relies on multidimensionality of GPCR signaling. Ligands were found that selectively confer activity in one pathway over another, and this phenomenon has been referred to as 'biased agonism' or 'functional selectivity'. While great strides in the understanding of this phenomenon have been made in recent years, two critical questions still dominate the field: How can we rationally design biased GPCR ligands, and ultimately, which physiological responses are due to G protein versus arrestin interactions? This review will discuss the current understanding of some of the key aspects of biased signaling that are related to these questions, including mechanistic insights in the nature of biased signaling and methods for measuring ligand bias, as well as relevant examples of drug discovery applications and medicinal chemistry strategies that highlight the challenges and opportunities in this rapidly evolving field.


Subject(s)
Arrestins/agonists , Drug Discovery , Receptors, G-Protein-Coupled/agonists , Animals , Arrestins/physiology , Humans , Ligands , Models, Molecular , Receptors, G-Protein-Coupled/physiology
18.
Mol Pharmacol ; 88(3): 579-88, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26134495

ABSTRACT

The fact that over 30% of current pharmaceuticals target heptahelical G protein-coupled receptors (GPCRs) attests to their tractability as drug targets. Although GPCR drug development has traditionally focused on conventional agonists and antagonists, the growing appreciation that GPCRs mediate physiologically relevant effects via both G protein and non-G protein effectors has prompted the search for ligands that can "bias" downstream signaling in favor of one or the other process. Biased ligands are novel entities with distinct signaling profiles dictated by ligand structure, and the potential prospect of biased ligands as better drugs has been pleonastically proclaimed. Indeed, preclinical proof-of-concept studies have demonstrated that both G protein and arrestin pathway-selective ligands can promote beneficial effects in vivo while simultaneously antagonizing deleterious ones. But along with opportunity comes added complexity and new challenges for drug discovery. If ligands can be biased, then ligand classification becomes assay dependent, and more nuanced screening approaches are needed to capture ligand efficacy across several dimensions of signaling. Moreover, because the signaling repertoire of biased ligands differs from that of the native agonist, unpredicted responses may arise in vivo as these unbalanced signals propagate. For any given GPCR target, establishing a framework relating in vitro efficacy to in vivo biologic response is crucial to biased drug discovery. This review discusses approaches to describing ligand efficacy in vitro, translating ligand bias into biologic response, and developing a systems-level understanding of biased agonism in vivo, with the overall goal of overcoming current barriers to developing biased GPCR therapeutics.


Subject(s)
Arrestins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Humans , Kinetics , Protein Binding , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/chemistry
19.
Mol Pharmacol ; 87(5): 866-77, 2015 May.
Article in English | MEDLINE | ID: mdl-25680753

ABSTRACT

Seven transmembrane receptors were originally named and characterized based on their ability to couple to heterotrimeric G proteins. The assortment of coupling partners for G protein-coupled receptors has subsequently expanded to include other effectors (most notably the ßarrestins). This diversity of partners available to the receptor has prompted the pursuit of ligands that selectively activate only a subset of the available partners. A biased or functionally selective ligand may be able to distinguish between different active states of the receptor, and this would result in the preferential activation of one signaling cascade more than another. Although application of the "standard" operational model for analyzing ligand bias is useful and suitable in most cases, there are limitations that arise when the biased agonist fails to induce a significant response in one of the assays being compared. In this article, we describe a quantitative method for measuring ligand bias that is particularly useful for such cases of extreme bias. Using simulations and experimental evidence from several κ opioid receptor agonists, we illustrate a "competitive" model for quantitating the degree and direction of bias. By comparing the results obtained from the competitive model with the standard model, we demonstrate that the competitive model expands the potential for evaluating the bias of very partial agonists. We conclude the competitive model provides a useful mechanism for analyzing the bias of partial agonists that exhibit extreme bias.


Subject(s)
Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Animals , Arrestins/metabolism , CHO Cells , Cell Line , Cricetulus , Heterotrimeric GTP-Binding Proteins/metabolism , Ligands , Receptors, Opioid, kappa/agonists , Signal Transduction/drug effects
20.
Mol Pharmacol ; 88(3): 534-5, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26162863

ABSTRACT

In August 2014, an international group of researchers gathered for 5 days at the Lorentz Center in Leiden, The Netherlands, to explore the technical and conceptual issues associated with the analysis of G protein-coupled receptor functions utilizing information from crystal structure models to the use of model organisms. This collection of review articles evolved from the 5-day meeting, with brief presentations and structured discussion periods that were designed to identify key questions remaining in understanding G protein-coupled receptor function and to propose novel strategies by integrating scientific disciplines to guide future research.


Subject(s)
Congresses as Topic , Receptors, G-Protein-Coupled/metabolism , Animals , Humans , Receptors, G-Protein-Coupled/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL