Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
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 ; 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
4.
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
5.
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
6.
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
7.
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
8.
Pharmacol Res ; 125(Pt B): 161-177, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28838808

ABSTRACT

The human cannabinoid subtype 1 receptor (hCB1R) is highly expressed in the CNS and serves as a therapeutic target for endogenous ligands as well as plant-derived and synthetic cannabinoids. Unfortunately, acute use of hCB1R agonists produces unwanted psychotropic effects and chronic administration results in development of tolerance and dependence, limiting the potential clinical use of these ligands. Studies in ß-arrestin knockout mice suggest that interaction of certain GPCRs, including µ-, δ-, κ-opioid and hCB1Rs, with ß-arrestins might be responsible for several adverse effects produced by agonists acting at these receptors. Indeed, agonists that bias opioid receptor activation toward G-protein, relative to ß-arrestin signaling, produce less severe adverse effects. These observations indicate that therapeutic utility of agonists acting at hCB1Rs might be improved by development of G-protein biased hCB1R agonists. Our laboratory recently reported a novel class of indole quinulidinone (IQD) compounds that bind cannabinoid receptors with relatively high affinity and act with varying efficacy. The purpose of this study was to determine whether agonists in this novel cannabinoid class exhibit ligand bias at hCB1 receptors. Our studies found that a novel IQD-derived hCB1 receptor agonist PNR-4-20 elicits robust G protein-dependent signaling, with transduction ratios similar to the non-biased hCB1R agonist CP-55,940. In marked contrast to CP-55,940, PNR-4-20 produces little to no ß-arrestin 2 recruitment. Quantitative calculation of bias factors indicates that PNR-4-20 exhibits from 5.4-fold to 29.5-fold bias for G protein, relative to ß-arrestin 2 signaling (when compared to G protein activation or inhibition of forskolin-stimulated cAMP accumulation, respectively). Importantly, as expected due to reduced ß-arrestin 2 recruitment, chronic exposure of cells to PNR-4-20 results in significantly less desensitization and down-regulation of hCB1Rs compared to similar treatment with CP-55,940. PNR-4-20 (i.p.) is active in the cannabinoid tetrad in mice and chronic treatment results in development of less persistent tolerance and no significant withdrawal signs when compared to animals repeatedly exposed to the non-biased full agoinst JWH-018 or Δ9-THC. Finally, studies of a structurally similar analog PNR- 4-02 show that it is also a G protein biased hCB1R agonist. It is predicted that cannabinoid agonists that bias hCB1R activation toward G protein, relative to ß-arrestin 2 signaling, will produce fewer and less severe adverse effects both acutely and chronically.


Subject(s)
Cannabinoid Receptor Agonists/pharmacology , GTP-Binding Proteins/metabolism , Quinuclidines/pharmacology , Animals , CHO Cells , Cricetulus , Cyclohexanols/pharmacology , Indoles/pharmacology , Male , Mice , Naphthalenes/pharmacology , Receptor, Cannabinoid, CB1/metabolism , beta-Arrestin 2/metabolism
9.
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
10.
Eur J Med Chem ; 276: 116627, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38971050

ABSTRACT

Kappa opioid receptor (KOR) agonists represent promising therapeutics for pain relief due to their analgesic properties along with lower abuse potential than opioids that act at the mu opioid receptor. However, typical KOR agonists produce sedation and dysphoria. Previous studies have shown that G protein signaling-biased KOR agonists may present a means to untangle the desired analgesic properties from undesired side effects. In this paper, we report a new series of G protein signaling-biased KOR agonists entailing -S- → -CH2- replacement in a previously reported KOR agonist, triazole 1.1. With an optimized carbon linker in hand, further development of the scaffold was undertaken to investigate the appendages of the triazole core. The structure-activity relationship study of this series is described, including several analogues that display enhanced potency while maintaining G protein-signaling bias compared to triazole 1.1.

11.
ACS Cent Sci ; 9(8): 1567-1574, 2023 Aug 23.
Article in English | MEDLINE | ID: mdl-37637743

ABSTRACT

The salvinorins serve as templates for next generation analgesics, antipruritics, and dissociative hallucinogens via selective and potent agonism of the kappa-opioid receptor (KOR). In contrast to most opioids, the salvinorins lack basic amines and bind with high affinity and selectivity via complex polyoxygenated scaffolds that have frustrated deep-seated modification by synthesis. Here we describe a short asymmetric synthesis that relies on a sterically confined organocatalyst to dissociate acidity from reactivity and effect Robinson annulation of an unactivated nucleophile/unstable electrophile pair. Combined with a cobalt-catalyzed polarized diene-alkyne cycloaddition, the route allows divergent access to a focused library of salvinorins. We appraise the synthesis by its generation of multiple analogs that exceed the potency, selectivity, stability, and functional bias of salvinorin A itself.

12.
Biomolecules ; 13(6)2023 06 02.
Article in English | MEDLINE | ID: mdl-37371516

ABSTRACT

Opioid analgesics such as morphine and fentanyl induce mu-opioid receptor (MOR)-mediated hyperactivity in mice. Herein, we show that morphine, fentanyl, SR-17018, and oliceridine have submaximal intrinsic efficacy in the mouse striatum using 35S-GTPγS binding assays. While all of the agonists act as partial agonists for stimulating G protein coupling in striatum, morphine, fentanyl, and oliceridine are fully efficacious in stimulating locomotor activity; meanwhile, the noncompetitive biased agonists SR-17018 and SR-15099 produce submaximal hyperactivity. Moreover, the combination of SR-17018 and morphine attenuates hyperactivity while antinociceptive efficacy is increased. The combination of oliceridine with morphine increases hyperactivity, which is maintained over time. These findings provide evidence that noncompetitive agonists at MOR can be used to suppress morphine-induced hyperactivity while enhancing antinociceptive efficacy; moreover, they demonstrate that intrinsic efficacy measured at the receptor level is not directly proportional to drug efficacy in the locomotor activity assay.


Subject(s)
Morphine , Spiro Compounds , Mice , Animals , Morphine/pharmacology , Analgesics, Opioid/pharmacology , Fentanyl/pharmacology
13.
Neuropsychopharmacology ; 45(2): 416-425, 2020 01.
Article in English | MEDLINE | ID: mdl-31443104

ABSTRACT

It has been demonstrated that opioid agonists that preferentially act at µ-opioid receptors to activate G protein signaling over ßarrestin2 recruitment produce antinociception with less respiratory suppression. However, most of the adverse effects associated with opioid therapeutics are realized after extended dosing. Therefore, we tested the onset of tolerance and dependence, and assessed for neurochemical changes associated with prolonged treatment with the biased agonist SR-17018. When chronically administered to mice, SR-17018 does not lead to hot plate antinociceptive tolerance, receptor desensitization in periaqueductal gray, nor a super-sensitization of adenylyl cyclase in the striatum, which are hallmarks of opioid neuronal adaptations that are seen with morphine. Interestingly, substitution with SR-17018 in morphine-tolerant mice restores morphine potency and efficacy, whereas the onset of opioid withdrawal is prevented. This is in contrast to buprenorphine, which can suppress withdrawal, but produces and maintains morphine antinociceptive tolerance. Biased agonists of this nature may therefore be useful for the treatment of opioid dependence while restoring opioid antinociceptive sensitivity.


Subject(s)
Analgesics, Opioid/metabolism , Drug Tolerance/physiology , Morphine Dependence/metabolism , Morphine/metabolism , Receptors, Opioid, mu/metabolism , Substance Withdrawal Syndrome/metabolism , Analgesics, Opioid/administration & dosage , Animals , Dose-Response Relationship, Drug , Female , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Infusion Pumps, Implantable , Male , Mice , Mice, Inbred C57BL , Morphine/administration & dosage , Oxycodone/administration & dosage , Oxycodone/metabolism , Pain Measurement/drug effects , Pain Measurement/methods , Receptors, Opioid, mu/agonists , Substance Withdrawal Syndrome/prevention & control
14.
Methods Mol Biol ; 1957: 235-247, 2019.
Article in English | MEDLINE | ID: mdl-30919358

ABSTRACT

G protein-coupled receptors (GPCRs) can interact with both G proteins and ß-arrestin proteins to propagate different signaling outputs. In some contexts, agonists may drive the receptor to preferentially engage one of these effectors over the other. Such "ligand bias" may present a means to impart pathway-selective signaling downstream of this class of receptors. In cases where physiological responses are mediated by diverse pathways, this could, in part, provide a means to refine GPCR therapeutics. Cell-based signaling assays are used to measure the potential for signaling bias in vitro, and these measures take into account potency, efficacy, and the overall capacity of the assay. However, narrow assay windows sometimes limit the confidence in estimating agonist activity, if a compound performs as a very weakly efficacious partial agonist. This lack of response in an assay hampers the ability to measure and compare potencies, and the degree of separation of an agonist's performance, between two assays. In this chapter, we describe in detail a method for the estimation of the relative activity of a partial agonist and provide a stepwise protocol for calculating bias when this case arises.


Subject(s)
Biological Assay/methods , Models, Biological , Bias , Confidence Intervals , Ligands , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism
15.
Sci Signal ; 11(542)2018 08 07.
Article in English | MEDLINE | ID: mdl-30087177

ABSTRACT

Biased agonists of G protein-coupled receptors may present a means to refine receptor signaling in a way that separates side effects from therapeutic properties. Several studies have shown that agonists that activate the κ-opioid receptor (KOR) in a manner that favors G protein coupling over ß-arrestin2 recruitment in cell culture may represent a means to treat pain and itch while avoiding sedation and dysphoria. Although it is attractive to speculate that the bias between G protein signaling and ß-arrestin2 recruitment is the reason for these divergent behaviors, little evidence has emerged to show that these signaling pathways diverge in the neuronal environment. We further explored the influence of cellular context on biased agonism at KOR ligand-directed signaling toward G protein pathways over ß-arrestin-dependent pathways and found that this bias persists in striatal neurons. These findings advance our understanding of how a G protein-biased agonist signal differs between cell lines and primary neurons, demonstrate that measuring [35S]GTPγS binding and the regulation of adenylyl cyclase activity are not necessarily orthogonal assays in cell lines, and emphasize the contributions of the environment to assessing biased agonism.


Subject(s)
GTP-Binding Proteins/metabolism , Neurons/metabolism , Receptors, Opioid, kappa/agonists , Signal Transduction , Animals , Animals, Newborn , Benzeneacetamides/pharmacology , CHO Cells , Cell Line, Tumor , Cells, Cultured , Corpus Striatum/cytology , Corpus Striatum/metabolism , Cricetinae , Cricetulus , HEK293 Cells , Humans , Mice, Knockout , Pyrrolidines/pharmacology , Receptors, Opioid, kappa/genetics , Receptors, Opioid, kappa/metabolism , beta-Arrestin 2/genetics , beta-Arrestin 2/metabolism
16.
Methods Enzymol ; 593: 259-279, 2017.
Article in English | MEDLINE | ID: mdl-28750807

ABSTRACT

G protein-coupled receptors, such as the cannabinoid type 1 receptor (CB1R), have been shown to interact with multiple binding partners to transmit signals. In both transfected cell systems and in endogenously expressing cell lines, CB1R signaling has been described as multifaceted. The question remains as to how this highly widely expressed receptor signals in a given cell at a given time in vivo. The concept of functional selectivity, or biased agonism, describes the ability of an agonist to engage the receptor in a manner that preferentially engages certain signaling interactions (e.g., G proteins) over others (e.g., ß-arrestins), presumably by stabilizing certain receptor conformations. There is growing interest in using such properties of ligands to direct signaling downstream of CB1R toward desirable therapeutic outcomes and to avoid adverse side effects. While it is not currently clear what pathways should be engaged and which should be avoided, the development of biased agonist tool compounds will aid in answering these questions. In this chapter, we discuss the approaches and caveats to assessing biased agonism at the CB1R.


Subject(s)
Cannabinoid Receptor Modulators/pharmacology , Receptor, Cannabinoid, CB1/physiology , Signal Transduction , Allosteric Regulation , Animals , Cells, Cultured , Humans , Kinetics , Ligands , Protein Processing, Post-Translational , Protein Transport
17.
Sci Signal ; 9(456): ra117, 2016 11 29.
Article in English | MEDLINE | ID: mdl-27899527

ABSTRACT

Agonists targeting the kappa opioid receptor (KOR) have been promising therapeutic candidates because of their efficacy for treating intractable itch and relieving pain. Unlike typical opioid narcotics, KOR agonists do not produce euphoria or lead to respiratory suppression or overdose. However, they do produce dysphoria and sedation, side effects that have precluded their clinical development as therapeutics. KOR signaling can be fine-tuned to preferentially activate certain pathways over others, such that agonists can bias signaling so that the receptor signals through G proteins rather than other effectors such as ßarrestin2. We evaluated a newly developed G protein signaling-biased KOR agonist in preclinical models of pain, pruritis, sedation, dopamine regulation, and dysphoria. We found that triazole 1.1 retained the antinociceptive and antipruritic efficacies of a conventional KOR agonist, yet it did not induce sedation or reductions in dopamine release in mice, nor did it produce dysphoria as determined by intracranial self-stimulation in rats. These data demonstrated that biased agonists may be used to segregate physiological responses downstream of the receptor. Moreover, the findings suggest that biased KOR agonists may present a means to treat pain and intractable itch without the side effects of dysphoria and sedation and with reduced abuse potential.


Subject(s)
Pain/drug therapy , Pruritus/drug therapy , Receptors, Opioid, kappa/agonists , Triazoles/pharmacology , Animals , Male , Mice , Pain/metabolism , Pruritus/metabolism , Rats , Rats, Inbred F344 , Receptors, Opioid, kappa/metabolism , Triazoles/chemistry , beta-Arrestin 2/metabolism
18.
ACS Chem Neurosci ; 6(8): 1411-9, 2015 Aug 19.
Article in English | MEDLINE | ID: mdl-25891774

ABSTRACT

Kappa opioid receptor (KOR) modulation is a promising target for drug discovery efforts due to KOR involvement in pain, depression, and addiction behaviors. We recently reported a new class of triazole KOR agonists that displays significant bias toward G protein signaling over ßarrestin2 recruitment; interestingly, these compounds also induce less activation of ERK1/2 map kinases than the balanced agonist, U69,593. We have identified structure-activity relationships around the triazole scaffold that allows for decreasing the bias for G protein signaling over ERK1/2 activation while maintaining the bias for G protein signaling over ßarrestin2 recruitment. The development of novel compounds, with different downstream signaling outcomes, independent of G protein/ßarrestin2 bias, provides a more diverse pharmacological toolset for use in defining complex KOR signaling and elucidating the significance of KOR-mediated signaling.


Subject(s)
Analgesics, Opioid/chemistry , Analgesics, Opioid/pharmacology , Receptors, Opioid, kappa/agonists , Animals , Arrestins/metabolism , Benzeneacetamides/pharmacology , CHO Cells , Cricetulus , Dose-Response Relationship, Drug , Drug Discovery , GTP-Binding Proteins/metabolism , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Pyrrolidines/pharmacology , Receptors, Opioid, kappa/metabolism , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship , beta-Arrestins
19.
Neuropharmacology ; 99: 131-41, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26160155

ABSTRACT

Differential modulation of kappa opioid receptor (KOR) signaling has been a proposed strategy for developing therapies for drug addiction and depression by either activating or blocking this receptor. Hence, there have been significant efforts to generate ligands with diverse pharmacological properties including partial agonists, antagonists, allosteric modulators as well as ligands that selectively activate some pathways while not engaging others (biased agonists). It is becoming increasingly evident that G protein coupled receptor signaling events are context dependent and that what may occur in cell based assays may not be fully indicative of signaling events that occur in the naturally occurring environment. As new ligands are developed, it is important to assess their signaling capacity in relevant endogenous systems in comparison to the performance of endogenous agonists. Since KOR is considered the cognate receptor for dynorphin peptides we have evaluated the selectivity profiles of dynorphin peptides in wild-type (WT), KOR knockout (KOR-KO), and mu opioid receptor knockout (MOR-KO) mice using [35S]GTPγS binding assay in striatal membrane preparations. We find that while the small molecule KOR agonist U69,593, is very selective for KOR, dynorphin peptides promiscuously stimulate G protein signaling in striatum. Furthermore, our studies demonstrate that norBNI and 5'GNTI are highly nonselective antagonists as they maintain full potency and efficacy against dynorphin signaling in the absence of KOR. Characterization of a new KOR antagonist, which may be more selective than NorBNI and 5'GNTI, is presented using this approach.


Subject(s)
Corpus Striatum/drug effects , Drug Evaluation, Preclinical/methods , Dynorphins/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Receptors, Opioid, kappa/metabolism , Analgesics, Opioid/pharmacology , Animals , Benzeneacetamides/pharmacology , Corpus Striatum/metabolism , Dynorphins/administration & dosage , Male , Mice, Inbred C57BL , Mice, Knockout , Narcotic Antagonists/pharmacology , Protein Binding , Pyrrolidines/pharmacology , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, kappa/genetics , Signal Transduction/drug effects , Sulfur Radioisotopes
SELECTION OF CITATIONS
SEARCH DETAIL