Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
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
2.
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
3.
Neuropharmacology ; 185: 108439, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33345829

ABSTRACT

The mu opioid receptor-selective agonist, SR-17018, preferentially activates GTPγS binding over ßarrestin2 recruitment in cellular assays, thereby demonstrating signaling bias. In mice, SR-17018 stimulates GTPγS binding in brainstem and produces antinociception with potencies similar to morphine. However, it produces much less respiratory suppression and mice do not develop antinociceptive tolerance in the hot plate assay upon repeated dosing. Herein we evaluate the effects of acute and repeated dosing of SR-17018, oxycodone and morphine in additional models of pain-related behaviors. In the mouse warm water tail immersion assay, an assessment of spinal reflex to thermal nociception, repeated administration of SR-17018 produces tolerance as does morphine and oxycodone. SR-17018 retains efficacy in a formalin-induced inflammatory pain model upon repeated dosing, while oxycodone does not. In a chemotherapeutic-induced neuropathy pain model SR-17018 is more potent and efficacious than morphine or oxycodone, moreover, this efficacy is retained upon repeated dosing of SR-17018. These findings demonstrate that, with the exception of the tail flick test, SR-17018 retains efficacy upon chronic treatment across several pain models.


Subject(s)
Analgesics, Opioid/administration & dosage , Benzimidazoles/administration & dosage , Morphine/administration & dosage , Neuralgia/drug therapy , Oxycodone/administration & dosage , Piperidines/administration & dosage , Receptors, Opioid, mu/agonists , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Tolerance , Female , Infusion Pumps, Implantable , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Neuralgia/pathology , Pain Measurement/drug effects , Pain Measurement/methods , Treatment Outcome
4.
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
5.
J Med Chem ; 61(19): 8895-8907, 2018 10 11.
Article in English | MEDLINE | ID: mdl-30199635

ABSTRACT

While mu opioid receptor (MOR) agonists are especially effective as broad-spectrum pain relievers, it has been exceptionally difficult to achieve a clear separation of analgesia from many problematic side effects. Recently, many groups have sought MOR agonists that induce minimal ßarrestin-mediated signaling because MOR agonist-treated ßarrestin2 knockout mice were found to display enhanced antinociceptive effects with significantly less respiratory depression and tachyphylaxis. Substantial data now exists to support the premise that G protein signaling biased MOR agonists can be effective analgesic agents. We recently showed that, within a chemical series, the degree of bias correlates linearly with the magnitude of the respiratory safety index. Herein we describe the synthesis and optimization of piperidine benzimidazolone MOR agonists that together display a wide range of bias (G/ßarr2). We identify structural features affecting potency and maximizing bias and show that many compounds have desirable properties, such as long half-lives and high brain penetration.


Subject(s)
Analgesics, Opioid/pharmacology , Blood-Brain Barrier/metabolism , Drug Discovery/standards , GTP-Binding Proteins/metabolism , Microsomes, Liver/metabolism , Receptors, Opioid, mu/agonists , Analgesics, Opioid/chemistry , Animals , Blood-Brain Barrier/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Microsomes, Liver/drug effects , Protein Conformation , Structure-Activity Relationship , beta-Arrestins/metabolism
6.
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
7.
Drug Alcohol Depend ; 191: 195-202, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30138791

ABSTRACT

Previous reports assessing morphine effects in two bottle choice (TBC) paradigms often use taste adulterants such as sweeteners (e.g., saccharin) and/or bitterants (e.g., quinine) to demonstrate morphine preference with C57BL6 mice. The effect of these additional components on the morphine preference of C57BL6 remains poorly understood. Thus, we sought to elucidate the interrelationship of morphine and quinine in the TBC paradigm. As expected, when morphine was included in the opposite bottle from quinine, a preference for the morphine solution was observed. Conversely, when quinine was included in each bottle, or when fentanyl without quinine was used, no preference was observed. All opioid-drinking mice displayed withdrawal signs, and morphine was detectable in plasma and brain. When these results were compared to previous results via conversion to quinine preference scores, quinine was revealed to determine largely the measured morphine preference. Thus, quinine is effective to drive morphine consumption and engender dependence but may confound the ability to measure oral abuse liability of morphine. Together, these results suggest future TBC procedures should consider the effect of quinine upon measured preference for compounds in the opposite bottle, and that excessively high quinine concentrations appear to influence preference more so than the opposite solute when using C57BL6 mice. Alternative conditions to assess oral abuse liability may be necessary to complement and confirm results from TBC experiments utilizing morphine or other opioids.


Subject(s)
Analgesics, Opioid/administration & dosage , Choice Behavior/drug effects , Quinine/administration & dosage , Substance Withdrawal Syndrome/metabolism , Analgesics, Opioid/pharmacokinetics , Animals , Brain/drug effects , Brain/metabolism , Choice Behavior/physiology , Male , Mice , Mice, Inbred C57BL , Morphine/administration & dosage , Quinine/pharmacokinetics , Saccharin/administration & dosage , Substance Withdrawal Syndrome/diagnosis , Taste/drug effects
8.
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
9.
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
10.
ACS Cent Sci ; 3(12): 1329-1336, 2017 Dec 27.
Article in English | MEDLINE | ID: mdl-29296674

ABSTRACT

Salvinorin A (SalA) is a plant metabolite that agonizes the human kappa-opioid receptor (κ-OR) with high affinity and high selectivity over mu- and delta-opioid receptors. Its therapeutic potential has stimulated extensive semisynthetic studies and total synthesis campaigns. However, structural modification of SalA has been complicated by its instability, and efficient total synthesis has been frustrated by its dense, complex architecture. Treatment of strategic bonds in SalA as dynamic and dependent on structural perturbation enabled the identification of an efficient retrosynthetic pathway. Here we show that deletion of C20 simultaneously stabilizes the SalA skeleton, simplifies its synthesis, and retains its high affinity and selectivity for the κ-OR. The resulting 10-step synthesis now opens the SalA scaffold to deep-seated property modification. Finally, we describe a workflow to identify structural changes that retain molecular complexity, but reduce synthetic complexity-two related, but independent ways of looking at complexity.

11.
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
12.
Mol Cell Biol ; 36(1): 223-35, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26503786

ABSTRACT

Primary cilia are essential sensory and signaling organelles present on nearly every mammalian cell type. Defects in primary cilia underlie a class of human diseases collectively termed ciliopathies. Primary cilia are restricted subcellular compartments, and specialized mechanisms coordinate the localization of proteins to cilia. Moreover, trafficking of proteins into and out of cilia is required for proper ciliary function, and this process is disrupted in ciliopathies. The somatostatin receptor subtype 3 (Sstr3) is selectively targeted to primary cilia on neurons in the mammalian brain and is implicated in learning and memory. Here, we show that Sstr3 localization to cilia is dynamic and decreases in response to somatostatin treatment. We further show that somatostatin treatment stimulates ß-arrestin recruitment into Sstr3-positive cilia and this recruitment can be blocked by mutations in Sstr3 that impact agonist binding or phosphorylation. Importantly, somatostatin treatment fails to decrease Sstr3 ciliary localization in neurons lacking ß-arrestin 2. Together, our results implicate ß-arrestin in the modulation of Sstr3 ciliary localization and further suggest a role for ß-arrestin in the mediation of Sstr3 ciliary signaling.


Subject(s)
Arrestins/metabolism , Cilia/metabolism , Memory/physiology , Neurons/metabolism , Receptors, Somatostatin/metabolism , Animals , Brain/metabolism , Cells, Cultured , Learning/physiology , Mice , Signal Transduction/physiology , beta-Arrestin 2 , beta-Arrestins
13.
Neuropsychopharmacology ; 39(8): 1902-13, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24531562

ABSTRACT

The G protein-coupled serotonin 2A receptor (5-HT2AR) is a prominent target for atypical antipsychotic drugs, such as clozapine. Although clozapine is known to inhibit 5-HT2AR signaling through G protein-dependent mechanisms, it differs from classic GPCR antagonists, in that it also induces 5-HT2AR internalization and activates Akt signaling via a 5-HT2AR-mediated event. In this regard, clozapine may also be considered a functionally selective agonist. The cognate neurotransmitter at the 5-HT2AR, serotonin, also induces 5-HT2AR internalization and Akt phosphorylation. Serotonin promotes interactions with the scaffolding and regulatory protein, ßarrestin2, which results in the recruitment and activation of Akt. These interactions prove to be critical for serotonin-induced, 5-HT2AR-mediated behavioral responses in mice. Herein, we sought to determine whether clozapine also utilizes ßarrestin2-mediated mechanisms to induce 5-HT2AR signaling, and whether this interaction contributes to its behavioral effects in mice. We demonstrate that unlike serotonin, clozapine-mediated 5-HT2AR internalization and Akt phosphorylation is independent of receptor interactions with ßarrestin2. Moreover, clozapine-mediated suppression of MK-801 and phencyclidine (PCP)-induced hyperlocomotion is ßarrestin2 independent, although it is dependent upon Akt. These results demonstrate that pharmacologically oppositional ligands, serotonin and clozapine, utilize differential mechanisms to achieve the same 5-HT2AR-meadiated downstream events: Akt phosphorylation and receptor internalization. Although ßarrestin2 has no effect on clozapine's actions in vivo, Akt phosphorylation is required for clozapine's efficacy in blocking MK-801- and PCP-induced models of schizophrenic behaviors in mice.


Subject(s)
Arrestins/metabolism , Clozapine/pharmacology , Motor Activity/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Serotonin 5-HT2 Receptor Agonists/pharmacology , Animals , Cells, Cultured , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , HEK293 Cells , Humans , Mice , Mice, Transgenic , Phosphorylation , beta-Arrestins
14.
J Biol Chem ; 288(51): 36703-16, 2013 Dec 20.
Article in English | MEDLINE | ID: mdl-24187130

ABSTRACT

The kappa opioid receptor (KOR) is widely expressed in the CNS and can serve as a means to modulate pain perception, stress responses, and affective reward states. Therefore, the KOR has become a prominent drug discovery target toward treating pain, depression, and drug addiction. Agonists at KOR can promote G protein coupling and ßarrestin2 recruitment as well as multiple downstream signaling pathways, including ERK1/2 MAPK activation. It has been suggested that the physiological effects of KOR activation result from different signaling cascades, with analgesia being G protein-mediated and dysphoria being mediated through ßarrestin2 recruitment. Dysphoria associated with KOR activation limits the therapeutic potential in the use of KOR agonists as analgesics; therefore, it may be beneficial to develop KOR agonists that are biased toward G protein coupling and away from ßarrestin2 recruitment. Here, we describe two classes of biased KOR agonists that potently activate G protein coupling but weakly recruit ßarrestin2. These potent and functionally selective small molecule compounds may prove to be useful tools for refining the therapeutic potential of KOR-directed signaling in vivo.


Subject(s)
Receptors, Opioid, kappa/agonists , Animals , Arrestins/metabolism , CHO Cells , Cricetinae , Cricetulus , Drug Discovery , GTP-Binding Proteins/metabolism , Humans , Ligands , Male , Mice , Mice, Inbred C57BL , Quinolones/chemical synthesis , Quinolones/pharmacology , Receptors, Opioid, kappa/metabolism , Signal Transduction , Triazoles/chemical synthesis , Triazoles/pharmacology , beta-Arrestins
15.
J Biol Chem ; 288(31): 22387-98, 2013 Aug 02.
Article in English | MEDLINE | ID: mdl-23775075

ABSTRACT

There is considerable evidence to suggest that drug actions at the κ-opioid receptor (KOR) may represent a means to control pain perception and modulate reward thresholds. As a G protein-coupled receptor (GPCR), the activation of KOR promotes Gαi/o protein coupling and the recruitment of ß-arrestins. It has become increasingly evident that GPCRs can transduce signals that originate independently via G protein pathways and ß-arrestin pathways; the ligand-dependent bifurcation of such signaling is referred to as "functional selectivity" or "signaling bias." Recently, a KOR agonist, 6'-guanidinonaltrindole (6'-GNTI), was shown to display bias toward the activation of G protein-mediated signaling over ß-arrestin2 recruitment. Therefore, we investigated whether such ligand bias was preserved in striatal neurons. Although the reference KOR agonist U69,593 induces the phosphorylation of ERK1/2 and Akt, 6'-GNTI only activates the Akt pathway in striatal neurons. Using pharmacological tools and ß-arrestin2 knock-out mice, we show that KOR-mediated ERK1/2 phosphorylation in striatal neurons requires ß-arrestin2, whereas Akt activation depends upon G protein signaling. These findings reveal a point of KOR signal bifurcation that can be observed in an endogenous neuronal setting and may prove to be an important indicator when developing biased agonists at the KOR.


Subject(s)
Corpus Striatum/drug effects , Guanidines/pharmacology , Naltrexone/analogs & derivatives , Neurons/drug effects , Receptors, Opioid, kappa/drug effects , Animals , CHO Cells , Corpus Striatum/cytology , Corpus Striatum/metabolism , Cricetinae , Cricetulus , MAP Kinase Signaling System , Male , Mice , Naltrexone/pharmacology , Neurons/metabolism , Phosphorylation
16.
Biol Psychiatry ; 71(8): 714-24, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22264443

ABSTRACT

BACKGROUND: Cannabinoid CB(1) receptors (CB(1)Rs) mediate the effects of ▵(9)-tetrahydrocannabinol (THC), the psychoactive component in marijuana. Repeated THC administration produces tolerance and dependence, which limit therapeutic development. Moreover, THC produces motor and psychoactive side effects. ß-arrestin2 mediates receptor desensitization, internalization, and signaling, but its role in these CB(1)R effects and receptor regulation is unclear. METHODS: CB(1)R signaling and behaviors (antinociception, hypothermia, catalepsy) were assessed in ß-arrestin2-knockout (ßarr2-KO) and wild-type mice after THC administration. Cannabinoid-stimulated [(35)S]GTPγS and [(3)H]ligand autoradiography were assessed by statistical parametric mapping and region-of-interest analysis. RESULTS: ß-arrestin2 deletion increased CB(1)R-mediated G-protein activity in subregions of the cortex but did not affect CB(1)R binding, in vehicle-treated mice. ßarr2-KO mice exhibited enhanced acute THC-mediated antinociception and hypothermia, with no difference in catalepsy. After repeated THC administration, ßarr2-KO mice showed reduced CB(1)R desensitization and/or downregulation in cerebellum, caudal periaqueductal gray, and spinal cord and attenuated tolerance to THC-mediated antinociception. In contrast, greater desensitization was found in hypothalamus, cortex, globus pallidus, and substantia nigra of ßarr2-KO compared with wild-type mice. Enhanced tolerance to THC-induced catalepsy was observed in ßarr2-KO mice. CONCLUSIONS: ß-arrestin2 regulation of CB(1)R signaling following acute and repeated THC administration was region-specific, and results suggest that multiple, overlapping mechanisms regulate CB(1)Rs. The observations that ßarr2-KO mice display enhanced antinociceptive responses to acute THC and decreased tolerance to the antinociceptive effects of the drug, yet enhanced tolerance to catalepsy, suggest that development of cannabinoid drugs that minimize CB(1)R interactions with ß-arrestin2 might produce improved cannabinoid analgesics with reduced motor suppression.


Subject(s)
Arrestins/metabolism , Dronabinol/metabolism , Drug Tolerance/physiology , Receptor, Cannabinoid, CB1/metabolism , Animals , Autoradiography , Catalepsy/metabolism , Central Nervous System/metabolism , Down-Regulation , Dronabinol/pharmacology , G-Protein-Coupled Receptor Kinases/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Hypothermia/metabolism , Mice , Mice, Knockout , Nociception/drug effects , Signal Transduction , Sulfur Radioisotopes , beta-Arrestins
17.
Pharmacol Rev ; 63(4): 1001-19, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21873412

ABSTRACT

Opioids are the most effective analgesic drugs for the management of moderate or severe pain, yet their clinical use is often limited because of the onset of adverse side effects. Drugs in this class produce most of their physiological effects through activation of the µ opioid receptor; however, an increasing number of studies demonstrate that different opioids, while presumably acting at this single receptor, can activate distinct downstream responses, a phenomenon termed functional selectivity. Functional selectivity of receptor-mediated events can manifest as a function of the drug used, the cellular or neuronal environment examined, or the signaling or behavioral measure recorded. This review summarizes both in vitro and in vivo work demonstrating functional selectivity at the µ opioid receptor in terms of G protein coupling, receptor phosphorylation, interactions with ß-arrestins, receptor desensitization, internalization and signaling, and details on how these differences may relate to the progression of analgesic tolerance after their extended use.


Subject(s)
Analgesics, Opioid/pharmacology , Analgesics/pharmacology , Pain/drug therapy , Receptors, Opioid, mu/physiology , Analgesics/therapeutic use , Analgesics, Opioid/therapeutic use , Drug Tolerance , GTP-Binding Protein Regulators/drug effects , GTP-Binding Protein Regulators/physiology , Humans , Pain/physiopathology
18.
J Biol Chem ; 286(36): 31731-41, 2011 Sep 09.
Article in English | MEDLINE | ID: mdl-21757712

ABSTRACT

Morphine and other opiates mediate their effects through activation of the µ-opioid receptor (MOR), and regulation of the MOR has been shown to critically affect receptor responsiveness. Activation of the MOR results in receptor phosphorylation, ß-arrestin recruitment, and internalization. This classical regulatory process can differ, depending on the ligand occupying the receptor. There are two forms of ß-arrestin, ß-arrestin1 and ß-arrestin2 (also known as arrestin2 and arrestin3, respectively); however, most studies have focused on the consequences of recruiting ß-arrestin2 specifically. In this study, we examine the different contributions of ß-arrestin1- and ß-arrestin2-mediated regulation of the MOR by comparing MOR agonists in cells that lack expression of individual or both ß-arrestins. Here we show that morphine only recruits ß-arrestin2, whereas the MOR-selective enkephalin [D-Ala(2),N-Me-Phe(4),Gly(5)-ol]enkephalin (DAMGO), recruits either ß-arrestin. We show that ß-arrestins are required for receptor internalization and that only ß-arrestin2 can rescue morphine-induced MOR internalization, whereas either ß-arrestin can rescue DAMGO-induced MOR internalization. DAMGO activation of the receptor promotes MOR ubiquitination over time. Interestingly, ß-arrestin1 proves to be critical for MOR ubiquitination as modification does not occur in the absence of ß-arrestin1 nor when morphine occupies the receptor. Moreover, the selective interactions between the MOR and ß-arrestin1 facilitate receptor dephosphorylation, which may play a role in the resensitization of the MOR and thereby contribute to overall development of opioid tolerance.


Subject(s)
Arrestins/agonists , Arrestins/metabolism , Receptors, Opioid, mu/metabolism , Analgesics, Opioid , Animals , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Enkephalins , Mice , Phosphorylation , Protein Transport , Ubiquitination , beta-Arrestins
19.
J Neurosci ; 30(40): 13513-24, 2010 Oct 06.
Article in English | MEDLINE | ID: mdl-20926677

ABSTRACT

Hallucinogens mediate many of their psychoactive effects by activating serotonin 2A receptors (5-HT(2A)R). Although serotonin is the cognate endogenous neurotransmitter and is not considered hallucinogenic, metabolites of serotonin also have high affinity at 5-HT(2A)R and can induce hallucinations in humans. Here we report that serotonin differs from the psychoactive N-methyltryptamines by its ability to engage a ß-arrestin2-mediated signaling cascade in the frontal cortex. Serotonin and 5-hydroxy-L-tryptophan (5-HTP) induce a head-twitch response in wild-type (WT) mice that is a behavioral proxy for 5-HT(2A)R activation. The response in ß-arrestin2 knock-out (ßarr2-KO) mice is greatly attenuated until the doses are elevated, at which point, ßarr2-KO mice display a head-twitch response that can exceed that of WT mice. Direct administration of N-methyltryptamines also produces a greater response in ßarr2-KO mice. Moreover, the inhibition of N-methyltransferase blocks 5-HTP-induced head twitches in ßarr2-KO mice, indicating that N-methyltryptamines, rather than serotonin, primarily mediate this response. Biochemical studies demonstrate that serotonin stimulates Akt phosphorylation in the frontal cortex and in primary cortical neurons through the activation of a ß-arrestin2/phosphoinositide 3-kinase/Src/Akt cascade, whereas N-methyltryptamines do not. Furthermore, disruption of any of the components of this cascade prevents 5-HTP-induced, but not N-methyltryptamine-induced, head twitches. We propose that there is a bifurcation of 5-HT(2A)R signaling that is neurotransmitter and ß-arrestin2 dependent. This demonstration of agonist-directed 5-HT(2A)R signaling in vivo may significantly impact drug discovery efforts for the treatment of disorders wherein hallucinations are part of the etiology, such as schizophrenia, or manifest as side effects of treatment, such as depression.


Subject(s)
Arrestins/physiology , Neurons/metabolism , Proto-Oncogene Proteins c-akt/physiology , Receptor, Serotonin, 5-HT2A/metabolism , Serotonin/physiology , Signal Transduction/physiology , Tryptamines/physiology , src-Family Kinases/physiology , Adaptor Proteins, Signal Transducing , Animals , Animals, Newborn , Arrestins/genetics , Cells, Cultured , Frontal Lobe/cytology , Frontal Lobe/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Neurons/enzymology , Serotonin/metabolism , Serotonin 5-HT2 Receptor Agonists , Synaptic Transmission/physiology , Tryptamines/metabolism , beta-Arrestins
20.
Crit Rev Biochem Mol Biol ; 45(6): 555-66, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20925600

ABSTRACT

Serotonin receptors are the product of 15 distinct genes, 14 of which are G protein-coupled receptors. These receptors are expressed in a wide range of cell types, including distinct neuronal populations, and promote diverse functional responses in multiple organ systems. These receptors are important for mediating the in vivo effects of their cognate neurotransmitter, serotonin, as well as the endogenous tryptamines. In addition, the actions of many drugs are mediated, either directly or indirectly, through serotonin receptors, including antidepressants, antipsychotics, anxiolytics, sleep aids, migraine therapies, gastrointestinal therapeutics and hallucinogenic drugs. It is becoming increasingly evident that serotonin receptors can engage in differential signaling that is determined by the chemical nature of the ligand and that ligands that demonstrate a predilection for inducing a particular signaling cascade are considered to have "functional selectivity". The elucidation of the cellular signaling pathways that mediate the physiological responses to serotonin and other agonists is an active area of investigation and will be an onward-looking focal point for determining how to effectively and selectively promote beneficial serotonergic mimicry while avoiding unwanted clinical side effects. This review highlights the modulation of serotonin 2A, 2C, and four receptors by ß-arrestins, which may represent a fulcrum for biasing receptor responsiveness in vivo.


Subject(s)
Arrestins/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Serotonin/metabolism , Serotonin/metabolism , Serotonin/pharmacology , Signal Transduction , Animals , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Antipsychotic Agents/pharmacology , Arrestins/genetics , Central Nervous System/metabolism , Hallucinogens/pharmacology , Humans , Neurons/drug effects , Neurons/metabolism , Peripheral Nervous System/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Serotonin/genetics , Serotonin Receptor Agonists/pharmacology , Signal Transduction/drug effects , beta-Arrestins
SELECTION OF CITATIONS
SEARCH DETAIL
...