Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 180(18): 2341-2360, 2023 09.
Article in English | MEDLINE | ID: mdl-37005796

ABSTRACT

BACKGROUND AND PURPOSE: The illicit use of fentanyl-like drugs (fentanyls), which are µ opioid receptor agonists, and the many overdose deaths that result, has become a major problem. Fentanyls are very potent in vivo, leading to respiratory depression and death. However, the efficacy and possible signalling bias of different fentanyls is not clearly known. Here, we compared the relative efficacy and bias of a series of fentanyls. EXPERIMENTAL APPROACH: For agonist signalling bias and efficacy measurements, Bioluminescence Resonance Energy Transfer experiments were undertaken in HEK293T cells transiently transfected with µ opioid receptors, to assess Gi protein activation and ß-arrestin 2 recruitment. Agonist-induced cell surface receptor loss was assessed using an enzyme-linked immunosorbent assay, whilst agonist-induced G protein-coupled inwardly rectifying potassium channel current activation was measured electrophysiologically from rat locus coeruleus slices. Ligand poses in the µ opioid receptor were determined in silico using molecular dynamics simulations. KEY RESULTS: Relative to the reference ligand DAMGO, carfentanil was ß-arrestin-biased, whereas fentanyl, sufentanil and alfentanil did not display bias. Carfentanil induced potent and extensive cell surface receptor loss, whilst the marked desensitisation of G protein-coupled inwardly rectifying potassium channel currents in the continued presence of carfentanil in neurones was prevented by a GRK2/3 inhibitor. Molecular dynamics simulations suggested unique interactions of carfentanil with the orthosteric site of the receptor that could underlie the bias. CONCLUSIONS AND IMPLICATIONS: Carfentanil is a ß-arrestin-biased opioid drug at the µ receptor. It is uncertain how such bias influences in vivo effects of carfentanil relative to other fentanyls.


Subject(s)
Potassium Channels, Inwardly Rectifying , Receptors, Opioid, mu , Rats , Humans , Animals , Receptors, Opioid, mu/metabolism , beta-Arrestins/metabolism , Arrestin/metabolism , Ligands , HEK293 Cells , Fentanyl/pharmacology , Analgesics, Opioid/pharmacology , GTP-Binding Proteins/metabolism , beta-Arrestin 1/metabolism
2.
Br J Pharmacol ; 180(7): 943-957, 2023 04.
Article in English | MEDLINE | ID: mdl-33245558

ABSTRACT

BACKGROUND AND PURPOSE: G protein-biased µ opioid receptor agonists have the potential to induce less receptor desensitisation and tolerance than balanced opioids. Here, we investigated if the cyclic endomorphin analogue Tyr-c[D-Lys-Phe-Tyr-Gly] (Compound 1) is a G protein-biased µ agonist and characterised its ability to induce rapid receptor desensitisation in mammalian neurones. EXPERIMENTAL APPROACH: The signalling and trafficking properties of opioids were characterised using bioluminescence resonance energy transfer assays, enzyme-linked immunosorbent assay and phosphosite-specific immunoblotting in human embryonic kidney 293 cells. Desensitisation of opioid-induced currents were studied in rat locus coeruleus neurones using whole-cell patch-clamp electrophysiology. The mechanism of Compound 1-induced µ receptor desensitisation was probed using kinase inhibitors. KEY RESULTS: Compound 1 has similar intrinsic activity for G protein signalling as morphine. As predicted for a G protein-biased µ agonist, Compound 1 induced minimal agonist-induced internalisation and phosphorylation at intracellular µ receptor serine/threonine residues known to be involved in G protein-coupled receptor kinase (GRK)-mediated desensitisation. However, Compound 1 induced robust rapid µ receptor desensitisation in locus coeruleus neurons, to a greater degree than morphine. The extent of Compound 1-induced desensitisation was unaffected by activation or inhibition of protein kinase C (PKC) but was significantly reduced by inhibition of GRK. CONCLUSION AND IMPLICATIONS: Compound 1 is a novel G protein-biased µ agonist that induces substantial rapid receptor desensitisation in mammalian neurons. Surprisingly, Compound 1-induced desensitisation was demonstrated to be GRK dependent despite its G protein bias. Our findings refute the assumption that G protein-biased agonists will evade receptor desensitisation and tolerance. LINKED ARTICLES: This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc.


Subject(s)
Analgesics, Opioid , Receptors, Opioid, mu , Rats , Humans , Animals , Analgesics, Opioid/pharmacology , Receptors, Opioid, mu/metabolism , GTP-Binding Proteins/metabolism , Morphine/pharmacology , Signal Transduction , G-Protein-Coupled Receptor Kinases/metabolism , Mammals/metabolism
3.
J Psychopharmacol ; 35(10): 1204-1215, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33691518

ABSTRACT

BACKGROUND: α7 Nicotinic acetylcholine receptors are implicated in the reinstatement of drug-seeking, an important component of relapse. We showed previously that the α7 nicotinic acetylcholine receptor antagonist, methyllycaconitine, specifically attenuated morphine-primed reinstatement of conditioned place preference in rodents and this effect was mediated in the ventral hippocampus. AIMS: The purpose of this study was to evaluate α7 nicotinic acetylcholine receptor antagonism in reinstatement of the conditioned place preference for the more widely abused opioid, heroin, and to compare the effect of α7 nicotinic acetylcholine receptor blockade on reinstatement of heroin-seeking and heroin self-administration in an intravenous self-administration model of addictive behaviour. METHODS: Rats were trained to acquire heroin conditioned place preference or heroin self-administration; both followed by extinction of responding. Methyllycaconitine or saline was given prior to reinstatement of drug-primed conditioned place preference, or drug-prime plus cue-induced reinstatement of intravenous self-administration, using two protocols: without delivery of heroin in response to lever pressing to model heroin-seeking, or with heroin self-administration, using fixed and progressive ratio reward schedules, to model relapse. RESULTS: Methyllycaconitine had no effect on acquisition of heroin conditioned place preference or lever-pressing for food rewards. Methyllycaconitine blocked reinstatement of heroin-primed conditioned place preference. Methyllycaconitine did not prevent drug-prime plus cue-induced reinstatement of heroin-seeking, reinstatement of heroin self-administration, or diminish the reinforcing effect of heroin. CONCLUSIONS: The α7 nicotinic acetylcholine receptor antagonist, methyllycaconitine, prevented reinstatement of the opioid conditioned place preference, consistent with a role for α7 nicotinic acetylcholine receptors in the retrieval of associative memories of drug liking. The lack of effect of methyllycaconitine in heroin-dependent rats in two intravenous self-administration models suggests that α7 nicotinic acetylcholine receptors do not play a role in later stages of heroin abuse.


Subject(s)
Aconitine/analogs & derivatives , Heroin Dependence/physiopathology , Heroin/administration & dosage , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors , Aconitine/pharmacology , Animals , Behavior, Addictive/physiopathology , Conditioning, Psychological/drug effects , Cues , Drug-Seeking Behavior/physiology , Extinction, Psychological/drug effects , Male , Nicotinic Antagonists/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Reinforcement, Psychology , Reward , Self Administration , alpha7 Nicotinic Acetylcholine Receptor/metabolism
4.
Br J Pharmacol ; 177(15): 3489-3504, 2020 08.
Article in English | MEDLINE | ID: mdl-32297669

ABSTRACT

BACKGROUND AND PURPOSE: Various GPCRs have been described as being modulated in a voltage-dependent manner. Opioid analgesics act via activation of µ receptors in various neurons. As neurons are exposed to large changes in membrane potential, we were interested in studying the effects of depolarization on µ receptor signalling. EXPERIMENTAL APPROACH: We investigated potential voltage sensitivity of µ receptors in heterologous expression systems (HEK293T cells) using electrophysiology in combination with Förster resonance energy transfer-based assays. Depolarization-induced changes in signalling were also tested in physiological rat tissue containing locus coeruleus neurons. We applied depolarization steps across the physiological range of membrane potentials. KEY RESULTS: Studying µ receptor function and signalling in cells, we discovered that morphine-induced signalling was strongly dependent on the membrane potential (VM ). This became apparent at the level of G-protein activation, G-protein coupled inwardly rectifying potassium channel (Kir 3.X) currents and binding of GPCR kinases and arrestin3 to µ receptors by a robust increase in signalling upon membrane depolarization. The pronounced voltage sensitivity of morphine-induced µ receptor activation was also observed at the level of Kir 3.X currents in rat locus coeruleus neurons. The efficacy of peptide ligands to activate µ receptors was not (Met-enkephalin) or only moderately ([D-Ala2 , N-Me-Phe4 , Gly5 -ol]-enkephalin) enhanced upon depolarization. In contrast, depolarization reduced the ability of the analgesic fentanyl to activate µ receptors. CONCLUSION AND IMPLICATIONS: Our results indicate a strong ligand-dependent modulation of µ receptor activity by the membrane potential, suggesting preferential activity of morphine in neurons with high neuronal activity.


Subject(s)
Locus Coeruleus , Receptors, Opioid, mu , Animals , Enkephalin, Ala(2)-MePhe(4)-Gly(5)- , HEK293 Cells , Humans , Ligands , Locus Coeruleus/metabolism , Morphine/pharmacology , Rats , Receptors, Opioid, mu/metabolism
5.
Br J Pharmacol ; 175(14): 2715-2716, 2018 07.
Article in English | MEDLINE | ID: mdl-29939423

ABSTRACT

This themed section of the British Journal of Pharmacology stems from an International Narcotics Research Conference (INRC) meeting held in July 2016 at The Assembly Rooms in Bath, UK. LINKED ARTICLES: This article is part of a themed section on Emerging Areas of Opioid Pharmacology. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.14/issuetoc.


Subject(s)
Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , Animals , Humans , Immune System/drug effects , Opioid-Related Disorders/drug therapy , Pain/drug therapy , Pain/metabolism , Receptors, Opioid/metabolism
6.
Mol Pharmacol ; 88(2): 347-56, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26013542

ABSTRACT

There is ongoing debate about the role of G protein-coupled receptor kinases (GRKs) in agonist-induced desensitization of the µ-opioid receptor (MOPr) in brain neurons. In the present paper, we have used a novel membrane-permeable, small-molecule inhibitor of GRK2 and GRK3, Takeda compound 101 (Cmpd101; 3-[[[4-methyl-5-(4-pyridyl)-4H-1,2,4-triazole-3-yl] methyl] amino]-N-[2-(trifuoromethyl) benzyl] benzamidehydrochloride), to study the involvement of GRK2/3 in acute agonist-induced MOPr desensitization. We observed that Cmpd101 inhibits the desensitization of the G protein-activated inwardly-rectifying potassium current evoked by receptor-saturating concentrations of methionine-enkephalin (Met-Enk), [d-Ala(2), N-MePhe(4), Gly-ol(5)]-enkephalin (DAMGO), endomorphin-2, and morphine in rat and mouse locus coeruleus (LC) neurons. In LC neurons from GRK3 knockout mice, Met-Enk-induced desensitization was unaffected, implying a role for GRK2 in MOPr desensitization. Quantitative analysis of the loss of functional MOPrs following acute agonist exposure revealed that Cmpd101 only partially reversed MOPr desensitization. Inhibition of extracellular signal-regulated kinase 1/2, protein kinase C, c-Jun N-terminal kinase, or GRK5 did not inhibit the Cmpd101-insensitive component of desensitization. In HEK 293 cells, Cmpd101 produced almost complete inhibition of DAMGO-induced MOPr phosphorylation at Ser(375), arrestin translocation, and MOPr internalization. Our data demonstrate a role for GRK2 (and potentially also GRK3) in agonist-induced MOPr desensitization in the LC, but leave open the possibility that another, as yet unidentified, mechanism of desensitization also exists.


Subject(s)
Benzamides/pharmacology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinase 3/metabolism , Locus Coeruleus/drug effects , Receptors, Opioid, mu/metabolism , Animals , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar
7.
Expert Opin Drug Discov ; 9(11): 1333-44, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25253272

ABSTRACT

INTRODUCTION: Psychostimulant and opioid addiction are poorly treated. The majority of abstinent users relapse back to drug-taking within a year of abstinence, making 'anti-relapse' therapies the focus of much current research. There are two fundamental challenges to developing novel treatments for drug addiction. First, there are three key stimuli that precipitate relapse back to drug-taking: stress, presentation of drug-conditioned cue, taking a small dose of drug. The most successful novel treatment would be effective against all three stimuli. Second, a large number of drug users are poly-drug users: taking more than one drug of abuse at a time. The ideal anti-addiction treatment would, therefore, be effective against all classes of drugs of abuse. AREAS COVERED: In this review, the authors discuss the clinical need and animal models used to uncover potential novel treatments. There is a very broad range of potential treatment approaches and targets currently being examined as potential anti-relapse therapies. These broadly fit into two categories: 'memory-based' and 'receptor-based' and the authors discuss the key targets here within. EXPERT OPINION: Opioid receptors and ligands have been widely studied, and research into how different opioid subtypes affect behaviours related to addiction (reward, dysphoria, motivation) suggests that they are tractable targets as anti-relapse treatments. Regarding opioid ligands as novel 'anti-relapse' medication targets, research suggests that a 'non-selective' approach to targeting opioid receptors will be the most effective.


Subject(s)
Receptors, Opioid/metabolism , Substance-Related Disorders/drug therapy , Analgesics, Opioid , Animals , Central Nervous System Stimulants , Disease Models, Animal , Humans , Recurrence , Substance-Related Disorders/metabolism
8.
Mol Pharmacol ; 84(2): 252-60, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23716621

ABSTRACT

Consumption of ethanol is a considerable risk factor for death in heroin overdose. We sought to determine whether a mildly intoxicating concentration of ethanol could alter morphine tolerance at the cellular level. In rat locus coeruleus (LC) neurons, tolerance to morphine was reversed by acute exposure of the brain slice to ethanol (20 mM). Tolerance to the opioid peptide [d-Ala(2),N-MePhe(4),Gly-ol]-enkephalin was not reversed by ethanol. Previous studies in LC neurons have revealed a role for protein kinase C (PKC)α in µ-opioid receptor (MOPr) desensitization by morphine and in the induction and maintenance of morphine tolerance, but we have been unable to demonstrate that 20 mM ethanol produces significant inhibition of PKCα. The ability of ethanol to reverse cellular tolerance to morphine in LC neurons was absent in the presence of the phosphatase inhibitor okadaic acid, indicating that dephosphorylation is involved. In human embryonic kidney 293 cells expressing the MOPr, ethanol reduced the level of MOPr phosphorylation induced by morphine. Ethanol reversal of tolerance did not appear to result from a direct effect on MOPr since acute exposure to ethanol (20 mM) did not modify the affinity of binding of morphine to the MOPr or the efficacy of morphine for G-protein activation as measured by guanosine 5'-O-(3-[(35)S]thio)triphosphate binding. Similarly, ethanol did not affect MOPr trafficking. We conclude that acute exposure to ethanol enhances the effects of morphine by reversing the processes underlying morphine cellular tolerance.


Subject(s)
Ethanol/pharmacology , Locus Coeruleus/drug effects , Morphine/pharmacology , Neurons/drug effects , Animals , Brain/drug effects , Brain/metabolism , Cell Line , Drug Interactions , Drug Tolerance , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Locus Coeruleus/metabolism , Male , Neurons/metabolism , Opioid Peptides/metabolism , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation/drug effects , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/metabolism , Rats , Rats, Wistar , Receptors, Opioid, mu/metabolism
9.
Eur J Neurosci ; 36(12): 3636-42, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23002724

ABSTRACT

There is considerable controversy over whether µ-opioid receptor (MOPr) desensitization is homologous or heterologous and over the mechanisms underlying such desensitization. In different cell types MOPr desensitization has been reported to involve receptor phosphorylation by various kinases, including G-protein-coupled receptor kinases (GRKs), second messenger and other kinases as well as perturbation of the MOPr effector pathway by GRK sequestration of G protein ßγ subunits or ion channel modulation. Here we report that in brainstem locus coeruleus (LC) neurons prepared from relatively mature rats (5-8 weeks old) rapid MOPr desensitization induced by the high-efficacy opioid peptides methionine enkephalin and DAMGO was homologous and not heterologous to α(2)-adrenoceptors and somatostatin SST(2) receptors. Given that these receptors all couple through G proteins to the same set of G-protein inwardly rectifying (GIRK) channels it is unlikely therefore that in mature neurons MOPr desensitization involves G protein ßγ subunit sequestration or ion channel modulation. In contrast, in slices from immature animals (less than postnatal day 20), MOPr desensitization was observed to be heterologous and could be downstream of the receptor. Heterologous MOPr desensitization was not dependent on protein kinase C or c-Jun N-terminal kinase activity, but the change from heterologous to homologous desensitization with age was correlated with a decrease in the expression levels of GRK2 in the LC and other brain regions. The observation that the mechanisms underlying MOPr desensitization change with neuronal development is important when extrapolating to the mature brain results obtained from experiments on expression systems, cell lines and immature neuronal preparations.


Subject(s)
Action Potentials , Receptors, Opioid, mu/metabolism , Animals , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Enkephalin, Methionine/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 2/metabolism , GTP-Binding Proteins/metabolism , Gene Expression , Locus Coeruleus/growth & development , Locus Coeruleus/physiology , Male , Mitogen-Activated Protein Kinase 8/metabolism , Neurons/metabolism , Neurons/physiology , Neurotransmitter Agents/pharmacology , Protein Kinase C/metabolism , Protein Subunits/metabolism , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Somatostatin/metabolism
10.
Mol Pharmacol ; 78(4): 756-66, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20647394

ABSTRACT

We have compared the ability of a number of µ-opioid receptor (MOPr) ligands to activate G proteins with their abilities to induce MOPr phosphorylation, to promote association of arrestin-3 and to cause MOPr internalization. For a model of G protein-coupled receptor (GPCR) activation where all agonists stabilize a single active conformation of the receptor, a close correlation between signaling outputs might be expected. Our results show that overall there is a very good correlation between efficacy for G protein activation and arrestin-3 recruitment, whereas a few agonists, in particular endomorphins 1 and 2, display apparent bias toward arrestin recruitment. The agonist-induced phosphorylation of MOPr at Ser(375), considered a key step in MOPr regulation, and agonist-induced internalization of MOPr were each found to correlate well with arrestin-3 recruitment. These data indicate that for the majority of MOPr agonists the ability to induce receptor phosphorylation, arrestin-3 recruitment, and internalization can be predicted from their ability as agonists to activate G proteins. For the prototypic MOPr agonist morphine, its relatively weak ability to induce MOPr internalization can be explained by its low agonist efficacy.


Subject(s)
Analgesics, Opioid/chemistry , Analgesics, Opioid/metabolism , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Signal Transduction/physiology , Animals , Cattle , Cell Line , Fluorescence Resonance Energy Transfer , Humans , Protein Binding/drug effects , Protein Binding/physiology , Protein Transport/drug effects , Protein Transport/physiology , Rats , Signal Transduction/drug effects
11.
Brain Res ; 1217: 70-7, 2008 Jun 27.
Article in English | MEDLINE | ID: mdl-18501877

ABSTRACT

We previously demonstrated that intracerebroventricular (i.c.v.) administration of protein kinase C (PKC) or protein kinase A (PKA) inhibitors reversed morphine antinociceptive tolerance in 3-day morphine-pelleted mice. The present study aimed at evaluating whether pre-treating mice with a PKC or PKA inhibitor prior to pellet implantation would prevent the development of morphine tolerance and physical dependence. Antinociception was assessed using the warm-water tail immersion test and physical dependence was evaluated by quantifying/scoring naloxone-precipitated withdrawal signs. While drug-naïve mice pelleted with a 75 mg morphine pellet for 3 days developed a 5.8-fold tolerance to morphine antinociception, mice pre-treated i.c.v. with the PKC inhibitors bisindolylmaleimide I, Go-7874 or Go-6976, or with the myristoylated PKA inhibitor, PKI-(14-22)-amide failed to develop any tolerance to morphine antinociception. Experiments were also conducted to determine whether morphine-pelleted mice were physically dependent when pre-treated with PKC or PKA inhibitors. The same inhibitor doses that prevented morphine tolerance were evaluated in other mice injected s.c. with naloxone and tested for precipitated withdrawal. The pre-treatment with PKC or PKA inhibitors failed to attenuate or block the signs of morphine withdrawal including jumping, wet-dog shakes, rearing, forepaw tremor, increased locomotion, grooming, diarrhea, tachypnea and ptosis. These data suggest that elevations in the activity of PKC and PKA in the brain are critical to the development of morphine tolerance. However, it appears that tolerance can be dissociated from physical dependence, indicating a role for PKC and PKA to affect antinociception but not those signs mediated through the complex physiological processes of withdrawal.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Drug Tolerance/physiology , Enzyme Inhibitors/administration & dosage , Morphine Dependence/enzymology , Protein Kinase C/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/drug effects , Injections, Intraventricular , Male , Mice , Morphine/pharmacology , Pain Threshold/drug effects , Protein Kinase C/drug effects , Substance Withdrawal Syndrome/enzymology
12.
Neuroreport ; 19(5): 589-93, 2008 Mar 26.
Article in English | MEDLINE | ID: mdl-18388744

ABSTRACT

There is confusion in the literature concerning the relative agonist efficacy of methadone at micro-opioid receptors (MOPrs). Here, we confirm that methadone is a full agonist in guanosine 5'-O-[gamma-thio]triphosphate (GTPgammaS) binding studies. Methadone, however, seems to have low efficacy in studies of MOPr activation of G-protein-gated potassium (GIRK) channels, but this is because it directly inhibits the GIRK channels. Methadone also inhibits alpha2-adrenoceptor-activated GIRK channels. Methadone is not a specific GIRK channel blocker. It also inhibits small conductance Ca2+-activated K+ (SK2) channels. We conclude that methadone is a full agonist at MOPrs that, as we and others have shown, induces MOPr desensitization and internalization.


Subject(s)
Methadone/pharmacology , Narcotics/pharmacology , Neurons/drug effects , Receptors, Opioid, mu/drug effects , Animals , Animals, Newborn , Cell Line, Transformed , Drug Interactions , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , In Vitro Techniques , Locus Coeruleus/cytology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Morphine/pharmacology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Neurons/physiology , Neurons/radiation effects , Protein Binding/drug effects , Rats , Transfection
13.
J Neurochem ; 104(4): 937-45, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18005002

ABSTRACT

The distribution of the mRNA of different C-terminal splice variants of the mu-opioid receptor in rat CNS was assessed by RT-PCR. The mRNA species for MOR1, MOR1A and MOR1B were readily detectable and distributed widely throughout the rat CNS, with levels of MOR1 and MOR1A mRNA being overall greater than for MOR1B. We did not find convincing evidence that significant levels of MOR1C, MOR1C1, MOR1C2 and MOR1D are present in rat CNS. To examine possible differences in the agonist-induced regulation of MOR1, MOR1A and MOR1B, we expressed these constructs in HEK293 cells along with G-protein-coupled inwardly rectifying K+ channel subunits and measured the rate and extent of desensitisation of (d-Ala2,N-Me-Phe4,glycinol5)-enkephalin (DAMGO)- and morphine-induced G-protein-coupled inwardly rectifying K+ currents. Morphine-induced desensitisation was rapid for all three splice variants (t1/2: 1.2-1.7 min) but DAMGO-induced desensitisation was significantly slower for MOR1B (t1/2 4.2 min). Inhibition of endocytosis by expression of a dynamin-dominant negative mutant increased the rate of DAMGO-induced desensitisation of MOR1B. These data show that some splice variants of mu-opioid receptor are widely expressed in rat CNS but question the existence of others that have been reported in the literature. In addition, whereas the rate of desensitisation of MOR1 and MOR1A is agonist-independent, that for MOR1B is agonist-dependent.


Subject(s)
Peptide Fragments/analysis , Peptide Fragments/physiology , Protein Isoforms/analysis , Protein Isoforms/physiology , Receptors, Opioid, mu/analysis , Receptors, Opioid, mu/physiology , Amino Acid Sequence , Analgesics, Opioid/pharmacology , Animals , Cell Line , Central Nervous System/chemistry , Central Nervous System/drug effects , Central Nervous System/physiology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Humans , Male , Molecular Sequence Data , Peptide Fragments/agonists , Protein Isoforms/agonists , Rats , Rats, Wistar , Receptors, Opioid, mu/agonists
14.
Brain Res ; 1159: 86-93, 2007 Jul 23.
Article in English | MEDLINE | ID: mdl-17582387

ABSTRACT

Acute morphine antinociception has been shown to be blocked by very low picogram doses of okadaic acid indicating that inhibition of protein phosphatase PP2A allows for increases in phosphorylation to inhibit antinociception. Comparative studies in morphine tolerant animals have not been reported. In the present study, we showed a significant increase in the total phosphatase activity in the periaqueductal gray matter (PAG) from morphine-pelleted versus placebo-pelleted mice, 72-h after pellet implantation. This supports our hypothesis that phosphatase activity is increased in tolerance as a compensatory mechanism for the increase in kinase activity during the development of tolerance. We also demonstrated that i.c.v. administration of the phosphatase inhibitor okadaic acid (3 pmol/mouse; a dose tested to be inert in placebo-pelleted mice) enhanced the level of morphine antinociceptive tolerance assessed by the tail immersion test, 72-h following pellet implantation. This was supported by the fact that the same treatment with okadaic acid blocked the increase in phosphatase activity in PAG of morphine tolerant mice indicating that selective inhibition of PP2A contributes to enhanced levels of morphine tolerance. We have previously reported that PKC or PKA inhibitors reversed morphine antinociceptive tolerance in mice. The current study shows that i.c.v. administration of the PKC inhibitors bisindolylmaleimide I or Go6976 reversed the enhanced level of morphine tolerance induced by okadaic acid treatment to the same level of tolerance observed in non-okadaic acid-treated tolerant mice. However, the PKA inhibitor PKI-(14-22)-amide only partially reversed the enhancement of morphine tolerance induced by okadaic acid. Our data suggest an important role for the balance between kinases and phosphatases in modulating tolerance levels. Further studies will be directed towards a better understanding of the role of different phosphatase isoforms in morphine tolerance.


Subject(s)
Behavior, Animal/drug effects , Drug Tolerance/physiology , Morphine/pharmacology , Narcotics/pharmacology , Phosphoprotein Phosphatases/physiology , Analysis of Variance , Animals , Drug Interactions , Enzyme Inhibitors/administration & dosage , Injections, Intraventricular , Male , Mice , Morphine/administration & dosage , Narcotics/administration & dosage , Pain Measurement , Periaqueductal Gray/drug effects , Periaqueductal Gray/metabolism , Reaction Time/drug effects
15.
Trends Pharmacol Sci ; 27(11): 558-65, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17000011

ABSTRACT

The repeated administration of opiate drugs such as morphine results in the development of tolerance to their analgesic, rewarding (euphoric) and respiratory-depressant effects; thus, to obtain the same level of response with subsequent administrations, a greater dose must be used. Tolerance can limit the clinical efficacy of opiate drugs and enhance the social problems that are inherent in recreational opioid abuse. Surprisingly, the mechanism (or mechanisms) underlying the development of morphine tolerance remains controversial. Here, we propose that protein kinase C could have a crucial role in the desensitization of mu-opioid receptors by morphine and that this cellular process could contribute to the development and maintenance of morphine tolerance in vivo.


Subject(s)
Analgesics, Opioid , Drug Tolerance , Morphine/pharmacology , Protein Kinase C/metabolism , Receptors, Opioid, mu/agonists , Animals , Humans , Isoenzymes/metabolism , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Receptors, Opioid, mu/metabolism
16.
Mol Pharmacol ; 70(2): 676-85, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16682505

ABSTRACT

The ability of two opioid agonists, [d-Ala(2),N-Me-Phe(4),Gly(5)-ol]-enkephalin (DAMGO) and morphine, to induce mu-opioid receptor (MOR) phosphorylation, desensitization, and internalization was examined in human embryonic kidney (HEK) 293 cells expressing rat MOR1 as well G protein-coupled inwardly rectifying potassium channel (GIRK) channel subunits. Both DAMGO and morphine activated GIRK currents, but the maximum response to DAMGO was greater than that of morphine, indicating that morphine is a partial agonist. The responses to DAMGO and morphine desensitized rapidly in the presence of either drug. Expression of a dominant negative mutant G protein-coupled receptor kinase 2 (GRK2), GRK2-K220R, markedly attenuated the DAMGO-induced desensitization of MOR1, but it had no effect on morphine-induced MOR1 desensitization. In contrast, inhibition of protein kinase C (PKC) either by the PKC inhibitory peptide PKC (19-31) or staurosporine reduced MOR1 desensitization by morphine but not that induced by DAMGO. Morphine and DAMGO enhanced MOR1 phosphorylation over basal. The PKC inhibitor bisindolylmaleimide 1 (GF109203X) inhibited MOR1 phosphorylation under basal conditions and in the presence of morphine, but it did not inhibit DAMGO-induced phosphorylation. DAMGO induced arrestin-2 translocation to the plasma membrane and considerable MOR1 internalization, whereas morphine did not induce arrestin-2 translocation and induced very little MOR1 internalization. Thus, DAMGO and morphine each induce desensitization of MOR1 signaling in HEK293 cells but by different molecular mechanisms; DAMGO-induced desensitization is GRK2-dependent, whereas morphine-induced desensitization is in part PKC-dependent. MORs desensitized by DAMGO activation are then readily internalized by an arrestin-dependent mechanism, whereas those desensitized by morphine are not. These data suggest that opioid agonists induce different conformations of the MOR that are susceptible to different desensitizing and internalization processes.


Subject(s)
Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Receptors, Opioid, mu/agonists , Arrestin/metabolism , Cell Line , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , Humans , Morphine/pharmacology , Phosphorylation , Protein Kinase C/physiology , Protein Transport/drug effects , Receptors, Opioid, mu/chemistry
17.
Curr Opin Pharmacol ; 5(1): 60-8, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15661627

ABSTRACT

Morphine and other opioids are used and abused for their analgesic and rewarding properties. Tolerance to these effects develops over hours/days to weeks, as can physical and psychological dependence. Despite much investigation, the precise cellular mechanisms underlying opioid tolerance and dependence remain elusive. Recent studies examining mu-opioid receptor desensitization and trafficking have revealed several potential mechanisms for acute receptor regulation. Other studies have reported changes in many other proteins that develop during chronic opioid treatment or withdrawal and such changes may be partly responsible for the cellular and synaptic adaptations to prolonged opioid exposure. While these studies have added to our knowledge of the cellular processes participating in opioid tolerance and dependence, the challenge remains to integrate these observations into a coherent explanation of the complex changes observed in whole animals chronically exposed to opioids.


Subject(s)
Molecular Biology/trends , Morphine/pharmacology , Opioid-Related Disorders/metabolism , Receptors, Opioid, mu/drug effects , Substance Withdrawal Syndrome/metabolism , Synaptic Transmission/drug effects , Animals , Drug Tolerance/physiology , Humans , Morphine/metabolism , Receptors, Opioid, mu/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...