Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
STAR Protoc ; 4(2): 102231, 2023 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-37104091

RESUMO

This protocol describes endogenous labeling of opioid receptors (ORs) using a ligand-directed reagent, naltrexamine-acylimidazole compounds (NAI-X). NAI acts by guiding and permanently tagging a small-molecule reporter (X)-such as fluorophores or biotin-to ORs. Here we detail syntheses and uses of NAI-X for OR visualization and functional studies. The NAI-X compounds overcome long-standing challenges in mapping and tracking endogenous ORs as the labeling can be done in situ with live tissues or cultured cells. For complete details on the use and execution of this protocol, please refer to Arttamangkul et al.1,2.

2.
Elife ; 102021 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-34477106

RESUMO

Class A G-protein-coupled receptors (GPCRs) normally function as monomers, although evidence from heterologous expression systems suggests that they may sometimes form homodimers and/or heterodimers. This study aims to evaluate possible functional interplay of endogenous µ- and δ-opioid receptors (MORs and DORs) in mouse neurons. Detecting GPCR dimers in native tissues, however, has been challenging. Previously, MORs and DORs co-expressed in transfected cells have been reported to form heterodimers, and their possible co-localization in neurons has been studied in knock-in mice expressing genetically engineered receptors fused to fluorescent proteins. Here, we find that single cholinergic neurons in the mouse striatum endogenously express both MORs and DORs. The receptors on neurons from live brain slices were fluorescently labeled with a ligand-directed labeling reagent, NAI-A594. The selective activation of MORs and DORs, with DAMGO (µ-agonist) and deltorphin (δ-agonist) inhibited spontaneous firing in all cells examined. In the continued presence of agonist, the firing rate returned to baseline as the result of receptor desensitization with the application of deltorphin but was less observed with the application of DAMGO. In addition, agonist-induced internalization of DORs but not MORs was detected. When MORs and DORs were activated simultaneously with [Met5]-enkephalin, desensitization of MORs was facilitated but internalization was not increased. Together, these results indicate that while MORs and DORs are expressed in single striatal cholinergic interneurons, the two receptors function independently.


Assuntos
Neurônios Colinérgicos/metabolismo , Corpo Estriado/metabolismo , Interneurônios/fisiologia , Receptores Opioides delta/genética , Receptores Opioides mu/genética , Animais , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo
3.
Mol Pharmacol ; 98(4): 497-507, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32362586

RESUMO

Based on studies using mutations of the µ-opioid receptor (MOR), phosphorylation of multiple sites on the C-terminus has been recognized as a critical step underlying acute desensitization and the development of cellular tolerance. The aim of this study is to explore which kinases mediate desensitization of MOR in brain slices from drug-naïve and morphine-treated animals. Whole-cell recordings from locus coeruleus neurons were made, and the agonist-induced increase in potassium conductance was measured. In slices from naïve animals, pharmacological inhibition of G-protein receptor kinase (GRK2/3) with compound 101 blocked acute desensitization. Following chronic treatment with morphine, compound 101 was less effective at blocking acute desensitization. Compound 101 blocked receptor internalization in tissue from both naïve and morphine-treated animals, suggesting that GRK2/3 remained active. Kinase inhibitors aimed at blocking protein kinase C and c-Jun N-terminal kinase had no effect on desensitization in tissue taken from naïve animals. However, in slices taken from morphine-treated animals, the combination of these blockers along with compound 101 was required to block acute desensitization. Acute desensitization of the potassium conductance induced by the somatostatin receptor was also blocked by compound 101 in slices from naïve but not morphine-treated animals. As was observed with MOR, it was necessary to use the combination of kinase inhibitors to block desensitization of the somatostatin receptor in slices from morphine-treated animals. The results show that chronic treatment with morphine results in a surprising and heterologous adaptation in kinase-dependent desensitization. SIGNIFICANCE STATEMENT: The results show that chronic treatment with morphine induced heterologous adaptations in kinase regulation of G protein coupled receptor (GPCR) desensitization. Although the canonical mechanism for acute desensitization through phosphorylation by G protein-coupled receptor kinase is supported in tissue taken from naïve animals, following chronic treatment with morphine, the acute kinase-dependent desensitization of GPCRs is disrupted such that additional kinases, including protein kinase C and c-Jun N-terminal kinase, contribute to desensitization.


Assuntos
Locus Cerúleo/metabolismo , Morfina/administração & dosagem , Receptores Opioides mu/metabolismo , Quinases de Receptores Adrenérgicos beta/metabolismo , Animais , Tolerância a Medicamentos , Feminino , Locus Cerúleo/efeitos dos fármacos , Masculino , Morfina/farmacologia , Técnicas de Patch-Clamp , Fosforilação , Potássio/metabolismo , Ratos
4.
Neuron ; 105(4): 663-677.e8, 2020 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-31837915

RESUMO

A major function of GPCRs is to inhibit presynaptic neurotransmitter release, requiring ligand-activated receptors to couple locally to effectors at terminals. The current understanding of how this is achieved is through receptor immobilization on the terminal surface. Here, we show that opioid peptide receptors, GPCRs that mediate highly sensitive presynaptic inhibition, are instead dynamic in axons. Opioid receptors diffuse rapidly throughout the axon surface and internalize after ligand-induced activation specifically at presynaptic terminals. We delineate a parallel regulated endocytic cycle for GPCRs operating at the presynapse, separately from the synaptic vesicle cycle, which clears activated receptors from the surface of terminals and locally reinserts them to maintain the diffusible surface pool. We propose an alternate strategy for achieving local control of presynaptic effectors that, opposite to using receptor immobilization and enforced proximity, is based on lateral mobility of receptors and leverages the inherent allostery of GPCR-effector coupling.


Assuntos
Endocitose/fisiologia , Terminações Pré-Sinápticas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Vesículas Sinápticas/metabolismo , Analgésicos Opioides/farmacologia , Animais , Células Cultivadas , Endocitose/efeitos dos fármacos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Terminações Pré-Sinápticas/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Receptores de Neurotransmissores/agonistas , Receptores de Neurotransmissores/metabolismo , Vesículas Sinápticas/efeitos dos fármacos
5.
Elife ; 82019 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-31589142

RESUMO

Identifying neurons that have functional opioid receptors is fundamental for the understanding of the cellular, synaptic and systems actions of opioids. Current techniques are limited to post hoc analyses of fixed tissues. Here we developed a fluorescent probe, naltrexamine-acylimidazole (NAI), to label opioid receptors based on a chemical approach termed 'traceless affinity labeling'. In this approach, a high affinity antagonist naltrexamine is used as the guide molecule for a transferring reaction of acylimidazole at the receptor. This reaction generates a fluorescent dye covalently linked to the receptor while naltrexamine is liberated and leaves the binding site. The labeling induced by this reagent allowed visualization of opioid-sensitive neurons in rat and mouse brains without loss of function of the fluorescently labeled receptors. The ability to locate endogenous receptors in living tissues will aid considerably in establishing the distribution and physiological role of opioid receptors in the CNS of wild type animals.


Assuntos
Química Encefálica , Neurônios/química , Receptores Opioides/análise , Coloração e Rotulagem/métodos , Animais , Corantes Fluorescentes/síntese química , Corantes Fluorescentes/metabolismo , Fluorometria/métodos , Camundongos Endogâmicos C57BL , Ratos Sprague-Dawley
6.
Mol Pharmacol ; 96(4): 505-514, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31383769

RESUMO

Phosphorylation of sites on the C terminus of the µ-opioid receptor (MOR) results in the induction of acute desensitization that is thought to be a precursor for the development of long-term tolerance. Alanine mutations of all 11 phosphorylation sites on the C terminus of MORs almost completely abolished desensitization and one measure of tolerance in locus coeruleus neurons when these phosphorylation-deficient MORs were virally expressed in MOR knockout rats. In the present work, we identified specific residues that underlie acute desensitization, receptor internalization, and tolerance and examined four MOR variants with different alanine or glutamate mutations in the C terminus. Alanine mutations in the sequence between amino acids 375 and 379 (STANT-3A) and the sequence between amino acids 363 and 394 having four additional alanine substitutions (STANT + 7A) reduced desensitization and two measures of long-term tolerance. After chronic morphine treatment, alanine mutations in the sequence between 354 and 357 (TSST-4A) blocked one measure of long-term tolerance (increased acute desensitization and slowed recovery from desensitization) but did not change a second (decreased sensitivity to morphine). With the expression of receptors having glutamate substitutions in the TSST sequence (TSST-4E), an increase in acute desensitization was present after chronic morphine treatment, but the sensitivity to morphine was not changed. The results show that all 11 phosphorylation sites contribute, in varying degrees, to acute desensitization and long-term tolerance. That acute desensitization and tolerance are not necessarily linked illustrates the complexity of events that are triggered by chronic treatment with morphine. SIGNIFICANCE STATEMENT: In this work, we showed that the degree of phosphorylation on the C terminus of the µ-opioid receptor alters acute desensitization and internalization, and in measures of long-term tolerance to morphine. The primary conclusion is that the degree of phosphorylation on the 11 possible sites of the C terminus has different roles for expression of the multiple adaptive mechanisms that follow acute and long-term agonist activation. Although the idea that acute desensitization and tolerance are intimately linked is generally supported, these results indicate that disruption of one phosphorylation cassette of the C terminus TSST (354-357) distinguishes the two processes.


Assuntos
Encefalina Metionina/farmacologia , Mutação , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Potenciais de Ação/efeitos dos fármacos , Alanina/metabolismo , Animais , Tolerância a Medicamentos , Feminino , Técnicas de Inativação de Genes , Ácido Glutâmico/metabolismo , Masculino , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/genética
7.
Elife ; 72018 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-29589831

RESUMO

Phosphorylation of the µ-opioid receptor (MOR) is known as a key step in desensitization and internalization but the role in the development of long-term tolerance at the cellular level is not known. Viral expression of wild type (exWT) and mutant MORs, where all phosphorylation sites on the C-terminus (Total Phosphorylation Deficient (TPD)) were mutated to alanine, were examined in locus coeruleus neurons in a MOR knockout rat. Both receptors activated potassium conductance similar to endogenous receptors in wild type animals. The exWT receptors, like endogenous receptors, acutely desensitized, internalized and, after chronic morphine treatment, displayed signs of tolerance. However, TPD receptors did not desensitize or internalize with agonist treatment. In addition the TPD receptors did not develop cellular tolerance following chronic morphine treatment. Thus C-terminal phosphorylation is necessary for the expression of acute desensitization, trafficking and one sign of long-term tolerance to morphine at the cellular level.


Assuntos
Processamento de Proteína Pós-Traducional , Receptores Opioides mu/metabolismo , Animais , Animais Geneticamente Modificados , Técnicas de Inativação de Genes , Locus Cerúleo/citologia , Morfina/metabolismo , Neurônios/metabolismo , Fosforilação , Potássio/metabolismo , Ratos
8.
Mol Pharmacol ; 88(4): 816-24, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25934731

RESUMO

Sustained activation of G protein-coupled receptors can lead to a rapid decline in signaling through acute receptor desensitization. In the case of the µ-opioid receptor (MOPr), this desensitization may play a role in the development of analgesic tolerance. It is understood that phosphorylation of MOPr promotes association with ß-arrestin proteins, which then facilitates desensitization and receptor internalization. Agonists that induce acute desensitization have been shown to induce a noncanonical high-affinity agonist binding state in MOPr, conferring a persistent memory of prior receptor activation. In the current study, live-cell confocal imaging was used to investigate the role of receptor phosphorylation in agonist binding to MOPr. A phosphorylation cluster in the C-terminal tail of MOPr was identified as a mediator of agonist-induced affinity changes in MOPr. This site is unique from the primary phosphorylation cluster responsible for ß-arrestin binding and internalization. Electrophysiologic measurements of receptor function suggest that both phosphorylation clusters may play a parallel role during acute receptor desensitization. Desensitization was unaffected by alanine mutation of either phosphorylation cluster, but was largely eliminated when both clusters were mutated. Overall, this work suggests that there are multiple effects of MOPr phosphorylation that appear to regulate MOPr function: one affecting ß-arrestin binding and a second affecting agonist binding.


Assuntos
Analgésicos Opioides/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Analgésicos Opioides/química , Analgésicos Opioides/farmacologia , Animais , Arrestinas/química , Arrestinas/metabolismo , Arrestinas/farmacologia , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Knockout , Técnicas de Cultura de Órgãos , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , beta-Arrestinas
9.
Br J Pharmacol ; 172(2): 583-92, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24697621

RESUMO

BACKGROUND AND PURPOSE: This study examined the role of agents known to activate PKC on morphine-induced desensitization of µ-opioid receptors (MOP receptors) in brain slices containing locus coeruleus neurons. EXPERIMENTAL APPROACH: Intracellular recordings were obtained from rat locus coeruleus neurons. Two measurements were used to characterize desensitization, the decline in hyperpolarization induced by application of a saturating concentration of agonist (acute desensitization) and the decrease in hyperpolarization induced by a subsaturating concentration of [Met](5) enkephalin (ME) following washout of the saturating concentration (sustained desensitization). Internalization of MOP receptors was studied in brain slices prepared from transgenic mice expressing Flag-MOP receptors. The subcellular distribution of activated PKC was examined using a novel fluorescent sensor of PKC in HEK293 cells. KEY RESULTS: The phorbol esters (PMA and PDBu) and muscarine increased acute desensitization induced by a saturating concentration of morphine and ME. These effects were not sensitive to staurosporine. Staurosporine did not block the decline in hyperpolarization induced by muscarine. PDBu and muscarine did not affect sustained desensitization induced by ME nor did phorbol esters or muscarine change the trafficking of MOP receptors induced by morphine or ME. The distribution of activated PKC measured in HEK293 cells differed depending on which phorbol ester was applied. CONCLUSIONS AND IMPLICATIONS: This study demonstrates a distinct difference in two measurements that are often used to evaluate desensitization. The measure of decline correlated well with the reduction in peak amplitudes caused by PKC activators implicating the modification of other factors rather than MOP receptors. LINKED ARTICLES: This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.


Assuntos
Proteína Quinase C/fisiologia , Receptores Opioides mu/fisiologia , Analgésicos Opioides/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encefalina Metionina/farmacologia , Células HEK293 , Humanos , Masculino , Camundongos Transgênicos , Morfina/farmacologia , Muscarina/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Dibutirato de 12,13-Forbol/farmacologia , Proteína Quinase C/antagonistas & inibidores , Transporte Proteico , Ratos Sprague-Dawley , Receptores Opioides mu/genética , Acetato de Tetradecanoilforbol/farmacologia
10.
J Neurosci ; 33(9): 4118-27, 2013 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-23447620

RESUMO

Prolonged exposure to high-efficacy agonists results in desensitization of the µ-opioid receptor (MOR). Desensitized receptors are thought to be unable to couple to G-proteins, preventing downstream signaling; however, the changes to the receptor itself are not well characterized. In the current study, confocal imaging was used to determine whether desensitizing conditions cause a change in agonist-receptor interactions. Using rapid solution exchange, the binding kinetics of fluorescently labeled opioid agonist, dermorphin Alexa594 (derm A594), to MORs was measured in live cells. The affinity of derm A594 binding increased after prolonged treatment of cells with multiple agonists that are known to cause receptor desensitization. In contrast, binding of a fluorescent antagonist, naltrexamine Alexa594, was unaffected by similar agonist pretreatment. The increased affinity of derm A594 for the receptor was long-lived and partially reversed after a 45 min wash. Treatment of the cells with pertussis toxin did not alter the increase in affinity of the derm A594 for MOR. Likewise, the affinity of derm A594 for MORs expressed in mouse embryonic fibroblasts derived from arrestin 1 and 2 knock-out animals increased after treatment of the cells with the desensitization protocol. Thus, opioid receptors were "imprinted" with a memory of prior agonist exposure that was independent of G-protein activation or arrestin binding that altered subsequent agonist-receptor interactions. The increased affinity suggests that acute desensitization results in a long-lasting but reversible conformational change in the receptor.


Assuntos
Membrana Celular/metabolismo , Fenômenos Farmacológicos/efeitos dos fármacos , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Analgésicos Opioides/farmacocinética , Análise de Variância , Animais , Arrestina/deficiência , Arrestina/metabolismo , Membrana Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacocinética , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Expressão Gênica/efeitos dos fármacos , Guanosina 5'-O-(3-Tiotrifosfato)/farmacologia , Células HEK293 , Humanos , Ligantes , Camundongos , Camundongos Knockout , Morfina/farmacologia , Naltrexona/análogos & derivados , Naltrexona/farmacocinética , Antagonistas de Entorpecentes/farmacocinética , Peptídeos Opioides/farmacocinética , Compostos Orgânicos/farmacocinética , Toxina Pertussis/farmacologia , Ligação Proteica/efeitos dos fármacos , Conformação Proteica/efeitos dos fármacos , Ensaio Radioligante , Receptores Opioides mu/genética , Especificidade por Substrato/efeitos dos fármacos , Fatores de Tempo , Transfecção , Trítio/farmacocinética
11.
J Neurophysiol ; 108(9): 2393-404, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22956800

RESUMO

Respiratory depression is a therapy-limiting side effect of opioid analgesics, yet our understanding of the brain circuits mediating this potentially lethal outcome remains incomplete. Here we studied the contribution of the rostral ventromedial medulla (RVM), a region long implicated in pain modulation and homeostatic regulation, to opioid-induced respiratory depression. Microinjection of the µ-opioid agonist DAMGO in the RVM of lightly anesthetized rats produced both analgesia and respiratory depression, showing that neurons in this region can modulate breathing. Blocking opioid action in the RVM by microinjecting the opioid antagonist naltrexone reversed the analgesic and respiratory effects of systemically administered morphine, showing that this region plays a role in both the analgesic and respiratory-depressant properties of systemically administered morphine. The distribution of neurons directly inhibited by RVM opioid microinjection was determined with a fluorescent opioid peptide, dermorphin-Alexa 594, and found to be concentrated in and around the RVM. The non-opioid analgesic improgan, like DAMGO, produced antinociception but, unlike DAMGO, stimulated breathing when microinjected into the RVM. Concurrent recording of RVM neurons during improgan microinjection showed that this agent activated RVM ON-cells, OFF-cells, and NEUTRAL-cells. Since opioids are known to activate OFF-cells but suppress ON-cell firing, the differential respiratory response to these two analgesic drugs is best explained by their opposing effects on the activity of RVM ON-cells. These findings show that pain relief can be separated pharmacologically from respiratory depression and identify RVM OFF-cells as important central targets for continued development of potent analgesics with fewer side effects.


Assuntos
Analgésicos Opioides/toxicidade , Bulbo/efeitos dos fármacos , Neurônios/fisiologia , Dor Nociceptiva/fisiopatologia , Insuficiência Respiratória/induzido quimicamente , Analgésicos Opioides/antagonistas & inibidores , Animais , Ala(2)-MePhe(4)-Gly(5)-Encefalina/antagonistas & inibidores , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Potenciais Evocados/efeitos dos fármacos , Potenciais Evocados/fisiologia , Masculino , Bulbo/citologia , Bulbo/fisiologia , Morfina/antagonistas & inibidores , Morfina/farmacologia , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Neurônios/efeitos dos fármacos , Nociceptividade/efeitos dos fármacos , Nociceptividade/fisiologia , Ratos , Ratos Sprague-Dawley , Insuficiência Respiratória/fisiopatologia
12.
Mol Pharmacol ; 81(3): 348-55, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22113080

RESUMO

The phosphorylation of µ-opioid receptors (MOPRs) by G protein-coupled receptor kinases (GRKs), followed by arrestin binding, is thought to be a key pathway leading to desensitization and internalization. The present study used the combination of intracellular and whole-cell recordings from rats and mice, as well as live cell imaging of Flag-tagged MOPRs from mouse locus ceruleus neurons, to examine the role of protein kinases in acute desensitization and receptor trafficking. Inhibition of GRKs by using heparin or GRK2-mutant mice did not block desensitization or alter the rate of recovery from desensitization. The nonselective kinase inhibitor staurosporine did not reduce the extent of [Met(5)]enkephalin (ME)-induced desensitization but increased the rate of recovery from desensitization. In the presence of staurosporine, ME-activated FlagMOPRs were internalized but did not traffic away from the plasma membrane. The increased rate of recovery from desensitization correlated with the enhancement in the recycling of receptors to the plasma membrane. ME-induced MOPR desensitization persisted and the trafficking of receptors was modified after inhibition of protein kinases. The results suggest that desensitization of MOPRs may be an early step after agonist binding that is modulated by but is not dependent on kinase activity.


Assuntos
Locus Cerúleo/metabolismo , Neurônios/metabolismo , Receptores Opioides mu/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Locus Cerúleo/citologia , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Neurônios/enzimologia , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico , Ratos , Ratos Sprague-Dawley , Estaurosporina/farmacologia
13.
J Neurosci ; 29(22): 7341-8, 2009 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-19494155

RESUMO

Buprenorphine is a weak partial agonist at mu-opioid receptors that is used for treatment of pain and addiction. Intracellular and whole-cell recordings were made from locus ceruleus neurons in rat brain slices to characterize the actions of buprenorphine. Acute application of buprenorphine caused a hyperpolarization that was prevented by previous treatment of slices with the irreversible opioid antagonist beta-chlornaltrexamine (beta-CNA) but was not reversed by a saturating concentration of naloxone. As expected for a partial agonist, subsaturating concentrations of buprenorphine decreased the [Met](5)enkephalin (ME)-induced hyperpolarization or outward current. When the ME-induced current was decreased below a critical value, desensitization and internalization of mu-opioid receptors was eliminated. The inhibition of desensitization by buprenorphine was not the result of previous desensitization, slow dissociation from the receptor, or elimination of receptor reserve. Treatment of slices with subsaturating concentrations of etorphine, methadone, oxymorphone, or beta-CNA also reduced the current induced by ME but did not block ME-induced desensitization. Treatment of animals with buprenorphine for 1 week resulted in the inhibition of the current induced by ME and a block of desensitization that was not different from the acute application of buprenorphine to brain slices. These observations show the unique characteristics of buprenorphine and further demonstrate the range of agonist-selective actions that are possible through G-protein-coupled receptors.


Assuntos
Buprenorfina/farmacologia , Entorpecentes/farmacologia , Inibição Neural/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Receptores Opioides mu/metabolismo , Agonistas alfa-Adrenérgicos/farmacologia , Antagonistas Adrenérgicos alfa/farmacologia , Análise de Variância , Animais , Biofísica , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Tartarato de Brimonidina , Relação Dose-Resposta a Droga , Estimulação Elétrica/métodos , Encefalina Metionina/farmacologia , Técnicas In Vitro , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Camundongos , Camundongos Transgênicos , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Inibição Neural/genética , Neurônios/fisiologia , Técnicas de Patch-Clamp/métodos , Quinoxalinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/agonistas , Receptores Opioides mu/deficiência , Ioimbina/farmacologia
14.
J Neurosci ; 29(1): 222-33, 2009 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-19129399

RESUMO

mu-Opioid receptors (MORs) are G-protein-coupled receptors (GPCRs) that mediate the physiological effects of endogenous opioid neuropeptides and opiate drugs such as morphine. MORs are coexpressed with neurokinin 1 receptors (NK1Rs) in several regions of the CNS that control opioid dependence and reward. NK1R activation affects opioid reward specifically, however, and the cellular basis for this specificity is unknown. We found that ligand-induced activation of NK1Rs produces a cell-autonomous and nonreciprocal inhibition of MOR endocytosis induced by diverse opioids. Studies using epitope-tagged receptors expressed in cultured striatal neurons and a neuroblastoma cell model indicated that this heterologous regulation is mediated by NK1R-dependent sequestration of arrestins on endosome membranes. First, endocytic inhibition mediated by wild-type NK1Rs was overcome in cells overexpressing beta-arrestin2, a major arrestin isoform expressed in striatum. Second, NK1R activation promoted sequestration of beta-arrestin2 on endosomes, whereas MOR activation did not. Third, heterologous inhibition of MOR endocytosis was prevented by mutational disruption of beta-arrestin2 sequestration by NK1Rs. NK1R-mediated regulation of MOR trafficking was associated with reduced opioid-induced desensitization of adenylyl cyclase signaling in striatal neurons. Furthermore, heterologous regulation of MOR trafficking was observed in both amygdala and locus ceruleus neurons that naturally coexpress these receptors. These results identify a cell-autonomous mechanism that may underlie the highly specific effects of NK1R on opioid signaling and suggest, more generally, that receptor-specific trafficking of arrestins may represent a fundamental mechanism for coordinating distinct GPCR-mediated signals at the level of individual CNS neurons.


Assuntos
Analgésicos Opioides/farmacologia , Corpo Estriado/citologia , Endocitose/efeitos dos fármacos , Morfina/farmacologia , Neurônios/efeitos dos fármacos , Receptores da Neurocinina-1/fisiologia , Receptores Opioides mu/metabolismo , Animais , Biotinilação/métodos , Linhagem Celular Tumoral , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Encefalina Metionina/farmacologia , Feminino , Locus Cerúleo/citologia , Camundongos , Mutação/genética , Neuroblastoma , Gravidez , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/genética , Substância P/farmacologia , Transfecção/métodos
15.
Mol Pharmacol ; 74(4): 972-9, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18612077

RESUMO

The activation of G protein-coupled receptors results in a cascade of events that include acute signaling, desensitization, and internalization, and it is thought that not all agonists affect each process to the same extent. The early steps in opioid receptor signaling, including desensitization, have been characterized electrophysiologically using brain slice preparations, whereas most previous studies of opioid receptor trafficking have been conducted in heterologous cell models. This study used transgenic mice that express an epitope-tagged (FLAG) micro-opioid receptor (FLAGMOR) targeted to catecholamine neurons by regulatory elements from the tyrosine hydroxylase gene. Brain slices from these mice were used to study tagged MOR receptors in neurons of the locus ceruleus. Activation of the FLAGMOR with [Met5]enkephalin (ME) produced a hyperpolarization that desensitized acutely to the same extent as native MOR in slices from wild-type mice. A series of opioid agonists were then used to study desensitization and receptor trafficking in brain slices, which was monitored with a monoclonal antibody against the FLAG epitope (M1) conjugated to Alexa 594. Three patterns of receptor trafficking and desensitization were observed: 1) ME, etorphine, and methadone resulted in both receptor desensitization and internalization; 2) morphine and oxymorphone caused significant desensitization without evidence for internalization; and 3) oxycodone was ineffective in both processes. These results show that two distinct forms of signaling were differentially engaged depending on the agonist used to activate the receptor, and they support the hypothesis that ligand-specific regulation of opioid receptors occurs in neurons maintained in brain slices from adult animals.


Assuntos
Encéfalo/metabolismo , Receptores Opioides mu/metabolismo , Receptores Opioides mu/fisiologia , Analgésicos Opioides/farmacologia , Animais , Quelantes/farmacologia , Ácido Egtázico/farmacologia , Eletrofisiologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Etorfina/farmacologia , Técnica Direta de Fluorescência para Anticorpo , Locus Cerúleo/citologia , Locus Cerúleo/metabolismo , Metadona/farmacologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Morfina/farmacologia , Neurônios/metabolismo , Oxicodona/farmacologia , Oximorfona/farmacologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Ensaio Radioligante , Receptores Opioides mu/agonistas , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Tempo
16.
J Neurosci ; 26(15): 4118-25, 2006 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-16611829

RESUMO

A close relationship between desensitization and internalization of mu-opioid receptors (MORs) has been proposed based on differential actions of series of agonists. The role that these two processes have in the development of tolerance and dependence to opioids has been a controversial subject that has been studied in a variety of model systems. Here, we examine desensitization and internalization of endogenous MORs simultaneously in primary cultures of locus ceruleus neurons using fluorescently tagged peptide agonists. With the use of two fluorescent opioid peptides, dermorphin-Bodipy Texas Red and dermorphin-Alexa594 (Derm-A594), desensitization was measured electrophysiologically and trafficking was followed by the accumulation of intracellular fluorescent puncta. Blocking endocytosis with concanavalin A eliminated the accumulation of fluorescent puncta but desensitization induced by Derm-A594 was unaffected. Likewise, after treatment with concanavalin A, there was no change in either desensitization or recovery from desensitization induced by [Met]5enkephalin. The results demonstrate that desensitization and the recovery from desensitization are not dependent on receptor internalization and suggest that the activity of endogenous MORs in primary neurons can be modulated at the level of the plasma membrane.


Assuntos
Encéfalo/fisiologia , Neurônios/fisiologia , Receptores Opioides mu/fisiologia , Animais , Técnicas de Cultura de Células , Genes Reporter , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Locus Cerúleo/fisiologia , Camundongos , Camundongos Transgênicos , Neurônios/citologia
17.
J Neurosci ; 22(13): 5769-76, 2002 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12097530

RESUMO

micro-Opioid receptor (MOR) desensitization and endocytosis have been implicated in tolerance and dependence to opioids. The efficiency of each process is known to be agonist dependent; however, it is not known what determines the relative efficiency of various agonists at either process. In the present study, homologous MOR desensitization in locus ceruleus (LC) neurons and MOR internalization in HEK293 cells were examined using a series of agonists. The results show that the rank order of this series of agonists was different when comparing the magnitude of hyperpolarization and the ability to cause desensitization in LC neurons. Endocytosis of MOR was also examined in HEK293 cells using the same agonists. The relative ability to cause endocytosis in HEK293 cells correlated with the degree of desensitization in LC cells. This strong correlation suggests that the two processes are closely linked. The results also suggest that agonist efficacy is not necessarily a predictor of the ability to cause MOR desensitization or endocytosis. Identification and characterization of the biophysical properties of agonists that favor desensitization and internalization of receptors will lead to a better understanding of opioid signaling.


Assuntos
Endocitose , Canais de Potássio/fisiologia , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Animais , Linhagem Celular , Condutividade Elétrica , Humanos , Cinética , Ligantes , Locus Cerúleo/fisiologia , Masculino , Potenciais da Membrana , Entorpecentes/agonistas , Entorpecentes/farmacologia , Oligopeptídeos/farmacologia , Peptídeos Opioides , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA