Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 171
Filtrar
1.
J Pharmacol Exp Ther ; 384(3): 343-352, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36456196

RESUMO

Major depressive disorder is a highly common disorder, with a lifetime prevalence in the United States of approximately 21%. Traditional antidepressant treatments are limited by a delayed onset of action and minimal efficacy in some patients. Ketamine is effective and fast-acting, but there are concerns over its abuse liability. Thus, there is a need for safe, fast-acting antidepressant drugs. The opioid buprenorphine shows promise but also has abuse liability due to its mu-agonist component. Preclinical evidence indicates that the delta-opioid system contributes to mood disorders, and delta-opioid agonists are effective in preclinical models of depression- and anxiety-like states. In this study, we test the hypothesis that the mu-opioid antagonist diprenorphine by virtue of its partial delta opioid agonist activity may offer a beneficial profile for an antidepressant medication without abuse liability. Diprenorphine was confirmed to bind with high affinity to all three opioid receptors, and functional experiments for G protein activation verified diprenorphine to be a partial agonist at delta- and kappa-opioid receptors and a mu-antagonist. Studies in C57BL/6 mice demonstrated that an acute dose of diprenorphine produced antidepressant-like effects in the tail suspension test and the novelty-induced hypophagia test that were inhibited in the presence of the delta-selective antagonist, naltrindole. Diprenorphine did not produce convulsions, a side effect of many delta agonists but rather inhibited convulsions caused by the full delta agonist SNC80; however, diprenorphine did potentiate pentylenetetrazole-induced convulsions. Diprenorphine, and compounds with a similar pharmacological profile, may provide efficient and safe rapidly acting antidepressants. SIGNIFICANCE STATEMENT: The management of major depressive disorder, particularly treatment-resistant depression, is a significant unmet medical need. Here we show that the opioid diprenorphine, a compound with mu-opioid receptor antagonist activity and delta- and kappa-opioid receptor partial agonist activities, has rapid onset antidepressant-like activity in animal models. Diprenorphine and compounds with a similar pharmacological profile to diprenorphine should be explored as novel antidepressant drugs.


Assuntos
Analgésicos Opioides , Transtorno Depressivo Maior , Diprenorfina , Animais , Camundongos , Analgésicos Opioides/farmacologia , Antidepressivos/farmacologia , Diprenorfina/farmacologia , Camundongos Endogâmicos C57BL , Receptores Opioides , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Convulsões/induzido quimicamente
2.
Cells ; 10(10)2021 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-34685631

RESUMO

Oxytocin (OT) influences various physiological functions such as uterine contractions, maternal/social behavior, and analgesia. Opioid signaling pathways are involved in one of the analgesic mechanisms of OT. We previously showed that OT acts as a positive allosteric modulator (PAM) and enhances µ-opioid receptor (MOR) activity. In this study, which focused on other opioid receptor (OR) subtypes, we investigated whether OT influences opioid signaling pathways as a PAM for δ-OR (DOR) or κ-OR (KOR) using human embryonic kidney-293 cells expressing human DOR or KOR, respectively. The CellKeyTM results showed that OT enhanced impedance induced by endogenous/exogenous KOR agonists on KOR-expressing cells. OT did not affect DOR activity induced by endogenous/exogenous DOR agonists. OT potentiated the KOR agonist-induced Gi/o protein-mediated decrease in intracellular cAMP, but did not affect the increase in KOR internalization caused by the KOR agonists dynorphin A and (-)-U-50488 hydrochloride (U50488). OT did not bind to KOR orthosteric binding sites and did not affect the binding affinities of dynorphin A and U50488 for KOR. These results suggest that OT is a PAM of KOR and MOR and enhances G protein signaling without affecting ß-arrestin signaling. Thus, OT has potential as a specific signaling-biased PAM of KOR.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Ocitocina/farmacologia , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Transdução de Sinais , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Regulação Alostérica/efeitos dos fármacos , Animais , Sítios de Ligação , Células CHO , Cricetulus , AMP Cíclico/metabolismo , Diprenorfina/farmacologia , Dinorfinas/farmacologia , Impedância Elétrica , Endocitose/efeitos dos fármacos , Células HEK293 , Humanos , Concentração Inibidora 50 , Receptores Opioides delta/agonistas , Receptores Opioides kappa/agonistas , Transdução de Sinais/efeitos dos fármacos
3.
J Wildl Dis ; 57(2): 357-367, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33822147

RESUMO

Aerial translocation of captured black rhinoceroses (Diceros bicornis) has been accomplished by suspending them by their feet. We expected this posture would compromise respiratory gas exchange more than would lateral recumbency. Because white rhinoceroses (Ceratotherium simum) immobilized with etorphine alone are hypermetabolic, with a high rate of carbon dioxide production (VCO2), we expected immobilized black rhinoceroses would also have a high VCO2. Twelve (nine male, three female; median age 8 yr old [range: 4-25]; median weight 1,137 kg [range: 804-1,234] body weight) wild black rhinoceroses were immobilized by aerial darting with etorphine and azaperone. The animals were in lateral recumbency or suspended by their feet from a crane for approximately 10 min before data were collected. Each rhinoceros received both treatments sequentially, in random order. Six were in lateral recumbency first and six were suspended first. All animals were substantially hypoxemic and hypercapnic in both postures. When suspended by the feet, mean arterial oxygen pressure (PaO2) was 42 mm Hg, 4 mm Hg greater than in lateral recumbency (P=0.030), and arterial carbon dioxide pressure (PaCO2) was 52 mm Hg, 3 mm Hg less than in lateral recumbency (P=0.016). Tidal volume and minute ventilation were similar between postures. The mean VCO2 was 2 mL/kg/min in both postures and was similar to, or marginally greater than, VCO2 predicted allometrically. Suspension by the feet for 10 min did not impair pulmonary function more than did lateral recumbency and apparently augmented gas exchange to a small degree relative to lateral recumbency. The biological importance in these animals of numerically small increments in PaO2 and decrements in PaCO2 with suspension by the feet is unknown. Black rhinoceroses immobilized with etorphine and azaperone were not as hypermetabolic as were white rhinoceroses immobilized with etorphine.


Assuntos
Metabolismo Energético/efeitos dos fármacos , Etorfina/farmacologia , Imobilização/veterinária , Perissodáctilos , Fenômenos Fisiológicos Respiratórios/efeitos dos fármacos , Animais , Animais Selvagens , Diprenorfina/administração & dosagem , Diprenorfina/farmacologia , Etorfina/administração & dosagem , Feminino , Hipnóticos e Sedativos/administração & dosagem , Hipnóticos e Sedativos/farmacologia , Masculino , Naltrexona/administração & dosagem , Naltrexona/farmacologia , Antagonistas de Entorpecentes/administração & dosagem , Antagonistas de Entorpecentes/farmacologia , Postura
4.
ChemMedChem ; 11(13): 1415-27, 2016 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-27193393

RESUMO

Ligand efficiency indices are widely used to guide chemical optimization in drug discovery, due to their predictive value in the early steps of optimization. At later stages, however, as more complex properties become critical for success, indices relying on calculated, rather than experimental, parameters become less informative. This problem is particularly acute when developing positron emission tomography (PET) imaging agents, for which nonspecific binding (NSB) to membranes and non-target proteins is a frequent cause of failure. NSB cannot be predicted using in silico parameters. To address this gap, we explored the use of the experimentally determined chromatographic hydrophobicity index on immobilized artificial membranes, CHI(IAM), to guide the optimization of NSB. The ligand specific efficiency (LSE) index was defined as the ratio between affinity (pIC50 or pKd ) and the logarithmic value of CHI(IAM). It allows for quantification of binding affinity to the target of interest, relative to NSB. Its use was illustrated by the optimization of PET tracer candidates for the prostacyclin receptor.


Assuntos
Compostos Radiofarmacêuticos/química , Compostos de Anilina/química , Compostos de Anilina/farmacologia , Animais , Benzamidas/química , Benzamidas/farmacologia , Cricetulus , Diprenorfina/química , Diprenorfina/farmacologia , Antagonistas dos Receptores de Dopamina D2/química , Antagonistas dos Receptores de Dopamina D2/farmacologia , Desenho de Fármacos , Humanos , Interações Hidrofóbicas e Hidrofílicas , Imidazóis/química , Imidazóis/farmacologia , Ligantes , Membranas Artificiais , Antagonistas de Entorpecentes/química , Antagonistas de Entorpecentes/farmacologia , Tomografia por Emissão de Pósitrons , Piridonas/síntese química , Piridonas/química , Compostos Radiofarmacêuticos/farmacologia , Receptores de Epoprostenol/química , Receptores de Epoprostenol/metabolismo , Trítio
5.
J Labelled Comp Radiopharm ; 56(3-4): 187-95, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24285325

RESUMO

Opiate receptors are found in the brain, the spinal cord, some peripheral sensory neurons, and the gastrointestinal tract. Naturally occurring and synthetic opiate ligands exert their influence on a wide variety of processes including analgesia, euphoria, dysphoria, sedation, respiratory depression, and miosis and are frequently topics for discussions on addiction and physical dependence. This review looks at the history of positron emission tomography radioligands for probing this receptor system.


Assuntos
Diprenorfina/farmacologia , Fentanila/farmacologia , Compostos Radiofarmacêuticos/farmacologia , Receptores Opioides/metabolismo , Animais , Encéfalo/diagnóstico por imagem , Diprenorfina/análogos & derivados , Diprenorfina/síntese química , Fentanila/análogos & derivados , Fentanila/síntese química , Humanos , Ligantes , Antagonistas de Entorpecentes , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/síntese química , Receptores Opioides/agonistas , Especificidade por Substrato
6.
Structure ; 19(11): 1683-90, 2011 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-22078567

RESUMO

Opioids that stimulate the µ-opioid receptor (MOR1) are the most frequently prescribed and effective analgesics. Here we present a structural model of MOR1. Molecular dynamics simulations show a ligand-dependent increase in the conformational flexibility of the third intracellular loop that couples with the G protein complex. These simulations likewise identified residues that form frequent contacts with ligands. We validated the binding residues using site-directed mutagenesis coupled with radioligand binding and functional assays. The model was used to blindly screen a library of ∼1.2 million compounds. From the 34 compounds predicted to be strong binders, the top three candidates were examined using biochemical assays. One compound showed high efficacy and potency. Post hoc testing revealed this compound to be nalmefene, a potent clinically used antagonist, thus further validating the model. In summary, the MOR1 model provides a tool for elucidating the structural mechanism of ligand-initiated cell signaling and for screening novel analgesics.


Assuntos
Simulação de Dinâmica Molecular , Receptores Opioides mu/química , Substituição de Aminoácidos , Animais , Sítios de Ligação , Ligação Competitiva , Bovinos , AMP Cíclico/química , AMP Cíclico/metabolismo , AMP Cíclico/farmacologia , Bases de Dados Factuais , Decapodiformes , Diprenorfina/química , Diprenorfina/farmacologia , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Morfina/química , Morfina/farmacologia , Mutagênese Sítio-Dirigida , Naltrexona/análogos & derivados , Naltrexona/química , Naltrexona/farmacologia , Ligação Proteica , Ensaio Radioligante , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Bibliotecas de Moléculas Pequenas
7.
J Pharmacol Exp Ther ; 335(3): 562-71, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20829393

RESUMO

Buprenorphine is a partial µ-opioid receptor agonist used for the treatment of opioid dependence that has several advantages over methadone. The principal route of buprenorphine disposition has been well established; however, little is known regarding the potential for buprenorphine to influence the metabolism and clearance of other drugs by affecting the expression of drug-metabolizing enzymes (DMEs). Here, we investigate the effects of buprenorphine on the activation of pregnane X receptor (PXR) and constitutive androstane receptor (CAR), as well as the induction of DMEs, in both HepG2 cells and human primary hepatocytes (HPHs). In HepG2 cells, buprenorphine significantly increased human PXR-mediated CYP2B6 and CYP3A4 reporter activities. CYP2B6 reporter activity was also enhanced by buprenorphine in HepG2 cells cotransfected with a chemical-responsive human CAR variant. Real-time reverse transcription-polymerase chain reaction analysis revealed that buprenorphine strongly induced CYP3A4 expression in both PXR- and CAR-transfected HepG2 cells. However, treatment with the same concentrations of buprenorphine in HPHs resulted in literally no induction of CYP3A4 or CYP2B6 expression. Further studies indicated that buprenorphine could neither translocate human CAR to the nucleus nor activate CYP2B6/CYP3A4 reporter activities in transfected HPHs. Subsequent experiments to determine whether the differential response was due to buprenorphine's metabolic stability revealed a dramatically differential rate of elimination for buprenorphine between HPHs and HepG2 cells. Taken together, these studies indicate that metabolic stability of buprenorphine defines the differential induction of DMEs observed in HepG2 and HPHs, and the results obtained from PXR and CAR reporter assays in immortalized cell line require cautious interpretation.


Assuntos
Buprenorfina/metabolismo , Buprenorfina/farmacologia , Hepatócitos/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/fisiologia , Analgésicos Opioides/metabolismo , Hidrocarboneto de Aril Hidroxilases/genética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Receptor Constitutivo de Androstano , Citocromo P-450 CYP2B6 , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Diprenorfina/metabolismo , Diprenorfina/farmacologia , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Genes Reporter/genética , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Humanos , Oxirredutases N-Desmetilantes/genética , Oxirredutases N-Desmetilantes/metabolismo , Receptor de Pregnano X , Receptores Citoplasmáticos e Nucleares/agonistas , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Receptores Citoplasmáticos e Nucleares/genética , Receptores de Esteroides/agonistas , Receptores de Esteroides/antagonistas & inibidores , Receptores de Esteroides/genética , Transfecção
8.
Acta Neurobiol Exp (Wars) ; 70(2): 147-64, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20628439

RESUMO

This longitudinal, case-control pilot study examined amygdala growth in rhesus macaque infants receiving the complete US childhood vaccine schedule (1994-1999). Longitudinal structural and functional neuroimaging was undertaken to examine central effects of the vaccine regimen on the developing brain. Vaccine-exposed and saline-injected control infants underwent MRI and PET imaging at approximately 4 and 6 months of age, representing two specific timeframes within the vaccination schedule. Volumetric analyses showed that exposed animals did not undergo the maturational changes over time in amygdala volume that was observed in unexposed animals. After controlling for left amygdala volume, the binding of the opioid antagonist [(11)C]diprenorphine (DPN) in exposed animals remained relatively constant over time, compared with unexposed animals, in which a significant decrease in [(11)C]DPN binding occurred. These results suggest that maturational changes in amygdala volume and the binding capacity of [(11)C]DPN in the amygdala was significantly altered in infant macaques receiving the vaccine schedule. The macaque infant is a relevant animal model in which to investigate specific environmental exposures and structural/functional neuroimaging during neurodevelopment.


Assuntos
Tonsila do Cerebelo/crescimento & desenvolvimento , Analgésicos Opioides/antagonistas & inibidores , Timerosal/toxicidade , Vacinação/efeitos adversos , Tonsila do Cerebelo/imunologia , Animais , Animais Recém-Nascidos , Estudos de Casos e Controles , Diprenorfina/farmacologia , Esquemas de Imunização , Ligantes , Estudos Longitudinais , Macaca mulatta , Imageamento por Ressonância Magnética/métodos , Masculino , Modelos Animais , Projetos Piloto , Tomografia por Emissão de Pósitrons/métodos
9.
J Pharm Sci ; 98(12): 4928-40, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19370547

RESUMO

Conclusions based on either in vitro or in vivo approach to evaluate the P-gp affinity status of opioids may be misleading. For example, in vitro studies indicated that fentanyl is a P-gp inhibitor while in vivo studies indicated that it is a P-gp substrate. Quite the opposite was evident for meperidine. The objective of this study was to evaluate the P-gp affinity status of methadone, buprenorphine and diprenorphine to predict P-gp-mediated drug-drug interactions and to determine a better candidate for management of opioid dependence. Two in vitro (P-gp ATPase and monolayer efflux) assays and two in vivo (tissue distribution and antinociceptive evaluation in mdr1a/b (-/-) mice) assays were used. Methadone stimulated the P-gp ATPase activity only at higher concentrations, while verapamil and GF120918 inhibited its efflux (p < 0.05). The brain distribution and antinociceptive activity of methadone were enhanced (p < 0.05) in P-gp knockout mice. Conversely, buprenorphine and diprenorphine were negative in all assays. P-gp can affect the PK/PD of methadone, but not buprenorphine or diprenorphine. Our report is in favor of buprenorphine over methadone for management of opioid dependence. Buprenorphine most likely is not a P-gp substrate and concerns regarding P-gp-mediated drug-drug interaction are not expected.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Analgésicos Opioides/farmacologia , Analgésicos/farmacologia , Buprenorfina/farmacologia , Diprenorfina/farmacologia , Absorção Intestinal/fisiologia , Metadona/farmacologia , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/metabolismo , Algoritmos , Analgésicos/farmacocinética , Analgésicos Opioides/farmacocinética , Animais , Buprenorfina/farmacocinética , Células CACO-2 , Proteínas de Transporte/genética , Diprenorfina/farmacocinética , Humanos , Manitol/análise , Manitol/metabolismo , Metadona/farmacocinética , Camundongos , Camundongos Knockout , Paclitaxel/análise , Paclitaxel/metabolismo , Conformação Proteica , Tempo de Reação/efeitos dos fármacos , Distribuição Tecidual
10.
Neuroreport ; 19(18): 1793-6, 2008 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-18955903

RESUMO

Opioid efficacy on mu-receptor may be influenced by various Gi/o-G-protein subunits interacting with intracellular face of receptor. Pertussis toxin-insensitive Galphai1 and Galphai2 subunits tethered with mu-receptor were stably transfected into AtT20 cells to (i) determine coupling of different alpha-subunits on opioid efficacy, and (ii) determine coupling to downstream effectors, for example, calcium and potassium channels. After pertussis toxin, stimulation of [35S]GTP-gamma-S incorporation persisted. Both constructs were able to couple to native calcium and potassium channels, with endomorphins 1 and 2 equally effective. However, pertussis toxin abolished opioid actions on calcium and potassium channels suggesting strong coupling to endogenous G-proteins, and that differences in coupling efficacy to Galphai1 and Galphai2 previously observed are restricted to initial step of signaling cascade.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Toxina Pertussis/farmacologia , Receptores Opioides mu/metabolismo , Analgésicos Opioides/farmacologia , Animais , Canais de Cálcio/metabolismo , Linhagem Celular Tumoral , Colo/metabolismo , Diprenorfina/farmacologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Camundongos , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Oligopeptídeos/farmacologia , Canais de Potássio/metabolismo , Ligação Proteica/efeitos dos fármacos , Receptores Opioides mu/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Transfecção/métodos
11.
Biochemistry ; 47(40): 10576-86, 2008 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-18778083

RESUMO

We previously demonstrated that D3.49(164)Y or T6.34(279)K mutation in the rat mu opioid receptor (MOPR) resulted in agonist-independent activation. Here, we identified the cysteine(s) within the transmembrane domains (TMs) of the D3.49(164)Y mutant that became accessible in the binding-site crevice by use of methanethiosulfonate ethylammonium (MTSEA) and inferred conformational changes associated with receptor activation. While the C7.38(321)S mutant was insensitive to MTSEA, the D3.49(164)Y/C7.38(321)S mutant showed similar sensitivity as the D3.49(164)Y, suggesting that, in the D3.49(164)Y mutant, C7.38(321) becomes inaccessible while other cysteines are accessible in the binding-site crevice. Each of the other seven cysteines in the TMs was mutated to serine on the background of D3.49(164)Y/C7.38(321)S, and the resulting triple mutants were evaluated for [3H]diprenorphine and [d-Ala2,NMe-Phe4,Gly5-ol]-enkephalin (DAMGO) binding and effect of MTSEA on [3H]diprenorphine binding. The D3.49(164)Y/C7.38(321)S mutant and the triple mutants, except the C6.47(292)S triple mutant, retained similar affinities for [3H]diprenorphine and DAMGO as the D3.49(164)Y mutant. The second-order rate constants for MTSEA reactions showed that C3.44(159)S, C4.48(190)S, C5.41(235)S, and C7.47(330)S significantly reduced sensitivity to MTSEA, compared with the D3.49(164)Y/C7.38(321)S. These results suggest that the four cysteines may be rotated and/or tilted to become accessible. While the D3.49(164)Y/C7.38(321)S was similarly sensitive to MTSEA as the D3.49(164)Y mutant, the T6.34(279)K/C7.38(321)S was much less sensitive to MTSEA than the T6.34(279)K mutant, suggesting that the two constitutively active mutants assume different conformations and/or possess different dynamic properties. Molecular models of the MOPR monomer and homodimer, using the crystal structures of rhodopsin, the beta2-adrenergic receptor, and the ligand-free opsin, which contains several features characteristic of the active state, were employed to analyze these experimental results in a structural context.


Assuntos
Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Diprenorfina/metabolismo , Diprenorfina/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ligação Proteica , Estrutura Terciária de Proteína/genética , Ratos , Receptores Opioides mu/genética
12.
Biochim Biophys Acta ; 1778(6): 1483-92, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18423369

RESUMO

Lipid rafts depicted as densely packed and thicker membrane microdomains, based on the dynamic clustering of cholesterol and sphingolipids, may help as platforms involved in a wide variety of cellular processes. The reasons why proteins segregate into rafts are yet to be clarified. The human delta opioid receptor (hDOR) reconstituted in a model system has been characterised after ligand binding by an elongation of its transmembrane part, inducing rearrangement of its lipid microenvironment [Alves, Salamon, Hruby, and Tollin (2005) Biochemistry 44, 9168-9178]. We used hDOR to understand better the correlation between its function and its membrane microdomain localisation. A fusion protein of hDOR with the Green Fluorescent Protein (DOR*) allows precise receptor membrane quantification. Here we report that (i) a fraction of the total receptor pool requires cholesterol for binding activity, (ii) G-proteins stabilize a high affinity state conformation which does not seem modulated by cholesterol. In relation to its distribution, and (iii) a fraction of DOR* is constitutively associated with detergent-resistant membranes (DRM) characterised by an enrichment in lipids and proteins raft markers. (iv) An increase in the quantity of DOR* was observed upon agonist addition. (v) This DRM relocation is prevented by uncoupling the receptor-G-protein interaction.


Assuntos
Colesterol/metabolismo , Diprenorfina/farmacologia , Microdomínios da Membrana/metabolismo , Antagonistas de Entorpecentes/farmacologia , Oligopeptídeos/farmacologia , Receptores Opioides delta/agonistas , Linhagem Celular , Humanos , Ligantes , Microdomínios da Membrana/genética , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína/fisiologia , Receptores Opioides delta/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Esfingolipídeos/metabolismo
13.
Mol Pharmacol ; 73(4): 1319-30, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18212250

RESUMO

Ubiquitination of the human kappa opioid receptor (hKOR) expressed in Chinese hamster ovary (CHO) cells was observed in the presence of the proteasomal inhibitor N-benzoyloxycarbonyl (Z)-Leu-Leu-leucinal (MG132) and enhanced by the agonists (-)(trans)-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidiny) cyclohexyl] benzeneacetamide (U50,488H) and dynorphin A (Dyn A). The dominant-negative (DN) mutants GRK2-K220R and beta-arrestin (319-418), but not dynamin I-K44A, reduced Dyn A-stimulated hKOR ubiquitination, and a phosphorylation-defective hKOR mutant (hKOR-S358N) did not undergo Dyn A-stimulated ubiquitination, indicating that hKOR ubiquitination is enhanced by receptor phosphorylation but not by receptor internalization. A hKOR mutant (hKOR-10 KR) in which all 10 intracellular Lys residues were changed to Arg showed greatly reduced basal and agonist-promoted receptor ubiquitination and substantially decreased Dyn A-induced receptor down-regulation, without changing ligand binding affinity, receptor-G protein coupling, or receptor internalization or desensitization. The ubiquitination sites were further determined to be the three Lys residues in the C-terminal domain. The K63R ubiquitin mutant decreased Dyn A-induced hKOR ubiquitination and down-regulation, but the K48R mutant did not. Expression of HN-CYLD, a DN mutant of deubiquitination enzyme cylindromatosis tumor suppressor gene (CYLD) that breaks Lys63-linked polyubiquitin chain, increased Dyn A-induced hKOR ubiquitination and down-regulation. These results indicate that ubiquitinated hKOR after agonist treatment contains predominantly Lys63-linked polyubiquitin chains and ubiquitination of the hKOR involved in agonist-induced down-regulation.


Assuntos
Regulação para Baixo , Lisina/metabolismo , Poliubiquitina/metabolismo , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/metabolismo , Ubiquitinação , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Sequência de Aminoácidos , Animais , Arginina/genética , Células CHO , Cricetinae , Cricetulus , Enzima Desubiquitinante CYLD , Diprenorfina/farmacologia , Regulação para Baixo/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Humanos , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutação/genética , Antagonistas de Entorpecentes/farmacologia , Fosforilação/efeitos dos fármacos , Estrutura Terciária de Proteína , Receptores Acoplados a Proteínas G/metabolismo , Receptores Opioides kappa/química , Proteínas Supressoras de Tumor/metabolismo , Ubiquitinação/efeitos dos fármacos
14.
Brain Res ; 1177: 9-18, 2007 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-17897634

RESUMO

We investigated the effects of morphine and other agonists on the human mu opioid receptor (MOP) expressed in M2 melanoma cells, lacking the actin cytoskeleton protein filamin A and in A7, a subclone of the M2 melanoma cells, stably transfected with filamin A cDNA. The results of binding experiments showed that after chronic morphine treatment (24 h) of A7 cells, MOP-binding sites were down-regulated to 63% of control, whereas, unexpectedly, in M2 cells, MOP binding was up-regulated to 188% of control naive cells. Similar up-regulation was observed with the agonists methadone and levorphanol. The presence of antagonists (naloxone or CTAP) during chronic morphine treatment inhibited MOP down-regulation in A7 cells. In contrast, morphine-induced up-regulation of MOP in M2 cells was further increased by these antagonists. Chronic morphine desensitized MOP in A7 cells, i.e., it decreased DAMGO-induced stimulation of GTPgammaS binding. In M2 cells DAMGO stimulation of GTPgammaS binding was significantly greater than in A7 cells and was not desensitized by chronic morphine. Pertussis toxin treatment abolished morphine-induced receptor up-regulation in M2 cells, whereas it had no effect on morphine-induced down-regulation in A7 cells. These results indicate that, in the absence of filamin A, chronic treatment with morphine, methadone or levorphanol leads to up-regulation of MOP, to our knowledge, the first instance of opioid receptor up-regulation by agonists in cell culture.


Assuntos
Proteínas Contráteis/fisiologia , Proteínas dos Microfilamentos/fisiologia , Morfina/farmacologia , Entorpecentes/farmacologia , Receptores Opioides mu/efeitos dos fármacos , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Proteínas Contráteis/deficiência , Interpretação Estatística de Dados , Diprenorfina/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Filaminas , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Ligantes , Melanoma/genética , Melanoma/patologia , Proteínas dos Microfilamentos/deficiência , Antagonistas de Entorpecentes/metabolismo , Antagonistas de Entorpecentes/farmacologia , Toxina Pertussis/farmacologia , Ensaio Radioligante , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Tubulina (Proteína)/farmacologia , Regulação para Cima/efeitos dos fármacos
15.
Biochemistry ; 46(38): 10960-70, 2007 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-17711303

RESUMO

We examined glycosylation of FLAG-hKOR expressed in CHO cells and determined its functional significance. FLAG-hKOR was resolved as a broad and diffuse 55-kDa band and a less diffuse 45-kDa band by immunoblotting, indicating that the receptor is glycosylated. Endoglycosidase H cleaved the 45-kDa band to approximately 38 kDa but did not change the 55-kDa band, demonstrating that the 45-kDa band is N-glycosylated with high-mannose or hybrid-type glycan. Peptide-N-glycosidase F digestion of solubilized hKOR or incubation of cells with tunicamycin resulted in two species of 43 and 38 kDa, suggesting that the 43-kDa band is O-glycosylated. FLAG-hKOR was reduced to lower Mr bands by neuraminidase and O-glycosidase, indicating that the hKOR contains O-linked glycan. Mutation of Asn25 or Asn39 to Gln in the N-terminal domain reduced the Mr by approximately 5 kDa, indicating that both residues were glycosylated. The double mutant hKOR-N25/39Q was resolved as a 43-kDa (mature form) and a 38-kDa (intermediate form) band. When transiently expressed, hKOR-N25/39Q had a lower expression level than the wild type. In CHO cells stably expressing the hKOR-N25/39Q, pulse-chase experiments revealed that the turnover rate constants (ke) of the intermediate and mature forms were approximately 3 times those of the wild type. In addition, the maturation rate constant (ka) of the 43-kDa form of hKOR-N25/39Q was 6 times that of the mature form of the wild type. The hKOR-N25/39Q mutant showed increased agonist-induced receptor phosphorylation, desensitization, internalization, and downregulation, without changing ligand binding affinity or receptor-G protein coupling. Thus, N-glycosylation of the hKOR plays important roles in stability and trafficking along the biosynthesis pathway of the receptor protein as well as agonist-induced receptor regulation.


Assuntos
Regulação da Expressão Gênica , Processamento de Proteína Pós-Traducional , Receptores de Superfície Celular/metabolismo , Receptores Opioides kappa/metabolismo , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Substituição de Aminoácidos , Analgésicos não Narcóticos/farmacologia , Animais , Células CHO , Cricetinae , Cricetulus , Diprenorfina/farmacologia , Regulação para Baixo , Eletroforese em Gel de Poliacrilamida , Regulação da Expressão Gênica/fisiologia , Glicosídeo Hidrolases/antagonistas & inibidores , Glicosídeo Hidrolases/metabolismo , Glicosilação , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Imunoprecipitação , Cinética , Mutagênese Sítio-Dirigida , Processamento de Proteína Pós-Traducional/fisiologia , Estrutura Terciária de Proteína , Transporte Proteico/fisiologia , Receptores de Superfície Celular/biossíntese , Receptores Opioides kappa/biossíntese , Proteínas Recombinantes de Fusão/efeitos dos fármacos , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Radioisótopos de Enxofre , Trítio , Tunicamicina/farmacologia
16.
J Pharmacol Exp Ther ; 323(2): 614-25, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17720886

RESUMO

Ligand-induced regulation of the rat kappa-opioid receptor (rKOR) was investigated in human embryonic kidney 293 cells stably expressing the FLAG-tagged rKOR. Incubation of rKOR cells with naltrexone for 24 h increased the B(max) >3-fold, with no change in the affinity of [(3)H]diprenorphine. Two immunoreactive receptor species were present in cell lysates: naltrexone treatment caused a >3-fold increase in the 52-kDa species while decreasing the level of the 42-kDa species. Dynorphin(1-13), U69,593 [(5alpha,7alpha,8beta)-(+)-N-methyl-N-(7-[1-pyrrolidinyl]-1-oxaspiro[4,5]dec-8-yl)benzeneacetamide], or salvinorin A [2S,4aR,6aR,7R,9S,10aS, 10bR)-9-(acetyloxy)-2-(3-furanyl)dodecahydro-6a,10b-dimethyl-4,10-dioxo-2H-naphtho[2,1c]pyran-7-carboxylic acid methyl ester] treatment did not alter the level of immunoreactive rKOR protein, whereas etorphine, cyclazocine, naloxone, and naloxone methiodide increased the 52-kDa and decreased the 42-kDa rKOR bands. Receptor up-regulation was associated with an increase in the number of cell surface receptors and a 2-fold increase in the E(max) for guanosine 5'-O-(3-[(35)S]thio)triphosphate binding. Glycosidase digestion indicated that the 52- and 42-kDa receptors contained complex and high-mannose N-glycans, respectively, Pulse-chase analysis and glycosidase digestion sensitivities suggested that the 42-kDa rKOR species was a precursor of the 52-kDa species. Naltrexone did not alter rKOR mRNA levels or translational efficiency, and rKOR up-regulation was not inhibited by cycloheximide. Brefeldin A caused accumulation of intracellular rKOR intermediates, and coincubation with naltrexone increased the levels of the brefeldin-induced species significantly. These results suggest that select opioid ligands up-regulate rKOR by enhancing the rate of receptor folding and maturation and by protecting the receptor from degradation, resulting in an increase in the number of rKOR binding sites, immunoreactive protein, and functional receptors.


Assuntos
Receptores Opioides kappa/genética , Animais , Benzenoacetamidas/farmacocinética , Brefeldina A/farmacologia , Células Cultivadas , Cicloeximida/farmacologia , Diprenorfina/farmacologia , Humanos , Ligantes , Naloxona/farmacocinética , Naloxona/farmacologia , Naltrexona/farmacologia , Pirrolidinas/farmacocinética , RNA Mensageiro/análise , Ratos , Receptores Opioides kappa/química , Receptores Opioides kappa/metabolismo , Transfecção , Regulação para Cima
17.
Biochem Pharmacol ; 73(4): 534-49, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17141202

RESUMO

Opioid receptors have been shown to be located in and regulated by lipid rafts/caveolae in caveolin-rich non-neuronal cells. Here, we found that caveolin-1 level was very low in rat brain and undetectable in NG108-15 cells, which endogenously express delta opioid receptors (DOR). Rat caudate putamen (CPu) membranes, NG108-15 cells and CHO cells stably transfected with FLAG-mouse-DOR (CHO-FLAG-mDOR) were homogenized, sonicated in a detergent-free 0.5M Na(2)CO(3) buffer and fractionated through discontinuous or continuous sucrose density gradients. About 70% of opioid receptors in CPu and DOR in both cell lines were present in low-density (5-20% sucrose) membrane domains enriched in cholesterol and ganglioside M1 (GM1), characteristics of lipid rafts in plasma membranes. In both cells, stimulation with permeable or non-permeable full agonists, but not with partial or inverse agonists, for 30min shifted approximately 25% of DORs out of rafts, by a naloxone-reversible and pertussis toxin-insensitive mechanism, which may undergo internalization. Methyl-beta-cyclodextrin (MCD) treatment greatly reduced cholesterol and shifted DOR to higher density fractions and decreased DPDPE affinities. MCD treatment attenuated DPDPE-induced [(35)S]GTPgammaS binding in CPu and NG108-15 cells, but enhanced it in CHO-FLAG-mDOR cells. In CHO-FLAG-mDOR cells, G(alphai) co-immunoprecipitated with caveolin-1, which was shown to inhibit G(alphai/o), and MCD treatment dramatically reduced the association leading to disinhibition. Thus, although localization in rafts and agonist-induced shift of DOR are independent of caveolin-1, lipid rafts sustain DOR-mediated signaling in caveolin-deficient neuronal cells, but appear to inhibit it in caveolin-enriched non-neuronal cells. Cholesterol-dependent association of caveolin-1 with and the resulting inhibition of G proteins may be a contributing factor.


Assuntos
Colesterol/metabolismo , Receptores Opioides delta/fisiologia , Transdução de Sinais/efeitos dos fármacos , beta-Ciclodextrinas/farmacologia , Animais , Anticolesterolemiantes/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Células CHO , Caveolina 1/metabolismo , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Diprenorfina/farmacologia , D-Penicilina (2,5)-Encefalina/farmacologia , Etorfina/farmacologia , Gangliosídeo G(M1)/metabolismo , Células Híbridas , Levorfanol/farmacologia , Microdomínios da Membrana/química , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Proteínas de Membrana/metabolismo , Naloxona/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Oligopeptídeos/farmacologia , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo
18.
Eur J Pharmacol ; 555(1): 1-7, 2007 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-17113072

RESUMO

Opioids, acting via G-protein coupled membrane receptors, induce analgesia. However their role is not limited to their anti-nociceptive action. They are found in several peripheral tissues acting as negative regulators of cellular processes. Even though that is not fully elucidated, it becomes obvious that opioids exert their effects in close relation to other neuropeptides such as somatostatin. Hepatocellular carcinoma is one tumor, among others, which secrete bioactive peptides while somatostatin analogs exert an inhibitory effect. We have used the human hepatocyte-derived cancer cell line HepG2, in order to examine the effect of opioids on cell growth and their possible mode of action. Our results show that the opioid ethylketocyclazocine (EKC) inhibits cell proliferation and induces apoptosis. This inhibitory effect is not exerted via opioids receptors since it was not reversed by the opioid antagonist diprenorphine and functional opioid receptors were not found on HepG2 cells. On the contrary, we show that EKC binds to somatostatin receptors, and activates a PTP signalling cascade. In this respect, the interaction of opioids with somatostatin receptors on hepatocellular carcinoma cells, and the fact that they are widely used for pain control, may provide some additional clues for the discrepancies during treatment with somatostatin analogues.


Assuntos
Analgésicos Opioides/farmacologia , Proliferação de Células/efeitos dos fármacos , Etilcetociclazocina/farmacologia , Receptores de Somatostatina/metabolismo , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Diprenorfina/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Leucina Encefalina-2-Alanina/farmacologia , Humanos , Antagonistas de Entorpecentes/farmacologia , Octreotida/farmacologia , RNA Mensageiro/metabolismo , Receptores Opioides/genética , Receptores Opioides/metabolismo , Somatostatina/metabolismo
19.
Eur J Pharmacol ; 530(1-2): 33-9, 2006 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-16386243

RESUMO

Matrix metalloproteinases (MMP) are ubiquitous enzymes involved in extracellular matrix remodeling, and as a consequence in a number of physiological and pathological states, including development, wound healing and cancer. A crucial feature of cancer progression and metastasis is the disruption of extracellular matrix, and spreading of proliferating cancer cells. Modulation of MMP is a main target of cancer research. Using the mouse fibrosarcoma cell line WEHI 164, producing high amounts of MMP-2, we investigated whether we could modulate its production. We report that MMP-2 is under the control of nitric oxide (NO)/nitric oxide synthase (NOS) system. In addition, we show that NOS activity is controlled by opioids in a non-opioid receptor-related manner. Finally, we provide evidence that morphine, when administrated at low, non-toxic concentrations (<10(-9) M) attenuates MMP-2 activity. We conclude that, as morphine is able to decrease metalloproteinase activity via the NO/NOS system, it may have a place in the treatment of several sarcomas including fibrosarcoma.


Assuntos
Metaloproteinase 2 da Matriz/metabolismo , Morfina/farmacologia , Óxido Nítrico/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Diprenorfina/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Fibrossarcoma/metabolismo , Fibrossarcoma/patologia , Citometria de Fluxo , Inibidores de Metaloproteinases de Matriz , Camundongos , Camundongos Endogâmicos BALB C , NG-Nitroarginina Metil Éster/farmacologia , Antagonistas de Entorpecentes/farmacologia , Óxido Nítrico/antagonistas & inibidores , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Opioides/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , S-Nitroso-N-Acetilpenicilamina/farmacologia , Células Tumorais Cultivadas
20.
J Pharmacol Exp Ther ; 316(2): 900-4, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16207834

RESUMO

The pharmacological profile of opioid-binding sites in zebrafish brain homogenates has been studied using radiolabeled binding techniques. The nonselective antagonist [(3)H]diprenorphine binds with high affinity (K(D) = 0.27 +/- 0.08 nM and a B(max) = 212 +/- 14.3 fmol/mg protein), displaying two different binding sites with affinities of K(D1) = 0.08 +/- 0.02 nM and K(D2) = 17.8 +/- 9.18 nM. The nonselective agonist [(3)H]bremazocine also binds with high affinity to zebrafish brain membranes but only displays one single binding site with a K(D) = 1.1 +/- 0.09 nM and a B(max) = 705 +/- 19.3 fmol/mg protein. Competition binding assays using [(3)H]diprenorphine and several unlabeled ligands were performed. The synthetic selective agonists for mammalian opioid receptors DPDPE ([DPen(2),D-Pen(5)]-enkephalin), DAMGO ([D-Ala(2),NMe-Phe(4),Gly(5)-ol]-enkephalin), and U69,593 [(5alpha,7alpha,8beta)-(+)-N-methyl-N-[7-(1-pyrrolidinyl)-1-oxaspiro[4.5]dec-8-yl]-benzeneacetamide] failed to effectively displace [(3)H]diprenorphine binding, whereas nonselective ligands and the endogenous opioid peptides such as dynorphin A showed good affinities in the nanomolar range, although several of the endogenous peptides only displaced approximately 50% of the specifically bound [(3)H]diprenorphine. Our results provide evidence that, although the selective synthetic compounds for mammalian receptors do not fully recognize the opioid-binding sites in zebrafish brain, the activity of the endogenous zebrafish opioid system might not significantly differ from that displayed by the mammalian opioid system. Hence, the study of zebrafish opioid activity may contribute to an understanding of endogenous opioid systems in higher vertebrates.


Assuntos
Benzomorfanos/farmacologia , Ligação Competitiva , Encéfalo/metabolismo , Diprenorfina/farmacologia , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Feminino , Técnicas In Vitro , Ligantes , Masculino , Ensaio Radioligante , Peixe-Zebra
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...