Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 162
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Chem Biol Drug Des ; 101(6): 1382-1392, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36813756

RESUMO

6,7-Benzomorphans have been investigated in medicinal chemistry for developing new drugs. This nucleus could be considered a versatile scaffold. The physicochemical properties of benzomorphan N-substituent are crucial in achieving a definite pharmacological profile at opioid receptors. Thus, the dual-target MOR/DOR ligands LP1 and LP2 were obtained through N-substituent modifications. Specifically, LP2, bearing as N-substituent the (2R/S)-2-methoxy-2- phenylethyl group, is a dual-target MOR/DOR agonist and is successful in animal models of inflammatory and neuropathic pain. To obtain new opioid ligands, we focused on the design and synthesis of LP2 analogs. First, the 2-methoxyl group of LP2 was replaced by an ester or acid functional group. Then, spacers of different lengths were introduced at N-substituent. In-vitro, their affinity profile versus opioid receptors has been performed through competition binding assays. Molecular modeling studies were conducted to deeply analyze the binding mode and the interactions between the new ligands and all opioid receptors.


Assuntos
Receptores Opioides delta , Receptores Opioides mu , Animais , Receptores Opioides mu/metabolismo , Receptores Opioides delta/metabolismo , Benzomorfanos/metabolismo , Benzomorfanos/farmacologia , Ligantes , Receptores Opioides , Relação Estrutura-Atividade
2.
J Chem Inf Model ; 55(3): 614-27, 2015 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-25642595

RESUMO

Human kappa opioid receptor (κ-OR), a G protein-coupled receptor (GPCR), has been identified as a drug target for treatment of such human disorders as pain perception, neuroendocrine physiology, affective behavior, and cognition. In order to find more selective and active agonists, one would like to do structure based drug design. Indeed, there is an X-ray structure for an antagonist bound to κ-OR, but structures for activated GPCRs are quite different from those for the inactive GPCRs. Here we predict the ensemble of 24 low-energy structures of human kappa opioid receptor (κ-OR), obtained by application of the GEnSeMBLE (GPCR Ensemble of Structures in Membrane Bilayer Environment) complete sampling method, which evaluates 13 trillion combinations of tilt and rotation angles for κ-OR to select the best 24. To validate these structures, we used the DarwinDock complete sampling method to predict the binding sites for five known agonists (ethylketocyclazocine, bremazocine, pentazocine, nalorphine, and morphine) bound to all 24 κ-OR conformations. We find that some agonists bind selectively to receptor conformations that lack the salt bridge between transmembrane domains 3 and 6 as expected for active conformations. These 3D structures for κ-OR provide a structural basis for understanding ligand binding and activation of κ-OR, which should be useful for guiding subtype specific drug design.


Assuntos
Modelos Moleculares , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/química , Benzomorfanos/metabolismo , Benzomorfanos/farmacologia , Sítios de Ligação , Desenho de Fármacos , Etilcetociclazocina/metabolismo , Humanos , Ligantes , Simulação de Acoplamento Molecular , Morfina/metabolismo , Morfina/farmacologia , Nalorfina/metabolismo , Nalorfina/farmacologia , Pentazocina/metabolismo , Pentazocina/farmacologia , Conformação Proteica , Receptores Acoplados a Proteínas G/química , Receptores Opioides kappa/metabolismo , Relação Estrutura-Atividade
3.
Eur J Pharmacol ; 731: 1-7, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24657279

RESUMO

Several methodological approaches suggest that receptor heteromers exist in cell systems, but their presence in physiological tissue is widely contentious. We describe a novel method to determine if heterodimers exist in brain tissue sections using autoradiographic binding comparisons from single and double gene knockout mice, where tissues either have a full receptor complement and can form heterodimers, or are incapable of making heterodimers. We have tested this model, which we have named Knockout Subtraction Autoradiography, to determine if heterodimerisation of the kappa (KOP) and delta opioid (DOP) receptors occurs, as evidence from binding studies in cell systems suggest they are present in the brain. Using labeling of putative KOP receptor/DOP receptor heterodimers with either [(3)H]bremazocine or with [(3)H]naltrindole, two ligands which were used to provide evidence suggesting that these opioid receptor subtypes heterodimerize, we have applied a subtraction equation model based on the principle that receptor gene double knockout of either MOP receptor/KOP receptor (DOP receptor expression only) or MOP receptor/DOP receptor (KOP receptor expression only) produces tissue incapable of making the KOP receptor/DOP receptor heterodimer. We have shown in most brain regions that the labeling fits a simple additive model of monomer labeling, but that in a few brain regions opioid receptor heterodimerization does occur. The data does not support the conclusion that KOP receptor/DOP receptor heterodimerisation is widespread in the central nervous system, but does indicate that this novel methodology can detect heterodimerisation, when ligands with distinct binding affinities for monomer and heterodimer forms exist.


Assuntos
Autorradiografia/métodos , Encéfalo/metabolismo , Técnicas de Inativação de Genes , Multimerização Proteica , Receptores Opioides delta/química , Receptores Opioides kappa/química , Técnica de Subtração , Animais , Benzomorfanos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Naltrexona/análogos & derivados , Naltrexona/metabolismo , Estrutura Quaternária de Proteína , Receptores Opioides delta/deficiência , Receptores Opioides delta/genética , Receptores Opioides kappa/deficiência , Receptores Opioides kappa/genética
4.
Neuropharmacology ; 71: 70-82, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23541722

RESUMO

Persistent pain states, such as those caused by nerve injury or inflammation, are associated with altered sensations, allodynia and hyperalgesia, that are resistant to traditional analgesics. A contribution to development and maintenance in altered pain perception comes from nociceptive processing and descending modulation from supraspinal sites. A multitarget ligand seems to be useful for pain relief with a decreased risk of adverse events and a considerable analgesic efficacy. The multitarget MOR agonist-DOR antagonist LP1, (3-[(2R,6R,11R)-8-hydroxy-6,11-dimethyl-1,4,5,6-tetrahydro-2,6-methano-3-benazocin-3(2H)-yl]-N-phenylpropanamide, is a central acting antinociceptive agent with low potential to induce tolerance. LP1 was tested in models of neuropathic pain - induced by chronic constriction injury (CCI) of the left sciatic nerve - and inflammatory pain - produced by intraplantar injection of carrageenan. In CCI rats, subcutaneous (s.c.) LP1 (3 mg/kg) showed a significant antiallodynic effect, measured with von Frey filaments, and antihyperalgesic effect, evoked in response to a radiant heat stimulus with plantar test. Analogously, LP1 significantly reduced allodynic and hyperalgesic thresholds in a model of inflammatory pain induced by carrageenan. To evaluate the contribution of opioid receptor subtypes in LP1 antinociceptive effects, the multitarget LP1 profile was assessed using selective opioid antagonists. Moreover, functional electrophysiological in vitro assays, using primary cortical and spinal cord networks, allowed to define the "pharmacological fingerprint" of LP1. The EC50 values in this functional screening seem to confirm LP1 as a potent opioid ligand (EC50 = 0.35 fM and EC50 = 44 pM in spinal cord and frontal cortex, respectively). Using a NeuroProof data-base of well characterised reference compounds, a similarity profile of LP1 to opioid and non-opioid drugs involved in pain modulation was detected. Our studies seem to support that multitarget ligand approach should be useful for persistent pain conditions in which mechanical allodynia and thermal hyperalgesia are significant components of the nociceptive response.


Assuntos
Analgésicos Opioides/uso terapêutico , Benzomorfanos/uso terapêutico , Dor Crônica/tratamento farmacológico , Modelos Animais de Doenças , Proteínas do Tecido Nervoso/metabolismo , Neuralgia/tratamento farmacológico , Neurônios/efeitos dos fármacos , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacologia , Animais , Benzomorfanos/metabolismo , Benzomorfanos/farmacologia , Células Cultivadas , Dor Crônica/imunologia , Dor Crônica/metabolismo , Embrião de Mamíferos , Lobo Frontal/citologia , Lobo Frontal/efeitos dos fármacos , Lobo Frontal/imunologia , Lobo Frontal/metabolismo , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos , Proteínas do Tecido Nervoso/agonistas , Proteínas do Tecido Nervoso/antagonistas & inibidores , Neuralgia/imunologia , Neuralgia/metabolismo , Neurônios/citologia , Neurônios/imunologia , Neurônios/metabolismo , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides delta/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Medula Espinal/citologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/imunologia , Medula Espinal/metabolismo
5.
Life Sci ; 90(25-26): 957-61, 2012 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-22580287

RESUMO

AIMS: Opioid drugs are the principal treatment option for moderate to severe pain and exert their biological effects through interactions with opioid receptors that are widely distributed throughout the CNS and peripheral tissues. Ligands capable of simultaneously targeting different receptors could be successful candidates for the treatment of chronic pain. Enhanced antinociception coupled with a low incidence of side effects has been demonstrated for ligands possessing mixed mu-opioid receptor (MOR) and delta-opioid receptor (DOR) activity. We previously reported that 3-[(2R,6R,11R)-8-hydroxy-6,11-dimethyl-1,4,5,6-tetrahydro-2,6-methano-3-benzazocin-3(2H)-yl]-N-phenylpropanamide (LP1) acted as a MOR-DOR ligand in in vitro functional assays and moreover this drug produced a valid antinociception that was longer lasting than that of morphine. The aim of this work was to determine whether the antinociceptive effect produced by LP1 was central or peripheral and to assess which opioid receptor subtypes are involved in its effects. MAIN METHODS: We explored the effects of naloxone methiodide (NX-M), a quaternary opioid antagonist, administered either intracerebroventricularly (i.c.v.) or subcutaneously (s.c.), on LP1-mediated antinociception in male Sprague-Dawley rats. In addition, we administered s.c. selective antagonists for MOR, DOR and kappa-opioid receptor (KOR) to investigate the effects of LP1. To characterise this drug's DOR profile better, we also investigated the effects of LP1 on DPDPE, a selective DOR agonist. KEY FINDINGS: Data obtained by tail flick test showed that LP1 induced predominantly MOR-mediated supraspinal antinociception and was able to counteract DPDPE analgesia. SIGNIFICANCE: LP1, a multitarget opioid ligand, is a supraspinal acting antinociceptive agent that is useful for the treatment of chronic pain.


Assuntos
Analgésicos/administração & dosagem , Benzomorfanos/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Medição da Dor/efeitos dos fármacos , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Analgésicos/metabolismo , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/metabolismo , Animais , Benzomorfanos/metabolismo , Ligantes , Masculino , Medição da Dor/métodos , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Reprodutibilidade dos Testes
6.
Life Sci ; 90(1-2): 66-70, 2012 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-22100511

RESUMO

AIMS: Powerful analgesics relieve pain primarily through activating mu opioid receptor (MOR), but the long-term use of MOR agonists, such as morphine, is limited by the rapid development of tolerance. Recently, it has been observed that simultaneous stimulation of the delta opioid receptor (DOR) and MOR limits the incidence of tolerance induced by MOR agonists. 3-[(2R,6R,11R)-8-hydroxy-6,11-dimethyl-1,4,5,6-tetrahydro-2,6-methano-3-benzazocin-3(2H)-yl]-N-phenylpropanamide (LP1) is a centrally acting agent with antinociceptive activity comparable to morphine and is able to bind and activate MOR and DOR. The aim of this work was to evaluate and compare the induction of tolerance to antinociceptive effects from treatment with LP1 and morphine. MAIN METHODS: Here, we evaluated the pharmacological effects of LP1 administered at a dose of 4 mg/kg subcutaneously (s.c.) twice per day for 9 days to male Sprague-Dawley rats. In addition, the LP1 mechanism of action was assessed by measurement of LP1-induced [(35)S]GTPγS binding to the MOR and DOR. KEY FINDINGS: Data obtained from the radiant heat tail flick test showed that LP1 maintained its antinociceptive profile until the ninth day, while tolerance to morphine (10mg/kg s.c. twice per day) was observed on day 3. Moreover, LP1 significantly enhanced [(35)S]GTPγS binding in the membranes of HEK293 cells expressing either the MOR or the DOR. SIGNIFICANCE: LP1 is a novel analgesic agent for chronic pain treatment, and its low tolerance-inducing capability may be correlated with its ability to bind both the MOR and DOR.


Assuntos
Analgésicos Opioides/uso terapêutico , Benzomorfanos/uso terapêutico , Dor Crônica/tratamento farmacológico , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacologia , Animais , Benzomorfanos/metabolismo , Benzomorfanos/farmacologia , Tolerância a Medicamentos/fisiologia , Células HEK293 , Humanos , Ligantes , Masculino , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Resultado do Tratamento
7.
Eur J Pharmacol ; 485(1-3): 119-25, 2004 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-14757131

RESUMO

Using Chinese Hamster Ovary cell membranes that stably expressed the human kappa-opioid receptor, we investigated the hypothesis that kappa(1)- and kappa(2)-opioid receptors, historically defined by their pharmacological selectivity for either arylacetamides or benzomorphans are, in fact, different affinity states or binding sites on the same kappa-opioid receptors. Receptor binding studies showed that GTP gamma S potently inhibited [3H](5 alpha,7 alpha,8 beta)-(+)-N-methyl-N-(7-[1-pyrrolidinyl]-1-oxaspiro [4.5]dec-8-yl)-benzeneacetamide (U69,593) binding, compared to virtually no inhibition of [3H]bremazocine binding. Saturation binding experiments showed a three-fold decrease in [3H]U69,593 affinity in the presence of GTP gamma S, but GTP gamma S had no effect on [3H]bremazocine affinity. The kappa-opioid receptor antagonist nor-binaltorphimine had a four-fold higher affinity for [3H]U69,593-labeled receptors than for [3H]bremazocine-labeled receptors. Functional selectivity studies, measuring the stimulation of [35S]GTP gamma S agonist-induced binding, showed a significantly higher U69,593-induced G protein-receptor activation in comparison to the stimulation observed with bremazocine. These results suggest that pharmacologically defined 1 kappa-opioid receptor subtypes may be different affinity states of the same receptor.


Assuntos
Acetamidas/metabolismo , Benzomorfanos/metabolismo , Receptores Opioides kappa/metabolismo , Acetamidas/farmacologia , Animais , Benzenoacetamidas/metabolismo , Benzenoacetamidas/farmacologia , Benzomorfanos/farmacologia , Células CHO , Cricetinae , Relação Dose-Resposta a Droga , Humanos , Ligação Proteica/fisiologia , Pirrolidinas/metabolismo , Pirrolidinas/farmacologia , Receptores Opioides kappa/agonistas
8.
Brain Res Mol Brain Res ; 118(1-2): 119-31, 2003 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-14559361

RESUMO

A mouse mu opioid receptor was engineered to contain a FLAG epitope at the amino-terminus and a hexahistidine tag at the carboxyl-terminus to facilitate purification. Selection of transfected human embryonic kidney (HEK) 293 cells yielded a cell line that expressed the receptor with a B(max) of 10 pmol/mg protein. 3[H]Bremazocine exhibited high affinity binding to the epitope-tagged mu opioid receptor with a KD of 1.0 nM. The agonists [D-Ala(2),N-Me-Phe(4),Gly(5)-ol]enkephalin (DAMGO), morphine and [D-Ala(2),D-Leu(5)]enkephalin (DADL) competitively inhibited bremazocine binding to the tagged mu receptor with KI's of 3.5, 17 and 70 nM, respectively. Chronic treatment of cells expressing the epitope-tagged mu receptor with DAMGO resulted in down-regulation of the receptor, indicating that the tagged receptor retained the capacity to mediate signal transduction. The mu receptor was solubilized from HEK 293 cell membranes with n-dodecyl-beta-D-maltoside in an active form that maintained high affinity bremazocine binding. Sequential use of wheat germ agglutinin (WGA)-agarose chromatography, Sephacryl S300 gel filtration chromatography, immobilized metal affinity chromatography, immunoaffinity chromatography, and sodium dodecyl sulfate/polyacrylamide gel electrophoresis (SDS/PAGE) permitted purification of the receptor. The purified mu opioid receptor was a glycoprotein that migrated on SDS/PAGE with an apparent molecular mass of 80 kDa. Matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) mass spectrometry was used to identify and characterize peptides derived from the mu opioid receptor following in-gel digestion with trypsin or chymotrypsin, and precursor-derived tandem mass spectrometry (ms/ms) confirmed the identity of several peptides derived from enzymatic digestion of the mu opioid receptor.


Assuntos
Membrana Celular/metabolismo , Receptores Opioides mu/química , Receptores Opioides mu/isolamento & purificação , Analgésicos/metabolismo , Analgésicos/farmacologia , Benzomorfanos/metabolismo , Benzomorfanos/farmacologia , Ligação Competitiva/efeitos dos fármacos , Ligação Competitiva/fisiologia , Linhagem Celular , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Epitopos , Humanos , Ligantes , Espectrometria de Massas , Entorpecentes/metabolismo , Entorpecentes/farmacologia , Peptídeos/química , Peptídeos/isolamento & purificação , Receptores Opioides mu/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Solubilidade
9.
Brain Res ; 945(1): 9-19, 2002 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-12113946

RESUMO

Using quantitative receptor autoradiography we have determined if deletion of the delta-opioid receptor gene (Oprd1) results in compensatory changes in the expression of other opioid receptors. Gene targeting was used to delete exon 1 of the mouse delta-opioid receptor gene and autoradiography was carried out on brains from wild-type, heterozygous and homozygous knockout mice. Delta-opioid receptors were labeled with [(3)H]deltorphin I (7 nM), mu- with [(3)H]DAMGO (4 nM), and kappa- with [(3)H]CI-977 (2.5 nM) or [(3)H]bremazocine (2 nM in the presence of DPDPE and DAMGO) and non-specific binding determined with naloxone. [(3)H]Deltorphin I binding was reduced by approximately 50% in heterozygous animals. In homozygous animals specific binding could only be detected after long-term film exposure (12 weeks). Regions exhibiting this residual [(3)H]deltorphin I binding correlated significantly with those demonstrating high levels of the mu-receptor and were abolished in the presence of the mu-agonist DAMGO. Autoradiographic mapping showed significant overall reductions in [(3)H]DAMGO and [(3)H]CI-977 binding throughout the brain following loss of both copies of the Oprd1 gene. In contrast, overall levels of [(3)H]bremazocine binding were higher in brains from -/- than +/+ mice. Our findings suggest that residual [(3)H]deltorphin I binding in the brain of delta-receptor gene knockout mice is the result of cross-reactivity with mu-sites and that there are no delta-receptor subtypes derived from a different gene. Changes in mu- and kappa-receptor labeling suggest compensatory changes in these subtypes in response to the absence of the delta-receptor. The differences in [(3)H]CI-977 and [(3)H]bremazocine binding indicate these ligands show differential recognition of the kappa-receptor.


Assuntos
Encéfalo/metabolismo , Camundongos Knockout/genética , Camundongos Knockout/metabolismo , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Animais , Autorradiografia , Benzofuranos/metabolismo , Benzomorfanos/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Feminino , Heterozigoto , Homozigoto , Masculino , Camundongos , Oligopeptídeos/metabolismo , Pirrolidinas/metabolismo , Receptores Opioides mu/agonistas
10.
Regul Pept ; 105(1): 9-14, 2002 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-11853866

RESUMO

We reported recently that the ubiquitin-proteasome pathway is involved in agonist-induced down regulation of mu and delta opioid receptors [J. Biol. Chem. 276 (2001) 12345]. While evaluating the effects of various protease inhibitors on agonist-induced opioid receptor down regulation, we observed that while the peptide aldehyde, leupeptin (acetyl-L-Leucyl-L-Leucyl-L-Arginal), did not affect agonist-induced down regulation, leupeptin at submillimolar concentrations directly inhibited radioligand binding to opioid receptors. In this study, the inhibitory activity of leupeptin on radioligand binding was characterized utilizing human embryonic kidney (HEK) 293 cell lines expressing transfected mu, delta, or kappa opioid receptors. The rank order of potency for leupeptin inhibition of [3H]bremazocine binding to opioid receptors was mu > delta > kappa. In contrast to the effect of leupeptin, the peptide aldehyde proteasome inhibitor, MG 132 (carbobenzoxy-L-Leucyl-L-Leucyl-L-Leucinal), had significantly less effect on bremazocine binding to mu, delta, or kappa opioid receptors. We propose that leupeptin inhibits ligand binding by reacting reversibly with essential sulfhydryl groups that are necessary for high-affinity ligand/receptor interactions.


Assuntos
Inibidores de Cisteína Proteinase/farmacologia , Leupeptinas/farmacologia , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides delta/metabolismo , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/metabolismo , Benzomorfanos/antagonistas & inibidores , Benzomorfanos/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Humanos , Ligantes , Ligação Proteica/efeitos dos fármacos , Receptores Opioides kappa/metabolismo
11.
Eur J Pharmacol ; 414(2-3): 189-95, 2001 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-11239918

RESUMO

Despite ample pharmacological evidence for the existence of multiple mu-, delta- and kappa-opioid receptor subtypes, only three genes encoding mu-(MOR), delta-(DOR) and kappa-(KOR) opioid receptor have been cloned. The KOR gene encodes kappa(1)-sites, which specifically bind arylacetamide compounds, and the possible existence of kappa-opioid receptor subtypes derived from another kappa-opioid-receptor gene, yet to be characterized, remains a very contentious issue. kappa(2)-Opioid receptors are described as binding sites typically labelled by the non-selective benzomorphan ligand [3H]bremazocine in the presence of mu-, delta- and kappa(1)-opioid receptor blocking ligands. To investigate the genetic origin of kappa(2)-opioid receptors, we have carried out homogenate binding experiments with [3H]bremazocine in brains of single MOR-, DOR-, KOR- and double MOR/DOR-deficient mice. Scatchard analysis showed that 68+/-12% of the binding sites arise from the MOR gene, 27+/-1% from the DOR gene and 14.5+/-0.2% from the KOR gene, indicating that the three known genes account for total [3H]bremazocine binding. Experiments in the presence of mu-, delta- and kappa(1)-opioid receptor suppressor ligands further showed that non-kappa(1)-opioid receptor labelling can be accounted for by binding to both the mu- and delta-opioid receptors. Finally, [3H]bremazocine binding experiments performed on brain membranes from the triple MOR/DOR/KOR-deficient mice revealed a complete absence of binding sites, confirming definitively that no additional gene is required to explain the total population of [3H]bremazocine binding sites. Altogether the data show that the putative kappa(2)-opioid receptors are in fact a mixed population of KOR, DOR and predominantly MOR gene products.


Assuntos
Analgésicos/metabolismo , Benzomorfanos/metabolismo , Encéfalo/metabolismo , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Animais , Camundongos , Camundongos Knockout , Receptores Opioides delta/deficiência , Receptores Opioides kappa/deficiência , Receptores Opioides mu/deficiência
12.
Biochemistry ; 39(45): 13904-15, 2000 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-11076532

RESUMO

Binding pockets of the opioid receptors are presumably formed among the transmembrane domains (TMDs) and are accessible from the extracellular medium. In this study, we determined the sensitivity of binding of [(3)H]diprenorphine, an antagonist, to mu, delta, and kappa opioid receptors to charged methanethiosulfonate (MTS) derivatives and identified the cysteine residues within the TMDs that conferred the sensitivity. Incubation of the mu opioid receptor expressed in HEK293 cells with MTS ethylammonium (MTSEA), MTS ethyltrimethylammonium (MTSET), or MTS ethylsulfonate (MTSES) inhibited [(3)H]diprenorphine binding with the potency order of MTSEA > MTSET > MTSES. Pretreatment of mu, delta, and kappa opioid receptors with MTSEA dose-dependently inhibited [(3)H]diprenorphine binding with MTSEA sensitivity in the order of kappa > mu >> delta. The effects of MTSEA occurred rapidly, reaching the maximal inhibition in 10 min. (-)-Naloxone, but not (+)-naloxone, prevented the MTSEA effect, demonstrating that the reaction occurs within or in the vicinity of the binding pockets. Each cysteine residue in the TMDs of the three receptors was mutated singly, and the effects of MTSEA treatment were examined. The mutants had similar affinities for [(3)H]diprenorphine, and C7. 38(321)S, C7.38(303)S, and C7.38(315)S mutations rendered mu, delta, and kappa opioid receptors less sensitive to the effect of MTSEA, respectively. These results indicate that the conserved Cys7.38 is differentially accessible in the binding-site crevice of these receptors. The second extracellular loop of the kappa receptor, which contains several acidic residues, appears to play a role, albeit small, in its higher sensitivity to MTSEA, whereas the negative charge of Glu6.58(297) did not. To the best of our knowledge, this is the first report to show that a conserved residue among highly homologous G protein-coupled receptors is differentially accessible in the binding-site crevice. In addition, this represents the first successful generation of MTSEA-insensitive mutants of mu, delta, and kappa opioid receptors, which will allow determination of residues accessible in the binding-site crevices of these receptors by the substituted cysteine accessibility method.


Assuntos
Sequência Conservada , Cisteína/metabolismo , Receptores Opioides/metabolismo , Sequência de Aminoácidos , Animais , Benzomorfanos/metabolismo , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/genética , Linhagem Celular , Sequência Conservada/efeitos dos fármacos , Cisteína/genética , Diprenorfina/antagonistas & inibidores , Diprenorfina/metabolismo , Relação Dose-Resposta a Droga , Metanossulfonato de Etila/análogos & derivados , Metanossulfonato de Etila/farmacologia , Ácido Glutâmico/genética , Ácido Glutâmico/metabolismo , Humanos , Indicadores e Reagentes , Mesilatos/farmacologia , Metionina/metabolismo , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Naloxona/farmacologia , Antagonistas de Entorpecentes , Estrutura Secundária de Proteína/efeitos dos fármacos , Ratos , Receptores Opioides/genética , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/antagonistas & inibidores , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Serina/metabolismo , Fatores de Tempo , Trítio
13.
Brain Res Mol Brain Res ; 80(2): 166-76, 2000 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11038249

RESUMO

Several non-peptidic opioids have been synthesized recently as part of a program to develop selective delta receptor agonists. In this study, the affinities of a set of compounds for cloned delta and mu opioid receptors expressed in HEK 293 cell lines were determined by competition analysis of [3H]bremazocine binding to membrane preparations. All compounds studied exhibited high affinity and selectivity, with apparent dissociation constants in the range of 0.6-1.7 nM for the delta opioid receptor and 240-1165 nM for the mu opioid receptor. We next sought to determine which domain of the delta receptor was critical for mediating the highly selective binding by analysis of ligand affinities for mu/delta receptor chimeras. Receptor binding profiles suggested that a critical site of receptor/ligand interaction was located between transmembrane domain 5 (TM5) and TM7 of the delta receptor. Substitution of tryptophan 284, located at the extracellular surface of TM6, with lysine, which is found at the equivalent position in the mu opioid receptor, led to a spectrum of effects on affinities, depending on the ligand tested. Affinities of SB 219825 and SB 222941 were particularly sensitive to the substitution, displaying a 50-fold and 70-fold decrease in affinity, respectively. Activities of the delta receptor-selective agonists were tested in two functional assays. Brief exposure of HEK 293 cells expressing delta opioid receptors with selective ligands induced phosphorylation of MAP kinase, although the non-peptidic ligands were less efficacious than the enkephalin derivative DADL (Tyr-D-Ala-Gly-Phe-D-Leu). Similarly, chronic exposure of HEK 293 cells expressing delta opioid receptors with selective, non-peptidic ligands, with the exception of SB 206848, caused receptor down-regulation, however, the SB compounds were less efficacious than DADL.


Assuntos
Receptores Opioides delta , Sequência de Aminoácidos , Analgésicos/metabolismo , Analgésicos/farmacologia , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacologia , Benzomorfanos/metabolismo , Benzomorfanos/farmacologia , Ligação Competitiva , Células Cultivadas , Clonagem Molecular , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Leucina Encefalina-2-Alanina/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Compostos Heterocíclicos de 4 ou mais Anéis/química , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Indóis/química , Indóis/farmacologia , Isoquinolinas/química , Isoquinolinas/farmacologia , Rim/citologia , Ligantes , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Dados de Sequência Molecular , Morfina/metabolismo , Morfina/farmacologia , Mutagênese Sítio-Dirigida , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Quinolinas/química , Quinolinas/metabolismo , Quinolinas/farmacologia , Ensaio Radioligante , Receptores Opioides delta/agonistas , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides delta/genética , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/genética , Trítio
14.
Eur J Pharmacol ; 403(1-2): 37-44, 2000 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10969141

RESUMO

We hypothesized that the selectivity profile of the rat mu-opioid receptor for opioid receptor-selective ligands is determined by the nature of the amino acid residues at highly divergent sites in the ligand-binding pocket. To determine which characteristics of these residues contribute to opioid receptor ligand selectivity, we made various mutant receptors that replaced the Lys(303) and Trp(318) residues near the extracellular interface of transmembrane domains VI and VII, respectively. Ligand binding determinations using transiently transfected monkey kidney epithelial (COS-1) cells show that Lys(303) mutations cause little change in the receptor binding profile, whereas the Trp(318) mutant receptors have considerably lower affinity for micro-opioid receptor-selective ligands and greatly increased affinity for delta-opioid receptor-selective ligands. The nature of these mutations show that this effect is not due to sterics or charge alone. [35S]guanosine-5'-O-(3-thio)-triphosphate ([35S]GTPgammaS) activity assays show that these residues may influence functional, as well as binding selection. We conclude that a primary role for Trp(318) is to form a basis for ligand selectivity.


Assuntos
Naloxona/análogos & derivados , Naltrexona/análogos & derivados , Receptores Opioides mu/metabolismo , Substituição de Aminoácidos , Animais , Benzamidas/metabolismo , Benzamidas/farmacologia , Benzomorfanos/metabolismo , Benzomorfanos/farmacologia , Sítios de Ligação/genética , Ligação Competitiva/efeitos dos fármacos , Células COS , Ala(2)-MePhe(4)-Gly(5)-Encefalina/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , D-Penicilina (2,5)-Encefalina/metabolismo , D-Penicilina (2,5)-Encefalina/farmacologia , Fentanila/metabolismo , Fentanila/farmacologia , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Ligantes , Morfina/metabolismo , Morfina/farmacologia , Mutação , Naloxona/metabolismo , Naloxona/farmacologia , Naltrexona/metabolismo , Naltrexona/farmacologia , Antagonistas de Entorpecentes/metabolismo , Antagonistas de Entorpecentes/farmacologia , Peptídeos/metabolismo , Peptídeos/farmacologia , Piperazinas/metabolismo , Piperazinas/farmacologia , Conformação Proteica , Ensaio Radioligante , Ratos , Receptores Opioides mu/química , Receptores Opioides mu/genética , Transdução de Sinais , Radioisótopos de Enxofre
15.
Arch Biochem Biophys ; 375(1): 31-49, 2000 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-10683246

RESUMO

Metal ions affect ligand binding to G-protein-coupled receptors by as yet unknown mechanisms. In particular, Na(+) increases the affinity for antagonists but decreases it for agonists. We had modeled the mu-opioid receptor (muR) based on the low-resolution structure of rhodopsin by G. F. X. Schertler, C. Villa, and R. Henderson (1993, Nature 362, 770-772) and proposed that metal ions may be directly involved in the binding of ligands and receptor activation (B. S. Zhorov and V. S. Ananthanarayanan, 1998, J. Biomol. Struct. Dyn. 15, 631-637). Developing this concept further, we present here homology models of muR using as templates the structure of rhodopsin elaborated by I. D. Pogozheva, A. L. Lomize, and H. I. Mosberg (1997, Biophys. J. 70, 1963-1985) and J. M. Baldwin, G. F. X. Schertler, and V. M. Unger (1997, J. Mol. Biol., 272, 144-164). Using the Monte Carlo minimization (MCM) method, we docked the Na(+)-bound forms of muR ligands: naloxone, bremazocine, and carfentanyl. The resultant low-energy complexes showed that the two positive charges in the protonated metal-bound ligands interact with the two negative charges at Asp(3.32) and Asp(2.50) (for notations, see J. A. Ballesteros and H. Weinstein, 1995, Methods Neurosci. 25, 366-426). MCM computation on morphine docked inside the model of muR by I. D. Pogozheva, A. L. Lomize, and H. I. Mosberg (1998, Biophys. J. 75, 612-634) yielded two binding modes with the ligand's ammonium group salt-bridged either to Asp(3.32) (generally regarded as the ligand recognition site) or to Asp(2.50). The latter is the presumed site for Na(+) ion, which is known to modulate ligand binding. Assuming that in the low-dielectric transmembrane region of muR, organic and inorganic cations would compete for Asp(3.32) and Asp(2.50), we propose that ligand binding, as visualized in the above models, would first displace Na(+) from Asp(3.32). A subsequent progress of the ligand toward Asp(2.50) would result in either the retention of Na(+) at Asp(2.50) in the case of antagonists or the displacement of Na(+) from Asp(2.50) in the case of agonists. The displaced Na(+) would move toward the salt-bridged Asp(3.49)-Arg(3.50) and disengage the salt bridge. This, in turn, would result in conformational changes at the cytoplasmic face of the receptor that facilitate the interaction with the G-protein.


Assuntos
Ácido Aspártico/metabolismo , Modelos Moleculares , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Sequência de Aminoácidos , Analgésicos Opioides/metabolismo , Animais , Benzomorfanos/metabolismo , Sítios de Ligação/genética , Sítios de Ligação/fisiologia , Cátions/metabolismo , Bovinos , Simulação por Computador , Sequência Conservada , Fentanila/análogos & derivados , Fentanila/metabolismo , Humanos , Ligantes , Metais/metabolismo , Dados de Sequência Molecular , Morfina/metabolismo , Naloxona/metabolismo , Antagonistas de Entorpecentes/metabolismo , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína/genética , Ratos , Receptores Opioides mu/genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade
16.
J Med Chem ; 43(1): 114-22, 2000 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-10633042

RESUMO

This report concerns the synthesis and preliminary pharmacological evaluation of a novel series of kappa agonists related to the morphinan (-)-cyclorphan (3a) and the benzomorphan (-)-cyclazocine (2) as potential agents for the pharmacotherapy of cocaine abuse. Recent evidence suggests that agonists acting at kappa opioid receptors may modulate the activity of dopaminergic neurons and alter the neurochemical and behavioral effects of cocaine. We describe the synthesis and chemical characterization of a series of morphinans 3a-c, structural analogues of cyclorphan [(-)-3-hydroxy-N-cyclopropylmethylmorphinan S(+)-mandelate, 3a], the 10-ketomorphinans 4a,b, and the 8-ketobenzomorphan 1b. Binding experiments demonstrated that the cyclobutyl analogue 3b [(-)-3-hydroxy-N-cyclobutylmethylmorphinan S(+)-mandelate, 3b, MCL-101] of cyclorphan (3a) had a high affinity for mu, delta, and kappa opioid receptors in guinea pig brain membranes. Both 3a,b were approximately 2-fold more selective for the kappa receptor than for the mu receptor. However 3b (the cyclobutyl analogue) was 18-fold more selective for the kappa receptor in comparison to the delta receptor, while cyclorphan (3a) had only 4-fold greater affinity for the kappa receptor in comparison to the delta receptor. These findings were confirmed in the antinociceptive tests (tail-flick and acetic acid writhing) in mice, which demonstrated that cyclorphan (3a) produced antinociception that was mediated by the delta receptor while 3b did not produce agonist or antagonist effects at the delta receptor. Both 3a,b had comparable kappa agonist properties. 3a,b had opposing effects at the mu receptor: 3b was a mu agonist whereas 3a was a mu antagonist.


Assuntos
Benzomorfanos/síntese química , Morfinanos/síntese química , Receptores Opioides kappa/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos , Ácido Acético , Animais , Benzomorfanos/metabolismo , Benzomorfanos/farmacologia , Encéfalo/metabolismo , Relação Dose-Resposta a Droga , Etilcetociclazocina/análogos & derivados , Etilcetociclazocina/farmacologia , Cobaias , Técnicas In Vitro , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos ICR , Morfinanos/metabolismo , Morfinanos/farmacologia , Morfina/antagonistas & inibidores , Antagonistas de Entorpecentes/síntese química , Antagonistas de Entorpecentes/farmacologia , Dor/induzido quimicamente , Dor/tratamento farmacológico , Medição da Dor , Tempo de Reação/efeitos dos fármacos , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inibidores , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/metabolismo
17.
J Med Chem ; 43(26): 5030-6, 2000 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-11150174

RESUMO

Two of the synthesized (-)-(1R,5R,9R)-N-homologues (N-but-3-enyl- and N-but-3-ynyl-5,9-dimethyl-2'-hydroxy-6,7-benzomorphan (9, 13)) were found to be about 20 times more potent than morphine in the mouse tail-flick assay (ED(50) = 0.05 mg/kg), and (-)-(1R,5R, 9R)-N-but-2-ynyl-5,9-dimethyl-2'-hydroxy-6,7-benzomorphan ((-)-(1R, 5R,9R)-N-but-2-ynylnormetazocine, 12) was about as potent as the opioid antagonist N-allylnormetazocine (AD(50) in the tail-flick vs morphine assay = 0.3 mg/kg). All of the homologues examined had higher affinity for the kappa-opioid receptor than the mu-receptor except (-)-N-but-2-ynyl-normetazocine (12), which had a kappa/mu ratio = 7.8 and a delta/mu ratio = 118. The (-)-N-2-cyanoethyl (3), -allyl (8), and -but-3-ynyl (13) analogues had good affinity (<10 nM) for delta-opioid receptors. Two homologues in the (+)-(1S,5S,9S)-normetazocine series, N-pent-4-enyl (24) and N-hex-5-enyl (25), were high-affinity and selective sigma(1)-ligands (K(i) = 2 nM, sigma(2)/sigma(1) = 1250, and 1 nM, sigma(2)/sigma(1) = 750, respectively); in contrast, N-allylnormetazocine (22) had relatively poor affinity at sigma(1), and its sigma(1)/sigma(2) ratio was <100.


Assuntos
Benzomorfanos/síntese química , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Analgésicos/síntese química , Analgésicos/química , Analgésicos/metabolismo , Analgésicos/farmacologia , Animais , Benzomorfanos/química , Benzomorfanos/metabolismo , Benzomorfanos/farmacologia , Ligação Competitiva , Córtex Cerebral/metabolismo , Ligantes , Macaca mulatta , Camundongos , Morfina/farmacologia , Dependência de Morfina , Antagonistas de Entorpecentes/síntese química , Antagonistas de Entorpecentes/química , Antagonistas de Entorpecentes/metabolismo , Antagonistas de Entorpecentes/farmacologia , Medição da Dor , Ensaio Radioligante , Receptores Opioides delta/agonistas , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Estereoisomerismo , Relação Estrutura-Atividade , Síndrome de Abstinência a Substâncias/tratamento farmacológico
18.
J Med Chem ; 42(22): 4621-9, 1999 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-10579824

RESUMO

A convenient enantioselective synthesis of (1R,5R,9R)- and (1S,5S, 9S)-9-alkyl-2-benzyl-6,7-benzomorphans (2a-c) which starts with naphthaldehyde is described. These compounds were designed to gain additional information on the structure-sigma binding relationship of the 6,7-benzomorphan class of sigma ligands. In contrast to pentazocine and most 6,7-benzomorphans, the (1R,5R,9R)-isomers of 2a-c showed greater affinity for the sigma(1) receptor than the (1S, 5S,9S)-isomers. Despite reversal of enantioselectivity at the sigma(1) sites, moderate affinity and enantioselectivity at the sigma(2) sites [greater affinity for (1R,5R,9R)-isomers than (1S,5S, 9S)-isomers] were maintained. A comparison of the binding affinities of 2a-c to the more conformationally flexible trans-2-alkyl-1-benzaminoethyl-1,2-dihydronaphthalenes (10a-c) suggested that the relatively rigid structure of 2a-c played an important part in their sigma(1) binding properties. These compounds, particularly (1R,5R,9R)-2-benzyl-9-methyl-6,7-benzomorphan [(-)-2a], which has a K(i) value of 0.96 nM, will be useful in further characterization of the sigma(1) receptor.


Assuntos
Benzomorfanos/síntese química , Receptores sigma/metabolismo , Animais , Benzomorfanos/química , Benzomorfanos/metabolismo , Encéfalo/metabolismo , Cobaias , Técnicas In Vitro , Ligantes , Fígado/metabolismo , Ensaio Radioligante , Ratos , Estereoisomerismo , Relação Estrutura-Atividade
19.
Brain Res ; 827(1-2): 46-50, 1999 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-10320692

RESUMO

Desglycinyl remacemide (DGR), a biologically active metabolite of remacemide, was radiolabeled in an attempt to develop a ligand binding assay to identify its site of action. Incubation of the radioligand with membranes obtained from P2 fractions of whole rat brain revealed a single population of specific [3H]-DGR binding sites having a Kd of 290 nM and a Bmax of 1.3 pmole/mg protein. The specific binding of [3H]-DGR is most enriched in the P2 subcellular fraction and is heterogeneously distributed throughout the brain. The binding of [3H]-DGR to rat brain membranes was inhibited most potently by MK-801 and SKF-10,047. In contrast, haloperidol, and other sigma receptor-active agents, were relatively inactive at this site. These data suggest that DGR interacts with a channel blocking site on the NMDA receptor.


Assuntos
Benzomorfanos/metabolismo , Antagonistas de Aminoácidos Excitatórios/metabolismo , Fenetilaminas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Antipsicóticos/metabolismo , Antipsicóticos/farmacologia , Benzomorfanos/química , Sítios de Ligação/fisiologia , Química Encefálica/efeitos dos fármacos , Maleato de Dizocilpina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Haloperidol/metabolismo , Haloperidol/farmacologia , Masculino , Proteínas de Membrana/metabolismo , Neurônios/química , Neurônios/metabolismo , Fenazocina/análogos & derivados , Fenazocina/farmacologia , Fenciclidina/farmacologia , Fenetilaminas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/química , Frações Subcelulares/química , Frações Subcelulares/metabolismo , Trítio
20.
Pharmacology ; 58(4): 190-9, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10077738

RESUMO

In addition to effects in the periphery through inhibition of prostaglandin synthesis, several lines of evidence suggest that nonsteroidal anti-inflammatory drugs (NSAIDs) act in the central nervous system. The possibility that the central action of NSAIDs involves regulation of opioid receptors was investigated by quantitative autoradiography of mu, delta, and kappa sites in rat brain slices. Increased (p < 0.05) labeling of mu receptors was observed in thalamic nuclei, gyrus dentate, and layers of the parietal cortex of rats treated for 10 days with lysine clonixinate. Labeling of delta receptors was lower in the lateral septum, and kappa sites decreased in thalamic nuclei. These effects were not mediated through direct interaction with opioid-binding sites, since receptor-binding assays using rat brain membranes confirmed that clonixinate up to 1 x 10(-4) mol/l does not inhibit mu, delta, and kappa receptor specific binding. Central effects of NSAIDs might, therefore, involve interaction with the opioid receptor system through indirect mechanisms.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Encéfalo/efeitos dos fármacos , Clonixina/análogos & derivados , Lisina/análogos & derivados , Receptores Opioides/efeitos dos fármacos , Analgésicos/farmacologia , Animais , Benzomorfanos/metabolismo , Benzomorfanos/farmacologia , Sítios de Ligação , Ligação Competitiva/efeitos dos fármacos , Encéfalo/metabolismo , Clonixina/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina , D-Penicilina (2,5)-Encefalina , Encefalinas/metabolismo , Encefalinas/farmacologia , Lisina/farmacologia , Masculino , Ensaio Radioligante , Ratos , Ratos Wistar , Receptores Opioides/metabolismo , Receptores Opioides delta/agonistas , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Sensibilidade e Especificidade , Trítio
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...