Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Med Chem ; 58(15): 5728-41, 2015 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-26125201

RESUMO

Glycopeptides related to ß-endorphin penetrate the blood-brain barrier (BBB) of mice to produce antinociception. Two series of glycopeptides were assessed for opioid receptor binding affinity. Attempts to alter the mu-selectivity of [D-Ala(2),N-MePhe(4),Gly-ol(5)]enkephalin (DAMGO)-related glycopeptides by altering the charged residues of the amphipathic helical address were unsuccessful. A series of pan-agonists was evaluated for antinociceptive activity (55 °C tail flick) in mice. A flexible linker was required to maintain antinociceptive activity. Circular dichroism (CD) in H2O, trifluoroethanol (TFE), and SDS micelles confirmed the importance of the amphipathic helices (11s → 11sG → 11) for antinociception. The glycosylated analogues showed only nascent helices and random coil conformations in H2O. Chemical shift indices (CSI) and nuclear Overhauser effects (NOE) with 600 MHz NMR and CD confirmed helical structures in micelles, which were rationalized by molecular dynamics calculations. Antinociceptive studies with mice confirm that these glycosylated endorphin analogues are potential drug candidates that penetrate the BBB to produce potent central effects.


Assuntos
Sistema Nervoso Central/efeitos dos fármacos , Glicopeptídeos/farmacologia , Peptídeos Opioides/farmacologia , Sequência de Aminoácidos , Animais , Dicroísmo Circular , Glicopeptídeos/química , Espectroscopia de Ressonância Magnética , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos Opioides/química , Conformação Proteica , Receptores Opioides mu/efeitos dos fármacos
2.
Chem Biol Drug Des ; 78(5): 749-56, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21801311

RESUMO

Phosphorylation of l-serine-containing enkephalin analogs has been explored as an alternative to glycosylation in an effort to increase blood-brain barrier permeability and CNS bioavailability of peptide pharmacophores. Two enkephalin-based peptides were modified for these studies, a set related to DTLES, a mixed µ/δ-agonist, and one related to DAMGO, a highly selective µ-agonist. Each unglycosylated peptide was compared to its phosphate, its mono-benzylphosphate ester, and its ß-d-glucoside. Binding was characterized in membrane preparations from Chinese hamster ovary cells expressing human µ, δ and κ-opiate receptors. Antinociception was measured in mice using the 55 °C tail-flick assay. To estimate bioavailability, the antinociceptive effect of each opioid agonist was evaluated after intracerebroventricular (i.c.v.) or intravenous administration (i.v.) of the peptides. Circular dichroism methods and high-field nuclear magnetic resonance were used in the presence and absence of sodium dodecylsulfate to understand how the presence of a membrane might influence the peptide conformations.


Assuntos
Dicroísmo Circular , Encefalinas/química , Espectroscopia de Ressonância Magnética , Peptídeos/química , Sequência de Aminoácidos , Animais , Barreira Hematoencefálica/metabolismo , Células CHO , Sistema Nervoso Central/metabolismo , Cricetinae , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Medição da Dor , Peptídeos/metabolismo , Peptídeos/farmacocinética , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Receptores Opioides delta/agonistas , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Dodecilsulfato de Sódio/química , Água/química
3.
J Pharmacol Exp Ther ; 336(3): 767-78, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21118955

RESUMO

We have previously reported the chemistry and antinociceptive properties of a series of glycosylated enkephalin analogs (glycopeptides) exhibiting approximately equal affinity and efficacy at δ opioid receptors (DORs) and µ opioid receptors (MORs). More detailed pharmacology of the lead glycopeptide MMP-2200 [H2N-Tyr-D-Thr-Gly-Phe-Leu-Ser-(O-ß-D-lactose)-CONH2] is presented. MMP-2200 produced dose-related antinociception in the 55°C tail-flick assay after various routes of administration. The antinociceptive effects of MMP-2200 were blocked by pretreatment with the general opioid antagonist naloxone and partially blocked by the MOR-selective antagonist ß-funaltrexamine and the DOR-selective antagonist naltrindole. The κ opioid receptor antagonist nor-binaltorphimine and the peripherally active opioid antagonist naloxone-methiodide were ineffective in blocking the antinociceptive effects of MMP-2200. At equi-antinociceptive doses, MMP-2200 produced significantly less stimulation of locomotor activity compared with morphine. Repeated administration of equivalent doses of morphine and MMP-2200 (twice daily for 3 days) produced antinociceptive tolerance (~13- and 5-fold rightward shifts, respectively). In acute and chronic physical dependence assays, naloxone precipitated a more severe withdrawal in mice receiving morphine compared with equivalent doses of the glycopeptide. Both morphine and MMP-2200 inhibited respiration and gastrointestinal transit. In summary, MMP-2200 acts as a mixed DOR/MOR agonist in vivo, which may in part account for its high antinociceptive potency after systemic administration, as well as its decreased propensity to produce locomotor stimulation, tolerance, and physical dependence in mice, compared with the MOR-selective agonist morphine. For other measures (e.g., gastrointestinal transit and respiration), the significant MOR component may not allow differentiation from morphine.


Assuntos
Analgésicos Opioides/farmacologia , Glicopeptídeos/farmacologia , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Analgésicos Opioides/química , Animais , Células CHO , Cricetinae , Cricetulus , Glicopeptídeos/química , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Receptores Opioides delta/fisiologia , Receptores Opioides mu/fisiologia
4.
Life Sci ; 86(15-16): 624-30, 2010 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-20159022

RESUMO

AIMS: The current study assessed the in vivo antagonist properties of nalmefene using procedures previously used to characterize the opioid antagonists naloxone, naltrexone, 6beta-naltrexol and nalbuphine. MAIN METHODS: ICR mice were used to generate antagonist dose-response curves with intraperitoneal (i.p.) nalmefene against fixed A(90) doses of morphine in models of morphine-stimulated hyperlocomotion and antinociception. Additional dose-response curves for antagonist precipitated opioid withdrawal were run in mice treated acutely (100mg/kg, s.c., -4h) or chronically (75mg pellet, s.c., -72h) with morphine. Comparisons were made between antagonist potency and degree of precipitated withdrawal. KEY FINDINGS: Nalmefene produced dose- and time-related antagonism of morphine-induced increases in locomotor activity with a calculated ID(50) (and 95% confidence interval) of 0.014 (0.007-0.027)mg/kg. Nalmefene produced rapid reversal of morphine-induced locomotor activity (5.1min for 50% reduction in morphine effect). A 0.32mg/kg dose of nalmefene produced blockade of morphine-induced antinociception in the 55 degrees C tail-flick test that lasted approximately 2h. Nalmefene was able to potently precipitate withdrawal in mice treated acutely or chronically with morphine. SIGNIFICANCE: These results demonstrate that nalmefene is similar to naloxone and naltrexone with respect to its in vivo pharmacology in mice. Specifically, nalmefene produces potent antagonism of morphine agonist effects while precipitating severe withdrawal. The compound has a slower onset and longer duration of action compared to naloxone and naltrexone. The data allows for a more complete preclinical comparison of nalmefene against other opioid antagonists including the putative opioid neutral antagonist 6beta-naltrexol.


Assuntos
Naltrexona/análogos & derivados , Antagonistas de Entorpecentes/farmacologia , Síndrome de Abstinência a Substâncias , Analgésicos Opioides/antagonistas & inibidores , Analgésicos Opioides/farmacologia , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Concentração Inibidora 50 , Masculino , Camundongos , Camundongos Endogâmicos ICR , Morfina/antagonistas & inibidores , Morfina/farmacologia , Atividade Motora/efeitos dos fármacos , Naltrexona/administração & dosagem , Naltrexona/farmacologia , Antagonistas de Entorpecentes/administração & dosagem , Dor/tratamento farmacológico , Fatores de Tempo
5.
Chem Biol Drug Des ; 69(1): 41-7, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17313456

RESUMO

A series of mu-agonist DAMGO analogs were synthesized and pharmacologically characterized to test the 'biousian' hypothesis of membrane hopping. DAMGO was altered by incorporating moieties of increasing water solubility into the C-terminus via carboxamide and simple glycoside additions. The hydrophilic C-terminal moieties were varied from glycinol in DAMGO (1) to l-serine amide (2), l-serine amide beta-d-xyloside (3), l-serine amide beta-d-glucoside (4), and finally to l-serine amide beta-lactoside (5). Opioid binding and mouse tail-flick studies were performed. Antinociceptive potency (intravenous) increased, passing through a maximum (A(50) approximately 0.2 micromol/kg) for 2 and 3 as membrane affinity versus water solubility became optimal, and dropped off (A(50) approximately 1.0 micromol/kg) for 4 and 5 as water solubility dominated molecular behavior. Intravenous A(50) values were plotted versus hydrodynamic values (glucose units, g.u.) for the glycoside moieties, or the hydrophilic/hydrophobic Connolly surface areas (A(50) versus e(-Awater/Alipid)), and provided either a V-shaped or a U-shaped curve, as predicted by the 'biousian' hypothesis. The mu-selective receptor profile was maintained (K(i)'s = 0.66-1.3 nm) upon modifications at the C-terminus. The optimal 'degree of glycosylation' for the DAMGO peptide message appears to be between 1.25 and 1.75 g.u. (hydrodynamic g.u.), or 0.75 and 0.90 in terms of the surface-derived amphipathicity values.


Assuntos
Analgésicos Opioides/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Receptores Opioides mu/efeitos dos fármacos , Amidas/química , Animais , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina/metabolismo , Glucose/análogos & derivados , Glicina/análogos & derivados , Glicosilação , Interações Hidrofóbicas e Hidrofílicas , Injeções Intraventriculares , Lactose/análogos & derivados , Masculino , Camundongos , Camundongos Endogâmicos ICR , Receptores Opioides mu/metabolismo , Serina/análogos & derivados , Solubilidade , Água/química , Xilose/análogos & derivados
6.
J Pharmacol Exp Ther ; 313(3): 1150-62, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15716384

RESUMO

The mu-opioid receptor displays basal signaling activity, which seems to be enhanced by exposure to opioid agonists. This study assesses the in vivo pharmacology of the putative "neutral" antagonist 6beta-naltrexol in comparison to other ligands with varying efficacy, such as naloxone, an inverse agonist in the opioid-dependent state. ICR mice were used to generate full antagonist dose-response curves for naloxone, naltrexone, nalbuphine, and 6beta-naltrexol in blocking acute antinociceptive effects of morphine and precipitating opioid withdrawal in models of physical dependence. 6beta-Naltrexol was roughly equipotent to naloxone and between 4.5- and 10-fold less potent than naltrexone in blocking morphine-induced antinociception and locomotor activity, showing that 6beta-naltrexol enters the central nervous system. In contrast to naloxone and naltrexone, 6beta-naltrexol precipitated only minimal withdrawal at high doses in an acute dependence model and was approximately 77- and 30-fold less potent than naltrexone and naloxone, respectively, in precipitating withdrawal in a chronic dependence model. 6beta-Naltrexol reduced the inverse agonist effects of naloxone in vitro and in vivo, as expected for a neutral antagonist. Therefore, the pharmacological effects of 6beta-naltrexol differ markedly from those of naloxone and naltrexone in the opioid-dependent state. A reduction of withdrawal effects associated with neutral mu-opioid receptor antagonists may offer advantages in treating opioid overdose and addiction.


Assuntos
Naloxona/farmacologia , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Animais , Compostos de Benzilideno/farmacologia , Relação Dose-Resposta a Droga , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Morfina/antagonistas & inibidores , Atividade Motora/efeitos dos fármacos
7.
J Pharmacol Exp Ther ; 311(1): 290-7, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15166257

RESUMO

Development of opioid peptides as therapeutic agents has historically been limited due to pharmacokinetic issues including stability and blood-brain barrier (BBB) permeability. Glycosylation of opioid peptides can increase peptide serum stability and BBB penetration. To further define the requirements for optimizing in vivo antinociceptive potency following intravenous administration, we synthesized a series of enkephalin-based glycopeptides using solid phase 9-fluorenylmethyloxy carbamate methods. The compounds differed in the sixth and subsequent amino acid residues (Ser or Thr) and in the attached carbohydrate moiety. In vitro binding and functional smooth muscle bioassays indicated that the addition of mono- or disaccharides did not significantly affect the opioid receptor affinity or agonist activity of the glycopeptides compared with their unglycosylated parent peptides. All of the glycopeptides tested produced potent antinociceptive effects in male ICR mice following intracerebroventricular injection in the 55 degrees C tail-flick test. The calculated A(50) values for the Ser/Thr and monosaccharide combinations were all very similar with values ranging from 0.02 to 0.09 nmol. Selected compounds were administered to mice intravenously and tested for antinociception to indirectly assess serum stability and BBB penetration. All compounds tested produced full antinociceptive effects with calculated A (50) values ranging from 2.2 to 46.4 micromol/kg with the disaccharides having potencies that equaled or exceeded that of morphine on a micromoles per kilogram basis. Substitution of a trisaccharide or bis- and tris-monosaccharides resulted in a decrease in antinociceptive potency. These results provide additional support for the utility of glycosylation to increase central nervous system bioavailability of small peptides and compliment our ongoing stability and blood-brain barrier penetration studies.


Assuntos
Analgésicos/uso terapêutico , Encefalinas/uso terapêutico , Glicopeptídeos/uso terapêutico , Dor/tratamento farmacológico , Analgésicos/química , Animais , Modelos Animais de Doenças , Encefalinas/química , Glicopeptídeos/química , Masculino , Camundongos , Camundongos Endogâmicos ICR , Peptídeos Opioides/química , Peptídeos Opioides/uso terapêutico , Medição da Dor/efeitos dos fármacos , Relação Estrutura-Atividade
8.
J Pharmacol Exp Ther ; 308(2): 512-20, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14600246

RESUMO

Narcotic analgesics cause addiction by poorly understood mechanisms, involving mu opoid receptor (MOR). Previous cell culture studies have demonstrated significant basal, spontaneous MOR signaling activity, but its relevance to narcotic addiction remained unclear. In this study, we tested basal MOR-signaling activity in brain tissue from untreated and morphine-pretreated mice, in comparison to antagonist-induced withdrawal in morphine-dependent mice. Using guanosine 5'-O-(3-[(35)S]thio)triphosphate ([(35)S]GTP gamma S) binding and adenylyl cyclase activity assay in brain homogenates, we demonstrated that morphine pretreatment of mice enhanced basal MOR signaling in mouse brain homogenates and, moreover, caused persistent changes in the effects of naloxone and naltrexone, antagonists that elicit severe withdrawal in dependent subjects. Naloxone and naltrexone suppressed basal [(35)S]GTP gamma S binding (acting as "inverse agonists") only after morphine pretreatment, but not in drug-naive animals. Moreover, naloxone and naltrexone stimulated adenylyl cyclase activity in striatum homogenates only after morphine pretreatment, by reversing the inhibitory effects of basal MOR activity. After cessation of morphine treatment, the time course of inverse naloxone effects on basal MOR signaling was similar to the time course of naltrexone-stimulated narcotic withdrawal over several days. The neutral antagonist 6 beta-naltrexol blocked MOR activation without affecting basal signaling (G protein coupling and adenylyl cyclase regulation) and also elicited substantially less severe withdrawal. These results demonstrate long-lasting regulation of basal MOR signaling as a potential factor in narcotic dependence.


Assuntos
Encéfalo/metabolismo , Naltrexona/análogos & derivados , Transtornos Relacionados ao Uso de Opioides/metabolismo , Receptores Opioides mu/metabolismo , Transdução de Sinais/fisiologia , Adenilil Ciclases/metabolismo , Animais , Encéfalo/enzimologia , Modelos Animais de Doenças , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Morfina/farmacologia , Atividade Motora/efeitos dos fármacos , Naltrexona/sangue , Naltrexona/líquido cefalorraquidiano , Antagonistas de Entorpecentes/sangue , Antagonistas de Entorpecentes/líquido cefalorraquidiano , Receptores Opioides mu/fisiologia , Síndrome de Abstinência a Substâncias/metabolismo , Radioisótopos de Enxofre
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...