Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters










Publication year range
1.
J Chromatogr A ; 1431: 138-144, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26774122

ABSTRACT

The α3ß4α5 nAChR has been recently shown to be a useful target for smoking cessation pharmacotherapies. Herein, we report on the development and characterization of the α3ß4α5 nicotinic receptor column by frontal displacement chromatography. The binding affinity of the nicotine and minor alkaloids found in tobacco smoke condensates were determined for both the α3ß4 and α3ß4α5 nicotinic receptors. It was demonstrated that while no subtype selectivity was observed for nicotine and nornicotine, anabasine was selective for the α3ß4α5 nicotinic receptor. The non-competitive inhibitor binding site was also studied and it was demonstrated while mecamylamine was not selective between subtypes, buproprion showed subtype selectivity for the α3ß4 nicotinic receptor. The application of this methodology to complex mixtures was then carried out by screening aqueous-alcoholic solutions of targeted plant extracts, including Lycopodium clavatum L. (Lycopodiaceae) and Trigonella foenum graecum L. (Fabaceae) against both the α3ß4 and α3ß4α5 nAChRs.


Subject(s)
Chemistry Techniques, Analytical/methods , Chromatography, Affinity , Plant Extracts/chemistry , Receptors, Nicotinic/chemistry , Alkaloids/chemistry , Anabasine/chemistry , Binding Sites , Fabaceae/chemistry , Lycopodiaceae/chemistry , Nicotine/analogs & derivatives , Nicotine/chemistry , Smoke/analysis
2.
Br J Pharmacol ; 172(2): 668-80, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24903063

ABSTRACT

BACKGROUND AND PURPOSE: Buprenorphine is a potent analgesic with high affinity at µ, δ and κ and moderate affinity at nociceptin opioid (NOP) receptors. Nevertheless, NOP receptor activation modulates the in vivo activity of buprenorphine. Structure activity studies were conducted to design buprenorphine analogues with high affinity at each of these receptors and to characterize them in in vitro and in vivo assays. EXPERIMENTAL APPROACH: Compounds were tested for binding affinity and functional activity using [(35) S]GTPγS binding at each receptor and a whole-cell fluorescent assay at µ receptors. BU08073 was evaluated for antinociceptive agonist and antagonist activity and for its effects on anxiety in mice. KEY RESULTS: BU08073 bound with high affinity to all opioid receptors. It had virtually no efficacy at δ, κ and NOP receptors, whereas at µ receptors, BU08073 has similar efficacy as buprenorphine in both functional assays. Alone, BU08073 has anxiogenic activity and produces very little antinociception. However, BU08073 blocks morphine and U50,488-mediated antinociception. This blockade was not evident at 1 h post-treatment, but is present at 6 h and remains for up to 3-6 days. CONCLUSIONS AND IMPLICATIONS: These studies provide structural requirements for synthesis of 'universal' opioid ligands. BU08073 had high affinity for all the opioid receptors, with moderate efficacy at µ receptors and reduced efficacy at NOP receptors, a profile suggesting potential analgesic activity. However, in vivo, BU08073 had long-lasting antagonist activity, indicating that its pharmacokinetics determined both the time course of its effects and what receptor-mediated effects were observed. LINKED ARTICLES: This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.


Subject(s)
Analgesics, Opioid/pharmacology , Buprenorphine/analogs & derivatives , Narcotic Antagonists/pharmacology , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/antagonists & inhibitors , Analgesics, Opioid/pharmacokinetics , Analgesics, Opioid/therapeutic use , Animals , Anxiety/drug therapy , Anxiety/physiopathology , Behavior, Animal/drug effects , Buprenorphine/pharmacokinetics , Buprenorphine/pharmacology , Buprenorphine/therapeutic use , CHO Cells , Cricetulus , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Hot Temperature , Humans , Male , Membrane Potentials/drug effects , Mice, Inbred ICR , Narcotic Antagonists/pharmacokinetics , Narcotic Antagonists/therapeutic use , Pain/drug therapy , Pain/physiopathology , Receptors, Opioid, mu/physiology
3.
J Pharmacol Exp Ther ; 336(2): 524-32, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21071556

ABSTRACT

Astrocytomas and glioblastomas have been particularly difficult to treat and refractory to chemotherapy. However, significant evidence has been presented that demonstrates a decrease in astrocytoma cell proliferation subsequent to an increase in cAMP levels. The 1321N1 astrocytoma cell line, as well as other astrocytomas and glioblastomas, expresses ß(2)-adrenergic receptors (ß(2)-ARs) that are coupled to G(s) activation and consequent cAMP production. Experiments were conducted to determine whether the ß(2)-AR agonist (R,R')-fenoterol and other ß(2)-AR agonists could attenuate mitogenesis and, if so, by what mechanism. Receptor binding studies were conducted to characterize ß(2)-AR found in 1321N1 and U118 cell membranes. In addition, cells were incubated with (R,R')-fenoterol and analogs to determine their ability to stimulate intracellular cAMP accumulation and inhibit [(3)H]thymidine incorporation into the cells. 1321N1 cells contain significant levels of ß(2)-AR as determined by receptor binding. (R,R')-fenoterol and other ß(2)-AR agonists, as well as forskolin, stimulated cAMP accumulation in a dose-dependent manner. Accumulation of cAMP induced a decrease in [(3)H]thymidine incorporation. There was a correlation between concentration required to stimulate cAMP accumulation and inhibit [(3)H]thymidine incorporation. U118 cells have a reduced number of ß(2)-ARs and a concomitant reduction in the ability of ß(2)-AR agonists to inhibit cell proliferation. These studies demonstrate the efficacy of ß(2)-AR agonists for inhibition of growth of the astrocytoma cell lines. Because a significant portion of brain tumors contain ß(2)-ARs to a greater extent than whole brain, (R,R')-fenoterol, or some analog, may be useful in the treatment of brain tumors after biopsy to determine ß(2)-AR expression.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Astrocytoma/drug therapy , Astrocytoma/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclic AMP/metabolism , Fenoterol/pharmacology , G1 Phase/drug effects , Humans , Propanolamines/metabolism , Thymidine/metabolism
4.
Br J Pharmacol ; 153(3): 609-19, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18059322

ABSTRACT

BACKGROUND AND PURPOSE: Compounds that activate both NOP and mu-opioid receptors might be useful as analgesics and drug abuse medications. Studies were carried out to better understand the biological activity of such compounds. EXPERIMENTAL APPROACH: Binding affinities were determined on membranes from cells transfected with NOP and opioid receptors. Functional activity was determined by [(35)S]GTPgammaS binding on cell membranes and using the mouse vas deferens preparation in vitro and the tail flick antinociception assay in vivo. KEY RESULTS: Compounds ranged in affinity from SR14150, 20-fold selective for NOP receptors, to buprenorphine, 50-fold selective for mu-opioid receptors. In the [(35)S]GTPgammaS assay, SR compounds ranged from full agonist to antagonist at NOP receptors and most were partial agonists at mu-opioid receptors. Buprenorphine was a low efficacy partial agonist at mu-opioid receptors, but did not stimulate [(35)S]GTPgammaS binding through NOP. In the mouse vas deferens, each compound, except for SR16430, inhibited electrically induced contractions. In each case, except for N/OFQ itself, the inhibition was due to mu-opioid receptor activation, as determined by equivalent results in NOP receptor knockout tissues. SR14150 showed antinociceptive activity in the tail flick test, which was reversed by the opioid antagonist naloxone. CONCLUSIONS AND IMPLICATIONS: Compounds that bind to both mu-opioid and NOP receptors have antinociceptive activity but the relative contribution of each receptor is unclear. These experiments help characterize compounds that bind to both receptors, to better understand the mechanism behind their biological activities, and identify new pharmacological tools to characterize NOP and opioid receptors.


Subject(s)
Pain/drug therapy , Receptors, Opioid, mu/drug effects , Receptors, Opioid/drug effects , Analgesics, Opioid/pharmacology , Animals , Buprenorphine/pharmacology , CHO Cells , Cell Membrane/metabolism , Cricetinae , Cricetulus , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Indoles/pharmacology , Ligands , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pain Measurement , Protein Binding , Receptors, Opioid/genetics , Receptors, Opioid/metabolism , Receptors, Opioid, mu/metabolism , Vas Deferens/drug effects , Vas Deferens/metabolism , Nociceptin Receptor
5.
J Pept Res ; 64(3): 87-94, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15317498

ABSTRACT

Nociceptin/orphanin FQ (N/OFQ) is a 17 amino acid peptide that is the endogenous ligand for the G-protein coupled receptor ORL1 (NOP), a member of the opioid receptor family. Although it is clear that this receptor system is involved in a variety of physiologic functions, including analgesia, the precise actions of N/OFQ remain largely uncharacterized. One reason for this has been limited number of high-affinity ligands to NOP, and particularly the lack of availability of useful specific antagonists. Herein, we describe the pharmacologic activity of a series of modified amino acid containing modifications of the hexapeptide Ac-RYYRWR-NH2, with high affinity for NOP. These compounds were tested for binding affinity using [3H]N/OFQ binding to human NOP in CHO cells, and functional activity by measuring stimulation of [35S]GTPgammaS-binding in CHO cell membranes. These studies suggest that each Arg of the hexapeptide is required to maintain high-binding affinity. The peptide maintains high affinity if the Tyr2 or Tyr3 are modified, but at least one of these residues must maintain its hydroxyl group or there is a large decrease in intrinsic activity of the peptide.


Subject(s)
Oligopeptides/chemistry , Oligopeptides/pharmacology , Receptors, Opioid/drug effects , Animals , Arginine/chemistry , CHO Cells , Cricetinae , DNA, Complementary/genetics , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , In Vitro Techniques , Kinetics , Ligands , Male , Mice , Oligopeptides/metabolism , Opioid Peptides/chemistry , Opioid Peptides/metabolism , Receptors, Opioid/genetics , Receptors, Opioid/metabolism , Recombinant Proteins/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship , Transfection , Tyrosine/chemistry , Vas Deferens/drug effects , Vas Deferens/physiology , Nociceptin Receptor , Nociceptin
6.
J Pept Res ; 62(5): 191-8, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14531842

ABSTRACT

Nociceptin/Orphanin FQ (N/OFQ) is a 17 amino acid peptide that is the endogenous ligand for the G protein-coupled receptor (opioid receptor like 1, ORL1), a member of the opioid receptor family. Although it is clear that this receptor system is involved in a variety of physiological functions, including analgesia, the precise actions of N/OFQ remain largely uncharacterized. One reason for this has been limited high affinity ligands to ORL1, and particularly the lack of availability of useful specific antagonists. Herein we describe the pharmacological activity of a series of N-terminally modified hexapeptides with high affinity for ORL1. These compounds were tested for binding affinity using [3H]N/OFQ binding to human ORL1 in CHO cells, and functional activity by measuring stimulation of [35S]GTPgammaS binding in CHO cell membranes. The N-terminal modifications have produced compounds that maintained very high receptor affinity, but led to significant changes in intrinsic activity. One compound, pentanoyl-RYYRWR-NH2, with barely measurable agonist activity was tested in vivo. It was found to possess modest analgesic activity, but it was unable to block the morphine modulatory activity of N/OFQ.


Subject(s)
Analgesics, Opioid/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Narcotic Antagonists/pharmacology , Oligopeptides/pharmacology , Receptors, Opioid/agonists , Animals , Binding, Competitive , CHO Cells , Colforsin , Cricetinae , Dose-Response Relationship, Drug , Humans , Mice , Oligopeptides/chemical synthesis , Opioid Peptides/pharmacology , Pain/drug therapy , Pain/metabolism , Receptors, Opioid/genetics , Sulfur Radioisotopes , Nociceptin Receptor , Nociceptin
7.
J Pharmacol Exp Ther ; 298(3): 1213-20, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11504823

ABSTRACT

Pregnancy and its hormonal simulation via 17beta-estradiol (E(2)) and progesterone (P) are associated with spinal opioid antinociception, primarily driven by augmented dynorphin/kappa-opioid activity. This study addresses the ovarian sex steroid-activated mechanism(s) that underlie this activation using an ex vivo spinal cord preparation. In lumbar spinal cord obtained from control animals, exogenous kappa- or delta-opioid agonists (but not mu), as well as nociceptin (orphanin FQ; N/OFQ), dose dependently inhibit the stimulated release of dynorphin. Consistent with these observations, stimulated dynorphin release is enhanced following selective blockade of opioid or N/OFQ receptors, indicating that their endogenous ligands are negative modulators of dynorphin release. In lumbar spinal cord obtained from ovariectomized animals exposed to pregnancy blood levels of E(2)/P, basal and stimulated rates of dynorphin release increase approximately 2-fold. Moreover, evoked dynorphin release is no longer negatively modulated by kappa- or delta-opioid agonists or N/OFQ. Interestingly, in these preparations, release can be facilitated by delta-opioid receptor activation, and neither spinal opioid nor N/OFQ receptor blockade enhances evoked dynorphin release. Consistent with these observations, guanosine-5'-O-3-[(35)S]-thio triphosphate binding analyses indicate a reduction in functional N/OFQ receptors. These data indicate that at least part of the E(2)/P-induced augmented activity of lumbar dynorphin neurons results from their disinhibition via the removal of negative opioid and N/OFQ modulation. These results underscore the plasticity of spinal opioid and N/OFQ systems and their dependence on the ovarian sex steroid milieu. Ovarian sex steroid-activated antinociception reveals mechanisms that enable sustained opioid activation without concomitant tolerance formation.


Subject(s)
Dynorphins/metabolism , Estradiol/pharmacology , Opioid Peptides/pharmacology , Opioid Peptides/physiology , Progesterone/pharmacology , Spinal Cord/metabolism , Animals , Female , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , In Vitro Techniques , Membranes/drug effects , Membranes/metabolism , Neuronal Plasticity/drug effects , Perfusion , Potassium/pharmacology , Radioimmunoassay , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects , Nociceptin
8.
Int J Cancer ; 91(1): 8-21, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11149424

ABSTRACT

Evaluation of retinoic acid receptor (RAR) subtype-selective alpha and gamma agonists and antagonists and a retinoid X receptor (RXR) class-selective agonist for efficacy at inhibiting both induction of ornithine decarboxylase (ODC) by the tumor promoter 12-O-tetradecanoylphorbol-13-acetate (TPA) in mouse epidermis and rat tracheal epithelial cells and the appearance of papillomas in mouse epidermis treated in the 2-stage tumor initiation-promotion model indicated that (i) RXR class-selective transcriptional agonists, such as MM11246, were not involved in ODC inhibition; (ii) RAR-selective agonists that induce gene transcription from RA-responsive elements (RAREs) were active at low concentrations; (iii) RAR-selective antagonists that bind RARs and inhibit AP-1 activation on the collagenase promoter but do not activate RAREs to induce gene transcription were less effective inhibitors; and (iv) RARgamma-selective retinoid agonists were more effective inhibitors of TPA-induced ODC activity than RARalpha-selective agonists. These results suggest that RARE activation has a more important role in inhibition of ODC activity than RXR activation or AP-1 inhibition and that RARgamma-selective agonists would be the most useful inhibitors of epithelial cell proliferation induced by tumor promoters. The natural retinoid all-trans-RA induced expression of transcription factor ZBP-89, which represses activation of the GC box in the ODC promoter by the transcription factor Sp1.


Subject(s)
DNA-Binding Proteins/physiology , Ornithine Decarboxylase Inhibitors , Receptors, Retinoic Acid/metabolism , Retinoids/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factors/physiology , 9,10-Dimethyl-1,2-benzanthracene , Animals , Antineoplastic Agents/pharmacology , Blotting, Northern , Blotting, Western , Carcinogens , Cell Survival/drug effects , Collagenases/genetics , Dose-Response Relationship, Drug , Epidermis/metabolism , Epithelial Cells/metabolism , Female , HeLa Cells , Humans , Mice , Mice, Hairless , Neoplasms, Experimental/metabolism , Papilloma/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Kinases/metabolism , RNA, Messenger/metabolism , Rats , Receptors, Retinoic Acid/chemistry , Response Elements , Retinoic Acid Receptor alpha , Retinoids/pharmacology , Time Factors , Trachea/metabolism , Transcription Factor AP-1/antagonists & inhibitors , Transcription, Genetic , Transcriptional Activation , Transfection , Ultraviolet Rays , Retinoic Acid Receptor gamma
9.
Eur J Pharmacol ; 428(1): 29-36, 2001 Sep 28.
Article in English | MEDLINE | ID: mdl-11779034

ABSTRACT

Nociceptin/orphanin FQ (N/OFQ) was recently identified as the endogenous ligand for the opioid-receptor like (ORL1) receptor. Although the ORL1 receptor shows sequence homology with the opioid receptors, the nociceptin/ORL1 ligand-receptor system has very distinct pharmacological actions compared to the opioid receptor system. Recently, several small-molecule ORLI receptor ligands were reported by pharmaceutical companies. Most of these ligands had close structural similarities with known neuroleptics and opiates. In this study, we screened several available neuroleptics and opiates for their binding affinity and functional activity at ORL1 and the opioid receptors. We also synthesized several analogs of known opiates with modified piperidine N-substituents in order to characterize the ORL1 receptor ligand binding pocket. Substitution with the large, lipophilic cyclooctylmethyl moiety increased ORL1 receptor affinity and decreased mu receptor affinity and efficacy in the fentanyl series of ligands but had a different effect in the oripavine class of opiate ligands. Our results indicate that opiates and neuroleptics may be good starting points for ORL1 receptor ligand design, and the selectivity may be modulated by appropriate structural modifications.


Subject(s)
Antipsychotic Agents/pharmacology , Narcotics/pharmacology , Receptors, Opioid/drug effects , Animals , CHO Cells , Cricetinae , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Humans , Kinetics , Structure-Activity Relationship , Nociceptin Receptor
10.
Biochem Biophys Res Commun ; 276(2): 710-7, 2000 Sep 24.
Article in English | MEDLINE | ID: mdl-11027536

ABSTRACT

We have cloned regions of the 5'-untranslated region of the human prepronociceptin (ppN/OFQ) gene into a luciferase reporter plasmid, pGL3 Basic Vector. By primer extension analysis, we determined that one start site of transcription lies within the known human cDNA sequence. There are two cyclic AMP response elements (CRE) with the consensus sequence CGTCA within 250 bp of the start of transcription. We designed various constructs around these sites, performed transient transfections, and measured luciferase activity in NS20Y cells. Transcriptional activity could be regulated by a variety of factors including cAMP levels, Ca(2+), and particularly by a 125-bp region adjacent to an intron located 23 bp upstream of the translation initiating ATG. These data should help in understanding the regulatory mechanisms of ppN/OFQ gene transcription.


Subject(s)
5' Untranslated Regions/genetics , Gene Expression Regulation , Protein Precursors/genetics , Receptors, Opioid/genetics , 5' Untranslated Regions/metabolism , Animals , Base Sequence , Binding Sites , Cloning, Molecular , Cricetinae , DNA, Complementary/analysis , Genes, Reporter , Humans , Luciferases/metabolism , Molecular Sequence Data , Transcription Factors/metabolism , Transcription, Genetic
11.
Peptides ; 21(7): 1147-54, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10998550

ABSTRACT

Recent studies have identified compounds with reduced efficacy relative to nociceptin/orphanin FQ at the opioid-like receptor ORL1. Utilizing stimulation of [(35)S]GTPgammaS binding as in vitro assays, it was determined that both [Phe(1)psi(CH(2)-NH)Gly(2)]N/OFQ(1-13)NH(2) and the hexapeptide Ac-RYYRIK-NH(2) act as partial agonists in CHO cells transfected with either human or mouse ORL1. Maximal activity for both [Phe(1)psi(CH(2)-NH)Gly(2)]N/OFQ(1-13)NH(2) and Ac-RYYRIK-NH(2) was significantly greater in cells transfected with the human receptor (90% and 73% in a high expressing clone, 76% and 68% in low expressing clone) rather than the mouse receptor (37.5 and 33%), regardless of receptor number in individual clones. In vitro studies in cells transfected with exaggerated receptor numbers can lead to unreliable estimates of agonist and antagonist activity, however, these studies suggest that animal experiments on the activity of novel compounds may not always be better predictors of the ultimate activity in humans.


Subject(s)
Peptides/metabolism , Receptors, Opioid/metabolism , Species Specificity , Animals , CHO Cells , Colforsin/pharmacology , Cricetinae , Cyclic AMP/metabolism , DNA, Complementary/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Inhibitory Concentration 50 , Kinetics , Mice , Oligopeptides/pharmacology , Opioid Peptides/pharmacology , Peptide Fragments/pharmacology , Protein Binding , Receptors, Opioid/genetics , Transfection , Nociceptin Receptor
12.
J Med Chem ; 43(9): 1852-7, 2000 May 04.
Article in English | MEDLINE | ID: mdl-10794701

ABSTRACT

A series of ethers of orvinol and isoorvinol has been prepared and evaluated in opioid receptor binding and in vitro functional assays. The most striking finding was the very large difference in kappa-opioid receptor activity between the diastereomeric ethyl ethers: 46-fold in binding, 150-fold in GPI, and 900-fold in the [(35)S]GTPgammaS assay in favor of the (R)-diastereomer. Additionally in the (R)-series there was a 700-fold increase in kappa-agonist potency in the [(35)S]GTPgammaS assay when OEt was replaced by OBn. The data can be explained in a triple binding site model: an H-bonding site, a lipophilic site, and an inhibitory site with which the 20-Me group in the (S)-ethers may interact. It appears that kappa-agonist binding of the orvinols avoids the inhibitory site in the intramolecular H-bonded conformation.


Subject(s)
Analgesics, Opioid/pharmacology , Hydroxamic Acids/chemical synthesis , Narcotics/pharmacology , Analgesics, Opioid/chemistry , Animals , Binding, Competitive/drug effects , CHO Cells , Cell Membrane/drug effects , Cell Membrane/metabolism , Cricetinae , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Guinea Pigs , Humans , Hydroxamic Acids/pharmacology , In Vitro Techniques , Magnetic Resonance Spectroscopy , Narcotics/chemical synthesis , Narcotics/chemistry , Spectrophotometry, Infrared , Stereoisomerism
14.
Neuropeptides ; 31(5): 403-8, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9413015

ABSTRACT

The relationship between binding affinity and functional activity of nociceptin/orphanin FQ binding was studied in brain membranes, membranes of Chinese hamster ovary cells transfected with ORL1, and intact CHO-ORL1 cells. Binding affinities were compared with potency for the stimulation of [35S]GTP gamma S binding in cell membranes, and inhibition of forskolin-stimulated cAMP accumulation in intact cells. Binding was conducted with [3H]14-Tyr-nociceptin, and in brain or cell membranes the affinity was found to be 50-100 pM. The binding of [3H]14-Tyr-nociceptin was found to be regulated by Na+ and GTP, as expected for an opioid-like receptor. In intact cells, saturation produced a curvilinear Scatchard Plot. Non-linear analysis indicated two states of the receptor, with the vast majority of binding being to a low affinity state of approximately 8 nM. This low affinity component is consistent with the lower potency derived from the inhibition of cAMP accumulation, stimulation of [35S]GTP gamma S binding, and other functional assays.


Subject(s)
Opioid Peptides/physiology , Receptors, Opioid/agonists , Animals , Binding, Competitive/physiology , CHO Cells , Cricetinae , Guinea Pigs , Logistic Models , Opioid Peptides/metabolism , Nociceptin
15.
J Pharmacol Exp Ther ; 283(2): 735-41, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9353393

ABSTRACT

Fifteen hexapeptides having high affinity for the opioid-like receptor ORL1 were identified from a combinatorial library containing more than 52 million different hexapeptides. The five compounds with the highest affinity were characterized further by use of a variety of in vitro models. Binding studies indicated that these five peptides have affinity for ORL1 in the nanomolar range, similar to the recently discovered endogenous ligand called nociceptin and orphanin FQ (N/OFQ). The activity of these compounds was investigated in three different assays: stimulation of [35S]GTPgammaS binding and inhibition of forskolin-stimulated cAMP accumulation in Chinese hamster ovary cells transfected with ORL1, and inhibition of electrically induced contractions in the mouse vas deferens. In each assay, the five hexapeptides acted as partial agonists. The EC50 values for stimulation of [35S]GTPgammaS binding and inhibition of cAMP accumulation were in the range of that for N/OFQ, but maximal effects ranged from 70 to 90% of N/OFQ in the cAMP assay, and 30 to 60% of N/OFQ in the GTPgammaS assay. The positive hexapeptides identified were found to have minimal structural similarity to N/OFQ. The peptides are positively charged, which could enable them to bind to the negatively charged second extracellular loop thought to be a likely binding site for N/OFQ.


Subject(s)
Oligopeptides/metabolism , Receptors, Opioid/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cyclic AMP/biosynthesis , Male , Mice , Molecular Sequence Data , Oligopeptides/pharmacology , Rats , Vas Deferens/drug effects , Nociceptin Receptor
16.
Eur J Pharmacol ; 323(2-3): 261-7, 1997 Apr 04.
Article in English | MEDLINE | ID: mdl-9128848

ABSTRACT

The delta-opioid receptor found in SH-SY5Y cells was characterized in terms of binding profile and ability to mediate the inhibition of forskolin-stimulated cAMP accumulation. Both DPDPE ([D-Pen2,D-Pen5]enkephalin) and deltorphin II, compounds reported to be selective for the delta 1- and delta 2-opioid receptor respectively, were potent agonists in these cells. Binding studies indicated that naltrindole benzofuran (NTB) had significantly higher affinity than 7-benzylidenenaltrexone (BNTX); however, both compounds have high affinity for the delta-opioid receptor found in SH-SY5Y cells. Naltrindole benzofuran was found to be a potent antagonist, with an IC50 of less than 1 nM, while 7-benzylidene naltrexone was found to be a relatively weak antagonist, requiring greater than 100 nM to inhibit 50% of agonist activity. Binding to intact SH-SY5Y cells was compared to binding to cell membranes and guinea-pig brain membranes. In each case, binding affinities were very similar. These studies suggest that the receptor found in SH-SY5Y cells could probably be classified as a delta 2-opioid receptor. However, the very similar binding characteristics of SH-SY5Y cells and guinea-pig brain membranes call into question the ability to label delta 1-opioid receptors.


Subject(s)
Neuroblastoma/metabolism , Receptors, sigma/metabolism , Animals , Binding, Competitive , Brain/metabolism , Cyclic AMP/metabolism , Guinea Pigs , Humans , Membranes/metabolism , Neuroblastoma/pathology , Receptors, sigma/drug effects , Tumor Cells, Cultured/metabolism
17.
Eur J Pharmacol ; 325(1): 29-34, 1997 Apr 23.
Article in English | MEDLINE | ID: mdl-9151935

ABSTRACT

Nociceptin (orphanin FQ) is an endogenous peptide agonist for the newly discovered receptor (opioid receptor-like 1 receptor, ORL1) that bears striking homology to opioid receptors. Initial reports claimed that this peptide had hypoalgesic effects following i.c.v. or i.t. administration. The present study demonstrates that, in the presence of opioid receptor blockade, nociceptin can substantially alter the magnitude of the stimulated release of methionine-enkephalin from the guinea pig myenteric plexus. This effect is concentration dependent. Low doses (1 or 10 nM) inhibit whereas higher concentrations (100 or 1000 nM) enhance evoked enkephalin release. In contrast, in the absence of opioid receptor blockade, a statistically significant inhibition of stimulated enkephalin release is observed in response to 1, 100 or 1000 nM nociceptin. However, the magnitude of this effect did not differ among these concentrations. Furthermore, at 10 nM nociceptin, either an inhibition or enhancement of stimulated enkephalin release is manifest. The ability of naloxone to alter the nociceptin modulation of enkephalin release suggests that a component of the nociceptin modulation of enkephalin release is mediated via opioid receptors. This is consistent with the observation that this peptide has modest affinity for opioid receptors (L > K > 8) which, under appropriate conditions, should be sufficient to permit interactions with multiple opioid receptor types. This complicates dose responsiveness for nociceptin since both the naloxone-resistant (ORL1-mediated) and naloxone-sensitive (opioid receptor-mediated) component exhibit a concentration-dependent bimodality (albeit in opposite directions). Determination of i.c.v. or i.t. nociceptin dose responsiveness over several orders of magnitude is suggested before concluding the physiological effects of this peptide.


Subject(s)
Enkephalin, Methionine/metabolism , Myenteric Plexus/drug effects , Myenteric Plexus/metabolism , Opioid Peptides/pharmacology , Animals , Dose-Response Relationship, Drug , Electric Stimulation , Guinea Pigs , In Vitro Techniques , Kinetics , Naloxone/pharmacology , Narcotic Antagonists , Opioid Peptides/metabolism , Receptors, Opioid/metabolism , Nociceptin Receptor , Nociceptin
18.
Synapse ; 24(2): 193-201, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8890461

ABSTRACT

Naloxone benzoylhydrazone (NalBzoH) has proved a valuable tool in the investigation of opioid receptor subtypes. In the present study, we have examined a series of derivatives of NalBzoH in which substitutions have been made on the benzoyl ring. Overall, we see dramatic effects on the binding affinities of derivatives against the various opioid receptor subtypes. Although the range of affinities against the mu receptors is quite modest, ranges of the others vary almost 30-fold for kappa 3, 50-fold for kappa 1 and 100-fold for delta and kappa 2 binding. Few substituted derivatives display greater affinity than NalBzoH for any of the receptors, except for delta sites where several derivatives have affinities almost tenfold greater than NalBzoH. Along with the wide variations in affinity, the compounds also appear to exhibited widely divergent activities in traditional bioassays.


Subject(s)
Binding, Competitive , Naloxone/analogs & derivatives , Naloxone/metabolism , Receptors, Opioid/drug effects , Animals , Cattle , Guinea Pigs
19.
Eur J Pharmacol ; 302(1-3): R1-2, 1996 Apr 29.
Article in English | MEDLINE | ID: mdl-8791013

ABSTRACT

The recently discovered neuropeptide nociceptin was found to inhibit electrically induced contractions of the mouse vas deferens. Nociceptin and its 14-Tyr analog were each partial agonists, but with high affinity (ED50 of 20 nM). This activity was not opioid in nature, as it was not inhibited by either selective or non-selective opiate antagonists.


Subject(s)
Muscle Contraction/drug effects , Opioid Peptides/pharmacology , Vas Deferens/drug effects , Animals , Dose-Response Relationship, Drug , Electrophysiology , Male , Mice , Narcotic Antagonists/pharmacology , Opioid Peptides/antagonists & inhibitors , Rats , Receptors, Opioid/agonists , Nociceptin
20.
Peptides ; 17(5): 873-6, 1996.
Article in English | MEDLINE | ID: mdl-8844779

ABSTRACT

In vitro biotransformation of a newly sequenced neuropeptide of 17 amino acid residues, named nociceptin and orphanin FQ by two separate research groups, was studied in human blood using matrix-assisted laser desorption/ionization mass spectrometry. Processing was carried out in freshly drawn blood incubated at 37 degrees C for various time periods. It was found that cleavage at peptide linkage Phe1-Gly2 was the predominant biotransformation pathway. Nociceptin (2-17) was the major biotransformation product. Further processing also occurred with the formation of a variety of minor biotransformation products. Cleavages at basic amino acid residues were observed, although these were not major biotransformation pathways found under these in vitro experimental conditions. Biotransformation of nociceptin followed a similar pattern to that of another neuropeptide, the endogenous opioid dynorphin A(1-17), but it appeared that nociceptin was more resistant to biotransformation in human blood in vitro than dynorphin A(1-17).


Subject(s)
Opioid Peptides/pharmacokinetics , Receptors, Opioid/agonists , Adult , Biotransformation , Humans , Male , Opioid Peptides/blood , Receptors, Opioid/blood , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Nociceptin
SELECTION OF CITATIONS
SEARCH DETAIL
...