Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Neuropharmacology ; 202: 108858, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34715121

ABSTRACT

Because of increased opioid misuse, there is a need to identify new targets for minimizing opioid tolerance, and physical and psychological dependence. Previous studies showed that fibroblast growth factor 21 (FGF21) decreased alcohol and sweet preference in mice. In this study, FGF21-transgenic (FGF21-Tg) mice, expressing high FGF21 serum levels, and wildtype (WT) C57BL/6J littermates were treated with morphine and saline to determine if differences exist in their physiological and behavioral responses to opioids. FGF21-Tg mice displayed reduced preference for morphine in the conditioned place preference assay compared to WT littermates. Similarly, FGF21-Tg mice had an attenuation of the magnitude and rate of acute morphine antinociceptive tolerance development, and acute and chronic morphine physical dependence, but exhibited no change in chronic morphine antinociceptive tolerance. The ED50 values for morphine-induced antinociception in the 55 °C hot plate and the 55 °C warm-water tail withdrawal assays were similar in both strains of mice. Likewise, FGF21-Tg and WT littermates had comparable responses to morphine-induced respiratory depression. Overall, FGF21-Tg mice had a decrease in the development of acute analgesic tolerance, and the development of physical dependence, and morphine preference. FGF21 and its receptor have therapeutic potential for reducing opioid withdrawal symptoms and craving, and augmenting opioid therapeutics for acute pain patients to minimize tolerance development.


Subject(s)
Drug Tolerance/genetics , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/physiology , Morphine Dependence/genetics , Morphine/adverse effects , Nociception/drug effects , Animals , Behavior, Animal/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/genetics , Substance Withdrawal Syndrome/genetics , Substance Withdrawal Syndrome/therapy
2.
Mol Pharmacol ; 98(4): 462-474, 2020 10.
Article in English | MEDLINE | ID: mdl-32958572

ABSTRACT

Opioid receptors (ORs) convert extracellular messages to signaling events by coupling to the heterotrimeric G proteins, Gα•ßγ Classic pharmacological methods, such as [35S]GTPγS binding and inhibition of cyclic AMP production, allow for general opioid characterization, but they are subject to the varying endogenous Gα proteins in a given cell type. Bioluminescence resonance energy transfer (BRET) technology offers new insight by allowing the direct observation of Gα subunit-specific effects on opioid pharmacology. Using a Venus-tagged Gßγ and nanoluciferase-tagged truncated G protein receptor kinase 3, an increase in BRET signal correlated with OR activation mediated by a specific Gα protein. The magnitude of the BRET signal was normalized to the maximum response obtained with 10 µM 2-(3,4-dichlorophenyl)-N-methyl-N-[(1R,2R)-2-pyrrolidin-1-ylcyclohexyl]acetamide (U50,488) for the kappa OR (KOR). Opioids reached equilibrium with the KOR, and concentration-response curves were generated. Although the full agonists U50,488, salvinorin A, nalfurafine, and dynorphin peptides were equally efficacious regardless of the Gα subunit present, the concentration-response curves were leftward shifted when the KOR was signaling through Gαz compared with other Gαi/o subunits. In contrast, the Gα subunit distinctly affected both the efficacy and potency of partial kappa agonists, such as the benzomorphans, and the classic mu opioid antagonists, naloxone, naltrexone, and nalmefene. For example, (-)pentazocine had EC50 values of 7.3 and 110 nM and maximal stimulation values of 79% and 35% when the KOR signaled through Gαz and Gαi1, respectively. Together, these observations suggest KOR pharmacology varies based on the specific Gα subunit coupled to the KOR. SIGNIFICANCE STATEMENT: Opioid receptors couple to various heterotrimeric Gαßγ proteins to convert extracellular cues to precise intracellular events. This paper focuses on how the various inhibitory Gα subunits influence the pharmacology of full and partial agonists at the kappa opioid receptor. Using a bioluminescent assay, the efficacy and potency of kappa opioids was determined. Opioid signaling was more potent through Gαz compared with other Gα proteins. These observations suggest that Gαz may impact opioid pharmacology and cellular physiology more than previously thought.


Subject(s)
Analgesics, Opioid/pharmacology , Bioluminescence Resonance Energy Transfer Techniques/methods , GTP-Binding Protein alpha Subunits/metabolism , Receptors, Opioid, kappa/metabolism , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Diterpenes, Clerodane/pharmacology , Dynorphins/pharmacology , HEK293 Cells , Humans , Morphinans/pharmacology , Signal Transduction/drug effects , Spiro Compounds/pharmacology
3.
J Pharmacol Exp Ther ; 367(2): 267-281, 2018 11.
Article in English | MEDLINE | ID: mdl-30108159

ABSTRACT

A combination of buprenorphine (BUP) and samidorphan (SAM) at a 1:1 (mg/mg) fixed-ratio dose is being investigated as an adjunctive treatment of major depressive disorder (BUP/SAM, ALKS 5461). Both [3H]BUP and [3H]SAM bound to the µ-, κ-, and δ-opioid receptors (MOR, KOR, and DOR, respectively) with Kd values of 3 nM or less. [3H]BUP dissociated from the MOR more slowly than [3H]SAM did. In the [35S]GTPγS assay, BUP was a partial agonist at the MOR, KOR, and DOR. SAM was an antagonist at the MOR and a partial agonist at the KOR and DOR. The pharmacology of the combination of SAM and BUP was characterized at ratios like the molar ratios of both compounds at steady state in humans. In all assessments, SAM reduced the efficacy of BUP at the MOR without altering its potency. At the KOR, SAM had no significant effect on the activity of BUP. In bioluminescent resonance energy transfer assays, SAM, naltrexone, and naloxone were partial agonists when the MOR was coupled to the Gα oB and Gα z, and were antagonists when coupled to Gα i At the KOR, SAM was a partial agonist activating Gα oA and Gα oB and a full agonist in stimulating Gα z SAM inhibited BUP's recruitment of ß-arrestin to the MOR, suggesting an attenuation of BUP's efficacy in activating G proteins correlated with an inhibition of ß-arrestin recruitment. The collective data suggest that SAM attenuates the efficacy of BUP under all conditions tested at the MOR and DOR but had little effect on BUP activity at the KOR.


Subject(s)
Buprenorphine/pharmacology , Depressive Disorder, Major/drug therapy , Naltrexone/analogs & derivatives , Animals , CHO Cells , Cell Line , Cricetulus , Drug Combinations , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Naloxone/pharmacology , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , beta-Arrestins/metabolism
4.
Brain Behav Immun ; 67: 152-162, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28844812

ABSTRACT

Patients receiving the cytokine immunotherapy, interferon-alpha (IFN-α) frequently present with neuropsychiatric consequences and cognitive impairments. Patients (25-80%) report symptoms of depression, including, anhedonia, irritability, fatigue and impaired motivation. Our lab has previously demonstrated treatment (170,000IU/kg sc, 3 times per week for 4weeks) of the pro-inflammatory cytokine, IFN-α, induced a depressive phenotype in rats in the forced swim test (FST). Here, we examine the biological mechanisms underlying behavioral changes induced by IFN-α, which may be reflective of mechanisms underlying inflammation associated depression. We also investigate the potential of 3-carboxamido seco-nalmefene (3CS-nalmefene), a novel opioid modulator (antagonist at mu and partial agonist at kappa and delta opioid receptors in vitro), to reverse IFN-α induced changes. In vitro radioligand receptor binding assays and the [35S] GTPγS were performed to determine the affinity of 3CS-nalmefene for the mu, kappa and delta opioid receptors. IFN-α treatment increased circulating and central markers of inflammation and hypothalamic-pituitaryadrenal (HPA) axis activity (IL-6, IL-1ß and corticosterone) while increasing immobility in the FST, impairing of object displacement learning in the object exploration task (OET), and decreasing neuronal proliferation and brain-derived neurotrophic factor (BDNF) in the hippocampus. Treatment with 3CS-nalmefene (0.3mg/kg/sc twice per day, 3 times per week for 4weeks) prevented IFN-α-induced immobility in the FST and impaired object displacement learning. In addition, 3CS-nalmefene prevented IFN-α-induced increases in inflammation and hyperactivity of the HPA-axis, the IFN-α-induced reduction in both neuronal proliferation and BDNF expression in the hippocampus. Overall, these preclinical data would support the hypothesis that opioid receptor modulation is a relevant target for treatment of depression.


Subject(s)
Antidepressive Agents/administration & dosage , Depressive Disorder/drug therapy , Naltrexone/analogs & derivatives , Narcotic Antagonists/administration & dosage , Receptors, Opioid/agonists , Animals , Anxiety/chemically induced , Anxiety/drug therapy , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Cell Proliferation/drug effects , Depression/chemically induced , Depression/drug therapy , Depressive Disorder/chemically induced , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Interferon-alpha/administration & dosage , Male , Naltrexone/administration & dosage , Neurons/drug effects , Neurons/metabolism , Rats, Wistar
5.
J Med Chem ; 58(15): 5728-41, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26125201

ABSTRACT

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.


Subject(s)
Central Nervous System/drug effects , Glycopeptides/pharmacology , Opioid Peptides/pharmacology , Amino Acid Sequence , Animals , Circular Dichroism , Glycopeptides/chemistry , Magnetic Resonance Spectroscopy , Mice , Models, Molecular , Molecular Sequence Data , Opioid Peptides/chemistry , Protein Conformation , Receptors, Opioid, mu/drug effects
6.
J Med Chem ; 57(6): 2237-46, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24576160

ABSTRACT

Glycosylated ß-endorphin analogues of various amphipathicity were studied in vitro and in vivo in mice. Opioid binding affinities of the O-linked glycopeptides (mono- or disaccharides) and unglycosylated peptide controls were measured in human receptors expressed in CHO cells. All were pan-agonists, binding to µ-, δ-, or κ-opioid receptors in the low nanomolar range (2.2-35 nM K(i)'s). The glycoside moiety was required for intravenous (i.v.) but not for intracerebroventricular (i.c.v.) activity. Circular dichroism and NMR indicated the degree of helicity in H2O, aqueous trifluoroethanol, or micelles. Glycosylation was essential for activity after i.v. administration. It was possible to manipulate the degree of helicity by the alteration of only two amino acid residues in the helical address region of the ß-endorphin analogues without destroying µ-, δ-, or κ-agonism, but the antinociceptive activity after i.v. administration could not be directly correlated to the degree of helicity in micelles.


Subject(s)
Analgesics/chemical synthesis , Analgesics/pharmacology , Glycopeptides/chemistry , Glycopeptides/pharmacology , beta-Endorphin/analogs & derivatives , beta-Endorphin/pharmacology , Animals , CHO Cells , Circular Dichroism , Cricetinae , Cricetulus , Drug Design , Glycopeptides/chemical synthesis , Humans , Injections, Intravenous , Injections, Intraventricular , Magnetic Resonance Spectroscopy , Male , Mice , Micelles , Models, Molecular , Protein Conformation , Protein Structure, Secondary , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/drug effects , Receptors, Opioid, mu/metabolism , Structure-Activity Relationship
7.
ACS Chem Neurosci ; 5(2): 93-9, 2014 Feb 19.
Article in English | MEDLINE | ID: mdl-24393077

ABSTRACT

A series of levo- and dextromorphinan pairs have been synthesized and evaluated for their affinities to the mu, kappa, and delta opioid receptors, the N-methyl-D-aspartate (NMDA) channel, and sigma 1 and 2 receptors. It was found that levo isomers tended to have higher affinities at the opioid receptors and moderate to high affinities to the NMDA and sigma receptors, while dextro isomers tended to have lower affinities to the opioid receptors but comparatively higher affinities to the NMDA and sigma receptors. This series of compounds have interesting and complex pharmacological profiles, and merit further investigation as potential therapies for drug abuse treatment.


Subject(s)
Morphinans/pharmacology , N-Methylaspartate/drug effects , Receptors, Opioid/drug effects , Receptors, sigma/drug effects , Animals , Humans , Male , Morphinans/chemical synthesis , Rats , Rats, Sprague-Dawley
8.
J Med Chem ; 56(21): 8872-8, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24107104

ABSTRACT

Previous studies with aminothiazolomorphinans suggested that this class of opioid ligands may be useful as a potential pharmacotherapeutic to decrease drug abuse. Novel aminothiazole derivatives of cyclorphan were prepared to evaluate a series of aminothiazolomorphinans with varying pharmacological properties at the κ opioid receptor (KOR) and µ opioid receptor (MOR). This study was focused on exploring the regioisomeric analogs with the aminothiazole on the C-ring of the morphinan skeleton. Receptor binding and [(35)S]GTPγS binding assays were used to characterize the affinity and pharmacological properties of the aminothiazolomorphinans. Intracranial self-stimulation (ICSS) was used to compare the effects of a representative aminothiazolomorphinan with the morphinan mixed-KOR/MOR agonist butorphan (MCL-101) on brain-stimulation reward.


Subject(s)
Morphinans/pharmacology , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, mu/antagonists & inhibitors , Thiazoles/pharmacology , Animals , CHO Cells , Cricetulus , Dose-Response Relationship, Drug , Humans , Male , Molecular Conformation , Morphinans/chemical synthesis , Morphinans/chemistry , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/chemistry
9.
Bioorg Med Chem Lett ; 23(7): 2128-33, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23434225

ABSTRACT

Derivatives of the lead compound N-BPE-8-CAC (1) where each CH of the biphenyl group was individually replaced by N were prepared in hopes of identifying high affinity ligands with improved aqueous solubility. Compared to 1, binding affinities of the five possible pyridinyl derivatives for the µ opioid receptor were between threefold lower to fivefold higher with the Ki of the most potent compound being 0.064 nM. Docking of 8-CAC (2) into the unliganded binding site of the mouse µ opioid receptor (pdb: 4DKL) revealed that 8-CAC and ß-FNA (from 4DKL) make nearly identical interactions with the receptor. However, for 1 and the new pyridinyl derivatives 4-8, binding is not tolerated in the 8-CAC binding mode due to the steric constraints of the large N-substituents. Either an alternative binding mode or rearrangement of the protein to accommodate these modifications may account for their high binding affinity.


Subject(s)
Biphenyl Compounds/pharmacology , Cyclopropanes/pharmacology , Pyridines/pharmacology , Receptors, Opioid/chemistry , Animals , Biphenyl Compounds/chemical synthesis , Biphenyl Compounds/chemistry , Cyclopropanes/chemical synthesis , Cyclopropanes/chemistry , Dose-Response Relationship, Drug , Humans , Ligands , Mice , Models, Molecular , Molecular Structure , Pyridines/chemical synthesis , Pyridines/chemistry , Receptors, Opioid/metabolism , Structure-Activity Relationship
10.
J Med Chem ; 55(8): 3878-90, 2012 Apr 26.
Article in English | MEDLINE | ID: mdl-22439881

ABSTRACT

A series of 3-benzylamino-3-desoxymorphinan (I) and 3-benzylamino-3-desoxymorphine (II) derivatives were synthesized and evaluated for their binding affinities, and functional activity data are presented at MOR, KOR, and DOR. Some of these ligands were found to have high binding affinity at MOR and KOR and displayed increased selectivity at MOR over KOR and DOR compared to butorphan or cyclorphan. The most selective compound, 3-(3'-hydroxybenzyl)amino-17-methylmorphinan (4g) (24-fold MOR to KOR and 1700-fold MOR to DOR) also showed high binding affinity (0.42 nM to MOR) and was a full agonist in the [(35)S]GTPγS binding assay. 2-(3'-Hydroxybenzyl)amino-17-cyclopropylmethylmorphinan (17) was found to be a KOR-selective ligand (150-fold over MOR and >10000-fold over the DORs). Most 3-benzylaminomorphinan derivatives were partial agonists at MOR and full agonists at KOR in the [(35)S]GTPγS binding assay.


Subject(s)
Benzylamines/chemical synthesis , Morphinans/chemical synthesis , Receptors, Opioid/metabolism , Animals , Benzylamines/metabolism , Binding, Competitive , CHO Cells , Cricetinae , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Kinetics , Morphinans/metabolism , Receptors, Opioid/drug effects , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Structure-Activity Relationship
11.
Chem Biol Drug Des ; 78(5): 749-56, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21801311

ABSTRACT

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.


Subject(s)
Circular Dichroism , Enkephalins/chemistry , Magnetic Resonance Spectroscopy , Peptides/chemistry , Amino Acid Sequence , Animals , Blood-Brain Barrier/metabolism , CHO Cells , Central Nervous System/metabolism , Cricetinae , Humans , Male , Mice , Mice, Inbred ICR , Pain Measurement , Peptides/metabolism , Peptides/pharmacokinetics , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/genetics , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/genetics , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Sodium Dodecyl Sulfate/chemistry , Water/chemistry
12.
J Med Chem ; 54(6): 1903-13, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21351746

ABSTRACT

A series of N-substituted and N'-substituted aminothiazole-derived morphinans (5) were synthesized for expanding the structure-activity relationships of aminothiazolo-morphinans. Although their affinities were somewhat lower than their prototype aminothiazolo-N-cyclopropylmorphinan (3), 3-aminothiazole derivatives of cyclorphan (1) containing a primary amino group displayed high affinity and selectivity at the κ and µ opioid receptors. [(35)S]GTPγS binding assays showed that the aminothiazolomorphinans were κ agonists with mixed agonist and antagonist activity at the µ opioid receptor. These novel N'-monosubstituted aminothiazole-derived morphinans may be valuable for the development of drug abuse medications.


Subject(s)
Morphinans/chemical synthesis , Receptors, Opioid, kappa/agonists , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/antagonists & inhibitors , Thiazoles/chemical synthesis , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Morphinans/chemistry , Morphinans/pharmacology , Radioligand Assay , Stereoisomerism , Structure-Activity Relationship , Thiazoles/chemistry , Thiazoles/pharmacology
13.
Pain ; 152(4): 878-887, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21316152

ABSTRACT

The search for the mechanism of action of improgan (a nonopioid analgesic) led to the recent discovery of CC12, a compound that blocks improgan antinociception. Because CC12 is a cytochrome P450 inhibitor, and brain P450 mechanisms were recently shown to be required in opioid analgesic signaling, pharmacological and transgenic studies were performed in rodents to test the hypothesis that improgan antinociception requires brain P450 epoxygenase activity. Intracerebroventricular (i.c.v.) administration of the P450 inhibitors miconazole and fluconazole, and the arachidonic acid (AA) epoxygenase inhibitor N-methylsulfonyl-6-(2-propargyloxyphenyl)hexanamide (MS-PPOH) potently inhibited improgan antinociception in rats at doses that were inactive alone. MW06-25, a new P450 inhibitor that combines chemical features of CC12 and miconazole, also potently blocked improgan antinociception. Although miconazole and CC12 were weakly active at opioid and histamine H(3) receptors, MW06-25 showed no activity at these sites, yet retained potent P450-inhibiting properties. The P450 hypothesis was also tested in Cpr(low) mice, a viable knock-in model with dramatically reduced brain P450 activity. Improgan (145 nmol, i.c.v.) antinociception was reduced by 37% to 59% in Cpr(low) mice, as compared with control mice. Moreover, CC12 pretreatment (200 nmol, i.c.v.) abolished improgan action (70% to 91%) in control mice, but had no significant effect in Cpr(low) mice. Thus, improgan's activation of bulbospinal nonopioid analgesic circuits requires brain P450 epoxygenase activity. A model is proposed in which (1) improgan activates an unknown receptor to trigger downstream P450 activity, and (2) brainstem epoxygenase activity is a point of convergence for opioid and nonopioid analgesic signaling.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Brain/drug effects , Cimetidine/analogs & derivatives , Cytochrome P-450 Enzyme System/metabolism , Gene Expression Regulation, Enzymologic/drug effects , 14-alpha Demethylase Inhibitors/pharmacology , Amides/pharmacology , Analgesics, Opioid/pharmacokinetics , Animals , Brain/metabolism , Cell Line, Transformed , Cimetidine/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Humans , Imidazoles/pharmacology , Injections, Intraventricular/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Miconazole/pharmacology , NADPH-Ferrihemoprotein Reductase/deficiency , Naltrexone/analogs & derivatives , Naltrexone/pharmacokinetics , Narcotic Antagonists/pharmacokinetics , Pain Measurement/drug effects , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Receptors, Histamine H3/metabolism , Sulfides/pharmacology , Time Factors , Tritium/pharmacokinetics
14.
Eur J Med Chem ; 46(2): 799-803, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21216504

ABSTRACT

Bifunctional ligands containing an ester linkage between morphine and the δ-selective pharmacophore Dmt-Tic were synthesized, and their binding affinity and functional bioactivity at the µ, δ and κ opioid receptors determined. Bifunctional ligands containing or not a spacer of ß-alanine between the two pharmacophores lose the µ agonism deriving from morphine becoming partial µ agonists 4 or µ antagonists 5. Partial κ agonism is evidenced only for compound 4. Finally, both compounds showed potent δ antagonism.


Subject(s)
Dipeptides/pharmacology , Esters/pharmacology , Morphine/pharmacology , Narcotic Antagonists , Tetrahydroisoquinolines/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Dipeptides/chemical synthesis , Dipeptides/chemistry , Esters/chemical synthesis , Esters/chemistry , Humans , Ligands , Molecular Conformation , Morphine/chemical synthesis , Morphine/chemistry , Stereoisomerism , Structure-Activity Relationship , Tetrahydroisoquinolines/chemical synthesis , Tetrahydroisoquinolines/chemistry
15.
ACS Chem Neurosci ; 1(2): 155-164, 2010 Feb 17.
Article in English | MEDLINE | ID: mdl-20352071

ABSTRACT

Based on a renewed importance recently attributed to bi- or multifunctional opioids, we report the synthesis and pharmacological evaluation of some analogues derived from our lead µ agonist / δ antagonist, H-Dmt-Tic-Gly-NH-Bzl. Our previous studies focused on the importance of the C-teminal benzyl function in the induction of such bifunctional activity. The introduction of some substituents in the para position of the phenyl ring (-Cl, -CH(3), partially -NO(2), inactive -NH(2)) was found to give a more potent µ agonist / antagonist effect associated with a relatively unmodified δ antagonist activity (pA(2) = 8.28-9.02). Increasing the steric hindrance of the benzyl group (using diphenylmethyl and tetrahydroisoquinoline functionalities) substantially maintained the µ agonist and δ antagonist activities of the lead compound. Finally and quite unexpectedly D-Tic2, considered as a wrong opioid message now; inserted into the reference compound in lieu of L-Tic, provided a µ agonist / δ agonist better than our reference ligand (H-Dmt-Tic-Gly-NH-Ph) and was endowed with the same pharmacological profile.

16.
Nat Neurosci ; 13(3): 284-6, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20139973

ABSTRACT

To assess the importance of brain cytochrome P450 (P450) activity in mu opioid analgesic action, we generated a mutant mouse with brain neuron-specific reductions in P450 activity; these mice showed highly attenuated morphine antinociception compared with controls. Pharmacological inhibition of brain P450 arachidonate epoxygenases also blocked morphine antinociception in mice and rats. Our findings indicate that a neuronal P450 epoxygenase mediates the pain-relieving properties of morphine.


Subject(s)
Analgesics, Opioid/pharmacology , Brain/drug effects , Cytochrome P-450 Enzyme System/drug effects , Neurons/drug effects , Pain/drug therapy , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/administration & dosage , Animals , Brain/enzymology , Brain/metabolism , Cytochrome P-450 CYP2J2 , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Female , Male , Mice , Mice, Transgenic , Morphine/administration & dosage , Morphine/pharmacology , Neural Pathways/drug effects , Neural Pathways/enzymology , Neural Pathways/metabolism , Neurons/enzymology , Neurons/metabolism , Pain/enzymology , Pain/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Time Factors
17.
J Med Chem ; 53(1): 402-18, 2010 Jan 14.
Article in English | MEDLINE | ID: mdl-19928862

ABSTRACT

The phenolic group of the potent mu and kappa opioid morphinan agonist/antagonists cyclorphan and butorphan was replaced by phenylamino and benzylamino groups including compounds with para-substituents in the benzene ring. These compounds are highly potent mu and kappa ligands, e.g., p-methoxyphenylaminocyclorphan showing a K(i) of 0.026 nM at the mu receptor and a K(i) of 0.03 nM at the kappa receptor. Phenyl carbamates and phenylureas were synthesized and investigated. Selective o-formylation of butorphan and levorphanol was achieved. This reaction opened the way to a large set of 2-substituted 3-hydroxymorphinans, including 2-hydroxymethyl-, 2-aminomethyl-, and N-substituted 2-aminomethyl-3-hydroxymorphinans. Bivalent ligands bridged in the 2-position were also synthesized and connected with secondary and tertiary aminomethyl groups, amide bonds, and hydroxymethylene groups, respectively. Although most of the 2-substituted morphinans showed considerably lower affinities compared to their parent compounds, the bivalent ligand approach led to significantly higher affinities compared to the univalent 2-substituted morphinans.


Subject(s)
Morphinans/chemical synthesis , Morphinans/pharmacology , Narcotic Antagonists , Binding Sites , Ligands , Molecular Conformation , Morphinans/chemistry , Structure-Activity Relationship
18.
J Med Chem ; 52(23): 7389-96, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-19634902

ABSTRACT

Bivalent morphinan compounds containing ester linkers were synthesized and their binding affinities at the mu, delta, and kappa opioid receptors determined. Addition of methyl groups adjacent to the hydrolytically labile ester linkage increased stability while only partially affecting binding affinity. The resulting bivalent ligands with optimized spacer length and structure show potent binding profiles with the most potent compound (4b), having K(i) values of 0.47 nM for both the mu and kappa opioid receptors, and 4a, having K(i) values of 0.95 and 0.62 nM for the mu and kappa receptors, respectively. Both 4a and 4b were partial agonists at the kappa and micro receptors in the [(35)S]GTPgammaS binding assay.


Subject(s)
Morphinans/chemistry , Morphinans/metabolism , Receptors, Opioid/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Hydrolysis , Ligands , Morphinans/chemical synthesis , Morphinans/pharmacology , Protein Binding , Receptors, Opioid/agonists
20.
Bioorg Med Chem Lett ; 18(16): 4474-6, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18674902

ABSTRACT

We synthesized several hydrophobic esters and ethers of butorphanol and assessed their affinities at opioid receptors in CHO cell membranes. Tested compounds displayed moderate to high affinities to the mu and kappa receptors. The findings accord with previous evidence of a lipophilic binding pocket in the opioid receptors that can be accessed to afford good binding affinity without the need for a phenolic hydrogen-bond donor group. The most potent (K(i)=61 pM at mu and 48 pM at kappa) novel agent was (-)-N-cyclobutylmethylmorphinan-3-yl-14-ol phenoxyacetate (4d).


Subject(s)
Butorphanol/chemistry , Butorphanol/chemical synthesis , Chemistry, Pharmaceutical/methods , Narcotic Antagonists , Receptors, Opioid/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Drug Design , Hydrophobic and Hydrophilic Interactions , Kinetics , Models, Chemical , Phenol , Protein Binding , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...