Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 51
1.
Neuropharmacology ; 166: 107718, 2020 04.
Article En | MEDLINE | ID: mdl-31351108

Binding and signaling kinetics have previously proven important in validation of biased agonism at GPCRs. Here we provide a comprehensive kinetic pharmacological comparison of clinically relevant µ-opioid receptor agonists, including the novel biased agonist oliceridine (TRV130) which is in clinical trial for pain management. We demonstrate that the bias profile observed for the selected agonists is not time-dependent and that agonists with dramatic differences in their binding kinetic properties can display the same degree of bias. Binding kinetics analyses demonstrate that buprenorphine has 18-fold higher receptor residence time than oliceridine. This is thus the largest pharmacodynamic difference between the clinically approved drug buprenorphine and the clinical candidate oliceridine, since their bias profiles are similar. Further, we provide the first pharmacological characterization of (S)-TRV130 demonstrating that it has a similar pharmacological profile as the (R)-form, oliceridine, but displays 90-fold lower potency than the (R)-form. This difference is driven by a significantly slower association rate. Finally, we show that the selected agonists are differentially affected by G protein-coupled receptor kinase 2 and 5 (GRK2 and GRK5) expression. GRK2 and GRK5 overexpression greatly increased µ-opioid receptor internalization induced by morphine, but only had modest effects on buprenorphine and oliceridine-induced internalization. Overall, our data reveal that the clinically available drug buprenorphine displays a similar pharmacological bias profile in vitro compared to the clinical candidate drug oliceridine and that this bias is independent of binding kinetics suggesting a mechanism driven by receptor-conformations. This article is part of the Special Issue entitled 'New Vistas in Opioid Pharmacology'.


Analgesics, Opioid/pharmacokinetics , Receptors, Opioid, mu/agonists , Signal Transduction/drug effects , Spiro Compounds/pharmacokinetics , Thiophenes/pharmacokinetics , Amino Acid Sequence , Analgesics, Opioid/metabolism , Buprenorphine/metabolism , Buprenorphine/pharmacokinetics , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/metabolism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , HEK293 Cells , Humans , Kinetics , Morphine/metabolism , Morphine/pharmacokinetics , Protein Binding/physiology , Receptors, Opioid, mu/metabolism , Signal Transduction/physiology , Spiro Compounds/metabolism , Thiophenes/metabolism
2.
PLoS One ; 14(6): e0218680, 2019.
Article En | MEDLINE | ID: mdl-31220174

Roux-en-Y gastric bypass surgery (RYGB) is the most common and effective weight loss procedure for severe obesity. However, a significant increase in addictive behaviors and new-onset substance use disorder (SUD) are sometimes observed post-surgery. The endogenous opioid system is known to play a major role in motivated behavior and reward, as well as the abuse of substances, including alcohol, tobacco, opioids and highly palatable foods. Here, we examined the effects of RYGB on mu-opioid receptor levels in the brain. Male Sprague-Dawley rats were assigned to one of four groups: standard diet with sham surgery (control), ad libitum high-energy high-fat (HF) diet with sham surgery, calorie restricted HF diet with sham surgery (Sham-FR), or HF diet with RYGB surgery. Control and HF groups were fed their respective diets for 8 weeks, with surgery performed on the eighth week. After 9 weeks on their respective diets post-surgery, animals were sacrificed for mu-opioid receptor autoradiography using the [3H] [D-Ala2,N-Me-Phe4-Gly5-ol]- enkephalin (DAMGO) ligand. Rats with RYGB showed reduced DAMGO binding in the central amygdala compared to sham-operated HF diet controls, and in the hypothalamus compared to high-fat fed Sham-FR. Diet alone did not change [3H] DAMGO binding in any region. These findings show that RYGB surgery, independent of diet or caloric restriction, decreases mu opioid signaling in specific regions important for stress and energy regulation. Thus, RYGB surgery may lead to greater stress sensitivity via downregulated mu opioid signaling in the central amygdala, which may contribute to the observed increased risk in some subjects for addictive behavior.


Brain/metabolism , Energy Metabolism/physiology , Gastric Bypass , Obesity, Morbid/surgery , Receptors, Opioid, mu/metabolism , Stress, Psychological/metabolism , Animals , Brain/pathology , Diet, High-Fat , Down-Regulation , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/metabolism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Gastric Bypass/methods , Male , Obesity, Morbid/etiology , Obesity, Morbid/metabolism , Obesity, Morbid/pathology , Rats , Rats, Sprague-Dawley , Stress, Psychological/complications , Stress, Psychological/surgery , Tritium/metabolism , Tritium/pharmacokinetics , Weight Loss/physiology
3.
Brain Res ; 1680: 105-109, 2018 02 01.
Article En | MEDLINE | ID: mdl-29269051

The analgesic mechanisms of mu opioid receptor (MOR) agonists, including receptor occupancy at the site of action, are not completely understood. The aims of the present study were to evaluate: (i) receptor occupancy in the rat brain after administration of MOR agonists; (ii) the relationship between occupancy and the antinociceptive effect. Morphine (2 or 4 mg/kg) or oxycodone (1 or 3 mg/kg) was subcutaneously administered to rats. The antinociceptive effect of these drugs was measured by the hot-plate test. MOR occupancy in the thalamus was assessed by conducting an ex vivo receptor binding assay using [3H] [D-Ala2, N-MePhe4, Gly-ol]-enkephalin, followed by autoradiographic analysis. Both drugs produced antinociception in a dose-dependent manner, and these effects disappeared after the time point at which the maximal effect was elicited. Thalamic MOR occupancy was observed in a dose-dependent manner at the time point at which maximal antinociception was elicited, and relatively low occupancy was observed when the antinociceptive effect was decreasing. Good correlation between thalamic MOR occupancy and the antinociceptive effect was observed. These findings provide direct evidence for the receptor occupancy of MOR agonists at the site of action and its relationship with the analgesic effect.


Analgesics, Opioid/pharmacology , Nociception/drug effects , Protein Binding/drug effects , Receptors, Opioid, mu/metabolism , Thalamus/drug effects , Thalamus/metabolism , Animals , Autoradiography , Dose-Response Relationship, Drug , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Male , Morphine/pharmacology , Oxycodone/pharmacology , Pain Measurement , Rats , Rats, Sprague-Dawley , Thalamus/diagnostic imaging , Time Factors , Tritium/pharmacokinetics
4.
Eur J Pharm Sci ; 92: 173-82, 2016 Sep 20.
Article En | MEDLINE | ID: mdl-27393342

The use of nanocarriers is an intriguing solution to increase the brain delivery of novel therapeutics. The aim of this paper was to use pharmacokinetic analysis and simulations to identify key factors that determine the effective drug concentration-time profile at the target site in the brain. Model building and simulations were based on experimental data obtained from the administration of the opioid peptide DAMGO in glutathione tagged PEGylated liposomes to rats. Different pharmacokinetic models were investigated to explore the mechanisms of increased brain delivery. Concentration-time profiles for a set of formulations with varying compound and carrier characteristics were simulated. By controlling the release rate from the liposome, the time profile and the extent of brain delivery can be regulated. The modeling did not support a mechanism of the liposomes passing the brain endothelial cell membrane in an intact form through endocytosis or transcytosis. The most likely process was found to be fusion of the liposome with the endothelial luminal membrane. The simulations revealed that low permeable compounds, independent on efflux, will gain the most from a nanocarrier formulation. The present model based approach is useful to explore and predict possibilities and limitations of carrier-based systems to the brain.


Analgesics, Opioid/pharmacokinetics , Brain/metabolism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Models, Biological , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/chemistry , Animals , Biological Transport , Computer Simulation , Drug Compounding , Endothelial Cells/metabolism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/administration & dosage , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/chemistry , Glutathione/chemistry , Liposomes , Nanostructures/chemistry , Polyethylene Glycols/chemistry , Rats
5.
Pharm Res ; 33(1): 177-85, 2016 Jan.
Article En | MEDLINE | ID: mdl-26275529

PURPOSE: The purpose of this study was to evaluate formulation factors causing improvement in brain delivery of a small peptide after encapsulation into a targeted nanocarrier in vivo. METHODS: The evaluation was performed in rats using microdialysis, which enabled continuous sampling of the released drug in both the brain (striatum) and blood. Uptake in brain could thereby be studied in terms of therapeutically active, released drug. RESULTS: We found that encapsulation of the peptide DAMGO in fast-releasing polyethylene glycol (PEG)ylated liposomes, either with or without the specific brain targeting ligand glutathione (GSH), doubled the uptake of DAMGO into the rat brain. The increased brain delivery was observed only when the drug was encapsulated into the liposomes, thus excluding any effects of the liposomes themselves on the blood-brain barrier integrity as a possible mechanism. The addition of a GSH coating on the liposomes did not result in an additional increase in DAMGO concentrations in the brain, in contrast to earlier studies on GSH coating. This may be caused by differences in the characteristics of the encapsulated compounds and the composition of the liposome formulations. CONCLUSIONS: We were able to show that encapsulation into PEGylated liposomes of a peptide with limited brain delivery could double the drug uptake into the brain without using a specific brain targeting ligand.


Brain/drug effects , Brain/metabolism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/administration & dosage , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Glutathione/chemistry , Liposomes/chemistry , Polyethylene Glycols/chemistry , Animals , Drug Carriers , Drug Compounding , Drug Delivery Systems , Male , Microdialysis , Neostriatum/metabolism , Phosphatidylcholines/chemistry , Rats , Rats, Sprague-Dawley
6.
Exp Brain Res ; 232(6): 1793-803, 2014 Jun.
Article En | MEDLINE | ID: mdl-24584836

The olfactory bulbectomized (OB) rat, an animal model of chronic depression with comorbid anxiety, exhibits a profound dysregulation of the brain serotonergic signalling, a neurotransmission system involved in pain transmission and modulation. We here report an increased nociceptive response of OB rats in the tail flick test which is reverted after chronic, but not acute, administration of fluoxetine. Autoradiographic studies demonstrated down-regulation of 5-HT transporters ([(3)H]citalopram binding) and decreased functionality of 5-HT1A receptors (8-OH-DPAT-stimulated [(35)S]GTPγS binding) in the dorsal horn of the lumbar spinal cord in OB rats. Acute administration of fluoxetine (5-40 mg/kg i.p.) did not modify tail flick latencies in OB rats. However, chronic fluoxetine (10 mg/kg/day s.c., 14 days; osmotic minipumps) progressively attenuated OB-associated thermal hyperalgesia, and a total normalization of the nociceptive response was achieved at the end of the treatment with the antidepressant. In these animals, autoradiographic studies revealed further down-regulation of 5-HT transporters and normalization in the functionality of 5-HT1A receptors on the spinal cord. On the other hand, acute morphine (0.5-10 mg/kg s.c.) produced a similar analgesic effect in OB and sham and OB rats, and no changes were detected in the density ([(3)H]DAMGO binding) and functionality (DAMGO-stimulated [(35)S]GTPγS binding) of spinal µ-opioid receptors in OB rats before and after chronic fluoxetine. Our findings demonstrate the participation of the spinal serotonergic system in the increased thermal nociception exhibited by the OB rat and the antinociceptive effect of chronic fluoxetine in this animal model of depression.


Depression/complications , Depression/pathology , Hyperalgesia/etiology , Serotonin/metabolism , Spinal Cord/metabolism , Animals , Antidepressive Agents/therapeutic use , Autoradiography , Depression/drug therapy , Disease Models, Animal , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Exploratory Behavior/physiology , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics , Hyperalgesia/drug therapy , Male , Morphine/therapeutic use , Neurotransmitter Agents/pharmacokinetics , Olfactory Bulb/injuries , Olfactory Bulb/surgery , Pain Measurement , Radiography , Radioisotopes/pharmacokinetics , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1A/metabolism , Receptors, Opioid, mu/metabolism , Serotonin Agents/pharmacokinetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Spinal Cord/diagnostic imaging , Spinal Cord/drug effects
7.
Neuropharmacology ; 72: 9-19, 2013 Sep.
Article En | MEDLINE | ID: mdl-23624290

The pharmacotherapy for the treatment of pain is an active area of investigation. There are effective drugs to treat this problem, but there is also a need to find alternative treatments free of undesirable side effects. In the present work the capacity of a series of flavonoids to bind to the µ opioid receptor was evaluated. The most active compound, 3,3-dibromoflavanone (31), a synthetic flavonoid, presented a significant inhibition of the binding of the selective µ opioid ligand [(3)H]DAMGO, with a Ki of 0.846 ± 0.263 µM. Flavanone 31 was further synthesized using a simple and cheap procedure with good yield. Its in vivo effects in mice, after acute treatments, were studied using antinociceptive and behavioral assays. It showed no sedative, anxiolytic, motor incoordination effects or inhibition of the gastrointestinal transit in mice at the doses tested. It evidenced antinociceptive activity on the acetic acid-induced nociception, hot plate and formalin tests (at 10 mg/kg and 30 mg/kg). The results showed that the 5-HT2 receptor and the adrenoceptors seem unlikely to be involved in its antinociceptive effects. Naltrexone, a nonselective opioid receptors antagonist, totally blocked compound 31 antinociceptive effects on the hot plate test, but naltrindole (δ opioid antagonist) and nor-binaltorphimine (κ opioid antagonist) did not. These findings demonstrated that 3,3-dibromoflavanone (31), at doses that did not interfere with the motor performance, exerted clear dose dependent antinociception when assessed in the chemical and thermal models of nociception in mice and it seems that its action is related to the activation of the µ opioid receptor.


Analgesics/therapeutic use , Flavonoids/therapeutic use , Receptors, Opioid, mu/metabolism , Visceral Pain/metabolism , Acetic Acid/pharmacology , Analgesics/pharmacology , Analgesics, Opioid/pharmacokinetics , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Compounding , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Flavanones/pharmacology , Flavanones/therapeutic use , Flavonoids/chemistry , Flavonoids/pharmacology , Gastrointestinal Transit/drug effects , Male , Maze Learning/drug effects , Mice , Motor Activity/drug effects , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Pain Measurement/drug effects , Pain Measurement/methods , Protein Binding/drug effects , Tritium/pharmacokinetics , Visceral Pain/diagnosis , Visceral Pain/drug therapy
8.
J Neurosci ; 33(10): 4369-77, 2013 Mar 06.
Article En | MEDLINE | ID: mdl-23467353

Regulator of G-protein signaling (RGS) proteins classically function as negative modulators of G-protein-coupled receptor signaling. In vitro, RGS proteins have been shown to inhibit signaling by agonists at the µ-opioid receptor, including morphine. The goal of the present study was to evaluate the contribution of endogenous RGS proteins to the antinociceptive effects of morphine and other opioid agonists. To do this, a knock-in mouse that expresses an RGS-insensitive (RGSi) mutant Gαo protein, Gαo(G184S) (Gαo RGSi), was evaluated for morphine or methadone antinociception in response to noxious thermal stimuli. Mice expressing Gαo RGSi subunits exhibited a naltrexone-sensitive enhancement of baseline latency in both the hot-plate and warm-water tail-withdrawal tests. In the hot-plate test, a measure of supraspinal nociception, morphine antinociception was increased, and this was associated with an increased ability of opioids to inhibit presynaptic GABA neurotransmission in the periaqueductal gray. In contrast, antinociception produced by either morphine or methadone was reduced in the tail-withdrawal test, a measure of spinal nociception. In whole-brain and spinal cord homogenates from mice expressing Gαo RGSi subunits, there was a small loss of Gαo expression and an accompanying decrease in basal G-protein activity. Our results strongly support a role for RGS proteins as negative regulators of opioid supraspinal antinociception and also reveal a potential novel function of RGS proteins as positive regulators of opioid spinal antinociceptive pathways.


Analgesics, Opioid/therapeutic use , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Hyperalgesia/drug therapy , Morphine/therapeutic use , RGS Proteins/metabolism , Analgesics, Opioid/pharmacology , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Diprenorphine/pharmacokinetics , Dose-Response Relationship, Drug , Electric Stimulation , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Excitatory Amino Acid Antagonists/pharmacology , Female , GABA Antagonists/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics , Hot Temperature/adverse effects , Humans , Hyperalgesia/genetics , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Isotopes/pharmacokinetics , Male , Methadone/pharmacology , Methadone/therapeutic use , Mice , Mice, Transgenic , Morphine/pharmacology , Mutation , Naloxone/pharmacology , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Neurons/drug effects , Neurons/physiology , Oligopeptides/pharmacology , Pain Measurement/drug effects , Patch-Clamp Techniques , Pertussis Toxin/pharmacology , Protein Binding/drug effects , Protein Binding/genetics , RGS Proteins/genetics , Reaction Time/drug effects , Reaction Time/genetics , Signal Transduction/drug effects , Spinal Cord/drug effects , Spinal Cord/metabolism , gamma-Aminobutyric Acid/metabolism
9.
J Neurosci ; 33(9): 4118-27, 2013 Feb 27.
Article En | MEDLINE | ID: mdl-23447620

Prolonged exposure to high-efficacy agonists results in desensitization of the µ-opioid receptor (MOR). Desensitized receptors are thought to be unable to couple to G-proteins, preventing downstream signaling; however, the changes to the receptor itself are not well characterized. In the current study, confocal imaging was used to determine whether desensitizing conditions cause a change in agonist-receptor interactions. Using rapid solution exchange, the binding kinetics of fluorescently labeled opioid agonist, dermorphin Alexa594 (derm A594), to MORs was measured in live cells. The affinity of derm A594 binding increased after prolonged treatment of cells with multiple agonists that are known to cause receptor desensitization. In contrast, binding of a fluorescent antagonist, naltrexamine Alexa594, was unaffected by similar agonist pretreatment. The increased affinity of derm A594 for the receptor was long-lived and partially reversed after a 45 min wash. Treatment of the cells with pertussis toxin did not alter the increase in affinity of the derm A594 for MOR. Likewise, the affinity of derm A594 for MORs expressed in mouse embryonic fibroblasts derived from arrestin 1 and 2 knock-out animals increased after treatment of the cells with the desensitization protocol. Thus, opioid receptors were "imprinted" with a memory of prior agonist exposure that was independent of G-protein activation or arrestin binding that altered subsequent agonist-receptor interactions. The increased affinity suggests that acute desensitization results in a long-lasting but reversible conformational change in the receptor.


Cell Membrane/metabolism , Pharmacological Phenomena/drug effects , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/pharmacokinetics , Analysis of Variance , Animals , Arrestin/deficiency , Arrestin/metabolism , Cell Membrane/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Embryo, Mammalian , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression/drug effects , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , HEK293 Cells , Humans , Ligands , Mice , Mice, Knockout , Morphine/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/pharmacokinetics , Narcotic Antagonists/pharmacokinetics , Opioid Peptides/pharmacokinetics , Organic Chemicals/pharmacokinetics , Pertussis Toxin/pharmacology , Protein Binding/drug effects , Protein Conformation/drug effects , Radioligand Assay , Receptors, Opioid, mu/genetics , Substrate Specificity/drug effects , Time Factors , Transfection , Tritium/pharmacokinetics
10.
Pain ; 154(4): 598-608, 2013 Apr.
Article En | MEDLINE | ID: mdl-23398938

Hyperalgesia is a cardinal symptom of opioid withdrawal. The transient receptor potential vanilloid 1 (TRPV1) is a ligand-gated ion channel expressed on sensory neurons responding to noxious heat, protons, and chemical stimuli such as capsaicin. TRPV1 can be inhibited via µ-opioid receptor (MOR)-mediated reduced activity of adenylyl cyclases (ACs) and decreased cyclic adenosine monophosphate (cAMP) levels. In contrast, opioid withdrawal following chronic activation of MOR uncovers AC superactivation and subsequent increases in cAMP and protein kinase A (PKA) activity. Here we investigated (1) whether an increase in cAMP during opioid withdrawal increases the activity of TRPV1 and (2) how opioid withdrawal modulates capsaicin-induced nocifensive behavior in rats. We applied whole-cell patch clamp, microfluorimetry, cAMP assays, radioligand binding, site-directed mutagenesis, and behavioral experiments. Opioid withdrawal significantly increased cAMP levels and capsaicin-induced TRPV1 activity in both transfected human embryonic kidney 293 cells and dissociated dorsal root ganglion (DRG) neurons. Inhibition of AC and PKA, as well as mutations of the PKA phosphorylation sites threonine 144 and serine 774, prevented the enhanced TRPV1 activity. Finally, capsaicin-induced nocifensive behavior was increased during opioid withdrawal in vivo. In summary, our results demonstrate an increased activity of TRPV1 in DRG neurons as a new mechanism contributing to opioid withdrawal-induced hyperalgesia.


Analgesics, Opioid/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Sensory Receptor Cells/drug effects , TRPV Cation Channels/metabolism , Animals , Calcium/metabolism , Capsaicin/adverse effects , Cells, Cultured , Cyclic AMP/metabolism , Disease Models, Animal , Diterpenes/pharmacokinetics , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Enzyme Inhibitors/pharmacology , Fentanyl/pharmacology , Ganglia, Spinal/cytology , Humans , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Morphine/pharmacology , Mutagenesis, Site-Directed , Protein Binding/drug effects , Rats , Receptors, Opioid, mu/metabolism , Sensory Receptor Cells/metabolism , Substance Withdrawal Syndrome/metabolism , TRPV Cation Channels/genetics , Tritium/pharmacokinetics
11.
Mol Pharm ; 10(5): 1533-41, 2013 May 06.
Article En | MEDLINE | ID: mdl-22934681

Glutathione PEGylated (GSH-PEG) liposomes were evaluated for their ability to enhance and prolong blood-to-brain drug delivery of the opioid peptide DAMGO (H-Tyr-d-Ala-Gly-MePhe-Gly-ol). An intravenous loading dose of DAMGO followed by a 2 h constant rate infusion was administered to rats, and after a washout period of 1 h, GSH-PEG liposomal DAMGO was administered using a similar dosing regimen. DAMGO and GSH-PEG liposomal DAMGO were also administered as a 10 min infusion to compare the disposition of the two formulations. Microdialysis made it possible to determine free DAMGO in brain and plasma, while the GSH-PEG liposomal encapsulated DAMGO was measured with regular plasma sampling. The antinociceptive effect of DAMGO was determined with the tail-flick method. All samples were analyzed using liquid chromatography-tandem mass spectrometry. The short infusion of DAMGO resulted in a fast decline of the peptide concentration in plasma with a half-life of 9.2 ± 2.1 min. Encapsulation in GSH-PEG liposomes prolonged the half-life to 6.9 ± 2.3 h. Free DAMGO entered the brain to a limited extent with a steady state ratio between unbound drug concentrations in brain interstitial fluid and in blood (Kp,uu) of 0.09 ± 0.04. GSH-PEG liposomes significantly increased the brain exposure of DAMGO to a Kp,uu of 0.21 ± 0.17 (p < 0.05). By monitoring the released, active substance in both blood and brain interstitial fluid over time, we were able to demonstrate that GSH-PEG liposomes offer a promising platform for enhancing and prolonging the delivery of drugs to the brain.


Analgesics, Opioid/administration & dosage , Brain/metabolism , Drug Delivery Systems , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/administration & dosage , Analgesics, Opioid/pharmacokinetics , Animals , Blood-Brain Barrier , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Glutathione/administration & dosage , Half-Life , Infusions, Intravenous , Liposomes/administration & dosage , Male , Microdialysis , Polyethylene Glycols/administration & dosage , Rats , Rats, Sprague-Dawley
12.
Article En | MEDLINE | ID: mdl-22672845

A liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS) method for the quantification of the opioid peptide DAMGO in rat plasma, as well as DAMGO and the microdialysis recovery calibrator [(13)C(2),(15)N]-DAMGO in microdialysis samples, is described. The microdialysis samples consisted of 15 µL Ringer solution containing 0.5% bovine serum albumin. Pretreatment of the samples involved protein precipitation with acetonitrile followed by dilution with 0.01% formic acid. The lower limits of quantification were 0.52 ng/mL and 0.24 ng/mL for DAMGO and [(13)C(2),(15)N]-DAMGO respectively and the response was linear up to 5000 fold higher concentrations. The plasma samples (50 µL) were precipitated with acetonitrile containing the isotope labeled analog [(13)C(2),(15)N]-DAMGO as internal standard. The method was linear in the range of 11-110,000 ng/mL. The separations were conducted on a HyPurity C18 column, 50×4.6 mm, 3 µm particle size, with a mobile phase consisting of acetonitrile, water and formic acid to the proportions of 17.5:82.5:0.01. Low energy collision dissociation tandem mass spectrometric (CID-MS/MS) analysis was carried out in the positive ion mode using multiple reaction monitoring (MRM) of the following mass transitions: m/z 514.2→453.2 for DAMGO and m/z 517.2→456.2 for [(13)C(2),(15)N]-DAMGO. The intra-day precision and accuracy did not exceed 5.2% and 93-104% for both compounds and sample types described. The inter-day precision an accuracy were <6.8% and 95-105% respectively. The method described is simple, reproducible and suitable for the analysis of small sample volumes at low concentrations.


Chromatography, Liquid/methods , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/blood , Microdialysis/methods , Tandem Mass Spectrometry/methods , Animals , Drug Stability , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Linear Models , Male , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Sensitivity and Specificity
13.
Neuropharmacology ; 61(8): 1229-38, 2011 Dec.
Article En | MEDLINE | ID: mdl-21803061

The biological effects of endomorphins (EMs) are short-lasting due to their rapid degradation by endogenous enzymes. Competing enzymatic degradation is an approach to prolong EM bioavailability. In the present study, a series of tetra- and tripeptides of similar to EMs structure was synthesized and tested in vitro and in vivo for their ability to inhibit degradation of EMs. The obtained results indicated that, among the series of analogs, the tetrapeptide Tyr-Pro-d-ClPhe-Phe-NH(2) and the tripeptide Tyr-Pro-Ala-NH(2), which did not bind to the µ-opioid receptors, were potent inhibitors of EM catabolism in rat brain homogenate. In vivo, these two peptides significantly prolonged the analgesic and antidepressant-like effects, induced by exogenous EMs, by blocking EM degrading enzymes. These new potent inhibitors may therefore increase the level and the half life of endogenous EMs and could be used in a new therapeutic strategy against pain and mood disorders, based on increasing of EM bioavailability.


Analgesics, Opioid/pharmacology , Antidepressive Agents/therapeutic use , Depression/drug therapy , Hyperalgesia/drug therapy , Oligopeptides/pharmacology , Analysis of Variance , Animals , Brain/drug effects , Brain/metabolism , Dipeptidyl Peptidase 4/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Injections, Intraventricular , Male , Mice , Motor Activity/drug effects , Oligopeptides/metabolism , Oligopeptides/pharmacokinetics , Pain Measurement/drug effects , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Swimming/psychology , Tritium/pharmacokinetics
14.
J Pharmacol Sci ; 110(1): 36-46, 2009 May.
Article En | MEDLINE | ID: mdl-19443999

In this study, we compared the effects of five short-, medium-, or long-acting benzodiazepine-receptor agonists (BZDs) [alprazolam (APZ), clonazepam (CLZ), flunitrazepam (FLZ), loprazolam (LPZ), zolpidem (ZLP)], at two distinct doses, 0.2 and 2 mg/kg, on the cell surface regulation of mu-opioid receptor induced by 0.15 mg/kg buprenorphine (BPN) in specific regions of the rat brain. Using 0.312 - 5 nM [(3)H]-DAMGO concentrations and Scatchard plot analysis, B(max) (maximal receptor density) and K(d) (dissociation constant) were determined at different brain regions of interest (amygdala, cortex, hippocampus, hypothalamus, thalamus). Acute BPN induced an expected down-regulation and addition of each of the BZDs to BPN induced less down-regulation than did BPN alone, sometimes while altering affinity. Some significant differences in the intensity of these effects were observed between BZDs. FLZ that is widely abused and enlarges BPN toxicity appeared the most potent to increase mu-cell surface receptor density at the lowest dose of 0.2 mg/kg. Besides, LPZ for which the effect on mu-opioid-receptor regulation appeared lower is considered to have a low risk of dependence in the epidemiological data banks. CLZ and ZLP (2 mg/kg) induced the strongest modification on mu-opioid-receptor density, but a substantial decrease in affinity could minimize the functional consequences. The reported changes were maximal in the amygdala, hippocampus, and thalamus. Among people using BPN and BZDs, the effects described here are likely to influence addictive behaviors and induce toxic effects that could be quantitatively different due to the quality of the BZD.


Analgesics, Opioid/pharmacology , Benzodiazepines/pharmacology , Buprenorphine/pharmacology , GABA-A Receptor Agonists , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/pharmacokinetics , Animals , Binding Sites , Brain Chemistry/drug effects , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/metabolism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Kinetics , Male , Rats , Rats, Wistar
15.
Neuroscience ; 156(4): 857-64, 2008 Oct 28.
Article En | MEDLINE | ID: mdl-18804151

Behavioral sensitization to ethanol is characterized by an increased locomotor activity after repeated exposure. A great variability exists among species and strains in the development of sensitization. There is a growing amount of evidence to indicate that the opioid system is involved in alcoholism; it is possible, therefore, that this system also modulates the sensitization to ethanol. In this study we evaluated the role of the opioid system in determining the variability of the sensitized response to ethanol. Mice received repeated administrations of ethanol (2.2 g/kg) or saline every other day for 10 days. According to their locomotor response on the last day of treatment, ethanol-treated animals were classified into two groups: sensitized or non-sensitized mice. After the treatment, mice were submitted to four challenges 48 h apart. In experiments 1 and 2, mice were challenged, respectively, with i.p. administration of opioid antagonists (naloxone or naltrexone) or an opioid agonist (morphine), followed immediately by 2.2 g/kg ethanol. In experiment 3, animals received morphine by i.c.v., followed by 2.2 g/kg of ethanol (i.p.). Pretreatment with opioid antagonists (naloxone or naltrexone) did not block the expression of ethanol sensitization; however pretreatment with morphine attenuated the increased locomotor activity after ethanol administration in sensitized mice. In experiment 4, after the ethanol or saline treatment, mice brains were processed and brain mu opioid binding was assessed by autoradiography using [3H]D-Ala2,N-mePhe4, Gly-ol5-enkephalin ([3H]DAMGO). No differences were seen between any of the groups of mice, so the agonist effect is not likely to be mediated by differences in binding to mu opioid receptors.


Analgesics, Opioid/pharmacology , Behavior, Animal/drug effects , Central Nervous System Depressants/administration & dosage , Ethanol/administration & dosage , Morphine/pharmacology , Analysis of Variance , Animals , Autoradiography , Brain/drug effects , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Male , Naloxone/pharmacology , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Protein Binding/drug effects , Rats , Rats, Wistar , Tritium/pharmacokinetics
16.
J Neurochem ; 103(1): 77-87, 2007 Oct.
Article En | MEDLINE | ID: mdl-17877633

The most common single nucleotide polymorphism in the coding region of the human mu opioid receptor gene is the A118G variant, an adenine to guanine transition at nucleotide position 118 of the coding sequence of the gene. This polymorphism codes for an asparagine to aspartic acid substitution at amino acid 40 in the amino-terminus, thereby removing a potential extracellular glycosylation site. Using in vitro cellular expression assays, this variant has been reported to change binding of the endogenous agonist beta-endorphin and signaling of the receptor following binding of beta-endorphin. Three clinical studies report that A118G genotype affects opioid antagonist-mediated increases in cortisol levels. These studies demonstrate a functional role of this variant in responses to endogenous and exogenous opioids. To further characterize function, we expressed the prototype and variant receptors in two types of cells (human 293 embryonic kidney cells and Syrian hamster adenovirus-12-induced tumor cells). Stable expression of variant and prototype receptors was characterized by differences in levels of cell surface binding capacity (B(max)), forskolin-induced cAMP accumulation, as well as agonist-induced accumulation of cAMP (EC(50)) for several agonists, but not for beta-endorphin. In contrast, transiently expressed variant receptors showed only a minor difference in cell surface binding capacity compared to the prototype, and no differences in cAMP EC(50) values.


Polymorphism, Single Nucleotide/genetics , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/pharmacokinetics , Animals , Binding, Competitive/drug effects , Binding, Competitive/genetics , Cell Line , Cell Membrane/metabolism , Colforsin/pharmacology , Cricetinae , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Fibroblasts/metabolism , Gene Transfer Techniques , Humans , Kidney/cytology , Kidney/metabolism , Mesocricetus , Receptors, Opioid, mu/drug effects
17.
Drug Alcohol Depend ; 89(2-3): 282-91, 2007 Jul 10.
Article En | MEDLINE | ID: mdl-17368966

Lobeline diminishes the behavioral and neurochemical effects of nicotine and amphetamines, and is considered a potential pharmacotherapy for drug abuse and addiction. Lobeline has high affinity for nicotinic acetylcholine receptors and inhibits the function of vesicular monoamine and dopamine transporters. The present study investigated the less-explored interaction of lobeline and the endogenous opioid system. In guinea pig brain homogenates, lobeline displaced (K(i)=0.74 microM) the binding of [(3)H]DAMGO [(D-Ala(2), N-ME-Phe(4), Gly(5)-ol)-enkephalin]. In a functional assay system comprised of MOR-1 mu opioid receptors and GIRK2 potassium channels expressed in Xenopus oocytes, lobeline had no effect on the resting current, but maximally inhibited (IC(50)=1.1 microM) morphine- and DAMGO-activated potassium current in a concentration-dependent manner. In a second functional assay, lobeline-evoked [(3)H]overflow from rat striatal slices preloaded with [(3)H]dopamine was not blocked by naltrexone. Importantly, concentrations of lobeline (0.1-0.3 microM) that did not have intrinsic activity attenuated ( approximately 50%) morphine-evoked [(3)H]overflow. Overall, the results suggest that lobeline functions as a mu opioid receptor antagonist. The ability of lobeline to block psychostimulant effects may be mediated by opioid receptor antagonism, and lobeline could be investigated as a treatment for opiate addiction.


Brain/drug effects , Lobeline/pharmacology , Morphine/pharmacology , Narcotic Antagonists/pharmacology , Narcotics/pharmacology , Receptors, Opioid, mu/drug effects , Receptors, Opioid/drug effects , Analgesics, Opioid/pharmacokinetics , Animals , Binding, Competitive , Dopamine/metabolism , Dose-Response Relationship, Drug , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Guinea Pigs , In Vitro Techniques , Mecamylamine/pharmacology , Naltrexone/pharmacology , Nicotine/pharmacology , Oocytes , Radioligand Assay , Receptors, Opioid/metabolism , Receptors, Opioid, mu/metabolism , Xenopus laevis
18.
Neuropsychopharmacology ; 32(3): 607-15, 2007 Mar.
Article En | MEDLINE | ID: mdl-16823391

Cannabis use is a hypothesized gateway to subsequent abuse of other drugs such as heroin. We currently assessed whether Delta-9-tetrahydrocannabinol (THC) exposure during adolescence modulates opiate reinforcement and opioid neural systems in adulthood. Long-Evan male rats received THC (1.5 mg/kg intraperitoneally (i.p.)) or vehicle every third day during postnatal days (PNDs) 28-49. Heroin self-administration behavior (fixed ratio-1; 3-h sessions) was studied from young adulthood (PND 57) into full adults (PND 102). THC-pretreated rats showed an upward shift throughout the heroin self-administration acquisition (30 microg/kg/infusion) phase, whereas control animals maintained the same pattern once stable intake was obtained. Heightened opiate sensitivity in THC animals was also evidenced by higher heroin consumption during the maintenance phase (30 and 60 microg/kg/infusion) and greater responding for moderate-low heroin doses (dose-response curve: 7.5, 15, 30, 60, and 100 microg/kg/injection). Specific disturbance of the endogenous opioid system was also apparent in the brain of adults with adolescent THC exposure. Striatal preproenkephalin mRNA expression was exclusively increased in the nucleus accumbens (NAc) shell; the relative elevation of preproenkephalin mRNA in the THC rats was maintained even after heroin self-administration. Moreover, mu opioid receptor (muOR) GTP-coupling was potentiated in mesolimbic and nigrostriatal brainstem regions in THC-pretreated animals. muOR function in the NAc shell was specifically correlated to heroin intake. The current findings support the gateway hypothesis demonstrating that adolescence cannabis exposure has an enduring impact on hedonic processing resulting in enhanced opiate intake, possibly as a consequence of alterations in limbic opioid neuronal populations.


Analgesics, Non-Narcotic/pharmacology , Analgesics, Opioid/administration & dosage , Dronabinol/pharmacology , Heroin/administration & dosage , Limbic System/cytology , Neurons/drug effects , Analysis of Variance , Animals , Animals, Newborn , Benzoxazines , Dose-Response Relationship, Drug , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Enkephalins/genetics , Enkephalins/metabolism , Gene Expression Regulation/drug effects , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics , In Situ Hybridization , Male , Morpholines/pharmacokinetics , Naphthalenes/pharmacokinetics , Neurons/metabolism , Piperidines/pharmacology , Protein Precursors/genetics , Protein Precursors/metabolism , Pyrazoles/pharmacology , Radioligand Assay/methods , Rats , Rats, Long-Evans , Reinforcement, Psychology , Rimonabant , Self Administration/methods , Sulfur Isotopes/pharmacokinetics , Tritium/pharmacokinetics
19.
Neuropharmacology ; 52(3): 931-48, 2007 Mar.
Article En | MEDLINE | ID: mdl-17161852

Lewis (LEW) and Fischer 344 (F344) rats show differential morphine self-administration rates. In this study, after animals of both strains self-administered morphine (1mg/kg) or extinguished this behaviour for 3, 7 or 15days, we measured the binding to, and functional state of mu opioid receptors (MORs) as well as proenkephalin (PENK) mRNA content in several brain regions. The results showed that in most brain areas: 1) LEW rats had less binding to MORs in basal conditions than F344 rats; 2) after morphine self-administration, either one of the strains or both (depending on the brain area) showed increased levels of binding to MORs as compared to basal groups; and 3) these binding levels in morphine self-administration animals came down in each extinction group. Moreover, F344 rats exhibited, in general, an increased functionality of MORs after morphine self-administration, as compared to basal groups, which also went down during extinction. Finally, the basal content of PENK mRNA was lower in LEW rats than in F344 rats and it decreased more after self-administration; during extinction, the levels of PENK mRNA got normalized in this strain. This differential modulation of the endogenous opioid system might be related to the different rates of morphine self-administration behavior exhibited by both inbred rat strains.


Enkephalins/metabolism , Extinction, Psychological/drug effects , Morphine/administration & dosage , Narcotics/administration & dosage , Protein Precursors/metabolism , Receptors, Opioid/metabolism , Analysis of Variance , Animals , Autoradiography , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Enkephalins/genetics , In Situ Hybridization/methods , Male , Narcotics/pharmacokinetics , Protein Precursors/genetics , Rats , Rats, Inbred F344 , Rats, Inbred Lew , Self Administration/methods , Time Factors , Tritium/pharmacokinetics
20.
Neuroscience ; 142(2): 481-91, 2006 Oct 13.
Article En | MEDLINE | ID: mdl-16893609

Numerous studies support a role for the endogenous opioid system in cocaine-influenced behavior. Few of these studies, however, selectively delineate a role for the mu opioid receptor (MOR) in this regard. This investigation examined if the MOR modulates cocaine-induced behavior in mice using a 17-base antisense oligodeoxynucleotide (AS ODN) directed against the MOR coding sequence 16-32. Specifically, cocaine-induced behavioral sensitization and conditioned reward were investigated. For the sensitization study, C57BL/6J mice received eight intermittent i.c.v. infusions of saline, mismatch oligodeoxynucleotide (ODN) (20 microg/4 microl) or AS ODN (20 microg/4 microl) over 20 days. Mice also received concomitant once daily i.p. injections of saline (4 ml/kg) or cocaine (15 mg/kg) for 10 days. There was a 7-day withdrawal period, after which all mice were challenged with cocaine (15 mg/kg) to test for behavioral sensitization. For the conditioned place preference (CPP) study, mice received five i.c.v. infusions of mismatch ODN or MOR AS ODN (days 1-5). An unbiased counterbalanced conditioning procedure was used where mice were conditioned with saline (4 ml/kg, i.p.) and cocaine (15 mg/kg, i.p.) on alternate days for four sessions (days 3-6). Mice were tested on day 7 for CPP. Immediately following testing, [3H]DAMGO (D-Ala2, N-Me-Phe4, Gly-ol5-enkephalin) receptor binding to brain homogenates was conducted. MOR AS attenuated cocaine-induced behavioral sensitization and conditioned reward. MOR AS ODN also reduced [3H]DAMGO binding. Collectively, these findings implicate the MOR as playing an important neuromodulatory role in the behavioral effects of cocaine in mice.


Cocaine-Related Disorders/drug therapy , Cocaine-Related Disorders/physiopathology , Oligodeoxyribonucleotides, Antisense/therapeutic use , Receptors, Opioid, mu/physiology , Reward , Analysis of Variance , Animals , Behavior, Animal , Cocaine-Related Disorders/etiology , Conditioning, Operant/drug effects , Drug Administration Routes , Drug Interactions , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Morphine/pharmacology , Narcotics/pharmacology , Protein Binding/drug effects , Radiography/methods , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/genetics , Time Factors , Tritium/pharmacokinetics
...