Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 822
Filter
1.
Acta Neurobiol Exp (Wars) ; 82(2): 217-225, 2022.
Article in English | MEDLINE | ID: mdl-35833821

ABSTRACT

The lateral hypothalamus (LH) sends neural pathways to structures involved on predator­related defensive behaviours, escape and antinociception. The aim of this study was to investigate the role played by µ-opioid receptors located on LH neurons in defensive behaviour and unconditioned fear­induced antinociception elicited by electric stimulation of LH. To achieve the goals, the µ1-opioid receptor selective antagonist naloxonazine was administered at different concentrations in the LH, and the defensive behaviour and fear­induced antinociception elicited by electrical stimulation of LH were evaluated. The electrical stimulation of LH caused escape behaviour followed by defensive antinociception. Microinjections of naloxonazine in a concentration of 5.0 µg/0.2 µL in the LH decreased the aversive stimulus­induced escape behaviour thresholds, but diminished defensive antinociception. These findings suggest that µ-opioid receptors of LH can be critical to panic attack­related symptoms and facilitate the unconditioned fear­induced antinociception produced by LH neurons activation.


Subject(s)
Behavior, Animal , Hypothalamic Area, Lateral , Panic Disorder , Receptors, Opioid, mu , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Bicuculline/pharmacology , Fear/physiology , Hypothalamic Area, Lateral/drug effects , Hypothalamic Area, Lateral/metabolism , Naloxone/analogs & derivatives , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Nociception , Panic/physiology , Panic Disorder/metabolism , Panic Disorder/psychology , Rats , Rats, Wistar , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/metabolism
2.
Eur J Clin Pharmacol ; 77(12): 1901-1908, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34327552

ABSTRACT

PURPOSE: Remifentanil has been shown to increase the bioavailability of nasally administered naloxone. The aim of this study was to explore the nature of this observation. METHODS: We analysed samples from three pharmacokinetic studies to determine the serum concentrations of naloxone-3-glucuronide (N3G), the main metabolite of naloxone, with or without exposure to remifentanil. To enable direct comparison of the three studies, the data are presented as metabolic ratios (ratio of metabolite to mother substance, N3G/naloxone) and dose-corrected values of the area under the curve and maximum concentration (Cmax). RESULTS: Under remifentanil exposure, the time to maximum concentration (Tmax) for N3G was significantly higher for intranasal administration of 71 min compared to intramuscular administration of 40 min. The dose-corrected Cmax of N3G after intranasal administration of naloxone under remifentanil exposure was significantly lower (4.5 ng/mL) than in subjects not exposed to remifentanil (7.8-8.4 ng/mL). The metabolic ratios after intranasal administration rose quickly after 30-90 min and were 2-3 times higher at 360 min compared to intravenous and intramuscular administration. Remifentanil exposure resulted in a much slower increase of the N3G/naloxone ratio after intranasal administration compared to intranasal administration with the absence of remifentanil. After remifentanil infusion was discontinued, this effect gradually diminished. From 240 min there was no significant difference between the ratios observed after intranasal naloxone administration. CONCLUSION: Remifentanil increases the bioavailability of naloxone after nasal administration by reducing the pre-systemic metabolism of the swallowed part of the nasal dose.


Subject(s)
Analgesics, Opioid/pharmacology , Naloxone/analogs & derivatives , Naloxone/pharmacokinetics , Narcotic Antagonists/pharmacokinetics , Remifentanil/pharmacology , Administration, Intranasal , Area Under Curve , Dose-Response Relationship, Drug , Healthy Volunteers , Humans , Injections, Intramuscular , Metabolic Clearance Rate , Naloxone/administration & dosage , Naloxone/blood , Narcotic Antagonists/administration & dosage
3.
Int J Mol Sci ; 22(14)2021 Jul 09.
Article in English | MEDLINE | ID: mdl-34299013

ABSTRACT

Mucosal CD4+ T lymphocytes display a potent opioid-mediated analgesic activity in interleukin (IL)-10 knockout mouse model of inflammatory bowel diseases (IBD). Considering that endogenous opioids may also exhibit anti-inflammatory activities in the periphery, we examined the consequences of a peripheral opioid receptor blockade by naloxone-methiodide, a general opioid receptor antagonist unable to cross the blood-brain barrier, on the development of piroxicam-accelerated colitis in IL-10-deficient (IL-10-/-) mice. Here, we show that IL-10-deficient mice treated with piroxicam exhibited significant alterations of the intestinal barrier function, including permeability, inflammation-related bioactive lipid mediators, and mucosal CD4+ T lymphocyte subsets. Opioid receptor antagonization in the periphery had virtually no effect on colitis severity but significantly worsened epithelial cell apoptosis and intestinal permeability. Thus, although the endogenous opioid tone is not sufficient to reduce the severity of colitis significantly, it substantially contributes to the protection of the physical integrity of the epithelial barrier.


Subject(s)
Colitis/metabolism , Interleukin-10/genetics , Intestinal Mucosa/drug effects , Naloxone/analogs & derivatives , Narcotic Antagonists/administration & dosage , Piroxicam/pharmacology , Receptors, Opioid/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , CD4-Positive T-Lymphocytes/drug effects , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Cytokines/genetics , Cytokines/metabolism , Epithelial Cells/drug effects , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Interleukin-10/metabolism , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Naloxone/pharmacology , Permeability/drug effects , Quaternary Ammonium Compounds/pharmacology , Severity of Illness Index
4.
Peptides ; 141: 170543, 2021 07.
Article in English | MEDLINE | ID: mdl-33794284

ABSTRACT

Endomorphin analogs containing unnatural amino acids have demonstrated potent analgesic effects in our previous studies. In the present study, the differences in antinociception and the mechanisms thereof for analogs 1-3 administered intracerebroventricularly and intrathecally were explored. All analogs at different routes of administration produced potent analgesia compared to the parent peptide endomorphin-1. Multiple antagonists and antibodies were used to explore the mechanisms of action of these analogs, and it was inferred that analogs 1-3 stimulated the µ opioid receptor to induce antinociception. Moreover, the antibody data suggested that analog 2 may induce the release of immunoreactive [Leu5]-enkephaline and [Met5]-enkephaline to produce a secondary component of antinociception at the spinal level and analog 3 may stimulate the the release of immunoreactive [Met5]-enkephaline at the spinal level. Finally, analogs 2 and 3 produced no acute tolerance in the spinal cord. We hypothesize that the unique characteristics of the endomorphin analogs result from their capacities to stimulate the release of endogenous antinociceptive substances.


Subject(s)
Analgesics/pharmacology , Enkephalins/metabolism , Oligopeptides/chemistry , Receptors, Opioid, mu/metabolism , Spinal Cord/drug effects , Analgesics/administration & dosage , Animals , Chronic Pain/drug therapy , Dose-Response Relationship, Drug , Dynorphins/metabolism , Enkephalin, Methionine/metabolism , Injections, Intraventricular , Mice , Naloxone/analogs & derivatives , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Oligopeptides/pharmacology , Receptors, Opioid, mu/antagonists & inhibitors
5.
Biochim Biophys Acta Gen Subj ; 1865(3): 129838, 2021 03.
Article in English | MEDLINE | ID: mdl-33373630

ABSTRACT

BACKGROUND: G protein-coupled receptors (GPCRs) comprise a family of membrane proteins that can be activated by a variety of external factors. The µ-opioid receptor (MOR), a class A GPCR, is the main target of morphine. Recently, enhanced sampling molecular dynamics simulations of a constitutively active mutant of MOR in its apo form allowed us to capture the novel intermediate states of activation, as well as the active state. This prompted us to apply the same techniques to wild type MOR in complex with ligands, in order to explore their contributions to the receptor conformational changes in the activation process. METHODS: MOR was modeled in complex with agonists (morphine, BU72), a partial agonist (naloxone benzoylhydrazone) and an antagonist (naloxone). Replica exchange with solute tempering (REST2) molecular dynamics simulations were carried out for all systems. Trajectory frames were clustered, and the activation state of each cluster was assessed by two different methods. RESULTS: Cluster sizes and activation indices show that while agonists stabilized structures in a higher activation state, the antagonist behaved oppositely. Morphine tends to drive the receptor towards increasing R165-T279 distances, while naloxone tends to increase the NPxxYA motif conformational change. CONCLUSIONS: Despite not observing a full transition between inactive and active states, an important conformational change of transmembrane helix 5 was observed and associated with a ligand-driven step of the process. GENERAL SIGNIFICANCE: The activation process of GPCRs is widely studied but still not fully understood. Here we carried out a step forward in the direction of gaining more details of this process.


Subject(s)
Amino Acids/chemistry , Apoproteins/chemistry , Morphine/chemistry , Phosphatidylcholines/chemistry , Receptors, Opioid, mu/chemistry , Amino Acids/metabolism , Apoproteins/metabolism , Binding Sites , Humans , Ligands , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Morphinans/chemistry , Morphinans/metabolism , Morphine/metabolism , Naloxone/analogs & derivatives , Naloxone/chemistry , Naloxone/metabolism , Phosphatidylcholines/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Pyrroles/chemistry , Pyrroles/metabolism , Receptors, Opioid, mu/metabolism , Solutions , Water/chemistry , Water/metabolism
7.
Sci Rep ; 10(1): 833, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31964994

ABSTRACT

While it is known that opioid receptors (ORs) are densely expressed in both the brain and periphery, it is widely accepted that hypoxic effects of opioids result solely from their direct action in the CNS. To examine the role of peripheral ORs in triggering brain hypoxia, we used oxygen sensors in freely moving rats to examine how naloxone-HCl and naloxone-methiodide, the latter which is commonly believed to be peripherally restricted, affect brain oxygen responses induced by intravenous heroin at low, human-relevant doses. Similar to naloxone-HCl, naloxone-methiodide at a relatively low dose (2 mg/kg) fully blocked heroin-induced decreases in brain oxygen levels. As measured by mass spectrometry, naloxone-methiodide was found to be ~40-fold less permeable than naloxone-HCl across the blood-brain barrier, thus acting as a selective blocker of peripheral ORs. Despite this selectivity, a low but detectable amount of naloxone was found in brain tissue after naloxone-methiodide administration, potentially influencing our results. Therefore, we examined the effects of naloxone-methiodide at a very low dose (0.2 mg/kg; at which naloxone was undetectable in brain tissue) and found that this drug still powerfully attenuates heroin-induced brain oxygen responses. These data demonstrate the role of peripheral ORs in triggering heroin-induced respiratory depression and subsequent brain hypoxia.


Subject(s)
Heroin/adverse effects , Hypoxia, Brain/etiology , Receptors, Opioid/physiology , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Hypoxia, Brain/drug therapy , Naloxone/administration & dosage , Naloxone/analogs & derivatives , Naloxone/metabolism , Naloxone/pharmacology , Oxygen/metabolism , Quaternary Ammonium Compounds/administration & dosage , Quaternary Ammonium Compounds/metabolism , Quaternary Ammonium Compounds/pharmacology , Rats , Receptors, Opioid/metabolism
9.
Neuropsychobiology ; 78(4): 218-228, 2019.
Article in English | MEDLINE | ID: mdl-31514182

ABSTRACT

BACKGROUND: Gamma-aminobutyric acid (GABA)ergic and opioid systems play a crucial role in the neural modulation of innate fear organised by the inferior colliculus (IC). In addition, the IC is rich in GABAergic fibres and opioid neurons, which are also connected to other mesencephalic structures, such as the superior colliculus and the substantia nigra. However, the contribution of distinct opioid receptors (ORs) in the IC during the elaboration and expression of innate fear and panic-like responses is unclear. The purpose of the present work was to investigate a possible integrated action exerted by ORs and the GABAA receptor-mediated system in the IC on panic-like responses. METHODS: The effect of the blockade of either µ1- or κ-ORs in the IC was evaluated in the unconditioned fear-induced responses elicited by GABAA antagonism with bicuculline. Microinjections of naloxonazine, a µ1-OR antagonist, or nor-binaltorphimine (nor-BNI), a κ-OR antagonist, were made into the IC, followed by intramesencephalic administration of the GABAA-receptor antagonist bicuculline. The defensive behaviours elicited by the treatments in the IC were quantitatively analysed, recording the number of escapes expressed as running (crossing), jumps, and rotations, over a 30-min period in a circular arena. The exploratory behaviour of rearing was also recorded. RESULTS: GABAA-receptor blockade with bicuculline in the IC increased defensive behaviours. However, pretreatment of the IC with higher doses (5 µg) of naloxonazine or nor-BNI followed by bicuculline resulted in a significant decrease in unconditioned fear-induced responses. CONCLUSIONS: These findings suggest a role played by µ1- and κ-OR-containing connexions and GABAA receptor-mediated neurotransmission on the organisation of panic attack-related responses elaborated by the IC neurons.


Subject(s)
Behavior, Animal/drug effects , Inferior Colliculi/drug effects , Mesencephalon/drug effects , Narcotic Antagonists/pharmacology , Panic/drug effects , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, mu/antagonists & inhibitors , Animals , Bicuculline/pharmacology , Exploratory Behavior/drug effects , GABA-A Receptor Antagonists/pharmacology , Male , Naloxone/analogs & derivatives , Naloxone/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Neurons/drug effects , Rats , Rats, Wistar
10.
Sci Signal ; 12(575)2019 04 02.
Article in English | MEDLINE | ID: mdl-30940767

ABSTRACT

Pain and inflammation are inherently linked responses to injury, infection, or chronic diseases. Given that acute inflammation in humans or mice enhances the analgesic properties of opioids, there is much interest in determining the inflammatory transducers that prime opioid receptor signaling in primary afferent nociceptors. Here, we found that activation of the transient receptor potential vanilloid type 1 (TRPV1) channel stimulated a mitogen-activated protein kinase (MAPK) signaling pathway that was accompanied by the shuttling of the scaffold protein ß-arrestin2 to the nucleus. The nuclear translocation of ß-arrestin2 in turn prevented its recruitment to the µ-opioid receptor (MOR), the subsequent internalization of agonist-bound MOR, and the suppression of MOR activity that occurs upon receptor desensitization. Using the complete Freund's adjuvant (CFA) inflammatory pain model to examine the role of TRPV1 in regulating endogenous opioid analgesia in mice, we found that naloxone methiodide (Nal-M), a peripherally restricted, nonselective, and competitive opioid receptor antagonist, slowed the recovery from CFA-induced hypersensitivity in wild-type, but not TRPV1-deficient, mice. Furthermore, we showed that inflammation prolonged morphine-induced antinociception in a mouse model of opioid receptor desensitization, a process that depended on TRPV1. Together, our data reveal a TRPV1-mediated signaling pathway that serves as an endogenous pain-resolution mechanism by promoting the nuclear translocation of ß-arrestin2 to minimize MOR desensitization. This previously uncharacterized mechanism may underlie the peripheral opioid control of inflammatory pain. Dysregulation of the TRPV1-ß-arrestin2 axis may thus contribute to the transition from acute to chronic pain.


Subject(s)
Acute Pain/metabolism , Analgesics, Opioid/pharmacology , Chronic Pain/metabolism , Naloxone/analogs & derivatives , Narcotic Antagonists/pharmacology , Signal Transduction/drug effects , TRPV Cation Channels/metabolism , Acute Pain/chemically induced , Acute Pain/drug therapy , Acute Pain/genetics , Analgesia , Animals , Chronic Pain/chemically induced , Chronic Pain/drug therapy , Chronic Pain/genetics , Disease Models, Animal , Freund's Adjuvant/adverse effects , Freund's Adjuvant/pharmacology , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Mice , Mice, Knockout , Naloxone/pharmacology , Quaternary Ammonium Compounds/pharmacology , Signal Transduction/genetics , TRPV Cation Channels/genetics , beta-Arrestin 2/genetics , beta-Arrestin 2/metabolism
11.
J Am Heart Assoc ; 8(2): e010152, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30636504

ABSTRACT

Background Morphine administration is a strong predictor of delayed onset of action of orally administered ticagrelor in patients with ST-segment-elevation myocardial infarction, likely because of impaired gastrointestinal motility. The aim of this study was to evaluate whether the peripheral opioid antagonist methylnaltrexone could improve pharmacodynamics and pharmacokinetics of orally administered ticagrelor in patients with ST-segment-elevation myocardial infarction receiving morphine. Methods and Results The MOVEMENT (Methylnaltrexone to Improve Platelet Inhibition of Ticagrelor in Morphine-Treated Patients With ST-Segment Elevation Myocardial Infarction) trial was a multicenter, prospective, randomized, controlled trial in patients with ST-segment-elevation myocardial infarction treated with morphine and ticagrelor. Upon arrival to the catheterization laboratory, patients were randomized to a blinded intravenous injection of either methylnaltrexone (8 or 12 mg according to weight) or 0.9% sodium chloride. The proportion of patients with high on-treatment platelet reactivity and plasma concentrations of ticagrelor and AR -C124910XX were assessed at baseline (arrival in the catheterization laboratory) and 1 and 2 hours later. A total of 82 patients received either methylnaltrexone (n=43) or placebo (n=39). Median (interquartile range) time from ticagrelor administration to randomization was 41 (31-50) versus 45.5 (37-60) minutes ( P=0.16). Intravenous methylnaltrexone administration did not significantly affect prevalence of high on-treatment platelet reactivity at 2 hours after inclusion, the primary end point, when compared with placebo (54% versus 51%, P=0.84). Plasma concentrations of ticagrelor and its active metabolite, the prespecified secondary end points, did not differ significantly between the groups over time. There was no significant difference in patient self-estimated pain between the groups. Conclusions Methylnaltrexone did not significantly improve platelet reactivity or plasma concentrations of orally administered ticagrelor in patients with ST-segment-elevation myocardial infarction receiving morphine. Clinical Trial Registration URL : http://www.clinicaltrials.gov . Unique identifier: NCT 02942550.


Subject(s)
Blood Platelets/metabolism , Morphine/therapeutic use , Naloxone/analogs & derivatives , ST Elevation Myocardial Infarction/drug therapy , Ticagrelor/pharmacokinetics , Administration, Oral , Aged , Analgesics, Opioid/therapeutic use , Blood Platelets/drug effects , Cardiac Catheterization , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , Follow-Up Studies , Humans , Injections, Intravenous , Male , Middle Aged , Naloxone/administration & dosage , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Function Tests , Prospective Studies , Quaternary Ammonium Compounds/administration & dosage , ST Elevation Myocardial Infarction/diagnosis , ST Elevation Myocardial Infarction/physiopathology , Single-Blind Method , Ticagrelor/administration & dosage , Treatment Outcome
12.
J Psychopharmacol ; 33(5): 577-588, 2019 05.
Article in English | MEDLINE | ID: mdl-30663473

ABSTRACT

BACKGROUND: The endogenous opioid peptide system has been implicated in the neural modulation of fear and anxiety organised by the dorsal midbrain. Furthermore, previous results indicate a fundamental role played by inferior colliculus (IC) opioid mechanisms during the expression of defensive behaviours, but the involvement of the IC µ1-opioid receptor in the modulation of anxiety- and panic attack-related behaviours remains unclear. Using a prey-versus-snake confrontation paradigm, we sought to investigate the effects of µ1-opioid receptor blockade in the IC on the defensive behaviour displayed by rats in a dangerous situation. METHODS: Specific pathogen-free Wistar rats were treated with microinjection of the selective µ1-opioid receptor antagonist naloxonazine into the IC at different concentrations (1.0, 3.0 and 5.0 µg/0.2 µL) and then confronted with rattlesnakes ( Crotalus durissus terrificus). The defensive behavioural repertoire, such as defensive attention, flat back approach (FBA), startle, defensive immobility, escape or active avoidance, displayed by rats either during the confrontations with wild snakes or during re-exposure to the experimental context without the predator was analysed. RESULTS: The blockade of µ1-opioid receptors in the IC decreased the expression of both anxiety-related behaviours (defensive attention, FBA) and panic attack-related responses (startle, defensive immobility and escape) during the confrontation with rattlesnakes. A significant decrease in defensive attention was also recorded during re-exposure of the prey to the experimental apparatus context without the predator. CONCLUSION: Taken together, these results suggest that a decrease in µ1-opioid receptor signalling activity within the IC modulates anxiety- and panic attack-related behaviours in dangerous environments.


Subject(s)
Anxiety/prevention & control , Behavior, Animal/drug effects , Fear , Inferior Colliculi/drug effects , Narcotic Antagonists/pharmacology , Panic Disorder/prevention & control , Receptors, Opioid, mu/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Crotalus , Disease Models, Animal , Food Chain , Naloxone/analogs & derivatives , Naloxone/pharmacology , Rats , Rats, Wistar
13.
Addict Biol ; 24(5): 874-885, 2019 09.
Article in English | MEDLINE | ID: mdl-29949228

ABSTRACT

Kratom, derived from the plant Mitragyna speciosa, is receiving increased attention as an alternative to traditional opiates and as a replacement therapy for opiate dependence. Mitragynine (MG) and 7-hydroxymitragynine (7-HMG) are major psychoactive constituents of kratom. While MG and 7-HMG share behavioral and analgesic properties with morphine, their reinforcing effects have not been examined to date. 7-HMG, but not MG, substituted for morphine self-administration in a dose-dependent manner in the rat self-administration paradigm. Following the substitution procedure, re-assessment of morphine self-administration revealed a significant increase following 7-HMG and a significant decrease following MG substitution. In a separate cohort, 7-HMG, but not MG, engendered and maintained intravenous self-administration in a dose-dependent manner. The effects of pretreatment with nalxonaxine (NLXZ), a µ1 opiate receptor antagonist, and naltrindole (NTI), a δ opiate receptor antagonist, on 7-HMG and morphine self-administration were also examined. Both NLXZ and NTI reduced 7-HMG self-administration, whereas only NLXZ decreased morphine intake. The present results are the first to demonstrate that 7-HMG is readily self-administered, and the reinforcing effects of 7-HMG are mediated in part by µ and δ opiate receptors. In addition, prior exposure to 7-HMG increased subsequent morphine intake whereas prior exposure to MG decreased morphine intake. The present findings indicate that MG does not have abuse potential and reduces morphine intake, desired characteristics of candidate pharmacotherapies for opiate addiction and withdrawal, whereas 7-HMG should be considered a kratom constituent with high abuse potential that may also increase the intake of other opiates.


Subject(s)
Analgesics, Opioid/administration & dosage , Behavior, Animal/drug effects , Morphine/administration & dosage , Narcotic Antagonists/pharmacology , Secologanin Tryptamine Alkaloids/pharmacology , Analgesics, Opioid/pharmacology , Animals , Mitragyna , Naloxone/analogs & derivatives , Naloxone/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Opioid-Related Disorders/drug therapy , Rats , Receptors, Opioid, delta , Receptors, Opioid, mu , Self Administration
14.
Pain ; 160(3): 619-631, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30422869

ABSTRACT

Bone cancer metastasis is extremely painful and decreases the quality of life of the affected patients. Available pharmacological treatments are not able to sufficiently ameliorate the pain, and as patients with cancer are living longer, new treatments for pain management are needed. Decitabine (5-aza-2'-deoxycytidine), a DNA methyltransferases inhibitor, has analgesic properties in preclinical models of postsurgical and soft-tissue oral cancer pain by inducing an upregulation of endogenous opioids. In this study, we report that daily treatment with decitabine (2 µg/g, intraperitoneally) attenuated nociceptive behavior in the 4T1-luc2 mouse model of bone cancer pain. We hypothesized that the analgesic mechanism of decitabine involved activation of the endogenous opioid system through demethylation and reexpression of the transcriptionally silenced endothelin B receptor gene, Ednrb. Indeed, Ednrb was hypermethylated and transcriptionally silenced in the mouse model of bone cancer pain. We demonstrated that expression of Ednrb in the cancer cells lead to release of ß-endorphin in the cell supernatant, which reduced the number of responsive dorsal root ganglia neurons in an opioid-dependent manner. Our study supports a role of demethylating drugs, such as decitabine, as unique pharmacological agents targeting the pain in the cancer microenvironment.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Bone Neoplasms/complications , Cancer Pain/drug therapy , Cancer Pain/etiology , Decitabine/therapeutic use , Animals , Bone Density , Bone Neoplasms/diagnostic imaging , Bone Neoplasms/pathology , Cancer Pain/diagnostic imaging , Cell Line, Tumor , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Endothelin-1/metabolism , Female , Ganglia, Spinal/cytology , Locomotion/drug effects , Mice , Mice, Inbred BALB C , Naloxone/analogs & derivatives , Naloxone/therapeutic use , Neurons/drug effects , Quaternary Ammonium Compounds/therapeutic use , Receptor, Endothelin B/genetics , Receptor, Endothelin B/metabolism , Weight-Bearing/physiology , beta-Endorphin/metabolism
16.
Sci Rep ; 8(1): 12170, 2018 08 15.
Article in English | MEDLINE | ID: mdl-30111876

ABSTRACT

Inhibition of regeneration and induction of tissue fibrosis are classic outcomes of tissue repair in adult mammals. Here, using a newly developed model of regeneration in adult mammals i.e. regeneration after massive resection of an inguinal fat pad, we demonstrate that both endogenous and exogenous opioids prevent tissue regeneration in adults, by inhibiting the early production of reactive oxygen species (ROS) that generally occurs after lesion and is required for regeneration. These effects can be overcome and regeneration induced by the use of an opioid antagonist. The results obtained in both our new model and the gold standard adult zebrafish demonstrate that this mechanism can be considered as a general paradigm in vertebrates. This work clearly demonstrates that ROS is required for tissue regeneration in adult mammals and shows the deleterious effect of opioids on tissue regeneration through the control of this ROS production. It thus raises questions about opioid-based analgesia in perioperative care.


Subject(s)
Analgesics, Opioid/pharmacology , Regeneration/drug effects , Adipose Tissue/pathology , Analgesics, Opioid/metabolism , Animal Fins , Animals , Female , Fibrosis/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Myocytes, Cardiac/pathology , Naloxone/analogs & derivatives , Naloxone/pharmacology , Quaternary Ammonium Compounds/pharmacology , Reactive Oxygen Species/metabolism , Regeneration/physiology , Tramadol/pharmacology , Zebrafish
17.
Eur J Pharmacol ; 833: 275-282, 2018 Aug 15.
Article in English | MEDLINE | ID: mdl-29886241

ABSTRACT

Fentanyl is an opioid commonly prescribed for cancer pain. Using melanoma-bearing mice, we investigated whether peripheral action would contribute to fentanyl analgesia in cancer pain. Intravenous injection of fentanyl inhibited mechanical nociception in healthy mice, which was markedly inhibited by the opioid antagonist naloxone, but not naloxone methiodide, a peripherally acting opioid antagonist. Melanoma-bearing mice showed mechanical allodynia and spontaneous licking, a pain-related behavior, which were suppressed by intravenous and local injections of fentanyl. Both naloxone and naloxone methiodide inhibited the analgesic effect of intravenous fentanyl to the same degree. Electrophysiological analysis showed that melanoma growth increased the spontaneous and mechanical stimuli-evoked activity of the tibial nerve, which were inhibited by intravenous fentanyl. There was a greater expression of µ- opioid receptors in skin with a melanoma mass than in the contralateral normal skin. In addition, we found µ-opioid receptors in cultured melanoma cells. There was no difference between the number of µ-opioid receptors in the dorsal root ganglia and spinal cord of the melanoma-bearing and contralateral skin side. These results suggest that the analgesic effect of systemic fentanyl is produced via central and peripheral µ- opioid receptors in cancer pain, and cancer cells are a key site of peripheral action.


Subject(s)
Analgesics, Opioid/pharmacology , Cancer Pain/drug therapy , Fentanyl/pharmacology , Hyperalgesia/drug therapy , Receptors, Opioid, mu/metabolism , Action Potentials/drug effects , Analgesics, Opioid/antagonists & inhibitors , Analgesics, Opioid/therapeutic use , Animals , Cancer Pain/etiology , Cell Line, Tumor , Disease Models, Animal , Fentanyl/antagonists & inhibitors , Fentanyl/therapeutic use , Humans , Hyperalgesia/etiology , Injections, Intramuscular , Injections, Intravenous , Male , Melanoma/complications , Mice , Mice, Inbred C57BL , Naloxone/analogs & derivatives , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Quaternary Ammonium Compounds/pharmacology , Receptors, Opioid , Skin/pathology , Skin Neoplasms/complications , Spinal Cord/drug effects , Spinal Cord/metabolism , Tibial Nerve/drug effects , Tibial Nerve/metabolism , Xenograft Model Antitumor Assays
18.
ACS Chem Neurosci ; 9(3): 563-567, 2018 03 21.
Article in English | MEDLINE | ID: mdl-29154536

ABSTRACT

The photolabile analogue of the broad-spectrum opioid antagonist naloxone, 3- O-(4,5-dimethoxy-2-nitrophenyl)carboxymethyl naloxone (also referred to as "caged naloxone", 3- O-(α-carboxy-6-nitroveratryl)naloxone, CNV-NLX), has been found to be a valuable biochemical probe. While the synthesis of CNV-NLX is simple, its characterization is complicated by the fact that it is produced as a mixture of α R,5 R,9 R,13 S,14 S and α S,5 R,9 R,13 S,14 S diastereomers. Using long-range and heteronuclear NMR correlations, the 1H NMR and 13C NMR resonances of both diastereomers have been fully assigned, confirming the structures. Monitoring of solutions of CNV-NLX in saline buffer, in methanol, and in DMSO has shown CNV-NLX to be stable for over a week under fluorescent laboratory lights at room temperature. Exposure of such solutions to λ 365 nm from a hand-held UV lamp led to the formation of naloxone and CNV-related breakdown products.


Subject(s)
Light , Naloxone/analogs & derivatives , Naloxone/chemistry , Narcotic Antagonists/chemistry , Analgesics, Opioid/chemistry , Methanol/chemistry , Ultraviolet Rays
19.
Eur Neuropsychopharmacol ; 27(12): 1298-1307, 2017 12.
Article in English | MEDLINE | ID: mdl-29102248

ABSTRACT

The endogenous neuropeptide nociceptin (N/OFQ), which mediates its actions via the nociceptin receptor (NOP), is implicated in multiple behavioural and physiological functions. This study examined the effects of the NOP agonists N/OFQ and the synthetic agonist Ro 64-6198, the antagonists NNN and NalBzoH, as well as deletion of the Pronociceptin gene on emotional memory in mice. The animals were tested in the passive avoidance (PA) task, dependent on hippocampal and amygdala functions. N/OFQ injected intraventricularly (i.c.v.) prior to training produced a biphasic effect on PA retention; facilitation at a low dose and impairment at higher doses. Ro 64-6198 also displayed a biphasic effect with memory facilitation at lower doses and impairment at a high dose. None of the agonists influenced PA training latencies. NNN did not significantly modulate retention in the PA task but antagonized the inhibitory effects of N/OFQ. NalBzoH facilitated memory retention in a dose-dependent manner and blocked the impairing effects of N/OFQ. However, neither NNN nor NalBzoH blocked the memory-impairing effects of Ro 64-6198. Finally, the Pnoc knockout mice exhibited enhanced PA retention latencies compared to the wild type mice. The biphasic effect of the natural ligand and Ro 64-6198 and the failure of the antagonists to block the action of Ro 64-6198 indicate complexity in ligand-receptor interaction. These results indicate that brain nociceptin and its NOP has a subtle role in regulation of mechanisms of relevance for treatment of disorders with processing disturbances of aversive events e.g. Alzheimer's disease, anxiety, depression and PTSD.


Subject(s)
Avoidance Learning/physiology , Opioid Peptides/metabolism , Receptors, Opioid/deficiency , Animals , Association Learning/drug effects , Avoidance Learning/drug effects , Dose-Response Relationship, Drug , Imidazoles/pharmacology , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Naloxone/analogs & derivatives , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Opioid Peptides/genetics , Opioid Peptides/pharmacology , Peptide Fragments/pharmacology , Receptors, Opioid/agonists , Receptors, Opioid/genetics , Retention, Psychology/drug effects , Spiro Compounds/pharmacology , Nociceptin Receptor , Nociceptin
20.
Psychopharmacology (Berl) ; 234(20): 3009-3025, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28856406

ABSTRACT

RATIONALE: Gamma-aminobutyric acid (GABA)ergic neurons of the substantia nigra pars reticulata (SNpr) are connected to the deep layers of the superior colliculus (dlSC). The dlSC, in turn, connect with the SNpr through opioid projections. Nociceptin/orphanin FQ peptide (N/OFQ) is a natural ligand of a Gi protein-coupled nociceptin receptor (ORL1; NOP) that is also found in the SNpr. Our hypothesis is that tectonigral opioid pathways and intranigral orphanin-mediated mechanisms modulate GABAergic nigrotectal connections. OBJECTIVES: Therefore, the aim of this work was to study the role of opioid and NOP receptors in the SNpr during the modulation of defence reactions organised by the dlSC. METHODS: The SNpr was pretreated with either opioid or NOP receptor agonists and antagonists, followed by dlSC treatment with bicuculline. RESULTS: Blockade of GABAA receptors in the dlSC elicited fear-related defensive behaviour. Pretreatment of the SNpr with naloxone benzoylhydrazone (NalBzoH), a µ-, δ-, and κ1-opioid receptor antagonist as well as a NOP receptor antagonist, decreased the aversive effect of bicuculline treatment on the dlSC. Either µ-opioid receptor activation or blockade by SNpr microinjection of endomorphin-1 (EM-1) and CTOP promoted pro-aversive and anti-aversive actions, respectively, that modulated the defensive responses elicited by bicuculline injection into the dlSC. Pretreatment of the SNpr with the selective NOP receptor antagonist JTC801 decreased the aversive effect of bicuculline, and microinjections of the selective NOP receptor agonist NNC 63-0532 promoted the opposite effect. CONCLUSIONS: These results demonstrate that opioid pathways and orphanin-mediated mechanisms have a critical role in modulating the activity of nigrotectal GABAergic pathways during the organisation of defensive behaviours.


Subject(s)
Aminoquinolines/administration & dosage , Benzamides/administration & dosage , Fear/drug effects , Oligopeptides/administration & dosage , Pars Reticulata/drug effects , Receptors, Opioid , Somatostatin/analogs & derivatives , Analgesics, Opioid/administration & dosage , Animals , Bicuculline/administration & dosage , Dose-Response Relationship, Drug , Fear/physiology , Male , Naloxone/administration & dosage , Naloxone/analogs & derivatives , Opioid Peptides/administration & dosage , Pars Reticulata/physiology , Rats , Rats, Wistar , Receptors, Opioid/physiology , Somatostatin/administration & dosage , Superior Colliculi/drug effects , Superior Colliculi/physiology , gamma-Aminobutyric Acid/administration & dosage , Nociceptin Receptor , Nociceptin
SELECTION OF CITATIONS
SEARCH DETAIL
...