Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Drug Chem Toxicol ; 45(1): 250-261, 2022 Jan.
Article in English | MEDLINE | ID: mdl-31656103

ABSTRACT

Long-term morphine use for therapeutic approaches may lead to serious side effects. Several studies have suggested opioid antagonist and antioxidant therapy for reducing adverse effects of morphine. Cinnamaldehyde has a potent anti-oxidant property. In this study, separate and combined effects of cinnamaldehyde and naloxone (an opioid receptor antagonist) on behavioral changes and cerebellar histological and biochemical outcomes were investigated after long-term morphine administration. Seventy-eight rats were divided into two major morphine-treated and morphine-untreated groups. Morphine-treated group was subdivided into seven subgroups for receiving vehicle, normal saline, cinnamaldehyde (1.25, 5, and 20 mg/kg), naloxone, and cinnamaldehyde plus naloxone before morphine. Morphine-untreated group was subdivided into six subgroups and treated with vehicle, cinnamaldehyde (1.25, 5, and 20 mg/kg), naloxone, and their combination. Chemical compounds were administered for 28 consecutive days. Behavioral tests including footprint, rotarod, and beam balance tests were employed. Histopathological and biochemical alterations of cerebellum were determined. Body and cerebellum weights, stride width, time spent on the rotarod, Purkinje cell number, thickness of molecular and granular layers, superoxide dismutase (SOD), and total antioxidant capacity (TAC) decreased as a result of administrating morphine. Morphine increased beam transverse time, malondealdehyde (MDA), tumor necrosis factor-α (TNF-α), and caspase-3 levels. Histopathological changes such as cellular vacuolation and loss were also produced as a result of treatment with morphine. Cinnamaldehyde, naloxone, and their combination treatments improved all the above-mentioned alterations induced by morphine. We concluded that cinnamaldehyde produced a neuroprotective effect through anti-oxidant, anti-inflammatory, apoptotic, and probably naloxone-sensitive opioid receptor interaction mechanisms.


Subject(s)
Morphine , Naloxone , Acrolein/analogs & derivatives , Animals , Cerebellum , Morphine/toxicity , Naloxone/toxicity , Narcotic Antagonists/toxicity , Rats
2.
Cardiovasc Toxicol ; 22(1): 29-34, 2022 01.
Article in English | MEDLINE | ID: mdl-34599474

ABSTRACT

This study aimed to assess the effects of Buprenorphine (BUP) on oxidative parameters in pups born to mothers exposed to the drug during gestation and lactation. Pregnant and lactating rats received BUP, 0.5 or 0.1 mg/kg subcutaneously for 21 and 28 days, respectively. At the end of the study, the pups were anesthetized, and the hearts were dissected out to measure oxidative stress indices, including the levels of Malondialdehyde (MDA), Nitric oxide (NO), Glutathione (GSH), and the activity of Superoxide dismutase (SOD). Our findings indicated that BUP did not alter MDA, NO, GSH levels, nor SOD activity in the cardiac tissue of pups exposed to this drug during the fetal period and through breast milk. We suggest performing additional studies to determine the association between BUP and oxidative modifications in cardiac tissues of pups born to mothers under BUP therapy during gestation and lactation.


Subject(s)
Buprenorphine/toxicity , Heart/drug effects , Myocardium/metabolism , Narcotic Antagonists/toxicity , Oxidative Stress/drug effects , Prenatal Exposure Delayed Effects , Animals , Biomarkers/metabolism , Female , Gestational Age , Glutathione/metabolism , Lactation , Nitric Oxide/metabolism , Pregnancy , Rats, Wistar , Superoxide Dismutase/metabolism
3.
Exp Neurol ; 343: 113787, 2021 09.
Article in English | MEDLINE | ID: mdl-34153321

ABSTRACT

Opioid addiction can produce severe side effects including physical dependence and withdrawal. Perturbations of the gut microbiome have recently been shown to alter opioid-induced side-effects such as addiction, tolerance and dependence. In the present study, we investigated the influence of the gut microbiome on opioid withdrawal by evaluating the effects of fecal microbiota transplantation (FMT), antibiotic and probiotic treatments, and pharmacological inhibition of gut permeability in a mouse model of opioid dependence. Repeated intraperitoneal (i.p.) morphine treatment produced physical dependence that was quantified by measuring somatic signs of withdrawal (i.e. number of jumps) precipitated using the opioid antagonist naloxone. Morphine-dependent mice that received FMT from morphine-treated donor mice exhibited fewer naloxone-precipitated jumps compared to morphine-dependent counterparts receiving FMT from saline-treated donor mice. Microbial contents in the mouse cecum were altered by morphine treatment but were not differentially impacted by FMT. A broad-spectrum antibiotic cocktail (ABX) regimen reduced the bacterial load and attenuated naloxone-precipitated morphine withdrawal in morphine-dependent mice, whereas commercially available probiotic strains did not reliably alter somatic signs of opioid withdrawal. ML-7, a pharmacological inhibitor of gut permeability, reduced the morphine-induced increase in gut permeability in vivo but did not reliably alter somatic signs of naloxone-precipitated opioid withdrawal. Our results suggest that the gut microbiome impacts the development of physical dependence induced by chronic morphine administration, and that therapeutic manipulations of the gut microbiome may reduce opioid withdrawal.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Fecal Microbiota Transplantation/methods , Morphine Dependence/therapy , Naloxone/toxicity , Narcotic Antagonists/toxicity , Substance Withdrawal Syndrome/therapy , Analgesics, Opioid/administration & dosage , Animals , Combined Modality Therapy/methods , Male , Mice , Mice, Inbred C57BL , Morphine Dependence/drug therapy , Morphine Dependence/metabolism , Probiotics/administration & dosage , Substance Withdrawal Syndrome/drug therapy , Substance Withdrawal Syndrome/metabolism
4.
Cardiovasc Toxicol ; 21(3): 248-254, 2021 03.
Article in English | MEDLINE | ID: mdl-33125619

ABSTRACT

Several case reports suggest QT prolongation leading to ventricular arrhythmias with fatal outcome after intoxication with the µ-opioid receptor agonist and anti-diarrheal agent loperamide. The number of cases of loperamide misuse are growing due to its potential stimulating effects. Loperamide intoxications can be treated by naloxone. However, previous reports described a further QT prolongation associated with naloxone administration. Therefore, the aim of this study was to investigate the effects of loperamide and naloxone on the cardiac electrophysiology in a sensitive whole-heart model. Twenty-six hearts of New Zealand White rabbits were retrogradely perfused in a modified Langendorff apparatus. Monophasic action potentials were recorded by endo- and epicardially positioned catheters. Hearts were stimulated at different cycle lengths, thereby obtaining action potential duration at 90% of repolarization (APD90) and QT intervals. Programmed ventricular stimulation was used to assess ventricular vulnerability. Fourteen hearts were perfused with ascending concentrations of loperamide (0.2 µM, 0.35 µM, and 0.5 µM) after obtaining baseline data. Another 12 hearts were treated with naloxone (0.1 µM, 0.5 µM, 2 µM). Loperamide led to a significant increase in QT interval, APD90, and ventricular tachycardia (VT) episodes. In contrast, naloxone led to a decrease in QT interval and APD90. Accordingly, the number of VT episodes was unaltered. To the best of our knowledge, this is the first experimental study that investigated the effects of loperamide and naloxone in a whole-heart model. Loperamide led to a significant increase in action potential duration and QT interval. Simultaneously, the number of ventricular tachycardias was significantly increased. In contrast, naloxone led to a shortening of the action potential duration without altering arrhythmia susceptibility.


Subject(s)
Analgesics, Opioid/toxicity , Antidiarrheals/toxicity , Heart/drug effects , Loperamide/toxicity , Naloxone/toxicity , Narcotic Antagonists/toxicity , Tachycardia, Ventricular/chemically induced , Action Potentials/drug effects , Animals , Cardiac Pacing, Artificial , Cardiotoxicity , Female , Heart/physiopathology , Heart Rate/drug effects , Isolated Heart Preparation , Rabbits , Tachycardia, Ventricular/physiopathology , Time Factors
5.
Toxicol Lett ; 325: 34-42, 2020 Jun 01.
Article in English | MEDLINE | ID: mdl-32070766

ABSTRACT

Carfentanil is an ultra-potent opioid with an analgesic potency 10,000 times that of morphine but has received little scientific investigation. Three experiments were conducted to evaluate the toxicity of carfentanil and the efficacy of naloxone in adult male African green monkeys. The first experiment determined the ED50 (found to be 0.71 µg/kg) of subcutaneous carfentanil for inducing bradypnea and/or loss of posture. Experiment 2 attempted to establish the ED50 of naloxone for rapidly reversing the bradypnea/loss of posture induced by carfentanil (1.15 µg/kg). Experiment 3 evaluated the effects of carfentanil (0.575 µg/kg) alone, the safety of naloxone (71-2841 µg/kg), and the efficacy of naloxone (71-710 µg/kg) administration at two time points following carfentanil (1.15 µg/kg) on operant choice reaction time. Collectively, these experiments characterized the temporal progression of carfentanil-induced toxic signs, determined the range of naloxone doses that restored respiratory and gross behavioral function, and determined the time course and range of naloxone doses that partially or completely reversed the effects of carfentanil on operant choice reaction time performance in African green monkeys. These results have practical relevance for the selection of opioid antagonists, initial doses, and expected functional outcomes following treatment of synthetic opioid overdose in a variety of operational/emergency response contexts.


Subject(s)
Analgesics, Opioid/toxicity , Fentanyl/analogs & derivatives , Naloxone/therapeutic use , Narcotic Antagonists/therapeutic use , Analgesics, Opioid/antagonists & inhibitors , Animals , Apnea/chemically induced , Apnea/prevention & control , Behavior, Animal/drug effects , Chlorocebus aethiops , Conditioning, Operant/drug effects , Drug Overdose/drug therapy , Fentanyl/antagonists & inhibitors , Fentanyl/toxicity , Male , Naloxone/toxicity , Narcotic Antagonists/toxicity , Reaction Time/drug effects
6.
Drug Res (Stuttg) ; 70(1): 23-25, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31525817

ABSTRACT

The experiment was conducted on 10 Wistar rats, male and female, with initial body weight 270-280 g (males) and 250-260 g (females). The drug was administered using a spray cap in 10 doses of 0.1 mg at 45 min intervals. The average cumulative dose of the drug per naloxone hydrochloride was 36.6 mg/kg for males and 39.4 mg/kg for females. The animals were monitored for 2 weeks after the exposure and then euthanized by a gentle decapitation.We noticed that after each drug administration the animals showed a decrease in motor activity. During the observation period there were no animal deaths or signs of abnormalities in their general state or behavior. Beginning on day 7 a significant increase in body weight of the animals was noted in comparison with the initial data. The relative mass of the internal organs of the treated rats remained within the physiological norm.We conclude that naloxone hydrochloride after an intranasal administration at 36.6 mg/kg for males and 39.4 mg/kg for females does not cause death of animals and or have a toxic effect on their general state, does not change their protein metabolism characteristics or the appearance of the internal organs and their mass.


Subject(s)
Motor Activity/drug effects , Naloxone/toxicity , Narcotic Antagonists/toxicity , Toxicity Tests, Acute , Administration, Intranasal , Administration, Oral , Animals , Behavior Observation Techniques , Behavior, Animal/drug effects , Dose-Response Relationship, Drug , Female , Male , Models, Animal , Naloxone/administration & dosage , Narcotic Antagonists/administration & dosage , Rats , Rats, Wistar , Sex Factors
7.
Psychopharmacology (Berl) ; 237(2): 375-384, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31712968

ABSTRACT

RATIONALE: Acute naloxone-precipitated morphine withdrawal (MWD) produces a conditioned place aversion (CPA) in rats even after one or two exposures to high-dose (20 mg/kg, sc) morphine followed 24-h later by naloxone (1 mg/kg, sc). However, the somatic withdrawal reactions produced by acute naloxone-precipitated MWD in rats have not been investigated. A recently discovered fatty acid amide, N-oleoylglycine (OlGly), which has been suggested to act as a fatty acid amide hydrolase (FAAH) inhibitor and as a peroxisome proliferator-activated receptor alpha (PPARα) agonist, was previously shown to interfere with a naloxone-precipitated MWD-induced CPA in rats. OBJECTIVES: The aims of these studies were to examine the somatic withdrawal responses produced by acute naloxone-precipitated MWD and determine whether OlGly can also interfere with these responses. RESULTS: Here, we report that following two exposures to morphine (20 mg/kg, sc) each followed by naloxone (1 mg/kg, sc) 24 h later, rats display nausea-like somatic reactions of lying flattened on belly, abdominal contractions and diarrhea, and display increased mouthing movements and loss of body weight. OlGly (5 mg/kg, ip) interfered with naloxone-precipitated MWD-induced abdominal contractions, lying on belly, diarrhea and mouthing movements in male Sprague-Dawley rats, by both a cannabinoid 1 (CB1) and a PPARα mechanism of action. Since these withdrawal reactions are symptomatic of nausea, we evaluated the potential of OlGly to interfere with lithium chloride (LiCl)-induced and MWD-induced conditioned gaping in rats, a selective measure of nausea; the suppression of MWD-induced gaping reactions by OlGly was both CB1 and PPARα mediated. CONCLUSION: These results suggest that the aversive effects of acute naloxone-precipitated MWD reflect nausea, which is suppressed by OlGly.


Subject(s)
Glycine/analogs & derivatives , Morphine/adverse effects , Naloxone/toxicity , Narcotic Antagonists/toxicity , Nausea/drug therapy , Oleic Acids/therapeutic use , Substance Withdrawal Syndrome/drug therapy , Animals , Female , Glycine/pharmacology , Glycine/therapeutic use , Male , Medically Unexplained Symptoms , Morphine Dependence/drug therapy , Morphine Dependence/physiopathology , Nausea/chemically induced , Nausea/physiopathology , Oleic Acids/pharmacology , Rats , Rats, Sprague-Dawley , Shrews , Substance Withdrawal Syndrome/etiology , Substance Withdrawal Syndrome/physiopathology
8.
Psychopharmacology (Berl) ; 236(9): 2623-2633, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30993360

ABSTRACT

RATIONALE: Oleoyl glycine (OlGly), a recently discovered fatty acid amide that is structurally similar to N- acylethanolamines, which include the endocannabinoid, anandamide (AEA), as well as endogenous peroxisome proliferator-activated receptor alpha (PPARα) agonists oleoylethanolamide (OEA) and palmitoylethanolamide (PEA), has been shown to interfere with nicotine reward and dependence in mice. OBJECTIVES AND METHODS: Behavioral and molecular techniques were used to investigate the ability of OlGly to interfere with the affective properties of morphine and morphine withdrawal (MWD) in male Sprague-Dawley rats. RESULTS: Synthetic OlGly (1-30 mg/kg, intraperitoneal [ip]) produced neither a place preference nor aversion on its own; however, at doses of 1 and 5 mg/kg, ip, it blocked the aversive effects of MWD in a place aversion paradigm. This effect was reversed by the cannabinoid 1 (CB1) receptor antagonist, AM251 (1 mg/kg, ip), but not the PPARα antagonist, MK886 (1 mg/kg, ip). OlGly (5 or 30 mg/kg, ip) did not interfere with a morphine-induced place preference or reinstatement of a previously extinguished morphine-induced place preference. Ex vivo analysis of tissue (nucleus accumbens, amygdala, prefrontal cortex, and interoceptive insular cortex) collected from rats experiencing naloxone-precipitated MWD revealed that OlGly was selectively elevated in the nucleus accumbens. MWD did not modify levels of the endocannabinoids 2-AG and AEA, nor those of the PPARα ligands, OEA and PEA, in any region evaluated. CONCLUSION: Here, we show that OlGly interferes with the aversive properties of acute naloxone-precipitated morphine withdrawal in rats. These results suggest that OlGly may reduce the impact of MWD and may possess efficacy in treating opiate withdrawal.


Subject(s)
Analgesics, Opioid/adverse effects , Glycine/analogs & derivatives , Morphine/adverse effects , Naloxone/toxicity , Oleic Acids/administration & dosage , Reward , Substance Withdrawal Syndrome/drug therapy , Amygdala/drug effects , Amygdala/metabolism , Animals , Dose-Response Relationship, Drug , Glycine/administration & dosage , Glycine/metabolism , Male , Mice , Narcotic Antagonists/toxicity , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Oleic Acids/metabolism , Rats , Rats, Sprague-Dawley , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/psychology
9.
Pain ; 160(2): 307-321, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30412056

ABSTRACT

Ocular pain is a core symptom of inflammatory or traumatic disorders affecting the anterior segment. To date, the management of chronic ocular pain remains a therapeutic challenge in ophthalmology. The main endogenous opioids (enkephalins) play a key role in pain control but exhibit only transient analgesic effects due to their rapid degradation. The aim of this study was to explore the antinociceptive and anti-inflammatory effects of topical administration of PL265 (a dual enkephalinase inhibitor) on murine models of corneal pain. On healthy corneas, chronic PL265 topical administration did not alter corneal integrity nor modify corneal mechanical and chemical sensitivity. Then, on murine models of corneal pain, we showed that repeated instillations of PL265 (10 mM) significantly reduced corneal mechanical and chemical hypersensitivity. PL265-induced corneal analgesia was completely antagonized by naloxone methiodide, demonstrating that PL265 antinociceptive effects were mediated by peripheral corneal opioid receptors. Moreover, flow cytometry (quantification of CD11b+ cells) and in vivo confocal microscopy analysis revealed that instillations of PL265 significantly decreased corneal inflammation in a corneal inflammatory pain model. Chronic PL265 topical administration also decreased Iba1 and neuronal injury marker (ATF3) staining in the nucleus of primary sensory neurons of ipsilateral trigeminal ganglion. These results open a new avenue for ocular pain treatment based on the enhancement of endogenous opioid peptides' analgesic effects in tissues of the anterior segment of the eye. Dual enkephalinase inhibitor PL265 seems to be a promising topical treatment for safe and effective alleviation of ocular pain and inflammation.


Subject(s)
Cornea/pathology , Enzyme Inhibitors/administration & dosage , Inflammation/drug therapy , Pain/drug therapy , Propionates/administration & dosage , Administration, Topical , Animals , Anti-Infective Agents, Local/therapeutic use , Benzalkonium Compounds/therapeutic use , Capsaicin/toxicity , Cornea/drug effects , Corneal Injuries/chemically induced , Corneal Injuries/complications , Disease Models, Animal , Hyperalgesia/physiopathology , Inflammation/chemically induced , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Naloxone/toxicity , Narcotic Antagonists/toxicity , Pain/etiology , Pain Threshold/drug effects , Sensory System Agents/toxicity , Trigeminal Ganglion/metabolism , Trigeminal Ganglion/pathology
10.
Reprod Toxicol ; 78: 81-89, 2018 06.
Article in English | MEDLINE | ID: mdl-29635048

ABSTRACT

Buprenorphine, a mu opioid receptor partial agonist and kappa opioid receptor (KOR) antagonist, is an emerging therapeutic agent for maternal opioid dependence in pregnancy and neonatal abstinence syndrome. However, the endogenous opioid system plays a critical role in modulating neurodevelopment and perinatal buprenorphine exposure may detrimentally influence this. To identify aspects of neurodevelopment vulnerable to perinatal buprenorphine exposure, we defined KOR protein expression and its cellular associations in normal rat brain from embryonic day 16 to postnatal day 23 with double-labelling immunohistochemistry. KOR was expressed on neural stem and progenitor cells (NSPCs), choroid plexus epithelium, subpopulations of cortical neurones and oligodendrocytes, and NSPCs and subpopulations of neurones in postnatal hippocampus. These distinct patterns of KOR expression suggest several pathways vulnerable to perinatal buprenorphine exposure, including proliferation, neurogenesis and neurotransmission. We thus suggest the cautious use of buprenorphine in both mothers and infants until its impact on neurodevelopment is better defined.


Subject(s)
Analgesics, Opioid/toxicity , Brain/drug effects , Buprenorphine/toxicity , Maternal-Fetal Exchange , Narcotic Antagonists/toxicity , Receptors, Opioid, kappa/metabolism , Animals , Animals, Newborn , Brain/metabolism , Cell Proliferation/drug effects , Embryonic Development/drug effects , Female , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Neurons/drug effects , Neurons/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Pregnancy , Rats, Wistar , Receptors, Opioid, kappa/antagonists & inhibitors
11.
Toxicol Appl Pharmacol ; 329: 85-95, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28554659

ABSTRACT

Naloxegol is an opioid antagonist which has been developed for the treatment of patients with opioid induced constipation. In the nonclinical safety program naloxegol was shown to have a very benign toxicity profile. In the rat, but not the mouse, 2-year carcinogenicity study a change in tumour pattern with an increase in testicular Leydig cell tumours (LCT) was observed after dosing at high (supra-pharmacological) concentrations. To establish the basis of the increase in LCT and to assess its potential relevance to humans, studies to exclude and potentially identify mode-of-action (MoA) were performed. A genotoxic mechanism was ruled out following negative results in the Ames, mouse lymphoma, and micronucleus assays. An effect on androgen metabolism was excluded since the treatment of rats with naloxegol for 14days did not result in any induction of CYP protein levels. It was demonstrated that administration of centrally restricted opioid antagonists naloxegol or methylnaltrexone at high doses induced an increase in LH release with no clear increase in testosterone, in contrast to the centrally acting opioid antagonist naloxone, which showed marked increases in both LH and testosterone. LCT due to increased LH stimulation is common in rats but not documented in humans. Collectively, the lack of genotoxicity signal, the lack of androgen effect, the increase in LH secretion in rats, which is no considered to be relevant for LCT formation in humans, and high margins to clinical exposures, the observed increase in LCT in the rat is not expected to be clinically relevant.


Subject(s)
Blood-Brain Barrier/metabolism , Leydig Cell Tumor/chemically induced , Luteinizing Hormone/blood , Morphinans/toxicity , Narcotic Antagonists/toxicity , Polyethylene Glycols/toxicity , Testicular Neoplasms/chemically induced , Animals , Biomarkers/blood , Capillary Permeability , Dogs , Female , Humans , Leydig Cell Tumor/pathology , Male , Mice , Morphinans/metabolism , Naltrexone/analogs & derivatives , Naltrexone/metabolism , Naltrexone/toxicity , Narcotic Antagonists/metabolism , Polyethylene Glycols/metabolism , Quaternary Ammonium Compounds/metabolism , Quaternary Ammonium Compounds/toxicity , Rabbits , Rats, Sprague-Dawley , Risk Assessment , Species Specificity , Testicular Neoplasms/pathology , Testosterone/blood , Time Factors , Toxicity Tests/methods , Up-Regulation
12.
Toxicol Sci ; 155(2): 389-399, 2017 02.
Article in English | MEDLINE | ID: mdl-27803387

ABSTRACT

Respiratory depression and fatalities have been attributed to ethanol/buprenorphine (BUP) combination in drug addicts maintained with BUP/naloxone or BUP alone. The exact mechanisms of the ethanol/BUP interaction and the contribution to the toxicity of norbuprenorphine (NBUP), the main BUP metabolite with respiratory depressant properties are unknown. We investigated the sedative and plethsymographic effects resulting from the co-administration of intragastric ethanol (3 g/kg) and intravenous BUP (30 mg/kg) in Sprague-Dawley rats. We determined the whole blood pharmacokinetics of ethanol (using gas chromatography coupled to mass spectrometry), BUP and its metabolites (using liquid chromatography coupled to tandem mass spectrometry) and investigated the mechanisms of drug-drug interactions in the presence or absence of naloxone (7.5 mg/kg). Ethanol/BUP and ethanol/BUP/naloxone combinations significantly deepened sedation in comparison to BUP alone (P < .01) and BUP/naloxone (P < .05), respectively. Ethanol/BUP combination significantly increased the inspiratory time and decreased the minute volume in comparison to BUP alone (P < .01 and P < .01, respectively) and ethanol/BUP/naloxone (P < .05 and P < .01, respectively). Neither naloxone nor flumazenil reversed ethanol/BUP-induced sedation and respiratory depression. In the presence of ethanol, the area under the BUP concentration-time curve was significantly decreased (P < .05), BUP volume of distribution increased (P < .05) and the metabolic ratios of NBUP and norbuprenorphine-3-glucuronide increased (P < .01). In conclusion, the ethanol/BUP combination results in marked sedation and respiratory depression in the rat, prevented but not reversed by naloxone. Ethanol/BUP interactions are mainly pharmacokinetic resulting in increased NBUP production. Despite the non-reversal by naloxone and flumazenil of the effects attributed to the ethanol/BUP combination, protection provided by naloxone suggests an additional pharmacodynamic interaction.


Subject(s)
Analgesics, Opioid/toxicity , Buprenorphine/toxicity , Ethanol/toxicity , Narcotic Antagonists/toxicity , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacokinetics , Animals , Buprenorphine/administration & dosage , Buprenorphine/analogs & derivatives , Buprenorphine/metabolism , Buprenorphine/pharmacokinetics , Drug Interactions , Ethanol/administration & dosage , Ethanol/pharmacokinetics , Gas Chromatography-Mass Spectrometry , Male , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/pharmacokinetics , Rats , Rats, Sprague-Dawley , Respiration/drug effects , Respiratory Insufficiency/chemically induced
13.
Cent Eur J Public Health ; 24(3): 248-251, 2016 09.
Article in English | MEDLINE | ID: mdl-27743517

ABSTRACT

Licit and illicit drug use in pregnant women constitutes a long lasting and serious problem worldwide. Information on long-term effects of maternal drug use on the child is limited. Nationwide registers provide a great potential to study short and long-term consequences for children exposed to licit and illicit drugs during pregnancy. We discuss this potential, with a special emphasis on exposure to methamphetamine, heroin and prescription drugs used for opioid maintenance treatment (OMT). We also discuss the advantages of register data and of merging such data from different regions. The Czech and Scandinavian registers are largely comparable and provide great opportunities to conduct innovative research. For instance, using Czech and Scandinavian cohorts we can compare groups with similar characteristics, such as mothers in OMT and mothers addicted to other drugs while also controlling for important confounding factors such as health and socio-economic status.


Subject(s)
Analgesics, Opioid/toxicity , Heroin/toxicity , Methamphetamine/toxicity , Prenatal Exposure Delayed Effects , Substance-Related Disorders/epidemiology , Adult , Czech Republic/epidemiology , Female , Humans , Illicit Drugs , Infant, Newborn , Middle Aged , Narcotic Antagonists/toxicity , Opiate Substitution Treatment , Pregnancy , Pregnancy Outcome , Registries , Scandinavian and Nordic Countries/epidemiology
14.
Pain ; 157(10): 2285-2296, 2016 10.
Article in English | MEDLINE | ID: mdl-27306045

ABSTRACT

Individuals with Alzheimer's disease (AD) are in susceptible patient groups in which pain is an important clinical issue that is often underdiagnosed. However, it is unclear whether decreased pain complaints in patients with AD result from elevated pain tolerance or an impaired ability to communicate sensations. Here, we explored if AD-related pathology is present in key regions of the pain pathway and assessed whether nociceptive thresholds to acute noxious stimulation are altered in the double-mutant APPswe × PS1.M146V (TASTPM) transgenic mouse model of AD. TASTPM mice exhibited an age-dependant cognitive deficit at the age of 6 months, but not at 4 months, a deficit that was accompanied by amyloid plaques in the cortex, hippocampus, and thalamus. In the spinal cord, ß-amyloid (APP/Aß) immunoreactivity was observed in dorsal and ventral horn neurons, and the expression of vesicular glutamate transporter 2 (VGLUT2) was significantly reduced, while the expression of the inhibitory peptides enkephalins was increased in TASTPM dorsal horn, consistent with an increased inhibitory tone. TASTPM mice displayed reduced sensitivity to acute noxious heat, which was reversed by naloxone, an opioid antagonist. This study suggests that increased inhibition and decreased excitation in the spinal cord may be responsible for the reduced thermal sensitivity associated with AD-related pathology.


Subject(s)
Alzheimer Disease/complications , Analgesics, Opioid/metabolism , Cognition Disorders/etiology , Sensory Thresholds/physiology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/drug effects , Brain/metabolism , Carrageenan/toxicity , Disease Models, Animal , Locomotion/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Naloxone/toxicity , Narcotic Antagonists/toxicity , Nerve Tissue Proteins/metabolism , Pain/chemically induced , Pain Measurement , Presenilin-1/genetics , Presenilin-1/metabolism , Recognition, Psychology/physiology , Sensory Thresholds/drug effects
15.
J Neurosci ; 36(11): 3391-7, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26985045

ABSTRACT

Mindfulness meditation, a cognitive practice premised on sustaining nonjudgmental awareness of arising sensory events, reliably attenuates pain. Mindfulness meditation activates multiple brain regions that contain a high expression of opioid receptors. However, it is unknown whether mindfulness-meditation-based analgesia is mediated by endogenous opioids. The present double-blind, randomized study examined behavioral pain responses in healthy human volunteers during mindfulness meditation and a nonmanipulation control condition in response to noxious heat and intravenous administration of the opioid antagonist naloxone (0.15 mg/kg bolus + 0.1 mg/kg/h infusion) or saline placebo. Meditation during saline infusion significantly reduced pain intensity and unpleasantness ratings when compared to the control + saline group. However, naloxone infusion failed to reverse meditation-induced analgesia. There were no significant differences in pain intensity or pain unpleasantness reductions between the meditation + naloxone and the meditation + saline groups. Furthermore, mindfulness meditation during naloxone produced significantly greater reductions in pain intensity and unpleasantness than the control groups. These findings demonstrate that mindfulness meditation does not rely on endogenous opioidergic mechanisms to reduce pain. SIGNIFICANCE STATEMENT: Endogenous opioids have been repeatedly shown to be involved in the cognitive inhibition of pain. Mindfulness meditation, a practice premised on directing nonjudgmental attention to arising sensory events, reduces pain by engaging mechanisms supporting the cognitive control of pain. However, it remains unknown if mindfulness-meditation-based analgesia is mediated by opioids, an important consideration for using meditation to treat chronic pain. To address this question, the present study examined pain reports during meditation in response to noxious heat and administration of the opioid antagonist naloxone and placebo saline. The results demonstrate that meditation-based pain relief does not require endogenous opioids. Therefore, the treatment of chronic pain may be more effective with meditation due to a lack of cross-tolerance with opiate-based medications.


Subject(s)
Analgesics, Opioid/metabolism , Meditation , Pain/metabolism , Pain/rehabilitation , Treatment Outcome , Adult , Analysis of Variance , Double-Blind Method , Female , Healthy Volunteers , Hot Temperature/adverse effects , Humans , Male , Meditation/psychology , Naloxone/toxicity , Narcotic Antagonists/toxicity , Pain/chemically induced , Pain Measurement , Psychophysics , Young Adult
16.
Can J Physiol Pharmacol ; 93(11): 935-44, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26440527

ABSTRACT

Long-term use of opioids for pain management results in rapid development of tolerance and dependence leading to severe withdrawal symptoms. We have previously demonstrated that endothelin-A (ETA) receptor antagonists potentiate opioid analgesia and eliminate analgesic tolerance. This study was designed to investigate the involvement of central ET mechanisms in opioid withdrawal. The effect of intracerebroventricular administration of ETA receptor antagonist BQ123 on morphine and oxycodone withdrawal was determined in male Swiss Webster mice. Opioid tolerance was induced and withdrawal was precipitated by the opioid antagonist naloxone. Expression of ETA and ETB receptors, nerve growth factor (NGF), and vascular endothelial growth factor was determined in the brain using Western blotting. BQ123 pretreatment reversed hypothermia and weight loss during withdrawal. BQ123 also reduced wet shakes, rearing behavior, and jumping behavior. No changes in expression of vascular endothelial growth factor, ETA receptors, and ETB receptors were observed during withdrawal. NGF expression was unaffected in morphine withdrawal but significantly decreased during oxycodone withdrawal. A decrease in NGF expression in oxycodone- but not in morphine-treated mice could be due to mechanistic differences in oxycodone and morphine. It is concluded that ETA receptor antagonists attenuate opioid-induced withdrawal symptoms.


Subject(s)
Endothelin A Receptor Antagonists/therapeutic use , Naloxone/toxicity , Opioid-Related Disorders/drug therapy , Peptides, Cyclic/therapeutic use , Substance Withdrawal Syndrome/drug therapy , Animals , Endothelin A Receptor Antagonists/pharmacology , Male , Mice , Narcotic Antagonists/toxicity , Opioid-Related Disorders/pathology , Opioid-Related Disorders/physiopathology , Pain Measurement/drug effects , Pain Measurement/methods , Peptides, Cyclic/pharmacology , Substance Withdrawal Syndrome/pathology , Substance Withdrawal Syndrome/physiopathology
17.
Int J Pharm ; 477(1-2): 631-42, 2014 Dec 30.
Article in English | MEDLINE | ID: mdl-25445974

ABSTRACT

Naltrexone hydrochloride (NTX) is an innovative drug used in ophthalmology for treatment of ocular surface diseases such as impaired corneal wound healing and severe dry eye. Poor chemical stability has been a major limitation for development of NTX in solution form. The aim of this study was to develop and characterise NTX in situ ocular films for enhanced chemical stability and improved ocular tolerability. The films were prepared from different amorphous polymers and characterised for physicochemical compatibility, moisture-sorption, surface pH, mechanical properties, sterilisability, surface morphology, mucoadhesion, in vitro release, conjunctival irritation and accelerated stability at 40°C/75% relative humidity for 3 months. Glycerin (GLY)-plasticised films exhibited significantly better mechanical properties, compared with polyethylene glycol (PEG) 400 and triethylcitrate (TEC)-plasticised formulations. Superior mucoadhesion was recorded for F7 and F9 plasticised with GLY and PEG 400, respectively. The stability of NTX was significantly enhanced more than 18-times, compared with the solution form. Combination of carboxymethylcellulose sodium (CMC) and sodium alginate (ALG) in a film formulation demonstrated minimal % moisture sorption, good mechanical properties, in vitro release, excellent chemical stability and minimal conjunctival irritation lending them as promising ocular formulations.


Subject(s)
Drug Carriers/chemistry , Naltrexone/administration & dosage , Narcotic Antagonists/administration & dosage , Receptors, Opioid/metabolism , Adhesiveness , Administration, Ophthalmic , Animals , Calorimetry, Differential Scanning , Chickens , Chorioallantoic Membrane/drug effects , Conjunctivitis/drug therapy , Disease Models, Animal , Drug Carriers/toxicity , Drug Liberation , Drug Stability , Drug Storage , Gels , In Vitro Techniques , Microscopy, Electron, Scanning , Naltrexone/therapeutic use , Naltrexone/toxicity , Narcotic Antagonists/therapeutic use , Narcotic Antagonists/toxicity , Spectroscopy, Fourier Transform Infrared , Stress, Mechanical , Surface Properties , Tensile Strength
18.
Toxicol Lett ; 228(2): 75-84, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24769261

ABSTRACT

Respiratory depression has been attributed to buprenorphine (BUP) misuse or combination with benzodiazepines. BUP/naloxone (NLX) has been marketed as maintenance treatment, aiming at preventing opiate addicts from self-injecting crushed pills. However, to date, BUP/NLX benefits in comparison to BUP alone remain debated. We investigated the plethysmography effects of BUP/NLX in comparison to BUP/solvent administered by intravenous route in naive and BUP-tolerant Sprague-Dawley rats, and in combination with diazepam (DZP) or its solvent. In naive rats, BUP/NLX in comparison to BUP significantly increased respiratory frequency (f, P<0.05) without altering minute volume (VE). In combination to DZP, BUP/NLX significantly increased expiratory time (P<0.01) and decreased f (P<0.01), tidal volume (VT, P<0.001), and VE (P<0.001) while BUP only decreased VT (P<0.5). In BUP-tolerant rats, no significant differences in respiratory effects were observed between BUP/NLX and BUP. In contrast, in combination to DZP, BUP/NLX did not significantly alter the plethysmography parameters, while BUP increased inspiratory time (P<0.001) and decreased f (P<0.01) and VE (P<0.001). In conclusion, differences in respiratory effects between BUP/NLX and BUP are only significant in combination with DZP, with increased depression in naive rats but reduced depression in BUP-tolerant rats. However, BUP/NLX benefits in humans remain to be determined.


Subject(s)
Analgesics, Opioid/toxicity , Buprenorphine/toxicity , Diazepam/toxicity , Hypnotics and Sedatives/toxicity , Naloxone/toxicity , Narcotic Antagonists/toxicity , Respiratory Insufficiency/chemically induced , Analysis of Variance , Animals , Drug Interactions , Drug Tolerance , Male , Plethysmography, Whole Body , Rats , Rats, Sprague-Dawley
19.
Curr HIV Res ; 12(6): 424-34, 2014.
Article in English | MEDLINE | ID: mdl-25760046

ABSTRACT

Few preclinical studies have compared the relative therapeutic efficacy of medications used to treat opiate addiction in relation to neuroAIDS. Here we compare the ability of methadone and buprenorphine, and the prototypic opiate morphine, to potentiate the neurotoxic and proinflammatory ([Ca²âº]i, ROS, H2O2, chemokines) effects of HIV-1 Tat in neuronal and/or mixed-glial co-cultures. Repeated observations of neurons during 48 h exposure to combinations of Tat, equimolar concentrations (500 nM) of morphine, methadone, or buprenorphine exacerbated neurotoxicity significantly above levels seen with Tat alone. Buprenorphine alone displayed marked neurotoxicity at 500 nM, prompting additional studies of its neurotoxic effects at 5 nM and 50 nM concentrations ± Tat. In combination with Tat, buprenorphine displayed paradoxical, concentration-dependent, neurotoxic and neuroprotective actions. Buprenorphine neurotoxicity coincided with marked elevations in [Ca²âº]i, but not increases in glial ROS or chemokine release. Tat by itself elevated the production of CCL5/RANTES, CCL4/MIP-1ß, and CCL2/MCP-1. Methadone and buprenorphine alone had no effect, but methadone interacted with Tat to further increase production of CCL5/RANTES. In combination with Tat, all drugs significantly increased glial [Ca²âº]i, but ROS was only significantly increased by co-exposure with morphine. Taken together, the increases in glial [Ca²âº]i, ROS, and neuroinflammatory chemokines were not especially accurate predictors of neurotoxicity. Despite similarities, opiates displayed differences in their neurotoxic and neuroinflammatory interactions with Tat. Buprenorphine, in particular, was partially neuroprotective at a low concentration, which may result from its unique pharmacological profile at multiple opioid receptors. Overall, the results reveal differences among addiction medications that may impact neuroAIDS.


Subject(s)
Calcium/metabolism , Chemokines/metabolism , Narcotic Antagonists/toxicity , Neuroglia/drug effects , Neurons/physiology , Reactive Oxygen Species/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Buprenorphine/metabolism , Buprenorphine/toxicity , Cell Survival/drug effects , Cells, Cultured , Humans , Methadone/metabolism , Methadone/toxicity , Morphine/metabolism , Morphine/toxicity , Narcotic Antagonists/metabolism , Narcotics/metabolism , Narcotics/toxicity
20.
J Reprod Dev ; 59(5): 479-84, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23877505

ABSTRACT

Puberty in mammals is timed by an increase in gonadotropin-releasing hormone (GnRH) secretion. Previous studies have shown involvement of the two neuropeptides, kisspeptin and neurokinin B (NKB), in controlling puberty onset. Little is known about the role of the other key neuropeptide, dynorphin, in controlling puberty onset, although these three neuropeptides colocalize in the arcuate kisspeptin neurons. The arcuate kisspeptin neuron, which is also referred to as the KNDy neuron, has recently been considered to play a role as an intrinsic source of the GnRH pulse generator. The present study aimed to determine if attenuation of inhibitory dynorphin-kappa-opioid receptor (KOR) signaling triggers the initiation of puberty in normal developing female rats. The present study also determined if stimulatory NKB-neurokinin 3 receptor (NK3R) signaling advances puberty onset. Female Wistar-Imamichi rats were weaned and intraperitoneally implanted with osmotic minipumps filled with nor-binaltorphimine (nor-BNI), a KOR antagonist, or senktide, a NK3R agonist, at 20 days of age. Fourteen days of intraperitoneal infusion of nor-BNI or senktide advanced puberty onset, manifested as vaginal opening and the first vaginal estrus in female rats. Frequent blood sampling showed that nor-BNI significantly increased luteinizing hormone (LH) pulse frequency at 29 days of age compared with vehicle-treated controls. Senktide tended to increase this frequency, but its effect was not statistically significant. The present results suggest that the inhibitory input of dynorphin-KOR signaling plays a role in the prepubertal restraint of GnRH/LH secretion in normal developing female rats and that attenuation of dynorphin-KOR signaling and increase in NKB-NK3R signaling trigger the onset of puberty in female rats.


Subject(s)
Dynorphins/metabolism , Luteinizing Hormone/metabolism , Neurokinin B/metabolism , Ovary/metabolism , Receptors, Neurokinin-3/metabolism , Receptors, Opioid, kappa/metabolism , Sexual Maturation , Animals , Drug Implants , Dynorphins/antagonists & inhibitors , Female , Luteinizing Hormone/blood , Naltrexone/administration & dosage , Naltrexone/analogs & derivatives , Naltrexone/toxicity , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/toxicity , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurokinin B/agonists , Neurons/drug effects , Neurons/metabolism , Ovary/drug effects , Ovary/growth & development , Peptide Fragments/administration & dosage , Peptide Fragments/toxicity , Puberty, Precocious/blood , Puberty, Precocious/chemically induced , Puberty, Precocious/metabolism , Random Allocation , Rats , Rats, Wistar , Receptors, Neurokinin-3/agonists , Receptors, Opioid, kappa/antagonists & inhibitors , Sexual Maturation/drug effects , Signal Transduction/drug effects , Substance P/administration & dosage , Substance P/analogs & derivatives , Substance P/toxicity , Weaning
SELECTION OF CITATIONS
SEARCH DETAIL