Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.360
Filter
1.
Sci Rep ; 14(1): 16978, 2024 07 23.
Article in English | MEDLINE | ID: mdl-39043899

ABSTRACT

Methadone is a synthetic long-acting opioid that is increasingly used in the replacement therapy of opioid-addicted patients, including pregnant women. However, methadone therapy in this population poses challenges, as it induces cognitive and behavioral impairments in infants exposed to this opioid during prenatal development. In animal models, prenatal methadone exposure results in detrimental consequences to the central nervous system, such as: (i) increased neuronal apoptosis; (ii) disruption of oligodendrocyte maturation and increased apoptosis and (iii) increased microglia and astrocyte activation. However, it remains unclear whether these deleterious effects result from a direct effect of methadone on brain cells. Therefore, our goal was to uncover the impact of methadone on single brain cell types in vitro. Primary cultures of rat neurons, oligodendrocytes, microglia, and astrocytes were treated for three days with 10 µM methadone to emulate a chronic administration. Apoptotic neurons were identified by cleaved caspase-3 detection, and synaptic density was assessed by the juxtaposition of presynaptic and postsynaptic markers. Apoptosis of oligodendrocyte precursors was determined by cleaved caspase-3 detection. Oligodendrocyte myelination was assessed by immunofluorescence, while microglia and astrocyte proinflammatory activation were assessed by both immunofluorescence and RT-qPCR. Methadone treatment increased neuronal apoptosis and reduced synaptic density. Furthermore, it led to increased oligodendrocyte apoptosis and a reduction in the myelinating capacity of these cells, and promoted the proinflammatory activation of microglia and astrocytes. We showed that methadone, the most widely used drug in opioid replacement therapy for pregnant women with opioid addiction, directly impairs brain cells in vitro, highlighting the need for developing alternative therapies to address opioid addiction in this population.


Subject(s)
Apoptosis , Astrocytes , Methadone , Microglia , Neurons , Oligodendroglia , Methadone/pharmacology , Animals , Rats , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Apoptosis/drug effects , Astrocytes/drug effects , Astrocytes/metabolism , Neurons/drug effects , Neurons/metabolism , Microglia/drug effects , Microglia/metabolism , Cells, Cultured , Female , Central Nervous System/drug effects , Central Nervous System/metabolism , Pregnancy , Analgesics, Opioid/pharmacology , Rats, Sprague-Dawley
2.
Sci Rep ; 14(1): 14350, 2024 06 21.
Article in English | MEDLINE | ID: mdl-38906975

ABSTRACT

Cardiac ischemic preconditioning (Pre) reduces cardiac ischemia-reperfusion injury (IRI) by stimulating opioid receptors. Chronic use of opioids can alter the signaling pathways. We investigated the effects of chronic methadone use on IRI and Pre. The experiments were performed on isolated hearts of male Wistar rats in four groups: IRI, Methadone + IRI (M-IRI), Pre + IRI (Pre-IRI), Methadone + Pre + IRI (M-Pre-IRI). The infarct size (IS) in the Pre-IRI group was smaller than the IRI group (26.8% vs. 47.8%, P < 0.05). In the M-IRI and M-Pre-IRI groups, the infarct size was similar to the IRI group. Akt (Ak strain transforming) phosphorylation in the Pre-IRI, M-IRI, and M-Pre-IRI groups was significantly higher than in the IRI group (0.56 ± 0.15, 0.63 ± 0.20, and 0.93 ± 0.18 vs 0.28 ± 0.17 respectively). STAT3 (signal transducer and activator of transcription 3) phosphorylation in the Pre-IRI and M-Pre-IRI groups (1.38 ± 0.14 and 1.46 ± 0.33) was significantly higher than the IRI and M-IRI groups (0.99 ± 0.1 and 0.98 ± 0.2). Thus, chronic use of methadone not only has no protective effect against IRI but also destroys the protective effects of ischemic preconditioning. This may be due to the hyperactivation of Akt and changes in signaling pathways.


Subject(s)
Ischemic Preconditioning, Myocardial , Methadone , Myocardial Reperfusion Injury , Proto-Oncogene Proteins c-akt , STAT3 Transcription Factor , Animals , Male , Rats , Ischemic Preconditioning, Myocardial/methods , Methadone/pharmacology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/pathology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats, Wistar , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Signal Transduction/drug effects , STAT3 Transcription Factor/metabolism
3.
Vet Anaesth Analg ; 51(3): 253-265, 2024.
Article in English | MEDLINE | ID: mdl-38580536

ABSTRACT

OBJECTIVE: To evaluate cardiovascular effects of oral tasipimidine on propofol-isoflurane anaesthesia with or without methadone and dexmedetomidine at equianaesthetic levels. STUDY DESIGN: Prospective, placebo-controlled, blinded, experimental trial. ANIMALS: A group of seven adult Beagle dogs weighing (mean ± standard deviation) 12.4 ± 2.6 kg and a mean age of 20.6 ± 1 months. METHODS: The dogs underwent four treatments 60 minutes before induction of anaesthesia with propofol. PP: placebo orally and placebo (NaCl 0.9%) intravenously (IV); TP: tasipimidine 30 µg kg-1 orally and placebo IV; TMP: tasipimidine 30 µg kg-1 orally and methadone 0.2 mg kg-1 IV; and TMPD: tasipimidine 30 µg kg-1 orally with methadone 0.2 mg kg-1 and dexmedetomidine 1 µg kg-1 IV followed by 1 µg kg-1 hour-1. Isoflurane in oxygen was maintained for 120 minutes at 1.2 individual minimum alveolar concentration preventing motor movement. Cardiac output (CO), tissue blood flow (tbf), tissue oxygen saturation (stO2) and relative haemoglobin content were determined. Arterial and mixed venous blood gases, arterial and pulmonary artery pressures and heart rate (HR) were measured at baseline; 60 minutes after oral premedication; 5 minutes after IV premedication; 15, 30, 60, 90 and 120 minutes after propofol injection; and 30 minutes after switching the vaporiser off. Data were analysed by two-way anova for repeated measures; p < 0.05. RESULTS: Tasipimidine induced a significant 20-30% reduction in HR and CO with decreases in MAP (10-15%), tbf (40%) and stO2 (43%). Blood pressure and oxygenation variables were mainly influenced by propofol-isoflurane-oxygen anaesthesia, preceded by short-lived alterations related to IV methadone and dexmedetomidine. CONCLUSIONS AND CLINICAL RELEVANCE: Tasipimidine induced mild to moderate cardiovascular depression. It can be incorporated into a common anaesthetic protocol without detrimental effects in healthy dogs, when anaesthetics are administered to effect and cardiorespiratory function is monitored.


Subject(s)
Dexmedetomidine , Isoflurane , Methadone , Propofol , Pyrazoles , Animals , Dogs , Dexmedetomidine/administration & dosage , Dexmedetomidine/pharmacology , Propofol/administration & dosage , Propofol/pharmacology , Methadone/administration & dosage , Methadone/pharmacology , Female , Isoflurane/administration & dosage , Isoflurane/pharmacology , Heart Rate/drug effects , Male , Blood Pressure/drug effects , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/administration & dosage , Quinolizines/pharmacology , Quinolizines/administration & dosage , Anesthetics, Intravenous/administration & dosage , Anesthetics, Intravenous/pharmacology , Anesthetics, Inhalation/administration & dosage , Anesthetics, Inhalation/pharmacology , Premedication/veterinary
4.
Int J Mol Sci ; 25(4)2024 Feb 10.
Article in English | MEDLINE | ID: mdl-38396814

ABSTRACT

Methadone is an effective and long-lasting analgesic drug that is also used in medication-assisted treatment for people with opioid use disorders. Although there is evidence that methadone activates µ-opioid and Toll-like-4 receptors (TLR-4s), its effects on distinct immune cells, including mast cells (MCs), are not well characterized. MCs express µ-opioid and Toll-like receptors (TLRs) and constitute an important cell lineage involved in allergy and effective innate immunity responses. In the present study, murine bone-marrow-derived mast cells (BMMCs) were treated with methadone to evaluate cell viability by flow cytometry, cell morphology with immunofluorescence and scanning electron microscopy, reactive oxygen species (ROS) production, and intracellular calcium concentration ([Ca2+]i) increase. We found that exposure of BMMCs to 0.5 mM or 1 mM methadone rapidly induced cell death by forming extracellular DNA traps (ETosis). Methadone-induced cell death depended on ROS formation and [Ca2+]i. Using pharmacological approaches and TLR4-defective BMMC cultures, we found that µ-opioid receptors were necessary for both methadone-induced ROS production and intracellular calcium increase. Remarkably, TLR4 receptors were also involved in methadone-induced ROS production as it did not occur in BMMCs obtained from TLR4-deficient mice. Finally, confocal microscopy images showed a significant co-localization of µ-opioid and TLR4 receptors that increased after methadone treatment. Our results suggest that methadone produces MCETosis by a mechanism requiring a novel crosstalk pathway between µ-opioid and TLR4 receptors.


Subject(s)
Analgesics, Opioid , Extracellular Traps , Humans , Animals , Mice , Analgesics, Opioid/pharmacology , Toll-Like Receptor 4/metabolism , Methadone/pharmacology , Mast Cells/metabolism , Reactive Oxygen Species/metabolism , Bone Marrow/metabolism , Calcium/metabolism , Extracellular Traps/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptors/metabolism
5.
Vet J ; 303: 106065, 2024 02.
Article in English | MEDLINE | ID: mdl-38228282

ABSTRACT

In this prospective, randomised, blinded clinical study, we compared the sedative, antinociceptive and cardiorespiratory effects of intranasal (IN) dexmedetomidine at 5 µg/kg (diluted with 0.03 mL/kg NaCl 0.9%, DEX) with or without methadone (0.3 mg/kg; DEXMET), through a mucosal atomization device to one nostril in twenty healthy client-owned dogs. At 5-min intervals over 45 min, sedation score, onset, cardiopulmonary variables, mechanical nociceptive thresholds (MNTs) were assessed, also ease of administration, adverse effects, and response to IV catheterization. Statistical analysis employed t-test, the Mann-Whitney U, repeated measures ANOVA and Chi-square tests as appropriate (P < 0.05). Higher sedation ocurred in DEXMET (7 [5-10]) compared to DEX (5 [2-7]) from 15 to 30 min (P < 0.01, median [interquartile range]). Heart rate was lower in DEXMET (P < 0.01; 65% reduction vs. 41% in DEX, P = 0.001). The MNTs were higher in DEXMET than DEX from 15 to 45 min (P < 0.01), peaking at T30 (17.1 ± 3.8, DEXMET and 8.5 ± 5.4 N, DEX). No differences were observed in mean arterial blood pressure and respiratory rate. Intranasal administration was considered easy for 8 dogs per group. Reverse sneezing (8 dogs; P < 0.001), sialorrhea and retching (4 and 2 dogs, respectively) occurred in DEXMET. Response to catheterisation was lower in DEXMET than DEX (P = 0.039; 2 and 7 dogs, respectively). In conclusion, intranasal methadone (0.3 mg/kg) increased the sedative and antinociceptive effects produced by dexmedetomidine (5 µg/kg) in healthy dogs and resulted in lower heart rate.


Subject(s)
Analgesics , Dexmedetomidine , Hypnotics and Sedatives , Methadone , Animals , Dogs , Administration, Intranasal/veterinary , Analgesics/pharmacology , Dexmedetomidine/pharmacology , Drug Combinations , Hypnotics and Sedatives/pharmacology , Methadone/pharmacology , Prospective Studies , Drug Synergism
6.
J Vet Pharmacol Ther ; 47(2): 73-79, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37930056

ABSTRACT

The purpose of this study was to assess antinociception and correlation of antinociception and hypothermic effects after intravenous opioids in dogs. Nine healthy male Beagles were enrolled in the study. They were acclimated to a thermal nociceptive device, then received three IV treatments (saline, butorphanol 0.4 mg/kg and methadone 0.5 mg/kg) in a randomized complete block design. Rectal temperature and thermal withdrawals were assessed prior to and 0.5-6 h after drug administration. One dog was excluded due to lack of withdrawal to thermal stimuli. Rectal temperatures were not significantly different between treatments at time 0, but significantly decreased from 0.5 to 5 h for both opioids compared to saline. Withdrawals were significantly decreased, compared to saline, from 0.5 to 4 h for butorphanol and 0.5-5 h for methadone. A significant (p = .0005) and moderate (R2 = .43) correlation between antinociception and hypothermia occurred. Based on these data, intravenous butorphanol (0.4 mg/kg) and methadone (0.5 mg/kg) provided 4 and 5 h of antinociception, respectively. Opioid hypothermia can serve as an easy, noninvasive and humane manner for preclinical assessment of opioid antinociception in dogs prior to evaluation in clinical trials. This is a major refinement in animal welfare for assessing novel opioids, opioid doses and dose intervals in dogs.


Subject(s)
Analgesics, Opioid , Hypothermia , Dogs , Male , Animals , Analgesics, Opioid/pharmacology , Butorphanol/pharmacology , Hypothermia/chemically induced , Hypothermia/prevention & control , Hypothermia/veterinary , Methadone/pharmacology , Administration, Intravenous/veterinary
7.
Mol Psychiatry ; 29(3): 624-632, 2024 03.
Article in English | MEDLINE | ID: mdl-38145984

ABSTRACT

(R,S)-methadone ((R,S)-MTD) is a µ-opioid receptor (MOR) agonist comprised of (R)-MTD and (S)-MTD enantiomers. (S)-MTD is being developed as an antidepressant and is considered an N-methyl-D-aspartate receptor (NMDAR) antagonist. We compared the pharmacology of (R)-MTD and (S)-MTD and found they bind to MORs, but not NMDARs, and induce full analgesia. Unlike (R)-MTD, (S)-MTD was a weak reinforcer that failed to affect extracellular dopamine or induce locomotor stimulation. Furthermore, (S)-MTD antagonized motor and dopamine releasing effects of (R)-MTD. (S)-MTD acted as a partial agonist at MOR, with complete loss of efficacy at the MOR-galanin Gal1 receptor (Gal1R) heteromer, a key mediator of the dopaminergic effects of opioids. In sum, we report novel and unique pharmacodynamic properties of (S)-MTD that are relevant to its potential mechanism of action and therapeutic use. One-sentence summary: (S)-MTD, like (R)-MTD, binds to and activates MORs in vitro, but (S)-MTD antagonizes the MOR-Gal1R heteromer, decreasing its abuse liability.


Subject(s)
Analgesics, Opioid , Methadone , Receptors, Opioid, mu , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/drug effects , Animals , Methadone/pharmacology , Male , Analgesics, Opioid/pharmacology , Humans , Mice , Dopamine/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Ligands , Stereoisomerism
8.
Can Vet J ; 64(11): 1058-1065, 2023 11.
Article in English | MEDLINE | ID: mdl-37915778

ABSTRACT

Opioid analgesics are routinely used during the perioperative period, to provide analgesia and reduce anesthetics doses required to maintain a surgical plane of anesthesia in companion animals. Acting on receptors in the brain, spinal cord, and peripheral nervous system, opioids provide reliable and consistent analgesia; however, they are not without adverse effects. Methadone, a mu agonist opioid analgesic, was recently licensed for veterinary use in Canada. In addition to its action on opioid receptors, methadone contributes to analgesia through other pathways, including inhibition of N-methyl-D-aspartate (NMDA) receptors. It has physiologic effects similar to other mu opioid agents, but fewer adverse gastrointestinal effects. This review discusses methadone's mechanism of action, pharmacologic characteristics, and clinical effects in dogs and cats. Current recommendations for using methadone in companion animals are also provided.


Le point sur l'anesthésie ­ Intégration de la méthadone dans les protocoles d'anesthésie et d'analgésie des animaux de compagnie : une revue descriptive. Les analgésiques opioïdes sont couramment utilisés pendant la période peropératoire, afin de fournir une analgésie et réduire les doses d'anesthésiques nécessaires pour maintenir un plan d'anesthésie chirurgical chez les animaux de compagnie. Agissant sur les récepteurs du cerveau, de la moelle épinière et du système nerveux périphérique, les opioïdes fournissent une analgésie fiable et constante; cependant, ils ne sont pas sans effets indésirables. La méthadone, un analgésique opioïde agoniste mu, a récemment été homologuée pour un usage vétérinaire au Canada. En plus de son action sur les récepteurs opioïdes, la méthadone contribue à l'analgésie par d'autres voies, notamment l'inhibition des récepteurs N-méthyl-D-aspartate (NMDA). Elle a des effets physiologiques similaires à ceux d'autres agents opioïdes mu, mais moins d'effets gastrointestinaux indésirables. Cette revue discute du mécanisme d'action de la méthadone, de ses caractéristiques pharmacologiques et de ses effets cliniques chez les chiens et les chats. Les recommandations actuelles concernant l'utilisation de la méthadone chez les animaux de compagnie sont également fournies.(Traduit par Dr Serge Messier).


Subject(s)
Anesthesia , Cat Diseases , Dog Diseases , Animals , Cats , Dogs , Methadone/therapeutic use , Methadone/pharmacology , Pets , Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , Anesthesia/veterinary , Pain/drug therapy , Pain/veterinary
9.
J Neurosci Res ; 101(11): 1737-1756, 2023 11.
Article in English | MEDLINE | ID: mdl-37551165

ABSTRACT

The circadian clock is one of the most important homeostatic systems regulating the majority of physiological functions. Its proper development contributes significantly to the maintenance of health in adulthood. Methadone is recommended for the treatment of opioid use disorders during pregnancy, increasing the number of children prenatally exposed to long-acting opioids. Although early-life opioid exposure has been studied for a number of behavioral and physiological changes observed later in life, information on the relationship between the effects of methadone exposure and circadian system development is lacking. Using a rat model, we investigated the effects of prenatal and early postnatal methadone administration on the maturation of the circadian clockwork in the suprachiasmatic nucleus (SCN) and liver, the rhythm of aralkylamine N-acetyltransferase (AA-NAT) activity in the pineal gland, and gene expression in the livers of 20-day-old rats. Our data show that repeated administration of methadone to pregnant and lactating mothers has significant effect on rhythmic gene expression in the SCN and livers and on the rhythm of AA-NAT in the offspring. Similar to previous studies with morphine, the rhythm amplitudes of the clock genes in the SCN and liver were unchanged or enhanced. However, six of seven specific genes in the liver showed significant downregulation of their expression, compared to the controls in at least one experimental group. Importantly, the amplitude of the AA-NAT rhythm was significantly reduced in all methadone-treated groups. As there is a strong correlation with melatonin levels, this result could be of importance for clinical practice.


Subject(s)
Melatonin , Pineal Gland , Pregnancy , Female , Rats , Animals , Methadone/metabolism , Methadone/pharmacology , Lactation , Circadian Rhythm/physiology , Pineal Gland/metabolism , Melatonin/pharmacology , Suprachiasmatic Nucleus/physiology
10.
Neuropharmacology ; 239: 109683, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37543137

ABSTRACT

Buprenorphine (BUP) and methadone (MTD) are used for medication-assisted treatment (MAT) in opioid use disorder. Although both medications show improved maternal and neonatal outcomes compared with illicit opioid use during pregnancy, BUP has exhibited more favorable outcomes to newborns than MTD. The underlying cellular and molecular mechanisms for the difference between BUP and MTD are largely unknown. Here, we examined the growth and neuronal activity in human cortical organoids (hCOs) exposed to BUP or MTD. We found that the growth of hCOs was significantly restricted in the MTD-treated but not in the BUP-treated hCOs and BUP attenuated the growth-restriction effect of MTD in hCOs. Furthermore, a κ-receptor agonist restricted while an antagonist alleviated the growth-restriction effect of MTD in hCOs. Since BUP is not only a µ-agonist but a κ-antagonist, the prevention of this growth-restriction by BUP is likely due to its κ-receptor-antagonism. In addition, using multielectrode array (MEA) technique, we discovered that both BUP and MTD inhibited neuronal activity in hCOs but BUP showed suppressive effects only at higher concentrations. Furthermore, κ-receptor antagonist nBNI did not prevent the MTD-induced suppression of neuronal activity in hCOs but the NMDA-antagonism of MTD (that BUP lacks) plays a role in the inhibition of neuronal activity. We conclude that, although both MTD and BUP are µ-opioid agonists, a) the additional κ-receptor antagonism of BUP mitigates the MTD-induced growth restriction during neurodevelopment and b) the lack of NMDA antagonism of BUP (in contrast to MTD) induces much less suppressive effect on neural network communications.


Subject(s)
Buprenorphine , Opioid-Related Disorders , Infant, Newborn , Humans , Buprenorphine/pharmacology , Buprenorphine/therapeutic use , Methadone/pharmacology , Methadone/therapeutic use , Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , N-Methylaspartate , Opioid-Related Disorders/drug therapy , Receptors, Opioid, kappa , Organoids , Brain , Narcotic Antagonists/pharmacology , Narcotic Antagonists/therapeutic use
11.
Addict Biol ; 28(9): e13320, 2023 09.
Article in English | MEDLINE | ID: mdl-37644895

ABSTRACT

Fatal poisonings where both methadone and quetiapine are detected post-mortem occurs frequently in legal autopsy cases. It is unclear whether quetiapine increases the risk of fatal methadone poisoning or if it is merely detected due to widespread use. We hypothesized that methadone and quetiapine would have additive toxic effects on respiratory rate, blood pressure, and the QTc-interval. To investigate this hypothesis, we used telemetry implants for measurements of respiratory rate, haemodynamic variables, the velocity of blood pressure changes, temperature, and movement in conscious, freely moving male Wistar rats aged 12-13 weeks. The combined effects of three accumulative i.p. doses of methadone (2.5, 10, 15 mg/kg) and quetiapine (3, 10, 30 mg/kg) were compared to rats treated with the same doses of each drug alone, and a vehicle-treated group in a randomized investigator blinded study. No additive effects of quetiapine and methadone on respiratory rate, haemodynamic variables, or movement were observed. However, body temperature was significantly lower by approximately 1.5°C on average in the group treated with both methadone and quetiapine (15 + 30 mg/kg) compared to the other groups. This indicates a synergistic effect of quetiapine and methadone on thermoregulation, which may increase the risk of fatal poisoning. We suggest studying this finding further in human settings.


Subject(s)
Methadone , Respiratory Rate , Humans , Rats , Animals , Male , Quetiapine Fumarate/pharmacology , Methadone/pharmacology , Temperature , Rats, Wistar , Hemodynamics
12.
Transl Psychiatry ; 13(1): 151, 2023 05 06.
Article in English | MEDLINE | ID: mdl-37147277

ABSTRACT

Opioid use disorder (OUD) among pregnant women has become an epidemic in the United States. Pharmacological interventions for maternal OUD most commonly involve methadone, a synthetic opioid analgesic that attenuates withdrawal symptoms and behaviors linked with drug addiction. However, evidence of methadone's ability to readily accumulate in neural tissue, and cause long-term neurocognitive sequelae, has led to concerns regarding its effect on prenatal brain development. We utilized human cortical organoid (hCO) technology to probe how this drug impacts the earliest mechanisms of cortico-genesis. Bulk mRNA sequencing of 2-month-old hCOs chronically treated with a clinically relevant dose of 1 µM methadone for 50 days revealed a robust transcriptional response to methadone associated with functional components of the synapse, the underlying extracellular matrix (ECM), and cilia. Co-expression network and predictive protein-protein interaction analyses demonstrated that these changes occurred in concert, centered around a regulatory axis of growth factors, developmental signaling pathways, and matricellular proteins (MCPs). TGFß1 was identified as an upstream regulator of this network and appeared as part of a highly interconnected cluster of MCPs, of which thrombospondin 1 (TSP1) was most prominently downregulated and exhibited dose-dependent reductions in protein levels. These results demonstrate that methadone exposure during early cortical development alters transcriptional programs associated with synaptogenesis, and that these changes arise by functionally modulating extra-synaptic molecular mechanisms in the ECM and cilia. Our findings provide novel insight into the molecular underpinnings of methadone's putative effect on cognitive and behavioral development and a basis for improving interventions for maternal opioid addiction.


Subject(s)
Opioid-Related Disorders , Substance Withdrawal Syndrome , Humans , Female , Pregnancy , Infant , Methadone/pharmacology , Methadone/therapeutic use , Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , Substance Withdrawal Syndrome/drug therapy , Brain , Opiate Substitution Treatment/methods
13.
Chin J Physiol ; 66(2): 65-72, 2023.
Article in English | MEDLINE | ID: mdl-37082994

ABSTRACT

Prenatal opioid exposure may impede the development of adaptive responses to environmental stimuli by altering the stress-sensitive brain circuitry located at the paraventricular nucleus of the hypothalamus (PVH) and locus coeruleus (LC). Corticotropin-releasing factor (CRF) released from neurons in the PVH has emerged as a key molecule to initiate and integrate the stress response. Methadone (Meth) and buprenorphine (Bu) are two major types of synthetic opioid agonists for first-line medication-assisted treatment of opioid (e.g., morphine, Mor) use disorder in pregnant women. No studies have compared the detrimental effects of prenatal exposure to Meth versus Bu on the stress response of their offspring upon reaching adulthood. In this study, we aimed to compare stress-related neuronal activation in the PVH and LC induced by restraint (RST) stress in adult male rat offspring with prenatal exposure to the vehicle (Veh), Bu, Meth, or Mor. CFos-immunoreactive cells were used as an indicator for neuronal activation. We found that RST induced less neuronal activation in the Meth or Mor exposure groups compared with that in the Bu or Veh groups; no significant difference was detected between the Bu and Veh exposure groups. RST-induced neuronal activation was completely prevented by central administration of a CRF receptor antagonist (α-helical CRF9-41, 10 µg/3 µL) in all exposure groups, suggesting the crucial role of CRF in this stress response. In offspring without RST, central administration of CRF (0.5 µg/3 µL)-induced neuronal activation in the PVH and LC. CRF-induced neuronal activation was lessened in the Meth or Mor exposure groups compared with that in the Bu or Veh groups; no significant difference was detected between the Bu and Veh exposure groups. Moreover, RST- or CRF-induced neuronal activation in the Meth exposure group was comparable with that in the Mor exposure group. Further immunohistochemical analysis revealed that the Meth and Mor exposure groups displayed less CRF neurons in the PVH of offspring with or without RST compared with the Bu or Veh groups. Thus, stress-induced neuronal activation in the PVH and LC was well preserved in adult male rat offspring with prenatal exposure to Bu, but it was substantially lessened in those with prenatal exposure to Meth or Mor. Lowered neuronal activation found in the Meth or Mor exposure groups may be, at least in part, due to the reduction in the density of CRF neurons in the PVH.


Subject(s)
Buprenorphine , Prenatal Exposure Delayed Effects , Rats , Male , Female , Pregnancy , Humans , Animals , Morphine/pharmacology , Methadone/pharmacology , Corticotropin-Releasing Hormone/pharmacology , Corticotropin-Releasing Hormone/physiology , Buprenorphine/pharmacology , Analgesics, Opioid/pharmacology , Rats, Sprague-Dawley , Neurons
14.
Asian Pac J Cancer Prev ; 24(3): 801-810, 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36974531

ABSTRACT

OBJECTIVES: Methadone is an opioid used in treating chronic and acute pains as well as opioid dependence. It induces death in neural cells. This study investigates Punica granatum oil's effects as a natural antioxidant on methadone-induced cell death. MATERIALS AND METHODS: The cell death index indicating the apoptosis occurrence is calculated using the TUNEL test. Rhodamine123 evaluated mitochondrial membrane permeability. Griess reaction was used to detect nitric oxide production. Furthermore, IL-1ß, IL-6, INFγ, and TNFα inflammatory cytokines were measured using the Rat inflammatory cytokine assay kit, Rat Kit V-Plex, and the caspase-3 activity was calculated through the Caspase-3 Colorimetric Assay Kit. RESULTS: Different treatment processes of Punica granatum oil reduced cell cytotoxicity and cell death index and increased viability and proliferation in methadone-treated PC12 cells. NO production decreased in different treatment processes compared to methadone-induced PC12 cells and decreased IL-1ß, IL-6, INFγ, and TNFα inflammatory cytokines. In these treatment processes, mitochondrial membrane potential increased, and caspase-3 activity decreased compared to methadone-induced PC12 cells. CONCLUSION: Punica granatum essential oil declined methadone-induced cell death in PC12 cells in a dose-dependent manner through suppressing NO production, IL-1ß, IL-6, INF-γ, and TNF-α inflammatory cytokines production, mitochondrial membrane disruption, and caspase-3 activities.


Subject(s)
Oils, Volatile , Pomegranate , Rats , Animals , Antioxidants/pharmacology , Antioxidants/metabolism , Tumor Necrosis Factor-alpha/metabolism , Caspase 3/metabolism , Interleukin-6/metabolism , Oils, Volatile/pharmacology , Methadone/pharmacology , Methadone/metabolism , Cytokines/metabolism , Apoptosis , Seeds
15.
Br J Pharmacol ; 180(7): 862-878, 2023 04.
Article in English | MEDLINE | ID: mdl-34128238

ABSTRACT

Opioid use disorder is a worldwide societal problem and public health burden. Strategies for treating opioid use disorder can be divided into those that target the opioid receptor system and those that target non-opioid receptor systems, including the dopamine and glutamate receptor systems. Currently, the clinical drugs used to treat opioid use disorder include the opioid receptor agonists methadone and buprenorphine, which are limited by their abuse liability, and the opioid receptor antagonist naltrexone, which is limited by poor compliance. Therefore, the development of effective medications with lower abuse liability and better potential for compliance is urgently needed. Based on recent advances in the understanding of the neurobiological mechanisms underlying opioid use disorder, potential treatment strategies and targets have emerged. This review focuses on the progress made in identifying potential targets and developing medications to treat opioid use disorder, including progress made by our laboratory, and provides insights for future medication development. LINKED ARTICLES: This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc.


Subject(s)
Buprenorphine , Opioid-Related Disorders , Humans , Opioid-Related Disorders/drug therapy , Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , Methadone/pharmacology , Methadone/therapeutic use , Buprenorphine/pharmacology , Buprenorphine/therapeutic use , Naltrexone/pharmacology , Naltrexone/therapeutic use
16.
Drug Metab Pharmacokinet ; 47: 100477, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36368298

ABSTRACT

Although methadone is effective in the management of acute pain, the complexity of its absorption-distribution-metabolism-excretion profile limits its use as an opioid of choice for perioperative analgesia. Because deuteration is known to improve the pharmacokinetic, pharmacodynamic and toxicological properties of some drugs, here we characterized the single dose pharmacokinetic properties and post-operative analgesic efficacy of d9-methadone. The pharmacokinetic profiles of d9-methadone and methadone administered intravenously to CD-1 male mice revealed that deuteration leads to a 5.7- and 4.4-fold increase in the area under the time-concentration curve and maximum concentration in plasma, respectively, as well as reduction in clearance (0.9 ± 0.3 L/h/kg vs 4.7 ± 0.8 L/h/kg). The lower brain-to-plasma ratio of d9-methadone compared to that of methadone (0.35 ± 0.12 vs 2.05 ± 0.62) suggested that deuteration decreases the transfer of the drug across the blood-brain barrier. The estimated LD50 value for a single intravenous dose of d9-methadone was 2.1-fold higher than that for methadone. Moreover, d9-methadone outperformed methadone in the efficacy against postoperative pain by primarily activating peripheral opioid receptors. Collectively, these data suggest that the replacement of three hydrogen atoms in three methyl groups of methadone altered its pharmacokinetic properties, improved safety, and enhanced its analgesic efficacy.


Subject(s)
Analgesics, Opioid , Methadone , Male , Animals , Mice , Methadone/pharmacology , Methadone/therapeutic use , Analgesics, Opioid/pharmacology , Blood-Brain Barrier , Kinetics , Brain
17.
Top Companion Anim Med ; 51: 100733, 2022.
Article in English | MEDLINE | ID: mdl-36272536

ABSTRACT

Intraocular pressure (IOP), pupil size (PS), and tear production are variables important in maintaining eye homeostasis. The purpose of this study was to determine the effects of methadone on IOP, PS, and tear production measured by Schirmer I tear test (STT-I) in healthy nonpainful dogs. A prospective, randomized, "double-blind" clinical study was performed. A total of 40 healthy conscious client-owned dogs were included in the study. Dogs were allocated randomly to 1 of 3 groups and given intravenous methadone 0.3 mg/kg (Met-IV, n = 15), intramuscular methadone 0.3 mg/kg (Met-IM, n = 15), or saline 0.3 mL/kg (SAL, n = 10). IOP, PS, STT-I, heart rate (HR), and mean arterial pressure (MAP) were measured prior to (baseline) and at 2, 5, 10, 20, and 30 minutes after drug administration. Data were analyzed using 1-way and 2-way repeated measures ANOVA or their nonparametric equivalents (P < .05). No significant differences in IOP and PS within or between the groups were detected. In the Met-IV group, the STT-I decreased significantly after 30 minutes (P = .025), however, the values remained within the physiological ranges. In Met-IV group, HR decreased significantly at 5, 10, 20, and 30 minutes, respectively. No other significant differences were observed. Methadone administered at a dose of 0.3 mg/kg intravenously or intramuscularly seems to cause within 30 minutes no clinically important effect on IOP, PS, and STT-I in healthy conscious nonpainful dogs without ocular abnormalities.


Subject(s)
Dog Diseases , Eye Diseases , Dogs , Animals , Intraocular Pressure , Pupil , Methadone/pharmacology , Prospective Studies , Tonometry, Ocular , Eye Diseases/veterinary
18.
EBioMedicine ; 85: 104283, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36182773

ABSTRACT

BACKGROUND: Opioid use disorder (OUD) is a chronic relapsing psychiatric disorder. An unconditioned stimulus (US)-triggers a memory reconsolidation updating procedure (MRUP) that has been developed and demonstrated its effectiveness in decreasing relapse to cocaine and heroin in preclinical models. However, utilizations of abused drugs as the US to initiate MRUP can be problematic. We therefore designed a translational rat study and human study to evaluate the efficacy of a novel methadone-initiated MRUP. METHODS: In the rodent study, male rats underwent heroin self-administration training for 10 consecutive days, and were randomly assigned to receive saline or methadone at 10 min, 1 h or 6 h before extinction training after 28-day withdrawal. The primary outcome was operant heroin seeking after reinstatement. In the human experimental study, male OUD patients were randomly assigned to get MRUP at 10 min or 6 h after methadone or methadone alone. The primary outcomes included experimental cue-induced heroin craving change, sustained abstinence and retention in the study at post intervention and the 5 monthly follow-up assessments. The secondary outcomes were changes in physiological responses including experimental cue-induced blood pressure and heart rate. FINDINGS: Methadone exposure but not saline exposure at 10 min or 1 h before extinction decreased heroin-induced reinstatement of heroin seeking after 28-day of withdrawal in rats (F (8,80) = 8.26, p < 0.001). In the human study, when the MRUP was performed 10 min, but not 6 h after methadone dosing, the MRUP promoted sustained abstinence from heroin throughout 5 monthly follow-up assessments compared to giving methadone alone without MRUP (Hazard Ratio [95%CI] of 0.43 [0.22, 0.83], p = 0.01). The MRUP at 10 min, but not at 6 h after dosing also decreased experimental cue-induced heroin craving and blood pressure increases during the 6-month study duration (group × months × cue types, F (12, 63·3) = 2.41, p = 0.01). INTERPRETATION: The approach of MRUP within about 1 to 6 h after a methadone dose potently improved several key outcomes of OUD patients during methadone maintenance treatment, and could be a potentially novel treatment to prevent opioid relapse. FUNDING: National Natural Science Foundation of China (NO. U1802283, 81761128036, 82001400, 82001404 and 31671143) and Chinese National Programs for Brain Science and Brain-like Intelligence Technology (NO. 2021ZD0200800).


Subject(s)
Opioid-Related Disorders , Substance Withdrawal Syndrome , Humans , Male , Animals , Rats , Methadone/pharmacology , Methadone/therapeutic use , Heroin/adverse effects , Narcotics/adverse effects , Substance Withdrawal Syndrome/drug therapy , Substance Withdrawal Syndrome/psychology , Substance Withdrawal Syndrome/rehabilitation , Neoplasm Recurrence, Local/drug therapy , Opioid-Related Disorders/drug therapy
19.
Addict Biol ; 27(5): e13215, 2022 09.
Article in English | MEDLINE | ID: mdl-36001432

ABSTRACT

Prenatal opioid exposures lead to extensive cognitive and emotion-regulation problems in children, persisting at least through school-age. Methadone, an opioid typically used for the treatment of opioid use disorder, has been approved for use in pregnant women for several decades. Importantly, however, the impacts of prenatal methadone exposure (PME), particularly on offspring as they progress into adulthood, has not been extensively examined. In recent years, children and young animal models have shown cognitive deficits related to PME, including evidence of hippocampal dysfunction. The present work aims to examine the persistent nature of these deficits, as well as determine how they may differ by sex. Pregnant Sprague-Dawley rats either received subcutaneous methadone or water injections twice daily from gestational days 3-20 or were left undisturbed. Following postnatal day 70, male and female offspring were behaviourally tested for impairments in recognition memory using the Novel Object Recognition task and working spatial memory through Spontaneous Alternation. Additionally, using whole-cell patch-clamp electrophysiology, hippocampal dentate granule cell function was examined in adult offspring. Results indicate that methadone-exposed females showed decreased excitability and increased inhibition of dentate granule cells compared to naïve controls, while males did not. These findings were accompanied by impairments in female working spatial memory and altered behaviour in the Object Recognition task. Overall, this work supports the continued investigation of the long-term effects of PME on adult male and female learning and memory, as well as promotes further exploration of adult hippocampal function as a neural mechanism impacted by this exposure.


Subject(s)
Dentate Gyrus , Prenatal Exposure Delayed Effects , Analgesics, Opioid/pharmacology , Animals , Female , Humans , Male , Memory, Long-Term , Methadone/pharmacology , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Rats , Rats, Sprague-Dawley , Spatial Memory
20.
Neuropsychopharmacology ; 47(12): 2132-2139, 2022 11.
Article in English | MEDLINE | ID: mdl-35906489

ABSTRACT

The high efficacy mu-opioid receptor (MOR) agonist methadone is an effective opioid use disorder (OUD) medication used exclusively in opioid-dependent patients. However, methadone has undesirable effects that limit its clinical efficacy. Intermediate efficacy MOR agonists may treat OUD with fewer undesirable effects. We compared the effects of methadone with the intermediate efficacy MOR agonist TRV130 (oliceridine) on fentanyl-vs.-food choice and somatic withdrawal signs in opioid-dependent and post-opioid-dependent rats. Male rats (n = 20) were trained under a fentanyl-vs.-food choice procedure. Rats were then provided extended fentanyl (3.2 µg/kg/infusion) access (6 p.m.-6 a.m.) for 10 days to produce opioid dependence/withdrawal. Rats were treated with vehicle (n = 7), TRV130 (3.2 mg/kg; n = 8), or methadone (3.2 mg/kg; n = 5) three times per day after each extended-access session (8:30 a.m., 11 a.m., 1:30 p.m.). Withdrawal sign scoring (1:55 p.m.) and choice tests (2-4 p.m.) were conducted daily. Vehicle, TRV130, and methadone effects on fentanyl choice were redetermined in post-opioid-dependent rats. Vehicle-, TRV130-, and methadone-treated rats had similar fentanyl intakes during extended access. Vehicle-treated rats exhibited increased withdrawal signs and decreased bodyweights. Both methadone and TRV130 decreased these withdrawal signs. TRV130 was less effective than methadone to decrease fentanyl choice and increase food choice in opioid-dependent rats. Neither methadone nor TRV130 decreased fentanyl choice in post-opioid-dependent rats. Results suggest that higher MOR activation is required to reduce fentanyl choice than withdrawal signs in fentanyl-dependent rats. Additionally, given that TRV130 did not precipitate withdrawal in opioid-dependent rats, intermediate efficacy MOR agonists like TRV130 may facilitate the transition of patients with OUD from methadone to lower efficacy treatments like buprenorphine.


Subject(s)
Buprenorphine , Opioid-Related Disorders , Substance Withdrawal Syndrome , Analgesics, Opioid , Animals , Buprenorphine/pharmacology , Fentanyl/pharmacology , Male , Methadone/pharmacology , Methadone/therapeutic use , Narcotics , Opioid-Related Disorders/drug therapy , Rats , Receptors, Opioid , Spiro Compounds , Substance Withdrawal Syndrome/drug therapy , Thiophenes
SELECTION OF CITATIONS
SEARCH DETAIL