Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.347
Filtrar
1.
Int Immunopharmacol ; 140: 112736, 2024 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-39088925

RESUMEN

Opioid addiction presents a relevant health challenge, with chronic heroin use linked to detrimental effects on various aspects of physical, mental, and sociological health. Opioid maintenance therapy (OMT), particularly using methadone, is the primary treatment option for heroin addiction. Previous studies using blood samples from current heroin addicts and OMT patients have shown immunomodulatory effects of heroin and methadone on T cell function. However, various additional factors beyond heroin and methadone affect these results, including the consumption of other substances, a stressful lifestyle, comorbid psychological and somatic disorders, as well as additional medications. Therefore, we here investigated the direct effects of heroin and methadone on purified human T cells in vitro. Our results reveal that both, heroin and methadone directly suppress Tcell activation and proliferation. Strikingly, this inhibitory effect was markedly stronger in the presence of methadone, correlating with a decrease in secretion of pro-inflammatory cytokines. While heroin did not interfere with the in vitro differentiation and expansion of regulatory Tcells (Tregs), methadone significantly impaired the proliferation of Tregs. Overall, our findings suggest a direct inhibitory impact of both opioids on effector T cell function in vitro, with methadone additionally interfering with Treg induction and expansion in contrast to heroin.


Asunto(s)
Proliferación Celular , Citocinas , Dependencia de Heroína , Heroína , Activación de Linfocitos , Metadona , Linfocitos T Reguladores , Metadona/farmacología , Humanos , Proliferación Celular/efectos de los fármacos , Activación de Linfocitos/efectos de los fármacos , Células Cultivadas , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Dependencia de Heroína/inmunología , Dependencia de Heroína/tratamiento farmacológico , Citocinas/metabolismo , Tratamiento de Sustitución de Opiáceos , Analgésicos Opioides/farmacología , Masculino , Adulto , Femenino , Diferenciación Celular/efectos de los fármacos
2.
J Psychiatr Res ; 177: 90-95, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38991269

RESUMEN

INTRODUCTION: Attentional bias (AB) is believed to be an important factor in the development and maintenance of both opioid use disorder (OUD) and chronic pain. Cannabis and its main psychoactive constituent, delta-9-tetrahydrocannabinol (THC), produce analgesic effects via processes that are potentially relevant to AB and is commonly used by persons with OUD. This exploratory study investigated if THC influences AB towards pain and opioid cues individuals with OUD. METHODS: Using a within-subject, crossover design, 27 adults receiving methadone were randomly assigned to receive single doses of oral THC (10 mg, 20 mg administered as dronabinol) or placebo across three, 5-h sessions. During each session, a visual probe task was used to measure AB to pain and opioid cues at baseline and 120 min post-THC administration. RESULTS: Mixed-effects models examined main effects of THC dose, time, and their interaction across all participants; findings were then stratified by methadone dose (low dose <90 mg/day and high dose ≥90 mg/day). Among individuals receiving high doses of methadone, a significant interaction was observed such that AB towards opioids increased following 10 mg THC administration and decreased following 20 mg THC administration. Additionally, participants receiving low doses of methadone showed significant increases in the variability of opioid-related AB post THC administration. CONCLUSION: We provide preliminary evidence showing that THC may cause dose-dependent effects on selective attention for opioid cues among methadone patients. These results underscore the need for further clinical investigation into the effects of cannabinoids and other substances with potential analgesic and addictive properties among persons with OUD.


Asunto(s)
Sesgo Atencional , Estudios Cruzados , Dronabinol , Trastornos Relacionados con Opioides , Humanos , Dronabinol/farmacología , Dronabinol/administración & dosificación , Masculino , Femenino , Adulto , Sesgo Atencional/efectos de los fármacos , Sesgo Atencional/fisiología , Trastornos Relacionados con Opioides/tratamiento farmacológico , Metadona/administración & dosificación , Metadona/farmacología , Adulto Joven , Dolor/tratamiento farmacológico , Persona de Mediana Edad , Método Doble Ciego , Relación Dosis-Respuesta a Droga , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/farmacología , Señales (Psicología)
3.
Sci Rep ; 14(1): 16978, 2024 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-39043899

RESUMEN

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.


Asunto(s)
Apoptosis , Astrocitos , Metadona , Microglía , Neuronas , Oligodendroglía , Metadona/farmacología , Animales , Ratas , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Apoptosis/efectos de los fármacos , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Microglía/efectos de los fármacos , Microglía/metabolismo , Células Cultivadas , Femenino , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Embarazo , Analgésicos Opioides/farmacología , Ratas Sprague-Dawley
4.
Sci Rep ; 14(1): 14350, 2024 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-38906975

RESUMEN

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.


Asunto(s)
Precondicionamiento Isquémico Miocárdico , Metadona , Daño por Reperfusión Miocárdica , Proteínas Proto-Oncogénicas c-akt , Factor de Transcripción STAT3 , Animales , Masculino , Ratas , Precondicionamiento Isquémico Miocárdico/métodos , Metadona/farmacología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/patología , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Wistar , Daño por Reperfusión/metabolismo , Daño por Reperfusión/prevención & control , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/patología , Transducción de Señal/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo
5.
Vet Anaesth Analg ; 51(3): 253-265, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38580536

RESUMEN

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.


Asunto(s)
Dexmedetomidina , Isoflurano , Metadona , Propofol , Pirazoles , Animales , Perros , Dexmedetomidina/administración & dosificación , Dexmedetomidina/farmacología , Propofol/administración & dosificación , Propofol/farmacología , Metadona/administración & dosificación , Metadona/farmacología , Femenino , Isoflurano/administración & dosificación , Isoflurano/farmacología , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Presión Sanguínea/efectos de los fármacos , Hipnóticos y Sedantes/farmacología , Hipnóticos y Sedantes/administración & dosificación , Quinolizinas/farmacología , Quinolizinas/administración & dosificación , Anestésicos Intravenosos/administración & dosificación , Anestésicos Intravenosos/farmacología , Anestésicos por Inhalación/administración & dosificación , Anestésicos por Inhalación/farmacología , Premedicación/veterinaria
6.
Psychopharmacology (Berl) ; 241(9): 1791-1813, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38642101

RESUMEN

RATIONALE AND OBJECTIVE: Avoidance of opioid withdrawal plays a key role in human opioid addiction. Here, we present a procedure for studying operant negative reinforcement in rats that was inspired by primate procedures where opioid-dependent subjects lever-press to prevent naloxone infusions. METHODS: In Experiment 1, we trained rats (n = 30, 15 females) to lever-press to escape and then avoid mild footshocks (0.13-0.27 mA) for 35 days (30 trials/d). Next, we catheterized them and implanted minipumps containing methadone (10 mg/kg/day) or saline. We then paired (4 times, single session) a light cue (20-s) with a naloxone infusion (20 µg/kg, i.v) that precipitated opioid withdrawal. Next, we trained the rats to escape naloxone injections for 10 days (30 trials/d). Each trial started with the onset of the opioid-withdrawal cue. After 20-s, the lever extended, and an infusion of naloxone (1 to 2.2 µg/kg/infusion) began; a lever-press during an 11-s window terminated the withdrawal-paired cue and the infusion. In Experiment 2, we trained rats (n = 34, 17 females) on the same procedure but decreased the footshock escape/avoidance training to 20 days. RESULTS: All rats learned to lever-press to escape or avoid mild footshocks. In both experiments, a subset, 56% (10/18) and 33% (8/24) of methadone-dependent rats learned to lever-press to escape naloxone infusions. CONCLUSIONS: We introduce an operant negative reinforcement procedure where a subset of opioid-dependent rats learned to lever-press to escape withdrawal-inducing naloxone infusions. The procedure can be used to study mechanisms of individual differences in opioid negative reinforcement-related behaviors in opioid-dependent rats.


Asunto(s)
Condicionamiento Operante , Naloxona , Antagonistas de Narcóticos , Trastornos Relacionados con Opioides , Refuerzo en Psicología , Síndrome de Abstinencia a Sustancias , Animales , Masculino , Femenino , Naloxona/farmacología , Naloxona/administración & dosificación , Ratas , Condicionamiento Operante/efectos de los fármacos , Antagonistas de Narcóticos/farmacología , Antagonistas de Narcóticos/administración & dosificación , Metadona/administración & dosificación , Metadona/farmacología , Ratas Sprague-Dawley , Analgésicos Opioides/farmacología , Analgésicos Opioides/administración & dosificación , Modelos Animales de Enfermedad , Señales (Psicología)
7.
Int J Mol Sci ; 25(4)2024 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-38396814

RESUMEN

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.


Asunto(s)
Analgésicos Opioides , Trampas Extracelulares , Humanos , Animales , Ratones , Analgésicos Opioides/farmacología , Receptor Toll-Like 4/metabolismo , Metadona/farmacología , Mastocitos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Médula Ósea/metabolismo , Calcio/metabolismo , Trampas Extracelulares/metabolismo , Receptor Toll-Like 2/metabolismo , Receptores Toll-Like/metabolismo
8.
Vet J ; 303: 106065, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38228282

RESUMEN

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.


Asunto(s)
Analgésicos , Dexmedetomidina , Hipnóticos y Sedantes , Metadona , Animales , Perros , Administración Intranasal/veterinaria , Analgésicos/farmacología , Dexmedetomidina/farmacología , Combinación de Medicamentos , Hipnóticos y Sedantes/farmacología , Metadona/farmacología , Estudios Prospectivos , Sinergismo Farmacológico
9.
J Vet Pharmacol Ther ; 47(2): 73-79, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37930056

RESUMEN

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.


Asunto(s)
Analgésicos Opioides , Hipotermia , Perros , Masculino , Animales , Analgésicos Opioides/farmacología , Butorfanol/farmacología , Hipotermia/inducido químicamente , Hipotermia/prevención & control , Hipotermia/veterinaria , Metadona/farmacología , Administración Intravenosa/veterinaria
10.
Mol Psychiatry ; 29(3): 624-632, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38145984

RESUMEN

(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.


Asunto(s)
Analgésicos Opioides , Metadona , Receptores Opioides mu , Receptores Opioides mu/metabolismo , Receptores Opioides mu/efectos de los fármacos , Animales , Metadona/farmacología , Masculino , Analgésicos Opioides/farmacología , Humanos , Ratones , Dopamina/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Ligandos , Estereoisomerismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA