Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 197
Filtrar
1.
Sci Rep ; 10(1): 5265, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32210319

RESUMO

Anesthetics are used to produce hypnosis and analgesic effects during surgery, but anesthesia for a long time after the operation is not conducive to the recovery of animals or patients. Therefore, finding appropriate treatments to counter the effects of anesthetics could enhance postoperative recovery. In the current study, we discovered the novel role of a GluN2A-selective positive allosteric modulator (PAM) in ketamine-induced anesthesia and investigated the effects of the PAM combined with nalmefene and flumazenil (PNF) in reversing the actions of an anesthetic combination (ketamine-fentanyl-dexmedetomidine, KFD). PAM treatment dose-dependently decreased the duration of the ketamine-induced loss of righting reflex (LORR). Compared with those in the KFD group, the duration of LORR and the analgesic effect of the KFD + PNF group were obviously decreased. Meanwhile, successive administration of PNF and KFD had no adverse effects on the cardiovascular and respiratory systems. Both the KFD group and the KFD + PNF group showed no changes in hepatic and renal function or cognitive function in rats. Moreover, the recovery of motor coordination of the KFD + PNF group was faster than that of the KFD group. In summary, our results suggest the potential application of the PNF combination as an antagonistic treatment strategy for anesthesia.


Assuntos
Analgesia , Anestesia , Dexmedetomidina/antagonistas & inibidores , Fentanila/antagonistas & inibidores , Flumazenil/farmacologia , Antagonistas de Receptores de GABA-A/farmacologia , Ketamina/antagonistas & inibidores , Naltrexona/análogos & derivados , Antagonistas de Entorpecentes/farmacologia , Receptores de N-Metil-D-Aspartato/agonistas , Adjuvantes Anestésicos , Regulação Alostérica , Animais , Recuperação Demorada da Anestesia/tratamento farmacológico , Combinação de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Feminino , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Naltrexona/farmacologia , Nociceptividade/efeitos dos fármacos , Medição da Dor , Ratos , Reflexo de Endireitamento/efeitos dos fármacos , Teste de Desempenho do Rota-Rod
2.
Toxicol Lett ; 325: 34-42, 2020 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-32070766

RESUMO

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.


Assuntos
Analgésicos Opioides/toxicidade , Fentanila/análogos & derivados , Naloxona/uso terapêutico , Antagonistas de Entorpecentes/uso terapêutico , Analgésicos Opioides/antagonistas & inibidores , Animais , Apneia/induzido quimicamente , Apneia/prevenção & controle , Comportamento Animal/efeitos dos fármacos , Chlorocebus aethiops , Condicionamento Operante/efeitos dos fármacos , Overdose de Drogas/tratamento farmacológico , Fentanila/antagonistas & inibidores , Fentanila/toxicidade , Masculino , Naloxona/toxicidade , Antagonistas de Entorpecentes/toxicidade , Tempo de Reação/efeitos dos fármacos
3.
Drug Alcohol Depend ; 204: 107501, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31479865

RESUMO

BACKGROUND: One emerging strategy to address the opioid crisis includes opioid-targeted immunopharmacotherapies. This study compared effectiveness of a heroin-tetanus toxoid (TT) conjugate vaccine to antagonize heroin, 6-acetylmorphine (6-AM), morphine, and fentanyl antinociception in rats. METHODS: Adult male and female Sprague Dawley rats received three doses of active or control vaccine at weeks 0, 2, and 4. Vaccine pharmacological selectivity was assessed by comparing opioid dose-effect curves in 50 °C warm-water tail-withdrawal procedure before and after active or control heroin-TT vaccine. Route of heroin administration [subcutaneous (SC) vs. intravenous [IV)] was also examined as a determinant of vaccine effectiveness. Continuous naltrexone treatment (0.0032-0.032 mg/kg/h) effects on heroin, 6-AM, and morphine antinociceptive potency were also determined as a benchmark for minimal vaccine effectiveness. RESULTS: The heroin-TT vaccine decreased potency of SC heroin (5-fold), IV heroin (3-fold), and IV 6-AM (3-fold) for several weeks without affecting IV morphine or SC and IV fentanyl potency. The control vaccine did not alter potency of any opioid. Naltrexone dose-dependently decreased antinociceptive potency of SC heroin, and treatment with 0.01 mg/kg/h naltrexone produced similar, approximate 8-fold decreases in potencies of SC and IV heroin, IV 6-AM, and IV morphine. The combination of naltrexone and active vaccine was more effective than naltrexone alone to antagonize SC heroin but not IV heroin. CONCLUSIONS: The heroin-TT vaccine formulation examined is less effective, but more selective, than chronic naltrexone to attenuate heroin antinociception in rats. Furthermore, these results provide an empirical framework for future preclinical opioid vaccine research to benchmark effectiveness against naltrexone.


Assuntos
Fentanila/antagonistas & inibidores , Heroína/antagonistas & inibidores , Heroína/farmacologia , Derivados da Morfina/antagonistas & inibidores , Morfina/antagonistas & inibidores , Naltrexona/farmacologia , Vacinação , Analgésicos Opioides/antagonistas & inibidores , Analgésicos Opioides/farmacologia , Animais , Relação Dose-Resposta a Droga , Vias de Administração de Medicamentos , Feminino , Fentanila/farmacologia , Masculino , Morfina/farmacologia , Derivados da Morfina/farmacologia , Antagonistas de Entorpecentes/farmacologia , Ratos , Toxoide Tetânico/farmacologia
4.
J Pharmacol Exp Ther ; 368(2): 282-291, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30409833

RESUMO

Fentanyl is an extremely potent synthetic opioid that has been increasingly used to adulterate heroin, cocaine, and counterfeit prescription pills, leading to an increase in opioid-induced fatal overdoses in the United States, Canada, and Europe. A vaccine targeting fentanyl could offer protection against the toxic effects of fentanyl in both recreational drug users and others in professions at risk of accidental exposure. This study focuses on the development of a vaccine consisting of a fentanyl-based hapten (F) conjugated to keyhole limpet hemocyanin (KLH) carrier protein or to GMP-grade subunit KLH (sKLH). Immunization with F-KLH in mice and rats reduced fentanyl-induced hotplate antinociception, and in rats reduced fentanyl distribution to the brain compared with controls. F-KLH did not reduce the antinociceptive effects of equianalgesic doses of heroin or oxycodone in rats. To assess the vaccine effect on fentanyl toxicity, rats immunized with F-sKLH or unconjugated sKLH were exposed to increasing subcutaneous doses of fentanyl. Vaccination with F-sKLH shifted the dose-response curves to the right for both fentanyl-induced antinociception and respiratory depression. Naloxone reversed fentanyl effects in both groups, showing that its ability to reverse respiratory depression was preserved. These data demonstrate preclinical selectivity and efficacy of a fentanyl vaccine and suggest that vaccines may offer a therapeutic option in reducing fentanyl-induced side effects.


Assuntos
Analgésicos Opioides/antagonistas & inibidores , Analgésicos Opioides/metabolismo , Fentanila/antagonistas & inibidores , Fentanila/metabolismo , Vacinas/farmacologia , Analgésicos Opioides/toxicidade , Animais , Bradicardia/sangue , Bradicardia/induzido quimicamente , Bradicardia/prevenção & controle , Fentanila/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Ratos , Ratos Sprague-Dawley , Insuficiência Respiratória/sangue , Insuficiência Respiratória/induzido quimicamente , Insuficiência Respiratória/prevenção & controle , Vacinas/uso terapêutico
5.
Eur J Pharmacol ; 833: 275-282, 2018 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-29886241

RESUMO

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


Assuntos
Analgésicos Opioides/farmacologia , Dor do Câncer/tratamento farmacológico , Fentanila/farmacologia , Hiperalgesia/tratamento farmacológico , Receptores Opioides mu/metabolismo , Potenciais de Ação/efeitos dos fármacos , Analgésicos Opioides/antagonistas & inibidores , Analgésicos Opioides/uso terapêutico , Animais , Dor do Câncer/etiologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Fentanila/antagonistas & inibidores , Fentanila/uso terapêutico , Humanos , Hiperalgesia/etiologia , Injeções Intramusculares , Injeções Intravenosas , Masculino , Melanoma/complicações , Camundongos , Camundongos Endogâmicos C57BL , Naloxona/análogos & derivados , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Compostos de Amônio Quaternário/farmacologia , Receptores Opioides , Pele/patologia , Neoplasias Cutâneas/complicações , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Nervo Tibial/efeitos dos fármacos , Nervo Tibial/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Bull Exp Biol Med ; 163(6): 737-741, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29063329

RESUMO

We compared samples of microencapsulated naloxone prepared by using spray drying technique. 2-Hydroxypropyl-ß-cyclodextrin, sodium alginate, polycaprolactone, and carboxymethyl cellulose were used as the carriers. It was found that the combination of naloxone with sodium alginate was characterized by the highest naloxone content in the matrix and the lowest release rate (100% release time was 60 min). Using the model of respiratory disturbances caused by 10 ED50 fentanyl (anesthetic effect), we studied the effects of naloxone-sodium alginate complex on the dynamics of CO2 concentration in the expired air. It was shown that treatment with the developed microencapsulated naloxone after fentanyl injection allowed reducing the therapeutic dose of the antagonist by more than 2 times and eliminated the necessity of repeated injections.


Assuntos
Portadores de Fármacos , Fentanila/intoxicação , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Entorpecentes/intoxicação , 2-Hidroxipropil-beta-Ciclodextrina/química , Alginatos/química , Animais , Animais não Endogâmicos , Carboximetilcelulose Sódica/química , Composição de Medicamentos/métodos , Liberação Controlada de Fármacos , Fentanila/antagonistas & inibidores , Fentanila/toxicidade , Ácido Glucurônico/química , Ácidos Hexurônicos/química , Cinética , Masculino , Naloxona/metabolismo , Antagonistas de Entorpecentes/metabolismo , Entorpecentes/toxicidade , Poliésteres/química , Ratos , Respiração/efeitos dos fármacos
7.
J Spec Oper Med ; 17(3): 120-122, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28910481

RESUMO

Law enforcement officers, whether working the streets or on narcotic detail, and even those who operate in strike teams, face a new danger from an old drug: carfentanil. Drug dealers seeking to increase profits cut this cheap synthetic drug into expensive heroin, providing an extreme high. As a potent synthetic opioid narcotic, it is finding its way to the streets of the United States and can pose a threat to life for law enforcement, first responders, and medical examiners.


Assuntos
Analgésicos Opioides/efeitos adversos , Médicos Legistas , Fentanila/análogos & derivados , Militares , Exposição Ocupacional/efeitos adversos , Polícia , Analgésicos Opioides/antagonistas & inibidores , Fentanila/efeitos adversos , Fentanila/antagonistas & inibidores , Humanos , Naloxona/uso terapêutico , Naltrexona/uso terapêutico , Antagonistas de Entorpecentes/uso terapêutico , Estados Unidos
8.
Med Sci Sports Exerc ; 49(12): 2404-2413, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28767527

RESUMO

PURPOSE: The purpose of this study was to investigate the role of the group III/IV muscle afferents in the bioenergetics of exercising skeletal muscle beyond constraining the magnitude of metabolic perturbation. METHODS: Eight healthy men performed intermittent isometric knee-extensor exercise to task failure at ~58% maximal voluntary contraction under control conditions (CTRL) and with lumbar intrathecal fentanyl to attenuate group III/IV leg muscle afferents (FENT). Intramuscular concentrations of phosphocreatine (PCr), inorganic phosphate (Pi), diprotonated phosphate (H2PO4), adenosine triphosphate (ATP), and pH were determined using phosphorous magnetic resonance spectroscopy (P-MRS). RESULTS: The magnitude of metabolic perturbation was significantly greater in FENT compared with CTRL for [Pi] (37.8 ± 16.8 vs 28.6 ± 8.6 mM), [H2PO4] (24.3 ± 12.2 vs 17.9 ± 7.1 mM), and [ATP] (75.8% ± 17.5% vs 81.9% ± 15.8% of baseline), whereas there was no significant difference in [PCr] (4.5 ± 2.4 vs 4.4 ± 2.3 mM) or pH (6.51 ± 0.10 vs 6.54 ± 0.14). The rate of perturbation in [PCr], [Pi], [H2PO4], and pH was significantly faster in FENT compared with CTRL. Oxidative ATP synthesis was not significantly different between conditions. However, anaerobic ATP synthesis, through augmented creatine kinase and glycolysis reactions, was significantly greater in FENT than in CTRL, resulting in a significantly greater ATP cost of contraction (0.049 ± 0.016 vs 0.038 ± 0.010 mM·min·N). CONCLUSION: Group III/IV muscle afferents not only constrain the magnitude of perturbation in intramuscular Pi, H2PO4, and ATP during small muscle mass exercise but also seem to play a role in maintaining efficient skeletal muscle contractile function in men.


Assuntos
Trifosfato de Adenosina/biossíntese , Vias Aferentes/fisiologia , Metabolismo Energético/fisiologia , Exercício Físico/fisiologia , Músculo Esquelético/inervação , Músculo Esquelético/fisiologia , Adulto , Vias Aferentes/efeitos dos fármacos , Creatina Quinase/metabolismo , Tolerância ao Exercício/fisiologia , Fentanila/antagonistas & inibidores , Fentanila/farmacologia , Glicólise/fisiologia , Humanos , Concentração de Íons de Hidrogênio , Joelho/fisiologia , Masculino , Músculo Esquelético/efeitos dos fármacos , Percepção , Fosfatos/metabolismo , Fosfocreatina/metabolismo , Esforço Físico/fisiologia
9.
Vet Anaesth Analg ; 44(1): 138-143, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27302030

RESUMO

OBJECTIVE: To characterize a propofol-medetomidine-ketamine total intravenous anaesthetic in impala (Aepyceros melampus). STUDY DESIGN: Prospective clinical study. ANIMALS: Ten adult female impala. MATERIALS AND METHODS: Impala were immobilized at 1253 m above sea level with 2.0 mg thiafentanil and 2.2 mg medetomidine via projectile darts. Propofol was given to effect (0.5 mg kg-1 boluses) to allow endotracheal intubation, following which oxygen was supplemented at 2 L minute-1. Anaesthesia was maintained with a constant-rate infusion of medetomidine and ketamine at 5 µg kg-1 hour-1 and 1.5 mg kg-1 hour-1, respectively, and propofol to effect (initially 0.2 mg kg-1 minute-1) for 120 minutes. The propofol infusion was titrated according to reaction to nociceptive stimuli every 15 minutes. Cardiopulmonary parameters were monitored continuously and arterial blood gas samples were analysed intermittently. After 120 minutes' maintenance, the thiafentanil and medetomidine were antagonized using naltrexone (10:1 thiafentanil) and atipamezole (5:1 medetomidine), respectively. RESULTS: All impala were successfully immobilized. The median dose [interquartile range (IQR)] of propofol required for intubation was 2.7 (1.9-3.3) mg kg-1. The propofol-medetomidine-ketamine combination abolished voluntary movement and ensured anaesthesia for the 120 minute period. Propofol titration showed a generally downward trend. Median (IQR) heart rate [57 (53-61) beats minute-1], respiratory rate [10 (9-12) breaths minute-1] and mean arterial blood pressure [101 (98-106) mmHg] were well maintained. Arterial blood gas analysis indicated hypoxaemia, hyper- capnia and acidaemia. Butorphanol (0.12 mg kg-1) was an essential rescue drug to counteract thiafentanil-induced respiratory depression. All impala regurgitated frequently during the maintenance period. Recovery was calm and rapid in all animals. Median (IQR) time to standing from antagonist administration was 4.4 (3.2-5.6) minutes. CONCLUSIONS AND CLINICAL RELEVANCE: A propofol-medetomidine-ketamine combination could provide adequate anaesthesia for invasive procedures in impala. The propofol infusion should begin at 0.2 mg kg-1 minute-1 and be titrated to clinical effect. Oxygen supplementation and airway protection with a cuffed endotracheal tube are essential.


Assuntos
Anestesia Intravenosa/veterinária , Anestésicos Combinados/administração & dosagem , Antílopes , Fentanila/análogos & derivados , Hipnóticos e Sedativos/administração & dosagem , Ketamina/administração & dosagem , Medetomidina/administração & dosagem , Propofol/administração & dosagem , Antagonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Anestesia Intravenosa/métodos , Animais , Feminino , Fentanila/administração & dosagem , Fentanila/antagonistas & inibidores , Frequência Cardíaca/efeitos dos fármacos , Hipnóticos e Sedativos/antagonistas & inibidores , Imidazóis/administração & dosagem , Medetomidina/antagonistas & inibidores , Naltrexona/administração & dosagem , Antagonistas de Entorpecentes/administração & dosagem , Estudos Prospectivos , Taxa Respiratória/efeitos dos fármacos
10.
Lab Anim ; 50(4): 264-74, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26860578

RESUMO

Injection anaesthesia is commonly used in laboratory mice; however, a disadvantage is that post-anaesthesia recovery phases are long. Here, we investigated the potential for shortening the recovery phase after injection anaesthesia with fentanyl-midazolam-medetomidine by antagonization with naloxone-flumazenil-atipamezole. In order to monitor side-effects, the depth of anaesthesia, heart rate (HR), core body temperature (BT) and concentration of blood gases, as well as reflex responses, were assessed during a 50 min anaesthesia. Mice were allowed to recover from the anaesthesia in their home cages either with or without antagonization, while HR, core BT and spontaneous home cage behaviours were recorded for 24 h. Mice lost righting reflex at 330 ± 47 s after intraperitoneal injection of fentanyl-midazolam-medetomidine. During anaesthesia, HR averaged 225 ± 23 beats/min, respiratory rate and core BT reached steady state at 131 ± 15 breaths/min and 34.3 ± 0.25℃, respectively. Positive pedal withdrawal reflex, movement triggered by tail pinch and by toe pinch, still occurred in 25%, 31.2% and 100% of animals, respectively. Arterial blood gas analysis revealed acidosis, hypoxia, hypercapnia and a marked increase in glucose concentration. After anaesthesia reversal by injection with naloxone-flumazenil-atipamezole, animals regained consciousness after 110 ± 18 s and swiftly returned to physiological baseline values, yet they displayed diminished levels of locomotion and disrupted circadian rhythm. Without antagonization, mice showed marked hypothermia (22 ± 1.9℃) and bradycardia (119 ± 69 beats/min) for several hours. Fentanyl-midazolam-medetomidine provided reliable anaesthesia in mice with reasonable intra-anaesthetic side-effects. Post-anaesthetic period and related adverse effects were both reduced substantially by antagonization with naloxone-flumazenil-atipamezole.


Assuntos
Anestésicos Combinados/farmacologia , Fentanila/farmacologia , Medetomidina/farmacologia , Midazolam/farmacologia , Assistência Perioperatória , Analgésicos/efeitos adversos , Analgésicos/farmacologia , Anestésicos Combinados/efeitos adversos , Animais , Temperatura Corporal/efeitos dos fármacos , Feminino , Fentanila/efeitos adversos , Fentanila/antagonistas & inibidores , Flumazenil/farmacologia , Frequência Cardíaca/efeitos dos fármacos , Imidazóis/farmacologia , Injeções Intraperitoneais/efeitos adversos , Medetomidina/efeitos adversos , Medetomidina/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Midazolam/efeitos adversos , Midazolam/antagonistas & inibidores , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Neurotransmissores/farmacologia
11.
J Pharmacol Exp Ther ; 357(1): 17-23, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26801398

RESUMO

Nociceptin/orphanin FQ receptor (NOP) agonists have been reported to produce antinociceptive effects in rhesus monkeys with comparable efficacy to µ-opioid receptor (MOP) agonists, but without their limiting side effects. There are also known to be species differences between rodents and nonhuman primates (NHPs) in the behavioral effects of NOP agonists. The aims of this study were the following: 1) to determine if the NOP agonist Ro 64-6198 could be trained as a discriminative stimulus; 2) to evaluate its pharmacological selectivity as a discriminative stimulus; and 3) to establish the order of potency with which Ro 64-6198 produces discriminative stimulus effects compared with analgesic effects in NHPs. Two groups of rhesus monkeys were trained to discriminate either fentanyl or Ro 64-6198 from vehicle. Four monkeys were trained in the warm-water tail-withdrawal procedure to measure antinociception. Ro 64-6198 produced discriminative stimulus effects that were blocked by the NOP antagonist J-113397 and not by naltrexone. The discriminative stimulus effects of Ro 64-6198 partially generalized to diazepam, but not to fentanyl, SNC 80, ketocyclazocine, buprenorphine, phencyclidine, or chlorpromazine. Fentanyl produced stimulus effects that were blocked by naltrexone and not by J-113397, and Ro 64-6198 did not produce fentanyl-appropriate responding in fentanyl-trained animals. In measures of antinociception, fentanyl, but not Ro 64-6198, produced dose-dependent increases in tail-withdrawal latency. Together, these results demonstrate that Ro 64-6198 produced stimulus effects in monkeys that are distinct from other opioid receptor agonists, but may be somewhat similar to diazepam. In contrast to previous findings, Ro 64-6198 did not produce antinociception in the majority of animals tested even at doses considerably greater than those that produced discriminative stimulus effects.


Assuntos
Discriminação Psicológica/efeitos dos fármacos , Imidazóis/farmacologia , Receptores Opioides/agonistas , Compostos de Espiro/farmacologia , Analgésicos Opioides/farmacologia , Animais , Benzimidazóis/farmacologia , Condicionamento Operante/efeitos dos fármacos , Diazepam/farmacologia , Relação Dose-Resposta a Droga , Feminino , Fentanila/antagonistas & inibidores , Fentanila/farmacologia , Moduladores GABAérgicos/farmacologia , Macaca mulatta , Masculino , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Medição da Dor/efeitos dos fármacos , Piperidinas/farmacologia , Receptor de Nociceptina
12.
J Clin Gastroenterol ; 50(3): e25-9, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25626630

RESUMO

BACKGROUND: Moderate sedation has been standard for noninvasive gastrointestinal procedures for decades yet there are limited data on reversal agent use and outcomes associated with need for reversal of sedation. AIM: To determine prevalence and clinical significance of reversal agent use during endoscopies and colonoscopies. METHODS: Individuals with adverse events requiring naloxone and/or flumazenil during endoscopy or colonoscopy from 2008 to 2013 were identified. A control group was obtained by random selection of patients matched by procedure type and date. Prevalence of reversal agent use and statistical comparison of patient demographics and risk factors against controls were determined. RESULTS: Prevalence of reversal agent use was 0.03% [95% confidence interval (CI), 0.02-0.04]. Events triggering reversal use were oxygen desaturation (64.4%), respiration changes (24.4%), hypotension (8.9%), and bradycardia (6.7%). Two patients required escalation of care and the majority of patients were stabilized and discharged home. Compared with the control group, the reversal group was older (61±1.8 vs. 55±1.6, P=0.01), mostly female (82% vs. 50%, P<0.01), and had lower body mass index (24±0.8 vs. 27±0.7, P=0.03) but received similar dosages of sedation. When adjusted for age, race, sex, and body mass index, the odds of reversal agent patients having a higher ASA score than controls was 4.7 (95% CI, 1.7-13.1), and the odds of having a higher Mallampati score than controls was 5.0 (95% CI, 2.1-11.7) with P<0.01. CONCLUSIONS: Prevalence of reversal agent use during moderate sedation is low and outcomes are generally good. Several clinically relevant risk factors for reversal agent use were found suggesting that certain groups may benefit from closer monitoring.


Assuntos
Antídotos/administração & dosagem , Colonoscopia , Sedação Consciente/efeitos adversos , Nível de Saúde , Hipnóticos e Sedativos/antagonistas & inibidores , Antagonistas de Entorpecentes/administração & dosagem , Fatores Etários , Antiarrítmicos/administração & dosagem , Atropina/administração & dosagem , Índice de Massa Corporal , Bradicardia/induzido quimicamente , Bradicardia/tratamento farmacológico , Estudos de Casos e Controles , Colonoscopia/efeitos adversos , Feminino , Fentanila/efeitos adversos , Fentanila/antagonistas & inibidores , Flumazenil/administração & dosagem , Humanos , Hipnóticos e Sedativos/efeitos adversos , Hipotensão/induzido quimicamente , Hipotensão/tratamento farmacológico , Hipóxia/induzido quimicamente , Hipóxia/tratamento farmacológico , Masculino , Midazolam/efeitos adversos , Midazolam/antagonistas & inibidores , Pessoa de Meia-Idade , Naloxona/administração & dosagem , Fatores Sexuais , Resultado do Tratamento
13.
Anesthesiology ; 122(2): 424-34, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25313880

RESUMO

BACKGROUND: There is an unmet clinical need to develop a pharmacological therapy to counter opioid-induced respiratory depression without interfering with analgesia or behavior. Several studies have demonstrated that 5-HT1A receptor agonists alleviate opioid-induced respiratory depression in rodent models. However, there are conflicting reports regarding their effects on analgesia due in part to varied agonist receptor selectivity and presence of anesthesia. Therefore the authors performed a study in rats with befiradol (F13640 and NLX-112), a highly selective 5-HT1A receptor agonist without anesthesia. METHODS: Respiratory neural discharge was measured using in vitro preparations. Plethysmographic recording, nociception testing, and righting reflex were used to examine respiratory ventilation, analgesia, and sedation, respectively. RESULTS: Befiradol (0.2 mg/kg, n = 6) reduced fentanyl-induced respiratory depression (53.7 ± 5.7% of control minute ventilation 4 min after befiradol vs. saline 18.7 ± 2.2% of control, n = 9; P < 0.001), duration of analgesia (90.4 ± 11.6 min vs. saline 130.5 ± 7.8 min; P = 0.011), duration of sedation (39.8 ± 4 min vs. saline 58 ± 4.4 min; P = 0.013); and induced baseline hyperventilation, hyperalgesia, and "behavioral syndrome" in nonsedated rats. Further, the befiradol-induced alleviation of opioid-induced respiratory depression involves sites or mechanisms not functioning in vitro brainstem-spinal cord and medullary slice preparations. CONCLUSIONS: The reversal of opioid-induced respiratory depression and sedation by befiradol in adult rats was robust, whereas involved mechanisms are unclear. However, there were adverse concomitant decreases in fentanyl-induced analgesia and altered baseline ventilation, nociception, and behavior.


Assuntos
Analgesia , Anestésicos Intravenosos/farmacologia , Sedação Consciente , Fentanila/antagonistas & inibidores , Piperidinas/farmacologia , Piridinas/farmacologia , Receptor 5-HT1A de Serotonina/efeitos dos fármacos , Insuficiência Respiratória/prevenção & controle , Agonistas do Receptor 5-HT1 de Serotonina/farmacologia , Envelhecimento/fisiologia , Anestésicos Intravenosos/toxicidade , Animais , Nível de Alerta , Comportamento Animal/efeitos dos fármacos , Tronco Encefálico/efeitos dos fármacos , Fentanila/farmacologia , Fentanila/toxicidade , Técnicas In Vitro , Bulbo/efeitos dos fármacos , Medição da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Insuficiência Respiratória/induzido quimicamente , Medula Espinal/efeitos dos fármacos
14.
Eur J Pharmacol ; 738: 153-7, 2014 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-24886878

RESUMO

Reversing the respiratory depression induced by carfentanil involves intravenous administration of naloxone or naltrexone, but this treatment has disadvantages. Hence, finding a more appropriate treatment to counter the depressive actions of carfentanil is needed. In the present study, with the naloxone as a control, we investigated the efficacy of nalmefene for countering the depressive actions of carfentanil. Rats were treated successively with carfentanil (10 µg/kg, i.v.) and nalmefene (9.4-150.0 µg/kg, i.m.), and the duration of loss of righting reflex (LORR) recorded. Respiratory parameters were measured in free-moving rats using a whole-body plethysmograph after rats were administered carfentanil (20 µg/kg, i.v.) and nalmefene (9.4-150.0 µg/kg, i.m.) sequentially. The parameters of arterial blood gases were also examined. Nalmefene (9.4-150.0 µg/kg, i.m.) treatment dose-dependently decreased the duration of carfentanil-induced LORR. The respiratory rate after 60 min of nalmefene (150.0 µg/kg, i.m.) treatment increased from 34.3 ± 5.3 bursts/min to 117.8 ± 18.9 bursts/min, and enhanced pause decreased from 1.1 ± 0.1 to 0.4 ± 0.1, and was close to those of normal rats. Furthermore, nalmefene (37.5-150.0 µg/kg) treatment could enable the PaO2, SaO2 and PaCO2 to approach normal levels 10 min (15 min after carfentanil injection) or 30 min (25 min after carfentanil injection) after injection. While, a single injection of naloxone (150.0 µg/kg, i.m.) only achieved partial remission of respiratory depression. These data suggest that nalmefene more effectively counters the depressive actions induced by carfentanil and is a more appropriate treatment to antagonize carfentanil toxicity compared with naloxone.


Assuntos
Analgésicos Opioides/efeitos adversos , Fentanila/análogos & derivados , Naltrexona/análogos & derivados , Reflexo de Endireitamento/efeitos dos fármacos , Insuficiência Respiratória/induzido quimicamente , Insuficiência Respiratória/tratamento farmacológico , Analgésicos Opioides/antagonistas & inibidores , Animais , Gasometria , Relação Dose-Resposta a Droga , Fentanila/efeitos adversos , Fentanila/antagonistas & inibidores , Masculino , Naltrexona/farmacologia , Naltrexona/uso terapêutico , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
16.
Respir Physiol Neurobiol ; 172(3): 94-105, 2010 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-20444435

RESUMO

Hypothetic mechanisms for respirogenic methylxanthine actions include blockade of adenosine receptors or phosphodiesterase-4 (PDE4) in inspiratory pre-Bötzinger complex (preBötC) networks. Here, we studied this by analyzing stimulating caffeine and theophylline actions on mu-opioid-depressed inspiratory-related rhythm in the ventrolateral aspect of rat brainstem slices. The methylxanthines restored DAMGO (0.5-1 microM) depressed rhythm only at >1mM, which is approximately 10-fold higher than selective for adenosine receptors. Adenosine receptor blockers did neither counter DAMGO inhibition nor change control rhythm, similar to adenosine (0.1-2.5 mM). The specific PDE4 blocker rolipram (5 microM) restored DAMGO-depressed rhythm incompletely, but effectively reversed similar inhibition by clinical mu-agonist (fentanyl, 0.1 microM). At 0.25 microM, rolipram boosted incomplete recovery by 1 mM theophylline of DAMGO-depressed rhythm. Findings indicate that methylxanthines excite rhythmogenic preBötC networks via blockade of cAMP dependent PDE4 and suggest that specific PDE4 inhibitors (plus low methylxanthine doses) stimulate breathing effectively. We discuss why methylxanthine doses for preBötC stimulation need to be higher than those for respirogenic effects in vivo.


Assuntos
Analgésicos Opioides/antagonistas & inibidores , Analgésicos Opioides/farmacologia , Inibidores da Fosfodiesterase 4 , Inibidores de Fosfodiesterase/farmacologia , Insuficiência Respiratória/induzido quimicamente , Insuficiência Respiratória/prevenção & controle , Xantinas/farmacologia , Antagonistas do Receptor A1 de Adenosina , Antagonistas do Receptor A2 de Adenosina , Animais , Animais Recém-Nascidos , Cafeína/farmacologia , Eletrofisiologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/antagonistas & inibidores , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Fentanila/antagonistas & inibidores , Fentanila/farmacologia , Técnicas In Vitro , Interneurônios/efeitos dos fármacos , Rede Nervosa/efeitos dos fármacos , Rede Nervosa/fisiologia , Antagonistas de Receptores Purinérgicos P1 , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Receptores Opioides mu/efeitos dos fármacos , Fenômenos Fisiológicos Respiratórios/efeitos dos fármacos , Rolipram/farmacologia , Teofilina/farmacologia
17.
Eur J Pharmacol ; 638(1-3): 72-7, 2010 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-20435034

RESUMO

Opioids provide effective analgesia in adult patients with painful inflammatory diseases. The proposed mechanism of action is the activation of peripheral opioid receptors, which may be up-regulated in such conditions. Here, by using a chronic inflammation model, namely subplantar injection of Complete Freund's adjuvant, we show a peripheral synergistic interaction between the histamine H(3) receptor agonist R-(alpha)-methylhistamine and fentanyl on the inhibition of thermal hyperalgesia and of peripheral substance P accumulation. Firstly, dose-related effects obtained for the subplantar antinociceptive effect of fentanyl (0.05-1 microg) in the presence of a fixed dose of R-(alpha)-methylhistamine (12.5 microg) showed a shift to the left when compared to that obtained with fentanyl alone. In a similar way, the subcutaneous administration of fentanyl (0.005-0.1mg/kg) plus a fixed dose of R-(alpha)-methylhistamine (0.5mg/kg) induced a supra additive effect on the inhibition of substance P accumulation in the hind-paw skin of inflamed mice. Interestingly, when a neurokinin-1 receptor antagonist was co-administered, the antinociceptive effects of the combined treatment were potentiated. The peripheral adjuvant effect of R-(alpha)-methylhistamine on fentanyl antinociception and inhibition of substance P accumulation was also demonstrated by means of opioid and histamine H(3) receptors selective antagonists: first, naloxone blockade of fentanyl-mediated effects were partially reversed by co-administration of R-(alpha)-methylhistamine, and second, thioperamide partially antagonised the combined R-(alpha)-methylhistamine/fentanyl effects. Overall, our results clearly show that R-(alpha)-methylhistamine enhances fentanyl effects at peripheral sites, and that the control of substance P levels might be one of the mechanisms responsible of such interaction.


Assuntos
Analgésicos/farmacologia , Fentanila/farmacologia , Agonistas dos Receptores Histamínicos/farmacologia , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Metilistaminas/farmacologia , Pele/efeitos dos fármacos , Substância P/metabolismo , Animais , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Quimioterapia Combinada , Fentanila/administração & dosagem , Fentanila/antagonistas & inibidores , Adjuvante de Freund , Agonistas dos Receptores Histamínicos/administração & dosagem , Hiperalgesia/tratamento farmacológico , Masculino , Metilistaminas/administração & dosagem , Metilistaminas/antagonistas & inibidores , Camundongos , Naloxona/farmacologia , Dor/tratamento farmacológico , Piperidinas/farmacologia , Receptores Histamínicos H3/efeitos dos fármacos , Pele/metabolismo
18.
Can J Vet Res ; 74(4): 286-98, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21197229

RESUMO

The objective of this study was to investigate the short-term cardiovascular effects of intravenous (IV) medetomidine-midazolam-fentanyl (MMF) injections in the rabbit using vascular ultrasonography and echocardiography.Anesthesia with MMF was induced intramuscularly (IM) in 8 female New Zealand White rabbits before 3 defined bolus injections of MMF were given IV. Before and for 10 min after each MMF injection the following vascular variables [at the left common carotid artery (ACC) after the first injection and at the abdominal aorta (AA) after the second injection]: vessel diameter (D), peak systolic, minimum diastolic, end-diastolic and average blood flow velocities (psBFV, mdBFV, edBFV, Vave), average volumetric flow (VFave), resistance index (RI) and pulsatility index (PI) and other clinical variables: mean arterial pressure (MAP), heart rate (HR), peripheral arterial oxygen saturation and end-tidal CO2 were recorded. Echocardiography was used after the third injection to investigate changes in cardiac parameters. Additionally, hemodynamic effects were observed at the ACC after complete subcutaneous antagonism of anesthesia by atipamezole-flumazenil-naloxone (AFN) until recovery of the animals.Medetomidine-midazolam-fentanyl IV caused a significant decrease of blood flow velocity in both investigated vessels which was associated with a significant decrease of HR and cardiac performance indicated by the decrease of FS and average volumetric blood flow. Mean arterial pressure significantly increased after each MMF injection; whereas, it significantly decreased after AFN injection. Therefore, MMF and AFN should be carefully used in rabbits and may not be suitable in patients with ventricular dysfunction.


Assuntos
Anestésicos Intravenosos/farmacologia , Sistema Cardiovascular/efeitos dos fármacos , Fentanila/farmacologia , Medetomidina/farmacologia , Midazolam/farmacologia , Coelhos/fisiologia , Animais , Aorta Abdominal/diagnóstico por imagem , Aorta Abdominal/fisiologia , Velocidade do Fluxo Sanguíneo/fisiologia , Velocidade do Fluxo Sanguíneo/veterinária , Pressão Sanguínea/fisiologia , Sistema Cardiovascular/diagnóstico por imagem , Artéria Carótida Primitiva/diagnóstico por imagem , Artéria Carótida Primitiva/fisiologia , Ecocardiografia/veterinária , Feminino , Fentanila/antagonistas & inibidores , Frequência Cardíaca/fisiologia , Modelos Lineares , Medetomidina/antagonistas & inibidores , Midazolam/antagonistas & inibidores
19.
Toxicol Lett ; 191(2-3): 327-40, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19819313

RESUMO

Opioids are known to induce respiratory depression. We aimed to characterize in rats the effects of four opioids on arterial blood gases and plethysmography after intraperitoneal administration at 80% of their LD(50) in order to identify opioid molecule-specific patterns and classify response severity. Opioid-receptor (OR) antagonists, including intravenous 10 mg kg(-1)-naloxonazine at 5 min [mu-OR antagonist], subcutaneous 30 mg kg(-1)-naloxonazine at 24 h [mu1-OR antagonist], subcutaneous 3 mg kg(-1)-naltrindole at 45 min [delta-OR antagonist], and subcutaneous 5 mg kg(-1)-Nor-binaltorphimine at 6 h [kappa-OR antagonist] were pre-administered to test the role of each OR. Methadone, morphine, and fentanyl significantly decreased PaO(2) (P<0.001) and increased PaCO(2) (P<0.05), while buprenorphine only decreased PaO(2) (P<0.05). While all opioids significantly increased inspiratory time (T(I), P<0.001), methadone and fentanyl also increased expiratory time (T(E), P<0.05). Intravenous 10 mg kg(-1)-naloxonazine at 5 min completely reversed opioid-related effects on PaO(2) (P<0.05), PaCO(2) (P<0.001), T(I) (P<0.05), and T(E) (P<0.01) except in buprenorphine. Subcutaneous 30 mg kg(-1)-naloxonazine at 24 h completely reversed effects on PaCO(2) (P<0.01) and T(E) (P<0.001), partially reversed effects on T(I) (P<0.001), and did not reverse effects on PaO(2). Naltrindole reversed methadone-induced T(E) increases (P<0.01) but worsened fentanyl's effect on PaCO(2) (P<0.05) and T(I) (P<0.05). Nor-binaltorphimine reversed morphine- and buprenorphine-induced T(I) increases (P<0.001) but worsened methadone's effect on PaO(2) (P<0.05) and morphine (P<0.001) and buprenorphine's (P<0.01) effects on pH. In conclusion, opioid-related respiratory patterns are not uniform. Opioid-induced hypoxemia as well as increases in T(I) and T(E) are caused by mu-OR, while delta and kappa-OR roles appear limited, depending on the specific opioid. Regarding severity of opioid-induced respiratory effects at 80% of their LD(50), all drugs increased T(I). Methadone and fentanyl induced hypoxemia, hypercapnia, and T(E) increases, morphine caused both hypoxemia and hypercapnia while buprenorphine caused only hypoxemia.


Assuntos
Entorpecentes/toxicidade , Insuficiência Respiratória/induzido quimicamente , Animais , Gasometria , Buprenorfina/antagonistas & inibidores , Buprenorfina/toxicidade , Cateterismo , Fentanila/antagonistas & inibidores , Fentanila/toxicidade , Ácido Láctico/metabolismo , Masculino , Metadona/antagonistas & inibidores , Metadona/toxicidade , Morfina/antagonistas & inibidores , Morfina/toxicidade , Derivados da Morfina/toxicidade , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Pletismografia Total , Ratos , Ratos Sprague-Dawley , Receptores Opioides/efeitos dos fármacos , Insuficiência Respiratória/fisiopatologia , Mecânica Respiratória/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...