Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Basic Clin Pharmacol Toxicol ; 132(5): 449-453, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36808477

RESUMO

Due to a narrow therapeutic index, prolonged lithium treatment and overdose may result in neurotoxicity. Neurotoxicity is deemed reversible with lithium clearance. However, echoing the report of syndrome of irreversible lithium-effectuated neurotoxicity (SILENT) in rare severe poisonings, lithium-induced histopathological brain injuries including extensive neuronal vacuolization, spongiosis and ageing-like neurodegenerative changes were described in the rat following acute toxic and pharmacological exposure. We aimed to investigate the histopathological consequences of lithium exposure in rat models mimicking prolonged treatment and all three patterns of acute, acute-on-chronic and chronic poisonings observed in humans. We performed histopathology and immunostaining-based analyses using optic microscopy of brains obtained from male Sprague-Dawley rats randomly assigned to lithium or saline (controls) and treated according to the therapeutic or to the three poisoning models. No lesion was observed in any brain structure in any of the models. Neuron and astrocyte counts did not differ significantly between lithium-treated rats and controls. Our findings support that lithium-induced neurotoxicity is reversible and brain injury not a common feature of toxicity.


Assuntos
Lesões Encefálicas , Síndromes Neurotóxicas , Intoxicação , Humanos , Masculino , Ratos , Animais , Lítio , Ratos Sprague-Dawley , Síndromes Neurotóxicas/etiologia , Encéfalo , Lesões Encefálicas/induzido quimicamente
2.
Pharmaceuticals (Basel) ; 15(10)2022 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-36297366

RESUMO

Tramadol overdose is frequently associated with the onset of seizures, usually considered as serotonin syndrome manifestations. Recently, the serotoninergic mechanism of tramadol-attributed seizures has been questioned. This study's aim was to identify the mechanisms involved in tramadol-induced seizures in overdose in rats. The investigations included (1) the effects of specific pretreatments on tramadol-induced seizure onset and brain monoamine concentrations, (2) the interaction between tramadol and γ-aminobutyric acid (GABA)A receptors in vivo in the brain using positron emission tomography (PET) imaging and 11C-flumazenil. Diazepam abolished tramadol-induced seizures, in contrast to naloxone, cyproheptadine and fexofenadine pretreatments. Despite seizure abolishment, diazepam significantly enhanced tramadol-induced increase in the brain serotonin (p < 0.01), histamine (p < 0.01), dopamine (p < 0.05) and norepinephrine (p < 0.05). No displacement of 11C-flumazenil brain kinetics was observed following tramadol administration in contrast to diazepam, suggesting that the observed interaction was not related to a competitive mechanism between tramadol and flumazenil at the benzodiazepine-binding site. Our findings do not support the involvement of serotoninergic, histaminergic, dopaminergic, norepinephrine or opioidergic pathways in tramadol-induced seizures in overdose, but they strongly suggest a tramadol-induced allosteric change of the benzodiazepine-binding site of GABAA receptors. Management of tramadol-poisoned patients should take into account that tramadol-induced seizures are mainly related to a GABAergic pathway.

3.
Br J Anaesth ; 128(3): 584-595, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34872716

RESUMO

BACKGROUND: The safety profile of buprenorphine has encouraged its widespread use. However, fatalities have been attributed to benzodiazepine/buprenorphine combinations, by poorly understood mechanisms of toxicity. Mechanistic hypotheses include (i) benzodiazepine-mediated increase in brain buprenorphine (pharmacokinetic hypothesis); (ii) benzodiazepine-mediated potentiation of buprenorphine interaction with opioid receptors (receptor hypothesis); and (iii) combined effects of buprenorphine and benzodiazepine on respiratory parameters (pharmacodynamic hypothesis). METHODS: We studied the neuro-respiratory effects of buprenorphine (30 mg kg-1, i.p.), diazepam (20 mg kg-1, s.c.), and diazepam/buprenorphine combination in rats using arterial blood gas analysis, plethysmography, and diaphragm electromyography. Pretreatments with various opioid and gamma-aminobutyric acid receptor antagonists were tested. Diazepam impact on brain 11C-buprenorphine kinetics and binding to opioid receptors was studied using positron emission tomography imaging. RESULTS: In contrast to diazepam and buprenorphine alone, diazepam/buprenorphine induced early-onset sedation (P<0.05) and respiratory depression (P<0.001). Diazepam did not alter 11C-buprenorphine brain kinetics or binding to opioid receptors. Diazepam/buprenorphine-induced effects on inspiratory time were additive, driven by buprenorphine (P<0.0001) and were blocked by naloxonazine (P<0.01). Diazepam/buprenorphine-induced effects on expiratory time were non-additive (P<0.001), different from buprenorphine-induced effects (P<0.05) and were blocked by flumazenil (P<0.01). Diazepam/buprenorphine-induced effects on tidal volume were non-additive (P<0.01), different from diazepam- (P<0.05) and buprenorphine-induced effects (P<0.0001) and were blocked by naloxonazine (P<0.05) and flumazenil (P<0.05). Compared with buprenorphine, diazepam/buprenorphine decreased diaphragm contraction amplitude (P<0.01). CONCLUSIONS: Pharmacodynamic parameters and antagonist pretreatments indicate that diazepam/buprenorphine-induced respiratory depression results from a pharmacodynamic interaction between both drugs on ventilatory parameters.


Assuntos
Buprenorfina , Diazepam , Insuficiência Respiratória , Animais , Masculino , Ratos , Analgésicos Opioides/farmacocinética , Benzodiazepinas/farmacocinética , Gasometria/métodos , Buprenorfina/efeitos adversos , Buprenorfina/farmacocinética , Diazepam/efeitos adversos , Diazepam/farmacocinética , Interações Medicamentosas/fisiologia , Flumazenil/farmacocinética , Antagonistas de Entorpecentes/farmacocinética , Ratos Sprague-Dawley , Receptores Opioides/metabolismo , Insuficiência Respiratória/induzido quimicamente , Insuficiência Respiratória/metabolismo
4.
Toxicol Sci ; 164(1): 153-165, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29945230

RESUMO

Baclofen, a γ-amino-butyric acid type-B receptor agonist with exponentially increased use at high-dose to facilitate abstinence in chronic alcoholics, is responsible for increasing poisonings. Tolerance and withdrawal syndromes have been reported during prolonged treatment but their contribution to the variability of baclofen-induced neurotoxicity in overdose is unknown. We studied baclofen-induced effects on rat sedation, temperature, and ventilation and modeled baclofen pharmacokinetics and effect/concentration relationships aiming to investigate the consequences of repeated baclofen pretreatment and to characterize withdrawal syndrome. Baclofen-induced dose-dependent sedation (p <0.01), hypothermia (p <.001) and respiratory depression (p <.01) were altered in repeatedly baclofen-pretreated rats (p <.05). Repeatedly baclofen-pretreated rats did not exhibit respiratory depression following baclofen overdose due to limitations on baclofen-induced increase in inspiratory (p <.01) and expiratory times (p <.01). Only slight hypoxemia without respiratory acidosis was observed. Baclofen discontinuation resulted in hyperlocomotion and non-anxiogenic withdrawal symptoms. Regarding pharmacokinetics, repeated baclofen pretreatment increased the peak concentration (p <.05) and absorption constant rate (p <.05) and reduced the distribution volume (p <.0001) and elimination half-life (p <.05). Analysis of the effect/concentration relationships indicated that plasma baclofen concentration decreases more rapidly than all studied neuro-respiratory effects, in tolerant and non-tolerant rats. Taken together, our findings supported the role of brain distribution in baclofen-induced neurotoxicity expression and its probable involvement in tolerance-related attenuation in addition to physiological adaptations of ventilation. In conclusion, repeated pretreatment attenuates baclofen-attributed neurotoxicity in overdose and results in post-discontinuation withdrawal syndrome. Our findings suggest both pharmacodynamic and pharmacokinetic mechanisms whose relative contributions to the variability of baclofen-induced neurotoxicity in overdose remain to be established.


Assuntos
Baclofeno/toxicidade , Tolerância a Medicamentos , Síndromes Neurotóxicas/etiologia , Insuficiência Respiratória/induzido quimicamente , Síndrome de Abstinência a Substâncias , Animais , Baclofeno/administração & dosagem , Baclofeno/farmacocinética , Encéfalo/metabolismo , Relação Dose-Resposta a Droga , Masculino , Ratos Sprague-Dawley , Distribuição Tecidual
5.
Artigo em Inglês | MEDLINE | ID: mdl-29782961

RESUMO

Baclofen, a γ-amino-butyric acid type-B receptor agonist with exponentially increased use at high-dose to facilitate abstinence in chronic alcoholics, is responsible for increasing poisonings. Baclofen overdose may induce severe encephalopathy and electroencephalographic (EEG) abnormalities. Whether prior prolonged baclofen treatment may influence the severity of baclofen-induced encephalopathy in overdose has not been established. We designed a rat study to characterize baclofen-induced encephalopathy, correlate its severity with plasma concentrations and investigate the contribution of tolerance. Baclofen-induced encephalopathy was assessed using continuous EEG and scored based on a ten-grade scale. Following the administration by gavage of 116 mg/kg baclofen, EEG rapidly and steadily impaired resulting in the successive onset of deepening sleep followed by generalized periodic epileptiform discharges and burst-suppressions. Thereafter, encephalopathy progressively recovered following similar phases in reverse. Periodic triphasic sharp waves, non-convulsive status epilepticus and even isoelectric signals were observed at the most critical stages. Prior repeated baclofen administration resulted in reduced severity (peak: grade 7 versus 9; peak effect length: 382 ±â€¯40 versus 123 ±â€¯14 min, P = 0.008) and duration of encephalopathy (18 versus > 24 h, P = 0.0007), supporting the acquisition of tolerance. The relationship between encephalopathy severity and plasma baclofen concentrations fitted a sigmoidal Emax model with an anticlockwise hysteresis loop suggesting a hypothetical biophase site of action. The baclofen concentration producing a response equivalent to 50% of Emax was significantly reduced (8947 µg/L, ±11.3% versus 12,728 µg/L, ±24.0% [mean, coefficient of variation], P = 0.03) with prior prolonged baclofen administration. In conclusion, baclofen overdose induces early-onset and prolonged marked encephalopathy that is significantly attenuated by prior repeated baclofen treatment. Our findings suggest a possible role for the blood-brain barrier in the development of tolerance; however, its definitive involvement remains to be demonstrated.


Assuntos
Baclofeno/efeitos adversos , Encefalopatias/induzido quimicamente , Encefalopatias/fisiopatologia , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Overdose de Drogas/fisiopatologia , Animais , Baclofeno/sangue , Relação Dose-Resposta a Droga , Tolerância a Medicamentos/fisiologia , Eletroencefalografia , Agonistas dos Receptores de GABA-B/efeitos adversos , Agonistas dos Receptores de GABA-B/sangue , Masculino , Modelos Biológicos , Distribuição Aleatória , Ratos Sprague-Dawley , Sono/efeitos dos fármacos , Sono/fisiologia
6.
Clin Toxicol (Phila) ; 56(8): 737-743, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29148295

RESUMO

CONTEXT: Since the banning of dextropropoxyphene from the market, overdoses, and fatalities attributed to tramadol, a WHO step-2 opioid analgesic, have increased markedly. Tramadol overdose results not only in central nervous system (CNS) depression attributed to its opioid properties but also in seizures, possibly related to non-opioidergic pathways, thus questioning the efficiency of naloxone to reverse tramadol-induced CNS toxicity. OBJECTIVE: To investigate the most efficient antidote to reverse tramadol-induced seizures and respiratory depression in overdose. MATERIALS AND METHODS: Sprague-Dawley rats overdosed with 75 mg/kg intraperitoneal (IP) tramadol were randomized into four groups to receive solvent (control group), diazepam (1.77 mg/kg IP), naloxone (2 mg/kg intravenous bolus followed by 4 mg/kg/h infusion), and diazepam/naloxone combination. Sedation depth, temperature, number of seizures, and intensity, whole-body plethysmography parameters and electroencephalography activity were measured. RESULTS: Naloxone reversed tramadol-induced respiratory depression (p < .05) but significantly increased seizures (p < .01) and prolonged their occurrence time. Diazepam abolished seizures but significantly deepened rat sedation (p < .05) without improving ventilation. Diazepam/naloxone combination completely abolished seizures, significantly improved rat ventilation by reducing inspiratory time (p < .05) but did not worsen sedation. None of these treatments significantly modified rat temperature. CONCLUSIONS: Diazepam/naloxone combination is the most efficient antidote to reverse tramadol-induced CNS toxicity in the rat.


Assuntos
Analgésicos Opioides/intoxicação , Antídotos/uso terapêutico , Overdose de Drogas/tratamento farmacológico , Naloxona/uso terapêutico , Insuficiência Respiratória/induzido quimicamente , Convulsões/induzido quimicamente , Tramadol/intoxicação , Animais , Ratos , Ratos Sprague-Dawley
7.
NMR Biomed ; 30(11)2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28703506

RESUMO

Lithium is the first-line mood stabilizer for the treatment of patients with bipolar disorder. However, its mechanisms of action and transport across the blood-brain barrier remain poorly understood. The contribution of lithium-7 magnetic resonance imaging (7 Li MRI) to investigate brain lithium distribution remains limited because of the modest sensitivity of the lithium nucleus and the expected low brain concentrations in humans and animal models. Therefore, we decided to image lithium distribution in the rat brain ex vivo using a turbo-spin-echo imaging sequence at 17.2 T. The estimation of lithium concentrations was performed using a phantom replacement approach accounting for B1 inhomogeneities and differential T1 and T2 weighting. Our MRI-derived lithium concentrations were validated by comparison with inductively coupled plasma-mass spectrometry (ICP-MS) measurements ([Li]MRI  = 1.18[Li]MS , R = 0.95). Overall, a sensitivity of 0.03 mmol/L was achieved for a spatial resolution of 16 µL. Lithium distribution was uneven throughout the brain (normalized lithium content ranged from 0.4 to 1.4) and was mostly symmetrical, with consistently lower concentrations in the metencephalon (cerebellum and brainstem) and higher concentrations in the cortex. Interestingly, low lithium concentrations were also observed close to the lateral ventricles. The average brain-to-plasma lithium ratio was 0.34 ± 0.04, ranging from 0.29 to 0.39. Brain lithium concentrations were reasonably correlated with plasma lithium concentrations, with Pearson correlation factors ranging from 0.63 to 0.90.


Assuntos
Encéfalo/metabolismo , Lítio/farmacocinética , Espectroscopia de Ressonância Magnética/métodos , Animais , Imageamento por Ressonância Magnética , Masculino , Ratos , Ratos Sprague-Dawley
8.
Bipolar Disord ; 19(2): 135-145, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28425670

RESUMO

OBJECTIVES: Lithium overdose may result in encephalopathy and electroencephalographic abnormalities. Three poisoning patterns have been identified based on the ingested dose, previous treatment duration and renal function. Whether the severity of lithium-induced encephalopathy depends on the poisoning pattern has not been established. We designed a rat study to investigate lithium-induced encephalopathy and correlate its severity to plasma, erythrocyte, cerebrospinal fluid and brain lithium concentrations previously determined in rat models mimicking human poisoning patterns. METHODS: Lithium-induced encephalopathy was assessed and scored using continuous electroencephalography. RESULTS: We demonstrated that lithium overdose was consistently responsible for encephalopathy, the severity of which depended on the poisoning pattern. Acutely poisoned rats developed rapid-onset encephalopathy which reached a maximal grade of 2/5 at 6 h and disappeared at 24 h post-injection. Acute-on-chronically poisoned rats developed persistent and slightly fluctuating encephalopathy which reached a maximal grade of 3/5. Chronically poisoned rats developed rapid-onset but gradually increasing life-threatening encephalopathy which reached a maximal grade of 4/5. None of the acutely, 20% of the acute-on-chronically and 57% of the chronically lithium-poisoned rats developed seizures. The relationships between encephalopathy severity and lithium concentrations fitted a sigmoidal Emax model based on cerebrospinal fluid concentrations in acute poisoning and brain concentrations in acute-on-chronic poisoning. In chronic poisoning, worsening of encephalopathy paralleled the increase in plasma lithium concentrations. CONCLUSIONS: The severity of lithium-induced encephalopathy is dependent on the poisoning pattern, which was previously shown to determine lithium accumulation in the brain. Our data support the proposition that electroencephalography is a sensitive tool for scoring lithium-related neurotoxicity.


Assuntos
Transtorno Bipolar/tratamento farmacológico , Eletroencefalografia/métodos , Compostos de Lítio , Lítio , Síndromes Neurotóxicas , Animais , Antimaníacos/farmacologia , Antimaníacos/toxicidade , Encéfalo/efeitos dos fármacos , Relação Dose-Resposta a Droga , Lítio/sangue , Lítio/farmacocinética , Compostos de Lítio/farmacologia , Compostos de Lítio/toxicidade , Síndromes Neurotóxicas/diagnóstico , Síndromes Neurotóxicas/etiologia , Ratos , Distribuição Tecidual
9.
Artigo em Inglês | MEDLINE | ID: mdl-28336491

RESUMO

Severity of lithium poisoning depends on the ingested dose, previous treatment duration and renal function. No animal study has investigated neurobehavioral differences in relation to the lithium poisoning pattern observed in humans, while differences in lithium pharmacokinetics have been reported in lithium-pretreated rats mimicking chronic poisonings with enhanced brain accumulation in rats with renal failure. Our objectives were: 1)-to investigate lithium-related effects in overdose on locomotor activity, anxiety-like behavior, spatial recognition memory and anhedonia in the rat; 2)-to model the relationships between lithium-induced effects on locomotion and plasma, erythrocyte, cerebrospinal fluid and brain concentrations previously obtained according to the poisoning pattern. Open-field, elevated plus-maze, Y-maze and sucrose consumption tests were used. In acutely lithium-poisoned rats, we observed horizontal (p<0.001) and vertical hypolocomotion (p<0.0001), increased anxiety-like behavior (p<0.05) and impaired memory (p<0.01) but no altered hedonic status. Horizontal (p<0.01) and vertical (p<0.001) hypolocomotion peaked more markedly 24h after lithium injection and was more prolonged in acute-on-chronically vs. acutely lithium-poisoned rats. Hypolocomotion in chronically lithium-poisoned rats with impaired renal function did not differ from acutely poisoned rats 24h after the last injection. Interestingly, hypolocomotion/concentration relationships best fitted a sigmoidal Emax model in acute poisoning and a linear regression model linked to brain lithium in acute-on-chronic poisoning. In conclusion, lithium overdose alters rat behavior and consistently induces hypolocomotion which is more marked and prolonged in repeatedly lithium-treated rats. Our data suggest that differences between poisoning patterns regarding lithium-induced hypolocomotion are better explained by the duration of lithium exposure than by its brain accumulation.


Assuntos
Anedonia/efeitos dos fármacos , Antidepressivos/intoxicação , Comportamento Animal/efeitos dos fármacos , Compostos de Lítio/intoxicação , Atividade Motora/efeitos dos fármacos , Memória Espacial/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Masculino , Ratos , Ratos Sprague-Dawley
10.
Pain ; 158(3): 505-515, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28135212

RESUMO

Drugs able to treat both nociceptive and neuropathic pain effectively without major side effects are lacking. We developed a bifunctional peptide-based hybrid (KGNOP1) that structurally combines a mu-opioid receptor agonist (KGOP1) with antinociceptive activity and a weak nociceptin receptor antagonist (KGNOP3) with anti-neuropathic pain activity. We investigated KGNOP1-related behavioral effects after intravenous administration in rats by assessing thermal nociception, cold hyperalgesia in a model of neuropathic pain induced by chronic constriction injury of the sciatic nerve, and plethysmography parameters including inspiratory time (TI) and minute ventilation (VM) in comparison to the well-known opioid analgesics, tramadol and morphine. Time-course and dose-dependent effects were investigated for all behavioral parameters to determine the effective doses 50% (ED50). Pain-related effects on cold hyperalgesia were markedly increased by KGNOP1 as compared to KGNOP3 and tramadol (ED50: 0.0004, 0.32, and 12.1 µmol/kg, respectively), whereas effects on thermal nociception were significantly higher with KGNOP1 as compared to morphine (ED50: 0.41 and 14.7 µmol/kg, respectively). KGNOP1 and KGOP1 produced a larger increase in TI and deleterious decrease in VM in comparison to morphine and tramadol (ED50(TI): 0.63, 0.52, 12.2, and 50.9 µmol/kg; ED50(VM): 0.57, 0.66, 10.6, and 50.0 µmol/kg, respectively). Interestingly, the calculated ratios of anti-neuropathic pain/antinociceptive to respiratory effects revealed that KGNOP1 was safer than tramadol (ED50 ratio: 5.44 × 10 vs 0.24) and morphine (ED50 ratio: 0.72 vs 1.39). We conclude that KGNOP1 is able to treat both experimental neuropathic and nociceptive pain, more efficiently and safely than tramadol and morphine, respectively, and thus should be a candidate for future clinical developments.


Assuntos
Analgésicos Opioides/uso terapêutico , Antagonistas de Entorpecentes/uso terapêutico , Neuralgia/tratamento farmacológico , Dor Nociceptiva/tratamento farmacológico , Peptídeos Opioides/antagonistas & inibidores , Peptídeos/uso terapêutico , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Hiperalgesia/tratamento farmacológico , Indóis/uso terapêutico , Ligantes , Masculino , Oligopeptídeos/química , Oligopeptídeos/uso terapêutico , Medição da Dor , Fenalenos/uso terapêutico , Pletismografia , Ratos , Ratos Sprague-Dawley , Receptores Opioides , Respiração/efeitos dos fármacos , Fatores de Tempo , Tramadol/uso terapêutico , Nociceptina
11.
Toxicol Sci ; 155(2): 389-399, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27803387

RESUMO

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


Assuntos
Analgésicos Opioides/toxicidade , Buprenorfina/toxicidade , Etanol/toxicidade , Antagonistas de Entorpecentes/toxicidade , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/farmacocinética , Animais , Buprenorfina/administração & dosagem , Buprenorfina/análogos & derivados , Buprenorfina/metabolismo , Buprenorfina/farmacocinética , Interações Medicamentosas , Etanol/administração & dosagem , Etanol/farmacocinética , Cromatografia Gasosa-Espectrometria de Massas , Masculino , Antagonistas de Entorpecentes/administração & dosagem , Antagonistas de Entorpecentes/farmacocinética , Ratos , Ratos Sprague-Dawley , Respiração/efeitos dos fármacos , Insuficiência Respiratória/induzido quimicamente
12.
Toxicol Appl Pharmacol ; 310: 108-119, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27641627

RESUMO

Poisoning with opioid analgesics including tramadol represents a challenge. Tramadol may induce respiratory depression, seizures and serotonin syndrome, possibly worsened when in combination to benzodiazepines. Our objectives were to investigate tramadol-related neurotoxicity, consequences of diazepam/tramadol combination, and mechanisms of drug-drug interactions in rats. Median lethal-doses were determined using Dixon-Bruce's up-and-down method. Sedation, seizures, electroencephalography and plethysmography parameters were studied. Concentrations of tramadol and its metabolites were measured using liquid-chromatography-high-resolution-mass-spectrometry. Plasma, platelet and brain monoamines were measured using liquid-chromatography coupled to fluorimetry. Median lethal-doses of tramadol and diazepam/tramadol combination did not significantly differ, although time-to-death was longer with combination (P=0.04). Tramadol induced dose-dependent sedation (P<0.05), early-onset seizures (P<0.001) and increase in inspiratory (P<0.01) and expiratory times (P<0.05). The diazepam/tramadol combination abolished seizures but significantly enhanced sedation (P<0.01) and respiratory depression (P<0.05) by reducing tidal volume (P<0.05) in addition to tramadol-related increase in respiratory times, suggesting a pharmacodynamic mechanism of interaction. Plasma M1 and M5 metabolites were mildly increased, contributing additionally to tramadol-related respiratory depression. Tramadol-induced early-onset increase in brain concentrations of serotonin and norepinephrine was not significantly altered by the diazepam/tramadol combination. Interestingly neither pretreatment with cyproheptadine (a serotonin-receptor antagonist) nor a benserazide/5-hydroxytryptophane combination (enhancing brain serotonin) reduced tramadol-induced seizures. Our study shows that diazepam/tramadol combination does not worsen tramadol-induced fatality risk but alters its toxicity pattern with enhanced respiratory depression but abolished seizures. Drug-drug interaction is mainly pharmacodynamic but increased plasma M1 and M5 metabolites may also contribute to enhancing respiratory depression. Tramadol-induced seizures are independent of brain serotonin.


Assuntos
Analgésicos Opioides/toxicidade , Diazepam/toxicidade , Overdose de Drogas , Sistema Nervoso/efeitos dos fármacos , Tramadol/toxicidade , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/farmacocinética , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Diazepam/administração & dosagem , Relação Dose-Resposta a Droga , Masculino , Norepinefrina/metabolismo , Ratos , Ratos Sprague-Dawley , Serotonina/metabolismo , Tramadol/administração & dosagem , Tramadol/farmacocinética
13.
Toxicol Sci ; 143(1): 185-95, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25354763

RESUMO

Lithium-induced neurotoxicity may be life threatening. Three patterns have been described, including acute, acute-on-chronic, and chronic poisoning, with unexplained discrepancies in the relationship between clinical features and plasma lithium concentrations. Our objective was to investigate differences in plasma, erythrocyte, cerebrospinal fluid, and brain lithium pharmacokinetics using a multicompartmental approach in rat models mimicking the three human intoxication patterns. We developed acute (intraperitoneal administration of 185 mg/kg Li2CO3 in naive rats), acute-on-chronic (intraperitoneal administration of 185 mg/kg Li2CO3 in rats receiving 800 mg/l Li2CO3 in water during 28 days), and chronic poisoning models (intraperitoneal administration of 74 mg/kg Li2CO3 during 5 days in rats with 15 mg/kg K2Cr2O7-induced renal failure). Delayed absorption (4.03 vs 0.31 h), increased plasma elimination (0.65 vs 0.37 l/kg/h) and shorter half-life (1.75 vs 2.68 h) were observed in acute-on-chronically compared with acutely poisoned rats. Erythrocyte and cerebrospinal fluid kinetics paralleled plasma kinetics in both models. Brain lithium distribution was rapid (as early as 15 min), inhomogeneous and with delayed elimination (over 78 h). However, brain lithium accumulation was more marked in acute-on-chronically than acutely poisoned rats [area-under-the-curve of brain concentrations (379 ± 41 vs 295 ± 26, P < .05) and brain-to-plasma ratio (45 ± 10 vs 8 ± 2, P < .0001) at 54 h]. Moreover, brain lithium distribution was increased in chronically compared with acute-on-chronically poisoned rats (brain-to-plasma ratio: 9 ± 1 vs 3 ± 0, P < .01). In conclusion, prolonged rat exposure results in brain lithium accumulation, which is more marked in the presence of renal failure. Our data suggest that differences in plasma and brain kinetics may at least partially explain the observed variability between human intoxication patterns.


Assuntos
Encéfalo/metabolismo , Carbonato de Lítio/farmacocinética , Carbonato de Lítio/toxicidade , Síndromes Neurotóxicas/metabolismo , Intoxicação/metabolismo , Doença Aguda , Animais , Área Sob a Curva , Doença Crônica , Modelos Animais de Doenças , Esquema de Medicação , Eritrócitos/metabolismo , Meia-Vida , Injeções Intraperitoneais , Carbonato de Lítio/administração & dosagem , Carbonato de Lítio/sangue , Carbonato de Lítio/líquido cefalorraquidiano , Masculino , Taxa de Depuração Metabólica , Modelos Biológicos , Síndromes Neurotóxicas/sangue , Síndromes Neurotóxicas/líquido cefalorraquidiano , Síndromes Neurotóxicas/etiologia , Intoxicação/sangue , Intoxicação/líquido cefalorraquidiano , Dicromato de Potássio , Ratos Sprague-Dawley , Insuficiência Renal/induzido quimicamente , Insuficiência Renal/metabolismo
14.
Toxicol Lett ; 228(2): 75-84, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24769261

RESUMO

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


Assuntos
Analgésicos Opioides/toxicidade , Buprenorfina/toxicidade , Diazepam/toxicidade , Hipnóticos e Sedativos/toxicidade , Naloxona/toxicidade , Antagonistas de Entorpecentes/toxicidade , Insuficiência Respiratória/induzido quimicamente , Análise de Variância , Animais , Interações Medicamentosas , Tolerância a Medicamentos , Masculino , Pletismografia Total , Ratos , Ratos Sprague-Dawley
15.
Toxicology ; 305: 99-108, 2013 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-23376546

RESUMO

While most deaths from asphyxia related to buprenorphine (BUP) overdose have been reported in males, higher plasma concentrations of BUP and its toxic metabolite norbuprenorphine (NBUP) have been observed in females. We previously demonstrated that P-glycoprotein (P-gp) modulation at the blood-brain barrier (BBB) contributes highly to BUP-related respiratory toxicity, by limiting NBUP entrance into the brain. In this work, we sought to investigate the role of P-gp-mediated transport at the BBB in gender and strain-related variability of BUP and NBUP-induced respiratory effects in mice. Ventilation was studied using plethysmography, P-gp expression using western blot, and transport at the BBB using in situ cerebral perfusion. In male Fvb and Swiss mice, BUP was responsible for ceiling respiratory effects. NBUP-related reduction in minute volume was dose-dependent but more marked in Fvb (p<0.01 at 1mg/kg NBUP and p<0.001 at 3 and 9mg/kg NBUP) than in Swiss mice (p<0.001 at 9mg/kg NBUP). Female Fvb mice were more susceptible to BUP than males with significantly increased inspiratory time (p<0.05) and to NBUP with significantly increased expiratory time (p<0.01). Following BUP administration, plasma BUP concentrations were significantly higher (p<0.01) and plasma NBUP concentrations significantly lower (p<0.001) in Fvb mice compared to Swiss mice. Plasma BUP concentrations were significantly higher (p<0.05) and plasma NBUP concentrations significantly lower (p<0.01) in male compared to female Fvb mice. In contrast, following NBUP administration, comparable plasma NBUP concentrations were observed in both genders and strains. No differences in P-gp expression or BUP and NBUP transport across the BBB were observed between male and female Fvb mice as well as between Swiss and Fvb mice. Our results suggest that P-gp-mediated transport across the BBB does not play a key-role in gender and strain-related variability in BUP and NBUP-induced respiratory toxicity in mice. Both gender- and strain-related differences in respiratory effects of BUP could be attributed to BUP itself rather than to its metabolite, NBUP.


Assuntos
Buprenorfina/toxicidade , Entorpecentes/toxicidade , Insuficiência Respiratória/induzido quimicamente , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/fisiologia , Animais , Barreira Hematoencefálica/metabolismo , Western Blotting , Capilares/efeitos dos fármacos , Capilares/metabolismo , Circulação Cerebrovascular , Relação Dose-Resposta a Droga , Feminino , Masculino , Camundongos , Pletismografia Total , Insuficiência Respiratória/fisiopatologia , Caracteres Sexuais , Especificidade da Espécie
16.
Toxicol Lett ; 217(3): 251-9, 2013 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-23296104

RESUMO

Morphine is responsible for severe poisonings in chronically treated patients. We hypothesize that toxicity could be related to the development of weaker tolerance for morphine-induced deleterious respiratory effects in comparison to analgesic effects. Our objectives were to compare tolerance to both effects in mice and investigate possible mechanisms for such possible differences. Tolerance to morphine-induced analgesia and respiratory effects was assessed using hot plate response latencies and plethysmography, respectively. Mechanisms of tolerance were investigated using binding studies to mu-opioid receptors (MOR) and adenylate cyclase (AC) activity measurement in homogenates of cell membranes from the periaqueductal gray region (PAG) and brainstem. Morphine (2.5 mg/kg) was responsible for analgesia with significant increase in inspiratory time. Acute tolerance to analgesia (p<0.01) and effects on respiratory frequency (p<0.05) was observed in mice pre-treated with 100 mg/kg morphine in comparison to saline. Following repetitive administration (2.5 mg/kg/day during 10 days), we observed a 13-fold increase in the effective dose-50% (ED50) of morphine-induced analgesia in comparison to a 2- or 4-fold increase in the ED50 of its related increase in inspiratory time determined in air and 4% CO2, respectively. No significant alteration in MOR expression was observed in either PAG or brainstem following repeated morphine administration. However, in PAG, in contrast to brainstem, superactivation of AC was observed in morphine-treated mice in comparison to controls (p<0.05). In conclusion, tolerance to morphine-induced respiratory effects is much more limited than tolerance to its analgesic effects in repeatedly morphine-treated mice. The difference in morphine-induced AC activation between the brainstem and the PAG contributes to the observed difference in tolerance between both morphine effects.


Assuntos
Analgésicos Opioides/toxicidade , Morfina/toxicidade , Respiração/efeitos dos fármacos , Adenilil Ciclases/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Encéfalo/metabolismo , Tolerância a Medicamentos , Masculino , Camundongos , Medição da Dor/métodos , Pletismografia Total , Distribuição Aleatória , Receptores Opioides mu/metabolismo
17.
Drug Alcohol Depend ; 131(3): 298-307, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23332448

RESUMO

BACKGROUND: Methadone may cause respiratory depression and fatalities. Concomitant use of benzodiazepines in methadone-treated patients for chronic pain or as maintenance therapy for opiate abuse is common. However, the exact contribution of benzodiazepines to methadone-induced respiratory toxicity remains debatable. METHODS: We investigated the respiratory effects of the combination diazepam (20mg/kg)/methadone (5mg/kg) in the rat, focusing on methadone concentration/effect relationships. Respiratory effects were studied using arterial blood gases and whole-body plethysmography. Plasma concentrations of both R- and S-methadone enantiomers were measured using high-performance liquid chiral chromatography coupled to mass spectrometry. To clarify mechanisms of diazepam/methadone interaction, methadone metabolism was investigated in vitro using rat liver microsomes. RESULTS: Diazepam/methadone co-administration significantly increased methadone-related effects on inspiratory time (p<0.001) but did not significantly alter the other respiratory parameters when compared with methadone alone, despite significant increase in the area under the curve of plasma R-methadone concentrations measured during 240 min (p<0.05). Diazepam/methadone co-incubation with microsomes in vitro resulted in a significant inhibition of methadone metabolism (p<0.01), with 50%-inhibitory diazepam concentrations of 25.02 ± 0.18 µmol/L and 25.18 ± 0.23 µmol/L for R- and S-methadone, respectively. CONCLUSION: We concluded that co-administration of high-doses of diazepam and methadone in rats is not responsible for additional respiratory depression in comparison to methadone alone, despite significant metabolic interaction between the drugs. In humans, although our experimental data may suggest the relative safety of benzodiazepine/methadone co-prescription, physicians should remain cautious as other underlying conditions may enhance this drug-drug interaction.


Assuntos
Diazepam/administração & dosagem , Metadona/administração & dosagem , Mecânica Respiratória/efeitos dos fármacos , Animais , Diazepam/sangue , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Interações Medicamentosas/fisiologia , Masculino , Metadona/sangue , Pletismografia/métodos , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Mecânica Respiratória/fisiologia
18.
Crit Care Med ; 40(12): 3215-23, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22975888

RESUMO

OBJECTIVES: Deaths due to asphyxia as well as following acute poisoning with severe respiratory depression have been attributed to buprenorphine in opioid abusers. However, in human and animal studies, buprenorphine exhibited ceiling respiratory effects, whereas its metabolite, norbuprenorphine, was assessed as being a potent respiratory depressor in rodents. Recently, norbuprenorphine, in contrast to buprenorphine, was shown in vitro to be a substrate of human P-glycoprotein, a drug-transporter involved in all steps of pharmacokinetics including transport at the blood-brain barrier. Our objectives were to assess P-glycoprotein involvement in norbuprenorphine transport in vivo and study its role in the modulation of buprenorphine-related respiratory effects in mice. SETTING: University-affiliated research laboratory, INSERM U705, Paris, France. SUBJECTS: Wild-type and P-glycoprotein knockout female Friend virus B-type mice. INTERVENTIONS: Respiratory effects were studied using plethysmography and the P-glycoprotein role at the blood-brain barrier using in situ brain perfusion. MEASUREMENTS AND MAIN RESULTS: Norbuprenorphine(≥ 1 mg/kg) and to a lesser extent buprenorphine (≥ 10 mg/kg) were responsible for dose-dependent respiratory depression combining increased inspiratory (TI) and expiratory times (TE). PSC833, a powerful P-glycoprotein inhibitor, significantly enhanced buprenorphine-related effects on TI (p < .01) and TE (p < .05) and norbuprenorphine-related effects on minute volume (VE, p < .05), TI, and TE (p < .001). In P-glycoprotein-knockout mice, buprenorphine-related effects on VE (p < .01), TE (p < .001), and TI (p < .05) and norbuprenorphine-related effects on VE (p < .05) and TI (p < .001) were significantly enhanced. Plasma norbuprenorphine concentrations were significantly increased in PSC833-treated mice (p < .001), supporting a P-glycoprotein role in norbuprenorphine pharmacokinetics. Brain norbuprenorphine efflux was significantly reduced in PSC833-treated and P-glycoprotein-knockout mice (p < .001), supporting P-glycoprotein-mediated norbuprenorphine transport at the blood-brain barrier. CONCLUSIONS: P-glycoprotein plays a key-protective role in buprenorphine-related respiratory effects, by allowing norbuprenorphine efflux at the blood-brain barrier. Our findings suggest a major role for drug-drug interactions that lead to P-glycoprotein inhibition in buprenorphine-associated fatalities and respiratory depression.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/fisiologia , Analgésicos Opioides/toxicidade , Barreira Hematoencefálica , Buprenorfina/análogos & derivados , Buprenorfina/toxicidade , Insuficiência Respiratória/induzido quimicamente , Animais , Transporte Biológico Ativo/efeitos dos fármacos , Buprenorfina/farmacocinética , Interações Medicamentosas , Feminino , França , Camundongos , Camundongos Knockout , Pletismografia
19.
Toxicology ; 302(2-3): 248-54, 2012 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-22951378

RESUMO

Citalopram, a selective serotonin reuptake inhibitor, is generally considered to be of low toxicity. However, serotonin syndrome, seizures, electrocardiographic abnormalities as well as respiratory failure and death have been described in patients with citalopram overdose. The mechanisms of severe toxicity remain unclear. Our objective was to study the mechanisms of death following high-dose citalopram administration in Sprague Dawley rats. The median lethal dose (MLD) of intraperitoneal (i.p.) citalopram was measured using Dixon & Bruce's up-and-down method at 102 mg/kg. Dose-effect relationships of citalopram-induced clinical features, alterations in arterial blood gas and plethysmography, and disturbances in blood lactate, plasma and platelet serotonin concentrations were studied. Seizures were significantly increased in rats receiving 80% and 120% of citalopram MLD versus controls (p<0.05 and p<0.01, respectively). A significant decrease in body temperature was observed after 90 min in rats treated with doses >60% MLD in comparison to controls (p<0.05). The occurrence of serotonin behavioural syndrome was comparable in all groups. Citalopram administration did not result in significant hypoxemia, hypercapnia and lactate elevation. However, a significant moderate increase in the inspiratory time (p<0.05) accompanied with an expiratory braking was observed. A significant dose-related linear decrease in platelet serotonin and increase in plasma serotonin concentrations were measured (p<0.05). Pre-treatments of rats receiving 120% of citalopram MLD with diazepam (1.77 mg/kg) and cyproheptadine (17.1mg/kg) prevented seizures and death, but propranolol pre-treatment was ineffective. Neuroprotection with diazepam and cyproheptadine was not associated with decreased serotonin plasma concentrations. In conclusion, citalopram-induced deaths resulted from seizures in relation to serotonin release, whilst respiratory and metabolic toxicity was mild. Our observations support the role of serotonin-induced neurotoxicity in citalopram overdose and suggest that cyproheptadine and benzodiazepines, but not beta-blockers, may have a role in the management of citalopram toxicity.


Assuntos
Citalopram/toxicidade , Convulsões/induzido quimicamente , Inibidores Seletivos de Recaptação de Serotonina/toxicidade , Síndrome da Serotonina/induzido quimicamente , Animais , Temperatura Corporal/efeitos dos fármacos , Ciproeptadina/farmacologia , Diazepam/farmacologia , Relação Dose-Resposta a Droga , Dose Letal Mediana , Masculino , Ratos , Ratos Sprague-Dawley , Convulsões/tratamento farmacológico , Serotonina/sangue , Síndrome da Serotonina/tratamento farmacológico
20.
Crit Care Med ; 39(4): 803-11, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21242797

RESUMO

OBJECTIVES: Pralidoxime is an organic cation used as an antidote in addition to atropine to treat organophosphate poisoning. Pralidoxime is rapidly eliminated by the renal route and thus has limited action. The objectives of this work were as follows. 1) Study the role of organic cation transporters in the renal secretion of pralidoxime using organic cation transporter substrates (tetraethylammonium) and knockout mice (Oct1/2⁻/⁻; Oct3⁻/⁻). 2) Assess whether sustained high plasma concentrations increase pralidoxime antidotal activity toward paraoxon-induced respiratory toxicity. SETTING: INSERM U705, Faculté de Pharmacie, Université Paris Descartes, 4 Avenue de l'Observatoire, 75006 Paris, France. SUBJECTS: Rodents: Knockout mice (Oct1/2⁻/⁻; Oct3⁻/⁻) and Sprague-Dawley rats. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: In rats, the renal clearance of pralidoxime was 3.6-fold higher than the creatinine clearance. Pretreatment with tetraethylammonium (75 mg/kg) in rats or deficiencies in organic cation transporters 1 and 2 in mice (Oct1/2⁻/⁻) resulted in a significant increase in plasma pralidoxime concentrations. Lack of Oct3 did not alter plasma pralidoxime concentrations. The antidotal activity of pralidoxime (50 mg/kg intramuscularly) was longer and with greater effect, resulting in a return to normal values when administered to rats pretreated with tetraethylammonium. CONCLUSIONS: Pralidoxime is secreted in rats and mice by renal Oct1 and/or Oct2 but not by Oct3. Modulation of organic cation transporter activity increased the plasma pralidoxime concentrations and the antidotal effect of pralidoxime with sustained return within the normal range of respiratory variables in paraoxon-poisoned rats. These results suggest a promising approach in an animal model toward the increase in efficiency of pralidoxime. However, further studies are needed before these results are extended to human poisoning.


Assuntos
Sistemas de Transporte de Aminoácidos Básicos/efeitos dos fármacos , Antídotos/uso terapêutico , Compostos Organotiofosforados/intoxicação , Compostos de Pralidoxima/uso terapêutico , Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Sistemas de Transporte de Aminoácidos Básicos/fisiologia , Animais , Antídotos/farmacocinética , Inseticidas/intoxicação , Masculino , Camundongos , Camundongos Knockout , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Transportador 1 de Cátions Orgânicos/metabolismo , Transportador 2 de Cátion Orgânico , Paraoxon/intoxicação , Pletismografia Total , Compostos de Pralidoxima/agonistas , Compostos de Pralidoxima/farmacocinética , Compostos de Amônio Quaternário/farmacologia , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...