Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Mol Neurobiol ; 58(10): 5396-5419, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34331199

RESUMO

Neuropathic pain is more prevalent in women. However, females are under-represented in animal experiments, and the mechanisms of sex differences remain inadequately understood. We used the spared nerve injury (SNI) model in rats to characterize sex differences in pain behaviour, unbiased RNA-Seq and proteomics to study the mechanisms. Male and female rats were subjected to SNI- and sham-surgery. Mechanical and cold allodynia were assessed. Ipsilateral lumbar dorsal root ganglia (DRG) and spinal cord (SC) segments were collected for RNA-seq analysis with DESeq2 on Day 7. Cerebrospinal fluid (CSF) samples for proteomic analysis and DRGs and SCs for analysis of IB-4 and CGRP, and IBA1 and GFAP, respectively, were collected on Day 21. Females developed stronger mechanical allodynia. There were no differences between the sexes in CGRP and IB-4 in the DRG or glial cell markers in the SC. No CSF protein showed change following SNI. DRG and SC showed abundant changes in gene expression. Sexually dimorphic responses were found in genes related to T-cells (cd28, ctla4, cd274, cd4, prf1), other immunological responses (dpp4, c5a, cxcr2 and il1b), neuronal transmission (hrh3, thbs4, chrna4 and pdyn), plasticity (atf3, c1qc and reg3b), and others (bhlhe22, mcpt1l, trpv6). We observed significantly stronger mechanical allodynia in females and numerous sexually dimorphic changes in gene expression following SNI in rats. Several genes have previously been linked to NP, while some are novel. Our results suggest gene targets for further studies in the development of new, possibly sex-specific, therapies for NP.


Assuntos
Gânglios Espinais/metabolismo , Hiperalgesia/genética , Hiperalgesia/metabolismo , Neuropatia Ciática/genética , Neuropatia Ciática/metabolismo , Diferenciação Sexual , Medula Espinal/metabolismo , Animais , Peptídeo Relacionado com Gene de Calcitonina/biossíntese , Peptídeo Relacionado com Gene de Calcitonina/genética , Proteínas de Ligação ao Cálcio/biossíntese , Proteínas de Ligação ao Cálcio/genética , Feminino , Expressão Gênica , Proteína Glial Fibrilar Ácida/biossíntese , Proteína Glial Fibrilar Ácida/genética , Masculino , Proteínas dos Microfilamentos/biossíntese , Proteínas dos Microfilamentos/genética , Medição da Dor/métodos , Proteômica/métodos , Ratos , Ratos Sprague-Dawley
2.
Eur J Pain ; 20(2): 297-306, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26031840

RESUMO

BACKGROUND: Oxycodone is increasingly being used in combination with pregabalin. Pregabalin use is prevalent in opioid-dependent individuals. A high number of deaths caused by the co-use of gabapentinoids and opioids occur. It is not known whether pregabalin affects concentrations of oxycodone or morphine in the central nervous system. METHODS: Effects of pregabalin on acute oxycodone or morphine-induced antinociception, tolerance and sedation were studied using tail-flick, hot plate and rotarod tests in male Sprague-Dawley rats. Concentrations of pregabalin, opioids and their major metabolites in the brain were quantified by mass spectrometry. RESULTS: In the hot plate test, morphine (2.5 mg/kg, s.c.) caused antinociception of 28% maximum possible effect (MPE), whereas pregabalin (50 mg/kg, i.p.) produced 8-10% MPE. Co-administration of pregabalin and morphine resulted in antinociception of 63% MPE. Oxycodone (0.6 mg/kg s.c.) produced antinociception of 18% MPE, which increased to 39% MPE after co-administration with pregabalin. When pregabalin 10 mg/kg was administered before oxycodone (0.6 mg/kg, s.c.) or morphine (2.5 mg/kg), only the effect of oxycodone was potentiated in the tail-flick and the hot plate tests. Brain concentrations of the opioids, their major metabolites and pregabalin were unchanged. Pregabalin co-administration (50 mg/kg, i.p., once daily) did not prevent the development of morphine tolerance. CONCLUSIONS: Pregabalin potentiated antinociceptive and sedative effects of oxycodone and morphine in acute nociception. Co-administration of pregabalin with the opioids did not affect the brain concentrations of oxycodone or morphine. Pregabalin did not prevent morphine tolerance.


Assuntos
Analgésicos Opioides/uso terapêutico , Morfina/uso terapêutico , Nociceptividade/efeitos dos fármacos , Oxicodona/uso terapêutico , Dor/tratamento farmacológico , Pregabalina/uso terapêutico , Analgésicos Opioides/farmacologia , Animais , Interações Medicamentosas , Quimioterapia Combinada , Temperatura Alta , Masculino , Morfina/farmacologia , Oxicodona/farmacologia , Medição da Dor/efeitos dos fármacos , Pregabalina/farmacologia , Ratos , Ratos Sprague-Dawley
3.
Br J Pharmacol ; 172(11): 2799-813, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25297798

RESUMO

BACKGROUND AND PURPOSE: The effects of ketamine in attenuating morphine tolerance have been suggested to result from a pharmacodynamic interaction. We studied whether ketamine might increase brain morphine concentrations in acute coadministration, in morphine tolerance and morphine withdrawal. EXPERIMENTAL APPROACH: Morphine minipumps (6 mg·day(-1) ) induced tolerance during 5 days in Sprague-Dawley rats, after which s.c. ketamine (10 mg·kg(-1) ) was administered. Tail flick, hot plate and rotarod tests were used for behavioural testing. Serum levels and whole tissue brain and liver concentrations of morphine, morphine-3-glucuronide, ketamine and norketamine were measured using HPLC-tandem mass spectrometry. KEY RESULTS: In morphine-naïve rats, ketamine caused no antinociception whereas in morphine-tolerant rats there was significant antinociception (57% maximum possible effect in the tail flick test 90 min after administration) lasting up to 150 min. In the brain of morphine-tolerant ketamine-treated rats, the morphine, ketamine and norketamine concentrations were 2.1-, 1.4- and 3.4-fold, respectively, compared with the rats treated with morphine or ketamine only. In the liver of morphine-tolerant ketamine-treated rats, ketamine concentration was sixfold compared with morphine-naïve rats. After a 2 day morphine withdrawal period, smaller but parallel concentration changes were observed. In acute coadministration, ketamine increased the brain morphine concentration by 20%, but no increase in ketamine concentrations or increased antinociception was observed. CONCLUSIONS AND IMPLICATIONS: The ability of ketamine to induce antinociception in rats made tolerant to morphine may also be due to increased brain concentrations of morphine, ketamine and norketamine. The relevance of these findings needs to be assessed in humans.


Assuntos
Analgésicos Opioides/farmacologia , Comportamento Animal/efeitos dos fármacos , Encéfalo/metabolismo , Tolerância a Medicamentos , Ketamina/farmacologia , Morfina/farmacologia , Analgésicos/metabolismo , Analgésicos/farmacologia , Analgésicos Opioides/metabolismo , Animais , Cromatografia Líquida de Alta Pressão , Quimioterapia Combinada , Injeções Subcutâneas , Ketamina/análogos & derivados , Ketamina/metabolismo , Fígado/metabolismo , Morfina/metabolismo , Derivados da Morfina/metabolismo , Dor/tratamento farmacológico , Medição da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Espectrometria de Massas em Tandem
4.
Eur J Pain ; 18(3): 386-95, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23900882

RESUMO

BACKGROUND: Spironolactone, a commonly used mineralocorticoid receptor antagonist, has been reported to potentiate the effect of morphine in the rat. The aim of this study was to investigate the effects of spironolactone on morphine antinociception and tissue distribution. METHODS: The effects of spironolactone on acute morphine-induced antinociception, induction of morphine tolerance and established morphine tolerance were studied with tail-flick and hot plate tests in male Sprague-Dawley rats. Serum, brain, and liver morphine and its metabolite concentrations were quantified using high-pressure liquid chromatography-tandem mass spectrometry. Spironolactone was also administered with the peripherally acting, P-glycoprotein (P-gp) substrate loperamide to test whether spironolactone allows loperamide to pass the blood-brain barrier. RESULTS: Spironolactone (50 mg/kg, i.p.) had no antinociceptive effects of its own, but it enhanced the antinociceptive effect of morphine in both thermal tests. Two doses of spironolactone enhanced the maximum possible effect (MPE) from 19.5% to 100% in the hot plate test 90 min after administration of 4 mg/kg morphine. Morphine concentrations in the brain were increased fourfold at 90 min by spironolactone. Spironolactone did not inhibit the formation of morphine-3-glucuronide. Acute spironolactone restored morphine antinociception in morphine-tolerant rats but did not inhibit the development of tolerance. The peripherally restricted opioid, loperamide (10 mg/kg), had no antinociceptive effects when administered alone, but co-administration with spironolactone produced a 40% MPE in the hot plate test. CONCLUSIONS: Spironolactone has no antinociceptive effects in thermal models of pain, but it enhances the antinociceptive effects of morphine mainly by increasing morphine central nervous system concentrations, probably by inhibiting P-gp.


Assuntos
Analgésicos/uso terapêutico , Antagonistas de Receptores de Mineralocorticoides/uso terapêutico , Morfina/uso terapêutico , Dor/tratamento farmacológico , Espironolactona/uso terapêutico , Analgésicos/farmacocinética , Animais , Interações Medicamentosas , Masculino , Morfina/farmacocinética , Medição da Dor , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
5.
Scand J Pain ; 1(1): 24-33, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-29913934

RESUMO

Oxycodone is a strong opioid and it is increasingly used in the management of acute and chronic pain. The pharmacodynamic effects of oxycodone are mainly mediated by the µ-opioid receptor. However, its affinity for the µ-opioid receptor is significantly lower compared with that of morphine and it has been suggested that active metabolites may play a role in oxycodone analgesia. Oxycodone is mainly metabolized by hepatic cytochrome (CYP) enzymes 2D6 and 3A4. Oxycodone is metabolized to oxymorphone, a potent µ-opioid receptor agonist by CYP2D6. However, CYP3A4 is quantitatively a more important metabolic pathway. Chronic pain patients often use multiple medications. Therefore it is important to understand how blocking or inducing these metabolic pathways may affect oxycodone induced analgesia. The aim of this study was to find out whether blocking CYP2D6 would decrease oxycodone induced analgesia in chronic pain patients. The effects of the antidepressant paroxetine, a potent inhibitor of CYP2D6, on the analgesic effects and pharmacokinetics of oral oxycodone were studied in 20 chronic pain patients using a randomized, double-blind, placebo-controlled cross-over study design. Pain intensity and rescue analgesics were recorded daily, and the pharmacokinetics and pharmacodynamics of oxycodone were studied on the 7th day of concomitant paroxetine (20 mg/day) or placebo administration. The patients were genotyped for CYP2D6, 3A4, 3A5 and ABCB1. Paroxetine had significant effects on the metabolism of oxycodone but it had no statistically significant effect on oxycodone analgesia or use of morphine for rescue analgesia. Paroxetine increased the dose-adjusted mean AUC0-12h of oxycodone by 19% (-23 to 113%; P = 0.003), and that of noroxycodone by 100% (5-280%; P < 0.0001) but decreased the AUC0-12 h of oxymorphone by 67% (-100 to -22%; P < 0.0001) and that of noroxymorphone by 68% (-100 to -16%; P < 0.0001). Adverse effects were also recorded in a pain diary for both 7-day periods (placebo/paroxetine). The most common adverse effects were drowsiness and nausea/vomiting. One patient out of four reported dizziness and headache during paroxetine co-administration, whereas no patient reported these during placebo administration (P = 0.0471) indicating that these adverse effects were due to paroxetine. No statistically significant associations of the CYP2D6 or CYP3A4/5 genotype of the patients and the pharmacokinetics of oxycodone or its metabolites, extent of paroxetine-oxycodone interaction, or analgesic effects were observed probably due to the limited number of patients studied. The results of this study strongly suggest that CYP2D6 inhibition does not significantly change oxycodone analgesia in chronic pain patients and that the analgesic activity of oxycodone is mainly due to the parent compound and that metabolites, e.g. oxymorphone, play an insignificant role. The clinical implication of these results is that induction of the metabolism of oxycodone may lead to inadequate analgesia while increased drug effects can be expected after addition of potent CYP3A4/5 inhibitors particularly if combined with CYP2D6 inhibitors or when administered to poor metabolizers of CYP2D6.

6.
Acta Oncol ; 39(8): 941-7, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11207001

RESUMO

Oxycodone is an opioid analgesic that closely resembles morphine. Oxymorphone, the active metabolite of oxycodone, is formed in a reaction catalyzed by CYP2D6, which is under polymorphic genetic control. The role of oxymorphone in the analgesic effect of oxycodone is not yet clear. In this study, controlled-release (CR) oxycodone and morphine were examined in cancer pain. CR oxycodone and morphine were administered to 45 adult patients with stable pain for 3-6 days after open-label titration in a randomized, double-blind, cross-over trial. Twenty patients were evaluable. Both opioids provided adequate analgesia. The variation in plasma morphine concentrations was higher than that of oxycodone, consistent with the lower bioavailability of morphine. Liver dysfunction affected selectively either oxycodone or morphine metabolism. Three patients with markedly aberrant plasma opioid concentrations are presented. Significant individual variation in morphine and oxycodone metabolism may account for abnormal responses during treatment of chronic cancer pain.


Assuntos
Analgésicos Opioides/farmacocinética , Morfina/farmacocinética , Neoplasias/metabolismo , Oxicodona/farmacocinética , Dor/tratamento farmacológico , Dor/metabolismo , Administração Oral , Adulto , Idoso , Analgésicos Opioides/sangue , Analgésicos Opioides/uso terapêutico , Estudos Cross-Over , Citocromo P-450 CYP2D6/metabolismo , Debrisoquina/metabolismo , Preparações de Ação Retardada , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Morfinanos/sangue , Morfina/sangue , Morfina/uso terapêutico , Derivados da Morfina/sangue , Neoplasias/sangue , Neoplasias/complicações , Oxicodona/sangue , Oxicodona/uso terapêutico , Dor/etiologia , Medição da Dor/efeitos dos fármacos , Fenótipo
7.
Exp Neurol ; 147(2): 428-36, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9344567

RESUMO

We studied whether a chronic neuropathy induced by unilateral spinal nerve ligation changes the response characteristics of spinal dorsal horn wide-dynamic range (WDR) neurons or their periaqueductal gray (PAG)-induced descending modulation. Experiments were performed in rats with behaviorally demonstrated allodynia induced by spinal nerve ligation and in a group of nonneuropathic control rats. The stimulus-response functions of WDR neurons for mechanical and thermal stimuli and the modulation of their peripherally evoked responses by electrical stimulation of the PAG were determined under pentobarbital anesthesia. The results showed that neuropathy caused a significant leftward shift in stimulus-response functions for mechanical stimuli. In contrast, stimulus-response functions for noxious heat stimuli in the neuropathic limb were, if anything, shifted rightward, although this shift was short of statistical significance. In neuropathic rats, PAG stimulation produced a significantly stronger attenuation of spinal neuronal responses induced by noxious heat in the unoperated than in the operated side. At the intensity that produced attenuation of noxious heat stimuli, PAG stimulation did not produce any significant change in spinal neuronal responses evoked by mechanical stimuli either from the operated or the nonoperated hindlimb of the neuropathic rats. Spontaneous activity of WDR neurons was higher in the operated side of neuropathic rats than in control rats. Afterdischarges evoked by peripheral stimuli were observed in 1/16 of the WDR neurons ipsilateral to spinal nerve ligation and not at all in other experimental groups. The WDR neurons studied were not activated by innocuous or noxious cold stimuli. The results indicate that spinal nerve ligation induces increased spontaneous activity and enhanced responses to mechanical stimuli in the spinal dorsal horn WDR neurons, whereas noxious heat-evoked responses are not significantly changed or if anything, attenuated. Moreover, the inhibition of noxious heat stimuli by PAG stimulation is attenuated in the neuropathic side. It is proposed that the observed changes in the response characteristics of the spinal dorsal horn WDR neurons and in their descending modulation may contribute to the neuropathic symptoms in these animals.


Assuntos
Hiperalgesia/fisiopatologia , Plasticidade Neuronal , Nociceptores/fisiologia , Dor/fisiopatologia , Substância Cinzenta Periaquedutal/fisiopatologia , Medula Espinal/fisiopatologia , Nervos Espinhais/lesões , Animais , Vias Eferentes/fisiopatologia , Estimulação Elétrica , Temperatura Alta , Ligadura , Masculino , Neurônios Aferentes/fisiologia , Limiar da Dor/fisiologia , Ratos , Ratos Sprague-Dawley , Medula Espinal/patologia , Nervos Espinhais/fisiopatologia
9.
Pharmacol Toxicol ; 76(6): 368-70, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7479577

RESUMO

A functional connection, evidenced by synergism and cross-tolerance, seems to exist between the mu-opioid and the alpha 2-adrenergic receptors in the superficial dorsal horn of the rat spinal cord. In this study possible cross-antagonism between these systems was studied by intrathecal administration of morphine 2.5 micrograms or dexmedetomidine 1.25 micrograms 15 min. after subcutaneous injection of naloxone 1.0 mg/kg or atipamezole 3.0 mg/kg in the morphine group and 4.0 mg/kg or 1.0 mg/kg in the dexmedetomidine group, respectively. No cross-antagonism of the antinociceptive effect was seen either in the tail flick or in the hot plate test at 15, 30, 45 and 60 min. from the pretreatment.


Assuntos
Antagonistas de Receptores Adrenérgicos alfa 2 , Receptores Opioides mu/antagonistas & inibidores , Medula Espinal/efeitos dos fármacos , Antagonistas Adrenérgicos alfa/farmacologia , Analgésicos Opioides/farmacologia , Animais , Interações Medicamentosas , Imidazóis/farmacologia , Masculino , Medetomidina , Morfina/farmacologia , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Ratos , Ratos Sprague-Dawley , Medula Espinal/metabolismo
10.
Peptides ; 16(5): 973-7, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-7479344

RESUMO

Neuropeptide FF (NPFF) has been found to act as an antiopioid peptide. However, IT NPFF has recently been shown to potentiate the antinociceptive effects of IT morphine and to produce antinociception on its own. The aim of this study was to find out whether pretreatment with NPFF causes a comparable potentiation of dexmedetomidine-induced antinociception. NPFF (0.05-10.0 nmol) produced no antinociceptive effects in the rat tail flick test. NPFF potentiated the antinociceptive effect of IT morphine (7.8 nmol). This potentiation was prevented by IT naltrindole (28 nmol), which did not attenuate the antinociceptive effect of morphine. Dexmedetomidine (1.6-6.4 nmol) produced a dose-dependent antinociceptive effect, which was not potentiated by NPFF. Activation of the endogenous delta-opioid system due to the antiopioid effect of IT NPFF is proposed as an explanation to the reported differential action of NPFF on the mu-opioid and the alpha 2-adrenergic systems.


Assuntos
Neuropeptídeos/farmacologia , Oligopeptídeos/farmacologia , Dor/fisiopatologia , Receptores Adrenérgicos alfa 2/fisiologia , Receptores Opioides mu/fisiologia , Medula Espinal/fisiologia , Agonistas alfa-Adrenérgicos/farmacologia , Analgésicos Opioides/farmacologia , Animais , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Imidazóis/farmacologia , Masculino , Medetomidina , Morfina/farmacologia , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos alfa 2/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Fatores de Tempo
11.
J Pharmacol Exp Ther ; 271(3): 1306-13, 1994 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7996439

RESUMO

The antinociceptive efficacies of both intrathecally (i.t.) and systemically administered dexmedetomidine (a selective alpha-2 adrenoceptor agonist) and U-50,488H [trans(+/-)-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl)-cyclohexyl]- benzene-acetamide] (a kappa-opioid receptor agonist) were studied during peripheral inflammation induced by carrageenan. The antinociceptive tests were the hot plate (HP), the tail flick (TF) and the paw pressure tests (PP). The motor incoordination, if any, produced by both i.t. and s.c. dexmedetomidine were evaluated with the rotarod. The interaction between dexmedetomidine and U-50,488H and between atipamezole (a selective alpha-2 adrenoceptor antagonist) and U-50,488H were also assessed. The carrageenan injection induced not only peripheral hyperalgesia but also central sensitization, as assessed by decreased PP and TF latencies, respectively. The i.t. dexmedetomidine (0.15, 0.45, 1.35, 4.05) micrograms) resulted in dose-dependent increases in the PP thresholds and TF latencies in both the control rats and the rats with unilateral inflammation, without causing changes in motor coordination, whereas on s.c. administration of dexmedetomidine (3-100 micrograms/kg), antinociception was produced in PP only at doses (30 micrograms/kg) that already interfered with rotarod performance. U-50,488H was ineffective i.t. (5-200 micrograms) but, on s.c. administration (2.5-22.5 mg/kg), dose-dependent increases were found in the PP thresholds and TF latencies of the rats with unilateral inflammation. Atipamezole, in a dose (3 mg/kg) that has been shown to block the antinociceptive effects of dexmedetomidine, did not modify the antinociceptive effects of U-50,488H.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Agonistas alfa-Adrenérgicos/farmacologia , Analgésicos não Narcóticos/farmacologia , Imidazóis/farmacologia , Pirrolidinas/farmacologia , Receptores Adrenérgicos alfa 2/efeitos dos fármacos , Receptores Opioides kappa/agonistas , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida , Animais , Relação Dose-Resposta a Droga , Inflamação/tratamento farmacológico , Masculino , Medetomidina , Limiar da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
12.
J Pharmacol Exp Ther ; 265(2): 551-8, 1993 May.
Artigo em Inglês | MEDLINE | ID: mdl-8098759

RESUMO

Intrathecal administration of morphine, Tyr-D-Ser(otbu)-Gly-Phe-Leu-Thr or dexmedetomidine for 5 to 10 days rendered rats tolerant to the test drug as measured by both behavioral and electrophysiological tests. Tolerance to the alpha-2 adrenergic agonist dexmedetomidine required a longer induction time and was not as pronounced as the tolerance to the opioid agonists, probably because lower doses of dexmedetomidine relative to the ED50 dose were used to avoid sedation. In the behavioral studies we used the tail-flick test and in the electrophysiological studies recordings were made from dorsal horn nociceptive neurons under halothane anesthesia. After completion of the behavioral testing the same animals were then used in the electrophysiological study. Cross-tolerance developed clearly between the mu opioid agonist morphine and the alpha-2 adrenergic agonist dexmedetomidine, whereas no cross-tolerance was seen between the delta opioid agonist Tyr-D-Ser(otbu)-Gly-Phe-Leu-Thr and either morphine or dexmedetomidine. This is further evidence to support the assumption that in the dorsal horn the mu opioid and the alpha-2 adrenergic receptor are linked functionally, whereas the delta opioid receptor operates independently. These results have also important clinical implications indicating the potential of delta opioid agonists to restore analgesia in a morphine-tolerant patient.


Assuntos
Receptores Adrenérgicos alfa/efeitos dos fármacos , Receptores Opioides delta/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Agonistas alfa-Adrenérgicos/farmacologia , Sequência de Aminoácidos , Animais , Comportamento Animal/efeitos dos fármacos , Tolerância a Medicamentos , Potenciais Evocados , Imidazóis/farmacologia , Masculino , Medetomidina , Dados de Sequência Molecular , Morfina/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Oligopeptídeos/farmacologia , Ratos , Ratos Sprague-Dawley
13.
Neurosci Lett ; 139(1): 65-8, 1992 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-1357605

RESUMO

The interaction between the spinal antinociceptive effects of selective mu or delta opioid agonists morphine and DSTBULET (Tyr-D-Ser(OtBu)-Gly-Phe-Leu-Thr), respectively, and the selective alpha 2 adrenergic agonist dexmedetomidine was examined on convergent dorsal horn neuronal responses in the intact anaesthetized rat. The coadministration of intrathecal morphine (0.5 microgram, 2.5 micrograms) and dexmedetomidine (0.5 microgram) produced a greater than additive inhibition of C fibre-evoked responses. Inhibitions were reversed by either the opioid antagonist naloxone or the alpha 2 adrenergic antagonist atipamezole. The coadministration of intrathecal DSTBULET (1 microgram, 2.5 micrograms) and dexmedetomidine did not result in a supra-additive inhibition of C fibre-evoked responses. The results suggest that mu rather than delta opioid receptors are involved in the synergism of spinal opioid and alpha 2 adrenergic antinociception.


Assuntos
Agonistas alfa-Adrenérgicos/farmacologia , Analgésicos/farmacologia , Entorpecentes/farmacologia , Receptores Opioides delta/fisiologia , Receptores Opioides mu/fisiologia , Medula Espinal/efeitos dos fármacos , Antagonistas Adrenérgicos alfa/farmacologia , Sequência de Aminoácidos , Animais , Sinergismo Farmacológico , Masculino , Dados de Sequência Molecular , Oligopeptídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos
14.
Eur J Pharmacol ; 216(1): 97-101, 1992 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-1326441

RESUMO

The spinal antinociceptive potency of the delta-opioid receptor agonist, Tyr-D-Ser(otbu)-Gly-Phe-Leu-Thr (DSTBULET), was studied in rats. The tail flick test was used as nociceptive stimulus and the rotarod test was used to detect any motor or sedative effects. A dose-response curve was also made for the mu-opioid receptor agonist, morphine. The ED50 for DSTBULET was 0.3 micrograms (0.4 nmol) and a near 100% maximum effect was achieved with 5 micrograms (7.5 nmol). No motor or sedative effects were detected. Antinociception by DSTBULET was antagonized by s.c. naltrindole (1 mg/kg), a selective delta-opioid receptor antagonist, and naloxone (1 mg/kg), a non-selective opioid receptor antagonist. The ED50 for morphine was 0.5 micrograms (1.0 nmol) and the antinociceptive effects were not antagonized by naltrindole (1 mg/kg). The results evidence further the important role of the delta-opioid receptor in spinal nociceptive processing.


Assuntos
Analgésicos/farmacologia , Naltrexona/análogos & derivados , Oligopeptídeos/farmacologia , Receptores Opioides/metabolismo , Sequência de Aminoácidos , Analgésicos/administração & dosagem , Animais , Relação Dose-Resposta a Droga , Indóis/farmacologia , Injeções Espinhais , Masculino , Dados de Sequência Molecular , Morfinanos/farmacologia , Morfina/farmacologia , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Oligopeptídeos/administração & dosagem , Ratos , Ratos Endogâmicos , Receptores Opioides delta
15.
Eur J Pharmacol ; 215(1): 127-33, 1992 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-1355441

RESUMO

The actions of the selective alpha 2-adrenoceptor agonist dexmedetomidine were examined on the nociceptive C and innocuous A beta fibre-evoked responses of dorsal horn neurones to transcutaneous electrical stimulation in the intact anaesthetized rat. C fibre-evoked responses were dose dependently reduced by intrathecal dexmedetomidine--to a maximum 86 +/- 6% inhibition by 10 micrograms of the agonist. The ED50 for inhibition of C fibre responses was estimated to be 2.5 micrograms. A beta-evoked responses were inhibited to a lesser degree--a maximum 54 +/- 8% inhibition after 10 micrograms dexmedetomidine. The antinociceptive effects of dexmedetomidine were reversed by the alpha 2-adrenoceptor antagonist atipamezole and the opioid antagonist naloxone. The results are discussed with reference to adrenergic and opioid mechanisms in the spinal cord.


Assuntos
Analgésicos/farmacologia , Imidazóis/farmacologia , Neurônios/efeitos dos fármacos , Nociceptores/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Antagonistas Adrenérgicos alfa/farmacologia , Analgésicos/antagonistas & inibidores , Anestesia , Animais , Clonidina/farmacologia , Relação Dose-Resposta a Droga , Eletrofisiologia , Imidazóis/antagonistas & inibidores , Masculino , Medetomidina , Naloxona/farmacologia , Neurônios/fisiologia , Ratos , Ratos Endogâmicos , Medula Espinal/citologia , Medula Espinal/fisiologia
16.
Pharmacol Toxicol ; 70(2): 125-30, 1992 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-1508838

RESUMO

Antinociception and central nervous system depression (CNSD) caused by intraperitoneal, intrathecal and subcutaneous administration of oxycodone or morphine were studied in a randomized and blind, saline controlled study in rats. Antinociception was assessed with the tail-flick and hot plate tests. CNSD was assessed by testing the corneal, placing and righting reflexes and with a 4-point catalepsy score. Intraperitoneally and subcutaneously administered oxycodone and morphine were given in doses of 2.5-10 and 5-20 mg kg-1 respectively. The intrathecal doses were 12.5 micrograms and 100 micrograms of oxycodone and 6.25 micrograms and 50 micrograms of morphine. In both nociceptive tests subcutaneously and intraperitoneally administered oxycodone was 2-4 times more potent than morphine, while intrathecal morphine was over 14 times more potent. CNSD was more profound with oxycodone than with morphine after intraperitoneal and subcutaneous administration, but was not observed after intratechal administration of either drug. Differences in opioid receptor affinities, liposolubilities and metabolism are discussed as possible explanations.


Assuntos
Analgésicos/farmacologia , Sistema Nervoso Central/efeitos dos fármacos , Morfina/farmacologia , Oxicodona/farmacologia , Animais , Relação Dose-Resposta a Droga , Feminino , Injeções Intraperitoneais , Injeções Espinhais , Injeções Subcutâneas , Morfina/administração & dosagem , Oxicodona/administração & dosagem , Ratos , Ratos Endogâmicos
17.
Br J Anaesth ; 67(6): 759-63, 1991 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-1768546

RESUMO

The first pass uptake, metabolism and recovery of bupivacaine were examined in an intact rabbit lung model using a multiple indicator technique with rapid sequential sampling. The rabbits were allocated to an acidotic group (pH 7.0-7.1) (n = 8) and a control group (n = 10) with normal pH. Bupivacaine recovery rates were not significantly different: median 93.2% (range 48.9-116.5%) and 94.5% (54.9-123.1%) in control and acidotic groups, respectively. Median peak percentage fractional concentrations of bupivacaine were greater in the acidotic group: 6.22% (2.5-7.65%) vs 4.1% (2.5-6.7%) (P less than 0.05). Median maximum instantaneous pulmonary percentage extraction was less in the acidotic animals than in animals with normal pH: 81.2% (47.1-91.9%) vs 91.0% (82.6-94.5%) (P less than 0.01). Median normalized mean percentage transit time was less in the acidotic group (245.3% (163.4-465.3%)) than in the control group (423.9% (313.9-740.4%)) (P less than 0.01). There was no evidence for bupivacaine metabolism by the lung. The results suggest that acidosis reduced bupivacaine lung uptake and increased its rate of passage through the lung, but did not influence overall drug recovery rates. This has clinical implications for bupivacaine related cardiac and cerebral toxicity.


Assuntos
Acidose/metabolismo , Bupivacaína/farmacocinética , Pulmão/metabolismo , Animais , Biotransformação , Bupivacaína/sangue , Débito Cardíaco/fisiologia , Modelos Biológicos , Coelhos
18.
Brain Res ; 562(2): 327-8, 1991 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-1685347

RESUMO

The interaction between FLFQPQRFamide and alpha 2-adrenergic spinal antinociception was examined in an electrophysiological study in the intact anaesthetised rat. The inhibition of C fibre-evoked neuronal responses by the selective alpha 2-adrenergic agonist dexmedetomidine was significantly reduced by intrathecal FLFQPQRFamide pretreatment. The results suggest a modulatory role of FLFQPQRFamide in spinal alpha 2-adrenergic antinociception.


Assuntos
Agonistas alfa-Adrenérgicos/farmacologia , Analgésicos/farmacologia , Imidazóis/farmacologia , Oligopeptídeos/fisiologia , Sequência de Aminoácidos , Animais , Estimulação Elétrica , Medetomidina , Dados de Sequência Molecular , Fibras Nervosas/efeitos dos fármacos , Ratos
19.
Acta Odontol Scand ; 49(4): 215-8, 1991 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-1927285

RESUMO

The use of opioids in chronic non-malignant pain conditions is largely rejected by the health authorities. Their concern is mostly due to the potential problems of addiction and other adverse effects of opioids. However, in certain pain conditions opioids may be the only effective remedy. This article presents some guidelines for the use of narcotics for non-cancer pain. A case is presented in which methadone in small doses in combination with an antidepressant was the first drug capable of alleviating the patient's suffering. The drug was effective during a period of 9 months.


Assuntos
Dor Facial/tratamento farmacológico , Metadona/uso terapêutico , Dor Intratável/tratamento farmacológico , Administração Oral , Feminino , Humanos , Metadona/administração & dosagem , Pessoa de Meia-Idade
20.
Pharmacol Toxicol ; 68(2): 140-3, 1991 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-1677190

RESUMO

The antinociceptive effects of dexmedetomidine, a highly selective new alpha 2-adrenoceptor agonist, were evaluated in rats after intrathecal, intraperitoneal and subcutaneous administration. Antinociception was tested using the tail-flick method. Both 3 and 6 micrograms of intrathecal dexmedetomidine produced maximal antinociception within 10 min. The effect lasted for up to 6 hr. The smaller dose of 1.5 micrograms produced a mean antinociception of 50% (of the maximum possible effect, MPE%) which lasted for about 2 hr. Subcutaneous atipamezole, a specific alpha 2-adrenergic antagonist completely abolished the antinociception produced by intrathecal dexmedetomidine. When given intraperitoneally, dexmedetomidine produced on average a 50% antinociceptive effect with the highest dose of 60 micrograms/kg. The lower doses of 10 and 30 micrograms/kg were ineffective. After subcutaneous administration a maximal effect was achieved with 120 micrograms/kg, a 70% effect, on average, with 60 micrograms/kg and a short lasting effect of 60% with 30 micrograms/kg. In conclusion, dexmedetomidine is a very potent antinociceptive agent when given intrathecally to rats.


Assuntos
Agonistas alfa-Adrenérgicos/farmacologia , Analgésicos , Imidazóis/farmacologia , Agonistas alfa-Adrenérgicos/administração & dosagem , Animais , Relação Dose-Resposta a Droga , Imidazóis/administração & dosagem , Imidazóis/antagonistas & inibidores , Injeções Intraperitoneais , Injeções Espinhais , Injeções Subcutâneas , Masculino , Medetomidina , Medição da Dor , Ratos , Ratos Endogâmicos , Tempo de Reação/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA