Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Neuroscience ; 146(3): 1275-88, 2007 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-17395382

RESUMO

Sustained exposure to opioid agonists such as morphine increases levels of calcitonin gene-related peptide (CGRP) in the spinal dorsal horn, a response implicated in the development of opioid tolerance and physical dependence. Recent evidence suggests that both the opioid-induced increase in CGRP and the development of opioid physical dependence are suppressed by blockade of spinal cannabinoid (CB1)-receptors. The present study examined whether CB1-receptor activity also has a role in the development of opioid tolerance. In rats implanted with spinal catheters, repeated acute injections of morphine (15 microg) delivered over 4 h resulted in a rapid decline of thermal and mechanical antinociception and a significant loss of analgesic potency, reflecting development of acute opioid tolerance. In another set of experiments, chronic administration of spinal morphine (15 microg) once daily for 5 days produced a similar loss of analgesic effect and a marked increase in CGRP-immunoreactivity in the superficial laminae of the dorsal horn. Consistent with the in vivo findings, primary cultures of adult dorsal root ganglion (DRG) neurons exposed to morphine for 5 days showed a significant increase in the number of CGRP-immunoreactive neurons. Co-administration of acute or chronic morphine with a CB1-receptor antagonist/inverse agonist, 1-(2,4-dichlorophenyl)-5-(4-iodophenyl)-4-methyl-N-1-piperidinyl-1H-pyrazole-3-carboxamide (AM-251), inhibited the development of both acute and chronic analgesic tolerance. In animals already exhibiting tolerance to morphine, intervention with AM-251 restored morphine analgesic potency. Co-administration with AM-251 attenuated the morphine-induced increase in CGRP-immunoreactivity in the spinal cord and in DRG cultured neurons. Collectively, the results of this study suggest that activity of endocannabinoids, mediated via CB1-receptors, contributes to both the development and maintenance of opioid tolerance by influencing the opioid-induced increase in spinal CGRP.


Assuntos
Analgésicos Opioides/farmacologia , Tolerância a Medicamentos/fisiologia , Receptor CB1 de Canabinoide/fisiologia , Animais , Células Cultivadas , Interpretação Estatística de Dados , Relação Dose-Resposta a Droga , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Injeções Espinhais , Masculino , Morfina/farmacologia , Nociceptores/efeitos dos fármacos , Medição da Dor/efeitos dos fármacos , Piperidinas , Pirazóis , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Receptor CB1 de Canabinoide/agonistas , Receptores de Peptídeo Relacionado com o Gene de Calcitonina/biossíntese
2.
Br J Pharmacol ; 151(6): 877-87, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17502848

RESUMO

BACKGROUND AND PURPOSE: Ultralow doses of naltrexone, a non-selective opioid antagonist, have previously been found to augment acute morphine analgesia and block the development of tolerance to this effect. Since morphine tolerance is dependent on the activity of micro and delta receptors, the present study investigated the effects of ultralow doses of antagonists selective for these receptor types on morphine analgesia and tolerance in tests of thermal and mechanical nociception. EXPERIMENTAL APPROACH: Effects of intrathecal administration of mu-receptor antagonists, CTOP (0.01 ng) or CTAP (0.001 ng), or a delta-receptor antagonist, naltrindole (0.01 ng), on spinal morphine analgesia and tolerance were evaluated using the tail-flick and paw-pressure tests in rats. KEY RESULTS: Both micro and delta antagonists augmented analgesia produced by a sub-maximal (5 microg) or maximal (15 microg) dose of morphine. Administration of the antagonists with morphine (15 microg) for 5 days inhibited the progressive decline of analgesia and prevented the loss of morphine potency. In animals exhibiting tolerance to morphine, administration of the antagonists with morphine produced a recovery of the analgesic response and restored morphine potency. CONCLUSIONS AND IMPLICATIONS: Combining ultralow doses of micro- or delta-receptor antagonists with spinal morphine augmented the acute analgesic effects, inhibited the induction of chronic tolerance and reversed established tolerance. The remarkably similar effects of micro- and delta-opioid receptor antagonists on morphine analgesia and tolerance are interpreted in terms of blockade of the latent excitatory effects of the agonist that limit expression of its full activity.


Assuntos
Analgésicos Opioides/farmacologia , Morfina/farmacologia , Antagonistas de Entorpecentes/farmacologia , Dor/tratamento farmacológico , Analgésicos Opioides/administração & dosagem , Animais , Relação Dose-Resposta a Droga , Interações Medicamentosas , Tolerância a Medicamentos , Injeções Espinhais , Masculino , Morfina/administração & dosagem , Naltrexona/administração & dosagem , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/administração & dosagem , Medição da Dor , Fragmentos de Peptídeos , Peptídeos/administração & dosagem , Peptídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides mu/antagonistas & inibidores , Somatostatina/administração & dosagem , Somatostatina/análogos & derivados , Somatostatina/farmacologia
3.
Peptides ; 27(5): 953-63, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16515821

RESUMO

Neuropeptide FF and related synthetic amidated peptides have been shown to elicit sustained anti-nociceptive responses and potently augment spinal anti-nociceptive actions of spinal morphine in tests of thermal and mechanical nociception. Recent studies have described the occurrence of another octapeptide, neuropeptide SF (NPSF) in the spinal cord and the cerebrospinal fluid and demonstrated its affinity for the NPFF receptors. This study examined the effects of NPSF and two putative precursor peptides, EFW-NPSF and NPAF, on the spinal actions of morphine in normal and opioid tolerant rats using the tailflick and pawpressure tests. In normal rats, NPSF demonstrated weak intrinsic activity but sub-effective doses of the peptide significantly increased the magnitude and duration of spinal morphine anti-nociception in both tests. A low-dose of NPSF also augmented the spinal actions of a delta receptor agonist, deltorphin. The morphine-potentiating effect of NPSF was shared by EFW-NPSF and the octadecapeptide NPAF. In animal rendered tolerant by continuous intrathecal infusion of morphine for 6 days, low dose NPSF itself elicited a significant anti-nociceptive response and potently increased morphine-induced response in both tests. In animals made tolerant by repeated injections of intrathecal morphine, administration of NPSF, EFW-NPSF, and NPAF with morphine reversed the loss of the anti-nociceptive effect and restored the agonist potency. The results demonstrate that in normal animals NPSF and related peptides exert strong potentiating effect on morphine anti-nociception at the spinal level and in tolerant animals these agents can reverse the loss of morphine potency.


Assuntos
Analgesia , Tolerância a Medicamentos/fisiologia , Neuropeptídeos/farmacologia , Medula Espinal/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Interações Medicamentosas , Masculino , Morfina/farmacologia , Oligopeptídeos/farmacologia , Medição da Dor , Peptídeos/farmacologia , Ratos , Ratos Sprague-Dawley
4.
Br J Pharmacol ; 58(1): 101-7, 1976 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-974367

RESUMO

1 The opiate antagonist naloxone, injected or topically applied to the cerebral cortex, had no significant effect on the spontaneous output of cortical acetylcholine (ACh) in rats. 2 Morphine (2.5 mg/kg) administered intravenously inhibited the release of cortical ACh. A subsequent injection of naloxone rapidly reversed morphine-induced inhibition, and produced a sustained increase in the release of ACh. Topical application of naloxone solutions, after morphine, produced a slow and weak reversal of its inhibitory action. 3 Destruction of the medial thalamus abolished both the inhibitory effects of morphine on the cortical ACh release, and its antagonism by naloxone administered after the agonist. 4 Injection of naloxone in a low dose (0.1 mg/kg) increased the release of cortical ACh provoked by electrical stimulation of either the medial thalamus or the reticular formation in normal rats. In the morphine-dependent rat, naloxone also facilitated the evoked release and its action was greater than in control animals. The facilitatory effect of naloxone on the cortical release evoked by stimulation of the medial thalamus was greater than its effect on the release evoked by stimulation of the reticular formation in both normal and morphine-dependent rats. 5 Naltrexone, a narcotic antagonist, also facilitated the electrically stimulated release of cortical ACh. 6 It is suggested that (a) morphine and naloxone act at a subcortical site, probably the medial thalamus, to modify the cortical ACh release and that (b) naloxone may facilitate the electrically-induced release of ACh in the CNS by antagonizing the effect of the endogenous morphine-like factor, enkephalin.


Assuntos
Encéfalo/fisiologia , Morfina/farmacologia , Naloxona/farmacologia , Sistema Nervoso Parassimpático/efeitos dos fármacos , Acetilcolina/metabolismo , Animais , Córtex Cerebral/metabolismo , Interações Medicamentosas , Estimulação Elétrica , Técnicas In Vitro , Naltrexona/farmacologia , Ratos , Fatores de Tempo
5.
Br J Pharmacol ; 78(2): 433-40, 1983 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-6831119

RESUMO

1--Electrical stimulation of nerves in the forepaw of anaesthetized rats caused an increase in the release of acetylcholine (ACh) from the cerebral cortex in vivo. Actions of naloxone (Nal) enantiomers and naltrexone (Ntx) were tested on this release in normal animals and those lacking pituitary gland for 3 weeks. 2--In normal animals systemic administration of (-)-Nal or Ntx, but not (+)-Nal caused a significant increase in the evoked release of ACh. Spinally administered (-)-Nal did not produce this effect. Cortical application of (-)-Nal produced a smaller increase in the evoked release of ACh. 3--In hypophysectomized rats the stimulatory action of (-)-Nal or Ntx on ACh release was significantly reduced. The ability of (-)-Nal to reverse inhibitory action of morphine or the enkephalin (FK 33,824) was not affected by hypophysectomy. 4--It is suggested that (-)-Nal and Ntx increase the stimulated release of cortical ACh by blocking the inhibitory action of an endogenous opioid at a subcortical site. An intact pituitary appears essential for a full expression of the Nal effect on evoked ACh release.


Assuntos
Acetilcolina/metabolismo , Encéfalo/metabolismo , Antagonistas de Entorpecentes/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Estimulação Elétrica , Encefalinas/metabolismo , Hipofisectomia , Naloxona/farmacologia , Naltrexona/farmacologia , Ratos , Ratos Endogâmicos , Estereoisomerismo
6.
Br J Pharmacol ; 71(1): 201-10, 1980.
Artigo em Inglês | MEDLINE | ID: mdl-7470736

RESUMO

1 Methionine (Met)-enkephalin, leucine (Leu)-enkephalin and their synthetic analogues were tested for effects on the spontaneous release of cortical acetylcholine (ACh) in vivo. The ability of naloxone to reverse the action of enkephalins on ACh release was compared with its action against morphine. An enkephalin analogue, structurally related to Met-enkephalin, was tested for opiate antagonistic activity in ACh release experiments. 2 Intraventricular administration of Met-enkephalin, Leu-enkephalin, D-Ala2-Met5-enkephalinamide (DALA) and D-Ala2-D-Leu5-enkephalin (DALEU) produced a dose-related inhibition of cortical ACh release. Met- and Leu-enkephalin were very similar both in their potency and the time course of their action on ACh release. Both DALA and DALEU were more potent and had a longer duration of action than Leu-enkephalin. Systemic injections of two pentapeptides, D-Met2-Pro5-enkephalinamide and D-Ala2-MePhe4-Met5(O)-ol-enkephalin (33,824), produced a sustained inhibition of cortical ACh release. 3 Naloxone, administered systemically following the depression of ACh release induced by either intraventricular injections of enkephalins (DALA or DALEU), or systemic injections of enkephalins (D-Met2-Pro5-enkephalinamide or 33,824), reversed this depression and restored the release to baseline levels. The effect of D-Met2-Pro5-enkephalinamide on the release of ACh was reversed by naloxone with difficulty. Naloxone also reversed the inhibitory effect of systemic morphine and this reversal was associated with a large overshoot of ACh release. The latter was never observed in the enkephalin experiments. 4 Intraventricular injection of the pentapeptide, D-Ala2-D-Ala3-Met5-enkephalinamide (TAAPM), at doses that did not influence the basal ACh release, blocked or reversed the inhibitory effect of morphine on this release. This peptide did not block the effect of the non-opiate, chlorpromazine, under similar conditions. In two experiments TAAPM failed to reverse the inhibition of ACh release produced by systemically injected enkephalin, D-Met2-Pro5-enkephalinamide. 5 Effects of morphine and enkephalin on ACh release are discussed in terms of their action on difference opiate receptors.


Assuntos
Acetilcolina/metabolismo , Encéfalo/metabolismo , Endorfinas/farmacologia , Encefalina Metionina/análogos & derivados , Encefalinas/farmacologia , Morfina/farmacologia , Naloxona/farmacologia , Animais , Encefalinas/antagonistas & inibidores , Injeções Intraventriculares , Morfina/antagonistas & inibidores , Ratos
7.
Br J Pharmacol ; 50(1): 57-62, 1974 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-4856723

RESUMO

1 The spontaneous release of acetylcholine (ACh) from the cerebral cortex of control and morphine-dependent rats was investigated. The rate of resting output of ACh in morphine-dependent animals was lower than that in the control animals.2 Administration of naloxone and nalorphine to morphine-dependent rats was followed by a significant rise in the release of cortical ACh. In control rats no such increase in the release of ACh occurred after similar injections of narcotic antagonists.3 Injections of morphine produced a consistent decrease in the rate of spontaneous release of cortical ACh in the control rats, but similar injections in the dependent rats did not produce a decrease in the rate of cortical ACh release.4 The relevance of these results with regard to development of the narcotic abstinence syndrome is discussed.


Assuntos
Acetilcolina/metabolismo , Encéfalo/efeitos dos fármacos , Dependência de Morfina/metabolismo , Morfina/farmacologia , Animais , Encéfalo/metabolismo , Humanos , Morfina/antagonistas & inibidores , Nalorfina/farmacologia , Naloxona/farmacologia , Ratos
8.
Br J Pharmacol ; 108(4): 1137-42, 1993 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8485623

RESUMO

1. The effects of intrathecal (i.t.) administration of beta-endorphin and two shorter fragments, human and ovine beta-endorphin1-27, were examined for antinociceptive activity in the tail-flick and paw-pressure tests in the rat. Additionally, the ability of ovine beta-endorphin1-27 to influence the action of i.t. beta-endorphin, morphine and [D-Pen2-D-Pen5]enkephalin (DPDPE) was also examined in these tests. 2. After i.t. injection, beta-endorphin produced potent dose-related antinociception in the tail-flick and paw-pressure tests. Shorter endorphins produced much weaker effects. The order of antinociceptive efficacy was beta-endorphin > human beta-endorphin1-27 > ovine beta-endorphin1-27. 3. Administration of ovine beta-endorphin1-27 (0.72, 1.44 nmol, i.t.) significantly attenuated the antinociceptive effect of beta-endorphin (2.88 nmol, i.t.) in the tail-flick and paw-pressure tests. 4. Both i.t. morphine and DPDPE produced dose-related antinociception in the tail-flick and paw-pressure tests. The potency of DPDPE was lower than that of morphine in both tests; however, the effect of DPDPE was weaker in the paw-pressure test. 5. Administration of ovine beta-endorphin1-27 (1.44 nmol, i.t.) significantly attenuated the antinociceptive effect of morphine (14.9 nmol, i.t.) in both tests and the effect of DPDPE (38.7 nmol) in the tail-flick test. 6. The results show that beta-endorphin1-27 acts as an opioid antagonist at the spinal level in the rat. Its ability to inhibit the action of morphine and DPDPE suggests that it may attenuate beta-endorphin action by an interaction with mu- and/or delta-opioid receptors.


Assuntos
Analgésicos/antagonistas & inibidores , Antagonistas de Entorpecentes/farmacologia , Entorpecentes/farmacologia , Fragmentos de Peptídeos/farmacologia , Medula Espinal/fisiologia , beta-Endorfina/farmacologia , Analgésicos/farmacologia , Animais , Relação Dose-Resposta a Droga , D-Penicilina (2,5)-Encefalina , Encefalinas/farmacologia , Humanos , Injeções Espinhais , Masculino , Morfina/farmacologia , Medição da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Ovinos
9.
Br J Pharmacol ; 80(4): 727-34, 1983 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-6571228

RESUMO

Theophylline (Theo) and caffeine antagonized the inhibitory effect of methionine (Met)-enkephalin, leucine (Leu)-enkephalin and morphine on the twitch height of the field stimulated myenteric plexus longitudinal muscle (MPLM) preparation of the guinea-pig ileum. Antagonism by Theo was observed only in tissues stimulated submaximally, but that by caffeine was observed in tissues stimulated submaximally and supramaximally. Injection of Theo (20, 40 mg kg-1) or caffeine (40 mg kg-1) reversed or blocked the inhibitory effects of Leu-enkephalin (50 micrograms i.c.v.) and a systemically active enkephalin FK 33,824 (0.5 mg kg-1) on the release of acetylcholine (ACh) from the rat cortex in vivo. Injections of morphine (2.5, 5.0 mg kg-1 i.v.) did not modify the in vivo release of radiolabelled purines from the cerebral cortex prelabelled with [3H]-adenosine (2.8 X 10(-7) M). Application of K+ (60 mM) to the cortex readily stimulated this release. Injection of morphine (5.0 mg kg-1 i.v.) increased the spontaneous release of radiolabelled purines from the cortex prelabelled with a higher concentration of [3H]-adenosine (10(-4) M) in six out of eleven experiments. Under similar conditions neither Leu-enkephalin (50 micrograms i.c.v.) nor FK 33,824 (0.5 mg kg-1 i.v.) stimulated purine release. It is concluded that methylxanthines can antagonize the inhibitory action of opioids on the peripheral and central release of ACh. However, this antagonism does not reflect an intermediary purine step in the action of opioids on the release of ACh.


Assuntos
Acetilcolina/metabolismo , Adenosina/fisiologia , Encefalinas/antagonistas & inibidores , Xantinas/farmacologia , Adenosina/metabolismo , Animais , Cafeína/farmacologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Cobaias , Técnicas In Vitro , Morfina/antagonistas & inibidores , Contração Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Plexo Mientérico/efeitos dos fármacos , Ratos , Teofilina/farmacologia
10.
Br J Pharmacol ; 117(3): 493-501, 1996 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8821539

RESUMO

1. Neuropeptide FF (NPFF) has been shown to produce antinociceptive effects and enhance morphine-induced antinociception after intrathecal (i.t.) injection. In this study, the spinal effects of two NPFF analogues, -D-Tyr1,(NMe)Phe3-NPFF (1DMe) and [D-Tyr1,D-Leu2,D-Phe3]NPFF (3D), which are resistant to degradation and exhibit a high affinity for NPFF binding sites, were examined in tests of thermal and mechanical nociception. 2. 1DMe and 3D produced potent dose-dependent spinal antinociception in the tail-flick test. On a molar basis, 1DMe was 20 and 50 times more potent than 3D and morphine, respectively, and high doses of 1DMe and 3D produced a sustained antinociceptive effect without visible signs of motor impairment. 3. Spinal antinociceptive effects produced by 1DMe (0.86 nmol) or 3D (8.6 nmol) were significantly reduced by i.t. co-administration of naloxone (11 nmol) or i.t. pre-administration of D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH2 (CTOP, 9.25 nmol) or beta-funaltrexamine (beta-FNA, 2 nmol) or naltrindole (2.2 nmol). The doses of the mu-antagonists (CTOP and beta-FNA) or the delta-antagonist (naltrindole) used in 1DMe and 3D experiments blocked the antinociceptive effects of mu- or delta-receptor-selective agonists. 4. When administered in combination with antinociceptive doses of the mu-receptor agonist, morphine (13.2 nmol) or the delta-receptor agonist, [D-Ala2]deltorphin I (20 nmol), sub-effective dose of 1DMe or 3D (0.009 nmol) enhanced and prolonged the spinal effects of these opioid agonists. 5. The results of this study show that spinal mu- and delta-opioid receptors play a role in antinociception produced by NPFF analogues. These results also suggest a role for NPFF in modulation of nociceptive signals at the spinal level.


Assuntos
Analgésicos/farmacologia , Oligopeptídeos/farmacologia , Receptores Opioides/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Sequência de Aminoácidos , Analgésicos Opioides/farmacologia , Animais , D-Penicilina (2,5)-Encefalina , Encefalinas/farmacologia , Injeções Espinhais , Masculino , Dados de Sequência Molecular , Morfina/farmacologia , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Medição da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Somatostatina/análogos & derivados , Somatostatina/farmacologia
11.
Br J Pharmacol ; 127(3): 631-44, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10401553

RESUMO

1. This study examined the effects of the COX inhibitors, ketorolac and ibuprofen, and the NOS inhibitor L-NAME for their potential to both inhibit the development and reverse tolerance to the antinociceptive action of morphine. 2. Repeated administration of intrathecal morphine (15 micrograms), once daily, resulted in a progressive decline of antinociceptive effect and an increase in the ED50 value in the tailflick and paw pressure tests. Co-administration of ketorolac (30 and 45 micrograms) or S(+) ibuprofen (10 micrograms) with morphine (15 micrograms) prevented the decline of antinociceptive effect and increase in ED50 value. Similar treatment with L-NAME (100 micrograms) exerted weaker effects. Administration of S(+) but not R(-) ibuprofen (10 mg kg-1) had similar effects on systemic administration of morphine (15 mg kg-1). 3. Intrathecal or systemic administration of the COX or NOS inhibitors did not alter the baseline responses in either tests. Acute keterolac or S(+) ibuprofen also did not potentiate the acute actions of spinal or systemic morphine, but chronic intrathecal administration of these agents increased the potency of acute morphine. 4. In animals already tolerant to intrathecal morphine, subsequent administration of ketorolac (30 micrograms) with morphine (15 micrograms) partially restored the antinociceptive effect and ED50 value of acute morphine, reflecting the reversal of tolerance. Intrathecal L-NAME (100 micrograms) exerted a weaker effect. 5. These data suggest that spinal COX activity, and to a lesser extent NOS activity, contributes to the development and expression of opioid tolerance. Inhibition of COX may represent a useful approach for the prevention as well as reversal of opioid tolerance.


Assuntos
Analgésicos Opioides/farmacologia , Inibidores de Ciclo-Oxigenase/farmacologia , Inibidores Enzimáticos/farmacologia , Morfina/efeitos adversos , Morfina/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Animais , Tolerância a Medicamentos , Ibuprofeno/farmacologia , Injeções Intraperitoneais , Injeções Espinhais , Cetorolaco , Masculino , NG-Nitroarginina Metil Éster/farmacologia , Nociceptores/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Medula Espinal/efeitos dos fármacos , Medula Espinal/fisiologia , Tolmetino/análogos & derivados , Tolmetino/farmacologia
12.
Br J Pharmacol ; 131(5): 875-84, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11053206

RESUMO

This study examined the effects of the peptide CGRP receptor antagonist CGRP(8-37) and the newly-developed non-peptide CGRP receptor antagonist BIBN4096BS for their potential to both inhibit the development and reverse tolerance to the antinociceptive action of morphine. Repeated administration of intrathecal morphine (15 microg), once daily, produced a progressive decline of antinociceptive effect and an increase in the ED(50) value in the tailflick and paw pressure tests. Co-administration of CGRP(8-37) (4 microg) or BIBN4096BS (0.05, 0.1 microg) with morphine (15 microg) prevented the decline of antinociceptive effect and increase in ED(50) value in the tailflick test. Intrathecal administration of the CGRP receptor antagonists did not alter the baseline responses in either tests. Acute CGRP(8-37) also did not potentiate the acute actions of spinal morphine. In animals rendered tolerant to intrathecal morphine, subsequent administration of CGRP(8-37) (4 microg) with morphine (15 microg) partially restored the antinociceptive effect and ED(50) value of acute morphine, reflecting the reversal of tolerance. Animals tolerant to intrathecal morphine expressed increased CGRP and substance P-like immunostaining in the dorsal horn of the spinal cord. The increase in CGRP, but not substance P-like immunostaining, was blocked by a co-treatment with CGRP(8-37) (4 microg). In animals already tolerant to morphine, the increase in CGRP but not substance P-like immunostaining was partially reversed by CGRP(8-37) (4 microg). These data suggest that activation of spinal CGRP receptors contributes to both the development and expression of spinal opioid tolerance. CGRP receptor antagonists may represent a useful therapeutic approach for preventing as well as reversing opioid tolerance.


Assuntos
Antagonistas do Receptor do Peptídeo Relacionado ao Gene de Calcitonina , Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Morfina/farmacologia , Fragmentos de Peptídeos/farmacologia , Piperazinas/farmacologia , Quinazolinas/farmacologia , Animais , Peptídeo Relacionado com Gene de Calcitonina/análise , Tolerância a Medicamentos , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Peptídeo Relacionado com o Gene de Calcitonina/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Substância P/análise
13.
Eur J Pharmacol ; 49(3): 309-12, 1978 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-658146

RESUMO

The inhibitory effect of morphine on the release of acetylcholine (ACh) from the rat cerebral cortex was antagonized by theophylline, caffeine and 3-isobutyl-1-methylxanthine (IBMX). Theophylline antagonism of morphine action was dose related and this agent failed to influence a comparable action of chlorpromazine on the release of ACh. Intraventricular injection of calcium also antagonized the anti-release effect of morphine. Neither methylxanthines nor calcium modified the spontaneous release of ACh in the absence of morphine. It is suggested that methylxanthine antagonism of morphine action is selective and that it could be related to the ability of methylxanthines to mobilize bound calcium.


Assuntos
Acetilcolina/metabolismo , Encéfalo/metabolismo , Cálcio/farmacologia , Morfina/antagonistas & inibidores , Xantinas/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Cafeína/farmacologia , Clorpromazina/farmacologia , Ratos , Teofilina/farmacologia
14.
Eur J Pharmacol ; 203(1): 93-103, 1991 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-1797559

RESUMO

This investigation was carried out to study the development of physical dependence on spinally administered morphine, and it was determined if this phenomenon is associated with altered levels of [Met5]enkephalin. Morphine was infused continuously into the intrathecal space of rats for three or six days. In morphine-dependent animals, an intrathecal naloxone challenge produced increased reaction to nociceptive stimuli, hypertension, hyperthermia, decreased urinary output, and loss of body weight. Chronic spinal infusion of morphine alone failed to alter levels of [Met5]enkephalin in sacral and lumbar spinal cord. However, 24 h after the naloxone challenge, there was a significant increase in spinal enkephalin levels in morphine-dependent animals. It is concluded that spinal morphine treatment leads to the development of physical dependence. Certain characteristics of this phenomenon, as reflected in the naloxone-precipitated withdrawal signs, differ from those associated with dependence on systemic morphine.


Assuntos
Encefalina Metionina/metabolismo , Morfina/farmacologia , Síndrome de Abstinência a Substâncias/metabolismo , Analgésicos/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Temperatura Corporal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Química Encefálica/efeitos dos fármacos , Tolerância a Medicamentos , Injeções Espinhais , Masculino , Morfina/administração & dosagem , Naloxona/farmacologia , Ratos , Ratos Endogâmicos , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Urodinâmica/efeitos dos fármacos
15.
Eur J Pharmacol ; 237(1): 73-81, 1993 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-8102975

RESUMO

The effects of intrathecal injections of F8Famide (Phe-Leu-Phe-Gln-Pro-Gln-Arg-Phe-NH2, 0.05-17.5 nmol) and FMRF-amide (Phe-Met-Arg-Phe-NH2, 0.002-25 nmol), known as anti-opioid agents, were investigated by using noxious thermal (tail flick) and mechanical (paw pressure) tests in the rat. Both peptides produced significant long-lasting (24-48 h) analgesia in both tests without causing detectable motor dysfunction. Pretreatment with systemic naloxone (5.5 mumol/kg i.p.) attenuated the initial antinociceptive effects (first hour) induced by both peptides (8.8 nmol) in the tail flick test and only by FMRFamide in the paw pressure test. A subeffective dose of F8Famide (0.05 nmol) enhanced both the intensity and the duration of spinal morphine (6.6 nmol) analgesia in both tests. In contrast, a subanalgesic dose of FMRFamide (0.002 nmol) decreased the intensity and enhanced the duration of the effect of morphine. These results show that, besides acting as antinociceptive agents in the spinal cord, F8Famide and FMRFamide could differentially modulate spinal opioid functions.


Assuntos
Analgésicos/farmacologia , Neuropeptídeos/farmacologia , Neurotransmissores/farmacologia , Oligopeptídeos/farmacologia , Sequência de Aminoácidos , Analgésicos/administração & dosagem , Animais , Relação Dose-Resposta a Droga , FMRFamida , Injeções Espinhais , Masculino , Dados de Sequência Molecular , Morfina/farmacologia , Naloxona/farmacologia , Neuropeptídeos/administração & dosagem , Neurotransmissores/administração & dosagem , Oligopeptídeos/administração & dosagem , Medição da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos
16.
Eur J Pharmacol ; 406(3): 391-401, 2000 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-11040346

RESUMO

The neuropeptide FF (Phe-Leu-Phe-Gln-Pro-Gln-Arg-Phe-NH(2)) and its synthetic analogs bind to specific receptors in the spinal cord to produce antinociceptive effects that are partially attenuated by opioid antagonists, and at sub-effective doses neuropeptide FF receptor agonists augment spinal opioid antinociception. Since adenosine plays an intermediary role in the production of spinal opioid antinociception, this study investigated whether this purine has a similar role in the expression of spinal effects produced by neuropeptide FF receptor agonists. In rats bearing indwelling spinal catheters, injection of adenosine receptor agonists, N6-cyclohexyladenosine (CHA, 1.72 nmol) and N-ethylcarboxiamidoadenosine (NECA, 1.95 nmol), as well as morphine (13.2 nmol) elicited antinociception in the tail-flick and paw-pressure tests. Pretreatment with intrathecal 8-phenyltheophylline (5.9 and 11.7 nmol), an adenosine receptor antagonist, blocked the effect of all three agents without influencing baseline responses. Administration of two synthetic neuropeptide FF (NPFF) analogs, [D-Tyr(1),(NMe)Phe(3)]NPFF (1DMe, 0. 86 nmol) and [D-Tyr(1),D-leu(2),D-Phe(3)]NPFF (3D, 8.6 nmol) produced sustained thermal and mechanical antinociception. Pretreatment with doses of intrathecal 8-phenyltheophylline (5.9, 11. 7 and 23.5 nmol), producing adenosine receptor blockade, significantly inhibited the antinociceptive effects of 1DMe or 3D. Injection of a sub-antinociceptive dose of 1DMe (0.009 nmol) significantly augmented the antinociceptive effect of intrathecal morphine (13.2 nmol) in the tail-flick and paw-pressure tests. Intrathecal 8-phenyltheophylline (11.7 nmol) reduced the effect of this combination. Administration of low dose of 1DMe (0.009 nmol) or 3D (0.009 nmol) very markedly potentiated the antinociceptive actions of the adenosine receptor agonist, N6-cyclohexyladenosine (0. 43, 0.86 and 1.72 nmol) in the tail-flick and paw-pressure tests 50 min after injection. The results suggest that the antinociceptive and morphine modulatory effects resulting from activation of spinal NPFF receptors could be due to an increase in the actions or availability of adenosine.


Assuntos
Adenosina/fisiologia , Analgésicos Opioides/farmacologia , Morfina/farmacologia , Oligopeptídeos/farmacologia , Medula Espinal/efeitos dos fármacos , Adenosina/análogos & derivados , Adenosina/farmacologia , Animais , Masculino , Ratos , Ratos Sprague-Dawley , Teofilina/análogos & derivados , Teofilina/farmacologia
18.
Br J Pharmacol ; 155(8): 1264-78, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18806811

RESUMO

BACKGROUND AND PURPOSE: Ultra-low doses of opioid receptor antagonists augment spinal morphine antinociception and block the induction of tolerance. Considering the evidence demonstrating functional and physical interactions between the opioid and alpha(2)-adrenoceptors, this study investigated whether ultra-low doses of alpha(2)-adrenoceptor antagonists also influence spinal morphine analgesia and tolerance. EXPERIMENTAL APPROACH: Effects of low doses of the competitive alpha(2)-adrenoceptor antagonists-atipamezole (0.08, 0.8 ng), yohimbine (0.02, 2 ng), mirtazapine (0.02 ng) and idazoxan (0.08 ng) were investigated on intrathecal morphine analgesia, as well as acute and chronic morphine antinociceptive tolerance using the rat tail flick and paw pressure tests. KEY RESULTS: At doses markedly lower than those producing alpha(2)-adrenoceptor blockade, atipamezole, yohimbine, mirtazapine and idazoxan, prolonged the antinociceptive effects of morphine. When co-administered with repeated acute spinal injections of morphine, all four agents blocked the induction of acute tolerance. Co-injection of atipamezole with morphine for 5 days inhibited the development of tolerance in a chronic treatment paradigm. Spinal administration of atipamezole also reversed established antinociceptive tolerance to morphine as indicated by the restoration of morphine antinociceptive potency. The effects of atipamezole on spinal morphine tolerance were not influenced by treatment with 6-hydroxydopamine. CONCLUSIONS AND IMPLICATIONS: Low doses of competitive alpha(2)-adrenoceptor antagonists can augment acute morphine analgesia and block or reverse tolerance to spinal administration of morphine. These actions are interpreted in terms of their interaction with an opioid-alpha(2)-adrenoceptor complex, whose activity may have a function in the genesis of analgesic tolerance.


Assuntos
Antagonistas de Receptores Adrenérgicos alfa 2 , Antagonistas Adrenérgicos alfa/farmacologia , Tolerância a Medicamentos , Morfina/administração & dosagem , Coluna Vertebral , Antagonistas Adrenérgicos alfa/administração & dosagem , Animais , Comportamento Animal , Relação Dose-Resposta a Droga , Idazoxano/administração & dosagem , Idazoxano/farmacologia , Imidazóis/administração & dosagem , Imidazóis/farmacologia , Masculino , Mianserina/administração & dosagem , Mianserina/análogos & derivados , Mianserina/farmacologia , Mirtazapina , Ratos , Ratos Sprague-Dawley , Ioimbina/administração & dosagem , Ioimbina/farmacologia
19.
Can J Physiol Pharmacol ; 59(3): 307-10, 1981 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7194726

RESUMO

Eseroline, an eserine derivative without anticholinesterase activity, was tested in several systems for opiatelike activity. Eseroline depressed the twitch of the field-stimulated guinea pig ileum myenteric plexus longitudinal muscle preparation but failed to depress the twitch of the rat vas deferens. Intraperitoneal injections of eseroline in rats induced naloxone-antagonizable analgesia and catalepsy. Eseroline failed to influence the release of acetylcholine from the cortex of anesthetized rats. These observations have implications for studies in which eserine is used as a pharmacological tool.


Assuntos
Indóis/farmacologia , Entorpecentes , Acetilcolina/metabolismo , Analgésicos , Animais , Catalepsia/induzido quimicamente , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Cobaias , Humanos , Indóis/antagonistas & inibidores , Masculino , Contração Muscular/efeitos dos fármacos , Naloxona/farmacologia , Ratos
20.
Can J Physiol Pharmacol ; 64(3): 263-8, 1986 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-2871911

RESUMO

The effect of intrathecal injections of dynorphin1-8 (DYN1-8), dynorphin1-13 (DYN1-13), and a putative kappa agonist, U50,488 was tested in the rat tail-flick test. DYN1-8 and DYN1-13 (5, 10, 20 micrograms) produced a dose-related biphasic antinociceptive response consisting of an initial and a delayed response. Injection of U50,488 (20, 40 60 micrograms) produced a monophasic response. The antinociceptive effect of DYN1-8 (5, 10, 20 micrograms) and DYN1-13 (20 micrograms), was present 24 h postintrathecal injection. Pretreatment with systemic naloxone (2 mg/kg s.c.) attenuated the delayed response, but not the initial response induced by DYN1-8 and DYN1-13. The initial response was attenuated by pretreatment with intrathecal naloxone at a dose of 0.5 and 2.0 micrograms. The antinociceptive effect of U50,488 (20, 60 micrograms) was not affected by pretreatment with 2.0 micrograms intrathecal naloxone, but was significantly reduced by 4 micrograms of the antagonist. Both DYN1-8 and DYN1-13 (5 micrograms) augmented the antinociceptive effect of intrathecally administered morphine (5, 10 micrograms). Intrathecal injection of DYN1-8 (5, 10, 20 micrograms), DYN1-13 (5 micrograms), and morphine (10 micrograms) reduced the spontaneous output of urine measured at 2 and 24 h postintrathecal injection. A similar injection of U50,488 (20 micrograms) had no significant action on the urinary output. The results show that long and short dynorphin fragments have a comparable activity and the spinal antinociceptive actions of dynorphin are sensitive to low doses of intrathecal naloxone. The activity profile of spinally administered dynorphins differs from that of the kappa agonist U50,488.


Assuntos
Analgésicos , Dinorfinas/farmacologia , Fragmentos de Peptídeos/farmacologia , Pirrolidinas/farmacologia , Medula Espinal/efeitos dos fármacos , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida , Animais , Relação Dose-Resposta a Droga , Injeções Espinhais , Masculino , Morfina/farmacologia , Naloxona/farmacologia , Ratos , Ratos Endogâmicos , Tempo de Reação/efeitos dos fármacos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa