Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuropharmacology ; 140: 302-309, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-30086291

RESUMO

Opioid neurotransmission has been implicated in psychiatric disorders featuring impaired control over appetitive motivation, such as addiction and binge-eating disorder. We have previously shown that infusions of the µ-opioid receptor (µOR) agonist DAMGO into the ventromedial prefrontal cortex (vmPFC) induced hyperphagia, increased motor activity, and augmented sucrose-reinforced responding in the task progressive ratio (PR) task, which assesses the motivational value of an incentive. These effects were not reproduced by intra-PFC infusion of a variety of dopamine (DA) agonists and antagonists, suggesting that manipulation of intra-PFC DA systems alone is not sufficient to reproduce µOR-like effects. Nevertheless, this does not rule out interactions between PFC DA and µ-opioid systems. Here we used intra-vmPFC drug cocktails containing DAMGO and SCH 23390 (a DA D1 receptor antagonist) to determine whether increases in appetitive motivation and motor activity elicited by intra-vmPFC µOR stimulation require intact signaling through vmPFC D1 receptors. Blockade of D1 receptors with SCH 23390 attenuated the enhancement of PR breakpoint, and increases in exploratory-like behavior and feeding initiation elicited by intra-vmPFC µOR stimulation. These results establish that intra-vmPFC D1 signaling is required for the expression of behavioral effects evoked by µOR stimulation within the PFC, and further suggest that D1 tone plays an enabling or permissive role in the expression of µOR -elicited effects. Simultaneous targeting of both µ-opioid and D1 systems may represent a more efficacious treatment strategy (compared to µOR blockade alone) for psychiatric disorders characterized by dysregulated appetitive motivation.


Assuntos
Motivação/fisiologia , Córtex Pré-Frontal/metabolismo , Receptores de Dopamina D1/fisiologia , Receptores Opioides mu/fisiologia , Transdução de Sinais , Animais , Benzazepinas/farmacologia , Condicionamento Operante/efeitos dos fármacos , Condicionamento Operante/fisiologia , Sinergismo Farmacológico , Ala(2)-MePhe(4)-Gly(5)-Encefalina/antagonistas & inibidores , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Masculino , Microinjeções , Motivação/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Ratos , Receptores de Dopamina D1/antagonistas & inibidores , Receptores Opioides mu/agonistas , Esquema de Reforço , Transdução de Sinais/efeitos dos fármacos
2.
J Psychopharmacol ; 27(3): 302-11, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23348755

RESUMO

Classically, opioids produce their effects by activating Gi-proteins that inhibit adenylate cyclase activity. Previous studies proposed that mu-opioid receptors can also stimulate adenylate cyclase due to an initial transient coupling to Gs-proteins. Treatment with ultra-low doses of the nonselective opioid antagonist (-)-naloxone or its inactive enantiomer (+)-naloxone blocks this excitatory effect and enhances Gi-coupling. Previously we reported that infusion of the mu-opioid receptor agonist [D-Ala2, N-Me-Phe4, Glycinol5]-Enkephalin (DAMGO) into the mu-opioid receptor expressing lateral parabrachial nucleus increases feeding. Pretreatment with (-)-naloxone blocks this effect. We used this parabrachial circuit as a model to assess cellular actions of ultra-low doses of (-)-naloxone and (+)-naloxone in modifying the effects of DAMGO. Our results showed that an ultra-low concentration of (-)-naloxone (0.001 nM) and several concentrations of (+)-naloxone (0.01-10 nM) enhanced DAMGO-stimulated guanosine-5'-0-(γ-thio)-triphosphate incorporation in parabrachial sections in vitro. Further, we analyzed the relevance of these effects in vivo. In the present study, we show that (+)-naloxone can potentiate DAMGO-induced feeding at doses at which (-)-naloxone was an antagonist. These results implicated (+)-naloxone as a novel tool for studying mu-opioid receptor functions and suggest that (+)-naloxone may have therapeutic value to enhance clinical actions of opiate drugs.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/agonistas , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Neurônios/efeitos dos fármacos , Ponte/efeitos dos fármacos , Receptores Opioides mu/agonistas , Transdução de Sinais/efeitos dos fármacos , Analgésicos Opioides/agonistas , Analgésicos Opioides/antagonistas & inibidores , Analgésicos Opioides/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Sinergismo Farmacológico , Ingestão de Alimentos/efeitos dos fármacos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/agonistas , Ala(2)-MePhe(4)-Gly(5)-Encefalina/antagonistas & inibidores , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Técnicas In Vitro , Masculino , Proteínas do Tecido Nervoso/agonistas , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Concentração Osmolar , Ponte/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/metabolismo , Estereoisomerismo
3.
J Neurophysiol ; 108(9): 2393-404, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22956800

RESUMO

Respiratory depression is a therapy-limiting side effect of opioid analgesics, yet our understanding of the brain circuits mediating this potentially lethal outcome remains incomplete. Here we studied the contribution of the rostral ventromedial medulla (RVM), a region long implicated in pain modulation and homeostatic regulation, to opioid-induced respiratory depression. Microinjection of the µ-opioid agonist DAMGO in the RVM of lightly anesthetized rats produced both analgesia and respiratory depression, showing that neurons in this region can modulate breathing. Blocking opioid action in the RVM by microinjecting the opioid antagonist naltrexone reversed the analgesic and respiratory effects of systemically administered morphine, showing that this region plays a role in both the analgesic and respiratory-depressant properties of systemically administered morphine. The distribution of neurons directly inhibited by RVM opioid microinjection was determined with a fluorescent opioid peptide, dermorphin-Alexa 594, and found to be concentrated in and around the RVM. The non-opioid analgesic improgan, like DAMGO, produced antinociception but, unlike DAMGO, stimulated breathing when microinjected into the RVM. Concurrent recording of RVM neurons during improgan microinjection showed that this agent activated RVM ON-cells, OFF-cells, and NEUTRAL-cells. Since opioids are known to activate OFF-cells but suppress ON-cell firing, the differential respiratory response to these two analgesic drugs is best explained by their opposing effects on the activity of RVM ON-cells. These findings show that pain relief can be separated pharmacologically from respiratory depression and identify RVM OFF-cells as important central targets for continued development of potent analgesics with fewer side effects.


Assuntos
Analgésicos Opioides/toxicidade , Bulbo/efeitos dos fármacos , Neurônios/fisiologia , Dor Nociceptiva/fisiopatologia , Insuficiência Respiratória/induzido quimicamente , Analgésicos Opioides/antagonistas & inibidores , Animais , Ala(2)-MePhe(4)-Gly(5)-Encefalina/antagonistas & inibidores , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Potenciais Evocados/efeitos dos fármacos , Potenciais Evocados/fisiologia , Masculino , Bulbo/citologia , Bulbo/fisiologia , Morfina/antagonistas & inibidores , Morfina/farmacologia , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Neurônios/efeitos dos fármacos , Nociceptividade/efeitos dos fármacos , Nociceptividade/fisiologia , Ratos , Ratos Sprague-Dawley , Insuficiência Respiratória/fisiopatologia
4.
J Leukoc Biol ; 89(2): 301-9, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20952659

RESUMO

Opioid receptor agonists induce broad immunomodulatory activity, which substantially alters host defense and the inflammatory response. Previous studies have shown that the MOR selective agonist DAMGO has the capacity to increase the expression of the proinflammatory chemokines CCL2, CCL5, and CXCL10 in human PBMCs. NF-κB is a transcription factor that plays a pivotal role in innate and adaptive immune responses. We report that NF-κB is a vital player in the DAMGO-induced, MOR-mediated regulation of chemokine expression. Results show that NF-κB inhibitors prevent the induction of CCL2 expression in response to DAMGO administration and that the NF-κB subunit, p65, is phosphorylated at serine residues 311 and 536 in response to MOR activation. Furthermore, we demonstrate that PKCζ is phosphorylated following DAMGO-induced MOR activation, and this kinase is essential for NF-κB activation as well as CCL2 expression and transcriptional activity. Finally, ChIP analysis shows that DAMGO administration induces binding of p65 to the enhancer region of the CCL2 promoter. These data are consistent with the notion that MOR activation promotes a proinflammatory response, which involves NF-κB activation. Our results also suggest a significant and novel role for PKCζ as an essential participant in the MOR-mediated regulation of proinflammatory chemokine expression.


Assuntos
Quimiocina CCL2/biossíntese , NF-kappa B/metabolismo , Proteína Quinase C/fisiologia , Receptores Opioides mu/biossíntese , Aldeídos/farmacologia , Analgésicos Opioides/farmacologia , Células Cultivadas , Quimiocina CCL2/antagonistas & inibidores , Quimiocina CCL2/genética , Ala(2)-MePhe(4)-Gly(5)-Encefalina/antagonistas & inibidores , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Células HEK293 , Humanos , Leupeptinas/farmacologia , NF-kappa B/antagonistas & inibidores , NF-kappa B/fisiologia , Nitrilas/farmacologia , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Receptores Opioides mu/agonistas , Receptores Opioides mu/fisiologia , Sulfonas/farmacologia
5.
Eur J Pharmacol ; 649(1-3): 336-41, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20863827

RESUMO

Pruritus is a common adverse effect of opioid treatment. However, the mechanism by which pruritus is induced by opioid administration is unclear. In this study, we examined the effects of the intradermal injection of loperamide, a peripherally restricted opioid receptor agonist, on the itch sensation. When injected intradermally into the rostral part of the back in mice, loperamide elicited scratching behavior. We also examined the effects of the selective mu opioid receptor agonist [d-Ala², N-Me-Phe4, Gly5-ol]-enkephalin acetate (DAMGO), the selective delta opioid receptor agonist [d-Pen(2,5)]-enkephalin (DPDPE), and the selective kappa opioid receptor agonist U-50488H on scratching behavior in mice in order to determine which subtype is involved in opioid-induced pruritus. Following intradermal injection into the rostral part of the back in mice, DAMGO elicited scratching behavior, while DPDPE and U-50488H did not. This suggests that peripheral mu opioid activation elicits the itch sensation. Next, we focused on the treatment of opioid-induced itch sensation without central adverse effects. Naloxone methiodide is a peripherally restricted opioid receptor antagonist. In the present study, naloxone methiodide significantly suppressed scratching behavior induced by loperamide and DAMGO. These findings suggest that mu opioid receptors play a primary role in peripheral pruritus and that naloxone methiodide may represent a possible remedy for opioid-induced itching.


Assuntos
Comportamento Animal , Prurido , Receptores Opioides mu/fisiologia , Pele/efeitos dos fármacos , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/uso terapêutico , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/toxicidade , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/antagonistas & inibidores , Analgésicos Opioides/farmacologia , Analgésicos Opioides/toxicidade , Animais , Antipruriginosos/administração & dosagem , Antipruriginosos/farmacologia , Antipruriginosos/uso terapêutico , Comportamento Animal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Ala(2)-MePhe(4)-Gly(5)-Encefalina/antagonistas & inibidores , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/toxicidade , D-Penicilina (2,5)-Encefalina/administração & dosagem , D-Penicilina (2,5)-Encefalina/farmacologia , D-Penicilina (2,5)-Encefalina/toxicidade , Injeções Intradérmicas , Loperamida/administração & dosagem , Loperamida/antagonistas & inibidores , Loperamida/farmacologia , Loperamida/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos ICR , Naloxona/administração & dosagem , Naloxona/análogos & derivados , Naloxona/farmacologia , Naloxona/uso terapêutico , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Prurido/induzido quimicamente , Prurido/tratamento farmacológico , Compostos de Amônio Quaternário/administração & dosagem , Compostos de Amônio Quaternário/farmacologia , Compostos de Amônio Quaternário/uso terapêutico , Receptores Opioides delta/agonistas , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inibidores , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores
6.
Respir Physiol Neurobiol ; 172(3): 94-105, 2010 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-20444435

RESUMO

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


Assuntos
Analgésicos Opioides/antagonistas & inibidores , Analgésicos Opioides/farmacologia , Inibidores da Fosfodiesterase 4 , Inibidores de Fosfodiesterase/farmacologia , Insuficiência Respiratória/induzido quimicamente , Insuficiência Respiratória/prevenção & controle , Xantinas/farmacologia , Antagonistas do Receptor A1 de Adenosina , Antagonistas do Receptor A2 de Adenosina , Animais , Animais Recém-Nascidos , Cafeína/farmacologia , Eletrofisiologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/antagonistas & inibidores , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Fentanila/antagonistas & inibidores , Fentanila/farmacologia , Técnicas In Vitro , Interneurônios/efeitos dos fármacos , Rede Nervosa/efeitos dos fármacos , Rede Nervosa/fisiologia , Antagonistas de Receptores Purinérgicos P1 , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Receptores Opioides mu/efeitos dos fármacos , Fenômenos Fisiológicos Respiratórios/efeitos dos fármacos , Rolipram/farmacologia , Teofilina/farmacologia
7.
Am J Physiol Regul Integr Comp Physiol ; 295(1): R243-51, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18480245

RESUMO

We have previously shown that ionotropic glutamate receptors in the caudal portion of the nucleus tractus solitarii (NTS), especially in the commissural NTS, play a prominent role in the mediation of tracheobronchial cough and that substance P potentiates this reflex. This NTS region could be a site of action of some centrally acting antitussive agents and a component of a drug-sensitive gating mechanism of cough. To address these issues, we investigated changes in baseline respiratory activity and cough responses to tracheobronchial mechanical stimulation following microinjections (30-50 nl) of centrally acting antitussive drugs into the caudal NTS of pentobarbitone-anesthetized, spontaneously breathing rabbits. [D-Ala2,N-Me-Phe4,Gly5-ol]-enkephalin (DAMGO) and baclofen decreased baseline respiratory frequency because of increases in the inspiratory time only at the higher concentration employed (5 mM and 1 mM, respectively). DAMGO (0.5 mM) and baclofen (0.1 mM) significantly decreased cough number, peak abdominal activity, peak tracheal pressure, and increased cough-related total cycle duration. At the higher concentrations, these agents suppressed the cough reflex. The effects of these two drugs were counteracted by specific antagonists (10 mM naloxone and 25 mM CGP-35348, respectively). The neurokinin-1 (NK1) receptor antagonist CP-99,994 (10 mM) abolished cough responses, whereas the NK2 receptor antagonist MEN 10376 (5 mM) had no effect. The results indicate that the caudal NTS is a site of action of some centrally acting drugs and a likely component of a neural system involved in cough regulation. A crucial role of substance P release in the mediation of reflex cough is also suggested.


Assuntos
Antitussígenos/farmacologia , Baclofeno/farmacologia , Tosse/tratamento farmacológico , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Reflexo/efeitos dos fármacos , Núcleo Solitário/efeitos dos fármacos , Animais , Antitussígenos/antagonistas & inibidores , Baclofeno/administração & dosagem , Baclofeno/antagonistas & inibidores , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Ala(2)-MePhe(4)-Gly(5)-Encefalina/antagonistas & inibidores , Masculino , Naloxona/farmacologia , Neurocinina A/análogos & derivados , Neurocinina A/farmacologia , Compostos Organofosforados/farmacologia , Fragmentos de Peptídeos/farmacologia , Piperidinas/farmacologia , Coelhos , Núcleo Solitário/fisiologia
8.
Naunyn Schmiedebergs Arch Pharmacol ; 372(3): 213-9, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16283255

RESUMO

The stimulation of peripheral opioid receptors yields analgesic responses in a model of bone cancer-induced pain in mice. In order to know the type(s) of peripheral opiate receptors involved, the paw thermal withdrawal latencies were measured in C3H/HeJ mice bearing a tibial osteosarcoma, after administering selective agonists of mu-,delta-and kappa-opiate receptors. The peritumoral administration of DAGO (0.6-6 microg) inhibited the osteosarcoma-induced hyperalgesia at doses ineffective in healthy animals, the highest one even increasing the withdrawal latencies over the control values. Naloxone-methiodide (2 mg/kg) and cyprodime (1 mg/kg), but not naltrindole (0.1 mg/kg) nor nor-binaltorphimine (10 mg/kg), antagonized DAGO-induced analgesic effects, these therefore probably being mediated through peripheral mu-opioid receptors. The peritumoral injection of DPDPE (100 microg) induced analgesia which was inhibited by naloxone-methiodide and naltrindole but not by nor-binaltorphimine. Cyprodime partially antagonized the analgesia induced by 100 microg of DPDPE, but did not modify the effect induced by 30 microg of this agonist-a dose that restores the hyperalgesic latencies up to the control values. The antihyperalgesic effect induced by the peritumoral administration of U-50,488H (1 microg) was antagonized by naloxone-methiodide and nor-binaltorphimine, but not by cyprodime nor naltrindole, thus suggesting the involvement of peripheral kappa-opioid receptors. In conclusion, the stimulation of peripheral mu-, delta- and kappa-opioid receptors is a pharmacological strategy useful for relieving this experimental type of bone cancer-induced pain, the greatest analgesic effect being achieved by stimulating peripheral mu-opioid receptors.


Assuntos
Analgésicos Opioides/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Hiperalgesia/tratamento farmacológico , Receptores Opioides/agonistas , Analgésicos Opioides/antagonistas & inibidores , Animais , Neoplasias Ósseas/complicações , Interações Medicamentosas , Ala(2)-MePhe(4)-Gly(5)-Encefalina/antagonistas & inibidores , Hiperalgesia/etiologia , Camundongos , Morfinanos/farmacologia , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Osteossarcoma/complicações
9.
Brain Res ; 1018(1): 78-85, 2004 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-15262208

RESUMO

Reward is an important factor motivating food intake in satiated animals. Two sites involved in the reward response are the ventral tegmental area (VTA) and the nucleus accumbens shell region (sNAcc), between which communication is partially regulated by opioids and dopamine (DA). Previous studies have shown that the mu-opioid agonist Tyr-D-Ala-Gly-MePhe-Gly(ol)-enkephalin (DAMGO) dose-dependently enhances food intake in satiated animals when injected into either the VTA or the sNAcc. The enhanced intake elicited by DAMGO injected into the sNAcc was dose-dependently blocked by injection of naltrexone (NTX) bilaterally into the VTA, indicating an opioid-dependent signaling pathway from the sNAcc to the VTA in mediation of food intake. In the present study, we cannulated animals bilaterally in both the VTA and the sNAcc to further study the nature of opioid- and DA-dependent communication between the sites. Food intake elicited by DAMGO (2 or 5 nmol) injected unilaterally into the VTA was dose-dependently diminished by bilateral injection of NTX (2.5, 5, and 25 g/side) or the D1 antagonist SCH 23390 (3, 1, 0.3, 0.15, 0.05, and 0.015 nmol/side) into the sNAcc. When DAMGO (5 nmol) was injected into the sNAcc, the resulting food intake was decreased by doses of SCH 23390 ranging from 0.05 to 100 nmol/side injected bilaterally into the VTA, but not by equimolar doses of Raclopride, a D2 antagonist. These results, combined with previous findings, suggest a signaling pathway between the VTA and the sNAcc in which opioids and DA facilitate feeding in an interdependent manner.


Assuntos
Dopamina/metabolismo , Ingestão de Alimentos/fisiologia , Vias Neurais/metabolismo , Núcleo Accumbens/metabolismo , Peptídeos Opioides/metabolismo , Área Tegmentar Ventral/metabolismo , Animais , Antagonistas de Dopamina/farmacologia , Interações Medicamentosas/fisiologia , Ingestão de Alimentos/efeitos dos fármacos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/antagonistas & inibidores , Comportamento Alimentar/efeitos dos fármacos , Comportamento Alimentar/fisiologia , Masculino , Antagonistas de Entorpecentes/farmacologia , Vias Neurais/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de Dopamina D1/efeitos dos fármacos , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2 , Receptores Opioides/efeitos dos fármacos , Receptores Opioides/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Área Tegmentar Ventral/efeitos dos fármacos
10.
Br J Pharmacol ; 140(4): 691-700, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14534152

RESUMO

In the intestine, opioids produce antidiarrhoeal and constipating actions that are mediated by enteric neurones. Through interactions with opioid receptors (ORs) on submucosal neurones, opioids suppress active ion transport evoked by transmural electrical stimulation (TES) in mucosa-submucosa sheets from the porcine ileum. In this study, we examined the pharmacological characteristics of the previously described OR, which is sensitive to the delta1-OR antagonist 7-benzylidenenaltrexone and modulates neurogenic transepithelial ion transport in this tissue preparation. Increases in short-circuit current (Isc, a measure of active anion transport) evoked by TES in ileal mucosa-submucosa sheets were inhibited by opioid agonists possessing high selectivity for either delta- or micro-ORs including [d-Pen2,5]enkephalin (DPDPE), [d-Ala2, Glu4]deltorphin II, and [d-Ala2, N-Me-Phe4, Gly5-ol]enkephalin (DAMGO). As determined by the Schild analysis, the actions of these agonists were competitively inhibited by 7-benzylidenenaltrexone. The nonequilibrium micro-OR antagonist beta-funaltrexamine inhibited the actions of DAMGO only at a high concentration (1 microm) but did not alter DPDPE or deltorphin II action. At concentrations up to 10 microm, the nonequilibrium delta-OR antagonist naltrindole 5'-isothiocyanate did not alter the actions of delta- or micro-OR agonists. Radioligand binding analyses of neuronal homogenates from the ileal submucosa revealed that the nonselective OR ligand [3H]diprenorphine bound to two populations of specific binding sites. One of these sites possessed binding characteristics similar to the delta-OR. In summary, neurogenic ion transport in the porcine intestine is modulated by an OR which shares pharmacological characteristics of both micro- and delta-ORs and may represent a novel receptor entity.


Assuntos
Compostos de Benzilideno/farmacologia , Íleo/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Receptores Opioides delta/efeitos dos fármacos , Proteínas de Anfíbios , Animais , Ânions/antagonistas & inibidores , Ânions/farmacocinética , Benzamidas/farmacologia , Transporte Biológico Ativo/efeitos dos fármacos , Transporte Biológico Ativo/fisiologia , Proteínas de Transporte/efeitos dos fármacos , Diprenorfina/antagonistas & inibidores , Diprenorfina/metabolismo , Diprenorfina/farmacologia , Relação Dose-Resposta a Droga , Estimulação Elétrica/métodos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/antagonistas & inibidores , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , D-Penicilina (2,5)-Encefalina/antagonistas & inibidores , D-Penicilina (2,5)-Encefalina/farmacologia , Feminino , Motilidade Gastrointestinal , Íleo/citologia , Íleo/inervação , Mucosa Intestinal/citologia , Masculino , Neurônios Aferentes/fisiologia , Oligopeptídeos/antagonistas & inibidores , Oligopeptídeos/farmacologia , Piperazinas/farmacologia , Quinolinas/farmacologia , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides mu/efeitos dos fármacos , Suínos , Trítio
11.
J Pharmacol Exp Ther ; 307(3): 1221-6, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14534368

RESUMO

Compound (+)-3-((alpha-R)-alpha-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-hydroxybenzyl)-N-(3-fluorophenyl)-N-methylbenzamide (DPI-3290), is one of a series of novel centrally acting agents with potent antinociceptive activity that binds specifically and with high affinity to opioid receptors. In saturation equilibrium binding studies performed at 25 degrees C using membranes from rat brain or guinea pig cerebellum, the Ki values measured for DPI-3290 at delta-, mu-, and kappa-opioid receptors were 0.18 +/- 0.02, 0.46 +/- 0.05, and 0.62 +/- 0.09 nM, respectively. In vas deferens isolated from laboratory mice, DPI-3290 decreased electrically induced tension development in a concentration-dependent manner with corresponding IC50 values of 1.0 +/- 0.3, 6.2 +/- 2.0, and 25.0 +/- 3.3 nM at delta-, mu-, and kappa-receptors, respectively. The activity of DPI-3290 in isolated vas deferens tissue was approximately 20,000, 175.8, and 1500 times more efficacious than morphine, and 492, 2.5, and 35 times more efficacious than fentanyl at delta-, mu-, and kappa-receptors, respectively. In ileal strips isolated from guinea pigs, DPI-3290 inhibited tension development with a corresponding IC50 value of 3.4 +/- 1.6 nM at mu-opioid receptors and 6.7 +/- 1.6 nM at kappa-opioid receptors. Intravenous administration of 0.05 +/- 0.007 mg/kg DPI-3290 produced a 50% antinociceptive response in rats. The antinociceptive properties of DPI-3290 were blocked by naloxone (0.5 mg/kg s.c.). Compared with morphine, this study demonstrated that DPI-3290 is more potent and elicited a similar magnitude of antinociceptive activity in the rat, actions mediated by its mixed opioid receptor agonist activity. The marked antinociceptive activity of DPI-3290 will likely provide a means for relieving severe pain in patients that require analgesic treatment.


Assuntos
Analgésicos/farmacologia , Benzamidas/farmacologia , Entorpecentes/agonistas , Piperazinas/farmacologia , Animais , Benzamidas/antagonistas & inibidores , Benzenoacetamidas/antagonistas & inibidores , Benzenoacetamidas/farmacologia , Ligação Competitiva/efeitos dos fármacos , Gasometria , Química Encefálica , Ala(2)-MePhe(4)-Gly(5)-Encefalina/antagonistas & inibidores , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , D-Penicilina (2,5)-Encefalina/antagonistas & inibidores , D-Penicilina (2,5)-Encefalina/farmacologia , Cobaias , Íleo/efeitos dos fármacos , Masculino , Músculo Liso/efeitos dos fármacos , Antagonistas de Entorpecentes/farmacologia , Medição da Dor/efeitos dos fármacos , Piperazinas/antagonistas & inibidores , Pirrolidinas/antagonistas & inibidores , Pirrolidinas/farmacologia , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/efeitos dos fármacos , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/efeitos dos fármacos , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/efeitos dos fármacos , Receptores Opioides mu/metabolismo , Estereoisomerismo , Ducto Deferente/efeitos dos fármacos
12.
J Neurosci ; 23(19): 7262-8, 2003 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-12917359

RESUMO

Glutamate neurotransmission plays an important role in the processing of pain and in chronic opioid-induced neural and behavioral plasticity, such as opioid withdrawal and opioid dependence. Kappa-opioid receptors also have been implicated in acute opioid modulation of pain and chronic opioid-induced plasticity, both of which are primarily mediated by mu-opioid receptors. Using whole-cell patch clamp recordings in brain slices in vitro and system analysis of pain behaviors in rats in vivo, this study investigated the functional role of glutamate synaptic transmission and kappa-opioid receptors in two behavioral pain conditions: m-opioid-induced analgesia (decreased pain) and mu-opioid withdrawal-induced hyperalgesia (increased pain). In the nucleus raphe magnus (NRM), a brainstem structure that controls spinal pain transmission, we found that kappa-receptor agonists presynaptically inhibited glutamate synaptic currents in both of the two cell types that are thought to respectively inhibit or facilitate spinal pain transmission. In rats, both glutamate receptor antagonists and the kappa agonist microinjected into the NRM attenuated mu-opioid-induced analgesia, which is most likely mediated through activation of such pain-inhibiting neurons. However, during opioid abstinence-induced withdrawal, the same doses of glutamate receptor antagonists and the kappa agonist administered in the NRM suppressed the withdrawal-induced hyperalgesia, which is thought to be mediated by activation of those pain-facilitating neurons during opioid withdrawal. These results demonstrate that kappa-opioid receptors antagonize mu-receptor-induced effects in both analgesic and hyperalgesic states, and suggest inhibition of glutamate synaptic transmission as a presynaptic mechanism for the kappa antagonism of these two mu receptor-mediated actions.


Assuntos
Analgésicos Opioides/antagonistas & inibidores , Benzenoacetamidas , Antagonistas de Aminoácidos Excitatórios/farmacologia , Hiperalgesia/etiologia , Receptores Opioides kappa/agonistas , Transmissão Sináptica/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Células Cultivadas , Dinorfinas/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/antagonistas & inibidores , Potenciais Pós-Sinápticos Excitadores , Ácido Glutâmico/fisiologia , Masculino , Técnicas de Patch-Clamp , Pirrolidinas/farmacologia , Núcleos da Rafe/efeitos dos fármacos , Núcleos da Rafe/fisiologia , Ratos , Ratos Wistar , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Receptores Pré-Sinápticos/agonistas
13.
J Pharmacol Exp Ther ; 304(1): 301-9, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12490605

RESUMO

D-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH2 (CTAP) is a peptide antagonist that demonstrates potent and selective affinity for micro-opioid receptors in radioligand binding assays and in vitro bioassays. However, previous studies indicate that CTAP may possess unusual pharmacology under certain conditions. Therefore, CTAP was evaluated as an antagonist of the antinociceptive effects of a range of structurally diverse high- and low-efficacy peptide and alkaloid opioid agonists and compared with the traditional antagonist naltrexone. Male Sprague-Dawley rats (N = 227) were loosely restrained and the latency for tail withdrawal from 55 degrees C water was measured. Morphine s.c. and i.c.v., buprenorphine s.c., etorphine s.c. and i.c.v., [N-Me-Phe3,D-Pro4]-morphiceptin and [D-Ala2,N-Me-Phe4,Gly5-ol]-enkephalin produced antinociceptive effects. CTAP was at least 10-fold more potent than naltrexone as an antagonist of the antinociceptive effects of all five agonists. High doses of CTAP produced a noncompetitive antagonism of etorphine s.c. and morphine s.c. suggesting that CTAP may interact with additional opioid receptors in vivo or produce insurmountable antagonism at these doses. CTAP was approximately 300-fold more potent as an antagonist of DAMGO than the other agonists, indicating that CTAP may distinguish some peptide agonists such as DAMGO from other agonists based on binding interactions within the micro-opioid receptor or pharmacodynamic properties of these peptides. Naltrexone, however, administered by either s.c. or i.c.v. routes of administration was approximately equipotent as an antagonist of the antinociceptive effects of most agonists. Taken together, these data indicate that the peptide antagonist CTAP possesses a unique pharmacology unlike traditional opioid antagonists such as naltrexone


Assuntos
Alcaloides/antagonistas & inibidores , Antagonistas de Entorpecentes/farmacologia , Peptídeos Opioides/antagonistas & inibidores , Medição da Dor/efeitos dos fármacos , Peptídeos/farmacologia , Receptores Opioides mu/agonistas , Analgésicos Opioides/antagonistas & inibidores , Analgésicos Opioides/farmacologia , Animais , Ligação Competitiva/efeitos dos fármacos , Buprenorfina/antagonistas & inibidores , Buprenorfina/farmacologia , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina/antagonistas & inibidores , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Etorfina/antagonistas & inibidores , Etorfina/farmacologia , Injeções Intraventriculares , Injeções Subcutâneas , Masculino , Morfina/antagonistas & inibidores , Morfina/farmacologia , Naltrexona/farmacologia , Fragmentos de Peptídeos , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Somatostatina
14.
J Pharmacol Exp Ther ; 300(1): 265-72, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11752125

RESUMO

When the opioid agonist morphine is given chronically and systemically to mice by pellet implantation for 3 days, the animals develop substantial tolerance to the antinociceptive effect of a test dose of morphine given systemically. When the test dose is administered to the spinal cord, however, very little tolerance is observed. We tested six strains of mice differing in the degree to which they develop systemic tolerance to morphine and found that none of them developed significant tolerance to spinal morphine. However, most of these strains did develop substantial spinal tolerance to antinociception induced by the selective mu-agonist [D-Ala(2),N-Me-Phe(4),Gly(5)-ol]-enkephalin (DAMGO) and by the selective delta-agonist [D-Pen(2),D-Pen(5)]-enkephalin (DPDPE). Moreover, in naïve animals, the antinociceptive effect of both DAMGO and DPDPE was blocked by D-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH(2), a selective mu-antagonist, indicating that both agonists mediate antinociception in the spinal cord through mu-receptors. In addition to directly mediating antinociception, however, DPDPE potentiated the antinociceptive activity of DAMGO in the spinal cord of naïve animals, and this antinociception was blocked by the delta-antagonist H-TyrTicPsi[CH(2)NH]Phe-Thr-OH (TIPPpsi), indicating mediation through delta-receptors. In contrast, in tolerant animals, TIPPpsi enhanced the antinociception of DAMGO. These results thus demonstrate not only that mu- and delta-opioid receptors interact in naïve animals, but that the nature of this interaction changes during tolerance, from a potentiation to an inhibition. The lack of tolerance to spinal morphine may result from the ability of morphine to act as a partial antagonist at delta-receptors.


Assuntos
Analgésicos Opioides/farmacologia , Morfina/farmacologia , Receptores Opioides delta/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos , Animais , Tolerância a Medicamentos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/antagonistas & inibidores , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , D-Penicilina (2,5)-Encefalina/antagonistas & inibidores , D-Penicilina (2,5)-Encefalina/farmacologia , Feminino , Injeções Espinhais , Injeções Subcutâneas , Masculino , Camundongos , Camundongos Endogâmicos , Medição da Dor/efeitos dos fármacos , Peptídeos/farmacologia , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides mu/antagonistas & inibidores , Especificidade da Espécie
15.
Brain Res ; 881(1): 1-8, 2000 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-11033087

RESUMO

To determine the role of spinal mu-opioid receptor subtypes in antinociception induced by intrathecal (i.t.) injection of endomorphin-1 and -2, we assessed the effects of beta-funaltrexamine (a selective mu-opioid receptor antagonist) naloxonazine (a selective antagonist at the mu(1)-opioid receptor) and a novel receptor antagonist (3-methoxynaltrexone) using the paw-withdrawal test. Antinociception of i.t. endomorphins and [D-Ala(2), MePhe(4), Gly(ol)(5)]enkephalin (DAMGO) was completely reversed by pretreatment with beta-funaltrexamine (40 mg/kg s.c.). Pretreatment with a variety of doses of i.t. or s.c. naloxonazine 24 h before testing antagonized the antinociception of endomorphin-1, -2 and DAMGO. Judging from the ID(50) values of naloxonazine, the antinociceptive effect of endomorphin-2 was more sensitive to naloxonazine than that of endomorphin-1 or DAMGO. The selective morphine-6beta-glucuronide antagonist, 3-methoxynaltrexone, which blocked endomorphin-2-induced antinociception at each dose (0.25 mg/kg s.c. or 2.5 ng i.t.) that was inactive against DAMGO, did not affect endomorphin-1-induced antinociception but shifted the dose-response curve of endomorphin-2 3-fold to the right. These findings may be interpreted as indicative of the existence of a novel mu-opioid receptor subtype in spinal sites, where antinociception of morphine-6beta-glucuronide and endomorphin-2 are antagonized by 3-methoxynaltrexone. The present results suggest that endomorphin-1 and endomorphin-2 may produce antinociception through different subtypes of mu-opioid receptor.


Assuntos
Heroína/agonistas , Naloxona/análogos & derivados , Naltrexona/análogos & derivados , Oligopeptídeos/antagonistas & inibidores , Medição da Dor/efeitos dos fármacos , Receptores Opioides mu/antagonistas & inibidores , Analgésicos Opioides/antagonistas & inibidores , Animais , Ala(2)-MePhe(4)-Gly(5)-Encefalina/antagonistas & inibidores , Injeções Espinhais , Masculino , Camundongos , Naloxona/farmacologia , Naltrexona/farmacologia , Receptores Opioides mu/agonistas
16.
Brain Res Bull ; 52(2): 143-50, 2000 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10808085

RESUMO

The effect of (+/-)-5-methyl-10,11-dihydro-5H-dibenzo(a,d) cyclohepten-5, 10-imine maleate (MK-801) or 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX) injected intrathecally (i.t.) on the inhibition of the tail-flick response induced by morphine, D-Ala(2)-NmePhe(4)-Gly-ol-enkephalin (DAMGO), beta-endorphin, D-Pen(2,5)-enkephalin (DPDPE), or ¿(trans-3, 4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl) cyclohexyl] benzeocetamide)¿ (U50, 488H) administered i.t. was studied in ICR mice. The i.t. injection of MK-801 (2 microg) or CNQX (1 microg) alone did not affect the basal tail-flick response. Morphine (0.2 microg), DAMGO (0.8 ng), beta-endorphin (0.1 microg), DPDPE (0.5 microg) or U50, 488H (6 microg) caused only slight inhibition of the tail-flick response. CNQX injected i.t., but not MK-801, enhanced the inhibition of the tail-flick response induced by i.t. administered morphine, DAMGO, DPDPE or U50, 488H. However, CNQX or MK-801 injected i.t. was not effective in enhancing the inhibition of the tail-flick response induced by beta-endorphin administered i.t. The potentiating effect of CNQX on tail-flick inhibition induced by morphine, DAMGO, DPDPE or U50, 488H was blocked by naloxone (from 1 to 20 microg), yohimbine (from 1 to 20 microg) or methysergide (from 1 to 20 microg) injected i.t. in a dose-dependent manner. Our results suggest that the blockade of AMPA/kainate receptors located in the spinal cord appears to be involved in enhancing the inhibition of the tail-flick response induced by stimulation of spinal mu-, delta-, and kappa-opioid receptors. Furthermore, this potentiating action may be mediated by spinal noradrenergic and serotonergic receptors. However, N-methyl-D-aspartate receptors may not be involved in modulating the inhibition of the tail-flick response induced by various opioids administered spinally.


Assuntos
6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Maleato de Dizocilpina/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , D-Penicilina (2,5)-Encefalina/farmacologia , Dor/fisiopatologia , Medula Espinal/fisiologia , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/antagonistas & inibidores , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , 6-Ciano-7-nitroquinoxalina-2,3-diona/administração & dosagem , Analgésicos Opioides/farmacologia , Animais , Maleato de Dizocilpina/administração & dosagem , Ala(2)-MePhe(4)-Gly(5)-Encefalina/antagonistas & inibidores , D-Penicilina (2,5)-Encefalina/antagonistas & inibidores , Antagonistas de Aminoácidos Excitatórios/farmacologia , Injeções Espinhais , Masculino , Camundongos , Camundongos Endogâmicos ICR , Dor/prevenção & controle , Medula Espinal/efeitos dos fármacos , Fatores de Tempo , beta-Endorfina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...