Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Neurosci ; 40(34): 6522-6535, 2020 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-32690613

RESUMO

Chronic pain caused by spinal cord injury (SCI) is notoriously resistant to treatment, particularly by opioids. After SCI, DRG neurons show hyperactivity and chronic depolarization of resting membrane potential (RMP) that is maintained by cAMP signaling through PKA and EPAC. Importantly, SCI also reduces the negative regulation by Gαi of adenylyl cyclase and its production of cAMP, independent of alterations in G protein-coupled receptors and/or G proteins. Opioid reduction of pain depends on coupling of opioid receptors to Gαi/o family members. Combining high-content imaging and cluster analysis, we show that in male rats SCI decreases opioid responsiveness in vitro within a specific subset of small-diameter nociceptors that bind isolectin B4. This SCI effect is mimicked in nociceptors from naive animals by a modest 5 min depolarization of RMP (15 mm K+; -45 mV), reducing inhibition of cAMP signaling by µ-opioid receptor agonists DAMGO and morphine. Disinhibition and activation of C-Raf by depolarization-dependent phosphorylation are central to these effects. Expression of an activated C-Raf reduces sensitivity of adenylyl cyclase to opioids in nonexcitable HEK293 cells, whereas inhibition of C-Raf or treatment with the hyperpolarizing drug retigabine restores opioid responsiveness and blocks spontaneous activity of nociceptors after SCI. Inhibition of ERK downstream of C-Raf also blocks SCI-induced hyperexcitability and depolarization, without direct effects on opioid responsiveness. Thus, depolarization-dependent C-Raf and downstream ERK activity maintain a depolarized RMP and nociceptor hyperactivity after SCI, providing a self-reinforcing mechanism to persistently promote nociceptor hyperexcitability and limit the therapeutic effectiveness of opioids.SIGNIFICANCE STATEMENT Chronic pain induced by spinal cord injury (SCI) is often permanent and debilitating, and usually refractory to treatment with analgesics, including opioids. SCI-induced pain in a rat model has been shown to depend on persistent hyperactivity in primary nociceptors (injury-detecting sensory neurons), associated with a decrease in the sensitivity of adenylyl cyclase production of cAMP to inhibitory Gαi proteins in DRGs. This study shows that SCI and one consequence of SCI (chronic depolarization of resting membrane potential) decrease sensitivity to opioid-mediated inhibition of cAMP and promote hyperactivity of nociceptors by enhancing C-Raf activity. ERK activation downstream of C-Raf is necessary for maintaining ongoing depolarization and hyperactivity, demonstrating an unexpected positive feedback loop to persistently promote pain.


Assuntos
Dor Crônica/fisiopatologia , Nociceptores/fisiologia , Proteínas Proto-Oncogênicas c-raf/fisiologia , Receptores Opioides mu/fisiologia , Transdução de Sinais , Traumatismos da Medula Espinal/fisiopatologia , Animais , Células Cultivadas , Dor Crônica/complicações , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/fisiopatologia , Células HEK293 , Humanos , Masculino , Potenciais da Membrana , Ratos Sprague-Dawley , Receptores Opioides mu/agonistas , Traumatismos da Medula Espinal/complicações
2.
Neurochem Int ; 131: 104521, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31419453

RESUMO

Chronic pain is a worldwide major health problem and many pain-suffering patients are under opioid based therapy. Epidemiological data show that pain intensity correlates with the risk of misuse of prescription opioids, and other drugs of abuse including alcohol. This increased vulnerability to suffer Substance Use Disorders could be, in part, caused by functional changes that occur over the mesocorticolimbic system, a brain pathway involved in reward processing and addiction. Previous data in rats revealed that inflammatory pain desensitizes mu opioid receptors (MORs) in the ventral tegmental area (VTA). As a consequence, pain alters dopamine release in the nucleus accumbens (NAc) derived from MOR activation in the VTA and also increases intake of high doses of heroine. Given that the VTA neurons target different brain regions, in the present study we first analyzed changes induced by inflammatory pain in the MOR dependent activation pattern of the main VTA projecting areas. To do that, we administered two doses (7 or 14 ng) of DAMGO (MORs agonist) or artificial cerebrospinal fluid (aCSF) focally into the VTA of rats and measured the activation in projection areas by cFos immunohistochemistry. Our results show that focal injections of DAMGO in the VTA increases cFos expression in the majority of its projecting areas, namely NAc, basolateral amygdala (BLA), cingulate cortex (ACC) and bed nucleus of the stria terminalis (BNST), as compared to aCSF. Second, we analyzed whether inflammatory pain would affect to cFos expression using a group of rats injected with CFA in the hind paw. In this case, we found that cFos expression was not significantly different between DAMGO and aCSF administered rats in BLA, ACC and BNST. Our results confirm that inflammatory pain induces desensitization of VTA MORs in a region dependent manner which can be very relevant for addictive behaviours.


Assuntos
Genes fos/genética , Inflamação/metabolismo , Dor/metabolismo , Receptores Opioides mu/agonistas , Área Tegmentar Ventral/metabolismo , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/farmacologia , Animais , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Adjuvante de Freund , Regulação da Expressão Gênica , Imuno-Histoquímica , Inflamação/induzido quimicamente , Masculino , Microinjeções , Vias Neurais/efeitos dos fármacos , Vias Neurais/metabolismo , Ratos , Área Tegmentar Ventral/efeitos dos fármacos
3.
Alcohol Clin Exp Res ; 40(10): 2114-2123, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27508965

RESUMO

BACKGROUND: The nucleus accumbens shell is a key brain area mediating the reinforcing effects of ethanol (EtOH). Previously, it has been shown that the density of µ-opioid receptors in the nucleus accumbens shell is higher in alcohol-preferring Alko Alcohol (AA) rats than in alcohol-avoiding Alko Non-Alcohol rats. In addition, EtOH releases opioid peptides in the nucleus accumbens and opioid receptor antagonists are able to modify EtOH intake, all suggesting an opioidergic mechanism in the control of EtOH consumption. As the exact mechanisms of opioidergic involvement remains to be elucidated, the aim of this study was to clarify the role of accumbal µ- and κ-opioid receptors in controlling EtOH intake in alcohol-preferring AA rats. METHODS: Microinfusions of the µ-opioid receptor antagonist CTOP (0.3 and 1 µg/site), µ-opioid receptor agonist DAMGO (0.03 and 0.1 µg/site), nonselective opioid receptor agonist morphine (30 µg/site), and κ-opioid receptor agonist U50488H (0.3 and 1 µg/site) were administered via bilateral guide cannulas into the nucleus accumbens shell of AA rats that voluntarily consumed 10% EtOH solution in an intermittent, time-restricted (90-minute) 2-bottle choice access paradigm. RESULTS: CTOP (1 µg/site) significantly increased EtOH intake. Conversely, DAMGO resulted in a decreasing trend in EtOH intake. Neither morphine nor U50488H had any effect on EtOH intake in the used paradigm. CONCLUSIONS: The results provide further evidence for the role of accumbens shell µ-opioid receptors but not κ-opioid receptors in mediating reinforcing effects of EtOH and in regulating EtOH consumption. The results also provide support for views suggesting that the nucleus accumbens shell has a major role in mediating EtOH reward.


Assuntos
Consumo de Bebidas Alcoólicas/fisiopatologia , Etanol/administração & dosagem , Núcleo Accumbens/fisiologia , Receptores Opioides kappa/fisiologia , Receptores Opioides mu/fisiologia , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Animais , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Masculino , Microinjeções , Morfina/administração & dosagem , Morfina/farmacologia , Ratos , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Recompensa , Somatostatina/administração & dosagem , Somatostatina/análogos & derivados , Somatostatina/farmacologia , Especificidade da Espécie
4.
Eur J Pharm Sci ; 92: 173-82, 2016 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-27393342

RESUMO

The use of nanocarriers is an intriguing solution to increase the brain delivery of novel therapeutics. The aim of this paper was to use pharmacokinetic analysis and simulations to identify key factors that determine the effective drug concentration-time profile at the target site in the brain. Model building and simulations were based on experimental data obtained from the administration of the opioid peptide DAMGO in glutathione tagged PEGylated liposomes to rats. Different pharmacokinetic models were investigated to explore the mechanisms of increased brain delivery. Concentration-time profiles for a set of formulations with varying compound and carrier characteristics were simulated. By controlling the release rate from the liposome, the time profile and the extent of brain delivery can be regulated. The modeling did not support a mechanism of the liposomes passing the brain endothelial cell membrane in an intact form through endocytosis or transcytosis. The most likely process was found to be fusion of the liposome with the endothelial luminal membrane. The simulations revealed that low permeable compounds, independent on efflux, will gain the most from a nanocarrier formulation. The present model based approach is useful to explore and predict possibilities and limitations of carrier-based systems to the brain.


Assuntos
Analgésicos Opioides/farmacocinética , Encéfalo/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacocinética , Modelos Biológicos , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/química , Animais , Transporte Biológico , Simulação por Computador , Composição de Medicamentos , Células Endoteliais/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Ala(2)-MePhe(4)-Gly(5)-Encefalina/química , Glutationa/química , Lipossomos , Nanoestruturas/química , Polietilenoglicóis/química , Ratos
5.
Pharm Res ; 33(1): 177-85, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26275529

RESUMO

PURPOSE: The purpose of this study was to evaluate formulation factors causing improvement in brain delivery of a small peptide after encapsulation into a targeted nanocarrier in vivo. METHODS: The evaluation was performed in rats using microdialysis, which enabled continuous sampling of the released drug in both the brain (striatum) and blood. Uptake in brain could thereby be studied in terms of therapeutically active, released drug. RESULTS: We found that encapsulation of the peptide DAMGO in fast-releasing polyethylene glycol (PEG)ylated liposomes, either with or without the specific brain targeting ligand glutathione (GSH), doubled the uptake of DAMGO into the rat brain. The increased brain delivery was observed only when the drug was encapsulated into the liposomes, thus excluding any effects of the liposomes themselves on the blood-brain barrier integrity as a possible mechanism. The addition of a GSH coating on the liposomes did not result in an additional increase in DAMGO concentrations in the brain, in contrast to earlier studies on GSH coating. This may be caused by differences in the characteristics of the encapsulated compounds and the composition of the liposome formulations. CONCLUSIONS: We were able to show that encapsulation into PEGylated liposomes of a peptide with limited brain delivery could double the drug uptake into the brain without using a specific brain targeting ligand.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacocinética , Glutationa/química , Lipossomos/química , Polietilenoglicóis/química , Animais , Portadores de Fármacos , Composição de Medicamentos , Sistemas de Liberação de Medicamentos , Masculino , Microdiálise , Neostriado/metabolismo , Fosfatidilcolinas/química , Ratos , Ratos Sprague-Dawley
6.
Bull Exp Biol Med ; 156(4): 423-5, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24771418

RESUMO

We studied the role of µ-, δ-, and κ-opioid receptors of the stomach in the regulation of natural feeding behavior, metabolism, and locomotor activity of rats. Locomotor activity (number of crossed squares), food and water intake, oxygen consumption, and carbon dioxide release in animals were estimated in the standard home cage using a Phenomaster device (TSE) for 24 h at 40-min intervals. Administration of a µ-opioid receptor agonist DAMGO suppressed feeding behavior of animals in the light phase, but had little effect on locomotor activity and metabolism. Treatment with a δ-opioid receptor agonist DADLE was followed by the increase in metabolism over 24 h. These changes were accompanied by a decrease in locomotor activity during the light phase and activation of feeding behavior in the transition period. Intragastric administration of a κ-opioid receptor agonist ICI-204,448 inhibited feeding behavior, metabolism, and locomotor activity of rats only in the nighttime. These data suggest that opioid peptides produced in the stomach during food digestion play an important role in the regulation of food motivation and metabolism in rats. Various subtypes of opioid receptors probably regulate feeding behavior and metabolism of animals in different phases of vital activity.


Assuntos
Metabolismo Energético/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Atividade Motora/efeitos dos fármacos , Receptores Opioides/agonistas , Animais , Dióxido de Carbono/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Leucina Encefalina-2-Alanina/administração & dosagem , Mucosa Gástrica/metabolismo , Consumo de Oxigênio , Pirrolidinas/administração & dosagem , Ratos Wistar , Receptores Opioides/fisiologia , Estômago/efeitos dos fármacos
7.
Mol Pharm ; 10(5): 1533-41, 2013 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-22934681

RESUMO

Glutathione PEGylated (GSH-PEG) liposomes were evaluated for their ability to enhance and prolong blood-to-brain drug delivery of the opioid peptide DAMGO (H-Tyr-d-Ala-Gly-MePhe-Gly-ol). An intravenous loading dose of DAMGO followed by a 2 h constant rate infusion was administered to rats, and after a washout period of 1 h, GSH-PEG liposomal DAMGO was administered using a similar dosing regimen. DAMGO and GSH-PEG liposomal DAMGO were also administered as a 10 min infusion to compare the disposition of the two formulations. Microdialysis made it possible to determine free DAMGO in brain and plasma, while the GSH-PEG liposomal encapsulated DAMGO was measured with regular plasma sampling. The antinociceptive effect of DAMGO was determined with the tail-flick method. All samples were analyzed using liquid chromatography-tandem mass spectrometry. The short infusion of DAMGO resulted in a fast decline of the peptide concentration in plasma with a half-life of 9.2 ± 2.1 min. Encapsulation in GSH-PEG liposomes prolonged the half-life to 6.9 ± 2.3 h. Free DAMGO entered the brain to a limited extent with a steady state ratio between unbound drug concentrations in brain interstitial fluid and in blood (Kp,uu) of 0.09 ± 0.04. GSH-PEG liposomes significantly increased the brain exposure of DAMGO to a Kp,uu of 0.21 ± 0.17 (p < 0.05). By monitoring the released, active substance in both blood and brain interstitial fluid over time, we were able to demonstrate that GSH-PEG liposomes offer a promising platform for enhancing and prolonging the delivery of drugs to the brain.


Assuntos
Analgésicos Opioides/administração & dosagem , Encéfalo/metabolismo , Sistemas de Liberação de Medicamentos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Analgésicos Opioides/farmacocinética , Animais , Barreira Hematoencefálica , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacocinética , Glutationa/administração & dosagem , Meia-Vida , Infusões Intravenosas , Lipossomos/administração & dosagem , Masculino , Microdiálise , Polietilenoglicóis/administração & dosagem , Ratos , Ratos Sprague-Dawley
8.
J Neurosci ; 31(5): 1591-9, 2011 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-21289167

RESUMO

The decision to perform, or not perform, actions known to lead to a rewarding outcome is strongly influenced by the current incentive value of the reward. Incentive value is largely determined by the affective experience derived during previous consumption of the reward-the process of incentive learning. We trained rats on a two-lever, seeking-taking chain paradigm for sucrose reward, in which responding on the initial seeking lever of the chain was demonstrably controlled by the incentive value of the reward. We found that infusion of the µ-opioid receptor antagonist, CTOP (d-Phe-Cys-Tyr-d-Trp-Orn-Thr-Pen-Thr-NH(2)), into the basolateral amygdala (BLA) during posttraining, noncontingent consumption of sucrose in a novel elevated-hunger state (a positive incentive learning opportunity) blocked the encoding of incentive value information normally used to increase subsequent sucrose-seeking responses. Similar treatment with δ [N, N-diallyl-Tyr-Aib-Aib-Phe-Leu-OH (ICI 174,864)] or κ [5'-guanidinonaltrindole (GNTI)] antagonists was without effect. Interestingly, none of these drugs affected the ability of the rats to encode a decrease in incentive value resulting from experiencing the sucrose in a novel reduced-hunger state. However, the µ agonist, DAMGO ([d-Ala2, NMe-Phe4, Gly5-ol]-enkephalin), appeared to attenuate this negative incentive learning. These data suggest that upshifts and downshifts in endogenous opioid transmission in the BLA mediate the encoding of positive and negative shifts in incentive value, respectively, through actions at µ-opioid receptors, and provide insight into a mechanism through which opiates may elicit inappropriate desire resulting in their continued intake in the face of diminishing affective experience.


Assuntos
Tonsila do Cerebelo/efeitos dos fármacos , Condicionamento Operante/efeitos dos fármacos , Impulso (Psicologia) , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Recompensa , Somatostatina/análogos & derivados , Tonsila do Cerebelo/metabolismo , Animais , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Encefalina Leucina/administração & dosagem , Encefalina Leucina/análogos & derivados , Encefalina Leucina/farmacologia , Alimentos , Guanidinas , Masculino , Microinjeções , Morfinanos , Naltrexona/administração & dosagem , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/administração & dosagem , Antagonistas de Entorpecentes/farmacologia , Testes Neuropsicológicos , Ratos , Ratos Long-Evans , Receptores Opioides kappa/antagonistas & inibidores , Somatostatina/administração & dosagem , Somatostatina/farmacologia , Sacarose
9.
J Neurosci ; 30(42): 14029-35, 2010 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-20962224

RESUMO

GABA transmission in the ventral tegmental area (VTA) is critical for fine tuning the activity of dopamine neurons in response to opioids. However, the precise mechanism by which GABA input shapes opioid reward is poorly understood. We observed a reduction of conditioned place preference for low doses of the opioid [d-Ala2, N-MePhe4, Gly5-ol]-enkephalin (DAMGO) and a switch in the functional effects of µ-opioid receptor modulation of GABA postsynaptic currents in the mouse VTA 1 d after chronic morphine treatment. Specifically, whereas in naive mice DAMGO inhibits GABA postsynaptic currents, GABAergic currents are potentiated by DAMGO after chronic morphine treatment. Importantly, pretreatment with the cAMP signaling inhibitor (R)-adenosine, cyclic 3',5'-(hydrogenphosphorothioate) triethylammonium both restored DAMGO reward and reversed the DAMGO-mediated potentiation, thereby reestablishing the inhibitory effects of opioids on GABA currents. Thus, a paradoxical bidirectionality in µ-receptor-mediated control of GABA transmission following chronic morphine treatment is a critical mechanism that determines the expression of opioid reward in the VTA.


Assuntos
Analgésicos Opioides/farmacologia , Morfina/farmacologia , Recompensa , Área Tegmentar Ventral/efeitos dos fármacos , Ácido gama-Aminobutírico/fisiologia , Analgésicos Opioides/administração & dosagem , Animais , Condicionamento Operante/efeitos dos fármacos , AMP Cíclico/análogos & derivados , AMP Cíclico/antagonistas & inibidores , AMP Cíclico/farmacologia , AMP Cíclico/fisiologia , Eletrofisiologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Injeções , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/administração & dosagem , Receptores Opioides mu/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Tionucleotídeos/farmacologia
10.
Brain Res ; 1317: 100-7, 2010 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-20051234

RESUMO

Activation of mu opioid receptors (MOR) makes animals hyperphagic and selectively increases their preference for a high fat diet independent of their dietary preference. The orexigenic peptide Agouti Related Peptide (AgRP) also produces hyperphagia and increased the preference for a high fat diet. In this paper, we tested the hypothesis that the effect of MOR on feeding behavior will be attenuated in the absence of the orexigenic peptide AgRP. Immunohistochemical studies demonstrated that MOR are co-localized on AgRP neurons located in the arcuate nucleus. This finding is consistent with a role of MOR in mediating the release of AgRP. Our data also demonstrated that the wild-type (FVB) animals preferred a diet high in fat whereas the AgRP knockout (AgRP KO) mice did not. mRNA expression of MOR in the hypothalamus was not significantly different between AgRP KO mice and their wild-type control. In a dose-response experiment, the low dose (0.025 microg) of a MOR agonist, DAMGO, increased cumulative food intake in wild-type and AgRP KO mice. The low and middle (0.25 microg) dose of DAMGO significantly increased the amount of high fat diet eaten by the wild-type animals, but did not significantly change the amount of high fat diet eaten by the AgRP KO mice. The highest dose of DAMGO (2.5 microg) reduced food intake in the control and AgRP KO mice, probably due to somnolence. These data demonstrate that the increased preference for a high fat diet after stimulation of MOR is attenuated in the absence of AgRP, but the increase in food intake (i.e. hyperphagia) is not.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Dieta , Gorduras na Dieta , Preferências Alimentares , Hiperfagia/metabolismo , Receptores Opioides mu/metabolismo , Proteína Relacionada com Agouti/genética , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Feminino , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurotransmissores/administração & dosagem , Neurotransmissores/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Opioides mu/agonistas
11.
J Appl Physiol (1985) ; 107(5): 1591-9, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19745186

RESUMO

Opioids are clinically important in the alleviation of pain. An undesirable side effect of opioids is depression of breathing. Data from isolated preparations suggest this effect is due to attenuation of discharge activity of neurons in the pre-Bötzinger complex (preBötzC), a medullary area with respiratory rhythmogenic properties. The purpose of this study was to examine how [d-Ala(2),N-Me-Phe(4),Gly(5)-ol]-enkephalin (DAMGO), a mu-opioid receptor agonist, affected breathing after injection into the presumed preBötzC of the adult awake goat. We hypothesized that DAMGO would cause breathing to decrease and become irregular when injected into the presumed preBötzC and the surrounding region of the conscious animal. We further hypothesized that ventilatory sensitivity to CO(2) and hypoxia would be blunted after the injection of DAMGO. Microtubules were bilaterally implanted into the presumed preBötzC of 10 adult female goats. After recovery from the surgery, DAMGO (0.5-10 mul, 1 nM-10 muM) was injected into the presumed preBötzC during the awake state. DAMGO had no effect on pulmonary ventilation [inspiratory minute ventilation (Vi)], respiratory rhythm and pattern, the activation pattern of inspiratory and expiratory muscles, or arterial blood gases during eupneic breathing conditions (P > 0.10). However, DAMGO attenuated (P < 0.05) the evoked increase in breathing frequency when inspired CO(2) was increased, and DAMGO attenuated the Vi response to reduction of inspired O(2) to 10.8% (P < 0.05). We conclude that our data do not provide support for the concept that in awake mammals opioid depression of breathing is due to a directed action of opioids on preBötzC neurons.


Assuntos
Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Bulbo/fisiologia , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Centro Respiratório/fisiologia , Mecânica Respiratória/fisiologia , Animais , Feminino , Cabras , Injeções , Bulbo/efeitos dos fármacos , Centro Respiratório/efeitos dos fármacos , Mecânica Respiratória/efeitos dos fármacos , Vigília
12.
Neuroscience ; 161(3): 718-33, 2009 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-19336249

RESUMO

Mu opioid receptor (MOR) signaling in the nucleus accumbens (NAcc) elicits marked increases in the consumption of palatable tastants. However, the mechanism and circuitry underlying this effect are not fully understood. Multiple downstream target regions have been implicated in mediating this effect but the role of the ventral pallidum (VP), a primary target of NAcc efferents, has not been well defined. To probe the mechanisms underlying increased consumption, we identified behavioral changes in rats' licking patterns following NAcc MOR stimulation. Because the temporal structure of licking reflects the physiological substrates modulating consumption, these measures provide a useful tool in dissecting the cause of increased consumption following NAcc MOR stimulation. Next, we used a combination of pharmacological inactivation and lesions to define the role of the VP in hyperphagia following infusion of the MOR-specific agonist [D-Ala2, N-MePhe4, Gly-ol]-enkephalin (DAMGO) into the NAcc. In agreement with previous studies, results from lick microstructure analysis suggest that NAcc MOR stimulation augments intake through a palatability-driven mechanism. Our results also demonstrate an important role for the VP in normal feeding behavior: pharmacological inactivation of the VP suppresses baseline and NAcc DAMGO-induced consumption. However, this interaction does not occur through a serial circuit requiring direct projections from the NAcc to the VP. Rather, our results indicate that NAcc and VP circuits converge on a common downstream target that regulates food intake.


Assuntos
Ingestão de Alimentos/fisiologia , Globo Pálido/fisiologia , Núcleo Accumbens/fisiologia , Receptores Opioides mu/metabolismo , Animais , Cateterismo , Ingestão de Alimentos/efeitos dos fármacos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Comportamento Alimentar/efeitos dos fármacos , Comportamento Alimentar/fisiologia , Agonistas GABAérgicos/administração & dosagem , Globo Pálido/efeitos dos fármacos , Hiperfagia/induzido quimicamente , Hiperfagia/fisiopatologia , Masculino , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Muscimol/administração & dosagem , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Neurotoxinas/toxicidade , Neurotransmissores/administração & dosagem , Núcleo Accumbens/efeitos dos fármacos , Ácido Quinolínico/toxicidade , Distribuição Aleatória , Ratos , Ratos Long-Evans , Receptores Opioides mu/agonistas , Fatores de Tempo
13.
Am J Physiol Regul Integr Comp Physiol ; 296(5): R1528-37, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19244586

RESUMO

Sighs, a well-known phenomenon in mammals, are substantially augmented by hypoxia and hypercapnia. Because (d-Ala(2),N-Me-Phe(4),Gly-ol)-enkephalin (DAMGO), a mu-receptor agonist, injected intravenously and locally in the caudal medullary raphe region (cMRR) decreased the ventilatory response to hypoxia and hypercapnia, we hypothesized that these treatments could inhibit sigh responses to these chemical stimuli. The number and amplitude of sighs were recorded during three levels of isocapnic hypoxia (15%, 10%, and 5% O(2) for 1.5 min) or hypercapnia (3%, 7%, and 10% CO(2) for 4 min) to test the dependence of sigh responses on the intensity of chemical drive in anesthetized and spontaneously breathing rats. The role of mu-receptors in modulating sigh responses to 10% O(2) or 7% CO(2) was subsequently evaluated by comparing the sighs before and after 1) intravenous administration of DAMGO (100 microg/kg), 2) microinjection of DAMGO (35 ng/100 nl) into the cMRR, and 3) intravenous administration of DAMGO after microinjection of d-Phe-Cys-Tyr-d-Trp-Arg-Thr-Pen-Thr-NH(2) (CTAP, 100 ng/100 nl), a micro-receptor antagonist, into the cMRR. Hypoxia and hypercapnia increased the number, but not amplitude, of sighs in a concentration-dependent manner, and the responses to hypoxia were significantly greater than those to hypercapnia. Systemic and local injection of DAMGO into the cMRR predominantly decreased the number of sighs, while microinjection into the rostral and middle MRR had no or limited effects. Microinjecting CTAP into the cMRR significantly diminished the systemic DAMGO-induced reduction of the number of sighs in response to hypoxia, but not to hypercapnia. Thus we conclude that hypoxia and hypercapnia elevate the number of sighs in a concentration-dependent manner in anesthetized rats, and this response is significantly depressed by activating systemic mu-receptors, especially those within the cMRR.


Assuntos
Núcleos da Rafe/metabolismo , Receptores Opioides mu/metabolismo , Respiração , Vocalização Animal/fisiologia , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/farmacologia , Animais , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Hipercapnia/metabolismo , Hipercapnia/fisiopatologia , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Masculino , Microinjeções , Modelos Animais , Peptídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/efeitos dos fármacos , Respiração/efeitos dos fármacos , Vocalização Animal/efeitos dos fármacos
14.
Brain Res Bull ; 74(5): 369-75, 2007 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-17845912

RESUMO

Peripheral micro-opioid receptors (MOR) have emerged as important components of inhibitory nociceptive pathways. Here, the antinociceptive effects of MOR agonists, the 6beta-glycine derivative of 14-O-methyloxymorphone (HS-731), DAMGO and morphine were evaluated in a mouse model of visceral pain. The abdominal acetic acid-induced writhing test was used to examine the peripheral, preemptive antinociceptive opioid action on visceral nociception. HS-731 administered subcutaneously (s.c.) or intracerebroventricularly (i.c.v.) dose-dependently and completely inhibited writhing, being 24-598-fold more potent, depending on the administration route, than two selective MOR agonists, the enkephalin analogue [D-Ala(2),N-Me-Phe(4),Gly-ol(5)]enkephalin (DAMGO) and morphine. A longer duration of action (2-3 h) was induced by HS-731 given before acetic acid, while shorter effect was produced by morphine (30-60 min) and DAMGO (30-45 min). The antinociceptive effects of systemic opioids were reversed by the s.c. opioid antagonist, naloxone. Blocking of central MOR by the selective MOR antagonist D-Phe-Cys-Tyr-d-Trp-Arg-Thr-Pen-Thr-NH(2) (CTAP, i.c.v.) resulted in a significant reduction of antinociception of s.c. morphine, whereas it completely failed to antagonize the effects of systemic HS-731 or DAMGO. In in vitro studies, HS-731 and DAMGO, but not morphine showed high intrinsic efficacy, naltrexone-sensitive agonist effect at MOR of the rat vas deferens. These data demonstrate that selective activation of peripheral MOR by systemic s.c. HS-731 or DAMGO produces potent peripheral, preemptive visceral antinociception, while morphine's effects are mediated primarily through central mechanisms. Our findings support the role of peripheral MOR in the pathology of pain states involving sensitization of peripheral nociceptors.


Assuntos
Analgésicos Opioides/administração & dosagem , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Compostos de Epóxi/administração & dosagem , Morfinanos/administração & dosagem , Morfina/administração & dosagem , Dor/tratamento farmacológico , Ducto Deferente/efeitos dos fármacos , Ácido Acético , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Vias de Administração de Medicamentos , Esquema de Medicação , Interações Medicamentosas , Técnicas In Vitro , Masculino , Camundongos , Dor/induzido quimicamente , Medição da Dor , Fragmentos de Peptídeos , Peptídeos/uso terapêutico , Ratos , Ratos Wistar , Somatostatina , Fatores de Tempo , Ducto Deferente/fisiologia
15.
Int J Urol ; 14(3): 226-31; discussion 232, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17430260

RESUMO

AIM: In order to determine the influence of different opioid receptor subtypes on detrusor overactivity after left middle cerebral artery (MCA) occlusion, cystometric recordings were obtained in conscious rats. METHODS: Female Sprague-Dawley rats were used in this study. Control cystometrography was followed by left MCA occlusion. The sham-operated (SO) rats underwent the same procedures except for MCA occlusion. [D-Ala(2), Phe(4), Gly(5)]-enkephalin (DAGO; mu-opioid agonist), [D-Pen(2,5)]-enkephalin (DPDPE; delta1-opioid agonist), deltorpin II (delta2-opioid agonist), and U-50488 (kappa-opioid agonist) were administered intracerebroventricularly at graded doses. The bladder capacity, residual volume, micturition threshold pressure, and bladder contraction pressure were determined. Finally, the volume of the infarction was measured. RESULTS: The intracerebroventricular administration of DAGO and DPDPE significantly increased the bladder capacity in the cerebrally infarcted (CI) and SO rats, but differences in the changes in bladder capacity between the CI and SO rats were not significant. Deltorpin II did not produce any changes in the bladder capacity in the CI or SO rats at any dose examined. However, the intracerebroventricular administration of U-50488 significantly increased the bladder capacity in the CI rats but not in the SO rats. None of the drugs affected the residual volume, micturition threshold pressure or bladder contraction pressure at any dosage examined. The mean infarcted volumes were not significantly different from those in the vehicle-treated rats. CONCLUSION: These results suggest that the opioid receptor subtypes, mu and delta1 in the brain, are related to the micturition reflex. Furthermore, the kappa opioid agonist might be useful for the suppression of detrusor overactivity caused by cerebral infarction.


Assuntos
Analgésicos não Narcóticos/uso terapêutico , Analgésicos Opioides/uso terapêutico , Infarto Cerebral/fisiopatologia , D-Penicilina (2,5)-Encefalina/uso terapêutico , Bexiga Urinária Hiperativa/tratamento farmacológico , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/administração & dosagem , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/uso terapêutico , Analgésicos não Narcóticos/administração & dosagem , Analgésicos Opioides/administração & dosagem , Animais , Infarto Cerebral/complicações , Ventrículos Cerebrais , Modelos Animais de Doenças , Vias de Administração de Medicamentos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Ala(2)-MePhe(4)-Gly(5)-Encefalina/uso terapêutico , D-Penicilina (2,5)-Encefalina/administração & dosagem , Feminino , Oligopeptídeos/administração & dosagem , Oligopeptídeos/uso terapêutico , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento , Bexiga Urinária Hiperativa/etiologia , Bexiga Urinária Hiperativa/fisiopatologia , Urodinâmica/efeitos dos fármacos
16.
Brain Res ; 1096(1): 120-4, 2006 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-16716266

RESUMO

The lateral hypothalamic area (LHa) is an important brain site for the regulation of food intake. Central injection of opioids increases food intake, and the LHa contains mu and kappa opioid receptors, both of which are involved in feeding behavior. It is unclear whether opioids impact feeding when injected directly into the rostral portion of the LHa (rLHa) in rats. We performed a series of studies in which free-feeding rLHa-cannulated rats were injected with opioid agonists (DAMGO, morphine, dynorphin, U-50488H) followed by the measurement of food intake at 1, 2, and 4 h postinjection. To determine whether opioid receptor ligands administered into the rLHa affect neuronal activation in this brain site, we studied cFos immunoreactivity (cFos IR) in response to rLHa stimulation with naltrexone. We found that the only compound that stimulated feeding behavior was morphine. The other agonists had no effect on food consumption. Naltrexone injection into the rLHa increased neural activation in the LHa, indicating the presence of functional opioid receptors in this region. These data suggest that although neuronal activity is affected by opioid agents acting in the rLHa, administration of selective mu and kappa opioid ligands in this subdivision of the LHa does not have a reliable effect on feeding behavior.


Assuntos
Comportamento Alimentar/efeitos dos fármacos , Genes fos/genética , Hipotálamo/fisiologia , Receptores Opioides/efeitos dos fármacos , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/farmacologia , Animais , Dinorfinas/farmacologia , Ingestão de Alimentos/efeitos dos fármacos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Expressão Gênica/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Ligantes , Masculino , Microinjeções , Morfina/administração & dosagem , Morfina/farmacologia , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inibidores , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores
17.
Neuropsychopharmacology ; 31(4): 739-50, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16123756

RESUMO

Clinically, it has been reported that chronic pain induces depression, anxiety, and reduced quality of life. The endogenous opioid system has been implicated in nociception, anxiety, and stress. The present study was undertaken to investigate whether chronic pain could induce anxiogenic effects and changes in the opioidergic function in the amygdala in mice. We found that either injection of complete Freund's adjuvant (CFA) or neuropathic pain induced by sciatic nerve ligation produced a significant anxiogenic effect at 4 weeks after the injection or surgery. Under these conditions, the selective mu-opioid receptor agonist [D-Ala2,N-MePhe4,Gly5-ol]-enkephalin (DAMGO)- and the selective delta-opioid receptor agonist (+)-4-[(alphaR)-alpha-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide (SNC80)-stimulated [35S]GTPgammaS binding in membranes of the amygdala was significantly suppressed by CFA injection or nerve ligation. CFA injection was associated with a significant increase in the kappa-opioid receptor agonist 2-(3,4-dichlorophenyl)-N-methyl-N-[(1S)-1-phenyl-2-(1-pyrrolidinyl)ethyl]acetamide hydrochloride (ICI199,441)-stimulated [35S]GTPgammaS binding in membranes of the amygdala. The intracerebroventricular administration and microinjection of a selective mu-opioid receptor antagonist, a selective delta-opioid receptor antagonist, and the endogenous kappa-opioid receptor ligand dynorphin A caused a significant anxiogenic effect in mice. We also found that thermal hyperalgesia induced by sciatic nerve ligation was reversed at 8 weeks after surgery. In the light-dark test, the time spent in the lit compartment was not changed at 8 weeks after surgery. Collectively, the present data constitute the first evidence that chronic pain has an anxiogenic effect in mice. This phenomenon may be associated with changes in opioidergic function in the amygdala.


Assuntos
Tonsila do Cerebelo/metabolismo , Ansiedade/etiologia , Ansiedade/metabolismo , Entorpecentes/metabolismo , Dor/complicações , Ciática/fisiopatologia , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/fisiopatologia , Analgésicos Opioides , Análise de Variância , Animais , Comportamento Animal , Benzamidas/administração & dosagem , Doença Crônica , Diazepam/análogos & derivados , Diazepam/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Interações Medicamentosas , Dinorfinas/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Adjuvante de Freund , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Injeções Intraventriculares/métodos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Dor/induzido quimicamente , Dor/fisiopatologia , Medição da Dor/métodos , Piperazinas/administração & dosagem , Ligação Proteica/efeitos dos fármacos , Pirrolidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Ciática/etiologia , Somatostatina/análogos & derivados , Somatostatina/farmacologia , Isótopos de Enxofre/farmacocinética , Fatores de Tempo , Tranquilizantes/farmacologia
18.
J Neurosci ; 21(7): 2536-45, 2001 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11264327

RESUMO

This study examined a mechanism responsible for the enhanced antihyperalgesic and antinociceptive effects of the mu opioid receptor agonist (ORA) [D-Ala(2), NMePhe(4), Gly(5)-ol]enkephalin (DAMGO) microinjected in the rostroventromedial medulla (RVM) of rats with inflammatory injury induced by injection of complete Freund's adjuvant (CFA) in one hindpaw. In rats injected with CFA 4 hr earlier, microinjection of the mu opioid receptor antagonist D-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH(2) (CTAP) in the RVM antagonized both the marginal enhancement of the potency of DAMGO and its antinociceptive effect. The delta opioid receptor antagonist naltriben (NTB) was without effect. In rats injected with CFA 2 weeks earlier, CTAP antagonized the effects of DAMGO to a lesser extent. However, NTB completely prevented the enhancement of the potency of DAMGO, whereas it did not antagonize DAMGO's antinociceptive effects. Microinjection of NTB alone, but not CTAP in the RVM of CFA-treated rats, enhanced the hyperalgesia present in the ipsilateral hindpaw and induced hyperalgesia in the contralateral, uninjured hindpaw. These results suggest that persistent inflammatory injury increased the release in the RVM of opioid peptides with preferential affinity for the delta opioid receptor, which can interact in a synergistic or additive manner with an exogenously administered mu opioid receptor agonist. Indeed, the levels of [Met(5)]enkephalin and [Leu(5)]enkephalin were increased in the RVM and in other brainstem nuclei in CFA-treated rats. This increase most likely presents a compensatory neuronal response of the CNS of the injured animal to mitigate the full expression of inflammatory pain and to enhance the antinociceptive and antihyperalgesic effects of exogenously administered mu opioid receptor analgesics.


Assuntos
Encéfalo/efeitos dos fármacos , Encefalinas/metabolismo , Inflamação/metabolismo , Dor/metabolismo , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/efeitos dos fármacos , Animais , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Masculino , Microinjeções , Naltrexona/administração & dosagem , Naltrexona/análogos & derivados , Fragmentos de Peptídeos , Peptídeos/administração & dosagem , Ratos , Ratos Sprague-Dawley , Somatostatina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA