Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neurosci ; 40(34): 6522-6535, 2020 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-32690613

RESUMEN

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.


Asunto(s)
Dolor Crónico/fisiopatología , Nociceptores/fisiología , Proteínas Proto-Oncogénicas c-raf/fisiología , Receptores Opioides mu/fisiología , Transducción de Señal , Traumatismos de la Médula Espinal/fisiopatología , Animales , Células Cultivadas , Dolor Crónico/complicaciones , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/fisiopatología , Células HEK293 , Humanos , Masculino , Potenciales de la Membrana , Ratas Sprague-Dawley , Receptores Opioides mu/agonistas , Traumatismos de la Médula Espinal/complicaciones
2.
J Neurosci ; 40(35): 6812-6819, 2020 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-32690616

RESUMEN

Parkinson's disease (PD) is characterized by severe locomotor deficits and is commonly treated with the dopamine precursor L-DOPA, but its prolonged usage causes dyskinesias referred to as L-DOPA-induced dyskinesia (LID). Several studies in animal models of PD have suggested that dyskinesias are associated with a heightened opioid cotransmitter tone, observations that have led to the notion of a LID-related hyperactive opioid transmission that should be corrected by µ opioid receptor antagonists. Reports that both antagonists and agonists of the µ opioid receptor may alleviate LID severity in primate models of PD and LID, together with the failure of nonspecific antagonist to improve LID in pilot clinical trials in patients, raises doubt about the reliability of the available data on the opioid system in PD and LID. After in vitro characterization of the functional activity at the µ opioid receptor, we selected prototypical agonists, antagonists, and partial agonists at the µ opioid receptor. We then showed that both oral and discrete intracerebral administration of a µ receptor agonist, but not of an antagonist as long thought, ameliorated LIDs in the gold-standard bilateral 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned female macaque model of PD and LID. The results call for a reappraisal of opioid pharmacology in the basal ganglia as well as for the development of brain nucleus-targeted µ opioid receptor agonists.SIGNIFICANCE STATEMENT µ opioid receptors have long been considered as a viable target for alleviating the severity of L-DOPA-induced hyperkinetic side effects, induced by the chronic treatment of Parkinson's disease motor symptoms with L-DOPA. Conflicting results between experimental parkinsonism and Parkinson's disease patients, however, dampened the enthusiasm for the target. Here we reappraise the pharmacology and then demonstrate that both oral and discrete intracerebral administration of a µ receptor agonist, but not of an antagonist as long thought, ameliorates LIDs in the gold-standard bilateral 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned macaque model of Parkinson's disease, calling for a reappraisal of the opioid pharmacology as well as for the development of brain nucleus-targeted µ receptor agonists.


Asunto(s)
Discinesias/tratamiento farmacológico , Intoxicación por MPTP/fisiopatología , Receptores Opioides mu/agonistas , Animales , Células CHO , Cricetinae , Cricetulus , Discinesias/etiología , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Encefalina Ala(2)-MeFe(4)-Gli(5)/uso terapéutico , Femenino , Humanos , Levodopa/efectos adversos , Levodopa/uso terapéutico , Intoxicación por MPTP/tratamiento farmacológico , Macaca fascicularis , Neurotransmisores/administración & dosificación , Neurotransmisores/farmacología , Neurotransmisores/uso terapéutico , Receptores Opioides mu/antagonistas & inhibidores
3.
Molecules ; 26(22)2021 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-34833919

RESUMEN

BACKGROUND: Recent studies suggest that lipids, including free fatty acids (FFAs), are necessary for proper µ opioid receptor (MOR) binding and that activation of opioid receptors (ORs) improves intestinal inflammation. The objective of the study was to investigate a possible interaction between the ORs and FFA receptors (FFARs) ligands in the colitis. METHODS: The potential synergistic effect of ORs and FFARs ligands was evaluated using mouse model of acute colitis induced by dextran sulfate sodium (DSS, 4%). Compounds were injected intraperitoneally (i.p.) once or twice daily at the doses of 0.01 or 0.02 mg/kg body weight (BW) (DAMGO-an MOR agonist), 0.3 mg/kg BW (DPDPE-a δ OR (DOR) agonist) and 1 mg/kg BW (naloxone-a non-selective OR antagonist, GLPG 0974-a FFAR2 antagonist, GSK 137647-a FFAR4 agonist and AH 7614-a FFAR4 antagonist) for 4 days. RESULTS: Myeloperoxidase (MPO) activity was significantly decreased after DAMGO (0.02 mg/kg BW) and GSK 137647 (1 mg/kg BW) administration and co-administration as compared to DSS group. CONCLUSIONS: Treatment with ligands of ORs and FFARs may affect the immune cells in the inflammation; however, no significant influence on the severity of colitis and no synergistic effect were observed.


Asunto(s)
Colitis/tratamiento farmacológico , Colitis/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Opioides/metabolismo , Compuestos de Anilina/administración & dosificación , Animales , Butiratos/administración & dosificación , Colitis/inmunología , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Encefalina D-Penicilamina (2,5)/administración & dosificación , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Ligandos , Masculino , Ratones , Ratones Endogámicos BALB C , Naloxona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Peroxidasa/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Opioides/agonistas , Receptores Opioides delta/agonistas , Receptores Opioides delta/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Sulfonamidas/administración & dosificación , Tiofenos/administración & dosificación , Xantenos/administración & dosificación
4.
Org Biomol Chem ; 17(21): 5305-5315, 2019 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-31094391

RESUMEN

Numerous studies demonstrate the promise of opioid peptides as analgesics, but poor oral bioavailability has limited their therapeutic development. This study sought to increase the oral bioavailability of opioid peptides by cyclization, using Hantzsch-based macrocyclization strategies to produce two new series of cyclized DAMGO and Leu/Met-enkephalin analogs. Opioid receptor affinity and selectivity for compounds in each series were assessed in vitro with radioligand competition binding assays. Compounds demonstrated modest affinity but high selectivity for the mu, delta, and kappa opioid receptors (MOR, DOR and KOR), while selectivity for mu opioid receptors varied by structure. Antinociceptive activity of each compound was initially screened in vivo following intracerebroventricular (i.c.v.) administration and testing in the mouse 55 °C warm-water tail-withdrawal test. The four most active compounds were then evaluated for dose- and time-dependent antinociception, and opioid receptor selectivity in vivo. Cyclic compounds 1924-10, 1936-1, 1936-7, and 1936-9 produced robust and long- lasting antinociception with ED50 values ranging from 0.32-0.75 nmol following i.c.v. administration mediated primarily by mu- and delta-opioid receptor agonism. Compounds 1924-10, 1936-1 and 1936-9 further displayed significant time-dependent antinociception after oral (10 mg kg-1, p.o.) administration. A higher oral dose (30 mg kg-1. p.o.) of all four cyclic peptides also reduced centrally-mediated respiration, suggesting successful penitration into the CNS. Overall, these data suggest cyclized opioid peptides synthesized by a Hantzsch-based macrocyclization strategy can retain opioid agonist activity to produce potent antinociception in vivo while conveying improved bioavailability following oral administration.


Asunto(s)
Analgésicos Opioides/farmacología , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Encefalina Metionina/farmacología , Receptores Opioides/agonistas , Tiazoles/farmacología , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/química , Animales , Ciclización , Relación Dosis-Respuesta a Droga , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Encefalina Ala(2)-MeFe(4)-Gli(5)/química , Encefalina Metionina/administración & dosificación , Encefalina Metionina/química , Inyecciones Intraventriculares , Masculino , Ratones , Ratones Endogámicos C57BL , Conformación Molecular , Frecuencia Respiratoria , Tiazoles/administración & dosificación , Tiazoles/química
5.
Am J Physiol Regul Integr Comp Physiol ; 314(5): R693-R699, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29341826

RESUMEN

µ-Opioid G protein-coupled receptors (MOR) interact with ion channels to decrease neuronal excitability. In humans, intrathecal administration of the MOR agonist fentanyl inhibits the exercise pressor reflex, an effect that can be attributed to either the opening of inward rectifying potassium channels (GIRK) or the closing of N-type calcium channels. The purpose of this study was to determine if the highly selective MOR agonist [d-Ala2, N-MePhe4,Gly-ol]-enkephalin (DAMGO) attenuates the exercise pressor reflex and which of these two channels are responsible for this effect. In decerebrate rats, we determined the effect of intrathecal injection of either tertiapin-LQ, which blocks the GIRK channel or ω-conotoxin-GVIA, which blocks the N-type calcium channel on the exercise pressor reflex, which was evoked by contracting the triceps surae muscles. Initially, we established that intrathecal injection of DAMGO inhibited the exercise pressor reflex relative to no intrathecal injection or intrathecal saline injection ( P < 0.001, n = 5). We then found that intrathecal injection of two doses of tertiapin-LQ (1 and 10 µg) had no effect on the exercise pressor reflex ( n = 6 and n = 7, respectively; P > 0.05). Importantly, neither dose of tertiapin-LQ prevented the DAMGO-induced inhibition of the exercise pressor reflex. Last, we found that intrathecal injection of ω-conotoxin-GVIA markedly attenuated the exercise pressor reflex ( P < 0.001, n = 7). The cardioaccelerator response to contraction did not appear to be effected in any of the experiments. We conclude that N-type voltage-gated calcium channel inhibition appears to be the mechanism by which MOR activation inhibits the exercise pressor reflex in decerebrate rats.


Asunto(s)
Canales de Calcio Tipo N/metabolismo , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Activación del Canal Iónico , Músculo Esquelético/inervación , Inhibición Neural , Esfuerzo Físico , Receptores Opioides mu/metabolismo , Reflejo , Médula Espinal/metabolismo , Analgésicos Opioides/administración & dosificación , Animales , Bloqueadores de los Canales de Calcio/administración & dosificación , Señalización del Calcio/efectos de los fármacos , Estado de Descerebración , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Inyecciones Espinales , Activación del Canal Iónico/efectos de los fármacos , Masculino , Contracción Muscular , Inhibición Neural/efectos de los fármacos , Neuronas Aferentes/metabolismo , Bloqueadores de los Canales de Potasio/administración & dosificación , Ratas Sprague-Dawley , Receptores Opioides mu/efectos de los fármacos , Reflejo/efectos de los fármacos , Médula Espinal/efectos de los fármacos
6.
J Neurosci ; 35(35): 12217-31, 2015 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-26338332

RESUMEN

Pain management in opioid abusers engenders ethical and practical difficulties for clinicians, often resulting in pain mismanagement. Although chronic opioid administration may alter pain states, the presence of pain itself may alter the propensity to self-administer opioids, and previous history of drug abuse comorbid with chronic pain promotes higher rates of opioid misuse. Here, we tested the hypothesis that inflammatory pain leads to increased heroin self-administration resulting from altered mu opioid receptor (MOR) regulation of mesolimbic dopamine (DA) transmission. To this end, the complete Freund's adjuvant (CFA) model of inflammation was used to assess the neurochemical and functional changes induced by inflammatory pain on MOR-mediated mesolimbic DA transmission and on rat intravenous heroin self-administration under fixed ratio (FR) and progressive ratio (PR) schedules of reinforcement. In the presence of inflammatory pain, heroin intake under an FR schedule was increased for high, but attenuated for low, heroin doses with concomitant alterations in mesolimbic MOR function suggested by DA microdialysis. Consistent with the reduction in low dose FR heroin self-administration, inflammatory pain reduced motivation for a low dose of heroin, as measured by responding under a PR schedule of reinforcement, an effect dissociable from high heroin dose PR responding. Together, these results identify a connection between inflammatory pain and loss of MOR function in the mesolimbic dopaminergic pathway that increases intake of high doses of heroin. These findings suggest that pain-induced loss of MOR function in the mesolimbic pathway may promote opioid dose escalation and contribute to opioid abuse-associated phenotypes. SIGNIFICANCE STATEMENT: This study provides critical new insights that show that inflammatory pain alters heroin intake through a desensitization of MORs located within the VTA. These findings expand our knowledge of the interactions between inflammatory pain and opioid abuse liability, and should help to facilitate the development of novel and safer opioid-based strategies for treating chronic pain.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Dolor , Receptores Opioides mu/metabolismo , Área Tegmental Ventral/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Condicionamiento Operante/efectos de los fármacos , Modelos Animales de Enfermedad , Antagonistas de Aminoácidos Excitadores/farmacología , Glicinérgicos/farmacología , Heroína/administración & dosificación , Hiperalgesia/tratamiento farmacológico , Inflamación/inducido químicamente , Inflamación/complicaciones , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Masculino , Neuronas/efectos de los fármacos , Dolor/tratamiento farmacológico , Dolor/patología , Dolor/psicología , Umbral del Dolor/efectos de los fármacos , Quinoxalinas/farmacología , Ratas , Ratas Sprague-Dawley , Estricnina/farmacología , Sacarosa/administración & dosificación , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/patología
7.
Alcohol Clin Exp Res ; 40(10): 2114-2123, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27508965

RESUMEN

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.


Asunto(s)
Consumo de Bebidas Alcohólicas/fisiopatología , Etanol/administración & dosificación , Núcleo Accumbens/fisiología , Receptores Opioides kappa/fisiología , Receptores Opioides mu/fisiología , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Animales , Relación Dosis-Respuesta a Droga , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Masculino , Microinyecciones , Morfina/administración & dosificación , Morfina/farmacología , Ratas , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inhibidores , Recompensa , Somatostatina/administración & dosificación , Somatostatina/análogos & derivados , Somatostatina/farmacología , Especificidad de la Especie
8.
Pharm Res ; 33(1): 177-85, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26275529

RESUMEN

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.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacocinética , Glutatión/química , Liposomas/química , Polietilenglicoles/química , Animales , Portadores de Fármacos , Composición de Medicamentos , Sistemas de Liberación de Medicamentos , Masculino , Microdiálisis , Neostriado/metabolismo , Fosfatidilcolinas/química , Ratas , Ratas Sprague-Dawley
9.
Bull Exp Biol Med ; 156(4): 423-5, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24771418

RESUMEN

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.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Receptores Opioides/agonistas , Animales , Dióxido de Carbono/metabolismo , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Leucina Encefalina-2-Alanina/administración & dosificación , Mucosa Gástrica/metabolismo , Consumo de Oxígeno , Pirrolidinas/administración & dosificación , Ratas Wistar , Receptores Opioides/fisiología , Estómago/efectos de los fármacos
10.
Mol Pharm ; 10(5): 1533-41, 2013 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-22934681

RESUMEN

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.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Encéfalo/metabolismo , Sistemas de Liberación de Medicamentos , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Analgésicos Opioides/farmacocinética , Animales , Barrera Hematoencefálica , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacocinética , Glutatión/administración & dosificación , Semivida , Infusiones Intravenosas , Liposomas/administración & dosificación , Masculino , Microdiálisis , Polietilenglicoles/administración & dosificación , Ratas , Ratas Sprague-Dawley
11.
J Neurosci ; 31(5): 1591-9, 2011 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-21289167

RESUMEN

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.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Impulso (Psicología) , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inhibidores , Recompensa , Somatostatina/análogos & derivados , Amígdala del Cerebelo/metabolismo , Animales , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Encefalina Leucina/administración & dosificación , Encefalina Leucina/análogos & derivados , Encefalina Leucina/farmacología , Alimentos , Guanidinas , Masculino , Microinyecciones , Morfinanos , Naltrexona/administración & dosificación , Naltrexona/análogos & derivados , Naltrexona/farmacología , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de Narcóticos/farmacología , Pruebas Neuropsicológicas , Ratas , Ratas Long-Evans , Receptores Opioides kappa/antagonistas & inhibidores , Somatostatina/administración & dosificación , Somatostatina/farmacología , Sacarosa
12.
J Neurosci ; 31(37): 13068-77, 2011 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-21917790

RESUMEN

Over the past few years, δ-opioid receptors (DOPRs) and µ-opioid receptors (MOPRs) have been shown to interact with each other. We have previously seen that expression of MOPR is essential for morphine and inflammation to potentiate the analgesic properties of selective DOPR agonists. In vivo, it is not clear whether MOPRs and DOPRs are expressed in the same neurons. Indeed, it was recently proposed that these receptors are segregated in different populations of nociceptors, with MOPRs and DOPRs expressed by peptidergic and nonpeptidergic fibers, respectively. In the present study, the role and the effects of DOPR- and MOPR-selective agonists in two different pain models were compared. Using preprotachykinin A knock-out mice, we first confirmed that substance P partly mediates intraplantar formalin- and capsaicin-induced pain behaviors. These mice had a significant reduction in pain behavior compared with wild-type mice. We then measured the effects of intrathecal deltorphin II (DOPR agonist) and DAMGO (MOPR agonist) on pain-like behavior, neuronal activation, and substance P release following formalin and capsaicin injection. We found that both agonists were able to decrease formalin- and capsaicin-induced pain, an effect that was correlated with a reduction in the number of c-fos-positive neurons in the superficial laminae of the lumbar spinal cord. Finally, visualization of NK(1) (neurokinin 1) receptor internalization revealed that DOPR and MOPR activation strongly reduced formalin- and capsaicin-induced substance P release via direct action on primary afferent fibers. Together, our results indicate that functional MOPRs and DOPRs are both expressed by peptidergic nociceptors.


Asunto(s)
Dolor/fisiopatología , Receptores Opioides delta/fisiología , Receptores Opioides mu/fisiología , Sustancia P/metabolismo , Sustancia P/fisiología , Animales , Capsaicina/antagonistas & inhibidores , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Formaldehído/antagonistas & inhibidores , Inyecciones Espinales , Masculino , Ratones , Ratones Noqueados , Neuronas/fisiología , Neuronas Aferentes/metabolismo , Oligopéptidos/administración & dosificación , Oligopéptidos/farmacología , Dolor/inducido químicamente , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Precursores de Proteínas/genética , Ratas , Ratas Sprague-Dawley , Receptores de Neuroquinina-1/metabolismo , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Médula Espinal/efectos de los fármacos , Médula Espinal/fisiología , Sustancia P/farmacología , Taquicininas/genética
13.
Anesthesiology ; 116(6): 1323-34, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22534246

RESUMEN

BACKGROUND: Peripheral application of opioids reduces inflammatory pain but is less effective in noninflamed tissue of rats and human patients. Hypertonic solutions can facilitate the antinociceptive activity of hydrophilic opioids in noninflamed tissue in vivo. However, the underlying mechanisms are not well understood. We hypothesized that the enhanced efficacy of opioids may be because of opening of the perineurial barrier formed by tight junction-proteins like claudin-1. METHODS: Male Wistar rats were treated intraplantarly with 10% NaCl. Pain behavior (n = 6) and electrophysiological recordings (n = 9 or more) from skin-nerve preparations after local application of the opioid [d-Ala2,N-Me-Phe4,Gly5-ol]enkephalin (DAMGO) were explored. Tight junction-proteins as well as permeability of the barrier were examined by immunohistochemistry and Western blot (n = 3 or more). RESULTS: Local administration of 10% NaCl facilitated increased mechanical nociceptive thresholds in response to DAMGO, penetration of horseradish peroxidase into the nerve, as well as a reduced response of C- but not Aδ-nociceptors to mechanical stimulation after application of DAMGO in the skin-nerve preparation. In noninflamed paw tissue, claudin-1 was expressed in the epidermis, blood vessels, and the perineurium, surrounding neurons immunoreactive for calcitonin gene-related peptide or protein gene product 9.5. Claudin-1 but not claudin-5 or occludin was significantly reduced after pretreatment with 10% NaCl. Intraplantar application of a metalloproteinase inhibitor (GM6001) completely reversed these effects. CONCLUSION: Hypertonic saline opens the perineurial barrier via metalloproteinase activation and claudin-1 regulation, thereby allowing access of hydrophilic drugs to peripheral opioid receptors. This principle may be used to specifically target hydrophilic drugs to peripheral neurons.


Asunto(s)
Analgesia , Analgésicos Opioides/farmacología , Proteínas del Tejido Nervioso/metabolismo , Nervios Periféricos/metabolismo , Uniones Estrechas/efectos de los fármacos , Animales , Western Blotting , Claudina-1 , Fenómenos Electrofisiológicos/efectos de los fármacos , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Técnica del Anticuerpo Fluorescente , Pie , Inyecciones , Masculino , Proteínas de la Membrana/metabolismo , Metaloproteasas/metabolismo , Microscopía Confocal , Fibras Nerviosas Mielínicas/efectos de los fármacos , Fibras Nerviosas Amielínicas/efectos de los fármacos , Nociceptores/efectos de los fármacos , Ocludina , Umbral del Dolor/efectos de los fármacos , Nervios Periféricos/efectos de los fármacos , Ratas , Ratas Wistar , Solución Salina Hipertónica/farmacología
14.
J Neurosci ; 30(42): 14029-35, 2010 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-20962224

RESUMEN

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.


Asunto(s)
Analgésicos Opioides/farmacología , Morfina/farmacología , Recompensa , Área Tegmental Ventral/efectos de los fármacos , Ácido gamma-Aminobutírico/fisiología , Analgésicos Opioides/administración & dosificación , Animales , Condicionamiento Operante/efectos de los fármacos , AMP Cíclico/análogos & derivados , AMP Cíclico/antagonistas & inhibidores , AMP Cíclico/farmacología , AMP Cíclico/fisiología , Electrofisiología , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Inyecciones , Masculino , Ratones , Ratones Endogámicos C57BL , Morfina/administración & dosificación , Receptores Opioides mu/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Tionucleótidos/farmacología
15.
Am J Physiol Regul Integr Comp Physiol ; 301(3): R690-700, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21697523

RESUMEN

The neural control of feeding involves many neuromodulators, including the endogenous opioids that bind µ-opioid receptors (MORs). Injections of the MOR agonist, Damgo, into limbic and hypothalamic forebrain sites increase intake, particularly of palatable foods. Indeed, forebrain Damgo injections increase sucrose-elicited licking but reduce aversive responding (gaping) to quinine, suggesting that MOR activation may enhance taste palatability. A µ-opioid influence on taste reactivity has not been assessed in the brain stem. However, MORs are present in the first-order taste relay, the rostral nucleus of the solitary tract (rNST), and in the immediately subjacent reticular formation (RF), a region known to be essential for consummatory responses. Thus, to evaluate the consequences of rNST/dorsal RF Damgo in this region, we implanted rats with intraoral cannulas, electromyographic electrodes, and brain cannulas aimed at the ventral border of the rNST. Licking and gaping elicited with sucrose, water, and quinine were assessed before and after intramedullary Damgo and saline infusions. Damgo slowed the rate, increased the amplitude, and decreased the size of fluid-induced lick and gape bouts. In addition, the neutral stimulus water, which typically elicits licks, began to evoke gapes. Thus, the current results demonstrate that µ-opioid activation in the rNST/dorsal RF exerts complex effects on oromotor responding that contrast with forebrain effects and are more indicative of a suppressive, rather than a facilitatory effect on ingestion.


Asunto(s)
Analgésicos Opioides/farmacología , Conducta Consumatoria/efectos de los fármacos , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Receptores Opioides mu/agonistas , Formación Reticular/efectos de los fármacos , Núcleo Solitario/efectos de los fármacos , Gusto/efectos de los fármacos , Analgésicos Opioides/administración & dosificación , Análisis de Varianza , Animales , Ingestión de Alimentos/efectos de los fármacos , Electromiografía , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Infusiones Parenterales , Inyecciones , Masculino , Actividad Motora/efectos de los fármacos , Quinina/administración & dosificación , Ratas , Ratas Sprague-Dawley , Receptores Opioides mu/metabolismo , Formación Reticular/metabolismo , Núcleo Solitario/metabolismo , Sacarosa/administración & dosificación , Factores de Tiempo
16.
Neurosci Lett ; 746: 135651, 2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33482313

RESUMEN

Stress-induced activation of locus coeruleus (LC)-norepinephrine (NE) projections to the prefrontal cortex are thought to promote cognitive responses to stressors. LC activation by stressors is modulated by endogenous opioids that restrain LC activation and facilitate a return to baseline activity upon stress termination. Sex differences in this opioid influence could be a basis for sex differences in stress vulnerability. Consistent with this, we recently demonstrated that µ-opioid receptor (MOR) expression is decreased in the female rat LC compared to the male LC, and this was associated with sexually distinct consequences of activating MOR in the LC on cognitive flexibility. Given that the LC-NE system affects cognitive flexibility through its projections to the medial prefrontal cortex (mPFC), the present study quantified and compared the effects of LC-MOR activation on mPFC neural activity in male and female rats. Local field potential (LFPs) were recorded from the mPFC of freely behaving male and female rats before and following local LC microinjection of the MOR agonist, DAMGO, or vehicle. Intra-LC DAMGO altered the LFP power spectrum selectively in male but not female rats, resulting in a time-dependent increase in the power in delta and alpha frequency bands. LC microinfusion of ACSF had no effect on either sex. Together, the results are consistent with previous evidence for decreased MOR function in the female rat LC and demonstrate that this translates to a diminished effect on cortical activity that can account for sex differences in cognitive consequences. Decreased LC-MOR function in females could contribute to greater stress-induced activation of the LC and increased vulnerability of females to hyperarousal symptoms of stress-related neuropsychiatric pathologies.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Locus Coeruleus/metabolismo , Corteza Prefrontal/metabolismo , Receptores Opioides mu/metabolismo , Caracteres Sexuales , Animales , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Femenino , Locus Coeruleus/efectos de los fármacos , Masculino , Microinyecciones/métodos , Corteza Prefrontal/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores Opioides mu/agonistas , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
17.
J Neurophysiol ; 103(1): 409-18, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19906886

RESUMEN

Systemic administration of mu-opioids at clinical doses for analgesia typically slows respiratory rate. Mu-opioid receptors (MORs) on pre-Bötzinger Complex (pre-BötC) respiratory neurons, the putative kernel of respiratory rhythmogenesis, are potential targets. The purpose of this study was to determine the contribution of pre-BötC MORs to the bradypnea produced in vivo by intravenous administration of clinically relevant infusion rates of remifentanil (remi), a short-acting, potent mu-opioid analgesic. In decerebrate dogs, multibarrel micropipettes were used to record pre-BötC neuronal activity and to eject the opioid antagonist naloxone (NAL, 0.5 mM), the glutamate agonist D-homocysteic acid (DLH, 20 mM), or the MOR agonist [D-Ala(2), N-Me-Phe(4), gly-ol(5)]-enkephalin (DAMGO, 100 microM). Inspiratory and expiratory durations (T(I) and T(E)) and peak phrenic nerve activity (PPA) were measured from the phrenic neurogram. The pre-BötC was functionally identified by its rate altering response (typically tachypnea) to DLH microinjection. During intravenous remi-induced bradypnea (approximately 60% decrease in central breathing frequency, f(B)), bilateral injections of NAL in the pre-BötC did not change T(I), T(E), f(B), and PPA. Also, NAL picoejected onto single pre-BötC neurons depressed by intravenous remi had no effect on their discharge. In contrast, approximately 60 microg/kg of intravenous NAL rapidly reversed all remi-induced effects. In a separate group of dogs, microinjections of DAMGO in the pre-BötC increased f(B) by 44%, while subsequent intravenous remi infusion more than offset this DAMGO induced tachypnea. These results indicate that mu-opioids at plasma concentrations that cause profound analgesia produce their bradypneic effect via MORs located outside the pre-BötC region.


Asunto(s)
Analgésicos Opioides/farmacología , Tronco Encefálico/efectos de los fármacos , Neuronas/efectos de los fármacos , Piperidinas/farmacología , Frecuencia Respiratoria/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Analgésicos Opioides/administración & dosificación , Animales , Tronco Encefálico/fisiología , Estado de Descerebración , Perros , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Agonistas de Aminoácidos Excitadores/administración & dosificación , Agonistas de Aminoácidos Excitadores/farmacología , Femenino , Lateralidad Funcional , Homocisteína/administración & dosificación , Homocisteína/análogos & derivados , Homocisteína/farmacología , Masculino , Microinyecciones , Naloxona/administración & dosificación , Naloxona/farmacología , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de Narcóticos/farmacología , Neuronas/fisiología , Nervio Frénico/efectos de los fármacos , Nervio Frénico/fisiología , Piperidinas/administración & dosificación , Remifentanilo , Frecuencia Respiratoria/fisiología , Factores de Tiempo
18.
J Neurophysiol ; 103(1): 172-82, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19889856

RESUMEN

The sphenopalatine ganglion (SPG) neurons represent the parasympathetic branch of the autonomic nervous system involved in controlling cerebral blood flow. In the present study, we examined the coupling mechanism between mu (mu) opioid receptors (MOR) and muscarinic acetylcholine receptors (mAChR) with Ca(2+) channels in acutely dissociated adult rat SPG neurons. Successful MOR activation was recorded in approximately 40-45% of SPG neurons employing the whole cell variant of the patch-clamp technique. In addition, immunofluorescence assays indicated that MOR are not expressed in all SPG neurons while M(2) mAChR staining was evident in all neurons. The concentration-response relationships generated with morphine and [d-Ala2-N-Me-Phe4-Glycol5]-enkephalin (DAMGO) showed IC(50) values of 15.2 and 56.1 nM and maximal Ca(2+) current inhibition of 26.0 and 38.7%, respectively. Activation of MOR or M(2) mAChR with morphine or oxotremorine-methiodide (Oxo-M), respectively, resulted in voltage-dependent inhibition of Ca(2+) currents via coupling with Galpha(i/o) protein subunits. The acute prolonged exposure (10 min) of neurons to morphine or Oxo-M led to the homologous desensitization of MOR and M(2) mAChR, respectively. The prolonged stimulation of M(2) mAChR with Oxo-M resulted in heterologous desensitization of morphine-mediated Ca(2+) current inhibition, and was sensitive to the M(2) mAChR blocker methoctramine. On the other hand, when the neurons were exposed to morphine or DAMGO for 10 min, heterologous desensitization of M(2) mAChR was not observed. These results suggest that in rat SPG neurons activation of M(2) mAChR likely modulates opioid transmission in the brain vasculature to adequately maintain cerebral blood flow.


Asunto(s)
Canales de Calcio/metabolismo , Ganglios Parasimpáticos/fisiología , Neuronas/fisiología , Receptor Muscarínico M2/metabolismo , Receptores Opioides mu/metabolismo , Animales , Células Cultivadas , Diaminas/farmacología , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Ganglios Parasimpáticos/efectos de los fármacos , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Morfina/administración & dosificación , Morfina/farmacología , Agonistas Muscarínicos/farmacología , Narcóticos/administración & dosificación , Narcóticos/farmacología , Neuronas/efectos de los fármacos , Neurotransmisores/administración & dosificación , Neurotransmisores/farmacología , Oxotremorina/farmacología , Parasimpatolíticos/farmacología , Ratas , Ratas Wistar , Receptor Muscarínico M2/agonistas
19.
Am J Physiol Gastrointest Liver Physiol ; 299(2): G494-506, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20489046

RESUMEN

We examined the effects of altering mu-opioid receptor (MOR) activity in the medial subnucleus of the tractus solitarius (mNTS) on several gastric end points including intragastric pressure (IGP), fundus tone, and the receptive relaxation reflex (RRR). Microinjection of the MOR agonist [d-Ala(2),MePhe(4),Gly(ol)(5)]enkephalin (DAMGO; 1-10 fmol) into the mNTS produced dose-dependent decreases in IGP. Microinjection of the endogenous MOR agonists endomorphin-1 and endomorphin-2 (20 fmol) into the mNTS mimicked the effects of 10 fmol DAMGO. Microinjection of 1 and 100 pmol DAMGO into the mNTS produced a triphasic response consisting of an initial decrease, a transient increase, and a persistent decrease in IGP. The increase in IGP appeared to be due to diffusion to the dorsal motor nucleus of the vagus. The effects of 10 fmol DAMGO in the mNTS were blocked by vagotomy and by blockade of MORs, GABA(A) receptors, and ionotropic glutamate receptors in the mNTS. The RRR response was abolished by bilateral microinjection of the opioid receptor antagonist naltrexone into the mNTS and reduced by intravenous administration of naltrexone. Our data demonstrate that 1) activation of MORs in the mNTS with femtomole doses of agonist inhibits gastric motility, 2) the mechanism of MOR effects in the mNTS is through suppression of local GABA activity, and 3) blockade of MORs in the mNTS prevents the RRR response. These data suggest that opioids play an important role in mediating a vagovagal reflex through release of an endogenous opioid in the mNTS, which, in turn, inhibits ongoing local GABA activity and allows vagal sensory input to excite second-order mNTS neurons.


Asunto(s)
Motilidad Gastrointestinal/fisiología , Receptores Opioides/metabolismo , Núcleo Solitario/metabolismo , Estómago/fisiología , Ácido gamma-Aminobutírico/metabolismo , Animales , Regulación hacia Abajo , Vías Eferentes/fisiología , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Antagonistas del GABA/administración & dosificación , Antagonistas de Receptores de GABA-A , Fundus Gástrico/efectos de los fármacos , Fundus Gástrico/fisiología , Motilidad Gastrointestinal/efectos de la radiación , Masculino , Microinyecciones , Tono Muscular/efectos de los fármacos , Naltrexona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Neurotransmisores/administración & dosificación , Oligopéptidos/administración & dosificación , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/metabolismo , Estómago/efectos de los fármacos , Nervio Vago/efectos de los fármacos , Nervio Vago/fisiología
20.
Am J Physiol Heart Circ Physiol ; 299(2): H557-65, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20543079

RESUMEN

Recently, opioid receptors have been shown to be expressed on group III and IV afferents, which comprise the sensory arm of the exercise pressor reflex. Although the stimulation of opioid receptors in the central nervous system has been shown to attenuate the exercise pressor reflex, the effect on the reflex of their stimulation in the periphery is unknown. We therefore tested the hypothesis that the activation of peripheral mu-opioid receptors attenuates the exercise pressor reflex. The pressor responses to static contraction were compared before and after the injection of the mu-opioid receptor agonist [d-Ala(2),N-MePhe(4),Gly-ol(5)]enkephalin (DAMGO; 1 microg) into the abdominal aorta of decerebrated rats in which one femoral artery had been occluded 72 h previously (n = 10) and in control rats whose femoral arteries were freely perfused (n = 8). DAMGO attenuated the peak pressor response to contraction in rats whose femoral arteries had been occluded (before: increase of 34 + or - 3 mmHg and after: increase of 22 + or - 2 mmHg, P = 0.008); the inhibitory effect of DAMGO was prevented by the injection of naloxone (100 microg) into the abdominal aorta (before: increase of 29 + or - 5 mmHg and after: increase of 29 + or - 5 mmHg, P = 0.646, n = 7). An intravenous injection of DAMGO (1 microg, n = 6) had no effect on the peak pressor response to contraction in both groups of rats. DAMGO had no effect on the peak pressor response to contraction in rats whose femoral arteries were freely perfused (before: Delta 23 + or - 4 mmHg, after: Delta 23 + or - 3 mmHg, n = 6) but appeared to have a small effect on topography of the response. DAMGO had no effect on the peak pressor response to tendon stretch in both groups of rats (both P > 0.05). We conclude that the stimulation of peripheral mu-opioid receptors attenuates the exercise pressor reflex in rats whose femoral arteries have been ligated for 72 h.


Asunto(s)
Arteriopatías Oclusivas/metabolismo , Barorreflejo , Arteria Femoral/inervación , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/inervación , Esfuerzo Físico , Receptores Opioides mu/metabolismo , Animales , Arteriopatías Oclusivas/fisiopatología , Barorreflejo/efectos de los fármacos , Enfermedad Crónica , Constricción Patológica , Modelos Animales de Enfermedad , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Arteria Femoral/cirugía , Miembro Posterior , Inyecciones Intraarteriales , Inyecciones Intravenosas , Ligadura , Masculino , Contracción Muscular , Naloxona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Opioides mu/antagonistas & inhibidores , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA