Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
J Ethnopharmacol ; 290: 115088, 2022 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-35149131

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Ben-Cha-Moon-Yai (BMY) remedy used in Thai traditional medicine as an anti-inflammatory, analgesic, and antipyretic agent compromises five herbal root extracts of equal weights: Aegle marmelos (L.) Corrêa (AM), Oroxylum indicum (L.) Kurz (OI), Dimocarpus longan Lour. (DL), Dolichandrone serrulata (Wall. ex DC.) Seem. (DS), and Walsura trichostemon Miq. (WT). AIM OF THE STUDY: To assess the anti-nociceptive and anti-inflammatory effects of the root extracts of all five species of BMY in experimental animal (mouse) models to ensure the rational use of herbal products in Thai traditional medicine. MATERIALS AND METHODS: Root extracts prepared by ethanol and water extraction were used for the biological assays in animal models at five dose levels: 25, 50,100,200 & 400 mg/kg. The anti-nociceptive activity was evaluated based on hot-plate latency, duration of paw licking induced by formalin, and abdominal writhing induced by acetic acid. Carrageenan- and prostaglandin-induced paw oedema models were used to determine the anti-inflammatory activity. RESULTS: The oral administration of AM, DS and WT root extracts displayed significant analgesic effects in the hot-plate test, both phases (early and late) of formalin test and acetic-acid induced writhing test at different dose levels. OI and DL only produced significant analgesia in the late phase of the formalin test and writhing test. The pretreatment of animals with the non-selective opioid receptor antagonist naloxone, reverse AM, DS and WT induced-antinociceptive activity. In both carrageenan and prostaglandin-induced paw oedema tests, all five herbal plant root extracts significantly reduced paw oedema at 3 h or more at different dose levels. Rotarod test results showed no effects of five herbal plant root extracts on the balance and the motor coordination at the highest dose level evaluated (400 mg/kg). CONCLUSION: The root extracts of AM, DS, and WT possess both central and peripheral anti-nociceptive properties, while OI and DL possess only peripheral analgesic properties. All five root extracts own anti-inflammatory properties, which might be due to their activity on the prostaglandin system. Altogether these findings ensure the rational use of BMY remedy in Thai traditional medicine.


Asunto(s)
Antiinflamatorios/farmacología , Nocicepción/efectos de los fármacos , Péptidos Opioides/efectos de los fármacos , Extractos Vegetales/farmacología , Raíces de Plantas , Prostaglandinas/metabolismo , Aegle , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Indometacina/farmacología , Masculino , Medicina Tradicional de Asia Oriental , Ratones , Ratones Endogámicos ICR , Morfina/farmacología , Naloxona/farmacología , Dimensión del Dolor , Preparaciones de Plantas/farmacología , Sapindaceae
2.
Eur J Pharmacol ; 871: 172918, 2020 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-31958457

RESUMEN

Mu opioid receptor (MOPr) agonists are thought to produce analgesia via modulation of G-protein-coupled intracellular signalling pathways whereas the ß-arrestin2 pathway is proposed to mediate opioid-related adverse effects. Here, we report the antinociception, constipation and respiratory depressant profile of CYX-6, a potent MOPr agonist that is also a delta and a kappa opioid receptor (DOPr/KOPr) antagonist and that lacks ß-arrestin2 recruitment at each of the MOPr, DOPr and the KOPr. In anaesthetised male Sprague Dawley rats, an intracerebroventricular (i.c.v.) guide cannula was stereotaxically implanted. After 5-7 days post-surgical recovery, rats received a single i.c.v. bolus dose of CYX-6 (3-30 nmol), morphine (100 nmol) or vehicle. Antinociception was assessed using the warm water tail flick test (52.5 ± 0.5 °C). Constipation was assessed using the charcoal meal gut motility test and the castor oil-induced diarrhoea test. Respiratory depression was measured by whole-body plethysmography in awake, freely moving animals, upon exposure to a hypercapnic gas mixture (8% CO2, 21% O2 and 71% N2). The intrinsic pharmacology of CYX-6 given by the i.c.v. route in rats showed that it produced dose-dependent antinociception. It also produced respiratory stimulation rather than depression and it had a minimal effect on intestinal motility in contrast to the positive control, morphine. CYX-6 is an endomorphin-2 analogue that dissociates antinociception from constipation and respiratory depression in rats. Our findings provide useful insight to inform the discovery and development of novel opioid analgesics with a superior tolerability profile compared with morphine.


Asunto(s)
Analgésicos Opioides/farmacología , Estreñimiento/inducido químicamente , Morfina/farmacología , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides kappa/antagonistas & inhibidores , Receptores Opioides mu/agonistas , Insuficiencia Respiratoria/inducido químicamente , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/metabolismo , Animales , Infusiones Intraventriculares , Ligandos , Masculino , Morfina/efectos adversos , Péptidos Opioides/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Nociceptina
3.
ACS Chem Neurosci ; 10(3): 1615-1626, 2019 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-30614675

RESUMEN

Leu-enkephalin and d-Ala2-Leu-enkephalin were modified at their N- and C-termini with guanidyl and tetrazole groups. The resulting molecules were prepared in solution or by solid phase peptide synthesis. The affinity of the different analogues at mu (MOP) and delta opioid receptors (DOP) was then assessed by competitive binding in stably transfected DOP and MOP HEK293 cells. Inhibition of cAMP production and recruitment of ß-arrestin were also investigated. Finally, lipophilicity (logD7.4) and plasma stability of each compound were measured. Compared to the native ligands, we found that the replacement of the terminal carboxylate by a tetrazole slightly decreased both the affinity at mu and delta opioid receptors as well as the half-life. By contrast, replacing the ammonium at the N-terminus with a guanidyl significantly improved the affinity, the potency, as well as the lipophilicity and the stability of the resulting peptides. Replacing the glycine residue with a d-alanine in position 2 consistently improved the potency as well as the stability of the analogues. The best peptidomimetic of the whole series, guanidyl-Tyr-d-Ala-Gly-Phe-Leu-tetrazole, displayed sub-nanomolar affinity and an increased lipophilicity. Moreover, it proved to be stable in plasma for up to 24 h, suggesting that the modifications are protecting the compound against protease degradation.


Asunto(s)
Encefalina Leucina/análogos & derivados , Oligopéptidos/farmacología , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Animales , Células HEK293 , Humanos , Péptidos Opioides/efectos de los fármacos , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo
4.
Acta Neuropathol Commun ; 6(1): 73, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30086801

RESUMEN

After spinal cord injury (SCI), neutrophil elastase (NE) released at injury site disrupts vascular endothelium integrity and stabilization. Angiopoietins (ANGPTs) are vascular growth factors that play an important role in vascular stabilization. We hypothesized that neutrophil elastase is one of the key determinants of vascular endothelium disruption/destabilization and affects angiopoietins expression after spinal cord injury. To test this, tubule formation and angiopoietins expression were assessed in endothelial cells exposed to different concentrations of recombinant neutropil elastase. Then, the expression of angiopoietin-1, angiopoietin-2, and neutrophil elastase was determined at 3 h and at 1, 3, 5, 7, 14, 21, and 28 days in a clinically relevant model of moderate compression (35 g for 5 min at T10) spinal cord injury. A dichotomy between the levels of angiopoietin-1 and angiopoietin-2 was observed; thus, we utilized a specific neutrophil elastase inhibitor (sivelestat sodium; 30 mg/kg, i.p., b.i.d.) after spinal cord injury. The expression levels of neutropil elastase and angiopoietin-2 increased, and that of angiopoietin-1 decreased after spinal cord injury in rats. The sivelestat regimen, optimized via a pharmacokinetics study, had potent effects on vascular stabilization by upregulating angiopoietin-1 via the AKT pathway and preventing tight junction protein degradation. Moreover, sivelestat attenuated the levels of inflammatory cytokines and chemokines after spinal cord injury and hence subsequently alleviated secondary damage observed as a reduction in glial scar formation and the promotion of blood vessel formation and stabilization. As a result, hindlimb locomotor function significantly recovered in the sivelestat-treated animals as determined by the Basso, Beattie, and Bresnahan scale and footprint analyses. Furthermore, sivelestat treatment attenuated neuropathic pain as assessed by responses to von Frey filaments after spinal cord injury. Thus, our result suggests that inhibiting neutropil elastase by administration of sivelestat is a promising therapeutic strategy to inhibit glial scar and promote functional recovery by upregulating angiopoietin-1 after spinal cord injury.


Asunto(s)
Angiopoyetina 1/metabolismo , Cicatriz/tratamiento farmacológico , Cicatriz/etiología , Elastasa de Leucocito/farmacología , Traumatismos de la Médula Espinal/complicaciones , Traumatismos de la Médula Espinal/patología , Angiopoyetina 2/metabolismo , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/fisiología , Glicina/análogos & derivados , Glicina/farmacología , Humanos , Laminina/metabolismo , Elastasa de Leucocito/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Ocludina/metabolismo , Péptidos Opioides/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Recuperación de la Función/efectos de los fármacos , Inhibidores de Serina Proteinasa/farmacología , Sulfonamidas/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Proteína de la Zonula Occludens-1/metabolismo , Nociceptina
5.
J Pharmacol Exp Ther ; 366(3): 509-518, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29945931

RESUMEN

The immunomodulatory prodrug 2-amino-2-(2-[4-octylphenyl]ethyl)-1,3-propanediol (FTY720), which acts as an agonist for sphingosine-1-phosphate (S1P) receptors (S1PR) when phosphorylated, is proposed as a novel pain therapeutic. In this study, we assessed FTY720-mediated antinociception in the radiant heat tail-flick test and in the chronic constriction injury (CCI) model of neuropathic pain in mice. FTY720 produced antinociception and antiallodynia, respectively, and these effects were dose-dependent and mimicked by the S1PR1-selective agonist CYM-5442. Repeated administration of FTY720 for 1 week produced tolerance to acute thermal antinociception, but not to antiallodynia in the CCI model. S1PR-stimulated [35S]GTPγS autoradiography revealed apparent desensitization of G protein activation by S1P or the S1PR1 agonist 5-[4-phenyl-5-(trifluoromethyl)-2-thienyl]-3-[3-(trifluoromethyl)phenyl]-1,2,4-oxadiazole (SEW-2871) throughout the brain. Similar results were seen in spinal cord membranes, whereby the Emax value of S1PR-stimulated [35S]GTPγS binding was greatly reduced in repeated FTY720-treated mice. These results suggest that S1PR1 is a primary target of FTY720 in alleviating both acute thermal nociception and chronic neuropathic nociception. Furthermore, the finding that tolerance develops to antinociception in the tail-flick test but not in chronic neuropathic pain suggests a differential mechanism of FTY720 action between these models. The observation that repeated FTY720 administration led to desensitized S1PR1 signaling throughout the central nervous system suggests the possibility that S1PR1 activation drives the acute thermal antinociceptive effects, whereas S1PR1 desensitization mediates the following: 1) tolerance to thermal antinociceptive actions of FTY720 and 2) the persistent antiallodynic effects of FTY720 in neuropathic pain by producing functional antagonism of pronociceptive S1PR1 signaling.


Asunto(s)
Clorhidrato de Fingolimod/farmacología , Neuralgia/tratamiento farmacológico , Neuralgia/metabolismo , Péptidos Opioides/efectos de los fármacos , Receptores de Lisoesfingolípidos/metabolismo , Temperatura , Animales , Modelos Animales de Enfermedad , Clorhidrato de Fingolimod/uso terapéutico , Masculino , Ratones , Ratones Endogámicos ICR , Neuralgia/fisiopatología , Receptores de Lisoesfingolípidos/agonistas , Nociceptina
6.
Am J Med ; 129(7): 755-8, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27039954

RESUMEN

BACKGROUND: Recent evidence supports the beneficial effects of mindfulness meditation on pain. However, the neural mechanisms underlying this effect remain poorly understood. We used an opioid blocker to examine whether mindfulness meditation-induced analgesia involves endogenous opioids. METHODS: Fifteen healthy experienced mindfulness meditation practitioners participated in a double-blind, randomized, placebo-controlled, crossover study. Participants rated the pain and unpleasantness of a cold stimulus prior to and after a mindfulness meditation session. Participants were then randomized to receive either intravenous naloxone or saline, after which they meditated again, and rated the same stimulus. RESULTS: A (3) × (2) repeated-measurements analysis of variance revealed a significant time effect for pain and unpleasantness scores (both P <.001) as well as a significant condition effect for pain and unpleasantness (both P <.2). Post hoc comparisons revealed that pain and unpleasantness scores were significantly reduced after natural mindfulness meditation and after placebo, but not after naloxone. Furthermore, there was a positive correlation between the pain scores following naloxone vs placebo and participants' mindfulness meditation experience. CONCLUSIONS: These findings show, for the first time, that meditation involves endogenous opioid pathways, mediating its analgesic effect and growing resilient with increasing practice to external suggestion. This finding could hold promising therapeutic implications and further elucidate the fine mechanisms involved in human pain modulation.


Asunto(s)
Frío , Meditación/métodos , Atención Plena/métodos , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Péptidos Opioides/efectos de los fármacos , Manejo del Dolor , Estudios Cruzados , Método Doble Ciego , Humanos , Péptidos Opioides/metabolismo , Dimensión del Dolor
7.
Subst Abuse Treat Prev Policy ; 10: 29, 2015 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-26238243

RESUMEN

INTRODUCTION: Zinc chelators were shown to facilitate some opioid-withdrawal signs in animals. Zinc deficiency, which affects more than 15% the world's population, is also common among opioid consumers and opioid-treated animals exhibit misbalances of zinc distribution. AIM: The present study focuses on how zinc ions interfere with opioid dependence/addiction and analgesia, trying to preliminary discuss if zinc supplementation in opioid-users should be recommended in order to reduce the risk of addiction. METHODS: All relevant literature was searched up to April 2015. The search was performed using the term "zinc" plus combinations of following terms: "opioid receptors", "opioid" or representatives of this class, "addiction", "dependence", "analgesia", and "pain". Human, animal, in vitro studies and reviews were including. RESULTS: Both human and animal studies revealed decreased serum zinc under opioid-administration conditions, attributed mainly to increased urinary elimination (humans) or redistribution (animals). Moreover, animal studies revealed decreased brain zinc levels in morphine-treated animals, with increased zinc hepatic levels, but also an enhancement of endogenous opioid system activity and a possible reduction of morphine withdrawal by zinc. In vitro studies revealed reduction of opioid ligands binding to receptors by zinc. However, the very few in vivo animal studies on opioid analgesia revealed controversial results, as zinc demonstrated clear analgesic effect, but zinc associated to opioids doesn't result in a potentiation of the analgesic effect. CONCLUSION: Zinc dietary supplementation in patients treated with opioids for cancer-related chronic pain should be considered, due to the high incidence of zinc deficiency, also well-documented in opioid consumers. The low toxicity of orally-administered zinc also pleads for this idea. The main contra-argument to zinc administration in opioid-treated persons is related to the way zinc influences opioid-induced analgesia.


Asunto(s)
Analgesia , Trastornos Relacionados con Opioides/fisiopatología , Síndrome de Abstinencia a Sustancias/fisiopatología , Zinc/metabolismo , Zinc/farmacología , Animales , Encéfalo/metabolismo , Humanos , Hígado/metabolismo , Péptidos Opioides/efectos de los fármacos , Péptidos Opioides/metabolismo , Receptores Opioides/efectos de los fármacos , Receptores Opioides/metabolismo , Zinc/análisis
8.
Rev Med Chir Soc Med Nat Iasi ; 118(3): 692-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25341287

RESUMEN

No doubt, alcoholism represents nowadays the toxicomany with the highest expansion rate among all population groups, being recognized by the specialists from the medical, social, economic and legal field as a true "toxic pandemy". Researchers consider ethanol, this small but highly aggressive molecule, to have supremacy if we were to consider the number of pages dedicated to it worldwide on daily bases, in the medical or any other specialty literature. Nonetheless, the large volume of data regarding ethanol toxicity does not seen to simplify things, on the contrary it points out new information about the its negative effects on human body. Ethanol represents a toxic that is rapidly and completely absorbed in the intestinal tract being distributed to most tissues and organs; ethanol is recognized as an enzymatic inductor of its own metabolization but also of the metabolization of numerous therapeutic agents.


Asunto(s)
Alcoholismo/fisiopatología , Alcoholismo/rehabilitación , Alcoholismo/epidemiología , Alcoholismo/metabolismo , Terapia Conductista , Calcio/metabolismo , Membrana Celular/efectos de los fármacos , Células/efectos de los fármacos , Medicina Basada en la Evidencia , Salud Global , Humanos , Intestinos/efectos de los fármacos , Hígado/efectos de los fármacos , Neurotransmisores/metabolismo , Péptidos Opioides/efectos de los fármacos , Prevalencia , Pronóstico , Rumanía/epidemiología , Ácido gamma-Aminobutírico/efectos de los fármacos
9.
J Neurol Sci ; 347(1-2): 82-9, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25282545

RESUMEN

Transcutaneous electrical nerve stimulation (TENS) is a non-pharmacological therapy for the treatment of pain. The present work investigated the effect of cannabidiol, naloxone and diazepam in combination with 10 Hz and 150 Hz TENS. Male Wistar rats were submitted to the tail-flick test (baseline), and each rodent received an acute administration (intraperitoneal) of naloxone (3.0mg/kg), diazepam (1.5mg/kg) or cannabidiol (0.75 mg/kg, 1.5mg/kg, 3.0mg/kg, 4.5mg/kg, 6.0mg/kg and 12.0mg/kg); 10 min after the acute administration, 10 Hz or 150 Hz TENS or a sham procedure was performed for 30 min. Subsequently, tail-flick measures were recorded over a 90-min period, at 5-min intervals. 10 Hz TENS increased the nociceptive threshold during the 90-min period. This antinociceptive effect was reversed by naloxone pre-treatment, was not altered by diazepam pre-treatment and was abolished by cannabidiol pre-treatment (1.5mg/kg). Moreover, 150 Hz TENS increased tail-flick latencies by 35 min post-treatment, which was partially inhibited by naloxone pre-treatment and totally inhibited by cannabidiol (1.5mg/kg). These data suggest the involvement of the endogenous opioid system and the cannabinoid-mediated neuromodulation of the antinociception induced by transcutaneous electrostimulation at 10 Hz and 150 Hz TENS.


Asunto(s)
Cannabidiol/metabolismo , Dolor Nociceptivo/metabolismo , Dolor Nociceptivo/terapia , Péptidos Opioides/metabolismo , Sistema Nervioso Periférico/fisiología , Estimulación Eléctrica Transcutánea del Nervio , Animales , Cannabidiol/farmacología , Diazepam/farmacología , Hipnóticos y Sedantes/farmacología , Masculino , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Péptidos Opioides/efectos de los fármacos , Dimensión del Dolor , Sistema Nervioso Periférico/efectos de los fármacos , Ratas , Ratas Wistar , Estimulación Eléctrica Transcutánea del Nervio/métodos
10.
Neuropeptides ; 48(4): 249-56, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24877897

RESUMEN

Angiotensin and endogenous opioids are important bioactive neuropeptides, which are widely distributed in the brain and peripheral regions to produce diverse biological and neurobiological activities. An endogenous opioid system includes proopiomelanocortin-derived enkephalin, dynorphin and endorphin that act on their specific receptors such as delta (δ), kappa (κ) and mu (µ) receptors. Research evidence demonstrates significant positive as well as negative interactions between renin angiotensin system (RAS) and endogenous opioids in the brain and periphery. The diverse actions of Ang II are possibly mediated indirectly through endogenous opioids, while opioids are also shown to activate RAS components suggesting the up-regulation of each system in concern with each other. On the contrary, there are reports suggesting a negative correlation between RAS and opioid system. Research evidence also supports the notion that Ang II acts as anti-opioid peptide to decrease the actions of opioids. Moreover, opioids-induced decline in angiotensin release and functioning has also been reported. Co-administration of ACE inhibitors with opioids exhibits significant interactions possibly due to decreased metabolism of opioids leading to potentiation of their actions. The present review describes the complexities of positive and negative interactions between RAS and opioids along with possible mechanisms responsible for these interactions.


Asunto(s)
Péptidos Opioides/fisiología , Receptores Opioides/fisiología , Sistema Renina-Angiotensina/fisiología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Humanos , Péptidos Opioides/efectos de los fármacos , Receptores Opioides/efectos de los fármacos , Sistema Renina-Angiotensina/efectos de los fármacos
11.
J Neuroendocrinol ; 26(5): 329-40, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24617903

RESUMEN

Orphanin FQ/nociceptin (OFQ/N) inhibits the activity of pro-opiomelanocortin (POMC) neurones located in the hypothalamic arcuate nucleus (ARH) that regulate female sexual behaviour and energy balance. We tested the hypothesis that gonadal steroids differentially modulate the ability of OFQ/N to inhibit these cells via presynaptic inhibition of transmitter release and postsynaptic activation of G protein-gated, inwardly-rectifying K(+) (GIRK)-1 channels. Whole-cell patch clamp recordings were performed in hypothalamic slices prepared from ovariectomised rats. OFQ/N (1 µm) decreased the frequency of miniature excitatory postsynaptic currents (mEPSCs) and miniature inhibitory postsynaptic currents (mIPSCs), and also caused a robust outward current in the presence of tetrodotoxin, in ARH neurones from vehicle-treated animals. A priming dose of oestradiol benzoate (EB; 2 µg) increased basal mEPSC frequency, markedly diminished both the OFQ/N-induced decrease in mEPSC frequency and the activation of GIRK-1 currents, and potentiated the OFQ/N-induced decrease in mIPSC frequency. Steroid treatment regimens that facilitate sexual receptivity reinstate the basal mEPSC frequency, the OFQ/N-induced decrease in mEPSC frequency and the activation of GIRK-1 currents to levels observed in vehicle-treated controls, and largely abolish the ability of OFQ/N to decrease mIPSC frequency. These effects were observed in an appreciable population of identified POMC neurones, almost one-half of which projected to the medial preoptic nucleus. Taken together, these data reveal that gonadal steroids influence the pleiotropic actions of OFQ/N on ARH neurones, including POMC neurones, in a disparate manner. These temporal changes in OFQ/N responsiveness further implicate this neuropeptide system as a critical mediator of the gonadal steroid regulation of reproductive behaviour.


Asunto(s)
Hormonas Esteroides Gonadales/farmacología , Péptidos Opioides/efectos de los fármacos , Conducta Sexual Animal/efectos de los fármacos , Animales , Estradiol/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Femenino , Ovariectomía , Técnicas de Placa-Clamp , Canales de Potasio/efectos de los fármacos , Proopiomelanocortina/fisiología , Progesterona/farmacología , Ratas , Ratas Long-Evans , Receptores Opioides/biosíntesis , Receptores Opioides/genética , Receptor de Nociceptina , Nociceptina
12.
Psychol Med ; 41(2): 385-93, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20444308

RESUMEN

BACKGROUND: The expanded suffocation false alarm theory (SFA) hypothesizes that dysfunction in endogenous opioidergic regulation increases sensitivity to CO2, separation distress and panic attacks. In panic disorder (PD) patients, both spontaneous clinical panics and lactate-induced panics markedly increase tidal volume (TV), whereas normals have a lesser effect, possibly due to their intact endogenous opioid system. We hypothesized that impairing the opioidergic system by naloxone could make normal controls parallel PD patients' response when lactate challenged. Whether actual separations and losses during childhood (childhood parental loss, CPL) affected naloxone-induced respiratory contrasts was explored. Subjective panic-like symptoms were analyzed although pilot work indicated that the subjective aspect of anxious panic was not well modeled by this specific protocol. METHOD: Randomized cross-over sequences of intravenous naloxone (2 mg/kg) followed by lactate (10 mg/kg), or saline followed by lactate, were given to 25 volunteers. Respiratory physiology was objectively recorded by the LifeShirt. Subjective symptomatology was also recorded. RESULTS: Impairment of the endogenous opioid system by naloxone accentuates TV and symptomatic response to lactate. This interaction is substantially lessened by CPL. CONCLUSIONS: Opioidergic dysregulation may underlie respiratory pathophysiology and suffocation sensitivity in PD. Comparing specific anti-panic medications with ineffective anti-panic agents (e.g. propranolol) can test the specificity of the naloxone+lactate model. A screen for putative anti-panic agents and a new pharmacotherapeutic approach are suggested. Heuristically, the experimental unveiling of the endogenous opioid system impairing effects of CPL and separation in normal adults opens a new experimental, investigatory area.


Asunto(s)
Ansiedad de Separación/metabolismo , Péptidos Opioides/metabolismo , Trastorno de Pánico/metabolismo , Adolescente , Adulto , Estudios Cruzados , Femenino , Humanos , Ácido Láctico , Masculino , Persona de Mediana Edad , Naloxona/farmacología , Péptidos Opioides/efectos de los fármacos , Fenómenos Fisiológicos Respiratorios/efectos de los fármacos
13.
Dev Psychobiol ; 52(8): 740-54, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21117244

RESUMEN

The present study investigated if oral exposure to milk or amniotic fluid (AF) alters responsiveness to sensory stimulation in the neonatal rat, and whether these effects are mediated by the opioid system. Facial wiping evoked by intraoral lemon infusion was used as a measure of sensory responsiveness. Pups were tested in a supine posture, because they showed more paw-face strokes during facial wiping than pups tested prone (Experiment 1). Moreover, pups orally exposed to milk (Experiment 2) or AF (Experiment 3) showed a diminished wiping response to lemon compared to controls exposed to water. Blockade of opioid receptors with the nonselective antagonist naltrexone (Experiment 4) or the kappa antagonist nor-binaltorphimine (Experiment 5) reinstated higher levels of facial wiping after AF exposure. These findings confirm developmental continuity between fetal and neonatal behavioral responses to AF and the ability of AF to induce activity at kappa receptors of the endogenous opioid system.


Asunto(s)
Líquido Amniótico , Conducta Animal/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Péptidos Opioides/efectos de los fármacos , Receptores Opioides/efectos de los fármacos , Administración Oral , Análisis de Varianza , Animales , Animales Recién Nacidos , Animales Lactantes , Femenino , Masculino , Leche , Naltrexona/análogos & derivados , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Péptidos Opioides/antagonistas & inhibidores , Embarazo , Ratas , Ratas Sprague-Dawley , Estimulación Química
14.
J Urol ; 184(4): 1529-35, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20723927

RESUMEN

PURPOSE: Painful bladder disorders vary in intensity with the menstrual cycle in women. We evaluated the influence of the correlate in rats (the estrous cycle) on the nociceptive visceromotor reflex to bladder distention in the presence/absence of inflammation and of spinal opioid blockade. MATERIALS AND METHODS: We recorded visceromotor reflexes as electromyogram responses of the abdominal musculature to graded (10 to 60 mm Hg) bladder distention in anesthetized female rats in the presence of intrathecal saline or naloxone (10 µg) 1 day after receiving intravesical zymosan or anesthesia alone. RESULTS: In saline treated rats visceromotor reflexes to bladder distention were significantly greater in those with an inflamed vs a noninflamed bladder when examined together. When separated into phases, rats with bladder inflammation showed complex estrous cycle effects with significantly greater visceromotor reflexes to bladder distention during metestrus and proestrus than diestrus. In naloxone treated rats visceromotor reflexes to bladder distention were significantly greater in those with an inflamed vs a noninflamed bladder when examined together. Naloxone enhanced the overall magnitude of visceromotor reflexes to bladder distention in the inflamed and noninflamed conditions. The magnitude of visceromotor reflexes to bladder distention in noninflamed and inflamed conditions in the presence of naloxone was estrous phase dependent in the order, estrus >metestrus >diestrus >proestrus. Similar findings were apparent on analysis of data on responses at threshold intensity (30 mm Hg). CONCLUSIONS: Data suggest that circulating hormones present during the estrous cycle alter bladder reactivity and opioid modulatory systems to maintain constancy of input from the bladder to the central nervous system.


Asunto(s)
Ciclo Estral/fisiología , Péptidos Opioides/fisiología , Reflejo Abdominal/fisiología , Vejiga Urinaria/fisiología , Animales , Femenino , Inflamación , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Péptidos Opioides/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
15.
Pain ; 149(2): 254-262, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20206445

RESUMEN

Endothelin-1 (ET-1) produced by various cancers is known to be responsible for inducing pain. While ET-1 binding to ETAR on peripheral nerves clearly mediates nociception, effects from binding to ETBR are less clear. The present study assessed the effects of ETBR activation and the role of endogenous opioid analgesia in carcinoma pain using an orthotopic cancer pain mouse model. mRNA expression analysis showed that ET-1 was nearly doubled while ETBR was significantly down-regulated in a human oral SCC cell line compared to normal oral keratinocytes (NOK). Squamous cell carcinoma (SCC) cell culture treated with an ETBR agonist (10(-4)M, 10(-5)M, and 10(-6) M BQ-3020) significantly increased the production of beta-endorphin without any effects on leu-enkephalin or dynorphin. Cancer inoculated in the hind paw of athymic mice with SCC induced significant pain, as indicated by reduction of paw withdrawal thresholds in response to mechanical stimulation, compared to sham-injected and NOK-injected groups. Intratumor administration of 3mg/kg BQ-3020 attenuated cancer pain by approximately 50% up to 3h post-injection compared to PBS-vehicle and contralateral injection, while intratumor ETBR antagonist BQ-788 treatment (100 and 300microg/kg and 3mg/kg) had no effects. Local naloxone methiodide (500microg/kg) or selective mu-opioid receptor antagonist (CTOP, 500microg/kg) injection reversed ETBR agonist-induced antinociception in cancer animals. We propose that these results demonstrate that peripheral ETBR agonism attenuates carcinoma pain by modulating beta-endorphins released from the SCC to act on peripheral opioid receptors found in the cancer microenvironment.


Asunto(s)
Endotelina-1/metabolismo , Nociceptores/metabolismo , Péptidos Opioides/metabolismo , Dolor/metabolismo , Receptor de Endotelina B/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Endotelina-1/genética , Endotelinas/farmacología , Humanos , Ratones , Ratones Desnudos , Antagonistas de Narcóticos/farmacología , Trasplante de Neoplasias/métodos , Neoplasias/complicaciones , Neoplasias/metabolismo , Neoplasias/fisiopatología , Nociceptores/efectos de los fármacos , Oligopéptidos/farmacología , Péptidos Opioides/efectos de los fármacos , Dolor/tratamiento farmacológico , Dolor/etiología , Fragmentos de Péptidos/farmacología , Piperidinas/farmacología , Receptor de Endotelina B/agonistas , Receptores Opioides/efectos de los fármacos , Receptores Opioides/metabolismo , Células Receptoras Sensoriales/efectos de los fármacos , Regulación hacia Arriba/genética , betaendorfina/efectos de los fármacos , betaendorfina/metabolismo
16.
CNS Neurol Disord Drug Targets ; 9(1): 87-93, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20201819

RESUMEN

Nociceptin (known also as orphanin FQ) is the most recently discovered member of the endogenous opioid peptide family, albeit nearly 15 years ago. Nociceptin renders or influences many behavioral, psychological and neurobiological processes, including memory, anxiety, stress and reward. Since its discovery, results of a steady stream of studies have suggested that endogenous nociceptin might be involved in responses to addictive drugs, and that targeting the nociceptin system may be beneficial in treating addictions. The current review summarizes and critically appraises those studies, particularly those that point to an application in treating alcoholism. Overall, most, studies suggest that the endogenous nociceptin system has a physiological role in mediating or regulating behavioral responses to alcohol, and that activating nociceptin receptors suppresses ongoing alcohol consumption or reinstatement of responding for alcohol. These findings encourage the development of therapies targeted at the nociceptin system for the treatment of alcoholism in humans, though a minor number of studies showing continuous activation of the nociceptin receptor can produce increased, rather than reduced, alcohol consumption emphasize the necessity of further investigation.


Asunto(s)
Trastornos del Sistema Nervioso Inducidos por Alcohol/tratamiento farmacológico , Alcoholismo/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Péptidos Opioides/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Trastornos del Sistema Nervioso Inducidos por Alcohol/metabolismo , Trastornos del Sistema Nervioso Inducidos por Alcohol/fisiopatología , Alcoholismo/metabolismo , Alcoholismo/fisiopatología , Animales , Conducta/efectos de los fármacos , Conducta/fisiología , Encéfalo/metabolismo , Encéfalo/fisiopatología , Química Encefálica/efectos de los fármacos , Química Encefálica/fisiología , Humanos , Péptidos Opioides/metabolismo , Receptores Opioides/efectos de los fármacos , Receptores Opioides/metabolismo , Recompensa , Transducción de Señal/fisiología , Receptor de Nociceptina , Nociceptina
17.
Life Sci ; 83(21-22): 693-9, 2008 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-18930741

RESUMEN

Several approaches have been taken for these in vivo studies. In many studies, the use of semi-quantitative immuno-electron microscopy is the approach of choice. Endogenous opioid receptors display differential subcellular distributions with mu opioid receptor (MOPR) being mostly present on the plasma membrane and delta-opioid receptor (DOPR) and kappa-opioid receptor (KOPR) having a significant intracellular pool. Etorphine and DAMGO cause endocytosis of the MOPR, but morphine does not, except in some dendrites. Interestingly, chronic inflammatory pain and morphine treatment promote trafficking of intracellular DOPR to the cell surface which may account for the enhanced antinociceptive effects of DOPR agonists. KOPR has been reported to be associated with secretory vesicles in the posterior pituitary and translocated to the cell surface upon salt loading along with the release of vasopressin. The study of endogenous opioid receptors using in vivo models has produced some interesting results that could not have been anticipated in vitro. In vivo studies, therefore, are essential to provide insight into the mechanisms underlying opioid receptor regulation.


Asunto(s)
Péptidos Opioides/fisiología , Receptores de Superficie Celular/fisiología , Analgésicos Opioides/farmacología , Animales , Humanos , Péptidos Opioides/efectos de los fármacos , Péptidos Opioides/genética , Receptores de Superficie Celular/efectos de los fármacos , Receptores de Superficie Celular/genética , Receptores Opioides delta/efectos de los fármacos , Receptores Opioides delta/genética , Receptores Opioides delta/fisiología , Receptores Opioides kappa/efectos de los fármacos , Receptores Opioides kappa/genética , Receptores Opioides kappa/fisiología , Receptores Opioides mu/efectos de los fármacos , Receptores Opioides mu/genética , Receptores Opioides mu/fisiología , Transducción de Señal , Fracciones Subcelulares
18.
Psychopharmacology (Berl) ; 201(2): 261-71, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18688603

RESUMEN

RATIONALE: There is experimental evidence that indicates that the endogenous opioid system of the central nucleus of the amygdala (CeA) may mediate some of the reinforcing effects of ethanol. However, the precise interactions of ethanol with the endogenous opioid system at the level of the CeA have not been investigated. OBJECTIVES: The aim of the current study was to investigate the hypothesis that acute systemic ethanol administration will increase the release of endogenous opioid peptides at the level of the CeA in a time- and dose-dependent manner. MATERIALS AND METHODS: Rats were implanted with a unilateral guide cannula to aim microdialysis probes at the CeA. Intraperitoneal injections of saline and various doses of ethanol (0.8, 1.6, 2.0, 2.4, and 2.8 g ethanol/kg body weight) were administered to the rats. Dialysate samples were collected at 30-min intervals at distinct time points prior to and following treatment. Radioimmunoassays specific for beta-endorphin, met-enkephalin, and dynorphin A1-8 were used to determine the effect of ethanol on the content of the opioid peptides in the dialysate. RESULTS: We report that the 2.8-g/kg dose of ethanol induced a long-lasting increase in beta-endorphin release from 60 min onwards following administration and, later, an ongoing increase in dynorphin A1-8 release. None of the ethanol doses tested elicited significant changes in dialysate met-enkephalin content compared to the saline treatment. CONCLUSIONS: Acute systemic ethanol administration induced a dose- and time-dependent increase in beta-endorphin and dynorphin A1-8 release at the level of the CeA, which may be involved in ethanol consumption.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Etanol/farmacología , Microdiálisis , Péptidos Opioides/efectos de los fármacos , Péptidos Opioides/metabolismo , Amígdala del Cerebelo/química , Animales , Relación Dosis-Respuesta a Droga , Dinorfinas/metabolismo , Encefalina Metionina/metabolismo , Etanol/administración & dosificación , Etanol/sangre , Inyecciones Intraperitoneales , Masculino , Péptidos Opioides/química , Fragmentos de Péptidos/metabolismo , Péptidos/metabolismo , Fotomicrografía , Ratas , Ratas Sprague-Dawley , betaendorfina/metabolismo
19.
Brain Res ; 1197: 85-93, 2008 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-18207137

RESUMEN

The internalization of mu-opioid receptors (MORs) provides an ideal way to locate areas of opioid peptide release. We used this method to study opioid release in the spinal cord evoked by noxious stimuli in anesthetized rats. Previous studies have shown that opioids released in the spinal cord produce MOR internalization only when they are protected from peptidase degradation. Accordingly, rats were implanted with chronic intrathecal catheters that were used to inject a mixture of peptidase inhibitors (amastatin, captopril and phosphoramidon) onto the lumbar spinal cord. Five minutes later, a noxious stimulus was delivered to the paw. Lumbar spinal segments were double-stained with antibodies against MORs and neurokinin 1 receptors (NK1Rs) using immunofluorescence. Mechanical stimulation of the hindpaw consisted of repeated 10 s clamps with a hemostat for 10 min. In the ipsilateral dorsal horn, the stimulus produced abundant NK1R internalization in segments L3-L6, and a more modest but significant MOR internalization in segments L5 and L6. In the contralateral dorsal horn, NK1R was substantially lower and MOR internalization was negligible. The same mechanical stimulus applied to a forepaw did not produce NK1R or MOR internalization in the lumbar spinal cord. Thermal stimulation consisted of immersing a hindpaw in water at 52 degrees C for 2 min. It produced substantial NK1R internalization ipsilaterally in segment L6, but no MOR internalization. These results show that mechanical stimulation induces segmental opioid release, i.e., in the dorsal horn receiving the noxious signals and not in other spinal segments.


Asunto(s)
Péptidos Opioides/metabolismo , Células del Asta Posterior/metabolismo , Inhibidores de Proteasas/administración & dosificación , Receptores Opioides mu/metabolismo , Animales , Técnica del Anticuerpo Fluorescente , Inyecciones Espinales , Masculino , Microscopía Confocal , Péptidos Opioides/efectos de los fármacos , Estimulación Física , Células del Asta Posterior/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de Neuroquinina-1/metabolismo , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo
20.
J Neurosci Res ; 86(5): 1077-86, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18027846

RESUMEN

Nociceptin/orphanin FQ (N/OFQ) is an opioid-related peptide that is markedly up-regulated in sensory neurons in vivo following peripheral inflammation and plays a key role in pain physiology. To identify substances that up-regulate N/OFQ expression in sensory neurons, we carried out an in vitro screen using purified adult mouse dorsal root ganglion (DRG) neurons and identified the potent proinflammatory agent bacterial lipopolysaccharide (LPS) as a very effective inducer of N/OFQ. The robust response of these neurons to LPS enabled us to identify the components of a putative neuronal LPS receptor complex. In contrast to the immune system, where the functional LPS receptor complex is composed of CD-14 together with either MD-2 and TLR4 on myeloid cells or the homologous receptors MD-1 and RP105 on mature B cells, DRG neurons express the unusual combination of CD-14, TLR4, and MD-1. Blocking antibodies against TLR4 and MD-1 prevented induction of N/OFQ by LPS, and, in immunoprecipitation experiments, MD-1 coprecipitated with TLR4. Our findings suggest that LPS regulates N/OFN expression in sensory neurons via a novel combination of LPS receptor components and demonstrate for the first time a direct action of a key initiator of innate immune responses on neurons.


Asunto(s)
Ganglios Espinales/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Neuronas Aferentes/metabolismo , Péptidos Opioides/metabolismo , Dolor/metabolismo , Animales , Anticuerpos/farmacología , Antígenos de Superficie/efectos de los fármacos , Antígenos de Superficie/metabolismo , Línea Celular , Células Cultivadas , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/inmunología , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/metabolismo , Mediadores de Inflamación/farmacología , Receptores de Lipopolisacáridos/efectos de los fármacos , Glicoproteínas de Membrana/efectos de los fármacos , Glicoproteínas de Membrana/metabolismo , Ratones , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/inmunología , Péptidos Opioides/efectos de los fármacos , Dolor/inducido químicamente , Dolor/inmunología , Receptor Toll-Like 4/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología , Nociceptina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...