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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Br J Cancer ; 127(8): 1565-1574, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35945243

RESUMEN

BACKGROUND: It has been considered that activation of peripheral µ-opioid receptors (MORs) induces side effects of opioids. In this study, we investigated the possible improvement of the immune system in tumour-bearing mice by systemic administration of the peripheral MOR antagonist naldemedine. METHODS: The inhibitory effect of naldemedine on MOR-mediated signalling was tested by cAMP inhibition and ß-arrestin recruitment assays using cultured cells. We assessed possible changes in tumour progression and the number of splenic lymphocytes in tumour-bearing mice under the repeated oral administration of naldemedine. RESULTS: Treatment with naldemedine produced a dose-dependent inhibition of both the decrease in the cAMP level and the increase in ß-arrestin recruitment induced by the MOR agonists. Repeated treatment with naldemedine at a dose that reversed the morphine-induced inhibition of gastrointestinal transport, but not antinociception, significantly decreased tumour volume and prolonged survival in tumour-transplanted mice. Naldemedine administration significantly decreased the increased expression of immune checkpoint-related genes and recovered the decreased level of toll-like receptor 4 in splenic lymphocytes in tumour-bearing mice. CONCLUSIONS: The blockade of peripheral MOR may induce an anti-tumour effect through the recovery of T-cell exhaustion and promotion of the tumour-killing system.


Asunto(s)
Neoplasias , Receptores Opioides mu , Analgésicos Opioides/efectos adversos , Animales , Sistema Inmunológico/metabolismo , Ratones , Derivados de la Morfina , Naltrexona/análogos & derivados , Neoplasias/inducido químicamente , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Receptor Toll-Like 4/metabolismo , beta-Arrestinas/metabolismo
2.
J Pharmacol Sci ; 149(3): 85-92, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35641032

RESUMEN

Neuropathic pain is refractory to opioid analgesics. Since there are functional linkages between µ-opioid receptors (MOR) and cannabinoid receptors (CBR), the present study was designed to investigate the interactions between MOR and CB1R based on antinociceptive effects for neuropathic pain mediated through G protein-coupled inwardly-rectifying potassium channels (GIRKs). The antinociceptive effects against pseudonociceptive response or neuropathic pain of MOR and CBR agonists were assessed in mice with or without partial sciatic nerve ligation. To investigate the functional interaction between MOR and CB1R, electrophysiological recording through GIRK was performed using the two-electrode voltage-clamp method in oocytes along with Western blotting in the spinal cord of mice. Co-administration of the MOR agonist DAMGO and the CB1R agonist CP55,940 augmented inwardly rectifying K+ currents in Xenopus oocytes co-expressing MOR, CB1R and GIRK1/2. Further, combination of morphine and the CBR agonist WIN-55,212-2 produced prominent antinociceptive effects in an i.t. GIRK1 inhibitor-reversible manner. Furthermore, CB1R was upregulated under neuropathic pain in the spinal cord, and such upregulation and antinociceptive effects were not altered by repeated treatment with morphine plus WIN-55,212-2. Our findings suggest that co-administration of MOR and CBR agonists could enhance their antinociceptive effects through GIRK1 in the spinal cord of mice.


Asunto(s)
Canales de Potasio Rectificados Internamente Asociados a la Proteína G , Neuralgia , Receptores de Cannabinoides , Receptores Opioides mu , Animales , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Ratones , Morfina/farmacología , Neuralgia/tratamiento farmacológico , Receptores de Cannabinoides/metabolismo , Receptores Opioides mu/metabolismo
3.
Biochem Biophys Res Commun ; 534: 624-631, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33220930

RESUMEN

In the present study, we demonstrated that there is a direct relationship between scratching behaviors induced by itch and functional changes in the brain reward system. Using a conditional place preference test, the rewarding effect was clearly evoked by scratching under both acute and chronic itch stimuli. The induction of ΔFosB, a member of the Fos family of transcription factors, was observed in dopamine transporter (DAT)-positive dopamine neurons in the ventral tegmental area (VTA) of mice suffering from a chronic itch sensation. Based on a cellular analysis of scratching-activated neurons, these neurons highly expressed tyrosine hydroxylase (TH) and DAT genes in the VTA. Furthermore, in an in vivo microdialysis study, the levels of extracellular dopamine in the nucleus accumbens (NAcc) were significantly increased by transient scratching behaviors. To specifically suppress the mesolimbic dopaminergic pathway using pharmacogenetics, we used the TH-cre/hM4Di mice. Pharmacogenetic suppression of mesolimbic dopaminergic neurons significantly decreased scratching behaviors. Under the itch condition with scratching behaviors restricted by an Elizabethan collar, the induction of ΔFosB was found mostly in corticotropin-releasing hormone (CRH)-containing neurons of the hypothalamic paraventricular nucleus (PVN). These findings suggest that repetitive abnormal scratching behaviors under acute and chronic itch stimuli may activate mesolimbic dopamine neurons along with pleasant emotions, while the restriction of such scratching behaviors may initially induce the activation of PVN-CRH neurons associated with stress.


Asunto(s)
Prurito/fisiopatología , Prurito/psicología , Recompensa , Área Tegmental Ventral/fisiopatología , Enfermedad Aguda , Animales , Conducta Animal/fisiología , Enfermedad Crónica , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Neuronas Dopaminérgicas/metabolismo , Expresión Génica , Histamina/administración & dosificación , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Núcleo Accumbens/fisiopatología , Pruebas de Farmacogenómica , Cloruro de Picrilo/administración & dosificación , Prurito/genética , Tirosina 3-Monooxigenasa/genética
4.
Biochem Biophys Res Commun ; 534: 988-994, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33139013

RESUMEN

TRV130 (oliceridine), a G protein-biased ligand for µ-opioid receptor, has recently been synthesized. It is considered to have strong antinociceptive effects and only minor adverse effects. However, whether or not oliceridine actually exhibits an ideal pharmacological profile as an analgesic has not yet been fully clarified in animal studies. This study examined the pharmacological profile of oliceridine in cells and animals. Oliceridine (10 µM) did not produce any µ-opioid receptor internalization in cells even though it increased impedance, which reflects the activation of Gi protein using the CellKey™ system, and inhibited the formation of cAMP. In mice, oliceridine (0.3-10 mg/kg) produced a dose-dependent antinociceptive effect with a rapid-onset and short-duration action in the hot-plate test, as well as antihyperalgesia after sciatic nerve ligation without the development of antinociceptive tolerance using the thermal hyperalgesia test. On the other hand, oliceridine inhibited gastrointestinal transit. Furthermore, oliceridine produced rapid-onset hyperlocomotion at antinociceptive doses; sensitization developed in mice and an emetic effect was observed in ferrets. These results indicate that, although oliceridine may produce dopamine-related behaviors even through selective stimulation of the G-protein-biased µ-opioid receptor pathway, it still offers advantages for breakthrough pain without antinociceptive tolerance with adequate doses.


Asunto(s)
Analgésicos/uso terapéutico , Proteínas de Unión al GTP/metabolismo , Neuralgia/tratamiento farmacológico , Receptores Opioides mu/metabolismo , Compuestos de Espiro/uso terapéutico , Tiofenos/uso terapéutico , Analgésicos/farmacología , Animales , Línea Celular , Humanos , Masculino , Ratones , Ratones Endogámicos ICR , Neuralgia/metabolismo , Receptores Opioides mu/agonistas , Transducción de Señal/efectos de los fármacos , Compuestos de Espiro/farmacología , Tiofenos/farmacología , Factores de Tiempo
5.
Addict Biol ; 26(4): e13000, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33372347

RESUMEN

Polydrug abuse is common among drug abusers. In particular, psychostimulants are often taken with ethanol, and the combination of 3,4-methylenedioxymethamphetamine (MDMA) and alcohol is one of the most common forms of polydrug abuse. However, the mechanism by which these drugs influence behavior remains unclear. The present study was designed to delineate the mechanisms that underlie the effects of the interaction between MDMA and ethanol on behavior in rodents. The combination of MDMA with ethanol enhanced their locomotor-increasing, rewarding, and discriminative stimulus effects without enhancing their effects on the release of dopamine from the nucleus accumbens in rodents. In addition, ethanol potently enhanced locomotor activity produced by the dopamine receptor agonist apomorphine in mice. In antagonism tests, the dopamine D1 -receptor antagonist SCH23390, but not the D2 -receptor antagonist haloperidol, completely suppressed hyperlocomotion induced by MDMA. However, hyperlocomotion induced by the co-administration of MDMA and ethanol was potently suppressed by haloperidol. These results suggest that the synergistic effects of MDMA and ethanol are mediated through dopamine transmission, especially through postsynaptical regulation of D2 -receptor-mediated functions.


Asunto(s)
Etanol/farmacología , N-Metil-3,4-metilenodioxianfetamina/farmacología , Receptores de Dopamina D2/efectos de los fármacos , Animales , Benzazepinas/farmacología , Antagonistas de Dopamina/farmacología , Haloperidol/farmacología , Masculino , Ratones , Actividad Motora/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Ratas , Receptores de Dopamina D1
6.
J Appl Toxicol ; 39(2): 221-230, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30175511

RESUMEN

Methylmercury (MeHg) is known to cause neurobehavioral impairment in human and experimental animals. We previously reported that MeHg (5 mg Hg/kg) induced severe neurobehavioral dysfunction in 4-week-old KK-Ay mice, although it is difficult to evaluate quantitatively the neurobehavioral impairment in MeHg-treated KK-Ay mice because of their obesity. The aim of this study was to evaluate MeHg-induced neurobehavioral dysfunction in KK-Ay mice using the dynamic weight-bearing test, which analyzes the animal's weight distribution between the four limbs. Male 12-week-old KK-Ay mice were treated with MeHg (5 mg Hg/kg) three times per week for 5 weeks. Body weight loss began after approximately 2 weeks of MeHg treatment, and decreased significantly at 4 weeks. Seven of the nine MeHg-treated mice exhibited overt neurological symptoms such as ataxia and gait disturbance. The weight-bearing load was lower for the forelimb than for the hindlimb at baseline and until 1 week after MeHg treatment was initiated. In weeks 2-4, the dynamic weight-bearing loads on the forelimb and hindlimb were similar. The load on the forelimb exceeded the load on the hindlimb after 5 weeks of treatment. This finding indicates that the dynamic weight-bearing test is useful for semi-quantitative evaluation of neurobehavioral impairment in MeHg-treated rodents, and is less stressful for the animals. Infiltration of CD204-positive macrophages was observed in the sciatic nerve of MeHg-treated mice, suggesting that CD204 can serve as a useful marker of tissue injury in peripheral nerves and a possible target in regenerating peripheral nerves and controlling neuropathies.


Asunto(s)
Conducta Animal/efectos de los fármacos , Intoxicación del Sistema Nervioso por Mercurio/fisiopatología , Compuestos de Metilmercurio/toxicidad , Actividad Motora/efectos de los fármacos , Soporte de Peso/fisiología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Masculino , Intoxicación del Sistema Nervioso por Mercurio/sangre , Intoxicación del Sistema Nervioso por Mercurio/orina , Compuestos de Metilmercurio/sangre , Compuestos de Metilmercurio/orina , Ratones , Ratones Endogámicos , Nervio Ciático/efectos de los fármacos , Nervio Ciático/metabolismo
7.
Mol Pain ; 13: 1744806917740030, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29056067

RESUMEN

Background: µ-Opioid receptor internalization is considered to be critically linked to antinociceptive tolerance. Although µ-opioid receptor agonists have been administered simultaneously with other drugs to control pain, little information is available regarding opioid­opioid interactions. Therefore, the present study was designed to further investigate the utility of a new G protein-biased ligand for µ-opioid receptors, TRV130, which has an antinociceptive effect without ß-arrestin-dependent µ-opioid receptor internalization, and its combination with fentanyl using µ-opioid receptor-expressing cells and mice. Results: In the present study, we confirmed that fentanyl produced a profound increase in ß-arrestin-2 recruitment accompanied by µ-opioid receptor internalization, whereas TRV130 did not induce either the recruitment of ß-arrestin-2 or µ-opioid receptor internalization in µ-opioid receptor-expressing cells. Under these conditions, ß-arrestin-2 recruitment accompanied by µ-opioid receptor internalization induced by fentanyl was abolished by TRV130, whereas TRV130 did not alter the reduction of cyclic adenosine monophosphate formation by fentanyl in µ-opioid receptor-expressing cells. In a behavioral assay, TRV130 exerted an antinociceptive effect in a hot-plate test in mice. In a combination test, the antinociceptive effect of TRV130 was synergistically increased by fentanyl. Fentanyl induced antihyperalgesia and development of its tolerance under a neuropathic pain-like state following sciatic nerve ligation. However, treatment of mice with an antinociceptive dose of TRV130 did not induce the rapid development of tolerance to its antihyperalgesic effect under a neuropathic pain-like state. Furthermore, the rapid development of tolerance to the antihyperalgesic effect induced by fentanyl plus TRV130 in mice with sciatic nerve ligation was not observed, unlike in the case of fentanyl alone. Conclusions: These findings provide evidence that activation of the G protein-biased pathway through µ-opioid receptors can alter signaling in the ß-arrestin-2 pathway linked to the stimulation of µ-opioid receptors. Furthermore, the combination of G protein-biased and ß-arrestin-biased ligands of µ-opioid receptors exerts an ideal antinociceptive effect without the rapid development of antinociceptive tolerance.


Asunto(s)
Tolerancia a Medicamentos/fisiología , Proteínas de Unión al GTP/metabolismo , Receptores Opioides mu/metabolismo , beta-Arrestinas/metabolismo , Analgésicos Opioides/farmacología , Animales , Fentanilo/farmacología , Ligandos , Masculino , Ratones , Morfina/farmacología , Neuralgia/tratamiento farmacológico , Receptores Opioides/metabolismo , Receptores Opioides mu/efectos de los fármacos
8.
Mol Pain ; 122016.
Artículo en Inglés | MEDLINE | ID: mdl-27317580

RESUMEN

BACKGROUND: Methadone is a unique µ-opioid receptor agonist. Although several researchers have insisted that the pharmacological effects of methadone are mediated through the blockade of NMDA receptor, the underlying mechanism by which methadone exerts its distinct pharmacological effects compared to those of other µ-opioid receptor agonists is still controversial. In the present study, we further investigated the pharmacological profile of methadone compared to those of fentanyl and morphine as measured mainly by the discriminative stimulus effect and in vitro assays for NMDA receptor binding, µ-opioid receptor-internalization, and µ-opioid receptor-mediated ß-arrestin recruitment. RESULTS: We found that fentanyl substituted for the discriminative stimulus effects of methadone, whereas a relatively high dose of morphine was required to substitute for the discriminative stimulus effects of methadone in rats. Under these conditions, the non-competitive NMDA receptor antagonist MK-801 did not substitute for the discriminative stimulus effects of methadone. In association with its discriminative stimulus effect, methadone failed to displace the receptor binding of MK801 using mouse brain membrane. Methadone and fentanyl, but not morphine, induced potent µ-opioid receptor internalization accompanied by the strong recruitment of ß-arrestin-2 in µ-opioid receptor-overexpressing cells. CONCLUSIONS: These results suggest that methadone may, at least partly, produce its pharmacological effect as a ß-arrestin-biased µ-opioid receptor agonist, similar to fentanyl, and NMDA receptor blockade is not the main contributor to the pharmacological profile of methadone.


Asunto(s)
Metadona/farmacología , Receptores Opioides mu/agonistas , beta-Arrestinas/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Maleato de Dizocilpina/farmacología , Endocitosis/efectos de los fármacos , Fentanilo/farmacología , Tránsito Gastrointestinal/efectos de los fármacos , Masculino , Ratones Endogámicos ICR , Nocicepción/efectos de los fármacos , Ratas Endogámicas F344 , Receptores de N-Metil-D-Aspartato/metabolismo
9.
Synapse ; 70(8): 317-24, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26990296

RESUMEN

A multiplex analysis for profiling the expression of candidate microRNAs (miRNAs), which are small noncoding RNAs that function as key post-transcriptional regulators, may lead to a better understanding of the complex machinery of neuropathic pain. In the present study, we performed a miRNA array analysis using tissues of the dorsal root ganglion (DRG), a primary site for pain processing, obtained from mice with partial sciatic nerve ligation. Among 1135 total miRNAs, 26 miRNAs showed up-regulation (more than 2-fold change) and only 4 miRNAs showed down-regulation (less than 0.5-fold change) in the DRG of nerve-ligated mice. In a RT-qPCR assay, the levels of miR-21, miR-431, and miR-511-3p were significantly increased on the ipsilateral side of the DRG from 3 to 7 days after sciatic nerve ligation. These elevations were almost absent in IL-6 knockout mice. Furthermore, the expression level of miR-21, but not those of miR-431 or miR511-3p, was significantly increased in exosomes extracted from blood of nerve-ligated mice. These findings suggest that the increased expression of IL-6-regulated miR-21, miR-431, and miR-511-3p in the DRG and increased exosomal miR-21 extracted from blood after sciatic nerve ligation may play at least a partial role in neuropathic pain. Synapse 70:317-324, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Ganglios Espinales/metabolismo , Interleucina-6/metabolismo , MicroARNs/genética , Neuralgia/metabolismo , Animales , Exosomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Neuralgia/genética , Células Receptoras Sensoriales/metabolismo
10.
Synapse ; 69(3): 166-71, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25559075

RESUMEN

Previous studies have shown that sigma-1 receptor chaperone (Sig-1R) ligands can regulate pain-related behaviors, and Sig-1R itself can regulate µ-opioid receptor functions as well as signal transduction. Even though (±)-pentazocine has been used clinically for the treatment of pain through opioid receptors, (+)-pentazocine is known to be a selective Sig-1R agonist. To the best of our knowledge, there is no information available regarding the involvement of Sig-1R agonistic action in the antinociceptive effects of (±)-pentazocine. Therefore, the present study was designed to investigate the effects of (+)-pentazocine on the antinociceptive effects of (-)-pentazocine in mice. Both and (-)-pentazocine induced biphasic antinociceptive effects as measured by the warm-plate test. The early phase, but not the delayed phase, of the antinociceptive effects induced by (-)-pentazocine, which are mediated by the activation of µ-opioid receptors, were suppressed by pretreatment with (+)-pentazocine. These results suggest that the innate antinociceptive action of (±)-pentazocine could be marginally reduced by the effects of (+)-pentazocine, but (+)-pentazocine can suppress the antinociceptive effects of (-)-pentazocine at certain time points.


Asunto(s)
Analgésicos Opioides/farmacología , Nocicepción/efectos de los fármacos , Pentazocina/farmacología , Analgésicos Opioides/química , Animales , Células CHO , Cricetinae , Cricetulus , Isomerismo , Masculino , Ratones , Pentazocina/química
11.
Addict Biol ; 20(4): 724-32, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25065832

RESUMEN

Most opioid receptor agonists have abuse potential, and the rewarding effects of opioids can be reduced in the presence of pain. While each of the enantiomers of pentazocine has a differential pharmacologic profile, (±)-pentazocine has been used clinically for the treatment of pain. However, little information is available regarding which components of pentazocine are associated with its rewarding effects, and whether the (±)-pentazocine-induced rewarding effects can be suppressed under pain. Therefore, the present study was performed to investigate the effects of pain on the acquisition of the rewarding effects of (±)-pentazocine, and to examine the mechanism of the rewarding effects of (±)-pentazocine using the conditioned place preference paradigm. (±)-Pentazocine and (-)-pentazocine, but not (+)-pentazocine, produced significant rewarding effects. Even though the rewarding effects induced by (±)-pentazocine were significantly suppressed under pain induced by formalin, accompanied by increase of preprodynorphin mRNA levels in the nucleus accumbens, a high dose of (±)-pentazocine produced significant rewarding effects under pain. In the normal condition, (±)-pentazocine-induced rewarding effects were blocked by a low dose of naloxone, whereas the rewarding effects induced by high doses of pentazocine under pain were suppressed by naltrindole (a δ-opioid receptor antagonist). Interestingly, (±)-pentazocine did not significantly affect dopamine levels in the nucleus accumbens. These findings suggest that the rewarding effects of (-)-pentazocine may contribute to the abuse potential of (±)-pentazocine through µ- as well as δ-opioid receptors, without robust activation of the mesolimbic dopaminergic system. We also found that neural adaptations can reduce the abuse potential of (±)-pentazocine under pain.


Asunto(s)
Analgésicos Opioides/farmacología , Dolor/fisiopatología , Pentazocina/farmacología , Receptores Opioides delta/fisiología , Receptores Opioides mu/fisiología , Recompensa , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/química , Análisis de Varianza , Animales , Condicionamiento Psicológico/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Isomerismo , Masculino , Naloxona/farmacología , Naltrexona/análogos & derivados , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Núcleo Accumbens/efectos de los fármacos , Pentazocina/administración & dosificación , Pentazocina/química , Ratas , Receptores Opioides delta/efectos de los fármacos , Receptores Opioides mu/efectos de los fármacos
12.
J Pharmacol Exp Ther ; 350(2): 403-11, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24917544

RESUMEN

Previous studies have demonstrated that methylphenidate, MDMA (3,4-methylenedioxymethamphetamine), and other psychostimulants exert stimulant-like subjective effects in humans. Furthermore, MDMA and methylphenidate substitute for the discriminative stimulus effects of psychostimulants, such as amphetamine and cocaine, in animals, which suggests that MDMA and methylphenidate may produce similar discriminative stimulus effects in rats. However, there is no evidence regarding the similarities between the discriminative stimulus effects of MDMA and methylphenidate. To explore this issue, cross-substitution, substitution, and combination tests were conducted in rats that had been trained to discriminate between MDMA (2.5 mg/kg) or methylphenidate (5.0 mg/kg) and saline. In the cross-substitution tests, MDMA and methylphenidate did not cross-substitute for each other. In the substitution test, methamphetamine substituted for the discriminative stimulus effects of methylphenidate, but not for those of MDMA. Furthermore, ephedrine and bupropion, which activate dopaminergic and noradrenergic systems, substituted for the discriminative stimulus effects of methylphenidate. On the other hand, serotonin (5-HT) receptor agonists 5-HT1A and 5-HT2 fully substituted for the discriminative stimulus effects of MDMA. These results suggest that activation of the noradrenergic and dopaminergic systems is important for the discriminative stimulus effects of methylphenidate, whereas activation of the serotonergic system is crucial for the discriminative stimulus effects of MDMA. Even though MDMA, like psychostimulants, exerts stimulant-like effects, our findings clearly indicate that the discriminative stimulus effects of MDMA are distinctly different from those of other psychostimulants in rats.


Asunto(s)
Estimulantes del Sistema Nervioso Central/farmacología , Aprendizaje Discriminativo/efectos de los fármacos , Metilfenidato/farmacología , N-Metil-3,4-metilenodioxianfetamina/farmacología , Animales , Benzazepinas/farmacología , Relación Dosis-Respuesta a Droga , Masculino , Ratas , Ratas Endogámicas F344 , Receptor de Serotonina 5-HT1A/fisiología , Receptores de Serotonina 5-HT2/fisiología , Receptores sigma/fisiología , Receptor Sigma-1
13.
J Pharmacol Sci ; 125(2): 233-6, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24881959

RESUMEN

Genetic factors affect locomotor activity, which mainly depends on the activation of dopaminergic systems. C57BL/6J-bg(J)bg(J) (beige-J) mice, which exhibit deficiencies in immunological function, show behavioral hyperactivity. The present study was designed to investigate the locomotor activity of beige-J mice accompanied by a change in the dopaminergic system. Beige-J mice showed higher locomotor activity and dopamine turnover, whereas splenectomy reduced this hyperlocomotion and dopamine turnover. These results suggest that beige-J mice could be suitable as an experimental animal model for investigating hyperactivation of the dopaminergic system, and the spleen may contribute to the susceptibility of dopaminergic systems to activation.


Asunto(s)
Conducta Animal/fisiología , Dopamina/fisiología , Hipercinesia/genética , Actividad Motora/genética , Animales , Modelos Animales de Enfermedad , Dopamina/metabolismo , Hipercinesia/fisiopatología , Masculino , Ratones Endogámicos C57BL , Actividad Motora/fisiología , Bazo/fisiología , Esplenectomía
14.
J Pharmacol Sci ; 126(3): 264-73, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25346041

RESUMEN

Oxaliplatin is a chemotherapeutic agent that induces chronic refractory neuropathy. To determine whether opioids effectively relieve this chronic neuropathy, we investigated the efficacies of morphine, oxycodone, and fentanyl, and the mechanisms underlying opioid antinociception, in oxaliplatin-induced neuropathy in rats. Rats exhibited significant mechanical allodynia following 2 weeks of chronic oxaliplatin administration. Within the range of doses that did not induce sedation and/or muscle rigidity, morphine (3 mg/kg, subcutaneously, s.c.) and oxycodone (0.3-0.56 mg/kg, s.c.) completely reversed oxaliplatin-induced mechanical allodynia, whereas fentanyl (0.017-0.03 mg/kg, s.c.) showed partial antinociception. The antinociception of the optimal doses of morphine and oxycodone were completely inhibited by pertussis toxin (PTX; 0.5 µg/rat, i.c.v.), a Gi/o protein inhibitor, while the partial effect of fentanyl was not affected in the oxaliplatin model. In the [(35)S]-GTPγS binding assay, activation of µ-opioid receptor by fentanyl, but not by morphine or oxycodone, in the mediodorsal thalamus was significantly reduced in oxaliplatin-treated rats. These results indicate that the lower antinociceptive potency of fentanyl in the oxaliplatin model might in part result from the loss of PTX-sensitive Gi/o protein activation, and the degree of Gi/o protein activation might be related to the potency of antinociception by opioids in this model.


Asunto(s)
Analgésicos Opioides/farmacología , Analgésicos/farmacología , Fentanilo/farmacología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Hiperalgesia/tratamiento farmacológico , Morfina/farmacología , Nocicepción/efectos de los fármacos , Compuestos Organoplatinos , Oxicodona/farmacología , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/antagonistas & inhibidores , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Masculino , Núcleo Talámico Mediodorsal/efectos de los fármacos , Núcleo Talámico Mediodorsal/metabolismo , Oxaliplatino , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/metabolismo , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Toxina del Pertussis/farmacología , Ratas Sprague-Dawley , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
15.
J Pharmacol Sci ; 126(1): 47-55, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25141998

RESUMEN

The rewarding effects of µ-receptor agonists can be suppressed under several pain conditions. We recently showed that clinically used µ-receptor agonists possess efficacies for relieving the neuropathic pain induced by chemotherapeutic drug in rats; however, it is possible that the use of µ-receptor agonists may trigger the rewarding effects even under chemotherapeutic drug-induced neuropathic pain. Nevertheless, no information is available regarding whether µ-receptor agonists produce psychological dependence under chemotherapeutic drug-induced neuropathic pain. Therefore, we examined the effects of neuropathy induced by chemotherapeutic drugs on the rewarding effects of morphine, oxycodone, and fentanyl in rats. Repeated treatment with oxaliplatin or paclitaxel produced neuropathy as measured by the von Frey test. Rewarding effects produced by antinociceptive doses of µ-receptor agonists were not suppressed under oxaliplatin- or paclitaxel-induced neuropathy. Furthermore, the morphine-induced increase in the release of dopamine from the nucleus accumbens, which is a critical step in the rewarding effects of µ-receptor agonists, was not altered in paclitaxel-treated rats. These results suggest that the rewarding effects of µ-receptor agonists can still be established under oxaliplatin- or paclitaxel-induced neuropathic pain. Therefore, patients should be carefully monitored for psychological dependence on µ-receptor agonists when they are used to control chemotherapeutic drug-induced neuropathic pain.


Asunto(s)
Analgésicos Opioides/farmacología , Analgésicos Opioides/uso terapéutico , Antineoplásicos Fitogénicos/efectos adversos , Antineoplásicos/efectos adversos , Fentanilo/farmacología , Fentanilo/uso terapéutico , Morfina/farmacología , Morfina/uso terapéutico , Neuralgia/inducido químicamente , Neuralgia/tratamiento farmacológico , Compuestos Organoplatinos/efectos adversos , Oxicodona/farmacología , Oxicodona/uso terapéutico , Paclitaxel/efectos adversos , Receptores Opioides mu/agonistas , Animales , Fentanilo/efectos adversos , Masculino , Morfina/efectos adversos , Oxaliplatino , Oxicodona/efectos adversos , Ratas Sprague-Dawley , Trastornos Relacionados con Sustancias/etiología
16.
Addict Biol ; 19(3): 362-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-22934790

RESUMEN

Previous findings have shown that sigma-1 receptors (Sig-1Rs) are upregulated by the self-administration of methamphetamine, whereas Sig-1R antisense can attenuate the behavioral effects of psychostimulants in rodents. Sig-1R is an endoplasmic reticulum chaperone protein. However, the effects of Sig-1R agonist on the rewarding effects of abused drugs are not fully understood. Therefore, we examined the effects of selective Sig-1R agonists, such as SA4503 and (+)-pentazocine, on the rewarding effects of abused drugs such as methamphetamine, cocaine and morphine in rats, as measured by the conditioned place preference. Methamphetamine, cocaine and morphine induced a significant place preference. SA4503, but not (+)-pentazocine, significantly attenuated the abused drug-induced place preference. We recently showed that (+)-pentazocine exerts U50,488H-like discriminative stimulus effects, which are related to its psychotomimetic/aversive effects. However, SA4503 did not generalize to the discriminative stimulus effects of U50,488H. These results suggest that SA4503 inhibits the rewarding effects of abused drugs, and that psychotomimetic/aversive effects may not play a role in the attenuating effects of SA4503 on the rewarding effects of abused drugs.


Asunto(s)
Inhibición Psicológica , Nootrópicos/farmacología , Piperazinas/farmacología , Recompensa , Animales , Cocaína/farmacología , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Psicológico/efectos de los fármacos , Discriminación en Psicología/efectos de los fármacos , Inhibidores de Captación de Dopamina/farmacología , Relación Dosis-Respuesta a Droga , Masculino , Metanfetamina/farmacología , Morfina/farmacología , Narcóticos/farmacología , Pentazocina/farmacología , Desempeño Psicomotor/efectos de los fármacos , Ratas Endogámicas F344 , Ratas Sprague-Dawley , Trastornos Relacionados con Sustancias/tratamiento farmacológico
17.
Mol Brain ; 17(1): 20, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38685046

RESUMEN

While the excessive inflammation in cancer cachexia is well-known to be induced by the overproduction of inflammatory mediators in the periphery, microflora disruption and brain dysfunction are also considered to contribute to the induction of cancer cachexia. Hypothalamic microglia play a crucial role in brain inflammation and central-peripheral immune circuits via the production of inflammatory mediators. In the present study, we evaluated possible changes in excessive secretion of gut microbiota-derived endotoxin and the expression timeline of several inflammation-regulatory mediators and their inhibiting modulators in hypothalamic microglia of a mouse model of cancer cachexia following transplantation of pancreatic cancer cells. We demonstrated that the plasma level of lipopolysaccharide (LPS) was significantly increased with an increase in anaerobic bacteria, especially Firmicutes, in the gut at the late stage of tumor-bearing mice that exhibited dramatic appetite loss, sarcopenia and severe peripheral immune suppression. At the early stage, in which tumor-bearing mice had not yet displayed "cachexia symptoms", the mRNA expression of pro-inflammatory cytokines, but not of the neurodegenerative and severe inflammatory modulator lipocalin-2 (LCN2), was significantly increased, whereas at the late "cachexia stage", the level of LCN2 mRNA was significantly increased along with significant decreases in levels of inhibitory immune checkpoint receptors programmed death receptor-1 (PD-1) and CD112R in hypothalamic microglia. In addition, a high density of activated neurons in the paraventricular nucleus (PVN) of the hypothalamus region and a significant increase in corticosterone secretion were found in cachexia model mice. Related to the cachexia state, released corticosterone was clearly increased in normal mice with specific activation of PVN neurons. A marked decrease in the natural killer cell population was also observed in the spleen of mice with robust activation of PVN neurons as well as mice with cancer cachexia. On the other hand, in vivo administration of LPS in normal mice induced hypothalamic microglia with low expression of inhibitory immune checkpoint receptors. These findings suggest that the induction of cancer cachexia may parallel exacerbation of the hypothalamic inflammatory status with polarization to microglia expressed with low levels of inhibitory immune checkpoint receptors following LPS release from the gut microflora.


Asunto(s)
Caquexia , Hipotálamo , Lipocalina 2 , Lipopolisacáridos , Microglía , Animales , Caquexia/complicaciones , Caquexia/patología , Microglía/metabolismo , Hipotálamo/metabolismo , Lipocalina 2/metabolismo , Lipopolisacáridos/farmacología , Masculino , Línea Celular Tumoral , Ratones , Receptor de Muerte Celular Programada 1/metabolismo , Microbioma Gastrointestinal , Citocinas/metabolismo , Neoplasias/complicaciones , Ratones Endogámicos C57BL , Mediadores de Inflamación/metabolismo , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico
18.
J Neurochem ; 125(5): 747-55, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23565710

RESUMEN

Benzodiazepines are commonly used as sedatives, sleeping aids, and anti-anxiety drugs. However, chronic treatment with benzodiazepines is known to induce dependence, which is considered related to neuroplastic changes in the mesolimbic system. This study investigated the involvement of K(+) -Cl(-) co-transporter 2 (KCC2) in the sensitization to morphine-induced hyperlocomotion after chronic treatment with zolpidem [a selective agonist of γ-aminobutyric acid A-type receptor (GABAA R) α1 subunit]. In this study, chronic treatment with zolpidem enhanced morphine-induced hyperlocomotion, which is accompanied by the up-regulation of KCC2 in the limbic forebrain. We also found that chronic treatment with zolpidem induced the down-regulation of protein phosphatase-1 (PP-1) as well as the up-regulation of phosphorylated protein kinase C γ (pPKCγ). Furthermore, PP-1 directly associated with KCC2 and pPKCγ, whereas pPKCγ did not associate with KCC2. On the other hand, pre-treatment with furosemide (a KCC2 inhibitor) suppressed the enhancing effects of zolpidem on morphine-induced hyperlocomotion. These results suggest that the mesolimbic dopaminergic system could be amenable to neuroplastic change through a pPKCγ-PP-1-KCC2 pathway by chronic treatment with zolpidem.


Asunto(s)
Sistema Límbico/efectos de los fármacos , Sistema Límbico/metabolismo , Morfina/administración & dosificación , Agitación Psicomotora/metabolismo , Piridinas/administración & dosificación , Simportadores/fisiología , Animales , Sinergismo Farmacológico , Masculino , Ratones , Ratones Endogámicos ICR , Simportadores/biosíntesis , Resultado del Tratamiento , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Zolpidem , Cotransportadores de K Cl
19.
J Pharmacol Exp Ther ; 347(1): 91-9, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23902939

RESUMEN

Reducing the side effects of pain treatment is one of the most important strategies for improving the quality of life of cancer patients. However, little is known about the mechanisms that underlie these side effects, especially constipation induced by opioid receptor agonists; i.e., do they involve naloxonazine-sensitive versus -insensitive sites or central-versus-peripheral µ-opioid receptors? The present study was designed to investigate the mechanisms of µ-opioid receptor agonist-induced constipation (i.e., the inhibition of gastrointestinal transit and colonic expulsion) that are antagonized by the peripherally restricted opioid receptor antagonist naloxone methiodide and naloxonazine in mice. Naloxonazine attenuated the fentanyl-induced inhibition of gastrointestinal transit more potently than the inhibition induced by morphine or oxycodone. Naloxone methiodide suppressed the oxycodone-induced inhibition of gastrointestinal transit more potently than the inhibition induced by morphine, indicating that µ-opioid receptor agonists induce the inhibition of gastrointestinal transit through different mechanisms. Furthermore, we found that the route of administration (intracerebroventricular, intrathecally, and/or intraperitoneally) of naloxone methiodide differentially influenced the suppressive effect on the inhibition of colorectal transit induced by morphine, oxycodone, and fentanyl. These results suggest that morphine, oxycodone, and fentanyl induce constipation through different mechanisms (naloxonazine-sensitive versus naloxonazine-insensitive sites and central versus peripheral opioid receptors), and these findings may help us to understand the characteristics of the constipation induced by each µ-opioid receptor agonist and improve the quality of life by reducing constipation in patients being treated for pain.


Asunto(s)
Analgésicos Opioides/metabolismo , Analgésicos Opioides/toxicidad , Estreñimiento/metabolismo , Tránsito Gastrointestinal/fisiología , Receptores Opioides mu/fisiología , Animales , Estreñimiento/inducido químicamente , Tránsito Gastrointestinal/efectos de los fármacos , Inyecciones Intraventriculares , Masculino , Ratones , Ratones Endogámicos ICR , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de Narcóticos/metabolismo , Técnicas de Cultivo de Órganos , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo
20.
Behav Pharmacol ; 24(4): 275-81, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23838963

RESUMEN

We have reported previously that the inhibition of both dopaminergic and psychotomimetic/hallucinogenic components plays a role in the discriminative stimulus effects of U-50,488H. However, the mechanisms that underlie the discriminative stimulus effects of U-50,488H, and especially the component that plays a significant role, have not yet been clarified. The present study was designed to further investigate the mechanism(s) of the discriminative stimulus effects of the κ-opioid receptor agonist U-50,488H in rats that had been trained to discriminate between 3.0 mg/kg U-50,488H and saline. The dopamine D2 receptor antagonist sulpiride, but not the D1 receptor antagonist SCH23390, generalized to the discriminative stimulus effects of U-50,488H. The mood-stabilizing agents lithium chloride and valproic acid, which have attenuating effects on the Akt/GSK3 pathway, also partially generalized to the discriminative stimulus effects of U-50,488H. In contrast, the 5-HT-related compound racemic 3,4-methylenedioxymethamphetamine, the cannabinoid receptor agonist WIN55,212-2, and the µ-opioid receptor agonist morphine failed to generalize to the discriminative stimulus effects of U-50,488H. These results suggest that the inhibition of the dopaminergic activity mediated by the postsynaptic D2 receptor, followed by suppression of the Akt/GSK3 pathway may be critical for the induction of the discriminative stimulus effects induced by U-50,488H.


Asunto(s)
3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Analgésicos no Narcóticos/farmacología , Discriminación en Psicología/efectos de los fármacos , Receptores de Dopamina D2/metabolismo , Transducción de Señal/efectos de los fármacos , Inhibidores de Captación Adrenérgica/farmacología , Animales , Antimaníacos/farmacología , Condicionamiento Operante/efectos de los fármacos , Dopaminérgicos/farmacología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Cloruro de Litio/farmacología , Masculino , Morfina/farmacología , N-Metil-3,4-metilenodioxianfetamina/farmacología , Narcóticos/farmacología , Ratas , Ratas Endogámicas F344
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA