Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Br J Pharmacol ; 180(7): 843-861, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-34986502

RESUMEN

BACKGROUND AND PURPOSE: In rodents, morphine antinociception is influenced by sex. However, conflicting results have been reported regarding the interaction between sex and morphine antinociceptive tolerance. Morphine is metabolised in the liver and brain into morphine-3-glucuronide (M3G). Sex differences in morphine metabolism and differential metabolic adaptations during tolerance development might contribute to behavioural discrepancies. This article investigates the differences in peripheral and central morphine metabolism after acute and chronic morphine treatment in male and female mice. EXPERIMENTAL APPROACH: Sex differences in morphine antinociception and tolerance were assessed using the tail-immersion test. After acute and chronic morphine treatment, morphine and M3G metabolic kinetics in the blood were evaluated using LC-MS/MS. They were also quantified in several CNS regions. Finally, the blood-brain barrier (BBB) permeability of M3G was assessed in male and female mice. KEY RESULTS: This study demonstrated that female mice showed weaker morphine antinociception and faster induction of tolerance than males. Additionally, female mice showed higher levels of M3G in the blood and in several pain-related CNS regions than male mice, whereas lower levels of morphine were observed in these regions. M3G brain/blood ratios after injection of M3G indicated no sex differences in M3G BBB permeability, and these ratios were lower than those obtained after injection of morphine. CONCLUSION: These differences are attributable mainly to morphine central metabolism, which differed between males and females in pain-related CNS regions, consistent with weaker morphine antinociceptive effects in females. However, the role of morphine metabolism in antinociceptive tolerance seemed limited. LINKED ARTICLES: This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc.


Asunto(s)
Morfina , Espectrometría de Masas en Tándem , Ratones , Masculino , Femenino , Animales , Cromatografía Liquida , Derivados de la Morfina/farmacología , Derivados de la Morfina/uso terapéutico , Analgésicos Opioides/farmacología , Dolor/tratamiento farmacológico
2.
Eur J Med Chem ; 258: 115608, 2023 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-37437352

RESUMEN

The compelling demand of a consummate analgesic medication without addiction is rising due to the clinical mistreatment. Additionally, the series of severe untoward effects usually deterred the utilization while coping with serious pain. As a possible turning point, we revealed that compound 14 is a dual agonist of mu opioid receptor (MOR) and nociceptin-orphanin FQ opioid peptide (NOP) receptor in this study. More importantly, compound 14 achieves pain relieving at very small doses, meanwhile, reduces several unwanted side effects such as constipation, reward, tolerance and withdrawal effects. Here, we evaluated the antinociception and side effects of this novel compound from wild type and humanized mice to further develop a safer prescription analgesic drug.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Receptores Opioides mu , Ratones , Animales , Receptores Opioides mu/agonistas , Receptores Opioides/agonistas , Receptor de Nociceptina , Péptidos Opioides/farmacología , Péptidos Opioides/uso terapéutico , Analgésicos Opioides/efectos adversos , Dolor/inducido químicamente , Dolor/tratamiento farmacológico , Analgésicos/efectos adversos , Nociceptina
3.
Antioxidants (Basel) ; 12(7)2023 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-37508016

RESUMEN

Morphine (MOR) is a commonly prescribed drug for the treatment of moderate to severe diabetic neuropathic pain (DNP). However, long-term MOR treatment is limited by morphine analgesic tolerance (MAT). The activation of microglial cells and the release of glia-derived proinflammatory cytokines are known to play an important role in the development of MAT. In this study, we aimed to investigate the effects of the dipeptidyl peptidase-4 inhibitor (DPP-4i) teneligliptin (TEN) on MOR-induced microglial cell activation and MAT in DNP rats. DNP was induced in four groups of male Wistar rats through a single intraperitoneal injection of streptozotocin (STZ) (50 mg/kg, freshly dissolved in 5 mmol/L citrate buffer, pH 4.5). Sham rats were administered with the vehicle. Seven days after STZ injection, all rats were implanted with an intrathecal (i.t) catheter connected to a mini-osmotic pump, divided into five groups, and infused with the following combinations: sham + saline (1 µL/h, i.t), DNP + saline (1 µL/h, i.t), DNP + MOR (15 µg/h, i.t), DNP + TEN (2 µg/h, i.t), and DNP + MOR (15 µg/h, i.t) + TEN (2 µg/h, i.t) for 7 days at a rate of 1 µL/h. The MAT was confirmed through the measurement of mechanical paw withdrawal threshold and tail-flick tests. The mRNA expression of neuroprotective proteins nuclear factor erythroid 2-related factor (Nrf2) and heme oxygenase-1 (HO-1) in the dorsal horn was evaluated by quantitative PCR (qPCR). Microglial cell activation and mononucleate cell infiltration in the spinal cord dorsal horn were assessed by immunofluorescence assay (IFA) and Western blotting (WB). The results showed that co-infusion of TEN with MOR significantly attenuated MAT in DNP rats through the restoration of neuroprotective proteins Nrf2 and HO-1 and suppression of microglial cell activation in the dorsal horn. Though TEN at a dose of 2 µg has mild antinociceptive effects, it is highly effective in limiting MAT.

4.
Front Pharmacol ; 13: 803331, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35529434

RESUMEN

Acutely, non-selective cannabinoid (CB) agonists have been shown to increase morphine antinociceptive effects, and we and others have also demonstrated that non-selective CB agonists attenuate morphine antinociceptive tolerance. Activation of cannabinoid CB2 receptors reverses allodynia and hyperalgesia in models of chronic pain, and co-administration of morphine with CB2 receptor selective agonists has been shown to be synergistic. CB2 receptor activation has also been shown to reduce morphine-induced hyperalgesia in rodents, an effect attributed to CB2 receptor modulation of inflammation. In the present set of experiments, we tested both the acute and chronic interactions between morphine and the CB2 receptor selective agonist O-1966 treatments on antinociception and antinociceptive tolerance in C57Bl6 mice. Co-administration of morphine and O-1966 was tested under three dosing regimens: simultaneous administration, morphine pre-treated with O-1966, and O-1966 pre-treated with morphine. The effects of O-1966 on mu-opioid receptor binding were determined using [3H]DAMGO and [35S]GTPγS binding assays, and these interactions were further examined by FRET analysis linked to flow cytometry. Results yielded surprising evidence of interactions between the CB2 receptor selective agonist O-1966 and morphine that were dependent upon the order of administration. When O-1966 was administered prior to or simultaneous with morphine, morphine antinociception was attenuated and antinociceptive tolerance was exacerbated. When O-1966 was administered following morphine, morphine antinociception was not affected and antinociceptive tolerance was attenuated. The [35S]GTPγS results suggest that O-1966 interrupts functional activity of morphine at the mu-opioid receptor, leading to decreased potency of morphine to produce acute thermal antinociceptive effects and potentiation of morphine antinociceptive tolerance. However, O-1966 administered after morphine blocked morphine hyperalgesia and led to an attenuation of morphine tolerance, perhaps due to well-documented anti-inflammatory effects of CB2 receptor agonism.

5.
Eur J Med Chem ; 243: 114728, 2022 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-36084534

RESUMEN

Currently, there is a significant unmet need for novel analgesics with fewer side effects. In this study, we carried out structural modification of a hit compound previously identified in an artificial-intelligence (AI) virtual screening and discovered the potent analgesic, benzo[b]thiophene-2-carboxamide analog (compound 25) with new structural scaffold. We investigated the signaling pathways of opioid receptors mediated by compound 25, and found this racemic compound activated mu-opioid receptor through the cyclic adenosine monophosphate (cAMP) and ß-arrestin-2-mediated pathways with strong potency and efficacy, and accompanying nociceptin-orphanin FQ opioid peptide and delta-opioid receptors through the cAMP pathway with weak potencies. Compound 25 elicited potent antinociception in thermal-stimulated pain (ED50 value of 127.1 ± 34.65 µg/kg) and inflammatory-induced allodynia models with less gastrointestinal transit inhibition and antinociceptive tolerance than morphine. Overall, this study revealed a novel analgesic with reduced risks of side effects.


Asunto(s)
Analgésicos Opioides , Tiofenos , Humanos , Tiofenos/farmacología , Tiofenos/uso terapéutico , Analgésicos Opioides/efectos adversos , Receptores Opioides mu/agonistas , Receptores Opioides/agonistas , Péptidos Opioides , Morfina/farmacología , Analgésicos/farmacología , Analgésicos/uso terapéutico , Analgésicos/química , Estreñimiento/inducido químicamente , Estreñimiento/tratamiento farmacológico
6.
Neuropharmacology ; 219: 109221, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36084794

RESUMEN

The present study evaluated the effects of compounds targeting extrasynaptic δ subunit-containing γ-aminobutyric acid type A receptors (δ*-GABAARs) to interrogate the role of tonic inhibition in the development of antinociceptive tolerance caused by repeated morphine administration. We investigated the effect of subchronic or acute treatment with non-steroidal positive allosteric modulators (PAMs) of δ*-GABAARs, such as 2-261, on the morphine-antinociceptive tolerance. Mice were treated twice daily with morphine for 9 days and antinociception was measured using the hot water tail immersion test. Co-treatment with 2-261 and morphine prevented morphine-antinociceptive tolerance and acute administration of 2-261 on day 9 was sufficient to reverse the tolerance. Other compounds with activity at δ*-GABAARs also reversed morphine tolerance, whereas an enaminone that lacked activity at δ*-GABAARs did not. Acute administration of 2-261 did not cause an additive or synergistic antinociceptive effect when combined with an acute submaximal dose of morphine. We then used Cre/LoxP recombination to generate GABAA δ-subunit knockout mice to corroborate the pharmacological results. Observations of male δ-knockout mice demonstrated that the δ*-GABAARs was necessary for 2-261 modulation of both analgesic tolerance and somatic withdrawal symptoms produced by subchronic morphine. While female mice still benefited from the positive effects of 2-261, the δ-subunit was not necessary for these effects, highlighting a distinction of the different pathways that could have implications for some of the sex-related differences seen in human opioid-induced outcomes. Consequently, subtype-specific allosteric modulators of GABAARs may warrant further investigation as pharmacological targets to manage tolerance and withdrawal from opioids.


Asunto(s)
Analgésicos Opioides , Morfina , Analgésicos/farmacología , Analgésicos Opioides/farmacología , Animales , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Receptores de GABA-A , Receptores Opioides delta , Agua , Ácido gamma-Aminobutírico
7.
Pharmaceuticals (Basel) ; 15(7)2022 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-35890089

RESUMEN

Analgesic tolerance is a major problem in the clinic for the maintenance of opioid-induced long-term pain relief. Opioids with mixed activity on multiple opioid receptors promise reduced antinociceptive tolerance in preclinical studies, but these compounds typically show poor bioavailability upon oral, subcutaneous, intraperitoneal, or intravenous administration. We designed UTA1003 as a novel opioid that acts as a mu (MOP) and kappa (KOP) opioid receptor agonist and a partial agonist for delta (DOP) opioid receptor. In the present study, its antinociceptive effects, as well as its effects on antinociceptive tolerance and motor behaviour, were investigated in male rats. Acute antinociception was measured before (basal) and at different time points after subcutaneous injection of UTA1003 or morphine using the tail flick and hot plate assays. Various motor behavioural activities, including horizontal locomotion, rearing, and turning, were automatically measured in an open-field arena. The antinociceptive and behavioural effects of repeated administration of UTA1003 and morphine were determined over eight days. UTA1003 induced mild antinociceptive effects after acute administration but induced no tolerance after repeated treatment. Importantly, UTA1003 co-treatment with morphine prevented antinociceptive tolerance compared to morphine alone. UTA1003 showed less motor suppression than morphine in both acute and sub-chronic treatment regimens, while it did not affect morphine-induced motor suppression or hyper-excitation. Based on these activities, we speculate that UTA1003 crosses the blood-brain barrier after subcutaneous administration and, therefore, could be developed as a lead molecule to avoid opioid-induced antinociceptive tolerance and motor suppression. Further structural modifications to improve its antinociceptive effects, toxicity profile, and ADME parameters are nevertheless required.

8.
Antioxidants (Basel) ; 10(2)2021 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-33572871

RESUMEN

Oxidative stress resulting from reactive oxygen species (ROS) is known to play a key role in numerous neurological disorders, including neuropathic pain. Morphine is one of the commonly used opioids for pain management. However, long-term administration of morphine results in morphine antinociceptive tolerance (MAT) through elevation of ROS and suppression of natural antioxidant defense mechanisms. Recently, mesoporous polydopamine (MPDA) nanoparticles (NPS) have been known to possess strong antioxidant properties. We speculated that morphine delivery through an antioxidant nanocarrier might be a reasonable strategy to alleviate MAT. MPDAs showed a high drug loading efficiency of ∼50%, which was much higher than conventional NPS. Spectral and in vitro studies suggest a superior ROS scavenging ability of NPS. Results from a rat neuropathic pain model demonstrate that MPDA-loaded morphine (MPDA@Mor) is efficient in minimizing MAT with prolonged analgesic effect and suppression of pro-inflammatory cytokines. Additionally, serum levels of liver enzymes and levels of endogenous antioxidants were measured in the liver. Treatment with free morphine resulted in elevated levels of liver enzymes and significantly lowered the activities of endogenous antioxidant enzymes in comparison with the control and MPDA@Mor-treated group. Histopathological examination of the liver revealed that MPDA@Mor can significantly reduce the hepatotoxic effects of morphine. Taken together, our current work will provide an important insight into the development of safe and effective nano-antioxidant platforms for neuropathic pain management.

9.
Rev Recent Clin Trials ; 16(3): 288-293, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33267764

RESUMEN

BACKGROUND: Severe postoperative pain is principally managed by opioids. While effective, opioids do not provide adequate relief in many patients and cause many side effects, including antinociceptive tolerance and opioid-induced hyperalgesia. To evaluate if a combination of intravenous Magnesium, Lidocaine, Ketorolac (MLK cocktail) is a useful rescue therapy through synergistic pharmacological mechanisms for acute pain relief. We present the intravenous combination of magnesium, lidocaine, and ketorolac (MLK cocktail) as a possible rescue for opioid insensitive severe post-operative pain. MATERIALS AND METHODS: The principal settings were the post-operative care unit (PACU) and the surgical ward. We retrospectively analyzed the electronic medical record and anesthesia documents of 14 patients experiencing severe postoperative pain, >7/10 visual-analogue pain score (VAS), despite receiving at least 8 mg of intravenous morphine milligram equivalents (MME) after arrival in the LAC+USC Medical Center PACU between September 2012 and January 2013. The data reviewed included patients' demographics, disease etiology, surgical procedure, opioids received perioperatively, and visual-analogue pain scores before and after each analgesic received, and after the MLK cocktail. The a priori primary outcome and a posteriori secondary outcome of this study are mean visual-analogue pain score and morphine milligram equivalent dose administered per hour, respectively. The main tool evaluated has been VAS score. RESULTS: In patients who failed to respond to opioid analgesics, administration of the MLK cocktail improved the VAS pain scores immediately from 9.4 ± 1.0 to 3.6 ± 3.5. The MLK cocktail also decreased the MME doses/hour in the immediate 12 hours postoperative period from 12.4 ± 5.6 to 1.1 ± 0.9. CONCLUSION: In patients experiencing opioid-resistant severe postoperative pain, the magnesium, lidocaine, and ketorolac combination may be an effective nonopioid rescue therapy. Additionally, magnesium, lidocaine, and ketorolac may be utilized in cases complicated by either antinociceptive tolerance or opioid-induced hyperalgesia and can restore opioid responsiveness.


Asunto(s)
Analgésicos Opioides , Ketorolaco , Antiinflamatorios no Esteroideos/uso terapéutico , Método Doble Ciego , Humanos , Ketorolaco/uso terapéutico , Lidocaína/uso terapéutico , Magnesio/uso terapéutico , Dolor Postoperatorio/tratamiento farmacológico , Estudios Retrospectivos
10.
Neuropharmacology ; 121: 158-166, 2017 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-28450061

RESUMEN

Antinociceptive tolerance after repetitive administration of morphine severely limits its clinical use. Despite increased mechanistic understanding of morphine tolerance, little is known about the influence of dosing regimens in its development. We hypothesized that the starting dose of morphine, dosing frequency and dose increments, influence antinociception and the manifestation of antinociceptive tolerance in rats. Male rats were randomly divided into four groups with different intermittent starting-doses of daily morphine (b.i.d.) followed by different increments of single-dose morphine upon development of antinociceptive tolerance, for 2-3 weeks: 2.5 (b.i.d.)→5 â†’ 10→15 mg/kg/day, 5 (b.i.d.)→10 mg/kg/day, 5 (b.i.d.)→15 mg/kg/day, 10 (b.i.d.)→20 mg/kg/day. Antinociception was assessed daily pre-treatment and at several time-points over 2 h post-administration, using tail-flick and hot-plate assays. Tolerance was defined as significant antinociceptive desensitization and was presented as significant reduction of the maximum and total antinociceptive efficacy upon morphine administration. Rats commenced on 2.5 mg/kg/day (b.i.d.) morphine developed tolerance faster than those started on 5 or 10 mg/kg/day (b.i.d.). Comparatively, higher starting and maintenance doses of morphine produced prolonged antinociception and delayed tolerance. Whereas, lower starting and maintenance doses of morphine produced less total antinociception during the course of treatment and did not delay the onset of tolerance, but require smaller dose-increments to reach antinociception after development of antinociceptive tolerance. These results suggest that morphine starting dose, dosing frequency, increments and timing determine the manifestation of antinociceptive tolerance and extent of antinociception. In addition, our results also highlight the need for generally standardized and validated assay protocols and procedures to compare different studies, as a prerequisite to translate pre-clinical results into the clinic.


Asunto(s)
Analgésicos/farmacología , Morfina/farmacología , Umbral del Dolor/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Interacciones Farmacológicas , Tolerancia a Medicamentos , Masculino , Dimensión del Dolor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
11.
Pharmacol Rep ; 68(2): 472-5, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26922555

RESUMEN

BACKGROUND: Prolonged morphine treatment leads to antinociceptive tolerance. Suppression of spinal astrocytes or d-amino acid oxidase (DAAO), an astroglial enzyme catalyzing oxidation of d-amino acids, has reversed morphine antinociceptive tolerance. Since the astrocyte-DAAO pathway generates hydrogen peroxide, an agonist of the TRPA1 channel expressed spinally on nociceptive nerve terminals and astrocytes, we tested a hypothesis that the spinal TRPA1 contributes to antinociceptive tolerance to prolonged spinal morphine treatment. METHODS: Nociception was assessed using hot-plate test in rats with an intrathecal (it) catheter. Drugs were administered it twice daily from day one to seven in five treatment groups: (i) Saline, (ii) Chembridge-5861528 (a TRPA1 antagonist; 10µg), (iii) morphine (10µg), (iv) Chembridge-5861528 (10µg)+morphine (10µg), (v) DMSO. Antinociceptive action of morphine was assessed at day one and eight. Additionally, mRNA for DAAO and TRPA1 in the spinal cord was determined on day 8. RESULTS: Morphine treatment produced antinociceptive tolerance, which was attenuated by co-administration of Chembridge-5861528 that alone had no effect on hot-plate latencies. In animals treated with morphine only, spinal mRNA for DAAO but not TRPA1 was increased. DAAO increase was prevented by co-administration of Chembridge-5861528. CONCLUSIONS: Antinociceptive morphine tolerance and up-regulation of spinal DAAO were attenuated in morphine-treated animals by blocking the spinal TRPA1. This finding suggests that spinal TRPA1 may contribute, at least partly, to facilitation of morphine antinociceptive tolerance through mechanisms that possibly involve TRPA1-mediated up-regulation of the astroglial DAAO, a generator of hydrogen peroxide, a pronociceptive compound acting also on TRPA1.


Asunto(s)
Analgésicos/farmacología , Tolerancia a Medicamentos/fisiología , Morfina/administración & dosificación , Médula Espinal/metabolismo , Canales Catiónicos TRPC/metabolismo , Analgésicos Opioides/administración & dosificación , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , D-Aminoácido Oxidasa/metabolismo , Masculino , Nocicepción/efectos de los fármacos , Dolor/tratamiento farmacológico , Dolor/metabolismo , Dimensión del Dolor/métodos , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Canal Catiónico TRPA1
12.
Br J Pharmacol ; 172(2): 364-74, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24611685

RESUMEN

UNLABELLED: Opiate analgesics such as morphine are often used for pain therapy. However, antinociceptive tolerance and dependence may develop with long-term use of these drugs. It was found that µ-opioid receptors can interact with δ-opioid receptors, and morphine antinociceptive tolerance can be reduced by blocking δ-opioid receptors. Recent studies have shown that µ- and δ-opioid receptors are co-expressed in a considerable number of small neurons in the dorsal root ganglion. The interaction of µ-opioid receptors with δ-opioid receptors in the nociceptive afferents is facilitated by the stimulus-induced cell-surface expression of δ-opioid receptors, and contributes to morphine tolerance. Further analysis of the molecular, cellular and neural circuit mechanisms that regulate the trafficking and interaction of opioid receptors and related signalling molecules in the pain pathway would help to elucidate the mechanism of opiate analgesia and improve pain therapy. LINKED ARTICLES: This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.


Asunto(s)
Nociceptores/metabolismo , Dolor/metabolismo , Receptores Opioides/metabolismo , Analgésicos Opioides/uso terapéutico , Animales , Tolerancia a Medicamentos , Proteínas de Unión al GTP/metabolismo , Ganglios Espinales/metabolismo , Humanos , Morfina/uso terapéutico , Neuronas Aferentes/metabolismo , Dolor/tratamiento farmacológico , Transporte de Proteínas
13.
Neuroscience ; 275: 384-94, 2014 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-24973657

RESUMEN

Opioids are the most widely used analgesics in the treatment of severe acute and chronic pain. However, opioids have many adverse side effects, including the development of antinociceptive tolerance after long-term use. The antinociceptive tolerance of opioids has limited their clinical use. A recent study has reported that autophagy is responsible for morphine-induced neuronal injury. However, little is known about the role of autophagy in morphine antinociceptive tolerance. In the present study, chronic morphine administration was found to induce the expression of autophagy-related proteins, including Beclin1 and microtubule-associated protein light chain 3 (LC3)-II, in GABAergic interneurons in the superficial layer (lamina I-II) of the spinal cord. A single intrathecal administration of autophagy inhibitors, 3-methyladenine (3MA) or wortmannin, inhibited the development of antinociceptive tolerance in a dose-dependent manner. Autophagy in the lamina I-II neurons was associated with increased level of cathepsin B (CatB), a lysosomal cysteine protease. The pharmacological blockade or gene deletion of CatB markedly prevented the development of morphine antinociceptive tolerance. Furthermore, the intrathecal administration of 3MA suppressed the upregulation of CatB 5 days after morphine administration. Finally, CatB deficiency inhibited the increased release probability of glutamate in the lamina I neurons after chronic morphine treatment. These observations suggest that the dysfunction of the spinal GABAergic system induced by CatB-dependent excessive autophagy is partly responsible for morphine antinociceptive tolerance following chronic treatment.


Asunto(s)
Analgésicos Opioides/toxicidad , Autofagia/efectos de los fármacos , Tolerancia a Medicamentos/fisiología , Morfina/toxicidad , Células del Asta Posterior/efectos de los fármacos , Animales , Autofagia/fisiología , Catepsina B/metabolismo , Potenciales Postsinápticos Excitadores , Immunoblotting , Inmunohistoquímica , Interneuronas/efectos de los fármacos , Interneuronas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Placa-Clamp , Células del Asta Posterior/patología
14.
Artículo en Zh | WPRIM | ID: wpr-694057

RESUMEN

Objective To investigate the effects of curcumin (Cur) on activation of spinal Toll-like receptor 4 (TLR4) and on the chronic antinociceptive tolerance of morphine.Methods Sixty male Sprague-Dawley rats with successful intrathecal catheterization were randomly divided into four groups (n=15):saline (NS) group;morphine (MOR) group;curcumin (Cur) group and morphine plus curcumin (MOR+Cur) group.A morphine tolerance model of rats was induced by intrathecal ifjection of morphine 15μg,once a day for 7 consecutive days in MOR and MOR+Cur group;100μg curcumin was administered intrathecally once a day for 7 consecutive days in Cur and MOR+Cur group,10μl saline was administered intrathecally once a day for 7 consecutive days in NS group.The effect of curcumin intrathecal catheterization on morphine antinociceptive tolerance was explored by the tail flick latency (TFL) method and mechanical withdrawal threshold (MWT),and then the maximum possible potential effect (MPE) was calculated.The immunofluorescence staining method was applied to detect the effect of curcumin on the activation of lumbar spinal microglia.Real-time PCR and Western blotting were used to evaluate the effect of curcumin on the expression of mRNA and protein of spinal TLR4.Results The %MPE TFL and %MPE MWT increased significantly in MOR+Cur group than in MOR group (P<0.05) after 7 days of intrathecal injection of morphine,intrathecal injection of curcumin and saline failed to induce analgesic effect.There was no significant difference in MPE between Cur group and NS group (P>0.05).The lumbar spinal microglia increased markedly and the expressions of polyclonal antibody IBA-1 and TLR4 were significantly up-regulated in MOR group than in NS group (P<0.05).The lumbar spinal microglia decreased obviously and the expressions of IBA-1 and TLR4 were significantly down-regulated in MOR+Cur group as compared with that in MOR group (P<0.0S),but there was no significant difference in the expressions of IBA-1 and TLR4 between Cur group and NS group (P>0.05).Conclusion Curcumin may attenuate chronic morphine antinociceptive tolerance through inhibiting spinal TLR4 up-regulation.

15.
Chinese Journal of Pathophysiology ; (12): 2163-2167, 2016.
Artículo en Zh | WPRIM | ID: wpr-506652

RESUMEN

AIM:To explore the roles of Akt ( also called protein kinase B ) and active caspase-3 in the leptin-mediated chronic morphine antinociceptive tolerance in rats .METHODS: A model of chronic morphine antinociceptive tolerance was established in the SD rats .The protein levels of spinal Akt and cleaved caspase-3 were tested by Western blotting.The technique of immunohistochemical staining was used to detect the immunoreactivity positive cells of phospho -rylated ( p)-Akt and cleaved caspase-3 in the spinal cord .Double staining of immunohistochemistry was used to examine the cellular location of the p-Akt and cleaved caspase-3 positive cells.RESULTS: The chronic intrathecal injection of morphine (15 μg) for 7 d markedly upregulated the spinal protein levels of p-Akt and cleaved caspase-3 in the rats.Thirty min before injection of morphine , intrathecal injection of leptin antagonist (3μg) for 7 d significantly attenuated the upreg-ulation of the protein levels of p-Akt and cleaved caspase-3 induced by chronic morphine treatment .The p-Akt was exclu-sively observed in the spinal neurons .The cleaved caspase-3 was only localized with the spinal astrocytes .Intrathecal injec-ting the inhibitors of leptin , Akt and caspase-3 ameliorated the chronic antinociceptive tolerance .CONCLUSION: The spinal Akt pathway and active caspase-3 are involved in the leptin-mediated chronic morphine antinociceptive tolerance in rats.

16.
Neurosci Lett ; 483(2): 85-9, 2010 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-20637262

RESUMEN

The role of superoxide and its active byproduct peroxynitrite as mediators of nociceptive signaling is emerging. We have recently reported that nitration and inactivation of spinal mitochondrial superoxide dismutase (MnSOD) provides a critical source of these reactive oxygen and nitrogen species during central sensitization associated with the development of morphine-induced hyperalgesia and antinociceptive tolerance. In this study, we demonstrate that activation of spinal NADPH oxidase is another critical source for superoxide generation. Indeed, the development of morphine-induced hyperalgesia and antinociceptive tolerance was associated with increased activation of NADPH oxidase and superoxide release. Co-administration of morphine with systemic delivery of two structurally unrelated NADPH oxidase inhibitors namely apocynin or diphenyleneiodonium (DPI), blocked NADPH oxidase activation and the development of hyperalgesia and antinociceptive tolerance at doses devoid of behavioral side effects. These results suggest that activation of spinal NADPH oxidase contributes to the development of morphine-induced hyperalgesia and antinociceptive tolerance. The role of spinal NADPH oxidase was confirmed by showing that intrathecal delivery of apocynin blocked these events. Our results are the first to implicate the contribution of NADPH oxidase as an enzymatic source of superoxide and thus peroxynitrite in the development of central sensitization associated with morphine-induced hyperalgesia and antinociceptive tolerance. These results continue to support the critical role of these reactive oxygen and nitrogen species in pain while advancing our knowledge of their biomolecular sources.


Asunto(s)
Tolerancia a Medicamentos/fisiología , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Morfina/toxicidad , NADPH Oxidasas/química , NADPH Oxidasas/fisiología , Médula Espinal/metabolismo , Superóxidos/química , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacología , Animales , Hiperalgesia/enzimología , Masculino , Ratones , Morfina/metabolismo , NADPH Oxidasas/antagonistas & inhibidores , Médula Espinal/efectos de los fármacos , Médula Espinal/enzimología , Superóxidos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA