morphine tolerance
Recently Published Documents


TOTAL DOCUMENTS

715
(FIVE YEARS 99)

H-INDEX

65
(FIVE YEARS 4)

2022 ◽  
Author(s):  
Jianlong Bu ◽  
Wu Xiaohong ◽  
Deng Lin ◽  
Ma Chao ◽  
Shi Xiaoding ◽  
...  

Abstract The pro-inflammatory (M1) and anti-inflammatory (M2) status of microglial determines the outcome of neuroinflammation, which contributes to the pathogenesis of chronic morphine tolerance. Studies report that α2-adrenoceptor agonist dexmedetomidine exerts anti-inflammatory effects in inhibiting morphine tolerance in normal and neuropathic pain animals, which has not been studied in cancer pain. Therefore, we investigate the effect of intrathecal DEX on morphine tolerance in cancer pain, and whether dexmedetomidine functions via modulating microglial activation and M1/M2 polarization. 54 Wistar rats with intrathecal catheterization were treated by morphine for 10 days. Test groups received intrathecal α2-adrenoceptor agonist dexmedetomidine or antagonist MK-467. The mRNA levels of TLR4 and NF-κB were tested by RT-PCR. The protein levels of TLR4, NF-κB, Iba-1, iNOS, CD206 were quantifed using Western blotting, and IL-10 and TNF-α were examined by ELISA. Dexmedetomidine attenuates mechanical threshold and thermal latency, and increased the expression of TLR4 and NF-κB in morphine tolerance of cancer pain. Dexmedetomidine attenuates mechanical and thermal nociception in morphine tolerance in cancer pain rats. Intrathecal DEX pre-treatment significantly increased the protein levels of microglia maker Iba-1, M2 marker CD206 and anti-inflammatory factor IL-10, while had no evident influence on the pro-inflammatory factor TNF-α and M1 marker iNOS in morphine tolerance. Our findings suggest that intrathecal dexmedetomidine attenuates morphine tolerance in cancer pain via α2-adrenoceptor pathway. Furthermore, dexmedetomidine upregulates TLR4/NF-κB pathway and induces microglia activation in chronic morphine tolerance of cancer pain. The anti-inflammatory effect of dexmedetomidine might be exerted by inducing microglia M2 polarization and increasing anti-inflammatory factor IL-10.


2022 ◽  
pp. 108938
Author(s):  
Meiling Deng ◽  
Zengli Zhang ◽  
Manyu Xing ◽  
Xia Liang ◽  
Zhengyiqi Li ◽  
...  

2021 ◽  
Vol 20 (9) ◽  
pp. 1881-1886
Author(s):  
Guihua Liu ◽  
Ying Xiong

Purpose: To determine the influence of miR-140-5p on morphine tolerance in rats.Methods: Sprague-Dawley (SD) rats were randomly divided into morphine tolerance (MT) and saline control (NS) groups, respectively. Rats in MT group were injected with 10 μL (10 μg) morphine twice daily for seven consecutive days while those in NS group were administered the equivalent volume of normal saline. The maximum effect of morphine (MPE) was computed from tail-flick test results. MiR-140-5p mimics and toll-like receptor 4 (TLR4) lentivirus were transfected separately or co-transfected into model rats. MiR-140-5p and TLR4 expression were determined by quantitative real-time polymerase chain reaction (RT-qPCR) or western blotting. Dual-luciferase reporter assay was used to verify the target relationship between miR-140-5p and TLR4.Results: The expression of miR-140-5p was decreased, while the expression of TLR4 increased in morphine-tolerant rats (p < 0.05). TLR4 was a target of miR-140-5p. At 24 and 48 h after injection, MPE clearly increased and TLR4 expression was reduced under miR-140-5p overexpression or TLR4 knockdown (p < 0.05). Moreover, there were no significant changes in MPE or levels of TLR4 when miR-140-5p and TLR4 were co-transfected into morphine-tolerant rats.Conclusion: MiR-140-5p inhibits morphine resistance in rats via targeted regulation of TLR4 expression. These provide a theoretical basis for the clinical management of morphine tolerance.


2021 ◽  
Vol 14 (10) ◽  
pp. 1034
Author(s):  
Lamees Alhassen ◽  
Khawla Nuseir ◽  
Allyssa Ha ◽  
Warren Phan ◽  
Ilias Marmouzi ◽  
...  

The opioid epidemic was triggered by an overprescription of opioid analgesics. In the treatment of chronic pain, repeated opioid administrations are required which ultimately lead to tolerance, physical dependence, and addiction. A possible way to overcome this conundrum consists of a co-medication that maintains the analgesic benefits of opioids while preventing their adverse liabilities. YHS, the extract of the plant Corydalis yanhusuo, has been used as analgesic in traditional Chinese medicine for centuries. More recently, it has been shown to promote analgesia in animal models of acute, inflammatory, and neuropathic pain. It acts, at least in part, by inhibiting the dopamine D2 receptor, suggesting that it may be advantageous to manage addiction. We first show that, in animals, YHS can increase the efficacy of morphine antinociceptive and, as such, decrease the need of the opioid. We then show that YHS, when coadministered with morphine, inhibits morphine tolerance, dependence, and addiction. Finally, we show that, in animals treated for several days with morphine, YHS can reverse morphine dependence and addiction. Together, these data indicate that YHS may be useful as a co-medication in morphine therapies to limit adverse morphine effects. Because YHS is readily available and safe, it may have an immediate positive impact to curb the opioid epidemic.


2021 ◽  
Author(s):  
Mauro Congiu ◽  
Laura Micheli ◽  
Michele Santoni ◽  
Claudia Sagheddu ◽  
Anna Lisa Muntoni ◽  
...  

AbstractOpioids are essential drugs for pain management, although long-term use is accompanied by tolerance, necessitating dose escalation, and dependence. Pharmacological treatments that enhance opioid analgesic effects and/or attenuate the development of tolerance (with a desirable opioid-sparing effect in treating pain) are actively sought. Among them, N-palmitoylethanolamide (PEA), an endogenous lipid neuromodulator with anti-inflammatory and neuroprotective properties, was shown to exert anti-hyperalgesic effects and to delay the emergence of morphine tolerance. A selective augmentation in endogenous PEA levels can be achieved by inhibiting N-acylethanolamine acid amidase (NAAA), one of its primary hydrolyzing enzymes. This study aimed to test the hypothesis that NAAA inhibition, with the novel brain permeable NAAA inhibitor AM11095, modulates morphine’s antinociceptive effects and attenuates the development of morphine tolerance in rats. We tested this hypothesis by measuring the pain threshold to noxious mechanical stimuli and, as a neural correlate, we conducted in vivo electrophysiological recordings from pain-sensitive locus coeruleus (LC) noradrenergic neurons in anesthetized rats. AM11095 dose-dependently (3–30 mg/kg) enhanced the antinociceptive effects of morphine and delayed the development of tolerance to chronic morphine in behaving rats. Consistently, AM11095 enhanced morphine-induced attenuation of the response of LC neurons to foot-shocks and prevented the attenuation of morphine effects following chronic treatment. Behavioral and electrophysiological effects of AM11095 on chronic morphine were paralleled by a decrease in glial activation in the spinal cord, an index of opioid-induced neuroinflammation. NAAA inhibition might represent a potential novel therapeutic approach to increase the analgesic effects of opioids and delay the development of tolerance.


2021 ◽  
Vol 0 (0) ◽  
Author(s):  
Mohammad Rahban ◽  
Samira Danyali ◽  
Jalal Zaringhalam ◽  
Homa Manaheji

Abstract Objectives The most notable adverse side effects of chronic morphine administration include tolerance and hyperalgesia. This study investigated the involvement of dorsal root ganglion (DRG) protein kinase Cɛ (PKCɛ) expression during chronic morphine administration and also considered the relationship between DRG PKCɛ expression and the substance P- neurokinin1 receptor (SP- NK1R) activity. Methods Thirty-six animals were divided into six groups (n=6) in this study. In the morphine and sham groups, rats received 10 µg intrathecal (i.t.) morphine or saline for eight consecutive days, respectively. Behavioral tests were performed on days 1 and 8 before and after the first injections and then 48 h after the last injection (day 10). In the treatment groups, rats received NK1R antagonist (L-732,138, 25 µg) daily, either alone or 10 min before a morphine injection, Sham groups received DMSO alone or 10 min before a morphine injection. Animals were sacrificed on days 8 and 10, and DRG PKCɛ and SP expression were analyzed by western blot and immunohistochemistry techniques, respectively. Results Behavioral tests indicated that tolerance developed following eight days of chronic morphine injection. Hyperalgesia was induced 48 h after the last morphine injection. Expression of SP and PKCɛ in DRG significantly increased in rats that developed morphine tolerance on day 8 and hyperalgesia on day 10, respectively. NK1R antagonist (L-732,138) not only blocked the development of hyperalgesia and the increase of PKCɛ expression but also alleviated morphine tolerance. Conclusions Our results provide evidence that DRG PKCɛ and SP-NK1R most likely participated in the generation of morphine tolerance and hyperalgesia. Pharmacological inhibition of SP-NK1R activity in the spinal cord suggests a role for NK1R and in restricting some side effects of chronic morphine. All experiments were performed by the National Institute of Health (NIH) Guidelines for the Care and Use of Laboratory Animals (NIH Publication No. 80-23, revised1996) and were approved by the Animal Ethics Committee of Shahid Beheshti University of Medical Sciences, Tehran, Iran (IR.SBMU.MSP.REC.1396.130).


PLoS ONE ◽  
2021 ◽  
Vol 16 (9) ◽  
pp. e0256870
Author(s):  
Yaqiong Yang ◽  
Yu Sun ◽  
Rong Hu ◽  
Jia Yan ◽  
Ziheng Wang ◽  
...  

Although they represent the cornerstone of analgesic therapy, opioids, such as morphine, are limited in efficacy by drug tolerance, hyperalgesia and other side effects. Activation of microglia and the consequent production of proinflammatory cytokines play a key pathogenic role in morphine tolerance, but the exact mechanisms are not well understood. This study aimed to investigate the regulatory mechanism of epidermal growth factor receptor (EGFR) on microglial activation induced by morphine in mouse microglial BV-2 cells. In this research, BV-2 cells were stimulated with morphine or pretreated with AG1478 (an inhibitor of EGFR). Expression levels of cluster of differentiation molecule 11b (CD11b), EGFR, and phospho-EGFR were detected by immunofluorescence staining. Cell signaling was assayed by Western blot. The migration ability of BV-2 cells was tested by Transwell assay. The production of interleukin-1beta (IL-1β) and tumor necrosis factor-alpha (TNF-α) in the cell supernatant was determined by ELISA. We observed that the expression of CD11b induced by morphine was increased in a dose- and time- dependent manner in BV-2 cells. Phosphorylation levels of EGFR and ERK1/2, migration of BV-2 cells, and production of IL-1β and TNFα were markedly enhanced by morphine treatment. The activation, migration, and production of proinflammatory cytokines in BV-2 cells were inhibited by blocking the EGFR signaling pathway with AG1478. The present study demonstrated that the EGFR/ERK signaling pathway may represent a novel pharmacological strategy to suppress morphine tolerance through attenuation of microglial activation.


PLoS ONE ◽  
2021 ◽  
Vol 16 (9) ◽  
pp. e0253902
Author(s):  
Ghazaleh Ghamkharinejad ◽  
Seyed Hossein Marashi ◽  
Forough Foolad ◽  
Mohammad Javan ◽  
Yaghoub Fathollahi

Background ɣ-aminobutyric acid (GABA) facilitator valproic acid may be able to curb memory disruption induced by morphine exposure. Objective The effects of the GABA facilitator valproic acid on the behavioral tolerance induced by morphine were investigated. Then hippocampal-dependent tasks named spatial-working and short-term memory procedures using the Y-maze apparatus were examined in morphine tolerant rats. Finally, the changes in the expression of hippocampal GABA-A receptors underlying morphine tolerance were also examined. Methods Rats were treated with daily morphine injections, with or without distinct contextual pairing. To examine the effect of valproic acid on morphine tolerance expression, valproic acid was pretreated an hour before morphine. Spatial-working and short-term memory procedures using the Y-maze apparatus were examined in morphine tolerant rats. Afterwards the changes in the expression of hippocampal GABAα receptors using the quantitative real-time PCR and western blot techniques to detect GABArα subunits mRNAs and protein level were studied. Results Our results showed that both learned and non-associative morphine tolerance influence short-term memory and the subjacent expression of GABArα mRNAs and protein level. Despite its attenuating effects on the development and expression of both learned and non-associative morphine tolerance, only associative morphine tolerance-induced memory dysfunction was ameliorated by valproic acid pretreatment. We also found that the expression of GABArα1, α2, α5 subunits mRNAs and GABAα protein level were affected heavier in associative morphine tolerant rats. Conclusion Our data supports the hypothesis that unconditioned and learned morphine tolerance influences short-term memory and the expression of GABArα 1, α2, α5 mRNAs and GABArα protein level differently, and adds to our understanding of the behavioral and molecular aspects of the learned tolerance to morphine effects.


2021 ◽  
Vol 171 (5) ◽  
pp. 623-626
Author(s):  
N. S. Sorokina ◽  
M. V. Starostina
Keyword(s):  

Sign in / Sign up

Export Citation Format

Share Document