Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.596
Filter
1.
Neuropeptides ; 90: 102185, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34419803

ABSTRACT

It has been shown that systemic and local administration of ultra-low dose morphine induced a hyperalgesic response via mu-opioid receptors. However, its exact mechanism(s) has not fully been clarified. It is documented that mu-opioid receptors functionally couple to T-type voltage dependent Ca+2 channels. Here, we investigated the role of T-type calcium channels, amiloride and mibefradil, on the induction of low-dose morphine hyperalgesia in male Wistar rats. The data showed that morphine (0.01 µg i.t. and 1 µg/kg i.p.) could elicit hyperalgesia as assessed by the tail-flick test. Administration of amiloride (5 and 10 µg i.t.) and mibefradil (2.5 and 5 µg i.t.) completely blocked low-dose morphine-induced hyperalgesia in spinal dorsal horn. Amiloride at doses of 1 and 5 mg/kg (i.p.) and mibefradil (9 mg/kg ip) 10 min before morphine (1 µg/kg i.p.) inhibited morphine-induced hyperalgesia. Our results indicate a role for T-type calcium channels in low dose morphine-induced hyperalgesia in rats.


Subject(s)
Analgesics, Opioid/adverse effects , Calcium Channels, T-Type/drug effects , Hyperalgesia/chemically induced , Morphine/adverse effects , Amiloride/pharmacology , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Injections, Intraperitoneal , Male , Mibefradil/pharmacology , Morphine/administration & dosage , Morphine/antagonists & inhibitors , Pain Measurement/drug effects , Pain Threshold/drug effects , Posterior Horn Cells/drug effects , Rats , Rats, Wistar , Receptors, Opioid, mu
2.
Neurosci Lett ; 752: 135825, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33727130

ABSTRACT

OBJECTIVE: To investigate the effect of minocycline on morphine withdrawal symptoms. METHODS: We established a rat model of morphine dependence, then injected the animals with naloxone to induce withdrawal symptoms. Minocycline was injected into the midbrain periaqueductal gray and its effect on withdrawal symptoms and Ca2+-dependent protein kinase (CaMKII), Ras, and phospho-extracellular signal-regulated kinase (p-ERK) expression was observed. RESULTS: Minocycline inhibited withdrawal symptoms such as "wet dog" shakes, teeth chatter, and ptosis, perhaps by inhibiting the activation of microglia and the expression of CaMKII, Ras, and p-ERK. Minocycline had no effect on the behavior of control rats or on CaMKII, Ras, or p-ERK expression. CONCLUSION: Minocycline alleviates morphine withdrawal symptoms by inhibiting the activation of microglia and downregulating the expression of CaMKII, Ras, and p-ERK.


Subject(s)
MAP Kinase Signaling System/drug effects , Minocycline/pharmacology , Morphine/adverse effects , Substance Withdrawal Syndrome/drug therapy , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Humans , Male , Microglia/drug effects , Microglia/pathology , Minocycline/therapeutic use , Morphine/antagonists & inhibitors , Naloxone/administration & dosage , Periaqueductal Gray/drug effects , Periaqueductal Gray/pathology , Rats , Substance Withdrawal Syndrome/pathology , ras Proteins/metabolism
3.
Neuropharmacology ; 185: 108437, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33316279

ABSTRACT

BACKGROUND: Antagonism of peripheral opioid receptors by methylnaltrexone (MNTX) was recently proposed as a potential mechanism to attenuate the development of opioid analgesic tolerance based on experiments conducted in mice. However, reports indicate that MNTX is demethylated to naltrexone (NTX) in mice, and NTX may subsequently cross the blood-brain barrier to antagonize centrally-mediated opioid effects. The goal of this study was to determine whether MNTX alters centrally-mediated behaviors elicited by the opioid analgesics, morphine and oxycodone, and to quantify concentrations of MNTX and NTX in blood and brain following their administration in mice. METHODS: Combinations of MNTX and morphine were tested under acute and chronic conditions in thermal nociceptive assays. Effects of MNTX and NTX pretreatment were assessed in an oxycodone discrimination operant procedure. Blood and brain concentrations of these antagonists were quantified after their administration using liquid chromatography-mass spectrometry. RESULTS: MNTX dose-dependently attenuated acute and chronic morphine antinociception. MNTX and NTX dose-dependently antagonized the discriminative stimulus effects of oxycodone. MNTX and NTX were detected in both blood and brain after administration of MNTX, confirming its demethylation and demonstrating that MNTX itself can cross the blood-brain barrier. CONCLUSIONS: These results provide converging behavioral and analytical evidence that MNTX administration in mice attenuates centrally-mediated effects produced by opioid analgesics and results in functional concentrations of MNTX and NTX in blood and brain. Collectively, these findings indicate that MNTX cannot be administered systemically in mice for making inferences that its effects are peripherally restricted.


Subject(s)
Analgesics, Opioid/pharmacology , Blood-Brain Barrier/drug effects , Morphine/pharmacology , Naltrexone/analogs & derivatives , Narcotic Antagonists/pharmacology , Oxycodone/pharmacology , Analgesics, Opioid/antagonists & inhibitors , Animals , Blood-Brain Barrier/metabolism , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Morphine/antagonists & inhibitors , Naltrexone/metabolism , Naltrexone/pharmacology , Narcotic Antagonists/metabolism , Oxycodone/antagonists & inhibitors , Pain Measurement/drug effects , Pain Measurement/methods , Quaternary Ammonium Compounds/metabolism , Quaternary Ammonium Compounds/pharmacology
4.
Bull Exp Biol Med ; 170(1): 30-34, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33222084

ABSTRACT

Activity of compound GSB-106, a low-molecular mimetic of loop 4 of the brain neurotrophic factor (BDNF), was studied in experimental morphine withdrawal syndrome simulated in outbred rats. Single and subchronic (5 intraperitoneal injections) administration of GSB-106 in a dose of 0.1 mg/kg significantly reduced the total index of morphine withdrawal syndrome by 55.2 and 45.6%, respectively. GSB-106 reduced the severity of some behavioral signs (piloerection, gnashing of teeth, wet-dog shaking, and runaway attempts), but had no effect on mechanical allodynia formed in the rats with dependence. Subchronic treatment with GSB-106 prevented the increase in the content of ΔFosB (product of early response gene) in the striatum induced by morphine withdrawal. The results confirmed the concept on the involvement of neurotrophins, specifically BDNF and its analogs, in the mechanisms associated with the formation of opiate dependence.


Subject(s)
Dipeptides/pharmacology , Morphine Dependence/drug therapy , Morphine/antagonists & inhibitors , Narcotic Antagonists/pharmacology , Peptidomimetics/pharmacology , Substance Withdrawal Syndrome/drug therapy , Animals , Animals, Outbred Strains , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Gene Expression , Hyperalgesia/genetics , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Injections, Intraperitoneal , Male , Morphine/adverse effects , Morphine Dependence/genetics , Morphine Dependence/metabolism , Morphine Dependence/physiopathology , Narcotics/adverse effects , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Rats , Substance Withdrawal Syndrome/genetics , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/physiopathology
5.
Psychopharmacology (Berl) ; 237(10): 3057-3065, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32772146

ABSTRACT

RATIONALE: Opioid abuse remains a serious public health problem. The pseudoirreversible mu opioid receptor antagonist methocinnamox (MCAM) might be useful for treating opioid abuse and overdose. Because endogenous opioid systems can modulate cognition and decision-making, it is important to evaluate whether long-term blockade of mu opioid receptors by MCAM adversely impacts complex operant behavior involving memory. OBJECTIVE: This study tested the effects of MCAM in rhesus monkeys responding under a delayed matching-to-sample task, with correct responses reinforced by sucrose pellets. Because MCAM did not alter performance, antagonism of the rate-decreasing effects of morphine was used to confirm that an effective dose of MCAM was administered. Moreover, the muscarinic receptor antagonist scopolamine and the N-methyl-D-aspartate antagonist phencyclidine were studied as positive controls to demonstrate sensitivity of this procedure to memory disruption. RESULTS: Neither MCAM (0.32 mg/kg) nor morphine (1-5.6 mg/kg) impaired delayed matching-to-sample accuracy. Morphine dose-dependently decreased the number of trials completed before MCAM administration, and a single injection of MCAM blocked the behavioral suppressant effects of morphine for at least 7 days. Scopolamine (0.01-0.056 mg/kg) and phencyclidine (0.1-0.56 mg/kg) dose-dependently decreased delayed matching-to-sample accuracy and the number of trials completed. CONCLUSIONS: MCAM did not impair memory (as measured by accuracy in a delayed matching-to-sample task) and did not decrease responding for or consumption of sucrose pellets. This dose of MCAM attenuates self-administration of opioids and reverses as well as prevents opioid-induced respiratory depression. These results provide further support for a favorable adverse effect profile for MCAM.


Subject(s)
Cinnamates/pharmacology , Memory/drug effects , Morphine Derivatives/pharmacology , Morphine/antagonists & inhibitors , Morphine/pharmacology , Narcotic Antagonists/pharmacology , Receptors, Opioid, mu/antagonists & inhibitors , Animals , Cognition/drug effects , Cognition/physiology , Dose-Response Relationship, Drug , Female , Macaca mulatta , Male , Memory/physiology , Receptors, Opioid, mu/physiology , Reinforcement, Psychology , Self Administration
6.
Neuropharmacology ; 171: 108110, 2020 07.
Article in English | MEDLINE | ID: mdl-32344007

ABSTRACT

MT-45 is a synthetic opioid that was developed in the 1970s as an analgesic compound. However, in recent years MT-45 has been associated with multiple deaths in Europe and has been included in the class of novel psychoactive substances known as novel synthetic opioids (NSOs). Little is known about the pharmaco-toxicological effects of MT-45. Therefore, we used a dynamic mass redistribution (DMR) assay to investigate the pharmacodynamic profile of this NSO in vitro compared with morphine. We then used in vivo studies to investigate the effect of the acute systemic administration of MT-45 (0.01-15 mg/kg i.p.) on motor and sensorimotor (visual, acoustic and tactile) responses, mechanical and thermal analgesia, muscle strength and body temperature in CD-1 male mice. Higher doses of MT-45 (6-30 mg/kg i.p.) were used to investigate cardiorespiratory changes (heart rate, respiratory rate, SpO2 saturation and pulse distention). All effects of MT-45 were compared with those of morphine. In vitro DMR assay results demonstrated that at human recombinant opioid receptors MT-45 behaves as a potent selective mu agonist with a slightly higher efficacy than morphine. In vivo results showed that MT-45 progressively induces tail elevation at the lowest dose tested (0.01 mg/kg), increased mechanical and thermal antinociception (starting from 1 to 6 mg/kg), decreased visual sensorimotor responses (starting from 3 to 6 mg/kg) and reduced tactile responses, modulated motor performance and induced muscle rigidity at higher doses (15 mg/kg). In addition, at higher doses (15-30 mg/kg) MT-45 impaired the cardiorespiratory functions. All effects were prevented by the administration of the opioid receptor antagonist naloxone. These findings reveal the risks associated with the ingestion of opioids and the importance of studying these drugs and undertaking more clinical studies of the current molecules to better understand possible therapeutic interventions in the case of toxicity.


Subject(s)
Analgesics, Opioid/pharmacology , Piperazines/pharmacology , Aggression/drug effects , Analgesics, Opioid/antagonists & inhibitors , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Hemodynamics/drug effects , Hot Temperature , Humans , Mice , Morphine/antagonists & inhibitors , Morphine/pharmacology , Motor Activity/drug effects , Muscle Strength/drug effects , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Pain Measurement/drug effects , Physical Stimulation , Piperazines/antagonists & inhibitors , Receptors, Opioid/drug effects , Respiration/drug effects , Sensation/drug effects
7.
QJM ; 113(12): 859-869, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-32176291

ABSTRACT

BACKGROUND: Our previous study has demonstrated that morphine post-conditioning (MpostC) protects cardiomyocytes from ischemia/reperfusion (I/R) injury partly through activating protein kinase-epsilon (PKCε) signaling pathway and subsequently inhibiting mitochondrial permeability transition pore (mPTP) opening. AIM: In this study, we aim to investigate the relationship between long non-coding RNA TINCR and PKCε in cardiomyocytes under MpostC-treated I/R injury. DESIGN: The myocardial I/R rat model was established by the ligation of lower anterior descending coronary artery for 45 min followed by the reperfusion for 1 h, and MpostC was performed before the reperfusion. METHOD: H/R and MpostC were performed in the rat cardiomyocyte cell line (H9C2), and the Cytochrome-c release in cytosol and mPTP opening were determined. Cell viability was detected by using Cell Counting Kit-8, and cell apoptosis was determined by using flow cytometry or TUNEL assay. RESULTS: The results indicated that MpostC restored the expression of TINCR in I/R rat myocardial tissues. In cardiomyocytes, the therapeutic effect of MpostC, including reduced mPTP opening, reduced Cytochrome-c expression, increased cell viability and reduced cell apoptosis, was dramatically negated by interfering TINCR. The expression of fibroblast growth factor 1 (FGF1), a protein that activates PKCε signaling pathway, was positively correlated with TINCR. The RNA immunoprecipitation and RNA pull-down assay further confirmed the binding between FGF1 and TINCR. Furthermore, TINCR was demonstrated to inhibit the degradation and ubiquitination of FGF1 in cardiomyocytes using the cycloheximide experiment and the ubiquitination assay. The TINCR/FGF1/PKCε axis was revealed to mediate the protective effect of MpostC against hypoxia/reoxygenation injury both in vitro and in vivo. CONCLUSION: In conclusion, our findings demonstrated that MpostC-induced up-regulation of TINCR protects cardiomyocytes from I/R injury via inhibiting degradation and ubiquitination of FGF1, and subsequently activating PKCε signaling pathway, which provides a novel insight in the mechanism of TINCR and PKCε during MpostC treatment of I/R injury.


Subject(s)
Fibroblast Growth Factor 1/genetics , Morphine/pharmacology , Myocardial Reperfusion Injury/prevention & control , Protein Kinase C-epsilon/genetics , RNA, Long Noncoding/genetics , Animals , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Cytochromes c/metabolism , Fibroblast Growth Factor 1/metabolism , Gene Expression Regulation , Ischemic Postconditioning/methods , Male , Morphine/antagonists & inhibitors , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Organ Culture Techniques , Protein Kinase C-epsilon/metabolism , RNA, Long Noncoding/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Ubiquitination , Up-Regulation
8.
Neurosci Lett ; 714: 134502, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31639423

ABSTRACT

The aim of this study was to investigate the relationship of dopamine D1 receptor (D1R) and its downstream factors with morphine withdrawal symptoms in rats. Rats were injected intraperitoneally with morphine in a dose-escalating manner. The midbrain periaqueductal gray (PAG) area was microinjected with D1R antagonist SCH23390 or D1R agonist SKF38393. Rats were intraperitoneally injected with naloxone (4 mg/kg) after the last morphine injection, and the withdrawal response was observed. The D1R antagonist reduced the withdrawal response in morphine-exposed rats and decreased the expression of Ca2+/calmodulin-dependent protein kinase II (CaMKII), phosphorylated extracellular signal-regulated kinase (p-ERK) and cAMP response element-binding protein (CREB) in the PAG. However, the ability of SKF38393 to increase the withdrawal response was weak and limited. Taken together, the results suggest that D1R antagonist decreased the withdrawal response in morphine-exposed rats by downregulating the downstream factors, CaMKII, p-ERK and CREB.


Subject(s)
Benzazepines/pharmacology , Morphine/adverse effects , Periaqueductal Gray/metabolism , Substance Withdrawal Syndrome/prevention & control , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/administration & dosage , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Animals , Benzazepines/administration & dosage , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Down-Regulation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Microinjections , Morphine/antagonists & inhibitors , Naloxone/pharmacology , Rats , Receptors, Dopamine D1/antagonists & inhibitors
9.
Neuroscience ; 422: 54-64, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31689388

ABSTRACT

The pancreatic peptide, Amylin (AMY), reportedly affects nociception in rodents. Here, we investigated the potential effect of AMY on the tolerance to morphine and on the expression of BDNF at both levels of protein and RNA in the lumbar spinal cord of morphine tolerant rats. Animals in both groups of control and test received a single daily dose of intrathecal (i.t.) morphine for 10 days. Rats in the test group received AMY (1, 10 and 60 pmoles) in addition to morphine from days 6 to10. Morphine tolerance was established at day 5. AMY alone showed enduring antinociceptive effects for 10 days. Real-Time PCR, western blotting and ELISA were used respectively to assess levels of BDNF transcripts and their encoded proteins. Rats tolerant to i.t. morphine showed increased expression of exons I, IV, and IX of the BDNF gene, and had elevated levels of pro-BDNF and BDNF protein in their lumbar spinal cord. AMY, when co-administered with morphine from days 6 to 10, reversed morphine tolerance and adversely affected the morphine-induced expression of the BDNF gene at both levels of protein and mRNAs containing exons I, IV and IX. AMY alone increased levels of exons I and IV transcripts. Levels of pro-BDNF and BDNF proteins remained unchanged in the lumbar spinal cord of rats treated by AMY alone. These results suggest that i.t. AMY not only abolished morphine tolerance, but also reduced the morphine induced increase in the expression of both BDNF transcripts and protein in the lumbar spinal cord.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Drug Tolerance/physiology , Islet Amyloid Polypeptide/pharmacology , Morphine/pharmacology , Animals , Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/metabolism , Dose-Response Relationship, Drug , Injections, Spinal , Islet Amyloid Polypeptide/administration & dosage , Male , Morphine/antagonists & inhibitors , Nociception/drug effects , Protein Precursors/metabolism , RNA, Messenger/biosynthesis , Rats , Spinal Cord/metabolism
10.
Drug Alcohol Depend ; 204: 107501, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31479865

ABSTRACT

BACKGROUND: One emerging strategy to address the opioid crisis includes opioid-targeted immunopharmacotherapies. This study compared effectiveness of a heroin-tetanus toxoid (TT) conjugate vaccine to antagonize heroin, 6-acetylmorphine (6-AM), morphine, and fentanyl antinociception in rats. METHODS: Adult male and female Sprague Dawley rats received three doses of active or control vaccine at weeks 0, 2, and 4. Vaccine pharmacological selectivity was assessed by comparing opioid dose-effect curves in 50 °C warm-water tail-withdrawal procedure before and after active or control heroin-TT vaccine. Route of heroin administration [subcutaneous (SC) vs. intravenous [IV)] was also examined as a determinant of vaccine effectiveness. Continuous naltrexone treatment (0.0032-0.032 mg/kg/h) effects on heroin, 6-AM, and morphine antinociceptive potency were also determined as a benchmark for minimal vaccine effectiveness. RESULTS: The heroin-TT vaccine decreased potency of SC heroin (5-fold), IV heroin (3-fold), and IV 6-AM (3-fold) for several weeks without affecting IV morphine or SC and IV fentanyl potency. The control vaccine did not alter potency of any opioid. Naltrexone dose-dependently decreased antinociceptive potency of SC heroin, and treatment with 0.01 mg/kg/h naltrexone produced similar, approximate 8-fold decreases in potencies of SC and IV heroin, IV 6-AM, and IV morphine. The combination of naltrexone and active vaccine was more effective than naltrexone alone to antagonize SC heroin but not IV heroin. CONCLUSIONS: The heroin-TT vaccine formulation examined is less effective, but more selective, than chronic naltrexone to attenuate heroin antinociception in rats. Furthermore, these results provide an empirical framework for future preclinical opioid vaccine research to benchmark effectiveness against naltrexone.


Subject(s)
Fentanyl/antagonists & inhibitors , Heroin/antagonists & inhibitors , Heroin/pharmacology , Morphine Derivatives/antagonists & inhibitors , Morphine/antagonists & inhibitors , Naltrexone/pharmacology , Vaccination , Analgesics, Opioid/antagonists & inhibitors , Analgesics, Opioid/pharmacology , Animals , Dose-Response Relationship, Drug , Drug Administration Routes , Female , Fentanyl/pharmacology , Male , Morphine/pharmacology , Morphine Derivatives/pharmacology , Narcotic Antagonists/pharmacology , Rats , Tetanus Toxoid/pharmacology
11.
Br J Pharmacol ; 176(23): 4434-4445, 2019 12.
Article in English | MEDLINE | ID: mdl-31347704

ABSTRACT

BACKGROUND AND PURPOSE: The concept of opioid ligands biased towards the G protein pathway with minimal recruitment of ß-arrestin-2 is a promising approach for the development of novel, efficient, and potentially nonaddictive opioid therapeutics. A recently discovered biased µ-opioid receptor agonist, PZM21, showed analgesic effects with reduced side effects. Here, we aimed to further investigate the behavioural and biochemical properties of PZM21. EXPERIMENT APPROACH: We evaluated antinociceptive effects of systemic and intrathecal PZM21 administration. Its addiction-like properties were determined using several behavioural approaches: conditioned place preference, locomotor sensitization, precipitated withdrawal, and self-administration. Also, effects of PZM21 on morphine-induced antinociception, tolerance, and reward were assessed. Effects of PZM21 on striatal release of monoamines were evaluated using brain microdialysis. KEY RESULTS: PZM21 caused long-lasting dose-dependent antinociception. It did not induce reward- and reinforcement-related behaviour; however, its repeated administration led to antinociceptive tolerance and naloxone-precipitated withdrawal symptoms. Pretreatment with PZM21 enhanced morphine-induced antinociception and attenuated the expression of morphine reward. In comparison to morphine, PZM21 administration induced a moderate release of dopamine and a robust release of 5-HT in the striatum. CONCLUSIONS AND IMPLICATIONS: PZM21 exhibited antinociceptive efficacy, without rewarding or reinforcing properties. However, its clinical application may be restricted, as it induces tolerance and withdrawal symptoms. Notably, its ability to diminish morphine reward implies that PZM21 may be useful in treatment of opioid use disorders.


Subject(s)
Analgesics, Opioid/pharmacology , Behavior, Animal/drug effects , Locomotion/drug effects , Morphine/antagonists & inhibitors , Thiophenes/pharmacology , Urea/analogs & derivatives , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/chemical synthesis , Animals , Dose-Response Relationship, Drug , Drug Delivery Systems , Injections, Intravenous , Injections, Spinal , Male , Mice , Mice, Inbred C57BL , Morphine/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Structure-Activity Relationship , Thiophenes/administration & dosage , Thiophenes/chemical synthesis , Urea/administration & dosage , Urea/chemical synthesis , Urea/pharmacology
12.
Neuropharmacology ; 141: 126-138, 2018 10.
Article in English | MEDLINE | ID: mdl-30170081

ABSTRACT

We have previously reported that the miR-181a/Prox1/Notch1 pathway mediates the effect of morphine on modulating lineage-specific differentiation of adult neural stem/progenitor cells (NSPCs) via a PKCε-dependent pathway, whereas fentanyl shows no such effect. However, the role of the PKCε/Prox1 pathway in mediating drug-associated contextual memory remains unknown. The current study investigated the effect of PKCε/Prox1 on morphine-induced inhibition of adult neurogenesis and drug-associated contextual memory in mice, while the effect of fentanyl was tested simultaneously. By using BrdU labeling, we were able to examine the lineages of differentiated NSPCs in adult DG. PKCε knockout blocked morphine's effects on inducing in vivo astrocyte-preferential differentiation of NSPCs, but did not alter NSPC lineages upon fentanyl treatment. Inhibited adult neurogenesis further resulted in prolonged extinction and enhanced reinstatement of morphine-induced CPP, as well as prolonged extinction of space reference memory indicated by the Morris water maze paradigm. However, after fentanyl administration, no significant changes were found between wild-type and PKCε knockout mice, during either CPP or water maze tasks. When the lentivirus encoding Nestin-promoter-controlled Prox1 cDNA was injected into hippocampi of wildtype and PKCε knockout adult mice to modulate PKCε/Prox1 activity, similar effects were discovered in adult mice injected with lentivirus encoding Prox1, and more dramatic effects were found in PKCε knockout mice with concurrent Prox1 overexpression. In conclusion, morphine mediates lineage-specific NSPC differentiation, inhibits adult neurogenesis and regulates contextual memory retention via the PKCε/Prox1 pathway, which are implicated in the eventual context-associated relapse.


Subject(s)
Extinction, Psychological/drug effects , Homeodomain Proteins/metabolism , Memory/drug effects , Morphine/pharmacology , Neurogenesis/drug effects , Protein Kinase C-epsilon/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Conditioning, Psychological/drug effects , Dentate Gyrus/metabolism , Fentanyl/pharmacology , Genetic Vectors/administration & dosage , Homeodomain Proteins/genetics , Lentivirus/genetics , Maze Learning/drug effects , Mice , Mice, Knockout , Microinjections , Morphine/antagonists & inhibitors , Neural Stem Cells/cytology , Protein Kinase C-epsilon/genetics , Signal Transduction/drug effects , Tumor Suppressor Proteins/genetics
13.
Pharmacol Rep ; 70(5): 955-962, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30103176

ABSTRACT

BACKGROUND: Morphine - the main pillar of nociceptive pain management - systemic use is associated with development of tolerance and dependence. Tolerance and dependence lay a heavy burden in clinical pain management settings. An added weight to this dilemma is that effective, safe, and tolerable solution to this problem is still beyond reach. Antidepressants were reported as possible alleviators of opioid tolerance and dependence. One of the increasingly used antidepressant in clinical practice is bupropion given its high safety and tolerability profile. METHODS: The study was performed on male Balb-c mice weighing 20-30g. Hot plate test was used for assessment of bupropion (5mg/kg, ip) possible analgesic activity and enhancement of morphine acute analgesia (1 and 5mg/kg, sc). Repeated morphine (5mg/kg, sc) administration for 9days developed tolerance and dependence, bupropion (5mg/kg, ip) was concurrently administered to evaluate its potential to modulate these processes. We also biochemically analyzed bupropion effect on these phenomena through modulation of neurotransmitters (glutamate and norepinephrine), inflammatory status (nitric oxide), and pro-antioxidant balance (malondialdehyde and reduced glutathione). RESULTS: Bupropion was devoid of intrinsic analgesic activity and did not enhance morphine acute analgesia. However, bupropion significantly attenuated morphine tolerance and dependence development and abstinence syndrome with corresponding suppression of morphine induced changes in glutamate, norepinephrine, inflammatory status, and prooxidant-antioxidant balance. CONCLUSION: Bupropion efficacy in attenuation of morphine tolerance and dependence with its high safety and tolerability profile provide an alternative option to conventional agents e.g., ketamine and clonidine to modulate these phenomena.


Subject(s)
Brain/metabolism , Bupropion/pharmacology , Drug Tolerance , Glutamic Acid/metabolism , Inflammation/metabolism , Morphine Dependence/prevention & control , Morphine/antagonists & inhibitors , Norepinephrine/metabolism , Oxidative Stress/drug effects , Animals , Dose-Response Relationship, Drug , Glutathione/metabolism , Male , Malondialdehyde , Mice , Morphine/pharmacology , Naloxone/pharmacology , Nitric Oxide/metabolism , Organophosphorus Compounds/metabolism , Pain Measurement/drug effects , Substance Withdrawal Syndrome/prevention & control
14.
Neuropharmacology ; 140: 174-183, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30048644

ABSTRACT

Cocaine addiction treatment is difficult due to the current lack of approved pharmacotherapuetics. Several preclinical and clinical studies have demonstrated that the mu opioid receptor (MOPr) antagonist/kappa opioid receptor (KOPr) partial agonist naltrexone (NTX) reduces the subjective effects and self-administration of cocaine. However, very limited research has examined the ability of the structurally similar MOPr antagonist/KOPr partial agonist nalmefene (NMF) to reduce cocaine reward. Here we examine the effect of low (1 mg/kg) and high (10 mg/kg) doses of NTX or NMF on cocaine place preference. In vivo characterization of these NTX and NMF doses were performed to examine their effectiveness at MOPr and KOPr. RESULTS: Both NTX doses and high dose NMF significantly reduced cocaine place preference. Conversely, a significant place avoidance was observed for high dose NTX and both NMF doses. Interestingly, neither NTX nor NMF blocked cocaine-induced hyperlocomotion. High dose NTX and both NMF doses fully blocked MOPr agonist morphine-induced thermal analgesia as well as KOPr agonist U50,488H-induced locomotor discoordination. However, low dose NTX fully blocked morphine analgesia but not U50,488H locomotor discoordination suggesting that low dose NTX is effective at MOPr but not KOPr. CONCLUSION: Both NTX and NMF block the place preference, but not locomotor activating, effects of cocaine. These results suggest that both NTX and NMF may be viable pharmacotheraputics for some aspects of cocaine addiction. This is an important step to understanding the potential mechanism(s) of action of NTX and NMF for the development of more efficacious pharmacological treatments for substance use disorders.


Subject(s)
Cocaine/antagonists & inhibitors , Conditioning, Psychological/drug effects , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/antagonists & inhibitors , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Analgesia , Analgesics, Opioid/pharmacology , Animals , Cocaine/pharmacology , Dose-Response Relationship, Drug , Locomotion/drug effects , Male , Mice , Morphine/antagonists & inhibitors , Morphine/pharmacology , Narcotic Antagonists/pharmacology
15.
Vet Anaesth Analg ; 45(2): 190-194, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29409803

ABSTRACT

OBJECTIVE: To evaluate whether subcutaneous (SC) metoclopramide (0.2 mg kg-1) administered 30 minutes prior to (T30) or simultaneously with (T0) intramuscular (IM) morphine (0.2 mg kg-1) and dexmedetomidine (0.003 mg kg-1) reduces the incidence of nausea and emesis in healthy dogs. STUDY DESIGN: Prospective, randomized and blinded study. ANIMALS: A total of 45 dogs scheduled for elective procedures. METHODS: Dogs were assigned randomly to three groups to be administered SC metoclopramide (0.2 mg kg-1) 30 minutes before (group M30) or simultaneously (group M0) to IM morphine (0.2 mg kg-1) and dexmedetomidine (0.003 mg kg-1). Dogs in the control group (group C) were administered SC saline at T30 and T0. Dogs were observed for 30 minutes after premedication to evaluate signs of nausea (continuous lip-licking and sialorrhoea) and emesis. Signs of pain or discomfort caused by SC injections were also recorded. RESULTS: There were no statistical differences amongst groups for age, body weight and sex. More dogs developed continuous lip-licking in group C (12/15, 80.0%) compared to dogs in group M30 (1/15, 6.7%) and dogs in group M0 (5/15, 33.3%; p = 0.0001 and p = 0.01, respectively). More dogs developed sialorrhoea in group M0 (8/15, 53.3%) and in group C (10/15, 66.7%) compared to dogs in group M30 (2/15, 13.3%; p = 0.03 and p = 0.004, respectively). More dogs vomited in group M0 (4/15, 26.7%) and in group C (9/15, 60.0%) compared to dogs in group M30 (0/15, 0.0%; p = 0.05 and p = 0.0003, respectively). None of the dogs demonstrated signs of pain or discomfort during SC metoclopramide injection. CONCLUSIONS AND CLINICAL RELEVANCE: Subcutaneous metoclopramide at 0.2 mg kg-1 may reduce IM morphine and dexmedetomidine-induced nausea and emesis if administered 30 minutes in advance. It is effective in reducing lip-licking even when administered concurrently with IM morphine-dexmedetomidine.


Subject(s)
Analgesics, Opioid/adverse effects , Antiemetics/therapeutic use , Dogs , Metoclopramide/therapeutic use , Morphine/adverse effects , Nausea/veterinary , Vomiting/veterinary , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/antagonists & inhibitors , Animals , Dexmedetomidine/administration & dosage , Dopamine Antagonists/therapeutic use , Female , Male , Morphine/administration & dosage , Morphine/antagonists & inhibitors , Nausea/chemically induced , Premedication/veterinary , Single-Blind Method , Vomiting/chemically induced
16.
Planta Med ; 84(4): 221-224, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28793355

ABSTRACT

This study sought to determine whether the cannabis constituent cannabidiol attenuates the development of morphine reward in the conditioned place preference paradigm. Separate groups of mice received either saline or morphine in combination with one of four doses of cannabidiol using three sets of drug/no-drug conditioning trials. After drug-place conditioning, morphine mice displayed robust place preference that was attenuated by 10 mg/kg cannabidiol. Further, when administered alone, this dose of cannabidiol was void of rewarding and aversive properties. The finding that cannabidiol blocks opioid reward suggests that this compound may be useful in addiction treatment settings.


Subject(s)
Cannabidiol/pharmacology , Morphine/pharmacology , Spatial Behavior/drug effects , Animals , Conditioning, Operant/drug effects , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Morphine/antagonists & inhibitors , Reward
17.
Neurochem Int ; 110: 101-113, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28958601

ABSTRACT

An increasing number of studies over the past few years have demonstrated ghrelin's role in alcohol, cocaine and nicotine abuse. However, the role of ghrelin in opioid effects has rarely been examined. Recently we substantiated in rats that ghrelin growth hormone secretagogue receptors (GHS-R1A) appear to be involved in acute opioid-induced changes in the mesolimbic dopaminergic system associated with the reward processing. The aim of the present study was to ascertain whether a ghrelin antagonist (JMV2959) was able to inhibit morphine-induced biased conditioned place preference and challenge-morphine-induced accumbens dopaminergic sensitization and behavioral sensitization in adult male rats. In the place preference model, the rats were conditioned for 8 days with morphine (10 mg/kg s.c.). On the experimental day, JMV2959 (3 and 6 mg/kg i.p.) or saline were administered before testing. We used in vivo microdialysis to determine changes of dopamine and its metabolites in the nucleus accumbens in rats following challenge-morphine dose (5 mg/kg s.c.) with or without JMV2959 (3 and 6 mg/kg i.p.) pretreatment, administered on the 12th day of spontaneous abstinence from morphine repeated treatment (5 days, 10-40 mg/kg). Induced behavioral changes were simultaneously monitored. Pretreatment with JMV2959 significantly and dose dependently reduced the morphine-induced conditioned place preference and significantly and dose dependently reduced the challenge-morphine-induced dopaminergic sensitization and affected concentration of by-products associated with dopamine metabolism in the nucleus accumbens. JMV2959 pretreatment also significantly reduced challenge-morphine-induced behavioral sensitization. Our present data suggest that GHS-R1A antagonists deserve to be further investigated as a novel treatment strategy for opioid addiction.


Subject(s)
Analgesics, Opioid/administration & dosage , Conditioning, Operant/drug effects , Dopamine , Morphine/administration & dosage , Nucleus Accumbens/drug effects , Receptors, Ghrelin/antagonists & inhibitors , Analgesics, Opioid/antagonists & inhibitors , Animals , Conditioning, Operant/physiology , Dopamine/metabolism , Glycine/analogs & derivatives , Glycine/pharmacology , Male , Microdialysis/methods , Morphine/antagonists & inhibitors , Nucleus Accumbens/metabolism , Rats , Rats, Wistar , Receptors, Ghrelin/metabolism , Triazoles/pharmacology
18.
Elife ; 62017 08 15.
Article in English | MEDLINE | ID: mdl-28826490

ABSTRACT

Transient receptor potential (TRP) ion channels in peripheral sensory neurons are functionally regulated by hydrolysis of the phosphoinositide PI(4,5)P2 and changes in the level of protein kinase mediated phosphorylation following activation of various G protein coupled receptors. We now show that the activity of TRPM3 expressed in mouse dorsal root ganglion (DRG) neurons is inhibited by agonists of the Gi-coupled µ opioid, GABA-B and NPY receptors. These agonist effects are mediated by direct inhibition of TRPM3 by Gßγ subunits, rather than by a canonical cAMP mediated mechanism. The activity of TRPM3 in DRG neurons is also negatively modulated by tonic, constitutive GPCR activity as TRPM3 responses can be potentiated by GPCR inverse agonists. GPCR regulation of TRPM3 is also seen in vivo where Gi/o GPCRs agonists inhibited and inverse agonists potentiated TRPM3 mediated nociceptive behavioural responses.


Subject(s)
GTP-Binding Protein beta Subunits/antagonists & inhibitors , GTP-Binding Protein gamma Subunits/antagonists & inhibitors , Ion Channels/drug effects , Sensory Receptor Cells/drug effects , TRPM Cation Channels/drug effects , Analgesics, Opioid/antagonists & inhibitors , Animals , Baclofen/antagonists & inhibitors , CHO Cells , Calcium/analysis , Capsaicin , Cricetulus , Electrophysiology/methods , Female , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Ganglia, Spinal/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphine/antagonists & inhibitors , Pain/metabolism , Pain Measurement , Phosphatidylinositols/metabolism , Receptor, Cannabinoid, CB1/agonists , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism
19.
PLoS One ; 12(4): e0175090, 2017.
Article in English | MEDLINE | ID: mdl-28380057

ABSTRACT

Opiates, one of the oldest known drugs, are the benchmark for treating pain. Regular opioid exposure also induces euphoria making these compounds addictive and often misused, as shown by the current epidemic of opioid abuse and overdose mortalities. In addition to the effect of opioids on their cognate receptors and signaling cascades, these compounds also induce multiple adaptations at cellular and behavioral levels. As omega-3 polyunsaturated fatty acids (n-3 PUFAs) play a ubiquitous role in behavioral and cellular processes, we proposed that supplemental n-3 PUFAs, enriched in docosahexanoic acid (DHA), could offset these adaptations following chronic opioid exposure. We used an 8 week regimen of n-3 PUFA supplementation followed by 8 days of morphine in the presence of this diet. We first assessed the effect of morphine in different behavioral measures and found that morphine increased anxiety and reduced wheel-running behavior. These effects were reduced by dietary n-3 PUFAs without affecting morphine-induced analgesia or hyperlocomotion, known effects of this opiate acting at mu opioid receptors. At the cellular level we found that morphine reduced striatal DHA content and that this was reversed by supplemental n-3 PUFAs. Chronic morphine also increased glutamatergic plasticity and the proportion of Grin2B-NMDARs in striatal projection neurons. This effect was similarly reversed by supplemental n-3 PUFAs. Gene analysis showed that supplemental PUFAs offset the effect of morphine on genes found in neurons of the dopamine receptor 2 (D2)-enriched indirect pathway but not of genes found in dopamine receptor 1(D1)-enriched direct-pathway neurons. Analysis of the D2 striatal connectome by a retrogradely transported pseudorabies virus showed that n-3 PUFA supplementation reversed the effect of chronic morphine on the innervation of D2 neurons by the dorsomedial prefontal and piriform cortices. Together these changes outline specific behavioral and cellular effects of morphine that can be reduced or reversed by dietary n-3 PUFAs.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Morphine/pharmacology , Animals , Anxiety/chemically induced , Corpus Striatum/chemistry , Drug Administration Schedule , Female , Frontal Lobe/chemistry , Lipids/analysis , Locomotion/drug effects , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Morphine/administration & dosage , Morphine/antagonists & inhibitors , Motor Activity/drug effects , Real-Time Polymerase Chain Reaction , Receptors, Glutamate/analysis
20.
Pain Physician ; 20(3): E401-E409, 2017 03.
Article in English | MEDLINE | ID: mdl-28339439

ABSTRACT

BACKGROUND: Dezocine is a powerful analgesic that can be less addictive than morphine, yet how the two drugs interact in vivo is poorly understood. Here we administered dezocine alone or in combination with morphine to different acute nociception paradigms to explore the interactions of the 2 drugs upon co-administration. OBJECTIVE: To evaluate how dezocine interacts with morphine in different acute nociception paradigms. STUDY DESIGN: Laboratory animal study. SETTING: Zhejiang University School of Medicine, Hangzhou, China. METHODS: Healthy mice were treated with saline, dezocine (0.625 - 2.5 µg), or a combination of dezocine with morphine (2.5 µg). Tail withdrawal latency (TWL) was analyzed prior to and 30 minutes after drug administration. Rats were treated with saline, morphine (3 mg/kg), dezocine (3 mg/kg), or a combination of both drugs. The animals were then left uninjured, subjected to plantar incision, or underwent formaldehyde-induced acute inflammation. Nociception was then analyzed in terms of mechanical threshold (MT) to von Frey stimulation and paw withdrawal latency (PWL) to thermal stimulation. Formaldehyde-induced pain score was calculated based on the duration of biting and elevating of the animal's legs. Phosphorylation of extracellular signal-regulated kinase (pERK) was also measured after plantar incision as a molecular index of nociception. RESULTS: Dezocine enhanced TWL but inhibited morphine analgesia in a dose-dependent fashion in mice. Usage of morphine or dezocine alone in uninjured rats increased MT, but co-administering both drugs did not further increase MT. Usage of one drug alone, and both drugs together increased MT and PWL relative to saline at 30 minutes after incision. Usage of one drug alone, but not both drugs together, increased MT and PWL at 120 minutes after incision. Dezocine reduced formaldehyde-induced nociception but co-administering both drugs did not further reduce pain behavior. LIMITATIONS: The results were obtained from animal study; clinical investigations will be needed to clarify their interaction. CONCLUSION: Dezocine antagonizes morphine analgesia on acute nociception upon simultaneous administration.Key words: Dezocine, morphine, acute nociception, analgesia.


Subject(s)
Analgesics, Opioid/antagonists & inhibitors , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Drug Interactions , Morphine/antagonists & inhibitors , Nociception , Tetrahydronaphthalenes/pharmacology , Analgesia , Animals , China , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Pain Measurement , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...