Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 158
Filter
Add more filters

Publication year range
1.
Cell Mol Neurobiol ; 41(5): 1131-1143, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33433723

ABSTRACT

Chronic administration of opioids produces physical dependence and opioid-induced hyperalgesia. Users claim the Thai traditional tea "kratom" and component alkaloid mitragynine ameliorate opioid withdrawal without increased sensitivity to pain. Testing these claims, we assessed the combined kratom alkaloid extract (KAE) and two individual alkaloids, mitragynine (MG) and the analog mitragynine pseudoindoxyl (MP), evaluating their ability to produce physical dependence and induce hyperalgesia after chronic administration, and as treatments for withdrawal in morphine-dependent subjects. C57BL/6J mice (n = 10/drug) were administered repeated saline, or graded, escalating doses of morphine (intraperitoneal; i.p.), kratom alkaloid extract (orally, p.o.), mitragynine (p.o.), or MP (subcutaneously, s.c.) for 5 days. Mice treated chronically with morphine, KAE, or mitragynine demonstrated significant drug-induced hyperalgesia by day 5 in a 48 °C warm-water tail-withdrawal test. Mice were then administered naloxone (10 mg/kg, s.c.) and tested for opioid withdrawal signs. Kratom alkaloid extract and the two individual alkaloids demonstrated significantly fewer naloxone-precipitated withdrawal signs than morphine-treated mice. Additional C57BL/6J mice made physically dependent on morphine were then used to test the therapeutic potential of combined KAE, mitragynine, or MP given twice daily over the next 3 days at either a fixed dose or in graded, tapering descending doses. When administered naloxone, mice treated with KAE, mitragynine, or MP under either regimen demonstrated significantly fewer signs of precipitated withdrawal than control mice that continued to receive morphine. In conclusion, while retaining some liabilities, kratom, mitragynine, and mitragynine pseudoindoxyl produced significantly less physical dependence and ameliorated precipitated withdrawal in morphine-dependent animals, suggesting some clinical value.


Subject(s)
Analgesics, Opioid/adverse effects , Mitragyna , Morphine Dependence/prevention & control , Secologanin Tryptamine Alkaloids/administration & dosage , Secologanin Tryptamine Alkaloids/chemical synthesis , Substance Withdrawal Syndrome/prevention & control , Analgesics, Opioid/administration & dosage , Animals , Male , Mice , Mice, Inbred C57BL , Morphine Dependence/metabolism , Morphine Dependence/psychology , Pain Measurement/drug effects , Pain Measurement/methods , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Secologanin Tryptamine Alkaloids/adverse effects , Secologanin Tryptamine Alkaloids/isolation & purification , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/psychology
2.
Pharm Res ; 38(7): 1263-1278, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34128146

ABSTRACT

A method to reproducibly mill abuse deterrent oxycodone hydrochloride (HCl) extended release (ER) tablets was developed for a nasal insufflation pharmacokinetic (PK) study. Several comminution methods were explored before determining that a conical mill resulted in controlled milling of tablets to a size range equal to or below 1000 µm. However, milling resulted in significant loss of oxycodone from abuse deterrent oxycodone HCl ER tablets compared to minimal oxycodone loss from oxycodone HCl immediate release (IR) tablets. Characterization of milled tablet powder showed that loss of oxycodone was not attributed to analytical procedures or oxycodone phase change during high intensity milling processes. The content uniformity of oxycodone in the milled tablet powder varied when ER and IR tablets were milled to a particle size distribution equal to or below 500 µm but did not vary when particles were sized above 500 µm to equal to or below 1000 µm. In addition, the initial excipient weight to drug substance weight ratio impacted the amount of oxycodone lost from the respective formulation. However, dissolution demonstrated that when oxycodone HCl ER tablets are milled, differences in excipient weight to drug substance weight ratio and particle size distribution of milled tablets did not result in significantly different release of oxycodone.


Subject(s)
Abuse-Deterrent Formulations , Analgesics, Opioid/chemistry , Drug Compounding/methods , Morphine Dependence/prevention & control , Oxycodone/chemistry , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacokinetics , Chemistry, Pharmaceutical , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Drug Liberation , Insufflation , Oxycodone/administration & dosage , Oxycodone/pharmacokinetics , Powders , Single Molecule Imaging , Tablets
3.
Neuropsychobiology ; 80(3): 264-270, 2021.
Article in English | MEDLINE | ID: mdl-33207349

ABSTRACT

OBJECTIVE: Hydrogen sulfide is an endogenous gaseous mediator that has been indicated to have a role in pain mechanisms. In this study, we aimed to detect brain and spinal cord hydrogen sulfide levels during different phases of tolerance and dependence to morphine and to determine the effects of inhibition of endogenous hydrogen sulfide production on the development of tolerance and dependence. METHODS: Morphine tolerance and dependence was developed by subcutaneous injection of morphine (10 mg/kg) twice daily for 12 days. Physical dependence was determined by counting the jumps for 20 min, which is a withdrawal symptom occurring after a single dose of naloxone (5 mg/kg) administered intraperitoneally (i.p.). Propargylglycine (30 mg/kg, i.p.), a cystathionine-γ-lyase inhibitor, and hydroxylamine (12.5 mg/kg, i.p.), a cystathionine-ß-synthase inhibitor, were used as hydrogen sulfide synthase inhibitors. The tail-flick and hot-plate tests were used to determine the loss of antinociceptive effects of morphine and development of tolerance. RESULTS: It was found that chronic and acute uses of both propargylglycine and hydroxylamine prevented the development of tolerance to morphine, whereas they had no effect on morphine dependence. Chronic and acute administrations of hydrogen sulfide synthase inhibitors did not exert any difference in hydrogen sulfide levels in brain and spinal cords of both morphine-tolerant and -dependent animals. CONCLUSION: It has been concluded that hydrogen sulfide synthase inhibitors may have utility in preventing morphine tolerance.


Subject(s)
Behavior, Animal/drug effects , Drug Tolerance , Enzyme Inhibitors/pharmacology , Hydrogen Sulfide/metabolism , Morphine Dependence/prevention & control , Alkynes/pharmacology , Animals , Disease Models, Animal , Enzyme Inhibitors/administration & dosage , Glycine/analogs & derivatives , Glycine/pharmacology , Hydroxylamine/pharmacology , Male , Mice , Mice, Inbred BALB C , Morphine/administration & dosage , Narcotics/administration & dosage
4.
Acta Pharmacol Sin ; 40(5): 583-588, 2019 May.
Article in English | MEDLINE | ID: mdl-30224637

ABSTRACT

Increasing preclinical evidence demonstrates that dopamine D3 receptor (D3R) antagonists are a potential option for the treatment of drug addiction. The reinstatement of the addiction can be triggered by environmental stimuli that acquire motivational salience through repeated associations with the drug's effects. YQA14 is a novel D3R antagonist that has exhibited pharmacotherapeutic efficacy in reducing cocaine and amphetamine reward and relapse to drug seeking in mice. In this study we investigated the effects of YQA14 on morphine-induced context-specific locomotor sensitization in mice. We showed that repeated injection of YQA14 (6.25-25 mg/kg every day ip) prior to morphine (10 mg/kg every day sc) not only inhibited the acquisition, but also significantly attenuated the expression of morphine-induced locomotor sensitization. Furthermore, in the expression phase, one single injection of YQA14 (6.25-25 mg/kg, ip) dose-dependently inhibited the expression of morphine-induced behavioral sensitization. Moreover, YQA14 inhibited the expression of morphine-induced behavioral sensitization in wild mice (WT), but not in D3R knockout (D3R-/-) mice in the expression phase. In addition, D3R-/- mice also displayed the reduction in the expression phase compared with WT mice. In summary, this study demonstrates that blockade or knockout of the D3R inhibits morphine-induced behavior sensitization, suggesting that D3R plays an important role in the pathogenesis and etiology of morphine addiction, and it might be a potential target for clinical management of opioid addiction.


Subject(s)
Benzoxazoles/therapeutic use , Dopamine Antagonists/therapeutic use , Morphine Dependence/prevention & control , Morphine/pharmacology , Motor Activity/drug effects , Piperazines/therapeutic use , Receptors, Dopamine D3/antagonists & inhibitors , Animals , Benzoxazoles/administration & dosage , Benzoxazoles/pharmacokinetics , Dopamine Antagonists/administration & dosage , Dopamine Antagonists/pharmacokinetics , Drug-Seeking Behavior , Injections, Intraperitoneal , Male , Mice, Knockout , Morphine/administration & dosage , Morphine Dependence/etiology , Morphine Dependence/psychology , Piperazines/administration & dosage , Piperazines/pharmacokinetics , Receptors, Dopamine D3/genetics
5.
Mol Pharmacol ; 93(2): 49-62, 2018 02.
Article in English | MEDLINE | ID: mdl-29192123

ABSTRACT

The CB2 cannabinoid agonist LY2828360 lacked both toxicity and efficacy in a clinical trial for osteoarthritis. Whether LY2828360 suppresses neuropathic pain has not been reported, and its signaling profile is unknown. In vitro, LY2828360 was a slowly acting but efficacious G protein-biased CB2 agonist, inhibiting cAMP accumulation and activating extracellular signal-regulated kinase 1/2 signaling while failing to recruit arrestin, activate inositol phosphate signaling, or internalize CB2 receptors. In wild-type (WT) mice, LY2828360 (3 mg/kg per day i.p. × 12 days) suppressed chemotherapy-induced neuropathic pain produced by paclitaxel without producing tolerance. Antiallodynic efficacy of LY2828360 was absent in CB2 knockout (KO) mice. Morphine (10 mg/kg per day i.p. × 12 days) tolerance developed in CB2KO mice but not in WT mice with a history of LY2828360 treatment (3 mg/kg per day i.p. × 12 days). LY2828360-induced antiallodynic efficacy was preserved in WT mice previously rendered tolerant to morphine (10 mg/kg per day i.p. × 12 days), but it was absent in morphine-tolerant CB2KO mice. Coadministration of LY2828360 (0.1 mg/kg per day i.p. × 12 days) with morphine (10 mg/kg per day × 12 days) blocked morphine tolerance in WT but not in CB2KO mice. WT mice that received LY2828360 coadministered with morphine exhibited a trend (P = 0.055) toward fewer naloxone-precipitated jumps compared with CB2KO mice. In conclusion, LY2828360 is a slowly signaling, G protein-biased CB2 agonist that attenuates chemotherapy-induced neuropathic pain without producing tolerance and may prolong effective opioid analgesia while reducing opioid dependence. LY2828360 may be useful as a first-line treatment in chemotherapy-induced neuropathic pain and may be highly efficacious in neuropathic pain states that are refractive to opioid analgesics.


Subject(s)
Analgesics, Opioid/administration & dosage , Cannabinoid Receptor Agonists/pharmacology , Drug Tolerance , GTP-Binding Proteins/metabolism , Morphine Dependence/prevention & control , Morphine/administration & dosage , Neuralgia/prevention & control , Purines/pharmacology , Receptor, Cannabinoid, CB2/agonists , Signal Transduction , Analgesics, Opioid/adverse effects , Animals , Antineoplastic Agents/administration & dosage , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphine/adverse effects , Naloxone/administration & dosage , Narcotic Antagonists/administration & dosage , Paclitaxel/administration & dosage , Purines/administration & dosage , Receptor, Cannabinoid, CB2/metabolism , Substance Withdrawal Syndrome
6.
Addict Biol ; 23(5): 1055-1066, 2018 09.
Article in English | MEDLINE | ID: mdl-28961365

ABSTRACT

Accumulating evidence indicated that minocycline, a glial cell modulator, is able to modify a variety of morphine effects. Here, we investigated minocycline effects on electrical activity of nucleus accumbens (NAc) neurons using single unit recording in urethane-anesthetized rats. In addition, we investigated whether minocycline can modify the effects of morphine on NAc neural activity during reinstatement of morphine-seeking behavior. Minocycline increased the NAc firing activity in intact animals. Electrophysiological recording in morphine-treated animals was performed, following the acquisition of morphine-induced conditioned place preference (5 mg/kg, s.c., 3 days) and a drug-free extinction period. In acutely minocycline- treated animals, the neurons were recorded for 40 minutes following a single injection of either minocycline (50 µg/5 µl, i.c.v.) or saline. Then a priming dose of morphine (1 mg/kg, s.c.) was injected while the recording was continued for an additional 40 minutes. Minocycline significantly increased the firing rates of neurons and significantly modified morphine inhibitory effects on NAc neurons. In subchronically minocycline-treated groups, the rats were given daily injections of minocycline (50 µg/5 µl, i.c.v) during the extinction period. Then, on the reinstatement day, NAc neurons were recorded for 10 minutes, the priming dose of morphine was administered and the recording was continued for 45 minutes. Our results showed the failure of minocycline to significantly modify the inhibitory effects of morphine. In conclusion, our findings indicated that minocycline modifies morphine-induced decreases in the firing rates of NAc neurons in the reinstatement phase.


Subject(s)
Minocycline/pharmacology , Morphine/pharmacology , Neurons/drug effects , Nucleus Accumbens/drug effects , Analgesics, Opioid/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Conditioning, Classical , Disease Models, Animal , Drug-Seeking Behavior/drug effects , Electrodiagnosis/methods , Male , Morphine Dependence/prevention & control , Rats , Rats, Wistar
7.
Behav Pharmacol ; 28(8): 681-689, 2017 12.
Article in English | MEDLINE | ID: mdl-28125510

ABSTRACT

Dendrosomal nanocurcumin (DNC) is fabricated from esterification of oleic acid and polyethylene glycol residues with curcumin. DNC has shown antioxidant, neuroprotective, and neurogenesis-enhancing effects. In addition, it can attenuate morphine tolerance. Morphine self-administration is associated with neurodegenerative changes of CA1 neurons in the adult hippocampus. The present study evaluated the effect of DNC pretreatment on morphine self-administration and hippocampal damage. Rats were pretreated with DNC (5 and 10 mg/kg, intraperitoneally) 30 min before a morphine self-administration paradigm performed in 2-h/sessions for 12 days under a FR-1 schedule. Pretreatment with both doses of DNC markedly suppressed morphine intake. Morphine self-administration resulted in a 71% reduction in the number of hippocampal CA1 neurons. DNC (5 mg/kg) pretreatment only marginally improved (by 22%) neuronal loss in this area. The data suggest that the effect of DNC on morphine self-administration is largely independent of the CA1 area. A functional restoration and regulation of reward circuit activity by DNC may reduce the motivation for morphine despite CA1 damage.


Subject(s)
Central Nervous System Agents/administration & dosage , Curcumin/administration & dosage , Morphine Dependence/prevention & control , Animals , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/pathology , Catheters, Indwelling , Cell Count , Disease Models, Animal , Male , Morphine/administration & dosage , Morphine Dependence/pathology , Narcotics/administration & dosage , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/pathology , Neurons/drug effects , Neurons/pathology , Random Allocation , Rats, Wistar , Self Administration
8.
Am J Drug Alcohol Abuse ; 43(3): 291-298, 2017 05.
Article in English | MEDLINE | ID: mdl-28448223

ABSTRACT

BACKGROUND: US FDA guidance recommends measuring the degree of effort needed to manipulate abuse-deterrent (AD) opioids. The ALERRT® instrument (PinneyAssociates; Bethesda, MD) uses visual analog scales to assess the labor, effort, and resources necessary to physically compromise AD product candidates in standardized settings. OBJECTIVE: Use the ALERRT® instrument for testing morphine abuse-deterrent, extended-release, injection-molded tablets (ADER-IMT) 60 and 100 mg and the comparators immediate-release (IR) morphine sulfate 30 mg and extended-release (ER) morphine sulfate 60 mg. METHODS: Four technicians tested the products using 10 household tools. The ALERRT instrument quantified effort (all tools) and time (3 preselected tools) required for manipulation. RESULTS: Morphine-ADER-IMT 60 and 100 mg were difficult to manipulate, as demonstrated by high scores (mean range, 71.0-99.0 and 77.0-99.5, respectively). IR and ER morphine sulfate were easy to manipulate (low scores; mean range, 2.0-14.8 and 2.3-17.5, respectively). Statistically significant mean differences between morphine-ADER-IMT and comparators' ALERRT scores were observed. Manipulations of morphine-ADER-IMT 60 and 100 mg for 300 seconds failed to produce substantial powdering. Manipulations of IR morphine sulfate (mean range, 65.5-175.8 seconds) and ER morphine sulfate (49.3-163.0 seconds) produced substantial to complete powdering in 92% of tablets. CONCLUSIONS: Morphine-ADER-IMT was extremely difficult to manipulate versus non-AD formulations of morphine. The ALERRT system differentiated the degree of effort for manipulation of morphine-ADER-IMT and non-AD morphine formulations, indicating sensitivity of this instrument as part of Category 1 testing. By measuring the degree of effort required for manipulation, the ALERRT instrument provides an empirical assessment into the relative difficulty of manipulating opioid analgesics for abuse.


Subject(s)
Analgesics, Opioid/administration & dosage , Morphine Dependence/prevention & control , Morphine/administration & dosage , Substance-Related Disorders/prevention & control , Analgesics, Opioid/chemistry , Delayed-Action Preparations , Humans , Morphine/chemistry , Opioid-Related Disorders/prevention & control , Tablets , United States , United States Food and Drug Administration
9.
Orv Hetil ; 157(18): 695-9, 2016 May 01.
Article in Hungarian | MEDLINE | ID: mdl-27106724

ABSTRACT

Short acting oral formulas make part of optimal opioid analgesia. The use of short acting oral morphine has not widely spread in Hungary, and these drugs completely lacked from the market for three years. Since December 2015 short acting morphine-sulphate has again been commercialised. The causes of the market failure are analysed in this article. The aim of the retrospective analysis is to help the accessibility of the medicine to every patient in need. Prescription morphine use depends on the harmonised cooperation of a number of actors. Besides regulating and financing authorities and professional organisations, patients and the opinion forming media are also responsible for building up the right routine.


Subject(s)
Analgesics, Opioid , Analgesics, Short-Acting , Health Services Accessibility , Morphine Dependence/prevention & control , Morphine Derivatives , Pain/drug therapy , Administration, Oral , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Analgesics, Opioid/pharmacology , Analgesics, Short-Acting/administration & dosage , Analgesics, Short-Acting/adverse effects , Drug Industry , Education, Medical, Continuing , Fentanyl/administration & dosage , Government Agencies , Health Services Accessibility/legislation & jurisprudence , Health Services Accessibility/organization & administration , Health Services Accessibility/statistics & numerical data , Health Services Accessibility/trends , Humans , Hungary , Insurance, Health , Mass Media , Morphine Derivatives/administration & dosage , Morphine Derivatives/adverse effects , Morphine Derivatives/pharmacology , Pain/etiology , Practice Patterns, Physicians' , Universities
11.
Ig Sanita Pubbl ; 72(1): 87-95, 2016.
Article in Italian | MEDLINE | ID: mdl-27077562

ABSTRACT

The treatment of drug addiction is a very wide-ranging sector within modern medicine. The use of immunotherapy in this context represents an innovative approach. The purpose of this paper is to illustrate, through a literature review, the main avenues of research and the results obtained with immunotherapy in the treatment of drug addiction.


Subject(s)
Substance-Related Disorders/prevention & control , Vaccines/therapeutic use , Cocaine-Related Disorders/prevention & control , Humans , Morphine Dependence/prevention & control , Substance-Related Disorders/immunology , Tobacco Use Disorder/prevention & control , Treatment Outcome
12.
Am J Drug Alcohol Abuse ; 41(5): 405-13, 2015.
Article in English | MEDLINE | ID: mdl-26337199

ABSTRACT

BACKGROUND: Salvia officinalis L. (SO) has effects on the central nervous system, including anti-addiction properties that may involve an opioid mechanism. OBJECTIVE: Effects of a hydroalcoholic extract of SO on nociception and on morphine-induced tolerance and dependence were evaluated in rats. METHODS: Tolerance and dependence were induced by injection of morphine (10 mg/kg, s.c.) or escalating doses of morphine (2.5, 2.5, 5, 10, 20, 40 and 50 mg/kg, s.c.) twice daily for 7 days. SO (400, 600 and 800 mg/kg, i.g.) was administered before morphine. The tail-flick and naloxone precipitation withdrawal tests were used to evaluate tolerance and dependence. Sedative effects as well as total polyphenolic and flavonoid were also measured. RESULTS: The morphine-treated group showed significant decrements in the percentage maximum possible effect (%MPE) on days 5 and 7 compared to the first day, illustrating morphine tolerance. Higher doses decreased morphine tolerance. Furthermore, SO (600 and 800 mg/kg) attenuated almost all of the withdrawal signs including weight loss, jumping, penis licking, teeth chattering, wet dog shakes, rearing, standing, sniffing, face grooming and paw tremor and increased sleep duration (64.5 ± 9.7, 100.3 ± 4.7, respectively). Total polyphenolic and flavonoid content of SO was 138 and 69 mg per g of dried extract, respectively. CONCLUSION: SO has antinociceptive effects and may decrease tolerance and dependence induced by repeated morphine administration. However, to determine whether treatment with SO blocks tolerance by interfering with neurobiological mechanisms that mediate the development of morphine tolerance will require further studies.


Subject(s)
Analgesics/pharmacology , Drug Tolerance , Hypnotics and Sedatives/pharmacology , Morphine Dependence/prevention & control , Morphine/pharmacology , Plant Extracts/pharmacology , Salvia officinalis/chemistry , Analgesics, Opioid/pharmacology , Animals , Dose-Response Relationship, Drug , Hypnotics and Sedatives/chemistry , Male , Plant Extracts/chemistry , Plant Leaves/chemistry , Rats , Substance Withdrawal Syndrome/prevention & control
13.
Pharm Biol ; 52(11): 1382-7, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25068674

ABSTRACT

CONTEXT: Scutellaria baicalensis Georgi (Lamiaceae) has been used as a traditional herbal preparation for the treatment of neuropsychiatric disorders in Asian countries for centuries. OBJECTIVE: To evaluate the effects of S. baicalensis on morphine-induced drug dependence in rats. MATERIALS AND METHODS: In order to evaluate the effect of S. baicalensis and baicalin on morphine-induced dependence-like behavior, a water extract of S. baicalensis [500 mg/kg, intraperitoneally (i.p.)] or baicalin (50 mg/kg, i.p., a flavonoid found in S. baicalensis) was administered prior to morphine injection [5 and 2.5 mg/kg, respectively, subcutaneously (s.c.)] to rats for 8 and 4 d, respectively. Morphine-induced conditioned place preference was assessed by measuring the time spent in a drug-paired chamber. The effect of S. baicalensis on dopamine receptor supersensitivity (locomotor activity) and dopamine agonist-induced climbing behavior due to a single apomorphine treatment (2 mg/kg, s.c.) was also measured. RESULTS: At 50 mg/kg, a water extract of S. baicalensis decreased morphine (5 mg/kg)-induced conditioned place preference by 86% in rats. Apomorphine (2 mg/kg)-induced locomotor activity (dopamine receptor supersensitivity) in rats and climbing behavior in mice were attenuated after pretreatment with 500 mg/kg of S. baicalensis water extract by 41% and 56%, respectively. In addition, baicalin-reduced morphine-induced conditioned places preference by 86% in rats at 50 mg/kg. DISCUSSION AND CONCLUSION: These results suggest that S. baicalensis can ameliorate drug addiction-related behavior through functional regulation of dopamine receptors.


Subject(s)
Conditioning, Operant/drug effects , Morphine Dependence/prevention & control , Morphine/pharmacology , Plant Extracts/pharmacology , Plant Roots , Animals , Conditioning, Operant/physiology , Male , Mice , Mice, Inbred ICR , Morphine Dependence/metabolism , Plant Extracts/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, Dopamine/metabolism , Scutellaria baicalensis , Water/pharmacology
14.
Pharmacol Biochem Behav ; 234: 173688, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38056696

ABSTRACT

There is accumulating evidence supporting the involvement of tissue-plasminogen activator (tPA) in the mechanisms underlying the effects of morphine and an enriched environment. This study was designed to investigate possible interactive roles of the glutamatergic and the dopaminergic systems regarding hippocampal tPA in the neurobiology of morphine dependence. For this purpose, Wistar albino rats, housed in either a standard- (SE) or an enriched environment (EE) were implanted subcutaneously with morphine (150 mg base) or placebo pellets. Behavioral and somatic signs of morphine abstinence precipitated by an opioid-receptor antagonist naloxone (1 mg/kg, i.p.) 72 h after the pellet implantation were observed individually for 15 min in all groups. Memantine (10 mg/kg i.p.), an antagonist of N-methyl-D-aspartic acid class of glutamatergic receptor-subtype decreased teeth-chattering, ptosis, diarrhea and the loss of body weight. SKF82958 (1 mg/kg, i.p.), a dopamine D1-receptor agonist decreased jumping and ptosis but increased rearing and loss of body weight. On the other hand, co-administration of SKF82958 with memantine prevented some of their effects that occur when administered alone at the same doses. Furthermore, the EE did not change the intensity of morphine abstinence. The level of hippocampal tPA mRNA was found to be lower in the SE morphine abstinence group than in the placebo group and close to the EE morphine abstinence group, whereas there was no significant alteration of its level in the memantine or SKF82958 groups. These findings suggest that the interaction between the glutamatergic and the dopaminergic systems may be an important component of the neurobiology of morphine dependence, and the role of tPA in this interaction should be further investigated.


Subject(s)
Morphine Dependence , Substance Withdrawal Syndrome , Rats , Animals , Morphine/pharmacology , Naloxone/pharmacology , Memantine/pharmacology , Morphine Dependence/prevention & control , Rats, Wistar , Substance Withdrawal Syndrome/drug therapy , Body Weight
15.
Psychopharmacology (Berl) ; 241(8): 1565-1575, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38630316

ABSTRACT

RATIONALE: Corticotropin-releasing factor (CRF), the apical stress-inducing hormone, exacerbates stress and addictive behaviors. TCAP-1 is a peptide that directly inhibits both CRF-mediated stress and addiction-related behaviors; however, the direct action of TCAP-1 on morphine withdrawal-associated behaviors has not previously been examined. OBJECTIVE: To determine whether TCAP-1 administration attenuates behavioral and physiological consequences of morphine withdrawal in mice. METHODS: Mice were administered via subcutaneous route TCAP-1 either before or after initial morphine exposure, after which jumping behavior was quantified to assess the effects of TCAP-1 on naloxone-precipitated morphine withdrawal. As a comparison, mice were treated with nonpeptide CRF1 receptor antagonist CP-154,526. In one experiment, plasma corticosterone (CORT) was also measured as a physiological stress indicator. RESULTS: Pretreatment with TCAP-1 (10-250 nmol/kg) before morphine treatment significantly inhibited the development of naloxone-precipitated withdrawal. TCAP-1 (250-500 nmol/kg) treatment administered after morphine treatment attenuated the behavioral expression of naloxone-precipitated withdrawal. TCAP-1 (250 nmol/kg) treatment during morphine treatment was more effective than the optimal dosing of CP-154,526 (20 mg/kg) at suppressing the behavioral expression of naloxone-precipitated withdrawal, despite similar reduction of withdrawal-induced plasma CORT level increases. CONCLUSIONS: These findings establish TCAP-1 as a potential therapeutic candidate for the prevention and treatment of morphine withdrawal.


Subject(s)
Corticosterone , Morphine , Naloxone , Narcotic Antagonists , Substance Withdrawal Syndrome , Animals , Male , Substance Withdrawal Syndrome/drug therapy , Substance Withdrawal Syndrome/metabolism , Naloxone/pharmacology , Naloxone/administration & dosage , Mice , Narcotic Antagonists/pharmacology , Narcotic Antagonists/administration & dosage , Morphine/administration & dosage , Morphine/pharmacology , Corticosterone/blood , Corticosterone/administration & dosage , Dose-Response Relationship, Drug , Pyrroles/pharmacology , Pyrroles/administration & dosage , Nerve Tissue Proteins/metabolism , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/metabolism , Behavior, Animal/drug effects , Morphine Dependence/metabolism , Morphine Dependence/drug therapy , Morphine Dependence/prevention & control , Pyrrolidines/pharmacology , Pyrrolidines/administration & dosage , Injections, Subcutaneous , Corticotropin-Releasing Hormone/metabolism , Corticotropin-Releasing Hormone/pharmacology , Pyrimidines
16.
Behav Pharmacol ; 24(2): 133-43, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23455447

ABSTRACT

Previous studies have reported that some of the central effects of morphine are counteracted by the administration of the methanolic extract of the root of Indian ginseng, Withania somnifera Dunal (WSE). The present study sought to determine whether WSE affects acquisition and expression of morphine-elicited conditioned place preference (CPP) in CD-1 mice. In CPP acquisition experiments, WSE (0, 25, 50, and 100 mg/kg) was administered, during conditioning, 30 min before morphine (10 mg/kg), whereas in expression experiments, WSE (0, 25, 50, and 100 mg/kg) was administered 30 min before the postconditioning test. The results demonstrate (i) that WSE was devoid of motivational properties; (ii) that WSE (100 mg/kg) was devoid of effects on spontaneous and morphine-stimulated motor activity and on spatial memory; and (iii) that WSE (50 and 100 mg/kg) significantly prevented the acquisition and expression of CPP. Further, to characterize the receptor(s) involved in these effects, we studied, by receptor-binding assay, the affinity of WSE for µ-opioid and γ-aminobutyric acid B receptors. These experiments revealed a higher affinity of WSE for γ-aminobutyric acid B than for µ-opioid receptors. Overall, these results point to WSE as an interesting alternative tool, worthy of further investigation, to study opiate addiction.


Subject(s)
Behavior, Addictive/prevention & control , Morphine Dependence/prevention & control , Phytotherapy , Plant Extracts/therapeutic use , Plant Roots/chemistry , Withania/chemistry , Animals , Animals, Outbred Strains , Behavior, Addictive/etiology , Behavior, Animal/drug effects , Binding, Competitive , Conditioning, Classical , Dose-Response Relationship, Drug , Kinetics , Ligands , Male , Medicine, Ayurvedic , Mice , Morphine Dependence/drug therapy , Morphine Dependence/physiopathology , Nerve Tissue Proteins/metabolism , Plant Extracts/chemistry , Plant Extracts/metabolism , Plant Extracts/pharmacology , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/metabolism
17.
Behav Brain Funct ; 8: 20, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22559224

ABSTRACT

BACKGROUND: Repeated morphine exposure can induce behavioral sensitization. There are evidences have shown that central gamma-aminobutyric acid (GABA) system is involved in morphine dependence. However, the effect of a GABAB receptor agonist baclofen on morphine-induced behavioral sensitization in rats is unclear. METHODS: We used morphine-induced behavioral sensitization model in rat to investigate the effects of baclofen on behavioral sensitization. Moreover, dopamine release in the shell of the nucleus accumbens was evaluated using microdialysis assay in vivo. RESULTS: The present study demonstrated that morphine challenge (3 mg/kg, s.c.) obviously enhanced the locomotor activity following 4-day consecutive morphine administration and 3-day withdrawal period, which indicated the expression of morphine sensitization. In addition, chronic treatment with baclofen (2.5, 5 mg/kg) significantly inhibited the development of morphine sensitization. It was also found that morphine challenge 3 days after repeated morphine administration produced a significant increase of extracellular dopamine release in nucleus accumbens. Furthermore, chronic treatment with baclofen decreased the dopamine release induced by morphine challenge. CONCLUSIONS: Our results indicated that gamma-aminobutyric acid system plays an important role in the morphine sensitization in rat and suggested that behavioral sensitization is a promising model to study the mechanism underlying drug abuse.


Subject(s)
Baclofen/pharmacology , Dopamine/metabolism , GABA-B Receptor Agonists/pharmacology , Morphine Dependence/prevention & control , Morphine/pharmacology , Nucleus Accumbens/drug effects , Analgesics, Opioid/pharmacology , Animals , Disease Models, Animal , Extracellular Space/metabolism , Male , Microdialysis , Morphine Dependence/drug therapy , Motor Activity/drug effects , Nucleus Accumbens/metabolism , Rats , Rats, Sprague-Dawley , Substance Withdrawal Syndrome/drug therapy , Substance Withdrawal Syndrome/prevention & control
18.
Addict Biol ; 17(2): 392-408, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21967557

ABSTRACT

Agmatine, an endogenous ligand for imidazoline receptor, has been shown to prevent opioid dependence, but not much is known about the mechanisms of the effect of agmatine. In the present study, we investigated the function of I1 imidazoline receptor and its candidate protein imidazoline receptor antisera-selected (IRAS)/Nischarin in morphine dependence as well as in the effect of agmatine inhibiting morphine dependence by pharmacological and molecular approaches. Results showed that inhibition of IRAS or Nischarin did not change the development of morphine dependence in vitro and in vivo under the basal condition. Agmatine could reduce the cyclic 3', 5' adenosine monophosphate (cAMP) overshoot at the concentration of 0.01-10 µM in the primary cultured rat hippocampal neurons and attenuated the withdrawal signals and the elevation of FosB and ΔFosB at the dose of 5 mg/kg in the morphine-dependent mice. The effect of agmatine was inhibited by efaroxan (I1 imidazoline receptor non-specific antagonist) and the RNA interference against IRAS or Nischarin. These findings indicate that I1 imidazoline receptor or IRAS/Nischarin mediates the effect of agmatine on morphine dependence and provide evidence that I1 imidazoline receptor may be a new target for treating morphine dependence.


Subject(s)
Agmatine/pharmacology , Imidazoline Receptors/physiology , Intracellular Signaling Peptides and Proteins/pharmacology , Morphine Dependence/prevention & control , Adenosine Monophosphate/metabolism , Adrenergic alpha-Antagonists/pharmacology , Animals , Benzofurans/pharmacology , Cells, Cultured , Drug Interactions , Gene Knockdown Techniques , Hippocampus/metabolism , Imidazoles/pharmacology , Imidazoline Receptors/antagonists & inhibitors , Immune Sera/pharmacology , Male , Mice , Mice, Inbred BALB C , Neurons/metabolism , Proto-Oncogene Proteins c-fos/metabolism , RNA, Small Interfering/pharmacology , Substance Withdrawal Syndrome/drug therapy
19.
Behav Brain Res ; 402: 113104, 2021 03 26.
Article in English | MEDLINE | ID: mdl-33417990

ABSTRACT

Oxidative stress and the nitric oxide (NO) pathway are involved in the development of opioid analgesic tolerance and dependence. Simvastatin modulates NO and oxidative stress, so the present study aimed to investigate its effect on the development and expression of morphine analgesic tolerance and withdrawal signs in mice. Morphine tolerance and dependence were induced by twice daily morphine injection (10 mg/kg, s.c.) for 5 consecutive days. Tolerance was assessed by the hot-plate test and dependence by naloxone challenge, on the sixth day. To determine if the NO is involved in the effects of simvastatin, mice were pre-treated with l-arginine (200 mg/kg) or the NO synthesis inhibitors (L-NAME; 30 mg/kg) along with simvastatin (300 mg/kg). The results showed that acute and chronic administration of simvastatin reversed the antinociceptive tolerance of morphine and attenuated withdrawal signs in morphine-dependent mice, and this effect is reversed by l-arginine and augmented by l-NAME. Also, the concentration of NO and oxidative stress factors such as malondialdehyde content, total thiol, and glutathione peroxidase (GPx) activity in brain tissues was evaluated. Chronic administration of simvastatin reduced NO and malondialdehyde, and increased total thiol and GPx levels in the cerebral cortex and hippocampus of morphine-dependent mice which were antagonized by l-arginine, and augmented by l-NAME. In summary, simvastatin attenuates morphine-induced antinociceptive tolerance and withdrawal symptoms, at least partly, through antioxidative properties and nitric oxide pathway.


Subject(s)
Antioxidants/pharmacology , Cerebral Cortex/drug effects , Drug Tolerance , Morphine Dependence/prevention & control , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Signal Transduction/drug effects , Simvastatin/pharmacology , Substance Withdrawal Syndrome/prevention & control , Animals , Hippocampus/drug effects , Male , Malondialdehyde/metabolism , Mice , Morphine Dependence/metabolism , Substance Withdrawal Syndrome/metabolism
20.
Neurochem Int ; 150: 105157, 2021 11.
Article in English | MEDLINE | ID: mdl-34390773

ABSTRACT

Besides their clinical application, chronic misuse of opioids has often been associated to drug addiction due to their addictive properties, underlying neuroadaptations of AMPA glutamate-receptor-dependent synaptic plasticity. Topiramate (TPM), an AMPAR antagonist, has been used to treat psychostimulants addiction, despite its harmful effects on memory. This study aimed to evaluate the effects of a novel topiramate nanosystem on molecular changes related to morphine reinstatement. Rats were previously exposed to morphine in conditioned place preference (CPP) paradigm and treated with topiramate-chitosan nanoparticles (TPM-CS-NP) or non-encapsulated topiramate in solution (S-TPM) during CPP extinction; following memory performance evaluation, they were re-exposed to morphine reinstatement. While morphine-CPP extinction was comparable among all experimental groups, TPM-CS-NP treatment prevented morphine reinstatement, preserving memory performance, which was impaired by both morphine-conditioning and S-TPM treatment. In the NAc, morphine increased D1R, D2R, D3R, DAT, GluA1 and MOR immunoreactivity. It also increased D1R, DAT, GluA1 and MOR in the dorsal hippocampus. TPM-CS-NP treatment decreased D1R, D3R and GluA1 and increased DAT in the NAc, decreasing GluA1 and increasing D2 and DAT in the dorsal hippocampus. Taken together, we may infer that TPM-CS-NP treatment was able to prevent the morphine reinstatement without memory impairment. Therefore, TPM-CS-NP may be considered an innovative therapeutic tool due to its property to prevent opioid reinstatement because it acts modifying both dopaminergic and glutamatergic neurotransmission, which are commonly related to morphine addiction.


Subject(s)
Chitosan/administration & dosage , Dopamine/metabolism , Glutamic Acid/metabolism , Morphine Dependence/metabolism , Nanoparticles/administration & dosage , Topiramate/administration & dosage , Analgesics, Opioid/pharmacology , Animals , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Drug Therapy, Combination , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Male , Memory/drug effects , Memory/physiology , Morphine/pharmacology , Morphine Dependence/prevention & control , Rats , Rats, Wistar , Receptors, AMPA/metabolism , Receptors, Dopamine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL