Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Molecules ; 26(16)2021 Aug 22.
Article in English | MEDLINE | ID: mdl-34443679

ABSTRACT

Alcohol consumption is associated with gut dysbiosis, increased intestinal permeability, endotoxemia, and a cascade that leads to persistent systemic inflammation, alcoholic liver disease, and other ailments. Craving for alcohol and its consequences depends, among other things, on the endocannabinoid system. We have analyzed the relative role of central vs. peripheral cannabinoid CB1 receptors (CB1R) using a "two-bottle" as well as a "drinking in the dark" paradigm in mice. The globally acting CB1R antagonist rimonabant and the non-brain penetrant CB1R antagonist JD5037 inhibited voluntary alcohol intake upon systemic but not upon intracerebroventricular administration in doses that elicited anxiogenic-like behavior and blocked CB1R-induced hypothermia and catalepsy. The peripherally restricted hybrid CB1R antagonist/iNOS inhibitor S-MRI-1867 was also effective in reducing alcohol consumption after oral gavage, while its R enantiomer (CB1R inactive/iNOS inhibitor) was not. The two MRI-1867 enantiomers were equally effective in inhibiting an alcohol-induced increase in portal blood endotoxin concentration that was caused by increased gut permeability. We conclude that (i) activation of peripheral CB1R plays a dominant role in promoting alcohol intake and (ii) the iNOS inhibitory function of MRI-1867 helps in mitigating the alcohol-induced increase in endotoxemia.


Subject(s)
Alcohol Drinking/pathology , Cannabinoid Receptor Antagonists/pharmacology , Endotoxemia/pathology , Ethanol/adverse effects , Nitric Oxide Synthase Type II/antagonists & inhibitors , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Alcohol Drinking/blood , Animals , Anxiety/blood , Anxiety/complications , Behavior, Animal/drug effects , Catalepsy/chemically induced , Catalepsy/complications , Cyclohexanols/administration & dosage , Elevated Plus Maze Test , Endotoxemia/blood , Endotoxemia/complications , Endotoxins/blood , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/metabolism , Hypothermia, Induced , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism , Pyrazoles/administration & dosage , Receptor, Cannabinoid, CB1/metabolism , Rimonabant/administration & dosage , Rimonabant/pharmacology , Stereoisomerism , Sulfonamides/administration & dosage
2.
Neuropharmacology ; 184: 108416, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33271186

ABSTRACT

Early-life stress induces an abnormal brain development and increases the risk of psychiatric diseases, including depression, anxiety and substance use disorders. We have developed a reliable model for maternal neglect, named maternal separation with early weaning (MSEW) in CD1 mice. In the present study, we evaluated the long-term effects on anxiety-like behaviours, nociception as well as the Iba1-positive microglial cells in this model in comparison to standard nest (SN) mice. Moreover, we investigated whether MSEW alters the cannabinoid agonist WIN55,212-2 effects regarding reward, spatial and emotional memories, tolerance to different cannabinoid responses, and physical dependence. Adult male offspring of MSEW group showed impaired responses on spatial and emotional memories after a repeated WIN55,212-2 treatment. These behavioural impairments were associated with an increase in basolateral amygdala and hippocampal CB1-expressing fibres and higher number of CB1-containing cells in cerebellum. Additionally, MSEW promotes a higher number of Iba1-positive microglial cells in basolateral amygdala and cerebellum. As for the cannabinoid-induced effects, rearing conditions did not influence the rewarding effects of WIN55,212-2 in the conditioned place preference paradigm. However, MSEW mice showed a delay in the development of tolerance to the cannabinoid effects. Moreover, CB1-positive fibres were reduced in limbic areas in MSEW mice after cannabinoid withdrawal precipitated with the CB1 antagonist SR141617A. These findings support that early-life stress promotes behavioural and molecular changes in the sensitivity to cannabinoids, which are mediated by alterations in CB1 signalling in limbic areas and it induces an increased Iba1-microglial marker which could interfere in emotional memories formation.


Subject(s)
Benzoxazines , Brain , Cannabinoid Receptor Agonists , Maternal Deprivation , Morpholines , Naphthalenes , Receptor, Cannabinoid, CB1 , Animals , Female , Male , Mice , Pregnancy , Animals, Newborn , Avoidance Learning/drug effects , Avoidance Learning/physiology , Benzoxazines/administration & dosage , Brain/drug effects , Brain/metabolism , Cannabinoid Receptor Agonists/administration & dosage , Cannabinoid Receptor Antagonists/administration & dosage , Gene Expression , Maze Learning/drug effects , Maze Learning/physiology , Morpholines/administration & dosage , Naphthalenes/administration & dosage , Pain Measurement/drug effects , Pain Measurement/methods , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/biosynthesis , Rimonabant/administration & dosage , Stress, Psychological
3.
Pharmacol Biochem Behav ; 195: 172966, 2020 08.
Article in English | MEDLINE | ID: mdl-32526216

ABSTRACT

Withdrawal from Δ9-tetrahyrocannibidol (THC) is associated with a host of dysphoric symptoms that increase probability of relapse. To date, many animal models of THC withdrawal rely on withdrawal-induced somatic withdrawal signs leaving withdrawal-suppressed behavior relatively unexplored. As compared with withdrawal-induced behaviors, ongoing behavior that is suppressed by withdrawal is a useful behavioral endpoint because it 1) more effectively models the subjective aspects of withdrawal and 2) identifies pharmacotherapies that restore behavior to baseline levels, rather than eliminate behavior induced by withdrawal. The current study assessed effects of spontaneous and rimonabant-precipitated THC withdrawal in mice responding on a progressive-ratio (PR) schedule of sucrose water reinforcement. Once behavior stabilized, male and female mice were administered THC (10 mg/kg, s.c.) or vehicle for five or six days. THC was either discontinued and behavior monitored for three days during abstinence, or the CB1 antagonist rimonabant (2 mg/kg, i.p.) was used to precipitate withdrawal. Whereas spontaneous THC withdrawal had no effect on PR performance, THC-treated mice were differentially sensitive to rimonabant administration via large decreases in break point, overall response rate, and run rate relative to vehicle-treated mice. Importantly, pretreatment with the CB1 positive allosteric modulator ZCZ011 (10 mg/kg, i.p.) did not prevent precipitated-withdrawal-induced behavioral impairment. These extend findings of earlier studies suggesting operant baselines are useful tools to study subjective effects of cannabinoid withdrawal. Additionally, operant baselines allow withdrawal pharmacotherapies to be tested in a restoration-of-function context, which may be more sensitive, selective, and clinically relevant.


Subject(s)
Cannabinoid Receptor Agonists/administration & dosage , Dronabinol/administration & dosage , Motivation , Reinforcement, Psychology , Substance Withdrawal Syndrome/psychology , Sucrose/pharmacology , Animals , Behavior, Animal/drug effects , Cannabinoid Receptor Antagonists/administration & dosage , Female , Indoles/administration & dosage , Male , Mice , Mice, Inbred C57BL , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Rimonabant/administration & dosage , Substance Withdrawal Syndrome/drug therapy , Thiophenes/administration & dosage
4.
Behav Brain Res ; 390: 112697, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32417279

ABSTRACT

Cannabinoid subtype 1 receptor (CB1R) antagonists were originally developed as anti-obesity agents. Unfortunately, SR1417116A (rimonabant), the first marketed inverse agonist of CB1R, produced CNS-related adverse effects including depression and suicidal ideation, and thus it was withdrawn from the market. These effects of rimonabant became evident in patients following chronic dosing. Standard preclinical toxicity studies failed to detect these adverse effects. The goal of these studies was to perform an integrated battery of behavioral assays to better understand the behavioral effects of rimonabant following both acute and chronic administration. In the present study, acute dosing with rimonabant in rats significantly decreased food consumption; decreased measures of locomotor activity; increased scratching, grooming and wet-dog shakes; and increased defecation. Subsequently, animals were tested using a chronic dosing regimen but prior to drug administration for that day. The highest dose of rimonabant tested significantly decreased marble burying behavior, presumably anxiolysis. There were also significant effects in social interaction after chronic dosing. Our results did not reveal significant rimonabant-induced anxiogenic behaviors. Future studies to characterize behavioral screens for anxiogenic effects of CB1 antagonists in rodents should further explore social interaction paradigms and potential comorbid factors of rimonabant dosing such as sex, age, and obesity.


Subject(s)
Behavior, Animal/drug effects , Cannabinoid Receptor Antagonists/pharmacology , Locomotion/drug effects , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Rimonabant/pharmacology , Social Behavior , Animals , Cannabinoid Receptor Antagonists/administration & dosage , Cannabinoid Receptor Antagonists/adverse effects , Male , Rats , Rats, Sprague-Dawley , Rimonabant/administration & dosage , Rimonabant/adverse effects
5.
Sleep Breath ; 24(4): 1441-1449, 2020 Dec.
Article in English | MEDLINE | ID: mdl-31898189

ABSTRACT

OBJECTIVE: The endocannabinoid system (ECS) regulates bone turn-over and remodeling. Chronic intermittent hypoxia (CIH) occurring during obstructive sleep apnea (OSA) may lead to disorders of the ECS and bone metabolism abnormalities. This study aimed to investigate whether or not the cannabinoid receptor 1 (CB1R) antagonist rimonabant (Ri) alleviates bone metabolism abnormalities and bone destruction induced by chronic intermittent hypoxia (CIH). METHODS: Healthy male Sprague Dawley (SD) rats (n=48) were randomly divided into 6 groups of 8 rats: 2 normal control (NC) groups, 2 intermittent hypoxia (IH) groups, and 2 IH + Ri groups. Rats in NC groups breathed room air for 4 weeks (4w NC group) and 6 weeks (6w NC group). Rats in IH groups experienced IH environment for 4 weeks (4w IH group) and 6 weeks (6w IH group). In addition to the same IH exposure, rats in IH + Ri group were given daily intraperitoneal injection of Ri at the dosage of 1.5 mg/kg/d for 4 weeks (4w IH + Ri group) and 6 weeks (6w IH + Ri group). Levels of serum tartrate-resistant acid phosphatase (TRAP, a marker of bone resorption) were determined by ELISA. Hematoxylin and eosin (HE) staining was performed on bone sections to observe the changes in bone microstructure. Expression of CB1R in bone tissue was determined by immunohistochemistry. RESULTS: TRAP levels were higher in the 4w IH and 6w IH groups than in the 4w NC and 6w NC groups; TRAP levels were lower in the 4w IH + Ri and 6w IH + Ri groups than in the 4w IH and 6w IH groups. HE staining showed that the morphology of bone cells in the NC group was normal, but the 4w IH group had mild edema of bone cells, reduction in trabecular bone, and destruction of bone microstructure. Changes were more severe in the 6w IH group than 4w IH. The 4w IH + Ri group was slightly improved compared with the 4w IH group. The 6w IH + Ri group was improved compared with the 4w IH + Ri group. The results of immunohistochemistry showed that the expression of CB1R in IH group was significantly higher than that in NC group. The expression of CB1R in the IH + Ri group was lower than that in the IH group. With the prolongation of hypoxia, the expression of CB1R in bone cells of IH group increased. The expression level of CB1R in IH + Ri group decreased with the prolongation of intervention time. Correlation analysis showed that the expression rate of CB1R in bone cells was positively correlated with the level of TRAP in serum. CONCLUSION: CIH increases serum TRAP levels and triggers metabolic bone disorder by activating bone CB1R. Intervention with CB1R antagonist (rimonabant) reduces the bone dysmetabolism in the CIH rat model.


Subject(s)
Bone Diseases, Metabolic/physiopathology , Cannabinoid Receptor Antagonists/administration & dosage , Hypoxia/physiopathology , Protective Agents/administration & dosage , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Rimonabant/administration & dosage , Animals , Bone Diseases, Metabolic/drug therapy , Bone Diseases, Metabolic/metabolism , Disease Models, Animal , Male , Osteoclasts/drug effects , Osteoclasts/metabolism , Rats, Sprague-Dawley , Tartrate-Resistant Acid Phosphatase/blood
6.
Addict Biol ; 25(1): e12691, 2020 01.
Article in English | MEDLINE | ID: mdl-30378732

ABSTRACT

Cessation from prolonged use of ∆9 -tetrahydrocannabinol (THC), the primary active compound responsible for the cannabimimetic effects of cannabis, results in a mild to moderate withdrawal syndrome in humans and laboratory animals. Whereas manipulations of the endogenous cannabinoid system (eg, cannabinoid receptors and endocannabinoid regulating enzymes) alter nicotine withdrawal, in this study we asked the reciprocal question. Do nicotinic acetylcholine receptors (nAChRs) modulate THC withdrawal? To assess the role of different nAChR subtypes in THC withdrawal, we used transgenic mouse, preclinical pharmacological, and human genetic correlation approaches. Our findings show that selective α3ß4* nAChR antagonist, AuIB, and α3ß4* nAChR partial agonist, AT-1001, dose-dependently attenuated somatic withdrawal signs in THC-dependent mice that were challenged with the cannabinoid-1 receptor antagonist rimonabant. Additionally, THC-dependent α5 and α6 nAChR knockout (KO) mice displayed decreased rimonabant precipitated somatic withdrawal signs compared with their wild-type counterparts. In contrast, ß2 and α7 nAChR KO mice showed no alterations in THC withdrawal signs. Moreover, deletion of ß2 nAChR did not alter the reduced expression of somatic signs by the preferred α6ß4* antagonist, BulA [T5A;P60]. Finally, the human genetic association studies indicated that variations in the genes that code for the α5, α3, ß4, and α6 nAChRs were associated with cannabis disorder phenotypes. Overall, these findings suggest that α3ß4* and α6ß4* nAChR subtypes represent viable targets for the development of medications to counteract THC dependence.


Subject(s)
Dronabinol/pharmacology , Marijuana Abuse/physiopathology , Receptors, Nicotinic/drug effects , Substance Withdrawal Syndrome/physiopathology , Animals , Cannabinoid Receptor Antagonists/administration & dosage , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Rimonabant/administration & dosage
7.
J Pharmacol Exp Ther ; 372(1): 119-127, 2020 01.
Article in English | MEDLINE | ID: mdl-31641018

ABSTRACT

Despite a growing acceptance that withdrawal symptoms can emerge following discontinuation of cannabis products, especially in high-intake chronic users, there are no Food and Drug Administration (FDA)-approved treatment options. Drug development has been hampered by difficulties studying cannabis withdrawal in laboratory animals. One preclinical approach that has been effective in studying withdrawal from drugs in several pharmacological classes is antagonist drug discrimination. The present studies were designed to examine this paradigm in squirrel monkeys treated daily with the long-acting CB1 agonist AM2389 (0.01 mg/kg) and trained to discriminate the CB1 inverse agonist/antagonist rimonabant (0.3 mg/kg) from saline. The discriminative-stimulus effects of rimonabant were both dose and time dependent and, importantly, could be reproduced by discontinuation of agonist treatment. Antagonist substitution tests with the CB1 neutral antagonists AM4113 (0.03-0.3 mg/kg), AM6527 (0.03-1.0 mg/kg), and AM6545 (0.03-1.0 mg/kg) confirmed that the rimonabant discriminative stimulus also could be reproduced by CB1 antagonists lacking inverse agonist action. Agonist substitution tests with the phytocannabinoid ∆9-tetrahydrocannabinol (0.1-1.0 mg/kg), synthetic CB1 agonists nabilone (0.01-0.1 mg/kg), AM4054 (0.01-0.03 mg/kg), K2/Spice compound JWH-018 (0.03-0.3 mg/kg), FAAH-selective inhibitors AM3506 (0.3-5.6 mg/kg), URB597 (3.0-5.6 mg/kg), and nonselective FAAH/MGL inhibitor AM4302 (3.0-10.0 mg/kg) revealed that only agonists with CB1 affinity were able to reduce the rimonabant-like discriminative stimulus effects of withholding daily agonist treatment. Although the present studies did not document physiologic disturbances associated with withdrawal, the results are consistent with the view that the cannabinoid antagonist drug discrimination paradigm provides a useful screening procedure for examining the ability of candidate medications to attenuate the interoceptive stimuli provoked by cannabis discontinuation. SIGNIFICANCE STATEMENT: Despite a growing acceptance that withdrawal symptoms can emerge following the discontinuation of cannabis products, especially in high-intake chronic users, there are no FDA-approved pharmacotherapies to assist those seeking treatment. The present studies systematically examined cannabinoid antagonist drug discrimination, a preclinical animal model that is designed to appraise the ability of candidate medications to attenuate the interoceptive effects that accompany abrupt cannabis abstinence.


Subject(s)
Cannabinoid Receptor Antagonists/therapeutic use , Discrimination, Psychological , Disease Models, Animal , Substance Withdrawal Syndrome/drug therapy , Animals , Benzopyrans/administration & dosage , Benzopyrans/adverse effects , Benzopyrans/therapeutic use , Cannabinoid Receptor Agonists/administration & dosage , Cannabinoid Receptor Agonists/adverse effects , Cannabinoid Receptor Agonists/therapeutic use , Cannabinoid Receptor Antagonists/administration & dosage , Cannabinoid Receptor Antagonists/adverse effects , Drug Evaluation, Preclinical/methods , Drug Substitution/methods , Male , Rimonabant/administration & dosage , Rimonabant/adverse effects , Rimonabant/therapeutic use , Saimiri , Substance Withdrawal Syndrome/psychology
8.
Pharmacol Biochem Behav ; 183: 72-79, 2019 08.
Article in English | MEDLINE | ID: mdl-31202811

ABSTRACT

One of most formidable problems in the treatment of addiction is the high rate of relapse. The discovery of medicines to help mitigate relapse are aided by animal models that currently involve weeks of training and require surgical preparations and drug delivery devices. The present set of experiments was initiated to investigate a rapid 8-day screening method that utilizes food instead of intravenous drug administration. Male Sprague-Dawley rats were trained in a reinstatement paradigm in which every lever press produced a 45 mg food pellet concurrently paired with a light and tone. Behavior was subsequently extinguished with lever responses producing neither food nor food-associated stimuli. Reinstatement of responding was evaluated under conditions in which the first three responses of every 5 min time bin produced a food pellet along with food-associated stimuli. The mGlu5 receptor antagonists MPEP and MTEP produced a significant reduction in reinstatement while failing to alter responding where every response produced food. The cannabinoid CB1 receptor antagonist rimonabant and the mGlu2/3 receptor agonist LY379268 also selectively reduced reinstatement. Other compounds including clozapine, d-amphetamine, chlordiazepoxide, ABT-431, naltrexone and citalopram were without effect. The results suggest that relapse-like behavioral effects can be extended to non-pharmacological reinforcers. Drug effects demonstrated both behavioral and pharmacological specificity. The present experimental design thus allows for efficient and rapid assessment of the effects of drugs that might be useful in the treatment of addiction-associated relapse.


Subject(s)
Behavior, Addictive/drug therapy , Behavior, Addictive/psychology , Behavior, Animal/drug effects , Drug Discovery/methods , Models, Animal , Amino Acids/administration & dosage , Amino Acids/pharmacology , Animals , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cannabinoid Receptor Antagonists/administration & dosage , Cannabinoid Receptor Antagonists/pharmacology , Conditioning, Operant/drug effects , Excitatory Amino Acid Agonists/administration & dosage , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/pharmacology , Extinction, Psychological/drug effects , Feeding Behavior/drug effects , Male , Pyridines/administration & dosage , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Recurrence , Rimonabant/administration & dosage , Rimonabant/pharmacology , Self Administration , Substance-Related Disorders/drug therapy , Thiazoles/administration & dosage , Thiazoles/pharmacology
9.
PLoS One ; 14(3): e0209947, 2019.
Article in English | MEDLINE | ID: mdl-30861005

ABSTRACT

Cannabis withdrawal upon discontinuation of long-term, heavy Cannabis use is reported in humans; however, methods to establish the nature and intensity of cannabinoid withdrawal, especially directly observable signs, have not been widely established. This study quantified activity in the home cage of rhesus monkeys and examined the extent to which activity can be used to quantify tolerance to and dependence on Δ9-tetrahydrocannabinol (Δ9-THC). Home-cage activity was measured in one group that received Δ9-THC (1 mg/kg s.c.) every 12 h (i.e., chronic Δ9-THC), and a second group that received Δ9-THC (0.1 mg/kg i.v.) once every 3 days (i.e., intermittent Δ9-THC). Treatment was temporarily discontinued in the chronic Δ9-THC group and the effects of rimonabant and Δ9-THC were examined in both groups. Activity counts were highest during the day (lights on 0600-2000 h) and were lower at night. Rimonabant (0.1-3.2 mg/kg i.v.) dose-dependently increased activity (maximum 20-fold) in the chronic Δ9-THC group but did not significantly alter activity in the intermittent Δ9-THC group. Δ9-THC (0.32-3.2 mg/kg i.v.) dose-dependently decreased activity counts (maximum 4-fold) in both groups but was somewhat more potent in the intermittent as compared with the Δ9-THC group. Discontinuation of Δ9-THC treatment resulted in an immediate (i.e., within 24 h) and time-related increase in activity. The time-related increase in home-cage activity upon abrupt discontinuation of chronic Δ9-THC treatment, as well as the effects of rimonabant to increase activity in monkeys receiving chronic, but not intermittent, Δ9-THC treatment, are consistent with signs of physical dependence on Δ9-THC in primates.


Subject(s)
Dronabinol/adverse effects , Drug Tolerance , Hallucinogens/adverse effects , Motor Activity/drug effects , Rimonabant/administration & dosage , Substance Withdrawal Syndrome/drug therapy , Administration, Intravenous , Animals , Disease Models, Animal , Female , Hallucinogens/pharmacology , Humans , Macaca mulatta , Male , Rimonabant/pharmacology
10.
PLoS One ; 14(1): e0205781, 2019.
Article in English | MEDLINE | ID: mdl-30645588

ABSTRACT

Reconsolidation is a process in which re-exposure to a reminder causes a previously acquired memory to undergo a process of destabilisation followed by subsequent restabilisation. Different molecular mechanisms have been postulated for destabilisation in the amygdala and hippocampus, including CB1 receptor activation, protein degradation and AMPA receptor exchange; however, most of the amygdala studies have used pre-reexposure interventions, while those in the hippocampus have usually performed them after reexposure. To test whether the temporal window for destabilisation is similar across both structures, we trained Lister Hooded rats in a contextual fear conditioning task, and 1 day later performed memory reexposure followed by injection of either the NMDA antagonist MK-801 (0.1 mg/kg) or saline in order to block reconsolidation. In parallel, we also performed local injections of either the CB1 antagonist SR141716A or its vehicle in the hippocampus or in the amygdala, either immediately before or immediately after reactivation. Infusion of SR141716A in the hippocampus prevented the reconsolidation-blocking effect of MK-801 when performed after reexposure, but not before it. In the amygdala, meanwhile, pre-reexposure infusions of SR141716A impaired reconsolidation blockade by MK-801, although the time-dependency of this effect was not as clear as in the hippocampus. Our results suggest the temporal windows for CB1-receptor-mediated memory destabilisation during reconsolidation vary between brain structures. Whether this reflects different time windows for engagement of these structures or different roles played by CB1 receptors in destabilisation across structures remains an open question for future studies.


Subject(s)
Amygdala/physiology , Fear/physiology , Hippocampus/physiology , Memory/physiology , Receptor, Cannabinoid, CB1/metabolism , Amygdala/drug effects , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Cannabinoid Receptor Antagonists/administration & dosage , Conditioning, Classical/drug effects , Dizocilpine Maleate/administration & dosage , Excitatory Amino Acid Antagonists/administration & dosage , Fear/drug effects , Hippocampus/drug effects , Male , Memory/drug effects , Models, Animal , Rats , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/metabolism , Rimonabant/administration & dosage , Time Factors
11.
Curr Opin Lipidol ; 30(2): 140-146, 2019 04.
Article in English | MEDLINE | ID: mdl-30649023

ABSTRACT

PURPOSE OF REVIEW: The endocannabinoid system affects several physiological functions. A family of endocannabinoid receptors is susceptible to cannabis constituents. Cannabis is widely used in our society and following its recent legalization in Canada, we focus on how exposure to cannabis and pharmacologic cannabinoid receptor type 1 (CB1) inhibition affect lipoprotein levels. RECENT FINDINGS: Several groups have reported that exposure to cannabis does not increase weight despite the marked increase in caloric intake. In observational studies, the effect of smoked cannabis exposure on plasma lipids is variable. Some studies in specific patient populations with longer exposure to cannabis seemed to identify slightly more favorable lipoprotein profiles in the exposed group. Several larger controlled clinical trials using orally administered rimonabant, a CB1 receptor antagonist, have consistently shown relative improvements in weight and plasma levels of triglyceride and high-density lipoprotein cholesterol among patients receiving the treatment. SUMMARY: The widely variable findings on the relationship of cannabis in various forms with plasma lipids preclude any definitive conclusions. Cannabis has complex effects on the cardiovascular system and its effects on lipid profile must be considered in this overall context. Further properly controlled research is required to better understand this topic.


Subject(s)
Cannabinoids/pharmacology , Cannabis/chemistry , Cholesterol, HDL/blood , Energy Intake/drug effects , Lipid Metabolism/drug effects , Triglycerides/blood , Anti-Obesity Agents/administration & dosage , Body Weight/drug effects , Cannabinoid Receptor Antagonists/administration & dosage , Case-Control Studies , Gene Expression , Humans , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Rimonabant/administration & dosage
12.
Folia Med (Plovdiv) ; 61(4): 540-544, 2019 12 31.
Article in English | MEDLINE | ID: mdl-32337866

ABSTRACT

INTRODUCTION: Endocannabinoid system is involved in neuropsychiatric disorders such as major depression. The bilaterally olfactory bulbectomized rat is widely used as an animal model of depression. The removal of the olfactory bulbs produces behavioural, physiological, and neurochemical alterations resembling clinical depression. There is increasing evidence that highlights the important role of cannabinoid signalling in depression and nociception. AIM: To investigate the effect of CB1 receptor agonist HU 210 and CB1 receptor antagonist SR 141716A administered icv subchronically (for 7 days) on nociception of rats with model of depression - bilateral olfactory bulbectomy (OBX). MATERIAL AND METHODS: Experimental model of depression - bilateral olfactory bulbectomy (OBX). Bilaterally olfactory bulbectomized rats were used as an experimental model of depression. HU 210 (5 µg) or SR 141716A (3 µg) were infused icv for 7 consecutive days, starting 15 days after the olfactory bulbectomy. Nociception was examined by applying paw pressure test (analgesy-meter) evaluating the rat pain threshold. On day 7, five minutes after the last microinjection, the rats were tested in an analgesy-meter and their mechanically evoked pain responses were measured in arbitrary units (AU). RESULTS: Microinjections of HU 210 (5 µg) significantly decreased the pain threshold in olfactory bulbectomized rats, while SR 141716A (3 µg) exerted antinociceptive effect by increasing the pain threshold. CONCLUSIONS: Data point to an involvement of CB1 receptors in depression-like behaviour and nociception in olfactory bulbectomized rats and support the data for the association between depressive disorder and pain pathways.


Subject(s)
Depression/drug therapy , Dronabinol/analogs & derivatives , Nociception/drug effects , Receptor, Cannabinoid, CB1/physiology , Rimonabant/administration & dosage , Animals , Disease Models, Animal , Dronabinol/administration & dosage , Ligands , Male , Olfactory Bulb/surgery , Pain Threshold/drug effects , Rats , Rats, Wistar
13.
J Pharm Biomed Anal ; 160: 119-125, 2018 Oct 25.
Article in English | MEDLINE | ID: mdl-30077950

ABSTRACT

A highly sensitive and selective liquid chromatography-tandem mass spectrometry method for the determination of tetrahydrocannabinol (THC), cannabidiol, and rimonabant in rat plasma was developed. Analytes and the internal standard were extracted from plasma using a combination of protein precipitation followed by liquid-liquid extraction. Chromatographic separation was done using Waters Symmetry C18, 4.6 × 150 mm, 5 um column using 10 mm ammonium formate buffer and methanol. The total run time was 6 min, and separation was achieved using isocratic elution at a flow rate of 1 mL/min using a 10:90 (aqueous: organic) ratio. The ionization of the analytes was optimized using electrospray ionization in positive mode, and multiple reaction mode was used for this analysis. This method showed linearity from 0.1 to 100 ng/ml for all the analytes and was validated according to FDA Bioanalytical Method Validation Guidance in terms of accuracy, precession, linearity, stability, matrix effect, recovery, and stability. This method was successfully applied to characterize the pharmacokinetics of THC in rats after continuous passive smoke exposure for 50 min when rimonabant was co-administered with cannabis smoke. Maximum concentration (Cmax) for THC was observed immediately after rats were removed from the exposure chamber (10 min post completion) which declined with a terminal half-life of 3.7 h and clearance was calculated to be 1.1 (L/h). Rimonabant (i.p) at a dose of 3 mg/kg was rapidly absorbed and maximum concentration (Cmax) was seen at 11 min which declined with a terminal half-life of 5.4 h and clearance was calculated to be 2.0 (L/h). Exposure AUCinf (h* µg/L) for THC and rimonabant were 13.9 and 457.6 respectively. As this method was highly sensitive and required only 50 µL of plasma, it is applicable in rodent models that assess the exposure-response relationships of these drugs.


Subject(s)
Cannabidiol/blood , Dronabinol/blood , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Rimonabant/pharmacokinetics , Animals , Chromatography, High Pressure Liquid/methods , Dose-Response Relationship, Drug , Injections, Intraperitoneal , Limit of Detection , Male , Models, Animal , Rats , Rats, Wistar , Rimonabant/administration & dosage , Sensitivity and Specificity , Tandem Mass Spectrometry/methods , Tobacco Smoke Pollution
14.
Neuroscience ; 392: 230-240, 2018 11 10.
Article in English | MEDLINE | ID: mdl-30031835

ABSTRACT

Postnatal ethanol exposure has been shown to cause persistent defects in hippocampal synaptic plasticity and disrupt learning and memory processes. However, the mechanisms responsible for these abnormalities are less well studied. We evaluated the influence of postnatal ethanol exposure on several signaling and epigenetic changes and on expression of the activity-regulated cytoskeletal (Arc) protein in the hippocampus of adult offspring under baseline conditions and after a Y-maze spatial memory (SP) behavior (activity). Postnatal ethanol treatment impaired pCaMKIV and pCREB in naïve mice without affecting H4K8ac, H3K14ac and H3K9me2 levels. The Y-maze increased pCaMKIV, pCREB, H4K8ac and H3K14ac levels in saline-treated mice but not in ethanol-treated mice; while H3K9me2 levels were enhanced in ethanol-exposed animals compared to saline groups. Like previous observations, ethanol not only reduced Arc expression in naïve mice but also behaviorally induced Arc expression. ChIP results suggested that reduced H3K14ac and H4K8ac in the Arc gene promoter is because of impaired CBP, and increased H3K9me2 is due to the enhanced recruitment of G9a. The CB1R antagonist and a G9a/GLP inhibitor, which were shown to rescue postnatal ethanol-triggered synaptic plasticity and learning and memory deficits, were able to prevent the negative effects of ethanol on activity-dependent signaling, epigenetics and Arc expression. Together, these findings provide a molecular mechanism involving signaling and epigenetic cascades that collectively are responsible for the neurobehavioral deficits associated with an animal model of fetal alcohol spectrum disorders (FASD).


Subject(s)
Epigenesis, Genetic , Ethanol/administration & dosage , Hippocampus/drug effects , Hippocampus/metabolism , Spatial Learning/drug effects , Spatial Memory/drug effects , Animals , Cannabinoid Receptor Antagonists/administration & dosage , Chromatin Assembly and Disassembly/drug effects , Cytoskeletal Proteins/metabolism , Female , Fetal Alcohol Spectrum Disorders/metabolism , Male , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/drug effects , Rimonabant/administration & dosage , Signal Transduction
15.
Neurobiol Aging ; 69: 199-208, 2018 09.
Article in English | MEDLINE | ID: mdl-29909177

ABSTRACT

Contradictory findings on the role of the type 1 cannabinoid receptor (CB1R) during the pathogenesis of Alzheimer's disease (AD) have been reported. Here, we evaluated the CB1R brain profile in an AD mouse model using longitudinal positron emission tomography with an inverse agonist for CB1R, [18F]FMPEP-d2. APP/PS1-21 and wild-type (n = 8 in each group) mice were repeatedly imaged between 6 to 15 months of age, accompanied by brain autoradiography, western blot, and CB1R immunohistochemistry with additional mice. [18F]FMPEP-d2 positron emission tomography demonstrated lower (p < 0.05) binding ratios in the parietotemporal cortex and hippocampus of APP/PS1-21 mice compared with age-matched wild-type mice. Western blot demonstrated no differences between APP/PS1-21 and wild-type mice in the CB1R abundance, whereas significantly lower (p < 0.05) receptor expression was observed in male than female mice. The results provide the first demonstration that [18F]FMPEP-d2 is a promising imaging tool for AD research in terms of CB1R availability, but not expression. This finding may further facilitate the development of novel therapeutic approaches based on endocannabinoid regulation.


Subject(s)
Aging , Alzheimer Disease/metabolism , Brain/metabolism , Genotype , Receptor, Cannabinoid, CB1/metabolism , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/genetics , Animals , Brain/drug effects , Cannabinoid Receptor Antagonists/administration & dosage , Disease Models, Animal , Female , Male , Mice, Inbred C57BL , Mice, Transgenic , Positron-Emission Tomography , Pyrrolidinones , Rimonabant/administration & dosage
16.
Sci Rep ; 7(1): 11678, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28916833

ABSTRACT

In a high percentage (≥85%) of both sporadic and familial adenomatous polyposis forms of colorectal cancer (CRC), the inactivation of the APC tumor suppressor gene initiates tumor formation and modulates the Wnt/ß-Catenin transduction pathways involved in the control of cell proliferation, adhesion and metastasis. Increasing evidence showed that the endocannabinoids control tumor growth and progression, both in vitro and in vivo. We evaluated the effect of Rimonabant, a Cannabinoid Receptor 1 (CB1) inverse agonist, on the Wnt/ß-Catenin pathway in HCT116 and SW48 cell lines carrying the genetic profile of metastatic CRC poorly responsive to chemotherapies. In these models, Rimonabant inhibited the Wnt/ß-Catenin canonical pathway and increased ß-Catenin phosphorylation; in HCT116 cells, but not in SW48, the compound also triggered the Wnt/ß-Catenin non canonical pathway activation through induction of Wnt5A and activation of CaMKII. The Rimonabant-induced downregulation of Wnt/ß-Catenin target genes was partially ascribable to a direct inhibition of p300/KAT3B histone acetyltransferase, a coactivator of ß-Catenin dependent gene regulation. Finally, in HCT116 xenografts, Rimonabant significantly reduced tumor growth and destabilized the nuclear localization of ß-Catenin. Obtained data heavily supported the rationale for the use of cannabinoids in combined therapies for metastatic CRC harbouring activating mutations of ß-Catenin.


Subject(s)
Cannabinoid Receptor Antagonists/pharmacology , Colonic Neoplasms/drug therapy , Colorectal Neoplasms/drug therapy , Histone Acetyltransferases/antagonists & inhibitors , Rimonabant/pharmacology , Wnt Signaling Pathway/drug effects , Animals , Cannabinoid Receptor Antagonists/administration & dosage , Cell Line, Tumor , Colorectal Neoplasms/pathology , Disease Models, Animal , Female , Heterografts , Humans , Mice, SCID , Models, Biological , Neoplasm Transplantation , Rimonabant/administration & dosage , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...