Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Clin Exp Pharmacol Physiol ; 50(8): 621-633, 2023 08.
Article in English | MEDLINE | ID: mdl-37194348

ABSTRACT

Mephedrone is a representative of synthetic cathinones that is known from its rewarding and psychostimulant effects. It exerts behavioural sensitization after repeated and then interrupted administration. In our study, we investigated a role of the L-arginine-NO-cGMP-dependent signalling in the expression of sensitization to hyperlocomotion evoked by mephedrone. The study was carried out in male albino Swiss mice. The tested mice received mephedrone (2.5 mg/kg) for 5 consecutive days and on the 20th day of the experiment (the 'challenge' day) animals received both mephedrone (2.5 mg/kg) and a given substance that affects the L-arginine-NO-cGMP signalling, that is, L-arginine hydrochloride (125 or 250 mg/kg), 7-nitroindazole (10 or 20 mg/kg), L-NAME (25 or 50 mg/kg) or methylene blue (5 or 10 mg/kg). We observed that 7-nitroindazole, L-NAME and methylene blue inhibited the expression of sensitization to the mephedrone-induced hyperlocomotion. Moreover, we demonstrated that the mephedrone-induced sensitization is accompanied by lowered levels of D1 receptors and NR2B subunits in the hippocampus, whereas a concurrent administration of L-arginine hydrochloride, 7-nitroindazole and L-NAME with the mephedrone challenge dose reversed these effects. Methylene blue only reversed the mephedrone-induced effects on hippocampal levels of the NR2B subunit. Our study confirms that the L-arginine-NO-cGMP pathway contributes to mechanisms underlying the expression of sensitization to the mephedrone-evoked hyperlocomotion.


Subject(s)
Methylene Blue , Nitric Oxide , Mice , Male , Animals , NG-Nitroarginine Methyl Ester/pharmacology , Methylene Blue/pharmacology , Nitric Oxide/metabolism , Arginine/pharmacology , Locomotion , Cyclic GMP/metabolism
2.
Int J Mol Sci ; 22(2)2021 Jan 08.
Article in English | MEDLINE | ID: mdl-33435576

ABSTRACT

A synthetic cathinone, mephedrone is widely abused by adolescents and young adults. Despite its widespread use, little is known regarding its long-term effects on cognitive function. Therefore, we assessed, for the first time, whether (A) repeated mephedrone (30 mg/kg, i.p., 10 days, once a day) exposure during adolescence (PND 40) induces deleterious effects on spatial memory and reversal learning (Barnes maze task) in adult (PND 71-84) rats and whether (B) these effects were comparable to amphetamine (2.5 mg/kg, i.p.). Furthermore, the influence of these drugs on MMP-9, NMDA receptor subunits (GluN1, GluN2A/2B) and PSD-95 protein expression were assessed in adult rats. The drug effects were evaluated at doses that per se induce rewarding/reinforcing effects in rats. Our results showed deficits in spatial memory (delayed effect of amphetamine) and reversal learning in adult rats that received mephedrone/amphetamine in adolescence. However, the reversal learning impairment may actually have been due to spatial learning rather than cognitive flexibility impairments. Furthermore, mephedrone, but not amphetamine, enhanced with delayed onset, MMP-9 levels in the prefrontal cortex and the hippocampus. Mephedrone given during adolescence induced changes in MMP-9 level and up-regulation of the GluN2B-containing NMDA receptor (prefrontal cortex and hippocampus) in young adult (PND 63) and adult (PND 87) rats. Finally, in adult rats, PSD-95 expression was increased in the prefrontal cortex and decreased in the hippocampus. In contrast, in adult rats exposed to amphetamine in adolescence, GluN2A subunit and PSD-95 expression were decreased (down-regulated) in the hippocampus. Thus, in mephedrone-but not amphetamine-treated rats, the deleterious effects on spatial memory were associated with changes in MMP-9 level. Because the GluN2B-containing NMDA receptor dominates in adolescence, mephedrone seems to induce more harmful effects on cognition than amphetamine does during this period of life.


Subject(s)
Amphetamine/pharmacology , Hippocampus/drug effects , Maze Learning/drug effects , Methamphetamine/analogs & derivatives , Prefrontal Cortex/drug effects , Spatial Memory/drug effects , Age Factors , Animals , Central Nervous System Stimulants/pharmacology , Cognition/drug effects , Disks Large Homolog 4 Protein/metabolism , Hippocampus/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Methamphetamine/pharmacology , Motor Activity/drug effects , Prefrontal Cortex/metabolism , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism
3.
Int J Mol Sci ; 20(17)2019 Sep 03.
Article in English | MEDLINE | ID: mdl-31484312

ABSTRACT

Opioid use disorder is classified as a chronic recurrent disease of the central nervous system (CNS) which leads to personality disorders, co-morbidities and premature death. It develops as a result of long-term administration of various abused substances, along with morphine. The pharmacological action of morphine is associated with its stimulation of opioid receptors. Opioid receptors are a group of G protein-coupled receptors and activation of these receptors by ligands induces significant molecular changes inside the cell, such as an inhibition of adenylate cyclase activity, activation of potassium channels and reductions of calcium conductance. Recent data indicate that other signalling pathways also may be involved in morphine activity. Among these are phospholipase C, mitogen-activated kinases (MAP kinases) or ß-arrestin. The present review focuses on major mechanisms which currently are considered as essential in morphine activity and dependence and may be important for further studies.


Subject(s)
Adenylyl Cyclases/metabolism , Morphine Dependence/metabolism , Adenylyl Cyclases/genetics , Animals , Humans , Morphine Dependence/genetics , Receptors, Opioid/genetics , Receptors, Opioid/metabolism , beta-Arrestins/metabolism
4.
Behav Brain Res ; 437: 114103, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36089098

ABSTRACT

Mephedrone, a popular psychostimulating substance widely used illegally in recreational purposes, exerts in rodents that regularly and intermittently were exposed to it a sensitized response to the drug. Behavioral sensitization is one of experimental models of drug dependency/abuse liability. In the present study we evaluated a potential involvement of the L-arginine-NO-cGMP pathway in the development of sensitization to the mephedrone-induced hyperlocomotion. Locomotor activity was measured automatically and experiments were performed on male Albino Swiss mice. We demonstrated that a 5-day administration of 7-nitroindazole (10 or 20 mg/kg/day) and L-NAME (50 mg/kg/day) suppressed the development of sensitization to the mephedrone-induced hyperlocomotion. As for L-arginine (125 or 250 mg/kg/day) and methylene blue (5 or 10 mg/kg/day) the obtained outcomes are inconclusive. Furthermore, the lower dose of L-NAME (25 mg/kg/day) surprisingly potentiated the development of sensitization to the mephedrone-induced effects on the spontaneous locomotor activity in mice. In conclusion, our data demonstrated that modulators of the L-arginine-NO-cGMP pathway may differently affect the development of sensitization to the locomotor stimulant effects of mephedrone. Inhibition of neuronal nitric oxide synthase (NOS) seems to prevent this process quite profoundly, non-selective inhibition of NOS may have a dual effect, whereas inhibition of soluble guanylate cyclase may only partially suppress the development of sensitization to the mephedrone-induced effects.


Subject(s)
Cyclic GMP , Nitric Oxide , Animals , Mice , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Cyclic GMP/metabolism , Arginine/pharmacology , Arginine/metabolism , Locomotion , Dose-Response Relationship, Drug
5.
Brain Sci ; 12(2)2022 Jan 30.
Article in English | MEDLINE | ID: mdl-35203952

ABSTRACT

Mephedrone belongs to the "party drugs" thanks to its psychostimulant effects, similar to the ones observed after amphetamines. Though mephedrone is used worldwide by humans and in laboratory animals, not all properties of this drug have been discovered yet. Therefore, the main aim of this study was to expand the knowledge about mephedrone's activity in living organisms. A set of behavioral tests (i.e., measurement of the spontaneous locomotor activity, rotarod, chimney, elevated plus maze with its modification, novel object recognition, and pentylenetetrazol seizure tests) were carried out in male albino Swiss mice. Different dose ranges of mephedrone (0.05-5 mg/kg) were administered. We demonstrated that mephedrone at a dose of 5 mg/kg rapidly increased the spontaneous locomotor activity of the tested mice and its repeated administration led to the development of tolerance to these effects. Mephedrone showed the anxiolytic-like potential and improved spatial memory, but it did not affect recognition memory. Moreover, the drug seemed not to have any anticonvulsant or proconvulsant activity. In conclusion, mephedrone induces many central effects. It easily crosses the blood-brain barrier and peaks in the brain quickly after exposure. Our experiment on inducing a hyperlocomotion effect showed that mephedrone's effects are transient and lasted for a relatively short time.

6.
Mol Neurobiol ; 58(9): 4413-4424, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34021482

ABSTRACT

Mephedrone is a widely used drug of abuse, exerting its effects by interacting with monoamine transporters. Although this mechanism has been widely studied heretofore, little is known about the involvement of glutamatergic transmission in mephedrone effects. In this study, we comprehensively evaluated glutamatergic involvement in rewarding effects of mephedrone using an interdisciplinary approach including (1) behavioural study on effects of memantine (non-selective NMDA antagonist) on expression of mephedrone-induced conditioned place preference (CPP) in rats; (2) evaluation of glutamate concentrations in the hippocampus of rats following 6 days of mephedrone administration, using in vivo magnetic resonance spectroscopy (MRS); and (3) determination of glutamate levels in the hippocampus of rats treated with mephedrone and subjected to MRS, using ion-exchange chromatography. In the presented research, we confirmed priorly reported mephedrone-induced rewarding effects in the CPP paradigm and showed that memantine (5 mg/kg) was able to reverse the expression of this effect. MRS study showed that subchronic mephedrone administration increased glutamate level in the hippocampus when measured in vivo 24 h (5 mg/kg, 10 mg/kg and 20 mg/kg) and 2 weeks (5 mg/kg and 20 mg/kg) after last injection. Ex vivo chromatographic analysis did not show significant changes in hippocampal glutamate concentrations; however, it showed similar results as obtained in the MRS study proving its validity. Taken together, the presented study provides new insight into glutamatergic involvement in rewarding properties of mephedrone.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/drug effects , Methamphetamine/analogs & derivatives , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Reward , Animals , Hippocampus/metabolism , Magnetic Resonance Spectroscopy , Male , Methamphetamine/pharmacology , Motor Activity/drug effects , Rats , Rats, Wistar
7.
Curr Med Chem ; 28(20): 4004-4022, 2021.
Article in English | MEDLINE | ID: mdl-33231144

ABSTRACT

Obesity is a major health concern for a growing fraction of the population, as its prevalence and related metabolic disorders are not fully understood. Over the last decade, many attempts have been undertaken to understand the mechanisms at the basis of this condition, in which the accumulation of fat occurring in adipose tissue leads to the pathogenesis of obesity- related disorders. Among the most recent studies, those on Peroxisome Proliferator Activated Receptors (PPARs) revealed that these nuclear receptor proteins acting as transcription factors, among others, regulate the expression of genes involved in energy, lipid, and glucose metabolisms, and chronic inflammation. The three different isotypes of PPARs, with different tissue expression and ligand binding specificity, exert similar or overlapping functions directly or indirectly linked to obesity. In this study, we reviewed the available scientific reports concerning the PPARs structure and functions, especially in obesity, considering both natural and synthetic ligands and their role in the therapy of obesity and obesity-associated disorders. On the whole, the collected data suggest that there are both natural and synthetic compounds that show beneficial and promising activity as PPAR agonists in chronic diseases related to obesity.


Subject(s)
Obesity , Peroxisome Proliferator-Activated Receptors , Humans , Ligands , Obesity/drug therapy , Receptors, Cytoplasmic and Nuclear , Transcription Factors
8.
Neurochem Int ; 133: 104616, 2020 02.
Article in English | MEDLINE | ID: mdl-31809774

ABSTRACT

Linagliptin is a selective dipeptidyl peptidase-4 (DPP-4) inhibitor which suppresses the rapid degradation of endogenous glucagon-like peptide-1 (GLP-1). In clinical practice, it is used as an antidiabetic drug, but recent studies have confirmed its role in the activity of the central nervous system (CNS). The reported study focused on the role of linagliptin (10 and 20 mg/kg, ip) in the morphine rewarding effect, analyzing how the agent had influenced the conditioned place preference (CPP) in rats via the expression, acquisition, extinction and reinstatement of the morphine rewarding effect. The obtained results clearly demonstrated linagliptin to inhibit the expression and acquisition, to accelerate the extinction and, eventually, to reduce the reinstatement of morphine-induced CPP. The undertaken experiments significantly extended our knowledge on the mechanisms behind the morphine rewarding effect.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Linagliptin/pharmacology , Morphine/pharmacology , Reward , Animals , Behavior, Animal/drug effects , Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl Peptidase 4/drug effects , Glucagon-Like Peptide 1/metabolism , Hypoglycemic Agents/pharmacology , Male , Rats, Wistar
9.
Neurotox Res ; 37(4): 1036-1046, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31792805

ABSTRACT

This study investigated the influence of sildenafil and methylene blue (MB), two modulators of the nitric oxide (NO)-cyclic guanosine-3',5'-monophosphate (cGMP) pathway on amnesic effects of two benzodiazepines (BZs) (diazepam (DZ) and flunitrazepam (FNZ)), in rodents-mice and rats. In the modified elevated plus maze (mEPM) and novel object recognition (NOR) tests, MB given ip at a dose of 5 mg/kg 5 min prior to DZ administration (0.25 or 1 mg/kg, sc) enhanced/induced memory impairment caused by DZ. When MB (2.5, 5, and 10 mg/kg) was applied 5 min prior to FNZ administration (0.05 and 0.1 mg/kg), an effect was opposite and memory impairment induced by FNZ was reduced. When sildenafil (2.5 and 5 mg/kg, ip) was applied 5 min prior to DZ, we observed a reduction of DZ-induced memory deficiency in the mEPM test. A similar effect of sildenafil was shown in the NOR test when the drug was applied at doses of 1.25, 2.5, and 5 mg/kg prior to DZ. In the mEPM test, sildenafil at abovementioned doses had no effects on FNZ-induced memory impairment. In turns, sildenafil administered at doses of 2.5 and 5 mg/kg increased the effect of FNZ on memory impairment in the NOR test. In conclusion, the NO-cGMP pathway is involved differentially into BZs-induced spatial and recognition memory impairments assessed using the NOR and mEPM tests. Modulators of the NO-cGMP pathway affect animal behavior in these tests in a different way depending on what benzodiazepine is applied.


Subject(s)
Cyclic GMP/metabolism , Diazepam/toxicity , Flunitrazepam/toxicity , Nitric Oxide/metabolism , Recognition, Psychology/drug effects , Spatial Memory/drug effects , Animals , Dose-Response Relationship, Drug , GABA Modulators/toxicity , Male , Maze Learning/drug effects , Maze Learning/physiology , Rats , Rats, Wistar , Recognition, Psychology/physiology , Rodentia , Signal Transduction/drug effects , Signal Transduction/physiology , Spatial Memory/physiology
10.
Brain Res ; 1719: 49-56, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31121160

ABSTRACT

Morphine is one of the most potent analgesics used in medicine and it's long-term use is associated with the risk of the state of dependence. The cessation of chronic morphine administration leads to withdrawal signs which are associated with neurotransmitter dysregulations within mesolimbic system. Adenosine 5'-triphosphate (ATP) and purinergic system play an important role in the activity of central nervous system (CNS). Purinergic receptors are widely distributed in neurons and glial cells throughout the CNS taking part in integration of functional activity between neurons, glial and vascular cells. In the present study the mRNA and protein expression of purinergic P2X4 and P2X7 receptors in selected mesolimbic structures (striatum, hippocampus and prefrontal cortex) during morphine withdrawal in rats was investigated by RT-PCR and Western Blot analysis. Two experimental models of morphine withdrawal were studied: single and repeated morphine withdrawal. We demonstrated that expression of P2X4 and P2X7 receptors was altered during morphine withdrawal period in rats. These alterations were varied in particular mesolimbic areas depending on the scheme of morphine administration. Our results extend the current knowledge on morphine withdrawal and for the first time high-light interactions between purinergic system and morphine withdrawal. It seems, the purinergic system may be a new, valuable tool in searching for a new strategy of management of opioid dependence.


Subject(s)
Receptors, Purinergic P2X4/genetics , Receptors, Purinergic P2X7/genetics , Substance Withdrawal Syndrome/metabolism , Adenosine Triphosphate/metabolism , Animals , Brain/metabolism , Corpus Striatum/metabolism , Gene Expression/genetics , Gene Expression Regulation/drug effects , Hippocampus/metabolism , Male , Morphine/metabolism , Morphine Dependence/genetics , Morphine Dependence/physiopathology , Neuroglia/metabolism , Neurons/metabolism , Prefrontal Cortex/metabolism , RNA, Messenger , Rats , Rats, Wistar , Receptors, Purinergic P2X4/metabolism , Receptors, Purinergic P2X7/metabolism , Substance Withdrawal Syndrome/genetics
11.
Eur J Pharmacol ; 858: 172512, 2019 Sep 05.
Article in English | MEDLINE | ID: mdl-31260653

ABSTRACT

The present study was conducted to evaluate the influence of AMN082, the metabotropic glutamate receptor subtype 7 (mGlu7) allosteric agonist on different stages of memory processes connected with fear conditioning in the passive avoidance (PA) learning task in mice and negative emotional state (anxiety-like) induced by ethanol- and morphine-withdrawal in the elevated plus maze (EPM) test in rats. To perform the PA test, AMN082 (1.25, 2.5 and 5 mg/kg, i. p.) was injected to interfere with (or inhibit) acquisition, consolidation, and retrieval processes. The retention latency in each group was recorded using a step-through passive avoidance task 24 h after training. In turn, in ethanol- and morphine-withdrawal rats, the influence of AMN082 on anxiety-like behavior was estimated in the EPM test 24 h- (ethanol) and 72- h (morphine) after the last dose of repeated drug administrations. In all experimental groups, AMN082 at the dose of 5 mg/kg significantly decreased the step-through latency of long-term memory in the PA task. These AMN082 effects were reversed by MMPIP (10 mg/kg), the antagonist of mGlu7 receptor. AMN082 (2.5 and 5 mg/kg) also decreased ethanol- and morphine withdrawal-induced anxiety-like behavior in the EPM test, and this AMN082 (5 mg/kg) effect was counteracted by MMPIP pretreatment. Taken together, the results show that mGlu7 is involved in fear learning to the context and anxiety-like state connected with unpleasant experiences after ethanol- and morphine withdrawal in rodents. However, it appears that functional dissociation exists between these two AMN082 effects.


Subject(s)
Anxiety/drug therapy , Behavior, Animal/drug effects , Benzhydryl Compounds/pharmacology , Fear/drug effects , Fear/physiology , Memory/drug effects , Receptors, Metabotropic Glutamate/agonists , Allosteric Regulation/drug effects , Animals , Anxiety/physiopathology , Anxiety/psychology , Benzhydryl Compounds/therapeutic use , Dose-Response Relationship, Drug , Male , Memory Consolidation/drug effects , Mice , Rats , Substance Withdrawal Syndrome/drug therapy
12.
Food Chem Toxicol ; 134: 110822, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31536753

ABSTRACT

Phosphodiesterases (PDEs) consisted of 11 subtypes (PDE1 to PDE11) and over 40 isoforms that regulate levels of cyclic guanosine monophosphate (cGMP) and cyclic adenosine monophosphate (cAMP), the second messengers in cell functions. PDE inhibitors (PDEIs) have been attractive therapeutic targets due to their involvement in diverse medical conditions, e.g. cardiovascular diseases, autoimmune diseases, Alzheimer's disease (AD), etc. Among them; AD with a complex pathology is a progressive neurodegenerative disorder which affect mostly senile people in the world and only symptomatic treatment particularly using cholinesterase inhibitors in clinic is available at the moment for AD. Consequently, novel treatment strategies towards AD are still searched extensively. Since PDEs are broadly expressed in the brain, PDEIs are considered to modulate neurodegenerative conditions through regulating cAMP and cGMP in the brain. In this sense, several synthetic or natural molecules inhibiting various PDE subtypes such as rolipram and roflumilast (PDE4 inhibitors), vinpocetine (PDE1 inhibitor), cilostazol and milrinone (PDE3 inhibitors), sildenafil and tadalafil (PDE5 inhibitors), etc have been reported showing encouraging results for the treatment of AD. In this review, PDE superfamily will be scrutinized from the view point of structural features, isoforms, functions and pharmacology particularly attributed to PDEs as target for AD therapy.


Subject(s)
Alzheimer Disease/drug therapy , Phosphodiesterase Inhibitors/pharmacology , Animals , Humans , Phosphodiesterase Inhibitors/therapeutic use
13.
Eur J Pharmacol ; 588(1): 72-7, 2008 Jun 24.
Article in English | MEDLINE | ID: mdl-18466897

ABSTRACT

In the present study, the effects of adenosine agonists on the development of sensitization to withdrawal signs precipitated after sporadic treatment with diazepam, in mice, were investigated. To obtain the sensitization, the animals were divided into groups: continuously and sporadically treated with diazepam (15.0 mg/kg, s.c.). The adenosine receptor agonists (CPA, CGS 21,680 and NECA) were administered in sporadically diazepam treated mice during two diazepam-free periods. Concomitant administration of pentetrazole (55.0 mg/kg, s.c.) with flumazenil (5.0 mg/kg, i.p.) after the last injection of diazepam or vehicle, induced the withdrawal signs, such as clonic seizures, tonic convulsion and death episodes. The major finding of our experiments is attenuation of withdrawal signs in sensitized mice, inducing by all adenosine agonists. Only higher dose of CPA produced significantly decreased the number of withdrawal incidents, while both used doses of CGS 21,680 and NECA produced more clear effects. These results support the hypothesis that adenosinergic system is involved in the mechanisms of sensitization to the benzodiazepine withdrawal signs, and adenosine A(2A) receptors play more important role in that process.


Subject(s)
Diazepam/adverse effects , Hypnotics and Sedatives/adverse effects , Purinergic P1 Receptor Agonists , Substance Withdrawal Syndrome/prevention & control , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A1 Receptor Agonists , Adenosine A2 Receptor Agonists , Adenosine-5'-(N-ethylcarboxamide)/pharmacology , Animals , Convulsants , Male , Mice , Pentylenetetrazole , Phenethylamines/pharmacology , Seizures/chemically induced , Seizures/etiology , Seizures/physiopathology , Substance Withdrawal Syndrome/psychology , Substance-Related Disorders/psychology
14.
Pharmacol Rep ; 60(5): 679-85, 2008.
Article in English | MEDLINE | ID: mdl-19066414

ABSTRACT

In the present study, the involvement of the selective adenosine A1 (CPA) and A2A (CGS 21680) and non-selective adenosine A1/A2A (NECA) receptor agonists on the development of hypersensitivity to acute morphine injection given during opiate withdrawal was investigated. Intraperitioneal (ip) injections of morphine at increasing doses (10, 20, 30, 40, 50 mg/kg) for 6 consecutive days produced a state of dependence. On the 6th day, in the morning, animals were injected with the last dose of morphine (50 mg/kg, ip). Each day, 20 min before each injection of morphine, adenosine receptor agonists were also administered. Seven days after cessation of the morphine treatment, on the 13th day of the experiment, all animals were challenged with a dose of morphine (10 mg/kg, ip). A clear increase in locomotor activity was observed, indicating that hypersensitivity had developed. Our study has demonstrated the presence of an attenuating effect of adenosinergic drugs, such as CGS 21680 and NECA, but not CPA, on the development of hypersensitivity. The results indicate that stimulation of the adenosine A2A receptor plays some role in modulating the neuroadaptive changes appearing during chronic opioid treatment and that adenosine A2A receptor agonists may serve as useful drugs in relapse protection. Our investigations focused on adenosine A2A agonists as possible vehicles for pharmacotherapy for morphine addiction.


Subject(s)
Analgesics, Opioid/pharmacology , Morphine/pharmacology , Purinergic P1 Receptor Agonists , Substance Withdrawal Syndrome/prevention & control , Substance Withdrawal Syndrome/psychology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine-5'-(N-ethylcarboxamide)/pharmacology , Animals , Male , Mice , Motor Activity/drug effects , Opioid-Related Disorders/psychology , Phenethylamines/pharmacology
15.
Pharmacol Rep ; 60(4): 475-82, 2008.
Article in English | MEDLINE | ID: mdl-18799815

ABSTRACT

Chronic treatment with the benzodiazepines is well known to produce tolerance, which has been extensively documented to be attributed to modifications in the gamma-aminobutyric acid (GABA)ergic neurotransmission. However, literature data have also suggested the participation of different neurotransmitter systems, including glutamatergic, in benzodiazepine tolerance. The purpose of the present study was to determine the role of nitric oxide (NO) in the development of tolerance to the motor dysfunction induced by chronic administration of diazepam. The motor performance was assessed on the 1st and 10th day of experiment, using the rotarod and chimney tests in mice. Treatment of animals with both non-selective NO synthase (NOS) inhibitors: N(G)-nitro-L-arginine methyl ester (L-NAME), N(G)-nitro-L-arginine (L-NOARG) and selective NOS inhibitor: 7-nitroindazole was able to prevent the development of tolerance to the motor impairing effect of diazepam. Moreover, administration of L-arginine, a NO precursor, facilitated the development of diazepam-induced tolerance in rotarod test. These findings suggest that NO may be involved, at least in part, in the tolerance to the motor dysfunction, developed during the chronic administration of diazepam in mice.


Subject(s)
Anticonvulsants/toxicity , Diazepam/toxicity , Free Radical Scavengers/pharmacology , Motor Activity/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide/physiology , Nitroarginine/pharmacology , Animals , Anticonvulsants/antagonists & inhibitors , Diazepam/antagonists & inhibitors , Drug Tolerance , Enzyme Inhibitors/pharmacology , Male , Mice , Nitric Oxide/biosynthesis , Rotarod Performance Test
16.
Neurotox Res ; 33(2): 422-432, 2018 02.
Article in English | MEDLINE | ID: mdl-28936791

ABSTRACT

The goal of the present study was to examine the effects of N-methyl-aspartate (NMDA) receptor antagonists-memantine and ketamine and the drugs modifying the NO:cGMP pathway-NG-nitro-L-arginine methyl ester (L-NAME) and 7-nitroindazole (7-NI), the endogenous precursor of NO-L-arginine, and the guanylyl cyclase inhibitor-methylene blue (MB) on the development of sensitization to withdrawal signs precipitated after chronic, interrupted treatment with diazepam, a benzodiazepine receptor agonist, in mice. To develop the sensitization, the mice were divided into groups: continuously and sporadically (with two diazepam-free periods) treated with diazepam (15 mg/kg, sc). To precipitate the withdrawal syndrome (clonic and tonic seizures, and death), pentylenetetrazole (55 mg/kg, sc) with the benzodiazepine receptor antagonist, flumazenil (5.0 mg/kg, ip), were administered after the last injection of diazepam or saline. Memantine (2.5, 5.0 mg/kg), and ketamine (2.5, 5.0 mg/kg), L-NAME (100, 200 mg/kg) and 7-NI (20 and 40 mg/kg), L-arginine (250, 500 mg/kg) and MB (5 and 10 mg/kg) were administered ip in sporadically diazepam-treated mice during the diazepam-free periods. Our results indicated that both NMDA receptor antagonists and drugs that inhibit the NO:cGMP pathway, except L-arginine (the endogenous donor of NO), attenuated the diazepam-induced sensitization to withdrawal signs in mice. Thus, NMDA receptors and the NO:cGMP pathway are involved in the mechanisms of sensitization to benzodiazepine withdrawal.


Subject(s)
Diazepam/pharmacology , Pentylenetetrazole/pharmacology , Receptors, N-Methyl-D-Aspartate/drug effects , Signal Transduction/drug effects , Substance Withdrawal Syndrome/physiopathology , Animals , Benzodiazepines/pharmacology , Cyclic GMP/metabolism , Male , Mice , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
17.
Mol Neurobiol ; 55(11): 8473-8485, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29557083

ABSTRACT

The present study focused upon the role of SB-334867, an orexin-1 receptor antagonist, in the acquisition of morphine-induced sensitization to locomotor activity in mice. Behavioral sensitization is an enhanced systemic reaction to the same dose of an addictive substance, which assumingly increases both the desire for the drug and the risk of relapse to addiction. Morphine-induced sensitization in mice was achieved by sporadic doses (five injections every 3 days) of morphine (10 mg/kg, i.p.), while a challenge dose of morphine (10 mg/kg) was injected 7 days later. In order to assess the impact of orexin system blockade on the acquisition of sensitization, SB-334867 was administered before each morphine injection, except the morphine challenge dose. The locomotor activity test was performed on each day of morphine administration. Brain structures (striatum, hippocampus, and prefrontal cortex) were collected after behavioral tests for molecular experiments in which mRNA expression of orexin, dopamine, and adenosine receptors was explored by the qRT-PCR technique. Additionally, the mRNA expression of markers, such as GFAP and Iba-1, was also analyzed by the same technique. SB-334867 inhibited the acquisition of morphine-induced sensitization to locomotor activity of mice. Significant alterations were observed in mRNA expression of orexin, dopamine, and adenosine receptors and in the expression of GFAP and Iba-1, showing a broad range of interactions in the mesolimbic system among orexin, dopamine, adenosine, and glial cells during behavioral sensitization. Summing up, the orexin system may be an effective measure to inhibit morphine-induced behavioral sensitization.


Subject(s)
Benzoxazoles/pharmacology , Morphine/pharmacology , Orexin Receptor Antagonists/pharmacology , Orexin Receptors/metabolism , Urea/analogs & derivatives , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Brain/pathology , Male , Mice , Microglia/drug effects , Microglia/metabolism , Motor Activity/drug effects , Naphthyridines , Orexin Receptors/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A2A/metabolism , Urea/pharmacology
18.
Behav Brain Res ; 338: 9-16, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29030082

ABSTRACT

Repeated exposure to and withdrawal from ethanol induces deficits in spatial reversal learning. Data indicate that metabotropic glutamate 5 (mGlu5) receptors are implicated in synaptic plasticity and learning and memory. These receptors functionally interact with N-methyl-d-aspartate (NMDA) receptors, and activation of one type results in the activation of the other. We examined whether (S)-(4-fluorophenyl)(3-(3-(4-fluorophenyl)-1,2,4-oxadiazol-5-yl)-piperidin-1-yl (ADX-47273), a positive allosteric modulator (PAM) of mGlu5 receptor, attenuates deficits in reversal learning induced by withdrawal (11-13days) from 'binge-like' ethanol input (5.0g/kg, i.g. for 5days) in the Barnes maze (a spatial learning) task in rats. We additionally examined the effects of ADX-47273 on the expression of the NMDA receptors subunit, GluN2B, in the hippocampus and prefrontal cortex, on the 13th day of ethanol withdrawal. Herein, withdrawal from repeated ethanol administration impaired reversal learning, but not the probe trial. Moreover, ADX-47273 (30mg/kg, i.p.) given prior to the first reversal learning trial for 3days in the Barnes maze, significantly enhanced performance in the ethanol-treated group. The 13th day of ethanol abstinence decreased the expression of the GluN2B subunit in the selected brain regions, but ADX-47273 administration increased it. In conclusion, positive allosteric modulation of mGlu5 receptors recovered spatial reversal learning impairment induced by withdrawal from 'binge-like' ethanol exposure. Such effect seems to be correlated with the mGlu5 receptors mediated potentiation of GluN2B-NMDA receptor mediated responses in the hippocampus and prefrontal cortex. Thus, our results emphasize the role of mGlu5 receptor PAM in the adaptive learning impaired by ethanol exposure.


Subject(s)
Cognition/drug effects , Ethanol/administration & dosage , Oxadiazoles/pharmacology , Piperidines/pharmacology , Receptor, Metabotropic Glutamate 5/metabolism , Reversal Learning/drug effects , Spatial Learning/drug effects , Substance Withdrawal Syndrome/psychology , Allosteric Regulation/drug effects , Animals , Male , Motor Activity/drug effects , Rats , Rats, Wistar , Substance Withdrawal Syndrome/metabolism
19.
Biofactors ; 43(6): 760-769, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28817221

ABSTRACT

The incidence of neurological disorders is growing in developed countries together with increased lifespan. Nowadays, there are still no effective treatments for neurodegenerative pathologies, which make necessary to search for new therapeutic agents. Natural products, most of them used in traditional medicine, are considered promising alternatives for the treatment of neurodegenerative diseases. Honokiol is a natural bioactive phenylpropanoid compound, belonging to the class of neolignan, found in notable amounts in the bark of Magnolia tree, and has been reported to exert diverse pharmacological properties including neuroprotective activities. Honokiol can permeate the blood brain barrier and the blood-cerebrospinal fluid to increase its bioavailability in neurological tissues. Diverse studies have provided evidence on the neuroprotective effect of honokiol in the central nervous system, due to its potent antioxidant activity, and amelioration of the excitotoxicity mainly related to the blockade of glutamate receptors and reduction in neuroinflammation. In addition, recent studies suggest that honokiol can attenuate neurotoxicity exerted by abnormally aggregated Aß in Alzheimer's disease. The present work summarizes what is currently known concerning the neuroprotective effects of honokiol and its potential molecular mechanisms of action, which make it considered as a promising agent in the treatment and management of neurodegenerative diseases. © 2017 BioFactors, 43(6):760-769, 2017.


Subject(s)
Antioxidants/pharmacology , Biphenyl Compounds/pharmacology , Central Nervous System/drug effects , Lignans/pharmacology , Magnolia/chemistry , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/pharmacology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/immunology , Antioxidants/isolation & purification , Antioxidants/pharmacokinetics , Biological Products/chemistry , Biological Transport , Biphenyl Compounds/isolation & purification , Biphenyl Compounds/pharmacokinetics , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , Central Nervous System/immunology , Central Nervous System/pathology , Cytokines/antagonists & inhibitors , Cytokines/genetics , Cytokines/immunology , Gene Expression Regulation , Humans , Lignans/isolation & purification , Lignans/pharmacokinetics , Nerve Growth Factors/antagonists & inhibitors , Nerve Growth Factors/genetics , Nerve Growth Factors/immunology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/pathology , Neuroprotective Agents/isolation & purification , Neuroprotective Agents/pharmacokinetics , Plant Bark/chemistry , Receptors, Glutamate/genetics , Receptors, Glutamate/immunology
20.
Neurotox Res ; 31(2): 309-316, 2017 02.
Article in English | MEDLINE | ID: mdl-27957675

ABSTRACT

The goal of the study was to investigate the effects of drugs modifying L-arginine:NO:cGMP pathway on the development of tolerance to flunitrazepam (FNZ)-induced motor impairment in mice. FNZ-induced motor incoordination was assessed on the 1st and 8th days of experiment, using the rotarod and chimney tests. It was found that (a) both a non-selective nitric oxide synthase (NOS) inhibitor: N G-nitro-L-arginine methyl ester (L-NAME) and an unselective neuronal NOS inhibitor: 7-nitroindazole (7-NI) inhibited the development of tolerance to the motor-impairing effects of FNZ in the rotarod and the chimney tests and (b) both a NO precursor: L-arginine and a selective inhibitor of phosphodiesterase 5 (PDE5): sildenafil did not affect the development of tolerance to FNZ-induced motor impairment in mice. Those findings provided behavioural evidence that NO could contribute an important role in the development of tolerance to FNZ in mice.


Subject(s)
Arginine/pharmacology , Drug Tolerance/physiology , Flunitrazepam/pharmacology , Nitric Oxide/physiology , Signal Transduction/physiology , Animals , Cyclic GMP/physiology , Drug Interactions , Indazoles/pharmacology , Male , Mice , Motor Skills/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Signal Transduction/drug effects , Sildenafil Citrate/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL