Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Int J Mol Sci ; 23(2)2022 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-35054944

RESUMO

Drug addiction causes constant serious health, social, and economic burden within the human society. The current drug dependence pharmacotherapies, particularly relapse prevention, remain limited, unsatisfactory, unreliable for opioids and tobacco, and even symptomatic for stimulants and cannabinoids, thus, new more effective treatment strategies are researched. The antagonism of the growth hormone secretagogue receptor type A (GHS-R1A) has been recently proposed as a novel alcohol addiction treatment strategy, and it has been intensively studied in experimental models of other addictive drugs, such as nicotine, stimulants, opioids and cannabinoids. The role of ghrelin signaling in these drugs effects has also been investigated. The present review aims to provide a comprehensive overview of preclinical and clinical studies focused on ghrelin's/GHS-R1A possible involvement in these nonalcohol addictive drugs reinforcing effects and addiction. Although the investigation is still in its early stage, majority of the existing reviewed experimental results from rodents with the addition of few human studies, that searched correlations between the genetic variations of the ghrelin signaling or the ghrelin blood content with the addictive drugs effects, have indicated the importance of the ghrelin's/GHS-R1As involvement in the nonalcohol abused drugs pro-addictive effects. Further research is necessary to elucidate the exact involved mechanisms and to verify the future potential utilization and safety of the GHS-R1A antagonism use for these drug addiction therapies, particularly for reducing the risk of relapse.


Assuntos
Grelina/metabolismo , Receptores de Grelina/metabolismo , Transdução de Sinais , Transtornos Relacionados ao Uso de Substâncias/etiologia , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Animais , Biomarcadores , Estimulantes do Sistema Nervoso Central/efeitos adversos , Estudos Clínicos como Assunto , Modelos Animais de Doenças , Suscetibilidade a Doenças , Predisposição Genética para Doença , Humanos , Nicotina/efeitos adversos , Reforço Psicológico , Uso de Tabaco/efeitos adversos
2.
Int J Mol Sci ; 22(5)2021 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-33673659

RESUMO

Cannabis/cannabinoids are widely used for recreational and therapy purposes, but their risks are largely disregarded. However, cannabinoid-associated use disorders and dependence are alarmingly increasing and an effective treatment is lacking. Recently, the growth hormone secretagogue receptor (GHSR1A) antagonism was proposed as a promising mechanism for drug addiction therapy. However, the role of GHS-R1A and its endogenous ligand ghrelin in cannabinoid abuse remains unclear. Therefore, the aim of our study was to investigate whether the GHS-R1A antagonist JMV2959 could reduce the tetrahydrocannabinol (THC)-induced conditioned place preference (CPP) and behavioral stimulation, the WIN55,212-2 intravenous self-administration (IVSA), and the tendency to relapse. Following an ongoing WIN55,212-2 self-administration, JMV2959 3 mg/kg was administered intraperitoneally 20 min before three consequent daily 120-min IVSA sessions under a fixed ratio FR1, which significantly reduced the number of the active lever-pressing, the number of infusions, and the cannabinoid intake. Pretreatment with JMV2959 suggested reduction of the WIN55,212-2-seeking/relapse-like behavior tested in rats on the twelfth day of the forced abstinence period. On the contrary, pretreatment with ghrelin significantly increased the cannabinoid IVSA as well as enhanced the relapse-like behavior. Co-administration of ghrelin with JMV2959 abolished/reduced the significant efficacy of the GHS-R1A antagonist in the cannabinoid IVSA. Pretreatment with JMV2959 significantly and dose-dependently reduced the manifestation of THC-induced CPP. The THC-CPP development was reduced after the simultaneous administration of JMV2959 with THC during conditioning. JMV2959 also significantly reduced the THC-induced behavioral stimulation in the LABORAS cage. Our findings suggest that GHS-R1A importantly participates in the rewarding/reinforcing effects of cannabinoids.


Assuntos
Comportamento Animal/efeitos dos fármacos , Canabinoides/farmacologia , Condicionamento Operante/efeitos dos fármacos , Condicionamento Psicológico/efeitos dos fármacos , Glicina/análogos & derivados , Receptores de Grelina/antagonistas & inibidores , Triazóis/farmacologia , Administração Intravenosa , Animais , Canabinoides/administração & dosagem , Glicina/farmacologia , Masculino , Ratos , Ratos Wistar , Reforço Psicológico , Autoadministração
3.
Addict Biol ; 25(6): e12845, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-31696597

RESUMO

The extended occurrence of fentanils abuse associated with the dramatic increase in opioid fatal overdoses and dependence strongly emphasizes insufficiencies in opioid addiction treatment. Recently, the growth hormone secretagogue receptor (GHS-R1A) antagonism was proposed as a promising mechanism for drug addiction therapy. However, the role of GHS-R1A and its endogenous ligand ghrelin in opioid abuse is still unclear. Therefore, the aim of our study was to clarify whether the GHS-R1A antagonist JMV2959 could reduce the fentanyl-induced conditioned place preference (CPP), the fentanyl intravenous self-administration (IVSA), and the tendency to relapse, but also whether JMV2959 could significantly influence the fentanyl-induced dopamine efflux in the nucleus accumbens (NAC) in rats, that importantly participates in opioids' reinforcing effects. Following an ongoing fentanyl self-administration, JMV2959 3 mg/kg was administered intraperitoneally 20 minutes before three consequent daily 360-minute IVSA sessions under a fixed ratio FR1, which significantly reduced the number of active lever-pressing, the number of infusions, and the fentanyl intake. Pretreatment with JMV2959 also reduced the fentanyl-seeking/relapse-like behaviour tested in rats on the 12th day of the forced abstinence period. Pretreatment with JMV2959 significantly and dose-dependently reduced the manifestation of fentanyl-CPP. The fentanyl-CPP development was reduced after the simultaneous administration of JMV2959 with fentanyl during conditioning. The JMV2959 significantly reduced the accumbens dopamine release induced by subcutaneous and intravenous fentanyl. Simultaneously, it affected the concentration of byproducts associated with dopamine metabolism in the NAC. Our findings suggest that GHS-R1A importantly participates in the rewarding/reinforcing effects of fentanyl.


Assuntos
Condicionamento Operante/efeitos dos fármacos , Dopamina/metabolismo , Fentanila/efeitos adversos , Glicina/análogos & derivados , Entorpecentes/efeitos adversos , Núcleo Accumbens/efeitos dos fármacos , Receptores de Grelina/antagonistas & inibidores , Triazóis/farmacologia , Administração Intravenosa , Animais , Fentanila/administração & dosagem , Grelina/metabolismo , Glicina/farmacologia , Masculino , Entorpecentes/administração & dosagem , Ratos , Ratos Wistar , Autoadministração
4.
Int J Mol Sci ; 22(1)2020 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-33379212

RESUMO

The endocannabinoid/CB1R system as well as the central ghrelin signalling with its growth hormone secretagogoue receptors (GHS-R1A) are importantly involved in food intake and reward/reinforcement processing and show distinct overlaps in distribution within the relevant brain regions including the hypothalamus (food intake), the ventral tegmental area (VTA) and the nucleus accumbens (NAC) (reward/reinforcement). The significant mutual interaction between these systems in food intake has been documented; however, the possible role of ghrelin/GHS-R1A in the cannabinoid reinforcement effects and addiction remain unclear. Therefore, the principal aim of the present study was to investigate whether pretreatment with GHS-R1A antagonist/JMV2959 could reduce the CB1R agonist/WIN55,212-2-induced dopamine efflux in the nucleus accumbens shell (NACSh), which is considered a crucial trigger impulse of the addiction process. The synthetic aminoalklylindol cannabinoid WIN55,212-2 administration into the posterior VTA induced significant accumbens dopamine release, which was significantly reduced by the 3 mg/kg i.p. JMV2959 pretreatment. Simultaneously, the cannabinoid-increased accumbens dopamine metabolic turnover was significantly augmented by the JMV2959 pretreament. The intracerebral WIN55,212-2 administration also increased the endocannabinoid arachidonoylethanolamide/anandamide and the 2-arachidonoylglycerol/2-AG extracellular levels in the NACSh, which was moderately but significantly attenuated by the JMV2959 pretreatment. Moreover, the cannabinoid-induced decrease in accumbens γ-aminobutyric acid/gamma-aminobutyric acid levels was reversed by the JMV2959 pretreatment. The behavioural study in the LABORAS cage showed that 3 mg/kg JMV2959 pretreatment also significantly reduced the systemic WIN55,212-2-induced behavioural stimulation. Our results demonstrate that the ghrelin/GHS-R1A system significantly participates in the rewarding/reinforcing effects of the cannabinoid/CB1 agonist that are involved in cannabinoid addiction processing.


Assuntos
Benzoxazinas/administração & dosagem , Dopamina/metabolismo , Grelina/metabolismo , Glicina/análogos & derivados , Morfolinas/administração & dosagem , Naftalenos/administração & dosagem , Núcleo Accumbens/efeitos dos fármacos , Triazóis/administração & dosagem , Animais , Ácidos Araquidônicos/metabolismo , Avaliação Pré-Clínica de Medicamentos , Endocanabinoides/metabolismo , Glicerídeos/metabolismo , Glicina/administração & dosagem , Masculino , Núcleo Accumbens/metabolismo , Alcamidas Poli-Insaturadas/metabolismo , Ratos Wistar , Ácido gama-Aminobutírico/metabolismo
5.
Int J Mol Sci ; 19(10)2018 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-30261633

RESUMO

Methamphetamine abuse imposes a significant burden on individuals and society worldwide, and an effective therapy of methamphetamine addiction would provide distinguished social benefits. Ghrelin significantly participates in reinforcing neurobiological mechanisms of stimulants, including amphetamines; thus, ghrelin antagonism is proposed as a promising addiction treatment. The aim of our study was to elucidate whether the pretreatment with growth hormone secretagogue receptor (GHS-R1A) antagonist, substance JMV2959, could reduce the methamphetamine intravenous self-administration (IVSA) and the tendency to relapse, and whether JMV2959 could reduce or prevent methamphetamine-induced conditioned place preference (CPP) in rats. Following an adequate maintenance period, JMV2959 3 mg/kg was administered intraperitoneally 20 min before three consequent daily 180 min sessions of methamphetamine IVSA under a fixed ratio FR1, which significantly reduced the number of active lever-pressings, the number of infusions, and the amount of the consumed methamphetamine dose. Pretreatment with JMV2959 also reduced or prevented relapse-like behavior tested in rats on the 12th day of the abstinence period. Pretreatment with JMV2959 significantly reduced the expression of methamphetamine-induced CPP. Simultaneous administration of JMV2959 with methamphetamine during the conditioning period significantly reduced the methamphetamine-CPP. Our results encourage further research of the ghrelin antagonism as a potential new pharmacological tool for methamphetamine addiction treatment.


Assuntos
Condicionamento Psicológico/efeitos dos fármacos , Glicina/análogos & derivados , Metanfetamina/farmacologia , Receptores de Grelina/antagonistas & inibidores , Comportamento Espacial/efeitos dos fármacos , Triazóis/farmacologia , Administração Intravenosa , Análise de Variância , Animais , Peso Corporal/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/administração & dosagem , Estimulantes do Sistema Nervoso Central/farmacologia , Glicina/administração & dosagem , Glicina/farmacologia , Masculino , Metanfetamina/administração & dosagem , Ratos Wistar , Receptores de Grelina/metabolismo , Autoadministração , Fatores de Tempo , Triazóis/administração & dosagem
6.
Int J Mol Sci ; 18(11)2017 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-29165386

RESUMO

The opioid-induced rise of extracellular dopamine, endocannabinoid anandamide and γ-aminobutyric acid (GABA) concentrations triggered by opioids in the nucleus accumbens shell (NACSh) most likely participate in opioid reward. We have previously demonstrated that systemic administration of ghrelin antagonist (JMV2959) significantly decreased morphine-induced dopamine and anandamide (N-arachidonoylethanolamine, AEA) increase in the NACSh. Fentanyl is considered as a µ-receptor-selective agonist. The aim of this study was to test whether JMV2959, a growth hormone secretagogue receptor (GHS-R1A) antagonist, can influence the fentanyl-induced effects on anandamide, 2-arachidonoylglycerol (2-AG) and GABA in the NACSh and specify the involvement of GHS-R1A located in the ventral tegmental area (VTA) and nucleus accumbens (NAC). Using in vivo microdialysis in rats, we have found that pre-treatment with JMV2959 reversed dose dependently fentanyl-induced anandamide increases in the NACSh, resulting in a significant AEA decrease and intensified fentanyl-induced decreases in accumbens 2-AG levels, with both JMV2959 effects more expressed when administered into the NACSh in comparison to the VTA. JMV2959 pre-treatment significantly decreased the fentanyl-evoked accumbens GABA efflux and reduced concurrently monitored fentanyl-induced behavioural stimulation. Our current data encourage further investigation to assess if substances affecting GABA or endocannabinoid concentrations and action, such as GHS-R1A antagonists, can be used to prevent opioid-seeking behaviour.


Assuntos
Endocanabinoides/metabolismo , Fentanila/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Comportamento Animal , Espaço Extracelular/metabolismo , Grelina/farmacologia , Glicina/análogos & derivados , Glicina/farmacologia , Masculino , Ratos , Receptores de Grelina/metabolismo , Triazóis/farmacologia , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo
7.
Pharmacol Biochem Behav ; 224: 173528, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36870422

RESUMO

Central ghrelin signaling seems to play important role in addiction as well as memory processing. Antagonism of the growth hormone secretagogue receptor (GHS-R1A) has been recently proposed as a promising tool for the unsatisfactory drug addiction therapy. However, molecular aspects of GHS-R1A involvement in specific brain regions remain unclear. The present study demonstrated for the first time that acute as well as subchronic (4 days) administration of the experimental GHS-R1A antagonist JMV2959 in usual intraperitoneal doses including 3 mg/kg, had no influence on memory functions tested in the Morris Water Maze in rats as well as no significant effects on the molecular markers linked with memory processing in selected brain areas in rats, specifically on the ß-actin, c-Fos, two forms of the calcium/calmodulin-dependent protein kinase II (CaMKII, p-CaMKII) and the cAMP-response element binding protein (CREB, p-CREB), within the medial prefrontal cortex (mPFC), nucleus accumbens (NAc), dorsal striatum, and hippocampus (HIPP). Furthermore, following the methamphetamine intravenous self-administration in rats, the 3 mg/kg JMV2959 pretreatment significantly reduced or prevented the methamphetamine-induced significant decrease of hippocampal ß-actin and c-Fos as well as it prevented the significant decrease of CREB in the NAC and mPFC. These results imply, that the GHS-R1A antagonist/JMV2959 might reduce/prevent some of the memory-linked molecular changes elicited by methamphetamine addiction within brain structures associated with memory (HIPP), reward (NAc), and motivation (mPFC), which may contribute to the previously observed significant JMV2959-induced reduction of the methamphetamine self-administration and drug-seeking behavior in the same animals. Further research is necessary to corroborate these results.


Assuntos
Metanfetamina , Receptores de Grelina , Ratos , Animais , Grelina/farmacologia , Actinas , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Metanfetamina/farmacologia
8.
Neuro Endocrinol Lett ; 30(1): 79-84, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19300401

RESUMO

OBJECTIVES: Some individually-housed male mice behave aggressively during encounters with strange males, while others are timid or sociable in the same situation. The objective of the present study was to examine concentrations of glutamate, aspartate, and GABA in the brain of aggressive, timid, and sociable mice. METHODS: Random-bred albino mice were housed individually for three weeks and then classified in three groups (aggressive, timid, and sociable mice) according to their behavior during social interaction with non-aggressive group-housed male mice in a neutral cage. One week after categorization, by means of the social conflict test, levels of glutamate, aspartate, and GABA were measured by in vivo microdialysis of the medial prefrontal cortex (mPFC) of the isolated and group-housed mice. RESULTS: Sociable mice had almost triple the levels of GABA in their mPFC than aggressive or timid mice. No significant differences in aspartate and glutamate levels were found in these three types of individually-housed mice. Forebrain chemistry of group-housed mice did not differ from that of individually-housed mice with the exception of levels of glutamate and GABA which were significantly lower in group-housed mice than in sociable individually-housed mice. CONCLUSION: The present results suggest that GABA might play a role in sociable behavior. Results also corroborate other findings indicating that the GABAergic system represents an important molecular and neuronal substrate for the selective attenuation of anxiety and aggression.


Assuntos
Comportamento Animal/fisiologia , Encéfalo/metabolismo , Aminoácidos Excitatórios/metabolismo , Comportamento Social , Ácido gama-Aminobutírico/metabolismo , Agressão/fisiologia , Animais , Ácido Aspártico/análise , Ácido Aspártico/metabolismo , Aminoácidos Excitatórios/análise , Ácido Glutâmico/análise , Ácido Glutâmico/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Microquímica/métodos , Microdiálise/métodos , Modelos Biológicos , Timidez , Desejabilidade Social , Ácido gama-Aminobutírico/análise
9.
Neurochem Int ; 110: 101-113, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28958601

RESUMO

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


Assuntos
Analgésicos Opioides/administração & dosagem , Condicionamento Operante/efeitos dos fármacos , Dopamina , Morfina/administração & dosagem , Núcleo Accumbens/efeitos dos fármacos , Receptores de Grelina/antagonistas & inibidores , Analgésicos Opioides/antagonistas & inibidores , Animais , Condicionamento Operante/fisiologia , Dopamina/metabolismo , Glicina/análogos & derivados , Glicina/farmacologia , Masculino , Microdiálise/métodos , Morfina/antagonistas & inibidores , Núcleo Accumbens/metabolismo , Ratos , Ratos Wistar , Receptores de Grelina/metabolismo , Triazóis/farmacologia
10.
Psychopharmacology (Berl) ; 233(3): 469-84, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26507196

RESUMO

RATIONALE AND OBJECTIVES: In addition to dopamine, endocannabinoids are thought to participate in neural reward mechanisms of opioids. Number of recent studies suggests crucial involvement of ghrelin in some addictive drugs effects. Our previous results showed that ghrelin participates in morphine-induced changes in the mesolimbic dopaminergic system associated with reward processing. The goal of the present study was to test whether the growth hormone secretagogue receptor (GHS-R1A) antagonist JMV2959 was able to influence morphine-induced effects on anandamide (N-arachidonoylethanolamine, AEA) and 2-arachidonoylglycerol (2-AG) in the nucleus accumbens shell (NACSh). METHODS: We used in vivo microdialysis to determine changes in levels of AEA and 2-AG in the NACSh in rats following (i) an acute morphine dose (5, 10 mg/kg s.c.) with and without JMV2959 pretreatment (3, 6 mg/kg i.p.) or (ii) a morphine challenge dose (5 mg/kg s.c.) with and without JMV2959 (3, 6 mg/kg i.p.) pretreatment, administered during abstinence following repeated doses of morphine (5 days, 10-40 mg/kg). Co-administration of ghrelin (40 ug/kg i.p.) was used to verify the ghrelin mechanisms involvement. RESULTS: Pretreatment with JMV2959 significantly and dose-dependently reversed morphine-induced anandamide increases in the NACSh in both the acute and longer-term models, resulting in a significant AEA decrease. JMV2959 significantly intensified acute morphine-induced decreases in accumbens 2-AG levels and attenuated morphine challenge-induced 2-AG decreases. JMV2959 pretreatment significantly reduced concurrent morphine challenge-induced behavioral sensitization. JMV2959 pretreatment effects were abolished by co-administration of ghrelin. CONCLUSIONS: Our results indicate significant involvement of ghrelin signaling in morphine-induced endocannabinoid changes in the NACSh.


Assuntos
Endocanabinoides/fisiologia , Grelina/fisiologia , Morfina/farmacologia , Entorpecentes/farmacologia , Núcleo Accumbens/metabolismo , Animais , Ácidos Araquidônicos/metabolismo , Relação Dose-Resposta a Droga , Endocanabinoides/metabolismo , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Glicerídeos/metabolismo , Glicina/análogos & derivados , Glicina/farmacologia , Masculino , Núcleo Accumbens/efeitos dos fármacos , Alcamidas Poli-Insaturadas/metabolismo , Ratos , Ratos Wistar , Receptores de Grelina/antagonistas & inibidores , Receptores da Somatotropina/antagonistas & inibidores , Comportamento Estereotipado/efeitos dos fármacos , Triazóis/farmacologia
11.
J Neurosci ; 22(19): 8676-83, 2002 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-12351742

RESUMO

In the present study, we describe a new role of the neuronal nitric oxide synthase (nNOS) gene in the regulation of alcohol drinking behavior. Mice deficient in the nNOS gene (nNOS -/-) and wild-type control mice were submitted to a two-bottle free-choice procedure with either water or increasing concentrations of alcohol (2-16%) for 6 weeks. nNOS -/- mice did not differ in consumption and preference for low alcohol concentrations from wild-type animals; however, nNOS -/- mice consumed sixfold more alcohol from highly concentrated alcohol solutions than wild-type mice. Taste studies with either sucrose or quinine solutions revealed that alcohol intake in nNOS -/- and wild-type mice is associated, at least in part, with sweet solution intake but not with the taste of bitterness. When compared with wild-type mice, the nNOS -/- mice were found to be less sensitive to the sedative effects of ethanol as measured by shorter recovery time from ethanol-induced sleep and did not develop rapid tolerance to ethanol-induced hypothermia, although plasma ethanol concentrations were not significantly different from those of controls. Our findings contrast with previous reports that showed that nonselective NOS inhibitors decrease alcohol consumption. However, because alcohol consumption was suppressed in wild-type as well as nNOS -/- mice by the NOS inhibitor N(G)-nitro-L-arginine methyl ester, we conclude that the effect of nonselective NOS inhibitors on alcohol drinking is not mediated by nNOS. Other NOS isoforms, most likely in the periphery or other splice variants of the NOS gene, might contribute to the effect of nonselective NOS inhibitors on alcohol drinking. In summary, the nNOS gene is critically involved in the regulation of neurobehavioral effects of alcohol.


Assuntos
Encéfalo/efeitos dos fármacos , Comportamento de Escolha/efeitos dos fármacos , Tolerância a Medicamentos/fisiologia , Etanol/farmacologia , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Administração Oral , Processamento Alternativo , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/enzimologia , Comportamento de Escolha/fisiologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Etanol/sangue , Homozigoto , Hipotermia/induzido quimicamente , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase Tipo I , Quinina/administração & dosagem , RNA Mensageiro/metabolismo , Reflexo/efeitos dos fármacos , Autoadministração , Sacarose/administração & dosagem , Paladar/efeitos dos fármacos
12.
World J Biol Psychiatry ; 16(7): 521-35, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26444572

RESUMO

OBJECTIVES: The self-medication hypothesis assumes that symptoms related to potential monoaminergic deficits in depression may be relieved by drug abuse. The aim of this study was to elucidate the neurotransmitter changes in a rat model of depression by measuring their levels in the nucleus accumbens shell, which is typically involved in the drug of abuse acquisition mechanism. METHODS: Depression was modelled by the olfactory bulbectomy (OBX) in Wistar male rats. In vivo microdialysis was performed, starting from the baseline and following after a single methamphetamine injection and behaviour was monitored. The determination of neurotransmitters and their metabolites was performed by high-performance liquid chromatography combined with mass spectrometry. RESULTS: OBX animals had lower basal levels of dopamine and serotonin and their metabolites. However, γ-aminobutyric acid (GABA) and glutamate levels were increased. The methamphetamine injection induced stronger dopamine and serotonin release in the OBX rats and lower release of glutamate in comparison with sham-operated rats; GABA levels did not differ significantly. CONCLUSIONS: This study provides an evidence of mesolimbic neurotransmitter changes in the rat model of depression which may elucidate mechanisms underlying intravenous self-administration studies in which OBX rats were demonstrated to have higher drug intake in comparison to intact controls.


Assuntos
Depressão/tratamento farmacológico , Dopaminérgicos/administração & dosagem , Metanfetamina/administração & dosagem , Neurotransmissores/metabolismo , Recompensa , Animais , Modelos Animais de Doenças , Dopamina/metabolismo , Ácido Glutâmico/metabolismo , Masculino , Microdiálise , Núcleo Accumbens/fisiopatologia , Ratos , Ratos Wistar , Autoadministração , Serotonina/metabolismo , Ácido gama-Aminobutírico/metabolismo
13.
Invest Radiol ; 39(3): 171-81, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15076009

RESUMO

RATIONALE AND OBJECTIVES: Newer radiologic techniques require fast bolus injections and thus low-viscosity, high-concentration, well-tolerated contrast media (CM), especially in vulnerable patients. To this end, we designed and developed iosimenol, a novel isotonic nonionic dimer, and have conducted tests to enable its clinical evaluation. METHODS: Standard physicochemical methods were used. Effects on erythrocyte morphology and coagulation were investigated in human and rat blood. Neural tolerance was assessed by behavioral tests in rats after intracisternal injection. Immunosensitizing potential was evaluated by the skin sensitization test in guinea pigs and by the popliteal lymph node assay in rats. Pharmacokinetics and biotransformation were investigated in rats and dogs. RESULTS: Iosimenol is extremely hydrophilic, it is less viscous than any other isotonic CM, has little effect on erythrocytes and blood coagulation, and has good neural tolerance. No immunosensitizing effect was found in validated animal models. Pharmacokinetics are identical with other angio- and urographic CM. CONCLUSIONS: Iosimenol is the only CM which, although isotonic, affords, unlike current nonionic dimers, at the same iodine concentration the low viscosity of monomeric, nonionic agents, which are all hypertonic. Iosimenol's pharmacologic characteristics closely resemble those of iotrolan and iodixanol.


Assuntos
Benzamidas/farmacologia , Meios de Contraste/farmacologia , Propanolaminas/farmacologia , Animais , Benzamidas/química , Benzamidas/imunologia , Benzamidas/metabolismo , Coagulação Sanguínea/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Meios de Contraste/química , Meios de Contraste/metabolismo , Cães , Relação Dose-Resposta a Droga , Eritrócitos/efeitos dos fármacos , Feminino , Cobaias , Humanos , Linfonodos/efeitos dos fármacos , Masculino , Sistema Nervoso/efeitos dos fármacos , Concentração Osmolar , Propanolaminas/química , Propanolaminas/imunologia , Propanolaminas/metabolismo , Ratos , Ratos Wistar , Testes de Irritação da Pele , Distribuição Tecidual , Ácidos Tri-Iodobenzoicos/química , Ácidos Tri-Iodobenzoicos/imunologia , Ácidos Tri-Iodobenzoicos/farmacologia , Viscosidade
14.
Psychopharmacology (Berl) ; 231(14): 2899-908, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24531567

RESUMO

RATIONALE AND OBJECTIVES: Ghrelin, an orexigenic (appetite stimulating) peptide activates binding sites in the ventral tegmental area (a structure linked with the neural reward system) allowing it to participate in reward-seeking behavior. An increasing number of studies over the past few years have demonstrated ghrelin's role in alcohol, cocaine, and nicotine abuse. However, the role of ghrelin, in opioid effects, has rarely been examined. The aim of the present study was to ascertain whether a ghrelin antagonist (JMV2959) was able to inhibit markers of morphine-induced activation of the neural reward system, namely morphine-induced increase of dopamine in the nucleus accumbens and behavioral changes in rats. METHODS: We used in vivo microdialysis to determine changes of dopamine and its metabolites in the nucleus accumbens shell in rats following morphine (MO, 5, 10 mg/kg s.c.) administration with and without ghrelin antagonist pretreatment (JMV2959, 3, 6 mg/kg i.p., 20 min before MO). Induced behavioral changes were simultaneously monitored. RESULTS: JMV2959 significantly and dose dependently reduced MO-induced dopamine release in the nucleus accumbens shell and affected concentration of by-products associated with dopamine metabolism: 3-methoxytyramine (3-MT), 3,4-dihydroxyphenylacetic acid (DOPAC), and homovanillic acid (HVA). JMV2959 pretreatment also significantly reduced MO-induced behavioral stimulation, especially stereotyped behavior. CONCLUSIONS: Ghrelin secretagogue receptors (GHS-R1A) appear to be involved in the opioid-induced changes in the mesolimbic dopaminergic system associated with the reward processing.


Assuntos
Dopamina/metabolismo , Glicina/análogos & derivados , Morfina/farmacologia , Receptores de Grelina/antagonistas & inibidores , Triazóis/farmacologia , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Dopamina/análogos & derivados , Dopamina/farmacologia , Relação Dose-Resposta a Droga , Grelina/metabolismo , Glicina/administração & dosagem , Glicina/farmacologia , Ácido Homovanílico/farmacologia , Masculino , Microdiálise , Morfina/administração & dosagem , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Ratos , Ratos Wistar , Receptores de Grelina/metabolismo , Recompensa , Comportamento Estereotipado/efeitos dos fármacos , Triazóis/administração & dosagem
15.
J Chromatogr A ; 1303: 94-9, 2013 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-23866123

RESUMO

A new variant of large-volume sample stacking injection (LVSS) was used in the capillary electrophoresis with capacitively coupled contactless conductivity detection (CE/C(4)D) determination of the neurotransmitters γ-aminobutyric acid (GABA), glycine (Gly) and glutamate (Glu) in microdialysates of periaqueductal gray matter (PAG). The separation capillary was filled to 98% from the injection side with a sample of microdialysate in acetonitrile. Simultaneously with turning on the separation voltage, the sample zone was forced out by the background electrolyte by increasing the pressure in the terminal capillary outlet vessel. As a consequence of the stacking effect, the analyte was concentrated from the large sample volume into a narrow zone at the sample/background electrolyte boundary close to the injection end of the capillary. Under these conditions, LOD values of 9, 10 and 15nM were determined in the model samples for GABA, Gly and Glu, respectively; RSD equalled 0.5% for the migration times and 1.0-1.9% for the peak areas, respectively. In analysis of microdialysates of PAG, LOD values of 29, 29 and 37nM were determined for GABA, Gly and Glu, respectively; RSD equalled 0.5-0.7% for the migration times and 2.6-8.2% for the peak areas, respectively. The determined basal levels of the neurotransmitters in PAG microdialysates are 0.08, 4.7 and 0.8µM for GABA, Gly and Glu, respectively. Carrageenan-induced hyperalgesia increases the Gly and Glu levels and reduces GABA in PAG microdialysate. Peroral administration of paracetamol in hyperalgesia effectively reduces the Gly value and has no effect on Glu and GABA.


Assuntos
Eletroforese Capilar/métodos , Ácido Glutâmico/análise , Glicina/análise , Neurotransmissores/análise , Substância Cinzenta Periaquedutal/química , Ácido gama-Aminobutírico/análise , Animais , Eletroforese Capilar/instrumentação , Microdiálise , Ratos
16.
Psychopharmacology (Berl) ; 234(1): 1-2, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27761587
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa