Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nitric Oxide ; 131: 1-7, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36513266

RESUMO

Besides enhanced feeding, the orexigenic peptide ghrelin activates the mesolimbic dopamine system to cause reward as measured by locomotor stimulation, dopamine release in nucleus accumbens shell (NAcS), and conditioned place preference. Although the ventral tegmental area (VTA) appears to be a central brain region for this ghrelin-reward, the underlying mechanisms within this area are unknown. The findings that the gaseous neurotransmitter nitric oxide (NO) modulate the ghrelin enhanced feeding, led us to hypothesize that ghrelin increases NO levels in the VTA, and thereby stimulates reward-related behaviors. We initially demonstrated that inhibition of NO synthesis blocked the ghrelin-induced activation of the mesolimbic dopamine system. We then established that antagonism of downstream signaling of NO in the VTA, namely sGC, prevents the ability of ghrelin to stimulate the mesolimbic dopamine system. The association of ghrelin to NO was further strengthened by in vivo electrochemical recordings showing that ghrelin enhances the NO release in the VTA. Besides a GABAB -receptor agonist, known to reduce NO and cGMP, blocks the stimulatory properties of ghrelin. The present series of experiments reveal that ablated NO signaling, through pharmacologically inhibiting the production of NO and/or cGMP, prevents the ability of ghrelin to induced reward-related behaviors.


Assuntos
Dopamina , Grelina , Óxido Nítrico , Recompensa , Área Tegmentar Ventral , Dopamina/metabolismo , Grelina/farmacologia , Grelina/fisiologia , Óxido Nítrico/metabolismo , Área Tegmentar Ventral/metabolismo , Animais , Camundongos , Ratos , Comportamento Animal
2.
Alcohol Alcohol ; 52(4): 425-430, 2017 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-28481975

RESUMO

AIMS: The orexigenic peptide ghrelin may enhance the incentive value of food-, drug- and alcohol-related rewards. Consistent with preclinical findings, human studies indicate a role of ghrelin in alcohol use disorders (AUD). In the present study an a priori hypothesis-driven analysis was conducted to investigate whether a Leu72Met missense polymorphism (rs696217) in the prepro-ghrelin gene (GHRL), is associated with AUD, alcohol consumption and subjective responses to alcohol. METHOD: Association analysis was performed using the National Institute on Alcohol Abuse and Alcoholism (NIAAA) clinical sample, comprising AUD individuals and controls (N = 1127). Then, a post-hoc analysis using data from a human laboratory study of intravenous alcohol self-administration (IV-ASA, N = 144) was performed to investigate the association of this SNP with subjective responses following a fixed dose of alcohol (priming phase) and alcohol self-administration (ad libitum phase). RESULTS: The case-control study revealed a trend association (N = 1127, OR = 0.665, CI = 0.44-1.01, P = 0.056) between AUD diagnosis and Leu72Met. In AUD subjects, the SNP was associated with significantly lower average drinks per day (n = 567, ß = -2.49, 95% CI = -4.34 to -0.64, P = 0.008) and significantly fewer heavy drinking days (n = 567, ß = -12.00, 95% CI = -19.10 to -4.89, P < 0.001). The IV-ASA study further revealed that 72Met carriers had greater subjective responses to alcohol (P < 0.05) when compared to Leu72Leu both at priming and during ad lib self-administration. CONCLUSION: Although preliminary, these findings suggest that the Leu72Leu genotype may lead to increased risk of AUD possibly via mechanisms involving a lower response to alcohol resulting in excessive alcohol consumption. Further investigations are warranted. SHORT SUMMARY: We investigated whether a Leu72Met missense polymorphism in the prepro-ghrelin gene, is associated with alcohol use disorder, alcohol consumption and subjective responses to alcohol. Although preliminary, results suggest that the Leu72Leu genotype may lead to increased risk of alcohol use disorder possibly via mechanisms involving a lower response to alcohol.


Assuntos
Consumo de Bebidas Alcoólicas/psicologia , Alcoolismo/genética , Grelina/genética , Administração Intravenosa , Adulto , Estudos de Casos e Controles , Etanol/administração & dosagem , Etanol/farmacologia , Predisposição Genética para Doença/genética , Humanos , Masculino , Mutação de Sentido Incorreto/genética , Autoadministração , Adulto Jovem
3.
Addict Biol ; 22(3): 640-651, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-26769653

RESUMO

By investigating the neurochemical mechanisms through which alcohol activates the brain reward systems, novel treatment strategies for alcohol use disorder (AUD), a chronic relapsing disease, can be developed. In contrast to the common view of the function of gut-brain peptides, such as neuromedin U (NMU), to regulate food intake and appetite, a novel role in reinforcement mediation has been implied. The anorexigenic effects of NMU are mediated via NMU2 receptors, preferably in the arcuate nucleus and paraventricular nucleus. The expression of NMU2 receptors is also expressed in several reward-related areas in the brain, suggesting a role in reward regulation. The present experiments were therefore set up to investigate the effect of intracerebroventricular administration of NMU on alcohol-mediated behaviors in rodents. We found that central administration of NMU attenuated alcohol-induced locomotor stimulation, accumbal dopamine release and the expression of conditioned place preference in mice. In addition, NMU dose dependently decreased alcohol intake in high, but not in low, alcohol-consuming rats. Central NMU administration did not alter the blood alcohol concentrations nor change the corticosterone levels in rodents. Given that AUD is a major health-care challenge causing an enormous cost to society and novel treatment strategies are warranted, our data suggest that NMU analogues deserve to be evaluated as novel treatment of AUD in humans.


Assuntos
Consumo de Bebidas Alcoólicas , Comportamento Animal/efeitos dos fármacos , Comportamento de Ingestão de Líquido/efeitos dos fármacos , Etanol/administração & dosagem , Neuropeptídeos/farmacologia , Recompensa , Animais , Masculino , Modelos Animais , Neuropeptídeos/administração & dosagem , Ratos
4.
PLoS One ; 11(5): e0154477, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27139195

RESUMO

Amphetamine dependence, besides its substantial economical consequence, is a serious cause of mortality and morbidity. By investigations of the neurochemical correlates through which addictive drugs, such as amphetamine, activate the mesoaccumbal dopamine system unique targets for treatment of drug addiction can be identified. This reward link consists of a dopamine projection from the ventral tegmental area to the nucleus accumbens (NAc) suggesting that these brain areas are important for reward. The physiological function of gut-brain peptides has expanded beyond food intake modulation and involves regulation of drug reinforcement. A novel candidate for reward regulation is the anorexigenic peptide neuromedin U (NMU). We therefore investigated the effects of intracerebroventricular (icv) administration of NMU on amphetamine's well-documented effects on the mesoaccumbal dopamine system, i.e. locomotor stimulation and accumbal dopamine release in mice. In addition, the effect of accumbal NMU administration on locomotor activity was examined. The effect of NMU, icv or intra-NAc, on the expression of conditioned place preference (CPP) was elucidated. Firstly, we showed that icv administration of NMU attenuate the amphetamine-induced locomotor stimulation, accumbal dopamine release and expression of CPP in mice. Secondly, we found that a lower dose of NMU (icv) reduce the amphetamine-induced locomotor stimulation in mice. Thirdly, we demonstrated that NMU administration into the NAc block the ability of amphetamine to cause a locomotor stimulation in mice. However, accumbal NMU administration did not attenuate the amphetamine-induced expression of CPP in mice. Our novel data suggest that central NMU signalling is involved in development of amphetamine dependence.


Assuntos
Anfetamina/farmacologia , Condicionamento Psicológico/fisiologia , Dopamina/metabolismo , Locomoção/efeitos dos fármacos , Neuropeptídeos/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Comportamento Espacial/efeitos dos fármacos , Animais , Comportamento de Procura de Droga/efeitos dos fármacos , Locomoção/fisiologia , Masculino , Camundongos , Núcleo Accumbens/metabolismo , Recompensa
5.
Alcohol Alcohol ; 51(2): 121-7, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26330568

RESUMO

AIMS: Ghrelin initially emerged as a gut-brain hormone controlling food intake, meal initiation and appetite mainly via hypothalamic circuits in both rodents and humans. The findings that ghrelin receptors (GHS-R1A) are expressed in reward-related areas, including the nucleus accumbens (NAc) and ventral tegmental area (VTA), suggest that ghrelin is a novel reward regulator. Indeed, ghrelin signalling mediates the rewarding and motivational properties of addictive drugs. In addition, daily co-administration of a GHS-R1A antagonist and various addictive drugs prevents the drug-induced locomotor sensitization in rats. METHODS: The present series of experiment were designed to evaluate the effect of repeated pharmacological GHS-R1A suppression on drug-induced locomotor stimulation in more detail. RESULTS: We showed that sub-chronic pre-treatment of the GHS-R1A antagonist, JMV2959, attenuated the ability of acute administration of alcohol as well as of amphetamine to stimulate locomotion. However, there was no effect of sub-chronic JMV2959 treatment on locomotor activity per se or on the expression of the GHS-R1A gene (Ghsr) in the VTA or the NAc compared with vehicle treatment. In addition, sub-chronic ghrelin treatment caused a locomotor sensitization. CONCLUSIONS: While previous research has pinpointed ghrelin as an appetite regulator the present study together with previous studies suggest that ghrelin signalling modulates various reward-mediated behaviours in rodents. Collectively, this suggests that the GHS-R1A could be a key target for novel treatment strategies for addiction.


Assuntos
Anfetamina/administração & dosagem , Etanol/administração & dosagem , Glicina/análogos & derivados , Locomoção/efeitos dos fármacos , Receptores de Grelina/antagonistas & inibidores , Triazóis/administração & dosagem , Animais , Estimulantes do Sistema Nervoso Central/administração & dosagem , Glicina/administração & dosagem , Locomoção/fisiologia , Masculino , Camundongos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Receptores de Grelina/biossíntese , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo
6.
Addict Biol ; 21(2): 422-37, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26303264

RESUMO

The incretin hormone, glucagon-like peptide 1 (GLP-1), regulates gastric emptying, glucose-dependent stimulation of insulin secretion and glucagon release, and GLP-1 analogs are therefore approved for treatment of type II diabetes. GLP-1 receptors are expressed in reward-related areas such as the ventral tegmental area and nucleus accumbens, and GLP-1 was recently shown to regulate several alcohol-mediated behaviors as well as amphetamine-induced, cocaine-induced and nicotine-induced reward. The present series of experiments were undertaken to investigate the effect of the GLP-1 receptor agonist, liraglutide, on several alcohol-related behaviors in rats that model different aspects of alcohol use disorder in humans. Acute liraglutide treatment suppressed the well-documented effects of alcohol on the mesolimbic dopamine system, namely alcohol-induced accumbal dopamine release and conditioned place preference in mice. In addition, acute administration of liraglutide prevented the alcohol deprivation effect and reduced alcohol intake in outbred rats, while repeated treatment of liraglutide decreased alcohol intake in outbred rats as well as reduced operant self-administration of alcohol in selectively bred Sardinian alcohol-preferring rats. Collectively, these data suggest that GLP-1 receptor agonists could be tested for treatment of alcohol dependence in humans.


Assuntos
Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Hipoglicemiantes/farmacologia , Liraglutida/farmacologia , Reforço Psicológico , Recompensa , Alcoolismo/tratamento farmacológico , Animais , Condicionamento Operante/efeitos dos fármacos , Modelos Animais de Doenças , Dopamina/metabolismo , Camundongos , Microdiálise/instrumentação , Núcleo Accumbens/efeitos dos fármacos , Próteses e Implantes , Ratos , Autoadministração
7.
Addict Biol ; 21(2): 481-8, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26059200

RESUMO

The multifaceted gut-brain peptide ghrelin and its receptor (GHSR-1a) are implicated in mechanisms regulating not only the energy balance but also the reward circuitry. In our pre-clinical models, we have shown that ghrelin increases whereas GHSR-1a antagonists decrease alcohol consumption and the motivation to consume alcohol in rodents. Moreover, ghrelin signaling is required for the rewarding properties of addictive drugs including alcohol and nicotine in rodents. Given the hereditary component underlying addictive behaviors and disorders, we sought to investigate whether single nucleotide polymorphisms (SNPs) located in the pre-proghrelin gene (GHRL) and GHSR-1a gene (GHSR) are associated with alcohol use, measured by the alcohol use disorders identification test (AUDIT) and smoking. Two SNPs located in GHRL, rs4684677 (Gln90Leu) and rs696217 (Leu72Met), and one in GHSR, rs2948694, were genotyped in a subset (n = 4161) of a Finnish population-based cohort, the Genetics of Sexuality and Aggression project. The effect of these SNPs on AUDIT scores and smoking was investigated using linear and logistic regressions, respectively. We found that the minor allele of the rs2948694 SNP was nominally associated with higher AUDIT scores (P = 0.0204, recessive model) and smoking (P = 0.0002, dominant model). Furthermore, post hoc analyses showed that this risk allele was also associated with increased likelihood of having high level of alcohol problems as determined by AUDIT scores ≥ 16 (P = 0.0043, recessive model). These convergent findings lend further support for the hypothesized involvement of ghrelin signaling in addictive disorders.


Assuntos
Transtornos Relacionados ao Uso de Álcool/genética , Grelina/genética , Receptores de Grelina/genética , Fumar/genética , Adulto , Feminino , Genótipo , Humanos , Masculino , Mutação de Sentido Incorreto/genética , Polimorfismo de Nucleotídeo Único/genética
8.
Eur Neuropsychopharmacol ; 25(12): 2364-71, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26508707

RESUMO

Gut-brain hormones such as ghrelin have recently been suggested to have a role in reward regulation. Ghrelin was traditionally known to regulate food intake and body weight homoeostasis. In addition, recent work has pin-pointed that this peptide has a novel role in drug-induced reward, including morphine-induced increase in the extracellular levels of accumbal dopamine in rats. Herein the effect of the ghrelin receptor (GHS-R1A) antagonist, JMV2959, on morphine-induced activation of the mesolimbic dopamine system was investigated in mice. In addition, the effects of JMV2959 administration on opioid peptide levels in reward related areas were investigated. In the present series of experiment we showed that peripheral JMV2959 administration, at a dose with no effect per se, attenuates the ability of morphine to cause locomotor stimulation, increase the extracellular levels of accumbal dopamine and to condition a place preference in mice. JMV2959 administration significantly increased tissue levels of Met-enkephalin-Arg(6)Phe(7) in the ventral tegmental area, dynorphin B in hippocampus and Leu-enkephalin-Arg(6) in striatum. We therefore hypothesise that JMV2959 prevents morphine-induced reward via stimulation of delta receptor active peptides in striatum and ventral tegmental areas. In addition, hippocampal peptides that activate kappa receptor may be involved in JMV2959׳s ability to regulate memory formation of reward. Given that development of drug addiction depends, at least in part, of the effects of addictive drugs on the mesolimbic dopamine system the present data suggest that GHS-R1A antagonists deserve to be elucidated as novel treatment strategies of opioid addiction.


Assuntos
Analgésicos Opioides/farmacologia , Encéfalo/metabolismo , Glicina/análogos & derivados , Morfina/farmacologia , Peptídeos Opioides/farmacologia , Receptores de Grelina/antagonistas & inibidores , Recompensa , Triazóis/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Condicionamento Operante/efeitos dos fármacos , Dopamina/metabolismo , Dinorfinas/metabolismo , Endorfinas/metabolismo , Líquido Extracelular/efeitos dos fármacos , Líquido Extracelular/enzimologia , Líquido Extracelular/metabolismo , Glicina/farmacologia , Masculino , Camundongos , Microdiálise , Atividade Motora/efeitos dos fármacos
9.
Psychopharmacology (Berl) ; 232(23): 4285-92, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26319159

RESUMO

RATIONALE: Schizophrenic-spectrum patients commonly display deficits in preattentive information processing as evidenced, for example, by disrupted prepulse inhibition (PPI), a measure of sensorimotor gating. Similar disruptions in PPI can be induced in rodents and primates by the psychotomimetic drug phencyclidine (PCP), a noncompetitive inhibitor of the NMDA receptor. Mounting evidence suggests that the hunger hormone ghrelin and its constitutively active receptor influences neuronal circuits involved in the regulation of mood and cognition. OBJECTIVES: In the present series of experiments, we investigated the effects of ghrelin and the growth hormone secretagogue receptor (GHS-R1A) neutral antagonist, JMV 2959, on acoustic startle responses (ASR), PPI, and PCP-induced alterations in PPI. RESULTS: Intraperitoneal (i.p.) administration of ghrelin (0.033, 0.1, and 0.33 mg/kg) did not alter the ASR or PPI in rats. Conversely, i.p. injection of JMV 2959 (1, 3, and 6 mg/kg), dose dependently decreased the ASR and increased PPI. Pretreatment with JMV 2959 at a dose with no effect on ASR or PPI per se, completely blocked PCP-induced (2 mg/kg) deficits in PPI while pretreatment with the highest dose of ghrelin did not potentiate or alter PPI responses of a sub-threshold dose of PCP (0.75 mg/kg). CONCLUSION: These findings indicate that the GHS-R1A is involved in specific behavioral effects of PCP and may have relevance for patients with schizophrenia.


Assuntos
Glicina/análogos & derivados , Fenciclidina/farmacologia , Inibição Pré-Pulso/efeitos dos fármacos , Receptores de Grelina/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Triazóis/farmacologia , Animais , Relação Dose-Resposta a Droga , Glicina/farmacologia , Masculino , Fenciclidina/antagonistas & inibidores , Inibição Pré-Pulso/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores de Grelina/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Reflexo de Sobressalto/efeitos dos fármacos , Reflexo de Sobressalto/fisiologia , Esquizofrenia/tratamento farmacológico , Filtro Sensorial/efeitos dos fármacos , Filtro Sensorial/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
10.
CNS Drugs ; 28(10): 875-86, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24958205

RESUMO

Food intake and appetite are regulated by various circulating hormones including ghrelin and glucagon-like-peptide 1 (GLP-1). Ghrelin, mainly released from the stomach, increases food intake, induces appetite, enhances adiposity as well as releases growth hormone. Hypothalamic "ghrelin receptors" (GHS-R1A) have a critical role in food intake regulation, but GHS-R1A are also expressed in reward related areas. GLP-1 is produced in the intestinal mucosa as well as in the hindbrain in response to nutrient ingestion. This gut-brain hormone reduces food intake as well as regulates glucose homeostasis, foremost via GLP-1 receptors in hypothalamus and brain stem. However, GLP-1 receptors are expressed in areas intimately associated with reward regulation. Given that regulation of food and drug intake share common neurobiological substrates, the possibility that ghrelin and GLP-1 play an important role in reward regulation should be considered. Indeed, this leading article describes that the orexigenic peptide ghrelin activates the cholinergic-dopaminergic reward link, an important part of the reward systems in the brain associated with reinforcement and thereby increases the incentive salience for motivated behaviors via this system. We also review the role of ghrelin signaling for reward induced by alcohol and addictive drugs from a preclinical, clinical and human genetic perspective. In addition, the recent findings showing that GLP-1 controls reward induced by alcohol, amphetamine, cocaine and nicotine in rodents are overviewed herein. Finally, the role of several other appetite regulatory hormones for reward and addiction is briefly discussed. Collectively, these data suggest that ghrelin and GLP-1 receptors may be novel targets for development of pharmacological treatments of alcohol and drug dependence.


Assuntos
Apetite/fisiologia , Comportamento Aditivo/fisiopatologia , Encéfalo/fisiopatologia , Grelina/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , Animais , Apetite/genética , Comportamento Aditivo/genética , Encéfalo/efeitos dos fármacos , Grelina/genética , Peptídeo 1 Semelhante ao Glucagon/genética , Humanos , Recompensa , Transtornos Relacionados ao Uso de Substâncias/genética
11.
Prog Brain Res ; 211: 201-33, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24968782

RESUMO

The reward systems are important for rewards, natural and artificial. An important part of these systems is the mesolimbic dopamine system, consisting of a dopamine projection from the ventral tegmental area to nucleus accumbens. There is an underlying disruption in the reward systems in individuals with alcohol dependence. Elucidation of the neurochemical mechanisms involved in the ability of alcohol to activate the mesolimbic dopamine system could identify novel targets for the treatment of alcohol dependence. The role of dopamine, for example, by using the dopamine stabilizer (-)-OSU6162, in alcohol dependence will be revised. Data showing that local perfusion of alcohol into reward nodes activates the mesolimbic dopamine system will be reviewed. The effects of alcohol on ligand-gated ion channels will be introduced. Finally, the findings demonstrating that gut-brain peptides, such as ghrelin and glucagon-like peptide-1, are important for alcohol-mediated will be presented.


Assuntos
Alcoolismo/fisiopatologia , Dopamina/fisiologia , Núcleo Accumbens/fisiopatologia , Recompensa , Área Tegmentar Ventral/fisiopatologia , Animais , Etanol/farmacologia , Humanos , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiopatologia , Núcleo Accumbens/efeitos dos fármacos , Área Tegmentar Ventral/efeitos dos fármacos
12.
Alcohol Clin Exp Res ; 38(4): 959-68, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24428428

RESUMO

BACKGROUND: Development of alcohol dependence, a chronic and relapsing disease, largely depends on the effects of alcohol on the brain reward systems. By elucidating the mechanisms involved in alcohol use disorder, novel treatment strategies may be developed. Ghrelin, the endogenous ligand for the growth hormone secretagogue receptor 1A, acts as an important regulator of energy balance. Recently ghrelin and its receptor were shown to mediate alcohol reward and to control alcohol consumption in rodents. However, the role of central versus peripheral ghrelin for alcohol reward needs to be elucidated. METHODS: Given that ghrelin mainly is produced by peripheral organs, the present study was designed to investigate the role of circulating endogenous ghelin for alcohol reward and for alcohol intake in rodents. RESULTS: We showed that the Spiegelmer NOX-B11-2, which binds and neutralizes acylated ghrelin in the periphery with high affinity and thus prevents its brain access, does not attenuate the alcohol-induced locomotor activity, accumbal dopamine release and expression of conditioned place preference in mice. Moreover, NOX-B11-2 does not affect alcohol intake using the intermittent access 20% alcohol 2-bottle-choice drinking paradigm in rats, suggesting that circulating ghrelin does not regulate alcohol intake or the rewarding properties of alcohol. In the present study, we showed however, that NOX-B11-2 reduced food intake in rats supporting a role for circulating ghrelin as physiological regulators of food intake. Moreover, NOX-B11-2 did not affect the blood alcohol concentration in mice. CONCLUSIONS: Collectively, the past and present studies suggest that central, rather than peripheral, ghrelin signaling may be a potential target for pharmacological treatment of alcohol dependence.


Assuntos
Consumo de Bebidas Alcoólicas/sangue , Grelina/sangue , Recompensa , Consumo de Bebidas Alcoólicas/fisiopatologia , Animais , Biomarcadores/sangue , Grelina/antagonistas & inibidores , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Oligonucleotídeos/farmacologia , Ratos , Ratos Wistar
13.
PLoS One ; 8(10): e77284, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24204788

RESUMO

The gastrointestinal peptide glucagon-like peptide 1 (GLP-1) is known to regulate consummatory behavior and is released in response to nutrient ingestion. Analogues of this peptide recently emerged as novel pharmacotherapies for treatment of type II diabetes since they reduce gastric emptying, glucagon secretion as well as enhance glucose-dependent insulin secretion. The findings that GLP-1 targets reward related areas including mesolimbic dopamine areas indicate that the physiological role of GLP-1 extends beyond food intake and glucose homeostasis control to include reward regulation. The present series of experiments was therefore designed to investigate the effects of the GLP-1 receptor agonist, Exendin-4 (Ex4), on established nicotine-induced effects on the mesolimbic dopamine system in mice. Specifically, we show that treatment with Ex4, at a dose with no effect per se, attenuate nicotine-induced locomotor stimulation, accumbal dopamine release as well as the expression of conditioned place preference in mice. In accordance, Ex4 also blocks nicotine-induced expression of locomotor sensitization in mice. Given that development of nicotine addiction largely depends on the effects of nicotine on the mesolimbic dopamine system these findings indicate that the GLP-1 receptor may be a potential target for the development of novel treatment strategies for nicotine cessations in humans.


Assuntos
Dopamina/metabolismo , Incretinas/farmacologia , Sistema Límbico/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Peptídeos/farmacologia , Receptores de Glucagon/agonistas , Tabagismo/tratamento farmacológico , Peçonhas/farmacologia , Animais , Condicionamento Psicológico/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Exenatida , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1 , Sistema Límbico/metabolismo , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Nicotina/farmacologia , Núcleo Accumbens/metabolismo , Receptores de Glucagon/metabolismo , Recompensa , Tabagismo/metabolismo , Tabagismo/psicologia
14.
PLoS One ; 8(8): e71284, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23977009

RESUMO

Alcohol dependence is a heterogeneous disorder where several signalling systems play important roles. Recent studies implicate that the gut-brain hormone ghrelin, an orexigenic peptide, is a potential mediator of alcohol related behaviours. Ghrelin increases whereas a ghrelin receptor (GHS-R1A) antagonist decreases alcohol consumption as well as operant self-administration of alcohol in rodents that have consumed alcohol for twelve weeks. In the present study we aimed at investigating the effect of acute and repeated treatment with the GHS-R1A antagonist JMV2959 on alcohol intake in a group of rats following voluntarily alcohol consumption for two, five and eight months. After approximately ten months of voluntary alcohol consumption the expression of the GHS-R1A gene (Ghsr) as well as the degree of methylation of a CpG island found in Ghsr was examined in reward related brain areas. In a separate group of rats, we examined the effect of the JMV2959 on alcohol relapse using the alcohol deprivation paradigm. Acute JMV2959 treatment was found to decrease alcohol intake and the effect was more pronounced after five, compared to two months of alcohol exposure. In addition, repeated JMV2959 treatment decreased alcohol intake without inducing tolerance or rebound increase in alcohol intake after the treatment. The GHS-R1A antagonist prevented the alcohol deprivation effect in rats. There was a significant down-regulation of the Ghsr expression in the ventral tegmental area (VTA) in high- compared to low-alcohol consuming rats after approximately ten months of voluntary alcohol consumption. Further analysis revealed a negative correlation between Ghsr expression in the VTA and alcohol intake. No differences in methylation degree were found between high- compared to low-alcohol consuming rats. These findings support previous studies showing that the ghrelin signalling system may constitute a potential target for development of novel treatment strategies for alcohol dependence.


Assuntos
Consumo de Bebidas Alcoólicas/tratamento farmacológico , Alcoolismo/tratamento farmacológico , Grelina/metabolismo , Glicina/análogos & derivados , Receptores de Grelina/antagonistas & inibidores , Síndrome de Abstinência a Substâncias/prevenção & controle , Triazóis/farmacologia , Área Tegmentar Ventral/efeitos dos fármacos , Administração Oral , Consumo de Bebidas Alcoólicas/metabolismo , Consumo de Bebidas Alcoólicas/fisiopatologia , Alcoolismo/metabolismo , Alcoolismo/fisiopatologia , Animais , Ilhas de CpG , Metilação de DNA/efeitos dos fármacos , Regulação da Expressão Gênica , Grelina/genética , Glicina/farmacologia , Masculino , Ratos , Ratos Wistar , Receptores de Grelina/genética , Receptores de Grelina/metabolismo , Autoadministração , Transdução de Sinais
15.
PLoS One ; 8(7): e69010, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23874851

RESUMO

Glucagon-like peptide 1 (GLP-1) is an incretine hormone that controls consummatory behavior and glucose homeostasis. It is released in response to nutrient ingestion from the intestine and production in the brain has also been identified. Given that GLP-1 receptors are expressed in reward areas, such as the nucleus accumbens and ventral tegmental area, and that common mechanisms regulate food and drug-induced reward we hypothesize that GLP-1 receptors are involved in reward regulation. Herein the effect of the GLP-1 receptor agonist Exendin-4 (Ex4), on amphetamine- and cocaine-induced activation of the mesolimbic dopamine system was investigated in mice. In a series of experiments we show that treatment with Ex4, at a dose with no effect per se, reduce amphetamine- as well as cocaine-induced locomotor stimulation, accumbal dopamine release as well as conditioned place preference in mice. Collectively these data propose a role for GLP-1 receptors in regulating drug reward. Moreover, the GLP-1 signaling system may be involved in the development of drug dependence since the rewarding effects of addictive drugs involves interferences with the mesolimbic dopamine system. Given that GLP-1 analogues, such as exenatide and liraglutide, are clinically available for treatment of type II diabetes, we propose that these should be elucidated as treatments of drug dependence.


Assuntos
Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Atividade Motora/efeitos dos fármacos , Receptores de Glucagon/metabolismo , Anfetamina/farmacologia , Animais , Estimulantes do Sistema Nervoso Central/farmacologia , Cocaína/farmacologia , Exenatida , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Receptor do Peptídeo Semelhante ao Glucagon 1 , Liraglutida , Masculino , Camundongos , Peptídeos , Peçonhas
16.
PLoS One ; 8(4): e61242, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23579732

RESUMO

The development of amphetamine dependence largely depends on the effects of amphetamine in the brain reward systems. Ghrelin, an orexigenic peptide, activates the reward systems and is required for reward induced by alcohol, nicotine, cocaine and amphetamine in mice. Human genetic studies have shown that polymorphisms in the pre-proghrelin (GHRL) as well as GHS-R1A (GHSR) genes are associated with high alcohol consumption, increased weight and smoking in males. Since the heritability factor underlying drug dependence is shared between different drugs of abuse, we here examine the association between single nucleotide polymorphisms (SNPs) and haplotypes in the GHRL and GHSR, and amphetamine dependence. GHRL and GHSR SNPs were genotyped in Swedish amphetamine dependent individuals (n = 104) and controls from the general population (n = 310). A case-control analysis was performed and SNPs and haplotypes were additionally tested for association against Addiction Severity Interview (ASI) composite score of drug use. The minor G-allele of the GHSR SNP rs2948694, was more common among amphetamine dependent individuals when compared to controls (pc  = 0.02). A significant association between the GHRL SNP rs4684677 and ASI composite score of drug use was also reported (pc  = 0.03). The haplotype analysis did not add to the information given by the individual polymorphisms. Although genetic variability of the ghrelin signalling system is not a diagnostic marker for amphetamine dependence and problem severity of drug use, the present results strengthen the notion that ghrelin and its receptor may be involved in the development of addictive behaviours and may thus serve as suitable targets for new treatments of such disorders.


Assuntos
Transtornos Relacionados ao Uso de Anfetaminas/genética , Transtornos Relacionados ao Uso de Anfetaminas/metabolismo , Anfetamina/metabolismo , Variação Genética , Grelina/genética , Transdução de Sinais , Adulto , Alelos , Estudos de Casos e Controles , Feminino , Frequência do Gene , Estudos de Associação Genética , Grelina/metabolismo , Haplótipos , Humanos , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Receptores de Grelina/genética , Adulto Jovem
17.
Psychoneuroendocrinology ; 38(8): 1259-70, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23219472

RESUMO

Development of alcohol use disorders largely depends on the effects of alcohol on the brain reward systems. Emerging evidence indicate that common mechanisms regulate food and alcohol intake and raise the possibility that endocrine signals from the gut may play an important role for alcohol consumption, alcohol-induced reward and the motivation to consume alcohol. Glucagon-like peptide 1 (GLP-1), a gastrointestinal peptide regulating food intake and glucose homeostasis, has recently been shown to target central brain areas involved in reward and motivation, including the ventral tegmental area and nucleus accumbens. Herein we investigated the effects of the GLP-1 receptor agonist, Exendin-4 (Ex4), on various measures of alcohol-induced reward as well as on alcohol intake and alcohol seeking behavior in rodents. Treatment with Ex4, at a dose with no effect per se, attenuated alcohol-induced locomotor stimulation and accumbal dopamine release in mice. Furthermore, conditioned place preference for alcohol was abolished by both acute and chronic treatment with Ex4 in mice. Finally we found that Ex4 treatment decreased alcohol intake, using the intermittent access 20% alcohol two-bottle-choice model, as well as alcohol seeking behavior, using the progressive ratio test in the operant self-administration model, in rats. These novel findings indicate that GLP-1 signaling attenuates the reinforcing properties of alcohol implying that the physiological role of GLP-1 extends beyond glucose homeostasis and food intake regulation. Collectively these findings implicate that the GLP-1 receptor may be a potential target for the development of novel treatment strategies for alcohol use disorders.


Assuntos
Comportamento de Escolha/efeitos dos fármacos , Comportamento de Procura de Droga/efeitos dos fármacos , Comportamento de Procura de Droga/fisiologia , Etanol/antagonistas & inibidores , Peptídeo 1 Semelhante ao Glucagon/fisiologia , Peptídeos/farmacologia , Recompensa , Peçonhas/farmacologia , Animais , Condicionamento Operante/efeitos dos fármacos , Condicionamento Psicológico/efeitos dos fármacos , Dopamina/metabolismo , Relação Dose-Resposta a Droga , Etanol/administração & dosagem , Etanol/farmacologia , Exenatida , Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Ratos , Autoadministração
18.
PLoS One ; 7(11): e49557, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23166710

RESUMO

Ghrelin, an orexigenic peptide, regulates energy balance specifically via hypothalamic circuits. Growing evidence suggest that ghrelin increases the incentive value of motivated behaviours via activation of the cholinergic-dopaminergic reward link. It encompasses the cholinergic afferent projection from the laterodorsal tegmental area (LDTg) to the dopaminergic cells of the ventral tegmental area (VTA) and the mesolimbic dopamine system projecting from the VTA to nucleus accumbens (N.Acc.). Ghrelin receptors (GHS-R1A) are expressed in these reward nodes and ghrelin administration into the LDTg increases accumbal dopamine, an effect involving nicotinic acetylcholine receptors in the VTA. The present series of experiments were undertaken directly to test this hypothesis. Here we show that ghrelin, administered peripherally or locally into the LDTg concomitantly increases ventral tegmental acetylcholine as well as accumbal dopamine release. A GHS-R1A antagonist blocks this synchronous neurotransmitter release induced by peripheral ghrelin. In addition, local perfusion of the unselective nicotinic antagonist mecamylamine into the VTA blocks the ability of ghrelin (administered into the LDTg) to increase N.Acc.-dopamine, but not VTA-acetylcholine. Collectively our data indicate that ghrelin activates the LDTg causing a release of acetylcholine in the VTA, which in turn activates local nicotinic acetylcholine receptors causing a release of accumbal dopamine. Given that a dysfunction in the cholinergic-dopaminergic reward system is involved in addictive behaviours, including compulsive overeating and alcohol use disorder, and that hyperghrelinemia is associated with such addictive behaviours, ghrelin-responsive circuits may serve as a novel pharmacological target for treatment of alcohol use disorder as well as binge eating.


Assuntos
Acetilcolina/metabolismo , Dopamina/metabolismo , Grelina/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo , Animais , Grelina/administração & dosagem , Masculino , Antagonistas Nicotínicos/administração & dosagem , Antagonistas Nicotínicos/farmacologia , Ratos , Receptores de Grelina/antagonistas & inibidores , Receptores Nicotínicos
19.
Addict Biol ; 17(1): 86-94, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21309944

RESUMO

The mechanisms involved in alcohol use disorders are complex. It has been shown that ghrelin is an important signal for the control of body weight homeostasis, preferably by interacting with hypothalamic circuits, as well as for drug reward by activating the mesolimbic dopamine system. The ghrelin receptor (GHS-R1A) has been shown to be required for alcohol-induced reward. Additionally, ghrelin increases and GHR-R1A antagonists reduce moderate alcohol consumption in mice, and a single nucleotide polymorphism in the GHS-R1A gene has been associated with high alcohol consumption in humans. However, the role of central ghrelin signaling in high alcohol consumption is not known. Therefore, the role of GHS-R1A in operant self-administration of alcohol in rats as well as for high alcohol consumption in Long-Evans rats and in alcohol preferring [Alko alcohol (AA)] rats was studied here. In the present study, the GHS-R1A antagonist, JMV2959, was found to reduce the operant self-administration of alcohol in rats and to decrease high alcohol intake in Long-Evans rats as well as in AA rats. These results suggest that the ghrelin receptor signaling system, specifically GHS-R1A, is required for operant self-administration of alcohol and for high alcohol intake in rats. Therefore, the GHS-R1A may be a therapeutic target for treatment of addictive behaviors, such as alcohol dependence.


Assuntos
Consumo de Bebidas Alcoólicas , Alcoolismo , Depressores do Sistema Nervoso Central/administração & dosagem , Condicionamento Operante/efeitos dos fármacos , Etanol/administração & dosagem , Receptores de Grelina/antagonistas & inibidores , Análise de Variância , Animais , Comportamento de Escolha/efeitos dos fármacos , Modelos Animais de Doenças , Glicina/análogos & derivados , Glicina/farmacologia , Masculino , Ratos , Ratos Long-Evans , Autoadministração/estatística & dados numéricos , Transdução de Sinais/efeitos dos fármacos , Triazóis/farmacologia
20.
Alcohol Clin Exp Res ; 36(1): 97-103, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21797890

RESUMO

BACKGROUND: Alcohol dependence has been associated with reduced function of serotonin, dopamine as well as noradrenaline activities in several neuroendocrine studies. To our knowledge, there is, however, no study investigating all these 3 systems with the use of neuroendocrine methods in one and the same alcohol-dependent individual. METHODS: Alcohol-dependent individuals (n = 42) and controls (n = 28) participated in the neuroendocrine test series. Central serotonergic neurotransmission was assessed by the prolactin (PRL) response to citalopram (CIT). The postsynaptic DRD2 function was measured by the growth hormone (GH) response to apomorphine (APO) and the postsynaptic α2-adrenoceptor function by GH response to clonidine (CLON). RESULTS: In the alcohol-dependent individuals, the PRL concentrations were significantly lower at the time points 240 minutes and 300 minutes after CIT administration and mean delta PRL value was significantly reduced by 45% in comparison with controls. There were no significant differences in APO-GH and CLON-GH concentrations at any time points or in mean delta GH values between the groups. An impaired monoaminergic profile, including all 3 systems, was significantly more frequent in alcohol-dependent individuals than controls (43% vs. 6% respectively). CONCLUSIONS: The monoaminergic dysfunction was restricted to an impairment of the serotonergic system, suggesting that this system is especially vulnerable to long-term and excessive alcohol consumption. Moreover, impaired monoaminergic profiles, including low responses in 2 or 3 systems, were more frequently observed in alcohol-dependent individuals than in controls. Such impaired profiles may be of clinical importance, but further studies are needed.


Assuntos
Alcoolismo/sangue , Dopamina/sangue , Norepinefrina/sangue , Serotonina/sangue , Adulto , Alcoolismo/diagnóstico , Biomarcadores/sangue , Estudos de Casos e Controles , Citalopram/farmacologia , Clonidina/farmacologia , Feminino , Hormônio do Crescimento Humano/sangue , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Sistemas Neurossecretores/efeitos dos fármacos , Sistemas Neurossecretores/metabolismo , Prolactina/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...