Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 181
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Behav Pharmacol ; 35(2-3): 114-121, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38451023

RESUMEN

We hypothesized that opioid receptor antagonists would inhibit motivated behavior produced by a natural reward. To evaluate motivated responses to a natural reward, mice were given access to running wheels for 71.5 h in a multi-configuration testing apparatus. In addition to a running wheel activity, locomotor activity (outside of the wheel), food and water intake, and access to a food container were measured in the apparatus. Mice were also tested separately for novel-object exploration to investigate whether naloxone affects behavior unrelated to natural reward. In untreated mice wheel running increased from day 1 to day 3. The selective µ-opioid receptor antagonist ß-funaltrexamine (ß-FNA) (5 mg/kg) slightly decreased wheel running, but did not affect the increase in wheel running from day 1 to day 3. The non-selective opioid receptor antagonist naloxone produced a greater reduction in wheel running than ß-FNA and eliminated the increase in wheel running that occurred over time in the other groups. Analysis of food access, locomotor behavior, and behavior in the novel-object test suggested that the reduction in wheel running was selective for this highly reinforcing behavior. These results indicate that opioid receptor antagonism reduces responses to the natural rewarding effects of wheel running and that these effects involve multiple opioid receptors since the non-selective opioid receptor antagonist had greater effects than the selective µ-opioid receptor antagonist. It is possible that at the doses employed, other receptor systems than opioid receptors might be involved, at least in part, in the effect of naloxone and ß-FNA.


Asunto(s)
Actividad Motora , Antagonistas de Narcóticos , Animales , Ratones , Antagonistas de Narcóticos/farmacología , Motivación , Naloxona/farmacología , Receptores Opioides
2.
Alcohol Alcohol ; 59(2)2024 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-38145944

RESUMEN

BACKGROUND: Alcohol is a major abused drug worldwide that contributes substantially to health and social problems. These problems result from acute alcohol overuse as well as chronic use, leading to alcohol use disorder (AUD). A major goal of this field is to establish a treatment for alcohol abuse and dependence in patients with AUD. The central molecular mechanisms of acute alcohol actions have been extensively investigated in rodent models. AIMS: One of the central mechanisms that may be involved is glycogen synthase kinase-3ß (GSK-3ß) activity, a key enzyme involved in glycogen metabolism but which has crucial roles in numerous cellular processes. Although the exact mechanisms leading from acute alcohol actions to these chronic changes in GSK-3ß function are not yet clear, GSK-3ß nonetheless constitutes a potential therapeutic target for AUD by reducing its function using GSK-3ß inhibitors. This review is focused on the correlation between GSK-3ß activity and the degree of alcohol consumption. METHODS: Research articles regarding investigation of effect of GSK-3ß on alcohol consumption in rodents were searched on PubMed, Embase, and Scopus databases using keywords "glycogen synthase kinase," "alcohol (or ethanol)," "intake (or consumption)," and evaluated by changes in ratios of pGSK-3ßSer9/pGSK-3ß. RESULTS: In animal experiments, GSK-3ß activity decreases in the brain under forced and voluntary alcohol consumption while GSK-3ß activity increases under alcohol-seeking behavior. CONCLUSIONS: Several pieces of evidence suggest that alterations in GSK-3ß function are important mediators of chronic ethanol actions, including those related to alcohol dependence and the adverse effects of chronic ethanol exposure.


Asunto(s)
Encéfalo , Etanol , Animales , Humanos , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Etanol/efectos adversos , Encéfalo/metabolismo , Consumo de Bebidas Alcohólicas/metabolismo , Fosforilación
3.
Neurochem Res ; 48(7): 2230-2240, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36907972

RESUMEN

We investigated morphine-induced Straub's tail reaction (STR) in mice pretreated with or without glycogen synthase kinase-3 (GSK-3) inhibitors (SB216763 and AR-A014418) by using a newly modified, infrared beam sensor-based automated apparatus. Mice treated with a single injection of morphine (30 mg/kg, i.p.) showed a significant STR with a plateau level at a time point of 20 min after morphine challenge. Pretreatment of mice with SB216763 (5 mg/kg, s.c.) or AR-A014418 (3 mg/kg, i.p.) significantly inhibited morphine-induced STR and attenuated the duration of STR in a dose-dependent fashion. In the striatum and the nucleus accumbens, expression of pGSK-3ßTyr216 but not GSK3ß or pGSK-3ßSer9 was largely but not significantly reduced after treatment with SB216763 (5 mg/kg, s.c.) in combination with/without morphine, indicating that the inhibitory effect of GSK-3 inhibitors on morphine-induced STR and hyperlocomotion might not depend on the direct blockade of GSK-3ß function. In constipated mice after morphine challenge (30 mg/kg), the effect of GSK-3 inhibitors on gastrointestinal transit was examined to reveal whether the action of GSK-3 inhibitors on morphine effects was central and/or peripheral. Pretreatment with SB216763 (5 mg/kg) did not block constipation in morphine-injected mice. The mechanism of action seems to be central but not peripheral, although the underlying subcellular mechanism of GSK-3 inhibitors is not clear. Our measurement system is a useful tool for investigating the excitatory effects of morphine in experimental animals.


Asunto(s)
Glucógeno Sintasa Quinasa 3 , Morfina , Ratones , Animales , Morfina/farmacología , Morfina/uso terapéutico , Glucógeno Sintasa Quinasa 3 beta , Cola (estructura animal)
4.
Behav Pharmacol ; 34(7): 393-403, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37668157

RESUMEN

The psychostimulant drug methamphetamine (METH) causes euphoria in humans and locomotor hyperactivity in rodents by acting on the mesolimbic dopamine (DA) pathway and has severe abuse and addiction liability. Behavioral sensitization, an increased behavioral response to a drug with repeated administration, can persist for many months after the last administration. Research has shown that the serotonin 1B (5-HT1B) receptor plays a critical role in the development and maintenance of drug addiction, as well as other addictive behaviors. This study examined the role of 5-HT1B receptors in METH-induced locomotor sensitization using 5-HT1B knockout (KO) mice. To clarify the action of METH in 5-HT1B KO mice the effects of METH on extracellular levels of DA (DAec) and 5-HT (5-HTec) in the caudate putamen (CPu) and the nucleus accumbens (NAc) were examined. Locomotor sensitization and extracellular monoamine levels were determined in wild-type mice (5-HT1B +/+), heterozygous 5-HT1B receptor KO (5-HT1B +/-) mice and homozygous 5-HT1B receptor KO mice (5-HT1B -/-). Behavioral sensitization to METH was enhanced in 5-HT1B -/- mice compared to 5-HT1B +/+ mice but was attenuated in 5-HT1B +/- mice compared to 5-HT1B +/+ and 5-HT1B -/- mice. In vivo, microdialysis demonstrated that acute administration of METH increases DAec levels in the CPu and NAc of 5-HT1B KO mice compared to saline groups. In 5-HT1B +/- mice, METH increased 5-HTec levels in the CPu, and DAec levels in the NAc were higher than in others.5-HT1B receptors play an important role in regulating METH-induced behavioral sensitization.


Asunto(s)
Estimulantes del Sistema Nervioso Central , Metanfetamina , Humanos , Animales , Ratones , Técnicas de Inactivación de Genes , Metanfetamina/farmacología , Receptor de Serotonina 5-HT1B/genética , Ratones Noqueados , Estimulantes del Sistema Nervioso Central/farmacología , Dopamina , Serotonina
5.
J Pharmacol Sci ; 151(3): 135-141, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36828615

RESUMEN

Previous pharmacological data have shown the possible existence of functional interactions between µ- (MOP), κ- (KOP), and δ-opioid receptors (DOP) in pain and mood disorders. We previously reported that MOP knockout (KO) mice exhibit a lower stress response compared with wildtype (WT) mice. Moreover, DOP agonists have been shown to exert antidepressant-like effects in numerous animal models. In the present study, the tail suspension test (TST) and forced swim test (FST) were used to examine the roles of MOP and DOP in behavioral despair. MOP-KO mice and WT mice were treated with KNT-127 (10 mg/kg), a selective DOP agonist. The results indicated a significant decrease in immobility time in the KNT-127 group compared with the saline group in all genotypes in both tests. In the saline groups, immobility time significantly decreased in MOP-KO mice compared with WT mice in both tests. In female MOP-KO mice, KNT-127 significantly decreased immobility time in the TST compared with WT mice. In male MOP-KO mice, however, no genotypic differences were found in the TST after either KNT-127 or saline treatment. Thus, at least in the FST and TST, the activation of DOP and absence of MOP had additive effects in reducing measures of behavioral despair, suggesting that effects on this behavior by DOP activation occur independently of MOP.


Asunto(s)
Morfinanos , Receptores Opioides mu , Masculino , Femenino , Ratones , Animales , Morfinanos/farmacología , Antidepresivos/farmacología , Analgésicos Opioides/farmacología , Dolor/tratamiento farmacológico
6.
Proc Natl Acad Sci U S A ; 115(45): 11597-11602, 2018 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-30348770

RESUMEN

Receptor-type protein tyrosine phosphatase D (PTPRD) is a neuronal cell-adhesion molecule/synaptic specifier that has been implicated in addiction vulnerability and stimulant reward by human genomewide association and mouse cocaine-conditioned place-preference data. However, there have been no reports of effects of reduced expression on cocaine self-administration. There have been no reports of PTPRD targeting by any small molecule. There are no data about behavioral effects of any PTPRD ligand. We now report (i) robust effects of heterozygous PTPRD KO on cocaine self-administration (These data substantially extend prior conditioned place-preference data and add to the rationale for PTPRD as a target for addiction therapeutics.); (ii) identification of 7-butoxy illudalic acid analog (7-BIA) as a small molecule that targets PTPRD and inhibits its phosphatase with some specificity; (iii) lack of toxicity when 7-BIA is administered to mice acutely or with repeated dosing; (iv) reduced cocaine-conditioned place preference when 7-BIA is administered before conditioning sessions; and (v) reductions in well-established cocaine self-administration when 7-BIA is administered before a session (in WT, not PTPRD heterozygous KOs). These results add to support for PTPRD as a target for medications to combat cocaine use disorders. 7-BIA provides a lead compound for addiction therapeutics.


Asunto(s)
Trastornos Relacionados con Cocaína/tratamiento farmacológico , Cumarinas/farmacología , Antagonistas de Narcóticos/farmacología , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/genética , Recompensa , Abuso de Sustancias por Vía Intravenosa/tratamiento farmacológico , Animales , Catéteres de Permanencia , Trastornos Relacionados con Cocaína/enzimología , Trastornos Relacionados con Cocaína/genética , Trastornos Relacionados con Cocaína/fisiopatología , Condicionamiento Psicológico , Cumarinas/síntesis química , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Inyecciones Intravenosas , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Antagonistas de Narcóticos/síntesis química , Neuronas/efectos de los fármacos , Neuronas/enzimología , Neuronas/patología , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/deficiencia , Autoadministración , Transducción de Señal , Abuso de Sustancias por Vía Intravenosa/enzimología , Abuso de Sustancias por Vía Intravenosa/genética , Abuso de Sustancias por Vía Intravenosa/fisiopatología , Pruebas de Toxicidad Aguda , Pruebas de Toxicidad Crónica
7.
Dev Psychopathol ; 32(2): 703-718, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31256767

RESUMEN

The study examined (a) whether alcohol use subgroups could be identified among African Americans assessed from adolescence through early adulthood, and (b) whether subgroup membership was associated with the interaction between internalizing symptoms and antisocial behavior polygenic risk scores (PRSs) and environmental characteristics (i.e., parental monitoring, community disadvantage). Participants (N = 436) were initially recruited for an elementary school-based prevention trial in a Mid-Atlantic city. Youths reported on the frequency of their past year alcohol use from ages 14-26. DNA was obtained from participants at age 21. Internalizing symptoms and antisocial behavior PRSs were created based on a genome-wide association study (GWAS) conducted by Benke et al. (2014) and Tielbeek et al. (2017), respectively. Parental monitoring and community disadvantage were assessed at age 12. Four classes of past year alcohol use were identified: (a) early-onset, increasing; (b) late-onset, moderate use; (c) low steady; and (d) early-onset, decreasing. In high community disadvantaged settings, participants with a higher internalizing symptoms PRS were more likely to be in the early-onset, decreasing class than the low steady class. When exposed to elevated community disadvantage, participants with a higher antisocial behavior PRS were more likely to be in the early-onset, increasing class than the early-onset, decreasing and late-onset, moderate use classes.


Asunto(s)
Negro o Afroamericano , Estudio de Asociación del Genoma Completo , Adolescente , Adulto , Negro o Afroamericano/genética , Consumo de Bebidas Alcohólicas , Trastorno de Personalidad Antisocial/genética , Niño , Humanos , Herencia Multifactorial , Adulto Joven
8.
Neural Plast ; 2018: 9803764, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29675039

RESUMEN

A variety of genetic approaches, including twin studies, linkage studies, and candidate gene studies, has established a firm genetic basis for addiction. However, there has been difficulty identifying the precise genes that underlie addiction liability using these approaches. This situation became especially clear in genome-wide association studies (GWAS) of addiction. Moreover, the results of GWAS brought into clarity many of the shortcomings of those early genetic approaches. GWAS studies stripped away those preconceived notions, examining genes that would not previously have been considered in the study of addiction, consequently creating a shift in our understanding. Most importantly, those studies implicated a class of genes that had not previously been considered in the study of addiction genetics: cell adhesion molecules (CAMs). Considering the well-documented evidence supporting a role for various CAMs in synaptic plasticity, axonal growth, and regeneration, it is not surprising that allelic variation in CAM genes might also play a role in addiction liability. This review focuses on the role of various cell adhesion molecules in neuroplasticity that might contribute to addictive processes and emphasizes the importance of ongoing research on CAM genes that have been implicated in addiction by GWAS.


Asunto(s)
Conducta Adictiva/genética , Moléculas de Adhesión Celular Neuronal/genética , Predisposición Genética a la Enfermedad , Plasticidad Neuronal/genética , Animales , Genotipo , Humanos , Neuronas/fisiología , Fenotipo , Polimorfismo de Nucleótido Simple
9.
Int J Neuropsychopharmacol ; 20(5): 403-409, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28031268

RESUMEN

Background: Opioid and dopamine systems play crucial roles in reward. Similarities and differences in the neural mechanisms of reward that are mediated by these 2 systems have remained largely unknown. Thus, in the present study, we investigated the differences in reward function in both µ-opioid receptor knockout mice and dopamine transporter knockout mice, important molecules in the opioid and dopamine systems. Methods: Mice were implanted with electrodes into the right lateral hypothalamus (l hour). Mice were then trained to put their muzzle into the hole in the head-dipping chamber for intracranial electrical stimulation, and the influences of gene knockout were assessed. Results: Significant differences are observed between opioid and dopamine systems in reward function. µ-Opioid receptor knockout mice exhibited enhanced intracranial electrical stimulation, which induced dopamine release. They also exhibited greater motility under conditions of "despair" in both the tail suspension test and water wheel test. In contrast, dopamine transporter knockout mice maintained intracranial electrical stimulation responding even when more active efforts were required to obtain the reward. Conclusions: The absence of µ-opioid receptor or dopamine transporter did not lead to the absence of intracranial electrical stimulation responsiveness but rather differentially altered it. The present results in µ-opioid receptor knockout mice are consistent with the suppressive involvement of µ-opioid receptors in both positive incentive motivation associated with intracranial electrical stimulation and negative incentive motivation associated with depressive states. In contrast, the results in dopamine transporter knockout mice are consistent with the involvement of dopamine transporters in positive incentive motivation, especially its persistence. Differences in intracranial electrical stimulation in µ-opioid receptor and dopamine transporter knockout mice underscore the multidimensional nature of reward.


Asunto(s)
Analgésicos Opioides/metabolismo , Dopamina/metabolismo , Área Hipotalámica Lateral/efectos de los fármacos , Área Hipotalámica Lateral/metabolismo , Receptores Opioides mu/deficiencia , Animales , Biofisica , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/deficiencia , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Estimulación Eléctrica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Motivación , Actividad Motora/efectos de los fármacos , Receptores Opioides mu/genética , Recompensa , Autoadministración , Factores de Tiempo
10.
Mol Med ; 22: 537-547, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27579475

RESUMEN

The cadherin 13 (CDH13) gene encodes a cell adhesion molecule likely to influence development and connections of brain circuits that modulate addiction, locomotion and cognition, including those that involve midbrain dopamine neurons. Human CDH13 mRNA expression differs by more than 80% in postmortem cerebral cortical samples from individuals with different CDH13 genotypes, supporting examination of mice with altered Cdh13 expression as models for common human variation at this locus. Constitutive cdh13 knockout mice display evidence for changed cocaine reward: shifted dose response relationship in tests of cocaine-conditioned place preference using doses that do not alter cocaine conditioned taste aversion. Reduced adult Cdh13 expression in conditional knockouts also alters cocaine reward in ways that correlate with individual differences in cortical Cdh13 mRNA levels. In control and comparison behavioral assessments, knockout mice display modestly-quicker acquisition of rotarod and water maze tasks, with a trend toward faster acquisition of 5 choice serial reaction time tasks that otherwise displayed no genotype-related differences. They display significant differences in locomotion in some settings, with larger effects in males. In assessments of brain changes that might contribute to these behavioral differences, there are selective alterations of dopamine levels, dopamine/metabolite ratios, dopaminergic fiber densities and mRNA encoding the activity dependent transcription factor npas4 in cerebral cortex of knockout mice. These novel data and previously reported human associations of CDH13 variants with addiction, individual differences in responses to stimulant administration and attention deficit hyperactivity disorder (ADHD) phenotypes suggest that levels of CDH13 expression, through mechanisms likely to include effects on mesocortical dopamine, influence stimulant reward and may contribute modestly to cognitive and locomotor phenotypes relevant to ADHD.

11.
Mol Med ; 21(1): 717-725, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26181631

RESUMEN

The receptor type protein tyrosine phosphatase D (PTPRD) gene encodes a cell adhesion molecule likely to influence development and connections of addiction-, locomotion- and sleep-related brain circuits in which it is expressed. The PTPRD gene harbors genome-wide association signals in studies of restless leg syndrome (Willis-Ekbom disease [WED]/restless leg syndrome [RLS]; p < 10-8) and addiction-related phenotypes (clusters of nearby single nucleotide polymorphisms [SNPs] with 10-2 > p > 10-8 associations in several reports). We now report work that seeks (a) association between PTPRD genotypes and expression of its mRNA in postmortem human brains and (b) RLS-related, addiction-related and comparison behavioral phenotypes in hetero- and homozygous PTPRD knockout mice. We identify associations between PTPRD SNPs and levels of PTPRD mRNA in human brain samples that support validity of mouse models with altered PTPRD expression. Knockouts display less behaviorally defined sleep at the end of their active periods. Heterozygotes move more despite motor weakness/impersistence. Heterozygotes display shifted dose-response relationships for cocaine reward. They display greater preference for places paired with 5 mg/kg cocaine and less preference for places paired with 10 or 20 mg/kg. The combined data provide support for roles for common, level-of-expression PTPRD variation in locomotor, sleep and drug reward phenotypes relevant to RLS and addiction. Taken together, mouse and human results identify PTPRD as a novel therapeutic target for RLS and addiction phenotypes.

12.
Behav Pharmacol ; 26(1-2): 167-79, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25485646

RESUMEN

Repeated administration of methamphetamine (METH) enhances acute locomotor responses to METH administered in the same context, a phenomenon termed as 'locomotor sensitization'. Although many of the acute effects of METH are mediated by its influences on the compartmentalization of dopamine, serotonin systems have also been suggested to influence the behavioral effects of METH in ways that are not fully understood. The present experiments examined serotonergic roles in METH-induced locomotor sensitization by assessing: (a) the effect of serotonin transporter (SERT; Slc6A4) knockout (KO) on METH-induced locomotor sensitization; (b) extracellular monoamine levels in METH-treated animals as determined by in-vivo microdialysis; and (c) effects of serotonin (5-HT) receptor antagonists on METH-induced behavioral sensitization, with focus on effects of the 5-HT1B receptor antagonist SB 216641 and a comparison with the 5-HT2 receptor antagonist ketanserin. Repeated METH administration failed to induce behavioral sensitization in homozygous SERT KO (SERT-/-) mice under conditions that produced substantial sensitization in wild-type or heterozygous SERT KO (SERT+/-) mice. The selective 5-HT1B antagonist receptor SB 216641 restored METH-induced locomotor sensitization in SERT-/- mice, whereas ketanserin was ineffective. METH-induced increases in extracellular 5-HT (5-HTex) levels were substantially reduced in SERT-/- mice, although SERT genotype had no effect on METH-induced increases in extracellular dopamine. These experiments demonstrate that 5-HT actions, including those at 5-HT1B receptors, contribute to METH-induced locomotor sensitization. Modulation of 5-HT1B receptors might aid therapeutic approaches to the sequelae of chronic METH use.


Asunto(s)
Benzamidas/farmacología , Metanfetamina/farmacología , Actividad Motora/efectos de los fármacos , Oxadiazoles/farmacología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Animales , Conducta Animal/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/administración & dosificación , Estimulantes del Sistema Nervioso Central/farmacología , Dopamina/metabolismo , Femenino , Ketanserina/farmacología , Masculino , Metanfetamina/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microdiálisis , Receptor de Serotonina 5-HT1B/efectos de los fármacos , Receptor de Serotonina 5-HT1B/metabolismo , Antagonistas del Receptor de Serotonina 5-HT1/farmacología , Antagonistas del Receptor de Serotonina 5-HT2/farmacología
13.
Behav Pharmacol ; 26(4): 345-52, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25794333

RESUMEN

Methylone (2-methylamino-1-[3,4-methylenedioxy-phenyl]propan-1-one), an amphetamine analog, has emerged as a popular drug of abuse worldwide. Methylone induces hyperthermia, which is thought to contribute toward the lethal consequences of methylone overdose. Methylone has been assumed to induce hyperthermic effects through inhibition of serotonin and/or dopamine transporters (SERT and DAT, respectively). To examine the roles of each of these proteins in methylone-induced toxic effects, we used SERT and DAT knockout (KO) mice and assessed the hyperthermic and lethal effects caused by a single administration of methylone. Methylone produced higher rates of lethal toxicity compared with other amphetamine analogs in wild-type mice. Compared with wild-type mice, lethality was significantly lower in DAT KO mice, but not in SERT KO mice. By contrast, only a slight diminution in the hyperthermic effects of methylone was observed in DAT KO mice, whereas a slight enhancement of these effects was observed in SERT KO mice. Administration of the selective D1 receptor antagonist SCH 23390 and the D2 receptor antagonist raclopride reduced methylone-induced hyperthermia, but these drugs also had hypothermic effects in saline-treated mice, albeit to a smaller extent than the effects observed in methylone-treated mice. In contradistinction to 3,4-methylenedioxymethamphetamine, which induces its toxicity through SERT and DAT, these data indicate that DAT, but not SERT, is strongly associated with the lethal toxicity produced by methylone, which did not seem to be dependent on the hyperthermic effects of methylone. DAT is therefore a strong candidate molecule for interventions aimed at preventing acute neurotoxic and lethal effects of methylone.


Asunto(s)
Estimulantes del Sistema Nervioso Central/toxicidad , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Fiebre/inducido químicamente , Metanfetamina/análogos & derivados , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Animales , Benzazepinas/farmacología , Temperatura Corporal/efectos de los fármacos , Temperatura Corporal/fisiología , Antagonistas de Dopamina/farmacología , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Femenino , Fiebre/tratamiento farmacológico , Fiebre/metabolismo , Fiebre/mortalidad , Masculino , Metanfetamina/toxicidad , Ratones Noqueados , Modelos Animales , Racloprida/farmacología , Receptores de Dopamina D1/antagonistas & inhibidores , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética
14.
Behav Pharmacol ; 25(2): 158-65, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24557322

RESUMEN

We investigated whether pretreatment with the neurotransmitter/neuromodulator agmatine (decarboxylated L-arginine) affected methamphetamine (METH)-induced hyperlocomotion and stereotypy in male ICR mice. Agmatine pretreatment alone had no effects on locomotion or stereotypy, but it produced a dose-dependent attenuation of locomotion and the total incidence of stereotyped behavior induced by a low dose of METH (5 mg/kg). The stereotypy induced by this dose was predominantly characterized by stereotyped sniffing. By contrast, agmatine did not affect the total incidence of stereotypy induced by a higher dose of METH (10 mg/kg). However, the nature of stereotypy induced by this dose of METH was substantially altered; agmatine pretreatment significantly reduced stereotyped biting but significantly increased stereotyped sniffing and persistent locomotion. Agmatine pretreatment therefore appears to produce a rightward shift in the dose-response curve for METH. Pretreatment of mice with piperazine-1-carboxamidine (a putative agmatinase inhibitor) had no effect on locomotion or stereotypy induced by a low dose of METH, suggesting that endogenous agmatine may not regulate the METH action.


Asunto(s)
Agmatina/farmacología , Estimulantes del Sistema Nervioso Central/efectos adversos , Metanfetamina/efectos adversos , Neurotransmisores/farmacología , Agitación Psicomotora/tratamiento farmacológico , Conducta Estereotipada/efectos de los fármacos , Trastornos Relacionados con Anfetaminas/tratamiento farmacológico , Animales , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Guanidinas/farmacología , Masculino , Ratones , Ratones Endogámicos ICR , Piperazinas/farmacología , Agitación Psicomotora/etiología , Factores de Tiempo , Ureohidrolasas/antagonistas & inhibidores
15.
Addict Biol ; 19(3): 343-53, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-22780223

RESUMEN

We have previously shown that a haplotype associated with decreased NrCAM expression in brain is protective against addiction vulnerability for polysubstance abuse in humans and that Nrcam knockout mice do not develop conditioned place preferences for morphine, cocaine or amphetamine. In order to gain insight into NrCAM involvement in addiction vulnerability, which may involve specific neural circuits underlying behavioral characteristics relevant to addiction, we evaluated several behavioral phenotypes in Nrcam knockout mice. Consistent with a potential general reduction in motivational function, Nrcam knockout mice demonstrated less curiosity for novel objects and for an unfamiliar conspecific, showed also less anxiety in the zero maze. Nrcam heterozygote knockout mice reduced alcohol preference and buried fewer marbles in home cage. These observations provide further support for a role of NrCAM in substance abuse including alcoholism vulnerability, possibly through its effects on behavioral traits that may affect addiction vulnerability, including novelty seeking, obsessive compulsion and responses to aversive or anxiety-provoking stimuli. Additionally, in order to prove glutamate homeostasis hypothesis of addiction, we analyzed glutamatergic molecules regulated by NRCAM expression. Glutaminase appears to be involved in NrCAM-related molecular pathway in two different tissues from human and mouse. An inhibitor of the enzyme, prolyl-leucyl-glycinamide, treatment produced, at least, some of the phenotypes of mice shown in alcohol preference and in anxiety-like behavior. Thus, NrCAM could affect addiction-related behaviors via at least partially modulation of some glutamatergic pathways and neural function in brain.


Asunto(s)
Conducta Adictiva/fisiopatología , Moléculas de Adhesión Celular/fisiología , Adaptación Psicológica/efectos de los fármacos , Consumo de Bebidas Alcohólicas/fisiopatología , Analgésicos Opioides/farmacología , Animales , Ansiedad/fisiopatología , Depresores del Sistema Nervioso Central/farmacología , Condicionamiento Psicológico/efectos de los fármacos , Etanol/farmacología , Conducta Exploratoria/efectos de los fármacos , Hormona Inhibidora de la Liberación de MSH/farmacología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Noqueados , Morfina/farmacología , Tiempo de Reacción/efectos de los fármacos , Conducta Social
16.
Prev Sci ; 15(6): 831-40, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24178584

RESUMEN

A variety of school-based, universal preventive interventions have been developed to address behavioral and mental health problems. Unfortunately, few have been evaluated within the context of randomized controlled trials with long-term follow-up. Even fewer still have examined the potential genetic factors that may drive differential impact of the intervention. In the present analysis, we examine the extent to which the longitudinal effects of two elementary school-based interventions were moderated by the brain-derived neurotrophic factor (BDNF) gene, which has been linked with aggression and impulsive behaviors. The sample included 678 urban, primarily African American children who were randomly assigned along with their teachers to one of three first grade classroom conditions: classroom-centered (CC) intervention, Family School Partnership (FSP), or a control condition. The teacher ratings of the youth's aggressive and impulsive behavior were obtained at baseline and in grades 6-12. Single-nucleotide polymorphisms (SNPs) from the BDNF gene were extracted from the genome-wide data. Longitudinal latent trait-state-error models indicated a significant interaction between a particular profile of the BDNF SNP cluster (46 % of sample) and CC intervention on impulsivity (ß = -.27, p < .05). A similar interaction was observed for the BDNF SNP cluster and the CC intervention on aggression (ß = -.14, p < .05). The results suggest that the impacts of preventive interventions in early elementary school on late adolescent outcomes of impulsivity and aggression can be potentially modified by genetic factors, such as BDNF. However, replication of these results is necessary before firm conclusions can be drawn.


Asunto(s)
Agresión , Factor Neurotrófico Derivado del Encéfalo/genética , Conducta Infantil , Promoción de la Salud/métodos , Conducta Impulsiva , Conducta de Reducción del Riesgo , Servicios de Salud Escolar , Adolescente , Niño , Conducta Infantil/etnología , Femenino , Humanos , Masculino , Polimorfismo de Nucleótido Simple , Psiquiatría Preventiva , Población Urbana
17.
Biochem Biophys Res Commun ; 432(3): 526-32, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23410751

RESUMEN

The vesicular monoamine transporter 2 (VMAT2) translocates monoamine neurotransmitters from the neuronal cytoplasm into synaptic vesicles. Since VMAT2-/- mice die within a few days of birth, it is difficult to analyze the detailed VMAT2 functions using these mice. In this study, we generated human VMAT2 transgenic mice that expressed VMAT2 in noradrenergic neurons with the aim to rescue the lethality of VMAT2 deletion. The expression of human VMAT2 in noradrenergic neurons extended the life of VMAT2-/- mice for up to three weeks, and these mice showed severe growth deficiency compared with VMAT2+/+ mice. These results may indicate that VMAT2 expressed in noradrenergic neurons has crucial roles in survival during the first several weeks after birth, and VMAT2 functions in other monoaminergic systems could be required for further extended survival. Although VMAT2 rescue in noradrenergic neurons did not eliminate the increased morbidity and lethality associated with VMAT2 deletion, the extension of the lifespan in VMAT2 transgenic mice will enable behavioral, pharmacological and pathophysiological studies of VMAT2 function.


Asunto(s)
Neuronas Adrenérgicas/metabolismo , Longevidad/genética , Proteínas de Transporte Vesicular de Monoaminas/fisiología , Animales , Ataxia/genética , Marcha/genética , Ratones , Ratones Noqueados , Actividad Motora/genética , Transgenes , Proteínas de Transporte Vesicular de Monoaminas/genética
18.
Int J Neuropsychopharmacol ; 16(1): 151-62, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22217458

RESUMEN

Most classical preclinical tests to predict antidepressant activity were initially developed to detect compounds that influenced noradrenergic and/or serotonergic activity, in accordance with the monoaminergic hypothesis of depression. However, central opioid systems are also known to influence the pathophysiology of depression. While the tail suspension test (TST) is very sensitive to several types of antidepressant, the traditional form of scoring the TST does not distinguish between different modes of action. The present study was designed to compare the behavioural effects of classical noradrenergic and/or serotonergic antidepressants in the TST with those of opioids. We developed a sampling technique to differentiate between behaviours in the TST, namely, curling, swinging and immobility. Antidepressants that inhibit noradrenaline and/or serotonin re-uptake (imipramine, venlafaxine, duloxetine, desipramine and citalopram) decreased the immobility of mice, increasing their swinging but with no effect on their curling behaviour. No differences were observed between antidepressants that act on noradrenergic or serotoninergic transmission. While opioid compounds also decreased the immobility of the mice [morphine, codeine, levorphanol, (-)-methadone, (±)-tramadol and (+)-tramadol], they selectively increased curling behaviour. Blocking opioid receptors with naloxone prevented the antidepressant-like effect of codeine, and µ-opioid receptor knockout decreased normal curling behaviour and blocked (±)-tramadol-induced curling, further demonstrating the reliability and validity of this approach. These results show that at least two behaviourally distinct processes occur in the TST, highlighting the antidepressant-like effects of opioids evident in this test. Furthermore, our data suggest that swinging and curling behaviours are mediated by enhanced monoamine and opioid neurotransmission, respectively.


Asunto(s)
Analgésicos Opioides/farmacología , Antidepresivos/farmacología , Suspensión Trasera/fisiología , Actividad Motora/fisiología , Animales , Femenino , Suspensión Trasera/métodos , Suspensión Trasera/psicología , Pérdida de Tono Postural/efectos de los fármacos , Pérdida de Tono Postural/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Receptores Opioides mu/deficiencia , Receptores Opioides mu/genética
19.
Front Psychiatry ; 14: 1031283, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37139308

RESUMEN

Substance use disorders provide challenges for development of effective medications. Use of abused substances is likely initiated, sustained and "quit" by complex brain and pharmacological mechanisms that have both genetic and environmental determinants. Medical utilities of prescribed stimulants and opioids provide complex challenges for prevention: how can we minimize their contribution to substance use disorders while retaining medical benefits for pain, restless leg syndrome, attention deficit hyperactivity disorder, narcolepsy and other indications. Data required to support assessments of reduced abuse liability and resulting regulatory scheduling differs from information required to support licensing of novel prophylactic or therapeutic anti-addiction medications, adding further complexity and challenges. I describe some of these challenges in the context of our current efforts to develop pentilludin as a novel anti-addiction therapeutic for a target that is strongly supported by human and mouse genetic and pharmacologic studies, the receptor type protein tyrosine phosphatase D (PTPRD).

20.
Int J Neurosci ; 122(7): 373-80, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22376027

RESUMEN

BACKGROUND: Dopamine transporter (DAT) homozygous knockout (DAT(-/-)) mice have a 10-fold higher extracellular (DA) concentration in the caudate-putamen and nucleus accumbens than do wildtype (DAT(+/+)) mice, but show reduced presynaptic DA synthesis and fewer postsynaptic D(2) receptors. One aspect of neurotransmission involves DA binding to postsynaptic D(2)-like receptors coupled to cytosolic phospholipase A(2) (cPLA(2)), which releases the second messenger, arachidonic acid (AA), from synaptic membrane phospholipid. We hypothesized that tonic overactivation of D(2)-like receptors in DAT(-/-) mice due to the excess DA would not increase brain AA signaling, because of compensatory downregulation of postsynaptic DA signaling mechanisms. METHODS: [1-(14)C]AA was infused intravenously for 3 min in unanesthetized DAT(+/+), heterozygous (DAT(+/-)), and DAT(-/-) mice. AA incorporation coefficients k* and rates J(in), markers of AA metabolism and signaling, were imaged in 83 brain regions using quantitative autoradiography; brain cPLA(2)-IV activity also was measured. RESULTS: Neither k* nor J(in) for AA in any brain region, or brain cPLA(2)-IV activity, differed significantly among DAT(-/-), DAT(+/-), and DAT(+/+) mice. CONCLUSIONS: These results differ from reported increases in k* and J(in) for AA, and in brain cPLA(2) expression, in serotonin reuptake transporter (5-HTT) knockout mice, and suggest that postsynaptic dopaminergic neurotransmission mechanisms involving AA are downregulated despite elevated DA in DAT(-/-) mice.


Asunto(s)
Ácido Araquidónico/metabolismo , Encéfalo/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/deficiencia , Transducción de Señal/genética , Análisis de Varianza , Animales , Autorradiografía , Peso Corporal/genética , Encéfalo/enzimología , Mapeo Encefálico , Isótopos de Carbono/metabolismo , Regulación hacia Abajo/genética , Ácidos Grasos/sangre , Masculino , Ratones , Ratones Noqueados , Fosfolipasas A2 , Potasio/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA