Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Addict Sci Clin Pract ; 19(1): 56, 2024 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-39061093

RESUMO

BACKGROUND: Despite continuing advancements in treatments for opioid use disorder (OUD), continued high rates of relapse indicate the need for more effective approaches, including novel pharmacological interventions. Glucagon-like peptide 1 receptor agonists (GLP-1RA) provide a promising avenue as a non-opioid medication for the treatment of OUD. Whereas GLP-1RAs have shown promise as a treatment for alcohol and nicotine use disorders, to date, no controlled clinical trials have been conducted to determine if a GLP-1RA can reduce craving in individuals with OUD. The purpose of the current protocol was to evaluate the potential for a GLP-1RA, liraglutide, to safely and effectively reduce craving in an OUD population in residential treatment. METHOD: This preliminary study was a randomized, double-blinded, placebo-controlled clinical trial designed to test the safety and efficacy of the GLP-1RA, liraglutide, in 40 participants in residential treatment for OUD. Along with taking a range of safety measures, efficacy for cue-induced craving was evaluated prior to (Day 1) and following (Day 19) treatment using a Visual Analogue Scale (VAS) in response to a cue reactivity task during functional near-infrared spectroscopy (fNIRS) and for craving. Efficacy of treatment for ambient craving was assessed using Ecological Momentary Assessment (EMA) prior to (Study Day 1), across (Study Days 2-19), and following (Study Days 20-21) residential treatment. DISCUSSION: This manuscript describes a protocol to collect clinical data on the safety and efficacy of a GLP-1RA, liraglutide, during residential treatment of persons with OUD, laying the groundwork for further evaluation in a larger, outpatient OUD population. Improved understanding of innovative, non-opioid based treatments for OUD will have the potential to inform community-based interventions and health policy, assist physicians and health care professionals in the treatment of persons with OUD, and to support individuals with OUD in their effort to live a healthy life. TRIAL REGISTRATION: ClinicalTrials.gov: NCT04199728. Registered 16 December 2019, https://clinicaltrials.gov/study/NCT04199728?term=NCT04199728 . PROTOCOL VERSION: 10 May 2023.


Assuntos
Fissura , Sinais (Psicologia) , Avaliação Momentânea Ecológica , Receptor do Peptídeo Semelhante ao Glucagon 1 , Liraglutida , Transtornos Relacionados ao Uso de Opioides , Humanos , Fissura/efeitos dos fármacos , Método Duplo-Cego , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Liraglutida/uso terapêutico , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Feminino , Masculino , Adulto , Tratamento Domiciliar/métodos , Pessoa de Meia-Idade , Ensaios Clínicos Controlados Aleatórios como Assunto
2.
Brain Res Bull ; 192: 142-155, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36410565

RESUMO

INTRODUCTION: Sleep disturbances are prominent in drug use disorders, including those involving opioids in both humans and animals. Recent studies have shown that administration of liraglutide, a glucagon-like peptide-1 agonist, significantly reduces heroin taking and seeking in rats. In an effort to further understand the action of this substance on physiological functions and to evaluate safety issues for its potential clinical use, the aim of the present study was to determine whether the dose of liraglutide found effective in reducing responding for an opioid also could improve sleep in drug-naïve rats. METHODS: Using a within-subjects design, adult male rats chronically implanted with EEG and EMG electrodes received subcutaneous injection of saline or 0.06, 0.10, 0.30 or 0.60 mg/kg liraglutide. The 0.10 and 0.30 mg/kg doses are known to be most effective in reducing responding for heroin in rats at light or dark onset during a 12:12 h light-dark cycle (0.10 mg/kg for taking and seeking, 0.30 mg/kg for seeking). EEG and EMG were recorded across the 24 h period following each injection. RESULTS: After both dark and light onset injections, liraglutide dose-dependently decreased wakefulness and increased non-rapid eye movement (NREM) sleep except at the lowest dose. The bout length of wakefulness and NREM sleep were decreased and increased, respectively. Whether administered at light or dark onset, the above alterations occurred primarily during the dark period (i.e., during the active period). The animals' body weight was decreased after liraglutide treatments as expected since it is clinically used for the treatment of obesity. CONCLUSION: These data indicate that liraglutide, at doses known to reduce responding for heroin and fentanyl, also increases NREM sleep, suggesting that the increase in sleep may contribute to the protective effects of liraglutide and may promote overall general health.


Assuntos
Receptor do Peptídeo Semelhante ao Glucagon 1 , Liraglutida , Humanos , Ratos , Masculino , Animais , Liraglutida/farmacologia , Eletroencefalografia , Sono , Vigília
3.
Brain Res Bull ; 191: 48-60, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36228871

RESUMO

As a drug of abuse tightens its hold on addicted individuals, aspects of life that once brought pleasure lose their appeal while attention and motivation are turned toward acquiring drug. In a rodent model of self-administration and reward devaluation, we previously showed that animals that suppress intake of a drug-paired saccharin cue show greater addiction-like behaviors, as well as increased gene-expression of elements of the corticotropin releasing factor (CRF) pathway in the prefrontal cortex (mPFC), hippocampus (Hipp), and ventral tegmental area (VTA). In the present study, we explored whether the observed differences in components of the CRF signaling pathway were a function of self-administration or devaluation of the cue. Moreover, as an increasing body of work illustrates, functional and molecular hemispheric differences in reward pathway components, we examined whether these CRF pathway components exhibited hemispheric differences in response to heroin administration. Over a period of 7 trials, 30 male rats received brief access to saccharin followed by passive (IP) injection of heroin (n = 20) or saline (n = 10). Saccharin intakes between large saccharin suppressors (LS; 12 animals) and small suppressors (SS; 8 animals) were statistically different after trial 1 and separated further with ensuing trials. We then assessed gene expression for components of the CRF pathway in the mPFC, Hipp, VTA, Amygdala, and nucleus accumbens (NAc). Within the Hipp, LS showed greater expression of CRF binding protein (CRFbp). No differences were observed in the mPFC, VTA, NAc or Amygdala. Several hemisphere differences in CRF signaling pathway genes were detected. These findings indicate that avoidance of the experimenter delivered heroin-paired saccharin cue, do not recapitulate findings observed for avoidance of the iv self-administered heroin-paired saccharin cue, at least in terms of the expression of genes within the CRF pathway, and provide further evidence that consideration should be given to hemisphere differences when exploring molecular phenomena.


Assuntos
Heroína , Sacarina , Ratos , Animais , Masculino , Heroína/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Sinais (Psicologia) , Hipocampo/metabolismo
4.
Brain Res Bull ; 191: 107-120, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36223840

RESUMO

Addiction is a disease of brain-reward circuitry whereby attention, motivation, memory and emotional systems become enslaved to the goal of seeking and acquiring drug, instead of responding to the natural rewards for which these systems evolved. At the intersection of reward/limbic structures, the medial prefrontal cortex (mPFC) receives and consolidates signals regarding environment and orchestrates the most appropriate response (i.e., decision-making and attention). As such, mPFC function plays a critical role in the vulnerability or resilience to drug addiction. In our model of drug-induced reward devaluation, an outbred group of Sprague-Dawley rats parsed into two distinct drug-taking phenotypes: those, referred to as small suppressors (SS) that readily ingest a heroin-paired sweet cue and then take little drug, and those, referred to large suppressors (LS), that avoid the heroin-paired cue, but then respond greatly for the drug of abuse. In the present study, we analyzed the mPFC transcriptome of rats from these divergent groups to discover differences in gene expression that underlie these distinct phenotypes. Genes found to be differentially expressed were those associated with schizophrenia and dopamine signaling, signal transduction, development and synaptic plasticity. These genes may underlie the circumstance whereby some individuals succumb to addiction, while others do not, and may reveal new pharmacological targets for the treatment of drug addiction.


Assuntos
Heroína , Córtex Pré-Frontal , Animais , Ratos , Heroína/metabolismo , RNA-Seq , Ratos Sprague-Dawley , Córtex Pré-Frontal/metabolismo , Fenótipo , Autoadministração
5.
Brain Res Bull ; 189: 155-162, 2022 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-36031011

RESUMO

Opioid Use Disorder (OUD) is a chronic relapsing disorder that has severe negative impacts on the individual, the family, and the community at large. In 2021, opioids contributed to nearly 70% of all drug overdose deaths in the United States. This number of opioid related deaths coincides with a significant rise in the use of fentanyl, a synthetic opioid that is 150 times more potent than morphine. Furthermore, this overdose trend has spared no demographic and costs the nation an estimated $51.2 billion annually. Thus, it is imperative to better understand the underlying mechanisms of OUD in an effort to identify new treatment targets. Using animal models, studies have shown that rats readily self-administer heroin and increase seeking following exposure to cues for drug, the drug itself, or stress. We have shown that treatment with the glucagon-like peptide-1 receptor (GLP-1R) agonist, liraglutide, can reduce heroin taking and seeking behavior in rats. Therefore, using our rodent model, we established a fentanyl self-administration paradigm to test whether acute treatment with the GLP-1R agonist also can reduce fentanyl seeking in fentanyl experienced rats. The results showed that rats readily self-administered fentanyl (2.5 ug/kg) intravenously, with marked individual differences in drug taking behavior. As with other drugs of abuse tested, rats exhibited high seeking behavior when challenged with a drug-related cue or, after a period of extinction, the drug itself. Here, acute treatment with the GLP-1R agonist, liraglutide (0.3 mg/kg s.c.), was found to attenuate both cue-induced fentanyl seeking and drug-induced reinstatement of fentanyl seeking with the same efficacy as the currently approved partial opioid agonist, buprenorphine. Taken together, these data suggest that a known satiety signal, GLP-1, may serve as an effective non-opioid alternative for the treatment of OUD.


Assuntos
Buprenorfina , Transtornos Relacionados ao Uso de Opioides , Analgésicos Opioides/farmacologia , Animais , Sinais (Psicologia) , Fentanila/farmacologia , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Heroína/farmacologia , Liraglutida , Ratos , Autoadministração
6.
Brain Res Bull ; 189: 163-173, 2022 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-36038016

RESUMO

Opioid use disorder (OUD), like other substance use disorders (SUDs), is widely understood to be a disorder of persistent relapse. Despite the use of three FDA-approved medications for OUD, typically in conjunction with behavioral treatments, relapse rates remain unacceptably high. Whereas medication assisted therapy (MAT) reduces the risk of opioid overdose mortality, the benefits of MAT are negated when people discontinue the medications. Currently approved medications present barriers to efficient use, including daily visits to a treatment center or work restrictions. With spiking increases in opioid relapse and death, it is imperative to identify new treatments that can reduce the risk of relapse. Recent evidence suggests that glucagon-like peptide-1 receptor agonists (GLP-1RAs), currently FDA-approved to treat obesity and type two diabetes, may be promising candidates to reduce relapse. GLP-1RAs have been shown to reduce relapse in rats, whether elicited by cues, drug, and/or stress. However, GLP-1RAs also can cause gastrointestinal malaise, and therefore, in humans, the medication typically is titrated up to full dose when initiating treatment. Here, we used a rodent model to test whether cue- and drug-induced heroin seeking can be reduced by the GLP-1RA, liraglutide, when the dose is titrated across the abstinence period and prior to test. The results show this titration regimen is effective in reducing both cue-induced heroin seeking and drug-induced reinstatement of heroin seeking, particularly in rats with a history of high drug-taking. Importantly, this treatment regimen had no effect on either circulating glucose or insulin. GLP-1RAs, then, appear strong candidates for the non-opioid prevention of relapse to opioids.


Assuntos
Diabetes Mellitus Tipo 2 , Liraglutida , Animais , Sinais (Psicologia) , Diabetes Mellitus Tipo 2/tratamento farmacológico , Peptídeo 1 Semelhante ao Glucagon , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Glucose , Heroína/farmacologia , Humanos , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Insulina , Liraglutida/farmacologia , Liraglutida/uso terapêutico , Ratos , Recidiva
7.
Behav Pharmacol ; 33(5): 364-378, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35695511

RESUMO

Substance use disorder is challenging to treat due to its relapsing nature. In the last decade, opioid use disorder has been a threat to public health, being declared an epidemic by the Centers for Disease Control and Prevention. This is a tragic situation, considering there currently are only three effective, yet not ideal, treatments to prevent relapse to opioids. Recent research has shown that hormones that modulate hunger and satiety also can modulate motivated behavior for drugs of abuse. For example, the short-acting analog of glucagon-like peptide-1 (GLP-1), an incretin hormone that regulates homeostatic feeding, has been shown to reduce responding for rewarding stimuli such as food, cocaine, heroin, and nicotine when administered over several days or weeks. This may serve as an effective adjuvant during treatment; however, whether it would be effective when used acutely to bridge a patient between cessation of use and onset of medication for the treatment of an opioid addiction is unknown. Here, we tested the acute effects of the longer acting GLP-1 analog, liraglutide, on heroin-seeking. In rats with heroin self-administration experience, we found that subcutaneous administration of an acute dose of 0.3-mg/kg liraglutide was effective in preventing drug-seeking after exposure to three major precipitators: drug-associated cues, stress (yohimbine-induced), and the drug itself. Finally, we confirmed that the reduction in drug-seeking is not due to a locomotor impairment, as liraglutide did not significantly alter performance in a rotarod test. As such, acute use of GLP-1 analogs may serve as a new and effective nonopioid bridge to treatment.


Assuntos
Sinais (Psicologia) , Heroína , Animais , Comportamento de Procura de Droga , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Heroína/farmacologia , Liraglutida/farmacologia , Ratos , Autoadministração
8.
Addict Biol ; 27(2): e13117, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34802173

RESUMO

Drug addiction is a chronic brain disease characterized by the uncontrolled use of a substance. Due to its relapsing nature, addiction is difficult to treat, as individuals can relapse following even long periods of abstinence and, it is during this time, that they are most vulnerable to overdose. In America, opioid overdose has been increasing for decades, making finding new treatments to help patients remain abstinent and prevent overdose deaths imperative. Recently, glucagon-like peptide-1 (GLP-1) receptor agonists have shown promise in reducing motivated behaviours for drugs of abuse. In this study, we test the effectiveness of the GLP-1 analogue, liraglutide (LIR), in reducing heroin addiction-like behaviour, and the potential side effects associated with the treatment. We show that daily treatment with LIR (0.1 mg/kg sc) increases the latency to take heroin, reduces heroin self-administration, prevents escalation of heroin self-administration and reduces drug-induced reinstatement of heroin-seeking behaviour in rats. A 1-h pretreatment time, however, was too short to reduce cue-induced seeking in our study. Moreover, we showed that, while LIR (0.1, 0.3, 0.6 and 1.0 mg/kg sc) supported conditioned taste avoidance of a LIR-paired saccharin cue, it did not elicit intake of the antiemetic kaolin in heroin-naïve or heroin-experienced rats. Further, 0.1 mg/kg LIR did not produce great disruptions in food intake or body weight. Overall, the data show that LIR is effective in reducing heroin taking and heroin seeking at doses that do not cause malaise and have a modest effect on food intake and body weight gain.


Assuntos
Receptor do Peptídeo Semelhante ao Glucagon 1 , Dependência de Heroína , Liraglutida , Animais , Sinais (Psicologia) , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Heroína/farmacologia , Dependência de Heroína/tratamento farmacológico , Liraglutida/farmacologia , Ratos , Autoadministração
9.
Behav Pharmacol ; 32(4): 265-277, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33229892

RESUMO

Opioid use disorder (OUD) causes the death of nearly 130 Americans daily. It is evident that new avenues for treatment are needed. To this end, studies have reported that 'satiety' agents such as the glucagon-like peptide-1 receptor (GLP-1R) agonist, exendin-4 (Ex-4), decreases responding for addictive drugs such as cocaine, nicotine, alcohol, and oxycodone, but no work has been done with heroin. In this study, we used a reward devaluation model in which rats avoid ingesting a saccharin solution that predicts drug availability to test the effects of 2.4 µg/kg Ex-4 on responding for a natural reward cue (i.e., saccharin) and on cue- and drug-induced heroin seeking. The results showed that treatment with Ex-4 during the 16-day abstinence period and on the test day decreased cue-induced heroin seeking. Drug-induced heroin seeking also was reduced by Ex-4, but only when using a 1 h, but not a 6 h, pretreatment time. Treatment with Ex-4 did not alter intake of the saccharin cue when the drug was on board, but a history of treatment with Ex-4 increased acceptance of the saccharin cue in later extinction trials. Finally, treatment with Ex-4 did not alter body weight, but was associated with increased Orexin 1 receptor (OX1) mRNA expression in the nucleus accumbens shell. Taken together, these findings are the first to show that treatment with a GLP-1R agonist can reduce both cue-induced seeking and drug-induced reinstatement of heroin seeking. As such, a GLP-1R agonist may serve as an effective treatment for OUD in humans.


Assuntos
Comportamento Apetitivo/efeitos dos fármacos , Exenatida/farmacologia , Heroína/farmacologia , Núcleo Accumbens/metabolismo , Receptores de Orexina/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Descoberta de Drogas , Regulação da Expressão Gênica/efeitos dos fármacos , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Hipoglicemiantes/farmacologia , Entorpecentes/farmacologia , Ratos , Resposta de Saciedade/efeitos dos fármacos
10.
J Neurosci Methods ; 343: 108857, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32652184

RESUMO

Animal models have significantly contributed to the understanding of reward-related behaviors, such as in Substance Use Disorder research. One of the most heavily utilized paradigms to date is conditioned place preference (CPP). However, CPP is limited by non-contingent exposure. Our new method advances this classic method by utilizing its benefits and simultaneously diminishing its limitations. We used a traditional 3-compartment CPP apparatus, where each chamber differs by both visual and tactile contexts. We restructured the apparatus allowing for insertion of bottles so that mice could orally self-administer sucrose or morphine-containing solutions in a specific context. Our results show that mice who self-administer sucrose or morphine show a place preference for the sucrose- or morphine-paired chamber. This place preference lasts for 21 d in sucrose-treated, but not morphine-treated mice. Additionally, we found that that mice will drink more water in the morphine-paired context during extinction tests. This model combines the distinct contextual cues associated with conditioned place preference and combines them with voluntary self-administration, thus enabling researchers to measure behavior using a model that incorporates spatial memory involved in affective states, while also providing a quantifiable readout of context/environment-specific drug seeking. In conclusion, we combined CPP and voluntary intake to establish a novel technique to assess not only preference for a context associated with rewarding stimuli (natural or drug), but also seeking, retention, and locomotor activity. This model can be further utilized to examine other drugs of abuse, extinction training, other learning models, or to allow for the assessment of neurobiological manipulations.


Assuntos
Comportamento de Procura de Droga , Recompensa , Animais , Condicionamento Clássico , Sinais (Psicologia) , Camundongos , Morfina/farmacologia
11.
Psychopharmacology (Berl) ; 235(10): 3005-3015, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30178302

RESUMO

RATIONALE: Drug addiction is a complex disease that is impacted by numerous factors. One such factor, time of day, influences drug intake, but there have been no investigations of how time of day affects the amount of drug taken and the development of addiction-like behavior. Previous data from our group show circadian disruption in rats given access to heroin during the light phase, which is important because circadian disruption, itself, can increase drug intake. Thus, the goal of this experiment was to determine how time of day of access affects heroin self-administration and the development of addiction-like behaviors including escalation of heroin intake, willingness to work for heroin on a progressive ratio schedule of reinforcement, seeking during extinction, incubation of seeking, and reinstatement of heroin-seeking behavior. MATERIALS AND METHODS: Male Sprague Dawley rats were given the opportunity to self-administer heroin for 6 h per trial during the second half of either the light or dark phase for 18 trials, including one progressive ratio challenge. Rats then underwent 14 days of abstinence, with a 5-h extinction test occurring on both the first and the 14th days of abstinence. The second extinction test was followed by a heroin prime and 1 h of reinstatement testing. On the following day, a subset of rats were tested in an additional extinction test where rats were tested either at the same time of the day as their previous self-administration sessions or during the opposite light/dark phase. RESULTS: Relative to Light Access rats, Dark Access rats took more heroin, exhibited more goal-directed behavior, exhibited more seeking during the dark phase, failed to extinguish seeking during the 5-h extinction test in the dark phase, and exhibited greater incubation of heroin seeking following abstinence. However, Dark Access rats did not escalate drug taking over trials, work harder for drug, or seek more during drug-induced reinstatement than Light Access rats. CONCLUSIONS: These results show that time of access to heroin affects overall heroin intake and seeking in extinction, but does not affect other addiction-like behaviors in rats.


Assuntos
Comportamento Aditivo/psicologia , Ritmo Circadiano/fisiologia , Extinção Psicológica/efeitos dos fármacos , Dependência de Heroína/psicologia , Heroína/administração & dosagem , Animais , Modelos Animais de Doenças , Masculino , Ratos , Ratos Sprague-Dawley , Reforço Psicológico , Autoadministração
12.
Neuropharmacology ; 139: 26-40, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29964093

RESUMO

Environmental factors profoundly affect the addictive potential of drugs of abuse and may also modulate the neuro-anatomical/neuro-chemical impacts of uncontrolled drug use and relapse propensity. This study examined the impact of environmental enrichment on heroin self-administration, addiction-related behaviors, and molecular processes proposed to underlie these behaviors. Male Sprague-Dawley rats in standard and enriched housing conditions intravenously self-administered similar amounts of heroin over 14 days. However, environmental enrichment attenuated progressive ratio, extinction, and reinstatement session responding after 14 days of enforced abstinence. Molecular mechanisms, namely DNA methylation and gene expression, are proposed to underlie abstinence-persistent behaviors. A global reduction in methylation is reported to coincide with addiction, but no differences in total genomic methylation or repeat element methylation were observed in CpG or non-CpG (CH) contexts across the mesolimbic circuitry as assessed by multiple methods including whole genome bisulfite sequencing. Immediate early gene expression associated with drug seeking, taking, and abstinence also were examined. EGR1 and EGR2 were suppressed in mesolimbic regions with heroin-taking and environmental enrichment. Site-specific methylation analysis of EGR1 and EGR2 promoter regions using bisulfite amplicon sequencing (BSAS) revealed hypo-methylation in the EGR2 promoter region and EGR1 intragenic CpG sites with heroin-taking and environmental enrichment that was associated with decreased mRNA expression. Taken together, these findings illuminate the impact of drug taking and environment on the epigenome in a locus and gene-specific manner and highlight the need for positive, alternative rewards in the treatment and prevention of drug addiction.


Assuntos
Meio Ambiente , Dependência de Heroína/metabolismo , Dependência de Heroína/terapia , Animais , Ilhas de CpG , Metilação de DNA/efeitos dos fármacos , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Epigênese Genética/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Heroína/administração & dosagem , Abrigo para Animais , Masculino , Entorpecentes/administração & dosagem , RNA Mensageiro/metabolismo , Distribuição Aleatória , Ratos Sprague-Dawley , Reforço Psicológico , Autoadministração
13.
Brain Res Bull ; 138: 80-87, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28899794

RESUMO

Rats avoid intake of an otherwise palatable taste cue when paired with drugs of abuse (Grigson and Twining, 2002). In male rats, avoidance of drug-paired taste cues is associated with conditioned blunting of dopamine in the nucleus accumbens (Grigson and Hajnal, 2007), conditioned elevation in circulating corticosterone (Gomez et al., 2000), and greater avoidance of the drug-paired cue predicts greater drug-taking (Grigson and Twining, 2002). While female rats generally are more responsive to drug than male rats, in this self-administration model, female rats consume more of a cocaine-paired saccharin cue and take less drug than males (Cason and Grigson, 2013). What is not known, however, is whether the same is true when a saccharin cue predicts availability of an opiate, particularly when the amount of drug experienced is held constant via passive administration by the experimenter. Here, avoidance of a saccharin cue was evaluated following pairings with experimenter delivered cocaine or morphine in male and female rats. Results showed that males and females avoided intake of a taste cue when paired with experimenter administered morphine or cocaine, and individual differences emerged whereby some male and female rats exhibited greater avoidance of the drug-paired cue than others. Female rats did not drink more of the saccharin cue than males when paired with morphine in Experiment 1, however, they did drink more of the saccharin cue than male rats when paired with cocaine in Experiment 2. While no pattern with estrous cycle emerged, avoidance of the cocaine-paired cue, like avoidance of a morphine-paired cue (Gomez et al., 2000), was associated with a conditioned elevation in corticosterone in both male and female rats.


Assuntos
Aprendizagem da Esquiva/efeitos dos fármacos , Cocaína/farmacologia , Sinais (Psicologia) , Morfina/farmacologia , Entorpecentes/farmacologia , Sacarina/administração & dosagem , Caracteres Sexuais , Análise de Variância , Animais , Condicionamento Clássico/efeitos dos fármacos , Corticosterona/sangue , Ciclo Estral/efeitos dos fármacos , Feminino , Masculino , Ratos , Ratos Sprague-Dawley , Recompensa , Paladar/efeitos dos fármacos
14.
Brain Res Bull ; 138: 88-95, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28899796

RESUMO

Evidence suggests that the addiction process may begin immediately in some vulnerable subjects. Specifically, some rats have been shown to exhibit aversive taste reactivity (gapes) following the intraoral delivery of a cocaine-predictive taste cue after as few as 1-2 taste-drug pairings. After only 3-4 trials, the number of gapes becomes a reliable predictor of later cocaine self-administration. Given that escalation of drug-taking behavior over time is recognized as a key feature of substance use disorder (SUD) and addiction, the present study examined the relationship between early aversion to the cocaine-predictive flavor cue and later escalation of cocaine self-administration in an extended-access paradigm. The data show that rats who exhibit the greatest conditioned aversion early in training to the intraorally delivered cocaine-paired cue exhibit the greatest escalation of cocaine self-administration over 15 extended-access trials. This finding suggests that early onset of the conditioned opponent process (i.e., the near immediate shift from ingestion to rejection of the drug-paired cue) is a reliable predictor of future vulnerability and resilience to cocaine addiction-like behavior. Future studies must determine the underlying neural mechanisms associated with this early transition and, hence, with early vulnerability to the later development of SUD and addiction. In so doing, we shall be in position to discover novel diagnostics and novel avenues of prevention and treatment.


Assuntos
Anestésicos Locais/administração & dosagem , Cocaína/administração & dosagem , Autoadministração , Paladar/efeitos dos fármacos , Administração Oral , Análise de Variância , Animais , Masculino , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Reflexo de Endireitamento/efeitos dos fármacos , Fatores de Tempo
15.
Brain Res Bull ; 138: 73-79, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28939474

RESUMO

The single nucleotide polymorphism of the µ-opioid receptor, OPRM1 A118G, has been associated with greater drug and alcohol use, increased sensitivity to pain, and reduced sensitivity to the antinociceptive effects of opiates. In the present studies, we employed a 'humanized' mouse model containing the wild-type (118AA) or variant (118GG) allele to examine behavior in a model of heroin-induced devaluation of an otherwise palatable saccharin cue when repeated saccharin-heroin pairings occurred every 24h (Experiment 1) or every 48h (Experiment 2). The results showed that, while both the 118AA and 118GG mice demonstrated robust avoidance of the heroin-paired saccharin cue following daily taste-drug pairings, only the 118AA mice suppressed intake of the heroin-paired saccharin cue when 48h elapsed between each taste-drug pairing. Humanized 118GG mice, then, defend their intake of the sweet cue despite saccharin-heroin pairings and this effect is illuminated by the use of spaced, rather than massed, trials. Given that this pattern of strain difference is not evident with saccharin-cocaine pairings (Freet et al., 2015), reduced avoidance of the heroin-paired saccharin cue by the 118GG mice may be due to an interaction between the opiate and the subjects' drive for the sweet or, alternatively, to differential downstream sensitivity to the aversive kappa mediated properties of the drug. These alternative hypotheses are addressed.


Assuntos
Analgésicos Opioides/farmacologia , Heroína/farmacologia , Polimorfismo de Nucleotídeo Único/genética , Receptores Opioides mu/genética , Sacarina/administração & dosagem , Edulcorantes/administração & dosagem , Análise de Variância , Animais , Aprendizagem por Associação/efeitos dos fármacos , Aprendizagem da Esquiva/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Peso Corporal/genética , Comportamento de Escolha/efeitos dos fármacos , Ritmo Circadiano/efeitos dos fármacos , Ritmo Circadiano/genética , Sinais (Psicologia) , Ingestão de Líquidos/efeitos dos fármacos , Ingestão de Líquidos/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores Opioides mu/metabolismo , Recompensa , Paladar
16.
Physiol Behav ; 167: 282-288, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27640134

RESUMO

Iron deficiency (ID) is the most prevalent single-nutrient deficiency worldwide. There is evidence that ID early in development (preweaning in rat) causes irreversible neurologic, behavioral, and motor development deficits. Many of these effects have been attributed to damage to dopamine systems, including ID-induced changes in transporter and receptor numbers in the striatum and nucleus accumbens. These mesolimbic dopaminergic neurons are, in part, responsible for mediating reward and thus play a key role in addiction. However, there has been relatively little investigation into the behavioral effects of ID on drug addiction. In 2002, we found that rats made ID from weaning (postnatal day 21) and throughout the experiment acquired cocaine self-administration significantly more slowly than controls and failed to increase responding when the dose of the drug was decreased. In the present study, we assessed addiction for self-administered cocaine in rats with a history of preweaning ID only during postnatal days 4 through 21, and iron replete thereafter. The results showed that while ID did not affect the number of cocaine infusions or the overall addiction-like behavior score, ID rats scored higher on a measure of continued responding for drug than did iron replete controls. This increase in responding, however, was less goal-directed as ID rats also responded more quickly to the non-rewarded manipulandum than did control rats. Thus, while ID early in infancy did not significantly increase addiction-like behaviors for cocaine in this small study, the pattern of data suggests a possible underlying learning or performance impairment. Future studies will be needed to elucidate the exact neuro-behavioral deficits that lead to the increase in indiscriminate responding for drug in rats with a history of perinatal ID.


Assuntos
Comportamento Aditivo/fisiopatologia , Cocaína/administração & dosagem , Inibidores da Captação de Dopamina/administração & dosagem , Deficiências de Ferro , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Análise de Variância , Animais , Comportamento Aditivo/induzido quimicamente , Condicionamento Operante/efeitos dos fármacos , Condicionamento Operante/fisiologia , Relação Dose-Resposta a Droga , Feminino , Hemoglobinas/metabolismo , Masculino , Gravidez , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/fisiologia , Esquema de Reforço , Autoadministração
17.
Physiol Behav ; 161: 104-115, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27083122

RESUMO

Rats avoid intake of a taste cue when paired with a drug of abuse or with the illness-inducing agent, lithium chloride (LiCl). Although progress has been made, it is difficult to compare the suppressive effects of abused agents and LiCl on intake of a gustatory conditioned stimulus (CS) because of the cross-laboratory use of different CSs, different unconditioned stimuli (USs), and different doses of the drugs, different conditioning regimens, and different restriction states. Here we have attempted to unify these variables by comparing the suppressive effects of a range of doses of morphine, cocaine, and LiCl on intake of a saccharin CS using a common regimen in non-restricted, food restricted, or water restricted male Sprague-Dawley rats. The results showed that, while the putatively aversive agent, LiCl, was effective in suppressing intake of the taste cue across nearly all doses, regardless of restriction state, the suppressive effects of both morphine and cocaine were greatly reduced when evaluated in either food or water restricted rats. Greater sensitivity to drug was revealed, at very low doses, when testing occurred in the absence of need (i.e., when the rats were non-restricted). Together, these results provide the first uniform and comprehensive analysis of the suppressive effects of morphine, cocaine, and LiCl as a function of dose and restriction state. In the present case, the suppressive effects of morphine and cocaine are found to differ from those of LiCl and, in some respects, from one another as well.


Assuntos
Antimaníacos/farmacologia , Aprendizagem da Esquiva/efeitos dos fármacos , Cocaína/farmacologia , Cloreto de Lítio/farmacologia , Morfina/farmacologia , Entorpecentes/farmacologia , Sacarina/administração & dosagem , Vasoconstritores/farmacologia , Animais , Relação Dose-Resposta a Droga , Ingestão de Alimentos/efeitos dos fármacos , Privação de Alimentos , Preferências Alimentares , Masculino , Ratos , Ratos Sprague-Dawley , Privação de Água
18.
Brain Res Bull ; 123: 71-80, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26733446

RESUMO

Heroin addiction is a disease of chronic relapse that harms the individual through devaluation of personal responsibilities in favor of finding and using drugs. Only some recreational heroin users devolve into addiction but the basis of these individual differences is not known. We have shown in rats that avoidance of a heroin-paired taste cue reliably identifies individual animals with greater addiction-like behavior for heroin. Here rats received 5min access to a 0.15% saccharin solution followed by the opportunity to self-administer either saline or heroin for 6h. Large Suppressors of the heroin-paired taste cue displayed increased drug escalation, motivation for drug, and drug loading behavior compared with Small Suppressors. Little is known about the molecular mechanisms of these individual differences in addiction-like behavior. We examined the individual differences in mRNA expression in the nucleus accumbens (NAc) of rats that were behaviorally stratified by addiction-like behavior using next-generation sequencing. We hypothesized that based on the avoidance of the drug-paired cue there will be a unique mRNA profile in the NAc. Analysis of strand-specific whole genome RNA-Seq data revealed a number of genes differentially regulated in NAc based on the suppression of the natural saccharine reward. Large Suppressors exhibited a unique mRNA prolife compared to Saline controls and Small Suppressors. Genes related to immunity, neuronal activity, and behavior were differentially expressed among the 3 groups. In total, individual differences in avoidance of a heroin-paired taste cue are associated with addiction-like behavior along with differential NAc gene expression.


Assuntos
Dependência de Heroína/genética , Núcleo Accumbens/efeitos dos fármacos , Animais , Encéfalo/efeitos dos fármacos , Condicionamento Operante/fisiologia , Sinais (Psicologia) , Heroína/metabolismo , Dependência de Heroína/metabolismo , Individualidade , Masculino , Núcleo Accumbens/metabolismo , Núcleo Accumbens/fisiologia , Ratos , Ratos Sprague-Dawley , Recompensa , Sacarina/administração & dosagem , Autoadministração , Paladar , Percepção Gustatória , Transcriptoma/genética
19.
Behav Neurosci ; 130(1): 103-13, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26653714

RESUMO

Rats suppress intake of a palatable taste cue when paired with a rewarding or an aversive stimulus in appetitive or aversive conditioning, respectively. A similar phenomenon occurs with drugs of abuse, but the nature of this conditioning has been subject for debate. While relatively little is known about the underlying neural circuitry, we recently reported bilateral lesions of the thalamic trigeminal orosensory area isolate drug-induced suppression of intake of a taste cue. The lesion blocks avoidance of the taste cue when paired with experimenter delivered drugs of abuse, yet has no effect on avoidance of the same cue when paired with an aversive agent or when it predicts access to a highly palatable sucrose solution. We hypothesize the lesion may blunt the rewarding properties of the drug. To test this, we used a runway apparatus, as running speed has been shown to increase with increasing reward value. Our hypothesis was supported by failure of the lesioned rats to increase running speed for morphine. Interestingly, lesioned rats did avoid intake of the drug-paired cue when presented in the runway apparatus and displayed naloxone-precipitated withdrawal. Using a partial crossover design, the lesion prevented avoidance of a cocaine-paired cue when presented in the home cage. We conclude that the lesion disrupts avoidance of a taste cue in anticipation of the rewarding properties of a drug but, at least in the presence of contextual cues, allows for avoidance of a taste cue as it elicits the onset of an aversive conditioned state of withdrawal.


Assuntos
Comportamento de Procura de Droga/fisiologia , Motivação/fisiologia , Percepção Gustatória/fisiologia , Paladar/fisiologia , Tálamo/lesões , Análise de Variância , Anestésicos Locais/farmacologia , Animais , Peso Corporal/efeitos dos fármacos , Cocaína/farmacologia , Condicionamento Psicológico , Comportamento de Procura de Droga/efeitos dos fármacos , Masculino , Motivação/efeitos dos fármacos , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Ratos , Ratos Sprague-Dawley , Recompensa , Sacarina/administração & dosagem , Estatística como Assunto , Edulcorantes/administração & dosagem , Paladar/efeitos dos fármacos , Percepção Gustatória/efeitos dos fármacos , Tálamo/efeitos dos fármacos
20.
Brain Res Bull ; 123: 61-70, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26494018

RESUMO

The ability to predict individual vulnerability to substance abuse would allow for a better understanding of the progression of the disease and development of better methods for prevention and/or early intervention. Here we use drug-induced devaluation of a saccharin cue in an effort to predict later addiction-like behavior in a model akin to that used by Deroche-Gamonet et al. (2004) and seek to link such vulnerability to changes in expression of various mu opioid receptor and D2 receptor-interacting proteins in brain. The results show that the greatest heroin-induced suppression of intake of a saccharin cue is associated with the greatest vulnerability to later addiction-like behavior and to differences in the expression of WLS, ß-catenin, and NCS-1 in brain compared to rats that exhibited the least suppression of intake of the heroin-paired cue and/or saline controls. Finally, because the self-administration model employed produced no significant differences in drug intake between groups, overall, the resultant changes in protein expression can be more closely linked to individual differences in motivation for drug.


Assuntos
Dependência de Heroína/metabolismo , Dependência de Heroína/prevenção & controle , Heroína/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Sinais (Psicologia) , Heroína/farmacologia , Masculino , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/metabolismo , Sacarina/administração & dosagem , Autoadministração , Paladar/fisiologia , Percepção Gustatória/fisiologia , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA