Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Transl Psychiatry ; 12(1): 530, 2022 12 31.
Article in English | MEDLINE | ID: mdl-36587026

ABSTRACT

Repeated cocaine exposure causes compensatory neuroadaptations in neurons in the nucleus accumbens (NAc), a region that mediates reinforcing effects of drugs. Previous studies suggested a role for adenosine monophosphate-activated protein kinase (AMPK), a cellular energy sensor, in modulating neuronal morphology and membrane excitability. However, the potential involvement of AMPK in cocaine use disorder is still unclear. The present study employed a cocaine self-administration model in rats to investigate the effect of AMPK and its target cyclic adenosine monophosphate response element binding protein-regulated transcriptional co-activator 1 (CRTC1) on cocaine reinforcement and the motivation for cocaine. We found that intravenous cocaine self-administration significantly decreased AMPK activity in the NAc shell (NAcsh), which persisted for at least 7 days of withdrawal. Cocaine reinforcement, reflected by self-administration behavior, was significantly prevented or enhanced by augmenting or suppressing AMPK activity pharmacologically and genetically, respectively. No difference in sucrose self-administration behavior was found after the same manipulations. The inhibition of AMPK activity in the NAcsh also increased the motivation for cocaine in progressive-ratio schedules of reinforcement, whereas the activation of AMPK had no effect. The knockdown of CRTC1 in the NAcsh significantly impaired cocaine reinforcement, which was rescued by pharmacologically increasing AMPK activity. Altogether, these results indicate that AMPK in the NAcsh is critical for cocaine reinforcement, possibly via the regulation of CRTC1 signaling. These findings may help reveal potential therapeutic targets and have important implications for the treatment of cocaine use disorder and relapse.


Subject(s)
Cocaine , Rats , Animals , Cocaine/pharmacology , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/pharmacology , Rats, Sprague-Dawley , Reinforcement, Psychology , Transcription Factors/metabolism , Adenosine Monophosphate/metabolism , Adenosine Monophosphate/pharmacology , Nucleus Accumbens , Self Administration
2.
CNS Neurosci Ther ; 27(2): 196-205, 2021 02.
Article in English | MEDLINE | ID: mdl-33118700

ABSTRACT

AIMS: Limited vs extended drug exposure has been proposed as one of the key factors in determining the risk of relapse, which is the primary characteristic of addiction behaviors. The current studies were designed to explore the related behavioral effects and neuronal alterations in the insular cortex (IC), an important brain region involved in addiction. METHODS: Experiments started with rats at the age of 35 days, a typical adolescent stage when initial drug exposure occurs often in humans. The drug-seeking/taking behaviors, and membrane properties and intrinsic excitability of IC pyramidal neurons were measured on withdrawal day (WD) 1 and WD 45-48 after limited vs extended cocaine intravenous self-administration (IVSA). RESULTS: We found higher cocaine-taking behaviors at the late withdrawal period after limited vs extended cocaine IVSA. We also found minor but significant effects of limited but not extended cocaine exposure on the kinetics and amplitude of action potentials on WD 45, in IC pyramidal neurons. CONCLUSION: Our results indicate potential high risks of relapse in young rats with limited but not extended drug exposure, although the adaptations detected in the IC may not be sufficient to explain the neural changes of higher drug-taking behaviors induced by limited cocaine IVSA.


Subject(s)
Action Potentials/drug effects , Administration, Intravenous , Cocaine/administration & dosage , Drug-Seeking Behavior/drug effects , Insular Cortex/drug effects , Action Potentials/physiology , Administration, Intravenous/methods , Animals , Dopamine Uptake Inhibitors/administration & dosage , Drug Administration Schedule , Drug-Seeking Behavior/physiology , Insular Cortex/physiology , Male , Rats , Rats, Sprague-Dawley , Self Administration
3.
J Neurosci Res ; 99(2): 649-661, 2021 02.
Article in English | MEDLINE | ID: mdl-33094531

ABSTRACT

Adolescent alcohol drinking, primarily in the form of binge-drinking episodes, is a serious public health concern. Binge drinking in laboratory animals has been modeled by a procedure involving chronic intermittent ethanol (CIE) administration, as compared with chronic intermittent water (CIW). The prolonged effects of adolescent binge alcohol exposure in adults, such as high risk of developing alcohol use disorder, are severe but available treatments in the clinic are limited. One reason is the lack of sufficient understanding about the associated neuronal alterations. The involvement of the insular cortex, particularly the anterior agranular insula (AAI), has emerged as a critical region to explain neuronal mechanisms of substance abuse. This study was designed to evaluate the functional output of the AAI by measuring the intrinsic excitability of pyramidal neurons from male rats 2 or 21 days after adolescent or adult CIE treatment. Decreases in intrinsic excitability in AAI pyramidal neurons were detected 21 days, relative to 2 days, after adolescent CIE. Interestingly, the decreased intrinsic excitability in the AAI pyramidal neurons was observed 2 days after adult CIE, compared to adult CIW, but no difference was found between 2 versus 21 days after adult CIE. These data indicate that, although the AAI is influenced within a limited period after adult but not adolescent CIE, neuronal alterations in AAI are affected during the prolonged period of withdrawal from adolescent but not adult CIE. This may explain the prolonged vulnerability to mental disorders of subjects with an alcohol binge history during their adolescent stage.


Subject(s)
Binge Drinking/physiopathology , Ethanol/toxicity , Insular Cortex/drug effects , Age Factors , Animals , Ethanol/administration & dosage , Insular Cortex/growth & development , Insular Cortex/physiopathology , Male , Patch-Clamp Techniques , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Rats , Rats, Sprague-Dawley , Sexual Maturation
4.
Sci Rep ; 10(1): 4465, 2020 Mar 06.
Article in English | MEDLINE | ID: mdl-32144354

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

5.
Addict Biol ; 25(4): e12793, 2020 07.
Article in English | MEDLINE | ID: mdl-31339209

ABSTRACT

Postretrieval extinction procedures are effective nonpharmacological interventions for disrupting drug-associated memories. Nonetheless, the conditioned stimulus (CS) memory retrieval-extinction procedure is ineffective in inhibiting drug craving and relapse after prolonged withdrawal, which significantly undermines its therapeutic potential. In the present study, we showed that, unlike the CS memory retrieval-extinction procedure, noncontingent heroin injections (unconditioned stimulus [UCS]) 1 hour before the extinction sessions decreased the heroin-priming-induced reinstatement, renewal, and spontaneous recovery of heroin seeking after 28 days of withdrawal (ie, remote heroin-associated memories) in rats. The UCS retrieval manipulation induced reactivation of the basolateral amygdala (BLA) after prolonged withdrawal, and this reactivation was absent with the CS retrieval manipulation. Chemogenetic inactivation of the BLA abolished the inhibitory effect of the UCS memory retrieval-extinction procedure on heroin-priming-induced reinstatement after prolonged withdrawal. Furthermore, the combination of chemogenetic reactivation of BLA and CS retrieval-extinction procedure resembled the inhibitory effect of UCS retrieval-extinction procedure on heroin seeking after prolonged withdrawal. We also observed that the inhibitory effect of the UCS retrieval-extinction procedure is mediated by regulation of AMPA receptor endocytosis in the BLA. Our results demonstrate critical engagement of the BLA in reconsolidation updating of heroin-associated memory after prolonged withdrawal, extending our knowledge of the boundary conditions of the reconsolidation of drug-associated memories.


Subject(s)
Basolateral Nuclear Complex/metabolism , Drug-Seeking Behavior/physiology , Extinction, Psychological/physiology , Heroin Dependence/metabolism , Heroin/pharmacology , Memory Consolidation/physiology , Narcotics/pharmacology , Animals , Basolateral Nuclear Complex/physiology , Central Amygdaloid Nucleus/metabolism , Central Amygdaloid Nucleus/physiology , Endocytosis , Heroin Dependence/physiopathology , Male , Rats , Receptors, AMPA/metabolism , Time Factors
6.
J Neurosci ; 37(37): 8938-8951, 2017 09 13.
Article in English | MEDLINE | ID: mdl-28821652

ABSTRACT

Exposure to drug-paired cues causes drug memories to be in a destabilized state and interfering with memory reconsolidation can inhibit relapse. Calpain, a calcium-dependent neutral cysteine protease, is involved in synaptic plasticity and the formation of long-term fear memory. However, the role of calpain in the reconsolidation of drug reward memory is still unknown. In the present study, using a conditioned place preference (CPP) model, we found that exposure to drug-paired contextual stimuli induced the activation of calpain and decreased the expression of glutamate receptor interacting protein 1 (GRIP1) in the nucleus accumbens (NAc) core, but not shell, of male rats. Infusions of calpain inhibitors in the NAc core immediately after retrieval disrupted the reconsolidation of cocaine/morphine cue memory and blocked retrieval-induced calpain activation and GRIP1 degradation. The suppressive effect of calpain inhibitors on the expression of drug-induced CPP lasted for at least 14 d. The inhibition of calpain without retrieval 6 h after retrieval or after exposure to an unpaired context had no effects on the expression of reward memory. Calpain inhibition after retrieval also decreased cocaine seeking in a self-administration model and this effect did not recover spontaneously after 28 d. Moreover, the knock-down of GRIP1 expression in the NAc core by lentivirus-mediated short-hairpin RNA blocked disruption of the reconsolidation of drug cue memories that was induced by calpain inhibitor treatment. These results suggest that calpain activity in the NAc core is crucial for the reconsolidation of drug reward memory via the regulation of GRIP1 expression.SIGNIFICANCE STATEMENT Calpain plays an important role in synaptic plasticity and long-term memory consolidation, however, its role in the reconsolidation of drug cue memory remains unknown. Using conditioned place preference and self-administration procedures, we found that exposure to drug-paired cues induced the activation of calpain and decreased glutamate receptor interacting protein 1 (GRIP1) expression in the nucleus accumbens (NAc) core. The inhibition of calpain activity in the NAc core immediately after retrieval disrupted the reconsolidation of cocaine/morphine cue memory that was blocked by prior GRIP1 knock-down. Our findings indicate that calpain-GRIP signaling is essential for the restabilization process that is associated with drug cue memory and the inhibition of calpain activity may be a novel strategy for the prevention of drug relapse.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Calpain/metabolism , Cues , Drug-Seeking Behavior/physiology , Memory Consolidation/physiology , Nerve Tissue Proteins/metabolism , Nucleus Accumbens/physiology , Reward , Animals , Gene Expression/physiology , Male , Rats , Rats, Sprague-Dawley
7.
Sci Rep ; 7(1): 9625, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28852144

ABSTRACT

The inability to successfully adapt to stress produces pathological changes that can lead to depression. Molecular hydrogen has anti-oxidative and anti-inflammatory activities and neuroprotective effects. However, the potential role of molecular hydrogen in stress-related disorders is still poorly understood. The present study aims to investigate the effects of hydrogen gas on resilience to stress in mice. The results showed that repeated inhalation of hydrogen-oxygen mixed gas [67%:33% (V/V)] significantly decreased both the acute and chronic stress-induced depressive- and anxiety-like behaviors of mice, assessed by tail suspension test (TST), forced swimming test (FST), novelty suppressed feeding (NSF) test, and open field test (OFT). ELISA analyses showed that inhalation of hydrogen-oxygen mixed gas blocked CMS-induced increase in the serum levels of corticosterone, adrenocorticotropic hormone, interleukin-6, and tumor necrosis factor-α in mice exposed to chronic mild stress. Finally, inhalation of hydrogen gas in adolescence significantly increased the resilience to acute stress in early adulthood, which illustrates the long-lasting effects of hydrogen on stress resilience in mice. This was likely mediated by inhibiting the hypothalamic-pituitary-adrenal axis and inflammatory responses to stress. These results warrant further exploration for developing molecular hydrogen as a novel strategy to prevent the occurrence of stress-related disorders.


Subject(s)
Adaptation, Physiological/drug effects , Gases/administration & dosage , Hydrogen/administration & dosage , Neuroprotective Agents/administration & dosage , Stress, Physiological/drug effects , Administration, Inhalation , Animals , Behavior, Animal , Blood Chemical Analysis , Mice
8.
Biol Psychiatry ; 82(11): 781-793, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28648649

ABSTRACT

BACKGROUND: Nicotine craving and relapse often occurs after reactivation of nicotine reward memories. We recently developed a memory retrieval-reconsolidation interference procedure in which reactivating nicotine reward memories by acute exposure to nicotine (the unconditioned stimulus [UCS]) and then pharmacologically interfering with memory reconsolidation decreased relapse to nicotine seeking in rats and nicotine craving in smokers. Here, we investigated underlying mechanisms. METHODS: In the first series of experiments, we trained rats for nicotine-induced conditioned place preference (CPP) or nicotine self-administration and ventricularly microinjected them with the protein synthesis inhibitor anisomycin immediately after UCS-induced memory retrieval. In the second series of experiments, we used tyramide-amplified immunohistochemistry-fluorescence in situ hybridization to examine neural ensembles in the basolateral amygdala (BLA) reactivated by nicotine conditioned stimulus- or UCS-induced memory retrieval. We then used the Daun02 chemogenetic inactivation procedure to selectively inhibit the nicotine UCS-reactivated BLA neuronal ensembles. RESULTS: Ventricular injections of the anisomycin immediately after nicotine UCS memory retrieval inhibited subsequent nicotine CPP and relapse to operant nicotine seeking after short or prolonged abstinence. More important, within BLA, distinct neuronal ensembles encoded pavlovian CPP and operant self-administration reward memories and nicotine (the UCS) injections in the home cage reactivated both neuronal ensembles. Daun02 chemogenetic inactivation of the nicotine-reactivated ensembles inhibited both nicotine CPP and relapse to nicotine seeking. CONCLUSIONS: Results demonstrate that the nicotine UCS-induced memory retrieval manipulation reactivates multiple nicotine reward memories that are encoded by distinct BLA neuronal ensembles that play a role in nicotine preference and relapse.


Subject(s)
Amygdala/cytology , Conditioning, Operant/physiology , Memory/drug effects , Neurons/physiology , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Animals , Conditioning, Classical/drug effects , Conditioning, Operant/drug effects , Extinction, Psychological/drug effects , Gene Expression Regulation/drug effects , Male , Neural Inhibition/drug effects , Neurons/drug effects , Oncogene Proteins v-fos/genetics , Oncogene Proteins v-fos/metabolism , Phosphopyruvate Hydratase/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Reward , Self Administration
9.
Sci Rep ; 7(1): 1038, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28432301

ABSTRACT

Relapse to drug seeking can be caused by exposure to drug-associated cues, provoking drug craving even after prolonged abstinence. Recent studies demonstrated that AMP-activated protein kinase (AMPK) regulates neuronal morphology and membrane excitability in neurons. Here, we investigated the role of AMPK activity in the nucleus accumbens (NAc) in relapse to cocaine seeking. We found that exposure to drug-related cues reinstated cocaine-seeking behavior and increased AMPK and p70s6k phosphorylation in the NAc core but not shell. Augmenting AMPK activity by intra-NAc core infusions of the AMPK activator 5-amino-1-ß-D-ribofuranosyl-imidazole-4-carboxamide (AICAR) or adenovirus expressing constitutively active subunits of AMPK decreased cue-induced reinstatement of cocaine seeking and inhibited the mammalian target of rapamycin complex 1 (mTORC1) and extracellular signal-regulated kinase 1/2 (ERK1/2) pathways. In contrast, inhibition of AMPK activity by intra-NAc core infusions of the AMPK inhibitor compound C or adenovirus expressing dominant-negative subunits of AMPK increased cue-induced reinstatement of cocaine seeking and enhanced mTORC1 and ERK1/2 activity. The regulation of AMPK activity in the NAc shell had no effect on cue-induced cocaine seeking. Altogether, these results indicate that AMPK activity in the NAc core is critical for the cue-induced reinstatement of cocaine seeking, which may be mediated by mTORC1 and ERK1/2 signaling.


Subject(s)
Adenylate Kinase/metabolism , Cocaine-Related Disorders/psychology , Drug-Seeking Behavior/physiology , Nucleus Accumbens/metabolism , Animals , Cocaine-Related Disorders/metabolism , Cues , Disease Models, Animal , MAP Kinase Signaling System , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Phosphorylation , Rats , Ribosomal Protein S6 Kinases, 70-kDa/metabolism
10.
JAMA Psychiatry ; 74(3): 224-232, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28146250

ABSTRACT

Importance: A relapse into nicotine addiction during abstinence often occurs after the reactivation of nicotine reward memories, either by acute exposure to nicotine (a smoking episode) or by smoking-associated conditioned stimuli (CS). Preclinical studies suggest that drug reward memories can undergo memory reconsolidation after being reactivated, during which they can be weakened or erased by pharmacological or behavioral manipulations. However, translational clinical studies using CS-induced memory retrieval-reconsolidation procedures to decrease drug craving reported inconsistent results. Objective: To develop and test an unconditioned stimulus (UCS)-induced retrieval-reconsolidation procedure to decrease nicotine craving among people who smoke. Design, Setting, and Participants: A translational rat study and human study in an academic outpatient medical center among 96 male smokers (aged 18- 45 years) to determine the association of propranolol administration within the time window of memory reconsolidation (after retrieval of the nicotine-associated memories by nicotine UCS exposure) with relapse to nicotine-conditioned place preference (CPP) and operant nicotine seeking in rats, and measures of preference to nicotine-associated CS and nicotine craving among people who smoke. Intervention: The study rats were injected noncontingently with the UCS (nicotine 0.15 mg/kg, subcutaneous) in their home cage, and the human study participants administered a dose of propranolol (40 mg, per os; Zhongnuo Pharma). Main Outcomes and Measures: Nicotine CPP and operant nicotine seeking in rats, and preference and craving ratings for newly learned and preexisting real-life nicotine-associated CS among people who smoke. Results: Sixty-nine male smokers completed the experiment and were included for statistical analysis: 24 in the group that received placebo plus 1 hour plus UCS, 23 who received propranolol plus 1 hour plus UCS, and 22 who received UCS plus 6 hours plus propranolol. In rat relapse models, propranolol injections administered immediately after nicotine UCS-induced memory retrieval inhibited subsequent nicotine CPP and operant nicotine seeking after short (CPP, d = 1.72, 95% CI, 0.63-2.77; operant seeking, d = 1.61, 95% CI, 0.59-2.60) or prolonged abstinence (CPP, d = 1.46, 95% CI, 0.42-2.47; operant seeking: d = 1.69, 95% CI, 0.66-2.69), as well as nicotine priming-induced reinstatement of nicotine CPP (d = 1.28, 95% CI, 0.27-2.26) and operant nicotine seeking (d = 1.61, 95% CI, 0.59-2.60) after extinction. Among the smokers, oral propranolol administered prior to nicotine UCS-induced memory retrieval decreased subsequent nicotine preference induced by newly learned nicotine CS (CS1, Cohen d = 0.61, 95% CI, 0.02-1.19 and CS2, d = 0.69, 95% CI, 0.10-1.28, respectively), preexisting nicotine CS (d = 0.57, 95% CI, -0.02 to 1.15), and nicotine priming (CS1, d = 0.82, 95% CI, 0.22-1.41 and CS2, d = 0.78, 95% CI, 0.18-1.37, respectively; preexisting nicotine CS, d = 0.92, 95% CI, 0.31-1.52), as well as nicotine craving induced by the preexisting nicotine CS (d = 0.64, 95% CI, 0.05-1.22), and nicotine priming (d = 1.15, 95% CI, 0.52-1.76). Conclusions and Relevance: In rat-to-human translational study, a novel UCS-induced memory retrieval-reconsolidation interference procedure inhibited nicotine craving induced by exposure to diverse nicotine-associated CS and nicotine itself. This procedure should be studied further in clinical trials.


Subject(s)
Association Learning/drug effects , Craving/drug effects , Mental Recall/drug effects , Nicotine , Propranolol/therapeutic use , Smoking Cessation/methods , Smoking Cessation/psychology , Tobacco Use Disorder/psychology , Tobacco Use Disorder/rehabilitation , Adolescent , Adult , Animals , Conditioning, Classical/drug effects , Female , Humans , Male , Memory Consolidation/drug effects , Middle Aged , Rats , Rats, Sprague-Dawley , Recurrence , Translational Research, Biomedical , Young Adult
11.
Sci Rep ; 6: 27895, 2016 06 10.
Article in English | MEDLINE | ID: mdl-27282818

ABSTRACT

Neuropeptides play important roles in modulating the rewarding value of abused drugs. Trefoil factor 3 (TFF3) was recently reported to modulate withdrawal syndrome of morphine, but the effects of TFF3 on the cocaine-induced behavioral changes are still elusive. In the present study, cocaine-induced hyperlocomotion and conditioned place preference (CPP) rat paradigms were provided to investigate the role of TFF3 in the reward response to cocaine. High-performance liquid chromatography (HPLC) analysis was used to analyse the dopamine concentration. The results showed that systemic TFF3 administration (0.1 mg/kg i.p.) significantly augmented cocaine- induced hyperlocomotion and CPP formation, without any effects on locomotor activity and aversive or rewarding effects per se. TFF3 significantly augmented the increment of the dopamine concentration in the NAc and the activity of the mTOR signalling pathway induced by acute cocaine exposure (10 mg/kg, i.p.) in the NAc shell, but not the core. The Intra-NAc shell infusion of rapamycin blocked TFF3-induced hyperactivity in cocaine-treatment rats. These findings indicated that TFF3 could potentiate behavioural response to cocaine, which may be associated with regulating dopamine concentration. Furthermore, the findings indicated that mTOR signalling pathway in the NAc shell is important for TFF3-induced enhancement on the cocaine-induced behavioral changes.


Subject(s)
Behavior, Animal/drug effects , Cocaine/pharmacology , Nucleus Accumbens/drug effects , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Trefoil Factor-3/pharmacology , Animals , Chromatography, High Pressure Liquid , Dopamine/analysis , Humans , Locomotion/drug effects , Male , Nucleus Accumbens/metabolism , Nucleus Accumbens/pathology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Trefoil Factor-3/genetics , Trefoil Factor-3/metabolism
12.
Sci Rep ; 5: 13327, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26289919

ABSTRACT

Drug addiction is considered an aberrant form of learning, and drug-associated memories evoked by the presence of associated stimuli (drug context or drug-related cues) contribute to recurrent craving and reinstatement. Epigenetic changes mediated by DNA methyltransferase (DNMT) have been implicated in the reconsolidation of fear memory. Here, we investigated the role of DNMT activity in the reconsolidation of cocaine-associated memories. Rats were trained over 10 days to intravenously self-administer cocaine by nosepokes. Each injection was paired with a light/tone conditioned stimulus (CS). After acquisition of stable self-administration behaviour, rats underwent nosepoke extinction (10 d) followed by cue-induced reactivation and subsequent cue-induced and cocaine-priming + cue-induced reinstatement tests or subsequently tested to assess the strength of the cocaine-associated cue as a conditioned reinforcer to drive cocaine seeking behaviour. Bilateral intra-basolateral amygdala (BLA) infusion of the DNMT inhibitor5-azacytidine (5-AZA, 1 µg per side) immediately following reactivation decreased subsequent reinstatement induced by cues or cocaine priming as well as cue-maintained cocaine-seeking behaviour. In contrast, delayed intra-BLA infusion of 5-AZA 6 h after reactivation or 5-AZA infusion without reactivation had no effect on subsequent cue-induced reinstatement. These findings indicate that memory reconsolidation for a cocaine-paired stimulus depends critically on DNMT activity in the BLA.


Subject(s)
Basolateral Nuclear Complex/enzymology , Cocaine/pharmacology , DNA (Cytosine-5-)-Methyltransferases/metabolism , Memory/drug effects , Animals , Azacitidine/pharmacology , Basolateral Nuclear Complex/drug effects , Behavior, Animal/drug effects , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Male , Rats, Sprague-Dawley , Reinforcement, Psychology
13.
Nat Commun ; 6: 7675, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26169171

ABSTRACT

We recently reported that a conditioned stimulus (CS) memory retrieval-extinction procedure decreases reinstatement of cocaine and heroin seeking in rats and heroin craving in humans. Here we show that non-contingent cocaine or methylphenidate injections (UCS retrieval) 1 h before the extinction sessions decreases cocaine-priming-induced reinstatement, spontaneous recovery, and renewal of cocaine seeking in rats. Unlike the CS-based memory retrieval-extinction procedure, the UCS memory retrieval manipulation decreases renewal and reinstatement of cocaine seeking in the presence of cocaine cues that were not present during extinction training and also decreases cocaine seeking when the procedure commences after 28 days of abstinence. The inhibitory effect of the UCS retrieval manipulation on cocaine-priming-induced reinstatement is mediated by regulation of AMPA-receptor endocytosis in the basolateral amygdala. The UCS memory retrieval-extinction procedure has superior relapse prevention characteristics than the CS memory retrieval-extinction procedure and could be a promising method for decreasing relapse in human addicts.


Subject(s)
Basolateral Nuclear Complex/metabolism , Behavior, Animal/physiology , Central Nervous System Stimulants/administration & dosage , Cocaine/administration & dosage , Drug-Seeking Behavior/physiology , Extinction, Psychological/physiology , Inhibition, Psychological , Mental Recall/physiology , Methylphenidate/administration & dosage , Receptors, AMPA/metabolism , Animals , Conditioning, Psychological/physiology , Cues , Endocytosis/physiology , Male , Rats , Rats, Sprague-Dawley , Recurrence , Self Administration
14.
J Neurosci ; 35(21): 8308-21, 2015 May 27.
Article in English | MEDLINE | ID: mdl-26019344

ABSTRACT

Fear extinction forms a new memory but does not erase the original fear memory. Exposure to novelty facilitates transfer of short-term extinction memory to long-lasting memory. However, the underlying cellular and molecular mechanisms are still unclear. Using a classical contextual fear-conditioning model, we investigated the effect of novelty on long-lasting extinction memory in rats. We found that exposure to a novel environment but not familiar environment 1 h before or after extinction enhanced extinction long-term memory (LTM) and reduced fear reinstatement. However, exploring novelty 6 h before or after extinction had no such effect. Infusion of the ß-adrenergic receptor (ßAR) inhibitor propranolol and glucocorticoid receptor (GR) inhibitor RU486 into the CA1 area of the dorsal hippocampus before novelty exposure blocked the effect of novelty on extinction memory. Propranolol prevented activation of the hippocampal PKA-CREB pathway, and RU486 prevented activation of the hippocampal extracellular signal-regulated kinase 1/2 (Erk1/2)-CREB pathway induced by novelty exposure. These results indicate that the hippocampal ßAR-PKA-CREB and GR-Erk1/2-CREB pathways mediate the extinction-enhancing effect of novelty exposure. Infusion of RU486 or the Erk1/2 inhibitor U0126, but not propranolol or the PKA inhibitor Rp-cAMPS, into the CA1 before extinction disrupted the formation of extinction LTM, suggesting that hippocampal GR and Erk1/2 but not ßAR or PKA play critical roles in this process. These results indicate that novelty promotes extinction memory via hippocampal ßAR- and GR-dependent pathways, and Erk1/2 may serve as a behavioral tag of extinction.


Subject(s)
Extinction, Psychological/physiology , Fear/physiology , Hippocampus/physiology , Receptors, Adrenergic, beta/physiology , Receptors, Glucocorticoid/physiology , Adrenergic beta-Antagonists/pharmacology , Animals , Extinction, Psychological/drug effects , Fear/drug effects , Hippocampus/drug effects , Male , Memory/drug effects , Memory/physiology , Rats , Rats, Sprague-Dawley , Receptors, Glucocorticoid/antagonists & inhibitors
15.
Nanomedicine ; 11(2): 391-400, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25461282

ABSTRACT

Leukocytes can cross intact blood-brain barrier under healthy conditions and in many neurological diseases, including psychiatric diseases. In present study, a cyclic RGD (cRGD) peptide with high affinity for integrin receptors of leukocytes was used to modify liposomes. The cRGD-modified liposomes (cRGDL) showed high affinity for monocytes in vitro and in vivo and co-migrated across in vitro BBB model with THP-1. The trefoil factor 3 (TFF3), a macromolecular drug, was rapidly and persistently delivered to brain for at least 12 h when loaded into cRGDL while 2.8-fold increase in drug concentration in basolateral amygdala regions related to depression was observed. A systemic administration of cRGDL-TFF3 mimicked antidepressant-like effect of direct intra-basolateral amygdala administration of TFF3 solution in rats subjected to chronic mild stress. The effective dual-brain targeting delivery resulting from the combination and co-migration of cRGDL with leukocyte cross BBB may be a promising strategy for targeted brain delivery. FROM THE CLINICAL EDITOR: In an effort to treat depression, brain targeted delivery via monocyte-cRGD liposome complexes capable of crossing the intact BBB was performed in this study in a murine model. Similar approaches may be helpful in the treatment of other neuropsychiatric conditions.


Subject(s)
Blood-Brain Barrier/drug effects , Depression/drug therapy , Drug Delivery Systems , Peptides, Cyclic/administration & dosage , Peptides/administration & dosage , Animals , Brain/drug effects , Brain/pathology , Depression/pathology , Drug Synergism , Humans , Leukocytes/drug effects , Liposomes/administration & dosage , Male , Mice , Rats , Trefoil Factor-3
16.
Addict Biol ; 20(3): 513-22, 2015 May.
Article in English | MEDLINE | ID: mdl-24698092

ABSTRACT

Time-dependent increases in cue-induced nicotine and methamphetamine craving during abstinence were recently reported in human drug-dependent individuals. In the present study, we sought to determine whether this 'incubation of craving' phenomenon also occurs in alcoholics. Four groups of 80 inpatient adult male alcoholics were assessed in a single session (between-group design) for cue-induced alcohol craving at 7, 14, 30 and 60 days of abstinence. Another group that included 19 patients was repeatedly tested for cue-induced alcohol craving at the same abstinence days as above. Other psychological and physiological measures were assessed at the four abstinence timepoints. Cue-induced alcohol craving measured with visual analogue scales was the highest at 60 days of abstinence both between and within groups. However, heart rate, blood pressure and skin conductance responses did not differ between abstinent groups. These results provide evidence of the incubation of alcohol craving in humans, extending previous reports with smokers and methamphetamine addicts.


Subject(s)
Alcohol Abstinence/psychology , Alcoholism/psychology , Craving , Adolescent , Adult , Analysis of Variance , Blood Pressure/physiology , Cues , Galvanic Skin Response/physiology , Heart Rate/physiology , Humans , Male , Middle Aged , Substance Withdrawal Syndrome/psychology , Young Adult
17.
Int J Neuropsychopharmacol ; 18(5)2014 Dec 07.
Article in English | MEDLINE | ID: mdl-25522425

ABSTRACT

BACKGROUND: Currently approved medications for opioid addiction have shown clinical efficacy, but undesired side effects, dependence induced by the medications themselves, and low treatment compliance necessitate the need for novel therapies. METHODS: A novel morphine-keyhole limpet hemocyanin conjugate vaccine was synthesized with 6-glutarylmorphine as the hapten and a lengthened linker of 6 carbon atoms. The titer and specificity of the triggered antibody were assessed by enzyme-linked immunosorbent assay. The effects of the vaccine on the morphine-induced elevation of dopamine levels in the nucleus accumbens were determined by high-performance liquid chromatography. The effects of the vaccine on morphine-induced locomotor sensitization and heroin-primed reinstatement of heroin self-administration were also assessed. RESULTS: After subcutaneous administration in rats, the vaccine triggered a high antibody titer, with comparable specificity for morphine, 6-acetylmorphine, and heroin, but no interaction with dissimilar therapeutic opioid compounds, including buprenorphine, naloxone, and nalorphine, was observed. The vaccine significantly prevented the elevation of dopamine levels in the nucleus accumbens induced by a single morphine challenge. Moreover, the vaccine prevented the expression of morphine-induced locomotor sensitization and heroin-primed reinstatement of heroin seeking, suggesting its potential for preventing relapse. CONCLUSION: These results demonstrate that active immunization with the present vaccine induces a robust morphine/heroin-specific antibody response in rats and attenuates the behavioral effects of morphine and heroin.


Subject(s)
Antibodies/administration & dosage , Behavior, Animal/drug effects , Dopamine/blood , Morphine/immunology , Vaccines, Conjugate/administration & dosage , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Animals , Antibodies/pharmacology , Chromatography, High Pressure Liquid , Drug-Seeking Behavior/drug effects , Heroin/administration & dosage , Heroin/adverse effects , Locomotion/drug effects , Male , Morphine/administration & dosage , Morphine/adverse effects , Morphine Derivatives/administration & dosage , Morphine Derivatives/adverse effects , Morphine Derivatives/immunology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Rats , Rats, Sprague-Dawley , Reinforcement, Psychology , Self Administration , Treatment Outcome , Vaccines, Conjugate/pharmacology
18.
J Neurosci ; 34(30): 10010-21, 2014 Jul 23.
Article in English | MEDLINE | ID: mdl-25057203

ABSTRACT

Maladaptive memories elicited by exposure to environmental stimuli associated with drugs of abuse are often responsible for relapse among addicts. Interference with the reconsolidation of drug memory can inhibit drug seeking. Previous studies have indicated that the dephosphorylation of the eukaryotic initiation factor 2 α-subunit (eIF2α) plays an important role in synaptic plasticity and long-term memory consolidation, but its role in the reconsolidation of drug memory remains unknown. The amygdala is required for the reconsolidation of a destabilized drug memory after retrieval of drug-paired stimuli. Here, we used conditioned place preference (CPP) and self-administration procedures to determine whether amygdala eIF2α dephosphorylation is required for the reconsolidation of morphine and cocaine memories in rats. We found that the levels of eIF2α phosphorylation (Ser51) and activating transcription factor 4 (ATF4) were decreased after reexposure to a previously morphine- or cocaine-paired context (i.e., a memory retrieval procedure) in the basolateral amygdala (BLA) but not in the central amygdala. Intra-BLA infusions of Sal003, a selective inhibitor of eIF2α dephosphorylation, immediately after memory retrieval disrupted the reconsolidation of morphine- or cocaine-induced CPP, leading to a long-lasting suppression of drug-paired stimulus-induced craving. Advanced knockdown of ATF4 expression in the BLA by lentivirus-mediated short-hairpin RNA blocked the disruption of the reconsolidation of morphine-induced CPP induced by Sal003 treatment. Furthermore, inhibition of eIF2α dephosphorylation in the BLA immediately after light/tone stimulus retrieval decreased subsequent cue-induced heroin-seeking behavior in the self-administration procedure. These results demonstrate that eIF2α dephosphorylation in the BLA mediates the memory reconsolidation of drug-paired stimuli.


Subject(s)
Amygdala/metabolism , Cues , Drug-Seeking Behavior/physiology , Eukaryotic Initiation Factor-2/metabolism , Memory/physiology , Amygdala/drug effects , Animals , Cocaine/administration & dosage , Drug-Seeking Behavior/drug effects , Heroin/administration & dosage , Male , Memory/drug effects , Morphine/administration & dosage , Phosphorylation/physiology , Rats , Rats, Sprague-Dawley , Self Administration
19.
J Neurosci ; 34(19): 6647-58, 2014 May 07.
Article in English | MEDLINE | ID: mdl-24806690

ABSTRACT

Extinction therapy has been suggested to suppress the conditioned motivational effect of drug cues to prevent relapse. However, extinction forms a new inhibiting memory rather than erasing the original memory trace and drug memories invariably return. Perineuronal nets (PNNs) are a specialized extracellular matrix around interneurons in the brain that have been suggested to be a permissive factor that allows synaptic plasticity in the adolescent brain. The degradation of PNNs caused by chondroitinase ABC (ChABC) may generate induced juvenile-like plasticity (iPlasticity) and promote experience-dependent plasticity in the adult brain. In the present study, we investigated the effect of removing PNNs in the amygdala of rat on the extinction of drug memories. We found that extinction combined with intra-amygdala injections of ChABC (0.01 U/side) prevented the subsequent priming-induced reinstatement of morphine-induced and cocaine-induced, but not food -induced, conditioned place preference (CPP). Intra-amygdala injections of ChABC alone had no effect on the retention, retrieval, or relearning of morphine-induced CPP and storage of acquired food-induced CPP. Moreover, we found that the procedure facilitated the extinction of heroin- and cocaine-seeking behavior and prevented the spontaneous recovery and drug-induced reinstatement of heroin- and cocaine-seeking behavior. We also found that the effect of PNNs degradation combined with extinction may be mediated by the potentiation of several plasticity-related proteins in the amygdala. Altogether, our findings demonstrate that a combination of extinction training with PNNs degradation in the amygdala erases drug memories and suggest that ChABC may be an attractive candidate for the prevention of relapse.


Subject(s)
Amygdala/drug effects , Amygdala/physiology , Memory , Nerve Net/physiology , Substance-Related Disorders/psychology , Animals , Blotting, Western , Chondroitin ABC Lyase/administration & dosage , Chondroitin ABC Lyase/pharmacology , Cocaine-Related Disorders/psychology , Conditioning, Operant , Extinction, Psychological , Food , Heroin Dependence/psychology , Male , Microinjections , Morphine Dependence/psychology , Neuronal Plasticity/drug effects , Rats , Rats, Sprague-Dawley , Reinforcement, Psychology , Secondary Prevention
20.
Psychopharmacology (Berl) ; 231(24): 4659-68, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24825609

ABSTRACT

RATIONALE: The persistence of physical dependence and craving in addicts is considered to contribute to relapse. Increasing evidence indicates that neuropeptide systems are associated with several phases of drug addiction, but little is known about whether the neuropeptide trefoil factor affects withdrawal symptoms. OBJECTIVES: This study aims to investigate the potential effects of the neuropeptide trefoil factor 3 (TFF3) on naloxone-precipitated withdrawal symptoms in morphine-dependent mice. RESULTS: Mice received increasing doses of morphine over 3 days. On day 4, the mice were injected with TFF3 (1.0 mg/kg, i.p.) 30 min after the last dose of morphine. Thirty minutes after TFF3 treatment, naloxone (1 mg/kg, i.p.) was injected, and body weight, jumping behavior, wet-dog shakes, and locomotor activity were assessed 30 min later. Naloxone caused significant weight loss and increased jumping behavior and wet-dog shakes in morphine-dependent mice. TFF3 (1.0 mg/kg) reversed these behavioral symptoms caused by morphine withdrawal, suggesting that TFF3 might ameliorate physical dependence associated with opiate addiction. Furthermore, TFF3 pretreatment significantly reduced morphine withdrawal-induced increases in plasma corticosterone and adrenocorticotropic hormone levels. The glucocorticoid receptor agonist RU486 blocked the behavioral effects of TFF3 on morphine withdrawal symptoms. Finally, Fos expression in the medial prefrontal cortex which was decreased during morphine withdrawal was increased by TFF3 pretreatment. CONCLUSION: These findings indicate that TFF3 might be a potential therapeutic candidate for opiate addiction by regulating glucocorticoid secretion and neuronal activation in the prefrontal cortex.


Subject(s)
Behavior, Animal/drug effects , Morphine/pharmacology , Naloxone/pharmacology , Narcotics/pharmacology , Opioid-Related Disorders/drug therapy , Peptides/therapeutic use , Substance Withdrawal Syndrome/drug therapy , Animals , Behavior, Animal/physiology , Male , Mice , Opioid-Related Disorders/metabolism , Peptides/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Substance Withdrawal Syndrome/metabolism , Trefoil Factor-2
SELECTION OF CITATIONS
SEARCH DETAIL
...