Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
eNeuro ; 10(5)2023 05.
Article in English | MEDLINE | ID: mdl-37193602

ABSTRACT

Mesolimbic nicotinic acetylcholine receptor (nAChRs) activation is necessary for nicotine reinforcement behavior, but it is unknown whether selective activation of nAChRs in the dopamine (DA) reward pathway is sufficient to support nicotine reinforcement. In this study, we tested the hypothesis that activation of ß2-containing (ß2*) nAChRs on VTA neurons is sufficient for intravenous nicotine self-administration (SA). We expressed ß2 nAChR subunits with enhanced sensitivity to nicotine (referred to as ß2Leu9'Ser) in the VTA of male Sprague Dawley (SD) rats, enabling very low concentrations of nicotine to selectively activate ß2* nAChRs on transduced neurons. Rats expressing ß2Leu9'Ser subunits acquired nicotine SA at 1.5 µg/kg/infusion, a dose too low to support acquisition in control rats. Saline substitution extinguished responding for 1.5 µg/kg/inf, verifying that this dose was reinforcing. ß2Leu9'Ser nAChRs also supported acquisition at the typical training dose in rats (30 µg/kg/inf) and reducing the dose to 1.5 µg/kg/inf caused a significant increase in the rate of nicotine SA. Viral expression of ß2Leu9'Ser subunits only in VTA DA neurons (via TH-Cre rats) also enabled acquisition of nicotine SA at 1.5 µg/kg/inf, and saline substitution significantly attenuated responding. Next, we examined electrically-evoked DA release in slices from ß2Leu9'Ser rats with a history of nicotine SA. Single-pulse evoked DA release and DA uptake rate were reduced in ß2Leu9'Ser NAc slices, but relative increases in DA following a train of stimuli were preserved. These results are the first to report that ß2* nAChR activation on VTA neurons is sufficient for nicotine reinforcement in rats.


Subject(s)
Nicotine , Receptors, Nicotinic , Rats , Male , Animals , Nicotine/pharmacology , Nicotine/metabolism , Nicotinic Agonists/pharmacology , Ventral Tegmental Area/metabolism , Rats, Sprague-Dawley , Receptors, Nicotinic/metabolism , Dopaminergic Neurons/metabolism
2.
eNeuro ; 6(6)2019.
Article in English | MEDLINE | ID: mdl-31744841

ABSTRACT

Ventral tegmental area (VTA) neurons receive glutamatergic and/or GABAergic input from other local neurons within the VTA. Nicotinic acetylcholine receptor (nAChR) activity is capable of modulating such intra-VTA transmission, but the mechanisms are unclear. Here, we isolated monosynaptic glutamate or GABA transmission from mouse medial VTA (mVTA) to lateral VTA (latVTA) using pharmacology and optogenetics, and we studied the ability of nicotine to modulate these modes of transmission. The action of nicotine on mVTA to latVTA glutamate transmission was bidirectional; nicotine enhanced glutamate release in half of the recorded latVTA cells and inhibited release in the other half. Nicotine-mediated reduction in glutamate release was reversed by blockade of GABAA receptors. This, coupled with expression data demonstrating coexpression of vesicular glutamate transporter 2 (VGluT2) and glutamate decarboxylase 2 (Gad2) in mVTA neurons, suggests that nicotine is able to stimulate GABA corelease from mVTA VGluT2+ neurons. Nicotine had an altogether different effect on mVTA to latVTA GABA release from Gad2+ cells; nicotine suppressed GABA release from mVTA Gad2+ terminals in nearly all cells tested. Together, these data uncover a complex system of local circuitry in the VTA that is modulated by nAChR activity. These actions of nicotine, which occurred at concentrations of nicotine found in the artificial CSF of cigarette smokers, may play a role in the adaptive response of the reward system to repeated nicotine exposure.


Subject(s)
GABAergic Neurons/metabolism , Glutamic Acid/metabolism , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Receptors, Nicotinic/metabolism , Ventral Tegmental Area/metabolism , Animals , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , GABAergic Neurons/drug effects , Glutamate Decarboxylase/metabolism , Male , Mice , Neural Pathways/drug effects , Neural Pathways/metabolism , Optogenetics , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Ventral Tegmental Area/drug effects , Vesicular Glutamate Transport Protein 2/metabolism
3.
J Neurosci ; 39(22): 4268-4281, 2019 05 29.
Article in English | MEDLINE | ID: mdl-30867261

ABSTRACT

Antagonism of nicotinic acetylcholine receptors (nAChRs) in the medial habenula (MHb) or interpeduncular nucleus (IPN) triggers withdrawal-like behaviors in mice chronically exposed to nicotine, implying that nicotine dependence involves the sensitization of nicotinic signaling. Identification of receptor and/or neurophysiological mechanisms underlying this sensitization is important, as it could promote novel therapeutic strategies to reduce tobacco use. Using an approach involving photoactivatable nicotine, we previously demonstrated that chronic nicotine (cNIC) potently enhances nAChR function in dendrites of MHb neurons. However, whether cNIC modulates downstream components of the habenulo-interpeduncular (Hb-IP) circuit is unknown. In this study, cNIC-mediated changes to Hb-IP nAChR function were examined in mouse (male and female) brain slices using molecular, electrophysiological, and optical techniques. cNIC enhanced action potential firing and modified spike waveform characteristics in MHb neurons. Nicotine uncaging revealed nAChR functional enhancement by cNIC on proximal axonal membranes. Similarly, nAChR-driven glutamate release from MHb axons was enhanced by cNIC. In IPN, the target structure of MHb axons, neuronal morphology, and nAChR expression is complex, with stronger nAChR function in the rostral subnucleus [rostral IPN (IPR)]. As in MHb, cNIC induced strong upregulation of nAChR function in IPN neurons. This, coupled with cNIC-enhanced nicotine-stimulated glutamate release, was associated with stronger depolarization responses to brief (1 ms) nicotine uncaging adjacent to IPR neurons. Together, these results indicate that chronic exposure to nicotine dramatically alters nicotinic cholinergic signaling and cell excitability in Hb-IP circuits, a key pathway involved in nicotine dependence.SIGNIFICANCE STATEMENT This study uncovers several neuropharmacological alterations following chronic exposure to nicotine in a key brain circuit involved in nicotine dependence. These results suggest that smokers or regular users of electronic nicotine delivery systems (i.e., "e-cigarettes") likely undergo sensitization of cholinergic circuitry in the Hb-IP system. Reducing the activity of Hb-IP nAChRs, either volitionally during smoking cessation or inadvertently via receptor desensitization during nicotine intake, may be a key trigger of withdrawal in nicotine dependence. Escalation of nicotine intake in smokers, or tolerance, may involve stimulation of these sensitized cholinergic pathways. Smoking cessation therapeutics are only marginally effective, and by identifying cellular/receptor mechanisms of nicotine dependence, our results take a step toward improved therapeutic approaches for this disorder.


Subject(s)
Habenula/drug effects , Interpeduncular Nucleus/drug effects , Neural Pathways/drug effects , Nicotine/pharmacology , Animals , Female , Habenula/metabolism , Interpeduncular Nucleus/metabolism , Male , Mice , Neural Pathways/metabolism , Nicotinic Agonists/pharmacology , Receptors, Nicotinic/metabolism , Synaptic Transmission/drug effects , Tobacco Use Disorder/metabolism
4.
Eur J Neurosci ; 50(3): 2224-2238, 2019 08.
Article in English | MEDLINE | ID: mdl-29779223

ABSTRACT

Nicotinic acetylcholine receptors (nAChRs), prototype members of the cys-loop ligand-gated ion channel family, are key mediators of cholinergic transmission in the central nervous system. Despite their importance, technical gaps exist in our ability to dissect the function of individual subunits in the brain. To overcome these barriers, we designed CRISPR/Cas9 small guide RNA sequences (sgRNAs) for the production of loss-of-function alleles in mouse nAChR genes. These sgRNAs were validated in vitro via deep sequencing. We subsequently targeted candidate nAChR genes in vivo by creating herpes simplex virus (HSV) vectors delivering sgRNAs and Cas9 expression to mouse brain. The production of loss-of-function insertions or deletions (indels) by these 'all-in-one' HSV vectors was confirmed using brain slice patch clamp electrophysiology coupled with pharmacological analysis. Next, we developed a scheme for cell type-specific gene editing in mouse brain. Knockin mice expressing Cas9 in a Cre-dependent manner were validated using viral microinjections and genetic crosses to common Cre-driver mouse lines. We subsequently confirmed functional Cas9 activity by targeting the ubiquitous neuronal protein, NeuN, using adeno-associated virus (AAV) delivery of sgRNAs. Finally, the mouse ß2 nAChR gene was successfully targeted in dopamine transporter (DAT)-positive neurons via CRISPR/Cas9. The sgRNA sequences and viral vectors, including our scheme for Cre-dependent gene editing, should be generally useful to the scientific research community. These tools could lead to new discoveries related to the function of nAChRs in neurotransmission and behavioral processes.


Subject(s)
Brain/physiology , Cholinergic Neurons/physiology , Gene Editing/methods , Genetic Vectors/genetics , Receptors, Nicotinic/physiology , Synaptic Transmission/physiology , Animals , CRISPR-Associated Protein 9/biosynthesis , CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems/physiology , Female , Genetic Vectors/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques
5.
Cell Rep ; 23(8): 2236-2244, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29791835

ABSTRACT

Ventral tegmental area (VTA) glutamate neurons are important components of reward circuitry, but whether they are subject to cholinergic modulation is unknown. To study this, we used molecular, physiological, and photostimulation techniques to examine nicotinic acetylcholine receptors (nAChRs) in VTA glutamate neurons. Cells in the medial VTA, where glutamate neurons are enriched, are responsive to acetylcholine (ACh) released from cholinergic axons. VTA VGLUT2+ neurons express mRNA and protein subunits known to comprise heteromeric nAChRs. Electrophysiology, coupled with two-photon microscopy and laser flash photolysis of photoactivatable nicotine, was used to demonstrate nAChR functional activity in the somatodendritic subcellular compartment of VTA VGLUT2+ neurons. Finally, optogenetic isolation of intrinsic VTA glutamatergic microcircuits along with gene-editing techniques demonstrated that nicotine potently modulates excitatory transmission within the VTA via heteromeric nAChRs. These results indicate that VTA glutamate neurons are modulated by cholinergic mechanisms and participate in the cascade of physiological responses to nicotine exposure.


Subject(s)
Glutamic Acid/metabolism , Neurons/metabolism , Receptors, Nicotinic/metabolism , Synaptic Transmission , Ventral Tegmental Area/metabolism , Animals , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism
6.
Proc Natl Acad Sci U S A ; 114(41): E8750-E8759, 2017 10 10.
Article in English | MEDLINE | ID: mdl-28973852

ABSTRACT

The basolateral amygdala (BLA) sends excitatory projections to the nucleus accumbens (NAc) and regulates motivated behaviors partially by activating NAc medium spiny neurons (MSNs). Here, we characterized a feedforward inhibition circuit, through which BLA-evoked activation of NAc shell (NAcSh) MSNs was fine-tuned by GABAergic monosynaptic innervation from adjacent fast-spiking interneurons (FSIs). Specifically, BLA-to-NAcSh projections predominantly innervated NAcSh FSIs compared with MSNs and triggered action potentials in FSIs preceding BLA-mediated activation of MSNs. Due to these anatomical and temporal properties, activation of the BLA-to-NAcSh projection resulted in a rapid FSI-mediated inhibition of MSNs, timing-contingently dictating BLA-evoked activation of MSNs. Cocaine self-administration selectively and persistently up-regulated the presynaptic release probability of BLA-to-FSI synapses, entailing enhanced FSI-mediated feedforward inhibition of MSNs upon BLA activation. Experimentally enhancing the BLA-to-FSI transmission in vivo expedited the acquisition of cocaine self-administration. These results reveal a previously unidentified role of an FSI-embedded circuit in regulating NAc-based drug seeking and taking.


Subject(s)
Action Potentials/physiology , Cocaine/administration & dosage , Drug-Seeking Behavior/physiology , Neural Inhibition , Neurons/physiology , Nucleus Accumbens/physiology , Vasoconstrictor Agents/administration & dosage , Animals , Basolateral Nuclear Complex , Female , Gene Knock-In Techniques , Long-Term Synaptic Depression , Male , Mice, Inbred C57BL , Neurons/cytology , Receptor, Cannabinoid, CB1/physiology , Self Administration
7.
PLoS One ; 12(7): e0182142, 2017.
Article in English | MEDLINE | ID: mdl-28759616

ABSTRACT

Nicotinic acetylcholine receptors containing α4 subunits (α4ß2* nAChRs) are critical for nicotinic cholinergic transmission and the addictive action of nicotine. To identify specific activities of these receptors in the adult mouse brain, we coupled targeted deletion of α4 nAChR subunits with behavioral and and electrophysiological measures of nicotine sensitivity. A viral-mediated Cre/lox approach allowed us to delete α4 from ventral midbrain (vMB) neurons. We used two behavioral assays commonly used to assess the motivational effects of drugs of abuse: home-cage oral self-administration, and place conditioning. Mice lacking α4 subunits in vMB consumed significantly more nicotine at the highest offered nicotine concentration (200 µg/mL) compared to control mice. Deletion of α4 subunits in vMB blocked nicotine-induced conditioned place preference (CPP) without affecting locomotor activity. Acetylcholine-evoked currents as well as nicotine-mediated increases in synaptic potentiation were reduced in mice lacking α4 in vMB. Immunostaining verified that α4 subunits were deleted from both dopamine and non-dopamine neurons in the ventral tegmental area (VTA). These results reveal that attenuation of α4* nAChR function in reward-related brain circuitry of adult animals may increase nicotine intake by enhancing the rewarding effects and/or reducing the aversive effects of nicotine.


Subject(s)
Nicotine/metabolism , Receptors, Nicotinic/metabolism , Reward , Ventral Tegmental Area/metabolism , Animals , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/physiology , Drug-Seeking Behavior , Female , Gene Deletion , Long-Term Potentiation , Male , Mice , Mice, Inbred C57BL , Receptors, Nicotinic/genetics , Synaptic Potentials , Ventral Tegmental Area/physiology
8.
Neuropsychopharmacology ; 41(9): 2399-410, 2016 08.
Article in English | MEDLINE | ID: mdl-27074816

ABSTRACT

Exposure to cocaine induces addiction-associated behaviors partially through remodeling neurocircuits in the nucleus accumbens (NAc). The paraventricular nucleus of thalamus (PVT), which projects to the NAc monosynaptically, is activated by cocaine exposure and has been implicated in several cocaine-induced emotional and motivational states. Here we show that disrupting synaptic transmission of select PVT neurons with tetanus toxin activated via retrograde trans-synaptic transport of cre from NAc efferents decreased cocaine self-administration in rats. This projection underwent complex adaptations after self-administration of cocaine (0.75 mg/kg/infusion; 2 h/d × 5 d, 1d overnight training). Specifically, 1d after cocaine self-administration, we observed increased levels of AMPA receptor (AMPAR)-silent glutamatergic synapses in this projection, accompanied by a decreased ratio of AMPAR-to-NMDA receptor (NMDAR)-mediated EPSCs. Furthermore, the decay kinetics of NMDAR EPSCs was significantly prolonged, suggesting insertion of new GluN2B-containing NMDARs to PVT-to-NAc synapses. After 45-d withdrawal, silent synapses within this projection returned to the basal levels, accompanied by a return of the AMPAR/NMDAR ratio and NMDAR decay kinetics to the basal levels. In amygdala and infralimbic prefrontal cortical projections to the NAc, a portion of cocaine-generated silent synapses becomes unsilenced by recruiting calcium-permeable AMPARs (CP-AMPARs) after drug withdrawal. However, the sensitivity of PVT-to-NAc synapses to CP-AMPAR-selective antagonists was not changed after withdrawal, suggesting that CP-AMPAR trafficking is not involved in the evolution of cocaine-generated silent synapses within this projection. Meanwhile, the release probability of PVT-to-NAc synapses was increased after short- and long-term cocaine withdrawal. These results reveal complex and profound alterations at PVT-to-NAc synapses after cocaine exposure and withdrawal.


Subject(s)
Cocaine/administration & dosage , Midline Thalamic Nuclei/drug effects , Midline Thalamic Nuclei/physiology , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiology , Synapses/drug effects , Synaptic Transmission/drug effects , Animals , Conditioning, Operant , Male , Neural Pathways , Rats, Sprague-Dawley , Self Administration , Synapses/physiology
9.
Addict Biol ; 19(1): 1-4, 2014 Jan.
Article in English | MEDLINE | ID: mdl-22260318

ABSTRACT

Methamphetamine (METH) dependence is becoming a serious socioeconomic health problem worldwide. The enhancement of the cholinergic nervous system is expected to greatly alleviate drug dependence. We investigated the effect of galantamine on the reinstatement of cue-induced METH-seeking behavior using a self-administration experiment. Treatment with galantamine (1 mg/kg, p.o.) 30 minutes before exposure to the cues suppressed the reinstatement of METH-seeking behavior. However, galantamine did not affect the cue-induced reinstatement of food-seeking behavior or locomotor activity. These results suggest that galantamine may be a candidate drug for treating relapses of METH-seeking behavior.


Subject(s)
Central Nervous System Stimulants/administration & dosage , Cholinesterase Inhibitors/pharmacology , Drug-Seeking Behavior/drug effects , Galantamine/pharmacology , Methamphetamine/administration & dosage , Amphetamine-Related Disorders/prevention & control , Analysis of Variance , Animals , Appetitive Behavior/drug effects , Cholinesterase Inhibitors/administration & dosage , Conditioning, Operant , Cues , Dose-Response Relationship, Drug , Galantamine/administration & dosage , Humans , Locomotion/drug effects , Mice , Nucleus Accumbens/drug effects , Secondary Prevention , Self Administration/statistics & numerical data
10.
Int J Neuropsychopharmacol ; 16(7): 1559-67, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23432945

ABSTRACT

Relapse of drug abuse after abstinence is a major challenge to the treatment of addicts. In our well-established mouse models of methamphetamine (Meth) self-administration and reinstatement, bilateral microinjection of adeno-associated virus vectors expressing GDNF (AAV-Gdnf) into the striatum significantly reduced Meth self-administration, without affecting locomotor activity. Moreover, the intrastriatal AAV-Gdnf attenuated cue-induced reinstatement of Meth-seeking behaviour in a sustainable manner. In addition, this manipulation showed that Meth-primed reinstatement of Meth-seeking behaviour was reduced. These findings suggest that the AAV vector-mediated Gdnf gene transfer into the striatum is an effective and sustainable approach to attenuate Meth self-administration and Meth-associated cue-induced relapsing behaviour and that the AAV-mediated Gdnf gene transfer in the brain may be a valuable gene therapy against drug dependence and protracted relapse in clinical settings.


Subject(s)
Corpus Striatum/metabolism , Genetic Therapy/methods , Glial Cell Line-Derived Neurotrophic Factor/therapeutic use , Methamphetamine/toxicity , Substance-Related Disorders/therapy , Analysis of Variance , Animals , Central Nervous System Stimulants , Cues , Dependovirus/genetics , Disease Models, Animal , Drug-Seeking Behavior/drug effects , Environment , Extinction, Psychological/drug effects , Glial Cell Line-Derived Neurotrophic Factor/biosynthesis , Glial Cell Line-Derived Neurotrophic Factor/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Microinjections , Reinforcement, Psychology , Self Administration , Substance-Related Disorders/mortality
11.
Am J Transl Res ; 4(4): 422-31, 2012.
Article in English | MEDLINE | ID: mdl-23145210

ABSTRACT

Extinction bursts are characterized by a temporary increase in responding when drug access is withheld from rats trained to self-administer drugs of abuse. Thus far, one study has examined extinction bursts for nicotine self-administration using a 23-h access paradigm [1]. Here we examined extinction bursts using previously published and unpublished data in which rats were trained to self-administer nicotine (0.03mg/kg/infusion) or food pellets (as a comparator) in 1-h sessions under an FR5 schedule of reinforcement followed by 1-h extinction sessions. Analysis of response rates during nicotine self-administration (NSA) was indicative of a loading phase, as response rates were significantly higher at the beginning of the session, which was not observed for food self-administration. At the start of extinction for both food and nicotine, although sessional response rates did not increase, there was an increase in response rate during the first 5-min of the first extinction session relative to self-administration. This transient extinction burst following nicotine was observed in a minority of subjects and correlated with the number of nicotine infusions obtained during self-administration. This transient extinction burst following food was observed in all subjects. Nicotine and food produce more transient extinction bursts compared to other drugs of abuse and only for a minority of animals in the case of nicotine. This study supports the presence of a loading phase in rats trained to self-administer nicotine in 1-r daily sessions and the presence of a transient extinction burst.

12.
Behav Brain Res ; 230(1): 34-9, 2012 Apr 21.
Article in English | MEDLINE | ID: mdl-22326373

ABSTRACT

The ability to examine genetically engineered mice in a chronic intravenous (IV) nicotine self-administration paradigm will be a powerful tool for investigating the contribution of specific genes to nicotine reinforcement and more importantly, to relapse behavior. Here we describe a reliable model of nicotine-taking and -seeking behavior in male C57BL/6J mice without prior operant training or food restriction. Mice were allowed to self-administer either nicotine (0.03mg/kg/infusion) or saline in 2-h daily sessions under fixed ratio 1 (FR1) followed by FR2 schedules of reinforcement. In the nicotine group, a dose-response curve was measured after the nose-poke behavior stabilized. Subsequently, nose-poke behavior was extinguished and ability of cue presentations, priming injections of nicotine, or intermittent footshock to reinstate responding was assessed in both groups. C57BL/6J mice given access to nicotine exhibited high levels of nose-poke behavior and self-administered a high number of infusions as compared to mice given access to saline. After this acquisition phase, changing the unit-dose of nicotine resulted in a flat dose-response curve for nicotine-taking and subsequently reinstatement of nicotine-seeking behavior was achieved by both nicotine-associated light cue presentation and intermittent footshock. Nicotine priming injections only triggered significant reinstatement on the second consecutive day of priming. In contrast, mice previously trained to self-administer saline did not increase their responding under those conditions. These results demonstrate the ability to produce nicotine-taking and nicotine-seeking behavior in naive C57BL/6J mice without both prior operant training and food restriction. Future work will utilize these models to evaluate nicotine-taking and relapsing behavior in genetically-altered mice.


Subject(s)
Conditioning, Operant/physiology , Food Deprivation/physiology , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Reinforcement, Psychology , Analysis of Variance , Animals , Cues , Dose-Response Relationship, Drug , Electroshock/adverse effects , Exploratory Behavior/drug effects , Extinction, Psychological/drug effects , Male , Mice , Mice, Inbred C57BL , Reinforcement Schedule , Self Administration
13.
Int J Neuropsychopharmacol ; 15(9): 1265-74, 2012 Oct.
Article in English | MEDLINE | ID: mdl-21939589

ABSTRACT

Effects of varenicline (Champix), a nicotinic partial agonist, were evaluated on subjective effects of nicotine (drug discrimination), motivation for nicotine taking (progressive-ratio schedule of intravenous nicotine self-administration) and reinstatement (cue-induced reinstatement of previously extinguished nicotine-seeking behaviour). Effects on motor performance were assessed in rats trained to discriminate nicotine (0.4 mg/kg) from saline under a fixed-ratio (FR 10) schedule of food delivery and in rats trained to respond for food under a progressive-ratio schedule. At short pretreatment times (5-40 min), varenicline produced full or high levels of partial generalization to nicotine's discriminative-stimulus effects and disrupted responding for food, while there were low levels of partial generalization and no disruption of responding for food at 2- or 4-h pretreatment times. Varenicline (1 and 3 mg/kg, 2-h pretreatment time) enhanced discrimination of low doses of nicotine and to a small extent decreased discrimination of the training dose of nicotine. It also dose-dependently decreased nicotine-taking behaviour, but had no effect on food-taking behaviour under progressive-ratio schedules. Finally, varenicline significantly reduced the ability of a nicotine-associated cue to reinstate extinguished nicotine-seeking behaviour. The ability of varenicline to reduce both nicotine-taking and nicotine-seeking behaviour can contribute to its relatively high efficacy in treating human smokers.


Subject(s)
Benzazepines/pharmacology , Cues , Drug-Seeking Behavior/drug effects , Quinoxalines/pharmacology , Tobacco Use Disorder/drug therapy , Animals , Conditioning, Operant/drug effects , Data Interpretation, Statistical , Discrimination, Psychological/drug effects , Dose-Response Relationship, Drug , Feeding Behavior/drug effects , Male , Motivation/drug effects , Nicotine/pharmacology , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Reinforcement Schedule , Self Administration , Tobacco Use Disorder/psychology , Varenicline
14.
Neuropsychopharmacology ; 37(3): 685-96, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22030716

ABSTRACT

Since cloning of the dopamine receptor D4 (DRD4), its role in the brain has remained unclear. It has been reported that polymorphism of the DRD4 gene in humans is associated with reactivity to cues related to tobacco smoking. However, the role of DRD4 in animal models of nicotine addiction has seldom been explored. In our study, male Long-Evans rats learned to intravenously self-administer nicotine under a fixed-ratio (FR) schedule of reinforcement. Effects of the selective DRD4 antagonist L-745,870 were evaluated on nicotine self-administration behavior and on reinstatement of extinguished nicotine-seeking behavior induced by nicotine-associated cues or by priming injections of nicotine. L-745,870 was also tested on reinstatement of extinguished food-seeking behavior as a control. In addition, the selective DRD4 agonist PD 168,077 was tested for its ability to reinstate extinguished nicotine-seeking behavior. Finally, L-745,870 was tested in Sprague Dawley rats trained to discriminate administration of 0.4 mg/kg nicotine from vehicle under an FR schedule of food delivery. L-745,870 significantly attenuated reinstatement of nicotine-seeking induced by both nicotine-associated cues and nicotine priming. In contrast, L-745,870 did not affect established nicotine self-administration behavior or reinstatement of food-seeking behavior induced by food cues or food priming. L-745,870 did not produce nicotine-like discriminative-stimulus effects and did not alter discriminative-stimulus effects of nicotine. PD 168,077 did not reinstate extinguished nicotine-seeking behavior. As DRD4 blockade by L-745,870 selectively attenuated both cue- and nicotine-induced reinstatement of nicotine-seeking behavior, without affecting cue- or food-induced reinstatement of food-seeking behavior, DRD4 antagonists are potential therapeutic agents against tobacco smoking relapse.


Subject(s)
Behavior, Animal/drug effects , Drug-Seeking Behavior/drug effects , Extinction, Psychological/drug effects , Nicotine/administration & dosage , Receptors, Dopamine D4/antagonists & inhibitors , Animals , Discrimination Learning/drug effects , Dopamine Antagonists/pharmacology , Male , Pyridines/pharmacology , Pyrroles/pharmacology , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Reinforcement, Psychology , Self Administration
15.
Psychopharmacology (Berl) ; 221(3): 427-36, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22160137

ABSTRACT

RATIONALE: During the development of addiction, addictive drugs induce transient and long-lasting changes in the brain including expression of endogenous molecules and alteration of morphological structure. Of the altered endogenous molecules, some facilitate but others slow the development of drug addiction. Previously, we have reported that tumor necrosis factor alpha (TNF-α) is a critical molecule among endogenous anti-addictive modulators using animal models of drug-conditioned place preference and drug discrimination. OBJECTIVES: Does targeted deletion of the TNF-α gene in mice affect methamphetamine (METH) self-administration, motivation to self-administer METH, cue-induced reinstatement of METH-seeking behavior, and food reinforcement or seeking behavior? METHODS: Both METH self-administration and reinstatement of drug-seeking behavior and food self-delivery and food-seeking behavior were measured in TNF-α (-/-) and wild-type mice. RESULTS: There were an upward shift of dose responses to METH self-administration under a fixed ratio schedule of reinforcement and higher breaking points under a progressive ratio schedule of reinforcement in TNF-α knockout (TNF-α (-/-)) mice as compared with wild-type mice. There was no significant difference in cue-induced reinstatement of METH-seeking behavior, food-maintained operant behavior, motivation to natural food, and cue-induced food-seeking behavior between TNF-α (-/-) and wild-type mice. CONCLUSION: TNF-α affects METH self-administration and motivation to self-administer METH but contributes to neither METH-associated cue-induced relapsing behavior nor food reward and food-seeking behavior. TNF-α may be explored for use as a diagnostic biomarker for the early stage of drug addiction.


Subject(s)
Behavior, Animal/drug effects , Central Nervous System Stimulants/administration & dosage , Methamphetamine/administration & dosage , Tumor Necrosis Factor-alpha/genetics , Animals , Conditioning, Operant , Cues , Dose-Response Relationship, Drug , Feeding Behavior , Gene Deletion , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motivation , Reinforcement Schedule , Self Administration
16.
Behav Brain Res ; 196(1): 1-10, 2009 Jan 03.
Article in English | MEDLINE | ID: mdl-18782591

ABSTRACT

To identify genetic risk factors involved in relapse to the abuse of drugs in humans, it is essential for researchers to develop a reliable mouse model of relapse by extending well-established extinction-reinstatement procedures in rats. Because of technical difficulties such as the relatively short duration of catheter patency in mice, few reports are available on the characterization of extinction-reinstatement behavior in wild-type and genetically engineered mutant mice. In this review, efforts are made to describe practical considerations during the establishment of extinction-reinstatement procedure in mice, including drug-primed, cue-induced, and stress-triggered reinstatement of previously extinguished drug-seeking behavior. Next, attention will be given to some characteristics of extinction-reinstatement behavior in mice. The present review might provide a new impetus in the exploration of genetic risk factors involved in relapse to drug dependence/addiction in humans using extinction-reinstatement procedures in widely available mutant mice.


Subject(s)
Behavior, Addictive/psychology , Disease Models, Animal , Reinforcement, Psychology , Substance-Related Disorders/psychology , Animals , Behavior, Addictive/physiopathology , Humans , Mice , Recurrence , Self Administration/methods , Substance-Related Disorders/physiopathology
17.
Ann N Y Acad Sci ; 1141: 76-95, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18991952

ABSTRACT

Recent evidence suggests that a variety of molecule products play critical roles in the transitions from recreational drug use to drug abuse, and then to drug dependence. Elucidation of the roles of specific molecules in the development of drug dependence can come from preclinical animal models and/or from clinical data. Among animal models, behavioral sensitization, conditioned place preference, drug discrimination, drug self-administration, and extensions of these basic procedures have been widely used to identify molecule products that might be involved in psychostimulant dependence. Repeated exposure to psychostimulants causes cellular adaptations in specific neuronal populations that are likely to contribute to dependence in some humans. In animal models, molecules that include shati, piccolo, tumor necrosis factor-alpha, and glial cell line-derived neurotrophic factor can act as antiaddictive factors. In some of these models, other molecules including matrix metalloproteinase and tissue plasminogen activator can act as proaddictive factors. We review evidence that the balance between levels of anti- and proaddictive factors induced by addictive drugs could play important roles in developing drug dependence. We focus on potential risk molecules in animal models for the development of methamphetamine dependence and their relevance to abusers. We propose that dynamic changes in the balance between levels of antiaddictive and proaddictive factors in the brain provide some of the determinants of susceptibility to drug dependence. Exploration of the roles that candidate molecules play in an appropriate repertoire of animal behavioral models, especially drug self-administration and extensions thereof, should thus help us to understand human stimulant dependence.


Subject(s)
Amphetamine-Related Disorders/genetics , Central Nervous System Stimulants/adverse effects , Methamphetamine/adverse effects , Models, Animal , Adaptation, Physiological , Amphetamine-Related Disorders/physiopathology , Animals , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Discrimination Learning/drug effects , Discrimination Learning/physiology , Drug Tolerance , Genetic Predisposition to Disease , Humans , Methamphetamine/administration & dosage , Methamphetamine/pharmacology , Methamphetamine/toxicity , Mice , Motor Activity/drug effects , Motor Activity/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Rats , Self Administration , Species Specificity , Substance Withdrawal Syndrome/genetics , Substance Withdrawal Syndrome/physiopathology , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/physiology , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/physiology
18.
FASEB J ; 21(9): 1994-2004, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17356005

ABSTRACT

Genetic factors are considered to play an important role in drug dependence/addiction including the development of drug dependence and relapse. With the use of a model of drug self-administration in mutant mice, several specific genes and proteins have been identified as potentially important in the development of drug dependence. In contrast, little is known about the role of specific genes in enduring vulnerability to relapse, a clinical hallmark of drug addiction. Using a mouse model of reinstatement, which models relapse of drug-seeking behavior in addicts, we provide evidence that a partial reduction in the expression of the glial cell line-derived neurotrophic factor (GDNF) potentiates methamphetamine (METH) self-administration, enhances motivation to take METH, increases vulnerability to drug-primed reinstatement, and prolongs cue-induced reinstatement of extinguished METH-seeking behavior. In contrast, there was no significant difference in novelty responses, METH-stimulated hyperlocomotion and locomotor sensitization, food-reinforced operant behavior and motivation, or reinstatement of food-seeking behavior between GDNF heterozygous knockout mice and wild-type littermates. These findings suggest that GDNF may be associated with enduring vulnerability to reinstatement of METH-seeking behavior and a potential target in the development of therapies to control relapse.


Subject(s)
Amphetamine-Related Disorders/genetics , Glial Cell Line-Derived Neurotrophic Factor/physiology , Methamphetamine , Amphetamine-Related Disorders/physiopathology , Amphetamine-Related Disorders/psychology , Animals , Appetitive Behavior/physiology , Catheters, Indwelling , Conditioning, Operant , Cues , Extinction, Psychological/physiology , Feeding Behavior , Food Deprivation , Genetic Predisposition to Disease , Glial Cell Line-Derived Neurotrophic Factor/deficiency , Glial Cell Line-Derived Neurotrophic Factor/genetics , Heterozygote , Infusion Pumps , Infusions, Intravenous , Male , Methamphetamine/administration & dosage , Mice , Mice, Knockout , Motivation , Recurrence , Reinforcement, Psychology , Self Administration
19.
Behav Brain Res ; 177(2): 261-8, 2007 Feb 27.
Article in English | MEDLINE | ID: mdl-17182116

ABSTRACT

It is essential to develop animal models to study the role of genetic factors in the relapse of drug-seeking behavior in genetically engineered mutant mice. This paper reports a typical model of drug-primed and cue-induced reinstatement of extinguished methamphetamine (METH)-seeking behavior in mice. C57BL/6J mice were trained to self-administer METH (0.1mg/kg/infusion) by poking their nose into an active hole under a fixed ratio schedule in daily 3-h sessions. After acquiring stable METH self-administration behavior, the mice were subjected to extinction training in the absence of both METH and METH-associated cues. Once the active nose-poking responses were extinguished, drug-primed and cue-induced reinstatement were investigated according to a within-subjects design. A priming injection of METH reliably reinstated the extinguished drug-seeking behavior in the absence of both METH and METH-associated cues. Interestingly, the drug-primed METH-seeking behavior disappeared within 2 months after withdrawal from METH, while cue-induced reinstatement of extinguished METH-seeking behavior lasted for at least 5 months after the withdrawal. A correlation study revealed that drug-primed, but not cue-induced, reinstatement behavior was positively correlated with the total amount of METH taken by individuals during METH self-administration. In conclusion, our findings suggest that the present reinstatement procedure for mouse model of relapse is useful and reliable, and different neural mechanisms may be involved in drug-primed and cue-induced METH-seeking behavior.


Subject(s)
Central Nervous System Stimulants/administration & dosage , Cues , Extinction, Psychological/drug effects , Methamphetamine/administration & dosage , Reinforcement, Psychology , Substance-Related Disorders/psychology , Analysis of Variance , Animals , Behavior, Animal , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Male , Mice , Mice, Inbred C57BL , Reinforcement Schedule , Self Administration
20.
Behav Brain Res ; 173(1): 39-46, 2006 Oct 02.
Article in English | MEDLINE | ID: mdl-16857277

ABSTRACT

Animal models of drug discrimination have been used to examine the subjective effects of addictive substances. The cAMP system is a crucial downstream signaling pathway implicated in the long-lasting neuroadaptations induced by addictive drugs. We examined effects of rolipram, nefiracetam, and dopamine D2-like receptor antagonists, all of which have been reported to modulate cAMP level in vivo, on the discriminative-stimulus effects of methamphetamine (METH) and morphine in rats. All these compounds inhibited the discriminative-stimulus effects of METH, while only rolipram and nefiracetam attenuated the discriminative-stimulus effects of morphine. In addition, neither nifedipine nor neomycin, two voltage-sensitive calcium channel blockers, was found to modulate the effect of nefiracetam on METH-associated discriminative stimuli, suggesting that the inhibitory effect of nefiracetam may not involve the activation of calcium channels. These findings suggest that the cAMP signaling cascade may play a key role in the discriminative-stimulus effects of METH and morphine and may be a potential target for the development of therapeutics to counter drugs of abuse.


Subject(s)
Central Nervous System Stimulants/pharmacology , Cyclic AMP/analogs & derivatives , Discrimination Learning/drug effects , Methamphetamine/pharmacology , Morphine/pharmacology , Narcotics/pharmacology , Signal Transduction/drug effects , Analysis of Variance , Animals , Calcium Channel Blockers/pharmacology , Central Nervous System Agents/pharmacology , Cyclic AMP/metabolism , Dopamine Antagonists/pharmacology , Dopamine D2 Receptor Antagonists , Male , Pyrrolidinones/pharmacology , Rats , Rats, Sprague-Dawley , Rolipram/pharmacology , Second Messenger Systems/drug effects , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...