Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Alcohol Clin Exp Res ; 46(4): 614-627, 2022 04.
Article in English | MEDLINE | ID: mdl-35102570

ABSTRACT

BACKGROUND: Alcohol is among the most commonly abused drugs worldwide. Cessation of chronic alcohol consumption can result in the appearance of withdrawal symptoms that commonly promote relapse in individuals with alcohol use disorder (AUD). Thus, preclinical models of voluntary alcohol consumption, in which animals manifest spontaneous signs of withdrawal after alcohol cessation, can be useful for studying AUD and its treatment. The intermittent two-bottle choice paradigm (I2BC) has been used extensively to examine alcohol intake in rodents. However, previous studies have reported conflicting observations regarding its potential to result in the spontaneous manifestation of withdrawal upon alcohol cessation. METHODS: We employed a battery of behavioral tests to examine the emergence of affective and physical signs of withdrawal in female and male mice exposed to alcohol in the I2BC for 10 weeks. Specifically, mice of both sexes undergoing 24-h withdrawal from the I2BC were tested for physical signs of withdrawal, anxiety-like behavior in the open field arena (OFA) and elevated plus maze (EPM), and anxiety/compulsive-like behavior in the marble burying test (MBT). The main outcomes from these tests were combined into a behavioral severity score to describe the overall behavioral phenotype. RESULTS: Both female and male mice undergoing withdrawal from the I2BC displayed elevated physical signs of withdrawal and anxiety-associated behavior in the EPM and MBT. Analysis of the overall behavioral severity score revealed more severe phenotypes in female and male mice undergoing withdrawal from the I2BC than controls. Additionally, stratification of the mice based on severity scores demonstrated a differential distribution of severities between the exposure groups. CONCLUSIONS: We confirmed that a significant fraction of mice chronically exposed to alcohol in the I2BC display spontaneous withdrawal. In addition, we showed that computing a severity score from a combination of behavioral metrics can be useful in preclinical research to model evaluation tools used in patients with AUD.


Subject(s)
Alcoholism , Substance Withdrawal Syndrome , Alcohol Drinking/psychology , Alcoholism/genetics , Animals , Anxiety/chemically induced , Anxiety/psychology , Ethanol , Female , Humans , Male , Mice , Mice, Inbred C57BL , Substance Withdrawal Syndrome/psychology
2.
Annu Rev Neurosci ; 34: 105-30, 2011.
Article in English | MEDLINE | ID: mdl-21438686

ABSTRACT

Nicotine is the principal addictive component that drives continued tobacco use despite users' knowledge of the harmful consequences. The initiation of addiction involves the mesocorticolimbic dopamine system, which contributes to the processing of rewarding sensory stimuli during the overall shaping of successful behaviors. Acting mainly through nicotinic receptors containing the α4 and ß2 subunits, often in combination with the α6 subunit, nicotine increases the firing rate and the phasic bursts by midbrain dopamine neurons. Neuroadaptations arise during chronic exposure to nicotine, producing an altered brain condition that requires the continued presence of nicotine to be maintained. When nicotine is removed, a withdrawal syndrome develops. The expression of somatic withdrawal symptoms depends mainly on the α5, α2, and ß4 (and likely α3) nicotinic subunits involving the epithalamic habenular complex and its targets. Thus, nicotine taps into diverse neural systems and an array of nicotinic acetylcholine receptor (nAChR) subtypes to influence reward, addiction, and withdrawal.


Subject(s)
Behavior, Addictive/physiopathology , Nicotine/adverse effects , Reward , Substance Withdrawal Syndrome/etiology , Substance Withdrawal Syndrome/psychology , Animals , Cerebral Cortex/pathology , Dopamine/metabolism , Humans , Limbic System/pathology , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Neurons/physiology , Receptors, Nicotinic/metabolism , Substance Withdrawal Syndrome/pathology
3.
Nicotine Tob Res ; 20(7): 789-799, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29065200

ABSTRACT

Introduction: Scientific discoveries over the past few decades have provided significant insight into the abuse liability and negative health consequences associated with tobacco and nicotine-containing products. While many of these advances have led to the development of policies and laws that regulate access to and formulations of these products, further research is critical to guide future regulatory efforts, especially as novel nicotine-containing products are introduced and selectively marketed to vulnerable populations. Discussion: In this narrative review, we provide an overview of the scientific findings that have impacted regulatory policy and discuss considerations for further translation of science into policy decisions. We propose that open, bidirectional communication between scientists and policy makers is essential to develop transformative preventive- and intervention-focused policies and programs to reduce appeal, abuse liability, and toxicity of the products. Conclusions: Through these types of interactions, collaborative efforts to inform and modify policy have the potential to significantly decrease the use of tobacco and alternative nicotine products and thus enhance health outcomes for individuals. Implications: This work addresses current topics in the nicotine and tobacco research field to emphasize the importance of basic science research and provide examples of how it can be utilized to inform public policy. In addition to relaying current thoughts on the topic from experts in the field, the article encourages continued efforts and communication between basic scientists and policy officials.


Subject(s)
Biomedical Research/legislation & jurisprudence , Nicotine , Public Policy/legislation & jurisprudence , Tobacco Products/legislation & jurisprudence , Biomedical Research/methods , Humans , Nicotine/standards , Tobacco Products/standards , Tobacco Use Disorder/prevention & control
4.
J Neurochem ; 142 Suppl 2: 130-143, 2017 08.
Article in English | MEDLINE | ID: mdl-28791703

ABSTRACT

Abstinence from chronic use of addictive drugs triggers an aversive withdrawal syndrome that compels relapse and deters abstinence. Many features of this syndrome are common across multiple drugs, involving both affective and physical symptoms. Some of the network signaling underlying withdrawal symptoms overlaps with activity that is associated with aversive mood states, including anxiety and depression. Given these shared features, it is not surprising that a particular circuit, the dorsal diencephalic conduction system, and the medial habenula (MHb) and interpeduncular nucleus (IPN), in particular, have been identified as critical to the emergence of aversive states that arise both as a result and, independently, of drug addiction. As the features of this circuit continue to be characterized, the MHb-IPN axis is emerging as a viable target for therapeutics to aid in the treatment of addiction to multiple drugs of abuse as well as mood-associated disorders. This is an article for the special issue XVth International Symposium on Cholinergic Mechanisms.


Subject(s)
Affect/drug effects , Anxiety/drug therapy , Behavior, Addictive/drug therapy , Interpeduncular Nucleus/physiopathology , Nicotinic Agonists/therapeutic use , Substance Withdrawal Syndrome/drug therapy , Affect/physiology , Animals , Anxiety/physiopathology , Behavior, Addictive/physiopathology , Humans , Interpeduncular Nucleus/drug effects , Substance Withdrawal Syndrome/physiopathology
5.
J Neurosci ; 34(12): 4273-84, 2014 Mar 19.
Article in English | MEDLINE | ID: mdl-24647947

ABSTRACT

The medial habenula (MHb) densely expresses nicotinic acetylcholine receptors (nAChRs) and participates in nicotine-related behaviors such as nicotine withdrawal and regulating nicotine intake. Although specific nAChR subunits are identified as being involved in withdrawal behavior, the cellular mechanisms through which nicotine acts to cause this aversive experience is unclear. Here, we demonstrate an interaction between the nicotinic and neurokinin signaling systems that may form the basis for some symptoms experienced during nicotine withdrawal. Using patch-clamp electrophysiology in mouse brain slices, we show that nicotine (1 µm) increases intrinsic excitability in MHb neurons. This nicotine-induced phenomenon requires α5-containing nAChRs and depends on intact neurokinin signaling. The effect is blocked by preincubation with neurokinin 1 (NK1; L-732138, 10 µm) and NK3 (SB222200, 2 µm) antagonists and mimicked by NK1 (substance P, 100 nm) and NK3 (neurokinin B [NKB], 100 nm) agonists. Microinjections (1 µl) of L-732138 (50 nm) and SB222200 (100 nm) into the MHb induces withdrawal behavior in chronic nicotine-treated (8.4 mg/kg/d, 2 weeks) mice. Conversely, withdrawal behavior is absent with analogous microinjections into the lateral habenula of nicotine-treated mice or in mice chronically treated with a vehicle solution. Further, chronic nicotine reduces nicotine's acute modulation of intrinsic excitability while sparing modulation by NKB. Our work elucidates the interplay between two neuromodulatory signaling systems in the brain through which nicotine acts to influence intrinsic excitability. More importantly, we document a neuroadaptation of this mechanism to chronic nicotine exposure and implicate these mechanisms collectively in the emergence of nicotine withdrawal behavior.


Subject(s)
Habenula/drug effects , Neurons/drug effects , Nicotine/pharmacology , Receptors, Neurokinin-1/metabolism , Receptors, Neurokinin-3/metabolism , Signal Transduction/drug effects , Action Potentials/drug effects , Animals , Female , Habenula/cytology , Habenula/metabolism , Male , Mice , Mice, Inbred C57BL , Neurokinin B/pharmacology , Neurokinin-1 Receptor Antagonists/pharmacology , Neurons/cytology , Neurons/metabolism , Receptors, Neurokinin-3/antagonists & inhibitors , Receptors, Nicotinic/metabolism , Substance P/pharmacology , Substance Withdrawal Syndrome/metabolism
6.
Nicotine Tob Res ; 15(5): 983-6, 2013 May.
Article in English | MEDLINE | ID: mdl-23042983

ABSTRACT

INTRODUCTION: Tobacco addiction has a strong social component. Therefore, nicotinic acetylcholine receptors (nAChR) may influence social behavior. Because the ß4 nicotinic receptor subunit is important for possibly related behaviors, such as anxiety-like behavior and the effects of nicotine, we studied the social behavior of mice null for the ß4 nAChR subunit. METHODS: To measure social behavior, we used the intruder test for social memory in wild-type and littermate ß4 null mice. In addition, we used a nonsocial olfactory memory test as a control. RESULTS: In the intruder test, ß4 null mice showed social amnesia: Wild-type mice spent less time actively interacting with a younger intruder on Day 2 than on Day 1, but ß4 null mice interacted for a similar time on both days. In the nonsocial olfactory memory test, control littermates and ß4 null mice learnt the associations to a similar extent, showing that the amnesic phenotype in the intruder test is specific for social settings. CONCLUSIONS: We conclude that nAChRs that contain the ß4 subunit are important for social behaviors. As those receptors are necessary to observe several effects of nicotine including withdrawal, it is tempting to speculate that the social component of tobacco use is related to the same neuronal circuits responsible for continuing tobacco use in smokers.


Subject(s)
Behavior, Animal/physiology , Nerve Tissue Proteins/genetics , Receptors, Nicotinic/genetics , Social Behavior , Amnesia , Animals , Anxiety/genetics , Female , Male , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/metabolism , Phenotype , Receptors, Nicotinic/metabolism , Smell/genetics
7.
Neuropharmacology ; 235: 109547, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37116611

ABSTRACT

Nicotine is the principal psychoactive component in tobacco that drives addiction through its action on neuronal nicotinic acetylcholine receptors (nAChR). The nicotinic receptor gene CHRNA5, which encodes the α5 subunit, is associated with nicotine use and dependence. In humans, the CHRNA5 missense variant rs16969968 (G > A) is associated with increased risk for nicotine dependence and other smoking-related phenotypes. In rodents, α5-containing nAChRs in dopamine (DA) neurons within the ventral tegmental area (VTA) powerfully modulate nicotine reward and reinforcement. Although the neuroadaptations caused by long-term nicotine exposure are being actively delineated at both the synaptic and behavioral levels, the contribution of α5-containing nAChRs to the cellular adaptations associated with long-term nicotine exposure remain largely unknown. To gain insight into the mechanisms behind the influence of α5-containing nAChRs and the rs16969968 polymorphism on nicotine use and dependence, we used electrophysiological approaches to examine changes in nAChR function arising in VTA neurons during chronic nicotine exposure and multiple stages of nicotine withdrawal. Our results demonstrate that CHRNA5 mutation leads to profound changes in VTA nAChR function at baseline, during chronic nicotine exposure, and during short-term and prolonged withdrawal. Whereas nAChR function was suppressed in DA neurons from WT mice undergoing withdrawal relative to drug-naïve or nicotine-drinking mice, α5-null mice exhibited an increase in nAChR function during nicotine exposure that persisted throughout 5-10 weeks of withdrawal. Re-expressing the hypofunctional rs16969968 CHRNA5 variant in α5-null VTA DA neurons did not rescue the phenotype, with α5-SNP neurons displaying a similar increased response to ACh during nicotine exposure and early stages of withdrawal. These results demonstrate the importance of VTA α5-nAChRs in the response to nicotine and implicate them in the time course of withdrawal.


Subject(s)
Nicotine , Receptors, Nicotinic , Humans , Mice , Animals , Nicotine/pharmacology , Dopaminergic Neurons/metabolism , Ventral Tegmental Area/metabolism , Receptors, Nicotinic/metabolism , Smoking , Mice, Knockout , Nerve Tissue Proteins/genetics
8.
eNeuro ; 10(6)2023 06.
Article in English | MEDLINE | ID: mdl-37253590

ABSTRACT

Nicotine is an addictive drug whose popularity has recently increased, particularly among adolescents, because of the availability of electronic nicotine devices (i.e., "vaping") and nicotine e-liquids containing additives with rich chemosensory properties. Some efforts to understand the role of these additives in nicotine reward suggest that they increase nicotine reward and reinforcement, but the sensory contributions of additives, especially in their vapor forms, are largely untested. Here, to better understand how a fruit-flavored (i.e., strawberry) additive influences nicotine reward and aversion, we used a conditioned place preference (CPP) procedure in which nicotine and a strawberry additive were delivered as a vapor to male and female adolescent mice. We found that nicotine vapor alone can lead to a dose-dependent CPP when using a biased design. The strawberry additive did not produce CPP on its own, and we did not observe an effect of the strawberry additive on nicotine vapor-induced reward. Nevertheless, mice exposed to nicotine plus strawberry additive vapor had higher plasma cotinine concentrations, which did not appear to reflect altered nicotine metabolism. Instead, by directly measuring vapor sampling through respiration monitoring, we uncovered an increase in the amount of sniffing toward strawberry-containing nicotine vapor compared with nicotine vapor alone. Together these data indicate that chemosensory-rich e-liquid additives may enhance the perceived sensory profile of nicotine vapors rather than the reward value per se, which leads to overall increased nicotine exposure.


Subject(s)
Electronic Nicotine Delivery Systems , Fragaria , Vaping , Male , Female , Mice , Animals , Nicotine/pharmacology , Nicotine/metabolism , Fragaria/metabolism , Reward
9.
J Neurosci ; 31(45): 16458-63, 2011 Nov 09.
Article in English | MEDLINE | ID: mdl-22072695

ABSTRACT

Attention depends on cholinergic stimulation of nicotinic and muscarinic acetylcholine receptors in the medial prefrontal cortex. Pyramidal neurons in layer VI of this region express cholinergic receptors of both families and play an important role in attention through their feedback projections to the thalamus. Here, we investigate how nicotinic and muscarinic cholinergic receptors affect the excitability of these neurons using whole-cell recordings in acute brain slices of prefrontal cortex. Since attention deficits have been documented in both rodents and humans having genetic abnormalities in nicotinic receptors, we focus in particular on how the cholinergic excitation of layer VI neurons is altered by genetic deletion of either of two key nicotinic receptor subunits, the accessory α5 subunit or the ligand-binding ß2 subunit. We find that the cholinergic excitation of layer VI neurons is dominated by nicotinic receptors in wild-type mice and that the reduction or loss of this nicotinic stimulation is accompanied by a surprising degree of plasticity in excitatory muscarinic receptors. These findings suggest that disrupting nicotinic receptors fundamentally alters the mechanisms and timing of excitation in prefrontal attentional circuitry.


Subject(s)
Neuronal Plasticity/genetics , Neurons/physiology , Prefrontal Cortex/cytology , Receptors, Muscarinic/metabolism , Receptors, Nicotinic/deficiency , Signal Transduction/genetics , Up-Regulation/genetics , Acetylcholine/pharmacology , Aconitine/analogs & derivatives , Aconitine/pharmacology , Age Factors , Analysis of Variance , Animals , Atropine/pharmacology , Dihydro-beta-Erythroidine/pharmacology , Intracellular Signaling Peptides and Proteins/pharmacology , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Knockout , Neuronal Plasticity/drug effects , Neurons/drug effects , Neuropeptides/pharmacology , Neurotransmitter Agents/pharmacology , Nicotine/metabolism , Nicotinic Antagonists/pharmacology , Orexins , Patch-Clamp Techniques/methods , Receptors, Muscarinic/genetics
10.
J Neurochem ; 122(1): 24-37, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22486777

ABSTRACT

The metabotropic glutamate receptors (mGluRs) fine-tune the efficacy of synaptic transmission. This unique feature makes mGluRs potential targets for the treatment of various CNS disorders. There is ample evidence to show that the ubiquitin proteasome system mediates changes in synaptic strength leading to multiple forms of synaptic plasticity. The present study describes a novel interaction between post-synaptic adaptors, long Homer-3 proteins, and one of the 26S proteasome regulatory subunits, the S8 ATPase, that influences the degradation of the metabotropic glutamate receptor 1α (mGluR1α). We have shown that the two human long Homer-3 proteins specifically interact with human proteasomal S8 ATPase. We identified that mGluR1α and long Homer-3s immunoprecipitate with the 26S proteasome both in vitro and in vivo. We further found that the mGluR1α receptor can be ubiquitinated and degraded by the 26S proteasome and that Homer-3A facilitates this process. Furthermore, the siRNA mediated silencing of Homer-3 led to increased levels of total and plasma membrane-associated mGluR1α receptors. These results suggest that long Homer-3 proteins control the degradation of mGluR1α receptors by shuttling ubiquitinated mGluR-1α receptors to the 26S proteasome via the S8 ATPase which may modulate synaptic transmission.


Subject(s)
Carrier Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction/physiology , ATPases Associated with Diverse Cellular Activities , Animals , Cadherins/metabolism , Calnexin/metabolism , Carrier Proteins/genetics , Cells, Cultured , Cerebral Cortex/cytology , Embryo, Mammalian , Hippocampus/cytology , Homer Scaffolding Proteins , Humans , Neurons/metabolism , Proteasome Endopeptidase Complex/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/genetics , Transfection , Ubiquitination/physiology
11.
Front Neurosci ; 16: 893587, 2022.
Article in English | MEDLINE | ID: mdl-35928010

ABSTRACT

While rates of smoking combustible cigarettes in the United States have trended down in recent years, use of electronic cigarettes (e-cigarettes) has dramatically increased, especially among adolescents. The vast majority of e-cigarette users consume "flavored" products that contain a variety of chemosensory-rich additives, and recent literature suggests that these additives have led to the current "teen vaping epidemic." This review, covering research from both human and rodent models, provides a comprehensive overview of the sensory implications of e-cigarette additives and what is currently known about their impact on nicotine use. In doing so, we specifically address the oronasal sensory contributions of e-cigarette additives. Finally, we summarize the existing gaps in the field and highlight future directions needed to better understand the powerful influence of these additives on nicotine use.

12.
Neuropharmacology ; 218: 109218, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35973602

ABSTRACT

The single nucleotide polymorphism (SNP) D398N (rs16969968) in CHRNA5, the gene encoding the α5 subunit of the nicotinic acetylcholine receptors (nAChR), has been associated with both nicotine and opiate dependence in human populations. Expression of this SNP on presynaptic VTA dopaminergic (DA) neurons is known to cause a reduction in calcium signaling, leading to alterations in transmitter signaling and altered responses to drugs of abuse. To examine the impact of the Chrna5 SNP on opiate reward and underlying dopaminergic mechanisms, mice harboring two copies of the risk-associated allele (Chrna5 A/A) at a location equivalent to human rs16969968 were generated via CRISPR/cas9 genome editing. We sought to determine whether Chrna5 A/A mice show differences in sensitivity to rewarding properties of morphine using the conditioned place preference paradigm. When mice were tested two weeks after conditioning, female Chrna5 A/A mice showed significantly enhanced preference for the morphine-paired chamber relative to WT females, suggesting that this genotype may enhance opioid reward specifically in females. In contrast, Chrna5 genotype had no effect on locomotor sensitization in male or female mice. Relative to WT females, peak amplitude of ACh-gated currents recorded from VTA DA neurons in Chrna5 A/A females was potentiated 1 day after conditioning with morphine. Increased FOS expression was also observed in Chrna5 A/A mice relative to WT mice following exposure to the morphine CPP chamber. We propose that impaired α5 nAChR subunit function alters DA neuron response following repeated morphine exposures, and that this early cellular response could contribute to enhanced opiate reward two weeks after conditioning.


Subject(s)
Polymorphism, Single Nucleotide , Receptors, Nicotinic , Animals , Female , Male , Mice , Morphine/pharmacology , Nerve Tissue Proteins/metabolism , Nicotine/pharmacology , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Reward
13.
Elife ; 112022 03 15.
Article in English | MEDLINE | ID: mdl-35290181

ABSTRACT

Sensory systems must account for both contextual factors and prior experience to adaptively engage with the dynamic external environment. In the central auditory system, neurons modulate their responses to sounds based on statistical context. These response modulations can be understood through a hierarchical predictive coding lens: responses to repeated stimuli are progressively decreased, in a process known as repetition suppression, whereas unexpected stimuli produce a prediction error signal. Prediction error incrementally increases along the auditory hierarchy from the inferior colliculus (IC) to the auditory cortex (AC), suggesting that these regions may engage in hierarchical predictive coding. A potential substrate for top-down predictive cues is the massive set of descending projections from the AC to subcortical structures, although the role of this system in predictive processing has never been directly assessed. We tested the effect of optogenetic inactivation of the auditory cortico-collicular feedback in awake mice on responses of IC neurons to stimuli designed to test prediction error and repetition suppression. Inactivation of the cortico-collicular pathway led to a decrease in prediction error in IC. Repetition suppression was unaffected by cortico-collicular inactivation, suggesting that this metric may reflect fatigue of bottom-up sensory inputs rather than predictive processing. We also discovered populations of IC units that exhibit repetition enhancement, a sequential increase in firing with stimulus repetition. Cortico-collicular inactivation led to a decrease in repetition enhancement in the central nucleus of IC, suggesting that it is a top-down phenomenon. Negative prediction error, a stronger response to a tone in a predictable rather than unpredictable sequence, was suppressed in shell IC units during cortico-collicular inactivation. These changes in predictive coding metrics arose from bidirectional modulations in the response to the standard and deviant contexts, such that the units in IC responded more similarly to each context in the absence of cortical input. We also investigated how these metrics compare between the anesthetized and awake states by recording from the same units under both conditions. We found that metrics of predictive coding and deviance detection differ depending on the anesthetic state of the animal, with negative prediction error emerging in the central IC and repetition enhancement and prediction error being more prevalent in the absence of anesthesia. Overall, our results demonstrate that the AC provides cues about the statistical context of sound to subcortical brain regions via direct feedback, regulating processing of both prediction and repetition.


Subject(s)
Auditory Cortex , Inferior Colliculi , Acoustic Stimulation , Animals , Auditory Cortex/physiology , Auditory Pathways/physiology , Auditory Perception/physiology , Inferior Colliculi/physiology , Mice , Optogenetics
14.
Neuropharmacology ; 216: 109170, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35752273

ABSTRACT

Alcohol and nicotine are commonly used during adolescence, establishing long-lasting neuroplastic alterations that influence subsequent drug use and abuse. Drinking- and smoking-related traits have been extensively associated with variation in CHRNA5 - the gene that encodes the α5 subunit of neuronal nicotinic acetylcholine receptors (nAChRs). The single nucleotide polymorphism (SNP) rs16969968 in CHRNA5 encodes an amino acid substitution (D398N) that alters the function and pharmacokinetics of α5-containing nAChR. When expressed in rodents, this variant results in increased ethanol and nicotine operant self-administration. How disruption of α5-containing nAChRs influences adolescent ethanol and nicotine intake, and how it modulates interactions between these drugs has not been previously explored. In the present study, we examined volitional ethanol and nicotine consumption in adolescent mice (post-natal day 30-43) of both sexes with mutated (SNP) or lacking (KO) the α5 nAChR subunit. The effect of adolescent alcohol or nicotine exposure on home cage consumption of the opposite drug in adulthood and its modulation by Chrna5 mutation and sex were examined. During adolescence, we found that α5 nAChR disruption increases nicotine intake in mice of both sexes, but the effect on alcohol intake was only observed in females. The sex-specific increase in alcohol consumption in α5 SNP and KO was replicated in adulthood. The effect of adolescent alcohol or nicotine exposure on subsequent intake of the opposite drug in adulthood is modulated by sex and Chrna5 mutation. These observations suggest sex differences in the genetic architecture of alcohol dependence, and modulators of alcohol and nicotine interactions.


Subject(s)
Receptors, Nicotinic/metabolism , Animals , Ethanol , Female , Male , Mice , Mutation , Nicotine , Smoking
15.
J Neurosci ; 30(27): 9241-52, 2010 Jul 07.
Article in English | MEDLINE | ID: mdl-20610759

ABSTRACT

Stimulation of the prefrontal cortex by acetylcholine is critical for attention; however, the cellular mechanisms underlying its influence on attention pathways within the brain are not well understood. Pyramidal neurons in layer VI of the prefrontal cortex are believed to play an important role in this process because they are excited by acetylcholine and provide a major source of feedback projections to the thalamus. Here, we show using whole-cell electrophysiology that the relatively rare alpha5 subunit of the nicotinic acetylcholine receptor powerfully enhances nicotinic currents in layer VI pyramidal neurons in prefrontal cortical brain slices from adult mice. In addition, behavioral experiments using the five-choice serial reaction time test show that the presence of the nicotinic receptor alpha5 subunit also increases the accuracy of adult mice on this visual attention task under highly demanding conditions. Together, these findings demonstrate a novel and important role for the nicotinic receptor alpha5 subunit in adult brain circuitry required for attentional performance.


Subject(s)
Attention/physiology , Pyramidal Cells/physiology , Receptors, Nicotinic/physiology , Acetylcholine/pharmacology , Analysis of Variance , Anesthetics, Local/pharmacology , Animals , Attention/drug effects , Choice Behavior/physiology , Cholinergic Agents/pharmacology , Dihydro-beta-Erythroidine/pharmacology , Dose-Response Relationship, Drug , In Vitro Techniques , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Motor Activity/genetics , Nerve Net/physiology , Neuropsychological Tests , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/pharmacology , Patch-Clamp Techniques/methods , Photic Stimulation/methods , Prefrontal Cortex/cytology , Reaction Time/drug effects , Reaction Time/genetics , Receptors, Nicotinic/deficiency , Tetrodotoxin/pharmacology , gamma-Aminobutyric Acid/metabolism
16.
FASEB J ; 24(12): 5113-23, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20739611

ABSTRACT

α6* (asterisk indicates the presence of additional subunits) nicotinic acetylcholine receptors (nAChRs) are broadly implicated in catecholamine-dependent disorders that involve attention, motor movement, and nicotine self-administration. Different molecular forms of α6 nAChRs mediate catecholamine release, but receptor differentiation is greatly hampered by a paucity of subtype selective ligands. α-Conotoxins are nAChR-targeted peptides used by Conus species to incapacitate prey. We hypothesized that distinct conotoxin-binding kinetics could be exploited to develop a series of selective probes to enable study of native receptor subtypes. Proline6 of α-conotoxin BuIA was found to be critical for nAChR selectivity; substitution of proline6 with 4-hydroyxproline increased the IC(50) by 2800-fold at α6/α3ß2ß3 but only by 6-fold at α6/α3ß4 nAChRs (to 1300 and 12 nM, respectively). We used conotoxin probes together with subunit-null mice to interrogate nAChR subtypes that modulate hippocampal norepinephrine release. Release was abolished in α6-null mutant mice. α-Conotoxin BuIA[T5A;P6O] partially blocked norepinephrine release in wild-type controls but failed to block release in ß4(-/-) mice. In contrast, BuIA[T5A;P6O] failed to block dopamine release in the wild-type striatum known to contain α6ß2* nAChRs. BuIA[T5A;P6O] is a novel ligand for distinguishing between closely related α6* nAChRs; α6ß4* nAChRs modulate norepinephrine release in hippocampus but not dopamine release in striatum.


Subject(s)
Conotoxins/metabolism , Nicotine/metabolism , Norepinephrine/metabolism , Receptors, Nicotinic/metabolism , Animals , Female , Hydroxylation , Ligands , Male , Mice , Mice, Inbred C57BL , Oocytes/metabolism , Proline/metabolism , Xenopus
17.
Neuron ; 109(5): 746-747, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33662269

ABSTRACT

The circuit that links stress and fear to feeding behavior is poorly understood. In this issue of Neuron, Yang et al. detail a trisynaptic, cannabinoid-dependent circuit that underlies appetite suppression in response to a fearful stimulus and provide evidence of noradrenaline and glutamate co-transmission in locus coeruleus.


Subject(s)
Appetite , Locus Coeruleus , Fear , Neurons , Norepinephrine
18.
Neuropharmacology ; 195: 108672, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34153314

ABSTRACT

The rise of e-cigarette popularity has sparked interest in the role of palatable flavors on nicotine use. Despite growing evidence that sweet flavorants enhance nicotine reward, their influence on nicotine consumption has not been studied extensively. In addition, the impact that flavored nicotine use in adolescence could have on nicotine reward and dependence in adulthood remains unclear. This study examined the role of flavored nicotine access on nicotine preference and consumption longitudinally, from adolescence to adulthood. Male and female adolescent mice preferred a fruit-flavored nicotine solution over an unflavored nicotine solution. However, only adolescent female mice with access to flavored nicotine consumed higher doses. Furthermore, while adolescent male mice escalated consumption of both flavored and unflavored nicotine, female mice only escalated nicotine consumption when given access to flavored nicotine. As mice matured into adulthood, there was no evidence that a history of flavored-nicotine access altered preference for unflavored nicotine compared to a nicotine-free control in a classic two-bottle choice design. However, when the nicotine concentration was progressively reduced, mice that had consumed strawberry-flavored nicotine in adolescence maintained baseline nicotine consumption levels longer than mice that initiated nicotine use without flavor in adolescence. Finally, addition of fruit-flavorants into the nicotine solution during adulthood led to nicotine preference and increased levels of nicotine consumption, regardless of previous flavored-nicotine access or of familiarity with the selected flavorant. These results indicate that flavorants increase nicotine consumption independent of life stage, possibly posing a disproportionate risk to adolescent females. Our results also point to an effect of adolescent flavored-nicotine use on nicotine dose maintenance in adulthood, which could have implications for the success of future quit attempts.


Subject(s)
Flavoring Agents/administration & dosage , Fruit , Nicotine/administration & dosage , Animals , Choice Behavior/drug effects , Female , Male , Mice , Self Administration , Sex Factors
19.
Neuropharmacology ; 199: 108783, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34509497

ABSTRACT

Alcohol use disorder (AUD) is a neuropsychiatric condition affecting millions of people worldwide. Topiramate (TPM) is an antiepileptic drug that has been shown to reduce ethanol drinking in humans. However, TPM is associated with a variety of adverse effects due to its interaction with many receptor systems and intracellular pathways. GluK1-containing kainate receptors (GluK1*KARs) are non-selectively inhibited by TPM, and genetic association studies suggest that this receptor system could be targeted to reduce drinking in AUD patients. We examined the efficacy of LY466195, a selective inhibitor of GluK1*KAR, in reducing ethanol consumption in the intermittent two-bottle choice paradigm in mice. The effect of LY466195 on various ethanol-related phenotypes was investigated by quantification of alcohol intake, physical signs of withdrawal, conditioned place preference (CPP) and in vivo microdialysis in the nucleus accumbens. Selective GluK1*KAR inhibition reduced ethanol intake and preference in a dose-dependent manner. LY466195 treatment attenuated the physical manifestations of ethanol withdrawal and influenced the rewarding properties of ethanol. Interestingly, LY466195 injection also normalized changes in dopamine levels in response to acute ethanol in ethanol-dependent mice, but had no effect in ethanol-naïve mice, suggesting ethanol state-dependent effects. The data point to GluK1*KARs as an attractive pharmacological target for the treatment of AUD.


Subject(s)
Alcohol Drinking/drug therapy , Alcoholism/drug therapy , Excitatory Amino Acid Antagonists/pharmacology , Isoquinolines/pharmacology , Receptors, Kainic Acid/antagonists & inhibitors , Reward , Substance Withdrawal Syndrome/drug therapy , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/administration & dosage , Isoquinolines/administration & dosage , Mice , Receptors, Ionotropic Glutamate
20.
Neuropsychopharmacology ; 46(10): 1757-1767, 2021 09.
Article in English | MEDLINE | ID: mdl-33953341

ABSTRACT

A major theme of addiction research has focused on the neural substrates of individual differences in the risk for addiction; however, little is known about how vulnerable populations differ from those that are relatively protected. Here, we prospectively measured dopamine (DA) neurotransmission prior to cocaine exposure to predict the onset and course of cocaine use. Using in vivo voltammetry, we first generated baseline profiles of DA release and uptake in the dorsomedial striatum (DMS) and nucleus accumbens of drug-naïve male rats prior to exposing them to cocaine using conditioned place preference (CPP) or operant self-administration. We found that the innate rate of DA uptake in the DMS strongly predicted motivation for cocaine and drug-primed reinstatement, but not CPP, responding when "price" was low, or extinction. We then assessed the impact of baseline variations in DA uptake on cocaine potency in the DMS using ex vivo voltammetry in naïve rats and in rats with DA transporter (DAT) knockdown. DA uptake in the DMS of naïve rats predicted the neurochemical response to cocaine, such that rats with innately faster rates of DA uptake demonstrated higher cocaine potency at the DAT and rats with DAT knockdown displayed reduced potency compared to controls. Together, these data demonstrate that inherent variability in DA uptake in the DMS predicts the behavioral response to cocaine, potentially by altering the apparent potency of cocaine.


Subject(s)
Cocaine , Animals , Cocaine/pharmacology , Dopamine , Dopamine Uptake Inhibitors/pharmacology , Individuality , Male , Motivation , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL