Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters










Publication year range
1.
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
2.
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
3.
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.

4.
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
5.
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
6.
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
7.
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
8.
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
10.
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
11.
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
12.
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
13.
Neuropharmacology ; 177: 108256, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32738308

ABSTRACT

Nicotine is a highly addictive drug found in tobacco that drives its continued use despite the harmful consequences. The initiation of nicotine abuse involves the mesolimbic dopamine system, which contributes to the rewarding sensory stimuli and associative learning processes in the beginning stages of addiction. Nicotine binds to neuronal nicotinic acetylcholine receptors (nAChRs), which come in a diverse collection of subtypes. The nAChRs that contain the α4 and ß2 subunits, often in combination with the α6 subunit, are particularly important for nicotine's ability to increase midbrain dopamine neuron firing rates and phasic burst firing. Chronic nicotine exposure results in numerous neuroadaptations, including the upregulation of particular nAChR subtypes associated with long-term desensitization of the receptors. When nicotine is no longer present, for example during attempts to quit smoking, a withdrawal syndrome develops. The expression of physical withdrawal symptoms depends mainly on the α2, α3, α5, and ß4 nicotinic subunits in the epithalamic habenular complex and its target regions. Thus, nicotine affects diverse neural systems and an array of nAChR subtypes to mediate the overall addiction process. This article is part of the special issue on 'Contemporary Advances in Nicotine Neuropharmacology'.


Subject(s)
Brain/metabolism , Nicotine/metabolism , Receptors, Nicotinic/metabolism , Tobacco Use Disorder/metabolism , Animals , Brain/drug effects , Humans , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Nicotinic Agonists/metabolism , Nicotinic Antagonists/administration & dosage , Nicotinic Antagonists/metabolism , Protein Subunits/agonists , Protein Subunits/antagonists & inhibitors , Protein Subunits/chemistry , Protein Subunits/metabolism , Receptors, Nicotinic/chemistry , Tobacco Use Disorder/psychology
14.
Neuropharmacology ; 177: 108162, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32497589

ABSTRACT

The popularity of e-cigarettes has skyrocketed in recent years, and most vapers use flavored e-cigarette products. Consumption of flavored e-cigarettes exceeds that of combustible cigarettes and other tobacco products among adolescents, who are particularly vulnerable to becoming nicotine dependent. Flavorings have been used by the tobacco industry since the 17th century, but the use of flavors by the e-cigarette industry to create products with "characterizing" flavors (i.e. flavors other than tobacco or menthol) has sparked a public health debate. This review addresses the possibility that characterizing flavors make nicotine more appealing, rewarding and addictive. It also discusses ways in which preclinical and clinical studies could improve our understanding of the mechanisms by which flavors may alter nicotine reward and reinforcement. This article is part of the special issue on 'Contemporary Advances in Nicotine Neuropharmacology'.


Subject(s)
Electronic Nicotine Delivery Systems , Flavoring Agents/administration & dosage , Nicotine/administration & dosage , Tobacco Use Disorder/psychology , Vaping/psychology , Vaping/trends , Animals , Flavoring Agents/adverse effects , Humans , Nicotine/adverse effects , Tobacco Use Disorder/diagnosis , Tobacco Use Disorder/prevention & control , Vaping/adverse effects
15.
Curr Top Behav Neurosci ; 45: 123-151, 2020.
Article in English | MEDLINE | ID: mdl-32451953

ABSTRACT

Human behavior can be controlled by physical or psychological dependencies associated with addiction. One of the most insidious addictions in our society is the use of tobacco products which contain nicotine. This addiction can be associated with specific receptors in the brain that respond to the natural neurotransmitter acetylcholine. These nicotinic acetylcholine receptors (nAChR) are ligand-gated ion channels formed by the assembly of one or multiple types of nAChR receptor subunits. In this paper, we review the structure and diversity of nAChR subunits and our understanding for how different nAChR subtypes play specific roles in the phenomenon of nicotine addiction. We focus on receptors containing ß2 and/or α6 subunits and the special significance of α5-containing receptors. These subtypes all have roles in regulating dopamine-mediated neurotransmission in the mesolimbic reward pathways of the brain. We also discuss the unique roles of homomeric α7 nAChR in behavioral responses to nicotine and how our knowledge of nAChR functional diversity may help guide pharmacotherapeutic approaches for treating nicotine addiction. While nicotine addiction is a truly global problem, the use of areca nut (betel) products is also a serious addiction associated with public health issues across most of South Asia, impacting as many as 600 million people. We discuss how cholinergic receptors of the brain are also involved with areca addiction and the unique challenges for dealing with addiction to this substance.


Subject(s)
Behavior, Addictive , Receptors, Nicotinic , Tobacco Use Disorder , Humans , Nicotine , Receptors, Cholinergic
16.
Elife ; 92020 01 31.
Article in English | MEDLINE | ID: mdl-32003747

ABSTRACT

The extensive feedback from the auditory cortex (AC) to the inferior colliculus (IC) supports critical aspects of auditory behavior but has not been extensively characterized. Previous studies demonstrated that activity in IC is altered by focal electrical stimulation and pharmacological inactivation of AC, but these methods lack the ability to selectively manipulate projection neurons. We measured the effects of selective optogenetic modulation of cortico-collicular feedback projections on IC sound responses in mice. Activation of feedback increased spontaneous activity and decreased stimulus selectivity in IC, whereas suppression had no effect. To further understand how microcircuits in AC may control collicular activity, we optogenetically modulated the activity of different cortical neuronal subtypes, specifically parvalbumin-positive (PV) and somatostatin-positive (SST) inhibitory interneurons. We found that modulating the activity of either type of interneuron did not affect IC sound-evoked activity. Combined, our results identify that activation of excitatory projections, but not inhibition-driven changes in cortical activity, affects collicular sound responses.


How do we hear the world around us? Hearing begins when hair cells in the inner ear translate incoming sound waves into electrical signals. These signals travel via the auditory nerve and the brainstem to the midbrain, where an area called the inferior colliculus processes them. The inferior colliculus then passes the signals on to another area deep within the brain, the thalamus, which processes the signals further before it too passes them on to an area of the brain's outer layer called the auditory cortex. At each stage of the auditory pathway, the signals undergo more complex processing than at the previous stage. Researchers have tended to think of this pathway as a one-way route from the ear to the brain. But in reality, feedback occurs at various points along the pathway, enabling areas that do higher processing to shape the responses of areas earlier in the pathway. This feedback is particularly prevalent in the auditory system, where one such strong feedback route is from the auditory cortex to the inferior colliculus. This reverse connection helps animals learn new behavioral responses to sounds, for example, to run away from a loud noise. By manipulating the activity of this pathway in mice using a technique called optogenetics, Blackwell et al. provide further clues to how the auditory pathway works. Optogenetics involves introducing light-sensitive ion channels into neurons, and then using light to activate or inhibit those neurons on demand. Blackwell et al. show that activating the feedback pathway from the auditory cortex to the inferior colliculus in awake mice changes how the inferior colliculus responds to sounds. By contrast, inhibiting the pathway has no effect on inferior colliculus responses. This suggests that the feedback pathway is not active all the time, but instead influences inferior colliculus activity only during specific behavior, for example, perhaps when we are listening for a specific sound like the ringing of a phone. Understanding how the brain processes sound is important for understanding how we communicate and why we appreciate music. It could also help in treating hearing loss. Stimulating the inferior colliculus using a device implanted in the brainstem can improve hearing in people with certain types of deafness. Strengthening or weakening the feedback pathway from the auditory cortex to the inferior colliculus could make these implants more effective. In the future, it may even be possible that stimulating the pathway directly could restore hearing without any implant being required.


Subject(s)
Auditory Cortex/physiology , Inferior Colliculi/physiology , Acoustic Stimulation , Animals , Feedback, Sensory/physiology , Female , Interneurons/physiology , Male , Mice , Mice, Inbred C57BL , Optogenetics
17.
Nat Commun ; 9(1): 3221, 2018 08 13.
Article in English | MEDLINE | ID: mdl-30104567

ABSTRACT

Genome-wide association studies (GWAS) identified the chromosome 15q25.1 locus as a leading susceptibility region for lung cancer. However, the pathogenic pathways, through which susceptibility SNPs within chromosome 15q25.1 affects lung cancer risk, have not been explored. We analyzed three cohorts with GWAS data consisting 42,901 individuals and lung expression quantitative trait loci (eQTL) data on 409 individuals to identify and validate the underlying pathways and to investigate the combined effect of genes from the identified susceptibility pathways. The KEGG neuroactive ligand receptor interaction pathway, two Reactome pathways, and 22 Gene Ontology terms were identified and replicated to be significantly associated with lung cancer risk, with P values less than 0.05 and FDR less than 0.1. Functional annotation of eQTL analysis results showed that the neuroactive ligand receptor interaction pathway and gated channel activity were involved in lung cancer risk. These pathways provide important insights for the etiology of lung cancer.


Subject(s)
Chromosomes, Human, Pair 15/genetics , Genetic Predisposition to Disease , Lung Neoplasms/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Cohort Studies , Female , Gene Ontology , Gene Regulatory Networks , Humans , Infant , Infant, Newborn , Male , Middle Aged , Polymorphism, Single Nucleotide , Quantitative Trait Loci/genetics , Reproducibility of Results , Risk Factors , Smoking/adverse effects , Young Adult
18.
Neuropharmacology ; 138: 341-348, 2018 08.
Article in English | MEDLINE | ID: mdl-29944862

ABSTRACT

Evidence suggests that there is an association between polymorphisms in the α5 nicotinic acetylcholine receptor (nAChR) subunit and risk of developing alcohol dependence in humans. The α5 nAChR subunit has also recently been shown to modulate some of the acute response to ethanol in mice. The aim of the current study was to further characterize the role of α5-containing (α5*) nAChRs in acute ethanol responsive behaviors, ethanol consumption and ethanol preference in mice. We conducted a battery of tests in male α5 knockout (KO) mice for a range of ethanol-induced behaviors including hypothermia, hypnosis, and anxiolysis. We also investigated the effects of α5* nAChR on ethanol reward using the Conditioned Place Preference (CPP) assay. Further, we tested the effects of gene deletion on drinking behaviors using the voluntary ethanol consumption in a two-bottle choice assay and Drinking in the Dark (DID, with or without stress) paradigm. We found that deletion of the α5 nAChR subunit enhanced ethanol-induced hypothermia, hypnosis, and an anxiolytic-like response in comparison to wild-type controls. The α5 KO mice showed reduced CPP for ethanol, suggesting that the rewarding properties of ethanol are decreased in mutant mice. Interestingly, Chrna5 gene deletion had no effect on basal ethanol drinking behavior, or ethanol metabolism, but did decrease ethanol intake in the DID paradigm following restraint stress. Taken together, we provide new evidence that α5 nAChRs are involved in some but not all of the behavioral effects of ethanol. Our results highlight the importance of nAChRs as a possible target for the treatment of alcohol dependence.


Subject(s)
Alcohol Drinking/metabolism , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Receptors, Nicotinic/deficiency , Reward , Alcohol Drinking/psychology , Animals , Anxiety/chemically induced , Anxiety/metabolism , Central Nervous System Depressants/blood , Choice Behavior/drug effects , Choice Behavior/physiology , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Ethanol/blood , Hypnotics and Sedatives/blood , Hypnotics and Sedatives/pharmacology , Hypothermia/chemically induced , Hypothermia/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Motor Activity/physiology , Receptors, Nicotinic/genetics , Reflex/drug effects , Reflex/physiology , Spatial Behavior/drug effects , Spatial Behavior/physiology
19.
Cancer Res ; 78(12): 3233-3242, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29661830

ABSTRACT

Mounting clinical and preclinical evidence supports a key role for sustained adrenergic signaling in the tumor microenvironment as a driver of tumor growth and progression. However, the mechanisms by which adrenergic neurotransmitters are delivered to the tumor microenvironment are not well understood. Here we present evidence for a feed-forward loop whereby adrenergic signaling leads to increased tumoral innervation. In response to catecholamines, tumor cells produced brain-derived neurotrophic factor (BDNF) in an ADRB3/cAMP/Epac/JNK-dependent manner. Elevated BDNF levels in the tumor microenvironment increased innervation by signaling through host neurotrophic receptor tyrosine kinase 2 receptors. In patients with cancer, high tumor nerve counts were significantly associated with increased BDNF and norepinephrine levels and decreased overall survival. Collectively, these data describe a novel pathway for tumor innervation, with resultant biological and clinical implications.Significance: Sustained adrenergic signaling promotes tumor growth and metastasis through BDNF-mediated tumoral innervation. Cancer Res; 78(12); 3233-42. ©2018 AACR.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Feedback, Physiological , Neoplasms/pathology , Norepinephrine/metabolism , Receptors, Adrenergic, beta-3/metabolism , Animals , Cell Line, Tumor , Cyclic AMP/metabolism , Female , Guanine Nucleotide Exchange Factors/metabolism , Humans , Membrane Glycoproteins/metabolism , Mice , Neoplasms/mortality , Peripheral Nerves/metabolism , Peripheral Nerves/pathology , Receptor, trkB/metabolism , Signal Transduction , Tumor Microenvironment/physiology , Xenograft Model Antitumor Assays
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...