Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Sci Rep ; 13(1): 8460, 2023 05 25.
Article in English | MEDLINE | ID: mdl-37231124

ABSTRACT

The effectiveness of current treatments for opioid use disorder (OUD) varies by sex. Our understanding of the neurobiological mechanisms mediating negative states during withdrawal is lacking, particularly with regard to sex differences. Based on preclinical research in male subjects, opioid withdrawal is accompanied by increased gamma-aminobutyric acid (GABA) release probability at synapses onto dopamine neurons in the ventral tegmental area (VTA). It is unclear, however, if the physiological consequences of morphine that were originally elucidated in male rodents extend to females. The effects of morphine on the induction of future synaptic plasticity are also unknown. Here, we show that inhibitory synaptic long-term potentiation (LTPGABA) is occluded in the VTA in male mice after repeated morphine injections and 1 day of withdrawal, while morphine-treated female mice maintain the ability to evoke LTPGABA and have basal GABA activity similar to controls. Our observation of this physiological difference between male and female mice connects previous reports of sex differences in areas upstream and downstream of the GABA-dopamine synapse in the VTA during opioid withdrawal. The sex differences highlight the mechanistic distinctions between males and females that can be targeted when designing and implementing treatments for OUD.


Subject(s)
Substance Withdrawal Syndrome , Ventral Tegmental Area , Female , Mice , Male , Animals , Analgesics, Opioid/pharmacology , Sex Characteristics , Narcotics/pharmacology , gamma-Aminobutyric Acid/pharmacology , Morphine/adverse effects , Neuronal Plasticity
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.
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
4.
Front Cell Neurosci ; 16: 887679, 2022.
Article in English | MEDLINE | ID: mdl-35496910

ABSTRACT

Processing of contextual information during a new episodic event is crucial for learning and memory. Neuromodulation in the hippocampus and prefrontal cortex plays an important role in the formation of associations between environmental cues and an aversive experience. Noradrenergic neurons in the locus coeruleus send dense projections to both regions, but their contribution to contextual associative learning has not been established. Here, we utilize selective optogenetic and pharmacological manipulations to control noradrenergic transmission in the hippocampus during the encoding of a contextual fear memory. We find that boosting noradrenergic terminal release in the dorsal CA1 enhances the acquisition of contextual associative learning and that this effect requires local activation of ß-adrenenergic receptors. Moreover, we show that increasing norepinephrine release can ameliorate contextual fear learning impairments caused by dopaminergic dysregulation in the hippocampus. Our data suggest that increasing of hippocampal noradrenergic activity can have important implications in the treatment of cognitive disorders that involve problems in contextual processing.

5.
Addict Biol ; 27(2): e13147, 2022 03.
Article in English | MEDLINE | ID: mdl-35229942

ABSTRACT

Disruption of neuronal chloride ion (Cl- ) homeostasis has been linked to several pathological conditions, including substance use disorder, yet targeted pharmacotherapies are lacking. In this study, we explored the potential of serotonin 2A receptor (5-HT2A R) agonism to reduce alcohol consumption in male wild-type C57Bl/6J mice and to ameliorate alcohol-induced inhibitory plasticity in the midbrain. We found that administration of the putative 5-HT2A R agonist TCB-2 attenuated alcohol consumption and preference but did not alter water or saccharin consumption. We hypothesized that the selective behavioural effects of TCB-2 on alcohol drinking were due, at least in part, to effects of the agonist on ventral tegmental area (VTA) neurocircuitry. Alcohol consumption impairs Cl- transport in VTA GABA neurons, which acts as a molecular adaptation leading to increased alcohol self-administration. Using ex vivo electrophysiological recordings, we found that exposure to either intermittent volitional alcohol drinking or an acute alcohol injection diminished homeostatic Cl- transport in VTA GABA neurons. Critically, in vivo TCB-2 administration normalized Cl- transport in the VTA after alcohol exposure. Thus, we show a potent effect of alcohol consumption on VTA inhibitory circuitry, in the form of dysregulated Cl- homeostasis that is reversible with agonism of 5-HT2A Rs. Our results provide insight into the potential therapeutic action of 5-HT2A R agonists for alcohol abuse.


Subject(s)
Receptor, Serotonin, 5-HT2A , Ventral Tegmental Area , Alcohol Drinking , Animals , Ethanol/pharmacology , GABAergic Neurons , Male , Mice
6.
Front Behav Neurosci ; 16: 1092420, 2022.
Article in English | MEDLINE | ID: mdl-36778837

ABSTRACT

The hippocampus is responsible for encoding behavioral episodes into short-term and long-term memory. The circuits that mediate these processes are subject to neuromodulation, which involves regulation of synaptic plasticity and local neuronal excitability. In this review, we present evidence to demonstrate the influence of dopaminergic neuromodulation on hippocampus-dependent memory, and we address the controversy surrounding the source of dopamine innervation. First, we summarize historical and recent retrograde and anterograde anatomical tracing studies of direct dopaminergic projections from the ventral tegmental area and discuss dopamine release from the adrenergic locus coeruleus. Then, we present evidence of dopaminergic modulation of synaptic plasticity in the hippocampus. Plasticity mechanisms are examined in brain slices and in recordings from in vivo neuronal populations in freely moving rodents. Finally, we review pharmacological, genetic, and circuitry research that demonstrates the importance of dopamine release for learning and memory tasks while dissociating anatomically distinct populations of direct dopaminergic inputs.

7.
Proc Natl Acad Sci U S A ; 118(40)2021 10 05.
Article in English | MEDLINE | ID: mdl-34580198

ABSTRACT

Aversive memories are important for survival, and dopaminergic signaling in the hippocampus has been implicated in aversive learning. However, the source and mode of action of hippocampal dopamine remain controversial. Here, we utilize anterograde and retrograde viral tracing methods to label midbrain dopaminergic projections to the dorsal hippocampus. We identify a population of midbrain dopaminergic neurons near the border of the substantia nigra pars compacta and the lateral ventral tegmental area that sends direct projections to the dorsal hippocampus. Using optogenetic manipulations and mutant mice to control dopamine transmission in the hippocampus, we show that midbrain dopamine potently modulates aversive memory formation during encoding of contextual fear. Moreover, we demonstrate that dopaminergic transmission in the dorsal CA1 is required for the acquisition of contextual fear memories, and that this acquisition is sustained in the absence of catecholamine release from noradrenergic terminals. Our findings identify a cluster of midbrain dopamine neurons that innervate the hippocampus and show that the midbrain dopamine neuromodulation in the dorsal hippocampus is sufficient to maintain aversive memory formation.


Subject(s)
Dopamine/metabolism , Hippocampus/metabolism , Memory/physiology , Animals , Dopaminergic Neurons , Fear/physiology , Female , Learning/physiology , Male , Mesencephalon/physiology , Mice , Mice, Inbred C57BL , Optogenetics/methods , Substantia Nigra/metabolism , Ventral Tegmental Area/physiology
8.
Hippocampus ; 31(10): 1154-1175, 2021 10.
Article in English | MEDLINE | ID: mdl-34418215

ABSTRACT

The hippocampus is essential for spatial learning and memory. To assess learning we used contextual fear conditioning (cFC), where animals learn to associate a place with aversive events like foot-shocks. Candidate memory mechanisms for cFC are long-term potentiation (LTP) and long-term depression (LTD), but there is little direct evidence of them operating in the hippocampus in vivo following cFC. Also, little is known about the behavioral state changes induced by cFC. To address these issues, we recorded local field potentials in freely behaving mice by stimulating in the left dorsal CA1 region and recording in the right dorsal CA1 region. Synaptic strength in the commissural pathway was monitored by measuring field excitatory postsynaptic potentials (fEPSPs) before and after cFC. After cFC, the commissural pathway's synaptic strength was potentiated. Although recordings occurred during the wake phase of the light/dark cycle, the mice slept more in the post-conditioning period than in the pre-conditioning period. Relative to awake periods, in non-rapid eye movement (NREM) sleep the fEPSPs were larger in both pre- and post-conditioning periods. We also found a significant negative correlation between the animal's speed and fEPSP size. Therefore, to avoid confounds in the fEFSP potentiation estimates, we controlled for speed-related and sleep-related fEPSP changes and still found that cFC induced long-term potentiation, but no significant long-term depression. Synaptic strength changes were not found in the control group that simply explored the fear-conditioning chamber, indicating that exploration of the novel place did not produce the measurable effects caused by cFC. These results show that following cFC, the CA1 commissural pathway is potentiated, likely contributing to the functional integration of the left and right hippocampi in fear memory consolidation. In addition, the cFC paradigm produces significant changes in an animal's behavioral state, which are observable as proximal changes in sleep patterns.


Subject(s)
Fear , Wakefulness , Animals , Hippocampus , Long-Term Potentiation , Mice , Sleep
9.
Addict Biol ; 26(2): e12899, 2021 03.
Article in English | MEDLINE | ID: mdl-32255261

ABSTRACT

Although alcohol (i.e., ethanol) is a major drug of abuse, the acute functional effects of ethanol on the reward circuitry are not well defined in vivo. In freely moving rats, we examined the effect of intravenous ethanol administration on neuronal unit activity in the posterior ventral tegmental area (VTA), a central component of the mesolimbic reward system. VTA units were classified as putative dopamine (DA) neurons, fast-firing GABA neurons, and unidentified neurons based on a combination of electrophysiological properties and DA D2 receptor pharmacological responses. A gradual infusion of ethanol significantly altered the firing rate of DA neurons in a concentration-dependent manner. The majority of DA neurons were stimulated by ethanol and showed enhanced burst firing activity, but a minority was inhibited. Ethanol also increased the proportion of DA neurons that exhibited pacemaker-like firing patterns. In contrast, ethanol mediated a variety of effects in GABA and other unidentified neurons that were distinct from DA neurons, including a nonlinear increase in firing rate, delayed inhibition, and more biphasic activity. These results provide evidence of discrete electrophysiological effects of ethanol on DA neurons compared with other VTA cell types, suggesting a complex role of the VTA in alcohol-induced responses in freely moving animals.


Subject(s)
Action Potentials/drug effects , Dopaminergic Neurons/drug effects , Ethanol/pharmacology , GABAergic Neurons/drug effects , Ventral Tegmental Area/drug effects , Animals , Dose-Response Relationship, Drug , Male , Rats , Rats, Long-Evans , Reward
10.
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
11.
eNeuro ; 7(2)2020.
Article in English | MEDLINE | ID: mdl-32102779

ABSTRACT

Nicotine use increases the risk for subsequent abuse of other addictive drugs, but the biological basis underlying this risk remains largely unknown. Interactions between nicotine and other drugs of abuse may arise from nicotine-induced neural adaptations in the mesolimbic dopamine (DA) system, a common pathway for the reinforcing effects of many addictive substances. Previous work identified nicotine-induced neuroadaptations that alter inhibitory transmission in the ventral tegmental area (VTA). Here, we test whether nicotine-induced dysregulation of GABAergic signaling within the VTA increases the vulnerability for benzodiazepine abuse that has been reported in smokers. We demonstrate in rats that nicotine exposure dysregulates diazepam-induced inhibition of VTA GABA neurons and increases diazepam consumption. In VTA GABA neurons, nicotine impaired KCC2-mediated chloride extrusion, depolarized the GABAA reversal potential, and shifted the pharmacological effect of diazepam on GABA neurons from inhibition toward excitation. In parallel, nicotine-related alterations in GABA signaling observed ex vivo were associated with enhanced diazepam-induced inhibition of lateral VTA DA neurons in vivo Targeting KCC2 with the agonist CLP290 normalized diazepam-induced effects on VTA GABA transmission and reduced diazepam consumption following nicotine administration to the control level. Together, our results provide insights into midbrain circuit alterations resulting from nicotine exposure that contribute to the abuse of other drugs, such as benzodiazepines.


Subject(s)
Nicotine , Ventral Tegmental Area , Animals , Diazepam/pharmacology , Dopaminergic Neurons , GABAergic Neurons , Nicotine/pharmacology , Rats
12.
Proc Natl Acad Sci U S A ; 116(52): 27028-27034, 2019 Dec 26.
Article in English | MEDLINE | ID: mdl-31806759

ABSTRACT

Stress is known to alter GABAergic signaling in the ventral tegmental area (VTA), and this inhibitory plasticity is associated with increased alcohol self-administration. In humans, serotonin 2A receptor (5-HT2AR) agonists can treat stress- and alcohol-related disorders, but the neural substrates are ill-defined. Thus, we reasoned that 5-HT2AR pharmacotherapies may ameliorate the stress-induced dysregulated inhibitory VTA circuitry that contributes to subsequent alcohol abuse. We found that acute stress exposure in mice compromised GABA-mediated inhibition of VTA GABA neurons corresponding with increased ethanol-induced GABAergic transmission. This stress-induced inhibitory plasticity was reversible by applying the 5-HT2AR agonist TCB-2 ex vivo via functional enhancement of the potassium-chloride cotransporter KCC2. The signaling pathway linking 5-HT2AR activation and normalization of KCC2 function was dependent on protein kinase C signaling and phosphorylation of KCC2 at serine 940 (S940), as mutation of S940 to alanine prevented restoration of chloride transport function by TCB-2. Through positive modulation of KCC2, TCB-2 also reduced elevated ethanol-induced GABAergic signaling after stress exposure that has previously been linked to increased ethanol consumption. Collectively, these findings provide mechanistic insights into the therapeutic action of 5-HT2AR agonists at the neuronal and circuit levels of brain reward circuitry.

13.
Trends Neurosci ; 41(12): 898-910, 2018 12.
Article in English | MEDLINE | ID: mdl-30149979

ABSTRACT

Behavioral adaptations occur through remodeling of brain circuits, as arising, for instance, from experience-dependent synaptic plasticity. Drugs of abuse and aversive stimuli, such as stress, act on the mesocorticolimbic system, dysregulating adaptive mechanisms and leading to a variety of aberrant behaviors associated with neuropsychiatric disorders. Until recently, research in the field has commonly focused on experience-dependent synaptic plasticity at excitatory synapses. However, there is growing evidence that synaptic plasticity within inhibitory circuits is an important contributor to maladaptive behaviors. We speculate that restoring normal inhibitory synaptic transmission is a promising therapeutic target for correcting some of the circuit abnormalities underlying neuropsychiatric disorders.


Subject(s)
Behavior, Addictive/physiopathology , Brain/physiology , Mental Disorders/physiopathology , Neural Inhibition/physiology , Neuronal Plasticity/physiology , Synaptic Transmission/physiology , Animals , Humans , Neural Pathways/physiology
14.
Cell Rep ; 23(1): 68-77, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29617674

ABSTRACT

Adolescent smoking is associated with pathological drinking later in life, but the biological basis for this vulnerability is unknown. To examine how adolescent nicotine exposure influences subsequent ethanol intake, nicotine was administered during adolescence or adulthood, and responses to alcohol were measured 1 month later. We found that adolescent, but not adult, nicotine exposure altered GABA signaling within the ventral tegmental area (VTA) and led to a long-lasting enhancement of alcohol self-administration. We detected depolarizing shifts in GABAA reversal potentials arising from impaired chloride extrusion in VTA GABA neurons. Alterations in GABA signaling were dependent on glucocorticoid receptor activation and were associated with attenuated dopaminergic neuron responses to alcohol in the lateral VTA. Importantly, enhancing chloride extrusion in adolescent nicotine-treated animals restored VTA GABA signaling and alcohol self-administration to control levels. Taken together, this work suggests that adolescent nicotine exposure increases the risk profile for increased alcohol drinking in adulthood.


Subject(s)
Ethanol/pharmacology , Nicotine/pharmacology , Receptors, GABA-A/metabolism , Synaptic Potentials , Ventral Tegmental Area/drug effects , Animals , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/physiology , Ethanol/administration & dosage , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , GABAergic Neurons/physiology , Male , Rats , Rats, Long-Evans , Receptors, Glucocorticoid/metabolism , Self Administration , Ventral Tegmental Area/growth & development
15.
Annu Rev Pharmacol Toxicol ; 58: 547-566, 2018 01 06.
Article in English | MEDLINE | ID: mdl-28977763

ABSTRACT

Stress and tobacco smoking are risk factors for alcoholism, but the underlying neural mechanisms are not well understood. Although stress, nicotine, and alcohol have broad, individual effects in the brain, some of their actions converge onto the same mechanisms and circuits. Stress and nicotine augment alcohol-related behaviors, in part via modulation of alcohol-evoked neuronal plasticity and metaplasticity mechanisms. Stress modulates alcohol-evoked plasticity via the release of signaling molecules that influence synaptic transmission. Nicotine also activates some of the same signaling molecules, cells, and circuits, producing a convergence of both stress and nicotine onto common plasticity mechanisms that influence alcohol self-administration. We describe several forms of alcohol-induced plasticity, including classic Hebbian plasticity at glutamatergic synapses, and we highlight less appreciated forms, such as non-Hebbian and GABAergic synaptic plasticity. Risk factors such as stress and nicotine initiate lasting neural changes that modify subsequent alcohol-induced synaptic plasticity and increase the vulnerability to alcohol addiction.


Subject(s)
Ethanol/adverse effects , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Nicotine/adverse effects , Stress, Physiological/physiology , Tobacco Smoking/adverse effects , Alcoholism/physiopathology , Animals , Humans , Stress, Physiological/drug effects
16.
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
17.
Eur J Neurosci ; 45(1): 138-146, 2017 01.
Article in English | MEDLINE | ID: mdl-27646422

ABSTRACT

Physiological and behavioral evidence supports that dopamine (DA) receptor signaling influences hippocampal function. While several recent studies examined how DA influences CA1 plasticity and learning, there are fewer studies investigating the influence of DA signaling to the dentate gyrus. The dentate gyrus receives convergent cortical input through the perforant path fiber tracts and has been conceptualized to detect novelty in spatial memory tasks. To test whether DA-receptor activity influences novelty-detection, we used a novel object recognition (NOR) task where mice remember previously presented objects as an indication of learning. Although DA innervation arises from other sources and the main DA signaling may be from those sources, our molecular approaches verified that midbrain dopaminergic fibers also sparsely innervate the dentate gyrus. During the NOR task, wild-type mice spent significantly more time investigating novel objects rather than previously observed objects. Dentate granule cells in slices cut from those mice showed an increased AMPA/NMDA-receptor current ratio indicative of potentiated synaptic transmission. Post-training injection of a D1-like receptor antagonist not only effectively blocked the preference for the novel objects, but also prevented the increased AMPA/NMDA ratio. Consistent with that finding, neither NOR learning nor the increase in the AMPA/NMDA ratio were observed in DA-receptor KO mice under the same experimental conditions. The results indicate that DA-receptor signaling contributes to the successful completion of the NOR task and to the associated synaptic plasticity of the dentate gyrus that likely contributes to the learning.


Subject(s)
Hippocampus/physiology , Neuronal Plasticity/physiology , Receptors, Dopamine/metabolism , Recognition, Psychology/physiology , Synapses/physiology , Synaptic Transmission/physiology , Animals , Dopamine/metabolism , Excitatory Postsynaptic Potentials/genetics , Excitatory Postsynaptic Potentials/physiology , Mice, Knockout , Neuronal Plasticity/genetics , Receptors, Dopamine/genetics , Receptors, N-Methyl-D-Aspartate/metabolism
18.
Elife ; 52016 12 13.
Article in English | MEDLINE | ID: mdl-27960077

ABSTRACT

Recreational drug use leads to compulsive substance abuse in some individuals. Studies on animal models of drug addiction indicate that persistent long-term potentiation (LTP) of excitatory synaptic transmission onto ventral tegmental area (VTA) dopamine (DA) neurons is a critical component of sustained drug seeking. However, little is known about the mechanism regulating such long-lasting changes in synaptic strength. Previously, we identified that translational control by eIF2α phosphorylation (p-eIF2α) regulates cocaine-induced LTP in the VTA (Huang et al., 2016). Here we report that in mice with reduced p-eIF2α-mediated translation, cocaine induces persistent LTP in VTA DA neurons. Moreover, selectively inhibiting eIF2α-mediated translational control with a small molecule ISRIB, or knocking down oligophrenin-1-an mRNA whose translation is controlled by p-eIF2α-in the VTA also prolongs cocaine-induced LTP. This persistent LTP is mediated by the insertion of GluR2-lacking AMPARs. Collectively, our findings suggest that eIF2α-mediated translational control regulates the progression from transient to persistent cocaine-induced LTP.


Subject(s)
Action Potentials/drug effects , Cocaine/metabolism , Dopamine Uptake Inhibitors/metabolism , Dopaminergic Neurons/physiology , Eukaryotic Initiation Factor-2/metabolism , Protein Biosynthesis , Ventral Tegmental Area/physiology , Animals , Dopaminergic Neurons/drug effects , Mice , Ventral Tegmental Area/drug effects
19.
Neuron ; 92(2): 493-504, 2016 Oct 19.
Article in English | MEDLINE | ID: mdl-27720487

ABSTRACT

Stress is a well-known risk factor for subsequent alcohol abuse, but the neural mechanisms underlying interactions between stress and alcohol remain largely unknown. Addictive drug reinforcement and stress signaling involve common neural circuitry, including the mesolimbic dopamine system. We demonstrate in rodents that pre-exposure to stress attenuates alcohol-induced dopamine responses and increases alcohol self-administration. The blunted dopamine signaling resulted from ethanol-induced excitation of GABA neurons in the ventral tegmental area. Excitation of GABA neurons was mediated by GABAA receptor activation and involved stress-induced functional downregulation of the K+, Cl- cotransporter, KCC2. Blocking stress hormone receptors, enhancing KCC2 function, or preventing excitatory GABA signaling by alternative methods all prevented the attenuated alcohol-induced dopamine response and prevented the increased alcohol self-administration. These results demonstrate that stress alters the neural and behavioral responses to alcohol through a neuroendocrine signal that shifts inhibitory GABA transmission toward excitation.


Subject(s)
Alcoholism/metabolism , Central Nervous System Depressants/administration & dosage , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Ethanol/administration & dosage , Self Administration , Stress, Psychological/metabolism , Ventral Tegmental Area/metabolism , gamma-Aminobutyric Acid/metabolism , Acetazolamide/pharmacology , Alcoholism/psychology , Animals , Carbonic Anhydrase Inhibitors/pharmacology , Conditioning, Operant , Hormone Antagonists/pharmacology , Male , Microdialysis , Mifepristone/pharmacology , Patch-Clamp Techniques , Rats , Rats, Long-Evans , Receptors, GABA-A/metabolism , Symporters/drug effects , Symporters/metabolism , K Cl- Cotransporters
20.
Elife ; 52016 Mar 01.
Article in English | MEDLINE | ID: mdl-26928076

ABSTRACT

Adolescents are particularly vulnerable to nicotine, the principal addictive component driving tobacco smoking. In a companion study, we found that reduced activity of the translation initiation factor eIF2α underlies the hypersensitivity of adolescent mice to the effects of cocaine. Here we report that nicotine potentiates excitatory synaptic transmission in ventral tegmental area dopaminergic neurons more readily in adolescent mice compared to adults. Adult mice with genetic or pharmacological reduction in p-eIF2α-mediated translation are more susceptible to nicotine's synaptic effects, like adolescents. When we investigated the influence of allelic variability of the Eif2s1 gene (encoding eIF2α) on reward-related neuronal responses in human smokers, we found that a single nucleotide polymorphism in the Eif2s1 gene modulates mesolimbic neuronal reward responses in human smokers. These findings suggest that p-eIF2α regulates synaptic actions of nicotine in both mice and humans, and that reduced p-eIF2α may enhance susceptibility to nicotine (and other drugs of abuse) during adolescence.


Subject(s)
Dopaminergic Neurons/physiology , Eukaryotic Initiation Factor-2/metabolism , Nicotine/metabolism , Protein Biosynthesis , Protein Processing, Post-Translational , Synapses/drug effects , Ventral Tegmental Area/physiology , Animals , Humans , Mice , Phosphorylation , Smoking , Nicotiana
SELECTION OF CITATIONS
SEARCH DETAIL
...