Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Biol Psychiatry ; 95(8): 774-784, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-37804900

ABSTRACT

BACKGROUND: Social status in humans, generally reflected by socioeconomic status, has been associated, when constrained, with heightened vulnerability to pathologies including psychiatric diseases. Social hierarchy in mice translates into individual and interdependent behavioral strategies of animals within a group. The rules leading to the emergence of a social organization are elusive, and detangling the contribution of social status from other factors, whether environmental or genetic, to normal and pathological behaviors remains challenging. METHODS: We investigated the mechanisms shaping the emergence of a social hierarchy in isogenic C57BL/6 mice raised in groups of 4 using conditional mutant mouse models and chemogenetic manipulation of dopamine midbrain neuronal activity. We further studied the evolution of behavioral traits and the vulnerability to psychopathological-like phenotypes according to the social status of the animals. RESULTS: Higher sociability predetermined higher social hierarchy in the colony. Upon hierarchy establishment, higher-ranked mice showed increased anxiety and better cognitive abilities in a working memory task. Strikingly, the higher-ranked mice displayed a reduced activity of dopaminergic neurons within the ventral tegmental area, paired with a decreased behavioral response to cocaine and a decreased vulnerability to depressive-like behaviors following repeated social defeats. The pharmacogenetic inhibition of this neuronal population and the genetic inactivation of glucocorticoid receptor signaling in dopamine-sensing brain areas that resulted in decreased dopaminergic activity promoted accession to higher social ranks. CONCLUSIONS: Dopamine activity and its modulation by the stress response shapes social organization in mice, potentially linking interindividual and social status differences in vulnerability to psychopathologies.


Subject(s)
Dopaminergic Neurons , Mental Disorders , Humans , Mice , Animals , Dopamine , Hierarchy, Social , Mice, Inbred C57BL , Ventral Tegmental Area
2.
Elife ; 122023 May 30.
Article in English | MEDLINE | ID: mdl-37249215

ABSTRACT

Nicotine intake is likely to result from a balance between the rewarding and aversive properties of the drug, yet the individual differences in neural activity that control aversion to nicotine and their adaptation during the addiction process remain largely unknown. Using a two-bottle choice experiment, we observed considerable heterogeneity in nicotine-drinking profiles in isogenic adult male mice, with about half of the mice persisting in nicotine consumption even at high concentrations, whereas the other half stopped consuming. We found that nicotine intake was negatively correlated with nicotine-evoked currents in the interpeduncular nucleus (IPN), and that prolonged exposure to nicotine, by weakening this response, decreased aversion to the drug, and hence boosted consumption. Lastly, using knock-out mice and local gene re-expression, we identified ß4-containing nicotinic acetylcholine receptors of IPN neurons as molecular and cellular correlates of nicotine aversion. Collectively, our results identify the IPN as a substrate for individual variabilities and adaptations in nicotine consumption.


Subject(s)
Habenula , Interpeduncular Nucleus , Receptors, Nicotinic , Mice , Male , Animals , Nicotine/pharmacology , Interpeduncular Nucleus/metabolism , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Mice, Knockout , Neurons/metabolism , Habenula/metabolism
3.
Neuropsychopharmacology ; 48(6): 963-974, 2023 05.
Article in English | MEDLINE | ID: mdl-36932179

ABSTRACT

A link between gut dysbiosis and the pathogenesis of brain disorders has been identified. A role for gut bacteria in drug reward and addiction has been suggested but very few studies have investigated their impact on brain and behavioral responses to addictive drugs so far. In particular, their influence on nicotine's addiction-like processes remains unknown. In addition, evidence shows that glial cells shape the neuronal activity of the mesolimbic system but their regulation, within this system, by the gut microbiome is not established. We demonstrate that a lack of gut microbiota in male mice potentiates the nicotine-induced activation of sub-regions of the mesolimbic system. We further show that gut microbiota depletion enhances the response to nicotine of dopaminergic neurons of the posterior ventral tegmental area (pVTA), and alters nicotine's rewarding and aversive effects in an intra-VTA self-administration procedure. These effects were not associated with gross behavioral alterations and the nicotine withdrawal syndrome was not impacted. We further show that depletion of the gut microbiome modulates the glial cells of the mesolimbic system. Notably, it increases the number of astrocytes selectively in the pVTA, and the expression of postsynaptic density protein 95 in both VTA sub-regions, without altering the density of the astrocytic glutamatergic transporter GLT1. Finally, we identify several sub-populations of microglia in the VTA that differ between its anterior and posterior sub-parts, and show that they are re-organized in conditions of gut microbiota depletion. The present study paves the way for refining our understanding of the pathophysiology of nicotine addiction.


Subject(s)
Gastrointestinal Microbiome , Substance Withdrawal Syndrome , Mice , Male , Animals , Nicotine/pharmacology , Ventral Tegmental Area , Dopamine/metabolism , Reward , Substance Withdrawal Syndrome/metabolism , Neuroglia/metabolism
4.
Nat Commun ; 13(1): 1807, 2022 04 04.
Article in English | MEDLINE | ID: mdl-35379786

ABSTRACT

Enduring behavioral changes upon stress exposure involve changes in gene expression sustained by epigenetic modifications in brain circuits, including the mesocorticolimbic pathway. Brahma (BRM) and Brahma Related Gene 1 (BRG1) are ATPase subunits of the SWI/SNF complexes involved in chromatin remodeling, a process essential to enduring plastic changes in gene expression. Here, we show that in mice, social defeat induces changes in BRG1 nuclear distribution. The inactivation of the Brg1/Smarca4 gene within dopamine-innervated regions or the constitutive inactivation of the Brm/Smarca2 gene leads to resilience to repeated social defeat and decreases the behavioral responses to cocaine without impacting midbrain dopamine neurons activity. Within striatal medium spiny neurons, Brg1 gene inactivation reduces the expression of stress- and cocaine-induced immediate early genes, increases levels of heterochromatin and at a global scale decreases chromatin accessibility. Altogether these data demonstrate the pivotal function of SWI/SNF complexes in behavioral and transcriptional adaptations to salient environmental challenges.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin , Adenosine Triphosphatases/metabolism , Animals , Cell Line, Tumor , Mice , Reward
5.
Nat Commun ; 12(1): 6945, 2021 11 26.
Article in English | MEDLINE | ID: mdl-34836948

ABSTRACT

Long-term exposure to nicotine alters brain circuits and induces profound changes in decision-making strategies, affecting behaviors both related and unrelated to drug seeking and consumption. Using an intracranial self-stimulation reward-based foraging task, we investigated in mice the impact of chronic nicotine on midbrain dopamine neuron activity and its consequence on the trade-off between exploitation and exploration. Model-based and archetypal analysis revealed substantial inter-individual variability in decision-making strategies, with mice passively exposed to nicotine shifting toward a more exploitative profile compared to non-exposed animals. We then mimicked the effect of chronic nicotine on the tonic activity of dopamine neurons using optogenetics, and found that photo-stimulated mice adopted a behavioral phenotype similar to that of mice exposed to chronic nicotine. Our results reveal a key role of tonic midbrain dopamine in the exploration/exploitation trade-off and highlight a potential mechanism by which nicotine affects the exploration/exploitation balance and decision-making.


Subject(s)
Exploratory Behavior/drug effects , Mesencephalon/drug effects , Nicotine/adverse effects , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Exploratory Behavior/physiology , Male , Mesencephalon/cytology , Mesencephalon/metabolism , Mice , Models, Animal , Nicotine/administration & dosage , Optogenetics , Prejudice , Reward , Self Administration , Stereotaxic Techniques
6.
Neuron ; 109(16): 2604-2615.e9, 2021 08 18.
Article in English | MEDLINE | ID: mdl-34242565

ABSTRACT

Nicotine stimulates dopamine (DA) neurons of the ventral tegmental area (VTA) to establish and maintain reinforcement. Nicotine also induces anxiety through an as yet unknown circuitry. We found that nicotine injection drives opposite functional responses of two distinct populations of VTA DA neurons with anatomically segregated projections: it activates neurons that project to the nucleus accumbens (NAc), whereas it inhibits neurons that project to the amygdala nuclei (Amg). We further show that nicotine mediates anxiety-like behavior by acting on ß2-subunit-containing nicotinic acetylcholine receptors of the VTA. Finally, using optogenetics, we bidirectionally manipulate the VTA-NAc and VTA-Amg pathways to dissociate their contributions to anxiety-like behavior. We show that inhibition of VTA-Amg DA neurons mediates anxiety-like behavior, while their activation prevents the anxiogenic effects of nicotine. These distinct subpopulations of VTA DA neurons with opposite responses to nicotine may differentially drive the anxiogenic and the reinforcing effects of nicotine.


Subject(s)
Anxiety/drug therapy , Neural Pathways/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Ventral Tegmental Area/drug effects , Amygdala/drug effects , Amygdala/metabolism , Animals , Anxiety/chemically induced , Anxiety/physiopathology , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/physiology , Male , Mice , Neural Pathways/physiology , Nicotine/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiology , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/metabolism , Reinforcement, Psychology , Ventral Tegmental Area/physiology
7.
Cell Rep ; 34(3): 108654, 2021 01 19.
Article in English | MEDLINE | ID: mdl-33472083

ABSTRACT

In humans, execution of unimanual movements requires lateralized activation of the primary motor cortex, which then transmits the motor command to the contralateral hand through the crossed corticospinal tract (CST). Mutations in NTN1 alter motor control lateralization, leading to congenital mirror movements. To address the role of midline Netrin-1 on CST development and subsequent motor control, we analyze the morphological and functional consequences of floor plate Netrin-1 depletion in conditional knockout mice. We show that depletion of floor plate Netrin-1 in the brainstem critically disrupts CST midline crossing, whereas the other commissural systems are preserved. The only associated defect is an abnormal entry of CST axons within the inferior olive. Alteration of CST midline crossing results in functional ipsilateral projections and is associated with abnormal symmetric movements. Our study reveals the role of Netrin-1 in CST development and describes a mouse model recapitulating the characteristics of human congenital mirror movements.


Subject(s)
Axons/metabolism , Movement Disorders/metabolism , Netrin-1/metabolism , Pyramidal Tracts/metabolism , Animals , Axons/pathology , Mice , Movement Disorders/pathology , Pyramidal Tracts/pathology
8.
Cell Metab ; 31(4): 773-790.e11, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32142669

ABSTRACT

Energy-dense food alters dopaminergic (DA) transmission in the mesocorticolimbic (MCL) system and can promote reward dysfunctions, compulsive feeding, and weight gain. Yet the mechanisms by which nutrients influence the MCL circuitry remain elusive. Here, we show that nutritional triglycerides (TGs), a conserved post-prandial metabolic signature among mammals, can be metabolized within the MCL system and modulate DA-associated behaviors by gating the activity of dopamine receptor subtype 2 (DRD2)-expressing neurons through a mechanism that involves the action of the lipoprotein lipase (LPL). Further, we show that in humans, post-prandial TG excursions modulate brain responses to food cues in individuals carrying a genetic risk for reduced DRD2 signaling. Collectively, these findings unveil a novel mechanism by which dietary TGs directly alter signaling in the reward circuit to regulate behavior, thereby providing a new mechanistic basis by which energy-rich diets may lead to (mal)adaptations in DA signaling that underlie reward deficit and compulsive behavior.


Subject(s)
Motivation , Neurons , Receptors, Dopamine D2/metabolism , Triglycerides/metabolism , Adolescent , Adult , Animals , Female , Humans , Male , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/metabolism , Young Adult
9.
Nat Commun ; 9(1): 4449, 2018 10 25.
Article in English | MEDLINE | ID: mdl-30361503

ABSTRACT

Stressful life events are primary environmental factors that markedly contribute to depression by triggering brain cellular maladaptations. Dysregulation of ventral tegmental area (VTA) dopamine neurons has been causally linked to the appearance of social withdrawal and anhedonia, two classical manifestations of depression. However, the relevant inputs that shape these dopamine signals remain largely unknown. We demonstrate that chronic social defeat (CSD) stress, a preclinical paradigm of depression, causes marked hyperactivity of laterodorsal tegmentum (LDTg) excitatory neurons that project to the VTA. Selective chemogenetic-mediated inhibition of cholinergic LDTg neurons prevent CSD-induced VTA DA neurons dysregulation and depressive-like behaviors. Pro-depressant outcomes are replicated by pairing activation of LDTg cholinergic terminals in the VTA with a moderate stress. Prevention of CSD outcomes are recapitulated by blocking corticotropin-releasing factor receptor 1 within the LDTg. These data uncover a neuro-circuitry of depressive-like disorders and demonstrate that stress, via a neuroendocrine signal, profoundly dysregulates the LDTg.


Subject(s)
Acetylcholine/metabolism , Behavior, Animal , Depression/psychology , Dopaminergic Neurons/pathology , Mesencephalon/pathology , Pons/pathology , Stress, Psychological/complications , Animals , Chronic Disease , Corticotropin-Releasing Hormone/metabolism , Depression/pathology , Dopaminergic Neurons/metabolism , Gene Silencing , Glutamic Acid/metabolism , Male , Mice, Inbred C57BL , Models, Biological , Pontine Tegmentum/pathology , Receptors, Corticotropin-Releasing Hormone/metabolism , Signal Transduction , Ventral Tegmental Area/pathology
10.
Elife ; 72018 09 04.
Article in English | MEDLINE | ID: mdl-30176987

ABSTRACT

Dopamine (DA) neurons of the ventral tegmental area (VTA) integrate cholinergic inputs to regulate key functions such as motivation and goal-directed behaviors. Yet the temporal dynamic range and mechanism of action of acetylcholine (ACh) on the modulation of VTA circuits and reward-related behaviors are not known. Here, we used a chemical-genetic approach for rapid and precise optical manipulation of nicotinic neurotransmission in VTA neurons in living mice. We provide direct evidence that the ACh tone fine-tunes the firing properties of VTA DA neurons through ß2-containing (ß2*) nicotinic ACh receptors (nAChRs). Furthermore, locally photo-antagonizing these receptors in the VTA was sufficient to reversibly switch nicotine reinforcement on and off. By enabling control of nicotinic transmission in targeted brain circuits, this technology will help unravel the various physiological functions of nAChRs and may assist in the design of novel therapies relevant to neuropsychiatric disorders.


Subject(s)
Dopaminergic Neurons/metabolism , Light , Mesencephalon/cytology , Receptors, Nicotinic/metabolism , Reward , Action Potentials/drug effects , Action Potentials/physiology , Action Potentials/radiation effects , Animals , Cell Line , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/radiation effects , Mice, Inbred C57BL , Nicotine/pharmacology , Signal Transduction/radiation effects , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism , Ventral Tegmental Area/radiation effects
11.
Nat Med ; 24(5): 591-597, 2018 05.
Article in English | MEDLINE | ID: mdl-29736027

ABSTRACT

Depression, a devastating psychiatric disorder, is a leading cause of disability worldwide. Current antidepressants address specific symptoms of the disease, but there is vast room for improvement 1 . In this respect, new compounds that act beyond classical antidepressants to target signal transduction pathways governing synaptic plasticity and cellular resilience are highly warranted2-4. The extracellular signal-regulated kinase (ERK) pathway is implicated in mood regulation5-7, but its pleiotropic functions and lack of target specificity prohibit optimal drug development. Here, we identified the transcription factor ELK-1, an ERK downstream partner 8 , as a specific signaling module in the pathophysiology and treatment of depression that can be targeted independently of ERK. ELK1 mRNA was upregulated in postmortem hippocampal tissues from depressed suicides; in blood samples from depressed individuals, failure to reduce ELK1 expression was associated with resistance to treatment. In mice, hippocampal ELK-1 overexpression per se produced depressive behaviors; conversely, the selective inhibition of ELK-1 activation prevented depression-like molecular, plasticity and behavioral states induced by stress. Our work stresses the importance of target selectivity for a successful approach for signal-transduction-based antidepressants, singles out ELK-1 as a depression-relevant transducer downstream of ERK and brings proof-of-concept evidence for the druggability of ELK-1.


Subject(s)
Antidepressive Agents/pharmacology , Signal Transduction/drug effects , ets-Domain Protein Elk-1/metabolism , Adult , Animals , Behavior, Animal , Depression/blood , Depression/genetics , Depression/physiopathology , Female , Hippocampus/metabolism , Humans , Male , Mice , Middle Aged , Neuronal Plasticity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stress, Psychological/complications , ets-Domain Protein Elk-1/blood , ets-Domain Protein Elk-1/genetics
12.
Sci Rep ; 7: 45116, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28332590

ABSTRACT

Alcohol and nicotine are the most widely co-abused drugs. Both modify the activity of dopaminergic (DA) neurons of the Ventral Tegmental Area (VTA) and lead to an increase in DA release in the Nucleus Accumbens, thereby affecting the reward system. Evidences support the hypothesis that distinct nicotinic acetylcholine receptors (nAChRs), the molecular target of acetylcholine (ACh) and exogenous nicotine, are also in addition implicated in the response to alcohol. The precise molecular and neuronal substrates of this interaction are however not well understood. Here we used in vivo electrophysiology in the VTA to characterise acute and chronic interactions between nicotine and alcohol. Simultaneous injections of the two drugs enhanced their responses on VTA DA neuron firing and chronic exposure to nicotine increased alcohol-induced DA responses and alcohol intake. Then, we assessed the role of ß4 * nAChRs, but not ß2 * nAChRs, in mediating acute responses to alcohol using nAChR subtypes knockout mice (ß2-/- and ß4-/- mice). Finally, we showed that nicotine-induced modifications of alcohol responses were absent in ß2-/- and ß4-/- mice, suggesting that nicotine triggers ß2* and ß4 * nAChR-dependent neuroadaptations that subsequently modify the responses to alcohol and thus indicating these receptors as key mediators in the complex interactions between these two drugs.


Subject(s)
Alcohols/pharmacology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Nerve Tissue Proteins/metabolism , Nicotine/pharmacology , Receptors, Nicotinic/metabolism , Action Potentials/drug effects , Animals , Mice , Reward
13.
Nat Neurosci ; 19(4): 560-3, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26878672

ABSTRACT

Dopamine (DA) neurons in the ventral tegmental area (VTA) help mediate stress susceptibility and resilience. However, upstream mechanisms controlling these neurons remain unknown. Noradrenergic (NE) neurons in the locus coeruleus, implicated in the pathophysiology of depression, have direct connections within the VTA. Here we demonstrate that NE neurons regulate vulnerability to social defeat through inhibitory control of VTA DA neurons.


Subject(s)
Adrenergic Neurons/physiology , Dopaminergic Neurons/metabolism , Neural Inhibition/physiology , Resilience, Psychological , Stress, Psychological/metabolism , Animals , Chronic Disease , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Norepinephrine/physiology , Stress, Psychological/genetics , Stress, Psychological/psychology , Ventral Tegmental Area/metabolism , Vesicular Monoamine Transport Proteins/deficiency , Vesicular Monoamine Transport Proteins/genetics
14.
Nat Neurosci ; 19(3): 471-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26780509

ABSTRACT

Cholinergic neurotransmission affects decision-making, notably through the modulation of perceptual processing in the cortex. In addition, acetylcholine acts on value-based decisions through as yet unknown mechanisms. We found that nicotinic acetylcholine receptors (nAChRs) expressed in the ventral tegmental area (VTA) are involved in the translation of expected uncertainty into motivational value. We developed a multi-armed bandit task for mice with three locations, each associated with a different reward probability. We found that mice lacking the nAChR ß2 subunit showed less uncertainty-seeking than their wild-type counterparts. Using model-based analysis, we found that reward uncertainty motivated wild-type mice, but not mice lacking the nAChR ß2 subunit. Selective re-expression of the ß2 subunit in the VTA was sufficient to restore spontaneous bursting activity in dopamine neurons and uncertainty-seeking. Our results reveal an unanticipated role for subcortical nAChRs in motivation induced by expected uncertainty and provide a parsimonious account for a wealth of behaviors related to nAChRs in the VTA expressing the ß2 subunit.


Subject(s)
Motivation/physiology , Receptors, Nicotinic/physiology , Uncertainty , Ventral Tegmental Area/physiology , Animals , Dopaminergic Neurons/physiology , Mice, Knockout , Mice, Transgenic , Receptors, Nicotinic/genetics , Reward , Self Stimulation/physiology
15.
Sci Rep ; 5: 8184, 2015 Feb 02.
Article in English | MEDLINE | ID: mdl-25640814

ABSTRACT

Midbrain dopamine (DA) neurons are key players in motivation and reward processing. Increased DA release is thought to be central in the initiation of drug addiction. Whereas dopamine neurons are generally considered to be activated by drugs such as nicotine, we report here that nicotine not only induces excitation of ventral tegmental area (VTA) DA cells but also induces inhibition of a subset of VTA DA neurons that are anatomically segregated in the medial part of the VTA. These opposite responses do not correlate with the inhibition and excitation induced by noxious stimuli. We show that this inhibition requires D2 receptor (D2-R) activation, suggesting that a dopaminergic release is involved in the mechanism. Our findings suggest a principle of concurrent excitation and inhibition of VTA DA cells in response to nicotine. It promotes unexplored roles for DA release in addiction contrasting with the classical views of reinforcement and motivation, and give rise to a new interpretation of the mode of operation of the reward system.


Subject(s)
Dopaminergic Neurons/drug effects , Nicotine/toxicity , Receptors, Dopamine D2/metabolism , Ventral Tegmental Area/drug effects , Action Potentials/drug effects , Animals , Dopaminergic Neurons/metabolism , Mice , Mice, Inbred C57BL , Receptors, Dopamine D2/chemistry , Ventral Tegmental Area/metabolism
16.
Science ; 339(6117): 332-5, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23329050

ABSTRACT

Repeated traumatic events induce long-lasting behavioral changes that are key to organism adaptation and that affect cognitive, emotional, and social behaviors. Rodents subjected to repeated instances of aggression develop enduring social aversion and increased anxiety. Such repeated aggressions trigger a stress response, resulting in glucocorticoid release and activation of the ascending dopamine (DA) system. We bred mice with selective inactivation of the gene encoding the glucocorticoid receptor (GR) along the DA pathway, and exposed them to repeated aggressions. GR in dopaminoceptive but not DA-releasing neurons specifically promoted social aversion as well as dopaminergic neurochemical and electrophysiological neuroadaptations. Anxiety and fear memories remained unaffected. Acute inhibition of the activity of DA-releasing neurons fully restored social interaction in socially defeated wild-type mice. Our data suggest a GR-dependent neuronal dichotomy for the regulation of emotional and social behaviors, and clearly implicate GR as a link between stress resiliency and dopaminergic tone.


Subject(s)
Anxiety/metabolism , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Receptors, Glucocorticoid/metabolism , Social Alienation , Social Isolation , Stress, Psychological/metabolism , Animals , Fear , Mice , Mice, Mutant Strains , Receptors, Dopamine/metabolism , Receptors, Glucocorticoid/genetics
17.
Neuropsychopharmacology ; 36(11): 2244-57, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21716264

ABSTRACT

Nicotine prominently mediates the behavioral effects of tobacco consumption, either through smoking or when taking tobacco by snuff or chew. However, many studies question the exclusive role of nicotine in these effects. The use of preparations containing all the components of tobacco, such as tobacco and smoke extracts, may be more suitable than nicotine alone to investigate the behavioral effects of smoking and tobacco intake. In the present study, the electrophysiological effects of tobacco and smoke on ventral tegmental area dopaminergic (DA) neurons were examined in vivo in anesthetized wild-type (WT), ß2-nicotinic acetylcholine receptor (nAChR) knockout (ß2-/-), α4-/-, and α6-/- mice and compared with those of nicotine alone. In WT mice, smoke and nicotine had similar potentiating effects on DA cell activity, but the action of tobacco on neuronal firing was weak and often inhibitory. In particular, nicotine triggered strong bursting activity, whereas no bursting activity was observed after tobacco extract (ToE) administration. In ß2-/- mice, nicotine or extract elicited no modification of the firing patterns of DA cells, indicating that extract acts predominantly through nAChRs. The differences between DA cell activation profiles induced by tobacco and nicotine alone observed in WT persisted in α6-/- mice but not in α4-/- mice. These results would suggest that tobacco has lower addiction-generating properties compared with either nicotine alone or smoke. The weak activation and prominent inhibition obtained with ToEs suggest that tobacco contains compounds that counteract some of the activating effects of nicotine and promote inhibition on DA cell acting through α4ß2*-nAChRs. The nature of these compounds remains to be elucidated. It nevertheless confirms that nicotine is the main substance involved in the tobacco addiction-related activation of mesolimbic DA neurons.


Subject(s)
Action Potentials/drug effects , Dopaminergic Neurons/drug effects , Nicotiana , Nicotine/pharmacology , Plant Extracts/pharmacology , Smoke , Ventral Tegmental Area/drug effects , Action Potentials/physiology , Animals , Dopaminergic Neurons/physiology , Drug Synergism , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nicotine/metabolism , Plant Extracts/isolation & purification , Ventral Tegmental Area/physiology
18.
Proc Natl Acad Sci U S A ; 108(18): 7577-82, 2011 May 03.
Article in English | MEDLINE | ID: mdl-21502501

ABSTRACT

Nicotine is the primary psychoactive component of tobacco. Its reinforcing and addictive properties depend on nicotinic acetylcholine receptors (nAChRs) located within the mesolimbic axis originating in the ventral tegmental area (VTA). The roles and oligomeric assembly of subunit α4- and subunit α6-containing nAChRs in dopaminergic (DAergic) neurons are much debated. Using subunit-specific knockout mice and targeted lentiviral re-expression, we have determined the subunit dependence of intracranial nicotine self-administration (ICSA) into the VTA and the effects of nicotine on dopamine (DA) neuron excitability in the VTA and on DA transmission in the nucleus accumbens (NAc). We show that the α4 subunit, but not the α6 subunit, is necessary for ICSA and nicotine-induced bursting of VTA DAergic neurons, whereas subunits α4 and α6 together regulate the activity dependence of DA transmission in the NAc. These data suggest that α4-dominated enhancement of burst firing in DA neurons, relayed by DA transmission in NAc that is gated by nAChRs containing α4 and α6 subunits, underlies nicotine self-administration and its long-term maintenance.


Subject(s)
Neurons/metabolism , Nicotine/metabolism , Receptors, Nicotinic/metabolism , Ventral Tegmental Area/metabolism , Analysis of Variance , Animals , Autoradiography , Dopamine/metabolism , Electrophysiology , Genetic Vectors/genetics , Lentivirus , Mice , Mice, Inbred C57BL , Mice, Knockout , Nicotine/pharmacology , Receptors, Nicotinic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL