Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38187775

ABSTRACT

Determining the localization of intracerebral implants in rodent brain stands as a critical final step in most physiological and behaviroral studies, especially when targeting deep brain nuclei. Conventional histological approaches, reliant on manual estimation through sectioning and slice examination, are error-prone, potentially complicating data interpretation. Leveraging recent advances in tissue-clearing techniques and light-sheet fluorescence microscopy, we introduce a method enabling virtual brain slicing in any orientation, offering precise implant localization without the limitations of traditional tissue sectioning. To illustrate the method's utility, we present findings from the implantation of linear silicon probes into the midbrain interpeduncular nucleus (IPN) of anesthetized transgenic mice expressing chanelrhodopsin-2 and enhanced yellow fluorescent protein under the choline acetyltransferase (ChAT) promoter/enhancer regions (ChAT-Chr2-EYFP mice). Utilizing a fluorescent dye applied to the electrode surface, we visualized both the targeted area and the precise localization, enabling enhanced inter-subject comparisons. Three dimensional (3D) brain renderings, presented effortlessly in video format across various orientations, showcase the versatility of this approach.

2.
Pharmacol Res ; 190: 106734, 2023 04.
Article in English | MEDLINE | ID: mdl-36933754

ABSTRACT

Over the last decade, the understanding of the habenula has rapidly advanced from being an understudied brain area with the Latin name 'habena" meaning "little rein", to being considered a "major rein" in the control of key monoaminergic brain centers. This ancient brain structure is a strategic node in the information flow from fronto-limbic brain areas to brainstem nuclei. As such, it plays a crucial role in regulating emotional, motivational, and cognitive behaviors and has been implicated in several neuropsychiatric disorders, including depression and addiction. This review will summarize recent findings on the medial (MHb) and lateral (LHb) habenula, their topographical projections, cell types, and functions. Additionally, we will discuss contemporary efforts that have uncovered novel molecular pathways and synaptic mechanisms with a focus on MHb-Interpeduncular nucleus (IPN) synapses. Finally, we will explore the potential interplay between the habenula's cholinergic and non-cholinergic components in coordinating related emotional and motivational behaviors, raising the possibility that these two pathways work together to provide balanced roles in reward prediction and aversion, rather than functioning independently.


Subject(s)
Habenula , Interpeduncular Nucleus , Motivation , Habenula/metabolism , Interpeduncular Nucleus/metabolism , Emotions
3.
Proc Natl Acad Sci U S A ; 119(46): e2209870119, 2022 Nov 15.
Article in English | MEDLINE | ID: mdl-36346845

ABSTRACT

Hedgehog-interacting protein (HHIP) sequesters Hedgehog ligands to repress Smoothened (SMO)-mediated recruitment of the GLI family of transcription factors. Allelic variation in HHIP confers risk of chronic obstructive pulmonary disease and other smoking-related lung diseases, but underlying mechanisms are unclear. Using single-cell and cell-type-specific translational profiling, we show that HHIP expression is highly enriched in medial habenula (MHb) neurons, particularly MHb cholinergic neurons that regulate aversive behavioral responses to nicotine. HHIP deficiency dysregulated the expression of genes involved in cholinergic signaling in the MHb and disrupted the function of nicotinic acetylcholine receptors (nAChRs) through a PTCH-1/cholesterol-dependent mechanism. Further, CRISPR/Cas9-mediated genomic cleavage of the Hhip gene in MHb neurons enhanced the motivational properties of nicotine in mice. These findings suggest that HHIP influences vulnerability to smoking-related lung diseases in part by regulating the actions of nicotine on habenular aversion circuits.


Subject(s)
Habenula , Lung Diseases , Receptors, Nicotinic , Mice , Animals , Nicotine/pharmacology , Nicotine/metabolism , Habenula/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Receptors, Nicotinic/metabolism , Cholinergic Neurons/metabolism , Lung Diseases/metabolism
4.
Proc Natl Acad Sci U S A ; 119(49): e2211454119, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36442105

ABSTRACT

Neuromodulatory substances can be released from distal afferents for communication between brain structures or produced locally to modulate neighboring circuit elements. Corticotropin-releasing hormone (CRH) from long-range neurons in the hypothalamus projecting to the medial prefrontal cortex (mPFC) has been shown to induce anxiety-like behaviors. However, the role of CRH produced in the mPFC has not been investigated. Here we demonstrate that a specific class of mPFC interneurons that express CRH (CrhINs) releases CRH upon high-frequency stimulation to enhance excitability of layer 2/3 pyramidal cells (L2/3 PCs) expressing the CRH receptors. When stimulated at low frequency, CrhINs release GABA resulting in the inhibition of oxytocin receptor-expressing interneurons (OxtrINs) and L2/3 PCs. Conditional deletion of CRH in mPFC CrhINs and chemogenetic activation of CrhINs have opposite effects on novelty exploration in male but not in female mice, and do not affect anxiety-related behaviors in either males or females. Our data reveal that CRH produced by local interneurons in the mPFC is required for sex-specific novelty exploration and suggest that our understanding of complex behaviors may require knowledge of local and remote neuromodulatory action.


Subject(s)
Corticotropin-Releasing Hormone , Prefrontal Cortex , Female , Male , Animals , Mice , Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone , Pyramidal Cells , Interneurons
5.
J Neurosci ; 40(17): 3465-3477, 2020 04 22.
Article in English | MEDLINE | ID: mdl-32184221

ABSTRACT

Nicotine addiction, through smoking, is the principal cause of preventable mortality worldwide. Human genome-wide association studies have linked polymorphisms in the CHRNA5-CHRNA3-CHRNB4 gene cluster, coding for the α5, α3, and ß4 nicotinic acetylcholine receptor (nAChR) subunits, to nicotine addiction. ß4*nAChRs have been implicated in nicotine withdrawal, aversion, and reinforcement. Here we show that ß4*nAChRs also are involved in non-nicotine-mediated responses that may predispose to addiction-related behaviors. ß4 knock-out (KO) male mice show increased novelty-induced locomotor activity, lower baseline anxiety, and motivational deficits in operant conditioning for palatable food rewards and in reward-based Go/No-go tasks. To further explore reward deficits we used intracranial self-administration (ICSA) by directly injecting nicotine into the ventral tegmental area (VTA) in mice. We found that, at low nicotine doses, ß4KO self-administer less than wild-type (WT) mice. Conversely, at high nicotine doses, this was reversed and ß4KO self-administered more than WT mice, whereas ß4-overexpressing mice avoided nicotine injections. Viral expression of ß4 subunits in medial habenula (MHb), interpeduncular nucleus (IPN), and VTA of ß4KO mice revealed dose- and region-dependent differences: ß4*nAChRs in the VTA potentiated nicotine-mediated rewarding effects at all doses, whereas ß4*nAChRs in the MHb-IPN pathway, limited VTA-ICSA at high nicotine doses. Together, our findings indicate that the lack of functional ß4*nAChRs result in deficits in reward sensitivity including increased ICSA at high doses of nicotine that is restored by re-expression of ß4*nAChRs in the MHb-IPN. These data indicate that ß4 is a critical modulator of reward-related behaviors.SIGNIFICANCE STATEMENT Human genetic studies have provided strong evidence for a relationship between variants in the CHRNA5-CHRNA3-CHRNB4 gene cluster and nicotine addiction. Yet, little is known about the role of ß4 nicotinic acetylcholine receptor (nAChR) subunit encoded by this cluster. We investigated the implication of ß4*nAChRs in anxiety-, food reward- and nicotine reward-related behaviors. Deletion of the ß4 subunit gene resulted in an addiction-related phenotype characterized by low anxiety, high novelty-induced response, lack of sensitivity to palatable food rewards and increased intracranial nicotine self-administration at high doses. Lentiviral vector-induced re-expression of the ß4 subunit into either the MHb or IPN restored a "stop" signal on nicotine self-administration. These results suggest that ß4*nAChRs provide a promising novel drug target for smoking cessation.


Subject(s)
Conditioning, Operant/drug effects , Motor Activity/drug effects , Nerve Tissue Proteins/metabolism , Nicotine/administration & dosage , Receptors, Nicotinic/metabolism , Reward , Self-Control , Ventral Tegmental Area/drug effects , Animals , Behavior, Animal/drug effects , Discrimination Learning/drug effects , Male , Mice , Mice, Knockout , Motivation/drug effects , Nerve Tissue Proteins/genetics , Nicotinic Agonists/administration & dosage , Receptors, Nicotinic/genetics , Self Administration
6.
Proc Natl Acad Sci U S A ; 117(10): 5502-5509, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32098843

ABSTRACT

The habenula, an ancient small brain area in the epithalamus, densely expresses nicotinic acetylcholine receptors and is critical for nicotine intake and aversion. As such, identification of strategies to manipulate habenular activity may yield approaches to treat nicotine addiction. Here we show that GPR151, an orphan G-protein-coupled receptor (GPCR) highly enriched in the habenula of humans and rodents, is expressed at presynaptic membranes and synaptic vesicles and associates with synaptic components controlling vesicle release and ion transport. Deletion of Gpr151 inhibits evoked neurotransmission but enhances spontaneous miniature synaptic currents and eliminates short-term plasticity induced by nicotine. We find that GPR151 couples to the G-alpha inhibitory protein Gαo1 to reduce cyclic adenosine monophosphate (cAMP) levels in mice and in GPR151-expressing cell lines that are amenable to ligand screens. Gpr151- knockout (KO) mice show diminished behavioral responses to nicotine and self-administer greater quantities of the drug, phenotypes rescued by viral reexpression of Gpr151 in the habenula. These data identify GPR151 as a critical modulator of habenular function that controls nicotine addiction vulnerability.


Subject(s)
Habenula/physiology , Neuronal Plasticity/physiology , Nicotine/metabolism , Nicotinic Agonists/metabolism , Receptors, G-Protein-Coupled/physiology , Substance-Related Disorders/metabolism , Animals , CHO Cells , Cricetulus , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Habenula/metabolism , Humans , Mice, Knockout , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Receptors, G-Protein-Coupled/genetics , Synaptic Transmission/genetics , Synaptic Transmission/physiology
7.
Nature ; 574(7778): 372-377, 2019 10.
Article in English | MEDLINE | ID: mdl-31619789

ABSTRACT

Diabetes is far more prevalent in smokers than non-smokers, but the underlying mechanisms of vulnerability are unknown. Here we show that the diabetes-associated gene Tcf7l2 is densely expressed in the medial habenula (mHb) region of the rodent brain, where it regulates the function of nicotinic acetylcholine receptors. Inhibition of TCF7L2 signalling in the mHb increases nicotine intake in mice and rats. Nicotine increases levels of blood glucose by TCF7L2-dependent stimulation of the mHb. Virus-tracing experiments identify a polysynaptic connection from the mHb to the pancreas, and wild-type rats with a history of nicotine consumption show increased circulating levels of glucagon and insulin, and diabetes-like dysregulation of blood glucose homeostasis. By contrast, mutant Tcf7l2 rats are resistant to these actions of nicotine. Our findings suggest that TCF7L2 regulates the stimulatory actions of nicotine on a habenula-pancreas axis that links the addictive properties of nicotine to its diabetes-promoting actions.


Subject(s)
Glucose Metabolism Disorders/genetics , Habenula/metabolism , Signal Transduction , Tobacco Use Disorder/complications , Transcription Factor 7-Like 2 Protein/metabolism , Animals , Cyclic AMP/metabolism , Glucose/metabolism , Glucose Metabolism Disorders/metabolism , Humans , Mice , Mutagenesis , Nicotine/metabolism , PC12 Cells , Pancreas/metabolism , Rats , Receptors, Nicotinic/metabolism , Tobacco Use Disorder/genetics , Tobacco Use Disorder/metabolism , Transcription Factor 7-Like 2 Protein/genetics
8.
J Neurosci ; 38(19): 4490-4504, 2018 05 09.
Article in English | MEDLINE | ID: mdl-29650697

ABSTRACT

Behavioral flexibility and impulse control are necessary for successful execution of adaptive behavior. They are impaired in patients with damage to the prefrontal cortex (PFC) and in some clinically important conditions, such as obsessive-compulsive disorder. Although the medial prefrontal cortex (mPFC) has been investigated as a critical structure for behavioral flexibility and impulse control, the contribution of the underlying pyramidal neuron cell types in the mPFC remained to be understood. Here we show that interneuron-mediated local inactivation of pyramidal neurons in the mPFC of male and female mice induces both premature responses and choice bias, and establish that these impulsive and compulsive responses are modulated independently. Cell-type-specific photoinhibition of pyramidal deep layer corticostriatal or corticothalamic neurons reduces behavioral flexibility without inducing premature responses. Together, our data confirm the role of corticostriatal neurons in behavioral flexibility and demonstrate that flexible behaviors are also modulated by direct projections from deep layer corticothalamic neurons in the mPFC to midline thalamic nuclei.SIGNIFICANCE STATEMENT Behavioral flexibility and impulse control are indispensable for animals to adapt to changes in the environment and often affected in patients with PFC damage and obsessive-compulsive disorder. We used a probabilistic reversal task to dissect the underlying neural circuitry in the mPFC. Through characterization of the three major pyramidal cell types in the mPFC with optogenetic silencing, we demonstrated that corticostriatal and corticothalamic but not corticocortical pyramidal neurons are temporally recruited for behavioral flexibility. Together, our findings confirm the role of corticostriatal projections in cognitive flexibility and identify corticothalamic neurons as equally important for behavioral flexibility.


Subject(s)
Behavior, Animal/physiology , Neurons/physiology , Prefrontal Cortex/physiology , Algorithms , Animals , Choice Behavior , Compulsive Behavior/psychology , Corpus Striatum/cytology , Corpus Striatum/physiology , Female , Impulsive Behavior , Male , Mice , Mice, Inbred C57BL , Neural Pathways/cytology , Neural Pathways/physiology , Optogenetics , Pyramidal Cells/physiology , Reaction Time , Thalamus/cytology , Thalamus/physiology
9.
Proc Natl Acad Sci U S A ; 114(49): 13012-13017, 2017 12 05.
Article in English | MEDLINE | ID: mdl-29158387

ABSTRACT

Repeated exposure to drugs of abuse can produce adaptive changes that lead to the establishment of dependence. It has been shown that allelic variation in the α5 nicotinic acetylcholine receptor (nAChR) gene CHRNA5 is associated with higher risk of tobacco dependence. In the brain, α5-containing nAChRs are expressed at very high levels in the interpeduncular nucleus (IPN). Here we identified two nonoverlapping α5 + cell populations (α5- Amigo1 and α5- Epyc ) in mouse IPN that respond differentially to nicotine. Chronic nicotine treatment altered the translational profile of more than 1,000 genes in α5- Amigo1 neurons, including neuronal nitric oxide synthase (Nos1) and somatostatin (Sst). In contrast, expression of few genes was altered in the α5- Epyc population. We show that both nitric oxide and SST suppress optically evoked neurotransmitter release from the terminals of habenular (Hb) neurons in IPN. Moreover, in vivo silencing of neurotransmitter release from the α5- Amigo1 but not from the α5- Epyc population eliminates nicotine reward, measured using place preference. This loss of nicotine reward was mimicked by shRNA-mediated knockdown of Nos1 in the IPN. These findings reveal a proaddiction adaptive response to chronic nicotine in which nitric oxide and SST are released by a specific α5+ neuronal population to provide retrograde inhibition of the Hb-IPN circuit and thereby enhance the motivational properties of nicotine.


Subject(s)
Interpeduncular Nucleus/drug effects , Nicotine/pharmacology , Nitric Oxide Synthase Type I/genetics , Receptors, Nicotinic/genetics , Somatostatin/genetics , Tobacco Use Disorder/genetics , Animals , Female , Gene Expression Profiling , Gene Expression Regulation , Habenula/drug effects , Habenula/metabolism , Habenula/pathology , Interpeduncular Nucleus/metabolism , Interpeduncular Nucleus/pathology , Male , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neurotransmitter Agents/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type I/metabolism , Protein Biosynthesis , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Nicotinic/metabolism , Reward , Somatostatin/metabolism , Stereotaxic Techniques , Synaptic Transmission , Tobacco Use Disorder/metabolism , Tobacco Use Disorder/pathology
10.
Cell ; 167(1): 60-72.e11, 2016 Sep 22.
Article in English | MEDLINE | ID: mdl-27641503

ABSTRACT

The frequency of human social and emotional disorders varies significantly between males and females. We have recently reported that oxytocin receptor interneurons (OxtrINs) modulate female sociosexual behavior. Here, we show that, in male mice, OxtrINs regulate anxiety-related behaviors. We demonstrate that corticotropin-releasing-hormone-binding protein (CRHBP), an antagonist of the stress hormone CRH, is specifically expressed in OxtrINs. Production of CRHBP blocks the CRH-induced potentiation of postsynaptic layer 2/3 pyramidal cell activity of male, but not female, mice, thus producing an anxiolytic effect. Our data identify OxtrINs as critical for modulation of social and emotional behaviors in both females and males and reveal a molecular mechanism that acts on local medial prefrontal cortex (mPFC) circuits to coordinate responses to OXT and CRH. They suggest that additional studies of the impact of the OXT/OXTR and CRHBP/CRH pathways in males and females will be important in development of gender-specific therapies.


Subject(s)
Anxiety/psychology , Carrier Proteins/metabolism , Corticotropin-Releasing Hormone/metabolism , Interneurons/metabolism , Oxytocin/metabolism , Prefrontal Cortex/metabolism , Receptors, Oxytocin/metabolism , Sex Characteristics , Animals , Anxiety/metabolism , Behavior, Animal , Female , Long-Term Potentiation , Male , Metabolic Networks and Pathways , Mice , Sex Factors
11.
Elife ; 4: e11396, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26623516

ABSTRACT

A great deal of interest has been focused recently on the habenula and its critical role in aversion, negative-reward and drug dependence. Using a conditional mouse model of the ACh-synthesizing enzyme choline acetyltransferase (Chat), we report that local elimination of acetylcholine (ACh) in medial habenula (MHb) neurons alters glutamate corelease and presynaptic facilitation. Electron microscopy and immuno-isolation analyses revealed colocalization of ACh and glutamate vesicular transporters in synaptic vesicles (SVs) in the central IPN. Glutamate reuptake in SVs prepared from the IPN was increased by ACh, indicating vesicular synergy. Mice lacking CHAT in habenular neurons were insensitive to nicotine-conditioned reward and withdrawal. These data demonstrate that ACh controls the quantal size and release frequency of glutamate at habenular synapses, and suggest that the synergistic functions of ACh and glutamate may be generally important for modulation of cholinergic circuit function and behavior.


Subject(s)
Acetylcholine/metabolism , Cholinergic Neurons/physiology , Glutamic Acid/metabolism , Habenula/physiology , Synapses/drug effects , Tobacco Use Disorder , Animals , Conditioning, Classical , Mice
12.
Nat Neurosci ; 18(7): 962-4, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26030846

ABSTRACT

Postsynaptic remodeling of glutamatergic synapses on ventral striatum (vSTR) medium spiny neurons (MSNs) is critical for shaping stress responses. However, it is unclear which presynaptic inputs are involved. Susceptible mice exhibited increased synaptic strength at intralaminar thalamus (ILT), but not prefrontal cortex (PFC), inputs to vSTR MSNs following chronic social stress. Modulation of ILT-vSTR versus PFC-vSTR neuronal activity differentially regulated dendritic spine plasticity and social avoidance.


Subject(s)
Dendritic Spines/physiology , Excitatory Postsynaptic Potentials/physiology , Neuronal Plasticity/physiology , Prefrontal Cortex/physiology , Stress, Psychological/physiopathology , Thalamus/physiology , Ventral Striatum/physiology , Animals , Behavior, Animal/physiology , Disease Susceptibility , Male , Mice , Mice, Inbred C57BL , Reward , Social Behavior , Ventral Striatum/cytology
13.
Arthritis Rheumatol ; 67(6): 1657-67, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25733371

ABSTRACT

OBJECTIVE: A hallmark of rheumatoid arthritis (RA) is the chronic pain that accompanies inflammation and joint deformation. Patients with RA rate pain relief as the highest priority; however, few studies have addressed the efficacy and safety of therapies directed specifically toward pain pathways. The ω-conotoxin MVIIA (ziconotide) is used in humans to alleviate persistent pain syndromes, because it specifically blocks the voltage-gated calcium 2.2 (CaV 2.2) channel, which mediates the release of neurotransmitters and proinflammatory mediators from peripheral nociceptor nerve terminals. The aims of this study were to investigate whether blockade of CaV 2.2 can suppress arthritis pain, and to examine the progression of induced arthritis during persistent CaV 2.2 blockade. METHODS: Transgenic mice expressing a membrane-tethered form of MVIIA under the control of a nociceptor-specific gene (MVIIA-transgenic mice) were used in the experiments. The mice were subjected to unilateral induction of joint inflammation using a combination of antigen and collagen. RESULTS: CaV 2.2 blockade mediated by tethered MVIIA effectively suppressed arthritis-induced pain; however, in contrast to their wild-type littermates, which ultimately regained use of their injured joint as inflammation subsided, MVIIA-transgenic mice showed continued inflammation, with up-regulation of the osteoclast activator RANKL and concomitant joint and bone destruction. CONCLUSION: Taken together, our results indicate that alleviation of peripheral pain by blockade of CaV 2.2- mediated calcium influx and signaling in nociceptor sensory neurons impairs recovery from induced arthritis and point to the potentially devastating effects of using CaV 2.2 channel blockers as analgesics during inflammation.


Subject(s)
Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Calcium Channels, N-Type/metabolism , Nociceptive Pain/metabolism , Nociceptors/metabolism , RANK Ligand/metabolism , Stifle/metabolism , omega-Conotoxins/genetics , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Calcium Channel Blockers/therapeutic use , Calcium Channels, N-Type/immunology , Disease Models, Animal , Mice , Mice, Transgenic , Nociceptive Pain/drug therapy , Nociceptive Pain/immunology , Stifle/pathology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , omega-Conotoxins/therapeutic use
14.
J Comp Neurol ; 523(3): 359-80, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25116430

ABSTRACT

The habenula is a phylogenetically conserved brain structure in the epithalamus. It is a major node in the information flow between fronto-limbic brain regions and monoaminergic brainstem nuclei, and is thus anatomically and functionally ideally positioned to regulate emotional, motivational, and cognitive behaviors. Consequently, the habenula may be critically important in the pathophysiology of psychiatric disorders such as addiction and depression. Here we investigated the expression pattern of GPR151, a G protein-coupled receptor (GPCR), whose mRNA has been identified as highly and specifically enriched in habenular neurons by in situ hybridization and translating ribosome affinity purification (TRAP). In the present immunohistochemical study we demonstrate a pronounced and highly specific expression of the GPR151 protein in the medial and lateral habenula of rodent brain. Specific expression was also seen in efferent habenular fibers projecting to the interpeduncular nucleus, the rostromedial tegmental area, the rhabdoid nucleus, the mesencephalic raphe nuclei, and the dorsal tegmental nucleus. Using confocal microscopy and quantitative colocalization analysis, we found that GPR151-expressing axons and terminals overlap with cholinergic, substance P-ergic, and glutamatergic markers. Virtually identical expression patterns were observed in rat, mouse, and zebrafish brains. Our data demonstrate that GPR151 is highly conserved, specific for a subdivision of the habenular neurocircuitry, and constitutes a promising novel target for psychiatric drug development.


Subject(s)
Axons/physiology , Habenula/cytology , Nerve Net/metabolism , Neurons/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Animals , Choline O-Acetyltransferase/metabolism , Humans , Interpeduncular Nucleus/physiology , Mesencephalon/anatomy & histology , Mesencephalon/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurofilament Proteins/metabolism , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/genetics , Receptors, Neuropeptide/genetics , Species Specificity , Substance P/metabolism , Tryptophan Hydroxylase/metabolism , Tyrosine 3-Monooxygenase/metabolism , Vertebrates/anatomy & histology , Vertebrates/metabolism , Vesicular Glutamate Transport Proteins/metabolism , Zebrafish , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
15.
Neuropharmacology ; 96(Pt B): 213-22, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25476971

ABSTRACT

Progress has been made over the last decade in our understanding of the brain areas and circuits involved in nicotine reward and withdrawal, leading to models of addiction that assign different addictive behaviors to distinct, yet overlapping, neural circuits (Koob and Volkow, 2010; Lobo and Nestler, 2011; Tuesta et al., 2011; Volkow et al., 2011). Recently the habenulo-interpeduncular (Hb-IPN) midbrain pathway has re-emerged as a new critical crossroad that influences the brain response to nicotine. This brain area is particularly enriched in nicotinic acetylcholine receptor (nAChR) subunits α5, α3 and ß4 encoded by the CHRNA5-A3-B4 gene cluster, which has been associated with vulnerability to tobacco dependence in human genetics studies. This finding, together with studies in mice involving deletion and replacement of nAChR subunits, and investigations of the circuitry, cell types and electrophysiological properties, have begun to identify the molecular mechanisms that take place in the MHb-IPN which underlie critical aspects of nicotine dependence. In the current review we describe the anatomical and functional connections of the MHb-IPN system, as well as the contribution of specific nAChRs subtypes in nicotine-mediated behaviors. Finally, we discuss the specific electrophysiological properties of MHb-IPN neuronal populations and how nicotine exposure alters their cellular physiology, highlighting the unique role of the MHb-IPN in the context of nicotine aversion and withdrawal. This article is part of the Special Issue entitled 'The Nicotinic Acetylcholine Receptor: From Molecular Biology to Cognition'.


Subject(s)
Habenula/metabolism , Interpeduncular Nucleus/metabolism , Neurons/metabolism , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Receptors, Nicotinic/metabolism , Substance Withdrawal Syndrome/metabolism , Animals , Habenula/cytology , Habenula/drug effects , Humans , Interpeduncular Nucleus/cytology , Interpeduncular Nucleus/drug effects , Mice , Neural Pathways/drug effects , Neural Pathways/metabolism , Neurons/drug effects , Substance Withdrawal Syndrome/physiopathology , Tobacco Use Disorder/complications
16.
Front Hum Neurosci ; 8: 12, 2014.
Article in English | MEDLINE | ID: mdl-24478678

ABSTRACT

The CHRNA5-CHRNA3-CHRNB4 gene cluster, encoding the α5, α3, and ß4 nicotinic acetylcholine receptor (nAChR) subunits, has been linked to nicotine dependence. The habenulo-interpeduncular (Hb-IPN) tract is particularly enriched in α3ß4 nAChRs. We recently showed that modulation of these receptors in the medial habenula (MHb) in mice altered nicotine consumption. Given that ß4 is rate-limiting for receptor activity and that single nucleotide polymorphisms (SNPs) in CHRNB4 have been linked to altered risk of nicotine dependence in humans, we were interested in determining the contribution of allelic variants of ß4 to nicotine receptor activity in the MHb. We screened for missense SNPs that had allele frequencies >0.0005 and introduced the corresponding substitutions in Chrnb4. Fourteen variants were analyzed by co-expression with α3. We found that ß4A90I and ß4T374I variants, previously shown to associate with reduced risk of smoking, and an additional variant ß4D447Y, significantly increased nicotine-evoked current amplitudes, while ß4R348C, the mutation most frequently encountered in sporadic amyotrophic lateral sclerosis (sALS), showed reduced nicotine currents. We employed lentiviruses to express ß4 or ß4 variants in the MHb. Immunoprecipitation studies confirmed that ß4 lentiviral-mediated expression leads to specific upregulation of α3ß4 but not ß2 nAChRs in the Mhb. Mice injected with the ß4-containing virus showed pronounced aversion to nicotine as previously observed in transgenic Tabac mice overexpressing Chrnb4 at endogenous sites including the MHb. Habenular expression of the ß4 gain-of-function allele T374I also resulted in strong aversion, while transduction with the ß4 loss-of function allele R348C failed to induce nicotine aversion. Altogether, these data confirm the critical role of habenular ß4 in nicotine consumption, and identify specific SNPs in CHRNB4 that modify nicotine-elicited currents and alter nicotine consumption in mice.

17.
Proc Natl Acad Sci U S A ; 110(42): 17077-82, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24082085

ABSTRACT

The discovery of genetic variants in the cholinergic receptor nicotinic CHRNA5-CHRNA3-CHRNB4 gene cluster associated with heavy smoking and higher relapse risk has led to the identification of the midbrain habenula-interpeduncular axis as a critical relay circuit in the control of nicotine dependence. Although clear roles for α3, ß4, and α5 receptors in nicotine aversion and withdrawal have been established, the cellular and molecular mechanisms that participate in signaling nicotine use and contribute to relapse have not been identified. Here, using translating ribosome affinity purification (TRAP) profiling, electrophysiology, and behavior, we demonstrate that cholinergic neurons, but not peptidergic neurons, of the medial habenula (MHb) display spontaneous tonic firing of 2-10 Hz generated by hyperpolarization-activated cyclic nucleotide-gated (HCN) pacemaker channels and that infusion of the HCN pacemaker antagonist ZD7288 in the habenula precipitates somatic and affective signs of withdrawal. Further, we show that a strong, α3ß4-dependent increase in firing frequency is observed in these pacemaker neurons upon acute exposure to nicotine. No change in the basal or nicotine-induced firing was observed in cholinergic MHb neurons from mice chronically treated with nicotine. We observe, however, that, during withdrawal, reexposure to nicotine doubles the frequency of pacemaking activity in these neurons. These findings demonstrate that the pacemaking mechanism of cholinergic MHb neurons controls withdrawal, suggesting that the heightened nicotine sensitivity of these neurons during withdrawal may contribute to smoking relapse.


Subject(s)
Biological Clocks/drug effects , Cholinergic Neurons , Habenula , Nicotine/adverse effects , Nicotinic Agonists/adverse effects , Substance Withdrawal Syndrome , Animals , Cardiotonic Agents/pharmacology , Cholinergic Neurons/metabolism , Cholinergic Neurons/pathology , Habenula/metabolism , Habenula/pathology , Habenula/physiopathology , Humans , Mice , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Pyrimidines/pharmacology , Smoking/metabolism , Smoking/pathology , Smoking/physiopathology , Smoking Cessation , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/pathology , Substance Withdrawal Syndrome/physiopathology
18.
PLoS One ; 7(6): e39572, 2012.
Article in English | MEDLINE | ID: mdl-22768092

ABSTRACT

Classical electron microscopic studies of the mammalian brain revealed two major classes of synapses, distinguished by the presence of a large postsynaptic density (PSD) exclusively at type 1, excitatory synapses. Biochemical studies of the PSD have established the paradigm of the synapse as a complex signal-processing machine that controls synaptic plasticity. We report here the results of a proteomic analysis of type 2, inhibitory synaptic complexes isolated by affinity purification from the cerebral cortex. We show that these synaptic complexes contain a variety of neurotransmitter receptors, neural cell-scaffolding and adhesion molecules, but that they are entirely lacking in cell signaling proteins. This fundamental distinction between the functions of type 1 and type 2 synapses in the nervous system has far reaching implications for models of synaptic plasticity, rapid adaptations in neural circuits, and homeostatic mechanisms controlling the balance of excitation and inhibition in the mature brain.


Subject(s)
Cerebral Cortex/metabolism , Neural Inhibition/physiology , Synapses/metabolism , Animals , HEK293 Cells , Humans , Mass Spectrometry , Mice , Mice, Transgenic , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/isolation & purification , Nerve Tissue Proteins/metabolism , Protein Transport , Receptors, GABA-A/metabolism , Synapses/ultrastructure , Xenopus
19.
Proc Natl Acad Sci U S A ; 109(28): 11360-5, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22733786

ABSTRACT

A large number of studies have demonstrated that the nucleus accumbens (NAC) is a critical site in the neuronal circuits controlling reward responses, motivation, and mood, but the neuronal cell type(s) underlying these processes are not yet known. Identification of the neuronal cell types that regulate depression-like states will guide us in understanding the biological basis of mood and its regulation by diseases like major depressive disorder. Taking advantage of recent findings demonstrating that the serotonin receptor chaperone, p11, is an important molecular regulator of depression-like states, here we identify cholinergic interneurons (CINs) as a primary site of action for p11 in the NAC. Depression-like behavior is observed in mice after decrease of p11 levels in NAC CINs. This phenotype is recapitulated by silencing neuronal transmission in these cells, demonstrating that accumbal cholinergic neuronal activity regulates depression-like behaviors and suggesting that accumbal CIN activity is crucial for the regulation of mood and motivation.


Subject(s)
Annexin A2/metabolism , Depression/physiopathology , Interneurons/metabolism , Nucleus Accumbens/metabolism , S100 Proteins/metabolism , Acetylcholine/metabolism , Animals , Antidepressive Agents/pharmacology , Behavior, Animal , Depression/metabolism , Immunohistochemistry/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Molecular Chaperones/metabolism , Neurons/metabolism , Neurotransmitter Agents/metabolism , Phenotype , Receptors, Cholinergic/metabolism
20.
Life Sci ; 91(21-22): 1003-8, 2012 Nov 27.
Article in English | MEDLINE | ID: mdl-22480510

ABSTRACT

AIMS: In the oviduct, muscarinic acetylcholine receptors (MR) are linked with motility regulation and nicotinic receptors (nAChR) with ectopic pregnancy. We here aimed to determine the repertoire of cholinergic receptor expression in the murine oviduct and their functional coupling to regulation of intracellular calcium concentration ([Ca(2+)](i)). MAIN METHODS: Cholinergic receptor transcripts were assessed by RT-PCR in oviductal segments (ampulla, isthmus, uterotubar junction) in all cyclic stages and pregnancy, and in laser-microdissected samples of epithelium and smooth muscle, nAChR subunit α3 distribution in tissue sections using an appropriate genetic reporter mouse strain. [Ca(2+)](i) responses were monitored in ciliated and non-ciliated oviductal cells isolated from wild-type and MR subtypes 1 and 3 gene deficient mice. KEY FINDINGS: Transcripts for all MR subtypes (M1-M5) are constantly expressed whereas there is some variability in nAChR expression from individual to individual. The qualitative expression pattern is independent from the hormonal status of the animal, except for nAChR α7, which is less present during pregnancy. The epithelium expresses M1, M3, nAChR α7 (data from laser-assisted microdissection) and nAChR α3 (ultrastructural investigation of reporter mice). MR dominate over nAChR in increasing [Ca(2+)](i) with being M3 the major, but not sole subtype driving this effect. The general nAChR inhibitor mecamylamine enhances muscarinic and purinergic responses. SIGNIFICANCE: In conclusion, the murine oviduct is endowed with a multiplicity of muscarinic and nicotinic receptors subtypes that, with respect to regulation of [Ca(2+)](i), are inversely linked to each other. The major, but not sole, cholinergic receptor driving increase in [Ca(2+)](i) is M3.


Subject(s)
Calcium/metabolism , Fallopian Tubes/metabolism , Receptors, Cholinergic/analysis , Receptors, Cholinergic/genetics , Animals , Fallopian Tubes/ultrastructure , Female , Mice , Mice, Inbred C57BL , Pregnancy , Receptors, Cholinergic/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...