Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros











Intervalo de año de publicación
1.
Neuropsychopharmacology ; 47(3): 641-651, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34326477

RESUMEN

A critical brain area implicated in nicotine dependence is the interpeduncular nucleus (IPN) located in the ventral midbrain and consisting primarily of GABAergic neurons. Previous studies indicate that IPN GABAergic neurons contribute to expression of somatic symptoms of nicotine withdrawal; however, whether IPN neurons are dynamically regulated during withdrawal in vivo and how this may contribute to both somatic and affective withdrawal behavior is unknown. To bridge this gap in knowledge, we expressed GCaMP in IPN GABAergic neurons and used in vivo fiber photometry to record changes in fluorescence, as a proxy for neuronal activity, in male mice during nicotine withdrawal. Mecamylamine-precipitated withdrawal significantly increased activity of IPN GABAergic neurons in nicotine-dependent, but not nicotine-naive mice. Analysis of GCaMP signals time-locked with somatic symptoms including grooming and scratching revealed reduced IPN GABAergic activity during these behaviors, specifically in mice undergoing withdrawal. In the elevated plus maze, used to measure anxiety-like behavior, an affective withdrawal symptom, IPN GABAergic neuron activity was increased during open-arm versus closed-arm exploration in nicotine-withdrawn, but not non-withdrawn mice. Optogenetic silencing IPN GABAergic neurons during withdrawal significantly reduced withdrawal-induced increases in somatic behavior and increased open-arm exploration. Together, our data indicate that IPN GABAergic neurons are dynamically regulated during nicotine withdrawal, leading to increased anxiety-like symptoms and somatic behavior, which inherently decrease IPN GABAergic neuron activity as a withdrawal-coping mechanism. These results provide a neuronal basis underlying the role of the IPN in the expression of somatic and affective behaviors of nicotine withdrawal.


Asunto(s)
Núcleo Interpeduncular , Síndrome de Abstinencia a Sustancias , Animales , Neuronas GABAérgicas , Núcleo Interpeduncular/metabolismo , Masculino , Mecamilamina/farmacología , Ratones , Nicotina/farmacología , Síndrome de Abstinencia a Sustancias/metabolismo
2.
Sci Rep ; 10(1): 813, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31965003

RESUMEN

Tobacco use is the leading preventable cause of mortality in the world. The limited number of smoking cessation aids currently available are minimally effective, highlighting the need for novel therapeutic interventions. We describe a genome-wide approach to identify potential candidates for such interventions. Next-generation sequencing was performed using RNA isolated from the habenulo-interpeduncular circuit of male mice withdrawn from chronic nicotine treatment. This circuit plays a central role in the nicotine withdrawal response. Differentially expressed miRNAs and mRNAs were validated using RT-qPCR. Many of the differentially expressed mRNAs are predicted targets of reciprocally expressed miRNAs. We illustrate the utility of the dataset by demonstrating that knockdown in the interpeduncular nucleus of a differentially expressed mRNA, that encoding profilin 2, is sufficient to induce anxiety-related behavior. Importantly, profilin 2 knockdown in the ventral tegmental area did not affect anxiety behavior. Our data reveal wide-spread changes in gene expression within the habenulo-interpeduncular circuit during nicotine withdrawal. This dataset should prove to be a valuable resource leading to the identification of substrates for the design of innovative smoking cessation aids.


Asunto(s)
Habénula/fisiología , Núcleo Interpeduncular/fisiología , MicroARNs/genética , Nicotina , ARN Mensajero/genética , Síndrome de Abstinencia a Sustancias/genética , Animales , Ansiedad/genética , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Masculino , Ratones Endogámicos C57BL , Profilinas/genética
3.
Handb Exp Pharmacol ; 248: 187-212, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29423839

RESUMEN

Ethanol and nicotine can modulate the activity of several neurotransmitter systems and signalling pathways. Interactions between ethanol and nicotine can also occur via common molecular targets including nicotinic acetylcholine receptors (nAChRs). These effects can induce molecular and synaptic adaptations that over time, are consolidated in brain circuits that reinforce drug-seeking behavior, contribute to the development of withdrawal symptoms during abstinence and increase the susceptibility to relapse. This chapter will discuss the acute and chronic effects of ethanol and nicotine within the mesolimbic reward pathway and brain circuits involved in learning, memory, and withdrawal. Individual and common molecular targets of ethanol and nicotine within these circuits are also discussed. Finally, we review studies that have identified potential molecular and neuronal processes underlying the high incidence of ethanol and nicotine co-use that may contribute to the development of ethanol and nicotine co-addiction.


Asunto(s)
Etanol/farmacología , Nicotina/farmacología , Receptores Nicotínicos , Síndrome de Abstinencia a Sustancias , Tabaquismo , Interacciones Farmacológicas , Humanos
4.
Elife ; 62017 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-28196335

RESUMEN

Paternal environmental conditions can influence phenotypes in future generations, but it is unclear whether offspring phenotypes represent specific responses to particular aspects of the paternal exposure history, or a generic response to paternal 'quality of life'. Here, we establish a paternal effect model based on nicotine exposure in mice, enabling pharmacological interrogation of the specificity of the offspring response. Paternal exposure to nicotine prior to reproduction induced a broad protective response to multiple xenobiotics in male offspring. This effect manifested as increased survival following injection of toxic levels of either nicotine or cocaine, accompanied by hepatic upregulation of xenobiotic processing genes, and enhanced drug clearance. Surprisingly, this protective effect could also be induced by a nicotinic receptor antagonist, suggesting that xenobiotic exposure, rather than nicotinic receptor signaling, is responsible for programming offspring drug resistance. Thus, paternal drug exposure induces a protective phenotype in offspring by enhancing metabolic tolerance to xenobiotics.


Asunto(s)
Exposición a Riesgos Ambientales , Nicotina/metabolismo , Agonistas Nicotínicos/metabolismo , Exposición Paterna , Herencia Paterna , Xenobióticos/metabolismo , Animales , Resistencia a Medicamentos , Femenino , Inactivación Metabólica , Hígado/metabolismo , Masculino , Ratones , Análisis de Supervivencia
5.
Trends Pharmacol Sci ; 38(2): 169-180, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27890353

RESUMEN

While innovative modern neuroscience approaches have aided in discerning brain circuitry underlying negative emotional behaviors including fear and anxiety responses, how these circuits are recruited in normal and pathological conditions remains poorly understood. Recently, genetic tools that selectively manipulate single neuronal populations have uncovered an understudied circuit, the medial habenula (mHb)-interpeduncular (IPN) axis, that modulates basal negative emotional responses. Interestingly, the mHb-IPN pathway also represents an essential circuit that signals heightened anxiety induced by nicotine withdrawal. Insights into how this circuit interconnects with regions more classically associated with anxiety, and how chronic nicotine exposure induces neuroadaptations resulting in an anxiogenic state, may thereby provide novel strategies and molecular targets for therapies that facilitate smoking cessation, as well as for anxiety relief.


Asunto(s)
Trastornos de Ansiedad/etiología , Habénula/fisiología , Núcleo Interpeduncular/fisiología , Tabaquismo/etiología , Animales , Trastornos de Ansiedad/tratamiento farmacológico , Hormona Liberadora de Corticotropina/fisiología , Emociones , Humanos , Ratones , Receptores Nicotínicos/fisiología , Tabaquismo/tratamiento farmacológico , Área Tegmental Ventral/fisiología
6.
Alcohol ; 57: 65-70, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27793544

RESUMEN

The prevalent co-abuse of nicotine and alcohol suggests a common neural mechanism underlying the actions of the two drugs. Nicotine, the addictive component of tobacco, activates nicotinic acetylcholine receptors (nAChRs) containing the α6 subunit (α6* nAChRs) in dopaminergic (DAergic) neurons of the ventral tegmental area (VTA), a region known to be crucial for drug reward. Recent evidence suggests that ethanol may potentiate ACh activation of these receptors as well, although whether α6* nAChR expression is necessary for behavioral effects of acute ethanol exposure is unknown. We compared binge-like ethanol consumption and ethanol reward sensitivity between knockout (KO) mice that do not express chrna6 (the gene encoding the α6 nAChR subunit, the α6 KO line) and wild-type (WT) littermates using the Drinking-in-the-Dark (DID) and Conditioned Place Preference (CPP) assay, respectively. In the DID assay, α6 KO female and male mice consumed ethanol similarly to WT mice at all concentrations tested. In the CPP assay, 2.0-g/kg and 3.0-g/kg, but not 0.5-mg/kg, ethanol conditioned a place preference in WT female and male mice, whereas only 2.0-g/kg ethanol conditioned a place preference in α6 KO mice. Acute challenge with ethanol reduced locomotor activity, an effect that developed tolerance with repeated injections, similarly between genotypes in both female and male mice. Together, these data indicate that expression of α6* nAChRs is not required for binge-like ethanol consumption and reward, but modulate sensitivity to the rewarding properties of the drug.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Etanol/administración & dosificación , Subunidades de Proteína/deficiencia , Receptores Nicotínicos/deficiencia , Recompensa , Consumo de Bebidas Alcohólicas/psicología , Animales , Relación Dosis-Respuesta a Droga , Femenino , Locomoción/efectos de los fármacos , Locomoción/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
7.
Neuropharmacology ; 107: 294-304, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27020042

RESUMEN

Cholinergic neurons in the medial habenula (MHb) modulate anxiety during nicotine withdrawal although the molecular neuroadaptation(s) within the MHb that induce affective behaviors during nicotine cessation is largely unknown. MHb cholinergic neurons are unique in that they robustly express neuronal nicotinic acetylcholine receptors (nAChRs), although their behavioral role as autoreceptors in these neurons has not been described. To test the hypothesis that nAChR signaling in MHb cholinergic neurons could modulate anxiety, we expressed novel "gain of function" nAChR subunits selectively in MHb cholinergic neurons of adult mice. Mice expressing these mutant nAChRs exhibited increased anxiety-like behavior that was alleviated by blockade with a nAChR antagonist. To test the hypothesis that anxiety induced by nicotine withdrawal may be mediated by increased MHb nicotinic receptor signaling, we infused nAChR subtype selective antagonists into the MHb of nicotine naïve and withdrawn mice. While antagonists had little effect on nicotine naïve mice, blocking α4ß2 or α6ß2, but not α3ß4 nAChRs in the MHb alleviated anxiety in mice undergoing nicotine withdrawal. Consistent with behavioral results, there was increased functional expression of nAChRs containing the α6 subunit in MHb neurons that also expressed the α4 subunit. Together, these data indicate that MHb cholinergic neurons regulate nicotine withdrawal-induced anxiety via increased signaling through nicotinic receptors containing the α6 subunit and point toward nAChRs in MHb cholinergic neurons as molecular targets for smoking cessation therapeutics.


Asunto(s)
Ansiedad/metabolismo , Neuronas Colinérgicas/metabolismo , Habénula/metabolismo , Nicotina/efectos adversos , Receptores Nicotínicos/biosíntesis , Síndrome de Abstinencia a Sustancias/metabolismo , Animales , Ansiedad/psicología , Neuronas Colinérgicas/efectos de los fármacos , Habénula/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Agonistas Nicotínicos/farmacología , Síndrome de Abstinencia a Sustancias/psicología
8.
Alcohol Clin Exp Res ; 40(1): 62-72, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26727524

RESUMEN

BACKGROUND: Ethanol (EtOH) and nicotine abuse are 2 leading causes of preventable mortality in the world, but little is known about the pharmacological mechanisms mediating co-abuse. Few studies have examined the interaction of the acute effects of EtOH and nicotine. Here, we examine the effects of nicotine administration on the duration of EtOH-induced loss of righting reflex (LORR) and characterize the nature of their pharmacological interactions in C57BL/6J mice. METHODS: We assessed the effects of EtOH and nicotine and the nature of their interaction in the LORR test using isobolographic analysis after acute injection in C57BL/6J male mice. Next, we examined the importance of receptor efficacy using nicotinic partial agonists varenicline and sazetidine. We evaluated the involvement of major nicotinic acetylcholine receptor (nAChR) subtypes using nicotinic antagonist mecamylamine and nicotinic α4- and α7-knockout mice. The selectivity of nicotine's actions on EtOH-induced LORR was examined by testing nicotine's effects on the hypnotic properties of ketamine and pentobarbital. We also assessed the development of tolerance after repeated nicotine exposure. Last, we assessed whether the effects of nicotine on EtOH-induced LORR extend to hypothermia and EtOH intake in the drinking in the dark (DID) paradigm. RESULTS: We found that acute nicotine injection enhances EtOH's hypnotic effects in a synergistic manner and that receptor efficacy plays an important role in this interaction. Furthermore, tolerance developed to the enhancement of EtOH's hypnotic effects by nicotine after repeated exposure of the drug. α4* and α7 nAChRs seem to play an important role in nicotine-EtOH interaction in the LORR test. In addition, the magnitude of EtOH-induced LORR enhancement by nicotine was more pronounced in C57BL/6J than DBA/2J mice. Furthermore, acute nicotine enhanced ketamine and pentobarbital hypnotic effects in the mouse. Finally, nicotine enhanced EtOH-induced hypothermia but decreased EtOH intake in the DID test. CONCLUSIONS: Our results demonstrate that nicotine synergistically enhances EtOH-induced LORR in the mouse.


Asunto(s)
Temperatura Corporal/efectos de los fármacos , Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Reflejo de Enderezamiento/efectos de los fármacos , Animales , Azetidinas/farmacología , Interacciones Farmacológicas , Hipnóticos y Sedantes/farmacología , Hipotermia , Ketamina/farmacología , Mecamilamina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Antagonistas Nicotínicos/farmacología , Pentobarbital/farmacología , Piridinas/farmacología , Receptores Nicotínicos/genética , Vareniclina/farmacología , Receptor Nicotínico de Acetilcolina alfa 7/genética
9.
J Neurosci ; 35(22): 8570-8, 2015 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-26041923

RESUMEN

Chronic nicotine exposure increases sensitivity to nicotine reward during a withdrawal period, which may facilitate relapse in abstinent smokers, yet the molecular neuroadaptation(s) that contribute to this phenomenon are unknown. Interestingly, chronic nicotine use induces functional upregulation of nicotinic acetylcholine receptors (nAChRs) in the mesocorticolimbic reward pathway potentially linking upregulation to increased drug sensitivity. In the ventral tegmental area (VTA), functional upregulation of nAChRs containing the α4 subunit (α4* nAChRs) is restricted to GABAergic neurons. To test the hypothesis that increased functional expression of α4* nAChRs in these neurons modulates nicotine reward behaviors, we engineered a Cre recombinase-dependent gene expression system to selectively express α4 nAChR subunits harboring a "gain-of-function" mutation [a leucine mutated to a serine residue at the 9' position (Leu9'Ser)] in VTA GABAergic neurons of adult mice. In mice expressing Leu9'Ser α4 nAChR subunits in VTA GABAergic neurons (Gad2(VTA):Leu9'Ser mice), subreward threshold doses of nicotine were sufficient to selectively activate VTA GABAergic neurons and elicit acute hypolocomotion, with subsequent nicotine exposures eliciting tolerance to this effect, compared to control animals. In the conditioned place preference procedure, nicotine was sufficient to condition a significant place preference in Gad2(VTA):Leu9'Ser mice at low nicotine doses that failed to condition control animals. Together, these data indicate that functional upregulation of α4* nAChRs in VTA GABAergic neurons confers increased sensitivity to nicotine reward and points to nAChR subtypes specifically expressed in GABAergic VTA neurons as molecular targets for smoking cessation therapeutics.


Asunto(s)
Neuronas GABAérgicas/fisiología , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Receptores Nicotínicos/metabolismo , Recompensa , Regulación hacia Arriba/genética , Área Tegmental Ventral/citología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Calbindina 2/metabolismo , Calbindinas/metabolismo , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Operante/fisiología , Dihidro-beta-Eritroidina/farmacología , Relación Dosis-Respuesta a Droga , Neuronas GABAérgicas/efectos de los fármacos , Glutamato Descarboxilasa/genética , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores Nicotínicos/genética , Tirosina 3-Monooxigenasa/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Área Tegmental Ventral/efectos de los fármacos
10.
Nat Commun ; 6: 6770, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25898242

RESUMEN

Increased anxiety is a prominent withdrawal symptom in abstinent smokers, yet the neuroanatomical and molecular bases underlying it are unclear. Here we show that withdrawal-induced anxiety increases activity of neurons in the interpeduncular intermediate (IPI), a subregion of the interpeduncular nucleus (IPN). IPI activation during nicotine withdrawal was mediated by increased corticotropin releasing factor (CRF) receptor-1 expression and signalling, which modulated glutamatergic input from the medial habenula (MHb). Pharmacological blockade of IPN CRF1 receptors or optogenetic silencing of MHb input reduced IPI activation and alleviated withdrawal-induced anxiety; whereas IPN CRF infusion in mice increased anxiety. We identified a mesointerpeduncular circuit, consisting of ventral tegmental area (VTA) dopaminergic neurons projecting to the IPN, as a potential source of CRF. Knockdown of CRF synthesis in the VTA prevented IPI activation and anxiety during nicotine withdrawal. These data indicate that increased CRF receptor signalling within a VTA-IPN-MHb circuit triggers anxiety during nicotine withdrawal.


Asunto(s)
Ansiedad/etiología , Hormona Liberadora de Corticotropina/metabolismo , Habénula/fisiología , Núcleo Interpeduncular/fisiología , Nicotina/efectos adversos , Área Tegmental Ventral/fisiología , Animales , Habénula/anatomía & histología , Masculino , Ratones , Ratones Endogámicos C57BL , Red Nerviosa , Receptores de Hormona Liberadora de Corticotropina/genética , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Transducción de Señal/fisiología , Síndrome de Abstinencia a Sustancias/fisiopatología
11.
Nat Neurosci ; 17(12): 1751-8, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25402857

RESUMEN

Dopaminergic neurons in the ventral tegmental area (VTA) are well known for mediating the positive reinforcing effects of drugs of abuse. Here we identify in rodents and humans a population of VTA dopaminergic neurons expressing corticotropin-releasing factor (CRF). We provide further evidence in rodents that chronic nicotine exposure upregulates Crh mRNA (encoding CRF) in dopaminergic neurons of the posterior VTA, activates local CRF1 receptors and blocks nicotine-induced activation of transient GABAergic input to dopaminergic neurons. Local downregulation of Crh mRNA and specific pharmacological blockade of CRF1 receptors in the VTA reversed the effect of nicotine on GABAergic input to dopaminergic neurons, prevented the aversive effects of nicotine withdrawal and limited the escalation of nicotine intake. These results link the brain reward and stress systems in the same brain region to signaling of the negative motivational effects of nicotine withdrawal.


Asunto(s)
Hormona Liberadora de Corticotropina/fisiología , Neuronas/metabolismo , Nicotina/efectos adversos , Síndrome de Abstinencia a Sustancias/metabolismo , Área Tegmental Ventral/metabolismo , Animales , Humanos , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , Ratas , Ratas Wistar , Síndrome de Abstinencia a Sustancias/psicología , Área Tegmental Ventral/efectos de los fármacos
12.
RNA ; 20(12): 1890-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25344397

RESUMEN

Nicotine binds to and activates a family of ligand-gated ion channels, neuronal nicotinic acetylcholine receptors (nAChRs). Chronic nicotine exposure alters the expression of various nAChR subtypes, which likely contributes to nicotine dependence; however, the underlying mechanisms regulating these changes remain unclear. A growing body of evidence indicates that microRNAs (miRNAs) may be involved in nAChR regulation. Using bioinformatics, miRNA library screening, site-directed mutagenesis, and gene expression analysis, we have identified a limited number of miRNAs that functionally interact with the 3'-untranslated regions (3' UTRs) of mammalian neuronal nAChR subunit genes. In silico analyses revealed specific, evolutionarily conserved sites within the 3' UTRs through which the miRNAs regulate gene expression. Mutating these sites disrupted miRNA regulation confirming the in silico predictions. In addition, the miRNAs that target nAChR 3' UTRs are expressed in mouse brain and are regulated by chronic nicotine exposure. Furthermore, we show that expression of one of these miRNAs, miR-542-3p, is modulated by nicotine within the mesocorticolimbic reward pathway. Importantly, overexpression of miR-542-3p led to a decrease in the protein levels of its target, the nAChR ß2 subunit. Bioinformatic analysis suggests that a number of the miRNAs play a general role in regulating cholinergic signaling. Our results provide evidence for a novel mode of nicotine-mediated regulation of the mammalian nAChR gene family.


Asunto(s)
Canales Iónicos/biosíntesis , MicroARNs/biosíntesis , Nicotina/metabolismo , Receptores Nicotínicos/genética , Regiones no Traducidas 3' , Animales , Encéfalo/metabolismo , Regulación de la Expresión Génica/genética , Canales Iónicos/genética , Ligandos , Mamíferos , Ratones , MicroARNs/genética , Mutagénesis Sitio-Dirigida , Especificidad de Órganos , Receptores Nicotínicos/biosíntesis , Transducción de Señal/genética
13.
Curr Biol ; 23(23): 2327-35, 2013 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-24239118

RESUMEN

BACKGROUND: Chronic exposure to nicotine elicits physical dependence in smokers, yet the mechanism and neuroanatomical bases for withdrawal symptoms are unclear. As in humans, rodents undergo physical withdrawal symptoms after cessation from chronic nicotine characterized by increased scratching, head nods, and body shakes. RESULTS: Here we show that induction of physical nicotine withdrawal symptoms activates GABAergic neurons within the interpeduncular nucleus (IPN). Optical activation of IPN GABAergic neurons via light stimulation of channelrhodopsin elicited physical withdrawal symptoms in both nicotine-naive and chronic-nicotine-exposed mice. Dampening excitability of GABAergic neurons during nicotine withdrawal through IPN-selective infusion of an NMDA receptor antagonist or through blockade of IPN neurotransmission from the medial habenula reduced IPN neuronal activation and alleviated withdrawal symptoms. During chronic nicotine exposure, nicotinic acetylcholine receptors containing the ß4 subunit were upregulated in somatostatin interneurons clustered in the dorsal region of the IPN. Blockade of these receptors induced withdrawal signs more dramatically in nicotine-dependent compared to nicotine-naive mice and activated nonsomatostatin neurons in the IPN. CONCLUSIONS: Together, our data indicate that therapeutic strategies to reduce IPN GABAergic neuron excitability during nicotine withdrawal, for example, by activating nicotinic receptors on somatostatin interneurons, may be beneficial for alleviating withdrawal symptoms and facilitating smoking cessation.


Asunto(s)
Neuronas GABAérgicas/metabolismo , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Animales , Neuronas GABAérgicas/efectos de la radiación , Ácido Glutámico/metabolismo , Luz , Masculino , Mecamilamina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Nicotina/administración & dosificación , Antagonistas Nicotínicos/farmacología , Fototerapia/métodos , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores Nicotínicos/biosíntesis , Rodopsina/biosíntesis , Rodopsina/genética , Somatostatina , Transmisión Sináptica/fisiología
14.
Front Physiol ; 4: 251, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24062692

RESUMEN

Ion channels modulate ion flux across cell membranes, activate signal transduction pathways, and influence cellular transport-vital biological functions that are inexorably linked to cellular processes that go awry during carcinogenesis. Indeed, deregulation of ion channel function has been implicated in cancer-related phenomena such as unrestrained cell proliferation and apoptotic evasion. As the prototype for ligand-gated ion channels, nicotinic acetylcholine receptors (nAChRs) have been extensively studied in the context of neuronal cells but accumulating evidence also indicate a role for nAChRs in carcinogenesis. Recently, variants in the nAChR genes CHRNA3, CHRNA5, and CHRNB4 have been implicated in nicotine dependence and lung cancer susceptibility. Here, we silenced the expression of these three genes to investigate their function in lung cancer. We show that these genes are necessary for the viability of small cell lung carcinomas (SCLC), the most aggressive type of lung cancer. Furthermore, we show that nicotine promotes SCLC cell viability whereas an α3ß4-selective antagonist, α-conotoxin AuIB, inhibits it. Our findings posit a mechanism whereby signaling via α3/α5/ß4-containing nAChRs promotes lung carcinogenesis.

15.
Front Psychiatry ; 4: 29, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23641218

RESUMEN

Alcohol and nicotine are often co-abused. As many as 80-95% of alcoholics are also smokers, suggesting that ethanol and nicotine, the primary addictive component of tobacco smoke, may functionally interact in the central nervous system and/or share a common mechanism of action. While nicotine initiates dependence by binding to and activating neuronal nicotinic acetylcholine receptors (nAChRs), ligand-gated cation channels normally activated by endogenous acetylcholine (ACh), ethanol is much less specific with the ability to modulate multiple gene products including those encoding voltage-gated ion channels, and excitatory/inhibitory neurotransmitter receptors. However, emerging data indicate that ethanol interacts with nAChRs, both directly and indirectly, in the mesocorticolimbic dopaminergic (DAergic) reward circuitry to affect brain reward systems. Like nicotine, ethanol activates DAergic neurons of the ventral tegmental area (VTA) which project to the nucleus accumbens (NAc). Blockade of VTA nAChRs reduces ethanol-mediated activation of DAergic neurons, NAc DA release, consumption, and operant responding for ethanol in rodents. Thus, ethanol may increase ACh release into the VTA driving activation of DAergic neurons through nAChRs. In addition, ethanol potentiates distinct nAChR subtype responses to ACh and nicotine in vitro and in DAergic neurons. The smoking cessation therapeutic and nAChR partial agonist, varenicline, reduces alcohol consumption in heavy drinking smokers and rodent models of alcohol consumption. Finally, single nucleotide polymorphisms in nAChR subunit genes are associated with alcohol dependence phenotypes and smoking behaviors in human populations. Together, results from pre-clinical, clinical, and genetic studies indicate that nAChRs may have an inherent role in the abusive properties of ethanol, as well as in nicotine and alcohol co-dependence.

16.
Neuropharmacology ; 73: 19-30, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23688922

RESUMEN

Nicotinic acetylcholine receptors (nAChRs) containing either the α4 and/or α6 subunit are robustly expressed in dopaminergic nerve terminals in dorsal striatum where they are hypothesized to modulate dopamine (DA) release via acetylcholine (ACh) stimulation from cholinergic interneurons. However, pharmacological blockade of nAChRs or genetic deletion of individual nAChR subunits, including α4 and α6, in mice, yields little effect on motor behavior. Based on the putative role of nAChRs containing the α4 subunit in modulation of DA in dorsal striatum, we hypothesized that mice expressing a single point mutation in the α4 nAChR subunit, Leu9'Ala, that renders nAChRs hypersensitive to agonist, would exhibit exaggerated differences in motor behavior compared to WT mice. To gain insight into these differences, we challenged WT and Leu9'Ala mice with the α4ß2 nAChR antagonist dihydro-ß-erythroidine (DHßE). Interestingly, in Leu9'Ala mice, DHßE elicited a robust, reversible motor impairment characterized by hypolocomotion, akinesia, catalepsy, clasping, and tremor; whereas the antagonist had little effect in WT mice at all doses tested. Pre-injection of nicotine (0.1 mg/kg) blocked DHßE-induced motor impairment in Leu9'Ala mice confirming that the phenotype was mediated by antagonism of nAChRs. In addition, SKF82958 (1 mg/kg) and amphetamine (5 mg/kg) prevented the motor phenotype. DHßE significantly activated more neurons within striatum and substantia nigra pars reticulata in Leu9'Ala mice compared to WT animals, suggesting activation of the indirect motor pathway as the circuit underlying motor dysfunction. ACh evoked DA release from Leu9'Ala striatal synaptosomes revealed agonist hypersensitivity only at α4(non-α6)* nAChRs. Similarly, α6 nAChR subunit deletion in an α4 hypersensitive nAChR (Leu9'Ala/α6 KO) background had little effect on the DHßE-induced phenotype, suggesting an α4(non-α6)* nAChR-dependent mechanism. Together, these data indicate that α4(non-α6)* nAChR have an impact on motor output and may be potential molecular targets for treatment of disorders associated with motor impairment.


Asunto(s)
Conducta Animal/fisiología , Receptores Nicotínicos/fisiología , Acetilcolina/farmacología , Anfetamina/farmacología , Animales , Conducta Animal/efectos de los fármacos , Benzazepinas/farmacología , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/fisiología , Dihidro-beta-Eritroidina/antagonistas & inhibidores , Dihidro-beta-Eritroidina/farmacología , Dopamina/metabolismo , Agonistas de Dopamina/farmacología , Femenino , Técnicas de Sustitución del Gen , Masculino , Ratones , Ratones Noqueados , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Antagonistas Nicotínicos/farmacología , Mutación Puntual , Receptores Nicotínicos/efectos de los fármacos , Receptores Nicotínicos/genética , Sustancia Negra/efectos de los fármacos , Sustancia Negra/fisiología , Sinaptosomas/efectos de los fármacos , Sinaptosomas/metabolismo
17.
Mol Pharmacol ; 81(4): 541-8, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22222765

RESUMEN

Nicotine is reinforcing because it activates dopaminergic (DAergic) neurons within the ventral tegmental area (VTA) of the brain's mesocorticolimbic reward circuitry. This increase in activity can occur for a period of several minutes up to an hour and is thought to be a critical component of nicotine dependence. However, nicotine concentrations that are routinely self-administered by smokers are predicted to desensitize high-affinity α4ß2 neuronal nicotinic acetylcholine receptors (nAChRs) in seconds. Thus, how physiologically relevant nicotine concentrations persistently activate VTA DAergic neurons is unknown. Here we show that nicotine can directly and robustly increase the firing frequency of VTA DAergic neurons for several minutes. In mouse midbrain slices, 300 nM nicotine elicited a persistent inward current in VTA DAergic neurons that was blocked by α-conotoxin MII[H9A;L15A], a selective antagonist of nAChRs containing the α6 subunit. α-conotoxin MII[H9A;L15A] also significantly reduced the long-lasting increase in DAergic neuronal activity produced by low concentrations of nicotine. In addition, nicotine failed to significantly activate VTA DAergic neurons in mice that did not express either α4 or α6 nAChR subunits. Conversely, selective activation of nAChRs containing the α4 subunit in knock-in mice expressing a hypersensitive version of these receptors yielded a biphasic response to nicotine consisting of an acute desensitizing increase in firing frequency followed by a sustained increase that lasted several minutes and was sensitive to α-conotoxin MII[H9A;L15A]. These data indicate that nicotine persistently activates VTA DAergic neurons via nAChRs containing α4 and α6 subunits.


Asunto(s)
Dopamina/metabolismo , Neuronas/efectos de los fármacos , Nicotina/farmacología , Receptores Nicotínicos/metabolismo , Área Tegmental Ventral/efectos de los fármacos , Animales , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Receptores Nicotínicos/química , Área Tegmental Ventral/citología , Área Tegmental Ventral/metabolismo
18.
PLoS One ; 6(9): e24813, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21980358

RESUMEN

Phosphorylation at murine Serine 18 (human Serine 15) is a critical regulatory process for the tumor suppressor function of p53. p53Ser18 residue is a substrate for ataxia-telangiectasia mutated (ATM) and ATM-related (ATR) protein kinases. Studies of mice with a germ-line mutation that replaces Ser18 with Ala (p53(S18A) mice) have demonstrated that loss of phosphorylation of p53Ser18 leads to the development of tumors, including lymphomas, fibrosarcomas, leukemia and leiomyosarcomas. The predominant lymphoma is B-cell lymphoma, which is in contrast to the lymphomas observed in Atm(-/-) animals. This observation and the fact that multiple kinases phosphorylate p53Ser18 suggest Atm-independent tumor suppressive functions of p53Ser18. Therefore, in order to examine p53Ser18 function in relationship to ATM, we analyzed the lifespan and tumorigenesis of mice with combined mutations in p53Ser18 and Atm. Surprisingly, we observed no cooperation in survival and tumorigenesis in compound p53(S18A) and Atm(-/-) animals. However, we observed embryonic lethality in the compound mutant animals. In addition, the homozygous p53Ser18 mutant allele impacted the weight of Atm(-/-) animals. These studies examine the genetic interaction of p53Ser18 and Atm in vivo. Furthermore, these studies demonstrate a role of p53Ser18 in regulating embryonic survival and motor coordination.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Regulación Neoplásica de la Expresión Génica , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/fisiología , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/fisiología , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proliferación Celular , Femenino , Fibroblastos/citología , Mutación de Línea Germinal , Humanos , Masculino , Ratones , Ratones Transgénicos , Modelos Genéticos , Mutación , Fosforilación
19.
PLoS One ; 6(9): e24132, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21931655

RESUMEN

BACKGROUND: Frontline treatment of small cell lung carcinoma (SCLC) relies heavily on chemotherapeutic agents and radiation therapy. Though SCLC patients respond well to initial cycles of chemotherapy, they eventually develop resistance. Identification of novel therapies against SCLC is therefore imperative. METHODS AND FINDINGS: We have designed a bioluminescence-based cell viability assay for high-throughput screening of anti-SCLC agents. The assay was first validated via standard pharmacological agents and RNA interference using two human SCLC cell lines. We then utilized the assay in a high-throughput screen using the LOPAC(1280) compound library. The screening identified several drugs that target classic cancer signaling pathways as well as neuroendocrine markers in SCLC. In particular, perturbation of dopaminergic and serotonergic signaling inhibits SCLC cell viability. CONCLUSIONS: The convergence of our pharmacological data with key SCLC pathway components reiterates the importance of neurotransmitter signaling in SCLC etiology and points to possible leads for drug development.


Asunto(s)
Antineoplásicos/farmacología , Mediciones Luminiscentes/métodos , Neurotransmisores/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cisplatino/farmacología , Dopaminérgicos/farmacología , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/genética , Células HEK293 , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serotoninérgicos/farmacología , Carcinoma Pulmonar de Células Pequeñas/genética , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Carcinoma Pulmonar de Células Pequeñas/patología , Estaurosporina/farmacología , Factores de Tiempo
20.
Biochem Pharmacol ; 82(8): 1015-21, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21640716

RESUMEN

Despite the known adverse health effects associated with tobacco use, over 45 million adults in the United States smoke. Cigarette smoking is the major etiologic factor associated with lung cancer. Cigarettes contain thousands of toxic chemicals, many of which are carcinogenic. Nicotine contributes directly to lung carcinogenesis through the activation of nicotinic acetylcholine receptors (nAChRs). nAChRs are ligand-gated ion channels, expressed in both normal and lung cancer cells, which mediate the proliferative, pro-survival, angiogenic, and metastatic effects of nicotine and its nitrosamine derivatives. The underlying molecular mechanisms involve increases in intracellular calcium levels and activation of cancer signal transduction pathways. In addition, acetylcholine (ACh) acts as an autocrine or paracrine growth factor in lung cancer. Other neurotransmitters and neuropeptides also activate similar growth loops. Recent genetic studies further support a role for nAChRs in the development of lung cancer. Several nAChR antagonists have been shown to inhibit lung cancer growth, suggesting that nAChRs may serve as valuable targets for biomarker-guided lung cancer interventions.


Asunto(s)
Neoplasias Pulmonares/metabolismo , Receptores Nicotínicos/metabolismo , Animales , Comunicación Autocrina , Calcio/metabolismo , Proliferación Celular/efectos de los fármacos , Humanos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/prevención & control , Nicotina/toxicidad , Antagonistas Nicotínicos/uso terapéutico , Receptores Nicotínicos/genética , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA