Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Science ; 373(6560): 1252-1256, 2021 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-34516792

RESUMEN

Compulsive drug use despite adverse consequences defines addiction. While mesolimbic dopamine signaling is sufficient to drive compulsion, psychostimulants such as cocaine also boost extracellular serotonin (5-HT) by inhibiting reuptake. We used SERT Met172 knockin (SertKI) mice carrying a transporter that no longer binds cocaine to abolish 5-HT transients during drug self-administration. SertKI mice showed an enhanced transition to compulsion. Conversely, pharmacologically elevating 5-HT reversed the inherently high rate of compulsion transition with optogenetic dopamine self-stimulation. The bidirectional effect on behavior is explained by presynaptic depression of orbitofrontal cortex­to­dorsal striatum synapses induced by 5-HT via 5-HT1B receptors. Consequently, in projection-specific 5-HT1B receptor knockout mice, the fraction of individuals compulsively self-administering cocaine was elevated.


Asunto(s)
Trastornos Relacionados con Cocaína/metabolismo , Receptor de Serotonina 5-HT1B/metabolismo , Serotonina/metabolismo , Transmisión Sináptica , Animales , Cocaína/administración & dosificación , Trastornos Relacionados con Cocaína/genética , Dopamina/metabolismo , Técnicas de Sustitución del Gen , Ratones , Ratones Noqueados , Optogenética , Receptor de Serotonina 5-HT1B/deficiencia , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo
2.
Cell Rep ; 23(13): 3852-3863, 2018 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-29949769

RESUMEN

The ventral pallidum (VP) is part of the basal ganglia circuitry and a target of both direct and indirect pathway projections from the nucleus accumbens. VP is important in cocaine reinforcement, and the firing of VP neurons is modulated in vivo during cocaine self-administration. This modulation of firing is thought to be indirect via cocaine actions on dopamine in the accumbens. Here, we show that cocaine directly inhibits synaptic transmission evoked by selective stimulation of indirect pathway projections to VP neurons. The inhibition is independent of dopamine receptor activation, absent in 5-HT1B knockout mice, and mimicked by a serotonin transporter (SERT) blocker. SERT-expressing neurons in dorsal raphe project to the VP. Optogenetic stimulation of these projections evokes serotonin transients and effectively inhibits GABAergic transmission to VP neurons. This study shows that cocaine increases endogenous serotonin in the VP to suppress synaptic transmission selectively from indirect pathway projections to VP neurons.


Asunto(s)
Prosencéfalo Basal/metabolismo , Cocaína/farmacología , Serotonina/metabolismo , Transmisión Sináptica/efectos de los fármacos , Animales , Núcleo Dorsal del Rafe/fisiología , Potenciales Evocados/efectos de los fármacos , Femenino , Luz , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Neuronas/metabolismo , Receptor de Serotonina 5-HT1B/deficiencia , Receptor de Serotonina 5-HT1B/genética , Receptores de Dopamina D2/metabolismo , Serotonina/farmacología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología
3.
Neuropsychopharmacology ; 42(6): 1182-1191, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27976680

RESUMEN

Impulsivity is an endophenotype found in many psychiatric disorders including substance use disorders, pathological gambling, and attention deficit hyperactivity disorder. Two behavioral features often considered in impulsive behavior are behavioral inhibition (impulsive action) and delayed gratification (impulsive choice). However, the extent to which these behavioral constructs represent distinct facets of behavior with discrete biological bases is unclear. To test the hypothesis that impulsive action and impulsive choice represent statistically independent behavioral constructs in mice, we collected behavioral measures of impulsivity in a single cohort of mice using well-validated operant behavioral paradigms. Mice with manipulation of serotonin 1B receptor (5-HT1BR) expression were included as a model of disordered impulsivity. A factor analysis was used to characterize correlations between the measures of impulsivity and to identify covariates. Using two approaches, we dissociated impulsive action from impulsive choice. First, the absence of 5-HT1BRs caused increased impulsive action, but not impulsive choice. Second, based on an exploratory factor analysis, a two-factor model described the data well, with measures of impulsive action and choice separating into two independent factors. A multiple-indicator multiple-causes analysis showed that 5-HT1BR expression and sex were significant covariates of impulsivity. Males displayed increased impulsivity in both dimensions, whereas 5-HT1BR expression was a predictor of increased impulsive action only. These data support the conclusion that impulsive action and impulsive choice are distinct behavioral phenotypes with dissociable biological influences that can be modeled in mice. Our work may help inform better classification, diagnosis, and treatment of psychiatric disorders, which present with disordered impulsivity.


Asunto(s)
Conducta Animal/fisiología , Descuento por Demora/fisiología , Conducta Impulsiva/fisiología , Inhibición Psicológica , Receptor de Serotonina 5-HT1B/fisiología , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones Transgénicos , Receptor de Serotonina 5-HT1B/deficiencia , Factores Sexuales
4.
Neuropsychopharmacology ; 41(12): 2941-2950, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27353308

RESUMEN

The effects of serotonin (5-HT) on anxiety and depression are mediated by a number of 5-HT receptors, including autoreceptors that act to inhibit 5-HT release. While the majority of anxiety and depression-related research has focused on the 5-HT1A receptor, the 5-HT1B receptor has a lesser known role in modulating emotional behavior. 5-HT1B receptors are inhibitory GPCRs located on the presynaptic terminal of both serotonin and non-serotonin neurons, where they act to inhibit neurotransmitter release. The autoreceptor population located on the axon terminals of 5-HT neurons is a difficult population to study due to their diffuse localization throughout the brain that overlaps with 5-HT1B heteroreceptors (receptors located on non-serotonergic neurons). In order to study the contribution of 5-HT1B autoreceptors to anxiety and depression-related behaviors, we developed a genetic mouse model that allows for selective ablation of 5-HT1B autoreceptors. Mice lacking 5-HT1B autoreceptors displayed the expected increases in extracellular serotonin levels in the ventral hippocampus following administration of a selective serotonin reuptake inhibitor. In behavioral studies, they displayed decreased anxiety-like behavior in the open field and antidepressant-like effects in the forced swim and sucrose preference tests. These results suggest that strategies aimed at blocking 5-HT1B autoreceptors may be useful for the treatment of anxiety and depression.


Asunto(s)
Ansiedad/metabolismo , Autorreceptores/metabolismo , Depresión/metabolismo , Hipocampo/metabolismo , Receptor de Serotonina 5-HT1B/deficiencia , Animales , Animales Recién Nacidos , Ansiedad/genética , Autorreceptores/genética , Depresión/genética , Modelos Animales de Enfermedad , Conducta Exploratoria/fisiología , Preferencias Alimentarias/efectos de los fármacos , Hipocampo/efectos de los fármacos , Isótopos de Yodo/farmacocinética , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Pindolol/análogos & derivados , Pindolol/farmacocinética , Receptor de Serotonina 5-HT1B/genética , Receptores de Serotonina 5-HT1/genética , Receptores de Serotonina 5-HT1/metabolismo , Antagonistas de la Serotonina/farmacocinética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo
5.
Neurochem Int ; 82: 52-68, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25684070

RESUMEN

The onset of attention-deficit-hyperactivity-disorder (ADHD) in childhood is characterized by developmentally inappropriate levels of hyperactivity, impulsivity and inattention. A chronic deficit of serotonin (5-HT) at the synapse may trigger symptoms of ADHD. This review focuses on neuro-anatomical, experimental and clinical pharmacological evidence, as well as the genetic underpinnings of serotoninergic involvement in the etiology of ADHD. Neuro-anatomical investigations suggest that serotonin through the orbitofrontal-striatal circuitry may regulate behavioral domains of hyperactivity and impulsivity in ADHD. Studies from animal models of ADHD indicate intimate interplay between 5-HT and dopaminergic neurotransmission. Selective serotonin re-uptake inhibitors, as also non-stimulant drugs acting on the 5-HT system are, however, clinically effective. They impart less severe side effects in patients with no risk of addiction. Oral administration of l-tryptophan, the amino acid precursor of 5-HT, significantly alleviates ADHD symptoms. Given the multifactorial nature of ADHD, candidate gene and genome-wide association studies have suggested that serotoninergic gene variants are associated with increased risk of ADHD with each locus individually exerting a modest effect on overall risk.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/genética , Serotonina/deficiencia , Adolescente , Animales , Animales Recién Nacidos , Trastorno por Déficit de Atención con Hiperactividad/tratamiento farmacológico , Trastorno por Déficit de Atención con Hiperactividad/fisiopatología , Estimulantes del Sistema Nervioso Central/uso terapéutico , Niño , Preescolar , Cuerpo Estriado/fisiopatología , Modelos Animales de Enfermedad , Dopamina/fisiología , Interacción Gen-Ambiente , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Inhibición Psicológica , Ratones Noqueados , Herencia Multifactorial , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Proteínas de Transporte de Neurotransmisores/deficiencia , Proteínas de Transporte de Neurotransmisores/genética , Oxidopamina/toxicidad , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiopatología , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptor de Serotonina 5-HT1B/deficiencia , Serotonina/fisiología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/uso terapéutico , Triptófano/deficiencia , Triptófano/uso terapéutico
6.
J Clin Invest ; 122(10): 3490-503, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22945629

RESUMEN

Serotonin is a critical regulator of bone mass, fulfilling different functions depending on its site of synthesis. Brain-derived serotonin promotes osteoblast proliferation, whereas duodenal-derived serotonin suppresses it. To understand the molecular mechanisms of duodenal-derived serotonin action on osteoblasts, we explored its transcriptional mediation in mice. We found that the transcription factor FOXO1 is a crucial determinant of the effects of duodenum-derived serotonin on bone formation We identified two key FOXO1 complexes in osteoblasts, one with the transcription factor cAMP-responsive element-binding protein 1 (CREB) and another with activating transcription factor 4 (ATF4). Under normal levels of circulating serotonin, the proliferative activity of FOXO1 was promoted by a balance between its interaction with CREB and ATF4. However, high circulating serotonin levels prevented the association of FOXO1 with CREB, resulting in suppressed osteoblast proliferation. These observations identify FOXO1 as the molecular node of an intricate transcriptional machinery that confers the signal of duodenal-derived serotonin to inhibit bone formation.


Asunto(s)
Remodelación Ósea/fisiología , Duodeno/metabolismo , Factores de Transcripción Forkhead/fisiología , Osteoblastos/fisiología , Serotonina/fisiología , Factor de Transcripción Activador 4/fisiología , Animales , Barrera Hematoencefálica , División Celular/efectos de los fármacos , Células Cultivadas/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/deficiencia , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica , Genes Reporteros , Genotipo , Homeostasis/fisiología , Factor I del Crecimiento Similar a la Insulina/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/deficiencia , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/fisiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Especificidad de Órganos , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteogénesis/fisiología , Regiones Promotoras Genéticas , Receptor de Serotonina 5-HT1B/deficiencia , Receptor de Serotonina 5-HT1B/genética , Receptor de Serotonina 5-HT1B/fisiología , Serotonina/sangre , Serotonina/farmacología , Estrés Fisiológico/genética , Transcripción Genética
7.
Eur J Pharmacol ; 667(1-3): 250-7, 2011 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-21723276

RESUMEN

The 5-HT(1B) receptor has been implicated in disorders such as depression, anxiety and obsessive-compulsive disorder. In mice lacking the 5-HT(1B) receptor (5-HT(1B) knockout mice), important changes in physiology and behavior exist. In the absence of presynaptic 5-HT(1B) receptor inhibition, chronic SSRI treatment may differentially affect 5-HT(1A) receptor functionality. The present studies tested the hypothesis that chronically reducing 5-HT transporter (5-HTT) function with selective serotonin reuptake inhibitor (SSRI) treatment would accelerate 5-HT(1A) receptor desensitization in 5-HT(1B) knockout mice. Moreover, as 5-HT(1B) knockout mice have been found to display exaggerated autonomic and locomotor responses to environmental stressors, the effects of chronic SSRI treatment on the hyperreactive phenotype of 5-HT(1B) knockout mice were investigated. The stress-reducing effect of the 5-HT(1A) receptor agonist flesinoxan on increases in body temperature, heart rate and locomotor activity was similar in wild type and 5-HT(1B) knockout mice before and after chronic 21-day treatment with the SSRI fluvoxamine, indicating no apparent alteration of 5-HT(1A) receptor sensitivity in 5-HT(1B) knockout mice. Also, chronic SSRI treatment did not alter the increased stress reactivity to mild environmental stressors in 5-HT(1B) knockout mice. We demonstrate that no apparent differences in 5-HT(1A) receptor sensitivity occur between 5-HT(1B) knockout and wild type mice after chronic fluvoxamine treatment. Also, the hyperreactive phenotype of 5-HT(1B) knockout mice is unresponsive to chronic SSRI treatment. Taken together, these results indicate that constitutive absence of 5-HT(1B) receptors does not result in adaptive changes in 5-HT(1A) receptor functionality and that chronic SSRI treatment does not modify stress reactivity in 5-HT(1B) knockout mice.


Asunto(s)
Fluvoxamina/farmacología , Técnicas de Inactivación de Genes , Receptor de Serotonina 5-HT1A/metabolismo , Receptor de Serotonina 5-HT1B/deficiencia , Receptor de Serotonina 5-HT1B/genética , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Animales , Temperatura Corporal/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Ratones , Actividad Motora/efectos de los fármacos , Receptor de Serotonina 5-HT1B/metabolismo , Estrés Fisiológico/efectos de los fármacos , Factores de Tiempo
8.
J Exp Med ; 208(1): 41-52, 2011 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-21187319

RESUMEN

Recent evidence indicates that leptin regulates appetite and energy expenditure, at least in part by inhibiting serotonin synthesis and release from brainstem neurons. To demonstrate that this pathway works postnatally, we used a conditional, brainstem-specific mouse CreER(T2) driver to show that leptin signals in brainstem neurons after birth to decrease appetite by inhibiting serotonin synthesis. Cell-specific gene deletion experiments and intracerebroventricular leptin infusions reveal that serotonin signals in arcuate nuclei of the hypothalamus through the Htr1a receptor to favor food intake and that this serotonin function requires the expression of Creb, which regulates the expression of several genes affecting appetite. Accordingly, a specific antagonist of the Htr1a receptor decreases food intake in leptin-deficient but not in Htr1a(-/-) mice. Collectively, these results establish that leptin inhibition of serotonin is necessary to inhibit appetite postnatally and provide a proof of principle that selective inhibition of this pathway may be a viable option to treat appetite disorders.


Asunto(s)
Regulación de la Expresión Génica , Leptina/metabolismo , Serotonina/metabolismo , Transcripción Genética , Animales , Ratones , Ratones Endogámicos C57BL , Análisis por Micromatrices , Receptor de Serotonina 5-HT1B/deficiencia , Receptor de Serotonina 5-HT1B/metabolismo , Transducción de Señal
9.
Physiol Behav ; 93(4-5): 659-65, 2008 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-18155098

RESUMEN

Serotonin (5-HT)(1A) and 5-HT(1B) receptors have been implicated in the incidence and treatment of depression in part through the examination of animals lacking these receptors. Although these receptors have been repeatedly implicated in ingestive behavior there is little information about how 5-HT(1A) and 5-HT(1B) receptor mutant mice react to solutions of varying palatability. In the present experiment male and female 5-HT(1A) and 5-HT(1B) mutant and wild-type mice were presented with increasing concentrations of sucrose using a two-bottle choice procedure. In addition fasting blood glucose levels were assessed. Both male and female 5-HT(1B) mutant mice drank more sucrose than WT mice but also consumed more water. Female, but not male, 5-HT(1A) mutant mice similarly showed increased sucrose consumption, but did not demonstrate increased consumption of water. In addition, the pattern of increased sucrose consumption over genotype and sex was related to fasting blood glucose concentrations such that levels in male 5-HT(1B) mutant mice were reduced relative to wild-type and 5-HT(1A) mutant males, but similar to those of females. The findings in 5-HT(1B) mutant mice emphasize the role of the 5-HT(1B) receptor in regulating ingestive behavior, whereas female sex hormones and 5-HT(1A) receptors may interact to alter sucrose consumption in 5-HT(1A) mutant mice. In addition, these findings may have implications for the role of these receptors in the incidence and treatment of depression since the intake of sucrose has been used as an index of anhedonia in animal models of depression and antidepressant efficacy.


Asunto(s)
Glucemia/fisiología , Conducta de Ingestión de Líquido/fisiología , Ayuno/sangre , Receptor de Serotonina 5-HT1A/deficiencia , Receptor de Serotonina 5-HT1B/deficiencia , Sacarosa , Análisis de Varianza , Animales , Conducta Animal , Peso Corporal/genética , Conducta de Elección/fisiología , Ingestión de Líquidos/genética , Femenino , Preferencias Alimentarias/fisiología , Masculino , Ratones , Ratones Noqueados , Factores Sexuales , Sacarosa/administración & dosificación
10.
Mol Psychiatry ; 12(11): 1042-56, 975, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17420766

RESUMEN

Normal aging of the brain differs from pathological conditions and is associated with increased risk for psychiatric and neurological disorders. In addition to its role in the etiology and treatment of mood disorders, altered serotonin (5-HT) signaling is considered a contributing factor to aging; however, no causative role has been identified in aging. We hypothesized that a deregulation of the 5-HT system would reveal its contribution to age-related processes and investigated behavioral and molecular changes throughout adult life in mice lacking the regulatory presynaptic 5-HT(1B) receptor (5-HT(1B)R), a candidate gene for 5-HT-mediated age-related functions. We show that the lack of 5-HT(1B)R (Htr1b(KO) mice) induced an early age-related motor decline and resulted in decreased longevity. Analysis of life-long transcriptome changes revealed an early and global shift of the gene expression signature of aging in the brain of Htr1b(KO) mice. Moreover, molecular changes reached an apparent maximum effect at 18-months in Htr1b(KO) mice, corresponding to the onset of early death in that group. A comparative analysis with our previous characterization of aging in the human brain revealed a phylogenetic conservation of age-effect from mice to humans, and confirmed the early onset of molecular aging in Htr1b(KO) mice. Potential mechanisms appear independent of known central mechanisms (Bdnf, inflammation), but may include interactions with previously identified age-related systems (IGF-1, sirtuins). In summary, our findings suggest that the onset of age-related events can be influenced by altered 5-HT function, thus identifying 5-HT as a modulator of brain aging, and suggesting age-related consequences to chronic manipulation of 5-HT.


Asunto(s)
Envejecimiento/fisiología , Regulación de la Expresión Génica/genética , Expresión Génica/genética , Actividad Motora/genética , Receptor de Serotonina 5-HT1B/deficiencia , Factores de Edad , Análisis de Varianza , Animales , Animales Recién Nacidos , Conducta Animal/fisiología , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Ensayo de Inmunoadsorción Enzimática/métodos , Fuerza de la Mano/fisiología , Hibridación in Situ , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Noqueados , Análisis por Micromatrices/métodos , Tiempo de Reacción/fisiología , Receptor de Serotonina 5-HT1B/genética , Análisis de Supervivencia
11.
Brain Res Bull ; 70(1): 81-93, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16750486

RESUMEN

Lesioning of serotonergic afferents increases hippocampal ACh release and attenuates memory deficits produced by cholinergic lesions. Improved memory performance described in 5-HT1B-knockout (KO) mice might thus be due to a weaker 5-HT1B-mediated inhibitory influence of 5-HT on hippocampal ACh release. The selective delay-dependent impairment of working memory observed in these KO mice suggests, however, that cortical regions also participate in task performance, possibly via indirect influences of 5-HT on ACh release. To provide neuropharmacological support for these hypotheses we measured evoked ACh and 5-HT release in hippocampal and cortical slices of wild-type (WT) and 5-HT1B KO mice. Superfused slices (preincubated with [3H]choline or [3H]5-HT) were electrically stimulated in the absence or presence of 5-HT1B receptor ligands. In hippocampus and cortex, 5-HT1B agonists decreased and antagonists increased 5-HT release in WT, but not in 5-HT1B KO mice. In 5-HT1B KO mice, 5-HT release was enhanced in both structures, while ACh release (in nCi) was reduced. ACh release was inhibited by 5-HT1B agonists in hippocampal (not cortical) slices of WT but not of 5-HT1B KO mice. Our data (i) confirm the absence of autoinhibition of 5-HT release in 5-HT1B-KO mice, (ii) demonstrate a reduced release of ACh, and the absence of 5-HT1B-receptor-mediated inhibition of ACh release, in the hippocampus and cortex of 5-HT1B-KO mice, and (iii) are compatible with an indirect role of cortical ACh in the working memory impairment observed in these KO mice.


Asunto(s)
Acetilcolina/metabolismo , Corteza Cerebral/citología , Hipocampo/citología , Terminales Presinápticos/metabolismo , Receptor de Serotonina 5-HT1B/deficiencia , Serotonina/metabolismo , Análisis de Varianza , Animales , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/efectos de la radiación , Colina/metabolismo , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica/métodos , Hipocampo/efectos de los fármacos , Hipocampo/efectos de la radiación , Masculino , Ratones , Ratones Noqueados , Terminales Presinápticos/efectos de los fármacos , Terminales Presinápticos/efectos de la radiación , Piridinas/farmacología , Pirroles/farmacología , Quipazina/análogos & derivados , Quipazina/farmacología , Antagonistas de la Serotonina/farmacología , Agonistas de Receptores de Serotonina/farmacología , Tritio/metabolismo
12.
Neurosci Lett ; 401(3): 209-13, 2006 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-16581182

RESUMEN

The hypothalamic suprachiasmatic nucleus (SCN) is a circadian oscillator that receives a dense serotonergic innervation from the median raphe nucleus. Serotonin (5-HT) modulates the effects of light on circadian behavior by acting on 5-HT1B receptors on retinohypothalamic (RHT) terminals in the SCN. Activation of 5-HT1B presynaptic receptors on RHT terminals inhibits glutamate release. However, 5-HT1B receptor knockout (5-HT1B KO) mice have attenuated behavioral responses to light [P.J. Sollars, M.D. Ogilvie, A.M. Simpson, G.E. Pickard, Photic entrainment is altered in the 5-HT1B receptor knockout mouse, J. Biol. Rhythms 21 (2006) 21-32]. To assess the cellular response of the 5-HT1B KO SCN to light, light-induced Fos expression was analyzed in 5-HT1B KO and wild-type (WT) mice. In addition, the distribution of melanopsin containing retinal ganglion cells that contribute the majority of axons to the RHT was examined in 5-HT1B KO mice and compared to that of WT mice. Light-induced Fos expression in the SCN was reduced in 5-HT1B KO mice compared to WT mice at circadian time (CT) 16 and CT 23 in a manner similar to the reduction previously described in light-induced behavioral phase shifts. The number of melanopsin retinal ganglion cells was similar in WT and 5-HT1B KO mice. These data taken together with previous data suggest that functional removal of the 5-HT1B receptor results in reduced functional light input to the SCN.


Asunto(s)
Ritmo Circadiano/fisiología , Expresión Génica/fisiología , Luz , Proteínas Oncogénicas v-fos/metabolismo , Receptor de Serotonina 5-HT1B/deficiencia , Núcleo Supraquiasmático/metabolismo , Animales , Expresión Génica/efectos de la radiación , Inmunohistoquímica/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Núcleo Supraquiasmático/efectos de la radiación
13.
Brain Res ; 1062(1-2): 26-31, 2005 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-16256085

RESUMEN

Serotonin (5-HT) is considered to play an important role in control of appetite. Enterostatin has been shown to alter 5-HT release in the brain, and non-specific 5-HT antagonists blocked the anorectic response to icv enterostatin. The aim of this study was to further identify which 5-HT receptor subtype mediates the enterostatin feeding behavior and whether this effect occurs due to action in the PVN. Wild-type and 5-HT2C receptor-/- (KO) mice and normal Sprague-Dawley rats were used in these experiments. All animals were fed a high fat diet. Enterostatin (120 nmol, i.p.) reduced the intake of high fat diet in 5-HT2C receptor mutant mice (saline 4.54 +/- 0.47 kcal vs. Ent 2.53 +/- 0.76 kcal) 1 h after injection. A selective 5-HT1B antagonist (GR55526, 40 mg/kg body weight, i.p.) blocked the enterostatin hypophagic effects in these KO mice. Rats were implanted with cannulas into the amygdala and the ipsilateral PVN. The 5-HT receptor antagonists metergoline (non-specific receptor subtypes 1 and 2), or ritanserin (selective 2C), or GR55562 (selective l B) was injected into the PVN prior to enterostatin (0.01 nmol) injection into the amygdala. Enterostatin reduced food intake (saline: 5.80 +/- 0.59 g vs. enterostatin 3.47 +/- 0.56 g, P < 0.05 at l h). Pretreatment with either metergoline (10 nmol) or GR55526 (10 nmol) but not ritanserin (10 nmol) into the PVN attenuated the anorectic response to amygdala enterostatin. The data imply that the enterostatin anorectic response may be modulated by 5-HT1B receptors and that a neuronal pathway from the amygdala to the PVN regulates the enterostatin response through activation of 5-HTlB receptors in PVN.


Asunto(s)
Regulación del Apetito/fisiología , Colipasas/fisiología , Conducta Alimentaria/fisiología , Núcleo Hipotalámico Paraventricular/fisiología , Precursores de Proteínas/fisiología , Receptor de Serotonina 5-HT1B/fisiología , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/fisiología , Animales , Regulación del Apetito/efectos de los fármacos , Grasas de la Dieta , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Precursores Enzimáticos , Conducta Alimentaria/efectos de los fármacos , Femenino , Masculino , Ratones , Ratones Noqueados , Microinyecciones , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/fisiología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptor de Serotonina 5-HT1B/deficiencia , Receptor de Serotonina 5-HT1B/efectos de los fármacos , Serotonina/fisiología , Antagonistas de la Serotonina/administración & dosificación
14.
J Neurophysiol ; 93(6): 3157-64, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15716370

RESUMEN

The suprachiasmatic nucleus (SCN) receives a dense serotonergic innervation that modulates photic input to the SCN via serotonin 1B (5-HT1B) presynaptic receptors on retinal glutamatergic terminals. However, the majority of 5-HT1B binding sites in the SCN are located on nonretinal terminals and most axonal terminals in the SCN are GABAergic. We therefore tested the hypothesis that 5-HT1B receptors might also be located on SCN GABAergic terminals by examining the effects of the highly selective 5-HT1B receptor agonist CP-93,129 on SCN miniature inhibitory postsynaptic currents (mIPSCs). Whole cell patch-clamp recordings of mIPSCs were obtained from rat and mouse SCN neurons in hypothalamic slices. Using CsCl-containing microelectrodes with QX314, we isolated mPSCs that were sensitive to the GABAA receptor antagonist, bicuculline. Bath application of CP-93,129 (1 microM) decreased the frequency of mIPSCs by an average of 22% (n = 7) in rat SCN neurons and by an average of 30% (n = 8) in mouse SCN neurons with no clear effect on mIPSC amplitude. In mice lacking functional 5-HT1B receptors, CP-93,129 (1 microM) had no clear effect on the frequency or the amplitude of mIPSCs recorded in any of the cells tested (n = 4). The decrease in the frequency of mIPSCs of SCN neurons produced by the selective 5-HT1B receptor agonist CP-93,129 is consistent with the interpretation that 5-HT1B receptors are located on GABA terminals in the SCN and that 5-HT inhibits GABA release via a 5-HT1B presynaptic receptor-mediated mechanism.


Asunto(s)
Terminales Presinápticos/metabolismo , Receptor de Serotonina 5-HT1B/metabolismo , Núcleo Supraquiasmático/metabolismo , Valina/análogos & derivados , Ácido gamma-Aminobutírico/metabolismo , Animales , Bicuculina/farmacología , Interacciones Farmacológicas , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Antagonistas del GABA/farmacología , Técnicas In Vitro , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Ratones Noqueados , Inhibición Neural/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Técnicas de Placa-Clamp/métodos , Piridinas/farmacología , Pirroles/farmacología , Quinoxalinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptor de Serotonina 5-HT1B/deficiencia , Agonistas de Receptores de Serotonina/farmacología , Núcleo Supraquiasmático/citología , Tetrodotoxina/farmacología , Factores de Tiempo , Valina/farmacología
15.
Therapie ; 60(5): 469-76, 2005.
Artículo en Francés | MEDLINE | ID: mdl-16433012

RESUMEN

The main hypothesis regarding the mechanism of action of antidepressant drugs is monoaminergic and mainly involves two neurotransmitters, serotonin and noradrenaline. Despite the well-recognized therapeutic efficacy of selective serotonin reuptake inhibitors (SSRIs), some disadvantages still occur. For example, they often require 4-6 weeks to achieve clinical benefits in depressed patients. In the past, some molecules that could shorten this long delay of action have been identified. The role of presynaptic autoreceptors - the activation of which leads to an inhibitory feedback control on neurotransmitter synthesis and release - has been extensively studied for antidepressant effects. In our laboratory, we studied the combined effects of an SSRI and a serotonin autoreceptor antagonist of the 5-HT1B subtype using intracerebral in vivo microdialysis in awake, freely moving mice. Important information on SSRIs has been obtained by applying this technique to genetically modified animals, such as constitutive knockout (KO) mice lacking 5-HT1B receptors (5-HT1B KO) generated by homologous recombination: we compared the effects of a combined treatment on extracellular/intrasynaptic levels of serotonin in various nerve terminals area in wild-type control and KO mice. Thus, we found that indirect activation of 5-HT1B autoreceptors limits the effects of SSRIs on dialysate 5-HT levels at serotonergic nerve terminals such as the ventral hippocampus. The study of substance P (neurokinin 1 receptor [R-NK1]) offers another example of the use of KO mice in the development of a new class of antidepressant drugs. NK1 receptor antagonists may display anxiolytic/antidepressant-like properties. The lack of selective compounds for each tachykinin receptor subtype (R-NK 1, R-NK2 or R-NK3) and differences in their affinity between animal species have made R-NK1 KO mice a very useful experimental tool. In collaborative work we found that genetic (R-NK1 KO mice) or pharmacological (GR205171) blockade of R-NK1 is associated with several changes: the increase in cortical 5-HT outflow caused by systemic injection of paroxetine was 4- to 6-fold higher in freely moving R-NK1 KO mice than in wild-type controls. The constitutive lack of NK1 receptors is associated with a functional desensitization of somatodendritic 5-HT1A autoreceptors, resembling that induced by chronic treatment with SSRI antidepressants. These results highlight the link between a neurotransmitter (serotonin) and a neuropeptide (substance P). This genetic strategy allowed us to point out that multiple targets participate to the effects of classical antidepressant drugs within the brain. We hope that, soon, some mice lines (constitutive or tissue specific, conditional rescue mice having alterations of sleep/wakefulness and/or food intake, altered central serotonin and/or noradrenaline neurotransmission, deficit in neurotrophic factors, but increases in intrasynaptic concentrations of substance P) could be a relevant model of the physiopathology of depressive disorders, and could help us understand the appearance of some symptoms. These recent findings suggest that instead of being rejected, the monoaminergic hypothesis of depression should be improved, corrected and completed by studying the role of other neurotransmitter, neuromodulatory compounds (substance P, BDNF [brain-derived neurotrophic factor]). By doing so, it thus could be possible to improve antidepressant drug treatment, i.e. shorten their long delay of action and/or to decrease treatment resistance or improve its tolerance.


Asunto(s)
Antidepresivos/uso terapéutico , Paroxetina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Animales , Antidepresivos/farmacología , Ratones , Ratones Noqueados , Receptor de Serotonina 5-HT1B/clasificación , Receptor de Serotonina 5-HT1B/deficiencia , Receptor de Serotonina 5-HT1B/genética , Receptor de Serotonina 5-HT1B/fisiología , Serotonina/fisiología
16.
Int J Dev Neurosci ; 22(5-6): 405-13, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15380839

RESUMEN

Serotonin is an important modulator of anxiety and thus drugs that act on this system have frequently been shown to be either anxiogenic or anxiolytic. In addition serotonin has important trophic functions during early development and disruption of serotonin homeostasis is likely to have long-lasting repercussions in the adult. In the present study we examined the contribution of two serotonin receptor subtypes (5HT(1A) and 5HT(1B)) to the pathophysiology of anxiety during development. For this, we have studied homozygous knockout mice lacking the 5HT(1B) receptor and examined the effect of pharmacological manipulations of 5HT(1A) and 5HT(1B) receptors on locomotor activity and emission of ultrasonic vocalization (USV) in 7-8 days old mice. As shown before, drug naïve 5HT(1B) knockout pups showed reduced USV and were hyperactive, in comparison to wild type controls. The administration of RU24969 (a 5HT(1A/1B) agonist) showed a dose-dependent decrease in USV in the wild type and a biphasic effect in the mutants and resulted in dose-dependent increase in activity in the wild type and, to a lesser extent, in the knockouts. The selective 5HT(1A) agonist, 8OH-DPAT, dose-dependently blocked vocalization in both genotypes and also increased locomotion. To differentially activate 5HT(1B) receptors we first blocked 5HT(1A) receptors with WAY100315 and then treated with RU24969. At a high testing temperature, pretreatment with WAY100315 resulted in an anxiogenic effect in wild type pups but not in the knockouts. In agreement with our findings that 5HT(1B) knockout mice were in general less sensitive to 5HT(1A) activation, 5HT(1A) receptor binding was reduced in the knockouts in comparison to controls. Finally, treatment with diazepam dose-dependently decreased USVs in both group with the knockouts showing enhanced sensitivity to this drug. Our results show that important adaptations to a disturbance of serotonin homeostasis occur during the first week of life within the serotonergic system. The observed decreased in sensitivity of 5HT(1B) knockout mice to 5HT(1A) and increased to GABA(A) manipulations are discussed within the context of serotonergic plasticity during development and the implication for clinical treatment of anxiety in genetically predisposed individuals.


Asunto(s)
Conducta Animal/fisiología , Receptor de Serotonina 5-HT1A/metabolismo , Receptor de Serotonina 5-HT1B/deficiencia , Agonistas del Receptor de Serotonina 5-HT1 , Agonistas de Receptores de Serotonina/farmacología , Vocalización Animal/fisiología , Animales , Animales Recién Nacidos , Conducta Animal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ratones , Vocalización Animal/efectos de los fármacos
17.
Psychopharmacology (Berl) ; 176(1): 39-49, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15138762

RESUMEN

RATIONALE: The possible role of compensatory changes in 5-HT2C receptors in the reduced hypophagic action of d-fenfluramine in 5-HT1B knockout (KO) mice was assessed by comparing their response to d-fenfluramine and the 5-HT2C receptor agonist mCPP. In addition we measured 5-HT(2C/A) receptor binding in 5-HT1B KO and wild-type (WT) mice and examined the effects of 5-HT1B receptor antagonists on d-fenfluramine-induced hypophagia in WT mice. METHODS: Hypophagic responses to d-fenfluramine (1-30 mg/kg) and mCPP (1-5.6 mg/kg) were measured using a behavioural satiety sequence paradigm. The effects of the 5-HT1B receptor antagonists GR 127,935 and SB 224289 in opposing the hypophagic action of d-fenfluramine were evaluated in WT mice. The binding of [3H]-mesulergine was compared in the brains of both mouse strains. RESULTS: The hypophagic effects of moderate doses of d-fenfluramine and mCPP were attenuated in 5-HT1B KO mice. Pretreatment of WT mice with the 5-HT(1B/1D) receptor antagonist GR 127,935, or food-deprived WT mice with the 5-HT1B receptor antagonist SB 224289, did not reproduce the reduction in sensitivity to the effects of d-fenfluramine on feeding behaviour observed in 5-HT1B KO mice. Estimates of 5-HT2C receptor binding were similar in 5-HT1B KO and WT mice. CONCLUSIONS: The hypophagic effect of d-fenfluramine in mice is unlikely to be mediated by the 5-HT1B receptor. Instead, the evidence suggests that an adaptive change in 5-HT2C receptor function occurs in 5-HT1B receptor KO mice and contributes to their reduced response to d-fenfluramine.


Asunto(s)
Fenfluramina/farmacocinética , Ratones Noqueados/genética , Piperazinas/farmacocinética , Receptor de Serotonina 5-HT1B/deficiencia , Receptor de Serotonina 5-HT1B/genética , Saciedad/efectos de los fármacos , Agonistas del Receptor de Serotonina 5-HT2 , Animales , Sitios de Unión/efectos de los fármacos , Sitios de Unión/fisiología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/ultraestructura , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Evaluación Preclínica de Medicamentos/métodos , Ingestión de Alimentos/efectos de los fármacos , Ergolinas/administración & dosificación , Ergolinas/farmacocinética , Fenfluramina/administración & dosificación , Genotipo , Inyecciones Intraperitoneales , Inyecciones Subcutáneas , Isomerismo , Mianserina/farmacología , Ratones , Piperazinas/administración & dosificación , Piperidonas/administración & dosificación , Piperidonas/farmacocinética , Receptor de Serotonina 5-HT2A , Receptor de Serotonina 5-HT2C/administración & dosificación , Saciedad/fisiología , Respuesta de Saciedad/efectos de los fármacos , Respuesta de Saciedad/fisiología , Especificidad de la Especie , Espiperona/farmacología , Compuestos de Espiro/administración & dosificación , Compuestos de Espiro/farmacocinética , Factores de Tiempo , Tritio , Reino Unido
18.
Neurosci Lett ; 354(2): 169-71, 2004 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-14698465

RESUMEN

Prior behavioral history in operant conditioning paradigms may induce impulsive-like responding as shown in rats. Little is known to what extent behavioral history influences subsequent behavior in mice, therefore the present study investigated the effects of lever-pressing under a fixed-ratio 5 schedule of reinforcement on subsequent differential-reinforcement-of-low-rate (DRL) 36 s performance in wild type mice compared to the behavior of 5-HT1B receptor knockout mice. Acquisition of both autoshaping and fixed-ratio 5 training was faster in 5-HT1B receptor knockout compared to wild type mice. Nevertheless, in the DRL 36 s procedure no differences were observed between genotypes. Both wild type and 5-HT1B receptor knockout mice displayed premature or impulsive-like responding in the DRL 36 s procedure, for example a peak location of responses around 20 s and high rates of responding. Taken together, the present data suggest that impulsive-like responding in the DRL 36 s procedure in mice depends on prior behavioral history.


Asunto(s)
Condicionamiento Operante/fisiología , Conducta Impulsiva/genética , Refuerzo en Psicología , Animales , Conducta Animal/fisiología , Química Encefálica/genética , Genotipo , Conducta Impulsiva/metabolismo , Conducta Impulsiva/psicología , Masculino , Ratones , Ratones Noqueados , Tiempo de Reacción/genética , Tiempo de Reacción/fisiología , Receptor de Serotonina 5-HT1B/deficiencia , Receptor de Serotonina 5-HT1B/genética , Serotonina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...