Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 13(1): 7708, 2022 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-36550097

RESUMEN

Appropriate processing of reward and aversive information is essential for survival. Although a critical role of serotonergic neurons in the dorsal raphe nucleus (DRN) in reward processing has been shown, the lack of rewarding effects with selective serotonin reuptake inhibitors (SSRIs) implies the presence of a discrete serotonergic system playing an opposite role to the DRN in the processing of reward and aversive stimuli. Here, we demonstrated that serotonergic neurons in the median raphe nucleus (MRN) of mice process reward and aversive information in opposite directions to DRN serotonergic neurons. We further identified MRN serotonergic neurons, including those projecting to the interpeduncular nucleus (5-HTMRN→IPN), as a key mediator of reward and aversive stimuli. Moreover, 5-HT receptors, including 5-HT2A receptors in the interpeduncular nucleus, are involved in the aversive properties of MRN serotonergic neural activity. Our findings revealed an essential function of MRN serotonergic neurons, including 5-HTMRN→IPN, in the processing of reward and aversive stimuli.


Asunto(s)
Núcleo Interpeduncular , Neuronas Serotoninérgicas , Ratones , Animales , Serotonina/fisiología , Núcleo Dorsal del Rafe/fisiología , Receptores de Serotonina
2.
Sci Rep ; 12(1): 6014, 2022 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-35399112

RESUMEN

Major psychiatric disorders such as attention-deficit/hyperactivity disorder and schizophrenia are often accompanied by elevated impulsivity. However, anti-impulsive drug treatments are still limited. To explore a novel molecular target, we examined the role of dopamine D5 receptors in impulse control using mice that completely lack D5 receptors (D5KO mice). We also measured spontaneous activity and learning/memory ability because these deficits could confound the assessment of impulsivity. We found small but significant effects of D5 receptor knockout on home cage activity only at specific times of the day. In addition, an analysis using the q-learning model revealed that D5KO mice displayed lower behavioral adjustment after impulsive actions. However, our results also showed that baseline impulsive actions and the effects of an anti-impulsive drug in D5KO mice were comparable to those in wild-type littermates. Moreover, unlike previous studies that used other D5 receptor-deficient mouse lines, we did not observe reductions in locomotor activity, working memory deficits, or severe learning deficits in our line of D5KO mice. These findings demonstrate that D5 receptors are dispensable for impulse control. Our results also indicate that time series analysis and detailed analysis of the learning process are necessary to clarify the behavioral functions of D5 receptors.


Asunto(s)
Dopamina , Receptores de Dopamina D5 , Animales , Humanos , Locomoción , Ratones , Ratones Noqueados , Receptores de Dopamina D1 , Receptores de Dopamina D5/fisiología
3.
Transl Psychiatry ; 12(1): 58, 2022 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-35145065

RESUMEN

Previous findings have proposed that drugs targeting 5-HT2C receptors could be promising candidates in the treatment of trauma- and stress-related disorders. However, the reduction of conditioned freezing observed in 5-HT2C receptor knock-out (KO) mice in previous studies could alternatively be accounted for by increased locomotor activity. To neutralize the confound of individual differences in locomotor activity, we measured a ratio of fear responses during versus before the presentation of a conditioned stimulus previously paired with a footshock (as a fear measure) by utilizing a conditioned licking suppression paradigm. We first confirmed that 5-HT2C receptor gene KO attenuated fear responses to distinct types of single conditioned stimuli (context or tone) independently of locomotor activity. We then assessed the effects of 5-HT2C receptor gene KO on compound fear responses by examining mice that were jointly conditioned to a context and a tone and later re-exposed separately to each. We found that separate re-exposure to individual components of a complex fear memory (i.e., context and tone) failed to elicit contextual fear extinction in both 5-HT2C receptor gene KO and wild-type mice, and also abolished differences between genotypes in tone-cued fear extinction. This study delineates a previously overlooked role of 5-HT2C receptors in conditioned fear responses, and invites caution in the future assessment of molecular targets and candidate therapies for the treatment of PTSD.


Asunto(s)
Señales (Psicología) , Miedo , Receptor de Serotonina 5-HT2C/metabolismo , Animales , Extinción Psicológica/fisiología , Miedo/fisiología , Ratones , Ratones Noqueados , Receptor de Serotonina 5-HT2C/genética , Serotonina
4.
Biochem Biophys Res Commun ; 562: 62-68, 2021 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-34038754

RESUMEN

In chronic smokers, nicotine withdrawal symptoms during tobacco cessation can lead to smoking relapse. In rodent models, chronic exposure to nicotine elicited physical dependence, whereas acute antagonism of nicotinic acetylcholine receptors (nAChRs) immediately precipitated withdrawal symptoms. Although the central serotonergic system plays an important role in nicotine withdrawal, the exact serotonergic raphe nuclei regulating these symptoms remain unknown. We used transgenic mice expressing archaerhodopsinTP009 or channelrhodopsin-2[C128S] exclusively in the central serotonergic neurons to selectively manipulate serotonergic neurons in each raphe nucleus. Nicotine withdrawal symptoms were precipitated by an acute injection of mecamylamine, a nonspecific nAChR antagonist, following chronic nicotine consumption. Somatic signs were used as measures of nicotine withdrawal symptoms. Acute mecamylamine administration significantly increased ptosis occurrence in nicotine-drinking mice compared with that in control-drinking mice. Optogenetic inhibition of the serotonergic neurons in the median raphe nucleus (MRN), but not of those in the dorsal raphe nucleus (DRN), mimicked the symptoms observed during mecamylamine-precipitated nicotine withdrawal even in nicotine-naïve mice following the administration of acute mecamylamine injection. Optogenetic activation of the serotonergic neurons in the MRN nearly abolished the occurrence of ptosis in nicotine-drinking mice. The serotonergic neurons in the MRN, but not those in the DRN, are necessary for the occurrence of somatic signs, a nicotine withdrawal symptom, and the activation of these neurons may act as a potential therapeutic strategy for preventing the somatic manifestations of nicotine withdrawal.


Asunto(s)
Nicotina/efectos adversos , Núcleos del Rafe/patología , Neuronas Serotoninérgicas/patología , Síndrome de Abstinencia a Sustancias/patología , Animales , Femenino , Masculino , Mecamilamina , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Optogenética , Receptores Nicotínicos/metabolismo , Serotonina/metabolismo
5.
Curr Biol ; 31(11): 2446-2454.e5, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-33838101

RESUMEN

Adapting to changing environmental conditions requires a prospective inference of future actions and their consequences, a strategy also known as model-based decision making.1-3 In stable environments, extensive experience of actions and their consequences leads to a shift from a model-based to a model-free strategy, whereby behavioral selection is primarily governed by retrospective experiences of positive and negative outcomes. Human and animal studies, where subjects are required to speculate about implicit information and adjust behavioral responses over multiple sessions, point to a role for the central serotonergic system in model-based decision making.4-8 However, to directly test a causal relationship between serotonergic activity and model-based decision making, phase-specific manipulation of serotonergic activity is needed in a one-shot test, where learning by trial and error is neutralized. Moreover, the serotonergic origin responsible for this effect is yet to be determined. Herein, we demonstrate that optogenetic silencing of serotonin neurons in the dorsal raphe nucleus, but not in the median raphe nucleus, disrupts model-based decision making in lithium-induced outcome devaluation tasks.9-11 Our data indicate that the serotonergic behavioral effects are not due to increased locomotor activity, anxiolytic effects, or working memory deficits. Our findings provide insights into the neural mechanisms underlying neural weighting between model-free and model-based strategies.


Asunto(s)
Núcleo Dorsal del Rafe , Serotonina , Animales , Toma de Decisiones , Humanos , Neuronas , Estudios Prospectivos , Estudios Retrospectivos , Neuronas Serotoninérgicas
6.
Am J Pathol ; 191(1): 144-156, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33339546

RESUMEN

Alzheimer disease (AD) is a progressive neurodegenerative disorder and the most common type of dementia worldwide. There is considerable evidence of age-related disruption of proteostasis being responsible for the development of AD. The proteasome is a multicatalytic enzyme complex that degrades both normal and damaged proteins, and an age-related decline in its activity has been implicated in age-related pathologies. Although proteasomal dysfunction is assumed to be a key AD hallmark, it remains unclear whether its role in disease onset is causative or secondary. In this study, we demonstrate that mice with proteasomal dysfunction exhibited memory impairment with associated neuronal loss, accumulation of phosphorylated tau, and activation of endoplasmic reticulum (ER) stress-related apoptosis pathways. Impaired proteasomal activity also activated ER stress-related apoptosis pathways in HT-22, a murine hippocampal neuronal cell line. HT-22 cell death, caused by proteasomal inhibition, was prevented by an inhibitor of c-Jun N-terminal kinase, an ER stress-related molecule. Collective evidence suggests that impaired proteasomal activity alters proteostasis, and subsequent ER stress-mediated pathways play pivotal roles in neuronal loss. Because aging decreases proteasomal function, age-related impairment of proteasomes may be involved in the development and progression of AD in elderly patients.


Asunto(s)
Encéfalo/patología , Estrés del Retículo Endoplásmico/fisiología , Trastornos de la Memoria/patología , Neuronas/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Apoptosis/fisiología , Encéfalo/metabolismo , Trastornos de la Memoria/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Proteínas tau/metabolismo
7.
Brain Res ; 1746: 146980, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32544501

RESUMEN

Juvenile stress, like that caused by childhood maltreatment, is a significant risk factor for psychiatric disorders such as depression later in life. Recently, the antidepressant effect of ketamine, a noncompetitive N-methyl-d-aspartate receptor antagonist, has been widely investigated. However, little is known regarding its efficacy against depressive-like alterations caused by juvenile stress, which is clinically relevant in human depression. In the present study, we evaluated the antidepressant-like effect of ketamine in adult rats that had been subjected to juvenile stress. Depressive-like behavior was assessed using the forced swim test (FST), and electrophysiological and morphological alterations in the layer V pyramidal cells of the prelimbic cortex were examined using whole-cell patch-clamp recordings and subsequent recording-cell specific fluorescence imaging. We demonstrated that ketamine (10 mg/kg) attenuated the increased immobility time caused by juvenile stress in the FST, restored the diminished excitatory postsynaptic currents, and caused atrophic changes in the apical dendritic spines. Ketamine's effects reversing impaired excitatory/inhibitory ratio of postsynaptic currents were also revealed. These results indicated that ketamine could be effective in reversing the depression-like alterations caused by juvenile stress.


Asunto(s)
Antidepresivos/farmacología , Corteza Cerebral/efectos de los fármacos , Depresión/etiología , Ketamina/farmacología , Estrés Psicológico/complicaciones , Animales , Corteza Cerebral/patología , Masculino , Células Piramidales/efectos de los fármacos , Ratas
8.
Int J Urol ; 27(8): 684-689, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32533581

RESUMEN

OBJECTIVES: To identify the types of serotonin (5-hydroxytryptamine) receptors of the prefrontal cortex related to the micturition reflex. METHODS: Female Sprague-Dawley rats and a microinjection method were used for this study. Stainless steel guide cannulas were implanted bilaterally into the prefrontal cortex, and a polyethylene catheter was inserted into the bladder. Cystometric parameters (intercontraction interval and maximum voiding pressure) were measured before and after injection of any one of six specific antagonists of 5-hydroxytriptamine receptors (5-hydroxytryptamine 1A, 5-hydroxytryptamine 2A, 5-hydroxytryptamine 2C, 5-hydroxytryptamine 3, 5-hydroxytryptamine 4 and 5-hydroxytryptamine 7) into the prefrontal cortex. The prefrontal cortex was divided into two regions, namely the prelimbic cortex and the infralimbic cortex. The experiments were carried out in conscious and free-moving rats. RESULTS: The intercontraction interval value increased significantly after injection of the 5-hydroxytriptamine 2A receptor antagonist, MDL11939, into the prelimbic cortex of the rat prefrontal cortex (7.68 ± 1.28 vs 9.02 ± 1.41 min, P < 0.05), whereas the intercontraction interval value decreased significantly after injection of the 5-hydroxytriptamine 7 antagonist SB269970 into the prelimbic cortex (9.42 ± 0.39 vs 8.14 ± 0.71 min, P < 0.05). The intercontraction interval was unaffected by injection of either of these two antagonists into the infralimbic cortex. The other four antagonists (5-hydroxytryptamine 1A, 5-hydroxytryptamine 2C, 5-hydroxytryptamine 3 and 5-hydroxytryptamine 4) had no effect on the intercontraction interval after injection into the prelimbic cortex and the infralimbic cortex. The maximum voiding pressure was unaffected by injection of any one of the six 5-hydroxytriptamine antagonists into the prelimbic cortex and infralimbic cortex. CONCLUSIONS: In the rat prefrontal cortex5-hydroxytryptamine 2A receptors excite the micturition reflex, whereas 5-hydroxytryptamine 7 receptors inhibit this reflex.


Asunto(s)
Serotonina , Micción , Animales , Femenino , Corteza Prefrontal , Ratas , Ratas Sprague-Dawley , Reflejo
9.
Neuropharmacology ; 167: 107703, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-31299228

RESUMEN

Serotonergic agents have been widely used for treatment of psychiatric disorders, but the therapeutic effects are insufficient and these drugs often induce severe side effects. We need to specify the distinct serotonergic pathways underlying each mental function to overcome these problems. Preclinical studies have demonstrated that the central serotonergic system is involved in several emotional/cognitive functions including anxiety, depression, and impulse control, but it remains unclear whether each function is regulated by a different serotonergic system. We used optogenetic strategy to increase central serotonergic activity in mice and evaluated the behavioral consequences. Pharmacological and genetic tools were used to determine the subtype of 5-HT receptors responsible for the observed effects. We demonstrated that the serotonergic activation in the median raphe nucleus enhanced anxiety-like behavior, the serotonergic activation in the dorsal raphe nucleus exerted antidepressant-like effects, and the serotonergic activation in the median or dorsal raphe nucleus suppressed impulsive action. We also found that different serotonergic terminals, ventral hippocampus, ventral tegmental area/substantia nigra, and subthalamic/parasubthalamic nucleus, are involved in regulating anxiety-like behavior, antidepressant-like, and anti-impulsive effects, respectively. Furthermore, we found, using triple-transgenic mice, that the stimulation of the 5-HT2C receptor is required to evoke anxiety-like behavior, but not to exert anti-impulsive effects. These results suggest the need for pathway-specific treatments and provide important insights that will help the development of more effective and safer therapeutics. This article is part of the special issue entitled 'Serotonin Research: Crossing Scales and Boundaries'.


Asunto(s)
Antidepresivos/administración & dosificación , Ansiedad/metabolismo , Conducta Impulsiva/fisiología , Receptor de Serotonina 5-HT2C/metabolismo , Neuronas Serotoninérgicas/metabolismo , Transducción de Señal/fisiología , Animales , Ansiedad/tratamiento farmacológico , Núcleo Dorsal del Rafe/efectos de los fármacos , Núcleo Dorsal del Rafe/metabolismo , Femenino , Conducta Impulsiva/efectos de los fármacos , Locomoción/efectos de los fármacos , Locomoción/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microinyecciones/métodos , Neuronas Serotoninérgicas/efectos de los fármacos , Agonistas del Receptor de Serotonina 5-HT2/administración & dosificación , Antagonistas del Receptor de Serotonina 5-HT2/administración & dosificación , Transducción de Señal/efectos de los fármacos
10.
Behav Brain Res ; 379: 112394, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31786274

RESUMEN

Pharmacological studies have suggested that the serotonin 5-HT2C receptor is involved in locomotor activity, anxiety, and fear memory. However, the results of locomotor activity and anxiety in 5-HT2C receptor knockout mice have been mixed, and the effects of 5-HT2C receptor knockout on contextual fear memory have not yet been addressed. In the present study, we reconcile these inconsistent results by analyzing behavioral data in detail and by examining the effects of 5-HT2C receptor knockout on contextual fear memory. We demonstrated that the higher locomotor activity in 5-HT2C receptor knockout mice was observed only in the late phase of the test, indicating that the analyses in the previous study using the total locomotor activity would lead to variable results. Moreover, by analyzing mouse behavior in detail, we found that 5-HT2C receptor knockout mice displayed a hesitating attitude by staying in the central area as well as risk assessment behavior in the elevated plus-maze test. However, the time spent in the open arms was longer in 5-HT2C receptor knockout mice than in wild-type littermates when a zero-maze test lacking the central area was used. In the contextual fear conditioning test, 5-HT2C receptor knockout mice showed rapid within-session extinction of fear, but not between-session extinction, compared with wild-type littermates. However, this remains inconclusive because the facilitation of extinction might be confounded with higher locomotor activity in 5-HT2C receptor knockout mice. Taken together, the present results provide reasonable explanations about previous inconsistent findings and partially filled the gaps between pharmacological and genetic findings.


Asunto(s)
Ansiedad/fisiopatología , Conducta Animal/fisiología , Extinción Psicológica/fisiología , Miedo/fisiología , Locomoción/fisiología , Aprendizaje por Laberinto/fisiología , Memoria/fisiología , Receptor de Serotonina 5-HT2C/fisiología , Animales , Reacción Cataléptica de Congelación/fisiología , Habituación Psicofisiológica/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Serotonina 5-HT2C/genética
11.
J Pharmacol Sci ; 141(3): 127-130, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31690490

RESUMEN

High impulsivity will increase the risk of criminal behavior, drug abuse, and suicide. We chose two drugs by following a strategy recently we proposed for identifying potential anti-impulsivity drugs, and examined the effects on impulsive action in rats by using a 3-choice serial reaction time task. We showed that the administration of blonanserin, an atypical antipsychotic, reduced impulsive actions in a U-shaped manner. 1-(2-Pyriidinyl)-piperazine, an active metabolite of buspirone or tandospirone, also slightly reduced impulsive actions, though it impaired motor functions. These results affirm the validity of our strategy, but require its refinement for developing anti-impulsivity drugs.


Asunto(s)
Antipsicóticos/farmacología , Conducta Impulsiva/efectos de los fármacos , Trastornos Mentales/tratamiento farmacológico , Piperazinas/farmacología , Piperidinas/farmacología , Animales , Conducta Animal/efectos de los fármacos , Masculino , Ratas , Ratas Wistar , Tiempo de Reacción
12.
Science ; 365(6459): 1308-1313, 2019 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-31604241

RESUMEN

The neural mechanisms underlying memory regulation during sleep are not yet fully understood. We found that melanin concentrating hormone-producing neurons (MCH neurons) in the hypothalamus actively contribute to forgetting in rapid eye movement (REM) sleep. Hypothalamic MCH neurons densely innervated the dorsal hippocampus. Activation or inhibition of MCH neurons impaired or improved hippocampus-dependent memory, respectively. Activation of MCH nerve terminals in vitro reduced firing of hippocampal pyramidal neurons by increasing inhibitory inputs. Wake- and REM sleep-active MCH neurons were distinct populations that were randomly distributed in the hypothalamus. REM sleep state-dependent inhibition of MCH neurons impaired hippocampus-dependent memory without affecting sleep architecture or quality. REM sleep-active MCH neurons in the hypothalamus are thus involved in active forgetting in the hippocampus.


Asunto(s)
Hipocampo/citología , Hormonas Hipotalámicas/fisiología , Melaninas/fisiología , Memoria , Hormonas Hipofisarias/fisiología , Células Piramidales/fisiología , Sueño REM , Animales , Conducta Animal , Hipocampo/fisiología , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
13.
Brain Res ; 1719: 243-252, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31194947

RESUMEN

Serotonin (5-HT) in the central nervous system regulates a variety of biological functions, from the basic homeostatic control to higher brain functions, by acting on fourteen known receptor subtypes. However, it is still usually unclear which receptor subtype is responsible for a specific function due to the lack of highly selective ligands for most of these receptors. Although 5-HT receptor knockout mice are useful, the brain-wide distribution of various receptors makes it difficult to dissect receptor functions in specific and brain regions and cell types. Recent advances in CRISPR/Cas9-mediated in vivo genome editing technology may overcome this problem. In this study, we constructed a viral vector expressing a single guide (sg)RNA targeting Htr1a (sgHtr1a) and Cre recombinase under the control of a neuron-specific promoter. Injection of the viral vector into the dorsal raphe nucleus (DRN) of Cre-dependent Cas9 knock-in mice induced Cre-dependent Cas9 expression mainly in DRN serotonin and GABA neurons. Mismatch cleavage assay and Sanger sequencing showed insertion or deletion formation at the target site. 5-HT1A receptor agonist-induced hypothermia was attenuated and antidepressant effect of a selective serotonin reuptake inhibitor (SSRI) was enhanced by microinjection of the viral vector expressing sgHtr1a into the DRN of Cre-dependent Cas9 knock-in mice. These results suggest that this in vivo CRISPR/Cas9-mediated 5-HT receptor gene knockout strategy provides a reliable and low-cost method for elucidating 5-HT receptor functions in specific cell types and brain regions. Further, we demonstrate that the neuronal 5-HT1A receptor in the DRN regulates body temperature and antidepressant effect of SSRI.


Asunto(s)
Regulación de la Temperatura Corporal/genética , Núcleo Dorsal del Rafe/fisiología , Receptor de Serotonina 5-HT1A/genética , Animales , Regulación de la Temperatura Corporal/fisiología , Sistemas CRISPR-Cas/genética , Núcleo Dorsal del Rafe/metabolismo , Femenino , Edición Génica/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Inhibición Neural/fisiología , Receptor de Serotonina 5-HT1A/metabolismo , Receptores de Serotonina/metabolismo , Neuronas Serotoninérgicas/metabolismo , Serotonina/metabolismo , Antagonistas de la Serotonina/farmacología , Agonistas de Receptores de Serotonina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología
14.
Eur J Pharmacol ; 844: 17-25, 2019 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-30503361

RESUMEN

Higher impulsivity is a risk factor for criminal involvement, substance abuse, and suicide. However, only a few drugs are clinically available for the treatment of deficient impulse control. We recently proposed a strategy for identifying potential drugs to treat such disorders by investigating clinically available drugs that increase extracellular dopamine levels in the medial prefrontal cortex and stimulate dopamine D1-like receptors without increasing extracellular dopamine levels in the ventral striatum. To determine whether this strategy is promising, we examined the effects of duloxetine, a serotonin-noradrenaline reuptake inhibitor that might meet these criteria, on impulsive action in adult male Wistar/ST rats using a 3-choice serial reaction time task. The effects of duloxetine on extracellular dopamine levels in the medial prefrontal cortex and nucleus accumbens, a part of the ventral striatum were evaluated using in vivo microdialysis, as the noradrenaline transporter transports dopamine in some brain regions. Our results showed that the administration of duloxetine reduced impulsive actions and increased extracellular dopamine levels in the mPFC but not in the nucleus accumbens. Microinjection of a selective D1-like receptor antagonist into the infralimbic cortex blocked the suppression of impulsive action by duloxetine. In addition, we demonstrated that the microinjection also blocked the suppression of impulsive action by atomoxetine, a noradrenaline reuptake inhibitor and an established anti-impulsive drug. These results support our proposed strategy for identifying and developing anti-impulsivity drugs.


Asunto(s)
Clorhidrato de Duloxetina/farmacología , Conducta Impulsiva/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Inhibidores de Captación de Serotonina y Norepinefrina/farmacología , Animales , Clorhidrato de Atomoxetina/farmacología , Conducta Animal/efectos de los fármacos , Benzazepinas/farmacología , Conducta de Elección/efectos de los fármacos , Dopamina/metabolismo , Masculino , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Corteza Prefrontal/metabolismo , Ratas Wistar , Receptores de Dopamina D1/antagonistas & inhibidores , Receptores de Dopamina D1/metabolismo , Serotonina/metabolismo
15.
Behav Brain Res ; 343: 61-70, 2018 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-29355672

RESUMEN

Immaturity in impulse control among adolescents could result in substance abuse, criminal involvement, and suicide. The brains of adolescents and adults are anatomically, neurophysiologically, and pharmacologically different. Therefore, preclinical models of adolescent impulsivity are required to screen drugs for adolescents and elucidate the neural mechanisms underlying age-related differences in impulsivity. The conventional 3- or 5-choice serial reaction time task, which is a widely used task to assess impulsivity in adult rodents, cannot be used for young mice because of two technical problems: impaired growth caused by food restriction and the very long training duration. To overcome these problems, we altered the conventional training process, optimizing the degree of food restriction for young animals and shortening the training duration. We found that almost all basal performance levels were similar between the novel and conventional procedures. We also confirmed the pharmacological validity of our results: the 5-hydroxytryptamine 2C (5-HT2C) receptor agonist Ro60-0175 (0.6 mg/kg, subcutaneous) reduced the occurrence of premature responses, whereas the 5-HT2C receptor antagonist SB242084 (0.5 mg/kg intraperitoneal) increased their occurrence, consistent with results of previous studies using conventional procedures. Furthermore, we detected age-related differences in impulsivity using the novel procedure: adolescent mice were found to be more impulsive than adult mice, congruent with the results of human studies. Thus, the new procedure enables the assessment of impulsivity in adolescent mice and facilitates a better understanding of the neurophysiological/pharmacological properties of adolescents.


Asunto(s)
Conducta Impulsiva , Ratones Endogámicos C57BL/crecimiento & desarrollo , Ratones Endogámicos C57BL/psicología , Modelos Animales , Pruebas Neuropsicológicas , Envejecimiento/psicología , Aminopiridinas/farmacología , Animales , Etilaminas/farmacología , Función Ejecutiva/efectos de los fármacos , Función Ejecutiva/fisiología , Privación de Alimentos/fisiología , Conducta Impulsiva/efectos de los fármacos , Conducta Impulsiva/fisiología , Indoles/farmacología , Inhibición Psicológica , Aprendizaje/efectos de los fármacos , Aprendizaje/fisiología , Masculino , Ratones Endogámicos C57BL/fisiología , Receptor de Serotonina 5-HT2C/metabolismo , Agonistas del Receptor de Serotonina 5-HT2/farmacología , Antagonistas del Receptor de Serotonina 5-HT2/farmacología , Factores de Tiempo
16.
J Pharmacol Sci ; 136(1): 1-8, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29277371

RESUMEN

Childhood maltreatment is associated with impaired adult brain function, particularly in the hippocampus, and is not only a major risk factor for some psychiatric diseases but also affects early social development and social adaptation in later life. The aims of this study were to determine whether early postnatal stress affects social behavior and whether repeated fluvoxamine treatment reverses these changes. Rat pups were exposed to footshock stress during postnatal days 21-25 (at 3 weeks old: 3wFS). During the post-adolescent period (10-14 weeks postnatal), the social interaction test and Golgi-cox staining of dorsal hippocampal pyramidal neurons were performed. Following exposure to footshock stress, 3wFS rats showed an increase in social interaction time, which might be practically synonymous with hypersociability, and a decrease in spine density in the CA3 hippocampal subregion, but not in CA1. These behavioral and morphological changes were both recovered by repeated oral administration of fluvoxamine at a dose of 10 mg/kg/day for 14 days. These findings suggest that the vulnerability of the hippocampal CA3 region is closely related to social impairments induced by physical stress during the juvenile period and shed some light on therapeutic alternatives for early postnatal stress-induced emotional dysfunction.


Asunto(s)
Fluvoxamina/administración & dosificación , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Trastorno de la Conducta Social/tratamiento farmacológico , Trastorno de la Conducta Social/etiología , Estrés Fisiológico , Administración Oral , Animales , Modelos Animales de Enfermedad , Hipocampo/patología , Humanos , Masculino , Ratas Wistar , Trastorno de la Conducta Social/patología , Trastorno de la Conducta Social/prevención & control , Trastornos por Estrés Postraumático
17.
Eur J Neurosci ; 47(1): 40-47, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29131433

RESUMEN

Pain is a complex experience with both sensory and affective components. Clinical and preclinical studies have shown that the affective component of pain can be reduced by doses of morphine lower than those necessary to reduce the sensory component. Although the neural mechanisms underlying the effects of morphine on the sensory component of pain have been investigated extensively, those influencing the affective component remain to be elucidated. The bed nucleus of the stria terminalis (BNST) has been implicated in the regulation of various negative emotional states, including aversion, anxiety and fear. Thus, this study aimed to clarify the role of the ventral part of the BNST (vBNST) in the actions of morphine on the affective and sensory components of pain. First, the effects of intra-vBNST injections of morphine on intraplantar formalin-induced conditioned place aversion (CPA) and nociceptive behaviors were investigated. Intra-vBNST injections of morphine reduced CPA without affecting nociceptive behaviors, which suggests that intra-vBNST morphine alters the affective, but not sensory, component of pain. Next, to examine the effects of morphine on neuronal excitability in type II vBNST neurons, whole-cell patch-clamp recordings were performed in brain slices. Bath application of morphine hyperpolarized type II vBNST neurons. Thus, the suppressive effects of intra-vBNST morphine on pain-induced aversion may be due to its inhibitory effects on neuronal excitability in type II vBNST neurons. These results suggest that the vBNST is a key brain region involved in the suppressive effects of morphine on the affective component of pain.


Asunto(s)
Analgésicos Opioides/farmacología , Condicionamiento Clásico , Morfina/farmacología , Nocicepción , Dolor/fisiopatología , Núcleos Septales/efectos de los fármacos , Potenciales de Acción , Animales , Masculino , Neuronas/fisiología , Ratas , Ratas Sprague-Dawley , Núcleos Septales/citología
18.
J Pharmacol Sci ; 134(3): 181-189, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28694090

RESUMEN

Serotonin/noradrenaline reuptake inhibitors (SNRIs) are widely used for the treatment for major depressive disorder, but these drugs induce several side effects including increased aggression and impulsivity, which are risk factors for substance abuse, criminal involvement, and suicide. To address this issue, milnacipran (0, 3, 10, or 30 mg/kg), an SNRI and antidepressant, was intraperitoneally administered to mice prior to the 3-choice serial reaction time task, resident-intruder test, and forced swimming test to measure impulsive, aggressive, and depressive-like behaviors, respectively. A milnacipran dose of 10 mg/kg suppressed all behaviors, which was accompanied by increased dopamine and serotonin levels in the medial prefrontal cortex (mPFC) but not in the nucleus accumbens (NAc). Although the most effective dose for depressive-like behavior was 30 mg/kg, the highest dose increased aggressive behavior and unaffected impulsive behavior. Increased dopamine levels in the NAc could be responsible for the effects. In addition, the mice basal impulsivity was negatively correlated with the latency to the first agonistic behavior. Thus, the optimal dose range of milnacipran is narrower than previously thought. Finding drugs that increase serotonin and dopamine levels in the mPFC without affecting dopamine levels in the NAc is a potential strategy for developing novel antidepressants.


Asunto(s)
Agresión/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Ciclopropanos/farmacología , Ciclopropanos/uso terapéutico , Trastorno Depresivo Mayor/tratamiento farmacológico , Conducta Impulsiva/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/uso terapéutico , Animales , Ciclopropanos/administración & dosificación , Dopamina/metabolismo , Relación Dosis-Respuesta a Droga , Inyecciones Intraperitoneales , Masculino , Ratones Endogámicos C57BL , Milnaciprán , Corteza Prefrontal/metabolismo , Serotonina/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación
19.
Proc Natl Acad Sci U S A ; 114(17): E3526-E3535, 2017 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-28396432

RESUMEN

Narcolepsy is a sleep disorder caused by the loss of orexin (hypocretin)-producing neurons and marked by excessive daytime sleepiness and a sudden weakening of muscle tone, or cataplexy, often triggered by strong emotions. In a mouse model for narcolepsy, we previously demonstrated that serotonin neurons of the dorsal raphe nucleus (DRN) mediate the suppression of cataplexy-like episodes (CLEs) by orexin neurons. Using an optogenetic tool, in this paper we show that the acute activation of DRN serotonin neuron terminals in the amygdala, but not in nuclei involved in regulating rapid eye-movement sleep and atonia, suppressed CLEs. Not only did stimulating serotonin nerve terminals reduce amygdala activity, but the chemogenetic inhibition of the amygdala using designer receptors exclusively activated by designer drugs also drastically decreased CLEs, whereas chemogenetic activation increased them. Moreover, the optogenetic inhibition of serotonin nerve terminals in the amygdala blocked the anticataplectic effects of orexin signaling in DRN serotonin neurons. Taken together, the results suggest that DRN serotonin neurons, as a downstream target of orexin neurons, inhibit cataplexy by reducing the activity of amygdala as a center for emotional processing.


Asunto(s)
Amígdala del Cerebelo , Catalepsia , Núcleo Dorsal del Rafe , Neuronas Serotoninérgicas/metabolismo , Transducción de Señal , Amígdala del Cerebelo/metabolismo , Amígdala del Cerebelo/patología , Amígdala del Cerebelo/fisiopatología , Animales , Catalepsia/genética , Catalepsia/metabolismo , Catalepsia/patología , Catalepsia/fisiopatología , Núcleo Dorsal del Rafe/metabolismo , Núcleo Dorsal del Rafe/patología , Núcleo Dorsal del Rafe/fisiopatología , Movimientos Oculares , Masculino , Ratones , Ratones Noqueados , Neuronas Serotoninérgicas/patología , Serotonina/metabolismo
20.
Neurosci Lett ; 650: 72-76, 2017 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-28432029

RESUMEN

Anhedonia, the loss of interest or pleasure in previously enjoyable activities, is a core symptom of major depressive disorder, suggesting that the brain reward system may be dysfunctional in this condition. Neurochemical changes in the mesolimbic dopamine (DA) system are not fully understood in animal models of depression. We investigated reward (30% sucrose intake)-induced DA release in the nucleus accumbens (NAc) and the effect of chronic treatment with the antidepressant escitalopram (5mg/kg, intraperitoneally twice daily for 3 weeks) in two animal models of depression. Exposure to chronic mild stress (CMS) during adulthood completely suppressed reward-induced intra-NAc DA release; however, this effect was reversed by chronic treatment with escitalopram. Our findings suggest that reward-induced intra-NAc DA release may be an indicator of depression severity and therapeutic efficacy. Exposure to neonatal maternal separation (MS) and CMS in adulthood completely suppressed reward-induced intra-NAc DA release. Chronic treatment with escitalopram did not restore reward-induced DA release in these animals, suggesting that this paradigm may serve as an animal model for treatment-resistant depression. Further study of the mesolimbic dopaminergic system in these animal models of depression may clarify the neural mechanisms underlying depression and treatment resistance.


Asunto(s)
Citalopram/administración & dosificación , Depresión/tratamiento farmacológico , Depresión/metabolismo , Modelos Animales de Enfermedad , Dopamina/metabolismo , Núcleo Accumbens/metabolismo , Recompensa , Animales , Antidepresivos/administración & dosificación , Depresión/etiología , Inhibición Psicológica , Masculino , Privación Materna , Inhibición Neural/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Estrés Psicológico/complicaciones , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/metabolismo , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA