Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Eur J Pharmacol ; 978: 176790, 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-38942263

RESUMO

Nicotine has been shown to enhance object recognition memory in the novel object recognition (NOR) test by activating excitatory neurons in the medial prefrontal cortex (mPFC). However, the exact neuronal mechanisms underlying the nicotine-induced activation of mPFC neurons and the resultant memory enhancement remain poorly understood. To address this issue, we performed brain-slice electrophysiology and the NOR test in male C57BL/6J mice. Whole-cell patch-clamp recordings from layer V pyramidal neurons in the mPFC revealed that nicotine augments the summation of evoked excitatory postsynaptic potentials (eEPSPs) and that this effect was suppressed by N-[3,5-Bis(trifluoromethyl)phenyl]-N'-[2,4-dibromo-6-(2H-tetrazol-5-yl)phenyl]urea (NS5806), a voltage-dependent potassium (Kv) 4.3 channel activator. In line with these findings, intra-mPFC infusion of NS5806 suppressed systemically administered nicotine-induced memory enhancement in the NOR test. Additionally, miRNA-mediated knockdown of Kv4.3 channels in mPFC pyramidal neurons enhanced object recognition memory. Furthermore, inhibition of A-type Kv channels by intra-mPFC infusion of 4-aminopyridine was found to enhance object recognition memory, while this effect was abrogated by prior intra-mPFC NS5806 infusion. These results suggest that nicotine augments the summation of eEPSPs via the inhibition of Kv4.3 channels in mPFC layer V pyramidal neurons, resulting in the enhancement of object recognition memory.


Assuntos
Camundongos Endogâmicos C57BL , Nicotina , Córtex Pré-Frontal , Reconhecimento Psicológico , Animais , Masculino , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/fisiologia , Córtex Pré-Frontal/metabolismo , Nicotina/farmacologia , Camundongos , Reconhecimento Psicológico/efeitos dos fármacos , Canais de Potássio Shal/metabolismo , Células Piramidais/efeitos dos fármacos , Células Piramidais/fisiologia , Memória/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos
2.
Neuropharmacology ; 239: 109672, 2023 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-37506875

RESUMO

Previous studies postulated that chronic administration of varenicline, a partial and full agonist at α4ß2 and α7 nicotinic acetylcholine receptors (nAChRs), respectively, enhances recognition memory. However, whether its acute administration is effective, on which brain region(s) it acts, and in what signaling it is involved, remain unknown. To address these issues, we conducted a novel object recognition test using male C57BL/6J mice, focusing on the medial prefrontal cortex (mPFC), a brain region associated with nicotine-induced enhancement of recognition memory. Systemic administration of varenicline before the training dose-dependently enhanced recognition memory. Intra-mPFC varenicline infusion also enhanced recognition memory, and this enhancement was blocked by intra-mPFC co-infusion of a selective α7, but not α4ß2, nAChR antagonist. Consistent with this, intra-mPFC infusion of a selective α7 nAChR agonist augmented object recognition memory. Furthermore, intra-mPFC co-infusion of U-73122, a phospholipase C (PLC) inhibitor, or 2-aminoethoxydiphenylborane (2-APB), an inositol trisphosphate (IP3) receptor inhibitor, suppressed the varenicline-induced memory enhancement, suggesting that α7 nAChRs may also act as Gq-coupled metabotropic receptors. Additionally, whole-cell recordings from mPFC layer V pyramidal neurons in vitro revealed that varenicline significantly increased the summation of evoked excitatory postsynaptic potentials, and this effect was suppressed by U-73122 or 2-APB. These findings suggest that varenicline might acutely enhance recognition memory via mPFC α7 nAChR stimulation, followed by mPFC neuronal excitation, which is mediated by the activation of PLC and IP3 receptor signaling. Our study provides evidence supporting the potential repositioning of varenicline as a treatment for cognitive impairment.


Assuntos
Receptores Nicotínicos , Receptor Nicotínico de Acetilcolina alfa7 , Camundongos , Masculino , Animais , Vareniclina/farmacologia , Receptores Nicotínicos/metabolismo , Camundongos Endogâmicos C57BL , Córtex Pré-Frontal/metabolismo
3.
Sci Rep ; 13(1): 8089, 2023 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-37208473

RESUMO

Stress is one of the critical facilitators for seizure induction in patients with epilepsy. However, the neural mechanisms underlying this facilitation remain poorly understood. Here, we investigated whether noradrenaline (NA) transmission enhanced by stress exposure facilitates the induction of medial prefrontal cortex (mPFC)-originated seizures. In mPFC slices, whole-cell current-clamp recordings revealed that bath application of picrotoxin induced sporadic epileptiform activities (EAs), which consisted of depolarization with bursts of action potentials in layer 5 pyramidal cells. Addition of NA dramatically shortened the latency and increased the number of EAs. Simultaneous whole-cell and field potential recordings revealed that the EAs are synchronous in the mPFC local circuit. Terazosin, but not atipamezole or timolol, inhibited EA facilitation, indicating the involvement of α1 adrenoceptors. Intra-mPFC picrotoxin infusion induced seizures in mice in vivo. Addition of NA substantially shortened the seizure latency, while co-infusion of terazosin into the mPFC inhibited the effect of NA. Finally, acute restraint stress shortened the latency of intra-mPFC picrotoxin infusion-induced seizures, whereas prior infusion of terazosin reversed this stress-induced shortening of seizure latency. Our findings suggest that stress facilitates the induction of mPFC-originated seizures via NA stimulation of α1 adrenoceptors.


Assuntos
Norepinefrina , Córtex Pré-Frontal , Ratos , Camundongos , Animais , Ratos Sprague-Dawley , Picrotoxina/farmacologia , Norepinefrina/farmacologia , Córtex Pré-Frontal/fisiologia , Convulsões/induzido quimicamente , Convulsões/tratamento farmacológico , Receptores Adrenérgicos
4.
Eur J Pharmacol ; 946: 175653, 2023 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-36907260

RESUMO

3,4-methylenedioxymethamphetamine (MDMA), a recreational drug, induces euphoric sensations and psychosocial effects, such as increased sociability and empathy. Serotonin, also called 5-hydroxytryptamine (5-HT), is a neurotransmitter that has been associated with MDMA-induced prosocial effects. However, the detailed neural mechanisms remain elusive. In the present study, we investigated whether 5-HT neurotransmission in the medial prefrontal cortex (mPFC) and the basolateral nucleus of amygdala (BLA) is involved in MDMA-induced prosocial effects using the social approach test in male ICR mice. Systemic administration of (S)-citalopram, a selective 5-HT transporter inhibitor, before administration of MDMA failed to suppress MDMA-induced prosocial effects. On the other hand, systemic administration of the 5-HT1A receptor antagonist WAY100635, but not 5-HT1B, 5-HT2A, 5-HT2C, or 5-HT4 receptor antagonist, significantly suppressed MDMA-induced prosocial effects. Furthermore, local administration of WAY100635 into the BLA but not into the mPFC suppressed MDMA-induced prosocial effects. Consistent with this finding, intra-BLA MDMA administration significantly increased sociability. Together, these results suggest that MDMA induces prosocial effects through the stimulation of 5-HT1A receptors in the BLA.


Assuntos
Complexo Nuclear Basolateral da Amígdala , N-Metil-3,4-Metilenodioxianfetamina , Camundongos , Masculino , Animais , N-Metil-3,4-Metilenodioxianfetamina/farmacologia , Receptor 5-HT1A de Serotonina , Camundongos Endogâmicos ICR , Antagonistas da Serotonina/farmacologia , Serotonina
5.
Cell Rep ; 42(3): 112149, 2023 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-36821440

RESUMO

Major depressive disorder (MDD) is among the most common mental illnesses. Serotonergic (5-HT) neurons are central to the pathophysiology and treatment of MDD. Repeatedly recalling positive episodes is effective for MDD. Stimulating 5-HT neurons of the dorsal raphe nucleus (DRN) or neuronal ensembles in the dorsal dentate gyrus (dDG) associated with positive memories reverses the stress-induced behavioral abnormalities. Despite this phenotypic similarity, their causal relationship is unclear. This study revealed that the DRN 5-HT neurons activate dDG neurons; surprisingly, this activation was specifically observed in positive memory ensembles rather than neutral or negative ensembles. Furthermore, we revealed that dopaminergic signaling induced by activation of DRN 5-HT neurons projecting to the ventral tegmental area mediates an increase in active coping behavior and positive dDG ensemble reactivation. Our study identifies a role of DRN 5-HT neurons as specific reactivators of positive memories and provides insights into how serotonin elicits antidepressive effects.


Assuntos
Transtorno Depressivo Maior , Núcleo Dorsal da Rafe , Humanos , Neurônios Serotoninérgicos , Serotonina/farmacologia , Giro Denteado
6.
Bio Protoc ; 12(23)2022 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-36561114

RESUMO

Pavlovian fear conditioning is a widely used procedure to assess learning and memory processes that has also been extensively used as a model of post-traumatic stress disorder (PTSD). Freezing, the absence of movement except for respiratory-related movements, is commonly used as a measure of fear response in non-human animals. However, this measure of fear responses can be affected by a different baseline of locomotor activity between groups and/or conditions. Moreover, fear conditioning procedures are usually restricted to a single conditioned stimulus (e.g., a tone cue, the context, etc.) and thus do not depict the complexity of real-life situations where traumatic memories are composed of a complex set of stimuli associated with the same aversive event. To overcome this issue, we use a conditioned lick suppression paradigm where water-deprived mice are presented with a single conditioned stimulus (CS, a tone cue or the context) previously paired with an unconditioned stimulus (US, a foot shock) while consuming water. We use the ratio of number of licks before and during the CS presentation as a fear measure, thereby neutralizing the potential effect of locomotor activity in fear responses. We further implemented the conditioned lick suppression ratio to assess the effect of cue competition using a compound of contextual and tone cue conditioned stimuli that were extinguished separately. This paradigm should prove useful in assessing potential therapeutics and/or behavioral therapies in PTSD, while neutralizing potential confounding effects between locomotor activity and fear responses on one side, and by considering potential cue-competition effects on the other side. This protocol was validated in: Transl Psychiatry (2022), DOI: 10.1038/s41398-022-01815-2 Graphical abstract Schematic representation of the compound context-cue condition lick suppression procedure. Illustration reproduced from Bouchekioua et al. (2022).

7.
Nat Commun ; 13(1): 7708, 2022 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-36550097

RESUMO

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.


Assuntos
Núcleo Interpeduncular , Neurônios Serotoninérgicos , Camundongos , Animais , Serotonina/fisiologia , Núcleo Dorsal da Rafe/fisiologia , Receptores de Serotonina
8.
Behav Brain Res ; 432: 113981, 2022 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-35777550

RESUMO

Spatiotemporal patterns of neuronal activity underlying the motivational effect of rotating running wheels (RWs) in rodents remain largely undetermined. Here, we investigated changes of neuronal activity among brain regions associated with motivation across different intensities of motivation for RWs in mice. Daily exposure to RWs gradually increased rotation number, then became stable after approximately 3 weeks. Immunohistochemical analyses revealed that the number of c-Fos (a neuronal activity marker)-positive cells increased in the medial prefrontal cortex (mPFC), core and shell of the nucleus accumbens (NAc), dorsal striatum (Str), and lateral septum (LS) at day 1, day 9, and days 20-24, in a time-dependent manner. Additionally, despite exposure to locked RWs for over 7 days after establishing stable rotation with 3-week RW access, increased c-Fos expression was still observed in most of these brain areas. Furthermore, daily overnight RW access developed stable rotation by day 6, with high and low rotation numbers at the start and end of the overnight session, respectively. The number of c-Fos-positive cells at the start of RW rotation was significantly higher than at the end of RW rotation in most brain regions. Furthermore, after establishing stable rotation, the number of c-Fos-positive cells increased in the mPFC and shell of the NAc of mice that only observed RWs. These findings suggest that the subareas of the mPFC and NAc may be critically involved in the motivational effects of RW rotations.


Assuntos
Motivação , Atividade Motora , Animais , Camundongos , Atividade Motora/fisiologia , Neurônios/metabolismo , Núcleo Accumbens , Proteínas Proto-Oncogênicas c-fos/metabolismo
9.
Sci Rep ; 12(1): 6014, 2022 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-35399112

RESUMO

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.


Assuntos
Dopamina , Receptores de Dopamina D5 , Animais , Humanos , Locomoção , Camundongos , Camundongos Knockout , Receptores de Dopamina D1 , Receptores de Dopamina D5/fisiologia
10.
Transl Psychiatry ; 12(1): 58, 2022 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-35145065

RESUMO

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.


Assuntos
Sinais (Psicologia) , Medo , Receptor 5-HT2C de Serotonina/metabolismo , Animais , Extinção Psicológica/fisiologia , Medo/fisiologia , Camundongos , Camundongos Knockout , Receptor 5-HT2C de Serotonina/genética , Serotonina
11.
J Pharmacol Sci ; 147(1): 58-61, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34294373

RESUMO

Nicotine administration enhances object recognition memory. However, target brain regions and cellular mechanisms underlying the nicotine effects remain unclear. In mice, the novel object recognition test revealed that systemic nicotine administration before training enhanced object recognition memory. Moreover, this effect was inhibited by infusion of retigabine, a selective voltage-dependent potassium 7 (Kv7) channel opener, into the medial prefrontal cortex (mPFC) before nicotine administration. Additionally, infusion of XE-991, a selective Kv7 channel blocker, into the mPFC before training enhanced object recognition memory. Therefore, Kv7 channels in the mPFC may be at least partly involved in nicotine-induced enhancement of object recognition memory.


Assuntos
Memória/efeitos dos fármacos , Nicotina/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Córtex Pré-Frontal/metabolismo , Reconhecimento Psicológico/efeitos dos fármacos , Animais , Antracenos/farmacologia , Carbamatos/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Fenilenodiaminas/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Estimulação Química
12.
Biol Pharm Bull ; 44(7): 1007-1013, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34193682

RESUMO

Nicotine has been known to enhance recognition memory in various species. However, the brain region where nicotine acts and exerts its effect remains unclear. Since the medial prefrontal cortex (mPFC) is associated with memory, we examined the role of the mPFC in nicotine-induced enhancement of recognition memory using the novel object recognition test in male C57BL/6J mice. Systemic nicotine administration 10 min before training session significantly enhanced object recognition memory in test session that was performed 24 h after the training. Intra-mPFC infusion of mecamylamine, a non-selective nicotinic acetylcholine receptor (nAChR) antagonist, 5 min before nicotine administration blocked the effect of nicotine. Additionally, intra-mPFC infusion of dihydro-ß-erythroidine, a selective α4ß2 nAChR antagonist, or methyllycaconitine, a selective α7 nAChR antagonist, significantly suppressed the nicotine-induced object recognition memory enhancement. Finally, intra-mPFC infusion of nicotine 1 min before the training session augmented object recognition memory in a dose-dependent manner. These findings suggest that mPFC α4ß2 and α7 nAChRs mediate the nicotine-induced object recognition memory enhancement.


Assuntos
Nicotina/farmacologia , Agonistas Nicotínicos/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Receptores Nicotínicos/fisiologia , Reconhecimento Psicológico/efeitos dos fármacos , Receptor Nicotínico de Acetilcolina alfa7/fisiologia , Aconitina/análogos & derivados , Aconitina/farmacologia , Animais , Di-Hidro-beta-Eritroidina/farmacologia , Masculino , Mecamilamina/farmacologia , Camundongos Endogâmicos C57BL , Antagonistas Nicotínicos/farmacologia , Córtex Pré-Frontal/fisiologia , Receptor Nicotínico de Acetilcolina alfa7/antagonistas & inibidores
13.
Biol Pharm Bull ; 44(5): 724-731, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33952828

RESUMO

Nicotine enhances attention, working memory and recognition. One of the brain regions associated with these effects of nicotine is the medial prefrontal cortex (mPFC). However, cellular mechanisms that induce the enhancing effects of nicotine remain unclear. To address this issue, we performed whole-cell patch-clamp recordings from mPFC layer 5 pyramidal neurons in slices of C57BL/6J mice. Shortly (approx. 2 min) after bath application of nicotine, the number of action potentials, which were elicited by depolarizing current injection, was increased, and this increase persisted for over 5 min. The effect of nicotine was blocked by the α4ß2 nicotinic acetylcholine receptor (nAChR) antagonist dihydro-ß-erythroidine, α7 nAChR antagonist methyllycaconitine, or intracellular perfusion with the Ca2+ chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA). Additionally, the voltage-dependent potassium 7 (Kv7) channel blocker, 10,10-bis(4-pyridinylmethyl)-9(10H)-anthracenone dihydrochloride (XE-991), as well as nicotine, shortened the spike threshold latency and increased the spike numbers. By contrast, the Kv7 channel opener, retigabine reduced the number of firings, and the addition of nicotine did not increase the spike numbers. These results indicate that nicotine induces long-lasting enhancement of firing activity in mPFC layer 5 pyramidal neurons, which is mediated by the stimulation of the α4ß2 and α7 nAChRs and subsequent increase in intracellular Ca2+ levels followed by the suppression of the Kv7 channels. The novel effect of nicotine might underlie the nicotine-induced enhancement of attention, working memory and recognition.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Nicotina/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/antagonistas & inibidores , Córtex Pré-Frontal/efeitos dos fármacos , Células Piramidais/efeitos dos fármacos , Animais , Antracenos/farmacologia , Células Cultivadas , Feminino , Masculino , Camundongos , Antagonistas Nicotínicos/farmacologia , Técnicas de Patch-Clamp , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Córtex Pré-Frontal/citologia , Córtex Pré-Frontal/metabolismo , Cultura Primária de Células , Células Piramidais/metabolismo , Receptores Nicotínicos/metabolismo
14.
Biochem Biophys Res Commun ; 562: 62-68, 2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34038754

RESUMO

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.


Assuntos
Nicotina/efeitos adversos , Núcleos da Rafe/patologia , Neurônios Serotoninérgicos/patologia , Síndrome de Abstinência a Substâncias/patologia , Animais , Feminino , Masculino , Mecamilamina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Optogenética , Receptores Nicotínicos/metabolismo , Serotonina/metabolismo
15.
Curr Biol ; 31(11): 2446-2454.e5, 2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-33838101

RESUMO

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.


Assuntos
Núcleo Dorsal da Rafe , Serotonina , Animais , Tomada de Decisões , Humanos , Neurônios , Estudos Prospectivos , Estudos Retrospectivos , Neurônios Serotoninérgicos
16.
Neurosci Lett ; 743: 135555, 2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33352288

RESUMO

Stress enhances cocaine craving. We recently reported that acute restraint stress increases cocaine conditioned place preference (CPP) in mice; however, the underlying mechanisms remain unclear. This study aimed to examine the role of serotonergic transmission in the medial prefrontal cortex (mPFC) in cocaine CPP enhancement by acute restraint stress, which increases extracellular serotonin (5-HT) levels in the mPFC. Intra-mPFC infusion of the selective serotonin reuptake inhibitor (S)-citalopram prior to the test session significantly increased the cocaine CPP score under non-stressed conditions. This is indicative of the substantial role of increased mPFC 5-HT levels in cocaine CPP enhancement. Moreover, intra-mPFC and systemic administration of the 5-HT1A receptor antagonist WAY100635 immediately before restraint stress exposure significantly attenuated stress-induced cocaine CPP enhancement. Our findings suggest that enhanced serotonergic transmission via 5-HT1A receptors in the mPFC is involved in acute stress-induced augmentation of rewarding memory of cocaine; moreover, the 5-HT1A receptor could be a therapeutic target for stress-induced cocaine craving.


Assuntos
Cocaína/administração & dosagem , Memória/fisiologia , Córtex Pré-Frontal/metabolismo , Receptor 5-HT1A de Serotonina/metabolismo , Recompensa , Estresse Psicológico/metabolismo , Animais , Comportamento Aditivo/tratamento farmacológico , Comportamento Aditivo/metabolismo , Comportamento Aditivo/psicologia , Inibidores da Captação de Dopamina/administração & dosagem , Infusões Intraventriculares , Masculino , Memória/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Piperazinas/administração & dosagem , Córtex Pré-Frontal/efeitos dos fármacos , Piridinas/administração & dosagem , Restrição Física/efeitos adversos , Restrição Física/psicologia , Neurônios Serotoninérgicos/efeitos dos fármacos , Neurônios Serotoninérgicos/metabolismo , Serotonina/metabolismo , Antagonistas do Receptor 5-HT1 de Serotonina/administração & dosagem , Estresse Psicológico/psicologia
17.
Int J Mol Sci ; 21(6)2020 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-32245184

RESUMO

BACKGROUND: Reward processing is fundamental for animals to survive and reproduce. Many studies have shown the importance of dorsal raphe nucleus (DRN) serotonin (5-HT) neurons in this process, but the strongly correlative link between the activity of DRN 5-HT neurons and rewarding/aversive potency is under debate. Our primary objective was to reveal this link using two different strategies to transduce DRN 5-HT neurons. METHODS: For transduction of 5-HT neurons in wildtype mice, adeno-associated virus (AAV) bearing the mouse tryptophan hydroxylase 2 (TPH2) gene promoter was used. For transduction in Tph2-tTA transgenic mice, AAVs bearing the tTA-dependent TetO enhancer were used. To manipulate the activity of 5-HT neurons, optogenetic actuators (CheRiff, eArchT) were expressed by AAVs. For measurement of rewarding/aversive potency, we performed a nose-poke self-stimulation test and conditioned place preference (CPP) test. RESULTS: We found that stimulation of DRN 5-HT neurons and their projections to the ventral tegmental area (VTA) increased the number of nose-pokes in self-stimulation test and CPP scores in both targeting methods. Concomitantly, CPP scores were decreased by inhibition of DRN 5-HT neurons and their projections to VTA. CONCLUSION: Our findings indicate that the activity of DRN 5-HT neurons projecting to the VTA is a key modulator of balance between reward and aversion.


Assuntos
Núcleo Dorsal da Rafe/fisiologia , Neurônios/fisiologia , Serotonina/metabolismo , Triptofano Hidroxilase/metabolismo , Área Tegmentar Ventral/fisiologia , Animais , Prosencéfalo Basal/metabolismo , Prosencéfalo Basal/fisiologia , Escala de Avaliação Comportamental , Núcleo Central da Amígdala/metabolismo , Núcleo Central da Amígdala/fisiologia , Dependovirus/genética , Núcleo Dorsal da Rafe/metabolismo , Elementos Facilitadores Genéticos , Vetores Genéticos , Região Hipotalâmica Lateral/metabolismo , Região Hipotalâmica Lateral/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/metabolismo , Núcleo Accumbens/metabolismo , Núcleo Accumbens/fisiologia , Optogenética , Regiões Promotoras Genéticas , Recompensa , Serotonina/fisiologia , Triptofano Hidroxilase/genética , Área Tegmentar Ventral/metabolismo
18.
Neuropharmacology ; 166: 107968, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32035731

RESUMO

Stress augments the rewarding memory of cocaine, which plays a critical role in inducing cocaine craving. However, the neurobiological mechanisms underlying the enhancing effect of stress remain unclear. Here, we show that noradrenaline (NA) transmission in the medial prefrontal cortex (mPFC) mediates stress-induced enhancement of cocaine craving. When mice were exposed to acute restraint stress immediately before the posttest session of the cocaine-induced conditioned place preference (CPP) paradigm, the CPP score was significantly higher than that in non-stressed mice. Because extracellular NA levels have been reported to be increased in the mPFC during stress exposure, we assessed the effects of NA on mPFC layer 5 pyramidal cell activity. Whole-cell recordings revealed that NA application induces depolarization and a concomitant increase in spontaneous excitatory postsynaptic currents (sEPSCs). The NA effects were inhibited by terazosin, but not by yohimbine or timolol, and the sEPSC increase was not observed in the presence of tetrodotoxin, suggesting the involvement of postsynaptic α1, but not α2 or ß, adrenoceptors in the NA effects. Additionally, intra-mPFC injection of terazosin before stress exposure attenuated the stress-induced increase in cocaine CPP. Intra-mPFC injection of phenylephrine, an α1 adrenoceptor agonist, before the posttest session without stress exposure significantly enhanced cocaine CPP. Furthermore, chemogenetic suppression of mPFC pyramidal cells with inhibitory DREADD (designer receptors exclusively activated by designer drugs) also suppressed the stress-induced CPP enhancement. These findings suggest that the stress-induced increase in NA transmission activates mPFC pyramidal cells via α1 adrenoceptor stimulation, leading to enhancement of cocaine craving-related behavior.


Assuntos
Cocaína/administração & dosagem , Memória/fisiologia , Córtex Pré-Frontal/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Recompensa , Estresse Psicológico/metabolismo , Agonistas de Receptores Adrenérgicos alfa 1/administração & dosagem , Antagonistas de Receptores Adrenérgicos alfa 1/administração & dosagem , Animais , Inibidores da Captação de Dopamina/administração & dosagem , Feminino , Masculino , Memória/efeitos dos fármacos , Camundongos , Microinjeções/métodos , Córtex Pré-Frontal/efeitos dos fármacos , Restrição Física , Estresse Psicológico/psicologia
19.
Neuropharmacology ; 167: 107703, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31299228

RESUMO

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'.


Assuntos
Antidepressivos/administração & dosagem , Ansiedade/metabolismo , Comportamento Impulsivo/fisiologia , Receptor 5-HT2C de Serotonina/metabolismo , Neurônios Serotoninérgicos/metabolismo , Transdução de Sinais/fisiologia , Animais , Ansiedade/tratamento farmacológico , Núcleo Dorsal da Rafe/efeitos dos fármacos , Núcleo Dorsal da Rafe/metabolismo , Feminino , Comportamento Impulsivo/efeitos dos fármacos , Locomoção/efeitos dos fármacos , Locomoção/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microinjeções/métodos , Neurônios Serotoninérgicos/efeitos dos fármacos , Agonistas do Receptor 5-HT2 de Serotonina/administração & dosagem , Antagonistas do Receptor 5-HT2 de Serotonina/administração & dosagem , Transdução de Sinais/efeitos dos fármacos
20.
Addict Biol ; 25(1): e12723, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-30734456

RESUMO

In drug addiction, environmental stimuli previously associated with cocaine use readily elicit cocaine-associated memories, which persist long after abstinence and trigger cocaine craving and consumption. Although previous studies suggest that the medial prefrontal cortex (mPFC) is involved in the expression of cocaine-addictive behaviors, it remains unclear whether excitatory and inhibitory neurons in the mPFC are causally related to the formation and retrieval of cocaine-associated memories. To address this issue, we used the designer receptors exclusively activated by designer drugs (DREADD) technology combined with a cocaine-induced conditioned place preference (CPP) paradigm. We suppressed mPFC neuronal activity in a cell-type- and timing-dependent manner. C57BL/6J wild-type mice received bilateral intra-mPFC infusion of an adeno-associated virus (AAV) expressing inhibitory DREADD (hM4Di) under the control of CaMKII promotor to selectively suppress mPFC pyramidal neurons. GAD67-Cre mice received bilateral intra-mPFC infusion of a Cre-dependent AAV expressing hM4Di to specifically silence GABAergic neurons. Chemogenetic suppression of mPFC pyramidal neurons significantly attenuated both the acquisition and expression of cocaine CPP, while suppression of mPFC GABAergic neurons affected neither the acquisition nor expression of cocaine CPP. Moreover, chemogenetic inhibition of mPFC glutamatergic neurons did not affect the acquisition and expression of lithium chloride-induced conditioned place aversion. These results suggest that the activation of glutamatergic, but not GABAergic, neurons in the mPFC mediates both the formation and retrieval of cocaine-associated memories.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/fisiopatologia , Cocaína/farmacologia , Neurônios GABAérgicos/efeitos dos fármacos , Memória/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/fisiopatologia , Animais , Modelos Animais de Doenças , Inibidores da Captação de Dopamina/farmacologia , Eletrofisiologia , Imunofluorescência , Masculino , Camundongos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA