Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 484
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Acta Neuropsychiatr ; 33(3): 148-155, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33593455

RESUMEN

OBJECTIVE: Whereas numerous experimental and clinical studies suggest a complex involvement of serotonin in the regulation of anxiety, it remains to be clarified if the dominating impact of this transmitter is best described as anxiety-reducing or anxiety-promoting. The aim of this study was to assess the impact of serotonin depletion on acquisition, consolidation, and expression of conditioned fear. METHODS: Male Sprague-Dawley rats were exposed to foot shocks as unconditioned stimulus and assessed with respect to freezing behaviour when re-subjected to context. Serotonin depletion was achieved by administration of a serotonin synthesis inhibitor, para-chlorophenylalanine (PCPA) (300 mg/kg daily × 3), (i) throughout the period from (and including) acquisition to (and including) expression, (ii) during acquisition but not expression, (iii) after acquisition only, and (iv) during expression only. RESULTS: The time spent freezing was significantly reduced in animals that were serotonin-depleted during the entire period from (and including) acquisition to (and including) expression, as well as in those being serotonin-depleted during either acquisition only or expression only. In contrast, PCPA administrated immediately after acquisition, that is during memory consolidation, did not impact the expression of conditioned fear. CONCLUSION: Intact serotonergic neurotransmission is important for both acquisition and expression of context-conditioned fear.


Asunto(s)
Miedo/efectos de los fármacos , Fenclonina/farmacología , Antagonistas de la Serotonina/farmacología , Serotonina/metabolismo , Animales , Ansiedad/metabolismo , Conducta Animal/efectos de los fármacos , Condicionamiento Psicológico , Modelos Animales de Enfermedad , Miedo/psicología , Fenclonina/administración & dosificación , Reacción Cataléptica de Congelación/efectos de los fármacos , Masculino , Ratas , Ratas Sprague-Dawley , Serotonina/deficiencia , Antagonistas de la Serotonina/administración & dosificación
2.
Synapse ; 74(12): e22180, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32644234

RESUMEN

Pharmacological magnetic resonance imaging (phMRI) allows the visualization of brain pharmacological effects of drugs using functional MRI (fMRI). phMRI can help us facilitate central nervous system (CNS) drug development. However, there have been few studies demonstrating the dose relationship of the fMRI response induced by CNS drugs to underlying target engagement or behavioral efficacy. To clarify these relationships, we examined receptor occupancy measurements using positron emission tomography (PET) (n = 3~5), fMRI (n = 5~8) and a cataleptic behavior (n = 6) with raclopride, a dopamine D2 receptor antagonist (8, 20, and 200 µg/kg) on Wistar rats. Dopamine D2 receptor occupancy was increased dose dependently by raclopride (41.8 ± 2.7%, 8 µg/kg; 64.9 ± 2.8%, 20 µg/kg; 83.1 ± 3.0%, 200 µg/kg). phMRI study revealed significant positive responses to raclopride at 200 µg/kg specifically in the striatum and nucleus accumbens, related to dopaminergic system. Slight fMRI responses were observed at 20 µg/kg in some areas corresponding to the striatum and nucleus accumbens. There were no noticeable fMRI responses at 8 µg/kg raclopride administration. Raclopride at 200 µg/kg significantly increased the cataleptic score, although, at 8 and 20 µg/kg, raclopride had no significant effects. These findings showed that raclopride-induced fMRI responses were observed at doses inducing cataleptic behavior and high D2 receptor occupancy, suggesting that phMRI can be useful for dose selection in clinical trial as an evaluation method of brain activity, which reflects behavioral responses induced by target engagements.


Asunto(s)
Cuerpo Estriado/metabolismo , Antagonistas de Dopamina/farmacocinética , Reacción Cataléptica de Congelación/efectos de los fármacos , Núcleo Accumbens/metabolismo , Racloprida/farmacocinética , Animales , Cuerpo Estriado/diagnóstico por imagen , Cuerpo Estriado/fisiología , Imagen por Resonancia Magnética , Masculino , Núcleo Accumbens/diagnóstico por imagen , Núcleo Accumbens/fisiología , Tomografía de Emisión de Positrones , Unión Proteica , Ratas , Ratas Wistar , Receptores de Dopamina D2/metabolismo
3.
Horm Behav ; 124: 104777, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32439347

RESUMEN

Neuropeptide oxytocin (OT) is involved in the regulation of social and non-social behaviour. The central nucleus of amygdala (CeA), part of the limbic system, plays an important role in learning, memory, anxiety and reinforcing mechanisms. CeA has been shown to be rich in OT receptors in rodents. Our previous findings indicated that OT in the rat CeA has a dose dependent rewarding and anxiolytic effect. The aim of our present study was to examine in the CeA the possible interaction of OT and D2 dopamine (DA) receptor antagonist Sulpiride on reinforcement in place preference test and on anxiety in elevated plus maze test. Wistar rats were microinjected bilaterally with 10 ng OT. In different group of animals 4 µg D2 DA receptor antagonist was applied. Other animals received D2 DA receptor antagonist 15 min before 10 ng OT treatment or vehicle solution into the CeA. Rats receiving 10 ng OT spent significantly longer time in the treatment quadrant during the test session in conditioned place preference test. Prior treatment with D2 DA receptor antagonist blocked the rewarding effects of OT. Antagonist in itself did not influence the time rats spent in the treatment quadrant. In elevated plus maze test, rats receiving 10 ng OT spent significantly longer time on the open arms. Prior treatment with D2 DA receptor antagonist blocked the effects of OT. Our results show that DA system plays a role in positive reinforcing and anxiolytic effects of OT because D2 DA receptor antagonist can block these actions.


Asunto(s)
Ansiolíticos/farmacología , Oxitocina/farmacología , Receptores de Dopamina D2/fisiología , Conducta Espacial/efectos de los fármacos , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Ansiedad/tratamiento farmacológico , Conducta Animal/efectos de los fármacos , Condicionamiento Clásico/efectos de los fármacos , Antagonistas de los Receptores de Dopamina D2/farmacología , Miedo/efectos de los fármacos , Reacción Cataléptica de Congelación/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratas , Ratas Wistar , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores de Oxitocina/metabolismo , Receptores de Oxitocina/fisiología , Refuerzo en Psicología , Recompensa , Sulpirida/farmacología
4.
Horm Behav ; 119: 104651, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31790664

RESUMEN

The estrogen receptor (ER) mechanisms by which 17ß-estradiol influences depressive-like behaviour have primarily been investigated acutely and not within an animal model of depression. Therefore, the current study aimed to dissect the contribution of ERα and ERß to the effects of 17ß-estradiol under non-stress and chronic stress conditions. Ovariectomized (OVX) or sham-operated mice were treated chronically (47 days) with 17ß-estradiol (E2), the ERß agonist diarylpropionitrile (DPN), the ERα agonist propylpyrazole-triol (PPT), or vehicle. On day 15 of treatment, mice from each group were assigned to chronic unpredictable stress (CUS; 28 days) or non-CUS conditions. Mice were assessed for anxiety- and depressive-like behaviour and hypothalamic-pituitary-adrenal (HPA) axis function. Cytokine and chemokine levels, and postsynaptic density protein 95 were measured in the hippocampus and frontal cortex, and adult hippocampal neurogenesis was assessed. Overall, the effects of CUS were more robust that those of estrogenic treatments, as seen by increased immobility in the tail suspension test (TST), reduced PSD-95 expression, reduced neurogenesis in the ventral hippocampus, and HPA axis negative feedback dysregulation. However, we also observe CUS-dependent and -independent effects of ovarian status and estrogenic treatments. The effects of CUS on PSD-95 expression, the cytokine milieu, and in TST were largely driven by PPT and DPN, indicating that these treatments were not protective. Independent of CUS, estradiol increased neurogenesis in the dorsal hippocampus, blunted the corticosterone response to an acute stressor, and increased anxiety-like behaviour. These findings provide insights into the complexities of estrogen signaling in modulating depressive-like phenotypes under non-stress and chronic stress conditions.


Asunto(s)
Depresión/metabolismo , Receptor alfa de Estrógeno/agonistas , Receptor beta de Estrógeno/agonistas , Estrés Psicológico/metabolismo , Animales , Enfermedad Crónica , Corticosterona/metabolismo , Depresión/etiología , Depresión/psicología , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Femenino , Reacción Cataléptica de Congelación/efectos de los fármacos , Reacción Cataléptica de Congelación/fisiología , Hipocampo/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/metabolismo , Ratones , Ratones Endogámicos C57BL , Nitrilos/farmacología , Ovariectomía , Fenoles/farmacología , Fenotipo , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/metabolismo , Propionatos/farmacología , Pirazoles/farmacología , Estrés Psicológico/complicaciones , Estrés Psicológico/patología , Estrés Psicológico/psicología
5.
Dev Psychobiol ; 62(3): 380-385, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31621064

RESUMEN

The ontogeny and NMDA-receptor (NMDAR) mechanisms of context conditioning were examined during standard contextual fear conditioning (sCFC) - involving context and context-shock learning in the same trial - as a comparison with our previous reports on the Context Preexposure Facilitation Effect (CPFE), which separates these two types of learning by 24 hr. In Experiment 1, systemic administration of the NMDAR antagonist, MK-801, prior to conditioning disrupted retention but not post-shock freezing during sCFC in PD31 rats. Experiment 2 replicated and extended this effect to PD17 versus PD31 rats. Consistent with Experiment 1, pre-training MK-801 spared post-shock freezing but impaired retention freezing in PD31 rats. In contrast, pre-training MK-801 disrupted post-shock freezing in PD17 rats, which showed no retention freezing regardless of drug. These results reveal developmental differences in the role of NMDAR activity in the acquisition versus retention of a context-shock association during sCFC in pre-weanling and adolescent rats.


Asunto(s)
Conducta Animal/fisiología , Condicionamiento Clásico/fisiología , Antagonistas de Aminoácidos Excitadores/farmacología , Miedo/fisiología , Receptores de N-Metil-D-Aspartato/fisiología , Retención en Psicología/fisiología , Animales , Conducta Animal/efectos de los fármacos , Condicionamiento Clásico/efectos de los fármacos , Maleato de Dizocilpina/farmacología , Miedo/efectos de los fármacos , Femenino , Reacción Cataléptica de Congelación/efectos de los fármacos , Reacción Cataléptica de Congelación/fisiología , Masculino , Ratas , Ratas Long-Evans , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Retención en Psicología/efectos de los fármacos
6.
Molecules ; 25(4)2020 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-32102354

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors with three isoforms (PPARα, PPARß/δ, PPARγ) and can regulate pain, anxiety, and cognition. However, their role in conditioned fear and pain-fear interactions has not yet been investigated. Here, we investigated the effects of systemically administered PPAR antagonists on formalin-evoked nociceptive behaviour, fear-conditioned analgesia (FCA), and conditioned fear in the presence of nociceptive tone in rats. Twenty-three and a half hours following fear conditioning to context, male Sprague-Dawley rats received an intraplantar injection of formalin and intraperitoneal administration of vehicle, PPARα (GW6471), PPARß/δ (GSK0660) or PPARγ (GW9662) antagonists, and 30 min later were re-exposed to the conditioning arena for 15 min. The PPAR antagonists did not alter nociceptive behaviour or fear-conditioned analgesia. The PPARα and PPARß/δ antagonists prolonged context-induced freezing in the presence of nociceptive tone without affecting its initial expression. The PPARγ antagonist potentiated freezing over the entire trial. In conclusion, pharmacological blockade of PPARα and PPARß/δ in the presence of formalin-evoked nociceptive tone, impaired short-term, within-trial fear-extinction in rats without affecting pain response, while blockade of PPARγ potentiated conditioned fear responding. These results suggest that endogenous signalling through these three PPAR isoforms may reduce the expression of conditioned fear in the presence of nociceptive tone.


Asunto(s)
Condicionamiento Psicológico/efectos de los fármacos , Miedo/efectos de los fármacos , Dolor Nociceptivo/tratamiento farmacológico , PPAR alfa/genética , PPAR delta/genética , PPAR gamma/genética , PPAR-beta/genética , Analgesia/métodos , Anilidas/farmacología , Animales , Extinción Psicológica/efectos de los fármacos , Formaldehído/administración & dosificación , Reacción Cataléptica de Congelación/efectos de los fármacos , Expresión Génica , Masculino , Dolor Nociceptivo/inducido químicamente , Dolor Nociceptivo/fisiopatología , Dolor Nociceptivo/psicología , Oxazoles/farmacología , PPAR alfa/antagonistas & inhibidores , PPAR alfa/metabolismo , PPAR delta/antagonistas & inhibidores , PPAR delta/metabolismo , PPAR gamma/antagonistas & inhibidores , PPAR gamma/metabolismo , PPAR-beta/antagonistas & inhibidores , PPAR-beta/metabolismo , Ratas , Ratas Sprague-Dawley , Sulfonas/farmacología , Tiofenos/farmacología , Tirosina/análogos & derivados , Tirosina/farmacología
7.
Learn Mem ; 26(7): 245-251, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31209119

RESUMEN

Vagus nerve stimulation (VNS) enhances extinction of conditioned fear in rats. Previous findings support the hypothesis that VNS effects on extinction are due to enhanced consolidation of extinction memories through promotion of plasticity in extinction-related brain pathways however, alternative explanations are plausible. According to one hypothesis, VNS may produce a hedonic effect and enhance extinction through counter-conditioning. According to another hypothesis, VNS reduces anxiety during exposure and this weakens the association of conditioned stimuli with aversive conditioned responses. The present set of experiments (1) used conditioned place preference (CPP) to identify potential rewarding effects associated with VNS and (2) examined the peripheral effects of VNS on anxiety and extinction enhancement. Male Sprague-Dawley rats were surgically implanted with cuff electrodes around the vagus nerve and subjected to a CPP task in which VNS and sham stimulation were each paired with one of two distinct contexts over the course of 5 d. Following this procedure, rats did not show a place preference, suggesting that VNS is not rewarding or aversive. The role of the peripheral parasympathetic system in the anxiolytic effect of VNS on the elevated plus maze was examined by blocking peripheral muscarinic receptors with intraperitoneal administration of methyl scopolamine prior to VNS. Methyl scopolamine blocked the VNS-induced reduction in anxiety but did not interfere with VNS enhancement of extinction of conditioned fear, indicating that the anxiety-reducing effect of VNS is not necessary for the extinction enhancement.


Asunto(s)
Ansiedad/fisiopatología , Extinción Psicológica/fisiología , Miedo/fisiología , Sistema Nervioso Parasimpático/fisiopatología , Estimulación del Nervio Vago , Animales , Ansiedad/tratamiento farmacológico , Condicionamiento Clásico/fisiología , Vías Eferentes/fisiología , Electrodos Implantados , Electrochoque , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Reacción Cataléptica de Congelación/efectos de los fármacos , Reacción Cataléptica de Congelación/fisiología , Masculino , Aprendizaje por Laberinto/fisiología , Modelos Neurológicos , Modelos Psicológicos , Antagonistas Muscarínicos/farmacología , Antagonistas Muscarínicos/uso terapéutico , N-Metilescopolamina/farmacología , N-Metilescopolamina/uso terapéutico , Ratas , Ratas Sprague-Dawley , Receptores Muscarínicos/fisiología
8.
Neurobiol Learn Mem ; 166: 107088, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31513850

RESUMEN

Female reproductive experience has been shown to alter the hormonal, neurobiological and behavioural features of fear extinction, which is the laboratory basis of exposure therapy. This raises uncertainties as to whether pharmacological agents that enhance fear extinction in reproductively inexperienced females are equally effective in reproductively experienced females. The aim of the current study was therefore to compare the effects of two pharmacological enhancers of fear extinction, d-cycloserine (DCS) and estradiol, between nulliparous (virgin) and primiparous (reproductively experienced) female rats. In Experiment 1, nulliparous and primiparous females received systemic administration of either DCS or saline immediately after extinction training, and were tested for extinction recall the following day. DCS enhanced extinction recall in nulliparous females that showed low levels of freezing at the end of extinction training, but not among those that showed high levels of freezing at the end of extinction training. DCS did not enhance fear extinction in primiparous females, regardless of their level of freezing at the end of extinction training. In Experiment 2, nulliparous and primiparous female rats received systemic administration of either estradiol or vehicle prior to extinction training. Estradiol enhanced extinction recall among nulliparous females, but not primiparous females. Increasing the dose of estradiol administered prior to extinction training did not alter the outcomes in primiparous females (Experiment 3). Together, these findings suggest that reproductive status may be an important individual difference factor associated with the response to pharmacological modulators of extinction in rats. The implications of these findings for the pharmacological augmentation of exposure therapy in clinical populations are discussed.


Asunto(s)
Cicloserina/farmacología , Estradiol/farmacología , Extinción Psicológica/efectos de los fármacos , Miedo/efectos de los fármacos , Paridad , Animales , Condicionamiento Clásico/efectos de los fármacos , Femenino , Reacción Cataléptica de Congelación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
9.
Stem Cells ; 36(8): 1273-1285, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29726088

RESUMEN

The adult neurogenesis occurs throughout the life of the mammalian hippocampus and is found to be essential for learning and memory. Identifying new ways to manipulate the number of neural stem cells (NSCs) and enhance endogenous neurogenesis in adults is very important. Here we found that a novel compound, N2-(4-isopropylphenyl)-5-(3-methoxyphenoxy)quinazoline-2,4-diamine (code-named Yhhu-3792), enhanced the self-renewal capability of NSCs in vitro and in vivo. In vitro, Yhhu-3792 increased the ratio of 5-Bromo-2-deoxyuridine+ /4'-6-diamidino-2-phenylindole+ embryonic NSCs and accelerated the growth of neurospheres significantly. We demonstrated that Yhhu-3792 activated Notch signaling pathway and promoted the expression of Notch target genes, Hes3 and Hes5. And the Notch signaling inhibitor DAPT could inhibit its function. Thus, we concluded Yhhu-3792 increased the number of embryonic NSCs via activating the Notch signaling pathway. We measured the effect of Yhhu-3792 on epidermal growth factor receptor signaling, which demonstrated Yhhu-3792 act via a different mechanism with the quinazoline parent chemical group. In the eight-week-old male C57BL/6 mice, chronic Yhhu-3792 administration expanded the NSCs pool and promoted endogenous neurogenesis in the hippocampal dentate gyrus (DG). It also increased the spatial and episodic memory abilities of mice, when evaluated with the Morris water maze and Fear conditioning tests. In conclusion, Yhhu-3792 could be a novel drug candidate to promote the self-renew of NSCs and adult neurogenesis. And it may have therapeutic potential in the impairment of learning and memory associated DG dysfunction. Stem Cells 2018;36:1273-1285.


Asunto(s)
Cognición/fisiología , Hipocampo/fisiología , Células-Madre Neurales/citología , Neurogénesis/efectos de los fármacos , Quinazolinas/farmacología , Animales , Recuento de Células , Proliferación Celular/efectos de los fármacos , Autorrenovación de las Células/efectos de los fármacos , Cognición/efectos de los fármacos , Condicionamiento Psicológico , Dipéptidos/farmacología , Embrión de Mamíferos/citología , Miedo , Reacción Cataléptica de Congelación/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones Endogámicos C57BL , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Quinazolinas/química , Tiempo de Reacción/efectos de los fármacos , Receptores Notch/metabolismo , Transducción de Señal/efectos de los fármacos
10.
Pharmacology ; 103(1-2): 68-75, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30513516

RESUMEN

The Medial Habenular (MHb) and the Lateral Habenular nuclei are 2 main parts of the habenular complex (Hb). Recent studies showed that MHb plays an important role in memory, and in the expression of ErbB4. However, the expression of MHb ErbB4 receptor and its role in fear memory is not well understood. In this study, western blotting and quantitative real-time polymerase chain reaction were used to assess the protein and mRNA levels of ErbB4 in the process of contextual fear conditioning. A pharmacological approach was used to block and stimulate the ErbB4 receptor. Contextual fear conditioning tests induced a significant increase on the expression of ErbB4 at various times in the Hb and the MHb. Moreover, the blockade and stimulation of MHb ErbB4 receptors did not affect the fear formation but impaired and improved the contextual-dependent fear expression. Furthermore, in vitro electrophysiological recordings showed that the blockade of the MHb ErbB4 receptor reduced the presynaptic gamma-amino butyric acid release. ErbB4 is a susceptible gene for schizophrenia and the above findings may provide new insights into the mechanisms of fear-related responses.


Asunto(s)
Miedo/fisiología , Habénula/metabolismo , Memoria/fisiología , Receptor ErbB-4/metabolismo , Animales , Escala de Evaluación de la Conducta , Condicionamiento Clásico , Miedo/psicología , Reacción Cataléptica de Congelación/efectos de los fármacos , Habénula/efectos de los fármacos , Habénula/fisiología , Masculino , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Potenciales Postsinápticos Miniatura/efectos de los fármacos , Neurregulina-1/farmacología , Pirimidinas/farmacología , Quinazolinas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor ErbB-4/agonistas , Receptor ErbB-4/antagonistas & inhibidores , Receptor ErbB-4/genética , Tirfostinos/farmacología
11.
Proc Natl Acad Sci U S A ; 113(17): 4830-5, 2016 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-27078100

RESUMEN

Stressful events evoke long-term changes in behavioral responses; however, the underlying mechanisms in the brain are not well understood. Previous work has shown that epigenetic changes and immediate-early gene (IEG) induction in stress-activated dentate gyrus (DG) granule neurons play a crucial role in these behavioral responses. Here, we show that an acute stressful challenge [i.e., forced swimming (FS)] results in DNA demethylation at specific CpG (5'-cytosine-phosphate-guanine-3') sites close to the c-Fos (FBJ murine osteosarcoma viral oncogene homolog) transcriptional start site and within the gene promoter region of Egr-1 (early growth response protein 1) specifically in the DG. Administration of the (endogenous) methyl donor S-adenosyl methionine (SAM) did not affect CpG methylation and IEG gene expression at baseline. However, administration of SAM before the FS challenge resulted in an enhanced CpG methylation at the IEG loci and suppression of IEG induction specifically in the DG and an impaired behavioral immobility response 24 h later. The stressor also specifically increased the expression of the de novo DNA methyltransferase Dnmt3a [DNA (cytosine-5-)-methyltransferase 3 alpha] in this hippocampus region. Moreover, stress resulted in an increased association of Dnmt3a enzyme with the affected CpG loci within the IEG genes. No effects of SAM were observed on stress-evoked histone modifications, including H3S10p-K14ac (histone H3, phosphorylated serine 10 and acetylated lysine-14), H3K4me3 (histone H3, trimethylated lysine-4), H3K9me3 (histone H3, trimethylated lysine-9), and H3K27me3 (histone H3, trimethylated lysine-27). We conclude that the DNA methylation status of IEGs plays a crucial role in FS-induced IEG induction in DG granule neurons and associated behavioral responses. In addition, the concentration of available methyl donor, possibly in conjunction with Dnmt3a, is critical for the responsiveness of dentate neurons to environmental stimuli in terms of gene expression and behavior.


Asunto(s)
Metilación de ADN , Giro Dentado/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Regulación de la Expresión Génica , Genes fos , S-Adenosilmetionina/farmacología , Estrés Fisiológico/genética , Estrés Psicológico/genética , Animales , Islas de CpG , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/efectos de los fármacos , ADN Metiltransferasa 3A , Giro Dentado/efectos de los fármacos , Reacción Cataléptica de Congelación/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Genes Inmediatos-Precoces/efectos de los fármacos , Código de Histonas/efectos de los fármacos , Masculino , Regiones Promotoras Genéticas/genética , Ratas , Ratas Wistar , Natación
12.
Acta Neuropsychiatr ; 31(1): 46-51, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30404671

RESUMEN

OBJECTIVE: Whereas long-term administration of selective serotonin reuptake inhibitors (SSRIs) is effective for the treatment of anxiety disorders, acute administration of these drugs may exert a paradoxical anxiogenic effect. The aim of the present study was to explore the possible effect of an SSRI in situations of unconditioned or limited conditioned fear. METHODS: Male Sprague Dawley rats were administered a single dose of an SSRI, escitalopram, before acquisition or expression of context conditioned fear, where noise bursts were used as the unconditioned stimulus. Freezing was assessed as a measure of unconditioned fear (=the acute response to noise bursts) or conditioned fear (=the response to the context), respectively. RESULTS: Noise bursts elicited an acute increase in freezing but no robust conditioned response 7 days after exposure. Administration of escitalopram before testing exacerbated the freezing response during presentation of the unconditioned stimulus and also unmasked a conditioned response; in contrast, administration of escitalopram prior to acquisition did not influence the conditioned response. CONCLUSION: The data suggest that freezing in rats exposed to a stimulus inducing relatively mild fear may be enhanced by acute pretreatment with an SSRI regardless of whether the freezing displayed by the animals is an acute unconditioned response to the stimulus in question or a conditioned response to the same stimulus.


Asunto(s)
Conducta Animal/efectos de los fármacos , Citalopram/farmacología , Condicionamiento Clásico/efectos de los fármacos , Miedo/efectos de los fármacos , Reacción Cataléptica de Congelación/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Animales , Citalopram/administración & dosificación , Masculino , Ruido , Ratas , Ratas Sprague-Dawley , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación
13.
Neurobiol Learn Mem ; 149: 68-76, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29432803

RESUMEN

Cannabidiol (CBD) is thought to have therapeutic potential for treating psychiatric conditions that affect cognitive aspects of learning and memory, including anxiety and post-traumatic stress disorder (PTSD). Studies have shown that CBD enhances extinction of fear memory when given after conditioning. This led us to hypothesize that CBD, if administered prior to fear conditioning, might modulate cognitive learning and memory processes in additional ways that would further guide its potential use for treating PTSD. Therefore, we designed a study to investigate effects of CBD on fear learning and memory when administered to mice prior to administering a trace fear conditioning protocol which imposes cognitive demands on the learning and memory process. We show that CBD-treated animals had increased levels of freezing during conditioning, enhanced generalized fear, inhibited cue-dependent memory extinction, slightly increased levels of freezing during an auditory-cued memory test, and increased contextual fear memory. Because synaptic plasticity is the fundamental mechanism of learning and memory, we also evaluated the impact of CBD on trace conditioning-dependent dendritic spine plasticity which occurred in the dorsal lateral amygdala and CA1 region of the ventral hippocampus. We showed that CBD mildly enhanced spine densities independent of conditioning, and inhibited conditioning-dependent spine increases in the hippocampi, but not the amygdala of fear conditioned animals. Overall, the memory-modulating effects of a single pre-conditioning dose of CBD, which we show here, demonstrate the need to more fully characterize its basic effects on memory, suggest caution when using it clinically as an anxiolytic, and point to a need for more research into its potential as a therapeutic for treating memory-loss disorders.


Asunto(s)
Aprendizaje por Asociación/efectos de los fármacos , Cannabidiol/farmacología , Condicionamiento Clásico/efectos de los fármacos , Miedo/efectos de los fármacos , Memoria/efectos de los fármacos , Animales , Espinas Dendríticas/efectos de los fármacos , Extinción Psicológica/efectos de los fármacos , Reacción Cataléptica de Congelación/efectos de los fármacos , Hipocampo/efectos de los fármacos , Masculino , Ratones
14.
Nat Chem Biol ; 12(7): 552-8, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27239788

RESUMEN

Humans and many animals show 'freezing' behavior in response to threatening stimuli. In humans, inappropriate threat responses are fundamental characteristics of several mental illnesses. To identify small molecules that modulate threat responses, we developed a high-throughput behavioral assay in zebrafish (Danio rerio) and evaluated 10,000 compounds for their effects on freezing behavior. We found three classes of compounds that switch the threat response from freezing to escape-like behavior. We then screened these for binding activity across 45 candidate targets. Using target profile clustering, we identified the sigma-1 (σ1) receptor as having a role in the mechanism of behavioral switching and confirmed that known σ1 ligands also disrupt freezing behavior. Furthermore, mutation of the gene encoding σ1 prevented the behavioral effect of escape-inducing compounds. One compound, which we call finazine, potently bound mammalian σ1 and altered threat-response behavior in mice. Thus, pharmacological and genetic interrogation of the freezing response revealed σ1 as a mediator of threat responses in vertebrates.


Asunto(s)
Reacción de Fuga/efectos de los fármacos , Reacción Cataléptica de Congelación/efectos de los fármacos , Larva/efectos de los fármacos , Receptores sigma/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Pez Cebra , Anilidas/química , Anilidas/metabolismo , Anilidas/farmacología , Animales , Reacción de Fuga/efectos de la radiación , Reacción Cataléptica de Congelación/efectos de la radiación , Ensayos Analíticos de Alto Rendimiento , Larva/efectos de la radiación , Ligandos , Luz , Ratones , Estructura Molecular , Piperazinas/química , Piperazinas/metabolismo , Piperazinas/farmacología , Receptores sigma/genética , Bibliotecas de Moléculas Pequeñas/química , Pez Cebra/crecimiento & desarrollo , Receptor Sigma-1
15.
Synapse ; 72(8): e22035, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29704283

RESUMEN

Post-traumatic stress disorder (PTSD) is a debilitating psychiatric syndrome that occurs in individuals exposed to extremely threatening or traumatic events. In both animals and humans, dopamine (DA) function appears to be dysregulated in brain areas involved in the conditioned fear response(s) that underlie PTSD. In this study, we determined the effect of the selective DA D3 receptor antagonists YQA14A (6.25, 12.5 and 25 mg/kg i.p.) and SB-277011A (6 mg/kg i.p.) on tone-induced fear (assessed by measuring freeze time) in a modified version of the single-prolonged stress (SPS) model of PTSD in adult male Sprague-Dawley rats. Rats pretreated with vehicle and then subjected to restraint stress, forced swim and random foot shock (SPS) in the presence of a distinctive tone, displayed a significantly increased tone-induced contextual freeze time and fecal pellet mass following re-exposure to the tone. Rats pretreated with a single i.p. injection of 6.25 or 12.5 mg/kg of YQA14 or 6 mg/kg of SB-277011A showed significantly attenuated contextual freeze time in the presence of the tone when tested 14 days after exposure to SPS. Overall, our results indicate that selectively antagonizing DA D3 receptors significantly decreases freezing time caused by an environment previously associated with stress. If our findings can be extrapolated to humans with PTSD, they suggest that DA D3 receptors may play a role in the pathophysiology of PTSD, and may have therapeutic utility for the clinical management of PTSD.


Asunto(s)
Benzoxazoles/farmacología , Nitrilos/farmacología , Piperazinas/farmacología , Psicotrópicos/farmacología , Receptores de Dopamina D3/antagonistas & inhibidores , Trastornos por Estrés Postraumático/tratamiento farmacológico , Tetrahidroisoquinolinas/farmacología , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Heces , Reacción Cataléptica de Congelación/efectos de los fármacos , Reacción Cataléptica de Congelación/fisiología , Masculino , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Distribución Aleatoria , Ratas Sprague-Dawley , Receptores de Dopamina D3/metabolismo , Trastornos por Estrés Postraumático/metabolismo , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/metabolismo
16.
Horm Behav ; 101: 113-124, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29107581

RESUMEN

Due of its structural similarity to the endogenous estrogen 17ß-estradiol (E2), the synthetic estrogen 17α-ethinyl estradiol (EE2) is widely used to study the effects of estrogenic substances on sensitive organs at multiple stages of development. Here, we investigated the effects of EE2 on maternal behavior and the maternal brain in females exposed during gestation and the perinatal period. We assessed several components of maternal behavior including nesting behavior and pup retrieval; characterized the expression of estrogen receptor (ER)α in the medial preoptic area (MPOA), a brain region critical for the display of maternal behavior; and measured expression of tyrosine hydroxylase, a marker for dopaminergic cells, in the ventral tegmental area (VTA), a brain region important in maternal motivation. We found that developmental exposure to EE2 induces subtle effects on several aspects of maternal behavior including time building the nest and time spent engaged in self-care. Developmental exposure to EE2 also altered ERα expression in the central MPOA during both early and late lactation and led to significantly reduced tyrosine hydroxylase immunoreactivity in the VTA. Our results demonstrate both dose- and postpartum stage-related effects of developmental exposure to EE2 on behavior and brain that manifest later in adulthood, during the maternal period. These findings provide further evidence for effects of exposure to exogenous estrogenic compounds during the critical periods of fetal and perinatal development.


Asunto(s)
Encéfalo/efectos de los fármacos , Disruptores Endocrinos/farmacología , Estrógenos/farmacología , Etinilestradiol/farmacología , Conducta Materna/efectos de los fármacos , Comportamiento de Nidificación/efectos de los fármacos , Animales , Encéfalo/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Disruptores Endocrinos/toxicidad , Femenino , Reacción Cataléptica de Congelación/efectos de los fármacos , Aseo Animal/efectos de los fármacos , Lactancia/efectos de los fármacos , Ratones , Embarazo
17.
Horm Behav ; 100: 20-28, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29501756

RESUMEN

The basolateral nucleus of the amygdala (BLA) plays a significant role in mediating individual differences in the effects of fear memory on sleep. Here, we assessed the effects of antagonizing corticotropin releasing factor receptor 1 (CRFR1) after shock training (ST) on fear-conditioned behaviors and sleep. Outbred Wistar rats were surgically implanted with electrodes for recording EEG and EMG and with bilateral guide cannulae directed at BLA. Data loggers were placed intraperitoneally to record core body temperature. The CRFR1 antagonist, antalarmin (ANT; 4.82 mM) was microinjected into BLA after shock training (ST: 20 footshocks, 0.8 mA, 0.5 s duration, 60 s interstimulus interval), and the effects on sleep, freezing and the stress response (stress-induced hyperthermia, SIH) were examined after ST and fearful context re-exposure alone at 7 days (CTX1) and 21 days (CTX2) post-ST. EEG and EMG recordings were scored for non-rapid eye movement sleep (NREM), rapid eye movement sleep (REM) and wakefulness. The rats were separated into 4 groups: Vehicle-vulnerable (Veh-Vul; n = 10), Veh-resilient (Veh-Res; n = 11), ANT-vulnerable (ANT-Vul; n = 8) and ANT-resilient (ANT-Res; n = 8) based on whether, compared to baseline, the rats showed a decrease or no change/increase in REM during the first 4 h following ST. Post-ST ANT microinjected into BLA attenuated the fear-conditioned reduction in REM in ANT-Vul rats on CTX1, but did not significantly alter REM in ANT-Res rats. However, compared to Veh treated rats, REM was reduced in ANT treated rats on CTX2. There were no group differences in freezing or SIH across conditions. Therefore, CRFR1 in BLA plays a role in mediating individual differences in sleep responses to stress and in the extinction of fear conditioned changes in sleep.


Asunto(s)
Adaptación Psicológica/efectos de los fármacos , Complejo Nuclear Basolateral/efectos de los fármacos , Temperatura Corporal/efectos de los fármacos , Miedo/efectos de los fármacos , Reacción Cataléptica de Congelación/efectos de los fármacos , Pirimidinas/farmacología , Pirroles/farmacología , Sueño/efectos de los fármacos , Animales , Complejo Nuclear Basolateral/metabolismo , Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Electroencefalografía , Miedo/psicología , Masculino , Memoria/efectos de los fármacos , Condicionamiento Físico Animal/fisiología , Condicionamiento Físico Animal/psicología , Ratas , Ratas Wistar , Sueño/fisiología , Sueño REM/efectos de los fármacos
18.
Metab Brain Dis ; 33(4): 1175-1185, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29633071

RESUMEN

Post-traumatic stress disorder (PTSD) is the serious psychiatric disorder. Paeoniflorin (PF) produces the antidepressant-like properties. However, few studies are concerned about its anti-PTSD-like effects and mechanisms. To investigate these, the single prolonged stress (SPS) model was utilized. PTSD-like behavioral deficits in rats after exposure to SPS were improved by PF (10 and 20 mg/kg, i.p.), evidenced by blocking increased freezing time in contextual fear paradigm (CFP) and increased time and entries in open arms in elevated plus maze (EPM) test without affecting the locomotor activity in open field (OF) test. We also found that increased levels of corticosterone (Cort), corticotropin releasing hormone (CRH) and adrenocorticotropic hormone (ACTH) after exposure to SPS were reversed by PF (10 and 20 mg/kg, i.p.) in serum, respectively. Moreover, the decreased levels of serotonin (5-HT) and 5-Hydroxyindoleacetic acid (5-HIAA) in prefrontal cortex and hippocampus were reversed by PF (10 and 20 mg/kg, i.p.), respectively. In summary, the anti-PTSD-like activities of PF were associated with the modulation of HPA axis and 5-HT system activation.


Asunto(s)
Ansiolíticos/uso terapéutico , Conducta Animal/efectos de los fármacos , Reacción Cataléptica de Congelación/efectos de los fármacos , Glucósidos/uso terapéutico , Monoterpenos/uso terapéutico , Trastornos por Estrés Postraumático/tratamiento farmacológico , Hormona Adrenocorticotrópica/sangre , Animales , Ansiolíticos/farmacología , Corticosterona/sangre , Hormona Liberadora de Corticotropina/sangre , Modelos Animales de Enfermedad , Glucósidos/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ácido Hidroxiindolacético/metabolismo , Masculino , Monoterpenos/farmacología , Actividad Motora/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Ratas , Ratas Sprague-Dawley , Serotonina/metabolismo , Trastornos por Estrés Postraumático/metabolismo , Resultado del Tratamiento
19.
Learn Mem ; 24(8): 375-380, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28716957

RESUMEN

Activity-regulated cytoskeleton-associated protein (Arc) supports fear memory through synaptic plasticity events requiring actin cytoskeleton rearrangements. We have previously shown that reducing hippocampal Arc levels through antisense knockdown leads to the premature extinction of contextual fear. Here we show that the AMPA receptor antagonist CNQX elevates hippocampal Arc levels during extinction and blocks extinction that can be rescued by reducing Arc. Increasing Arc levels with CNQX also overcomes the actin-destabilizing properties of cytochalasin D and promotes extinction. Therefore, extinction is dependent on AMPA-mediated reductions of Arc via a mechanism consistent with a role for Arc in stabilizing the actin cytoskeleton to constrain extinction.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Extinción Psicológica/fisiología , Miedo/fisiología , Proteínas del Tejido Nervioso/metabolismo , Receptores AMPA/metabolismo , 6-Ciano 7-nitroquinoxalina 2,3-diona/farmacología , Animales , Condicionamiento Clásico/efectos de los fármacos , Condicionamiento Clásico/fisiología , Citocalasina D/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Extinción Psicológica/efectos de los fármacos , Miedo/efectos de los fármacos , Reacción Cataléptica de Congelación/efectos de los fármacos , Reacción Cataléptica de Congelación/fisiología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Pruebas Neuropsicológicas , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Ratas , Receptores AMPA/antagonistas & inhibidores
20.
Learn Mem ; 24(9): 385-391, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28814463

RESUMEN

Retrieval of fear memories can be state-dependent, meaning that they are best retrieved if the brain states at encoding and retrieval are similar. Such states can be induced by activating extrasynaptic γ-aminobutyric acid type A receptors (GABAAR) with the broad α-subunit activator gaboxadol. However, the circuit mechanisms and specific subunits underlying gaboxadol's effects are not well understood. Here we show that gaboxadol induces profound changes of local and network oscillatory activity, indicative of discoordinated hippocampal-cortical activity, that were accompanied by robust and long-lasting state-dependent conditioned fear. Episodic memories typically are hippocampus-dependent for a limited period after learning, but become cortex-dependent with the passage of time. In contrast, state-dependent memories continued to rely on hippocampal GABAergic mechanisms for memory retrieval. Pharmacological approaches with α-subunit-specific agonists targeting the hippocampus implicated the prototypic extrasynaptic subunits (α4) as the mediator of state-dependent conditioned fear. Together, our findings suggest that continued dependence on hippocampal rather than cortical mechanisms could be an important feature of state-dependent memories that contributes to their conditional retrieval.


Asunto(s)
Condicionamiento Clásico/fisiología , Miedo/fisiología , Recuerdo Mental/fisiología , Animales , Condicionamiento Clásico/efectos de los fármacos , Miedo/efectos de los fármacos , Femenino , Reacción Cataléptica de Congelación/efectos de los fármacos , Agonistas del GABA/farmacología , Hipocampo/efectos de los fármacos , Masculino , Recuerdo Mental/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA