Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 14.814
Filtrar
1.
Behav Neurosci ; 138(2): 125-141, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38661671

RESUMEN

Selenium is an essential trace element that is delivered to the brain by the selenium transport protein selenoprotein P (SEPP1), primarily by binding to its receptor low-density lipoprotein receptor-related protein 8 (LRP8), also known as apolipoprotein E receptor 2 (ApoER2), at the blood-brain barrier. Selenium transport is required for several important brain functions, with transgenic deletion of either Sepp1 or Lrp8 resulting in severe neurological dysfunction and death in mice fed a selenium-deficient diet. Previous studies have reported that although feeding a standard chow diet can prevent these severe deficits, some motor coordination and cognitive dysfunction remain. Importantly, no single study has directly compared the motor and cognitive performance of the Sepp1 and Lrp8 knockout (KO) lines. Here, we report the results of a comprehensive parallel analysis of the motor and spatial learning and memory function of Sepp1 and Lrp8 knockout mice fed a standard mouse chow diet. Our results revealed that Sepp1 knockout mice raised on a selenium-replete diet displayed motor and cognitive function that was indistinguishable from their wild-type littermates. In contrast, we found that although Lrp8-knockout mice fed a selenium-replete diet had normal motor function, their spatial learning and memory showed subtle deficits. We also found that the deficit in baseline adult hippocampal neurogenesis exhibited by Lrp8-deficit mice could not be rescued by dietary selenium supplementation. Taken together, these findings further highlight the importance of selenium transport in maintaining healthy brain function. (PsycInfo Database Record (c) 2024 APA, all rights reserved).


Asunto(s)
Proteínas Relacionadas con Receptor de LDL , Ratones Noqueados , Selenio , Aprendizaje Espacial , Animales , Ratones , Selenio/administración & dosificación , Selenio/deficiencia , Selenio/farmacología , Aprendizaje Espacial/fisiología , Aprendizaje Espacial/efectos de los fármacos , Proteínas Relacionadas con Receptor de LDL/genética , Proteínas Relacionadas con Receptor de LDL/metabolismo , Masculino , Selenoproteína P/genética , Selenoproteína P/metabolismo , Dieta , Memoria Espacial/fisiología , Memoria Espacial/efectos de los fármacos , Ratones Endogámicos C57BL , Hipocampo/metabolismo , Memoria/fisiología , Memoria/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Aprendizaje por Laberinto/efectos de los fármacos
3.
J Biol Chem ; 299(5): 104693, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37037305

RESUMEN

The Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a central regulator of learning and memory, which poses a problem for targeting it therapeutically. Indeed, our study supports prior conclusions that long-term interference with CaMKII signaling can erase pre-formed memories. By contrast, short-term pharmacological CaMKII inhibition with the neuroprotective peptide tatCN19o interfered with learning in mice only mildly and transiently (for less than 1 h) and did not at all reverse pre-formed memories. These results were obtained with ≥500-fold of the dose that protected hippocampal neurons from cell death after a highly clinically relevant pig model of transient global cerebral ischemia: ventricular fibrillation followed by advanced life support and electrical defibrillation to induce the return of spontaneous circulation. Of additional importance for therapy development, our preliminary cardiovascular safety studies in mice and pig did not indicate any concerns with acute tatCN19o injection. Taken together, although prolonged interference with CaMKII signaling can erase memory, acute short-term CaMKII inhibition with tatCN19o did not cause such retrograde amnesia that would pose a contraindication for therapy.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Memoria , Animales , Ratones , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Hipocampo/metabolismo , Memoria/efectos de los fármacos , Memoria/fisiología , Neuronas/metabolismo , Fosforilación/fisiología , Porcinos , Péptidos/farmacología
4.
J Alzheimers Dis ; 92(4): 1413-1426, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36911940

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is a neurodegenerative disorder characterized by amyloid-ß peptide (Aß) deposition. Aß accumulation induces oxidative stress, leading to mitochondrial dysfunction, apoptosis, and so forth. Octadecaneuropeptide (ODN), a diazepam-binding inhibitor (DBI)-derived peptide, has been reported to have antioxidant properties. However, it is unclear whether ODN has neuroprotective effects in AD. OBJECTIVE: To profile the potential effects of ODN on AD. METHODS: We established a mouse model of AD via microinjection of Aß in the lateral ventricle. Utilizing a combination of western blotting assays, electrophysiological recordings, and behavioral tests, we investigated the neuroprotective effects of ODN on AD. RESULTS: DBI expression was decreased in AD model mice and cells. Meanwhile, ODN decreased Aß generation by downregulating amyloidogenic AßPP processing in HEK-293 cells stably expressing human Swedish mutant APP695 and BACE1 (2EB2). Moreover, ODN could inhibit Aß-induced oxidative stress in primary cultured cells and mice, as reflected by a dramatic increase in antioxidants and a decrease in pro-oxidants. We also found that ODN could reduce oxidative stress-induced apoptosis by restoring mitochondrial membrane potential, intracellular Ca2+ and cleaved caspase-3 levels in Aß-treated primary cultured cells and mice. More importantly, intracerebroventricular injection of ODN attenuated cognitive impairments as well as long-term potentiation in Aß-treated mice. CONCLUSION: These results suggest that ODN may exert a potent neuroprotective effect against Aß-induced neurotoxicity and memory decline via its antioxidant effects, indicating that ODN may be a potential therapeutic agent for AD.


Asunto(s)
Enfermedad de Alzheimer , Encéfalo , Disfunción Cognitiva , Inhibidor de la Unión a Diazepam , Neuropéptidos , Fármacos Neuroprotectores , Estrés Oxidativo , Fragmentos de Péptidos , Animales , Humanos , Ratones , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Antioxidantes/metabolismo , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Apoptosis/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Región CA1 Hipocampal/efectos de los fármacos , Células Cultivadas , Disfunción Cognitiva/complicaciones , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/prevención & control , Inhibidor de la Unión a Diazepam/farmacología , Inhibidor de la Unión a Diazepam/uso terapéutico , Modelos Animales de Enfermedad , Células HEK293 , Potenciación a Largo Plazo/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Memoria/efectos de los fármacos , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuropéptidos/farmacología , Neuropéptidos/uso terapéutico , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Fragmentos de Péptidos/uso terapéutico
5.
Food Res Int ; 158: 111576, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35840261

RESUMEN

Pseudostellaria heterophylla, has historically been used as medicine food homology plant for thousand years in China. Our previous studies had indicated that daily intake of Pseudostellaria heterophylla extract enhanced cognitive memory. Herein, heterophyllin B (HET-B), a brain permeable cyclopeptide from Pseudostellaria heterophylla was determined, and the molecular mechanism underlying its memory improvement effects was investigated. Pseudostellaria heterophylla extract as well as HET-B reversed Aß25-35-induced axonal atrophy and neuronal apoptosis in cultured cortical neurons of mice. HET-B could enhance memory retrieval, modulate splenic T helper cell, and ameliorate neuroinflammation in i.c.v. Aß1-42 injected Alzheimer's disease (AD) mice. To explore the mechanism of action, network pharmacology was performed to predict protein targets and pathways of HET-B against AD. Five key targets were identified related to the effect of HET-B in AD intervention, and were clarified involved in axonal regeneration. We revealed for the first time that HET-B promoted memory retrieval through axonal regeneration and anti-neuroinflammation. This study provides a basis to research on HET-B as nutritional supplements for brain healthy.


Asunto(s)
Caryophyllaceae , Memoria , Neuritas , Péptidos Cíclicos , Animales , Caryophyllaceae/química , Caryophyllaceae/metabolismo , Inmunomodulación/efectos de los fármacos , Memoria/efectos de los fármacos , Ratones , Péptidos Cíclicos/farmacología , Extractos Vegetales/metabolismo , Extractos Vegetales/farmacología , Regeneración/efectos de los fármacos
6.
Cells ; 11(15)2022 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-35892581

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disorder, and no effective treatments are available to treat this disorder. Therefore, researchers have been investigating Hericium erinaceus, or the monkey head mushroom, an edible medicinal mushroom, as a possible treatment for AD. In this narrative review, we evaluated six preclinical and three clinical studies of the therapeutic effects of Hericium erinaceus on AD. Preclinical trials have successfully demonstrated that extracts and bioactive compounds of Hericium erinaceus have potential beneficial effects in ameliorating cognitive functioning and behavioral deficits in animal models of AD. A limited number of clinical studies have been conducted and several clinical trials are ongoing, which have thus far shown analogous outcomes to the preclinical studies. Nonetheless, future research on Hericium erinaceus needs to focus on elucidating the specific neuroprotective mechanisms and the target sites in AD. Additionally, standardized treatment parameters and universal regulatory systems need to be established to further ensure treatment safety and efficacy. In conclusion, Hericium erinaceus has therapeutic potential and may facilitate memory enhancement in patients with AD.


Asunto(s)
Enfermedad de Alzheimer , Hericium , Memoria , Enfermedad de Alzheimer/tratamiento farmacológico , Animales , Extractos Celulares/farmacología , Extractos Celulares/uso terapéutico , Modelos Animales de Enfermedad , Hericium/química , Humanos , Memoria/efectos de los fármacos , Neuroprotección/efectos de los fármacos
7.
Food Funct ; 13(6): 3247-3257, 2022 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-35233585

RESUMEN

Lycium ruthenicum Murr. fruit (LRF) is an edible berry known for its rich anthocyanin content. Our previous study has shown that LRF-derived anthocyanins have neuroprotective effects in rats, which may be due to their effective antioxidant activity. Therefore, this study performed online HPLC-DPPH screening as a bioactivity-guided method for the preparative separation of anthocyanins from LRF. Finally, the main fraction was isolated and identified as petunidin-3,5-O-diglucoside (Pn3G5G). Pn3G5G exhibited strong antioxidant capacity during DPPH and ABTS free radical scavenge assays. Furthermore, Pn3G5G exhibited protective effects on Nε-carboxymethyllysine (CML)-treated Neuro-2a cells by enhancing cell viability in a dose-dependent manner. CML-induced apoptosis was also reduced by Pn3G5G potentially by suppressing oxidative stress and inflammation. More importantly, Pn3G5G significantly improved cognitive impairment, neuroinflammation and neuronal apoptosis in D-galactose-induced aging mice. The result suggests the development of Pn3G5G as a healthcare product or a potent dietary supplement with antioxidant and neuroprotective effects.


Asunto(s)
Antocianinas , Antioxidantes , Lycium , Neuronas , Fármacos Neuroprotectores , Animales , Masculino , Ratones , Envejecimiento , Antocianinas/química , Antocianinas/aislamiento & purificación , Antocianinas/farmacología , Antioxidantes/química , Antioxidantes/aislamiento & purificación , Antioxidantes/farmacología , Antioxidantes/fisiología , Apoptosis/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Disfunción Cognitiva , Frutas/química , Galactosa/farmacología , Lycium/química , Memoria/efectos de los fármacos , Ratones Endogámicos C57BL , Enfermedades Neuroinflamatorias , Neuronas/efectos de los fármacos , Neuronas/fisiología , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/aislamiento & purificación , Fármacos Neuroprotectores/farmacología , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos
8.
Pak J Pharm Sci ; 35(1(Supplementary)): 239-245, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35228183

RESUMEN

Geraniol, a component of essential oil, is reported to have various pharmacological properties. The current study was conducted to demonstrate the dose-dependent neurobehavioral effects of geraniol. Rats were divided into 5 groups (n=7), comprising of control and four test groups for different doses of geraniol including 10, 30, 50 and 100 mg/kg. Geraniol was given for 15 days through intraperitoneal route. Following the administration, anxiety-, depression-like behaviors and memory function were evaluated. Extent of oxidative stress in rat's brain was also assessed by determining the levels of malondialdehyde and antioxidant enzymes activity. The present study revealed that low doses of geraniol produced more potent anxiolytic, antidepressant, nootropic, and antioxidant effects as compared to the higher doses. The findings highlight the dual characteristic of geraniol, acting as antioxidant at lower doses while at higher doses it produces pro-oxidant effects. The results are discussed in the context of dual characteristic of antioxidant compounds.


Asunto(s)
Monoterpenos Acíclicos/farmacología , Ansiedad/tratamiento farmacológico , Malondialdehído/sangre , Memoria/efectos de los fármacos , Aceites Volátiles/química , Animales , Conducta Animal/efectos de los fármacos , Encéfalo , Glutatión/metabolismo , Masculino , Ratas , Ratas Wistar
9.
Mol Med Rep ; 25(4)2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35119079

RESUMEN

Exenatide could reduce blood glucose and alleviate cognitive dysfunction induced by diabetes mellitus (DM). In the present study, a diabetic model was established in Sprague­Dawley rats to further explore the mechanism of exenatide on diabetes­induced cognitive impairment. Notably, the model rats performed poorly in the Morris water maze test and had more apoptotic neurons compared with the control rats. By contrast, exenatide attenuated cognitive impairment and inhibited neuronal apoptosis in the DM rat model. To explore the neuroprotective mechanisms of exenatide, western blotting was performed to detect the expression levels of markers of endoplasmic reticulum stress, including cytochrome c (Cyt­c), Caspase­3, JNK and c­JUN, in hippocampal tissue. Reverse transcription­quantitative PCR was also performed to measure the mRNA expression levels of Cyt­c and Caspase­3. After 16 weeks of treatment, exenatide treatment downregulated Cyt­c, Caspase­3, phosphorylated (p)­JNK and p­c­JUN expression in the hippocampal tissue of diabetic rats. Moreover, Cyt­c, Caspase­3, JNK and JUN expression levels were detected following treatment with a specific inhibitor of JNK (SP600125). The results revealed that SP600125 had similar inhibitory effects on the JNK pathway and ERS­related protein expression (Cyt­t, Caspase­3, p­JNK and p­c­JUN). These results suggested that exenatide improved cognitive dysfunction in DM rats and that the underlying mechanism may be associated with inhibiting apoptosis by suppressing the activation of JNK/c­JUN.


Asunto(s)
Apoptosis/efectos de los fármacos , Disfunción Cognitiva/prevención & control , Diabetes Mellitus Experimental/tratamiento farmacológico , Exenatida/farmacología , Genes jun/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Animales , Glucemia/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Caspasa 3/genética , Caspasa 3/metabolismo , Disfunción Cognitiva/etiología , Citocromos c/genética , Citocromos c/metabolismo , Diabetes Mellitus Experimental/complicaciones , Exenatida/uso terapéutico , Hipocampo/efectos de los fármacos , Hipocampo/patología , Insulina/metabolismo , Aprendizaje/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Ratas Sprague-Dawley
10.
Sci Rep ; 12(1): 2701, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-35177771

RESUMEN

Traumatic brain injury (TBI) is an important cause of death in young adults and children. Till now, the treatment of TBI in the short- and long-term complications is still a challenge. Our previous evidence implied aquaporin 4 (AQP4) and hypoxia inducible factor-1α (HIF-1α) might be potential targets for TBI. In this study, we explored the roles of AQP4 and HIF-1α on brain edema formation, neuronal damage and neurological functional deficits after TBI using the controlled cortical injury (CCI) model. The adult male Sprague Dawley rats were randomly divided into sham and TBI group, the latter group was further divided into neutralized-AQP4 antibody group, 2-methoxyestradiol (2-ME2) group, and their corresponding control, IgG and isotonic saline groups, respectively. Brain edema was examined by water content. Hippocampal neuronal injury was assessed by neuron loss and neuronal skeleton related protein expressions. Spatial learning and memory deficits were evaluated by Morris water maze test and memory-related proteins were detected by western blot. Our data showed that increased AQP4 protein level was closely correlated with severity of brain edema after TBI. Compared with that in the control group, both blockage of AQP4 with neutralized-AQP4 antibody and inhibition of HIF-1α with 2-ME2 for one-time treatment within 30-60 min post TBI significantly ameliorated brain edema on the 1st day post-TBI, and markedly alleviated hippocampal neuron loss and spatial learning and memory deficits on the 21st day post-TBI. In summary, our preliminary study revealed the short-term and long-term benefits of targeting HIF-1α-AQP4 axis after TBI, which may provide new clues for the selection of potential therapeutic targets for TBI in clinical practice.


Asunto(s)
Acuaporina 4/antagonistas & inhibidores , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/metabolismo , Corteza Cerebral/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Neuronas/metabolismo , 2-Metoxiestradiol/administración & dosificación , Animales , Anticuerpos/administración & dosificación , Acuaporina 4/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Edema Encefálico/etiología , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/lesiones , Trastornos de Conversión/tratamiento farmacológico , Trastornos de Conversión/etiología , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Inyecciones Intravenosas , Aprendizaje/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/etiología , Neuronas/efectos de los fármacos , Ratas Sprague-Dawley
11.
Sci Rep ; 12(1): 2285, 2022 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-35145138

RESUMEN

Disrupting memory reconsolidation provides an opportunity to abruptly reduce the behavioural expression of fear memories with long-lasting effects. The success of a reconsolidation intervention is, however, not guaranteed as it strongly depends on the destabilization of the memory. Identifying the necessary conditions to trigger destabilization remains one of the critical challenges in the field. We aimed to replicate a study from our lab, showing that the occurrence of a prediction error (PE) during reactivation is necessary but not sufficient for destabilization. We tested the effectiveness of a reactivation procedure consisting of a single PE, compared to two control groups receiving no or multiple PEs. All participants received propranolol immediately after reactivation and were tested for fear retention 24 h later. In contrast to the original results, we found no evidence for a reconsolidation effect in the single PE group, but a straightforward interpretation of these results is complicated by the lack of differential fear retention in the control groups. Our results corroborate other failed reconsolidation studies and exemplify the complexity of experimentally investigating this process in humans. Thorough investigation of the interaction between learning and memory reactivation is essential to understand the inconsistencies in the literature and to improve reconsolidation interventions.


Asunto(s)
Conducta/fisiología , Miedo/psicología , Consolidación de la Memoria/fisiología , Memoria/fisiología , Adolescente , Adulto , Conducta/efectos de los fármacos , Extinción Psicológica/efectos de los fármacos , Extinción Psicológica/fisiología , Miedo/efectos de los fármacos , Femenino , Humanos , Aprendizaje/efectos de los fármacos , Aprendizaje/fisiología , Masculino , Memoria/efectos de los fármacos , Consolidación de la Memoria/efectos de los fármacos , Propranolol/farmacología , Retención en Psicología/efectos de los fármacos , Retención en Psicología/fisiología , Adulto Joven
12.
Acta Neuropathol Commun ; 10(1): 10, 2022 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-35090569

RESUMEN

Clinical trials of therapeutics for traumatic brain injury (TBI) demonstrating preclinical efficacy for TBI have failed to replicate these results in humans, in part due to the absence of clinically feasible therapeutic windows for administration. Minocycline, an inhibitor of microglial activation, has been shown to be neuroprotective when administered early after experimental TBI but detrimental when administered chronically to human TBI survivors. Rather than focusing on the rescue of primary injury with early administration of therapeutics which may not be clinically feasible, we hypothesized that minocycline administered at a clinically feasible time point (24 h after injury) would be neuroprotective in a model of TBI plus delayed hypoxemia. We first explored several different regimens of minocycline dosing with the initial dose 24 h after injury and 2 h prior to hypoxemia, utilizing short-term neuropathology to select the most promising candidate. We found that a short course of minocycline reduced acute microglial activation, monocyte infiltration and hippocampal neuronal loss at 1 week post injury. We then conducted a preclinical trial to assess the long-term efficacy of a short course of minocycline finding reductions in hippocampal neurodegeneration and synapse loss, preservation of white matter myelination, and improvements in fear memory performance at 6 months after injury. Timing in relation to injury and duration of minocycline treatment and its impact on neuroinflammatory response may be responsible for extensive neuroprotection observed in our studies.


Asunto(s)
Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Hipoxia/tratamiento farmacológico , Minociclina/farmacología , Fármacos Neuroprotectores/farmacología , Recuperación de la Función/efectos de los fármacos , Animales , Femenino , Masculino , Memoria/efectos de los fármacos , Ratones , Minociclina/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico
13.
Sci Rep ; 12(1): 1450, 2022 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-35087146

RESUMEN

The calcitonin gene-related peptide (CGRP) suppresses fear memory retention in mice. Although intracerebroventricular administration of CGRP alters the fear memory processes, making it a promising therapeutic strategy for post-traumatic stress disorder (PTSD), direct brain injection into patients is not practical. Therefore, we propose that intranasal application may be an effective way to deliver CGRP to the brain. This study tested whether CGRP nasal administration exerts the same effect as intracerebroventricular administration using C57BL6J mice. The amount of CGRP in the cerebrospinal fluid and hippocampus 30 min after nasal administration of CGRP was significantly higher when compared with saline. Intranasal CGRP also elicited photophobic behaviors similar to intracerebroventricular injection. Moreover, intranasal CGRP decreased fear memory retention but did not affect reactivation and extinction of fear memory. We found intranasal CGRP significantly increased the expression of protein kinase D (PKD), phosphorylated histone deacetylase 5 (p-HDAC5) and neuronal PAS domain protein 4 (Npas4) in the hippocampus. CGRP-mediated impairment of fear memory and Npas4 expression increases were attenuated significantly by the CGRP receptor antagonist BIBN4096. Together, our data demonstrate that intranasal CGRP delivery activates the PKD/p-HDAC5/Npas4 pathway, decreases fear memory retention.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/administración & dosificación , Miedo , Hipocampo/efectos de los fármacos , Memoria/efectos de los fármacos , Retención en Psicología/efectos de los fármacos , Administración Intranasal , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Hipocampo/metabolismo , Histona Desacetilasas/metabolismo , Masculino , Ratones , Modelos Animales , Proteína Quinasa C/metabolismo , Transducción de Señal/efectos de los fármacos
14.
Biomed Pharmacother ; 147: 112663, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35093759

RESUMEN

Memory-enhancing agents have long been required for various reasons such as for obtaining a good score in a test in the young and for retaining memory in the aged. Although many studies have found that several natural products may be good candidates for memory enhancement, there is still a need for better agents. The present study investigated whether rubrofusarin, an active ingredient in Cassiae semen, enhances learning and memory in normal mice. Passive avoidance and Morris water maze tests were performed to determine the memory-enhancing ability of rubrofusarin. To investigate synaptic function, hippocampal long-term potentiation (LTP) was measured. Western blotting was performed to determine protein levels. To investigate neurite outgrowth, DCX immunohistochemistry and cell culture were utilised. Rubrofusarin (1, 3, 10, 30 mg/kg) enhanced memory in passive avoidance and Morris water maze tests. Moreover, rubrofusarin ameliorated scopolamine-induced memory impairment. In the rubrofusarin-treated group, high-frequency stimulation induced higher LTP in the hippocampal Schaffer-collateral pathway compared to the control group. The rubrofusarin-treated group showed a higher number of DCX-positive immature neurons with an increase in the length of dendrites compared to the control group in the hippocampal dentate gyrus region. In vitro experiments showed that rubrofusarin facilitated neurite outgrowth in neuro2a cells through extracellular signal-regulated kinase (ERK). Finally, we found that extracellular signal-regulated kinase (ERK) is required for rubrofusarin-induced enhancement of neurite outgrowth, learning and memory. These results demonstrate that rubrofusarin enhances learning and memory and neurite outgrowth, and these might need activation of ERK pathway.


Asunto(s)
Cognición/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Proyección Neuronal/efectos de los fármacos , Pironas/farmacología , Animales , Técnicas de Cultivo de Célula , Relación Dosis-Respuesta a Droga , Hipocampo/efectos de los fármacos , Aprendizaje/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Ratones , Pironas/administración & dosificación
15.
PLoS One ; 17(1): e0262474, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35081152

RESUMEN

Alzheimer's disease (AD) is the leading cause of dementia in the elderly, but therapeutic options are lacking. Despite long being able to effectively treat the ill-effects of pathology present in various rodent models of AD, translation of these strategies to the clinic has so far been disappointing. One potential contributor to this situation is the fact that the vast majority of AD patients have other dementia-contributing comorbid pathologies, the most common of which are vascular in nature. This situation is modeled relatively infrequently in basic AD research, and almost never in preclinical studies. As part of our efforts to develop small molecule, anti-inflammatory therapeutics for neurological injury and disease, we have recently been exploring potentially promising treatments in preclinical multi-morbidity contexts. In the present study, we generated a mouse model of mixed amyloid and hyperhomocysteinemia (HHcy) pathology in which to test the efficacy of one of our anti-inflammatory compounds, MW151. HHcy can cause cerebrovascular damage and is an independent risk factor for both AD dementia and vascular contributions to cognitive impairment and dementia. We found that MW151 was able to partially rescue hippocampal-dependent spatial memory and learning deficits in this comorbidity context, and further, that the benefit is associated with a normalization of hippocampal metabolites detectable via magnetic resonance spectroscopy. These findings provide evidence that MW151 in particular, and potentially anti-inflammatory treatment more generally, may be beneficial in AD patients with comorbid vascular pathology.


Asunto(s)
Antiinflamatorios/uso terapéutico , Demencia/tratamiento farmacológico , Hipocampo/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Memoria/efectos de los fármacos , Animales , Antiinflamatorios/farmacología , Conducta Animal/efectos de los fármacos , Demencia/diagnóstico por imagen , Demencia/metabolismo , Modelos Animales de Enfermedad , Hipocampo/diagnóstico por imagen , Hipocampo/metabolismo , Imagen por Resonancia Magnética , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/diagnóstico por imagen , Trastornos de la Memoria/metabolismo , Ratones
16.
Life Sci ; 293: 120349, 2022 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-35065162

RESUMEN

AIMS: Propofol, the most commonly used intravenous anesthetic, is known for its protective effect in various human and animal disease models such as post-traumatic stress disease (PTSD). However, it still needs efforts to clarify the effect of propofol on fear memory extinction and the relevant mechanisms. METHODS: Fear memory extinction was examined in PTSD mice model. Thirty-six mice were randomly divided into three groups: a shock + propofol group (sh + Pro), shock + normal saline group (sh + NS), and sham group. The mice were treated with propofol (150 mg/kg) or normal saline (of the same volume) intraperitoneally 30 min after the conditioning. These mice's behavior was analysed with contextual test, sucrose preference test (SPT) and Morris water maze (MWM). Additionally, the synaptic plasticity of the hippocampus was examined by long-term potentiation (LTP) and long-term depression (LTD). KEY FINDINGS: Compared with the sham group, the sh + NS group showed increased freezing time and depressive behavior, meanwhile the sh + Pro group showed minor behavioral changes. What's more, we found that propofol rescued the impaired long-term potentiation (LTP) and long-term depression (LTD) in hippocampus of PTSD mice. All these suggest that propofol can accelerate fear memory extinction and change synaptic plasticity of PTSD mice. SIGNIFICANCE: The study proved that propofol can protect the mice from PTSD by reserving synaptic plasticity and brought a new insight into PTSD treatment indicating that propofol maybe a potential cure for PTSD.


Asunto(s)
Hipocampo/efectos de los fármacos , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Propofol/uso terapéutico , Trastornos por Estrés Postraumático/tratamiento farmacológico , Animales , Hipocampo/fisiología , Hipnóticos y Sedantes/farmacología , Hipnóticos y Sedantes/uso terapéutico , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Masculino , Aprendizaje por Laberinto/fisiología , Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal/fisiología , Propofol/farmacología , Trastornos por Estrés Postraumático/fisiopatología , Trastornos por Estrés Postraumático/psicología
17.
Int J Mol Sci ; 23(2)2022 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-35054842

RESUMEN

Tris (2-butoxyethyl) phosphate (TBEP) is an organophosphate flame retardant and used as a plasticizer in various household products such as plastics, floor polish, varnish, textiles, furniture, and electronic equipment. However, little is known about the effects of TBEP on the brain and behavior. We aimed to examine the effects of dietary exposure of TBEP on memory functions, their-related genes, and inflammatory molecular markers in the brain of allergic asthmatic mouse models. C3H/HeJSlc male mice were given diet containing TBEP (0.02 (TBEP-L), 0.2 (TBEP-M), or 2 (TBEP-H) µg/kg/day) and ovalbumin (OVA) intratracheally every other week from 5 to 11 weeks old. A novel object recognition test was conducted in each mouse at 11 weeks old. The hippocampi were collected to detect neurological, glia, and immunological molecular markers using the real-time RT-PCR method and immunohistochemical analyses. Mast cells and microglia were examined by toluidine blue staining and ionized calcium-binding adapter molecule (Iba)-1 immunoreactivity, respectively. Impaired discrimination ability was observed in TBEP-H-exposed mice with or without allergen. The mRNA expression levels of N-methyl-D aspartate receptor subunits Nr1 and Nr2b, inflammatory molecular markers tumor necrosis factor-α oxidative stress marker heme oxygenase 1, microglia marker Iba1, and astrocyte marker glial fibrillary acidic protein were significantly increased in TBEP-H-exposed mice with or without allergen. Microglia and mast cells activation were remarkable in TBEP-H-exposed allergic asthmatic mice. Our results indicate that chronic exposure to TBEP with or without allergen impaired object recognition ability accompanied with alteration of molecular expression of neuronal and glial markers and inflammatory markers in the hippocampus of mice. Neuron-glia-mast cells interaction may play a role in TBEP-induced neurobehavioral toxicity.


Asunto(s)
Asma/psicología , Retardadores de Llama/efectos adversos , Compuestos Organofosforados/efectos adversos , Ovalbúmina/efectos adversos , Animales , Asma/etiología , Asma/genética , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Exposición Dietética/efectos adversos , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Mastocitos/metabolismo , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C3H , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Microglía/metabolismo , Proteínas del Tejido Nervioso/genética , Ovalbúmina/inmunología , Estrés Oxidativo/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/genética
18.
Pharmacol Biochem Behav ; 213: 173319, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34990706

RESUMEN

We aimed to determine whether REM sleep deprivation (RSD) affects extinction and reinstatement of methamphetamine (METH) reward memory in male rats and also to evaluate the possible role of dopamine D1-like and D2-like dopamine (DA) receptors in these processes. Male rats were trained to acquire METH-induced place preference (2 mg/kg, i.p.). METH reward memory was then reinstated following a 10-day extinction period. The animals underwent a 72-hour sleep deprivation episode by multiple platforms method (in separate groups), either before the extraction or before the reinstatement of METH reward memory. The animals received SCH 23390 (0.01 or 0.05 mg/kg, i.p.) or sulpiride (20 or 60 mg/kg, i.p.) as antagonists of D1-like and D2-like DA receptors, respectively, either immediately following each daily extinction session or before the reinstatement of METH-seeking behavior. The RSD episode postponed extinction and facilitated reinstatement of METH reward memory. Administration of SCH 23390, but not sulpiride, facilitated METH extinction and decreased reinstatement of the extinguished METH-seeking behavior. Moreover, locomotor activity was not affected by METH and/or the RSD paradigm. The results would seem to suggest that the D1-like, but not the D2-like, DA receptors may be involved in the extinction and reinstatement of the extinguished METH reward memory in RSD animals. Nonetheless, more investigations are needed to elucidate the exact mechanisms involved.


Asunto(s)
Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Metanfetamina/farmacología , Trastorno de la Conducta del Sueño REM/metabolismo , Receptores de Dopamina D1/metabolismo , Privación de Sueño/metabolismo , Animales , Benzazepinas/farmacología , Condicionamiento Psicológico/efectos de los fármacos , Dopaminérgicos/farmacología , Extinción Psicológica/efectos de los fármacos , Locomoción/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Ratas , Ratas Wistar , Receptores de Dopamina D2/metabolismo , Recompensa , Sueño REM , Sulpirida/farmacología
19.
Med Sci Monit ; 28: e933978, 2022 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-34980874

RESUMEN

BACKGROUND To reveal the mechanism underlying the effect of alpha7 nicotinic acetylcholine receptor (nAChR) on neurodegeneration in Alzheimer disease (AD), the influence of the receptor on recognition in APP/PS1 mice was evaluated by using its selective agonist (PNU-282987). MATERIAL AND METHODS APP/PS1 and wild-type (WT) mice were treated with PNU or saline, respectively, for 7 days at the ages of 6 and 10 months. RESULTS Morris water maze analysis showed that both at 6 and 10 months of age, PNU treatment enhanced the learning and memory of APP/PS1 mice. However, PNU treatment did not alter the number of senile plaques. Furthermore, a higher protein expression of Nrf2/HO-1, ADAM10, SYP, and SNAP-25, and a lower level of oxidative stress, were observed in the hippocampus of APP/PS1 mice treated with PNU compared with the control group. CONCLUSIONS The results indicated that the activation of alpha7 nAChR by PNU improved the learning and memory of mice carrying the APP/PS1 mutation, regulated the levels of enzymes that mediate APP metabolization to reduce ß-amyloid peptide damage, and decreased the level of oxidative stress and maintained synaptic plasticity, in which the mechanism might be enhancement of the Nrf2/HO-1 pathway.


Asunto(s)
Enfermedad de Alzheimer , Benzamidas/farmacología , Compuestos Bicíclicos con Puentes/farmacología , Hemo-Oxigenasa 1/metabolismo , Proteínas de la Membrana/metabolismo , Memoria , Factor 2 Relacionado con NF-E2/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7 , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Modelos Animales de Enfermedad , Aprendizaje/efectos de los fármacos , Aprendizaje/fisiología , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Memoria/fisiología , Ratones , Ratones Transgénicos , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Agonistas Nicotínicos/farmacología , Presenilina-1/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor Nicotínico de Acetilcolina alfa 7/agonistas , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo
20.
Neurobiol Learn Mem ; 188: 107584, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35032676

RESUMEN

General anesthesia is widely utilized in the clinic for surgical and diagnostic procedures. However, growing evidence suggests that anesthetic exposure may affect cognitive function negatively. Unfortunately, little is known about the underlying mechanisms and efficient prevention and therapeutic strategies for the anesthesia-induced cognitive dysfunction. 5-HT7R, a serotonin receptor family member, is functionally associated with learning and memory. It has recently become a potential therapeutic target in various neurological diseases as its ligands have a wide range of neuropharmacological effects. However, it remains unknown the role of 5-HT7R in the long-term isoflurane anesthesia-induced memory impairment and whether prior activation or blockade of 5-HT7R before anesthesia has modulating effects on this memory impairment. In this study, 5-HT7R selective agonist LP-211 and 5-HT7R selective antagonist SB-269970 were pretreated intraperitoneally to mice before anesthesia; their effects on the cognitive performance of mice were assessed using fear conditioning test and novel object recognition test. Furthermore, the transcriptional level of 5-HT7R in the hippocampus was detected using qRT-PCR, and proteomics was conducted to probe the underlying mechanisms. As a result, long-term exposure to isoflurane anesthesia caused memory impairment and an increase in hippocampal 5-HT7R mRNA expression, which could be attenuated by SB-269970 pretreatment but not LP-211pretreatment. According to the proteomics results, the antiamnestic effect of SB-269970 pretreatment was probably attributed to its action on the gene expression of Slc6a11, Itpka, Arf3, Srcin1, and Epb41l2, and synapse organization in the hippocampus. In conclusion, 5-HT7R is involved in the memory impairment induced by long-term isoflurane anesthesia, and the prior blockade of 5-HT7R with SB-269970 protects the memory impairment. This finding may help to improve the understanding of the long-term isoflurane anesthesia-induced memory impairment and to construct potential preventive and therapeutic strategies for the adverse effects after long-term isoflurane exposure.


Asunto(s)
Anestésicos por Inhalación/administración & dosificación , Disfunción Cognitiva/inducido químicamente , Isoflurano/administración & dosificación , Memoria/efectos de los fármacos , Animales , Hipocampo/metabolismo , Aprendizaje , Masculino , Ratones , Ratones Endogámicos C57BL , Fenoles/farmacología , Piperazinas/farmacología , Receptores de Serotonina/metabolismo , Serotonina/metabolismo , Sulfonamidas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA