Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Cereb Blood Flow Metab ; 41(11): 3111-3126, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34176340

RESUMEN

Repetitive hypoxia (RH) exposure affects the initiation and progression of cognitive dysfunction, but little is known about the mechanisms of hypoxic brain damage. In this study, we show that sublethal RH increased anxiety, impaired learning and memory (L/M), and triggered downregulation of brain levels of glucose and several glucose metabolites in zebrafish, and that supplementation of glucose or glucosamine (GlcN) restored RH-induced L/M impairment. Fear conditioning (FC)-induced brain activation of and PKA/CREB signaling was abrogated by RH, and this effect was reversed by GlcN supplementation. RH was associated with decreased brain O-GlcNAcylation and an increased O-GlcNAcase (OGA) level. RH increased brain inflammation and p-Tau and amyloid ß accumulation, and these effects were suppressed by GlcN. Our observations collectively suggest that changes in O-GlcNAc flux during hypoxic exposure could be an important causal factor for neurodegeneration, and that supplementation of the HBP/O-GlcNAc flux may be a potential novel therapeutic or preventive target for addressing hypoxic brain damage.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Disfunción Cognitiva/metabolismo , Glucosamina/farmacología , Hipoxia/metabolismo , Pez Cebra/metabolismo , Proteínas tau/metabolismo , Animales , Ansiedad/metabolismo , Encéfalo/metabolismo , Estudios de Casos y Controles , Disfunción Cognitiva/etiología , Encefalitis/metabolismo , Femenino , Cromatografía de Gases y Espectrometría de Masas/métodos , Glucosamina/metabolismo , Glucosamina/uso terapéutico , Glucosa/metabolismo , Hipoxia/complicaciones , Hipoxia Encefálica/metabolismo , Hipoxia Encefálica/prevención & control , Discapacidades para el Aprendizaje/metabolismo , Masculino , Trastornos de la Memoria/metabolismo , N-Acetilglucosaminiltransferasas/metabolismo , Proteínas de Pez Cebra/metabolismo , beta-N-Acetilhexosaminidasas/metabolismo
2.
J Inorg Biochem ; 212: 111252, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32950828

RESUMEN

The study was to investigate the effects of flavonoids (rutin, puerarin, and silymarin) on learning and memory function in rats exposed to aluminum chloride (AlCl3). Wistar rats were administered flavonoids at a dose of 100 mg/(kg·bw)/day or 200 mg/(kg·bw)/day after exposed to 281.40 mg/(kg·bw)/day AlCl3·6H2O. The results of Morris water maze suggested that rutin and puerarin increased the frequency of crossing the platform and swimming time spent in the target quadrant of AlCl3-induced rats significantly. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay indicated that three flavonoids could alleviate apoptosis of hippocampal neurons induced by AlCl3. Real time-PCR and western blot suggested that rutin, puerarin and 100 mg/(kg·bw)/day silymarin could decrease the AlCl3-induced high expression of Bcl-2 associated X protein (Bax) mRNA and protein in hippocampus, but the expression of B cell lymphoma/leukemia-2 (Bcl-2) mRNA and protein was not significantly different among groups. Flavonoids could up regulate the low expression of autophagy related proteins (Beclin 1 (Bcl-2-interacting protein with a coiled-coil domain 1) and LC3 (microtubule-associated protein 1 light chain 3)) caused by AlCl3 exposure. Flavonoids could also adjust the change in adenosine triphosphatase, superoxide dismutase, glutathione peroxidase and malondialdehyde induced by intake of AlCl3. The results of inductively coupled plasma atomic emission spectroscopy (ICP-AES) suggested that flavonoids could effectively reduce the high Al level in brain and serum of AlCl3 exposed rats. In conclusion, three flavonoids may improve learning and memory function by inhibiting excessive apoptosis and oxidative stress in AlCl3 exposed rats.


Asunto(s)
Cloruro de Aluminio/toxicidad , Apoptosis/efectos de los fármacos , Flavonoides/uso terapéutico , Discapacidades para el Aprendizaje/tratamiento farmacológico , Trastornos de la Memoria/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Flavonoides/administración & dosificación , Flavonoides/farmacología , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Discapacidades para el Aprendizaje/inducido químicamente , Discapacidades para el Aprendizaje/metabolismo , Masculino , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/metabolismo , Neuronas/efectos de los fármacos , Ratas , Ratas Wistar
3.
Nat Neurosci ; 23(4): 533-543, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32203497

RESUMEN

Learning disabilities are hallmarks of congenital conditions caused by prenatal exposure to harmful agents. These include fetal alcohol spectrum disorders (FASDs) with a wide range of cognitive deficiencies, including impaired motor skill development. Although these effects have been well characterized, the molecular effects that bring about these behavioral consequences remain to be determined. We previously found that the acute molecular responses to alcohol in the embryonic brain are stochastic, varying among neural progenitor cells. However, the pathophysiological consequences stemming from these heterogeneous responses remain unknown. Here we show that acute responses to alcohol in progenitor cells altered gene expression in their descendant neurons. Among the altered genes, an increase of the calcium-activated potassium channel Kcnn2 in the motor cortex correlated with motor learning deficits in a mouse model of FASD. Pharmacologic blockade of Kcnn2 improves these learning deficits, suggesting Kcnn2 blockers as a new intervention for learning disabilities in FASD.


Asunto(s)
Conducta Animal/efectos de los fármacos , Trastornos del Espectro Alcohólico Fetal/tratamiento farmacológico , Discapacidades para el Aprendizaje/tratamiento farmacológico , Aprendizaje/efectos de los fármacos , Corteza Motora/efectos de los fármacos , Venenos de Escorpión/farmacología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/antagonistas & inhibidores , Animales , Forma de la Célula/efectos de los fármacos , Dendritas/efectos de los fármacos , Dendritas/metabolismo , Modelos Animales de Enfermedad , Discapacidades para el Aprendizaje/metabolismo , Ratones , Actividad Motora/efectos de los fármacos , Corteza Motora/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Venenos de Escorpión/uso terapéutico , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo
4.
Neuromolecular Med ; 21(3): 250-261, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31037465

RESUMEN

Depression is one of the most prevalent and crucial public health problem connected to significant mortality and co-morbidity. Recently, numerous studies suggested that dietary flavanones exhibit neuroprotective and antidepressant effects against various psycho-physiological conditions including depression. The present study is focused on the antidepressant and neuroprotective effects of naringenin (NAR) and the involvement of sonic hedgehog (Shh) signaling in the chronic unpredictable mild stress (CUMS)-induced depression. Twenty-four male Wistar rats were randomly assigned into four groups: CON group (saline s.c.), NAR group (NAR 50 mg/kg, p.o.), CUMS group (subjected to CUMS along with saline p.o.), and CUMS + NAR group (NAR 50 mg/kg p.o. along with CUMS) for 28 days including 1-week pre-treatment with NAR. The results showed that NAR was found to inhibit behavioral abnormalities including increased despair in force swim test, and reduced locomotor activity caused by CUMS in open field test. Moreover, Morris water maze revealed that NAR also mitigates CUMS-associated cognitive impairment. In addition to the antidepressant-like effect, NAR mitigates morphological anomalies in the hippocampal CA1 region and cortex. Furthermore, we observed brain-derived neurotrophic factor (BDNF), Shh, GLI1, NKX2.2, and PAX6 were downregulated in the hippocampus of CUMS-exposed rats, which can be upregulated by NAR pre-treatment. GLI1 is main downstream signaling component of Shh signaling cascade, which further regulates the expression of homeodomain transcription factors PAX6 and NKX2.2.


Asunto(s)
Antidepresivos/uso terapéutico , Depresión/tratamiento farmacológico , Flavanonas/uso terapéutico , Proteínas Hedgehog/fisiología , Discapacidades para el Aprendizaje/tratamiento farmacológico , Trastornos de la Memoria/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Transducción de Señal/efectos de los fármacos , Estrés Psicológico/tratamiento farmacológico , Proteína con Dedos de Zinc GLI1/fisiología , Animales , Antidepresivos/farmacología , Enfermedad Crónica , Depresión/etiología , Depresión/metabolismo , Depresión/prevención & control , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Conducta Exploratoria/efectos de los fármacos , Flavanonas/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/química , Hipocampo/efectos de los fármacos , Proteína Homeobox Nkx-2.2 , Discapacidades para el Aprendizaje/etiología , Discapacidades para el Aprendizaje/metabolismo , Discapacidades para el Aprendizaje/prevención & control , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/etiología , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/prevención & control , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Fármacos Neuroprotectores/farmacología , Distribución Aleatoria , Ratas , Ratas Wistar , Estrés Psicológico/fisiopatología , Natación
5.
J Med Food ; 21(7): 678-688, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29851371

RESUMEN

Schisantherin A (SCA) was evaluated for possible function in restoring the learning and memory impairment induced by D-galactose in mice. ICR mice were treated with D-galactose subcutaneously (220 mg·kg-1), and followed by SCA in different doses (1.25, 2.50 and 5.00 mg·kg-1, administered orally) for 42 days. Effects of SCA on learning and memory were examined by step-through tests and Morris water maze tests. The activity of superoxide dismutase (SOD), the content of malondialdehyde (MDA) in the peripheral blood and hippocampus of mice were assayed by water-soluble tetrazolium-1 (WST-1) and thiobarbituric acid (TBA) methods. The contents of 8 hydroxy deoxy guanosine (8-OHdG) in the hippocampus of mice were detected by immunosorbent assay methods, respectively. Quantitative real-time PCR and Western Blot were respectively used to detect the expression of p19, p53, p21, cyclin D1, CDK4 and RB genes, and the phosphorylation of RB in the hippocampus of mice. We found that SCA significantly improved the learning and memory impairment induced by D-galactose in mice. After SCA treatment, SOD activity was increased and the content of MDA was decreased in both peripheral blood and hippocampus of mice. 8-OHDG content was also decreased in the hippocampus of mice. Furthermore, the expression of p19, p53 and p21 genes was reduced and the expression of cyclin D1 and CDK4 and the phosphorylation of RB protein were elevated in the hippocampus. SCA may improve the learning and memory impairment induced by D-galactose by enhancing the antioxidant capacity, and regulating the expression of p19/p53/p21/cyclinD1/CDK4 genes, and the phosphorylation of RB protein in the hippocampus of mice.


Asunto(s)
Ciclooctanos/administración & dosificación , Dioxoles/administración & dosificación , Medicamentos Herbarios Chinos/administración & dosificación , Discapacidades para el Aprendizaje/tratamiento farmacológico , Lignanos/administración & dosificación , Trastornos de la Memoria/tratamiento farmacológico , Schisandra/química , Animales , Ciclina D1/genética , Ciclina D1/metabolismo , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Inhibidor p19 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p19 de las Quinasas Dependientes de la Ciclina/metabolismo , Galactosa/efectos adversos , Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Discapacidades para el Aprendizaje/genética , Discapacidades para el Aprendizaje/metabolismo , Discapacidades para el Aprendizaje/psicología , Masculino , Memoria/efectos de los fármacos , Trastornos de la Memoria/genética , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/psicología , Ratones , Ratones Endogámicos ICR , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo
6.
Toxicol Appl Pharmacol ; 354: 153-175, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29524501

RESUMEN

The Adverse Outcome Pathways (AOPs) are designed to provide mechanistic understanding of complex biological systems and pathways of toxicity that result in adverse outcomes (AOs) relevant to regulatory endpoints. AOP concept captures in a structured way the causal relationships resulting from initial chemical interaction with biological target(s) (molecular initiating event) to an AO manifested in individual organisms and/or populations through a sequential series of key events (KEs), which are cellular, anatomical and/or functional changes in biological processes. An AOP provides the mechanistic detail required to support chemical safety assessment, the development of alternative methods and the implementation of an integrated testing strategy. An example of the AOP relevant to developmental neurotoxicity (DNT) is described here following the requirements of information defined by the OECD Users' Handbook Supplement to the Guidance Document for developing and assessing AOPs. In this AOP, the binding of an antagonist to glutamate receptor N-methyl-d-aspartate (NMDAR) receptor is defined as MIE. This MIE triggers a cascade of cellular KEs including reduction of intracellular calcium levels, reduction of brain derived neurotrophic factor release, neuronal cell death, decreased glutamate presynaptic release and aberrant dendritic morphology. At organ level, the above mentioned KEs lead to decreased synaptogenesis and decreased neuronal network formation and function causing learning and memory deficit at organism level, which is defined as the AO. There are in vitro, in vivo and epidemiological data that support the described KEs and their causative relationships rendering this AOP relevant to DNT evaluation in the context of regulatory purposes.


Asunto(s)
Rutas de Resultados Adversos , Encéfalo/efectos de los fármacos , Conducta Infantil/efectos de los fármacos , Desarrollo Infantil/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/toxicidad , Discapacidades para el Aprendizaje/inducido químicamente , Aprendizaje/efectos de los fármacos , Trastornos de la Memoria/inducido químicamente , Síndromes de Neurotoxicidad/etiología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Adolescente , Adulto , Factores de Edad , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Niño , Preescolar , Humanos , Lactante , Recién Nacido , Discapacidades para el Aprendizaje/metabolismo , Discapacidades para el Aprendizaje/fisiopatología , Discapacidades para el Aprendizaje/psicología , Memoria/efectos de los fármacos , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/fisiopatología , Trastornos de la Memoria/psicología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Síndromes de Neurotoxicidad/metabolismo , Síndromes de Neurotoxicidad/fisiopatología , Síndromes de Neurotoxicidad/psicología , Receptores de N-Metil-D-Aspartato/metabolismo , Medición de Riesgo
7.
FASEB J ; 31(11): 4998-5011, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28739640

RESUMEN

Obesity, which is caused by an energy imbalance between calorie intake and consumption, has become a major international health burden. Obesity increases the risk of insulin resistance and age-related cognitive decline, accompanied by peripheral inflammation. (-)-Epigallocatechin-3-gallate (EGCG), the major polyphenol in green tea, possesses antioxidant, anti-inflammatory, and cardioprotective activities; however, few reports have focused on its potential effect on cognitive disorders. In this study, our goal was to investigate the protective effects of EGCG treatment on insulin resistance and memory impairment induced by a high-fat and high-fructose diet (HFFD). We randomly assigned 3-mo-old C57BL/6J mice to 3 groups with different diets: control group, HFFD group, and HFFD plus EGCG group. Memory loss was assessed by using the Morris water maze test, during which EGCG was observed to prevent HFFD-elicited memory impairment and neuronal loss. Consistent with these results, EGCG attenuated HFFD-induced neuronal damage. Of note, EGCG significantly ameliorated insulin resistance and cognitive disorder by up-regulating the insulin receptor substrate-1 (IRS-1)/AKT and ERK/cAMP response element binding protein (CREB)/brain-derived neurotrophic factor (BDNF) signaling pathways. Long-term HFFD-triggered neuroinflammation was restored by EGCG supplementation by inhibiting the MAPK and NF-κB pathways, as well as the expression of inflammatory mediators, such as TNF-α. EGCG also reversed high glucose and glucosamine-induced insulin resistance in SH-SY5Y neuronal cells by improving the oxidized cellular status and mitochondrial function. To our knowledge, this study is the first to provide compelling evidence that the nutritional compound EGCG has the potential to ameliorate HFFD-triggered learning and memory loss.-Mi, Y., Qi, G., Fan, R., Qiao, Q., Sun, Y., Gao, Y., Liu, X. EGCG ameliorates high-fat- and high-fructose-induced cognitive defects by regulating the IRS/AKT and ERK/CREB/BDNF signaling pathways in the CNS.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Catequina/análogos & derivados , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Carbohidratos de la Dieta/efectos adversos , Grasas de la Dieta/efectos adversos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fructosa/efectos adversos , Proteínas Sustrato del Receptor de Insulina/metabolismo , Discapacidades para el Aprendizaje/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Trastornos de la Memoria/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Catequina/farmacología , Línea Celular , Carbohidratos de la Dieta/farmacología , Grasas de la Dieta/farmacología , Fructosa/farmacología , Discapacidades para el Aprendizaje/inducido químicamente , Discapacidades para el Aprendizaje/patología , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/patología , Ratones
8.
Neuromolecular Med ; 19(2-3): 309-321, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28623611

RESUMEN

Parkinson's disease (PD) is the most common neurodegenerative movement disorder, and its causes remain unknown. A major hallmark of the disease is the increasing presence of aggregated alpha-synuclein (aSyn). Furthermore, there is a solid consensus on iron (Fe) accumulation in several regions of PD brains during disease progression. In our study, we focused on the interaction of Fe and aggregating aSyn in vivo in a transgenic mouse model overexpressing human aSyn bearing the A53T mutation (prnp.aSyn.A53T). We utilized a neonatal iron-feeding model to exacerbate the motor phenotype of the transgenic mouse model. Beginning from day 100, mice were treated with deferiprone (DFP), a ferric chelator that is able to cross the blood-brain barrier and is currently used in clinics as treatment for hemosiderosis. Our paradigm resulted in an impairment of the learning abilities in the rotarod task and the novel object recognition test. DFP treatment significantly improved the performance in both tasks. Although this was not accompanied by alterations in aSyn aggregation, our results support DFP as possible therapeutic option in PD.


Asunto(s)
Trastornos Neurológicos de la Marcha/tratamiento farmacológico , Quelantes del Hierro/uso terapéutico , Hierro/toxicidad , Discapacidades para el Aprendizaje/tratamiento farmacológico , Trastornos Parkinsonianos/tratamiento farmacológico , Piridonas/uso terapéutico , alfa-Sinucleína/genética , Animales , Recuento de Células , Deferiprona , Evaluación Preclínica de Medicamentos , Femenino , Trastornos Neurológicos de la Marcha/etiología , Trastornos Neurológicos de la Marcha/metabolismo , Humanos , Hierro/metabolismo , Discapacidades para el Aprendizaje/etiología , Discapacidades para el Aprendizaje/metabolismo , Masculino , Ratones , Ratones Transgénicos , Neuronas/patología , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/psicología , Agregación Patológica de Proteínas/tratamiento farmacológico , Agregación Patológica de Proteínas/metabolismo , Reconocimiento en Psicología/efectos de los fármacos , Prueba de Desempeño de Rotación con Aceleración Constante , alfa-Sinucleína/metabolismo
9.
Acta Neuropathol ; 133(6): 967-982, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28130638

RESUMEN

Deposition of amyloid ß protein (Aß) to form neuritic plaques in the brain is the unique pathological hallmark of Alzheimer's disease (AD). Aß is derived from amyloid ß precursor protein (APP) by ß- and γ-secretase cleavages and turned over by glia in the central nervous system (CNS). Vitamin A deficiency (VAD) has been shown to affect cognitive functions. Marginal vitamin A deficiency (MVAD) is a serious and widespread public health problem among pregnant women and children in developing countries. However, the role of MVAD in the pathogenesis of AD remains elusive. Our study showed that MVAD is approximately twofold more prevalent than VAD in the elderly, and increased cognitive decline is positively correlated with lower VA levels. We found that MVAD, mostly prenatal MVAD, promotes beta-site APP cleaving enzyme 1 (BACE1)-mediated Aß production and neuritic plaque formation, and significantly exacerbates memory deficits in AD model mice. Supplementing a therapeutic dose of VA rescued the MVAD-induced memory deficits. Taken together, our study demonstrates that MVAD facilitates AD pathogenesis and VA supplementation improves cognitive deficits. These results suggest that VA supplementation might be a potential approach for AD prevention and treatment.


Asunto(s)
Encéfalo/metabolismo , Encéfalo/patología , Deficiencia de Vitamina A/metabolismo , Deficiencia de Vitamina A/patología , Anciano , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Biomarcadores/sangre , Línea Celular , Suplementos Dietéticos , Modelos Animales de Enfermedad , Femenino , Humanos , Discapacidades para el Aprendizaje/dietoterapia , Discapacidades para el Aprendizaje/metabolismo , Discapacidades para el Aprendizaje/patología , Masculino , Trastornos de la Memoria/dietoterapia , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/patología , Ratones Transgénicos , Placa Amiloide/metabolismo , Placa Amiloide/patología , Vitamina A/administración & dosificación , Vitamina A/sangre , Deficiencia de Vitamina A/dietoterapia , Deficiencia de Vitamina A/psicología
10.
J Nutr Biochem ; 35: 87-95, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27469996

RESUMEN

Docosahexaenoic acid (22:6n-3; DHA) is known to play a critical role in postnatal brain development. However, there have been no studies investigating the preventive effect of DHA on prenatal valproic acid (VPA)-induced behavioral and molecular alterations in offspring. The present study was to evaluate the neuroprotective effects in offspring using maternal feeding of DHA to rats exposed to VPA in pregnancy. In the present study, rats were exposed to VPA on day 12.5 of pregnancy; DHA was administered at the dosages of 100, 300 and 500 mg/kg/day for 3 weeks from day 1 to 21 of pregnancy. The results showed that maternal feeding of DHA to the prenatal exposed to VPA (1) prevented VPA-induced learning and memory impairment but did not change social-related behavior, (2) increased total DHA content in offspring plasma and hippocampus, (3) rescued VPA-induced neuronal loss and apoptosis of pyramidal cells in hippocampal CA1, (4) influenced the content of malondialdehyde and glutathione and the activities of superoxide dismutase and glutathione in the hippocampus, (5) altered levels of apoptosis-related proteins (Bcl-2, Bax and caspase-3) and inhibited the activity of caspase-3 in offspring hippocampus and (6) enhanced relative levels of p-CaMKII and p-CREB proteins in the hippocampus. These findings suggest that maternal feeding with DHA may prevent prenatal VPA-induced impairment of learning and memory, normalize several different molecules associated with oxidative stress and apoptosis in the hippocampus of offspring, and exert preventive effects on prenatal VPA-induced brain dysfunction.


Asunto(s)
Anticonvulsivantes/efectos adversos , Ácidos Docosahexaenoicos/uso terapéutico , Discapacidades para el Aprendizaje/prevención & control , Fenómenos Fisiologicos Nutricionales Maternos , Trastornos de la Memoria/prevención & control , Efectos Tardíos de la Exposición Prenatal , Ácido Valproico/efectos adversos , Animales , Anticonvulsivantes/química , Conducta Animal/efectos de los fármacos , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/patología , Suplementos Dietéticos , Ácidos Docosahexaenoicos/administración & dosificación , Ácidos Docosahexaenoicos/metabolismo , Femenino , Discapacidades para el Aprendizaje/inducido químicamente , Discapacidades para el Aprendizaje/metabolismo , Discapacidades para el Aprendizaje/patología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/patología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/uso terapéutico , Fosforilación/efectos de los fármacos , Embarazo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ratas Wistar , Conducta Social , Ácido Valproico/antagonistas & inhibidores
11.
Int J Mol Med ; 38(3): 869-75, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27430591

RESUMEN

Caffeic acid is a type of phenolic acid and organic acid. It is found in food (such as tomatoes, carrots, strawberries, blueberries and wheat), beverages (such as wine, tea, coffee and apple juice) as well as Chinese herbal medicines. In the present study, we examined the effects of caffeic acid on learning deficits in a rat model of Alzheimer's disease (AD). The rats were randomly divided into three groups: i) control group, ii) AD model group and iii) caffeic acid group. Caffeic acid significantly rescued learning deficits and increased cognitive function in the rats with AD as demonstrated by the Morris water maze task. Furthermore, caffeic acid administration resulted in a significant decrease in acetylcholinesterase activity and nitrite generation in the rats with AD compared with the AD model group. Furthermore, caffeic acid suppressed oxidative stress, inflammation, nuclear factor­κB­p65 protein expression and caspase­3 activity as well as regulating the protein expression of p53 and phosphorylated (p-)p38 MAPK expression in the rats with AD. These experimental results indicate that the beneficial effects of caffeic acid on learning deficits in a model of AD were due to the suppression of oxidative stress and inflammation through the p38 MAPK signaling pathway.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Ácidos Cafeicos/farmacología , Modelos Animales de Enfermedad , Discapacidades para el Aprendizaje/prevención & control , Acetilcolinesterasa/metabolismo , Animales , Western Blotting , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Caspasa 3/metabolismo , Cognición/efectos de los fármacos , Inflamación/prevención & control , Discapacidades para el Aprendizaje/etiología , Discapacidades para el Aprendizaje/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Nitritos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Distribución Aleatoria , Ratas Sprague-Dawley , Factor de Transcripción ReIA/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Rev. int. med. cienc. act. fis. deporte ; 16(62): 297-315, jun. 2016. tab, graf
Artículo en Español | IBECS | ID: ibc-153358

RESUMEN

Estudios recientes observaron que las imágenes motrices se desarrollan de forma entrelazada con el desarrollo de las habilidades motrices en niños. La finalidad de este estudio es analizar en qué medida la imagen motriz de los elementos necesarios para resolver un problema motor (la recepción de un balón), se relaciona con los niveles de habilidad en niños (3 - 9 años). La muestra estuvo formada por 215 participantes (87 chicos y 118 chicas), (M = 5,94, DT = 1,47). Se ha utilizado una metodología mixta: dibujos, indicaciones gestuales, verbalización del pensamiento y una prueba práctica de recepción de balón. El MANOVA reveló diferencias significativas en las capacidades meta-cognitivas y motrices en función de las etapas de desarrollo. Un análisis de ecuaciones estructurales reveló que las capacidades meta-cognitivas median la relación entre las etapas de desarrollo y la habilidad de recepción de móviles. Se discuten sus repercusiones en el aprendizaje motor (AU)


Recent studies have found that motor imaginery is developed linked to the development of motor skills in children. The purpose of this study is to analyze how the motor imaginery of theprincipal elements to solve a motor problem (ball reception) relates to the motor skill levels in children (3-9 years). The sample consisted of 215 participants (87 boys and 118 girls), (M = 5.94, SD = 1.47).We used a mixed methodology: drawings, gestural prompts, verbalization of thought and a practical test of ball reception. The MANOVA revealed significant differences in the meta-cognitive abilities and motor function of the developmental stages. A structural equation analysis revealed that meta-cognitive abilities mediate the relationship between the stages of development and the ability in the reception of moving objects. Their implications in motor learning are discussed (AU)


Asunto(s)
Humanos , Masculino , Femenino , Niño , Destreza Motora/fisiología , Discapacidades para el Aprendizaje/metabolismo , Discapacidades para el Aprendizaje/psicología , Baloncesto/educación , Estudios Transversales/métodos , Actividad Motora/genética , Destreza Motora/clasificación , Discapacidades para el Aprendizaje/rehabilitación , Discapacidades para el Aprendizaje/terapia , Educación Primaria y Secundaria , Baloncesto/normas , Estudios Transversales , Actividad Motora/fisiología
13.
Chin J Nat Med ; 13(11): 823-830, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26614457

RESUMEN

The present study was aimed at determining the effects of Tongqiao Huoxue Decoction (TQHXD) on the Ca(2+)-CaMKII-CREB pathway and the memory and learning capacities of rats with vascular dementia (VD). The rat VD model was established by using an improved bilateral carotid artery ligation method. The Morris water maze experiment was used to evaluate the ethology of the VD rats following treatments with TQHXD at 3.01, 6.02, and 12.04 g·kg(-1) per day for 31 days. At the end of experiment, the hippocampus were harvested and analyzed. Western blotting and RT-PCR were used to measure the expression levels of calmodulin-binding protein kinase II(CaMKII), protein kinase A(PKA), cAMP-response element binding protein(CREB), and three N-methyl-D-aspartic acid receptor subunits (NR1, NR2A, and NR2B). Our results revealed that TQHXD could alleviate the loss of learning abilities and increase the memory capacity (P < 0.05 and P < 0.01 vs the model group, respectively). The treatment with 6.02 and 12.04 g·kg(-1) of TQHXD significantly up-regulated the Ca(2+)-CaMKII-CREB pathway in the hippocampus. In conclusion, TQHXD showed therapeutic effects on a bilateral carotid artery ligation-induced vascular dementia model, through the up-regulation of calcium signalling pathways.


Asunto(s)
Calcio/metabolismo , Demencia Vascular/tratamiento farmacológico , Medicamentos Herbarios Chinos/uso terapéutico , Hipocampo/efectos de los fármacos , Discapacidades para el Aprendizaje/tratamiento farmacológico , Trastornos de la Memoria/tratamiento farmacológico , Memoria/efectos de los fármacos , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Demencia Vascular/complicaciones , Demencia Vascular/metabolismo , Demencia Vascular/psicología , Medicamentos Herbarios Chinos/farmacología , Femenino , Hipocampo/metabolismo , Discapacidades para el Aprendizaje/etiología , Discapacidades para el Aprendizaje/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/etiología , Trastornos de la Memoria/metabolismo , Fitoterapia , Ratas Sprague-Dawley , Transducción de Señal , Regulación hacia Arriba
14.
Biochem Biophys Res Commun ; 464(4): 1090-1095, 2015 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-26188513

RESUMEN

Isoliquiritigenin (ILG), a chalcone from Glycyrrhiza uralensis, has various biological properties. ILG markedly inhibited inflammation, but the effects on the brain inflammation and insulin resistance induced by high-fat diet (HFD) are still unknown, so our study intended to investigate its effect on cognitive dysfunction induced by HFD and the relevant mechanisms. ICR mice were treated with HFD diet for 8 weeks to induce peripheral insulin resistance prior to being intervened with rosiglitazone, ILG (30, 60 mg/kg). 4 weeks later, Morris Water Maze (MWM) was used to assess the learning and memory, the insulin resistance index was measured, and the brain inflammation cytokines (IL-1ß, TNF-α) were assessed. Meanwhile, the p-JNK, p-IRS Ser(307) protein expressions in the hippocampus were also detected using the western blot to explore the corresponding mechanisms. Our results suggested that ILG could significantly alleviate the cognitive impairments in the MWM test and attenuate peripheral insulin resistance. The IL-1ß, TNF-α levels declined with the administration of ILG, meanwhile the p-IRS Ser(307) expression decreased with the inhibition of p-JNK. In conclusion, ILG could improve the spatial learning and memory lesions induced by HFD via the inhibition of TNF-α/JNK/IRS pathway.


Asunto(s)
Chalconas/farmacología , Dieta Alta en Grasa/efectos adversos , Discapacidades para el Aprendizaje/tratamiento farmacológico , Trastornos de la Memoria/tratamiento farmacológico , Nootrópicos/farmacología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Proteínas Sustrato del Receptor de Insulina/metabolismo , Resistencia a la Insulina , Interleucina-1beta/metabolismo , Discapacidades para el Aprendizaje/etiología , Discapacidades para el Aprendizaje/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/etiología , Trastornos de la Memoria/metabolismo , Ratones , Ratones Endogámicos ICR , Fitoterapia , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
15.
Zhen Ci Yan Jiu ; 40(6): 431-8, 2015 Dec.
Artículo en Chino | MEDLINE | ID: mdl-26887202

RESUMEN

OBJECTIVE: To observe the effect of electroacupuncture (EA) therapy on levels of oxygen free radicals (OFR) and hippocampal apoptosis-related protein expression in ischemic learning-memory disorder rats so as to investigate its mechanisms underlying improvement of ischemic learning-memory impairment. METHODS: A total of 60 SD rats were randomly divided into sham operation (sham), model, medication, and EA groups, with 15 rats in each group. The learning-memory disorder model was made by occlusion of bilateral carotid arteries. EA (2- 3 Hz, 2 mA) was applied to "Zhi San Zhen" ["Shenting" (GV 24) and bilateral "Benshen" (GB 13)] for 30 min, once a day for 3 weeks. The rats of the medication group were treated by lavage of Aricept (0.03 mg . kg(-1) . d(-1)), once daily for 3 weeks. The rats' learning-memory ability was detected by Morris water maze tests and the state of hippocampal apoptosis cells was observed by light microscope after TUNEL staining and the expression of hippocampal Bcl-2, Bax and Caspase-3 proteins was detected by immunohistochemistry. Serum and hippocampal superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) activity and malondialdehyde (MDA) contents were detected by chemical colorimetric analysis. RESULTS: Compared with the sham group, the escape latencies (place-navigation) after modeling were evidently prolonged, and the times of target-platform crossing in 90 sec (spatial probe test) considerably reduced in the model group (P<0.01), suggesting an impairment of learning-memory ability. After the treatment for 21 d, the increased escape latency and the reduced target-platform crossing time in both EA and medication groups were reversed in comparison with the model group (P<0.01), suggesting an improvement of memory ability, and the effect of the EA group was significantly superior to that of the medication group (P<0.05). Compared with the sham group, the number of apoptotic cells in hippocampal CA 1- CA 3 regions, and the expression levels of hippocampal Bcl-2, Bax and Caspase-3 proteins, and serum and hippocampal MDA contents were significantly increased in the model group (P<0.01), while serum and hippocampal SOD and GSH-Px levels obviously decreased in the model group (P<0.01). After the treatment for 21 days, compared to the model group, the number of the apoptotic cells, the expression levels of hippocampal Bax and Caspase--3 proteins, and the contents of serum and hippocampal MDA were notably decreased in the EA and medication groups (P<0.01), whereas, Bcl-2 protein expression levels, and serum and hippocampal SOD and GSH-Px activity were notably up-regulated in the EA and medication groups (P<0.01). The effects of EA group were obviously superior to those of medication group in increasing hippocampal Bcl-2 immunoactivity, serum SOD and GSH-Px and hippocampal GSH-Px activity and in down-regulating serum MDA level (P<0.01, P<0.05). CONCLUSION: Electroacupuncture intervention can improve learning-memory ability in ischemic learning-memory disorder rats which may be associated with its effects in reducing blood and hippocampal OFR contents and hippocampal cellular apoptosis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Electroacupuntura , Isquemia/complicaciones , Discapacidades para el Aprendizaje/terapia , Trastornos de la Memoria/terapia , Especies Reactivas de Oxígeno/metabolismo , Puntos de Acupuntura , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Glutatión Peroxidasa/metabolismo , Humanos , Isquemia/psicología , Aprendizaje , Discapacidades para el Aprendizaje/etiología , Discapacidades para el Aprendizaje/genética , Discapacidades para el Aprendizaje/metabolismo , Masculino , Malondialdehído/metabolismo , Memoria , Trastornos de la Memoria/etiología , Trastornos de la Memoria/genética , Trastornos de la Memoria/metabolismo , Ratas , Ratas Sprague-Dawley , Superóxido Dismutasa/metabolismo
16.
BMC Complement Altern Med ; 14: 482, 2014 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-25496367

RESUMEN

BACKGROUND: The physiological effects of the non-anthocyanin fraction (NAF) in a black soybean seed coat extract on Aß-induced oxidative stress were investigated to confirm neuroprotection. In addition, we examined the preventive effect of NAF on cognitive defects induced by the intracerebroventricular (ICV) injection of Aß. METHODS: Levels of cellular oxidative stress were measured using 2',7'-dichlorofluorescein diacetate (DCF-DA). Neuronal cell viability was investigated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and lactate dehydrogenase (LDH) assay. To investigate in vivo anti-amnesic effects of NAF by using Y-maze and passive avoidance tests, the learning and memory impairment in mice was induced by Aß. After in vivo assays, acetylcholinesterase (AChE) activity and level of malondialdehyde (MDA) in the mouse brain were determined to confirm the cognitive effect. Individual phenolics of NAF were qualitatively analyzed by using an ultra-performance liquid chromatography (UPLC) Accurate-Mass Quadrupole Time of-Flight (Q-TOF) UPLC/MS. RESULTS: A NAF showed cell protective effects against oxidative stress-induced cytotoxicity. Intracellular ROS accumulated through Aß1-40 treatment was significantly reduced in comparison to cells only treated with Aß1-40. In MTT and LDH assay, the NAF also presented neuroprotective effects on Aß1-40-treated cytotoxicity. Finally, the administration of this NAF in mice significantly reversed the Aß1-40-induced cognitive defects in in vivo behavioral tests. After behavioral tests, the mice brains were collected in order to examine lipid peroxidation and AChE activity. AChE, preparation was inhibited by NAF in a dose-dependent manner. MDA generation in the brain homogenate of mice treated with the NAF was decreased. Q-TOF UPLC/MS analyses revealed three major phenolics from the non-anthocyanin fraction; epicatechin, procyanidin B1, and procyanidin B2. CONCLUSIONS: The results suggest that the NAF in black soybean seed coat extracts may improve the cytotoxicity of Aß in PC12 cells, possibly by reducing oxidative stress, and also have an anti-amnesic effect on the in vivo learning and memory deficits caused by Aß. Q-TOF UPLC/MS analyses showed three major phenolics; (-)-epicatechin, procyanidin B1, and procyanidin B2. Above results suggest that (-)-epicatechins are the major components, and contributors to the anti-amnesic effect of the NAF from black soybean seed coat.


Asunto(s)
Encéfalo/efectos de los fármacos , Glycine max/química , Trastornos de la Memoria/tratamiento farmacológico , Memoria/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Polifenoles/farmacología , Acetilcolinesterasa/metabolismo , Péptidos beta-Amiloides/efectos adversos , Péptidos beta-Amiloides/metabolismo , Animales , Antioxidantes/análisis , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Biflavonoides/análisis , Biflavonoides/farmacología , Biflavonoides/uso terapéutico , Encéfalo/metabolismo , Catequina/análisis , Catequina/farmacología , Catequina/uso terapéutico , Supervivencia Celular/efectos de los fármacos , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/metabolismo , Aprendizaje/efectos de los fármacos , Discapacidades para el Aprendizaje/tratamiento farmacológico , Discapacidades para el Aprendizaje/metabolismo , Masculino , Trastornos de la Memoria/metabolismo , Ratones Endogámicos ICR , Fármacos Neuroprotectores/análisis , Fármacos Neuroprotectores/uso terapéutico , Células PC12 , Fragmentos de Péptidos/efectos adversos , Fragmentos de Péptidos/metabolismo , Fitoterapia , Extractos Vegetales/química , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Polifenoles/análisis , Polifenoles/uso terapéutico , Proantocianidinas/análisis , Proantocianidinas/farmacología , Proantocianidinas/uso terapéutico , Ratas , Semillas
17.
Neurol Sci ; 35(8): 1261-8, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24590842

RESUMEN

Baicalein has been shown to possess various pharmacological actions. The current work was designed to assess the neuroprotection of baicalein against cognitive deficits in epilepsy-like tremor rat (TRM). Epileptic characteristics and memory functions were assessed by electroencephalograms recording and Morris water maze test, respectively. The changes of oxidative indicators including malondialdehyde (MDA), catalase (CAT), Cu/Zn-superoxide dismutase (Cu/Zn-SOD), Mn-SOD, glutathione (GSH), glutathione peroxidase (GSH-PX) and 8-isoprostane were measured using corresponding commercial kits. Real-time RT-PCR and immunoassay were employed to detect activities of various inflammatory mediators such as NF-κB p65, TNF-α, IL-1ß, IL-6 and IL-10. Western blot analysis was performed to determine heat shock protein (HSP) 70 and mitogen-activated protein kinases (MAPKs) (including ERK, JNK and p38) proteins. Our results illustrated that baicalein significantly ameliorated epileptiform activity and cognitive deficits in TRM. Besides, reduced oxidative stress and inflammatory responses were also found in TRM treated with baicalein. Furthermore, there were evident alterations of HSP70 and MAPK cascades at protein levels after 14-day pretreatment with baicalein. It was concluded that the neuroprotective effect of baicalein against cognitive dysfunction might be associated with suppressing oxidative stress, inhibiting inflammation and mediating HSP70 as well as MAPK cascades in absence-like TRM.


Asunto(s)
Química Encefálica/efectos de los fármacos , Medicamentos Herbarios Chinos/uso terapéutico , Epilepsia Tipo Ausencia/psicología , Flavanonas/uso terapéutico , Discapacidades para el Aprendizaje/prevención & control , Fármacos Neuroprotectores/uso terapéutico , Nootrópicos/uso terapéutico , Convulsiones/psicología , Animales , Antiinflamatorios/uso terapéutico , Antioxidantes/uso terapéutico , Citocinas/análisis , Evaluación Preclínica de Medicamentos , Electroencefalografía , Epilepsia Tipo Ausencia/genética , Epilepsia Tipo Ausencia/metabolismo , Proteínas de Choque Térmico/análisis , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Mediadores de Inflamación/análisis , Discapacidades para el Aprendizaje/tratamiento farmacológico , Discapacidades para el Aprendizaje/etiología , Discapacidades para el Aprendizaje/metabolismo , Aprendizaje por Laberinto/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/análisis , Proteínas del Tejido Nervioso/análisis , Estrés Oxidativo/efectos de los fármacos , Distribución Aleatoria , Ratas , Ratas Mutantes , Convulsiones/genética , Convulsiones/metabolismo , Transducción de Señal/efectos de los fármacos
18.
Zhong Yao Cai ; 36(3): 438-40, 2013 Mar.
Artículo en Chino | MEDLINE | ID: mdl-24010328

RESUMEN

OBJECTIVE: To investigate the improving effect of Litchi Seed Aqueous Extracts on learning and memory obstacles model and its mechanism. METHODS: The learning and memory obstacles model was incluced by subcutoneous injection of D-galactose (500 mg/kg) for 8 weeks. The model group and treatment groups were given huperzine A (0.4 mg/kg) and Litchi Seed Aqueous Extracts (15,60 g/kg) respectively for 4 weeks by ig at the 5th week. After huperzine A and Litchi Seed Aqueous Extracts treatment for 4 weeks, water maze test was used to determine the ability of mice's spatial learning and memory. The contents of advanced glycation end products (AGEs) in serum, the content of nitric oxide (NO) and acetylcholine (Ach), the activity of nitric oxide synthase (NOS) and acetylcholinesterase (AchE) in the brain tissue were detected. RESULTS: Litchi Seed Aqueous Extracts significantly ameliorated the learning and memory ability in mice, decreased the level of AGEs in serum, and reduced the content of NO and activity of NOS in brain tissues. No significantly influence was observed for the Ach and Ach-E in brain tissues. CONCLUSION: Litchi Seed Aqueous Extracts possesses improving the learning and memory effects on the model mice induced by D-galactose, which may be related to inhibiting too much AGEs and NO formation and reducing damage in the brain cells.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Litchi/química , Trastornos de la Memoria/tratamiento farmacológico , Memoria/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Semillas/química , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/administración & dosificación , Femenino , Galactosa/farmacología , Productos Finales de Glicación Avanzada/sangre , Discapacidades para el Aprendizaje/tratamiento farmacológico , Discapacidades para el Aprendizaje/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/metabolismo , Ratones , Óxido Nítrico/metabolismo , Extractos Vegetales/administración & dosificación , Extractos Vegetales/farmacología , Distribución Aleatoria
19.
J Med Food ; 16(4): 280-7, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23514232

RESUMEN

The present study aimed to evaluate the effect of kai xin san (KXS, at doses of 500, 250, and 125 mg/kg body weight per day), a well-known traditional Chinese medicine, on learning and memory in paradoxical sleep deprivation (PSD)-induced cognition deficit rats. Two behavior tests (the Open Field test and the Morris water maze task) were used for testing the effects of KXS on a PSD-induced learning and memory deficit model. Furthermore, its effect on the glutamic acid (GLU) and γ-amino-butyric acid (GABA) levels in the brain tissue, brain-derived neurotrophic factor (BDNF), cyclic AMP response element binding protein (CREB), and phosphorylated-CREB (p-CREB) expression in the hippocampus was also tested. KXS exerted the greatest cognition against the 48 h PSD-induced cognitive deficit and these effects may be mediated by decreasing the GLU and GABA levels and increasing the levels of BDNF, CREB, and p-CREB. This study indicates that the effect of KXS on learning and memory in a rat model of PSD could be associated with the modulation of neurotransmitter levels and the expression of some genes in the brain that contribute to memory functions.


Asunto(s)
Encéfalo/efectos de los fármacos , Trastornos del Conocimiento/tratamiento farmacológico , Medicamentos Herbarios Chinos/uso terapéutico , Discapacidades para el Aprendizaje/tratamiento farmacológico , Trastornos de la Memoria/tratamiento farmacológico , Privación de Sueño/complicaciones , Sueño REM , Animales , Conducta Animal/efectos de los fármacos , Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Medicamentos Herbarios Chinos/farmacología , Expresión Génica , Ácido Glutámico/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Aprendizaje/efectos de los fármacos , Discapacidades para el Aprendizaje/etiología , Discapacidades para el Aprendizaje/metabolismo , Masculino , Memoria/efectos de los fármacos , Trastornos de la Memoria/etiología , Trastornos de la Memoria/metabolismo , Fitoterapia , Ratas , Ratas Sprague-Dawley , Privación de Sueño/metabolismo , Ácido gamma-Aminobutírico/metabolismo
20.
Phytomedicine ; 20(1): 84-8, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23079229

RESUMEN

The rhizome of Codonopsis lanceolata (family Campanulaceae), which contains lancemaside A as a main constituent, has been used as herbal medicine to treat inflammation, insomnia, and hypomnesia. Lancemaside A and echinocystic acid, which is its metabolite by intestinal microflora, potently inhibited acetylcholinesterase activity in a dose-dependent manner, with IC50 value 13.6 µM and 12.2 µM, respectively. Its inhibitory potency is comparable with that of donepezil (IC50=10.9 µM). Lancemaside A and echinocystic acid significantly reversed scopolamine-induced memory and learning deficits on passive avoidance task. Lancemaside A orally administered 5h before treatment with scopolamine reversed scopolamine-induced memory and learning deficits more potently than one orally administered 1h before. Echinocystic acid more potently reversed it than lancemaside A. Lancemaside A and echinocystic acid significantly reversed scopolamine-induced memory and learning deficits on the Y-maze and Morris water maze tasks. Lancemaside A and echinocystic acid also increased the expression of brain-derived neurotrophic factor (BDNF) and phosphorylated cAMP response element binding protein (p-CREB). Based on these findings, orally administered lancemaside A may be metabolized to echinocystic acid, which may be absorbed into the blood and ameliorate memory and learning deficits by inhibiting AChE activity and inducing BDNF and p-CREB expressions.


Asunto(s)
Acetilcolinesterasa/metabolismo , Inhibidores de la Colinesterasa/uso terapéutico , Codonopsis/química , Discapacidades para el Aprendizaje/tratamiento farmacológico , Trastornos de la Memoria/tratamiento farmacológico , Ácido Oleanólico/análogos & derivados , Fitoterapia , Animales , Reacción de Prevención/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Inhibidores de la Colinesterasa/aislamiento & purificación , Inhibidores de la Colinesterasa/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Donepezilo , Relación Dosis-Respuesta a Droga , Indanos/farmacología , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Discapacidades para el Aprendizaje/inducido químicamente , Discapacidades para el Aprendizaje/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/metabolismo , Ratones , Ratones Endogámicos ICR , Ácido Oleanólico/aislamiento & purificación , Ácido Oleanólico/metabolismo , Ácido Oleanólico/farmacología , Ácido Oleanólico/uso terapéutico , Piperidinas/farmacología , Extractos Vegetales/química , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Rizoma , Escopolamina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA