Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 235
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Comp Neurol ; 529(4): 657-693, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32621762

RESUMEN

The parabrachial nucleus (PB) is a complex structure located at the junction of the midbrain and hindbrain. Its neurons have diverse genetic profiles and influence a variety of homeostatic functions. While its cytoarchitecture and overall efferent projections are known, we lack comprehensive information on the projection patterns of specific neuronal subtypes in the PB. In this study, we compared the projection patterns of glutamatergic neurons here with a subpopulation expressing the transcription factor Foxp2 and a further subpopulation expressing the neuropeptide Pdyn. To do this, we injected an AAV into the PB region to deliver a Cre-dependent anterograde tracer (synaptophysin-mCherry) in three different strains of Cre-driver mice. We then analyzed 147 neuroanatomical regions for labeled boutons in every brain (n = 11). Overall, glutamatergic neurons in the PB region project to a wide variety of sites in the cerebral cortex, basal forebrain, bed nucleus of the stria terminalis, amygdala, diencephalon, and brainstem. Foxp2 and Pdyn subpopulations project heavily to the hypothalamus, but not to the cortex, basal forebrain, or amygdala. Among the few differences between Foxp2 and Pdyn cases was a notable lack of Pdyn projections to the ventromedial hypothalamic nucleus. Our results indicate that genetic identity determines connectivity (and therefore, function), providing a framework for mapping all PB output projections based on the genetic identity of its neurons. Using genetic markers to systematically classify PB neurons and their efferent projections will enhance the translation of research findings from experimental animals to humans.


Asunto(s)
Encefalinas/biosíntesis , Factores de Transcripción Forkhead/biosíntesis , Núcleos Parabraquiales/metabolismo , Precursores de Proteínas/biosíntesis , Proteínas Represoras/biosíntesis , Proteína 2 de Transporte Vesicular de Glutamato/biosíntesis , Animales , Tronco Encefálico/química , Tronco Encefálico/metabolismo , Corteza Cerebral/química , Corteza Cerebral/metabolismo , Vías Eferentes/química , Vías Eferentes/metabolismo , Encefalinas/análisis , Encefalinas/genética , Femenino , Factores de Transcripción Forkhead/análisis , Factores de Transcripción Forkhead/genética , Hipotálamo/química , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Núcleos Parabraquiales/química , Precursores de Proteínas/análisis , Precursores de Proteínas/genética , Proteínas Represoras/análisis , Proteínas Represoras/genética , Tálamo/química , Tálamo/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/análisis , Proteína 2 de Transporte Vesicular de Glutamato/genética
2.
Artículo en Inglés | MEDLINE | ID: mdl-33271211

RESUMEN

Wernicke-Korsakoff Syndrome (WKS) is a neuropsychiatric disorder whose etiology is a thiamine deficiency (TD), with alcoholism being the main underlying cause. Previous evidence suggests the presence of initial neuroinflammation and oxidative/nitrosative stress in the physiopathology, although the specific molecular mechanisms underlying TD-induced brain damage and behavioral disabilities are unknown. We explored the specific role of the innate immune receptor TLR4 in three murine models of WKS, based on the combination of a thiamine-deficient diet and pyrithiamine injections (0.25 mg/kg, i.p.) over time. The Symptomatic Model (SM) allowed us to describe the complete neurological/neurobehavioral symptomatology over 16 days of TD. Animals showed an upregulation of the TLR4 signaling pathway both in the frontal cortex (FC) and cerebellum and clear motor impairments related with cerebellar dysfunction. However, in the Pre-Symptomatic Model (PSM), 12 days of TD induced the TLR4 pathway upregulation in the FC, which correlated with disinhibited-like behavior, but not in the cerebellum, and no motor impairments. In addition, we tested the effects of the biolipid oleoylethanolamide (OEA, 10 mg/kg, i.p., once daily, starting before any symptom of the pathology is manifested) through the Glucose-Precipitated Model (GPM), which was generated by glucose loading (5 g/kg, i.v., last day) in thiamine-deficient animals to accelerate damage. Pretreatment with OEA prevented the TLR4-induced signature in the FC, as well as an underlying incipient memory disability and disinhibited-like behavior. This study suggests a key role for TLR4 in TD-induced neuroinflammation in the FC and cerebellum, and it reveals different vulnerability of these brain regions in WKS over time. Pre-treatment with OEA counteracts TD-induced TLR4-associated neuroinflammation and may serve as co-adjuvant therapy to prevent WKS-induced neurobehavioral alterations.


Asunto(s)
Cerebelo/metabolismo , Corteza Cerebral/metabolismo , Endocannabinoides/uso terapéutico , Síndrome de Korsakoff/tratamiento farmacológico , Ácidos Oléicos/uso terapéutico , Receptor Toll-Like 4/metabolismo , Animales , Cerebelo/química , Corteza Cerebral/química , Citocinas/análisis , Citocinas/metabolismo , Modelos Animales de Enfermedad , Prueba de Laberinto Elevado , Masculino , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/etiología , Prueba de Campo Abierto , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Prueba de Desempeño de Rotación con Aceleración Constante , Deficiencia de Tiamina/complicaciones , Receptor Toll-Like 4/análisis
3.
J Neurosci ; 40(30): 5785-5796, 2020 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-32532890

RESUMEN

The cerebral cortex, with all its computational power, can only influence behavior via corticofugal connections originating from layer 5 (L5) cells (Sherman and Guillery, 2013). To begin to establish the global pattern of these outputs, we examined L5 efferents originating from four cortical areas: somatosensory, visual, motor, and prefrontal (i.e., ventromedial orbitofrontal) cortex. We injected Cre-dependent adeno-associated virus in an Rbp4-Cre transgenic mouse line (both sexes) to label these L5 efferents selectively. Our study reveals that, across this diverse series of cortical regions, L5 commonly projects to multiple thalamic and extrathalamic sites. We also identified several novel corticofugal targets (i.e., the lateral dorsal nucleus, submedial nucleus) previously unidentified as L5 targets. We identified common patterns for these projections: all areas innervated both thalamus and the midbrain, and all areas innervated multiple thalamic targets, including those with core and matrix cell types (Jones, 1998). An examination of the terminal size within each of these targets suggests that terminal populations of L5 efferents are not consistently large but vary with cortical area and target; and in some cases, these include small terminals only. Overall, our data reveal more widespread and diverse L5 efferents than previously appreciated, suggesting a generalizable role for this cortical layer in influencing motor commands and cognitive processes.SIGNIFICANCE STATEMENT While the neocortex is responsible for coordination of complex behavior, it requires communication with subcortical regions to do so. It is specifically cortical layer 5 (L5) that is thought to underlie these behaviors, although it is unknown whether this holds true across functionally different cortical areas. Using a selective viral tracing method and transgenic mice, we examined the connectivity of four cortical regions (somatosensory, visual, motor and prefrontal cortex) to assess the generalizability of these L5 projections. All areas of cortex projected to overlapping as well as distinct thalamic and brainstem structures. Terminals within these regions varied in size, implicating that L5 has a broad and diverse impact on behavior.


Asunto(s)
Corteza Cerebral/química , Corteza Cerebral/fisiología , Tálamo/química , Tálamo/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Vías Nerviosas/química , Vías Nerviosas/fisiología
4.
Lipids Health Dis ; 19(1): 104, 2020 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-32450867

RESUMEN

BACKGROUND: Glycerophospholipids were the main components of cerebral cortex lipids, and there was a close association between lipid homeostasis and human health. It has been reported that dietary DHA-enriched phosphatidylcholine (DHA-PC) and phosphatidylserine (DHA-PS) could improve brain function. However, it was unclear that whether supplementation of DHA-PC and DHA-PS could change lipid profiles in the brain of dementia animals. METHODS: SAMP8 mice was fed with different diet patterns for 2 months, including high-fat diet and low-fat diet. After intervention with DHA-PC and DHA-PS for another 2 months, the lipid profile in cerebral cortex was determined by lipidomics in dementia mice. RESULTS: High-fat diet could significantly decrease the levels of DHA-containing PS/pPE, DPA-containing PS, and AA-containing PE, which might exhibit the potential of lipid biomarkers for the prevention and diagnosis of AD. Notably, DHA-PC and DHA-PS remarkably recovered the lipid homeostasis in dementia mice. These might provide a potential novel therapy strategy and direction of dietary intervention for patients with cognitive decline. CONCLUSIONS: DHA-PC and DHA-PS could recover the content of brain DHA-containing PS and pPE in SAMP8 mice fed with high-fat diet.


Asunto(s)
Corteza Cerebral/química , Dieta Alta en Grasa , Ácidos Docosahexaenoicos/análisis , Fosfatidilcolinas/química , Fosfatidilserinas/análisis , Plasmalógenos/análisis , Enfermedad de Alzheimer , Animales , Corteza Cerebral/efectos de los fármacos , Modelos Animales de Enfermedad , Lipidómica , Masculino , Ratones , Fosfatidilcolinas/farmacología , Fosfatidilserinas/química , Fosfatidilserinas/metabolismo , Fosfatidilserinas/farmacología , Plasmalógenos/química , Plasmalógenos/metabolismo
5.
Neuromolecular Med ; 22(3): 341-358, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32048142

RESUMEN

Alzheimer's disease (AD) is a common neurodegenerative disease that is always accompanied by synaptic loss in the brain. Safflower yellow (SY) is the extract of safflower, a traditional Chinese medicine, which has shown neuroprotective effects in recent studies. However, the mechanism of SY in protecting synapses remains unclear. In this study, we are going to study the mechanism of how SY treats AD in terms of synaptic plasticity. We found, via behavioral experiments, that SY treatment could improve the abilities of learning and memory in APP/PS1 mice. In addition, using Golgi staining and HE staining, we found that SY treatment could reduce the loss of dendritic spines in the pathological condition and could maintain the normal physiological state of the cells in cortex and in hippocampus. In addition, the results of immunofluorescence staining and western blotting showed that SY treatment could significantly increase the expression of synapse-related proteins. Moreover, after being treated with SY, the expression of iNOS (marker of M1 microglia) declined remarkably, and the level of Arginase-1 (marker of M2 microglia) increased significantly. Finally, we found BDNF/TrkB/ERK signaling cascade was activated. These results indicate that SY enhances synaptic plasticity in APP/PS1 mice by regulating microglia activation phenotypes and BDNF/TrkB/ERK signaling pathway.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Factor Neurotrófico Derivado del Encéfalo/fisiología , Chalcona/análogos & derivados , Medicamentos Herbarios Chinos/uso terapéutico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Glicoproteínas de Membrana/fisiología , Microglía/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Fitoterapia , Proteínas Tirosina Quinasas/fisiología , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Arginasa/biosíntesis , Arginasa/genética , Corteza Cerebral/química , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Chalcona/uso terapéutico , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/ultraestructura , Modelos Animales de Enfermedad , Donepezilo/farmacología , Donepezilo/uso terapéutico , Inducción Enzimática/efectos de los fármacos , Reacción de Fuga/efectos de los fármacos , Femenino , Hipocampo/química , Hipocampo/efectos de los fármacos , Hipocampo/patología , Masculino , Memoria a Largo Plazo/efectos de los fármacos , Memoria a Corto Plazo/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/fisiología , Prueba del Laberinto Acuático de Morris/efectos de los fármacos , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Plasticidad Neuronal/fisiología , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Óxido Nítrico Sintasa de Tipo II/genética , Presenilina-1/genética , Distribución Aleatoria
6.
Basic Clin Pharmacol Toxicol ; 125(5): 430-438, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31222944

RESUMEN

The main sites of the analgesic action of oxycodone are the brain and spinal cord. The present study describes the concentrations of oxycodone and its metabolites in the brain and spinal cord after epidural administration to the ewe. Twenty pregnant ewes undergoing laparotomy were randomized into two groups to receive epidural oxycodone: infusion group (n = 10, 0.1 mg·kg-1 bolus followed by continuous infusion of 0.05 mg·kg-1 ·h-1 for five days) or repeated boluses group (n = 10, 0.2 + 2x0.1 mg·kg-1 bolus followed by a 0.2 mg·kg-1 bolus every 12 hours for five days). After five days of oxycodone administration, arterial blood samples were collected, the sheep were killed, and a CSF sample and tissue samples from the cortex, thalamus, cerebellum and spinal cord were obtained for the quantification of oxycodone and its main metabolites. The median plasma and CSF concentrations of oxycodone were 9.0 and 14.2 ng·mL-1 after infusion and 0.4 and 1.1 ng·mL-1 after repeated boluses. In the infusion group, the cortex, thalamus and cerebellum oxycodone concentrations were 4-8 times higher and in the spinal cord 1310 times higher than in plasma. In the repeated boluses group, brain tissue concentrations were similar in the three areas, and in the spinal cord were 720 times higher than in plasma. Oxymorphone was the main metabolite detected, which accumulated in the brain and spinal cord tissue. In conclusion, first, accumulation of oxycodone and oxymorphone in the CNS was observed, and second, high spinal cord concentrations suggest that epidural oxycodone may provide segmental analgesia.


Asunto(s)
Analgésicos Opioides/farmacocinética , Química Encefálica , Oxicodona/farmacocinética , Oximorfona/farmacocinética , Médula Espinal/química , Analgesia/métodos , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/sangre , Analgésicos Opioides/líquido cefalorraquídeo , Animales , Cerebelo/química , Corteza Cerebral/química , Femenino , Inyecciones Epidurales , Modelos Animales , Oxicodona/administración & dosificación , Oxicodona/sangre , Oxicodona/líquido cefalorraquídeo , Oximorfona/sangre , Oximorfona/líquido cefalorraquídeo , Embarazo , Ovinos , Tálamo/química , Distribución Tisular
7.
Nutrients ; 11(5)2019 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-31121888

RESUMEN

The pharmacological properties of Eleutherococcus senticosus leaf have not been clarified although it is taken as a food item. In this study, the effects of water extract of Eleutherococcus senticosus leaves on memory function were investigated in normal mice. Oral administration of the extract for 17 days significantly enhanced object recognition memory. Compounds absorbed in blood and the brain after oral administration of the leaf extract were detected by LC-MS/MS analyses. Primarily detected compounds in plasma and the cerebral cortex were ciwujianoside C3, eleutheroside M, ciwujianoside B, and ciwujianoside A1. Pure compounds except for ciwujianoside A1 were administered orally for 17 days to normal mice. Ciwujianoside C3, eleutheroside M, and ciwujianoside B significantly enhanced object recognition memory. These results demonstrated that oral administration of the leaf extract of E. senticosus enhances memory function, and that active ingredients in the extract, such as ciwujianoside C3, eleutheroside M, and ciwujianoside B, were able to penetrate and work in the brain. Those three compounds as well as the leaf extract had dendrite extension activity against primary cultured cortical neurons. The effect might relate to memory enhancement.


Asunto(s)
Encéfalo/efectos de los fármacos , Eleutherococcus/química , Memoria/efectos de los fármacos , Extractos Vegetales/farmacología , Hojas de la Planta/química , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Células Cultivadas , Corteza Cerebral/química , Corteza Cerebral/embriología , Corteza Cerebral/ultraestructura , Dendritas/efectos de los fármacos , Dendritas/fisiología , Glicósidos/análisis , Glicósidos/farmacocinética , Glicósidos/farmacología , Masculino , Ratones , Extractos Vegetales/análisis , Extractos Vegetales/farmacocinética , Saponinas/análisis , Saponinas/farmacocinética , Saponinas/farmacología
8.
Spectrochim Acta A Mol Biomol Spectrosc ; 220: 117128, 2019 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-31146210

RESUMEN

Aging brain is characterized by a change in biomolecular composition leading to a diverse range of neurological diseases. Anti-aging research is of current interest, to lessen the burden of age-related macromolecular damage through antioxidant supplementation and caloric restriction. However, data concerning the effect of these anti-aging regimens on age-related biomolecular changes in rat brain is still lacking. In the present study, for the first time, we employed Fourier transform infrared (FTIR) spectroscopy, to investigate the effect of quercetin, caloric restriction (CR) and combination of both on alterations in the composition of lipids and proteins of aged rat brain cerebral cortex. Aged male Wistar rats (21 months old) were divided into four groups: Control (CONT), fed pellet diet; Quercetin (QUER), fed quercetin (50 mg/kg/day); CR (caloric restriction) (fed 40% reduced CONT), and CRQ (40% CR and 50 mg/kg/day QUER). Three-month-old rats served as young control (YOUNG). Our short-term study (45 days) shows decreased band area of unsaturated lipids, decreased area ratios of olefinic/lipid and CH2 antisymmetric stretching (2925 cm-1)/lipids in CONT group compared to young rats, suggesting age-associated lipid peroxidation in aged rats. A slight decrease in the frequency of CH2 antisymmetric mode of lipids (whereas no change in CH2 symmetric mode), but a decrease in bandwidths of both CH2 antisymmetric and symmetric modes of lipids was observed for CONT group compared to YOUNG. Further, a significant decrease in the peak area of infrared bands of proteins and an increase in the peak area of the CO band of lipids was observed in the CONT group. Our data also show that lower levels of α-helical structures and higher levels of random coils, representing altered protein secondary structure composition in the CONT group compared to YOUNG group. Reduction in neuronal cell density and shrinked nucleus was also observed in aged rats. Increase in the accumulation of oxidative mediated damage to macromolecules and diminished antioxidant levels, could be the possible reason for the age-related alterations in the composition of lipids and proteins. However, the combination of quercetin and CR, but not either treatment alone, significantly prevented the age associated alterations in the lipid and protein profiles in the rat cerebral cortex. Further, our results help to understand the mechanism of action of antioxidants under non-restriction and CR conditions, this might help in the development of novel anti-aging treatments to ameliorate oxidative stress in age-related disorders.


Asunto(s)
Envejecimiento/efectos de los fármacos , Restricción Calórica , Corteza Cerebral/química , Lípidos/química , Quercetina/farmacología , Envejecimiento/fisiología , Amidas/química , Animales , Recuento de Células , Corteza Cerebral/efectos de los fármacos , Suplementos Dietéticos , Peroxidación de Lípido/efectos de los fármacos , Masculino , Neuronas/citología , Neuronas/efectos de los fármacos , Proteínas/química , Ratas Wistar , Espectroscopía Infrarroja por Transformada de Fourier
9.
Molecules ; 24(9)2019 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-31058813

RESUMEN

INTRODUCTION: Alcohol overuse may be related to increased aluminum (Al) exposure, the brain accumulation of which contributes to dementia. However, some reports indicate that silicon (Si) may have a protective role over Al-induced toxicity. Still, no study has ever explored the brain content of Al and Si in alcoholic use disorder (AUD). MATERIALS AND METHODS: To fill this gap, the present study employed inductively coupled plasma optical emission spectrometry to investigate levels of Al and Si in 10 brain regions and in the liver of AUD patients (n = 31) and control (n = 32) post-mortem. RESULTS: Al content was detected only in AUD patients at mean ± SD total brain content of 1.59 ± 1.19 mg/kg, with the highest levels in the thalamus (4.05 ± 12.7 mg/kg, FTH), inferior longitudinal fasciculus (3.48 ± 9.67 mg/kg, ILF), insula (2.41 ± 4.10 mg/kg) and superior longitudinal fasciculus (1.08 ± 2.30 mg/kg). Si content displayed no difference between AUD and control, except for FTH. Positive inter-region correlations between the content of both elements were identified in the cingulate cortex, hippocampus, and ILF. CONCLUSIONS: The findings of this study suggest that AUD patients may potentially be prone to Al-induced neurodegeneration in their brain-although this hypothesis requires further exploration.


Asunto(s)
Alcoholismo/complicaciones , Aluminio/análisis , Química Encefálica , Enfermedades Neurodegenerativas/diagnóstico , Silicio/análisis , Adulto , Anciano , Aluminio/toxicidad , Autopsia , Estudios de Casos y Controles , Corteza Cerebral/química , Femenino , Humanos , Masculino , Persona de Mediana Edad , Enfermedades Neurodegenerativas/inducido químicamente , Espectrofotometría Atómica , Tálamo/química
10.
Fitoterapia ; 134: 165-171, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30825572

RESUMEN

Two novel phenanthrenoids, juncuenin H (1) and dijuncuenin B (2), together with eight known phenanthrenoids, effusol (3), dehydroeffusol (4), juncusol (5), dehydrojuncusol (6), juncuenin B (7), dehydrojuncuenin B (8), juncuenin A (9), and dehydrojuncuenin A (10), were isolated from the underground parts of Juncus setchuenensis. The structures of the compounds were determined by 1D and 2D NMR and mass spectroscopy. The anxiolytic activities of compounds 1, 6, 9, and 10 were evaluated. In order to explore the mechanisms underlying their anxiolytic activities, the levels of serotonin (5-HT), dopamine (DA), and their metabolites in the cerebral cortex and hippocampus of mice treated with compound 1 were determined by quantitative mass spectrometry. The mice treated with compound 1 had significantly lower levels of 5-HT, 3-methoxytyramine (3-MT), 5-hydroxyindole-3-acetic acid (5-HIAA), homovanillic acid (HVA), and 3, 4-dihydroxyphenylacetic acid (DOPAC) in the cerebral cortex than those of the vehicle control-treated mice. The levels of HVA and 5-HIAA in the hippocampus were also significantly lower in the mice treated with compound 1 than in the control group mice. These results suggest that the metabolic changes, reflected in the levels of DA and/or 5-HT, may contribute to the anxiolytic activity of the phenanthrenoids studied herein.


Asunto(s)
Ansiolíticos/farmacología , Corteza Cerebral/efectos de los fármacos , Hipocampo/efectos de los fármacos , Magnoliopsida/química , Fenantrenos/farmacología , Ácido 3,4-Dihidroxifenilacético/análisis , Animales , Ansiolíticos/aislamiento & purificación , Corteza Cerebral/química , China , Dopamina/análogos & derivados , Dopamina/análisis , Hipocampo/química , Ácido Homovanílico/análisis , Masculino , Ratones , Estructura Molecular , Fenantrenos/aislamiento & purificación , Fitoquímicos/aislamiento & purificación , Fitoquímicos/farmacología , Raíces de Plantas/química , Serotonina/análisis
11.
Drug Chem Toxicol ; 42(2): 167-175, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29745257

RESUMEN

This study was conducted to investigate protective effects of Urtica dioica extract on acetylcholinesterase (AChE) activity and the oxidative damage of brain tissues in scopolamine-induced memory impairment model. The rats were treated with (1) saline (control), (2) scopolamine, and (3-5) the plant extract (20, 50, or 100 mg/kg) before scopolamine. The traveled distance and the latency to find the platform in Morris water maze (MWM) by scopolamine-treated group were longer while the time spent in target quadrant was shorter than those of the control. Scopolamine decreased the latency to enter the dark in passive avoidance test. Besides, it also increased AChE activity and malondialdehyde (MDA) concentration in the hippocampal and cortical tissues while decreased thiols content and superoxide dismutase (SOD) and catalase (CAT) activities in the brain (p < 0.01-p <0.001). Treatment by the extract reversed all the effects of scopolamine (p < 0.05-p <0.001). According to the results of present study, the beneficial effects of U. dioica on memory can be attributed to its protective effects on oxidative damage of brain tissue and AChE activity.


Asunto(s)
Acetilcolinesterasa/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/farmacología , Escopolamina/farmacología , Urtica dioica/química , Acetilcolinesterasa/metabolismo , Animales , Química Encefálica/efectos de los fármacos , Corteza Cerebral/química , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/enzimología , Hipocampo/química , Hipocampo/efectos de los fármacos , Hipocampo/enzimología , Masculino , Malondialdehído/análisis , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/inducido químicamente , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Wistar , Superóxido Dismutasa/metabolismo
12.
Undersea Hyperb Med ; 44(4): 345-355, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28783891

RESUMEN

This study aimed to explore the effects of hyperbaric oxygen (HBO2) on blood-brain barrier (BBB) integrity in rats, when administered for one (at 2.5 ATA, 3 HBO2 sessions a day) and five days (at 2.5 ATA, 3 HBO2 sessions a day for the first two days, and twice a day for the last three days). Horseradish peroxidase (HRP) was used to evaluate the BBB permeability. Superoxide dismutase (SOD) activity, glutathione (GSH) and malondialdehyde (MDA) levels were measured in the cerebral cortex and hippocampus regions. Frequent vesicles containing HRP reaction products were observed in capillary endothelial cells in the cerebral cortex and hippocampus of rats subjected to HBO2. The accumulation of HRP reaction products in these brain regions was significantly higher than that of control animals (P ⟨ 0.01). In animals that received HBO2, MDA levels (P ⟨ 0.01 for five days) and GSH (p ⟨ 0.05 for one day, and P ⟨ 0.01 for five days) were decreased in the cerebral cortex, whereas SOD activities slightly increased in this region. In animals that received HBO2 significant decreases in MDA (P ⟨ 0.05 for one day; P ⟨ 0.01 for five days) and GSH (P ⟨ 0.05 for five days) levels were observed in the hippocampus region, but SOD activities decreased in this region. We showed that HBO2 administered with the doses described above impaired BBB integrity in otherwise healthy rats. Therefore, we suggest that the results of this study should be taken into consideration when patients are exposed to HBO2 with the same doses.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Corteza Cerebral/química , Glutatión Peroxidasa/análisis , Hipocampo/química , Oxigenoterapia Hiperbárica/efectos adversos , Malondialdehído/análisis , Superóxido Dismutasa/análisis , Animales , Capilares/ultraestructura , Corteza Cerebral/irrigación sanguínea , Femenino , Hipocampo/irrigación sanguínea , Peroxidasa de Rábano Silvestre/farmacocinética , Oxigenoterapia Hiperbárica/métodos , Microscopía Electrónica , Permeabilidad , Ratas , Ratas Wistar , Factores de Tiempo
13.
J Mol Histol ; 48(2): 133-146, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28255782

RESUMEN

Galectin-3 is a member of the lectin subfamily that enables the specific binding of ß-galactosides. It is expressed in a broad spectrum of species and organs, and is known to have various functions related to cell adhesion, signal transduction, and proinflammatory responses. Although, expression of galectin-3 in some activated neuroglia under neuroinflammation has been well documented in the central nervous system, little is known about the neuronal expression and distribution of galectin-3 in normal brain. To describe the cellular and neuroanatomical expression map of galectin-3, we performed galectin-3 immunohistochemistry on the entire normal rat brain and subsequently analyzed the neuronal distribution. Galectin-3 expression was observed not only in some neuroglia but also in neurons. Neuronal expression of galectin-3 was observed in many functional parts of the cerebral cortex and various other subcortical nuclei in the hypothalamus and brainstem. Neuroanatomical analysis revealed that robust galectin-3 immuno-signals were present in many hypothalamic nuclei related to a variety of physiological functions responsible for mediating anxiety responses, energy balance, and neuroendocrine regulation. In addition, the regions highly connected with these hypothalamic nuclei also showed intense galectin-3 expression. Moreover, multiple key regions involved in regulating autonomic functions exhibited high levels of galectin-3 expression. In contrast, the subcortical nuclei responsible for the control of voluntary motor functions and limbic system exhibited no galectin-3 immunoreactivity. These observations suggest that galectin-3 expression in the rat brain seems to be regulated by developmental cascades, and that functionally and neuroanatomically related brain nuclei constitutively express galectin-3 in adulthood.


Asunto(s)
Encéfalo/anatomía & histología , Galectina 3/análisis , Neuronas/química , Factores de Edad , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/fisiología , Tronco Encefálico/química , Núcleo Celular/química , Corteza Cerebral/química , Hipotálamo/química , Inmunohistoquímica , Neuroglía/química , Ratas
14.
Food Funct ; 8(3): 1271-1281, 2017 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-28232989

RESUMEN

Herein, we investigated the potential relationship between sphingolipids and Alzheimer's disease (AD) with special attention to the relationship between dietary sea cucumber glucocerebrosides (SCGs) and sphingolipid metabolism. We assessed animal behavior using the Morris water maze test, determined Aß1-42 concentration in the hippocampus using ELISA, and assessed the sphingolipid profile of the hippocampus and the cortex in normal mice (SAMR1), AD mice (SAMP8), and AD mice (SAMP8) fed with SCG using liquid chromatography-triple quadrupole mass spectrometry. We found that the SAMP8 mice had impaired memory and an SCG diet significantly rescued spatial memory deficits in these mice. As expected, we found that the profiles of sphingolipid species and the levels of total cerebrosides (CBS), ceramides (Cer), and sulfatides (ST) were significantly different in both the hippocampus and the cortex between the three groups; moreover, there were significantly lower ST levels and higher Cer and CBS levels in these regions in the SAMP8 mice. In the AD-SCG group, Cer and ST levels were altered only in the hippocampus, in contrast to the AD group. The major molecular species ST (d18:1-C24:1) and Cer (d18:1/18:0) were especially different between those of the two groups. Unexpectedly, sphingolipid profiles, including the nonhydroxylated fatty acid-ST/hydroxylated fatty acid-ST, very long fatty acid-galactocerebroside/long fatty acid-galactocerebroside, nonhydroxylated fatty acid-galactocerebroside/hydroxylated fatty acid-galactocerebroside and galactocerebroside/glucocerebroside ratios, were affected by AD. Thus, the ST and Cer levels and the profiles of sphingolipid species in the AD-SCG group were significantly different compared to those of the AD model group. Therefore, SCG has potential ameliorative effects in AD, and exogenous sphingolipid intake may potentially influence sphingolipid metabolism in vivo.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Corteza Cerebral/metabolismo , Glucosilceramidas/farmacología , Hipocampo/metabolismo , Pepinos de Mar/química , Esfingolípidos/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/psicología , Animales , Corteza Cerebral/química , Corteza Cerebral/efectos de los fármacos , Suplementos Dietéticos/análisis , Modelos Animales de Enfermedad , Hipocampo/química , Hipocampo/efectos de los fármacos , Humanos , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Transgénicos
15.
Artículo en Inglés | MEDLINE | ID: mdl-27926457

RESUMEN

The relative amounts of arachidonic acid (AA) and docosahexaenoic acid (DHA) govern the different functions of the brain. Their brain levels depend on structures considered, on fatty acid dietary supply and the age of animals. To have a better overview of the different models available in the literature we here compared the brain fatty acid composition in various mice models (C57BL/6J, CD1, Fat-1, SAMP8 mice) fed with different n-3 PUFA diets (deficient, balanced, enriched) in adults and aged animals. Our results demonstrated that brain AA and DHA content is 1) structure-dependent; 2) strain-specific; 3) differently affected by dietary approaches when compared to genetic model of PUFA modulation; 4) different in n-3 PUFA deficient aged C57BL6/J when compared to SAMP8 mouse model of aging. From these experiments, we highlight the difficulty to compare results obtained in different mouse models, different strains, different brain regions and different ages.


Asunto(s)
Ácido Araquidónico/química , Química Encefálica , Grasas Insaturadas en la Dieta/administración & dosificación , Ácidos Docosahexaenoicos/química , Animales , Tronco Encefálico/química , Cerebelo/química , Corteza Cerebral/química , Femenino , Hipocampo/química , Hipotálamo/química , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Teóricos , Corteza Prefrontal/química
16.
Amino Acids ; 47(5): 1053-63, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25691144

RESUMEN

TFF3 is a member of the trefoil factor family (TFF) predominantly secreted by mucous epithelia. Minute amounts are also expressed in the immune system and the brain. In the latter, particularly the hypothalamo-pituitary axis has been investigated in detail in the past. Functionally, cerebral TFF3 has been reported to be involved in several processes such as fear, depression, learning and object recognition, and opiate addiction. Furthermore, TFF3 has been linked with neurodegenerative and neuropsychiatric disorders (e.g., Alzheimer's disease, schizophrenia, and alcoholism). Here, using immunohistochemistry, a systematic survey of the TFF3 localization in the adult human brain is presented focusing on extrahypothalamic brain areas. In addition, the distribution of TFF3 in the developing human brain is described. Taken together, neurons were identified as the predominant cell type to express TFF3, but to different extent; TFF3 was particularly enriched in various midbrain and brain stem nuclei. Besides, TFF3 immunostaining staining was observed in oligodendroglia and the choroid plexus epithelium. The wide cerebral distribution should help to explain its multiple effects in the CNS.


Asunto(s)
Plexo Coroideo/metabolismo , Mesencéfalo/metabolismo , Neuronas/metabolismo , Oligodendroglía/metabolismo , Péptidos/genética , Aborto Espontáneo , Adulto , Amígdala del Cerebelo/química , Amígdala del Cerebelo/metabolismo , Mapeo Encefálico , Cerebelo/química , Cerebelo/metabolismo , Corteza Cerebral/química , Corteza Cerebral/metabolismo , Plexo Coroideo/química , Femenino , Feto , Expresión Génica , Hipocampo/química , Hipocampo/metabolismo , Humanos , Hipotálamo/química , Hipotálamo/metabolismo , Inmunohistoquímica , Masculino , Mesencéfalo/química , Persona de Mediana Edad , Neuronas/química , Oligodendroglía/química , Especificidad de Órganos , Péptidos/metabolismo , Hipófisis/química , Hipófisis/metabolismo , Neurohipófisis/química , Neurohipófisis/metabolismo , Factor Trefoil-3 , Sustancia Blanca/química , Sustancia Blanca/metabolismo
17.
NMR Biomed ; 27(10): 1167-75, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25088346

RESUMEN

The (1)H resonances of γ-aminobutyric acid (GABA) in the human brain in vivo are extensively overlapped with the neighboring abundant resonances of other metabolites and remain indiscernible in short-TE MRS at 7 T. Here we report that the GABA resonance at 2.28 ppm can be fully resolved by means of echo time optimization of a point-resolved spectroscopy (PRESS) scheme. Following numerical simulations and phantom validation, the subecho times of PRESS were optimized at (TE, TE2) = (31, 61) ms for detection of GABA, glutamate (Glu), glutamine (Gln), and glutathione (GSH). The in vivo feasibility of the method was tested in several brain regions in nine healthy subjects. Spectra were acquired from the medial prefrontal, left frontal, medial occipital, and left occipital brain and analyzed with LCModel. Following the gray and white matter (GM and WM) segmentation of T1 -weighted images, linear regression of metabolite estimates was performed against the fractional GM contents. The GABA concentration was estimated to be about seven times higher in GM than in WM. GABA was overall higher in frontal than in occipital brain. Glu was about twice as high in GM as in WM in both frontal and occipital brain. Gln was significantly different between frontal GM and WM while being similar between occipital GM and WM. GSH did not show significant dependence on tissue content. The signals from N-acetylaspartylglutamate were clearly resolved, giving the concentration more than 10 times higher in WM than in GM. Our data indicate that the PRESS TE = 92 ms method provides an effective means for measuring GABA and several challenging J-coupled spin metabolites in human brain at 7 T.


Asunto(s)
Corteza Cerebral/química , Espectroscopía de Protones por Resonancia Magnética/métodos , Ácido gamma-Aminobutírico/análisis , Adulto , Corteza Cerebral/anatomía & histología , Colina/análisis , Simulación por Computador , Creatina/análisis , Dipéptidos/análisis , Estudios de Factibilidad , Femenino , Lóbulo Frontal/química , Glutamatos/análisis , Glutamina/análisis , Glutatión/análisis , Humanos , Masculino , Lóbulo Occipital/química , Fantasmas de Imagen , Corteza Prefrontal/química , Protones , Sustancia Blanca/química , Adulto Joven
18.
Brain Behav Immun ; 41: 22-31, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24735929

RESUMEN

Low dietary intake of the n-3 polyunsaturated fatty acids (PUFAs) is a causative factor of neurodevelopmental disorders. However the mechanisms linking n-3 PUFAs low dietary intake and neurodevelopmental disorders are poorly understood. Microglia, known mainly for their immune function in the injured or infected brain, have recently been demonstrated to play a pivotal role in regulating maturation of neuronal circuits during normal brain development. Disruption of this role during the perinatal period therefore could significantly contribute to psychopathologies with a neurodevelopmental neurodevelopmental component. N-3 PUFAs, essential lipids and key structural components of neuronal membrane phospholipids, are highly incorporated in cell membranes during the gestation and lactation phase. We previously showed that in a context of perinatal n-3 PUFAs deficiency, accretion of these latter is decreased and this is correlated to an alteration of endotoxin-induced inflammatory response. We thus postulated that dietary n-3 PUFAs imbalance alters the activity of microglia in the developing brain, leading to abnormal formation of neuronal networks. We first confirmed that mice fed with a n-3 PUFAs deficient diet displayed decreased n-3 PUFAs levels in the brain at post-natal days (PND)0 and PND21. We then demonstrated that n-3 PUFAs deficiency altered microglia phenotype and motility in the post-natal developing brain. This was paralleled by an increase in pro-inflammatory cytokines expression at PND21 and to modification of neuronal plasticity-related genes expression. Overall, our findings show for the first time that a dietary n-3 PUFAs deficiency from the first day of gestation leads to the development of a pro-inflammatory condition in the central nervous system that may contribute to neurodevelopmental alterations.


Asunto(s)
Encéfalo/inmunología , Ácidos Grasos Omega-3/fisiología , Regulación del Desarrollo de la Expresión Génica , Lípidos/deficiencia , Microglía/inmunología , Proteínas del Tejido Nervioso/biosíntesis , Plasticidad Neuronal/inmunología , Efectos Tardíos de la Exposición Prenatal , Animales , Recuento de Células , Movimiento Celular , Corteza Cerebral/química , Cruzamientos Genéticos , Citocinas/biosíntesis , Citocinas/genética , Grasas de la Dieta/administración & dosificación , Ácidos Grasos Omega-3/administración & dosificación , Ácidos Grasos Omega-3/análisis , Femenino , Aceites de Pescado , Hipocampo/inmunología , Hipocampo/metabolismo , Hipocampo/patología , Inmunidad Innata , Lactancia , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/fisiología , Proteínas del Tejido Nervioso/genética , Neuroinmunomodulación , Plasticidad Neuronal/genética , Aceites de Plantas/administración & dosificación , Embarazo , Aceite de Girasol
19.
Ann Neurol ; 75(1): 22-32, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24122892

RESUMEN

OBJECTIVE: A high incidence of structural brain abnormalities has been reported in individuals with pyridoxine-dependent epilepsy (PDE). PDE is caused by mutations in ALDH7A1, also known as antiquitin. How antiquitin dysfunction leads to cerebral dysgenesis is unknown. In this study, we analyzed tissue from a child with PDE as well as control human and murine brain to determine the normal distribution of antiquitin, its distribution in PDE, and associated brain malformations. METHODS: Formalin-fixed human brain sections were subjected to histopathology and fluorescence immunohistochemistry studies. Frozen brain tissue was utilized for measurement of PDE-associated metabolites and Western blot analysis. Comparative studies of antiquitin distribution were performed in developing mouse brain sections. RESULTS: Histologic analysis of PDE cortex revealed areas of abnormal radial neuronal organization consistent with type Ia focal cortical dysplasia. Heterotopic neurons were identified in subcortical white matter, as was cortical astrogliosis, hippocampal sclerosis, and status marmoratus of the basal ganglia. Highly elevated levels of lysine metabolites were present in postmortem PDE cortex. In control human and developing mouse brain, antiquitin immunofluorescence was identified in radial glia, mature astrocytes, ependyma, and choroid plexus epithelium, but not in neurons. In PDE cortex, antiquitin immunofluorescence was greatly attenuated with evidence of perinuclear accumulation in astrocytes. INTERPRETATION: Antiquitin is expressed within glial cells in the brain, and its dysfunction in PDE is associated with neuronal migration abnormalities and other structural brain defects. These malformations persist despite postnatal pyridoxine supplementation and likely contribute to neurodevelopmental impairments.


Asunto(s)
Aldehído Deshidrogenasa/biosíntesis , Corteza Cerebral/metabolismo , Epilepsia/diagnóstico , Epilepsia/metabolismo , Neuroglía/metabolismo , Adolescente , Animales , Animales Recién Nacidos , Movimiento Celular/fisiología , Corteza Cerebral/química , Corteza Cerebral/patología , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Ratones , Neuroglía/química , Neuroglía/patología , Embarazo
20.
Zhen Ci Yan Jiu ; 39(6): 466-71, 2014 Dec.
Artículo en Chino | MEDLINE | ID: mdl-25632571

RESUMEN

OBJECTIVE: To observe the effect of acupuncture therapy on 14-3-3, Bcl-2 and Bax expression levels in the cerebral cortex in neonatal rats with hypoxic-ischemic brain damage(HIBD). METHODS: Timed pregnant Sprague-Dawley rat dams were delivered either vaginally (normal group), or by C-section (sham-operation group) or by C-section with 5 min of global anoxia (anoxia group), with 8 rats in each group. The rat pups of the anoxia group were randomly divided into model group and acupuncture group (n =8). Acupuncture stimulation of "Naosanzhen" "Niesanzhen" and "Zhisanzhen" acupoints was given begin- ning from the 14th day after birth, once daily for 7 consecutive days. All rat pups were killed by decapitation on day 21 after birth, and then 14-3-3, Bcl-2 and Bax immunoactivity (expression) in the cerebral cortex were detected by immunohistochemistry. RESULTS: In comparison with the normal group, the expression level of cerebral cortical 14-3-3 was significantly decreased, and that of Bax remarkably increased in the model group (P<0. 01, P<0. 05). Compared to the model group, cortical 14-3-3 and Bcl-2 expression levels were markedly up-regulated in the acupuncture group (P<0.01, P<0.05). Compared to the normal group, cortical 14-3-3 expression level was obviously lower, but Bax expression level significantly higher in the sham-operation group (P<0. 05, P<0. 01). No significant differences were found between the model and normal groups in the expression levels of Bcl-2, and between the acupuncture and model groups in the expression levels of Bax (P>0. 05). CONCLUSION: Acupuncture intervention can increase the expression of 14-3-3 and Bcl-2 in the cerebral cortex in HIBD rats.


Asunto(s)
Proteínas 14-3-3/análisis , Terapia por Acupuntura , Corteza Cerebral/química , Hipoxia-Isquemia Encefálica/terapia , Animales , Femenino , Hipoxia-Isquemia Encefálica/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/análisis , Ratas , Ratas Sprague-Dawley , Proteína X Asociada a bcl-2/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA