Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 144
Filtrar
Más filtros

Base de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Neurobiol ; 58(11): 5602-5617, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34368932

RESUMEN

Subarachnoid hemorrhage (SAH) is a catastrophic form of stroke responsible for significant morbidity and mortality. Oxidative stress, inflammation, and neuronal apoptosis are important in the pathogenesis of early brain injury (EBI) following SAH. Preconditioning exercise confers neuroprotective effects, mitigating EBI; however, the basis for such protection is unknown. We investigated the effects of preconditioning exercise on brain damage and sensorimotor function after SAH. Male rats were assigned to either a sham-operated (Sham) group, exercise (Ex) group, or no-exercise (No-Ex) group. After a 3-week exercise program, they underwent SAH by endovascular perforation. Consciousness level, neurological score, and sensorimotor function were studied. The expression of nuclear factor erythroid 2 p45-related factor 2 (Nrf2), heme oxygenase 1 (HO-1), 4-hydroxynonenal (4HNE), nitrotyrosine (NT), ionized calcium-binding adaptor molecule 1 (Iba1), tumor necrosis factor alpha (TNF-α), interleukin 6 (IL-6), interleukin 1ß (IL-1ß), 14-3-3γ, p-ß-catenin Ser37, Bax, and caspase-3 were evaluated by immunohistochemistry or western blotting. The terminal deoxynucleotidyl transferase-mediated biotinylated dUTP nick end labeling (TUNEL) assay was also performed. After SAH, the Ex group had significantly reduced neurological deficits, sensorimotor dysfunction, and consciousness disorder compared with the No-Ex group. Nrf2, HO-1, and 14-3-3γ were significantly higher in the Ex group, while 4HNE, NT, Iba1, TNF-α, IL-6, IL-1ß, Bax, caspase-3, and TUNEL-positive cells were significantly lower. Our findings suggest that preconditioning exercise ameliorates EBI after SAH. The expression of 4HNE and NT was reduced by Nrf2/HO-1 pathway activation; additionally, both oxidative stress and inflammation were reduced. Furthermore, preconditioning exercise reduced apoptosis, likely via the 14-3-3γ/p-ß-catenin Ser37/Bax/caspase-3 pathway.


Asunto(s)
Daño Encefálico Crónico/prevención & control , Neuronas/patología , Condicionamiento Físico Animal , Hemorragia Subaracnoidea/complicaciones , Proteínas 14-3-3/fisiología , Animales , Apoptosis , Daño Encefálico Crónico/diagnóstico por imagen , Daño Encefálico Crónico/etiología , Daño Encefálico Crónico/metabolismo , Citocinas/biosíntesis , Citocinas/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Procesamiento de Imagen Asistido por Computador , Etiquetado Corte-Fin in Situ , Masculino , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Enfermedades Neuroinflamatorias/etiología , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/prevención & control , Estrés Oxidativo , Condicionamiento Físico Animal/fisiología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Factores de Tiempo , Microtomografía por Rayos X
2.
Neuromolecular Med ; 23(1): 211-223, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32914259

RESUMEN

Sphingosine 1-phosphate (S1P) is an important lipid biomolecule that exerts pleiotropic cellular actions as it binds to and activates its five G-protein-coupled receptors, S1P1-5. Through these receptors, S1P can mediate diverse biological activities in both healthy and diseased conditions. S1P is produced by S1P-producing enzymes, sphingosine kinases (SphK1 and SphK2), and is abundantly present in different organs, including the brain. The medically important roles of receptor-mediated S1P signaling are well characterized in multiple sclerosis because FTY720 (Gilenya™, Novartis), a non-selective S1P receptor modulator, is currently used as a treatment for this disease. In cerebral ischemia, its role is also notable because of FTY720's efficacy in both rodent models and human patients with cerebral ischemia. In particular, some of the S1P receptors, including S1P1, S1P2, and S1P3, have been identified as pathogenic players in cerebral ischemia. Other than these receptors, S1P itself and S1P-producing enzymes have been shown to play certain roles in cerebral ischemia. This review aims to compile the current updates and overviews about the roles of S1P signaling, along with a focus on S1P receptors in cerebral ischemia, based on recent studies that used in vivo rodent models of cerebral ischemia.


Asunto(s)
Isquemia Encefálica/metabolismo , Lisofosfolípidos/fisiología , Proteínas del Tejido Nervioso/fisiología , Receptores de Esfingosina-1-Fosfato/fisiología , Esfingosina/análogos & derivados , Animales , Daño Encefálico Crónico/etiología , Daño Encefálico Crónico/metabolismo , Isquemia Encefálica/complicaciones , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Clorhidrato de Fingolimod/uso terapéutico , Humanos , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/metabolismo , Inflamación , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Neovascularización Fisiológica/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Ratas , Transducción de Señal/fisiología , Esfingosina/fisiología
3.
Medicina (Kaunas) ; 56(3)2020 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-32204311

RESUMEN

Traumatic brain injury represents physical damage to the brain tissue that induces transitory or permanent neurological disabilities. The traumatic injury activates an important inflammatory response, followed by a cascade of events that lead to neuronal loss and further brain damage. Maintaining proper ventilation, a normal level of oxygenation, and adequate blood pressure are the main therapeutic strategies performed after injury. Surgery is often necessary for patients with more serious injuries. However, to date, there are no therapies that completely resolve the brain damage suffered following the trauma. Stem cells, due to their capacity to differentiate into neuronal cells and through releasing neurotrophic factors, seem to be a valid strategy to use in the treatment of traumatic brain injury. The purpose of this review is to provide an overview of clinical trials, aimed to evaluate the use of stem cell-based therapy in traumatic brain injury. These studies aim to assess the safety and efficacy of stem cells in this disease. The results available so far are few; therefore, future studies need in order to evaluate the safety and efficacy of stem cell transplantation in traumatic brain injury.


Asunto(s)
Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/terapia , Inflamación/etiología , Trasplante de Células Madre/métodos , Adolescente , Adulto , Anciano , Daño Encefálico Crónico/metabolismo , Daño Encefálico Crónico/fisiopatología , Daño Encefálico Crónico/terapia , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/fisiopatología , Niño , Preescolar , Ensayos Clínicos como Asunto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Factores de Crecimiento Nervioso/metabolismo , Neuronas/patología , Seguridad , Resultado del Tratamiento , Adulto Joven
4.
Pharmacol Res ; 144: 331-335, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31042564

RESUMEN

Dickkopf-1 (DKK1), a secretory glycoprotein discovered for 'inducing generation of head', is an endogenous inhibitor of the canonical Wnt/ß-catenin signaling pathway. It was found to be involved in many pathophysiological processes in vivo. Abnormal expression of DKK1 will alter expressions of related proteins and genes not only in canonical Wnt/ß-catenin signaling pathway but also in other signaling pathways. Previous studies of DKK1 focused on its function in tumors. In recent years, a large number of studies have shown that it plays an important role in embryonic development, neural regeneration, synaptogenesis and so on. Therefore, its role in neuropsychiatric disorders, such as neurodysplasia, cognitive impairment and emotional disorder, has attracted increasing attention. At present, the role of DKK1 in Alzheimer's disease (AD) is one of the research hot topics. This article reviewed the research progress of its role in AD in order to provide new ideas and directions for further studies on the pathogenesis and treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Daño Encefálico Crónico/etiología , Encéfalo/patología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Encéfalo/metabolismo , Daño Encefálico Crónico/metabolismo , Daño Encefálico Crónico/patología , Humanos , Péptidos y Proteínas de Señalización Intercelular/análisis , Vía de Señalización Wnt
5.
Am J Forensic Med Pathol ; 39(1): 8-13, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29293100

RESUMEN

The polysialylated isoform of the neural cell adhesion molecule (PSA-NCAM) has been shown to be a key player in neuroplastic changes and is expressed in various disorders. We investigated the PSA-NCAM expression on brain cortical tissue in a cohort of drug-related deaths. Brains from 25 drug abusers and 10 control subjects were removed at autopsy, and 2 samples of the right parietal lobe of each case were obtained. The polysialylated isoform of NCAM was evaluated on formalin-fixed and paraffin-embedded tissues. Eleven patients were polydrug abusers; 14 used a single substance. The mechanisms of death were acute respiratory failure (n = 19), cardiorespiratory failure (n = 4), acute heart failure (n = 1), and brain injury (n = 1). Toxicological analyses of blood were available for all cases, and urine and bile analyses for 19 of 25 cases. The polysialylated isoform of NCAM immunoexpression in the neuronal soma and dendritic spines was observed in 18 (72%) of 25 drug abusers and in 2 (20%) of 10 control subjects. Drug abusers were statistically more positive for PSA-NCAM than control subjects (P = 0.0082). The expression of PSA-NCAM in the parietal cortex could be an indicator of brain damage due to drug abuse, and its availability could allow the forensic pathologists to develop rapid and low-cost additional or alternative method to improve detection of drug-related deaths.


Asunto(s)
Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Lóbulo Parietal/metabolismo , Ácidos Siálicos/metabolismo , Trastornos Relacionados con Sustancias/diagnóstico , Adulto , Biomarcadores/metabolismo , Daño Encefálico Crónico/diagnóstico , Daño Encefálico Crónico/metabolismo , Estudios de Casos y Controles , Femenino , Patologia Forense , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Isoformas de Proteínas/metabolismo , Trastornos Relacionados con Sustancias/metabolismo , Adulto Joven
6.
Neuromolecular Med ; 19(4): 541-554, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28916896

RESUMEN

The pro-inflammatory activity of interleukin 17, which is produced by the IL-23/IL-17 axis, has been associated with the pathogenesis of traumatic brain injury (TBI). The study investigated the potential role of IL-17 in secondary brain injury of TBI in a rat model. Our data showed that the levels of IL-17 increased from 6 h to 7 days and peaked at 3 days, in both the CNS and serum, which were consistent with the severity of secondary brain injury. The IL-23 inhibitor suberoylanilide hydroxamic acid (SAHA) treatment markedly decreased the expressions of IL-17 and apoptosis-associated proteins cleaved caspase-3 and increased the protein ratio of Bcl-2 (B cell lymphoma/leukemia-2)/Bax (Bcl-2-associated X protein). Meanwhile, neuronal apoptosis was reduced, and neural function was improved after SAHA treatment. This study suggests that IL-17 is involved in secondary brain injury after TBI. Administering an IL-23 inhibitor and thereby blocking the IL-23/IL-17 axis may be beneficial in the treatment of TBI.


Asunto(s)
Daño Encefálico Crónico/fisiopatología , Lesiones Traumáticas del Encéfalo/fisiopatología , Interleucina-17/fisiología , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas Reguladoras de la Apoptosis/genética , Daño Encefálico Crónico/etiología , Daño Encefálico Crónico/metabolismo , Daño Encefálico Crónico/prevención & control , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Ácidos Hidroxámicos/farmacología , Ácidos Hidroxámicos/uso terapéutico , Inflamación , Interleucina-17/sangre , Interleucina-17/líquido cefalorraquídeo , Interleucina-17/genética , Interleucina-23/antagonistas & inhibidores , Interleucina-23/fisiología , Masculino , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/sangre , Proteínas del Tejido Nervioso/líquido cefalorraquídeo , Proteínas del Tejido Nervioso/genética , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Vorinostat
7.
J Nutr ; 144(7): 1058-66, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24744313

RESUMEN

Gestational iron deficiency (ID) has been associated with a wide variety of central nervous system (CNS) impairments in developing offspring. However, a focus on singular regions has impeded an understanding of the CNS-wide effects of this micronutrient deficiency. Because the developing brain requires iron during specific phases of growth in a region-specific manner, we hypothesized that maternal iron deprivation would lead to region-specific impairments in the CNS of offspring. Female rats were fed an iron control (Fe+) or iron-deficient (Fe-) diet containing 240 or 6 µg/g iron during gestation and lactation. The corpus callosum (CC), hippocampus, and cortex of the offspring were analyzed at postnatal day 21 (P21) and/or P40 using structural and functional measures. In the CC at P40, ID was associated with reduced peak amplitudes of compound action potentials specific to myelinated axons, in which diameters were reduced by ∼20% compared with Fe+ controls. In the hippocampus, ID was associated with a 25% reduction in basal dendritic length of pyramidal neurons at P21, whereas branching complexity was unaffected. We also identified a shift toward increased proximal branching of apical dendrites in ID without an effect on overall length compared with Fe+ controls. ID also affected cortical neurons, but unlike the hippocampus, both apical and basal dendrites displayed a uniform decrease in branching complexity, with no significant effect on overall length. These deficits culminated in significantly poorer performance of P40 Fe- offspring in the novel object recognition task. Collectively, these results demonstrate that non-anemic gestational ID has a significant and region-specific impact on neuronal development and may provide a framework for understanding and recognizing the presentation of clinical symptoms of ID.


Asunto(s)
Daño Encefálico Crónico/etiología , Corteza Cerebral/diagnóstico por imagen , Cuerpo Calloso/diagnóstico por imagen , Deficiencias de Hierro , Lactancia , Fenómenos Fisiologicos Nutricionales Maternos , Neuronas/diagnóstico por imagen , Animales , Axones/metabolismo , Axones/ultraestructura , Daño Encefálico Crónico/congénito , Daño Encefálico Crónico/metabolismo , Daño Encefálico Crónico/patología , Corteza Cerebral/metabolismo , Cuerpo Calloso/metabolismo , Dendritas/metabolismo , Dendritas/ultraestructura , Femenino , Hipocampo/diagnóstico por imagen , Hipocampo/metabolismo , Masculino , Fibras Nerviosas Mielínicas/diagnóstico por imagen , Fibras Nerviosas Mielínicas/metabolismo , Neurogénesis , Neuronas/metabolismo , Embarazo , Células Piramidales/diagnóstico por imagen , Células Piramidales/metabolismo , Distribución Aleatoria , Ratas , Ratas Endogámicas F344 , Reproducibilidad de los Resultados , Ultrasonografía
8.
Biol Trace Elem Res ; 158(2): 176-85, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24573406

RESUMEN

Selenium (Se) is an important dietary micronutrient with antioxidative roles. Cadmium (Cd), a ubiquitous environmental pollutant, is known to cause brain lesion in rats and humans. However, little is reported about the deleterious effects of subchronic Cd exposure on the brain of poultry and the protective roles on the brain by Se against Cd. The aim of this study was to investigate the protective effects of Se on Cd-induced brain damage in chickens. One hundred twenty 100-day-old chickens were randomly assigned to four groups and were fed a basal diet, or Se (as 10 mg Na2SeO3/kg dry weight of feed), Cd (as 150 mg CdCl2/kg dry weight of feed), or Cd + Se in their basic diets for 60 days. Then, concentrations of Cd and Se, production of nitric oxide (NO), messenger RNA (mRNA) level and activity of inducible NO synthase (iNOS), level of oxidative stress, and histological and ultrastructural changes of the cerebrum and cerebellum were examined. The results showed that Cd exposure significantly increased Cd accumulation, NO production, iNOS activities, iNOS mRNA level, and MDA content in the cerebrum and cerebellum. Cd treatment obviously decreased Se content and antioxidase activities and caused histopathological changes in the cerebrum and cerebellum. Se supplementation during dietary Cd obviously reduced Cd accumulation, NO production, mRNA level and activity of iNOS, oxidative stress, and histopathological damage in the cerebrum and cerebellum of chickens. It indicated that Se ameliorates Cd-induced brain damage in chickens by regulating iNOS-NO system changes, and oxidative stress induced by Cd and Se can serve as a potential therapeutic for Cd-induced brain lesion of chickens.


Asunto(s)
Daño Encefálico Crónico/inducido químicamente , Daño Encefálico Crónico/prevención & control , Cadmio/toxicidad , Selenio/farmacología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Encéfalo/metabolismo , Daño Encefálico Crónico/enzimología , Daño Encefálico Crónico/metabolismo , Cadmio/administración & dosificación , Pollos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Estrés Oxidativo/efectos de los fármacos , Selenio/administración & dosificación
9.
Resuscitation ; 85(2): 299-305, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24200890

RESUMEN

OBJECTIVE: The aim of this study was to investigate whether the DOR agonist BW373U86 conferred neuroprotection following ACA when given after resuscitation and to determine the long-term effects of chronic BW373U86 treatment on ACA-elicited brain injury. METHODS: Animals were divided into acute and chronic treatment groups. Each group consisted of four sub-groups, including Sham, ACA, BW373U86 (BW373U86+ACA), and Naltrindole groups (Naltrindole and BW373U86+ACA). The DOR antagonist Naltrindole was used to confirm the possible receptor-dependent effects of BW373U86. ACA was induced by 8min of asphyxiation followed by resuscitation. All drugs were administered either immediately after the restoration of spontaneous circulation (ROSC) in acute-treatment groups or over 6 consecutive days in chronic-treatment groups. Alterations of cAMP response element-binding protein (CREB) and phosphorylated CREB (pCREB) were analyzed by western blot and immunohistochemistry. Neurological functions were assessed by neurological deficit score (NDS) and Morris Water Maze performance. Neurodegeneration was monitored by immunofluorescence and Nissl staining. RESULTS: ACA induced massive neuron loss and serious neurological function deficits. BW373U86 significantly reduced both of these negative effects and increased CREB and pCREB expression in the hippocampus; these effects were reversed with acute Naltrindole treatment. The protective effects of BW373U86 persisted until 28d post-ROSC with chronic treatment, but these effects were not reversed by Naltrindole. CONCLUSIONS: BW373U86 attenuates global cerebral ischemic injury induced by ACA through both DOR-dependent and DOR-independent mechanisms. CREB might be an important molecule in mediating these neuroprotective effects.


Asunto(s)
Asfixia/terapia , Benzamidas/farmacología , Daño Encefálico Crónico/prevención & control , Reanimación Cardiopulmonar , Paro Cardíaco/terapia , Fármacos Neuroprotectores/farmacología , Piperazinas/farmacología , Animales , Western Blotting , Daño Encefálico Crónico/metabolismo , Daño Encefálico Crónico/patología , Proteína de Unión a CREB/metabolismo , Modelos Animales de Enfermedad , Técnicas para Inmunoenzimas , Masculino , Naltrexona/análogos & derivados , Naltrexona/farmacología , Ratas Sprague-Dawley
10.
Brain Res ; 1455: 114-23, 2012 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-22483792

RESUMEN

Excess activation of ionotropic glutamate receptors and iron is believed to contribute to free radical production and neuronal death following hypoxic ischemia. We examined the possibility that both NMDA receptor activation and iron overload determine spatial and temporal patterns of free radical production after transient middle cerebral artery occlusion (tMCAO) in male Sprague-Dawley rats. Mitochondrial free radical (MFR) levels were maximally increased in neurons in the core at 1 h and 24 h after tMCAO. Early MFR production was blocked by administration of MK-801, an NMDA receptor antagonist, but not deferoxamine, an iron chelator. Neither MK-801 nor deferoxamine attenuated late MFR production in the core. Increased MFRs were observed in penumbral neurons within 6 h and gradually increased over 24 h after tMCAO. Slowly-evolving MFRs in the core and penumbra were accompanied by iron overload. Deferoxamine blocked iron overload but reduced MFR production only in the penumbra. Combined MK-801/deferoxamine reduced late MFR production in both core and penumbra in an additive manner. Combination therapy significantly ameliorated infarction compared with monotherapy. These findings suggest that the NMDA receptor activation and iron overload mediate late MFR production and infarction after tMCAO.


Asunto(s)
Daño Encefálico Crónico/metabolismo , Radicales Libres/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Hierro/metabolismo , Estrés Oxidativo/fisiología , Receptores de N-Metil-D-Aspartato/fisiología , Animales , Daño Encefálico Crónico/tratamiento farmacológico , Daño Encefálico Crónico/fisiopatología , Deferoxamina/farmacología , Modelos Animales de Enfermedad , Maleato de Dizocilpina/farmacología , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/fisiopatología , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores
11.
Eur J Pharmacol ; 680(1-3): 41-8, 2012 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-22314218

RESUMEN

Chronic cerebral hypoperfusion, induced by permanent occlusion of bilateral common carotid arteries (2VO), is related to neurological disorders and contributes to cognitive decline. Chrysin (5,7-dihydroxyflavone) is an important member of the flavonoid family. The aim of this study is to investigate the effects of chrysin on cognitive deficits and brain damage in this rat 2VO model. At 52days after ligation, the escape latency in Morris water maze was significantly increased in rats subjected to 2VO, the neuronal damage was also increased accompanied by a large proliferation in glial fibrillary acidic protein (GFAP) immunoreactivity with marked white matter lesions, and neuronal cell apoptosis, all of which were significantly alleviated by long treatment of chrysin (30mg/kg). Biochemical examinations revealed that chrysin decreased lipid peroxide, reduced the increased activities of superoxide dismutase, and attenuated the decreased activities of glutathione peroxidase in 2VO rats. The results suggest that chrysin may have therapeutic potential for the treatment of neurodegeneration and dementia caused by decreased cerebral blood flow, which is most likely related, at least in part, to its anti-inflammatory and antioxidant properties.


Asunto(s)
Daño Encefálico Crónico/tratamiento farmacológico , Isquemia Encefálica/complicaciones , Trastornos del Conocimiento/tratamiento farmacológico , Flavonoides/farmacología , Fármacos Neuroprotectores/farmacología , Animales , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Daño Encefálico Crónico/metabolismo , Daño Encefálico Crónico/patología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Arteria Carótida Común/efectos de los fármacos , Arteria Carótida Común/metabolismo , Arteria Carótida Común/patología , Circulación Cerebrovascular/efectos de los fármacos , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/patología , Demencia/tratamiento farmacológico , Demencia/metabolismo , Demencia/patología , Proteína Ácida Fibrilar de la Glía/metabolismo , Glutatión Peroxidasa/metabolismo , Discapacidades para el Aprendizaje/tratamiento farmacológico , Discapacidades para el Aprendizaje/metabolismo , Discapacidades para el Aprendizaje/patología , Peróxidos Lipídicos/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Ratas , Ratas Wistar , Superóxido Dismutasa/metabolismo
12.
Brain Pathol ; 22(2): 142-9, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21714827

RESUMEN

While a history of a single traumatic brain injury (TBI) is associated with the later development of syndromes of cognitive impairment such as Alzheimer's disease, the long-term pathology evolving after single TBI is poorly understood. However, a progressive tauopathy, chronic traumatic encephalopathy, is described in selected cohorts with a history of repetitive concussive/mild head injury. Here, post-mortem brains from long-term survivors of just a single TBI (1-47 years survival; n=39) vs. uninjured, age-matched controls (n=47) were examined for neurofibrillary tangles (NFTs) and amyloid-ß (Aß) plaques using immunohistochemistry and thioflavine-S staining. Detailed maps of findings permitted classification of pathology using semiquantitative scoring systems. NFTs were exceptionally rare in young, uninjured controls, yet were abundant and widely distributed in approximately one-third of TBI cases. In addition, Aß-plaques were found in a greater density following TBI vs. controls. Moreover, thioflavine-S staining revealed that while all plaque-positive control cases displayed predominantly diffuse plaques, 64% of plaque-positive TBI cases displayed predominantly thioflavine-S-positive plaques or a mixed thioflavine-S-positive/diffuse pattern. These data demonstrate that widespread NFT and Aß plaque pathologies are present in up to a third of patients following survival of a year or more from a single TBI. This suggests that a single TBI induces long-term neuropathological changes akin to those found in neurodegenerative disease.


Asunto(s)
Péptidos beta-Amiloides/efectos adversos , Daño Encefálico Crónico/patología , Lesiones Encefálicas/patología , Ovillos Neurofibrilares/patología , Placa Amiloide/patología , Proteínas tau/efectos adversos , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Daño Encefálico Crónico/metabolismo , Daño Encefálico Crónico/fisiopatología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/fisiopatología , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
13.
Glia ; 59(12): 1958-73, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21956849

RESUMEN

NG2-expressing cells comprise a population of cycling precursors that can exit the cell cycle and differentiate into mature oligodendrocytes. As a whole, they display heterogeneous properties and behaviors that remain unresolved at the molecular level, although partly interpretable as distinct maturation stages. To address this issue, we analyzed the expression of the GPR17 receptor, recently shown to decorate NG2-expressing cells and to operate as an early sensor of brain damage, in immature and adult oligodendrocyte progenitors in the intact brain and after injury. In both the early postnatal and adult cerebral cortex, distinct GPR17 protein localizations and expression levels define different stages of oligodendroglial maturation, ranging from the precursor phase to the premyelinating phenotype. As soon as cells exit mitosis, a fraction of NG2-expressing cells displays accumulation of GPR17 protein in the Golgi apparatus. GPR17 expression is subsequently upregulated and distributed to processes of cells that stop dividing, progressively lose NG2 positivity and assume premyelinating features. Absence of colabeling with mature markers or myelin proteins indicates that GPR17 is downregulated when cells complete their final maturation. BrdU-based fate-mapping demonstrated that a significant fraction of newly generated oligodendrocyte progenitors transiently upregulates GPR17 during maturation. Importantly, we also found that GPR17 does not participate to the early reaction of NG2-expressing cells to damage, while it is induced at postacute stages after injury. These findings identify GPR17 as a marker for progenitor progression within the oligodendroglial lineage and highlight its participation to postacute reactivity of NG2 cells in different injury paradigms.


Asunto(s)
Antígenos/biosíntesis , Daño Encefálico Crónico/metabolismo , Lesiones Encefálicas/metabolismo , Diferenciación Celular/fisiología , Proteínas del Tejido Nervioso/metabolismo , Oligodendroglía/metabolismo , Proteoglicanos/biosíntesis , Receptores Acoplados a Proteínas G/metabolismo , Células Madre/metabolismo , Enfermedad Aguda , Animales , Antígenos/genética , Biomarcadores/metabolismo , Daño Encefálico Crónico/patología , Lesiones Encefálicas/patología , Modelos Animales de Enfermedad , Aparato de Golgi/metabolismo , Aparato de Golgi/patología , Ratones , Ratones Endogámicos C57BL , Regeneración Nerviosa/fisiología , Oligodendroglía/patología , Cultivo Primario de Células , Proteoglicanos/genética , Células Madre/patología
14.
Hypertension ; 58(4): 635-42, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21859961

RESUMEN

The role of the renin-angiotensin system in cognitive impairment is unclear. This work was undertaken to test our hypothesis that renin-angiotensin system may contribute to cognitive decline and brain damage caused by chronic cerebral ischemia. C57BL/6J mice were subjected to bilateral common carotid artery stenosis with microcoil to prepare mice with chronic cerebral hypoperfusion, a model of subcortical vascular dementia. The effects of aliskiren, a direct renin inhibitor, or Tempol, a superoxide scavenger, on brain damage and working memory in these mice were examined. Chronic cerebral hypoperfusion significantly increased brain renin activity and angiotensinogen expression in C57BL/6J mice, which was attributed to the increased renin in activated astrocytes and microvessels and the increased angiotensinogen in activated astrocytes in white matter. Aliskiren pretreatment significantly inhibited brain renin activity and ameliorated brain p67(phox)-related NADPH oxidase activity, oxidative stress, glial activation, white matter lesion, and spatial working memory deficits in C57BL/6J mice with bilateral common carotid artery stenosis. To elucidate the role of oxidative stress in brain protective effects of aliskiren, we also examined the effect of Tempol in the same mice with bilateral common carotid artery stenosis. Tempol pretreatment mimicked the brain protective effects of aliskiren in this mouse model. Posttreatment of mice with aliskiren or Tempol after bilateral common carotid artery stenosis also prevented cognitive decline. In conclusion, chronic cerebral hypoperfusion induced the activation of the brain renin-angiotensin system. Aliskiren ameliorated brain damage and working memory deficits in the model of chronic cerebral ischemia through the attenuation of oxidative stress. Thus, direct renin inhibition seems to be a promising therapeutic strategy for subcortical vascular dementia.


Asunto(s)
Daño Encefálico Crónico/prevención & control , Isquemia Encefálica/metabolismo , Trastornos del Conocimiento/prevención & control , Demencia Vascular/metabolismo , Renina/antagonistas & inhibidores , Renina/metabolismo , Amidas/farmacología , Angiotensinógeno/metabolismo , Animales , Antihipertensivos/farmacología , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Daño Encefálico Crónico/epidemiología , Daño Encefálico Crónico/metabolismo , Trastornos del Conocimiento/epidemiología , Trastornos del Conocimiento/metabolismo , Óxidos N-Cíclicos/farmacología , Modelos Animales de Enfermedad , Fumaratos/farmacología , Hipertensión/complicaciones , Masculino , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Sistema Renina-Angiotensina/efectos de los fármacos , Sistema Renina-Angiotensina/fisiología , Factores de Riesgo , Marcadores de Spin
15.
J Neurotrauma ; 28(6): 983-93, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21501066

RESUMEN

Inflammatory and ischemic processes contribute to the development of secondary brain damage after mechanical brain injury. Recent data suggest that thiazolidinediones (TZDs), a class of drugs approved for the treatment of non-insulin-dependent diabetes mellitus, effectively reduces inflammation and brain lesion by stimulation of the peroxisome proliferator-activated receptor-γ (PPAR-γ). The present study investigates the influence of the TZD pioglitazone and rosiglitazone on inflammation and secondary brain damage after experimental traumatic brain injury (TBI). A controlled cortical impact (CCI) injury was induced in male C57BL/6 mice to investigate following endpoints: (1) mRNA expression of PPAR-γ and PPAR-γ target genes (LPL, GLT1, and IRAP/Lnpep), and inflammatory markers (TNF-α, IL-1ß, IL-6, and iNOS), at 15 min, 3 h, 6 h, 12 h, and 24 h post-trauma; (2) contusion volume, neurological function, and gene expression after 24 h in mice treated with pioglitazone (0.5 and 1 mg/kg) or rosiglitazone (5 and 10 mg/kg IP at 30 min post-trauma); and (3) the role of PPAR-γ to mediate protection was determined in animals treated with pioglitazone, the PPAR-γ inhibitor T0070907, and a combination of both. Inflammatory marker genes, but not PPAR-γ gene expression, was upregulated after trauma. Pioglitazone reduced the histological damage and inflammation in a dose-dependent fashion. In contrast, rosiglitazone failed to suppress inflammation and histological damage. PPAR-γ and PPAR-γ target gene expression was not induced by pioglitazone and rosiglitazone. In line with these results, pioglitazone-mediated protection was not reversed by T0070907. The results indicate that the neuroprotective effects of pioglitazone are not solely related to PPAR-γ-dependent mechanisms.


Asunto(s)
Daño Encefálico Crónico/tratamiento farmacológico , Lesiones Encefálicas/tratamiento farmacológico , Hipoglucemiantes/farmacología , Fármacos Neuroprotectores/farmacología , PPAR gamma/fisiología , Tiazolidinedionas/farmacología , Animales , Daño Encefálico Crónico/metabolismo , Lesiones Encefálicas/metabolismo , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Pioglitazona
16.
Brain Res ; 1382: 259-65, 2011 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-21241675

RESUMEN

Pristanic acid (Prist) is accumulated in various peroxisomal disorders characterized by severe neurological dysfunction whose pathogenesis is poorly understood. Since oxidative damage has been demonstrated in brain of patients affected by neurodegenerative disorders, in the present work we investigated the in vitro effects of Prist on important parameters of oxidative stress in cerebral cortex from young rats. Prist significantly increased malondialdehyde levels, reflecting an increase of lipid peroxidation. This effect was totally prevented by the free radical scavenger melatonin, suggesting the involvement of reactive species. Prist also provoked protein oxidative damage, as determined by increased carbonyl formation and sulfhydryl oxidation. Otherwise, it did not alter nitric oxide production, indicating that nitrogen reactive species were not implicated in the lipid and oxidative damage provoked by Prist. Furthermore, the concentration of glutathione (GSH), the major brain non-enzymatic antioxidant defense, was significantly decreased by Prist and this decrease was fully prevented by melatonin and attenuated by α-tocopherol. It is therefore presumed that Prist elicits oxidative stress in the brain probably via reactive oxygen species formation and that this pathomechanism may possibly be involved in the brain damage found in patients affected by peroxisomal disorders where Prist accumulates.


Asunto(s)
Química Encefálica/efectos de los fármacos , Daño Encefálico Crónico/inducido químicamente , Corteza Cerebral/efectos de los fármacos , Ácidos Grasos/toxicidad , Estrés Oxidativo/efectos de los fármacos , Trastorno Peroxisomal/inducido químicamente , Animales , Química Encefálica/fisiología , Daño Encefálico Crónico/metabolismo , Daño Encefálico Crónico/fisiopatología , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/fisiopatología , Modelos Animales de Enfermedad , Ácidos Grasos/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Peroxidación de Lípido/fisiología , Masculino , Estrés Oxidativo/fisiología , Trastorno Peroxisomal/metabolismo , Trastorno Peroxisomal/fisiopatología , Ratas , Ratas Wistar
17.
Brain Res Rev ; 67(1-2): 157-83, 2011 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-21238488

RESUMEN

Almost a quarter of a century ago, interleukin-6 (IL-6) was discovered as an inflammatory cytokine involved in B cell differentiation. Today, IL-6 is recognized to be a highly versatile cytokine, with pleiotropic actions not only in immune cells, but also in other cell types, such as cells of the central nervous system (CNS). The first evidence implicating IL-6 in brain-related processes originated from its dysregulated expression in several neurological disorders such as multiple sclerosis, Alzheimer's disease and Parkinson's disease. In addition, IL-6 was shown to be involved in multiple physiological CNS processes such as neuron homeostasis, astrogliogenesis and neuronal differentiation. The molecular mechanisms underlying IL-6 functions in the brain have only recently started to emerge. In this review, an overview of the latest discoveries concerning the actions of IL-6 in the nervous system is provided. The central position of IL-6 in the neuroinflammatory reaction pattern, and more specifically, the role of IL-6 in specific neurodegenerative processes, which accompany Alzheimer's disease, multiple sclerosis and excitotoxicity, are discussed. It is evident that IL-6 has a dichotomic action in the CNS, displaying neurotrophic properties on the one hand, and detrimental actions on the other. This is in agreement with its central role in neuroinflammation, which evolved as a beneficial process, aimed at maintaining tissue homeostasis, but which can become malignant when exaggerated. In this perspective, it is not surprising that 'well-meant' actions of IL-6 are often causing harm instead of leading to recovery.


Asunto(s)
Interleucina-6/fisiología , Trastornos Mentales/metabolismo , Trastornos Mentales/patología , Animales , Daño Encefálico Crónico/etiología , Daño Encefálico Crónico/metabolismo , Daño Encefálico Crónico/patología , Muerte Celular/fisiología , Humanos , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Interleucina-6/metabolismo , Interleucina-6/toxicidad , Trastornos Mentales/etiología , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología
18.
Minerva Pediatr ; 62(3 Suppl 1): 141-3, 2010 Jun.
Artículo en Italiano | MEDLINE | ID: mdl-21090084

RESUMEN

Hypoxia-ischemia (H-I) constitutes the main phenomenon responsible for brain-blood barrier permeability modifications leading to cerebral vascular autoregulation loss in newborns. Hypotension, cerebral ischemia, and reperfusion are the main events involved in vascular auto-regulation loss leading to cell death and tissue damage. Reperfusion could be critical since organ damage, particularly of the brain, may be amplified during this period. An exaggerated activation of vasoactive agents, of calcium mediated effects could be responsible for reperfusion injury (R-I), which, in turns, leads to cerebral hemorrhage and damage. These phenomena represent a common repertoire in newborns complicated by perinatal acute or chronic hypoxia treated by risky procedures such as mechanical ventilation, nitric oxide supplementation, brain cooling, and extracorporeal membrane oxygenation (ECMO). Despite accurate monitoring, the post-insult period is crucial, as clinical symptoms and standard monitoring parameters may be silent at a time when brain damage is already occurring and the therapeutic window for pharmacological intervention is limited. Therefore, the measurement of circulating biochemical markers of brain damage, such as vasoactive agents and nervous tissue peptides is eagerly awaited in clinical practice to detect high risk newborns. The present article is aimed at investigating the role of dosage biochemical markers in non-invasive biological fluids such as S100B, a calcium binding protein, activin A, a protein expressed in Central nervous System (CNS).


Asunto(s)
Activinas/orina , Daño Encefálico Crónico/prevención & control , Hipoxia-Isquemia Encefálica/metabolismo , Factores de Crecimiento Nervioso/análisis , Proteínas S100/análisis , Saliva/química , Biomarcadores/análisis , Daño Encefálico Crónico/etiología , Daño Encefálico Crónico/metabolismo , Estudios de Casos y Controles , Dimerización , Humanos , Hipoxia-Isquemia Encefálica/complicaciones , Hipoxia-Isquemia Encefálica/fisiopatología , Recién Nacido , Enfermedades del Recién Nacido/metabolismo , Factores de Crecimiento Nervioso/química , Factores de Crecimiento Nervioso/orina , Daño por Reperfusión/prevención & control , Subunidad beta de la Proteína de Unión al Calcio S100 , Proteínas S100/química , Proteínas S100/orina , Urinálisis
19.
Neuroscience ; 167(3): 716-23, 2010 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-20188153

RESUMEN

Glutamate excitotoxicity is among the main cellular mechanisms leading to perinatal insults in human newborns. We used intracerebral injection of the glutamatergic glutamate N-methyl-D-aspartate-receptor agonist ibotenate to produce excitotoxic lesions mimicking the acquired white matter lesions seen in human preterm infants. We evaluated whether nonsteroidal antiinflammatory drugs (NSAIDs) protected against glutamate excitotoxicity. Aspirin (0.01-100 microg/d), indomethacin (0.1-10 microg/d), paracetamol (10-100 microg/d), or NS-398 (12.5 microg/d) was given daily before ibotenate (P1 to P5) or after ibotenate (P5 to P9). Lesion size was measured on Cresyl Violet-stained brain sections collected on P10. None of the drugs tested alone or in combination increased lesion size. Pretreatment with low- or high-dose aspirin and post-treatment with paracetamol or NS-398 protected against white matter lesions, whereas cortical lesions were decreased by pretreatment with low- or high-dose aspirin or post-treatment with NS-398. The corticosteroid betamethasone (0.18 microg/d) was neuroprotective when given before or after ibotenate and this effect was reversed by concomitant aspirin therapy (10 microg/d). In conclusion, perinatal NSAID administration may have beneficial effects on brain injury if appropriately timed.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Daño Encefálico Crónico/tratamiento farmacológico , Encefalitis/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Neurotoxinas/antagonistas & inhibidores , Complicaciones del Embarazo/tratamiento farmacológico , Acetaminofén/farmacología , Acetaminofén/uso terapéutico , Corticoesteroides/antagonistas & inhibidores , Corticoesteroides/farmacología , Animales , Animales Recién Nacidos , Antiinflamatorios no Esteroideos/uso terapéutico , Aspirina/farmacología , Aspirina/uso terapéutico , Betametasona/antagonistas & inhibidores , Betametasona/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Daño Encefálico Crónico/metabolismo , Daño Encefálico Crónico/fisiopatología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Quimioterapia Combinada , Encefalitis/metabolismo , Encefalitis/fisiopatología , Femenino , Ácido Iboténico/antagonistas & inhibidores , Ácido Iboténico/toxicidad , Indometacina/farmacología , Indometacina/uso terapéutico , Ratones , Fibras Nerviosas Mielínicas/efectos de los fármacos , Fibras Nerviosas Mielínicas/patología , Fármacos Neuroprotectores/uso terapéutico , Neurotoxinas/toxicidad , Embarazo , Complicaciones del Embarazo/fisiopatología , Complicaciones del Embarazo/prevención & control , Resultado del Tratamiento
20.
Acta Neurochir (Wien) ; 151(11): 1419-25, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19499176

RESUMEN

PURPOSE: Radiation-induced brain injury (RBI) is an insidious side-effect of radiotherapy mediated by vascular alterations, inflammation and ischaemia. In previous studies we had shown potential increases in loco-regional blood flow and glucose metabolism in brain tumours by using electrical cervical spinal cord stimulation (SCS). In this preliminary report we demonstrate the effect of cervical SCS on RBI-tissue metabolism, as assessed using [(18)F]fluorodeoxyglucose-positron emission tomography (FDG-PET). METHODS: SCS devices were inserted in eight patients with diagnosis of potential RBI in previously irradiated areas. While the SCS device was deactivated, each patient underwent an initial FDG-PET study to evaluate the clinical status. A second FDG-PET study was performed later the same day while the SCS device was activated in order to evaluate the effect of cervical SCS on glucose metabolism. RESULTS: Basal glucose metabolism in RBI areas was 31% lower than peri-RBI areas (p = 0.009) and 32% lower than healthy contra-lateral areas (p = 0.020). There was a significant increase in glucose uptake during SCS in both the RBI (p = 0.005) and the peri-RBI (p = 0.004) areas, with measured increases of 38 and 42%, respectively. The estimated potential maximal residual activity of the first FDG dose's contribution to the activity on the second scan was

Asunto(s)
Daño Encefálico Crónico/metabolismo , Encéfalo/metabolismo , Glucosa/metabolismo , Traumatismos por Radiación/metabolismo , Radioterapia/efectos adversos , Médula Espinal/fisiología , Adulto , Vías Aferentes/fisiología , Anciano , Encéfalo/diagnóstico por imagen , Encéfalo/fisiopatología , Daño Encefálico Crónico/diagnóstico por imagen , Daño Encefálico Crónico/fisiopatología , Mapeo Encefálico/métodos , Neoplasias Encefálicas/radioterapia , Circulación Cerebrovascular/fisiología , Vértebras Cervicales , Estimulación Eléctrica/métodos , Metabolismo Energético/fisiología , Femenino , Fluorodesoxiglucosa F18 , Glucosa/análisis , Humanos , Masculino , Persona de Mediana Edad , Tomografía de Emisión de Positrones/métodos , Valor Predictivo de las Pruebas , Traumatismos por Radiación/diagnóstico por imagen , Traumatismos por Radiación/fisiopatología , Sensibilidad y Especificidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA