Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 361
Filtrar
Más filtros










Intervalo de año de publicación
1.
J Mol Neurosci ; 67(4): 613-621, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30734244

RESUMEN

Traumatic brain injury (TBI) is one of the most common causes of neurological damage in young people. It was previously reported that dietary restriction, by either intermittent fasting (IF) or daily caloric restriction (CR), could protect neurons against dysfunction and degeneration in animal models of stroke and Parkinson's disease. Recently, several studies have shown that the protein Sirtuin 1 (SIRT1) plays a significant role in the induced neuroprotection following dietary restriction. In the present study, we found a significant reduction of SIRT1 levels in the cortex and hippocampus in a mouse model of mild weight-drop closed head TBI. This reduction was prevented in mice maintained on IF (alternate day fasting) and CR initiated after the head trauma. Hippocampus-dependent learning and memory (measured using a novel object recognition test) was impaired 30 days post-injury in mice fed ad libitum, but not in mice in the IF and CR groups. These results suggest a clinical potential for IF and/or CR as an intervention to reduce brain damage and improve functional outcome in TBI patients.


Asunto(s)
Lesiones Traumáticas del Encéfalo/complicaciones , Restricción Calórica/métodos , Disfunción Cognitiva/dietoterapia , Animales , Corteza Cerebral/metabolismo , Disfunción Cognitiva/etiología , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Sirtuina 1/metabolismo
3.
Cell Death Dis ; 4: e790, 2013 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-24008734

RESUMEN

Multi-protein complexes called inflammasomes have recently been identified and shown to contribute to cell death in tissue injury. Intravenous immunoglobulin (IVIg) is an FDA-approved therapeutic modality used for various inflammatory diseases. The objective of this study is to investigate dynamic responses of the NLRP1 and NLRP3 inflammasomes in stroke and to determine whether the NLRP1 and NLRP3 inflammasomes can be targeted with IVIg for therapeutic intervention. Primary cortical neurons were subjected to glucose deprivation (GD), oxygen-glucose deprivation (OGD) or simulated ischemia-reperfusion (I/R). Ischemic stroke was induced in C57BL/6J mice by middle cerebral artery occlusion, followed by reperfusion. Neurological assessment was performed, brain tissue damage was quantified, and NLRP1 and NLRP3 inflammasome protein levels were evaluated. NLRP1 and NLRP3 inflammasome components were also analyzed in postmortem brain tissue samples from stroke patients. Ischemia-like conditions increased the levels of NLRP1 and NLRP3 inflammasome proteins, and IL-1ß and IL-18, in primary cortical neurons. Similarly, levels of NLRP1 and NLRP3 inflammasome proteins, IL-1ß and IL-18 were elevated in ipsilateral brain tissues of cerebral I/R mice and stroke patients. Caspase-1 inhibitor treatment protected cultured cortical neurons and brain cells in vivo in experimental stroke models. IVIg treatment protected neurons in experimental stroke models by a mechanism involving suppression of NLRP1 and NLRP3 inflammasome activity. Our findings provide evidence that the NLRP1 and NLRP3 inflammasomes have a major role in neuronal cell death and behavioral deficits in stroke. We also identified NLRP1 and NLRP3 inflammasome inhibition as a novel mechanism by which IVIg can protect brain cells against ischemic damage, suggesting a potential clinical benefit of therapeutic interventions that target inflammasome assembly and activity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Portadoras/metabolismo , Inmunoglobulinas Intravenosas/farmacología , Inflamasomas/metabolismo , Neuronas/metabolismo , Accidente Cerebrovascular/patología , Animales , Isquemia Encefálica/complicaciones , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Caspasa 1/metabolismo , Inhibidores de Caspasas/farmacología , Muerte Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/patología , Citoprotección/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR , Proteínas NLR , Neuronas/efectos de los fármacos , Neuronas/enzimología , Neuronas/patología , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/metabolismo , Resultado del Tratamiento
4.
Transl Psychiatry ; 3: e304, 2013 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-24022509

RESUMEN

Alzheimer's disease (AD) is the most common form of dementia in the elderly. Although there are no drugs that modify the disease process, exposure to an enriched environment (EE) can slow the disease progression. Here, we characterize the effects of AD and EE on the post-transcriptional regulators, microRNAs (miRNAs), which may contribute to the detrimental and beneficial effects of AD and EE, respectively, on synaptic plasticity-related proteins and AD pathology. We found for the first time miRNAs that were inversely regulated in AD and EE, and may affect synaptic proteins and modulators, molecular factors associated with AD pathology, and survival and neuroprotective factors. MiRNAs that were upregulated only in 3xTgAD mice model of AD compared with their control mice were localized to synapses, predicted to downregulate essential synaptic proteins and are highly associated with regulating apoptosis, AD-associated processes and axon guidance. Studying the progressive change in miRNAs modulation during aging of 3xTgAD mice, we identified miRNAs that were regulated in earlier stages of AD, suggesting them as potential AD biomarkers. Last, we characterized AD- and EE-related effects in the mouse hippocampus on tomosyn protein levels, an inhibitor of the synaptic transmission machinery. While EE reduced tomosyn levels, tomosyn levels were increased in old 3xTgAD mice, suggesting a role for tomosyn in the impairment of synaptic transmission in AD. Interestingly, we found that miR-325 regulates the expression levels of tomosyn as demonstrated by a luciferase reporter assay, and that miR-325 was downregulated in AD and upregulated following EE. These findings improve our understanding of the molecular and cellular processes in AD pathology, following EE, and the interplay between the two processes, and open new avenues for the studies of understanding and controlling AD.


Asunto(s)
Envejecimiento/genética , Enfermedad de Alzheimer/genética , Ambiente , Regulación de la Expresión Génica/fisiología , Hipocampo/metabolismo , MicroARNs/genética , Plasticidad Neuronal/genética , Envejecimiento/metabolismo , Animales , Apoptosis/genética , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Ratones , MicroARNs/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas R-SNARE/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sinapsis/genética , Sinapsis/metabolismo , Transmisión Sináptica/genética
5.
Neuroscience ; 239: 228-40, 2013 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-23079624

RESUMEN

During development of the nervous system, the formation of connections (synapses) between neurons is dependent upon electrical activity in those neurons, and neurotrophic factors produced by target cells play a pivotal role in such activity-dependent sculpting of the neural networks. A similar interplay between neurotransmitter and neurotrophic factor signaling pathways mediates adaptive responses of neural networks to environmental demands in adult mammals, with the excitatory neurotransmitter glutamate and brain-derived neurotrophic factor (BDNF) being particularly prominent regulators of synaptic plasticity throughout the central nervous system. Optimal brain health throughout the lifespan is promoted by intermittent challenges such as exercise, cognitive stimulation and dietary energy restriction, that subject neurons to activity-related metabolic stress. At the molecular level, such challenges to neurons result in the production of proteins involved in neurogenesis, learning and memory and neuronal survival; examples include proteins that regulate mitochondrial biogenesis, protein quality control, and resistance of cells to oxidative, metabolic and proteotoxic stress. BDNF signaling mediates up-regulation of several such proteins including the protein chaperone GRP-78, antioxidant enzymes, the cell survival protein Bcl-2, and the DNA repair enzyme APE1. Insufficient exposure to such challenges, genetic factors may conspire to impair BDNF production and/or signaling resulting in the vulnerability of the brain to injury and neurodegenerative disorders including Alzheimer's, Parkinson's and Huntington's diseases. Further, BDNF signaling is negatively regulated by glucocorticoids. Glucocorticoids impair synaptic plasticity in the brain by negatively regulating spine density, neurogenesis and long-term potentiation, effects that are potentially linked to glucocorticoid regulation of BDNF. Findings suggest that BDNF signaling in specific brain regions mediates some of the beneficial effects of exercise and energy restriction on peripheral energy metabolism and the cardiovascular system. Collectively, the findings described in this article suggest the possibility of developing prescriptions for optimal brain health based on activity-dependent BDNF signaling.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Encéfalo/fisiología , Ejercicio Físico/fisiología , Animales , Humanos , Plasticidad Neuronal/fisiología , Transducción de Señal , Estrés Fisiológico
6.
Neurosignals ; 21(1-2): 75-88, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22572473

RESUMEN

A decline in cognitive ability is a typical feature of the normal aging process, and of neurodegenerative disorders such as Alzheimer's, Parkinson's and Huntington's diseases. Although their etiologies differ, all of these disorders involve local activation of innate immune pathways and associated inflammatory cytokines. However, clinical trials of anti-inflammatory agents in neurodegenerative disorders have been disappointing, and it is therefore necessary to better understand the complex roles of the inflammatory process in neurological dysfunction. The dietary phytochemical curcumin can exert anti-inflammatory, antioxidant and neuroprotective actions. Here we provide evidence that curcumin ameliorates cognitive deficits associated with activation of the innate immune response by mechanisms requiring functional tumor necrosis factor α receptor 2 (TNFR2) signaling. In vivo, the ability of curcumin to counteract hippocampus-dependent spatial memory deficits, to stimulate neuroprotective mechanisms such as upregulation of BDNF, to decrease glutaminase levels, and to modulate N-methyl-D-aspartate receptor levels was absent in mice lacking functional TNFRs. Curcumin treatment protected cultured neurons against glutamate-induced excitotoxicity by a mechanism requiring TNFR2 activation. Our results suggest the possibility that therapeutic approaches against cognitive decline designed to selectively enhance TNFR2 signaling are likely to be more beneficial than the use of anti-inflammatory drugs per se.


Asunto(s)
Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/metabolismo , Curcumina/uso terapéutico , Lipopolisacáridos/toxicidad , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/fisiología , Animales , Células Cultivadas , Trastornos del Conocimiento/inducido químicamente , Curcumina/farmacología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
7.
Braz. j. med. biol. res ; 45(1): 58-67, Jan. 2012. ilus
Artículo en Inglés | LILACS | ID: lil-610544

RESUMEN

Wnt proteins are involved in tissue development and their signaling pathways play an important role during embryogenesis. Wnt signaling can promote cell survival, which is beneficial for neurons, but could also lead to tumor development in different tissues. The present study investigated the effects of a Wnt protein on the susceptibility of a neural tumor cell line (PC12 cells) to the cytotoxic compounds ferrous sulfate (10 mM), staurosporine (100 and 500 nM), 3-nitropropionic acid (5 mM), and amyloid β-peptide (Aβ25-35; 50 µM). Cells (1 x 10(6) cells/mL) were treated with the Wnt-3a recombinant peptide (200 ng/mL) for 24 h before exposure to toxic insults. The Wnt-3a protein partially protected PC12 cells, with a 6-15 percent increase in cell viability in the presence of toxic agents, similar to the effect measured using the MTT and lactate dehydrogenase cell viability assays. The Wnt-3a protein increased protein expression of β-catenin by 52 percent compared to control. These findings suggest that Wnt signaling can protect neural cells against apoptosis induced by toxic agents, which are relevant to the pathogenesis of Alzheimer’s and Huntington’s diseases.


Asunto(s)
Animales , Ratas , Apoptosis/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , /efectos de los fármacos , Péptidos beta-Amiloides/farmacología , Apoptosis/fisiología , Diferenciación Celular , Proliferación Celular , Compuestos Ferrosos/farmacología , Nitrocompuestos/farmacología , Estrés Oxidativo/fisiología , Propionatos/farmacología , Transducción de Señal/fisiología , Estaurosporina/farmacología , /fisiología
8.
Braz J Med Biol Res ; 45(1): 58-67, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22124704

RESUMEN

Wnt proteins are involved in tissue development and their signaling pathways play an important role during embryogenesis. Wnt signaling can promote cell survival, which is beneficial for neurons, but could also lead to tumor development in different tissues. The present study investigated the effects of a Wnt protein on the susceptibility of a neural tumor cell line (PC12 cells) to the cytotoxic compounds ferrous sulfate (10 mM), staurosporine (100 and 500 nM), 3-nitropropionic acid (5 mM), and amyloid ß-peptide (Aß25-35; 50 µM). Cells (1 x 10(6) cells/mL) were treated with the Wnt-3a recombinant peptide (200 ng/mL) for 24 h before exposure to toxic insults. The Wnt-3a protein partially protected PC12 cells, with a 6-15% increase in cell viability in the presence of toxic agents, similar to the effect measured using the MTT and lactate dehydrogenase cell viability assays. The Wnt-3a protein increased protein expression of ß-catenin by 52% compared to control. These findings suggest that Wnt signaling can protect neural cells against apoptosis induced by toxic agents, which are relevant to the pathogenesis of Alzheimer's and Huntington's diseases.


Asunto(s)
Apoptosis/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteína Wnt3A/efectos de los fármacos , Péptidos beta-Amiloides/farmacología , Animales , Apoptosis/fisiología , Diferenciación Celular , Proliferación Celular , Compuestos Ferrosos/farmacología , Nitrocompuestos/farmacología , Estrés Oxidativo/fisiología , Células PC12 , Propionatos/farmacología , Ratas , Transducción de Señal/fisiología , Estaurosporina/farmacología , Proteína Wnt3A/fisiología
9.
Int J Obes (Lond) ; 35(5): 714-27, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-20921964

RESUMEN

BACKGROUND: The problems of adherence to energy restriction in humans are well known. OBJECTIVE: To compare the feasibility and effectiveness of intermittent continuous energy (IER) with continuous energy restriction (CER) for weight loss, insulin sensitivity and other metabolic disease risk markers. DESIGN: Randomized comparison of a 25% energy restriction as IER (∼ 2710 kJ/day for 2 days/week) or CER (∼ 6276 kJ/day for 7 days/week) in 107 overweight or obese (mean (± s.d.) body mass index 30.6 (± 5.1) kg m(-2)) premenopausal women observed over a period of 6 months. Weight, anthropometry, biomarkers for breast cancer, diabetes, cardiovascular disease and dementia risk; insulin resistance (HOMA), oxidative stress markers, leptin, adiponectin, insulin-like growth factor (IGF)-1 and IGF binding proteins 1 and 2, androgens, prolactin, inflammatory markers (high sensitivity C-reactive protein and sialic acid), lipids, blood pressure and brain-derived neurotrophic factor were assessed at baseline and after 1, 3 and 6 months. RESULTS: Last observation carried forward analysis showed that IER and CER are equally effective for weight loss: mean (95% confidence interval ) weight change for IER was -6.4 (-7.9 to -4.8) kg vs -5.6 (-6.9 to -4.4) kg for CER (P-value for difference between groups = 0.4). Both groups experienced comparable reductions in leptin, free androgen index, high-sensitivity C-reactive protein, total and LDL cholesterol, triglycerides, blood pressure and increases in sex hormone binding globulin, IGF binding proteins 1 and 2. Reductions in fasting insulin and insulin resistance were modest in both groups, but greater with IER than with CER; difference between groups for fasting insulin was -1.2 (-1.4 to -1.0) µU ml(-1) and for insulin resistance was -1.2 (-1.5 to -1.0) µU mmol(-1) l(-1) (both P = 0.04). CONCLUSION: IER is as effective as CER with regard to weight loss, insulin sensitivity and other health biomarkers, and may be offered as an alternative equivalent to CER for weight loss and reducing disease risk.


Asunto(s)
Restricción Calórica , Resistencia a la Insulina , Síndrome Metabólico/terapia , Sobrepeso/terapia , Pérdida de Peso , Adulto , Biomarcadores/metabolismo , Neoplasias de la Mama/prevención & control , Enfermedades Cardiovasculares/prevención & control , Estudios de Factibilidad , Femenino , Humanos , Síndrome Metabólico/metabolismo , Persona de Mediana Edad , Sobrepeso/metabolismo , Cooperación del Paciente/estadística & datos numéricos , Factores de Riesgo
11.
Genes Brain Behav ; 9(5): 512-8, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20345896

RESUMEN

Brain-derived neurotrophic factor (BDNF) regulates synaptic plasticity and neurogenesis, and BDNF plasma and serum levels have been associated with depression, Alzheimer's disease, and other psychiatric and neurodegenerative disorders. In a relatively large community sample, drawn from the Baltimore Longitudinal Study of Aging (BLSA), we examine whether BDNF plasma concentration is associated with the Val66Met functional polymorphism of the BDNF gene (n = 335) and with depression-related personality traits assessed with the NEO-PI-R (n = 391). Plasma concentration of BDNF was not associated with the Val66Met variant in either men or women. However, in men, but not in women, BDNF plasma level was associated with personality traits linked to depression. Contrary to the notion that low BDNF is associated with negative outcomes, we found lower plasma levels in men who score lower on depression and vulnerability to stress (two facets of Neuroticism) and higher on Conscientiousness and Extraversion. These findings challenge the prevailing hypothesis that lower peripheral levels of BDNF are a marker of depression.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/sangre , Factor Neurotrófico Derivado del Encéfalo/genética , Depresión/sangre , Personalidad/genética , Adulto , Anciano , Anciano de 80 o más Años , Envejecimiento/fisiología , Biomarcadores/sangre , Depresión/genética , Femenino , Variación Genética , Humanos , Masculino , Persona de Mediana Edad , Personalidad/fisiología , Polimorfismo de Nucleótido Simple , Valores de Referencia , Factores Sexuales , Estadísticas no Paramétricas
12.
Neuroscience ; 166(2): 508-21, 2010 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-20045450

RESUMEN

Matrix metalloproteinases (MMPs) are a family of zinc-dependent endopeptidases that can be released or activated in a neuronal activity dependent manner. Although pathologically elevated levels of MMPs may be synaptotoxic, physiologically appropriate levels of MMPs may instead enhance synaptic transmission. MMP inhibitors can block long term potentiation (LTP), and at least one family member can affect an increase in the volume of dendritic spines. While the mechanism by which MMPs affect these changes is not completely understood, one possibility is that the cleavage of specific synaptic cell adhesion molecules plays a role. In the present study, we have examined the ability of neuronal activity to stimulate rapid MMP dependent shedding of the intercellular adhesion molecule-5 (ICAM-5), a synaptic adhesion molecule that is thought to inhibit the maturation and enlargement of dendritic spines. Since such cleavage would likely occur within minutes if it were relevant to a process such as LTP, we focused on post stimulus time points of 30 min or less. We show that NMDA can stimulate rapid shedding of ICAM-5 from cortical neurons in dissociated cell cultures and that such shedding is diminished by pretreatment of cultures with inhibitors that target MMP-3 and -9, proteases thought to influence synaptic plasticity. Additional studies suggest that MMP mediated cleavage of ICAM-5 occurs at amino acid 780, so that the major portion of the ectodomain is released. Since reductions in ICAM-5 have been linked to changes in dendritic spine morphology that are associated with LTP, we also examined the possibility that MMP dependent ICAM-5 shedding occurs following high frequency tetanic stimulation of murine hippocampal slices. Results show that the shedding of ICAM-5 occurs in association with LTP, and that both LTP and the associated ICAM-5 shedding are reduced when slices are pretreated with an MMP inhibitor. Together, these findings suggest that neuronal activity is linked to the shedding of a molecule that may inhibit dendritic spine enlargement and that MMPs can affect this change. While further studies will be necessary to determine the extent to which cleavage of ICAM-5 in particular contributes to MMP dependent LTP, our data support an emerging body of literature suggesting that MMPs are critical mediators of synaptic plasticity.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Hipocampo/metabolismo , Potenciación a Largo Plazo/fisiología , Metaloproteinasas de la Matriz/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Análisis de Varianza , Animales , Western Blotting , Células Cultivadas , Espinas Dendríticas/metabolismo , Estimulación Eléctrica , Electrofisiología , Hipocampo/citología , Hipocampo/efectos de los fármacos , Inmunohistoquímica , N-Metilaspartato/farmacología , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Neuronas/citología , Neuronas/efectos de los fármacos , Ratas
13.
Neuromolecular Med ; 11(2): 63-75, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19562525

RESUMEN

Overproduction of the beta-amyloid fragment 1-42 (A beta(1-42)) is thought to contribute to synaptic dysfunction and neuronal death in Alzheimer's disease. Mounting evidence suggests that purinergic receptors play critical roles in synaptic plasticity and neuronal survival, but the potential involvement of these receptors in A beta(1-42)-induced synaptic dysfunction and neuronal death has not been addressed. Here we report that A beta(1-42) promoted accumulation of the calcium-permeable purinergic receptor P2X4 in neurons. We also report evidence that A beta(1-42) induced a caspase-3-mediated cleavage of the receptor that slowed channel closure times and prevented agonist-induced internalization of the receptor. Molecular interference to reduce the expression of P2X4 in primary rodent neurons attenuated A beta(1-42)-induced neuronal death while induced expression of P2X4 in a neuronal cell line that does not normally express P2-receptors enhanced the toxic effect of A beta(1-42). Together these findings suggest that A beta(1-42)-induced synaptic dysfunction and neuronal death may involve perturbations in P2X4 purinergic receptors.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Caspasa 3/metabolismo , Muerte Celular/fisiología , Neuronas/metabolismo , Fragmentos de Péptidos/metabolismo , Purinas/toxicidad , Receptores Purinérgicos P2/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Secuencia de Aminoácidos , Péptidos beta-Amiloides/genética , Animales , Encéfalo/citología , Encéfalo/metabolismo , Encéfalo/patología , Calcio/metabolismo , Hipocampo/citología , Hipocampo/patología , Humanos , Datos de Secuencia Molecular , Neuronas/citología , Técnicas de Placa-Clamp , Fragmentos de Péptidos/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Purinérgicos P2X4 , Sinapsis/metabolismo
14.
Neuroscience ; 161(2): 403-12, 2009 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-19344753

RESUMEN

Numb is an evolutionarily conserved protein that controls the differentiation of neuronal progenitor cells by unknown mechanisms. Here we report that the neural cells expressing Numb isoforms with short phosphotyrosine-binding (SPTB) domain undergo extensive neurite outgrowth, an effect that can be blocked by voltage-gated Ca2+ channel (VGCC) inhibitor or by Ca2+ chelator. In contrast, tyrosine kinase inhibitor, genistein, and selective receptor tyrosine kinase (TrkA) inhibitor, K252alpha did not affect SPTB Numb-mediated neurite outgrowth. MAP kinase inhibitor, PD98059 partially reduced SPTB Numb-mediated neurite outgrowth. Cells expressing SPTB Numbs exhibit increased whole-cell Ca2+ current densities (ICa) which can be prevented by preincubation of either nifedipine or PD98095. Cells expressing LPTB Numbs expressed little ICa (density) and were not able to grow neurites. Our results indicate that Ca2+ influx through VGCC may be required for SPTB Numb-mediated neurite outgrowth, suggesting that Numb promotes neuronal differentiation by a mechanism involving PTB domain-specific regulation of Ca2+ influx and MAP kinase activation.


Asunto(s)
Canales de Calcio Tipo L/fisiología , Proteínas de la Membrana/fisiología , Proteínas del Tejido Nervioso/fisiología , Neuritas/fisiología , Animales , Calcio/metabolismo , Agonistas de los Canales de Calcio/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/biosíntesis , Activación del Canal Iónico , Cinética , Proteínas de la Membrana/genética , Factor de Crecimiento Nervioso/farmacología , Proteínas del Tejido Nervioso/genética , Células PC12 , Técnicas de Placa-Clamp , Fosfotirosina/metabolismo , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Estructura Terciaria de Proteína , Subunidades de Proteína/biosíntesis , Ratas
15.
Neuroscience ; 158(3): 1074-89, 2009 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-18691639

RESUMEN

Activation of the complement system occurs in a variety of neuroinflammatory diseases and neurodegenerative processes of the CNS. Studies in the last decade have demonstrated that essentially all of the activation components and receptors of the complement system are produced by astrocytes, microglia, and neurons. There is also rapidly growing evidence to indicate an active role of the complement system in cerebral ischemic injury. In addition to direct cell damage, regional cerebral ischemia and reperfusion (I/R) induces an inflammatory response involving complement activation and generation of active fragments, such as C3a and C5a anaphylatoxins, C3b, C4b, and iC3b. The use of specific inhibitors to block complement activation or their mediators such as C5a, can reduce local tissue injury after I/R. Consistent with therapeutic approaches that have been successful in models of autoimmune disorders, many of the same complement inhibition strategies are proving effective in animal models of cerebral I/R injury. One new form of therapy, which is less specific in its targeting of complement than monodrug administration, is the use of immunoglobulins. Intravenous immunoglobulin (IVIG) has the potential to inhibit multiple components of inflammation, including complement fragments, pro-inflammatory cytokine production and leukocyte cell adhesion. Thus, IVIG may directly protect neurons, reduce activation of intrinsic inflammatory cells (microglia) and inhibit transendothelial infiltration of leukocytes into the brain parenchyma following an ischemic stroke. The striking neuroprotective actions of IVIG in animal models of ischemic stroke suggest a potential therapeutic potential that merits consideration for clinical trials in stroke patients.


Asunto(s)
Infarto Encefálico/tratamiento farmacológico , Infarto Encefálico/inmunología , Inactivadores del Complemento/uso terapéutico , Citoprotección/inmunología , Inmunoglobulinas Intravenosas/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/inmunología , Anafilatoxinas/antagonistas & inhibidores , Anafilatoxinas/metabolismo , Animales , Infarto Encefálico/fisiopatología , Quimiotaxis de Leucocito/efectos de los fármacos , Quimiotaxis de Leucocito/inmunología , Proteínas del Sistema Complemento/efectos de los fármacos , Proteínas del Sistema Complemento/metabolismo , Citoprotección/efectos de los fármacos , Encefalitis/tratamiento farmacológico , Encefalitis/inmunología , Encefalitis/fisiopatología , Humanos , Inmunoglobulinas Intravenosas/farmacología , Microglía/efectos de los fármacos , Microglía/inmunología , Accidente Cerebrovascular/fisiopatología
16.
J Neurosci Res ; 86(4): 845-60, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17969100

RESUMEN

Amyloid beta-peptide (A beta) likely causes functional alterations in neurons well prior to their death. Nuclear factor-kappaB (NF-kappaB), a transcription factor that is known to play important roles in cell survival and apoptosis, has been shown to be modulated by A beta in neurons and glia, but the mechanism is unknown. Because A beta has also been shown to enhance activation of N-methyl-D-aspartate (NMDA) receptors, we investigated the role of NMDA receptor-mediated intracellular signaling pathways in A beta-induced NF-kappaB activation in primary cultured rat cerebellar cells. Cells were treated with different concentrations of A beta1-40 (1 or 2 microM) for different periods (6, 12, or 24 hr). MK-801 (NMDA antagonist), manumycin A and FTase inhibitor 1 (farnesyltransferase inhibitors), PP1 (Src-family tyrosine kinase inhibitor), PD98059 [mitogen-activated protein kinase (MAPK) inhibitor], and LY294002 [phosphatidylinositol 3-kinase (PI3-k) inhibitor] were added 20 min before A beta treatment of the cells. A beta induced a time- and concentration-dependent activation of NF-kappaB (1 microM, 12 hr); both p50/p65 and p50/p50 NF-kappaB dimers were involved. This activation was abolished by MK-801 and attenuated by manumycin A, FTase inhibitor 1, PP1, PD98059, and LY294002. A beta at 1 microM increased the expression of inhibitory protein I kappaB, brain-derived neurotrophic factor, inducible nitric oxide synthase, tumor necrosis factor-alpha, and interleukin-1 beta as shown by RT-PCR assays. Collectively, these findings suggest that A beta activates NF-kappaB by an NMDA-Src-Ras-like protein through MAPK and PI3-k pathways in cultured cerebellar cells. This pathway may mediate an adaptive, neuroprotective response to A beta.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , N-Metilaspartato/metabolismo , FN-kappa B/metabolismo , Neuronas/metabolismo , Transducción de Señal/fisiología , Animales , Células Cultivadas , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Maleato de Dizocilpina/farmacología , Ensayo de Cambio de Movilidad Electroforética , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Técnica del Anticuerpo Fluorescente , Expresión Génica/efectos de los fármacos , Immunoblotting , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , N-Metilaspartato/efectos de los fármacos , Neuronas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos
17.
Neurology ; 68(18): 1481-7, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17470750

RESUMEN

BACKGROUND: Infection with HIV can result in a debilitating CNS disorder known as HIV dementia (HIV-D). Since the advent of highly active antiretroviral therapy (HAART), the incidence of HIV-D has declined, but the prevalence continues to increase. In this new era of HIV-D, traditional biomarkers such as CSF viral load and monocyte chemotactic protein 1 levels are less likely to be associated with dementia in patients on HAART and biomarkers that can predict HIV-D have not yet been identified. OBJECTIVE: To identify biomarkers that are associated with and can predict HIV-D. METHODS: We grouped patients with HIV based on changes in cognitive status over a 1-year period and analyzed sphingolipid, sterol, triglyceride, antioxidant, and lipid peroxidation levels in CSF. RESULTS: We found that increased levels of the vitamin E and triglyceride C52 predicted the onset or worsening of dementia. Elevated levels of sphingomyelin were associated with inactive dementia. Elevated levels of ceramide and the accumulation of 4-hydroxynonenals were associated with active dementia. CONCLUSIONS: We interpret these findings to indicate that early in the pathogenesis of HIV dementia, there is an up-regulation of endogenous antioxidant defenses in brain. The failure of this attempted neuroprotective mechanism leads to the accumulation of sphingomyelin and moderate cognitive dysfunction. The breakdown of this enlarged pool of sphingomyelin to ceramide and the accumulation of highly reactive aldehydes are associated with declining cognitive function. Thus, elevations in endogenous protective mechanisms may identify patients who are at increased risk of the development of HIV dementia.


Asunto(s)
Complejo SIDA Demencia/líquido cefalorraquídeo , Complejo SIDA Demencia/diagnóstico , Líquido Cefalorraquídeo/química , Infecciones por VIH/complicaciones , VIH-1 , Complejo SIDA Demencia/fisiopatología , Adulto , Aldehídos/análisis , Aldehídos/líquido cefalorraquídeo , Antioxidantes/análisis , Antioxidantes/metabolismo , Biomarcadores/líquido cefalorraquídeo , Encéfalo/inmunología , Encéfalo/fisiopatología , Encéfalo/virología , Ceramidas/análisis , Ceramidas/líquido cefalorraquídeo , Femenino , Humanos , Peroxidación de Lípido , Masculino , Persona de Mediana Edad , Estrés Oxidativo , Valor Predictivo de las Pruebas , Esfingolípidos/análisis , Esfingolípidos/líquido cefalorraquídeo , Esteroles/análisis , Esteroles/líquido cefalorraquídeo , Triglicéridos/análisis , Triglicéridos/líquido cefalorraquídeo , Regulación hacia Arriba , Vitamina E/análisis , Vitamina E/líquido cefalorraquídeo
18.
Neuroscience ; 145(4): 1439-48, 2007 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-17207936

RESUMEN

Postmitotic neurons must survive for the entire life of the organism and be able to respond adaptively to adverse conditions of oxidative and genotoxic stress. Unrepaired DNA damage can trigger apoptosis of neurons which is typically mediated by the ataxia telangiectasia mutated (ATM)-p53 pathway. As in all mammalian cells, telomeres in neurons consist of TTAGGG DNA repeats and several associated proteins that form a nucleoprotein complex that prevents chromosome ends from being recognized as double strand breaks. Proteins that stabilize telomeres include TRF1 and TRF2, and proteins known to play important roles in DNA damage responses and DNA repair including ATM, Werner and the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs). We have been performing studies of developing and adult neurons aimed at understanding the effects of global and telomere-directed DNA damage responses in neuronal plasticity and survival in the contexts of aging and neurodegenerative disorders. Deficits in specific DNA repair proteins, including DNA-PKcs and uracil DNA glycosylase (UDG), render neurons vulnerable to adverse conditions of relevance to the pathogenesis of neurodegenerative disorders such as Alzheimer's disease and stroke. Similarly, early postmitotic neurons with reduced telomerase activity exhibit accentuated responses to DNA damage and are prone to apoptosis demonstrating a pivotal role for telomere maintenance in both mitotic cells and postmitotic neurons. Our recent findings suggest key roles for TRF2 in regulating the differentiation and survival of neurons. TRF2 affects cell survival and differentiation by modulating DNA damage pathways, and gene expression. A better understanding of the molecular mechanisms by which neurons respond to global and telomere-specific DNA damage may reveal novel strategies for prevention and treatment of neurodegenerative disorders. Indeed, work in this and other laboratories has shown that dietary folic acid can protect neurons against Alzheimer's disease by keeping homocysteine levels low and thereby minimizing the misincorporation of uracil into DNA in neurons.


Asunto(s)
Senescencia Celular/genética , Daño del ADN/genética , Sistema Nervioso/metabolismo , Enfermedades Neurodegenerativas/genética , Neuronas/metabolismo , Telómero/genética , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/genética , Trastornos por Deficiencias en la Reparación del ADN/genética , Trastornos por Deficiencias en la Reparación del ADN/metabolismo , Trastornos por Deficiencias en la Reparación del ADN/fisiopatología , Proteínas de Unión al ADN/genética , Humanos , Sistema Nervioso/citología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/fisiopatología , Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Similares a la Proteína de Unión a TATA-Box/genética , Proteína 2 de Unión a Repeticiones Teloméricas , Proteínas Supresoras de Tumor/genética
19.
Amino Acids ; 32(3): 299-304, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-16998712

RESUMEN

The amino acid glutamate, the major excitatory neurotransmitter in the central nervous system, activates receptors coupled to calcium influx. Excessive activation of glutamate receptors in conditions such as severe epileptic seizures or stroke can kill neurons in a process called excitotoxicity. However, subtoxic levels of activation of the N-methyl-D-aspartate (NMDA) type of glutamate receptor elicit adaptive responses in neurons that enhance their ability to withstand more severe stress. A variety of stimuli induce adaptive responses to protect neurons. For example, sublethal ischemic episodes or a mild epileptic insult can protect neurons in a process referred to as tolerance. The molecular mechanisms that protect neurons by these different stressful stimuli are largely unknown but they share common features such as the transcription factor, nuclear factor kappa B (NF-kappaB), which is activated by ischemic and epileptic preconditioning as well as exposure to subtoxic NMDA concentrations. In this article, we describe stress-induced neuroprotective mechanisms highlighting the role of brain-derived neurotrophic factor (BDNF), a protein that plays a crucial role in neuronal survival and maintenance, neurogenesis and learning and memory.


Asunto(s)
Adaptación Fisiológica , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Precondicionamiento Isquémico , Receptores de N-Metil-D-Aspartato/metabolismo , Convulsiones/metabolismo , Animales , Calcio/metabolismo , Señalización del Calcio , Ácido Glutámico/metabolismo , Humanos , Aprendizaje , Memoria , N-Metilaspartato/metabolismo , FN-kappa B/metabolismo , Neuronas/metabolismo
20.
J Neurochem ; 98(4): 1176-84, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16895583

RESUMEN

The hippocampal glucocorticoid receptor (GR) is involved in negative feedback regulation of the hypothalamo-pituitary-adrenal axis and is believed to transduce the deleterious effects of glucocorticoids in depression and age-related memory loss. Regulation and intracellular trafficking of the GR are critical determinants of GR action in both health and disease. Here, we show dynamic regulation of GR and its interaction with its principal intracellular chaperone, heat-shock protein (HSP) 90, across the circadian cycle. Our initial experiments indicate that cytosolic hippocampal GR protein is elevated in the evening (PM), whereas nuclear GR and cytosolic HSP90, HSP70 and heat-shock cognate 70 (HSC70), are unchanged. In contrast, there are no changes in examined proteins in the hypothalamus. Immunoprecipitation experiments reveal increased GR-HSP90 associations in the hippocampus in the PM, whereas binding in the hypothalamus is decreased in the PM. Given that GR requires HSP90 for ligand binding, the data suggest that circadian GR signaling capacity is regulated in a region-specific pattern.


Asunto(s)
Química Encefálica/fisiología , Proteínas HSP90 de Choque Térmico/biosíntesis , Receptores de Glucocorticoides/metabolismo , Hormona Adrenocorticotrópica/sangre , Animales , Núcleo Celular , Ritmo Circadiano/fisiología , Corticosterona/farmacología , Citosol/metabolismo , Hipocampo/metabolismo , Hidrocortisona/sangre , Hipotálamo/metabolismo , Immunoblotting , Inmunoprecipitación , Masculino , Radioinmunoensayo , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...