Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 18(1)2017 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-28106772

RESUMEN

CSP-1103 (formerly CHF5074) has been shown to reverse memory impairment and reduce amyloid plaque as well as inflammatory microglia activation in preclinical models of Alzheimer's disease. Moreover, it was found to improve cognition and reduce brain inflammation in patients with mild cognitive impairment. Recent evidence suggests that CSP-1103 acts through a single molecular target, the amyloid precursor protein intracellular domain (AICD), a transcriptional regulator implicated in inflammation and apoptosis. We here tested the possible anti-apoptotic and neuroprotective activity of CSP-1103 in a cell-based model of post-ischemic injury, wherein the primary mouse cortical neurons were exposed to oxygen-glucose deprivation (OGD). When added after OGD, CSP-1103 prevented the apoptosis cascade by reducing cytochrome c release and caspase-3 activation and the secondary necrosis. Additionally, CSP-1103 limited earlier activation of p38 and nuclear factor κB (NF-κB) pathways. These results demonstrate that CSP-1103 is neuroprotective in a model of post-ischemic brain injury and provide further mechanistic insights as regards its ability to reduce apoptosis and potential production of pro-inflammatory cytokines. In conclusion, these findings suggest a potential use of CSP-1103 for the treatment of brain ischemia.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclopropanos/farmacología , Flurbiprofeno/análogos & derivados , Glucosa/deficiencia , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Oxígeno/farmacología , Animales , Caspasa 3/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Corteza Cerebral/patología , Citocromos c/metabolismo , Activación Enzimática/efectos de los fármacos , Flurbiprofeno/farmacología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Ibuprofeno/farmacología , Ratones Endogámicos C57BL , Necrosis , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Factor de Transcripción ReIA/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
2.
Sci Rep ; 4: 4618, 2014 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-24714650

RESUMEN

Amyloid precursor protein (APP) intracellular domain (AICD) is a product of APP processing with transcriptional modulation activity, whose overexpression causes various Alzheimer's disease (AD)-related dysfunctions. Here we report that 1-(3',4'-dichloro-2-fluoro[1,1'-biphenyl]-4-yl)-cyclopropanecarboxylic acid) (CHF5074), a compound that favorably affects neurodegeneration, neuroinflammation and memory deficit in transgenic mouse models of AD, interacts with the AICD and impairs its nuclear activity. In neuroglioma-APPswe cells, CHF5074 shifted APP cleavage from Aß42 to the less toxic Aß38 peptide without affecting APP-C-terminal fragment, nor APP levels. As revealed by photoaffinity labeling, CHF5074 does not interact with γ-secretase, but binds to the AICD and lowers its nuclear translocation. In vivo treatment with CHF5074 reduced AICD occupancy as well as histone H3 acetylation levels and transcriptional output of the AICD-target gene KAI1. The data provide new mechanistic insights on this compound, which is under clinical investigation for AD treatment/prevention, as well as on the contribution of the AICD to AD pathology.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Ciclopropanos/farmacología , Flurbiprofeno/análogos & derivados , Fragmentos de Péptidos/metabolismo , Acetilación , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/prevención & control , Línea Celular Tumoral , Flurbiprofeno/farmacología , Histonas/metabolismo , Humanos , Proteína Kangai-1/biosíntesis , Proteína Kangai-1/genética , Estructura Terciaria de Proteína , Transcripción Genética
3.
Neurobiol Dis ; 49: 177-89, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22971966

RESUMEN

UNLABELLED: Nuclear factor-kappaB (NF-κB) p50/RelA is a key molecule with a dual effect in the progression of ischemic stroke. In harmful ischemia, but not in preconditioning insult, neurotoxic activation of p50/RelA is characterized by RelA-specific acetylation at Lys310 (K310) and deacetylation at other Lys residues. The derangement of RelA acetylation is associated with activation of Bim promoter. OBJECTIVE: With the aim of producing neuroprotection by correcting altered acetylation of RelA in brain ischemia, we combined the pharmacological inhibition of histone deacetylase (HDAC) 1-3, the enzymes known to reduce global RelA acetylation, and the activation of sirtuin 1, endowed with a specific deacetylase activity on the K310 residue of RelA. To afford this aim, we tested the clinically used HDAC 1-3 inhibitor entinostat (MS-275) and the sirtuin 1 activator resveratrol. METHODS: We used the mouse model of transient middle cerebral artery occlusion (MCAO) and primary cortical neurons exposed to oxygen glucose deprivation (OGD). RESULTS: The combined use of MS-275 and resveratrol, by restoring normal RelA acetylation, elicited a synergistic neuroprotection in neurons exposed to OGD. This effect correlated with MS-275 capability to increase total RelA acetylation and resveratrol capability to reduce RelA K310 acetylation through the activation of an AMP-activated protein kinase-sirtuin 1 pathway. The synergistic treatment reproduced the acetylation state of RelA peculiar of preconditioning ischemia. Neurons exposed to the combined drugs totally recovered the optimal histone H3 acetylation. Neuroprotection was reproduced in mice subjected to MCAO and treated with MS-275 (20µg/kg and 200µg/kg) or resveratrol (6800µg/kg) individually. However, the administration of lowest doses of MS-275 (2µg/kg) and resveratrol (68µg/kg) synergistically reduced infarct volume and neurological deficits. Importantly, the treatment was effective even when administered 7h after the stroke onset. Chromatin immunoprecipitation analysis of cortices harvested from treated mice showed that the RelA binding and histone acetylation increased at the Bcl-xL promoter and decreased at the Bim promoter. CONCLUSION: Our study reveals that epigenetic therapy shaping acetylation of both RelA and histones may be a promising strategy to limit post-ischemic injury with an extended therapeutic window.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Epigénesis Genética/efectos de los fármacos , Histonas/metabolismo , FN-kappa B/metabolismo , Fármacos Neuroprotectores/farmacología , Factor de Transcripción ReIA/metabolismo , Acetilación/efectos de los fármacos , Animales , Benzamidas/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Modelos Animales de Enfermedad , Epigénesis Genética/fisiología , Glucosa/deficiencia , Inhibidores de Histona Desacetilasas/farmacología , Infarto de la Arteria Cerebral Media , Masculino , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/patología , Piridinas/farmacología , Resveratrol , Sirtuina 1/metabolismo , Estilbenos/farmacología
4.
PLoS One ; 7(5): e38019, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22666436

RESUMEN

The molecular mechanisms responsible for increasing iron and neurodegeneration in brain ischemia are an interesting area of research which could open new therapeutic approaches. Previous evidence has shown that activation of nuclear factor kappa B (NF-κB) through RelA acetylation on Lys310 is the prerequisite for p50/RelA-mediated apoptosis in cellular and animal models of brain ischemia. We hypothesized that the increase of iron through a NF-κB-regulated 1B isoform of the divalent metal transporter-1 (1B/DMT1) might contribute to post-ischemic neuronal damage. Both in mice subjected to transient middle cerebral artery occlusion (MCAO) and in neuronally differentiated SK-N-SH cells exposed to oxygen-glucose-deprivation (OGD), 1A/DMT1 was only barely expressed while the 1B/DMT1 without iron-response-element (-IRE) protein and mRNA were early up-regulated. Either OGD or over-expression of 1B/(-)IRE DMT1 isoform significantly increased iron uptake, as detected by total reflection X-ray fluorescence, and iron-dependent cell death. Iron chelation by deferoxamine treatment or (-)IRE DMT1 RNA silencing displayed significant neuroprotection against OGD which concomitantly decreased intracellular iron levels. We found evidence that 1B/(-)IRE DMT1 was a target gene for RelA activation and acetylation on Lys310 residue during ischemia. Chromatin immunoprecipitation analysis of the 1B/DMT1 promoter showed there was increased interaction with RelA and acetylation of H3 histone during OGD exposure of cortical neurons. Over-expression of wild-type RelA increased 1B/DMT1 promoter-luciferase activity, the (-)IRE DMT1 protein, as well as neuronal death. Expression of the acetylation-resistant RelA-K310R construct, which carried a mutation from lysine 310 to arginine, but not the acetyl-mimic mutant RelA-K310Q, down-regulated the 1B/DMT1 promoter, consequently offering neuroprotection. Our data showed that 1B/(-)IRE DMT1 expression and intracellular iron influx are early downstream responses to NF-κB/RelA activation and acetylation during brain ischemia and contribute to the pathogenesis of stroke-induced neuronal damage.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Proteínas de Transporte de Catión/metabolismo , Regulación de la Expresión Génica , Lisina , Factor de Transcripción ReIA/química , Factor de Transcripción ReIA/metabolismo , Acetilación , Animales , Transporte Biológico , Isquemia Encefálica/genética , Proteínas de Transporte de Catión/genética , Muerte Celular , Línea Celular Tumoral , Glucosa/deficiencia , Histonas/metabolismo , Humanos , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Espacio Intracelular/metabolismo , Hierro/metabolismo , Masculino , Ratones , Neuronas/patología , Oxígeno/metabolismo , Regiones Promotoras Genéticas/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología , Activación Transcripcional , Regulación hacia Arriba
5.
Brain Res ; 1476: 203-10, 2012 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-22575713

RESUMEN

Brain cells display an amazing ability to respond to several different types of environmental stimuli and integrate this response physiologically. Some of these responses can outlive the original stimulus by days, weeks or even longer. Long-lasting changes in both physiological and pathological conditions occurring in response to external stimuli are almost always mediated by changes in gene expression. To effect these changes, cells have developed an impressive repertoire of signaling systems designed to modulate the activity of numerous transcription factors and epigenetic mechanisms affecting the chromatin structure. Since its initial characterization in the nervous system, NF-κB has shown to respond to multiple signals and elicit pleiotropic activities suggesting that it may play a pivotal role in integration of different types of information within the brain. Ample evidence demonstrates that NF-κB factors are engaged in and necessary for neuronal development and synaptic plasticity, but they also regulate brain response to environmental noxae. By focusing on the complexity of NF-κB transcriptional activity in neuronal cell death, it emerged that the composition of NF-κB active dimers finely tunes the neuronal vulnerability to brain ischemia. Even though we are only beginning to understand the contribution of distinct NF-κB family members to the regulation of gene transcription in the brain, an additional level of regulation of NF-κB activity has emerged as operated by the epigenetic mechanisms modulating histone acetylation. We will discuss NF-κB and epigenetic mechanisms as integrative regulators of brain resilience to anoxic stress and useful drug targets for restoration of brain function. This article is part of a Special Issue entitled: Brain Integration.


Asunto(s)
Isquemia Encefálica/patología , Encéfalo/metabolismo , Encéfalo/fisiopatología , Epigénesis Genética , FN-kappa B/metabolismo , Transducción de Señal/genética , Animales , Encéfalo/patología , Humanos , Modelos Biológicos , Neuronas/metabolismo
6.
J Alzheimers Dis ; 24(4): 799-816, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21321397

RESUMEN

Abnormal amyloid-ß (Aß) production and deposition is believed to represent one of the main causes of Alzheimer's disease (AD). γ-Secretase is the enzymatic complex responsible for Aß generation from its precursor protein. Inhibition or modulation of γ-secretase represents an attractive therapeutic approach. CHF5074 is a new γ-secretase modulator that has been shown to inhibit brain plaque deposition and to attenuate memory deficit in adult AD transgenic mice after chronic treatment. To date, it is not known whether the positive behavioral effects of this compound also occur in young transgenic mice without plaque deposition. Here, we evaluated the effects of acute and subchronic treatment with CHF5074 on contextual and recognition memory and on hippocampal synaptic plasticity in plaque-free Tg2576 mice. We found that at 5 months of age, contextual memory impairment was significantly attenuated after acute subcutaneous administration of 30 mg/kg CHF5074. At 6 months of age, recognition memory impairment was fully reversed after a 4-week oral treatment in the diet (≈60 mg/kg/day). These cognitive effects were associated with a reversal of long-term potentiation (LTP) impairment in the hippocampus. A significant reduction in brain intraneuronal AßPP/Aß levels and hyperphosphorylated tau, but no change in soluble or oligomeric Aß levels was detected in Tg2576 mice showing functional recovery following CHF5074 treatment. We conclude that the beneficial effects of CHF5074 treatment in young transgenic mice occurred at a stage that precedes plaque formation and were associated with a reduction in intraneuronal AßPP/Aß and hyperphosphorylated tau.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/fisiología , Ciclopropanos/farmacología , Flurbiprofeno/análogos & derivados , Hipocampo/enzimología , Trastornos de la Memoria/enzimología , Memoria/fisiología , Sinapsis/enzimología , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Cricetinae , Ciclopropanos/uso terapéutico , Femenino , Flurbiprofeno/farmacología , Flurbiprofeno/uso terapéutico , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Humanos , Memoria/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/genética , Plasticidad Neuronal/fisiología , Placa Amiloide , Sinapsis/efectos de los fármacos , Sinapsis/genética
7.
J Mol Neurosci ; 45(1): 22-31, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21181298

RESUMEN

The relationship between ß-amyloid (Aß) and tau is not fully understood, though it is proposed that in the pathogenesis of Alzheimer's disease (AD) Aß accumulation precedes and promotes tau hyperphosphorylation via activation of glycogen synthase kinase-3beta (GSK-3ß). Both events contribute to learning and memory impairments. Modulation of γ-secretase activity has proved to reduce the Aß burden and cognitive deficits in mouse models of AD, but its ability in reducing the tau pathology remains elusive. Chronic treatments with two γ-secretase modulators, ibuprofen and CHF5074, disclosed higher activity of CHF5074 in ameliorating brain plaque deposition and spatial memory deficits in transgenic mice expressing human amyloid precursor protein (hAPP) with Swedish and London mutations (APP(SL) mice). The aim of our study was to investigate in APP(SL) mice the effect of the two compounds on the accumulation of native hyperphosphorylated tau as well as on the GSK-3ß signaling. CHF5074 was more effective than ibuprofen in reducing tau pathology, though both compounds decreased the GSK-3ß level and increased the GSK-3ß inhibitory phosphorylation near to the non-Tg values. The inhibition of GSK-3ß appeared to be secondary to the reduction of Aß generation as, differently from LiCl, CHF5074 reproduced its effect in hAPP-overexpressing neuroglioma cells, but not in wild-type primary neurons. Our data show that the novel γ-secretase modulator CHF5074 can fully reverse ß-amyloid-associated tau pathology, thus representing a promising therapeutic agent for AD.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Ciclopropanos/farmacología , Flurbiprofeno/análogos & derivados , Neuronas/efectos de los fármacos , Proteínas tau/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Animales , Células Cultivadas , Inhibidores de la Ciclooxigenasa/farmacología , Dieta , Modelos Animales de Enfermedad , Flurbiprofeno/farmacología , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Ibuprofeno/farmacología , Ratones , Ratones Transgénicos , Neuronas/citología , Neuronas/metabolismo , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteínas tau/genética
8.
Int Rev Neurobiol ; 85: 351-62, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19607980

RESUMEN

Nuclear factor-kappaB (NF-kappaB) is a dimeric transcription factor composed of five members, p50, RelA/p65, c-Rel, RelB, and p52 that can diversely combine to form the active transcriptional dimer. NF-kappaB controls the expression of genes that regulate a broad range of biological processes in the central nervous system such as synaptic plasticity, neurogenesis, and differentiation. Although NF-kappaB is essential for neuron survival and its activation may protect neurons against oxidative-stresses or ischemia-induced neurodegeneration, NF-kappaB activation can contribute to inflammatory reactions and apoptotic cell death after brain injury and stroke. It was proposed that the death or survival of neurons might depend on the cell type and the timing of NF-kappaB activation. We here discuss recent evidence suggesting that within the same neuronal cell, activation of diverse NF-kappaB dimers drives opposite effects on neuronal survival. Unbalanced activation of NF-kappaB p50/RelA dimer over c-Rel-containing complexes contributes to cell death secondary to the ischemic insult. While p50/RelA acts as transcriptional inducer of Bcl-2 family proapoptotic Bim and Noxa genes, c-Rel dimers specifically promote transcription of antiapototic Bcl-xL gene. Changes in the nuclear content of c-Rel dimers strongly affect the threshold of neuron vulnerability to ischemic insult and agents, likewise leptin, activating a NF-kappaB/c-Rel-dependent transcription elicit neuroprotection in animal models of brain ischemia.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Supervivencia Celular/fisiología , FN-kappa B/fisiología , Neuronas/patología , Neuronas/fisiología , Animales , Encéfalo/metabolismo , Encéfalo/fisiología , Muerte Celular/fisiología , Dimerización , FN-kappa B/biosíntesis , Neuronas/metabolismo , Activación Transcripcional
9.
J Neurochem ; 108(2): 475-85, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19094066

RESUMEN

Diverse nuclear factor-kappaB subunits mediate opposite effects of extracellular signals on neuron survival. While RelA is activated by neurotoxic agents, c-Rel drives neuroprotective effects. In brain ischaemia RelA and p50 factors rapidly activate, but how they associate with c-Rel to form active dimers and contribute to the changes in diverse dimer activation for neuron susceptibility is unknown. We show that in both cortical neurons exposed to oxygen glucose deprivation (OGD) and mice subjected to brain ischaemia, activation of p50/RelA was associated with inhibition of c-Rel/RelA dimer and no change p50/c-Rel. Targeting c-Rel and RelA expression revealed that c-Rel dimers reduced while p50/RelA enhanced neuronal susceptibility to anoxia. Activation of p50/RelA complex is known to induce the pro-apoptotic Bim and Noxa genes. We now show that c-Rel-containing dimers, p50/c-Rel and RelA/c-Rel, but not p50/RelA, promoted Bcl-xL transcription. Accordingly, the OGD exposure induced Bim, but reduced Bcl-xL promoter activity and decreased the content of endogenous Bcl-xL protein. These findings demonstrate that within the same neuronal cell, the balance between activation of p50/RelA and c-Rel-containing complexes fine tunes the threshold of neuron vulnerability to the ischaemic insult. Selective targeting of different dimers will unravel new approaches to limit ischaemia-associated apoptosis.


Asunto(s)
Infarto de la Arteria Cerebral Media/patología , Subunidad p50 de NF-kappa B/metabolismo , Neuronas/fisiología , Proteínas Proto-Oncogénicas c-rel/fisiología , Factor de Transcripción ReIA/fisiología , Animales , Supervivencia Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Embrión de Mamíferos , Regulación de la Expresión Génica/fisiología , Glucosa/deficiencia , Humanos , Hipoxia , Inmunoprecipitación/métodos , Etiquetado Corte-Fin in Situ , Infarto de la Arteria Cerebral Media/fisiopatología , Ratones , Ratones Endogámicos C57BL , Subunidad p50 de NF-kappa B/genética , Neuroblastoma , Proteínas Proto-Oncogénicas c-rel/genética , ARN Interferente Pequeño/farmacología , Factor de Transcripción ReIA/genética , Transfección/métodos , Proteína bcl-X/metabolismo
10.
Stroke ; 40(2): 610-7, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19023096

RESUMEN

BACKGROUND AND PURPOSE: Leptin is an adipose hormone endowed with angiopoietic, neurotrophic, and neuroprotective properties. We tested the hypothesis that leptin might act as an endogenous mediator of recovery after ischemic stroke and investigated whether nuclear transcription factors kappaB activation is involved in leptin-mediated neuroprotection. METHODS: The antiapoptotic effects of leptin were evaluated in cultured mouse cortical neurons from wild-type or NF-kappaB/c-Rel(-/-) mice exposed to oxygen-glucose deprivation. Wild-type, c-Rel(-/-) and leptin-deficient ob/ob mice were subjected to permanent middle cerebral artery occlusion. Leptin production was measured in brains from wild-type mice with quantitative reverse transcriptase-polymerase chain reaction and immunostaining. Mice received a leptin bolus (20 microg/g) intraperitoneally at the onset of ischemia. RESULTS: Leptin treatment activated the nuclear translocation of nuclear transcription factors kappaB dimers containing the c-Rel subunit, induced the expression of the antiapoptotic c-Rel target gene Bcl-xL in both control and oxygen-glucose deprivation conditions, and counteracted the oxygen-glucose deprivation-mediated apoptotic death of cultured cortical neurons. Leptin-mediated Bcl-xL induction and neuroprotection against oxygen-glucose deprivation were hampered in cortical neurons from c-Rel(-/-) mice. Leptin mRNA was induced and the protein was detectable in microglia/macrophage cells from the ischemic penumbra of wild-type mice subjected to permanent middle cerebral artery occlusion. Ob/ob mice were more susceptible than wild-type mice to the permanent middle cerebral artery occlusion injury. Leptin injection significantly reduced the permanent middle cerebral artery occlusion-mediated cortical damage in wild-type and ob/ob mice, but not in c-Rel(-/-) mice. CONCLUSIONS: Leptin acts as an endogenous mediator of neuroprotection during cerebral ischemia. Exogenous leptin administration protects against ischemic neuronal injury in vitro and in vivo in a c-Rel-dependent manner.


Asunto(s)
Isquemia Encefálica/metabolismo , Corteza Cerebral/metabolismo , Leptina/biosíntesis , Leptina/fisiología , FN-kappa B/genética , FN-kappa B/fisiología , Animales , Western Blotting , Células Cultivadas , Infarto Cerebral/patología , ADN/biosíntesis , ADN/genética , Femenino , Técnica del Anticuerpo Fluorescente , Glucosa/deficiencia , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Hipoxia/metabolismo , Inmunohistoquímica , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Embarazo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética , Regulación hacia Arriba/genética , Proteína bcl-X/biosíntesis , Proteína bcl-X/genética
11.
FEBS J ; 276(1): 27-35, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19087197

RESUMEN

Nuclear factor-kappaB (NF-kappaB) has been proposed to serve a dual function as a regulator of neuron survival in pathological conditions associated with neurodegeneration. NF-kappaB is a transcription family of factors comprising five different proteins, namely p50, RelA/p65, c-Rel, RelB and p52, which can combine differently to form active dimers in response to external stimuli. Recent research shows that diverse NF-kappaB dimers lead to cell death or cell survival in neurons exposed to ischemic injury. While the p50/p65 dimer participates in the pathogenesis of post-ischemic injury by inducing pro-apoptotic gene expression, c-Rel-containing dimers increase neuron resistance to ischemia by inducing anti-apoptotic gene transcription. We present, in this report, the latest findings and consider the therapeutic potential of targeting different NF-kappaB dimers to limit ischemia-associated neurodegeneration.


Asunto(s)
Daño Encefálico Crónico/etiología , Isquemia Encefálica/fisiopatología , FN-kappa B/fisiología , Neuronas/patología , Animales , Apoptosis/genética , Daño Encefálico Crónico/patología , Daño Encefálico Crónico/fisiopatología , Isquemia Encefálica/patología , Dimerización , Regulación de la Expresión Génica , Humanos , Factores de Transcripción/metabolismo
12.
J Neurochem ; 106(2): 560-77, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18410503

RESUMEN

Progressive degeneration and intraneuronal Lewy bodies made of filamentous alpha-synuclein (alpha-syn) in dopaminergic cells of the nigrostriatal system are characteristics of Parkinson's disease (PD). Glucose uptake is reduced in some of the brain regions affected by PD neurodegenerative changes. Defects in mitochondrial activity in the substantia nigra have been observed in the brain of patients affected by PD and substantia nigra lesions can induce the onset of a secondary parkinsonism. Thus, energy starvation and consequently metabolic impairment to dopaminergic neurons may be related to the onset of PD. On this line, we evaluated the effect of nutrient starvation, reproduced 'in vitro' by glucose deprivation (GD), in primary mesecephalic neuronal cultures and dopaminergic-differentiated SH-SY5Y cells, to evaluate if decreased glucose support to dopaminergic cells can lead to mitochondrial damage, neurodegeneration and alpha-syn misfolding. Furthermore, we investigated the effect of dopamine (DA) treatment in the presence of a DA-uptake inhibitor or of the D(2)/D(3) receptor (D(2)R/D(3)R) agonist quinpirole on GD-treated cells, to evaluate the efficacy of these therapeutic compounds. We found that GD induced the formation of fibrillary aggregated alpha-syn inclusions containing the DA transporter in dopaminergic cells. These alterations were accompanied by dopaminergic cell death and were exacerbated by DA overload. Conversely, the block of DA uptake and D(2)R/D(3)R agonist treatment exerted neuroprotective effects. These data indicate that glucose starvation is likely involved in the induction of PD-related pathological changes in dopaminergic neurons. These changes may be counteracted by the block of DA uptake and by dopaminergic agonist treatment.


Asunto(s)
Dopamina/metabolismo , Glucosa/deficiencia , Degeneración Nerviosa/etiología , Receptores de Dopamina D2/fisiología , alfa-Sinucleína/metabolismo , Análisis de Varianza , Animales , Benzotiazoles , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Células Cultivadas , Dopamina/farmacología , Dopaminérgicos/farmacología , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Embrión de Mamíferos , Formazáns/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Mesencéfalo/citología , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Sales de Tetrazolio/metabolismo , Tiazoles/metabolismo
13.
Neuropsychopharmacology ; 32(4): 872-80, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16936710

RESUMEN

We originally suggested that inhibition of tumor necrosis factor-related apoptosis inducing ligand (TRAIL) death pathway could be taken into consideration as a potential therapeutic strategy for Alzheimer's disease (AD). However, because the critical role of TRAIL in immune surveillance, the neutralization of TRAIL protein by an antibody to prevent its binding to death receptors is definitely a risky approach. Here, we demonstrated that the blockade of the TRAIL death receptor DR5 with a specific antibody completely prevented amyloid beta peptide (A beta) neurotoxicity in both neuronal cell line and primary cortical neurons. DR5 was demonstrated to be a key factor in TRAIL death pathway. In fact, whereas TRAIL expression was enhanced dose-dependently by concentrations of beta amyloid ranging from 10 nM to 1 microM, only the highest toxic dose of A beta (25 microM) induced the increased expression of DR5 and neuronal cell death. In addition, the increased expression of DR5 receptor after beta amyloid treatment was sustained by p53 transcriptional activity, as demonstrated by the data showing that the p53 inhibitor Pifithrin alpha prevented both beta amyloid-induced DR5 induction and cell death. These data suggest a sequential activation of p53 and DR5 upon beta amyloid exposure. Further insight into the key role of DR5 in AD was suggested by data showing a significant increase of DR5 receptor in cortical slices of AD brain. Thus, these findings may give intracellular TRAIL pathway a role in AD pathophysiology, making DR5 receptor a possible candidate as a pharmacological target.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Apoptosis/efectos de los fármacos , Neuronas/efectos de los fármacos , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Anciano , Enfermedad de Alzheimer/patología , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Neuroblastoma
14.
J Neurosci ; 26(50): 12896-903, 2006 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-17167080

RESUMEN

The transcription factor nuclear factor kappaB (NF-kappaB) is well known for its antiapoptotic action. However, in some disorders, such as cerebral ischemia, a proapoptotic function of NF-kappaB has been demonstrated. To analyze which subunit of NF-kappaB is functional in cerebral ischemia, we induced focal cerebral ischemia in mice with a germline deletion of the p52 or c-Rel gene or with a conditional deletion of RelA in the brain. Only RelA deficiency reduced infarct size. Interestingly, expression of the proapoptotic BH3 (Bcl-2 homology domain 3)-only genes Bim and Noxa in cerebral ischemia depended on RelA and the upstream kinase IKK (IkappaB kinase). RelA stimulated Bim and Noxa gene transcription in primary cortical neurons and bound to the promoter of both genes. Thus, the deleterious function in cerebral ischemia is specific for the NF-kappaB subunit RelA and may be mediated through Bim and Noxa.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/biosíntesis , Isquemia Encefálica/metabolismo , Proteínas de la Membrana/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Proto-Oncogénicas/biosíntesis , Factor de Transcripción ReIA/biosíntesis , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteína 11 Similar a Bcl2 , Isquemia Encefálica/genética , Células Cultivadas , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , FN-kappa B/biosíntesis , FN-kappa B/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Factor de Transcripción ReIA/deficiencia , Factor de Transcripción ReIA/genética , Transcripción Genética/fisiología
15.
Eur J Neurosci ; 23(7): 1711-20, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16623827

RESUMEN

Beta-amyloid (Abeta) peptides are key proteins in the pathophysiology of Alzheimer's disease (AD). While Abeta42 aggregates very rapidly to form early diffuse plaques, supplemental Abeta40 deposition is required to form mature neuritic plaques. We here investigated the role of nuclear factor-kappaB (NF-kappaB) pathway in Abeta40-mediated neuronal damage and amyloid pathology. In rat primary neurons and human postmitotic neuronal cells, the Abeta peptide induced a dose-dependent neuronal death, reduced the levels of the anti-apoptotic protein Bcl-XL, enhanced the cytosolic release of cytochrome c, and elicited the intracellular accumulation and secretion of Abeta42 oligomers. Moreover, Abeta40 activated the NF-kappaB pathway by selectively inducing the nuclear translocation of p65 and p50 subunits, and promoted an apoptotic profile of gene expression. As inhibitors of the NF-kappaB pathway, we tested the capability of a double-stranded kappaB decoy oligonucleotide, the anti-inflammatory drug aspirin and the selective IkappaB kinase 2 inhibitor, AS602868, to modify the Abeta40-mediated effects. These treatments, transiently applied before Abeta exposure, completely inhibited p50/p65 nuclear translocation and neuronal damage. The kappaB decoy also inhibited the Abeta-induced release of cytochrome c, restored the levels of Bcl-XL, and prevented intraneuronal accumulation and secretion of Abeta42. These results open up interesting perspectives on the development of novel strategies targeting out NF-kappaB p50/p65 dimers for pharmacological intervention in AD.


Asunto(s)
Péptidos beta-Amiloides/biosíntesis , FN-kappa B/fisiología , Neuronas/patología , Fragmentos de Péptidos/biosíntesis , Péptidos beta-Amiloides/toxicidad , Animales , Apoptosis , Línea Celular Tumoral , Células Cultivadas , Cerebelo/citología , Humanos , FN-kappa B/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/toxicidad , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Factor de Transcripción ReIA/metabolismo
16.
Mol Cell Neurosci ; 25(2): 301-11, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15019946

RESUMEN

We investigated the effects of IL-6 and a chimeric derivative of IL-6 and soluble IL-6 receptor (IL6RIL6 chimera) on excitotoxic injury in rat organotypic hippocampal slices. Brief application of N-methyl-d-aspartate (NMDA) induced astrocyte reactivity, neuron cell death, and oligodendrocyte degeneration, the latter caused by secondary activation of AMPA/kainate receptors. Both these cytokines rescued neurons and oligodendrocytes, albeit the chimeric compound was much more potent and efficient than IL-6. No change was produced on reactive astrocytosis. The cytokines preserved myelin basic protein (MBP) production in slices exposed to excitotoxic insult, and when applied singularly for a week, they also enhanced both MBP and proteolipid protein expression. These effects occurred through activating the signal transducer gp130 and were associated with stimulation of transcription factors STAT1 and STAT3. Our results suggest that IL-6 and IL6RIL6 may prove to be valuable in treating neurodegenerative and demyelinating diseases.


Asunto(s)
Interleucina-6/farmacología , Degeneración Nerviosa/prevención & control , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Proteínas Recombinantes de Fusión/farmacología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/genética , Animales , Antígenos CD/metabolismo , Astrocitos/efectos de los fármacos , Astrocitos/fisiología , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatología , Receptor gp130 de Citocinas , Proteínas de Unión al ADN/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Gliosis/fisiopatología , Gliosis/prevención & control , Hipocampo/citología , Técnicas In Vitro , Interleucina-6/genética , Glicoproteínas de Membrana/metabolismo , Proteína Básica de Mielina/genética , Proteína Proteolipídica de la Mielina/genética , N-Metilaspartato/antagonistas & inhibidores , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/metabolismo , Neuronas/metabolismo , Neurotoxinas/antagonistas & inhibidores , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Fosforilación/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Receptores de Interleucina-6/genética , Proteínas Recombinantes de Fusión/genética , Factor de Transcripción STAT1 , Factor de Transcripción STAT3 , Transactivadores/efectos de los fármacos , Transactivadores/metabolismo
17.
Eur J Neurosci ; 16(12): 2342-50, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12492429

RESUMEN

The present study demonstrates that human SK-N-SH neuroblastoma cells, differentiated by retinoic acid (RA), express functional NMDA receptors and become vulnerable to glutamate toxicity. During exposure to RA, SK-N-SH cells switched from non-neuronal to neuronal phenotype by showing antigenic changes typical of postmitotic neurons together with markers specific for cholinergic cells. Neuronally differentiated cells displayed positive immunoreactivity to the vesicular acetylcholine transporter and active acetylcholine release in response to depolarizing stimuli. The differentiation correlated with the expression of NMDA receptors. RT-PCR and immunoblotting analysis identified NMDA receptor subunits NR1 and NR2B, in RA-differentiated cultures. The NR1 protein immunolocalized to the neuronal cell population and assembled with the NR2B subunit to form functional N-methyl-D-aspartate (NMDA) receptors. Glutamate or NMDA application, concentration-dependently increased the intracellular Ca2+ levels and acetylcholine release in differentiated cultures, but not in undifferentiated SK-N-SH cells. Moreover, differentiated cultures became vulnerable to NMDA receptor-mediated excitotoxicity. The glutamate effects were enhanced by glycine application and were prevented by the NMDA receptor blocker MK 801, as well as by the NR2B selective antagonist ifenprodil. These data suggest that SK-N-SH cells differentiated by brief treatment with RA may represent an unlimited source of neuron-like cells suitable for studying molecular events associated with activation of human NR1/NR2B receptors.


Asunto(s)
Diferenciación Celular/fisiología , Ácido Glutámico/metabolismo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transmisión Sináptica/fisiología , Células Tumorales Cultivadas/metabolismo , Acetilcolina/metabolismo , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Diferenciación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Ácido Glutámico/farmacología , Glicina/metabolismo , Glicina/farmacología , Humanos , Inmunohistoquímica , Deficiencia de Magnesio/metabolismo , Neuroblastoma , Neuronas/citología , Neuronas/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Tretinoina/farmacología , Células Tumorales Cultivadas/citología , Células Tumorales Cultivadas/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...