Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Biochim Biophys Acta ; 1808(1): 236-43, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21036142

RESUMEN

The fluidity of neuronal membranes plays a pivotal role in brain aging and neurodegeneration. In this study, we investigated the role of the omega-3 fatty acid docosahexaenoic acid (DHA) in modulation of membrane fluidity, APP processing, and protection from cytotoxic stress. To this end, we applied unilamellar transfer liposomes, which provided protection from oxidation and effective incorporation of DHA into cell membranes. Liposomes transferring docosanoic acid (DA), the completely saturated form of DHA, to the cell cultures served as controls. In HEK-APP cells, DHA significantly increased membrane fluidity and non-amyloidogenic processing of APP, leading to enhanced secretion of sAPPα. This enhanced secretion of sAPPα was associated with substantial protection against apoptosis induced by ER Ca(2+) store depletion. sAPPα-containing supernatants obtained from HEK-APP cells exerted similar protective effects as DHA in neuronal PC12 cells and HEK293 control cells. Correlating to further increased sAPPα levels, supernatants obtained from DHA-treated HEK-APP cells enhanced protection, whereas supernatants obtained from DHA-treated HEK293 control cells did not inhibit apoptosis, likely due to the low expression of endogenous APP and negligible sAPPα secretion in these cells. Further experiments with the small molecule inhibitors LY294002 and SP600125 indicated that sAPPα-induced cytoprotection relied on activation of the anti-apoptotic PI3K/Akt pathway and inhibition of the stress-triggered JNK signaling pathway in PC12 cells. Our data suggest that liposomal DHA is able to restore or maintain physiological membrane properties, which are required for neuroprotective sAPPα secretion and autocrine modulation of neuronal survival.


Asunto(s)
Precursor de Proteína beta-Amiloide/química , Ácidos Docosahexaenoicos/química , Liposomas/química , Neuronas/metabolismo , Animales , Calcio/química , Caspasa 3/química , Supervivencia Celular , Retículo Endoplásmico/metabolismo , Humanos , Peroxidación de Lípido , Liposomas/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Potenciales de la Membrana , Oxígeno/química , Ratas
2.
Hepatology ; 51(6): 2183-92, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20513003

RESUMEN

UNLABELLED: Hypoxia-inducible factors (HIFs) provoke adaptation to hypoxic stress occurring in rapidly growing tumor tissues. Therefore, overexpression of HIF-1 or HIF-2 is a common feature in hepatocellular carcinoma but their specific function is still controversially discussed. To analyze HIF function in hypoxia-induced cell death we created a stable knockdown of HIF-1alpha and HIF-2alpha in HepG2 cells and generated tumor spheroids as an in vitro hepatocellular carcinoma model. Knockdown of HIF-1alpha enhanced expression of HIF-2alpha and vice versa. Unexpectedly, knockdown of HIF-1alpha or HIF-2alpha increased cell viability as well as spheroid size and decreased caspase-3 activity. Antiapoptotic Bcl-X(L) expression increased in both knockdown spheroids, whereas proapoptotic Bax was only reduced in HIF-1alpha-knockdown cells. Furthermore, an HIF-2alpha-knockdown significantly increased Bcl-2/adenovirus E1B 19 kDa-interacting protein 3 (BNIP3) expression in an HIF-1alpha-dependent manner. Concomitantly, electron microscopy revealed a substantial increase in autophagosomal structures in HIF-2alpha-knockdown spheroids and mito-/lysotracker costaining confirmed lysosomal activity of these autophagosomes. Blocking autophagosome maturation using 3-methyladenine restored cell death in HIF-2alpha-knockdown clones comparable to wildtype cells. CONCLUSION: An HIF-1alpha-knockdown increases HIF-2alpha expression and shifts the balance of Bcl-2 family members toward survival. The knockdown of HIF-2alpha raises autophagic activity and attenuates apoptosis by enhancing HIF-1alpha expression. Our data indicate that enhanced expression of one HIF-isoform causes a survival advantage in hepatocellular carcinoma development.


Asunto(s)
Apoptosis , Autofagia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinoma Hepatocelular/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Hepáticas/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Caspasa 3/metabolismo , Aumento de la Célula , Supervivencia Celular , Técnicas de Silenciamiento del Gen , Células Hep G2 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Esferoides Celulares/metabolismo , Células Tumorales Cultivadas , Regulación hacia Arriba , Proteína X Asociada a bcl-2/metabolismo , Proteína bcl-X/metabolismo
3.
Mol Cell Neurosci ; 44(4): 386-93, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20472066

RESUMEN

Impaired proteasomal function is a major hallmark in the pathophysiology of neurodegenerative diseases, including Alzheimer's disease (AD). Here we investigated the biological properties of the secreted cleavage product of APP (sAPPalpha) in antagonizing stress signalling, dendritic degeneration and neuronal cell death induced by the proteasome inhibitor epoxomicin. Analysis of executioner caspase activation demonstrated that sAPPalpha was able to protect PC12 cells from apoptosis triggered by epoxomicin, as well as by genotoxic stress (UV irradiation). This anti-apoptotic effect of sAPPalpha was associated with inhibition of the stress-triggered c-Jun N-terminal kinase (JNK)-signalling pathway. The anti-apoptotic effect of sAPPalpha could also be confirmed in organotypic slice cultures of Thy1-GFP mouse hippocampi. Confocal time-lapse imaging of CA1 pyramidal neurons revealed that preincubation with sAPPalpha preserves the structural integrity of neurons after epoxomicin treatment. Taken together, our data demonstrate that sAPPalpha is neuroprotective under conditions of proteasomal stress.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Apoptosis , Región CA1 Hipocampal/citología , Espinas Dendríticas/patología , Degeneración Nerviosa , Complejo de la Endopetidasa Proteasomal/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Apoptosis/fisiología , Apoptosis/efectos de la radiación , Células Cultivadas , Cromonas/farmacología , Medios de Cultivo Condicionados/química , Técnicas de Cultivo , Inhibidores Enzimáticos/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Ratones , Microscopía Confocal , Morfolinas/farmacología , Oligopéptidos/farmacología , Células PC12 , Inhibidores de las Quinasa Fosfoinosítidos-3 , Ratas , Proteínas Recombinantes de Fusión , Antígenos Thy-1 , Rayos Ultravioleta
4.
J Neurosci ; 29(4): 1017-33, 2009 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-19176811

RESUMEN

The spine apparatus is an essential component of dendritic spines of cortical and hippocampal neurons, yet its functions are still enigmatic. Synaptopodin (SP), an actin-binding protein, is tightly associated with the spine apparatus and it may play a role in synaptic plasticity, but it has not yet been linked mechanistically to synaptic functions. We studied endogenous and transfected SP in dendritic spines of cultured hippocampal neurons and found that spines containing SP generate larger responses to flash photolysis of caged glutamate than SP-negative ones. An NMDA-receptor-mediated chemical long-term potentiation caused the accumulation of GFP-GluR1 in spine heads of control but not of shRNA-transfected, SP-deficient neurons. SP is linked to calcium stores, because their pharmacological blockade eliminated SP-related enhancement of glutamate responses, and release of calcium from stores produced an SP-dependent increase of GluR1 in spines. Thus, SP plays a crucial role in the calcium store-associated ability of neurons to undergo long-term plasticity.


Asunto(s)
Espinas Dendríticas/fisiología , Hipocampo/citología , Proteínas de Microfilamentos/fisiología , Plasticidad Neuronal/fisiología , Neuronas/citología , Acetaminofén/farmacología , Actinas/metabolismo , Animales , Animales Recién Nacidos , Antipirina/análogos & derivados , Antipirina/farmacología , Cafeína/farmacología , Calcio/metabolismo , Células Cultivadas , Codeína/farmacología , Espinas Dendríticas/efectos de los fármacos , Combinación de Medicamentos , Estimulación Eléctrica/métodos , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Proteínas Fluorescentes Verdes/genética , Proteínas de Microfilamentos/genética , Modelos Biológicos , Plasticidad Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp/métodos , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , ARN Interferente Pequeño/farmacología , Ratas , Receptores AMPA/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Sinaptofisina/metabolismo , Transfección/métodos
5.
Biochim Biophys Acta ; 1788(5): 964-72, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19366591

RESUMEN

Gradual changes in steady-state levels of beta amyloid peptides (Abeta) in brain are considered an initial step in the amyloid cascade hypothesis of Alzheimer's disease. Abeta is a product of the secretase cleavage of amyloid precursor protein (APP). There is evidence that the membrane lipid environment may modulate secretase activity and alters its function. Cleavage of APP strongly depends on membrane properties. Since Abeta perturbs cell membrane fluidity, the cell membrane may be the location where the neurotoxic cascade of Abeta is initiated. Therefore, we tested effects of oligomeric Abeta on membrane fluidity of whole living cells, the impact of exogenous and cellular Abeta on the processing of APP and the role of GM-1 ganglioside. We present evidence that oligoAbeta((1-40)) stimulates the amyloidogenic processing of APP by reducing membrane fluidity and complexing with GM-1 ganglioside. This dynamic action of Abeta may start a vicious circle, where endogenous Abeta stimulates its own production. Based on our novel findings, we propose that oligoAbeta((1-40)) accelerates the proteolytic cleavage of APP by decreasing membrane fluidity.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Membrana Celular/metabolismo , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/biosíntesis , Péptidos beta-Amiloides/química , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Fenómenos Biofísicos , Línea Celular , Colesterol/metabolismo , Gangliósido G(M1)/metabolismo , Humanos , Fluidez de la Membrana , Lípidos de la Membrana/metabolismo , Ratones , Microscopía Confocal , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Procesamiento Proteico-Postraduccional , Estructura Cuaternaria de Proteína
6.
Neurosci Lett ; 453(1): 41-4, 2009 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-19429012

RESUMEN

Alzheimer's disease (AD) is characterized by the pathological deposition of amyloid-beta protein in the aged brain. Inefficient clearance of amyloid-beta from brain tissue is believed to play a major role in the pathogenesis of these deposits. Since amyloid-beta clearance likely involves activation of microglial cells via toll-like receptors and since these receptors and their signaling pathways are regarded as potential therapeutic targets, we have studied the expression of toll-like receptor (tlr) mRNAs in an animal model of AD (APP23 transgenic mice). Laser microdissection was used to harvest plaques, tissue surrounding plaques and plaque-free tissue from cortex of aged APP23 transgenic mice and age-matched controls. Real-time RT-PCR was employed to quantify expression levels of different tlr mRNAs in these tissues. This revealed a strong upregulation of tlr2, tlr4, tlr5, tlr7 and tlr9 mRNAs in plaque material compared to plaque-free tissue. In contrast, tlr3 was not significantly upregulated. Plaque-free tissue did not show an increased expression of any tlr mRNAs compared to age-matched control mice. Double-immunofluorescence for TLR2 and the microglial marker Iba1 was used to demonstrate localization of TLR2 on plaque-associated microglia. Taken together, these data show a strong upregulation of mRNAs encoding surface TLRs in plaque-associated brain tissue of aged APP23 transgenic mice. Since TLR-upregulation is restricted to plaques, modifying TLR-signaling may be a promising therapeutic strategy for plaque removal.


Asunto(s)
Envejecimiento , Enfermedad de Alzheimer/metabolismo , Corteza Cerebral/metabolismo , Placa Amiloide/metabolismo , ARN Mensajero/metabolismo , Receptores Toll-Like/metabolismo , Animales , Proteínas de Unión al Calcio/metabolismo , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Transgénicos , Proteínas de Microfilamentos , Microglía/fisiología , Placa Amiloide/genética , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 9/metabolismo , Receptores Toll-Like/genética , Regulación hacia Arriba
7.
Biochim Biophys Acta ; 1773(2): 157-65, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17113167

RESUMEN

Here we investigated the role of the amyloid precursor protein (APP) in regulation of Ca(2+) store depletion-induced neural cell death. Ca(2+) store depletion from the endoplasmic reticulum (ER) was induced by the SERCA (Sarco/Endoplasmic Reticulum Calcium ATPase) inhibitor thapsigargin which led to a rapid induction of the unfolded protein response (UPR) and a delayed activation of executioner caspases in the cultures. Overexpression of APP potently enhanced cytosolic Ca(2+) levels and cell death after ER Ca(2+) store depletion in comparison to vector-transfected controls. GeneChip and RT-PCR analysis revealed that the expression of classical UPR chaperone genes was not altered by overexpression of APP. Interestingly, the induction of the ER stress-responsive pro-apoptotic transcription factor CHOP was significantly upregulated in APP-overexpressing cells in comparison to vector-transfected controls. Chelation of intracellular Ca(2+) with BAPTA-AM revealed that enhanced CHOP expression after store depletion occurred in a Ca(2+)-dependent manner in APP-overexpressing cells. Prevention of CHOP induction by BAPTA-AM and by RNA interference was also able to abrogate the potentiating effect of APP on thapsigargin-induced apoptosis. Application of the store-operated channel (SOC)-inhibitors SK & F96365 and 2-APB downmodulated APP-triggered potentiation of cytosolic Ca(2+) levels and apoptosis after treatment with thapsigargin. Our data demonstrate that APP significantly modulates Ca(2+) store depletion-induced cell death in a SOC- and CHOP-dependent manner, but independent of the UPR.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Calcio/deficiencia , Retículo Endoplásmico/metabolismo , Regulación de la Expresión Génica , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , Animales , Tampones (Química) , Canales de Calcio/metabolismo , Muerte Celular/efectos de los fármacos , Quelantes/farmacología , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Retículo Endoplásmico/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Activación del Canal Iónico/efectos de los fármacos , Células PC12 , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Tapsigargina/farmacología , Factor de Transcripción CHOP/deficiencia , Transcripción Genética/efectos de los fármacos
8.
Neuro Oncol ; 15(7): 840-52, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23486688

RESUMEN

BACKGROUND: Diffuse infiltration remains the fulcrum of glioblastoma's incurability, leading inevitably to recurrence. Therefore, uncovering the pathological mechanism is imperative. Because signal transducer and activator of transcription 3 (STAT3) correlates with glioma malignancy and predicts poor clinical outcome, we determined its role in glioma single cell infiltration and tumor growth. METHODS: STAT3 was silenced in Tu-2449 glioma cells via lentiviral gene transfer. Target gene expression was measured by real-time reverse transcription PCR, Western blotting, and immunohistochemistry. Microvilli were visualized by staining with wheat germ agglutinin. Migration and invasion were measured by Scratch and Matrigel chamber assays. Diffuse infiltration was studied in 350-µm-thick organotypic tissue cultures over 14 days using cells tagged with enhanced green fluorescent protein and live confocal laser scanning microscopy. Survival of tumor-bearing syngeneic, immunocompetent B6C3F1 mice was analyzed by Kaplan-Meier plots. RESULTS: STAT3 silencing reduced cell migration and invasion in vitro and stopped single cell infiltration ex vivo, while STAT3-expressing cells disseminated through the neuropil at ∼100 µm/day. STAT3 silencing reduced transcription of several tumor progression genes. Mice with intracranial STAT3 knockdown tumors had a significant (P< .0007) survival advantage over controls, yielding 27% long-term survival. STAT3 knockdown reduced podoplanin expression 50-fold and inhibited concurrent microvilli formation. STAT3 knockdown tumors exhibited a weaker podoplanin immunoreactivity compared with controls. Podoplanin staining was diffuse, preferentially at tumor margins, and absent in normal brain. CONCLUSIONS: Our results show compelling evidence that STAT3 is a key driver of diffuse infiltration and glioma growth and might therefore represent a promising target for an anti-invasive therapy.


Asunto(s)
Neoplasias Encefálicas/prevención & control , Proliferación Celular , Glioma/prevención & control , ARN Interferente Pequeño/genética , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Apoptosis , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Glioma/patología , Técnicas para Inmunoenzimas , Ratones , Microscopía Confocal , Técnicas de Cultivo de Órganos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Células Tumorales Cultivadas , Aglutininas del Germen de Trigo/metabolismo
9.
J Neuropathol Exp Neurol ; 69(2): 168-75, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20084017

RESUMEN

Vascular endothelial growth factor receptor 1 (VEGFR-1) is highly expressed in endothelial cells and regulates developmental angiogenesis by acting as a decoy receptor and trapping VEGF-A. Vascular endothelial growth factor receptor 1 is also expressed in monocytes and macrophages; mice lacking the VEGFR-1 tyrosine kinase (TK) domain (VEGFR-1 TK mice) display impaired macrophage function. Because macrophages are recruited to sites of cerebral ischemic infarcts, we hypothesized that lack of VEGFR-1 TK in bone marrow(BM) cells would affect the outcome in an experimental stroke model. We performed BM transplantation experiments in C57BL/6J mice using VEGFR-1 TK and VEGFR-1 TK mice as BM donors and analyzed cell infiltration after cerebral ischemia. There was reduced initial recruitment of VEGFR-1 TK myeloid cells into the infarcted tissue and reduced postischemic angiogenesis at 3days postischemia. By 10 days, the numbers of infiltrating cells and the densities of vessels in the infarct peri-infarct zone were similar for both groups. Neither infarct size at 3 and 10 days postischemia nor neurological performance at 24 hours was different between the experimental groups. These results support a role of VEGFR-1 signaling in the early regulation of BM infiltration and angiogenesis after brain ischemia.


Asunto(s)
Trasplante de Médula Ósea , Infarto Cerebral/cirugía , Macrófagos , Microglía , Transducción de Señal , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Células de la Médula Ósea/citología , Isquemia Encefálica/complicaciones , Diferenciación Celular , Células Cultivadas , Infarto Cerebral/etiología , Infarto Cerebral/metabolismo , Infarto Cerebral/patología , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptor 1 de Factores de Crecimiento Endotelial Vascular/deficiencia
10.
J Neurochem ; 99(3): 952-64, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16942595

RESUMEN

Oxygen and glucose deprivation are direct consequences of tissue ischaemia. We explored the interaction of hypoxia and hypoglycaemia on cell survival and gene expression in the absence of glutamatergic signalling using human SH-SY5Y neuroblastoma cells as a model. In agreement with previous investigations in non-neural cells, prolonged hypoxia (0.5% O(2)) failed to induce significant cell death in this system. In contrast, exposure to hypoglycaemia induced significant necrotic cell death (> 80% after 72 h). Interestingly, hypoglycaemia-induced cell death was completely abrogated by simultaneous exposure to hypoxia, suggesting strong cytoprotective effects of hypoxia. Subsequent microarray analysis of the underlying transcriptional responses revealed that the transcription factor CEBP homology protein (CHOP) was strongly induced by hypoglycaemia, and suppressed by simultaneous hypoxia. RNA interference against CHOP significantly protected cells from glucose deprivation-induced cell death. Hypoxia-induced vascular endothelial growth factor (VEGF) activation also protected cells against hypoglycaemia-induced cell death, but VEGF failed to modify hypoglycaemia-induced CHOP induction. Our data suggest that hypoglycaemia-induced necrotic cell death of neuroblastoma cells is an active process mediated via the induction of the transcription factor CHOP, and that hypoxia counteracts this cell death via at least two distinct mechanisms: repression of CHOP and induction of VEGF.


Asunto(s)
Neoplasias Encefálicas/patología , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Hipoglucemia/metabolismo , Neuroblastoma/patología , Apoptosis/genética , Apoptosis/fisiología , Western Blotting , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/fisiología , Caspasa 3/metabolismo , Muerte Celular , Línea Celular Tumoral , Colorantes Fluorescentes , Glucosa/deficiencia , Humanos , Hipoxia/patología , Necrosis , Análisis de Secuencia por Matrices de Oligonucleótidos , Transducción de Señal/fisiología , Transfección , Factor A de Crecimiento Endotelial Vascular/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA