Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Psychiatry ; 20(1): 109-17, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25349165

RESUMEN

Certain mutant Alzheimer's amyloid-ß (Aß) peptides (that is, Dutch mutant APP(E693Q)) form complexes with gangliosides (GAß). These mutant Aß peptides may also undergo accelerated aggregation and accumulation upon exposure to GM2 and GM3. We hypothesized that increasing ß-hexosaminidase (ß-hex) activity would lead to a reduction in GM2 levels, which in turn, would cause a reduction in Aß aggregation and accumulation. The small molecule OT1001 is a ß-hex-targeted pharmacological chaperone with good bioavailability, blood-brain barrier penetration, high selectivity for ß-hex and low cytotoxicity. Dutch APP(E693Q) transgenic mice accumulate oligomeric Aß as they age, as well as Aß oligomer-dose-dependent anxiety and impaired novel object recognition (NOR). Treatment of Dutch APP(E693Q) mice with OT1001 caused a dose-dependent increase in brain ß-hex levels up to threefold over those observed at baseline. OT1001 treatment was associated with reduced anxiety, improved learning behavior in the NOR task and dramatically reduced GAß accumulation in the subiculum and perirhinal cortex, both of which are brain regions required for normal NOR. Pharmacological chaperones that increase ß-hex activity may be useful in reducing accumulation of certain mutant species of Aß and in preventing the associated behavioral pathology.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Antipsicóticos/uso terapéutico , Trastornos del Conocimiento , Gangliósidos/metabolismo , beta-N-Acetilhexosaminidasas/metabolismo , Enfermedad de Alzheimer/genética , Animales , Barrera Hematotesticular/efectos de los fármacos , Células Cultivadas , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Gangliósidos/uso terapéutico , Humanos , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Reconocimiento en Psicología/efectos de los fármacos , Factores de Tiempo
2.
Curr Alzheimer Res ; 8(1): 4-23, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21143159

RESUMEN

At diagnosis, Alzheimer's disease (AD) brains are extensively burdened with plaques and tangles and display a degree of synaptic failure most likely beyond therapeutic treatment. It is therefore crucial to identify early pathological events in the progression of the disease. While it is not currently feasible to identify and study early, pre-clinical stages of AD, transgenic (Tg) models offer a valuable tool in this regard. Here we investigated cognitive, structural and biochemical CNS alterations occurring in our newly developed McGill-Thyl-APP Tg mice (over-expressing the human amyloid precursor protein with the Swedish and Indiana mutations) prior to extracellular plaque deposition. Pre-plaque, 3-month old Tg mice already displayed cognitive deficits concomitant with reorganization of cortical cholinergic pre-synaptic terminals. Conformational specific antibodies revealed the early appearance of intracellular amyloid ß (Aß)-oligomers and fibrillar oligomers in pyramidal neurons of cerebral cortex and hippocampus. At the same age, the cortical levels of insulin degrading enzyme -a well established Aß-peptidase, were found to be significantly down-regulated. Our results suggest that, in the McGill-Thy1-APP Tg model, functional, structural and biochemical alterations are already present in the CNS at early, pre-plaque stages of the pathology. Accumulation of intraneuronal neurotoxic Aß-oligomers (possibly caused by a failure in the clearance machinery) is likely to be the culprit of such early, pre-plaque pathology. Similar neuronal alterations might occur prior to clinical diagnosis in AD, during a yet undefined 'latent' stage. A better understanding of such pre-clinical AD might yield novel therapeutic targets and or diagnostic tools.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Modelos Animales de Enfermedad , Factores de Edad , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Análisis de Varianza , Animales , Cerebelo/metabolismo , Corteza Cerebral/metabolismo , Trastornos del Conocimiento/etiología , Evaluación Preclínica de Medicamentos , Regulación de la Expresión Génica/genética , Hipocampo/metabolismo , Humanos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Mutación/genética , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Fragmentos de Péptidos/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Reconocimiento en Psicología/fisiología , Proteínas de Transporte Vesicular de Acetilcolina/metabolismo
3.
J Nutr Health Aging ; 12(1): 51S-7S, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18165846

RESUMEN

Individuals with early-stage Alzheimer's disease (AD) suffer from profound failure to form new memories. A novel molecular mechanism with implications for therapeutics and diagnostics is now emerging in which the specificity of AD for memory derives from disruption of plasticity at synapses targeted by neurologically active A beta oligomers (1). We have named these oligomers "ADDLs" (for pathogenic A beta-Derived Diffusible Ligands). ADDLs constitute metastable alternatives to the disease-defining A beta fibrils deposited in amyloid plaques. In AD brain, ADDLs accumulate primarily as A beta 12mers (2) (approximately 54 kDa) and can be found in dot-like clusters distinct from senile plaques (3). Oligomers of equal mass have been reported to occur in tgmouse AD models where they emerge concomitantly with memory failure (4), consistent with ADDL inhibition of LTP (1). In cell biology studies, ADDLs act as pathogenic gain-of-function ligands that target particular synapses, binding to synaptic spines at or near NMDA receptors (5,6). Binding produces ectopic expression of the memory-linked immediate early gene Arc. Subsequent ADDL-induced abnormalities in spine morphology and synaptic receptor composition (7) are predicted consequences of Arc overexpression, a pathology associated with memory dysfunction in tg-Arc mice. Significantly, the attack on synapses provides a plausible mechanism unifying memory dysfunction with major features of AD neuropathology; recent findings show that ADDL binding instigates synapse loss, oxidative damage, and AD-type tau hyperphosphorylation. Acting as novel neurotoxins that putatively account for memory loss and neuropathology, ADDLs present significant targets for disease-modifying therapeutics in AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Memoria , Vías Nerviosas/patología , Sinapsis/patología , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/toxicidad , Química Encefálica , Humanos , Ligandos , Memoria/fisiología , Neuronas/patología
4.
Neurobiol Aging ; 25(5): 569-80, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15172732

RESUMEN

Pioneering work in the 1950s by Christian Anfinsen on the folding of ribonuclease has shown that the primary structure of a protein "encodes" all of the information necessary for a nascent polypeptide to fold into its native, physiologically active, three-dimensional conformation (for his classic review, see [Science 181 (1973) 223]). In Alzheimer's disease (AD), the amyloid beta-protein (Abeta) appears to play a seminal role in neuronal injury and death. Recent data have suggested that the proximate effectors of neurotoxicity are oligomeric Abeta assemblies. A fundamental question, of relevance both to the development of therapeutic strategies for AD and to understanding basic laws of protein folding, is how Abeta assembly state correlates with biological activity. Evidence suggests, as argued by Anfinsen, that the formation of toxic Abeta structures is an intrinsic feature of the peptide's amino acid sequence-one requiring no post-translational modification or invocation of peptide-associated enzymatic activity.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Neurotoxinas/toxicidad , Pliegue de Proteína , Secuencia de Aminoácidos , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/toxicidad , Animales , Proteínas de Unión a Calmodulina/metabolismo , Humanos , Metionina/metabolismo , Neurotoxinas/química , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/toxicidad , Procesamiento Proteico-Postraduccional/fisiología
5.
J Neurochem ; 79(3): 595-605, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11701763

RESUMEN

In recent studies of transgenic models of Alzheimer's disease (AD), it has been reported that antibodies to aged beta amyloid peptide 1-42 (Abeta(1-42)) solutions (mixtures of Abeta monomers, oligomers and amyloid fibrils) cause conspicuous reduction of amyloid plaques and neurological improvement. In some cases, however, neurological improvement has been independent of obvious plaque reduction, and it has been suggested that immunization might neutralize soluble, non-fibrillar forms of Abeta. It is now known that Abeta toxicity resides not only in fibrils, but also in soluble protofibrils and oligomers. The current study has investigated the immune response to low doses of Abeta(1-42) oligomers and the characteristics of the antibodies they induce. Rabbits that were injected with Abeta(1-42) solutions containing only monomers and oligomers produced antibodies that preferentially bound to assembled forms of Abeta in immunoblots and in physiological solutions. The antibodies have proven useful for assays that can detect inhibitors of oligomer formation, for immunofluorescence localization of cell-attached oligomers to receptor-like puncta, and for immunoblots that show the presence of SDS-stable oligomers in Alzheimer's brain tissue. The antibodies, moreover, were found to neutralize the toxicity of soluble oligomers in cell culture. Results support the hypothesis that immunizations of transgenic mice derive therapeutic benefit from the immuno-neutralization of soluble Abeta-derived toxins. Analogous immuno-neutralization of oligomers in humans may be a key in AD vaccines.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/inmunología , Especificidad de Anticuerpos , Fragmentos de Péptidos/inmunología , Vacunación , Péptidos beta-Amiloides/toxicidad , Animales , Epítopos , Técnica del Anticuerpo Fluorescente , Hipocampo/citología , Humanos , Neuronas/citología , Neuronas/inmunología , Fármacos Neuroprotectores/inmunología , Células PC12 , Fragmentos de Péptidos/toxicidad , Ratas , Solubilidad
6.
Trends Neurosci ; 24(4): 219-24, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11250006

RESUMEN

Amyloid beta (Abeta) is a small self-aggregating peptide produced at low levels by normal brain metabolism. In Alzheimer's disease (AD), self-aggregation of Abeta becomes rampant, manifested most strikingly as the amyloid fibrils of senile plaques. Because fibrils can kill neurons in culture, it has been argued that fibrils initiate the neurodegenerative cascades of AD. An emerging and different view, however, is that fibrils are not the only toxic form of Abeta, and perhaps not the neurotoxin that is most relevant to AD: small oligomers and protofibrils also have potent neurological activity. Immuno-neutralization of soluble Abeta-derived toxins might be the key to optimizing AD vaccines that are now on the horizon.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Transducción de Señal/fisiología , Sinaptofisina/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/efectos de los fármacos , Péptidos beta-Amiloides/farmacología , Precursor de Proteína beta-Amiloide/efectos de los fármacos , Animales , Anticuerpos/farmacología , Anticuerpos/uso terapéutico , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Ratones , Ratones Transgénicos , Fragmentos de Péptidos/efectos de los fármacos , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Placa Amiloide/efectos de los fármacos , Placa Amiloide/metabolismo , Transducción de Señal/efectos de los fármacos , Vacunas/farmacología , Vacunas/uso terapéutico
7.
J Neurochem ; 75(5): 1977-85, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11032887

RESUMEN

The activity of the superoxide-sensitive enzyme aconitase was monitored to evaluate the generation of superoxide in neuronal cell lines treated with beta-amyloid (Abeta) peptide 1-42. Treatment of differentiated and undifferentiated rat PC12 and human neuroblastoma SK-N-SH cells with soluble Abeta1-42 (Abeta-derived diffusible ligands) or fibrillar Abeta1-42 caused a 35% reversible inactivation of aconitase, which preceded loss of viability and was correlated with altered cellular function. Aconitase was reactivated upon incubation of cellular extracts with iron and sulfur, suggesting that Abeta causes the release of iron from 4Fe-4S clusters. Abeta neurotoxicity was partially blocked by the iron chelator deferoxamine. These data suggest that increased superoxide generation and the release of iron from 4Fe-4S clusters are early events in Abeta1-42 neurotoxicity.


Asunto(s)
Aconitato Hidratasa/metabolismo , Péptidos beta-Amiloides/metabolismo , Chaperonas Moleculares , Neuronas/enzimología , Fragmentos de Péptidos/metabolismo , Superóxidos/metabolismo , Aconitato Hidratasa/antagonistas & inhibidores , Péptidos beta-Amiloides/toxicidad , Animales , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Clusterina , Relación Dosis-Respuesta a Droga , Reactivadores Enzimáticos/farmacología , Glicoproteínas/metabolismo , Humanos , Líquido Intracelular/metabolismo , Hierro/farmacología , Quelantes del Hierro/farmacología , Ligandos , Sustancias Macromoleculares , Neuroblastoma , Neuronas/citología , Neuronas/efectos de los fármacos , Óxido Nítrico/biosíntesis , Células PC12 , Fragmentos de Péptidos/toxicidad , Ratas , Compuestos de Azufre/farmacología , Células Tumorales Cultivadas
8.
Proc Natl Acad Sci U S A ; 95(11): 6448-53, 1998 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-9600986

RESUMEN

Abeta1-42 is a self-associating peptide whose neurotoxic derivatives are thought to play a role in Alzheimer's pathogenesis. Neurotoxicity of amyloid beta protein (Abeta) has been attributed to its fibrillar forms, but experiments presented here characterize neurotoxins that assemble when fibril formation is inhibited. These neurotoxins comprise small diffusible Abeta oligomers (referred to as ADDLs, for Abeta-derived diffusible ligands), which were found to kill mature neurons in organotypic central nervous system cultures at nanomolar concentrations. At cell surfaces, ADDLs bound to trypsin-sensitive sites and surface-derived tryptic peptides blocked binding and afforded neuroprotection. Germ-line knockout of Fyn, a protein tyrosine kinase linked to apoptosis and elevated in Alzheimer's disease, also was neuroprotective. Remarkably, neurological dysfunction evoked by ADDLs occurred well in advance of cellular degeneration. Without lag, and despite retention of evoked action potentials, ADDLs inhibited hippocampal long-term potentiation, indicating an immediate impact on signal transduction. We hypothesize that impaired synaptic plasticity and associated memory dysfunction during early stage Alzheimer's disease and severe cellular degeneration and dementia during end stage could be caused by the biphasic impact of Abeta-derived diffusible ligands acting upon particular neural signal transduction pathways.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Neurotoxinas/toxicidad , Enfermedad de Alzheimer , Péptidos beta-Amiloides/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células Cultivadas , Ligandos , Ratones
9.
J Neurosci Res ; 50(6): 979-89, 1997 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-9452012

RESUMEN

Beta-amyloid1-42 (Abeta) is a naturally occuring peptide whose accumulation in the brain is putatively coupled to Alzheimer's disease pathogenesis. Deleterious effects of Abeta on neurons have been linked to the inappropriate activation of signaling pathways within the cell (reviewed in Yankner, 1996), including tyrosine phosphorylation of focal adhesion kinase (FAK) (Zhang et al., 1994, 1996a,b). Here we have investigated the effects of Abeta on paxillin in a neural cell line. Paxillin, a substrate for FAK, is thought to act as a signal "integrator," functioning to link other proteins into multi-molecular signaling complexes (reviewed in Turner, 1994). Treatment of the rat central nervous system B103 cell line with aggregates of Abeta was found to induce the tyrosine phosphorylation of paxillin within 30 min, nearly 24 hr prior to significant cell death. Particularly striking was a subsequent "mobilization" of paxillin to the cytoskeleton in Abeta-treated cells. The amount of paxillin associated with the cytoskeleton in Abeta-treated cells was increased 10-fold over controls. The Abeta-induced paxillin accumulation could be visualized immunocytochemically, with an increase in number and size of paxillin-labeled focal contacts upon treatment with Abeta. This effect was specific, in that vinculin, another focal contact protein, was unaffected by Abeta. Disruption of f-actin, which inhibits both Abeta-induced neurotoxicity (Furukawa and Mattson, 1995) and focal contact signaling in B103 cells (Zhang et al., 1996b) was found to block the cytoskeletal paxillin accumulation. The rapid tyrosine phosphorylation and cytoskeletal mobilization of paxillin links Abeta to the activation of focal contact signaling events that may influence neuronal cytoskeletal architecture, gene expression, synaptic plasticity and cell viability.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Proteínas del Citoesqueleto/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Fosfoproteínas/metabolismo , Actinas/fisiología , Animales , Línea Celular , Citoesqueleto/metabolismo , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Paxillin , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Ratas , Factores de Tiempo
10.
J Neurosci Res ; 46(4): 445-55, 1996 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-8950704

RESUMEN

Focal adhesion kinase (FAK) is a non-receptor protein tyrosine kinase that appears to play a central role in integrin-mediated signal transduction in non-neuronal cells, linking the extracellular matrix to the actin-based cytoskeleton at focal adhesion contacts. Biochemical analysis has revealed the presence of FAK immunoreactivity in cells of neuronal lineage (Zhang et al., 1994) and in the CNS (Burgaya et al. 1995; Grant et al., 1995). In the current work, we have examined the immunodistribution of FAK in nerve cell cultures and tissue sections from the rat CNS. Cultures of B103 CNS neuroblastoma cells and primary cultures of hippocampal neurons both showed abundant FAK immunoreactivity in nerve cell bodies. Immunoreactivity also extended into neurites and growth cones. The most striking feature of FAK distribution was the presence of short, punctate clusters of high FAK concentration. These FAK clusters were maintained in triton-extracted cell ghosts, indicating association with the cytoskeleton. Double-label confocal imaging showed that clusters of FAK coincided with clusters of vinculin, another actin-associated signal transduction molecule implicated in control of growth cone motility. Data from hippocampal sections verified the presence of FAK in adult neurons where it was enriched in somato-dendritic domains and showed a non-uniform distribution. Quantitative FAK immunoprecipitation to compare adult with embryonic brain showed a 7-fold developmental down-regulation of FAK and a 21-fold down-regulation of FAK TyrP. The data suggest that neuronal FAK may participate in signal transduction complexes relevant to neuronal morphogenesis and plasticity.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Hipocampo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Vinculina/metabolismo , Factores de Edad , Animales , Transporte Biológico , Moléculas de Adhesión Celular/genética , Citoesqueleto/metabolismo , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Regulación del Desarrollo de la Expresión Génica , Hipocampo/citología , Hipocampo/embriología , Sustancias Macromoleculares , Microscopía Confocal , Proteínas del Tejido Nervioso/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Plasticidad Neuronal , Neuronas/metabolismo , Neuronas/ultraestructura , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Tirosina Quinasas/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Células Tumorales Cultivadas
11.
Neurosci Lett ; 211(3): 187-90, 1996 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-8817572

RESUMEN

Neurotoxicity of the amyloid beta protein (A beta) is known to correlate with a selective change in protein tyrosine phosphorylation (Tyr(P)) of focal adhesion kinase (FAK) (Zhang et al., J. Biol. Chem., 269 (1994) 25247-25250). The current work has found that exposure of neuronal cells to A beta upregulates the stable association of FAK with Fyn, a neuronally-enriched protein tyrosine kinase of the Src-family. In cells incubated with aged A beta 1-42, the amount of immunoprecipitable FAK-Fyn complex increased approximately 280%. Equivalent results were obtained whether anti-FAK or anti-Fyn was used to precipitate the complex. Cells incubated with non-toxic A beta 17-42, which makes aggregates and attaches to cells but does not upregulate FAK Tyr(P), exhibited no increase in FAK-Fyn complex. Aberrant Fyn activity due to the A beta evoked association with FAK could play a role in neuronal degeneration and also cause anomalies in synaptic plasticity. These possibilities are of particular significance because of the reported increase in Fyn immunoreactivity in Alzheimer's-afflicted neurons (Shirazi and Wood, NeuroReport, 4 (1993) 435-437).


Asunto(s)
Precursor de Proteína beta-Amiloide/toxicidad , Moléculas de Adhesión Celular/efectos de los fármacos , Sistema Nervioso Central/efectos de los fármacos , Proteínas Tirosina Quinasas/efectos de los fármacos , Animales , Línea Celular/efectos de los fármacos , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Fosforilación/efectos de los fármacos , Ratas
12.
FEBS Lett ; 386(2-3): 185-8, 1996 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-8647278

RESUMEN

Focal adhesion kinase (FAK) is a protein tyrosine kinase implicated in signal transduction pathways for integrins, neuropeptides, and lysophosphatidic acid. FAK, first discovered in non-neuronal cells, recently has been reported to occur in neurons, where its tyrosine phosphorylation is upregulated by fibronectin and by the Alzheimer's Abeta peptide. The current work has elucidated molecular events leading to tyrosine phosphorylation of FAK in the rat B103 CNS nerve cell line. Activation of receptor-coupled G-proteins by Mas-7 was found to evoke rapid upregulation of FAK tyrosine phosphorylation (Tyr(P)). Upregulation by Mas-7 was blocked by GF109203X, a potent inhibitor of protein kinase C (PKC). Phorbol ester also upregulated FAK-YP, verifying a role for PKC in the transduction cascade. Upregulation of FAK-YP by activation of G-proteins and PKC was dependent upon intact F-actin, as cytochalasin D abolished stimulation by Mas-7 and by phorbol ester. The relatively slow increase in FAK-YP evoked by chronic exposure to Abeta also was abolished by GF109203X and by cytochalasin D. The results show that tyrosine phosphorylation of FAK in neurons is regulated positively by PKC, functioning down-stream from G-proteins through an F-actin-dependent mechanism. The Alzheimer's Abeta peptide is capable of activating elements of this same signal transduction pathway, via membrane events that remain to be determined.


Asunto(s)
Actinas/metabolismo , Péptidos beta-Amiloides/metabolismo , Moléculas de Adhesión Celular/metabolismo , Proteínas de Unión al GTP/metabolismo , Neuronas/enzimología , Proteína Quinasa C/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Animales , Citocalasina D/farmacología , Activación Enzimática , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Indoles/farmacología , Maleimidas/farmacología , Neuronas/citología , Ésteres del Forbol/farmacología , Fosforilación , Proteína Quinasa C/antagonistas & inhibidores , Ratas , Células Tumorales Cultivadas
13.
Neurosci Lett ; 207(1): 17-20, 1996 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-8710199

RESUMEN

Use of fluorescent probes to monitor f-actin in living cells typically relies on difficult microinjection procedures. The current work has developed cationic lipotransfer of membrane-impermeant probes as an alternative to microinjection. BODIPY FL-phallacidin, a fluorescent f-actin probe, was packaged into 40-50 nm cationic liposomes. Packaging, verified by gel filtration, enabled delivery of the probe into living nerve cells and provided an image of f-actin that was identical to that seen in fixed, permeabilized cells. Phallacidin alone did not enter living cells, nor was its uptake stimulated by the presence of empty liposomes. All predicted f-actin structures were fluorescent in living cells, indicating a high efficacy of delivery. Cationic lipotransfer of fluorescent probes was rapid, not disruptive to cells, and delivered a probe en masse to a large sample population. Lipotransfer appears to be a promising alternative to microinjection for introducing membrane-impermeant probes and reagents into living cells.


Asunto(s)
Actinas/metabolismo , Colorantes Fluorescentes/farmacología , Neuronas/metabolismo , Péptidos Cíclicos/farmacología , Animales , Línea Celular , Portadores de Fármacos , Liposomas , Ratas
14.
J Biol Chem ; 271(8): 4436-40, 1996 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-8626795

RESUMEN

Amyloid precursor protein (APP) and cholesterol metabolism are genetically linked to Alzheimer's disease, the latter through apolipoprotein E, a lipid and cholesterol transport protein. We have examined the hypothesis that the processing of APP is disrupted by elevated cholesterol, which is known to modulate the activity of several transmembrane proteins. In the current study, cholesterol, solubilized by methyl- beta-cyclodextrin or ethanol, was added to the culture media of APP 751 stably transfected HEK 293 cells. Radiolabeled APP and APPsol (the soluble N-terminal derivative following alpha-secretase cleavage) were precipitated from lysates and conditioned media of stably transfected HEK 293 cells; the relative levels were determined by quantitative densitometry following separation by SDS-polyacrylamide gel electrophoresis. The data show that cholesterol, solubilized by methyl-beta-cyclodextrin, greatly reduced the levels of APPsol. Low doses of ethanol-solubilized cholesterol similarly caused a dramatic reduction of APPsol. By contrast, levels of APP holoprotein remained the same or increased. The large decrease seen in APPsol production was not due to nonspecific inhibition of secretion because several secreted proteins increased in level. Cholesterol, which impedes membrane fluidity, may lower APPsol production by impeding the interaction of the substrate with its protease(s). If APPsol were to function trophically, as suggested by other studies, the current conclusion suggests that changes in cellular cholesterol levels in Alzheimer's disease could contribute to neuronal degeneration by decreasing the production of APPsol.


Asunto(s)
Péptidos beta-Amiloides/biosíntesis , Precursor de Proteína beta-Amiloide/metabolismo , Colesterol/farmacología , Endopeptidasas/metabolismo , Procesamiento Proteico-Postraduccional , Secretasas de la Proteína Precursora del Amiloide , Péptidos beta-Amiloides/análisis , Péptidos beta-Amiloides/aislamiento & purificación , Ácido Aspártico Endopeptidasas , Línea Celular , Inhibidores Enzimáticos/farmacología , Humanos , Riñón , Cinética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/metabolismo , Solubilidad , Transfección
15.
Neurobiol Aging ; 16(4): 583-9, 1995.
Artículo en Inglés | MEDLINE | ID: mdl-8544909

RESUMEN

Paired helical filaments (PHFs) of Alzheimer's disease (AD) largely comprise hyperphosphorylated forms of the cytoskeletal protein tau. AD-type tau phosphoepitopes, detected by various monoclonal antibodies, are absent from normal adult neurons, but recent studies have shown that their expression may contribute to neuritogenesis and axon differentiation in the developing nervous system. Therefore, we have examined a brain nerve cell line that is spontaneously neuritogenic for possible expression of AD-type tau epitopes. The neuritogenic rat brain cell line B103 was found to constitutively produce two-AD related epitopes of tau, detected by cellular immunofluorescence studies with the PHF-1 and Alz-50 monoclonal antibodies. Biochemical studies showed that the antibodies bound to proteins within the molecular, weight range expected for phosphorylated tau isoforms. Further verification was established by use of tau antisense oligomers, which eliminated cellular immunofluorescence due to the AD-related monoclonals and polyclonal anti-tau but did not eliminate fluorescence due to anti-tubulin. Cells treated with tau antisense were not neurite-free. Neurites that remained, however, were abnormal, generally short and wavy in appearance. Cellular distribution of the tau epitopes was found to be particularly interesting. Alz-50 recognized only cytoplasmic tau whereas PHF-1 recognized nuclear tau as well as cytoplasmic. Thus, the two epitopes, are morphologically segregated within the cell. Because subcellular segregation of tau is compromised in Alzheimer's disease, mechanisms that segregate AD-type phosphotau epitopes in B103 cells may have relevance to this neurodegenerative disorder.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Sistema Nervioso Central/inmunología , Epítopos/metabolismo , Proteínas tau/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Línea Celular , Núcleo Celular/inmunología , Sistema Nervioso Central/citología , Citoplasma/inmunología , Técnica del Anticuerpo Fluorescente , Neuritas/inmunología , Oligonucleótidos Antisentido/farmacología , Fosforilación , Pruebas de Precipitina , Ensayo de Radioinmunoprecipitación , Ratas
16.
Neuroscience ; 66(4): 769-79, 1995 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-7651608

RESUMEN

Alzheimer's-afflicted neurons contain phosphorylated forms of tau that are not present in healthy adults. these can be recognized with great specificity by monoclonal antibodies such as paired helical filament-1 (PHF-1) [Greenberg S. G. and Davies P. (1990) Proc. natn. Acad. Sci. U.S.A. 87, 5827-5831; Greenberg S. G. et al. (1992) J. biol. Chem. 267, 564-569]. The PHF-1 phospho-tau epitope is also present in immature neurons undergoing axodendritic differentiation [Pope W. B. et al. (1993) Expl Neurol. 120, 106-113]. Analogous to its presence in immature neurons, we report here that the PHF-1 tau epitope spontaneously occurs in the human neuroblastoma cell line SHSY5Y, where its level can be regulated by differentiation and by molecules found in the extracellular matrix. Confocal immunofluorescence studies showed PHF-1 epitope to be constitutively expressed in the somatic cytoplasm as well as in short neurites typical of undifferentiated SHSY5Y cells. Induction of differentiation with retinoic acid produced cells with a neuronal morphology and a redistribution of the expression of PHF-1 tau in the long neurites. Protracted exposure to retinoic acid decreased the levels of PHF-1 immunofluorescence without a loss of neurites, similar to the developmental down-regulation seen in situ. The effects of retinoic acid on PHF-1 immunofluorescence were modifiable by fibronectin, which can be released by some neuroblastoma cell lines [Ciccarone V. et al. (1989) Cancer Res. 49, 219-225; Yoshihara T. et al. (1992) Int. J. Cancer 51, 620-626]. Exogenous human fibronectin caused a marked up-regulation of PHF-1 immunofluorescence. Quantitative analysis of 15 multicellular areas, from six different cultures, per experimental condition showed a 16-fold increase compared to untreated controls. Up-regulation by fibronectin was also evident in undifferentiated cells. Cell counts indicated no proliferative effects of the fibronectin under the conditions used. Laminin also caused an increase of PHF-1 tau in retinoic acid-treated cells. Data obtained from immunoblots verified the results observed with immunofluorescence. The data show that the PHF-1 tau epitope is spontaneously expressed by non-degenerating human neuroblastoma cells, down-regulated by cellular differentiation, induced by retinoic acid and up-regulated by the extracellular matrix components fibronectin and laminin. One explanation of the data is that fibronectin maintains a population of SHSY5Y cells in a biochemical state of differentiation in which PHF-1 tau is expressed.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Proteínas tau/metabolismo , Anticuerpos Monoclonales/inmunología , Biomarcadores , Western Blotting , División Celular/efectos de los fármacos , Línea Celular , Células Cultivadas , Fibronectinas/farmacología , Humanos , Immunoblotting , Inmunohistoquímica , Laminina/farmacología , Neuroblastoma , Regulación hacia Arriba/efectos de los fármacos
17.
Exp Neurol ; 132(1): 42-53, 1995 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-7720825

RESUMEN

The principle externalized forms of amyloid precursor protein (APP) are soluble and well-characterized, but some evidence has suggested the additional presence of externalized APP in a nonsoluble form. To further assess this possibility, the current study has applied high resolution microscopy protocols in addition to immunoprecipitation to characterize externalized APP in three commonly used cell culture models (SH-SY5Y human neuroblastoma cells, fetal rat brain cells, and HEK 293 human embryonic kidney cells). Confocal immunofluorescence microscopy, using an antiserum against the c-terminal domain of APP, showed typical cell-associated APP, but hot spots of APP also were evident in cell-free areas, apparently associated with the culture substrata. These hot-spots were examined for evidence of cellular deterioration by whole mount transmission electron microscopy. Neither cell debris nor disrupted cells were present. Instead, the hot spots of substratum-bound APP comprised discrete microparticles, approximately 50-100 nm across. These microparticles also could be found near cells and in some cases were attached to cell surface fibrils. Substratum-bound APP also could be found clustered within the extracellular matrix made by primary cell cultures. Occurrence of APP in extracellular microparticles was verified by centrifugation-immunoprecipitation analysis of media conditioned by APP-transfected cells. Radiolabeling data showed that particulate APP was from metabolically active cells. Metabolic labeling of particle-associated APP, as well as the absence of cellular debris near the APP-containing particles, suggests that the occurrence of nonsoluble APP in the extracellular milieu derives from a physiologically active process.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Espacio Extracelular/metabolismo , Animales , Anticuerpos/análisis , Encéfalo/metabolismo , Encéfalo/ultraestructura , Línea Celular , Riñón/embriología , Riñón/metabolismo , Riñón/ultraestructura , Neuroblastoma/metabolismo , Neuroblastoma/ultraestructura , Ratas , Células Tumorales Cultivadas
18.
J Neurochem ; 64(1): 307-15, 1995 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-7798927

RESUMEN

The amyloid precursor protein (APP) is a membrane-spanning glycoprotein that is the source of beta A4 peptides, which aggregate in Alzheimer's disease to form senile plaques. APP is cleaved within the beta A4 sequence to release a soluble N-terminal derivative (APPsol), which has a wide range of trophic and protective functions. In the current study we have examined the hypothesis that iron availability may modulate expression or processing of APP, whose mRNA contains, based on sequence homology, a putative iron response element (IRE). Radiolabeled APP and its catabolites were precipitated from lysates and conditioned medium of stably transfected HEK 293 cells using antibodies selective for C-terminal, beta A4, and N-terminal domains. The relative abundance of the different APP catabolites under different conditions of iron availability was determined by quantitative densitometry after separation by sodium dodecyl sulfate-polyacrylamide gel electrophoresis. The data show a specific effect on the production of APPsol. Using standard conditions previously established for IRE studies, it was found that iron chelation reduces APPsol production, whereas iron level elevation augments it. No changes were observed in levels of immature and mature APP holoprotein or in the C-terminal alpha-secretase derivative C83, beta A4, and p3 peptides. The specificity for modulatory changes in APPsol suggests that iron acts at the level of alpha-secretase activity. In addition to its modulatory effects, iron at very high levels was found to inhibit maturation of APP and production of its downstream catabolites without blocking formation of immature APP. The data establish a potential physiological role for iron in controlling the processing of APP.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Endopeptidasas/fisiología , Hierro/farmacología , Secretasas de la Proteína Precursora del Amiloide , Precursor de Proteína beta-Amiloide/genética , Animales , Autorradiografía , Línea Celular , Densitometría , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Poliacrilamida , Endopeptidasas/metabolismo , Pruebas de Precipitina , ARN Mensajero/análisis , ARN Mensajero/genética , Transfección
19.
Neurosci Lett ; 184(1): 25-8, 1995 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-7739799

RESUMEN

beta-Amyloid accumulates as extracellular aggregates in Alzheimer's-afflicted brain tissue, but it also is secreted by healthy tissue, for reasons not yet established. One possibility is that beta-amyloid, which contains a sequence (RHDS) homologous to the cell-binding domain of fibronectin, may modulate integrin function, a possibility supported by previous data from non-neuronal cells (Ghiso et al., Biochem. J., 288 (1992) 1053-1059). The current work shows that functional interaction with beta-amyloid peptides is also supported by integrins in neuronal cells. Experiments used the SH-SY5Y human neuroblastoma cell line, which was shown to contain integrins that mediated cell adhesion to substratum-bound fibronectin. Adhesion to fibronectin was partially blocked by synthetic beta-amyloid peptides containing the RHDS sequence. beta-Amyloid sequences adsorbed to substratum themselves were found to mediate cell adhesion, although less effectively than fibronectin. Anti-integrin blocked the peptide-mediated adhesion, at doses commensurate with those blocking fibronectin-mediated adhesion. The data support the hypothesis that beta-amyloid peptides could physiologically, and perhaps pathogenically, modulate the activity of neuronal integrins, important cell surface receptors known to control protein kinase activities, Ca2+ levels, gene expression and organization of the cytoskeleton.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Integrinas/metabolismo , Anticuerpos , Adhesión Celular , Fibronectinas/farmacología , Humanos , Inmunohistoquímica , Neuroblastoma/metabolismo , Neuronas/metabolismo , Péptidos/sangre , Células Tumorales Cultivadas
20.
J Neurochem ; 63(6): 2225-30, 1994 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-7964742

RESUMEN

The effect of the Kunitz proteinase inhibitor (KPI) on potential beta-amyloid precursor protein (beta PP)-processing activities from control and Alzheimer's disease (AD) brains was examined using fluorogenic substrates designed to mimic the secretory and amyloidogenic cleavages in beta PP. In addition, the level of secretion of KPI-containing beta PP751 and KPI-lacking beta PP695 from transfected cells was examined to assess the effect of the KPI on beta PP secretion. beta PP751 and beta PP695, obtained from conditioned media of transfected cells, had no effect on proteinase activities against the secretory and amyloidogenic substrates in extracts from control and AD brains. At similar concentrations beta PP751, but not beta PP695, completely inhibited the activity of trypsin against these substrates. Serine proteinase inhibitors had only modest effects on activities from brain, whereas cysteine modification completely inhibited them, indicating that these proteinase activities were not of the serine type. Thus, the results do not support a role for the KPI in the secretion of beta PP or in the amyloidogenic cleavage of beta PP. The amounts of beta PP695 and beta PP751 collected from the media of transfected cells after 48 h of growth were similar, indicating an equal rate of secretion. This result suggests that the KPI domain in beta PP751 did not inhibit the secretory cleavage in transfected cells.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Inhibidor de la Tripsina de Soja de Kunitz/farmacología , Anciano , Anciano de 80 o más Años , Secuencia de Aminoácidos , Precursor de Proteína beta-Amiloide/genética , Sitios de Unión , Línea Celular , Medios de Cultivo Condicionados , Endopeptidasas/metabolismo , Humanos , Immunoblotting , Persona de Mediana Edad , Datos de Secuencia Molecular , Proteínas Recombinantes/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...