Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Hum Mol Genet ; 21(3): 495-510, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22045698

RESUMEN

Huntington's disease (HD) is the most common of nine inherited neurological disorders caused by expanded polyglutamine (polyQ) sequences which confer propensity to self-aggregate and toxicity to their corresponding mutant proteins. It has been postulated that polyQ expression compromises the folding capacity of the cell which might affect other misfolding-prone proteins. α-Synuclein (α-syn) is a small neural-specific protein with propensity to self-aggregate that forms Parkinson's disease (PD) Lewy bodies. Point mutations in α-syn that favor self-aggregation or α-syn gene duplications lead to familial PD, thus indicating that increased α-syn aggregation or levels are sufficient to induce neurodegeneration. Since polyQ inclusions in HD and other polyQ disorders are immunopositive for α-syn, we speculated that α-syn might be recruited as an additional mediator of polyQ toxicity. Here, we confirm in HD postmortem brains and in the R6/1 mouse model of HD the accumulation of α-syn in polyQ inclusions. By isolating the characteristic filaments formed by aggregation-prone proteins, we found that N-terminal mutant huntingtin (N-mutHtt) and α-syn form independent filamentous microaggregates in R6/1 mouse brain as well as in the inducible HD94 mouse model and that N-mutHtt expression increases the load of α-syn filaments. Accordingly, α-syn knockout results in a diminished number of N-mutHtt inclusions in transfected neurons and also in vivo in the brain of HD mice. Finally, α-syn knockout attenuates body weight loss and early motor phenotype of HD mice. This study therefore demonstrates that α-syn is a modifier of polyQ toxicity in vivo and raises the possibility that potential PD-related therapies aimed to counteract α-syn toxicity might help to slow HD.


Asunto(s)
Enfermedad de Huntington/etiología , Cuerpos de Inclusión/química , alfa-Sinucleína/análisis , Animales , Apoptosis , Atrofia , Modelos Animales de Enfermedad , Femenino , Humanos , Proteína Huntingtina , Enfermedad de Huntington/patología , Longevidad , Masculino , Ratones , Ratones Noqueados , Actividad Motora , Mutación , Neostriado/patología , Proteínas del Tejido Nervioso/análisis , Proteínas del Tejido Nervioso/genética , Neuronas/química , Proteínas Nucleares/genética , Fenotipo , Pérdida de Peso , alfa-Sinucleína/genética
2.
Hippocampus ; 21(8): 910-22, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20575007

RESUMEN

Overexpression of GSK3ß in transgenic mice induces learning deficits and some features associated with Alzheimer's disease (AD), including dentate gyrus (DG) atrophy. Here, we assessed whether these mice also recapitulate DG atrophy as well as impaired neurogenesis reported in AD. Ultrastructural analysis revealed that there were fewer and more disorganized neurogenic niches in these animals, coupled with an increase in the proportion of immature neurons. Indeed, the maturation of granule cells is delayed as witnessed by the alterations to the length and patterning of their dendritic trees and to the mossy fiber terminals. Together with an increase in neuronal death, these phenomena lead to a marked decrease in the number and disorganization of granule cells of the DG. Our results suggest that GSK3ß overexpression perturbs proliferation and maturation, resulting in the loss of immature neurons. In turn, the activation of microglia is stimulated in conjunction with a decrease in the birth of new functional neurons, leading to the deterioration of this structure. These data support the idea that by inducing degeneration of the DG, GSK3ß could be involved in the pathogenesis of AD.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Giro Dentado , Glucógeno Sintasa Quinasa 3 , Neurogénesis , Animales , Muerte Celular , Diferenciación Celular , Proliferación Celular , Giro Dentado/patología , Giro Dentado/ultraestructura , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía
3.
Neurobiol Dis ; 37(3): 622-9, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20004245

RESUMEN

It has been proposed that deregulation of neuronal glycogen synthase kinase 3 (GSK3) activity may be a key feature in Alzheimer disease pathogenesis. We have previously generated transgenic mice that overexpress GSK3beta in forebrain regions including dentate gyrus (DG), a region involved in learning and memory acquisition. We have found that GSK3 overexpression results in DG degeneration. To test whether tau protein modified by GSK3 plays a role in that neurodegeneration, we have brought GSK3 overexpressing mice to a tau knockout background. Our results indicate that the toxic effect of GSK3 overexpression is milder and slower in the absence of tau. Thus, we suggest that the hyperphosphorylated tau mediates, at least in part, the pathology observed in the brain of GSK3 overexpressing mice.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Hipocampo/metabolismo , Discapacidades para el Aprendizaje/metabolismo , Degeneración Nerviosa/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Animales , Atrofia/genética , Atrofia/metabolismo , Atrofia/patología , Biomarcadores/metabolismo , Núcleo Celular/metabolismo , Núcleo Celular/patología , Giro Dentado/metabolismo , Giro Dentado/patología , Giro Dentado/fisiopatología , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Regulación Enzimológica de la Expresión Génica/genética , Gliosis/genética , Gliosis/metabolismo , Gliosis/patología , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Hipocampo/patología , Hipocampo/fisiopatología , Discapacidades para el Aprendizaje/genética , Discapacidades para el Aprendizaje/fisiopatología , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Ovillos Neurofibrilares/genética , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Neuronas/metabolismo , Neuronas/patología , Fosforilación , beta Catenina/metabolismo , Proteínas tau/genética
4.
Neurosci Res ; 63(4): 244-50, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19167438

RESUMEN

Changes in the retino-collicular projection and in the number of optic nerve (ON) axons in adult rats were analyzed after partial loss of retinal ganglion cells (RGCs), induced by intravitreal injections of kainic acid (KA) on postnatal days 2-3 (P2-P3) or 10-12 (P10-P12). KA injected at P2-P3 decreased the volume of the adult contralateral superior colliculus (SC) and the density of the retino-collicular contralateral projection, but maintained the neonatal pattern in the ipsilateral projection from the un-injected eye. ON axon number was significantly increased in the un-injected eye but decreased in the KA-injected eye. Thus, restriction of the ipsilateral retino-collicular projection and RGC death in the un-injected eye are modified by KA at P2-P3, during the postnatal critical period, but not at P10-P12, after it is over. We suggest that, in the SC contralateral to the KA-injected eye, the disappearance of axon terminals belonging to RGC killed by KA would decrease competition between ipsilateral and contralateral terminals, thus contributing to maintaining the neonatal pattern in the ipsilateral retino-collicular projection. The reduction in RGC death in the un-injected eye could also be related to the disappearance of RGC terminals in the contralateral SC, which would have increased neurotrophic factor availability.


Asunto(s)
Agonistas de Aminoácidos Excitadores/farmacología , Ácido Kaínico/farmacología , Plasticidad Neuronal/efectos de los fármacos , Retina/efectos de los fármacos , Células Ganglionares de la Retina/efectos de los fármacos , Vías Visuales , Factores de Edad , Animales , Animales Recién Nacidos , Lateralidad Funcional/efectos de los fármacos , Lateralidad Funcional/fisiología , Degeneración Nerviosa/inducido químicamente , Plasticidad Neuronal/fisiología , Nervio Óptico/efectos de los fármacos , Nervio Óptico/crecimiento & desarrollo , Ratas , Ratas Sprague-Dawley , Retina/citología , Retina/crecimiento & desarrollo , Células Ganglionares de la Retina/fisiología , Colículos Superiores , Vías Visuales/citología , Vías Visuales/efectos de los fármacos , Vías Visuales/crecimiento & desarrollo
5.
Acta Neuropathol ; 116(4): 409-18, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18679696

RESUMEN

We have previously reported that double-transgenic APP(SW)/Tau(VLW) mice show enhanced amyloid deposition, stronger tau hyperphosphorylation, increased sarkosyl tau polymers, and wider tau filaments when compared to simple mutant models. To validate these transgenic mice as models of Alzheimer disease pathology, in the present study we analyze tau phosphorylation at 12E8 and AT-8 epitopes in amyloid plaques. In APP(SW) mice, phospho-tau in plaque-associated neurites suggests a local direct effect of plaque-amyloid (and/or APP(SW)) on tau phosphorylation. In vitro, attempts to identify which kinases are induced by fibrillar amyloid reveal to Protein Kinase C as responsible for phosphorylation at the 12E8 epitope. Tau(VLW) mice, without plaques, show increased tau phosphorylation at the 12E8 epitope, particularly in pyramidal neurons. APP(SW)/Tau(VLW) mice show earlier and stronger 12E8 tau phosphorylation. Ultrastructurally, the same two types of neurites are found in plaques from APP(SW)/Tau(VLW) and Alzheimer disease (AD) brains: (a) dystrophic giant neurites filled with degenerating organelles and/or phospho-tau-positive filaments and (b) non-dystrophic phospho-tau-positive small punctiform neurites. Both types of plaque-associated neurites are AT-8 positive in APP(SW)/Tau(VLW) mice and AD, but 12E8-positive dystrophic neurites are only detected in AD. We conclude that the simultaneous presence of human mutated Tau(VLW) and plaque-amyloid (and/or APP(SW)) potentiates and anticipates tau phosphorylation at the 12E8 epitope, intensifying pyramidal neuron immunostaining and tau filament formation in this double-transgenic model. Thus, the APP(SW)/Tau(VLW) mouse is a useful model to study neuritic plaques, since they reproduce most of the characteristics that these structures have in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Placa Amiloide/metabolismo , Proteínas tau/metabolismo , Anciano , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/farmacología , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Fosforilación , Placa Amiloide/patología , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , Células Piramidales/patología , Proteínas tau/genética
6.
J Alzheimers Dis ; 11(1): 53-9, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17361035

RESUMEN

Autopsied brain tissue from Alzheimer's disease patients and old non-demented controls was studied after immunocytochemistry with the 4G8 monoclonal antibody that recognizes amyloid-beta peptides. Intraneuronal 4G8-positive reaction product was detected in all of the studied brains. The same brain regions in the Alzheimer's disease samples consistently showed both more immunopositive neurons and more stained reaction product per neuron than those from the non-demented brains. Ultrastructurally, the immunopositive reaction product accumulated in clusters of cytoplasmic elements that had a lipofuscin-like appearance, showed a fibrogranular content and were also closely apposed to lipid droplets located either on their periphery or within them. The most strongly 4G8-immunopositive elements had diffuse limits with their fibrogranular content free in the cytoplasm, whereas elements either without or showing only light immunoreaction had a limiting membrane. All immunopositive neurons displayed a similar pattern of clumping heterochromatin. The hypothetical neurotoxic role of intraneuronal amyloid-beta peptides free in the cytoplasm is discussed as is the possible relationship between the amyloid-beta peptides recognized by the 4G8 antibody and the lipid droplets which would presumably contain esterified cholesterol.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/ultraestructura , Encéfalo/patología , Citoplasma/diagnóstico por imagen , Anciano , Anciano de 80 o más Años , Ésteres del Colesterol/análisis , Femenino , Humanos , Técnicas para Inmunoenzimas , Cuerpos de Inclusión/diagnóstico por imagen , Lipofuscina/análisis , Masculino , Microscopía Electrónica , Neuronas/patología , Valores de Referencia , Ultrasonografía
7.
J Neurosci ; 25(42): 9773-81, 2005 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-16237181

RESUMEN

The primary mechanism responsible for Huntington's disease remains unknown. Postulated early pathogenic events include the following: impaired protein folding, altered protein degradation, mitochondrial dysfunction, and transcriptional dysregulation. Although related therapies can delay disease progression in mouse models, they target downstream and probably indirect effects of mutant-huntingtin expression. Accordingly, in case they prove beneficial in humans, they might only palliate some aspects of disease. Our previous studies in the Tet/HD94 conditional model and the recently reported efficacy of RNA interference against mutant huntingtin in another mouse model support silencing mutant-huntingtin expression as a valid therapeutic approach that has the advantage of targeting toxicity at its root. Here, we address whether gene silencing can still be beneficial in the late stages of disease with detectable striatal neuron loss. Stereological analysis was applied to determine an age at which Tet/HD94 mice show a decrease in the number of striatal neurons. Then, progression of neuropathology and motor phenotype were analyzed in mice that were allowed to continue expressing mutant huntingtin and in mice that no longer expressed it. Neuronal loss did not revert in gene-off mice, but the additional loss that takes place in gene-on mice was prevented. The total number of huntingtin-containing inclusions dramatically reverted, but a small fraction of inclusions positive for the amyloid dye thioflavin-S remained. Interestingly, despite a 20% decrease in striatal neurons and the presence of amyloid-like irreversible inclusions, gene-off mice fully recover from their motor deficit, thus ruling out amyloid-like huntingtin inclusions as the main toxic species and suggesting that gene-silencing therapies might work in late stages of disease.


Asunto(s)
Amiloide/metabolismo , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Enfermedad de Huntington/patología , Destreza Motora , Neuronas/patología , Amiloide/genética , Amiloide/fisiología , Animales , Recuento de Células/métodos , Muerte Celular/fisiología , Cuerpo Estriado/fisiología , Enfermedad de Huntington/genética , Ratones , Ratones Transgénicos , Destreza Motora/fisiología , Recuperación de la Función/fisiología
8.
J Neurosci ; 24(42): 9361-71, 2004 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-15496672

RESUMEN

Huntington's disease (HD) and eight additional inherited neurological disorders are caused by CAG triplet-repeat expansions leading to expanded polyglutamine-sequences in their respective proteins. These triplet-CAG repeat disorders have in common the formation of aberrant intraneuronal proteinaceous inclusions containing the expanded polyglutamine sequences. These aggregates have been postulated to contribute to pathogenesis caused by conformational toxicity, sequestration of other polyglutamine-containing proteins, or by interfering with certain enzymatic activities. Testing these hypotheses has been hampered by the difficulty to isolate these aggregates from brain. Here we report that polyglutamine aggregates can be isolated from the brain of the Tet/HD94 conditional mouse model of HD, by following a method based on high salt buffer homogenization, nonionic detergent extraction, and gradient fractionation. We then verified that the method can be successfully applied to postmortem HD brains. Immunoelectron microscopy, both in human and mouse samples, revealed that the stable component of the inclusions are mutant huntingtin-containing and ubiquitin-containing fibrils. Atomic-force microscopy revealed that these fibrils have a "beads on a string" morphology. Thus, they resemble the in vitro assembled filaments made of recombinant mutant-huntingtin, as well as the Abeta and alpha-synuclein amyloid protofibrils. Finally, by shutting down transgene expression in the Tet/HD94 conditional mouse model of HD, we were able to demonstrate that these filaments, although stable in vitro, are susceptible to revert in vivo, thus demonstrating that the previously reported reversal of ubiquitin-immunoreactive inclusions does not simply reflect disassembling of the inclusions into their constituent fibrils and suggesting that any associated conformational or protein-sequestration toxicity is also likely to revert.


Asunto(s)
Encéfalo/metabolismo , Enfermedad de Huntington/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas Nucleares/química , Anciano , Anciano de 80 o más Años , Animales , Western Blotting , Encéfalo/ultraestructura , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Proteína Huntingtina , Enfermedad de Huntington/patología , Cuerpos de Inclusión/ultraestructura , Masculino , Ratones , Ratones Transgénicos , Microscopía de Fuerza Atómica , Microscopía Inmunoelectrónica , Complejos Multiproteicos/aislamiento & purificación , Proteínas del Tejido Nervioso/aislamiento & purificación , Proteínas del Tejido Nervioso/ultraestructura , Proteínas Nucleares/aislamiento & purificación , Proteínas Nucleares/ultraestructura , Péptidos/análisis , Ubiquitina/análisis
9.
J Neurosci ; 23(37): 11653-61, 2003 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-14684867

RESUMEN

Huntington's disease (HD) inclusions are stained with anti-ubiquitin and anti-proteasome antibodies. This, together with proteasome activity studies on transfected cells, suggest that an impairment of the ubiquitin-proteasome system (UPS) may be key in HD pathogenesis. To test whether proteasome activity is impaired in vivo, we performed enzymatic assays for the three peptidase activities of the proteasome in brain extracts from the HD94 conditional mouse model of HD. We found no inhibition of any of the activities, suggesting that if UPS impairment happens in vivo, it is not at the level of the proteasome catalytic core. Intriguingly, the chymotrypsin- and trypsin-like activities increased selectively in the affected and aggregate-containing regions: cortex and striatum. Western blot analysis revealed no difference in total proteasome content whereas an increase in the interferon-inducible subunits of the immunoproteasome, LMP2 and LMP7, was observed. These subunits confer to the proteasome catalytic properties that are optimal for MHC-I peptide presentation. Immunohistochemistry in control mouse brain revealed LMP2 and LMP7 mainly in neurons. Accordingly, their increase in HD94 mice predominantly took place in neurons, and 5% of the ubiquitin-positive cortical aggregates were also LMP2-positive. Ultrastructural analysis of neurons with high level of immunoproteasome subunits revealed signs of neurodegeneration like nuclear indentation or fragmentation and dark cell appearance. The neuronal induction of LMP2 and LMP7 and the associated signs of neurodegeneration were also found in HD postmortem brains. Our results indicate that LMP2 and LMP7 participate in normal neuronal physiology and suggest a role in HD neurodegeneration.


Asunto(s)
Cisteína Endopeptidasas/biosíntesis , Enfermedad de Huntington/enzimología , Complejos Multienzimáticos/biosíntesis , Neuronas/enzimología , Anciano , Animales , Encéfalo/patología , Corteza Cerebral/enzimología , Quimotripsina/metabolismo , Cisteína Endopeptidasas/inmunología , Cisteína Endopeptidasas/metabolismo , Femenino , Humanos , Enfermedad de Huntington/patología , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Complejos Multienzimáticos/inmunología , Complejos Multienzimáticos/metabolismo , Neostriado/enzimología , Neuronas/ultraestructura , Complejo de la Endopetidasa Proteasomal , Tripsina/metabolismo , Ubiquitinas/análisis
10.
J Alzheimers Dis ; 19(1): 37-56, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20061625

RESUMEN

RhoGTPases control cytoskeleton dynamics thereby modulating synaptic plasticity. Because Alzheimer's disease (AD) is characterized by synaptic dysfunction, we sought to determine whether the expression, activity, or localization of the GTPases RhoA, Rac1 and Cdc42, as well as p21-PAK, a downstream target of Rac1/Cdc42, were altered in 18-month-old AbetaPP Tg2576 mice (Swedish mutation) or in brains from patients with AD and, for comparison in the case of RhoA, Pick's disease (PiD), a neurodegenerative disorder characterized by hyper-phosphorylated tau accumulation. Immunohistochemical analyses revealed a distinct localization of each RhoGTPase in synapses, dendrite shafts, neuronal bodies, or astrocytes. The association of RhoA with synapses and dendritic microtubules was confirmed by electron microscopy. In AbetaPP mice, RhoA expression decreased in synapses and increased in dystrophic neurites, suggesting altered subcellular targeting of RhoA. In AD, RhoA immunostaining decreased in the neuropil and markedly increased in neurons, co-localizing with hyperphosphorylated tau inclusions, as though RhoA were sequestered by neurofibrillary tangles. Additionally, total RhoA protein was lower in the AD brain hippocampus, reflecting loss of the membrane bound, presumably active, GTPase. RhoA colocalized with hyperphosphorylated tau in PiD, again suggesting that altered subcellular targeting of RhoA is related to neurodegeneration. No major immunohistochemical changes were observed for Rac1, Cdc42, or p21-PAK, thus identifying RhoA among RhoGTPases as a possible therapeutic target in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/biosíntesis , Precursor de Proteína beta-Amiloide/genética , Animales , Regulación hacia Abajo/genética , Humanos , Cuerpos de Inclusión/genética , Cuerpos de Inclusión/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuritas/metabolismo , Neuritas/patología , Ovillos Neurofibrilares/genética , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Fosforilación , Enfermedad de Pick/genética , Enfermedad de Pick/metabolismo , Enfermedad de Pick/patología , Sinapsis/genética , Sinapsis/metabolismo , Regulación hacia Arriba/genética , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/biosíntesis , Proteína de Unión al GTP rhoA/genética , Proteínas tau/genética , Proteínas tau/metabolismo
11.
Neurobiol Aging ; 27(9): 1258-68, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16054268

RESUMEN

Here we have tested whether tau modification either by point mutation or by hyperphosphorylation can exert maximal pathogenic effects or if, on the contrary, both types of tau modifications can act synergistically to induce neuropathology. For this, we have combined transgenic mice overexpressing the enzyme GSK-3beta (Tet/GSK-3beta mice), with transgenic mice expressing Tau with a triple FTDP-17 mutation which develop prefibrillar tau-aggregates (VLW mice). Tet/GSK-3beta/VLW transgenic mice show tau hyperphosphorylation in hippocampal neurons. This is accompanied by thioflavin-S staining, and formation of filaments similar in width to those found in tauophaties. Finally, the atrophy of the hippocampal dentate gyrus observed in Tet/GSK-3beta mice develops much faster in Tet/GSK-3beta/VLW mice. All these morphological and biochemical data demonstrate that there is a synergistic contribution of both types of tau modifications and that the potential of GSK-3 inhibitors for AD therapeutics also extends to tauopathies caused by point mutations in tau gene.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Degeneración Nerviosa/diagnóstico , Proteínas tau/metabolismo , Animales , Benzotiazoles , Biomarcadores/metabolismo , Western Blotting/métodos , Giro Dentado/patología , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Inmunohistoquímica/métodos , Ratones , Ratones Transgénicos , Proteínas de Neurofilamentos/metabolismo , Proteínas de Neurofilamentos/ultraestructura , Fosforilación , Tiazoles , Proteínas tau/genética
12.
J Neurochem ; 98(5): 1585-96, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16787406

RESUMEN

In Huntington's disease (HD), as in the rest of CAG triplet-repeat disorders, the expanded polyglutamine (polyQ)-containing proteins form intraneuronal fibrillar aggregates that are gathered into inclusion bodies (IBs). Since IBs contain ubiquitin and proteasome subunits, it was proposed that inhibition of proteasome activity might underlie pathogenesis of polyQ disorders. Recent in vitro enzymatic studies revealed the inability of eukaryotic proteasomes to digest expanded polyQ, thus suggesting that occasional failure of polyQ to exit the proteasome may interfere with its proteolytic function. However, it has also recently been found that in vitro assembled aggregates made of synthetic polyQ fail to inhibit proteasome activity. Because synthetic polyQ aggregates lack the post-translational modifications found inside affected neurons, such as poly ubiquitylation, we decided to study the effect of mutant huntingtin (htt) aggregates isolated from the Tet/HD94 mouse model and from human HD brain tissue. Here, we show that isolated ubiquitylated filamentous htt aggregates, extracted from IBs by a previously reported method, selectively inhibited the in vitro peptidase activity of the 26S but not of the 20S proteasome in a non-competitive manner. In good agreement, immuno-electron microscopy revealed a direct interaction of htt filaments with the 19S ubiquitin-interacting regulatory caps of the 26S proteasome. Here, we also report a new method for isolation of IBs based on magnetic sorting. Interestingly, isolated IBs did not modify proteasome activity. Our results therefore show that mutant htt filamentous aggregates can inhibit proteasome activity, but only when not recruited into IBs, thus strengthening the notion that IB formation is protective by neutralizing toxicity of dispersed filamentous htt aggregates.


Asunto(s)
Enfermedad de Huntington/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/química , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Anciano , Anciano de 80 o más Años , Animales , Cumarinas/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Citometría de Flujo/métodos , Humanos , Enfermedad de Huntington/patología , Cuerpos de Inclusión/metabolismo , Cuerpos de Inclusión/patología , Cuerpos de Inclusión/ultraestructura , Masculino , Ratones , Ratones Transgénicos , Microscopía de Fuerza Atómica , Microscopía Inmunoelectrónica/métodos , Mutación/fisiología , Oligopéptidos/farmacología , Péptidos , Factores de Tiempo , Ubiquitina/química , Ubiquitina/genética , Ubiquitina/metabolismo
13.
Neurobiol Dis ; 20(3): 814-22, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16125396

RESUMEN

Even though the idea that amyloid beta peptide accumulation is the primary event in the pathogenesis of Alzheimer's disease has become the leading hypothesis, the causal link between aberrant amyloid precursor protein processing and tau alterations in this type of dementia remains controversial. We further investigated the role of beta-amyloid production/deposition in tau pathology and neuronal cell death in the mouse brain by crossing Tg2576 and VLW lines expressing human mutant amyloid precursor protein and human mutant tau, respectively. The resulting double transgenic mice showed enhanced amyloid deposition accompanied by neurofibrillary degeneration and overt neuronal loss in selectively vulnerable brain limbic areas. These findings challenge the idea that tau pathology in Alzheimer's disease is merely a downstream effect of amyloid production/deposition and suggest that reciprocal interactions between beta-amyloid and tau alterations may take place in vivo.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Degeneración Nerviosa/metabolismo , Ovillos Neurofibrilares/metabolismo , Placa Amiloide/metabolismo , Proteínas tau/genética , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/biosíntesis , Animales , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/fisiopatología , Muerte Celular/genética , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación/genética , Degeneración Nerviosa/genética , Degeneración Nerviosa/fisiopatología , Ovillos Neurofibrilares/genética , Neuronas/metabolismo , Neuronas/patología , Placa Amiloide/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA