Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Neurosci ; 38(50): 10595-10606, 2018 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-30355631

RESUMO

Intracellular neurofibrillary tangles (NFTs) composed of tau protein are a neuropathological hallmark of several neurodegenerative diseases, the most common of which is Alzheimer's disease (AD). For some time NFTs were considered the primary cause of synaptic dysfunction and neuronal death, however, more recent evidence suggests that soluble aggregates of tau are key drivers of disease. Here we investigated the effect of different tau species on synaptic plasticity in the male rat hippocampus in vivo Intracerebroventricular injection of soluble aggregates formed from either wild-type or P301S human recombinant tau potently inhibited hippocampal long-term potentiation (LTP) at CA3-to-CA1 synapses. In contrast, tau monomers and fibrils appeared inactive. Neither baseline synaptic transmission, paired-pulse facilitation nor burst response during high-frequency conditioning stimulation was affected by the soluble tau aggregates. Similarly, certain AD brain soluble extracts inhibited LTP in a tau-dependent manner that was abrogated by either immunodepletion with, or coinjection of, a mid-region anti-tau monoclonal antibody (mAb), Tau5. Importantly, this tau-mediated block of LTP was prevented by administration of mAbs selective for the prion protein (PrP). Specifically, mAbs to both the mid-region (6D11) and N-terminus (MI-0131) of PrP prevented inhibition of LTP by both recombinant and brain-derived tau. These findings indicate that PrP is a mediator of tau-induced synaptic dysfunction.SIGNIFICANCE STATEMENT Here we report that certain soluble forms of tau selectively disrupt synaptic plasticity in the live rat hippocampus. Further, we show that monoclonal antibodies to cellular prion protein abrogate the impairment of long-term potentiation caused both by recombinant and Alzheimer's disease brain-derived soluble tau. These findings support a critical role for cellular prion protein in the deleterious synaptic actions of extracellular soluble tau in tauopathies, including Alzheimer's disease. Thus, approaches targeting cellular prion protein, or downstream pathways, might provide an effective strategy for developing therapeutics.


Assuntos
Hipocampo/metabolismo , Hipocampo/patologia , Plasticidade Neuronal/fisiologia , Proteínas PrPC/metabolismo , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Inibidores da Angiogênese/farmacologia , Animais , Feminino , Hipocampo/efeitos dos fármacos , Humanos , Masculino , Plasticidade Neuronal/efeitos dos fármacos , Proteínas Priônicas/metabolismo , Ratos
2.
Curr Opin Neurobiol ; 52: 182-187, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30121451

RESUMO

Gamma oscillations (∼25-100 Hz) are believed to play a role in cognition. Accordingly, aberrant gamma oscillations have been observed in several cognitive disorders, including Alzheimer's disease and Fragile X syndrome. Here, we review how recent results showing abnormal gamma rhythms in Alzheimer's disease and Fragile X syndrome help reveal links between cellular disturbances and cognitive impairments. We also discuss how gamma results from rodent models of Alzheimer's disease and Fragile X syndrome may provide insights about unique functions of distinct slow (∼25-50 Hz) and fast gamma (∼55-100 Hz) subtypes. Finally, we consider studies employing brain stimulation paradigms in Alzheimer's disease and discuss how such studies may reveal causal relationships between gamma impairments and memory disturbances.


Assuntos
Doença de Alzheimer/fisiopatologia , Encéfalo/fisiopatologia , Transtornos Cognitivos/fisiopatologia , Estimulação Encefálica Profunda , Síndrome do Cromossomo X Frágil/fisiopatologia , Ritmo Gama/fisiologia , Transtornos da Memória/fisiopatologia , Doença de Alzheimer/terapia , Animais , Transtornos Cognitivos/terapia , Síndrome do Cromossomo X Frágil/terapia , Humanos , Transtornos da Memória/terapia
3.
J Neurophysiol ; 119(2): 476-489, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29070630

RESUMO

CA1 place cells become more anticipatory with experience, an effect thought to be caused by NMDA receptor-dependent plasticity in the CA3-CA1 network. Theta (~5-12 Hz), slow gamma (~25-50 Hz), and fast gamma (~50-100 Hz) rhythms are thought to route spatial information in the hippocampal formation and to coordinate place cell ensembles. Yet, it is unknown whether these rhythms exhibit experience-dependent changes concurrent with those observed in place cells. Slow gamma rhythms are thought to indicate inputs from CA3 to CA1, and such inputs are thought to be strengthened with experience. Thus, we hypothesized that slow gamma rhythms would become more evident with experience. We tested this hypothesis using mice freely traversing a familiar circular track for three 10-min sessions per day. We found that slow gamma amplitude was reduced in the early minutes of the first session of each day, even though both theta and fast gamma amplitudes were elevated during this same period. However, in the first minutes of the second and third sessions of each day, all three rhythms were elevated. Interestingly, theta was elevated to a greater degree in the first minutes of the first session than in the first minutes of later sessions. Additionally, all three rhythms were strongly influenced by running speed in dynamic ways, with the influence of running speed on theta and slow gamma changing over time within and across sessions. These results raise the possibility that experience-dependent changes in hippocampal rhythms relate to changes in place cell activity that emerge with experience. NEW & NOTEWORTHY We show that CA1 theta, slow gamma, and fast gamma rhythms exhibit characteristic changes over time within sessions in familiar environments. These effects in familiar environments evolve across repeated sessions.


Assuntos
Região CA1 Hipocampal/fisiologia , Ritmo Gama , Corrida , Ritmo Teta , Animais , Aprendizagem , Camundongos , Camundongos Endogâmicos C57BL
4.
Cereb Cortex ; 27(7): 3724-3735, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27390019

RESUMO

Dysregulation of glutamate homeostasis in the interstitial fluid of the brain is strongly implicated in causing synaptic dysfunction in many neurological and psychiatric illnesses. In the case of Alzheimer's disease (AD), amyloid ß (Aß)-mediated disruption of synaptic plasticity and memory can be alleviated by interventions that directly remove glutamate or block certain glutamate receptors. An alternative strategy is to facilitate the removal of excess glutamate from the nervous system by activating peripheral glutamate clearance systems. One such blood-based system, glutamate oxaloacetate transaminase (GOT), is activated by oxaloacetate, which acts as a co-substrate. We report here that synthetic and AD brain-derived Aß-mediated inhibition of synaptic long-term potentiation in the hippocampus is alleviated by oxaloacetate. Moreover the effect of oxaloacetate was GOT-dependent. The disruptive effects of a general inhibitor of excitatory amino acid transport or TNFα, a pro-inflammatory mediator of Aß action, were also reversed by oxaloacetate. Furthermore, another intervention that increases peripheral glutamate clearance, peritoneal dialysis, mimicked the beneficial effect of oxaloacetate. These findings lend support to the promotion of the peripheral clearance of glutamate as a means to alleviate synaptic dysfunction that is caused by impaired glutamate homeostasis in the brain.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Ácido Glutâmico/sangue , Hipocampo/metabolismo , Homeostase/fisiologia , Sinapses/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Aspartato Aminotransferase Citoplasmática/farmacologia , Ácido Aspártico/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Feminino , Hipocampo/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Humanos , Injeções Intraperitoneais , Masculino , Ácido Oxaloacético/farmacologia , Fragmentos de Peptídeos/farmacologia , Ratos , Ratos Wistar , Sinapses/fisiologia , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo
5.
Hippocampus ; 27(4): 378-392, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28032686

RESUMO

Alzheimer's disease (AD) is an irreversible and highly progressive neurodegenerative disease. Clinically, patients with AD display impairments in episodic and spatial memory. However, the underlying neuronal dysfunctions that result in these impairments remain poorly understood. The hippocampus is crucial for spatial and episodic memory, and thus we tested the hypothesis that abnormal neuronal representations of space in the hippocampus contribute to memory deficits in AD. To test this hypothesis, we recorded spikes from place cells in hippocampal subfield CA1, together with corresponding rhythmic activity in local field potentials, in the 3xTg AD mouse model. We observed disturbances in place cell firing patterns, many of which were consistent with place cell disturbances reported in other rodent models of AD. We found place cell representations of space to be unstable in 3xTg mice compared to control mice. Furthermore, coordination of place cell firing by hippocampal rhythms was disrupted in 3xTg mice. Specifically, a smaller proportion of place cells from 3xTg mice were significantly phase-locked to theta and slow gamma rhythms, and the theta and slow gamma phases at which spikes occurred were also altered. Remarkably, these disturbances were observed at an age before detectable Aß pathology had developed. Consistencies between these findings in 3xTg mice and previous findings from other AD models suggest that disturbances in place cell firing and hippocampal rhythms are related to AD rather than reflecting peculiarities inherent to a particular transgenic model. Thus, disturbed rhythmic organization of place cell activity may contribute to unstable spatial representations, and related spatial memory deficits, in AD. © 2017 Wiley Periodicals, Inc.


Assuntos
Doença de Alzheimer/fisiopatologia , Hipocampo/fisiopatologia , Células de Lugar/fisiologia , Percepção Espacial/fisiologia , Potenciais de Ação/fisiologia , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Modelos Animais de Doenças , Eletrodos Implantados , Ritmo Gama/fisiologia , Hipocampo/patologia , Humanos , Imuno-Histoquímica , Masculino , Camundongos da Linhagem 129 , Camundongos Transgênicos , Células de Lugar/patologia , Comportamento Espacial/fisiologia , Ritmo Teta/fisiologia , Proteínas tau/genética , Proteínas tau/metabolismo
6.
Neurobiol Dis ; 82: 372-384, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26215784

RESUMO

Compelling genetic evidence links the amyloid precursor protein (APP) to Alzheimer's disease (AD). A leading hypothesis proposes that a small amphipathic fragment of APP, the amyloid ß-protein (Aß), self-associates to form soluble assemblies loosely referred to as "oligomers" and that these are primary mediators of synaptic dysfunction. As such, Aß, and specifically Aß oligomers, are targets for disease modifying therapies. Currently, the most advanced experimental treatment for AD relies on the use of anti-Aß antibodies. In this study, we tested the ability of the monomer-preferring antibody, m266 and a novel aggregate-preferring antibody, 1C22, to attenuate spatial reference memory impairments in J20 mice. Chronic treatment with m266 resulted in a ~70-fold increase in Aß detected in the bloodstream, and a ~50% increase in water-soluble brain Aß--and in both cases Aß was bound to m266. In contrast, 1C22 increased the levels of free Aß in the bloodstream, and bound to amyloid deposits in J20 brain. However, neither 1C22 nor m266 attenuated the cognitive deficits evident in 12month old J20 mice. Moreover, both antibodies failed to alter the levels of soluble Aß oligomers in J20 brain. These results suggest that Aß oligomers may mediate the behavioral deficits seen in J20 mice and highlight the need for the development of aggregate-preferring antibodies that can reach the brain in sufficient levels to neutralize bioactive Aß oligomers. Aside from the lack of positive effect of m266 and 1C22 on cognition, a substantial number of deaths occurred in m266- and 1C22-immunized J20 mice. These fatalities were specific to anti-Aß antibodies and to the J20 mouse line since treatment of wild type or PDAPP mice with these antibodies did not cause any deaths. These and other recent results indicate that J20 mice are particularly susceptible to targeting of the APP/Aß/tau axis. Notwithstanding the specificity of fatalities for J20 mice, it is worrying that the murine precursor (m266) of a lead experimental therapeutic, Solanezumab, did not engage with putatively pathogenic Aß oligomers.


Assuntos
Peptídeos beta-Amiloides/imunologia , Anticorpos/administração & dosagem , Encéfalo/metabolismo , Imunização Passiva , Transtornos da Memória/imunologia , Transtornos da Memória/terapia , Nootrópicos/administração & dosagem , Peptídeos beta-Amiloides/sangue , Animais , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Infusões Parenterais , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Memória Espacial/efeitos dos fármacos , Memória Espacial/fisiologia
7.
J Neurosci ; 35(16): 6265-76, 2015 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-25904780

RESUMO

Alzheimer's disease (AD) and familial Danish dementia (FDD) are degenerative neurological diseases characterized by amyloid pathology. Normal human sera contain IgG antibodies that specifically bind diverse preamyloid and amyloid proteins and have shown therapeutic potential in vitro and in vivo. We cloned one of these antibodies, 3H3, from memory B cells of a healthy individual using a hybridoma method. 3H3 is an affinity-matured IgG that binds a pan-amyloid epitope, recognizing both Aß and λ Ig light chain (LC) amyloids, which are associated with AD and primary amyloidosis, respectively. The pan-amyloid-binding properties of 3H3 were demonstrated using ELISA, immunohistochemical studies, and competition binding assays. Functional studies showed that 3H3 inhibits both Aß and LC amyloid formation in vitro and abrogates disruption of hippocampal synaptic plasticity by AD-patient-derived soluble Aß in vivo. A 3H3 single-chain variable fragment (scFv) retained the binding specificity of the 3H3 IgG and, when expressed in the brains of transgenic mice using an adeno-associated virus (AAV) vector, decreased parenchymal Aß amyloid deposition in TgCRND8 mice and ADan (Danish Amyloid) cerebral amyloid angiopathy in the mouse model of FDD. These data indicate that naturally occurring human IgGs can recognize a conformational, amyloid-specific epitope and have potent anti-amyloid activities, providing a rationale to test their potential as antibody therapeutics for diverse neurological and other amyloid diseases.


Assuntos
Peptídeos beta-Amiloides/imunologia , Amiloide/metabolismo , Anticorpos Monoclonais/imunologia , Imunoglobulina G/imunologia , Amiloide/efeitos dos fármacos , Animais , Anticorpos Monoclonais/farmacologia , Encéfalo/metabolismo , Catarata/imunologia , Ataxia Cerebelar/imunologia , Angiopatia Amiloide Cerebral/imunologia , Surdez/imunologia , Demência/imunologia , Humanos , Imunoglobulina G/farmacologia , Masculino , Camundongos , Camundongos Transgênicos , Ratos
8.
Alzheimers Dement ; 11(11): 1286-305, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25846299

RESUMO

INTRODUCTION: Much knowledge about amyloid ß (Aß) aggregation and toxicity has been acquired using synthetic peptides and mouse models, whereas less is known about soluble Aß in human brain. METHODS: We analyzed aqueous extracts from multiple AD brains using an array of techniques. RESULTS: Brains can contain at least four different Aß assembly forms including: (i) monomers, (ii) a ∼7 kDa Aß species, and larger species (iii) from ∼30-150 kDa, and (iv) >160 kDa. High molecular weight species are by far the most prevalent and appear to be built from ∼7 kDa Aß species. The ∼7 kDa Aß species resist denaturation by chaotropic agents and have a higher Aß42/Aß40 ratio than monomers, and are unreactive with antibodies to Asp1 of Ab or APP residues N-terminal of Asp1. DISCUSSION: Further analysis of brain-derived ∼7 kDa Aß species, the mechanism by which they assemble and the structures they form should reveal therapeutic and diagnostic opportunities.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Lobo Frontal/metabolismo , Lobo Temporal/metabolismo , Água/metabolismo , Idoso , Idoso de 80 Anos ou mais , Western Blotting , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imunoprecipitação , Masculino , Espectrometria de Massas , Pessoa de Meia-Idade , Peso Molecular , Multimerização Proteica
9.
Neurobiol Aging ; 36(3): 1316-32, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25619661

RESUMO

The amyloid ß (Aß)-protein and microtubule-associated protein, tau, are the major components of the amyloid plaques and neurofibrillary tangles that typify Alzheimer's disease (AD) pathology. As such both Aß and tau have long been proposed as therapeutic targets. Immunotherapy, particularly targeting Aß, is currently the most advanced clinical strategy for treating AD. However, several Aß-directed clinical trials have failed, and there is concern that targeting this protein may not be useful. In contrast, there is a growing optimism that tau immunotherapy may prove more efficacious. Here, for the first time, we studied the effects of chronic administration of an anti-tau monoclonal antibody (5E2) in amyloid precursor protein transgenic mice. For our animal model, we chose the J20 mouse line because prior studies had shown that the cognitive deficits in these mice require expression of tau. Despite the fact that 5E2 was present and active in the brains of immunized mice and that this antibody appeared to engage with extracellular tau, 5E2-treatment did not recover age-dependent spatial reference memory deficits. These results indicate that the memory impairment evident in J20 mice is unlikely to be mediated by a form of extracellular tau recognized by 5E2. In addition to the lack of positive effect of anti-tau immunotherapy, we also documented a significant increase in mortality among J20 mice that received 5E2. Because both the J20 mice used here and tau transgenic mice used in prior tau immunotherapy trials are imperfect models of AD our results recommend extensive preclinical testing of anti-tau antibody-based therapies using multiple mouse models and a variety of different anti-tau antibodies.


Assuntos
Doença de Alzheimer/terapia , Anticorpos Monoclonais/uso terapêutico , Imunoterapia , Proteínas tau/imunologia , Doença de Alzheimer/psicologia , Animais , Modelos Animais de Doenças , Feminino , Masculino , Memória , Camundongos Transgênicos , Falha de Tratamento
10.
Nat Commun ; 5: 3374, 2014 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-24594908

RESUMO

NMDA-type glutamate receptors (NMDARs) are currently regarded as paramount in the potent and selective disruption of synaptic plasticity by Alzheimer's disease amyloid ß-protein (Aß). Non-NMDAR mechanisms remain relatively unexplored. Here we describe how Aß facilitates NMDAR-independent long-term depression of synaptic transmission in the hippocampus in vivo. Synthetic Aß and Aß in soluble extracts of Alzheimer's disease brain usurp endogenous acetylcholine muscarinic receptor-dependent long-term depression, to enable long-term depression that required metabotropic glutamate-5 receptors (mGlu5Rs). We also find that mGlu5Rs are essential for Aß-mediated inhibition of NMDAR-dependent long-term potentiation in vivo. Blocking Aß binding to cellular prion protein with antibodies prevents the facilitation of long-term depression. Our findings uncover an overarching role for Aß-PrP(C)-mGlu5R interplay in mediating both LTD facilitation and LTP inhibition, encompassing NMDAR-mediated processes that were previously considered primary.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Depressão Sináptica de Longo Prazo/fisiologia , Receptor de Glutamato Metabotrópico 5/metabolismo , Animais , Hipocampo/metabolismo , Masculino , Príons/metabolismo , Ratos , Ratos Wistar , Receptor de Glutamato Metabotrópico 5/genética
11.
Philos Trans R Soc Lond B Biol Sci ; 369(1633): 20130147, 2014 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-24298149

RESUMO

Many endogenous factors influence the time course and extent of the detrimental effects of amyloid ß-protein (Aß) on synaptic function. Here, we assessed the impact of varying endogenous glutamatergic and cholinergic transmission by pharmacological means on the disruption of plasticity at hippocampal CA3-to-CA1 synapses in the anaesthetized rat. NMDA receptors (NMDARs) are considered critical in mediating Aß-induced inhibition of long-term potentiation (LTP). However, intracerebroventricular injection of Aß1-42 inhibited not only NMDAR-dependent LTP but also voltage-activated Ca(2+)-dependent LTP induced by strong conditioning stimulation during NMDAR blockade. On the other hand, another form of NMDAR-independent synaptic plasticity, endogenous acetylcholine-induced muscarinic receptor-dependent long-term enhancement, was not hindered by Aß1-42. Interestingly, augmenting endogenous acetylcholine activation of nicotinic receptors prior to the injection of Aß1-42 prevented the inhibition of NMDAR-dependent LTP, whereas the same intervention when introduced after the infusion of Aß was ineffective. We also examined the duration of action of Aß, including water soluble Aß from Alzheimer's disease (AD) brain. Remarkably, the inhibition of LTP induction caused by a single injection of sodium dodecyl sulfate-stable Aß dimer-containing AD brain extract persisted for at least a week. These findings highlight the need to increase our understanding of non-NMDAR mechanisms and of developing novel means of overcoming, rather than just preventing, the deleterious synaptic actions of Aß.


Assuntos
Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/metabolismo , Hipocampo/fisiologia , Potenciação de Longa Duração/fisiologia , Fragmentos de Peptídeos/metabolismo , Sinapses/fisiologia , Acetilcolina/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/farmacologia , Análise de Variância , Animais , Química Encefálica , Potenciais Pós-Sinápticos Excitadores/fisiologia , Líquido Extracelular/metabolismo , Humanos , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Fragmentos de Peptídeos/farmacologia , Ratos , Ratos Wistar , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores Nicotínicos/metabolismo , Sinapses/efeitos dos fármacos
12.
Mol Brain ; 6: 47, 2013 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-24284042

RESUMO

BACKGROUND: Exosomes, small extracellular vesicles of endosomal origin, have been suggested to be involved in both the metabolism and aggregation of Alzheimer's disease (AD)-associated amyloid ß-protein (Aß). Despite their ubiquitous presence and the inclusion of components which can potentially interact with Aß, the role of exosomes in regulating synaptic dysfunction induced by Aß has not been explored. RESULTS: We here provide in vivo evidence that exosomes derived from N2a cells or human cerebrospinal fluid can abrogate the synaptic-plasticity-disrupting activity of both synthetic and AD brain-derived Aß. Mechanistically, this effect involves sequestration of synaptotoxic Aß assemblies by exosomal surface proteins such as PrPC rather than Aß proteolysis. CONCLUSIONS: These data suggest that exosomes can counteract the inhibitory action of Aß, which contributes to perpetual capability for synaptic plasticity.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Exossomos/metabolismo , Plasticidade Neuronal , Sinapses/metabolismo , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/fisiopatologia , Líquido Cefalorraquidiano/metabolismo , Difusão , Exossomos/efeitos dos fármacos , Feminino , Humanos , Ligantes , Potenciação de Longa Duração , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Plasticidade Neuronal/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Proteínas PrPC/metabolismo , Ratos , Ratos Wistar , Sinapses/efeitos dos fármacos
13.
Neurobiol Aging ; 34(5): 1315-27, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23182244

RESUMO

Aggregation of the amyloid ß-protein (Aß) is believed to play a central role in initiating the molecular cascade that culminates in Alzheimer-type dementia (AD), a disease which in its early stage is characterized by synaptic loss and impairment of episodic memory. Here we show that intracerebroventricular injection of Aß-containing water-soluble extracts of AD brain inhibits consolidation of the memory of avoidance learning in the rat and that this effect is highly dependent on the interval between learning and administration. When injected at 1 hour post training extracts from 2 different AD brains significantly impaired recall tested at 48 hours. Ultrastructural examination of hippocampi from animals perfused after 48 hours revealed that Aß-mediated impairment of avoidance memory was associated with lower density of synapses and altered synaptic structure in the dentate gyrus and CA1 fields. These behavioral and ultrastructural data suggest that human brain-derived Aß impairs formation of long-term memory by compromising the structural plasticity essential for consolidation and that Aß targets processes initiated very early in the consolidation pathway.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/farmacologia , Aprendizagem da Esquiva/efeitos dos fármacos , Encéfalo/metabolismo , Plasticidade Neuronal/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Idoso , Idoso de 80 Anos ou mais , Animais , Feminino , Humanos , Masculino , Memória Episódica , Ratos , Ratos Wistar
14.
J Neurosci ; 31(20): 7259-63, 2011 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-21593310

RESUMO

Synthetic amyloid-ß protein (Aß) oligomers bind with high affinity to cellular prion protein (PrP(C)), but the role of this interaction in mediating the disruption of synaptic plasticity by such soluble Aß in vitro is controversial. Here we report that intracerebroventricular injection of Aß-containing aqueous extracts of Alzheimer's disease (AD) brain robustly inhibits long-term potentiation (LTP) without significantly affecting baseline excitatory synaptic transmission in the rat hippocampus in vivo. Moreover, the disruption of LTP was abrogated by immunodepletion of Aß. Importantly, intracerebroventricular administration of antigen-binding antibody fragment D13, directed to a putative Aß-binding site on PrP(C), prevented the inhibition of LTP by AD brain-derived Aß. In contrast, R1, a Fab directed to the C terminus of PrP(C), a region not implicated in binding of Aß, did not significantly affect the Aß-mediated inhibition of LTP. These data support the pathophysiological significance of SDS-stable Aß dimer and the role of PrP(C) in mediating synaptic plasticity disruption by soluble Aß.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/administração & dosagem , Hipocampo/patologia , Potenciação de Longa Duração/fisiologia , Inibição Neural/imunologia , Fragmentos de Peptídeos/administração & dosagem , Proteínas PrPC/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Animais , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Feminino , Hipocampo/metabolismo , Humanos , Injeções Intraventriculares , Masculino , Inibição Neural/fisiologia , Plasticidade Neuronal/fisiologia , Proteínas PrPC/imunologia , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA