Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Philos Trans R Soc Lond B Biol Sci ; 379(1906): 20230234, 2024 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-38853565

RESUMO

How the two pathognomonic proteins of Alzheimer's disease (AD); amyloid ß (Aß) and tau, cause synaptic failure remains enigmatic. Certain synthetic and recombinant forms of these proteins are known to act concurrently to acutely inhibit long-term potentiation (LTP). Here, we examined the effect of early amyloidosis on the acute disruptive action of synaptotoxic tau prepared from recombinant protein and tau in patient-derived aqueous brain extracts. We also explored the persistence of the inhibition of LTP by different synaptotoxic tau preparations. A single intracerebral injection of aggregates of recombinant human tau that had been prepared by either sonication of fibrils (SτAs) or disulfide bond formation (oTau) rapidly and persistently inhibited LTP in rat hippocampus. The threshold for the acute inhibitory effect of oTau was lowered in amyloid precursor protein (APP)-transgenic rats. A single injection of synaptotoxic tau-containing AD or Pick's disease brain extracts also inhibited LTP, for over two weeks. Remarkably, the persistent disruption of synaptic plasticity by patient-derived brain tau was rapidly reversed by a single intracerebral injection of different anti-tau monoclonal antibodies, including one directed to a specific human tau amino acid sequence. We conclude that patient-derived LTP-disrupting tau species persist in the brain for weeks, maintaining their neuroactivity often in concert with Aß. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Encéfalo , Potenciação de Longa Duração , Proteínas tau , Potenciação de Longa Duração/efeitos dos fármacos , Animais , Proteínas tau/metabolismo , Peptídeos beta-Amiloides/metabolismo , Ratos , Humanos , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Ratos Transgênicos , Masculino , Hipocampo/metabolismo , Hipocampo/efeitos dos fármacos
2.
Eur J Neurosci ; 58(6): 3402-3411, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37655756

RESUMO

Non-invasive sensory stimulation in the range of the brain's gamma rhythm (30-100 Hz) is emerging as a new potential therapeutic strategy for the treatment of Alzheimer's disease (AD). Here, we investigated the effect of repeated combined exposure to 40 Hz synchronized sound and light stimuli on hippocampal long-term potentiation (LTP) in vivo in three rat models of early AD. We employed a very complete model of AD amyloidosis, amyloid precursor protein (APP)-overexpressing transgenic McGill-R-Thy1-APP rats at an early pre-plaque stage, systemic treatment of transgenic APP rats with corticosterone modelling certain environmental AD risk factors and, importantly, intracerebral injection of highly disease-relevant AD patient-derived synaptotoxic beta-amyloid and tau in wild-type animals. We found that daily treatment with 40 Hz sensory stimulation for 2 weeks fully abrogated the inhibition of LTP in all three models. Moreover, there was a negative correlation between the magnitude of LTP and the level of active caspase-1 in the hippocampus of transgenic APP animals, which suggests that the beneficial effect of 40 Hz stimulation was dependent on modulation of pro-inflammatory mechanisms. Our findings support ongoing clinical trials of gamma-patterned sensory stimulation in early AD.


Assuntos
Doença de Alzheimer , Animais , Ratos , Doença de Alzheimer/terapia , Plasticidade Neuronal , Potenciação de Longa Duração , Ratos Transgênicos , Precursor de Proteína beta-Amiloide/genética
3.
J Neurosci ; 43(32): 5870-5879, 2023 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-37491315

RESUMO

Amyloid ß protein (Aß) and tau, the two main proteins implicated in causing Alzheimer's disease (AD), are posited to trigger synaptic dysfunction long before significant synaptic loss occurs in vulnerable circuits. Whereas soluble Aß aggregates from AD brain are well recognized potent synaptotoxins, less is known about the synaptotoxicity of soluble tau from AD or other tauopathy patient brains. Minimally manipulated patient-derived aqueous brain extracts contain the more diffusible native forms of these proteins. Here, we explore how intracerebral injection of Aß and tau present in such aqueous extracts of patient brain contribute to disruption of synaptic plasticity in the CA1 area of the male rat hippocampus. Aqueous extracts of certain AD brains acutely inhibited long-term potentiation (LTP) of synaptic transmission in a manner that required both Aß and tau. Tau-containing aqueous extracts of a brain from a patient with Pick's disease (PiD) also impaired LTP, and diffusible tau from either AD or PiD brain lowered the threshold for AD brain Aß to inhibit LTP. Remarkably, the disruption of LTP persisted for at least 2 weeks after a single injection. These findings support a critical role for diffusible tau in causing rapid onset, persistent synaptic plasticity deficits, and promoting Aß-mediated synaptic dysfunction.SIGNIFICANCE STATEMENT The microtubule-associated protein tau forms relatively insoluble fibrillar deposits in the brains of people with neurodegenerative diseases including Alzheimer's and Pick's diseases. More soluble aggregates of disease-associated tau may diffuse between cells and could cause damage to synapses in vulnerable circuits. We prepared aqueous extracts of diseased cerebral cortex and tested their ability to interfere with synaptic function in the brains of live rats. Tau in these extracts rapidly and persistently disrupted synaptic plasticity and facilitated impairments caused by amyloid ß protein, the other major pathologic protein in Alzheimer's disease. These findings show that certain diffusible forms of tau can mediate synaptic dysfunction and may be a target for therapy.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Masculino , Ratos , Animais , Peptídeos beta-Amiloides/metabolismo , Potenciação de Longa Duração , Doença de Alzheimer/metabolismo , Proteínas tau/metabolismo , Plasticidade Neuronal , Sinapses/metabolismo , Hipocampo/metabolismo , Encéfalo/metabolismo
4.
Neural Regen Res ; 18(6): 1213-1219, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36453396

RESUMO

Cognitive decline in Alzheimer's disease correlates with the extent of tau pathology, in particular tau hyperphosphorylation that initially appears in the transentorhinal and related regions of the brain including the hippocampus. Recent evidence indicates that tau hyperphosphorylation caused by either amyloid-ß or long-term depression, a form of synaptic weakening involved in learning and memory, share similar mechanisms. Studies from our group and others demonstrate that long-term depression-inducing low-frequency stimulation triggers tau phosphorylation at different residues in the hippocampus under different experimental conditions including aging. Conversely, certain forms of long-term depression at hippocampal glutamatergic synapses require endogenous tau, in particular, phosphorylation at residue Ser396. Elucidating the exact mechanisms of interaction between tau and long-term depression may help our understanding of the physiological and pathological functions of tau/tau (hyper)phosphorylation. We first summarize experimental evidence regarding tau-long-term depression interactions, followed by a discussion of possible mechanisms by which this interplay may influence the pathogenesis of Alzheimer's disease. Finally, we conclude with some thoughts and perspectives on future research about these interactions.

5.
J Alzheimers Dis ; 89(1): 335-350, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35871344

RESUMO

BACKGROUND: Cognitive decline in Alzheimer's disease (AD) correlates with the extent of tau pathology, in particular tau hyperphosphorylation, which is strongly age-associated. Although elevation of cerebrospinal fluid or blood levels of phosphorylated tau (p-Tau) at residues Thr181 (p-Tau181), Thr217 (p-Tau217), and Thr231 (p-Tau231) are proposed to be particularly sensitive markers of preclinical AD, the generation of p-Tau during brain activity is poorly understood. OBJECTIVE: To study whether the expression levels of p-Tau181, p-Tau217, and p-Tau231 can be enhanced by physiological synaptic long-term depression (LTD) which has been linked to the enhancement of p-Tau in hippocampus. METHODS: In vivo electrophysiology was performed in urethane anesthetized young adult and aged male rats. Low frequency electrical stimulation (LFS) was used to induce LTD at CA3 to CA1 synapses. The expression level of p-Tau and total tau was measured in dorsal hippocampus using immunofluorescent staining and/or western blotting. RESULTS: We found that LFS enhanced p-Tau181 and p-Tau217 in an age-dependent manner in the hippocampus of live rats. In contrast, phosphorylation at residues Thr231, Ser202/Thr205, and Ser396 appeared less sensitive to LFS. Pharmacological antagonism of either N-methyl-D-aspartate or metabotropic glutamate 5 receptors inhibited the elevation of both p-Tau181 and p-Tau217. Targeting the integrated stress response, which increases with aging, using a small molecule inhibitor ISRIB, prevented the enhancement of p-Tau by LFS in aged rats. CONCLUSION: Together, our data provide a novel in vivo means to uncover brain plasticity-related cellular and molecular processes of tau phosphorylation at key sites in health and aging.


Assuntos
Doença de Alzheimer , Depressão , Doença de Alzheimer/líquido cefalorraquidiano , Animais , Biomarcadores/líquido cefalorraquidiano , Masculino , Plasticidade Neuronal , Fosforilação , Ratos , Sinapses/metabolismo , Proteínas tau/metabolismo
6.
Neuropsychopharmacology ; 46(12): 2170-2179, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34188184

RESUMO

Synaptic dysfunction is a likely proximate cause of subtle cognitive impairment in early Alzheimer's disease. Soluble oligomers are the most synaptotoxic forms of amyloid ß-protein (Aß) and mediate synaptic plasticity disruption in Alzheimer's disease amyloidosis. Because the presence and extent of cortisol excess in prodromal Alzheimer's disease predicts the onset of cognitive symptoms we hypothesised that corticosteroids would exacerbate the inhibition of hippocampal synaptic long-term potentiation in a rat model of Alzheimer's disease amyloidosis. In a longitudinal experimental design using freely behaving pre-plaque McGill-R-Thy1-APP male rats, three injections of corticosterone or the glucocorticoid methylprednisolone profoundly disrupted long-term potentiation induced by strong conditioning stimulation for at least 2 months. The same treatments had a transient or no detectible detrimental effect on synaptic plasticity in wild-type littermates. Moreover, corticosterone-mediated cognitive dysfunction, as assessed in a novel object recognition test, was more persistent in the transgenic animals. Evidence for the involvement of pro-inflammatory mechanisms was provided by the ability of the selective the NOD-leucine rich repeat and pyrin containing protein 3 (NLRP3) inflammasome inhibitor Mcc950 to reverse the synaptic plasticity deficit in corticosterone-treated transgenic animals. The marked prolongation of the synaptic plasticity disrupting effects of brief corticosteroid excess substantiates a causal role for hypothalamic-pituitary-adrenal axis dysregulation in early Alzheimer's disease.


Assuntos
Doença de Alzheimer , Amiloidose , Doença de Alzheimer/induzido quimicamente , Peptídeos beta-Amiloides/metabolismo , Animais , Glucocorticoides , Sistema Hipotálamo-Hipofisário/metabolismo , Masculino , Plasticidade Neuronal , Sistema Hipófise-Suprarrenal/metabolismo , Ratos
7.
J Neurochem ; 157(6): 2128-2144, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33583024

RESUMO

Neuronal network dysfunction is a hallmark of Alzheimer's disease (AD). However, the underlying pathomechanisms remain unknown. We analyzed the hippocampal micronetwork in transgenic McGill-R-Thy1-APP rats (APPtg) at the beginning of extracellular amyloid beta (Aß) deposition. We established two-photon Ca2+ -imaging in vivo in the hippocampus of rats and found hyperactivity of CA1 neurons. Patch-clamp recordings in brain slices in vitro revealed increased neuronal input resistance and prolonged action potential width in CA1 pyramidal neurons. We did neither observe changes in synaptic inhibition, nor in excitation. Our data support the view that increased intrinsic excitability of CA1 neurons may precede inhibitory dysfunction at an early stage of Aß-deposition and disease progression.


Assuntos
Doença de Alzheimer/metabolismo , Modelos Animais de Doenças , Potenciais Pós-Sinápticos Excitadores/fisiologia , Hipocampo/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Feminino , Hipocampo/patologia , Masculino , Técnicas de Cultura de Órgãos , Ratos , Ratos Transgênicos
8.
BMJ Open Ophthalmol ; 5(1): e000510, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33024827

RESUMO

The biomedical use of silk as a suture dates back to antiquity. Fibroin is the structural element that determines the strength of silk and here we consider the safety of fibroin in its role in ophthalmology. The high mechanical strength of silk meant sufficiently thin threads could be made for eye microsurgery, but such usage was all but superseded by synthetic polymer sutures, primarily because silk in its entirety was more inflammatory. Significant immunological response can normally be avoided by careful manufacturing to provide high purity fibroin, and it has been utilised in this form for tissue engineering an array of fibre and film substrata deployed in research with cells of the eye. Films of fibroin can also be made transparent, which is a required property in the visual pathway. Transparent layers of corneal epithelial, stromal and endothelial cells have all been demonstrated with maintenance of phenotype, as have constructs supporting retinal cells. Fibroin has a lack of demonstrable infectious agent transfer, an ability to be sterilised and prepared with minimal contamination, long-term predictable degradation and low direct cytotoxicity. However, there remains a known ability to be involved in amyloid formation and potential amyloidosis which, without further examination, is enough to currently question whether fibroin should be employed in the eye given its innervation into the brain.

9.
Front Neurosci ; 13: 861, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31474823

RESUMO

How endogenously produced soluble amyloid ß-protein (Aß) affects synaptic plasticity in vulnerable circuits should provide insight into early Alzheimer's disease pathophysiology. McGill-R-Thy1-APP transgenic rats, modeling Alzheimer's disease amyloidosis, exhibit an age-dependent soluble Aß-mediated impairment of the induction of long-term potentiation (LTP) by 200 Hz conditioning stimulation at apical CA3-to-CA1 synapses. Here, we investigated if synaptic weakening at these synapses in the form of activity-dependent persistent reversal (depotentiation) of LTP is also altered in pre-plaque rats in vivo. In freely behaving transgenic rats strong, 400 Hz, conditioning stimulation induced stable LTP that was NMDA receptor- and voltage-gated Ca2+ channel-dependent. Surprisingly, the ability of novelty exploration to induce depotentiation of 400 Hz-induced LTP was impaired in an Aß-dependent manner in the freely behaving transgenic rats. Moreover, at apical synapses, low frequency conditioning stimulation (1 Hz) did not trigger depotentiation in anaesthetized transgenic rats, with an age-dependence similar to the LTP deficit. In contrast, at basal synapses neither LTP, induced by 100 or 200 Hz, nor novelty exploration-induced depotentiation was impaired in the freely behaving transgenic rats. These findings indicate that activity-dependent weakening, as well as strengthening, is impaired in a synapse- and age-dependent manner in this model of early Alzheimer's disease amyloidosis.

10.
Neurobiol Dis ; 127: 582-590, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30910746

RESUMO

Soluble synaptotoxic aggregates of the main pathological proteins of Alzheimer's disease, amyloid ß-protein (Aß) and tau, have rapid and potent inhibitory effects on long-term potentiation (LTP). Although the promotion of synaptic weakening mechanisms, including long-term depression (LTD), is posited to mediate LTP inhibition by Aß, little is known regarding the action of exogenous tau on LTD. The present study examined the ability of different assemblies of full-length human tau to affect LTD in the dorsal hippocampus of the anaesthetized rat. Unlike Aß, intracerebroventricular injection of soluble aggregates of tau (SτAs), but not monomers or fibrils, potently increased the threshold for LTD induction in a manner that required cellular prion protein. However, MTEP, an antagonist of the putative prion protein coreceptor metabotropic glutamate receptor 5, did not prevent the disruption of synaptic plasticity by SτAs. In contrast, systemic treatment with Ro 25-6981, a selective antagonist at GluN2B subunit-containing NMDA receptors, reduced SτA-mediated inhibition of LTD, but not LTP. Intriguingly, SτAs completely blocked Aß-facilitated LTD, whereas a subthreshold dose of SτAs facilitated Aß-mediated inhibition of LTP. Overall, these findings support the importance of cellular prion protein in mediating a range of, sometimes opposing, actions of soluble Aß and tau aggregates with different effector mechanisms on synaptic plasticity.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Hipocampo/efeitos dos fármacos , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Agregados Proteicos/fisiologia , Proteínas tau/metabolismo , Animais , Hipocampo/metabolismo , Depressão Sináptica de Longo Prazo/fisiologia , Masculino , Piridinas/farmacologia , Ratos , Receptor de Glutamato Metabotrópico 5/agonistas , Sinapses/efeitos dos fármacos , Sinapses/fisiologia , Tiazóis/farmacologia
11.
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
12.
Cell Rep ; 23(7): 1932-1938, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29768194

RESUMO

The early stages of Alzheimer's disease are associated with synaptic dysfunction prior to overt loss of neurons. To identify extracellular molecules that impair synaptic plasticity in the brain, we studied the secretomes of human iPSC-derived neuronal models of Alzheimer's disease. When introduced into the rat brain, secretomes from human neurons with either a presenilin-1 mutation, amyloid precursor protein duplication, or trisomy of chromosome 21 all strongly inhibit hippocampal long-term potentiation. Synaptic dysfunction caused by presenilin-1 mutant and amyloid precusor protein duplication secretomes is mediated by Aß peptides, whereas trisomy of chromosome 21 (trisomy 21) neuronal secretomes induce dysfunction through extracellular tau. In all cases, synaptotoxicity is relieved by antibody blockade of cellular prion protein. These data indicate that human models of Alzheimer's disease generate distinct proteins that converge at the level of cellular prion protein to induce synaptic dysfunction in vivo.


Assuntos
Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/metabolismo , Espaço Extracelular/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Biológicos , Plasticidade Neuronal , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Animais , Genótipo , Humanos , Potenciação de Longa Duração , Masculino , Neurônios/metabolismo , Presenilina-1/metabolismo , Ratos
13.
Neurobiol Dis ; 114: 24-30, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29477641

RESUMO

Pro-inflammatory mechanisms have recently emerged as an important component of early Alzheimer's disease (AD) pathogenesis. A particularly attractive therapeutic strategy is to selectively prevent the disruptive effects of activation of the innate immune system in the brain at an early transitional stage by reducing the production or directly neutralizing pro-inflammatory cytokines, in particular IL-1ß and TNF-α. Here we tested their in vivo effects on synaptic plasticity deficits, which provide sensitive and robust measures of synaptic failure, in a rat model of AD amyloidosis. Using electrophysiological techniques we longitudinally studied the effects of the NLRP3 inflammasome inhibitor Mcc950, the IL-1 receptor antagonist (anakinra) and an anti-TNF-α agent (etanercept) in awake freely moving transgenic rats overexpressing AD associated ß-amyloid precursor protein at a pre-plaque stage of amyloidosis. Repeated treatment with Mcc950 reversibly abrogated the inhibition of long-term potentiation. The IL-1 receptor antagonist and etanercept also had a similar beneficial effect on the deficit in synaptic plasticity. Our findings support the clinical development of Mcc950 and clinically available IL-1- and TNF-α-neutralizing agents in early AD.


Assuntos
Doença de Alzheimer/fisiopatologia , Amiloidose/fisiopatologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Plasticidade Neuronal/fisiologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Amiloidose/tratamento farmacológico , Amiloidose/genética , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Anti-Inflamatórios não Esteroides/uso terapêutico , Etanercepte/farmacologia , Etanercepte/uso terapêutico , Humanos , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Proteína Antagonista do Receptor de Interleucina 1/uso terapêutico , Masculino , Proteína 3 que Contém Domínio de Pirina da Família NLR/agonistas , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Plasticidade Neuronal/efeitos dos fármacos , Ratos , Ratos Transgênicos
14.
Neuropharmacology ; 121: 231-246, 2017 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-28390893

RESUMO

Alzheimer's disease amyloid-ß (Aß) oligomers are synaptotoxic, inappropriately increasing extracellular glutamate concentration and glutamate receptor activation to thereby rapidly disrupt synaptic plasticity. Thus, acutely promoting brain glutamate homeostasis with a blood-based scavenging system, glutamate-oxaloacetate transaminase (GOT), and blocking metabotropic glutamate 5 (mGlu5) receptor or its co-receptor cellular prion protein (PrP), prevent the acute inhibition of long-term potentiation (LTP) by exogenous Aß. Here, we evaluated the time course of the effects of such interventions in the persistent disruptive effects of Aß oligomers, either exogenously injected in wild type rats or endogenously generated in transgenic rats that model Alzheimer's disease amyloidosis. We report that repeated, but not acute, systemic administration of recombinant GOT type 1, with or without the glutamate co-substrate oxaloacetate, reversed the persistent deleterious effect of exogenous Aß on synaptic plasticity. Moreover, similar repetitive treatment reversibly abrogated the inhibition of LTP monitored longitudinally in freely behaving transgenic rats. Remarkably, brief repeated treatment with an mGlu5 receptor antagonist, basimglurant, or an antibody that prevents Aß oligomer binding to PrP, ICSM35, also had similar reversible ameliorative effects in the transgenic rat model. Overall, the present findings support the ongoing development of therapeutics for early Alzheimer's disease based on these complementary approaches.


Assuntos
Amiloidose/patologia , Amiloidose/fisiopatologia , Região CA1 Hipocampal/fisiologia , Potenciação de Longa Duração/efeitos dos fármacos , Proteínas Priônicas/metabolismo , Receptor de Glutamato Metabotrópico 5/metabolismo , Peptídeos beta-Amiloides/imunologia , Peptídeos beta-Amiloides/farmacologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidose/sangue , Amiloidose/genética , Animais , Anticorpos/farmacologia , Aspartato Aminotransferase Citoplasmática/farmacologia , Região CA1 Hipocampal/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Imidazóis/farmacologia , Potenciação de Longa Duração/genética , Masculino , Mutação/genética , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/farmacologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Piridinas/farmacologia , Ratos , Ratos Transgênicos , Receptor de Glutamato Metabotrópico 5/antagonistas & inibidores , Vigília
15.
J Biol Chem ; 290(47): 28343-28352, 2015 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-26221033

RESUMO

Despite significant advances, the molecular identity of the cytotoxic species populated during in vivo amyloid formation crucial for the understanding of neurodegenerative disorders is yet to be revealed. In this study lysozyme prefibrillar oligomers and fibrils in both mature and sonicated states have been isolated through an optimized ultrafiltration/ultracentrifugation method and characterized with various optical spectroscopic techniques, atomic force microscopy, and transmission electron microscopy. We examined their level and mode of toxicity on rat pheochromocytoma (PC12) cells in both differentiated and undifferentiated states. We find that oligomers and fibrils display cytotoxic capabilities toward cultured cells in vitro, with oligomers producing elevated levels of cellular injury toward undifferentiated PC12 cells (PC12(undiff)). Furthermore, dual flow cytometry staining experiments demonstrate that the oligomers and mature fibrils induce divergent cellular death pathways (apoptosis and secondary necrosis, respectively) in these PC12 cells. We have also shown that oligomers but not sonicated mature fibrils inhibit hippocampal long term potentiation, a form of synaptic plasticity implicated in learning and memory, in vivo. We conclude that our in vitro and in vivo findings confer a level of resistance toward amyloid fibrils, and that the PC 12-based comparative cytotoxicity assay can provide insights into toxicity differences between differently aggregated protein species.


Assuntos
Amiloide/metabolismo , Biopolímeros/metabolismo , Morte Celular , Amiloide/química , Animais , Biopolímeros/química , Células PC12 , Ratos
16.
Acta Neuropathol Commun ; 2: 175, 2014 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-25540024

RESUMO

Long before synaptic loss occurs in Alzheimer's disease significant harbingers of disease may be detected at the functional level. Here we examined if synaptic long-term potentiation is selectively disrupted prior to extracellular deposition of Aß in a very complete model of Alzheimer's disease amyloidosis, the McGill-R-Thy1-APP transgenic rat. Longitudinal studies in freely behaving animals revealed an age-dependent, relatively rapid-onset and persistent inhibition of long-term potentiation without a change in baseline synaptic transmission in the CA1 area of the hippocampus. Thus the ability of a standard 200 Hz conditioning protocol to induce significant NMDA receptor-dependent short- and long-term potentiation was lost at about 3.5 months of age and this deficit persisted for at least another 2-3 months, when plaques start to appear. Consistent with in vitro evidence for a causal role of a selective reduction in NMDA receptor-mediated synaptic currents, the deficit in synaptic plasticity in vivo was associated with a reduction in the synaptic burst response to the conditioning stimulation and was overcome using stronger 400 Hz stimulation. Moreover, intracerebroventricular treatment for 3 days with an N-terminally directed monoclonal anti- human Aß antibody, McSA1, transiently reversed the impairment of synaptic plasticity. Similar brief treatment with the BACE1 inhibitor LY2886721 or the γ-secretase inhibitor MRK-560 was found to have a comparable short-lived ameliorative effect when tracked in individual rats. These findings provide strong evidence that endogenously generated human Aß selectively disrupts the induction of long-term potentiation in a manner that enables potential therapeutic options to be assessed longitudinally at the pre-plaque stage of Alzheimer's disease amyloidosis.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Hipocampo/fisiopatologia , Potenciação de Longa Duração/fisiologia , Transmissão Sináptica/fisiologia , Fatores Etários , Doença de Alzheimer/fisiopatologia , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Peptídeos beta-Amiloides/imunologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/imunologia , Animais , Anticorpos/farmacologia , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Comportamento Animal/fisiologia , Modelos Animais de Doenças , Compostos Heterocíclicos com 2 Anéis/farmacologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Ácidos Picolínicos/farmacologia , Ratos , Ratos Transgênicos , Ratos Wistar , Sulfonamidas/farmacologia , Transmissão Sináptica/efeitos dos fármacos
17.
Biochemistry ; 53(24): 3908-21, 2014 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-24840308

RESUMO

Evidence for a central role of amyloid ß-protein (Aß) in the genesis of Alzheimer's disease (AD) has led to advanced human trials of Aß-lowering agents. The "amyloid hypothesis" of AD postulates deleterious effects of small, soluble forms of Aß on synaptic form and function. Because selectively targeting synaptotoxic forms of soluble Aß could be therapeutically advantageous, it is important to understand the full range of soluble Aß derivatives. We previously described a Chinese hamster ovary (CHO) cell line (7PA2 cells) that stably expresses mutant human amyloid precursor protein (APP). Here, we extend this work by purifying an sodium dodecyl sulfate (SDS)-stable, ∼8 kDa Aß species from the 7PA2 medium. Mass spectrometry confirmed its identity as a noncovalently bonded Aß40 homodimer that impaired hippocampal long-term potentiation (LTP) in vivo. We further report the detection of Aß-containing fragments of APP in the 7PA2 medium that extend N-terminal from Asp1 of Aß. These N-terminally extended Aß-containing monomeric fragments are distinct from soluble Aß oligomers formed from Aß1-40/42 monomers and are bioactive synaptotoxins secreted by 7PA2 cells. Importantly, decreasing ß-secretase processing of APP elevated these alternative synaptotoxic APP fragments. We conclude that certain synaptotoxic Aß-containing species can arise from APP processing events N-terminal to the classical ß-secretase cleavage site.


Assuntos
Peptídeos beta-Amiloides/fisiologia , Precursor de Proteína beta-Amiloide/metabolismo , Plasticidade Neuronal , Sinapses/efeitos dos fármacos , Doença de Alzheimer/fisiopatologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/isolamento & purificação , Peptídeos beta-Amiloides/farmacologia , Peptídeos beta-Amiloides/toxicidade , Animais , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Meios de Cultivo Condicionados , Humanos , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Camundongos , Plasticidade Neuronal/efeitos dos fármacos , Fragmentos de Peptídeos , Ratos
18.
J Neurosci ; 34(18): 6140-5, 2014 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-24790184

RESUMO

Alzheimer's disease (AD) is associated with pathological assembly states of amyloid-ß protein (Aß). Aß-related synaptotoxicity can be blocked by anti-prion protein (PrP) antibodies, potentially allowing therapeutic targeting of this aspect of AD neuropathogenesis. Here, we show that intravascular administration of a high-affinity humanized anti-PrP antibody to rats can prevent the plasticity-disrupting effects induced by exposure to soluble AD brain extract. These results provide an in vivo proof of principle for such a therapeutic strategy.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/farmacologia , Anticorpos Monoclonais/administração & dosagem , Região CA1 Hipocampal/efeitos dos fármacos , Potenciação de Longa Duração/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Príons/imunologia , Idoso de 80 Anos ou mais , Análise de Variância , Animais , Biofísica , Vias de Administração de Medicamentos , Estimulação Elétrica , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Masculino , Príons/metabolismo , Ratos , Ratos Wistar , Lobo Temporal/química , Lobo Temporal/metabolismo
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...