Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
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
2.
Nature ; 564(7736): 415-419, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30546139

RESUMO

We previously reported1 the presence of amyloid-ß protein (Aß) deposits in individuals with Creutzfeldt-Jakob disease (CJD) who had been treated during childhood with human cadaveric pituitary-derived growth hormone (c-hGH) contaminated with prions. The marked deposition of parenchymal and vascular Aß in these relatively young individuals with treatment-induced (iatrogenic) CJD (iCJD), in contrast to other prion-disease patients and population controls, allied with the ability of Alzheimer's disease brain homogenates to seed Aß deposition in laboratory animals, led us to argue that the implicated c-hGH batches might have been contaminated with Aß seeds as well as with prions. However, this was necessarily an association, and not an experimental, study in humans and causality could not be concluded. Given the public health importance of our hypothesis, we proceeded to identify and biochemically analyse archived vials of c-hGH. Here we show that certain c-hGH batches to which patients with iCJD and Aß pathology were exposed have substantial levels of Aß40, Aß42 and tau proteins, and that this material can seed the formation of Aß plaques and cerebral Aß-amyloid angiopathy in intracerebrally inoculated mice expressing a mutant, humanized amyloid precursor protein. These results confirm the presence of Aß seeds in archived c-hGH vials and are consistent with the hypothesized iatrogenic human transmission of Aß pathology. This experimental confirmation has implications for both the prevention and the treatment of Alzheimer's disease, and should prompt a review of the risk of iatrogenic transmission of Aß seeds by medical and surgical procedures long recognized to pose a risk of accidental prion transmission2,3.


Assuntos
Doença de Alzheimer/induzido quimicamente , Peptídeos beta-Amiloides/metabolismo , Cadáver , Síndrome de Creutzfeldt-Jakob/induzido quimicamente , Contaminação de Medicamentos , Hormônio do Crescimento/farmacologia , Doença Iatrogênica , Doença de Alzheimer/etiologia , Peptídeos beta-Amiloides/análise , Precursor de Proteína beta-Amiloide/administração & dosagem , Precursor de Proteína beta-Amiloide/efeitos adversos , Animais , Estudos de Casos e Controles , Síndrome de Creutzfeldt-Jakob/etiologia , Modelos Animais de Doenças , Transmissão de Doença Infecciosa/prevenção & controle , Transmissão de Doença Infecciosa/estatística & dados numéricos , Contaminação de Medicamentos/prevenção & controle , Contaminação de Medicamentos/estatística & dados numéricos , Feminino , Hormônio do Crescimento/administração & dosagem , Humanos , Masculino , Camundongos , Modelos Biológicos , Príons/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Reprodutibilidade dos Testes , Proteínas tau/análise , Proteínas tau/metabolismo
3.
Nat Chem Biol ; 17(3): 317-325, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33432237

RESUMO

Epitope-specific enzymes are powerful tools for site-specific protein modification but generally require genetic manipulation of the target protein. Here, we describe the laboratory evolution of the bacterial transpeptidase sortase A to recognize the LMVGG sequence in endogenous amyloid-ß (Aß) protein. Using a yeast display selection for covalent bond formation, we evolved a sortase variant that prefers LMVGG substrates from a starting enzyme that prefers LPESG substrates, resulting in a >1,400-fold change in substrate preference. We used this evolved sortase to label endogenous Aß in human cerebrospinal fluid, enabling the detection of Aß with sensitivities rivaling those of commercial assays. The evolved sortase can conjugate a hydrophilic peptide to Aß42, greatly impeding the ability of the resulting protein to aggregate into higher-order structures. These results demonstrate laboratory evolution of epitope-specific enzymes toward endogenous targets as a strategy for site-specific protein modification without target gene manipulation and enable potential future applications of sortase-mediated labeling of Aß peptides.


Assuntos
Aminoaciltransferases/farmacologia , Peptídeos beta-Amiloides/química , Proteínas de Bactérias/farmacologia , Cisteína Endopeptidases/farmacologia , Fragmentos de Peptídeos/química , Agregados Proteicos/efeitos dos fármacos , Sequência de Aminoácidos , Aminoaciltransferases/química , Aminoaciltransferases/metabolismo , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Cisteína Endopeptidases/química , Cisteína Endopeptidases/metabolismo , Evolução Molecular Direcionada , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Especificidade por Substrato , Técnicas do Sistema de Duplo-Híbrido
4.
Ann Neurol ; 88(5): 878-892, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32829532

RESUMO

OBJECTIVE: There is an urgent need for sensitive, widely available, blood-based screening tests to identify presymptomatic individuals destined to develop Alzheimer's disease (AD). We investigated whether tau detected in plasma by our in-house NT1 assay is specifically altered in AD, and when applied to patients with subjective cognitive decline (SCD) or mild cognitive impairment (MCI) can serve to predict progression to AD dementia. The predictive value of NT1 versus tau measured using assays from Quanterix and Roche, and the specificity of NT1 for AD versus a nonspecific marker of neurodegeneration (neurofilament light [NfL]) were also examined. METHODS: NT1 tau and NfL were measured in plasma from prospectively followed patients with SCD or MCI who remained cognitively stable, converted to AD dementia, or converted to non-AD dementias, and in cognitively unimpaired participants. Tau was measured using Quanterix and Roche assays in baseline subjects with SCD and MCI. RESULTS: Plasma NT1 tau was specifically elevated in AD, but not in non-AD dementia compared with controls, whereas NfL was increased in both AD and non-AD dementias. Baseline specimens from individuals who had SCD or MCI revealed that NT1 tau, but not tau measured using Quanterix or Roche assays, is elevated in subjects who progress to AD dementia. As expected, baseline plasma NfL is elevated in those who progress to AD and non-AD dementias. INTERPRETATION: Plasma NT1 tau is a specific marker of AD, which is elevated early in disease and may prove useful as a first round screen to identify individuals at risk of developing AD. ANN NEUROL 2020;88:878-892.


Assuntos
Doença de Alzheimer/sangue , Biomarcadores/sangue , Marcadores Genéticos , Proteínas tau/sangue , Idade de Início , Idoso , Doença de Alzheimer/líquido cefalorraquidiano , Biomarcadores/líquido cefalorraquidiano , Disfunção Cognitiva/sangue , Disfunção Cognitiva/genética , Estudos de Coortes , Progressão da Doença , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Doenças Neurodegenerativas/sangue , Doenças Neurodegenerativas/genética , Proteínas de Neurofilamentos/sangue , Proteínas tau/líquido cefalorraquidiano , Proteínas tau/genética
5.
Nat Rev Neurosci ; 17(4): 251-60, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26988744

RESUMO

There has been an explosion in the number of papers discussing the hypothesis of 'pathogenic spread' in neurodegenerative disease - the idea that abnormal forms of disease-associated proteins, such as tau or α-synuclein, physically move from neuron to neuron to induce disease progression. However, whether inter-neuronal spread of protein aggregates actually occurs in humans and, if so, whether it causes symptom onset remain uncertain. Even if pathogenic spread is proven in humans, it is unclear how much this would alter the specific therapeutic approaches that are in development. A critical appraisal of this increasingly popular hypothesis thus seems both important and timely.


Assuntos
Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Neurônios/patologia , Agregação Patológica de Proteínas/etiologia , Animais , Humanos , Doenças Neurodegenerativas/complicações , Transporte Proteico , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo
6.
Brain ; 143(11): 3234-3241, 2020 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-33068398

RESUMO

Plasma levels of tau phosphorylated at threonine-217 (p-tau217) is a candidate tool to monitor Alzheimer's disease. We studied 150 cognitively unimpaired participants and 100 patients with mild cognitive impairment in the Swedish BioFINDER study. P-tau217 was measured repeatedly for up to 6 years (median three samples per person, median time from first to last sample, 4.3 years). Preclinical (amyloid-ß-positive cognitively unimpaired, n = 62) and prodromal (amyloid-ß-positive mild cognitive impairment, n = 49) Alzheimer's disease had accelerated p-tau217 compared to amyloid-ß-negative cognitively unimpaired (ß = 0.56, P < 0.001, using linear mixed effects models) and amyloid-ß-negative mild cognitive impairment patients (ß = 0.67, P < 0.001), respectively. Mild cognitive impairment patients who later converted to Alzheimer's disease dementia (n = 40) had accelerated p-tau217 compared to other mild cognitive impairment patients (ß = 0.79, P < 0.001). P-tau217 did not change in amyloid-ß-negative participants, or in patients with mild cognitive impairment who did not convert to Alzheimer's disease dementia. For 80% power, 109 participants per arm were required to observe a slope reduction in amyloid-ß-positive cognitively unimpaired (71 participants per arm in amyloid-ß-positive mild cognitive impairment). Longitudinal increases in p-tau217 correlated with longitudinal worsening of cognition and brain atrophy. In summary, plasma p-tau217 increases during early Alzheimer's disease and can be used to monitor disease progression.


Assuntos
Doença de Alzheimer/sangue , Proteínas tau/sangue , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/psicologia , Peptídeos beta-Amiloides/sangue , Biomarcadores/sangue , Disfunção Cognitiva/sangue , Disfunção Cognitiva/psicologia , Progressão da Doença , Feminino , Humanos , Estudos Longitudinais , Masculino , Testes de Estado Mental e Demência , Pessoa de Meia-Idade , Testes Neuropsicológicos , Fosforilação , Estudos Prospectivos , Treonina/metabolismo
7.
Ann Neurol ; 86(2): 215-224, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31168802

RESUMO

OBJECTIVE: Oligomeric forms of amyloid ß protein (oAß) are believed to be principally responsible for neurotoxicity in Alzheimer disease (AD), but it is not known whether anti-Aß antibodies are capable of lowering oAß levels in humans. METHODS: We developed an ultrasensitive immunoassay and used it to measure oAß in cerebrospinal fluid (CSF) from 104 AD subjects participating in the ABBY and BLAZE phase 2 trials of the anti-Aß antibody crenezumab. Patients received subcutaneous (SC) crenezumab (300mg) or placebo every 2 weeks, or intravenous (IV) crenezumab (15mg/kg) or placebo every 4 weeks for 68 weeks. Ninety-eight of the 104 patients had measurable baseline oAß levels, and these were compared to levels at week 69 in placebo (n = 28), SC (n = 35), and IV (n = 35) treated patients. RESULTS: Among those receiving crenezumab, 89% of SC and 86% of IV patients had lower levels of oAß at week 69 versus baseline. The difference in the proportion of patients with decreasing levels was significant for both treatment arms: p = 0.0035 for SC and p = 0.01 for IV crenezumab versus placebo. The median percentage change was -48% in the SC arm and -43% in the IV arm. No systematic change was observed in the placebo group, with a median change of -13% and equivalent portions with negative and positive change. INTERPRETATION: Crenezumab lowered CSF oAß levels in the large majority of treated patients tested. These results support engagement of the principal pathobiological target in AD and identify CSF oAß as a novel pharmacodynamic biomarker for use in trials of anti-Aß agents. ANN NEUROL 2019;86:215-224.


Assuntos
Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Anticorpos Monoclonais Humanizados/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/líquido cefalorraquidiano , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
8.
Acta Neuropathol ; 139(3): 503-526, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31853635

RESUMO

Neurodegenerative diseases are an enormous public health problem, affecting tens of millions of people worldwide. Nearly all of these diseases are characterized by oligomerization and fibrillization of neuronal proteins, and there is great interest in therapeutic targeting of these aggregates. Here, we show that soluble aggregates of α-synuclein and tau bind to plate-immobilized PrP in vitro and on mouse cortical neurons, and that this binding requires at least one of the same N-terminal sites at which soluble Aß aggregates bind. Moreover, soluble aggregates of tau, α-synuclein and Aß cause both functional (impairment of LTP) and structural (neuritic dystrophy) compromise and these deficits are absent when PrP is ablated, knocked-down, or when neurons are pre-treated with anti-PrP blocking antibodies. Using an all-human experimental paradigm involving: (1) isogenic iPSC-derived neurons expressing or lacking PRNP, and (2) aqueous extracts from brains of individuals who died with Alzheimer's disease, dementia with Lewy bodies, and Pick's disease, we demonstrate that Aß, α-synuclein and tau are toxic to neurons in a manner that requires PrPC. These results indicate that PrP is likely to play an important role in a variety of late-life neurodegenerative diseases and that therapeutic targeting of PrP, rather than individual disease proteins, may have more benefit for conditions which involve the aggregation of more than one protein.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Príons/metabolismo , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo , Animais , Encéfalo/metabolismo , Humanos , Camundongos , Ligação Proteica
9.
Brain ; 142(5): 1441-1457, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31032851

RESUMO

The primary structure of canonical amyloid-ß-protein was elucidated more than 30 years ago, yet the forms of amyloid-ß that play a role in Alzheimer's disease pathogenesis remain poorly defined. Studies of Alzheimer's disease brain extracts suggest that amyloid-ß, which migrates on sodium dodecyl sulphate polyacrylamide gel electrophoresis with a molecular weight of ∼7 kDa (7kDa-Aß), is particularly toxic; however, the nature of this species has been controversial. Using sophisticated mass spectrometry and sensitive assays of disease-relevant toxicity we show that brain-derived bioactive 7kDa-Aß contains a heterogeneous mixture of covalently cross-linked dimers in the absence of any other detectable proteins. The identification of amyloid-ß dimers may open a new phase of Alzheimer's research and allow a better understanding of Alzheimer's disease, and how to monitor and treat this devastating disorder. Future studies investigating the bioactivity of individual dimers cross-linked at known sites will be critical to this effort.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Multimerização Proteica/fisiologia , Idoso , Idoso de 80 Anos ou mais , Peptídeos beta-Amiloides/análise , Animais , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Órgãos
10.
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
11.
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
12.
Alzheimers Dement ; 15(3): 487-496, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30419228

RESUMO

INTRODUCTION: The tau protein plays a central role in Alzheimer's disease (AD), and there is huge interest in measuring tau in blood and cerebrospinal fluid (CSF). METHODS: We developed a set of immunoassays to measure tau in specimens from humans diagnosed based on current best clinical and CSF biomarker criteria. RESULTS: In CSF, mid-region- and N-terminal-detected tau predominated and rose in disease. In plasma, an N-terminal assay (NT1) detected elevated levels of tau in AD and AD-mild cognitive impairment (MCI). Plasma NT1 measurements separated controls from AD-MCI (area under the curve [AUC] = 0.88) and AD (AUC = 0.96) in a discovery cohort and in a Validation Cohort (with AUCs = 0.79 and 0.75, respectively). DISCUSSION: The forms of tau in CSF and plasma are distinct, but in each specimen type, the levels of certain fragments are increased in AD. Measurement of plasma NT1 tau should be aggressively pursued as a potential blood-based screening test for AD/AD-MCI.


Assuntos
Doença de Alzheimer/sangue , Disfunção Cognitiva/sangue , Imunoensaio , Proteínas tau/sangue , Idoso , Doença de Alzheimer/líquido cefalorraquidiano , Biomarcadores/sangue , Biomarcadores/líquido cefalorraquidiano , Disfunção Cognitiva/líquido cefalorraquidiano , Estudos de Coortes , Diagnóstico Diferencial , Espaço Extracelular , Feminino , Humanos , Imunoensaio/métodos , Masculino , Pessoa de Meia-Idade , Sensibilidade e Especificidade , Proteínas tau/líquido cefalorraquidiano
13.
J Neurosci ; 37(1): 152-163, 2017 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-28053038

RESUMO

Soluble oligomers of amyloid ß-protein (oAß) isolated from the brains of Alzheimer's disease (AD) patients have been shown experimentally (in the absence of amyloid plaques) to impair hippocampal synaptic plasticity, decrease synapses, induce tau hyperphosphorylation and neuritic dystrophy, activate microglial inflammation, and impair memory in normal adult rodents. Nevertheless, there has been controversy about what types of oligomers actually confer these AD-like phenotypes. Here, we show that the vast majority of soluble Aß species obtained from brains of humans who died with confirmed AD elute at high molecular weight (HMW) on nondenaturing size-exclusion chromatography. These species have little or no cytotoxic activity in several bioassays. However, incubation of HMW oAß in mildly alkaline buffer led to their quantitative dissociation into low molecular weight oligomers (∼8-70 kDa), and these were now far more bioactive: they impaired hippocampal LTP, decreased neuronal levels of ß2-adrenergic receptors, and activated microglia in wt mice in vivo Thus, most soluble Aß assemblies in AD cortex are large and inactive but under certain circumstances can dissociate into smaller, highly bioactive species. Insoluble amyloid plaques likely sequester soluble HMW oligomers, limiting their potential to dissociate. We conclude that conditions that destabilize HMW oligomers or retard the sequestration of their smaller, more bioactive components are important drivers of Aß toxicity. Selectively targeting these small, cytotoxic forms should be therapeutically beneficial. SIGNIFICANCE STATEMENT: Oligomers of amyloid ß-protein (oAß) are tought to play an important role in Alzheimer's disease (AD), but there is confusion and controversy about what types and sizes of oligomers have disease-relevant activity. Using size-exclusion chromatography and three distinct measures of bioactivity, we show that the predominant forms of Aß in aqueous extracts of AD brain are high molecular weight (HMW) and relatively inactive. Importantly, under certain conditions, the abundant HMW oAß can dissociate into low molecular weight species, and these low molecular weight oligomers are significantly more bioactive on synapses and microglia than the HMW species from which they are derived. We conclude that conditions that destabilize HMW oAß or retard the sequestration of smaller, more bioactive components are important drivers of Aß toxicity.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/toxicidade , Química Encefálica , Animais , Cromatografia em Gel , Feminino , Hipocampo/fisiopatologia , Humanos , Técnicas In Vitro , Potenciação de Longa Duração , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peso Molecular , Receptores Adrenérgicos beta 2/metabolismo , Sinaptossomos/metabolismo
14.
J Neurosci ; 37(49): 11947-11966, 2017 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-29101243

RESUMO

Compelling genetic evidence links the amyloid precursor protein (APP) to Alzheimer's disease (AD) and several theories have been advanced to explain the relationship. A leading hypothesis proposes that a small amphipathic fragment of APP, the amyloid ß-protein (Aß), self-associates to form soluble aggregates that impair synaptic and network activity. Here, we used the most disease-relevant form of Aß, protein isolated from AD brain. Using this material, we show that the synaptotoxic effects of Aß depend on expression of APP and that the Aß-mediated impairment of synaptic plasticity is accompanied by presynaptic effects that disrupt the excitatory/inhibitory (E/I) balance. The net increase in the E/I ratio and inhibition of plasticity are associated with Aß localizing to synapses and binding of soluble Aß aggregates to synapses requires the expression of APP. Our findings indicate a role for APP in AD pathogenesis beyond the generation of Aß and suggest modulation of APP expression as a therapy for AD.SIGNIFICANCE STATEMENT Here, we report on the plasticity-disrupting effects of amyloid ß-protein (Aß) isolated from Alzheimer's disease (AD) brain and the requirement of amyloid precursor protein (APP) for these effects. We show that Aß-containing AD brain extracts block hippocampal LTP, augment glutamate release probability, and disrupt the excitatory/inhibitory balance. These effects are associated with Aß localizing to synapses and genetic ablation of APP prevents both Aß binding and Aß-mediated synaptic dysfunctions. Our results emphasize the importance of APP in AD and should stimulate new studies to elucidate APP-related targets suitable for pharmacological manipulation.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/biossíntese , Encéfalo/metabolismo , Plasticidade Neuronal/fisiologia , Fragmentos de Peptídeos/metabolismo , Sinapses/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/deficiência , Animais , Encéfalo/patologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Técnicas de Cultura de Órgãos , Ligação Proteica/fisiologia , Sinapses/patologia
15.
Acta Neuropathol ; 136(1): 19-40, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29687257

RESUMO

Significant data suggest that soluble Aß oligomers play an important role in Alzheimer's disease (AD), but there is great confusion over what exactly constitutes an Aß oligomer and which oligomers are toxic. Most studies have utilized synthetic Aß peptides, but the relevance of these test tube experiments to the conditions that prevail in AD is uncertain. A few groups have studied Aß extracted from human brain, but they employed vigorous tissue homogenization which is likely to release insoluble Aß that was sequestered in plaques during life. Several studies have found such extracts to possess disease-relevant activity and considerable efforts are being made to purify and better understand the forms of Aß therein. Here, we compared the abundance of Aß in AD extracts prepared by traditional homogenization versus using a far gentler extraction, and assessed their bioactivity via real-time imaging of iPSC-derived human neurons plus the sensitive functional assay of long-term potentiation. Surprisingly, the amount of Aß retrieved by gentle extraction constituted only a small portion of that released by traditional homogenization, but this readily diffusible fraction retained all of the Aß-dependent neurotoxic activity. Thus, the bulk of Aß extractable from AD brain was innocuous, and only the small portion that was aqueously diffusible caused toxicity. This unexpected finding predicts that generic anti-oligomer therapies, including Aß antibodies now in trials, may be bound up by the large pool of inactive oligomers, whereas agents that specifically target the small pool of diffusible, bioactive Aß would be more useful. Furthermore, our results indicate that efforts to purify and target toxic Aß must employ assays of disease-relevant activity. The approaches described here should enable these efforts, and may assist the study of other disease-associated aggregation-prone proteins.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Fragmentos de Peptídeos/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/líquido cefalorraquidiano , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Células CHO , Cricetulus , Feminino , Humanos , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/genética , Potenciação de Longa Duração/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Peso Molecular , Mutação/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fragmentos de Peptídeos/farmacologia , Células-Tronco Pluripotentes/metabolismo
16.
Acta Neuropathol ; 136(4): 537-555, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29982852

RESUMO

MicroRNAs (miRNA) regulate fundamental biological processes, including neuronal plasticity, stress response, and survival. Here, we describe a neuroprotective function of miR-132, the miRNA most significantly downregulated in neurons in Alzheimer's disease. We demonstrate that miR-132 protects primary mouse and human wild-type neurons and more vulnerable Tau-mutant neurons against amyloid ß-peptide (Aß) and glutamate excitotoxicity. It lowers the levels of total, phosphorylated, acetylated, and cleaved forms of Tau implicated in tauopathies, promotes neurite elongation and branching, and reduces neuronal death. Similarly, miR-132 attenuates PHF-Tau pathology and neurodegeneration, and enhances long-term potentiation in the P301S Tau transgenic mice. The neuroprotective effects are mediated by direct regulation of the Tau modifiers acetyltransferase EP300, kinase GSK3ß, RNA-binding protein Rbfox1, and proteases Calpain 2 and Caspases 3/7. These data suggest miR-132 as a master regulator of neuronal health and indicate that miR-132 supplementation could be of therapeutic benefit for the treatment of Tau-associated neurodegenerative disorders.


Assuntos
MicroRNAs/genética , Transdução de Sinais/genética , Tauopatias/genética , Peptídeos beta-Amiloides/genética , Animais , Morte Celular , Ácido Glutâmico/toxicidade , Humanos , Camundongos , Camundongos Transgênicos , MicroRNAs/fisiologia , Mutação/genética , Degeneração Neural/genética , Degeneração Neural/patologia , Neuritos/patologia , Neurônios/patologia , Cultura Primária de Células , Processamento de Proteína Pós-Traducional , RNA Longo não Codificante/genética , Proteínas tau/genética
17.
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
18.
Int J Mol Sci ; 19(3)2018 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-29495441

RESUMO

Progressive cerebral accumulation of tau aggregates is a defining feature of Alzheimer's disease (AD). A popular theory that seeks to explain the apparent spread of neurofibrillary tangle pathology proposes that aggregated tau is passed from neuron to neuron. Such a templated seeding process requires that the transferred tau contains the microtubule binding repeat domains that are necessary for aggregation. While it is not clear how a protein such as tau can move from cell to cell, previous reports have suggested that this may involve extracellular vesicles (EVs). Thus, measurement of tau in EVs may both provide insights on the molecular pathology of AD and facilitate biomarker development. Here, we report the use of sensitive immunoassays specific for full-length (FL) tau and mid-region tau, which we applied to analyze EVs from human induced pluripotent stem cell (iPSC)-derived neuron (iN) conditioned media, cerebrospinal fluid (CSF), and plasma. In each case, most tau was free-floating with a small component inside EVs. The majority of free-floating tau detected by the mid-region assay was not detected by our FL assays, indicating that most free-floating tau is truncated. Inside EVs, the mid-region assay also detected more tau than the FL assay, but the ratio of FL-positive to mid-region-positive tau was higher inside exosomes than in free solution. These studies demonstrate the presence of minute amounts of free-floating and exosome-contained FL tau in human biofluids. Given the potential for FL tau to aggregate, we conclude that further investigation of these pools of extracellular tau and how they change during disease is merited.


Assuntos
Vesículas Extracelulares/metabolismo , Neurônios/metabolismo , Agregados Proteicos , Agregação Patológica de Proteínas , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Alelos , Doença de Alzheimer/sangue , Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Apolipoproteínas E/genética , Biomarcadores/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Estudos de Casos e Controles , Diferenciação Celular , Disfunção Cognitiva/sangue , Disfunção Cognitiva/líquido cefalorraquidiano , Disfunção Cognitiva/metabolismo , Exossomos/metabolismo , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Neurônios/citologia , Proteínas tau/líquido cefalorraquidiano
19.
J Neurosci ; 35(30): 10851-65, 2015 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-26224867

RESUMO

Recent evidence suggests that tau aggregation may spread via extracellular release and subsequent uptake by synaptically connected neurons, but little is known about the processes by which tau is released or the molecular forms of extracellular tau. To gain insight into the nature of extracellular tau, we used highly sensitive ELISAs, which, when used in tandem, are capable of differentiating between full-length (FL) tau, mid-region-bearing fragments, and C-terminal (CT) fragments. We applied these assays to the systematic study of the conditioned media of N2a cells, induced pluripotent stem cell-derived human cortical neurons, and primary rat cortical neurons, each of which was carefully assessed for viability. In all three neuronal models, the bulk of extracellular tau was free-floating and unaggregated and <0.2% was encapsulated in exosomes. Although most intracellular tau was FL, the majority of extracellular tau was CT truncated and appeared to be released both actively by living neurons and passively by dead cells. In contrast, only a small amount of extracellular tau was aggregation-competent tau (i.e., contained the microtubule-binding regions) and this material appears to be released solely due to a low level of cell death that occurs in all cell culture systems. Importantly, amyloid ß-protein (Aß)-induced neuronal compromise significantly increased the quantity of all forms of extracellular tau, but the presence of Aß before detectable cell compromise did not increase extracellular tau. Collectively, these results suggest that factors that induce neuronal death are likely to be necessary to initiate the extracellular spread of tau aggregation. SIGNIFICANCE STATEMENT: Recent studies suggest that the transfer of tau between neurons underlies the characteristic spatiotemporal progression of neurofibrillary pathology. We searched for tau in the conditioned medium of N2a cells, induced pluripotent stem cell-derived human cortical neurons, and primary rat cortical neurons and analyzed the material present using four different tau ELISAs. We demonstrate that the majority of tau released from healthy neurons is C-terminally truncated and lacks the microtubule-binding region (MTBR) thought necessary for self-aggregation. A small amount of MTBR-containing tau is present outside of cells, but this appears to be solely due to cell death. Therefore, if propagation of tau aggregation is mediated by extracellular tau, our findings suggest that neuronal compromise is required to facilitate this process.


Assuntos
Neurônios/metabolismo , Fragmentos de Peptídeos/metabolismo , Proteínas tau/química , Proteínas tau/metabolismo , Animais , Morte Celular/fisiologia , Linhagem Celular , Meios de Cultivo Condicionados/química , Ensaio de Imunoadsorção Enzimática , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Neurais/metabolismo , Ratos
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA