Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 298(4): 101774, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35218773

RESUMO

Microtubule-associated protein tau is a naturally unfolded protein that can modulate a vast array of physiological processes through direct or indirect binding with molecular partners. Aberrant tau homeostasis has been implicated in the pathogenesis of several neurodegenerative disorders, including Alzheimer's disease. In this study, we performed an unbiased high-content protein profiling assay by incubating recombinant human tau on microarrays containing thousands of human polypeptides. Among the putative tau-binding partners, we identify SAH hydrolase-like protein 1/inositol 1,4,5-trisphosphate receptor (IP3R)-binding protein (AHCYL1/IRBIT), a member of the SAH hydrolase family and a previously described modulator of IP3R activity. Using coimmunoprecipitation assays, we show that endogenous as well as overexpressed tau can physically interact with AHCYL1/IRBIT in brain tissues and cultured cells. Proximity ligation assay experiments demonstrate that tau overexpression may modify the close localization of AHCYL1/IRBIT to IP3R at the endoplasmic reticulum. Together, our experimental evidence indicates that tau interacts with AHCYL1/IRBIT and potentially modulates AHCYL1/IRBIT function.


Assuntos
Lectinas Tipo C , Proteínas de Membrana , Proteômica , Proteínas tau , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Expressão Gênica , Humanos , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Ligação Proteica , Proteínas tau/genética , Proteínas tau/metabolismo
2.
Int J Mol Sci ; 24(11)2023 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-37298211

RESUMO

The accumulation of tau is a hallmark of several neurodegenerative diseases and is associated with neuronal hypoactivity and presynaptic dysfunction. Oral administration of the adenosine A1 receptor antagonist rolofylline (KW-3902) has previously been shown to reverse spatial memory deficits and to normalize the basic synaptic transmission in a mouse line expressing full-length pro-aggregant tau (TauΔK) at low levels, with late onset of disease. However, the efficacy of treatment remained to be explored for cases of more aggressive tauopathy. Using a combination of behavioral assays, imaging with several PET-tracers, and analysis of brain tissue, we compared the curative reversal of tau pathology by blocking adenosine A1 receptors in three mouse models expressing different types and levels of tau and tau mutants. We show through positron emission tomography using the tracer [18F]CPFPX (a selective A1 receptor ligand) that intravenous injection of rolofylline effectively blocks A1 receptors in the brain. Moreover, when administered to TauΔK mice, rolofylline can reverse tau pathology and synaptic decay. The beneficial effects are also observed in a line with more aggressive tau pathology, expressing the amyloidogenic repeat domain of tau (TauRDΔK) with higher aggregation propensity. Both models develop a progressive tau pathology with missorting, phosphorylation, accumulation of tau, loss of synapses, and cognitive decline. TauRDΔK causes pronounced neurofibrillary tangle assembly concomitant with neuronal death, whereas TauΔK accumulates only to tau pretangles without overt neuronal loss. A third model tested, the rTg4510 line, has a high expression of mutant TauP301L and hence a very aggressive phenotype starting at ~3 months of age. This line failed to reverse pathology upon rolofylline treatment, consistent with a higher accumulation of tau-specific PET tracers and inflammation. In conclusion, blocking adenosine A1 receptors by rolofylline can reverse pathology if the pathological potential of tau remains below a threshold value that depends on concentration and aggregation propensity.


Assuntos
Receptor A1 de Adenosina , Tauopatias , Camundongos , Animais , Camundongos Transgênicos , Receptor A1 de Adenosina/genética , Receptor A1 de Adenosina/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Hipocampo/metabolismo , Tauopatias/tratamento farmacológico , Tauopatias/genética , Tauopatias/metabolismo , Cognição , Modelos Animais de Doenças
3.
J Biol Chem ; 295(52): 18213-18225, 2020 12 25.
Artigo em Inglês | MEDLINE | ID: mdl-33106314

RESUMO

Abnormal changes of neuronal Tau protein, such as phosphorylation and aggregation, are considered hallmarks of cognitive deficits in Alzheimer's disease. Abnormal phosphorylation is thought to precede aggregation and therefore to promote aggregation, but the nature and extent of phosphorylation remain ill-defined. Tau contains ∼85 potential phosphorylation sites, which can be phosphorylated by various kinases because the unfolded structure of Tau makes them accessible. However, methodological limitations (e.g. in MS of phosphopeptides, or antibodies against phosphoepitopes) led to conflicting results regarding the extent of Tau phosphorylation in cells. Here we present results from a new approach based on native MS of intact Tau expressed in eukaryotic cells (Sf9). The extent of phosphorylation is heterogeneous, up to ∼20 phosphates per molecule distributed over 51 sites. The medium phosphorylated fraction Pm showed overall occupancies of ∼8 Pi (± 5) with a bell-shaped distribution; the highly phosphorylated fraction Ph had 14 Pi (± 6). The distribution of sites was highly asymmetric (with 71% of all P-sites in the C-terminal half of Tau). All sites were on Ser or Thr residues, but none were on Tyr. Other known posttranslational modifications were near or below our detection limit (e.g. acetylation, ubiquitination). These findings suggest that normal cellular Tau shows a remarkably high extent of phosphorylation, whereas other modifications are nearly absent. This implies that abnormal phosphorylations at certain sites may not affect the extent of phosphorylation significantly and do not represent hyperphosphorylation. By implication, the pathological aggregation of Tau is not likely a consequence of high phosphorylation.


Assuntos
Cromatografia Líquida/métodos , Processamento de Proteína Pós-Traducional , Espectrometria de Massas em Tandem/métodos , Proteínas tau/química , Proteínas tau/metabolismo , Sequência de Aminoácidos , Humanos , Fosforilação , Homologia de Sequência
4.
Hum Mol Genet ; 28(9): 1498-1514, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30590647

RESUMO

Mutations in the microtubule-associated protein tau (MAPT) underlie multiple neurodegenerative disorders, yet the pathophysiological mechanisms are unclear. A novel variant in MAPT resulting in an alanine to threonine substitution at position 152 (A152T tau) has recently been described as a significant risk factor for both frontotemporal lobar degeneration and Alzheimer's disease. Here we use complementary computational, biochemical, molecular, genetic and imaging approaches in Caenorhabditis elegans and mouse models to interrogate the effects of the A152T variant on tau function. In silico analysis suggests that a threonine at position 152 of tau confers a new phosphorylation site. This finding is borne out by mass spectrometric survey of A152T tau phosphorylation in C. elegans and mouse. Optical pulse-chase experiments of Dendra2-tau demonstrate that A152T tau and phosphomimetic A152E tau exhibit increased diffusion kinetics and the ability to traverse across the axon initial segment more efficiently than wild-type (WT) tau. A C. elegans model of tauopathy reveals that A152T and A152E tau confer patterns of developmental toxicity distinct from WT tau, likely due to differential effects on retrograde axonal transport. These data support a role for phosphorylation of the variant threonine in A152T tau toxicity and suggest a mechanism involving impaired retrograde axonal transport contributing to human neurodegenerative disease.


Assuntos
Alelos , Substituição de Aminoácidos , Variação Genética , Proteínas tau/genética , Proteínas tau/metabolismo , Animais , Animais Geneticamente Modificados , Transporte Axonal , Axônios/metabolismo , Caenorhabditis elegans , Modelos Animais de Doenças , Suscetibilidade a Doenças , Humanos , Camundongos , Mutação , Fosforilação , Ligação Proteica , Vesículas Sinápticas/metabolismo , Tauopatias/etiologia , Tauopatias/metabolismo , Tauopatias/patologia
5.
Adv Exp Med Biol ; 1184: 97-103, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32096031

RESUMO

Mutations in MAPT (Tau) have been implicated in several types of tauopathy, but the pathways leading to neurodegeneration have remained elusive and are heterogeneous. Here we describe the effects of two mutations, both linked to AD or FTD, that are located in different domains of Tau and show different pathways of toxicity. The deletion mutation ΔK280 lies in the repeat domain and strongly increases ß-structure and hence aggregation, whereas the mutation A152T lies in the N-terminal projection domain, has little effect on aggregation but instead on signalling. Both mutations cause presynaptic dysfunction, but in opposite ways, leading to hypoexcitability/hypoactivity vs. hyperexcitability/excitotoxicity, respectively. In organotypic slices these abnormal states can be reversed by drugs, e.g. Tau aggregation inhibitors or modulators of glutamate uptake. This information could contribute to the understanding of "normal" Tau biology and possible therapeutical strategies.


Assuntos
Tauopatias/metabolismo , Tauopatias/fisiopatologia , Proteínas tau/química , Proteínas tau/metabolismo , Ácido Glutâmico/metabolismo , Humanos , Mutação , Domínios Proteicos/genética , Tauopatias/genética , Tauopatias/patologia , Proteínas tau/genética
6.
Proc Natl Acad Sci U S A ; 113(41): 11597-11602, 2016 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-27671637

RESUMO

Accumulation of Tau is a characteristic hallmark of several neurodegenerative diseases but the mode of toxic action of Tau is poorly understood. Here, we show that the Tau protein is toxic due to its aggregation propensity, whereas phosphorylation and/or missorting is not sufficient to cause neuronal dysfunction. Aggregate-prone Tau accumulates, when expressed in vitro at near-endogenous levels, in axons as spindle-shaped grains. These axonal grains contain Tau that is folded in a pathological (MC-1) conformation. Proaggregant Tau induces a reduction of neuronal ATP, concomitant with loss of dendritic spines. Counterintuitively, axonal grains of Tau are not targeted for degradation and do not induce a molecular stress response. Proaggregant Tau causes neuronal and astrocytic hypoactivity and presynaptic dysfunction instead. Here, we show that the adenosine A1 receptor antagonist rolofylline (KW-3902) is alleviating the presynaptic dysfunction and restores neuronal activity as well as dendritic spine levels in vitro. Oral administration of rolofylline for 2-wk to 14-mo-old proaggregant Tau transgenic mice restores the spatial memory deficits and normalizes the basic synaptic transmission. These findings make rolofylline an interesting candidate to combat the hypometabolism and neuronal dysfunction associated with Tau-induced neurodegenerative diseases.


Assuntos
Antagonistas do Receptor A1 de Adenosina/farmacologia , Axônios/metabolismo , Deleção de Sequência , Xantinas/farmacologia , Proteínas tau/genética , Trifosfato de Adenosina/metabolismo , Animais , Espinhas Dendríticas/efeitos dos fármacos , Modelos Animais de Doenças , Imunofluorescência , Expressão Gênica , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Memória de Longo Prazo/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Fosforilação , Agregados Proteicos , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Tauopatias/tratamento farmacológico , Tauopatias/genética , Tauopatias/metabolismo , Tauopatias/patologia
7.
Nano Lett ; 18(5): 3271-3281, 2018 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-29644863

RESUMO

Misfolding and aggregation of the neuronal, microtubule-associated protein tau is involved in the pathogenesis of Alzheimer's disease and tauopathies. It has been proposed that neuronal membranes could play a role in tau release, internalization, and aggregation and that tau aggregates could exert toxicity via membrane permeabilization. Whether and how tau interacts with lipid membranes remains a matter of discussion. Here, we characterize the interaction of full-length human tau (htau40) with supported lipid membranes (SLMs) made from brain total lipid extract by time-lapse high-resolution atomic force microscopy (AFM). We observe that tau attaches to brain lipid membranes where it self-assembles in a cation-dependent manner. Sodium triggers the attachment, self-assembly, and growth, whereas potassium inhibits these processes. Moreover, tau assemblies are stable in the presence of sodium and lithium but disassemble in the presence of potassium and rubidium. Whereas the pseudorepeat domains (R1-R4) of htau40 promote the sodium-dependent attachment to the membrane and stabilize the tau assemblies, the N-terminal region promotes tau self-assembly and growth.


Assuntos
Encéfalo/metabolismo , Membrana Celular/metabolismo , Lipídeos de Membrana/metabolismo , Proteínas tau/metabolismo , Cátions/metabolismo , Humanos , Microscopia de Força Atômica , Agregados Proteicos , Ligação Proteica , Domínios Proteicos , Estabilidade Proteica , Proteínas tau/química
8.
J Biol Chem ; 292(29): 12192-12207, 2017 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-28536263

RESUMO

Subcellular mislocalization of the microtubule-associated protein Tau is a hallmark of Alzheimer disease (AD) and other tauopathies. Six Tau isoforms, differentiated by the presence or absence of a second repeat or of N-terminal inserts, exist in the human CNS, but their physiological and pathological differences have long remained elusive. Here, we investigated the properties and distributions of human and rodent Tau isoforms in primary forebrain rodent neurons. We found that the Tau diffusion barrier (TDB), located within the axon initial segment (AIS), controls retrograde (axon-to-soma) and anterograde (soma-to-axon) traffic of Tau. Tau isoforms without the N-terminal inserts were sorted efficiently into the axon. However, the longest isoform (2N4R-Tau) was partially retained in cell bodies and dendrites, where it accelerated spine and dendrite growth. The TDB (located within the AIS) was impaired when AIS components (ankyrin G, EB1) were knocked down or when glycogen synthase kinase-3ß (GSK3ß; an AD-associated kinase tethered to the AIS) was overexpressed. Using superresolution nanoscopy and live-cell imaging, we observed that microtubules within the AIS appeared highly dynamic, a feature essential for the TDB. Pathomechanistically, amyloid-ß insult caused cofilin activation and F-actin remodeling and decreased microtubule dynamics in the AIS. Concomitantly with these amyloid-ß-induced disruptions, the AIS/TDB sorting function failed, causing AD-like Tau missorting. In summary, we provide evidence that the human and rodent Tau isoforms differ in axodendritic sorting and amyloid-ß-induced missorting and that the axodendritic distribution of Tau depends on AIS integrity.


Assuntos
Segmento Inicial do Axônio/metabolismo , Córtex Cerebral/metabolismo , Dendritos/metabolismo , Microtúbulos/metabolismo , Neurônios/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Segmento Inicial do Axônio/patologia , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/patologia , Dendritos/patologia , Difusão , Embrião de Mamíferos/citologia , Deleção de Genes , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microtúbulos/patologia , Mutagênese Insercional , Neurônios/citologia , Neurônios/patologia , Domínios e Motivos de Interação entre Proteínas , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte Proteico , Interferência de RNA , Ratos Wistar , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Sequências Repetitivas de Aminoácidos , Proteínas tau/antagonistas & inibidores , Proteínas tau/química , Proteínas tau/genética
9.
Neurobiol Dis ; 117: 189-202, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29894752

RESUMO

Mutations in the gene encoding Tau (MAPT-microtubule-associated protein tau) cause a group of neurodegenerative diseases called tauopathies. A recently identified Tau variant, p.A152T, has been reported as a risk factor for frontotemporal dementia-related disorders and Alzheimer disease. However, the mechanism for the pathologies still remain poorly understood. Transgenic Caenorhabditis elegans expressing mutant 2N4R-TauA152T (TauAT) panneuronally show locomotor defects, neurodegeneration and accelerated aging. Here we report that, in TauAT animals, the glutamatergic nervous system is at a high risk of progressive neuronal loss. We present genetic data that this loss occurs predominantly through necrosis. The neuronal loss is caused by several determinants, such as altered adenylyl cyclase (type AC9) pathway, prevalence of excitotoxicity-like conditions, aging-related factors and finally dyshomeostasis of intracellular calcium (Ca2+). The study provides novel insights into the mechanisms involved in selective loss of glutamatergic neurons in a TauAT tauopathy model which could point to new therapeutic targets.


Assuntos
Proteínas de Caenorhabditis elegans/biossíntese , Sinalização do Cálcio/fisiologia , Ácido Glutâmico/metabolismo , Degeneração Neural/metabolismo , Tauopatias/metabolismo , Proteínas tau/biossíntese , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Ácido Glutâmico/genética , Degeneração Neural/genética , Degeneração Neural/patologia , Tauopatias/genética , Tauopatias/patologia , Proteínas tau/genética
10.
EMBO Rep ; 17(4): 552-69, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26931569

RESUMO

We report on a novel transgenic mouse model expressing human full-length Tau with the Tau mutation A152T (hTau(AT)), a risk factor for FTD-spectrum disorders including PSP and CBD Brain neurons reveal pathological Tau conformation, hyperphosphorylation, mis-sorting, aggregation, neuronal degeneration, and progressive loss, most prominently in area CA3 of the hippocampus. The mossy fiber pathway shows enhanced basal synaptic transmission without changes in short- or long-term plasticity. In organotypic hippocampal slices, extracellular glutamate increases early above control levels, followed by a rise in neurotoxicity. These changes are normalized by inhibiting neurotransmitter release or by blocking voltage-gated sodium channels. CA3 neurons show elevated intracellular calcium during rest and after activity induction which is sensitive to NR2B antagonizing drugs, demonstrating a pivotal role of extrasynaptic NMDA receptors. Slices show pronounced epileptiform activity and axonal sprouting of mossy fibers. Excitotoxic neuronal death is ameliorated by ceftriaxone, which stimulates astrocytic glutamate uptake via the transporter EAAT2/GLT1. In summary, hTau(AT) causes excitotoxicity mediated by NR2B-containing NMDA receptors due to enhanced extracellular glutamate.


Assuntos
Região CA3 Hipocampal/metabolismo , Região CA3 Hipocampal/patologia , Demência Frontotemporal/fisiopatologia , Neurônios/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Animais , Cálcio/metabolismo , Meios de Cultura/química , Modelos Animais de Doenças , Ácido Glutâmico/análise , Humanos , Camundongos , Camundongos Transgênicos , Mutação , Plasticidade Neuronal , Neurônios/química , Neurônios/patologia , Técnicas de Cultura de Órgãos , Transmissão Sináptica , Proteínas tau/química
11.
EMBO J ; 32(22): 2920-37, 2013 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-24065130

RESUMO

Mislocalization and aggregation of Aß and Tau combined with loss of synapses and microtubules (MTs) are hallmarks of Alzheimer disease. We exposed mature primary neurons to Aß oligomers and analysed changes in the Tau/MT system. MT breakdown occurs in dendrites invaded by Tau (Tau missorting) and is mediated by spastin, an MT-severing enzyme. Spastin is recruited by MT polyglutamylation, induced by Tau missorting triggered translocalization of TTLL6 (Tubulin-Tyrosine-Ligase-Like-6) into dendrites. Consequences are spine loss and mitochondria and neurofilament mislocalization. Missorted Tau is not axonally derived, as shown by axonal retention of photoconvertible Dendra2-Tau, but newly synthesized. Recovery from Aß insult occurs after Aß oligomers lose their toxicity and requires the kinase MARK (Microtubule-Affinity-Regulating-Kinase). In neurons derived from Tau-knockout mice, MTs and synapses are resistant to Aß toxicity because TTLL6 mislocalization and MT polyglutamylation are prevented; hence no spastin recruitment and no MT breakdown occur, enabling faster recovery. Reintroduction of Tau re-establishes Aß-induced toxicity in TauKO neurons, which requires phosphorylation of Tau's KXGS motifs. Transgenic mice overexpressing Tau show TTLL6 translocalization into dendrites and decreased MT stability. The results provide a rationale for MT stabilization as a therapeutic approach.


Assuntos
Adenosina Trifosfatases/fisiologia , Peptídeos beta-Amiloides/fisiologia , Microtúbulos/fisiologia , Peptídeo Sintases/fisiologia , Sinapses/patologia , Proteínas tau/fisiologia , Peptídeos beta-Amiloides/química , Animais , Células Cultivadas , Ácido Glutâmico/metabolismo , Camundongos , Camundongos Knockout , Ratos , Espastina , Proteínas tau/genética
12.
Alzheimers Dement ; 13(11): 1270-1291, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28528849

RESUMO

INTRODUCTION: Tau-mediated toxicity in Alzheimer's disease is thought to operate through low-n oligomers, rather than filamentous aggregates. However, the nature of oligomers and pathways of toxicity are poorly understood. Therefore, we investigated structural and functional aspects of highly purified oligomers of a pro-aggregant tau species. METHODS: Purified oligomers of the tau repeat domain were characterized by biophysical and structural methods. Functional aspects were investigated by cellular assays ((3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay of cell viability, lactate dehydrogenase release assay [for cell toxicity], reactive oxygen species production, and calcium assay), combined with analysis of neuronal dendritic spines exposed to oligomers. RESULTS: Purified low-n oligomers are roughly globular, with sizes around 1.6 to 5.4 nm, exhibit an altered conformation, but do not have substantial ß-structure. Treatment of primary neurons with oligomers impairs spine morphology and density, accompanied by increased reactive oxygen species and intracellular calcium, but without affecting cell viability (by (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay of cell viability and lactate dehydrogenase release assay [for cell toxicity]). DISCUSSION: Tau oligomers are toxic to synapses but not lethal to cells.


Assuntos
Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Fragmentos de Peptídeos/toxicidade , Sinapses/efeitos dos fármacos , Proteínas tau/química , Animais , Cálcio/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Dicroísmo Circular , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/ultraestrutura , Líquido Extracelular/efeitos dos fármacos , Hipocampo/citologia , Humanos , Imunoprecipitação , Técnicas In Vitro , Microscopia de Força Atômica , Microscopia Eletrônica de Transmissão , Neuroblastoma , Técnicas de Cultura de Órgãos , Células PC12 , Ratos , Espécies Reativas de Oxigênio/metabolismo , Espectrofotometria , Sinapses/metabolismo , Proteínas tau/toxicidade
13.
Alzheimers Dement ; 13(6): 701-709, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27750032

RESUMO

INTRODUCTION: The inhibition of the ß-site amyloid precursor protein-cleaving enzyme 1 (BACE1) is a main therapeutic approach for the treatment of Alzheimer's disease (AD). We previously reported an age-related increase of tau protein in the cerebrospinal fluid (CSF) of amyloid ß (Aß) precursor protein (APP) transgenic mice. METHODS: APP transgenic mice were treated with a potent BACE1 inhibitor. CSF tau and CSF Aß levels were assessed. A novel high-sensitivity tau sandwich immunoassay was developed. RESULTS: We demonstrate that long-term BACE1 inhibition prevents CSF tau increase both in early-depositing APP transgenic mice and APP transgenic mice with moderate Aß pathology. DISCUSSION: Our results demonstrate that BACE1 inhibition not only reduces Aß generation but also downstream AD pathophysiology. The tight correlation between Aß aggregation in brain and CSF tau levels renders CSF tau a valuable marker to predict the effectiveness of BACE1 inhibitors in current clinical trials.


Assuntos
Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Ácidos Picolínicos/farmacologia , Tiazinas/farmacologia , Proteínas tau/líquido cefalorraquidiano , Envelhecimento/efeitos dos fármacos , Envelhecimento/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Imunoensaio , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Placa Amiloide/tratamento farmacológico , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/metabolismo , Prosencéfalo/patologia
14.
Alzheimers Dement ; 12(10): 1033-1039, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27154059

RESUMO

Alzheimer's Association Research Roundtable Fall 2015-Tau: From research to clinical development. Tau pathology is recognized as the key driver of disease progression in Alzheimer's and other neurodegenerative diseases. Although this makes tau an attractive target for the development of novel diagnostic and therapeutic strategies, the mechanisms underlying the onset and progression of tau-related neurotoxicity remain elusive. Recent strides in the development of sophisticated preclinical models and the emergence of tau PET imaging and fluid biomarkers provide new opportunities to increase our understanding of tau biology, overcome translational challenges, and accelerate the advancement of tau therapeutics from bench to bedside. With this in mind, the Alzheimer's Association convened a Research Roundtable in October 2015, bringing together experts from academia, industry, and regulatory agencies to discuss the latest understanding of tau pathogenic pathways and review the evolution of tau therapeutics and biomarkers currently in development. The meeting provided a forum to share experience and expertise with the common goal of advancing the discovery and development of new treatment strategies and expediting the design and implementation of efficient clinical trials.


Assuntos
Progressão da Doença , Tauopatias , Proteínas tau/metabolismo , Doença de Alzheimer/patologia , Animais , Biomarcadores/metabolismo , Modelos Animais de Doenças , Humanos , Emaranhados Neurofibrilares/patologia , Fosforilação
15.
J Biol Chem ; 289(29): 20318-32, 2014 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-24825901

RESUMO

Several neurodegenerative diseases are characterized by the aggregation and posttranslational modifications of Tau protein. Its "repeat domain" (TauRD) is mainly responsible for the aggregation properties, and oligomeric forms are thought to dominate the toxic effects of Tau. Here we investigated the conformational transitions of this domain during oligomerization and aggregation in different states of ß-propensity and pseudo-phosphorylation, using several complementary imaging and spectroscopic methods. Although the repeat domain generally aggregates more readily than full-length Tau, its aggregation was greatly slowed down by phosphorylation or pseudo-phosphorylation at the KXGS motifs, concomitant with an extended phase of oligomerization. Analogous effects were observed with pro-aggregant variants of TauRD. Oligomers became most evident in the case of the pro-aggregant mutant TauRDΔK280, as monitored by atomic force microscopy, and the fluorescence lifetime of Alexa-labeled Tau (time-correlated single photon counting (TCSPC)), consistent with its pronounced toxicity in mouse models. In cell models or primary neurons, neither oligomers nor fibrils of TauRD or TauRDΔK280 had a toxic effect, as seen by assays with lactate dehydrogenase and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, respectively. However, oligomers of pro-aggregant TauRDΔK280 specifically caused a loss of spine density in differentiated neurons, indicating a locally restricted impairment of function.


Assuntos
Neurônios/metabolismo , Neurônios/patologia , Proteínas tau/química , Proteínas tau/metabolismo , Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Motivos de Aminoácidos , Animais , Células , Células Cultivadas , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/patologia , Humanos , Camundongos , Microscopia de Força Atômica , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Fosforilação , Conformação Proteica , Multimerização Proteica , Estrutura Terciária de Proteína , Sequências Repetitivas de Aminoácidos , Proteínas tau/genética
16.
J Biol Chem ; 289(49): 34389-407, 2014 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-25339173

RESUMO

Abnormal phosphorylation ("hyperphosphorylation") and aggregation of Tau protein are hallmarks of Alzheimer disease and other tauopathies, but their causative connection is still a matter of debate. Tau with Alzheimer-like phosphorylation is also present in hibernating animals, mitosis, or during embryonic development, without leading to pathophysiology or neurodegeneration. Thus, the role of phosphorylation and the distinction between physiological and pathological phosphorylation needs to be further refined. So far, the systematic investigation of highly phosphorylated Tau was difficult because a reliable method of preparing reproducible quantities was not available. Here, we generated full-length Tau (2N4R) in Sf9 cells in a well defined phosphorylation state containing up to ∼20 phosphates as judged by mass spectrometry and Western blotting with phospho-specific antibodies. Despite the high concentration in living Sf9 cells (estimated ∼230 µm) and high phosphorylation, the protein was not aggregated. However, after purification, the highly phosphorylated protein readily formed oligomers, whereas fibrils were observed only rarely. Exposure of mature primary neuronal cultures to oligomeric phospho-Tau caused reduction of spine density on dendrites but did not change the overall cell viability.


Assuntos
Córtex Cerebral/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Multimerização Proteica/genética , Processamento de Proteína Pós-Traducional , Sequência de Aminoácidos , Animais , Anticorpos Fosfo-Específicos/química , Baculoviridae/genética , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Expressão Gênica , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Humanos , Camundongos , Dados de Sequência Molecular , Neurônios/citologia , Neurônios/efeitos dos fármacos , Mapeamento de Peptídeos , Fosforilação , Cultura Primária de Células , Agregados Proteicos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Células Sf9 , Spodoptera , Proteínas tau/genética , Proteínas tau/metabolismo , Proteínas tau/farmacologia
17.
J Biol Chem ; 289(2): 956-67, 2014 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-24235150

RESUMO

Understanding the formation and propagation of aggregates of the Alzheimer disease-associated Tau protein in vivo is vital for the development of therapeutics for this devastating disorder. Using our recently developed live-cell aggregation sensor in neuron-like cells, we demonstrate that different variants of exogenous monomeric Tau, namely full-length Tau (hTau40) and the Tau-derived construct K18 comprising the repeat domain, initially accumulate in endosomal compartments, where they form fibrillar seeds that subsequently induce the aggregation of endogenous Tau. Using superresolution imaging, we confirm that fibrils consisting of endogenous and exogenous Tau are released from cells and demonstrate their potential to spread Tau pathology. Our data indicate a greater pathological risk and potential toxicity than hitherto suspected for extracellular soluble Tau.


Assuntos
Endocitose , Emaranhados Neurofibrilares/metabolismo , Neurônios/metabolismo , Proteínas tau/metabolismo , Animais , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Endossomos/metabolismo , Exocitose , Espaço Extracelular/metabolismo , Humanos , Lisossomos/metabolismo , Microscopia Confocal , Microscopia Eletrônica , Modelos Biológicos , Emaranhados Neurofibrilares/ultraestrutura , Neurônios/patologia , Tauopatias/metabolismo , Vesículas Transportadoras/metabolismo
18.
EMBO J ; 30(23): 4825-37, 2011 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-22009197

RESUMO

Missorting of Tau from axons to the somatodendritic compartment of neurons is a hallmark of Alzheimer's disease, but the mechanisms underlying normal sorting and pathological failure are poorly understood. Here, we used several Tau constructs labelled with photoconvertible Dendra2 to analyse its mobility in polarized neurons. This revealed a novel mechanism of sorting-a retrograde barrier in the axon initial segment (AIS) operating as cellular rectifier. It allows anterograde flow of axonal Tau but prevents retrograde flow back into soma and dendrites. The barrier requires binding of Tau to microtubules but does not require F-actin and thus is distinct from the sorting of membrane-associated proteins at the AIS. The barrier breaks down when Tau is phosphorylated in its repeat domain and detached from microtubules, for example, by the kinase MARK/Par1. These observations link the pathological hallmarks of Tau missorting and hyperphosphorylation in neurodegenerative diseases.


Assuntos
Transporte Axonal/fisiologia , Axônios/metabolismo , Microtúbulos/metabolismo , Transporte Proteico/fisiologia , Proteínas tau/metabolismo , Actinas/metabolismo , Doença de Alzheimer/fisiopatologia , Animais , Axônios/ultraestrutura , Polaridade Celular/fisiologia , Células Cultivadas , Córtex Cerebral/citologia , Dendritos/metabolismo , Dendritos/ultraestrutura , Vetores Genéticos/genética , Humanos , Proteínas Luminescentes/análise , Proteínas de Membrana/metabolismo , Microtúbulos/ultraestrutura , Fosforilação , Fotoquímica/métodos , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Ratos Sprague-Dawley
19.
Hum Mol Genet ; 21(8): 1681-92, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22194194

RESUMO

Microdeletions encompassing the MAPT (Tau) locus resulting in intellectual disability raised the hypothesis that Tau may regulate early functions in the developing brain. Our results indicate that neuronal migration was inhibited in mouse brains following Tau reduction. In addition, the leading edge of radially migrating neurons was aberrant in spite of normal morphology of radial glia. Furthermore, intracellular mitochondrial transport and morphology were affected. In early postnatal brains, a portion of Tau knocked down neurons reached the cortical plate. Nevertheless, they exhibited far less developed dendrites and a striking reduction in connectivity evident by the size of boutons. Our novel results strongly implicate MAPT as a dosage-sensitive gene in this locus involved in intellectual disability. Furthermore, our results are likely to impact our understanding of other diseases involving Tau.


Assuntos
Encéfalo/embriologia , Deficiência Intelectual/genética , Neurônios/fisiologia , Proteínas tau/genética , Proteínas tau/metabolismo , Animais , Axônios/ultraestrutura , Encéfalo/citologia , Encéfalo/metabolismo , Movimento Celular , Forma Celular , Células Cultivadas , Dendritos/ultraestrutura , Eletroporação , Embrião de Mamíferos , Desenvolvimento Embrionário , Técnicas de Silenciamento de Genes , Deficiência Intelectual/metabolismo , Camundongos , Mitocôndrias/ultraestrutura , Neuroglia/ultraestrutura , Neurônios/citologia , Neurônios/ultraestrutura , Terminações Pré-Sinápticas/ultraestrutura , RNA Interferente Pequeno
20.
Hum Mol Genet ; 21(16): 3587-603, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22611162

RESUMO

Increased Tau protein amyloidogenicity has been causatively implicated in several neurodegenerative diseases, collectively called tauopathies. In pathological conditions, Tau becomes hyperphosphorylated and forms intracellular aggregates. The deletion of K280, which is a mutation that commonly appears in patients with frontotemporal dementia with Parkinsonism linked to chromosome 17, enhances Tau aggregation propensity (pro-aggregation). In contrast, introduction of the I277P and I308P mutations prevents ß-sheet formation and subsequent aggregation (anti-aggregation). In this study, we created a tauopathy model by expressing pro- or anti-aggregant Tau species in the nervous system of Caenorhabditis elegans. Animals expressing the highly amyloidogenic Tau species showed accelerated Tau aggregation and pathology manifested by severely impaired motility and evident neuronal dysfunction. In addition, we observed that the axonal transport of mitochondria was perturbed in these animals. Control animals expressing the anti-aggregant combination had rather mild phenotype. We subsequently tested several Tau aggregation inhibitor compounds and observed a mitigation of Tau proteotoxicity. In particular, a novel compound that crosses the blood-brain barrier of mammals proved effective in ameliorating the motility as well as delaying the accumulation of neuronal defects. Our study establishes a new C. elegans model of Tau aggregation-mediated toxicity and supports the emerging notion that inhibiting the nucleation of Tau aggregation can be neuroprotective.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Neurônios/patologia , Tauopatias/etiologia , Proteínas tau/metabolismo , Animais , Animais Geneticamente Modificados , Transporte Axonal , Barreira Hematoencefálica/efeitos dos fármacos , Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Modelos Animais de Doenças , Humanos , Hidrazinas/farmacologia , Azul de Metileno/farmacologia , Mitocôndrias/metabolismo , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Mutação , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Fenótipo , Fosforilação , Estrutura Terciária de Proteína , Células Receptoras Sensoriais/metabolismo , Tauopatias/patologia , Tiazóis/farmacologia , Proteína 1 Associada à Membrana da Vesícula/metabolismo , Proteínas tau/antagonistas & inibidores , Proteínas tau/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA