Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 41(11): e108882, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35298090

RESUMEN

Biomolecular condensation of the neuronal microtubule-associated protein Tau (MAPT) can be induced by coacervation with polyanions like RNA, or by molecular crowding. Tau condensates have been linked to both functional microtubule binding and pathological aggregation in neurodegenerative diseases. We find that molecular crowding and coacervation with RNA, two conditions likely coexisting in the cytosol, synergize to enable Tau condensation at physiological buffer conditions and to produce condensates with a strong affinity to charged surfaces. During condensate-mediated microtubule polymerization, their synergy enhances bundling and spatial arrangement of microtubules. We further show that different Tau condensates efficiently induce pathological Tau aggregates in cells, including accumulations at the nuclear envelope that correlate with nucleocytoplasmic transport deficits. Fluorescent lifetime imaging reveals different molecular packing densities of Tau in cellular accumulations and a condensate-like density for nuclear-envelope Tau. These findings suggest that a complex interplay between interaction partners, post-translational modifications, and molecular crowding regulates the formation and function of Tau condensates. Conditions leading to prolonged existence of Tau condensates may induce the formation of seeding-competent Tau and lead to distinct cellular Tau accumulations.


Asunto(s)
Enfermedades Neurodegenerativas , ARN , Humanos , Microtúbulos/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , Unión Proteica , ARN/metabolismo , Proteínas tau/metabolismo
2.
Nature ; 580(7803): 381-385, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32296178

RESUMEN

The spread of protein aggregates during disease progression is a common theme underlying many neurodegenerative diseases. The microtubule-associated protein tau has a central role in the pathogenesis of several forms of dementia known as tauopathies-including Alzheimer's disease, frontotemporal dementia and chronic traumatic encephalopathy1. Progression of these diseases is characterized by the sequential spread and deposition of protein aggregates in a predictable pattern that correlates with clinical severity2. This observation and complementary experimental studies3,4 have suggested that tau can spread in a prion-like manner, by passing to naive cells in which it templates misfolding and aggregation. However, although the propagation of tau has been extensively studied, the underlying cellular mechanisms remain poorly understood. Here we show that the low-density lipoprotein receptor-related protein 1 (LRP1) controls the endocytosis of tau and its subsequent spread. Knockdown of LRP1 significantly reduced tau uptake in H4 neuroglioma cells and in induced pluripotent stem cell-derived neurons. The interaction between tau and LRP1 is mediated by lysine residues in the microtubule-binding repeat region of tau. Furthermore, downregulation of LRP1 in an in vivo mouse model of tau spread was found to effectively reduce the propagation of tau between neurons. Our results identify LRP1 as a key regulator of tau spread in the brain, and therefore a potential target for the treatment of diseases that involve tau spread and aggregation.


Asunto(s)
Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteínas tau/metabolismo , Animales , Línea Celular , Endocitosis , Femenino , Humanos , Ligandos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Masculino , Ratones , Neuronas/metabolismo
3.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-33619090

RESUMEN

Tau protein plays an important role in the biology of stress granules and in the stress response of neurons, but the nature of these biochemical interactions is not known. Here we show that the interaction of tau with RNA and the RNA binding protein TIA1 is sufficient to drive phase separation of tau at physiological concentrations, without the requirement for artificial crowding agents such as polyethylene glycol (PEG). We further show that phase separation of tau in the presence of RNA and TIA1 generates abundant tau oligomers. Prior studies indicate that recombinant tau readily forms oligomers and fibrils in vitro in the presence of polyanionic agents, including RNA, but the resulting tau aggregates are not particularly toxic. We discover that tau oligomers generated during copartitioning with TIA1 are significantly more toxic than tau aggregates generated by incubation with RNA alone or phase-separated tau complexes generated by incubation with artificial crowding agents. This pathway identifies a potentially important source for generation of toxic tau oligomers in tau-related neurodegenerative diseases. Our results also reveal a general principle that phase-separated RBP droplets provide a vehicle for coassortment of selected proteins. Tau selectively copartitions with TIA1 under physiological conditions, emphasizing the importance of TIA1 for tau biology. Other RBPs, such as G3BP1, are able to copartition with tau, but this happens only in the presence of crowding agents. This type of selective mixing might provide a basis through which membraneless organelles bring together functionally relevant proteins to promote particular biological activities.


Asunto(s)
Agregado de Proteínas , Agregación Patológica de Proteínas , Multimerización de Proteína , Antígeno Intracelular 1 de las Células T/metabolismo , Proteínas tau/metabolismo , Amiloide/química , Amiloide/metabolismo , Humanos , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuronas/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas con Motivos de Reconocimiento de ARN/química , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , Proteínas Recombinantes , Proteínas tau/química
4.
Brain Behav Immun ; 110: 245-259, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36906076

RESUMEN

Remodeling of synapses by microglia is essential for synaptic plasticity in the brain. However, during neuroinflammation and neurodegenerative diseases, microglia can induce excessive synaptic loss, although the precise underlying mechanisms are unknown. To directly observe microglia-synapse interactions under inflammatory conditions, we performed in vivo two-photon time-lapse imaging of microglia-synapse interactions after bacterial lipopolysaccharide administration to model systemic inflammation, or after inoculation of Alzheimer's disease (AD) brain extracts to model disease-associated neuroinflammatory microglial response. Both treatments prolonged microglia-neuron contacts, decreased basal surveillance of synapses and promoted synaptic remodeling in response to synaptic stress induced by focal single-synapse photodamage. Spine elimination correlated with the expression of microglial complement system/phagocytic proteins and the occurrence of synaptic filopodia. Microglia were observed contacting spines, then stretching and phagocytosing spine head filopodia. Thus, in response to inflammatory stimuli microglia exacerbated spine remodeling through prolonged microglial contact and elimination of spines 'tagged' by synaptic filopodia.


Asunto(s)
Enfermedad de Alzheimer , Tauopatías , Humanos , Microglía/metabolismo , Tauopatías/metabolismo , Enfermedad de Alzheimer/metabolismo , Sinapsis/metabolismo , Inflamación/metabolismo
5.
Semin Cell Dev Biol ; 99: 202-214, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31260737

RESUMEN

Cells contain multiple compartments dedicated to the regulation and control of biochemical reactions. Cellular compartments that are not surrounded by membranes can rapidly form and dissolve in response to changes in the cellular environment. The physicochemical processes that underlie the formation of non-membrane-bound compartments in vivo are connected to liquid-liquid phase separation of proteins and nucleic acids in vitro. Recent evidence suggests that the protein tau, which plays an important role in Alzheimer's disease and other neurodegenerative disorders, phase separates in solution, forms tau phases with microtubules, and associates with phase-separated RNA-binding protein granules in cells. Here we review the experimental evidence that supports the ability of tau to phase separate in solution and form biomolecular condensates in cells. As for other disease-relevant proteins, the physiological and pathological functions of tau are tightly connected - through loss of normal function or gain of toxic function - and we therefore discuss how tau phase separation plays a role for both, and with respect to different cellular functions of tau.


Asunto(s)
Proteínas tau/química , Proteínas tau/metabolismo , Animales , Humanos , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología
6.
EMBO J ; 37(7)2018 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-29472250

RESUMEN

The transition between soluble intrinsically disordered tau protein and aggregated tau in neurofibrillary tangles in Alzheimer's disease is unknown. Here, we propose that soluble tau species can undergo liquid-liquid phase separation (LLPS) under cellular conditions and that phase-separated tau droplets can serve as an intermediate toward tau aggregate formation. We demonstrate that phosphorylated or mutant aggregation prone recombinant tau undergoes LLPS, as does high molecular weight soluble phospho-tau isolated from human Alzheimer brain. Droplet-like tau can also be observed in neurons and other cells. We found that tau droplets become gel-like in minutes, and over days start to spontaneously form thioflavin-S-positive tau aggregates that are competent of seeding cellular tau aggregation. Since analogous LLPS observations have been made for FUS, hnRNPA1, and TDP43, which aggregate in the context of amyotrophic lateral sclerosis, we suggest that LLPS represents a biophysical process with a role in multiple different neurodegenerative diseases.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Encéfalo/metabolismo , Neuronas/metabolismo , Agregación Patológica de Proteínas/metabolismo , Proteínas tau/química , Proteínas tau/aislamiento & purificación , Proteínas tau/metabolismo , Anciano de 80 o más Años , Secuencia de Aminoácidos , Animales , Benzotiazoles/metabolismo , Fenómenos Biofísicos , Clonación Molecular , Proteínas de Unión al ADN/metabolismo , Escherichia coli/genética , Femenino , Células HEK293 , Ribonucleoproteína Nuclear Heterogénea A1/metabolismo , Humanos , Extracción Líquido-Líquido , Ratones , Ratones Transgénicos , Peso Molecular , Neuroblastoma/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Ovillos Neurofibrilares/metabolismo , Fosforilación , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Análisis de Secuencia de Proteína , Células Sf9
7.
Int J Mol Sci ; 23(7)2022 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-35408855

RESUMEN

Tau is a neuronal protein that stabilizes axonal microtubules (MTs) in the central nervous system. In Alzheimer's disease (AD) and other tauopathies, phosphorylated Tau accumulates in intracellular aggregates, a pathological hallmark of these diseases. However, the chronological order of pathological changes in Tau prior to its cytosolic aggregation remains unresolved. These include its phosphorylation and detachment from MTs, mislocalization into the somatodendritic compartment, and oligomerization in the cytosol. Recently, we showed that Tau can interact with phenylalanine-glycine (FG)-rich nucleoporins (Nups), including Nup98, that form a diffusion barrier inside nuclear pore complexes (NPCs), leading to defects in nucleocytoplasmic transport. Here, we used surface plasmon resonance (SPR) and bio-layer interferometry (BLI) to investigate the molecular details of Tau:Nup98 interactions and determined how Tau phosphorylation and oligomerization impact the interactions. Importantly, phosphorylation, but not acetylation, strongly facilitates the accumulation of Tau with Nup98. Oligomerization, however, seems to inhibit Tau:Nup98 interactions, suggesting that Tau-FG Nup interactions occur prior to oligomerization. Overall, these results provide fundamental insights into the molecular mechanisms of Tau-FG Nup interactions within NPCs, which might explain how stress-and disease-associated posttranslational modifications (PTMs) may lead to Tau-induced nucleocytoplasmic transport (NCT) failure. Intervention strategies that could rescue Tau-induced NCT failure in AD and tauopathies will be further discussed.


Asunto(s)
Proteínas de Complejo Poro Nuclear , Tauopatías , Transporte Activo de Núcleo Celular , Humanos , Poro Nuclear/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Fosforilación , Tauopatías/metabolismo , Proteínas tau/metabolismo
8.
J Biol Chem ; 293(34): 13247-13256, 2018 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-29950521

RESUMEN

Apolipoprotein E (ApoE) is a secreted apolipoprotein with three isoforms, E2, E3, and E4, that binds to lipids and facilitates their transport in the extracellular environment of the brain and the periphery. The E4 allele is a major genetic risk factor for the sporadic form of Alzheimer's disease (AD), and studies of human brain and mouse models have revealed that E4 significantly exacerbates the deposition of amyloid beta (Aß). It has been suggested that this deposition could be attributed to the formation of soluble ApoE isoform-specific ApoE-Aß complexes. However, previous studies have reported conflicting results regarding the directionality and strength of those interactions. In this study, using a series of flow cytometry assays that maintain the physiological integrity of ApoE-Aß complexes, we systematically assessed the association of Aß with ApoE2, E3, or E4. We used ApoE secreted from HEK cells or astrocytes overexpressing ApoE fused with a GFP tag. As a source of soluble Aß peptide, we used synthetic Aß40 or Aß42 or physiological Aß secreted from CHO cell lines overexpressing WT or V717F variant amyloid precursor protein (APP). We observed significant interactions between the different ApoE isoforms and Aß, with E4 interacting with Aß more strongly than the E2 and E3 isoforms. We also found subtle differences depending on the Aß type and the ApoE-producing cell type. In conclusion, these results indicate that the strength of the ApoE-Aß association depends on the source of Aß or ApoE.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Apolipoproteína E2/metabolismo , Apolipoproteína E3/metabolismo , Apolipoproteína E4/metabolismo , Astrocitos/metabolismo , Citometría de Flujo/métodos , Neuronas/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Astrocitos/citología , Bioensayo , Linaje de la Célula , Células HEK293 , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Transgénicos , Neuronas/citología , Isoformas de Proteínas
9.
EMBO J ; 34(24): 3028-41, 2015 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-26538322

RESUMEN

In Alzheimer's disease and tauopathies, tau protein aggregates into neurofibrillary tangles that progressively spread to synaptically connected brain regions. A prion-like mechanism has been suggested: misfolded tau propagating through the brain seeds neurotoxic aggregation of soluble tau in recipient neurons. We use transgenic mice and viral tau expression to test the hypotheses that trans-synaptic tau propagation, aggregation, and toxicity rely on the presence of endogenous soluble tau. Surprisingly, mice expressing human P301Ltau in the entorhinal cortex showed equivalent tau propagation and accumulation in recipient neurons even in the absence of endogenous tau. We then tested whether the lack of endogenous tau protects against misfolded tau aggregation and toxicity, a second prion model paradigm for tau, using P301Ltau-overexpressing mice with severe tangle pathology and neurodegeneration. Crossed onto tau-null background, these mice had similar tangle numbers but were protected against neurotoxicity. Therefore, misfolded tau can propagate across neural systems without requisite templated misfolding, but the absence of endogenous tau markedly blunts toxicity. These results show that tau does not strictly classify as a prion protein.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Proteínas tau/genética , Animales , Células Cultivadas , Corteza Entorrinal/citología , Corteza Entorrinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Mutación Missense , Neuronas/metabolismo , Proteínas tau/deficiencia , Proteínas tau/metabolismo
10.
Adv Exp Med Biol ; 1184: 341-357, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32096048

RESUMEN

Tau is an intrinsically unfolded protein that, aside from its important role in the regulation of microtubule stability, harbors an emerging number of other functions. In order to find explanations for some longtime unsolved aspects of neuronal tau biology in the brain, we may have to step aside from observing tau molecules in dilute solutions, and from assuming a mono-molecular physicochemical behavior of molecules in the cell. Liquid condensed phases of tau proteins, which form through the biophysical process of liquid-liquid phase separation (LLPS), behave like liquids and thereby offer a new regime of interactions in the cell. So far, there is evidence that tau condensates (i) play a role for neurodegenerative diseases by transitioning into aggregated forms of tau, (ii) are involved in microtubule binding, nucleation, and bundling, and (iii) are interacting with RNA molecules, which could impact RNA homeostasis and transcription. Likewise the functions of monomeric tau, also tau condensation is regulated by post-translational modifications and can be influenced by the local environment, for example in neuronal sub-compartments. However, we are just beginning to understand the physicochemistry of tau LLPS, and the biological role of tau condensation has to be explored in the next years.


Asunto(s)
Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuronas/metabolismo , Neuronas/patología , Proteínas tau/química , Proteínas tau/metabolismo , Humanos , Microtúbulos/metabolismo , Neurobiología , Patología , Proteínas tau/aislamiento & purificación
11.
Nano Lett ; 18(5): 3271-3281, 2018 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-29644863

RESUMEN

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.


Asunto(s)
Encéfalo/metabolismo , Membrana Celular/metabolismo , Lípidos de la Membrana/metabolismo , Proteínas tau/metabolismo , Cationes/metabolismo , Humanos , Microscopía de Fuerza Atómica , Agregado de Proteínas , Unión Proteica , Dominios Proteicos , Estabilidad Proteica , Proteínas tau/química
12.
J Biol Chem ; 292(36): 14720-14729, 2017 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-28684412

RESUMEN

Apolipoprotein E (apoE) has an important role in the pathogenesis of Alzheimer's disease with its three isoforms having distinct effects on disease risk. Here, we assessed the conformational differences between those isoforms using a novel flow cytometry-Forster resonance energy transfer (FRET) assay. We showed that the conformation of intracellular apoE within HEK cells and astrocytes adopts a directional pattern; in other words, E4 adopts the most closed conformation, E2 adopts the most open conformation, and E3 adopts an intermediate conformation. However, this pattern was not maintained upon secretion of apoE from astrocytes. Intermolecular interactions between apoE molecules were isoform-specific, indicating a great diversity in the structure of apoE lipoparticles. Finally, we showed that secreted E4 is the most lipidated isoform in astrocytes, suggesting that increased lipidation acts as a folding chaperone enabling E4 to adopt a closed conformation. In conclusion, this study gives insights into apoE biology and establishes a robust screening system to monitor apoE conformation.


Asunto(s)
Apolipoproteínas E/química , Astrocitos/química , Transferencia Resonante de Energía de Fluorescencia , Apolipoproteínas E/metabolismo , Citometría de Flujo , Células HEK293 , Humanos , Conformación Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo
13.
Am J Pathol ; 187(6): 1399-1412, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28408124

RESUMEN

The clinical progression of Alzheimer disease (AD) is associated with the accumulation of tau neurofibrillary tangles, which may spread throughout the cortex by interneuronal tau transfer. If so, targeting extracellular tau species may slow the spreading of tau pathology and possibly cognitive decline. To identify suitable target epitopes, we tested the effects of a panel of tau antibodies on neuronal uptake and aggregation in vitro. Immunodepletion was performed on brain extract from tau-transgenic mice and postmortem AD brain and added to a sensitive fluorescence resonance energy transfer-based tau uptake assay to assess blocking efficacy. The antibodies reduced tau uptake in an epitope-dependent manner: N-terminal (Tau13) and middomain (6C5 and HT7) antibodies successfully prevented uptake of tau species, whereas the distal C-terminal-specific antibody (Tau46) had little effect. Phosphorylation-dependent (40E8 and p396) and C-terminal half (4E4) tau antibodies also reduced tau uptake despite removing less total tau by immunodepletion, suggesting specific interactions with species involved in uptake. Among the seven antibodies evaluated, 6C5 most efficiently blocked uptake and subsequent aggregation. More important, 6C5 also blocked neuron-to-neuron spreading of tau in a unique three-chamber microfluidic device. Furthermore, 6C5 slowed down the progression of tau aggregation even after uptake had begun. Our results imply that not all antibodies/epitopes are equally robust in terms of blocking tau uptake of human AD-derived tau species.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Neuronas/metabolismo , Proteínas tau/metabolismo , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Epítopos/inmunología , Femenino , Humanos , Interneuronas/metabolismo , Masculino , Ratones Transgénicos , Técnicas Analíticas Microfluídicas , Terapia Molecular Dirigida/métodos , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Fosforilación , Proteínas tau/antagonistas & inhibidores , Proteínas tau/inmunología
14.
Ann Neurol ; 80(3): 355-67, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27351289

RESUMEN

OBJECTIVE: Cerebrospinal fluid (CSF) tau is an excellent surrogate marker for assessing neuropathological changes that occur in Alzheimer's disease (AD) patients. However, whether the elevated tau in AD CSF is just a marker of neurodegeneration or, in fact, a part of the disease process is uncertain. Moreover, it is unknown how CSF tau relates to the recently described soluble high-molecular-weight (HMW) species that is found in the postmortem AD brain and can be taken up by neurons and seed aggregates. METHODS: We have examined seeding and uptake properties of brain extracellular tau from various sources, including interstitial fluid (ISF) and CSF from an AD transgenic mouse model and postmortem ventricular and antemortem lumbar CSF from AD patients. RESULTS: We found that brain ISF and CSF tau from the AD mouse model can be taken up by cells and induce intracellular aggregates. Ventricular CSF from AD patients contained a rare HMW tau species that exerted a higher seeding activity. Notably, the HMW tau species was also detected in lumbar CSF from AD patients, and its levels were significantly elevated compared to control subjects. HMW tau derived from CSF of AD patients was seed competent in vitro. INTERPRETATION: These findings suggest that CSF from an AD brain contains potentially bioactive HMW tau species, giving new insights into the role of CSF tau and biomarker development for AD. Ann Neurol 2016;80:355-367.


Asunto(s)
Enfermedad de Alzheimer/líquido cefalorraquídeo , Encéfalo/metabolismo , Proteínas tau/líquido cefalorraquídeo , Anciano , Animales , Biomarcadores/líquido cefalorraquídeo , Líquido Extracelular/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad
15.
Synapse ; 71(6)2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28196395

RESUMEN

Synaptic dysfunction and loss is the strongest pathological correlate of cognitive decline in Alzheimer's disease (AD) with increasing evidence implicating neuropathological tau protein in this process. Despite the knowledge that tau spreads through defined synaptic circuits, it is currently unknown whether synapse loss occurs before the accumulation of tau or as a consequence. To address this, we have used array tomography to examine an rTgTauEC mouse model expressing a P301L human tau transgene and a transgene labeling cytoplasm red (tdTomato) and presynaptic terminals green (Synaptophysin-EGFP). All transgenes are restricted primarily to the entorhinal cortex using the neuropsin promotor to drive tTA expression. It has previously been shown that rTgTauEC mice exhibit neuronal loss in the entorhinal cortex and synapse density loss in the middle molecular layer (MML) of the dentate gyrus at 24 months of age. Here, we observed the density of tau-expressing and total presynapses, and the spread of tau into the postsynapse in the MML of 3-6, 9, and 18 month old red-green-rTgTauEC mice. We observe no loss of synapse density in the MML up to 18 months even in axons expressing tau. Despite the maintenance of synapse density, we see spread of human tau from presynaptic terminals to postsynaptic compartments in the MML at very early ages, indicating that the spread of tau through neural circuits is not due to the degeneration of axon terminals and is an early feature of the disease process.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Neuronas/metabolismo , Terminales Presinápticos/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Axones/metabolismo , Axones/patología , Muerte Celular , Corteza Entorrinal/metabolismo , Corteza Entorrinal/patología , Femenino , Masculino , Ratones , Neuronas/patología , Terminales Presinápticos/patología , Proteínas tau/genética
16.
Proc Natl Acad Sci U S A ; 111(1): 510-4, 2014 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-24368848

RESUMEN

Alzheimer's disease (AD) is pathologically characterized by the deposition of extracellular amyloid-ß plaques and intracellular aggregation of tau protein in neurofibrillary tangles (NFTs) (1, 2). Progression of NFT pathology is closely correlated with both increased neurodegeneration and cognitive decline in AD (3) and other tauopathies, such as frontotemporal dementia (4, 5). The assumption that mislocalization of tau into the somatodendritic compartment (6) and accumulation of fibrillar aggregates in NFTs mediates neurodegeneration underlies most current therapeutic strategies aimed at preventing NFT formation or disrupting existing NFTs (7, 8). Although several disease-associated mutations cause both aggregation of tau and neurodegeneration, whether NFTs per se contribute to neuronal and network dysfunction in vivo is unknown (9). Here we used awake in vivo two-photon calcium imaging to monitor neuronal function in adult rTg4510 mice that overexpress a human mutant form of tau (P301L) and develop cortical NFTs by the age of 7-8 mo (10). Unexpectedly, NFT-bearing neurons in the visual cortex appeared to be completely functionally intact, to be capable of integrating dendritic inputs and effectively encoding orientation and direction selectivity, and to have a stable baseline resting calcium level. These results suggest a reevaluation of the common assumption that insoluble tau aggregates are sufficient to disrupt neuronal function.


Asunto(s)
Ovillos Neurofibrilares/metabolismo , Proteínas tau/genética , Enfermedad de Alzheimer/patología , Animales , Encéfalo/patología , Calcio/metabolismo , Dependovirus/metabolismo , Humanos , Procesamiento de Imagen Asistido por Computador , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Modelos Neurológicos , Mutación , Neuronas/patología , Fotones , Tauopatías/patología , Transgenes , Proteínas tau/metabolismo
17.
J Biol Chem ; 290(4): 1966-78, 2015 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-25468905

RESUMEN

Interfering with the assembly of Amyloid ß (Aß) peptides from monomer to oligomeric species and fibrils or promoting their clearance from the brain are targets of anti-Aß-directed therapies in Alzheimer disease. Here we demonstrate that cromolyn sodium (disodium cromoglycate), a Food and Drug Administration-approved drug already in use for the treatment of asthma, efficiently inhibits the aggregation of Aß monomers into higher-order oligomers and fibrils in vitro without affecting Aß production. In vivo, the levels of soluble Aß are decreased by over 50% after only 1 week of daily intraperitoneally administered cromolyn sodium. Additional in vivo microdialysis studies also show that this compound decreases the half-life of soluble Aß in the brain. These data suggest a clear effect of a peripherally administered, Food and Drug Administration-approved medication on Aß economy, supporting further investigation of the potential long-term efficacy of cromolyn sodium in Alzheimer disease.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Cromolin Sódico/farmacología , Aprobación de Drogas , Fragmentos de Péptidos/metabolismo , Animales , Células Cultivadas , Cromolin Sódico/química , Modelos Animales de Enfermedad , Flavonoides/química , Flavonoles , Humanos , Ratones , Ratones Transgénicos , Microglía/metabolismo , Microscopía Electrónica de Transmisión , Estados Unidos , United States Food and Drug Administration
18.
J Neurochem ; 139(6): 1163-1174, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27731899

RESUMEN

Tau is a neuronal microtubule-binding protein that, in Alzheimer's disease and other neurodegenerative diseases, can form oligomeric and large fibrillar aggregates, which deposit in neurofibrillary tangles. Tau's physiological state of multimerization appears to vary across conditions, and a stable dimeric form of soluble tau has been suggested from experiments using recombinant tau in vitro. We tested if tau dimerization or oligomerization, also occurs in cells, and if soluble tau oligomers are relevant for the release and internalization of tau. We developed a sensitive tau split-luciferase assay to show the rapid intracellular formation of stable tau dimers that are released and taken up by cells. Our data further suggest that tau dimerization can be accelerated slightly by aggregation catalysts. We conclude that tau oligomers are a stable physiological form of tau, and that tau oligomerization does not necessarily lead to tau aggregation.


Asunto(s)
Multimerización de Proteína/fisiología , Proteínas tau/metabolismo , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Ratones , Ratones Transgénicos , Ovillos Neurofibrilares/metabolismo , Unión Proteica/fisiología , Proteínas tau/toxicidad
19.
Eur J Neurosci ; 44(12): 3056-3066, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27748574

RESUMEN

Alzheimer's disease is characterized by the presence of aggregates of amyloid beta (Aß) in senile plaques and tau in neurofibrillary tangles, as well as marked neuron and synapse loss. Of these pathological changes, synapse loss correlates most strongly with cognitive decline. Synapse loss occurs prominently around plaques due to accumulations of oligomeric Aß. Recent evidence suggests that tau may also play a role in synapse loss but the interactions of Aß and tau in synapse loss remain to be determined. In this study, we generated a novel transgenic mouse line, the APP/PS1/rTg21221 line, by crossing APP/PS1 mice, which develop Aß-plaques and synapse loss, with rTg21221 mice, which overexpress wild-type human tau. When compared to the APP/PS1 mice without human tau, the cross-sectional area of ThioS+ dense core plaques was increased by ~50%. Along with increased plaque size, we observed an increase in plaque-associated dystrophic neurites containing misfolded tau, but there was no exacerbation of neurite curvature or local neuron loss around plaques. Array tomography analysis similarly revealed no worsening of synapse loss around plaques, and no change in the accumulation of Aß at synapses. Together, these results indicate that adding human wild-type tau exacerbates plaque pathology and neurite deformation but does not exacerbate plaque-associated synapse loss.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Modelos Animales de Enfermedad , Placa Amiloide/metabolismo , Sinapsis/metabolismo , Sinapsis/patología , Proteínas tau/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Astrocitos/metabolismo , Humanos , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Fosforilación , Presenilina-1/genética , Presenilina-1/metabolismo , Proteínas tau/genética
20.
Proc Natl Acad Sci U S A ; 110(4): E313-21, 2013 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-23269837

RESUMEN

The structure and properties of amyloid-like Tau fibrils accumulating in neurodegenerative diseases have been debated for decades. Although the core of Tau fibrils assembles from short ß-strands, the properties of the much longer unstructured Tau domains protruding from the fibril core remain largely obscure. Applying immunogold transmission EM, and force-volume atomic force microscopy (AFM), we imaged human Tau fibrils at high resolution and simultaneously mapped their mechanical and adhesive properties. Tau fibrils showed a ≈ 16-nm-thick fuzzy coat that resembles a two-layered polyelectrolyte brush, which is formed by the unstructured short C-terminal and long N-terminal Tau domains. The mechanical and adhesive properties of the fuzzy coat are modulated by electrolytes and pH, and thus by the cellular environment. These unique properties of the fuzzy coat help in understanding how Tau fibrils disturb cellular interactions and accumulate in neurofibrillary tangles.


Asunto(s)
Proteínas tau/química , Proteínas tau/ultraestructura , Adhesividad , Fenómenos Biomecánicos , Electrólitos/química , Humanos , Concentración de Iones de Hidrógeno , Microscopía de Fuerza Atómica , Microscopía Inmunoelectrónica , Modelos Moleculares , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/ultraestructura , Proteínas tau/genética , Proteínas tau/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA