Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 164
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 156(5): 963-74, 2014 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-24581495

RESUMEN

Protein folding in the cell relies on the orchestrated action of conserved families of molecular chaperones, the Hsp70 and Hsp90 systems. Hsp70 acts early and Hsp90 late in the folding path, yet the molecular basis of this timing is enigmatic, mainly because the substrate specificity of Hsp90 is poorly understood. Here, we obtained a structural model of Hsp90 in complex with its natural disease-associated substrate, the intrinsically disordered Tau protein. Hsp90 binds to a broad region in Tau that includes the aggregation-prone repeats. Complementarily, a 106-Å-long substrate-binding interface in Hsp90 enables many low-affinity contacts. This allows recognition of scattered hydrophobic residues in late folding intermediates that remain after early burial of the Hsp70 sites. Our model resolves the paradox of how Hsp90 specifically selects for late folding intermediates but also for some intrinsically disordered proteins-through the eyes of Hsp90 they look the same.


Asunto(s)
Proteínas tau/química , Enfermedad de Alzheimer/tratamiento farmacológico , Secuencia de Aminoácidos , Proteínas HSP90 de Choque Térmico/química , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Pliegue de Proteína , Dispersión del Ángulo Pequeño , Difracción de Rayos X , Proteínas tau/metabolismo
2.
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
3.
Cell ; 142(3): 356-8, 2010 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-20691893

RESUMEN

The axonal protein tau and amyloid beta-peptide (Abeta) are key players in the pathogenesis of Alzheimer's disease, and tau mediates Abeta toxicity, but it is not clear how. Ittner et al. (2010) now report an unexpected physiological function for tau in neuronal dendrites that may explain how tau mediates Abeta toxicity.

4.
J Biol Chem ; 298(4): 101774, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35218773

RESUMEN

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.


Asunto(s)
Lectinas Tipo C , Proteínas de la Membrana , Proteómica , Proteínas tau , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Expresión Génica , Humanos , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Unión Proteica , Proteínas tau/genética , Proteínas tau/metabolismo
5.
Chemistry ; 29(17): e202203493, 2023 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-36579699

RESUMEN

Reorientational dynamics of intrinsically disordered proteins (IDPs) contain multiple motions often clustered around three motional modes: ultrafast librational motions of amide groups, fast local backbone conformational fluctuations and slow chain segmental motions. This dynamic picture is mainly based on 15 N NMR relaxation studies of IDPs at relatively low temperatures where the amide-water proton exchange rates are sufficiently small. Less is known, however, about the dynamics of IDPs at more physiological temperatures. Here, we investigate protein dynamics in a 441-residue long IDP, tau protein, in the temperature range from 0-25 °C, using 15 N NMR relaxation rates and spectral density analysis. While at these temperatures relaxation rates are still better described in terms of amide group librational motions, local backbone dynamics and chain segmental motions, the temperature-dependent trend of spectral densities suggests that the timescales of fast backbone conformational fluctuations and slower chain segmental motions might become inseparable at higher temperatures. Our data demonstrate the remarkable dynamic plasticity of this prototypical IDP and highlight the need for dynamic studies of IDPs at multiple temperatures.


Asunto(s)
Proteínas Intrínsecamente Desordenadas , Proteínas tau , Temperatura , Conformación Proteica , Espectroscopía de Resonancia Magnética , Proteínas Intrínsecamente Desordenadas/química , Amidas
6.
Int J Mol Sci ; 24(11)2023 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-37298211

RESUMEN

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.


Asunto(s)
Receptor de Adenosina A1 , Tauopatías , Ratones , Animales , Ratones Transgénicos , Receptor de Adenosina A1/genética , Receptor de Adenosina A1/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Hipocampo/metabolismo , Tauopatías/tratamiento farmacológico , Tauopatías/genética , Tauopatías/metabolismo , Cognición , Modelos Animales de Enfermedad
7.
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
8.
J Biol Chem ; 295(52): 18213-18225, 2020 12 25.
Artículo en Inglés | MEDLINE | ID: mdl-33106314

RESUMEN

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.


Asunto(s)
Cromatografía Liquida/métodos , Procesamiento Proteico-Postraduccional , Espectrometría de Masas en Tándem/métodos , Proteínas tau/química , Proteínas tau/metabolismo , Secuencia de Aminoácidos , Humanos , Fosforilación , Homología de Secuencia
9.
Chembiochem ; 22(21): 3049-3059, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34375027

RESUMEN

Alzheimer's disease and other Tauopathies are associated with neurofibrillary tangles composed of Tau protein, as well as toxic Tau oligomers. Therefore, inhibitors of pathological Tau aggregation are potentially useful candidates for future therapies targeting Tauopathies. Two hexapeptides within Tau, designated PHF6* (275-VQIINK-280) and PHF6 (306-VQIVYK-311), are known to promote Tau aggregation. Recently, the PHF6* segment has been described as the more potent driver of Tau aggregation. We therefore employed mirror-image phage display with a large peptide library to identify PHF6* fibril binding peptides consisting of D-enantiomeric amino acids. The suitability of D-enantiomeric peptides for in vivo applications, which are protease stable and less immunogenic than L-peptides, has already been demonstrated. The identified D-enantiomeric peptide MMD3 and its retro-inverso form, designated MMD3rev, inhibited in vitro fibrillization of the PHF6* peptide, the repeat domain of Tau as well as full-length Tau. Dynamic light scattering, pelleting assays and atomic force microscopy demonstrated that MMD3 prevents the formation of tau ß-sheet-rich fibrils by diverting Tau into large amorphous aggregates. NMR data suggest that the D-enantiomeric peptides bound to Tau monomers with rather low affinity, but ELISA (enzyme-linked immunosorbent assay) data demonstrated binding to PHF6* and full length Tau fibrils. In addition, molecular insight into the binding mode of MMD3 to PHF6* fibrils were gained by in silico modelling. The identified PHF6*-targeting peptides were able to penetrate cells. The study establishes PHF6* fibril binding peptides consisting of D-enantiomeric amino acids as potential molecules for therapeutic and diagnostic applications in AD research.


Asunto(s)
Péptidos/farmacología , Proteínas tau/antagonistas & inhibidores , Humanos , Biblioteca de Péptidos , Péptidos/química , Agregado de Proteínas/efectos de los fármacos , Proteínas tau/metabolismo
10.
Nat Rev Neurosci ; 17(1): 5-21, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26631930

RESUMEN

Tau is a microtubule-associated protein that has a role in stabilizing neuronal microtubules and thus in promoting axonal outgrowth. Structurally, tau is a natively unfolded protein, is highly soluble and shows little tendency for aggregation. However, tau aggregation is characteristic of several neurodegenerative diseases known as tauopathies. The mechanisms underlying tau pathology and tau-mediated neurodegeneration are debated, but considerable progress has been made in the field of tau research in recent years, including the identification of new physiological roles for tau in the brain. Here, we review the expression, post-translational modifications and functions of tau in physiology and in pathophysiology.


Asunto(s)
Tauopatías/metabolismo , Proteínas tau/metabolismo , Animales , Humanos , Fosforilación/genética , Procesamiento Proteico-Postraduccional , Tauopatías/patología
11.
Angew Chem Int Ed Engl ; 60(2): 726-730, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33017094

RESUMEN

Cellular condensation of intrinsically disordered proteins (IDPs) through liquid-liquid phase separation (LLPS) allows dynamic compartmentalization and regulation of biological processes. The IDP tau, which promotes the assembly of microtubules and is hyperphosphorylated in Alzheimer's disease, undergoes LLPS in solution and on the surface of microtubules. Little is known, however, about the influence of tau phosphorylation on its ability to nucleate microtubule bundles in conditions of tau LLPS. Herein, we show that unmodified tau as well as tau phosphorylated at disease-associated epitopes condense into liquid-like droplets. Although tubulin partitioned into and reached high concentrations inside all tau droplets, it was unable to grow into microtubules form the inside of droplets formed by tau phosphorylated at the AT180 epitope (T231/S235). In contrast, neither phosphorylation of tau in the repeat domain nor at its tyrosine residues inhibited the assembly of tubulin from tau droplets. Because LLPS of IDPs has been shown to promote different types of cytoskeletal assembly, our study suggests that IDP phosphorylation might be a broadly used mechanism for the modulation of condensate-mediated cytoskeletal assembly.


Asunto(s)
Tubulina (Proteína)/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Humanos , Microscopía Fluorescente , Mutagénesis Sitio-Dirigida , Resonancia Magnética Nuclear Biomolecular , Fosforilación , Unión Proteica , Tubulina (Proteína)/química , Proteínas tau/química , Proteínas tau/genética
12.
PLoS Biol ; 15(6): e2001336, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28654636

RESUMEN

The accumulation of amyloidogenic proteins is a pathological hallmark of neurodegenerative disorders. The aberrant accumulation of the microtubule associating protein tau (MAPT, tau) into toxic oligomers and amyloid deposits is a primary pathology in tauopathies, the most common of which is Alzheimer's disease (AD). Intrinsically disordered proteins, like tau, are enriched with proline residues that regulate both secondary structure and aggregation propensity. The orientation of proline residues is regulated by cis/trans peptidyl-prolyl isomerases (PPIases). Here we show that cyclophilin 40 (CyP40), a PPIase, dissolves tau amyloids in vitro. Additionally, CyP40 ameliorated silver-positive and oligomeric tau species in a mouse model of tau accumulation, preserving neuronal health and cognition. Nuclear magnetic resonance (NMR) revealed that CyP40 interacts with tau at sites rich in proline residues. CyP40 was also able to interact with and disaggregate other aggregating proteins that contain prolines. Moreover, CyP40 lacking PPIase activity prevented its capacity for disaggregation in vitro. Finally, we describe a unique structural property of CyP40 that may permit disaggregation to occur in an energy-independent manner. This study identifies a novel human protein disaggregase and, for the first time, demonstrates its capacity to dissolve intracellular amyloids.


Asunto(s)
Amiloide/metabolismo , Ciclofilinas/metabolismo , Enfermedades Neurodegenerativas/metabolismo , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Amiloide/genética , Amiloide/ultraestructura , Animales , Western Blotting , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/fisiopatología , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/fisiopatología , Peptidil-Prolil Isomerasa F , Ciclofilinas/genética , Ciclosporina/farmacología , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Humanos , Masculino , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Enfermedades Neurodegenerativas/genética , Agregado de Proteínas/efectos de los fármacos , Agregación Patológica de Proteínas , Tauopatías/genética , Tauopatías/metabolismo , alfa-Sinucleína/genética , Proteínas tau/genética
13.
Adv Exp Med Biol ; 1184: 69-77, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32096029

RESUMEN

Tau is a microtubule-associated protein (MAP) that is mainly sorted into the axons in physiological conditions, but missorted in Alzheimer Disease and related tauopathies. The mechanism(s) of axonal targeting of Tau protein are still a matter of debate. Several possibilities for the axonal localization of Tau protein have been proposed: (1) Targeting of Tau mRNA into axons which is then translated locally. (2) Preferred axonal translation of Tau mRNA. (3) Specific dendritic degradation of Tau protein. (4) Active axonal sorting of somatically translated Tau protein. (5) Axonal retention of Tau protein by specific association of Tau protein with axonal structures, namely particularly modified microtubules. (6) Restriction of Tau diffusion by a selective filter function of the Axon Initial Segment (AIS). In our research we focused on the Tau Diffusion Barrier (TDB), located within the AIS, which controls anterograde and retrograde propagation of Tau. It shows both sensitivity to size of the Tau protein isoforms, and to disruption of the molecular structure of the AIS. Here, we review proposed mechanisms of axonal targeting of Tau and potential influences of the TDB/AIS on the subcellular distribution of Tau.


Asunto(s)
Segmento Inicial del Axón , Polaridad Celular , Proteínas tau/metabolismo , Humanos , Microtúbulos/metabolismo
14.
Proc Natl Acad Sci U S A ; 113(41): 11597-11602, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27671637

RESUMEN

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.


Asunto(s)
Antagonistas del Receptor de Adenosina A1/farmacología , Axones/metabolismo , Eliminación de Secuencia , Xantinas/farmacología , Proteínas tau/genética , Adenosina Trifosfato/metabolismo , Animales , Espinas Dendríticas/efectos de los fármacos , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Expresión Génica , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Memoria a Largo Plazo/efectos de los fármacos , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Neuronas/metabolismo , Neuronas/patología , Fosforilación , Agregado de Proteínas , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/metabolismo , Transmisión Sináptica/efectos de los fármacos , Tauopatías/tratamiento farmacológico , Tauopatías/genética , Tauopatías/metabolismo , Tauopatías/patología
15.
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
16.
J Biol Chem ; 292(29): 12192-12207, 2017 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-28536263

RESUMEN

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.


Asunto(s)
Segmento Inicial del Axón/metabolismo , Corteza Cerebral/metabolismo , Dendritas/metabolismo , Microtúbulos/metabolismo , Neuronas/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Segmento Inicial del Axón/patología , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/patología , Dendritas/patología , Difusión , Embrión de Mamíferos/citología , Eliminación de Gen , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Microtúbulos/patología , Mutagénesis Insercional , Neuronas/citología , Neuronas/patología , Dominios y Motivos de Interacción de Proteínas , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte de Proteínas , Interferencia de ARN , Ratas Wistar , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Secuencias Repetitivas de Aminoácido , Proteínas tau/antagonistas & inhibidores , Proteínas tau/química , Proteínas tau/genética
17.
Neurobiol Dis ; 117: 189-202, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29894752

RESUMEN

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.


Asunto(s)
Proteínas de Caenorhabditis elegans/biosíntesis , Señalización del Calcio/fisiología , Ácido Glutámico/metabolismo , Degeneración Nerviosa/metabolismo , Tauopatías/metabolismo , Proteínas tau/biosíntesis , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Ácido Glutámico/genética , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Tauopatías/genética , Tauopatías/patología , Proteínas tau/genética
18.
FASEB J ; 31(12): 5137-5148, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29191965

RESUMEN

One of the hallmarks of the tauopathies, which include the neurodegenerative disorders, such as Alzheimer disease (AD), corticobasal degeneration, frontotemporal dementia, and progressive supranuclear palsy (PSP), is the abnormal accumulation of post-translationally modified, insoluble tau. The result is a loss of neurons, decreased mental function, and complete dependence of patients on others. Aggregation of tau, which under physiologic conditions is a highly soluble protein, is thought to be central to the pathogenesis of these diseases. Indeed one of the strongest lines of evidence is the MAPT gene polymorphisms that lead to the familial forms of tauopathy. Extensive research in animal models over the years has contributed some of the most important findings regarding the pathogenesis of these diseases. Despite this, the precise molecular mechanisms that lead to abnormal tau folding, accumulation, and spreading remain unknown. Owing to the fact that most of the biochemical pathways are conserved, Caenorhabditis elegans provides an alternative approach to identify cellular mechanisms and druggable genes that operate in such disorders. Many human genes implicated in neurodegenerative diseases have counterparts in C. elegans, making it an excellent model in which to study their pathogenesis. In this article, we review some of the important findings gained from C. elegans tauopathy models.-Pir, G. J., Choudhary, B., Mandelkow, E. Caenorhabditiselegans models of tauopathy.


Asunto(s)
Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/patogenicidad , Tauopatías/metabolismo , Tauopatías/patología , Animales , Caenorhabditis elegans/genética , Modelos Animales de Enfermedad , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Tauopatías/genética , Proteínas tau/genética , Proteínas tau/metabolismo
19.
EMBO Rep ; 17(4): 552-69, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26931569

RESUMEN

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.


Asunto(s)
Región CA3 Hipocampal/metabolismo , Región CA3 Hipocampal/patología , Demencia Frontotemporal/fisiopatología , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Animales , Calcio/metabolismo , Medios de Cultivo/química , Modelos Animales de Enfermedad , Ácido Glutámico/análisis , Humanos , Ratones , Ratones Transgénicos , Mutación , Plasticidad Neuronal , Neuronas/química , Neuronas/patología , Técnicas de Cultivo de Órganos , Transmisión Sináptica , Proteínas tau/química
20.
Proc Natl Acad Sci U S A ; 112(24): 7501-6, 2015 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-26034266

RESUMEN

The structure, dynamic behavior, and spatial organization of microtubules are regulated by microtubule-associated proteins. An important microtubule-associated protein is the protein Tau, because its microtubule interaction is impaired in the course of Alzheimer's disease and several other neurodegenerative diseases. Here, we show that Tau binds to microtubules by using small groups of evolutionary conserved residues. The binding sites are formed by residues that are essential for the pathological aggregation of Tau, suggesting competition between physiological interaction and pathogenic misfolding. Tau residues in between the microtubule-binding sites remain flexible when Tau is bound to microtubules in agreement with a highly dynamic nature of the Tau-microtubule interaction. By binding at the interface between tubulin heterodimers, Tau uses a conserved mechanism of microtubule polymerization and, thus, regulation of axonal stability and cell morphology.


Asunto(s)
Microtúbulos/metabolismo , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Proteínas tau/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Unión Competitiva , Fenómenos Biofísicos , Humanos , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Porcinos , Vinblastina/metabolismo , Proteínas tau/química , Proteínas tau/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA