Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 1679, 2024 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-38396035

RESUMEN

Tauopathies such as Alzheimer's disease are characterized by aggregation and increased phosphorylation of the microtubule-associated protein tau. Tau's pathological changes are closely linked to neurodegeneration, making tau a prime candidate for intervention. We developed an approach to monitor pathological changes of aggregation-prone human tau in living neurons. We identified 2-phenyloxazole (PHOX) derivatives as putative polypharmacological small molecules that interact with tau and modulate tau kinases. We found that PHOX15 inhibits tau aggregation, restores tau's physiological microtubule interaction, and reduces tau phosphorylation at disease-relevant sites. Molecular dynamics simulations highlight cryptic channel-like pockets crossing tau protofilaments and suggest that PHOX15 binding reduces the protofilament's ability to adopt a PHF-like conformation by modifying a key glycine triad. Our data demonstrate that live-cell imaging of a tauopathy model enables screening of compounds that modulate tau-microtubule interaction and allows identification of a promising polypharmacological drug candidate that simultaneously inhibits tau aggregation and reduces tau phosphorylation.


Asunto(s)
Enfermedad de Alzheimer , Tauopatías , Humanos , Tauopatías/tratamiento farmacológico , Tauopatías/metabolismo , Proteínas tau/metabolismo , Microtúbulos/metabolismo , Enfermedad de Alzheimer/metabolismo , Citoesqueleto/metabolismo , Fosforilación
2.
Cytoskeleton (Hoboken) ; 81(1): 47-52, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37694806

RESUMEN

Tau is a microtubule-associated protein that is enriched in the axonal process of neurons. Post-translational modifications of tau have been implicated in the development of tauopathies characterized by defects in axonal transport, neuronal atrophy, and microtubule disassembly. Although tau is almost quantitatively bound to microtubules under physiological conditions, it does not significantly affect axonal transport. Furthermore, acute or chronic tau deficiency does not result in significant destabilization of neuronal microtubules, challenging the classical view that disease-related tau modifications directly cause axonal microtubule collapse. Here, we discuss how the rapid interaction kinetics of the tau-microtubule interaction, which we previously termed the kiss-and-hop interaction, explains why tau does not affect microtubule-dependent axonal transport but still allows tau to modulate microtubule polymerization. In contrast, tau modifications that slow down the kinetics of the tau-microtubule interaction and increase the residence time of tau at a microtubule interaction site can disrupt axonal transport and cause dendritic atrophy. We discuss the consequences of such a gain-of-toxicity mechanism in terms of the development of disease-modulating drugs that target the tau protein.


Asunto(s)
Tauopatías , Proteínas tau , Humanos , Transporte Axonal , Microtúbulos/metabolismo , Tauopatías/metabolismo , Atrofia/metabolismo
3.
Brain Res Bull ; 194: 13-22, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36626968

RESUMEN

The unique morphology of neurons consists of a long axon and a highly variable arbour of dendritic processes, which assort neuronal cells into the main classes. The dendritic tree serves as the main domain for receiving synaptic input. Therefore, to maintain the structure and to be able to plastically change according to the incoming stimuli, molecules and organelles need to be readily available. This is achieved mainly via bi-directional transport of cargo along the microtubule lattices. Analysis of dendritic transport is lagging behind the investigation of axonal transport. Moreover, addressing transport mechanisms in tissue environment is very challenging and, therefore, rare. We employed high-speed volumetric lattice light-sheet microscopy and single particle tracking of truncated KIF1A motor protein lacking the cargo-binding domain. We focused our analysis on dendritic processes of CA1 pyramidal neurons in cultured hippocampal tissue. Analysis of individual trajectories revealed detailed information about stalling and high variability in movement and speed, and biased directionality of KIF1A. Furthermore, we could also observe KIF1A shortly entering into dendritic spines. We provide a workflow to analyse variations in the speed and direction of motor protein movement in dendrites that are either intrinsic properties of the motor domain or depend on the structure and modification of the microtubule trails.


Asunto(s)
Espinas Dendríticas , Cinesinas , Microscopía , Ratones , Axones/metabolismo , Dendritas , Espinas Dendríticas/metabolismo , Hipocampo/metabolismo , Cinesinas/metabolismo , Cinesinas/fisiología , Microscopía/métodos , Neuronas/metabolismo , Animales
4.
Brain Res Bull ; 190: 234-243, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36244582

RESUMEN

Microtubules are essential for the development of neurons and the regulation of their structural plasticity. Microtubules also provide the structural basis for the long-distance transport of cargo. Various factors influence the organization and dynamics of neuronal microtubules, and disturbance of microtubule regulation is thought to play a central role in neurodegenerative diseases. However, imaging and quantitative assessment of the microtubule organization in the densely packed neuronal processes is challenging. The development of super-resolution techniques combined with the use of nanobodies offers new possibilities to visualize microtubules in neurites in high resolution. In combination with recently developed computational analysis tools, this allows automated quantification of neuronal microtubule organization with high precision. Here we have implemented three-dimensional DNA-PAINT (Point Accumulation in Nanoscale Topography), a single-molecule localization microscopy (SMLM) technique, which allows us to acquire 3D arrays of the microtubule lattice in axons of model neurons (neuronally differentiated PC12 cells) and dendrites of primary neurons. For the quantitative analysis of the microtubule organization, we used the open-source software package SMLM image filament extractor (SIFNE). We found that treatment with nanomolar concentrations of the microtubule-targeting drug epothilone D (EpoD) increased microtubule density in axon-like processes of model neurons and shifted the microtubule length distribution to shorter ones, with a mean microtubule length of 2.39 µm (without EpoD) and 1.98 µm (with EpoD). We also observed a significant decrease in microtubule straightness after EpoD treatment. The changes in microtubule density were consistent with live-cell imaging measurements of ensemble microtubule dynamics using a previously established Fluorescence Decay After Photoactivation (FDAP) assay. For comparison, we determined the organization of the microtubule array in dendrites of primary hippocampal neurons. We observed that dendritic microtubules have a very similar length distribution and straightness compared to microtubules in axon-like processes of a neuronal cell line. Our data show that super-resolution imaging of microtubules followed by algorithm-based image analysis represents a powerful tool to quantitatively assess changes in microtubule organization in neuronal processes, useful to determine the effect of microtubule-modulating conditions. We also provide evidence that the approach is robust and can be applied to neuronal cell lines or primary neurons, both after incorporation of labeled tubulin and by anti-tubulin antibody staining.


Asunto(s)
Axones , Microtúbulos , Ratas , Animales , Microtúbulos/metabolismo , Axones/metabolismo , Neuronas/metabolismo , Células PC12
5.
Mol Psychiatry ; 27(7): 3010-3023, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35393558

RESUMEN

The microtubule-associated protein tau plays a central role in tauopathies such as Alzheimer's disease (AD). The exact molecular mechanisms underlying tau toxicity are unclear, but aging is irrefutably the biggest risk factor. This raises the question of how cellular senescence affects the function of tau as a microtubule regulator. Here we report that the proportion of tau that is proteolytically cleaved at the caspase-3 site (TauC3) doubles in the hippocampus of senescent mice. TauC3 is also elevated in AD patients. Through quantitative live-cell imaging, we show that TauC3 has a drastically reduced dynamics of its microtubule interaction. Single-molecule tracking of tau confirmed that TauC3 has a longer residence time on axonal microtubules. The reduced dynamics of the TauC3-microtubule interaction correlated with a decreased transport of mitochondria, a reduced processivity of APP-vesicle transport and an induction of region-specific dendritic atrophy in CA1 neurons of the hippocampus. The microtubule-targeting drug Epothilone D normalized the interaction of TauC3 with microtubules and modulated the transport of APP-vesicles dependent on the presence of overexpressed human tau. The results indicate a novel toxic gain of function, in which a post-translational modification of tau changes the dynamics of the tau-microtubule interaction and thus leads to axonal transport defects and neuronal degeneration. The data also introduce microtubule-targeting drugs as pharmacological modifiers of the tau-microtubule interaction with the potential to restore the physiological interaction of pathologically altered tau with microtubules.


Asunto(s)
Enfermedad de Alzheimer , Tauopatías , Enfermedad de Alzheimer/metabolismo , Animales , Transporte Axonal , Caspasas/metabolismo , Mutación con Ganancia de Función , Humanos , Ratones , Microtúbulos/metabolismo , Tauopatías/metabolismo , Proteínas tau/metabolismo
6.
Curr Neuropharmacol ; 20(4): 782-798, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34852744

RESUMEN

The microtubule skeleton plays an essential role in nerve cells as the most important structural determinant of morphology and as a highway for axonal transport processes. Many neurodegenerative diseases are characterized by changes in the structure and organization of microtubules and microtubule-regulating proteins such as the microtubule-associated protein tau, which exhibits characteristic changes in a whole class of diseases collectively referred to as tauopathies. Changes in the dynamics of microtubules appear to occur early under neurodegenerative conditions and are also likely to contribute to age-related dysfunction of neurons. Thus, modulating microtubule dynamics and correcting impaired microtubule stability can be a useful neuroprotective strategy to counteract the disruption of the microtubule system in disease and aging. In this article, we review current microtubule- directed approaches for the treatment of neurodegenerative diseases with microtubules as a drug target, tau as a drug target, and post-translational modifications as potential modifiers of the microtubule system. We discuss limitations of the approaches that can be traced back to the rather unspecific mechanism of action, which causes undesirable side effects in non-neuronal cell types or which are due to the disruption of non-microtubule-related interactions. We also develop some thoughts on how the specificity of the approaches can be improved and what further targets could be used for modulating substances.


Asunto(s)
Enfermedades Neurodegenerativas , Tauopatías , Humanos , Microtúbulos/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , Procesamiento Proteico-Postraduccional , Tauopatías/tratamiento farmacológico , Tauopatías/metabolismo
7.
Front Neurol ; 11: 590059, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193056

RESUMEN

Tau protein (MAPT) is classified as a microtubule-associated protein (MAP) and is believed to regulate the axonal microtubule arrangement. It belongs to the tau/MAP2/MAP4 family of MAPs that have a similar microtubule binding region at their carboxy-terminal half. In tauopathies, such as Alzheimer's disease, tau is distributed more in the somatodendritic compartment, where it aggregates into filamentous structures, the formation of which correlates with cognitive impairments in patients. While microtubules are the dominant interaction partners of tau under physiological conditions, tau has many additional interaction partners that can contribute to its physiological and pathological role. In particular, the amino-terminal non-microtubule binding domain (N-terminal projection region, NTR) of tau interacts with many partners that are involved in membrane organization. The NTR contains intrinsically disordered regions (IDRs) that show a strong evolutionary increase in the disorder and may have been the basis for the development of new, tau-specific interactions. In this review we discuss the functional organization of the tau protein and the special features of the tau non-microtubule binding region also in the connection with the results of Tau KO models. We consider possible physiological and pathological functions of tau's non-microtubule interactions, which could indicate that interactions mediated by tau's NTR and regulated by far-reaching functional interactions of the PRR and the extreme C-terminus of tau contribute to the pathological processes.

8.
Artículo en Inglés | MEDLINE | ID: mdl-32390822

RESUMEN

Alzheimer's disease is regarded as a synaptopathy with a long presymptomatic phase. Soluble, oligomeric amyloid-ß (Aß) is thought to play a causative role in this disease, which eventually leads to cognitive decline. However, most animal studies have employed mice expressing high levels of the Aß precursor protein (APP) transgene to drive pathology. Here, to understand how the principal neurons in different brain regions cope with moderate, chronically present levels of Aß, we employed transgenic mice expressing equal levels of mouse and human APP carrying a combination of three familial AD (FAD)-linked mutations (Swedish, Dutch, and London), that develop plaques only in old age. We analyzed dendritic spine parameters in hippocampal and cortical brain regions after targeted expression of EGFP to allow high-resolution imaging, followed by algorithm-based evaluation of mice of both sexes from adolescence to old age. We report that Aß species gradually accumulated throughout the life of APPSDL mice, but not the oligomeric forms, and that the amount of membrane-associated oligomers decreased at the onset of plaque formation. We observed an age-dependent loss of thin spines under most conditions as an indicator of a loss of synaptic plasticity in older mice. We further found that hippocampal pyramidal neurons respond to increased Aß levels by lowering spine density and shifting spine morphology, which reached significance in the CA1 subfield. In contrast, the spine density in cortical pyramidal neurons of APPSDL mice was unchanged. We also observed an increase in the protein levels of PSD-95 and Arc in the hippocampus and cortex, respectively. Our data demonstrated that increased concentrations of Aß have diverse effects on dendritic spines in the brain and suggest that hippocampal and cortical neurons have different adaptive and compensatory capacity during their lifetime. Our data also indicated that spine morphology differs between sexes in a region-specific manner.

9.
Artículo en Inglés | MEDLINE | ID: mdl-32116638

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disorder that leads to impaired memory and cognitive deficits. Spine loss as well as changes in spine morphology correlates with cognitive impairment in this neurological disorder. Many studies in animal models and ex vivo cultures indicate that amyloid ß-peptide (Aß) oligomers induce synaptic damage early during the progression of the disease. Here, in order to determine the events that initiate synaptic alterations, we acutely applied oligomeric Aß to primary hippocampal neurons and an ex vivo model of organotypic hippocampal cultures from a mouse after targeted expression of EGFP to allow high-resolution imaging and algorithm-based evaluation of spine changes. Dendritic spines were classified as thin, stubby or mushroom, based on morphology. In vivo, time-lapse imaging showed that the three spine types were relatively stable, although their stability significantly decreased after treatment with Aß oligomers. Unexpectedly, we observed that the density of total dendritic spines increased in organotypic hippocampal slices treated with Aß compared to control cultures. Specifically, the fraction of stubby spines significantly increased, while mushroom and thin spines remained unaltered. Pharmacological tools revealed that acute Aß oligomers induced spine changes through mechanisms involving CaMKII and integrin ß1 activities. Additionally, analysis of dendritic complexity based on a 3D reconstruction of the whole neuron morphology showed an increase in the apical dendrite length and branching points in CA1 organotypic hippocampal slices treated with Aß. In contrast to spines, the morphological changes were affected by integrin ß1 but not by CaMKII inhibition. Altogether, these data indicate that the Aß oligomers exhibit early dual effects by acutely enhancing dendritic complexity and spine density.

10.
Front Aging Neurosci ; 11: 256, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31619983

RESUMEN

Tau is a neuronal microtubule-associated protein (MAP) that is involved in the regulation of axonal microtubule assembly. However, as a protein with intrinsically disordered regions (IDRs), tau also interacts with many other partners in addition to microtubules. Phosphorylation at selected sites modulates tau's various intracellular interactions and regulates the properties of IDRs. In Alzheimer's disease (AD) and other tauopathies, tau exhibits pathologically increased phosphorylation (hyperphosphorylation) at selected sites and aggregates into neurofibrillary tangles (NFTs). By bioinformatics means, we tested the hypothesis that the sequence of tau has changed during the vertebrate evolution in a way that novel interactions developed and also the phosphorylation pattern was affected, which made tau prone to the development of tauopathies. We report that distinct regions of tau show functional specialization in their molecular interactions. We found that tau's amino-terminal region, which is involved in biological processes related to "membrane organization" and "regulation of apoptosis," exhibited a strong evolutionary increase in protein disorder providing the basis for the development of novel interactions. We observed that the predicted phosphorylation sites have changed during evolution in a region-specific manner, and in some cases the overall number of phosphorylation sites increased owing to the formation of clusters of phosphorylatable residues. In contrast, disease-specific hyperphosphorylated sites remained highly conserved. The data indicate that novel, non-microtubule related tau interactions developed during evolution and suggest that the biological processes, which are mediated by these interactions, are of pathological relevance. Furthermore, the data indicate that predicted phosphorylation sites in some regions of tau, including a cluster of phosphorylatable residues in the alternatively spliced exon 2, have changed during evolution. In view of the "antagonistic pleiotropy hypothesis" it may be worth to take disease-associated phosphosites with low evolutionary conservation as relevant biomarkers into consideration.

11.
Transl Psychiatry ; 9(1): 7, 2019 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-30664629

RESUMEN

In humans, genetic variants of DLGAP1-4 have been linked with neuropsychiatric conditions, including autism spectrum disorder (ASD). While these findings implicate the encoded postsynaptic proteins, SAPAP1-4, in the etiology of neuropsychiatric conditions, underlying neurobiological mechanisms are unknown. To assess the contribution of SAPAP4 to these disorders, we characterized SAPAP4-deficient mice. Our study reveals that the loss of SAPAP4 triggers profound behavioural abnormalities, including cognitive deficits combined with impaired vocal communication and social interaction, phenotypes reminiscent of ASD in humans. These behavioural alterations of SAPAP4-deficient mice are associated with dramatic changes in synapse morphology, function and plasticity, indicating that SAPAP4 is critical for the development of functional neuronal networks and that mutations in the corresponding human gene, DLGAP4, may cause deficits in social and cognitive functioning relevant to ASD-like neurodevelopmental disorders.


Asunto(s)
Trastorno del Espectro Autista/genética , Disfunción Cognitiva/genética , Proteínas del Tejido Nervioso/genética , Proteínas Asociadas a SAP90-PSD95/genética , Animales , Conducta Animal , Modelos Animales de Enfermedad , Femenino , Relaciones Interpersonales , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Conducta Social , Sinapsis/metabolismo
12.
J Biol Chem ; 293(21): 8065-8076, 2018 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-29636414

RESUMEN

During neuronal development, the microtubule-associated protein tau becomes enriched in the axon, where it remains concentrated in the healthy brain. In tauopathies such as Alzheimer's disease, tau redistributes from the axon to the somatodendritic compartment. However, the cellular mechanism that regulates tau's localization remains unclear. We report here that tau interacts with the Ca2+-regulated plasma membrane-binding protein annexin A2 (AnxA2) via tau's extreme N terminus encoded by the first exon (E1). Bioinformatics analysis identified two conserved eight-amino-acids-long motifs within E1 in mammals. Using a heterologous yeast system, we found that disease-related mutations and pseudophosphorylation of Tyr-18, located within E1 but outside of the two conserved regions, do not influence tau's interaction with AnxA2. We further observed that tau interacts with the core domain of AnxA2 in a Ca2+-induced open conformation and interacts also with AnxA6. Moreover, lack of E1 moderately increased tau's association rate to microtubules, consistent with the supposition that the presence of the tau-annexin interaction reduces the availability of tau to interact with microtubules. Of note, intracellular competition through overexpression of E1-containing constructs reduced tau's axonal enrichment in primary neurons. Our results suggest that the E1-mediated tau-annexin interaction contributes to the enrichment of tau in the axon and is involved in its redistribution in pathological conditions.


Asunto(s)
Anexina A2/metabolismo , Anexina A6/metabolismo , Axones/metabolismo , Microtúbulos/metabolismo , Proteínas tau/metabolismo , Animales , Anexina A2/genética , Anexina A6/genética , Membrana Celular/metabolismo , Células Cultivadas , Humanos , Ratones , Ratones Endogámicos C57BL , Células PC12 , Fosforilación , Unión Proteica , Ratas , Proteínas tau/genética
13.
J Neurochem ; 143(4): 409-417, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28267200

RESUMEN

Alzheimer's disease (AD) is the most common neurodegenerative disorder and is, on a histopathological level, characterized by the presence of extracellular amyloid plaques composed of the protein fragment Aß, and intracellular neurofibrillary tangles, which contain the microtubule-associated protein tau in a hyperphosphorylated state. In AD defects in microtubule (MT) assembly and organization have also been reported; however, it is unclear whether MT abnormalities have a causal and early role in the disease process or represent a common end point downstream of the neurodegenerative cascade. Recent evidence indicates that microtubule-stabilizing drugs prevent axonopathy in animal models of tauopathies and reverse Aß-induced loss of synaptic connectivity in an ex vivo model of amyloidosis. This could suggest that MT dysfunction connects some of the degenerative events and provides a useful target to simultaneously prevent several neurodegenerative processes in AD. Here, we describe how changes in the structure and dynamics of MTs are involved in the different aspects of the neurodegenerative triad of AD. We discuss evidence that MTs are affected both by tau-dependent and tau-independent mechanisms but appear to be regulated in a distinct way in different neuronal compartments. We argue that modulation of MT dynamics could be of potential benefit but needs to be precisely controlled in a cell and compartment-specific manner to avoid harmful side effects. This article is part of the series "Beyond Amyloid".


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Microtúbulos/metabolismo , Microtúbulos/patología , Péptidos beta-Amiloides/metabolismo , Animales , Dendritas/metabolismo , Dendritas/patología , Humanos , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Placa Amiloide/metabolismo , Placa Amiloide/patología , Proteínas tau/metabolismo
14.
Mol Cell Neurosci ; 84: 132-141, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28318914

RESUMEN

Tau is a microtubule-associated neuronal protein, whose primary role was long thought to regulate axonal microtubule assembly. Tau is subject to many posttranslational modifications and can aggregate into neurofibrillary tangles, which are considered to be a hallmark of several neurodegenerative diseases collectively called "tauopathies". The most common tauopathy is Alzheimer's disease, where tau pathology correlates with sites of neurodegeneration. Tau belongs to the class of intrinsically disordered proteins, which are known to interact with many partners and are considered to be involved in various signaling, regulation and recognition processes. Thus more recent evidence indicates that tau functionally interacts with many proteins and different cellular structures, which may have an important physiological role and may be involved in neurodegenerative processes. Furthermore, tau can be released from neurons and exert functional effects on other cells. This review article weighs the evidence that tau has subtle but important systemic effects on neuronal network function by maintaining physiological neuronal transmission and synaptic plasticity, which are possibly independent from tau's microtubule modulating activities. Implications for tau-based therapeutic approaches are discussed.


Asunto(s)
Enfermedad de Alzheimer/terapia , Encéfalo/metabolismo , Ovillos Neurofibrilares/metabolismo , Tauopatías/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Humanos , Neuronas/metabolismo
15.
Neuropharmacology ; 113(Pt A): 434-444, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27773645

RESUMEN

DMSO has a widespread use as a vehicle for water-insoluble therapeutic drug candidates but may also exert disease-relevant pharmacological effects by itself. However, its influence on the CNS has hardly been addressed. Here we examined the brain structure and function following chronic exposure to low DMSO dose at a paradigm with flawed synaptic connectivity in a preclinical transgenic mouse model for Alzheimer's disease (APPSDL mice). DMSO treatment increased spine density in a region-specific manner in the hippocampus of APPSDL mice ex vivo and in vivo. Moreover, DMSO exhibited clear influence on the behavior of this mouse line by enhancing hippocampal-dependent spatial memory accuracy, modulating hippocampal-independent olfactory habituation and displaying anxiolytic effect. Despite that most of the action of DMSO was observed in animals with elevated Aß levels, the drug did not exert its function via decreasing the oligomeric Aß species. However, challenging organotypic hippocampal slice cultures with NMDA receptor antagonist MK-801 recapitulated the effect of DMSO on spine density, indicating a tuning influence of DMSO on receptor signalization. Our findings demonstrate that DMSO should be considered as a true bioactive compound, which has the potential to be a beneficial adjuvant to counteract Aß-mediated synaptotoxicity and behavioral impairment.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Espinas Dendríticas/efectos de los fármacos , Dimetilsulfóxido/uso terapéutico , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Espinas Dendríticas/patología , Dimetilsulfóxido/farmacología , Evaluación Preclínica de Medicamentos/métodos , Depuradores de Radicales Libres/farmacología , Depuradores de Radicales Libres/uso terapéutico , Hipocampo/patología , Masculino , Ratones , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Memoria Espacial/efectos de los fármacos , Memoria Espacial/fisiología
16.
Mol Biol Cell ; 27(22): 3537-3549, 2016 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-27582388

RESUMEN

A current challenge of cell biology is to investigate molecular interactions in subcellular compartments of living cells to overcome the artificial character of in vitro studies. To dissect the interaction of the neuronal microtubule (MT)-associated protein tau with MTs in axon-like processes, we used a refined fluorescence decay after photoactivation approach and single-molecule tracking. We found that isoform variation had only a minor influence on the tau-MT interaction, whereas the presence of a C-terminal pseudorepeat region (PRR) greatly increased MT binding by a greater-than-sixfold reduction of the dissociation rate. Bioinformatic analysis revealed that the PRR contained a highly conserved motif of 18 amino acids. Disease-associated tau mutations in the PRR (K369I, G389R) did not influence apparent MT binding but increased its dynamicity. Simulation of disease-like tau hyperphosphorylation dramatically diminished the tau-MT interaction by a greater-than-fivefold decrease of the association rate with no major change in the dissociation rate. Apparent binding of tau to MTs was similar in axons and dendrites but more sensitive to increased phosphorylation in axons. Our data indicate that under the conditions of high MT density that prevail in the axon, tau's MT binding and localization are crucially affected by the presence of the PRR and tau hyperphosphorylation.


Asunto(s)
Proteínas tau/metabolismo , Secuencia de Aminoácidos , Animales , Axones/metabolismo , Técnicas de Cultivo de Célula , Biología Computacional , Secuencia Conservada , Humanos , Microtúbulos/metabolismo , Microtúbulos/fisiología , Neuronas , Imagen Óptica/métodos , Células PC12 , Fosforilación , Unión Proteica , Dominios Proteicos , Isoformas de Proteínas/metabolismo , Ratas , Proteínas tau/genética , Proteínas tau/fisiología
17.
Int Rev Cell Mol Biol ; 321: 89-169, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26811287

RESUMEN

Neurons are the basic information-processing units of the nervous system. In fulfilling their task, they establish a structural polarity with an axon that can be over a meter long and dendrites with a complex arbor, which can harbor ten-thousands of spines. Microtubules and their associated proteins play important roles during the development of neuronal morphology, the plasticity of neurons, and neurodegenerative processes. They are dynamic structures, which can quickly adapt to changes in the environment and establish a structural scaffold with high local variations in composition and stability. This review presents a comprehensive overview about the role of microtubules and their dynamic behavior during the formation and maturation of processes and spines in the healthy brain, during aging and under neurodegenerative conditions. The review ends with a discussion of microtubule-targeted therapies as a perspective for the supportive treatment of neurodegenerative disorders.


Asunto(s)
Microtúbulos/metabolismo , Neuronas/fisiología , Envejecimiento , Enfermedad de Alzheimer/metabolismo , Animales , Axones/metabolismo , Axones/fisiología , Encéfalo/embriología , Citoesqueleto/metabolismo , Dendritas/metabolismo , Dendritas/fisiología , Espinas Dendríticas/metabolismo , Humanos , Enfermedades Neurodegenerativas/metabolismo , Neurogénesis , Plasticidad Neuronal , Neuronas/metabolismo , Procesamiento Proteico-Postraduccional
18.
Neuropharmacology ; 105: 84-95, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26772969

RESUMEN

Dendritic spines represent the major postsynaptic input of excitatory synapses. Loss of spines and changes in their morphology correlate with cognitive impairment in Alzheimer's disease (AD) and are thought to occur early during pathology. Therapeutic intervention at a preclinical stage of AD to modify spine changes might thus be warranted. To follow the development and to potentially interfere with spine changes over time, we established a long term ex vivo model from organotypic cultures of the hippocampus from APP transgenic and control mice. The cultures exhibit spine loss in principal hippocampal neurons, which closely resembles the changes occurring in vivo, and spine morphology progressively changes from mushroom-shaped to stubby. We demonstrate that spine changes are completely reversed within few days after blocking amyloid-ß (Aß) production with the gamma-secretase inhibitor DAPT. We show that the microtubule disrupting drug nocodazole leads to spine loss similar to Aß expressing cultures and suppresses DAPT-mediated spine recovery in slices from APP transgenic mice. Finally, we report that epothilone D (EpoD) at a subnanomolar concentration, which slightly stabilizes microtubules in model neurons, completely reverses Aß-induced spine loss and increases thin spine density. Taken together the data indicate that Aß causes spine changes by microtubule destabilization and that spine recovery requires microtubule polymerization. Moreover, our results suggest that a low, subtoxic concentration of EpoD is sufficient to reduce spine loss during the preclinical stage of AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/metabolismo , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/patología , Epotilonas/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/patología , Moduladores de Tubulina/farmacología , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Células Cultivadas , Diaminas/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Nocodazol/farmacología , Células PC12 , Ratas , Tiazoles/farmacología
19.
Drugs ; 76(3): 301-13, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26729186

RESUMEN

Alzheimer's disease (AD) is characterised by a progressive loss of cognitive functions. Histopathologically, AD is defined by the presence of extracellular amyloid plaques containing Aß and intracellular neurofibrillary tangles composed of hyperphosphorylated tau proteins. According to the now well-accepted amyloid cascade hypothesis is the Aß pathology the primary driving force of AD pathogenesis, which then induces changes in tau protein leading to a neurodegenerative cascade during the progression of disease. Since many earlier drug trials aiming at preventing Aß pathology failed to demonstrate efficacy, tau and microtubules have come into focus as prominent downstream targets. The article aims to develop the current concept of the involvement of tau in the neurodegenerative triad of synaptic loss, cell death and dendritic simplification. The function of tau as a microtubule-associated protein and versatile interaction partner will then be introduced and the rationale and progress of current tau-directed therapy will be discussed in the biological context.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Terapia Molecular Dirigida , Agregación Patológica de Proteínas/prevención & control , Proteínas tau/metabolismo , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Epotilonas/administración & dosificación , Epotilonas/efectos adversos , Epotilonas/uso terapéutico , Humanos , Inmunización Pasiva , Azul de Metileno/administración & dosificación , Azul de Metileno/efectos adversos , Azul de Metileno/uso terapéutico , Oligopéptidos/administración & dosificación , Oligopéptidos/efectos adversos , Oligopéptidos/uso terapéutico , Proteínas tau/antagonistas & inhibidores , Proteínas tau/genética , Proteínas tau/inmunología
20.
Mol Neurodegener ; 10: 60, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26541821

RESUMEN

BACKGROUND: Dendritic simplification, a key feature of the neurodegenerative triad of Alzheimer's disease (AD) in addition to spine changes and neuron loss, occurs in a region-specific manner. However, it is unknown how changes in dendritic complexity are mediated and how they relate to spine changes and neuron loss. RESULTS: To investigate the mechanisms of dendritic simplification in an authentic CNS environment we employed an ex vivo model, based on targeted expression of enhanced green fluorescent protein (EGFP)-tagged constructs in organotypic hippocampal slices of mice. Algorithm-based 3D reconstruction of whole neuron morphology in different hippocampal regions was performed on slices from APPSDL-transgenic and control animals. We demonstrate that induction of dendritic simplification requires the combined action of amyloid beta (Aß) and human tau. Simplification is restricted to principal neurons of the CA1 region, recapitulating the region specificity in AD patients, and occurs at sites of Schaffer collateral input. We report that γ-secretase inhibition and treatment with the NMDA-receptor antagonist, CPP, counteract dendritic simplification. The microtubule-stabilizing drug epothilone D (EpoD) induces simplification in control cultures per se. Similar morphological changes were induced by a phosphoblocking tau construct, which also increases microtubule stability. In fact, low nanomolar concentrations of naturally secreted Aß decreased phosphorylation at S262 in a cellular model, a site which is known to directly modulate tau-microtubule interactions. CONCLUSIONS: The data provide evidence that dendritic simplification is mechanistically distinct from other neurodegenerative events and involves microtubule stabilization by dendritic tau, which becomes dephosphorylated at certain sites. They imply that treatments leading to an overall decrease of tau phosphorylation might have a negative impact on neuronal connectivity.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Dendritas/patología , Espinas Dendríticas/patología , Hipocampo/metabolismo , Microtúbulos/metabolismo , Neuronas/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/patología , Animales , Dendritas/metabolismo , Espinas Dendríticas/metabolismo , Femenino , Hipocampo/patología , Masculino , Ratones , Microtúbulos/patología , Degeneración Nerviosa/patología , Neuronas/patología , Sinapsis/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...