Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Mol Neurosci ; 73(9-10): 693-712, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37606769

RESUMO

The stereotypical progression of Tau pathology during Alzheimer disease has been attributed to trans-neuronal spreading of misfolded Tau proteins, followed by prion-like templated aggregation of Tau. The nature of Tau and the cellular mechanisms of Tau spreading are still under debate. We hypothesized that Tau's propensity for aggregation would correlate with its ability to spread across synapses and propagate pathology. To study the progressive propagation of Tau proteins in brain regions relevant for Alzheimer disease, we used mice expressing near-physiological levels of full-length human Tau protein carrying pro-aggregant (TauΔK280, TauΔK) or anti-aggregant (TauΔK280-PP, TauΔK-PP) mutations in the entorhinal cortex (EC). To enhance Tau expression in the EC, we performed EC injections of adeno-associated virus (AAV) particles encoding TauΔK or TauΔK-PP. The brains of injected and non-injected EC/TauΔK and EC/TauΔK-PP mice were studied by immunohistological and biochemical techniques to detect Tau propagation to dentate gyrus (DG) neurons and Tau-induced pathological changes. Pro- and anti-aggregant mice had comparable low transgene expression (~0.2 times endogenous mouse Tau). They accumulated human Tau at similar rates and only in expressing EC neurons, including their axonal projections of the perforant path and presynaptic terminals in the molecular layer of the DG. Pro-aggregant EC/TauΔK mice showed misfolded Tau and synaptic protein alterations in EC neurons, not observed in anti-aggregant EC/TauΔK-PP mice. Additional AAV-mediated expression of TauΔK or TauΔK-PP in EC/TauΔK or EC/TauΔK-PP mice, respectively, increased the human Tau expression to ~0.65 times endogenous mouse Tau, with comparable spreading of TauΔK and TauΔK-PP throughout the EC. There was a low level of transcellular propagation of Tau protein, without pathological phosphorylation or misfolding, as judged by diagnostic antibodies. Additionally, TauΔK but not TauΔK-PP expression induced hippocampal astrogliosis. Low levels of pro- or anti-aggregant full-length Tau show equivalent distributions in EC neurons, independent of their aggregation propensity. Increasing the expression via AAV induce local Tau misfolding in the EC neurons, synaptotoxicity, and astrogliosis and lead to a low level of detectable trans-neuronal spreading of Tau. This depends on its concentration in the EC, but, contrary to expectations, does not depend on Tau's aggregation propensity/misfolding and does not lead to templated misfolding in recipient neurons.


Assuntos
Doença de Alzheimer , Tauopatias , Camundongos , Animais , Humanos , Proteínas tau/genética , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Tauopatias/metabolismo , Gliose , Hipocampo/metabolismo , Modelos Animais de Doenças , Camundongos Transgênicos
2.
Int J Mol Sci ; 24(11)2023 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-37298211

RESUMO

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


Assuntos
Receptor A1 de Adenosina , Tauopatias , Camundongos , Animais , Camundongos Transgênicos , Receptor A1 de Adenosina/genética , Receptor A1 de Adenosina/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Hipocampo/metabolismo , Tauopatias/tratamento farmacológico , Tauopatias/genética , Tauopatias/metabolismo , Cognição , Modelos Animais de Doenças
3.
J Biol Chem ; 298(4): 101774, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35218773

RESUMO

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


Assuntos
Lectinas Tipo C , Proteínas de Membrana , Proteômica , Proteínas tau , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Expressão Gênica , Humanos , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Ligação Proteica , Proteínas tau/genética , Proteínas tau/metabolismo
4.
Mol Neurobiol ; 56(5): 3751-3767, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30196394

RESUMO

Tau aggregation is a hallmark of a group of neurodegenerative diseases termed Tauopathies. Reduction of aggregation-prone Tau has emerged as a promising therapeutic approach. Here, we show that an anti-aggregant Tau fragment (F3ΔKPP, residues 258-360) harboring the ΔK280 mutation and two proline substitutions (I277P & I308P) in the repeat domain can inhibit aggregation of Tau constructs in vitro, in cultured cells and in vivo in a Caenorhabditis elegans model of Tau aggregation. The Tau fragment reduced Tau-dependent cytotoxicity in a N2a cell model, suppressed the Tau-mediated neuronal dysfunction and ameliorated the defective locomotion in C. elegans. In vitro the fragment competes with full-length Tau for polyanionic aggregation inducers and thus inhibits Tau aggregation. Our combined in vitro and in vivo results suggest that the anti-aggregant Tau fragment may potentially be used to address the consequences of Tau aggregation in Tauopathies.


Assuntos
Fragmentos de Peptídeos/farmacologia , Agregados Proteicos/efeitos dos fármacos , Proteínas tau/toxicidade , Animais , Caenorhabditis elegans/efeitos dos fármacos , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Camundongos , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Mitocôndrias/metabolismo , Modelos Biológicos , Estrutura Secundária de Proteína , Proteínas tau/química
5.
Autophagy ; 14(12): 2139-2154, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30145931

RESUMO

Missorting of MAPT/Tau represents one of the early signs of neurodegeneration in Alzheimer disease. The triggers for this are still a matter of debate. Here we investigated the sorting mechanisms of endogenous MAPT in mature primary neurons using microfluidic chambers (MFCs) where cell compartments can be observed separately. Blocking protein degradation pathways with proteasomal or autophagy inhibitors dramatically increased the missorting of MAPT in dendrites on the neuritic side, suggesting that degradation of MAPT in dendrites is a major determinant for the physiological axonal distribution of MAPT. Such missorted dendritic MAPT differed in its phosphorylation pattern from axonal MAPT. By contrast, enhancing autophagy or proteasomal pathways strongly reduced MAPT missorting, thereby confirming the role of protein degradation pathways in the polar distribution of MAPT. Dendritic missorting of MAPT by blocking protein degradation resulted in the loss of spines but not in overall cell toxicity. Inhibition of local protein synthesis in dendrites eliminated the missorting of MAPT, indicating that the accumulation of dendritic MAPT is locally generated. In support of this, a substantial fraction of Mapt/Tau mRNA was detected in dendrites. Taken together, our results indicate that the autophagy and proteasomal pathways play important roles in fine-tuning dendritic MAPT levels and thereby prevent synaptic toxicity caused by MAPT accumulation. Abbreviations Ani: anisomycin; Baf: bafilomycin A1; BSA: bovine serum albumin; cAMP: cyclic adenosine monophosphate; CHX: cycloheximide; DMSO: dimethyl sulfoxide; DIV: days in vitro; Epo: epoxomicin; E18: embryonic day 18; FISH: fluorescence in situ hybridization; IgG: immunoglobulin; kDa: kilodalton; Lac: lactacystin; LDH: lactate dehydrogenase; MFC: microfluidic chambers; MAPs: microtubule-associated proteins; MAPT/Tau: microtubule-associated protein tau; PVDF: polyvinylidene difluoride; PBS: phosphate-buffered saline; PRKA: protein kinase AMP-activated; RD150: round device 150; RT: room temperature; SDS: sodium dodecyl sulfate; SEM: standard error of the mean; Wor: wortmannin.


Assuntos
Neurônios/metabolismo , Proteólise , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Autofagia/genética , Células Cultivadas , Embrião de Mamíferos , Camundongos , Camundongos Knockout , Neuritos/metabolismo , Neuritos/patologia , Neurônios/patologia , Transporte Proteico , Ratos , Ratos Sprague-Dawley , Sinapses/metabolismo , Sinapses/patologia , Proteínas tau/genética
6.
Neurobiol Aging ; 57: 47-63, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28600952

RESUMO

The aggregation of Tau protein is a hallmark of neurodegenerative diseases including Alzheimer's disease. Previously, we generated a cell model of tauopathy based on the 4-repeat domain with the FTDP-17 mutation ΔK280 (Tau4RDΔK) which is expressed in a regulatable fashion (tet-on). The deletion variant ΔK280 is highly amyloidogenic and forms fibrous aggregates in neuroblastoma N2a cells staining with the reporter dye Thioflavin S. The aggregation of Tau4RDΔK is toxic, contrary to wildtype or anti-aggregant variants of the protein. Using a novel approach for monitoring in situ Tau aggregation and toxicity by combination of microscopic analysis with FACS and biochemical analysis of cells enabled the dissection of the aggregating species which cause a time-dependent increase of toxicity. The dominant initiating step is the dimerization of Tau4RDΔK which leads to further aggregation and induces a strong increase in reactive oxygen species (ROS) and cytoplasmic Ca2+ which damage the membranes and cause cell death. Tau-based treatments using Tau aggregation inhibitors reduce both soluble oligomeric and fully aggregated Tau species and decrease their toxicity.


Assuntos
Hidrazinas/farmacologia , Hidrazinas/uso terapêutico , Tauopatias/prevenção & controle , Tiazóis/farmacologia , Tiazóis/uso terapêutico , Proteínas tau/metabolismo , Proteínas tau/toxicidade , Cálcio/metabolismo , Morte Celular , Células Cultivadas , Citoplasma/metabolismo , Dimerização , Relação Dose-Resposta a Droga , Mutação , Agregados Proteicos , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo
7.
Alzheimers Dement ; 13(11): 1270-1291, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28528849

RESUMO

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


Assuntos
Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Fragmentos de Peptídeos/toxicidade , Sinapses/efeitos dos fármacos , Proteínas tau/química , Animais , Cálcio/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Dicroísmo Circular , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/ultraestrutura , Líquido Extracelular/efeitos dos fármacos , Hipocampo/citologia , Humanos , Imunoprecipitação , Técnicas In Vitro , Microscopia de Força Atômica , Microscopia Eletrônica de Transmissão , Neuroblastoma , Técnicas de Cultura de Órgãos , Células PC12 , Ratos , Espécies Reativas de Oxigênio/metabolismo , Espectrofotometria , Sinapses/metabolismo , Proteínas tau/toxicidade
8.
Proc Natl Acad Sci U S A ; 113(41): 11597-11602, 2016 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-27671637

RESUMO

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


Assuntos
Antagonistas do Receptor A1 de Adenosina/farmacologia , Axônios/metabolismo , Deleção de Sequência , Xantinas/farmacologia , Proteínas tau/genética , Trifosfato de Adenosina/metabolismo , Animais , Espinhas Dendríticas/efeitos dos fármacos , Modelos Animais de Doenças , Imunofluorescência , Expressão Gênica , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Memória de Longo Prazo/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Fosforilação , Agregados Proteicos , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Tauopatias/tratamento farmacológico , Tauopatias/genética , Tauopatias/metabolismo , Tauopatias/patologia
9.
Brain Struct Funct ; 220(5): 3067-73, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24889163

RESUMO

Focal epilepsies often originate in the hippocampal formation of the temporal lobe (temporal lobe epilepsy) and are generally acquired after transient brain insults. Such insults induce cellular and structural reorganization processes of the hippocampus, referred to as epileptogenesis that finally convert the brain spontaneous epileptic. Here, we developed a new molecular imaging strategy in a state-of-the-art animal model to provide insights into key epileptogenic mechanisms. Our new approach combines recombinant adeno-associated virus (rAAV) gene delivery with in vivo bioluminescence imaging. rAAV particles harboring the luciferase reporter gene under control of the minimal T type Ca(2+)-channel subunit Ca V 3.2-promoter were generated and injected stereotaxically in the hippocampal region of mice. Bioluminescent signals, corresponding to Ca V 3.2 promoter activation, were imaged in vivo in the pilocarpine model of status epilepticus (SE). We detected activation of key Ca V 3.2 promoter motifs at 3 and 10 days after SE but not after the onset of chronic seizures. These data suggest Ca V 3.2 promoter activation as novel anti-epileptogenic target. In more general terms, we have established an experimental approach that allows to follow cerebral gene promoter dynamics longitudinally and to correlate this activity to behavioral parameters in the same mice.


Assuntos
Canais de Cálcio/metabolismo , Epilepsia do Lobo Temporal/metabolismo , Hipocampo/metabolismo , Imagem Molecular , Pilocarpina/farmacologia , Animais , Modelos Animais de Doenças , Epilepsia do Lobo Temporal/patologia , Camundongos , Convulsões/fisiopatologia , Estado Epiléptico/metabolismo , Lobo Temporal/metabolismo , Lobo Temporal/patologia
10.
J Alzheimers Dis ; 44(3): 937-47, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25374103

RESUMO

One of the hallmarks of Alzheimer's disease is the formation of neurofibrillary tangles, intracellular aggregates of hyperphosphorylated, mislocalized tau protein, which are associated with neuronal loss. Changes in tau are known to impair cellular transport (including that of mitochondria) and are associated with cell death in cell culture and mouse models of tauopathy. Thus clearing pathological forms of tau from cells is a key therapeutic strategy. One critical modulator in the degradation and clearance of misfolded proteins is the co-chaperone CHIP (Carboxy terminus Hsp70 interacting Protein), which is known to play a role in refolding and clearance of hyperphosphorylated tau. Here, we tested the hypothesis that CHIP could ameliorate pathological changes associated with tau. We find that co-expressing CHIP with full-length tau, tau truncated at D421 mimicking caspase cleavage, or the short tauRDΔK280 tau construct containing only the tau repeat domain with a tauopathy mutation, decreases tau protein levels in human H4 neuroglioma cells in a manner dependent on the Hsp70-binding TPR domain of CHIP. The observed reduction in tau levels by CHIP is associated with a decrease of tau phosphorylation and reduced levels of cleaved Caspase 3 indicating that CHIP plays an important role in preventing tau-induced pathological changes. Furthermore, tau-associated mitochondrial transport deficits are rescued by CHIP co-expression in H4 cells. Together, these data suggest that the co-chaperone CHIP can rescue the pathological effects of tau, and indicate that other diseases of protein misfolding and accumulation may also benefit from CHIP upregulation.


Assuntos
Regulação da Expressão Gênica/genética , Proteínas de Choque Térmico HSP70/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas tau/metabolismo , Caspase 3/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Choque Térmico HSP70/genética , Humanos , Mitocôndrias/metabolismo , Mutação/genética , Neuroblastoma/patologia , Ligação Proteica/efeitos dos fármacos , Transfecção , Ubiquitina-Proteína Ligases/genética , Proteínas tau/genética
11.
J Biol Chem ; 289(2): 956-67, 2014 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-24235150

RESUMO

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


Assuntos
Endocitose , Emaranhados Neurofibrilares/metabolismo , Neurônios/metabolismo , Proteínas tau/metabolismo , Animais , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Endossomos/metabolismo , Exocitose , Espaço Extracelular/metabolismo , Humanos , Lisossomos/metabolismo , Microscopia Confocal , Microscopia Eletrônica , Modelos Biológicos , Emaranhados Neurofibrilares/ultraestrutura , Neurônios/patologia , Tauopatias/metabolismo , Vesículas Transportadoras/metabolismo
12.
EMBO J ; 32(22): 2920-37, 2013 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-24065130

RESUMO

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


Assuntos
Adenosina Trifosfatases/fisiologia , Peptídeos beta-Amiloides/fisiologia , Microtúbulos/fisiologia , Peptídeo Sintases/fisiologia , Sinapses/patologia , Proteínas tau/fisiologia , Peptídeos beta-Amiloides/química , Animais , Células Cultivadas , Ácido Glutâmico/metabolismo , Camundongos , Camundongos Knockout , Ratos , Espastina , Proteínas tau/genética
13.
Mol Neurobiol ; 47(3): 868-82, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23192390

RESUMO

Bioluminescence imaging (BLI) takes advantage of the light-emitting properties of luciferase enzymes, which produce light upon oxidizing a substrate (i.e., D-luciferin) in the presence of molecular oxygen and energy. Photons emitted from living tissues can be detected and quantified by a highly sensitive charge-coupled device camera, enabling the investigator to noninvasively analyze the dynamics of biomolecular reactions in a variety of living model organisms such as transgenic mice. BLI has been used extensively in cancer research, cell transplantation, and for monitoring of infectious diseases, but only recently experimental models have been designed to study processes and pathways in neurological disorders such as Alzheimer disease, Parkinson disease, or amyotrophic lateral sclerosis. In this review, we highlight recent applications of BLI in neuroscience, including transgene expression in the brain, longitudinal studies of neuroinflammatory responses to neurodegeneration and injury, and in vivo imaging studies of neurogenesis and mitochondrial toxicity. Finally, we highlight some new developments of BLI compounds and luciferase substrates with promising potential for in vivo studies of neurological dysfunctions.


Assuntos
Encéfalo/metabolismo , Encéfalo/patologia , Medições Luminescentes/métodos , Degeneração Neural/diagnóstico , Degeneração Neural/patologia , Animais , Modelos Animais de Doenças , Neurogênese , Transgenes
14.
Methods Mol Biol ; 849: 85-99, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22528085

RESUMO

The microtubule-associated protein Tau plays a physiological role of stabilizing neuronal microtubules by binding to their lateral surface. Tau belongs to the category of natively unfolded protein as it shows typical features of random coil, as analyzed by various biophysical techniques. In cells, it is subjected to several posttranslational modifications (e.g., phosphorylation, cleavage, ubiquitination, and glycosylation). In neurodegenerative diseases, Tau forms insoluble aggregates called paired helical filaments (PHFs). We have applied fluorescence resonance energy transfer (FRET) to examine the conformations of soluble Tau. We created a series of Tau mutants, each carrying one tryptophan and one cysteine (labeled by IEADANS). This made it possible to measure the distance between these FRET pairs placed in different domains of Tau. This approach enables one to analyze the global folding of soluble Tau and its alteration upon phosphorylation and denaturation.


Assuntos
Transferência Ressonante de Energia de Fluorescência/métodos , Proteínas tau/química , Guanidina/farmacologia , Fosforilação , Conformação Proteica/efeitos dos fármacos , Desdobramento de Proteína/efeitos dos fármacos
15.
J Biol Chem ; 287(11): 8174-86, 2012 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-22238344

RESUMO

The kinase MARK2/Par-1 plays key roles in several cell processes, including neurodegeneration such as Alzheimer disease by phosphorylating tau and detaching it from microtubules. In search of interaction partners of MARK2, we identified phosphatase and tensin homolog (PTEN)-induced kinase 1 (PINK1), which is important for the survival of neurons and whose mutations are linked to familial Parkinson disease (PD). MARK2 phosphorylated and activated the cleaved form of PINK1 (ΔN-PINK1; amino acids 156-581). Thr-313 was the primary phosphorylation site, a residue mutated to a non-phosphorylatable form (T313M) in a frequent variant of PD. Mutation of Thr-313 to Met or Glu in PINK1 showed toxic effects with abnormal mitochondrial distribution in neurons. MARK2 and PINK1 were found to colocalize with mitochondria and regulate their transport. ΔN-PINK1 promoted anterograde transport and increased the fraction of stationary mitochondria, whereas full-length PINK1 promoted retrograde transport. In both cases, MARK2 enhanced the effects. The results identify MARK2 as an upstream regulator of PINK1 and ΔN-PINK1 and provide insights into the regulation of mitochondrial trafficking in neurons and neurodegeneration in PD.


Assuntos
Mitocôndrias/enzimologia , Mutação de Sentido Incorreto , Neurônios/enzimologia , Doença de Parkinson/enzimologia , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Substituição de Aminoácidos , Animais , Transporte Biológico Ativo/genética , Células CHO , Sobrevivência Celular , Cricetinae , Cricetulus , Células HEK293 , Humanos , Mitocôndrias/genética , Mitocôndrias/patologia , Neurônios/patologia , Células PC12 , Doença de Parkinson/genética , Doença de Parkinson/patologia , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases/genética , Ratos , Spodoptera , Treonina
16.
J Biol Chem ; 286(48): 41711-41722, 2011 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-21984823

RESUMO

Protein kinases of the microtubule affinity regulating kinase (MARK)/Par-1 family play important roles in the establishment of cellular polarity, cell cycle control, and intracellular signal transduction. Disturbance of their function is linked to cancer and brain diseases, e.g. lissencephaly and Alzheimer disease. To understand the biological role of MARK family kinases, we searched for specific inhibitors and a biosensor for MARK activity. A screen of the ChemBioNet library containing ~18,000 substances yielded several compounds with inhibitory activity in the low micromolar range and capable of inhibiting MARK activity in cultured cells and primary neurons, as judged by MARK-dependent phosphorylation of microtubule-associated proteins and its consequences for microtubule integrity. Four of the compounds share a 9-oxo-9H-acridin-10-yl structure as a basis that will serve as a lead for optimization of inhibition efficiency. To test these inhibitors, we developed a cellular biosensor for MARK activity based on a MARK target sequence attached to the 14-3-3 scaffold protein and linked to enhanced cyan or teal and yellow fluorescent protein as FRET donor and acceptor pairs. Transfection of the teal/yellow fluorescent protein sensor into neurons and imaging by fluorescence lifetime imaging revealed that MARK was particularly active in the axons and growth cones of differentiating neurons.


Assuntos
Proteínas 14-3-3/metabolismo , Técnicas Biossensoriais/métodos , Transferência Ressonante de Energia de Fluorescência/métodos , Cones de Crescimento/metabolismo , Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas 14-3-3/genética , Animais , Células CHO , Diferenciação Celular/fisiologia , Cricetinae , Cricetulus , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Microtúbulos/genética , Células PC12 , Proteínas Serina-Treonina Quinases/genética , Ratos
17.
Neurobiol Aging ; 32(1): 1-14, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20961659

RESUMO

The amyloid cascade hypothesis of Alzheimer's disease (AD) posits that the generation of ß-amyloid (Aß) triggers Tau neurofibrillary pathology. Recently a "17 kD" calpain-induced Tau fragment, comprising residues 45-230 (molecular weight [MW], 18.7 kD), was proposed to mediate Aß-induced toxicity. Here, we demonstrate that the "17 kD" fragment is actually much smaller, containing residues 125-230 (molecular weight, 10.7 kD). Inducing Tau phosphorylation by okadaic acid or mimicking phosphorylation by Glu mutations at the epitopes of Alzheimer-diagnostic antibodies AT100/AT8/PHF1 could not prevent the generation of this fragment. The fragment can be induced not only by Aß oligomers, but also by other cell stressors, e.g., thapsigargin (a Ca(2+)-ATPase inhibitor) or glutamate (an excitatory neurotransmitter). However, overexpression of neither Tau(45-230) nor Tau(125-230) fragment is toxic to Chinese hamster ovary (CHO) cells, neuroblastoma cells (N2a) or primary hippocampal neurons. Finally, the calpain-induced fragment can be observed both in Alzheimer's disease brains and in control normal human brains. We conclude that the 17 kD Tau fragment is not a mediator of Aß-induced toxicity, leaving open the possibility that upstream calpain activation might cause both Tau fragmentation and toxicity.


Assuntos
Doença de Alzheimer/metabolismo , Calpaína/farmacologia , Córtex Cerebral/metabolismo , Proteínas tau/efeitos dos fármacos , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/toxicidade , Animais , Proteínas de Ligação a Calmodulina/farmacologia , Contagem de Células , Córtex Cerebral/citologia , Cricetinae , Cricetulus , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Ácido Glutâmico/farmacologia , Proteínas de Fluorescência Verde/genética , Humanos , Marcação In Situ das Extremidades Cortadas/métodos , Peso Molecular , Mutação/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fragmentos de Peptídeos/toxicidade , Peptídeos/metabolismo , Peptídeos/farmacologia , Fosforilação/efeitos dos fármacos , Ratos , Tapsigargina/farmacologia , Transfecção/métodos , Proteínas tau/genética
19.
J Neurosci ; 30(36): 11938-50, 2010 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-20826658

RESUMO

Aggregation of amyloid-beta (Abeta) and Tau protein are hallmarks of Alzheimer's disease (AD), and according to the Abeta-cascade hypothesis, Abeta is considered toxic for neurons and Tau a downstream target of Abeta. We have investigated differentiated primary hippocampal neurons for early localized changes following exposure to Abeta oligomers. Initial events become evident by missorting of endogenous Tau into the somatodendritic compartment, in contrast to axonal sorting in normal neurons. In missorted dendritic regions there is a depletion of spines and local increase in Ca(2+), and breakdown of microtubules. Tau in these regions shows elevated phosphorylation at certain sites diagnostic of AD-Tau (e.g., epitope of antibody 12E8, whose phosphorylation causes detachment of Tau from microtubules, and AT8 epitope), and local elevation of certain kinase activities (e.g., MARK/par-1, BRSK/SADK, p70S6K, cdk5, but not GSK3beta, JNK, MAPK). These local effects occur without global changes in Tau, tubulin, or kinase levels. Somatodendritic missorting occurs not only with Tau, but also with other axonal proteins such as neurofilaments, and correlates with pronounced depletion of microtubules and mitochondria. The Abeta-induced effects on microtubule and mitochondria depletion, Tau missorting, and loss of spines are prevented by taxol, indicating that Abeta-induced microtubule destabilization and corresponding traffic defects are key factors in incipient degeneration. By contrast, the rise in Ca(2+) levels, kinase activities, and Tau phosphorylation cannot be prevented by taxol. Incipient and local changes similar to those of Abeta oligomers can be evoked by cell stressors (e.g., H(2)O(2), glutamate, serum deprivation), suggesting some common mechanism of signaling.


Assuntos
Peptídeos beta-Amiloides/química , Cálcio/metabolismo , Microtúbulos/metabolismo , Neurônios/ultraestrutura , Fragmentos de Peptídeos/farmacologia , Proteínas tau/metabolismo , Trifosfato de Adenosina/metabolismo , Peptídeos beta-Amiloides/farmacologia , Análise de Variância , Animais , Células Cultivadas , Dendritos , Espinhas Dendríticas , Embrião de Mamíferos , Hipocampo/citologia , L-Lactato Desidrogenase/metabolismo , Microscopia Eletrônica de Transmissão/métodos , Microtúbulos/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Proteínas de Neurofilamentos/metabolismo , Neurônios/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteínas Quinases/metabolismo , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
20.
Neuropharmacology ; 59(4-5): 276-89, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20149808

RESUMO

Alzheimer disease is characterized by pathological aggregation of two proteins, tau and Abeta-amyloid, both of which are considered to be toxic to neurons. In this review we summarize recent advances on small molecule inhibitors of protein aggregation with emphasis on tau, with activities mediated by the direct interference of self-assembly. The inhibitors can be clustered in several compound classes according to their chemical structure, with subsequent description of the structure-activity relationships, showing that hydrophobic interactions are prevailing. The description is extended to the pharmacological profile of the compounds in order to evaluate their drug-likeness, with special attention to toxicity and bioavailability. The collected data indicate that following the improvements of the in vitro inhibitory potencies, the consideration of the in vivo pharmacokinetics is an absolute prerequisite for the development of compounds suitable for a transfer from bench to bedside.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Proteínas tau/antagonistas & inibidores , Proteínas tau/metabolismo , Aldeído Redutase/antagonistas & inibidores , Aldeído Redutase/metabolismo , Doença de Alzheimer/metabolismo , Animais , Flavonoides/farmacocinética , Flavonoides/farmacologia , Flavonoides/uso terapêutico , Humanos , Interações Hidrofóbicas e Hidrofílicas/efeitos dos fármacos , Fenóis/farmacocinética , Fenóis/farmacologia , Fenóis/uso terapêutico , Polifenóis , Rodanina/análogos & derivados , Rodanina/farmacocinética , Rodanina/farmacologia , Rodanina/uso terapêutico , Relação Estrutura-Atividade , Tiazolidinas/farmacocinética , Tiazolidinas/farmacologia , Tiazolidinas/uso terapêutico , Proteínas tau/efeitos adversos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA