Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 136
Filtrar
1.
Hum Mol Genet ; 30(21): 1955-1967, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34137825

RESUMEN

Accumulation of microtubule-associated tau protein is thought to cause neuron loss in a group of neurodegenerative diseases called tauopathies. In diseased brains, tau molecules adopt pathological structures that propagate into insoluble forms with disease-specific patterns. Several types of posttranslational modifications in tau are known to modulate its aggregation propensity in vitro, but their influence on tau accumulation and toxicity at the whole-organism level has not been fully elucidated. Herein, we utilized a series of transgenic Drosophila models to compare systematically the toxicity induced by five tau constructs with mutations or deletions associated with aggregation, including substitutions at seven disease-associated phosphorylation sites (S7A and S7E), deletions of PHF6 and PHF6* sequences (ΔPHF6 and ΔPHF6*), and substitutions of cysteine residues in the microtubule binding repeats (C291/322A). We found that substitutions and deletions resulted in different patterns of neurodegeneration and accumulation, with C291/322A having a dramatic effect on both tau accumulation and neurodegeneration. These cysteines formed disulfide bonds in mouse primary cultured neurons and in the fly retina, and stabilized tau proteins. Additionally, they contributed to tau accumulation under oxidative stress. We also found that each of these cysteine residues contributes to the microtubule polymerization rate and microtubule levels at equilibrium, but none of them affected tau binding to polymerized microtubules. Since tau proteins expressed in the Drosophila retina are mostly present in the early stages of tau filaments self-assembly, our results suggest that disulfide bond formation by these cysteine residues could be attractive therapeutic targets.


Asunto(s)
Agregación Patológica de Proteínas/metabolismo , Tauopatías/metabolismo , Proteínas tau/metabolismo , Animales , Animales Modificados Genéticamente , Biomarcadores , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Drosophila , Microtúbulos/metabolismo , Neuronas/metabolismo , Estrés Oxidativo , Unión Proteica , Multimerización de Proteína , Tauopatías/etiología , Tauopatías/patología , Proteínas tau/genética
2.
Nutr Neurosci ; 26(8): 766-777, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35816407

RESUMEN

Neurodegenerative diseases involving pathological tau protein aggregation are collectively known as tauopathies and include Alzheimer's disease and Pick's disease. Recent studies show that the intake of tryptophan-tyrosine (Trp-Tyr)-related ß-lactopeptides, including ß-lactolin, attenuates cognitive decline in the elderly and prevents the amyloid pathology in mouse models of Alzheimer's disease. However, the effects of Trp-Tyr-related ß-lactopeptides on tau-related pathology have not been investigated. In the present study, we examined the effects of Trp-Tyr dipeptide intake on tauopathy in PS19 transgenic mice, a well-established tauopathy model. Intake of Trp-Tyr dipeptide improved the behavioral deficits observed in the open field test, prevented tau phosphorylation, and increased the dopamine turnover and synaptophysin expression in the frontal cortex. Levels of short-chain fatty acids in the cecum were lower in PS19 mice than those in wild-type mice and were increased by treatment with Trp-Tyr dipeptide. In addition, intake of Trp-Tyr dipeptide extended the lifespan of PS19 mice. These findings suggest that the intake of Trp-Tyr-related peptides improves tauopathy symptoms, resulting in improvements in behavioral deficits and longevity. Hence, the intake of Trp-Tyr-related peptides, including ß-lactolin, may be beneficial for preventing dementia.


Asunto(s)
Enfermedad de Alzheimer , Tauopatías , Ratones , Animales , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Triptófano/uso terapéutico , Dipéptidos/uso terapéutico , Tirosina , Tauopatías/tratamiento farmacológico , Tauopatías/prevención & control , Tauopatías/metabolismo , Ratones Transgénicos , Proteínas tau/metabolismo , Modelos Animales de Enfermedad
3.
J Neurosci ; 39(34): 6781-6797, 2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31235644

RESUMEN

Tau is a microtubule (MT)-associated protein that is localized to the axon. In Alzheimer's disease, the distribution of tau undergoes a remarkable alteration, leading to the formation of tau inclusions in the somatodendritic compartment. To investigate how this mislocalization occurs, we recently developed immunohistochemical tools that can separately detect endogenous mouse and exogenous human tau with high sensitivity, which allows us to visualize not only the pathological but also the pre-aggregated tau in mouse brain tissues of both sexes. Using these antibodies, we found that in tau-transgenic mouse brains, exogenous human tau was abundant in dendrites and somata even in the presymptomatic period, whereas the axonal localization of endogenous mouse tau was unaffected. In stark contrast, exogenous tau was properly localized to the axon in human tau knock-in mice. We tracked this difference to the temporal expression patterns of tau. Endogenous mouse tau and exogenous human tau in human tau knock-in mice exhibited high expression levels during the neonatal period and strong suppression into the adulthood. However, human tau in transgenic mice was expressed continuously and at high levels in adult animals. These results indicated the uncontrolled expression of exogenous tau beyond the developmental period as a cause of mislocalization in the transgenic mice. Superresolution microscopic and biochemical analyses also indicated that the interaction between MTs and exogenous tau was impaired only in the tau-transgenic mice, but not in knock-in mice. Thus, the ectopic expression of tau may be critical for its somatodendritic mislocalization, a key step of the tauopathy.SIGNIFICANCE STATEMENT Somatodendritic localization of tau may be an early step leading to the neuronal degeneration in tauopathies. However, the mechanisms of the normal axonal distribution of tau and the mislocalization of pathological tau remain obscure. Our immunohistochemical and biochemical analyses demonstrated that the endogenous mouse tau is transiently expressed in neonatal brains, that exogenous human tau expressed corresponding to such tau expression profile can distribute into the axon, and that the constitutive expression of tau into adulthood (e.g., human tau in transgenic mice) results in abnormal somatodendritic localization. Thus, the expression profile of tau is tightly associated with the localization of tau, and the ectopic expression of tau in matured neurons may be involved in the pathogenesis of tauopathy.


Asunto(s)
Química Encefálica/fisiología , Encéfalo/citología , Dendritas/fisiología , Expresión Génica Ectópica/genética , Proteínas tau/biosíntesis , Animales , Animales Recién Nacidos , Axones/metabolismo , Encéfalo/crecimiento & desarrollo , Femenino , Técnicas de Sustitución del Gen , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Cultivo Primario de Células , Tauopatías/metabolismo
4.
Bioorg Med Chem Lett ; 29(16): 2107-2111, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31311732

RESUMEN

(R,S)-Isoproterenol inhibits the formation of toxic granular tau oligomers associated with neuronal loss and development of cognitive disorders, and is an attractive drug candidate for Alzheimer's disease. To elucidate its behavior in the brain by positron emission tomography, we synthesize (R,S)-[11C]isoproterenol by reductive alkylation of (R,S)-norepinephrine with [2-11C]acetone, which was in turn synthesized in situ under improved conditions afforded a decay-corrected radiochemical yield of 54%. The reductive alkylation using NaBH(OAc)3 as reducing agent in the presence of benzoic acid in DMSO/DMF (60:40 v/v) at 100 °C for 10 min gave (R,S)-[11C]isoproterenol in an 87% radio-high performance liquid chromatography (HPLC) analytical yield. HPLC separation using a strong cation exchange column, followed by pharmaceutical formulation in the presence of d/l-tartaric acid, afforded (R,S)-[11C]isoproterenol with a total radioactivity of 2.0 ±â€¯0.2 GBq, a decay-corrected radiochemical yield of 19 ±â€¯2%, chemical and radiochemical purities of 71% and >99%, respectively, and a molar activity of 100 ±â€¯13 GBq/µmol (n = 3). The overall synthesis time from the end of the bombardment to pharmaceutical formulation was 48 min. A preliminary preclinical PET study in a rat demonstrated the potential of the radioligand for the evaluation of the penetration of (R,S)-isoproterenol in human brain.


Asunto(s)
Acetona/química , Isoproterenol/síntesis química , Norepinefrina/química , Radiofármacos/síntesis química , Acetona/síntesis química , Alquilación , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Radioisótopos de Carbono/química , Isoproterenol/farmacología , Masculino , Tomografía de Emisión de Positrones , Radiofármacos/farmacología , Ratas Wistar , Estereoisomerismo
5.
Adv Exp Med Biol ; 1184: 373-380, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32096050

RESUMEN

The accumulation of tau filaments in neurons is a pathological hallmark of various neurodegenerative diseases, including Alzheimer's disease. However, it is not the filamentous aggregates themselves, but non-filamentous tau species, tau oligomer, that is thought to be the culprit in tau-mediated neurodegeneration. The definition of and methodology for isolating tau oligomers vary among researchers. Here we describe how tau oligomers are identified, summarize the differences of tau oligomers among research groups, and discuss their hypothesized functions.


Asunto(s)
Enfermedades Neurodegenerativas/metabolismo , Proteínas tau/química , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Humanos , Enfermedades Neurodegenerativas/patología , Neuronas/metabolismo , Neuronas/patología , Proteínas tau/aislamiento & purificación
6.
J Biol Chem ; 292(9): 3720-3728, 2017 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-28087694

RESUMEN

Alongside the rapid growth in aging populations worldwide, prevention and therapy for age-related memory decline and dementia are in great demand to maintain a long, healthy life. Here we found that iso-α-acids, hop-derived bitter compounds in beer, enhance microglial phagocytosis and suppress inflammation via activation of the peroxisome proliferator-activated receptor γ. In normal mice, oral administration of iso-α-acids led to a significant increase both in CD11b and CD206 double-positive anti-inflammatory type microglia (p < 0.05) and in microglial phagocytosis in the brain. In Alzheimer's model 5xFAD mice, oral administration of iso-α-acids resulted in a 21% reduction in amyloid ß in the cerebral cortex as observed by immunohistochemical analysis, a significant reduction in inflammatory cytokines such as IL-1ß and chemokines including macrophage inflammatory protein-1α in the cerebral cortex (p < 0.05) and a significant improvement in a novel object recognition test (p < 0.05), as compared with control-fed 5xFAD mice. The differences in iso-α-acid-fed mice were due to the induction of microglia to an anti-inflammatory phenotype. The present study is the first to report that amyloid ß deposition and inflammation are suppressed in a mouse model of Alzheimer's disease by a single component, iso-α-acids, via the regulation of microglial activation. The suppression of neuroinflammation and improvement in cognitive function suggests that iso-α-acids contained in beer may be useful for the prevention of dementia.


Asunto(s)
Ácidos/química , Enfermedad de Alzheimer/metabolismo , Cerveza , Disfunción Cognitiva/prevención & control , Inflamación/prevención & control , Administración Oral , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Animales , Antiinflamatorios/química , Barrera Hematoencefálica , Antígeno CD11b/metabolismo , Separación Celular , Células Cultivadas , Quimiocina CCL3/metabolismo , Cromatografía Líquida de Alta Presión , Cromatografía Liquida , Modelos Animales de Enfermedad , Humanos , Inmunohistoquímica , Lectinas Tipo C/metabolismo , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Ratones , Ratones Transgénicos , Microglía/metabolismo , Fagocitosis , Fenotipo , Receptores de Superficie Celular/metabolismo , Espectrometría de Masas en Tándem
7.
Molecules ; 23(12)2018 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-30501069

RESUMEN

Due to the growth in aging populations, prevention for cognitive decline and dementia are in great demand. We previously demonstrated that the consumption of iso-α-acids (IAA), the hop-derived bitter compounds in beer, prevents inflammation and Alzheimer's disease pathology in model mice. However, the effects of iso-α-acids on inflammation induced by other agents aside from amyloid ß have not been investigated. In this study, we demonstrated that the consumption of iso-α-acids suppressed microglial inflammation in the frontal cortex of rTg4510 tauopathy mice. In addition, the levels of inflammatory cytokines and chemokines, including IL-1ß and MIP-1ß, in the frontal cortex of rTg4510 mice were greater than those of wild-type mice, and were reduced in rTg4510 mice fed with iso-α-acids. Flow cytometry analysis demonstrated that the expression of cells producing CD86, CD68, TSPO, MIP-1α, TNF-α, and IL-1ß in microglia was increased in rTg4510 mice compared with wild-type mice. Furthermore, the expression of CD86- and MIP-1α-producing cells was reduced in rTg4510 mice administered with iso-α-acids. Moreover, the consumption of iso-α-acids reduced the levels of phosphorylated tau in the frontal cortex. Collectively, these results suggest that the consumption of iso-α-acids prevents the inflammation induced in tauopathy mice. Thus, iso-α-acids may help in preventing inflammation-related brain disorders.


Asunto(s)
Ácidos/uso terapéutico , Cerveza/análisis , Inflamación/patología , Microglía/patología , Gusto , Tauopatías/patología , Ácidos/química , Ácidos/farmacología , Animales , Encéfalo/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/efectos de los fármacos , Fenotipo , Fosforilación/efectos de los fármacos
8.
J Biol Chem ; 291(43): 22714-22720, 2016 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-27601475

RESUMEN

Neurofibrillar tangles caused by intracellular hyperphosphorylated tau inclusion and extracellular amyloid ß peptide deposition are hallmarks of Alzheimer's disease. Tau contains one or two cysteine residues in three or four repeats of the microtubule binding region following alternative splicing of exon 10, and formation of intermolecular cysteine disulfide bonds accelerates tau aggregation. 8-Nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP) acts as a novel second messenger of nitric oxide (NO) by covalently binding cGMP to cysteine residues by electrophilic properties, a process termed protein S-guanylation. Here we studied S-guanylation of tau and its effects on tau aggregation. 8-Nitro-cGMP exposure induced S-guanylation of tau both in vitro and in tau-overexpressed HEK293T cells. S-guanylated tau inhibited heparin-induced tau aggregation in a thioflavin T assay. Atomic force microscopy observations indicated that S-guanylated tau could not form tau granules and fibrils. Further biochemical analyses showed that S-guanylated tau was inhibited at the step of tau oligomer formation. In P301L tau-expressing Neuro2A cells, 8-nitro-cGMP treatment significantly reduced the amount of sarcosyl-insoluble tau. NO-linked chemical modification on cysteine residues of tau could block tau aggregation, and therefore, increasing 8-nitro-cGMP levels in the brain could become a potential therapeutic strategy for Alzheimer's disease.


Asunto(s)
GMP Cíclico/análogos & derivados , Óxido Nítrico/metabolismo , Agregado de Proteínas , Procesamiento Proteico-Postraduccional , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/terapia , GMP Cíclico/química , GMP Cíclico/metabolismo , Células HEK293 , Humanos , Proteínas tau/química , Proteínas tau/genética
9.
Hum Mol Genet ; 24(17): 4879-900, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26056228

RESUMEN

Leucine-rich repeat kinase 2 (LRRK2) is the causative molecule of the autosomal dominant hereditary form of Parkinson's disease (PD), PARK8, which was originally defined in a study of a Japanese family (the Sagamihara family) harboring the I2020T mutation in the kinase domain. Although a number of reported studies have focused on cell death mediated by mutant LRRK2, details of the pathogenetic effect of LRRK2 still remain to be elucidated. In the present study, to elucidate the mechanism of neurodegeneration in PD caused by LRRK2, we generated induced pluripotent stem cells (iPSC) derived from fibroblasts of PD patients with I2020T LRRK2 in the Sagamihara family. We found that I2020T mutant LRRK2 iPSC-derived neurons released less dopamine than control-iPSC-derived neurons. Furthermore, we demonstrated that patient iPSC-derived neurons had a lower phospho-AKT level than control-iPSC-derived neurons, and that the former showed an increased incidence of apoptosis relative to the controls. Interestingly, patient iPSC-derived neurons exhibited activation of glycogen synthase kinase-3ß (GSK-3ß) and high Tau phosphorylation. In addition, the postmortem brain of the patient from whom the iPSC had been established exhibited deposition of neurofibrillary tangles as well as increased Tau phosphorylation in neurons. These results suggest that I2020T LRRK2-iPSC could be a promising new tool for reproducing the pathology of PD in the brain caused by the I2020T mutation, and applicable as a model in studies of targeted therapeutics.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Mutación , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Proteínas tau/metabolismo , Animales , Apoptosis/genética , Autofagia , Caspasa 3/metabolismo , Línea Celular , Dopamina/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Células Madre Pluripotentes Inducidas/citología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Ratones , Neuronas/citología , Estrés Oxidativo , Fosforilación
10.
J Biol Chem ; 290(49): 29438-48, 2015 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-26472923

RESUMEN

Protein interacting with C kinase 1 (PICK1) is a synaptic protein interacting with the AMPA receptor subunits GluA2/3. The interaction between GluA2 and PICK1 is required for the removal of GluA2 from the synaptic plasma membrane during long-term depression (LTD). It has been suggested that glycogen synthase kinase 3ß (GSK-3ß) is activated during LTD, but the relationships between GluA2, PICK1, and GSK-3ß are not well understood. In particular, the substrate(s) of GSK-3ß have not yet been determined. Here we showed that PICK1 is a substrate of GSK-3ß. We found that Ser(339), Ser(342), Ser(412), and Ser(416) of PICK1 were putative GSK-3ß-mediated phosphorylation sites. Among these sites, Ser(416) played a crucial role in regulating the interaction between GluA2 and PICK1. We showed that replacing Ser(416) with Ala disrupted the GluA2-PICK1 interaction, whereas substituting Ser(416) with Glu or Asp retained this interaction. However, deletion of Ser(416) did not affect the GluA2-PICK1 interaction, and substitution of Ser(416) with Ala did not alter the PICK1-PICK1 interaction. Using image analysis in COS-7 cells with AcGFP1-fused PICK1, we showed that substitution of Ser(416) with Ala increased the formation of AcGFP1-positive clusters, suggesting an increase in the association of PICK1 with the membrane. This may have resulted in the dissociation of the GluA2-PICK1 complexes. Our results indicated that GSK-3ß-mediated phosphorylation of PICK1 at Ser(416) was required for its association with the AMPA receptor subunit. Therefore, the GSK-3ß-mediated phosphorylation of PICK1 may be a regulating factor during LTD induction.


Asunto(s)
Proteínas Portadoras/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Proteínas Nucleares/metabolismo , Secuencia de Aminoácidos , Animales , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Proteínas del Citoesqueleto , Ácido Glutámico/química , Glucógeno Sintasa/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Inmunoprecipitación , Datos de Secuencia Molecular , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína , Ratas , Receptores AMPA/metabolismo , Homología de Secuencia de Aminoácido , Serina/química
11.
Am J Pathol ; 183(1): 211-25, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23680655

RESUMEN

Frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) is a neurodegenerative disorder caused by mutations in the tau gene. Many mutations identified in FTDP-17 have been shown to affect tau exon 10 splicing in vitro, which presumably causes pathologic imbalances in exon 10(-) [3-repeat (3R)] and exon 10(+) [4-repeat (4R)] tau expression and leads to intracellular inclusions of hyperphosphorylated tau in patient brains. However, no reports have investigated this theory using model mice with a tau intronic mutation. Herein, we generated new transgenic mice harboring the tau intron 10 +16C → T mutation. We prepared a transgene construct containing intronic sequences required for exon 10 splicing in the longest tau isoform cDNA. Although mice bearing the construct without the intronic mutation showed normal developmental changes of the tau isoform from 3R tau to equal amounts of 3R and 4R tau, mice with the mutation showed much higher levels of 4R tau at the adult stage. 4R tau was selectively recovered in insoluble brain fractions in their old age. Furthermore, these mice displayed abnormal tau phosphorylation, synapse loss and dysfunction, memory impairment, glial activation, tangle formation, and neuronal loss in an age-dependent manner. These findings provide the first evidence in a mouse model that a tau intronic mutation-induced imbalance of 3R and 4R tau could be a cause of tauopathy.


Asunto(s)
Exones , Demencia Frontotemporal/genética , Intrones , Mutación , Empalme del ARN , Tauopatías/genética , Proteínas tau/genética , Animales , Western Blotting , Demencia Frontotemporal/patología , Demencia Frontotemporal/fisiopatología , Marcadores Genéticos , Masculino , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tauopatías/patología , Tauopatías/fisiopatología
12.
Acta Neuropathol ; 127(5): 685-98, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24531886

RESUMEN

Senile plaques comprised of Aß aggregates and neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau filaments are the hallmarks of Alzheimer's disease (AD). A number of amyloid precursor protein (APP) transgenic (Tg) mice harboring APP mutations have been generated as animal models of AD. These mice successfully display amyloid plaque formation and subsequent tau hyperphosphorylation, but seldom induce NFT formations. We have demonstrated that the APPOSK-Tg mice, which possess the E693Δ (Osaka) mutation in APP and thereby accumulate Aß oligomers without plaques, exhibit tau hyperphosphorylation at 8 months, but not NFT formation even at 24 months. We assumed that APP-Tg mice, including ours, failed to form NFTs because NFT formation requires human tau. To test this hypothesis, we crossbred APPOSK-Tg mice with tau-Tg mice (tau264), which express low levels of 3-repeat and 4-repeat wild-type human tau without any pathology. The resultant double Tg mice displayed tau hyperphosphorylation at 6 months and NFT formation at 18 months in the absence of tau mutations. Importantly, these NFTs contained both 3-repeat and 4-repeat human tau, similar to those in AD. Furthermore, the double Tg mice exhibited Aß oligomer accumulation, synapse loss, and memory impairment at 6 months and neuronal loss at 18 months, all of which appeared earlier than in the parent APPOSK-Tg mice. These results suggest that Aß and human tau synergistically interact to accelerate each other's pathology, that the presence of human tau is critical for NFT formation, and that Aß oligomers can induce NFTs in the absence of amyloid plaques.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Ovillos Neurofibrilares/metabolismo , Proteínas tau/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/patología , Progresión de la Enfermedad , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/patología , Ratones Transgénicos , Mutación , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Ovillos Neurofibrilares/patología , Neuronas/metabolismo , Neuronas/patología , Fosforilación , Sinapsis/metabolismo , Sinapsis/patología , Tauopatías/metabolismo , Tauopatías/patología , Factores de Tiempo , Proteínas tau/genética
13.
Structure ; 32(10): 1793-1807.e6, 2024 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-39032487

RESUMEN

Intracellular tau aggregation requires a local protein concentration increase, referred to as "droplets". However, the cellular mechanism for droplet formation is poorly understood. Here, we expressed OptoTau, a P301L mutant tau fused with CRY2olig, a light-sensitive protein that can form homo-oligomers. Under blue light exposure, OptoTau increased tau phosphorylation and was sequestered in aggresomes. Suppressing aggresome formation by nocodazole formed tau granular clusters in the cytoplasm. The granular clusters disappeared by discontinuing blue light exposure or 1,6-hexanediol treatment suggesting that intracellular tau droplet formation requires microtubule collapse. Expressing OptoTau-ΔN, a species of N-terminal cleaved tau observed in the Alzheimer's disease brain, formed 1,6-hexanediol and detergent-resistant tau clusters in the cytoplasm with blue light stimulation. These intracellular stable tau clusters acted as a seed for tau fibrils in vitro. These results suggest that tau droplet formation and N-terminal cleavage are necessary for neurofibrillary tangles formation in neurodegenerative diseases.


Asunto(s)
Proteínas tau , Humanos , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Citoplasma/metabolismo , Luz , Ovillos Neurofibrilares/metabolismo , Fosforilación , Agregación Patológica de Proteínas/metabolismo , Proteínas tau/metabolismo , Proteínas tau/química , Proteínas tau/genética
14.
J Alzheimers Dis ; 101(2): 651-660, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39240637

RESUMEN

Background: The entorhinal cortex is the very earliest involvement of Alzheimer's disease (AD). Grid cells in the medial entorhinal cortex form part of the spatial navigation system. Objective: We aimed to determine whether path integration performance can be used to detect patients with mild cognitive impairment (MCI) at high risk of developing AD, and whether it can predict cognitive decline. Methods: Path integration performance was assessed in 71 patients with early MCI (EMCI) and late MCI (LMCI) using a recently developed 3D virtual reality navigation task. Patients with LMCI were further divided into those displaying characteristic brain imaging features of AD, including medial temporal lobe atrophy on magnetic resonance imaging and posterior hypoperfusion on single-photon emission tomography (LMCI+), and those not displaying such features (LMCI-). Results: Path integration performance was significantly lower in patients with LMCI+than in those with EMCI and LMCI-. A significantly lower performance was observed in patients who showed progression of MCI during 12 months, than in those with stable MCI. Path integration performance distinguished patients with progressive MCI from those with stable MCI, with a high classification accuracy (a sensitivity of 0.88 and a specificity of 0.70). Conclusions: Our results suggest that the 3D virtual reality navigation task detects prodromal AD patients and predicts cognitive decline after 12 months. Our navigation task, which is simple, short (12-15 minutes), noninvasive, and inexpensive, may be a screening tool for therapeutic choice of disease-modifiers in individuals with prodromal AD.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Imagen por Resonancia Magnética , Síntomas Prodrómicos , Humanos , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/patología , Masculino , Femenino , Anciano , Disfunción Cognitiva/diagnóstico por imagen , Disfunción Cognitiva/diagnóstico , Imagen por Resonancia Magnética/métodos , Progresión de la Enfermedad , Pruebas Neuropsicológicas , Tomografía Computarizada de Emisión de Fotón Único/métodos , Persona de Mediana Edad , Navegación Espacial/fisiología , Realidad Virtual , Anciano de 80 o más Años , Corteza Entorrinal/diagnóstico por imagen , Corteza Entorrinal/patología
15.
Sci Rep ; 14(1): 16391, 2024 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-39060263

RESUMEN

Prior to the formation of amyloid fibrils, the pathological hallmark in tau-related neurodegenerative disease, tau monomers aggregate into a diverse range of oligomers. Granular tau oligomers, consisting of approximately 40 tau protein molecules, are present in the prefrontal cortex of patients at Braak stages I-II, preclinical stages of Alzheimer's disease (AD). Antibodies to granular tau oligomers as antigens have not been reported. Therefore, we generated new rat monoclonal antibodies by immunization with granular tau oligomers. Three antibodies from different hybridoma clones showed stronger immunoreactivity to granular tau oligomers and tau fibrils compared with monomeric tau. Of the three antibodies, 2D6-2C6 showed 3000-fold greater immunoreactivity in P301L-tau transgenic (rTg4510) mice than in non-transgenic mice, while MC1 antibody, which detects pathological conformations of tau, showed a 5.5-fold increase. These results suggest that 2D6-2C6 recognizes aggregates more specifically than MC1. In AD subjects, 2D6-2C6 recognized neurofibrillary tangles and pretangles, and co-localized within AT8-positive cells containing phosphorylated tau aggregates. The epitope of 2D6-2C6 is the 423-430 amino acid (AA) sequence of C-terminal regions. Taken together, a novel monoclonal antibody, 2D6-2C6, generated by immunization with granular tau oligomers binds to tau aggregates at the 423-430 AA sequence.


Asunto(s)
Enfermedad de Alzheimer , Anticuerpos Monoclonales , Ratones Transgénicos , Proteínas tau , Proteínas tau/metabolismo , Proteínas tau/inmunología , Proteínas tau/química , Anticuerpos Monoclonales/inmunología , Animales , Humanos , Ratones , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/inmunología , Ratas , Inmunización , Femenino , Secuencia de Aminoácidos , Epítopos/inmunología , Masculino , Anciano , Agregado de Proteínas , Ovillos Neurofibrilares/metabolismo
16.
Brain Commun ; 6(1): fcad359, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38347945

RESUMEN

Alzheimer's disease is a devastating disease that is accompanied by dementia, and its incidence increases with age. However, no interventions have exhibited clear therapeutic effects. We aimed to develop and characterize behavioural tasks that allow the earlier identification of signs preceding dementia that would facilitate the development of preventative and therapeutic interventions for Alzheimer's disease. To this end, we developed a 3D virtual reality task sensitive to the activity of grid cells in the entorhinal cortex, which is the region that first exhibits neurofibrillary tangles in Alzheimer's disease. We investigated path integration (assessed by error distance) in a spatial navigation task sensitive to grid cells in the entorhinal cortex in 177 volunteers, aged 20-89 years, who did not have self-reported dementia. While place memory was intact even in old age, path integration deteriorated with increasing age. To investigate the relationship between neurofibrillary tangles in the entorhinal cortex and path integration deficit, we examined a mouse model of tauopathy (P301S mutant tau-overexpressing mice; PS19 mice). At 6 months of age, PS19 mice showed a significant accumulation of phosphorylated tau only in the entorhinal cortex, associated with impaired path integration without impairments in spatial cognition. These data are consistent with the idea that path integration deficit is caused by the accumulation of phosphorylated tau in the entorhinal cortex. This method may allow the early identification of individuals likely to develop Alzheimer's disease.

17.
Mov Disord Clin Pract ; 11(6): 720-727, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38605589

RESUMEN

BACKGROUND: MAPT is a causative gene in frontotemporal dementia with parkinsonism linked to chromosome 17 (FTDP-17), a hereditary degenerative disease with various clinical manifestations, including progressive supranuclear palsy, corticobasal syndrome, Parkinson's disease, and frontotemporal dementia. OBJECTIVES: To analyze genetically, biochemically, and pathologically multiple members of two families who exhibited various phenotypes of the disease. METHODS: Genetic analysis included linkage analysis, homozygosity haplotyping, and exome sequencing. We conducted tau protein microtubule polymerization assay, heparin-induced tau aggregation, and western blotting with brain lysate from an autopsy case. We also evaluated abnormal tau aggregation by using anti-tau antibody and PM-PBB3. RESULTS: We identified a variant, c.896_897insACA, p.K298_H299insQ, in the MAPT gene of affected patients. Similar to previous reports, most patients presented with atypical parkinsonism. Biochemical analysis revealed that the mutant tau protein had a reduced ability to polymerize microtubules and formed abnormal fibrous aggregates. Pathological study revealed frontotemporal lobe atrophy, midbrain atrophy, depigmentation of the substantia nigra, and four-repeat tau-positive inclusions in the hippocampus, brainstem, and spinal cord neurons. The inclusion bodies also stained positively with PM-PBB3. CONCLUSIONS: This study confirmed that the insACA mutation caused FTDP-17. The affected patients showed symptoms resembling Parkinson's disease initially and symptoms of progressive supranuclear palsy later. Despite the initial clinical diagnosis of frontotemporal dementia in the autopsy case, the spread of lesions could explain the process of progressive supranuclear palsy. The study of more cases in the future will help clarify the common pathogenesis of MAPT mutations or specific pathogeneses of each mutation.


Asunto(s)
Demencia Frontotemporal , Mutación , Proteínas tau , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Encéfalo/patología , Encéfalo/metabolismo , Cromosomas Humanos Par 17/genética , Demencia Frontotemporal/genética , Demencia Frontotemporal/patología , Demencia Frontotemporal/metabolismo , Demencia Frontotemporal/diagnóstico , Trastornos Parkinsonianos/genética , Trastornos Parkinsonianos/patología , Trastornos Parkinsonianos/metabolismo , Linaje , Parálisis Supranuclear Progresiva/genética , Parálisis Supranuclear Progresiva/patología , Proteínas tau/genética , Proteínas tau/metabolismo
18.
J Biol Chem ; 287(18): 14631-43, 2012 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-22393064

RESUMEN

Cerebral deposition of amyloid ß protein (Aß) is an invariant feature of Alzheimer disease (AD), and epidemiological evidence suggests that moderate consumption of foods enriched with phenolic compounds reduce the incidence of AD. We reported previously that the phenolic compounds myricetin (Myr) and rosmarinic acid (RA) inhibited Aß aggregation in vitro and in vivo. To elucidate a mechanistic basis for these results, we analyzed the effects of five phenolic compounds in the Aß aggregation process and in oligomer-induced synaptic toxicities. We now report that the phenolic compounds blocked Aß oligomerization, and Myr promoted significant NMR chemical shift changes of monomeric Aß. Both Myr and RA reduced cellular toxicity and synaptic dysfunction of the Aß oligomers. These results suggest that Myr and RA may play key roles in blocking the toxicity and early assembly processes associated with Aß through different binding.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Antioxidantes/farmacología , Cinamatos/farmacología , Depsidos/farmacología , Flavonoides/farmacología , Multimerización de Proteína/efectos de los fármacos , Sinapsis/metabolismo , Animales , Células HEK293 , Humanos , Ratones , Sinapsis/patología , Ácido Rosmarínico
19.
Biochem Biophys Res Commun ; 440(4): 677-82, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24099767

RESUMEN

Glycogen synthase kinase (GSK)-3ß plays an important role in osteoblastogenesis by regulating the Wnt/ß-catenin signaling pathway. Therefore, we investigated whether GSK-3ß deficiency affects bone development and regeneration using mice heterozygously deficient for GSK-3ß (GSK-3ß(+/-)). The amounts of ß-catenin, c-Myc, cyclin D1, and runt-related transcription factor-2 (Runx2) in the bone marrow cells of GSK-3ß(+/-) mice were significantly increased compared with those of wild-type mice, indicating that Wnt/ß-catenin signals were enhanced in GSK-3ß(+/-) mice. Microcomputed tomography of the distal femoral metaphyses demonstrated that the volumes of both the cortical and trabecular bones were increased in GSK-3ß(+/-) mice compared with those in wild-type mice. Subsequently, to investigate the effect of GSK-3ß deficiency on bone regeneration, we established a partial bone defect in the femur and observed new bone at 14 days after surgery. The volume and mineral density of the new bone were significantly higher in GSK-3ß(+/-) mice than those in wild-type mice. These results suggest that bone formation and regeneration in vivo are accelerated by inhibition of GSK-3ß, probably through activation of the Wnt/ß-catenin signaling pathway.


Asunto(s)
Desarrollo Óseo , Regeneración Ósea , Glucógeno Sintasa Quinasa 3/metabolismo , Osteoblastos/fisiología , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Ciclina D1/metabolismo , Glucógeno Sintasa Quinasa 3/genética , Ratones , Osteoblastos/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal
20.
J Neurosci ; 31(21): 7840-7, 2011 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-21613497

RESUMEN

Stressful life experiences are likely etiological factors in sporadic forms of Alzheimer's disease (AD). Many AD patients hypersecrete glucocorticoids (GCs), and their GC levels correlate with the rate of cognitive impairment and extent of neuronal atrophy. Severity of cognitive deficits in AD correlates strongly with levels of hyperphosphorylated forms of the cytoskeletal protein TAU, an essential mediator of the actions of amyloid ß (Aß), another molecule with a key pathogenic role in AD. Our objective was to investigate the sequential interrelationships between these various pathogenic elements, in particular with respect to the mechanisms through which stress might precipitate cognitive decline. We thus examined whether stress, through the mediation of GCs, influences TAU hyperphosphorylation, a critical and early event in the cascade of processes leading to AD pathology. Results from healthy, wild-type, middle-aged rats show that chronic stress and GC induce abnormal hyperphosphorylation of TAU in the hippocampus and prefrontal cortex (PFC), with contemporaneous impairments of hippocampus- and PFC-dependent behaviors. Exogenous GC potentiated the ability of centrally infused Aß to induce hyperphosphorylation of TAU epitopes associated with AD and cytoplasmic accumulation of TAU, while previous exposure to stress aggravated the biochemical and behavioral effects of GC in Aß-infused animals. Thus, lifetime stress/GC exposure may have a cumulative impact on the onset and progress of AD pathology, with TAU hyperphosphorylation serving to transduce the negative effects of stress and GC on cognition.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Trastornos del Conocimiento/metabolismo , Estrés Psicológico/metabolismo , Proteínas tau/biosíntesis , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/patología , Animales , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/patología , Glucocorticoides/fisiología , Glucocorticoides/toxicidad , Masculino , Fosforilación/fisiología , Distribución Aleatoria , Ratas , Ratas Wistar , Estrés Psicológico/inducido químicamente , Estrés Psicológico/complicaciones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA