Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(12): e2117723119, 2022 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-35290109

RESUMO

Type 2 diabetes mellitus is known to be a risk factor for Alzheimer's disease (AD), but the underlying mechanisms remain unclear. In AD, the cerebral accumulation of amyloid ß (Aß) triggers a pathological cascade leading to neurodegeneration. Plasma Aß levels are thought to reflect the brain amyloid pathology and currently used as a diagnostic biomarker of AD. However, amyloid precursor protein and Aß-generating enzymes, ß- and γ-secretases, are widely expressed in various peripheral tissues. Previous reports have shown that glucose and insulin loading cause a transient increase of plasma Aß in mice and humans. These findings led us to speculate that plasma Aß is produced from glucose- and insulin-susceptible peripheral tissues to play a role in glucose and insulin metabolism. To test this hypothesis, we investigated the effects of glucose and insulin on Aß secretion and the effect of Aß on insulin secretion in vivo, ex vivo, and in vitro. Aß was found to be secreted from ß-cells of the pancreas along with insulin upon glucose stimulation. Upon insulin stimulation, Aß was secreted from cells of insulin-targeted organs, such as adipose tissues, skeletal muscles, and the liver, along with their organokines. Furthermore, Aß inhibited the glucose-triggered insulin secretion from ß-cells, slowing down glucose clearance from the blood. These results suggest that peripheral Aß acts as a negative modulator of insulin secretion. Our findings provide a possible mechanism linking diabetes to AD and call attention to how plasma Aß levels are used in AD diagnosis.


Assuntos
Doença de Alzheimer , Diabetes Mellitus Tipo 2 , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Glucose/metabolismo , Humanos , Insulina/metabolismo , Secreção de Insulina
2.
J Nanobiotechnology ; 21(1): 36, 2023 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-36721182

RESUMO

BACKGROUND: Although a large amount of evidence has revealed that amyloid ß (Aß), especially Aß oligomers, protofibrils, and pyroglutamated Aßs, participate primarily in the pathophysiological processes of Alzheimer's disease, most clinical trials of anti-Aß antibody therapy have never acquired successful efficacy in human clinical trials, partly because peripheral administration of antibody medications was unable to deliver sufficient amounts of the molecules to the brain. Recently, we developed polymeric nanomicelles capable of passing through the blood-brain barrier that function as chaperones to deliver larger amounts of heavy molecules to the brain. Herein, we aimed to evaluate the efficacy of newly developed antibody 6H4 fragments specific to Aß oligomers encapsulated in polymeric nanomicelles on the development of Alzheimer's disease pathology in Alzheimer's disease model mice at the age of emergence of early Alzheimer's disease pathology. RESULTS: During the 10-week administration of 6H4 antibody fragments in polymeric nanomicelles, a significant reduction in the amounts of various toxic Aß species, such as Aß oligomers, toxic Aß conformers, and pyroglutamated Aßs in the brain was observed. In addition, immunohistochemistry indicated inhibition of diameters of Aß plaques, Aß-antibody immunoreactive areas, and also plaque core formation. Behavioral analysis of the mice model revealed that the 6H4 fragments-polymeric nanomicelle group was significantly better at maintaining long-term spatial reference memory in the probe and platform tests of the water maze, thereby indicating inhibition of the pathophysiological process of Alzheimer's disease. CONCLUSIONS: The results indicated that the strategy of reducing toxic Aß species in early dementia owing to Alzheimer's disease by providing sufficient antibodies in the brain may modify Alzheimer's disease progression.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Humanos , Animais , Camundongos , Doença de Alzheimer/tratamento farmacológico , Encéfalo , Barreira Hematoencefálica , Anticorpos , Placa Amiloide , Polímeros
3.
Int J Mol Sci ; 22(16)2021 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-34445158

RESUMO

α-Synuclein oligomers are thought to play an important role in the pathogenesis of dementia with Lewy bodies (DLB). There is no effective cure for DLB at present. Previously, we demonstrated that in APP- and tau-transgenic mice, oral or intranasal rifampicin reduced brain Aß and tau oligomers and improved mouse cognition. In the present study, we expanded our research to DLB. Rifampicin was intranasally administered to 6-month-old A53T-mutant α-synuclein-transgenic mice at 0.1 mg/day for 1 month. The mice displayed memory impairment but no motor deficit at this age, indicating a suitable model of DLB. α-Synuclein pathologies were examined by the immunohistochemical/biochemical analyses of brain tissues. Cognitive function was evaluated by the Morris water maze test. Intranasal rifampicin significantly reduced the levels of [pSer129] α-synuclein in the hippocampus and α-synuclein oligomers in the visual cortex and hippocampus. The level of the presynaptic marker synaptophysin in the hippocampus was recovered to the level in non-transgenic littermates. In the Morris water maze, a significant improvement in spatial reference memory was observed in rifampicin-treated mice. Taken together with our previous findings, these results suggest that intranasal rifampicin is a promising remedy for the prevention of neurodegenerative dementia, including Alzheimer's disease, frontotemporal dementia, and DLB.


Assuntos
Cognição/efeitos dos fármacos , Demência/tratamento farmacológico , Doença por Corpos de Lewy/tratamento farmacológico , Rifampina/uso terapêutico , alfa-Sinucleína/metabolismo , Administração Intranasal , Animais , Demência/metabolismo , Demência/patologia , Modelos Animais de Doenças , Feminino , Corpos de Lewy/efeitos dos fármacos , Corpos de Lewy/metabolismo , Corpos de Lewy/patologia , Doença por Corpos de Lewy/metabolismo , Doença por Corpos de Lewy/patologia , Masculino , Camundongos Transgênicos , Multimerização Proteica/efeitos dos fármacos , Rifampina/administração & dosagem , alfa-Sinucleína/análise
4.
Molecules ; 26(5)2021 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-33806326

RESUMO

Recent evidence suggests that the formation of soluble amyloid ß (Aß) aggregates with high toxicity, such as oligomers and protofibrils, is a key event that causes Alzheimer's disease (AD). However, understanding the pathophysiological role of such soluble Aß aggregates in the brain in vivo could be difficult due to the lack of a clinically available method to detect, visualize, and quantify soluble Aß aggregates in the brain. We had synthesized a novel fluorinated curcumin derivative with a fixed keto form, named as Shiga-Y51, which exhibited high selectivity to Aß oligomers in vitro. In this study, we investigated the in vivo detection of Aß oligomers by fluorine-19 (19F) magnetic resonance imaging (MRI) using Shiga-Y51 in an APP/PS1 double transgenic mouse model of AD. Significantly high levels of 19F signals were detected in the upper forebrain region of APP/PS1 mice compared with wild-type mice. Moreover, the highest levels of Aß oligomers were detected in the upper forebrain region of APP/PS1 mice in enzyme-linked immunosorbent assay. These findings suggested that 19F-MRI using Shiga-Y51 detected Aß oligomers in the in vivo brain. Therefore, 19F-MRI using Shiga-Y51 with a 7 T MR scanner could be a powerful tool for imaging Aß oligomers in the brain.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/patologia , Curcumina/metabolismo , Modelos Animais de Doenças , Imagem por Ressonância Magnética de Flúor-19/métodos , Placa Amiloide/patologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/química , Animais , Encéfalo/metabolismo , Curcumina/química , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Placa Amiloide/metabolismo
5.
J Neurosci ; 39(34): 6781-6797, 2019 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-31235644

RESUMO

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.


Assuntos
Química Encefálica/fisiologia , Encéfalo/citologia , Dendritos/fisiologia , Expressão Ectópica do Gene/genética , Proteínas tau/biossíntese , Animais , Animais Recém-Nascidos , Axônios/metabolismo , Encéfalo/crescimento & desenvolvimento , Feminino , Técnicas de Introdução de Genes , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Cultura Primária de Células , Tauopatias/metabolismo
6.
Int J Mol Sci ; 21(4)2020 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-32093100

RESUMO

Alzheimer's disease is believed to begin with synaptic dysfunction caused by soluble Aß oligomers. When this oligomer hypothesis was proposed in 2002, there was no direct evidence that Aß oligomers actually disrupt synaptic function to cause cognitive impairment in humans. In patient brains, both soluble and insoluble Aß species always coexist, and therefore it is difficult to determine which pathologies are caused by Aß oligomers and which are caused by amyloid fibrils. Thus, no validity of the oligomer hypothesis was available until the Osaka mutation was discovered. This mutation, which was found in a Japanese pedigree of familial Alzheimer's disease, is the deletion of codon 693 of APP gene, resulting in mutant Aß lacking the 22nd glutamate. Only homozygous carriers suffer from dementia. In vitro studies revealed that this mutation has a very unique character that accelerates Aß oligomerization but does not form amyloid fibrils. Model mice expressing this mutation demonstrated that all pathologies of Alzheimer's disease can be induced by Aß oligomers alone. In this review, we describe the story behind the discovery of the Osaka mutation, summarize the mutant's phenotypes, and propose a mechanism of its recessive inheritance.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Doença de Alzheimer/congênito , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Modelos Animais de Doenças , Hereditariedade , Homozigoto , Humanos , Japão , Camundongos , Linhagem , Fenótipo , Placa Amiloide/patologia , Deleção de Sequência
7.
Int J Mol Sci ; 21(12)2020 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-32580499

RESUMO

We previously identified a novel mutation in amyloid precursor protein from a Japanese pedigree of familial Alzheimer's disease, FAD (Osaka). Our previous positron emission tomography (PET) study revealed that amyloid ß (Aß) accumulation was negligible in two sister cases of this pedigree, indicating a possibility that this mutation induces dementia without forming senile plaques. To further explore the relationship between Aß, tau and neurodegeneration, we performed tau and Aß PET imaging in the proband of FAD (Osaka) and in patients with sporadic Alzheimer's disease (SAD) and healthy controls (HCs). The FAD (Osaka) patient showed higher uptake of tau PET tracer in the frontal, lateral temporal, and parietal cortices, posterior cingulate gyrus and precuneus than the HCs (>2.5 SD) and in the lateral temporal and parietal cortices than the SAD patients (>2 SD). Most noticeably, heavy tau tracer accumulation in the cerebellum was found only in the FAD (Osaka) patient. Scatter plot analysis of the two tracers revealed that FAD (Osaka) exhibits a distinguishing pattern with a heavy tau burden and subtle Aß accumulation in the cerebral cortex and cerebellum. These observations support our hypothesis that Aß can induce tau accumulation and neuronal degeneration without forming senile plaques.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Cerebelo/metabolismo , Córtex Cerebral/metabolismo , Mutação , Proteínas tau/metabolismo , Idoso , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Cerebelo/patologia , Córtex Cerebral/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
8.
Brain ; 139(Pt 5): 1568-86, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27020329

RESUMO

Amyloid-ß, tau, and α-synuclein, or more specifically their soluble oligomers, are the aetiologic molecules in Alzheimer's disease, tauopathies, and α-synucleinopathies, respectively. These proteins have been shown to interact to accelerate each other's pathology. Clinical studies of amyloid-ß-targeting therapies in Alzheimer's disease have revealed that the treatments after disease onset have little benefit on patient cognition. These findings prompted us to explore a preventive medicine which is orally available, has few adverse effects, and is effective at reducing neurotoxic oligomers with a broad spectrum. We initially tested five candidate compounds: rifampicin, curcumin, epigallocatechin-3-gallate, myricetin, and scyllo-inositol, in cells expressing amyloid precursor protein (APP) with the Osaka (E693Δ) mutation, which promotes amyloid-ß oligomerization. Among these compounds, rifampicin, a well-known antibiotic, showed the strongest activities against the accumulation and toxicity (i.e. cytochrome c release from mitochondria) of intracellular amyloid-ß oligomers. Under cell-free conditions, rifampicin inhibited oligomer formation of amyloid-ß, tau, and α-synuclein, indicating its broad spectrum. The inhibitory effects of rifampicin against amyloid-ß and tau oligomers were evaluated in APPOSK mice (amyloid-ß oligomer model), Tg2576 mice (Alzheimer's disease model), and tau609 mice (tauopathy model). When orally administered to 17-month-old APPOSK mice at 0.5 and 1 mg/day for 1 month, rifampicin reduced the accumulation of amyloid-ß oligomers as well as tau hyperphosphorylation, synapse loss, and microglial activation in a dose-dependent manner. In the Morris water maze, rifampicin at 1 mg/day improved memory of the mice to a level similar to that in non-transgenic littermates. Rifampicin also inhibited cytochrome c release from the mitochondria and caspase 3 activation in the hippocampus. In 13-month-old Tg2576 mice, oral rifampicin at 0.5 mg/day for 1 month decreased amyloid-ß oligomer accumulation, tau hyperphosphorylation, synapse loss, and microglial activation, but not amyloid deposition. Rifampicin treatment to 14-15-month-old tau609 mice at 0.5 and 1 mg/day for 1 month also reduced tau oligomer accumulation, tau hyperphosphorylation, synapse loss, and microglial activation in a dose-dependent fashion, and improved the memory almost completely at 1 mg/day. In addition, rifampicin decreased the level of p62/sequestosome-1 in the brain without affecting the increased levels of LC3 (microtubule-associated protein light chain 3) conversion, suggesting the restoration of autophagy-lysosomal function. Considering its prescribed dose and safety in humans, these results indicate that rifampicin could be a promising, ready-to-use medicine for the prevention of Alzheimer's disease and other neurodegenerative diseases.


Assuntos
Doença de Alzheimer/prevenção & controle , Peptídeos beta-Amiloides/efeitos dos fármacos , Rifampina/farmacologia , Rifampina/uso terapêutico , Tauopatias/prevenção & controle , Proteínas tau/efeitos dos fármacos , Doença de Alzheimer/complicações , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Caspase 3/metabolismo , Células Cultivadas , Citocromos c/metabolismo , Relação Dose-Resposta a Droga , Feminino , Hipocampo/metabolismo , Aprendizagem em Labirinto/efeitos dos fármacos , Transtornos da Memória/complicações , Transtornos da Memória/tratamento farmacológico , Camundongos , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/metabolismo , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Fosforilação/efeitos dos fármacos , Proteína Sequestossoma-1/metabolismo , Sinapses/efeitos dos fármacos , Sinucleínas/efeitos dos fármacos , Sinucleínas/metabolismo , Tauopatias/complicações , Tauopatias/metabolismo , Proteínas tau/metabolismo
9.
Neuropathology ; 37(6): 495-501, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28731225

RESUMO

Protein disulfide isomerase (PDI) is a chaperone protein located in the endoplasmic reticulum (ER). Nitric oxide-induced S-nitrosylation of PDI inhibits its enzymatic activity, leading to protein accumulation and activation of the unfolded protein response. Protein disulfide isomerase P5 (P5) is a member of the PDI family that mostly localizes to the ER lumen. Both S-nitrosylated PDI and S-nitrosylated P5 are found in Alzheimer's disease (AD) brain. Previously, we showed that expression of the ER stress marker, growth arrest, and DNA damage protein (GADD34) was significantly increased in neurons and oligodendrocytes in AD brain. In the present study, we showed that PDI and P5 levels were significantly decreased in oligodendrocytes in the brains of AD patients and an AD mouse model. Interestingly, these decreases were evident before the animals displayed typical AD pathology. Because we previously showed that small short interfering RNA knockdown of PDI or P5 could affect the viability of neuronal cells under ER stress, dysfunction of PDI and P5 under ER stress could cause apoptosis of neuronal cells. In summary, we showed that the levels of PDI and P5 were significantly decreased in the oligodendrocytes of AD patients. This phenomenon was also found in an AD mouse model before the animals displayed AD pathology. The overall findings suggest that oligodendrocytes may play important roles in AD pathogenesis.


Assuntos
Doença de Alzheimer/enzimologia , Encéfalo/enzimologia , Oligodendroglia/enzimologia , Isomerases de Dissulfetos de Proteínas/biossíntese , Idoso , Idoso de 80 Anos ou mais , Animais , Feminino , Humanos , Masculino , Camundongos , Isomerases de Dissulfetos de Proteínas/análise
10.
Am J Pathol ; 183(1): 211-25, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23680655

RESUMO

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.


Assuntos
Éxons , Demência Frontotemporal/genética , Íntrons , Mutação , Splicing de RNA , Tauopatias/genética , Proteínas tau/genética , Animais , Western Blotting , Demência Frontotemporal/patologia , Demência Frontotemporal/fisiopatologia , Marcadores Genéticos , Masculino , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tauopatias/patologia , Tauopatias/fisiopatologia
11.
Acta Neuropathol ; 127(5): 685-98, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24531886

RESUMO

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.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Emaranhados Neurofibrilares/metabolismo , Proteínas tau/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/patologia , Progressão da Doença , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Transtornos da Memória/metabolismo , Transtornos da Memória/patologia , Camundongos Transgênicos , Mutação , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Emaranhados Neurofibrilares/patologia , Neurônios/metabolismo , Neurônios/patologia , Fosforilação , Sinapses/metabolismo , Sinapses/patologia , Tauopatias/metabolismo , Tauopatias/patologia , Fatores de Tempo , Proteínas tau/genética
12.
Geroscience ; 46(2): 1971-1987, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37783918

RESUMO

Neurodegenerative diseases including Alzheimer's disease, frontotemporal dementia, and dementia with Lewy bodies are age-related disorders and the main cause of dementia. They are characterized by the cerebral accumulation of Aß, tau, α-synuclein, and TDP-43. Because the accumulation begins decades before disease onset, treatment should be started in the preclinical stage. Such intervention would be long-lasting, and therefore, prophylactic agents should be safe, non-invasively taken by the patients, and inexpensive. In addition, the agents should be broadly effective against etiologic proteins and capable of repairing neurons damaged by toxic oligomers. These requirements are difficult to meet with single-ingredient pharmaceuticals but may be feasible by taking proper diets composed of multiple ingredients. As a source of such diets, we focused on the Hawaiian native herb Mamaki. From its dried leaves and fruits, we made three preparations: hot water extract of the leaves, non-extracted simple crush powder of the leaves, and simple crush powder of the fruits, and examined their effects on the cognitive function and neuropathologies in four different mouse models of neurodegenerative dementia. Hot water extract of the leaves attenuated neuropathologies, restored synaptophysin levels, suppressed microglial activation, and improved memory when orally administered for 1 month. Simply crushed leaf powder showed a higher efficacy, but simply crushed fruit powder displayed the strongest effects. Moreover, the fruit powder significantly enhanced the levels of brain-derived neurotrophic factor expression and neurogenesis, indicating its ability to repair neurons. These results suggest that crushed Mamaki leaves and fruits are promising sources of dementia-preventive diets.


Assuntos
Doença de Alzheimer , Doenças Neurodegenerativas , Camundongos , Animais , Humanos , Doenças Neurodegenerativas/prevenção & controle , Havaí , Pós , Doença de Alzheimer/prevenção & controle , Doença de Alzheimer/metabolismo , Neurônios/metabolismo , Água
13.
Brain Res ; 1838: 148987, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38718851

RESUMO

Dynamin is a microtubule (MT) binding protein playing a key role in vesicle endocytosis. In a brain slice model, tau loaded in presynaptic terminals assembles MTs, thereby impairing vesicle endocytosis via depletion of cytosolic dynamin. The peptide PHDP5, derived from the pleckstrin homology domain of dynamin 1, inhibits dynamin-MT interaction and rescues endocytosis and synaptic transmission impaired by tau when co-loaded in presynaptic terminals. We tested whether in vivo administration of PHDP5 could rescue the learning/memory deficits observed in Alzheimer's disease (AD) model mice. A modified PHDP5 incorporating a cell-penetrating peptide (CPP) and a FITC fluorescent marker was delivered intranasally to Tau609 transgenic (Tg) and 3xTg-AD mice. FITC-positive puncta were observed in the hippocampus of mice infused with PHDP5 or scrambled (SPHDP5) peptide, but not in saline-infused controls. In the Morris water maze (MWM) test for spatial learning/memory, AD model mice treated with FITC-PHDP5-CPP showed prominent improvements in learning and memory, performing close to the level of saline-infused WT mice control. In contrast, mice treated with a scrambled construct (FITC-SPHDP5-CPP) showed no significant improvement. We conclude that PHDP5 can be a candidate for human AD therapy.


Assuntos
Doença de Alzheimer , Modelos Animais de Doenças , Transtornos da Memória , Camundongos Transgênicos , Aprendizagem Espacial , Animais , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Camundongos , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/metabolismo , Aprendizagem Espacial/efeitos dos fármacos , Microtúbulos/metabolismo , Microtúbulos/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/efeitos dos fármacos , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Dinaminas/metabolismo , Masculino , Proteínas tau/metabolismo
14.
Nutrients ; 16(11)2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38892521

RESUMO

The rhizomes of Acorus tatarinowii Schott and Acorus gramineus Solander are widely used for treating amnesia in traditional Chinese medicine. In contrast, their leaves are usually discarded without their medicinal properties being known. Here, we found that the hot water extract of leaves improved cognition and tau pathology in model mice of frontotemporal dementia, similar to or even better than that of rhizomes. To explore the optimal method of processing, we made three preparations from dried leaves: hot water extract, extraction residue, and non-extracted simple crush powder. Among them, the simple crush powder had the strongest effect on tauopathy in mice. The crush powder also ameliorated Aß and α-synuclein pathologies and restored cognition in mouse models of Alzheimer's disease and dementia with Lewy bodies. These findings suggest the potential of Acorus tatarinowii/gramineus leaves as a dietary source for dementia prevention and reveal that simple crushing is a better way to maximize their efficacy.


Assuntos
Acorus , Demência , Extratos Vegetais , Folhas de Planta , Animais , Folhas de Planta/química , Acorus/química , Camundongos , Extratos Vegetais/farmacologia , Demência/prevenção & controle , Modelos Animais de Doenças , Cognição/efeitos dos fármacos , Peptídeos beta-Amiloides/metabolismo , Masculino , Doença de Alzheimer/prevenção & controle , Proteínas tau/metabolismo
15.
J Control Release ; 367: 515-521, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38237689

RESUMO

This study explored the effectiveness of nasal administration in delivering magnetic nanoparticles into the brain for magnetic particle imaging of target regions. Successful delivery of iron oxide nanoparticles, which serve as contrast agents, to specific sites within the brain is crucial for achieving magnetic particle imaging. Nasal administration has gained attention as a method to bypass the blood-brain barrier and directly deliver therapeutics to the brain. In this study, we investigated surface modification techniques for administering magnetic nanoparticles into the nasal cavity, and provided experimental validation through in vivo studies. By compositing magnetic nanoparticles with gold nanoparticles, we enabled additional surface modification via AuS bonds without compromising their magnetic properties. The migration of the designed PEGylated magnetic nanoparticles into the brain following nasal administration was confirmed by magnetization measurements. Furthermore, we demonstrated the accumulation of these nanoparticles at specific target sites using probe molecules immobilized on the PEG terminus. Thus, the efficacy of delivering magnetic nanoparticles to the brain via nasal administration was demonstrated in this study. The findings of this research are expected to contribute significantly to the realization of magnetic particle imaging of target regions within the brain.


Assuntos
Nanopartículas de Magnetita , Nanopartículas , Administração Intranasal , Nanopartículas de Magnetita/química , Ouro , Encéfalo/diagnóstico por imagem , Nanopartículas/química , Fenômenos Magnéticos , Tamanho da Partícula , Sistemas de Liberação de Medicamentos
16.
J Neurosci Res ; 91(12): 1541-50, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24105984

RESUMO

It has been shown that amyloid ß (Aß) secretion regulates cholesterol efflux from cells and that the E693Δ (Osaka) mutation in amyloid precursor protein (APP) promotes intracellular accumulation of Aß and thus reduces its secretion. These findings led us to speculate that APP with the Osaka mutation (APPOSK ) might have a defect in cholesterol efflux and thus cause cellular malfunction. We therefore examined the effects of this mutation on intracellular cholesterol transport and efflux in cultured cells. Upon cholesterol loading, APPOSK -expressing cells exhibited higher levels of cellular cholesterol than wild-type APP-expressing cells, suggesting impaired cholesterol efflux. It is known that, after its internalization, cholesterol is transported from the endosomes to the endoplasmic reticulum (ER) and Golgi apparatus and then to the plasma membrane. In APPOSK -expressing cells, cholesterol accumulated with Aß in the ER and Golgi apparatus and alone in endosomes/lysosomes. These results imply that the mutation-induced disturbance of Aß trafficking from the ER to the plasma membrane affects cholesterol transport to cause cholesterol accumulation in the ER and Golgi apparatus and, consequently, in endosomes. Furthermore, we detected an enhanced mitochondrial accumulation of Aß and cholesterol in APPOSK -expressing cells, and this was accompanied by an increase in the generation of reactive oxygen species (ROS). The present findings suggest that Aß trafficking is important for intracellular cholesterol transport and efflux and that the Osaka mutation potentiates cholesterol-dependent exacerbation of intracellular Aß toxicity, i.e. Aß-induced ROS generation, by disturbing Aß-mediated cholesterol efflux from the cell.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Colesterol/metabolismo , Mutação , Transporte Proteico/genética , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Células HEK293 , Humanos , Imuno-Histoquímica , Espaço Intracelular/metabolismo , Transfecção
17.
J Neurosci ; 31(12): 4720-30, 2011 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-21430171

RESUMO

Core pathologies of Alzheimer's disease (AD) are aggregated amyloid-ß peptides (Aß) and tau, and the latter is also characteristic of diverse neurodegenerative tauopathies. These amyloid lesions provoke microglial activation, and recent neuroimaging technologies have enabled visualization of this response in living brains using radioligands for the peripheral benzodiazepine receptor also known as the 18 kDa translocator protein (TSPO). Here, we elucidated contributions of Aß and tau deposits to in vivo TSPO signals in pursuit of mechanistic and diagnostic significance of TSPO imaging in AD and other tauopathies. A new antibody to human TSPO revealed induction of TSPO-positive microgliosis by tau fibrils in tauopathy brains. Emergence of TSPO signals before occurrence of brain atrophy and thioflavin-S-positive tau amyloidosis was also demonstrated in living mice transgenic for mutant tau by positron emission tomography (PET) with two classes of TSPO radioligands, [(11)C]AC-5216 and [(18)F]fluoroethoxy-DAA1106. Meanwhile, only modest TSPO elevation was observed in aged mice modeling Aß plaque deposition, despite the notably enhanced in vivo binding of amyloid radiotracer, [(11)C]Pittsburgh Compound-B, to plaques. In these animals, [(11)C]AC-5216 yielded better TSPO contrasts than [(18)F]fluoroethoxy-DAA1106, supporting the possibility of capturing early neurotoxicity with high-performance TSPO probes. Furthermore, an additional line of mice modeling intraneuronal Aß accumulation displayed elevated TSPO signals following noticeable neuronal loss, unlike TSPO upregulation heralding massive neuronal death in tauopathy model mice. Our data corroborate the utility of TSPO-PET imaging as a biomarker for tau-triggered toxicity, and as a complement to amyloid scans for diagnostic assessment of tauopathies with and without Aß pathologies.


Assuntos
Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Neuroglia/diagnóstico por imagem , Neuroglia/patologia , Proteínas tau/metabolismo , Acetamidas/síntese química , Compostos de Anilina , Animais , Autorradiografia , Encéfalo/patologia , Humanos , Imuno-Histoquímica , Marcação por Isótopo/métodos , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos C57BL , Neurite (Inflamação)/patologia , Doença de Pick/patologia , Placa Amiloide/patologia , Tomografia por Emissão de Pósitrons , Purinas/síntese química , Compostos Radiofarmacêuticos/síntese química , Receptores de GABA/metabolismo , Paralisia Supranuclear Progressiva/patologia , Tiazóis
18.
Biomedicines ; 10(2)2022 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-35203506

RESUMO

The cell-to-cell transmission of tau aggregates is considered a mechanism underlying the intracerebral spreading of tau pathology in Alzheimer's disease (AD) and other tauopathies. Recent studies suggest that tau oligomers, rather than fibrils, participate in this process. We previously showed that intranasal rifampicin inhibits tau oligomer accumulation and improves cognition in tauopathy mice. In the present study, we examined the effects of nasal rifampicin on tau propagation in a new mouse model of tauopathy. A tau oligomer-rich fraction prepared from the brain of an AD patient was injected into a unilateral hippocampus of tau264 mice that express both 3-repeat and 4-repeat wild-type human tau. Rifampicin administration was started one week after the injection and performed three times a week for 24 weeks. Cognitive function and tau pathology were assessed by the Morris water maze test and brain section staining. Rifampicin treatment inhibited the spreading of tau oligomers from the injection site to other brain regions and neurofibrillary tangle formation in the entorhinal cortex. Synapse and neuronal loss in the hippocampus were also prevented, and cognitive function remained normal. These results suggest that intranasal rifampicin could be a promising remedy that halts the progression of tauopathy by inhibiting tau oligomer propagation.

19.
Biomedicines ; 10(5)2022 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-35625816

RESUMO

The non-coding GGGGCC hexanucleotide repeat expansion (HRE) in C9orf72 gene is a dominant cause of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). This intronic mutation elicits the formation of nuclear and cytoplasmic inclusions containing RNA, RNA-binding proteins, and HRE-derived dipeptide repeat proteins (DPRs), leading to neurodegeneration via the gain-of-toxic function or loss-of-function of relevant proteins. Using C9-500 mice harboring ~500 repeats of the GGGGCC sequence in human C9orf72 gene, we investigated the effects of rifampicin against HRE-related pathological phenotypes. Rifampicin was administered intranasally to 4.5- to 5-month-old mice for 1 month, and their cognitive function and neuropathology were assessed by the Morris water maze test and immunohistochemical staining. Rifampicin treatment reduced the formation of RNA foci and cytoplasmic inclusions containing DPRs or phosphorylated TDP-43, and furthermore, the levels of phosphorylated double-strand RNA-dependent protein kinase (PKR) that regulates repeat-associated non-ATG (RAN) translation. Synapse loss in the hippocampus and neuronal loss and microglial activation in the prefrontal and motor cortices were also attenuated, and mouse memory was significantly improved. Our findings suggest a therapeutic potential of nasal rifampicin in the prevention of C9orf72-linked neurodegenerative disorders.

20.
J Neurosci ; 30(14): 4845-56, 2010 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-20371804

RESUMO

Although amyloid beta (Abeta) oligomers are presumed to cause synaptic and cognitive dysfunction in Alzheimer's disease (AD), their contribution to other pathological features of AD remains unclear. To address the latter, we generated APP transgenic mice expressing the E693Delta mutation, which causes AD by enhanced Abeta oligomerization without fibrillization. The mice displayed age-dependent accumulation of intraneuronal Abeta oligomers from 8 months but no extracellular amyloid deposits even at 24 months. Hippocampal synaptic plasticity and memory were impaired at 8 months, at which time the presynaptic marker synaptophysin began to decrease. Furthermore, we detected abnormal tau phosphorylation from 8 months, microglial activation from 12 months, astrocyte activation from 18 months, and neuronal loss at 24 months. These findings suggest that Abeta oligomers cause not only synaptic alteration but also other features of AD pathology and that these mice are a useful model of Abeta oligomer-induced pathology in the absence of amyloid plaques.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/fisiologia , Neuroglia/metabolismo , Neurônios/metabolismo , Sinapses/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Morte Celular/genética , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neuroglia/patologia , Neurônios/patologia , Fosforilação/genética , Sinapses/patologia , Proteínas tau/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA