Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Proc Natl Acad Sci U S A ; 119(12): e2117723119, 2022 03 22.
Article in English | MEDLINE | ID: mdl-35290109

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Diabetes Mellitus, Type 2 , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Humans , Insulin/metabolism , Insulin Secretion
2.
Sensors (Basel) ; 21(5)2021 Mar 09.
Article in English | MEDLINE | ID: mdl-33803456

ABSTRACT

The 10-s grip and release is a method to evaluate hand dexterity. Current evaluations only visually determine the presence or absence of a disability, but experienced physicians may also make other diagnoses. In this study, we investigated a method for evaluating hand movement function by acquiring and analyzing fingertip data during a 10-s grip and release using a wearable sensor that can measure triaxial acceleration and strain. The subjects were two healthy females. The analysis was performed on the x-, y-, and z-axis data, and absolute acceleration and contact force of all fingertips. We calculated the variability of the data, the number of grip and release, the frequency response, and each finger's correlation. Experiments with some grip-and-release patterns have resulted in different characteristics for each. It was suggested that this could be expressed in radar charts to intuitively know the state of grip and release. Contact-force data of each finger were found to be useful for understanding the characteristics of grip and release and improving the accuracy of calculating the number of times to grip and release. Frequency analysis suggests that knowing the periodicity of grip and release can detect unnatural grip and release and tremor states. The correlations between the fingers allow us to consider the finger's grip-and-release characteristics, considering the hand's anatomy. By taking these factors into account, it is thought that the 10-s grip-and-release test could give us a new value by objectively assessing the motor functions of the hands other than the number of times of grip and release.


Subject(s)
Fingers , Hand Strength , Acceleration , Female , Hand , Humans , Movement
3.
Int J Mol Sci ; 22(16)2021 Aug 06.
Article in English | MEDLINE | ID: mdl-34445158

ABSTRACT

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.


Subject(s)
Cognition/drug effects , Dementia/drug therapy , Lewy Body Disease/drug therapy , Rifampin/therapeutic use , alpha-Synuclein/metabolism , Administration, Intranasal , Animals , Dementia/metabolism , Dementia/pathology , Disease Models, Animal , Female , Lewy Bodies/drug effects , Lewy Bodies/metabolism , Lewy Bodies/pathology , Lewy Body Disease/metabolism , Lewy Body Disease/pathology , Male , Mice, Transgenic , Protein Multimerization/drug effects , Rifampin/administration & dosage , alpha-Synuclein/analysis
4.
Brain ; 139(Pt 5): 1568-86, 2016 05.
Article in English | MEDLINE | ID: mdl-27020329

ABSTRACT

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.


Subject(s)
Alzheimer Disease/prevention & control , Amyloid beta-Peptides/drug effects , Rifampin/pharmacology , Rifampin/therapeutic use , Tauopathies/prevention & control , tau Proteins/drug effects , Alzheimer Disease/complications , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Caspase 3/metabolism , Cells, Cultured , Cytochromes c/metabolism , Dose-Response Relationship, Drug , Female , Hippocampus/metabolism , Maze Learning/drug effects , Memory Disorders/complications , Memory Disorders/drug therapy , Mice , Mice, Transgenic , Microglia/drug effects , Microtubule-Associated Proteins/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Phosphorylation/drug effects , Sequestosome-1 Protein/metabolism , Synapses/drug effects , Synucleins/drug effects , Synucleins/metabolism , Tauopathies/complications , Tauopathies/metabolism , tau Proteins/metabolism
5.
Am J Pathol ; 183(1): 211-25, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23680655

ABSTRACT

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.


Subject(s)
Exons , Frontotemporal Dementia/genetics , Introns , Mutation , RNA Splicing , Tauopathies/genetics , tau Proteins/genetics , Animals , Blotting, Western , Frontotemporal Dementia/pathology , Frontotemporal Dementia/physiopathology , Genetic Markers , Male , Mice , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Tauopathies/pathology , Tauopathies/physiopathology
6.
Acta Neuropathol ; 127(5): 685-98, 2014 May.
Article in English | MEDLINE | ID: mdl-24531886

ABSTRACT

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.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Neurofibrillary Tangles/metabolism , tau Proteins/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Brain/pathology , Disease Progression , Hippocampus/metabolism , Hippocampus/pathology , Humans , Memory Disorders/metabolism , Memory Disorders/pathology , Mice, Transgenic , Mutation , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurofibrillary Tangles/pathology , Neurons/metabolism , Neurons/pathology , Phosphorylation , Synapses/metabolism , Synapses/pathology , Tauopathies/metabolism , Tauopathies/pathology , Time Factors , tau Proteins/genetics
7.
Geroscience ; 46(2): 1971-1987, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37783918

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Mice , Animals , Humans , Neurodegenerative Diseases/prevention & control , Hawaii , Powders , Alzheimer Disease/prevention & control , Alzheimer Disease/metabolism , Neurons/metabolism , Water
8.
Nutrients ; 16(11)2024 May 23.
Article in English | MEDLINE | ID: mdl-38892521

ABSTRACT

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.


Subject(s)
Acorus , Dementia , Plant Extracts , Plant Leaves , Animals , Plant Leaves/chemistry , Acorus/chemistry , Mice , Plant Extracts/pharmacology , Dementia/prevention & control , Disease Models, Animal , Cognition/drug effects , Amyloid beta-Peptides/metabolism , Male , Alzheimer Disease/prevention & control , tau Proteins/metabolism
9.
Brain Res ; 1838: 148987, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38718851

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Memory Disorders , Spatial Learning , Animals , Male , Mice , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/drug effects , Maze Learning/drug effects , Maze Learning/physiology , Memory Disorders/drug therapy , Memory Disorders/metabolism , Mice, Transgenic , Microtubules/metabolism , Microtubules/drug effects , Spatial Learning/drug effects , tau Proteins/metabolism , Dynamin I/metabolism
10.
J Control Release ; 367: 515-521, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38237689

ABSTRACT

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.


Subject(s)
Magnetite Nanoparticles , Nanoparticles , Administration, Intranasal , Magnetite Nanoparticles/chemistry , Gold , Brain/diagnostic imaging , Nanoparticles/chemistry , Magnetic Phenomena , Particle Size , Drug Delivery Systems
11.
J Neurosci Res ; 91(12): 1541-50, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24105984

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Cholesterol/metabolism , Mutation , Protein Transport/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , HEK293 Cells , Humans , Immunohistochemistry , Intracellular Space/metabolism , Transfection
12.
Biomedicines ; 10(2)2022 Jan 27.
Article in English | MEDLINE | ID: mdl-35203506

ABSTRACT

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.

13.
Biomedicines ; 10(5)2022 May 06.
Article in English | MEDLINE | ID: mdl-35625816

ABSTRACT

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.

14.
J Neurosci ; 30(14): 4845-56, 2010 Apr 07.
Article in English | MEDLINE | ID: mdl-20371804

ABSTRACT

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.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/physiology , Neuroglia/metabolism , Neurons/metabolism , Synapses/metabolism , tau Proteins/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Animals , Cell Death/genetics , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Neuroglia/pathology , Neurons/pathology , Phosphorylation/genetics , Synapses/pathology , tau Proteins/genetics
15.
J Neurosci Res ; 89(7): 1031-42, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21488093

ABSTRACT

Intraneuronal accumulation of amyloid ß (Aß) is an early pathological change in Alzheimer's disease. Previously, we showed that the E693Δ mutation (referred to as the "Osaka" mutation) of amyloid precursor protein (APP) caused intracellular accumulation of Aß oligomers and apoptosis in transfected COS-7 cells. We also showed that transgenic mice expressing APP(E693Δ) (APP(OSK) ) displayed both an age-dependent accumulation of intraneuronal Aß oligomers from 8 months of age and apparent neuronal loss in the hippocampus at 24 months of age. These findings indicate that intraneuronal Aß oligomers cause cell death, but the mechanism of this process remains unclear. Accordingly, here we investigated the subcellular localization and toxicity of intraneuronal Aß oligomers in APP(OSK) -transgenic mice. We found Aß oligomer accumulation in the endoplasmic reticulum (ER), endosomes/lysosomes, and mitochondria in hippocampal neurons of 22-month-old mice. We also detected up-regulation of Grp78 and HRD1 (an E3 ubiquitin ligase), leakage of cathepsin D from endosomes/lysosomes into cytoplasm, cytochrome c release from mitochondria, and activation of caspase-3 in the hippocampi of 18-month-old mice. Collectively, our findings suggest that intraneuronal Aß oligomers cause cell death by inducing ER stress, endosomal/lysosomal leakage, and mitochondrial dysfunction in vivo. © 2011 Wiley-Liss, Inc.


Subject(s)
Amyloid beta-Peptides/toxicity , Endoplasmic Reticulum/pathology , Hippocampus/pathology , Mitochondria/pathology , Nerve Degeneration/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Animals , Apoptosis Regulatory Proteins/metabolism , COS Cells , Cell Death/physiology , Chlorocebus aethiops , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Female , Hippocampus/metabolism , Humans , Male , Mice , Mice, Transgenic , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Nerve Degeneration/metabolism , Stress, Physiological/genetics
16.
Front Neurosci ; 15: 763476, 2021.
Article in English | MEDLINE | ID: mdl-34966254

ABSTRACT

Amyloidogenic protein oligomers are thought to play an important role in the pathogenesis of neurodegenerative dementia, including Alzheimer's disease, frontotemporal dementia, and dementia with Lewy bodies. Previously we demonstrated that oral or intranasal rifampicin improved the cognition of APP-, tau-, and α-synuclein-transgenic mice by reducing the amount of Aß, tau, and α-synuclein oligomers in the brain. In the present study, to explore more effective and safer medications for dementia, we tested the drug combination of rifampicin and resveratrol, which is a multifunctional natural polyphenol with the potential to antagonize the adverse effects of rifampicin. The mixture was intranasally administered to APP-, tau-, and α-synuclein-transgenic mice, and their memory and oligomer-related pathologies were evaluated. Compared with rifampicin and resveratrol alone, the combinatorial medicine significantly improved mouse cognition, reduced amyloid oligomer accumulation, and recovered synaptophysin levels in the hippocampus. The plasma levels of liver enzymes, which reflect hepatic injury and normally increase by rifampicin treatment, remained normal by the combination treatment. Notably, resveratrol alone and the combinatorial medicine, but not rifampicin alone, enhanced the levels of brain-derived neurotrophic factor (BDNF) and its precursor, pro-BDNF, in the hippocampus. Furthermore, the combination showed a synergistic effect in ameliorating mouse cognition. These results show the advantages of this combinatorial medicine with regards to safety and effectiveness over single-drug rifampicin. Our findings may provide a feasible means for the prevention of neurodegenerative dementia that targets toxic oligomers.

17.
J Neurosci Res ; 88(9): 1985-94, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20155813

ABSTRACT

Amyloid beta (Abeta) is a key molecule in the pathogenesis of Alzheimer's disease, but its physiological function remains unclear. Abeta is produced from amyloid precursor protein (APP) by beta- and gamma-secretases, which is enhanced by high levels of cellular cholesterol, so cholesterol is a risk factor for Alzheimer's disease. This linkage led us to hypothesize that Abeta is produced to regulate cellular cholesterol levels in response to high-cholesterol stimulation. Here we show that Abeta production caused a reduction of cellular cholesterol levels in transfected HEK293 cells and neuronal IMR-32 and Neuro2a cells, which was accompanied by an increase in efflux of cholesterol from cells. Fractionation of the culture media by ultracentrifugation and subsequent immunoelectron microscopic observation revealed that Abeta assembled high-density lipoprotein-like particles with cellular cholesterol during its secretion. This assembly was mediated by the ATP-binding cassette transporter A1. APP transgenic and knockout mice exhibited lower and higher levels of cellular cholesterol in their brains, suggesting that Abeta-mediated regulation of cellular cholesterol is physiological. Furthermore, we found that, when injected into mouse cerebral ventricle, reconstituted lipoproteins with Abeta were excreted into the peripheral tissues more efficiently than those without Abeta. This result suggests that Abeta mediates cholesterol transport from the brain to the circulation. We propose, based on these findings, a novel, apolipoprotein-like function for Abeta that is involved in maintenance of cellular and cerebral cholesterol homeostasis.


Subject(s)
Amyloid beta-Peptides/metabolism , Cholesterol/metabolism , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Cell Line , Cell Line, Tumor , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Immunoelectron , Neurons/metabolism , Neurons/ultrastructure , Protease Nexins , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism
18.
R Soc Open Sci ; 6(5): 181649, 2019 May.
Article in English | MEDLINE | ID: mdl-31218023

ABSTRACT

The fabrication conditions of bone-haemostasis sheet were examined by using (i) phosphoryl oligosaccharides of calcium (POs-Ca), sugar-containing hydroxyapatite (s-Ca10(PO4)6(OH)2: s-HAp) derived from POs-Ca and (ii) natural plant-derived polymers (locust bean gum (LBG), guar gum (GG) and alginate (AG)). The sol, which had been prepared by dissolving 2 mass% LBG/GG and 2 mass% AG into 200 cm3 deionized water and then by agitating at the speed of 20 000 r.p.m., was immersed into 3 mass% POs-Ca solution at room temperature for 24 h; it was hydrothermally treated at 100°C for 5 h, and then freeze-dried at -50°C for 24 h to form porous composite sheet. The microscopic observation showed that the pore sizes were controlled in the range of 5-100 µm by the optimization of LBG/GG ratio. The composite sheet showed the noted uptake of simulated body fluid (1426%) at 37.0°C and also the human blood. Thus, the porous composite sheet was found to be a promising candidate of the bone haemostasis, on the basis of the data of haemostasis, uptake ability of SBF and solubility in acetic acid-sodium acetate buffer solution.

19.
Alzheimers Dement (N Y) ; 4: 304-313, 2018.
Article in English | MEDLINE | ID: mdl-30094330

ABSTRACT

INTRODUCTION: Oral rifampicin has been shown to significantly reduce amyloid ß (Aß) and tau pathologies in mice. However, it shows occasional adverse effects such as liver injury in humans, making its use difficult for a long period. METHODS: To explore safer rifampicin treatment, APPOSK mice, a model of Alzheimer's disease, were treated with rifampicin for 1 month via oral, intranasal, and subcutaneous administration, and its therapeutic efficacy and safety were compared. RESULTS: Intranasal or subcutaneous administration of rifampicin improved memory more effectively than oral administration. The improvement of memory was accompanied with the reduction of neuropathologies, including Aß oligomer accumulation, tau abnormal phosphorylation, and synapse loss. Serum levels of a liver enzyme significantly rose only by oral administration. Pharmacokinetic study revealed that the level of rifampicin in the brain was highest with intranasal administration. DISCUSSION: Considering its easiness and noninvasiveness, intranasal administration would be the best way for long-term dosing of rifampicin.

20.
Acta Neuropathol Commun ; 5(1): 59, 2017 07 31.
Article in English | MEDLINE | ID: mdl-28760161

ABSTRACT

The E693Δ (Osaka) mutation in APP is linked to familial Alzheimer's disease. While this mutation accelerates amyloid ß (Aß) oligomerization, only patient homozygotes suffer from dementia, implying that this mutation is recessive and causes loss-of-function of amyloid precursor protein (APP). To investigate the recessive trait, we generated a new mouse model by knocking-in the Osaka mutation into endogenous mouse APP. The produced homozygous, heterozygous, and non-knockin littermates were compared for memory, neuropathology, and synaptic plasticity. Homozygotes showed memory impairment at 4 months, whereas heterozygotes did not, even at 8 months. Immunohistochemical and biochemical analyses revealed that only homozygotes displayed intraneuronal accumulation of Aß oligomers at 8 months, followed by abnormal tau phosphorylation, synapse loss, glial activation, and neuron loss. These pathologies were not observed at younger ages, suggesting that a certain mechanism other than Aß accumulation underlies the memory disturbance at 4 months. For the electrophysiology studies at 4 months, high-frequency stimulation evoked long-term potentiation in all mice in the presence of picrotoxin, but in the absence of picrotoxin, such potentiation was observed only in homozygotes, suggesting their GABAergic deficit. In support of this, the levels of GABA-related proteins and the number of dentate GABAergic interneurons were decreased in 4-month-old homozygotes. Since APP has been shown to play a role in dentate GABAergic synapse formation, the observed GABAergic depletion is likely associated with an impairment of the APP function presumably caused by the Osaka mutation. Oral administration of diazepam to homozygotes from 6 months improved memory at 8 months, and furthermore, prevented Aß oligomer accumulation, indicating that GABAergic deficiency is a cause of memory impairment and also a driving force of Aß accumulation. Our findings suggest that the Osaka mutation causes loss of APP function, leading to GABAergic depletion and memory disorder when wild-type APP is absent, providing a mechanism of the recessive heredity.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Disease Models, Animal , gamma-Aminobutyric Acid/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Diazepam/pharmacology , GABA Modulators/pharmacology , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Gene Knock-In Techniques , Genes, Recessive , Genetic Predisposition to Disease , Humans , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Mutation , Spatial Memory/drug effects , Spatial Memory/physiology , Tissue Culture Techniques , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL