Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Nat Commun ; 6: 10216, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26671725

ABSTRACT

Neurofibrillary tangles, composed of hyperphosphorylated tau fibrils, are a pathological hallmark of Alzheimer's disease; the neurofibrillary tangle load correlates strongly with clinical progression of the disease. A growing body of evidence indicates that tau oligomer formation precedes the appearance of neurofibrillary tangles and contributes to neuronal loss. Here we show that tau oligomer formation can be inhibited by compounds whose chemical backbone includes 1,2-dihydroxybenzene. Specifically, we demonstrate that 1,2-dihydroxybenzene-containing compounds bind to and cap cysteine residues of tau and prevent its aggregation by hindering interactions between tau molecules. Further, we show that orally administered DL-isoproterenol, an adrenergic receptor agonist whose skeleton includes 1,2-dihydroxybenzene and which penetrates the brain, reduces the levels of detergent-insoluble tau, neuronal loss and reverses neurofibrillary tangle-associated brain dysfunction. Thus, compounds that target the cysteine residues of tau may prove useful in halting the progression of Alzheimer's disease and other tauopathies.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Alzheimer Disease/metabolism , Catechols/pharmacology , Cysteine/drug effects , Isoproterenol/pharmacology , Neurofibrillary Tangles/drug effects , Neurons/drug effects , tau Proteins/drug effects , Adrenergic beta-Agonists/chemistry , Animals , Behavior, Animal/drug effects , Blotting, Western , Brain/drug effects , Brain/metabolism , Catechols/chemistry , Catechols/metabolism , Cell Line, Tumor , Cysteine/metabolism , Disease Models, Animal , Disease Progression , Drug Evaluation, Preclinical , Isoproterenol/chemistry , Mice , Mice, Transgenic , Neurofibrillary Tangles/metabolism , Neurons/pathology , Polymerization , tau Proteins/genetics , tau Proteins/metabolism
2.
Sci Rep ; 5: 10821, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-26027742

ABSTRACT

Methylene blue (MB) inhibits the aggregation of tau, a main constituent of neurofibrillary tangles. However, MB's mode of action in vivo is not fully understood. MB treatment reduced the amount of sarkosyl-insoluble tau in Drosophila that express human wild-type tau. MB concurrently ameliorated the climbing deficits of transgenic tau flies to a limited extent and diminished the climbing activity of wild-type flies. MB also decreased the survival rate of wild-type flies. Based on its photosensitive efficacies, we surmised that singlet oxygen generated through MB under light might contribute to both the beneficial and toxic effects of MB in vivo. We identified rose bengal (RB) that suppressed tau accumulation and ameliorated the behavioral deficits to a lesser extent than MB. Unlike MB, RB did not reduce the survival rate of flies. Our findings indicate that singlet oxygen generators with little toxicity may be suitable drug candidates for treating tauopathies.


Subject(s)
Singlet Oxygen/metabolism , Tauopathies/metabolism , tau Proteins/metabolism , Animals , Animals, Genetically Modified , Autophagy , Behavior, Animal , Disease Models, Animal , Drosophila , Female , Humans , Male , Methylene Blue/pharmacology , Oxidative Stress , Solubility , Tauopathies/drug therapy , tau Proteins/chemistry
3.
Small ; 8(23): 3631-9, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-22915547

ABSTRACT

Amyloids are pathogenic hallmarks in many neurodegenerative diseases such as amyloid-ß (Aß) fibrils in Alzheimer's disease (AD). Here, the effect of gold nanoparticles (AuNPs) on amyloids is examined using Aß as a model system. It is found that bare AuNPs inhibited Aß fibrillization to form fragmented fibrils and spherical oligomers. Adding bare AuNPs to preformed Aß fibrils results in ragged species where AuNPs bind preferentially to fibrils. Similar results are demonstrated with carboxyl- but not amine-conjugated AuNPs. Co-incubation of negatively charged AuNPs with Aß relieved Aß toxicity to neuroblastoma. Overall, it is demonstrated that AuNPs possessing negative surface potential serve as nano-chaperones to inhibit and redirect Aß fibrillization, which could contribute to applications for AD.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid/chemistry , Gold/chemistry , Metal Nanoparticles/chemistry , Metal Nanoparticles/toxicity , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid/antagonists & inhibitors , Amyloid/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Cell Survival , Humans , Microscopy, Electron, Transmission , Neuroblastoma/metabolism , Neuroblastoma/pathology , Tumor Cells, Cultured
4.
J Biol Chem ; 287(18): 14631-43, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22393064

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides/metabolism , Antioxidants/pharmacology , Cinnamates/pharmacology , Depsides/pharmacology , Flavonoids/pharmacology , Protein Multimerization/drug effects , Synapses/metabolism , Animals , HEK293 Cells , Humans , Mice , Synapses/pathology , Rosmarinic Acid
5.
Aging Cell ; 11(1): 51-62, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21981382

ABSTRACT

Oxidative stress is considered to promote aging and age-related disorders such as tauopathy. Although recent reports suggest that oxidative stress under certain conditions possesses anti-aging properties, no such conditions have been reported to ameliorate protein-misfolding diseases. Here, we used neuronal and murine models that overexpress human tau to demonstrate that mild oxidative stress generated by alloxan suppresses several phenotypes of tauopathy. Alloxan treatment reduced HSP90 levels and promoted proteasomal degradation of tau, c-Jun N-amino terminal kinase, and histone deacetylase (HDAC) 6. Moreover, reduced soluble tau (phosphorylated tau) levels suppressed the formation of insoluble tau in tau transgenic mice, while reduced HDAC6 levels contributed to microtubule stability by increasing tubulin acetylation. Age-dependent decreases in HDAC2 and phospho-tau levels correlated with spatial memory enhancement in alloxan-injected tau mice. These results suggest that mild oxidative stress, through adaptive stress responses, operates counteractively against some of the tauopathy phenotypes.


Subject(s)
Aging/psychology , Alloxan/administration & dosage , Neurons/metabolism , Oxidative Stress/physiology , Tauopathies/metabolism , tau Proteins/metabolism , Acetylation , Adaptation, Physiological/drug effects , Aging/drug effects , Alloxan/therapeutic use , Animals , Disease Models, Animal , Female , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Histone Deacetylase 6 , Histone Deacetylases/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Maze Learning/drug effects , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/pathology , Phenotype , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Tauopathies/drug therapy , Tauopathies/pathology , Tubulin/metabolism , tau Proteins/genetics
6.
J Alzheimers Dis ; 27(2): 271-80, 2011.
Article in English | MEDLINE | ID: mdl-21811022

ABSTRACT

Inhibition of amyloid-ß (Aß) aggregation is an attractive therapeutic strategy for treatment of Alzheimer's disease (AD). We previously reported that vitamin A and ß-carotene inhibit fibrillation of Aß40 and Aß42 (Ono et al, 2004, Exp Neurol). In this study, we firstly examined the effects of vitamin A (retinoic acid, retinol, and retinal), ß-carotene, vitamin B2, vitamin B6, vitamin C, vitamin E, coenzyme Q10, and α-lipoic acid on oligomerization of Aß40 and Aß42 in vitro; vitamin A and ß-carotene dose-dependently inhibited oligomerization of Aß40 and Aß42. Furthermore, retinoic acid decreased cellular toxicity by inhibition of Aß42 oligomerization. Second, we analyzed how vitamin A inhibits Aß aggregation by using fluorescence spectroscopy and thioflavin T assay with two Aß fragments, Aß1-16 and Aß25-35. A fluorescence peak of retinoic acid was greatly restrained in the presence of Aß25-35, and retinoic acid inhibited aggregation of Aß25-35, but not of Aß1-16, which suggest the specific binding of retinoic acid to the C-terminal portion of Aß. Thus, vitamin A and ß-carotene might be key molecules for prevention of AD.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Peptide Fragments/antagonists & inhibitors , Vitamin A/pharmacology , Biopolymers/antagonists & inhibitors , Biopolymers/metabolism , Cell Survival/drug effects , Cell Survival/physiology , HEK293 Cells , Humans , Peptide Fragments/metabolism
8.
J Biol Chem ; 285(49): 38692-9, 2010 Dec 03.
Article in English | MEDLINE | ID: mdl-20921222

ABSTRACT

Neurofibrillary tangles (NFTs), which consist of highly phosphorylated tau, are hallmarks of neurodegenerative diseases including Alzheimer disease (AD). In neurodegenerative diseases, neuronal dysfunction due to neuronal loss and synaptic loss accompanies NFT formation, suggesting that a process associated with NFT formation may be involved in neuronal dysfunction. To clarify the relationship between the tau aggregation process and synapse and neuronal loss, we compared two lines of mice expressing human tau with or without an aggregation-prone P301L mutation. P301L tau transgenic (Tg) mice exhibited neuronal loss and produced sarcosyl-insoluble tau in old age but did not exhibit synaptic loss and memory impairment. By contrast, wild-type tau Tg mice neither exhibited neuronal loss nor produced sarcosyl-insoluble tau but did exhibit synaptic loss and memory impairment. Moreover, P301L tau was less phosphorylated than wild-type tau, suggesting that the tau phosphorylation state is involved in synaptic loss, whereas the tau aggregation state is involved in neuronal loss. Finally, increasing concentrations of insoluble tau aggregates leads to the formation of fibrillar tau, which causes NFTs to form.


Subject(s)
Alzheimer Disease/metabolism , Detergents/chemistry , Mutation, Missense , Neurofibrillary Tangles/metabolism , Neurons/metabolism , tau Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amino Acid Substitution , Animals , Humans , Mice , Mice, Transgenic , Neurofibrillary Tangles/genetics , Neurofibrillary Tangles/pathology , Neurons/pathology , Phosphorylation/genetics , tau Proteins/genetics
9.
PLoS One ; 3(10): e3540, 2008.
Article in English | MEDLINE | ID: mdl-18958152

ABSTRACT

Activation of GSK-3beta is presumed to be involved in various neurodegenerative diseases, including Alzheimer's disease (AD), which is characterized by memory disturbances during early stages of the disease. The normal function of GSK-3beta in adult brain is not well understood. Here, we analyzed the ability of heterozygote GSK-3beta knockout (GSK+/-) mice to form memories. In the Morris water maze (MWM), learning and memory performance of GSK+/- mice was no different from that of wild-type (WT) mice for the first 3 days of training. With continued learning on subsequent days, however, retrograde amnesia was induced in GSK+/- mice, suggesting that GSK+/- mice might be impaired in their ability to form long-term memories. In contextual fear conditioning (CFC), context memory was normally consolidated in GSK+/- mice, but once the original memory was reactivated, they showed reduced freezing, suggesting that GSK+/- mice had impaired memory reconsolidation. Biochemical analysis showed that GSK-3beta was activated after memory reactivation in WT mice. Intraperitoneal injection of a GSK-3 inhibitor before memory reactivation impaired memory reconsolidation in WT mice. These results suggest that memory reconsolidation requires activation of GSK-3beta in the adult brain.


Subject(s)
Brain/physiopathology , Glycogen Synthase Kinase 3/physiology , Memory Disorders/genetics , Memory Disorders/rehabilitation , Memory/physiology , Amnesia, Retrograde/genetics , Animals , Brain/metabolism , Enzyme Activation/genetics , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Heterozygote , Male , Maze Learning/physiology , Memory Disorders/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Physical Conditioning, Animal/methods
10.
PLoS One ; 3(9): e3235, 2008 Sep 18.
Article in English | MEDLINE | ID: mdl-18800165

ABSTRACT

Recent in vitro and in vivo studies suggest that destabilized proteins with defective folding induce aggregation and toxicity in protein-misfolding diseases. One such unstable protein state is called amyloid oligomer, a precursor of fully aggregated forms of amyloid. Detection of various amyloid oligomers with A11, an anti-amyloid oligomer conformation-specific antibody, revealed that the amyloid oligomer represents a generic conformation and suggested that toxic beta-aggregation processes possess a common mechanism. By using A11 antibody as a probe in combination with mass spectrometric analysis, we identified GroEL in bacterial lysates as a protein that may potentially have an amyloid oligomer conformation. Surprisingly, A11 reacted not only with purified GroEL but also with several purified heat shock proteins, including human Hsp27, 40, 70, 90; yeast Hsp104; and bovine Hsc70. The native folds of A11-reactive proteins in purified samples were characterized by their anti-beta-aggregation activity in terms of both functionality and in contrast to the beta-aggregation promoting activity of misfolded pathogenic amyloid oligomers. The conformation-dependent binding of A11 with natively folded Hsp27 was supported by the concurrent loss of A11 reactivity and anti-beta-aggregation activity of heat-treated Hsp27 samples. Moreover, we observed consistent anti-beta-aggregation activity not only by chaperones containing an amyloid oligomer conformation but also by several A11-immunoreactive non-chaperone proteins. From these results, we suggest that the amyloid oligomer conformation is present in a group of natively folded proteins. The inhibitory effects of A11 antibody on both GroEL/ES-assisted luciferase refolding and Hsp70-mediated decelerated nucleation of Abeta aggregation suggested that the A11-binding sites on these chaperones might be functionally important. Finally, we employed a computational approach to uncover possible A11-binding sites on these targets. Since the beta-sheet edge was a common structural motif having the most similar physicochemical properties in the A11-reactive proteins we analyzed, we propose that the beta-sheet edge in some natively folded amyloid oligomers is designed positively to prevent beta aggregation.


Subject(s)
Amyloid/chemistry , Animals , Binding Sites , Cattle , Chaperonin 60/chemistry , Epitopes , HSP27 Heat-Shock Proteins/chemistry , Heat-Shock Proteins , Humans , Models, Statistical , Molecular Chaperones , Molecular Conformation , Protein Conformation , Protein Denaturation , Protein Folding , Protein Structure, Secondary
11.
PLoS One ; 3(8): e3029, 2008 Aug 21.
Article in English | MEDLINE | ID: mdl-18716656

ABSTRACT

Advanced age and mutations in the genes encoding amyloid precursor protein (APP) and presenilin (PS1) are two serious risk factors for Alzheimer's disease (AD). Finding common pathogenic changes originating from these risks may lead to a new therapeutic strategy. We observed a decline in memory performance and reduction in hippocampal long-term potentiation (LTP) in both mature adult (9-15 months) transgenic APP/PS1 mice and old (19-25 months) non-transgenic (nonTg) mice. By contrast, in the presence of bicuculline, a GABA(A) receptor antagonist, LTP in adult APP/PS1 mice and old nonTg mice was larger than that in adult nonTg mice. The increased LTP levels in bicuculline-treated slices suggested that GABA(A) receptor-mediated inhibition in adult APP/PS1 and old nonTg mice was upregulated. Assuming that enhanced inhibition of LTP mediates memory decline in APP/PS1 mice, we rescued memory deficits in adult APP/PS1 mice by treating them with another GABA(A) receptor antagonist, picrotoxin (PTX), at a non-epileptic dose for 10 days. Among the saline vehicle-treated groups, substantially higher levels of synaptic proteins such as GABA(A) receptor alpha1 subunit, PSD95, and NR2B were observed in APP/PS1 mice than in nonTg control mice. This difference was insignificant among PTX-treated groups, suggesting that memory decline in APP/PS1 mice may result from changes in synaptic protein levels through homeostatic mechanisms. Several independent studies reported previously in aged rodents both an increased level of GABA(A) receptor alpha1 subunit and improvement of cognitive functions by long term GABA(A) receptor antagonist treatment. Therefore, reduced LTP linked to enhanced GABA(A) receptor-mediated inhibition may be triggered by aging and may be accelerated by familial AD-linked gene products like Abeta and mutant PS1, leading to cognitive decline that is pharmacologically treatable at least at this stage of disease progression in mice.


Subject(s)
Aging/physiology , Amyloid beta-Protein Precursor/metabolism , GABA Antagonists/therapeutic use , Memory Disorders/prevention & control , Memory Disorders/physiopathology , Picrotoxin/therapeutic use , Presenilin-1/metabolism , Receptors, GABA-A/physiology , Amyloid beta-Protein Precursor/drug effects , Amyloid beta-Protein Precursor/genetics , Animals , Cognition Disorders/physiopathology , Cognition Disorders/prevention & control , Humans , Learning/drug effects , Learning/physiology , Long-Term Potentiation/drug effects , Long-Term Potentiation/genetics , Long-Term Potentiation/physiology , Maze Learning/drug effects , Mice , Mice, Transgenic , Presenilin-1/drug effects , Receptors, GABA-A/drug effects
12.
EMBO J ; 26(24): 5143-52, 2007 Dec 12.
Article in English | MEDLINE | ID: mdl-18007595

ABSTRACT

To investigate how tau affects neuronal function during neurofibrillary tangle (NFT) formation, we examined the behavior, neural activity, and neuropathology of mice expressing wild-type human tau. Here, we demonstrate that aged (>20 months old) mice display impaired place learning and memory, even though they do not form NFTs or display neuronal loss. However, soluble hyperphosphorylated tau and synapse loss were found in the same regions. Mn-enhanced MRI showed that the activity of the parahippocampal area is strongly correlated with the decline of memory as assessed by the Morris water maze. Taken together, the accumulation of hyperphosphorylated tau and synapse loss in aged mice, leading to inhibition of neural activity in parahippocampal areas, including the entorhinal cortex, may underlie place learning impairment. Thus, the accumulation of hyperphosphorylated tau that occurs before NFT formation in entorhinal cortex may contribute to the memory problems seen in Alzheimer's disease (AD).


Subject(s)
Aging/physiology , Entorhinal Cortex/metabolism , Maze Learning/physiology , Memory/physiology , tau Proteins/metabolism , Animals , Behavior, Animal/physiology , Brain Mapping , Entorhinal Cortex/cytology , Humans , Magnetic Resonance Imaging , Mice , Mice, Transgenic , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Phosphorylation , Synapses/metabolism , Synapses/pathology , tau Proteins/genetics
13.
Neuromolecular Med ; 9(3): 270-5, 2007.
Article in English | MEDLINE | ID: mdl-17914185

ABSTRACT

Degenerative diseases such as Alzheimer's, Parkinson's, and Huntington's diseases are believed to be causally related to the accumulation of amyloid oligomers that exhibit a common structure and may be toxic by a common mechanism involving permeabilization of membranes. We discovered that amyloid oligomers and the pore-forming bacterial toxin, alpha-hemolysin (alpha HL), as well as human perforin from cytotoxic T lymphocytes, share a structural and functional homology at the level of their common reactivity with a conformation-dependent antibody that is specific for amyloid oligomers, A11. The alpha HL oligomeric pores and partially folded alpha HL protomer, but not the monomer alpha HL precursor reacts with A11 antibody. A11 antibody inhibits the hemolytic activity of alpha HL, indicating that the structural homology is functionally significant. Perforin oligomers were also recognized by A11. Amyloidogenic properties of alpha HL and perforin were confirmed spectroscopically and morphologically. These results indicate that pore forming proteins (PFP) and amyloid oligomers share structural homology and suggest that PFPs and amyloid oligomers share the same mechanism of membrane permeabilization.


Subject(s)
Amyloid beta-Peptides/chemistry , Bacterial Toxins/chemistry , Escherichia coli Proteins/chemistry , Hemolysin Proteins/chemistry , Peptide Fragments/chemistry , Perforin/chemistry , Amino Acid Sequence , Antibodies/immunology , Bacterial Toxins/isolation & purification , Deoxycholic Acid/chemistry , Escherichia coli Proteins/isolation & purification , Hemolysin Proteins/isolation & purification , Humans , Protein Structure, Secondary , Structure-Activity Relationship , T-Lymphocytes, Cytotoxic/chemistry
14.
Biochemistry ; 46(34): 9805-12, 2007 Aug 28.
Article in English | MEDLINE | ID: mdl-17676931

ABSTRACT

Amyloid beta (Abeta) toxicity has been hypothesized to initiate the pathogenesis of Alzheimer's disease (AD). The characteristic fibrillar morphology of Abeta-aggregates, that constitute the main components of senile plaque, has long been considered to account for the neurotoxicity. But recent reports argue against a primary role for mature fibrils in AD pathogenesis because of the lack of a robust correlation between the severity of neurological impairment and the extent of amyloid deposition. Toxicity from the soluble prefibrillar intermediate entity of aggregates often called oligomer has recently proposed a plausible explanation for this inconsistency. An alternative explanation is based on the observation that certain amyloid fibril morphologies are more toxic than others, indicating that not all amyloid fibrils are equally toxic. Here, we report that it is not only the beta-sheeted fibrillar structure but also the surface physicochemical composition that affects the toxicity of Abeta fibrils. For the first time, colloidal gold was used to visualize by electron microscopy positive-charge clusters on Abeta fibrils. Chemical modifications as well as point-mutated Abeta synthesis techniques were applied to change the surface structures of Abeta and to show how local structure affects surface properties that are responsible for electrostatic and hydrophobic interactions with cells. We also report that covering the surface of Abeta fibers with myelin basic protein, which has surface properties contrary to those of Abeta, suppresses Abeta toxicity. On the basis of these results, we propose that the surface structure of Abeta fibrils plays an important role in Abeta toxicity.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/toxicity , Cell Survival , Peptide Fragments/chemistry , Peptide Fragments/toxicity , Plaque, Amyloid/chemistry , Acetylation , Animals , Benzothiazoles , Carrier Proteins/metabolism , Cattle , Cells, Cultured , Fluorescence , Humans , Kidney/metabolism , Maltose-Binding Proteins , Microscopy, Electron, Transmission , Thiazoles/metabolism , Tryptophan/chemistry
15.
Biochemistry ; 46(12): 3856-61, 2007 Mar 27.
Article in English | MEDLINE | ID: mdl-17338548

ABSTRACT

Neurofibrillary tangles (NFTs) are pathological hallmarks of several neurodegenerative disorders, including Alzheimer's disease (AD). NFTs are composed of microtubule-binding protein tau, which assembles to form paired helical filaments (PHFs) and straight filaments. Here we show by atomic force microscopy that AD brain tissue and in vitro tau form granular and fibrillar tau aggregates. CD spectral analysis and immunostaining with conformation-dependent antibodies indicated that tau may undergo conformational changes during fibril formation. Enriched granules generated filaments, suggesting that granular tau aggregates may be an intermediate form of tau fibrils. The amount of granular tau aggregates was elevated in prefrontal cortex of Braak stage I cases compared to that of Braak stage 0 cases, suggesting that granular tau aggregation precedes PHF formation. Thus, granular tau aggregates may be a relevant marker for the early diagnosis of tauopathy. Reducing the level of these aggregates may be a promising therapy for tauopathies and for promoting healthy brain aging.


Subject(s)
Neurofibrillary Tangles/chemistry , tau Proteins/chemistry , Aging/genetics , Aging/metabolism , Aging/pathology , Alzheimer Disease/diagnosis , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Biomarkers/chemistry , Biomarkers/metabolism , Brain Chemistry , Circular Dichroism , Frontal Lobe/metabolism , Frontal Lobe/pathology , Humans , Microscopy, Atomic Force , Neurofibrillary Tangles/genetics , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/ultrastructure , tau Proteins/genetics , tau Proteins/metabolism
16.
Exp Neurol ; 189(2): 380-92, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15380488

ABSTRACT

Cerebral deposition of amyloid beta-peptide (Abeta) in the brain is an invariant feature of Alzheimer disease (AD). Plasma or cerebrospinal fluid concentrations of antioxidant vitamins and carotenoids, such as vitamins A, C, E, and beta-carotene, have been reported to be lower in AD patients, and these vitamins clinically have been demonstrated to slow the progression of dementia. In this study, we used fluorescence spectroscopy with thioflavin T (ThT) and electron microscopy to examine the effects of vitamin A (retinol, retinal, and retinoic acid), beta-carotene, and vitamins B2, B6, C, and E on the formation, extension, and destabilization of beta-amyloid fibrils (fAbeta) in vitro. Among them, vitamin A and beta-carotene dose-dependently inhibited formation of fAbeta from fresh Abeta, as well as their extension. Moreover, they dose-dependently destabilized preformed fAbetas. The overall activity of the molecules examined was in the order of retinol = retinal > beta-carotene > retinoic acid. Although the exact mechanisms are still unclear, vitamins A and beta-carotene could be key molecules for the prevention and therapy of AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Neurofibrils/drug effects , Vitamin A/pharmacology , Vitamins/pharmacology , Amyloid beta-Peptides/biosynthesis , Antioxidants/pharmacology , Benzothiazoles , Cell Line , Dose-Response Relationship, Drug , Humans , Kinetics , Microscopy, Electron , Molecular Structure , Neurofibrils/metabolism , Neurofibrils/ultrastructure , Retinaldehyde/pharmacology , Thiazoles , Tretinoin/pharmacology , beta Carotene/pharmacology
17.
J Neurochem ; 87(1): 172-81, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12969264

ABSTRACT

Cerebral deposition of amyloid beta-peptide (Abeta) in the brain is an invariant feature of Alzheimer's disease (AD). A consistent protective effect of wine consumption on AD has been documented by epidemiological studies. In the present study, we used fluorescence spectroscopy with thioflavin T and electron microscopy to examine the effects of wine-related polyphenols (myricetin, morin, quercetin, kaempferol (+)-catechin and (-)-epicatechin) on the formation, extension, and destabilization of beta-amyloid fibrils (fAbeta) at pH 7.5 at 37 degrees C in vitro. All examined polyphenols dose-dependently inhibited formation of fAbeta from fresh Abeta(1-40) and Abeta(1-42), as well as their extension. Moreover, these polyphenols dose-dependently destabilized preformed fAbetas. The overall activity of the molecules examined was in the order of: myricetin = morin = quercetin > kaempferol > (+)-catechin = (-)-epicatechin. The effective concentrations (EC50) of myricetin, morin and quercetin for the formation, extension and destabilization of fAbetas were in the order of 0.1-1 micro m. In cell culture experiments, myricetin-treated fAbeta were suggested to be less toxic than intact fAbeta, as demonstrated by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide assay. Although the mechanisms by which these polyphenols inhibit fAbeta formation from Abeta, and destabilize pre-formed fAbetain vitro are still unclear, polyphenols could be a key molecule for the development of preventives and therapeutics for AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides/chemistry , Phenols/chemistry , Phenols/pharmacology , Polymers/chemistry , Polymers/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/prevention & control , Amyloid beta-Peptides/drug effects , Benzothiazoles , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Flavonoids/chemistry , Flavonoids/pharmacology , Humans , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Kinetics , Microscopy, Electron , Peptide Fragments/chemistry , Quercetin/chemistry , Quercetin/pharmacology , Spectrometry, Fluorescence , Tetrazolium Salts , Thiazoles/chemistry
18.
J Biol Chem ; 278(26): 23648-55, 2003 Jun 27.
Article in English | MEDLINE | ID: mdl-12716908

ABSTRACT

Alzheimer's disease (AD) may be caused by toxic aggregates formed from amyloid-beta (Abeta) peptides. By using Thioflavin T, a dye that specifically binds to beta-sheet structures, we found that highly toxic forms of Abeta-aggregates were formed at the initial stage of fibrillogenesis, which is consistent with recent reports on Abeta oligomers. Formation of such aggregates depends on factors that affect both nucleation and elongation. As reported previously, addition of Abeta42 systematically accelerated the nucleation of Abeta40, most likely because of the extra hydrophobic residues at the C terminus of Abeta42. At Abeta42-increased specific ratio (Abeta40: Abeta42 = 10: 1), on the other hand, not only accelerated nucleation but also induced elongation were observed, suggesting pathogenesis of early-onset AD. Because a larger proportion of Abeta40 than Abeta42 was still required for this phenomenon, we assumed that elongation does not depend only on hydrophobic interactions. Without any change in the C-terminal hydrophobic nature, elongation was effectively induced by mixing wild type Abeta40 with Italian variant Abeta40 (E22K) or Dutch variant (E22Q). We suggest that Abeta peptides in specific compositions that balance hydrophilic and hydrophobic interactions promote the formation of toxic beta-aggregates. These results may introduce a new therapeutic approach through the disruption of this balance.


Subject(s)
Alzheimer Disease/etiology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Benzothiazoles , Cell Line , Dimerization , Genetic Variation , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Static Electricity , Thiazoles
19.
J Neurochem ; 81(3): 434-40, 2002 May.
Article in English | MEDLINE | ID: mdl-12065652

ABSTRACT

Inhibition of the accumulation of amyloid beta-peptide (Abeta) and the formation of beta-amyloid fibrils (fAbeta) from Abeta, as well as the degradation of pre-formed fAbeta in the CNS would be attractive therapeutic objectives for the treatment of Alzheimer's disease (AD). We previously reported that nordihydroguaiaretic acid (NDGA) inhibited fAbeta formation from Abeta(1-40) and Abeta(1-42) dose-dependently in the range of 10-30 micromin vitro. Utilizing fluorescence spectroscopic analysis with thioflavin T and electron microscopic study, we show here that NDGA dose-dependently breaks down fAbeta(1-40) and fAbeta(1-42) within a few hours at pH 7.5 at 37 degrees C. At 4 h, the fluorescence of fAbeta(1-40) and fAbeta(1-42) incubated with 50 microm NDGA was 5% and 10% of the initial fluorescence, respectively. The activity of NDGA to break down these fAbetas was observed even at a low concentration of 0.1 microm. At 1 h, many short, sheared fibrils were observed in the mixture incubated with 50 microm NDGA, and at 4 h, the number of fibrils reduced markedly, and small amorphous aggregates were observed. We next compared the activity of NDGA to break down fAbeta(1-40) and fAbeta(1-42), with other molecules reported to inhibit fAbeta formation from Abeta and/or to degrade pre-formed fAbeta both in vivo and in vitro. At a concentration of 50 microm, the overall activity of the molecules examined in this study was in the order of: NDGA >> rifampicin = tetracycline > poly(vinylsulfonic acid, sodium salt) = 1,3-propanedisulfonic acid, disodium salt > beta-sheet breaker peptide (iAbeta5). In cell culture experiments, fAbeta disrupted by NDGA were less toxic than intact fAbeta, as demonstrated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Although the mechanisms by which NDGA inhibits fAbeta formation from Abeta, as well as breaking down pre-formed fAbetain vitro, are still unclear, NDGA could be a key molecule for the development of therapeutics for AD.


Subject(s)
Amyloid beta-Peptides/chemistry , Masoprocol/chemistry , Peptide Fragments/chemistry , Amyloid beta-Peptides/toxicity , Amyloid beta-Peptides/ultrastructure , Benzothiazoles , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Kidney/cytology , Kidney/drug effects , Macromolecular Substances , Masoprocol/pharmacology , Microscopy, Electron , Peptide Fragments/chemical synthesis , Peptide Fragments/pharmacology , Peptide Fragments/toxicity , Peptide Fragments/ultrastructure , Protein Binding/drug effects , Spectrometry, Fluorescence , Thiazoles
SELECTION OF CITATIONS
SEARCH DETAIL
...