Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Neurosci ; 34(21): 7124-36, 2014 May 21.
Article in English | MEDLINE | ID: mdl-24849348

ABSTRACT

Hyperphosphorylation and accumulation of tau aggregates are prominent features in tauopathies, including Alzheimer's disease, but the impact of loss of tau function on synaptic and cognitive deficits remains poorly understood. We report that old (19-20 months; OKO) but not middle-aged (8-9 months; MKO) tau knock-out mice develop Morris Water Maze (MWM) deficits and loss of hippocampal acetylated α-tubulin and excitatory synaptic proteins. Mild motor deficits and reduction in tyrosine hydroxylase (TH) in the substantia nigra were present by middle age, but did not affect MWM performance, whereas OKO mice showed MWM deficits paralleling hippocampal deficits. Deletion of tau, a microtubule-associated protein (MAP), resulted in increased levels of MAP1A, MAP1B, and MAP2 in MKO, followed by loss of MAP2 and MAP1B in OKO. Hippocampal synaptic deficits in OKO mice were partially corrected with dietary supplementation with docosahexaenoic acid (DHA) and both MWM and synaptic deficits were fully corrected by combining DHA with α-lipoic acid (ALA), which also prevented TH loss. DHA or DHA/ALA restored phosphorylated and total GSK3ß and attenuated hyperactivation of the tau C-Jun N-terminal kinases (JNKs) while increasing MAP1B, dephosphorylated (active) MAP2, and acetylated α-tubulin, suggesting improved microtubule stability and maintenance of active compensatory MAPs. Our results implicate the loss of MAP function in age-associated hippocampal deficits and identify a safe dietary intervention, rescuing both MAP function and TH in OKO mice. Therefore, in addition to microtubule-stabilizing therapeutic drugs, preserving or restoring compensatory MAP function may be a useful new prevention strategy.


Subject(s)
Aging/pathology , Hippocampus/pathology , Maze Learning/physiology , Synapses/metabolism , tau Proteins/deficiency , Aging/drug effects , Aging/genetics , Alzheimer Disease/complications , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Disease Models, Animal , Docosahexaenoic Acids/administration & dosage , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hippocampus/drug effects , Learning Disabilities/diet therapy , Learning Disabilities/etiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Movement Disorders/diet therapy , Movement Disorders/etiology , Psychomotor Performance/physiology , Reaction Time/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology , Synapses/drug effects , Synapses/genetics , Thioctic Acid/administration & dosage
2.
J Biol Chem ; 288(6): 4056-65, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23264626

ABSTRACT

The mechanisms underlying Tau-related synaptic and cognitive deficits and the interrelationships between Tau species, their clearance pathways, and synaptic impairments remain poorly understood. To gain insight into these mechanisms, we examined these interrelationships in aged non-mutant genomic human Tau mice, with established Tau pathology and neuron loss. We also examined how these interrelationships changed with an intervention by feeding mice either a control diet or one containing the brain permeable beta-amyloid and Tau aggregate binding molecule curcumin. Transgene-dependent elevations in soluble and insoluble phospho-Tau monomer and soluble Tau dimers accompanied deficits in behavior, hippocampal excitatory synaptic markers, and molecular chaperones (heat shock proteins (HSPs)) involved in Tau degradation and microtubule stability. In human Tau mice but not control mice, HSP70, HSP70/HSP72, and HSP90 were reduced in membrane-enriched fractions but not in cytosolic fractions. The synaptic proteins PSD95 and NR2B were reduced in dendritic fields and redistributed into perikarya, corresponding to changes observed by immunoblot. Curcumin selectively suppressed levels of soluble Tau dimers, but not of insoluble and monomeric phospho-Tau, while correcting behavioral, synaptic, and HSP deficits. Treatment increased PSD95 co-immunoprecipitating with NR2B and, independent of transgene, increased HSPs implicated in Tau clearance. It elevated HSP90 and HSC70 without increasing HSP mRNAs; that is, without induction of the heat shock response. Instead curcumin differentially impacted HSP90 client kinases, reducing Fyn without reducing Akt. In summary, curcumin reduced soluble Tau and elevated HSPs involved in Tau clearance, showing that even after tangles have formed, Tau-dependent behavioral and synaptic deficits can be corrected.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Behavior, Animal/drug effects , Curcumin/pharmacology , Heat-Shock Proteins/metabolism , Protein Multimerization/drug effects , Synapses/metabolism , Tauopathies/metabolism , tau Proteins/metabolism , Aged , Aged, 80 and over , Animals , Disks Large Homolog 4 Protein , Female , Heat-Shock Proteins/genetics , Hippocampus/metabolism , Hippocampus/pathology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Protein Multimerization/genetics , Proteolysis/drug effects , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Solubility/drug effects , Synapses/genetics , Synapses/pathology , Tauopathies/drug therapy , Tauopathies/genetics , Tauopathies/pathology , tau Proteins/genetics
3.
J Neurosci ; 29(28): 9078-89, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19605645

ABSTRACT

Both insulin resistance (type II diabetes) and beta-amyloid (Abeta) oligomers are implicated in Alzheimer's disease (AD). Here, we investigate the role of Abeta oligomer-induced c-Jun N-terminal kinase (JNK) activation leading to phosphorylation and degradation of the adaptor protein insulin receptor substrate-1 (IRS-1). IRS-1 couples insulin and other trophic factor receptors to downstream kinases and neuroprotective signaling. Increased phospho-IRS-1 is found in AD brain and insulin-resistant tissues from diabetics. Here, we report Abeta oligomers significantly increased active JNK and phosphorylation of IRS-1 (Ser616) and tau (Ser422) in cultured hippocampal neurons, whereas JNK inhibition blocked these responses. The omega-3 fatty acid docosahexaenoic acid (DHA) similarly inhibited JNK and the phosphorylation of IRS-1 and tau in cultured hippocampal neurons. Feeding 3xTg-AD transgenic mice a diet high in saturated and omega-6 fat increased active JNK and phosphorylated IRS-1 and tau. Treatment of the 3xTg-AD mice on high-fat diet with fish oil or curcumin or a combination of both for 4 months reduced phosphorylated JNK, IRS-1, and tau and prevented the degradation of total IRS-1. This was accompanied by improvement in Y-maze performance. Mice fed with fish oil and curcumin for 1 month had more significant effects on Y-maze, and the combination showed more significant inhibition of JNK, IRS-1, and tau phosphorylation. These data indicate JNK mediates Abeta oligomer inactivation of IRS-1 and phospho-tau pathology and that dietary treatment with fish oil/DHA, curcumin, or a combination of both has the potential to improve insulin/trophic signaling and cognitive deficits in AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/pharmacology , Curcumin/pharmacology , Enzyme Inhibitors/pharmacology , Fatty Acids, Omega-3/pharmacology , Insulin Receptor Substrate Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Neurons/drug effects , Peptide Fragments/pharmacology , Signal Transduction/drug effects , tau Proteins/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/diet therapy , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Behavior, Animal , Cells, Cultured , Curcumin/therapeutic use , Disease Models, Animal , Embryo, Mammalian , Enzyme Inhibitors/therapeutic use , Fatty Acids, Omega-3/therapeutic use , Hippocampus/cytology , Humans , Mice , Mice, Transgenic , Middle Aged , Phosphorylation/drug effects , Postmortem Changes , Presenilin-1/genetics , Rats , Rats, Sprague-Dawley , Serine/metabolism
4.
Arch Neurol ; 66(4): 448-57, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19364929

ABSTRACT

BACKGROUND: The sortilin-related receptor SorLA/LR11 (LR11) is a transmembrane neuronal sorting protein that reduces beta-amyloid precursor protein trafficking to secretases, notably BACE1 that generates beta-amyloid, the principal component of senile plaques in Alzheimer disease (AD). LR11 protein is reduced in patients with late-onset AD, and LR11 polymorphisms have been associated with late-onset AD. OBJECTIVE: T o detect soluble LR11 and APP in cerebrospinal fluid (CSF) from patients with AD and control subjects, as (like beta-amyloid precursor protein) LR11 is cleaved near the membrane to release a large N-terminal fragment that is secreted to media from cultured cells. DESIGN: Case-control study. SETTING: Academic research. PARTICIPANTS: Patients with AD and control subjects. MAIN OUTCOME MEASURES: We evaluated CSF LR11, beta-amyloid precursor protein, and apolipoprotein E levels by Western blot in lumbar and postmortem CSF samples. RESULTS: LR11 levels were detectable and stable during 6 months in the CSF of patients with AD. LR11 levels were significantly reduced in lumbar samples from patients with mild to moderate probable AD, as well as in ventricular CSF from patients with autopsy-confirmed AD (predominantly Braak stage III-IV). Bivariate analysis with beta-amyloid 42 and LR11 levels improved diagnostic specificity for AD. Reduced LR11 levels are significantly correlated with soluble beta-amyloid precursor protein but not apolipoprotein E levels. CONCLUSION: Reduced LR11 levels in CSF of patients with AD may have potential as a diagnostic biomarker for patients with LR11 deficits that promote beta-amyloid production or as an index of therapeutic response in late-onset AD.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , Brain/metabolism , LDL-Receptor Related Proteins/cerebrospinal fluid , LDL-Receptor Related Proteins/genetics , Membrane Transport Proteins/cerebrospinal fluid , Membrane Transport Proteins/genetics , Aged , Aged, 80 and over , Alzheimer Disease/diagnosis , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/cerebrospinal fluid , Apolipoproteins E/cerebrospinal fluid , Blotting, Western , Brain/pathology , Case-Control Studies , Cell Line , Gene Expression Regulation/genetics , Humans , Neurofibrillary Tangles/pathology , Peptide Fragments/cerebrospinal fluid , Plaque, Amyloid/pathology , Predictive Value of Tests , Reference Values
5.
Neurobiol Dis ; 33(2): 193-206, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19038340

ABSTRACT

The dysregulation of glycogen synthase kinase-3 (GSK3) has been implicated in Alzheimer disease (AD) pathogenesis and in Abeta-induced neurotoxicity, leading us to investigate it as a therapeutic target in an intracerebroventricular Abeta infusion model. Infusion of a specific GSK3 inhibitor SB216763 (SB) reduced a downstream target, phospho-glycogen synthase 39%, and increased glycogen levels 44%, suggesting effective inhibition of enzyme activity. Compared to vehicle, Abeta increased GSK3 activity, and was associated with elevations in levels of ptau, caspase-3, the tau kinase phospho-c-jun N-terminal kinase (pJNK), neuronal DNA fragmentation, and gliosis. Co-infusion of SB corrected all responses to Abeta infusion except the induction of gliosis and behavioral deficits in the Morris water maze. Nevertheless, SB alone was associated with induction of neurodegenerative markers and behavioral deficits. These data support a role for GSK3 hyperactivation in AD pathogenesis, but emphasize the importance of developing inhibitors that do not suppress constitutive activity.


Subject(s)
Alzheimer Disease/therapy , Enzyme Inhibitors/therapeutic use , Glycogen Synthase Kinase 3/antagonists & inhibitors , Indoles/therapeutic use , Maleimides/therapeutic use , Alzheimer Disease/chemically induced , Amyloid beta-Peptides/pharmacology , Animals , Caspase 3/metabolism , Cells, Cultured , DNA Fragmentation , Disease Models, Animal , Enzyme Inhibitors/adverse effects , Gliosis/chemically induced , Glycogen/metabolism , Glycogen Synthase Kinase 3/metabolism , Hippocampus/drug effects , Hippocampus/physiology , Indoles/adverse effects , JNK Mitogen-Activated Protein Kinases/metabolism , Maleimides/adverse effects , Maze Learning , Nerve Degeneration/drug therapy , Neurons/drug effects , Neurons/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , tau Proteins/metabolism
6.
J Alzheimers Dis ; 15(4): 625-40, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19096161

ABSTRACT

The rat amyloid-beta (Abeta) intracerebroventricular infusion can model aspects of Alzheimer's disease (AD) and has predicted efficacy of therapies such as ibuprofen and curcumin in transgenic mouse models. High density lipoprotein (HDL), a normal plasma carrier of Abeta, is used to attenuate Abeta aggregation within the pump, causing Abeta-dependent toxicity and cognitive deficits within 3 months. Our goal was to identify factors that might accelerate onset of Abeta-dependent deficits to improve efficiency and cost-effectiveness of model. We focused on: 1) optimizing HDL-Abeta preparation for maximal toxicity; 2) evaluating the role of copper, a factor typically in water that can impact oligomer stability; and 3) determining impact of insulin resistance (type II diabetes), a risk factor for AD. In vitro studies were performed to determine doses of copper and methods of Abeta-HDL preparation that maximized toxicity. These preparations when infused resulted in earlier onset of cognitive deficits within 6 weeks post-infusion. Induction of insulin resistance did not exacerbate Abeta-dependent cognitive deficits, but did exacerbate synaptic protein loss. In summary, the newly described in vivo infusion model may be useful cost-effective method for screening for new therapeutic drugs for AD.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Cognition Disorders/genetics , Cognition Disorders/psychology , Copper/toxicity , Insulin Resistance/genetics , Nerve Tissue Proteins/metabolism , Synapses/metabolism , Alzheimer Disease/pathology , Animals , Blotting, Western , Cells, Cultured , Cognition Disorders/pathology , Diet , Fructose/pharmacology , Insulin Resistance/physiology , Maze Learning/physiology , Osmosis , Rats , Rats, Sprague-Dawley
7.
J Biol Chem ; 283(20): 14132-43, 2008 May 16.
Article in English | MEDLINE | ID: mdl-18347024

ABSTRACT

Defects in dendritic spines and synapses contribute to cognitive deficits in mental retardation syndromes and, potentially, Alzheimer disease. p21-activated kinases (PAKs) regulate actin filaments and morphogenesis of dendritic spines regulated by the Rho family GTPases Rac and Cdc42. We previously reported that active PAK was markedly reduced in Alzheimer disease cytosol, accompanied by downstream loss of the spine actin-regulatory protein Drebrin. beta-Amyloid (Abeta) oligomer was implicated in PAK defects. Here we demonstrate that PAK is aberrantly activated and translocated from cytosol to membrane in Alzheimer disease brain and in 22-month-old Tg2576 transgenic mice with Alzheimer disease. This active PAK coimmunoprecipitated with the small GTPase Rac and both translocated to granules. Abeta42 oligomer treatment of cultured hippocampal neurons induced similar effects, accompanied by reduction of dendrites that were protected by kinase-active but not kinase-dead PAK. Abeta42 oligomer treatment also significantly reduced N-methyl-d-aspartic acid receptor subunit NR2B phosphotyrosine labeling. The Src family tyrosine kinase inhibitor PP2 significantly blocked the PAK/Rac translocation but not the loss of p-NR2B in Abeta42 oligomer-treated neurons. Src family kinases are known to phosphorylate the Rac activator Tiam1, which has recently been shown to be Abeta-responsive. In addition, anti-oligomer curcumin comparatively suppressed PAK translocation in aged Tg2576 transgenic mice with Alzheimer amyloid pathology and in Abeta42 oligomer-treated cultured hippocampal neurons. Our results implicate aberrant PAK in Abeta oligomer-induced signaling and synaptic deficits in Alzheimer disease.


Subject(s)
Alzheimer Disease/metabolism , p21-Activated Kinases/chemistry , Amyloid beta-Peptides/chemistry , Animals , Cytosol/metabolism , GTP Phosphohydrolases/chemistry , Humans , Mice , Mice, Transgenic , Models, Biological , Neurons/metabolism , Protein Transport , Rats , Rats, Sprague-Dawley , rac GTP-Binding Proteins/metabolism
8.
J Neurosci ; 27(52): 14299-307, 2007 Dec 26.
Article in English | MEDLINE | ID: mdl-18160637

ABSTRACT

Environmental and genetic factors, notably ApoE4, contribute to the etiology of late-onset Alzheimer's disease (LOAD). Reduced mRNA and protein for an apolipoprotein E (ApoE) receptor family member, SorLA (LR11) has been found in LOAD but not early-onset AD, suggesting that LR11 loss is not secondary to pathology. LR11 is a neuronal sorting protein that reduces amyloid precursor protein (APP) trafficking to secretases that generate beta-amyloid (Abeta). Genetic polymorphisms that reduce LR11 expression are associated with increased AD risk. However these polymorphisms account for only a fraction of cases with LR11 deficits, suggesting involvement of environmental factors. Because lipoprotein receptors are typically lipid-regulated, we postulated that LR11 is regulated by docosahexaenoic acid (DHA), an essential omega-3 fatty acid related to reduced AD risk and reduced Abeta accumulation. In this study, we report that DHA significantly increases LR11 in multiple systems, including primary rat neurons, aged non-Tg mice and an aged DHA-depleted APPsw AD mouse model. DHA also increased LR11 in a human neuronal line. In vivo elevation of LR11 was also observed with dietary fish oil in young rats with insulin resistance, a model for type II diabetes, another AD risk factor. These data argue that DHA induction of LR11 does not require DHA-depleting diets and is not age dependent. Because reduced LR11 is known to increase Abeta production and may be a significant genetic cause of LOAD, our results indicate that DHA increases in SorLA/LR11 levels may play an important role in preventing LOAD.


Subject(s)
Alzheimer Disease/diet therapy , Alzheimer Disease/metabolism , Docosahexaenoic Acids/administration & dosage , Gene Expression Regulation/drug effects , Membrane Transport Proteins/metabolism , Receptors, LDL/metabolism , Age Factors , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Cell Line, Tumor , Disease Models, Animal , Embryo, Mammalian , Female , Humans , Male , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neuroblastoma , Rats , Rats, Sprague-Dawley , Receptors, LDL/genetics , Time Factors
9.
J Neurochem ; 103(4): 1594-607, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17760871

ABSTRACT

Extracellular-signal regulated kinase (ERK) signaling is critical for memory and tightly regulated by acute environmental stimuli. In Alzheimer disease transgenic models, active ERK is shown to first be increased, then later reduced, but whether these baseline changes reflect disruptions in ERK signaling is less clear. We investigated the influence of the familial Alzheimer's disease transgene APPsw and beta-amyloid peptide (Abeta) immunoneutralization on cannulation injury-associated (i.c.v. infusion) ERK activation. At both 12 and 22 months of age, the trauma-associated activation of ERK observed in Tg(-) mice was dramatically attenuated in Tg(+). In cortices of 22-month-old non-infused mice, a reduction in ERK activation was observed in Tg(+), relative to Tg(-) mice. Intracerebroventricular (i.c.v.) anti-Abeta infusion significantly increased phosphorylated ERK, its substrate cAMP-response element-binding protein (CREB) and a downstream target, the NMDA receptor subunit. We also demonstrated that Abeta oligomer decreased active ERK and subsequently active CREB in human neuroblastoma cells, which could be prevented by oligomer immunoneutralization. Abeta oligomers also inhibited active ERK and CREB in primary neurons, in addition to reducing the downstream post-synaptic protein NMDA receptor subunit. These effects were reversed by anti-oligomer. Our data strongly support the existence of an APPsw transgene-dependent and Abeta oligomer-mediated defect in regulation of ERK activation.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Peptides/physiology , CREB-Binding Protein/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , MAP Kinase Signaling System/genetics , Transgenes/physiology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/genetics , Animals , CREB-Binding Protein/genetics , Cell Line, Tumor , Disease Models, Animal , Enzyme Activation/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Humans , Mice , Mice, Transgenic
10.
Nat Neurosci ; 9(2): 234-42, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16415866

ABSTRACT

Defects in dendritic spines are common to several forms of cognitive deficits, including mental retardation and Alzheimer disease. Because mutation of p21-activated kinase (PAK) can lead to mental retardation and because PAK-cofilin signaling is critical in dendritic spine morphogenesis and actin dynamics, we hypothesized that the PAK pathway is involved in synaptic and cognitive deficits in Alzheimer disease. Here, we show that PAK and its activity are markedly reduced in Alzheimer disease and that this is accompanied by reduced and redistributed phosphoPAK, prominent cofilin pathology and downstream loss of the spine actin-regulatory protein drebrin, which cofilin removes from actin. We found that beta-amyloid (Abeta) was directly involved in PAK signaling deficits and drebrin loss in Abeta oligomer-treated hippocampal neurons and in the Appswe transgenic mouse model bearing a double mutation leading to higher Abeta production. In addition, pharmacological PAK inhibition in adult mice was sufficient to cause similar cofilin pathology, drebrin loss and memory impairment, consistent with a potential causal role of PAK defects in cognitive deficits in Alzheimer disease.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Peptides/metabolism , Cognition Disorders/enzymology , Protein Serine-Threonine Kinases/metabolism , Actin Depolymerizing Factors/metabolism , Alzheimer Disease/complications , Alzheimer Disease/pathology , Animals , Cells, Cultured , Cognition Disorders/etiology , Cognition Disorders/pathology , Dendritic Spines/metabolism , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic , Neuropeptides/metabolism , Rats , p21-Activated Kinases
11.
J Neurosci Res ; 83(3): 374-84, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16385556

ABSTRACT

Although active and passive immunization against the beta-amyloid peptide (Abeta) of amyloid plaque-bearing transgenic mice markedly reduces amyloid plaque deposition and improves cognition, the mechanisms of neuroprotection and impact on toxic oligomer species are not understood. We demonstrate that compared to control IgG2b, passive immunization with intracerebroventricular (icv) anti-Abeta (1-15) antibody into the AD HuAPPsw (Tg2576) transgenic mouse model reduced specific oligomeric forms of Abeta, including the dodecamers that correlate with cognitive decline. Interestingly, the reduction of soluble Abeta oligomers, but not insoluble Abeta, significantly correlated with reduced tau phosphorylation by glycogen synthase kinase-3beta (GSK-3beta), a major tau kinase implicated previously in mediating Abeta toxicity. A conformationally-directed antibody against amyloid oligomers (larger than tetramer) also reduced Abeta oligomer-induced activation of GSK3beta and protected human neuronal SH-SY5Y cells from Abeta oligomer-induced neurotoxicity, supporting a role for Abeta oligomers in human tau kinase activation. These data suggest that antibodies that are highly specific for toxic oligomer subspecies may reduce toxicity via reduction of GSK-3beta, which could be an important strategy for Alzheimer's disease (AD) therapeutics.


Subject(s)
Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Antibodies/pharmacology , Enzyme Reactivators/pharmacology , Glycogen Synthase Kinase 3/metabolism , tau Proteins/metabolism , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Blotting, Western/methods , Cell Line, Tumor , Cell Survival/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Enzyme Activation/drug effects , Enzyme-Linked Immunosorbent Assay/methods , Female , Glycogen Synthase Kinase 3 beta , Humans , Immunohistochemistry/methods , In Vitro Techniques , Male , Mice , Mice, Transgenic , Neuroblastoma , Peptide Fragments/metabolism , Phosphorylation/drug effects , Plaque, Amyloid/pathology , Random Allocation , Silver Staining/methods
12.
J Biol Chem ; 280(7): 5892-901, 2005 Feb 18.
Article in English | MEDLINE | ID: mdl-15590663

ABSTRACT

Alzheimer's disease (AD) involves amyloid beta (Abeta) accumulation, oxidative damage, and inflammation, and risk is reduced with increased antioxidant and anti-inflammatory consumption. The phenolic yellow curry pigment curcumin has potent anti-inflammatory and antioxidant activities and can suppress oxidative damage, inflammation, cognitive deficits, and amyloid accumulation. Since the molecular structure of curcumin suggested potential Abeta binding, we investigated whether its efficacy in AD models could be explained by effects on Abeta aggregation. Under aggregating conditions in vitro, curcumin inhibited aggregation (IC(50) = 0.8 microM) as well as disaggregated fibrillar Abeta40 (IC(50) = 1 microM), indicating favorable stoichiometry for inhibition. Curcumin was a better Abeta40 aggregation inhibitor than ibuprofen and naproxen, and prevented Abeta42 oligomer formation and toxicity between 0.1 and 1.0 microM. Under EM, curcumin decreased dose dependently Abeta fibril formation beginning with 0.125 microM. The effects of curcumin did not depend on Abeta sequence but on fibril-related conformation. AD and Tg2576 mice brain sections incubated with curcumin revealed preferential labeling of amyloid plaques. In vivo studies showed that curcumin injected peripherally into aged Tg mice crossed the blood-brain barrier and bound plaques. When fed to aged Tg2576 mice with advanced amyloid accumulation, curcumin labeled plaques and reduced amyloid levels and plaque burden. Hence, curcumin directly binds small beta-amyloid species to block aggregation and fibril formation in vitro and in vivo. These data suggest that low dose curcumin effectively disaggregates Abeta as well as prevents fibril and oligomer formation, supporting the rationale for curcumin use in clinical trials preventing or treating AD.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Curcumin/metabolism , Curcumin/pharmacology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/toxicity , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzoates/chemistry , Benzoates/pharmacology , Benzothiazoles , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiology , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Congo Red , Curcumin/chemistry , Diamines/chemistry , Diamines/pharmacology , Humans , Ibuprofen/pharmacology , Mice , Mice, Transgenic , Naproxen/pharmacology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Plaque, Amyloid/chemistry , Plaque, Amyloid/genetics , Protein Binding/drug effects , Protein Denaturation/drug effects , Protein Structure, Quaternary/drug effects , Pyridazines/chemistry , Pyridazines/pharmacology , Solubility , Thiazoles
13.
J Neurosci ; 24(49): 11120-6, 2004 Dec 08.
Article in English | MEDLINE | ID: mdl-15590928

ABSTRACT

Insulin-degrading enzyme (IDE) is one of the proteins that has been demonstrated to play a key role in degrading beta-amyloid (Abeta) monomer in vitro and in vivo, raising the possibility of upregulating IDE as an approach to reduce Abeta. Little is known, however, about the cellular and molecular regulation of IDE protein. Because one of the main functions of IDE is to degrade insulin, we hypothesized that there is a negative feedback mechanism whereby stimulation of insulin receptor-mediated signaling upregulates IDE to prevent chronic activation of the pathway. We show that treatment of primary hippocampal neurons with insulin increased IDE protein levels by approximately 25%. Insulin treatment also led to phosphatidylinositol-3 (PI3) kinase activation evidenced by Akt phosphorylation, which was blocked by PI3 kinase inhibitors, wortmannin and LY 294002. Inhibition of PI3 kinase abolished the IDE upregulation by insulin, indicating a cause-effect relationship between insulin signaling and IDE upregulation. Further support for this link was provided by the findings that deficient insulin signaling (decreased PI3 kinase subunit P85) was correlated with reduced IDE in Alzheimer's disease (AD) brains and in Tg2576 Swedish amyloid precursor protein transgenic mice fed a safflower oil-enriched ("Bad") diet used to accelerate pathogenesis. Consistent with IDE function in the degradation of Abeta monomer, the IDE decrease in the Bad diet-fed Tg2576 mice was associated with increased Abeta monomer levels. These in vitro and in vivo analyses validate the use of enhanced CNS insulin signaling as a potential strategy for AD intervention to correct the IDE defects occurring in AD.


Subject(s)
Alzheimer Disease/enzymology , Brain/enzymology , Insulin/physiology , Insulysin/metabolism , Aged , Aged, 80 and over , Amyloid beta-Peptides/metabolism , Animals , Cells, Cultured , Diabetes Mellitus/metabolism , Female , Hippocampus/enzymology , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Neurons/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Insulin/physiology , Safflower Oil , Signal Transduction/physiology , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...