Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
ACS Chem Neurosci ; 12(19): 3719-3732, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34519476

ABSTRACT

Protein citrullination (deimination of arginine residue) is a well-known biomarker of inflammation. Elevated protein citrullination has been shown to colocalize with extracellular amyloid plaques in postmortem AD patient brains. Amyloid-ß (Aß) peptides which aggregate and accumulate in the plaques of Alzheimer's disease (AD) have sequential N-terminal truncations and multiple post-translational modifications (PTM) such as isomerization, pyroglutamate formation, phosphorylation, nitration, and dityrosine cross-linking. However, no conclusive biochemical evidence exists whether citrullinated Aß is present in AD brains. In this study, using high-resolution mass spectrometry, we have identified citrullination of Aß in sporadic and familial AD brains by characterizing the tandem mass spectra of endogenous N-truncated citrullinated Aß peptides. Our quantitative estimations demonstrate that ∼ 35% of pyroglutamate3-Aß pool was citrullinated in plaques in the sporadic AD temporal cortex and ∼ 22% in the detergent-insoluble frontal cortex fractions. Similarly, hypercitrullinated pyroglutamate3-Aß (∼ 30%) was observed in both the detergent-soluble as well as insoluble Aß pool in familial AD cases. Our results indicate that a common mechanism for citrullination of Aß exists in both the sporadic and familial AD. We establish that citrullination of Aß is a remarkably common PTM, closely associated with pyroglutamate3-Aß formation and its accumulation in AD. This may have implications for Aß toxicity, autoantigenicity of Aß, and may be relevant for the design of diagnostic assays and therapeutic targeting.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Amyloid beta-Peptides/metabolism , Brain/metabolism , Citrullination , Humans , Plaque, Amyloid
2.
Brain Commun ; 3(2): fcab028, 2021.
Article in English | MEDLINE | ID: mdl-33928245

ABSTRACT

Plaques that characterize Alzheimer's disease accumulate over 20 years as a result of decreased clearance of amyloid-ß peptides. Such long-lived peptides are subjected to multiple post-translational modifications, in particular isomerization. Using liquid chromatography ion mobility separations mass spectrometry, we characterized the most common isomerized amyloid-ß peptides present in the temporal cortex of sporadic Alzheimer's disease brains. Quantitative assessment of amyloid-ß N-terminus revealed that > 80% of aspartates (Asp-1 and Asp-7) in the N-terminus was isomerized, making isomerization the most dominant post-translational modification of amyloid-ß in Alzheimer's disease brain. Total amyloid-ß1-15 was ∼85% isomerized at Asp-1 and/or Asp-7 residues, with only 15% unmodified amyloid-ß1-15 left in Alzheimer's disease. While amyloid-ß4-15 the next most abundant N-terminus found in Alzheimer's disease brain, was only ∼50% isomerized at Asp-7 in Alzheimer's disease. Further investigations into different biochemically defined amyloid-ß-pools indicated a distinct pattern of accumulation of extensively isomerized amyloid-ß in the insoluble fibrillar plaque and membrane-associated pools, while the extent of isomerization was lower in peripheral membrane/vesicular and soluble pools. This pattern correlated with the accumulation of aggregation-prone amyloid-ß42 in Alzheimer's disease brains. Isomerization significantly alters the structure of the amyloid-ß peptide, which not only has implications for its degradation, but also for oligomer assembly, and the binding of therapeutic antibodies that directly target the N-terminus, where these modifications are located.

3.
J Alzheimers Dis ; 44(4): 1131-43, 2015.
Article in English | MEDLINE | ID: mdl-25408207

ABSTRACT

Biomarkers enabling the preclinical identification of Alzheimer's disease (AD) remain one of the major unmet challenges in the field. The blood cellular fractions offer a viable alternative to current cerebrospinal fluid and neuroimaging modalities. The current study aimed to replicate our earlier reports of altered binding within the AD-affected blood cellular fraction to copper-loaded immobilized metal affinity capture (IMAC) arrays. IMAC and anti-amyloid-ß (Aß) antibody arrays coupled with mass spectrometry were used to analyze blood samples collected from 218 participants from within the AIBL Study of Aging. Peripheral Aß was fragile and prone to degradation in the AIBL samples, even when stored at -80°C. IMAC analysis of the AIBL samples lead to the isolation and identification of alpha-defensins 1 and 2 at elevated levels in the AD periphery, validating earlier findings. Alpha-defensins 1 and 2 were elevated in AD patients indicating that an inflammatory phenotype is present in the AD periphery; however, peripheral Aß levels are required to supplement their prognostic power.


Subject(s)
Alzheimer Disease/blood , alpha-Defensins/blood , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Apolipoproteins E/genetics , Case-Control Studies , Female , Follow-Up Studies , Humans , Male , Neuropsychological Tests , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Statistics, Nonparametric
5.
FEBS Lett ; 588(20): 3739-43, 2014 Oct 16.
Article in English | MEDLINE | ID: mdl-25171862

ABSTRACT

Expression of human amyloid-ß (Aß) in Drosophila is frequently used to investigate its toxicity in vivo. We expressed Aß1-42 in the fly using a secretion signal derived from the Drosophila necrotic gene, as described in several previous publications. Surface-enhanced laser desorption/ionization TOF MS analysis revealed that the Aß produced contained an additional glutamine residue at the N-terminus. AßQ+1-42 was found to have increased protein abundance and to cause more severe neurodegenerative effects than wild type Aß1-42 as assessed by locomotor activity and lifespan assays. These data reveal that a commonly used model of Alzheimer's disease generates incorrect Aß peptide.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Drosophila/metabolism , Peptide Fragments/chemistry , Phenotype , Alzheimer Disease/genetics , Amino Acid Sequence , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Drosophila/genetics , Drosophila/physiology , Glutamine/chemistry , Molecular Sequence Data , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Sorting Signals , Protein Structure, Tertiary
6.
Acta Neuropathol ; 127(6): 803-10, 2014.
Article in English | MEDLINE | ID: mdl-24803227

ABSTRACT

Reducing amyloid-ß peptide (Aß) burden at the pre-symptomatic stages of Alzheimer's disease (AD) is currently the advocated clinical strategy for treating this disease. The most developed method for targeting Aß is the use of monoclonal antibodies including bapineuzumab, solanezumab and crenezumab. We have synthesized these antibodies and used surface plasmon resonance (SPR) and mass spectrometry to characterize and compare the ability of these antibodies to target Aß in transgenic mouse tissue as well as human AD tissue. SPR analysis showed that the antibodies were able to bind Aß with high affinity. All of the antibodies were able to bind Aß in mouse tissue. However, significant differences were observed in human brain tissue. While bapineuzumab was able to capture a variety of N-terminally truncated Aß species, the Aß detected using solanezumab was barely above detection limits while crenezumab did not detect any Aß. None of the antibodies were able to detect any Aß species in human blood. Immunoprecipitation experiments using plasma from AD subjects showed that both solanezumab and crenezumab have extensive cross-reactivity with non-Aß related proteins. Bapineuzumab demonstrated target engagement with brain Aß, consistent with published clinical data. Solanezumab and crenezumab did not, most likely as a result of a lack of specificity due to cross-reactivity with other proteins containing epitope overlap. This lack of target engagement raises questions as to whether solanezumab and crenezumab are suitable drug candidates for the preventative clinical trials for AD.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Antibodies/metabolism , Nootropic Agents/pharmacology , Alzheimer Disease/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Brain/drug effects , Brain/metabolism , Female , Humans , Mice, Transgenic , Surface Plasmon Resonance
7.
Alzheimers Dement ; 10(1): 53-61, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23491263

ABSTRACT

BACKGROUND: A practical biomarker is required to facilitate the preclinical diagnosis of Alzheimer's disease (AD). METHODS: Plasma amyloid beta (Aß)1-40, Aß1-42, Aßn-40, and Aßn-42 peptides were measured at baseline and after 18 months in 771 participants from the Australian Imaging Biomarkers and Lifestyle (AIBL) study of aging. Aß peptide levels were compared with clinical pathology, neuroimaging and neuropsychological measurements. RESULTS: Although inflammatory and renal function covariates influenced plasma Aß levels significantly, a decrease in Aß1-42/Aß1-40 was observed in patients with AD, and was also inversely correlated with neocortical amyloid burden. During the 18 months, plasma Aß1-42 decreased in subjects with mild cognitive impairment (MCI) and in those transitioning from healthy to MCI. CONCLUSION: Our findings are consistent with a number of published plasma Aß studies and, although the prognostic value of individual measures in any given subject is limited, the diagnostic contribution of plasma Aß may demonstrate utility when combined with a panel of peripheral biomarkers.


Subject(s)
Aging/blood , Alzheimer Disease/blood , Amyloid beta-Peptides/blood , Peptide Fragments/blood , Aged , Aged, 80 and over , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/genetics , Apolipoprotein E4/genetics , Chi-Square Distribution , Cognitive Dysfunction/blood , Cognitive Dysfunction/diagnostic imaging , Cohort Studies , Disease Progression , Female , Humans , Male , Middle Aged , Neuropsychological Tests , Positron-Emission Tomography
8.
Acta Neuropathol ; 125(4): 549-64, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23354835

ABSTRACT

The formation of low-order oligomers of ß-amyloid (Aß) within the brain is widely believed to be a central component of Alzheimer's disease (AD) pathogenesis. However, despite advances in high-throughput and high-resolution techniques such as xMAP and mass spectrometry (MS), investigations into these oligomeric species have remained reliant on low-resolution Western blots and enzyme-linked immunosorbent assays. The current investigation compared Aß profiles within human cortical tissue using sodium dodecyl sulphate (SDS) polyacrylamide gel electrophoresis (PAGE), xMAP and surface enhanced laser desorption/ionization time-of-flight MS and found that whilst there was significant correlation across the techniques regarding levels of monomeric Aß, only SDS-PAGE was capable of detecting dimeric isoforms of Aß. The addition of synthetic di-tyrosine cross-linked Aß(1-40)Met(35)(O) to the AD tissue demonstrated that the MS methodology was capable of observing dimeric Aß at femto-molar concentrations, with no noticeable effect on monomeric Aß levels. Focus turned to the association between SDS-PAGE and levels of observable dimeric Aß within the AD brain tissue. These investigations revealed that increased levels of dimeric Aß were observed with increasing concentrations of SDS in the sample buffer. This finding was subsequently confirmed using synthetic Aß(1-42) and suggests that SDS was inducing the formation of dimeric Aß. The findings that SDS promotes Aß dimerization have significant implications for the putative role of low-order oligomers in AD pathogenesis and draw into question the utility of oligomeric Aß as a therapeutic target.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Artifacts , Brain/metabolism , Electrophoresis, Polyacrylamide Gel , Alzheimer Disease/pathology , Brain/pathology , Dimerization , Humans
9.
J Alzheimers Dis ; 30(2): 323-36, 2012.
Article in English | MEDLINE | ID: mdl-22426018

ABSTRACT

Effective therapeutic interventions for Alzheimer's disease (AD) will require treatment regimes to move toward the earliest stages of the disease. For this to occur the field has to identify biomarkers that are able to accurately identify individuals at risk for progression toward AD in the presymptomatic stage. One very significant implication is that some form of population-based screening will need to be undertaken in order to identify those at risk. To date, efforts in neuroimaging brain amyloid-ß (Aß) and changes in cerebrospinal fluid Aß and tau levels shows promise, however, it is questionable as to whether these methods are applicable for screening the general population. The Aß peptide is also found in blood which is the most economical and efficient biological fluid to analyze. Unfortunately, investigations into blood-based diagnostic markers have produced mixed results. This variability is likely to be the result of differences in the preanalytical processing of samples and as such is delaying progress in the field. Reported preanalytical processing techniques from 87 recent articles focusing on the measurement of Aß in blood were compared, to investigate whether basic sample-handling techniques were comparable between studies. This comparison revealed that not only is it likely that some of the variability in blood-based results is attributable to discrepancies in preanalytical methodologies but also that the field is failing to adequately report sample processing techniques. This review highlights the current shortcomings in methodological reporting and recommends a standardized blood collection methodology based on the limited consensus of the reviewed articles.


Subject(s)
Alzheimer Disease/blood , Alzheimer Disease/diagnosis , Amyloid beta-Peptides/blood , Chemistry, Clinical/standards , Chemistry, Clinical/trends , Alzheimer Disease/epidemiology , Biomarkers/blood , Blood Specimen Collection/standards , Blood Specimen Collection/trends , Humans , Risk Factors
10.
Methods Mol Biol ; 752: 229-40, 2011.
Article in English | MEDLINE | ID: mdl-21713641

ABSTRACT

Alzheimer's disease (AD) is a highly heterogeneous and progressive dementia which is characterised by a progressive decline in cognitive functioning, selective neuronal atrophy, and loss of cortical volume in areas involved in learning and memory. However, recent research has indicated that the AD-affected brain is also besieged by increases in oxidative stress as well as perturbations to the homeostasis of biometals, such as copper and iron. These metals are known to interact with the neuropathological hallmark of AD, the ß-amyloid peptide (Aß), in a manner which increases Aß's neurotoxic effects. This knowledge has led to the development of therapeutic measures which act to restore biometal homeostasis within the AD brain. This chapter outlines how Surface-Enhanced Laser Desorption/Ionisation Time-of-Flight Mass Spectrometry can be used to monitor Aß levels within biological systems as well as describing the use of immobilised metal affinity capture in the observation of synthetic Aß peptides.


Subject(s)
Alzheimer Disease/metabolism , Metals/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Animals , Antibodies/immunology , Coumaric Acids/chemistry , Humans , Immobilized Proteins/chemistry , Immobilized Proteins/immunology , Immobilized Proteins/metabolism , Mice , Mice, Transgenic , Surface Properties
11.
J Alzheimers Dis ; 24(1): 47-59, 2011.
Article in English | MEDLINE | ID: mdl-21157020

ABSTRACT

Diagnostic measures for Alzheimer's disease (AD) commonly rely on evaluating the levels of amyloid-ß (Aß) peptides within the cerebrospinal fluid (CSF) of affected individuals. These levels are often combined with levels of an additional non-Aß marker to increase predictive accuracy. Recent efforts to overcome the invasive nature of CSF collection led to the observation of Aß species within the blood cellular fraction, however, little is known of what additional biomarkers may be found in this membranous fraction. The current study aimed to undertake a discovery-based proteomic investigation of the blood cellular fraction from AD patients (n = 18) and healthy controls (HC; n = 15) using copper immobilized metal affinity capture and Surface Enhanced Laser Desorption/Ionisation Time-Of-Flight Mass Spectrometry. Three candidate biomarkers were observed which could differentiate AD patients from HC (ROC AUC > 0.8). Bivariate pairwise comparisons revealed significant correlations between these markers and measures of AD severity including; MMSE, composite memory, brain amyloid burden, and hippocampal volume. A partial least squares regression model was generated using the three candidate markers along with blood levels of Aß. This model was able to distinguish AD from HC with high specificity (90%) and sensitivity (77%) and was able to separate individuals with mild cognitive impairment (MCI) who converted to AD from MCI non-converters. While requiring further characterization, these candidate biomarkers reaffirm the potential efficacy of blood-based investigations into neurodegenerative conditions. Furthermore, the findings indicate that the incorporation of non-amyloid markers into predictive models, function to increase the accuracy of the diagnostic potential of Aß.


Subject(s)
Alzheimer Disease/blood , Alzheimer Disease/diagnosis , Amyloid beta-Peptides/blood , Aged , Aged, 80 and over , Biomarkers/blood , Cognition Disorders/blood , Cognition Disorders/diagnosis , Cohort Studies , Female , Follow-Up Studies , Hippocampus/pathology , Humans , Male , Middle Aged , Predictive Value of Tests , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/standards
12.
J Neurosci ; 30(18): 6315-22, 2010 May 05.
Article in English | MEDLINE | ID: mdl-20445057

ABSTRACT

Alzheimer's disease (AD) is the most common age-related dementia. Unfortunately due to a lack of validated biomarkers definitive diagnosis relies on the histological demonstration of amyloid-beta (Abeta) plaques and tau neurofibrillary tangles. Abeta processing is implicated in AD progression and many therapeutic strategies target various aspects of this biology. While Abeta deposition is the most prominent feature of AD, oligomeric forms of Abeta have been implicated as the toxic species inducing the neuronal dysfunction. Currently there are no methods allowing routine monitoring of levels of such species in living populations. We have used surface enhanced laser desorption ionization time of flight (SELDI-TOF) mass spectrometry incorporating antibody capture to investigate whether the cellular membrane-containing fraction of blood provides a new source of biomarkers. There are significant differences in the mass spectra profiles of AD compared with HC subjects, with significantly higher levels of Abeta monomer and dimer in the blood of AD subjects. Furthermore, levels of these species correlated with clinical markers of AD including brain Abeta burden, cognitive impairment and brain atrophy. These results indicate that fundamental biochemical events relevant to AD can be monitored in blood, and that the species detected may be useful clinical biomarkers for AD.


Subject(s)
Alzheimer Disease/blood , Alzheimer Disease/diagnosis , Amyloid beta-Peptides/blood , Biomarkers/blood , Aged , Alzheimer Disease/diagnostic imaging , Amyloid beta-Peptides/metabolism , Brain/metabolism , Brain/pathology , Female , Humans , Male , Protein Multimerization , Radionuclide Imaging , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
13.
Eur Biophys J ; 39(10): 1407-19, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20309679

ABSTRACT

Inclusions of aggregated alpha-synuclein (alpha-syn) in dopaminergic neurons are a characteristic histological marker of Parkinson's disease (PD). In vitro, alpha-syn in the presence of dopamine (DA) at physiological pH forms SDS-resistant non-amyloidogenic oligomers. We used a combination of biophysical techniques, including sedimentation velocity analysis, small angle X-ray scattering (SAXS) and circular dichroism spectroscopy to study the characteristics of alpha-syn oligomers formed in the presence of DA. Our SAXS data show that the trimers formed by the action of DA on alpha-syn consist of overlapping worm-like monomers, with no end-to-end associations. This lack of structure contrasts with the well-established, extensive beta-sheet structure of the amyloid fibril form of the protein and its pre-fibrillar oligomers. We propose on the basis of these and earlier data that oxidation of the four methionine residues at the C- and N-terminal ends of alpha-syn molecules prevents their end-to-end association and stabilises oligomers formed by cross linking with DA-quinone/DA-melanin, which are formed as a result of the redox process, thus inhibiting formation of the beta-sheet structure found in other pre-fibrillar forms of alpha-syn.


Subject(s)
Dopamine/pharmacology , Protein Multimerization/drug effects , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , Amyloid/chemistry , Amyloid/metabolism , Benzoquinones/chemistry , Benzoquinones/metabolism , Circular Dichroism , Cross-Linking Reagents/pharmacology , Electron Spin Resonance Spectroscopy , Hydrogen-Ion Concentration , Melanins/chemistry , Melanins/metabolism , Methionine/chemistry , Methionine/metabolism , Oxidation-Reduction , Parkinson Disease/metabolism , Protein Folding/drug effects , Protein Multimerization/physiology , Protein Structure, Secondary/drug effects , Scattering, Small Angle , Ultracentrifugation
14.
J Biol Chem ; 284(34): 22697-702, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19574211

ABSTRACT

Transgenic expression of human amyloid beta (A beta) peptide in body wall muscle cells of Caenorhabditis elegans has been used to better understand aspects of Alzheimer disease (AD). In human aging and AD, A beta undergoes post-translational changes including covalent modifications, truncations, and oligomerization. Amino truncated A beta is increasingly recognized as potentially contributing to AD pathogenesis. Here we describe surface-enhanced laser desorption ionization-time of flight mass spectrometry mass spectrometry of A beta peptide in established transgenic C. elegans lines. Surprisingly, the A beta being expressed is not full-length 1-42 (amino acids) as expected but rather a 3-42 truncation product. In vitro analysis demonstrates that A beta(3-42) self-aggregates like A beta(1-42), but more rapidly, and forms fibrillar structures. Similarly, A beta(3-42) is also the more potent initiator of A beta(1-40) aggregation. Seeded aggregation via A beta(3-42) is further enhanced via co-incubation with the transition metal Cu(II). Although unexpected, the C. elegans model of A beta expression can now be co-opted to study the proteotoxic effects and processing of A beta(3-42).


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Caenorhabditis elegans/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/ultrastructure , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/ultrastructure , Humans , Immunoblotting , Microscopy, Electron, Transmission , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
15.
J Neurochem ; 108(5): 1198-207, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19141082

ABSTRACT

Accumulation of neurotoxic amyloid-beta (Abeta) is central to the pathology of Alzheimer's disease (AD). Elucidating the mechanisms of Abeta accumulation will therefore expedite the development of Abeta-targeting AD therapeutics. We examined activity of an Abeta-degrading protease (matrix metalloprotease 2) to investigate whether biochemical factors consistent with conditions in the AD brain contribute to Abeta accumulation by altering Abeta sensitivity to proteolytic degradation. An Abeta amino acid mutation found in familial AD, Abeta interactions with zinc (Zn), and increased Abeta hydrophobicity all strongly prevented Abeta degradation. Consistent to all of these factors is the promotion of specific Abeta aggregates where the protease cleavage site, confirmed by mass spectrometry, is inaccessible within an amyloid structure. These data indicate decreased degradation due to amyloid formation initiates Abeta accumulation by preventing normal protease activity. Zn also prevented Abeta degradation by the proteases neprilysin and insulin degrading enzyme. Treating Zn-induced Abeta amyloid with the metal-protein attenuating compound clioquinol reversed amyloid formation and restored the peptide's sensitivity to degradation by matrix metalloprotease 2. This provides new data indicating that therapeutic compounds designed to modulate Abeta-metal interactions can inhibit Abeta accumulation by restoring the catalytic potential of Abeta-degrading proteases.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid/metabolism , Amyloid/drug effects , Amyloid beta-Peptides/drug effects , Amyloid beta-Peptides/genetics , Clioquinol/pharmacology , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay/methods , Glutamic Acid/genetics , Glutamine/genetics , Humans , Insulysin/pharmacology , Matrix Metalloproteinase 2/metabolism , Microscopy, Electron, Transmission/methods , Mutation , Neprilysin/pharmacology , Peptide Fragments/drug effects , Peptide Fragments/genetics , Peptide Fragments/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Time Factors , Zinc/pharmacology
16.
Proc Natl Acad Sci U S A ; 106(2): 381-6, 2009 Jan 13.
Article in English | MEDLINE | ID: mdl-19122148

ABSTRACT

Cognitive decline in Alzheimer's disease (AD) involves pathological accumulation of synaptotoxic amyloid-beta (Abeta) oligomers and hyperphosphorylated tau. Because recent evidence indicates that glycogen synthase kinase 3beta (GSK3beta) activity regulates these neurotoxic pathways, we developed an AD therapeutic strategy to target GSK3beta. The strategy involves the use of copper-bis(thiosemicarbazonoto) complexes to increase intracellular copper bioavailability and inhibit GSK3beta through activation of an Akt signaling pathway. Our lead compound Cu(II)(gtsm) significantly inhibited GSK3beta in the brains of APP/PS1 transgenic AD model mice. Cu(II)(gtsm) also decreased the abundance of Abeta trimers and phosphorylated tau, and restored performance of AD mice in the Y-maze test to levels expected for cognitively normal animals. Improvement in the Y-maze correlated directly with decreased Abeta trimer levels. This study demonstrates that increasing intracellular copper bioavailability can restore cognitive function by inhibiting the accumulation of neurotoxic Abeta trimers and phosphorylated tau.


Subject(s)
Amyloid beta-Peptides/drug effects , Copper/pharmacology , tau Proteins/drug effects , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Cognition/drug effects , Copper/pharmacokinetics , Copper/therapeutic use , Dimerization , Disease Models, Animal , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Glycogen Synthase Kinases/antagonists & inhibitors , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organometallic Compounds/pharmacokinetics , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...