Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 313
Filter
1.
Anal Chem ; 96(24): 9799-9807, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38830618

ABSTRACT

Cerebral accumulation of amyloid-ß (Aß) initiates molecular and cellular cascades that lead to Alzheimer's disease (AD). However, amyloid deposition does not invariably lead to dementia. Amyloid-positive but cognitively unaffected (AP-CU) individuals present widespread amyloid pathology, suggesting that molecular signatures more complex than the total amyloid burden are required to better differentiate AD from AP-CU cases. Motivated by the essential role of Aß and the key lipid involvement in AD pathogenesis, we applied multimodal mass spectrometry imaging (MSI) and machine learning (ML) to investigate amyloid plaque heterogeneity, regarding Aß and lipid composition, in AP-CU versus AD brain samples at the single-plaque level. Instead of focusing on a population mean, our analytical approach allowed the investigation of large populations of plaques at the single-plaque level. We found that different (sub)populations of amyloid plaques, differing in Aß and lipid composition, coexist in the brain samples studied. The integration of MSI data with ML-based feature extraction further revealed that plaque-associated gangliosides GM2 and GM1, as well as Aß1-38, but not Aß1-42, are relevant differentiators between the investigated pathologies. The pinpointed differences may guide further fundamental research investigating the role of amyloid plaque heterogeneity in AD pathogenesis/progression and may provide molecular clues for further development of emerging immunotherapies to effectively target toxic amyloid assemblies in AD therapy. Our study exemplifies how an integrative analytical strategy facilitates the unraveling of complex biochemical phenomena, advancing our understanding of AD from an analytical perspective and offering potential avenues for the refinement of diagnostic tools.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Brain , Plaque, Amyloid , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/diagnosis , Humans , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/analysis , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Plaque, Amyloid/chemistry , Brain/metabolism , Brain/pathology , Lipids/analysis , Lipids/chemistry , Machine Learning , Aged
2.
Chemistry ; 30(34): e202400322, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38629212

ABSTRACT

This study presents the synthesis and characterization of two fluorescent norbornadiene (NBD) photoswitches, each incorporating two conjugated pyrene units. Expanding on the limited repertoire of reported photoswitchable fluorescent NBDs, we explore their properties with a focus on applications in bioimaging of amyloid beta (Aß) plaques. While the fluorescence emission of the NBD decreases upon photoisomerization, aligning with what has been previously reported, for the first time we observed luminescence after irradiation of the quadricyclane (QC) isomer. We deduce how the observed emission is induced by photoisomerization to the excited state of the parent isomer (NBD) which is then the emitting species. Thorough characterizations including NMR, UV-Vis, fluorescence, X-ray structural analysis and density functional theory (DFT) calculations provide a comprehensive understanding of these systems. Notably, one NBD-QC system exhibits exceptional durability. Additionally, these molecules serve as effective fluorescent stains targeting Aß plaques in situ, with observed NBD/QC switching within the plaques. Molecular docking simulations explore NBD interactions with amyloid, unveiling novel binding modes. These insights mark a crucial advancement in the comprehension and design of future photochromic NBDs for bioimaging applications and beyond, emphasizing their potential in studying and addressing protein aggregates.


Subject(s)
Amyloid beta-Peptides , Fluorescent Dyes , Pyrenes , Fluorescent Dyes/chemistry , Pyrenes/chemistry , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Humans , Molecular Docking Simulation , Norbornanes/chemistry , Plaque, Amyloid/chemistry , Plaque, Amyloid/diagnostic imaging , Density Functional Theory , Isomerism , Spectrometry, Fluorescence
3.
Bioorg Med Chem ; 95: 117513, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37931520

ABSTRACT

In this article, the development of fluorescent imaging probes for the detection of Alzheimer's disease (AD)-associated protein aggregates is described. Indane derivatives with a donor-π-acceptor (D-π-A) structure were designed and synthesized. The probes were evaluated for their ability to bind to ß-amyloid (Aß) protein aggregates, which are a key pathological hallmark of AD. The results showed that several probes exhibited significant changes in fluorescence intensity at wavelengths greater than 600 nm when they were bound to Aß aggregates compared to the Aß monomeric form. Among the tested probes, four D-π-A type indane derivatives showed promising binding selectivity to Aß aggregates over non-specific proteins such as bovine serum albumin (BSA). The molecular docking study showed that our compounds were appropriately located along the Aß fibril axis through the hydrophobic tunnel structure. Further analysis revealed that the most active compound having dimethylaminopyridyl group as an election donor and dicyano group as an electron acceptor could effectively stain Aß plaques in brain tissue samples from AD transgenic mice. These findings suggest that our indane-based compounds have the potential to serve as fluorescent probes for the detection and monitoring of Aß aggregation in AD.


Subject(s)
Alzheimer Disease , Mice , Animals , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Fluorescent Dyes/chemistry , Protein Aggregates , Molecular Docking Simulation , Amyloid beta-Peptides/metabolism , Mice, Transgenic , Brain/metabolism , Plaque, Amyloid/chemistry , Plaque, Amyloid/diagnosis , Plaque, Amyloid/pathology
4.
Acta Biomater ; 170: 260-272, 2023 10 15.
Article in English | MEDLINE | ID: mdl-37574159

ABSTRACT

Amyloid-ß (Aß) plaques from Alzheimer's Disease (AD) can be visualized ex vivo in label-free brain samples using synchrotron X-ray phase-contrast tomography (XPCT). However, for XPCT to be useful as a screening method for amyloid pathology, it is essential to understand which factors drive the detection of Aß plaques. The current study was designed to test the hypothesis that Aß-related contrast in XPCT could be caused by Aß fibrils and/or by metals trapped in the plaques. Fibrillar and elemental compositions of Aß plaques were probed in brain samples from different types of AD patients and AD models to establish a relationship between XPCT contrast and Aß plaque characteristics. XPCT, micro-Fourier-Transform Infrared spectroscopy and micro-X-Ray Fluorescence spectroscopy were conducted on human samples (one genetic and one sporadic case) and on four transgenic rodent strains (mouse: APPPS1, ArcAß, J20; rat: TgF344). Aß plaques from the genetic AD patient were visible using XPCT, and had higher ß-sheet content and higher metal levels than those from the sporadic AD patient, which remained undetected by XPCT. Aß plaques in J20 mice and TgF344 rats appeared hyperdense on XPCT images, while they were hypodense with a hyperdense core in the case of APPPS1 and ArcAß mice. In all four transgenic strains, ß-sheet content was similar, while metal levels were highly variable: J20 (zinc and iron) and TgF344 (copper) strains showed greater metal accumulation than APPPS1 and ArcAß mice. Hence, a hyperdense contrast formation of Aß plaques in XPCT images was associated with biometal entrapment within plaques. STATEMENT OF SIGNIFICANCE: The role of metals in Alzheimer's disease (AD) has been a subject of continuous interest. It was already known that amyloid-ß plaques (Aß), the earliest hallmark of AD, tend to trap endogenous biometals like zinc, iron and copper. Here we show that this metal accumulation is the main reason why Aß plaques are detected with a new technique called X-ray phase contrast tomography (XPCT). XPCT enables to map the distribution of Aß plaques in the whole excised brain without labeling. In this work we describe a unique collection of four transgenic models of AD, together with a human sporadic and a rare genetic case of AD, thus exploring the full spectrum of amyloid contrast in XPCT.


Subject(s)
Alzheimer Disease , Trace Elements , Humans , Mice , Animals , Rats , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/pathology , Copper/chemistry , X-Rays , Mice, Transgenic , Amyloid beta-Peptides/metabolism , Metals , Zinc/chemistry , Iron , Brain/metabolism , Amyloid , Plaque, Amyloid/diagnostic imaging , Plaque, Amyloid/chemistry , Disease Models, Animal
5.
Transl Neurodegener ; 11(1): 30, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35641972

ABSTRACT

BACKGROUND: The molecular heterogeneity of Alzheimer's amyloid-ß (Aß) deposits extends well beyond the classic Aß1-40/Aß1-42 dichotomy, substantially expanded by multiple post-translational modifications that increase the proteome diversity. Numerous truncated fragments consistently populate the brain Aß peptidome, and their homeostatic regulation and potential contribution to disease pathogenesis are largely unknown. Aß4-x peptides have been reported as major components of plaque cores and the limited studies available indicate their relative abundance in Alzheimer's disease (AD). METHODS: Immunohistochemistry was used to assess the topographic distribution of Aß4-x species in well-characterized AD cases using custom-generated monoclonal antibody 18H6-specific for Aß4-x species and blind for full-length Aß1-40/Aß1-42-in conjunction with thioflavin-S and antibodies recognizing Aßx-40 and Aßx-42 proteoforms. Circular dichroism, thioflavin-T binding, and electron microscopy evaluated the biophysical and aggregation/oligomerization properties of full-length and truncated synthetic homologues, whereas stereotaxic intracerebral injections of monomeric and oligomeric radiolabeled homologues in wild-type mice were used to evaluate their brain clearance characteristics. RESULTS: All types of amyloid deposits contained the probed Aß epitopes, albeit expressed in different proportions. Aß4-x species showed preferential localization within thioflavin-S-positive cerebral amyloid angiopathy and cored plaques, strongly suggesting poor clearance characteristics and consistent with the reduced solubility and enhanced oligomerization of their synthetic homologues. In vivo clearance studies demonstrated a fast brain efflux of N-terminally truncated and full-length monomeric forms whereas their oligomeric counterparts-particularly of Aß4-40 and Aß4-42-consistently exhibited enhanced brain retention. CONCLUSIONS: The persistence of aggregation-prone Aß4-x proteoforms likely contributes to the process of amyloid formation, self-perpetuating the amyloidogenic loop and exacerbating amyloid-mediated pathogenic pathways.


Subject(s)
Alzheimer Disease , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Antibodies, Monoclonal , Mice , Peptide Fragments , Plaque, Amyloid/chemistry , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology
6.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Article in English | MEDLINE | ID: mdl-34725161

ABSTRACT

Although amyloid plaques composed of fibrillar amyloid-ß (Aß) assemblies are a diagnostic hallmark of Alzheimer's disease (AD), quantities of amyloid similar to those in AD patients are observed in brain tissue of some nondemented elderly individuals. The relationship between amyloid deposition and neurodegeneration in AD has, therefore, been unclear. Here, we use solid-state NMR to investigate whether molecular structures of Aß fibrils from brain tissue of nondemented elderly individuals with high amyloid loads differ from structures of Aß fibrils from AD tissue. Two-dimensional solid-state NMR spectra of isotopically labeled Aß fibrils, prepared by seeded growth from frontal lobe tissue extracts, are similar in the two cases but with statistically significant differences in intensity distributions of cross-peak signals. Differences in solid-state NMR data are greater for 42-residue amyloid-ß (Aß42) fibrils than for 40-residue amyloid-ß (Aß40) fibrils. These data suggest that similar sets of fibril polymorphs develop in nondemented elderly individuals and AD patients but with different relative populations on average.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Frontal Lobe/pathology , Plaque, Amyloid/pathology , Aged, 80 and over , Case-Control Studies , Female , Humans , Magnetic Resonance Spectroscopy , Male , Plaque, Amyloid/chemistry
7.
Acta Neuropathol Commun ; 9(1): 168, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34663480

ABSTRACT

Progressive accumulation of Amyloid-ß (Aß) deposits in the brain is a characteristic neuropathological hallmark of Alzheimer's disease (AD). During disease progression, extracellular Aß plaques undergo specific changes in their composition by the sequential deposition of different modified Aß species. Microglia are implicated in the restriction of amyloid deposits and play a major role in internalization and degradation of Aß. Recent studies showed that rare variants of the Triggering Receptor Expressed on Myeloid cells 2 (TREM2) are associated with an increased risk for AD. Post-translational modifications of Aß could modulate the interaction with TREM2, and the uptake by microglia. Here, we demonstrate that genetic deletion of TREM2 or expression of a disease associated TREM2 variant in mice lead to differential accumulation of modified and non-modified Aß species in extracellular plaques and intraneuronal deposits. Human brains with rare TREM2 AD risk variants also showed altered deposition of modified Aß species in the different brain lesions as compared to cases with the common variant of TREM2. These findings indicate that TREM2 plays a critical role in the development and the composition of Aß deposits, not only in extracellular plaques, but also intraneuronally, that both could contribute to the pathogenesis of AD.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Membrane Glycoproteins/metabolism , Neurons/pathology , Plaque, Amyloid/pathology , Receptors, Immunologic/metabolism , Aged , Aged, 80 and over , Amyloid beta-Peptides/chemistry , Animals , Female , Humans , Male , Membrane Glycoproteins/genetics , Mice , Plaque, Amyloid/chemistry , Receptors, Immunologic/genetics
8.
Mol Neurodegener ; 16(1): 55, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34384464

ABSTRACT

Mass spectrometry-based proteomics empowers deep profiling of proteome and protein posttranslational modifications (PTMs) in Alzheimer's disease (AD). Here we review the advances and limitations in historic and recent AD proteomic research. Complementary to genetic mapping, proteomic studies not only validate canonical amyloid and tau pathways, but also uncover novel components in broad protein networks, such as RNA splicing, development, immunity, membrane transport, lipid metabolism, synaptic function, and mitochondrial activity. Meta-analysis of seven deep datasets reveals 2,698 differentially expressed (DE) proteins in the landscape of AD brain proteome (n = 12,017 proteins/genes), covering 35 reported AD genes and risk loci. The DE proteins contain cellular markers enriched in neurons, microglia, astrocytes, oligodendrocytes, and epithelial cells, supporting the involvement of diverse cell types in AD pathology. We discuss the hypothesized protective or detrimental roles of selected DE proteins, emphasizing top proteins in "amyloidome" (all biomolecules in amyloid plaques) and disease progression. Comprehensive PTM analysis represents another layer of molecular events in AD. In particular, tau PTMs are correlated with disease stages and indicate the heterogeneity of individual AD patients. Moreover, the unprecedented proteomic coverage of biofluids, such as cerebrospinal fluid and serum, procures novel putative AD biomarkers through meta-analysis. Thus, proteomics-driven systems biology presents a new frontier to link genotype, proteotype, and phenotype, accelerating the development of improved AD models and treatment strategies.


Subject(s)
Alzheimer Disease/metabolism , Nerve Tissue Proteins/metabolism , Proteome , Alzheimer Disease/etiology , Alzheimer Disease/genetics , Asymptomatic Diseases , Biomarkers , Blood Proteins/analysis , Cerebrospinal Fluid Proteins/analysis , Chromatography, Liquid , Cognitive Dysfunction/metabolism , Data Mining , Databases, Protein , Datasets as Topic , Humans , Meta-Analysis as Topic , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/genetics , Plaque, Amyloid/chemistry , Protein Processing, Post-Translational , Proteomics/methods , Tandem Mass Spectrometry
9.
Mol Neurodegener ; 16(1): 59, 2021 08 28.
Article in English | MEDLINE | ID: mdl-34454574

ABSTRACT

Alzheimer's disease (AD) is pathologically defined by the presence of fibrillar amyloid ß (Aß) peptide in extracellular senile plaques and tau filaments in intracellular neurofibrillary tangles. Extensive research has focused on understanding the assembly mechanisms and neurotoxic effects of Aß during the last decades but still we only have a brief understanding of the disease associated biological processes. This review highlights the many other constituents that, beside Aß, are accumulated in the plaques, with the focus on extracellular proteins. All living organisms rely on a delicate network of protein functionality. Deposition of significant amounts of certain proteins in insoluble inclusions will unquestionably lead to disturbances in the network, which may contribute to AD and copathology. This paper provide a comprehensive overview of extracellular proteins that have been shown to interact with Aß and a discussion of their potential roles in AD pathology. Methods that can expand the knowledge about how the proteins are incorporated in plaques are described. Top-down methods to analyze post-mortem tissue and bottom-up approaches with the potential to provide molecular insights on the organization of plaque-like particles are compared. Finally, a network analysis of Aß-interacting partners with enriched functional and structural key words is presented.


Subject(s)
Alzheimer Disease/metabolism , Nerve Tissue Proteins/metabolism , Plaque, Amyloid/chemistry , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Apolipoproteins/metabolism , Autopsy , Blood Coagulation Factors/metabolism , Carrier Proteins/metabolism , Cell Adhesion Molecules/metabolism , Complement System Proteins/metabolism , Extracellular Fluid/chemistry , Extracellular Matrix Proteins/metabolism , Humans , Immunoglobulins/metabolism , Laser Capture Microdissection , Lipid Metabolism , Microscopy, Confocal , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/isolation & purification , Protein Interaction Maps , Protein Isoforms , Proteoglycans/metabolism , Tandem Mass Spectrometry
10.
Bioorg Med Chem Lett ; 46: 128164, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34082067

ABSTRACT

Positron emission tomographic (PET) studies of amyloid ß (Aß) accumulation in Alzheimer's disease (AD) have shown clinical utility. The aim of this study was to develop and evaluate the effectiveness of a new fluorine-18 radiotracer [18F]Flotaza (2-{2-[2-[18F]fluoroethoxy]ethoxy}ethoxy)-4'-N,N-dimethylaminoazobenzene), for Aß plaque imaging. Nucleophilic [18F]fluoride was used in a one-step radiosynthesis for [18F]flotaza. Using post mortem human AD brain tissues consisting of anterior cingulate (AC) and corpus callosum (CC), binding affinity of Flotaza, Ki = 1.68 nM for human Aß plaques and weak (>10-5 M) for Tau protein. Radiosynthesis of [18F]Flotaza was very efficient in high radiochemical yields (>25%) with specific activities >74 GBq/µmol. Brain slices from all AD subjects were positively immunostained with anti-Aß. Ratio of [18F]Flotaza in gray matter AC to white matter CC was >100 in all the 6 subjects. Very little white matter binding was seen. [18F]Flotaza binding in AC strongly correlated with anti-Aß immunostains. [18F]Flotaza is therefore a suitable fluorine-18 PET radiotracer for PET imaging studies of human Aß plaques.


Subject(s)
Alzheimer Disease/diagnosis , Brain/diagnostic imaging , Drug Development , Plaque, Amyloid/chemistry , Alzheimer Disease/metabolism , Humans , Molecular Structure , Positron-Emission Tomography
11.
J Neurochem ; 159(2): 330-342, 2021 10.
Article in English | MEDLINE | ID: mdl-33048341

ABSTRACT

Over the last 10 years, considerable technical advances in mass spectrometry (MS)-based bioanalysis have enabled the investigation of lipid signatures in neuropathological structures. In Alzheimer´s Disease (AD) research, it is now well accepted that lipid dysregulation plays a key role in AD pathogenesis and progression. This review summarizes current MS-based strategies, notably MALDI and ToF-SIMS imaging as well as laser capture microdissection combined with LC-ESI-MS. It also presents recent advances to assess lipid alterations associated with Amyloid-ß plaques, one of the hallmarks of AD. Collectively, these methodologies offer new opportunities for the study of lipids, thus pushing forward our understanding of their role in such a complex and still untreatable disease as AD.


Subject(s)
Lipids/chemistry , Mass Spectrometry/methods , Plaque, Amyloid/chemistry , Animals , Humans , Laser Capture Microdissection , Microglia/pathology , Microglia/ultrastructure , Neuroimaging , Plaque, Amyloid/pathology , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
12.
Inorg Chem ; 59(16): 11658-11669, 2020 Aug 17.
Article in English | MEDLINE | ID: mdl-32799487

ABSTRACT

Alzheimer's disease is characterized by the presence of extracellular amyloid-ß plaques. Positron emission tomography (PET) imaging with tracers radiolabeled with positron-emitting radionuclides that bind to amyloid-ß plaques can assist in the diagnosis of Alzheimer's disease. With the goal of designing new imaging agents radiolabeled with positron-emitting copper-64 radionuclides that bind to amyloid-ß plaques, a family of bis(thiosemicarbazone) ligands with appended substituted stilbenyl functional groups has been prepared. The ligands form charge-neutral and stable complexes with copper(II). The new ligands can be radiolabeled with copper-64 at room temperature. Two lead complexes were demonstrated to bind to amyloid-ß plaques present in post-mortem brain tissue from subjects with clinically diagnosed Alzheimer's disease and crossed the blood-brain barrier in mice. The work presented here provides strategies to prepare compounds with radionuclides of copper that can be used for targeted brain PET imaging.


Subject(s)
Amyloid beta-Peptides/chemistry , Coordination Complexes/chemistry , Copper/chemistry , Plaque, Amyloid/chemistry , Stilbenes/chemistry , Thiosemicarbazones/chemistry , Crystallography, X-Ray , Molecular Structure , Protein Binding
13.
Metab Brain Dis ; 35(8): 1371-1383, 2020 12.
Article in English | MEDLINE | ID: mdl-32852699

ABSTRACT

Although there are multiple histochemical tracers available to label plaques and tangles in the brain to evaluate neuropathology in Alzheimer disease (AD), few of them are versatile in nature and compatible with immunohistochemical procedures. Congo Red (CR) is an anisotropic organic stain discovered to label amyloid beta (Aß) plaques in the brain. Unfortunately, its use is underappreciated due to its low resolution and brightness as stated in previous studies using bright field microscopy. Here, we modified a previous method to localize both plaques and tangles in brains from humans and a transgenic rodent model of AD for fluorescence microscopic visualization. The plaque staining affinities displayed by CR were compared with fibrillar pattern labeling seen with Thioflavin S. This study summarizes the optimization of protocols in which various parameters have been finetuned. To determine the target CR potentially binds, we have performed double labeling with different antibodies against Aß as well as phosphorylated Tau. The plaque staining affinities exhibited by CR are compared with those associated with the diffuse pattern of labeling seen with antibodies directed against different epitopes of Aß. Neither CP13, TNT2 or TOC1 binds all the neurofibrillary tangles as revealed by CR labeling in the human brain. Additionally, we also evaluated double labeling with AT8, AT180, and PHF1. Interestingly, PHF-1 shows 40% colocalization and AT8 shows 15% colocalization with NFT. Thus, CR is a much better marker to detect AD pathologies in human and rodent brains with higher fluorescence intensity relative to other conventional fluorescence markers.


Subject(s)
Brain/metabolism , Coloring Agents/metabolism , Congo Red/metabolism , Neurofibrillary Tangles/metabolism , Plaque, Amyloid/metabolism , Staining and Labeling/methods , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Brain/pathology , Brain Chemistry/physiology , Coloring Agents/analysis , Congo Red/analysis , Humans , Mice , Mice, Transgenic , Neurofibrillary Tangles/chemistry , Neurofibrillary Tangles/pathology , Optical Imaging/methods , Plaque, Amyloid/chemistry , Plaque, Amyloid/pathology , Rats , Rodentia
14.
Biochem Biophys Res Commun ; 532(1): 82-87, 2020 10 29.
Article in English | MEDLINE | ID: mdl-32828536

ABSTRACT

Curcumin and related compounds have been validated to remove even well-developed human ß-amyloid plaques from the brain of transgenic mice, in vivo. However, their molecular mechanism of the plaque buster activity is rather unknown. Computational chemistry was employed here to better understand the ß-amyloid protein elimination. According to our docking studies, a tautomeric "keto-enol" flip-flop mechanism is proposed that may chop up ß-amyloid plaques in Alzheimer's due to removing each hairpin-foldamers one by one from both ends of aggregated fibrils. According to the experimented models, other bi-stable "keto-enol" pharmacophores might be identified to break up amyloid plaques and enhance rapid clearance of toxic aggregates in Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Curcumin/pharmacology , Plaque, Amyloid/drug therapy , Plaque, Amyloid/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/drug effects , Animals , Brain/drug effects , Brain/metabolism , Curcumin/chemistry , Dietary Supplements , Humans , In Vitro Techniques , Mice , Mice, Transgenic , Molecular Docking Simulation , Phytotherapy , Plaque, Amyloid/chemistry , Protein Aggregates/drug effects , Protein Aggregation, Pathological , Protein Binding/drug effects
15.
BMC Res Notes ; 13(1): 128, 2020 Mar 04.
Article in English | MEDLINE | ID: mdl-32131889

ABSTRACT

OBJECTIVE: We present a method to prepare an amyloid model at scalable quantities for phantom studies to evaluate small-angle x-ray scattering systems for amyloid detection. Two amyloid models were made from a plasma protein with and without heating. Both models mimic the [Formula: see text]-sheet structure of the [Formula: see text]-amyloid ([Formula: see text]) plaques in Alzheimer's disease. Amyloid detection is based on the distinct peaks in the scattering signature of the [Formula: see text]-sheet structure. We characterized the amyloid models using a spectral small-angle x-ray scattering (sSAXS) prototype with samples in a plastic syringe and within a cylindrical polymethyl methacrylate (PMMA) phantom. RESULTS: sSAXS data show that we can detect the scattering peaks characteristic of amyloid [Formula: see text]-sheet structure in both models around 6 and 13 [Formula: see text]. The [Formula: see text] model prepared without heating provides a stronger signal in the PMMA phantom. The methods described can be used to prepare models in sufficiently large quantities and used in samples with different packing density to assess the performance of [Formula: see text] quantification systems.


Subject(s)
Phantoms, Imaging , Plaque, Amyloid/ultrastructure , Polymethyl Methacrylate/chemistry , Serum Albumin, Bovine/chemistry , Alzheimer Disease/diagnostic imaging , Animals , Cattle , Hot Temperature , Humans , Models, Biological , Plaque, Amyloid/chemistry , Protein Conformation, beta-Strand , Scattering, Small Angle , X-Ray Diffraction
16.
Chemistry ; 26(43): 9449-9453, 2020 Aug 03.
Article in English | MEDLINE | ID: mdl-32167218

ABSTRACT

HIV transactivator of transcription (Tat) protein could interact with amyloid ß (Aß) peptide which cause the growth of Aß plaques in the brain and result in Alzheimer's disease in HIV-infected patients. Herein, we employ high-resolution atomic force microscopy and quantitative nanomechanical mapping to investigate the effects of Tat protein in Aß peptide aggregation. Our results demonstrate that the Tat protein could bind to the Aß fibril surfaces and result in the formation of Tat-Aß multifibrillar structures. The resultant Tat-Aß multifibrillar aggregates represent an increase in stiffness compared with Aß fibrils due to the increase in ß-sheet formation. The identification and characterization of the Tat-Aß intermediate aggregates is important to understanding the interactions between Tat protein and Aß peptide, and the development of novel therapeutic strategy for Alzheimer's disease-like disorder in HIV infected individuals.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Peptides/chemistry , Amyloid/chemistry , Gene Products, tat/chemistry , Microscopy, Atomic Force/methods , Plaque, Amyloid/chemistry , Amyloid beta-Peptides/analysis , Gene Products, tat/metabolism , Humans , Plaque, Amyloid/metabolism
17.
Biosci Biotechnol Biochem ; 84(1): 1-16, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31538538

ABSTRACT

Recent investigations suggest that soluble oligomeric amyloid ß (Aß) species may be involved in early onset of Alzheimer's disease (AD). Using systematic proline replacement, solid-state NMR, and ESR, we identified a toxic turn at position 22 and 23 of Aß42, the most potent neurotoxic Aß species. Through radicalization, the toxic turn can induce formation of the C-terminal hydrophobic core to obtain putative Aß42 dimers and trimers. Synthesized dimer and trimer models showed that the C-terminal hydrophobic core plays a critical role in the formation of high molecular weight oligomers with neurotoxicity. Accordingly, an anti-toxic turn antibody (24B3) that selectively recognizes a toxic dimer model of E22P-Aß42 was developed. Sandwich enzyme-linked immunosorbent assay with 24B3 and 82E1 detected a significantly higher ratio of Aß42 with a toxic turn to total Aß42 in cerebrospinal fluid of AD patients compared with controls, suggesting that 24B3 could be useful for early onset of AD diagnosis.


Subject(s)
Alzheimer Disease/diagnosis , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/immunology , Antibodies, Monoclonal/therapeutic use , Peptide Fragments/cerebrospinal fluid , Peptide Fragments/immunology , Alzheimer Disease/cerebrospinal fluid , Amyloid beta-Peptides/chemistry , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Mice , Models, Molecular , Neurofibrillary Tangles/chemistry , Peptide Fragments/chemistry , Plaque, Amyloid/chemistry , Proline/chemistry , Protein Aggregates , Protein Aggregation, Pathological , Protein Structure, Tertiary
18.
ACS Chem Neurosci ; 11(1): 14-24, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31774647

ABSTRACT

Ganglioside metabolism is significantly altered in Alzheimer's disease (AD), which is a progressive neurodegenerative disease prominently characterized by one of its pathological hallmarks, amyloid deposits or "senile plaques". While the plaques mainly consist of aggregated variants of amyloid-ß protein (Aß), recent studies have revealed a number of lipid species including gangliosides in amyloid plaques along with Aß peptides. It has been widely suggested that long chain (sphingosine) base (LCBs), C18:1-LCB and C20:1-LCB, containing gangliosides might play different roles in neuronal function in vivo. In order to elucidate region-specific aspects of amyloid-plaque associated C18:1-LCB and C20:1-LCB ganglioside accumulations, high spatial resolution (10 µm per pixel) matrix assisted laser desorption ionization imaging mass spectrometry (MALDI-IMS) of gangliosides in amyloid plaques was performed in hippocampal and adjacent cortical regions of 12 month old 5xFAD mouse coronal brain sections from two different stereotaxic coordinates (bregma points, -2.2 and -2.7 mm). MALDI-IMS uncovered brain-region (2 and 3D) and/or LCB specific accumulations of monosialogangliosides (GMs): GM1, GM2, and GM3 in the hippocampal and cortical amyloid plaques. The results reveal monosialogangliosides to be an important component of amyloid plaques and the accumulation of different gangliosides is region and LCB specific in 12 month old 5xFAD mouse brain. This is discussed in relation to amyloid-associated AD pathogenesis such as lipid related immune changes in amyloid plaques, AD specific ganglioside metabolism, and, notably, AD-associated impaired neurogenesis in the subgranular zone.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , Gangliosides/chemistry , Plaque, Amyloid/chemistry , Animals , Lipidomics , Mice , Mice, Transgenic , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
19.
Neurobiol Dis ; 134: 104629, 2020 02.
Article in English | MEDLINE | ID: mdl-31669752

ABSTRACT

The loss of native function of the DJ-1 protein has been linked to the development of Parkinson's (PD) and other neurodegenerative diseases. Here we show that DJ-1 aggregates into ß-sheet structured soluble and fibrillar aggregates in vitro under physiological conditions and that this process is promoted by the oxidation of its catalytic Cys106 residue. This aggregation resulted in the loss of its native biochemical glyoxalase function and in addition oxidized DJ-1 aggregates were observed to localize within Lewy bodies, neurofibrillary tangles and amyloid plaques in human PD and Alzheimer's (AD) patients' post-mortem brain tissue. These findings suggest that the aggregation of DJ-1 may be a critical player in the development of the pathology of PD and AD and demonstrate that loss of DJ-1 function can happen through DJ-1 aggregation. This could then contribute to AD and PD disease onset and progression.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , Parkinson Disease/pathology , Protein Aggregation, Pathological/metabolism , Protein Deglycase DJ-1/metabolism , Alzheimer Disease/metabolism , Brain/metabolism , Humans , Lewy Bodies/chemistry , Lewy Bodies/metabolism , Lewy Bodies/pathology , Neurofibrillary Tangles/chemistry , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Parkinson Disease/metabolism , Plaque, Amyloid/chemistry , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Protein Aggregates , Protein Aggregation, Pathological/pathology , Protein Conformation, beta-Strand , Protein Deglycase DJ-1/chemistry
20.
ACS Chem Neurosci ; 10(12): 4847-4853, 2019 12 18.
Article in English | MEDLINE | ID: mdl-31790189

ABSTRACT

Alzheimer's disease (AD) is one of the most prevalent forms of dementia. The current diagnosis methods based on the behavior and cognitive decline or imaging of core biomarkers, namely, amyloid-ß (Aß) plaques and neurofibrillary tangles (NFTs), in the brain offer poor to moderate success. Detection and imaging of biomarkers that cause additional traits of pathophysiological aberrations in the brain are invaluable to monitor early disease onset and progression of AD pathology. The pathological hallmark of AD is associated with generation of excessive reactive oxygen species (ROS) in the brain, which aggravate oxidative stress and inflammation. ROS production involves elevated levels of hypochlorous acid (HOCl) and can serve as one of the potential biomarkers for the diagnosis of AD. We report the design, synthesis, and characterization of switchable coumarin-morpholine (CM) conjugates as off-on fluorescence probes for the specific detection of HOCl produced and proximally localized with amyloid plaques. The nonfluorescent thioamide probe CM2 undergoes regioselective transformation to fluorescent amide probe CM1 in the presence of HOCl (∼90-fold fluorescence enhancement and 0.32 quantum yield) with high selectivity and sensitivity (detection limit: 0.17 µM). The excellent cellular uptake and blood-brain barrier (BBB) crossing ability of CM2 allowed unambiguous and differential detection, imaging, and quantification of HOCl in cellular milieu and in the wild type (WT) and AD mouse brains. This study demonstrates the elevated level of HOCl in the AD mouse brain and the potential to expand the repertoire of biomarkers for the diagnosis of AD.


Subject(s)
Alzheimer Disease/metabolism , Brain Chemistry , Hypochlorous Acid/analysis , Animals , Biomarkers , Blood-Brain Barrier , Cell Line , Disease Models, Animal , Fluorescent Dyes/analysis , Fluorescent Dyes/pharmacokinetics , Male , Mice , Mice, Transgenic , Microscopy, Confocal , Microscopy, Fluorescence , Neurons/chemistry , Plaque, Amyloid/chemistry , Thioamides/analysis , Thioamides/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...