Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters











Publication year range
1.
ACS Chem Neurosci ; 15(14): 2586-2599, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38979921

ABSTRACT

Aß oligomers are being investigated as cytotoxic agents in Alzheimer's disease (AD). Because of their transient nature and conformational heterogeneity, the relationship between the structure and activity of these oligomers is still poorly understood. Hence, methods for stabilizing Aß oligomeric species relevant to AD are needed to uncover the structural determinants of their cytotoxicity. Here, we build on the observation that metal ions and metabolites have been shown to interact with Aß, influencing its aggregation and stabilizing its oligomeric species. We thus developed a method that uses zinc ions, Zn(II), to stabilize oligomers produced by the 42-residue form of Aß (Aß42), which is dysregulated in AD. These Aß42-Zn(II) oligomers are small in size, spanning the 10-30 nm range, stable at physiological temperature, and with a broad toxic profile in human neuroblastoma cells. These oligomers offer a tool to study the mechanisms of toxicity of Aß oligomers in cellular and animal AD models.


Subject(s)
Amyloid beta-Peptides , Peptide Fragments , Zinc , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Humans , Zinc/chemistry , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Cell Line, Tumor , Alzheimer Disease/metabolism , Cell Survival/drug effects
2.
ACS Chem Neurosci ; 15(6): 1125-1134, 2024 03 20.
Article in English | MEDLINE | ID: mdl-38416693

ABSTRACT

Oligomeric assemblies of the amyloid ß peptide (Aß) have been investigated for over two decades as possible neurotoxic agents in Alzheimer's disease. However, due to their heterogeneous and transient nature, it is not yet fully established which of the structural features of these oligomers may generate cellular damage. Here, we study distinct oligomer species formed by Aß40 (the 40-residue form of Aß) in the presence of four different metal ions (Al3+, Cu2+, Fe2+, and Zn2+) and show that they differ in their structure and toxicity in human neuroblastoma cells. We then describe a correlation between the size of the oligomers and their neurotoxic activity, which provides a type of structure-toxicity relationship for these Aß40 oligomer species. These results provide insight into the possible role of metal ions in Alzheimer's disease by the stabilization of Aß oligomers.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Humans , Amyloid beta-Peptides/chemistry , Metals , Ions , Peptide Fragments/chemistry
3.
Mol Pharm ; 20(1): 183-193, 2023 01 02.
Article in English | MEDLINE | ID: mdl-36374974

ABSTRACT

The presence of amyloid fibrils of α-synuclein is closely associated with Parkinson's disease and related synucleinopathies. It is still very challenging, however, to systematically discover small molecules that prevent the formation of these aberrant aggregates. Here, we describe a structure-based approach to identify small molecules that specifically inhibit the surface-catalyzed secondary nucleation step in the aggregation of α-synuclein by binding to the surface of the amyloid fibrils. The resulting small molecules are screened using a range of kinetic and thermodynamic assays for their ability to bind α-synuclein fibrils and prevent the further generation of α-synuclein oligomers. This study demonstrates that the combination of structure-based and kinetic-based drug discovery methods can lead to the identification of small molecules that selectively inhibit the autocatalytic proliferation of α-synuclein aggregates.


Subject(s)
Parkinson Disease , alpha-Synuclein , Humans , alpha-Synuclein/metabolism , Amyloid/metabolism , Parkinson Disease/metabolism , Kinetics , Cell Proliferation , Protein Aggregates
4.
Molecules ; 27(4)2022 Feb 15.
Article in English | MEDLINE | ID: mdl-35209093

ABSTRACT

A wide variety of oligomeric structures are formed during the aggregation of proteins associated with neurodegenerative diseases. Such soluble oligomers are believed to be key toxic species in the related disorders; therefore, identification of the structural determinants of toxicity is of upmost importance. Here, we analysed toxic oligomers of α-synuclein and its pathological variants in order to identify structural features that could be related to toxicity and found a novel structural polymorphism within G51D oligomers. These G51D oligomers can adopt a variety of ß-sheet-rich structures with differing degrees of α-helical content, and the helical structural content of these oligomers correlates with the level of induced cellular dysfunction in SH-SY5Y cells. This structure-function relationship observed in α-synuclein oligomers thus presents the α-helical structure as another potential structural determinant that may be linked with cellular toxicity in amyloid-related proteins.


Subject(s)
Mutation , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism , Protein Multimerization , alpha-Synuclein/chemistry , alpha-Synuclein/genetics , Humans , Neurodegenerative Diseases , Protein Aggregates , Protein Binding , Protein Multimerization/genetics , Spectrum Analysis , alpha-Synuclein/metabolism
5.
Front Mol Biosci ; 8: 757425, 2021.
Article in English | MEDLINE | ID: mdl-34790701

ABSTRACT

The aggregation of the human islet amyloid polypeptide (IAPP) is associated with diabetes type II. A quantitative understanding of this connection at the molecular level requires that the aggregation mechanism of IAPP is resolved in terms of the underlying microscopic steps. Here we have systematically studied recombinant IAPP, with amidated C-terminus in oxidised form with a disulphide bond between residues 3 and 7, using thioflavin T fluorescence to monitor the formation of amyloid fibrils as a function of time and IAPP concentration. We used global kinetic analyses to connect the macroscopic measurements of aggregation to the microscopic mechanisms, and show that the generation of new aggregates is dominated by the secondary nucleation of monomers on the fibril surface. We then exposed insulinoma cells to aliquots extracted from different time points of the aggregation process, finding the highest toxicity at the midpoint of the reaction, when the secondary nucleation rate reaches its maximum. These results identify IAPP oligomers as the most cytotoxic species generated during IAPP aggregation, and suggest that compounds that target secondary nucleation of IAPP could be most effective as therapeutic candidates for diabetes type II.

6.
Commun Biol ; 4(1): 843, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34234268

ABSTRACT

Age-related changes in cellular metabolism can affect brain homeostasis, creating conditions that are permissive to the onset and progression of neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. Although the roles of metabolites have been extensively studied with regard to cellular signaling pathways, their effects on protein aggregation remain relatively unexplored. By computationally analysing the Human Metabolome Database, we identified two endogenous metabolites, carnosine and kynurenic acid, that inhibit the aggregation of the amyloid beta peptide (Aß) and rescue a C. elegans model of Alzheimer's disease. We found that these metabolites act by triggering a cytosolic unfolded protein response through the transcription factor HSF-1 and downstream chaperones HSP40/J-proteins DNJ-12 and DNJ-19. These results help rationalise previous observations regarding the possible anti-ageing benefits of these metabolites by providing a mechanism for their action. Taken together, our findings provide a link between metabolite homeostasis and protein homeostasis, which could inspire preventative interventions against neurodegenerative disorders.


Subject(s)
Alzheimer Disease/metabolism , Caenorhabditis elegans/metabolism , Carnosine/metabolism , Disease Models, Animal , Kynurenic Acid/metabolism , Unfolded Protein Response/physiology , Alzheimer Disease/prevention & control , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Animals , Caenorhabditis elegans Proteins/metabolism , Carnosine/pharmacology , Cytosol/metabolism , HSP40 Heat-Shock Proteins/metabolism , Humans , Kynurenic Acid/pharmacology , Protein Aggregates , Protein Aggregation, Pathological/prevention & control , Transcription Factors/metabolism , Unfolded Protein Response/drug effects
7.
Front Neurosci ; 15: 680026, 2021.
Article in English | MEDLINE | ID: mdl-34220435

ABSTRACT

The aberrant aggregation of proteins is a key molecular event in the development and progression of a wide range of neurodegenerative disorders. We have shown previously that squalamine and trodusquemine, two natural products in the aminosterol class, can modulate the aggregation of the amyloid-ß peptide (Aß) and of α-synuclein (αS), which are associated with Alzheimer's and Parkinson's diseases. In this work, we expand our previous analyses to two squalamine derivatives, des-squalamine and α-squalamine, obtaining further insights into the mechanism by which aminosterols modulate Aß and αS aggregation. We then characterize the ability of these small molecules to alter the physicochemical properties of stabilized oligomeric species in vitro and to suppress the toxicity of these aggregates to varying degrees toward human neuroblastoma cells. We found that, despite the fact that these aminosterols exert opposing effects on Aß and αS aggregation under the conditions that we tested, the modifications that they induced to the toxicity of oligomers were similar. Our results indicate that the suppression of toxicity is mediated by the displacement of toxic oligomeric species from cellular membranes by the aminosterols. This study, thus, provides evidence that aminosterols could be rationally optimized in drug discovery programs to target oligomer toxicity in Alzheimer's and Parkinson's diseases.

8.
Sci Rep ; 11(1): 14391, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34257326

ABSTRACT

Misfolded protein oligomers are increasingly recognized as highly cytotoxic agents in a wide range of human disorders associated with protein aggregation. In this study, we assessed the possible uptake and resulting toxic effects of model protein oligomers administered to C. elegans through the culture medium. We used an automated machine-vision, high-throughput screening procedure to monitor the phenotypic changes in the worms, in combination with confocal microscopy to monitor the diffusion of the oligomers, and oxidative stress assays to detect their toxic effects. Our results suggest that the oligomers can diffuse from the intestinal lumen to other tissues, resulting in a disease phenotype. We also observed that pre-incubation of the oligomers with a molecular chaperone (αB-crystallin) or a small molecule inhibitor of protein aggregation (squalamine), reduced the oligomer absorption. These results indicate that exogenous misfolded protein oligomers can be taken up by the worms from their environment and spread across tissues, giving rise to pathological effects in regions distant from their place of absorbance.


Subject(s)
Caenorhabditis elegans , Intestines , Animals , High-Throughput Screening Assays , Phenotype
10.
Immunology ; 164(2): 358-371, 2021 10.
Article in English | MEDLINE | ID: mdl-34043816

ABSTRACT

Increasing evidence indicates that peripheral immune cells play a prominent role in neurodegeneration connected to protein misfolding, which are associated with formation of aberrant aggregates, including soluble protein misfolded oligomers. The precise links, however, between the physicochemical features of diverse oligomers and their effects on the immune system, particularly on adaptive immunity, remain currently unexplored, due partly to the transient and heterogeneous nature of the oligomers themselves. To overcome these limitations, we took advantage of two stable and well-characterized types of model oligomers (A and B), formed by HypF-N bacterial protein, type B oligomers displaying lower solvent-exposed hydrophobicity. Exposure to oligomers of human peripheral blood mononuclear cells (PBMCs) revealed differential effects, with type B, but not type A, oligomers leading to a reduction in CD4+ cells. Type A oligomers promoted enhanced differentiation towards CD4+ CD25High FoxP3+ Tregs and displayed a higher suppressive effect on lymphocyte proliferation than Tregs treated with oligomers B or untreated cells. Moreover, our results reveal Th1 and Th17 lymphocyte differentiation mediated by type A oligomers and a differential balance of TGF-ß, IL-6, IL-23, IFN-γ and IL-10 mediators. These results indicate that type B oligomers recapitulate some of the biological responses associated with Parkinson's disease in peripheral immunocompetent cells, while type A oligomers resemble responses associated with Alzheimer's disease. We anticipate that further studies characterizing the differential effects of protein misfolded oligomers on the peripheral immune system may lead to the development of blood-based diagnostics, which could report on the type and properties of oligomers present in patients.


Subject(s)
Leukocytes, Mononuclear/metabolism , Proteostasis Deficiencies/metabolism , Adult , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/physiology , Cells, Cultured , Cytokines/metabolism , Humans , Lymphocyte Activation/physiology , Middle Aged , Protein Folding , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/metabolism , Th17 Cells/metabolism
11.
ACS Chem Neurosci ; 12(4): 766-781, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33538575

ABSTRACT

Alzheimer's disease, which is the most common form of dementia, is characterized by the aggregation of the amyloid ß peptide (Aß) and by an impairment of calcium homeostasis caused by excessive activation of glutamatergic receptors (excitotoxicity). Here, we studied the effects on calcium homeostasis caused by the formation of Aß oligomeric assemblies. We found that Aß oligomers cause a rapid influx of calcium ions (Ca2+) across the cell membrane by rapidly activating extrasynaptic N-methyl-d-aspartate (NMDA) receptors and, to a lower extent, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. We also observed, however, that misfolded oligomers do not interact directly with these receptors. Further experiments with lysophosphatidylcholine and arachidonic acid, which cause membrane compression and stretch, respectively, indicated that these receptors are activated through a change in membrane tension induced by the oligomers and transmitted mechanically to the receptors via the lipid bilayer. Indeed, lysophosphatidylcholine is able to neutralize the oligomer-induced activation of the NMDA receptors, whereas arachidonic acid activates the receptors similarly to the oligomers with no additive effects. An increased rotational freedom observed for a fluorescent probe embedded within the membrane in the presence of the oligomers also indicates a membrane stretch. These results reveal a mechanism of toxicity of Aß oligomers in Alzheimer's disease through the perturbation of the mechanical properties of lipid membranes sensed by NMDA and AMPA receptors.


Subject(s)
Alzheimer Disease , Receptors, N-Methyl-D-Aspartate , Amyloid beta-Peptides/metabolism , Calcium/metabolism , Homeostasis , Humans , Neurons/metabolism , Peptide Fragments/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid
13.
Commun Biol ; 4(1): 19, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33398040

ABSTRACT

Aberrant soluble oligomers formed by the amyloid-ß peptide (Aß) are major pathogenic agents in the onset and progression of Alzheimer's disease. A variety of biomolecules can influence the formation of these oligomers in the brain, although their mechanisms of action are still largely unknown. Here, we studied the effects on Aß aggregation of DOPAL, a reactive catecholaldehyde intermediate of dopamine metabolism. We found that DOPAL is able to stabilize Aß oligomeric species, including dimers and trimers, that exert toxic effects on human neuroblastoma cells, in particular increasing cytosolic calcium levels and promoting the generation of reactive oxygen species. These results reveal an interplay between Aß aggregation and key biochemical processes regulating cellular homeostasis in the brain.


Subject(s)
Alzheimer Disease/etiology , Amyloid beta-Peptides/metabolism , Dopamine/metabolism , Alzheimer Disease/metabolism , Cell Line, Tumor , Escherichia coli , Humans
14.
Int J Mol Sci ; 21(22)2020 Nov 17.
Article in English | MEDLINE | ID: mdl-33212787

ABSTRACT

The aberrant aggregation of proteins is implicated in the onset and pathogenesis of a wide range of neurodegenerative disorders, including Alzheimer's and Parkinson's diseases. Mounting evidence indicates that misfolded protein oligomers produced as intermediates in the aggregation process are potent neurotoxic agents in these diseases. Because of the transient and heterogeneous nature of these elusive aggregates, however, it has proven challenging to develop therapeutics that can effectively target them. Here, we review approaches aimed at reducing oligomer toxicity, including (1) modulating the oligomer populations (e.g., by altering the kinetics of aggregation by inhibiting, enhancing, or redirecting the process), (2) modulating the oligomer properties (e.g., through the size-hydrophobicity-toxicity relationship), (3) modulating the oligomer interactions (e.g., by protecting cell membranes by displacing oligomers), and (4) reducing oligomer toxicity by potentiating the protein homeostasis system. We analyze examples of these complementary approaches, which may lead to the development of compounds capable of preventing or treating neurodegenerative disorders associated with protein aggregation.


Subject(s)
Protein Aggregation, Pathological/therapy , Protein Multimerization , Proteostasis Deficiencies/therapy , Animals , Humans , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/pathology
15.
Sci Adv ; 6(45)2020 11.
Article in English | MEDLINE | ID: mdl-33148639

ABSTRACT

Disordered proteins are challenging therapeutic targets, and no drug is currently in clinical use that modifies the properties of their monomeric states. Here, we identify a small molecule (10074-G5) capable of binding and sequestering the intrinsically disordered amyloid-ß (Aß) peptide in its monomeric, soluble state. Our analysis reveals that this compound interacts with Aß and inhibits both the primary and secondary nucleation pathways in its aggregation process. We characterize this interaction using biophysical experiments and integrative structural ensemble determination methods. We observe that this molecule increases the conformational entropy of monomeric Aß while decreasing its hydrophobic surface area. We also show that it rescues a Caenorhabditis elegans model of Aß-associated toxicity, consistent with the mechanism of action identified from the in silico and in vitro studies. These results illustrate the strategy of stabilizing the monomeric states of disordered proteins with small molecules to alter their behavior for therapeutic purposes.


Subject(s)
Alzheimer Disease , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Drug Discovery , Humans , Hydrophobic and Hydrophilic Interactions , Peptide Fragments/metabolism
16.
Sci Rep ; 10(1): 15280, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32943652

ABSTRACT

Bicyclic peptides have great therapeutic potential since they can bridge the gap between small molecules and antibodies by combining a low molecular weight of about 2 kDa with an antibody-like binding specificity. Here we apply a recently developed in silico rational design strategy to produce a bicyclic peptide to target the C-terminal region (residues 31-42) of the 42-residue form of the amyloid ß peptide (Aß42), a protein fragment whose aggregation into amyloid plaques is linked with Alzheimer's disease. We show that this bicyclic peptide is able to remodel the aggregation process of Aß42 in vitro and to reduce its associated toxicity in vivo in a C. elegans worm model expressing Aß42. These results provide an initial example of a computational approach to design bicyclic peptides to target specific epitopes on disordered proteins.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Caenorhabditis elegans/metabolism , Protein Aggregation, Pathological/metabolism , Amyloid/metabolism , Animals , Disease Models, Animal , Peptide Fragments , Plaque, Amyloid/metabolism
17.
Commun Biol ; 3(1): 435, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32792544

ABSTRACT

The onset and progression of numerous protein misfolding diseases are associated with the presence of oligomers formed during the aberrant aggregation of several different proteins, including amyloid-ß (Aß) in Alzheimer's disease and α-synuclein (αS) in Parkinson's disease. These small, soluble aggregates are currently major targets for drug discovery. In this study, we show that trodusquemine, a naturally-occurring aminosterol, markedly reduces the cytotoxicity of αS, Aß and HypF-N oligomers to human neuroblastoma cells by displacing the oligomers from cell membranes in the absence of any substantial morphological and structural changes to the oligomers. These results indicate that the reduced toxicity results from a mechanism that is common to oligomers from different proteins, shed light on the origin of the toxicity of the most deleterious species associated with protein aggregation and suggest that aminosterols have the therapeutically-relevant potential to protect cells from the oligomer-induced cytotoxicity associated with numerous protein misfolding diseases.


Subject(s)
Cell Membrane/metabolism , Cholestanes/pharmacology , Protein Folding , Protein Multimerization , Spermine/analogs & derivatives , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/toxicity , Biophysical Phenomena/drug effects , Carboxyl and Carbamoyl Transferases/chemistry , Carboxyl and Carbamoyl Transferases/toxicity , Cell Death/drug effects , Cell Line, Tumor , Cell Membrane/drug effects , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/toxicity , Humans , Protein Folding/drug effects , Protein Multimerization/drug effects , Spermine/pharmacology , alpha-Synuclein/chemistry , alpha-Synuclein/toxicity
18.
Int J Mol Sci ; 21(12)2020 Jun 25.
Article in English | MEDLINE | ID: mdl-32630615

ABSTRACT

Alzheimer's disease is associated with the aggregation of the amyloid-ß peptide (Aß), resulting in the deposition of amyloid plaques in brain tissue. Recent scrutiny of the mechanisms by which Aß aggregates induce neuronal dysfunction has highlighted the importance of the Aß oligomers of this protein fragment. Because of the transient and heterogeneous nature of these oligomers, however, it has been challenging to investigate the detailed mechanisms by which these species exert cytotoxicity. To address this problem, we demonstrate here the use of rationally designed single-domain antibodies (DesAbs) to characterize the structure-toxicity relationship of Aß oligomers. For this purpose, we use Zn2+-stabilized oligomers of the 40-residue form of Aß (Aß40) as models of brain Aß oligomers and two single-domain antibodies (DesAb18-24 and DesAb34-40), designed to bind to epitopes at residues 18-24 and 34-40 of Aß40, respectively. We found that the DesAbs induce a change in structure of the Zn2+-stabilized Aß40 oligomers, generating a simultaneous increase in their size and solvent-exposed hydrophobicity. We then observed that these increments in both the size and hydrophobicity of the oligomers neutralize each other in terms of their effects on cytotoxicity, as predicted by a recently proposed general structure-toxicity relationship, and observed experimentally. These results illustrate the use of the DesAbs as research tools to investigate the biophysical and cytotoxicity properties of Aß oligomers.


Subject(s)
Amyloid beta-Peptides/immunology , Antibodies/immunology , Antibodies/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Antibody Formation/immunology , Brain/metabolism , Drug Design , Humans , Neurons/metabolism , Peptide Fragments/metabolism , Plaque, Amyloid/metabolism , Protein Aggregates/physiology , Protein Engineering/methods , Structure-Activity Relationship
19.
Nat Commun ; 11(1): 2945, 2020 06 10.
Article in English | MEDLINE | ID: mdl-32522983

ABSTRACT

The chemical and structural properties of biomolecules determine their interactions, and thus their functions, in a wide variety of biochemical processes. Innovative imaging methods have been developed to characterise biomolecular structures down to the angstrom level. However, acquiring vibrational absorption spectra at the single molecule level, a benchmark for bulk sample characterization, has remained elusive. Here, we introduce off-resonance, low power and short pulse infrared nanospectroscopy (ORS-nanoIR) to allow the acquisition of infrared absorption spectra and chemical maps at the single molecule level, at high throughput on a second timescale and with a high signal-to-noise ratio (~10-20). This high sensitivity enables the accurate determination of the secondary structure of single protein molecules with over a million-fold lower mass than conventional bulk vibrational spectroscopy. These results pave the way to probe directly the chemical and structural properties of individual biomolecules, as well as their interactions, in a broad range of chemical and biological systems.


Subject(s)
Nanotechnology/methods , Spectrophotometry, Infrared/methods , Biophysics , Molecular Conformation
20.
Proc Natl Acad Sci U S A ; 117(24): 13509-13518, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32493749

ABSTRACT

Protein misfolding and aggregation is the hallmark of numerous human disorders, including Alzheimer's disease. This process involves the formation of transient and heterogeneous soluble oligomers, some of which are highly cytotoxic. A major challenge for the development of effective diagnostic and therapeutic tools is thus the detection and quantification of these elusive oligomers. Here, to address this problem, we develop a two-step rational design method for the discovery of oligomer-specific antibodies. The first step consists of an "antigen scanning" phase in which an initial panel of antibodies is designed to bind different epitopes covering the entire sequence of a target protein. This procedure enables the determination through in vitro assays of the regions exposed in the oligomers but not in the fibrillar deposits. The second step involves an "epitope mining" phase, in which a second panel of antibodies is designed to specifically target the regions identified during the scanning step. We illustrate this method in the case of the amyloid ß (Aß) peptide, whose oligomers are associated with Alzheimer's disease. Our results show that this approach enables the accurate detection and quantification of Aß oligomers in vitro, and in Caenorhabditis elegans and mouse hippocampal tissues.


Subject(s)
Amyloid beta-Peptides/metabolism , Antibodies/immunology , Protein Aggregates , Alzheimer Disease/diagnosis , Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Animals , Antibodies/chemistry , Antibodies/metabolism , Antibody Specificity , Caenorhabditis elegans , Disease Models, Animal , Epitopes , Hippocampus/metabolism , Mice , Protein Binding , Protein Conformation , Single-Domain Antibodies
SELECTION OF CITATIONS
SEARCH DETAIL