Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Acta Neuropathol Commun ; 11(1): 181, 2023 11 14.
Article in English | MEDLINE | ID: mdl-37964332

ABSTRACT

Tau seed amplification assays (SAAs) directly measure the seeding activity of tau and would therefore be ideal biomarkers for clinical trials targeting seeding-competent tau in Alzheimer's disease (AD). However, the precise relationship between tau seeding measured by SAA and the levels of pathological forms of tau in the AD brain remains unknown. We developed a new tau SAA based on full-length 0N3R tau with sensitivity in the low fg/ml range and used it to characterize 103 brain samples from three independent cohorts. Tau seeding clearly discriminated between AD and control brain samples. Interestingly, seeding was absent in Progressive Supranuclear Palsy (PSP) putamen, suggesting that our tau SAA did not amplify 4R tau aggregates from PSP brain. The specificity of our tau SAA for AD brain was further supported by analysis of matched hippocampus and cerebellum samples. While seeding was detected in hippocampus from Braak stages I-II, no seeding was present in AD cerebellum that is devoid of tau inclusions. Analysis of 40 middle frontal gyrus samples encompassing all Braak stages showed that tau SAA seeding activity gradually increased with Braak stage. This relationship between seeding activity and the presence of tau inclusions in AD brain was further supported by robust correlations between tau SAA results and the levels of phosphorylated tau212/214, phosphorylated tau181, aggregated tau, and sarkosyl-insoluble tau. Strikingly, we detected tau seeding in the middle frontal gyrus already at Braak stage II-III, suggesting that tau SAA can detect tau pathology earlier than conventional immunohistochemical staining. In conclusion, our data suggest a quantitative relationship between tau seeding activity and pathological forms of tau in the human brain and provides an important basis for further development of tau SAA for accessible human samples.


Subject(s)
Alzheimer Disease , Supranuclear Palsy, Progressive , Humans , Alzheimer Disease/pathology , tau Proteins/metabolism , Brain/pathology , Supranuclear Palsy, Progressive/pathology , Cerebellum/pathology
2.
Neurobiol Aging ; 109: 64-77, 2022 01.
Article in English | MEDLINE | ID: mdl-34655982

ABSTRACT

In Alzheimer disease, Tau pathology is thought to propagate from cell to cell throughout interconnected brain areas. However, the forms of Tau released into the brain interstitial fluid (ISF) in vivo during the development of Tauopathy and their pathological relevance remain unclear. Combining in vivo microdialysis and biochemical analysis, we find that in Tau transgenic mice, human Tau (hTau) present in brain ISF is truncated and comprises at least 10 distinct fragments spanning the entire Tau protein. The fragmentation pattern is similar across different Tau transgenic models, pathological stages and brain areas. ISF hTau concentration decreases during Tauopathy progression, while its phosphorylation increases. ISF from mice with established Tauopathy induces Tau aggregation in HEK293-Tau biosensor cells. Notably, immunodepletion of ISF phosphorylated Tau, but not Tau fragments, significantly reduces its ability to seed Tau aggregation and only a fraction of Tau, separated by ultracentrifugation, is seeding-competent. These results indicate that ISF seeding competence is driven by a small subset of Tau, which potentially contribute to the propagation of Tau pathology.


Subject(s)
Brain/metabolism , Extracellular Fluid/metabolism , Tauopathies/metabolism , tau Proteins/metabolism , Animals , Disease Models, Animal , HEK293 Cells , Humans , Mice, Transgenic , Microdialysis , Peptide Fragments/metabolism , Phosphorylation , Protein Aggregation, Pathological/metabolism
3.
MAbs ; 13(1): 1887628, 2021.
Article in English | MEDLINE | ID: mdl-33596779

ABSTRACT

ABT-736 is a humanized monoclonal antibody generated to target a specific conformation of the amyloid-beta (Aß) protein oligomer. Development of ABT-736 for Alzheimer's disease was discontinued due to severe adverse effects (AEs) observed in cynomolgus monkey toxicity studies. The acute nature of AEs observed only at the highest doses suggested potential binding of ABT-736 to an abundant plasma protein. Follow-up investigations indicated polyspecificity of ABT-736, including unintended high-affinity binding to monkey and human plasma protein platelet factor 4 (PF-4), known to be involved in heparin-induced thrombocytopenia (HIT) in humans. The chronic AEs observed at the lower doses after repeat administration in monkeys were consistent with HIT pathology. Screening for a backup antibody revealed that ABT-736 possessed additional unintended binding characteristics to other, unknown factors. A subsequently implemented screening funnel focused on nonspecific binding led to the identification of h4D10, a high-affinity Aß oligomer binding antibody that did not bind PF-4 or other unintended targets and had no AEs in vivo. This strengthened the hypothesis that ABT-736 toxicity was not Aß target-related, but instead was the consequence of polyspecificity including PF-4 binding, which likely mediated the acute and chronic AEs and the HIT-like pathology. In conclusion, thorough screening of antibody candidates for nonspecific interactions with unrelated molecules at early stages of discovery can eliminate candidates with polyspecificity and reduce potential for toxicity caused by off-target binding.


Subject(s)
Alzheimer Vaccines/immunology , Amyloid beta-Peptides/antagonists & inhibitors , Antibodies, Monoclonal, Humanized/toxicity , Blood Platelets/drug effects , Immunity, Heterologous , Platelet Factor 4/antagonists & inhibitors , Purpura, Thrombocytopenic, Idiopathic/chemically induced , Alzheimer Vaccines/pharmacokinetics , Alzheimer Vaccines/toxicity , Amyloid beta-Peptides/immunology , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibody Specificity , Blood Platelets/immunology , Blood Platelets/metabolism , Female , Humans , Macaca fascicularis , Male , Mice, Inbred BALB C , No-Observed-Adverse-Effect Level , Platelet Activation/drug effects , Platelet Factor 4/immunology , Purpura, Thrombocytopenic, Idiopathic/blood , Purpura, Thrombocytopenic, Idiopathic/immunology , Risk Assessment , Time Factors , Toxicity Tests, Acute , Toxicity Tests, Chronic
4.
J Neurosci ; 30(31): 10369-79, 2010 Aug 04.
Article in English | MEDLINE | ID: mdl-20685980

ABSTRACT

Oligomers of the beta-amyloid (Abeta) peptide have been indicated in early neuropathologic changes in Alzheimer's disease. Here, we present a synthetic Abeta(20-42) oligomer (named globulomer) with a different conformation to monomeric and fibrillar Abeta peptide, enabling the generation of highly Abeta oligomer-specific monoclonal antibodies. The globulomer-derived antibodies specifically detect oligomeric but not monomeric or fibrillar Abeta in various Abeta preparations. The globulomer-specific antibody A-887755 was able to prevent Abeta oligomer binding and dynamin cleavage in primary hippocampal neurons and to reverse globulomer-induced reduced synaptic transmission. In amyloid precursor protein (APP) transgenic mice, vaccination with Abeta globulomer and treatment with A-887755 improved novel object recognition. The cognitive improvement is likely attributable to reversing a deficit in hippocampal synaptic spine density in APP transgenic mice as observed after treatment with A-887755. Our findings demonstrate that selective reduction of Abeta oligomers by immunotherapy is sufficient to normalize cognitive behavior and synaptic deficits in APP transgenic mice.


Subject(s)
Amyloid beta-Peptides/immunology , Amyloid beta-Protein Precursor/genetics , Antibodies, Monoclonal/therapeutic use , Alzheimer Disease/drug therapy , Alzheimer Disease/immunology , Analysis of Variance , Animals , Antibodies, Monoclonal/immunology , Cells, Cultured , Disease Models, Animal , Female , Hippocampus/cytology , Hippocampus/immunology , Immunoprecipitation , Male , Mice , Mice, Transgenic , Neurons/cytology , Neurons/immunology , Rats , Rats, Wistar , Recognition, Psychology
5.
Neurobiol Dis ; 30(2): 212-20, 2008 May.
Article in English | MEDLINE | ID: mdl-18353662

ABSTRACT

Soluble A beta-oligomers are currently discussed as the major causative species for the development of Alzheimer's disease (AD). Consequently, the beta-amyloid cascade hypothesis was extended by A beta-oligomers and their central neuropathogenic role in AD. However, the molecular structure of A beta-oligomers and their relation to amyloid fibril formation remains elusive. Previously we demonstrated that incubation of A beta(1-42) with SDS or fatty acids induces the formation of a homogeneous globular A beta-oligomer termed A beta-globulomer. In this study we investigated the role of A beta-globulomers in the aggregation pathway of A beta-peptide. We used in vitro assays such as thioflavin-T binding and aggregation inhibitors like Congo red to reveal that A beta-peptide in its A beta-globulomer conformation is a structural entity which is independent from amyloid fibril formation. In addition, cellular Alzheimer's-like plaque forming assays show the resistance of A beta-globulomers to deposition as amyloid plaques. We hypothesize that a conformational switch of A beta is decisive for either fibril formation or alternatively and independently A beta-globulomer formation.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid/metabolism , Peptide Fragments/metabolism , Plaque, Amyloid/metabolism , Signal Transduction/physiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid/chemistry , Amyloid/physiology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/physiology , Animals , Astrocytes/chemistry , Astrocytes/metabolism , Astrocytes/pathology , Cells, Cultured , Mice , Mice, Inbred C57BL , Neurofibrillary Tangles/chemistry , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Peptide Fragments/chemistry , Peptide Fragments/physiology , Plaque, Amyloid/chemistry , Protein Conformation
6.
J Neurochem ; 95(3): 834-47, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16135089

ABSTRACT

Amyloid beta-peptide (Abeta)(1-42) oligomers have recently been discussed as intermediate toxic species in Alzheimer's disease (AD) pathology. Here we describe a new and highly stable Abeta(1-42) oligomer species which can easily be prepared in vitro and is present in the brains of patients with AD and Abeta(1-42)-overproducing transgenic mice. Physicochemical characterization reveals a pure, highly water-soluble globular 60-kDa oligomer which we named 'Abeta(1-42) globulomer'. Our data indicate that Abeta(1-42) globulomer is a persistent structural entity formed independently of the fibrillar aggregation pathway. It is a potent antigen in mice and rabbits eliciting generation of Abeta(1-42) globulomer-specific antibodies that do not cross-react with amyloid precursor protein, Abeta(1-40) and Abeta(1-42) monomers and Abeta fibrils. Abeta(1-42) globulomer binds specifically to dendritic processes of neurons but not glia in hippocampal cell cultures and completely blocks long-term potentiation in rat hippocampal slices. Our data suggest that Abeta(1-42) globulomer represents a basic pathogenic structural principle also present to a minor extent in previously described oligomer preparations and that its formation is an early pathological event in AD. Selective neutralization of the Abeta globulomer structure epitope is expected to have a high potential for treatment of AD.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Neurons/metabolism , Neurons/pathology , Peptide Fragments/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/immunology , Animals , Antibodies , Antibody Specificity , Cells, Cultured , Epitopes/chemistry , Epitopes/immunology , Epitopes/metabolism , Fatty Acids , Hippocampus/cytology , Humans , Long-Term Potentiation , Male , Mice , Mice, Transgenic , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/immunology , Protein Conformation , Rabbits , Rats , Rats, Sprague-Dawley , Solubility , Water/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...