Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Alzheimers Dement ; 20(2): 1013-1025, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37849026

ABSTRACT

INTRODUCTION: Signatures of a type-I interferon (IFN-I) response are observed in the post mortem brain in Alzheimer's disease (AD) and other tauopathies. However, the effect of the IFN-I response on pathological tau accumulation remains unclear. METHODS: We examined the effects of IFN-I signaling in primary neural culture models of seeded tau aggregation and P301S-tau transgenic mouse models in the context of genetic deletion of the IFN-I receptor (IFNAR). RESULTS: Polyinosinic:polycytidylic acid (PolyI:C), a synthetic analog of viral nucleic acids, evoked a potent cytokine response that enhanced seeded aggregation of tau in an IFN-I-dependent manner. IFN-I-induced vulnerability could be pharmacologically prevented and was intrinsic to neurons. Aged P301S-tau mice lacking Ifnar1 had significantly reduced tau pathology compared to mice with intact IFN signaling. DISCUSSION: We identify a critical role for IFN-I in potentiating tau aggregation. IFN-I is therefore identified as a potential therapeutic target in AD and other tauopathies. HIGHLIGHTS: Type-I IFN (IFN-I) promotes seeded tau aggregation in neural cultures. IFNAR inhibition prevents IFN-I driven sensitivity to tau aggregation. IFN-I driven vulnerability is intrinsic to neurons. Tau pathology is significantly reduced in aged P301S-tau mice lacking IFNAR.


Subject(s)
Alzheimer Disease , Interferon Type I , Tauopathies , Mice , Animals , tau Proteins/genetics , Interferon Type I/therapeutic use , Tauopathies/pathology , Mice, Transgenic , Alzheimer Disease/pathology , Disease Models, Animal
2.
Cell Rep ; 42(7): 112725, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37393617

ABSTRACT

Tau is a soluble protein interacting with tubulin to stabilize microtubules. However, under pathological conditions, it becomes hyperphosphorylated and aggregates, a process that can be induced by treating cells with exogenously added tau fibrils. Here, we employ single-molecule localization microscopy to resolve the aggregate species formed in early stages of seeded tau aggregation. We report that entry of sufficient tau assemblies into the cytosol induces the self-replication of small tau aggregates, with a doubling time of 5 h inside HEK cells and 1 day in murine primary neurons, which then grow into fibrils. Seeding occurs in the vicinity of the microtubule cytoskeleton, is accelerated by the proteasome, and results in release of small assemblies into the media. In the absence of seeding, cells still spontaneously form small aggregates at lower levels. Overall, our work provides a quantitative picture of the early stages of templated seeded tau aggregation in cells.


Subject(s)
Alzheimer Disease , tau Proteins , Mice , Animals , tau Proteins/metabolism , Microtubules/metabolism , Tubulin/metabolism , Cytosol/metabolism , Neurons/metabolism , Alzheimer Disease/metabolism , Protein Aggregates
3.
Science ; 380(6651): 1258-1265, 2023 06 23.
Article in English | MEDLINE | ID: mdl-37347855

ABSTRACT

During initiation of antiviral and antitumor T cell-mediated immune responses, dendritic cells (DCs) cross-present exogenous antigens on major histocompatibility complex (MHC) class I molecules. Cross-presentation relies on the unusual "leakiness" of endocytic compartments in DCs, whereby internalized proteins escape into the cytosol for proteasome-mediated generation of MHC I-binding peptides. Given that type 1 conventional DCs excel at cross-presentation, we searched for cell type-specific effectors of endocytic escape. We devised an assay suitable for genetic screening and identified a pore-forming protein, perforin-2 (Mpeg1), as a dedicated effector exclusive to cross-presenting cells. Perforin-2 was recruited to antigen-containing compartments, where it underwent maturation, releasing its pore-forming domain. Mpeg1-/- mice failed to efficiently prime CD8+ T cells to cell-associated antigens, revealing an important role for perforin-2 in cytosolic entry of antigens during cross-presentation.


Subject(s)
Antigen Presentation , CD8-Positive T-Lymphocytes , Endocytosis , Pore Forming Cytotoxic Proteins , Animals , Mice , Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Cross-Priming/genetics , Cross-Priming/immunology , Dendritic Cells/immunology , Endocytosis/genetics , Endocytosis/immunology , Genetic Testing , Histocompatibility Antigens Class I , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/metabolism , Proteolysis
4.
Science ; 379(6639): 1336-1341, 2023 03 31.
Article in English | MEDLINE | ID: mdl-36996217

ABSTRACT

Aggregates of the protein tau are proposed to drive pathogenesis in neurodegenerative diseases. Tau can be targeted by using passively transferred antibodies (Abs), but the mechanisms of Ab protection are incompletely understood. In this work, we used a variety of cell and animal model systems and showed that the cytosolic Ab receptor and E3 ligase TRIM21 (T21) could play a role in Ab protection against tau pathology. Tau-Ab complexes were internalized to the cytosol of neurons, which enabled T21 engagement and protection against seeded aggregation. Ab-mediated protection against tau pathology was lost in mice that lacked T21. Thus, the cytosolic compartment provides a site of immunotherapeutic protection, which may help in the design of Ab-based therapies in neurodegenerative disease.


Subject(s)
Antibodies, Monoclonal , Immunization, Passive , Ribonucleoproteins , Tauopathies , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , tau Proteins , Animals , Mice , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Cytosol/metabolism , Disease Models, Animal , Receptors, Fc , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , tau Proteins/immunology , Tauopathies/therapy , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
5.
Cell Rep ; 39(5): 110776, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35508140

ABSTRACT

Assemblies of tau can transit between neurons, seeding aggregation in a prion-like manner. To accomplish this, tau must cross cell-limiting membranes, a process that is poorly understood. Here, we establish assays for the study of tau entry into the cytosol as a phenomenon distinct from uptake, in real time, and at physiological concentrations. The entry pathway of tau is cell type specific and, in neurons, highly sensitive to cholesterol. Depletion of the cholesterol transporter Niemann-Pick type C1 or extraction of membrane cholesterol renders neurons highly permissive to tau entry and potentiates seeding even at low levels of exogenous tau assemblies. Conversely, cholesterol supplementation reduces entry and almost completely blocks seeded aggregation. Our findings establish entry as a rate-limiting step to seeded aggregation and demonstrate that dysregulated cholesterol, a feature of several neurodegenerative diseases, potentiates tau aggregation by promoting entry of tau assemblies into the cell interior.


Subject(s)
Alzheimer Disease , Prions , Alzheimer Disease/metabolism , Cholesterol/metabolism , Cytosol/metabolism , Humans , Neurons/metabolism , Prions/metabolism , tau Proteins/metabolism
6.
Acta Neuropathol Commun ; 9(1): 41, 2021 03 12.
Article in English | MEDLINE | ID: mdl-33712082

ABSTRACT

A fundamental property of infectious agents is their particulate nature: infectivity arises from independently-acting particles rather than as a result of collective action. Assemblies of the protein tau can exhibit seeding behaviour, potentially underlying the apparent spread of tau aggregation in many neurodegenerative diseases. Here we ask whether tau assemblies share with classical pathogens the characteristic of particulate behaviour. We used organotypic hippocampal slice cultures from P301S tau transgenic mice in order to precisely control the concentration of extracellular tau assemblies in neural tissue. Whilst untreated slices displayed no overt signs of pathology, exposure to recombinant tau assemblies could result in the formation of intraneuronal, hyperphosphorylated tau structures. However, seeding ability of tau assemblies did not titrate in a one-hit manner in neural tissue. The results suggest that seeding behaviour of tau arises at high concentrations, with implications for the interpretation of high-dose intracranial challenge experiments and the possible contribution of seeded aggregation to human disease.


Subject(s)
Prions/pathogenicity , Protein Aggregation, Pathological/pathology , Protein Aggregation, Pathological/physiopathology , Tauopathies/pathology , Tauopathies/physiopathology , tau Proteins/metabolism , Alzheimer Disease , Animals , Disease Models, Animal , HEK293 Cells , Hippocampus/metabolism , Hippocampus/pathology , Humans , In Vitro Techniques , Mice , Mice, Transgenic , Phosphorylation , Protein Aggregation, Pathological/metabolism , Tauopathies/metabolism , Tissue Culture Techniques , tau Proteins/genetics
7.
Front Immunol ; 10: 1139, 2019.
Article in English | MEDLINE | ID: mdl-31214163

ABSTRACT

Ordered assemblies of proteins are found in the postmortem brains of sufferers of several neurodegenerative diseases. The cytoplasmic microtubule associated protein tau and alpha-synuclein (αS) are found in an assembled state in Alzheimer's disease and Parkinson's disease, respectively. An accumulating body of evidence suggests a "prion-like" mechanism of spread of these assemblies through the diseased brain. Under this hypothesis, assembled variants of these proteins promote the conversion of native proteins to the assembled state. This likely inflicts pathology on cells of the brain through a toxic gain-of-function mechanism. Experiments in animal models of tau and αS pathology have demonstrated that the passive transfer of anti-tau or anti-αS antibodies induces a reduction in the levels of assembled proteins. This is further accompanied by improvements in neurological function and preservation of brain volume. Immunotherapy is therefore considered one of the brightest hopes as a therapeutic avenue in an area currently without disease-modifying therapy. Following a series of disappointing clinical trials targeting beta-amyloid, a peptide that accumulates in the extracellular spaces of the AD brain, attention is turning to active and passive immunotherapies that target tau and αS. However, there are several remaining uncertainties concerning the mechanism by which antibodies afford protection against self-propagating protein conformations. This review will discuss current understanding of how antibodies and their receptors can be brought to bear on proteins involved in neurodegeneration. Parallels will be made to antibody-mediated protection against classical viral infections. Common mechanisms that may contribute to protection against self-propagating protein conformations include blocking the entry of protein "seeds" to cells, clearance of immune complexes by microglia, and the intracellular protein degradation pathway initiated by cytoplasmic antibodies via the Fc receptor TRIM21. As with anti-viral immunity, protective mechanisms may be accompanied by the activation of immune signaling pathways and we will discuss the suitability of such activation in the neurological setting.


Subject(s)
Autoantibodies/metabolism , Brain/metabolism , Immunotherapy/methods , Neurodegenerative Diseases/immunology , Vaccines/immunology , alpha-Synuclein/metabolism , tau Proteins/metabolism , Animals , Brain/pathology , Disease Models, Animal , Humans , Neurodegenerative Diseases/therapy
SELECTION OF CITATIONS
SEARCH DETAIL