Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Brain Commun ; 6(2): fcae100, 2024.
Article in English | MEDLINE | ID: mdl-38585667

ABSTRACT

Monoclonal antibodies have emerged as a leading therapeutic agent for the treatment of disease, including Alzheimer's disease. In the last year, two anti-amyloid monoclonal antibodies, lecanemab and aducanumab, have been approved in the USA for the treatment of Alzheimer's disease, whilst several tau-targeting monoclonal antibodies are currently in clinical trials. Such antibodies, however, are expensive and timely to produce and require frequent dosing regimens to ensure disease-modifying effects. Synthetic in vitro-transcribed messenger RNA encoding antibodies for endogenous protein expression holds the potential to overcome many of the limitations associated with protein antibody production. Here, we have generated synthetic in vitro-transcribed messenger RNA encoding a tau-specific antibody as a full-sized immunoglobulin and as a single-chain variable fragment. In vitro transfection of human neuroblastoma SH-SY5Y cells demonstrated the ability of the synthetic messenger RNA to be translated into a functional tau-specific antibody. Furthermore, we show that the translation of the tau-specific single-chain variable fragment as an intrabody results in the specific engagement of intracellular tau. This work highlights the utility of messenger RNA for the delivery of antibody therapeutics, including intrabodies, for the targeting of tau in Alzheimer's disease and other tauopathies.

2.
Cytoskeleton (Hoboken) ; 81(1): 24-29, 2024 01.
Article in English | MEDLINE | ID: mdl-37632370

ABSTRACT

Aggregates of the microtubule-associated protein Tau define more than a dozen primary tauopathies, and together with amyloid-ß, the secondary tauopathy Alzheimer's disease (AD). Historically, Tau has been viewed as executor of amyloid-ß toxicity, with the two molecules working together as "trigger and bullet." Given the two protein's opposing roles in protein translation, we wish to introduce another metaphor, borrowing from the mechanics of a car, with amyloid-ß boosting Tau translation, whereas Tau puts a break on global translation. The underlying studies entail an alternative hypothesis regarding Tau's subcellular accumulation in AD, namely its de novo synthesis in the somatodendritic domain rather than the relocalization from the axon upon dissociation from microtubules. We contest that it may be worth (given Tau's 50th birthday) to revisit some entrenched dogmas about Tau's pathophysiology.


Subject(s)
Alzheimer Disease , tau Proteins , Humans , Alzheimer Disease/metabolism , Axons , Microtubules/metabolism , Protein Binding , Phosphorylation
3.
J Huntingtons Dis ; 12(1): 1-13, 2023.
Article in English | MEDLINE | ID: mdl-37092231

ABSTRACT

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder. The disease, characterized by motor, cognitive, and psychiatric impairments, is caused by the expansion of a CAG repeat in the huntingtin gene. Despite the discovery of the mutation in 1993, no disease-modifying treatments are yet available. Understanding the molecular and cellular mechanisms involved in HD is therefore crucial for the development of novel treatments. Emerging research has found that HD might be classified as a secondary tauopathy, with the presence of tau insoluble aggregates in late HD. Increased total tau protein levels have been observed in both HD patients and animal models of HD. Tau hyperphosphorylation, the main feature of tau pathology, has also been investigated and our own published results suggest that the protein phosphorylation machinery is dysregulated in the early stages of HD in R6/1 transgenic mice, primarily in the cortex and striatum. Protein phosphorylation, catalysed by kinases, regulates numerous cellular mechanisms and has been shown to be dysregulated in other neurodegenerative disorders, including Alzheimer's disease. While it is still unclear how the mutation in the huntingtin gene leads to tau dysregulation in HD, several hypotheses have been explored. Evidence suggests that the mutant huntingtin does not directly interact with tau, but instead interacts with tau kinases, phosphatases, and proteins involved in tau alternative splicing, which could result in tau dysregulation as observed in HD. Altogether, there is increasing evidence that tau is undergoing pathological changes in HD and may be a good therapeutic target.


Subject(s)
Huntington Disease , tau Proteins , Animals , Mice , Alzheimer Disease , Brain/metabolism , Disease Models, Animal , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Mice, Transgenic , Neurons/metabolism
4.
Science ; 379(6639): 1300-1301, 2023 03 31.
Article in English | MEDLINE | ID: mdl-36996197
5.
Phlebology ; 38(2): 80-90, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36541140

ABSTRACT

OBJECTIVE: The study aimed to evaluate the early follow-up quality of life (QoL), pain and mental health of patients with congenital vascular malformation (CVM) from a variety of treatment options. METHODS: All patients with CVM who received care and had follow-up between February 1st 2018 and January 31st 2020 were included. The health-related QoL, pain, and mental health were assessed with RAND Health Care 36-Item Short Form Survey (SF-36), visual analogue score for pain (VAS-P) and Hospital Anxiety and Depression Scale (HADS). Paired t-test was used for all analyses. p < .05 were considered significant. RESULTS: In total, 110 patients with a mean age of 36.9 years were included in this study. In all patients following care, significant improvement was found in the bodily pain domain of SF-36 and VAS-P (both p = .01). This was largely driven by high-flow vascular malformation patients who responded better to embolo-sclerotherapy, which revealed significant improvement in the bodily pain domain of SF-36 (p = .002) and VAS-P (p = .02). Patients who received supportive treatment only reported significant improvement in mental health (p = .004) and social functioning (p = .03) domains of SF-36. Meanwhile, patients treated with embolo-sclerotherapy reported significant improvement only in VAS-P (p = .02). CONCLUSIONS: This study concluded that the effects of care on early follow-up QoL, pain and mental health of patients with CVM were heterogenous. Future research should therefore, include larger sample size and longer term follow-up to understand the various factors that affect the QoL and mental health of these patients, as well as the holistic approaches to manage them.


Subject(s)
Vascular Diseases , Vascular Malformations , Humans , Adult , Follow-Up Studies , Quality of Life/psychology , Mental Health , Vascular Diseases/therapy , Vascular Malformations/therapy , Pain , Treatment Outcome
6.
J Neurochem ; 164(4): 529-552, 2023 02.
Article in English | MEDLINE | ID: mdl-36271678

ABSTRACT

The two hallmarks of Alzheimer's disease (AD) are amyloid-ß (Aß) plaques and neurofibrillary tangles marked by phosphorylated tau. Increasing evidence suggests that aggregating Aß drives tau accumulation, a process that involves synaptic degeneration leading to cognitive impairment. Conversely, there is a realization that non-fibrillar (oligomeric) forms of Aß mediate toxicity in AD. Fibrillar (filamentous) aggregates of proteins across the spectrum of the primary and secondary tauopathies were the focus of recent structural studies with a filament structure-based nosologic classification, but less emphasis was given to non-filamentous co-aggregates of insoluble proteins in the fractions derived from post-mortem human brains. Here, we revisited sarkosyl-soluble and -insoluble extracts to characterize tau and Aß species by quantitative targeted mass spectrometric proteomics, biochemical assays, and electron microscopy. AD brain sarkosyl-insoluble pellets were greatly enriched with Aß42 at almost equimolar levels to N-terminal truncated microtubule-binding region (MTBR) isoforms of tau with multiple site-specific post-translational modifications (PTMs). MTBR R3 and R4 tau peptides were most abundant in the sarkosyl-insoluble materials with a 10-fold higher concentration than N-terminal tau peptides. This indicates that the major proportion of the enriched tau was the aggregation-prone N-terminal and proline-rich region (PRR) of truncated mixed 4R and 3R tau with more 4R than 3R isoforms. High concentration and occupancies of site-specific phosphorylation pT181 (~22%) and pT217 (~16%) (key biomarkers of AD) along with other PTMs in the PRR and MTBR indicated a regional susceptibility of PTMs in aggregated tau. Immunogold labelling revealed that tau may exist in globular non-filamentous form (N-terminal intact tau) co-localized with Aß in the sarkosyl-insoluble pellets along with tau filaments (N-truncated MTBR tau). Our results suggest a model that Aß and tau interact forming globular aggregates, from which filamentous tau and Aß emerge. These characterizations contribute towards unravelling the sequence of events which lead to end-stage AD changes.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/metabolism , Detergents/chemistry , Detergents/metabolism , Proteomics/methods , Amyloid beta-Peptides/metabolism , Brain/metabolism , Protein Isoforms/metabolism , tau Proteins/metabolism
7.
Theranostics ; 12(16): 6826-6847, 2022.
Article in English | MEDLINE | ID: mdl-36276649

ABSTRACT

Rationale: The blood-brain barrier (BBB) is a major impediment to therapeutic intracranial drug delivery for the treatment of neurodegenerative diseases, including Alzheimer's disease (AD). Focused ultrasound applied together with microbubbles (FUS+MB) is a novel technique to transiently open the BBB and increase drug delivery. Evidence suggests that FUS+MB is safe, however, the effects of FUS+MB on human BBB cells, especially in the context of AD, remain sparsely investigated. In addition, there currently are no cell platforms to test for FUS+MB-mediated drug delivery. Methods: Here we generated BBB cells (induced brain endothelial-like cells (iBECs) and astrocytes (iAstrocytes)) from apolipoprotein E gene allele E4 (APOE4, high sporadic AD risk) and allele E3 (APOE3, lower AD risk) carrying patient-derived induced pluripotent stem cells (iPSCs). We established mono- and co-culture models of human sporadic AD and control BBB cells to investigate the effects of FUS+MB on BBB cell phenotype and to screen for the delivery of two potentially therapeutic AD antibodies, an Aducanumab-analogue (AduhelmTM; anti-amyloid-ß) and a novel anti-Tau antibody, RNF5. We then developed a novel hydrogel-based 2.5D BBB model as a step towards a more physiologically relevant FUS+MB drug delivery platform. Results: When compared to untreated cells, the delivery of Aducanumab-analogue and RNF5 was significantly increased (up to 1.73 fold), across the Transwell-based BBB models following FUS+MB treatment. Our results also demonstrated the safety of FUS+MB indicated by minimal changes in iBEC transcriptome as well as little or no changes in iBEC or iAstrocyte viability and inflammatory responses within the first 24 h post FUS+MB. Furthermore, we demonstrated successful iBEC barrier formation in our novel 2.5D hydrogel-based BBB model with significantly increased delivery (1.4 fold) of Aducanumab-analogue following FUS+MB. Conclusion: Our results demonstrate a robust and reproducible approach to utilize patient cells for FUS+MB-mediated drug delivery screening in vitro. With such a cell platform for FUS+MB research previously not reported, it has the potential to identify novel FUS+MB-deliverable drugs as well as screen for cell- and patient-specific effects of FUS+MB, accelerating the use of FUS+MB as a therapeutic modality in AD.


Subject(s)
Alzheimer Disease , Antibodies, Monoclonal, Humanized , Blood-Brain Barrier , Humans , Alzheimer Disease/drug therapy , Apolipoprotein E3/metabolism , Apolipoprotein E4/metabolism , Brain/physiology , Drug Delivery Systems/methods , Hydrogels , Microbubbles , Antibodies, Monoclonal, Humanized/administration & dosage
8.
J Control Release ; 349: 634-648, 2022 09.
Article in English | MEDLINE | ID: mdl-35901857

ABSTRACT

Tau-specific immunotherapy is an attractive strategy for the treatment of Alzheimer's disease and other tauopathies. However, effectively targeting tau in the brain remains a considerable challenge due to the restrictive nature of the blood-brain barrier (BBB), which excludes an estimated >99% of peripherally administered antibodies. However, their transport across the BBB can be facilitated by a novel modality, low-intensity scanning ultrasound used in combination with intravenously injected microbubbles (SUS+MB). We have previously shown that SUS+MB-mediated delivery of a tau-specific antibody in a single-chain (scFv) format to tau transgenic mice enhanced brain and neuronal uptake and subsequently, reduced tau pathology and improved behavioural outcomes to a larger extent than either scFv or SUS+MB on its own. Here we generated a novel tau-specific monoclonal antibody, RNF5, and validated it in its IgG format in the presence or absence of SUS+MB by treating K369I tau transgenic K3 mice once weekly for 12 weeks. We found that both RNF5 and SUS+MB treatments on their own significantly reduced tau pathology. In the combination group (RNF5 + SUS+MB), however, despite increased antibody localization in the brain, there were no further reductions in tau pathology when compared to RNF5 treatment alone. Furthermore, following SUS+MB, RNF5 accumulated heavily within cells across the pyramidal cell layer of the hippocampus, that were negative for MAP2 and p-tau, suggesting that SUS+MB may not facilitate enhanced RNF5 engagement of intraneuronal tau. Overall, our new findings reveal the complexities of combining tau immunotherapy with SUS+MB and challenge the view that this is a straight-forward approach.


Subject(s)
Alzheimer Disease , Tauopathies , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Animals , Antibodies, Monoclonal , Brain/metabolism , Disease Models, Animal , Immunoglobulin G , Immunologic Factors , Membrane Proteins , Mice , Mice, Transgenic , Tauopathies/pathology , Tauopathies/therapy , Ubiquitin-Protein Ligases , tau Proteins/metabolism
9.
J Vasc Surg Venous Lymphat Disord ; 10(1): 159-169, 2022 01.
Article in English | MEDLINE | ID: mdl-33872818

ABSTRACT

OBJECTIVE: Patients with vascular malformations suffer from chronic debilitating symptoms that have been shown to contribute negatively to their quality of life (QoL) and mental health. Despite this, the current literature evaluating the QoL and mental health of patients with vascular malformations remains scarce. Our aim was to evaluate the QoL and mental health of patients with vascular malformations. METHODS: We prospectively analyzed the validated health-related QoL (HRQoL) questionnaires: the RAND Health Care 36-Item Short Form Survey (SF-36), Hospital Anxiety and Depression Scale (HADS), and visual analogue score for pain reported by 253 patients with vascular malformations in a specialist center of vascular anomalies in the UK over 2 years. RESULTS: Patients with vascular malformations reported significantly poorer SF-36 scores in all domains compared with the UK general population. Patients with low-flow vascular malformations and arteriovenous malformations reported little variations in SF-36, HADS, and visual analogue score for pain scores. No significant association was found between age and any of the health-related QoL scores, other than the physical functioning in SF-36. Female patients reported significantly lower physical and social functioning of SF-36 and worse HADS-Depression than their male counterparts. Patients with syndromic vascular malformations reported significantly lower SF-36 scores in role-physical, role-emotional and bodily pain than nonsyndromic vascular malformations. CONCLUSIONS: This study concluded that patients with vascular malformations reported worse QoL than the UK general population. Therefore, the assessment and management of QoL and mental health should be incorporated into the overall treatment strategies of patients with vascular malformations.


Subject(s)
Mental Health , Quality of Life , Vascular Malformations/psychology , Adult , Diagnostic Self Evaluation , Female , Humans , Male , Middle Aged , Prospective Studies , Self Report , United Kingdom
10.
Pharmaceutics ; 13(12)2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34959296

ABSTRACT

For the treatment of neurological diseases, achieving sufficient exposure to the brain parenchyma is a critical determinant of drug efficacy. The blood-brain barrier (BBB) functions to tightly control the passage of substances between the bloodstream and the central nervous system, and as such poses a major obstacle that must be overcome for therapeutics to enter the brain. Monoclonal antibodies have emerged as one of the best-selling treatment modalities available in the pharmaceutical market owing to their high target specificity. However, it has been estimated that only 0.1% of peripherally administered antibodies can cross the BBB, contributing to the low success rate of immunotherapy seen in clinical trials for the treatment of neurological diseases. The development of new strategies for antibody delivery across the BBB is thereby crucial to improve immunotherapeutic efficacy. Here, we discuss the current strategies that have been employed to enhance antibody delivery across the BBB. These include (i) focused ultrasound in combination with microbubbles, (ii) engineered bi-specific antibodies, and (iii) nanoparticles. Furthermore, we discuss emerging strategies such as extracellular vesicles with BBB-crossing properties and vectored antibody genes capable of being encapsulated within a BBB delivery vehicle.

11.
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
12.
Acta Neuropathol Commun ; 9(1): 42, 2021 03 12.
Article in English | MEDLINE | ID: mdl-33712083

ABSTRACT

One of the main pathological hallmarks of Alzheimer's disease (AD) is the intraneuronal accumulation of hyperphosphorylated tau. Passive immunotherapy is a promising strategy for the treatment of AD and there are currently a number of tau-specific monoclonal antibodies in clinical trials. A proposed mechanism of action is to engage and clear extracellular, pathogenic forms of tau. This process has been shown in vitro to be facilitated by microglial phagocytosis through interactions between the antibody-tau complex and microglial Fc-receptors. As this interaction is mediated by the conformation of the antibody's Fc domain, this suggests that the antibody isotype may affect the microglial phagocytosis and clearance of tau, and hence, the overall efficacy of tau antibodies. We therefore aimed to directly compare the efficacy of the tau-specific antibody, RN2N, cloned into a murine IgG1/κ framework, which has low affinity Fc-receptor binding, to that cloned into a murine IgG2a/κ framework, which has high affinity Fc-receptor binding. Our results demonstrate, for RN2N, that although enhanced microglial activation via the IgG2a/κ isotype increased extracellular tau phagocytosis in vitro, the IgG1/κ isoform demonstrated enhanced ability to reduce tau pathology and microgliosis following passive immunisation of the P301L tau transgenic pR5 mouse model.


Subject(s)
Antibodies, Monoclonal/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Tauopathies/immunology , tau Proteins/immunology , Alzheimer Disease/pathology , Animals , Brain/pathology , Disease Models, Animal , Immunization, Passive/methods , Immunoglobulin G/isolation & purification , Luminescent Measurements , Mice , Mice, Transgenic , Microglia/immunology , Microglia/pathology , Phosphorylation/genetics , Tauopathies/therapy , tau Proteins/metabolism
13.
J Vis Exp ; (161)2020 07 18.
Article in English | MEDLINE | ID: mdl-32744519

ABSTRACT

Only a small fraction of therapeutic antibodies targeting brain diseases are taken up by the brain. Focused ultrasound offers a possibility to increase uptake of antibodies and engagement through transient opening of the blood-brain barrier (BBB). In our laboratory, we are developing therapeutic approaches for neurodegenerative diseases in which an antibody in various formats is delivered across the BBB using microbubbles, concomitant with focused ultrasound application through the skull targeting multiple spots, an approach we refer to as scanning ultrasound (SUS). The mechanical effects of microbubbles and ultrasound on blood vessels increases paracellular transport across the BBB by transiently separating tight junctions and enhances vesicle- mediated transcytosis, allowing antibodies and therapeutic agents to effectively cross. Moreover, ultrasound also facilitates the uptake of antibodies from the interstitial brain into brain cells such as neurons where the antibody distributes throughout the cell body and even into neuritic processes. In our studies, fluorescently labeled antibodies are prepared, mixed with in-house prepared lipid-based microbubbles and injected into mice immediately before SUS is applied to the brain. The increased antibody concentration in the brain is then quantified. To account for alterations in normal brain homeostasis, microglial phagocytosis can be used as a cellular marker. The generated data suggest that ultrasound delivery of antibodies is an attractive approach to treat neurodegenerative diseases.


Subject(s)
Antibodies/administration & dosage , Drug Delivery Systems , Microbubbles , Animals , Blood-Brain Barrier , Fluorescent Antibody Technique , Mice , Tight Junctions , Ultrasonic Waves
14.
Sci Rep ; 9(1): 9255, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31239479

ABSTRACT

The microtubule-associated protein tau is an attractive therapeutic target for the treatment of Alzheimer's disease and related tauopathies as its aggregation strongly correlates with disease progression and is considered a key mediator of neuronal toxicity. Delivery of most therapeutics to the brain is, however, inefficient, due to their limited ability to cross the blood-brain barrier (BBB). Therapeutic ultrasound is an emerging non-invasive technology which transiently opens the BBB in a focused manner to allow peripherally delivered molecules to effectively enter the brain. In order to open a large area of the BBB, we developed a scanning ultrasound (SUS) approach by which ultrasound is applied in a sequential pattern across the whole brain. We have previously shown that delivery of an anti-tau antibody in a single-chain variable fragment (scFv) format to the brain is increased with SUS allowing for an enhanced therapeutic effect. Here we compared the delivery of an anti-tau antibody, RN2N, in an scFv, fragment antigen-binding (Fab) and full-sized immunoglobulin G (IgG) format, with and without sonication, into the brain of pR5 tau transgenic mice, a model of tauopathy. Our results revealed that the full-sized IgG reaches a higher concentration in the brain compared with the smaller formats by bypassing renal excretion. No differences in either the ultrasound-mediated uptake or distribution in the brain from the sonication site was observed across the different antibody formats, suggesting that ultrasound can be used to successfully increase the delivery of therapeutic molecules of various sizes into the brain for the treatment of neurological diseases.


Subject(s)
Antibodies, Monoclonal/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Drug Delivery Systems , Hippocampus/metabolism , Sonication/methods , tau Proteins/immunology , Animals , Mice , Mice, Transgenic
15.
Annu Rev Pathol ; 14: 239-261, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30355155

ABSTRACT

The tauopathies constitute a group of diseases that have Tau inclusions in neurons or glia as their common denominator. In this review, we describe the biochemical and histological differences in Tau pathology that are characteristic of the spectrum of frontotemporal lobar degeneration as primary tauopathies and of Alzheimer's disease as a secondary tauopathy, as well as the commonalities and differences between the familial and sporadic forms. Furthermore, we discuss selected advances in transgenic animal models in delineating the different pathomechanisms of Tau.


Subject(s)
Alzheimer Disease/pathology , Frontotemporal Dementia/pathology , Frontotemporal Lobar Degeneration/pathology , Tauopathies/pathology , Alzheimer Disease/genetics , Animals , Frontotemporal Dementia/genetics , Frontotemporal Lobar Degeneration/genetics , Humans , Neuroglia/pathology , Neurons/pathology , Tauopathies/genetics , tau Proteins/genetics
16.
J Alzheimers Dis ; 64(s1): S517-S527, 2018.
Article in English | MEDLINE | ID: mdl-29562514

ABSTRACT

Accumulation of the peptide amyloid-ß (Aß) and the protein tau in Alzheimer's disease (AD) brains is a gradual process that involves the post-translational modification and assembly of monomeric forms into larger structures that eventually form fibrillar inclusions. This process is thought to both drive and initiate AD. However, why the axonally enriched tau in the course of AD accumulates in the somatodendritic domain is not fully understood. We discuss new data that provide a possible explanation that involves de novo protein synthesis, induced by Aß and mediated through the kinase Fyn. We further discuss how in a pathological state, tau, being a scaffolding protein, impairs nuclear and mitochondrial functions and reduces action potential generation at the axon initial segment. Pathological tau can further be packaged into exosomes, released by one neuron and taken up by another, contributing to its pathogenicity. We also present our new work that suggests ultrasound as a new treatment modality to clear pathological Aß and tau. We put this work into perspective, discussing current vaccination strategies and improved brain delivery methods involving antibody engineering and viral approaches. We propose that rather than reducing post-translational modifications of tau, its levels and de novo synthesis need to be reduced. We anticipate a surge in combinatorial strategies, simultaneously targeting multiple pathologies, and an improved drug delivery to the brain facilitated by emerging technologies such as ultrasound.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , tau Proteins/metabolism , Animals , Humans
17.
J Alzheimers Dis ; 61(3): 899-905, 2018.
Article in English | MEDLINE | ID: mdl-29332046

ABSTRACT

Alzheimer's disease is characterized by two main pathological hallmarks in the human brain: the extracellular deposition of amyloid-ß as plaques and the intracellular accumulation of the hyperphosphorylated protein tau as neurofibrillary tangles (NFTs). Phosphorylated tau (p-tau) specific-antibodies and silver staining have been used to reveal three morphological stages of NFT formation: pre-NFTs, intraneuronal NFTs (iNFTs), and extraneuronal NFTs (eNFTs). Here we characterize a novel monoclonal antibody, RN235, which is specific for tau phosphorylated at serine 235, and detects iNFTs and eNFTs in brain tissue, suggesting that phosphorylation at this site is indicative of late stage changes in tau.


Subject(s)
Alzheimer Disease , Antibodies, Monoclonal/immunology , Antibody Specificity , Neurofibrillary Tangles/pathology , tau Proteins/immunology , Aged , Aged, 80 and over , Animals , Brain/pathology , Disease Models, Animal , Humans , Mice , Phosphorylation , Plaque, Amyloid/pathology , tau Proteins/chemistry
19.
Brain ; 140(5): 1220-1230, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28379300

ABSTRACT

Alzheimer's disease is characterized by the deposition of amyloid-ß as extracellular plaques and hyperphosphorylated tau as intracellular neurofibrillary tangles. Tau pathology characterizes not only Alzheimer's disease, but also many other tauopathies, presenting tau as an attractive therapeutic target. Passive tau immunotherapy has been previously explored; however, because only a small fraction of peripherally delivered antibodies crosses the blood-brain barrier, enters the brain and engages with tau that forms intracellular aggregates, more efficient ways of antibody delivery and neuronal uptake are warranted. In the brain, tau exists as multiple isoforms. Here, we investigated the efficacy of a novel 2N tau isoform-specific single chain antibody fragment, RN2N, delivered by passive immunization in the P301L human tau transgenic pR5 mouse model. We demonstrate that, in treated mice, RN2N reduces anxiety-like behaviour and phosphorylation of tau at distinct sites. When administration of RN2N was combined with focused ultrasound in a scanning mode (scanning ultrasound), RN2N delivery into the brain and uptake by neurons were markedly increased, and efficacy was significantly enhanced. Our study provides evidence that scanning ultrasound is a viable tool to enhance the delivery of biologics across the blood-brain barrier and improve therapeutic outcomes and further presents single-chain antibodies as an alternative to full-length antibodies.


Subject(s)
Antibodies/immunology , Antibodies/pharmacology , Combined Modality Therapy/methods , Tauopathies/immunology , Tauopathies/therapy , tau Proteins/immunology , Animals , Brain/immunology , Brain/metabolism , Disease Models, Animal , Humans , Immunization, Passive/psychology , Maze Learning/drug effects , Mice , Mice, Transgenic , Neurons/immunology , Neurons/metabolism , Phosphorylation/immunology , Protein Isoforms/immunology , Tauopathies/metabolism , Ultrasonic Therapy , tau Proteins/genetics , tau Proteins/metabolism
20.
J Biol Chem ; 291(15): 8173-88, 2016 Apr 08.
Article in English | MEDLINE | ID: mdl-26861879

ABSTRACT

Alternative splicing generates multiple isoforms of the microtubule-associated protein Tau, but little is known about their specific function. In the adult mouse brain, three Tau isoforms are expressed that contain either 0, 1, or 2 N-terminal inserts (0N, 1N, and 2N). We generated Tau isoform-specific antibodies and performed co-immunoprecipitations followed by tandem mass tag multiplexed quantitative mass spectrometry. We identified novel Tau-interacting proteins of which one-half comprised membrane-bound proteins, localized to the plasma membrane, mitochondria, and other organelles. Tau was also found to interact with proteins involved in presynaptic signal transduction. MetaCore analysis revealed one major Tau interaction cluster that contained 33 Tau pulldown proteins. To explore the pathways in which these proteins are involved, we conducted an ingenuity pathway analysis that revealed two significant overlapping pathways, "cell-to-cell signaling and interaction" and "neurological disease." The functional enrichment tool DAVID showed that in particular the 2N Tau-interacting proteins were specifically associated with neurological disease. Finally, for a subset of Tau interactions (apolipoprotein A1 (apoA1), apoE, mitochondrial creatine kinase U-type, ß-synuclein, synaptogyrin-3, synaptophysin, syntaxin 1B, synaptotagmin, and synapsin 1), we performed reverse co-immunoprecipitations, confirming the preferential interaction of specific isoforms. For example, apoA1 displayed a 5-fold preference for the interaction with 2N, whereas ß-synuclein showed preference for 0N. Remarkably, a reverse immunoprecipitation with apoA1 detected only the 2N isoform. This highlights distinct protein interactions of the different Tau isoforms, suggesting that they execute different functions in brain tissue.


Subject(s)
Immunoprecipitation , Protein Interaction Mapping , Protein Interaction Maps , Tauopathies/metabolism , tau Proteins/analysis , tau Proteins/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Apolipoproteins A/analysis , Apolipoproteins A/metabolism , Apolipoproteins E/analysis , Apolipoproteins E/metabolism , Mice , Mice, Inbred C57BL , Protein Isoforms/analysis , Protein Isoforms/metabolism , Tauopathies/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...