Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
2.
Mol Ther ; 32(4): 1080-1095, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38310353

ABSTRACT

Abnormal tau accumulation is the hallmark of several neurodegenerative diseases, named tauopathies. Strategies aimed at reducing tau in the brain are promising therapeutic interventions, yet more precise therapies would require targeting specific nuclei and neuronal subpopulations affected by disease while avoiding global reduction of physiological tau. Here, we developed artificial microRNAs directed against the human MAPT mRNA to dwindle tau protein by engaging the endogenous RNA interference pathway. In human differentiated neurons in culture, microRNA-mediated tau reduction diminished neuronal firing without affecting neuronal morphology or impairing axonal transport. In the htau mouse model of tauopathy, we locally expressed artificial microRNAs in the prefrontal cortex (PFC), an area particularly vulnerable to initiating tau pathology in this model. Tau knockdown prevented the accumulation of insoluble and hyperphosphorylated tau, modulated firing activity of putative pyramidal neurons, and improved glucose uptake in the PFC. Moreover, such tau reduction prevented cognitive decline in aged htau mice. Our results suggest target engagement of designed tau-microRNAs to effectively reduce tau pathology, providing a proof of concept for a potential therapeutic approach based on local tau knockdown to rescue tauopathy-related phenotypes.


Subject(s)
MicroRNAs , Tauopathies , Mice , Humans , Animals , Aged , tau Proteins/genetics , tau Proteins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Tauopathies/genetics , Tauopathies/therapy , Tauopathies/metabolism , Neurons/metabolism , Phenotype , Mice, Transgenic , Disease Models, Animal
3.
J Prev Alzheimers Dis ; 11(2): 366-374, 2024.
Article in English | MEDLINE | ID: mdl-38374743

ABSTRACT

BACKGROUND: PNT001 is a humanized full-length IgG4 S228P monoclonal antibody that binds the cis conformation of the phosphorylated Thr231-Pro232 motif in human full-length (2N4R) tau (cis-pT231 tau) with high selectivity and affinity. It binds selectively to cis-pT231 tau in human tauopathy brain sections, inhibits aggregation of tau, and has shown efficacy in preclinical models of tauopathy. Good Laboratory Practice six-month toxicology studies in cynomolgous monkeys have shown no test article-related findings. OBJECTIVES: To evaluate the safety, tolerability, pharmacokinetics, and immunogenicity of single escalating intravenous doses of PNT001 in healthy volunteers. DESIGN: Phase 1, randomized, double-blind, and placebo-controlled 16-week study. SETTING: Subjects were recruited across three clinical research sites in the United States. PARTICIPANTS: Fifty healthy volunteer subjects enrolled, with 49 receiving the double-blind study drug. INTERVENTION: Six cohorts were administered single escalating doses of PNT001 (33, 100, 300, 900, 2,700, and 4,000 mg). The subjects were randomized 6:2 (PNT001:placebo). MEASUREMENTS: Safety was evaluated by the occurrence of adverse events, electrocardiography, physical examinations, neurological examinations, vital signs, and suicidality. Pharmacokinetics and biomarkers were assessed via serum and cerebrospinal fluid sample analyses. RESULTS: Dose continuation after review of sentinel group data and dose escalation after completion of full cohort data were determined by an external, independent safety board. There were no study pauses or safety concerns identified by the safety board. A total of 49 subjects received the study drugs, with 36 receiving PNT001 and 13 receiving placebo. There were three related non-serious adverse events, each Grade 1, which occurred at the lowest doses and resolved without sequelae. No maximum tolerated dose was identified, and no premature discontinuations, dose reductions, or interruptions due to treatment-related adverse events occurred. One unrelated serious adverse event occurred in a placebo subject with an undisclosed medical condition. No other safety findings were identified. Doses of 900-4,000 mg produced concentrations in the cerebrospinal fluid exceeding the binding affinity constant of PNT001 for cis-pT231 tau (45 ng/mL), indicating that concentrations sufficient for target engagement can be obtained in the cerebrospinal fluid within the tested dose range. The serum pharmacokinetic profile was as expected for a monoclonal antibody. The terminal half-lives ranged from 23.8-33.8 days, and the cerebrospinal fluid exposures were approximately 0.1% of the plasma concentration and dose-proportional. Of the 36 subjects receiving PNT001, one post-baseline positive anti-drug antibody result was observed at Day 112 in a subject who received PNT001 (300 mg). CONCLUSIONS: Single doses of PNT001 were safe and well-tolerated at all dose levels studied, including those doses expected to produce therapeutic benefit. These results support multiple ascending dose trials in patients with neurodegenerative tauopathies for this novel mid-domain tau antibody.


Subject(s)
Antibodies, Monoclonal , Tauopathies , Humans , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Double-Blind Method , Healthy Volunteers , Tauopathies/immunology , Tauopathies/therapy , United States , tau Proteins/immunology
4.
Handb Clin Neurol ; 196: 611-619, 2023.
Article in English | MEDLINE | ID: mdl-37620094

ABSTRACT

Tauopathies are heterogeneous clinicopathological entities characterized by abnormal neuronal and/or glial inclusions of the microtubule-binding protein tau. In secondary tauopathies, i.e., Alzheimer's disease (AD), tau deposition can be observed, but tau may coexist with another protein, i.e., amyloid-ß. In the last 20 years, little progress has been made in developing disease-modifying drugs for primary and secondary tauopathies and available symptomatic drugs have limited efficacy. Treatments are being developed to interfere with the aggregation process or to promote the clearance of tau protein. Several tau-targeted passive immunotherapy approaches are in development for treating tauopathies. At present, 12 anti-tau antibodies have entered clinical trials, and 7 of them are still in clinical testing for primary tauopathies and AD (semorinemab, bepranemab, E2814, JNJ-63733657, Lu AF87908, PNT00, and APNmAb005). However, none of these seven agents have reached Phase III. The most advanced anti-tau monoclonal antibody for treating AD is semorinemab, while bepranemab is the only anti-tau monoclonal antibody still in clinical testing for treating progressive supranuclear palsy syndrome. Two other anti-tau monoclonal antibodies have been discontinued for the treatment of primary tauopathies, i.e., gosuranemab and tilavonemab. Further evidence will come from ongoing Phase I/II trials on passive immunotherapeutics for treating primary and secondary tauopathies.


Subject(s)
Alzheimer Disease , Tauopathies , Humans , tau Proteins , Tauopathies/therapy , Antibodies, Monoclonal , Immunization, Passive
5.
Int J Mol Sci ; 24(15)2023 Aug 03.
Article in English | MEDLINE | ID: mdl-37569786

ABSTRACT

Alzheimer's disease (AD) is the leading cause of dementia. No treatments have led to clinically meaningful impacts. A major obstacle for peripherally administered therapeutics targeting the central nervous system is related to the blood-brain barrier (BBB). Ultrasounds associated with microbubbles have been shown to transiently and safely open the BBB. In AD mouse models, the sole BBB opening with no adjunct drugs may be sufficient to reduce lesions and mitigate cognitive decline. However, these therapeutic effects are for now mainly assessed in preclinical mouse models of amyloidosis and remain less documented in tau lesions. The aim of the present study was therefore to evaluate the effects of repeated BBB opening using low-intensity pulsed ultrasounds (LIPU) in tau transgenic P301S mice with two main readouts: tau-positive lesions and microglial cells. Our results show that LIPU-induced BBB opening does not decrease tau pathology and may even potentiate the accumulation of pathological tau in selected brain regions. In addition, LIPU-BBB opening in P301S mice strongly reduced microglia densities in brain parenchyma, suggesting an anti-inflammatory action. These results provide a baseline for future studies using LIPU-BBB opening, such as adjunct drug therapies, in animal models and in AD patients.


Subject(s)
Alzheimer Disease , Tauopathies , Mice , Animals , Alzheimer Disease/genetics , Alzheimer Disease/therapy , Alzheimer Disease/pathology , Blood-Brain Barrier/pathology , Tauopathies/therapy , Tauopathies/pathology , Mice, Transgenic , Ultrasonic Waves
6.
J Clin Invest ; 133(12)2023 06 15.
Article in English | MEDLINE | ID: mdl-37317972

ABSTRACT

Tauopathies are disorders associated with tau protein dysfunction and insoluble tau accumulation in the brain at autopsy. Multiple lines of evidence from human disease, as well as nonclinical translational models, suggest that tau has a central pathologic role in these disorders, historically thought to be primarily related to tau gain of toxic function. However, a number of tau-targeting therapies with various mechanisms of action have shown little promise in clinical trials in different tauopathies. We review what is known about tau biology, genetics, and therapeutic mechanisms that have been tested in clinical trials to date. We discuss possible reasons for failures of these therapies, such as use of imperfect nonclinical models that do not predict human effects for drug development; heterogeneity of human tau pathologies which may lead to variable responses to therapy; and ineffective therapeutic mechanisms, such as targeting of the wrong tau species or protein epitope. Innovative approaches to human clinical trials can help address some of the difficulties that have plagued our field's development of tau-targeting therapies thus far. Despite limited clinical success to date, as we continue to refine our understanding of tau's pathogenic mechanism(s) in different neurodegenerative diseases, we remain optimistic that tau-targeting therapies will eventually play a central role in the treatment of tauopathies.


Subject(s)
Neurodegenerative Diseases , Tauopathies , Humans , Neurodegenerative Diseases/therapy , Tauopathies/therapy , Autopsy , Brain , Drug Development
7.
Rev Med Suisse ; 19(824): 817-819, 2023 Apr 26.
Article in French | MEDLINE | ID: mdl-37133943

ABSTRACT

The tauopathies are one of the families of proteinopathies causing neurodegenerative diseases. They are characterized by a combination of cognitive and motor disorders. In this article, we summarize the clinical features of progressive supranuclear palsy and cortico-basal degeneration, focusing on their cognitive-behavioral impairment profiles, which in some cases allow them to be differentiated from other neurodegenerative entities. Finally, we propose tools for therapeutic management.


Les tauopathies sont une des familles de protéinopathies engendrant des maladies neurodégénératives. Elles se caractérisent par l'association de troubles cognitifs et moteurs. Dans cet article, nous résumons les caractéristiques cliniques de la paralysie supranucléaire progressive et de la dégénérescence cortico-basale, en nous attardant sur leurs profils d'atteinte cognitivo-comportementale, qui permettent, dans certains cas, de les différencier d'autres entités neurodégénératives. Enfin, nous proposons des outils de prise en charge thérapeutique.


Subject(s)
Cognitive Dysfunction , Neurodegenerative Diseases , Supranuclear Palsy, Progressive , Tauopathies , Humans , Tauopathies/therapy , Supranuclear Palsy, Progressive/diagnosis , Supranuclear Palsy, Progressive/therapy , Neurodegenerative Diseases/therapy , Cognition , tau Proteins
8.
Science ; 379(6639): 1300-1301, 2023 03 31.
Article in English | MEDLINE | ID: mdl-36996197
9.
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
10.
J Alzheimers Dis ; 90(3): 1103-1122, 2022.
Article in English | MEDLINE | ID: mdl-36189593

ABSTRACT

BACKGROUND: Tau oligomers are one of the most toxic species, displaying prion-like strains which have different conformations resulting in different tauopathies. Passive immunotherapy targeting different tau species is a promising therapeutic approach. Age is one of the greatest risk factors; however, most immunotherapy studies are done in young to middle-aged mice tauopathy models, which is not representative of the many clinical trials done with older humans with established tauopathies. OBJECTIVE: We utilized two different clones of tau oligomer monoclonal antibodies (TOMAs) in aged Htau and JNPL3 mouse models to investigate the potential of passive immunotherapy. METHODS: Aged mice received a single intravenous injection of 120 µg/animal of either TOMA1, TOMA3 clones or a non-specific IgG. Their cognitive functions were assessed one-week post-injection using Y-maze and novel object recognition tests. Brain tissues were analyzed using biochemical and immunological assays. RESULTS: TOMA 1 and 3 rescues cognitive phenotypes in aged animals in a mouse model-specific manner, indicative by a reduction in tau oligomers levels. The TOMAs were shown to have strong reactivity with different tau oligomeric species in the different mouse models in vitro and ex vivo. CONCLUSION: This is the first study testing tau passive immunotherapy in aged animals and supports our previous reports on of the role of oligomeric tau in disease progression further validating the potential of TOMAs to rescue the late-stage disease pathology and phenotype. Moreover, this study suggests that multiple tau oligomeric strains exist in aged animals; therefore, it is of great importance to further characterize these strains.


Subject(s)
Immunization, Passive , Tauopathies , Animals , Humans , Mice , Antibodies, Monoclonal/genetics , Disease Models, Animal , Mice, Transgenic , Phenotype , tau Proteins/genetics , Tauopathies/pathology , Tauopathies/therapy
11.
Neuropeptides ; 96: 102285, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36087426

ABSTRACT

Traumatic brain injury (TBI), characterized by acute neurological impairment, is associated with a higher incidence of neurodegenerative diseases, particularly chronic traumatic encephalopathy (CTE), Alzheimer's disease (AD), and Parkinson's disease (PD), whose hallmarks include hyperphosphorylated tau protein. Recently, phosphorylated tau at Thr231 has been shown to exist in two distinct cis and trans conformations. Moreover, targeted elimination of cis P-tau by passive immunotherapy with an appropriate mAb that efficiently suppresses tau-mediated neurodegeneration in severe TBI mouse models has proven to be a useful tool to characterize the neurotoxic role of cis P-tau as an early driver of the tauopathy process after TBI. Here, we investigated whether active immunotherapy can develop sufficient neutralizing antibodies to specifically target and eliminate cis P-tau in the brain of TBI mouse models. First, we explored the therapeutic efficacy of two different vaccines. C57BL/6 J mice were immunized with either cis or trans P-tau conformational peptides plus adjuvant. After rmTBI in mice, we found that cis peptide administration developed a specific Ab that precisely targeted and neutralized cis P-tau, inhibited the development of neuropathology and brain dysfunction, and restored various structural and functional sequelae associated with TBI in chronic phases. In contrast, trans P-tau peptide application not only lacked neuroprotective properties, but also contributed to a number of neuropathological features, including progressive TBI-induced neuroinflammation, widespread tau-mediated neurodegeneration, worsening functional deficits, and brain atrophy. Taken together, our results suggest that active immunotherapy strategies against pathogenic cis P-tau can halt the process of tauopathy and would have profound clinical implications.


Subject(s)
Brain Injuries, Traumatic , Neurodegenerative Diseases , Tauopathies , Animals , Mice , Brain/metabolism , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/therapy , Brain Injuries, Traumatic/metabolism , Disease Models, Animal , Immunotherapy, Active , Mice, Inbred C57BL , tau Proteins/metabolism , Tauopathies/therapy , Tauopathies/complications , Tauopathies/metabolism
12.
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
13.
Rev Neurol (Paris) ; 178(5): 460-471, 2022 May.
Article in English | MEDLINE | ID: mdl-35562199

ABSTRACT

The two commonest groups of neurodegenerative disorders causing movement disorders are synucleinopathies and tauopathies. These disorders are characterised by the accumulation of abnormally misfolded forms of α-synuclein and tau proteins. Our current understanding of their pathogenesis suggests that extracellular forms of these proteins are of major relevance to the mechanism of pathology propagation throughout the brain and disease progression. The most novel approaches to find disease-modifying therapies aim to reduce or block these forms of tau and α-synuclein. This article reviews therapeutic strategies targeting α-synuclein and tau protein which have entered clinical development.


Subject(s)
Movement Disorders , Neurodegenerative Diseases , Tauopathies , Humans , Movement Disorders/therapy , Neurodegenerative Diseases/therapy , Tauopathies/pathology , Tauopathies/therapy , alpha-Synuclein , tau Proteins/metabolism
14.
Neuropathol Appl Neurobiol ; 48(5): e12819, 2022 08.
Article in English | MEDLINE | ID: mdl-35508761

ABSTRACT

AIM: Tau truncation (tr-tau) by active caspase-6 (aCasp-6) generates tau fragments that may be toxic. Yet the relationship between aCasp-6, different forms of tr-tau and hyperphosphorylated tau (p-tau) accumulation in human brains with Alzheimer's disease (AD) and other tauopathies remains unclear. METHODS: We generated two neoepitope monoclonal antibodies against tr-tau sites (D402 and D13) targeted by aCasp-6. Then, we used five-plex immunofluorescence to quantify the neuronal and astroglial burden of aCasp-6, tr-tau, p-tau and their co-occurrence in healthy controls, AD and primary tauopathies. RESULTS: Casp-6 activation was strongest in AD and Pick's disease (PiD) but almost absent in 4-repeat (4R) tauopathies. In neurons, the tr-tau burden was much more abundant in AD and PiD than in 4R tauopathies and disproportionally higher when normalising by p-tau pathology. Tr-tau astrogliopathy was detected in low numbers in 4R tauopathies. Unexpectedly, about half of tr-tau positive neurons in AD and PiD lacked p-tau aggregates, a finding we confirmed using several p-tau antibodies. CONCLUSIONS: Early modulation of aCasp-6 to reduce tr-tau pathology is a promising therapeutic strategy for AD and PiD but is unlikely to benefit 4R tauopathies. The large percentage of tr-tau-positive neurons lacking p-tau suggests that many vulnerable neurons to tau pathology go undetected when using conventional p-tau antibodies. Therapeutic strategies against tr-tau pathology could be necessary to modulate the extent of tau abnormalities in AD. The disproportionally higher burden of tr-tau in AD and PiD supports the development of biofluid biomarkers against tr-tau to detect AD and PiD and differentiate them from 4R tauopathies at a patient level.


Subject(s)
Alzheimer Disease , Tauopathies , Alzheimer Disease/diagnosis , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Brain/pathology , Caspase 6 , Humans , Neurons/pathology , Tauopathies/diagnosis , Tauopathies/pathology , Tauopathies/therapy , tau Proteins/metabolism
15.
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
16.
Science ; 371(6532)2021 02 26.
Article in English | MEDLINE | ID: mdl-33632820

ABSTRACT

Several lines of evidence implicate the protein tau in the pathogenesis of multiple brain disorders, including Alzheimer's disease, other neurodegenerative conditions, autism, and epilepsy. Tau is abundant in neurons and interacts with microtubules, but its main functions in the brain remain to be defined. These functions may involve the regulation of signaling pathways relevant to diverse biological processes. Informative disease models have revealed a plethora of abnormal tau species and mechanisms that might contribute to neuronal dysfunction and loss, but the relative importance of their respective contributions is uncertain. This knowledge gap poses major obstacles to the development of truly impactful therapeutic strategies. The current expansion and intensification of efforts to translate mechanistic insights into tau-related therapeutics should address this issue and could deliver better treatments for a host of devastating conditions.


Subject(s)
Brain Diseases/metabolism , Brain Diseases/therapy , Tauopathies/metabolism , Tauopathies/therapy , tau Proteins/metabolism , Animals , Brain/physiology , Humans , Microtubules/metabolism , Neurons/physiology , tau Proteins/chemistry , tau Proteins/genetics
17.
PLoS Biol ; 18(8): e3000851, 2020 08.
Article in English | MEDLINE | ID: mdl-32822389

ABSTRACT

High levels of the amyloid-beta (Aß) peptide have been shown to disrupt neuronal function and induce hyperexcitability, but it is unclear what effects Aß-associated hyperexcitability may have on tauopathy pathogenesis or propagation in vivo. Using a novel transgenic mouse line to model the impact of human APP (hAPP)/Aß accumulation on tauopathy in the entorhinal cortex-hippocampal (EC-HIPP) network, we demonstrate that hAPP overexpression aggravates EC-Tau aggregation and accelerates pathological tau spread into the hippocampus. In vivo recordings revealed a strong role for hAPP/Aß, but not tau, in the emergence of EC neuronal hyperactivity and impaired theta rhythmicity. Chronic chemogenetic attenuation of EC neuronal hyperactivity led to reduced hAPP/Aß accumulation and reduced pathological tau spread into downstream hippocampus. These data strongly support the hypothesis that in Alzheimer's disease (AD), Aß-associated hyperactivity accelerates the progression of pathological tau along vulnerable neuronal circuits, and demonstrates the utility of chronic, neuromodulatory approaches in ameliorating AD pathology in vivo.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Entorhinal Cortex/metabolism , Tauopathies/genetics , tau Proteins/genetics , Action Potentials/physiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Amyloid beta-Protein Precursor/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Electrodes, Implanted , Entorhinal Cortex/pathology , Female , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Neurons/pathology , Protein Aggregates , Stereotaxic Techniques , Tauopathies/metabolism , Tauopathies/pathology , Tauopathies/therapy , Theta Rhythm/physiology , Transduction, Genetic , Transgenes , tau Proteins/metabolism
18.
J Cell Mol Med ; 24(16): 9176-9188, 2020 08.
Article in English | MEDLINE | ID: mdl-32648620

ABSTRACT

The therapeutic hypothermia is an effective tool for TBI-associated brain impairment, but its side effects limit in clinical routine use. Hypothermia up-regulates RNA-binding motif protein 3 (RBM3), which is verified to protect synaptic plasticity. Here, we found that cognitive and LTP deficits, loss of spines, AD-like tau pathologies are displayed one month after TBI in mice. In contrast, the deficits of LTP and cognitive, loss of spines and tau abnormal phosphorylation at several sites are obviously reversed in TBI mice combined with hypothermia pre-treatment (HT). But, the neuroprotective role of HT disappears in TBI mouse models under condition of blocking RBM3 expression with RBM3 shRNA. In other hand, overexpressing RBM3 by AAV-RBM3 plasmid can mimic HT-like neuroprotection against TBI-induced chronic brain injuries, such as improving LTP and cognitive, loss of spines and tau hyperphosphorylation in TBI mouse models. Taken together, hypothermia pre-treatment reverses TBI-induced chronic AD-like pathology and behaviour deficits in RBM3 expression dependent manner, RBM3 may be a potential target for neurodegeneration diseases including Alzheimer disease.


Subject(s)
Alzheimer Disease/therapy , Behavior, Animal , Brain Injuries, Traumatic/complications , Hypothermia, Induced/methods , Neuroprotective Agents , RNA-Binding Proteins/metabolism , Tauopathies/therapy , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Male , Mice , Mice, Inbred C57BL , RNA-Binding Proteins/genetics , Tauopathies/etiology , Tauopathies/metabolism , Tauopathies/pathology
19.
Neurotherapeutics ; 17(4): 1563-1581, 2020 10.
Article in English | MEDLINE | ID: mdl-32676851

ABSTRACT

Four-repeat tauopathies are a neurodegenerative disease characterized by brain parenchymal accumulation of a specific isoform of the protein tau, which gives rise to a wide breadth of clinical syndromes encompassing diverse symptomatology, with the most common syndromes being progressive supranuclear palsy-Richardson's and corticobasal syndrome. Despite the lack of effective disease-modifying therapies, targeted treatment of symptoms can improve quality of life for patients with 4-repeat tauopathies. However, managing these symptoms can be a daunting task, even for those familiar with the diseases, as they span motor, sensory, cognitive, affective, autonomic, and behavioral domains. This review describes current approaches to symptomatic management of common clinical symptoms in 4-repeat tauopathies with a focus on practical patient management, including pharmacologic and nonpharmacologic strategies, and concludes with a discussion of the history and future of disease-modifying therapeutics and clinical trials in this population.


Subject(s)
Disease Management , Motor Disorders/diagnosis , Motor Disorders/therapy , Tauopathies/diagnosis , Tauopathies/therapy , Clinical Trials as Topic/methods , Forecasting , Humans , Motor Disorders/genetics , Tauopathies/genetics , Treatment Outcome
20.
Neuropharmacology ; 175: 108104, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32360477

ABSTRACT

The majority of clinical trials targeting the tau protein in Alzheimer's disease and other tauopathies are tau immunotherapies. Because tau pathology correlates better with the degree of dementia than amyloid-ß lesions, targeting tau is likely to be more effective in improving cognition than clearing amyloid-ß in Alzheimer's disease. However, the development of tau therapies is in many ways more complex than for amyloid-ß therapies as briefly outlined in this review. Most of the trials are on humanized antibodies, which may have very different properties than the original mouse antibodies. The impact of these differences are to a large extent unknown, can be difficult to decipher, and may not always be properly considered. Furthermore, the ideal antibody properties for efficacy are not well established and can depend on several factors. However, considering the varied approaches in clinical trials, there is a general optimism that at least some of these trials may provide functional benefits to patients suffering of various tauopathies. This article is part of the special issue entitled 'The Quest for Disease-Modifying Therapies for Neurodegenerative Disorders'.


Subject(s)
Alzheimer Disease/therapy , Immunotherapy , tau Proteins/immunology , Alzheimer Disease/immunology , Animals , Antibodies/therapeutic use , Clinical Trials as Topic , Humans , Tauopathies/immunology , Tauopathies/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...