Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.330
Filtrer
1.
Sci Transl Med ; 16(754): eadj5958, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38959324

RÉSUMÉ

Pathological tau aggregates cause cognitive decline in neurodegenerative tauopathies, including Alzheimer's disease (AD). These aggregates are prevalent within intracellular compartments. Current tau immunotherapies have shown limited efficacy in clearing intracellular tau aggregates and improving cognition in clinical trials. In this study, we developed toxic tau conformation-specific monoclonal antibody-2 (TTCM2), which selectively recognized pathological tau aggregates in brain tissues from patients with AD, dementia with Lewy bodies (DLB), and progressive supranuclear palsy (PSP). TTCM2 potently inhibited tau-seeding activity, an essential mechanism underlying tauopathy progression. To effectively target intracellular tau aggregates and ensure rapid delivery to the brain, TTCM2 was loaded in micelles (TTCM2-ms) and administered through the intranasal route. We found that intranasally administered TTCM2-ms efficiently entered the brain in hTau-tauopathy mice, targeting pathological tau in intracellular compartments. Moreover, a single intranasal dose of TTCM2-ms effectively cleared pathological tau, elevated synaptic proteins, and improved cognitive functions in aged tauopathy mice. Mechanistic studies revealed that TTCM2-ms cleared intracellular, synaptic, and seed-competent tau aggregates through tripartite motif-containing 21 (TRIM21), an intracellular antibody receptor and E3 ubiquitin ligase known to facilitate proteasomal degradation of cytosolic antibody-bound proteins. TRIM21 was found to be essential for TTCM2-ms-mediated clearance of tau pathology. Our study collectively provides evidence of the effectiveness of nasal tau immunotherapy in targeting and clearing intracellular tau pathology through TRIM21 and enhancing cognition in aged tauopathy mice. This study could be valuable in designing effective tau immunotherapies for AD and other tauopathies.


Sujet(s)
Administration par voie nasale , Cognition , Immunothérapie , Souris transgéniques , Tauopathies , Protéines tau , Animaux , Protéines tau/métabolisme , Tauopathies/thérapie , Tauopathies/anatomopathologie , Tauopathies/métabolisme , Immunothérapie/méthodes , Humains , Souris , Vieillissement/anatomopathologie , Encéphale/anatomopathologie , Encéphale/métabolisme , Anticorps monoclonaux/pharmacologie , Modèles animaux de maladie humaine , Agrégats de protéines/effets des médicaments et des substances chimiques
2.
Sci Transl Med ; 16(754): eadq6489, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38959325

RÉSUMÉ

Nasal delivery of an oligomeric tau antibody loaded into micelles reduces pathology and ameliorates cognition in a mouse model of tauopathy.


Sujet(s)
Administration par voie nasale , Tauopathies , Protéines tau , Animaux , Protéines tau/métabolisme , Souris , Tauopathies/métabolisme , Tauopathies/anatomopathologie , Humains , Modèles animaux de maladie humaine , Micelles , Nez , Anticorps/usage thérapeutique , Anticorps/immunologie
3.
Adv Protein Chem Struct Biol ; 141: 467-493, 2024.
Article de Anglais | MEDLINE | ID: mdl-38960483

RÉSUMÉ

In Alzheimer's disease, the microtubule-associated protein, Tau misfolds to form aggregates and filaments in the intra- and extracellular region of neuronal cells. Microglial cells are the resident brain macrophage cells involved in constant surveillance and activated by the extracellular deposits. Purinergic receptors are involved in the chemotactic migration of microglial cells towards the site of inflammation. From our recent study, we have observed that the microglial P2Y12 receptor is involved in phagocytosis of full-length Tau species such as monomers, oligomers and aggregates by actin-driven chemotaxis. This study shows the interaction of repeat-domain of Tau (TauRD) with the microglial P2Y12 receptor and the corresponding residues for interaction have been analyzed by various in-silico approaches. In the cellular studies, TauRD was found to interact with microglial P2Y12R and induces its cellular expression confirmed by co-immunoprecipitation and western blot analysis. Furthermore, the P2Y12R-mediated TauRD internalization has demonstrated activation of microglia with an increase in the Iba1 level, and TauRD becomes accumulated at the peri-nuclear region for the degradation.


Sujet(s)
Tauopathies , Humains , Tauopathies/métabolisme , Tauopathies/anatomopathologie , Protéines tau/métabolisme , Microglie/métabolisme , Microglie/anatomopathologie , Récepteurs purinergiques P2Y12/métabolisme , Animaux , Récepteurs couplés aux protéines G/métabolisme
4.
Cell Mol Life Sci ; 81(1): 304, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39009859

RÉSUMÉ

The autophagy-lysosomal pathway plays a critical role in the clearance of tau protein aggregates that deposit in the brain in tauopathies, and defects in this system are associated with disease pathogenesis. Here, we report that expression of Tau35, a tauopathy-associated carboxy-terminal fragment of tau, leads to lipid accumulation in cell lines and primary cortical neurons. Our findings suggest that this is likely due to a deleterious block of autophagic clearance and lysosomal degradative capacity by Tau35. Notably, upon induction of autophagy by Torin 1, Tau35 inhibited nuclear translocation of transcription factor EB (TFEB), a key regulator of lysosomal biogenesis. Both cell lines and primary cortical neurons expressing Tau35 also exhibited changes in endosomal protein expression. These findings implicate autophagic and endolysosomal dysfunction as key pathological mechanisms through which disease-associated tau fragments could lead to the development and progression of tauopathy.


Sujet(s)
Autophagie , Endosomes , Métabolisme lipidique , Lysosomes , Neurones , Protéines tau , Protéines tau/métabolisme , Protéines tau/génétique , Lysosomes/métabolisme , Humains , Neurones/métabolisme , Animaux , Endosomes/métabolisme , Tauopathies/métabolisme , Tauopathies/anatomopathologie , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/métabolisme , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/génétique , Souris
5.
Cells ; 13(13)2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38994964

RÉSUMÉ

Aggregation of the microtubule-associated protein tau (MAPT) is the hallmark pathology in a spectrum of neurodegenerative disorders collectively called tauopathies. Physiologically, tau is an inherent neuronal protein that plays an important role in the assembly of microtubules and axonal transport. However, disease-associated mutations of this protein reduce its binding to the microtubule components and promote self-aggregation, leading to formation of tangles in neurons. Tau is also expressed in oligodendrocytes, where it has significant developmental roles in oligodendrocyte maturation and myelin synthesis. Oligodendrocyte-specific tau pathology, in the form of fibrils and coiled coils, is evident in major tauopathies including progressive supranuclear palsy (PSP), corticobasal degeneration (CBD), and Pick's disease (PiD). Multiple animal models of tauopathy expressing mutant forms of MAPT recapitulate oligodendroglial tau inclusions with potential to cause degeneration/malfunction of oligodendrocytes and affecting the neuronal myelin sheath. Till now, mechanistic studies heavily concentrated on elucidating neuronal tau pathology. Therefore, more investigations are warranted to comprehensively address tau-induced pathologies in oligodendrocytes. The present review provides the current knowledge available in the literature about the intricate relations between tau and oligodendrocytes in health and diseases.


Sujet(s)
Oligodendroglie , Tauopathies , Protéines tau , Humains , Tauopathies/métabolisme , Tauopathies/anatomopathologie , Oligodendroglie/métabolisme , Oligodendroglie/anatomopathologie , Animaux , Protéines tau/métabolisme , Maladies neurodégénératives/métabolisme , Maladies neurodégénératives/anatomopathologie
6.
Nat Commun ; 15(1): 4706, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38830849

RÉSUMÉ

The neuromodulatory subcortical nuclei within the isodendritic core (IdC) are the earliest sites of tauopathy in Alzheimer's disease (AD). They project broadly throughout the brain's white matter. We investigated the relationship between IdC microstructure and whole-brain white matter microstructure to better understand early neuropathological changes in AD. Using multiparametric quantitative magnetic resonance imaging we observed two covariance patterns between IdC and white matter microstructure in 133 cognitively unimpaired older adults (age 67.9 ± 5.3 years) with familial risk for AD. IdC integrity related to 1) whole-brain neurite density, and 2) neurite orientation dispersion in white matter tracts known to be affected early in AD. Pattern 2 was associated with CSF concentration of phosphorylated-tau, indicating AD specificity. Apolipoprotein-E4 carriers expressed both patterns more strongly than non-carriers. IdC microstructure variation is reflected in white matter, particularly in AD-affected tracts, highlighting an early mechanism of pathological development.


Sujet(s)
Maladie d'Alzheimer , Imagerie par résonance magnétique , Tauopathies , Substance blanche , Protéines tau , Humains , Substance blanche/imagerie diagnostique , Substance blanche/anatomopathologie , Substance blanche/métabolisme , Femelle , Mâle , Sujet âgé , Adulte d'âge moyen , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/liquide cérébrospinal , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/imagerie diagnostique , Tauopathies/imagerie diagnostique , Tauopathies/métabolisme , Tauopathies/anatomopathologie , Tauopathies/génétique , Tauopathies/liquide cérébrospinal , Protéines tau/métabolisme , Protéines tau/liquide cérébrospinal , Encéphale/anatomopathologie , Encéphale/imagerie diagnostique , Encéphale/métabolisme , Apolipoprotéines E/génétique , Apolipoprotéines E/métabolisme , Apolipoprotéine E4/génétique , Apolipoprotéine E4/métabolisme , Neurites/métabolisme , Neurites/anatomopathologie
7.
Cell Death Dis ; 15(6): 429, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38890273

RÉSUMÉ

Tauopathies are characterised by the pathological accumulation of misfolded tau. The emerging view is that toxic tau species drive synaptic dysfunction and potentially tau propagation before measurable neurodegeneration is evident, but the underlying molecular events are not well defined. Human non-mutated 0N4R tau (tauWT) and P301L mutant 0N4R tau (tauP301L) were expressed in mouse primary cortical neurons using adeno-associated viruses to monitor early molecular changes and synaptic function before the onset of neuronal loss. In this model tauP301L was differentially phosphorylated relative to tauwt with a notable increase in phosphorylation at ser262. Affinity purification - mass spectrometry combined with tandem mass tagging was used to quantitatively compare the tauWT and tauP301L interactomes. This revealed an enrichment of tauP301L with ribosomal proteins but a decreased interaction with the proteasome core complex and reduced tauP301L degradation. Differences in the interaction of tauP301L with members of a key synaptic calcium-calmodulin signalling pathway were also identified, most notably, increased association with CaMKII but reduced association with calcineurin and the candidate AD biomarker neurogranin. Decreased association of neurogranin to tauP301L corresponded with the appearance of enhanced levels of extracellular neurogranin suggestive of potential release or leakage from synapses. Finally, analysis of neuronal network activity using micro-electrode arrays showed that overexpression of tauP301L promoted basal hyperexcitability coincident with these changes in the tau interactome and implicating tau in specific early alterations in synaptic function.


Sujet(s)
Neurones , Protéines tau , Animaux , Protéines tau/métabolisme , Protéines tau/génétique , Humains , Souris , Neurones/métabolisme , Phosphorylation , Calcium-Calmodulin-Dependent Protein Kinase Type 2/métabolisme , Calcium-Calmodulin-Dependent Protein Kinase Type 2/génétique , Tauopathies/métabolisme , Tauopathies/anatomopathologie , Tauopathies/génétique , Synapses/métabolisme , Neurogranine/métabolisme , Neurogranine/génétique , Calcineurine/métabolisme
8.
Mol Neurodegener ; 19(1): 51, 2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-38915105

RÉSUMÉ

BACKGROUND: Tau is aberrantly acetylated in various neurodegenerative conditions, including Alzheimer's disease, frontotemporal lobar degeneration (FTLD), and traumatic brain injury (TBI). Previously, we reported that reducing acetylated tau by pharmacologically inhibiting p300-mediated tau acetylation at lysine 174 reduces tau pathology and improves cognitive function in animal models. METHODS: We investigated the therapeutic efficacy of two different antibodies that specifically target acetylated lysine 174 on tau (ac-tauK174). We treated PS19 mice, which harbor the P301S tauopathy mutation that causes FTLD, with anti-ac-tauK174 and measured effects on tau pathology, neurodegeneration, and neurobehavioral outcomes. Furthermore, PS19 mice received treatment post-TBI to evaluate the ability of the immunotherapy to prevent TBI-induced exacerbation of tauopathy phenotypes. Ac-tauK174 measurements in human plasma following TBI were also collected to establish a link between trauma and acetylated tau levels, and single nuclei RNA-sequencing of post-TBI brain tissues from treated mice provided insights into the molecular mechanisms underlying the observed treatment effects. RESULTS: Anti-ac-tauK174 treatment mitigates neurobehavioral impairment and reduces tau pathology in PS19 mice. Ac-tauK174 increases significantly in human plasma 24 h after TBI, and anti-ac-tauK174 treatment of PS19 mice blocked TBI-induced neurodegeneration and preserved memory functions. Anti-ac-tauK174 treatment rescues alterations of microglial and oligodendrocyte transcriptomic states following TBI in PS19 mice. CONCLUSIONS: The ability of anti-ac-tauK174 treatment to rescue neurobehavioral impairment, reduce tau pathology, and rescue glial responses demonstrates that targeting tau acetylation at K174 is a promising neuroprotective therapeutic approach to human tauopathies resulting from TBI or genetic disease.


Sujet(s)
Tauopathies , Protéines tau , Animaux , Tauopathies/métabolisme , Protéines tau/métabolisme , Souris , Acétylation , Humains , Immunothérapie/méthodes , Modèles animaux de maladie humaine , Souris transgéniques , Lésions traumatiques de l'encéphale/métabolisme , Lésions encéphaliques/métabolisme , Encéphale/métabolisme , Encéphale/anatomopathologie , Neuroprotecteurs/pharmacologie
9.
Brain ; 147(7): 2428-2439, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38842726

RÉSUMÉ

Four-repeat (4R) tauopathies are neurodegenerative diseases characterized by cerebral accumulation of 4R tau pathology. The most prominent 4R tauopathies are progressive supranuclear palsy (PSP) and corticobasal degeneration characterized by subcortical tau accumulation and cortical neuronal dysfunction, as shown by PET-assessed hypoperfusion and glucose hypometabolism. Yet, there is a spatial mismatch between subcortical tau deposition patterns and cortical neuronal dysfunction, and it is unclear how these two pathological brain changes are interrelated. Here, we hypothesized that subcortical tau pathology induces remote neuronal dysfunction in functionally connected cortical regions to test a pathophysiological model that mechanistically links subcortical tau accumulation to cortical neuronal dysfunction in 4R tauopathies. We included 51 Aß-negative patients with clinically diagnosed PSP variants (n = 26) or corticobasal syndrome (n = 25) who underwent structural MRI and 18F-PI-2620 tau-PET. 18F-PI-2620 tau-PET was recorded using a dynamic one-stop-shop acquisition protocol to determine an early 0.5-2.5 min post tracer-injection perfusion window for assessing cortical neuronal dysfunction, as well as a 20-40 min post tracer-injection window to determine 4R-tau load. Perfusion-PET (i.e. early window) was assessed in 200 cortical regions, and tau-PET was assessed in 32 subcortical regions of established functional brain atlases. We determined tau epicentres as subcortical regions with the highest 18F-PI-2620 tau-PET signal and assessed the connectivity of tau epicentres to cortical regions of interest using a resting-state functional MRI-based functional connectivity template derived from 69 healthy elderly controls from the ADNI cohort. Using linear regression, we assessed whether: (i) higher subcortical tau-PET was associated with reduced cortical perfusion; and (ii) cortical perfusion reductions were observed preferentially in regions closely connected to subcortical tau epicentres. As hypothesized, higher subcortical tau-PET was associated with overall lower cortical perfusion, which remained consistent when controlling for cortical tau-PET. Using group-average and subject-level PET data, we found that the seed-based connectivity pattern of subcortical tau epicentres aligned with cortical perfusion patterns, where cortical regions that were more closely connected to the tau epicentre showed lower perfusion. Together, subcortical tau-accumulation is associated with remote perfusion reductions indicative of neuronal dysfunction in functionally connected cortical regions in 4R-tauopathies. This suggests that subcortical tau pathology may induce cortical dysfunction, which may contribute to clinical disease manifestation and clinical heterogeneity.


Sujet(s)
Cortex cérébral , Tomographie par émission de positons , Paralysie supranucléaire progressive , Tauopathies , Protéines tau , Humains , Mâle , Femelle , Tomographie par émission de positons/méthodes , Sujet âgé , Tauopathies/imagerie diagnostique , Tauopathies/métabolisme , Tauopathies/anatomopathologie , Protéines tau/métabolisme , Adulte d'âge moyen , Cortex cérébral/imagerie diagnostique , Cortex cérébral/métabolisme , Cortex cérébral/anatomopathologie , Paralysie supranucléaire progressive/imagerie diagnostique , Paralysie supranucléaire progressive/métabolisme , Paralysie supranucléaire progressive/anatomopathologie , Paralysie supranucléaire progressive/physiopathologie , Imagerie par résonance magnétique/méthodes
10.
J Neurol Sci ; 462: 123079, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38878650

RÉSUMÉ

BACKGROUND: Reactive astrocytes play an important role in the development of Alzheimer's disease and primary tauopathies. Here, we aimed to investigate the relationships between reactive astrocytes. Microgliosis and glucose metabolism with Tau and amyloid beta pathology by using multi-tracer imaging in widely used tauopathy and familial Alzheimer's disease mouse models. RESULTS: Positron emission tomography imaging using [18F]PM-PBB3 (tau), [18F]florbetapir (amyloid-beta), [18F]SMBT-1 (monoamine oxidase-B), [18F]DPA-714 (translocator protein) and [18F]fluorodeoxyglucose was carried out in 3- and 7-month-old rTg4510 tau mice, 5 × FAD familial Alzheimer's disease mice and wild-type mice. Immunofluorescence staining was performed to validate the pathological distribution in the mouse brain after in vivo imaging. We found increased regional levels of [18F]PM-PBB3, [18F]SMBT-1, and [18F]DPA-714 and hypoglucose metabolism in the brains of 7-month-old rTg4510 mice compared to age-matched wild-type mice. Increased [18F]SMBT-1 uptake was observed in the brains of 3, 7-month-old 5 × FAD mice, with elevated regional [18F]florbetapir and [18F]DPA-714 uptakes in the brains of 7-month-old 5 × FAD mice, compared to age-matched wild-type mice. Positive correlations were shown between [18F]SMBT-1 and [18F]PM-PBB3, [18F]DPA-714 and [18F]PM-PBB3 in rTg4510 mice, and between [18F]florbetapir and [18F]DPA-714 SUVRs in 5 × FAD mice. CONCLUSION: In summary, these findings provide in vivo evidence that reactive astrocytes, microglial activation, and cerebral hypoglucose metabolism are associated with tau and amyloid pathology development in animal models of tauopathy and familial Alzheimer's disease.


Sujet(s)
Maladie d'Alzheimer , Astrocytes , Encéphale , Modèles animaux de maladie humaine , Souris transgéniques , Tomographie par émission de positons , Tauopathies , Animaux , Maladie d'Alzheimer/imagerie diagnostique , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/génétique , Astrocytes/métabolisme , Tomographie par émission de positons/méthodes , Tauopathies/imagerie diagnostique , Tauopathies/métabolisme , Tauopathies/anatomopathologie , Souris , Encéphale/imagerie diagnostique , Encéphale/métabolisme , Encéphale/anatomopathologie , Radio-isotopes du fluor , Mâle , Peptides bêta-amyloïdes/métabolisme , Humains , Protéines tau/métabolisme , Protéines tau/génétique , Fluorodésoxyglucose F18 , Radiopharmaceutiques
11.
Proc Natl Acad Sci U S A ; 121(27): e2311831121, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38941274

RÉSUMÉ

TMEM16F is a calcium-activated phospholipid scramblase and nonselective ion channel, which allows the movement of lipids bidirectionally across the plasma membrane. While the functions of TMEM16F have been extensively characterized in multiple cell types, the role of TMEM16F in the central nervous system remains largely unknown. Here, we sought to study how TMEM16F in the brain may be involved in neurodegeneration. Using a mouse model that expresses the pathological P301S human tau (PS19 mouse), we found reduced tauopathy and microgliosis in 6- to 7-mo-old PS19 mice lacking TMEM16F. Furthermore, this reduction of pathology can be recapitulated in the PS19 mice with TMEM16F removed from neurons, while removal of TMEM16F from microglia of PS19 mice did not significantly impact tauopathy at this time point. Moreover, TMEM16F mediated aberrant phosphatidylserine exposure in neurons with phospho-tau burden. These studies raise the prospect of targeting TMEM16F in neurons as a potential treatment of neurodegeneration.


Sujet(s)
Anoctamines , Neurones , Phosphatidylsérine , Tauopathies , Protéines tau , Animaux , Anoctamines/métabolisme , Anoctamines/génétique , Phosphatidylsérine/métabolisme , Neurones/métabolisme , Neurones/anatomopathologie , Protéines tau/métabolisme , Protéines tau/génétique , Souris , Tauopathies/métabolisme , Tauopathies/anatomopathologie , Humains , Microglie/métabolisme , Microglie/anatomopathologie , Phosphorylation , Souris transgéniques , Modèles animaux de maladie humaine , Protéines de transfert des phospholipides/métabolisme , Protéines de transfert des phospholipides/génétique , Encéphale/métabolisme , Encéphale/anatomopathologie , Souris knockout
12.
eNeuro ; 11(6)2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38858068

RÉSUMÉ

Sleep disruption and impaired synaptic processes are common features in neurodegenerative diseases, including Alzheimer's disease (AD). Hyperphosphorylated Tau is known to accumulate at neuronal synapses in AD, contributing to synapse dysfunction. However, it remains unclear how sleep disruption and synapse pathology interact to contribute to cognitive decline. Here, we examined sex-specific onset and consequences of sleep loss in AD/tauopathy model PS19 mice. Using a piezoelectric home-cage monitoring system, we showed PS19 mice exhibited early-onset and progressive hyperarousal, a selective dark-phase sleep disruption, apparent at 3 months in females and 6 months in males. Using the Morris water maze test, we report that chronic sleep disruption (CSD) accelerated the onset of decline of hippocampal spatial memory in PS19 males only. Hyperarousal occurs well in advance of robust forebrain synaptic Tau burden that becomes apparent at 6-9 months. To determine whether a causal link exists between sleep disruption and synaptic Tau hyperphosphorylation, we examined the correlation between sleep behavior and synaptic Tau, or exposed mice to acute or chronic sleep disruption at 6 months. While we confirm that sleep disruption is a driver of Tau hyperphosphorylation in neurons of the locus ceruleus, we were unable to show any causal link between sleep loss and Tau burden in forebrain synapses. Despite the finding that hyperarousal appears earlier in females, female cognition was resilient to the effects of sleep disruption. We conclude sleep disruption interacts with the synaptic Tau burden to accelerate the onset of cognitive decline with greater vulnerability in males.


Sujet(s)
Dysfonctionnement cognitif , Modèles animaux de maladie humaine , Souris transgéniques , Prosencéphale , Synapses , Protéines tau , Animaux , Protéines tau/métabolisme , Mâle , Femelle , Dysfonctionnement cognitif/métabolisme , Dysfonctionnement cognitif/étiologie , Synapses/métabolisme , Synapses/anatomopathologie , Souris , Prosencéphale/métabolisme , Caractères sexuels , Tauopathies/métabolisme , Tauopathies/anatomopathologie , Troubles de la veille et du sommeil/métabolisme , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Souris de lignée C57BL
13.
Nat Commun ; 15(1): 5109, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38877019

RÉSUMÉ

Positron emission tomography (PET) imaging of tau aggregation in Alzheimer's disease (AD) is helping to map and quantify the in vivo progression of AD pathology. To date, no high-affinity tau-PET radiopharmaceutical has been optimized for imaging non-AD tauopathies. Here we show the properties of analogues of a first-in-class 4R-tau lead, [18F]OXD-2115, using ligand-based design. Over 150 analogues of OXD-2115 were synthesized and screened in post-mortem brain tissue for tau affinity against [3H]OXD-2115, and in silico models were used to predict brain uptake. [18F]OXD-2314 was identified as a selective, high-affinity non-AD tau PET radiotracer with favorable brain uptake, dosimetry, and radiometabolite profiles in rats and non-human primate and is being translated for first-in-human PET studies.


Sujet(s)
Maladie d'Alzheimer , Encéphale , Radio-isotopes du fluor , Tomographie par émission de positons , Radiopharmaceutiques , Tauopathies , Protéines tau , Tomographie par émission de positons/méthodes , Animaux , Humains , Tauopathies/imagerie diagnostique , Tauopathies/métabolisme , Encéphale/imagerie diagnostique , Encéphale/métabolisme , Ligands , Radiopharmaceutiques/composition chimique , Radiopharmaceutiques/pharmacocinétique , Radiopharmaceutiques/synthèse chimique , Rats , Maladie d'Alzheimer/imagerie diagnostique , Maladie d'Alzheimer/métabolisme , Radio-isotopes du fluor/composition chimique , Protéines tau/métabolisme , Mâle
14.
Alzheimers Dement ; 20(7): 4985-4998, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38923171

RÉSUMÉ

INTRODUCTION: A noncoding variant (rs35349669) within INPP5D, a lipid and protein phosphatase restricted to microglia in the brain, is linked to increased susceptibility to Alzheimer's disease (AD). While Inpp5d is well-studied in amyloid pathology, its role in tau pathology remains unclear. METHODS: PS19 Tauopathy mice were crossed with Inpp5d-haplodeficient (Inpp5d+/-) mice to examine the impact of Inpp5d in tau pathology. RESULTS: Increased INPP5D expression correlated positively with phospho-Tau AT8 in PS19 mice. Inpp5d haplodeficiency mitigated hyperphosphorylated tau levels (AT8, AT180, AT100, and PHF1) and motor deficits in PS19 mice. Transcriptomic analysis revealed an up-regulation of genes associated with immune response and cell migration. DISCUSSION: Our findings define an association between INPP5D expression and tau pathology in PS19 mice. Alleviation in hyperphosphorylated tau, motor deficits, and transcriptomics changes in haplodeficient-Inpp5d PS19 mice indicate that modulation in INPP5D expression may provide therapeutic potential for mitigating tau pathology and improving motor deficits. HIGHLIGHTS: The impact of Inpp5d in the context of tau pathology was studied in the PS19 mouse model. INPP5D expression is associated with tau pathology. Reduced Inpp5d expression in PS19 mice improved motor functions and decreased total and phospho-Tau levels. Inpp5d haplodeficiency in PS19 mice modulates gene expression patterns linked to immune response and cell migration. These data suggest that inhibition of Inpp5d may be a therapeutic approach in tauopathies.


Sujet(s)
Modèles animaux de maladie humaine , Souris transgéniques , Tauopathies , Protéines tau , Animaux , Tauopathies/anatomopathologie , Tauopathies/métabolisme , Tauopathies/génétique , Souris , Protéines tau/métabolisme , Encéphale/anatomopathologie , Encéphale/métabolisme , Phosphorylation
15.
Sci Rep ; 14(1): 11533, 2024 05 21.
Article de Anglais | MEDLINE | ID: mdl-38773170

RÉSUMÉ

Tauopathies, including Alzheimer's disease and Frontotemporal Dementia, are debilitating neurodegenerative disorders marked by cognitive decline. Despite extensive research, achieving effective treatments and significant symptom management remains challenging. Accurate diagnosis is crucial for developing effective therapeutic strategies, with hyperphosphorylated protein units and tau oligomers serving as reliable biomarkers for these conditions. This study introduces a novel approach using nanotechnology to enhance the diagnostic process for tauopathies. We developed humanized ferritin nanocages, a novel nanoscale delivery system, designed to encapsulate and transport a tau-specific fluorophore, BT1, into human retinal cells for detecting neurofibrillary tangles in retinal tissue, a key marker of tauopathies. The delivery of BT1 into living cells was successfully achieved through these nanocages, demonstrating efficient encapsulation and delivery into retinal cells derived from human induced pluripotent stem cells. Our experiments confirmed the colocalization of BT1 with pathological forms of tau in living retinal cells, highlighting the method's potential in identifying tauopathies. Using ferritin nanocages for BT1 delivery represents a significant contribution to nanobiotechnology, particularly in neurodegenerative disease diagnostics. This method offers a promising tool for the early detection of tau tangles in retinal tissue, with significant implications for improving the diagnosis and management of tauopathies. This study exemplifies the integration of nanotechnology with biomedical science, expanding the frontiers of nanomedicine and diagnostic techniques.


Sujet(s)
Ferritines , Rétine , Tauopathies , Protéines tau , Humains , Protéines tau/métabolisme , Ferritines/métabolisme , Rétine/métabolisme , Rétine/anatomopathologie , Tauopathies/métabolisme , Tauopathies/anatomopathologie , Tauopathies/diagnostic , Cellules souches pluripotentes induites/métabolisme , Enchevêtrements neurofibrillaires/métabolisme , Enchevêtrements neurofibrillaires/anatomopathologie
17.
J Neuroinflammation ; 21(1): 130, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38750510

RÉSUMÉ

Epidemiological studies have unveiled a robust link between exposure to repetitive mild traumatic brain injury (r-mTBI) and elevated susceptibility to develop neurodegenerative disorders, notably chronic traumatic encephalopathy (CTE). The pathogenic lesion in CTE cases is characterized by the accumulation of hyperphosphorylated tau in neurons around small cerebral blood vessels which can be accompanied by astrocytes that contain phosphorylated tau, the latter termed tau astrogliopathy. However, the contribution of tau astrogliopathy to the pathobiology and functional consequences of r-mTBI/CTE or whether it is merely a consequence of aging remains unclear. We addressed these pivotal questions by utilizing a mouse model harboring tau-bearing astrocytes, GFAPP301L mice, subjected to our r-mTBI paradigm. Despite the fact that r-mTBI did not exacerbate tau astrogliopathy or general tauopathy, it increased phosphorylated tau in the area underneath the impact site. Additionally, gene ontology analysis of tau-bearing astrocytes following r-mTBI revealed profound alterations in key biological processes including immunological and mitochondrial bioenergetics. Moreover, gene array analysis of microdissected astrocytes accrued from stage IV CTE human brains revealed an immunosuppressed astroglial phenotype similar to tau-bearing astrocytes in the GFAPP301L model. Additionally, hippocampal reduction of proteins involved in water transport (AQP4) and glutamate homeostasis (GLT1) was found in the mouse model of tau astrogliopathy. Collectively, these findings reveal the importance of understanding tau astrogliopathy and its role in astroglial pathobiology under normal circumstances and following r-mTBI. The identified mechanisms using this GFAPP301L model may suggest targets for therapeutic interventions in r-mTBI pathogenesis in the context of CTE.


Sujet(s)
Aquaporine-4 , Astrocytes , Transporteur-2 d'acides aminés excitateurs , Souris transgéniques , Tauopathies , Protéines tau , Astrocytes/métabolisme , Astrocytes/anatomopathologie , Animaux , Souris , Protéines tau/métabolisme , Protéines tau/génétique , Aquaporine-4/métabolisme , Aquaporine-4/génétique , Tauopathies/métabolisme , Tauopathies/anatomopathologie , Tauopathies/génétique , Humains , Transporteur-2 d'acides aminés excitateurs/métabolisme , Transporteur-2 d'acides aminés excitateurs/génétique , Transporteur-2 d'acides aminés excitateurs/biosynthèse , Commotion de l'encéphale/métabolisme , Commotion de l'encéphale/anatomopathologie , Mâle , Phénotype , Souris de lignée C57BL
18.
ACS Chem Neurosci ; 15(11): 2112-2120, 2024 06 05.
Article de Anglais | MEDLINE | ID: mdl-38776461

RÉSUMÉ

Neuroinflammation plays an important role in Alzheimer's disease and primary tauopathies. The aim of the current study was to map [18F]GSK1482160 for imaging of purinergic P2X7R in Alzheimer's disease and primary tauopathy mouse models. Small animal PET was performed using [18F]GSK1482160 in widely used mouse models of Alzheimer's disease (APP/PS1, 5×FAD, and 3×Tg), 4-repeat tauopathy (rTg4510) mice, and age-matched wild-type mice. Increased uptake of [18F]GSK1482160 was observed in the brains of 7-month-old rTg4510 mice compared to wild-type mice and compared to 3-month-old rTg4510 mice. A positive correlation between hippocampal tau [18F]APN-1607 and [18F]GSK1482160 uptake was found in rTg4510 mice. No significant differences in the uptake of [18F]GSK1482160 was observed for APP/PS1 mice, 5×FAD mice, or 3×Tg mice. Immunofluorescence staining further indicated the distribution of P2X7Rs in the brains of 7-month-old rTg4510 mice with accumulation of tau inclusion. These findings provide in vivo imaging evidence for an increased level of P2X7R in the brains of tauopathy mice.


Sujet(s)
Tomographie par émission de positons , Récepteurs purinergiques P2X7 , Tauopathies , Animaux , Souris , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/imagerie diagnostique , Encéphale/métabolisme , Encéphale/imagerie diagnostique , Modèles animaux de maladie humaine , Radio-isotopes du fluor , Souris transgéniques , Tomographie par émission de positons/méthodes , Récepteurs purinergiques P2X7/métabolisme , Protéines tau/métabolisme , Tauopathies/imagerie diagnostique , Tauopathies/métabolisme
19.
Cell Death Dis ; 15(5): 333, 2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38740758

RÉSUMÉ

Precise polyamine metabolism regulation is vital for cells and organisms. Mutations in spermine synthase (SMS) cause Snyder-Robinson intellectual disability syndrome (SRS), characterized by significant spermidine accumulation and autophagy blockage in the nervous system. Emerging evidence connects polyamine metabolism with other autophagy-related diseases, such as Tauopathy, however, the functional intersection between polyamine metabolism and autophagy in the context of these diseases remains unclear. Here, we altered SMS expression level to investigate the regulation of autophagy by modulated polyamine metabolism in Tauopathy in Drosophila and human cellular models. Interestingly, while complete loss of Drosophila spermine synthase (dSms) impairs lysosomal function and blocks autophagic flux recapitulating SRS disease phenotype, partial loss of dSms enhanced autophagic flux, reduced Tau protein accumulation, and led to extended lifespan and improved climbing performance in Tauopathy flies. Measurement of polyamine levels detected a mild elevation of spermidine in flies with partial loss of dSms. Similarly, in human neuronal or glial cells, partial loss of SMS by siRNA-mediated knockdown upregulated autophagic flux and reduced Tau protein accumulation. Importantly, proteomics analysis of postmortem brain tissue from Alzheimer's disease (AD) patients showed a significant albeit modest elevation of SMS level. Taken together, our study uncovers a functional correlation between polyamine metabolism and autophagy in AD: SMS reduction upregulates autophagy, suppresses Tau accumulation, and ameliorates neurodegeneration and cell death. These findings provide a new potential therapeutic target for AD.


Sujet(s)
Autophagie , Spermine synthase , Protéines tau , Animaux , Protéines tau/métabolisme , Humains , Spermine synthase/métabolisme , Spermine synthase/génétique , Drosophila melanogaster/métabolisme , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Tauopathies/métabolisme , Tauopathies/anatomopathologie , Neurones/métabolisme , Neurones/anatomopathologie , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/génétique , Spermidine/métabolisme , Modèles animaux de maladie humaine , Lysosomes/métabolisme , Drosophila/métabolisme , Retard mental lié à l'X
20.
J Math Biol ; 89(1): 4, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38750128

RÉSUMÉ

A system of partial differential equations is developed to study the spreading of tau pathology in the brain for Alzheimer's and other neurodegenerative diseases. Two cases are considered with one assuming intracellular diffusion through synaptic activities or the nanotubes that connect the adjacent cells. The other, in addition to intracellular spreading, takes into account of the secretion of the tau species which are able to diffuse, move with the interstitial fluid flow and subsequently taken up by the surrounding cells providing an alternative pathway for disease spreading. Cross membrane transport of the tau species are considered enabling us to examine the role of extracellular clearance of tau protein on the disease status. Bifurcation analysis is carried out for the steady states of the spatially homogeneous system yielding the results that fast cross-membrane transport combined with effective extracellular clearance is key to maintain the brain's healthy status. Numerical simulations of the first case exhibit solutions of travelling wave form describing the gradual outward spreading of the pathology; whereas the second case shows faster spreading with the buildup of neurofibrillary tangles quickly elevated throughout. Our investigation thus indicates that the gradual progression of the intracellular spreading case is more consistent with the clinical observations of the development of Alzheimer's disease.


Sujet(s)
Maladie d'Alzheimer , Encéphale , Simulation numérique , Concepts mathématiques , Maladies neurodégénératives , Protéines tau , Protéines tau/métabolisme , Humains , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Maladies neurodégénératives/métabolisme , Maladies neurodégénératives/anatomopathologie , Encéphale/métabolisme , Encéphale/anatomopathologie , Modèles neurologiques , Enchevêtrements neurofibrillaires/métabolisme , Enchevêtrements neurofibrillaires/anatomopathologie , Modèles biologiques , Évolution de la maladie , Tauopathies/métabolisme , Tauopathies/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...