Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.315
Filter
1.
Sci Transl Med ; 16(754): eadj5958, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38959324

ABSTRACT

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.


Subject(s)
Administration, Intranasal , Cognition , Immunotherapy , Mice, Transgenic , Tauopathies , tau Proteins , Animals , tau Proteins/metabolism , Tauopathies/therapy , Tauopathies/pathology , Tauopathies/metabolism , Immunotherapy/methods , Humans , Mice , Aging/pathology , Brain/pathology , Brain/metabolism , Antibodies, Monoclonal/pharmacology , Disease Models, Animal , Protein Aggregates/drug effects
2.
Sci Transl Med ; 16(754): eadq6489, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38959325

ABSTRACT

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


Subject(s)
Administration, Intranasal , Tauopathies , tau Proteins , Animals , tau Proteins/metabolism , Mice , Tauopathies/metabolism , Tauopathies/pathology , Humans , Disease Models, Animal , Micelles , Nose , Antibodies/therapeutic use , Antibodies/immunology
3.
Cells ; 13(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38994964

ABSTRACT

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.


Subject(s)
Oligodendroglia , Tauopathies , tau Proteins , Humans , Tauopathies/metabolism , Tauopathies/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Animals , tau Proteins/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology
4.
JASA Express Lett ; 4(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38980136

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder in which changes in hearing sensitivity precede cognitive decline. Despite a well-known link between dementia and hearing loss, few AD model mouse lines have hearing characterized. We screened for hearing loss using auditory brainstem responses (ABR) in young (3-4 months) and aging (9-10 months) mice with a P301S tauopathy (PS19 mice). Compared to wild types, aging PS19 mice did not show accelerated hearing loss but did show latency differences in centrally generated ABR waveform components. These results suggest that tauopathy causes mild central auditory dysfunction in the absence of overt hearing loss.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem , Tauopathies , Animals , Alzheimer Disease/physiopathology , Mice , Tauopathies/physiopathology , Tauopathies/pathology , Mice, Transgenic , Hearing Loss/physiopathology , Hearing Loss/etiology , Humans , Audiometry/methods
5.
Cell Mol Life Sci ; 81(1): 304, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39009859

ABSTRACT

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.


Subject(s)
Autophagy , Endosomes , Lipid Metabolism , Lysosomes , Neurons , tau Proteins , tau Proteins/metabolism , tau Proteins/genetics , Lysosomes/metabolism , Humans , Neurons/metabolism , Animals , Endosomes/metabolism , Tauopathies/metabolism , Tauopathies/pathology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Mice
6.
Adv Protein Chem Struct Biol ; 141: 467-493, 2024.
Article in English | MEDLINE | ID: mdl-38960483

ABSTRACT

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.


Subject(s)
Tauopathies , Humans , Tauopathies/metabolism , Tauopathies/pathology , tau Proteins/metabolism , Microglia/metabolism , Microglia/pathology , Receptors, Purinergic P2Y12/metabolism , Animals , Receptors, G-Protein-Coupled/metabolism
7.
Neuropathol Appl Neurobiol ; 50(4): e13000, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39036836

ABSTRACT

AIMS: Astrocytic tau pathology is a major feature of tauopathies and ageing-related tau astrogliopathy (ARTAG). The substantia nigra (SN) is one of the important degenerative areas in tauopathies with parkinsonism. Nigral tau pathology is usually reported as neuronal predominant with less prominent astrocytic involvement. We aimed to identify cases with prominent astrocytic tau pathology in the SN. METHODS: We use the term nigral tau-astrogliopathy (NITAG) to describe cases showing an unusually high density of ARTAG with less neuronal tau pathology in the SN. We collected clinical information and studied the distribution of tau pathology, morphological features and immunostaining profiles in three cases. RESULTS: Three cases, all males with parkinsonism, were identified with the following clinicopathological diagnoses: (i) atypical parkinsonism with tau pathology reminiscent to that in postencephalitic parkinsonism (69-year-old); (ii) multiple system atrophy (73-year-old); (iii) traumatic encephalopathy syndrome/chronic traumatic encephalopathy (84-year-old). Double-labelling immunofluorescence confirmed co-localization of GFAP and phosphorylated tau in affected astrocytes. Staining profiles of NITAG revealed immunopositivity for various phosphorylated tau antibodies. Some astrocytic tau lesions were also seen in other brainstem regions and cerebral grey matter. CONCLUSIONS: We propose NITAG is a rare neuropathological feature, and not a distinct disease entity, in the frame of multiple system ARTAG, represented by abundant tau-positive astrocytes in various brain regions but having the highest density in the SN. The concept of NITAG allows the stratification of cases with various background pathologies to understand its relevance and contribution to neuronal dysfunction.


Subject(s)
Aging , Astrocytes , Substantia Nigra , Tauopathies , tau Proteins , Humans , Male , Substantia Nigra/pathology , Substantia Nigra/metabolism , Aged , Astrocytes/pathology , Astrocytes/metabolism , Tauopathies/pathology , Tauopathies/metabolism , Aged, 80 and over , Aging/pathology , tau Proteins/metabolism , Parkinsonian Disorders/pathology , Parkinsonian Disorders/metabolism
8.
Cell Death Dis ; 15(6): 429, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890273

ABSTRACT

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.


Subject(s)
Neurons , tau Proteins , Animals , tau Proteins/metabolism , tau Proteins/genetics , Humans , Mice , Neurons/metabolism , Phosphorylation , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Tauopathies/metabolism , Tauopathies/pathology , Tauopathies/genetics , Synapses/metabolism , Neurogranin/metabolism , Neurogranin/genetics , Calcineurin/metabolism
9.
Nat Commun ; 15(1): 4706, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830849

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Magnetic Resonance Imaging , Tauopathies , White Matter , tau Proteins , Humans , White Matter/diagnostic imaging , White Matter/pathology , White Matter/metabolism , Female , Male , Aged , Middle Aged , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/metabolism , Alzheimer Disease/diagnostic imaging , Tauopathies/diagnostic imaging , Tauopathies/metabolism , Tauopathies/pathology , Tauopathies/genetics , Tauopathies/cerebrospinal fluid , tau Proteins/metabolism , tau Proteins/cerebrospinal fluid , Brain/pathology , Brain/diagnostic imaging , Brain/metabolism , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Neurites/metabolism , Neurites/pathology
10.
Brain ; 147(7): 2428-2439, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38842726

ABSTRACT

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.


Subject(s)
Cerebral Cortex , Positron-Emission Tomography , Supranuclear Palsy, Progressive , Tauopathies , tau Proteins , Humans , Male , Female , Positron-Emission Tomography/methods , Aged , Tauopathies/diagnostic imaging , Tauopathies/metabolism , Tauopathies/pathology , tau Proteins/metabolism , Middle Aged , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Supranuclear Palsy, Progressive/diagnostic imaging , Supranuclear Palsy, Progressive/metabolism , Supranuclear Palsy, Progressive/pathology , Supranuclear Palsy, Progressive/physiopathology , Magnetic Resonance Imaging/methods
11.
Alzheimers Dement ; 20(7): 4540-4558, 2024 07.
Article in English | MEDLINE | ID: mdl-38884283

ABSTRACT

INTRODUCTION: Intraneuronal inclusions composed of tau protein are found in Alzheimer's disease (AD) and other tauopathies. Tau normally binds microtubules (MTs), and its disengagement from MTs and misfolding in AD is thought to result in MT abnormalities. We previously identified triazolopyrimidine-containing MT-stabilizing compounds that provided benefit in AD mouse models and herein describe the characterization and efficacy testing of an optimized candidate, CNDR-51997. METHODS: CNDR-51997 underwent pharmacokinetic, pharmacodynamic, safety pharmacology, and mouse tolerability testing. In addition, the compound was examined for efficacy in 5XFAD amyloid beta (Aß) plaque mice and PS19 tauopathy mice. RESULTS: CNDR-51997 significantly reduced Aß plaques in 5XFAD mice and tau pathology in PS19 mice, with the latter also showing attenuated axonal dystrophy and gliosis. CNDR-51997 was well tolerated at doses that exceeded efficacy doses, with a good safety pharmacology profile. DISCUSSION: CNDR-51997 may be a candidate for advancement as a potential therapeutic agent for AD and/or other tauopathies. Highlights There is evidence of microtubule alterations (MT) in Alzheimer's disease (AD) brain and in mouse models of AD pathology. Intermittent dosing with an optimized, brain-penetrant MT-stabilizing small-molecule, CNDR-51997, reduced both Aß plaque and tau inclusion pathology in established mouse models of AD. CNDR-51997 attenuated axonal dystrophy and gliosis in a tauopathy mouse model, with a strong trend toward reduced hippocampal neuron loss. CNDR-51997 is well tolerated in mice at doses that are meaningfully greater than required for efficacy in AD mouse models, and the compound has a good safety pharmacology profile.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Mice, Transgenic , Microtubules , Plaque, Amyloid , tau Proteins , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Mice , Plaque, Amyloid/drug therapy , Plaque, Amyloid/pathology , tau Proteins/metabolism , Microtubules/drug effects , Microtubules/metabolism , Brain/drug effects , Brain/pathology , Brain/metabolism , Tauopathies/drug therapy , Tauopathies/pathology , Humans , Tubulin Modulators/pharmacology , Tubulin Modulators/therapeutic use , Amyloid beta-Peptides/metabolism
12.
Alzheimers Dement ; 20(7): 4985-4998, 2024 07.
Article in English | MEDLINE | ID: mdl-38923171

ABSTRACT

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.


Subject(s)
Disease Models, Animal , Mice, Transgenic , Tauopathies , tau Proteins , Animals , Mice , Brain/pathology , Brain/metabolism , Phosphorylation , tau Proteins/metabolism , Tauopathies/pathology , Tauopathies/metabolism , Tauopathies/genetics
13.
eNeuro ; 11(6)2024 Jun.
Article in English | MEDLINE | ID: mdl-38858068

ABSTRACT

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.


Subject(s)
Cognitive Dysfunction , Disease Models, Animal , Mice, Transgenic , Prosencephalon , Synapses , tau Proteins , Animals , tau Proteins/metabolism , Male , Female , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/etiology , Synapses/metabolism , Synapses/pathology , Mice , Prosencephalon/metabolism , Sex Characteristics , Tauopathies/metabolism , Tauopathies/pathology , Sleep Wake Disorders/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Mice, Inbred C57BL
14.
J Neurol Sci ; 462: 123079, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38878650

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Astrocytes , Brain , Disease Models, Animal , Mice, Transgenic , Positron-Emission Tomography , Tauopathies , Animals , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Astrocytes/metabolism , Positron-Emission Tomography/methods , Tauopathies/diagnostic imaging , Tauopathies/metabolism , Tauopathies/pathology , Mice , Brain/diagnostic imaging , Brain/metabolism , Brain/pathology , Fluorine Radioisotopes , Male , Amyloid beta-Peptides/metabolism , Humans , tau Proteins/metabolism , tau Proteins/genetics , Fluorodeoxyglucose F18 , Radiopharmaceuticals
15.
Proc Natl Acad Sci U S A ; 121(27): e2311831121, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38941274

ABSTRACT

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.


Subject(s)
Anoctamins , Neurons , Phosphatidylserines , Tauopathies , tau Proteins , Animals , Anoctamins/metabolism , Anoctamins/genetics , Phosphatidylserines/metabolism , Neurons/metabolism , Neurons/pathology , tau Proteins/metabolism , tau Proteins/genetics , Mice , Tauopathies/metabolism , Tauopathies/pathology , Humans , Microglia/metabolism , Microglia/pathology , Phosphorylation , Mice, Transgenic , Disease Models, Animal , Phospholipid Transfer Proteins/metabolism , Phospholipid Transfer Proteins/genetics , Brain/metabolism , Brain/pathology , Mice, Knockout
16.
J Math Biol ; 89(1): 4, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750128

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Brain , Computer Simulation , Mathematical Concepts , Neurodegenerative Diseases , tau Proteins , tau Proteins/metabolism , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Brain/metabolism , Brain/pathology , Models, Neurological , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Models, Biological , Disease Progression , Tauopathies/metabolism , Tauopathies/pathology
17.
J Neuroinflammation ; 21(1): 130, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750510

ABSTRACT

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.


Subject(s)
Aquaporin 4 , Astrocytes , Excitatory Amino Acid Transporter 2 , Mice, Transgenic , Tauopathies , tau Proteins , Astrocytes/metabolism , Astrocytes/pathology , Animals , Mice , tau Proteins/metabolism , tau Proteins/genetics , Aquaporin 4/metabolism , Aquaporin 4/genetics , Tauopathies/metabolism , Tauopathies/pathology , Tauopathies/genetics , Humans , Excitatory Amino Acid Transporter 2/metabolism , Excitatory Amino Acid Transporter 2/genetics , Excitatory Amino Acid Transporter 2/biosynthesis , Brain Concussion/metabolism , Brain Concussion/pathology , Male , Phenotype , Mice, Inbred C57BL
18.
J Alzheimers Dis ; 99(3): 1023-1032, 2024.
Article in English | MEDLINE | ID: mdl-38728190

ABSTRACT

Background: TAR DNA binding protein 43 (TDP-43) has been shown to be associated with whole hippocampal atrophy in primary age-related tauopathy (PART). It is currently unknown which subregions of the hippocampus are contributing to TDP-43 associated whole hippocampal atrophy in PART. Objective: To identify which specific hippocampal subfield regions are contributing to TDP-43-associated whole hippocampal atrophy in PART. Methods: A total of 115 autopsied cases from the Mayo Clinic Alzheimer Disease Research Center, Neurodegenerative Research Group, and the Mayo Clinic Study of Aging were analyzed. All cases underwent antemortem brain volumetric MRI, neuropathological assessment of the distribution of Aß (Thal phase), and neurofibrillary tangle (Braak stage) to diagnose PART, as well as assessment of TDP-43 presence/absence in the amygdala, hippocampus and beyond. Hippocampal subfield segmentation was performed using FreeSurfer version 7.4.1. Statistical analyses using logistic regression were performed to assess for associations between TDP-43 and hippocampal subfield volumes, accounting for potential confounders. Results: TDP-43 positive patients (n = 37, 32%), of which 15/15 were type-α, had significantly smaller whole hippocampal volumes, and smaller volumes of the body and tail of the hippocampus compared to TDP-43 negative patients. Subfield analyses revealed an association between TDP-43 and the molecular layer of hippocampal body and the body of cornu ammonis 1 (CA1), subiculum, and presubiculum regions. There was no association between TDP-43 stage and subfield volumes. Conclusions: Whole hippocampal volume loss linked to TDP-43 in PART is mainly due to volume loss occurring in the molecular layer, CA1, subiculum and presubiculum of the hippocampal body.


Subject(s)
Atrophy , DNA-Binding Proteins , Hippocampus , Tauopathies , Humans , Male , Female , Atrophy/pathology , Tauopathies/pathology , Tauopathies/diagnostic imaging , Aged , DNA-Binding Proteins/metabolism , Hippocampus/pathology , Hippocampus/diagnostic imaging , Aged, 80 and over , Magnetic Resonance Imaging , Middle Aged
19.
Acta Neuropathol ; 147(1): 86, 2024 05 17.
Article in English | MEDLINE | ID: mdl-38758288

ABSTRACT

Dominantly inherited mutation D395G in the gene encoding valosin-containing protein causes vacuolar tauopathy, a type of behavioural-variant frontotemporal dementia, with marked vacuolation and abundant filamentous tau inclusions made of all six brain isoforms. Here we report that tau inclusions were concentrated in layers II/III of the frontotemporal cortex in a case of vacuolar tauopathy. By electron cryomicroscopy, tau filaments had the chronic traumatic encephalopathy (CTE) fold. Tau inclusions of vacuolar tauopathy share this cortical location and the tau fold with CTE, subacute sclerosing panencephalitis and amyotrophic lateral sclerosis/parkinsonism-dementia complex, which are believed to be environmentally induced. Vacuolar tauopathy is the first inherited disease with the CTE tau fold.


Subject(s)
Chronic Traumatic Encephalopathy , Mutation , Tauopathies , Valosin Containing Protein , tau Proteins , Humans , Tauopathies/genetics , Tauopathies/pathology , Chronic Traumatic Encephalopathy/pathology , Chronic Traumatic Encephalopathy/genetics , tau Proteins/genetics , tau Proteins/metabolism , Valosin Containing Protein/genetics , Vacuoles/pathology , Vacuoles/ultrastructure , Male , Adenosine Triphosphatases/genetics , Cell Cycle Proteins/genetics , Middle Aged , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Brain/pathology , Female
20.
Cell Death Dis ; 15(5): 333, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38740758

ABSTRACT

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.


Subject(s)
Autophagy , Spermine Synthase , tau Proteins , Animals , tau Proteins/metabolism , Humans , Spermine Synthase/metabolism , Spermine Synthase/genetics , Drosophila melanogaster/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Tauopathies/metabolism , Tauopathies/pathology , Neurons/metabolism , Neurons/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Spermidine/metabolism , Disease Models, Animal , Lysosomes/metabolism , Drosophila/metabolism , Mental Retardation, X-Linked
SELECTION OF CITATIONS
SEARCH DETAIL