Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.759
Filtrer
1.
Clin Nucl Med ; 49(8): 754-756, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38967508

RÉSUMÉ

ABSTRACT: Currently, monoamine oxidase B is recognized as the primary target of 18F-THK5351, although 18F-THK5351 was initially developed to target neurofibrillary tangles (NFTs) in Alzheimer disease. When clinically applying 18F-THK5351 PET to visualize ongoing astrogliosis via estimating monoamine oxidase B levels, a crucial concern is how much degree 18F-THK5351 uptake reflects NFTs in in vivo images. To unravel this concern, a head-to-head comparison between 18F-THK5351 and 18F-MK-6240 (estimating NFT) images in the NFT lesion ideally without accompanying astrogliosis is essential. Here, we present such a case suggesting that 18F-THK5351 uptake may not estimate NFTs in in vivo images.


Sujet(s)
Enchevêtrements neurofibrillaires , Enchevêtrements neurofibrillaires/métabolisme , Enchevêtrements neurofibrillaires/anatomopathologie , Humains , Tomographie par émission de positons , Aminopyridines , Transport biologique , Sujet âgé , Mâle , Femelle , Maladie d'Alzheimer/imagerie diagnostique , Maladie d'Alzheimer/métabolisme , Isoquinoléines , Quinoléines
2.
Acta Neuropathol ; 148(1): 3, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38980423

RÉSUMÉ

This study investigates various pathological tau isoforms in the retina of individuals with early and advanced Alzheimer's disease (AD), exploring their connection with disease status. Retinal cross-sections from predefined superior-temporal and inferior-temporal subregions and corresponding brains from neuropathologically confirmed AD patients with a clinical diagnosis of either mild cognitive impairment (MCI) or dementia (n = 45) were compared with retinas from age- and sex-matched individuals with normal cognition (n = 30) and non-AD dementia (n = 4). Retinal tau isoforms, including tau tangles, paired helical filament of tau (PHF-tau), oligomeric-tau (Oligo-tau), hyperphosphorylated-tau (p-tau), and citrullinated-tau (Cit-tau), were stereologically analyzed by immunohistochemistry and Nanostring GeoMx digital spatial profiling, and correlated with clinical and neuropathological outcomes. Our data indicated significant increases in various AD-related pretangle tau isoforms, especially p-tau (AT8, 2.9-fold, pS396-tau, 2.6-fold), Cit-tau at arginine residue 209 (CitR209-tau; 4.1-fold), and Oligo-tau (T22+, 9.2-fold), as well as pretangle and mature tau tangle forms like MC-1-positive (1.8-fold) and PHF-tau (2.3-fold), in AD compared to control retinas. MCI retinas also exhibited substantial increases in Oligo-tau (5.2-fold), CitR209-tau (3.5-fold), and pS396-tau (2.2-fold). Nanostring GeoMx analysis confirmed elevated retinal p-tau at epitopes: Ser214 (2.3-fold), Ser396 (2.6-fold), Ser404 (2.4-fold), and Thr231 (1.8-fold), particularly in MCI patients. Strong associations were found between retinal tau isoforms versus brain pathology and cognitive status: a) retinal Oligo-tau vs. Braak stage, neurofibrillary tangles (NFTs), and CDR cognitive scores (ρ = 0.63-0.71), b) retinal PHF-tau vs. neuropil threads (NTs) and ABC scores (ρ = 0.69-0.71), and c) retinal pS396-tau vs. NTs, NFTs, and ABC scores (ρ = 0.67-0.74). Notably, retinal Oligo-tau strongly correlated with retinal Aß42 and arterial Aß40 forms (r = 0.76-0.86). Overall, this study identifies and quantifies diverse retinal tau isoforms in MCI and AD patients, underscoring their link to brain pathology and cognition. These findings advocate for further exploration of retinal tauopathy biomarkers to facilitate AD detection and monitoring via noninvasive retinal imaging.


Sujet(s)
Maladie d'Alzheimer , Isoformes de protéines , Rétine , Protéines tau , Humains , Protéines tau/métabolisme , Mâle , Femelle , Sujet âgé , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/métabolisme , Rétine/anatomopathologie , Rétine/métabolisme , Sujet âgé de 80 ans ou plus , Dysfonctionnement cognitif/anatomopathologie , Dysfonctionnement cognitif/métabolisme , Adulte d'âge moyen , Enchevêtrements neurofibrillaires/anatomopathologie , Enchevêtrements neurofibrillaires/métabolisme , Encéphale/anatomopathologie , Encéphale/métabolisme
3.
Neurobiol Aging ; 141: 160-170, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38964013

RÉSUMÉ

Women have a higher incidence of Alzheimer's disease (AD), even after adjusting for increased longevity. Thus, there is an urgent need to identify genes that underpin sex-associated risk of AD. PIN1 is a key regulator of the tau phosphorylation signaling pathway; however, potential differences in PIN1 expression, in males and females, are still unknown. We analyzed brain transcriptomic datasets focusing on sex differences in PIN1 mRNA levels in an aging and AD cohort, which revealed reduced PIN1 levels primarily within females. We validated this observation in an independent dataset (ROS/MAP), which also revealed that PIN1 is negatively correlated with multiregional neurofibrillary tangle density and global cognitive function in females only. Additional analysis revealed a decrease in PIN1 in subjects with mild cognitive impairment (MCI) compared with aged individuals, again driven predominantly by female subjects. Histochemical analysis of PIN1 in AD and control male and female neocortex revealed an overall decrease in axonal PIN1 protein levels in females. These findings emphasize the importance of considering sex differences in AD research.


Sujet(s)
Maladie d'Alzheimer , Cognition , Dysfonctionnement cognitif , NIMA-interacting peptidylprolyl isomerase , Néocortex , Enchevêtrements neurofibrillaires , Caractères sexuels , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/métabolisme , NIMA-interacting peptidylprolyl isomerase/génétique , NIMA-interacting peptidylprolyl isomerase/métabolisme , Humains , Femelle , Néocortex/anatomopathologie , Néocortex/métabolisme , Mâle , Dysfonctionnement cognitif/génétique , Dysfonctionnement cognitif/anatomopathologie , Dysfonctionnement cognitif/métabolisme , Sujet âgé , Sujet âgé de 80 ans ou plus , Enchevêtrements neurofibrillaires/anatomopathologie , Enchevêtrements neurofibrillaires/métabolisme , Phénotype , Système limbique/anatomopathologie , Système limbique/métabolisme , Expression des gènes , Vieillissement/anatomopathologie , Vieillissement/génétique , Vieillissement/métabolisme , ARN messager/métabolisme , ARN messager/génétique , Protéines tau/métabolisme , Protéines tau/génétique , Phosphorylation
4.
Molecules ; 29(12)2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38930877

RÉSUMÉ

Tau protein is a microtubule-associated protein that is widely distributed in the central nervous system and maintains and regulates neuronal morphology and function. Tau protein aggregates abnormally and forms neurofibrillary tangles in neurodegenerative diseases, disrupting the structure and function of neurons and leading to neuronal death, which triggers the initiation and progression of neurological disorders. The aggregation of tau protein in neurodegenerative diseases is associated with post-translational modifications, which may affect the hydrophilicity, spatial conformation, and stability of tau protein, promoting tau protein aggregation and the formation of neurofibrillary tangles. Therefore, studying the role of tau protein in neurodegenerative diseases and the mechanism of aberrant aggregation is important for understanding the mechanism of neurodegenerative diseases and finding therapeutic approaches. This review describes the possible mechanisms by which tau protein promotes neurodegenerative diseases, the post-translational modifications of tau protein and associated influencing factors, and the current status of drug discovery and development related to tau protein, which may contribute to the development of new therapeutic approaches to alleviate or treat neurodegenerative diseases.


Sujet(s)
Maladies neurodégénératives , Maturation post-traductionnelle des protéines , Protéines tau , Protéines tau/métabolisme , Humains , Maladies neurodégénératives/traitement médicamenteux , Maladies neurodégénératives/métabolisme , Animaux , Développement de médicament , Agrégation pathologique de protéines/métabolisme , Agrégation pathologique de protéines/traitement médicamenteux , Enchevêtrements neurofibrillaires/métabolisme , Thérapie moléculaire ciblée
5.
Nat Commun ; 15(1): 4803, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38839876

RÉSUMÉ

Our current understanding of the spread and neurodegenerative effects of tau neurofibrillary tangles (NFTs) within the medial temporal lobe (MTL) during the early stages of Alzheimer's Disease (AD) is limited by the presence of confounding non-AD pathologies and the two-dimensional (2-D) nature of conventional histology studies. Here, we combine ex vivo MRI and serial histological imaging from 25 human MTL specimens to present a detailed, 3-D characterization of quantitative NFT burden measures in the space of a high-resolution, ex vivo atlas with cytoarchitecturally-defined subregion labels, that can be used to inform future in vivo neuroimaging studies. Average maps show a clear anterior to poster gradient in NFT distribution and a precise, spatial pattern with highest levels of NFTs found not just within the transentorhinal region but also the cornu ammonis (CA1) subfield. Additionally, we identify granular MTL regions where measures of neurodegeneration are likely to be linked to NFTs specifically, and thus potentially more sensitive as early AD biomarkers.


Sujet(s)
Maladie d'Alzheimer , Imagerie par résonance magnétique , Enchevêtrements neurofibrillaires , Lobe temporal , Protéines tau , Humains , Maladie d'Alzheimer/imagerie diagnostique , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Lobe temporal/imagerie diagnostique , Lobe temporal/métabolisme , Lobe temporal/anatomopathologie , Protéines tau/métabolisme , Mâle , Femelle , Sujet âgé , Imagerie par résonance magnétique/méthodes , Enchevêtrements neurofibrillaires/métabolisme , Enchevêtrements neurofibrillaires/anatomopathologie , Sujet âgé de 80 ans ou plus , Autopsie , Neuroimagerie/méthodes , Adulte d'âge moyen ,
6.
Acta Neuropathol Commun ; 12(1): 94, 2024 06 12.
Article de Anglais | MEDLINE | ID: mdl-38867338

RÉSUMÉ

Down syndrome (DS) is a common genetic condition caused by trisomy of chromosome 21. Among their complex clinical features, including musculoskeletal, neurological, and cardiovascular disabilities, individuals with DS have an increased risk of developing progressive dementia and early-onset Alzheimer's disease (AD). This dementia is attributed to the increased gene dosage of the amyloid-ß (Aß) precursor protein gene, the formation of self-propagating Aß and tau prion conformers, and the deposition of neurotoxic Aß plaques and tau neurofibrillary tangles. Tau amyloid fibrils have previously been established to adopt many distinct conformations across different neurodegenerative conditions. Here, we report the characterization of brain samples from four DS cases spanning 36-63 years of age by spectral confocal imaging with conformation-specific dyes and cryo-electron microscopy (cryo-EM) to determine structures of isolated tau fibrils. High-resolution structures revealed paired helical filament (PHF) and straight filament (SF) conformations of tau that were identical to those determined from AD cases. The PHFs and SFs are made of two C-shaped protofilaments, each containing a cross-ß/ß-helix motif. Similar to filaments from AD cases, most filaments from the DS cases adopted the PHF form, while a minority (approximately 20%) formed SFs. Samples from the youngest individual with no documented dementia had sparse tau deposits. To isolate tau for cryo-EM from this challenging sample we used a novel affinity-grid method involving a graphene oxide surface derivatized with anti-tau antibodies. This method improved isolation and revealed that primarily tau PHFs and a minor population of chronic traumatic encephalopathy type II-like filaments were present in this youngest case. These findings expand the similarities between AD and DS to the molecular level, providing insight into their related pathologies and the potential for targeting common tau filament folds by small-molecule therapeutics and diagnostics.


Sujet(s)
Maladie d'Alzheimer , Cryomicroscopie électronique , Syndrome de Down , Protéines tau , Humains , Syndrome de Down/anatomopathologie , Syndrome de Down/métabolisme , Protéines tau/métabolisme , Protéines tau/ultrastructure , Cryomicroscopie électronique/méthodes , Adulte d'âge moyen , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/métabolisme , Femelle , Adulte , Mâle , Enchevêtrements neurofibrillaires/anatomopathologie , Enchevêtrements neurofibrillaires/métabolisme , Encéphale/anatomopathologie , Encéphale/métabolisme , Encéphale/ultrastructure
7.
Acta Neuropathol ; 147(1): 98, 2024 06 11.
Article de Anglais | MEDLINE | ID: mdl-38861157

RÉSUMÉ

Widespread cortical accumulation of misfolded pathological tau proteins (ptau) in the form of paired helical filaments is a major hallmark of Alzheimer's disease. Subcellular localization of ptau at various stages of disease progression is likely to be informative of the cellular mechanisms involving its spread. Here, we found that the density of ptau within several distinct rostral thalamic nuclei in post-mortem human tissue (n = 25 cases) increased with the disease stage, with the anterodorsal nucleus (ADn) consistently being the most affected. In the ADn, ptau-positive elements were present already in the pre-cortical (Braak 0) stage. Tau pathology preferentially affected the calretinin-expressing subpopulation of glutamatergic neurons in the ADn. At the subcellular level, we detected ptau immunoreactivity in ADn cell bodies, dendrites, and in a specialized type of presynaptic terminal that expresses vesicular glutamate transporter 2 (vGLUT2) and likely originates from the mammillary body. The ptau-containing terminals displayed signs of degeneration, including endosomal/lysosomal organelles. In contrast, corticothalamic axon terminals lacked ptau. The data demonstrate the involvement of a specific cell population in ADn at the onset of the disease. The presence of ptau in subcortical glutamatergic presynaptic terminals supports hypotheses about the transsynaptic spread of tau selectively affecting specialized axonal pathways.


Sujet(s)
Maladie d'Alzheimer , Protéines tau , Humains , Protéines tau/métabolisme , Femelle , Mâle , Sujet âgé , Sujet âgé de 80 ans ou plus , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/métabolisme , Adulte d'âge moyen , Neurones/métabolisme , Neurones/anatomopathologie , Transporteur vésiculaire-2 du glutamate/métabolisme , Acide glutamique/métabolisme , Noyaux antérieurs du thalamus/métabolisme , Noyaux antérieurs du thalamus/anatomopathologie , Calbindine-2/métabolisme , Enchevêtrements neurofibrillaires/anatomopathologie , Enchevêtrements neurofibrillaires/métabolisme , Terminaisons présynaptiques/métabolisme , Terminaisons présynaptiques/anatomopathologie
9.
Alzheimers Dement ; 20(7): 4499-4511, 2024 07.
Article de Anglais | MEDLINE | ID: mdl-38856164

RÉSUMÉ

INTRODUCTION: The ɛ4 allele of the apolipoprotein E gene (APOE ɛ4) is the strongest genetic risk factor for Alzheimer's disease (AD), but the mechanisms connecting APOE ɛ4 to AD are not clear. METHODS: Participants (n = 596) were from two clinical-pathological studies. Tissues from dorsolateral prefrontal cortex were examined to identify 8425 proteins. Post mortem pathological assessment used immunohistochemistry to obtain amyloid beta (Aß) load and tau tangle density. RESULTS: In separate models, APOE ɛ4 was associated with 18 proteins, which were associated with Aß and tau tangles. Examining the proteins in a single model identified Netrin-1 and secreted frizzled-related protein 1 (SFRP1) as the two proteins linking APOE ɛ4 with Aß with the largest effect sizes and Netrin-1 and testican-3 linking APOE ɛ4 with tau tangles. DISCUSSION: We identified Netrin-1, SFRP1, and testican-3 as the most promising proteins that link APOE ɛ4 with Aß and tau tangles. HIGHLIGHTS: Of 8425 proteins extracted from prefrontal cortex, 18 were related to APOE ɛ4. The 18 proteins were also related to amyloid beta (Aß) and tau. The 18 proteins were more related to APOE ɛ4 than other AD genetic risk variants. Netrin-1 and secreted frizzled-related protein 1 were the two most promising proteins linking APOE ɛ4 with Aß. Netrin-1 and testican-3 were two most promising proteins linking APOE ɛ4 with tau.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Apolipoprotéine E4 , Nétrine-1 , Cortex préfrontal , Humains , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/métabolisme , Femelle , Mâle , Apolipoprotéine E4/génétique , Peptides bêta-amyloïdes/métabolisme , Sujet âgé , Nétrine-1/métabolisme , Nétrine-1/génétique , Cortex préfrontal/métabolisme , Sujet âgé de 80 ans ou plus , Protéines tau/métabolisme , Enchevêtrements neurofibrillaires/métabolisme , Enchevêtrements neurofibrillaires/anatomopathologie
10.
Acta Neuropathol ; 147(1): 101, 2024 06 17.
Article de Anglais | MEDLINE | ID: mdl-38884806

RÉSUMÉ

Insoluble pathogenic proteins accumulate along blood vessels in conditions of cerebral amyloid angiopathy (CAA), exerting a toxic effect on vascular cells and impacting cerebral homeostasis. In this work, we provide new evidence from three-dimensional human brain histology that tau protein, the main component of neurofibrillary tangles, can similarly accumulate along brain vascular segments. We quantitatively assessed n = 6 Alzheimer's disease (AD), and n = 6 normal aging control brains and saw that tau-positive blood vessel segments were present in all AD cases. Tau-positive vessels are enriched for tau at levels higher than the surrounding tissue and appear to affect arterioles across cortical layers (I-V). Further, vessels isolated from these AD tissues were enriched for N-terminal tau and tau phosphorylated at T181 and T217. Importantly, tau-positive vessels are associated with local areas of increased tau neurofibrillary tangles. This suggests that accumulation of tau around blood vessels may reflect a local clearance failure. In sum, these data indicate that tau, like amyloid beta, accumulates along blood vessels and may exert a significant influence on vasculature in the setting of AD.


Sujet(s)
Maladie d'Alzheimer , Encéphale , Enchevêtrements neurofibrillaires , Protéines tau , Humains , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/métabolisme , Protéines tau/métabolisme , Enchevêtrements neurofibrillaires/anatomopathologie , Enchevêtrements neurofibrillaires/métabolisme , Encéphale/anatomopathologie , Encéphale/métabolisme , Femelle , Mâle , Sujet âgé , Sujet âgé de 80 ans ou plus , Adulte d'âge moyen , Phosphorylation
11.
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
12.
Commun Biol ; 7(1): 569, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38750228

RÉSUMÉ

Accumulation of amyloid-ß (Aß) and tau tangles are hallmarks of Alzheimer's disease. Aß is extracellular while tau tangles are typically intracellular, and it is unknown how these two proteinopathies are connected. Here, we use data of 1206 elders and test that RNA expression levels of GPER1, a transmembrane protein, modify the association of Aß with tau tangles. GPER1 RNA expression is related to more tau tangles (p = 0.001). Moreover, GPER1 expression modifies the association of immunohistochemistry-derived Aß load with tau tangles (p = 0.044). Similarly, GPER1 expression modifies the association between Aß proteoforms and tau tangles: total Aß protein (p = 0.030) and Aß38 peptide (p = 0.002). Using single nuclei RNA-seq indicates that GPER1 RNA expression in astrocytes modifies the relation of Aß load with tau tangles (p = 0.002), but not GPER1 in excitatory neurons or endothelial cells. We conclude that GPER1 may be a link between Aß and tau tangles driven mainly by astrocytic GPER1 expression.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Récepteurs des oestrogènes , Récepteurs couplés aux protéines G , Protéines tau , Humains , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Protéines tau/métabolisme , Protéines tau/génétique , Femelle , Mâle , Peptides bêta-amyloïdes/métabolisme , Peptides bêta-amyloïdes/génétique , Sujet âgé , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/génétique , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/génétique , Sujet âgé de 80 ans ou plus , Enchevêtrements neurofibrillaires/métabolisme , Enchevêtrements neurofibrillaires/anatomopathologie , Astrocytes/métabolisme
13.
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
14.
Brain ; 147(6): 2144-2157, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38667631

RÉSUMÉ

Recent longitudinal PET imaging studies have established methods to estimate the age at which amyloid becomes abnormal at the level of the individual. Here we recontextualized amyloid levels into the temporal domain to better understand the downstream Alzheimer's disease processes of tau neurofibrillary tangle (NFT) accumulation and cognitive decline. This cohort study included a total of 601 individuals from the Wisconsin Registry for Alzheimer's Prevention and Wisconsin Alzheimer's Disease Research Center that underwent amyloid and tau PET, longitudinal neuropsychological assessments and met clinical criteria for three clinical diagnosis groups: cognitively unimpaired (n = 537); mild cognitive impairment (n = 48); or dementia (n = 16). Cortical 11C-Pittsburgh compound B (PiB) distribution volume ratio (DVR) and sampled iterative local approximation were used to estimate amyloid positive (A+; global PiB DVR > 1.16 equivalent to 17.1 centiloids) onset age and years of A+ duration at tau PET (i.e. amyloid chronicity). Tau PET burden was quantified using 18F-MK-6240 standardized uptake value ratios (70-90 min, inferior cerebellar grey matter reference region). Whole-brain and region-specific approaches were used to examine tau PET binding along the amyloid timeline and across the Alzheimer's disease clinical continuum. Voxel-wise 18F-MK-6240 analyses revealed that with each decade of A+, the spatial extent of measurable tau spread (i.e. progressed) from regions associated with early to late NFT tau stages. Regional analyses indicated that tau burden in the entorhinal cortex was detectable, on average, within 10 years of A+ onset. Additionally, the entorhinal cortex was the region most sensitive to early amyloid pathology and clinical impairment in this predominantly preclinical sample. Among initially cognitively unimpaired (n = 472) individuals with longitudinal cognitive follow-up, mixed effects models showed significant linear and non-linear interactions of A+ duration and entorhinal tau on cognitive decline, suggesting a synergistic effect whereby greater A+ duration, together with a higher entorhinal tau burden, increases the likelihood of cognitive decline beyond their separable effects. Overall, the amyloid time framework enabled a spatiotemporal characterization of tau deposition patterns across the Alzheimer's disease continuum. This approach, which examined cross-sectional tau PET data along the amyloid timeline to make longitudinal disease course inferences, demonstrated that A+ duration explains a considerable amount of variability in the magnitude and topography of tau spread, which largely recapitulated NFT staging observed in human neuropathological studies. By anchoring disease progression to the onset of amyloid, this study provides a temporal disease context, which may help inform disease prognosis and timing windows for anti-amyloid therapies.


Sujet(s)
Maladie d'Alzheimer , Encéphale , Dysfonctionnement cognitif , Tomographie par émission de positons , Protéines tau , Humains , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/imagerie diagnostique , Maladie d'Alzheimer/anatomopathologie , Sujet âgé , Mâle , Femelle , Protéines tau/métabolisme , Tomographie par émission de positons/méthodes , Dysfonctionnement cognitif/métabolisme , Dysfonctionnement cognitif/imagerie diagnostique , Dysfonctionnement cognitif/anatomopathologie , Encéphale/métabolisme , Encéphale/imagerie diagnostique , Encéphale/anatomopathologie , Sujet âgé de 80 ans ou plus , Enchevêtrements neurofibrillaires/anatomopathologie , Enchevêtrements neurofibrillaires/métabolisme , Évolution de la maladie , Dérivés de l'aniline , Études de cohortes , Peptides bêta-amyloïdes/métabolisme , Adulte d'âge moyen , Études longitudinales , Thiazoles , Tests neuropsychologiques , Amyloïde/métabolisme
15.
Methods Mol Biol ; 2754: 193-203, 2024.
Article de Anglais | MEDLINE | ID: mdl-38512668

RÉSUMÉ

Alzheimer's disease (AD) is characterized by the abnormal accumulation of disordered protein, that is, extracellular senile plaques of amyloid-ß (Aß) and intracellular neurofibrillary tangles of Tau. Tau protein has gained the attention in recent years owing to the ability to propagate in a "prion-like" nature. The disordered protein Tau possesses a high positive charge, which allows its binding to anionic proteins and factors. The native disorder of proteins attends the ß-sheet structure from its random-coiled conformation upon charge compensation by various polyanionic agents such as heparin, RNA, etc. Anionic lipids such as arachidonic acid (AA) and oleic acid (OA) are also one of the factors which can induce aggregation of Tau in physiological conditions. The free units of Tau protein can bind to lipid membranes through its repeat domain (RD), the anionic side chains of the membrane lipids induce aggregation of Tau by reducing the activation barrier. In this study, we investigated the role of α-linolenic acid (ALA) as an inducing agent for Tau aggregation in vitro conditions. Omega-3 fatty acids bear a capacity to reduce the pathology of Tau by downregulating the Tau phosphorylation pathway. We have studied by using various biochemical or biophysical methods the potency of ALA as an aggregating agent for Tau. We have implemented different techniques such as SDS-PAGE, transmission electron microscopy, CD spectroscopy to evaluated higher-order aggregates of Tau upon induction by ALA.


Sujet(s)
Maladie d'Alzheimer , Protéines tau , Humains , Protéines tau/métabolisme , Acide alpha-linolénique/pharmacologie , Acide alpha-linolénique/métabolisme , Maladie d'Alzheimer/métabolisme , Peptides bêta-amyloïdes/composition chimique , Enchevêtrements neurofibrillaires/métabolisme
16.
Int J Mol Sci ; 25(5)2024 Feb 27.
Article de Anglais | MEDLINE | ID: mdl-38473986

RÉSUMÉ

Elderly human brains are vulnerable to multiple proteinopathies, although each protein has a different transmission pathway. Tau-immunoreactive astrocytes are well-known in elderly brains. In contrast, astrocytic plaques, a hallmark in corticobasal degeneration (CBD), rarely occur in aging and neurodegenerative disease other than CBD. To elucidate the clinicopathological correlation of aging-related pathology in CBD, we examined 21 pathologically proven CBD cases in our institute (12 males and 9 females, with a mean age of death 70.6 years). All CBD cases showed grains and neurofibrillary tangles (NFTs). Fifteen cases (71.4%) showed beta-amyloid deposition such as senile plaques or cerebral amyloid angiopathy. Three cases (14.3%) had Lewy body pathology. One case was classified as amygdala-predominant Lewy body disease, although no cases met the pathological criteria for Alzheimer's disease. Five cases (23.8%) displayed Limbic-predominant and age-related TDP-43 encephalopathy (LATE). NFTs, grains, and TDP-43-positive neuronal inclusions were widely distributed throughout the limbic system of CBD patients, but their densities were low. CBD might a have similar cell vulnerability and transmission pathway to that of multiple proteinopathy in aging brains.


Sujet(s)
Maladie d'Alzheimer , Dégénérescence corticobasale , Maladie à corps de Lewy , Maladies neurodégénératives , Mâle , Femelle , Humains , Sujet âgé , Maladies neurodégénératives/métabolisme , Maladie d'Alzheimer/métabolisme , Maladie à corps de Lewy/métabolisme , Enchevêtrements neurofibrillaires/métabolisme , Protéines de liaison à l'ADN/métabolisme , Protéines tau/métabolisme
17.
Int J Mol Sci ; 25(6)2024 Mar 08.
Article de Anglais | MEDLINE | ID: mdl-38542104

RÉSUMÉ

Synaptic transmission is essential for nervous system function and the loss of synapses is a known major contributor to dementia. Alzheimer's disease dementia (ADD) is characterized by synaptic loss in the mesial temporal lobe and cerebral neocortex, both of which are brain areas associated with memory and cognition. The association of synaptic loss and ADD was established in the late 1980s, and it has been estimated that 30-50% of neocortical synaptic protein is lost in ADD, but there has not yet been a quantitative profiling of different synaptic proteins in different brain regions in ADD from the same individuals. Very recently, positron emission tomography (PET) imaging of synapses is being developed, accelerating the focus on the role of synaptic loss in ADD and other conditions. In this study, we quantified the densities of two synaptic proteins, the presynaptic protein Synaptosome Associated Protein 25 (SNAP25) and the postsynaptic protein postsynaptic density protein 95 (PSD95) in the human brain, using enzyme-linked immunosorbent assays (ELISA). Protein was extracted from the cingulate gyrus, hippocampus, frontal, primary visual, and entorhinal cortex from cognitively unimpaired controls, subjects with mild cognitive impairment (MCI), and subjects with dementia that have different levels of Alzheimer's pathology. SNAP25 is significantly reduced in ADD when compared to controls in the frontal cortex, visual cortex, and cingulate, while the hippocampus showed a smaller, non-significant reduction, and entorhinal cortex concentrations were not different. In contrast, all brain areas showed lower PSD95 concentrations in ADD when compared to controls without dementia, although in the hippocampus, this failed to reach significance. Interestingly, cognitively unimpaired cases with high levels of AD pathology had higher levels of both synaptic proteins in all brain regions. SNAP25 and PSD95 concentrations significantly correlated with densities of neurofibrillary tangles, amyloid plaques, and Mini Mental State Examination (MMSE) scores. Our results suggest that synaptic transmission is affected by ADD in multiple brain regions. The differences were less marked in the entorhinal cortex and the hippocampus, most likely due to a ceiling effect imposed by the very early development of neurofibrillary tangles in older people in these brain regions.


Sujet(s)
Maladie d'Alzheimer , Dysfonctionnement cognitif , Humains , Sujet âgé , Maladie d'Alzheimer/métabolisme , Enchevêtrements neurofibrillaires/métabolisme , Encéphale/métabolisme , Dysfonctionnement cognitif/métabolisme , Protéines tau/métabolisme , Tomographie par émission de positons
18.
Mech Ageing Dev ; 219: 111930, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38554950

RÉSUMÉ

This study aimed to elucidate the specific biochemical pathways linked to changes in proteins in the Alzheimer's disease (AD) human hippocampus. Our data demonstrate a constant rise in the expression of four proteins (VGF, GFAP, HSPB1, and APP) across all eleven studies. Notably, UBC was the most centrally involved and had increased expression in the hippocampus tissue of individuals with AD. Modified proteins in the hippocampal tissue were found to activate the innate immune system and disrupt communication across chemical synapses. Four hub proteins (CD44, APP, ITGB2, and APOE) are connected to amyloid plaques, whereas two hub proteins (RPL24 and RPS23) are related to neurofibrillary tangles (NFTs). The presence of modified proteins was discovered to trigger the activation of microglia and decrease the functioning of ribosomes and mitochondria in the hippocampus. Three significant microRNAs (hsa-miR-106b-5p, hsa-miR-17-5p, and hsa-miR-16-5p) and transcription factors (MYT1L, PIN1, and CSRNP3) have been discovered to improve our understanding of the alterations in proteins within the hippocampal tissues that lead to the progression of AD. These findings establish a path for possible treatments for AD to employ therapeutic strategies that specifically focus on the proteins or processes linked to the illness.


Sujet(s)
Maladie d'Alzheimer , Hippocampe , microARN , Humains , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Hippocampe/métabolisme , Hippocampe/anatomopathologie , microARN/métabolisme , Protéines du choc thermique HSP27/métabolisme , Précurseur de la protéine bêta-amyloïde/métabolisme , Mâle , Femelle , Chaperons moléculaires/métabolisme , Enchevêtrements neurofibrillaires/métabolisme , Enchevêtrements neurofibrillaires/anatomopathologie , Immunité innée , NIMA-interacting peptidylprolyl isomerase/métabolisme , NIMA-interacting peptidylprolyl isomerase/génétique , Apolipoprotéines E/métabolisme , Apolipoprotéines E/génétique , Plaque amyloïde/métabolisme , Plaque amyloïde/anatomopathologie , Protéines de tissu nerveux/métabolisme , Protéine gliofibrillaire acide , Protéines du choc thermique
19.
J Alzheimers Dis ; 98(4): 1235-1241, 2024.
Article de Anglais | MEDLINE | ID: mdl-38552113

RÉSUMÉ

 Alzheimer's disease is the leading cause of dementia in the world. It affects 6 million people in the United States and 50 million people worldwide. Alzheimer's disease is characterized by the accumulation of amyloid-ß plaques (Aß), an increase in tau protein neurofibrillary tangles, and a loss of synapses. Since the 1990s, removing and reducing Aß has been the focus of Alzheimer's treatment and prevention research. The accumulation of Aß can lead to oxidative stress, inflammation, neurotoxicity, and eventually apoptosis. These insults impair signaling systems in the brain, potentially leading to memory loss and cognitive decline. Aniracetam is a safe, effective, cognitive-enhancing drug that improves memory in both human and animal studies. Aniracetam may prevent the production and accumulation of Aß by increasing α-secretase activity through two distinct pathways: 1) increasing brain derived neurotrophic factor expression and 2) positively modulating metabotropic glutamate receptors. This is the first paper to propose an evidence-based model for aniracetam reducing the accumulation and production of Aß.


Sujet(s)
Maladie d'Alzheimer , Animaux , Humains , Maladie d'Alzheimer/métabolisme , Peptides bêta-amyloïdes/métabolisme , Pyrrolidones/pharmacologie , Pyrrolidones/usage thérapeutique , Protéines tau/métabolisme , Enchevêtrements neurofibrillaires/métabolisme , Plaque amyloïde/traitement médicamenteux , Plaque amyloïde/métabolisme
20.
J Neuroinflammation ; 21(1): 78, 2024 Mar 27.
Article de Anglais | MEDLINE | ID: mdl-38539208

RÉSUMÉ

BACKGROUND: Accumulation of tau leads to neuroinflammation and neuronal cell death in tauopathies, including Alzheimer's disease. As the disease progresses, there is a decline in brain energy metabolism. However, the role of tau protein in regulating lipid metabolism remains less characterized and poorly understood. METHODS: We used a transgenic rat model for tauopathy to reveal metabolic alterations induced by neurofibrillary pathology. Transgenic rats express a tau fragment truncated at the N- and C-terminals. For phenotypic profiling, we performed targeted metabolomic and lipidomic analysis of brain tissue, CSF, and plasma, based on the LC-MS platform. To monitor disease progression, we employed samples from transgenic and control rats aged 4, 6, 8, 10, 12, and 14 months. To study neuron-glia interplay in lipidome changes induced by pathological tau we used well well-established multicomponent cell model system. Univariate and multivariate statistical approaches were used for data evaluation. RESULTS: We showed that tau has an important role in the deregulation of lipid metabolism. In the lipidomic study, pathological tau was associated with higher production of lipids participating in protein fibrillization, membrane reorganization, and inflammation. Interestingly, significant changes have been found in the early stages of tauopathy before the formation of high-molecular-weight tau aggregates and neurofibrillary pathology. Increased secretion of pathological tau protein in vivo and in vitro induced upregulated production of phospholipids and sphingolipids and accumulation of lipid droplets in microglia. We also found that this process depended on the amount of extracellular tau. During the later stages of tauopathy, we found a connection between the transition of tau into an insoluble fraction and changes in brain metabolism. CONCLUSION: Our results revealed that lipid metabolism is significantly affected during different stages of tau pathology. Thus, our results demonstrate that the dysregulation of lipid composition by pathological tau disrupts the microenvironment, further contributing to the propagation of pathology.


Sujet(s)
Maladie d'Alzheimer , Tauopathies , Rats , Animaux , Souris , Protéines tau/génétique , Protéines tau/métabolisme , Enchevêtrements neurofibrillaires/métabolisme , Métabolisme lipidique , Tauopathies/anatomopathologie , Maladie d'Alzheimer/anatomopathologie , Encéphale/métabolisme , Rats transgéniques , Souris transgéniques , Modèles animaux de maladie humaine
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...