Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.590
Filter
1.
Clin Nucl Med ; 49(8): 754-756, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38967508

ABSTRACT

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.


Subject(s)
Neurofibrillary Tangles , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Humans , Positron-Emission Tomography , Aminopyridines , Biological Transport , Aged , Male , Female , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Isoquinolines , Quinolines
2.
Acta Neuropathol Commun ; 12(1): 94, 2024 06 12.
Article in English | MEDLINE | ID: mdl-38867338

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Cryoelectron Microscopy , Down Syndrome , tau Proteins , Humans , Down Syndrome/pathology , Down Syndrome/metabolism , tau Proteins/metabolism , tau Proteins/ultrastructure , Cryoelectron Microscopy/methods , Middle Aged , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Female , Adult , Male , Neurofibrillary Tangles/pathology , Neurofibrillary Tangles/metabolism , Brain/pathology , Brain/metabolism , Brain/ultrastructure
3.
Nat Commun ; 15(1): 4803, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38839876

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Magnetic Resonance Imaging , Neurofibrillary Tangles , Temporal Lobe , tau Proteins , Humans , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Temporal Lobe/diagnostic imaging , Temporal Lobe/metabolism , Temporal Lobe/pathology , tau Proteins/metabolism , Male , Female , Aged , Magnetic Resonance Imaging/methods , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Aged, 80 and over , Autopsy , Neuroimaging/methods , Middle Aged , Postmortem Imaging
4.
Acta Neuropathol ; 147(1): 98, 2024 06 11.
Article in English | MEDLINE | ID: mdl-38861157

ABSTRACT

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.


Subject(s)
Alzheimer Disease , tau Proteins , Humans , tau Proteins/metabolism , Female , Male , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Middle Aged , Neurons/metabolism , Neurons/pathology , Vesicular Glutamate Transport Protein 2/metabolism , Glutamic Acid/metabolism , Anterior Thalamic Nuclei/metabolism , Anterior Thalamic Nuclei/pathology , Calbindin 2/metabolism , Neurofibrillary Tangles/pathology , Neurofibrillary Tangles/metabolism , Presynaptic Terminals/metabolism , Presynaptic Terminals/pathology
5.
Acta Neuropathol ; 147(1): 101, 2024 06 17.
Article in English | MEDLINE | ID: mdl-38884806

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Brain , Neurofibrillary Tangles , tau Proteins , Humans , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , tau Proteins/metabolism , Neurofibrillary Tangles/pathology , Neurofibrillary Tangles/metabolism , Brain/pathology , Brain/metabolism , Female , Male , Aged , Aged, 80 and over , Middle Aged , Phosphorylation
6.
Molecules ; 29(12)2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38930877

ABSTRACT

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.


Subject(s)
Neurodegenerative Diseases , Protein Processing, Post-Translational , tau Proteins , tau Proteins/metabolism , Humans , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Animals , Drug Development , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/drug therapy , Neurofibrillary Tangles/metabolism , Molecular Targeted Therapy
8.
Commun Biol ; 7(1): 569, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750228

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Receptors, Estrogen , Receptors, G-Protein-Coupled , tau Proteins , Humans , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , tau Proteins/metabolism , tau Proteins/genetics , Female , Male , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/genetics , Aged , Receptors, Estrogen/metabolism , Receptors, Estrogen/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Aged, 80 and over , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Astrocytes/metabolism
9.
Sci Rep ; 14(1): 11533, 2024 05 21.
Article in English | MEDLINE | ID: mdl-38773170

ABSTRACT

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.


Subject(s)
Ferritins , Retina , Tauopathies , tau Proteins , Humans , tau Proteins/metabolism , Ferritins/metabolism , Retina/metabolism , Retina/pathology , Tauopathies/metabolism , Tauopathies/pathology , Tauopathies/diagnosis , Induced Pluripotent Stem Cells/metabolism , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology
10.
BMJ Ment Health ; 27(1)2024 May 24.
Article in English | MEDLINE | ID: mdl-38796179

ABSTRACT

QUESTION: Does neurodegenerative disease underlie the increased rate of dementia observed in older people with schizophrenia? Several studies have reported a higher prevalence of dementia in people with schizophrenia compared with the general population. This may reflect a higher risk of developing neurodegenerative diseases such as vascular dementia or Alzheimer's disease (AD). Alternatively, this may reflect non-pathological, age-related cognitive decline in a population with low cognitive reserve. STUDY SELECTION AND ANALYSIS: We reviewed papers that compared postmortem findings, hippocampal MRI volume or cerebrospinal fluid (CSF) markers of AD, between patients with schizophrenia with evidence of cognitive impairment (age ≥45 years) with controls. We subsequently performed a meta-analysis of postmortem studies that compared amyloid-ß plaques (APs) or neurofibrillary tangles (NFTs) in cognitively impaired patients with schizophrenia to normal controls or an AD group. FINDINGS: No studies found a significant increase of APs or NFTs in cognitively impaired patients with schizophrenia compared with controls. All postmortem studies that compared APs or NFTs in patients with schizophrenia to an AD group found significantly more APs or NFTs in AD. No studies found a significant differences in CSF total tau or phosphorylated tau between patients with schizophrenia and controls. The two studies which compared CSF Aß42 between patients with schizophrenia and controls found significantly decreased CSF Aß42 in schizophrenia compared with controls. Hippocampal volume findings were mixed. CONCLUSIONS: Studies have not found higher rates of AD-related pathology in cognitively impaired individuals with schizophrenia compared with controls. Higher rates of dementia identified in population studies may reflect a lack of specificity in clinical diagnostic tools used to diagnose dementia.


Subject(s)
Biomarkers , Schizophrenia , Humans , Schizophrenia/cerebrospinal fluid , Schizophrenia/pathology , Biomarkers/cerebrospinal fluid , Neurodegenerative Diseases/cerebrospinal fluid , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/diagnostic imaging , Neurodegenerative Diseases/diagnosis , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/pathology , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/diagnosis , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/metabolism , Neurofibrillary Tangles/pathology , Plaque, Amyloid/pathology , Plaque, Amyloid/diagnostic imaging
11.
Alzheimers Dement ; 20(6): 4147-4158, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38747539

ABSTRACT

INTRODUCTION: Typical MRI measures of neurodegeneration have limited sensitivity in early disease stages. Diffusion MRI (dMRI) microstructural measures may allow for detection in preclinical stages. METHODS: Participants had dMRI and either beta-amyloid PET or plasma biomarkers of Alzheimer's pathology within 18 months of MRI. Microstructure was measured in portions of the medial temporal lobe (MTL) with high neurofibrillary tangle (NFT) burden based on a previously developed post mortem 3D-map. Regressions examined relationships between microstructure and markers of Alzheimer's pathology in preclinical disease and then across disease stages. RESULTS: There was higher isometric volume fraction in amyloid-positive compared to amyloid-negative cognitively unimpaired individuals in high tangle MTL regions. Similarly, plasma biomarkers and 18F-flortaucipir were associated with microstructural changes in preclinical disease. Additional microstructural effects were seen across disease stages. DISCUSSION: Combining a post mortem atlas of NFT pathology with microstructural measures allows for detection of neurodegeneration in preclinical Alzheimer's disease. Highlights Typical markers of neurodegeneration are not sensitive in preclinical Alzheimer's. dMRI measured microstructure in regions with high NFT. Microstructural changes occur in medial temporal regions in preclinical disease. Microstructural changes occur in other typical Alzheimer's regions in later stages. Combining post mortem pathology atlases with in vivo MRI is a powerful framework.


Subject(s)
Alzheimer Disease , Biomarkers , Gray Matter , Positron-Emission Tomography , Temporal Lobe , Humans , Alzheimer Disease/pathology , Alzheimer Disease/diagnostic imaging , Temporal Lobe/pathology , Temporal Lobe/diagnostic imaging , Male , Female , Aged , Gray Matter/pathology , Gray Matter/diagnostic imaging , Biomarkers/blood , Amyloid beta-Peptides/metabolism , Neurofibrillary Tangles/pathology , Diffusion Magnetic Resonance Imaging
12.
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
13.
J Alzheimers Dis ; 99(1): 121-143, 2024.
Article in English | MEDLINE | ID: mdl-38640149

ABSTRACT

Background: Previous work from our group has shown that chronic exposure to Vanadium pentoxide (V2O5) causes cytoskeletal alterations suggesting that V2O5 can interact with cytoskeletal proteins through polymerization and tyrosine phosphatases inhibition, causing Alzheimer's disease (AD)-like hippocampal cell death. Objective: This work aims to characterize an innovative AD experimental model through chronic V2O5 inhalation, analyzing the spatial memory alterations and the presence of neurofibrillary tangles (NFTs), amyloid-ß (Aß) senile plaques, cerebral amyloid angiopathy, and dendritic spine loss in AD-related brain structures. Methods: 20 male Wistar rats were divided into control (deionized water) and experimental (0.02 M V2O5 1 h, 3/week for 6 months) groups (n = 10). The T-maze test was used to assess spatial memory once a month. After 6 months, histological alterations of the frontal and entorhinal cortices, CA1, subiculum, and amygdala were analyzed by performing Congo red, Bielschowsky, and Golgi impregnation. Results: Cognitive results in the T-maze showed memory impairment from the third month of V2O5 inhalation. We also noted NFTs, Aß plaque accumulation in the vascular endothelium and pyramidal neurons, dendritic spine, and neuronal loss in all the analyzed structures, CA1 being the most affected. Conclusions: This model characterizes neurodegenerative changes specific to AD. Our model is compatible with Braak AD stage IV, which represents a moment where it is feasible to propose therapies that have a positive impact on stopping neuronal damage.


Subject(s)
Alzheimer Disease , Brain , Disease Models, Animal , Spatial Memory , Vanadium Compounds , Animals , Male , Administration, Inhalation , Alzheimer Disease/chemically induced , Alzheimer Disease/pathology , Amygdala/drug effects , Amygdala/pathology , Brain/drug effects , Brain/pathology , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/pathology , Cerebral Amyloid Angiopathy/chemically induced , Cerebral Amyloid Angiopathy/pathology , Dendritic Spines/drug effects , Dendritic Spines/pathology , Entorhinal Cortex/drug effects , Entorhinal Cortex/pathology , Frontal Lobe/drug effects , Frontal Lobe/pathology , Maze Learning/drug effects , Neurofibrillary Tangles/drug effects , Neurofibrillary Tangles/pathology , Plaque, Amyloid/chemically induced , Plaque, Amyloid/pathology , Rats, Wistar , Spatial Memory/drug effects , Vanadium Compounds/administration & dosage , Vanadium Compounds/toxicity
14.
Acta Neuropathol Commun ; 12(1): 52, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38576010

ABSTRACT

The transcellular propagation of the aberrantly modified protein tau along the functional brain network is a key hallmark of Alzheimer's disease and related tauopathies. Inoculation-based tau propagation models can recapitulate the stereotypical spread of tau and reproduce various types of tau inclusions linked to specific tauopathy, albeit with varying degrees of fidelity. With this systematic review, we underscore the significance of judicious selection and meticulous functional, biochemical, and biophysical characterization of various tau inocula. Furthermore, we highlight the necessity of choosing suitable animal models and inoculation sites, along with the critical need for validation of fibrillary pathology using confirmatory staining, to accurately recapitulate disease-specific inclusions. As a practical guide, we put forth a framework for establishing a benchmark of inoculation-based tau propagation models that holds promise for use in preclinical testing of disease-modifying drugs.


Subject(s)
Alzheimer Disease , Tauopathies , Animals , Alzheimer Disease/pathology , Neurofibrillary Tangles/pathology , Disease Models, Animal , Tauopathies/pathology , tau Proteins/metabolism , Brain/pathology
15.
Brain ; 147(6): 2144-2157, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38667631

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Brain , Cognitive Dysfunction , Positron-Emission Tomography , tau Proteins , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/pathology , Aged , Male , Female , tau Proteins/metabolism , Positron-Emission Tomography/methods , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/pathology , Brain/metabolism , Brain/diagnostic imaging , Brain/pathology , Aged, 80 and over , Neurofibrillary Tangles/pathology , Neurofibrillary Tangles/metabolism , Disease Progression , Aniline Compounds , Cohort Studies , Amyloid beta-Peptides/metabolism , Middle Aged , Longitudinal Studies , Thiazoles , Neuropsychological Tests , Amyloid/metabolism
16.
JAMA Neurol ; 81(6): 619-629, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38619853

ABSTRACT

Importance: Factors associated with clinical heterogeneity in Alzheimer disease (AD) lay along a continuum hypothesized to associate with tangle distribution and are relevant for understanding glial activation considerations in therapeutic advancement. Objectives: To examine clinicopathologic and neuroimaging characteristics of disease heterogeneity in AD along a quantitative continuum using the corticolimbic index (CLix) to account for individuality of spatially distributed tangles found at autopsy. Design, Setting, and Participants: This cross-sectional study was a retrospective medical record review performed on the Florida Autopsied Multiethnic (FLAME) cohort accessioned from 1991 to 2020. Data were analyzed from December 2022 to December 2023. Structural magnetic resonance imaging (MRI) and tau positron emission tomography (PET) were evaluated in an independent neuroimaging group. The FLAME cohort includes 2809 autopsied individuals; included in this study were neuropathologically diagnosed AD cases (FLAME-AD). A digital pathology subgroup of FLAME-AD cases was derived for glial activation analyses. Main Outcomes and Measures: Clinicopathologic factors of heterogeneity that inform patient history and neuropathologic evaluation of AD; CLix score (lower, relative cortical predominance/hippocampal sparing vs higher, relative cortical sparing/limbic predominant cases); neuroimaging measures (ie, structural MRI and tau-PET). Results: Of the 2809 autopsied individuals in the FLAME cohort, 1361 neuropathologically diagnosed AD cases were evaluated. A digital pathology subgroup included 60 FLAME-AD cases. The independent neuroimaging group included 93 cases. Among the 1361 FLAME-AD cases, 633 were male (47%; median [range] age at death, 81 [54-96] years) and 728 were female (53%; median [range] age at death, 81 [53-102] years). A younger symptomatic onset (Spearman ρ = 0.39, P < .001) and faster decline on the Mini-Mental State Examination (Spearman ρ = 0.27; P < .001) correlated with a lower CLix score in FLAME-AD series. Cases with a nonamnestic syndrome had lower CLix scores (median [IQR], 13 [9-18]) vs not (median [IQR], 21 [15-27]; P < .001). Hippocampal MRI volume (Spearman ρ = -0.45; P < .001) and flortaucipir tau-PET uptake in posterior cingulate and precuneus cortex (Spearman ρ = -0.74; P < .001) inversely correlated with CLix score. Although AD cases with a CLix score less than 10 had higher cortical tangle count, we found lower percentage of CD68-activated microglia/macrophage burden (median [IQR], 0.46% [0.32%-0.75%]) compared with cases with a CLix score of 10 to 30 (median [IQR], 0.75% [0.51%-0.98%]) and on par with a CLix score of 30 or greater (median [IQR], 0.40% [0.32%-0.57%]; P = .02). Conclusions and Relevance: Findings show that AD heterogeneity exists along a continuum of corticolimbic tangle distribution. Reduced CD68 burden may signify an underappreciated association between tau accumulation and microglia/macrophages activation that should be considered in personalized therapy for immune dysregulation.


Subject(s)
Alzheimer Disease , Magnetic Resonance Imaging , Neuroglia , Positron-Emission Tomography , Humans , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Male , Female , Aged , Aged, 80 and over , Neuroglia/pathology , Neuroglia/metabolism , Cross-Sectional Studies , Retrospective Studies , Neurofibrillary Tangles/pathology , tau Proteins/metabolism , Middle Aged , Neuroimaging , Cohort Studies , Brain/diagnostic imaging , Brain/pathology , Brain/metabolism , Autopsy
17.
J Alzheimers Dis ; 99(1): 105-111, 2024.
Article in English | MEDLINE | ID: mdl-38607758

ABSTRACT

 Tau accumulation in and neurodegeneration of locus coeruleus (LC) neurons is observed in Alzheimer's disease (AD). We investigated whether tangle and neuronal density in the rostral and caudal LC is characterized by an asymmetric pattern in 77 autopsy cases of the Rush Memory and Aging Project. We found left-right equivalence for tangle density across individuals with and without AD pathology. However, neuronal density, particularly in the caudal-rostral axis of the LC, is asymmetric among individuals with AD pathology. Asymmetry in LC neuronal density may signal advanced disease progression and should be considered in AD neuroimaging studies of LC neurodegeneration.


Subject(s)
Alzheimer Disease , Locus Coeruleus , Humans , Locus Coeruleus/pathology , Alzheimer Disease/pathology , Female , Male , Aged, 80 and over , Aged , Neurons/pathology , Neurons/metabolism , Neurofibrillary Tangles/pathology , tau Proteins/metabolism , Cell Count
18.
Brain ; 147(5): 1667-1679, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38634687

ABSTRACT

Glial fibrillary acidic protein (GFAP), a proxy of astrocyte reactivity, has been proposed as biomarker of Alzheimer's disease. However, there is limited information about the correlation between blood biomarkers and post-mortem neuropathology. In a single-centre prospective clinicopathological cohort of 139 dementia patients, for which the time-frame between GFAP level determination and neuropathological assessment was exceptionally short (on average 139 days), we analysed this biomarker, measured at three time points, in relation to proxies of disease progression such as cognitive decline and brain weight. Most importantly, we investigated the use of blood GFAP to detect the neuropathological hallmarks of Alzheimer's disease, while accounting for potential influences of the most frequent brain co-pathologies. The main findings demonstrated an association between serum GFAP level and post-mortem tau pathology (ß = 12.85; P < 0.001) that was independent of amyloid deposits (ß = 13.23; P = 0.02). A mediation analysis provided additional support for the role of astrocytic activation as a link between amyloid and tau pathology in Alzheimer's disease. Furthermore, a negative correlation was observed between pre-mortem serum GFAP and brain weight at post-mortem (r = -0.35; P < 0.001). This finding, together with evidence of a negative correlation with cognitive assessments (r = -0.27; P = 0.005), supports the role of GFAP as a biomarker for disease monitoring, even in the late phases of Alzheimer's disease. Moreover, the diagnostic performance of GFAP in advanced dementia patients was explored, and its discriminative power (area under the receiver operator characteristic curve at baseline = 0.91) in differentiating neuropathologically-confirmed Alzheimer's disease dementias from non-Alzheimer's disease dementias was determined, despite the challenging scenario of advanced age and frequent co-pathologies in these patients. Independently of Alzheimer's disease, serum GFAP levels were shown to be associated with two other pathologies targeting the temporal lobes-hippocampal sclerosis (ß = 3.64; P = 0.03) and argyrophilic grain disease (ß = -6.11; P = 0.02). Finally, serum GFAP levels were revealed to be correlated with astrocyte reactivity, using the brain GFAP-immunostained area as a proxy (ρ = 0.21; P = 0.02). Our results contribute to increasing evidence suggesting a role for blood GFAP as an Alzheimer's disease biomarker, and the findings offer mechanistic insights into the relationship between blood GFAP and Alzheimer's disease neuropathology, highlighting its ties with tau burden. Moreover, the data highlighting an independent association between serum GFAP levels and other neuropathological lesions provide information for clinicians to consider when interpreting test results. The longitudinal design and correlation with post-mortem data reinforce the robustness of our findings. However, studies correlating blood biomarkers and neuropathological assessments are still scant, and further research is needed to replicate and validate these results in diverse populations.


Subject(s)
Alzheimer Disease , Astrocytes , Atrophy , Biomarkers , Brain , Glial Fibrillary Acidic Protein , Neurofibrillary Tangles , Humans , Glial Fibrillary Acidic Protein/blood , Astrocytes/pathology , Astrocytes/metabolism , Female , Male , Neurofibrillary Tangles/pathology , Aged , Atrophy/pathology , Atrophy/blood , Alzheimer Disease/blood , Alzheimer Disease/pathology , Brain/pathology , Brain/metabolism , Aged, 80 and over , Biomarkers/blood , Autopsy , tau Proteins/blood , Prospective Studies , Middle Aged , Disease Progression , Dementia/blood , Dementia/pathology
19.
Microb Biotechnol ; 17(4): e14462, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38593310

ABSTRACT

Alzheimer's disease is a complex and progressive condition that affects essential neurological functions such as memory and reasoning. In the brain, neuronal loss, synaptic dysfunction, proteinopathy, neurofibrillary tangles, and neuroinflammation are the hallmarks of Alzheimer's disease pathophysiology. In addition, recent evidence has highlighted that microbes, whether commensal or pathogenic, also have the ability to interact with their host and to regulate its immune system, therefore participating in the exchanges that lead to peripheral inflammation and neuropathology. Because of this intimate relationship, bacteria, viruses, fungi, and protozoa have been implicated in the development of Alzheimer's disease. Here, we bring together current and most recent evidence of the role of microbes in Alzheimer's disease, raising burning questions that need to be addressed to guide therapeutic approaches and potential prophylactic strategies.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Neurofibrillary Tangles/pathology , Brain , Inflammation/pathology
20.
Ageing Res Rev ; 96: 102270, 2024 04.
Article in English | MEDLINE | ID: mdl-38484981

ABSTRACT

Among the central features of Alzheimer's disease (AD) progression are altered levels of the neuropeptide somatostatin (SST), and the colocalisation of SST-positive interneurons (SST-INs) with amyloid-ß plaques, leading to cell death. In this theoretical review, I propose a molecular model for the pathogenesis of AD based on SST-IN hypofunction and hyperactivity. Namely, hypofunctional and hyperactive SST-INs struggle to control hyperactivity in medial regions in early stages, leading to axonal Aß production through excessive presynaptic GABAB inhibition, GABAB1a/APP complex downregulation and internalisation. Concomitantly, excessive SST-14 release accumulates near SST-INs in the form of amyloids, which bind to Aß to form toxic mixed oligomers. This leads to differential SST-IN death through excitotoxicity, further disinhibition, SST deficits, and increased Aß release, fibrillation and plaque formation. Aß plaques, hyperactive networks and SST-IN distributions thereby tightly overlap in the brain. Conversely, chronic stimulation of postsynaptic SST2/4 on gulutamatergic neurons by hyperactive SST-INs promotes intense Mitogen-Activated Protein Kinase (MAPK) p38 activity, leading to somatodendritic p-tau staining and apoptosis/neurodegeneration - in agreement with a near complete overlap between p38 and neurofibrillary tangles. This model is suitable to explain some of the principal risk factors and markers of AD progression, including mitochondrial dysfunction, APOE4 genotype, sex-dependent vulnerability, overactive glial cells, dystrophic neurites, synaptic/spine losses, inter alia. Finally, the model can also shed light on qualitative aspects of AD neuropsychology, especially within the domains of spatial and declarative (episodic, semantic) memory, under an overlying pattern of contextual indiscrimination, ensemble instability, interference and generalisation.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Somatostatin/metabolism , Neurons/metabolism , Neurofibrillary Tangles/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...