Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.442
Filter
1.
Brain Nerve ; 76(10): 1119-1125, 2024 Oct.
Article in Japanese | MEDLINE | ID: mdl-39370836

ABSTRACT

Alzheimer's disease (AD) is pathologically characterized by deposition of amyloid plaques (comprising amyloid ß [Aß] protein) and neurofibrillary tangles (comprising tau protein), and neuronal death. Aß monomers aggregate to form oligomers, protofibrils, and mature fibrils. Previously, the mature fibrils and plaques were implicated as contributors to neurotoxicity and neurodegeneration. However, a growing body of evidence proves stronger toxicity of oligomers and protofibrils. Among the many recent phase 3 clinical trials that have investigated the role of anti-Aß antibodies in AD, some have shown the clinical efficacy of aducanumab, lecanemab, and donanemab in these patients. Lecanemab showed selectivity towards protofibrils over fibrils, and donanemab was specifically directed against Aß only in brain-specific amyloid plaques. In contrast, other anti-Aß antibodies did not show efficacy in AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Alzheimer Disease/drug therapy , Alzheimer Disease/immunology , Humans , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Plaque, Amyloid/immunology , Animals
2.
Lancet Neurol ; 23(10): 1025-1034, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39304242

ABSTRACT

Progressive cerebral volume loss on MRI is a hallmark of Alzheimer's disease and has been widely used as an outcome measure in clinical trials, with the prediction that disease-modifying treatments would slow loss. However, in trials of anti-amyloid immunotherapy, the participants who received treatment had excess volume loss. Explanations for this observation range from reduction of amyloid ß plaque burden and related inflammatory changes through to treatment-induced toxicity. The excess volume changes are characteristic of only those immunotherapies that achieve amyloid ß lowering; are compatible with plaque removal; and evidence to date does not suggest an association with harmful effects. Based on the current evidence, we suggest that these changes can be described as amyloid-removal-related pseudo-atrophy. Better understanding of the causes and consequences of these changes is important to enable informed decisions about treatments. Patient-level analyses of data from the trials are urgently needed, along with longitudinal follow-up and neuroimaging data, to determine the long-term trajectory of these volume changes and their clinical correlates. Post-mortem examination of cerebral tissue from treated patients and evaluation of potential correlation with antemortem neuroimaging findings are key priorities.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Atrophy , Brain , Immunotherapy , Humans , Alzheimer Disease/therapy , Alzheimer Disease/diagnostic imaging , Immunotherapy/methods , Amyloid beta-Peptides/metabolism , Brain/diagnostic imaging , Brain/pathology , Plaque, Amyloid/pathology , Magnetic Resonance Imaging
3.
Nat Commun ; 15(1): 8303, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39333107

ABSTRACT

Brain-wide profiling of diverse biological components is fundamental for understanding complex brain pathology. Despite the availability in whole-brain imaging, it is still challenging to conduct multiplexed, brain-wide analysis with current tissue clearing techniques. Here, we propose SOLID, a hydrophobic tissue clearing method that can minimize tissue distortion while offering impressive clearing performance. SOLID achieves high-quality imaging of multi-color labeled mouse brain, and the acquired datasets can be effectively registered to the Allen Brain Atlas via commonly-used algorithms. SOLID enables generation of neural and vascular maps within one mouse brain, as well as tracing of specific neural projections labeled with viruses. SOLID also allows cross-channel investigations of ß-amyloid plaques and neurovascular lesions in the reconstructed all-in-one panorama, providing quantitative insights into structural interactions at different stages of Alzheimer's disease. Altogether, SOLID provides a robust pipeline for whole-brain mapping, which may widen the utility of tissue clearing techniques in diverse neuroscience research.


Subject(s)
Alzheimer Disease , Brain , Plaque, Amyloid , Brain/diagnostic imaging , Brain/pathology , Animals , Mice , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/pathology , Plaque, Amyloid/pathology , Plaque, Amyloid/metabolism , Algorithms , Brain Mapping/methods , Mice, Inbred C57BL , Humans , Male
4.
Endocrinology ; 165(10)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39276028

ABSTRACT

The identification of critical factors that can worsen the mechanisms contributing to the pathophysiology of Alzheimer disease is of paramount importance. Thyroid hormones (TH) fit this criterion. Epidemiological studies have identified an association between altered circulating TH levels and Alzheimer disease. The study of human and animal models indicates that TH can affect all the main cellular, molecular, and genetic mechanisms known as hallmarks of Alzheimer disease. This is true not only for the excessive production in the brain of protein aggregates leading to amyloid plaques and neurofibrillary tangles but also for the clearance of these molecules from the brain parenchyma via the blood-brain barrier and for the escalated process of neuroinflammation-and even for the effects of carrying Alzheimer-associated genetic variants. Suboptimal TH levels result in a greater accumulation of protein aggregates in the brain. The direct TH regulation of critical genes involved in amyloid beta production and clearance is remarkable, affecting the expression of multiple genes, including APP (related to amyloid beta production), APOE, LRP1, TREM2, AQP4, and ABCB1 (related to amyloid beta clearance). TH also affects microglia by increasing their migration and function and directly regulating the immunosuppressor gene CD73, impacting the immune response of these cells. Studies aiming to understand the mechanisms that could explain how changes in TH levels can contribute to the brain alterations seen in patients with Alzheimer disease are ongoing. These studies have potential implications for the management of patients with Alzheimer disease and ultimately can contribute to devising new interventions for these conditions.


Subject(s)
Alzheimer Disease , Thyroid Hormones , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/epidemiology , Alzheimer Disease/etiology , Thyroid Hormones/metabolism , Animals , Brain/metabolism , Amyloid beta-Peptides/metabolism , Plaque, Amyloid/metabolism
5.
J Comp Neurol ; 532(9): e25670, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39315417

ABSTRACT

The brain changes of Alzheimer's disease (AD) include Abeta (Aß) amyloid plaques ("A"), abnormally phosphorylated tau tangles ("T"), and neurodegeneration ("N"). These have been used to construct in vivo and postmortem diagnostic and staging classifications for evaluating the spectrum of AD in the "ATN" and "ABC" ("B" for Braak tau stage, "C" for Consortium to Establish a Registry for Alzheimer's Disease [CERAD] neuritic plaque density) systems. Another common AD feature involves cerebral amyloid angiopathy (CAA). We report the first experiment to examine relationships among cognition, brain distribution of amyloid plaques, CAA, tau/tangles, and magnetic resonance imaging (MRI)-determined volume changes (as a measure of "N") in the same group of behaviorally characterized nonhuman primates. Both ATN and ABC systems were applied to a group of 32 rhesus macaques aged between 7 and 33 years. When an immunohistochemical method for "T" and "B" was used, some monkeys were "triple positive" on ATN, with a maximum ABC status of A1B2C3. With silver or thioflavin S methods, however, all monkeys were classified as T-negative and B0, indicating the absence of mature neurofibrillary tangles (NFTs) and hence neuropathologically defined AD. Although monkeys at extremes of the ATN and ABC classifications, or with frequent CAA, had significantly lower scores on some cognitive tests, the lack of fully mature NFTs or dementia-consistent cognitive impairment indicates that fully developed AD may not occur in rhesus macaques. There were sex differences noted in the types of histopathology present, and only CAA was significantly related to gray matter volume.


Subject(s)
Aging , Alzheimer Disease , Brain , Gray Matter , Macaca mulatta , Magnetic Resonance Imaging , Animals , Alzheimer Disease/pathology , Alzheimer Disease/diagnostic imaging , Male , Female , Magnetic Resonance Imaging/methods , Aging/pathology , Aging/physiology , Humans , Gray Matter/diagnostic imaging , Gray Matter/pathology , Brain/pathology , Brain/diagnostic imaging , Plaque, Amyloid/pathology , Plaque, Amyloid/diagnostic imaging , Neurofibrillary Tangles/pathology , Cognition/physiology , Cognition Disorders/pathology , Cognition Disorders/diagnostic imaging , tau Proteins/metabolism
6.
Nat Commun ; 15(1): 7978, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39266547

ABSTRACT

Systemic amyloidosis involves the deposition of misfolded proteins in organs/tissues, leading to progressive organ dysfunction and failure. Congo red is the gold-standard chemical stain for visualizing amyloid deposits in tissue, showing birefringence under polarization microscopy. However, Congo red staining is tedious and costly to perform, and prone to false diagnoses due to variations in amyloid amount, staining quality and manual examination of tissue under a polarization microscope. We report virtual birefringence imaging and virtual Congo red staining of label-free human tissue to show that a single neural network can transform autofluorescence images of label-free tissue into brightfield and polarized microscopy images, matching their histochemically stained versions. Blind testing with quantitative metrics and pathologist evaluations on cardiac tissue showed that our virtually stained polarization and brightfield images highlight amyloid patterns in a consistent manner, mitigating challenges due to variations in chemical staining quality and manual imaging processes in the clinical workflow.


Subject(s)
Amyloid , Deep Learning , Microscopy, Fluorescence , Staining and Labeling , Humans , Birefringence , Amyloid/metabolism , Microscopy, Fluorescence/methods , Staining and Labeling/methods , Congo Red , Microscopy, Polarization/methods , Amyloidosis/pathology , Amyloidosis/metabolism , Amyloidosis/diagnostic imaging , Optical Imaging/methods , Plaque, Amyloid/pathology , Plaque, Amyloid/metabolism , Plaque, Amyloid/diagnostic imaging , Myocardium/pathology , Myocardium/metabolism
7.
J Infect Dis ; 230(Supplement_2): S165-S172, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39255396

ABSTRACT

BACKGROUND: Toxoplasma gondii infection of Alzheimer's disease model mice decreases amyloid ß plaques. We aimed to determine if there is a brain regional difference in amyloid ß reduction in the brains of T. gondii-infected compared to control mice. METHOD: Three-month-old 5xFAD (AD model) mice were injected with T. gondii or with phosphate-buffered saline as a control. Intact brains were harvested at 6 weeks postinfection, optically cleared using iDISCO+, and brain-wide amyloid burden was visualized using volumetric light-sheet imaging. Amyloid signal was quantified across each brain and computationally mapped to the Allen Institute Brain Reference Atlas to determine amyloid density in each region. RESULTS: A brain-wide analysis of amyloid in control and T. gondii-infected 5xFAD mice revealed that T. gondii infection decreased amyloid burden in the brain globally as well as in the cortex and hippocampus, and many daughter regions. Daughter regions that showed reduced amyloid burden included the prelimbic cortex, visual cortex, and retrosplenial cortex. The olfactory tubercle, a region known to have increased monocytes following T. gondii infection, also showed reduced amyloid after infection. CONCLUSIONS: T. gondii infection of AD mice reduces amyloid burden in a brain region-specific manner that overlaps with known regions of T. gondii infection and peripheral immune cell infiltration.


Subject(s)
Alzheimer Disease , Brain , Disease Models, Animal , Mice, Transgenic , Toxoplasma , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/parasitology , Alzheimer Disease/pathology , Mice , Brain/parasitology , Brain/metabolism , Brain/pathology , Amyloid beta-Peptides/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Toxoplasmosis/metabolism , Female
8.
J Infect Dis ; 230(Supplement_2): S109-S116, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39255392

ABSTRACT

Periodontitis is a chronic inflammatory disease driven by dysbiosis in subgingival microbial communities leading to increased abundance of a limited number of pathobionts, including Porphyromonas gingivalis and Treponema denticola. Oral health, particularly periodontitis, is a modifiable risk factor for Alzheimer disease (AD) pathogenesis, with components of both these bacteria identified in postmortem brains of persons with AD. Repeated oral inoculation of mice with P. gingivalis results in brain infiltration of bacterial products, increased inflammation, and induction of AD-like biomarkers. P. gingivalis displays synergistic virulence with T. denticola during periodontitis. The aim of the current study was to determine the ability of P. gingivalis and T. denticola, grown in physiologically relevant conditions, individually and in combination, to induce AD-like pathology following chronic oral inoculation of female mice over 12 weeks. P. gingivalis alone significantly increased all 7 brain pathologies examined: neuronal damage, activation of astrocytes and microglia, expression of inflammatory cytokines interleukin 1ß (IL-1ß) and interleukin 6 and production of amyloid-ß plaques and hyperphosphorylated tau, in the hippocampus, cortex and midbrain, compared to control mice. T. denticola alone significantly increased neuronal damage, activation of astrocytes and microglia, and expression of IL-1ß, in the hippocampus, cortex and midbrain, compared to control mice. Coinoculation of P. gingivalis with T. denticola significantly increased activation of astrocytes and microglia in the hippocampus, cortex and midbrain, and increased production of hyperphosphorylated tau and IL-1ß in the hippocampus only. The host brain response elicited by oral coinoculation was less than that elicited by each bacterium, suggesting coinoculation was less pathogenic.


Subject(s)
Alzheimer Disease , Bacteroidaceae Infections , Brain , Disease Models, Animal , Porphyromonas gingivalis , Treponema denticola , Animals , Alzheimer Disease/microbiology , Alzheimer Disease/pathology , Mice , Female , Brain/pathology , Brain/microbiology , Bacteroidaceae Infections/microbiology , Periodontitis/microbiology , Periodontitis/pathology , Microglia/microbiology , Treponemal Infections/microbiology , Treponemal Infections/pathology , Mice, Inbred C57BL , Astrocytes/microbiology , Astrocytes/pathology , Plaque, Amyloid/pathology , Plaque, Amyloid/microbiology , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Amyloid beta-Peptides/metabolism
9.
Int J Mol Sci ; 25(15)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39125687

ABSTRACT

Human brain aging is characterized by the production and deposition of ß-amyloid (Aß) in the form of senile plaques and cerebral amyloid angiopathy and the intracellular accumulation of hyper-phosphorylated tau (Hp-tau) to form neurofibrillary tangles (NFTs) and dystrophic neurites of senile plaques. The process progresses for years and eventually manifests as cognitive impairment and dementia in a subgroup of aged individuals. Aß is produced and deposited first in the neocortex in most aged mammals, including humans; it is usually not accompanied by altered behavior and cognitive impairment. Hp-tau is less frequent than Aß pathology, and NFTs are rare in most mammals. In contrast, NFTs are familiar from middle age onward in humans; NFTs first appear in the paleocortex and selected brain stem nuclei. NFTs precede for decades or years Aß deposition and correlate with dementia in about 5% of individuals at the age of 65 and 25% at the age of 85. Based on these comparative data, (a) Aß deposition is the most common Alzheimer's disease neuropathological change (ADNC) in the brain of aged mammals; (b) Hp-tau is less common, and NFTs are rare in most aged mammals; however, NFTs are the principal cytoskeletal pathology in aged humans; (c) NFT in aged humans starts in selected nuclei of the brain stem and paleocortical brain regions progressing to the most parts of the neocortex and other regions of the telencephalon; (d) human brain aging is unique among mammalian species due to the early appearance and dramatic progression of NFTs from middle age onward, matching with cognitive impairment and dementia in advanced cases; (e) neither mammalian nor human brain aging supports the concept of the amyloid cascade hypothesis.


Subject(s)
Aging , Alzheimer Disease , Neurofibrillary Tangles , tau Proteins , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Animals , Humans , Aging/pathology , Aging/metabolism , Neurofibrillary Tangles/pathology , Neurofibrillary Tangles/metabolism , tau Proteins/metabolism , Brain/pathology , Brain/metabolism , Amyloid beta-Peptides/metabolism , Mammals/metabolism , Plaque, Amyloid/pathology , Plaque, Amyloid/metabolism
10.
Acta Neuropathol Commun ; 12(1): 134, 2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39154006

ABSTRACT

Accurate and scalable quantification of amyloid-ß (Aß) pathology is crucial for deeper disease phenotyping and furthering research in Alzheimer Disease (AD). This multidisciplinary study addresses the current limitations on neuropathology by leveraging a machine learning (ML) pipeline to perform a granular quantification of Aß deposits and assess their distribution in the temporal lobe. Utilizing 131 whole-slide-images from consecutive autopsied cases at the University of California Davis Alzheimer Disease Research Center, our objectives were threefold: (1) Validate an automatic workflow for Aß deposit quantification in white matter (WM) and gray matter (GM); (2) define the distributions of different Aß deposit types in GM and WM, and (3) investigate correlates of Aß deposits with dementia status and the presence of mixed pathology. Our methodology highlights the robustness and efficacy of the ML pipeline, demonstrating proficiency akin to experts' evaluations. We provide comprehensive insights into the quantification and distribution of Aß deposits in the temporal GM and WM revealing a progressive increase in tandem with the severity of established diagnostic criteria (NIA-AA). We also present correlations of Aß load with clinical diagnosis as well as presence/absence of mixed pathology. This study introduces a reproducible workflow, showcasing the practical use of ML approaches in the field of neuropathology, and use of the output data for correlative analyses. Acknowledging limitations, such as potential biases in the ML model and current ML classifications, we propose avenues for future research to refine and expand the methodology. We hope to contribute to the broader landscape of neuropathology advancements, ML applications, and precision medicine, paving the way for deep phenotyping of AD brain cases and establishing a foundation for further advancements in neuropathological research.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Machine Learning , Temporal Lobe , Humans , Temporal Lobe/pathology , Temporal Lobe/metabolism , Amyloid beta-Peptides/metabolism , Female , Male , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Tissue Banks , Gray Matter/pathology , Gray Matter/metabolism , White Matter/pathology , White Matter/metabolism , Plaque, Amyloid/pathology , Plaque, Amyloid/metabolism , Middle Aged
11.
Sci Adv ; 10(32): eadn5181, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39110801

ABSTRACT

Episodic memory in older adults is varied and perceived to rely on numbers of synapses or dendritic spines. We analyzed 2157 neurons among 128 older individuals from the Religious Orders Study and Rush Memory and Aging Project. Analysis of 55,521 individual dendritic spines by least absolute shrinkage and selection operator regression and nested model cross-validation revealed that the dendritic spine head diameter in the temporal cortex, but not the premotor cortex, improved the prediction of episodic memory performance in models containing ß amyloid plaque scores, neurofibrillary tangle pathology, and sex. These findings support the emerging hypothesis that, in the temporal cortex, synapse strength is more critical than quantity for memory in old age.


Subject(s)
Dendritic Spines , Memory, Episodic , Humans , Dendritic Spines/physiology , Male , Female , Aged , Aged, 80 and over , Aging/physiology , Temporal Lobe/physiology , Plaque, Amyloid/pathology
12.
Life Sci ; 355: 122973, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39142510

ABSTRACT

AIMS: Microglial cells are integral to the pathogenesis of Alzheimer's disease (AD). The observed sex disparity in AD prevalence, with a notable predominance in women, implies a potential influence of sex hormones, such as androgens, on disease mechanisms. Despite this, the specific effects of androgens on microglia remain unclear. This study is designed to delineate the interplay between androgens and the survival and inflammatory profile of microglial cells, as well as to explore their contribution to the progression of AD. METHODS AND KEY FINDINGS: To create a chronic androgen deficiency model, 3-month-old wild-type (WT) mice and APP/PS1 mice underwent bilateral orchiectomy (ORX), with age-matched sham-operated controls. Cognitive and memory were evaluated at 5 and 12 months, paralleled by assessments of amyloid-beta (Aß) and microglial morphology in hippocampal and cortical areas. The ORX treatment in mice resulted in diminished microglial populations and morphological alterations, alongside an increase in Aß plaques and a concomitant decline in cognitive performance that exacerbated over time. In vitro, dihydrotestosterone (DHT) was found to stimulate microglial proliferation and ameliorate Aß1-42-induced apoptosis. SIGNIFICANCE: These findings suggested that androgens may exert a protective role, maintaining the normal proliferation and functionality of microglial cells. This preservation could potentially slow the progression of AD. As a result, our study provided a conceptual framework for the development of novel therapeutic strategies for AD.


Subject(s)
Alzheimer Disease , Androgens , Mice, Transgenic , Microglia , Animals , Microglia/pathology , Microglia/metabolism , Microglia/drug effects , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Male , Mice , Androgens/pharmacology , Androgens/metabolism , Androgens/deficiency , Orchiectomy , Amyloid beta-Peptides/metabolism , Mice, Inbred C57BL , Dihydrotestosterone/pharmacology , Disease Models, Animal , Hippocampus/pathology , Hippocampus/metabolism , Apoptosis/drug effects , Cell Proliferation/drug effects , Age Factors , Plaque, Amyloid/pathology , Plaque, Amyloid/metabolism
13.
Nat Neurosci ; 27(10): 1880-1891, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39187705

ABSTRACT

Proteomics can shed light on the dynamic and multifaceted alterations in neurodegenerative disorders like Alzheimer's disease (AD). Combining radioligands measuring ß-amyloid (Aß) plaques and tau tangles with cerebrospinal fluid proteomics, we uncover molecular events mirroring different stages of AD pathology in living humans. We found 127 differentially abundant proteins (DAPs) across the AD spectrum. The strongest Aß-related proteins were mainly expressed in glial cells and included SMOC1 and ITGAM. A dozen proteins linked to ATP metabolism and preferentially expressed in neurons were independently associated with tau tangle load and tau accumulation. Only 20% of the DAPs were also altered in other neurodegenerative diseases, underscoring AD's distinct proteome. Two co-expression modules related, respectively, to protein metabolism and microglial immune response encompassed most DAPs, with opposing, staggered trajectories along the AD continuum. We unveil protein signatures associated with Aß and tau proteinopathy in vivo, offering insights into complex neural responses and potential biomarkers and therapeutics targeting different disease stages.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Neurofibrillary Tangles , Plaque, Amyloid , Proteomics , tau Proteins , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Humans , tau Proteins/metabolism , Plaque, Amyloid/pathology , Plaque, Amyloid/metabolism , Proteomics/methods , Female , Aged , Male , Amyloid beta-Peptides/metabolism , Neurofibrillary Tangles/pathology , Neurofibrillary Tangles/metabolism , Aged, 80 and over , Proteome/metabolism , Brain/metabolism , Brain/pathology , Biomarkers/metabolism
14.
J Neurochem ; 168(9): 3235-3249, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39115041

ABSTRACT

The accumulation of ß-amyloid in Alzheimer's disease greatly impacts neuronal health and synaptic function. To maintain network stability in the face of altered synaptic activity, neurons engage a feedback mechanism termed homeostatic scaling; however, this process is thought to be disrupted during disease progression. Previous proteomics studies have shown that one of the most highly regulated proteins in cell culture models of homeostatic scaling is the small secretory chaperone proSAAS. Our prior work has shown that proSAAS exhibits anti-aggregant behavior against alpha-synuclein and ß-amyloid fibrillation in vitro and is up-regulated in cell models of proteostatic stress. However, the specific role that this protein might play in homeostatic scaling, and its anti-aggregant role in Alzheimer's progression, is not clear. To learn more about the role of proSAAS in maintaining hippocampal proteostasis, we compared its expression in a primary neuron model of homeostatic scaling to other synaptic components using western blotting and qPCR, revealing that proSAAS protein responses to homeostatic up- and down-regulation were significantly higher than those of two other synaptic vesicle components, 7B2 and carboxypeptidase E. However, proSAAS mRNA expression was static, suggesting translational control and/or altered protein degradation. ProSAAS was readily released upon depolarization of differentiated hippocampal cultures, supporting its synaptic localization. Immunohistochemical analysis demonstrated abundant proSAAS within the mossy fiber layer of the hippocampus in both wild-type and 5xFAD mice; in the latter, proSAAS was also concentrated around amyloid plaques. Importantly, overexpression of proSAAS in the CA1 region via stereotaxic injection of proSAAS-encoding AAV2/1 significantly decreased amyloid plaque burden in 5xFAD mice. We hypothesize that dynamic changes in proSAAS expression play a critical role in hippocampal proteostatic processes, both in the context of normal homeostatic plasticity and in the control of protein aggregation during Alzheimer's disease progression.


Subject(s)
Alzheimer Disease , Hippocampus , Homeostasis , Mice, Transgenic , Plaque, Amyloid , Up-Regulation , Animals , Mice , Hippocampus/metabolism , Hippocampus/pathology , Plaque, Amyloid/pathology , Plaque, Amyloid/metabolism , Homeostasis/physiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Male , Humans , Mice, Inbred C57BL , Female , Cells, Cultured , Neurons/metabolism , Neurons/pathology
15.
JAMA Neurol ; 81(10): 1066-1072, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39158850

ABSTRACT

Importance: Anti-ß-amyloid immunotherapy using lecanemab is becoming increasingly available to patients with Alzheimer disease (AD). Individuals with Down syndrome (DS) develop AD neuropathology by age 40 years, representing a significant cohort of genetically determined AD. Objective: To investigate the binding properties of lecanemab in the brains of people with DS, in anticipation of their inclusion in clinical trials or access to antiamyloid immunotherapies. Design, Setting, Participants: The study included cases of postmortem brain tissue analysis from 15 individuals with DS aged 43 to 68 years that were acquired from Alzheimer Disease research centers at the University of California, Irvine and the University of Kentucky from 2008 to 2021. Data were analyzed from August 2023 through May 2024. Exposure: The binding properties of lecanemab were assessed in brain tissue. Main Outcome: The primary outcome was the extent of lecanemab binding to amyloid plaques and brain blood vessels. Results: Tissue from 15 people (8 were female [53%]) with DS ranging in age from 43 to 68 (mean, 56.6) years were included in the study. Lecanemab-labeled amyloid plaques appeared in all 15 DS cases studied, indicating potential target engagement. However, extensive binding of lecanemab to brain blood vessels in DS was observed, raising significant safety concerns. These findings underscore the necessity for clinical trials of lecanemab in people with DS to evaluate both safety and efficacy, particularly in individuals older than 43 years. Conclusions and Relevance: These findings suggest significant binding of lecanemab to cerebral amyloid angiopathy in DS. Lecanemab should be rigorously tested in clinical trials for AD in the DS population to determine its safety and efficacy, especially in those older than 43 years.


Subject(s)
Brain , Down Syndrome , Plaque, Amyloid , Humans , Down Syndrome/metabolism , Down Syndrome/pathology , Female , Male , Middle Aged , Adult , Brain/metabolism , Brain/diagnostic imaging , Brain/pathology , Aged , Plaque, Amyloid/pathology , Plaque, Amyloid/metabolism , Amyloid beta-Peptides/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology
16.
Mol Metab ; 89: 102019, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39216535

ABSTRACT

OBJECTIVE: The development of glucagon-like peptide-1 receptor (GLP-1R) agonists for the treatment of type 2 diabetes and obesity has been accompanied by evidence for anti-inflammatory and cytoprotective actions in the heart, blood vessels, kidney, and brain. Whether GLP-1R agonists might be useful clinically for attenuating deterioration of cognitive dysfunction and reducing the progression of Alzheimer's disease remains uncertain. METHODS: Here we evaluated the actions of semaglutide and tirzepatide, clinically distinct GLP-1 medicines, in two mouse models of neurodegeneration. RESULTS: Semaglutide reduced body weight and improved glucose tolerance in 12-month-old male and female 5XFAD and APP/PS1 mice, consistent with pharmacological engagement of the GLP-1R. Nevertheless, amyloid plaque density was not different in the cerebral cortex, hippocampus, or subiculum of semaglutide-treated 12-month-old 5XFAD and APP/PS1 mice. IBA1 and GFAP expression were increased in the hippocampus of 5XFAD and APP/PS1 mice but were not reduced by semaglutide. Moreover, parameters of neurobehavioral and cognitive function evaluated using Open Field testing or the Morris water maze were not improved following treatment with semaglutide. To explore whether incretin therapies might be more effective in younger mice, we studied semaglutide and tirzepatide action in 6-month-old male and female 5XFAD mice. Neither semaglutide nor tirzepatide modified the extent of plaque accumulation, hippocampal IBA1+ or GFAP+ cells, or parameters of neurobehavioral testing, despite improving glucose tolerance and reducing body weight. mRNA biomarkers of inflammation and neurodegeneration were increased in the hippocampus of male and female 5XFAD mice but were not reduced after treatment with semaglutide or tirzepatide. CONCLUSIONS: Collectively, these findings reveal preservation of the metabolic actions of two GLP-1 medicines, semaglutide and tirzepatide, yet inability to detect improvement in structural and functional parameters of neurodegeneration in two mouse models of Alzheimer's disease.


Subject(s)
Alzheimer Disease , Cognition , Glucagon-Like Peptides , Mice, Transgenic , Animals , Mice , Glucagon-Like Peptides/pharmacology , Glucagon-Like Peptides/therapeutic use , Male , Female , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Cognition/drug effects , Disease Models, Animal , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Plaque, Amyloid/metabolism , Plaque, Amyloid/drug therapy , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Glucagon-Like Peptide 1/metabolism
17.
Acta Neuropathol ; 148(1): 20, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39147931

ABSTRACT

Cotton wool plaques (CWPs) have been described as features of the neuropathologic phenotype of dominantly inherited Alzheimer disease (DIAD) caused by some missense and deletion mutations in the presenilin 1 (PSEN1) gene. CWPs are round, eosinophilic amyloid-ß (Aß) plaques that lack an amyloid core and are recognizable, but not fluorescent, in Thioflavin S (ThS) preparations. Amino-terminally truncated and post-translationally modified Aß peptide species are the main component of CWPs. Tau immunopositive neurites may be present in CWPs. In addition, neurofibrillary tangles coexist with CWPs. Herein, we report the structure of Aß and tau filaments isolated from brain tissue of individuals affected by DIAD caused by the PSEN1 V261I and A431E mutations, with the CWP neuropathologic phenotype. CWPs are predominantly composed of type I Aß filaments present in two novel arrangements, type Ic and type Id; additionally, CWPs contain type I and type Ib Aß filaments. Tau filaments have the AD fold, which has been previously reported in sporadic AD and DIAD. The formation of type Ic and type Id Aß filaments may be the basis for the phenotype of CWPs. Our data are relevant for the development of PET imaging methodologies to best detect CWPs in DIAD.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Plaque, Amyloid , Presenilin-1 , tau Proteins , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Humans , Plaque, Amyloid/pathology , Plaque, Amyloid/metabolism , tau Proteins/metabolism , tau Proteins/genetics , Amyloid beta-Peptides/metabolism , Presenilin-1/genetics , Brain/pathology , Brain/metabolism , Brain/diagnostic imaging , Mutation , Female , Male
18.
Cell Rep Med ; 5(8): 101683, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39168096

ABSTRACT

Levites et al. demonstrate that mouse models of Alzheimer disease (AD), exhibiting amyloid-beta (Αß) plaque formation, share Αß responsome proteins with humans. Their work underscores the value of these models in studying Αß aggregation, cellular vulnerability, and early-stage AD pathology.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Proteome , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Animals , Humans , Proteome/metabolism , Mice , Amyloid beta-Peptides/metabolism , Plaque, Amyloid/pathology , Plaque, Amyloid/metabolism
19.
Neuroscience ; 557: 56-66, 2024 Oct 04.
Article in English | MEDLINE | ID: mdl-39127343

ABSTRACT

The anterior thalamic nuclei are important for cognition, and memory in particular. However, little is known about how the anterior thalamic nuclei are affected in many neurological disorders partly due to difficulties in selective segmentation in in vivo scans, due to their size and location. Post-mortem studies, therefore, remain a valuable source of information about the status of the anterior thalamic nuclei. We used post-mortem tissue to assess the status of the anteroventral thalamic nucleus in Down syndrome using samples from males and females ranging from 22-65 years in age and comparing to tissue from age matched controls. As expected, there was increased beta-amyloid plaque expression in the Down syndrome group. While there was a significant increase in neuronal density in the Down syndrome group, the values showed more variation consistent with a heterogeneous population. The surface area of the anteroventral thalamic nucleus was smaller in the Down syndrome group suggesting the increased neuronal density was due to greater neuronal packing but likely fewer overall neurons. There was a marked reduction in the proportion of neurons immunoreactive for the calcium-binding proteins calbindin, calretinin, and parvalbumin in individuals with Down syndrome. These findings highlight the vulnerability of calcium-binding proteins in the anteroventral nucleus in Down syndrome, which could both be driven by, and exacerbate, Alzheimer-related pathology in this region.


Subject(s)
Anterior Thalamic Nuclei , Down Syndrome , Neurons , Humans , Down Syndrome/metabolism , Down Syndrome/pathology , Male , Female , Middle Aged , Adult , Aged , Anterior Thalamic Nuclei/metabolism , Anterior Thalamic Nuclei/pathology , Neurons/metabolism , Neurons/pathology , Young Adult , Calcium-Binding Proteins/metabolism , Parvalbumins/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology
20.
Cells ; 13(15)2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39120323

ABSTRACT

Alzheimer's disease is the primary neurodegenerative disease affecting the elderly population. Despite the first description of its pathology over a century ago, its precise cause and molecular mechanism remain unknown. Numerous factors, including beta-amyloid, tau protein, the APOEε4 gene, and different metals, have been extensively investigated in relation to this disease. However, none of them have been proven to have a decisive causal relationship. Furthermore, no single theory has successfully integrated these puzzle pieces thus far. In this review article, we propose the most probable molecular mechanism for AD, which clearly shows the relationship between the main aspects of the disease, and addresses fundamental questions such as: Why is aging the major risk factor for the disease? Are amyloid plaques and tau tangles the causes or consequences of AD? Why are the distributions of senile plaques and tau tangles in the brain different and independent of each other? Why is the APOEε4 gene a risk factor for AD? Finally, why is the disease more prevalent in women?


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , tau Proteins , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , tau Proteins/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Animals , Brain/metabolism , Brain/pathology , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL