Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.662
Filter
1.
Acta Neuropathol ; 147(1): 105, 2024 06 19.
Article in English | MEDLINE | ID: mdl-38896306

ABSTRACT

Alzheimer's disease (AD) is a progressive neurological condition characterized by impaired cognitive function and behavioral alterations. While AD research historically centered around mis-folded proteins, advances in mass spectrometry techniques have triggered increased exploration of the AD lipidome with lipid dysregulation emerging as a critical player in AD pathogenesis. Gangliosides are a class of glycosphingolipids enriched within the central nervous system. Previous work has suggested a shift in a-series gangliosides from complex (GM1) to simple (GM2 and GM3) species may be related to the development of neurodegenerative disease. In addition, complex gangliosides with 20 carbon sphingosine chains have been shown to increase in the aging brain. In this study, we utilized matrix-assisted laser desorption ionization mass spectrometry imaging (MALDI-MSI) to interrogate the in situ relationship of a-series gangliosides with either 18 or 20 carbon sphingosine chains (d18:1 or d20:1, respectively) in the post-mortem human AD brain. Here, we expanded upon previous literature and demonstrated a significant decrease in the GM1 d20:1 to GM1 d18:1 ratio in regions of the dentate gyrus and entorhinal cortex in AD relative to control brain tissue. Then, we demonstrated that the MALDI-MSI profile of GM3 co-localizes with histologically confirmed amyloid beta (Aß) plaques and found a significant increase in both GM1 and GM3 in proximity to Aß plaques. Collectively, this study demonstrates a perturbation of the ganglioside profile in AD, and validates a pipeline for MALDI-MSI and classic histological staining in the same tissue sections. This demonstrates feasibility for integrating untargeted mass spectrometry imaging approaches into a digital pathology framework.


Subject(s)
Alzheimer Disease , Gangliosides , Plaque, Amyloid , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Humans , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Gangliosides/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Plaque, Amyloid/pathology , Plaque, Amyloid/metabolism , Aged , Aged, 80 and over , Brain/pathology , Brain/metabolism , Male , Female
2.
Nat Commun ; 15(1): 5000, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38866763

ABSTRACT

To date, earlier diagnosis of Alzheimer's disease (AD) is still challenging. Recent studies revealed the elevated expression of connective tissue growth factor (CTGF) in AD brain is an upstream regulator of amyloid-beta (Aß) plaque, thus CTGF could be an earlier diagnostic biomarker of AD than Aß plaque. Herein, we develop a peptide-coated gold nanocluster that specifically targets CTGF with high affinity (KD ~ 21.9 nM). The probe can well penetrate the blood-brain-barrier (BBB) of APP/PS1 transgenic mice at early-stage (earlier than 3-month-old) in vivo, allowing non-invasive NIR-II imaging of CTGF when there is no appearance of Aß plaque deposition. Notably, this probe can also be applied to measuring CTGF on postmortem brain sections by multimodal analysis, including fluorescence imaging, peroxidase-like chromogenic imaging, and ICP-MS quantitation, which enables distinguishment between the brains of AD patients and healthy people. This probe possesses great potential for precise diagnosis of earlier AD before Aß plaque formation.


Subject(s)
Alzheimer Disease , Brain , Connective Tissue Growth Factor , Mice, Transgenic , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Connective Tissue Growth Factor/metabolism , Animals , Humans , Mice , Brain/diagnostic imaging , Brain/metabolism , Brain/pathology , Gold/chemistry , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/diagnostic imaging , Plaque, Amyloid/diagnostic imaging , Plaque, Amyloid/metabolism , Metal Nanoparticles/chemistry , Disease Models, Animal , Amyloid beta-Peptides/metabolism , Female , Male , Multimodal Imaging/methods , Biomarkers/metabolism , Optical Imaging/methods
3.
Int J Mol Sci ; 25(11)2024 May 31.
Article in English | MEDLINE | ID: mdl-38892278

ABSTRACT

Alzheimer's disease (AD) presents a significant challenge due to its multifaceted nature, characterized by cognitive decline, memory loss, and neuroinflammation. Though AD is an extensively researched topic, effective pharmacological interventions remain elusive, prompting explorations into non-pharmacological approaches. Microcurrent (MC) therapy, which utilizes imperceptible currents, has emerged as a potent clinical protocol. While previous studies have focused on its therapeutic effects, this study investigates the impact of MC on neuronal damage and neuroinflammation in an AD mouse model, specifically addressing potential side effects. Utilizing 5xFAD transgenic mice, we examined the effects of MC therapy on neuronal integrity and inflammation. Our findings suggest that MC therapy attenuates memory impairment and reduces neurodegeneration, as evidenced by improved performance in memory tests and the preservation of the neuronal structure. Additionally, MC therapy significantly decreases amyloid-beta (Aß) plaque deposition and inhibits apoptosis, indicating its potential to mitigate AD pathology. This study determined that glial activation is effectively reduced by using MC therapy to suppress the TLR4-MyD88-NFκB pathway, which consequently causes the levels of inflammatory factors TNF-α, IL-1ß, and IL-6 to decrease, thus implicating TLR4 in neurodegenerative disease-related neuroinflammation. Furthermore, while our study did not observe significant adverse effects, a further clinical trial into potential side effects and neuroinflammatory responses associated with MC therapy is warranted.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Disease Models, Animal , Mice, Transgenic , Neurons , Animals , Alzheimer Disease/therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Mice , Cognitive Dysfunction/therapy , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Neurons/metabolism , Neurons/pathology , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , Toll-Like Receptor 4/metabolism , Amyloid beta-Peptides/metabolism , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/pathology , Plaque, Amyloid/pathology , Plaque, Amyloid/metabolism , NF-kappa B/metabolism , Apoptosis
4.
Alzheimers Res Ther ; 16(1): 121, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38831312

ABSTRACT

BACKGROUND: Beta-amyloid (Aß) deposition in the brain parenchyma is a crucial initiating step in the amyloid cascade hypothesis of Alzheimer's disease (AD) pathology. Furthermore, dysfunction of plaque-associated microglia, also known as disease-associated microglia (DAM) has been reported to accelerate Aß deposition and cognitive impairment. Our previous research demonstrated that intermittent hypoxia training (IHT) improved AD pathology by upregulating autophagy in DAM, thereby enhancing oligomeric Aß (oAß) clearance. Considering that oAß internalization is the initial stage of oAß clearance, this study focused on the IHT mechanism involved in upregulating Aß uptake by DAM. METHODS: IHT was administered to 8-month-old APP/PS1 mice or 6-month-old microglial vacuolar protein sorting 35 (VPS35) knockout mice in APP/PS1 background (MG VPS35 KO: APP/PS1) for 28 days. After the IHT, the spatial learning-memory capacity of the mice was assessed. Additionally, AD pathology was determined by estimating the nerve fiber and synapse density, Aß plaque deposition, and Aß load in the brain. A model of Aß-exposed microglia was constructed and treated with IHT to explore the related mechanism. Finally, triggering receptor expressed on myeloid cells 2 (TREM2) intracellular recycling and Aß internalization were measured using a fluorescence tracing technique. RESULTS: Our results showed that IHT ameliorated cognitive function and Aß pathology. In particular, IHT enhanced Aß endocytosis by augmenting the intracellular transport function of microglial TREM2, thereby contributing to Aß clearance. Furthermore, IHT specifically upregulated VPS35 in DAM, the primary cause for the enhanced intracellular recycling of TREM2. IHT lost ameliorative effect on Aß pathology in MG VPS35 KO: APP/PS1 mice brain. Lastly, the IHT mechanism of VPS35 upregulation in DAM was mediated by the transcriptional regulation of VPS35 by transcription factor EB (TFEB). CONCLUSION: IHT enhances Aß endocytosis in DAM by upregulating VPS35-dependent TREM2 recycling, thereby facilitating oAß clearance and mitigation of Aß pathology. Moreover, the transcriptional regulation of VPS35 by TFEB demonstrates a close link between endocytosis and autophagy in microglia. Our study further elucidates the IHT mechanism in improving AD pathology and provides evidence supporting the potential application of IHT as a complementary therapy for AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Endocytosis , Membrane Glycoproteins , Microglia , Plaque, Amyloid , Receptors, Immunologic , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Microglia/metabolism , Mice , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Amyloid beta-Peptides/metabolism , Endocytosis/physiology , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , Mice, Transgenic , Hypoxia/metabolism , Mice, Knockout , Disease Models, Animal , Male , Brain/metabolism , Brain/pathology , Mice, Inbred C57BL
5.
Anal Chem ; 96(24): 9799-9807, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38830618

ABSTRACT

Cerebral accumulation of amyloid-ß (Aß) initiates molecular and cellular cascades that lead to Alzheimer's disease (AD). However, amyloid deposition does not invariably lead to dementia. Amyloid-positive but cognitively unaffected (AP-CU) individuals present widespread amyloid pathology, suggesting that molecular signatures more complex than the total amyloid burden are required to better differentiate AD from AP-CU cases. Motivated by the essential role of Aß and the key lipid involvement in AD pathogenesis, we applied multimodal mass spectrometry imaging (MSI) and machine learning (ML) to investigate amyloid plaque heterogeneity, regarding Aß and lipid composition, in AP-CU versus AD brain samples at the single-plaque level. Instead of focusing on a population mean, our analytical approach allowed the investigation of large populations of plaques at the single-plaque level. We found that different (sub)populations of amyloid plaques, differing in Aß and lipid composition, coexist in the brain samples studied. The integration of MSI data with ML-based feature extraction further revealed that plaque-associated gangliosides GM2 and GM1, as well as Aß1-38, but not Aß1-42, are relevant differentiators between the investigated pathologies. The pinpointed differences may guide further fundamental research investigating the role of amyloid plaque heterogeneity in AD pathogenesis/progression and may provide molecular clues for further development of emerging immunotherapies to effectively target toxic amyloid assemblies in AD therapy. Our study exemplifies how an integrative analytical strategy facilitates the unraveling of complex biochemical phenomena, advancing our understanding of AD from an analytical perspective and offering potential avenues for the refinement of diagnostic tools.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Brain , Plaque, Amyloid , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/diagnosis , Humans , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/analysis , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Plaque, Amyloid/chemistry , Brain/metabolism , Brain/pathology , Lipids/analysis , Lipids/chemistry , Machine Learning , Aged
6.
Nat Commun ; 15(1): 3996, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38734693

ABSTRACT

SPI1 was recently reported as a genetic risk factor for Alzheimer's disease (AD) in large-scale genome-wide association studies. However, it is unknown whether SPI1 should be downregulated or increased to have therapeutic benefits. To investigate the effect of modulating SPI1 levels on AD pathogenesis, we performed extensive biochemical, histological, and transcriptomic analyses using both Spi1-knockdown and Spi1-overexpression mouse models. Here, we show that the knockdown of Spi1 expression significantly exacerbates insoluble amyloid-ß (Aß) levels, amyloid plaque deposition, and gliosis. Conversely, overexpression of Spi1 significantly ameliorates these phenotypes and dystrophic neurites. Further mechanistic studies using targeted and single-cell transcriptomics approaches demonstrate that altered Spi1 expression modulates several pathways, such as immune response pathways and complement system. Our data suggest that transcriptional reprogramming by targeting transcription factors, like Spi1, might hold promise as a therapeutic strategy. This approach could potentially expand the current landscape of druggable targets for AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Amyloidosis , Proto-Oncogene Proteins , Trans-Activators , Transcriptome , Animals , Male , Mice , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloidosis/genetics , Amyloidosis/metabolism , Amyloidosis/pathology , Disease Models, Animal , Gene Expression Profiling , Gene Knockdown Techniques , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Plaque, Amyloid/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Trans-Activators/metabolism
7.
FEBS Lett ; 598(13): 1591-1604, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38724485

ABSTRACT

Inhibition of the cyclic-AMP degrading enzyme phosphodiesterase type 4 (PDE4) in the brains of animal models is protective in Alzheimer's disease (AD). We show for the first time that enzymes from the subfamily PDE4D not only colocalize with beta-amyloid (Aß) plaques in a mouse model of AD but that Aß directly associates with the catalytic machinery of the enzyme. Peptide mapping suggests that PDE4D is the preferential PDE4 subfamily for Aß as it possesses a unique binding site. Intriguingly, exogenous addition of Aß to cells overexpressing the PDE4D5 longform caused PDE4 activation and a decrease in cAMP. We suggest a novel mechanism where PDE4 longforms can be activated by Aß, resulting in the attenuation of cAMP signalling to promote loss of cognitive function in AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Cyclic AMP , Cyclic Nucleotide Phosphodiesterases, Type 4 , Neurons , Animals , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Amyloid beta-Peptides/metabolism , Cyclic AMP/metabolism , Mice , Neurons/metabolism , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Protein Binding , Enzyme Activation , Mice, Transgenic , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology
8.
Mol Neurodegener ; 19(1): 42, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802940

ABSTRACT

Microglia play diverse pathophysiological roles in Alzheimer's disease (AD), with genetic susceptibility factors skewing microglial cell function to influence AD risk. CD33 is an immunomodulatory receptor associated with AD susceptibility through a single nucleotide polymorphism that modulates mRNA splicing, skewing protein expression from a long protein isoform (CD33M) to a short isoform (CD33m). Understanding how human CD33 isoforms differentially impact microglial cell function in vivo has been challenging due to functional divergence of CD33 between mice and humans. We address this challenge by studying transgenic mice expressing either of the human CD33 isoforms crossed with the 5XFAD mouse model of amyloidosis and find that human CD33 isoforms have opposing effects on the response of microglia to amyloid-ß (Aß) deposition. Mice expressing CD33M have increased Aß levels, more diffuse plaques, fewer disease-associated microglia, and more dystrophic neurites compared to 5XFAD control mice. Conversely, CD33m promotes plaque compaction and microglia-plaque contacts, and minimizes neuritic plaque pathology, highlighting an AD protective role for this isoform. Protective phenotypes driven by CD33m are detected at an earlier timepoint compared to the more aggressive pathology in CD33M mice that appears at a later timepoint, suggesting that CD33m has a more prominent impact on microglia cell function at earlier stages of disease progression. In addition to divergent roles in modulating phagocytosis, scRNAseq and proteomics analyses demonstrate that CD33m+ microglia upregulate nestin, an intermediate filament involved in cell migration, at plaque contact sites. Overall, our work provides new functional insights into how CD33, as a top genetic susceptibility factor for AD, modulates microglial cell function.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Mice, Transgenic , Microglia , Protein Isoforms , Sialic Acid Binding Ig-like Lectin 3 , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Microglia/metabolism , Sialic Acid Binding Ig-like Lectin 3/metabolism , Humans , Mice , Protein Isoforms/metabolism , Amyloid beta-Peptides/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology
9.
Biomed Pharmacother ; 175: 116616, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38723516

ABSTRACT

Fluorescent probes are a powerful tool for imaging amyloid ß (Aß) plaques, the hallmark of Alzheimer's disease (AD). Herein, we report the synthesis and comprehensive characterization of 21 novel probes as well as their optical properties and binding affinities to Aß fibrils. One of these dyes, 1Ae, exhibited several improvements over FDDNP, an established biomarker for Aß- and Tau-aggregates. First, 1Ae had large Stokes shifts (138-213 nm) in various solvents, thereby reducing self-absorption. With a high quantum yield ratio (φ(dichloromethane/methanol) = 104), 1Ae also ensures minimal background emission in aqueous environments and high sensitivity. In addition, compound 1Ae exhibited low micromolar binding affinity to Aß fibrils in vitro (Kd = 1.603 µM), while increasing fluorescence emission (106-fold) compared to emission in buffer alone. Importantly, the selective binding of 1Ae to Aß1-42 fibrils was confirmed by an in cellulo assay, supported by ex vivo fluorescence microscopy of 1Ae on postmortem AD brain sections, allowing unequivocal identification of Aß plaques. The intermolecular interactions of fluorophores with Aß were elucidated by docking studies and molecular dynamics simulations. Density functional theory calculations revealed the unique photophysics of these rod-shaped fluorophores, with a twisted intramolecular charge transfer (TICT) excited state. These results provide valuable insights into the future application of such probes as potential diagnostic tools for AD in vitro and ex vivo such as determination of Aß1-42 in cerebrospinal fluid or blood.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Fluorescent Dyes , Alzheimer Disease/metabolism , Alzheimer Disease/diagnosis , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Humans , Fluorescent Dyes/chemistry , Peptide Fragments/metabolism , Peptide Fragments/cerebrospinal fluid , Brain/metabolism , Brain/pathology , Brain/diagnostic imaging , Molecular Docking Simulation , Molecular Dynamics Simulation , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Microscopy, Fluorescence/methods
10.
Brain Res Bull ; 212: 110969, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38705540

ABSTRACT

Alzheimer's disease (AD) stands as the most prevalent neurodegenerative condition worldwide, and its correlation with microglial function is notably significant. Dl-3-n-butylphthalide (NBP), derived from the seeds of Apium graveolens L. (Chinese celery), has demonstrated the capacity to diminish Aß levels in the brain tissue of Alzheimer's transgenic mice. Despite this, its connection to neuroinflammation and microglial phagocytosis, along with the specific molecular mechanism involved, remains undefined. In this study, NBP treatment exhibited a substantial improvement in learning deficits observed in AD transgenic mice (APP/PS1 transgenic mice). Furthermore, NBP treatment significantly mitigated the total cerebral Aß plaque deposition. This effect was attributed to the heightened presence of activated microglia surrounding Aß plaques and an increase in microglial phagocytosis of Aß plaques. Transcriptome sequencing analysis unveiled the potential involvement of the AGE (advanced glycation end products) -RAGE (receptor for AGE) signaling pathway in NBP's impact on APP/PS1 mice. Subsequent investigation disclosed a reduction in the secretion of AGEs, RAGE, and proinflammatory factors within the hippocampus and cortex of NBP-treated APP/PS1 mice. In summary, NBP alleviates cognitive impairment by augmenting the number of activated microglia around Aß plaques and ameliorating AGE-RAGE-mediated neuroinflammation. These findings underscore the related mechanism of the crucial neuroprotective roles of microglial phagocytosis and anti-inflammation in NBP treatment for AD, offering a potential therapeutic target for the disease.


Subject(s)
Alzheimer Disease , Benzofurans , Mice, Transgenic , Microglia , Phagocytosis , Receptor for Advanced Glycation End Products , Animals , Microglia/drug effects , Microglia/metabolism , Benzofurans/pharmacology , Mice , Phagocytosis/drug effects , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Receptor for Advanced Glycation End Products/metabolism , Signal Transduction/drug effects , Male , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Peptides/metabolism , Inflammation/metabolism , Inflammation/drug therapy , Disease Models, Animal , Presenilin-1/genetics , Presenilin-1/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Plaque, Amyloid/drug therapy , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism
11.
Sci Adv ; 10(22): eadl1123, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38809977

ABSTRACT

Immunosenescence contributes to systematic aging and plays a role in the pathogenesis of Alzheimer's disease (AD). Therefore, the objective of this study was to investigate the potential of immune rejuvenation as a therapeutic strategy for AD. To achieve this, the immune systems of aged APP/PS1 mice were rejuvenated through young bone marrow transplantation (BMT). Single-cell RNA sequencing revealed that young BMT restored the expression of aging- and AD-related genes in multiple cell types within blood immune cells. The level of circulating senescence-associated secretory phenotype proteins was decreased following young BMT. Notably, young BMT resulted in a significant reduction in cerebral Aß plaque burden, neuronal degeneration, neuroinflammation, and improvement of behavioral deficits in aged APP/PS1 mice. The ameliorated cerebral amyloidosis was associated with an enhanced Aß clearance of peripheral monocytes. In conclusion, our study provides evidence that immune system rejuvenation represents a promising therapeutic approach for AD.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Rejuvenation , Animals , Alzheimer Disease/therapy , Alzheimer Disease/metabolism , Alzheimer Disease/immunology , Mice , Mice, Transgenic , Bone Marrow Transplantation , Behavior, Animal , Amyloid beta-Peptides/metabolism , Monocytes/immunology , Monocytes/metabolism , Plaque, Amyloid/pathology , Plaque, Amyloid/metabolism , Aging/immunology , Humans
12.
Int J Mol Sci ; 25(9)2024 May 03.
Article in English | MEDLINE | ID: mdl-38732223

ABSTRACT

Alzheimer's disease (AD) is characterized by a loss of neurons in the cortex and subcortical regions. Previously, we showed that the progressive degeneration of subcortical monoaminergic (MAergic) neurons seen in human AD is recapitulated in the APPswe/PS1ΔE9 (APP/PS) transgenic mouse model. Because degeneration of cholinergic (Ach) neurons is also a prominent feature of AD, we examined the integrity of the Ach system in the APP/PS model. The overall density of Ach fibers is reduced in APP/PS1 mice at 12 and 18 months of age but not at 4 months of age. Analysis of basal forebrain Ach neurons shows no loss of Ach neurons in the APP/PS model. Thus, since MAergic systems show overt cell loss at 18 months of age, the Ach system is less vulnerable to neurodegeneration in the APP/PS1 model. We also examined whether the proximity to Aß deposition affected the degeneration of Ach and 5-HT afferents. We found that the areas closer to the edges of compact Aß deposits exhibit a more severe loss of afferents than the areas that are more distal to Aß deposits. Collectively, the results indicate that the APP/PS model recapitulates the degeneration of multiple subcortical neurotransmitter systems, including the Ach system. In addition, the results indicate that Aß deposits cause global as well as local toxicity to subcortical afferents.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Cholinergic Neurons , Disease Models, Animal , Plaque, Amyloid , Presenilin-1 , Animals , Humans , Mice , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Cholinergic Neurons/metabolism , Cholinergic Neurons/pathology , Mice, Transgenic , Plaque, Amyloid/pathology , Plaque, Amyloid/metabolism , Presenilin-1/genetics , Presenilin-1/metabolism
13.
Int J Mol Sci ; 25(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38731870

ABSTRACT

Transcranial magneto-acoustic stimulation (TMAS), which is characterized by high spatiotemporal resolution and high penetrability, is a non-invasive neuromodulation technology based on the magnetic-acoustic coupling effect. To reveal the effects of TMAS treatment on amyloid-beta (Aß) plaque and synaptic plasticity in Alzheimer's disease, we conducted a comparative analysis of TMAS and transcranial ultrasound stimulation (TUS) based on acoustic effects in 5xFAD mice and BV2 microglia cells. We found that the TMAS-TUS treatment effectively reduced amyloid plaque loads and plaque-associated neurotoxicity. Additionally, TMAS-TUS treatment ameliorated impairments in long-term memory formation and long-term potentiation. Moreover, TMAS-TUS treatment stimulated microglial proliferation and migration while enhancing the phagocytosis and clearance of Aß. In 5xFAD mice with induced microglial exhaustion, TMAS-TUS treatment-mediated Aß plaque reduction, synaptic rehabilitation improvement, and the increase in phospho-AKT levels were diminished. Overall, our study highlights that stimulation of hippocampal microglia by TMAS treatment can induce anti-cognitive impairment effects via PI3K-AKT signaling, providing hope for the development of new strategies for an adjuvant therapy for Alzheimer's disease.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Microglia , Plaque, Amyloid , Animals , Microglia/metabolism , Mice , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Transcranial Magnetic Stimulation/methods , Acoustic Stimulation , Mice, Transgenic , Disease Models, Animal , Synapses/metabolism , Hippocampus/metabolism , Male , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Neuronal Plasticity , Long-Term Potentiation , Signal Transduction
14.
Cell Rep ; 43(6): 114216, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38819990

ABSTRACT

The amyloid plaque niche is a pivotal hallmark of Alzheimer's disease (AD). Here, we employ two high-resolution spatial transcriptomics (ST) platforms, CosMx and Spatial Enhanced Resolution Omics-sequencing (Stereo-seq), to characterize the transcriptomic alterations, cellular compositions, and signaling perturbations in the amyloid plaque niche in an AD mouse model. We discover heterogeneity in the cellular composition of plaque niches, marked by an increase in microglial accumulation. We profile the transcriptomic alterations of glial cells in the vicinity of plaques and conclude that the microglial response to plaques is consistent across different brain regions, while the astrocytic response is more heterogeneous. Meanwhile, as the microglial density of plaque niches increases, astrocytes acquire a more neurotoxic phenotype and play a key role in inducing GABAergic signaling and decreasing glutamatergic signaling in hippocampal neurons. We thus show that the accumulation of microglia around hippocampal plaques disrupts astrocytic signaling, in turn inducing an imbalance in neuronal synaptic signaling.


Subject(s)
Alzheimer Disease , Astrocytes , Disease Models, Animal , Microglia , Plaque, Amyloid , Transcriptome , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Microglia/metabolism , Microglia/pathology , Astrocytes/metabolism , Astrocytes/pathology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Transcriptome/genetics , Mice , Hippocampus/metabolism , Hippocampus/pathology , Mice, Transgenic , Cell Communication , Signal Transduction , Neurons/metabolism , Neurons/pathology , Male
15.
Nat Commun ; 15(1): 3872, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38719797

ABSTRACT

The gut microbiota and microglia play critical roles in Alzheimer's disease (AD), and elevated Bacteroides is correlated with cerebrospinal fluid amyloid-ß (Aß) and tau levels in AD. We hypothesize that Bacteroides contributes to AD by modulating microglia. Here we show that administering Bacteroides fragilis to APP/PS1-21 mice increases Aß plaques in females, modulates cortical amyloid processing gene expression, and down regulates phagocytosis and protein degradation microglial gene expression. We further show that administering Bacteroides fragilis to aged wild-type male and female mice suppresses microglial uptake of Aß1-42 injected into the hippocampus. Depleting murine Bacteroidota with metronidazole decreases amyloid load in aged 5xFAD mice, and activates microglial pathways related to phagocytosis, cytokine signaling, and lysosomal degradation. Taken together, our study demonstrates that members of the Bacteroidota phylum contribute to AD pathogenesis by suppressing microglia phagocytic function, which leads to impaired Aß clearance and accumulation of amyloid plaques.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Disease Models, Animal , Mice, Transgenic , Microglia , Phagocytosis , Plaque, Amyloid , Animals , Microglia/metabolism , Microglia/drug effects , Alzheimer Disease/metabolism , Alzheimer Disease/microbiology , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Plaque, Amyloid/metabolism , Female , Mice , Male , Bacteroides fragilis/metabolism , Gastrointestinal Microbiome , Humans , Mice, Inbred C57BL , Hippocampus/metabolism , Hippocampus/pathology
16.
ACS Chem Neurosci ; 15(10): 2058-2069, 2024 05 15.
Article in English | MEDLINE | ID: mdl-38652895

ABSTRACT

Amyloid plaques composed of fibrils of misfolded Aß peptides are pathological hallmarks of Alzheimer's disease (AD). Aß fibrils are polymorphic in their tertiary and quaternary molecular structures. This structural polymorphism may carry different pathologic potencies and can putatively contribute to clinical phenotypes of AD. Therefore, mapping of structural polymorphism of Aß fibrils and structural evolution over time is valuable to understanding disease mechanisms. Here, we investigated how Aß fibril structures in situ differ in Aß plaque of different mouse models expressing familial mutations in the AßPP gene. We imaged frozen brains with a combination of conformation-sensitive luminescent conjugated oligothiophene (LCO) ligands and Aß-specific antibodies. LCO fluorescence mapping revealed that mouse models APP23, APPPS1, and AppNL-F have different fibril structures within Aß-amyloid plaques depending on the AßPP-processing genotype. Co-staining with Aß-specific antibodies showed that individual plaques from APP23 mice expressing AßPP Swedish mutation have two distinct fibril polymorph regions of core and corona. The plaque core is predominantly composed of compact Aß40 fibrils, and the corona region is dominated by diffusely packed Aß40 fibrils. Conversely, the AßPP knock-in mouse AppNL-F, expressing the AßPP Iberian mutation along with Swedish mutation has tiny, cored plaques consisting mainly of compact Aß42 fibrils, vastly different from APP23 even at elevated age up to 21 months. Age-dependent polymorph rearrangement of plaque cores observed for APP23 and APPPS1 mice >12 months, appears strongly promoted by Aß40 and was hence minuscule in AppNL-F. These structural studies of amyloid plaques in situ can map disease-relevant fibril polymorph distributions to guide the design of diagnostic and therapeutic molecules.


Subject(s)
Amyloid beta-Peptides , Amyloid beta-Protein Precursor , Plaque, Amyloid , Animals , Humans , Mice , Aging/metabolism , Aging/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Brain/pathology , Disease Models, Animal , Mice, Transgenic , Mutation , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Protein Conformation
17.
Brain Behav Immun ; 119: 286-300, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38608739

ABSTRACT

Alzheimer's disease is a progressive neurological disorder causing memory loss and cognitive decline. The underlying causes of cognitive deterioration and neurodegeneration remain unclear, leading to a lack of effective strategies to prevent dementia. Recent evidence highlights the role of neuroinflammation, particularly involving microglia, in Alzheimer's disease onset and progression. Characterizing the initial phase of Alzheimer's disease can lead to the discovery of new biomarkers and therapeutic targets, facilitating timely interventions for effective treatments. We used the AppNL-G-F knock-in mouse model, which resembles the amyloid pathology and neuroinflammatory characteristics of Alzheimer's disease, to investigate the transition from a pre-plaque to an early plaque stage with a combined functional and molecular approach. Our experiments show a progressive decrease in the power of cognition-relevant hippocampal gamma oscillations during the early stage of amyloid pathology, together with a modification of fast-spiking interneuron intrinsic properties and postsynaptic input. Consistently, transcriptomic analyses revealed that these effects are accompanied by changes in synaptic function-associated pathways. Concurrently, homeostasis- and inflammatory-related microglia signature genes were downregulated. Moreover, we found a decrease in Iba1-positive microglia in the hippocampus that correlates with plaque aggregation and neuronal dysfunction. Collectively, these findings support the hypothesis that microglia play a protective role during the early stages of amyloid pathology by preventing plaque aggregation, supporting neuronal homeostasis, and overall preserving the oscillatory network's functionality. These results suggest that the early alteration of microglia dynamics could be a pivotal event in the progression of Alzheimer's disease, potentially triggering plaque deposition, impairment of fast-spiking interneurons, and the breakdown of the oscillatory circuitry in the hippocampus.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Disease Progression , Hippocampus , Mice, Transgenic , Microglia , Plaque, Amyloid , Animals , Microglia/metabolism , Microglia/pathology , Hippocampus/metabolism , Hippocampus/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Mice , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Amyloid beta-Peptides/metabolism , Male , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Interneurons/metabolism , Interneurons/pathology
18.
J Neuroimmunol ; 390: 578342, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38640827

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease characterized by cognitive decline that severely affects patients and their families. Genetic and environmental risk factors, such as viral infections, synergize to accelerate the aging-associated neurodegeneration. Genetic risk factors for late-onset AD (LOAD), which accounts for most AD cases, are predominantly implicated in microglial and immune cell functions. As such, microglia play a major role in formation of amyloid beta (Aß) plaques, the major pathological hallmark of AD. This review aims to provide an overview of the current knowledge regarding the role of microglia in Aß plaque formation, as well as their impact on morphological and functional diversity of Aß plaques. Based on this discussion, we seek to identify challenges and opportunities in this field with potential therapeutic implications.


Subject(s)
Alzheimer Disease , Microglia , Plaque, Amyloid , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Humans , Plaque, Amyloid/pathology , Plaque, Amyloid/metabolism , Microglia/metabolism , Microglia/pathology , Animals , Amyloid beta-Peptides/metabolism
19.
Alzheimers Dement ; 20(5): 3551-3566, 2024 May.
Article in English | MEDLINE | ID: mdl-38624088

ABSTRACT

INTRODUCTION: Ozone (O3) is an air pollutant associated with Alzheimer's disease (AD) risk. The lung-brain axis is implicated in O3-associated glial and amyloid pathobiology; however, the role of disease-associated astrocytes (DAAs) in this process remains unknown. METHODS: The O3-induced astrocyte phenotype was characterized in 5xFAD mice by spatial transcriptomics and proteomics. Hmgb1fl/fl LysM-Cre+ mice were used to assess the role of peripheral myeloid cell high mobility group box 1 (HMGB1). RESULTS: O3 increased astrocyte and plaque numbers, impeded the astrocyte proteomic response to plaque deposition, augmented the DAA transcriptional fingerprint, increased astrocyte-microglia contact, and reduced bronchoalveolar lavage immune cell HMGB1 expression in 5xFAD mice. O3-exposed Hmgb1fl/fl LysM-Cre+ mice exhibited dysregulated DAA mRNA markers. DISCUSSION: Astrocytes and peripheral myeloid cells are critical lung-brain axis interactors. HMGB1 loss in peripheral myeloid cells regulates the O3-induced DAA phenotype. These findings demonstrate a mechanism and potential intervention target for air pollution-induced AD pathobiology. HIGHLIGHTS: Astrocytes are part of the lung-brain axis, regulating how air pollution affects plaque pathology. Ozone (O3) astrocyte effects are associated with increased plaques and modified by plaque localization. O3 uniquely disrupts the astrocyte transcriptomic and proteomic disease-associated astrocyte (DAA) phenotype in plaque associated astrocytes (PAA). O3 changes the PAA cell contact with microglia and cell-cell communication gene expression. Peripheral myeloid cell high mobility group box 1 regulates O3-induced transcriptomic changes in the DAA phenotype.


Subject(s)
Alzheimer Disease , Astrocytes , HMGB1 Protein , Ozone , Animals , Astrocytes/metabolism , Astrocytes/pathology , HMGB1 Protein/metabolism , Mice , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Mice, Transgenic , Disease Models, Animal , Brain/pathology , Brain/metabolism , Plaque, Amyloid/pathology , Plaque, Amyloid/metabolism , Microglia/metabolism , Air Pollutants , Lung/pathology , Amyloid beta-Peptides/metabolism
20.
Chem Biol Interact ; 395: 111012, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38648920

ABSTRACT

Acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) are associated with amyloid-ß (Aß) plaques and exhibit altered biochemical properties in human Alzheimer's disease (AD), as well as in the transgenic 5XFAD mouse model of AD amyloidosis. In the brains of the 5XFAD mouse model devoid of BChE enzyme (5XFAD/BChE-KO), incubation of tissue sections with exogenous BChE purified from human plasma (pl-BChE) leads to its association with Aß plaques and its biochemical properties are comparable to those reported for endogenous BChE associated with plaques in both human AD and in 5XFAD mouse brain tissue. We sought to determine whether these observations in 5XFAD/BChE-KO mice also apply to human brain tissues. To do so, endogenous ChE activity in human AD brain tissue sections was quenched with 50 % aqueous acetonitrile (MeCNaq) leaving the tissue suitable for further studies. Quenched sections were then incubated with recombinant AChE (r-AChE) or pl-BChE and stained for each enzymes' activity. Exogenous r-AChE or pl-BChE became associated with Aß plaques, and when bound, had properties that were comparable to the endogenous ChE enzymes associated with plaques in AD brain tissues without acetonitrile treatment. These findings in human AD brain tissue extend previous observations in the 5XFAD/BChE-KO mouse model and demonstrate that exogenously applied r-AChE and pl-BChE have high affinity for Aß plaques in human brain tissues. This association alters the biochemical properties of these enzymes, most likely due a conformational change. If incorporation of AChE and BChE in Aß plaques facilitates AD pathogenesis, blocking this association could lead to disease-modifying approaches to AD. This work provides a method to study the mechanism of AChE and BChE interaction with Aß plaque pathology in post-mortem human brain tissue.


Subject(s)
Acetylcholinesterase , Alzheimer Disease , Brain , Butyrylcholinesterase , Plaque, Amyloid , Humans , Butyrylcholinesterase/metabolism , Butyrylcholinesterase/chemistry , Brain/metabolism , Brain/pathology , Acetylcholinesterase/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Animals , Mice , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Mice, Transgenic , Mice, Knockout , Aged , Recombinant Proteins/metabolism , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...