Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 3.470
1.
Nat Commun ; 15(1): 3996, 2024 May 11.
Article En | MEDLINE | ID: mdl-38734693

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.


Alzheimer Disease , Amyloid beta-Peptides , Amyloidosis , Disease Models, Animal , Proto-Oncogene Proteins , Transcriptome , Animals , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Mice , Amyloidosis/genetics , Amyloidosis/metabolism , Amyloidosis/pathology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Phenotype , Mice, Transgenic , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Plaque, Amyloid/genetics , Humans , Male , Mice, Inbred C57BL , Gene Expression Profiling , Gene Knockdown Techniques , Trans-Activators
2.
Alzheimers Res Ther ; 16(1): 101, 2024 May 06.
Article En | MEDLINE | ID: mdl-38711159

BACKGROUND: In Alzheimer's disease (AD), microglia surround extracellular plaques and mount a sustained inflammatory response, contributing to the pathogenesis of the disease. Identifying approaches to specifically target plaque-associated microglia (PAMs) without interfering in the homeostatic functions of non-plaque associated microglia would afford a powerful tool and potential therapeutic avenue. METHODS: Here, we demonstrated that a systemically administered nanomedicine, hydroxyl dendrimers (HDs), can cross the blood brain barrier and are preferentially taken up by PAMs in a mouse model of AD. As proof of principle, to demonstrate biological effects in PAM function, we treated the 5xFAD mouse model of amyloidosis for 4 weeks via systemic administration (ip, 2x weekly) of HDs conjugated to a colony stimulating factor-1 receptor (CSF1R) inhibitor (D-45113). RESULTS: Treatment resulted in significant reductions in amyloid-beta (Aß) and a stark reduction in the number of microglia and microglia-plaque association in the subiculum and somatosensory cortex, as well as a downregulation in microglial, inflammatory, and synaptic gene expression compared to vehicle treated 5xFAD mice. CONCLUSIONS: This study demonstrates that systemic administration of a dendranib may be utilized to target and modulate PAMs.


Alzheimer Disease , Dendrimers , Disease Models, Animal , Mice, Transgenic , Microglia , Plaque, Amyloid , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Microglia/drug effects , Microglia/metabolism , Plaque, Amyloid/drug therapy , Plaque, Amyloid/pathology , Mice , Amyloid beta-Peptides/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Humans
3.
Int J Mol Sci ; 25(9)2024 Apr 24.
Article En | MEDLINE | ID: mdl-38731870

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.


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
4.
Int J Mol Sci ; 25(9)2024 May 03.
Article En | MEDLINE | ID: mdl-38732223

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.


Alzheimer Disease , Amyloid beta-Protein Precursor , Cholinergic Neurons , Disease Models, Animal , Mice, Transgenic , Plaque, Amyloid , Presenilin-1 , Animals , Plaque, Amyloid/pathology , Plaque, Amyloid/metabolism , Mice , Cholinergic Neurons/metabolism , Cholinergic Neurons/pathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Presenilin-1/genetics , Presenilin-1/metabolism , Humans , Amyloid beta-Peptides/metabolism
5.
Int J Mol Sci ; 25(8)2024 Apr 13.
Article En | MEDLINE | ID: mdl-38673909

Recruitment and accumulation of reactive astrocytes around senile plaques are common pathological features of Alzheimer's disease (AD), with unclear mechanisms. Chemerin, an adipokine implicated in neuroinflammation, acts through its receptor, chemokine-like receptor 1 (CMKLR1), which also functions as a receptor for amyloid ß (Aß). The impact of the chemerin/CMKLR1 axis on astrocyte migration towards Aß plaques is unknown. Here we investigated the effect of CMKLR1 on astrocyte migration around Aß deposition in APP/PS1 mice with Cmklr1 knockout (APP/PS1-Cmklr1-/-). CMKLR1-expressed astrocytes were upregulated in the cortices and hippocampi of 9-month-old APP/PS1 mice. Chemerin mainly co-localized with neurons, and its expression was reduced in the brains of APP/PS1 mice, compared to WT mice. CMKLR1 deficiency decreased astrocyte colocalization with Aß plaques in APP/PS1-Cmklr1-/- mice, compared to APP/PS1 mice. Activation of the chemerin/CMKLR1 axis promoted the migration of primary cultured astrocytes and U251 cells, and reduced astrocyte clustering induced by Aß42. Mechanistic studies revealed that chemerin/CMKLR1 activation induced STING phosphorylation. Deletion of STING attenuated the promotion of the chemerin/CMKLR1 axis relative to astrocyte migration and abolished the inhibitory effect of chemerin on Aß42-induced astrocyte clustering. These findings suggest the involvement of the chemerin/CMKLR1/STING pathway in the regulation of astrocyte migration and recruitment to Aß plaques/Aß42.


Alzheimer Disease , Astrocytes , Chemokines , Intercellular Signaling Peptides and Proteins , Plaque, Amyloid , Receptors, Chemokine , Animals , Astrocytes/metabolism , Chemokines/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Mice , Receptors, Chemokine/metabolism , Receptors, Chemokine/genetics , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Humans , Amyloid beta-Peptides/metabolism , Mice, Knockout , Cell Movement , Signal Transduction , Mice, Transgenic , Mice, Inbred C57BL
6.
J Neuroimmunol ; 390: 578342, 2024 May 15.
Article En | MEDLINE | ID: mdl-38640827

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.


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
7.
J Theor Biol ; 587: 111823, 2024 Jun 21.
Article En | MEDLINE | ID: mdl-38608804

This paper introduces a new model to simulate the progression of senile plaques, focusing on scenarios where concentrations of amyloid beta (Aß) monomers and aggregates vary between neurons. Extracellular variations in these concentrations may arise due to limited diffusivity of Aß monomers and a high rate of Aß monomer production at lipid membranes, requiring a substantial concentration gradient for diffusion-driven transport of Aß monomers. The dimensionless formulation of the model is presented, which identifies four key dimensionless parameters governing the solutions for Aß monomer and aggregate concentrations, as well as the radius of a growing Aß plaque within the control volume. These parameters include the dimensionless diffusivity of Aß monomers, the dimensionless rate of Aß monomer production, and the dimensionless half-lives of Aß monomers and aggregates. A dimensionless parameter is then introduced to evaluate the validity of the lumped capacitance approximation. An approximate solution is derived for the scenario involving large diffusivity of Aß monomers and dysfunctional protein degradation machinery, resulting in infinitely long half-lives for Aß monomers and aggregates. In this scenario, the concentrations of Aß aggregates and the radius of the Aß plaque depend solely on a single dimensionless parameter that characterizes the rate of Aß monomer production. According to the approximate solution, the concentration of Aß aggregates is linearly dependent on the rate of monomer production, and the radius of an Aß plaque is directly proportional to the cube root of the rate of monomer production. However, when departing from the conditions of the approximate solution (e.g., finite half-lives), the concentrations of Aß monomers and aggregates, along with the plaque radius, exhibit complex dependencies on all four dimensionless parameters. For instance, under physiological half-life conditions, the plaque radius reaches a maximum value and stabilizes thereafter.


Amyloid beta-Peptides , Plaque, Amyloid , Amyloid beta-Peptides/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Models, Biological , Diffusion
8.
J Mol Neurosci ; 74(2): 49, 2024 Apr 26.
Article En | MEDLINE | ID: mdl-38668787

The pathogenesis of Alzheimer's disease (AD) is complex and involves an imbalance between production and clearance of amyloid-ß peptides (Aß), resulting in accumulation of Aß in senile plaques. Hypercholesterolemia is a major risk factor for developing AD, with cholesterol shown to accumulate in senile plaques and increase production of Aß. ABCG4 is a member of the ATP-binding cassette transporters predominantly expressed in the CNS and has been suggested to play a role in cholesterol and Aß efflux from the brain. In this study, we bred Abcg4 knockout (KO) with the APPSwe,Ind (J9) mouse model of AD to test the hypothesis that loss of Abcg4 would exacerbate the AD phenotype. Unexpectedly, no differences were observed in novel object recognition (NOR) and novel object placement (NOP) behavioral tests, or on histologic examinations of brain tissues for senile plaque numbers. Furthermore, clearance of radiolabeled Aß from the brains did not differ between Abcg4 KO and control mice. Metabolic testing by indirect calorimetry, glucose tolerance test (GTT), and insulin tolerance test (ITT) were also mostly similar between groups with only a few mild metabolic differences noted. Overall, these data suggest that the loss of ABCG4 did not exacerbate the AD phenotype.


ATP Binding Cassette Transporter, Subfamily G , Alzheimer Disease , Animals , Male , Mice , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , ATP Binding Cassette Transporter, Subfamily G/metabolism , ATP Binding Cassette Transporter, Subfamily G/genetics , Brain/metabolism , Brain/pathology , Mice, Inbred C57BL , Plaque, Amyloid/pathology
9.
ACS Chem Neurosci ; 15(10): 2058-2069, 2024 May 15.
Article En | MEDLINE | ID: mdl-38652895

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.


Amyloid beta-Peptides , Amyloid beta-Protein Precursor , Mice, Transgenic , Plaque, Amyloid , Animals , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Mice , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Disease Models, Animal , Brain/metabolism , Brain/pathology , Mutation , Aging/metabolism , Aging/pathology , Protein Conformation , Humans
10.
Chem Biol Interact ; 395: 111012, 2024 May 25.
Article En | MEDLINE | ID: mdl-38648920

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.


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
11.
Alzheimers Dement ; 20(5): 3551-3566, 2024 May.
Article En | MEDLINE | ID: mdl-38624088

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.


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
12.
Mol Brain ; 17(1): 21, 2024 Apr 30.
Article En | MEDLINE | ID: mdl-38685105

Dopamine plays important roles in cognitive function and inflammation and therefore is involved in the pathogenesis of neurodegenerative diseases, including Alzheimer's disease (AD). Drugs that increase or maintain dopamine levels in the brain could be a therapeutic strategy for AD. However, the effects of dopamine and its precursor levodopa (L-DOPA) on Aß/tau pathology in vivo and the underlying molecular mechanisms have not been studied in detail. Here, we investigated whether L-DOPA treatment alters neuroinflammation, Aß pathology, and tau phosphorylation in 5xFAD mice, a model of AD. We found that L-DOPA administration significantly reduced microgliosis and astrogliosis in 5xFAD mice. In addition, L-DOPA treatment significantly decreased Aß plaque number by upregulating NEP and ADAM17 levels in 5xFAD mice. However, L-DOPA-treated 5xFAD mice did not exhibit changes in tau hyperphosphorylation or tau kinase levels. These data suggest that L-DOPA alleviates neuroinflammatory responses and Aß pathology but not tau pathology in this mouse model of AD.


ADAM17 Protein , Alzheimer Disease , Amyloid beta-Peptides , Disease Models, Animal , Levodopa , Mice, Transgenic , Neuroinflammatory Diseases , tau Proteins , Animals , Levodopa/pharmacology , Alzheimer Disease/pathology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , ADAM17 Protein/metabolism , Amyloid beta-Peptides/metabolism , tau Proteins/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/metabolism , Phosphorylation/drug effects , Plaque, Amyloid/pathology , Plaque, Amyloid/metabolism , Mice , Brain/pathology , Brain/drug effects , Brain/metabolism
13.
PLoS One ; 19(4): e0299534, 2024.
Article En | MEDLINE | ID: mdl-38574297

Alzheimer's disease (AD) is the most common neuronal disorder that leads to the development of dementia. Until nowadays, some therapies may alleviate the symptoms, but there is no pharmacological treatment. Microdosing lithium has been used to modify the pathological characteristics of the disease, with effects in both experimental and clinical conditions. The present work aimed to analyze the effects of this treatment on spatial memory, anxiety, and molecular mechanisms related to long-term memory formation during the aging process of a mouse model of accelerated aging (SAMP-8). Female SAMP-8 showed learning and memory impairments together with disruption of memory mechanisms, neuronal loss, and increased density of senile plaques compared to their natural control strain, the senescence-accelerated mouse resistant (SAMR-1). Chronic treatment with lithium promoted memory maintenance, reduction in anxiety, and maintenance of proteins related to memory formation and neuronal density. The density of senile plaques was also reduced. An increase in the density of gamma-aminobutyric acid A (GABAA) and α7 nicotinic cholinergic receptors was also observed and related to neuroprotection and anxiety reduction. In addition, this microdose of lithium inhibited the activation of glycogen synthase kinase-3beta (GSK-3ß), the classical mechanism of lithium cell effects, which could contribute to the preservation of the memory mechanism and reduction in senile plaque formation. This work shows that lithium effects in neuroprotection along the aging process are not related to a unique cellular mechanism but produce multiple effects that slowly protect the brain along the aging process.


Alzheimer Disease , Lithium , Phenylmercury Compounds , Mice , Female , Animals , Lithium/pharmacology , Lithium/therapeutic use , Plaque, Amyloid/pathology , Glycogen Synthase Kinase 3 beta , Alzheimer Disease/pathology , Aging/metabolism , Disease Models, Animal
14.
J Alzheimers Dis ; 99(1): 121-143, 2024.
Article En | MEDLINE | ID: mdl-38640149

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.


Alzheimer Disease , Disease Models, Animal , Memory Disorders , Rats, Wistar , Vanadium Compounds , Animals , Alzheimer Disease/pathology , Alzheimer Disease/chemically induced , Male , Vanadium Compounds/pharmacology , Rats , Memory Disorders/pathology , Memory Disorders/chemically induced , Maze Learning/drug effects , Brain/pathology , Brain/drug effects , Brain/metabolism , Spatial Memory/drug effects , Neurofibrillary Tangles/pathology , Neurofibrillary Tangles/drug effects , Plaque, Amyloid/pathology , Dendritic Spines/drug effects , Dendritic Spines/pathology , Administration, Inhalation
15.
Nat Neurosci ; 27(5): 886-900, 2024 May.
Article En | MEDLINE | ID: mdl-38539015

Microglia are central players in Alzheimer's disease pathology but analyzing microglial states in human brain samples is challenging due to genetic diversity, postmortem delay and admixture of pathologies. To circumvent these issues, here we generated 138,577 single-cell expression profiles of human stem cell-derived microglia xenotransplanted in the brain of the AppNL-G-F model of amyloid pathology and wild-type controls. Xenografted human microglia adopt a disease-associated profile similar to that seen in mouse microglia, but display a more pronounced human leukocyte antigen or HLA state, likely related to antigen presentation in response to amyloid plaques. The human microglial response also involves a pro-inflammatory cytokine/chemokine cytokine response microglia or CRM response to oligomeric Aß oligomers. Genetic deletion of TREM2 or APOE as well as APOE polymorphisms and TREM2R47H expression in the transplanted microglia modulate these responses differentially. The expression of other Alzheimer's disease risk genes is differentially regulated across the distinct cell states elicited in response to amyloid pathology. Thus, we have identified multiple transcriptomic cell states adopted by human microglia in a multipronged response to Alzheimer's disease-related pathology, which should be taken into account in translational studies.


Alzheimer Disease , Amyloid beta-Peptides , Microglia , Receptors, Immunologic , Transcriptome , Humans , Microglia/metabolism , Microglia/pathology , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Animals , Amyloid beta-Peptides/metabolism , Mice , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Mice, Transgenic , Heterografts , Plaque, Amyloid/pathology , Plaque, Amyloid/metabolism , Brain/metabolism , Brain/pathology
16.
JCI Insight ; 9(6)2024 Feb 06.
Article En | MEDLINE | ID: mdl-38516884

Substantial evidence suggests a role for immunotherapy in treating Alzheimer's disease (AD). While the precise pathophysiology of AD is incompletely understood, clinical trials of antibodies targeting aggregated forms of ß amyloid (Aß) have shown that reducing amyloid plaques can mitigate cognitive decline in patients with early-stage AD. Here, we describe what we believe to be a novel approach to target and degrade amyloid plaques by genetically engineering macrophages to express an Aß-targeting chimeric antigen receptor (CAR-Ms). When injected intrahippocampally, first-generation CAR-Ms have limited persistence and fail to significantly reduce plaque load, which led us to engineer next-generation CAR-Ms that secrete M-CSF and self-maintain without exogenous cytokines. Cytokine secreting "reinforced CAR-Ms" have greater survival in the brain niche and significantly reduce plaque load locally in vivo. These findings support CAR-Ms as a platform to rationally target, resorb, and degrade pathogenic material that accumulates with age, as exemplified by targeting Aß in AD.


Alzheimer Disease , Receptors, Chimeric Antigen , Mice , Animals , Humans , Mice, Transgenic , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Alzheimer Disease/pathology , Cytokines/metabolism , Macrophages/metabolism
17.
Mol Ther ; 32(5): 1373-1386, 2024 May 01.
Article En | MEDLINE | ID: mdl-38504517

Epidemiological studies show that individuals who carry the relatively uncommon APOE ε2 allele rarely develop Alzheimer disease, and if they do, they have a later age of onset, milder clinical course, and less severe neuropathological findings than people without this allele. The contrast is especially stark when compared with the major genetic risk factor for Alzheimer disease, APOE ε4, which has an age of onset several decades earlier, a more aggressive clinical course and more severe neuropathological findings, especially in terms of the amount of amyloid deposition. Here, we demonstrate that brain exposure to APOE ε2 via a gene therapy approach, which bathes the entire cortical mantle in the gene product after transduction of the ependyma, reduces Aß plaque deposition, neurodegenerative synaptic loss, and, remarkably, reduces microglial activation in an APP/PS1 mouse model despite continued expression of human APOE ε4. This result suggests a promising protective effect of exogenous APOE ε2 and reveals a cell nonautonomous effect of the protein on microglial activation, which we show is similar to plaque-associated microglia in the brain of Alzheimer disease patients who inherit APOE ε2. These data increase the potential that an APOE ε2 therapeutic could be effective in Alzheimer disease, even in individuals born with the risky ε4 allele.


Alzheimer Disease , Apolipoprotein E2 , Disease Models, Animal , Genetic Therapy , Mice, Transgenic , Microglia , Plaque, Amyloid , Animals , Alzheimer Disease/therapy , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/etiology , Mice , Genetic Therapy/methods , Humans , Apolipoprotein E2/genetics , Apolipoprotein E2/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Microglia/metabolism , Brain/metabolism , Brain/pathology , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/therapy , Neuroinflammatory Diseases/metabolism , Amyloid beta-Peptides/metabolism , Biomarkers
18.
J Transl Med ; 22(1): 291, 2024 Mar 18.
Article En | MEDLINE | ID: mdl-38500108

BACKGROUND: Biologic TNF-α inhibitors (bTNFIs) can block cerebral TNF-α in Alzheimer's disease (AD) if these macromolecules can cross the blood-brain barrier (BBB). Thus, a model bTNFI, the extracellular domain of type II TNF-α receptor (TNFR), which can bind to and sequester TNF-α, was fused with a mouse transferrin receptor antibody (TfRMAb) to enable brain delivery via BBB TfR-mediated transcytosis. Previously, we found TfRMAb-TNFR to be protective in a mouse model of amyloidosis (APP/PS1) and tauopathy (PS19), and herein we investigated its effects in mice that combine both amyloidosis and tauopathy (3xTg-AD). METHODS: Eight-month-old female 3xTg-AD mice were injected intraperitoneally with saline (n = 11) or TfRMAb-TNFR (3 mg/kg; n = 11) three days per week for 12 weeks. Age-matched wild-type (WT) mice (n = 9) were treated similarly with saline. Brains were processed for immunostaining and high-resolution multiplex NanoString GeoMx spatial proteomics. RESULTS: We observed regional differences in proteins relevant to Aß, tau, and neuroinflammation in the hippocampus of 3xTg-AD mice compared with WT mice. From 64 target proteins studied using spatial proteomics, a comparison of the Aß-plaque bearing vs. plaque-free regions in the 3xTg-AD mice yielded 39 differentially expressed proteins (DEP) largely related to neuroinflammation (39% of DEP) and Aß and tau pathology combined (31% of DEP). Hippocampal spatial proteomics revealed that the majority of the proteins modulated by TfRMAb-TNFR in the 3xTg-AD mice were relevant to microglial function (⁓ 33%). TfRMAb-TNFR significantly reduced mature Aß plaques and increased Aß-associated microglia around larger Aß deposits in the 3xTg-AD mice. Further, TfRMAb-TNFR increased mature Aß plaque-associated microglial TREM2 in 3xTg-AD mice. CONCLUSION: Overall, despite the low visual Aß load in the 11-month-old female 3xTg-AD mice, our results highlight region-specific AD-relevant DEP in the hippocampus of these mice. Chronic TfRMAb-TNFR dosing modulated several DEP involved in AD pathology and showed a largely microglia-centric mechanism of action in the 3xTg-AD mice.


Alzheimer Disease , Amyloidosis , Biological Products , Mice , Female , Animals , Alzheimer Disease/drug therapy , Tumor Necrosis Factor-alpha/metabolism , Amyloid beta-Protein Precursor/metabolism , Neuroinflammatory Diseases , Mice, Transgenic , Brain/pathology , Hippocampus/metabolism , Hippocampus/pathology , Amyloidosis/metabolism , Amyloidosis/pathology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Antibodies/metabolism , Biological Products/metabolism , Disease Models, Animal
19.
Sci Rep ; 14(1): 6287, 2024 03 15.
Article En | MEDLINE | ID: mdl-38491154

The absence of a natural animal model is one of the main challenges in Alzheimer's disease research. Despite the challenges of using nonhuman primates in studies, these animals can bridge mouse models and humans, as nonhuman primates are phylogenetically closer to humans and can spontaneously develop AD-type pathology. The capuchin monkey, a New World primate, has recently attracted attention due to its skill in creating and using instruments. We analyzed one capuchin brain using structural 7 T MRI and performed a neuropathological evaluation of three animals. Alzheimer-type pathology was found in the two of the capuchins. Widespread ß-amyloid pathology was observed, mainly in focal deposits with variable morphology and a high density of mature plaques. Notably, plaque-associated dystrophic neurites associated with disruption of axonal transport and early cytoskeletal alteration were frequently found. Unlike in other species of New World monkeys, cerebral arterial angiopathy was not the predominant form of ß-amyloid pathology. Additionally, abnormal aggregates of hyperphosphorylated tau, resembling neurofibrillary pathology, were observed in the temporal and frontal cortex. Astrocyte hypertrophy surrounding plaques was found, suggesting a neuroinflammatory response. These findings indicate that aged capuchin monkeys can spontaneously develop Alzheimer-type pathology, indicating that they may be an advantageous animal model for research in Alzheimer's disease.


Alzheimer Disease , Cebinae , Humans , Animals , Mice , Aged , Alzheimer Disease/pathology , Cebus , Haplorhini , Amyloid beta-Peptides/metabolism , Brain/metabolism , Plaque, Amyloid/pathology , tau Proteins/metabolism
20.
Alzheimers Dement ; 20(4): 2794-2816, 2024 Apr.
Article En | MEDLINE | ID: mdl-38426371

INTRODUCTION: Alzheimer's disease (AD) is a neurodegenerative disorder with multifactorial etiology, including genetic factors that play a significant role in disease risk and resilience. However, the role of genetic diversity in preclinical AD studies has received limited attention. METHODS: We crossed five Collaborative Cross strains with 5xFAD C57BL/6J female mice to generate F1 mice with and without the 5xFAD transgene. Amyloid plaque pathology, microglial and astrocytic responses, neurofilament light chain levels, and gene expression were assessed at various ages. RESULTS: Genetic diversity significantly impacts AD-related pathology. Hybrid strains showed resistance to amyloid plaque formation and neuronal damage. Transcriptome diversity was maintained across ages and sexes, with observable strain-specific variations in AD-related phenotypes. Comparative gene expression analysis indicated correlations between mouse strains and human AD. DISCUSSION: Increasing genetic diversity promotes resilience to AD-related pathogenesis, relative to an inbred C57BL/6J background, reinforcing the importance of genetic diversity in uncovering resilience in the development of AD. HIGHLIGHTS: Genetic diversity's impact on AD in mice was explored. Diverse F1 mouse strains were used for AD study, via the Collaborative Cross. Strain-specific variations in AD pathology, glia, and transcription were found. Strains resilient to plaque formation and plasma neurofilament light chain (NfL) increases were identified. Correlations with human AD transcriptomics were observed.


Alzheimer Disease , Resilience, Psychological , Mice , Humans , Female , Animals , Alzheimer Disease/pathology , Plaque, Amyloid/pathology , Mice, Inbred C57BL , Microglia/metabolism , Genetic Variation/genetics , Disease Models, Animal , Mice, Transgenic , Amyloid beta-Peptides/metabolism
...