Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 23.000
Filtrer
1.
Neurology ; 103(3): e209665, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39008782

RÉSUMÉ

BACKGROUND AND OBJECTIVES: Amyloid pathology, vascular disease pathology, and pathologies affecting the medial temporal lobe are associated with cognitive trajectories in older adults. However, only limited evidence exists on how these pathologies influence cognition in the oldest old. We evaluated whether amyloid burden, white matter hyperintensity (WMH) volume, and hippocampal volume (HV) are associated with cognitive level and decline in the oldest old. METHODS: This was a longitudinal, observational community-based cohort study. We included participants with 18F-florbetapir PET and MRI data from the 90+ Study. Amyloid load was measured using the standardized uptake value ratio in the precuneus/posterior cingulate with eroded white matter mask as reference. WMH volume was log-transformed. All imaging measures were standardized using sample means and SDs. HV and log-WMH volume were normalized by total intracranial volume using the residual approach. Global cognitive performance was measured by the Mini-Mental State Examination (MMSE) and modified MMSE (3MS) tests, repeated every 6 months. We used linear mixed-effects models with random intercepts; random slopes; and interaction between time, time squared, and imaging variables to estimate the associations of imaging variables with cognitive level and cognitive decline. Models were adjusted for demographics, APOE genotype, and health behaviors. RESULTS: The sample included 192 participants. The mean age was 92.9 years, 125 (65.1%) were female, 71 (37.0%) achieved a degree beyond college, and the median follow-up time was 3.0 years. A higher amyloid load was associated with a lower cognitive level (ßMMSE = -0.82, 95% CI -1.17 to -0.46; ß3MS = -2.77, 95% CI -3.69 to -1.84). A 1-SD decrease in HV was associated with a 0.70-point decrease in the MMSE score (95% CI -1.14 to -0.27) and a 2.27-point decrease in the 3MS score (95% CI -3.40 to -1.14). Clear nonlinear cognitive trajectories were detected. A higher amyloid burden and smaller HV were associated with faster cognitive decline. WMH volume was not significantly associated with cognitive level or decline. DISCUSSION: Amyloid burden and hippocampal atrophy are associated with both cognitive level and cognitive decline in the oldest old. Our findings shed light on how different pathologies contributed to driving cognitive function in the oldest old.


Sujet(s)
Dysfonctionnement cognitif , Hippocampe , Imagerie par résonance magnétique , Tomographie par émission de positons , Substance blanche , Humains , Femelle , Mâle , Substance blanche/imagerie diagnostique , Substance blanche/anatomopathologie , Substance blanche/métabolisme , Hippocampe/imagerie diagnostique , Hippocampe/anatomopathologie , Hippocampe/métabolisme , Sujet âgé de 80 ans ou plus , Études longitudinales , Dysfonctionnement cognitif/imagerie diagnostique , Dysfonctionnement cognitif/anatomopathologie , Dysfonctionnement cognitif/métabolisme , Cognition/physiologie , Études de cohortes , Taille d'organe , Éthylène glycols , Dérivés de l'aniline , Peptides bêta-amyloïdes/métabolisme , Amyloïde/métabolisme
2.
Life Sci Alliance ; 7(10)2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39009412

RÉSUMÉ

Treatments for Alzheimer's disease have primarily focused on removing brain amyloid plaques to improve cognitive outcomes in patients. We developed small compounds, known as BK40143 and BK40197, and we hypothesize that these drugs alleviate microglial-mediated neuroinflammation and induce autophagic clearance of neurotoxic proteins to improve behavior in models of neurodegeneration. Specificity binding assays of BK40143 and BK40197 showed primary binding to c-KIT/Platelet Derived Growth Factor Receptors (PDGFR)α/ß, whereas BK40197 also differentially binds to FYVE finger-containing phosphoinositide kinase (PIKFYVE). Both compounds penetrate the CNS, and treatment with these drugs inhibited the maturation of peripheral mast cells in transgenic mice, correlating with cognitive improvements on measures of memory and anxiety. In the brain, microglial activation was profoundly attenuated and amyloid-beta and tau were reduced via autophagy. Multi-kinase inhibition, including c-KIT, exerts multifunctional effects to reduce neurodegenerative pathology via autophagy and microglial activity and may represent a potential therapeutic option for neurodegeneration.


Sujet(s)
Maladie d'Alzheimer , Modèles animaux de maladie humaine , Souris transgéniques , Microglie , Protéines proto-oncogènes c-kit , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Animaux , Souris , Protéines proto-oncogènes c-kit/métabolisme , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Autophagie/effets des médicaments et des substances chimiques , Humains , Peptides bêta-amyloïdes/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Encéphale/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Encéphale/anatomopathologie , Mastocytes/effets des médicaments et des substances chimiques , Mastocytes/métabolisme , Protéines tau/métabolisme , Plaque amyloïde/métabolisme , Plaque amyloïde/traitement médicamenteux , Comportement animal/effets des médicaments et des substances chimiques , Mâle
3.
J Am Chem Soc ; 146(28): 19077-19087, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38973199

RÉSUMÉ

Deposition of amyloid plaques in the brains of Alzheimer's disease (AD) patients is a hallmark of the disease. AD plaques consist primarily of the beta-amyloid (Aß) peptide but can contain other factors such as lipids, proteoglycans, and chaperones. So far, it is unclear how the cellular environment modulates fibril polymorphism and how differences in fibril structure affect cell viability. The small heat-shock protein (sHSP) alpha-B-Crystallin (αBC) is abundant in brains of AD patients, and colocalizes with Aß amyloid plaques. Using solid-state NMR spectroscopy, we show that the Aß40 fibril seed structure is not replicated in the presence of the sHSP. αBC prevents the generation of a compact fibril structure and leads to the formation of a new polymorph with a dynamic N-terminus. We find that the N-terminal fuzzy coat and the stability of the C-terminal residues in the Aß40 fibril core affect the chemical and thermodynamic stability of the fibrils and influence their seeding capacity. We believe that our results yield a better understanding of how sHSP, such as αBC, that are part of the cellular environment, can affect fibril structures related to cell degeneration in amyloid diseases.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Chaîne B de la cristalline alpha , Peptides bêta-amyloïdes/composition chimique , Peptides bêta-amyloïdes/métabolisme , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Humains , Chaîne B de la cristalline alpha/composition chimique , Chaîne B de la cristalline alpha/métabolisme , Chaîne B de la cristalline alpha/génétique , Fragments peptidiques/composition chimique , Fragments peptidiques/métabolisme , Fragments peptidiques/génétique , Amyloïde/composition chimique , Amyloïde/métabolisme
4.
ACS Chem Neurosci ; 15(14): 2545-2564, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38979773

RÉSUMÉ

Alzheimer's disease (AD) is a progressive multifaceted neurodegenerative disease and remains a formidable global health challenge. The current medication for AD gives symptomatic relief and, thus, urges us to look for alternative disease-modifying therapies based on a multitarget directed approach. Looking at the remarkable progress made in peptide drug development in the last decade and the benefits associated with peptides, they offer valuable chemotypes [multitarget directed ligands (MTDLs)] as AD therapeutics. This review recapitulates the current developments made in harnessing peptides as MTDLs in combating AD by targeting multiple key pathways involved in the disease's progression. The peptides hold immense potential and represent a convincing avenue in the pursuit of novel AD therapeutics. While hurdles remain, ongoing research offers hope that peptides may eventually provide a multifaceted approach to combat AD.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Stress oxydatif , Protéines tau , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Humains , Stress oxydatif/effets des médicaments et des substances chimiques , Peptides bêta-amyloïdes/métabolisme , Protéines tau/métabolisme , Peptides/pharmacologie , Animaux , Cholinesterases/métabolisme , Agrégats de protéines/effets des médicaments et des substances chimiques , Agrégats de protéines/physiologie , Agrégation pathologique de protéines/traitement médicamenteux , Agrégation pathologique de protéines/métabolisme
5.
ACS Chem Neurosci ; 15(14): 2586-2599, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38979921

RÉSUMÉ

Aß oligomers are being investigated as cytotoxic agents in Alzheimer's disease (AD). Because of their transient nature and conformational heterogeneity, the relationship between the structure and activity of these oligomers is still poorly understood. Hence, methods for stabilizing Aß oligomeric species relevant to AD are needed to uncover the structural determinants of their cytotoxicity. Here, we build on the observation that metal ions and metabolites have been shown to interact with Aß, influencing its aggregation and stabilizing its oligomeric species. We thus developed a method that uses zinc ions, Zn(II), to stabilize oligomers produced by the 42-residue form of Aß (Aß42), which is dysregulated in AD. These Aß42-Zn(II) oligomers are small in size, spanning the 10-30 nm range, stable at physiological temperature, and with a broad toxic profile in human neuroblastoma cells. These oligomers offer a tool to study the mechanisms of toxicity of Aß oligomers in cellular and animal AD models.


Sujet(s)
Peptides bêta-amyloïdes , Fragments peptidiques , Zinc , Peptides bêta-amyloïdes/composition chimique , Peptides bêta-amyloïdes/métabolisme , Humains , Zinc/composition chimique , Fragments peptidiques/composition chimique , Fragments peptidiques/métabolisme , Lignée cellulaire tumorale , Maladie d'Alzheimer/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques
6.
Commun Biol ; 7(1): 861, 2024 Jul 14.
Article de Anglais | MEDLINE | ID: mdl-39004677

RÉSUMÉ

Oxidative stress is a key contributor to AD pathology. However, the earliest role of pre-plaque neuronal oxidative stress, remains elusive. Using laser microdissected hippocampal neurons extracted from McGill-R-Thy1-APP transgenic rats we found that intraneuronal amyloid beta (iAß)-burdened neurons had increased expression of genes related to oxidative stress and DNA damage responses including Ercc2, Fancc, Sod2, Gsr, and Idh1. DNA damage was further evidenced by increased neuronal levels of XPD (Ercc2) and γH2AX foci, indicative of DNA double stranded breaks (DSBs), and by increased expression of Ercc6, Rad51, and Fen1, and decreased Sirt6 in hippocampal homogenates. We also found increased expression of synaptic plasticity genes (Grin2b (NR2B), CamkIIα, Bdnf, c-fos, and Homer1A) and increased protein levels of TOP2ß. Our findings indicate that early accumulation of iAß, prior to Aß plaques, is accompanied by incipient oxidative stress and DSBs that may arise directly from oxidative stress or from maladaptive synaptic plasticity.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Altération de l'ADN , Modèles animaux de maladie humaine , Hippocampe , Neurones , Stress oxydatif , Rats transgéniques , Animaux , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/anatomopathologie , Hippocampe/métabolisme , Hippocampe/anatomopathologie , Neurones/métabolisme , Peptides bêta-amyloïdes/métabolisme , Peptides bêta-amyloïdes/génétique , Rats , Mâle , Plasticité neuronale
7.
ACS Chem Neurosci ; 15(14): 2600-2611, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38957957

RÉSUMÉ

Over a century has passed since Alois Alzheimer first described Alzheimer's disease (AD), and since then, researchers have made significant strides in understanding its pathology. One key feature of AD is the presence of amyloid-ß (Aß) peptides, which form amyloid plaques, and therefore, it is a primary target for treatment studies. Naturally occurring peptides have garnered attention for their potential pharmacological benefits, particularly in the central nervous system. In this study, nine peptide derivatives of Crotamine, a polypeptide from Crotalus durissus terrificus Rattlesnake venom, as well as one d-enantiomer, were evaluated for their ability to modulate Aß42 aggregation through various assays such as ThT, QIAD, SPR, and sFIDA. All tested peptides were able to decrease Aß42 aggregation and eliminate Aß42 aggregates. Additionally, all of the peptides showed an affinity for Aß42. This study is the first to describe the potential of crotamine derivative peptides against Aß42 aggregation and to identify a promising d-peptide that could be used as an effective pharmacological tool against AD in the future.


Sujet(s)
Peptides bêta-amyloïdes , Venins de crotalidé , Fragments peptidiques , Peptides bêta-amyloïdes/métabolisme , Humains , Animaux , Agrégats de protéines/effets des médicaments et des substances chimiques , Venins de serpent/composition chimique , Peptides/pharmacologie , Peptides/composition chimique , Crotalus
8.
Acta Neuropathol Commun ; 12(1): 113, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38992700

RÉSUMÉ

BACKGROUND: Despite extensive studies on the neurobiological correlates of traumatic brain injury (TBI), little is known about its molecular determinants on long-term consequences, such as dementia and Alzheimer's disease (AD). METHODS: Here, we carried out behavioural studies and an extensive biomolecular analysis, including inflammatory cytokines, gene expression and the combination of LC-HRMS and MALDI-MS Imaging to elucidate the targeted metabolomics and lipidomics spatiotemporal alterations of brains from wild-type and APP-SWE mice, a genetic model of AD, at the presymptomatic stage, subjected to mild TBI. RESULTS: We found that brain injury does not affect cognitive performance in APP-SWE mice. However, we detected an increase of key hallmarks of AD, including Aß1-42 levels and BACE1 expression, in the cortices of traumatized transgenic mice. Moreover, significant changes in the expanded endocannabinoid (eCB) system, or endocannabinoidome (eCBome), occurred, including increased levels of the endocannabinoid 2-AG in APP-SWE mice in both the cortex and hippocampus, and N-acylserotonins, detected for the first time in the brain. The gene expression of enzymes for the biosynthesis and inactivation of eCBs and eCB-like mediators, and some of their main molecular targets, also underwent significant changes. We also identified the formation of heteromers between cannabinoid 1 (CB1) and serotonergic 2A (5HT2A) receptors, whose levels increased in the cortex of APP-SWE mTBI mice, possibly contributing to the exacerbated pathophysiology of AD induced by the trauma. CONCLUSIONS: Mild TBI induces biochemical changes in AD genetically predisposed mice and the eCBome may play a role in the pathogenetic link between brain injury and neurodegenerative disorders also by interacting with the serotonergic system.


Sujet(s)
Maladie d'Alzheimer , Dysfonctionnement cognitif , Endocannabinoïdes , Souris transgéniques , Animaux , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/génétique , Souris , Endocannabinoïdes/métabolisme , Dysfonctionnement cognitif/métabolisme , Sérotonine/métabolisme , Marqueurs biologiques/métabolisme , Mâle , Commotion de l'encéphale/métabolisme , Modèles animaux de maladie humaine , Souris de lignée C57BL , Encéphale/métabolisme , Précurseur de la protéine bêta-amyloïde/génétique , Précurseur de la protéine bêta-amyloïde/métabolisme , Symptômes prodromiques , Peptides bêta-amyloïdes/métabolisme
9.
CNS Neurosci Ther ; 30(7): e14823, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38992870

RÉSUMÉ

BACKGROUND: Caloric restriction (CR) might be effective for alleviating/preventing Alzheimer's disease (AD), but the biological mechanisms remain unclear. In the current study, we explored whether CR caused an alteration of gut microbiome and resulted in the attenuation of cognitive impairment of AD animal model. METHODS: Thirty-week-old male APP/PS1 transgenic mice were used as AD models (AD mouse). CR was achieved by 30% reduction of daily free feeding (ad libitum, AL) amount. The mice were fed with CR protocol or AL protocol for six consecutive weeks. RESULTS: We found that with CR treatment, AD mice showed improved ability of learning and spatial memory, and lower levels of Aß40, Aß42, IL-1ß, TNF-α, and ROS in the brain. By sequencing 16S rDNA, we found that CR treatment resulted in significant diversity in composition and abundance of gut flora. At the phylum level, Deferribacteres (0.04%), Patescibacteria (0.14%), Tenericutes (0.03%), and Verrucomicrobia (0.5%) were significantly decreased in CR-treated AD mice; at the genus level, Dubosiella (10.04%), Faecalibaculum (0.04%), and Coriobacteriaceae UCG-002 (0.01%) were significantly increased in CR-treated AD mice by comparing with AL diet. CONCLUSIONS: Our results demonstrate that the attenuation of AD following CR treatment in APP/PS1 mice may result from alterations in the gut microbiome. Thus, gut flora could be a new target for AD prevention and therapy.


Sujet(s)
Maladie d'Alzheimer , Précurseur de la protéine bêta-amyloïde , Restriction calorique , Microbiome gastro-intestinal , Souris transgéniques , Animaux , Microbiome gastro-intestinal/physiologie , Restriction calorique/méthodes , Maladie d'Alzheimer/microbiologie , Maladie d'Alzheimer/diétothérapie , Maladie d'Alzheimer/prévention et contrôle , Mâle , Souris , Précurseur de la protéine bêta-amyloïde/génétique , Préséniline-1/génétique , Peptides bêta-amyloïdes/métabolisme , Modèles animaux de maladie humaine , Apprentissage du labyrinthe/physiologie , Encéphale/métabolisme , Souris de lignée C57BL
10.
Cells ; 13(13)2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38995015

RÉSUMÉ

The emergence of sustained neuropsychiatric symptoms (NPS) among non-demented individuals in later life, defined as mild behavioral impairment (MBI), is linked to a higher risk of cognitive decline. However, the underlying pathophysiological mechanisms remain largely unexplored. A growing body of evidence has shown that MBI is associated with alterations in structural and functional neuroimaging studies, higher genetic predisposition to clinical diagnosis of Alzheimer's disease (AD), as well as amyloid and tau pathology assessed in the blood, cerebrospinal fluid, positron-emission tomography (PET) imaging and neuropathological examination. These findings shed more light on the MBI-related potential neurobiological mechanisms, paving the way for the development of targeted pharmacological approaches. In this review, we aim to discuss the available clinical evidence on the role of amyloid and tau pathology in MBI and the potential underlying pathophysiological mechanisms. Dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, disruption of neurotrophic factors, such as the brain-derived neurotrophic factor (BDNF), abnormal neuroinflammatory responses including the kynurenine pathway, dysregulation of transforming growth factor beta (TGF-ß1), epigenetic alterations including micro-RNA (miR)-451a and miR-455-3p, synaptic dysfunction, imbalance in neurotransmitters including acetylcholine, dopamine, serotonin, gamma-aminobutyric acid (GABA) and norepinephrine, as well as altered locus coeruleus (LC) integrity are some of the potential mechanisms connecting MBI with amyloid and tau pathology. The elucidation of the underlying neurobiology of MBI would facilitate the design and efficacy of relative clinical trials, especially towards amyloid- or tau-related pathways. In addition, we provide insights for future research into our deeper understanding of its underlying pathophysiology of MBI, and discuss relative therapeutic implications.


Sujet(s)
Protéines tau , Humains , Protéines tau/métabolisme , Dysfonctionnement cognitif/métabolisme , Dysfonctionnement cognitif/anatomopathologie , Dysfonctionnement cognitif/physiopathologie , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Animaux , Amyloïde/métabolisme , Peptides bêta-amyloïdes/métabolisme
11.
Int J Mol Sci ; 25(13)2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-39000427

RÉSUMÉ

The amyloid-beta peptide (Aß) is the neurotoxic component in senile plaques of Alzheimer's disease (AD) brains. Previously we have reported that Aß toxicity is mediated by the induction of sonic hedgehog (SHH) to trigger cell cycle re-entry (CCR) and apoptosis in post-mitotic neurons. Basella alba is a vegetable whose polysaccharides carry immunomodulatory and anti-cancer actions, but their protective effects against neurodegeneration have never been reported. Herein, we tested whether polysaccharides derived from Basella alba (PPV-6) may inhibit Aß toxicity and explored its underlying mechanisms. In differentiated rat cortical neurons, Aß25-35 reduced cell viability, damaged neuronal structure, and compromised mitochondrial bioenergetic functions, all of which were recovered by PPV-6. Immunocytochemistry and western blotting revealed that Aß25-35-mediated induction of cell cycle markers including cyclin D1, proliferating cell nuclear antigen (PCNA), and histone H3 phosphorylated at Ser-10 (p-Histone H3) in differentiated neurons was all suppressed by PPV-6, along with mitigation of caspase-3 cleavage. Further studies revealed that PPV-6 inhibited Aß25-35 induction of SHH; indeed, PPV-6 was capable of suppressing neuronal CCR and apoptosis triggered by the exogenous N-terminal fragment of sonic hedgehog (SHH-N). Our findings demonstrated that, in the fully differentiated neurons, PPV-6 exerts protective actions against Aß neurotoxicity via the downregulation of SHH to suppress neuronal CCR and apoptosis.


Sujet(s)
Peptides bêta-amyloïdes , Apoptose , Cycle cellulaire , Protéines Hedgehog , Neurones , Polyosides , Peptides bêta-amyloïdes/métabolisme , Peptides bêta-amyloïdes/toxicité , Protéines Hedgehog/métabolisme , Animaux , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Rats , Polyosides/pharmacologie , Polyosides/composition chimique , Cycle cellulaire/effets des médicaments et des substances chimiques , Fragments peptidiques , Survie cellulaire/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie
12.
Int J Mol Sci ; 25(13)2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-39000459

RÉSUMÉ

Accumulation of hyper-phosphorylated tau and amyloid beta (Aß) are key pathological hallmarks of Alzheimer's disease (AD). Increasing evidence indicates that in the early pre-clinical stages of AD, phosphorylation and build-up of tau drives impairments in hippocampal excitatory synaptic function, which ultimately leads to cognitive deficits. Consequently, limiting tau-related synaptic abnormalities may have beneficial effects in AD. There is now significant evidence that the hippocampus is an important brain target for the endocrine hormone leptin and that leptin has pro-cognitive properties, as activation of synaptic leptin receptors markedly influences higher cognitive processes including learning and memory. Clinical studies have identified a link between the circulating leptin levels and the risk of AD, such that AD risk is elevated when leptin levels fall outwith the physiological range. This has fuelled interest in targeting the leptin system therapeutically. Accumulating evidence supports this possibility, as numerous studies have shown that leptin has protective effects in a variety of models of AD. Recent findings have demonstrated that leptin has beneficial effects in the preclinical stages of AD, as leptin prevents the early synaptic impairments driven by tau protein and amyloid ß. Here we review recent findings that implicate the leptin system as a potential novel therapeutic target in AD.


Sujet(s)
Maladie d'Alzheimer , Leptine , Synapses , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/traitement médicamenteux , Humains , Leptine/métabolisme , Animaux , Synapses/métabolisme , Peptides bêta-amyloïdes/métabolisme , Protéines tau/métabolisme , Récepteurs à la leptine/métabolisme , Hippocampe/métabolisme
13.
Int J Mol Sci ; 25(13)2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-39000499

RÉSUMÉ

General anesthetics may accelerate the neuropathological changes related to Alzheimer's disease (AD), of which amyloid beta (Aß)-induced toxicity is one of the main causes. However, the interaction of general anesthetics with different Aß-isoforms remains unclear. In this study, we investigated the effects of sevoflurane (0.4 and 1.2 maximal alveolar concentration (MAC)) on four Aß species-induced changes on dendritic spine density (DSD) in hippocampal brain slices of Thy1-eGFP mice and multiple epidermal growth factor-like domains 10 (MEGF10)-related astrocyte-mediated synaptic engulfment in hippocampal brain slices of C57BL/6 mice. We found that both sevoflurane and Aß downregulated CA1-dendritic spines. Moreover, compared with either sevoflurane or Aß alone, pre-treatment with Aß isoforms followed by sevoflurane application in general further enhanced spine loss. This enhancement was related to MEGF10-related astrocyte-dependent synaptic engulfment, only in AßpE3 + 1.2 MAC sevoflurane and 3NTyrAß + 1.2 MAC sevoflurane condition. In addition, removal of sevoflurane alleviated spine loss in Aß + sevoflurane. In summary, these results suggest that both synapses and astrocytes are sensitive targets for sevoflurane; in the presence of 3NTyrAß, 1.2 MAC sevoflurane alleviated astrocyte-mediated synaptic engulfment and exerted a lasting effect on dendritic spine remodeling.


Sujet(s)
Peptides bêta-amyloïdes , Astrocytes , Région CA1 de l'hippocampe , Épines dendritiques , Souris de lignée C57BL , Sévoflurane , Synapses , Sévoflurane/pharmacologie , Animaux , Épines dendritiques/métabolisme , Épines dendritiques/effets des médicaments et des substances chimiques , Peptides bêta-amyloïdes/métabolisme , Astrocytes/métabolisme , Astrocytes/effets des médicaments et des substances chimiques , Synapses/effets des médicaments et des substances chimiques , Synapses/métabolisme , Souris , Région CA1 de l'hippocampe/métabolisme , Région CA1 de l'hippocampe/effets des médicaments et des substances chimiques , Région CA1 de l'hippocampe/cytologie , Mâle , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Anesthésiques par inhalation/pharmacologie
14.
Zool Res ; 45(4): 845-856, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39004862

RÉSUMÉ

SIL1, an endoplasmic reticulum (ER)-resident protein, is reported to play a protective role in Alzheimer's disease (AD). However, the effect of SIL1 on amyloid precursor protein (APP) processing remains unclear. In this study, the role of SIL1 in APP processing was explored both in vitro and in vivo. In the in vitro experiment, SIL1 was either overexpressed or knocked down in cells stably expressing the human Swedish mutant APP695. In the in vivo experiment, AAV-SIL1-EGFP or AAV-EGFP was microinjected into APP23/PS45 mice and their wild-type littermates. Western blotting (WB), immunohistochemistry, RNA sequencing (RNA-seq), and behavioral experiments were performed to evaluate the relevant parameters. Results indicated that SIL1 expression decreased in APP23/PS45 mice. Overexpression of SIL1 significantly decreased the protein levels of APP, presenilin-1 (PS1), and C-terminal fragments (CTFs) of APP in vivo and in vitro. Conversely, knockdown of SIL1 increased the protein levels of APP, ß-site APP cleavage enzyme 1 (BACE1), PS1, and CTFs, as well as APP mRNA expression in 2EB2 cells. Furthermore, SIL1 overexpression reduced the number of senile plaques in APP23/PS45 mice. Importantly, Y-maze and Morris Water maze tests demonstrated that SIL1 overexpression improved cognitive impairment in APP23/PS45 mice. These findings indicate that SIL1 improves cognitive impairment in APP23/PS45 mice by inhibiting APP amyloidogenic processing and suggest that SIL1 is a potential therapeutic target for AD by modulating APP processing.


Sujet(s)
Précurseur de la protéine bêta-amyloïde , Dysfonctionnement cognitif , Souris transgéniques , Animaux , Précurseur de la protéine bêta-amyloïde/métabolisme , Précurseur de la protéine bêta-amyloïde/génétique , Souris , Dysfonctionnement cognitif/métabolisme , Dysfonctionnement cognitif/génétique , Peptides bêta-amyloïdes/métabolisme , Préséniline-1/génétique , Préséniline-1/métabolisme , Régulation de l'expression des gènes , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/génétique , Humains
15.
Sci Rep ; 14(1): 16097, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38997334

RÉSUMÉ

The relationship between the triglyceride glucose-body mass index (TyG-BMI) index and Alzheimer's disease (AD) pathology, cognition, and brain structure remains unclear. This study aimed to investigate these associations, focusing on cerebrospinal fluid (CSF) biomarkers, cognitive measures, and brain imaging data. Eight hundred and fifty-five non-demented participants were included. Linear regression was used to explore associations between the TyG-BMI index and AD pathology, cognition, and brain structure. The association between the TyG-BMI index and AD risk was assessed using Kaplan-Meier and Cox proportional hazards models. Longitudinal relationships were assessed using linear mixed-effects models. Mediation analyses were conducted to examine AD pathology's potential mediating role between the TyG-BMI index and cognition as well as brain structure. In the linear regression analyses, higher TyG-BMI levels were associated with increased Aß42 and decreased Tau, pTau, Tau/Aß42, pTau/Aß42, and pTau/Tau. Positive correlations were observed with mini-mental state examination (MMSE), memory (MEM), executive function (EF), and the volumes of the hippocampus, entorhinal cortex, and middle temporal regions, while negative correlations were found with Alzheimer's Disease Assessment Scale (ADAS). Longitudinally, the TyG-BMI index was inversely associated with ADAS, and positively with MMSE, MEM, EF, hippocampus, entorhinal, and middle temporal. High TyG-BMI levels were correlated with lower AD risk (HR 0.996 [0.994, 0.999]). Mediation analyses revealed AD pathology mediated the association between TyG-BMI index and cognition as well as brain structure. Additionally, the TyG-BMI index could mediate cognitive changes by influencing brain structure. The TyG-BMI index is associated with AD pathology, cognition, and brain structure.


Sujet(s)
Maladie d'Alzheimer , Indice de masse corporelle , Encéphale , Cognition , Triglycéride , Humains , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/sang , Maladie d'Alzheimer/liquide cérébrospinal , Mâle , Femelle , Sujet âgé , Encéphale/anatomopathologie , Encéphale/imagerie diagnostique , Encéphale/métabolisme , Triglycéride/sang , Marqueurs biologiques/sang , Adulte d'âge moyen , Glucose/métabolisme , Peptides bêta-amyloïdes/liquide cérébrospinal , Peptides bêta-amyloïdes/métabolisme , Protéines tau/liquide cérébrospinal , Protéines tau/métabolisme
16.
Fluids Barriers CNS ; 21(1): 56, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38997764

RÉSUMÉ

BACKGROUND: The cerebrospinal fluid (CSF), primarily generated by the choroid plexus (ChP), is the major carrier of the glymphatic system. The alternations of CSF production and the ChP can be associated with the Alzheimer's disease (AD). The present work investigated the roles of the ChP in the AD based on a proposed ChP image segmentation pipeline. METHODS: A human-in-the-loop ChP image segmentation pipeline was implemented with intermediate and active learning datasets. The performance of the proposed pipeline was evaluated on manual contours by five radiologists, compared to the FreeSurfer and FastSurfer toolboxes. The ChP volume and blood flow were investigated among AD groups. The correlations between the ChP volume and AD CSF biomarkers including phosphorylated tau (p-tau), total tau (t-tau), amyloid-ß42 (Aß42), and amyloid-ß40 (Aß40) was investigated using three models (univariate, multiple variables, and stepwise regression) on two datasets with 806 and 320 subjects. RESULTS: The proposed ChP segmentation pipeline achieved superior performance with a Dice coefficient of 0.620 on the test dataset, compared to the FreeSurfer (0.342) and FastSurfer (0.371). Significantly larger volumes (p < 0.001) and higher perfusion (p = 0.032) at the ChP were found in AD compared to CN groups. Significant correlations were found between the tau and the relative ChP volume (the ChP volume and ChP/parenchyma ratio) in each patient groups and in the univariate regression analysis (p < 0.001), the multiple regression model (p < 0.05 except for the t-tau in the LMCI), and in the step-wise regression model (p < 0.021). In addition, the correlation coefficients changed from - 0.32 to - 0.21 along with the AD progression in the multiple regression model. In contrast, the Aß42 and Aß40 shows consistent and significant associations with the lateral ventricle related measures in the step-wise regression model (p < 0.027). CONCLUSIONS: The proposed pipeline provided accurate ChP segmentation which revealed the associations between the ChP and tau level in the AD. The proposed pipeline is available on GitHub ( https://github.com/princeleeee/ChP-Seg ).


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Plexus choroïde , Protéines tau , Maladie d'Alzheimer/imagerie diagnostique , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/liquide cérébrospinal , Humains , Protéines tau/liquide cérébrospinal , Protéines tau/métabolisme , Plexus choroïde/imagerie diagnostique , Plexus choroïde/métabolisme , Peptides bêta-amyloïdes/liquide cérébrospinal , Peptides bêta-amyloïdes/métabolisme , Mâle , Femelle , Sujet âgé , Apprentissage machine supervisé , Marqueurs biologiques/liquide cérébrospinal , Marqueurs biologiques/métabolisme , Imagerie par résonance magnétique/méthodes , Traitement d'image par ordinateur/méthodes , Adulte d'âge moyen , Sujet âgé de 80 ans ou plus
17.
Int J Mol Sci ; 25(13)2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-39000011

RÉSUMÉ

Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder and affects millions of individuals globally. AD is associated with cognitive decline and memory loss that worsens with aging. A statistical report using U.S. data on AD estimates that approximately 6.9 million individuals suffer from AD, a number projected to surge to 13.8 million by 2060. Thus, there is a critical imperative to pinpoint and address AD and its hallmark tau protein aggregation early to prevent and manage its debilitating effects. Amyloid-ß and tau proteins are primarily associated with the formation of plaques and neurofibril tangles in the brain. Current research efforts focus on degrading amyloid-ß and tau or inhibiting their synthesis, particularly targeting APP processing and tau hyperphosphorylation, aiming to develop effective clinical interventions. However, navigating this intricate landscape requires ongoing studies and clinical trials to develop treatments that truly make a difference. Genome-wide association studies (GWASs) across various cohorts identified 40 loci and over 300 genes associated with AD. Despite this wealth of genetic data, much remains to be understood about the functions of these genes and their role in the disease process, prompting continued investigation. By delving deeper into these genetic associations, novel targets such as kinases, proteases, cytokines, and degradation pathways, offer new directions for drug discovery and therapeutic intervention in AD. This review delves into the intricate biological pathways disrupted in AD and identifies how genetic variations within these pathways could serve as potential targets for drug discovery and treatment strategies. Through a comprehensive understanding of the molecular underpinnings of AD, researchers aim to pave the way for more effective therapies that can alleviate the burden of this devastating disease.


Sujet(s)
Maladie d'Alzheimer , Protéines tau , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/étiologie , Humains , Protéines tau/métabolisme , Peptides bêta-amyloïdes/métabolisme , Animaux , Étude d'association pangénomique , Protéolyse
18.
Int J Mol Sci ; 25(13)2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-39000060

RÉSUMÉ

Neurodegenerative diseases (NDs) represent an unsolved problem to date with an ever-increasing population incidence. Particularly, Alzheimer's disease (AD) is the most widespread ND characterized by an accumulation of amyloid aggregates of beta-amyloid (Aß) and Tau proteins that lead to neuronal death and subsequent cognitive decline. Although neuroimaging techniques are needed to diagnose AD, the investigation of biomarkers within body fluids could provide important information on neurodegeneration. Indeed, as there is no definitive solution for AD, the monitoring of these biomarkers is of strategic importance as they are useful for both diagnosing AD and assessing the progression of the neurodegenerative state. In this context, exercise is known to be an effective non-pharmacological management strategy for AD that can counteract cognitive decline and neurodegeneration. However, investigation of the concentration of fluid biomarkers in AD patients undergoing exercise protocols has led to unclear and often conflicting results, suggesting the need to clarify the role of exercise in modulating fluid biomarkers in AD. Therefore, this critical literature review aims to gather evidence on the main fluid biomarkers of AD and the modulatory effects of exercise to clarify the efficacy and usefulness of this non-pharmacological strategy in counteracting neurodegeneration in AD.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Marqueurs biologiques , Exercice physique , Protéines tau , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/thérapie , Humains , Marqueurs biologiques/métabolisme , Exercice physique/physiologie , Peptides bêta-amyloïdes/métabolisme , Protéines tau/métabolisme , Traitement par les exercices physiques/méthodes
19.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 46(3): 425-434, 2024 Jun.
Article de Chinois | MEDLINE | ID: mdl-38953267

RÉSUMÉ

Alzheimer's disease (AD) is a severe threat to human health and one of the three major causes of human death.Double-stranded RNA-dependent protein kinase (PKR) is an interferon-induced protein kinase involved in innate immunity.In the occurrence and development of AD,PKR is upregulated and continuously activated.On the one hand,the activation of PKR triggers an integrated stress response in brain cells.On the other hand,it indirectly upregulates the expression of ß-site amyloid precursor protein cleaving enzyme 1 and facilitates the accumulation of amyloid-ß protein (Aß),which could activate PKR activator to further activate PKR,thus forming a sustained accumulation cycle of Aß.In addition,PKR can promote Tau phosphorylation,thereby reducing microtubule stability in nerve cells.Inflammation in brain tissue,neurotoxicity resulted from Aß accumulation,and disruption of microtubule stability led to the progression of AD and the declines of memory and cognitive function.Therefore,PKR is a key molecule in the development and progression of AD.Effective PKR detection can aid in the diagnosis and prediction of AD progression and provide opportunities for clinical treatment.The inhibitors targeting PKR are expected to control the activity of PKR,thereby controlling the progression of AD.Therefore,PKR could be a target for the development of therapeutic drugs for AD.


Sujet(s)
Maladie d'Alzheimer , eIF-2 Kinase , Maladie d'Alzheimer/métabolisme , Humains , eIF-2 Kinase/métabolisme , Peptides bêta-amyloïdes/métabolisme , Protéines tau/métabolisme , Phosphorylation , Encéphale/métabolisme , Précurseur de la protéine bêta-amyloïde/métabolisme
20.
FASEB J ; 38(13): e23800, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38979931

RÉSUMÉ

Insulin resistance, the hallmark of type 2 diabetes mellitus (T2DM), has emerged as a pathological feature in Alzheimer's disease (AD). Given the shared role of insulin resistance in T2DM and AD, repurposing peripheral insulin sensitizers is a promising strategy to preserve neuronal insulin sensitivity and prevent AD. 1-Deoxynojirimycin (DNJ), a bioactive iminosugar, exhibited insulin-sensitizing effects in metabolic tissues and was detected in brain tissue post-oral intake. However, its impact on brain and neuronal insulin signaling has not been described. Here, we investigated the effect of DNJ treatment on insulin signaling and AD markers in insulin-resistant human SK-N-SH neuroblastoma, a cellular model of neuronal insulin resistance. Our findings show that DNJ increased the expression of insulin signaling genes and the phosphorylation status of key molecules implicated in insulin resistance (Y1146-pIRß, S473-pAKT, S9-GSK3B) while also elevating the expression of glucose transporters Glut3 and Glut4, resulting in higher glucose uptake upon insulin stimuli. DNJ appeared to mitigate the insulin resistance-driven increase in phosphorylated tau and Aß1-42 levels by promoting insulin-induced phosphorylation of GSK3B (a major tau kinase) and enhancing mRNA expression of the insulin-degrading enzyme (IDE) pivotal for insulin and Aß clearance. Overall, our study unveils probable mechanisms underlying the potential benefits of DNJ for AD, wherein DNJ attenuates tau and amyloid pathologies by reversing neuronal insulin resistance. This provides a scientific basis for expanding the use of DNJ-containing products for neuroprotective purposes and prompts further research into compounds with similar mechanisms of action.


Sujet(s)
1-Désoxynojirimycine , Maladie d'Alzheimer , Insulinorésistance , Neurones , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/anatomopathologie , Humains , 1-Désoxynojirimycine/pharmacologie , 1-Désoxynojirimycine/analogues et dérivés , Neurones/métabolisme , Neurones/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Peptides bêta-amyloïdes/métabolisme , Protéines tau/métabolisme , Transporteur de glucose de type 3/métabolisme , Transporteur de glucose de type 3/génétique , Insuline/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Transporteur de glucose de type 4/métabolisme , Transporteur de glucose de type 4/génétique , Glycogen synthase kinase 3 beta/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Marqueurs biologiques/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...