Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22.748
Filter
1.
Sci Rep ; 14(1): 21560, 2024 09 16.
Article in English | MEDLINE | ID: mdl-39284855

ABSTRACT

While previous research has shown the potential links between taste perception pathways and brain-related conditions, the area involving Alzheimer's disease remains incompletely understood. Taste perception involves neurotransmitter signaling, including serotonin, glutamate, and dopamine. Disruptions in these pathways are implicated in neurodegenerative diseases. The integration of olfactory and taste signals in flavor perception may impact brain health, evident in olfactory dysfunction as an early symptom in neurodegenerative conditions. Shared immune response and inflammatory pathways may contribute to the association between altered taste perception and conditions like neurodegeneration, present in Alzheimer's disease. This study consists of an exploration of expression-quantitative trait loci (eQTL), utilizing whole-blood transcriptome profiles, of 28 taste perception genes, from a combined cohort of 475 African American subjects. This comprehensive dataset was subsequently intersected with single-nucleotide polymorphisms (SNPs) identified in Genome-Wide Association Studies (GWAS) of Alzheimer's Disease (AD). Finally, the investigation delved into assessing the association between eQTLs reported in GWAS of AD and the profiles of 741 proteins from the Olink Neurological Panel. The eQTL analysis unveiled 3,547 statistically significant SNP-Gene associations, involving 412 distinct SNPs that spanned all 28 taste genes. In 17 GWAS studies encompassing various traits, a total of 14 SNPs associated with 12 genes were identified, with three SNPs consistently linked to Alzheimer's disease across four GWAS studies. All three SNPs demonstrated significant associations with the down-regulation of TAS2R41, and two of them were additionally associated with the down-regulation of TAS2R60. In the subsequent pQTL analysis, two of the SNPs linked to TAS2R41 and TAS2R60 genes (rs117771145 and rs10228407) were correlated with the upregulation of two proteins, namely EPHB6 and ADGRB3. Our investigation introduces a new perspective to the understanding of Alzheimer's disease, emphasizing the significance of bitter taste receptor genes in its pathogenesis. These discoveries set the stage for subsequent research to delve into these receptors as promising avenues for both intervention and diagnosis. Nevertheless, the translation of these genetic insights into clinical practice requires a more profound understanding of the implicated pathways and their pertinence to the disease's progression across diverse populations.


Subject(s)
Alzheimer Disease , Black or African American , Genome-Wide Association Study , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Taste Perception , Humans , Alzheimer Disease/genetics , Black or African American/genetics , Taste Perception/genetics , Female , Male , Aged , Genetic Predisposition to Disease , Middle Aged , Transcriptome
2.
Nat Commun ; 15(1): 7880, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39251599

ABSTRACT

Progressive supranuclear palsy (PSP), a rare Parkinsonian disorder, is characterized by problems with movement, balance, and cognition. PSP differs from Alzheimer's disease (AD) and other diseases, displaying abnormal microtubule-associated protein tau by both neuronal and glial cell pathologies. Genetic contributors may mediate these differences; however, the genetics of PSP remain underexplored. Here we conduct the largest genome-wide association study (GWAS) of PSP which includes 2779 cases (2595 neuropathologically-confirmed) and 5584 controls and identify six independent PSP susceptibility loci with genome-wide significant (P < 5 × 10-8) associations, including five known (MAPT, MOBP, STX6, RUNX2, SLCO1A2) and one novel locus (C4A). Integration with cell type-specific epigenomic annotations reveal an oligodendrocytic signature that might distinguish PSP from AD and Parkinson's disease in subsequent studies. Candidate PSP risk gene prioritization using expression quantitative trait loci (eQTLs) identifies oligodendrocyte-specific effects on gene expression in half of the genome-wide significant loci, and an association with C4A expression in brain tissue, which may be driven by increased C4A copy number. Finally, histological studies demonstrate tau aggregates in oligodendrocytes that colocalize with C4 (complement) deposition. Integrating GWAS with functional studies, epigenomic and eQTL analyses, we identify potential causal roles for variation in MOBP, STX6, RUNX2, SLCO1A2, and C4A in PSP pathogenesis.


Subject(s)
Genetic Predisposition to Disease , Genome-Wide Association Study , Quantitative Trait Loci , Supranuclear Palsy, Progressive , tau Proteins , Humans , Supranuclear Palsy, Progressive/genetics , Supranuclear Palsy, Progressive/pathology , Supranuclear Palsy, Progressive/metabolism , Aged , Male , Female , tau Proteins/genetics , tau Proteins/metabolism , Transcriptome , Polymorphism, Single Nucleotide , Neuroglia/metabolism , Neuroglia/pathology , Aged, 80 and over , Oligodendroglia/metabolism , Oligodendroglia/pathology , Middle Aged , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Case-Control Studies , Myelin Proteins
3.
Commun Biol ; 7(1): 1103, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39251807

ABSTRACT

Neurofilament light chain (NfL) levels in circulation have been established as a sensitive biomarker of neuro-axonal damage across a range of neurodegenerative disorders. Elucidation of the genetic architecture of blood NfL levels could provide new insights into molecular mechanisms underlying neurodegenerative disorders. In this meta-analysis of genome-wide association studies (GWAS) of blood NfL levels from eleven cohorts of European ancestry, we identify two genome-wide significant loci at 16p12 (UMOD) and 17q24 (SLC39A11). We observe association of three loci at 1q43 (FMN2), 12q14, and 12q21 with blood NfL levels in the meta-analysis of African-American ancestry. In the trans-ethnic meta-analysis, we identify three additional genome-wide significant loci at 1p32 (FGGY), 6q14 (TBX18), and 4q21. In the post-GWAS analyses, we observe the association of higher NfL polygenic risk score with increased plasma levels of total-tau, Aß-40, Aß-42, and higher incidence of Alzheimer's disease in the Rotterdam Study. Furthermore, Mendelian randomization analysis results suggest that a lower kidney function could cause higher blood NfL levels. This study uncovers multiple genetic loci of blood NfL levels, highlighting the genes related to molecular mechanism of neurodegeneration.


Subject(s)
Genome-Wide Association Study , Neurodegenerative Diseases , Neurofilament Proteins , Humans , Neurofilament Proteins/genetics , Neurofilament Proteins/blood , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/blood , Genetic Predisposition to Disease , Genetic Loci , Biomarkers/blood , Polymorphism, Single Nucleotide , Male , Female , Alzheimer Disease/genetics , Alzheimer Disease/blood
4.
J Alzheimers Dis ; 101(2): 693-704, 2024.
Article in English | MEDLINE | ID: mdl-39240638

ABSTRACT

Background: Bridging integrator 1 (BIN1) gene polymorphism has been reported to play a role in the pathological processes of Alzheimer's disease (AD). Objective: To explore the association of BIN1 loci with neuroinflammation and AD pathology. Methods: Alzheimer's Disease Neuroimaging Initiative (ADNI, N = 495) was the discovery cohort, and Chinese Alzheimer's Biomarker and LifestylE (CABLE, N = 619) study was used to replicate the results. Two BIN1 gene polymorphism (rs7561528 and rs744373) were included in the analysis. Multiple linear regression model and causal mediation analysis conducted through 10,000 bootstrapped iterations were used to examine the BIN1 loci relationship with cerebrospinal fluid (CSF) AD biomarkers and alternative biomarker of microglial activation microglia-soluble triggering receptor expressed on myeloid cells 2 (sTREM2). Results: In ADNI database, we found a significant association between BIN1 loci (rs7561528 and rs744373) and levels of CSF phosphorylated-tau (P-tau) (pc = 0.017; 0.010, respectively) and total-tau (T-tau) (pc = 0.011; 0.013, respectively). The BIN1 loci were also correlated with CSF sTREM2 levels (pc = 0.010; 0.008, respectively). Mediation analysis demonstrated that CSF sTREM2 partially mediated the association of BIN1 loci with P-tau (Proportion of rs7561528 : 20.8%; Proportion of rs744373 : 24.8%) and T-tau (Proportion of rs7561528 : 36.5%; Proportion of rs744373 : 43.9%). The analysis in CABLE study replicated the mediation role of rs7561528. Conclusions: This study demonstrated the correlation between BIN1 loci and CSF AD biomarkers as well as microglia biomarkers. Additionally, the link between BIN1 loci and tau pathology was partially mediated by CSF sTREM2.


Subject(s)
Adaptor Proteins, Signal Transducing , Alzheimer Disease , Biomarkers , Membrane Glycoproteins , Receptors, Immunologic , Tumor Suppressor Proteins , tau Proteins , Humans , Alzheimer Disease/genetics , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/pathology , tau Proteins/cerebrospinal fluid , tau Proteins/genetics , Adaptor Proteins, Signal Transducing/genetics , Female , Tumor Suppressor Proteins/genetics , Male , Aged , Receptors, Immunologic/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Polymorphism, Single Nucleotide/genetics , Aged, 80 and over , Nuclear Proteins
5.
PLoS One ; 19(9): e0289435, 2024.
Article in English | MEDLINE | ID: mdl-39240956

ABSTRACT

Mutations in the presenilin (PS) genes are a predominant cause of familial Alzheimer's disease (fAD). An ortholog of PS in the genetic model organism Caenorhabditis elegans (C. elegans) is sel-12. Mutations in the presenilin genes are commonly thought to lead to fAD by upregulating the expression of amyloid beta (Aß), however this hypothesis has been challenged by recent evidence. As C. elegans lack amyloid beta (Aß), the goal of this work was to examine Aß-independent effects of mutations in sel-12 and PS1/PS2 on behaviour and sensory neuron morphology across the lifespan in a C. elegans model. Olfactory chemotaxis experiments were conducted on sel-12(ok2078) loss-of-function mutant worms. Adult sel-12 mutant worms showed significantly lower levels of chemotaxis to odorants compared to wild-type worms throughout their lifespan, and this deficit increased with age. The chemotaxis phenotype in sel-12 mutant worms is rescued by transgenic over-expression of human wild-type PS1, but not the classic fAD-associated variant PS1C410Y, when expression was driven by either the endogenous sel-12 promoter (Psel-12), a pan-neuronal promoter (Primb-1), or by a promoter whose primary expression was in the sensory neurons responsible for the chemotaxis behavior (Psra-6, Podr-10). The behavioural phenotype was also rescued by over-expressing an atypical fAD-linked mutation in PS1 (PS1ΔS169) that has been reported to leave the Notch pathway intact. An examination of the morphology of polymodal nociceptive (ASH) neurons responsible for the chemotaxis behavior also showed increased neurodegeneration over time in sel-12 mutant worms that could be rescued by the same transgenes that rescued the behaviour, demonstrating a parallel with the observed behavioral deficits. Thus, we report an Aß-independent neurodegeneration in C. elegans that was rescued by cell specific over-expression of wild-type human presenilin.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Caenorhabditis elegans Proteins , Caenorhabditis elegans , Mutation , Presenilin-1 , Animals , Humans , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Chemotaxis/genetics , Disease Models, Animal , Presenilin-1/genetics , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/pathology
6.
Transl Psychiatry ; 14(1): 367, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39256358

ABSTRACT

This article delves into Alzheimer's disease (AD), a prevalent neurodegenerative condition primarily affecting the elderly. It is characterized by progressive memory and cognitive impairments, severely disrupting daily life. Recent research highlights the potential involvement of microRNAs in the pathogenesis of AD. MicroRNAs (MiRNAs), short non-coding RNAs comprising 20-24 nucleotides, significantly influence gene regulation by hindering translation or promoting degradation of target genes. This review explores the role of specific miRNAs in AD progression, focusing on their impact on ß-amyloid (Aß) peptide accumulation, intracellular aggregation of hyperphosphorylated tau proteins, mitochondrial dysfunction, neuroinflammation, oxidative stress, and the expression of the APOE4 gene. Our insights contribute to understanding AD's pathology, offering new avenues for identifying diagnostic markers and developing novel therapeutic targets.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , MicroRNAs , Oxidative Stress , tau Proteins , Alzheimer Disease/genetics , Alzheimer Disease/therapy , Alzheimer Disease/diagnosis , Alzheimer Disease/metabolism , Humans , MicroRNAs/genetics , Amyloid beta-Peptides/metabolism , tau Proteins/metabolism , tau Proteins/genetics , Oxidative Stress/genetics , Animals , Apolipoprotein E4/genetics , Gene Expression Regulation
8.
Cereb Cortex ; 34(9)2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39323397

ABSTRACT

Glaucoma and Alzheimer's disease are critical degenerative neuropathies with global impact. Previous studies have indicated that glaucomatous damage could extend beyond ocular structures, leading to brain alterations potentially associated with Alzheimer's disease risk. This study aimed to explore the causal associations among glaucoma, brain alterations, and Alzheimer's disease. We conducted a comprehensive investigation into the genetic correlation and causality between glaucoma, glaucoma endophenotypes, cerebral cortical surficial area and thickness, and Alzheimer's disease (including late-onset Alzheimer's disease, cognitive performance, and reaction time) using linkage disequilibrium score regression and Mendelian randomization. This study showed suggestive genetic correlations between glaucoma, cortical structures, and Alzheimer's disease. The genetically predicted all-caused glaucoma was nominally associated with a decreased risk of Alzheimer's disease (OR = 0.96, 95% CI: 0.93-0.99, P = 0.013). We found evidence for suggestive causality between glaucoma (endophenotypes) and 20 cortical regions and between 29 cortical regions and Alzheimer's disease (endophenotypes). Four cortical regions were causally associated with cognitive performance or reaction time at a significant threshold (P < 6.2E-04). Thirteen shared cortical regions between glaucoma (endophenotypes) and Alzheimer's disease (endophenotypes) were identified. Our findings complex causal relationships among glaucoma, cerebral cortical structures, and Alzheimer's disease. More studies are required to clarify the mediation effect of cortical alterations in the relationship between glaucoma and Alzheimer's disease.


Subject(s)
Alzheimer Disease , Cerebral Cortex , Glaucoma , Mendelian Randomization Analysis , Humans , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Glaucoma/genetics , Cerebral Cortex/pathology , Female , Male , Aged , Genetic Predisposition to Disease/genetics , Endophenotypes , Polymorphism, Single Nucleotide
9.
PLoS Comput Biol ; 20(9): e1012346, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39236079

ABSTRACT

Inferring the driving regulatory programs from comparative analysis of gene expression data is a cornerstone of systems biology. Many computational frameworks were developed to address this problem, including our iPAGE (information-theoretic Pathway Analysis of Gene Expression) toolset that uses information theory to detect non-random patterns of expression associated with given pathways or regulons. Our recent observations, however, indicate that existing approaches are susceptible to the technical biases that are inherent to most real world annotations. To address this, we have extended our information-theoretic framework to account for specific biases and artifacts in biological networks using the concept of conditional information. To showcase pyPAGE, we performed a comprehensive analysis of regulatory perturbations that underlie the molecular etiology of Alzheimer's disease (AD). pyPAGE successfully recapitulated several known AD-associated gene expression programs. We also discovered several additional regulons whose differential activity is significantly associated with AD. We further explored how these regulators relate to pathological processes in AD through cell-type specific analysis of single cell and spatial gene expression datasets. Our findings showcase the utility of pyPAGE as a precise and reliable biomarker discovery in complex diseases such as Alzheimer's disease.


Subject(s)
Alzheimer Disease , Gene Expression Profiling , Alzheimer Disease/genetics , Humans , Gene Expression Profiling/methods , Computational Biology/methods , Gene Regulatory Networks/genetics , Software , Databases, Genetic , Systems Biology/methods
10.
J Am Heart Assoc ; 13(18): e033850, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39258525

ABSTRACT

BACKGROUND: Sleep apnea (SA) has been linked to an increased risk of dementia in numerous observational studies; whether this is driven by neurodegenerative, vascular, or other mechanisms is not clear. We sought to examine the bidirectional causal relationships between SA, Alzheimer disease (AD), coronary artery disease (CAD), and ischemic stroke using Mendelian randomization. METHODS AND RESULTS: Using summary statistics from 4 recent, large genome-wide association studies of SA (n=523 366), AD (n=94 437), CAD (n=1 165 690), and stroke (n=1 308 460), we conducted bidirectional 2-sample Mendelian randomization analyses. Our primary analytic method was fixed-effects inverse variance-weighted (IVW) Mendelian randomization; diagnostics tests and sensitivity analyses were conducted to verify the robustness of the results. We identified a significant causal effect of SA on the risk of CAD (odds ratio [ORIVW]=1.35 per log-odds increase in SA liability [95% CI=1.25-1.47]) and stroke (ORIVW=1.13 [95% CI=1.01-1.25]). These associations were somewhat attenuated after excluding single-nucleotide polymorphisms associated with body mass index (ORIVW=1.26 [95% CI=1.15-1.39] for CAD risk; ORIVW=1.08 [95% CI=0.96-1.22] for stroke risk). SA was not causally associated with a higher risk of AD (ORIVW=1.14 [95% CI=0.91-1.43]). We did not find causal effects of AD, CAD, or stroke on risk of SA. CONCLUSIONS: These results suggest that SA increased the risk of CAD, and the identified causal association with stroke risk may be confounded by body mass index. Moreover, no causal effect of SA on AD risk was found. Future studies are warranted to investigate cardiovascular pathways between sleep disorders, including SA, and dementia.


Subject(s)
Alzheimer Disease , Genome-Wide Association Study , Mendelian Randomization Analysis , Sleep Apnea Syndromes , Humans , Alzheimer Disease/genetics , Alzheimer Disease/epidemiology , Alzheimer Disease/diagnosis , Sleep Apnea Syndromes/genetics , Sleep Apnea Syndromes/epidemiology , Sleep Apnea Syndromes/complications , Sleep Apnea Syndromes/diagnosis , Risk Factors , Polymorphism, Single Nucleotide , Risk Assessment/methods , Coronary Artery Disease/genetics , Coronary Artery Disease/epidemiology , Coronary Artery Disease/diagnosis , Genetic Predisposition to Disease , Cardiovascular Diseases/genetics , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/etiology , Ischemic Stroke/genetics , Ischemic Stroke/epidemiology , Ischemic Stroke/etiology
11.
Transl Psychiatry ; 14(1): 371, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39266518

ABSTRACT

Observational and genetic studies have reported the relationship between dyslexia and Alzheimer's disease (AD). Until now, the causal effect of dyslexia on AD risk has remained unclear. We conducted a two-sample univariable Mendelian randomization (MR) analysis to determine the causal association between dyslexia and the risk of AD, vascular dementia (VD), Lewy body dementia (LBD), and frontotemporal dementia (FTD) and its four subtypes. First, we selected 42 dyslexia genetic variants from a large-scale genome-wide association studies (GWAS) dataset and extracted their corresponding GWAS summary statistics from AD, VD, LBD, and FTD. Second, we selected four MR methods, including inverse-variance weighted (IVW), weighted median, MR-Egger, and MR-PRESSO. Heterogeneity, horizontal pleiotropy, and leave-one-out sensitivity analysis were then used to evaluate the reliability of all causal estimates. We also conducted multivariable MR (MVMR) and mediation analysis to assess the potential mediating role of cognitive performance (CP) or educational achievement (EA) on the causal association between dyslexia and AD. Two MVMR methods, including MV IVW and MV-Egger, and two-step MR were used to perform the analysis. Using IVW, we found a significant causal association between increased dyslexia and increased risk of AD (OR = 1.15, 95% CI: 1.04-1.28, P = 0.006), but not VD, LBD, FTD, or its four subtypes. MR-PRESSO further supported the statistically significant association between dyslexia and AD (OR = 1.15, 95% CI: 1.05-1.27, P = 0.006). All sensitivity analyses confirmed the reliability of causal estimates. Using MV IVW and mediation analysis, we found no causal relationship between dyslexia and AD after adjusting for CP but not EA, CP mediated the total effect of dyslexia on AD with a proportion of 46.32%. We provide genetic evidence to support a causal effect of increased dyslexia on increased risk of AD, which was largely mediated by CP. Reading activity may be a potential intervention strategy for AD by improving cognitive function.


Subject(s)
Alzheimer Disease , Dyslexia , Frontotemporal Dementia , Genome-Wide Association Study , Mendelian Randomization Analysis , Humans , Dyslexia/genetics , Frontotemporal Dementia/genetics , Alzheimer Disease/genetics , Lewy Body Disease/genetics , Dementia, Vascular/genetics , Dementia/genetics , Dementia/etiology , Dementia/epidemiology , Risk Factors , Causality
12.
Sci Rep ; 14(1): 21163, 2024 09 10.
Article in English | MEDLINE | ID: mdl-39256511

ABSTRACT

The generation of new neurons at the hippocampal neurogenic niche, known as adult hippocampal neurogenesis (AHN), and its impairment, have been implicated in Alzheimer's disease (AD). MicroRNA-132 (miR-132), the most consistently downregulated microRNA (miRNA) in AD, was recently identified as a potent regulator of AHN, exerting multilayered proneurogenic effects in adult neural stem cells (NSCs) and their progeny. Supplementing miR-132 in AD mouse brain restores AHN and relevant memory deficits, yet the exact mechanisms involved are still unknown. Here, we identify NACC2 as a novel miR-132 target implicated in both AHN and AD. miR-132 deficiency in mouse hippocampus induces Nacc2 expression and inflammatory signaling in adult NSCs. We show that miR-132-dependent regulation of NACC2 is involved in the initial stages of human NSC differentiation towards astrocytes and neurons. Later, NACC2 function in astrocytic maturation becomes uncoupled from miR-132. We demonstrate that NACC2 is present in reactive astrocytes surrounding amyloid plaques in mouse and human AD hippocampus, and that there is an anticorrelation between miR-132 and NACC2 levels in AD and upon induction of inflammation. Unraveling the molecular mechanisms by which miR-132 regulates neurogenesis and cellular reactivity in AD, will provide valuable insights towards its possible application as a therapeutic target.


Subject(s)
Alzheimer Disease , Astrocytes , Hippocampus , MicroRNAs , Neural Stem Cells , Neurogenesis , MicroRNAs/genetics , MicroRNAs/metabolism , Neurogenesis/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Humans , Neural Stem Cells/metabolism , Mice , Hippocampus/metabolism , Hippocampus/pathology , Astrocytes/metabolism , Neurons/metabolism , Cell Differentiation , Gene Expression Regulation
13.
Protein Sci ; 33(10): e5147, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39276018

ABSTRACT

Alzheimer's disease (AD) is one of the most common forms of dementia and neurodegenerative diseases, characterized by the formation of neuritic plaques and neurofibrillary tangles. Many different proteins participate in this complicated pathogenic mechanism, and missense mutations can alter the folding and functions of these proteins, significantly increasing the risk of AD. However, many methods to identify AD-causing variants did not consider the effect of mutations from the perspective of a protein three-dimensional environment. Here, we present a machine learning-based analysis to classify the AD-causing mutations from their benign counterparts in 21 AD-related proteins leveraging both sequence- and structure-based features. Using computational tools to estimate the effect of mutations on protein stability, we first observed a bias of the pathogenic mutations with significant destabilizing effects on family AD-related proteins. Combining this insight, we built a generic predictive model, and improved the performance by tuning the sample weights in the training process. Our final model achieved the performance on area under the receiver operating characteristic curve up to 0.95 in the blind test and 0.70 in an independent clinical validation, outperforming all the state-of-the-art methods. Feature interpretation indicated that the hydrophobic environment and polar interaction contacts were crucial to the decision on pathogenic phenotypes of missense mutations. Finally, we presented a user-friendly web server, AlzDiscovery, for researchers to browse the predicted phenotypes of all possible missense mutations on these 21 AD-related proteins. Our study will be a valuable resource for AD screening and the development of personalized treatment.


Subject(s)
Alzheimer Disease , Mutation, Missense , Alzheimer Disease/genetics , Humans , Machine Learning , Computational Biology/methods , Software , Protein Conformation
14.
Nutrients ; 16(17)2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39275195

ABSTRACT

The causal association of circulating metabolites with dementia remains uncertain. We assessed the causal association of circulating metabolites with dementia utilizing Mendelian randomization (MR) methods. We performed univariable MR analysis to evaluate the associations of 486 metabolites with dementia, Alzheimer's disease (AD), and vascular dementia (VaD) risk. For secondary validation, we replicated the analyses using an additional dataset with 123 metabolites. We observed 118 metabolites relevant to the risk of dementia, 59 of which were lipids, supporting the crucial role of lipids in dementia pathogenesis. After Bonferroni adjustment, we identified nine traits of HDL particles as potential causal mediators of dementia. Regarding dementia subtypes, protective effects were observed for epiandrosterone sulfate on AD (OR = 0.60, 95% CI: 0.48-0.75) and glycoproteins on VaD (OR = 0.89, 95% CI: 0.83-0.95). Bayesian model averaging MR (MR-BMA) analysis was further conducted to prioritize the predominant metabolites for dementia risk, which highlighted the mean diameter of HDL particles and the concentration of very large HDL particles as the predominant protective factors against dementia. Moreover, pathway analysis identified 17 significant and 2 shared metabolic pathways. These findings provide support for the identification of promising predictive biomarkers and therapeutic targets for dementia.


Subject(s)
Alzheimer Disease , Biomarkers , Dementia , Mendelian Randomization Analysis , Humans , Dementia/blood , Dementia/genetics , Alzheimer Disease/blood , Alzheimer Disease/genetics , Biomarkers/blood , Risk Factors , Bayes Theorem , Dementia, Vascular/blood , Dementia, Vascular/genetics , Male , Female
15.
Int J Mol Sci ; 25(17)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39273347

ABSTRACT

In patients with Alzheimer's disease (AD) and in animal models, the increased accumulation of amyloid ß (Aß) in retinal blood vessels strongly correlates with brain amyloid deposits and cognitive decline. The accumulation of Aß in blood vessels may result from impaired transcytosis and a dysfunctional ocular glymphatic system in AD. High-dose fish oil (FO) supplementation has been shown to significantly change the expression of major facilitator superfamily domain-containing protein 2a (Mfsd2a), a key regulator of transcytosis, and Aquaporin 4 (Aqp4), an essential component of the glymphatic system in the retinas of WT mice. We examined the expression of Mfsd2a and Aqp4 in the retinas of 4-month-old 5xFAD female mice supplemented with high-dose FO for three weeks. There was a significant increase in Mfsd2a expression in 5xFAD retinas supplemented with FO compared to control 5xFAD mice. Additionally, the increase in Aqp4 expression observed in 4-month-old 5xFAD retinas, indicative of an impaired glymphatic system, was significantly decreased. Simultaneously, Aß accumulation in 5xFAD retinal blood vessels was reduced following FO supplementation. These findings suggest that high-dose FO supplementation could serve as an adjunct in developing new treatments aimed at improving the regulation of transcytosis or the function of the glymphatic system in the AD retina.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Aquaporin 4 , Dietary Supplements , Disease Models, Animal , Fish Oils , Mice, Transgenic , Retinal Vessels , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Aquaporin 4/metabolism , Aquaporin 4/genetics , Amyloid beta-Peptides/metabolism , Mice , Female , Retinal Vessels/metabolism , Retinal Vessels/drug effects , Fish Oils/pharmacology , Fish Oils/administration & dosage , Symporters/metabolism , Symporters/genetics , Humans
16.
Zhen Ci Yan Jiu ; 49(8): 858-866, 2024 Aug 25.
Article in English, Chinese | MEDLINE | ID: mdl-39318316

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease with high incidence in the elderly population, and the synaptic changes in central neurons are the key pathological feature. The clinical effect of acupuncture and moxibustion in the treatment of AD is positive, and the research on the mechanism of acupuncture intervention of AD from the perspective of central synaptic plasticity regulation has been conducted uninterruptedly. In the present paper, we made a summation about the relevant experimental studies in recent years, and analyzed its mechanisms underlying improvement of AD by regulating synaptic plasticity from 1) repairing synaptic structure (synaptic contact area ï¼»total number of synapses, synaptic surface density, synaptic number densityï¼½, postsynaptic dense zone thickness, synaptic gap width, and interface curvature), 2) improving synaptic transmission efficiency (regulating long-term potentiation and long-term depression), 3) promoting the expression of synapse related proteins (synaptophysin, postsynaptic density protein 95, growth associated protein 43), 4) regulating the expression of neurotransmitters (acetylcholine, monoamines, amino acids, etc.) and receptors (α7 nicotinic acetylcholine receptor, glutaminergic receptor, etc.), and 5) improving the level of neurotrophic factors (brain derived neurotrophic factor, BDNF) and BDNF/SYN/microtubule-associated protein 2 signaling, etc., hoping to provide a reference for future studies.


Subject(s)
Acupuncture Therapy , Alzheimer Disease , Neuronal Plasticity , Alzheimer Disease/therapy , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Humans , Animals , Synapses/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics
17.
Alzheimers Res Ther ; 16(1): 207, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39322953

ABSTRACT

BACKGROUND: Several blood-based biomarkers offer the opportunity of in vivo detection of brain pathology and neurodegeneration in Alzheimer disease with high specificity and sensitivity, but the performance of amyloid-ß (Aß) measurements remains under evaluation. Autosomal dominant Alzheimer disease (ADAD) with mutations in PSEN1, PSEN2 and APP can be studied as a model for sporadic Alzheimer disease. However, clarifying the genetic effects on the Aß-levels in different matrices such as cerebrospinal fluid or plasma is crucial for generalizability and utility of data. We aimed to explore plasma Aß concentrations over the Alzheimer disease continuum in a longitudinal cohort of genetic Alzheimer disease. METHODS: 92 plasma samples were collected from at-risk individuals (n = 47) in a Swedish cohort of ADAD, including 18 mutation carriers (13 APPswe (p.KM670/671NL) MC), 5 PSEN1 (p.H163Y) MC) and 29 non-carriers (NC) as the reference group. Concentrations of Aß1-38, Aß1-40 and Aß1-42 were analyzed in plasma using immunoprecipitation coupled to tandem liquid chromatography mass spectrometry (IP-LC-MS/MS). Cross-sectional and repeated-measures data analyses were investigated family-wise, applying non-parametric tests as well as mixed-effects models. RESULTS: Cross-sectional analysis at baseline showed more than a 3-fold increase in all plasma Aß peptides in APPswe MC, regardless of clinical status, compared to controls (p < 0.01). PSEN1 (p.H163Y) presymptomatic MC had a decrease of plasma Aß1-38 compared to controls (p < 0.05). There was no difference in Aß1-42/1-40 ratio between APPswe MC (PMC and SMC), PSEN1 MC (PMC) and controls at baseline. Notably, both cross-sectional data and repeated-measures analysis suggested that APPswe MC have a stable Aß1-42/1-40 ratio with increasing age, in contrast to the decrease seen with aging in both controls and PSEN1 (p.H163Y) MC. CONCLUSION: These data show very strong mutation-specific effects on Aß profiles in blood, most likely due to a ubiquitous production outside of the CNS. Hence, analyses in an unselected clinical setting might unintentionally disclose genetic status. Furthermore, our findings suggest that the Aß ratio might be a poor indicator of brain Aß pathology in selected genetic cases. The very small sample size is a limitation that needs to be considered but reflects the scarcity of longitudinal in vivo data from genetic cohorts.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Amyloid beta-Protein Precursor , Presenilin-1 , Presenilin-2 , Humans , Alzheimer Disease/blood , Alzheimer Disease/genetics , Alzheimer Disease/cerebrospinal fluid , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/cerebrospinal fluid , Male , Female , Sweden , Middle Aged , Presenilin-1/genetics , Presenilin-2/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/blood , Aged , Mutation , Adult , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Longitudinal Studies , Cohort Studies , Peptide Fragments/blood , Peptide Fragments/cerebrospinal fluid , Peptide Fragments/genetics
18.
Elife ; 132024 Sep 27.
Article in English | MEDLINE | ID: mdl-39329365

ABSTRACT

The recent, controversial approval of antibody-based treatments for Alzheimer's disease (AD) is fueling a heated debate on the molecular determinants of this condition. The discussion should also incorporate a critical revision of the limitations of preclinical mouse models in advancing our understanding of AD. We critically discuss the limitations of animal models, stressing the need for careful consideration of how experiments are designed and results interpreted. We identify the shortcomings of AD models to recapitulate the complexity of the human disease. We dissect these issues at the quantitative, qualitative, temporal, and context-dependent levels. We argue that these models are based on the oversimplistic assumptions proposed by the amyloid cascade hypothesis (ACH) of AD and fail to account for the multifactorial nature of the condition. By shedding light on the constraints of current experimental tools, this review aims to foster the development and implementation of more clinically relevant tools. While we do not rule out a role for preclinical models, we call for alternative approaches to be explored and, most importantly, for a re-evaluation of the ACH.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Alzheimer Disease/genetics , Animals , Mice , Humans
19.
Cells ; 13(18)2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39329779

ABSTRACT

A characteristic feature of Alzheimer's disease (AD) is the formation of neuronal extracellular senile plaques composed of aggregates of fibrillar amyloid ß (Aß) peptides, with the Aß1-42 peptide being the most abundant species. These Aß peptides have been proposed to contribute to the pathophysiology of the disease; however, there are few tools available to test this hypothesis directly. In particular, there are no data that establish a dose-response relationship between Aß peptide expression level and disease. We have generated a panel of transgenic Caenorhabditis elegans strains expressing the human Aß1-42 peptide under the control of promoter regions of two pan-neuronal expressed genes, snb-1 and rgef-1. Phenotypic data show strong age-related defects in motility, subtle changes in chemotaxis, reduced median and maximum lifespan, changes in health span indicators, and impaired learning. The Aß1-42 expression level of these strains differed as a function of promoter identity and transgene copy number, and the timing and severity of phenotypes mediated by Aß1-42 were strongly positively correlated with expression level. The pan-neuronal expression of varying levels of human Aß1-42 in a nematode model provides a new tool to investigate the in vivo toxicity of neuronal Aß expression and the molecular and cellular mechanisms underlying AD progression in the absence of endogenous Aß peptides. More importantly, it allows direct quantitative testing of the dose-response relationship between neuronal Aß peptide expression and disease for the first time. These strains may also be used to develop screens for novel therapeutics to treat Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides , Animals, Genetically Modified , Caenorhabditis elegans , Neurons , Phenotype , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/genetics , Amyloid beta-Peptides/metabolism , Animals , Neurons/metabolism , Neurons/pathology , Humans , Peptide Fragments/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Longevity/genetics , Promoter Regions, Genetic/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics
20.
J Alzheimers Dis ; 101(3): 813-822, 2024.
Article in English | MEDLINE | ID: mdl-39269833

ABSTRACT

Background: There is no molecular test for Alzheimer's disease (AD) using self-collected samples, nor is there a definitive molecular test for AD. We demonstrate an accurate and potentially definitive TempO-Seq® gene expression test for AD using fingerstick blood spotted and dried on filter paper, a sample that can be collected in any doctor's office or can be self-collected. Objective: Demonstrate the feasibility of developing an accurate test for the classification of persons with AD from a minimally invasive sample of fingerstick blood spotted on filter paper which can be obtained in any doctor's office or self-collected to address health disparities. Methods: Fingerstick blood samples from patients clinically diagnosed with AD, Parkinson's disease (PD), or asymptomatic controls were spotted onto filter paper in the doctor's office, dried, and shipped to BioSpyder for testing. Three independent patient cohorts were used for training/retraining and testing/retesting AD and PD classification algorithms. Results: After initially identifying a 770 gene classification signature, a minimum set of 68 genes was identified providing classification test areas under the ROC curve of 0.9 for classifying patients as having AD, and 0.94 for classifying patients as having PD. Conclusions: These data demonstrate the potential to develop a screening and/or definitive, minimally invasive, molecular diagnostic test for AD and PD using dried fingerstick blood spot samples that are collected in a doctor's office or clinic, or self-collected, and thus, can address health disparities. Whether the test can classify patients with AD earlier then possible with cognitive testing remains to be determined.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/genetics , Alzheimer Disease/diagnosis , Alzheimer Disease/blood , Female , Male , Aged , Parkinson Disease/genetics , Parkinson Disease/blood , Parkinson Disease/diagnosis , Middle Aged , Aged, 80 and over , Blood Specimen Collection , Gene Expression Profiling/methods
SELECTION OF CITATIONS
SEARCH DETAIL