Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 42
Filtrer
1.
Sci Adv ; 10(14): eadk3674, 2024 Apr 05.
Article de Anglais | MEDLINE | ID: mdl-38569027

RÉSUMÉ

The immune system substantially influences age-related cognitive decline and Alzheimer's disease (AD) progression, affected by genetic and environmental factors. In a Mayo Clinic Study of Aging cohort, we examined how risk factors like APOE genotype, age, and sex affect inflammatory molecules and AD biomarkers in cerebrospinal fluid (CSF). Among cognitively unimpaired individuals over 65 (N = 298), we measured 365 CSF inflammatory molecules, finding age, sex, and diabetes status predominantly influencing their levels. We observed age-related correlations with AD biomarkers such as total tau, phosphorylated tau-181, neurofilament light chain (NfL), and YKL40. APOE4 was associated with lower Aß42 and higher SNAP25 in CSF. We explored baseline variables predicting cognitive decline risk, finding age, CSF Aß42, NfL, and REG4 to be independently correlated. Subjects with older age, lower Aß42, higher NfL, and higher REG4 at baseline had increased cognitive impairment risk during follow-up. This suggests that assessing CSF inflammatory molecules and AD biomarkers could predict cognitive impairment risk in the elderly.


Sujet(s)
Maladie d'Alzheimer , Dysfonctionnement cognitif , Humains , Sujet âgé , Maladie d'Alzheimer/diagnostic , Maladie d'Alzheimer/étiologie , Maladie d'Alzheimer/liquide cérébrospinal , Dysfonctionnement cognitif/diagnostic , Dysfonctionnement cognitif/étiologie , Protéines tau , Marqueurs biologiques , Peptides bêta-amyloïdes , Fragments peptidiques
2.
bioRxiv ; 2024 Jan 19.
Article de Anglais | MEDLINE | ID: mdl-38293184

RÉSUMÉ

Loss-of-function mutations in the genes encoding PINK1 and PRKN result in early-onset Parkinson disease (EOPD). Together the encoded enzymes direct a neuroprotective pathway that ensures the elimination of damaged mitochondria via autophagy. We performed a genome-wide high content imaging miRNA screen for inhibitors of the PINK1-PRKN pathway and identified all three members of the miRNA family 29 (miR-29). Using RNAseq we identified target genes and found that siRNA against ATG9A phenocopied the effects of miR-29 and inhibited the initiation of PINK1-PRKN mitophagy. Furthermore, we discovered two rare, potentially deleterious, missense variants (p.R631W and p.S828L) in our EOPD cohort and tested them experimentally in cells. While expression of wild-type ATG9A was able to rescue the effects of miR-29a, the EOPD-associated variants behaved like loss-of-function mutations. Together, our study validates miR-29 and its target gene ATG9A as novel regulators of mitophagy initiation. It further serves as proof-of-concept of finding novel, potentially disease-causing EOPD-linked variants specifically in mitophagy regulating genes. The nomination of genetic variants and biological pathways is important for the stratification and treatment of patients that suffer from devastating diseases, such as EOPD.

3.
Parkinsonism Relat Disord ; 119: 105935, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38072719

RÉSUMÉ

INTRODUCTION: Substantial heterogeneity between individual patients in the clinical presentation of Parkinson's disease (PD) has led to the classification of distinct PD subtypes. However, genetic susceptibility factors for specific PD subtypes are not well understood. Therefore, the present study aimed to investigate the genetics of PD heterogeneity by performing a genome-wide association study (GWAS) of PD subtypes. METHODS: A total of 799 PD patients were included and classified into tremor-dominant (TD) (N = 345), akinetic-rigid (AR) (N = 227), gait-difficulty (GD) (N = 82), and mixed (MX) (N = 145) phenotypic subtypes. After array genotyping and subsequent imputation, a total of 7,918,344 variants were assessed for association with each PD subtype using logistic regression models that were adjusted for age, sex, and the top five principal components of GWAS data. RESULTS: We identified one genome-wide significant association (P < 5 × 10-8), which was between the MIR3976HG rs7504760 variant and the AR subtype (Odds ratio [OR] = 6.12, P = 2.57 × 10-8). Suggestive associations (P < 1 × 10-6) were observed regarding TD for RP11-497G19.3/RP11-497G19.1 rs7304254 (OR = 3.33, P = 3.89 × 10-7), regarding GD for HES2 rs111473931 (OR = 3.18, P = 6.85 × 10-7), RP11-400D2.3/CTD-2012I17.1 rs149082205 (OR = 8.96, P = 9.08 × 10-7), and RN7SL408P/SGK1 rs56161738 (OR = 2.97, P = 6.19 × 10-7), and regarding MX for MMRN2 rs112991171 (OR = 4.98, P = 1.02 × 10-7). CONCLUSION: Our findings indicate that genetic variation may account for part of the clinical heterogeneity of PD. In particular, we found a novel genome-wide significant association between MIR3976HG variation and the AR PD subtype. Replication of these findings will be important in order to better define the genetic architecture of clinical variability in PD disease presentation.


Sujet(s)
Maladie de Parkinson , Humains , Maladie de Parkinson/génétique , Maladie de Parkinson/complications , Étude d'association pangénomique , Tremblement/complications , Odds ratio
4.
Mol Psychiatry ; 29(3): 809-819, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38135757

RÉSUMÉ

ABCA7 loss-of-function variants are associated with increased risk of Alzheimer's disease (AD). Using ABCA7 knockout human iPSC models generated with CRISPR/Cas9, we investigated the impacts of ABCA7 deficiency on neuronal metabolism and function. Lipidomics revealed that mitochondria-related phospholipids, such as phosphatidylglycerol and cardiolipin were reduced in the ABCA7-deficient iPSC-derived cortical organoids. Consistently, ABCA7 deficiency-induced alterations of mitochondrial morphology accompanied by reduced ATP synthase activity and exacerbated oxidative damage in the organoids. Furthermore, ABCA7-deficient iPSC-derived neurons showed compromised mitochondrial respiration and excess ROS generation, as well as enlarged mitochondrial morphology compared to the isogenic controls. ABCA7 deficiency also decreased spontaneous synaptic firing and network formation in iPSC-derived neurons, in which the effects were rescued by supplementation with phosphatidylglycerol or NAD+ precursor, nicotinamide mononucleotide. Importantly, effects of ABCA7 deficiency on mitochondria morphology and synapses were recapitulated in synaptosomes isolated from the brain of neuron-specific Abca7 knockout mice. Together, our results provide evidence that ABCA7 loss-of-function contributes to AD risk by modulating mitochondria lipid metabolism.


Sujet(s)
Transporteurs ABC , Cellules souches pluripotentes induites , Métabolisme lipidique , Souris knockout , Mitochondries , Neurones , Mitochondries/métabolisme , Neurones/métabolisme , Humains , Animaux , Métabolisme lipidique/physiologie , Souris , Cellules souches pluripotentes induites/métabolisme , Transporteurs ABC/métabolisme , Transporteurs ABC/génétique , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/génétique , Encéphale/métabolisme
5.
Sci Rep ; 13(1): 16855, 2023 10 06.
Article de Anglais | MEDLINE | ID: mdl-37803069

RÉSUMÉ

Mounting evidence highlights the crucial role of aging in the pathogenesis of Alzheimer's disease (AD). We have previously explored human apoE-targeted replacement mice across different ages and identified distinct molecular pathways driven by aging. However, the specific contribution of different brain cell types to the gene modules underlying these pathways remained elusive. To bridge this knowledge gap, we employed a computational deconvolution approach to examine cell-type-specific gene expression profiles in major brain cell types, including astrocytes (AS), microglia (MG), oligodendroglia (OG), neurons (NEU), and vascular cells (VC). Our findings revealed that immune module genes were predominantly expressed in MG, OG, and VC. The lipid metabolism module genes were primarily expressed in AS, MG, and OG. The mitochondria module genes showed prominent expression in VC, and the synapse module genes were primarily expressed in NEU and VC. Furthermore, we identified intra- and inter-cell-type interactions among these module genes and validated their aging-associated expression changes using published single cell studies. Our study dissected bulk brain transcriptomics data at the cellular level, providing a closer examination of the cell-type contributions to the molecular pathways driven by aging.


Sujet(s)
Maladie d'Alzheimer , Transcriptome , Souris , Humains , Animaux , Analyse de profil d'expression de gènes , Maladie d'Alzheimer/métabolisme , Vieillissement/génétique , Encéphale/métabolisme
6.
Nat Immunol ; 24(11): 1854-1866, 2023 Nov.
Article de Anglais | MEDLINE | ID: mdl-37857825

RÉSUMÉ

Microglial involvement in Alzheimer's disease (AD) pathology has emerged as a risk-determining pathogenic event. While apolipoprotein E (APOE) is known to modify AD risk, it remains unclear how microglial apoE impacts brain cognition and AD pathology. Here, using conditional mouse models expressing apoE isoforms in microglia and central nervous system-associated macrophages (CAMs), we demonstrate a cell-autonomous effect of apoE3-mediated microglial activation and function, which are negated by apoE4. Expression of apoE3 in microglia/CAMs improves cognitive function, increases microglia surrounding amyloid plaque and reduces amyloid pathology and associated toxicity, whereas apoE4 expression either compromises or has no effects on these outcomes by impairing lipid metabolism. Single-cell transcriptomic profiling reveals increased antigen presentation and interferon pathways upon apoE3 expression. In contrast, apoE4 expression downregulates complement and lysosomal pathways, and promotes stress-related responses. Moreover, in the presence of mouse endogenous apoE, microglial apoE4 exacerbates amyloid pathology. Finally, we observed a reduction in Lgals3-positive responsive microglia surrounding amyloid plaque and an increased accumulation of lipid droplets in APOE4 human brains and induced pluripotent stem cell-derived microglia. Our findings establish critical isoform-dependent effects of microglia/CAM-expressed apoE in brain function and the development of amyloid pathology, providing new insight into how apoE4 vastly increases AD risk.


Sujet(s)
Maladie d'Alzheimer , Souris , Animaux , Humains , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/anatomopathologie , Apolipoprotéine E4/génétique , Apolipoprotéine E4/métabolisme , Microglie/métabolisme , Apolipoprotéine E3/génétique , Apolipoprotéine E3/métabolisme , Plaque amyloïde/métabolisme , Plaque amyloïde/anatomopathologie , Apolipoprotéines E/génétique , Apolipoprotéines E/métabolisme , Encéphale , Homéostasie , Souris transgéniques
7.
Stem Cell Res Ther ; 14(1): 214, 2023 08 21.
Article de Anglais | MEDLINE | ID: mdl-37605285

RÉSUMÉ

BACKGROUND: The apolipoprotein E (APOE) gene is the strongest genetic risk factor for Alzheimer's disease (AD); however, how it modulates brain homeostasis is not clear. The apoE protein is a major lipid carrier in the brain transporting lipids such as cholesterol among different brain cell types. METHODS: We generated three-dimensional (3-D) cerebral organoids from human parental iPSC lines and its isogenic APOE-deficient (APOE-/-) iPSC line. To elucidate the cell-type-specific effects of APOE deficiency in the cerebral organoids, we performed scRNA-seq in the parental and APOE-/- cerebral organoids at Day 90. RESULTS: We show that APOE deficiency in human iPSC-derived cerebral organoids impacts brain lipid homeostasis by modulating multiple cellular and molecular pathways. Molecular profiling through single-cell RNA sequencing revealed that APOE deficiency leads to changes in cellular composition of isogenic cerebral organoids likely by modulating the eukaryotic initiation factor 2 (EIF2) signaling pathway as these events were alleviated by the treatment of an integrated stress response inhibitor (ISRIB). APOE deletion also leads to activation of the Wnt/ß-catenin signaling pathway with concomitant decrease of secreted frizzled-related protein 1 (SFRP1) expression in glia cells. Importantly, the critical role of apoE in cell-type-specific lipid homeostasis was observed upon APOE deletion in cerebral organoids with a specific upregulation of cholesterol biosynthesis in excitatory neurons and excessive lipid accumulation in astrocytes. Relevant to human AD, APOE4 cerebral organoids show altered neurogenesis and cholesterol metabolism compared to those with APOE3. CONCLUSIONS: Our work demonstrates critical roles of apoE in brain homeostasis and offers critical insights into the APOE4-related pathogenic mechanisms.


Sujet(s)
Apolipoprotéines E , Cerveau , Cellules souches pluripotentes induites , Humains , Apolipoprotéine E4 , Apolipoprotéines E/génétique , Différenciation cellulaire , Organoïdes , Cerveau/métabolisme
8.
JCI Insight ; 8(7)2023 04 10.
Article de Anglais | MEDLINE | ID: mdl-37036005

RÉSUMÉ

Cerebrovasculature is critical in maintaining brain homeostasis; its dysregulation often leads to vascular cognitive impairment and dementia (VCID) during aging. VCID is the second most prevalent cause of dementia in the elderly, after Alzheimer's disease (AD), with frequent cooccurrence of VCID and AD. While multiple factors are involved in the pathogenesis of AD and VCID, APOE4 increases the risk for both diseases. A major apolipoprotein E (apoE) receptor, the low-density lipoprotein receptor-related protein 1 (LRP1), is abundantly expressed in vascular mural cells (pericytes and smooth muscle cells). Here, we investigated how deficiency of vascular mural cell LRP1 affects the cerebrovascular system and cognitive performance using vascular mural cell-specific Lrp1-KO mice (smLrp1-/-) in a human APOE3 or APOE4 background. We found that spatial memory was impaired in the 13- to 16-month-old APOE4 smLrp1-/- mice but not in the APOE3 smLrp1-/- mice, compared with their respective littermate control mice. These disruptions in the APOE4 smLrp1-/- mice were accompanied with excess paravascular glial activation and reduced cerebrovascular collagen IV. In addition, blood-brain barrier (BBB) integrity was disrupted in the APOE4 smLrp1-/- mice. Together, our results suggest that vascular mural cell LRP1 modulates cerebrovasculature integrity and function in an APOE genotype-dependent manner.


Sujet(s)
Maladie d'Alzheimer , Apolipoprotéine E4 , Humains , Souris , Animaux , Sujet âgé , Nourrisson , Apolipoprotéine E4/génétique , Apolipoprotéine E3/métabolisme , Apolipoprotéines E/métabolisme , Barrière hémato-encéphalique/métabolisme , Maladie d'Alzheimer/anatomopathologie , Protéine-1 apparentée au récepteur des LDL/métabolisme
9.
Mol Neurobiol ; 60(1): 26-35, 2023 Jan.
Article de Anglais | MEDLINE | ID: mdl-36215026

RÉSUMÉ

Wnt and R-spondin (Rspo) proteins are two major types of endogenous Wnt/ß-catenin signaling agonists. While Wnt/ß-catenin signaling is greatly diminished in Alzheimer's disease (AD), it remains to be elucidated whether the inhibition of this pathway is associated with dysregulation of Wnt and Rspo proteins. By analyzing temporal cortex RNA-seq data of the human postmortem brain samples, we found that WNT1 and RRPO2 were significantly downregulated in human AD brains. In addition, the expression of Wnt acyltransferase porcupine (PORCN), which is essential for Wnt maturation and secretion, was greatly deceased in these human AD brains. Interestingly, the lowest levels of WNT1, PORCN, and RSPO2 expression were found in human AD brains carrying two copies of APOE4 allele, the strongest genetic risk factor of late-onset AD. Importantly, there were positive correlations among the levels of WNT1, PORCN, and RSPO2 expression in human AD brains. Supporting observations in humans, Wnt1, PORCN, and Rspo2 were downregulated and Wnt/ß-catenin signaling was diminished in the 5xFAD amyloid model mice. In human APOE-targeted replacement mice, downregulation of WNT1, PORCN, and RSPO2 expression was positively associated with aging and APOE4 genotype. Finally, WNT1 and PORCN expression and Wnt/ß-catenin signaling were inhibited in human APOE4 iPSC-derived astrocytes when compared to the isogenic APOE3 iPSC-derived astrocytes. Altogether, our findings suggest that the dysregulations of Wnt1, PORCN, and Rspo2 could be coordinated together to diminish Wnt/ß-catenin signaling in aging- and APOE4-dependent manners in the AD brain.


Sujet(s)
Maladie d'Alzheimer , Protéines et peptides de signalisation intercellulaire , Protéines membranaires , Voie de signalisation Wnt , Animaux , Humains , Souris , Acyltransferases/métabolisme , Maladie d'Alzheimer/génétique , Apolipoprotéine E4/génétique , Régulation négative , Protéines membranaires/métabolisme , Protéines et peptides de signalisation intercellulaire/métabolisme
10.
Front Genet ; 13: 984338, 2022.
Article de Anglais | MEDLINE | ID: mdl-36186441

RÉSUMÉ

The recent methodological advances in multi-omics approaches, including genomic, transcriptomic, metabolomic, lipidomic, and proteomic, have revolutionized the research field by generating "big data" which greatly enhanced our understanding of the molecular complexity of the brain and disease states. Network approaches have been routinely applied to single-omics data to provide critical insight into disease biology. Furthermore, multi-omics integration has emerged as both a vital need and a new direction to connect the different layers of information underlying disease mechanisms. In this review article, we summarize popular network analytic approaches for single-omics data and multi-omics integration and discuss how these approaches have been utilized in studying neurodegenerative diseases.

11.
J Exp Med ; 219(12)2022 12 05.
Article de Anglais | MEDLINE | ID: mdl-36107206

RÉSUMÉ

TREM2 is exclusively expressed by microglia in the brain and is strongly linked to the risk for Alzheimer's disease (AD). As microglial responses modulated by TREM2 are central to AD pathogenesis, enhancing TREM2 signaling has been explored as an AD therapeutic strategy. However, the effective therapeutic window targeting TREM2 is unclear. Here, by using microglia-specific inducible mouse models overexpressing human wild-type TREM2 (TREM2-WT) or R47H risk variant (TREM2-R47H), we show that TREM2-WT expression reduces amyloid deposition and neuritic dystrophy only during the early amyloid seeding stage, whereas TREM2-R47H exacerbates amyloid burden during the middle amyloid rapid growth stage. Single-cell RNA sequencing reveals suppressed disease-associated microglia (DAM) signature and reduced DAM population upon TREM2-WT expression in the early stage, whereas upregulated antigen presentation pathway is detected with TREM2-R47H expression in the middle stage. Together, our findings highlight the dynamic effects of TREM2 in modulating AD pathogenesis and emphasize the beneficial effect of enhancing TREM2 function in the early stage of AD development.


Sujet(s)
Maladie d'Alzheimer , Amyloïdose , Maladie d'Alzheimer/anatomopathologie , Amyloïde/métabolisme , Amyloïdose/anatomopathologie , Animaux , Encéphale/anatomopathologie , Humains , Glycoprotéines membranaires/génétique , Glycoprotéines membranaires/métabolisme , Souris , Microglie/métabolisme , Récepteurs immunologiques/génétique , Récepteurs immunologiques/métabolisme
12.
Nat Neurosci ; 25(8): 1020-1033, 2022 08.
Article de Anglais | MEDLINE | ID: mdl-35915180

RÉSUMÉ

The ε4 allele of the apolipoprotein E (APOE) gene, a genetic risk factor for Alzheimer's disease, is abundantly expressed in both the brain and periphery. Here, we present evidence that peripheral apoE isoforms, separated from those in the brain by the blood-brain barrier, differentially impact Alzheimer's disease pathogenesis and cognition. To evaluate the function of peripheral apoE, we developed conditional mouse models expressing human APOE3 or APOE4 in the liver with no detectable apoE in the brain. Liver-expressed apoE4 compromised synaptic plasticity and cognition by impairing cerebrovascular functions. Plasma proteome profiling revealed apoE isoform-dependent functional pathways highlighting cell adhesion, lipoprotein metabolism and complement activation. ApoE3 plasma from young mice improved cognition and reduced vessel-associated gliosis when transfused into aged mice, whereas apoE4 compromised the beneficial effects of young plasma. A human induced pluripotent stem cell-derived endothelial cell model recapitulated the plasma apoE isoform-specific effect on endothelial integrity, further supporting a vascular-related mechanism. Upon breeding with amyloid model mice, liver-expressed apoE4 exacerbated brain amyloid pathology, whereas apoE3 reduced it. Our findings demonstrate pathogenic effects of peripheral apoE4, providing a strong rationale for targeting peripheral apoE to treat Alzheimer's disease.


Sujet(s)
Maladie d'Alzheimer , Cellules souches pluripotentes induites , Maladie d'Alzheimer/métabolisme , Animaux , Apolipoprotéine E3/génétique , Apolipoprotéine E3/métabolisme , Apolipoprotéine E4/génétique , Apolipoprotéine E4/métabolisme , Apolipoprotéines E/génétique , Encéphale/métabolisme , Cognition , Humains , Cellules souches pluripotentes induites/métabolisme , Souris , Souris transgéniques , Isoformes de protéines/métabolisme
14.
Brain ; 145(7): 2472-2485, 2022 07 29.
Article de Anglais | MEDLINE | ID: mdl-34918030

RÉSUMÉ

Frontotemporal lobar degeneration with TDP-43 inclusions (FTLD-TDP) is a complex heterogeneous neurodegenerative disorder for which mechanisms are poorly understood. To explore transcriptional changes underlying FTLD-TDP, we performed RNA-sequencing on 66 genetically unexplained FTLD-TDP patients, 24 FTLD-TDP patients with GRN mutations and 24 control participants. Using principal component analysis, hierarchical clustering, differential expression and coexpression network analyses, we showed that GRN mutation carriers and FTLD-TDP-A patients without a known mutation shared a common transcriptional signature that is independent of GRN loss-of-function. After combining both groups, differential expression as compared to the control group and coexpression analyses revealed alteration of processes related to immune response, synaptic transmission, RNA metabolism, angiogenesis and vesicle-mediated transport. Deconvolution of the data highlighted strong cellular alterations that were similar in FTLD-TDP-A and GRN mutation carriers with NSF as a potentially important player in both groups. We propose several potentially druggable pathways such as the GABAergic, GDNF and sphingolipid pathways. Our findings underline new disease mechanisms and strongly suggest that affected pathways in GRN mutation carriers extend beyond GRN and contribute to genetically unexplained forms of FTLD-TDP-A.


Sujet(s)
Démence frontotemporale , Dégénérescence lobaire frontotemporale , Progranulines , Encéphale/métabolisme , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Démence frontotemporale/génétique , Démence frontotemporale/métabolisme , Dégénérescence lobaire frontotemporale/génétique , Dégénérescence lobaire frontotemporale/métabolisme , Humains , Protéines et peptides de signalisation intercellulaire/génétique , Protéines et peptides de signalisation intercellulaire/métabolisme , Mutation , Progranulines/génétique , Progranulines/métabolisme , Transcriptome
15.
Sci Transl Med ; 13(613): eabc9375, 2021 Sep 29.
Article de Anglais | MEDLINE | ID: mdl-34586832

RÉSUMÉ

Apolipoprotein E (APOE) genetic variants have been shown to modify Alzheimer's disease (AD) risk. We previously identified an APOE3 variant (APOE3-V236E), named APOE3-Jacksonville (APOE3-Jac), associated with healthy brain aging and reduced risk for AD and dementia with Lewy bodies (DLB). Herein, we resolved the functional mechanism by which APOE3-Jac reduces APOE aggregation and enhances its lipidation in human brains, as well as in cellular and biochemical assays. Compared to APOE3, expression of APOE3-Jac in astrocytes increases several classes of lipids in the brain including phosphatidylserine, phosphatidylethanolamine, phosphatidic acid, and sulfatide, critical for synaptic functions. Mice expressing APOE3-Jac have reduced amyloid pathology, plaque-associated immune responses, and neuritic dystrophy. The V236E substitution is also sufficient to reduce the aggregation of APOE4, whose gene allele is a major genetic risk factor for AD and DLB. These findings suggest that targeting APOE aggregation might be an effective strategy for treating a subgroup of individuals with AD and DLB.


Sujet(s)
Apolipoprotéine E3/génétique , Démence , Apolipoprotéines E , Démence/génétique , Humains
16.
Acta Neuropathol ; 142(5): 807-825, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34453582

RÉSUMÉ

APOE4 is a strong genetic risk factor for Alzheimer's disease and Dementia with Lewy bodies; however, how its expression impacts pathogenic pathways in a human-relevant system is not clear. Here using human iPSC-derived cerebral organoid models, we find that APOE deletion increases α-synuclein (αSyn) accumulation accompanied with synaptic loss, reduction of GBA levels, lipid droplet accumulation and dysregulation of intracellular organelles. These phenotypes are partially rescued by exogenous apoE2 and apoE3, but not apoE4. Lipidomics analysis detects the increased fatty acid utilization and cholesterol ester accumulation in apoE-deficient cerebral organoids. Furthermore, APOE4 cerebral organoids have increased αSyn accumulation compared to those with APOE3. Carrying APOE4 also increases apoE association with Lewy bodies in postmortem brains from patients with Lewy body disease. Our findings reveal the predominant role of apoE in lipid metabolism and αSyn pathology in iPSC-derived cerebral organoids, providing mechanistic insights into how APOE4 drives the risk for synucleinopathies.


Sujet(s)
Apolipoprotéines E/métabolisme , Métabolisme lipidique/physiologie , Organoïdes/anatomopathologie , Synucléinopathies/métabolisme , alpha-Synucléine/métabolisme , Animaux , Humains , Cellules souches pluripotentes induites , Souris , Organoïdes/métabolisme , Isoformes de protéines/métabolisme , Synucléinopathies/anatomopathologie
18.
Front Neurosci ; 15: 647974, 2021.
Article de Anglais | MEDLINE | ID: mdl-33897360

RÉSUMÉ

The ATP binding cassette subfamily A member 7 (ABCA7) gene is one of the significant susceptibility loci for Alzheimer's disease (AD). Furthermore, ABCA7 loss of function variants resulting from premature termination codon in the gene are associated with increased risk for AD. ABCA7 belongs to the ABC transporter family, which mediates the transport of diverse metabolites across the cell membrane. ABCA7 is also involved in modulating immune responses. Because the immune system and lipid metabolism causatively engage in the pathogenesis of AD, we investigated how ABCA7 haplodeficiency modulates the metabolic profile in mouse brains during acute immune response using a metabolomics approach through LC/Q-TOF-MS. Peripheral lipopolysaccharide (LPS) stimulation substantially influenced the metabolite content in the cortex, however, the effect on metabolic profiles in Abca7 heterozygous knockout mice (Abca7 ±) was modest compared to that in the control wild-type mice. Weighted gene co-expression network analysis (WGCNA) of the metabolomics dataset identified two modules influenced by LPS administration and ABCA7 haplodeficiency, in which glycerophospholipid metabolism, linoleic acid metabolism, and α-linolenic acid metabolism were identified as major pathways. Consistent with these findings, we also found that LPS stimulation increased the brain levels of eicosapentaenoic acid, oleic acid, and palmitic acid in Abca7 ± mice, but not control mice. Together, our results indicate that ABCA7 is involved in the crosstalk between fatty acid metabolism and inflammation in the brain, and disturbances in these pathways may contribute to the risk for AD.

19.
PLoS One ; 16(4): e0249305, 2021.
Article de Anglais | MEDLINE | ID: mdl-33861770

RÉSUMÉ

Genetic studies have shifted to sequencing-based rare variants discovery after decades of success in identifying common disease variants by Genome-Wide Association Studies using Single Nucleotide Polymorphism chips. Sequencing-based studies require large sample sizes for statistical power and therefore often inadvertently introduce batch effects because samples are typically collected, processed, and sequenced at multiple centers. Conventionally, batch effects are first detected and visualized using Principal Components Analysis and then controlled by including batch covariates in the disease association models. For sequencing-based genetic studies, because all variants included in the association analyses have passed sequencing-related quality control measures, this conventional approach treats every variant as equal and ignores the substantial differences still remaining in variant qualities and characteristics such as genotype quality scores, alternative allele fractions (fraction of reads supporting alternative allele at a variant position) and sequencing depths. In the Alzheimer's Disease Sequencing Project (ADSP) exome dataset of 9,904 cases and controls, we discovered hidden variant-level differences between sample batches of three sequencing centers and two exome capture kits. Although sequencing centers were included as a covariate in our association models, we observed differences at the variant level in genotype quality and alternative allele fraction between samples processed by different exome capture kits that significantly impacted both the confidence of variant detection and the identification of disease-associated variants. Furthermore, we found that a subset of top disease-risk variants came exclusively from samples processed by one exome capture kit that was more effective at capturing the alternative alleles compared to the other kit. Our findings highlight the importance of additional variant-level quality control for large sequencing-based genetic studies. More importantly, we demonstrate that automatically filtering out variants with batch differences may lead to false negatives if the batch discordances come largely from quality differences and if the batch-specific variants have better quality.


Sujet(s)
Étude d'association pangénomique , Séquençage nucléotidique à haut débit/méthodes , Allèles , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/anatomopathologie , Apolipoprotéines E/génétique , Bases de données génétiques , Exome , Femelle , Fréquence d'allèle , Prédisposition génétique à une maladie , Génotype , Humains , Mâle , Protéines de transport membranaire/génétique , Protéines du complexe d'import des protéines précurseurs mitochondriales , Polymorphisme de nucléotide simple , Analyse en composantes principales , Analyse de séquence d'ADN
20.
Brain Pathol ; 31(3): e12945, 2021 05.
Article de Anglais | MEDLINE | ID: mdl-33709463

RÉSUMÉ

TMEM106B has been recently implicated in multiple neurodegenerative diseases. Here, Rademakers et al. report a late-onset cerebellar Purkinje cell loss and progressive decline in motor function and gait deficits in a conventional Tmem106b-/- mouse model. By using high-power microscopy and bulk RNA sequencing, the authors further identify lysosomal and immune dysfunction as potential underlying mechanisms of the Purkinje cell loss.


Sujet(s)
Cellules de Purkinje , Animaux , Modèles animaux de maladie humaine , Souris
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...