20 results on '"Kanekiyo, Takahisa"'
Search Results
2. Relationships between PET and blood plasma biomarkers in corticobasal syndrome.
- Author
-
Singh, Neha Atulkumar, Alnobani, Alla, Graff‐Radford, Jonathan, Machulda, Mary M., Mielke, Michelle M., Schwarz, Christopher G., Senjem, Matthew L., Jack, Clifford R., Lowe, Val J., Kanekiyo, Takahisa, Josephs, Keith A., and Whitwell, Jennifer L.
- Abstract
INTRODUCTION: Corticobasal syndrome (CBS) can result from underlying Alzheimer's disease (AD) pathologies. Little is known about the utility of blood plasma metrics to predict positron emission tomography (PET) biomarker‐confirmed AD in CBS. METHODS: A cohort of eighteen CBS patients (8 amyloid beta [Aβ]+; 10 Aβ−) and 8 cognitively unimpaired (CU) individuals underwent PET imaging and plasma analysis. Plasma concentrations were compared using a Kruskal–Wallis test. Spearman correlations assessed relationships between plasma concentrations and PET uptake. RESULTS: CBS Aβ+ group showed a reduced Aβ42/40 ratio, with elevated phosphorylated tau (p‐tau)181, glial fibrillary acidic protein (GFAP), and neurofilament light (NfL) concentrations, while CBS Aβ− group only showed elevated NfL concentration compared to CU. Both p‐tau181 and GFAP were able to differentiate CBS Aβ− from CBS Aβ+ and showed positive associations with Aβ and tau PET uptake. DISCUSSION: This study supports use of plasma p‐tau181 and GFAP to detect AD in CBS. NfL shows potential as a non‐specific disease biomarker of CBS regardless of underlying pathology. Highlights: Plasma phosphorylated tau (p‐tau)181 and glial fibrillary acidic protein (GFAP) concentrations differentiate corticobasal syndrome (CBS) amyloid beta (Aβ)− from CBS Aβ+.Plasma neurofilament light concentrations are elevated in CBS Aβ− and Aβ+ compared to controls.Plasma p‐tau181 and GFAP concentrations were associated with Aβ and tau positron emission tomography (PET) uptake.Aβ42/40 ratio showed a negative correlation with Aβ PET uptake. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Cross‐species systems biology target prioritization leads to the development of antisense oligonucleotides as a potential therapeutic approach for progressive supranuclear palsy.
- Author
-
Min, Yuhao, Wang, Xue, Bergman, Jeremiah, Sotelo, Katie D, Reddy, Joseph S., Quicksall, Zachary, Nguyen, Thuy, Mitchell, Adriana O, Dickson, Dennis W., Kawatani, Keiji, Inoue, Yasuteru, Kanekiyo, Takahisa, Allen, Mariet, Is, Ozkan, and Ertekin‐Taner, Nilüfer
- Abstract
Background: Progressive supranuclear palsy (PSP) is a devastating primary tauopathy with rapid progression to death. Although several therapies currently in the development pipeline show promising safety profiles and robust target engagement, few demonstrated significant efficacy in patients, underscoring the need to interrogate additional targets with novel therapeutic modalities to expand the potential therapeutic arsenal. To diversify the therapeutic avenues for PSP and related tauopathies (e.g. Alzheimer's disease), we systematically integrated multi‐omics data from human brains of PSP and control donors with cross‐species validation to nominate high‐confidence therapeutic targets. We plan to translate our findings into safe and effective treatments for PSP using antisense oligonucleotides (ASO). Method: We analyzed brain gene expression profiles in PSP and control individuals at bulk tissue (N = 408) and single‐cell levels (snRNAseq, N = 36). Cell‐type‐specific expression perturbations were systematically prioritized using a cross‐species validation paradigm, including rTG4510 tau mice and tau‐overexpressing Drosophila models. We conducted in vitro screening of ASO candidates against our prioritized target genes. ASO efficacy and toxicity were measured. RNAseq experiments will be performed to interrogate off‐target effects. The safety and efficacy of the lead ASOs will be assessed in PSP‐patient‐derived iPSC models. Result: We previously reported discovery of novel genes with significant differential expression in PSP brains, characterized their brain cell‐specificity and validated them using snRNAseq data. We prioritized 21 genes using gene expression data from a mouse tauopathy model. Validation of these 21 high‐priority genes using a Drosophila tau model nominated astrocytic STOM, KANK2, and DDR2 as potential therapeutic targets for PSP. Knocking down their expression in Drosophila significantly rescues tau‐mediated neurodegenerative pathology. In vitro screening identified ASO leads that reduced the target expressions at mRNA and protein levels with low cellular toxicity. Conclusion: We developed a systems biology pipeline and nominated STOM, KANK2, and DDR2 as candidate gene targets for PSP. We identified ASOs that modulate these targets' expression without cellular toxicity, suggesting they may be suitable as potential therapeutic candidates. Importantly, the shared pathophysiology and molecular aberrations between PSP and other tauopathies, such as Alzheimer's disease, suggests that such therapeutic candidates may be repurposed for multiple neurodegenerative diseases, further accelerating, and streamlining the therapeutic pipeline. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. ApoE in brain mitochondria and energetics: From mice to human.
- Author
-
Borkowski, Kamil, Liang, Nuanyi, Arnold, Matthias, Bu, Guojun, Kaddurah‐Daouk, Rima, Zhao, Na, and Kanekiyo, Takahisa
- Abstract
Background: Alzheimer's disease (AD) risk and progression are significantly influenced by ApoE genotypes, with ApoE4 increasing and ApoE2 decreasing the susceptibility compared to ApoE3. Understanding metabolic pathways affected by ApoE genotypes will help decipher disease development and identify new therapeutic targets. Method: This study investigates the impact of ApoE genotypes on aging brain metabolic trajectories using human ApoE‐targeted replacement mice. Applying Biocrates P180 targeted metabolomics platform, we analyzed the metabolic impact of ApoE2/2, ApoE3/3, and ApoE4/4 on fatty acid β‐oxidation, amino acids, and phospholipids, which are known to be altered in AD. Furthermore, we compared our rodent model results with human dorsolateral prefrontal cortex data from the Religious Orders Study/Memory and Aging Project (ROS‐MAP). Result: We found aging mice carrying ApoE2/2 had altered branch‐chain amino acid metabolism and increased C5 acylcarnitine and its ratio to precursor isoleucine, pointing towards increased β‐oxidation and branched‐chain amino acid (BCAA) utilization. Furthermore, ROS‐MAP data revealed the ApoE2 genotype affects similar areas of metabolism in humans. Additionally, our data provide comprehensive insight into age‐related metabolic changes of the current mice model, independent of ApoE genotype, among phospholipids, sphingomyelins, amino acids, and biogenic amines. Many of the observed differences are also known aging markers in humans, with a connection to cognition and Alzheimer's disease. Conclusion: Together, these results suggest a potential involvement of ApoE2/2 genotype in energy metabolism and characterize the current mice model for further study of ApoE in AD, brain aging, and brain BCAA utilization for energy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. The P301L tau mutation alters extracellular vesicle biogenesis in astrocyte and contributes to astrocyte‐mediated tau pathology in a human iPSC‐derived model of tauopathies.
- Author
-
You, Yang, Mann, Sean D, Lu, Wenyan, Poon, Wayne W., Ikezu, Seiko, Kanekiyo, Takahisa, Dickson, Dennis W., Sławek, Jarosaw, Wszolek, Zbigniew, and Ikezu, Tsuneya
- Abstract
Background: Tauopathies are a group of neurodegenerative disorders which are characterized by the accumulation of abnormal tau protein in the brain. However, the mechanistic understanding of pathogenic tau formation and spread within the brain remains elusive. Astrocytes are major immune reactive cells in the brain and have been implicated in exacerbating tau pathology by releasing extracellular vesicles (AEVs) containing pro‐inflammatory cytokines and chemokines upon activation. Our prior investigation revealed a significant association between AEVs and tau pathology development, as well as cognitive function, by analyzing brain‐derived EV proteins from AD patients. In this study, we explore the potential roles of AEVs in tau pathogenesis using a human induced pluripotent stem cell (iPSC) model. Method: We obtained two male P301L tau mutant iPSC lines from a Polish family with frontotemporal dementia. By including two male control lines, these iPSCs were differentiated into astrocytes (iAs) and characterized by immunocytochemistry and subjected for bulk RNAseq. Bioinformatics analysis was conducted to compare the transcriptome profile between wild‐type (WT) and P301L iAs. EVs from WT and P301L iAs were isolated by ultracentrifugation combined with size exclusion chromatography. Characterization of WT and P301L iAEVs involved nanoparticle tracking analysis, nano‐flow cytometry and super‐resolution microscopy. Result: We successfully differentiated WT and P301L mutant iPSC lines into astrocytes with >99% purity. P301L iAs displayed distinctive astrocyte reactivity compared to WT cells, with elevated levels of pan‐reactive astrocyte genes (e.g., GFAP, CD44) and decreased expression of neuroprotective A2 astrocyte‐specific genes (e.g., TM4SF1, PTGS2). Additionally, gene enrichment set analysis of RNAseq data revealed dysregulation in the endo‐lysosomal pathway and extracellular matrix in P301L iAs compared to WT iAs. The count of intraluminal vesicles marked by CD9+ were reduced in P301L iAs compared to WT cells. Moreover, we observed a significant increase in the internalization of Tau by P301L iAs compared to WT iAs following incubation with preformed Tau fibrils, resulting in an augmented release of tau‐containing EVs from P301L iAs. Conclusion: Our findings suggest a potential alteration in EV biogenesis in P301L iAs, potentially contributing to astrocyte‐mediated tau pathology. Future investigations will focus on understanding how AEVs contribute to tau propagation and accumulation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Integrative multi‐omics epigenome‐wide association study identifies dysregulated DNA methylation near oligodendrocyte genes that associate with tau levels in the AD brain.
- Author
-
Oatman, Stephanie R, Reddy, Joseph S., Wang, Xue, Quicksall, Zachary, Atashgaran, Amin, Vanelderen, Floor, Bergman, Jeremiah, Is, Ozkan, Carrasquillo, Minerva M., Liu, Chia‐Chen, Yamazaki, Yu, Nguyen, Thuy, Heckman, Michael G., Zhao, Na, DeTure, Michael, Murray, Melissa E., Bu, Guojun, Kanekiyo, Takahisa, Dickson, Dennis W., and Allen, Mariet
- Abstract
Background: Alzheimer's disease (AD) is neuropathologically characterized by amyloid‐β (Aβ) plaques and tau neurofibrillary tangles often quantified by Thal phase and Braak stage, respectively. Aβ also frequently deposits in the cerebrovasculature with severity categorized by a cerebral amyloid angiopathy (CAA) score. These and related measures often show high variability within AD suggesting distinct underlying mechanisms. We hypothesize that, within the AD brain, neuropathology and levels of core AD‐related proteins are influenced by variations in DNA methylation (DNAm). To test this, we performed epigenome‐wide association studies (EWAS) using DNAm measures from the temporal cortex (TCX) and cerebellum (CER) with AD‐related neuropathologic measures (Braak, Thal, CAA) and brain biochemical levels of five proteins (apoE, Aβ40, Aβ42, tau, p‐tau). Methods: DNAm from neuropathologically‐confirmed AD cases was measured by reduced representation bisulfite sequencing (RRBS) from 471 TCX samples, 200 of which also had CER RRBS. TCX levels of five AD‐related proteins from three tissue fractions (buffer‐, detergent‐, and in‐soluble) were measured previously by ELISA (Liu 2020). CpG methylation (CpGm) was binned by a 15‐state chromatin model (Kundaje 2015) and averaged into CpGm clusters (rCpGm). rCpGms were tested for association with each AD endophenotype and expression levels of nearby genes through multi‐variable linear regression. Replication of significant rCpGms was performed in two independent datasets with AD‐related endophenotypes (Shireby 2022, De Jager 2014). Results: Our innovative binning method demonstrated biologically relevant CpGm patterns. We found epigenome‐wide significant associations primarily with tau‐related endophenotypes including 93 that had significant and concordant associations in the replication datasets. These rCpGms also significantly associated with expression of nearby genes showing enrichment in oligodendrocyte marker genes including those related to myelination. In vitro validation of tau effects on oligodendrocyte gene expression through DNAm is ongoing. Conclusions: Although all endophenotypes tested are core to AD pathophysiology, our results suggest each has a distinct epigenetic architecture underlying their variability in the AD brain. In particular, we found evidence of DNAm variability associating with brain oligodendrocyte gene expression and TCX tau levels. By discovering AD brain endophenotype‐specific DNAm changes, we can identify core components of complex mechanisms revealing important biological insights into AD pathophysiology. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Brain‐targeting liposome‐based APOE2 gene delivery exacerbates soluble amyloid‐β accumulation in AppNL‐G‐F mice.
- Author
-
Wang, Ni, Parsons, Tammee M, Ren, Yingxue, Pan, Yining, Starling, Skylar C, Muolokwu, Chinenye, Singh, Jagdish, and Kanekiyo, Takahisa
- Abstract
Background: Alzheimer's disease (AD) is a progressive neurodegenerative disease and the most prevalent form of late‐life dementia. The ε2 allele of the APOE gene encoding apolipoprotein E (APOE2) is associated with lower susceptibility to AD among the three genotypes (ε2, ε3, ε4), while APOE4 is the strongest genetic risk factor for late‐onset AD. APOE plays a critical role in maintaining synaptic plasticity and neuronal function by controlling lipid homeostasis, with APOE2 having a superior function. Gene therapy that increases APOE2 levels in the brain has, therefore, emerged as a potential therapeutic strategy to treat AD. Method: We conjugated PEGylated liposomes with transferrin and Penetratin, a cell‐penetrating peptide, sufficiently deliver chitosan‐APOE2 cDNA plasmid complex into amyloid model AppNL‐G‐F knockin mice at 12‐month‐old. Biochemical studies and brain transcriptome analysis were employed to investigate how brain‐targeting liposome‐based APOE2 gene delivery influences amyloid‐β (Aβ)‐related pathologies in AppNL‐G‐F knockin mice. Result: One month after APOE2 gene therapy, there was a trend of reduced insoluble Aβ levels in the mouse cortices. Furthermore, in the AppNL‐G‐F knockin mice that received the APOE2 gene therapy, brain transcriptome analysis through RNA‐sequencing identified the upregulation of genes/pathways related to neuronal development. This was supported by increases of Dlg4 and Syp mRNAs coding synaptic proteins in the experimental group. On the other hand, we found that APOE2 gene delivery increased soluble Aβ levels, including oligomers, as well as exacerbated neurite dystrophy and decreased synaptophysin. Conclusion: Our results suggest that brain‐targeting liposome‐based APOE2 gene therapy is potentially beneficial for synaptic formation at the transcriptional level. Forced APOE2 expressions, however, may exacerbate Aβ toxicity by increasing the dissociation of Aβ oligomers from aggregates in the presence of considerable amyloid burden. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. Gliovascular molecular alterations in Alzheimer's disease: a cross‐tissue, cross‐species study.
- Author
-
Is, Ozkan, Wang, Xue, Reddy, Joseph S., Patel, Tulsi, Min, Yuhao, Quicksall, Zachary, Heckman, Michael G., Gao, Junli, Bergman, Jeremiah, Da Mesquita, Sandro Gabriel Ferreira, Kizil, Caghan, Bhattarai, Prabesh, Cosacak, Mehmet I., Lee, Annie J, Vardarajan, Badri N, Mayeux, Richard, Koga, Shunsuke, Kanekiyo, Takahisa, White, Launia J, and Kouri, Naomi
- Abstract
Background: Inter‐cellular communication within the gliovascular unit (GVU) is critical for cerebral blood flow regulation, and maintenance of the blood‐brain‐barrier (BBB) properties. The breakdown of BBB in Alzheimer's disease (AD) is well‐established, but precise underlying molecular changes remain unclear. Additionally, whether GVU molecular alterations observed in AD brains are also detected in blood from living patients is unknown. Further, these GVU molecular perturbations require further investigation in different model systems to identify both human brain‐specific and cross‐species conserved alterations. In this study, we investigated prioritized GVU molecules altered in AD brains for their conservation in blood and cross‐species model systems. Methods: We performed single nucleus RNA sequencing (snRNAseq) of temporal cortex tissue in AD and control brains. We analyzed this data to detect cell‐specific GVU molecular perturbations and their interactions. We investigated molecular interactions between vascular and astrocyte clusters, the major cell types of the GVU of the BBB. To determine whether GVU transcriptional alterations detected in the brain are preserved in the blood, existing blood expression, genetic, and neuroimaging data from two longitudinal antemortem cohorts were analyzed. Using model systems, including mouse, drosophila, and zebrafish, we evaluated the cross‐species conservation of the top GVU alterations detected in AD brains. Results: Brain snRNAseq revealed transcriptional profiles of 6,541 astrocytes and 2,210 vascular cells. The latter formed three distinct vascular clusters characterized as pericytes, endothelia and perivascular fibroblasts. We identified differentially expressed genes and their enriched pathways within these clusters and observed the highest levels of transcriptional changes within pericytes. Vascular targets that interact with astrocytic ligands have biological functions in cell signaling, angiogenesis, amyloid ß metabolism, and cytoskeletal architecture. We discovered that genetic variants influencing blood expression levels of some of the prioritized GVU genes were associated with neuroimaging burden of cerebrovascular disease in living human cohorts. Our ongoing studies in model systems revealed conservation of some of the top prioritized molecular perturbations across species. Conclusion: Our findings prioritized by multiscale, cross‐tissue human data revealed GVU perturbations within interacting pericyte and astrocyte molecules, which are conserved across multiple cross‐species models. These results nominate new molecular targets and mechanistic insights for BBB disruptions in AD. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
9. DNA methylation clustered by predicted chromatin state reveals distinct association patterns with neuropathologic and biochemical measures in the AD brain.
- Author
-
Oatman, Stephanie R, Quicksall, Zachary, Reddy, Joseph S., Carrasquillo, Minerva M., Wang, Xue, Liu, Chia‐Chen, Yamazaki, Yu, Nguyen, Thuy, Malphrus, Kimberly G., Heckman, Michael G., Martens, Yuka A, Zhao, Na, DeTure, Michael, Murray, Melissa E., Kanekiyo, Takahisa, Bu, Guojun, Dickson, Dennis W., Allen, Mariet, and Ertekin‐Taner, Nilufer
- Abstract
Background: Alzheimer's disease (AD) is neuropathologically characterized by amyloid‐β (Aβ) plaques and tau neurofibrillary tangles often quantified by Thal phase and Braak stage, respectively. Aβ also frequently deposits in the cerebrovasculature with severity categorized by a cerebral amyloid angiopathy (CAA) score. These and related measures often show high variability within AD suggesting distinct underlying mechanisms, the identification of which may reveal important biological insights into AD pathophysiology. We hypothesize that, within the AD brain, neuropathology and levels of core AD‐related proteins are influenced by variations in DNA methylation (DNAm). To test this, we performed epigenome‐wide association studies (EWAS) using DNAm clusters from the temporal cortex (TCX) and cerebellum (CER) with AD‐related neuropathologic measures (Braak, Thal, and CAA) and brain biochemical levels of five proteins (apoE, Aβ40, Aβ42, tau, and p‐tau). Methods: DNAm from neuropathologically‐confirmed AD cases available through the Mayo Clinic brain bank was measured by reduced representation bisulfite sequencing (RRBS) from 471 brain samples, 200 of which had both TCX and CER measures. Neuropathology was scored by an experienced neuropathologist. TCX levels of five AD‐related proteins from three tissue fractions (buffer‐, detergent‐, and in‐soluble) were measured by ELISA (Liu 2020). CpG methylation (CpGm) was binned by a 15‐state chromatin model (Kundaje 2015) and averaged. Averaged CpGm clusters were tested for their association with each endophenotype through multi‐variable linear regression. The influence of CpGm clusters on expression levels of nearby genes was also investigated through multi‐variable regression. Results: Our innovative binning method demonstrated expected and biologically relevant CpGm patterns. We identified multiple epigenome‐wide significant CpGm clusters unique to each AD‐related endophenotype and brain region. Comparisons between the TCX and CER further revealed brain region‐specific enrichment of distinct CpGm clusters. Significant CpGm clusters were further characterized by their influence on gene expression. Conclusions: Although all neuropathologic and biochemical endophenotypes tested are core to AD pathophysiology, our results suggest each has a distinct epigenetic architecture underlying their variability in the AD brain. By discovering brain region‐specific DNAm changes that contribute to each neuropathologic or biochemical outcome, we can identify core components of complex mechanisms that individually and jointly contribute to AD. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
10. Transcriptome profiling in AD cases to identify distinct associations with AD‐related neuropathology and protein levels in the brain.
- Author
-
Allen, Mariet, Quicksall, Zachary, Wang, Xue, Reddy, Joseph S., Bergman, Jeremiah, Oatman, Stephanie R, Nguyen, Thuy, Malphrus, Kimberly G., Lincoln, Sarah J., Yamazaki, Yu, Martens, Yuka A, Zhao, Na, DeTure, Michael, Murray, Melissa E., Liu, Chia‐Chen, Kanekiyo, Takahisa, Dickson, Dennis W., Bu, Guojun, and Ertekin‐Taner, Nilufer
- Abstract
Background: Gene expression changes have been identified in post‐mortem brain tissue of AD cases when compared with controls, implicating various genes and pathways. Such findings provide important insights into molecular dysregulation associated with this disease and nominate therapeutic targets. Here we investigate brain gene and co‐expression network changes in a cohort of AD cases to identify associations with degree of AD neuropathology including Braak stage (Tau), Thal phase (Amyloid beta) and brain tissue levels of five AD‐related proteins (apoE, Aβ40, Aβ42, tau, and p‐tau). Method: Gene expression measures were collected from temporal cortex (TCX) tissue of 477 AD cases, available through the Mayo Clinic Brain Bank, using RNA sequencing (Illumina TruSeq mRNA) performed on the HiSeq4000. Following quality control 456 cases remained for analysis. Braak and Thal were provided by an experienced neuropathologist and TCX levels of five AD‐related proteins from three fractions (buffer‐soluble, detergent‐soluble, and insoluble) were measured by ELISA (Liu et al. 2020). Weighted gene coexpression network analysis (WGCNA) R package was used to build co‐expression networks. Genes and networks were tested for association with neuropathology and AD‐related protein levels, adjusting for relevant covariates. Result: A total of 57,353 genes were profiled, of which 19,044 were reliably expressed above background levels and formed 20 co‐expression modules. 36 genes were associated with Braak stage, 10 with Thal phase and up to 9,000 with the AD‐related protein fractions. Brain levels of pTau had the greatest number of associations, with an overrepresentation of genes involved in synaptic signalling and immune response gene ontology biological processes. 16 of the coexpression modules associated with at least one trait after Bonferroni correction for number of tests. Conclusion: We have identified a number of genes and networks associated with different aspects of AD neuropathology and associated proteins, further lending support to the involvement of synaptic and immune biology in AD. However it is not clear whether observed changes cause disease or are a consequence of the neurodegenerative process. Comparison of these findings across different proteins and fractions is expected to reveal common and distinct vulnerabilities associated with amyloid and tau, and different stages of protein aggregation. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
11. Pharmacological inhibition of sFRP1 stimulates the non‐amyloidogenic pathway of amyloid precursor protein proteolytic processing.
- Author
-
Lu, Wenyan, Macyczko, Jesse R, Martens, Yuka A, Bu, Guojun, Kanekiyo, Takahisa, and Li, Yonghe
- Abstract
Background: The accumulation of the amyloid‐β (Aβ) aggregates in the brain is a key pathological event in the development of Alzheimer's disease (AD). The α‐secretase ADAM10 is responsible for cleavage of the amyloid precursor protein (APP) through a non‐amyloidogenic pathway, thereby preventing the generation of pathogenic Aβ in the brain. Secreted frizzled related protein‐1 (sFRP1), an endogenous antagonist of Wnt signaling, has recently been identified as a key negative modulator of ADAM10. Importantly, proteomic studies reveal that unregulated sFRP1 is one of the top Aβ‐correlated proteins in the human AD brains. Methods: We determined the therapeutic potential of specific sFRP1 WAY316606 in the N2a/APP695swe cells and patient‐specific iPSC‐derived cerebral organoids. Results: We herein demonstrated that treatment of sFRP1 inhibitor WAY316606 resulted in augmentation of ADAM10 activity, upregulation of soluble APP‐α (sAPPα) production, and inhibition of Aβ generation in the N2a/APP695swe cells. Moreover, WAY316606 significantly enhanced ADAM10 activity, decreased Aβ production and inhibited tau phosphorylation in human AD patient‐specific iPSC‐derived cerebral organoids. Conclusion: Our findings indicate that sFRP1 is a promising therapeutic target for AD. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
12. Multiple system atrophy and apolipoprotein E.
- Author
-
Ogaki, Kotaro, Martens, Yuka A., Heckman, Michael G., Koga, Shunsuke, Labbé, Catherine, Lorenzo‐Betancor, Oswaldo, Wernick, Anna I., Walton, Ronald L., Soto, Alexandra I., Vargas, Emily R., Nielsen, Henrietta M., Fujioka, Shinsuke, Kanekiyo, Takahisa, Uitti, Ryan J., van Gerpen, Jay A., Cheshire, William P., Wszolek, Zbigniew K., Low, Phillip A., Singer, Wolfgang, and Dickson, Dennis W.
- Abstract
Background: Dysregulation of the specialized lipid metabolism involved in myelin synthesis and maintenance by oligodendrocytes has been associated with the unique neuropathology of MSA. We hypothesized that apolipoprotein E, which is associated with neurodegeneration, may also play a role in the pathogenesis of MSA.Objective: This study evaluated genetic associations of Apolipoprotein E alleles with risk of MSA and α-synuclein pathology, and also examined whether apolipoprotein E isoforms differentially affect α-synuclein uptake in a oligodendrocyte cell.Methods: One hundred sixty-eight pathologically confirmed MSA patients, 89 clinically diagnosed MSA patients, and 1,277 control subjects were genotyped for Apolipoprotein E. Human oligodendrocyte cell lines were incubated with α-synuclein and recombinant human apolipoprotein E, with internalized α-synuclein imaged by confocal microscopy and cells analyzed by flow cytometry.Results: No significant association with risk of MSA or was observed for either Apolipoprotein E ɛ2 or ɛ4. α-Synuclein burden was also not associated with Apolipoprotein E alleles in the pathologically confirmed patients. Interestingly, in our cell assays, apolipoprotein E ɛ4 significantly reduced α-synuclein uptake in the oligodendrocytic cell line.Conclusions: Despite differential effects of apolipoprotein E isoforms on α-synuclein uptake in a human oligodendrocytic cell, we did not observe a significant association at the Apolipoprotein E locus with risk of MSA or α-synuclein pathology. © 2018 International Parkinson and Movement Disorder Society. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
13. Association of DNA methylation from the temporal cortex and cerebellum with AD‐related neuropathology and biochemistry.
- Author
-
Oatman, Stephanie R, Allen, Mariet, Quicksall, Zachary, Reddy, Joseph S., Carrasquillo, Minerva M., Wang, Xue, Liu, Chia‐Chen, Yamazaki, Yu, Nguyen, Thuy, Heckman, Michael G., Martens, Yuka A, Zhao, Na, DeTure, Michael, Murray, Melissa E., Kanekiyo, Takahisa, Dickson, Dennis W., Bu, Guojun, and Ertekin‐Taner, Nilufer
- Abstract
Background: Alzheimer's disease (AD) is a progressive neurodegenerative disorder neuropathologically characterized by amyloid‐β (Aβ) plaques and tau neurofibrillary tangles often measured by Thal phase and Braak stage, respectively. Frequently, Aβ also deposits in the brain vasculature with severity categorized by a cerebral amyloid angiopathy (CAA) score. Assessment of these and related endophenotypes can reveal biological insights into the phenotypic variability in AD. We hypothesize that, amongst AD patients, severity of neuropathology and brain levels of AD‐related proteins are influenced by variation in DNA methylation (DNAm), the identification of which may uncover biological pathways for AD pathogenesis. To test this, we performed epigenome‐wide association studies (EWAS) using DNAm from the temporal cortex (TCX) and cerebellum (CER) with AD‐related neuropathology measures (Braak, Thal, and CAA) and brain levels of five proteins (apoE, Aβ40, Aβ42, tau, and p‐tau). Methods: DNAm from autopsy‐confirmed AD cases available through the Mayo Clinic Brain Bank was measured by reduced representation bisulfite sequencing (RRBS) from 471 brain samples, 200 of which had both TCX and CER measures. Bisulfite converted reads were sequenced on the Illumina High Seq4000 and quality filtered. Neuropathology scoring was performed by an experienced neuropathologist. TCX levels of five AD‐related proteins from three fractions (buffer‐soluble, detergent‐soluble, and insoluble) were measured by ELISA (Liu et al. 2020). Individual CpG methylation ratios were tested for association with each neuropathological and biochemical measure using the R package CpGassoc. Results: We identified multiple epigenome‐wide significant CpGs unique to each AD‐related measure. Comparisons between the TCX and CER revealed region specific CpG associations. The effects of sex, age at death, APOE genotype, and presence of genetic variants for each significant CpG association were also identified. Conclusions: Although all neuropathological and biochemical measures tested reflect core AD pathologies, our results demonstrate unique associations of epigenetic factors with these individual phenotypes, suggesting that their variability may be governed by distinct epigenetic processes. Discovery of DNAm changes that underlie specific neuropathological or biochemical outcomes can enhance our understanding of AD pathophysiology and may lead to the identification of novel gene or pathway targets for the different facets of this complex condition. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
14. APOE2 eases cognitive decline during Aging: Clinical and preclinical evaluations.
- Author
-
Shinohara, Mitsuru, Kanekiyo, Takahisa, Yang, Longyu, Linthicum, Duane, Shinohara, Motoko, Fu, Yuan, Price, Laura, Frisch ‐ Daiello, Jessica L., Han, Xianlin, Fryer, John D., Bu, Guojun, and Frisch-Daiello, Jessica L
- Abstract
Objective: Apolipoprotein E (apoE), a major cholesterol carrier in the brain, is associated with a strong risk for Alzheimer disease. Compared to the risky APOE4 gene allele, the effects of the protective APOE2 gene allele are vastly understudied, and thus need to be further clarified.Methods: We reviewed National Alzheimer's Coordinating Center clinical records and performed preclinical experiments using human apoE-targeted replacement (apoE-TR) mice, which do not show amyloid pathology.Results: Clinically, the APOE2 allele was associated with less cognitive decline during aging. This effect was also seen in subjects with little amyloid pathology, or after adjusting for Alzheimer disease-related pathologies. In animal studies, aged apoE2-TR mice also exhibited preserved memory function in water maze tests. Regardless, apoE2-TR mice showed similar or greater age-related changes in synaptic loss, neuroinflammation, and oxidative stress compared to apoE3-TR or apoE4-TR mice. Interestingly, apoE concentrations in the cortex, hippocampus, plasma, and cerebrospinal fluid (CSF) were positively correlated with memory performance across apoE isoforms, where apoE2-TR mice had higher apoE levels. Moreover, apoE2-TR mice exhibited the lowest levels of cholesterol in the cortex, despite higher levels in CSF and plasma. These cholesterol levels were associated with apoE levels and memory performance across apoE isoforms.Interpretation: APOE2 is associated with less cognitive decline during aging. This can occur independently of age-related synaptic/neuroinflammatory changes and amyloid accumulation. Higher levels of apoE and associated cholesterol metabolism in APOE2 carriers might contribute to this protective effect. Ann Neurol 2016;79:758-774. [ABSTRACT FROM AUTHOR]- Published
- 2016
- Full Text
- View/download PDF
15. Genome‐wide analysis identifies novel genetic variants and unique biological pathways associated with AD‐related proteins in the brain.
- Author
-
Oatman, Stephanie R, Reddy, Joseph S., Quicksall, Zachary, Carrasquillo, Minerva M., Wang, Xue, Liu, Chia‐Chen, Yamazaki, Yu, Nguyen, Thuy, Malphrus, Kimberly G., Heckman, Michael G., Martens, Yuka A, Zhao, Na, DeTure, Michael, Murray, Melissa E, Kanekiyo, Takahisa, Dickson, Dennis W, Bu, Guojun, Allen, Mariet, and Ertekin‐Taner, Nilufer
- Abstract
Background: Alzheimer's disease (AD) is a progressive neurodegenerative disorder neuropathologically characterized by amyloid‐β (Aβ) plaques and neurofibrillary tangles. Assessment of AD‐related endophenotypes can reveal novel insights into aspects of the heterogeneity of AD. We hypothesize that, amongst AD patients, the levels and solubility of AD‐related proteins are influenced by genetic variants; identifying these may provide key insights into disease pathogenesis. To test this, we performed a genome‐wide association study (GWAS) for the levels of five AD‐related proteins (apoE, Aβ40, Aβ42, tau, and p‐tau) in the brain. Methods: Genome‐wide genotypes were collected from 441 autopsy‐confirmed AD cases on the Infinium Omni2.5Exome‐8 v1.3 genotyping array, imputed to the haplotype reference consortium (HRC) panel, and quality filtered. Temporal cortex levels of the five AD‐related proteins from three fractions, i.e. buffer‐soluble (TBS), detergent‐soluble (Triton‐X=TX), and insoluble (Formic acid=FA) were measured by ELISA. Using linear regression, genetic variants were tested for association with each biochemical measure, and the Aβ40/42 ratio. Pathway analysis was done using GSA‐SNP2 to identify enriched Gene Ontology (GO) terms. Results: We identified 9 unique GWS variants (genome‐wide significant, p<2.89E‐08), 2 at the APOE locus and 7 novel. Variants within the APOE locus were significantly associated with Aβ40 levels in the TX and FA fractions, the Aβ40/42 ratio in the FA fraction, and apoE levels in the TBS and FA fractions. Of the novel GWS variants, four were associated with Aβ40 residing on chromosomes 17 (TBS), 3, 4, and 15 (TX), and three with apoE levels on chromosomes 19 (TBS), 7, and 14 (TX). Pathway analysis revealed both shared and distinct pathways significantly enriched for the GWAS results. Conclusions: Although all biochemical measures tested reflect proteins core to AD pathology, our results strongly suggest each have unique genetic factors and enriched pathways which influence their brain levels. Furthermore, discovery of genetic factors that underlie specific biochemical outcomes may lead to the identification of novel genes that influence biochemical properties of AD proteins. These findings can enhance our understanding of the pathophysiology of proteostasis in AD and may have implications for other neurodegenerative diseases also characterized by abnormal protein accumulation in the brain. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
16. Cell type‐specific gene expression changes in Alzheimer's patients with varying levels of cerebral amyloid angiopathy.
- Author
-
Patel, Tulsi, Udine, Evan, Wang, Xue, Allen, Mariet, Is, Ozkan, Tutor‐New, Frederick Q, Carnwath, Troy, Deniz, Kaancan, Lincoln, Sarah J., Nguyen, Thuy, Malphrus, Kimberly G., Quicksall, Zachary, Lewis‐Tuffin, Laura, Reddy, Joseph S., Carrasquillo, Minerva M., Crook, Julia E., Kanekiyo, Takahisa, Murray, Melissa E, Bu, Guojun, and Dickson, Dennis W
- Abstract
Background: Tau neurofibrillary tangles and senile plaques comprised of insoluble amyloid beta are the major histopathological hallmarks of Alzheimer's disease (AD). More than 85% of autopsy‐confirmed AD cases also exhibit some degree of cerebral amyloid angiopathy (CAA), which is characterized by amyloid beta peptide deposits predominantly in blood vessels in the meningeal and intracerebral blood vessels. Consequently, CAA predisposes individuals with AD to cerebral infarction and hemorrhages, accounting for ∼20% of cases in the elderly, which could lead to faster cognitive decline through neurovascular pathway dysfunction. We sought to identify cellular composition changes and characterize cell type‐specific expression associated with CAA pathology in AD patients. Methods: Using 10x Genomics single nuclei RNA sequencing, we profiled nuclei from frozen post‐mortem temporal cortex tissue of 80 individuals with neuropathologic diagnosis of AD and varying levels of CAA, ranging from zero to severe CAA pathology. Braak and Thal measures were also obtained. Downstream analyses were performed in R using Seurat v3.2.3. Quality control filtering based on percentage of genes mapped to the mitochondrial genome and number of genes and UMIs detected resulted in 135,092 nuclei for analysis. Differential gene expression analyses were performed using R package MAST. Results: We identified 30 clusters which were annotated using established cell type marker genes. There were 3 oligodendrocyte, 14 neuronal (2 excitatory and 3 inhibitory), 5 microglial, 3 endothelial, 4 astrocyte and 1 oligodendrocyte precursor cell cluster. We observed a significant increase in microglial cell proportions with increasing CAA severity, for 3 of the microglial clusters. Conversely, the excitatory neuronal proportion were decreased in individuals with more severe CAA. Further investigation is ongoing to identify the genes underlying the dysregulation of these cell types in CAA. Conclusion: We observed cellular proportion changes for multiple cell types with increasing CAA severity. Additional analyses to determine the effects of CAA independent of AD severity will be performed by adjusting for Braak and Thal measures. Deeper characterization of cell type‐specific expression profiles is expected to provide novel insights into cell‐type specific transcriptome changes associated with vascular risk factors in AD. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
17. Lipocalin-type prostaglandin D synthase is up-regulated in oligodendrocytes in lysosomal storage diseases and binds gangliosides.
- Author
-
Mohri, Ikuko, Taniike, Masako, Okazaki, Issei, Kagitani-Shimono, Kuriko, Aritake, Kosuke, Kanekiyo, Takahisa, Yagi, Takashi, Takikita, Shoichi, Hyung-Suk Kim, Urade, Yoshihiro, and Suzuki, Kinuko
- Subjects
GANGLIOSIDES ,GLYCOSPHINGOLIPIDS ,OLIGODENDROGLIA ,PROSTAGLANDINS ,LYSOSOMES ,ENZYMES - Abstract
Lipocalin-type prostaglandin (PG) D synthase (L-PGDS) is a dually functional protein, acting both as a PGD
2 -synthesizing enzyme and as an extracellular transporter of various lipophilic small molecules. L-PGDS is expressed in oligodendrocytes (OLs) in the central nervous system and is up-regulated in OLs of the twitcher mouse, a model of globoid cell leukodystrophy (Krabbe's disease). We investigated whether up-regulation of L-PGDS is either unique to Krabbe's disease or is a more generalized phenomenon in lysosomal storage disorders (LSDs), using LSD mouse models of Tay–Sachs disease, Sandhoff disease, GM1 gangliosidosis and Niemann–Pick type C1 disease. Quantitative RT-PCR revealed that L-PGDS mRNA was up-regulated in the brains of all these mouse models. In addition, strong L-PGDS immunoreactivity was observed in OLs, but not in either astrocytes or microglia in these models. Thus, up-regulation of L-PGDS appears to be a common response of OLs in LSDs. Moreover, surface plasmon resonance analyses revealed that L-PGDS binds GM1 and GM2 gangliosides, accumulated in neurons in the course of LSD, with high affinities ( KD = 65 and 210 nm, respectively). This suggests that L-PGDS may play a role in scavenging harmful lipophilic substrates in LSD. [ABSTRACT FROM AUTHOR]- Published
- 2006
- Full Text
- View/download PDF
18. P4‐229: LIPOSOMAL NANOPARTICLES FOR GENE DELIVERY TO THE BRAIN.
- Author
-
Rodrigues, Bruna, Oue, Hiroshi, Kanekiyo, Takahisa, and Singh, Jagdish
- Published
- 2018
- Full Text
- View/download PDF
19. P4‐065: APOE4 CONDITIONALLY EXPRESSED IN CEREBROVASCULATURE IMPAIRS ENDOTHELIAL FUNCTIONS AND INDUCES COGNITIVE DEFICITS.
- Author
-
Yamazaki, Yu, Liu, Chia-Chen, Oue, Hiroshi, Kurti, Aishe, Yamazaki, Akari, Fryer, John D., Kanekiyo, Takahisa, and Bu, Guojun
- Published
- 2018
- Full Text
- View/download PDF
20. P3‐088: LIPOSOME‐MEDIATED NGF GENE TRANSFECTION AND POTENTIAL THERAPEUTIC APPLICATION.
- Author
-
Rodrigues, Bruna, Kanekiyo, Takahisa, and Singh, Jagdish
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.