33 results on '"Yuka A Martens"'
Search Results
2. APOE4 exacerbates α-synuclein seeding activity and contributes to neurotoxicity in Alzheimer’s disease with Lewy body pathology
- Author
-
Yunjung Jin, Fuyao Li, Berkiye Sonoustoun, Naveen Chandra Kondru, Yuka A. Martens, Wenhui Qiao, Michael G. Heckman, Tadafumi C. Ikezu, Zonghua Li, Jeremy D. Burgess, Danilyn Amerna, Justin O’Leary, Michael A. DeTure, Jing Zhao, Pamela J. McLean, Dennis W. Dickson, Owen A. Ross, Guojun Bu, and Na Zhao
- Subjects
Cellular and Molecular Neuroscience ,nervous system ,Neurology (clinical) ,Pathology and Forensic Medicine - Abstract
Approximately half of Alzheimer’s disease (AD) brains have concomitant Lewy pathology at autopsy, suggesting that α-synuclein (α-SYN) aggregation is a regulated event in the pathogenesis of AD. Genome-wide association studies revealed that the ε4 allele of the apolipoprotein E (APOE4) gene, the strongest genetic risk factor for AD, is also the most replicated genetic risk factor for Lewy body dementia (LBD), signifying an important role of APOE4 in both amyloid-β (Aβ) and α-SYN pathogenesis. How APOE4 modulates α-SYN aggregation in AD is unclear. In this study, we aimed to determine how α-SYN is associated with AD-related pathology and how APOE4 impacts α-SYN seeding and toxicity. We measured α-SYN levels and their association with other established AD-related markers in brain samples from autopsy-confirmed AD patients (N = 469), where 54% had concomitant LB pathology (AD + LB). We found significant correlations between the levels of α-SYN and those of Aβ40, Aβ42, tau and APOE, particularly in insoluble fractions of AD + LB. Using a real-time quaking-induced conversion (RT-QuIC) assay, we measured the seeding activity of soluble α-SYN and found that α-SYN seeding was exacerbated by APOE4 in the AD cohort, as well as a small cohort of autopsy-confirmed LBD brains with minimal Alzheimer type pathology. We further fractionated the soluble AD brain lysates by size exclusion chromatography (SEC) ran on fast protein liquid chromatography (FPLC) and identified the α-SYN species (~ 96 kDa) that showed the strongest seeding activity. Finally, using human induced pluripotent stem cell (iPSC)-derived neurons, we showed that amplified α-SYN aggregates from AD + LB brain of patients with APOE4 were highly toxic to neurons, whereas the same amount of α-SYN monomer was not toxic. Our findings suggest that the presence of LB pathology correlates with AD-related pathologies and that APOE4 exacerbates α-SYN seeding activity and neurotoxicity, providing mechanistic insight into how APOE4 affects α-SYN pathogenesis in AD.
- Published
- 2022
- Full Text
- View/download PDF
3. ApoE Cascade Hypothesis in the pathogenesis of Alzheimer’s disease and related dementias
- Author
-
Yuka A. Martens, Na Zhao, Chia-Chen Liu, Takahisa Kanekiyo, Austin J. Yang, Alison M. Goate, David M. Holtzman, and Guojun Bu
- Subjects
Apolipoproteins E ,Alzheimer Disease ,Apolipoprotein E2 ,General Neuroscience ,Apolipoprotein E4 ,Humans ,Protein Isoforms ,Amino Acids ,Article - Abstract
The ε4 allele of the apolipoprotein E gene (APOE4) is a strong genetic risk factor for Alzheimer's disease (AD) and several other neurodegenerative conditions, including Lewy body dementia (LBD). The three APOE alleles encode protein isoforms that differ from one another only at amino acid positions 112 and 158: apoE2 (C112, C158), apoE3 (C112, R158), and apoE4 (R112, R158). Despite progress, it remains unclear how these small amino acid differences in apoE sequence among the three isoforms lead to profound effects on aging and disease-related pathways. Here, we propose a novel "ApoE Cascade Hypothesis" in AD and age-related cognitive decline, which states that the biochemical and biophysical properties of apoE impact a cascade of events at the cellular and systems levels, ultimately impacting aging-related pathogenic conditions including AD. As such, apoE-targeted therapeutic interventions are predicted to be more effective by addressing the biochemical phase of the cascade.
- Published
- 2022
- Full Text
- View/download PDF
4. Trem2 H157Y increases soluble TREM2 production and reduces amyloid pathology
- Author
-
Wenhui Qiao, Yixing Chen, Jun Zhong, Benjamin J. Madden, Cristine M. Charlesworth, Yuka A. Martens, Chia-Chen Liu, Joshua Knight, Tadafumi C. Ikezu, Aishe Kurti, Yiyang Zhu, Axel Meneses, Cassandra L. Rosenberg, Lindsey A. Kuchenbecker, Lucy K. Vanmaele, Fuyao Li, Kai Chen, Francis Shue, Maxwell V. Dacquel, John Fryer, Akhilesh Pandey, Na Zhao, and Guojun Bu
- Subjects
Cellular and Molecular Neuroscience ,Neurology (clinical) ,Molecular Biology - Abstract
Background The rare p.H157Y variant of TREM2 (Triggering Receptor Expressed on Myeloid Cells 2) was found to increase Alzheimer’s disease (AD) risk. This mutation is located at the cleavage site of TREM2 extracellular domain. Ectopic expression of TREM2-H157Y in HEK293 cells resulted in increased TREM2 shedding. However, the physiological outcomes of the TREM2 H157Y mutation remain unknown in the absence and presence of AD related pathologies. Methods We generated a novel Trem2 H157Y knock-in mouse model through CRISPR/Cas9 technology and investigated the effects of Trem2 H157Y on TREM2 proteolytic processing, synaptic function, and AD-related amyloid pathologies by conducting biochemical assays, targeted mass spectrometry analysis of TREM2, hippocampal electrophysiology, immunofluorescent staining, in vivo micro-dialysis, and cortical bulk RNA sequencing. Results Consistent with previous in vitro findings, Trem2 H157Y increases TREM2 shedding with elevated soluble TREM2 levels in the brain and serum. Moreover, Trem2 H157Y enhances synaptic plasticity without affecting microglial density and morphology, or TREM2 signaling. In the presence of amyloid pathology, Trem2 H157Y accelerates amyloid-β (Aβ) clearance and reduces amyloid burden, dystrophic neurites, and gliosis in two independent founder lines. Targeted mass spectrometry analysis of TREM2 revealed higher ratios of soluble to full-length TREM2-H157Y compared to wild-type TREM2, indicating that the H157Y mutation promotes TREM2 shedding in the presence of Aβ. TREM2 signaling was further found reduced in Trem2 H157Y homozygous mice. Transcriptomic profiling revealed that Trem2 H157Y downregulates neuroinflammation-related genes and an immune module correlated with the amyloid pathology. Conclusion Taken together, our findings suggest beneficial effects of the Trem2 H157Y mutation in synaptic function and in mitigating amyloid pathology. Considering the genetic association of TREM2 p.H157Y with AD risk, we speculate TREM2 H157Y in humans might increase AD risk through an amyloid-independent pathway, such as its effects on tauopathy and neurodegeneration which merit further investigation.
- Published
- 2023
- Full Text
- View/download PDF
5. Novel Wnt modulators for the treatment of Alzheimer’s disease
- Author
-
Yonghe Li, Thomas R. Caulfield, Jesse R Macyczko, Na Wang, Lin Jia, Francis Shue, Wenyan Lu, Jing Zhao, Yuka A Martens, Chia‐Chen Liu, and Guojun Bu
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2022
- Full Text
- View/download PDF
6. Association of DNA methylation from the temporal cortex and cerebellum with AD‐related neuropathology and biochemistry
- Author
-
Stephanie R Oatman, Mariet Allen, Zachary Quicksall, Joseph S. Reddy, Minerva M. Carrasquillo, Xue Wang, Chia‐Chen Liu, Yu Yamazaki, Thuy Nguyen, Michael G. Heckman, Yuka A Martens, Na Zhao, Michael DeTure, Melissa E. Murray, Takahisa Kanekiyo, Dennis W. Dickson, Guojun Bu, and Nilufer Ertekin‐Taner
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2022
- Full Text
- View/download PDF
7. Determining Etiologic Diagnoses in Patients with Rapidly Progressive Dementia
- Author
-
Gregory S Day, Philip W Tipton, Evelyn Lazar, Yuka A Martens, S Richard Dunham, Michael D Geschwind, Guojun Bu, John C. Morris, and Neill R. Graff‐Radford
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2022
- Full Text
- View/download PDF
8. Elevating microglia TREM2 reduces amyloid seeding and suppresses disease-associated microglia
- Author
-
Na Zhao, Wenhui Qiao, Fuyao Li, Yingxue Ren, Jiaying Zheng, Yuka A. Martens, Xusheng Wang, Ling Li, Chia-Chen Liu, Kai Chen, Yiyang Zhu, Tadafumi C. Ikezu, Zonghua Li, Axel D. Meneses, Yunjung Jin, Joshua A. Knight, Yixing Chen, Ligia Bastea, Cynthia Linares, Berkiye Sonustun, Lucy Job, Madeleine L. Smith, Manling Xie, Yong U. Liu, Anthony D. Umpierre, Koichiro Haruwaka, Zachary S. Quicksall, Peter Storz, Yan W. Asmann, Long-Jun Wu, and Guojun Bu
- Subjects
Amyloid ,Mice ,Membrane Glycoproteins ,Alzheimer Disease ,Immunology ,Animals ,Brain ,Humans ,Immunology and Allergy ,Amyloidosis ,Microglia ,Receptors, Immunologic - Abstract
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.
- Published
- 2022
- Full Text
- View/download PDF
9. LRP1 is a neuronal receptor for α-synuclein uptake and spread
- Author
-
Kai Chen, Yuka A. Martens, Axel Meneses, Daniel H. Ryu, Wenyan Lu, Ana Caroline Raulin, Fuyao Li, Jing Zhao, Yixing Chen, Yunjung Jin, Cynthia Linares, Marshall Goodwin, Yonghe Li, Chia-Chen Liu, Takahisa Kanekiyo, David M. Holtzman, Todd E. Golde, Guojun Bu, and Na Zhao
- Subjects
Mice ,Cellular and Molecular Neuroscience ,Induced Pluripotent Stem Cells ,alpha-Synuclein ,Animals ,Humans ,Infant ,Parkinson Disease ,tau Proteins ,Neurology (clinical) ,Synapsins ,Molecular Biology ,Low Density Lipoprotein Receptor-Related Protein-1 - Abstract
Background The aggregation and spread of α-synuclein (α-Syn) protein and related neuronal toxicity are the key pathological features of Parkinson’s disease (PD) and Lewy body dementia (LBD). Studies have shown that pathological species of α-Syn and tau can spread in a prion-like manner between neurons, although these two proteins have distinct pathological roles and contribute to different neurodegenerative diseases. It is reported that the low-density lipoprotein receptor-related protein 1 (LRP1) regulates the spread of tau proteins; however, the molecular regulatory mechanisms of α-Syn uptake and spread, and whether it is also regulated by LRP1, remain poorly understood. Methods We established LRP1 knockout (LRP1-KO) human induced pluripotent stem cells (iPSCs) isogenic lines using a CRISPR/Cas9 strategy and generated iPSC-derived neurons (iPSNs) to test the role of LRP1 in α-Syn uptake. We treated the iPSNs with fluorescently labeled α-Syn protein and measured the internalization of α-Syn using flow cytometry. Three forms of α-Syn species were tested: monomers, oligomers, and pre-formed fibrils (PFFs). To examine whether the lysine residues of α-Syn are involved in LRP1-mediated uptake, we capped the amines of lysines on α-Syn with sulfo-NHS acetate and then measured the internalization. We also tested whether the N-terminus of α-Syn is critical for LRP1-mediated internalization. Lastly, we investigated the role of Lrp1 in regulating α-Syn spread with a neuronal Lrp1 conditional knockout (Lrp1-nKO) mouse model. We generated adeno-associated viruses (AAVs) that allowed for distinguishing the α-Syn expression versus spread and injected them into the hippocampus of six-month-old Lrp1-nKO mice and the littermate wild type (WT) controls. The spread of α-Syn was evaluated three months after the injection. Results We found that the uptake of both monomeric and oligomeric α-Syn was significantly reduced in iPSNs with LRP1-KO compared with the WT controls. The uptake of α-Syn PFFs was also inhibited in LRP1-KO iPSNs, albeit to a much lesser extent compared to α-Syn monomers and oligomers. The blocking of lysine residues on α-Syn effectively decreased the uptake of α-Syn in iPSNs and the N-terminus of α-Syn was critical for LRP1-mediated α-Syn uptake. Finally, in the Lrp1-nKO mice, the spread of α-Syn was significantly reduced compared with the WT littermates. Conclusions We identified LRP1 as a key regulator of α-Syn neuronal uptake, as well as an important mediator of α-Syn spread in the brain. This study provides new knowledge on the physiological and pathological role of LRP1 in α-Syn trafficking and pathology, offering insight for the treatment of synucleinopathies.
- Published
- 2022
- Full Text
- View/download PDF
10. Genome-wide association study of brain biochemical phenotypes reveals distinct genetic architecture of Alzheimer's disease related proteins
- Author
-
Stephanie R, Oatman, Joseph S, Reddy, Zachary, Quicksall, Minerva M, Carrasquillo, Xue, Wang, Chia-Chen, Liu, Yu, Yamazaki, Thuy T, Nguyen, Kimberly, Malphrus, Michael, Heckman, Kristi, Biswas, Kwangsik, Nho, Matthew, Baker, Yuka A, Martens, Na, Zhao, Jun Pyo, Kim, Shannon L, Risacher, Rosa, Rademakers, Andrew J, Saykin, Michael, DeTure, Melissa E, Murray, Takahisa, Kanekiyo, Dennis W, Dickson, Guojun, Bu, Mariet, Allen, and Nilüfer, Ertekin-Taner
- Subjects
Cellular and Molecular Neuroscience ,Neurology (clinical) ,Human medicine ,Molecular Biology - Abstract
Background Alzheimer’s disease (AD) is neuropathologically characterized by amyloid-beta (Aβ) plaques and neurofibrillary tangles. The main protein components of these hallmarks include Aβ40, Aβ42, tau, phosphor-tau, and APOE. We hypothesize that genetic variants influence the levels and solubility of these AD-related proteins in the brain; identifying these may provide key insights into disease pathogenesis. Methods Genome-wide genotypes were collected from 441 AD cases, imputed to the haplotype reference consortium (HRC) panel, and filtered for quality and frequency. Temporal cortex levels of five AD-related proteins from three fractions, buffer-soluble (TBS), detergent-soluble (Triton-X = TX), and insoluble (Formic acid = FA), were available for these same individuals. Variants were tested for association with each quantitative biochemical measure using linear regression, and GSA-SNP2 was used to identify enriched Gene Ontology (GO) terms. Implicated variants and genes were further assessed for association with other relevant variables. Results We identified genome-wide significant associations at seven novel loci and the APOE locus. Genes and variants at these loci also associate with multiple AD-related measures, regulate gene expression, have cell-type specific enrichment, and roles in brain health and other neuropsychiatric diseases. Pathway analysis identified significant enrichment of shared and distinct biological pathways. Conclusions Although all biochemical measures tested reflect proteins core to AD pathology, our results strongly suggest that each have unique genetic architecture and biological pathways that influence their specific biochemical states in the brain. Our novel approach of deep brain biochemical endophenotype GWAS has implications for pathophysiology of proteostasis in AD that can guide therapeutic discovery efforts focused on these proteins.
- Published
- 2022
11. Correction to: Apolipoprotein E regulates lipid metabolism and α‑synuclein pathology in human iPSC‑derived cerebral organoids
- Author
-
Mary D. Davis, Yan W. Asmann, Guojun Bu, Yuan Fu, Nilufer Ertekin-Taner, Zbigniew K. Wszolek, Jing Zhao, Meixia Pan, Kai Chen, Chia Chen Liu, Neill R. Graff-Radford, Yingxue Ren, Fuyao Li, Dennis W. Dickson, Steven G. Younkin, Takahisa Kanekiyo, Xianlin Han, Francis Shue, Yuka A. Martens, Wenyan Lu, Ziying Xu, David A. Brafman, and Xue Wang
- Subjects
Apolipoprotein E ,Cellular and Molecular Neuroscience ,Chemistry ,Organoid ,Lipid metabolism ,α synuclein ,Neurology (clinical) ,Pathology and Forensic Medicine ,Cell biology - Published
- 2021
- Full Text
- View/download PDF
12. APOE deficiency impacts neural differentiation and cholesterol biosynthesis in human iPSC-derived cerebral organoids
- Author
-
Jing Zhao, Tadafumi C. Ikezu, Wenyan Lu, Jesse R. Macyczko, Yonghe Li, Laura J. Lewis-Tuffin, Yuka A. Martens, Yingxue Ren, Yiyang Zhu, Yan W. Asmann, Nilüfer Ertekin-Taner, Takahisa Kanekiyo, and Guojun Bu
- Abstract
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. Here, 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 EIF2 signaling pathway as these events were alleviated by the treatment of a pathway inhibitor ISRIB. APOE deletion also leads to activation of the Wnt/β-catenin signaling pathway with concomitant decrease of 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. Our work demonstrates critical roles of apoE in brain homeostasis and offers critical insights into the APOE4-related pathogenic mechanisms.
- Published
- 2022
- Full Text
- View/download PDF
13. Genome-wide association study of brain biochemical phenotypes reveals distinct genetic architecture of Alzheimer’s Disease related proteins
- Author
-
Stephanie R. Oatman, Joseph S. Reddy, Zachary Quicksall, Minerva M. Carrasquillo, Xue Wang, Chia-Chen Liu, Yu Yamazaki, Thuy T. Nguyen, Kimberly Malphrus, Michael Heckman, Kristi Biswas, Matthew Baker, Yuka A. Martens, Na Zhao, Rosa Rademakers, Michael DeTure, Melissa E. Murray, Takahisa Kanekiyo, Dennis W. Dickson, Guojun Bu, Mariet Allen, and Nilüfer Ertekin-Taner
- Abstract
Alzheimer’s disease (AD) is neuropathologically characterized by amyloid-beta (Aβ) plaques and neurofibrillary tangles. Main protein components of these hallmarks include Aβ40, Aβ42, tau, phospho-tau and APOE. With the exception of the APOE-ε4 variant, genetic risk factors associated with brain biochemical measures of these proteins have yet to be characterized. We performed a genome-wide association study in brains of 441 AD patients for quantitative levels of these proteins collected from three distinct fractions reflecting soluble, membrane-bound and insoluble biochemical states. We identified 123 genome-wide significant associations at seven novel loci and the APOE locus. Genes and variants at these loci also associate with multiple AD- related measures, regulate gene expression, have cell-type specific enrichment, and roles in brain health and other neuropsychiatric diseases. Pathway analysis identified significant enrichment of shared and distinct biological pathways. Although all biochemical measures tested reflect proteins core to AD pathology, our results strongly suggest that each have unique genetic architecture and biological pathways that influence their specific biochemical states in the brain. Our novel approach of deep brain biochemical endophenotype GWAS has implications for pathophysiology of proteostasis in AD that can guide therapeutic discovery efforts focused on these proteins.
- Published
- 2022
- Full Text
- View/download PDF
14. Opposing effects of apoE2 and apoE4 on microglial activation and lipid metabolism in response to demyelination
- Author
-
Na Wang, Minghui Wang, Suren Jeevaratnam, Cassandra Rosenberg, Tadafumi C. Ikezu, Francis Shue, Sydney V. Doss, Alla Alnobani, Yuka A. Martens, Melissa Wren, Yan W. Asmann, Bin Zhang, Guojun Bu, and Chia-Chen Liu
- Subjects
Cellular and Molecular Neuroscience ,Mice ,Cuprizone ,Apolipoproteins E ,Apolipoprotein E2 ,Apolipoprotein E4 ,Apolipoprotein E3 ,Animals ,Neurology (clinical) ,Microglia ,Lipid Metabolism ,Molecular Biology ,Demyelinating Diseases - Abstract
Background Abnormal lipid accumulation has been recognized as a key element of immune dysregulation in microglia whose dysfunction contributes to neurodegenerative diseases. Microglia play essential roles in the clearance of lipid-rich cellular debris upon myelin damage or demyelination, a common pathogenic event in neuronal disorders. Apolipoprotein E (apoE) plays a pivotal role in brain lipid homeostasis; however, the apoE isoform-dependent mechanisms regulating microglial response upon demyelination remain unclear. Methods To determine how apoE isoforms impact microglial response to myelin damage, 2-month-old apoE2-, apoE3-, and apoE4-targeted replacement (TR) mice were fed with normal diet (CTL) or 0.2% cuprizone (CPZ) diet for four weeks to induce demyelination in the brain. To examine the effects on subsequent remyelination, the cuprizone diet was switched back to regular chow for an additional two weeks. After treatment, brains were collected and subjected to immunohistochemical and biochemical analyses to assess the myelination status, microglial responses, and their capacity for myelin debris clearance. Bulk RNA sequencing was performed on the corpus callosum (CC) to address the molecular mechanisms underpinning apoE-mediated microglial activation upon demyelination. Results We demonstrate dramatic isoform-dependent differences in the activation and function of microglia upon cuprizone-induced demyelination. ApoE2 microglia were hyperactive and more efficient in clearing lipid-rich myelin debris, whereas apoE4 microglia displayed a less activated phenotype with reduced clearance efficiency, compared with apoE3 microglia. Transcriptomic profiling revealed that key molecules known to modulate microglial functions had differential expression patterns in an apoE isoform-dependent manner. Importantly, apoE4 microglia had excessive buildup of lipid droplets, consistent with an impairment in lipid metabolism, whereas apoE2 microglia displayed a superior ability to metabolize myelin enriched lipids. Further, apoE2-TR mice had a greater extent of remyelination; whereas remyelination was compromised in apoE4-TR mice. Conclusions Our findings provide critical mechanistic insights into how apoE isoforms differentially regulate microglial function and the maintenance of myelin dynamics, which may inform novel therapeutic avenues for targeting microglial dysfunctions in neurodegenerative diseases.
- Published
- 2022
15. APOE4 exacerbates α-synuclein seeding activity and contributes to neurotoxicity in Alzheimer's disease with Lewy body pathology
- Author
-
Yunjung, Jin, Fuyao, Li, Berkiye, Sonoustoun, Naveen Chandra, Kondru, Yuka A, Martens, Wenhui, Qiao, Michael G, Heckman, Tadafumi C, Ikezu, Zonghua, Li, Jeremy D, Burgess, Danilyn, Amerna, Justin, O'Leary, Michael A, DeTure, Jing, Zhao, Pamela J, McLean, Dennis W, Dickson, Owen A, Ross, Guojun, Bu, and Na, Zhao
- Subjects
Lewy Body Disease ,Apolipoproteins E ,Alzheimer Disease ,Apolipoprotein E4 ,Induced Pluripotent Stem Cells ,alpha-Synuclein ,Humans ,Lewy Bodies ,Neurotoxicity Syndromes ,tau Proteins ,Genome-Wide Association Study - Abstract
Approximately half of Alzheimer's disease (AD) brains have concomitant Lewy pathology at autopsy, suggesting that α-synuclein (α-SYN) aggregation is a regulated event in the pathogenesis of AD. Genome-wide association studies revealed that the ε4 allele of the apolipoprotein E (APOE4) gene, the strongest genetic risk factor for AD, is also the most replicated genetic risk factor for Lewy body dementia (LBD), signifying an important role of APOE4 in both amyloid-β (Aβ) and α-SYN pathogenesis. How APOE4 modulates α-SYN aggregation in AD is unclear. In this study, we aimed to determine how α-SYN is associated with AD-related pathology and how APOE4 impacts α-SYN seeding and toxicity. We measured α-SYN levels and their association with other established AD-related markers in brain samples from autopsy-confirmed AD patients (N = 469), where 54% had concomitant LB pathology (AD + LB). We found significant correlations between the levels of α-SYN and those of Aβ40, Aβ42, tau and APOE, particularly in insoluble fractions of AD + LB. Using a real-time quaking-induced conversion (RT-QuIC) assay, we measured the seeding activity of soluble α-SYN and found that α-SYN seeding was exacerbated by APOE4 in the AD cohort, as well as a small cohort of autopsy-confirmed LBD brains with minimal Alzheimer type pathology. We further fractionated the soluble AD brain lysates by size exclusion chromatography (SEC) ran on fast protein liquid chromatography (FPLC) and identified the α-SYN species (~ 96 kDa) that showed the strongest seeding activity. Finally, using human induced pluripotent stem cell (iPSC)-derived neurons, we showed that amplified α-SYN aggregates from AD + LB brain of patients with APOE4 were highly toxic to neurons, whereas the same amount of α-SYN monomer was not toxic. Our findings suggest that the presence of LB pathology correlates with AD-related pathologies and that APOE4 exacerbates α-SYN seeding activity and neurotoxicity, providing mechanistic insight into how APOE4 affects α-SYN pathogenesis in AD.
- Published
- 2021
16. TREM2-H157Y Increases Soluble TREM2 Production and Reduces Amyloid Pathology
- Author
-
Wenhui Qiao, Kurti I. Aishe, Na Zhao, Francis Shue, Chia Chen Liu, Yuka A. Martens, John D. Fryer, Yixing Chen, Fuyao Li, Maxwell V. Dacquel, Kai Chen, Joshua Knight, and Guojun Bu
- Subjects
Mutation ,Chemistry ,TREM2 ,Synaptic plasticity ,Neurodegeneration ,Mutant ,medicine ,Wild type ,Tauopathy ,Microgliosis ,medicine.disease_cause ,medicine.disease ,Cell biology - Abstract
Background: The p.H157Y variant of TREM2 (Triggering Receptor Expressed on Myeloid Cells 2) has been reported to increase Alzheimer’s disease (AD) risk. This mutation in the extracellular domain of TREM2 localizes at the cleavage site and was shown to enhance shedding and impair phagocytosis in vitro by ectopic expression of TREM2-H157Y in HEK293 cells. However, the physiological and AD-related outcomes of TREM2 H157Y mutation in vivo remain unknown.Methods: We generated a novel Trem2 H157Y knock-in mouse model through CRISPR-Cas9 technology and investigated how Trem2 H157Y mutation impacts TREM2 proteolytic processing, synaptic function, and AD-related amyloid pathology by conducting biochemical assays, immunofluorescent staining, hippocampal electrophysiology, and in vivo micro-dialysis in awake, free-moving animals.Results: Consistent with previous in vitro findings, TREM2-H157Y increases the amount of soluble TREM2 (sTREM2) in the cortex and serum of mutant mice compared to the wild type controls. Interestingly, the Trem2 H157Y variant enhances synaptic plasticity without affecting microglial density and morphology. In the presence of amyloid pathology, TREM2-H157Y surprisingly accelerates Aβ clearance and reduces amyloid burden and microgliosis. Conclusion: Taken together, our findings support a beneficial effect of the Trem2 H157Y mutation in synaptic function and in mitigating amyloid pathology. Considering the genetic association of TREM2 p.H157Y with AD, we speculate TREM2-H157Y might increase AD risk through an amyloid-independent pathway, as such its effects on tauopathy and neurodegeneration merit further investigation.
- Published
- 2021
- Full Text
- View/download PDF
17. APOE3 -Jacksonville (V236E) variant reduces self-aggregation and risk of dementia
- Author
-
Hu Wang, Bradley F. Boeve, Michael G. Heckman, Yingxue Ren, Yan W. Asmann, Ana-Caroline Raulin, Yuan Fu, Thomas R. Caulfield, Tadafumi C. Ikezu, Tanis J. Ferman, Cynthia Linares, Lin Jia, Dennis W. Dickson, Carl Frieden, Jing Zhao, Owen A. Ross, Steven G. Younkin, Zbigniew K. Wszolek, Val J. Lowe, Cassandra L. Rosenberg, David S. Knopman, Xia Li, Yonghe Li, John D. Fryer, Xue Wang, Sydney V. Doss, Melissa E. Murray, Wenyan Lu, Clifford R. Jack, Melissa C. Wren, Xianlin Han, Joshua Knight, Zachary A. Trottier, Na Wang, Yuka A. Martens, Joseph E. Parisi, Yixing Chen, Guojun Bu, Aishe Kurti, Neill R. Graff-Radford, Kejal Kantarci, Chia Chen Liu, Glenn E. Smith, Ronald C. Petersen, Prashanthi Vemuri, Na Zhao, and Francis Shue
- Subjects
Apolipoprotein E ,business.industry ,Self aggregation ,Extramural ,Genetic variants ,General Medicine ,Disease ,medicine.disease ,Bioinformatics ,Apolipoproteins E ,Text mining ,Medicine ,Dementia ,business - Abstract
A rare APOE variant APOE3 -Jac reduces self-aggregation and promotes healthy brain aging.
- Published
- 2021
- Full Text
- View/download PDF
18. Clinicopathologic Factors Associated With Reversion to Normal Cognition in Patients With Mild Cognitive Impairment
- Author
-
Zonghua Li, Michael G Heckman, Takahisa Kanekiyo, Yuka A Martens, Gregory S Day, Maria Vassilaki, Chia-Chen Liu, David A. Bennett, Ronald C Petersen, Na Zhao, and Guojun Bu
- Subjects
Aging ,Amyloid ,Cognition ,Glucose ,Alzheimer Disease ,Disease Progression ,Humans ,Cognitive Dysfunction ,Neurology (clinical) ,Amyloidosis ,Neuropsychological Tests ,Research Article - Abstract
Objective:This study aims to identify clinicopathological factors contributing to mild cognitive impairment (MCI) reversion to normal cognition.Methods:We analyzed three longitudinal cohorts in this study, including Mayo Clinic Study of Aging (MCSA), the Religious Orders Study and Memory and Aging Project (ROSMAP), and the National Alzheimer’s Coordinating Center (NACC). The demographic characteristics and clinical outcomes were compared between MCI subjects with or without an experience of reversion to normal cognition (referred to as reverters and non-reverters, respectively). We also compared longitudinal changes in cortical thickness, glucose metabolism, amyloid and tau load in a sub-cohort of reverters and non-reverters in MCSA with MRI or PET imaging information from multiple visits.ResultsWe identified 164 (56.4%) individuals in MCSA, 508 (66.8%) individuals in ROSMAP, and 280 (34.1%) individuals in NACC who experienced MCI reversion to normal cognition. Cox proportional hazards regression models showed that MCI reverters had an increased chance of being cognitively normal at the last visit in MCSA (HR, 3.31; 95% CI, 2.14 to 5.12), ROSMAP (HR, 3.72; 95% CI, 2.50 to 5.56), and NACC (HR, 9.29; 95% CI, 6.45 to 13.40) and a reduced risk of progression to dementia (HR, 0.12; 95% CI, 0.05 to 0.29 in MCSA; HR, 0.41; 95% CI, 0.32 to 0.53 in ROSMAP and HR, 0.29; 95% CI, 0.21 to 0.40 in NACC). Compared to MCI non-reverters, reverters had better-preserved cortical thickness (β = 0.082, P < 0.001) and glucose metabolism (β = 0.119, P = 0.001) and lower levels of amyloid, albeit, statistically non-significant (β = -0.172, P = 0.090). However, no difference in tau load was found between reverters and non-reverters (β = 0.073, P = 0.24).Conclusion:MCI reversion to normal cognition is likely attributed to the better-preserved cortical structure and glucose metabolism.
- Published
- 2021
19. TREM2 interacts with TDP-43 and mediates microglial neuroprotection against TDP-43-related neurodegeneration
- Author
-
Manling Xie, Yong U. Liu, Shunyi Zhao, Lingxin Zhang, Dale B. Bosco, Yuan-Ping Pang, Jun Zhong, Udit Sheth, Yuka A. Martens, Na Zhao, Chia-Chen Liu, Yongxian Zhuang, Liewei Wang, Dennis W. Dickson, Mark P. Mattson, Guojun Bu, and Long-Jun Wu
- Subjects
DNA-Binding Proteins ,Mice ,Membrane Glycoproteins ,General Neuroscience ,Animals ,Mice, Transgenic ,Neurodegenerative Diseases ,Microglia ,Receptors, Immunologic ,Neuroprotection - Abstract
Triggering receptor expressed on myeloid cell 2 (TREM2) is a surface receptor that, in the central nervous system, is exclusively expressed on microglia. TREM2 variants have been linked to increased risk for neurodegenerative diseases, but the functional effects of microglial TREM2 remain largely unknown. To this end, we investigated TAR-DNA binding protein 43 kDa (TDP-43)-related neurodegenerative disease via viral-mediated expression of human TDP-43 protein (hTDP-43) in neonatal and adult mice or inducible expression of hTDP43 with defective nuclear localization signals in transgenic mice. We found that TREM2 deficiency impaired microglia phagocytic clearance of pathological TDP-43, and enhanced neuronal damage and motor function impairments. Mass cytometry analysis revealed that hTDP-43 induced a TREM2-dependent subpopulation of microglia with high CD11c expression and higher phagocytic ability. Using mass spectrometry and surface plasmon resonance analysis, we further demonstrated an interaction between TDP-43 and TREM2,in vitroandin vivo,in hTDP-43-expressing transgenic mouse brains. We computationally identified the region within hTDP-43 that interacts with TREM2 and observed the potential interaction in ALS patient tissues. Our data reveal the novel interaction between TREM2 and TDP-43, highlighting that TDP-43 is a possible ligand for microglial TREM2 and the interaction mediates neuroprotection of microglial TREM2 in TDP-43-related neurodegeneration.
- Published
- 2021
- Full Text
- View/download PDF
20. ABCA7 haplodeficiency disturbs microglial immune responses in the mouse brain
- Author
-
Yingxue Ren, Yan W. Asmann, Tomonori Aikawa, Takahisa Kanekiyo, Guojun Bu, Takaomi C. Saido, Marie Louise Holm, Yu Yamazaki, Casey T. Anderson, Michael L. Fitzgerald, Takashi Saito, Masaya Tachibana, Yuka A. Martens, and Madeleine R. Johnson
- Subjects
Lipopolysaccharide ,Mice, Transgenic ,Inflammation ,Haploinsufficiency ,ABCA7 ,Proinflammatory cytokine ,Pathogenesis ,Mice ,chemistry.chemical_compound ,Immune system ,Alzheimer Disease ,medicine ,Animals ,Amyloid beta-Peptides ,Multidisciplinary ,Microglia ,biology ,business.industry ,Gene Expression Profiling ,Brain ,Biological Sciences ,Immunity, Innate ,medicine.anatomical_structure ,chemistry ,Knockout mouse ,Immunology ,biology.protein ,ATP-Binding Cassette Transporters ,medicine.symptom ,Transcriptome ,business - Abstract
Carrying premature termination codons in 1 allele of the ABCA7 gene is associated with an increased risk for Alzheimer’s disease (AD). While the primary function of ABCA7 is to regulate the transport of phospholipids and cholesterol, ABCA7 is also involved in maintaining homeostasis of the immune system. Since inflammatory pathways causatively or consequently participate in AD pathogenesis, we studied the effects of Abca7 haplodeficiency in mice on brain immune responses under acute and chronic conditions. When acute inflammation was induced through peripheral lipopolysaccharide injection in control or heterozygous Abca7 knockout mice, partial ABCA7 deficiency diminished proinflammatory responses by impairing CD14 expression in the brain. On breeding to App NL-G-F knockin mice, we observed increased amyloid-β (Aβ) accumulation and abnormal endosomal morphology in microglia. Taken together, our results demonstrate that ABCA7 loss of function may contribute to AD pathogenesis by altering proper microglial responses to acute inflammatory challenges and during the development of amyloid pathology, providing insight into disease mechanisms and possible treatment strategies.
- Published
- 2019
- Full Text
- View/download PDF
21. APOE4-mediated amyloid-β pathology depends on its neuronal receptor LRP1
- Author
-
Simon Glerup, Yu Yamazaki, Melissa E. Murray, Yuka A. Martens, Marie Louise Holm, Takahisa Kanekiyo, Dennis W. Dickson, Mitsuru Shinohara, Kathrin Weyer, Patrick Sullivan, Masaya Tachibana, Tomonori Aikawa, Guojun Bu, Chia Chen Liu, and Hiroshi Oue
- Subjects
0301 basic medicine ,Apolipoprotein E ,Pathology ,medicine.medical_specialty ,CLEARANCE ,Amyloid ,Mice, Knockout, ApoE ,Apolipoprotein E4 ,Hippocampus ,APOLIPOPROTEIN-E ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Alzheimer Disease ,mental disorders ,Genotype ,ANGIOPATHY ,medicine ,Animals ,Humans ,BRAIN ,DEPOSITION ,Allele ,Receptor ,A-BETA ,RISK ,Amyloid beta-Peptides ,Chemistry ,General Medicine ,Metabolism ,LRP1 ,Peptide Fragments ,ALZHEIMERS-DISEASE ,Disease Models, Animal ,030104 developmental biology ,030220 oncology & carcinogenesis ,Commentary ,lipids (amino acids, peptides, and proteins) ,human activities ,APOE ,Low Density Lipoprotein Receptor-Related Protein-1 - Abstract
Carrying the ε4 allele of the APOE gene encoding apolipoprotein E (APOE4) markedly increases the risk for late-onset Alzheimer's disease (AD), in which APOE4 exacerbates the brain accumulation and subsequent deposition of amyloid-β (Aβ) peptides. While the LDL receptor-related protein 1 (LRP1) is a major apoE receptor in the brain, we found that its levels are associated with those of insoluble Aβ depending on APOE genotype status in postmortem AD brains. Thus, to determine the functional interaction of apoE4 and LRP1 in brain Aβ metabolism, we crossed neuronal LRP1-knockout mice with amyloid model APP/PS1 mice and APOE3-targeted replacement (APO3-TR) or APOE4-TR mice. Consistent with previous findings, mice expressing apoE4 had increased Aβ deposition and insoluble amounts of Aβ40 and Aβ42 in the hippocampus of APP/PS1 mice compared with those expressing apoE3. Intriguingly, such effects were reversed in the absence of neuronal LRP1. Neuronal LRP1 deficiency also increased detergent-soluble apoE4 levels, which may contribute to the inhibition of Aβ deposition. Together, our results suggest that apoE4 exacerbates Aβ pathology through a mechanism that depends on neuronal LRP1. A better understanding of apoE isoform-specific interaction with their metabolic receptor LRP1 on Aβ metabolism is crucial for defining APOE4-related risk for AD.
- Published
- 2019
- Full Text
- View/download PDF
22. Generation and validation of
- Author
-
Yuka A, Martens, Siming, Xu, Richard, Tait, Gary, Li, Xinping C, Zhao, Wenyan, Lu, Chia-Chen, Liu, Takahisa, Kanekiyo, Guojun, Bu, and Jing, Zhao
- Subjects
Organoids ,Gene Knockdown Techniques ,CRISPR ,Stem Cells ,Induced Pluripotent Stem Cells ,Protocol ,Brain ,Humans ,lipids (amino acids, peptides, and proteins) ,CRISPR-Cas Systems ,Neuroscience - Abstract
Summary Apolipoprotein E (apoE) is a major lipid carrier in the brain and closely associated with the pathogenesis of Alzheimer's disease (AD). Here, we describe a protocol for efficient knockout of APOE in human induced pluripotent stem cells (iPSCs) using the CRISPR-Cas9 system. We obtain homozygous APOE knockout (APOE-/-) iPSCs and further validate the deficiency of apoE in iPSC-derived cerebral organoids. APOE-/- cerebral organoids can serve as a useful tool to study apoE functions and apoE-related pathogenic mechanisms in AD. For complete details on the use and execution of this protocol, please refer to Zhao et al. (2020)., Graphical abstract, Highlights • Detailed protocol to generate APOE knockout human iPSC lines via CRISPR-Cas9 technology • Detailed protocol to generate cerebral organoids from parental and isogenic iPSC lines • Steps for quality control and assessment of apoE deletion in cerebral organoids • Provides a valuable tool for apoE function study using iPSC-derived cerebral organoids, Apolipoprotein E (apoE) is a major lipid carrier in the brain and closely associated with the pathogenesis of Alzheimer's disease (AD). Here, we describe a protocol for efficient knockout of APOE in human induced pluripotent stem cells (iPSCs) using the CRISPR-Cas9 system. We obtain homozygous APOE knockout (APOE-/-) iPSCs and further validate the deficiency of apoE in iPSC-derived cerebral organoids. APOE-/- cerebral organoids can serve as a useful tool to study apoE functions and apoE-related pathogenic mechanisms in AD.
- Published
- 2021
23. TREM2 interacts with TDP-43 and mediates microglial neuroprotection against TDP-43-related neurodegeneration
- Author
-
Yong Liu, Yuka A. Martens, Yongxian Zhuang, Dennis W. Dickson, Dale B. Bosco, Udit Sheth, Yuan Ping Pang, Chia Chen Liu, Manling Xie, Shuyi Zhao, Na Zhao, Liewei Wang, Lingxin Zhang, Long Jun Wu, Mark P. Mattson, and Guojun Bu
- Subjects
Genetically modified mouse ,medicine.anatomical_structure ,Microglia ,Chemistry ,TREM2 ,Neurodegeneration ,Central nervous system ,medicine ,medicine.disease ,Ligand (biochemistry) ,Receptor ,Neuroprotection ,Cell biology - Abstract
Triggering receptor expressed on myeloid cell 2 (TREM2) is a surface receptor that, in the central nervous system, is exclusively expressed on microglia. TREM2 variants have been linked to increased risk for neurodegenerative diseases, but the functional effects of microglial TREM2 remain largely unknown. To this end, we investigated TAR-DNA binding protein 43 kDa (TDP-43)-related neurodegenerative disease via viral-mediated expression of human TDP-43 protein (hTDP-43) in neonatal and adult mice or inducible expression of hTDP43 with defective nuclear localization signals in transgenic mice. We found that TREM2 deficiency impaired microglia phagocytic clearance of pathological TDP-43, and enhanced neuronal damage and motor function impairments. Mass cytometry analysis revealed that hTDP-43 induced a TREM2-dependent subpopulation of microglia with high CD11c expression and higher phagocytic ability. Using mass spectrometry and surface plasmon resonance analysis, we further demonstrated an interaction between TDP-43 and TREM2, in vitro and in vivo, in hTDP-43-expressing transgenic mouse brains. We computationally identified the region within hTDP-43 that interacts with TREM2 and observed the potential interaction in ALS patient tissues. Our data reveal the novel interaction between TREM2 and TDP-43, highlighting that TDP-43 is a possible ligand for microglial TREM2 and the interaction mediates neuroprotection of microglial TREM2 in TDP-43-related neurodegeneration.
- Published
- 2021
- Full Text
- View/download PDF
24. APOE4 exacerbates synapse loss and neurodegeneration in Alzheimer’s disease patient iPSC-derived cerebral organoids
- Author
-
Yan W. Asmann, Yuan Fu, Wenyan Lu, Francis Shue, Yesesri Cherukuri, Nilufer Ertekin-Taner, Yuka A. Martens, Chia Chen Liu, Jing Zhao, Guojun Bu, Lin Jia, Neill R. Graff-Radford, Zbigniew K. Wszolek, Yu Yamazaki, Takahisa Kanekiyo, Mary D. Davis, Lucy Job, Yingxue Ren, Steven G. Younkin, David A. Brafman, Thanh Thanh L. Nguyen, Kai Chen, and Xue Wang
- Subjects
0301 basic medicine ,Apolipoprotein E ,Science ,General Physics and Astronomy ,General Biochemistry, Genetics and Molecular Biology ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,mental disorders ,medicine ,Organoid ,Dementia ,lcsh:Science ,Induced pluripotent stem cell ,Multidisciplinary ,business.industry ,Neurodegeneration ,General Chemistry ,medicine.disease ,Phenotype ,030104 developmental biology ,lipids (amino acids, peptides, and proteins) ,lcsh:Q ,business ,human activities ,Neuroscience ,030217 neurology & neurosurgery ,Cerebral organoid - Abstract
APOE4 is the strongest genetic risk factor associated with late-onset Alzheimer’s disease (AD). To address the underlying mechanism, we develop cerebral organoid models using induced pluripotent stem cells (iPSCs) with APOE ε3/ε3 or ε4/ε4 genotype from individuals with either normal cognition or AD dementia. Cerebral organoids from AD patients carrying APOE ε4/ε4 show greater apoptosis and decreased synaptic integrity. While AD patient-derived cerebral organoids have increased levels of Aβ and phosphorylated tau compared to healthy subject-derived cerebral organoids, APOE4 exacerbates tau pathology in both healthy subject-derived and AD patient-derived organoids. Transcriptomics analysis by RNA-sequencing reveals that cerebral organoids from AD patients are associated with an enhancement of stress granules and disrupted RNA metabolism. Importantly, isogenic conversion of APOE4 to APOE3 attenuates the APOE4-related phenotypes in cerebral organoids from AD patients. Together, our study using human iPSC-organoids recapitulates APOE4-related phenotypes and suggests APOE4-related degenerative pathways contributing to AD pathogenesis.
- Published
- 2020
- Full Text
- View/download PDF
25. Multiple system atrophy and apolipoprotein E
- Author
-
Jay A. van Gerpen, Zbigniew K. Wszolek, Alexandra I. Soto, Oswaldo Lorenzo-Betancor, Shinsuke Fujioka, Owen A. Ross, Wolfgang Singer, Anna I. Wernick, Kotaro Ogaki, Phillip A. Low, Ronald L. Walton, Catherine Labbé, Yuka A. Martens, Shunsuke Koga, Takahisa Kanekiyo, Emily R. Vargas, Guojun Bu, Michael G. Heckman, Henrietta M. Nielsen, Ryan J. Uitti, William P. Cheshire, and Dennis W. Dickson
- Subjects
Male ,0301 basic medicine ,Apolipoprotein E ,medicine.medical_specialty ,Genotype ,Apolipoprotein B ,animal diseases ,Neuropathology ,Article ,Pathogenesis ,03 medical and health sciences ,Myelin ,Apolipoproteins E ,0302 clinical medicine ,Atrophy ,Internal medicine ,mental disorders ,medicine ,Humans ,Genetic Testing ,Aged ,Cell Line, Transformed ,biology ,Neurodegeneration ,Multiple System Atrophy ,medicine.disease ,Oligodendrocyte ,nervous system diseases ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,nervous system ,Neurology ,Astrocytes ,alpha-Synuclein ,biology.protein ,Female ,lipids (amino acids, peptides, and proteins) ,Neurology (clinical) ,030217 neurology & neurosurgery - 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.
- Published
- 2018
- Full Text
- View/download PDF
26. APOE ε4/ε4 diminishes neurotrophic function of human iPSC-derived astrocytes
- Author
-
Jing Zhao, Mary D. Davis, Neill R. Graff-Radford, Yuka A. Martens, Mitsuru Shinohara, Takahisa Kanekiyo, Guojun Bu, Steven G. Younkin, and Zbigniew K. Wszolek
- Subjects
0301 basic medicine ,Apolipoprotein E ,Genotype ,Apolipoprotein E4 ,Induced Pluripotent Stem Cells ,Central nervous system ,Apolipoprotein E3 ,Synaptogenesis ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,medicine ,Humans ,Cognitive decline ,Induced pluripotent stem cell ,Molecular Biology ,Alleles ,Cells, Cultured ,Genetics (clinical) ,Neurons ,Articles ,General Medicine ,Coculture Techniques ,Neural stem cell ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Astrocytes ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Cellular model ,030217 neurology & neurosurgery ,Neurotrophin - Abstract
The ε4 allele of the APOE gene encoding apolipoprotein E (apoE) is a strong genetic risk factor for aging-related cognitive decline as well as late-onset Alzheimer's disease (AD) compared to the common ε3 allele. In the central nervous system, apoE is produced primarily by astrocytes and functions in transporting lipids including cholesterol to support neuronal homeostasis and synaptic integrity. Although mouse models and corresponding primary cells have provided valuable tools for studying apoE isoform-dependent functions, recent studies have shown that human astrocytes have a distinct gene expression profile compare with rodent astrocytes. Human induced pluripotent stem cells (iPSCs) derived from individuals carrying specific gene variants or mutations provide an alternative cellular model more relevant to humans upon differentiation into specific cell types. Thus, we reprogramed human skin fibroblasts from cognitively normal individuals carrying APOE ε3/ε3 or ε4/ε4 genotype to iPSC clones and further differentiated them into neural progenitor cells and then astrocytes. We found that human iPSC-derived astrocytes secreted abundant apoE with apoE4 lipoprotein particles less lipidated compared to apoE3 particles. More importantly, human iPSC-derived astrocytes were capable of promoting neuronal survival and synaptogenesis when co-cultured with iPSC-derived neurons with APOE ε4/ε4 astrocytes less effective in supporting these neurotrophic functions than those with APOE ε3/ε3 genotype. Taken together, our findings demonstrate APOE genotype-dependent effects using human iPSC-derived astrocytes and provide novel evidence that the human iPSC-based model system is a strong tool to explore how apoE isoforms contribute to neurodegenerative diseases.
- Published
- 2017
- Full Text
- View/download PDF
27. Alzheimer's Risk Factors Age, APOE Genotype, and Sex Drive Distinct Molecular Pathways
- Author
-
Yingxue Ren, Zonghua Li, Alexandra Kueider-Paisley, Yu Yamazaki, Olivia N. Attrebi, Rima Kaddurah-Daouk, Fuyao Li, Yuka A. Martens, Takahisa Kanekiyo, Akari Yamazaki, Lucy Job, Nilufer Ertekin-Taner, Matthias Arnold, Ligia I. Bastea, Tomonori Aikawa, Siamak MahmoudianDehkordi, Kai Chen, Lisa St. John-Williams, Na Zhao, Francis Shue, Berkiye Sonoustoun, Axel D. Meneses, Guojun Bu, Wenhui Qiao, J. Will Thompson, Peter Storz, Lindsey M. Felton, Jiaying Zheng, Hiroshi Oue, Masaya Tachibana, Chia Chen Liu, and Yan W. Asmann
- Subjects
0301 basic medicine ,Apolipoprotein E ,Male ,Aging ,Genotype ,Apolipoprotein E2 ,Apolipoprotein E4 ,Apolipoprotein E3 ,Gene Expression ,Mice, Transgenic ,Disease ,Biology ,Article ,Transcriptome ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Apolipoproteins E ,Sex Factors ,Downregulation and upregulation ,Alzheimer Disease ,Risk Factors ,Metabolome ,Animals ,Humans ,Gene Regulatory Networks ,Receptors, Immunologic ,Gene ,Serpins ,Adaptor Proteins, Signal Transducing ,Genetics ,Membrane Glycoproteins ,TREM2 ,General Neuroscience ,Gene Expression Profiling ,Age Factors ,Brain ,Membrane Proteins ,Protective Factors ,030104 developmental biology ,Unfolded Protein Response ,Female ,030217 neurology & neurosurgery - Abstract
Evidence suggests interplay among the three major risk factors for Alzheimer's disease (AD): age, APOE genotype, and sex. Here, we present comprehensive datasets and analyses of brain transcriptomes and blood metabolomes from human apoE2-, apoE3-, and apoE4-targeted replacement mice across young, middle, and old ages with both sexes. We found that age had the greatest impact on brain transcriptomes highlighted by an immune module led by Trem2 and Tyrobp, whereas APOE4 was associated with upregulation of multiple Serpina3 genes. Importantly, these networks and gene expression changes were mostly conserved in human brains. Finally, we observed a significant interaction between age, APOE genotype, and sex on unfolded protein response pathway. In the periphery, APOE2 drove distinct blood metabolome profile highlighted by the upregulation of lipid metabolites. Our work identifies unique and interactive molecular pathways underlying AD risk factors providing valuable resources for discovery and validation research in model systems and humans.
- Published
- 2019
28. APOE4 exacerbates α-synuclein pathology and related toxicity independent of amyloid
- Author
-
Mary D. Davis, Chia Chen Liu, Joshua Knight, Aishe Kurti, Fuyao Li, Guojun Bu, Cynthia Linares, Wenhui Qiao, John D. Fryer, Olivia N. Attrebi, Yingxue Ren, Marion Delenclos, Jiaying Zheng, Yan W. Asmann, Dennis W. Dickson, Pamela J. McLean, Owen A. Ross, Na Zhao, Francis Shue, Axel D. Meneses, Berkiye Sonustun, Yuka A. Martens, Yixing Chen, and Alexandra J. Van Ingelgom
- Subjects
Apolipoprotein E ,Lewy Body Disease ,Pathology ,medicine.medical_specialty ,Synucleinopathies ,Mice, Knockout, ApoE ,Apolipoprotein E4 ,Disease ,Article ,Transcriptome ,Mice ,mental disorders ,medicine ,Dementia ,Animals ,Allele ,Amyloid beta-Peptides ,Lewy body ,business.industry ,Dementia with Lewy bodies ,General Medicine ,medicine.disease ,Astrogliosis ,nervous system diseases ,alpha-Synuclein ,lipids (amino acids, peptides, and proteins) ,business ,human activities - Abstract
The apolipoprotein E (APOE) ε4 allele is the strongest genetic risk factor for late-onset Alzheimer’s disease mainly by driving amyloid-β pathology. Recently, APOE4 has also been found to be a genetic risk factor for Lewy body dementia (LBD), which includes dementia with Lewy bodies (DLB) and Parkinson’s disease dementia (PDD). How APOE4 drives risk of LBD and whether it has a direct effect on α-synuclein pathology is not clear. Here we generated a mouse model of synucleinopathy using an adeno-associated virus (AAV) gene delivery of α-synuclein in human APOE-targeted replacement mice expressing APOE2, APOE3 or APOEE4. We found that APOE4, but not APOE2 or APOE3, increased α-synuclein pathology, impaired behavioral performances, worsened neuronal and synaptic loss, and increased astrogliosis at nine months of age. Transcriptomic profiling in APOE4-expressing α-synuclein mice highlighted altered lipid and energy metabolism, and synapse-related pathways. We also observed an effect of APOE4 on α-synuclein pathology in human postmortem brains with LBD and minimal amyloid pathology. Our data demonstrates a pathogenic role of APOE4 in exacerbating α-synuclein pathology independent of amyloid, providing mechanistic insights into how APOE4 increases the risk of LBD.
- Published
- 2019
29. Vascular apoE4 Impairs Brain Cognition by Modulating Glio-Vascular Functions
- Author
-
Hiroshi Oue, Cherukuri Yesesri, Akari Yamazaki, John D. Fryer, Yan W. Asmann, Wenhui Qiao, Chia Chen Liu, Yingxue Ren, Betty Y. S. Kim, Yuanxin Chen, Yu Yamazaki, Yuka A. Martens, Guojun Bu, Takahisa Kanekiyo, Aishe Kurti, and Ying Li
- Subjects
Apolipoprotein E ,Cell type ,Microglia ,Biology ,Phenotype ,Mural cell ,Pathogenesis ,medicine.anatomical_structure ,Arteriole ,medicine.artery ,medicine ,lipids (amino acids, peptides, and proteins) ,Neuroscience ,Astrocyte - Abstract
The e4 allele of the apolipoprotein E gene (APOE4) is the strongest genetic risk factor for late-onset Alzheimer’s disease (AD) compared to the common e3 allele. Because APOE4 is also associated with an increased risk for several vascular conditions including small vessel disease and vascular cognitive impairment (VCI), apoE4 effects on cerebrovasculature might be a converging pathway impacting brain homeostasis. As apoE is abundantly expressed in multiple brain cell types including astrocytes, microglia, and vascular mural cells (VMCs), defining the in vivo effect of apoE4 expressed by individual cell types is critical for addressing its pathogenic mechanism. Here, using conditional mouse models, we show that VMC-specific apoE4 expression impairs cognition and glio-vascular functions. Expression of either apoE3 or apoE4 in VMCs was sufficient to rescue the hypercholesterolemia and atherosclerosis phenotypes seen in the Apoe-knockout mice. Intriguingly, vascular specific expression of apoE4, but not apoE3, led to decreased arteriole blood flow, impaired spatial learning, and increased anxiety-like phenotypes. Single cell RNA sequencing of vascular and glial cells isolated from these mice revealed that conditional expression of apoE4 in VMCs was associated with astrocyte activation while expression of apoE3 in VMCs was associated with angiogenic signatures in pericytes. Together, our data support cell-autonomous effects of vascular apoE on multiple brain homeostatic processes in an isoform-dependent manner, suggesting a critical contribution of vascular apoE in the pathogenesis of AD and VCI.
- Published
- 2019
- Full Text
- View/download PDF
30. Alzheimer's Risk Factors Age, APOE Genotype, and Sex Drive Distinct Molecular Pathways
- Author
-
Yan W. Asmann, Rima Kaddurah-Daouk, Guojun Bu, Jiaying Zheng, Hiroshi Oue, Wenhui Qiao, Lindsey M. Felton, Yuka A. Martens, Tomonori Aikawa, Chia Chen Liu, Yu Yamazaki, Olivia N. Attrebi, Takahisa Kanekiyo, Na Zhao, Francis Shue, Berkiye Sonoustoun, Alexandra Kueider-Paisley, Nilufer Ertekin-Taner, Masaya Tachibana, Lucy Job, Yingxue Ren, Siamak MahmoudianDehkordi, Zonghua Li, and Akari Yamazaki
- Subjects
Genetics ,Apolipoprotein E ,Transcriptome ,Downregulation and upregulation ,TREM2 ,Genotype ,Metabolome ,Disease ,Biology ,Gene - Abstract
Evidence suggests interplay among the three major risk factors for Alzheimer’s disease (AD): age, APOE genotype, and sex. Here, we present comprehensive datasets and analyses of brain transcriptomes and blood metabolomes from human APOE2, APOE3 and APOE4-targeted replacement mice across young, middle, and old ages with both sexes. We found that age had the greatest impact on brain transcriptomes highlighted by an immune module led by Trem2 and Tyrobp, whereas APOE4 was associated with upregulation of multiple Serpina3 genes. Importantly, these networks and gene expression changes were mostly conserved in human brains. Finally, we observed a significant interaction between age, APOE genotype, and sex on unfolded protein response pathway. In the periphery, APOE2 drove distinct blood metabolome profile highlighted by the upregulation of lipid metabolites. Our work identifies unique and interactive molecular pathways underlying AD risk factors providing valuable resources for discovery and validation research in model systems and humans.
- Published
- 2019
- Full Text
- View/download PDF
31. Amyloid-beta modulates microglial responses by binding to the triggering receptor expressed on myeloid cells 2 (TREM2)
- Author
-
Ying Xu, Xiao Fen Chen, Guojun Bu, Huaxi Xu, Kai Wang, Yuka A. Martens, Li Zhong, Linbei Wu, Daxin Wang, Ruizhi Huang, Zhe Wang, Dan Can, Jianguo Li, and Zongqi Wang
- Subjects
0301 basic medicine ,Amyloid beta ,microglia ,Syk ,lcsh:Geriatrics ,migration ,lcsh:RC346-429 ,Mice ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Alzheimer Disease ,TREM2 ,medicine ,Animals ,Humans ,Receptors, Immunologic ,Receptor ,Molecular Biology ,Protein kinase B ,lcsh:Neurology. Diseases of the nervous system ,Mice, Knockout ,Amyloid beta-Peptides ,Membrane Glycoproteins ,biology ,Microglia ,β-amyloid ,Chemistry ,Cell biology ,Mice, Inbred C57BL ,lcsh:RC952-954.6 ,030104 developmental biology ,medicine.anatomical_structure ,biology.protein ,Phosphorylation ,Neurology (clinical) ,Signal transduction ,Alzheimer’s disease ,Research Article - Abstract
Background TREM2 is an innate immune receptor specifically expressed in microglia. Coding variations in TREM2 have been reported to increase the risk for Alzheimer’s disease (AD) and other neurodegenerative diseases. While multiple studies support a role for TREM2 in microglial recruitment to amyloid plaques, the chemoattractant factor modulating TREM2-dependent microglial responses has not been defined. Methods Potential binding of oligomeric amyloid-β 1–42 (oAβ1–42) to TREM2 was tested by complementary approaches including solid phase binding, surface plasmon resonance and immunoprecipitation assays. The ability of oAβ1–42 to activate TREM2 signaling pathways was examined by analyzing the phosphorylation of Syk and Akt in primary microglia as well as TREM2-mediated signaling in a reporter cell system. Lastly, the functional outcome of oAβ1–42-TREM2 interaction was tested by examining impacts on microglial migration in vitro and clustering around oAβ1–42-bearing brain areas in vivo. Results We found that oAβ1–42 bound to TREM2 with high affinity and activated TREM2-dependent signaling pathway. Neither monomeric nor scrambled Aβ bound to TREM2 supporting a specific interaction between oAβ and TREM2. The disease-associated mutations of TREM2 reduced its binding affinity to oAβ1–42. Furthermore, we identified several positively charged amino acids within residues 31–91 of TREM2 that were crucial for its interaction with oAβ1–42. Importantly, oAβ1–42 promoted microglial migration in vitro and clustering in vivo in a TREM2-dependent manner. Conclusions Our data establish a critical link between oAβ1–42, a major pathological component of AD, and TREM2, a strong genetic risk factor for AD expressed in microglia, and suggest that such interaction contributes to the pathogenic events in AD by modulating microglial responses. Electronic supplementary material The online version of this article (10.1186/s13024-018-0247-7) contains supplementary material, which is available to authorized users.
- Published
- 2018
- Full Text
- View/download PDF
32. Association study between multiple system atrophy and TREM2 p.R47H
- Author
-
Zbigniew K. Wszolek, Wolfgang Singer, Ronald L. Walton, Alexandra I. Soto, William P. Cheshire, Phillip A. Low, Guojun Bu, Yuka A. Martens, Ryan J. Uitti, Shunsuke Koga, Kotaro Ogaki, Michael G. Heckman, Emily R. Vargas, Dennis W. Dickson, Catherine Labbé, Jay A. van Gerpen, Steven G. Younkin, Oswaldo Lorenzo-Betancor, Owen A. Ross, and Shinsuke Fujioka
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Disease ,Gastroenterology ,Article ,03 medical and health sciences ,symbols.namesake ,0302 clinical medicine ,Atrophy ,stomatognathic system ,Internal medicine ,parasitic diseases ,mental disorders ,medicine ,Risk factor ,Genotyping ,Genetics (clinical) ,Sanger sequencing ,business.industry ,TREM2 ,Odds ratio ,medicine.disease ,3. Good health ,nervous system diseases ,030104 developmental biology ,nervous system ,symbols ,Neurology (clinical) ,Alzheimer's disease ,business ,030217 neurology & neurosurgery - Abstract
ObjectiveThe triggering receptor expressed on myeloid cells 2 (TREM2) p.R47H substitution (rs75932628) is a risk factor for Alzheimer disease (AD) but has not been well studied in relation to the risk of multiple system atrophy (MSA); the aim of this study was to evaluate the association between the TREM2 p.R47H variant and the risk of MSA.MethodsA total of 168 patients with pathologically confirmed MSA, 89 patients with clinically diagnosed MSA, and 1,695 controls were included. TREM2 p.R47H was genotyped and assessed for association with MSA. Positive results in the Taqman genotyping assay were confirmed by Sanger sequencing. The primary comparison involved patients with pathologically confirmed MSA and controls due to the definitive MSA diagnosis in the pathologically confirmed series.ResultsWe identified TREM2 p.R47H in 3 patients with pathologically confirmed MSA (1.79%), 1 patient with clinically diagnosed MSA (1.12%), and 7 controls (0.41%). Minimal AD pathology was observed for the pathologically confirmed MSA p.R47H carriers. For the primary comparison of patients with pathologically confirmed MSA and controls, risk of disease was significantly higher for p.R47H carriers (odds ratio [OR]: 4.39, p = 0.033). When supplementing the 168 pathologically confirmed patients with the 89 clinically diagnosed and examining the combined MSA series, the association with TREM2 p.R47H remained significant (OR: 3.81, p = 0.034).ConclusionsOur preliminary results suggest that the TREM2 p.R47H substitution may be a risk factor for MSA, implying a link to neuroinflammatory processes, especially microglial activation. Validation of this finding will be important, given our relatively small sample size; meta-analytic approaches will be needed to better define the role of this variant in MSA.
- Published
- 2018
33. Apolipoprotein E4 Impairs Neuronal Insulin Signaling by Trapping Insulin Receptor in the Endosomes
- Author
-
Chia Chen Liu, Joshua Knight, Alexandra J. Van Ingelgom, Meghan M. Painter, Na Zhao, Cynthia Linares, Guojun Bu, Yuka A. Martens, and Patrick Sullivan
- Subjects
Male ,0301 basic medicine ,Apolipoprotein E ,Aging ,medicine.medical_specialty ,Endosome ,Apolipoprotein E4 ,Cell Respiration ,Primary Cell Culture ,Apolipoprotein E3 ,Mice, Transgenic ,Endosomes ,Diet, High-Fat ,Article ,Pathogenesis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Insulin resistance ,Internal medicine ,Diabetes mellitus ,mental disorders ,medicine ,Animals ,Insulin ,Glycolysis ,Allele ,Mice, Knockout ,Neurons ,biology ,General Neuroscience ,medicine.disease ,Receptor, Insulin ,Mitochondria ,Insulin receptor ,030104 developmental biology ,Endocrinology ,biology.protein ,Female ,lipids (amino acids, peptides, and proteins) ,Insulin Resistance ,human activities ,030217 neurology & neurosurgery - Abstract
Diabetes and impaired brain insulin signaling are linked to the pathogenesis of Alzheimer’s disease (AD). The association between diabetes and AD-associated amyloid pathology is stronger among carriers of the apolipoprotein E (APOE) ε4 gene allele, the strongest genetic risk factor for late-onset AD. Here we report that apoE4 impairs neuronal insulin signaling in human apoE-targeted replacement (TR) mice in an age-dependent manner. High fat diet (HFD) accelerates these effects in apoE4-TR mice at middle age. In primary neurons, apoE4 interacts with insulin receptor and impairs its trafficking by trapping it in the endosomes, leading to impaired insulin signaling and insulin-stimulated mitochondrial respiration and glycolysis. In aging brains, the increased apoE4 aggregation and compromised endosomal function further exacerbate the inhibitory effects of apoE4 on insulin signaling and related functions. Together, our study provides novel mechanistic insights into the pathogenic mechanisms of apoE4 and insulin resistance in AD.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.