- Browse by Author
Browsing by Author "Harari, Oscar"
Now showing 1 - 8 of 8
Results Per Page
Sort Options
Item Analysis of whole genome-transcriptomic organization in brain to identify genes associated with alcoholism(Springer Nature, 2019-02-14) Kapoor, Manav; Wang, Jen-Chyong; Farris, Sean P.; Liu, Yunlong; McClintick, Jeanette; Gupta, Ishaan; Meyers, Jacquelyn L.; Bertelsen, Sarah; Chao, Michael; Nurnberger, John; Tischfield, Jay; Harari, Oscar; Zeran, Li; Hesselbrock, Victor; Bauer, Lance; Raj, Towfique; Porjesz, Bernice; Agrawal, Arpana; Foroud, Tatiana; Edenberg, Howard J.; Mayfield, R. Dayne; Goate, Alison; Medical and Molecular Genetics, School of MedicineAlcohol exposure triggers changes in gene expression and biological pathways in human brain. We explored alterations in gene expression in the Pre-Frontal Cortex (PFC) of 65 alcoholics and 73 controls of European descent, and identified 129 genes that showed altered expression (FDR < 0.05) in subjects with alcohol dependence. Differentially expressed genes were enriched for pathways related to interferon signaling and Growth Arrest and DNA Damage-inducible 45 (GADD45) signaling. A coexpression module (thistle2) identified by weighted gene co-expression network analysis (WGCNA) was significantly correlated with alcohol dependence, alcohol consumption, and AUDIT scores. Genes in the thistle2 module were enriched with genes related to calcium signaling pathways and showed significant downregulation of these pathways, as well as enrichment for biological processes related to nicotine response and opioid signaling. A second module (brown4) showed significant upregulation of pathways related to immune signaling. Expression quantitative trait loci (eQTLs) for genes in the brown4 module were also enriched for genetic associations with alcohol dependence and alcohol consumption in large genome-wide studies included in the Psychiatric Genetic Consortium and the UK Biobank's alcohol consumption dataset. By leveraging multi-omics data, this transcriptome analysis has identified genes and biological pathways that could provide insight for identifying therapeutic targets for alcohol dependence.Item Circular RNA detection identifies circPSEN1 alterations in brain specific to autosomal dominant Alzheimer's disease(BMC, 2022-03-04) Chen, Hsiang‑Han; Eteleeb, Abdallah; Wang, Ciyang; Fernandez, Maria Victoria; Budde, John P.; Bergmann, Kristy; Norton, Joanne; Wang, Fengxian; Ebl, Curtis; Morris, John C.; Perrin, Richard J.; Bateman, Randall J.; McDade, Eric; Xiong, Chengjie; Goate, Alison; Farlow, Martin; Chhatwal, Jasmeer; Schofield, Peter R.; Chui, Helena; Harari, Oscar; Cruchaga, Carlos; Ibanez, Laura; Dominantly Inherited Alzheimer Network; Neurology, School of MedicineBackground: Autosomal-dominant Alzheimer's disease (ADAD) is caused by pathogenic mutations in APP, PSEN1, and PSEN2, which usually lead to an early age at onset (< 65). Circular RNAs are a family of non-coding RNAs highly expressed in the nervous system and especially in synapses. We aimed to investigate differences in brain gene expression of linear and circular transcripts from the three ADAD genes in controls, sporadic AD, and ADAD. Methods: We obtained and sequenced RNA from brain cortex using standard protocols. Linear counts were obtained using the TOPMed pipeline; circular counts, using python package DCC. After stringent quality control (QC), we obtained the counts for PSEN1, PSEN2 and APP genes. Only circPSEN1 passed QC. We used DESeq2 to compare the counts across groups, correcting for biological and technical variables. Finally, we performed in-silico functional analyses using the Circular RNA interactome website and DIANA mirPath software. Results: Our results show significant differences in gene counts of circPSEN1 in ADAD individuals, when compared to sporadic AD and controls (ADAD = 21, AD = 253, Controls = 23-ADADvsCO: log2FC = 0.794, p = 1.63 × 10-04, ADADvsAD: log2FC = 0.602, p = 8.22 × 10-04). The high gene counts are contributed by two circPSEN1 species (hsa_circ_0008521 and hsa_circ_0003848). No significant differences were observed in linear PSEN1 gene expression between cases and controls, indicating that this finding is specific to the circular forms. In addition, the high circPSEN1 levels do not seem to be specific to PSEN1 mutation carriers; the counts are also elevated in APP and PSEN2 mutation carriers. In-silico functional analyses suggest that circPSEN1 is involved in several pathways such as axon guidance (p = 3.39 × 10-07), hippo signaling pathway (p = 7.38 × 10-07), lysine degradation (p = 2.48 × 10-05) or Wnt signaling pathway (p = 5.58 × 10-04) among other KEGG pathways. Additionally, circPSEN1 counts were able to discriminate ADAD from sporadic AD and controls with an AUC above 0.70. Conclusions: Our findings show the differential expression of circPSEN1 is increased in ADAD. Given the biological function previously ascribed to circular RNAs and the results of our in-silico analyses, we hypothesize that this finding might be related to neuroinflammatory events that lead or that are caused by the accumulation of amyloid-beta.Item Genome-wide association study identifies four novel loci associated with Alzheimer's endophenotypes and disease modifiers(Springer Verlag, 2017-05) Deming, Yuetiva; Li, Zeran; Kapoor, Manav; Harari, Oscar; Del-Aguila, Jorge L.; Black, Kathleen; Carrell, David; Cai, Yefei; Fernandez, Maria Victoria; Budde, John; Ma, Shengmei; Saef, Benjamin; Howells, Bill; Huang, Kuanlin; Bertelsen, Sarah; Fagan, Anne M.; Holtzman, David M.; Morris, John C.; Kim, Sungeun; Saykin, Andrew J.; De Jager, Philip L.; Albert, Marilyn; Moghekar, Abhay; O’Brien, Richard; Riemenschneider, Matthias; Petersen, Ronald C.; Blennow, Kaj; Zetterberg, Henrik; Minthon, Lennart; Van Deerlin, Vivianna M.; Lee, Virginia Man-Yee; Shaw, Leslie M.; Trojanowski, John Q.; Schellenberg, Gerard; Haines, Jonathan L.; Mayeux, Richard; Pericak-Vance, Margaret A.; Farrer, Lindsay A.; Peskind, Elaine R.; Li, Ge; Di Narzo, Antonio F.; Alzheimer’s Disease Neuroimaging Initiative (ADGC). The Alzheimer Disease Genetic Consortium (ADGC); Kauwe, John S. K.; Goate, Alison M.; Cruchaga, Carlos; Medicine, School of MedicineMore than 20 genetic loci have been associated with risk for Alzheimer's disease (AD), but reported genome-wide significant loci do not account for all the estimated heritability and provide little information about underlying biological mechanisms. Genetic studies using intermediate quantitative traits such as biomarkers, or endophenotypes, benefit from increased statistical power to identify variants that may not pass the stringent multiple test correction in case-control studies. Endophenotypes also contain additional information helpful for identifying variants and genes associated with other aspects of disease, such as rate of progression or onset, and provide context to interpret the results from genome-wide association studies (GWAS). We conducted GWAS of amyloid beta (Aβ42), tau, and phosphorylated tau (ptau181) levels in cerebrospinal fluid (CSF) from 3146 participants across nine studies to identify novel variants associated with AD. Five genome-wide significant loci (two novel) were associated with ptau181, including loci that have also been associated with AD risk or brain-related phenotypes. Two novel loci associated with Aβ42 near GLIS1 on 1p32.3 (β = -0.059, P = 2.08 × 10-8) and within SERPINB1 on 6p25 (β = -0.025, P = 1.72 × 10-8) were also associated with AD risk (GLIS1: OR = 1.105, P = 3.43 × 10-2), disease progression (GLIS1: β = 0.277, P = 1.92 × 10-2), and age at onset (SERPINB1: β = 0.043, P = 4.62 × 10-3). Bioinformatics indicate that the intronic SERPINB1 variant (rs316341) affects expression of SERPINB1 in various tissues, including the hippocampus, suggesting that SERPINB1 influences AD through an Aβ-associated mechanism. Analyses of known AD risk loci suggest CLU and FERMT2 may influence CSF Aβ42 (P = 0.001 and P = 0.009, respectively) and the INPP5D locus may affect ptau181 levels (P = 0.009); larger studies are necessary to verify these results. Together the findings from this study can be used to inform future AD studies.Item Meningeal lymphatics affect microglia responses and anti-Aβ immunotherapy(Springer Nature, 2021) Da Mesquita, Sandro; Papadopoulos, Zachary; Dykstra, Taitea; Brase, Logan; Farias, Fabiana Geraldo; Wall, Morgan; Jiang, Hong; Kodira, Chinnappa Dilip; de Lima, Kalil Alves; Herz, Jasmin; Louveau, Antoine; Goldman, Dylan H.; Salvador, Andrea Francesca; Onengut-Gumuscu, Suna; Farber, Emily; Dabhi, Nisha; Kennedy, Tatiana; Milam, Mary Grace; Baker, Wendy; Smirnov, Igor; Rich, Stephen S.; Dominantly Inherited Alzheimer Network; Benitez, Bruno A.; Karch, Celeste M.; Perrin, Richard J.; Farlow, Martin; Chhatwal, Jasmeer P.; Holtzman, David M.; Cruchaga, Carlos; Harari, Oscar; Kipnis, Jonathan; Neurology, School of MedicineAlzheimer's disease (AD) is the most prevalent cause of dementia1. Although there is no effective treatment for AD, passive immunotherapy with monoclonal antibodies against amyloid beta (Aβ) is a promising therapeutic strategy2,3. Meningeal lymphatic drainage has an important role in the accumulation of Aβ in the brain4, but it is not known whether modulation of meningeal lymphatic function can influence the outcome of immunotherapy in AD. Here we show that ablation of meningeal lymphatic vessels in 5xFAD mice (a mouse model of amyloid deposition that expresses five mutations found in familial AD) worsened the outcome of mice treated with anti-Aβ passive immunotherapy by exacerbating the deposition of Aβ, microgliosis, neurovascular dysfunction, and behavioural deficits. By contrast, therapeutic delivery of vascular endothelial growth factor C improved clearance of Aβ by monoclonal antibodies. Notably, there was a substantial overlap between the gene signature of microglia from 5xFAD mice with impaired meningeal lymphatic function and the transcriptional profile of activated microglia from the brains of individuals with AD. Overall, our data demonstrate that impaired meningeal lymphatic drainage exacerbates the microglial inflammatory response in AD and that enhancement of meningeal lymphatic function combined with immunotherapies could lead to better clinical outcomes.Item Metabolomic and lipidomic signatures in autosomal dominant and late-onset Alzheimer's disease brains(Wiley, 2023) Novotny, Brenna C.; Fernandez, Maria Victoria; Wang, Ciyang; Budde, John P.; Bergmann, Kristy; Eteleeb, Abdallah M.; Bradley, Joseph; Webster, Carol; Ebl, Curtis; Norton, Joanne; Gentsch, Jen; Dube, Umber; Wang, Fengxian; Morris, John C.; Bateman, Randall J.; Perrin, Richard J.; McDade, Eric; Xiong, Chengjie; Chhatwal, Jasmeer; Dominantly Inherited Alzheimer Network (DIAN) Study Group; Alzheimer's Disease Neuroimaging Initiative; Alzheimer's Disease Metabolomics Consortium (ADMC); Goate, Alison; Farlow, Martin; Schofield, Peter; Chui, Helena; Karch, Celeste M.; Cruchaga, Carlos; Benitez, Bruno A.; Harari, Oscar; Neurology, School of MedicineIntroduction: The identification of multiple genetic risk factors for Alzheimer's disease (AD) suggests that many pathways contribute to AD onset and progression. However, the metabolomic and lipidomic profiles in carriers of distinct genetic risk factors are not fully understood. The metabolome can provide a direct image of dysregulated pathways in the brain. Methods: We interrogated metabolomic signatures in the AD brain, including carriers of pathogenic variants in APP, PSEN1, and PSEN2 (autosomal dominant AD; ADAD), APOE ɛ4, and TREM2 risk variant carriers, and sporadic AD (sAD). Results: We identified 133 unique and shared metabolites associated with ADAD, TREM2, and sAD. We identified a signature of 16 metabolites significantly altered between groups and associated with AD duration. Discussion: AD genetic variants show distinct metabolic perturbations. Investigation of these metabolites may provide greater insight into the etiology of AD and its impact on clinical presentation. Highlights: APP/PSEN1/PSEN2 and TREM2 variant carriers show distinct metabolic changes. A total of 133 metabolites were differentially abundant in AD genetic groups. β-citrylglutamate is differentially abundant in autosomal dominant, TREM2, and sporadic AD. A 16-metabolite profile shows differences between Alzheimer's disease (AD) genetic groups. The identified metabolic profile is associated with duration of disease.Item Parenchymal border macrophages regulate the flow dynamics of the cerebrospinal fluid(Springer, 2022-11-09) Drieu, Antoine; Du, Siling; Storck, Steffen E.; Rustenhoven, Justin; Papadopoulos, Zachary; Dykstra, Taitea; Zhong, Fenghe; Kim, Kyungdeok; Blackburn, Susan; Mamuladze, Tornike; Harari, Oscar; Karch, Celeste M.; Bateman, Randall J.; Perrin, Richard; Farlow, Martin; Chhatwal, Jasmeer; Dominantly Inherited Alzheimer Network; Hu, Song; Randolph, Gwendalyn J.; Smirnov, Igor; Kipnis, Jonathan; Neurology, School of MedicineMacrophages are important players for the maintenance of tissue homeostasis1. Perivascular and leptomeningeal macrophages reside in close proximity to the central nervous system (CNS) parenchyma2, and their role in CNS physiology has not been well enough studied to date. Given their continuous interaction with the cerebrospinal fluid (CSF) and strategic positioning, we refer to these cells collectively as parenchymal border macrophages (PBMs). Here, we demonstrate that PBMs regulate CSF flow dynamics. We identify a subpopulation of PBMs expressing high levels of CD163 and Lyve1 (scavenger receptor proteins), located in close proximity to the brain arterial tree, and show that Lyve1+ PBMs regulate arterial motion that drives CSF flow. Pharmacological or genetic depletion of PBMs led to accumulation of extracellular matrix proteins, obstructing CSF access to perivascular spaces hence impairing CNS perfusion and clearance. Aging-associated alterations in PBMs and impairment of CSF dynamics were restored upon intracisternal injection of macrophage colony-stimulating growth factor (M-CSF). Human single-nuclei RNA sequencing data obtained from Alzheimer’s disease (AD) patients and healthy controls point to changes in phagocytosis/endocytosis and interferon-gamma (IFNγ) signaling on PBMs, pathways that are corroborated in a mouse AD model. Collectively, our results identify PBMs as novel cellular regulators of CSF flow dynamics, which could potentially be targeted pharmacologically to alleviate brain clearance deficits associated with aging and AD.Item Proteomics of brain, CSF, and plasma identifies molecular signatures for distinguishing sporadic and genetic Alzheimer's disease(American Association for the Advancement of Science, 2023) Sung, Yun Ju; Yang, Chengran; Norton, Joanne; Johnson, Matt; Fagan, Anne; Bateman, Randall J.; Perrin, Richard J.; Morris, John C.; Farlow, Martin R.; Chhatwal, Jasmeer P.; Schofield, Peter R.; Chui, Helena; Wang, Fengxian; Novotny, Brenna; Eteleeb, Abdallah; Karch, Celeste; Schindler, Suzanne E.; Rhinn, Herve; Johnson, Erik C. B.; Oh, Hamilton Se-Hwee; Rutledge, Jarod Evert; Dammer, Eric B.; Seyfried, Nicholas T.; Wyss-Coray, Tony; Harari, Oscar; Cruchaga, Carlos; Neurology, School of MedicineProteomic studies for Alzheimer's disease (AD) are instrumental in identifying AD pathways but often focus on single tissues and sporadic AD cases. Here, we present a proteomic study analyzing 1305 proteins in brain tissue, cerebrospinal fluid (CSF), and plasma from patients with sporadic AD, TREM2 risk variant carriers, patients with autosomal dominant AD (ADAD), and healthy individuals. We identified 8 brain, 40 CSF, and 9 plasma proteins that were altered in individuals with sporadic AD, and we replicated these findings in several external datasets. We identified a proteomic signature that differentiated TREM2 variant carriers from both individuals with sporadic AD and healthy individuals. The proteins associated with sporadic AD were also altered in patients with ADAD, but with a greater effect size. Brain-derived proteins associated with ADAD were also replicated in additional CSF samples. Enrichment analyses highlighted several pathways, including those implicated in AD (calcineurin and Apo E), Parkinson's disease (α-synuclein and LRRK2), and innate immune responses (SHC1, ERK-1, and SPP1). Our findings suggest that combined proteomics across brain tissue, CSF, and plasma can be used to identify markers for sporadic and genetically defined AD.Item Single-nucleus RNA-sequencing of autosomal dominant Alzheimer disease and risk variant carriers(Springer Nature, 2023-04-21) Brase, Logan; You, Shih-Feng; D'Oliveira Albanus, Ricardo; Del-Aguila, Jorge L.; Dai, Yaoyi; Novotny, Brenna C.; Soriano-Tarraga, Carolina; Dykstra, Taitea; Fernandez, Maria Victoria; Budde, John P.; Bergmann, Kristy; Morris, John C.; Bateman, Randall J.; Perrin, Richard J.; McDade, Eric; Xiong, Chengjie; Goate, Alison M.; Farlow, Martin; Dominantly Inherited Alzheimer Network (DIAN); Sutherland, Greg T.; Kipnis, Jonathan; Karch, Celeste M.; Benitez, Bruno A.; Harari, Oscar; Neurology, School of MedicineGenetic studies of Alzheimer disease (AD) have prioritized variants in genes related to the amyloid cascade, lipid metabolism, and neuroimmune modulation. However, the cell-specific effect of variants in these genes is not fully understood. Here, we perform single-nucleus RNA-sequencing (snRNA-seq) on nearly 300,000 nuclei from the parietal cortex of AD autosomal dominant (APP and PSEN1) and risk-modifying variant (APOE, TREM2 and MS4A) carriers. Within individual cell types, we capture genes commonly dysregulated across variant groups. However, specific transcriptional states are more prevalent within variant carriers. TREM2 oligodendrocytes show a dysregulated autophagy-lysosomal pathway, MS4A microglia have dysregulated complement cascade genes, and APOEε4 inhibitory neurons display signs of ferroptosis. All cell types have enriched states in autosomal dominant carriers. We leverage differential expression and single-nucleus ATAC-seq to map GWAS signals to effector cell types including the NCK2 signal to neurons in addition to the initially proposed microglia. Overall, our results provide insights into the transcriptional diversity resulting from AD genetic architecture and cellular heterogeneity. The data can be explored on the online browser (http://web.hararilab.org/SNARE/).