Ancestry-related differences in chromatin accessibility and gene expression of APOE ε4 are associated with Alzheimer's disease risk
Katrina Celis
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Search for more papers by this authorMaria D. M. Muniz Moreno
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Search for more papers by this authorFarid Rajabli
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Search for more papers by this authorPatrice Whitehead
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Search for more papers by this authorKara Hamilton-Nelson
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Search for more papers by this authorDerek M. Dykxhoorn
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
Search for more papers by this authorKaren Nuytemans
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
Search for more papers by this authorLiyong Wang
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
Search for more papers by this authorMargaret Flanagan
Northwestern ADC Neuropathology Core, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
Search for more papers by this authorSandra Weintraub
Northwestern ADC Neuropathology Core, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
Search for more papers by this authorChangiz Geula
Northwestern ADC Neuropathology Core, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
Search for more papers by this authorMarla Gearing
Goizueta Alzheimer's Disease Research Center, Emory University, Atlanta, Georgia, USA
Search for more papers by this authorClifton L. Dalgard
The American Genome Center, Uniformed Services University, Bethesda, Maryland, USA
Collaborative Health Initiative Research Program, Henry Jackson Foundation, Bethesda, Maryland, USA
Department of Anatomy Physiology & Genetics, Uniformed Services University, Bethesda, Maryland, USA
Search for more papers by this authorFulai Jin
Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
Search for more papers by this authorDavid A. Bennett
Department of Neurological Sciences, Rush University, Chicago, Illinois, USA
Search for more papers by this authorTheresa Schuck
The Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
Search for more papers by this authorMargaret A. Pericak-Vance
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
Search for more papers by this authorAnthony J. Griswold
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
Search for more papers by this authorJuan I. Young
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
Search for more papers by this authorCorresponding Author
Jeffery M. Vance
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
Correspondence
Jeffery M. Vance, University of Miami Miller School of Medicine, John P. Hussman Institute for Human Genomics, 1501 NW 10th Ave, BRB 509, Miami, FL 33136, USA.
Email: [email protected]
Search for more papers by this authorKatrina Celis
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Search for more papers by this authorMaria D. M. Muniz Moreno
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Search for more papers by this authorFarid Rajabli
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Search for more papers by this authorPatrice Whitehead
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Search for more papers by this authorKara Hamilton-Nelson
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Search for more papers by this authorDerek M. Dykxhoorn
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
Search for more papers by this authorKaren Nuytemans
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
Search for more papers by this authorLiyong Wang
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
Search for more papers by this authorMargaret Flanagan
Northwestern ADC Neuropathology Core, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
Search for more papers by this authorSandra Weintraub
Northwestern ADC Neuropathology Core, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
Search for more papers by this authorChangiz Geula
Northwestern ADC Neuropathology Core, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
Search for more papers by this authorMarla Gearing
Goizueta Alzheimer's Disease Research Center, Emory University, Atlanta, Georgia, USA
Search for more papers by this authorClifton L. Dalgard
The American Genome Center, Uniformed Services University, Bethesda, Maryland, USA
Collaborative Health Initiative Research Program, Henry Jackson Foundation, Bethesda, Maryland, USA
Department of Anatomy Physiology & Genetics, Uniformed Services University, Bethesda, Maryland, USA
Search for more papers by this authorFulai Jin
Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
Search for more papers by this authorDavid A. Bennett
Department of Neurological Sciences, Rush University, Chicago, Illinois, USA
Search for more papers by this authorTheresa Schuck
The Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
Search for more papers by this authorMargaret A. Pericak-Vance
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
Search for more papers by this authorAnthony J. Griswold
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
Search for more papers by this authorJuan I. Young
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
Search for more papers by this authorCorresponding Author
Jeffery M. Vance
John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
Correspondence
Jeffery M. Vance, University of Miami Miller School of Medicine, John P. Hussman Institute for Human Genomics, 1501 NW 10th Ave, BRB 509, Miami, FL 33136, USA.
Email: [email protected]
Search for more papers by this authorJuan I. Young and Jeffery M. Vance contributed equally.
Abstract
Introduction
European local ancestry (ELA) surrounding apolipoprotein E (APOE) ε4 confers higher risk for Alzheimer's disease (AD) compared to African local ancestry (ALA). We demonstrated significantly higher APOE ε4 expression in ELA versus ALA in AD brains from APOE ε4/ε4 carriers. Chromatin accessibility differences could contribute to these expression changes.
Methods
We performed single nuclei assays for transposase accessible chromatin sequencing from the frontal cortex of six ALA and six ELA AD brains, homozygous for local ancestry and APOE ε4.
Results
Our results showed an increased chromatin accessibility at the APOE ε4 promoter area in ELA versus ALA astrocytes. This increased accessibility in ELA astrocytes extended genome wide. Genes with increased accessibility in ELA in astrocytes were enriched for synapsis, cholesterol processing, and astrocyte reactivity.
Discussion
Our results suggest that increased chromatin accessibility of APOE ε4 in ELA astrocytes contributes to the observed elevated APOE ε4 expression, corresponding to the increased AD risk in ELA versus ALA APOE ε4/ε4 carriers.
CONFLICTS OF INTEREST STATEMENT
K.C., M.D.M.M., F.R., P.W., K.H., D.M.D., K.N., L.W., M.F., S.W., C.G., M.G., C.L.D., F.J., D.A.B., T.S., M.A.P., A.J.G., J.I.Y., and J.M.V. have nothing to disclose. The authors declare that they have no competing interests. Author disclosures are available in the Supporting information.
Open Research
DATA AVAILABILITY STATEMENT
Data are available through the National Institute on Aging Genetics of Alzheimer's Disease Data Storage Site (NIAGADS) Data Sharing Service (DSS): https://dss.niagads.org/datasets/ng00067/
Supporting Information
Filename | Description |
---|---|
alz13075-sup-0001-FiguresS1-S6.pdf5.7 MB | Supporting Information |
alz13075-sup-0002-TablesS1-S7.docx336.2 KB | Supporting Information |
alz13075-sup-0003-SuppMat.pdf2.9 MB | Supporting Information |
Please note: The publisher is not responsible for the content or functionality of any supporting information supplied by the authors. Any queries (other than missing content) should be directed to the corresponding author for the article.
REFERENCES
- 1 GBD 2019 Diseases and Injuries Collaborators. Global burden of 369 diseases and injuries in 204 countries and territories, 1990-2019: a systematic analysis for the Global Burden of Disease Study. Lancet. 2020; 10258: 1204-1222.
- 2Yamazaki Y, Zhao N, Caulfield TR, Liu CC, Bu G. Apolipoprotein E and Alzheimer disease: pathobiology and targeting strategies. Nat Rev Neurol. 2019; 9: 501-518.
- 3Corder EH, Saunders AM, Strittmatter WJ, et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer's disease in late onset families. Science. 1993; 5123: 921-923.
- 4Farrer LA, Cupples LA, Haines JL, et al. Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. A meta-analysis. APOE and Alzheimer Disease Meta Analysis Consortium. JAMA. 1997; 16: 1349-1356.
- 5Graff-Radford NR, Green RC, Go RCP, et al. Association between apolipoprotein E genotype and Alzheimer disease in African American subjects. Arch Neurol. 2002; 4: 594-600.
- 6Neu SC, Pa J, Kukull W, et al. Apolipoprotein E genotype and sex risk factors for Alzheimer disease: a meta-analysis. JAMA Neurol. 2017; 10: 1178-1189.
- 7Tang MX, Stern Y, Marder K, et al. The APOE-epsilon4 allele and the risk of Alzheimer disease among African Americans, whites, and Hispanics. JAMA. 1998; 10: 751-755.
- 8Rajabli F, Feliciano BE, Celis K, et al. Ancestral origin of ApoE epsilon4 Alzheimer disease risk in Puerto Rican and African American populations. PLoS Genet. 2018; 12:e1007791.
- 9Blue EE, Horimoto ARVR, Mukherjee S, Wijsman EM, Thornton TA. Local ancestry at APOE modifies Alzheimer's disease risk in Caribbean Hispanics. Alzheimers Dement. 2019; 12: 1524-1532.
- 10Naslavsky MS, Suemoto CK, Brito LA, et al. Global and local ancestry modulate APOE association with Alzheimer's neuropathology and cognitive outcomes in an admixed sample. medRxiv 7/20.2022.
- 11Griswold AJ, Celis K, Bussies PL, et al. Increased APOEε4 expression is associated with the difference in Alzheimer's disease risk from diverse ancestral backgrounds. Alzheimers Dement. 2021; 7: 1179-1188.
- 12Bonn S, Furlong E. cis-Regulatory networks during development: a view of Drosophila. Curr Opin Genet Dev. 2008; 18(6): 513-520. doi: 10.1016/j.gde.2008.09.005 Epub 2008 Oct 16
- 13Wilczynski B, Furlong E. Challenges for modeling global gene regulatory networks during development: insights from Drosophila. Dev Biol. 2010; 340(2): 161-169. doi: 10.1016/j.ydbio.2009.10.032 Epub 2009 Oct 27
- 14Zinzen R, Girardot C, Gagneur J, Braun M, Furlong E. Combinatorial binding predicts spatio-temporal cis-regulatory activity. Nature. 2009; 462(7269): 65-70. doi: 10.1038/nature08531
- 15Gross D, Garrard W. Nuclease hypersensitive sites in chromatin. Annu Rev Biochem. 1988; 57: 159-197. doi: 10.1146/annurev.bi.57.070188.001111
- 16Kornberg R. Structure of chromatin. Annu Rev Biochem. 1977; 46: 931-954. doi: 10.1146/annurev.bi.46.070177.004435
- 17Kornberg R, Lorch Y. Chromatin-modifying and -remodeling complexes. Curr Opin Genet Dev. 1999; 9(2): 148-151. doi: 10.1016/S0959-437X(99)80022-7
- 18Luger K, Dechassa ML, Tremethick DJ. New insights into nucleosome and chromatin structure: an ordered state or a disordered affair? Nat Rev Mol Cell Biol. 2012; 7: 436-447.
- 19Tremethick DJ. Higher-order structures of chromatin: the elusive 30 nm fiber. Cell. 2007; 4: 651-654.
- 20Corces MR, Shcherbina A, Kundu S, et al. Single-cell epigenomic analyses implicate candidate causal variants at inherited risk loci for Alzheimer's and Parkinson's diseases. Nat Genet. 2020; 11: 1158-1168.
- 21Wang Y, Zhang X, Song Q, et al. Characterization of the chromatin accessibility in an Alzheimer's disease (AD) mouse model. Alzheimers Res Ther. 2020; 1. 29-020-00598-2.
- 22Morabito S, Miyoshi E, Michael N, et al. Single-nucleus chromatin accessibility and transcriptomic characterization of Alzheimer's disease. Nat Genet. 2021; 8: 1143-1155.
- 23Corces MR, Trevino AE, Hamilton EG, et al. An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues. Nat Methods. 2017; 10: 959-962.
- 24Corces R, Greenleaf WJ, Chang HY. Isolation of Nuceli from frozen tissue for ATAC-seq and other epigenomic assays V.1. Nat Methods. 2019. https://doi.org/10.17504/protocols.io.6t8herw
- 25Granja JM, Corces MR, Pierce SE, et al. ArchR is a scalable software package for integrative single-cell chromatin accessibility analysis. Nat Genet. 2021; 3: 403-411.
- 26Satpathy AT, Granja JM, Yost KE, et al. Massively parallel single-cell chromatin landscapes of human immune cell development and intratumoral T cell exhaustion. Nat Biotechnol. 2019; 8: 925-936.
- 27Granja JM, Klemm S, McGinnis LM, et al. Single-cell multiomic analysis identifies regulatory programs in mixed-phenotype acute leukemia. Nat Biotechnol. 2019; 12: 1458-1465.
- 28Korsunsky I, Millard N, Fan J, et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat Methods. 2019; 12: 1289-1296.
- 29Stuart T, Butler A, Hoffman P, et al. Comprehensive integration of single-cell data. Cell. 2019; 7: 1888-1902.e21.
- 30Zhang Y, Liu T, Meyer CA, et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 2008; 9: R137.
- 31Heinz S, Benner C, Spann N, et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol Cell. 2010; 4: 576-589.
- 32Duttke SH, Chang MW, Heinz S, Benner C. Identification and dynamic quantification of regulatory elements using total RNA. Genome Res. 2019; 11: 1836-1846.
- 33McLean CY, Bristor D, Hiller M, et al. GREAT improves functional interpretation of cis-regulatory regions. Nat Biotechnol. 2010; 5: 495-501.
- 34Fishilevich S, Nudel R, Rappaport N, et al. GeneHancer: genome-wide integration of enhancers and target genes in GeneCards. Database (Oxford). 2017; 2017:bax028. doi: 10.1093/database/bax028
- 35Quinlan AR, Hall IM. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics. 2010; 6: 841-842.
- 36Grant CE, Bailey TL, Noble WS. FIMO: scanning for occurrences of a given motif. Bioinformatics. 2011; 7: 1017-1018.
- 37Bailey TL, Johnson J, Grant CE, Noble WS. The MEME suite. Nucleic Acids Res. 2015; W1: W39-49.
- 38Castro-Mondragon JA, Riudavets-Puig R, Rauluseviciute I, et al. JASPAR 2022: the 9th release of the open-access database of transcription factor binding profiles. Nucleic Acids Res. 2022; D1: D165-D173.
- 39Jolma A, Yan J, Whitington T, et al. DNA-binding specificities of human transcription factors. Cell. 2013; 1-2: 327-339.
- 40Pachkov M, Balwierz PJ, Arnold P, Ozonov E, van Nimwegen E. SwissRegulon, a database of genome-wide annotations of regulatory sites: recent updates. Nucleic Acids Res Database issue. 2013; 41: D214-20.
- 41Karolchik D, Hinrichs AS, Furey TS, et al. The UCSC Table Browser data retrieval tool. Nucleic Acids Res Database issue. 2004; 32: D493-6.
- 42Chen EY, Tan CM, Kou Y, et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics. 2013. 128-2105-14-128.
- 43Ogata H, Goto S, Fujibuchi W, Kanehisa M. Computation with the KEGG pathway database. BioSystems. 1998; 1-2: 119-128.
- 44Ashburner M, Ball CA, Blake JA, et al. Gene ontology: tool for the unification of biology. The gene ontology consortium. Nat Genet. 2000; 1: 25-29.
- 45Joshi-Tope G, Gillespie M, Vastrik I, et al. Reactome: a knowledgebase of biological pathways. Nucleic Acids Res Database issue. 2005; 33: D428-32.
- 46 ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012; 7414: 57-74.
- 47Hao Y, Hao S, Andersen-Nissen E, et al. Integrated analysis of multimodal single-cell data. Cell. 2021; 13: 3573-3587.e29.
- 48Franzén O, Gan L, Björkegren J. PanglaoDB: a web server for exploration of mouse and human single-cell RNA sequencing data. Database (Oxford). 2019; 2019:baz046. doi: 10.1093/database/baz046
- 49Culhane AC, Schwarzl T, Sultana R, et al. GeneSigDB–a curated database of gene expression signatures. Nucleic Acids Res Database issue. 2010; 38: D716-25.
- 50Aaronson J, Beaumont V, Blevins RA, et al. HDinHD: a rich data portal for Huntington's disease research. J Huntingtons Dis. 2021; 3: 405-412.
- 51Liberzon A, Subramanian A, Pinchback R, Thorvaldsdottir H, Tamayo P, Mesirov JP. Molecular signatures database (MSigDB) 3.0. Bioinformatics. 2011; 12: 1739-1740.
- 52Grubman A, Chew G, Ouyang JF, et al. A single-cell atlas of entorhinal cortex from individuals with Alzheimer's disease reveals cell-type-specific gene expression regulation. Nat Neurosci. 2019; 12: 2087-2097.
- 53Sadick JS, O'Dea MR, Hasel P, Dykstra T, Faustin A, Liddelow SA. Astrocytes and oligodendrocytes undergo subtype-specific transcriptional changes in Alzheimer's disease. Neuron. 2022; 11: 1788-1805.e10.
- 54Srinivasan K, Friedman BA, Larson JL, et al. Untangling the brain's neuroinflammatory and neurodegenerative transcriptional responses. Nat Commun. 2016; 7:11295.
- 55Lin L, Park J, Ramachandran S, et al. Transcriptome sequencing reveals aberrant alternative splicing in Huntington's disease. Hum Mol Genet. 2016; 25(16): 3454-3466. doi: 10.1093/hmg/ddw187. Epub 2016 Jul 4.
- 56Lee H, Fenster RJ, Pineda SS, et al. Cell type-specific transcriptomics reveals that mutant huntingtin leads to mitochondrial RNA release and neuronal innate immune activation. Neuron. 2020; 5: 891-908.e8.
- 57Kunkle BW, Schmidt M, Klein HU, et al, Writing Group for the Alzheimer's Disease Genetics Consortium (ADGC). Novel Alzheimer disease risk loci and pathways in African American individuals using the African genome resources panel: a meta-analysis. JAMA Neurol. 2021; 1: 102-113.
- 58Kunkle BW, Grenier-Boley B, Sims R, et al, Alzheimer Disease Genetics Consortium (ADGC), European Alzheimer's Disease Initiative (EADI), Cohorts for Heart and Aging Research in Genomic Epidemiology Consortium (CHARGE), and Genetic and Environmental Risk in AD/Defining Genetic, Polygenic and Environmental Risk for Alzheimer's Disease Consortium (GERAD/PERADES). Genetic meta-analysis of diagnosed Alzheimer's disease identifies new risk loci and implicates Abeta, tau, immunity and lipid processing. Nat Genet. 2019; 3: 414-430.
- 59Nuytemans K, Lipkin Vasquez M, Wang L, et al. Identifying differential regulatory control of APOE varepsilon4 on African versus European haplotypes as potential therapeutic targets. Alzheimers Dement. 2022; 18(10): 1930-1942.
- 60Fernandez CG, Hamby ME, McReynolds ML, Ray WJ. The role of APOE4 in disrupting the homeostatic functions of astrocytes and microglia in aging and Alzheimer's disease. Front Aging Neurosci. 2019; 11: 14.
- 61Zhao H, Ji Q, Wu Z, et al. Destabilizing heterochromatin by APOE mediates senescence. Nat Aging. 2022; 2: 303-316.
- 62Narlikar GJ, Fan HY, Kingston RE. Cooperation between complexes that regulate chromatin structure and transcription. Cell. 2002; 4: 475-487.
- 63Merrill CB, Montgomery AB, Pabon MA, et al. Harnessing changes in open chromatin determined by ATAC-seq to generate insulin-responsive reporter constructs. BMC Genomics. 2022; 1. 399-022-08637-y.
- 64Klemm SL, Shipony Z, Greenleaf WJ. Chromatin accessibility and the regulatory epigenome. Nat Rev Genet. 2019; 4: 207-220.
- 65Belanger M, Magistretti PJ. The role of astroglia in neuroprotection. Dialogues Clin Neurosci. 2009; 3: 281-295.
- 66Belanger M, Allaman I, Magistretti PJ. Brain energy metabolism: focus on astrocyte-neuron metabolic cooperation. Cell Metab. 2011; 6: 724-738.
- 67Gibbs ME, Hutchinson D, Hertz L. Astrocytic involvement in learning and memory consolidation. Neurosci Biobehav Rev. 2008; 5: 927-944.
- 68Halassa MM, Florian C, Fellin T, et al. Astrocytic modulation of sleep homeostasis and cognitive consequences of sleep loss. Neuron. 2009; 2: 213-219.
- 69Phatnani H, Maniatis T. Astrocytes in neurodegenerative disease. Cold Spring Harb Perspect Biol. 2015; 6:a020628. doi: 10.1101/cshperspect.a020628
- 70Monterey MD, Wei H, Wu X, Wu JQ. The many faces of astrocytes in Alzheimer's. Disease Front Neurol. 2021; 12:619626.
- 71Liu Y. Clinical implications of chromatin accessibility in human cancers. Oncotarget. 2020; 18: 1666-1678.
- 72Harris RA, Raveendran M, Worley KC, Rogers J. Unusual sequence characteristics of human chromosome 19 are conserved across 11 nonhuman primates. BMC Evol Biol. 2020; 1: 33. 020-1595-9.
- 73Castresana J, Guigo R, Alba MM. Clustering of genes coding for DNA binding proteins in a region of atypical evolution of the human genome. J Mol Evol. 2004; 1: 72-79.
- 74Grimwood J, Gordon LA, Olsen A, et al. The DNA sequence and biology of human chromosome 19. Nature. 2004; 6982: 529-535.
- 75Spell C, Kolsch H, Lutjohann D, et al. SREBP-1a polymorphism influences the risk of Alzheimer's disease in carriers of the ApoE4 allele. Dement Geriatr Cogn Disord. 2004; 3-4: 245-249.
- 76Shi X, Ren S, Zhang B, et al. Analysis of the role of Puralpha in the pathogenesis of Alzheimer's disease based on RNA-seq and ChIP-seq. Sci Rep. 2021; 1. 12178-021-90982-1.
- 77Fang X, Zhong X, Yu G, Shao S, Yang Q. Vascular protective effects of KLF2 on Abeta-induced toxicity: implications for Alzheimer's disease. Brain Res. 2017; 1663: 174-183.