Abstract
To characterize the dysregulation of chromatin accessibility in Alzheimer’s disease (AD), we generated 636 ATAC-seq libraries from neuronal and nonneuronal nuclei isolated from the superior temporal gyrus and entorhinal cortex of 153 AD cases and 56 controls. By analyzing a total of ~20 billion read pairs, we expanded the repertoire of known open chromatin regions (OCRs) in the human brain and identified cell-type-specific enhancer–promoter interactions. We show that interindividual variability in OCRs can be leveraged to identify cis-regulatory domains (CRDs) that capture the three-dimensional structure of the genome (3D genome). We identified AD-associated effects on chromatin accessibility, the 3D genome and transcription factor (TF) regulatory networks. For one of the most AD-perturbed TFs, USF2, we validated its regulatory effect on lysosomal genes. Overall, we applied a systematic approach to understanding the role of the 3D genome in AD. We provide all data as an online resource for widespread community-based analysis.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
£14.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
£139.00 per year
only £11.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout
Similar content being viewed by others
Data availability
Raw data (FASTQ files) and processed data (BigWig files, peaks, and raw/normalized count matrices) are available via the AD Knowledge Portal (https://adknowledgeportal.org). The AD Knowledge Portal is a platform for accessing data, analyses and tools generated by the Accelerating Medicines Partnership (AMP-AD) Target Discovery Program and other National Institute on Aging (NIA)-supported programs to enable open-science practices and accelerate translational learning. The data, analyses and tools are shared early in the research cycle without a publication embargo on secondary use. Data are available for general research use according to the following requirements for data access and data attribution (https://adknowledgeportal.org/DataAccess/Instructions). For access to the content described in this manuscript, see https://doi.org/10.7303/syn21513145. Browsable UCSC genome browser tracks of processed data are available at http://icahn.mssm.edu/atacad.
External validation sets: MSBB RNA-seq of postmortem brains (Synapse ID: syn3157743), ATAC-seq on FANS-sorted NeuN+/− from postmortem brains (Synapse ID: syn20755767), H3K9ac ChIP–seq of postmortem brains (Synapse ID: syn4896408). ATAC-seq iPSC-derived neurons overexpressing MAPT gene (GEO: GSE97409), ROSMAP RNA-seq of postmortem brains (Synapse ID: syn3388564), fine-mapped eQTLs (https://alkesgroup.broadinstitute.org/LDSCORE/LDSC_QTL/, version ‘FE_META_TISSUE_GTEx_Brain_MaxCPP’), CTCF ChIP–seq peaks on human neural cell (GEO: GSE127577). OCRs (peaks) from The Cancer Genome Atlas (https://gdc.cancer.gov/about-data/publications/ATACseq-AWG), BOCA/BOCA2 brain epigenome atlas (https://icahn.mssm.edu/boca, https://icahn.mssm.edu/boca2), Dong. et al. 2021 (Synapse ID: syn25716684), Nott et al. 2019 (dbGaP ID: phs001373), and Meuleman et al. 2020 (ENCODE ID: ENCSR857UZV), fine-mapped eQTLs (https://alkesgroup.broadinstitute.org/LDSCORE/LDSC_QTL/, version ‘FE_META_TISSUE_GTEx_Brain_MaxCPP’), CTCF ChIP–seq on human neural cell (GEO: GSE127577), The Cancer Genome Atlas (https://gdc.cancer.gov/about-data/publications/ATACseq-AWG), REMC (http://www.roadmapepigenomics.org), mSigDB 7.0 (http://www.gsea-msigdb.org/), dbSNP v.151 (https://www.ncbi.nlm.nih.gov/snp/), PsychENCODE SNP-array: Capstone collection (https://psychencode.synapse.org/).
Code availability
The code used to perform the analysis described in this study is available at https://doi.org/10.7303/syn34034120.
References
Andrews, S. J., Fulton-Howard, B. & Goate, A. Interpretation of risk loci from genome-wide association studies of Alzheimer’s disease. Lancet Neurol. 19, 326–335 (2020).
Marzi, S. J. et al. A histone acetylome-wide association study of Alzheimer’s disease identifies disease-associated H3K27ac differences in the entorhinal cortex. Nat. Neurosci. 21, 1618–1627 (2018).
Klein, H.-U. et al. Epigenome-wide study uncovers large-scale changes in histone acetylation driven by tau pathology in aging and Alzheimer’s human brains. Nat. Neurosci. 22, 37–46 (2019).
Gasparoni, G. et al. DNA methylation analysis on purified neurons and glia dissects age and Alzheimer’s disease-specific changes in the human cortex. Epigenet. Chromatin 11, 41 (2018).
Li, P. et al. Epigenetic dysregulation of enhancers in neurons is associated with Alzheimer’s disease pathology and cognitive symptoms. Nat. Commun. 10, 2246 (2019).
Raj, T. et al. Integrative transcriptome analyses of the aging brain implicate altered splicing in Alzheimer’s disease susceptibility. Nat. Genet. 50, 1584–1592 (2018).
Nott, A. et al. Brain cell type-specific enhancer–promoter interactome maps and disease-risk association. Science 366, 1134–1139 (2019).
Kozlenkov, A. et al. Substantial DNA methylation differences between two major neuronal subtypes in human brain. Nucleic Acids Res. 44, 2593–2612 (2016).
Wang, M. et al. The Mount Sinai cohort of large-scale genomic, transcriptomic and proteomic data in Alzheimer’s disease. Sci. Data 5, 180185 (2018).
Fillenbaum, G. G. et al. Consortium to Establish a Registry for Alzheimer’s Disease (CERAD): the first twenty years. Alzheimers Dement. 4, 96–109 (2008).
Braak, H. & Braak, E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 82, 239–259 (1991).
Braak, H., Alafuzoff, I., Arzberger, T., Kretzschmar, H. & Del Tredici, K. Staging of Alzheimer disease-associated neurofibrillary pathology using paraffin sections and immunocytochemistry. Acta Neuropathol. 112, 389–404 (2006).
Morris, J. C. The Clinical Dementia Rating (CDR): current version and scoring rules. Neurology 43, 2412–2414 (1993).
Nelson, P. T. et al. Correlation of Alzheimer disease neuropathologic changes with cognitive status: a review of the literature. J. Neuropathol. Exp. Neurol. 71, 362–381 (2012).
Fullard, J. F. et al. An atlas of chromatin accessibility in the adult human brain. Genome Res. 28, 1243–1252 (2018).
Hauberg, M. E. et al. Common schizophrenia risk variants are enriched in open chromatin regions of human glutamatergic neurons. Nat. Commun. 11, 5581 (2020).
Hu, B. et al. Neuronal and glial 3D chromatin architecture informs the cellular etiology of brain disorders. Nat. Commun. 12, 3968 (2021).
Hoffman, G. E. & Schadt, E. E. variancePartition: interpreting drivers of variation in complex gene expression studies. BMC Bioinformatics 17, 483 (2016).
Fulco, C. P. et al. Activity-by-contact model of enhancer–promoter regulation from thousands of CRISPR perturbations. Nat. Genet. 51, 1664–1669 (2019).
Mifsud, B. et al. Mapping long-range promoter contacts in human cells with high-resolution capture Hi-C. Nat. Genet. 47, 598–606 (2015).
Corces, M. R. et al. Single-cell epigenomic analyses implicate candidate causal variants at inherited risk loci for Alzheimer’s and Parkinson’s diseases. Nat. Genet. 52, 1158–1168 (2020).
Hormozdiari, F. et al. Leveraging molecular quantitative trait loci to understand the genetic architecture of diseases and complex traits. Nat. Genet. 50, 1041–1047 (2018).
Pickar-Oliver, A. & Gersbach, C. A. The next generation of CRISPR–Cas technologies and applications. Nat. Rev. Mol. Cell Biol. 20, 490–507 (2019).
Wang, M. et al. Integrative network analysis of nineteen brain regions identifies molecular signatures and networks underlying selective regional vulnerability to Alzheimer’s disease. Genome Med. 8, 104 (2016).
Khan, U. A. et al. Molecular drivers and cortical spread of lateral entorhinal cortex dysfunction in preclinical Alzheimer’s disease. Nat. Neurosci. 17, 304–311 (2014).
Barrera, J. et al. Sex dependent glial-specific changes in the chromatin accessibility landscape in late-onset Alzheimer’s disease brains. Mol. Neurodegener. 16, 58 (2021).
Bowles, K. et al. 17q21.31 sub-haplotypes underlying H1-associated risk for Parkinson’s disease are associated with LRRC37A/2 expression in astrocytes. Mol. Neurodegener. 16, 1–21 (2022).
Jansen, I. E. et al. Genome-wide meta-analysis identifies new loci and functional pathways influencing Alzheimer’s disease risk. Nat. Genet. 51, 404–413 (2019).
Finucane, H. K. et al. Partitioning heritability by functional annotation using genome-wide association summary statistics. Nat. Genet. 47, 1228–1235 (2015).
Kunkle, B. W. et al. Genetic meta-analysis of diagnosed Alzheimer’s disease identifies new risk loci and implicates Aβ, tau, immunity and lipid processing. Nat. Genet. 51, 414–430 (2019).
Yokoyama, K. et al. NYAP: a phosphoprotein family that links PI3K to WAVE1 signalling in neurons. EMBO J. 30, 4739–4754 (2011).
Chen, X. et al. Cholecystokinin release triggered by NMDA receptors produces LTP and sound–sound associative memory. Proc. Natl. Acad. Sci. U.S.A. 116, 6397–6406 (2019).
Li, X., Long, J., He, T., Belshaw, R. & Scott, J. Integrated genomic approaches identify major pathways and upstream regulators in late onset Alzheimer’s disease. Sci. Rep. 5, 12393 (2015).
Castillo, E. et al. Comparative profiling of cortical gene expression in Alzheimer’s disease patients and mouse models demonstrates a link between amyloidosis and neuroinflammation. Sci. Rep. 7, 17762 (2017).
Hokama, M. et al. Altered expression of diabetes-related genes in Alzheimer’s disease brains: the Hisayama study. Cereb. Cortex 24, 2476–2488 (2014).
Chan, P. M. & Manser, E. PAKs in human disease. Prog. Mol. Biol. Transl. Sci. 106, 171–187 (2012).
Bell, R. D. & Zlokovic, B. V. Neurovascular mechanisms and blood–brain barrier disorder in Alzheimer’s disease. Acta Neuropathol. 118, 103–113 (2009).
Yuan, Z. et al. Regulation of neuronal cell death by MST1–FOXO1 signaling. J. Biol. Chem. 284, 11285–11292 (2009).
Greer, E. L. & Brunet, A. FOXO transcription factors at the interface between longevity and tumor suppression. Oncogene 24, 7410–7425 (2005).
Webb, A. E., Kundaje, A. & Brunet, A. Characterization of the direct targets of FOXO transcription factors throughout evolution. Aging Cell 15, 673–685 (2016).
Koopmans, F. et al. SynGO: an evidence-based, expert-curated knowledge base for the synapse. Neuron 103, 217–234 (2019).
Bentsen, M. et al. ATAC-seq footprinting unravels kinetics of transcription factor binding during zygotic genome activation. Nat. Commun. 11, 4267 (2020).
Rahman, M. R. et al. Network-based approach to identify molecular signatures and therapeutic agents in Alzheimer’s disease. Comput. Biol. Chem. 78, 431–439 (2019).
Acquaah-Mensah, G. K. & Taylor, R. C. Brain in situ hybridization maps as a source for reverse-engineering transcriptional regulatory networks: Alzheimer’s disease insights. Gene 586, 77–86 (2016).
Qin, L. et al. Ethnicity-specific and overlapping alterations of brain hydroxymethylome in Alzheimer’s disease. Hum. Mol. Genet. 29, 149–158 (2020).
Malik, B. R., Maddison, D. C., Smith, G. A. & Peters, O. M. Autophagic and endo-lysosomal dysfunction in neurodegenerative disease. Mol. Brain 12, 100 (2019).
Yamanaka, T. et al. Genome-wide analyses in neuronal cells reveal that upstream transcription factors regulate lysosomal gene expression. FEBS J. 283, 1077–1087 (2016).
Johnson, D. E., Ostrowski, P., Jaumouillé, V. & Grinstein, S. The position of lysosomes within the cell determines their luminal pH. J. Cell Biol. 212, 677–692 (2016).
Hayek, S. R., Rane, H. S. & Parra, K. J. Reciprocal regulation of v-ATPase and glycolytic pathway elements in health and disease. Front. Physiol. 10, 127 (2019).
Couoh-Cardel, S., Milgrom, E. & Wilkens, S. Affinity purification and structural features of the yeast vacuolar ATPase V0 membrane sector. J. Biol. Chem. 290, 27959–27971 (2015).
Delaneau, O. et al. Chromatin three-dimensional interactions mediate genetic effects on gene expression. Science 364, eaat8266 (2019).
Ambroise, C., Dehman, A., Neuvial, P., Rigaill, G. & Vialaneix, N. Adjacency-constrained hierarchical clustering of a band similarity matrix with application to genomics. Algorithms Mol. Biol. 14, 22 (2019).
Hoffman, G. E., Bendl, J., Girdhar, K. & Roussos, P. decorate: differential epigenetic correlation test. Bioinformatics 36, 2856–2861 (2020).
Rao, S. S. P. et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 159, 1665–1680 (2014).
Lieberman-Aiden, E. et al. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science 326, 289–293 (2009).
De Jager, P. L. et al. A multi-omic atlas of the human frontal cortex for aging and Alzheimer’s disease research. Sci. Data 5, 180142 (2018).
Meuleman, W. et al. Constitutive nuclear lamina–genome interactions are highly conserved and associated with A/T-rich sequence. Genome Res. 23, 270–280 (2013).
Roadmap Epigenomics Consortiumet al. Integrative analysis of 111 reference human epigenomes. Nature 518, 317–330 (2015).
Corces, M.R. et al. The chromatin accessibility landscape of primary human cancers. Science 362, eaav1898 (2018).
Ishizuka, Y. & Hanamura, K. Drebrin in Alzheimer’s disease. Adv. Exp. Med. Biol. 1006, 203–223 (2017).
Turi, Z., Lacey, M., Mistrik, M. & Moudry, P. Impaired ribosome biogenesis: mechanisms and relevance to cancer and aging. Aging 11, 2512–2540 (2019).
Chambers, D. M., Peters, J. & Abbott, C. M. The lethal mutation of the mouse wasted (wst) is a deletion that abolishes expression of a tissue-specific isoform of translation elongation factor 1α, encoded by the Eef1a2 gene. Proc. Natl. Acad. Sci. U.S.A. 95, 4463–4468 (1998).
Zeisel, A. et al. Cell types in the mouse cortex and hippocampus revealed by single-cell RNA-seq. Science 347, 1138–1142 (2015).
Zhang, W. et al. Integrative transcriptome imputation reveals tissue-specific and shared biological mechanisms mediating susceptibility to complex traits. Nat. Commun. 10, 3834 (2019).
Ernst, J. & Kellis, M. Chromatin-state discovery and genome annotation with ChromHMM. Nat. Protoc. 12, 2478–2492 (2017).
Meuleman, W. et al. Index and biological spectrum of human DNase I hypersensitive sites. Nature 584, 244–251 (2020).
Dong, P. et al. Population-level variation of enhancer expression identifies novel disease mechanisms in the human brain. Nat. Genet. https://doi.org/10.1038/s41588-022-01170-4 (2022).
Ernst, J. & Kellis, M. Large-scale imputation of epigenomic datasets for systematic annotation of diverse human tissues. Nat. Biotechnol. 33, 364–376 (2015).
Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).
Hoffman, G. E. & Roussos, P. Dream: powerful differential expression analysis for repeated measures designs. Bioinformatics 37, 192–201 (2021).
de Leeuw, C. A., Mooij, J. M., Heskes, T. & Posthuma, D. MAGMA: generalized gene-set analysis of GWAS data. PLoS Comput. Biol. 11, e1004219 (2015).
Haroutunian, V., Katsel, P. & Schmeidler, J. Transcriptional vulnerability of brain regions in Alzheimer’s disease and dementia. Neurobiol. Aging 30, 561–573 (2009).
Morris, J. C. et al. The Consortium to Establish a Registry for Alzheimer’s Disease (CERAD). Part I. Clinical and neuropsychological assessment of Alzheimer’s disease. Neurology 39, 1159–1165 (1989).
Buenrostro, J. D., Wu, B., Chang, H. Y. & Greenleaf, W. J. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr. Protoc. Mol. Biol. 109, 21.29.1–21.29.9 (2015).
Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).
Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).
McKenna, A. et al. The genome analysis toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 20, 1297–1303 (2010).
Manichaikul, A. et al. Robust relationship inference in genome-wide association studies. Bioinformatics 26, 2867–2873 (2010).
Servant, N. et al. HiC-Pro: an optimized and flexible pipeline for Hi-C data processing. Genome Biol. 16, 259 (2015).
Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).
Durand, N. C. et al. Juicer provides a one-click system for analyzing loop-resolution Hi-C experiments. Cell Syst. 3, 95–98 (2016).
Shin, H. et al. TopDom: an efficient and deterministic method for identifying topological domains in genomes. Nucleic Acids Res. 44, e70 (2016).
Yu, G., Wang, L.-G. & He, Q.-Y. ChIPseeker: an R/Bioconductor package for ChIP peak annotation, comparison and visualization. Bioinformatics 31, 2382–2383 (2015).
Lawrence, M. et al. Software for computing and annotating genomic ranges. PLoS Comput. Biol. 9, e1003118 (2013).
Zhang, Y. et al. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J. Neurosci. 34, 11929–11947 (2014).
Liberzon, A. et al. Molecular Signatures Database (MSigDB) 3.0. Bioinformatics 27, 1739–1740 (2011).
McLean, C. Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).
Law, C.W. et al. RNA-seq analysis is easy as 1-2-3 with limma, Glimma and edgeR. F1000Res 5, 1408 (2016).
Yoshida, H. et al. The cis-regulatory atlas of the mouse immune system. Cell 176, 897–912 (2019).
Anscombe, F. J. The transformation of Poisson, binomial and negative-binomial data. Biometrika 35, 246–254 (1948).
Francis Harrison, P. Varistran: Anscombe’s variance stabilizing transformation for RNA-seq gene expression data. JOSS 2, 257 (2017).
Gandal, M. J. et al. Transcriptome-wide isoform-level dysregulation in ASD, schizophrenia, and bipolar disorder. Science 362, eaat8127 (2018).
Nagpal, S. et al. TIGAR: an improved Bayesian tool for transcriptomic data imputation enhances gene mapping of complex traits. Am. J. Hum. Genet. 105, 258–266 (2019).
Becker, M., Kuhse, J. & Kirsch, J. Effects of two elongation factor 1A isoforms on the formation of gephyrin clusters at inhibitory synapses in hippocampal neurons. Histochem. Cell Biol. 140, 603–609 (2013).
Shirao, T. et al. The role of drebrin in neurons. J. Neurochem. 141, 819–834 (2017).
Hetman, M. & Slomnicki, L. P. Ribosomal biogenesis as an emerging target of neurodevelopmental pathologies. J. Neurochem. 148, 325–347 (2019).
Schrode, N. et al. Synergistic effects of common schizophrenia risk variants. Nat. Genet. 51, 1475–1485 (2019).
Weirauch, M. T. et al. Determination and inference of eukaryotic transcription factor sequence specificity. Cell 158, 1431–1443 (2014).
Gupta, S., Stamatoyannopoulos, J. A., Bailey, T. L. & Noble, W. S. Quantifying similarity between motifs. Genome Biol. 8, R24 (2007).
Reyna, M. A., Leiserson, M. D. M. & Raphael, B. J. Hierarchical HotNet: identifying hierarchies of altered subnetworks. Bioinformatics 34, i972–i980 (2018).
Acknowledgements
We thank the patients and families who donated material for these studies. We thank the members of the Roussos laboratory for thoughtful advice and critique and the computational resources and staff expertise provided by the Scientific Computing at the Icahn School of Medicine at Mount Sinai. This study was supported by grants from the National Institute on Aging, NIH grants R01-AG067025 (P.R. and V.H.), R01-AG065582 (P.R. and V.H.) and R01-AG050986 (P.R.) and by grants from the National Institute of Mental Health, NIH grants, R56-MH101454 (K.J.B.), R01-MH106056 (P.R. and K.J.B.), R01-MH109897 (P.R. and K.J.B.) and R01-MH121074 (K.J.B.). J.B. was supported in part by Alzheimer’s Association Research Fellowship AARF-21-722200. K.G. was supported in part by Alzheimer’s Association Research Fellowship AARF-21-722582. G.E.H. and P.D. were supported in part by NARSAD Young Investigator grants 26313 and 29683, respectively, from the Brain & Behavior Research Foundation. S.P.K. is a recipient of an NIH LRP award. Research reported in this paper was supported by the Office of Research Infrastructure of the National Institutes of Health under award numbers S10OD018522 and S10OD026880. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
Author information
Authors and Affiliations
Contributions
R.A.N., V.H. and P.R. conceived of and designed the project. V.H. provided human brain tissue. J.F.F. and P.R. designed experimental strategies for epigenome profiling in human postmortem tissue. R.M., S.K., S.M.R. and J.F.F. performed ATAC-seq data generation. S.R. performed Hi-C data generation. P.A. performed ChIP–seq data generation. E.I., J.V. and R.A.N. performed USF2 in vitro validation studies. M.B.F., K.G.T., J.V., S.R. and K.J.B. performed the CRISPRi in vitro validation studies. J.B., M.E.H., K.G., G.E.H. and P.R. designed analytical strategies. J.B., M.E.H., K.G. and G.E.H. conducted initial bioinformatics sample processing and quality control. J.B., M.E.H., K.G. and G.E.H. developed and performed all downstream omics data analyses and interpreted results. B.Z. performed eQTL fine-mapping analysis. W.Z. and G.V. performed transcriptome imputation analysis. P.D. performed ChIP–seq and Hi-C data analysis. J.B., M.E.H., K.G., G.E.H., J.F.F. and P.R. wrote the manuscript with the help of all authors. G.E.H., J.F.F. and P.R. supervised overall data generation and analysis.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Neuroscience thanks the anonymous reviewers for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary information
Supplementary Information
Supplementary Methods. Supplementary Figures 1–35 and Supplementary Note.
Supplementary Tables
Supplementary Tables 1–19.
Rights and permissions
About this article
Cite this article
Bendl, J., Hauberg, M.E., Girdhar, K. et al. The three-dimensional landscape of cortical chromatin accessibility in Alzheimer’s disease. Nat Neurosci 25, 1366–1378 (2022). https://doi.org/10.1038/s41593-022-01166-7
Received:
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/s41593-022-01166-7
This article is cited by
-
Chromatin accessibility: biological functions, molecular mechanisms and therapeutic application
Signal Transduction and Targeted Therapy (2024)
-
Single cell transcriptomes and multiscale networks from persons with and without Alzheimer’s disease
Nature Communications (2024)
-
Mechanisms of autophagy–lysosome dysfunction in neurodegenerative diseases
Nature Reviews Molecular Cell Biology (2024)
-
Usf2 Deficiency Promotes Autophagy to Alleviate Cerebral Ischemia-Reperfusion Injury Through Suppressing YTHDF1-m6A-Mediated Cdc25A Translation
Molecular Neurobiology (2024)
-
A multi-region single nucleus transcriptomic atlas of Parkinson’s disease
Scientific Data (2024)