Single-cell anatomical analysis of human hippocampus and entorhinal cortex uncovers early-stage molecular pathology in Alzheimer’s disease

The human hippocampal formation plays a central role in Alzheimer’s disease (AD) progression, cognitive traits, and the onset of dementia; yet its molecular states in AD remain uncharacterized. Here, we report a comprehensive single-cell transcriptomic dissection of the human hippocampus and entorhinal cortex across 489,558 cells from 65 individuals with varying stages of AD pathology. We transcriptionally characterize major brain cell types and neuronal classes, including 17 glutamatergic and 8 GABAergic neuron subpopulations. Combining evidence from human and mouse tissue-microdissection, neuronal cell isolation and spatial transcriptomics, we show that single-cell expression patterns capture fine-resolution neuronal anatomical topography. By stratifying subjects into early and late pathology groups, we uncover stage-dependent and cell-type specific transcriptional modules altered during AD progression. These include early-stage cell-type specific dysregulation of cellular and cholesterol metabolism, late-stage neuron-glia alterations in neurotransmission, and late-stage signatures of cellular stress, apoptosis, and DNA damage broadly shared across cell types. Late-stage signatures show signs of convergence in hippocampal and cortical cells, while early changes diverge; highlighting the relevance of characterizing molecular pathology across brain regions and AD progression. Finally, we characterize neuron subregion-specific responses to AD pathology and show that CA1 pyramidal neurons are the most transcriptionally altered while CA3 and dentate gyrus granule neurons the least. Our study provides a valuable resource to extend cell type-specific studies of AD to clinically relevant brain regions affected early by pathology in disease progression.

[1]  D. Bennett,et al.  Neuronal ApoE upregulates MHC-I expression to drive selective neurodegeneration in Alzheimer’s disease , 2021, Nature Neuroscience.

[2]  J. Davila-Velderrain,et al.  Single-cell dissection of schizophrenia reveals neurodevelopmental-synaptic axis and transcriptional resilience , 2020, medRxiv.

[3]  Liang He,et al.  NEBULA: a fast negative binomial mixed model for differential expression and co-expression analyses of large-scale multi-subject single-cell data , 2020, bioRxiv.

[4]  J. Vilo,et al.  gprofiler2 -- an R package for gene list functional enrichment analysis and namespace conversion toolset g:Profiler , 2020, F1000Research.

[5]  James A. Eddy,et al.  Meta-Analysis of the Alzheimer’s Disease Human Brain Transcriptome and Functional Dissection in Mouse Models , 2020, Cell reports.

[6]  Matheus B. Victor,et al.  Reconstruction of the human blood–brain barrier in vitro reveals a pathogenic mechanism of APOE4 in pericytes , 2020, Nature Medicine.

[7]  Jamie L. Marshall,et al.  Disease-associated astrocytes in Alzheimer’s disease and aging , 2020, Nature Neuroscience.

[8]  A. Fagan,et al.  APOE4 leads to blood-brain barrier dysfunction predicting cognitive decline , 2020, Nature.

[9]  Hongkui Zeng,et al.  A taxonomy of transcriptomic cell types across the isocortex and hippocampal formation , 2020, Cell.

[10]  J. Arellano,et al.  Molecular Diversity Among Adult Human Hippocampal and Entorhinal Cells , 2020, bioRxiv.

[11]  F. Sanz,et al.  The DisGeNET knowledge platform for disease genomics: 2019 update , 2019, Nucleic Acids Res..

[12]  John F. Ouyang,et al.  A single-cell atlas of entorhinal cortex from individuals with Alzheimer’s disease reveals cell-type-specific gene expression regulation , 2019, Nature Neuroscience.

[13]  D. Holtzman,et al.  Alzheimer Disease: An Update on Pathobiology and Treatment Strategies , 2019, Cell.

[14]  Joakim Lundeberg,et al.  Molecular atlas of the adult mouse brain , 2019, Science Advances.

[15]  Jose Davila-Velderrain,et al.  A multiresolution framework to characterize single-cell state landscapes , 2019, Nature Communications.

[16]  Richard Reynolds,et al.  Neuronal vulnerability and multilineage diversity in multiple sclerosis , 2019, Nature.

[17]  Maximilian Haeussler,et al.  Single-cell genomics identifies cell type–specific molecular changes in autism , 2019, Science.

[18]  Manolis Kellis,et al.  Single-cell transcriptomic analysis of Alzheimer’s disease , 2019, Nature.

[19]  Oscar Franzén,et al.  PanglaoDB: a web server for exploration of mouse and human single-cell RNA sequencing data , 2019, Database J. Biol. Databases Curation.

[20]  Dheeraj Malhotra,et al.  Altered human oligodendrocyte heterogeneity in multiple sclerosis , 2019, Nature.

[21]  Bradley T. Hyman,et al.  Beyond the neuron–cellular interactions early in Alzheimer disease pathogenesis , 2019, Nature Reviews Neuroscience.

[22]  Timothy J. Hohman,et al.  Genome-wide meta-analysis identifies new loci and functional pathways influencing Alzheimer’s disease risk , 2019, Nature Genetics.

[23]  P. Greengard,et al.  Selective Neuronal Vulnerability in Alzheimer’s Disease: A Network-Based Analysis , 2018, Neuron.

[24]  Prashant S. Emani,et al.  Comprehensive functional genomic resource and integrative model for the human brain , 2018, Science.

[25]  Hans-Ulrich Klein,et al.  Epigenome-wide study uncovers large-scale changes in histone acetylation driven by tau pathology in the aging and Alzheimer human brain , 2018, Nature Neuroscience.

[26]  Fan Zhang,et al.  Fast, sensitive, and accurate integration of single cell data with Harmony , 2018, bioRxiv.

[27]  S. Beck,et al.  A histone acetylome-wide association study of Alzheimer’s disease identifies disease-associated H3K27ac differences in the entorhinal cortex , 2018, Nature Neuroscience.

[28]  Vincent A. Traag,et al.  From Louvain to Leiden: guaranteeing well-connected communities , 2018, Scientific Reports.

[29]  Bin Zhang,et al.  Integrative transcriptome analyses of the aging brain implicate altered splicing in Alzheimer’s disease susceptibility , 2018, Nature Genetics.

[30]  John Hardy,et al.  Selective vulnerability in neurodegenerative diseases , 2018, Nature Neuroscience.

[31]  Evan Z. Macosko,et al.  Molecular Diversity and Specializations among the Cells of the Adult Mouse Brain , 2018, Cell.

[32]  J. Dorszewska,et al.  Alzheimer's Disease - The 21st Century Challenge , 2018 .

[33]  David A Bennett,et al.  Religious Orders Study and Rush Memory and Aging Project. , 2018, Journal of Alzheimer's disease : JAD.

[34]  Fabian J Theis,et al.  SCANPY: large-scale single-cell gene expression data analysis , 2018, Genome Biology.

[35]  Allan R. Jones,et al.  Shared and distinct transcriptomic cell types across neocortical areas , 2017, bioRxiv.

[36]  Yue-Ming Li,et al.  The role of astrocytes in amyloid production and Alzheimer's disease , 2017, Open Biology.

[37]  A. Regev,et al.  Temporal Tracking of Microglia Activation in Neurodegeneration at Single-Cell Resolution , 2017, Cell reports.

[38]  Aviv Regev,et al.  Massively-parallel single nucleus RNA-seq with DroNc-seq , 2017, Nature Methods.

[39]  Anastasia G. Efthymiou,et al.  Late onset Alzheimer’s disease genetics implicates microglial pathways in disease risk , 2017, Molecular Neurodegeneration.

[40]  Anastasia G. Efthymiou,et al.  Late onset Alzheimer’s disease genetics implicates microglial pathways in disease risk , 2017, Molecular Neurodegeneration.

[41]  B. Wieringa,et al.  The SH-SY5Y cell line in Parkinson’s disease research: a systematic review , 2017, Molecular Neurodegeneration.

[42]  Andrew D. Rouillard,et al.  The harmonizome: a collection of processed datasets gathered to serve and mine knowledge about genes and proteins , 2016, Database J. Biol. Databases Curation.

[43]  Andrew D. Rouillard,et al.  Enrichr: a comprehensive gene set enrichment analysis web server 2016 update , 2016, Nucleic Acids Res..

[44]  Nelson Spruston,et al.  Hipposeq: a comprehensive RNA-seq database of gene expression in hippocampal principal neurons , 2016, eLife.

[45]  Eric Karran,et al.  The Cellular Phase of Alzheimer’s Disease , 2016, Cell.

[46]  S. Linnarsson,et al.  Cell types in the mouse cortex and hippocampus revealed by single-cell RNA-seq , 2015, Science.

[47]  Manolis Kellis,et al.  Conserved epigenomic signals in mice and humans reveal immune basis of Alzheimer’s disease , 2015, Nature.

[48]  H. Braak,et al.  Neuroanatomy and Pathology of Sporadic Alzheimer's Disease , 2014 .

[49]  S. Horvath,et al.  Genes and pathways underlying regional and cell type changes in Alzheimer's disease , 2013, Genome Medicine.

[50]  Mikko Niemi,et al.  Genetics is a major determinant of expression of the human hepatic uptake transporter OATP1B1, but not of OATP1B3 and OATP2B1 , 2013, Genome Medicine.

[51]  Allan R. Jones,et al.  An anatomically comprehensive atlas of the adult human brain transcriptome , 2012, Nature.

[52]  E. Walker,et al.  Diagnostic and Statistical Manual of Mental Disorders , 2013 .

[53]  W Noble,et al.  Astrocytes are important mediators of Aβ-induced neurotoxicity and tau phosphorylation in primary culture , 2011, Cell Death and Disease.

[54]  J. Morris,et al.  Alzheimer’s Disease: The Challenge of the Second Century , 2011, Science Translational Medicine.

[55]  J. Schneider,et al.  Neuropathology of older persons without cognitive impairment from two community-based studies , 2006, Neurology.

[56]  By Michael Marron-stearns 2019 Update : What to , 2019 .

[57]  Andrea Klug,et al.  The Hippocampus Book , 2016 .

[58]  G. Arbanas Diagnostic and Statistical Manual of Mental Disorders (DSM-5) , 2015 .