Skip to main content

Single-cell ATAC-Seq in human pancreatic islets and deep learning upscaling of rare cells reveals cell-specific type 2 diabetes regulatory signatures.

Citation
Rai, V., et al. “Single-Cell Atac-Seq In Human Pancreatic Islets And Deep Learning Upscaling Of Rare Cells Reveals Cell-Specific Type 2 Diabetes Regulatory Signatures.”. Molecular Metabolism, pp. 109-121.
Center University of Michigan
Author Vivek Rai, Daniel X Quang, Michael R Erdos, Darren A Cusanovich, Riza M Daza, Narisu Narisu, Luli S Zou, John P Didion, Yuanfang Guan, Jay Shendure, Stephen C J Parker, Francis S Collins
Keywords chromatin, Deep learning, Epigenomics, islet, single cell, type 2 diabetes
Abstract

OBJECTIVE: Type 2 diabetes (T2D) is a complex disease characterized by pancreatic islet dysfunction, insulin resistance, and disruption of blood glucose levels. Genome-wide association studies (GWAS) have identified > 400 independent signals that encode genetic predisposition. More than 90% of associated single-nucleotide polymorphisms (SNPs) localize to non-coding regions and are enriched in chromatin-defined islet enhancer elements, indicating a strong transcriptional regulatory component to disease susceptibility. Pancreatic islets are a mixture of cell types that express distinct hormonal programs, so each cell type may contribute differentially to the underlying regulatory processes that modulate T2D-associated transcriptional circuits. Existing chromatin profiling methods such as ATAC-seq and DNase-seq, applied to islets in bulk, produce aggregate profiles that mask important cellular and regulatory heterogeneity.

METHODS: We present genome-wide single-cell chromatin accessibility profiles in >1,600 cells derived from a human pancreatic islet sample using single-cell combinatorial indexing ATAC-seq (sci-ATAC-seq). We also developed a deep learning model based on U-Net architecture to accurately predict open chromatin peak calls in rare cell populations.

RESULTS: We show that sci-ATAC-seq profiles allow us to deconvolve alpha, beta, and delta cell populations and identify cell-type-specific regulatory signatures underlying T2D. Particularly, T2D GWAS SNPs are significantly enriched in beta cell-specific and across cell-type shared islet open chromatin, but not in alpha or delta cell-specific open chromatin. We also demonstrate, using less abundant delta cells, that deep learning models can improve signal recovery and feature reconstruction of rarer cell populations. Finally, we use co-accessibility measures to nominate the cell-specific target genes at 104 non-coding T2D GWAS signals.

CONCLUSIONS: Collectively, we identify the islet cell type of action across genetic signals of T2D predisposition and provide higher-resolution mechanistic insights into genetically encoded risk pathways.

Year of Publication
2020
Journal
Molecular metabolism
Volume
32
Number of Pages
109-121
Date Published
12/2020
ISSN Number
2212-8778
DOI
10.1016/j.molmet.2019.12.006
Alternate Journal
Mol Metab
PMID
32029221
PMCID
PMC6961712
Download citation