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

2019 ◽  
Author(s):  
Vivek Rai ◽  
Daniel X. Quang ◽  
Michael R. Erdos ◽  
Darren A. Cusanovich ◽  
Riza M. Daza ◽  
...  

ABSTRACTObjectiveType 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 the 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, and 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.MethodsWe 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 the U-Net architecture to accurately predict open chromatin peak calls in rare cell populations.ResultsWe 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, we find that T2D GWAS SNPs are significantly enriched in beta cell-specific and cross 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.ConclusionsCollectively, 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.

2020 ◽  
Vol 32 ◽  
pp. 109-121 ◽  
Author(s):  
Vivek Rai ◽  
Daniel X. Quang ◽  
Michael R. Erdos ◽  
Darren A. Cusanovich ◽  
Riza M. Daza ◽  
...  

2016 ◽  
Vol 24 (4) ◽  
pp. 593-607 ◽  
Author(s):  
Åsa Segerstolpe ◽  
Athanasia Palasantza ◽  
Pernilla Eliasson ◽  
Eva-Marie Andersson ◽  
Anne-Christine Andréasson ◽  
...  

2020 ◽  
Author(s):  
Agata Wesolowska-Andersen ◽  
Grace Zhuo Yu ◽  
Vibe Nylander ◽  
Fernando Abaitua ◽  
Matthias Thurner ◽  
...  

2019 ◽  
Vol 9 (1) ◽  
Author(s):  
Madhusudhan Bysani ◽  
Rasmus Agren ◽  
Cajsa Davegårdh ◽  
Petr Volkov ◽  
Tina Rönn ◽  
...  

Phenomics ◽  
2021 ◽  
Author(s):  
Kaixuan Bao ◽  
Zhicheng Cui ◽  
Hui Wang ◽  
Hui Xiao ◽  
Ting Li ◽  
...  

eLife ◽  
2020 ◽  
Vol 9 ◽  
Author(s):  
Agata Wesolowska-Andersen ◽  
Grace Zhuo Yu ◽  
Vibe Nylander ◽  
Fernando Abaitua ◽  
Matthias Thurner ◽  
...  

Genome-wide association analyses have uncovered multiple genomic regions associated with T2D, but identification of the causal variants at these remains a challenge. There is growing interest in the potential of deep learning models - which predict epigenome features from DNA sequence - to support inference concerning the regulatory effects of disease-associated variants. Here, we evaluate the advantages of training convolutional neural network (CNN) models on a broad set of epigenomic features collected in a single disease-relevant tissue – pancreatic islets in the case of type 2 diabetes (T2D) - as opposed to models trained on multiple human tissues. We report convergence of CNN-based metrics of regulatory function with conventional approaches to variant prioritization – genetic fine-mapping and regulatory annotation enrichment. We demonstrate that CNN-based analyses can refine association signals at T2D-associated loci and provide experimental validation for one such signal. We anticipate that these approaches will become routine in downstream analyses of GWAS.


2016 ◽  
Vol 27 (2) ◽  
pp. 208-222 ◽  
Author(s):  
Nathan Lawlor ◽  
Joshy George ◽  
Mohan Bolisetty ◽  
Romy Kursawe ◽  
Lili Sun ◽  
...  

2020 ◽  
Vol 10 (1) ◽  
Author(s):  
Madhusudhan Bysani ◽  
Rasmus Agren ◽  
Cajsa Davegårdh ◽  
Petr Volkov ◽  
Tina Rönn ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document