https://spdx.org/licenses/CC0-1.0.htmlhttps://spdx.org/licenses/CC0-1.0.html
The human adult intestinal system is a complex organ that is approximately 9 meters long and performs a variety of complex functions including digestion, nutrient absorption, and immune surveillance. We performed snRNA-seq on 8 regions of of the human intestine (duodenum, proximal-jejunum, mid-jejunum, ileum, ascending colon, transverse colon, descending colon, and sigmoid colon) from 9 donors (B001, B004, B005, B006, B008, B009, B010, B011, and B012). In the corresponding paper, we find cell compositions differ dramatically across regions of the intestine and demonstrate the complexity of epithelial subtypes. We map gene regulatory differences in these cells suggestive of a regulatory differentiation cascade, and associate intestinal disease heritability with specific cell types. These results describe the complexity of the cell composition, regulation, and organization in the human intestine, and serve as an important reference map for understanding human biology and disease. Methods For a detailed description of each of the steps to obtain this data see the detailed materials and methods in the associated manuscript. Briefly, intestine pieces from 8 different sites across the small intestine and colon were flash frozen. Nuclei were isolated from each sample and the resulting nuclei were processed with either 10x scRNA-seq using Chromium Next GEM Single Cell 3’ Reagent Kits v3.1 (10x Genomics, 1000121) or Chromium Next GEM Chip G Single Cell Kits (10x Genomics, 1000120) or 10x multiome sequencing using Chromium Next GEM Single Cell Multiome ATAC + Gene Expression Kits (10x Genomics, 1000283). Initial processing of snRNA-seq data was done with the Cell Ranger Pipeline (https://support.10xgenomics.com/single-cell-gene-expression/software/pipelines/latest/what-is-cell-ranger) by first running cellranger mkfastq to demultiplex the bcl files and then running cellranger count. Since nuclear RNA was sequenced, data were aligned to a pre-mRNA reference. Initial processing of the mutiome data, including alignment and generation of fragments files and expression matrices, was performed with the Cell Ranger ARC Pipeline. The raw expression matrices from these pipelines are included here. Downstream processing was performed in R, using the Seurat package.
https://spdx.org/licenses/CC0-1.0.htmlhttps://spdx.org/licenses/CC0-1.0.html
Transcribed enhancer maps can reveal nuclear interactions underpinning each cell type and connect specific cell types to diseases. Using a 5′ single-cell RNA sequencing approach, we defined transcription start sites of enhancer RNAs and other classes of coding and non-coding RNAs in human CD4+ T cells, revealing cellular heterogeneity and differentiation trajectories. Integration of these datasets with single-cell chromatin profiles showed that active enhancers with bidirectional RNA transcription are highly cell type–specific, and disease heritability is strongly enriched in these enhancers. The resulting cell type–resolved multimodal atlas of bidirectionally transcribed enhancers, which we linked with promoters using fine-scale chromatin contact maps, enabled us to systematically interpret genetic variants associated with a range of immune-mediated diseases. Methods All experiments using human samples were approved by the ethical review committee of RIKEN [approval no. H30-9(13)]. Written informed consent was obtained from all donors. CD4+ T cells were isolated by the immunomagnetic negative selection method. Stained CD4+ T cells were sorted using a FACSAria IIu Cell Sorter (BD Biosciences). Human CD4+ T cells and FACS-sorted heterogenous populations were processed with a Chromium Next GEM Single Cell 5′ kit (10x Genomics). Libraries were sequenced on an Illumina NovaSeq 6000 sequencing platform using 2 × 150 bp paired-end sequencing. Multiome assay (10x Genomics) was performed according to the manufacturer’s instructions. Multiome libraries were pooled and sequenced as above with 10 cycles for i7 index and 24 cycles for i5 index. Micro-C libraries were generated using a Dovetail Micro-C Kit (Cantata Bio, Cat#21006) and were sequenced on an Illumina NovaSeq 6000 platform using 2 × 150 bp paired-end sequencing. Chromium scRNA-seq, snRNA-seq, and CITE-seq data were processed using Cell Ranger Software version 5.0.1 (10x Genomics) and R package Seurat version 5 (4.9.9.9067). Multiome data were processed by Cell Ranger ARC version 2.0.0 (10x Genomics), Seurat version 5 (4.9.9.9067), and Signac version 1.10.0. scRNA-seq, snRNA-seq, and Multiome 3′ snRNA-seq data were integrated using canonical correlation analysis. snATAC peaks were identified from fragment files of each cluster using MACS2 version 2.2.6 with default settings as implemented in Signac version 1.10.0. For ReapTEC, paired-end reads were mapped again using STAR (STARsolo) to obtain reads with unencoded G, which was tagged as a soft-clipped G by STARsolo. Reads were deduplicated, and those with the barcodes of each cell type were extracted. A count file was generated for each transcription start site (TSS) using the “bamToBed” function in BEDTools version 2.30.0. TSS peaks were generated by merging TSSs located within 10 bp of each other. To identify btcEnhs, TSS peak pairs were detected using scripts provided at https://github.com/anderssonrobin/enhancers/blob/master/scripts/bidir_enhancers with minor modifications. Micro-C data were processed with the dovetail_tools pipeline (Cantata Bio). Chromatin loop contacts were identified by the HiCCUPS algorithm using the Juicer Tools package version 2.20.0 and the scale-space representation algorithm using the Mustache package. Loops were called at a 1-kb resolution with SCALE-normalized contact matrices for HiCCUPS and with ICE-normalized contact matrices for Mustache, and were filtered for an FDR < 0.05.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
In the context of the Human Cell Atlas, we have created a single-cell-driven taxonomy of cell types and states in human tonsils. This repository contains the Seurat objects derived from this effort. In particular, we have datasets for each modality (scRNA-seq, scATAC-seq, CITE-seq, spatial transcriptomics), as well as cell type-specific datasets. Most importantly, this is the input that we used to create the HCATonsilData package, which allows programmatic access to all this datasets within R.
Version 2 of this repository includes cells from 7 additional donors, which we used as a validation cohort to validate the cell types and states defined in the atlas. In addition, in this version we also provide the Seurat object associated with the spatial transcriptomics data (10X Visium), as well as the fragments files for scATAC-seq and Multiome
Not seeing a result you expected?
Learn how you can add new datasets to our index.
https://spdx.org/licenses/CC0-1.0.htmlhttps://spdx.org/licenses/CC0-1.0.html
The human adult intestinal system is a complex organ that is approximately 9 meters long and performs a variety of complex functions including digestion, nutrient absorption, and immune surveillance. We performed snRNA-seq on 8 regions of of the human intestine (duodenum, proximal-jejunum, mid-jejunum, ileum, ascending colon, transverse colon, descending colon, and sigmoid colon) from 9 donors (B001, B004, B005, B006, B008, B009, B010, B011, and B012). In the corresponding paper, we find cell compositions differ dramatically across regions of the intestine and demonstrate the complexity of epithelial subtypes. We map gene regulatory differences in these cells suggestive of a regulatory differentiation cascade, and associate intestinal disease heritability with specific cell types. These results describe the complexity of the cell composition, regulation, and organization in the human intestine, and serve as an important reference map for understanding human biology and disease. Methods For a detailed description of each of the steps to obtain this data see the detailed materials and methods in the associated manuscript. Briefly, intestine pieces from 8 different sites across the small intestine and colon were flash frozen. Nuclei were isolated from each sample and the resulting nuclei were processed with either 10x scRNA-seq using Chromium Next GEM Single Cell 3’ Reagent Kits v3.1 (10x Genomics, 1000121) or Chromium Next GEM Chip G Single Cell Kits (10x Genomics, 1000120) or 10x multiome sequencing using Chromium Next GEM Single Cell Multiome ATAC + Gene Expression Kits (10x Genomics, 1000283). Initial processing of snRNA-seq data was done with the Cell Ranger Pipeline (https://support.10xgenomics.com/single-cell-gene-expression/software/pipelines/latest/what-is-cell-ranger) by first running cellranger mkfastq to demultiplex the bcl files and then running cellranger count. Since nuclear RNA was sequenced, data were aligned to a pre-mRNA reference. Initial processing of the mutiome data, including alignment and generation of fragments files and expression matrices, was performed with the Cell Ranger ARC Pipeline. The raw expression matrices from these pipelines are included here. Downstream processing was performed in R, using the Seurat package.