15 datasets found
  1. n

    Data from: Large-scale integration of single-cell transcriptomic data...

    • data.niaid.nih.gov
    • dataone.org
    • +1more
    zip
    Updated Dec 14, 2021
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    David McKellar; Iwijn De Vlaminck; Benjamin Cosgrove (2021). Large-scale integration of single-cell transcriptomic data captures transitional progenitor states in mouse skeletal muscle regeneration [Dataset]. http://doi.org/10.5061/dryad.t4b8gtj34
    Explore at:
    zipAvailable download formats
    Dataset updated
    Dec 14, 2021
    Dataset provided by
    Cornell University
    Authors
    David McKellar; Iwijn De Vlaminck; Benjamin Cosgrove
    License

    https://spdx.org/licenses/CC0-1.0.htmlhttps://spdx.org/licenses/CC0-1.0.html

    Description

    Skeletal muscle repair is driven by the coordinated self-renewal and fusion of myogenic stem and progenitor cells. Single-cell gene expression analyses of myogenesis have been hampered by the poor sampling of rare and transient cell states that are critical for muscle repair, and do not inform the spatial context that is important for myogenic differentiation. Here, we demonstrate how large-scale integration of single-cell and spatial transcriptomic data can overcome these limitations. We created a single-cell transcriptomic dataset of mouse skeletal muscle by integration, consensus annotation, and analysis of 23 newly collected scRNAseq datasets and 88 publicly available single-cell (scRNAseq) and single-nucleus (snRNAseq) RNA-sequencing datasets. The resulting dataset includes more than 365,000 cells and spans a wide range of ages, injury, and repair conditions. Together, these data enabled identification of the predominant cell types in skeletal muscle, and resolved cell subtypes, including endothelial subtypes distinguished by vessel-type of origin, fibro/adipogenic progenitors defined by functional roles, and many distinct immune populations. The representation of different experimental conditions and the depth of transcriptome coverage enabled robust profiling of sparsely expressed genes. We built a densely sampled transcriptomic model of myogenesis, from stem cell quiescence to myofiber maturation and identified rare, transitional states of progenitor commitment and fusion that are poorly represented in individual datasets. We performed spatial RNA sequencing of mouse muscle at three time points after injury and used the integrated dataset as a reference to achieve a high-resolution, local deconvolution of cell subtypes. We also used the integrated dataset to explore ligand-receptor co-expression patterns and identify dynamic cell-cell interactions in muscle injury response. We provide a public web tool to enable interactive exploration and visualization of the data. Our work supports the utility of large-scale integration of single-cell transcriptomic data as a tool for biological discovery.

    Methods Mice. The Cornell University Institutional Animal Care and Use Committee (IACUC) approved all animal protocols, and experiments were performed in compliance with its institutional guidelines. Adult C57BL/6J mice (mus musculus) were obtained from Jackson Laboratories (#000664; Bar Harbor, ME) and were used at 4-7 months of age. Aged C57BL/6J mice were obtained from the National Institute of Aging (NIA) Rodent Aging Colony and were used at 20 months of age. For new scRNAseq experiments, female mice were used in each experiment.

    Mouse injuries and single-cell isolation. To induce muscle injury, both tibialis anterior (TA) muscles of old (20 months) C57BL/6J mice were injected with 10 µl of notexin (10 µg/ml; Latoxan; France). At 0, 1, 2, 3.5, 5, or 7 days post-injury (dpi), mice were sacrificed and TA muscles were collected and processed independently to generate single-cell suspensions. Muscles were digested with 8 mg/ml Collagenase D (Roche; Switzerland) and 10 U/ml Dispase II (Roche; Switzerland), followed by manual dissociation to generate cell suspensions. Cell suspensions were sequentially filtered through 100 and 40 μm filters (Corning Cellgro #431752 and #431750) to remove debris. Erythrocytes were removed through incubation in erythrocyte lysis buffer (IBI Scientific #89135-030).

    Single-cell RNA-sequencing library preparation. After digestion, single-cell suspensions were washed and resuspended in 0.04% BSA in PBS at a concentration of 106 cells/ml. Cells were counted manually with a hemocytometer to determine their concentration. Single-cell RNA-sequencing libraries were prepared using the Chromium Single Cell 3’ reagent kit v3 (10x Genomics, PN-1000075; Pleasanton, CA) following the manufacturer’s protocol. Cells were diluted into the Chromium Single Cell A Chip to yield a recovery of 6,000 single-cell transcriptomes. After preparation, libraries were sequenced using on a NextSeq 500 (Illumina; San Diego, CA) using 75 cycle high output kits (Index 1 = 8, Read 1 = 26, and Read 2 = 58). Details on estimated sequencing saturation and the number of reads per sample are shown in Sup. Data 1.

    Spatial RNA sequencing library preparation. Tibialis anterior muscles of adult (5 mo) C57BL6/J mice were injected with 10µl notexin (10 µg/ml) at 2, 5, and 7 days prior to collection. Upon collection, tibialis anterior muscles were isolated, embedded in OCT, and frozen fresh in liquid nitrogen. Spatially tagged cDNA libraries were built using the Visium Spatial Gene Expression 3’ Library Construction v1 Kit (10x Genomics, PN-1000187; Pleasanton, CA) (Fig. S7). Optimal tissue permeabilization time for 10 µm thick sections was found to be 15 minutes using the 10x Genomics Visium Tissue Optimization Kit (PN-1000193). H&E stained tissue sections were imaged using Zeiss PALM MicroBeam laser capture microdissection system and the images were stitched and processed using Fiji ImageJ software. cDNA libraries were sequenced on an Illumina NextSeq 500 using 150 cycle high output kits (Read 1=28bp, Read 2=120bp, Index 1=10bp, and Index 2=10bp). Frames around the capture area on the Visium slide were aligned manually and spots covering the tissue were selected using Loop Browser v4.0.0 software (10x Genomics). Sequencing data was then aligned to the mouse reference genome (mm10) using the spaceranger v1.0.0 pipeline to generate a feature-by-spot-barcode expression matrix (10x Genomics).

    Download and alignment of single-cell RNA sequencing data. For all samples available via SRA, parallel-fastq-dump (github.com/rvalieris/parallel-fastq-dump) was used to download raw .fastq files. Samples which were only available as .bam files were converted to .fastq format using bamtofastq from 10x Genomics (github.com/10XGenomics/bamtofastq). Raw reads were aligned to the mm10 reference using cellranger (v3.1.0).

    Preprocessing and batch correction of single-cell RNA sequencing datasets. First, ambient RNA signal was removed using the default SoupX (v1.4.5) workflow (autoEstCounts and adjustCounts; github.com/constantAmateur/SoupX). Samples were then preprocessed using the standard Seurat (v3.2.1) workflow (NormalizeData, ScaleData, FindVariableFeatures, RunPCA, FindNeighbors, FindClusters, and RunUMAP; github.com/satijalab/seurat). Cells with fewer than 750 features, fewer than 1000 transcripts, or more than 30% of unique transcripts derived from mitochondrial genes were removed. After preprocessing, DoubletFinder (v2.0) was used to identify putative doublets in each dataset, individually. BCmvn optimization was used for PK parameterization. Estimated doublet rates were computed by fitting the total number of cells after quality filtering to a linear regression of the expected doublet rates published in the 10x Chromium handbook. Estimated homotypic doublet rates were also accounted for using the modelHomotypic function. The default PN value (0.25) was used. Putative doublets were then removed from each individual dataset. After preprocessing and quality filtering, we merged the datasets and performed batch-correction with three tools, independently- Harmony (github.com/immunogenomics/harmony) (v1.0), Scanorama (github.com/brianhie/scanorama) (v1.3), and BBKNN (github.com/Teichlab/bbknn) (v1.3.12). We then used Seurat to process the integrated data. After initial integration, we removed the noisy cluster and re-integrated the data using each of the three batch-correction tools.

    Cell type annotation. Cell types were determined for each integration method independently. For Harmony and Scanorama, dimensions accounting for 95% of the total variance were used to generate SNN graphs (Seurat::FindNeighbors). Louvain clustering was then performed on the output graphs (including the corrected graph output by BBKNN) using Seurat::FindClusters. A clustering resolution of 1.2 was used for Harmony (25 initial clusters), BBKNN (28 initial clusters), and Scanorama (38 initial clusters). Cell types were determined based on expression of canonical genes (Fig. S3). Clusters which had similar canonical marker gene expression patterns were merged.

    Pseudotime workflow. Cells were subset based on the consensus cell types between all three integration methods. Harmony embedding values from the dimensions accounting for 95% of the total variance were used for further dimensional reduction with PHATE, using phateR (v1.0.4) (github.com/KrishnaswamyLab/phateR).

    Deconvolution of spatial RNA sequencing spots. Spot deconvolution was performed using the deconvolution module in BayesPrism (previously known as “Tumor microEnvironment Deconvolution”, TED, v1.0; github.com/Danko-Lab/TED). First, myogenic cells were re-labeled, according to binning along the first PHATE dimension, as “Quiescent MuSCs” (bins 4-5), “Activated MuSCs” (bins 6-7), “Committed Myoblasts” (bins 8-10), and “Fusing Myoctes” (bins 11-18). Culture-associated muscle stem cells were ignored and myonuclei labels were retained as “Myonuclei (Type IIb)” and “Myonuclei (Type IIx)”. Next, highly and differentially expressed genes across the 25 groups of cells were identified with differential gene expression analysis using Seurat (FindAllMarkers, using Wilcoxon Rank Sum Test; results in Sup. Data 2). The resulting genes were filtered based on average log2-fold change (avg_logFC > 1) and the percentage of cells within the cluster which express each gene (pct.expressed > 0.5), yielding 1,069 genes. Mitochondrial and ribosomal protein genes were also removed from this list, in line with recommendations in the BayesPrism vignette. For each of the cell types, mean raw counts were calculated across the 1,069 genes to generate a gene expression profile for BayesPrism. Raw counts for each spot were then passed to the run.Ted function, using

  2. Markers of all cells and subset clusters. In the first tab: markers for the...

    • plos.figshare.com
    xlsx
    Updated Jun 1, 2023
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Adi Egozi; Oluwabunmi Olaloye; Lael Werner; Tatiana Silva; Blake McCourt; Richard W. Pierce; Xiaojing An; Fujing Wang; Kong Chen; Jordan S. Pober; Dror Shouval; Shalev Itzkovitz; Liza Konnikova (2023). Markers of all cells and subset clusters. In the first tab: markers for the 8 cell type clusters, identified by the FindAllMarkers command in Seurat. [Dataset]. http://doi.org/10.1371/journal.pbio.3002124.s007
    Explore at:
    xlsxAvailable download formats
    Dataset updated
    Jun 1, 2023
    Dataset provided by
    PLOShttp://plos.org/
    Authors
    Adi Egozi; Oluwabunmi Olaloye; Lael Werner; Tatiana Silva; Blake McCourt; Richard W. Pierce; Xiaojing An; Fujing Wang; Kong Chen; Jordan S. Pober; Dror Shouval; Shalev Itzkovitz; Liza Konnikova
    License

    Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
    License information was derived automatically

    Description

    Markers included have a log-fold above 1 and expressed in at least 50% of the cluster cells. In all the rest of the tabs: Markers for each of cell subtype (Myeloid, T/NK cells, vascular/lymphatic endothelial cells, enterocytes, and fibroblasts) clusters (compared internally to the other clusters in the same cell subtype), identified by the FindAllMarkers command in Seurat. Markers included have a log-fold above 0.6 and expressed in at least 25% of the cluster cells. (XLSX)

  3. f

    Skin sc-RNASeq from seven body sites (face, scalp, axilla, palmoplantar,...

    • plus.figshare.com
    bin
    Updated Mar 11, 2025
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Lam C Tsoi; Rachael Bogle; Johann Gudjonsson; Meri Oliva; Bridget Riley-Gillis (2025). Skin sc-RNASeq from seven body sites (face, scalp, axilla, palmoplantar, arm, leg, and back) [Dataset]. http://doi.org/10.25452/figshare.plus.25696620.v2
    Explore at:
    binAvailable download formats
    Dataset updated
    Mar 11, 2025
    Dataset provided by
    Figshare+
    Authors
    Lam C Tsoi; Rachael Bogle; Johann Gudjonsson; Meri Oliva; Bridget Riley-Gillis
    License

    CC0 1.0 Universal Public Domain Dedicationhttps://creativecommons.org/publicdomain/zero/1.0/
    License information was derived automatically

    Description

    This sc-RNAseq dataset is composed of disease-unaffected epidermal samples from 96 skin biopsies: 18 from published datasets - GSE173706, GSE249279 – and 78 newly generated ones. Biopsy sample and protocol details, and curated cell-type signature genes, are available in the scRNASeq_source_info_FigShare spreadsheet of this dataset. Processed Seurat object are provided herein. Raw data are available in SRA (id PRJNA1054546). Biopsies originated from seven body sites (face, scalp, axilla, palmoplantar, arm, leg, and back). The skin biopsies were separated into epidermis and dermis before dissociated and enriched for various cell fractions (keratinocytes, fibroblasts, and endothelial cells) and immune cells (myeloid and lymphoid cells) to up sample rare cell types. In total, across body sites, 274,834 cells were profiled, including 96,194 keratinocytes. Seurat v3.0. was utilized to normalize, scale, and reduce the dimensionality of the data. Low quality cells containing less than 200 genes per cell as well as greater than 5,000 genes per cell were filtered out. Cells containing more mitochondrial genes than the permitted quantile of 0.05 were removed. Ambient RNA was removed using R package SoupX v1.6.2. Doublets were removed using scDblFinder v1.12.0. Principal components (PC) were obtained from the topmost 2,000 variable genes, and the Uniform Manifold Approximation and Projection (UMAP) dimensional reduction technique was applied to the 30 topmost variable PC-reduced dataset. Batch effect correction was performed utilizing harmony v1.0, using donor as batch. After batch correction, cells were clustered using shared nearest neighbor modularity optimization-based clustering. Cluster marker genes were identified with FindAllMarkers; cluster corresponding cell type was identified by comparing marker genes to curated cell-type signature genes. Differential expression by keratinocyte subtype was performed with Seurat (v4.3.0) FindMarkers function by comparing keratinocyte subtype to non-keratinocyte clusters. The log fold-change of the average expression between a keratinocyte subtype cluster compared to the rest of clusters is utilized as keratinocyte-subtype gene expression statistic.

  4. scRNAseq_Dataset Merge AMI d5 (CD45+Fibroblast) + AAA Kinetik +...

    • zenodo.org
    Updated Mar 28, 2023
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Alexander Lang; Alexander Lang (2023). scRNAseq_Dataset Merge AMI d5 (CD45+Fibroblast) + AAA Kinetik + Cite-Seq_Dataset AG Gerdes [Dataset]. http://doi.org/10.5281/zenodo.7774809
    Explore at:
    Dataset updated
    Mar 28, 2023
    Dataset provided by
    Zenodohttp://zenodo.org/
    Authors
    Alexander Lang; Alexander Lang
    Description

    Integration Skript:

    library(Seurat)
    library(tidyverse)
    library(Matrix)

    #cite <- readRDS("C:/Users/alex/sciebo/ALL_NGS/scRNAseq/scRNAseq/Merge AAA mit Cite AAA/Cite_seq_v0.41.rds")
    #CD45 <- readRDS("C:/Users/alex/sciebo/ALL_NGS/scRNAseq/scRNAseq/Schrader/Fertige_Analysen/TS_d5_paper/CD45.rds")
    AAA <- readRDS("C:/Users/alex/sciebo/AAA_Zhao_v4.rds")
    cite <- readRDS("C:/Users/alex/sciebo/CITE_Seq_v0.5.rds")
    all4 <- readRDS("C:/Users/alex/sciebo/ALL_NGS/scRNAseq/scRNAseq/Schrader/Fertige_Analysen/Schrader_All4_Rohanalyse/all4_220228.rds")

    #fuse lists
    c <- list(cite, all4, AAA)
    names(c) <- c("cite", "all4", "AAA")

    pancreas.list <- c[c("cite", "all4", "AAA")]
    for (i in 1:length(pancreas.list)) {
    pancreas.list[[i]] <- SCTransform(pancreas.list[[i]], verbose = FALSE)
    }

    pancreas.features <- SelectIntegrationFeatures(object.list = pancreas.list, nfeatures = 3000)
    #options(future.globals.maxSize= 6091289600)
    #pancreas.list <- PrepSCTIntegration(object.list = pancreas.list, anchor.features = pancreas.features,
    #verbose = FALSE) #future.globals.maxsize was to low. changed it to options(future.globals.maxSize= 1091289600)
    #identify anchors

    #alternative from tutorial (https://satijalab.org/seurat/articles/integration_introduction.html)
    #memory.limit(9999999999)
    features <- SelectIntegrationFeatures(object.list = pancreas.list, nfeatures = 3000)
    pancreas.list <- PrepSCTIntegration(object.list = pancreas.list, anchor.features = features)
    pancreas.anchors <- FindIntegrationAnchors(object.list = pancreas.list, normalization.method = "SCT", anchor.features = pancreas.features, verbose = FALSE)
    pancreas.integrated <- IntegrateData(anchorset = pancreas.anchors, normalization.method = "SCT",
    verbose = FALSE)

    setwd("C:/Users/alex/sciebo/ALL_NGS/scRNAseq/scRNAseq/Schrader/Fertige_Analysen/TS_d5_paper")

    saveRDS(pancreas.integrated, file = "integrated_AAA_Cite_AMI.rds")

    saveRDS(cd45, file = "integrated_AAA_Cite_CD45.rds")

    seurat <- pancreas.integrated

    #seurat <- readRDS("C:/Users/alex/sciebo/ALL_NGS/scRNAseq/scRNAseq/Schrader/Fertige_Analysen/TS_d5_paper/integrated_d5_cite.rds")

    DefaultAssay(object = seurat) <- "integrated"
    seurat <- FindVariableFeatures(seurat, selection.method = "vst", nfeatures = 3000)
    seurat <- ScaleData(seurat, verbose = FALSE)
    seurat <- RunPCA(seurat, npcs = 30, verbose = FALSE)
    seurat <- FindNeighbors(seurat, dims = 1:30)
    seurat <- FindClusters(seurat, resolution = 0.5)
    seurat <- RunUMAP(seurat, reduction = "pca", dims = 1:30)
    DimPlot(seurat, reduction = "umap", split.by = "treatment") + NoLegend()


    DimPlot(seurat, label = T, repel = T) + NoLegend()

    DefaultAssay(object = seurat) <- "ADT"
    adt_marker_integrated <- FindAllMarkers(seurat, logfc.threshold = 0.3)
    write.csv(adt_marker_integrated, file = "adt_marker_all4_integrated.csv")

    DefaultAssay(object = seurat) <- "RNA"
    RNA_marker_integrated <- FindAllMarkers(seurat, logfc.threshold = 0.5)
    write.csv(RNA_marker_integrated, file = "RNA_marker_all4_integrated.csv")

    DimPlot(seurat, label = T, repel = T, split.by = "tissue") + NoLegend()

    FeaturePlot(seurat, features = "Cd40", order = T, label = T)
    FeaturePlot(seurat, features = "Ms.CD40", order = T, label = T)


    #####
    #leanup:
    > seurat@meta.data[["sen_score1"]] <- NULL
    > seurat@meta.data[["sen_score2"]] <- NULL
    > seurat@meta.data[["sen_score3"]] <- NULL
    > seurat@meta.data[["sen_score4"]] <- NULL
    > seurat@meta.data[["sen_score5"]] <- NULL
    > seurat@meta.data[["sen_score6"]] <- NULL
    > seurat@meta.data[["sen_score7"]] <- NULL
    > seurat@meta.data[["pANN_0.25_0.1_1211"]] <- NULL
    > seurat@meta.data[["DF.classifications_0.25_0.1_1211"]] <- NULL
    > seurat@meta.data[["DF.classifications_0.25_0.1_466"]] <- NULL
    > seurat@assays[["prediction.score.celltype"]] <- NULL
    > seurat@meta.data[["predicted.celltype"]] <- NULL
    > seurat@meta.data[["DF.classifications_0.25_0.1_184"]] <- NULL
    > seurat@meta.data[["DF.classifications_0.25_0.1_953"]] <- NULL
    > seurat@meta.data[["integrated_snn_res.3"]] <- NULL
    > seurat@meta.data[["RNA_snn_res.3"]] <- NULL
    > seurat@meta.data[["SingleR"]] <- NULL
    > seurat@meta.data[["SingleR_fine"]] <- NULL
    > seurat@meta.data[["ImmGen"]] <- NULL
    > seurat@meta.data[["ImmGen_fine"]] <- NULL
    > seurat@meta.data[["percent.mt"]] <- NULL
    > seurat@meta.data[["nCount_integrated"]] <- NULL
    > seurat@meta.data[["nFeature_integrated"]] <- NULL
    > seurat@meta.data[["S.Score"]] <- NULL
    > seurat@meta.data[["G2M.Score"]] <- NULL
    > seurat@meta.data[["Phase"]] <- NULL
    > seurat@meta.data[["sen_score8"]] <- NULL
    > seurat@meta.data[["sen_score9"]] <- NULL
    > seurat@meta.data[["sen_score10"]] <- NULL
    > seurat@meta.data[["sen_score11"]] <- NULL
    > seurat@meta.data[["sen_score12"]] <- NULL
    > seurat@meta.data[["sen_score13"]] <- NULL
    > seurat@meta.data[["sen_score14"]] <- NULL
    > seurat@meta.data[["sen_score15"]] <- NULL
    > seurat@meta.data[["sen_score16"]] <- NULL
    > seurat@meta.data[["sen_score17"]] <- NULL
    > seurat@meta.data[["sen_score18"]] <- NULL
    > seurat@meta.data[["sen_score19"]] <- NULL
    seurat@meta.data[["pANN_0.25_0.1_184"]] <- NULL
    seurat@meta.data[["pANN_0.25_0.1_953"]] <- NULL
    seurat@meta.data[["pANN_0.25_0.1_466"]] <- NULL

  5. n

    Data from: Dermomyotome-derived endothelial cells migrate to the dorsal...

    • data.niaid.nih.gov
    • datadryad.org
    zip
    Updated Oct 4, 2023
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    David Traver; Pankaj Sahai-Hernandez; Claire Pouget; Shai Eyal; Ondrej Svoboda; Jose Chacon; Lin Grimm; Tor Gjøen (2023). Dermomyotome-derived endothelial cells migrate to the dorsal aorta to support hematopoietic stem cell emergence [Dataset]. http://doi.org/10.6075/J0GB22J0
    Explore at:
    zipAvailable download formats
    Dataset updated
    Oct 4, 2023
    Dataset provided by
    University of Oslo
    University of California, San Diego
    Authors
    David Traver; Pankaj Sahai-Hernandez; Claire Pouget; Shai Eyal; Ondrej Svoboda; Jose Chacon; Lin Grimm; Tor Gjøen
    License

    https://spdx.org/licenses/CC0-1.0.htmlhttps://spdx.org/licenses/CC0-1.0.html

    Description

    Development of the dorsal aorta is a key step in the establishment of the adult blood-forming system since hematopoietic stem and progenitor cells (HSPCs) arise from ventral aortic endothelium in all vertebrate animals studied. Work in zebrafish has demonstrated that arterial and venous endothelial precursors arise from distinct subsets of lateral plate mesoderm. Here, we profile the transcriptome of the earliest detectable endothelial cells (ECs) during zebrafish embryogenesis to demonstrate that tissue-specific EC programs initiate much earlier than previously appreciated, by the end of gastrulation. Classic studies in the chick embryo showed that paraxial mesoderm generates a subset of somite-derived endothelial cells (SDECs) that incorporate into the dorsal aorta to replace HSPCs as they exit the aorta and enter circulation. We describe a conserved program in the zebrafish, where a rare population of endothelial precursors delaminates from the dermomyotome to incorporate exclusively into the developing dorsal aorta. Although SDECs lack hematopoietic potential, they act as a local niche to support the emergence of HSPCs from neighboring hemogenic endothelium. Thus, at least three subsets of ECs contribute to the developing dorsal aorta: vascular ECs, hemogenic ECs, and SDECs. Taken together, our findings indicate that the distinct spatial origins of endothelial precursors dictate different cellular potentials within the developing dorsal aorta. Methods Single-cell RNA sample preparation After FACS, total cell concentration and viability were ascertained using a TC20 Automated Cell Counter (Bio-Rad). Samples were then resuspended in 1XPBS with 10% BSA at a concentration between 800-3000 per ml. Samples were loaded on the 10X Chromium system and processed as per manufacturer’s instructions (10X Genomics). Single cell libraries were prepared as per the manufacturer’s instructions using the Single Cell 3’ Reagent Kit v2 (10X Genomics). Single cell RNA-seq libraries and barcode amplicons were sequenced on an Illumina HiSeq platform. Single-cell RNA sequencing analysis The Chromium 3’ sequencing libraries were generated using Chromium Single Cell 3’ Chip kit v3 and sequenced with (actually, I don’t know:( what instrument was used?). The Ilumina FASTQ files were used to generate filtered matrices using CellRanger (10X Genomics) with default parameters and imported into R for exploration and statistical analysis using a Seurat package (La Manno et al., 2018). Counts were normalized according to total expression, multiplied by a scale factor (10,000), and log-transformed. For cell cluster identification and visualization, gene expression values were also scaled according to highly variable genes after controlling for unwanted variation generated by sample identity. Cell clusters were identified based on UMAP of the first 14 principal components of PCA using Seurat’s method, Find Clusters, with an original Louvain algorithm and resolution parameter value 0.5. To find cluster marker genes, Seurat’s method, FindAllMarkers. Only genes exhibiting significant (adjusted p-value < 0.05) a minimal average absolute log2-fold change of 0.2 between each of the clusters and the rest of the dataset were considered as differentially expressed. To merge individual datasets and to remove batch effects, Seurat v3 Integration and Label Transfer standard workflow (Stuart et al., 2019)

  6. Marker genes of each MDS-based cluster of PBMCs.

    • plos.figshare.com
    xlsx
    Updated Oct 10, 2024
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Yutaro Kumagai (2024). Marker genes of each MDS-based cluster of PBMCs. [Dataset]. http://doi.org/10.1371/journal.pcbi.1012480.s003
    Explore at:
    xlsxAvailable download formats
    Dataset updated
    Oct 10, 2024
    Dataset provided by
    PLOShttp://plos.org/
    Authors
    Yutaro Kumagai
    License

    Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
    License information was derived automatically

    Description

    The output of FindAllMarkers function in Seurat was printed as a table. (XLSX)

  7. Marker genes of each MDS-based cluster of cells undergoing hematopoiesis.

    • plos.figshare.com
    xlsx
    Updated Oct 10, 2024
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Yutaro Kumagai (2024). Marker genes of each MDS-based cluster of cells undergoing hematopoiesis. [Dataset]. http://doi.org/10.1371/journal.pcbi.1012480.s006
    Explore at:
    xlsxAvailable download formats
    Dataset updated
    Oct 10, 2024
    Dataset provided by
    PLOShttp://plos.org/
    Authors
    Yutaro Kumagai
    License

    Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
    License information was derived automatically

    Description

    The output of FindAllMarkers function in Seurat was printed as a table. (XLSX)

  8. Marker genes of each BootCellNet2 clustering results of PBMCs based on...

    • plos.figshare.com
    xlsx
    Updated Oct 10, 2024
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Yutaro Kumagai (2024). Marker genes of each BootCellNet2 clustering results of PBMCs based on CITE-seq. [Dataset]. http://doi.org/10.1371/journal.pcbi.1012480.s010
    Explore at:
    xlsxAvailable download formats
    Dataset updated
    Oct 10, 2024
    Dataset provided by
    PLOShttp://plos.org/
    Authors
    Yutaro Kumagai
    License

    Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
    License information was derived automatically

    Description

    The output of FindAllMarkers function in Seurat was printed as a table. (XLSX)

  9. Marker genes of each BootCellNet2 cluster of BAL cells from COVID-19...

    • plos.figshare.com
    xlsx
    Updated Oct 10, 2024
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Yutaro Kumagai (2024). Marker genes of each BootCellNet2 cluster of BAL cells from COVID-19 patients. [Dataset]. http://doi.org/10.1371/journal.pcbi.1012480.s014
    Explore at:
    xlsxAvailable download formats
    Dataset updated
    Oct 10, 2024
    Dataset provided by
    PLOShttp://plos.org/
    Authors
    Yutaro Kumagai
    License

    Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
    License information was derived automatically

    Description

    The output of FindAllMarkers function in Seurat was printed as a table. (XLSX)

  10. f

    Additional file 10 of Single-cell transcriptomics highlights immunological...

    • springernature.figshare.com
    xlsx
    Updated Feb 7, 2024
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Qiqing Huang; Yuanyuan Wang; Lili Zhang; Wei Qian; Shaoran Shen; Jingshen Wang; Shuangshuang Wu; Wei Xu; Bo Chen; Mingyan Lin; Jianqing Wu (2024). Additional file 10 of Single-cell transcriptomics highlights immunological dysregulations of monocytes in the pathobiology of COPD [Dataset]. http://doi.org/10.6084/m9.figshare.22601786.v1
    Explore at:
    xlsxAvailable download formats
    Dataset updated
    Feb 7, 2024
    Dataset provided by
    figshare
    Authors
    Qiqing Huang; Yuanyuan Wang; Lili Zhang; Wei Qian; Shaoran Shen; Jingshen Wang; Shuangshuang Wu; Wei Xu; Bo Chen; Mingyan Lin; Jianqing Wu
    License

    Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
    License information was derived automatically

    Description

    Additional file 10: Dataset 8. Lists of markers in 2 sub-clusters of club cells, namely autoimmune-prone sub-cluster of club cells and mix sub-cluster of club cells, related to Fig. 5D and Fig. S5M. The resulting sub-cluster markers were identified by the seurat FindAllMarkers, including the p-value, the average log2(fold change) of the gene in the sub-cluster compared to all other sub-clusters, the percent of cells expressing the gene in the sub-cluster (pct.1), the percent of cells expressing the gene in all other sub-clusters (pct.2), and the adjusted p-value.

  11. f

    Marker genes for cell clusters in the integrated scRNA-seq dataset.

    • plos.figshare.com
    csv
    Updated Oct 24, 2024
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Alexander Ferrena; Xusheng Zhang; Rupendra Shrestha; Deyou Zheng; Wei Liu (2024). Marker genes for cell clusters in the integrated scRNA-seq dataset. [Dataset]. http://doi.org/10.1371/journal.pone.0308839.s008
    Explore at:
    csvAvailable download formats
    Dataset updated
    Oct 24, 2024
    Dataset provided by
    PLOS ONE
    Authors
    Alexander Ferrena; Xusheng Zhang; Rupendra Shrestha; Deyou Zheng; Wei Liu
    License

    Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
    License information was derived automatically

    Description

    Related to Fig 1. These cluster markers were identified when one cluster was compared with all other clusters in the integrated scRNA-seq dataset using the function FindAllMarkers in Seurat. (CSV)

  12. f

    Table_1_In-depth single-cell and bulk-RNA sequencing developed a...

    • frontiersin.figshare.com
    xlsx
    Updated Oct 19, 2023
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Liangyu Zhang; Xun Zhang; Maohao Guan; Fengqiang Yu; Fancai Lai (2023). Table_1_In-depth single-cell and bulk-RNA sequencing developed a NETosis-related gene signature affects non-small-cell lung cancer prognosis and tumor microenvironment: results from over 3,000 patients.xlsx [Dataset]. http://doi.org/10.3389/fonc.2023.1282335.s001
    Explore at:
    xlsxAvailable download formats
    Dataset updated
    Oct 19, 2023
    Dataset provided by
    Frontiers
    Authors
    Liangyu Zhang; Xun Zhang; Maohao Guan; Fengqiang Yu; Fancai Lai
    License

    Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
    License information was derived automatically

    Description

    BackgroundCell death caused by neutrophil extracellular traps (NETs) is known as NETosis. Despite the increasing importance of NETosis in cancer diagnosis and treatment, its role in Non-Small-Cell Lung Cancer (NSCLC) remains unclear.MethodsA total of 3298 NSCLC patients from different cohorts were included. The AUCell method was used to compute cells’ NETosis scores from single-cell RNA-sequencing data. DEGs in sc-RNA dataset were obtained by the Seurat’s “FindAllMarkers” function, and DEGs in bulk-RNA dataset were acquired by the DESeq2 package. ConsensusClusterPlus package was used to group patients into different NETosis subtypes, and the Enet algorithm was used to construct the NETosis-Related Riskscore (NETRS). Enrichment analyses were conducted using the GSVA and ClusterProfiler packages. Six distinct algorithms were utilized to evaluate patients’ immune cell infiltration level. Patients’ SNV and CNV data were analyzed by maftools and GISTIC2.0, respectively. Drug information was obtained from the GDSC1, and predicted by the Oncopredict package. Patient response to immunotherapy was evaluated by the TIDE algorithm in conjunction with the phs000452 immunotherapy cohort. Six NRGs’ differential expression was verified using qRT-PCR and immunohistochemistry.ResultsAmong all cell types, neutrophils had the highest AUCell score. By Intersecting the DEGs between high and low NETosis classes, DEGs between normal and LUAD tissues, and prognostic related genes, 61 prognostic related NRGs were identified. Based on the 61 NRGs, all LUAD patients can be divided into two clusters, showing different prognostic and TME characteristics. Enet regression identified the NETRS composed of 18 NRGs. NETRS significantly associated with LUAD patients’ clinical characteristics, and patients at different NETRS groups showed significant differences on prognosis, TME characteristics, immune-related molecules’ expression levels, gene mutation frequencies, response to immunotherapy, and drug sensitivity. Besides, NETRS was more powerful than 20 published gene signatures in predicting LUAD patients’ survival. Nine independent cohorts confirmed that NETRS is also valuable in predicting the prognosis of all NSCLC patients. Finally, six NRGs’ expression was confirmed using three independent datasets, qRT-PCR and immunohistochemistry.ConclusionNETRS can serves as a valuable prognostic indicator for patients with NSCLC, providing insights into the tumor microenvironment and predicting the response to cancer therapy.

  13. Shared equine BAL cell-type marker genes across HIVE and Drop-seq data.

    • plos.figshare.com
    xls
    Updated Jan 24, 2025
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Kim Fegraeus; Miia Riihimäki; Jessica Nordlund; Srinivas Akula; Sara Wernersson; Amanda Raine (2025). Shared equine BAL cell-type marker genes across HIVE and Drop-seq data. [Dataset]. http://doi.org/10.1371/journal.pone.0317343.t003
    Explore at:
    xlsAvailable download formats
    Dataset updated
    Jan 24, 2025
    Dataset provided by
    PLOShttp://plos.org/
    Authors
    Kim Fegraeus; Miia Riihimäki; Jessica Nordlund; Srinivas Akula; Sara Wernersson; Amanda Raine
    License

    Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
    License information was derived automatically

    Description

    The table lists common cluster markers among the top 25 genes with the highest log2FC in each respective dataset, as identified by Seurat’s (v.4.3) FindAllMarkers function.

  14. Additional Data

    • figshare.com
    application/gzip
    Updated Aug 5, 2021
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Jennifer Nguyen (2021). Additional Data [Dataset]. http://doi.org/10.6084/m9.figshare.15109422.v7
    Explore at:
    application/gzipAvailable download formats
    Dataset updated
    Aug 5, 2021
    Dataset provided by
    Figsharehttp://figshare.com/
    Authors
    Jennifer Nguyen
    License

    Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
    License information was derived automatically

    Description

    Here, we provide:Robjects pertaining to scRNA-seq (Seurat) and snATAC-seq (Signac) analysis. These contain the single-cell and single-nuclei used in downstream analyses. Tables containing information about the gene markers identified for each cluster in scRNA-seq, peak markers identified for each cluster in snATAC-seq, and motif enrichment analyses using chromVAR motif scores. Differential gene expression and motif enrichment analyses was performed using Wilcoxon rank sum test comparing the distribution of gene expression or chromVAR motif scores between cells in the cluster and all other cells. Differential peak analyses was performed using FindAllMarkers in Signac.

  15. Significant Differential Tumor-Intrinsic Markers.

    • plos.figshare.com
    xlsx
    Updated Jun 16, 2023
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Aparna Jorapur; Lisa A. Marshall; Scott Jacobson; Mengshu Xu; Sachie Marubayashi; Mikhail Zibinsky; Dennis X. Hu; Omar Robles; Jeffrey J. Jackson; Valentin Baloche; Pierre Busson; David Wustrow; Dirk G. Brockstedt; Oezcan Talay; Paul D. Kassner; Gene Cutler (2023). Significant Differential Tumor-Intrinsic Markers. [Dataset]. http://doi.org/10.1371/journal.ppat.1010200.s017
    Explore at:
    xlsxAvailable download formats
    Dataset updated
    Jun 16, 2023
    Dataset provided by
    PLOShttp://plos.org/
    Authors
    Aparna Jorapur; Lisa A. Marshall; Scott Jacobson; Mengshu Xu; Sachie Marubayashi; Mikhail Zibinsky; Dennis X. Hu; Omar Robles; Jeffrey J. Jackson; Valentin Baloche; Pierre Busson; David Wustrow; Dirk G. Brockstedt; Oezcan Talay; Paul D. Kassner; Gene Cutler
    License

    Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
    License information was derived automatically

    Description

    Gene markers distinguishing the tumor cells between tumor types in the single cell RNA-Sequencing data, as identified by the “FindAllMarkers” function of the Seurat analysis package. Additional details are given at the top of the table. (XLSX)

  16. Not seeing a result you expected?
    Learn how you can add new datasets to our index.

Share
FacebookFacebook
TwitterTwitter
Email
Click to copy link
Link copied
Close
Cite
David McKellar; Iwijn De Vlaminck; Benjamin Cosgrove (2021). Large-scale integration of single-cell transcriptomic data captures transitional progenitor states in mouse skeletal muscle regeneration [Dataset]. http://doi.org/10.5061/dryad.t4b8gtj34

Data from: Large-scale integration of single-cell transcriptomic data captures transitional progenitor states in mouse skeletal muscle regeneration

Related Article
Explore at:
zipAvailable download formats
Dataset updated
Dec 14, 2021
Dataset provided by
Cornell University
Authors
David McKellar; Iwijn De Vlaminck; Benjamin Cosgrove
License

https://spdx.org/licenses/CC0-1.0.htmlhttps://spdx.org/licenses/CC0-1.0.html

Description

Skeletal muscle repair is driven by the coordinated self-renewal and fusion of myogenic stem and progenitor cells. Single-cell gene expression analyses of myogenesis have been hampered by the poor sampling of rare and transient cell states that are critical for muscle repair, and do not inform the spatial context that is important for myogenic differentiation. Here, we demonstrate how large-scale integration of single-cell and spatial transcriptomic data can overcome these limitations. We created a single-cell transcriptomic dataset of mouse skeletal muscle by integration, consensus annotation, and analysis of 23 newly collected scRNAseq datasets and 88 publicly available single-cell (scRNAseq) and single-nucleus (snRNAseq) RNA-sequencing datasets. The resulting dataset includes more than 365,000 cells and spans a wide range of ages, injury, and repair conditions. Together, these data enabled identification of the predominant cell types in skeletal muscle, and resolved cell subtypes, including endothelial subtypes distinguished by vessel-type of origin, fibro/adipogenic progenitors defined by functional roles, and many distinct immune populations. The representation of different experimental conditions and the depth of transcriptome coverage enabled robust profiling of sparsely expressed genes. We built a densely sampled transcriptomic model of myogenesis, from stem cell quiescence to myofiber maturation and identified rare, transitional states of progenitor commitment and fusion that are poorly represented in individual datasets. We performed spatial RNA sequencing of mouse muscle at three time points after injury and used the integrated dataset as a reference to achieve a high-resolution, local deconvolution of cell subtypes. We also used the integrated dataset to explore ligand-receptor co-expression patterns and identify dynamic cell-cell interactions in muscle injury response. We provide a public web tool to enable interactive exploration and visualization of the data. Our work supports the utility of large-scale integration of single-cell transcriptomic data as a tool for biological discovery.

Methods Mice. The Cornell University Institutional Animal Care and Use Committee (IACUC) approved all animal protocols, and experiments were performed in compliance with its institutional guidelines. Adult C57BL/6J mice (mus musculus) were obtained from Jackson Laboratories (#000664; Bar Harbor, ME) and were used at 4-7 months of age. Aged C57BL/6J mice were obtained from the National Institute of Aging (NIA) Rodent Aging Colony and were used at 20 months of age. For new scRNAseq experiments, female mice were used in each experiment.

Mouse injuries and single-cell isolation. To induce muscle injury, both tibialis anterior (TA) muscles of old (20 months) C57BL/6J mice were injected with 10 µl of notexin (10 µg/ml; Latoxan; France). At 0, 1, 2, 3.5, 5, or 7 days post-injury (dpi), mice were sacrificed and TA muscles were collected and processed independently to generate single-cell suspensions. Muscles were digested with 8 mg/ml Collagenase D (Roche; Switzerland) and 10 U/ml Dispase II (Roche; Switzerland), followed by manual dissociation to generate cell suspensions. Cell suspensions were sequentially filtered through 100 and 40 μm filters (Corning Cellgro #431752 and #431750) to remove debris. Erythrocytes were removed through incubation in erythrocyte lysis buffer (IBI Scientific #89135-030).

Single-cell RNA-sequencing library preparation. After digestion, single-cell suspensions were washed and resuspended in 0.04% BSA in PBS at a concentration of 106 cells/ml. Cells were counted manually with a hemocytometer to determine their concentration. Single-cell RNA-sequencing libraries were prepared using the Chromium Single Cell 3’ reagent kit v3 (10x Genomics, PN-1000075; Pleasanton, CA) following the manufacturer’s protocol. Cells were diluted into the Chromium Single Cell A Chip to yield a recovery of 6,000 single-cell transcriptomes. After preparation, libraries were sequenced using on a NextSeq 500 (Illumina; San Diego, CA) using 75 cycle high output kits (Index 1 = 8, Read 1 = 26, and Read 2 = 58). Details on estimated sequencing saturation and the number of reads per sample are shown in Sup. Data 1.

Spatial RNA sequencing library preparation. Tibialis anterior muscles of adult (5 mo) C57BL6/J mice were injected with 10µl notexin (10 µg/ml) at 2, 5, and 7 days prior to collection. Upon collection, tibialis anterior muscles were isolated, embedded in OCT, and frozen fresh in liquid nitrogen. Spatially tagged cDNA libraries were built using the Visium Spatial Gene Expression 3’ Library Construction v1 Kit (10x Genomics, PN-1000187; Pleasanton, CA) (Fig. S7). Optimal tissue permeabilization time for 10 µm thick sections was found to be 15 minutes using the 10x Genomics Visium Tissue Optimization Kit (PN-1000193). H&E stained tissue sections were imaged using Zeiss PALM MicroBeam laser capture microdissection system and the images were stitched and processed using Fiji ImageJ software. cDNA libraries were sequenced on an Illumina NextSeq 500 using 150 cycle high output kits (Read 1=28bp, Read 2=120bp, Index 1=10bp, and Index 2=10bp). Frames around the capture area on the Visium slide were aligned manually and spots covering the tissue were selected using Loop Browser v4.0.0 software (10x Genomics). Sequencing data was then aligned to the mouse reference genome (mm10) using the spaceranger v1.0.0 pipeline to generate a feature-by-spot-barcode expression matrix (10x Genomics).

Download and alignment of single-cell RNA sequencing data. For all samples available via SRA, parallel-fastq-dump (github.com/rvalieris/parallel-fastq-dump) was used to download raw .fastq files. Samples which were only available as .bam files were converted to .fastq format using bamtofastq from 10x Genomics (github.com/10XGenomics/bamtofastq). Raw reads were aligned to the mm10 reference using cellranger (v3.1.0).

Preprocessing and batch correction of single-cell RNA sequencing datasets. First, ambient RNA signal was removed using the default SoupX (v1.4.5) workflow (autoEstCounts and adjustCounts; github.com/constantAmateur/SoupX). Samples were then preprocessed using the standard Seurat (v3.2.1) workflow (NormalizeData, ScaleData, FindVariableFeatures, RunPCA, FindNeighbors, FindClusters, and RunUMAP; github.com/satijalab/seurat). Cells with fewer than 750 features, fewer than 1000 transcripts, or more than 30% of unique transcripts derived from mitochondrial genes were removed. After preprocessing, DoubletFinder (v2.0) was used to identify putative doublets in each dataset, individually. BCmvn optimization was used for PK parameterization. Estimated doublet rates were computed by fitting the total number of cells after quality filtering to a linear regression of the expected doublet rates published in the 10x Chromium handbook. Estimated homotypic doublet rates were also accounted for using the modelHomotypic function. The default PN value (0.25) was used. Putative doublets were then removed from each individual dataset. After preprocessing and quality filtering, we merged the datasets and performed batch-correction with three tools, independently- Harmony (github.com/immunogenomics/harmony) (v1.0), Scanorama (github.com/brianhie/scanorama) (v1.3), and BBKNN (github.com/Teichlab/bbknn) (v1.3.12). We then used Seurat to process the integrated data. After initial integration, we removed the noisy cluster and re-integrated the data using each of the three batch-correction tools.

Cell type annotation. Cell types were determined for each integration method independently. For Harmony and Scanorama, dimensions accounting for 95% of the total variance were used to generate SNN graphs (Seurat::FindNeighbors). Louvain clustering was then performed on the output graphs (including the corrected graph output by BBKNN) using Seurat::FindClusters. A clustering resolution of 1.2 was used for Harmony (25 initial clusters), BBKNN (28 initial clusters), and Scanorama (38 initial clusters). Cell types were determined based on expression of canonical genes (Fig. S3). Clusters which had similar canonical marker gene expression patterns were merged.

Pseudotime workflow. Cells were subset based on the consensus cell types between all three integration methods. Harmony embedding values from the dimensions accounting for 95% of the total variance were used for further dimensional reduction with PHATE, using phateR (v1.0.4) (github.com/KrishnaswamyLab/phateR).

Deconvolution of spatial RNA sequencing spots. Spot deconvolution was performed using the deconvolution module in BayesPrism (previously known as “Tumor microEnvironment Deconvolution”, TED, v1.0; github.com/Danko-Lab/TED). First, myogenic cells were re-labeled, according to binning along the first PHATE dimension, as “Quiescent MuSCs” (bins 4-5), “Activated MuSCs” (bins 6-7), “Committed Myoblasts” (bins 8-10), and “Fusing Myoctes” (bins 11-18). Culture-associated muscle stem cells were ignored and myonuclei labels were retained as “Myonuclei (Type IIb)” and “Myonuclei (Type IIx)”. Next, highly and differentially expressed genes across the 25 groups of cells were identified with differential gene expression analysis using Seurat (FindAllMarkers, using Wilcoxon Rank Sum Test; results in Sup. Data 2). The resulting genes were filtered based on average log2-fold change (avg_logFC > 1) and the percentage of cells within the cluster which express each gene (pct.expressed > 0.5), yielding 1,069 genes. Mitochondrial and ribosomal protein genes were also removed from this list, in line with recommendations in the BayesPrism vignette. For each of the cell types, mean raw counts were calculated across the 1,069 genes to generate a gene expression profile for BayesPrism. Raw counts for each spot were then passed to the run.Ted function, using

Search
Clear search
Close search
Google apps
Main menu