--- title: "ASURAT" author: "Keita Iida" date: "`r Sys.Date()`" output: rmarkdown::html_vignette: toc: true toc_depth: 2 number_section: true fig_width: 6 fig_height: 5.4 vignette: > %\VignetteIndexEntry{ASURAT} %\VignetteEngine{knitr::rmarkdown} %\VignetteEncoding{UTF-8} --- ```{r, include = FALSE} knitr::opts_chunk$set( collapse = TRUE, comment = "#>" ) ``` # Installations Attach necessary libraries: ```{r, message = FALSE, warning = FALSE} library(ASURAT) library(SingleCellExperiment) library(SummarizedExperiment) ```
# Goal A goal of ASURAT is to cluster and characterize individual samples (cells) in terms of cell type (or disease), biological function, signaling pathway activity, and so on (see [here](#multilayered)).
# Quick start by SingleCellExperiment objects Having a SingleCellExperiment object (e.g., `sce`), one can use ASURAT by confirming the following requirements: * `assays(sce)` contains gene expression data with row and column names as variable (gene) and sample (cell), respectively, If `sce` contains normalized expression data (e.g., `assay(sce, "logcounts")`), set `assay(sce, "centered")` by subtracting the data with the mean expression levels across samples (cells). ```{r, eval = FALSE} mat <- as.matrix(assay(sce, "logcounts")) assay(sce, "centered") <- sweep(mat, 1, apply(mat, 1, mean), FUN = "-") ``` One may use a Seurat function `Seurat::as.SingleCellExperiment()` for converting Seurat objects into SingleCellExperiment ones. Now, ready for the next step [here](#sign).
# Preprocessing ## Prepare SingleCellExperiment objects Load single-cell RNA sequencing (scRNA-seq) data. ```{r, message = FALSE} sce <- TENxPBMCData::TENxPBMCData(dataset = c("pbmc4k")) pbmc_counts <- as.matrix(assay(sce, "counts")) rownames(pbmc_counts) <- make.unique(rowData(sce)$Symbol_TENx) colnames(pbmc_counts) <- make.unique(colData(sce)$Barcode) ``` Here, `pbmc_counts` is a read count table of peripheral blood mononuclear cells (PBMCs). Below is a head of `pbmc_counts`: ```{r} pbmc_counts[1:5, 1:3] ``` Create SingleCellExperiment objects by inputting gene expression data. ```{r} pbmc <- SingleCellExperiment(assays = list(counts = pbmc_counts), rowData = data.frame(gene = rownames(pbmc_counts)), colData = data.frame(cell = colnames(pbmc_counts))) ``` Check data sizes. ```{r} dim(pbmc) ```
## Control data quality Remove variables (genes) and samples (cells) with low quality, by processing the following three steps: 1. remove variables based on expression profiles across samples, 2. remove samples based on the numbers of reads and nonzero expressed variables, 3. remove variables based on the mean read counts across samples. First of all, add metadata for both variables and samples using ASURAT function `add_metadata()`. The arguments are * `sce`: SingleCellExperiment object, and * `mitochondria_symbol`: a string representing for mitochondrial genes. ```{r} pbmc <- add_metadata(sce = pbmc, mitochondria_symbol = "^MT-") ``` One can check the results in `rowData(sce)` and `colData(sce)` slots.
### Remove variables based on expression profiles ASURAT function `remove_variables()` removes variable (gene) data such that the numbers of non-zero expressing samples (cells) are less than `min_nsamples`. ```{r} pbmc <- remove_variables(sce = pbmc, min_nsamples = 10) ```
### Remove samples based on expression profiles Qualities of sample (cell) data are confirmed based on proper visualization of `colData(sce)`. ```{r} df <- data.frame(x = colData(pbmc)$nReads, y = colData(pbmc)$nGenes) ggplot2::ggplot() + ggplot2::geom_point(ggplot2::aes(x = df$x, y = df$y), size = 1, alpha = 1) + ggplot2::labs(x = "Number of reads", y = "Number of genes") + ggplot2::theme_classic(base_size = 20) ``` ```{r} df <- data.frame(x = colData(pbmc)$nReads, y = colData(pbmc)$percMT) ggplot2::ggplot() + ggplot2::geom_point(ggplot2::aes(x = df$x, y = df$y), size = 1, alpha = 1) + ggplot2::labs(x = "Number of reads", y = "Perc of MT reads") + ggplot2::theme_classic(base_size = 20) ``` ASURAT function `remove_samples()` removes sample (cell) data by setting cutoff values for the metadata. The arguments are * `sce`: SingleCellExperiment object, * `min_nReads` and `max_nReads`: minimum and maximum number of reads, * `min_nGenes` and `max_nGenes`: minimum and maximum number of non-zero expressed genes, and * `min_percMT` and `max_percMT`: minimum and maximum percent of reads that map to mitochondrial genes, respectively. If there is no mitochondrial genes, set them as `NULL`. ```{r} pbmc <- remove_samples(sce = pbmc, min_nReads = 5000, max_nReads = 20000, min_nGenes = 100, max_nGenes = 1e+10, min_percMT = 0, max_percMT = 10) ``` **Tips:** Take care not to set excessive values for the arguments of `remove_samples()`, or it may produce biased results. Note that `min_nReads = 5000` is somewhat large, which is used only for the tutorial.
### Remove variables based on the mean read counts Qualities of variable (gene) data are confirmed based on proper visualization of `rowData(sce)`. ```{r} df <- data.frame(x = 1:nrow(rowData(pbmc)), y = sort(rowData(pbmc)$nSamples, decreasing = TRUE)) ggplot2::ggplot() + ggplot2::scale_y_log10() + ggplot2::geom_point(ggplot2::aes(x = df$x, y = df$y), size = 1, alpha = 1) + ggplot2::labs(x = "Rank of genes", y = "Mean read counts") + ggplot2::theme_classic(base_size = 20) ``` ASURAT function `remove_variables_second()` removes variable (gene) data such that the mean read counts across samples are less than `min_meannReads`. ```{r} pbmc <- remove_variables_second(sce = pbmc, min_meannReads = 0.05) ```
## Normalize data Normalize data using log transformation with a pseudo count and center the data by subtracting with the mean expression levels across samples (cells). The resulting normalized-and-centered data are used for downstream analyses. Perform log-normalization with a pseudo count. ```{r} assay(pbmc, "logcounts") <- log(assay(pbmc, "counts") + 1) ``` Center each row data. ```{r} mat <- assay(pbmc, "logcounts") assay(pbmc, "centered") <- sweep(mat, 1, apply(mat, 1, mean), FUN = "-") ``` Set gene expression data into `altExp(sce)`. **Tips:** Take care not to use a slot name "log-normalized" for `altExp(sce)`, which may produce an error when using a Seurat (version 4.0.5) function `as.Seurat()` in the downstream analysis. ```{r} sname <- "logcounts" altExp(pbmc, sname) <- SummarizedExperiment(list(counts = assay(pbmc, sname))) ``` Add ENTREZ Gene IDs to `rowData(sce)`. ```{r, message = FALSE} dictionary <- AnnotationDbi::select(org.Hs.eg.db::org.Hs.eg.db, key = rownames(pbmc), columns = "ENTREZID", keytype = "SYMBOL") dictionary <- dictionary[!duplicated(dictionary$SYMBOL), ] rowData(pbmc)$geneID <- dictionary$ENTREZID ```
# Multifaceted sign analysis {#sign} Infer cell or disease types, biological functions, and signaling pathway activity at the single-cell level by inputting related databases. ASURAT transforms centered read count tables to functional feature matrices, termed sign-by-sample matrices (SSMs). Using SSMs, perform unsupervised clustering of samples (cells).
## Compute correlation matrices Prepare correlation matrices of gene expressions. ```{r} set.seed(1) nrand_samples <- 1000 mat <- t(as.matrix(assay(pbmc, "centered"))) ids <- sample(rownames(mat), nrand_samples, prob = NULL) cormat <- cor(mat[ids, ], method = "spearman") ```
## Load databases Load databases. ```{r} urlpath <- "https://github.com/keita-iida/ASURATDB/blob/main/genes2bioterm/" load(url(paste0(urlpath, "20201213_human_CO.rda?raw=TRUE"))) # CO load(url(paste0(urlpath, "20220308_human_MSigDB.rda?raw=TRUE"))) # MSigDB load(url(paste0(urlpath, "20201213_human_GO_red.rda?raw=TRUE"))) # GO load(url(paste0(urlpath, "20201213_human_KEGG.rda?raw=TRUE"))) # KEGG ``` These knowledge-based data were available from the following repositories: * [DOI:10.6084/m9.figshare.19102598](https://figshare.com/s/0599d2de970c2deb675c) * [Github ASURATDB](https://github.com/keita-iida/ASURATDB) Create a custom-built (CB) cell type-related database by combining different databases for analyzing human single-cell transcriptome data. ```{r} d <- list(human_CO[["cell"]], human_MSigDB[["cell"]]) res <- do.call("rbind", d) human_CB <- list(cell = res) ``` Add formatted databases to `metadata(sce)$sign`. ```{r} pbmcs <- list(CB = pbmc, GO = pbmc, KG = pbmc) metadata(pbmcs$CB) <- list(sign = human_CB[["cell"]]) metadata(pbmcs$GO) <- list(sign = human_GO[["BP"]]) metadata(pbmcs$KG) <- list(sign = human_KEGG[["pathway"]]) ```
## Create signs ASURAT function `remove_signs()` redefines functional gene sets for the input database by removing genes, which are not included in `rownames(sce)`, and further removes biological terms including too few or too many genes. The arguments are * `sce`: SingleCellExperiment object, * `min_ngenes`: minimal number of genes> 1 (the default value is 2), and * `max_ngenes`: maximal number of genes> 1 (the default value is 1000). ```{r} pbmcs$CB <- remove_signs(sce = pbmcs$CB, min_ngenes = 2, max_ngenes = 1000) pbmcs$GO <- remove_signs(sce = pbmcs$GO, min_ngenes = 2, max_ngenes = 1000) pbmcs$KG <- remove_signs(sce = pbmcs$KG, min_ngenes = 2, max_ngenes = 1000) ``` The results are stored in `metadata(sce)$sign`. ASURAT function `cluster_genes()` clusters functional gene sets using a correlation graph-based decomposition method, which produces strongly, variably, and weakly correlated gene sets (SCG, VCG, and WCG, respectively). The arguments are * `sce`: SingleCellExperiment object, * `cormat`: correlation matrix of gene expressions, * `th_posi` and `th_nega`: threshold values of positive and negative correlation coefficients, respectively. **Tips:** Empirically, typical values of `th_posi` and `th_nega` are $0.15 \le {\rm th{\_}posi} \le 0.4$ and $-0.4 \le {\rm th{\_}nega} \le -0.15$, but one cannot avoid trial and error for setting these values. An exhaustive parameter searching is time-consuming but helpful for obtaining interpretable results. ```{r} set.seed(1) pbmcs$CB <- cluster_genesets(sce = pbmcs$CB, cormat = cormat, th_posi = 0.30, th_nega = -0.30) set.seed(1) pbmcs$GO <- cluster_genesets(sce = pbmcs$GO, cormat = cormat, th_posi = 0.30, th_nega = -0.30) set.seed(1) pbmcs$KG <- cluster_genesets(sce = pbmcs$KG, cormat = cormat, th_posi = 0.20, th_nega = -0.20) ``` The results are stored in `metadata(sce)$sign`. ASURAT function `create_signs()` creates signs by the following criteria: 1. the number of genes in SCG>= `min_cnt_strg` (the default value is 2) and 2. the number of genes in VCG>= `min_cnt_vari` (the default value is 2), which are independently applied to SCGs and VCGs, respectively. **Tips:** Empirically, typical values of `min_cnt_strg` and `min_cnt_vari` are $2 \le {\rm min\_cnt\_strg} = {\rm min\_cnt\_vari} \le 4$, but one cannot avoid trial and error for setting these values. An exhaustive parameter searching is time-consuming but helpful for obtaining interpretable results. ```{r} pbmcs$CB <- create_signs(sce = pbmcs$CB, min_cnt_strg = 2, min_cnt_vari = 2) pbmcs$GO <- create_signs(sce = pbmcs$GO, min_cnt_strg = 4, min_cnt_vari = 4) pbmcs$KG <- create_signs(sce = pbmcs$KG, min_cnt_strg = 3, min_cnt_vari = 3) ``` The results are stored in `metadata(sce)$sign_all`, where "CorrType" indicates SCG or VCG, "Corr" means the average correlation coefficients of SCG or VCG, "CorrWeak" means the average correlation coefficients of WCG, "CorrGene" means SCG or VCG, and "WeakCorrGene" means WCG. The orders of gene symbols and ENTREZ IDs, separated by "/", are consistent. **Tips:** If one would like to recreate signs, reset the list of objects by, e.g., (`pbmcs <- list(CB = pbmc, GO = pbmc, KG = pbmc)`), and go back to `remove_signs()`.
## Select signs If signs have semantic similarity information, one can use ASURAT function `remove_signs_redundant()` for removing redundant sings using the semantic similarity matrices. The arguments are * `sce`: SingleCellExperiment object, * `similarity_matrix`: a semantic similarity matrix, * `threshold`: a threshold value of semantic similarity, used for regarding biological terms as similar ones, and * `keep_rareID`: if `TRUE`, biological terms with the larger ICs are kept. **Tips:** The optimal value of `threshold` depends on the ontology structure as well as the method for computing semantic similarity matrix. ```{r} pbmcs$GO <- remove_signs_redundant(sce = pbmcs$GO, similarity_matrix = human_GO$similarity_matrix$BP, threshold = 0.70, keep_rareID = TRUE) ``` The results are stored in `metadata(sce)$sign_SCG`, `metadata(sce)$sign_VCG`, `metadata(sce)$sign_all`, and if there exist, `metadata(sce)$sign_SCG_redundant` and `metadata(sce)$sign_VCG_redundant`. ASURAT function `remove_signs_manually()` removes signs by specifying IDs (e.g., `GOID:XXX`) or descriptions (e.g., `COVID`) using `grepl()`. The arguments are `sce` and `keywords` (keywords separated by `|`). ```{r} keywords <- "Covid|COVID" pbmcs$KG <- remove_signs_manually(sce = pbmcs$KG, keywords = keywords) ``` The results are stored in `metadata(sce)$sign_SCG`, `metadata(sce)$sign_VCG`, and `metadata(sce)$sign_all`. There is another ASURAT function `select_signs_manually()`, a counter part of `remove_signs_manually()`, which removes signs that do not include `keywords` (keywords separated by `|`). ```{r, eval = FALSE} keywords <- "cell|cyte" test <- select_signs_manually(sce = pbmcs$CB, keywords = keywords) ``` The results are stored in `metadata(sce)$sign_SCG`, `metadata(sce)$sign_VCG`, and `metadata(sce)$sign_all`.
## Create sign-by-sample matrices ASURAT function `create_sce_signmatrix()` creates a new SingleCellExperiment object `new_sce`, consisting of the following information: * `assayNames(new_sce)`: counts (SSM whose entries are termed sign scores), * `names(colData(new_sce))`: nReads, nGenes, percMT, * `names(rowData(new_sce))`: ParentSignID, Description, CorrGene, etc., * `names(metadata(new_sce))`: sign_SCG, sign_VCG, etc., * `altExpNames(new_sce)`: something if there is data in `altExp(sce)`. The arguments are * `sce`: SingleCellExperiment object, * `weight_strg`: weight parameter for SCG (the default value is 0.5), and * `weight_vari`: weight parameter for VCG (the default is 0.5). ```{r} pbmcs$CB <- makeSignMatrix(sce = pbmcs$CB, weight_strg = 0.5, weight_vari = 0.5) pbmcs$GO <- makeSignMatrix(sce = pbmcs$GO, weight_strg = 0.5, weight_vari = 0.5) pbmcs$KG <- makeSignMatrix(sce = pbmcs$KG, weight_strg = 0.5, weight_vari = 0.5) ``` Below are head and tail of `assay(sce, "counts")`: ```{r} rbind(head(assay(pbmcs$CB, "counts")[, 1:3], n = 4), tail(assay(pbmcs$CB, "counts")[, 1:3], n = 4)) ```
## Reduce dimensions of sign-by-sample matrices Perform principal component analysis and t-distributed stochastic neighbor embedding (t-SNE). ```{r} pca_dims <- c(30, 30, 50) tsne_dims <- c(2, 2, 3) for(i in seq_along(pbmcs)){ set.seed(1) mat <- t(as.matrix(assay(pbmcs[[i]], "counts"))) res <- Rtsne::Rtsne(mat, dim = tsne_dims[i], pca = TRUE, initial_dims = pca_dims[i]) reducedDim(pbmcs[[i]], "TSNE") <- res[["Y"]] } ``` Show the results of dimensional reduction in t-SNE spaces. ```{r} df <- as.data.frame(reducedDim(pbmcs$CB, "TSNE")) ggplot2::ggplot() + ggplot2::geom_point(ggplot2::aes(x = df[, 1], y = df[, 2]), color = "black", size = 1, alpha = 1) + ggplot2::labs(title = "PBMC (cell type)", x = "tSNE_1", y = "tSNE_2") + ggplot2::theme_classic(base_size = 15) ``` ```{r} df <- as.data.frame(reducedDim(pbmcs$GO, "TSNE")) ggplot2::ggplot() + ggplot2::geom_point(ggplot2::aes(x = df[, 1], y = df[, 2]), color = "black", size = 1, alpha = 1) + ggplot2::labs(title = "PBMC (function)", x = "tSNE_1", y = "tSNE_2") + ggplot2::theme_classic(base_size = 15) ``` Use ASURAT function `plot_dataframe3D()` for plotting three-dimensional data. See `?plot_dataframe3D` for details. ```{r} df <- as.data.frame(reducedDim(pbmcs$KG, "TSNE")) plot_dataframe3D(dataframe3D = df, theta = 45, phi = 30, title = "PBMC (pathway)", xlabel = "tSNE_1", ylabel = "tSNE_2", zlabel = "tSNE_3") ```
## Cluster cells ### Use Seurat functions To date (December, 2021), one of the most useful clustering methods in scRNA-seq data analysis is a combination of a community detection algorithm and graph-based unsupervised clustering, developed in Seurat package. Here, our strategy is as follows: 1. convert SingleCellExperiment objects into Seurat objects (note that `rowData()` and `colData()` must have data), 2. perform `ScaleData()`, `RunPCA()`, `FindNeighbors()`, and `FindClusters()`, 3. convert Seurat objects into temporal SingleCellExperiment objects `temp`, 4. add `colData(temp)$seurat_clusters` into `colData(sce)$seurat_clusters`. ```{r, message = FALSE, warning = FALSE, results = "hide"} resolutions <- c(0.20, 0.20, 0.10) ds <- list(seq_len(20), seq_len(20), seq_len(20)) for(i in seq_along(pbmcs)){ surt <- Seurat::as.Seurat(pbmcs[[i]], counts = "counts", data = "counts") mat <- as.matrix(assay(pbmcs[[i]], "counts")) surt[["SSM"]] <- Seurat::CreateAssayObject(counts = mat) Seurat::DefaultAssay(surt) <- "SSM" surt <- Seurat::ScaleData(surt, features = rownames(surt)) surt <- Seurat::RunPCA(surt, features = rownames(surt)) surt <- Seurat::FindNeighbors(surt, reduction = "pca", dims = ds[[i]]) surt <- Seurat::FindClusters(surt, resolution = resolutions[i]) temp <- Seurat::as.SingleCellExperiment(surt) colData(pbmcs[[i]])$seurat_clusters <- colData(temp)$seurat_clusters } ``` Show the clustering results in t-SNE spaces. ```{r} labels <- colData(pbmcs$CB)$seurat_clusters df <- as.data.frame(reducedDim(pbmcs$CB, "TSNE")) ggplot2::ggplot() + ggplot2::geom_point(ggplot2::aes(x = df[, 1], y = df[, 2], color = labels), size = 1, alpha = 1) + ggplot2::labs(title = "PBMC (cell type)", x = "tSNE_1", y = "tSNE_2", color = "") + ggplot2::theme_classic(base_size = 15) + ggplot2::scale_colour_hue() + ggplot2::guides(colour = ggplot2::guide_legend(override.aes = list(size = 4))) ``` ```{r} labels <- colData(pbmcs$GO)$seurat_clusters df <- as.data.frame(reducedDim(pbmcs$GO, "TSNE")) ggplot2::ggplot() + ggplot2::geom_point(ggplot2::aes(x = df[, 1], y = df[, 2], color = labels), size = 1, alpha = 1) + ggplot2::labs(title = "PBMC (function)", x = "tSNE_1", y = "tSNE_2", color = "") + ggplot2::theme_classic(base_size = 15) + ggplot2::scale_colour_brewer(palette = "Set1") + ggplot2::guides(colour = ggplot2::guide_legend(override.aes = list(size = 4))) ``` Use ASURAT function `plot_dataframe3D()` for plotting three-dimensional data. See `?plot_dataframe3D` for details. ```{r} labels <- colData(pbmcs$KG)$seurat_clusters colors <- scales::brewer_pal(palette = "Set2")(length(unique(labels)))[labels] df <- as.data.frame(reducedDim(pbmcs$KG, "TSNE")[, seq_len(3)]) plot_dataframe3D(dataframe3D = df, labels = labels, colors = colors, theta = 45, phi = 30, title = "PBMC (pathway)", xlabel = "tSNE_1", ylabel = "tSNE_2", zlabel = "tSNE_3") ```
### Cell cycle inference using Seurat functions If there is gene expression data in `altExp(sce)`, one can infer cell cycle phases by using Seurat functions in the similar manner as above. ```{r, message = FALSE, warning = FALSE} surt <- Seurat::as.Seurat(pbmcs$CB, counts = "counts", data = "counts") mat <- as.matrix(assay(altExp(pbmcs$CB), "counts")) surt[["GEM"]] <- Seurat::CreateAssayObject(counts = mat) Seurat::DefaultAssay(surt) <- "GEM" surt <- Seurat::ScaleData(surt, features = rownames(surt)) surt <- Seurat::RunPCA(surt, features = rownames(surt)) surt <- Seurat::CellCycleScoring(surt, s.features = Seurat::cc.genes$s.genes, g2m.features = Seurat::cc.genes$g2m.genes) temp <- Seurat::as.SingleCellExperiment(surt) colData(pbmcs$CB)$Phase <- colData(temp)$Phase ```
## Investigate significant signs Significant signs are analogous to differentially expressed genes but bear biological meanings. Note that naïve usages of statistical tests should be avoided because the row vectors of SSMs are centered. Instead, ASURAT function `compute_sepI_all()` computes separation indices for each cluster against the others. Briefly, a separation index "sepI", ranging from -1 to 1, is a nonparametric measure of significance of a given sign score for a given subpopulation. The larger (resp. smaller) sepI is, the more reliable the sign is as a positive (resp. negative) marker for the cluster. The arguments are * `sce`: SingleCellExperiment object, * `labels`: a vector of labels of all the samples, and * `nrand_samples`: an integer for the number of samples used for random sampling, which samples at least one sample per cluster. ```{r, message = FALSE, results = "hide"} for(i in seq_along(pbmcs)){ set.seed(1) labels <- colData(pbmcs[[i]])$seurat_clusters pbmcs[[i]] <- compute_sepI_all(sce = pbmcs[[i]], labels = labels, nrand_samples = 200) } ``` The results are stored in `metadata(sce)$marker_signs`. When computing separation indices between given clusters, e.g., cluster 1 versus clusters 2 and 3, use an ASURAT function `compute_sepI_clusters()`. See `?compute_sepI_clusters` for details.
## Investigate significant genes ### Use Seurat function To date (December, 2021), one of the most useful methods of multiple statistical tests in scRNA-seq data analysis is to use a Seurat function `FindAllMarkers()`. If there is gene expression data in `altExp(sce)`, one can investigate differentially expressed genes by using Seurat functions in the similar manner as described before. ```{r, message = FALSE} set.seed(1) surt <- Seurat::as.Seurat(pbmcs$CB, counts = "counts", data = "counts") mat <- as.matrix(assay(altExp(pbmcs$CB), "counts")) surt[["GEM"]] <- Seurat::CreateAssayObject(counts = mat) Seurat::DefaultAssay(surt) <- "GEM" surt <- Seurat::SetIdent(surt, value = "seurat_clusters") res <- Seurat::FindAllMarkers(surt, only.pos = TRUE, min.pct = 0.50, logfc.threshold = 0.50) metadata(pbmcs$CB)$marker_genes$all <- res ```
## Multifaceted analysis {#multilayered} Simultaneously analyze multiple sign-by-sample matrices, which helps us characterize individual samples (cells) from multiple biological aspects. ASURAT function `plot_multiheatmaps()` shows heatmaps (ComplexHeatmap object) of sign scores and gene expression levels (if there are), where rows and columns stand for sign (or gene) and sample (cell), respectively. See `?plot_multiheatmaps` for details. First, remove unrelated signs by setting keywords, followed by selecting top significant signs and genes for the clustering results with respect to separation index and adjusted p-value, respectively. ```{r} # Significant signs marker_signs <- list() keywords <- "MESENCHYMAL|LIMB|PANCREAS" for(i in seq_along(pbmcs)){ marker_signs[[i]] <- metadata(pbmcs[[i]])$marker_signs$all marker_signs[[i]] <- marker_signs[[i]][!grepl(keywords, marker_signs[[i]]$Description), ] marker_signs[[i]] <- dplyr::group_by(marker_signs[[i]], Ident_1) marker_signs[[i]] <- dplyr::slice_max(marker_signs[[i]], sepI, n = 2) marker_signs[[i]] <- dplyr::slice_min(marker_signs[[i]], Rank, n = 1) } # Significant genes marker_genes_CB <- metadata(pbmcs$CB)$marker_genes$all marker_genes_CB <- dplyr::group_by(marker_genes_CB, cluster) marker_genes_CB <- dplyr::slice_min(marker_genes_CB, p_val_adj, n = 2) marker_genes_CB <- dplyr::slice_max(marker_genes_CB, avg_log2FC, n = 1) ``` Next, prepare the arguments. ```{r} # ssm_list sces_sub <- list() ; ssm_list <- list() for(i in seq_along(pbmcs)){ sces_sub[[i]] <- pbmcs[[i]][rownames(pbmcs[[i]]) %in% marker_signs[[i]]$SignID, ] ssm_list[[i]] <- assay(sces_sub[[i]], "counts") } names(ssm_list) <- c("SSM_cell", "SSM_function", "SSM_pathway") # gem_list expr_sub <- altExp(pbmcs$CB, "logcounts") expr_sub <- expr_sub[rownames(expr_sub) %in% marker_genes_CB$gene] gem_list <- list(GeneExpr = assay(expr_sub, "counts")) # ssmlabel_list labels <- list() ; ssmlabel_list <- list() for(i in seq_along(pbmcs)){ tmp <- colData(sces_sub[[i]])$seurat_clusters labels[[i]] <- data.frame(label = tmp) n_groups <- length(unique(tmp)) if(i == 1){ labels[[i]]$color <- scales::hue_pal()(n_groups)[tmp] }else if(i == 2){ labels[[i]]$color <- scales::brewer_pal(palette = "Set1")(n_groups)[tmp] }else if(i == 3){ labels[[i]]$color <- scales::brewer_pal(palette = "Set2")(n_groups)[tmp] } ssmlabel_list[[i]] <- labels[[i]] } names(ssmlabel_list) <- c("Label_cell", "Label_function", "Label_pathway") # gemlabel_list label_CC <- data.frame(label = colData(pbmcs$CB)$Phase, color = NA) gemlabel_list <- list(CellCycle = label_CC) ``` Show heatmaps for the selected signs and genes. ```{r, message = FALSE, warning = FALSE} set.seed(1) plot_multiheatmaps(ssm_list = ssm_list, gem_list = gem_list, ssmlabel_list = ssmlabel_list, gemlabel_list = gemlabel_list, nrand_samples = 100, show_row_names = TRUE, title = "PBMC") ``` Show violin plots for sign score distributions across cell type-related clusters. ```{r} labels <- colData(pbmcs$CB)$seurat_clusters variable <- "GO:0042100-V" description <- "B cell proliferation" subsce <- pbmcs$GO[which(rownames(pbmcs$GO) == variable), ] df <- as.data.frame(t(as.matrix(assay(subsce, "counts")))) ggplot2::ggplot() + ggplot2::geom_violin(ggplot2::aes(x = as.factor(labels), y = df[, 1], fill = labels), trim = FALSE, size = 0.5) + ggplot2::geom_boxplot(ggplot2::aes(x = as.factor(labels), y = df[, 1]), width = 0.15, alpha = 0.6) + ggplot2::labs(title = paste0(variable, "\n", description), x = "Cluster (cell type)", y = "Sign score") + ggplot2::theme_classic(base_size = 20) + ggplot2::theme(legend.position = "none") + ggplot2::scale_fill_hue() ``` Show violin plots for gene expression distributions across cell type-related clusters. ```{r} vname <- "CD79A" sub <- altExp(pbmcs$CB, "logcounts") sub <- sub[rownames(sub) == vname, ] labels <- colData(pbmcs$CB)$seurat_clusters df <- as.data.frame(t(assay(sub, "counts"))) ggplot2::ggplot() + ggplot2::geom_violin(ggplot2::aes(x = as.factor(labels), y = df[, 1], fill = labels), trim = FALSE, size = 0.5) + ggplot2::geom_boxplot(ggplot2::aes(x = as.factor(labels), y = df[, 1]), width = 0.15, alpha = 0.6) + ggplot2::labs(title = vname, x = "Cluster (cell type)", y = "Normalized expression") + ggplot2::theme_classic(base_size = 20) + ggplot2::theme(legend.position = "none") + ggplot2::scale_fill_hue() ```
# Session information ```{r} sessionInfo() ```