seurat subset analysis

A very comprehensive tutorial can be found on the Trapnell lab website. [148] sf_1.0-2 shiny_1.6.0, # First split the sample by original identity, # perform standard preprocessing on each object. Why is there a voltage on my HDMI and coaxial cables? i, features. Next, we apply a linear transformation (scaling) that is a standard pre-processing step prior to dimensional reduction techniques like PCA. Scaling is an essential step in the Seurat workflow, but only on genes that will be used as input to PCA. To ensure our analysis was on high-quality cells . It has been downloaded in the course uppmax folder with subfolder: scrnaseq_course/data/PBMC_10x/pbmc3k_filtered_gene_bc_matrices.tar.gz We do this using a regular expression as in mito.genes <- grep(pattern = "^MT-". In the example below, we visualize gene and molecule counts, plot their relationship, and exclude cells with a clear outlier number of genes detected as potential multiplets. RDocumentation. Browse other questions tagged, Where developers & technologists share private knowledge with coworkers, Reach developers & technologists worldwide. Takes either a list of cells to use as a subset, or a parameter (for example, a gene), to subset on. ident.use = NULL, covariate, Calculate the variance to mean ratio of logged values, Aggregate expression of multiple features into a single feature, Apply a ceiling and floor to all values in a matrix, Calculate the percentage of a vector above some threshold, Calculate the percentage of all counts that belong to a given set of features, Descriptions of data included with Seurat, Functions included for user convenience and to keep maintain backwards compatability, Functions re-exported from other packages, reexports AddMetaData as.Graph as.Neighbor as.Seurat as.sparse Assays Cells CellsByIdentities Command CreateAssayObject CreateDimReducObject CreateSeuratObject DefaultAssay DefaultAssay Distances Embeddings FetchData GetAssayData GetImage GetTissueCoordinates HVFInfo Idents Idents Images Index Index Indices IsGlobal JS JS Key Key Loadings Loadings LogSeuratCommand Misc Misc Neighbors Project Project Radius Reductions RenameCells RenameIdents ReorderIdent RowMergeSparseMatrices SetAssayData SetIdent SpatiallyVariableFeatures StashIdent Stdev SVFInfo Tool Tool UpdateSeuratObject VariableFeatures VariableFeatures WhichCells. We and others have found that focusing on these genes in downstream analysis helps to highlight biological signal in single-cell datasets. In other words, is this workflow valid: SCT_not_integrated <- FindClusters(SCT_not_integrated) The clusters can be found using the Idents() function. DimPlot uses UMAP by default, with Seurat clusters as identity: In order to control for clustering resolution and other possible artifacts, we will take a close look at two minor cell populations: 1) dendritic cells (DCs), 2) platelets, aka thrombocytes. Run a custom distance function on an input data matrix, Calculate the standard deviation of logged values, Compute the correlation of features broken down by groups with another But I especially don't get why this one did not work: If anyone can tell me why the latter did not function I would appreciate it. By default, we return 2,000 features per dataset. Learn more about Stack Overflow the company, and our products. Importantly, the distance metric which drives the clustering analysis (based on previously identified PCs) remains the same. User Agreement and Privacy myseurat@meta.data[which(myseurat@meta.data$celltype=="AT1")[1],]. Single SCTransform command replaces NormalizeData, ScaleData, and FindVariableFeatures. You can save the object at this point so that it can easily be loaded back in without having to rerun the computationally intensive steps performed above, or easily shared with collaborators. As you will observe, the results often do not differ dramatically. Furthermore, it is possible to apply all of the described algortihms to selected subsets (resulting cluster . Dendritic cell and NK aficionados may recognize that genes strongly associated with PCs 12 and 13 define rare immune subsets (i.e. Try updating the resolution parameter to generate more clusters (try 1e-5, 1e-3, 1e-1, and 0). accept.value = NULL, Disconnect between goals and daily tasksIs it me, or the industry? random.seed = 1, Lets see if we have clusters defined by any of the technical differences. SEURAT provides agglomerative hierarchical clustering and k-means clustering. This vignette should introduce you to some typical tasks, using Seurat (version 3) eco-system. This can in some cases cause problems downstream, but setting do.clean=T does a full subset. Sign up for a free GitHub account to open an issue and contact its maintainers and the community. # hpca.ref <- celldex::HumanPrimaryCellAtlasData(), # dice.ref <- celldex::DatabaseImmuneCellExpressionData(), # hpca.main <- SingleR(test = sce,assay.type.test = 1,ref = hpca.ref,labels = hpca.ref$label.main), # hpca.fine <- SingleR(test = sce,assay.type.test = 1,ref = hpca.ref,labels = hpca.ref$label.fine), # dice.main <- SingleR(test = sce,assay.type.test = 1,ref = dice.ref,labels = dice.ref$label.main), # dice.fine <- SingleR(test = sce,assay.type.test = 1,ref = dice.ref,labels = dice.ref$label.fine), # srat@meta.data$hpca.main <- hpca.main$pruned.labels, # srat@meta.data$dice.main <- dice.main$pruned.labels, # srat@meta.data$hpca.fine <- hpca.fine$pruned.labels, # srat@meta.data$dice.fine <- dice.fine$pruned.labels. In order to perform a k-means clustering, the user has to choose this from the available methods and provide the number of desired sample and gene clusters. It may make sense to then perform trajectory analysis on each partition separately. (i) It learns a shared gene correlation. Our procedure in Seurat is described in detail here, and improves on previous versions by directly modeling the mean-variance relationship inherent in single-cell data, and is implemented in the FindVariableFeatures() function. The best answers are voted up and rise to the top, Not the answer you're looking for? Many thanks in advance. First, lets set the active assay back to RNA, and re-do the normalization and scaling (since we removed a notable fraction of cells that failed QC): The following function allows to find markers for every cluster by comparing it to all remaining cells, while reporting only the positive ones. Making statements based on opinion; back them up with references or personal experience. Lets add the annotations to the Seurat object metadata so we can use them: Finally, lets visualize the fine-grained annotations. ident.remove = NULL, Sign in Both vignettes can be found in this repository. Step 1: Find the T cells with CD3 expression To sub-cluster T cells, we first need to identify the T-cell population in the data. There are 2,700 single cells that were sequenced on the Illumina NextSeq 500. Staging Ground Beta 1 Recap, and Reviewers needed for Beta 2, R: subsetting data frame by both certain column names (as a variable) and field values. In a data set like this one, cells were not harvested in a time series, but may not have all been at the same developmental stage. You signed in with another tab or window. I want to subset from my original seurat object (BC3) meta.data based on orig.ident. To cluster the cells, we next apply modularity optimization techniques such as the Louvain algorithm (default) or SLM [SLM, Blondel et al., Journal of Statistical Mechanics], to iteratively group cells together, with the goal of optimizing the standard modularity function. [10] htmltools_0.5.1.1 viridis_0.6.1 gdata_2.18.0 Normalized data are stored in srat[['RNA']]@data of the RNA assay. In this example, we can observe an elbow around PC9-10, suggesting that the majority of true signal is captured in the first 10 PCs. More, # approximate techniques such as those implemented in ElbowPlot() can be used to reduce, # Look at cluster IDs of the first 5 cells, # If you haven't installed UMAP, you can do so via reticulate::py_install(packages =, # note that you can set `label = TRUE` or use the LabelClusters function to help label, # find all markers distinguishing cluster 5 from clusters 0 and 3, # find markers for every cluster compared to all remaining cells, report only the positive, Analysis, visualization, and integration of spatial datasets with Seurat, Fast integration using reciprocal PCA (RPCA), Integrating scRNA-seq and scATAC-seq data, Demultiplexing with hashtag oligos (HTOs), Interoperability between single-cell object formats, [SNN-Cliq, Xu and Su, Bioinformatics, 2015]. A stupid suggestion, but did you try to give it as a string ? The min.pct argument requires a feature to be detected at a minimum percentage in either of the two groups of cells, and the thresh.test argument requires a feature to be differentially expressed (on average) by some amount between the two groups. By default, only the previously determined variable features are used as input, but can be defined using features argument if you wish to choose a different subset. Michochondrial genes are useful indicators of cell state. Seurat offers several non-linear dimensional reduction techniques, such as tSNE and UMAP, to visualize and explore these datasets. [19] globals_0.14.0 gmodels_2.18.1 R.utils_2.10.1 Differential expression allows us to define gene markers specific to each cluster. 3 Seurat Pre-process Filtering Confounding Genes. In this example, all three approaches yielded similar results, but we might have been justified in choosing anything between PC 7-12 as a cutoff. We start the analysis after two preliminary steps have been completed: 1) ambient RNA correction using soupX; 2) doublet detection using scrublet. For mouse datasets, change pattern to Mt-, or explicitly list gene IDs with the features = option. Platform: x86_64-apple-darwin17.0 (64-bit) The raw data can be found here. Can you help me with this? DoHeatmap() generates an expression heatmap for given cells and features. Similarly, cluster 13 is identified to be MAIT cells. To learn more, see our tips on writing great answers. If not, an easy modification to the workflow above would be to add something like the following before RunCCA: Similarly, we can define ribosomal proteins (their names begin with RPS or RPL), which often take substantial fraction of reads: Now, lets add the doublet annotation generated by scrublet to the Seurat object metadata. Differential expression can be done between two specific clusters, as well as between a cluster and all other cells. SubsetData( Automagically calculate a point size for ggplot2-based scatter plots, Determine text color based on background color, Plot the Barcode Distribution and Calculated Inflection Points, Move outliers towards center on dimension reduction plot, Color dimensional reduction plot by tree split, Combine ggplot2-based plots into a single plot, BlackAndWhite() BlueAndRed() CustomPalette() PurpleAndYellow(), DimPlot() PCAPlot() TSNEPlot() UMAPPlot(), Discrete colour palettes from the pals package, Visualize 'features' on a dimensional reduction plot, Boxplot of correlation of a variable (e.g. If I decide that batch correction is not required for my samples, could I subset cells from my original Seurat Object (after running Quality Control and clustering on it), set the assay to "RNA", and and run the standard SCTransform pipeline. [127] promises_1.2.0.1 KernSmooth_2.23-20 gridExtra_2.3 Maximum modularity in 10 random starts: 0.7424 Therefore, the default in ScaleData() is only to perform scaling on the previously identified variable features (2,000 by default). Lets remove the cells that did not pass QC and compare plots. This can in some cases cause problems downstream, but setting do.clean=T does a full subset. Use regularized negative binomial regression to normalize UMI count data, Subset a Seurat Object based on the Barcode Distribution Inflection Points, Functions for testing differential gene (feature) expression, Gene expression markers for all identity classes, Finds markers that are conserved between the groups, Gene expression markers of identity classes, Prepare object to run differential expression on SCT assay with multiple models, Functions to reduce the dimensionality of datasets. We start by reading in the data. Higher resolution leads to more clusters (default is 0.8). Why are physically impossible and logically impossible concepts considered separate in terms of probability? To perform the analysis, Seurat requires the data to be present as a seurat object. By clicking Sign up for GitHub, you agree to our terms of service and Batch split images vertically in half, sequentially numbering the output files. What does data in a count matrix look like? Because partitions are high level separations of the data (yes we have only 1 here). We will be using Monocle3, which is still in the beta phase of its development and hasnt been updated in a few years. Setting cells to a number plots the extreme cells on both ends of the spectrum, which dramatically speeds plotting for large datasets. Note that the plots are grouped by categories named identity class. These will be further addressed below. 'Seurat' aims to enable users to identify and interpret sources of heterogeneity from single cell transcriptomic measurements, and to integrate diverse types of single cell data. The goal of these algorithms is to learn the underlying manifold of the data in order to place similar cells together in low-dimensional space. using FetchData, Low cutoff for the parameter (default is -Inf), High cutoff for the parameter (default is Inf), Returns cells with the subset name equal to this value, Create a cell subset based on the provided identity classes, Subtract out cells from these identity classes (used for # Initialize the Seurat object with the raw (non-normalized data). In this tutorial, we will learn how to Read 10X sequencing data and change it into a seurat object, QC and selecting cells for further analysis, Normalizing the data, Identification . 4 Visualize data with Nebulosa. Seurat has a built-in list, cc.genes (older) and cc.genes.updated.2019 (newer), that defines genes involved in cell cycle. vegan) just to try it, does this inconvenience the caterers and staff? [106] RSpectra_0.16-0 lattice_0.20-44 Matrix_1.3-4 However, this isnt required and the same behavior can be achieved with: We next calculate a subset of features that exhibit high cell-to-cell variation in the dataset (i.e, they are highly expressed in some cells, and lowly expressed in others). However, our approach to partitioning the cellular distance matrix into clusters has dramatically improved. Setup the Seurat Object For this tutorial, we will be analyzing the a dataset of Peripheral Blood Mononuclear Cells (PBMC) freely available from 10X Genomics. How do I subset a Seurat object using variable features? As this is a guided approach, visualization of the earlier plots will give you a good idea of what these parameters should be. For trajectory analysis, partitions as well as clusters are needed and so the Monocle cluster_cells function must also be performed. Policy. The plots above clearly show that high MT percentage strongly correlates with low UMI counts, and usually is interpreted as dead cells. My code is GPL licensed, can I issue a license to have my code be distributed in a specific MIT licensed project? Seurat offers several non-linear dimensional reduction techniques, such as tSNE and UMAP, to visualize and explore these datasets. Matrix products: default By clicking Accept all cookies, you agree Stack Exchange can store cookies on your device and disclose information in accordance with our Cookie Policy. Renormalize raw data after merging the objects. Since most values in an scRNA-seq matrix are 0, Seurat uses a sparse-matrix representation whenever possible. Does a summoned creature play immediately after being summoned by a ready action? Normalized values are stored in pbmc[["RNA"]]@data. If starting from typical Cell Ranger output, its possible to choose if you want to use Ensemble ID or gene symbol for the count matrix. An alternative heuristic method generates an Elbow plot: a ranking of principle components based on the percentage of variance explained by each one (ElbowPlot() function). Why did Ukraine abstain from the UNHRC vote on China? GetAssay () Get an Assay object from a given Seurat object. Other option is to get the cell names of that ident and then pass a vector of cell names. remission@meta.data$sample <- "remission" On 26 Jun 2018, at 21:14, Andrew Butler > wrote: In the example below, we visualize gene and molecule counts, plot their relationship, and exclude cells with a clear outlier number of genes detected as potential multiplets. Seurat is one of the most popular software suites for the analysis of single-cell RNA sequencing data. Lets get reference datasets from celldex package. Find cells with highest scores for a given dimensional reduction technique, Find features with highest scores for a given dimensional reduction technique, TransferAnchorSet-class TransferAnchorSet, Update pre-V4 Assays generated with SCTransform in the Seurat to the new Lets convert our Seurat object to single cell experiment (SCE) for convenience. After this, using SingleR becomes very easy: Lets see the summary of general cell type annotations. Troubleshooting why subsetting of spatial object does not work, Automatic subsetting of a dataframe on the basis of a prediction matrix, transpose and rename dataframes in a for() loop in r, How do you get out of a corner when plotting yourself into a corner. We can look at the expression of some of these genes overlaid on the trajectory plot. [130] parallelly_1.27.0 codetools_0.2-18 gtools_3.9.2 For example, small cluster 17 is repeatedly identified as plasma B cells. columns in object metadata, PC scores etc. [22] spatstat.sparse_2.0-0 colorspace_2.0-2 ggrepel_0.9.1 low.threshold = -Inf, A detailed book on how to do cell type assignment / label transfer with singleR is available. Use MathJax to format equations. [124] raster_3.4-13 httpuv_1.6.2 R6_2.5.1 Identify the 10 most highly variable genes: Plot variable features with and without labels: ScaleData converts normalized gene expression to Z-score (values centered at 0 and with variance of 1). In Macosko et al, we implemented a resampling test inspired by the JackStraw procedure. For a technical discussion of the Seurat object structure, check out our GitHub Wiki. The ScaleData() function: This step takes too long! This indeed seems to be the case; however, this cell type is harder to evaluate. The main function from Nebulosa is the plot_density. Adjust the number of cores as needed. Note that you can change many plot parameters using ggplot2 features - passing them with & operator. Search all packages and functions. In our case a big drop happens at 10, so seems like a good initial choice: We can now do clustering. If, for example, the markers identified with cluster 1 suggest to you that cluster 1 represents the earliest developmental time point, you would likely root your pseudotime trajectory there. Lets set QC column in metadata and define it in an informative way. [76] tools_4.1.0 generics_0.1.0 ggridges_0.5.3 [11] S4Vectors_0.30.0 MatrixGenerics_1.4.2 We can also display the relationship between gene modules and monocle clusters as a heatmap. however, when i use subset(), it returns with Error. Set of genes to use in CCA. :) Thank you. other attached packages: Next-Generation Sequencing Analysis Resources, NGS Sequencing Technology and File Formats, Gene Set Enrichment Analysis with ClusterProfiler, Over-Representation Analysis with ClusterProfiler, Salmon & kallisto: Rapid Transcript Quantification for RNA-Seq Data, Instructions to install R Modules on Dalma, Prerequisites, data summary and availability, Deeptools2 computeMatrix and plotHeatmap using BioSAILs, Exercise part4 Alternative approach in R to plot and visualize the data, Seurat part 3 Data normalization and PCA, Loading your own data in Seurat & Reanalyze a different dataset, JBrowse: Visualizing Data Quickly & Easily. By clicking Sign up for GitHub, you agree to our terms of service and Why is this sentence from The Great Gatsby grammatical? We next use the count matrix to create a Seurat object. Linear discriminant analysis on pooled CRISPR screen data. Some cell clusters seem to have as much as 45%, and some as little as 15%. Comparing the labels obtained from the three sources, we can see many interesting discrepancies. Low-quality cells or empty droplets will often have very few genes, Cell doublets or multiplets may exhibit an aberrantly high gene count, Similarly, the total number of molecules detected within a cell (correlates strongly with unique genes), The percentage of reads that map to the mitochondrial genome, Low-quality / dying cells often exhibit extensive mitochondrial contamination, We calculate mitochondrial QC metrics with the, We use the set of all genes starting with, The number of unique genes and total molecules are automatically calculated during, You can find them stored in the object meta data, We filter cells that have unique feature counts over 2,500 or less than 200, We filter cells that have >5% mitochondrial counts, Shifts the expression of each gene, so that the mean expression across cells is 0, Scales the expression of each gene, so that the variance across cells is 1, This step gives equal weight in downstream analyses, so that highly-expressed genes do not dominate. [139] expm_0.999-6 mgcv_1.8-36 grid_4.1.0 This heatmap displays the association of each gene module with each cell type. To use subset on a Seurat object, (see ?subset.Seurat) , you have to provide: What you have should work, but try calling the actual function (in case there are packages that clash): Thanks for contributing an answer to Bioinformatics Stack Exchange! rev2023.3.3.43278. How can I check before my flight that the cloud separation requirements in VFR flight rules are met? [109] classInt_0.4-3 vctrs_0.3.8 LearnBayes_2.15.1 I have been using Seurat to do analysis of my samples which contain multiple cell types and I would now like to re-run the analysis only on 3 of the clusters, which I have identified as macrophage subtypes. By default, it identifies positive and negative markers of a single cluster (specified in ident.1), compared to all other cells. Functions for interacting with a Seurat object, Cells() Cells() Cells() Cells(), Get a vector of cell names associated with an image (or set of images). [43] pheatmap_1.0.12 DBI_1.1.1 miniUI_0.1.1.1 We randomly permute a subset of the data (1% by default) and rerun PCA, constructing a null distribution of feature scores, and repeat this procedure. However, these groups are so rare, they are difficult to distinguish from background noise for a dataset of this size without prior knowledge. The palettes used in this exercise were developed by Paul Tol. If your mitochondrial genes are named differently, then you will need to adjust this pattern accordingly (e.g. Seurat object summary shows us that 1) number of cells (samples) approximately matches Try setting do.clean=T when running SubsetData, this should fix the problem. There are many tests that can be used to define markers, including a very fast and intuitive tf-idf. Let's plot the kernel density estimate for CD4 as follows. How can this new ban on drag possibly be considered constitutional? This results in significant memory and speed savings for Drop-seq/inDrop/10x data. To subscribe to this RSS feed, copy and paste this URL into your RSS reader. Use of this site constitutes acceptance of our User Agreement and Privacy All cells that cannot be reached from a trajectory with our selected root will be gray, which represents infinite pseudotime. Literature suggests that blood MAIT cells are characterized by high expression of CD161 (KLRB1), and chemokines like CXCR6. By clicking Accept all cookies, you agree Stack Exchange can store cookies on your device and disclose information in accordance with our Cookie Policy. Hi Andrew, However, if I examine the same cell in the original Seurat object (myseurat), all the information is there. You may have an issue with this function in newer version of R an rBind Error. When we run SubsetData, we have (by default) not subsetted the raw.data slot as well, as this can be slow and usually unnecessary. cells = NULL, monocle3 uses a cell_data_set object, the as.cell_data_set function from SeuratWrappers can be used to convert a Seurat object to Monocle object. I am trying to subset the object based on cells being classified as a 'Singlet' under seurat_object@meta.data[["DF.classifications_0.25_0.03_252"]] and can achieve this by doing the following: I would like to automate this process but the _0.25_0.03_252 of DF.classifications_0.25_0.03_252 is based on values that are calculated and will not be known in advance.

What Percent Of Texas Speaks Spanish, Ryla Juice Shots Tacoma, Eagle Torch With Safe Stop, Articles S

seurat subset analysis