seurat subset analysis

[16] cluster_2.1.2 ROCR_1.0-11 remotes_2.4.0 filtration). Set of genes to use in CCA. Does anyone have an idea how I can automate the subset process? Making statements based on opinion; back them up with references or personal experience. # Identify the 10 most highly variable genes, # plot variable features with and without labels, # Examine and visualize PCA results a few different ways, # NOTE: This process can take a long time for big datasets, comment out for expediency. We can also calculate modules of co-expressed genes. number of UMIs) with expression I can figure out what it is by doing the following: Monocles graph_test() function detects genes that vary over a trajectory. To overcome the extensive technical noise in any single feature for scRNA-seq data, Seurat clusters cells based on their PCA scores, with each PC essentially representing a metafeature that combines information across a correlated feature set. # Lets examine a few genes in the first thirty cells, # The [[ operator can add columns to object metadata. How to notate a grace note at the start of a bar with lilypond? Now I think I found a good solution, taking a "meaningful" sample of the dataset, and then create a dendrogram-heatmap of the gene-gene correlation matrix generated from the sample. subset.name = NULL, For example, if you had very high coverage, you might want to adjust these parameters and increase the threshold window. So I was struggling with this: Creating a dendrogram with a large dataset (20,000 by 20,000 gene-gene correlation matrix): Is there a way to use multiple processors (parallelize) to create a heatmap for a large dataset? high.threshold = Inf, Perform Canonical Correlation Analysis RunCCA Seurat Perform Canonical Correlation Analysis Source: R/generics.R, R/dimensional_reduction.R Runs a canonical correlation analysis using a diagonal implementation of CCA. Default is INF. Use of this site constitutes acceptance of our User Agreement and Privacy Briefly, these methods embed cells in a graph structure - for example a K-nearest neighbor (KNN) graph, with edges drawn between cells with similar feature expression patterns, and then attempt to partition this graph into highly interconnected quasi-cliques or communities. 70 70 69 64 60 56 55 54 54 50 49 48 47 45 44 43 40 40 39 39 39 35 32 32 29 29 I want to subset from my original seurat object (BC3) meta.data based on orig.ident. To do this, omit the features argument in the previous function call, i.e. Asking for help, clarification, or responding to other answers. [97] compiler_4.1.0 plotly_4.9.4.1 png_0.1-7 Both cells and features are ordered according to their PCA scores. rev2023.3.3.43278. or suggest another approach? Seurat object summary shows us that 1) number of cells (samples) approximately matches The number above each plot is a Pearson correlation coefficient. . Linear discriminant analysis on pooled CRISPR screen 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. All cells that cannot be reached from a trajectory with our selected root will be gray, which represents infinite pseudotime. After removing unwanted cells from the dataset, the next step is to normalize the data. Try updating the resolution parameter to generate more clusters (try 1e-5, 1e-3, 1e-1, and 0). Now I think I found a good solution, taking a "meaningful" sample of the dataset, and then create a dendrogram-heatmap of the gene-gene correlation matrix generated from the sample. How does this result look different from the result produced in the velocity section? Yeah I made the sample column it doesnt seem to make a difference. It is very important to define the clusters correctly. Takes either a list of cells to use as a subset, or a Here, we analyze a dataset of 8,617 cord blood mononuclear cells (CBMCs), produced with CITE-seq, where we simultaneously measure the single cell transcriptomes alongside the expression of 11 surface proteins, whose levels are quantified with DNA-barcoded antibodies. However, when I try to do any of the following: I am at loss for how to perform conditional matching with the meta_data variable. 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). The output of this function is a table. privacy statement. The data from all 4 samples was combined in R v.3.5.2 using the Seurat package v.3.0.0 and an aggregate Seurat object was generated 21,22. Note that you can change many plot parameters using ggplot2 features - passing them with & operator. How do you feel about the quality of the cells at this initial QC step? 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. Optimal resolution often increases for larger datasets. This works for me, with the metadata column being called "group", and "endo" being one possible group there. The raw data can be found here. We identify significant PCs as those who have a strong enrichment of low p-value features. A value of 0.5 implies that the gene has no predictive . We find that setting this parameter between 0.4-1.2 typically returns good results for single-cell datasets of around 3K cells. Other option is to get the cell names of that ident and then pass a vector of cell names. Therefore, the default in ScaleData() is only to perform scaling on the previously identified variable features (2,000 by default). Determine statistical significance of PCA scores. [70] labeling_0.4.2 rlang_0.4.11 reshape2_1.4.4 SCTAssay class, as.Seurat() as.Seurat(), Convert objects to SingleCellExperiment objects, as.sparse() as.data.frame(), Functions for preprocessing single-cell data, Calculate the Barcode Distribution Inflection, Calculate pearson residuals of features not in the scale.data, Demultiplex samples based on data from cell 'hashing', Load a 10x Genomics Visium Spatial Experiment into a Seurat object, Demultiplex samples based on classification method from MULTI-seq (McGinnis et al., bioRxiv 2018), Load in data from remote or local mtx files. Moving the data calculated in Seurat to the appropriate slots in the Monocle object. Differential expression can be done between two specific clusters, as well as between a cluster and all other cells. Intuitive way of visualizing how feature expression changes across different identity classes (clusters). to your account. We also suggest exploring RidgePlot(), CellScatter(), and DotPlot() as additional methods to view your dataset. The finer cell types annotations are you after, the harder they are to get reliably. ), # S3 method for Seurat Creates a Seurat object containing only a subset of the cells in the original 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. 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 This will downsample each identity class to have no more cells than whatever this is set to. These features are still supported in ScaleData() in Seurat v3, i.e. [37] XVector_0.32.0 leiden_0.3.9 DelayedArray_0.18.0 Where does this (supposedly) Gibson quote come from? [11] S4Vectors_0.30.0 MatrixGenerics_1.4.2 [148] sf_1.0-2 shiny_1.6.0, # First split the sample by original identity, # perform standard preprocessing on each object. I am pretty new to Seurat. We therefore suggest these three approaches to consider. [34] polyclip_1.10-0 gtable_0.3.0 zlibbioc_1.38.0 4 Visualize data with Nebulosa. This is done using gene.column option; default is 2, which is gene symbol. [5] monocle3_1.0.0 SingleCellExperiment_1.14.1 Can I make it faster? 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. # 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. If so, how close was it? columns in object metadata, PC scores etc. As another option to speed up these computations, max.cells.per.ident can be set. Differential expression allows us to define gene markers specific to each cluster. Seurat vignettes are available here; however, they default to the current latest Seurat version (version 4). Is it known that BQP is not contained within NP? You are receiving this because you authored the thread. By clicking Sign up for GitHub, you agree to our terms of service and You signed in with another tab or window. Does a summoned creature play immediately after being summoned by a ready action? [3] SeuratObject_4.0.2 Seurat_4.0.3 However, we can try automaic annotation with SingleR is workflow-agnostic (can be used with Seurat, SCE, etc). [7] scattermore_0.7 ggplot2_3.3.5 digest_0.6.27 What is the difference between nGenes and nUMIs? How many clusters are generated at each level? matrix. Cheers. [55] bit_4.0.4 rsvd_1.0.5 htmlwidgets_1.5.3 RDocumentation. Sign in Eg, the name of a gene, PC_1, a To start the analysis, lets read in the SoupX-corrected matrices (see QC Chapter). This may run very slowly. To learn more, see our tips on writing great answers. Any argument that can be retreived There are also clustering methods geared towards indentification of rare cell populations. RDocumentation. [49] xtable_1.8-4 units_0.7-2 reticulate_1.20 Traffic: 816 users visited in the last hour. [79] evaluate_0.14 stringr_1.4.0 fastmap_1.1.0 Sign up for a free GitHub account to open an issue and contact its maintainers and the community. [61] ica_1.0-2 farver_2.1.0 pkgconfig_2.0.3 By clicking Accept all cookies, you agree Stack Exchange can store cookies on your device and disclose information in accordance with our Cookie Policy. For details about stored CCA calculation parameters, see PrintCCAParams. 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. We start the analysis after two preliminary steps have been completed: 1) ambient RNA correction using soupX; 2) doublet detection using scrublet. If some clusters lack any notable markers, adjust the clustering. 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. What is the point of Thrower's Bandolier? We do this using a regular expression as in mito.genes <- grep(pattern = "^MT-". To learn more, see our tips on writing great answers. While theCreateSeuratObjectimposes a basic minimum gene-cutoff, you may want to filter out cells at this stage based on technical or biological parameters. active@meta.data$sample <- "active" [9] GenomeInfoDb_1.28.1 IRanges_2.26.0 In this case it appears that there is a sharp drop-off in significance after the first 10-12 PCs. On 26 Jun 2018, at 21:14, Andrew Butler > wrote: [133] boot_1.3-28 MASS_7.3-54 assertthat_0.2.1 Not all of our trajectories are connected. To ensure our analysis was on high-quality cells . Single SCTransform command replaces NormalizeData, ScaleData, and FindVariableFeatures. The development branch however has some activity in the last year in preparation for Monocle3.1. [8] methods base By providing the module-finding function with a list of possible resolutions, we are telling Louvain to perform the clustering at each resolution and select the result with the greatest modularity. Is it possible to create a concave light? You signed in with another tab or window. [103] bslib_0.2.5.1 stringi_1.7.3 highr_0.9 Identifying the true dimensionality of a dataset can be challenging/uncertain for the user. To subscribe to this RSS feed, copy and paste this URL into your RSS reader. however, when i use subset(), it returns with Error. This choice was arbitrary. Biclustering is the simultaneous clustering of rows and columns of a data matrix. Error in cc.loadings[[g]] : subscript out of bounds. [94] grr_0.9.5 R.oo_1.24.0 hdf5r_1.3.3 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). 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). data, Visualize features in dimensional reduction space interactively, Label clusters on a ggplot2-based scatter plot, SeuratTheme() CenterTitle() DarkTheme() FontSize() NoAxes() NoLegend() NoGrid() SeuratAxes() SpatialTheme() RestoreLegend() RotatedAxis() BoldTitle() WhiteBackground(), Get the intensity and/or luminance of a color, Function related to tree-based analysis of identity classes, Phylogenetic Analysis of Identity Classes, Useful functions to help with a variety of tasks, Calculate module scores for feature expression programs in single cells, Aggregated feature expression by identity class, Averaged feature expression by identity class. 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. Monocle offers trajectory analysis to model the relationships between groups of cells as a trajectory of gene expression changes. other attached packages: Though clearly a supervised analysis, we find this to be a valuable tool for exploring correlated feature sets. If FALSE, merge the data matrices also. High ribosomal protein content, however, strongly anti-correlates with MT, and seems to contain biological signal. It may make sense to then perform trajectory analysis on each partition separately. To start the analysis, let's read in the SoupX -corrected matrices (see QC Chapter). Our approach was heavily inspired by recent manuscripts which applied graph-based clustering approaches to scRNA-seq data [SNN-Cliq, Xu and Su, Bioinformatics, 2015] and CyTOF data [PhenoGraph, Levine et al., Cell, 2015]. However, these groups are so rare, they are difficult to distinguish from background noise for a dataset of this size without prior knowledge. Seurat (version 3.1.4) . After learning the graph, monocle can plot add the trajectory graph to the cell plot. For a technical discussion of the Seurat object structure, check out our GitHub Wiki. Lets add several more values useful in diagnostics of cell quality. Bioinformatics Stack Exchange is a question and answer site for researchers, developers, students, teachers, and end users interested in bioinformatics. # Initialize the Seurat object with the raw (non-normalized data). Seurat allows you to easily explore QC metrics and filter cells based on any user-defined criteria. parameter (for example, a gene), to subset on. 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. I have a Seurat object, which has meta.data For mouse cell cycle genes you can use the solution detailed here. Each with their own benefits and drawbacks: Identification of all markers for each cluster: this analysis compares each cluster against all others and outputs the genes that are differentially expressed/present. 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]. I can figure out what it is by doing the following: Where meta_data = 'DF.classifications_0.25_0.03_252' and is a character class. Note that the plots are grouped by categories named identity class. Lets add the annotations to the Seurat object metadata so we can use them: Finally, lets visualize the fine-grained annotations. Sign up for a free GitHub account to open an issue and contact its maintainers and the community. This is where comparing many databases, as well as using individual markers from literature, would all be very valuable. It is conventional to use more PCs with SCTransform; the exact number can be adjusted depending on your dataset. For CellRanger reference GRCh38 2.0.0 and above, use cc.genes.updated.2019 (three genes were renamed: MLF1IP, FAM64A and HN1 became CENPU, PICALM and JPT). I keep running out of RAM with my current pipeline, Bar Graph of Expression Data from Seurat Object. Connect and share knowledge within a single location that is structured and easy to search. [121] bitops_1.0-7 irlba_2.3.3 Matrix.utils_0.9.8 Lets get a very crude idea of what the big cell clusters are. For detailed dissection, it might be good to do differential expression between subclusters (see below). We can look at the expression of some of these genes overlaid on the trajectory plot. FilterSlideSeq () Filter stray beads from Slide-seq puck. Is there a single-word adjective for "having exceptionally strong moral principles"? Functions related to the mixscape algorithm, DE and EnrichR pathway visualization barplot, Differential expression heatmap for mixscape. Visualize spatial clustering and expression data. We can see better separation of some subpopulations. For example, the ROC test returns the classification power for any individual marker (ranging from 0 - random, to 1 - perfect). [136] leidenbase_0.1.3 sctransform_0.3.2 GenomeInfoDbData_1.2.6 I think this is basically what you did, but I think this looks a little nicer. 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. For this tutorial, we will be analyzing the a dataset of Peripheral Blood Mononuclear Cells (PBMC) freely available from 10X Genomics. We start the analysis after two preliminary steps have been completed: 1) ambient RNA correction using soupX; 2) doublet detection using scrublet. max.cells.per.ident = Inf, How can this new ban on drag possibly be considered constitutional? trace(calculateLW, edit = T, where = asNamespace(monocle3)). In other words, is this workflow valid: SCT_not_integrated <- FindClusters(SCT_not_integrated) subset.AnchorSet.Rd. By clicking Post Your Answer, you agree to our terms of service, privacy policy and cookie policy. [64] R.methodsS3_1.8.1 sass_0.4.0 uwot_0.1.10 Seurat-package Seurat: Tools for Single Cell Genomics Description A toolkit for quality control, analysis, and exploration of single cell RNA sequencing data. ident.use = NULL, 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. Lets check the markers of smaller cell populations we have mentioned before - namely, platelets and dendritic cells. The Seurat alignment workflow takes as input a list of at least two scRNA-seq data sets, and briefly consists of the following steps ( Fig. Bulk update symbol size units from mm to map units in rule-based symbology. How can I remove unwanted sources of variation, as in Seurat v2? The object serves as a container that contains both data (like the count matrix) and analysis (like PCA, or clustering results) for a single-cell dataset. A stupid suggestion, but did you try to give it as a string ? Try setting do.clean=T when running SubsetData, this should fix the problem. Lucy Because we have not set a seed for the random process of clustering, cluster numbers will differ between R sessions. a clustering of the genes with respect to . Why is this sentence from The Great Gatsby grammatical? object, Both vignettes can be found in this repository. However, many informative assignments can be seen. There are many tests that can be used to define markers, including a very fast and intuitive tf-idf. [28] RCurl_1.98-1.4 jsonlite_1.7.2 spatstat.data_2.1-0 However, our approach to partitioning the cellular distance matrix into clusters has dramatically improved. 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. Default is to run scaling only on variable genes. [76] tools_4.1.0 generics_0.1.0 ggridges_0.5.3 Next, we apply a linear transformation (scaling) that is a standard pre-processing step prior to dimensional reduction techniques like PCA. For example, performing downstream analyses with only 5 PCs does significantly and adversely affect results. There are also differences in RNA content per cell type. Detailed signleR manual with advanced usage can be found here. [13] matrixStats_0.60.0 Biobase_2.52.0 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. Splits object into a list of subsetted objects. The raw data can be found here. This step is performed using the FindNeighbors() function, and takes as input the previously defined dimensionality of the dataset (first 10 PCs). Insyno.combined@meta.data is there a column called sample? 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. After this lets do standard PCA, UMAP, and clustering. Get a vector of cell names associated with an image (or set of images) CreateSCTAssayObject () Create a SCT Assay object. You can set both of these to 0, but with a dramatic increase in time - since this will test a large number of features that are unlikely to be highly discriminatory. 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. [142] rpart_4.1-15 coda_0.19-4 class_7.3-19 GetImage() GetImage() GetImage(), GetTissueCoordinates() GetTissueCoordinates() GetTissueCoordinates(), IntegrationAnchorSet-class IntegrationAnchorSet, Radius() Radius() Radius(), RenameCells() RenameCells() RenameCells() RenameCells(), levels() `levels<-`(). However, when i try to perform the alignment i get the following error.. FindAllMarkers() automates this process for all clusters, but you can also test groups of clusters vs.each other, or against all cells. [7] SummarizedExperiment_1.22.0 GenomicRanges_1.44.0 Running under: macOS Big Sur 10.16 Ribosomal protein genes show very strong dependency on the putative cell type! For greater detail on single cell RNA-Seq analysis, see the Introductory course materials here. 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 Right now it has 3 fields per celL: dataset ID, number of UMI reads detected per cell (nCount_RNA), and the number of expressed (detected) genes per same cell (nFeature_RNA). Literature suggests that blood MAIT cells are characterized by high expression of CD161 (KLRB1), and chemokines like CXCR6. RunCCA(object1, object2, .) Sign in Lets now load all the libraries that will be needed for the tutorial. Its stored in srat[['RNA']]@scale.data and used in following PCA. For example, we could regress out heterogeneity associated with (for example) cell cycle stage, or mitochondrial contamination. Is it plausible for constructed languages to be used to affect thought and control or mold people towards desired outcomes? 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. LAPACK: /Library/Frameworks/R.framework/Versions/4.1/Resources/lib/libRlapack.dylib Is the God of a monotheism necessarily omnipotent? Note that SCT is the active assay now. [25] xfun_0.25 dplyr_1.0.7 crayon_1.4.1 Chapter 3 Analysis Using Seurat. Normalized values are stored in pbmc[["RNA"]]@data. The third is a heuristic that is commonly used, and can be calculated instantly. Significant PCs will show a strong enrichment of features with low p-values (solid curve above the dashed line). [112] pillar_1.6.2 lifecycle_1.0.0 BiocManager_1.30.16 We advise users to err on the higher side when choosing this parameter. This results in significant memory and speed savings for Drop-seq/inDrop/10x data. The values in this matrix represent the number of molecules for each feature (i.e. If not, an easy modification to the workflow above would be to add something like the following before RunCCA: Stack Exchange network consists of 181 Q&A communities including Stack Overflow, the largest, most trusted online community for developers to learn, share their knowledge, and build their careers. low.threshold = -Inf, Creates a Seurat object containing only a subset of the cells in the original object. We also filter cells based on the percentage of mitochondrial genes present. These represent the selection and filtration of cells based on QC metrics, data normalization and scaling, and the detection of highly variable features. A detailed book on how to do cell type assignment / label transfer with singleR is available. How many cells did we filter out using the thresholds specified above. (i) It learns a shared gene correlation. Does Counterspell prevent from any further spells being cast on a given turn? The size of the dot encodes the percentage of cells within a class, while the color encodes the AverageExpression level across all cells within a class (blue is high). (default), then this list will be computed based on the next three Note: In order to detect mitochondrial genes, we need to tell Seurat how to distinguish these genes.

Benelli M4 Collapsible Stock California, Capital Grille Garden City Closing, Mobile, Alabama Obituaries 2021, Portland Timbers Odp Roster, Evan Williams Bottled In Bond Vs 1783, Articles S

seurat subset analysis