Welcome to the blog

Posts

My thoughts and ideas

Quality Assessment/Clustering | Griffith Lab

RNA-seq Bioinformatics

Introduction to bioinformatics for RNA sequence analysis

Quality Assessment/Clustering

Quality assessment

We are going to begin our single-cell analysis by loading in the output from CellRanger. We will load in our different samples, create a Seurat object with them, and take a look at the quality of the cells.

Note, we have provided the raw data for this exercise in your cloud workspace. They are also available here.

The general steps to preprocessing your single-cell data with Seurat:

  1. Create a Seurat object
  2. Filter low-quality cells
  3. Merge samples
  4. Normalize counts
  5. Find variable features
  6. Scale data
  7. Determine PCs for Clustering
  8. Clustering -> FindNeighbors, FindClusters, RunUMAP

Load in Data

First, create a new R script and load the needed packages. The most important package for this step is Seurat. Seurat provides a unique data structure and tools for quality control, analysis, and exploration of single-cell RNA sequencing data. Seurat is very popular and is considered a standard tool for single-cell RNA analysis. Another example of a toolkit with similar functionality is Scanpy, which is implemented in Python.

library("Seurat") 
library("ggplot2") # creating plots
library("cowplot") # add-on to ggplot, we use the plot_grid function to put multiple violin plots in one image file
library("dplyr")   # a set of functions for data frame manipulation -- a core package of Tidyverse (THE R data manipulation package)
library("Matrix")  # a set of functions for operating on matrices
library("viridis") # color maps for graphs that are more readable than default colors
library("gprofiler2") 

Let’s make sure our workspace is configured the way we like. We will set our working directory where files will be saved. It is always good practice to verify what directory you are in and to ensure that you set the directory to where you want your files to be saved.

getwd()                         # list your current working directory
dir()                           # list the contents of your current working directory
setwd("/cloud/project/")        # set your working directory to where your data files are located
getwd()                         # verify that your working directory has been set correctly 

outdir="/cloud/project/outdir_single_cell_rna" # a path to where you want your plots to be saved
dir.create(outdir)

We already have our single-cell data files loaded into the posit session. Let’s take a look at them:

list.files("data/single_cell_rna/cellranger_outputs") # use this command or navigate to the file in the finder pane

You should see six h5 files:

[1] "Rep1_ICB-sample_filtered_feature_bc_matrix.h5"   "Rep1_ICBdT-sample_filtered_feature_bc_matrix.h5"
[3] "Rep3_ICB-sample_filtered_feature_bc_matrix.h5"   "Rep3_ICBdT-sample_filtered_feature_bc_matrix.h5"
[5] "Rep5_ICB-sample_filtered_feature_bc_matrix.h5"   "Rep5_ICBdT-sample_filtered_feature_bc_matrix.h5"

Loading in one sample

Next, we will read in our data matrix generated by Cellranger. Cellranger provides several output files, but the one we need is sample_filtered_feature_bc_matrix.h5, which is a matrix of the raw read counts for all cells per sample. This file is not human-readable but can be mentally visualized as a table where the cell barcodes are the column headers and the genes (or features) are the row names.

We will be using 3 replicates, which are separated into pairs of ICB-treated and CD4 depleted ICB-treated samples.

The CreateSeuratObject function is given 4 parameters. The first is the counts, which we supply with the data read in from the h5 files. The project parameter is basically what you want this Seurat object’s identification to be, so we pass it the sample name. We also apply a little bit of filtering in this step by saying that we only want to create an object where each gene is found in a minimum of 10 cells and each cell has a minimum of 100 features (genes).

For example, two samples could be processed like this:

Rep1_ICBdT_data = Read10X_h5("data/single_cell_rna/cellranger_outputs/Rep1_ICBdT-sample_filtered_feature_bc_matrix.h5") # read in the cellranger h5 file

Rep1_ICB_data = Read10X_h5("data/single_cell_rna/cellranger_outputs/Rep1_ICB-sample_filtered_feature_bc_matrix.h5")

Rep1_ICBdT_data_seurat_obj = CreateSeuratObject(counts = Rep1_ICBdT_data, project = "Rep1_ICBdT", min.cells = 10, min.features = 100)

Rep1_ICB_data_seurat_obj = CreateSeuratObject(counts = Rep1_ICB_data, project = "Rep1_ICB", min.cells = 10, min.features = 100)

Understanding our Seurat object

Let’s get a feel for what this Seurat object looks like and how we can access the information we will use for quality assessment, clustering, and further analysis.

print(Rep1_ICB_data_seurat_obj)
An object of class Seurat 
14423 features across 4169 samples within 1 assay 
Active assay: RNA (14423 features, 0 variable features)
 1 layer present: counts

We see we have 14423 features which are our genes, 4269 samples which are our cells, and 1 assay which is our counts matrix.

Assays(Rep1_ICB_data_seurat_obj) # seurat objects are made up of assays

Layers(Rep1_ICB_data_seurat_obj) # assays contain layers

The raw counts are stored in an assay called RNA.

Rep1_ICB_data_seurat_obj[["RNA"]] # an assay with layers
Assay (v5) data with 14423 features for 4169 cells
First 10 features:
 Mrpl15, Lypla1, Tcea1, Atp6v1h, Rb1cc1, 4732440D04Rik, Pcmtd1, Gm26901, Sntg1, Rrs1 
Layers:
 counts 

Let’s view our counts.

Rep1_ICB_data_seurat_obj[["RNA"]]$counts
14423 x 4169 sparse Matrix of class "dgCMatrix"
  [[ suppressing 59 column names ‘AAACCTGAGCGGATCA-1’, ‘AAACCTGCAAGAGGCT-1’, ‘AAACCTGCATACTCTT-1’ ... ]]
  [[ suppressing 59 column names ‘AAACCTGAGCGGATCA-1’, ‘AAACCTGCAAGAGGCT-1’, ‘AAACCTGCATACTCTT-1’ ... ]]
                                                                                                                                           
Mrpl15        . . . 1 . . . . . . . 1 1 . 2 . . . . . . . . 1 1 1 . . .  5 . . . 1 1 . . . 1 1 . . . . . . . . . . . . . . . 1 . . . ......
Lypla1        . . . 1 . . . . . . . . . . 1 1 . . . . . . . 2 1 . . 1 .  6 . . 1 . . . . . . 1 . . 1 . . . . . . . . . . . 1 . . . . ......
Tcea1         . . . . . . 2 . . . 1 . 1 . 1 . 2 1 1 . . . . . . . . . 1 20 . . 2 . 2 . 1 . 3 . . 1 1 . . . . 2 . 3 1 . 1 . . 3 . . 1 ......
Atp6v1h       . 1 . . . . . . . . . . 1 . . 1 . . . . . . . . 1 . 1 . .  2 . . . 2 1 . . . . . . 2 1 . 1 . . 1 2 . 1 . . . . . . . 1 ......
Rb1cc1        . . . . 2 . 1 . . . . . 3 . 1 1 . . . . . 1 . . 2 4 . . .  1 . . . 1 1 . . . 1 1 . . . . . 2 . 2 1 . 2 1 . . . . 2 1 . ......
4732440D04Rik . . . . . . . . . . . . . . . . . . . . . . . . . . . . .  1 . . . . . . . . 1 . . . . . . . . . . . 1 . . . . . . . . ......
Pcmtd1        . . . . . 1 . . . . . . . . . 1 1 . . . . 1 . . 1 . . 1 .  3 . . . . 1 . . . . . . . . 1 . 1 . . . . . . . . . . 1 1 . ......
Gm26901       . . . . . . . . . . . . . . . . . . . . . . . . . . . . .  . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ......

 ..............................
 ........suppressing 4110 columns and 14407 rows in show(); maybe adjust options(max.print=, width=)
 ..............................

The counts matrix is not very human readable and not at all human interpretable. Again, if we could see the whole matrix it would look like a table with the columns as cell barcodes and the row names as genes. The numbers are how many reads of that gene were observed in each cell.

We store our human-readable annotations in a place called meta.data. We can see that this is much more interpretable with row names as cell barcodes and columns as labels for each cell. As we analyze our data we will add a lot of columns to meta.data so get used to looking at it! Automatically meta.data is populated with three columns:

  1. orig.ident: the sample this cell came from
  2. nCount_RNA: the total number of reads in that cell
  3. nFeature_RNA: the number of genes in that cell
head(Rep1_ICB_data_seurat_obj@meta.data)
                   orig.ident nCount_RNA nFeature_RNA
AAACCTGAGCGGATCA-1   Rep1_ICB       5679         1758
AAACCTGCAAGAGGCT-1   Rep1_ICB       1397          496
AAACCTGCATACTCTT-1   Rep1_ICB       8463         2444
AAACCTGCATCATCCC-1   Rep1_ICB       4640         1721
AAACCTGGTTCGGGCT-1   Rep1_ICB       3810         1291
AAACCTGTCAGTCCCT-1   Rep1_ICB       7810         2079

Note that there are actually two ways to access the meta.data information:

# viewing meta.data
head(Rep1_ICB_data_seurat_obj@meta.data)

head(Rep1_ICB_data_seurat_obj[[]])

# viewing the orig.ident column in meta.data

head(Rep1_ICB_data_seurat_obj@meta.data$orig.ident)

head(Rep1_ICB_data_seurat_obj[['orig.ident']])

Seurat Object Diagram

We can view the cell’s identities by using the Idents function. Our cell’s identites are set to be the orig.ident column in the meta.data. When we use our table function to create a summary, we see that all cells are from the same sample, which is expected since we are just looking at one sample’s Seurat object right now.

Idents(Rep1_ICB_data_seurat_obj) # View cell identities

table(Idents(Rep1_ICB_data_seurat_obj)) # get summary table

It is very easy to add a column to meta.data. Let’s start by adding the percentage of mitochondrial genes, this will be a useful number for when we begin removing low-quality cells. We will call our column percent.mt and will use the function PercentageFeatureSet to calculate the percentage of all the counts belonging to a subset of the possible features for each cell. We will use the pattern parameter to match all genes that start with mt (the carrot ^ character means “starts with”) and specify that we want this calculation to be done on our RNA assay.

Rep1_ICB_data_seurat_obj[["percent.mt"]] = 
    PercentageFeatureSet(Rep1_ICB_data_seurat_obj, pattern = "^mt-", assay = "RNA")

head(Rep1_ICB_data_seurat_obj[[]]) # check to see the column in meta.data

We will use percent.mt to assess the quality if our cells, lets take a quick look at this one sample data before moving on to process all our samples. Using violin plots we take a look at the number of reads, the number of genes, and the percentage of mitochondrial genes in each cell.

p1 <- VlnPlot(Rep1_ICB_data_seurat_obj, features = c("nCount_RNA"), pt.size = 0) 
p2 <- VlnPlot(Rep1_ICB_data_seurat_obj, features = c("nFeature_RNA"), pt.size = 0) + scale_y_continuous(breaks = c(0, 300, 500, 1000, 2000, 4000))
p3 <- VlnPlot(Rep1_ICB_data_seurat_obj, features = c("percent.mt"), pt.size = 0) + scale_y_continuous(breaks = c(0, 12.5, 25, 50))
p <- plot_grid(p1, p2, p3, ncol = 3)
p

We see that a majority of cells have greater than 1000 genes and a mitochondrial percentage of less 12. Cells that fall outside of this range are most likely dead or of low quality, we will want to filter these out.

rep1_icb prefiltered violen plot

Creating Seurat Objects for all Samples

The above code accomplishes what we want to do just fine but it requires a lot of paths and sample names to be exactly correct for it to work. There is a lot of room for small mistakes to be made and it would take a long time to write out all the commands.

Can you write a for-loop to read in all six sample files?

Our for-loop will accomplish two major steps. The first is to read in the cellranger files and the second is to create a Seurat object. So this for-loop can be read: “For every sample in the sample_names list, paste together a path where the h5 file for that sample can be found, print that path for verification, use the function Read10X_h5 to read the h5 file, create a Seurat object using the function CreateSeuratObject, and finally add that sample’s Seurat object to a list of Seurat objects. When the for-loop completes, our sample.data list should contain all 6 samples.

Here is how I will write the for loop:

# List of sample names
sample_names <- c("Rep1_ICBdT", "Rep1_ICB", "Rep3_ICBdT", "Rep3_ICB",
                  "Rep5_ICBdT", "Rep5_ICB")

sample.data  = list() # create a list to hold seurat object

for (sample in sample_names) {
  path = paste("data/single_cell_rna/cellranger_outputs/", sample, "-sample_filtered_feature_bc_matrix.h5", sep="") # paste together a path name for the h5 file
  print(path) # check to make sure the path is correct

  data = Read10X_h5(path) # read in the cellranger h5 file

  seurat_obj = CreateSeuratObject(counts = data, project = sample, min.cells = 10, min.features = 100)

  sample.data[[sample]] = seurat_obj # add sample seurat object to list
}

Filtering Out Low-Quality Cells

Calculate the Percent of Mitochondrial Genes within each cell

As we did for a single sample, we want to calculate the percentage of mitochondrial genes for each cell. We will again use a for-loop to accomplish this for all samples:

for (sample in sample_names) {
  sample.data[[sample]][["percent.mt"]] = 
    PercentageFeatureSet(sample.data[[sample]], pattern = "^mt-", assay = "RNA")
}

View the Number of Cells Before Filtering

for (sample in sample_names) {
  print(table(Idents(sample.data[[sample]]))) 
}
Rep1_ICBdT 
      4012 

Rep1_ICB 
    4169 

Rep3_ICBdT 
      5656 

Rep3_ICB 
    6474 

Rep5_ICBdT 
      6066 

Rep5_ICB 
    2993

Quality Assessment Violin Plots

We can produce violin plots to view the quality of the cells for each sample. Take time to examine the outputted plots and think about what threshold would be best to remove the low-quality cells.

for (sample in sample_names) {
  print(sample)
  jpeg(sprintf("%s/%s_unfilteredQC.jpg", outdir, sample), width = 16, height = 5, units = 'in', res = 150)
  p1 <- VlnPlot(sample.data[[sample]], features = c("nCount_RNA"), pt.size = 0) 
  p2 <- VlnPlot(sample.data[[sample]], features = c("nFeature_RNA"), pt.size = 0) + scale_y_continuous(breaks = c(0, 300, 500, 1000, 2000, 4000))
  p3 <- VlnPlot(sample.data[[sample]], features = c("percent.mt"), pt.size = 0) + scale_y_continuous(breaks = c(0, 12.5, 25, 50))
  p <- plot_grid(p1, p2, p3, ncol = 3)
  
  print(p)
  dev.off()
}

Remove low-quality cells based on the QC plots

Let’s say we decide to keep all cells with gene counts greater than 1000 and mitochondrial percentages less than 12.

We will mark cells that we want to keep. This ifelse function statement reads: if percent.mt is less than or equal to 12 we will mark it as TRUE to keep it, otherwise we will mark it as FALSE to filter it out.

Rep1_ICB_data_seurat_obj[["keep_cell_percent.mt"]] = ifelse(Rep1_ICB_data_seurat_obj[["percent.mt"]] <= 12, TRUE, FALSE)

head(Rep1_ICB_data_seurat_obj[[]])
                   orig.ident nCount_RNA nFeature_RNA percent.mt kept_cell_percent.mt
AAACCTGAGCGGATCA-1   Rep1_ICB       5679         1758   2.341962                 TRUE
AAACCTGCAAGAGGCT-1   Rep1_ICB       1397          496  50.894775                FALSE
AAACCTGCATACTCTT-1   Rep1_ICB       8463         2444   2.197802                 TRUE
AAACCTGCATCATCCC-1   Rep1_ICB       4640         1721   3.750000                 TRUE
AAACCTGGTTCGGGCT-1   Rep1_ICB       3810         1291   2.755906                 TRUE
AAACCTGTCAGTCCCT-1   Rep1_ICB       7810         2079   3.354673                 TRUE

We will do the same for the number of genes.

Rep1_ICB_data_seurat_obj[["keep_cell_nFeature"]] = ifelse(Rep1_ICB_data_seurat_obj[["nFeature_RNA"]] > 1000, TRUE, FALSE)

head(Rep1_ICB_data_seurat_obj[[]])
                   orig.ident nCount_RNA nFeature_RNA percent.mt kept_cell_percent.mt keep_cell_nFeature
AAACCTGAGCGGATCA-1   Rep1_ICB       5679         1758   2.341962                 TRUE               TRUE
AAACCTGCAAGAGGCT-1   Rep1_ICB       1397          496  50.894775                FALSE              FALSE
AAACCTGCATACTCTT-1   Rep1_ICB       8463         2444   2.197802                 TRUE               TRUE
AAACCTGCATCATCCC-1   Rep1_ICB       4640         1721   3.750000                 TRUE               TRUE
AAACCTGGTTCGGGCT-1   Rep1_ICB       3810         1291   2.755906                 TRUE               TRUE
AAACCTGTCAGTCCCT-1   Rep1_ICB       7810         2079   3.354673                 TRUE               TRUE

Create this column for all samples:

for (sample in sample_names) {
  sample.data[[sample]][["keep_cell_percent.mt"]] = ifelse(sample.data[[sample]][["percent.mt"]] <= 12, TRUE, FALSE)
  sample.data[[sample]][["keep_cell_nFeature"]] = ifelse(sample.data[[sample]][["nFeature_RNA"]] > 1000, TRUE, FALSE)

}

Let’s see how many cells we will have after filtering:

for (sample in sample_names) {
  print(sample)
  print(sum(sample.data[[sample]][["keep_cell_nFeature"]] & sample.data[[sample]][["keep_cell_percent.mt"]]))
}
[1] "Rep1_ICBdT"
[1] 3106
[1] "Rep1_ICB"
[1] 2986
[1] "Rep3_ICBdT"
[1] 5072
[1] "Rep3_ICB"
[1] 5680
[1] "Rep5_ICBdT"
[1] 4547
[1] "Rep5_ICB"
[1] 1794

Post-filtration number of cells

Let’s use the subset function to view the spread of our data after filtering.

for (sample in sample_names) {
  print(sample)
  jpeg(sprintf("%s/%s_filteredQC.jpg", outdir, sample), width = 16, height = 5, units = 'in', res = 150)
  p1 <- VlnPlot(subset(sample.data[[sample]], nFeature_RNA > 1000 & percent.mt <= 12), features = c("nCount_RNA"), pt.size = 0) 
  p2 <- VlnPlot(subset(sample.data[[sample]], nFeature_RNA > 1000 & percent.mt <= 12), features = c("nFeature_RNA"), pt.size = 0) + scale_y_continuous(breaks = c(0, 300, 500, 1000, 2000, 4000))
  p3 <- VlnPlot(subset(sample.data[[sample]], nFeature_RNA > 1000 & percent.mt <= 12), features = c("percent.mt"), pt.size = 0) + scale_y_continuous(breaks = c(0, 12.5, 25, 50))
  p <- plot_grid(p1, p2, p3, ncol = 3)
  
  print(p)
  dev.off()
}

Merge the Samples

Now that we have assessed the quality on a per-sample level we can merge all our samples together to create one seurat object. The merge function will accomplish this for us and add the sample name to the end of the cell barcodes in the case that there are duplicate barcodes across samples.


sample_names <- c("Rep1_ICBdT", "Rep1_ICB", "Rep3_ICBdT", "Rep3_ICB",
                  "Rep5_ICBdT", "Rep5_ICB")

merged <- merge(x = sample.data[["Rep1_ICBdT"]], y = c(sample.data[["Rep1_ICB"]], sample.data[["Rep3_ICBdT"]], sample.data[["Rep3_ICB"]], sample.data[["Rep5_ICBdT"]], sample.data[["Rep5_ICB"]]), add.cell.ids = sample_names)  

Filtering our Data

Let’s now filter out merged objects with the cutoffs decided above.

unfiltered_merged <- merged # save a copy of unfiltered merge for later
merged <- subset(merged, nFeature_RNA > 1000 & percent.mt <= 12) 
merged
An object of class Seurat 
18187 features across 23185 samples within 1 assay 
Active assay: RNA (18187 features, 0 variable features)
 6 layers present: counts.Rep1_ICBdT, counts.Rep1_ICB, counts.Rep3_ICBdT, counts.Rep3_ICB, counts.Rep5_ICBdT, counts.Rep5_ICB

As of now, the various replicates are in their own layers. They need to be merged into 1 single layer for further analysis.

merged <- JoinLayers(merged)
merged
An object of class Seurat 
18187 features across 23185 samples within 1 assay 
Active assay: RNA (18187 features, 0 variable features)
 1 layer present: counts

We can use Idents to verify that we have all our cells in our merged object.

Idents(merged) # View cell identities

table(Idents(merged)) # get summary table

Normalize Data

Standard practice for scRNA data is to normalize your counts. Many functions will only use the normalized counts and not look at the raw counts. The NormalizeData function takes our merged object and will log normalize our RNA assay. Log normalized in this context means that the feature counts for each cell are divided by the total counts for that cell and multiplied by the scale.factor, then natural-log transformed using log1p.

merged <- NormalizeData(merged, assay = "RNA", normalization.method = "LogNormalize", scale.factor = 10000)

merged
An object of class Seurat 
18187 features across 23185 samples within 1 assay 
Active assay: RNA (18187 features, 0 variable features)
 2 layers present: data, counts

Seurat Object Normalize

Find Variable Features

Next, we identify genes that are highly expressed in some cells, and lowly expressed in others. features that are outliers on a ‘mean variability plot’. We will use vst to choose the top variable features. These genes will be used in downstream analysis, like PCA.

merged <- FindVariableFeatures(merged, assay = "RNA", selection.method = "vst", nfeatures = 2000, mean.cutoff = c(0.1, 8), dispersion.cutoff = c(1,Inf))

We can visualize the highly variable genes with a dot plot.

# Identify the 10 most highly variable genes
top10 <- head(VariableFeatures(merged), 10)
top10

# plot variable features with and without labels
plot1 <- VariableFeaturePlot(object=merged)
HoverLocator(plot = plot1)

Understanding where the information about the variable features are stored is a little tricky.

merged[["RNA"]]
Assay (v5) data with 18187 features for 23185 cells
Top 10 variable features:
 Jchain, Ighg1, Igkc, Sprr2d, Mzb1, Slpi, Sprr2f, Sprr2h, Saa3, Cxcl3 
Layers:
 data, counts 

Seurat FindVariableFeatures

Scale Data

Now we will scale and center the genes in the dataset.

merged <- ScaleData(merged, verbose = TRUE) 

merged
An object of class Seurat 
18187 features across 23185 samples within 1 assay 
Active assay: RNA (18187 features, 2000 variable features)
 3 layers present: data, counts, scale.data

Seurat FindVariableFeatures

Cell Cycle Scoring

Cell Cycle scoring is another step that can add extremely useful information to your Seurat object. We will use gprofiler2 to grab a list of mouse genes which are associated with different phases of the the cell cycle. The Seurat function ‘CellCycleScoring’ will then calculate a score for each cell based on the given genes and also assign the cell a phase. This function will add the columns S.Score, G2M.Score, and Phase to the meta.data.

library(gprofiler2) 
s.genes = gorth(cc.genes.updated.2019$s.genes, source_organism = "hsapiens", target_organism = "mmusculus")$ortholog_name
g2m.genes = gorth(cc.genes.updated.2019$g2m.genes, source_organism = "hsapiens", target_organism = "mmusculus")$ortholog_name

merged <- CellCycleScoring(object=merged, s.features=s.genes, g2m.features=g2m.genes, set.ident=FALSE)

head(merged[[]])
                              orig.ident nCount_RNA nFeature_RNA percent.mt keep_cell_percent.mt keep_cell_nFeature     S.Score   G2M.Score Phase
Rep1_ICBdT_AAACCTGAGCCAACAG-1 Rep1_ICBdT      20585         4384   1.675978                 TRUE               TRUE  0.61857647  0.19639961     S
Rep1_ICBdT_AAACCTGAGCCTTGAT-1 Rep1_ICBdT       4528         1967   3.577739                 TRUE               TRUE -0.06225445 -0.13230705    G1
Rep1_ICBdT_AAACCTGAGTACCGGA-1 Rep1_ICBdT      12732         3327   2.764687                 TRUE               TRUE -0.14015383 -0.17780932    G1
Rep1_ICBdT_AAACCTGCACGGCCAT-1 Rep1_ICBdT       4903         2074   1.427697                 TRUE               TRUE -0.05258375 -0.06400598    G1
Rep1_ICBdT_AAACCTGCACGGTAAG-1 Rep1_ICBdT      10841         3183   2.472097                 TRUE               TRUE -0.10176717 -0.06637093    G1
Rep1_ICBdT_AAACCTGCATGCCACG-1 Rep1_ICBdT      10981         2788   2.030780                 TRUE               TRUE -0.08794336 -0.21230015    G1

Determine how many PCA should be used for clustering

Looking at the count matrix for our Seurat object is a good reminder that there is no way we can process the number of genes and cells present with just our eyes. We need a way of compressing this information into something we can more easy comprehend and manipulate. PCAs are a dimension reduction strategy that aims to show similarity without losing the patterns that drive variation. We can visualize this by picturing what it would look like if we had just two cells and a handful of genes. If one cell’s gene expression value was on the x-axis and the other on the y-axis you would get a simple dot plot and could draw two lines through those points to measure the spread of the data points in two directions. Those lines that you draw are a PC, they generalize the data points into a more manageable, single object.

If we added another cell, we would add another axis to our graph AND we would add another direction in which we could have variation. So for the 23185 cells in our dataset, we would have 23185 directions of variation or principal components (PC). The PC that deals with the largest variant is PC1. For further explanation and visuals please view this page.

Calculating PCAs

Now we will calculate PCs on our dataset. We will calculate the default 50 PCs which should capture plenty of information on this dataset.

merged <- RunPCA(merged, npcs = 50, assay = "RNA") 

merged
An object of class Seurat 
18187 features across 23185 samples within 1 assay 
Active assay: RNA (18187 features, 2000 variable features)
 3 layers present: data, counts, scale.data
 1 dimensional reduction calculated: pca
merged[["pca"]]
A dimensional reduction object with key PC_ 
 Number of dimensions: 50 
 Number of cells: 23185 
 Projected dimensional reduction calculated:  FALSE 
 Jackstraw run: FALSE 
 Computed using assay: RNA 

After the PCs have been calculated, then each cell is “fitted” back into that PCs context. So for each cell we ask, “whats a cell’s score/embedding for a given PC based on gene expression in that cell”?

head(Embeddings(merged, reduction = "pca")[, 1:5])
                                    PC_1        PC_2        PC_3      PC_4       PC_5
Rep1_ICBdT_AAACCTGAGCCAACAG-1   3.835173  -0.8115711   9.8921410 2.2594305 14.1203451
Rep1_ICBdT_AAACCTGAGCCTTGAT-1   3.663907  -2.2085850  -1.2447077 1.2005316 -1.6541640
Rep1_ICBdT_AAACCTGAGTACCGGA-1 -65.634614 -46.0958039  -9.3464346 7.9993856  3.6250345
Rep1_ICBdT_AAACCTGCACGGCCAT-1   4.139736  -0.8617120  -0.3006448 0.4993232 -0.6362336
Rep1_ICBdT_AAACCTGCACGGTAAG-1   3.905916  -0.3761172   1.3514115 1.0349043  3.2202961
Rep1_ICBdT_AAACCTGCATGCCACG-1 -67.302103 -45.6771707 -11.5863126 3.4400124  4.9678837

Seurat FindVariableFeatures

Further resources here.

Visualizing PCAs

Now we have to decide what are the most important PCs, which capture the most similarity and diversity without introducing too much noise. There are several methods for analyzing your PCs.

Elbow Plot

One of the most common ways to understand PCs is an elbow plot. We choose the PC that is the ‘elbow’, that is where the standard deviation stops dramatically decreasing and levels out.

elbow <- ElbowPlot(merged, ndims = 30)
jpeg(sprintf("%s/Elbow.jpg", outdir), width = 8, height = 6, units = 'in', res = 150)
print(elbow)
dev.off()

Seurat FindVariableFeatures

Dimension Heat maps

We can also view the PCs as heatmaps. We see the top genes that drive the PC and how much they are expressed or not expressed in each cell. We want our heatmaps to show contrast and not just look like a blobby mess. More importantly, we look at what genes are driving the PC, and depending on what we would like to search for in our analysis, we would want to make sure our PCs include genes that are relavent.

jpeg(sprintf("%s/DimHm1_12.jpg", outdir), width = 10, height = 20, units = 'in', res = 150)
DimHeatmap(merged, dims = 1:12, balanced = TRUE, cells = 500)
dev.off()

jpeg(sprintf("%s/DimHm13_24.jpg", outdir), width = 10, height = 20, units = 'in', res = 150)
DimHeatmap(merged, dims = 13:24, balanced = TRUE, cells = 500)
dev.off()

jpeg(sprintf("%s/DimHm25_36.jpg", outdir), width = 10, height = 20, units = 'in', res = 150)
DimHeatmap(merged, dims = 25:36, balanced = TRUE, cells = 500)
dev.off()
DimHm1_12 DimHm13_24 DimHm25_36
Jackstaw

Jackstaw is an older and computationally expensive way to analyze the variability of PCs. However, it could be useful for further understanding what PCs to use. Usually, p-values over -100 are indicative of useful PCs.

# merged <- JackStraw(merged, num.replicate = 100, dims = 30)
# merged <- ScoreJackStraw(merged, dims = 1:30)

# plot <- JackStrawPlot(merged, dims = 1:30)
# jpeg(sprintf("JackStraw.jpg",), width = 8, height = 6, units = 'in', res = 150)
# print(plot)
# dev.off()

Choosing the number of PCAs

Using the elbow, heatmap, and possibly jackstraw we can make an informed decision on how many PCs we should use for cell clustering. We are trying to include as much information as possible without suffering from too much noise.

There is one tactic we can use to deduce our PC cutoff. We can see how many PCs have a standard deviation greater than 2.

ndims = length(which(merged@reductions$pca@stdev > 2)) # determines which PCs are important (stdev>2) 
ndims

After looking at the elbow plot, the PC heatmaps, and JackStraw plot (maybe this takes a long time to run so we might want to get rid of it) we decided to select 26 PCs. This is a great place to play around and take your time to notice the differences that adding or removing PCs make. When in doubt, slightly more PCs is better than not enough.

Cell Clustering

We are finally on the Cell Clustering step. The first step to clustering is the FindNeighbors which computes the k.param nearest neighbors for a given dataset. The concept is that given a data point, you want to identify the closest data points to it based on some similarity metric, such as Euclidean distance or cosine similarity. This helps to identify similar points in the dataset, which can be useful for making predictions or understanding the distribution of the data.

PC = 22

merged <- FindNeighbors(merged, dims = 1:PC)

We then run FindClusters where our cells will be grouped together based on similarity. FindNeighbors focuses on finding the nearest neighbors of a single data point, while FindClusters deals with grouping multiple data points into clusters based on their similarities. More extensive explanations are found here,

merged <- FindClusters(merged, resolution = 1.2, cluster.name = 'seurat_clusters_res1.2')

Finally, we get to RunUMAP, which uses the clustering information to project our cells into a 2D space for visualization.

merged <- RunUMAP(merged, dims = 1:PC)

merged
An object of class Seurat 
18187 features across 23185 samples within 1 assay 
Active assay: RNA (18187 features, 2000 variable features)
 3 layers present: data, counts, scale.data
 2 dimensional reductions calculated: pca, umap

Seurat FindVariableFeatures

Plotting

Let’s view our clusters with DimPlot.

jpeg(sprintf("%s/UMAP.jpg",outdir), width = 5, height = 4, units = 'in', res = 150)
DimPlot(merged, label = TRUE, group.by = 'seurat_clusters_res1.2')
dev.off()

Now, let’s color by our samples to make sure that no sample is clustering by itself – this would be an indication of batch effects. We want similar cells to be clustered together because they have similar gene expression. We do NOT want our cells to cluster together because of technical reasons (e.g., different sequencing batch, different replicate, etc).

# UMAP by sample
jpeg(sprintf("%s/UMAP_orig.ident.jpg",outdir), width = 5, height = 4, units = 'in', res = 150)
DimPlot(merged, label = TRUE, group.by = "orig.ident")
dev.off()

It looks like our samples are mixed nicely! We can check further by highlighting the cells from one sample.

# UMAP with one day highlighted (not saved)
highlighted_cells <- WhichCells(merged, expression = orig.ident == "Rep1_ICBdT")
DimPlot(merged, reduction = 'umap', group.by = 'orig.ident', cells.highlight = highlighted_cells)

Lets take a look at the cell cycle scoring calculations.

FeaturePlot(merged, features = c("S.Score", "G2M.Score")) + DimPlot(merged,  group.by = "Phase")  

Exploring Clustering Resolution

Now it’s time to explore the nuances of clustering resolution. Choosing cluster resolution is somewhat arbitrary and affects the number of clusters called (higher resolution calls more clusters). The shape of the UMAP does not change if you change the cluster resolution. The shape of the UMAP is determined by the number of PCs used to create the UMAP.

merged <- FindClusters(merged, resolution = 0.8, cluster.name = 'seurat_clusters_res0.8')

merged <- FindClusters(merged, resolution = 0.5, cluster.name = 'seurat_clusters_res0.5')

jpeg(sprintf("%s/UMAP_compare_res.jpg",outdir), width = 20, height = 5, units = 'in', res = 150)
DimPlot(merged, label = TRUE, group.by = 'seurat_clusters_res0.5') +
  DimPlot(merged, label = TRUE, group.by = 'seurat_clusters_res0.8') + 
  DimPlot(merged, label = TRUE, group.by = 'seurat_clusters_res1.2') 
dev.off()

Finishing up

Finally, let’s save our object for further analysis.

saveRDS(merged, file = "outdir_single_cell_rna/rep135_clustered.rds")

Exercise

Early we made a copy of our object before filtering. This object has two meta.data columns marking cells that are going to be removed.

Can we visualize these cells on a UMAP?

The unfiltered_merge object did not go through any processing after the merging step, so those will have to be repeated to produce a UMAP. Pay attention to what changes when you run the steps on the unfiltered object. Is the number of PCs different? Visualize the cell cycle scores, are there any patterns that you notice? Are the cells that are filtered out clustered together or scattered throughout the UMAP?

unfiltered_merged
An object of class Seurat 
18187 features across 23185 samples within 1 assay 
Active assay: RNA (18187 features, 0 variable features)
 1 layer present: counts

Resources

Seurat Command Cheat Sheet

Graphical Explanation of Seurat

scRNA-seq Data | Griffith Lab

RNA-seq Bioinformatics

Introduction to bioinformatics for RNA sequence analysis

scRNA-seq Data

Obtain RNA-seq test data.

The data that will be used to demonstrate scRNA analysis in this module has been published by Freshour et al. 2023. If you use this data in your own benchmarking, tool development, or research please cite this article. The complete raw sequence dataset can be obtained from SRA at accession: PRJNA934380.


MCBC6-overview


Model system

The full details can be found in the publication but briefly these data are generated from a murine bladder cancer model system with the following characteristics:

Experimental details

Briefly, the experimental details are depicted below and summarized briefly here.


MCBC6-experiment


Data generation

The method and types of data generation are depicted below and summarized briefly here.


MCBC6-data-generation


Description of samples/replicates and QC reports

In the following demonstration analyses we will focus on a simplified comparison of two biological conditions: treatment with checkpoint (“ICB”) or treatment with ICB after depletion of CD4 cells in the mice (“ICB-dT”). For each of these two conditions we will have three replicates (of five total reported in the publication).

List of replicates with labels and links to CellRanger 7.0.0 multi QC reports:

QC report discussion

Using the report for Rep1_ICB as an example, explore and discuss the following points:

Input files for the demonstration analysis

We will not be running Cell Ranger ourselves and will instead be starting the scRNA analysis in R using matrix files from Cell Ranger (cellrange multi to be precise) and a few other input files briefly described here:

Loupe browser demonstration for preliminary exploration of the MCB6C

Briefly explore the MCB6C data using the 10X Loupe browser. Use the following two samples as examples:

Input data files
Sample GEX Cloupe TCR Vloupe BCR Vloupe
REP3_ICB: Rep3_ICB GEX cloupe Rep3_ICB TCR vloupe Rep3_ICB BCR vloupe
REP3_ICBdT: Rep3_ICBdT GEX cloupe Rep3_ICBdT TCR vloupe Rep3_ICBdT BCR vloupe

.

Exercise

Work through the following very basic intro to this tool with the following steps: