Welcome to the blog

Posts

My thoughts and ideas

Gene set/Regulatory network annotation | Griffith Lab

RNA-seq Bioinformatics

Introduction to bioinformatics for RNA sequence analysis

Differential expression analysis | Griffith Lab

RNA-seq Bioinformatics

Introduction to bioinformatics for RNA sequence analysis

Differential expression analysis


In this section we will use the previously generated Seurat object that has gone through the various preprocessing steps, clustering, and celltyping, and use it for gene expression and differential expression analyses. We will carry out two differential expression analyses in this module. Firstly, since we know that the tumor cells should be epithelial cells, we will begin by trying to identify epithelial cells in our data using expression of Epcam as a marker. Subsequently, we will carry out a DE analysis within the Epcam-positive population(s). Secondly, we will compare the T cell populations of mice treated with ICB therapy against mice with their T cells depleted that underwent ICB therapy to determine differences in T cell phenotypes.


Read-in the saved seurat object from the previous step if it is not already loaded in your current R session.

TODO SEE WHAT THIS NEW OBJECT IS NAMED AND WHERE IT’S KEPT FROM PREVIOUS STEPS

library(Seurat)
library(dplyr)
library(EnhancedVolcano)
library(presto)
merged <- readRDS('processed_object_0409.rds')

Gene expression analysis for epithelial cells

TODO we may take care of the step below in a previous section

As of now the various replicates are in their own layers. They need to be merged into 1 single layer for further analysis. Also add (draft) SingleR labels to the object.

library(celldex)
library(SingleR)

merged
merged <- JoinLayers(merged)
merged

#load singler immgen reference
ref_immgen <- celldex::ImmGenData()
#generate predictions for our seurat object
predictions_main = SingleR(test = GetAssayData(merged), 
                      ref = ref_immgen,
                      labels = ref_immgen$label.main)

predictions_fine = SingleR(test = GetAssayData(merged), 
                           ref = ref_immgen,
                           labels = ref_immgen$label.fine)

#add main labels to object
merged[['immgen_singler_main']] = rep('NA', ncol(merged))
merged$immgen_singler_main[rownames(predictions_main)] = predictions_main$labels

#add fine labels to object
merged[['immgen_singler_fine']] = rep('NA', ncol(merged))
merged$immgen_singler_fine[rownames(predictions_fine)] = predictions_fine$labels


Compare the number of layers present before and after merging. JoinLayers is an important step because many of the DE functions use the log-normalized data (held in the data layer) instead of the data kept in the scale.data layer.

Use Seurat’s FeaturePlot function to color each cell by its Epcam expression on a UMAP. FeaturePlot requires at least 2 arguments- the seurat object, and the ‘feature’ you want to plot (where a ‘feature’ can be a gene, PC scores, any of the metadata columns, etc.). To customize the FeaturePlot, please refer to Seurat’s documentation here

FeaturePlot(merged, features = 'Epcam')

While there are some Epcam positive cells scattered on the UMAP, there appear to be 2 distinct groups of cells at the top of the UMAP that we may be able to pull apart as potentially being malignant cells.

There are a few different ways to go about identifying what those clusters are. We can start by trying to use the DimPlot plotting function from before along with the FeaturePlot function. Separating plots by the + symbol allows us to plot multiple plots side-by-side.

TODO UPDATE GROUP.BY ARGUMENT BASED ON WHAT THE CELLTYPE COLUMN IS CALLED

DimPlot(merged, group.by = 'seurat_clusters_res0.8', label = TRUE) + 
FeaturePlot(merged, features = 'Epcam') + 
DimPlot(merged, group.by = 'immgen_singler_main', label = TRUE)

While the plots generated by the above commands make it pretty clear that the clusters of interest are clusters 10 and 12, sometimes it is trickier to determine which cluster we are interested in solely from the UMAP as the clusters may be overlapping. In this case, a violin plot VlnPlot may be more helpful. Similar to FeaturePlot, VlnPlot also takes the Seurat object and features as input. It also requires a group.by argument that determines the x-axis groupings of the cells.
To learn more about customizing a Violin plot, please refer to the Seurat documentation

VlnPlot(merged, group.by = 'seurat_clusters_res0.8', features = 'Epcam')

Thus we were able to confirm that clusters 10 and 12 have the highest expression of Epcam. However, it is interesting that they are split into 2 clusters. Let’s use differential expression analysis to determine how these clusters differ from each other.

Differential expression for epithelial cells

We can begin by restricting the Seurat object to the cells we are interested in. We will do so using Seurat’s subset function, that allows us to create an object that is filtered to any values of interest in the metadata column. However, we first need to set the default identity of the Seurat object to the metadata column we want to use for the ‘subset’, and we can do so using the SetIdent function. After subsetting the object, we can plot the original object and subsetted object side-by-side to ensure the subsetting happened as expected. We can also count the number of cells of each type to confirm the same.

#set ident to seurat clusters metadata column and subset object to Epcam positive clusters
merged <- SetIdent(merged, value = 'seurat_clusters_res0.8')
merged_epithelial <- subset(merged, idents = c('10', '12'))

#confirm that we have subset the object as expected visually using a UMAP
DimPlot(merged, group.by = 'seurat_clusters_res0.8', label = TRUE) + 
DimPlot(merged_epithelial, group.by = 'seurat_clusters_res0.8', label = TRUE)

#confirm that we have subset the object as expected by looking at the individual cell counts
table(merged$seurat_clusters_res0.8)
table(merged_epithelial$seurat_clusters_res0.8)

Now we will use Seurat’s FindMarkers function to carry out a differential expression analysis between both groups. FindMarkers also requires that we use SetIdent to change the default ‘Ident’ to the metadata column we want to use for our comparison. More information about FindMarkers is available here.

Note that here we use FindMarkers to compare clusters 10 and 12. The default syntax of FindMarkers requires that we provide each group of cells as ident.1 and ident.2. The output of FindMarkers is a table with each gene that is differentially expressed and its corresponding log2FC. The direction of the log2FC is of ident.1 with respect to ident.2. Therefore, genes upregulated in ident.1 have positive log2FC, while those downregulated in ident.1 have negative log2FC. Here, we also provide a min.pct=0.25 argument so that we only test genes that are expressed in 25% of cells in either of the ident.1 or ident.2 groups. This can help reduce false positives as the genes must be expressed in a greater proportion of the cells compared to the default value of 1%. We also specify the logfc.threshold=0.1 parameter, which ensures our results only include genes that have a fold change of less than -0.1 or more than 0.1. Increasing the min.pct and logfc.threshold parameters can also result in the function running faster as they reduce the number of genes being tested.

#carry out DE analysis between both groups
merged_epithelial <- SetIdent(merged_epithelial, value = "seurat_clusters_res0.8")
epithelial_de <- FindMarkers(merged_epithelial, ident.1 = "10", ident.2 = "12", min.pct=0.25, logfc.threshold=0.1) #how cluster 10 changes wrt cluster 12

On opening epithelial_de in your RStudio session, you’ll see that it is a dataframe with the genes as rownames, and the following columns- p_val, avg_log2FC, pct.1, pct.2, p_val_adj. The p-values are dependent on the test used while running FindMarkers, and the adjusted p-value is based on the bonferroni correction test. pct.1 and pct.2 are the percentages of cells where the gene is detected in the ident.1 and ident.2 groups respectively.

Next we can subset this dataframe to only include DE genes that have a significant p-value, and then further subset the ‘significant DE genes only’ dataframe to the top 20 genes with the highest absolute log2FC. Looking a the absolute log2FC allows us to capture both, the upregulated and downregulated genes.

#restrict differentially expressed genes to those with an adjusted p-value less than 0.001 
epithelial_de_sig <- epithelial_de[epithelial_de$p_val_adj < 0.001,]

#get the top 20 genes by fold change
epithelial_de_sig %>%
  top_n(n = 20, wt = abs(avg_log2FC)) -> epithelial_de_sig_top20

epithelial_de_sig_top20 is a dataframe that is restricted to the top20 most differentially expressed genes by log2FC.

There are a few different ways we can visualize the differentially expressed genes. We’ll start by with the Violin and Feature plots from before. We can also visualize DEs using a DotPlot that allows us to capture both the average expression of a gene and the % of cells expressing it. In addition to these in-built Seurat functions, we can also generate a volcano plot using the EnhancedVolcano package. For the volcano plot, we can use the unfiltered DE results as the function colors and labels genes based on the parameters (pCutoff, FCcutoff) we specify.

#get list of top 20 DE genes for ease
epithelial_de_sig_top20_genes <- rownames(epithelial_de_sig_top20)

#plot all 20 genes in violin plots
VlnPlot(merged_epithelial, features = epithelial_de_sig_top20_genes, group.by = 'seurat_clusters_res0.8', ncol = 5, pt.size = 0)

#plot all 20 genes in UMAP plots
FeaturePlot(merged_epithelial, features = epithelial_de_sig_top20_genes, ncol = 5)

#plot all 20 genes in a DotPlot
DotPlot(merged_epithelial, features = epithelial_de_sig_top20_genes, group.by = 'seurat_clusters_res0.8') + RotatedAxis()

#plot all differentially expressed genes in a volcano plot
EnhancedVolcano(epithelial_de,
  lab = rownames(epithelial_de),
  x = 'avg_log2FC',
  y = 'p_val_adj',
  title = 'Cluster10 wrt Cluster 12',
  pCutoff = 0.05,
  FCcutoff = 0.5,
  pointSize = 3.0,
  labSize = 5.0,
  colAlpha = 0.3)

To find out how we can figure out what these genes mean, stay tuned! The next module on pathway analysis will help shed some light on that. Let’s create a TSV file containing our DE results for use later on. We will need to rerun FindMarkers with slightly different parameters for this- we will change the logfc.threshold parameter to 0, as one of the pathway analysis tools requires all genes to be included in the analysis (more on that later).

#rerun FindMarkers
epithelial_de_gsea <- FindMarkers(merged_epithelial, ident.1 = "10", ident.2 = "12", min.pct=0.25, logfc.threshold=0)
#save this table as a TSV file
write.table(x = epithelial_de_gsea, file = 'epithelial_de_gsea.tsv', sep='\t')

Differential expression for T cells

For the T cell focused analysis, we will start by subsetting our merged object to only have T cells, by combining the various T cell annotations from celltyping section. We’ll start by seeing all the possible celltypes we have, and picking the ones that are related to T cells. Next, we will SetIdent to the celltype metadata column, and subset to the celltypes that correspond to T cells. Finally, we’ll doublecheck that the subsetting happened as we expected it to.

TODO UPDATE THESE BASED ON FINAL CELLTYPING

#check all the annotated celltypes
unique(merged$immgen_singler_main)

#pick the ones that are related to T cells
t_celltypes_names <- c('T cells', 'NKT', 'Tgd')
merged <- SetIdent(merged, value = 'immgen_singler_main')
merged_tcells <- subset(merged, idents = t_celltypes_names)

#confirm that we have subset the object as expected visually using a UMAP
DimPlot(merged, group.by = 'immgen_singler_main', label = TRUE) + 
DimPlot(merged_tcells, group.by = 'immgen_singler_main', label = TRUE)

#confirm that we have subset the object as expected by looking at the individual cell counts
table(merged$immgen_singler_main)
table(merged_tcells$immgen_singler_main)

Now we want to compare cells from mice treated with ICB vs mice with their T cells depleted treated with ICB (ICBdT). First, we need to distinguish the ICB and ICBdT cells from each other. Start by clicking on the object in RStudio and expand meta.data to get a snapshot of the columns and the what kind of data they hold.

Looks like orig.ident has information about the condition and replicates, but for the purposes of this DE analysis, we want a meta.data column that combines the replicates of each condition. So, we want to combine the replicates of each condition together into a single category.

#we'll start by checking the possible names each replicate has.
unique(merged_tcells$orig.ident)

#there are 6 possible values, 3 replicates for the ICB treatment condition, and 3 for the ICBdT condition
#so we can combine "Rep1_ICB", "Rep3_ICB", "Rep5_ICB" to ICB, and "Rep1_ICBdT", "Rep3_ICBdT", "Rep5_ICBdT" to ICBdT. 
#first initialize a metadata column for experimental_condition
merged@meta.data$experimental_condition <- NA
#Now we can take all cells that are in each replicate-condition, and assign them to the appropriate condition
merged@meta.data$experimental_condition[merged@meta.data$orig.ident %in% c("Rep1_ICB", "Rep3_ICB", "Rep5_ICB")] <- "ICB"
merged@meta.data$experimental_condition[merged@meta.data$orig.ident %in% c("Rep1_ICBdT", "Rep3_ICBdT", "Rep5_ICBdT")] <- "ICBdT"
#double check that the new column we generated makes sense (each replicate should correspond to its experimental condition)
table(merged@meta.data$orig.ident, merged@meta.data$experimental_condition)

With the experimental conditions now defined, we can compare the T cells from both groups. We’ll start by using FindMarkers using similar parameters to last time, and see how ICBdT changes with respect to ICB. Next, restrict the dataframe to significant genes only, and then look at the top 5 most upregulated and downregulated DE genes by log2FC.

#carry out DE analysis between both groups
merged_tcells <- SetIdent(merged_tcells, value = "experimental_condition")
tcells_de <- FindMarkers(merged_tcells, ident.1 = "ICBdT", ident.2 = "ICB", min.pct=0.25)

#restrict differentially expressed genes to those with an adjusted p-value less than 0.001 
epithelial_de_sig <- epithelial_de[epithelial_de$p_val_adj < 0.001,]

#find the top 5 most downregulated genes
tcells_de_sig %>%
  top_n(n = 5, wt = -avg_log2FC)
#find the top 5 most upregulated genes
tcells_de_sig %>%
  top_n(n = 5, wt = avg_log2FC)

The most downregulated gene in the ICBdT condition based on foldchange is Cd4. That makes sense, because the monoclonal antibody (GK1.5) used for T cell depletion specifically targets CD4 T cells! You can read more about the antibody here.

Interestingly, for the list of genes that are upregulated, we see Cd8b1 show up. It could be interesting to see if the CD8 T cells’ phenotype changes based on the treatment condition. So now, let’s subset the object to CD8 T cells only, find DE genes to see how ICBdT CD8 T cells change compared to ICB CD8 T cells, and visualize these similar to before.

#subset object to CD8 T cells
cd8t_celltypes_names <- c("T cells (T.8EFF.OT1.12HR.LISOVA)","T cells (T.8EFF.OT1.24HR.LISOVA)","T cells (T.8EFF.OT1.48HR.LISOVA)","T cells (T.8EFF.OT1.D10LIS)","T cells (T.8EFF.OT1.D15.LISOVA)","T cells (T.8EFF.OT1.D45VSV)","T cells (T.8EFF.OT1.D5.VSVOVA)","T cells (T.8EFF.OT1.D8.VSVOVA)","T cells (T.8EFF.OT1.D8LISO)","T cells (T.8EFF.OT1.LISOVA)","T cells (T.8EFF.OT1.VSVOVA)","T cells (T.8EFF.OT1LISO)","T cells (T.8EFF.TBET-.OT1LISOVA)","T cells (T.8EFF.TBET+.OT1LISOVA)","T cells (T.8EFFKLRG1+CD127-.D8.LISOVA)","T cells (T.8MEM.OT1.D100.LISOVA)","T cells (T.8MEM.OT1.D106.VSVOVA)","T cells (T.8MEM.OT1.D45.LISOVA)","T cells (T.8Mem)","T cells (T.8MEM)","T cells (T.8MEMKLRG1-CD127+.D8.LISOVA)","T cells (T.8NVE.OT1)","T cells (T.8Nve)","T cells (T.8NVE)","T cells (T.8SP24-)","T cells (T.8SP69+)","T cells (T.CD8.1H)","T cells (T.CD8.24H)","T cells (T.CD8.48H)","T cells (T.CD8.5H)","T cells (T.CD8.96H)","T cells (T.CD8.CTR)")
merged_tcells <- SetIdent(merged_tcells, value = 'immgen_singler_fine')
merged_cd8tcells <- subset(merged_tcells, idents = cd8t_celltypes_names)

#carry out DE analysis between both groups
merged_cd8tcells <- SetIdent(merged_cd8tcells, value = "experimental_condition")
cd8tcells_de <- FindMarkers(merged_cd8tcells, ident.1 = "ICBdT", ident.2 = "ICB", min.pct=0.25) #how ICBdT changes wrt ICB

#restrict differentially expressed genes to those with an adjusted p-value less than 0.001 
cd8tcells_de_sig <- cd8tcells_de[cd8tcells_de$p_val_adj < 0.001,]

#get the top 20 genes by fold change
cd8tcells_de_sig %>%
  top_n(n = 20, wt = abs(avg_log2FC)) -> cd8tcells_de_sig_top20

#get list of top 20 DE genes for ease
cd8tcells_de_sig_top20_genes <- rownames(cd8tcells_de_sig_top20)


#plot all 20 genes in violin plots
VlnPlot(merged_cd8tcells, features = cd8tcells_de_sig_top20_genes, group.by = 'experimental_condition', ncol = 5, pt.size = 0)

#plot all 20 genes in UMAP plots
FeaturePlot(merged_cd8tcells, features = cd8tcells_de_sig_top20_genes, ncol = 5)

#plot all 20 genes in a DotPlot
DotPlot(merged_cd8tcells, features = cd8tcells_de_sig_top20_genes, group.by = 'experimental_condition') + RotatedAxis()

#plot all differentially expressed genes in a volcano plot
EnhancedVolcano(cd8tcells_de,
  lab = rownames(cd8tcells_de),
  x = 'avg_log2FC',
  y = 'p_val_adj',
  title = 'ICBdT wrt ICB',
  pCutoff = 0.05,
  FCcutoff = 0.5,
  pointSize = 3.0,
  labSize = 5.0,
  colAlpha = 0.3)

At this point, you can either start doing literature searches for some of these genes and maybe find that antigen-specific memory CD8 T cells have higher expression of Bcl2 and Cdk8.

But we can also use gene set and pathway analyses to try and determine what processes the cells may be involved in.