Welcome to the blog

Posts

My thoughts and ideas

DE Visualization Advanced | Griffith Lab

RNA-seq Bioinformatics

Introduction to bioinformatics for RNA sequence analysis

DE Visualization Advanced


RNA-seq_Flowchart4


Supplementary R DE Visualization

Occasionally you may wish to reformat and work with expression estimates in R ad hoc. Therefore we provide an optional/advanced tutorial on how to visualize your results for R and perform “old school” (non-ballgown, non-DESeq2) visualization of your data.

In this tutorial you will:

  • Learn basic R usage and commands (common plots, and data manipulation tasks)
  • Examine the expression estimates
  • Create an MDS plot to visualize the differences between/among replicates, library prep methods and UHR versus HBR
  • Examine the differential expression estimates
  • Visualize the expression estimates and highlight those genes that appear to be differentially expressed
  • Generate a list of the top differentially expressed genes
  • Ask how reproducible technical replicates are.

Expression and differential expression files will be read into R. The R analysis will make use of the transcript-level expression and differential expression files from stringtie/ballgown.

Start RStudio, or launch a posit Cloud session, or if you are using AWS, navigate to the correct directory and then launch R:

cd $RNA_HOME/de/ballgown/ref_only/
R
#First you'll load your libraries and your data.
#Load libraries
library(ggplot2)
library(gplots)
library(GenomicRanges)
library(ggrepel)

#### Import the gene expression data from the HISAT2/StringTie/Ballgown tutorial

#Set working directory where results files exist
#datadir = "~/workspace/rnaseq/de/ballgown/ref_only"
datadir = "/cloud/project/data/bulk_rna"
outdir = "/cloud/project/outdir"

setwd(datadir)

# List the current contents of this directory
dir()

#Import expression results (TPM values) from the HISAT2/Stringtie pipeline
gene_expression=read.table("gene_tpm_all_samples.tsv", header=TRUE, stringsAsFactors=FALSE, row.names=1)
gene_names=read.table("ENSG_ID2Name.txt", header=TRUE, stringsAsFactors=FALSE)
colnames(gene_names)=c("gene_id","gene_name")

#Import DE results from the HISAT2/htseq-count/DESeq2 pipeline
setwd(outdir)
results_genes <-read.table("DE_all_genes_DESeq2.tsv", sep="\t", header=T, stringsAsFactors = F)

Next, you’ll load the data into a R dataframe.


#### Working with 'dataframes'
#View the first five rows of data (all columns) in one of the dataframes created
head(gene_expression)

#View the column names
colnames(gene_expression)

#View the row names
row.names(gene_expression)

#Determine the dimensions of the dataframe.  'dim()' will return the number of rows and columns
dim(gene_expression)

You’ll then get the first 3 rows of data and view expression and transcript information.

#Get the first 3 rows of data and a selection of columns
gene_expression[1:3,c(1:3,6)]

#Do the same thing, but using the column names instead of numbers
gene_expression[1:3, c("HBR_Rep1","HBR_Rep2","HBR_Rep3","UHR_Rep3")]

#Assign colors to each.  You can specify color by RGB, Hex code, or name
#To get a list of color names:
colours()
data_colors=c("tomato1","tomato2","tomato3","royalblue1","royalblue2","royalblue3")

The following code blocks are to generate various plots using the above data set.


#### Plot #1 - View the range of values and general distribution of TPM values for all 6 libraries
#Create boxplots for this purpose
#Display on a log2 scale and add the minimum non-zero value to avoid log2(0)

#Set the minimum non-zero TPM values for use later. Alternatively just set min value to 1
min_nonzero=1

# Set the columns for finding TPM and create shorter names for figures
data_columns=c(1:6)
short_names=c("HBR_1","HBR_2","HBR_3","UHR_1","UHR_2","UHR_3")

boxplot(log2(gene_expression[,data_columns]+min_nonzero), col=data_colors, names=short_names, las=2, ylab="log2(TPM)", main="Distribution of TPMs for all 6 libraries")
#Note that the bold horizontal line on each boxplot is the median

#### Plot #2 - plot a pair of replicates to assess reproducibility of technical replicates
#Tranform the data by converting to log2 scale after adding an arbitrary small value to avoid log2(0)
x = gene_expression[,"UHR_Rep1"]
y = gene_expression[,"UHR_Rep2"]
plot(x=log2(x+min_nonzero), y=log2(y+min_nonzero), pch=16, col="blue", cex=0.25, xlab="FPKM (UHR, Replicate 1)", ylab="FPKM (UHR, Replicate 2)", main="Comparison of expression values for a pair of replicates")

#Add a straight line of slope 1, and intercept 0
abline(a=0,b=1)

#Calculate the correlation coefficient and display in a legend
rs=cor(x,y)^2
legend("topleft", paste("R squared = ", round(rs, digits=3), sep=""), lwd=1, col="black")


#### Plot #3 - Scatter plots with a large number of data points can be misleading ... regenerate this figure as a density scatter plot
colors = colorRampPalette(c("white", "blue", "#007FFF", "cyan","#7FFF7F", "yellow", "#FF7F00", "red", "#7F0000"))
smoothScatter(x=log2(x+min_nonzero), y=log2(y+min_nonzero), xlab="TPM (UHR, Replicate 1)", ylab="TPM (UHR, Replicate 2)", main="Comparison of expression values for a pair of replicates", colramp=colors, nbin=200)


#### Plot #4 - Scatter plots of all sets of replicates on a single plot
#Create a function that generates an R plot.  This function will take as input the two libraries to be compared and a plot name
plotCor = function(lib1, lib2, name){
	x=gene_expression[,lib1]
	y=gene_expression[,lib2]
	zero_count = length(which(x==0)) + length(which(y==0))
	colors = colorRampPalette(c("white", "blue", "#007FFF", "cyan","#7FFF7F", "yellow", "#FF7F00", "red", "#7F0000"))
	smoothScatter(x=log2(x+min_nonzero), y=log2(y+min_nonzero), xlab=lib1, ylab=lib2, main=name, colramp=colors, nbin=275)
	abline(a=0,b=1)
	rs=cor(x,y, method="pearson")^2
	legend_text = c(paste("R squared = ", round(rs, digits=3), sep=""), paste("Zero count = ", zero_count, sep=""))
	legend("topleft", legend_text, lwd=c(1,NA), col="black", bg="white", cex=0.8)
}

#Now make a call to our custom function created above, once for each library comparison
par(mfrow=c(1,3))
plotCor("UHR_Rep1", "UHR_Rep2", "UHR_1 vs UHR_2")
plotCor("UHR_Rep2", "UHR_Rep3", "UHR_2 vs UHR_3")
plotCor("UHR_Rep1", "UHR_Rep3", "UHR_1 vs UHR_3")


#### Compare the correlation 'distance' between all replicates
#Do we see the expected pattern for all eight libraries (i.e. replicates most similar, then tumor vs. normal)?

#Calculate the TPM sum for all 6 libraries
gene_expression[,"sum"]=apply(gene_expression[,data_columns], 1, sum)

#Identify the genes with a grand sum TPM of at least 5 - we will filter out the genes with very low expression across the board
i = which(gene_expression[,"sum"] > 5)

#Calculate the correlation between all pairs of data
r=cor(gene_expression[i,data_columns], use="pairwise.complete.obs", method="pearson")

#Print out these correlation values
r

#### Plot #5 - Convert correlation to 'distance', and use 'multi-dimensional scaling' to display the relative differences between libraries
#This step calculates 2-dimensional coordinates to plot points for each library
#Libraries with similar expression patterns (highly correlated to each other) should group together
#What pattern do we expect to see, given the types of libraries we have (technical replicates, biologal replicates, tumor/normal)?
d=1-r
mds=cmdscale(d, k=2, eig=TRUE)
par(mfrow=c(1,1))
plot(mds$points, type="n", xlab="", ylab="", main="MDS distance plot (all non-zero genes)", xlim=c(-0.12,0.12), ylim=c(-0.12,0.12))
points(mds$points[,1], mds$points[,2], col="grey", cex=2, pch=16)
text(mds$points[,1], mds$points[,2], short_names, col=data_colors)

#### Plot #6 - View the distribution of differential expression values as a histogram
#Display only those results that are significant according to DESeq2 (loaded above)
sig=which(results_genes$pvalue<0.05)
hist(results_genes[sig,"log2FoldChange"], breaks=50, col="seagreen", xlab="log2(Fold change) UHR vs HBR", main="Distribution of differential expression values")
abline(v=-2, col="black", lwd=2, lty=2)
abline(v=2, col="black", lwd=2, lty=2)
legend("topleft", "Fold-change > 4", lwd=2, lty=2)

#### Plot #7 - Display the grand expression values from UHR and HBR and mark those that are significantly differentially expressed
gene_expression[,"HBR_mean"]=apply(gene_expression[,c(1:3)], 1, mean)
gene_expression[,"UHR_mean"]=apply(gene_expression[,c(4:6)], 1, mean)

x=log2(gene_expression[,"UHR_mean"]+min_nonzero)
y=log2(gene_expression[,"HBR_mean"]+min_nonzero)
plot(x=x, y=y, pch=16, cex=0.25, xlab="UHR TPM (log2)", ylab="HBR TPM (log2)", main="UHR vs HBR TPMs")
abline(a=0, b=1)
xsig=x[sig]
ysig=y[sig]
points(x=xsig, y=ysig, col="magenta", pch=16, cex=0.5)
legend("topleft", "Significant", col="magenta", pch=16)

#Get the gene symbols for the top N (according to corrected p-value) and display them on the plot
#topn = order(abs(results_genes[sig,"log2FoldChange"]), decreasing=TRUE)[1:25]
topn = order(results_genes[sig,"padj"])[1:25]
text(x[topn], y[topn], results_genes[topn,"Symbol"], col="black", cex=0.75, srt=45)

#### Plot #8 - Create a heatmap to vizualize expression differences between the six samples
#Define custom dist and hclust functions for use with heatmaps
mydist=function(c) {dist(c,method="euclidian")}
myclust=function(c) {hclust(c,method="average")}

#Create a subset of significant genes with p-value<0.05 and log2 fold-change >= 2
sigpi = which(results_genes[,"pvalue"]<0.05)
sigp = results_genes[sigpi,]
sigfc = which(abs(sigp[,"log2FoldChange"]) >= 2)
sigDE = sigp[sigfc,]

main_title="sig DE Genes"
par(cex.main=0.8)
sigDE_genes=sigDE[,"ensemblID"]
sigDE_genenames=sigDE[,"Symbol"]

data=log2(as.matrix(gene_expression[as.vector(sigDE_genes),data_columns])+1)
heatmap.2(data, hclustfun=myclust, distfun=mydist, na.rm = TRUE, scale="none", dendrogram="both", margins=c(10,4), Rowv=TRUE, Colv=TRUE, symbreaks=FALSE, key=TRUE, symkey=FALSE, density.info="none", trace="none", main=main_title, cexRow=0.3, cexCol=1, labRow=sigDE_genenames,col=rev(heat.colors(75)))

#### Plot #9 - Volcano plot

# Set differential expression status for each gene - default all genes to "no change"
results_genes$diffexpressed <- "No"
# if log2Foldchange > 2 and pvalue < 0.05, set as "Up regulated"
results_genes$diffexpressed[results_genes$log2FoldChange >= 2 & results_genes$pvalue < 0.05] <- "Higher in UHR"
# if log2Foldchange < -2 and pvalue < 0.05, set as "Down regulated"
results_genes$diffexpressed[results_genes$log2FoldChange <= -2 & results_genes$pvalue < 0.05] <- "Higher in HBR"

# write the gene names of those significantly upregulated/downregulated to a new column
results_genes$gene_label <- NA
results_genes$gene_label[results_genes$diffexpressed != "No"] <- results_genes$Symbol[results_genes$diffexpressed != "No"]

ggplot(data=results_genes[results_genes$diffexpressed != "No",], aes(x=log2FoldChange, y=-log10(pvalue), label=gene_label, color = diffexpressed)) +
             xlab("log2Foldchange") +
             scale_color_manual(name = "Differentially expressed", values=c("blue", "red")) +
             geom_point() +
             theme_minimal() +
             geom_text_repel() +
             geom_vline(xintercept=c(-0.6, 0.6), col="red") +
             geom_hline(yintercept=-log10(0.05), col="red") +
             guides(colour = guide_legend(override.aes = list(size=5))) +
             geom_point(data = results_genes[results_genes$diffexpressed == "No",], aes(x=log2FoldChange, y=-log10(pvalue)), colour = "black")


#To exit R type:
#quit(save="no")

The output file can be viewed in your browser at the following url. Note, you must replace YOUR_PUBLIC_IPv4_ADDRESS with your own amazon instance IP (e.g., 101.0.1.101)).

  • http://YOUR_PUBLIC_IPv4_ADDRESS/rnaseq/de/ballgown/ref_only/Tutorial_Part3_Supplementary_R_output.pdf

Visual comparison of example genes from the volcano plot

One can manually explore interesting looking genes from the volcano plot. In this case our analysis involves comparison of RNA isolated from tissues of different types (HBR -> brain tissue, UHR -> a collection of cancer cell lines). So, in this analysis it might make sense to explore candidates in a tissue expression atlas such as GTEX.

Looking at our gene plot, two example genes we could look at are: SEPT3 (significantly higher in HBR) and PRAME (significantly higher in UHR).

Expression of SEPT3 across tissues according to GTEX SEPT3 Note that this gene appears to be highly expressed in brain tissues.

Expression of PRAME across tissues according to GTEX PRAME Note that this gene appears to be almost uniquely expressed in testis tissue. Since one of the cell lines in the UHR sample is a testicular cancer cell line, this makes sense.


PRACTICAL EXERCISE 10 (ADVANCED)

Assignment: Use R to create a volcano plot for the differentially expressed genes you identified with Ballgown in Practical Exercise 9.

  • Hint: Follow the example R code above.
  • Hint: You could import the ballgown data object (e.g., bg.rda) that you should have saved in Practical Exercise 9 as a source of DE results.

Solution: When you are ready you can check your approach against the Solutions


DE Visualization with DESeq2 | Griffith Lab

RNA-seq Bioinformatics

Introduction to bioinformatics for RNA sequence analysis

DE Visualization with DESeq2


RNA-seq_Flowchart4


Differential Expression Visualzation

In this section we will be going over some basic visualizations of the DESeq2 results generated in the “Differential Expression with DESeq2” section of this course. Our goal is to quickly obtain some interpretable results using built-in visualization functions from DESeq2 or recommended packages. For a very extensive overview of DESeq2 and how to visualize and interpret the results, refer to the DESeq2 vignette.

Setup

If it is not already in your R environment, load the DESeqDataSet object and the results table into the R environment.

# set the working directory
setwd('/cloud/project/outdir')

# view the contents of this directory
dir()

# load libs
library(DESeq2)
library(data.table)
library(pheatmap)

# Load in the DESeqDataSet object
dds <- readRDS('dds.rds')

# Load in the results objects before and after shrinkage
res <- readRDS('res.rds')
resLFC <- readRDS('resLFC.rds')

# Load in the final results file with all sorted DE results
deGeneResultSorted <- fread('DE_all_genes_DESeq2.tsv')

MA-plot before LFC shrinkage

MA-plots were originally used to evaluate microarray expression data where M is the log ratio and A is the mean average (both based on scanned intensity measurements from the microarray).

These types of plots are still usefull in RNAseq DE experiments with two conditions, as they can immediately give us information on the number of signficantly differentially expressed genes, the ratio of up vs down regulated genes, and any outliers. To interpret these plots it is important to keep a couple of things in mind. The Y axis (M) is the log2 fold change between the two conditions tested, a higher fold-change indicates greater difference between condition A and condition B. The X axis (A) is a measure of read alignment to a gene, so as you go higher on on the X axis you are looking at regions which have higher totals of aligned reads, in other words the gene is “more” expressed overall (with the caveat that gene length is not being taken into account by raw read counts here).

Using the build in plotMA function from DESeq2 we also see that the genes are color coded by a significance threshold. Genes with higher expression values and higher fold-changes are more often significant as one would expect.

# use DESeq2 built in MA-plot function
plotMA(res, ylim=c(-2, 2))

MA-plot after LFC shrinkage

When we ran DESeq2 we obtained two results, one with and one without log-fold change shrinkage. When you have genes with low hits you can get some very large fold changes. For example 1 hit on a gene vs 6 hits on a gene is a 6x fold change. This high level of variance though is probably quantifying noise instead of real biology. Running plotMA on our results where we applied an algorithm for log fold change shrinkage we can see that this “effect” is somewhat controlled for.

# ma plot
plotMA(resLFC, ylim=c(-2,2))

The effect is very subtle here due to the focused nature of our dataset (chr22 genes onle), but if you toggle between the two plots and look in the upper left and bottom left corners you can see some fold change values are moving closer to 0.

Viewing individual gene counts between two conditions

Often it may be useful to view the normalized counts for a gene amongst our samples. DESeq2 provides a built in function for that which works off of the dds object. Here we view SEPT3 which we can see in our DE output is significantly higher in the UHR cohort. This is useful as we can see the per-sample distribution of our corrected counts, we can immediately determine if there are any outliers within each group and investigate further if need be.

# view SEPT3 normalized counts
plotCounts(dds, gene='ENSG00000100167', intgroup = 'Condition')

# view PRAME normalized counts
plotCounts(dds, gene='ENSG00000185686', intgroup = 'Condition')

Viewing pairwise sample clustering

It may often be useful to view inter-sample relatedness. In other words, how similar or disimilar samples are to one another overall. While not part of the DESeq2 package, there is a convenient library that can easily construct a hierarchically clustered heatmap from our DESeq2 data. It should be noted that when doing any sort of distance calculation the count data should be transformed using vst() or rlog(), this can be done directly on the dds object.

# note that we use rlog because we don't have a large number of genes, for a typical DE experiment with 1000's of genes use the vst() function
rld <- rlog(dds, blind=F)

# view the structure of this object
rld

# compute sample distances (the dist function uses the euclidean distance metric by default)
# in this command we will pull the rlog transformed data ("regularized" log2 transformed, see ?rlog for details) using "assay"
# then we transpose that data using t()
# then we calculate distance values using dist() 
# the distance is calculated for each vector of sample gene values, in a pairwise fashion comparing all samples

# view the first few lines of raw data
head(assay(dds))

# see the rlog transformed data
head(assay(rld))

# see the impact of transposing the matrix
t(assay(rld))[1:6,1:5]

# see the distance values
dist(t(assay(rld)))

# put it all together and store the result
sampleDists <- dist(t(assay(rld)))

# convert the distance result to a matrix
sampleDistMatrix <- as.matrix(sampleDists)

# view the distance numbers directly in the pairwise distance matrix
head(sampleDistMatrix)

# construct clustered heatmap, important to use the computed sample distances for clustering
pheatmap(sampleDistMatrix, clustering_distance_rows=sampleDists, clustering_distance_cols=sampleDists)

Instead of a distance metric we could also use a similarity metric such as a Peason correlation

There are many correlation and distance options:

Correlation: “pearson”, “kendall”, “spearman” Distance: “euclidean”, “maximum”, “manhattan”, “canberra”, “binary” or “minkowski”

sampleCorrs <- cor(assay(rld), method="pearson")
sampleCorrMatrix <- as.matrix(sampleCorrs)
head(sampleCorrMatrix)

pheatmap(sampleCorrMatrix)

Instead of boiling all the gene count data for each sample down to a distance metric you can get a similar sense of the pattern by just visualizing all the genes at once


# because there are so many gene we choose not to display them
pheatmap(mat=t(assay(rld)), show_colnames = FALSE)