Gene set enrichment analysis

Last updated on 2024-03-05 | Edit this page

Overview

Questions

  • What is the aim of performing gene set enrichment analysis?
  • What is the method of over-representation analysis?
  • What are the commonly-used gene set databases?

Objectives

  • Learn the method of gene set enrichment analysis.
  • Learn how to obtain gene sets from various resources in R.
  • Learn how to perform gene set enrichment analysis and how to visualize enrichment results.

After we have obtained a list of differentially expressed (DE) genes, the next question naturally to ask is what biological functions these DE genes may affect. Gene set enrichment analysis (GSEA) evaluates the associations of a list of DE genes to a collection of pre-defined gene sets, where each gene set has a specific biological meaning. Once DE genes are significantly enriched in a gene set, the conclusion is made that the corresponding biological meaning (e.g. a biological process or a pathway) is significantly affected.

The definition of a gene set is very flexible and the construction of gene sets is straightforward. In most cases, gene sets are from public databases where huge efforts from scientific curators have already been made to carefully categorize genes into gene sets with clear biological meanings. Nevertheless, gene sets can also be self-defined from individual studies, such as a set of genes in a network module from a co-expression network analysis, or a set of genes that are up-regulated in a certain disease.

There are a huge amount of methods available for GSEA analysis. In this episode, we will learn the simplest but the mostly used one: the over-representation analysis (ORA). ORA is directly applied to the list of DE genes and it evaluates the association of the DE genes and the gene set by the numbers of genes in different categories.

Please note, ORA is a universal method that it can not only be applied to the DE gene list, but also any type of gene list of interest to look for their statistically associated biological meanings.

In this episode, we will start with a tiny example to illustrate the statistical method of ORA. Next we will go through several commonly-used gene set databases and how to access them in R. Then, we will learn how to perform ORA analysis with the Bioconductor package clusterProfiler. And in the end, we will learn several visualization methods on the GSEA results.

Following is a list of packages that will be used in this episode:

R

library(SummarizedExperiment)
library(DESeq2)
library(gplots)
library(microbenchmark)
library(org.Hs.eg.db)
library(org.Mm.eg.db)
library(msigdbr)
library(clusterProfiler)
library(enrichplot)
library(ggplot2)
library(simplifyEnrichment)

The statistical method


To demonstrate the ORA analysis, we use a list of DE genes from a comparison between genders. The following code performs DESeq2 analysis which you should have already learnt in the previous episode. In the end, we have a list of DE genes filtered by FDR < 0.05, and save it in the object sexDEgenes.

The file data/GSE96870_se.rds contains a RangedSummarizedExperiment that contains RNA-Seq counts that were downloaded in Episode 2 and constructed in Episode 3 (minimal codes for downloading and constructing in the script download_data.R. In the following code, there are also comments that explain every step of the analysis.

R

library(SummarizedExperiment)
library(DESeq2)

# read the example dataset which is a `RangedSummarizedExperiment` object
se <- readRDS("data/GSE96870_se.rds")

# only restrict to mRNA (protein-coding genes)
se <- se[rowData(se)$gbkey == "mRNA"]

# construct a `DESeqDataSet` object where we also specify the experimental design
dds <- DESeqDataSet(se, design = ~ sex + time)
# perform DESeq2 analysis
dds <- DESeq(dds)
# obtain DESeq2 results, here we only want Male vs Female in the "sex" variable
resSex <- results(dds, contrast = c("sex", "Male", "Female"))
# extract DE genes with padj < 0.05
sexDE <- as.data.frame(subset(resSex, padj < 0.05))
# the list of DE genes
sexDEgenes <- rownames(sexDE)

Let’s check the number of DE genes and how they look like. It seems the number of DE genes is very small, but it is OK for this example.

R

length(sexDEgenes)

OUTPUT

[1] 54

R

head(sexDEgenes)

OUTPUT

[1] "Lgr6"   "Myoc"   "Fibcd1" "Kcna4"  "Ctxn2"  "S100a9"

Next we construct a gene set which contains genes on sex chromosomes (let’s call it the “XY gene set”). Recall the RangedSummarizedExperiment object also includes genomic locations of genes, thus we can simply obtain sex genes by filtering the chromosome names.

In the following code, geneGR is a GRanges object on which seqnames() is applied to extract chromosome names. seqnames() returns a special data format and we need to explicitly convert it to a normal vector by as.vector().

R

geneGR <- rowRanges(se)
totalGenes <- rownames(se)
XYGeneSet <- totalGenes[as.vector(seqnames(geneGR)) %in% c("X", "Y")]
head(XYGeneSet)

OUTPUT

[1] "Gm21950"   "Gm14346"   "Gm14345"   "Gm14351"   "Spin2-ps1" "Gm3701"   

R

length(XYGeneSet)

OUTPUT

[1] 1134

The format of a single gene set is very straightforward, which is simply a vector. The ORA analysis is applied on the DE gene vector and gene set vector.

Before we move on, one thing worth to mention is that ORA deals with two gene vectors. To correctly map between them, gene ID types must be consistent in the two vectors. In this tiny example, since both DE genes and the XY gene set are from the same object se, they are ensured to be in the same gene ID types (the gene symbol). But in general, DE genes and gene sets are from two different sources (e.g. DE genes are from researcher’s experiment and gene sets are from public databases), it is very possible that gene IDs are not consistent in the two. Later in this episode, we will learn how to perform gene ID conversion in the ORA analysis.

Since the DE genes and the gene set can be mathematically thought of as two sets, a natural way is to first visualize them with a Venn diagram.

R

library(gplots)
plot(venn(list("sexDEgenes"  = sexDEgenes, 
               "XY gene set" = XYGeneSet)))
title(paste0("|universe| = ", length(totalGenes)))

In the Venn diagram, we can observe that around 1.1% (13/1134) of genes in the XY gene set are DE. Compared to the global fraction of DE genes (54/21198 = 0.25%), it seems there is a strong relations between DE genes and the gene set. We can also compare the fraction of DE genes that belong to the gene set (13/54 = 24.1%) and the global fraction of XY gene set in the genome (1134/21198 = 5.3%). On the other hand, it is quite expected because the two events are actually biologically relevant where one is from a comparison between genders and the other is the set of gender-related genes.

Then, how to statistically measure the enrichment or over-representation? Let’s go to the next section.

Fisher’s exact test

To statistically measure the enrichment, the relationship of DE genes and the gene set is normally formatted into the following 2x2 contingency table, where in the table are the numbers of genes in different categories. \(n_{+1}\) is the size of the XY gene set (i.e. the number of member genes), \(n_{1+}\) is the number of DE genes, \(n\) is the number of total genes.

In the gene set Not in the gene set Total
DE \(n_{11}\) \(n_{12}\) \(n_{1+}\)
Not DE \(n_{21}\) \(n_{22}\) \(n_{2+}\)
Total \(n_{+1}\) \(n_{+2}\) \(n\)

These numbers can be obtained as in the following code1. Note we replace + with 0 in the R variable names.

R

n    <- nrow(se)
n_01 <- length(XYGeneSet)
n_10 <- length(sexDEgenes)
n_11 <- length(intersect(sexDEgenes, XYGeneSet))

Other values can be obtained by:

R

n_12 <- n_10 - n_11
n_21 <- n_01 - n_11
n_20 <- n    - n_10
n_02 <- n    - n_01
n_22 <- n_02 - n_12

All the values are:

R

matrix(c(n_11, n_12, n_10, n_21, n_22, n_20, n_01, n_02, n),
    nrow = 3, byrow = TRUE)

OUTPUT

     [,1]  [,2]  [,3]
[1,]   13    41    54
[2,] 1121 20023 21144
[3,] 1134 20064 21198

And we fill these numbers into the 2x2 contingency table:

In the gene set Not in the gene set Total
DE 13 41 54
Not DE 1121 20023 21144
Total 1134 20064 21198

Fisher’s exact test can be used to test the associations of the two marginal attributes, i.e. is there a dependency of a gene to be a DE gene and to be in the XY gene set? In R, we can use the function fisher.test() to perform the test. The input is the top-left 2x2 sub-matrix. We specify alternative = "greater" in the function because we are only interested in over-representation.

R

fisher.test(matrix(c(n_11, n_12, n_21, n_22), nrow = 2, byrow = TRUE),
    alternative = "greater")

OUTPUT


	Fisher's Exact Test for Count Data

data:  matrix(c(n_11, n_12, n_21, n_22), nrow = 2, byrow = TRUE)
p-value = 3.906e-06
alternative hypothesis: true odds ratio is greater than 1
95 percent confidence interval:
 3.110607      Inf
sample estimates:
odds ratio 
  5.662486 

In the output, we can see the p-value is very small (3.906e-06), then we can conclude DE genes have a very strong enrichment in the XY gene set.

Results of the Fisher’s Exact test can be saved into an object t, which is a simple list, and the p-value can be obtained by t$p.value.

R

t <- fisher.test(matrix(c(n_11, n_12, n_21, n_22), nrow = 2, byrow = TRUE),
    alternative = "greater")
t$p.value

OUTPUT

[1] 3.9059e-06

Odds ratio from the Fisher’s exact test is defined as follows:

\[ \mathrm{Odds\_ratio} = \frac{n_{11}/n_{21}}{n_{12}/n_{22}} = \frac{n_{11}/n_{12}}{n_{21}/n_{22}} = \frac{n_{11} * n_{22}}{n_{12} * n_{21}} \]

If there is no association between DE genes and the gene set, odds ratio is expected to be 1. And it is larger than 1 if there is an over-representation of DE genes on the gene set.

Further reading

The 2x2 contingency table can be transposed and it does not affect the Fisher’s exact test. E.g. let’s put whether genes are in the gene sets on rows, and put whether genes are DE on columns.

DE Not DE Total
In the gene set 13 1121 1134
Not in the gene set 41 20023 20064
Total 54 21144 21198

And the corresponding fisher.test() is:

R

fisher.test(matrix(c(13, 1121, 41, 20023), nrow = 2, byrow = TRUE),
    alternative = "greater")

OUTPUT


	Fisher's Exact Test for Count Data

data:  matrix(c(13, 1121, 41, 20023), nrow = 2, byrow = TRUE)
p-value = 3.906e-06
alternative hypothesis: true odds ratio is greater than 1
95 percent confidence interval:
 3.110607      Inf
sample estimates:
odds ratio 
  5.662486 

The hypergeometric distribution

We can look at the problem from another aspect. This time we treat all genes as balls in a big box where all the genes have the same probability to be picked up. Some genes are marked as DE genes (in red in the figure) and other genes are marked as non-DE genes (in blue). We grab \(n_{+1}\) genes (the size of the gene set) from the box and we want to ask what is the probability of having \(n_{11}\) DE genes in our hand?

We first calculate the total number of ways of picking \(n_{+1}\) genes from total \(n\) genes, without distinguishing whether they are DE or not: \(\binom{n}{n_{+1}}\).

Next, in the \(n_{+1}\) genes that have been picked, there are \(n_{11}\) DE genes which can only be from the total \(n_{1+}\) DE genes. Then the number of ways of picking \(n_{11}\) DE genes from \(n_{1+}\) total DE genes is \(\binom{n_{1+}}{n_{11}}\).

Similarly, there are still \(n_{21}\) non-DE genes in our hand, which can only be from the total \(n_{2+}\) non-DE genes. Then the number of ways of picking \(n_{21}\) non-DE genes from \(n_{2+}\) total non-DE genes: \(\binom{n_{2+}}{n_{21}}\).

Since picking DE genes and picking non-DE genes are independent, the number of ways of picking \(n_{+1}\) genes which contain \(n_{11}\) DE genes and \(n_{21}\) non-DE genes is their multiplication: \(\binom{n_{1+}}{n_{11}} \binom{n_{2+}}{n_{21}}\).

And the probability \(P\) is:

\[P = \frac{\binom{n_{1+}}{n_{11}} \binom{n_{2+}}{n_{21}}}{\binom{n}{n_{+1}}} = \frac{\binom{n_{1+}}{n_{11}} \binom{n - n_{1+}}{n_{+1} -n_{11}}}{\binom{n}{n_{+1}}} \]

where in the denominator is the number of ways of picking \(n_{+1}\) genes without distinguishing whether they are DE or not.

If \(n\) (number of total genes), \(n_{1+}\) (number of DE genes) and \(n_{+1}\) (size of gene set) are all fixed values, the number of DE genes that are picked can be denoted as a random variable \(X\). Then \(X\) follows the hypergeometric distribution with parameters \(n\), \(n_{1+}\) and \(n_{+1}\), written as:

\[ X \sim \mathrm{Hyper}(n, n_{1+}, n_{+1})\]

The p-value of the enrichment is calculated as the probability of having an observation equal to or larger than \(n_{11}\) under the assumption of independence:

\[ \mathrm{Pr}( X \geqslant n_{11} ) = \sum_{x \in \{ {n_{11}, n_{11}+1, ..., \min\{n_{1+}, n_{+1}\} \}}} \mathrm{Pr}(X = x) \]

In R, the function phyper() calculates p-values from the hypergeometric distribution. There are four arguments:

R

phyper(q, m, n, k)

which are:

  • q: the observation,
  • m: number of DE genes,
  • n: number of non-DE genes,
  • k: size of the gene set.

phyper() calculates \(\mathrm{Pr}(X \leqslant q)\). To calculate \(\mathrm{Pr}(X \geqslant q)\), we need to transform it a little bit:

\[ \mathrm{Pr}(X \geqslant q) = 1 - \mathrm{Pr}(X < q) = 1 - \mathrm{Pr}(X \leqslant q-1)\]

Then, the correct use of phyper() is:

R

1 - phyper(q - 1, m, n, k)

Let’s plugin our variables:

R

1 - phyper(n_11 - 1, n_10, n_20, n_01)

OUTPUT

[1] 3.9059e-06

Optionally, lower.tail argument can be specified which directly calculates p-values from the upper tail of the distribution.

R

phyper(n_11 - 1, n_10, n_20, n_01, lower.tail = FALSE)

OUTPUT

[1] 3.9059e-06

If we switch n_01 and n_10, the p-values are identical:

R

1 - phyper(n_11 - 1, n_01, n_02, n_10)

OUTPUT

[1] 3.9059e-06

fisher.test() and phyper() give the same p-value. Actually the two methods are identical because in Fisher’s exact test, hypergeometric distribution is the exact distribution of its statistic.

Let’s test the runtime of the two functions:

R

library(microbenchmark)
microbenchmark(
    fisher = fisher.test(matrix(c(n_11, n_12, n_21, n_22), nrow = 2, byrow = TRUE),
        alternative = "greater"),
    hyper = 1 - phyper(n_11 - 1, n_10, n_20, n_01)
)

OUTPUT

Unit: microseconds
   expr     min      lq      mean  median       uq      max neval
 fisher 238.955 243.980 270.67608 248.163 259.2635 1920.491   100
  hyper   1.333   1.488   2.28346   2.550   2.8505    6.953   100

It is very astonishing that phyper() is hundreds of times faster than fisher.test(). Main reason is in fisher.test(), there are many additional calculations besides calculating the p-value. So if you want to implement ORA analysis by yourself, always consider to use phyper()2.

Further reading

Current tools also use Binomial distribution or chi-square test for ORA analysis. These two are just approximations. Please refer to Rivals et al., Enrichment or depletion of a GO category within a class of genes: which test? Bioinformatics 2007 which gives an overview of statistical methods used in ORA analysis.

Gene set resources


We have learnt the basic methods of ORA analysis. Now we go to the second component of the analysis: the gene sets.

Gene sets represent prior knowledge of what is the general shared biological attribute of genes in the gene set. For example, in a “cell cycle” gene set, all the genes are involved in the cell cycle process. Thus, if DE genes are significantly enriched in the “cell cycle” gene set, which means there are significantly more cell cycle genes differentially expressed than expected, we can conclude that the normal function of cell cycle process may be affected.

As we have mentioned, genes in the gene set share the same “biological attribute” where “the attribute” will be used for making conclusions. The definition of “biological attribute” is very flexible. It can be a biological process such as “cell cycle”. It can also be from a wide range of other definitions, to name a few:

  • Locations in the cell, e.g. cell membrane or cell nucleus.
  • Positions on chromosomes, e.g. sex chromosomes or the cytogenetic band p13 on chromome 10.
  • Target genes of a transcription factor or a microRNA, e.g. all genes that are transcriptionally regulationed by NF-κB.
  • Signature genes in a certain tumor type, i.e. genes that are uniquely highly expressed in a tumor type.

The MSigDB database contains gene sets in many topics. We will introduce it later in this section.

You may have encountered many different ways to name gene sets: “gene sets”, “biological terms”, “GO terms”, “GO gene sets”, “pathways”, and so on. They basically refer to the same thing, but from different aspects. As shown in the following figure, “gene set” corresponds to a vector of genes and it is the representation of the data for computation. “Biological term” is a textual entity that contains description of its biological meaning; It corresponds to the knowledge of the gene set and is for the inference of the analysis. “GO gene sets” and “pathways” specifically refer to the enrichment analysis using GO gene sets and pahtway gene sets.

Before we touch the gene set databases, we first summarize the general formats of gene sets in R. In most analysis, a gene set is simply treated as a vector of genes. Thus, naturally, a collection of gene sets can be represented as a list of vectors. In the following example, there are three gene sets with 3, 5 and 2 genes. Some genes exist in multiple gene sets.

R

lt <- list(gene_set_1 = c("gene_1", "gene_2", "gene_3"),
           gene_set_2 = c("gene_1", "gene_3", "gene_4", "gene_5", "gene_6"),
           gene_set_3 = c("gene_4", "gene_7")
)
lt

OUTPUT

$gene_set_1
[1] "gene_1" "gene_2" "gene_3"

$gene_set_2
[1] "gene_1" "gene_3" "gene_4" "gene_5" "gene_6"

$gene_set_3
[1] "gene_4" "gene_7"

It is also very common to store the relations of gene sets and genes as a two-column data frame. The order of the gene set column and the gene column, i.e. which column locates as the first column, are quite arbitrary. Different tools may require differently.

R

data.frame(gene_set = rep(names(lt), times = sapply(lt, length)),
           gene = unname(unlist(lt)))

OUTPUT

     gene_set   gene
1  gene_set_1 gene_1
2  gene_set_1 gene_2
3  gene_set_1 gene_3
4  gene_set_2 gene_1
5  gene_set_2 gene_3
6  gene_set_2 gene_4
7  gene_set_2 gene_5
8  gene_set_2 gene_6
9  gene_set_3 gene_4
10 gene_set_3 gene_7

Or genes be in the first column:

OUTPUT

     gene   gene_set
1  gene_1 gene_set_1
2  gene_2 gene_set_1
3  gene_3 gene_set_1
4  gene_1 gene_set_2
5  gene_3 gene_set_2
6  gene_4 gene_set_2
7  gene_5 gene_set_2
8  gene_6 gene_set_2
9  gene_4 gene_set_3
10 gene_7 gene_set_3

Not very often, gene sets are represented as a matrix where one dimension corresponds to gene sets and the other dimension corresponds to genes. The values in the matix are binary where a value of 1 represents the gene is a member of the corresponding gene sets. In some methods, 1 is replaced by \(w_{ij}\) to weight the effect of the genes in the gene set.

#            gene_1 gene_2 gene_3 gene_4
# gene_set_1      1      1      0      0
# gene_set_2      1      0      1      1

Challenge

Can you convert between different gene set representations? E.g. convert a list to a two-column data frame?

R

lt <- list(gene_set_1 = c("gene_1", "gene_2", "gene_3"),
           gene_set_2 = c("gene_1", "gene_3", "gene_4", "gene_5", "gene_6"),
           gene_set_3 = c("gene_4", "gene_7")
)

To convert lt to a data frame (e.g. let’s put gene sets in the first column):

R

df = data.frame(gene_set = rep(names(lt), times = sapply(lt, length)),
                gene = unname(unlist(lt)))
df

OUTPUT

     gene_set   gene
1  gene_set_1 gene_1
2  gene_set_1 gene_2
3  gene_set_1 gene_3
4  gene_set_2 gene_1
5  gene_set_2 gene_3
6  gene_set_2 gene_4
7  gene_set_2 gene_5
8  gene_set_2 gene_6
9  gene_set_3 gene_4
10 gene_set_3 gene_7

To convert df back to the list:

R

split(df$gene, df$gene_set)

OUTPUT

$gene_set_1
[1] "gene_1" "gene_2" "gene_3"

$gene_set_2
[1] "gene_1" "gene_3" "gene_4" "gene_5" "gene_6"

$gene_set_3
[1] "gene_4" "gene_7"

Next, let’s go through gene sets from several major databases: the GO, KEGG and MSigDB databases.

Gene Ontology gene sets

Gene Ontology (GO) is the standard source for gene set enrichment analysis. GO contains three namespaces of biological process (BP), cellular components (CC) and molecular function (MF) which describe a biological entity from different aspect. The associations between GO terms and genes are integrated in the Bioconductor standard packages: the organism annotation packages. In the current Bioconductor release (3.17), there are the following organism packages:

Package Organism Package Organism
org.Hs.eg.db Human org.Mm.eg.db Mouse
org.Rn.eg.db Rat org.Dm.eg.db Fly
org.At.tair.db Arabidopsis org.Dr.eg.db Zebrafish
org.Sc.sgd.db Yeast org.Ce.eg.db Worm
org.Bt.eg.db Bovine org.Gg.eg.db Chicken
org.Ss.eg.db Pig org.Mmu.eg.db Rhesus
org.Cf.eg.db Canine org.EcK12.eg.db E coli strain K12
org.Xl.eg.db Xenopus org.Pt.eg.db Chimp
org.Ag.eg.db Anopheles org.EcSakai.eg.db E coli strain Sakai

There are four sections in the name of an organism package. The naming convention is: org simply means “organism”. The second section corresponds to a specific organism, e.g. Hs for human and Mm for mouse. The third section corresponds to the primary gene ID type used in the package, where normally eg is used which means “Entrez genes” because data is mostly retrieved from the NCBI database. However, for some organisms, the primary ID can be from its own primary database, e.g. sgd for Yeast which corresponds to the Saccharomyces Genome Database, the primary database for yeast. The last section is always “db”, which simply implies it is a database package.

Taking the org.Hs.eg.db package as an example, all the data is stored in a database object org.Hs.eg.db in the OrgDb class. The object contains a connection to a local SQLite database. Users can simply think org.Hs.eg.db as a huge table that contains ID mappings between various databases. GO gene sets are essentially mappings between GO terms and genes. Let’s try to extract it from the org.Hs.eg.db object.

All the columns (the key column or the source column) can be obtained by keytypes():

R

library(org.Hs.eg.db)
keytypes(org.Hs.eg.db)

OUTPUT

 [1] "ACCNUM"       "ALIAS"        "ENSEMBL"      "ENSEMBLPROT"  "ENSEMBLTRANS"
 [6] "ENTREZID"     "ENZYME"       "EVIDENCE"     "EVIDENCEALL"  "GENENAME"    
[11] "GENETYPE"     "GO"           "GOALL"        "IPI"          "MAP"         
[16] "OMIM"         "ONTOLOGY"     "ONTOLOGYALL"  "PATH"         "PFAM"        
[21] "PMID"         "PROSITE"      "REFSEQ"       "SYMBOL"       "UCSCKG"      
[26] "UNIPROT"     

To get the GO gene sets, we first obtain all GO IDs under the BP (biological process) namespace. As shown in the output from keytype(), "ONTOLOGY" is also a valid “key column”, thus we can query “select all GO IDs where the corresponding ONTOLOGY is BP”, which is translated into the following code:

R

BP_Id = mapIds(org.Hs.eg.db, keys = "BP", keytype = "ONTOLOGY", 
               column = "GO", multiVals = "list")[[1]]
head(BP_Id)

OUTPUT

[1] "GO:0008150" "GO:0001553" "GO:0001869" "GO:0002438" "GO:0006953"
[6] "GO:0007584"

mapIds() maps IDs between two sources. Since a GO namespace have more than one GO terms, we have to set multiVals = "list" to obtain all GO terms under that namespace. And since we only query for one GO “ONTOLOGY”, we directly take the first element from the list returned by mapIds().

Next we do mapping from GO IDs to gene Entrez IDs. Now the query becomes “providing a vector of GO IDs, select ENTREZIDs which correspond to every one of them”.

R

BPGeneSets = mapIds(org.Hs.eg.db, keys = BP_Id, keytype = "GOALL", 
                    column = "ENTREZID", multiVals = "list")

You may have noticed there is a “GO” key column as well a “GOALL” column in the database. As GO has a hierarchical structure where a child term is a sub-class of a parent term. All the genes annotated to a child term are also annotated to its parent terms. To reduce the duplicated information when annotating genes to GO terms, genes are normally annotated to the most specific offspring terms in the GO hierarchy. Upstream merging of gene annotations should be done by the tools which perform analysis. In this way, the mapping between "GO" and "ENTREZID" only contains “primary” annotations which is not complete, and mapping between "GOALL" and "ENTREZID" is the correct one to use.

We filter out GO gene sets with no gene annotated.

R

BPGeneSets = BPGeneSets[sapply(BPGeneSets, length) > 0]
BPGeneSets[2:3] # BPGeneSets[[1]] is too long

OUTPUT

$`GO:0001553`
 [1] "2"     "2516"  "2661"  "2661"  "3624"  "4313"  "5156"  "5798"  "6777" 
[10] "8322"  "8879"  "56729" "59338"

$`GO:0001869`
[1] "2"   "710"

In most cases, because OrgDb is a standard Bioconductor data structure, most tools can automatically construct GO gene sets internally. There is no need for users to touch such low-level processings.

Further reading

Mapping between various databases can also be done with the general select() interface. If the OrgDb object is provided by a package such as org.Hs.eg.db, there is also a separated object that specifically contains mapping between GO terms and genes. Readers can check the documentation of org.Hs.egGO2ALLEGS. Additional information on GO terms such as GO names and long descriptions are available in the package GO.db.

Bioconductor has already provided a large number of organism packages. However, if the organism you are working on is not supported there, you may consider to look for it with the AnnotationHub package, which additionally provide OrgDb objects for approximately 2000 organisms. The OrgDb object can be directly used in the ORA analysis introduced in the next section.

KEGG gene sets

A biological pathway is a series of interactions among molecules in a cell that leads to a certain product or a change in a cell3. A pathway involves a list of genes playing different roles which constructs the “pathway gene set”. KEGG pathway is the mostly used database for pathways. It provides its data via a REST API (https://rest.kegg.jp/). There are several commands to retrieve specific types of data. To retrieve the pathway gene sets, we can use the “link” command as shown in the following URL (“link” means to link genes to pathways). When you enter the URL in the web browser:

https://rest.kegg.jp/link/pathway/hsa

there will be a text table which contains a column of genes and a column of pathway IDs.

hsa:10327   path:hsa00010
hsa:124 path:hsa00010
hsa:125 path:hsa00010
hsa:126 path:hsa00010

We can directly read the text output with read.table(). Wrapping the URL with the function url(), you can pretend to directly read data from the remote web server.

R

keggGeneSets = read.table(url("https://rest.kegg.jp/link/pathway/hsa"), sep = "\t")
head(keggGeneSets)

OUTPUT

         V1            V2
1 hsa:10327 path:hsa00010
2   hsa:124 path:hsa00010
3   hsa:125 path:hsa00010
4   hsa:126 path:hsa00010
5   hsa:127 path:hsa00010
6   hsa:128 path:hsa00010

In this two-column table, the first column contains genes in the Entrez ID type. Let’s remove the "hsa:" prefix, also we remove the "path:" prefix for pathway IDs in the second column.

R

keggGeneSets[, 1] = gsub("hsa:", "", keggGeneSets[, 1])
keggGeneSets[, 2] = gsub("path:", "", keggGeneSets[, 2])
head(keggGeneSets)

OUTPUT

     V1       V2
1 10327 hsa00010
2   124 hsa00010
3   125 hsa00010
4   126 hsa00010
5   127 hsa00010
6   128 hsa00010

The full pathway names can be obtained via the “list” command.

R

keggNames = read.table(url("https://rest.kegg.jp/list/pathway/hsa"), sep = "\t")
head(keggNames)

OUTPUT

        V1                                                     V2
1 hsa01100              Metabolic pathways - Homo sapiens (human)
2 hsa01200               Carbon metabolism - Homo sapiens (human)
3 hsa01210 2-Oxocarboxylic acid metabolism - Homo sapiens (human)
4 hsa01212           Fatty acid metabolism - Homo sapiens (human)
5 hsa01230     Biosynthesis of amino acids - Homo sapiens (human)
6 hsa01232           Nucleotide metabolism - Homo sapiens (human)

In both commands, we obtained data for human where the corresponding KEGG code is "hsa". The code for other organisms can be found from the KEGG website (e.g. "mmu" for mouse), or via https://rest.kegg.jp/list/organism.

Keep in mind, KEGG pathways are only free for academic users. If you use it for commercial purposes, please contact the KEGG team to get a licence.

Further reading

Instead directly reading from the URLs, there are also R packages which help to obtain data from the KEGG database, such as the KEGGREST package or the download_KEGG() function from the clusterProfiler package. But essentially, they all obtain KEGG data with the REST API.

MSigDB gene sets

Molecular signature database (MSigDB) is a manually curated gene set database. Initially, it was proposed as a supplementary dataset for the original GSEA paper. Later it has been separated out and developed independently. In the first version in 2005, there were only two gene sets collections and in total 843 gene sets. Now in the newest version of MSigDB (v2023.1.Hs), it has grown into nine gene sets collections, covering > 30K gene sets. It provides gene sets on a variety of topics.

MSigDB categorizes gene sets into nine collections where each collection focuses on a specific topic. For some collections, they are additionally split into sub-collections. There are several ways to obtain gene sets from MSigDB. One convenient way is to use the msigdbr package. The advantages is msigdbr supports many other organisms by mapping to orthologs.

Let’s check which organisms are supported and which gene sets collections it provides.

R

library(msigdbr)
msigdbr_species()

OUTPUT

# A tibble: 20 × 2
   species_name                    species_common_name                          
   <chr>                           <chr>                                        
 1 Anolis carolinensis             Carolina anole, green anole                  
 2 Bos taurus                      bovine, cattle, cow, dairy cow, domestic cat…
 3 Caenorhabditis elegans          <NA>                                         
 4 Canis lupus familiaris          dog, dogs                                    
 5 Danio rerio                     leopard danio, zebra danio, zebra fish, zebr…
 6 Drosophila melanogaster         fruit fly                                    
 7 Equus caballus                  domestic horse, equine, horse                
 8 Felis catus                     cat, cats, domestic cat                      
 9 Gallus gallus                   bantam, chicken, chickens, Gallus domesticus 
10 Homo sapiens                    human                                        
11 Macaca mulatta                  rhesus macaque, rhesus macaques, Rhesus monk…
12 Monodelphis domestica           gray short-tailed opossum                    
13 Mus musculus                    house mouse, mouse                           
14 Ornithorhynchus anatinus        duck-billed platypus, duckbill platypus, pla…
15 Pan troglodytes                 chimpanzee                                   
16 Rattus norvegicus               brown rat, Norway rat, rat, rats             
17 Saccharomyces cerevisiae        baker's yeast, brewer's yeast, S. cerevisiae 
18 Schizosaccharomyces pombe 972h- <NA>                                         
19 Sus scrofa                      pig, pigs, swine, wild boar                  
20 Xenopus tropicalis              tropical clawed frog, western clawed frog    

R

msigdbr_collections()

OUTPUT

# A tibble: 23 × 3
   gs_cat gs_subcat         num_genesets
   <chr>  <chr>                    <int>
 1 C1     ""                         299
 2 C2     "CGP"                     3384
 3 C2     "CP"                        29
 4 C2     "CP:BIOCARTA"              292
 5 C2     "CP:KEGG"                  186
 6 C2     "CP:PID"                   196
 7 C2     "CP:REACTOME"             1615
 8 C2     "CP:WIKIPATHWAYS"          664
 9 C3     "MIR:MIRDB"               2377
10 C3     "MIR:MIR_Legacy"           221
11 C3     "TFT:GTRD"                 518
12 C3     "TFT:TFT_Legacy"           610
13 C4     "CGN"                      427
14 C4     "CM"                       431
15 C5     "GO:BP"                   7658
16 C5     "GO:CC"                   1006
17 C5     "GO:MF"                   1738
18 C5     "HPO"                     5071
19 C6     ""                         189
20 C7     "IMMUNESIGDB"             4872
21 C7     "VAX"                      347
22 C8     ""                         700
23 H      ""                          50

The first column in the above output is the primary category of gene sets. Some gene sets collections may have sub-collections, and they are shown in the second column. The description of gene sets collections from MSigDB is in the following figure.

The core function msigdbr() retrieves gene sets in a specific category (or a subcategory if it exists).

R

msigdbr(species, category, subcategory)

For example, we want to obtain the hallmark gene sets for mouse.

R

MSigDBGeneSets = msigdbr(species = "mouse", category = "H")
head(MSigDBGeneSets)

OUTPUT

# A tibble: 6 × 18
  gs_cat gs_subcat gs_name               gene_symbol entrez_gene ensembl_gene   
  <chr>  <chr>     <chr>                 <chr>             <int> <chr>          
1 H      ""        HALLMARK_ADIPOGENESIS Abca1             11303 ENSMUSG0000001…
2 H      ""        HALLMARK_ADIPOGENESIS Abcb8             74610 ENSMUSG0000002…
3 H      ""        HALLMARK_ADIPOGENESIS Acaa2             52538 ENSMUSG0000003…
4 H      ""        HALLMARK_ADIPOGENESIS Acadl             11363 ENSMUSG0000002…
5 H      ""        HALLMARK_ADIPOGENESIS Acadm             11364 ENSMUSG0000006…
6 H      ""        HALLMARK_ADIPOGENESIS Acads             11409 ENSMUSG0000002…
# ℹ 12 more variables: human_gene_symbol <chr>, human_entrez_gene <int>,
#   human_ensembl_gene <chr>, gs_id <chr>, gs_pmid <chr>, gs_geoid <chr>,
#   gs_exact_source <chr>, gs_url <chr>, gs_description <chr>, taxon_id <int>,
#   ortholog_sources <chr>, num_ortholog_sources <dbl>

The output is in the tibble class. If you have no experience with it, don’t worry, just take it as a table. As you can see, the three major gene IDs type "gene_symbol", "entrez_gene" and "ensembl_gene" are all included in the table. So users can easily pick one with the same gene ID type as in the DE gene list. For example:

R

MSigDBGeneSets[, c("gs_name", "ensembl_gene")]

OUTPUT

# A tibble: 7,384 × 2
   gs_name               ensembl_gene      
   <chr>                 <chr>             
 1 HALLMARK_ADIPOGENESIS ENSMUSG00000015243
 2 HALLMARK_ADIPOGENESIS ENSMUSG00000028973
 3 HALLMARK_ADIPOGENESIS ENSMUSG00000036880
 4 HALLMARK_ADIPOGENESIS ENSMUSG00000026003
 5 HALLMARK_ADIPOGENESIS ENSMUSG00000062908
 6 HALLMARK_ADIPOGENESIS ENSMUSG00000029545
 7 HALLMARK_ADIPOGENESIS ENSMUSG00000020917
 8 HALLMARK_ADIPOGENESIS ENSMUSG00000022477
 9 HALLMARK_ADIPOGENESIS ENSMUSG00000020777
10 HALLMARK_ADIPOGENESIS ENSMUSG00000022994
# ℹ 7,374 more rows

R

# or put genes in the first column
MSigDBGeneSets[, c("ensembl_gene", "gs_name")]

OUTPUT

# A tibble: 7,384 × 2
   ensembl_gene       gs_name              
   <chr>              <chr>                
 1 ENSMUSG00000015243 HALLMARK_ADIPOGENESIS
 2 ENSMUSG00000028973 HALLMARK_ADIPOGENESIS
 3 ENSMUSG00000036880 HALLMARK_ADIPOGENESIS
 4 ENSMUSG00000026003 HALLMARK_ADIPOGENESIS
 5 ENSMUSG00000062908 HALLMARK_ADIPOGENESIS
 6 ENSMUSG00000029545 HALLMARK_ADIPOGENESIS
 7 ENSMUSG00000020917 HALLMARK_ADIPOGENESIS
 8 ENSMUSG00000022477 HALLMARK_ADIPOGENESIS
 9 ENSMUSG00000020777 HALLMARK_ADIPOGENESIS
10 ENSMUSG00000022994 HALLMARK_ADIPOGENESIS
# ℹ 7,374 more rows

If you only want to use a sub-category gene sets, e.g. "CP:KEGG" from "C2", you can simply specify both category and subcategory arguments:

R

msigdbr(species = "mouse", category = "C2", subcategory = "CP:KEGG")

ORA with clusterProfiler


The ORA method itself is quite simple and it has been implemented in a large number of R packages. Among them, the clusterProfiler package especially does a good job in that it has a seamless integration to the Bioconductor annotation resources which allows extending GSEA analysis to other organisms easily; it has pre-defined functions for common analysis tasks, e.g. GO enrichment, KEGG enrichment; and it implements a variety of different visualization methods on the GSEA results. In this section, we will learn how to perform ORA analysis with clusterProfiler.

Here we will use a list of DE genes from a different comparison. As you may still remember, there are only 54 DE genes between genders, which may not be a good case for GSEA analysis, since after overlapping to a collection of gene sets, the majority of the gene sets will have few or even no gene overlapped. In this example we use the list of DE genes from the comparison between different time points.

Another thing worth to mention is, in the following code where we filter DE genes, we additionally add a filtering on the log2 fold change. This is recommended when the number of DE genes is too large. The filtering on log2 fold change can be thought as a filtering from the biology aspect.

R

resTime <- DESeq2::results(dds, contrast = c("time", "Day8", "Day0"))
timeDE <- as.data.frame(subset(resTime, 
                               padj < 0.05 & abs(log2FoldChange) > log2(1.5)
                       ))
timeDEgenes <- rownames(timeDE)
head(timeDEgenes)

OUTPUT

[1] "3110035E14Rik" "Sgk3"          "Kcnb2"         "Sbspon"       
[5] "Gsta3"         "Lman2l"       

R

length(timeDEgenes)

OUTPUT

[1] 1134

Let’s confirm that there are around one thousand DE genes, and the DE genes are in gene symbols.

GO enrichment

In clusterProfiler, there is an enrichGO() function which performs ORA on GO gene sets. To use it, we need to provide the DE genes, the organism OrgDb object which is from the organism package org.Mm.eg.db (because our data is from mouse), also the GO namespace (one of "BP", "CC" and "MF"). The GO gene sets are automatically retrieved and processed from org.Mm.eg.db in enrichGO().

R

library(clusterProfiler)
library(org.Mm.eg.db)
resTimeGO = enrichGO(gene = timeDEgenes, 
                     ont = "BP", 
                     OrgDb = org.Mm.eg.db)

OUTPUT

--> No gene can be mapped....

OUTPUT

--> Expected input gene ID: 23920,12589,22068,330470,21808,74716

OUTPUT

--> return NULL...

Oops, something seems wrong. Well, this is a common mistake where the gene ID types do not match between DE genes and the gene sets. Thankfully, the message clearly explains the reason. The ID type for gene sets is Entrez ID and it cannot match any DE gene.

There are two ways to solve this problem. 1. Convert gene IDs in timeDEgenes to Entrez IDs in advance; or 2. Simply specify the ID type of DE genes and let enrichGO() do the conversion job (recall various gene ID types are also stored in the OrgDb object). Let’s choose the second way.

In the next code, we additionally specify keyType = "SYMBOL" to explicitly tell the function that DE genes are in gene symbols. Recall that all valid values for keyType are in keytypes(org.Mm.eg.db).

R

resTimeGO = enrichGO(gene = timeDEgenes, 
                     keyType = "SYMBOL",
                     ont = "BP", 
                     OrgDb = org.Mm.eg.db)
resTimeGOTable = as.data.frame(resTimeGO)
head(resTimeGOTable)

OUTPUT

                   ID                 Description GeneRatio   BgRatio
GO:0050900 GO:0050900         leukocyte migration    49/969 386/28564
GO:0030595 GO:0030595        leukocyte chemotaxis    35/969 230/28564
GO:0071674 GO:0071674  mononuclear cell migration    30/969 185/28564
GO:0060326 GO:0060326             cell chemotaxis    39/969 308/28564
GO:0035456 GO:0035456 response to interferon-beta    17/969  71/28564
GO:0097529 GO:0097529 myeloid leukocyte migration    31/969 242/28564
                 pvalue     p.adjust       qvalue
GO:0050900 2.163956e-15 1.096260e-11 8.402981e-12
GO:0030595 1.066463e-13 2.701351e-10 2.070622e-10
GO:0071674 1.143223e-12 1.930522e-09 1.479771e-09
GO:0060326 1.728979e-12 2.189752e-09 1.678475e-09
GO:0035456 1.681085e-10 1.703276e-07 1.305584e-07
GO:0097529 2.447325e-10 2.066358e-07 1.583892e-07
                                                                                                                                                                                                                                                                                       geneID
GO:0050900 Tnfsf18/Sell/Slamf9/Fut7/Itga4/Mdk/Grem1/Ada/Prex1/Edn3/P2ry12/Il12a/S100a8/S100a9/Nbl1/Padi2/Bst1/Cxcl5/Ppbp/Pf4/Cxcl1/Ptn/Alox5/Trpm4/Hsd3b7/Itgam/Adam8/Calr/Ccl17/Enpp1/Aire/Ccl2/Ccl7/Ccl5/Ccl6/Ccr7/Aoc3/Itgb3/Ccl28/Lgals3/Ptk2b/Emp2/Apod/Retnlg/Plg/Dusp1/Ager/Il33/Ch25h
GO:0030595                                                                           Tnfsf18/Sell/Slamf9/Mdk/Grem1/Prex1/Edn3/Il12a/S100a8/S100a9/Nbl1/Padi2/Bst1/Cxcl5/Ppbp/Pf4/Cxcl1/Ptn/Alox5/Trpm4/Hsd3b7/Itgam/Adam8/Calr/Ccl17/Ccl2/Ccl7/Ccl5/Ccl6/Ccr7/Lgals3/Ptk2b/Retnlg/Dusp1/Ch25h
GO:0071674                                                                                                         Tnfsf18/Slamf9/Fut7/Itga4/Mdk/Grem1/Il12a/Nbl1/Padi2/Alox5/Trpm4/Hsd3b7/Adam8/Calr/Ccl17/Aire/Ccl2/Ccl7/Ccl5/Ccl6/Ccr7/Itgb3/Lgals3/Ptk2b/Apod/Retnlg/Plg/Dusp1/Ager/Ch25h
GO:0060326                                                  Tnfsf18/Sell/Slamf9/Mdk/Grem1/Prex1/Edn3/Il12a/S100a8/S100a9/Lpar1/Nbl1/Padi2/Bst1/Cxcl5/Ppbp/Pf4/Cxcl1/Ptn/Alox5/Trpm4/Hsd3b7/Itgam/Adam8/Calr/Ccl17/Ccl2/Ccl7/Ccl5/Ccl6/Ccr7/Ccl28/Lgals3/Ptk2b/Nr4a1/Retnlg/Dusp1/Ch25h/Plxnb3
GO:0035456                                                                                                                                                                      Aim2/Ifi204/Gbp6/Oas1c/Ifitm6/Bst2/Irgm1/Tgtp1/Tgtp2/Ifi47/Igtp/Irgm2/Ifitm7/Gm4951/F830016B08Rik/Iigp1/Ifit1
GO:0097529                                                                                                    Tnfsf18/Sell/Fut7/Mdk/Grem1/Prex1/Edn3/P2ry12/S100a8/S100a9/Nbl1/Bst1/Cxcl5/Ppbp/Pf4/Cxcl1/Itgam/Adam8/Ccl17/Enpp1/Ccl2/Ccl7/Ccl5/Ccl6/Ccr7/Lgals3/Ptk2b/Emp2/Retnlg/Dusp1/Ager
           Count
GO:0050900    49
GO:0030595    35
GO:0071674    30
GO:0060326    39
GO:0035456    17
GO:0097529    31

Now enrichGO() went through! The returned object resTimeGO is in a special format which looks like a table but actually is not! To get rid of the confusion, in the code it is converted to a real data frame resTimeGOTable.

In the output data frame, there are the following columns:

  • ID: ID of the gene set. In this example analysis, it is the GO ID.
  • Description: Readable description. Here it is the name of the GO term.
  • GeneRatio: Number of DE genes in the gene set / total number of DE genes.
  • BgRatio: Size of the gene set / total number of genes.
  • pvalue: p-value calculated from the hypergeometric distribution.
  • p.adjust: Adjusted p-value by the BH method.
  • qvalue: q-value which is another way for controlling false positives in multiple testings.
  • geneID: A list of DE genes in the gene set.
  • Count: Number of DE genes in the gene set.

You may have found the total number of DE genes changes. There are 1134 in timeDEgenes, but only 983 DE genes are included in the enrichment result table (in the GeneRatio column). The main reason is by default DE genes not annotated to any GO gene set are filtered out. This relates to the “universe” of all genes in ORA, which we will touch in the end of this section.

There are several additional arguments in enrichGO():

  • universe: the universe set of genes, i.e. total genes to use. By default it uses the union of the genes in all gene sets. If it is set, DE genes and all gene sets will take intersections with it. We will discuss it in the end of this section.
  • minGSSize: Minimal size of gene sets. Normally gene sets with very small size have very specific biological meanings which are not helpful too much for the interpretation. Gene sets with size smaller than it will be removed from the analysis. By default it is 10.
  • maxGSSize: Maximal size of gene sets. Normally gene sets with huge size provide too general biological meanings and are not helpful either. By default is 500.
  • pvalueCutoff: Cutoff for both p-values and adjusted p-values. By default is 0.05.
  • qvalueCutoff: Cutoff for q-values. by default is 0.2.

Note, enrichGO() only returns significant gene sets that pass the cutoffs4. This function design might not be proper because a function should return all the results no matter they are significant or not. Later users may need to use the complete enrichment table for downstream anlaysis. Second, the meaning of pvalueCutoff is not precise and there is redundancy between pvalueCutoff and qvalueCutoff (adjusted p-values and q-values are always non-smaller than raw p-values). Thus it is suggested to set both pvalueCutoff and qvalueCutoff to 1 in enrichGO().

R

resTimeGO = enrichGO(gene = timeDEgenes, 
                     keyType = "SYMBOL",
                     ont = "BP", 
                     OrgDb = org.Mm.eg.db,
                     pvalueCutoff = 1,
                     qvalueCutoff = 1)
resTimeGOTable = as.data.frame(resTimeGO)
head(resTimeGOTable)

OUTPUT

                   ID                 Description GeneRatio   BgRatio
GO:0050900 GO:0050900         leukocyte migration    49/969 386/28564
GO:0030595 GO:0030595        leukocyte chemotaxis    35/969 230/28564
GO:0071674 GO:0071674  mononuclear cell migration    30/969 185/28564
GO:0060326 GO:0060326             cell chemotaxis    39/969 308/28564
GO:0035456 GO:0035456 response to interferon-beta    17/969  71/28564
GO:0097529 GO:0097529 myeloid leukocyte migration    31/969 242/28564
                 pvalue     p.adjust       qvalue
GO:0050900 2.163956e-15 1.096260e-11 8.402981e-12
GO:0030595 1.066463e-13 2.701351e-10 2.070622e-10
GO:0071674 1.143223e-12 1.930522e-09 1.479771e-09
GO:0060326 1.728979e-12 2.189752e-09 1.678475e-09
GO:0035456 1.681085e-10 1.703276e-07 1.305584e-07
GO:0097529 2.447325e-10 2.066358e-07 1.583892e-07
                                                                                                                                                                                                                                                                                       geneID
GO:0050900 Tnfsf18/Sell/Slamf9/Fut7/Itga4/Mdk/Grem1/Ada/Prex1/Edn3/P2ry12/Il12a/S100a8/S100a9/Nbl1/Padi2/Bst1/Cxcl5/Ppbp/Pf4/Cxcl1/Ptn/Alox5/Trpm4/Hsd3b7/Itgam/Adam8/Calr/Ccl17/Enpp1/Aire/Ccl2/Ccl7/Ccl5/Ccl6/Ccr7/Aoc3/Itgb3/Ccl28/Lgals3/Ptk2b/Emp2/Apod/Retnlg/Plg/Dusp1/Ager/Il33/Ch25h
GO:0030595                                                                           Tnfsf18/Sell/Slamf9/Mdk/Grem1/Prex1/Edn3/Il12a/S100a8/S100a9/Nbl1/Padi2/Bst1/Cxcl5/Ppbp/Pf4/Cxcl1/Ptn/Alox5/Trpm4/Hsd3b7/Itgam/Adam8/Calr/Ccl17/Ccl2/Ccl7/Ccl5/Ccl6/Ccr7/Lgals3/Ptk2b/Retnlg/Dusp1/Ch25h
GO:0071674                                                                                                         Tnfsf18/Slamf9/Fut7/Itga4/Mdk/Grem1/Il12a/Nbl1/Padi2/Alox5/Trpm4/Hsd3b7/Adam8/Calr/Ccl17/Aire/Ccl2/Ccl7/Ccl5/Ccl6/Ccr7/Itgb3/Lgals3/Ptk2b/Apod/Retnlg/Plg/Dusp1/Ager/Ch25h
GO:0060326                                                  Tnfsf18/Sell/Slamf9/Mdk/Grem1/Prex1/Edn3/Il12a/S100a8/S100a9/Lpar1/Nbl1/Padi2/Bst1/Cxcl5/Ppbp/Pf4/Cxcl1/Ptn/Alox5/Trpm4/Hsd3b7/Itgam/Adam8/Calr/Ccl17/Ccl2/Ccl7/Ccl5/Ccl6/Ccr7/Ccl28/Lgals3/Ptk2b/Nr4a1/Retnlg/Dusp1/Ch25h/Plxnb3
GO:0035456                                                                                                                                                                      Aim2/Ifi204/Gbp6/Oas1c/Ifitm6/Bst2/Irgm1/Tgtp1/Tgtp2/Ifi47/Igtp/Irgm2/Ifitm7/Gm4951/F830016B08Rik/Iigp1/Ifit1
GO:0097529                                                                                                    Tnfsf18/Sell/Fut7/Mdk/Grem1/Prex1/Edn3/P2ry12/S100a8/S100a9/Nbl1/Bst1/Cxcl5/Ppbp/Pf4/Cxcl1/Itgam/Adam8/Ccl17/Enpp1/Ccl2/Ccl7/Ccl5/Ccl6/Ccr7/Lgals3/Ptk2b/Emp2/Retnlg/Dusp1/Ager
           Count
GO:0050900    49
GO:0030595    35
GO:0071674    30
GO:0060326    39
GO:0035456    17
GO:0097529    31

Perform GO enrichment on other organisms

Gene sets are provided as an OrgDb object in enrichGO(), thus you can perform ORA analysis on any organism as long as there is a corresponding OrgDb object.

  • For model organisms, the OrgDb object can be obtained from the corresponding org.*.db package.
  • For other organisms, the OrgDb object can be found with the AnnotationHub package.

KEGG pathway enrichment

To perform KEGG pathway enrichment analysis, there is also a function enrichKEGG() for that. Unfortunately, it cannot perform gene ID conversion automatically. Thus, if the ID type is not Entrez ID, we have to convert it by hand.

Note if you have also set universe, it should be converted to Entrez IDs as well.

We use the mapIds() function to convert genes from symbols to Entrez IDs. Since the ID mapping is not always one-to-one. We only take the first one if there are multiple hits by setting multiVals = "first"(but of course you can choose other options for multiVals, check the documentation). We also remove genes with no mapping available (with NA after the mapping)5.

R

EntrezIDs = mapIds(org.Mm.eg.db, keys = timeDEgenes, 
                   keytype = "SYMBOL", column = "ENTREZID", multiVals = "first")

OUTPUT

'select()' returned 1:1 mapping between keys and columns

R

EntrezIDs = EntrezIDs[!is.na(EntrezIDs)]
head(EntrezIDs)

OUTPUT

    Sgk3    Kcnb2   Sbspon    Gsta3   Lman2l  Ankrd39 
"170755"  "98741" "226866"  "14859" "214895" "109346" 

We have to set the KEGG organism code if it is not human. Similarly it is suggested to set pvalueCutoff and qvalueCutoff both to 1 and convert the result to a data frame.

R

resTimeKEGG = enrichKEGG(gene = EntrezIDs, 
                         organism = "mmu",
                         pvalueCutoff = 1,
                         qvalueCutoff = 1)
resTimeKEGGTable = as.data.frame(resTimeKEGG)
head(resTimeKEGGTable)

OUTPUT

               ID
mmu00590 mmu00590
mmu00565 mmu00565
mmu00592 mmu00592
mmu00591 mmu00591
mmu04913 mmu04913
mmu04061 mmu04061
                                                                                        Description
mmu00590                                   Arachidonic acid metabolism - Mus musculus (house mouse)
mmu00565                                        Ether lipid metabolism - Mus musculus (house mouse)
mmu00592                               alpha-Linolenic acid metabolism - Mus musculus (house mouse)
mmu00591                                      Linoleic acid metabolism - Mus musculus (house mouse)
mmu04913                                       Ovarian steroidogenesis - Mus musculus (house mouse)
mmu04061 Viral protein interaction with cytokine and cytokine receptor - Mus musculus (house mouse)
         GeneRatio BgRatio       pvalue     p.adjust       qvalue
mmu00590    16/454 85/9465 2.224011e-06 0.0006827712 0.0005946302
mmu00565    11/454 48/9465 1.234403e-05 0.0014156451 0.0012328951
mmu00592     8/454 25/9465 1.383367e-05 0.0014156451 0.0012328951
mmu00591    11/454 50/9465 1.870748e-05 0.0014357990 0.0012504473
mmu04913    12/454 63/9465 3.669398e-05 0.0022530101 0.0019621620
mmu04061    14/454 95/9465 1.606325e-04 0.0072719317 0.0063331756
                                                                                                       geneID
mmu00590 18783/19215/211429/329502/78390/19223/67103/242546/13118/18781/18784/11689/232889/15446/237625/11687
mmu00565                               18783/211429/329502/78390/22239/18781/18784/232889/320981/237625/53897
mmu00592                                                  18783/211429/329502/78390/18781/18784/232889/237625
mmu00591                               18783/211429/329502/78390/242546/18781/18784/13113/232889/237625/11687
mmu04913                          18783/211429/329502/78390/242546/11689/232889/13076/13070/15485/13078/16867
mmu04061                  16174/20311/57349/56744/14825/20295/20296/20306/20304/20305/12775/56838/16185/16186
         Count
mmu00590    16
mmu00565    11
mmu00592     8
mmu00591    11
mmu04913    12
mmu04061    14

Perform KEGG pathway enrichment on other organisms

Extending ORA to other organisms is rather simple.

  1. Make sure the DE genes are Entrez IDs.
  2. Choose the corresponding KEGG organism code.

MSigDB enrichment

For MSigDB gene sets, there is no pre-defined enrichment function. We need to directly use the low-level enrichment function enricher() which accepts self-defined gene sets. The gene sets should be in a format of a two-column data frame of genes and gene sets (or a class that can be converted to a data frame).

R

library(msigdbr)
gene_sets = msigdbr(category = "H", species = "mouse")
head(gene_sets)

OUTPUT

# A tibble: 6 × 18
  gs_cat gs_subcat gs_name               gene_symbol entrez_gene ensembl_gene   
  <chr>  <chr>     <chr>                 <chr>             <int> <chr>          
1 H      ""        HALLMARK_ADIPOGENESIS Abca1             11303 ENSMUSG0000001…
2 H      ""        HALLMARK_ADIPOGENESIS Abcb8             74610 ENSMUSG0000002…
3 H      ""        HALLMARK_ADIPOGENESIS Acaa2             52538 ENSMUSG0000003…
4 H      ""        HALLMARK_ADIPOGENESIS Acadl             11363 ENSMUSG0000002…
5 H      ""        HALLMARK_ADIPOGENESIS Acadm             11364 ENSMUSG0000006…
6 H      ""        HALLMARK_ADIPOGENESIS Acads             11409 ENSMUSG0000002…
# ℹ 12 more variables: human_gene_symbol <chr>, human_entrez_gene <int>,
#   human_ensembl_gene <chr>, gs_id <chr>, gs_pmid <chr>, gs_geoid <chr>,
#   gs_exact_source <chr>, gs_url <chr>, gs_description <chr>, taxon_id <int>,
#   ortholog_sources <chr>, num_ortholog_sources <dbl>

As mentioned before, it is important the gene ID type in the gene sets should be the same as in the DE genes, so here we choose the "gene_symbol" column.

R

resTimeHallmark = enricher(gene = timeDEgenes, 
                           TERM2GENE = gene_sets[, c("gs_name", "gene_symbol")],
                           pvalueCutoff = 1,
                           qvalueCutoff = 1)
resTimeHallmarkTable = as.data.frame(resTimeHallmark)
head(resTimeHallmarkTable)

OUTPUT

                                                             ID
HALLMARK_MYOGENESIS                         HALLMARK_MYOGENESIS
HALLMARK_COMPLEMENT                         HALLMARK_COMPLEMENT
HALLMARK_COAGULATION                       HALLMARK_COAGULATION
HALLMARK_ALLOGRAFT_REJECTION       HALLMARK_ALLOGRAFT_REJECTION
HALLMARK_ESTROGEN_RESPONSE_LATE HALLMARK_ESTROGEN_RESPONSE_LATE
HALLMARK_INFLAMMATORY_RESPONSE   HALLMARK_INFLAMMATORY_RESPONSE
                                                    Description GeneRatio
HALLMARK_MYOGENESIS                         HALLMARK_MYOGENESIS    31/291
HALLMARK_COMPLEMENT                         HALLMARK_COMPLEMENT    26/291
HALLMARK_COAGULATION                       HALLMARK_COAGULATION    20/291
HALLMARK_ALLOGRAFT_REJECTION       HALLMARK_ALLOGRAFT_REJECTION    23/291
HALLMARK_ESTROGEN_RESPONSE_LATE HALLMARK_ESTROGEN_RESPONSE_LATE    23/291
HALLMARK_INFLAMMATORY_RESPONSE   HALLMARK_INFLAMMATORY_RESPONSE    22/291
                                 BgRatio       pvalue     p.adjust       qvalue
HALLMARK_MYOGENESIS             201/4394 5.710171e-06 0.0002740882 0.0002284068
HALLMARK_COMPLEMENT             196/4394 4.300844e-04 0.0103220246 0.0086016872
HALLMARK_COAGULATION            139/4394 7.088433e-04 0.0113414921 0.0094512434
HALLMARK_ALLOGRAFT_REJECTION    201/4394 6.397659e-03 0.0767719033 0.0639765861
HALLMARK_ESTROGEN_RESPONSE_LATE 206/4394 8.584183e-03 0.0824081536 0.0686734614
HALLMARK_INFLAMMATORY_RESPONSE  201/4394 1.256794e-02 0.0960740794 0.0800617329
                                                                                                                                                                                                                geneID
HALLMARK_MYOGENESIS             Myl1/Casq1/Aplnr/Tnnc2/Ptgis/Gja5/Col15a1/Cav3/Tnnt1/Ryr1/Cox6a2/Tnni2/Lsp1/Nqo1/Hspb2/Cryab/Erbb3/Stc2/Gpx3/Sparc/Myh3/Myh1/Col1a1/Cacng1/Itgb4/Bdkrb2/Mapk12/Apod/Spdef/Cdkn1a/Actn3
HALLMARK_COMPLEMENT                                                Serpinb2/Pla2g4a/Cd46/Hspa5/Cp/Gnb4/S100a9/Cda/Cxcl1/Apoc1/Itgam/Irf7/Klkb1/Mmp15/Mmp8/Ccl5/Lgals3/Gzmb/Tmprss6/Maff/Plg/Psmb9/Hspa1a/C3/Dusp5/Phex
HALLMARK_COAGULATION                                                                                         Serpinb2/Ctse/C8g/Ctsk/Masp2/Pf4/Sh2b2/Vwf/Apoc1/Klkb1/Mmp15/Mmp8/Trf/Sparc/Itgb3/Dct/Tmprss6/Maff/Plg/C3
HALLMARK_ALLOGRAFT_REJECTION                                                           Il18rap/Cd247/Il12a/Pf4/Capg/Ccnd2/Igsf6/Irf7/Il12rb1/Icosl/Itk/Nos2/Ccl2/Ccl7/Ccl5/Gpr65/Gzmb/Il2rb/Tap1/Tap2/H2-T23/Cfp/Il2rg
HALLMARK_ESTROGEN_RESPONSE_LATE                                                    Clic3/Ass1/Mdk/S100a9/Cdc20/Nbl1/Cav1/Idh2/Th/Pdlim3/Ascl1/Xbp1/Top2a/Dcxr/Fos/Serpina3n/Dhrs2/Tst/Emp2/Fkbp5/Mapk13/Cyp4f15/Kif20a
HALLMARK_INFLAMMATORY_RESPONSE                                                              Il18rap/Sell/Aplnr/Abca1/Lpar1/Cxcl5/Adm/Irf7/Msr1/Bst2/Ccl17/Icosl/Ccl2/Ccl7/Ccl5/Ccr7/Itgb3/Has2/Il2rb/Cdkn1a/Cd70/Best1
                                Count
HALLMARK_MYOGENESIS                31
HALLMARK_COMPLEMENT                26
HALLMARK_COAGULATION               20
HALLMARK_ALLOGRAFT_REJECTION       23
HALLMARK_ESTROGEN_RESPONSE_LATE    23
HALLMARK_INFLAMMATORY_RESPONSE     22

Further reading

Implementing ORA is rather simple. The following function ora() performs ORA on a list of gene sets. Try to read and understand the code.

R

ora = function(genes, gene_sets, universe = NULL) {
    if(is.null(universe)) {
        universe = unique(unlist(gene_sets))
    } else {
        universe = unique(universe)
    }

    # make sure genes are unique
    genes = intersect(genes, universe)
    gene_sets = lapply(gene_sets, intersect, universe)

    # calculate different numbers
    n_11 = sapply(gene_sets, function(x) length(intersect(genes, x)))
    n_10 = length(genes)
    n_01 = sapply(gene_sets, length)
    n = length(universe)

    # calculate p-values
    p = 1 - phyper(n_11 - 1, n_10, n - n_10, n_01)

    df = data.frame(
        gene_set = names(gene_sets),
        hits = n_11,
        n_genes = n_10,
        gene_set_size = n_01,
        n_total = n,
        p_value = p,
        p_adjust = p.adjust(p, "BH")
    )
}

Test on the MSigDB hallmark gene sets:

R

HallmarkGeneSets = split(gene_sets$gene_symbol, gene_sets$gs_name)
df = ora(timeDEgenes, HallmarkGeneSets, rownames(se))
head(df)

OUTPUT

                                                 gene_set hits n_genes
HALLMARK_ADIPOGENESIS               HALLMARK_ADIPOGENESIS    9    1134
HALLMARK_ALLOGRAFT_REJECTION HALLMARK_ALLOGRAFT_REJECTION   23    1134
HALLMARK_ANDROGEN_RESPONSE     HALLMARK_ANDROGEN_RESPONSE    6    1134
HALLMARK_ANGIOGENESIS               HALLMARK_ANGIOGENESIS    4    1134
HALLMARK_APICAL_JUNCTION         HALLMARK_APICAL_JUNCTION   14    1134
HALLMARK_APICAL_SURFACE           HALLMARK_APICAL_SURFACE    3    1134
                             gene_set_size n_total     p_value    p_adjust
HALLMARK_ADIPOGENESIS                  185   21198 0.662392856 0.871569547
HALLMARK_ALLOGRAFT_REJECTION           191   21198 0.000234807 0.002935087
HALLMARK_ANDROGEN_RESPONSE             107   21198 0.512665196 0.753919406
HALLMARK_ANGIOGENESIS                   36   21198 0.124351058 0.260206520
HALLMARK_APICAL_JUNCTION               186   21198 0.124899130 0.260206520
HALLMARK_APICAL_SURFACE                 41   21198 0.376915129 0.588929889

Choose a proper universe

Finally, it is time to talk about the “universe” of ORA analysis which is normally ignored in many analyses. In current tools, there are mainly following different universe settings:

  1. Using all genes in the genome, this also includes non-protein coding genes. For human, the size of universe is 60k ~ 70k.
  2. Using all protein-coding genes. For human, the size of universe is ~ 20k.
  3. In the era of microarray, total genes that are measured on the chip is taken as the universe. For RNASeq, since reads are aligned to all genes, we can set a cutoff and only use those “expressed” genes as the universe.
  4. Using all genes in a gene sets collection. Then the size of the universe depends on the size of the gene sets collection. For example GO gene sets collection is much larger than the KEGG pathway gene sets collection.

If the universe is set, DE genes as well as genes in the gene sets are first intersected to the universe. However, in general the universe affects three values of \(n_{22}\), \(n_{02}\) and \(n_{20}\) more, which correspond to the non-DE genes or non-gene-set genes.

In the gene set Not in the gene set Total
DE \(n_{11}\) \(n_{12}\) \(n_{1+}\)
Not DE \(n_{21}\) \(\color{red}{n_{22}}\) \(\color{red}{n_{2+}}\)
Total \(n_{+1}\) \(\color{red}{n_{+2}}\) \(\color{red}{n}\)

In the contingency table, we are testing the dependency of whether genes being DE and whether genes being in the gene set. In the model, each gene has a definite attribute of being either DE or non-DE and each gene has a second definite attribute of either belonging to the gene set or not. If a larger universe is used, such as total genes where there are genes not measured nor will never annotated to gene sets (let’s call them non-informative genes, e.g. non-protein coding genes or not-expressed genes), all the non-informative genes are implicitly assigned with an attribute of being non-DE or not in the gene set. This implicit assignment is not proper because these genes provide no information and they should not be included in the analysis. Adding them to the analysis increases \(n_{22}\), \(n_{02}\) or \(n_{20}\), makes the observation \(n_{11}\) getting further away from the null distribution, eventually generates a smaller p-value. For the similar reason, small universes tend to generate large p-values.

In enrichGO()/enrichKEGG()/enricher(), universe genes can be set via the universe argument. By default the universe is the total genes in the gene sets collection. When a self-defined universe is provided, this might be different from what you may think, the universe is the intersection of user-provided universe and total genes in the gene set collection. Thus the universe setting in clusterProfiler is very conservative.

Check the more discusstions at https://twitter.com/mdziemann/status/1626407797939384320.

We can do a simple experiment on the small MSigDB hallmark gene sets. We use the ora() function which we have implemented in previous “Further reading” section and we compare three different universe settings.

R

# all genes in the gene sets collection ~ 4k genes
df1 = ora(timeDEgenes, HallmarkGeneSets)
# all protein-coding genes, ~ 20k genes
df2 = ora(timeDEgenes, HallmarkGeneSets, rownames(se))
# all genes in org.Mm.eg.db ~ 70k genes
df3 = ora(timeDEgenes, HallmarkGeneSets, 
    keys(org.Mm.eg.db, keytype = "SYMBOL"))

# df1, df2, and df3 are in the same row order, 
# so we can directly compare them
plot(df1$p_value, df2$p_value, col = 2, pch = 16, 
    xlim = c(0, 1), ylim = c(0, 1), 
    xlab = "all hallmark genes as universe (p-values)", ylab = "p-values",
    main = "compare universes")
points(df1$p_value, df3$p_value, col = 4, pch = 16)
abline(a = 0, b = 1, lty = 2)
legend("topleft", legend = c("all protein-coding genes as universe", "all genes as universe"), 
    pch = 16, col = c(2, 4))

It is very straightforward to see, with a larger universe, there are more significant gene sets, which may produce potentially more false positives. This is definitely worse when using all genes in the genome as universe.

Based on the discussion in this section, the recommendation of using universe is:

  1. using protein-coding genes,
  2. using measured genes,
  3. or using a conservative way with clusterProfiler.

Visualization


clusterProfiler provides a rich set of visualization methods on the GSEA results, from simple visualization to complex ones. Complex visualizations are normally visually fancy but do not transfer too much useful information, and they should only be applied in very specific scenarios under very specific settings; while simple graphs normally do better jobs. Recently the visualization code in clusterProfiler has been moved to a new package enrichplot. Let’s first load the enrichplot package. The full sets of visualizations that enrichplot supports can be found from https://yulab-smu.top/biomedical-knowledge-mining-book/enrichplot.html.

We first re-generate the enrichment table.

R

library(enrichplot)
resTimeGO = enrichGO(gene = timeDEgenes, 
                     keyType = "SYMBOL",
                     ont = "BP", 
                     OrgDb = org.Mm.eg.db,
                     pvalueCutoff = 1,
                     qvalueCutoff = 1)
resTimeGOTable = as.data.frame(resTimeGO)
head(resTimeGOTable)

OUTPUT

                   ID                 Description GeneRatio   BgRatio
GO:0050900 GO:0050900         leukocyte migration    49/969 386/28564
GO:0030595 GO:0030595        leukocyte chemotaxis    35/969 230/28564
GO:0071674 GO:0071674  mononuclear cell migration    30/969 185/28564
GO:0060326 GO:0060326             cell chemotaxis    39/969 308/28564
GO:0035456 GO:0035456 response to interferon-beta    17/969  71/28564
GO:0097529 GO:0097529 myeloid leukocyte migration    31/969 242/28564
                 pvalue     p.adjust       qvalue
GO:0050900 2.163956e-15 1.096260e-11 8.402981e-12
GO:0030595 1.066463e-13 2.701351e-10 2.070622e-10
GO:0071674 1.143223e-12 1.930522e-09 1.479771e-09
GO:0060326 1.728979e-12 2.189752e-09 1.678475e-09
GO:0035456 1.681085e-10 1.703276e-07 1.305584e-07
GO:0097529 2.447325e-10 2.066358e-07 1.583892e-07
                                                                                                                                                                                                                                                                                       geneID
GO:0050900 Tnfsf18/Sell/Slamf9/Fut7/Itga4/Mdk/Grem1/Ada/Prex1/Edn3/P2ry12/Il12a/S100a8/S100a9/Nbl1/Padi2/Bst1/Cxcl5/Ppbp/Pf4/Cxcl1/Ptn/Alox5/Trpm4/Hsd3b7/Itgam/Adam8/Calr/Ccl17/Enpp1/Aire/Ccl2/Ccl7/Ccl5/Ccl6/Ccr7/Aoc3/Itgb3/Ccl28/Lgals3/Ptk2b/Emp2/Apod/Retnlg/Plg/Dusp1/Ager/Il33/Ch25h
GO:0030595                                                                           Tnfsf18/Sell/Slamf9/Mdk/Grem1/Prex1/Edn3/Il12a/S100a8/S100a9/Nbl1/Padi2/Bst1/Cxcl5/Ppbp/Pf4/Cxcl1/Ptn/Alox5/Trpm4/Hsd3b7/Itgam/Adam8/Calr/Ccl17/Ccl2/Ccl7/Ccl5/Ccl6/Ccr7/Lgals3/Ptk2b/Retnlg/Dusp1/Ch25h
GO:0071674                                                                                                         Tnfsf18/Slamf9/Fut7/Itga4/Mdk/Grem1/Il12a/Nbl1/Padi2/Alox5/Trpm4/Hsd3b7/Adam8/Calr/Ccl17/Aire/Ccl2/Ccl7/Ccl5/Ccl6/Ccr7/Itgb3/Lgals3/Ptk2b/Apod/Retnlg/Plg/Dusp1/Ager/Ch25h
GO:0060326                                                  Tnfsf18/Sell/Slamf9/Mdk/Grem1/Prex1/Edn3/Il12a/S100a8/S100a9/Lpar1/Nbl1/Padi2/Bst1/Cxcl5/Ppbp/Pf4/Cxcl1/Ptn/Alox5/Trpm4/Hsd3b7/Itgam/Adam8/Calr/Ccl17/Ccl2/Ccl7/Ccl5/Ccl6/Ccr7/Ccl28/Lgals3/Ptk2b/Nr4a1/Retnlg/Dusp1/Ch25h/Plxnb3
GO:0035456                                                                                                                                                                      Aim2/Ifi204/Gbp6/Oas1c/Ifitm6/Bst2/Irgm1/Tgtp1/Tgtp2/Ifi47/Igtp/Irgm2/Ifitm7/Gm4951/F830016B08Rik/Iigp1/Ifit1
GO:0097529                                                                                                    Tnfsf18/Sell/Fut7/Mdk/Grem1/Prex1/Edn3/P2ry12/S100a8/S100a9/Nbl1/Bst1/Cxcl5/Ppbp/Pf4/Cxcl1/Itgam/Adam8/Ccl17/Enpp1/Ccl2/Ccl7/Ccl5/Ccl6/Ccr7/Lgals3/Ptk2b/Emp2/Retnlg/Dusp1/Ager
           Count
GO:0050900    49
GO:0030595    35
GO:0071674    30
GO:0060326    39
GO:0035456    17
GO:0097529    31

barplot() and dotplot() generate plots for a small number of significant gene sets. Note the two functions are directly applied on resTimeGO returned by enrichGO().

R

barplot(resTimeGO, showCategory = 20)

R

dotplot(resTimeGO, showCategory = 20)

Barplots can map two variables to the plot, one to the height of bars and the other to the colors of bars; while for dotplot, sizes of dots can be mapped to a third variable. The variable names are in the colum names of the result table. Both plots include the top 20 most significant terms. On dotplot, terms are ordered by the values on x-axis (the GeneRatio).

Now we need to talk about “what is a good visualization?”. The essential two questions are “what is the key message a plot transfers to readers?” and “what is the major graphical element in the plot?”. In the barplot or dotplot, the major graphical element which readers may notice the easiest is the height of bars or the offset of dots to the origin. The most important message of the ORA analysis is of course “the enrichment”. The two examples from barplot() and dotplot() actually fail to transfer such information to readers. In the first barplot where "Count" is used as values on x-axis, the numer of DE genes in gene sets is not a good measure of the enrichment because it has a positive relation to the size of gene sets. A high value of "Count" does not mean the gene set is more enriched.

It is the same reason for dotplot where "GeneRatio" is used as values on x-axis. Gene ratio is calculated as the fraction of DE genes from a certain gene set (GeneRatio = Count/Total_DE_Genes). The dotplot puts multiple gene sets in the same plot and the aim is to compare between gene sets, thus gene sets should be “scaled” to make them comparable. "GeneRatio" is not scaled for different gene sets and it still has a positive relation to the gene set size, which can be observed in the dotplot where higher the gene ratio, larger the dot size. Actually “GeneRatio” has the same effect as “Count” (GeneRatio = Count/Total_DE_Genes), so as has been explained in the previous paragraph, "GeneRatio" is not a good measure for enrichment either.

Now let’s try to make a more reasonable barplot and dotplot to show the enrichment of ORA.

First, let’s define some metrics which measure the “enrichment” of DE genes on gene sets. Recall the denotations in the 2x2 contingency table (we are too far from that!). Let’s take these numbers from the enrichment table.

R

n_11 = resTimeGOTable$Count
n_10 = 983  # length(intersect(resTimeGO@gene, resTimeGO@universe))
n_01 = as.numeric(gsub("/.*$", "", resTimeGOTable$BgRatio))
n = 28943  # length(resTimeGO@universe)

Instead of using GeneRatio, we use the fraction of DE genes in the gene sets which are kind of like a “scaled” value for all gene sets. Let’s calculate it:

R

resTimeGOTable$DE_Ratio = n_11/n_01
resTimeGOTable$GS_size = n_01  # size of gene sets

Then intuitively, if a gene set has a higher DE_Ratio value, we could say DE genes have a higher enrichment6 in it.

We can measure the enrichment in two other ways. First, the log2 fold enrichment, defined as:

\[ \log_2(\mathrm{Fold\_enrichment}) = \frac{n_{11}/n_{10}}{n_{01}/n} = \frac{n_{11}/n_{01}}{n_{10}/n} = \frac{n_{11}n}{n_{10}n_{01}} \]

which is the log2 of the ratio of DE% in the gene set and DE% in the universe or the log2 of the ratio of gene_set% in the DE genes and gene_set% in the universe. The two are identical.

R

resTimeGOTable$log2_Enrichment = log( (n_11/n_10)/(n_01/n) )

Second, it is also common to use z-score which is

\[ z = \frac{n_{11} - \mu}{\sigma} \]

where \(\mu\) and \(\sigma\) are the mean and standard deviation of the hypergeometric distribution. They can be calculated as:

R

hyper_mean = n_01*n_10/n

n_02 = n - n_01
n_20 = n - n_10
hyper_var = n_01*n_10/n * n_20*n_02/n/(n-1)
resTimeGOTable$zScore = (n_11 - hyper_mean)/sqrt(hyper_var)

We will use log2 fold change as the primary variable to map to bar heights and DE_Ratio as the secondary variable to map to colors. This can be done directly with the ggplot2 package. We also add the adjusted p-values as labels on the bars.

In resTimeGOTable, gene sets are already ordered by the significance, so we take the first 10 gene sets which are the 10 most significant gene sets.

R

library(ggplot2)
ggplot(resTimeGOTable[1:10, ], 
        aes(x = log2_Enrichment, y = factor(Description, levels = rev(Description)), 
            fill = DE_Ratio)) +
    geom_bar(stat = "identity") +
    geom_text(aes(x = log2_Enrichment, 
        label = sprintf("%.2e", p.adjust)), hjust = 1, col = "white") +
    ylab("")

In the next example, we use z-score as the primary variable to map to the offset to origin, DE_Ratio and Count to map to dot colors and sizes.

R

ggplot(resTimeGOTable[1:10, ], 
        aes(x = zScore, y = factor(Description, levels = rev(Description)), 
            col = DE_Ratio, size = Count)) +
    geom_point() +
    ylab("")

Both plots can highlight the gene set “leukocyte migration involved in inflammatory response” is relatively small but highly enriched.

Another useful visualization is the volcano plot. You may be aware of in differential expression analysis, in the volcano plot, x-axis corresponds to log2 fold changes of the differential expression, and y-axis corresponds to the adjusted p-values. It is actually similar here where we use log2 fold enrichment on x-axis.

Since we only look at the over-representation, the volcano plot is one-sided. We can set two cutoffs on the log2 fold enrichment and adjusted p-values, then the gene sets on the top right region can be thought as being both statistically significant and also biologically sensible.

R

ggplot(resTimeGOTable, 
    aes(x = log2_Enrichment, y = -log10(p.adjust), 
        color = DE_Ratio, size = GS_size)) +
    geom_point() +
    geom_hline(yintercept = -log10(0.01), lty = 2, col = "#444444") +
    geom_vline(xintercept = 1.5, lty = 2, col = "#444444")

In the “volcano plot”, we can observe the plot is composed by a list of curves. The trends are especially clear in the right bottom of the plot. Actually each “curve” corresponds to a same "Count" value (number of DE genes in a gene set). The volcano plot shows very clearly that the enrichment has a positive relation to the gene set size where a large gene set can easily reach a small p-value with a small DE_ratio and a small log2 fold enrichment, while a small gene set needs to have a large DE ratio to be significant.

It is also common that we perform ORA analysis on up-regulated genes and down-regulated separately. And we want to combine the significant gene sets from the two ORA analysis in one plot. In the following code, we first generate two enrichment tables for up-regulated genes and down-regulated separately.

R

# up-regulated genes
timeDEup <- as.data.frame(subset(resTime, padj < 0.05 & log2FoldChange > log2(1.5)))
timeDEupGenes <- rownames(timeDEup)

resTimeGOup = enrichGO(gene = timeDEupGenes, 
                       keyType = "SYMBOL",
                       ont = "BP", 
                       OrgDb = org.Mm.eg.db,
                       universe = rownames(se),
                       pvalueCutoff = 1,
                       qvalueCutoff = 1)
resTimeGOupTable = as.data.frame(resTimeGOup)
n_11 = resTimeGOupTable$Count
n_10 = length(intersect(resTimeGOup@gene, resTimeGOup@universe))
n_01 = as.numeric(gsub("/.*$", "", resTimeGOupTable$BgRatio))
n = length(resTimeGOup@universe)
resTimeGOupTable$log2_Enrichment = log( (n_11/n_10)/(n_01/n) )

# down-regulated genes
timeDEdown <- as.data.frame(subset(resTime, padj < 0.05 & log2FoldChange < -log2(1.5)))
timeDEdownGenes <- rownames(timeDEdown)

resTimeGOdown = enrichGO(gene = timeDEdownGenes, 
                       keyType = "SYMBOL",
                       ont = "BP", 
                       OrgDb = org.Mm.eg.db,
                       universe = rownames(se),
                       pvalueCutoff = 1,
                       qvalueCutoff = 1)
resTimeGOdownTable = as.data.frame(resTimeGOdown)
n_11 = resTimeGOdownTable$Count
n_10 = length(intersect(resTimeGOdown@gene, resTimeGOdown@universe))
n_01 = as.numeric(gsub("/.*$", "", resTimeGOdownTable$BgRatio))
n = length(resTimeGOdown@universe)
resTimeGOdownTable$log2_Enrichment = log( (n_11/n_10)/(n_01/n) )

As an example, let’s simply take the first 5 most significant terms for up-regulated genes and the first 5 most significant terms for down-regulated genes. The following ggplot2 code should be easy to read.

R

# The name of the 3rd term is too long, we wrap it into two lines.
resTimeGOupTable[3, "Description"] = paste(strwrap(resTimeGOupTable[3, "Description"]), collapse = "\n")

direction = c(rep("up", 5), rep("down", 5))
ggplot(rbind(resTimeGOupTable[1:5, ],
             resTimeGOdownTable[1:5, ]),
        aes(x = log2_Enrichment, y = factor(Description, levels = rev(Description)), 
            fill = direction)) +
    geom_bar(stat = "identity") +
    scale_fill_manual(values = c("up" = "red", "down" = "darkgreen")) +
    geom_text(aes(x = log2_Enrichment, 
        label = sprintf("%.2e", p.adjust)), hjust = 1, col = "white") +
    ylab("")

Specifically for GO enrichment, it is often that GO enrichment returns a long list of significant GO terms (e.g. several hundreds). This makes it difficult to summarize the common functions from the long list. The last package we will introduce is the simplifyEnrichment package which partitions GO terms into clusters based on their semantic similarity7 and summarizes their common functions via word clouds.

The input of the simplifyGO() function is a vector of GO IDs. It is recommended to have at least 100 GO IDs for summarization and visualization.

R

GO_ID = resTimeGOTable$ID[resTimeGOTable$p.adjust < 0.1]
library(simplifyEnrichment)
simplifyGO(GO_ID)

Further reading

ORA analysis actually applies a binary conversion on genes where genes pass the cutoff are set as 1 (DE gene) and others are set as 0 (non-DE gene). This binary transformation over-simplifies the problem and a lot of information are lost. There is second class of gene set enrichment analysis methods which takes the continuous gene-level score as input and weights the importance of a gene in the gene set. Please refer to Subramanian et. al. Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles, PNAS 2005 for more information.

Key Points

  • ORA analysis is based on the gene counts and it is based on Fisher’s exact test or the hypergeometric distribution.
  • In R, it is easy to obtain gene sets from a large number of sources.

  1. Genes must be unique in each vector.↩︎

  2. Also note phyper() can be vectorized.↩︎

  3. The definition is from Wikipedia: https://en.wikipedia.org/wiki/Biological_pathway.↩︎

  4. This is actually not true. Indeed as.data.frame(resTimeGO) only returns the significant GO terms, but the complete enrichment table is still stored in resTimeGO@result. However, directly retrieving the slot of an S4 object is highly unrecommended.↩︎

  5. You can also use select() function: select(org.Mm.eg.db, keys = timeDEgenes, keytype = "SYMBOL", column = "ENTREZID")↩︎

  6. If here the term “enrichment” does mean statistically.↩︎

  7. The semantic similarity between GO terms considers the topological relations in the GO hierarchy.↩︎