Cell type annotation

Last updated on 2024-09-08 | Edit this page

Estimated time: 45 minutes

Overview

Questions

  • How can we identify groups of cells with similar expression profiles?
  • How can we identify genes that drive separation between these groups of cells?
  • How to leverage reference datasets and known marker genes for the cell type annotation of new datasets?

Objectives

  • Identify groups of cells by clustering cells based on gene expression patterns.
  • Identify marker genes through testing for differential expression between clusters.
  • Annotate cell types through annotation transfer from reference datasets.
  • Annotate cell types through marker gene set enrichment testing.

Setup


Again we’ll start by loading the libraries we’ll be using:

R

library(AUCell)
library(MouseGastrulationData)
library(SingleR)
library(bluster)
library(scater)
library(scran)
library(pheatmap)
library(GSEABase)

Data retrieval


We’ll be using the fifth processed sample from the WT chimeric mouse embryo data:

R

sce <- WTChimeraData(samples = 5, type = "processed")
sce

OUTPUT

class: SingleCellExperiment
dim: 29453 2411
metadata(0):
assays(1): counts
rownames(29453): ENSMUSG00000051951 ENSMUSG00000089699 ...
  ENSMUSG00000095742 tomato-td
rowData names(2): ENSEMBL SYMBOL
colnames(2411): cell_9769 cell_9770 ... cell_12178 cell_12179
colData names(11): cell barcode ... doub.density sizeFactor
reducedDimNames(2): pca.corrected.E7.5 pca.corrected.E8.5
mainExpName: NULL
altExpNames(0):

To speed up the computations, we take a random subset of 1,000 cells.

R

set.seed(123)

ind <- sample(ncol(sce), 1000)

sce <- sce[,ind]

Preprocessing


The SCE object needs to contain log-normalized expression counts as well as PCA coordinates in the reduced dimensions, so we compute those here:

R

sce <- logNormCounts(sce)

sce <- runPCA(sce)

Clustering


Clustering is an unsupervised learning procedure that is used to empirically define groups of cells with similar expression profiles. Its primary purpose is to summarize complex scRNA-seq data into a digestible format for human interpretation. This allows us to describe population heterogeneity in terms of discrete labels that are easily understood, rather than attempting to comprehend the high-dimensional manifold on which the cells truly reside. After annotation based on marker genes, the clusters can be treated as proxies for more abstract biological concepts such as cell types or states.

Popularized by its use in Seurat, graph-based clustering is a flexible and scalable technique for clustering large scRNA-seq datasets. We first build a graph where each node is a cell that is connected to its nearest neighbors in the high-dimensional space. Edges are weighted based on the similarity between the cells involved, with higher weight given to cells that are more closely related. We then apply algorithms to identify “communities” of cells that are more connected to cells in the same community than they are to cells of different communities. Each community represents a cluster that we can use for downstream interpretation.

Here, we use the clusterCells() function from the scran package to perform graph-based clustering using the Louvain algorithm for community detection. All calculations are performed using the top PCs to take advantage of data compression and denoising. This function returns a vector containing cluster assignments for each cell in our SingleCellExperiment object. We use the colLabels() function to assign the cluster labels as a factor in the column data.

R

colLabels(sce) <- clusterCells(sce, use.dimred = "PCA",
                               BLUSPARAM = NNGraphParam(cluster.fun = "louvain"))

table(colLabels(sce))

OUTPUT


  1   2   3   4   5   6   7   8   9  10  11
100 160  99 141  63  93  60 108  44  91  41 

You can see we ended up with 11 clusters of varying sizes.

We can now overlay the cluster labels as color on a UMAP plot:

R

sce <- runUMAP(sce, dimred = "PCA")

plotReducedDim(sce, "UMAP", color_by = "label")

Challenge

Our clusters look semi-reasonable, but what if we wanted to make them less granular? Look at the help documentation for ?clusterCells and ?NNGraphParam to find out what we’d need to change to get fewer, larger clusters.

We see in the help documentation for ?clusterCells that all of the clustering algorithm details are handled through the BLUSPARAM argument, which needs to provide a BlusterParam object (of which NNGraphParam is a sub-class). Each type of clustering algorithm will have some sort of hyper-parameter that controls the granularity of the output clusters. Looking at ?NNGraphParam specifically, we see an argument called k which is described as “An integer scalar specifying the number of nearest neighbors to consider during graph construction.” If the clustering process has to connect larger sets of neighbors, the graph will tend to be cut into larger groups, resulting in less granular clusters. Try the two code blocks above once more with k = 20. Given their visual differences, do you think one set of clusters is “right” and the other is “wrong”?

Marker gene detection


To interpret clustering results as obtained in the previous section, we identify the genes that drive separation between clusters. These marker genes allow us to assign biological meaning to each cluster based on their functional annotation. In the simplest case, we have a priori knowledge of the marker genes associated with particular cell types, allowing us to treat the clustering as a proxy for cell type identity.

The most straightforward approach to marker gene detection involves testing for differential expression between clusters. If a gene is strongly DE between clusters, it is likely to have driven the separation of cells in the clustering algorithm.

Here, we use findMarkers() to perform a Wilcoxon rank sum test against a log2 fold change threshold of 1, focusing on up-regulated (positive) markers in one cluster when compared to another cluster.

R

rownames(sce) <- rowData(sce)$SYMBOL

markers <- findMarkers(sce, test.type = "wilcox", direction = "up", lfc = 1)

markers

OUTPUT

List of length 11
names(11): 1 2 3 4 5 6 7 8 9 10 11

The resulting object contains a sorted marker gene list for each cluster, in which the top genes are those that contribute the most to the separation of that cluster from mall other clusters.

Here, we inspect the ranked marker gene list for the first cluster.

R

markers[[1]]

OUTPUT

DataFrame with 29453 rows and 14 columns
                 Top     p.value         FDR summary.AUC     AUC.2     AUC.3
           <integer>   <numeric>   <numeric>   <numeric> <numeric> <numeric>
Crabp2             1 7.31206e-35 1.95784e-31    0.938625  0.938625 0.9159596
Ptn                1 2.26190e-43 6.66197e-39    0.983313  0.983313 0.9812121
Crabp1             1 1.13915e-32 2.09696e-29    0.926687  0.926687 0.7927273
Zeb2               2 3.98744e-10 1.38167e-07    0.801500  0.553125 0.3943434
Mest               2 1.10835e-24 9.60122e-22    0.883000  0.883000 0.0249495
...              ...         ...         ...         ...       ...       ...
AC125149.2     29448           1           1           0         0         0
AC125149.4     29449           1           1           0         0         0
AC234645.1     29450           1           1           0         0         0
AC168977.2     29451           1           1           0         0         0
Vmn2r122       29453           1           1           0         0         0
               AUC.4     AUC.5     AUC.6     AUC.7     AUC.8     AUC.9
           <numeric> <numeric> <numeric> <numeric> <numeric> <numeric>
Crabp2      0.861986  0.494444  0.690000  0.763833  0.845370  0.769091
Ptn         0.911773  0.771429  0.198710  0.830667  0.563333  0.898409
Crabp1      0.540071  0.714127  0.695591  0.767167  0.620556  0.524773
Zeb2        0.604184  0.390317  0.337849  0.801500  0.447963  0.341364
Mest        0.136028  0.476190  0.091828  0.386833  0.314444  0.773864
...              ...       ...       ...       ...       ...       ...
AC125149.2         0         0         0         0         0         0
AC125149.4         0         0         0         0         0         0
AC234645.1         0         0         0         0         0         0
AC168977.2         0         0         0         0         0         0
Vmn2r122           0         0         0         0         0         0
              AUC.10    AUC.11
           <numeric> <numeric>
Crabp2     0.9416484  0.882439
Ptn        0.9463736  0.967317
Crabp1     0.8584615  0.714878
Zeb2       0.6175824  0.501463
Mest       0.0154945  0.290976
...              ...       ...
AC125149.2         0         0
AC125149.4         0         0
AC234645.1         0         0
AC168977.2         0         0
Vmn2r122           0         0

The Top field provides the the minimum rank across all pairwise comparisons. The p.value field provides the combined p-value across all comparisons, and the FDR field the BH-adjusted p-value for each gene. The summary.AUC provides area under the curve (here the concordance probability) from the comparison with the lowest p-value, the AUC.n fields provide the AUC for each pairwise comparison. The AUC is the probability that a randomly selected cell in cluster A has a greater expression of gene X than a randomly selected cell in B.

We can then inspect the top marker genes for the first cluster using the plotExpression function from the scater package.

R

top.markers <- head(rownames(markers[[1]]))
plotExpression(sce, features = top.markers, x = "label", color_by = "label")

Clearly, not every marker gene distinguishes cluster 1 from every other cluster. However, with a combination of multiple marker genes it’s possible to clearly identify gene patterns that are unique to cluster 1. It’s sort of like the 20 questions game - with answers to the right questions about a cell (e.g. “Do you highly express Ptn?”), you can clearly identify what cluster it falls in.

Challenge

Why do you think marker genes are found by aggregating pairwise comparisons rather than iteratively comparing each cluster to all other clusters?

One important reason why is because averages over all other clusters can be sensitive to the cell type composition. If a rare cell type shows up in one sample, the most discriminative marker genes found in this way could be very different from those found in another sample where the rare cell type is absent.

Generally, it’s good to keep in mind that the concept of “everything else” is not a stable basis for comparison. Read that sentence again, because its a subtle but broadly applicable point. Think about it and you can probably identify analogous issues in fields outside of single-cell analysis. It frequently comes up when comparisons between multiple categories are involved.

Cell type annotation


The most challenging task in scRNA-seq data analysis is arguably the interpretation of the results. Obtaining clusters of cells is fairly straightforward, but it is more difficult to determine what biological state is represented by each of those clusters. Doing so requires us to bridge the gap between the current dataset and prior biological knowledge, and the latter is not always available in a consistent and quantitative manner. Indeed, even the concept of a “cell type” is not clearly defined, with most practitioners possessing a “I’ll know it when I see it” intuition that is not amenable to computational analysis. As such, interpretation of scRNA-seq data is often manual and a common bottleneck in the analysis workflow.

To expedite this step, we can use various computational approaches that exploit prior information to assign meaning to an uncharacterized scRNA-seq dataset. The most obvious sources of prior information are the curated gene sets associated with particular biological processes, e.g., from the Gene Ontology (GO) or the Kyoto Encyclopedia of Genes and Genomes (KEGG) collections. Alternatively, we can directly compare our expression profiles to published reference datasets where each sample or cell has already been annotated with its putative biological state by domain experts. Here, we will demonstrate both approaches on the wild-type chimera dataset.

Assigning cell labels from reference data

A conceptually straightforward annotation approach is to compare the single-cell expression profiles with previously annotated reference datasets. Labels can then be assigned to each cell in our uncharacterized test dataset based on the most similar reference sample(s), for some definition of “similar”. This is a standard classification challenge that can be tackled by standard machine learning techniques such as random forests and support vector machines. Any published and labelled RNA-seq dataset (bulk or single-cell) can be used as a reference, though its reliability depends greatly on the expertise of the original authors who assigned the labels in the first place.

In this section, we will demonstrate the use of the SingleR method for cell type annotation Aran et al., 2019. This method assigns labels to cells based on the reference samples with the highest Spearman rank correlations, using only the marker genes between pairs of labels to focus on the relevant differences between cell types. It also performs a fine-tuning step for each cell where the correlations are recomputed with just the marker genes for the top-scoring labels. This aims to resolve any ambiguity between those labels by removing noise from irrelevant markers for other labels. Further details can be found in the SingleR book from which most of the examples here are derived.

R

ref <- EmbryoAtlasData(samples = 29)

ref

OUTPUT

class: SingleCellExperiment
dim: 29452 7569
metadata(0):
assays(1): counts
rownames(29452): ENSMUSG00000051951 ENSMUSG00000089699 ...
  ENSMUSG00000096730 ENSMUSG00000095742
rowData names(2): ENSEMBL SYMBOL
colnames(7569): cell_95727 cell_95728 ... cell_103294 cell_103295
colData names(17): cell barcode ... colour sizeFactor
reducedDimNames(2): pca.corrected umap
mainExpName: NULL
altExpNames(0):

In order to reduce the computational load, we subsample the dataset to 1,000 cells.

R

set.seed(123)

ind <- sample(ncol(ref), 1000)

ref <- ref[,ind]

You can see we have an assortment of different cell types in the reference (with varying frequency):

R

tab <- sort(table(ref$celltype), decreasing = TRUE)
tab

OUTPUT


  Forebrain/Midbrain/Hindbrain                     Erythroid3
                           131                             75
             Paraxial mesoderm                            NMP
                            69                             51
                  ExE mesoderm               Surface ectoderm
                            49                             47
                     Allantois                     Mesenchyme
                            46                             45
                   Spinal cord            Pharyngeal mesoderm
                            45                             41
                  ExE endoderm                   Neural crest
                            38                             35
                           Gut Haematoendothelial progenitors
                            30                             27
         Intermediate mesoderm                 Cardiomyocytes
                            27                             26
              Somitic mesoderm                    Endothelium
                            25                             23
                    Erythroid2                  Def. endoderm
                            11                              3
                    Erythroid1            Blood progenitors 1
                             2                              1
           Blood progenitors 2                Caudal Mesoderm
                             1                              1
                           PGC
                             1 

We need the normalized log counts, so we add those on:

R

ref <- logNormCounts(ref)

Some cleaning - remove cells of the reference dataset for which the cell type annotation is missing:

R

nna <- !is.na(ref$celltype)

ref <- ref[,nna]

Also remove cell types of very low abundance (here less than 10 cells) to remove noise prior to subsequent annotation tasks.

R

abu.ct <- names(tab)[tab >= 10]

ind <- ref$celltype %in% abu.ct

ref <- ref[,ind] 

Restrict to genes shared between query and reference dataset.

R

rownames(ref) <- rowData(ref)$SYMBOL

shared_genes <- intersect(rownames(sce), rownames(ref))

sce <- sce[shared_genes,]

ref <- ref[shared_genes,]

Convert sparse assay matrices to regular dense matrices for input to SingleR:

R

sce.mat <- as.matrix(assay(sce, "logcounts"))

ref.mat <- as.matrix(assay(ref, "logcounts"))

Finally, run SingleR with the query and reference datasets:

R

res <- SingleR(test = sce.mat, 
               ref = ref.mat,
               labels = ref$celltype)
res

OUTPUT

DataFrame with 1000 rows and 4 columns
                                   scores                 labels delta.next
                                 <matrix>            <character>  <numeric>
cell_11995 0.348586:0.335451:0.314515:... Forebrain/Midbrain/H..  0.1285110
cell_10294 0.273570:0.260013:0.298932:...             Erythroid3  0.1381951
cell_9963  0.328538:0.291288:0.475611:...            Endothelium  0.2193295
cell_11610 0.281161:0.269245:0.299961:...             Erythroid3  0.0359215
cell_10910 0.422454:0.346897:0.355947:...           ExE mesoderm  0.0984285
...                                   ...                    ...        ...
cell_11597 0.323805:0.292967:0.300485:...                    NMP  0.1663369
cell_9807  0.464466:0.374189:0.381698:...             Mesenchyme  0.0833019
cell_10095 0.341721:0.288215:0.485324:...            Endothelium  0.0889931
cell_11706 0.267487:0.240215:0.286012:...             Erythroid2  0.0350557
cell_11860 0.345786:0.343437:0.313994:... Forebrain/Midbrain/H..  0.0117001
                    pruned.labels
                      <character>
cell_11995 Forebrain/Midbrain/H..
cell_10294             Erythroid3
cell_9963             Endothelium
cell_11610             Erythroid3
cell_10910           ExE mesoderm
...                           ...
cell_11597                    NMP
cell_9807              Mesenchyme
cell_10095            Endothelium
cell_11706             Erythroid2
cell_11860 Forebrain/Midbrain/H..

We inspect the results using a heatmap of the per-cell and label scores. Ideally, each cell should exhibit a high score in one label relative to all of the others, indicating that the assignment to that label was unambiguous.

R

plotScoreHeatmap(res)

We obtained fairly unambiguous predictions for mesenchyme and endothelial cells, whereas we see expectedly more ambiguity between the two erythroid cell populations.

We can also compare the cell type assignments with the unsupervised clustering results to determine the identity of each cluster. Here, several cell type classes are nested within the same cluster, indicating that these clusters are composed of several transcriptomically similar cell populations. On the other hand, there are also instances where we have several clusters for the same cell type, indicating that the clustering represents finer subdivisions within these cell types.

R

tab <- table(anno = res$pruned.labels, cluster = colLabels(sce))

pheatmap(log2(tab + 10), color = colorRampPalette(c("white", "blue"))(101))

As it so happens, we are in the fortunate position where our test dataset also contains independently defined labels. We see strong consistency between the two sets of labels, indicating that our automatic annotation is comparable to that generated manually by domain experts.

R

tab <- table(res$pruned.labels, sce$celltype.mapped)

pheatmap(log2(tab + 10), color = colorRampPalette(c("white", "blue"))(101))

Challenge

SingleR can be computationally expensive. How do you set it to run in parallel?

Use BiocParallel and the BPPARAM argument! This example will set it to use four cores on your laptop, but you can also configure BiocParallel to use cluster jobs.

R

library(BiocParallel)

my_bpparam = MulticoreParam(workers = 4)

res <- SingleR(test = sce.mat, 
               ref = ref.mat,
               labels = ref$celltype,
               BPPARAM = my_bpparam)

BiocParallel is the most common way to enable parallel computation in Bioconductor packages, so you can expect to see it elsewhere outside of SingleR.

Assigning cell labels from gene sets

A related strategy is to explicitly identify sets of marker genes that are highly expressed in each individual cell. This does not require matching of individual cells to the expression values of the reference dataset, which is faster and more convenient when only the identities of the markers are available. We demonstrate this approach using cell type markers derived from the mouse embryo atlas dataset.

R

wilcox.z <- pairwiseWilcox(ref, ref$celltype, lfc = 1, direction = "up")

markers.z <- getTopMarkers(wilcox.z$statistics, wilcox.z$pairs, 
                           pairwise = FALSE, n = 50)

lengths(markers.z)

OUTPUT

                     Allantois                 Cardiomyocytes
                           106                            106
                   Endothelium                     Erythroid2
                           103                             54
                    Erythroid3                   ExE endoderm
                            84                            102
                  ExE mesoderm   Forebrain/Midbrain/Hindbrain
                            97                             97
                           Gut Haematoendothelial progenitors
                            90                             71
         Intermediate mesoderm                     Mesenchyme
                            70                            118
                  Neural crest                            NMP
                            66                             91
             Paraxial mesoderm            Pharyngeal mesoderm
                            88                             85
              Somitic mesoderm                    Spinal cord
                            86                             91
              Surface ectoderm
                            92 

Our test dataset will be as before the wild-type chimera dataset.

R

sce

OUTPUT

class: SingleCellExperiment
dim: 29411 1000
metadata(0):
assays(2): counts logcounts
rownames(29411): Xkr4 Gm1992 ... Vmn2r122 CAAA01147332.1
rowData names(2): ENSEMBL SYMBOL
colnames(1000): cell_11995 cell_10294 ... cell_11706 cell_11860
colData names(12): cell barcode ... sizeFactor label
reducedDimNames(4): pca.corrected.E7.5 pca.corrected.E8.5 PCA UMAP
mainExpName: NULL
altExpNames(0):

We use the AUCell package to identify marker sets that are highly expressed in each cell. This method ranks genes by their expression values within each cell and constructs a response curve of the number of genes from each marker set that are present with increasing rank. It then computes the area under the curve (AUC) for each marker set, quantifying the enrichment of those markers among the most highly expressed genes in that cell. This is roughly similar to performing a Wilcoxon rank sum test between genes in and outside of the set, but involving only the top ranking genes by expression in each cell.

R

all.sets <- lapply(names(markers.z), 
                   function(x) GeneSet(markers.z[[x]], setName = x))

all.sets <- GeneSetCollection(all.sets)

all.sets

OUTPUT

GeneSetCollection
  names: Allantois, Cardiomyocytes, ..., Surface ectoderm (19 total)
  unique identifiers: Prrx2, Spin2c, ..., Igf2bp3 (560 total)
  types in collection:
    geneIdType: NullIdentifier (1 total)
    collectionType: NullCollection (1 total)

R

rankings <- AUCell_buildRankings(as.matrix(counts(sce)),
                                 plotStats = FALSE, verbose = FALSE)

cell.aucs <- AUCell_calcAUC(all.sets, rankings)

results <- t(assay(cell.aucs))

head(results)

OUTPUT

            gene sets
cells        Allantois Cardiomyocytes Endothelium Erythroid2 Erythroid3
  cell_11995    0.2199          0.234       0.195     0.1223      0.164
  cell_10294    0.1007          0.125       0.110     0.6073      0.608
  cell_9963     0.3427          0.316       0.541     0.1359      0.180
  cell_11610    0.0824          0.134       0.105     0.5789      0.571
  cell_10910    0.3176          0.278       0.245     0.0818      0.135
  cell_11021    0.2055          0.232       0.187     0.1135      0.177
            gene sets
cells        ExE endoderm ExE mesoderm Forebrain/Midbrain/Hindbrain   Gut
  cell_11995       0.0605        0.437                        0.632 0.378
  cell_10294       0.0637        0.190                        0.267 0.148
  cell_9963        0.0947        0.417                        0.497 0.381
  cell_11610       0.0572        0.206                        0.279 0.147
  cell_10910       0.1179        0.577                        0.572 0.445
  cell_11021       0.0929        0.474                        0.601 0.399
            gene sets
cells        Haematoendothelial progenitors Intermediate mesoderm Mesenchyme
  cell_11995                          0.333                 0.540      0.246
  cell_10294                          0.162                 0.178      0.108
  cell_9963                           0.601                 0.462      0.357
  cell_11610                          0.153                 0.197      0.112
  cell_10910                          0.433                 0.548      0.351
  cell_11021                          0.334                 0.520      0.240
            gene sets
cells        Neural crest   NMP Paraxial mesoderm Pharyngeal mesoderm
  cell_11995        0.654 0.484             0.551               0.508
  cell_10294        0.284 0.211             0.219               0.205
  cell_9963         0.537 0.374             0.501               0.462
  cell_11610        0.295 0.224             0.235               0.213
  cell_10910        0.547 0.523             0.551               0.556
  cell_11021        0.586 0.573             0.499               0.471
            gene sets
cells        Somitic mesoderm Spinal cord Surface ectoderm
  cell_11995            0.465       0.581            0.525
  cell_10294            0.214       0.229            0.187
  cell_9963             0.431       0.437            0.417
  cell_11610            0.234       0.256            0.205
  cell_10910            0.518       0.517            0.505
  cell_11021            0.519       0.553            0.505

We assign cell type identity to each cell in the test dataset by taking the marker set with the top AUC as the label for that cell. Our new labels mostly agree with the original annotation (and, thus, also with the reference-based annotation). Instances where the original annotation is divided into several new label groups typically points to large overlaps in their marker sets. In the absence of prior annotation, a more general diagnostic check is to compare the assigned labels to cluster identities, under the expectation that most cells of a single cluster would have the same label (or, if multiple labels are present, they should at least represent closely related cell states). We only print out the top-left corner of the table here, but you should try looking at the whole thing:

R

new.labels <- colnames(results)[max.col(results)]

tab <- table(new.labels, sce$celltype.mapped)

tab[1:4,1:4]

OUTPUT


new.labels       Allantois Blood progenitors 1 Blood progenitors 2
  Allantois             28                   0                   0
  Cardiomyocytes         0                   0                   0
  Endothelium            0                   0                   0
  Erythroid2             0                   0                   1

new.labels       Cardiomyocytes
  Allantois                   0
  Cardiomyocytes             27
  Endothelium                 0
  Erythroid2                  0

As a diagnostic measure, we examine the distribution of AUCs across cells for each label. In heterogeneous populations, the distribution for each label should be bimodal with one high-scoring peak containing cells of that cell type and a low-scoring peak containing cells of other types. The gap between these two peaks can be used to derive a threshold for whether a label is “active” for a particular cell. (In this case, we simply take the single highest-scoring label per cell as the labels should be mutually exclusive.) In populations where a particular cell type is expected, lack of clear bimodality for the corresponding label may indicate that its gene set is not sufficiently informative.

R

par(mfrow = c(3,3))

AUCell_exploreThresholds(cell.aucs[1:9], plotHist = TRUE, assign = TRUE) 

Shown is the distribution of AUCs in the wild-type chimera dataset for each label in the embryo atlas dataset. The blue curve represents the density estimate, the red curve represents a fitted two-component mixture of normals, the pink curve represents a fitted three-component mixture, and the grey curve represents a fitted normal distribution. Vertical lines represent threshold estimates corresponding to each estimate of the distribution.

Challenge

The diagnostics don’t look so good for some of the examples here. Which ones? Why?

The example that jumps out most strongly to the eye is ExE endoderm, which doesn’t show clear separate modes. Simultaneously, Endothelium seems to have three or four modes.

Remember, this is an exploratory diagnostic, not the final word! At this point it’d be good to engage in some critical inspection of the results. Maybe we don’t have enough / the best marker genes. In this particular case, the fact that we subsetted the reference set to 1000 cells probably didn’t help.

Session Info


R

sessionInfo()

OUTPUT

R version 4.4.1 (2024-06-14)
Platform: x86_64-pc-linux-gnu
Running under: Ubuntu 22.04.4 LTS

Matrix products: default
BLAS:   /usr/lib/x86_64-linux-gnu/blas/libblas.so.3.10.0
LAPACK: /usr/lib/x86_64-linux-gnu/lapack/liblapack.so.3.10.0

locale:
 [1] LC_CTYPE=C.UTF-8       LC_NUMERIC=C           LC_TIME=C.UTF-8
 [4] LC_COLLATE=C.UTF-8     LC_MONETARY=C.UTF-8    LC_MESSAGES=C.UTF-8
 [7] LC_PAPER=C.UTF-8       LC_NAME=C              LC_ADDRESS=C
[10] LC_TELEPHONE=C         LC_MEASUREMENT=C.UTF-8 LC_IDENTIFICATION=C

time zone: UTC
tzcode source: system (glibc)

attached base packages:
[1] stats4    stats     graphics  grDevices utils     datasets  methods
[8] base

other attached packages:
 [1] GSEABase_1.66.0              graph_1.82.0
 [3] annotate_1.82.0              XML_3.99-0.16.1
 [5] AnnotationDbi_1.66.0         pheatmap_1.0.12
 [7] scran_1.32.0                 scater_1.32.0
 [9] ggplot2_3.5.1                scuttle_1.14.0
[11] bluster_1.14.0               SingleR_2.6.0
[13] MouseGastrulationData_1.18.0 SpatialExperiment_1.14.0
[15] SingleCellExperiment_1.26.0  SummarizedExperiment_1.34.0
[17] Biobase_2.64.0               GenomicRanges_1.56.0
[19] GenomeInfoDb_1.40.1          IRanges_2.38.0
[21] S4Vectors_0.42.0             BiocGenerics_0.50.0
[23] MatrixGenerics_1.16.0        matrixStats_1.3.0
[25] AUCell_1.26.0                BiocStyle_2.32.0

loaded via a namespace (and not attached):
  [1] RColorBrewer_1.1-3        jsonlite_1.8.8
  [3] magrittr_2.0.3            ggbeeswarm_0.7.2
  [5] magick_2.8.3              farver_2.1.2
  [7] rmarkdown_2.27            zlibbioc_1.50.0
  [9] vctrs_0.6.5               memoise_2.0.1
 [11] DelayedMatrixStats_1.26.0 htmltools_0.5.8.1
 [13] S4Arrays_1.4.1            AnnotationHub_3.12.0
 [15] curl_5.2.1                BiocNeighbors_1.22.0
 [17] SparseArray_1.4.8         htmlwidgets_1.6.4
 [19] plotly_4.10.4             cachem_1.1.0
 [21] igraph_2.0.3              mime_0.12
 [23] lifecycle_1.0.4           pkgconfig_2.0.3
 [25] rsvd_1.0.5                Matrix_1.7-0
 [27] R6_2.5.1                  fastmap_1.2.0
 [29] GenomeInfoDbData_1.2.12   digest_0.6.35
 [31] colorspace_2.1-0          dqrng_0.4.1
 [33] irlba_2.3.5.1             ExperimentHub_2.12.0
 [35] RSQLite_2.3.7             beachmat_2.20.0
 [37] labeling_0.4.3            filelock_1.0.3
 [39] fansi_1.0.6               httr_1.4.7
 [41] abind_1.4-5               compiler_4.4.1
 [43] bit64_4.0.5               withr_3.0.0
 [45] BiocParallel_1.38.0       viridis_0.6.5
 [47] DBI_1.2.3                 highr_0.11
 [49] R.utils_2.12.3            MASS_7.3-60.2
 [51] rappdirs_0.3.3            DelayedArray_0.30.1
 [53] rjson_0.2.21              tools_4.4.1
 [55] vipor_0.4.7               beeswarm_0.4.0
 [57] R.oo_1.26.0               glue_1.7.0
 [59] nlme_3.1-164              grid_4.4.1
 [61] cluster_2.1.6             generics_0.1.3
 [63] gtable_0.3.5              R.methodsS3_1.8.2
 [65] tidyr_1.3.1               data.table_1.15.4
 [67] BiocSingular_1.20.0       ScaledMatrix_1.12.0
 [69] metapod_1.12.0            utf8_1.2.4
 [71] XVector_0.44.0            ggrepel_0.9.5
 [73] BiocVersion_3.19.1        pillar_1.9.0
 [75] limma_3.60.2              BumpyMatrix_1.12.0
 [77] splines_4.4.1             dplyr_1.1.4
 [79] BiocFileCache_2.12.0      lattice_0.22-6
 [81] survival_3.6-4            FNN_1.1.4
 [83] renv_1.0.7                bit_4.0.5
 [85] tidyselect_1.2.1          locfit_1.5-9.9
 [87] Biostrings_2.72.1         knitr_1.47
 [89] gridExtra_2.3             edgeR_4.2.0
 [91] xfun_0.44                 mixtools_2.0.0
 [93] statmod_1.5.0             UCSC.utils_1.0.0
 [95] lazyeval_0.2.2            yaml_2.3.8
 [97] evaluate_0.23             codetools_0.2-20
 [99] kernlab_0.9-32            tibble_3.2.1
[101] BiocManager_1.30.23       cli_3.6.2
[103] uwot_0.2.2                xtable_1.8-4
[105] segmented_2.1-0           munsell_0.5.1
[107] Rcpp_1.0.12               dbplyr_2.5.0
[109] png_0.1-8                 parallel_4.4.1
[111] blob_1.2.4                sparseMatrixStats_1.16.0
[113] viridisLite_0.4.2         scales_1.3.0
[115] purrr_1.0.2               crayon_1.5.2
[117] rlang_1.1.3               cowplot_1.1.3
[119] KEGGREST_1.44.0          

Exercises


Exercise 1: Clustering

The Leiden algorithm is similar to the Louvain algorithm, but it is faster and has been shown to result in better connected communities. Modify the above call to clusterCells to carry out the community detection with the Leiden algorithm instead. Visualize the results in a UMAP plot.

The NNGraphParam constructor has an argument cluster.args. This allows to specify arguments passed on to the cluster_leiden function from the igraph package. Use the cluster.args argument to parameterize the clustering to use modularity as the objective function and a resolution parameter of 0.5.

TODO

Exercise 2: Cluster annotation

Another strategy for annotating the clusters is to perform a gene set enrichment analysis on the marker genes defining each cluster. This identifies the pathways and processes that are (relatively) active in each cluster based on upregulation of the associated genes compared to other clusters. Focus on the top 100 up-regulated genes in a cluster of your choice and perform a gene set enrichment analysis of biological process (BP) gene sets from the Gene Ontology (GO).

Use the goana() function from the limma package to identify GO BP terms that are overrepresented in the list of marker genes.

TODO

Exercise 3: Workflow

The scRNAseq package provides gene-level counts for a collection of public scRNA-seq datasets, stored as SingleCellExperiment objects with annotated cell- and gene-level metadata. Consult the vignette of the scRNAseq package to inspect all available datasets and select a dataset of your choice. Perform a typical scRNA-seq analysis on this dataset including QC, normalization, feature selection, dimensionality reduction, clustering, and marker gene detection.

TODO

Further Reading

Key Points

  • The two main approaches for cell type annotation are 1) manual annotation of clusters based on marker gene expression, and 2) computational annotation based on annotation transfer from reference datasets or marker gene set enrichment testing.
  • For manual annotation, cells are first clustered with unsupervised methods such as graph-based clustering followed by community detection algorithms such as Louvain or Leiden.
  • The clusterCells function from the scran package provides different algorithms that are commonly used for the clustering of scRNA-seq data.
  • Once clusters have been obtained, cell type labels are then manually assigned to cell clusters by matching cluster-specific upregulated marker genes with prior knowledge of cell-type markers.
  • The findMarkers function from the scran package package can be used to find candidate marker genes for clusters of cells by testing for differential expression between pairs of clusters.
  • Computational annotation using published reference datasets or curated gene sets provides a fast, automated, and reproducible alternative to the manual annotation of cell clusters based on marker gene expression.
  • The SingleR package is a popular choice for reference-based annotation and assigns labels to cells based on the reference samples with the highest Spearman rank correlations.
  • The AUCell package provides an enrichment test to identify curated marker sets that are highly expressed in each cell.