Multi-sample analyses

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

Estimated time: 45 minutes

Overview

Questions

  • How can we integrate data from multiple batches, samples, and studies?
  • How can we identify differentially expressed genes between experimental conditions for each cell type?
  • How can we identify changes in cell type abundance between experimental conditions?

Objectives

  • Correct batch effects and diagnose potential problems such as over-correction.
  • Perform differential expression comparisons between conditions based on pseudo-bulk samples.
  • Perform differential abundance comparisons between conditions.

Setup and data exploration


As before, we will use the the wild-type data from the Tal1 chimera experiment:

  • Sample 5: E8.5 injected cells (tomato positive), pool 3
  • Sample 6: E8.5 host cells (tomato negative), pool 3
  • Sample 7: E8.5 injected cells (tomato positive), pool 4
  • Sample 8: E8.5 host cells (tomato negative), pool 4
  • Sample 9: E8.5 injected cells (tomato positive), pool 5
  • Sample 10: E8.5 host cells (tomato negative), pool 5

Note that this is a paired design in which for each biological replicate (pool 3, 4, and 5), we have both host and injected cells.

We start by loading the data and doing a quick exploratory analysis, essentially applying the normalization and visualization techniques that we have seen in the previous lectures to all samples. Note that this time we’re selecting samples 5 to 10, not just 5 by itself.

R

library(MouseGastrulationData)
library(batchelor)
library(edgeR)
library(scater)
library(ggplot2)
library(scran)
library(pheatmap)
library(scuttle)

sce <- WTChimeraData(samples = 5:10, type = "processed")

sce

OUTPUT

class: SingleCellExperiment
dim: 29453 20935
metadata(0):
assays(1): counts
rownames(29453): ENSMUSG00000051951 ENSMUSG00000089699 ...
  ENSMUSG00000095742 tomato-td
rowData names(2): ENSEMBL SYMBOL
colnames(20935): cell_9769 cell_9770 ... cell_30702 cell_30703
colData names(11): cell barcode ... doub.density sizeFactor
reducedDimNames(2): pca.corrected.E7.5 pca.corrected.E8.5
mainExpName: NULL
altExpNames(0):

R

colData(sce)

OUTPUT

DataFrame with 20935 rows and 11 columns
                  cell          barcode    sample       stage    tomato
           <character>      <character> <integer> <character> <logical>
cell_9769    cell_9769 AAACCTGAGACTGTAA         5        E8.5      TRUE
cell_9770    cell_9770 AAACCTGAGATGCCTT         5        E8.5      TRUE
cell_9771    cell_9771 AAACCTGAGCAGCCTC         5        E8.5      TRUE
cell_9772    cell_9772 AAACCTGCATACTCTT         5        E8.5      TRUE
cell_9773    cell_9773 AAACGGGTCAACACCA         5        E8.5      TRUE
...                ...              ...       ...         ...       ...
cell_30699  cell_30699 TTTGTCACAGCTCGCA        10        E8.5     FALSE
cell_30700  cell_30700 TTTGTCAGTCTAGTCA        10        E8.5     FALSE
cell_30701  cell_30701 TTTGTCATCATCGGAT        10        E8.5     FALSE
cell_30702  cell_30702 TTTGTCATCATTATCC        10        E8.5     FALSE
cell_30703  cell_30703 TTTGTCATCCCATTTA        10        E8.5     FALSE
                pool stage.mapped        celltype.mapped closest.cell
           <integer>  <character>            <character>  <character>
cell_9769          3        E8.25             Mesenchyme   cell_24159
cell_9770          3         E8.5            Endothelium   cell_96660
cell_9771          3         E8.5              Allantois  cell_134982
cell_9772          3         E8.5             Erythroid3  cell_133892
cell_9773          3        E8.25             Erythroid1   cell_76296
...              ...          ...                    ...          ...
cell_30699         5         E8.5             Erythroid3   cell_38810
cell_30700         5         E8.5       Surface ectoderm   cell_38588
cell_30701         5        E8.25 Forebrain/Midbrain/H..   cell_66082
cell_30702         5         E8.5             Erythroid3  cell_138114
cell_30703         5         E8.0                Doublet   cell_92644
           doub.density sizeFactor
              <numeric>  <numeric>
cell_9769    0.02985045    1.41243
cell_9770    0.00172753    1.22757
cell_9771    0.01338013    1.15439
cell_9772    0.00218402    1.28676
cell_9773    0.00211723    1.78719
...                 ...        ...
cell_30699   0.00146287   0.389311
cell_30700   0.00374155   0.588784
cell_30701   0.05651258   0.624455
cell_30702   0.00108837   0.550807
cell_30703   0.82369305   1.184919

To speed up computations, after removing doublets, we randomly select 50% cells per sample.

R

drop <- sce$celltype.mapped %in% c("stripped", "Doublet")

sce <- sce[,!drop]

set.seed(29482)

idx <- unlist(tapply(colnames(sce), sce$sample, function(x) {
    perc <- round(0.50 * length(x))
    sample(x, perc)
}))

sce <- sce[,idx]

We now normalize the data, run some dimensionality reduction steps, and visualize them in a tSNE plot.

R

sce <- logNormCounts(sce)

dec <- modelGeneVar(sce, block = sce$sample)

chosen.hvgs <- dec$bio > 0

sce <- runPCA(sce, subset_row = chosen.hvgs, ntop = 1000)

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

sce$sample <- as.factor(sce$sample)

plotTSNE(sce, colour_by = "sample")

R

plotTSNE(sce, colour_by = "celltype.mapped") +
    scale_color_discrete() +
    theme(legend.position = "bottom")

There are evident sample effects. Depending on the analysis that you want to perform you may want to remove or retain the sample effect. For instance, if the goal is to identify cell types with a clustering method, one may want to remove the sample effects with “batch effect” correction methods.

For now, let’s assume that we want to remove this effect.

Challenge

It seems like samples 5 and 6 are separated off from the others in gene expression space. Given the group of cells in each sample, why might this make sense versus some other pair of samples? What is the factor presumably leading to this difference?

Samples 5 and 6 were from the same “pool” of cells. Looking at the documentation for the dataset under ?WTChimeraData we see that the pool variable is defined as: “Integer, embryo pool from which cell derived; samples with same value are matched.” So samples 5 and 6 have an experimental factor in common which causes a shared, systematic difference in gene expression profiles compared to the other samples. That’s why you can see many of isolated blue/orange clusters on the first TSNE plot. If you were developing single-cell library preparation protocols you might want to preserve this effect to understand how variation in pools leads to variation in expression, but for now, given that we’re investigating other effects, we’ll want to remove this as undesired technical variation.

Correcting batch effects


We “correct” the effect of samples with the correctExperiment function in the batchelor package and using the sample column as batch.

R

set.seed(10102)

merged <- correctExperiments(
    sce, 
    batch = sce$sample, 
    subset.row = chosen.hvgs,
    PARAM = FastMnnParam(
        merge.order = list(
            list(1,3,5), # WT (3 replicates)
            list(2,4,6)  # td-Tomato (3 replicates)
        )
    )
)

merged <- runTSNE(merged, dimred = "corrected")

plotTSNE(merged, colour_by = "batch")

Once we removed the sample batch effect, we can proceed with the Differential Expression Analysis.

Challenge

Having multiple independent samples in each experimental group is always helpful, but it particularly important when it comes to batch effect correction. Why?

It’s important to have multiple samples within each experimental group because it helps the batch effect correction algorithm distinguish differences due to batch effects (uninteresting) from differences due to biology (interesting).

Imagine you had one sample that received a drug treatment and one that did not, each with 10,000 cells. They differ substantially in expression of gene X. Is that an important scientific finding? You can’t tell for sure, because the effect of drug is indistinguishable from a sample-wise batch effect. But if the difference in gene X holds up when you have five treated samples and five untreated samples, now you can be a bit more confident. Many batch effect correction methods will take information on experimental factors as additional arguments, which they can use to help remove batch effects while retaining experimental differences.

Differential Expression


In order to perform a differential expression analysis, we need to identify groups of cells across samples/conditions (depending on the experimental design and the final aim of the experiment).

As previously seen, we have two ways of grouping cells, cell clustering and cell labeling. In our case we will focus on this second aspect to group cells according to the already annotated cell types to proceed with the computation of the pseudo-bulk samples.

Pseudo-bulk samples

To compute differences between groups of cells, a possible way is to compute pseudo-bulk samples, where we mediate the gene signal of all the cells for each specific cell type. In this manner, we are then able to detect differences between the same cell type across two different conditions.

To compute pseudo-bulk samples, we use the aggregateAcrossCells function in the scuttle package, which takes as input not only a SingleCellExperiment, but also the id to use for the identification of the group of cells. In our case, we use as id not just the cell type, but also the sample, because we want be able to discern between replicates and conditions during further steps.

R

# Using 'label' and 'sample' as our two factors; each column of the output
# corresponds to one unique combination of these two factors.

summed <- aggregateAcrossCells(
    merged, 
    id = colData(merged)[,c("celltype.mapped", "sample")]
)

summed

OUTPUT

class: SingleCellExperiment
dim: 13641 179
metadata(2): merge.info pca.info
assays(1): counts
rownames(13641): ENSMUSG00000051951 ENSMUSG00000025900 ...
  ENSMUSG00000096730 ENSMUSG00000095742
rowData names(3): rotation ENSEMBL SYMBOL
colnames: NULL
colData names(15): batch cell ... sample ncells
reducedDimNames(5): corrected pca.corrected.E7.5 pca.corrected.E8.5 PCA
  TSNE
mainExpName: NULL
altExpNames(0):

Differential Expression Analysis

The main advantage of using pseudo-bulk samples is the possibility to use well-tested methods for differential analysis like edgeR and DESeq2, we will focus on the former for this analysis. edgeR uses a Negative Binomial Generalized Linear Model.

First, let’s start with a specific cell type, for instance the “Mesenchymal stem cells”, and look into differences between this cell type across conditions.

R

label <- "Mesenchyme"

current <- summed[,label == summed$celltype.mapped]

y <- DGEList(counts(current), samples = colData(current))

y

OUTPUT

An object of class "DGEList"
$counts
                   Sample1 Sample2 Sample3 Sample4 Sample5 Sample6
ENSMUSG00000051951       2       0       0       0       1       0
ENSMUSG00000025900       0       0       0       0       0       0
ENSMUSG00000025902       4       0       2       0       3       6
ENSMUSG00000033845     765     130     508     213     781     305
ENSMUSG00000002459       2       0       1       0       0       0
13636 more rows ...

$samples
        group lib.size norm.factors batch cell barcode sample stage tomato pool
Sample1     1  2478901            1     5 <NA>    <NA>      5  E8.5   TRUE    3
Sample2     1   548407            1     6 <NA>    <NA>      6  E8.5  FALSE    3
Sample3     1  1260187            1     7 <NA>    <NA>      7  E8.5   TRUE    4
Sample4     1   578699            1     8 <NA>    <NA>      8  E8.5  FALSE    4
Sample5     1  2092329            1     9 <NA>    <NA>      9  E8.5   TRUE    5
Sample6     1   904929            1    10 <NA>    <NA>     10  E8.5  FALSE    5
        stage.mapped celltype.mapped closest.cell doub.density sizeFactor
Sample1         <NA>      Mesenchyme         <NA>           NA         NA
Sample2         <NA>      Mesenchyme         <NA>           NA         NA
Sample3         <NA>      Mesenchyme         <NA>           NA         NA
Sample4         <NA>      Mesenchyme         <NA>           NA         NA
Sample5         <NA>      Mesenchyme         <NA>           NA         NA
Sample6         <NA>      Mesenchyme         <NA>           NA         NA
        celltype.mapped.1 sample.1 ncells
Sample1        Mesenchyme        5    151
Sample2        Mesenchyme        6     28
Sample3        Mesenchyme        7    127
Sample4        Mesenchyme        8     75
Sample5        Mesenchyme        9    239
Sample6        Mesenchyme       10    146

A typical step is to discard low quality samples due to low sequenced library size. We discard these samples because they can affect further steps like normalization and/or DEGs analysis.

We can see that in our case we don’t have low quality samples and we don’t need to filter out any of them.

R

discarded <- current$ncells < 10

y <- y[,!discarded]

summary(discarded)

OUTPUT

   Mode   FALSE
logical       6 

The same idea is typically applied to the genes, indeed we need to discard low expressed genes to improve accuracy for the DEGs modeling.

R

keep <- filterByExpr(y, group = current$tomato)

y <- y[keep,]

summary(keep)

OUTPUT

   Mode   FALSE    TRUE
logical    9121    4520 

We can now proceed to normalize the data There are several approaches for normalizing bulk, and hence pseudo-bulk data. Here, we use the Trimmed Mean of M-values method, implemented in the edgeR package within the calcNormFactors function. Keep in mind that because we are going to normalize the pseudo-bulk counts, we don’t need to normalize the data in “single cell form”.

R

y <- calcNormFactors(y)

y$samples

OUTPUT

        group lib.size norm.factors batch cell barcode sample stage tomato pool
Sample1     1  2478901    1.0506857     5 <NA>    <NA>      5  E8.5   TRUE    3
Sample2     1   548407    1.0399112     6 <NA>    <NA>      6  E8.5  FALSE    3
Sample3     1  1260187    0.9700083     7 <NA>    <NA>      7  E8.5   TRUE    4
Sample4     1   578699    0.9871129     8 <NA>    <NA>      8  E8.5  FALSE    4
Sample5     1  2092329    0.9695559     9 <NA>    <NA>      9  E8.5   TRUE    5
Sample6     1   904929    0.9858611    10 <NA>    <NA>     10  E8.5  FALSE    5
        stage.mapped celltype.mapped closest.cell doub.density sizeFactor
Sample1         <NA>      Mesenchyme         <NA>           NA         NA
Sample2         <NA>      Mesenchyme         <NA>           NA         NA
Sample3         <NA>      Mesenchyme         <NA>           NA         NA
Sample4         <NA>      Mesenchyme         <NA>           NA         NA
Sample5         <NA>      Mesenchyme         <NA>           NA         NA
Sample6         <NA>      Mesenchyme         <NA>           NA         NA
        celltype.mapped.1 sample.1 ncells
Sample1        Mesenchyme        5    151
Sample2        Mesenchyme        6     28
Sample3        Mesenchyme        7    127
Sample4        Mesenchyme        8     75
Sample5        Mesenchyme        9    239
Sample6        Mesenchyme       10    146

To investigate the effect of our normalization, we use a Mean-Difference (MD) plot for each sample in order to detect possible normalization problems due to insufficient cells/reads/UMIs composing a particular pseudo-bulk profile.

In our case, we verify that all these plots are centered in 0 (on y-axis) and present a trumpet shape, as expected.

R

par(mfrow = c(2,3))

for (i in seq_len(ncol(y))) {
    plotMD(y, column = i)
}

R

par(mfrow = c(1,1))

Furthermore, we want to check if the samples cluster together based on their known factors (like the tomato injection in this case).

To do so, we use the MDS plot, which is very close to a PCA representation.

R

limma::plotMDS(cpm(y, log = TRUE), 
               col = ifelse(y$samples$tomato, "red", "blue"))

We then construct a design matrix by including both the pool and the tomato as factors. This design indicates which samples belong to which pool and condition, so we can use it in the next step of the analysis.

R

design <- model.matrix(~factor(pool) + factor(tomato),
                       data = y$samples)
design

OUTPUT

        (Intercept) factor(pool)4 factor(pool)5 factor(tomato)TRUE
Sample1           1             0             0                  1
Sample2           1             0             0                  0
Sample3           1             1             0                  1
Sample4           1             1             0                  0
Sample5           1             0             1                  1
Sample6           1             0             1                  0
attr(,"assign")
[1] 0 1 1 2
attr(,"contrasts")
attr(,"contrasts")$`factor(pool)`
[1] "contr.treatment"

attr(,"contrasts")$`factor(tomato)`
[1] "contr.treatment"

Now we can estimate the Negative Binomial (NB) overdispersion parameter, to model the mean-variance trend.

R

y <- estimateDisp(y, design)

summary(y$trended.dispersion)

OUTPUT

    Min.  1st Qu.   Median     Mean  3rd Qu.     Max.
0.009325 0.016271 0.024233 0.021603 0.026868 0.027993 

The BCV plot allows us to investigate the relation between the Biological Coefficient of Variation and the Average log CPM for each gene. Additionally, the Common and Trend BCV are shown in red and blue.

R

plotBCV(y)

We then fit a Quasi-Likelihood (QL) negative binomial generalized linear model for each gene. The robust = TRUE parameter avoids distortions from highly variable clusters. The QL method includes an additional dispersion parameter, useful to handle the uncertainty and variability of the per-gene variance, which is not well estimated by the NB dispersions, so the two dispersion types complement each other in the final analysis.

R

fit <- glmQLFit(y, design, robust = TRUE)

summary(fit$var.prior)

OUTPUT

   Min. 1st Qu.  Median    Mean 3rd Qu.    Max.
 0.3189  0.9705  1.0901  1.0251  1.1486  1.2151 

R

summary(fit$df.prior)

OUTPUT

   Min. 1st Qu.  Median    Mean 3rd Qu.    Max.
 0.3046  8.7242  8.7242  8.6466  8.7242  8.7242 

QL dispersion estimates for each gene as a function of abundance. Raw estimates (black) are shrunk towards the trend (blue) to yield squeezed estimates (red).

R

plotQLDisp(fit)

We then use an empirical Bayes quasi-likelihood F-test to test for differential expression (due to tomato injection) per each gene at a False Discovery Rate (FDR) of 5%. The low amount of DGEs highlights that the tomato injection effect has a low influence on the mesenchyme cells.

R

res <- glmQLFTest(fit, coef = ncol(design))

summary(decideTests(res))

OUTPUT

       factor(tomato)TRUE
Down                    5
NotSig               4510
Up                      5

R

topTags(res)

OUTPUT

Coefficient:  factor(tomato)TRUE
                        logFC   logCPM          F       PValue          FDR
ENSMUSG00000010760 -4.1515323 9.973704 1118.17411 3.424939e-12 1.548073e-08
ENSMUSG00000096768  1.9987246 8.844258  375.41194 1.087431e-09 2.457594e-06
ENSMUSG00000035299  1.7963926 6.904163  119.08173 3.853318e-07 5.805666e-04
ENSMUSG00000086503 -6.4701526 7.411257  238.72531 1.114877e-06 1.259812e-03
ENSMUSG00000101609  1.3784805 7.310009   79.94279 2.682051e-06 2.424574e-03
ENSMUSG00000019188 -1.0191494 7.545530   62.01494 8.860823e-06 6.675153e-03
ENSMUSG00000024423  0.9940616 7.391075   56.84876 1.322645e-05 8.540506e-03
ENSMUSG00000042607 -0.9508732 7.468203   45.43086 3.625976e-05 2.048676e-02
ENSMUSG00000036446 -0.8280894 9.401028   43.03008 4.822988e-05 2.293136e-02
ENSMUSG00000027520  1.5929714 6.952923   42.86686 5.073310e-05 2.293136e-02

All the previous steps can be easily performed with the following function for each cell type, thanks to the pseudoBulkDGE function in the scran package.

R

summed.filt <- summed[,summed$ncells >= 10]

de.results <- pseudoBulkDGE(
    summed.filt, 
    label = summed.filt$celltype.mapped,
    design = ~factor(pool) + tomato,
    coef = "tomatoTRUE",
    condition = summed.filt$tomato 
)

The returned object is a list of DataFrames each with the results for a cell type. Each of these contains also the intermediate results in edgeR format to perform any intermediate plot or diagnostic.

R

cur.results <- de.results[["Allantois"]]

cur.results[order(cur.results$PValue),]

OUTPUT

DataFrame with 13641 rows and 5 columns
                       logFC    logCPM         F      PValue         FDR
                   <numeric> <numeric> <numeric>   <numeric>   <numeric>
ENSMUSG00000037664 -7.993129  11.55290  2730.230 1.05747e-13 4.44242e-10
ENSMUSG00000010760 -2.575007  12.40592  1049.284 1.33098e-11 2.79572e-08
ENSMUSG00000086503 -7.015618   7.49749   788.150 5.88102e-11 8.23539e-08
ENSMUSG00000096768  1.828366   9.33239   343.044 3.58836e-09 3.76868e-06
ENSMUSG00000022464  0.970431  10.28302   126.467 4.59369e-07 3.85961e-04
...                      ...       ...       ...         ...         ...
ENSMUSG00000095247        NA        NA        NA          NA          NA
ENSMUSG00000096808        NA        NA        NA          NA          NA
ENSMUSG00000079808        NA        NA        NA          NA          NA
ENSMUSG00000096730        NA        NA        NA          NA          NA
ENSMUSG00000095742        NA        NA        NA          NA          NA

Challenge

Clearly some of the results have low p-values. What about the effect sizes? What does logFC stand for?

“logFC” stands for log fold-change. Rather than reporting e.g. a 5-fold increase, it’s better to report a logFC of log(5) = 1.61. Additive log scales are easier to work with than multiplicative identity scales, once you get used to it.

ENSMUSG00000037664 seems to have an estimated logFC of about -8. That’s a big difference if it’s real.

Differential Abundance


With DA we look for differences in cluster abundance across conditions (the tomato injection in our case), rather than differences in gene expression.

Our first steps are quantifying the number of cells per each cell type and fitting a model to catch differences between the injected cells and the background.

The process is very similar differential expression modeling, but this time we start our analysis on the computed abundances and without normalizing the data with TMM.

R

abundances <- table(merged$celltype.mapped, merged$sample) 

abundances <- unclass(abundances) 

extra.info <- colData(merged)[match(colnames(abundances), merged$sample),]

y.ab <- DGEList(abundances, samples = extra.info)

design <- model.matrix(~factor(pool) + factor(tomato), y.ab$samples)

y.ab <- estimateDisp(y.ab, design, trend = "none")

fit.ab <- glmQLFit(y.ab, design, robust = TRUE, abundance.trend = FALSE)

Background on compositional effect

As mentioned before, in DA we don’t normalize our data with calcNormFactors function, because this approach considers that most of the input features do not vary between conditions. This cannot be applied to DA analysis because we have a small number of cell populations that all can change due to the treatment. This means that here we will normalize only for library depth, which in pseudo-bulk data means by the total number of cells in each sample (cell type).

On the other hand, this can lead our data to be susceptible to compositional effect. “Compositional” refers to the fact that the cluster abundances in a sample are not independent of one another because each cell type is effectively competing for space in the sample. They behave like proportions. If cell type A abundance increases in a new condition, that means we’ll observe less of everything else, even if everything else is unaffected by the new condition.

Compositionality means that our conclusions can be biased by the amount of cells present in each cell type. And this amount of cells can be totally unbalanced between cell types. This is particularly problematic for cell types that start at or end up near 0 or 100 percent.

For example, a specific cell type can be 40% of the total amount of cells present in the experiment, while another just the 3%. The differences in terms of abundance of these cell types are detected between the different conditions, but our final interpretation could be biased if we don’t consider this aspect.

We now look at different approaches for handling the compositional effect.

Assuming most labels do not change

We can use a similar approach used during the DEGs analysis, assuming that most labels are not changing, in particular if we think about the low number of DEGs resulted from the previous analysis.

To do so, we first normalize the data with calcNormFactors and then we fit and estimate a QL-model for our abundance data.

R

y.ab2 <- calcNormFactors(y.ab)

y.ab2$samples$norm.factors

OUTPUT

[1] 1.1029040 1.0228173 1.0695358 0.7686501 1.0402941 1.0365354

We then use edgeR in a manner similar to what we ran before:

R

y.ab2 <- estimateDisp(y.ab2, design, trend = "none")

fit.ab2 <- glmQLFit(y.ab2, design, robust = TRUE, abundance.trend = FALSE)

res2 <- glmQLFTest(fit.ab2, coef = ncol(design))

summary(decideTests(res2))

OUTPUT

       factor(tomato)TRUE
Down                    4
NotSig                 29
Up                      1

R

topTags(res2, n = 10)

OUTPUT

Coefficient:  factor(tomato)TRUE
                       logFC   logCPM         F       PValue          FDR
ExE ectoderm      -5.7983253 13.13490 34.326044 1.497957e-07 5.093053e-06
Parietal endoderm -6.9219242 12.36649 21.805721 1.468320e-05 2.496144e-04
Erythroid3        -0.9115099 17.34677 12.478845 7.446554e-04 8.439428e-03
Mesenchyme         0.9796446 16.32654 11.692412 1.064808e-03 9.050865e-03
Neural crest      -1.0469872 14.83912  7.956363 6.274678e-03 4.266781e-02
Endothelium        0.9241543 14.12195  4.437179 3.885736e-02 2.201917e-01
Erythroid2        -0.6029365 15.97357  3.737927 5.735479e-02 2.682206e-01
Cardiomyocytes     0.6789803 14.93321  3.569604 6.311073e-02 2.682206e-01
ExE endoderm      -3.9996258 10.75172  3.086597 8.344133e-02 3.125815e-01
Allantois          0.5462287 15.54924  2.922074 9.193574e-02 3.125815e-01

Testing against a log-fold change threshold

This other approach assumes that the composition bias introduces a spurious log2-fold change of no more than a quantity for a non-DA label.

In other words, we interpret this as the maximum log-fold change in the total number of cells given by DA in other labels. On the other hand, when choosing , we should not consider fold-differences in the totals due to differences in capture efficiency or the size of the original cell population are not attributable to composition bias. We then mitigate the effect of composition biases by testing each label for changes in abundance beyond .

R

res.lfc <- glmTreat(fit.ab, coef = ncol(design), lfc = 1)

summary(decideTests(res.lfc))

OUTPUT

       factor(tomato)TRUE
Down                    2
NotSig                 32
Up                      0

R

topTags(res.lfc)

OUTPUT

Coefficient:  factor(tomato)TRUE
                         logFC unshrunk.logFC   logCPM       PValue
ExE ectoderm        -5.5156915     -5.9427658 13.06465 5.730409e-06
Parietal endoderm   -6.5897795    -27.4235942 12.30091 1.215896e-04
ExE endoderm        -3.9307381    -23.9369433 10.76159 7.352966e-02
Mesenchyme           1.1615857      1.1628182 16.35239 1.335104e-01
Endothelium          1.0564619      1.0620564 14.14422 2.136590e-01
Caudal neurectoderm -1.4588627     -1.6095620 11.09613 3.257325e-01
Cardiomyocytes       0.8521199      0.8545967 14.96579 3.649535e-01
Neural crest        -0.8366836     -0.8392250 14.83184 3.750471e-01
Def. endoderm        0.7335519      0.7467590 12.50001 4.219471e-01
Allantois            0.7637525      0.7650565 15.54528 4.594987e-01
                             FDR
ExE ectoderm        0.0001948339
Parietal endoderm   0.0020670230
ExE endoderm        0.8333361231
Mesenchyme          0.9866105512
Endothelium         0.9866105512
Caudal neurectoderm 0.9866105512
Cardiomyocytes      0.9866105512
Neural crest        0.9866105512
Def. endoderm       0.9866105512
Allantois           0.9866105512

Addionally, the choice of can be guided by other external experimental data, like a previous or a pilot experiment.

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] pheatmap_1.0.12              scran_1.32.0
 [3] scater_1.32.0                ggplot2_3.5.1
 [5] scuttle_1.14.0               edgeR_4.2.0
 [7] limma_3.60.2                 batchelor_1.20.0
 [9] MouseGastrulationData_1.18.0 SpatialExperiment_1.14.0
[11] SingleCellExperiment_1.26.0  SummarizedExperiment_1.34.0
[13] Biobase_2.64.0               GenomicRanges_1.56.0
[15] GenomeInfoDb_1.40.1          IRanges_2.38.0
[17] S4Vectors_0.42.0             BiocGenerics_0.50.0
[19] MatrixGenerics_1.16.0        matrixStats_1.3.0
[21] 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         cachem_1.1.0
 [19] ResidualMatrix_1.14.0     igraph_2.0.3
 [21] mime_0.12                 lifecycle_1.0.4
 [23] pkgconfig_2.0.3           rsvd_1.0.5
 [25] Matrix_1.7-0              R6_2.5.1
 [27] fastmap_1.2.0             GenomeInfoDbData_1.2.12
 [29] digest_0.6.35             colorspace_2.1-0
 [31] AnnotationDbi_1.66.0      dqrng_0.4.1
 [33] irlba_2.3.5.1             ExperimentHub_2.12.0
 [35] RSQLite_2.3.7             beachmat_2.20.0
 [37] filelock_1.0.3            labeling_0.4.3
 [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] rappdirs_0.3.3            DelayedArray_0.30.1
 [51] rjson_0.2.21              bluster_1.14.0
 [53] tools_4.4.1               vipor_0.4.7
 [55] beeswarm_0.4.0            glue_1.7.0
 [57] grid_4.4.1                Rtsne_0.17
 [59] cluster_2.1.6             generics_0.1.3
 [61] gtable_0.3.5              BiocSingular_1.20.0
 [63] ScaledMatrix_1.12.0       metapod_1.12.0
 [65] utf8_1.2.4                XVector_0.44.0
 [67] ggrepel_0.9.5             BiocVersion_3.19.1
 [69] pillar_1.9.0              BumpyMatrix_1.12.0
 [71] splines_4.4.1             dplyr_1.1.4
 [73] BiocFileCache_2.12.0      lattice_0.22-6
 [75] renv_1.0.7                bit_4.0.5
 [77] tidyselect_1.2.1          locfit_1.5-9.9
 [79] Biostrings_2.72.1         knitr_1.47
 [81] gridExtra_2.3             xfun_0.44
 [83] statmod_1.5.0             UCSC.utils_1.0.0
 [85] yaml_2.3.8                evaluate_0.23
 [87] codetools_0.2-20          tibble_3.2.1
 [89] BiocManager_1.30.23       cli_3.6.2
 [91] munsell_0.5.1             Rcpp_1.0.12
 [93] dbplyr_2.5.0              png_0.1-8
 [95] parallel_4.4.1            blob_1.2.4
 [97] sparseMatrixStats_1.16.0  viridisLite_0.4.2
 [99] scales_1.3.0              purrr_1.0.2
[101] crayon_1.5.2              rlang_1.1.3
[103] cowplot_1.1.3             KEGGREST_1.44.0
[105] formatR_1.14             

Exercises


Exercise 1: Replicates

Test differential expressed genes with just 2 replicates per condition and look into the changes in the results and possible emerging issues.

Remember, you can subset SingleCellExperiments with logical indices, just like a matrix. You can also access their column data with the $ accessor, like a data frame.

R

summed.filt.subset = summed.filt[,summed.filt$pool != 3]

de.results <- pseudoBulkDGE(
    summed.filt.subset, 
    label = summed.filt.subset$celltype.mapped,
    design = ~factor(pool) + tomato,
    coef = "tomatoTRUE",
    condition = summed.filt.subset$tomato 
)

Exercise 2:

Use the pheatmap package to create a heatmap of the abundances table. Does it comport with the model results?

You can just hand pheatmap() a matrix as its only argument. It has a million options, but the defaults are usually pretty good.

R

pheatmap(y.ab$counts)

The top DA result was a decrease in ExE ectoderm in the tomato condition, which you can sort of see, especially if you log1p() the counts or discard rows that show much higher values. ExE ectoderm counts were much higher in samples 8 and 10 compared to 5, 7, and 9.

Further Reading

Key Points

  • Batch effects are systematic technical differences in the observed expression in cells measured in different experimental batches.
  • Computational removal of batch-to-batch variation with the correctExperiment function from the batchelor package allows us to combine data across multiple batches for a consolidated downstream analysis.
  • Differential expression (DE) analysis of replicated multi-condition scRNA-seq experiments is typically based on pseudo-bulk expression profiles, generated by summing counts for all cells with the same combination of label and sample.
  • The aggregateAcrossCells function from the scater package facilitates the creation of pseudo-bulk samples.
  • The pseudoBulkDGE function from the scran package can be used to detect significant changes in expression between conditions for pseudo-bulk samples consisting of cells of the same type.
  • Differential abundance (DA) analysis aims at identifying significant changes in cell type abundance across conditions.
  • DA analysis uses bulk DE methods such as edgeR and DESeq2, which provide suitable statistical models for count data in the presence of limited replication - except that the counts are not of reads per gene, but of cells per label.