Bulk functional analysis

Bulk RNA-seq yields many molecular readouts that are hard to interpret by themselves. One way of summarizing this information is by inferring pathway and transcription factor activities from prior knowledge.

In this notebook we showcase how to use decoupler for transcription factor (TF) and pathway activity inference from a human data-set. The data consists of 6 samples of hepatic stellate cells (HSC) where three of them were activated by the cytokine Transforming growth factor (TGF-β), it is available at GEO here.

Note

This tutorial assumes that you already know the basics of decoupler. Else, check out the Usage tutorial first.

Loading packages

First, we need to load the relevant packages, scanpy to handle RNA-seq data and decoupler to use statistical methods.

[1]:
import scanpy as sc
import decoupler as dc

# Only needed for processing
import numpy as np
import pandas as pd
from anndata import AnnData

Loading the data

We can download the data easily from GEO:

[2]:
!wget 'https://www.ncbi.nlm.nih.gov/geo/download/?acc=GSE151251&format=file&file=GSE151251%5FHSCs%5FCtrl%2Evs%2EHSCs%5FTGFb%2Ecounts%2Etsv%2Egz' -O counts.txt.gz
!gzip -d -f counts.txt.gz
--2022-09-01 12:46:55--  https://www.ncbi.nlm.nih.gov/geo/download/?acc=GSE151251&format=file&file=GSE151251%5FHSCs%5FCtrl%2Evs%2EHSCs%5FTGFb%2Ecounts%2Etsv%2Egz
Resolving www.ncbi.nlm.nih.gov (www.ncbi.nlm.nih.gov)... 130.14.29.110, 2607:f220:41e:4290::110
Connecting to www.ncbi.nlm.nih.gov (www.ncbi.nlm.nih.gov)|130.14.29.110|:443... connected.
HTTP request sent, awaiting response... 200 OK
Length: 1578642 (1,5M) [application/octet-stream]
Saving to: ‘counts.txt.gz’

counts.txt.gz       100%[===================>]   1,50M   349KB/s    in 4,6s

2022-09-01 12:47:00 (336 KB/s) - ‘counts.txt.gz’ saved [1578642/1578642]

We can then read it using pandas:

[3]:
# Read raw data and process it
adata = pd.read_csv('counts.txt', index_col=2, sep='\t').iloc[:, 5:].T
adata
[3]:
GeneName DDX11L1 WASH7P MIR6859-1 MIR1302-11 MIR1302-9 FAM138A OR4G4P OR4G11P OR4F5 RP11-34P13.7 ... MT-ND4 MT-TH MT-TS2 MT-TL2 MT-ND5 MT-ND6 MT-TE MT-CYB MT-TT MT-TP
25_HSCs-Ctrl1 0 9 10 1 0 0 0 0 0 33 ... 93192 342 476 493 54466 17184 1302 54099 258 475
26_HSCs-Ctrl2 0 12 14 0 0 0 0 0 0 66 ... 114914 355 388 436 64698 21106 1492 62679 253 396
27_HSCs-Ctrl3 0 14 10 0 0 0 0 0 0 52 ... 155365 377 438 480 85650 31860 2033 89559 282 448
31_HSCs-TGFb1 0 11 16 0 0 0 0 0 0 54 ... 110866 373 441 481 60325 19496 1447 66283 172 341
32_HSCs-TGFb2 0 5 8 0 0 0 0 0 0 44 ... 45488 239 331 343 27442 9054 624 27535 96 216
33_HSCs-TGFb3 0 12 5 0 0 0 0 0 0 32 ... 70704 344 453 497 45443 13796 1077 43415 192 243

6 rows × 64253 columns

Note

In case your data does not contain gene symbols but rather gene ids like ENSMBL, you can use the function sc.queries.biomart_annotations to retrieve them. In this other vignette, ENSMBL ids are transformed into gene symbols.

Note

In case your data is not from human but rather a model organism, you can find their human orthologs using pypath. Check this GitHub issue where mouse genes are transformed into human.

The obtained data consist of raw read counts for six different samples (three controls, three treatments) for ~60k genes. Before continuing, we will transform our expression data into an AnnData object. It handles annotated data matrices efficiently in memory and on disk and is used in the scverse framework. You can read more about it here and here.

[4]:
# Transform to AnnData object
adata = AnnData(adata)
adata.var_names_make_unique()
adata
Variable names are not unique. To make them unique, call `.var_names_make_unique`.
/home/badi/miniconda3/envs/decoupler/lib/python3.8/site-packages/anndata/utils.py:111: UserWarning: Suffix used (-[0-9]+) to deduplicate index values may make index values difficult to interpret. There values with a similar suffixes in the index. Consider using a different delimiter by passing `join={delimiter}`Example key collisions generated by the make_index_unique algorithm: ['SNORD116-1', 'SNORD116-2', 'SNORD116-3', 'SNORD116-4', 'SNORD116-5']
  warnings.warn(
[4]:
AnnData object with n_obs × n_vars = 6 × 64253

Inside an AnnData object, there is the .obs attribute where we can store the metadata of our samples. Here we will infer the metadata by processing the sample ids, but this could also be added from a separate dataframe:

[5]:
# Process treatment information
adata.obs['condition'] = ['control' if '-Ctrl' in sample_id else 'treatment' for sample_id in adata.obs.index]

# Process sample information
adata.obs['sample_id'] = [sample_id.split('_')[0] for sample_id in adata.obs.index]

# Visualize metadata
adata.obs
[5]:
condition sample_id
25_HSCs-Ctrl1 control 25
26_HSCs-Ctrl2 control 26
27_HSCs-Ctrl3 control 27
31_HSCs-TGFb1 treatment 31
32_HSCs-TGFb2 treatment 32
33_HSCs-TGFb3 treatment 33

Quality control

Before doing anything we need to ensure that our data passes some quality control thresholds. In transcriptomics it can happen that some genes were not properly profiled and thus need to be removed. We can check for lowly expressed genes by running this line:

[6]:
dc.plot_violins(adata, log=True, thr=4, title='Log-unnormalized counts per sample')
../_images/notebooks_bulk_15_0.png

It can be observed that across samples there seems to be a bimodal distribution of lowly and highly expressed genes.

We can set the lowly expressed genes to 0, in this case less than 4:

[7]:
# Set features to 0
dc.mask_features(adata, log=True, thr=4)

Then we can remove samples with less than 200 highly expressed genes and genes that are not expressed across all samples.

Note

This thresholds can vary a lot between datasets, manual assessment of them needs to be considered. For example, it might be the case that many genes are not expressed in just one sample which they would get removed by the current setting. For this specific dataset it is fine.

[8]:
# Filter out samples
sc.pp.filter_cells(adata, min_genes=200)

# Filter out features not expressed in all samples (n=6)
sc.pp.filter_genes(adata, min_cells=adata.shape[0])

Now we can visualize again how the distributions look like after filtering:

[9]:
dc.plot_violins(adata, log=True, thr=4, title='Log-unnormalized counts per sample')
../_images/notebooks_bulk_21_0.png

Normalization

In order to work with transcriptomics data we need to normalize it due to the high variance between readouts. Many normalization techniques exist but for simplicity, here we will use the log transformed Counts Per Million (CPM) normalization.

[10]:
# Log-CPM counts
sc.pp.normalize_total(adata, target_sum=1e6)
sc.pp.log1p(adata)

Now we visualize how the samples look like after normalization

[11]:
dc.plot_violins(adata, title='Log-CPM counts per sample')
../_images/notebooks_bulk_25_0.png

Differential expression analysis

In order to identify which are the genes that are changing the most between treatment and control we can perform differential expression analysis (DEA). Like in normalization, many approaches exists but for simplicity here we use a t-test between groups.

[12]:
# Run DEA
logFCs, pvals = dc.get_contrast(adata,
                                group_col=None,
                                condition_col='condition',
                                condition='treatment',
                                reference='control',
                                method='t-test'
                               )
logFCs
[12]:
CICP27 MTND1P23 MTND2P28 AC114498.1 MIR6723 RP5-857K21.7 MTATP8P1 MTATP6P1 RP5-857K21.11 LINC01128 ... MT-ND4 MT-TH MT-TS2 MT-TL2 MT-ND5 MT-ND6 MT-TE MT-CYB MT-TT MT-TP
treatment.vs.control -0.135726 -0.601097 -0.450566 -0.536081 -0.870987 -0.559754 -0.841359 -0.685194 -0.650889 0.557 ... -0.718909 -0.168917 -0.075884 -0.08817 -0.64625 -0.726885 -0.649676 -0.624246 -0.809369 -0.724974

1 rows × 14254 columns

After running DEA, we obtain how much each gene is changing in treatment compared to controls (logFCs), and how statistically singificant is this change (pvals).

We can further explore these changes by plotting them into a volcano plot.

[13]:
dc.plot_volcano(logFCs, pvals, 'treatment.vs.control', top=15, sign_thr=0.05, lFCs_thr=0.5)
../_images/notebooks_bulk_29_0.png

To extract the top significant genes after FDR correction (adj_pvals), we can run this:

[14]:
top_genes = dc.get_top_targets(logFCs, pvals, 'treatment.vs.control', sign_thr=0.05, lFCs_thr=0.5)
top_genes
[14]:
contrast name logFCs pvals adj_pvals
0 treatment.vs.control LPAR1 -2.856200 1.555664e-08 0.000222
1 treatment.vs.control FTH1 -2.028517 1.330181e-07 0.000948
2 treatment.vs.control NTNG2 -0.901692 2.986280e-07 0.000957
3 treatment.vs.control FTL -1.825337 3.792117e-07 0.000957
4 treatment.vs.control APCDD1L 2.706221 4.179862e-07 0.000957
... ... ... ... ... ...
3766 treatment.vs.control VAMP1 -0.892178 2.064460e-02 0.049842
3767 treatment.vs.control RP11-214K3.25 -0.652381 2.065199e-02 0.049852
3768 treatment.vs.control SLC26A6 -0.787473 2.071529e-02 0.049963
3769 treatment.vs.control ANKDD1A -1.015187 2.071575e-02 0.049963
3770 treatment.vs.control BCKDHB -0.627068 2.072337e-02 0.049965

3771 rows × 5 columns

Pathway activity inference

To estimate pathway activities we will use the resource PROGENy and the consensus method.

For another example on pathway activities please visit this other notebook: Pathway activity inference.

[15]:
# Retrieve PROGENy model weights
progeny = dc.get_progeny(top=300)

# Infer pathway activities with consensus
pathway_acts, pathway_pvals = dc.run_consensus(mat=logFCs, net=progeny)
pathway_acts
[15]:
Androgen EGFR Estrogen Hypoxia JAK-STAT MAPK NFkB PI3K TGFb TNFa Trail VEGF WNT p53
treatment.vs.control 0.073365 0.118366 0.193759 0.553585 -1.439839 0.346702 -0.469478 0.389559 2.781179 -1.26574 -0.077649 0.246556 0.080174 -0.516285

After running the consensus method, we obtained activities and p-values for each pathway.

We can visualize by runnig this:

[16]:
dc.plot_barplot(pathway_acts, 'treatment.vs.control', top=25, vertical=False)
../_images/notebooks_bulk_35_0.png

As expected, after treating cells with the cytokine TGFb we see an increase of activity for this pathway.

On the other hand, it seems that this treatment has decreased the activity of other pathways like JAK-STAT or TNFa.

We can visualize the targets of TFGb in a volcano plot:

[17]:
dc.plot_volcano(logFCs, pvals, 'treatment.vs.control', name='TGFb', net=progeny, top=5, sign_thr=0.05, lFCs_thr=0.5)
../_images/notebooks_bulk_37_0.png

In this plot we can see that genes with positive weights towards TFGb have positive logFCs, while genes with negative weights have negative logFCs. This justifies the high activity that we observed.

On the contrary if we look at JAK-STAT:

[18]:
dc.plot_volcano(logFCs, pvals, 'treatment.vs.control', name='JAK-STAT', net=progeny, top=5, sign_thr=0.05, lFCs_thr=0.5)
../_images/notebooks_bulk_39_0.png

Here most of the downstream genes with positive weights of JAK-STAT seem to have negative logFCs, indicating that the pathway may be inactive compared to controls.

Transcription factor activity inference

Similarly to pathways, we can estimate transcription factor activities using the resource DoRothEA and the consensus method.

For another example on transcription factor activities please visit this other notebook: Transcription factor activity inference.

[19]:
# Retrieve DoRothEA gene regulatory network
dorothea = dc.get_dorothea()

# Infer pathway activities with consensus
tf_acts, tf_pvals = dc.run_consensus(mat=logFCs, net=dorothea)
tf_acts
[19]:
AHR AR ARID2 ARID3A ARNT ARNTL ASCL1 ATF1 ATF2 ATF3 ... ZNF217 ZNF24 ZNF263 ZNF274 ZNF384 ZNF584 ZNF592 ZNF639 ZNF644 ZNF740
treatment.vs.control -1.366347 -0.223558 0.404413 -0.129955 -0.018745 0.494533 -0.04347 0.283429 0.212837 -0.244554 ... -0.610369 -0.002195 -1.472273 -0.889196 0.754258 -0.240206 -0.039853 0.712266 0.654209 0.159676

1 rows × 278 columns

After running the consensus method, we obtained activities and p-values for each transcription factor.

We can visualize the most active and inactive transcription factor by runnig this:

[20]:
dc.plot_barplot(tf_acts, 'treatment.vs.control', top=25, vertical=True)
../_images/notebooks_bulk_44_0.png

EPAS1, SRF, and TP63 seem to be the most activated in this treatment while STAT2, HNF4A AND SOX10 seem to be inactivated.

As before, we can manually inspect the downstream targets of each transcription factor:

[21]:
dc.plot_volcano(logFCs, pvals, 'treatment.vs.control', name='EPAS1', net=dorothea, top=5, sign_thr=0.05, lFCs_thr=0.5)
../_images/notebooks_bulk_46_0.png
[22]:
dc.plot_volcano(logFCs, pvals, 'treatment.vs.control', name='STAT2', net=dorothea, top=5, sign_thr=0.05, lFCs_thr=0.5)
../_images/notebooks_bulk_47_0.png

Functional enrichment of biological terms

We can also assign the obtained DEG biological terms using the resource MSigDB and the ora method.

For another example on functional enrichment of biological terms please visit this other notebook: Functional enrichment of biological terms.

[23]:
# Retrieve MSigDB resource
msigdb = dc.get_resource('MSigDB')
msigdb

# Filter by a desired geneset collection, for example hallmarks
msigdb = msigdb[msigdb['collection']=='hallmark']
msigdb = msigdb.drop_duplicates(['geneset', 'genesymbol'])

# Infer enrichment with ora using significant deg
top_genes = dc.get_top_targets(logFCs, pvals, 'treatment.vs.control', sign_thr=0.05, lFCs_thr=1.5)
enr_pvals = dc.get_ora_df(top_genes, msigdb, groupby='contrast', features='name', source='geneset', target='genesymbol')
enr_pvals
[23]:
HALLMARK_ADIPOGENESIS HALLMARK_ALLOGRAFT_REJECTION HALLMARK_ANDROGEN_RESPONSE HALLMARK_ANGIOGENESIS HALLMARK_APICAL_JUNCTION HALLMARK_APOPTOSIS HALLMARK_COAGULATION HALLMARK_COMPLEMENT HALLMARK_EPITHELIAL_MESENCHYMAL_TRANSITION HALLMARK_ESTROGEN_RESPONSE_EARLY ... HALLMARK_MITOTIC_SPINDLE HALLMARK_MTORC1_SIGNALING HALLMARK_MYOGENESIS HALLMARK_P53_PATHWAY HALLMARK_REACTIVE_OXYGEN_SPECIES_PATHWAY HALLMARK_TGF_BETA_SIGNALING HALLMARK_TNFA_SIGNALING_VIA_NFKB HALLMARK_UV_RESPONSE_DN HALLMARK_UV_RESPONSE_UP HALLMARK_XENOBIOTIC_METABOLISM
treatment.vs.control 5.764578e-11 7.876590e-17 7.876590e-17 4.889991e-08 1.373635e-28 1.373635e-28 3.564059e-24 1.205612e-25 0.0 4.072404e-27 ... 1.680132e-09 9.129221e-20 1.052105e-22 1.373635e-28 2.308692e-15 1.975060e-12 0.0 7.275542e-42 7.876590e-17 3.564059e-24

1 rows × 31 columns

We can then transform these p-values to their -log10 (so that the higher the value, the more significant the p-value). We will also set 0s to a minimum p-value so that we do not get infinites.

[24]:
# Set 0s to min p-value
enr_pvals.values[enr_pvals.values == 0] = np.min(enr_pvals.values[enr_pvals.values != 0])

# Log-transform
enr_pvals = -np.log10(enr_pvals)

Then we can visualize the most enriched terms:

[25]:
dc.plot_barplot(enr_pvals, 'treatment.vs.control', top=25, vertical=True)
../_images/notebooks_bulk_53_0.png

TNFa and interferons response (JAK-STAT) processes seem to be enriched. We previously observed a similar result with the PROGENy pathways, were they were significantly downregulated. Therefore, one of the limitations of using a prior knowledge resource without weights is that it doesn’t provide direction.